<<

Int J Clin Exp Med 2018;11(1):59-68 www.ijcem.com /ISSN:1940-5901/IJCEM0056886

Review Article and β-oxidation, which one is the culprit of ischemic ?

Qing Gao1*, Hao Deng2*, Huhu Li1*, Chun Sun1, Yingxin Sun1, Bing Wei1, Maojuan Guo1, Xijuan Jiang1

1School of Integrative Medicine, 2First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China. *Equal contributors Received May 5, 2017; Accepted November 14, 2017; Epub January 15, 2018; Published January 30, 2018

Abstract: Thrombolysis therapy and percutaneous coronary intervention are common methods in the treatment of acute myocardial infarction. These methods can recover the cardiac function in most cases. But in almost one-third circumstances, cardiac dysfunction and structural damage aggravated, which is known as ischemia-reperfusion injury. Normally, most ATP in cardiomyocytes was produced from fatty acid β-oxidation. However, both fatty acid β-oxidation and glycolysis accelerated due to AMPK activation during ischemic. Glycolysis uncoupled from oxidation results in intermediate metabolite accumulation, such as lactate, proton, succinate and NADH. During reperfusion, the recovering rate of fatty acid β-oxidation even exceed the rate under physiological condition due to the sudden influx of high concentration of . High rate of fatty acid β-oxidation inhibits glycose oxidation and results in proton and Ca2+ overload, especially huge amount of ROS production, which leads to mitochondria damage and death. Clearly, energy disorder result from the sudden change of oxygen supply during ischemic and reperfusion is the main cause of ischemic reperfusion injury. However, glycolysis and fatty acid β-oxidation, which one is the real culprit in ischemic reperfusion injury is controversial. In this review, we will discuss the process of metabolism and fatty acid β-oxidation thoroughly, as well as the energy sensor AMPK signaling, in order to clarify how to modulate energy metabolism to reduce injury during ischemic and reperfusion.

Keywords: Glycolysis, fatty acid β-oxidation, intermediate metabolite, ischemic reperfusion injury

Introduction FFA in the plasma and the intracellular level of malonyl-CoA can regulate the rate of fatty Although cardiomyocytes were supplied by mul- acid β-oxidation [5, 6]. Malonyl-CoA is synthe- tiple energy sources, fatty acid and glucose are sized from cytosolic acetyl-CoA via acetyl-CoA the main ones. Under physiological condition, carboxylase (ACC), while it is degraded thro- most of its energy was produced from fatty acid ugh malonyl-CoA decarboxylase (MCD) [7, 8]. β-oxidation (FAO) (All abbreviations are listed in Malonyl-CoA regulates fatty acid β-oxidation by Table 1), due to its high efficiency in ATP pro- inhibiting the activity of CPT-I, which is the rate duction. Free fatty acids (FFA) in cardiomyo- limiting of mitochondrial fatty acid cytes generate fatty acyl-CoA following este- uptake, thereby it controls the rate of fatty acids rification reaction, the process of which is cata- entering into the mitochondria for subsequent lyzed by a family of fatty acyl-CoA synthase oxidation [9, 10]. (FACS) [1]. The mitochondrial uptake of fatty acyl-CoA into its matrix is mediated by Since the well-known fact that fatty acid are palmitoyl- I and II (CPT-I, normally the predominant fuel for cardiac ener- II), which are localized to the mitochondrial gy production, aerobic glucose metabolism has outer membrane and inner membrane respec- been neglected in heart. Actually, it is respon- tively [2]. Once enter the mitochondrial matrix, sible for 10%-40% of ATP production in cardio- fatty acyl-CoA are catalyzed via the process of myocytes [11]. Glucose transportation into car- fatty acid β-oxidation, eventually they were dis- diomyocytes was regulated by glucose trans- membered to acyl-CoA that were metabolized porter family members such as GLUT 1 in TCA cycle [3, 4]. Both the level of circulating and 4, which are predominantly expressed at Glycolysis and fatty acid β-oxidation disorder in IR

Table 1. List of abbreviations Actually, the heart is an organ that can Full name Abbreviate exert maximum function when it apply different energy sources simultane- Fatty acid β-oxidation FAO ously [11]. In aerobic condition, high Free fatty acids FFA rate of fatty acid β-oxidation can inhibit Acyl-CoA synthase FACS glucose oxidation in cardiomyocytes. Carnitine palmitoyl-transferase I and II CPT-I, II This phenomenon is based on the Malonyl-CoA decarboxylase MCD Randle cycle [20], in which fatty acid- Acetyl-CoA carboxylase ACC derived acetyl CoA can decrease the Pyruvate decarboxylase PDC production of glucose-derived acetyl 6-phosphofructo-1- PFK-1 CoA via inhibition of the pyruvate dehy- drogenase complex. On the other side, Reactive oxygen species ROS under anoxic condition, energy-provi- Ischemic reperfusion IR sion way switch into the more efficient ETC way, glycolysis but brings harmful Activated AMPK metabolites. Any alterations in energy adenine dinucleotide NDAH metabolism can contribute to develop- Reverse electron transport RET ment of heart diseases, including IR Coenzyme Q CoQ injury. Optimizing energy metabolism binding module CBM in the heart is a feasible and important approach to treat IRI. Under this con- Cystathionine-b-synthase CBS cept, we recapitulate myocardial ener- kinase B1 LKB1 gy metabolism and its relevance to IR injury. the surface of adult cardiomyocytes [12]. Alterations of glycolysis and fatty acid Intracellular glucose is rapidly phosphorylated β-oxidation and becomes a for the glycolytic pathway, synthesis, and syn- Ischemic thesis [13, 14]. Once entering the glycolytic pathway, the process will be examined by key During ischemia, ATP production from electron enzymes such as , 6-phosphofruc- transport chain (ETC) is almost terminated to-1-kinase (PFK-1) and [15]. without oxygen. It results in AMP accumulated, Pyruvate enters the mitochondria via a mono- which activate Adenosine Monophosphate Ac- carboxylate carrier, and becomes a cross point tivated Protein Kinase (AMPK) signaling (Figure for several metabolic pathways. For example, it 1) [21]. Activated AMPK can accelerate both can produce lactate glycolysis; it can convert to glucose and fatty acid β-oxidation through relo- acetyl-CoA by pyruvate decarboxylase (PDC), cating GLUT4 and /CD36 to sarcolemma as and transform into oxaloacetate [16]. well as phosphorylating PFK-1 and inhibiting ACC [22]. Subsequently, malonyl-CoA decre- Although ischemic treatment such as coronary ase thus relieves the inhibition of CPT-1 [23]. bypass surgery, thrombolysis, and percutane- However, Krebs cycle cannot disposal the huge ous coronary intervention achieved significant amount of Acetyl-CoA from glucose and fatty accomplishment, ischemic reperfusion (IR) in- acid oxidation with blocked ETC [24]. As a con- jury is still to be solved [17]. Reactive oxygen sequence, Acetyl-CoA produced from fatty acid species (ROS) and Ca2+ overload are the main β-oxidation will inhibit PDC, i.e. Randle cycle as culprits as supported by many researchers previously described, and results in glycolysis [18]. But the resources of ROS and Ca2+ are still that uncoupled with ATP production [25]. In under debates. During ischemic and reperfu- addition, fatty acid accumulate in cytoplasma sion process, oxygen supply in cardiomyocytes under both prandial state and catecholamine changed suddenly, which cause energy metab- discharge. Catecholamine discharge was up- olism disorder and further damage [19]. The regulated in the ischemic stress, together with heart has a very high energy demand, and of plasma levels [26]. Catech- course, oxygen demand. Energy metabolism olamines stimulate adipose , pathway changes with oxygen concentration. decrease pancreatic release, and des-

60 Int J Clin Exp Med 2018;11(1):59-68 Glycolysis and fatty acid β-oxidation disorder in IR

reuptake of Ca2+ into the sar- coplasmic reticulum, respec- tively, leads to intracellular Ca2+ overload, which in turn increases the production of free radicals. This also chang- es the affinity of different pro- teins, such as enzymes and troponin C, to Ca2+ [35], modi- fies tertiary protein structures, inhibits enzymes and disrupts the function of sarcoplasmic pumps and carriers [36-38]. Low dose of Na+/Ca2+ exch- ange inhibitor protects isch- emic reperfusion injury in rat hearts [39]. In general, glycol- ysis generates ATP for cardio- Figure 1. Alterations of glucose and fatty acid β-oxidation metabolism dur- ing ischemic. AMP accumulation during ischemic leads to AMPK activation, myocytes utilization, but also which accelerate both glucose and fatty acid β-oxidation. High rate of fatty results in intracellular acido- acid β-oxidation results in glycolysis uncoupled from oxidation. Then the in- sis, NADH and Ca2+ overload termediate of glycolysis accumulated, including lactate, protons and NDAH. during ischemic. Intracellular impair the activity of Na+/K+ ATPase, which extrudes + + + 3Na ions in exchange for 2K ions, leads to intracellular Na overload, sub- Reperfusion sequently activation of Na+/Ca2+ exchangers, results in intracellular Ca2+ overload. Excess NADH in enter the mitochondria membrane through malate/aspartate shuttle. Moreover, ischemic succinate accumu- At reperfusion, the rates of lation arises from reversal of succinate , which is driven by glucose oxidation, cardiac effi- fumarate overflow from purine break down and partial reversal ciency, and mechanical func- of the malate/aspartate shuttle. Green arrow means normal physiological tion remain depressed [40. process. Red arrow means pathophysiological process. Red X mark means 41]. The accelerated rates of the pathway is inhibited. glycolysis during ischemic pe- riod can resolve during reper- ensitize peripheral insulin [27-29]. Meanwhile, fusion, but still remain uncoupled from glucose plasma levels of hydrocortisone elevate, which oxidation (Figure 2) [42-44]. Furthermore, as also desensitize insulin [30]. All these effects reperfusion rapidly normalizes extracellular pH, promote lipolysis that leads to it generates a large trans-sarcolemmal proton increase plasma concentrations of FFA and gradient that increases Na+/H+ exchange and increase delivery of FFA to the myocardium. The exacerbates intracellular Na+ overload followed increased delivery of FFA to the myocardium ischemia. In return, Na+ overload promotes can alter fatty acid utilization during both isch- reverse activation of the Na+/Ca2+ exchanger, emia, and reperfusion following ischemia. thereby contributing to intracellular Ca2+ over- load and aggravation during reperfusion [45]. Although ATP production from glycolysis may be These disturbances in ionic dur- sufficient to maintain ionic homeostasis during mild to moderate ischemia, the of ing ischemia and during reperfusion contribute ATP derived from glycolysis uncoupled from to the deficits in both cardiac function and car- subsequent pyruvate oxidation results in the diac efficiency, both of which can be improved increased generation of lactate, protons and by therapies that correct myocardial energy nicotinamide adenine dinucleotide (NDAH) [31- production. By activating the PDC, by products 33]. Intracellular acidosis impair the activity of accumulation can be limited, and cardiac func- Na+/K+ ATPase [34], which leads to intracellular tional recovery is improved. Improving the cou- Na+ overload and subsequent activation of Na+/ pling of glucose metabolism by stimulating glu- Ca2+ exchangers. Also, impaired activity of the cose oxidation accelerates the recovery of PH sarcolemmal and sarcoplasmic Ca2+ ATPase and improves both cardiac mechanical function which are responsible for the extrusion and and cardiac efficiency [44].

61 Int J Clin Exp Med 2018;11(1):59-68 Glycolysis and fatty acid β-oxidation disorder in IR

theyenhance oxygen efficien- cy during myocardial ischemic [48, 49].

Succinate accumulates and reverse electron transport chain

Accumulation of the Krebs cy- cle intermediate succinate is a universal metabolic marker of ischemia due to its role in tissues arrangement and mi- tochondrial ROS production during reperfusion. Most im- portantly, succinate accumu- lation arises from reversal of succinate dehydrogenase un- Figure 2. Alterations of glucose and fatty acid β-oxidation metabolism in re- der ischemic conditions, wh- perfusion. At reperfusion, fatty acid β-oxidation is rapidly recovered due to the sudden restore of oxygen, with rates exceeding pre-ischemic levels. The ich in turn is driven by fum- combination of high-circulating fatty acid levels and a decrease in malonyl arate overflow from purine CoA control of mitochondrial fatty acid uptake results in the preferential use nucleotide break down and of fatty acids as an oxidative substrate over glucose at reperfusion. High partial reversal of the malate/ fatty acid oxidation rates still inhibit glucose oxidation. Most importantly, the aspartate shuttle [50]. Elev- accumulated succinate is rapidly re-oxidized by succinate dehydrogenase, driving extensive ROS generation by reverse electron transport at mitochon- ated NADH level in the cyto- drial complex I in the first few minutes. MPTP opening because of ROS burst plasm that traveled out from and Ca2+ overload. Green arrow means normal physiological process. Red mitochondria during glycolysis arrow means pathophysiological process. Red X mark means the pathway relies on malate aspartate is inhibited. shuttle in cardiomyocyte [51]. Deamination of aspartate to Paradoxically, fatty acid β-oxidation is rapidly oxaloacetate transfers electrons from NADH in recovered at reperfusion, due to the sudden the to form malate. The malate in the restore of oxygen, with its rates exceeding pre- cytosol is then exchanged with mitochondrial ischemic levels [46]. The combination of high- succinate through dicarboxylate carrier [52]. circulating fatty acid levels with decreased Once in the matrix, the succinate fails to form malonyl CoA mitochondrial fatty acid uptake succinyl CoA due to the lack of CoA and GTP results in the preferential use of fatty acids as during ischemia. In parallel, AMPs, which accu- an energy source over glucose at reperfusion. mulates during ischemia, was metabolized into High fatty acid oxidation rates also inhibit glu- fumarate through purine nucleotide cycle [53]. cose oxidation. Because the regulation of malo- This fumarate can then be hydrolyzed to malate nyl CoA levels is central in administration of the by cytosolic fumarate hydratase. rate of fatty acid β-oxidation in the heart, MCD may play a major role in controlling the extent of After reperfusion, the accumulated succinates ischemic injury by promoting glucose oxidation. are rapidly re-oxidized by succinate dehydroge- This hypothesis was proved by the study that nase, which drives extensive ROS generation MCD inhibitors can increase myocardial malo- by reverse electron transport (RET) at mito- nyl CoA levels, decrease fatty acid β-oxidation, chondrial complex I in the first few minutes [54, accelerate glucose oxidation in both in vitro 55]. Under physiological condition, transpo- and in vivo [47]. This suggests that switching tation of electron across the differences in in energy substrate preference improves cardi- reduction potential between the NAD+/NADH ac function during and after ischemia. Per- and the Coenzyme Q (CoQ) pool across com- hexiline, trimetazidine, ranolazine, and etomox- plex I (ΔEh) has to be sufficient to pump prot- ir, all stimulate glucose metabolism while down ons across the mitochondrial inner membrane regulate free-fatty-acid metabolism. Therefore, against the proton motive force, Δp [56]. As

62 Int J Clin Exp Med 2018;11(1):59-68 Glycolysis and fatty acid β-oxidation disorder in IR

specific pathway through wh- ich AMPK works seems am- biguous in the previous lite- ratures. AMPK is a heterotrim- eric complex, and composed of a catalytic α-subunit com- prising a typical Ser/Thr kin- ase domain and regulatory β and γ subunits [63]. Each subunit has multiple isoforms (α1 and α2, β1 and β2, and γ1, γ2, and γ3) with tissue- Figure 3. Role of AMPK in ischemic reperfusion injury. Ca2+-dependent path- specific distribution. In heart, way mediated by CaMKKβ and an AMP-dependent pathway mediated by all isoforms have been report- liver kinase B1 (LKB1) can activate AMPK. Firstly, activated AMPK promote glucose utilization. AMPK stimulate glucose uptake by translocating GLUT4- ed to be expressed [64]. The containing intracellular vesicles across the plasma membrane by phosphor- α-subunit of AMPK is a 63- ylating PKC and p38MAPK/TAB1 pathway. Besides, AMPK promotes glycoly- kDa protein that exhibits ca- sis by increasing PFK2 activity. Secondly, AMPK inhibits FA synthesis while talytic activity. The α1 isoform increase Fatty acid oxidation through PGC1α/PPAR pathway. is widely expressed, whereas the α2 isoform has its highest four protons are pumped for every two elec- levels of expression in liver, heart, and skele- trons that pass through complex I, 2ΔEh > 4Δp tal muscle. The β and γ subunits appear to is requirement for the forward reaction to oc- be important in substrate specificity and ma- cur [57]. Electrons can be driven backward intenance of heterotrimer stability [65]. The from the CoQ pool onto the FMN of complex I, β-subunit acts as a scaffold for the binding of reduces the FMN which can donate a pair of the α- and γ-subunits and, by virtue of having a electrons to NAD+ to form NADH, or pass one carbohydrate binding module (CBM), also func- electron to oxygen to generate superoxide. The tion in the regulation of glycogen metabolism condition to be met for RET to occur is 4Δp > 2 [66]. The γ-subunit containing four cystathio- ΔEh [58]. The rapid oxidation of the succinate nine-b-synthase (CBS) domains, that serve to that accumulates during ischemia favors re- bind adenine and has also been duction of the CoQ pool, thereby maintaining a implicated in regulating glycogen metabolism, large ΔEh [59]. The reduced CoQ pool also since mutations in the CBS domains lead to favors proton pumping by complexes III and altered glycogen metabolism in both skeletal IV helping maintain a large Δp upon reperfu- muscle and heart. sion [60]. In addition, the degradation of ade- nine nucleotides during ischemia limits ADP The mechanism of AMPK activation involves 2+ availability upon reperfusion that would other- two distinct signals: a Ca -dependent pathway wise diminish Δp by stimulating ATP synthesis. mediated by CaMKKβ and an AMP-dependent In this scenario accumulated succinate act as pathway mediated by liver kinase B1 (LKB1) electron sink during ischemia, which is then (Figure 3) [67]. Theupstream phos- used to drive ROS by RET at complex I upon phorylated at Thr172 on α subunit, stimulate reperfusion. Excessive ROS will bring further allosteric effect upon binding of AMP within damage to the mitochondrial, such as Mito- the CBS domain of the γ subunit, thereby main- chondrial Permeability Transition Pore (MPTP) taining the enzyme in the activated state, as opening. well as protecting Thr172 from dephosphoryla- tion [68]. Thus, AMPK serves as a unique meta- AMPK signaling in IR bolic control node as it senses cellular energy status through modulation of its activities via AMPK, known as energy sensor, has been and allosteric activation by reported to be related to ischemic reperfusion AMP [69]. Numerous pathological processes injury. Intrinsic modulation of AMPK is critical have been shown to stimulate AMPK, including to prevent irreversible mitochondrial damage conditions that lead to alterations of the intra- and myocardial injury [61, 62]. However, the cellular AMP/ATP ratio (e.g., hypoxia, glucose

63 Int J Clin Exp Med 2018;11(1):59-68 Glycolysis and fatty acid β-oxidation disorder in IR deprivation) and calcium overload, which are AMPK activation is harmful for cardiomyocytes. the notable symptoms in ischemic reperfusion Since high rates of fatty acid oxidation can con- injury [70, 71]. tribute to ischemic damage by inhibiting glu- cose oxidation, it is important to maintain prop- Once AMPK activated, cardiomyocytes con- er control of fatty acid oxidation both during ducted energy consumption rather than reser- and following ischemic. vation, which means overwhelms . On one hand, AMPK promotes glu- Conclusion and future perspective cose utilization. AMPK stimulates glucose up- take by translocating GLUT4-containing intra- In the present review, we analyzed the altera- cellular vesicles across the plasma membrane tions of glucose and fatty acid β-oxidation dur- [72]. AMPK involves phosphorylation and acti- ing IR, as well as the role of AMPK signaling. In vation of Akt substrate of 160 kDa [73], protein brief, glucose uncoupled from oxidation due to kinase C [74], endothelial nitric oxide synthase high rates of fatty acid β-oxidation result in [75] and p38 mitogen-activated protein kinase/ intermediate metabolites, which are the main transforming growth factor β-activated protein pathogenic factor of reperfusion during isch- complex 1 [76], regulate GLUT4 translocation emic. In this case, glycolysis is the leading to the plasma membrane. Besides, AMPK pro- cause of IR injury. For this reason, the inhibition motes glycolysis by increasing phosphofructo- of glycolysis during ischemia will lessen lactate kinase 2 activity [77, 78], which produces fruc- and proton production, and improve cardiac tose 2, 6-bisphosphate, a potent stimulator of efficiency [80]. Previous clinical studies focused glycolysis. On the other hand, AMPK inhibits FA on inhibiting fatty acid oxidation and increasing synthesis while increase Fatty acid oxidation. glucose oxidation during ischemic heart dis- The phosphorylation process of acetyl-coA car- ease, but barely no study paid attention to the boxylase 1, which catalyzes the rate-limiting metabolites of glycolysis. Since AMPK signaling step in and sterol regulato- has dual effect in regulating glucose and fatty ry element-binding protein 1c, a transcription acid metabolism, we need to elucidate the pre- factor that promotes the expression of multi- cise molecular pathway when we want to inhibit ple lipogenic enzymes were inhibited. While glycolysis and promote glucose oxidation. Also, fatty acid uptake is increased by promoting the decreasing ischemic succinate accumulation translocation of fatty acid transporter CD36 to by pharmacological inhibition is sufficient to the plasma membrane, the mechanism under- ameliorate in vivo IR injury in murine models of lying this is unclear. In cytoplasm, fatty acids heart attack and stroke. In the future, we hope are transported into the mitochondria for more studies will focus on the bad influence of β-oxidation by CPT-1. AMPK activation enh- glycolysis and try everything to eliminate it in ances PGC-1a transcription [79], which incre- the ischemic reperfusion process. ases CPT-1 activity and activates fatty acid β-oxidation by inhibiting phosphorylation of Acknowledgements ACC2. ACC2 is localized to the outer membrane of the mitochondria near CPT-1 where it inhibits This work was supported by the National Na- production of malonyl-CoA, a potent allosteric tural Science Foundation of China (81503504, inhibitor of CPT-1. 81573733, 830472117, 81202797).

During ischemic, high AMP/ATP rate activates Disclosure of conflict of interest AMPK, which exerts catabolism to produce more ATP for cardiomyocytes utilization. Glyco- None. lysis dominates the main ATP production way Address correspondence to: Dr. Xijuan Jiang, Sc- due to lack of oxygen. In this point, AMPK hool of Integrative Medicine, Tianjin University presents beneficial effect for cardiomyocytes. of Traditional Chinese Medicine, 88th Yuquan However, during reperfusion, AMPK also pro- Road, Nankai District, Tianjin, China. Tel: +86-22- motes high rate of fatty acid β-oxidation, si- 59596287; E-mail: [email protected] nce oxygen content suddenly recover. In return, the increased rate of fatty acid β-oxidation References can inhibit glucose oxidation, which leads to increase lactate and proton production and [1] Lopaschuk GD, Ussher JR, Folmes CD, Jaswal decrease cardiac efficiency. In this situation, JS, Stanley WC. Myocardial fatty acid metabo-

64 Int J Clin Exp Med 2018;11(1):59-68 Glycolysis and fatty acid β-oxidation disorder in IR

lism in health and disease. Physiol Rev 2010; [16] Chanda D, Luiken JJ, Glatz JF. Signaling path- 90: 207-258. ways involved in cardiac energy metabolism. [2] Murthy MS, Pande SV. Malonyl-CoA binding FEBS Lett 2016; 590: 2364-2374. site and the overt carnitine palmitoyltransfer- [17] Turer AT, Hill JA. Pathogenesis of myocardial ase activity reside on the opposite sides of ischemia-reperfusion injury and rationale for the outer mitochondrial membrane. Proc Natl therapy. Am J Cardiol 2010; 106: 360-368. Acad Sci U S A 1987; 84: 378-382. [18] Hausenloy DJ, Yellon DM. Myocardial ischemia- [3] Stanley WC, Chandler MP. Energy metabolism reperfusion injury: a neglected therapeutic tar- in the normal and failing heart: potential get. J Clin Invest 2013; 123: 92-100. for therapeutic interventions. Heart Fail Rev [19] Kolwicz SC Jr, Purohit S, Tian R. Cardiac me- 2002; 7: 115-130. tabolism and its interactions with contraction, [4] McGarry JD, Brown NF. The mitochondrial car- growth, and survival of cardiomyocytes. Circ nitine palmitoyltransferase system: from con- Res 2013; 113: 603-616. cept to molecular analysis. Eur J Biochem 997; [20] Randle PJ, Garland PB, Hales CN, Newsholme 244: 1-14. EA. The glucose fatty-acid cycle. Its role in insu- [5] Dyck JR, Lopaschuk GD. Malonyl CoA control lin sensitivity and the metabolic disturbances of fatty acid oxidation in the ischemic heart. J of mellitus. Lancet 1963; 1: 785- Mol Cell Cardiol 2002; 34: 1099-1109. 789. [6] Kudo N, Barr AJ, Barr RL, Desai S, Lopaschuk [21] Qi D, Young LH. AMPK: energy sensor and sur- GD. High rates of fatty acid oxidation during re- vival mechanism in the ischemic heart. Trends perfusion of ischemic hearts are associated Endocrinol Metab 2016; 26: 422-429. with a decrease in malonyl-CoA levels due to [22] Zordoky BN, Nagendran J, Pulinilkunnil T, an increase in 5’-AMP-activated protein kin- Kienesberger PC, Masson G, Waller TJ, Kemp ase inhibition of acetyl-CoA carboxylase. J Biol BE, Steinberg GR, Dyck JR. AMPK-dependent Chem 1995; 270: 17513-17520. inhibitory phosphorylation of ACC is not essen- [7] Ussher JR, Lopaschuk GD. The malonyl CoA tial for maintaining myocardial fatty acid oxida- axis as a potential target for treating ischaemic tion. Circ Res 2014; 115: 518-524. heart disease. Cardiovasc Res 2008; 79: 259- [23] Fillmore N, Mori J, Lopaschuk GD. Mitochon- 268. drial fatty acid oxidation alterations in heart [8] Ussher JR, Lopaschuk GD. Targeting malonyl failure, ischaemic heart disease and diabetic CoA inhibition of mitochondrial fatty acid up- cardiomyopathy. Br J Pharmacol 2014; 171: take as an approach to treat cardiac ischemia/ 2080-2090. reperfusion. Basic Res Cardiol 2009; 104: [24] Czibik G, Steeples V, Yavari A, Ashrafian H. Cit- 203-210. ric acid cycle intermediates in cardioprotec- [9] Paulson DJ, Ward KM, Shug AL. Malonyl CoA tion. Circ Cardiovasc Genet 2014; 7: 711-719. inhibition of carnitine palmityltransferase in [25] FPietrocola F, Galluzzi L, Bravo-San Pedro JM, rat heart mitochondria. FEBS Lett 1984; 176: Madeo F, Kroemer G. Acetyl : a 381-384. central metabolite and second messenger. [10] Saggerson ED. Carnitine acyltransferase activi- Cell Metabolism 2015; 21: 805-821. ties in rat liver and heart measured with palmi- [26] Mueller HS, Ayres SM. Propranolol decreases toyl-CoA and octanoyl-CoA. Latency, effects of sympathetic nervous activity reflected by plas- K+, bivalent metal ions and malonyl-CoA. Bio- ma catecholamines during of myo- chem J 1982; 202: 397-405. cardial infarction in man. J Clin Invest 1980; [11] Christophe Depre, Jean-Louis J. Vanoversc- 65: 338-346. helde, heinrich taegtmeyer. Glucose for the [27] Robertson RP, Porte D Jr. Adrenergic modula- Heart. Circulation 1999; 99: 578-588. tion of basal insulin secretion in man. Diabetes [12] Shao D, Tian R. Glucose transporters in cardi- 1973; 22: 1-8. ac metabolism and hypertrophy. Compr Physiol [28] Lerner RL, Porte D Jr. : selective 2015; 6: 331-351. inhibition of the acute insulin response to glu- [13] Mapanga RF, Essop MF. Damaging effects cose. J Clin Invest 1971; 50: 2453-2457. of on cardiovascular function: [29] Christensen NJ, Videbaek J. Plasma catechol- spotlight on glucose metabolic pathways. Am J amines and in pa- Physiol Heart Circ Physiol 2016; 310: 153-173. tients with acute myocardial infarction. J Clin [14] Stephen C. Kolwicz Jr and Rong Tian. Glucose Invest 1974; 54: 278-286. metabolism and cardiac hypertrophy. Cardio- [30] Rizza RA, Mandarino LJ, Gerich JE. Cortisol-in- vascular Research 2011; 90: 194-201. duced in man: impaired sup- [15] Guoqiang Jiang and Bei B. Zhang. pression of glucose production and stimula- and regulation of glucose metabolism. Am J tion of glucose utilization due to a postreceptor Physiol Endocrinol Metab 2003; 284: 671- defect of insulin action. J Clin Endocrinol 678. Metab 1982; 54: 131-138.

65 Int J Clin Exp Med 2018;11(1):59-68 Glycolysis and fatty acid β-oxidation disorder in IR

[31] Heather LC, Pates KM, Atherton HJ, Cole MA, hearts as a result of adenosine A1 receptor Ball DR, Evans RD, Glatz JF, Luiken JJ, Griffin stimulation during reperfusion following isch- JL, Clarke K. Differential Translocation of the aemia. Br J Pharmacol 1996; 118: 355-363. fatty acid transporter, FAT/CD36, and the glu- [44] Liu Q, Docherty JC, Rendell JC, Clanachan AS, cose transporter, GLUT4, coordinates changes Lopaschuk GD. High levels of fatty acids delay in cardiac substrate metabolism during isch- the recovery of intracellular pH and cardiac ef- emia and reperfusion. Circ Heart Fail 2013; 6: ficiency in postischemic hearts by inhibiting 1058-1066. glucose oxidation. J Am Coll Cardiol 2002; 39: [32] King LM, Opie LH. Glucose and glycogen utili- 718-725. sation in myocardial ischemia—changes in me- [45] Rowe GT, Manson NH, Caplan M, Hess ML. Hy- tabolism and consequences for the myocyte. drogen peroxide and hydroxyl radical media- Mol Cell Biochem 1998; 180: 3-26. tion of activated leukocyte depression of car- [33] Depre C, Rider MH, Veitch K, Hue L. Role of diac sarcoplasmic reticulum. Participation of 2, 6-bisphosphate in the control of the pathway. Circ Res 1983; heart glycolysis. J Biol Chem 1993; 268: 53: 584-591. 13274-13279. [46] Hendrickson SC, St Louis JD, Lowe JE, Abdel- + [34] Bers DM, Barry WH, Despa S. Intracellular Na aleem S. Free during regulation in cardiac myocytes. Cardiovasc Res myocardial ischemia and reperfusion. Mol Cell 2003; 57: 897-912. Biochem 1997; 166: 85-94. [35] Hool LC. What cardiologists should know about [47] Dyck JR, Cheng JF, Stanley WC, Barr R, Chan- calcium ion channels and their regulation by dler MP, Brown S, Wallace D, Arrhenius T, Har- reactive oxygen species. Heart Lung Circ 2007; mon C, Yang G, Nadzan AM, Lopaschuk GD. 16: 361-372. Malonyl coenzyme a decarboxylase inhibition [36] McHowat J, Yamada KA, Wu J, Yan GX, Corr PB. protects the ischemic heart by inhibiting fatty Recent insights pertaining to sarcolemmal acid oxidation and stimulating glucose oxida- alterations underlying arrhythmo- tion. Circ Res 2004; 94: 78-84. genesis in the ischemic heart. J Cardiovasc [48] Kantor PF, Lucien A, Kozak R, Lopaschuk GD. Electrophysiol 1993; 4: 288-310. The antianginal drug trimetazidine shifts car- [37] Yamada KA, McHowat J, Yan GX, Donahue K, diac energy metabolism from fatty acid oxida- Peirick J, Kleber AG and Corr PB. Cellular un- tion to glucose oxidation by inhibiting mito- coupling induced by accumulation of long- chondrial long-chain 3-ketoacyl coenzyme a chain acylcarnitine during ischemia. Circ Res . Circ Res 2000; 86: 580-588. 1994; 74: 83-95. [49] Lee L, Horowitz J, Frenneaux M. Metabolic ma- [38] Neely JR, Feuvray D. Metabolic products and nipulation in ischaemic heart disease, a novel myocardial ischemia. Am J Pathol 1981; 102: approach to treatment. Eur Heart J 2004; 25: 282-291. 634-641. [39] Yamamura K, Tani M, Hasegawa H, Gen W. [50] Chouchani ET, Pell VR, Gaude E, Aksentijević Very low dose of the Na+/Ca2+ exchange in- D, Sundier SY, Robb EL, Logan A, Nadtochiy hibitor, KB-R7943, protects ischemic reper- SM, Ord ENJ, Smith AC, Eyassu F, Shirley R, Hu fused aged Fischer 344 rat hearts: consider- able strain difference in the sensitivity to CH, Dare AJ, James AM, Rogatti S, Hartley RC, KB-R7943. Cardiovasc Res 2001; 523: 97- Eaton S, Costa ASH, Brookes PS, Davidson SM, 406. Duchen MR, Saeb-Parsy K, Shattock MJ, Rob- [40] McVeigh JJ, Lopaschuk GD. Dichloroacetate inson AJ, Work LM, Frezza C, Krieg T, Murphy stimulation of glucose oxidation improves re- MP. Ischaemic accumulation of succinate con- covery of ischemic rat hearts. Am J Physiol trols reperfusion injury through mitochondrial 1990; 259: 1079-1085. ROS. Nature 2014; 515: 431-437. [41] Taniguchi M, Wilson C, Hunter CA, Pehowich [51] Chouchani ET, Pell VR, James AM, Work LM, DJ, Clanachan AS, Lopaschuk GD. Dichloroac- Saeb-Parsy K, Frezza C, Krieg T, Murphy MP. A etate improves cardiac efficiency after isch- unifying mechanism for mitochondrial super- emia independent of changes in mitochond- oxide production during ischemia-reperfusion rial proton leak. Am J Physiol Heart Circ Physiol injury. Cell Metab 2016; 23: 254-263. 2001; 280: 1762-1769. [52] Ozpinar A, Weiner GM, Ducruet AF. Succinate: [42] Finegan BA, Lopaschuk GD, Coulson CS, Cla- a promising therapeutic target for reperfusion nachan AS. Adenosine alters glucose use dur- injury. Neurosurgery 2015; 77: 13-24. ing ischemia and reperfusion in isolated rat [53] O’Neill LA. : succinate strikes. Na- hearts. Circulation 1993; 87: 900-908. ture 2014; 515: 350-351. [43] Finegan BA, Lopaschuk GD, Gandhi M, Cla- [54] Murphy MP. How mitochondria produce reac- nachan AS. Inhibition of glycolysis and en- tive oxygen species. Biochem J 2009; 417: hanced mechanical function of working rat 1-13.

66 Int J Clin Exp Med 2018;11(1):59-68 Glycolysis and fatty acid β-oxidation disorder in IR

[55] St-Pierre J, Buckingham JA, Roebuck SJ, Brand DG. A novel domain in AMP activated protein MD. Topology of superoxide production from kinase causes glycogen storage bodies similar different sites in the mitochondrial electron to those seen in hereditary cardiac arrhyth- transport chain. J Biol Chem 2002; 277: mias. Curr Biol 2003; 13: 861-866. 44784-90. [66] Polekhina G, Gupta A, Michell BJ, van Den- [56] Chouchani ET, Methner C, Nadtochiy SM, Lo- deren B, Murthy S, Feil SC, Jennings IG, Camp- gan A, Pell VR, Ding S, James AM, Cochemé bell DJ, Witters LA, Parker MW, Kemp BE, Sta- HM, Reinhold J, Lilley KS, Partridge L, Fearnley pleton D. AMPK beta subunit targets metabolic IM, Robinson AJ, Hartley RC, Smith RA, Krieg T, stress sensing to glycogen. Curr Biol 2003; 13: Brookes PS, Murphy MP. Cardioprotection by S- 867-871. nitrosation of a cysteine switch on mitochon- [67] Young LH. A crystallized view of AMPK activa- drial complex I. Nat Med 2013; 19: 753-759. tion. Cell Metab 2009; 10: 5-6. [57] Ashrafian H, Czibik G, Bellahcene M, [68] Hardie DG. AMPK-Sensing energy while talking Aksentijević D, Smith AC, Mitchell SJ, Dodd to other signaling pathways. Cell Metab 2014; MS, Kirwan J, Byrne JJ, Ludwig C, Isackson H, 20: 939-952. Yavari A, Støttrup NB, Contractor H, Cahill TJ, [69] Zhang BB, Zhou G, Li C. AMPK: an emerging Sahgal N, Ball DR, Birkler RI, Hargreaves I, Ten- drug target for diabetes and the metabolic nant DA, Land J, Lygate CA, Johannsen M, syndrome. Cell Metab 2009; 9: 407-16. Kharbanda RK, Neubauer S, Redwood C, de [70] Mu J, Brozinick JT Jr, Valladares O, Bucan M, Cabo R, Ahmet I, Talan M, Günther UL, Robin- Birnbaum MJ. A role for AMP-activated protein son AJ, Viant MR, Pollard PJ, Tyler DJ, Watkins kinase in contraction-and hypoxiaregulated H. Fumarate is cardioprotective via activation glucose transport in . Mol Cell of the Nrf2 pathway. Cell Metab 2001; 7: 1085-1094. 2012; 15: 361-371. [71] Russell RR 3rd, Li J, Coven DL, Pypaert M, [58] Pell VR, Chouchani ET, Frezza C, Murphy MP, Zechner C, Palmeri M, Giordano FJ, Mu J, Birn- Krieg T. Succinate metabolism: a new thera- baum MJ, Young LH. AMP-activated protein ki- peutic target for myocardial reperfusion injury. nase mediates ischemic glucose uptake and Cardiovasc Res 2016; 111: 134-141. prevents postischemic cardiac dysfunction, [59] Scialò F, Sriram A, Fernández-Ayala D, Gubina apoptosis, and injury. J Clin Invest 2004; 114: N, Lõhmus M, Nelson G, Logan A, Cooper HM, 495-503. Navas P, Enríquez JA, Murphy MP, Sanz A. Mito- [72] Fujii N, Jessen N, Goodyear LJ. AMP-activated chondrial ROS produced via reverse electron protein kinase and the regulation of gluc- transport extend lifespan. Cell Metab ose transport. Am J Physiol Endocrinol Metab 2016; 23: 725-734. 2006; 291: E867-E877. [60] Mills EL, Kelly B, Logan A, Costa ASH, Varma M, [73] Samovski D, Su X, Xu Y, Abumrad NA, Stahl PD. Bryant CE, Tourlomousis P, Däbritz JHM, Gott- Insulin and AMPK regulate FA translocase/ lieb E, Latorre I, Corr SC, McManus G, Ryan D, CD36 plasma membrane recruitment in car- Jacobs HT, Szibor M, Xavier RJ, Braun T, Frezza diomyocytes via Rab GAP AS160 and Rab8a C, Murphy MP, O’Neill LA. Succinate dehydro- Rab GTPase. J Res 2012; 53: 709-717. genase supports metabolic repurposing of [74] Nishino Y, Miura T, Miki T, Sakamoto J, Naka- mitochondria to drive inflammatory macro- mura Y, Ikeda Y, Kobayashi H, Shimamoto K. phages. Cell 2016; 167: 457-470. Ischemic preconditioning activates AMPK in a [61] Zaha VG, Qi D, Su KN, Palmeri M, Lee HY, Hu X, PKC-dependent manner and induces GLUT4 Wu X, Shulman GI, Rabinovitch PS, Russell RR up-regulation in the late phase of cardioprotec- 3rd, Young LH. AMPK is critical for mitochon- tion. Cardiovasc Res 2004; 61: 610-619. drial function during reperfusion after myocar- [75] Li J, Hu X, Selvakumar P, Russell RR 3rd, Cush- dial ischemia. Journal of Molecular and Cellu- man SW, Holman GD, Young LH. Role of the lar Cardiology 2016; 91: 104-113. nitric oxide pathway in AMPK-mediated glu- [62] Jeon SM. Regulation and function of AMPK in cose uptake and GLUT4 translocation in heart physiology and diseases. Exp Mol Med 2016; muscle. Am J Physiol Endocrinol Metab 2004; 245: e245. 287: E834-E841. [63] Carling D. The AMP-activated protein kinase [76] Li J, Miller EJ, Ninomiya-Tsuji J, Russell RR 3rd, cascade-a unifying system for energy control. Young LH. AMP-activated protein kinase acti- Trends Biochem Sci 2004; 29: 18-24. vates p38 mitogen-activated protein kinase by [64] Hardie DG. The AMP-activated protein kinase increasing recruitment of p38 MAPK to TAB1 cascade: the key sensor of cellular energy sta- in the ischemic heart. Circ Res 2005; 97: 872- tus. Endocrinology 2003; 144: 5179-5183. 879. [65] Hudson ER, Pan DA, James J, Lucocq JM, Haw- [77] Nagendran J, Waller TJ, Dyck JR. AMPK signal- ley SA, Green KA, Baba O, Terashima T, Hardie ling and the control of substrate use in the

67 Int J Clin Exp Med 2018;11(1):59-68 Glycolysis and fatty acid β-oxidation disorder in IR

heart. Mol Cell Endocrinol 2013; 366: 180- [80] Folmes CD, Sowah D, Clanachan AS, Lo- 193. paschuk GD. High rates of residual fatty acid [78] Marsin AS, Bertrand L, Rider MH, Deprez J, oxidation during mild ischemia decrease car- Beauloye C, Vincent MF, Van den Berghe G, diac work and efficiency. J Mol Cell Cardiol Carling D, Hue L. Phosphorylation and activa- 2009; 47: 142-148. tion of heart PFK-2 by AMPK has a role in the stimulation of glycolysis during ischaemia. Curr Biol 2000; 101: 247-1255. [79] Lin J, Handschin C, Spiegelman BM. Metabolic control through the PGC-1 family of transcrip- tion coactivators. Cell Metab 2005; 1: 361- 370.

68 Int J Clin Exp Med 2018;11(1):59-68