The tortuous path of lactate shuttle discovery: From cinders and boards to the lab and ICU

George A Brooks, George A Brooks

Abstract

Once thought to be a waste product of oxygen limited (anaerobic) metabolism, lactate is now known to form continuously under fully oxygenated (aerobic) conditions. Lactate shuttling between producer (driver) and consumer cells fulfills at least 3 purposes; lactate is: (1) a major energy source, (2) the major gluconeogenic precursor, and (3) a signaling molecule. The Lactate Shuttle theory is applicable to diverse fields such as sports nutrition and hydration, resuscitation from acidosis and Dengue, treatment of traumatic brain injury, maintenance of glycemia, reduction of inflammation, cardiac support in heart failure and following a myocardial infarction, and to improve cognition. Yet, dysregulated lactate shuttling disrupts metabolic flexibility, and worse, supports oncogenesis. Lactate production in cancer (the Warburg effect) is involved in all main sequela for carcinogenesis: angiogenesis, immune escape, cell migration, metastasis, and self-sufficient metabolism. The history of the tortuous path of discovery in lactate metabolism and shuttling was discussed in the 2019 American College of Sports Medicine Joseph B. Wolffe Lecture in Orlando, FL.

Keywords: Anaerobic metabolism; Exercise; Glycolysis; Oxidative metabolism, Warburg Effect.

Copyright © 2020. Production and hosting by Elsevier B.V.

Figures

Graphical abstract
Graphical abstract
Fig. 1
Fig. 1
The lactate shuttle concept, depicting lactate as the vehicle linking glycolytic and oxidative metabolism. Linkages between lactate “producer” and “consumer” exist within and among cells, tissues, and organs. As the product of one metabolic pathway (glycolysis) and the substrate for a downstream pathway of disposal (mitochondrial respiration), lactate is the link between the glycolytic and aerobic pathways. Importantly, according to the lactate shuttle hypothesis, this linkage occurs continuously under fully aerobic conditions, can transcend compartment barriers and occur within and among cells, tissues and organs. Modified from Refs , , with permission.
Fig. 2
Fig. 2
Lactate production occurs continuously under fully aerobic conditions in intact animals, mammalian tissue preparations, intact animals, and humans in vivo. In muscles and arterial blood of resting healthy humans, lactate concentration approximates 1.0 mmol/L, while pyruvate concentration approximates 0.1 mmol/L. The lactate/pyruvate (L/P) approximates 10, with net lactate production and release from resting muscle of healthy individuals occurring when arterial partial pressure of oxygen (PO2) approximates 100 Torr and intramuscular PO2 approximates 40 Torr, well above the critical mitochondrial PO2 for maximal mitochondrial respiration (1–2 Torr)., , During exercise at about 65% of maximal oxygen consumption (VO2max), lactate production and net lactate release from working muscle beds rise and the L/P rises more than an order of magnitude (to approximately 500). However, the intramuscular PO2 remains at 3–4 Torr, well above the critical mitochondrial O2 level. Hence, it is appropriate to conclude that in healthy humans, glycolysis proceeds to lactate under fully aerobic conditions. Importantly, most (75%–80%) lactate is disposed of immediately within the tissue or subsequent to release and reuptake by working muscle, with significant uptake and oxidation by heart or oxidation by liver for gluconeogenesis. From diverse sources,,,,, with permission.
Fig. 3
Fig. 3
The Meyerhof calorimeter. m1 is the device for indirect electrical stimulation of frog hemicorpus thighs; the horizontal line (m2) is Ringers solution level. This device was used to demonstrate quantitative conversion of glycogen to lactate under nonperfused and nonoxygenated conditions. From Ref. 1 with permission.
Fig. 4
Fig. 4
Devices use to support the presence of lactate shuttling in resting and exercising mammals in vivo. These include (A) a motorized treadmill with sensors to determine oxygen consumption (VO2), rate of elimination of carbon dioxide (VCO2), electrocardiogram (ECG), RER (= VCO2/VO2) and (B) blood-specific activities of glucose, lactate glucose (Gluc), fructose di-phosphate (FDP), and other metabolic intermediates using 2-dimensional paper chromatography, autoradiography and enzymatic analyses. See original papers, for details on how lactate flux (production and disposal, oxidation and gluconeogenic) rates were determined in a mammalian model organism during physical exercise. From Ref. 23 with permission.
Fig. 5
Fig. 5
Depiction of the lactate shuttle as it fulfills 3 physiologic functions: (1) lactate as a major energy source, (2) lactate as the major gluconeogenic precursor, and (3) lactate as a signaling molecule with autocrine-, paracrine-, and endocrine-like effects (called a “lactormone”). “Cell–Cell” and “Intracellular Lactate Shuttle” concepts describe the roles of lactate in the delivery of oxidative and gluconeogenic substrates as well as in cell signaling. Examples of the Cell–Cell Lactate Shuttles include lactate exchanges between white glycolytic and red oxidative fibers within a working muscle bed and between working skeletal muscle and heart, brain, liver, and kidneys. Examples of Intracellular Lactate Shuttles include cytosol–mitochondrial and cytosol–peroxisome exchanges. Indeed, most, if not all, lactate shuttles are driven by a concentration or pH gradient or by redox state. FG = fast glycolytic; SO = slow oxidative. The figure is annotated because the original model did not anticipate cerebral lactate oxidation and hepatic and renal gluconeogenesis. Compiled from diverse sources, , and with permission.
Fig. 6
Fig. 6
A schematic showing the putative mitochondrial lactate oxidation complex (mLOC). The lactate-pyruvate transporter (MCT1) is inserted into the mitochondrial inner membrane, strongly interacting with its chaperone protein CD147, and is also associated with cytochrome oxidase (COx) as well as mitochondrial lactate dehydrogenase (mLDH), which could be located at the outer side of the inner membrane. Lactate, which is always produced in the cytosol of muscle and other tissues because of the abundance, activity, and characteristics of cytosolic LDH, is oxidized to pyruvate via the lactate oxidation complex in mitochondria of the same cell. This endergonic lactate oxidation reaction is coupled to the exergonic redox change in COx during mitochondrial electron transport. ETC = electron transport chain; GP = glycerol phosphate; Mal-Asp = malate-aspartate; MCT = monocarboxylate (lactate) transporter; mPC = mitochondrial pyruvate carrier; TCA = tricarboxylic acid. Redrawn from Ref. 75 with permission.
Fig. 7
Fig. 7
Immunohistochemical images demonstrating some components of the mitochondrial lactate oxidation complex (mLOC) in L6 cells. The mLOC contains the inner mitochondrial membrane cytochrome oxidase (COX), the lactate-pyruvate transporter (MCT1), lactate dehydrogenase (LDH) and the MCT anchoring protein, CD147 (Basigin). MCT1 was detected at both sarcolemmal and intracellular domains (A-1). Mitochondrial reticulum (MR), identified by MitoTracker, was extensively elaborated in L6 cells (A-2). The merged images of MCT1 (green, A-1) and MR (red, A-2) showed intense yellow, indicating co-localization of MCT1 and components of the MR, particularly at perinuclear cell domains (A-3). (B) LDH (B-1) and COX (B-2) are imaged. Superposition of signals for LDH (red, B-1) and COX (green, B-2) shows co-localization of LDH in the MR (yellow) of muscle cells (B-3). Depth of field approximately 1 μm, scale bar = 10 μm. Similar data have been obtained on rat plantaris leg muscles in vivo. From Ref. 75 with permission.
Fig. 8
Fig. 8
First images assessing co-localization of the monocarboxylate (lactate, pyruvate, β-hydroxybutyrate) transporter (MCT1) and mitochondrial the pyruvate carrier (mPC) in L6 cells, which shows the localization of DAPI-positive nuclei (A), MCT1 (B), mPC1 (C), and Mito Tracker-positive MR (D) in L6 cells. The merged images are shown in E. Co-localization analysis of mPC1 (C) and mitochondria (D) showed a Pearson correlation coefficient (r2) value of 0.8. Co-localization analysis of MCT1 (B) and mPC1 (C) showed an r2 of 0.3, largely because MCT1 occupies sarcolemmal, mitochondrial, and peroxisomal compartments. A channel to represent the co-localization of MCT1 and mitochondria was created to image mMCT1; subsequent co-localization of mMCT1 with mPC1 resulted in an r2 of 0.8 (F). White dots indicate the co-localization of mMCT1 and mPC1 as observed in Image J software. Whole images were contrast enhanced in A, B, C, D, and E. Similar results were observed for mPC2. Scale bar = 20 μm. It appears that both MCT1 and the putative mPC are co-localized to the mitochondria (r2 = 0.8). However, at the light microscopic level, it is impossible to know if the 2 proteins interact physically and functionally. Also, with benefit of the Orbitrap liquid chromatography/mass spectrometry device, we would be able to determine fractional synthesis rates of mitochondrial lactate oxidation complex and mPC proteins.
Fig. 9
Fig. 9
Illustration of how lactatemia affects blood (glucose) and peripheral glucose uptake as well as the production, uptake and oxidation of FFA, giving rise to metabolic inflexibility in muscle. Lactate is the inevitable consequence of glycolysis, the minimal muscle lactate (L) to pyruvate (P) ratio (L/P) being 10 and rising to an L/P of >100 when glycolytic flux is high. Lactate is the favored oxidizable substrate and provides product inhibition of glucose and FFA oxidation. As the products of glycolysis, lactate and pyruvate provide negative feedback inhibition of glucose disposal (blue dashed lines). Also, as the predominant mitochondrial substrate, lactate gives rise to acetyl-coenzyme A (CoA), and in turn malonyl-CoA. Acetyl-CoA inhibits β-ketothiolase and, hence, β-oxidation, while malonyl-CoA inhibits mitochondrial FFA-derivative uptake via CPT1 (T). Moreover, lactate is the main gluconeogenic precursor raising glucose production and blood (glucose) (red lines). Via GPR81 binding, lactate inhibits lipolysis in WAT (T), depressing circulating FFA., This model explains the paradoxical presence of lactatemia in high-intensity exercise and insulin-resistant states with limited ability to oxidize fat (green lines). Modified from. CPT1 = carnitine palmitoyl transporter-1; FAT = fatty acid translocator comprised of CD36 and FABPc; FFA = free fatty acid; GLUT = glucose transporter; m = mitochondrial; Malonyl = CoA formed from exported TCA citrate controlled by the interactions of malonyl-CoA decarboxylase (MCD) and acetyl-CoA carboxylase (ACC); MCT = monocarboxylate transporter; mPC = mitochondrial pyruvate transporter; PDH = pyruvate dehydrogenase; s = sarcolemmal; T = inhibition; WAT = white adipose tissue. Not shown is fatty acyl-Co (FA-CoA) that will accumulate if FFAs are taken up by myocytes, but blocked from mitochondrial entry by the effect of malonyl-CoA on CPT1. Accumulated intracellular FA-CoA will give rise to intramyocellular triglyceride (IMTG) and the formulation of LC-FA, DAG, and ceramides via inhibition of PI3 Kinase (PI3-K) and reducing GLUT4 translocation; from Ref. with permission.

References

    1. Meyerhof O. Uber's energy conversions in muscle II. The € Fate of the milk sour in the recovery period of the muscle (Über die Energieumwandlungen im Muskel II. Das Schicksal der Milchsaure in der Erholungsperiode des Muskels) Pflügers Archiv ges Physiol Mensch Tiere. 1920;182:284–317. [in German]
    1. Hill A.V. Muscular activity and carbohydrate metabolism. Science. 1924;60:505–514.
    1. Mitchell P. Coupling of phosphorylation to electron and hydrogen transfer by a chemi-osmotic type of mechanism. Nature. 1961;191:144–148.
    1. Chance B., Williams G.R. Respiratory enzymes in oxidative phosphorylation. VI. The effects of adenosine diphosphate on azide-treated mitochondria. J Biol Chem. 1956;221:477–489.
    1. Huxley H.E. Recent x-ray diffraction and electron microscope studies of striated muscle. J Gen Physiol. 1967;50(Suppl.):S71–83.
    1. Brooks G.A. Circulation, respiration, and metabolism: current comparative approaches. Springer; Berlin, Heidelberg: 1984. Glycolytic end product and oxidative substrate during sustained exercise in mammals–the “lactate shuttle; pp. 208–218.
    1. Brooks G.A. Lactate shuttles in nature. Biochem Soc Trans. 2002;30:258–264.
    1. Brooks G.A. Cell-cell and intracellular lactate shuttles. J Physiol. 2009;587:5591–5600.
    1. Bergman B.C., Wolfel E.E., Butterfield G.E., Lopaschuk G.D., Casazza G.A., Horning M.A. Active muscle and whole body lactate kinetics after endurance training in men. J Appl Physiol (1985) 1999;87:1684–1696.
    1. Gertz E.W., Wisneski J.A., Stanley W.C., Neese R.A. Myocardial substrate utilization during exercise in humans. Dual carbon-labeled carbohydrate isotope experiments. J Clin Invest. 1988;82:2017–2025.
    1. Glenn T.C., Martin N.A., Horning M.A., McArthur D.L., Hovda D.A., Vespa P. Lactate: brain fuel in human traumatic brain injury: a comparison to normal healthy control subjects. J Neurotrauma. 2015;32:820–832.
    1. Glenn T.C., Martin N.A., McArthur D.L., Hovda D.A., Vespa P., Johnson M.L. Endogenous nutritive support following traumatic brain injury: peripheral lactate production for glucose supply via gluconeogenesis. J Neurotrauma. 2015;32:811–819.
    1. Bergman B.C., Horning M.A., Casazza G.A., Wolfel E.E., Butterfield G.E., Brooks G.A. Endurance training increases gluconeogenesis during rest and exercise in men. Am J Physiol Endocrinol Metab. 2000;278:E244–E251.
    1. Hashimoto T., Hussien R., Oommen S., Gohil K., Brooks G.A. Lactate sensitive transcription factor network in L6 cells: activation of MCT1 and mitochondrial biogenesis. FASEB J. 2007;21:2602–2612.
    1. de Paoli F.V., Overgaard K., Pedersen T.H., Nielsen O.B. Additive protective effects of the addition of lactic acid and adrenaline on excitability and force in isolated rat skeletal muscle depressed by elevated extracellular K+ J Physiol. 2007;581:829–839.
    1. Robergs R.A., Ghiasvand F., Parker D. Biochemistry of exercise-induced metabolic acidosis. Am J Physiol Regul Integr Comp Physiol. 2004;287:R502–R516.
    1. Robergs R.A. Invited review: quantifying proton exchange from chemical reactions - implications for the biochemistry of metabolic acidosis. Comp Biochem Physiol A Mol Integr Physiol. 2019;235:29–45.
    1. Rogatzki M.J., Ferguson B.S., Goodwin M.L., Gladden L.B. Lactate is always the end product of glycolysis. Front Neurosci. 2015;9:22. doi: 10.3389/fnins.2015.00022.
    1. Brooks G.A., Martin N.A. Cerebral metabolism following traumatic brain injury: new discoveries with implications for treatment. Front Neurosci. 2015;8:408. doi: 10.3389/fnins.2014.00408.
    1. Garcia-Alvarez M., Marik P., Bellomo R. Stress hyperlactataemia: present understanding and controversy. Lancet Diabetes Endocrinol. 2014;2:339–347.
    1. Somasetia D.H., Setiati T.E., Sjahrodji A.M., Idjradinata P.S., Setiabudi D., Roth H. Early resuscitation of dengue shock syndrome in children with hyperosmolar sodium-lactate: a randomized single-blind clinical trial of efficacy and safety. Crit Care. 2014;18:466. doi: 10.1186/s13054-014-0466-4.
    1. Einstein A. Letter to Max Born. In: Born I., editor. The Born-Einstein letters. 1st ed. Walker and Company; New York, NY: 1926.
    1. Brooks G.A. The science and translation of lactate shuttle theory. Cell Metab. 2018;27:757–785.
    1. Hill A.V., Lupton H. Muscular exercise, lactic acid and the supply and utilization of oxygen. Q J Med. 1923;16:135–171.
    1. Wasserman K., McIlroy M.B. Detecting the threshold of anaerobic metabolism in cardiac patients during exercise. Am J Cardiol. 1964;14:844–852.
    1. Cori C.F., Cori G.T. Carbohydrate metabolism. Annu Rev Biochem. 1946;15:193–218.
    1. Warburg O., Wind F., Negelein E. The metabolism of tumors in the body. J Gen Physiol. 1927;8:519–530.
    1. Hussien R., Brooks G.A. Mitochondrial and plasma membrane lactate transporter and lactate dehydrogenase isoform expression in breast cancer cell lines. Physiol Genomics. 2011;43:255–264.
    1. Wasserman K., Beaver W.L., Davis J.A., Pu J.Z., Heber D., Whipp B.J. Lactate, pyruvate, and lactate-to-pyruvate ratio during exercise and recovery. J Appl Physiol (1985) 1985;59:935–940.
    1. Brooks G.A., Brauner K.E., Cassens R.G. Glycogen synthesis and metabolism of lactic acid after exercise. Am J Physiol. 1973;224:1162–1166.
    1. Brooks G.A., Gaesser G.A. End points of lactate and glucose metabolism after exhausting exercise. J Appl Physiol Respir Environ Exerc Physiol. 1980;49:1057–1069.
    1. Brooks G.A., White T.P. Determination of metabolic and heart rate responses of rats to treadmill exercise. J Appl Physiol Respir Environ Exerc Physiol. 1978;45:1009–1015.
    1. Brooks G.A., Bissell M.J., Bassham J.A. Ion-retardation desalting of blood and other animal tissues for separation of soluble metabolites by two-dimensional chromatography. Anal Biochem. 1977;83:580–588.
    1. Brooks G.A., Donovan C.M. Effect of endurance training on glucose kinetics during exercise. Am J Physiol. 1983;244:E505–E512.
    1. Donovan C.M., Brooks G.A. Endurance training affects lactate clearance, not lactate production. Am J Physiol. 1983;244:E83–E92.
    1. Emhoff C.A., Messonnier L.A., Horning M.A., Fattor J.A., Carlson T.J., Brooks G.A. Gluconeogenesis and hepatic glycogenolysis during exercise at the lactate threshold. J Appl Physiol (1985) 2013;114:297–306.
    1. Emhoff C.A., Messonnier L.A., Horning M.A., Fattor J.A., Carlson T.J., Brooks G.A. Direct and indirect lactate oxidation in trained and untrained men. J Appl Physiol (1985) 2013;115:829–838.
    1. Mazzeo R.S., Brooks G.A., Schoeller D.A., Budinger T.F. Disposal of blood [1-13C]lactate in humans during rest and exercise. J Appl Physiol (1985) 1986;60:232–241.
    1. Stanley W.C., Gertz E.W., Wisneski J.A., Morris D.L., Neese R.A., Brooks G.A. Systemic lactate kinetics during graded exercise in man. Am J Physiol. 1985;249:E595–E602.
    1. Bergman B.C., Butterfield G.E., Wolfel E.E., Lopaschuk G.D., Casazza G.A., Horning M.A. Muscle net glucose uptake and glucose kinetics after endurance training in men. Am J Physiol. 1999;277:E81–E92.
    1. Hooker A.M., Baldwin K.M. Substrate oxidation specificity in different types of mammalian muscle. Am J Physiol. 1979;236:C66–C69.
    1. Molé P.A., Baldwin K.M., Terjung R.L., Holloszy J.O. Enzymatic pathways of pyruvate metabolism in skeletal muscle: adaptations to exercise. Am J Physiol. 1973;224:50–54.
    1. Brooks G.A. Anaerobic threshold: review of the concept and directions for future research. Med Sci Sports Exerc. 1985;17:22–34.
    1. Brooks G.A. Lactate production under fully aerobic conditions: the lactate shuttle during rest and exercise. Fed Proc. 1986;45:2924–2929.
    1. Stanley W.C., Gertz E.W., Wisneski J.A., Neese R.A., Morris D.L., Brooks G.A. Lactate extraction during net lactate release in legs of humans during exercise. J Appl Physiol (1985) 1986;60:1116–1120.
    1. Gertz E.W., Wisneski J.A., Neese R., Bristow J.D., Searle G.L., Hanlon J.T. Myocardial lactate metabolism: evidence of lactate release during net chemical extraction in man. Circulation. 1981;63:1273–1279.
    1. Baldwin K.M., Campbell P.J., Cooke D.A. Glycogen, lactate, and alanine changes in muscle fiber types during graded exercise. J Appl Physiol. 1977;43:288–291.
    1. Barnard R.J., Edgerton V.R., Furukawa T., Peter J.B. Histochemical, biochemical, and contractile properties of red, white, and intermediate fibers. Am J Physiol. 1971;220:410–414.
    1. Peter J.B., Sawaki S., Barnard R.J., Edgerton V.R., Gillespie C.A. Lactate dehydrogenase isoenzymes: distribution in fast-twitch red, fast-twitch white, and slow-twitch intermediate fibers of guinea pig skeletal muscle. Arch Biochem Biophys. 1971;144:304–307.
    1. Johnson J.A., Fusaro R.M. The role of the skin in carbohydrate metabolism. Adv Metab Disord. 1972;60:1–55.
    1. Bergman B.C., Tsvetkova T., Lowes B., Wolfel E.E. Myocardial glucose and lactate metabolism during rest and atrial pacing in humans. J Physiol. 2009;587:2087–2099.
    1. Depocas F., Minaire Y., Chatonnet J. Rates of formation and oxidation of lactic acid in dogs at rest and during moderate exercise. Can J Physiol Pharmacol. 1969;47:603–610.
    1. Hochachka P.W. Cross-species studies of glycolytic function. Adv Exp Med Biol. 1999;474:219–229.
    1. Depocas F., Minaire Y., Chatonnet J. Rates of formation and oxidation of lactic acid in dogs at rest and during moderate exercise. Can J Physiol Pharmacol. 1969;47:603–610.
    1. Freminet A., Bursaux E., Poyart C.F. Effect of elevated lactataemia on the rates of lactate turnover and oxidation in rats. Pflugers Arch. 1974;346:75–86.
    1. Zinker B.A., Lacy D.B., Bracy D., Jacobs J., Wasserman D.H. Regulation of glucose uptake and metabolism by working muscle. An in vivo analysis. Diabetes. 1993;42:956–965.
    1. Henderson G.C., Horning M.A., Wallis G.A., Brooks G.A. Pyruvate metabolism in working human skeletal muscle. Am J Physiol Endocrinol Metab. 2007;292:E366. doi: 10.1152/ajpendo.00363.2006.
    1. Johnson M.L., Emhoff C.A., Horning M.A., Brooks G.A. Transpulmonary lactate shuttle. Am J Physiol Regul Integr Comp Physiol. 2012;302:R143–R149.
    1. Johnson M.L., Hussien R., Horning M.A., Brooks G.A. Transpulmonary pyruvate kinetics. Am J Physiol Regul Integr Comp Physiol. 2011;301:R769–R774.
    1. Brandt R.B., Laux J.E., Spainhour S.E., Kline E.S. Lactate dehydrogenase in rat mitochondria. Arch Biochem Biophys. 1987;259:412–422.
    1. Kline E.S., Brandt R.B., Laux J.E., Spainhour S.E., Higgins E.S., Rogers K.S. Localization of L-lactate dehydrogenase in mitochondria. Arch Biochem Biophys. 1986;246:673–680.
    1. Baba N., Sharma H.M. Histochemistry of lactic dehydrogenase in heart and pectoralis muscles of rat. J Cell Biol. 1971;51:621–635.
    1. De Bari L., Atlante A., Valenti D., Passarella S. Partial reconstruction of in vitro gluconeogenesis arising from mitochondrial l-lactate uptake/metabolism and oxaloacetate export via novel L-lactate translocators. Biochem J. 2004;380:231–242.
    1. Brooks G.A., Brown M.A., Butz C.E., Sicurello J.P., Dubouchaud H. Cardiac and skeletal muscle mitochondria have a monocarboxylate transporter MCT1. J Appl Physiol (1985) 1999;87:1713–1718.
    1. Passarella S., Paventi G., Pizzuto R. The mitochondrial L-lactate dehydrogenase affair. Front Neurosci. 2014;8:407. doi: 10.3389/fnins.2014.00407.
    1. Yoshida Y., Holloway G.P., Ljubicic V., Hatta H., Spriet L.L., Hood D.A. Negligible direct lactate oxidation in subsarcolemmal and intermyofibrillar mitochondria obtained from red and white rat skeletal muscle. J Physiol. 2007;582:1317–1335.
    1. Rasmussen H.N., van Hall G., Rasmussen U.F. Lactate dehydrogenase is not a mitochondrial enzyme in human and mouse vastus lateralis muscle. J Physiol. 2002;541:575–580.
    1. Sahlin K., Fernstrom M., Svensson M., Tonkonogi M. No evidence of an intracellular lactate shuttle in rat skeletal muscle. J Physiol. 2002;541:569–574.
    1. Kirkwood S.P., Munn E.A., Brooks G.A. Mitochondrial reticulum in limb skeletal muscle. Am J Physiol. 1986;251:C395–C402.
    1. Kirkwood S.P., Packer L., Brooks G.A. Effects of endurance training on a mitochondrial reticulum in limb skeletal muscle. Arch Biochem Biophys. 1987;255:80–88.
    1. Glancy B., Hartnell L.M., Combs C.A., Femnou A., Sun J., Murphy E. Power grid protection of the muscle mitochondrial reticulum. Cell Rep. 2018;23:2832. doi: 10.1016/j.celrep.2018.05.055.
    1. Jacobs R.A., Meinild A.K., Nordsborg N.B., Lundby C. Lactate oxidation in human skeletal muscle mitochondria. Am J Physiol Endocrinol Metab. 2013;304:E686–E694.
    1. Chen Y.J., Mahieu N.G., Huang X., Singh M., Crawford P.A., Johnson S.L. Lactate metabolism is associated with mammalian mitochondria. Nat Chem Biol. 2016;12:937–943.
    1. Park J.M., Josan S., Mayer D., Hurd R.E., Chung Y., Bendahan D. Hyperpolarized 13C NMR observation of lactate kinetics in skeletal muscle. J Exp Biol. 2015;218:3308–3318.
    1. Hashimoto T., Hussien R., Brooks G.A. Colocalization of MCT1, CD147, and LDH in mitochondrial inner membrane of L6 muscle cells: evidence of a mitochondrial lactate oxidation complex. Am J Physiol Endocrinol Metab. 2006;290:1237–1244.
    1. Hashimoto T., Hussien R., Cho H.S., Kaufer D., Brooks G.A. Evidence for the mitochondrial lactate oxidation complex in rat neurons: demonstration of an essential component of brain lactate shuttles. PloS one. 2008;3:e2915. doi: 10.1371/journal.pone.0002915.
    1. Pande S.V., Blanchaer M.C. Preferential loss of ATP-dependent long-chain fatty acid activating enzyme in mitochondria prepared using Nagarse. Biochim Biophys Acta. 1970;202:43–48.
    1. Butz C.E., McClelland G.B., Brooks G.A. MCT1 confirmed in rat striated muscle mitochondria. J Appl Physiol (1985) 2004;97:1059–1066.
    1. Hashimoto T., Masuda S., Taguchi S., Brooks G.A. Immunohistochemical analysis of MCT1, MCT2 and MCT4 expression in rat plantaris muscle. J Physiol. 2005;567:121–129.
    1. Pagliarini D.J., Calvo S.E., Chang B., Sheth S.A., Vafai S.B., Ong S.E. A mitochondrial protein compendium elucidates complex I disease biology. Cell. 2008;134:112–123.
    1. Calvo S.E., Clauser K.R., Mootha V.K. MitoCarta2.0: an updated inventory of mammalian mitochondrial proteins. Nucleic Acids Res. 2016;44:D1251–D1257.
    1. Chretien D., Pourrier M., Bourgeron T., Séné M., Rötig A., Munnich A. An improved spectrophotometric assay of pyruvate dehydrogenase in lactate dehydrogenase contaminated mitochondrial preparations from human skeletal muscle. Clin Chim Acta. 1995;240:129–136.
    1. Schurr A. Lactate: a major and crucial player in normal function of both muscle and brain. J Physiol. 2008;586:2665–2666.
    1. Atlante A., de Bari L., Bobba A., Marra E., Passarella S. Transport and metabolism of L-lactate occur in mitochondria from cerebellar granule cells and are modified in cells undergoing low potassium dependent apoptosis. Biochim Biophys Acta. 2007;1767:1285–1299.
    1. Sonveaux P., Végran F., Schroeder T., Wergin M.C., Verrax J., Rabbani Z.N. Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice. J Clin Invest. 2008;118:3930–3942.
    1. Roth D.A., Brooks G.A. Lactate transport is mediated by a membrane-bound carrier in rat skeletal muscle sarcolemmal vesicles. Arch Biochem Biophys. 1990;279:377–385.
    1. Roth D.A., Brooks G.A. Lactate and pyruvate transport is dominated by a pH gradient-sensitive carrier in rat skeletal muscle sarcolemmal vesicles. Arch Biochem Biophys. 1990;279:386–394.
    1. Herzig S., Raemy E., Montessuit S., Veuthey J.L., Zamboni N., Westermann B. Identification and functional expression of the mitochondrial pyruvate carrier. Science. 2012;337:93–96.
    1. Bricker D.K., Taylor E.B., Schell J.C., Orsak T., Boutron A., Chen Y.C. A mitochondrial pyruvate carrier required for pyruvate uptake in yeast, Drosophila, and humans. Science. 2012;337:96–100.
    1. Pellerin L., Pellegri G., Bittar P.G., Charnay Y., Bouras C., Martin J.L. Evidence supporting the existence of an activity-dependent astrocyte-neuron lactate shuttle. Dev Neurosci. 1998;20:291–299.
    1. Liu L., MacKenzie K.R., Putluri N., Maletic-Savatic M., Bellen H.J. The glia-neuron lactate shuttle and elevated ROS promote lipid synthesis in neurons and lipid droplet accumulation in glia via APOE/D. Cell Metabolism. 2017;27:719–737.
    1. Boussouar F., Benahmed M. Lactate and energy metabolism in male germ cells. Trends Endocrinol Metab. 2004;15:345–350.
    1. Storey B.T., Kayne F.J. Energy metabolism of spermatozoa. VI. Direct intramitochondrial lactate oxidation by rabbit sperm mitochondria. Biol Reprod. 1977;16:549–556.
    1. Ahmed K., Tunaru S., Tang C., Müller M., Gille A., Sassmann A. An autocrine lactate loop mediates insulin-dependent inhibition of lipolysis through GPR81. Cell Metab. 2010;11:311–319.
    1. Cai T.Q., Ren N., Jin L., Cheng K., Kash S., Chen R. Role of GPR81 in lactate-mediated reduction of adipose lipolysis. Biochem Biophys Res Commun. 2008;377:987–991.
    1. Liu C., Wu J., Zhu J., Kuei C., Yu J., Shelton J. Lactate inhibits lipolysis in fat cells through activation of an orphan G-protein-coupled receptor, GPR81. J Biol Chem. 2009;284:2811–2822.
    1. McClelland G.B., Khanna S., González G.F., Butz C.E., Brooks G.A. Peroxisomal membrane monocarboxylate transporters: evidence for a redox shuttle system? Biochem Biophys Res Commun. 2003;304:130–135.
    1. Brooks G.A., Mercier J. Balance of carbohydrate and lipid utilization during exercise: the “crossover” concept. J Appl Physiol (1985) 1994;76:2253–2261.
    1. Issekutz B., Jr, Miller H. Plasma free fatty acids during exercise and the effect of lactic acid. Proc Soc Exp Biol Med. 1962;110:237–239.
    1. Rodahl K., Miller H.I., Issekutz B., Jr Plasma free fatty acids in exercise. J Appl Physiol. 1964;19:489–492.
    1. Gold M., Miller H.I., Issekutz B., Jr., Spitzer J.J. Effect of exercise and lactic acid infusion on individual free fatty acids of plasma. Am J Physiol. 1963;205:902–904.
    1. Miller H.I., Issekutz B., Jr., Rodahl K., Paul P. Effect of lactic acid on plasma free fatty acids in pancreatectomized dogs. Am J Physiol. 1964;207:1226–1230.
    1. Ge H., Weiszmann J., Reagan J.D., Gupte J., Baribault H., Gyuris T. Elucidation of signaling and functional activities of an orphan GPCR, GPR81. J Lipid Res. 2008;49:797–803.
    1. Hoque R., Farooq A., Ghani A., Gorelick F., Mehal W.Z. Lactate reduces liver and pancreatic injury in Toll-like receptor- and inflammasome-mediated inflammation via GPR81-mediated suppression of innate immunity. Gastroenterology. 2014;146:1763–1774.
    1. Bergersen L.H. Lactate transport and signaling in the brain: potential therapeutic targets and roles in body-brain interaction. J Cereb Blood Flow Metab. 2015;35:176–185.
    1. Lauritzen K.H., Morland C., Puchades M., Holm-Hansen S., Hagelin E.M., Lauritzen F. Lactate receptor sites link neurotransmission, neurovascular coupling, and brain energy metabolism. Cereb Cortex. 2014;24:2784–2795.
    1. Takahashi H., Alves C.R.R., Stanford K.I., Middelbeek R.J.W., Nigro Pasquale, Ryan R.E. TGF-β2 is an exercise-induced adipokine that regulates glucose and fatty acid metabolism. Nat Metab. 2019;1:291–303.
    1. Friedman A., Kaufer D., Heinemann U. Blood-brain barrier breakdown-inducing astrocytic transformation: novel targets for the prevention of epilepsy. Epilepsy Res. 2009;85:142–149.
    1. Henderson G.C., Fattor J.A., Horning M.A., Faghihnia N., Johnson M.L., Mau T.L. Lipolysis and fatty acid metabolism in men and women during the postexercise recovery period. J Physiol. 2007;584:963–981.
    1. Wu B.U., Hwang J.Q., Gardner T.H., Repas K., Delee R., Yu S. Lactated Ringer's solution reduces systemic inflammation compared with saline in patients with acute pancreatitis. Clin Gastroenterol Hepatol. 2011;9:710.
    1. Carteron L., Solari D., Patet C., Quintard H., Miroz J.P., Bloch J. Hypertonic lactate to improve cerebral perfusion and glucose availability after acute brain injury. Crit Care Med. 2018;46:1649–1655.
    1. Oddo M., Vespa P., Menon D.K. Boosting the injured brain with supplemental energy fuels. Intensive Care Med. 2019;45:872–875.
    1. Patet C., Suys T., Carteron L., Oddo M. Cerebral lactate metabolism after traumatic brain injury. Curr Neurol Neurosci Rep. 2016;16:31. doi: 10.1007/s11910-016-0638-5.
    1. Warburg O., Minami S. Tests on over-living carcinoma tissue (Versuche an Überlebendem Carcinom-gewebe) Klinische Wochenschrift. 1923;2:776–777. [in German]
    1. Racker E. Bioenergetics and the problem of tumor growth. Am Sci. 1972;60:56–63.
    1. Hanahan D., Weinberg R.A. The hallmarks of cancer. Cell. 2000;100:57–70.
    1. Koppenol W.H., Bounds P.L., Dang C.V. Otto Warburg's contributions to current concepts of cancer metabolism. Nat Rev Cancer. 2011;11:325–337.
    1. Hensley C.T., Faubert B., Yuan Q., Lev-Cohain N., Jin E., Kim J. Metabolic heterogeneity in human lung tumors. Cell. 2016;164:681–694.
    1. San-Millan I., Brooks G.A. Reexamining cancer metabolism: lactate production for carcinogenesis could be the purpose and explanation of the Warburg Effect. Carcinogenesis. 2017;38:119–133.
    1. Goodwin M.L., Gladden L.B., Nijsten M.W., Jones K.B. Lactate and cancer: revisiting the Warburg effect in an era of lactate shuttling. Front Nutr. 2014;1:27. doi: 10.3389/fnut.2014.00027.
    1. Semenza G.L. Tumor metabolism: cancer cells give and take lactate. J Clin Invest. 2008;118:3835–3837.
    1. Doherty J.R., Yang C., Scott K.E., Cameron M.D., Fallahi M., Li W. Blocking lactate export by inhibiting the Myc target MCT1 disables glycolysis and glutathione synthesis. Cancer Res. 2014;74:908–920.
    1. Draoui N., Feron O. Lactate shuttles at a glance: from physiological paradigms to anti-cancer treatments. Dis Model Mech. 2011;4:727–732.
    1. Draoui N., Schicke O., Seront E., Bouzin C., Sonveaux P., Riant O. Antitumor activity of 7-aminocarboxycoumarin derivatives, a new class of potent inhibitors of lactate influx but not efflux. Mol Cancer Ther. 2014;13:1410–1418.
    1. Doherty J.R., Cleveland J.L. Targeting lactate metabolism for cancer therapeutics. J Clin Invest. 2013;123:3685–3692.
    1. Ovens M.J., Davies A.J., Wilson M.C., Murray C.M., Halestrap A.P. AR-C155858 is a potent inhibitor of monocarboxylate transporters MCT1 and MCT2 that binds to an intracellular site involving transmembrane helices 7–10. Biochem J. 2010;425:523–530.
    1. Baba M., Inoue M., Itoh K., Nishizawa Y. Blocking CD147 induces cell death in cancer cells through impairment of glycolytic energy metabolism. Biochem Biophys Res Commun. 2008;374:111–116.
    1. Schneiderhan W., Scheler M., Holzmann K.H., Marx M., Gschwend J.E., Bucholz M. CD147 silencing inhibits lactate transport and reduces malignant potential of pancreatic cancer cells in in vivo and in vitro models. Gut. 2009;58:1391–1398.
    1. Su J., Chen X., Kanekura T. A CD147-targeting siRNA inhibits the proliferation, invasiveness, and VEGF production of human malignant melanoma cells by down-regulating glycolysis. Cancer Lett. 2009;273:140–147.
    1. Zou W., Yang H., Hou X., Zhang W., Chen B., Xin X. Inhibition of CD147 gene expression via RNA interference reduces tumor cell invasion, tumorigenicity and increases chemosensitivity to paclitaxel in HO-8910pm cells. Cancer Lett. 2007;248:211–218.
    1. Powell K.E., King A.C., Buchner D.M., Campbell W.W., DiPietro L., Erickson K.I. The scientific foundation for the physical activity guidelines for Americans, 2nd ed. J Phys Act Health. 2018;16:1–11.
    1. Hultman E.A. Physiological role of muscle glycogen in man. Physiol Muscular Exerc. 1967:I99–I112.
    1. Meyer C., Dostou J.M., Welle S.L., Gerich J.E. Role of human liver, kidney, and skeletal muscle in postprandial glucose homeostasis. Am J Physiol Endocrinol Metab. 2002;282:E419–E427.
    1. Meyer C., Stumvoll M., Dostou J., Welle S., Haymond M., Gerich J. Renal substrate exchange and gluconeogenesis in normal postabsorptive humans. Am J Physiol Endocrinol Metab. 2002;282:E428–E434.
    1. Garcia C.K., Goldstein J.L., Pathak R.K., Anderson R.G., Brown M.S. Molecular characterization of a membrane transporter for lactate, pyruvate, and other monocarboxylates: implications for the Cori cycle. Cell. 1994;76:865–873.
    1. Price N.T., Jackson V.N., Halestrap A.P. Cloning and sequencing of four new mammalian monocarboxylate transporter (MCT) homologues confirms the existence of a transporter family with an ancient past. Biochem J. 1998;329:321–328.
    1. Pullen T.J., Khan A.M., Barton G., Butcher S.A., Sun G., Rutter G.A. Identification of genes selectively disallowed in the pancreatic islet. Islets. 2010;2:89–95.
    1. Rutter G.A., Pullen T.J., Hodson D.J., Martinez-Sanchez A. Pancreatic β-cell identity, glucose sensing and the control of insulin secretion. Biochem J. 2015;466:203–218.
    1. Bender K., Newsholme P., Brennan L., Maechler P. The importance of redox shuttles to pancreatic beta-cell energy metabolism and function. Biochem Soc Trans. 2006;34:811–814.
    1. Otonkoski T., Jiao H., Kaminen-Ahola N., Tapia-Paez I., Ullah M.S., Parton L.E. Physical exercise-induced hypoglycemia caused by failed silencing of monocarboxylate transporter 1 in pancreatic β cells. Am J Hum Genet. 2007;81:467–474.
    1. Casaburi R., Barstow T.J., Robinson T., Wasserman K. Influence of work rate on ventilatory and gas exchange kinetics. J Appl Physiol (1985) 1989;67:547–555.
    1. Messonnier A.L., Emhoff C.A., Fattor J.A., Horning M.A., J. T., Brooks G.A. Lactate kinetics at the lactate threshold in trained and untrained men. J Appl Physiol (1985) 2013;114:1593–1602.
    1. Ferguson B.S., Rogatzki M.J., Goodwin M.L., Kane D.A., Rightmire Z., Gladden L.B. Lactate metabolism: historical context, prior misinterpretations, and current understanding. Eur J Appl Physiol. 2018;118:691–728.
    1. Richardson R.S., Noyszewski E.A., Leigh J.S., Wagner P.D. Lactate efflux from exercising human skeletal muscle: role of intracellular PO2. J Appl Physiol (1985) 1998;85:627–634.
    1. Mazzeo R.S., Brooks G.A., Butterfield G.E., Podolin D.A., Wolfel E.E., Reeves J.T. Acclimatization to high altitude increase muscle sympathetic activity both at rest and during exercise. Am J Physiol. 1995;269:R201–R207.
    1. Mercier J.G., Hokanson J.F., Brooks G.A. Effects of cyclosporine A on skeletal muscle mitochondrial respiration and endurance time in rats. Am J Respir Crit Care Med. 1995;151:1532–1536.
    1. Hughes E.F., Turner S.C., Brooks G.A. Effects of glycogen depletion and pedaling speed on “anaerobic threshold”. J Appl Physiol Respir Environ Exerc Physiol. 1982;52:1598–1607.
    1. Brooks G.A., Butterfield G.E., Wolfe R.R., Groves B.M., Mazzeo R.S., Sutton J.R. Decreased reliance on lactate during exercise after acclimatization to 4,300 m. J Appl Physiol (1985) 1991;71:333–341.
    1. Gallagher C.N., Carpenter K.L., Grice P., Howe D.J., Mason A., Timofeev I. The human brain utilizes lactate via the tricarboxylic acid cycle: a 13C-labelled microdialysis and high-resolution nuclear magnetic resonance study. Brain. 2009;132:2839–2849.
    1. van Hall G., Str ø mstad M., Rasmussen P., Jans O., Zaar M., Gam C. Blood lactate is an important energy source for the human brain. J Cereb Blood Flow Metab. 2009;29:1121–1129.
    1. Smith D., Pernet A., Hallett W.A., Bingham E., Marsden P.K., Amiel S.A. Lactate: a preferred fuel for human brain metabolism in vivo. J Cereb Blood Flow Metab. 2003;23:658–664.
    1. Quistorff B., Secher N.H., Van Lieshout J.J. Lactate fuels the human brain during exercise. FASEB J. 2008;22:3443–3449.
    1. Schurr A. Lactate: the ultimate cerebral oxidative energy substrate? J Cereb Blood Flow Metab. 2006;26:142–152.
    1. Schurr A., Payne R.S. Lactate, not pyruvate, is neuronal aerobic glycolysis end product: an in vitro electrophysiological study. Neuroscience. 2007;147:613–619.
    1. Coco M., Caggia S., Musumeci G., Perciavalle V., Graziano A.C., Pannuzzo G. Sodium L-lactate differently affects brain-derived neurothrophic factor, inducible nitric oxide synthase, and heat shock protein 70 kDa production in human astrocytes and SH-SY5Y cultures. J Neurosci Res. 2013;91:313–320.
    1. Seifert T., Brassard P., Wissenberg M., Rasmussen P., Nordby P., Stallknecht B. Endurance training enhances BDNF release from the human brain. Am J Physiol Regul Integr Comp Physiol. 2010;298:R372–R377.
    1. Cotman C.W., Berchtold N.C. Exercise: a behavioral intervention to enhance brain health and plasticity. Trends Neurosci. 2002;25:295–301.
    1. Tsai S.J. Brain-derived neurotrophic factor: a bridge between major depression and Alzheimer's disease? Med Hypotheses. 2003;61:110–113.
    1. Krabbe K.S., Nielsen A.R., Krogh-Madsen R., Plomgaard P., Rasmussen P., Erikstrup C. Brain-derived neurotrophic factor (BDNF) and type 2 diabetes. Diabetologia. 2007;50:431–438.
    1. Holloway R., Zhou Z., Harvey H.B., Levasseur J.E., Rice A.C., Sun D. Effect of lactate therapy upon cognitive deficits after traumatic brain injury in the rat. Acta Neurochirurgica (Wien) 2007;149:919–927. discussion 927.
    1. Rice A.C., Zsoldos R., Chen T., Wilson M.S., Alessandri B., Hamm R.J. Lactate administration attenuates cognitive deficits following traumatic brain injury. Brain Res. 2002;928:156–159.
    1. Bisri T., Utomo B.A., Fuadi I. Exogenous lactate infusion improved neurocognitive function of patients with mild traumatic brain injury. Asian J Neurosurg. 2016;11:151–159.
    1. Hashimoto T., Tsukamoto H., Takenaka S., Olesen N.D., Petersen L.G., Sørensen H. Exercise improves brain executive function related to cerebral lactate metabolism in men. FASEB J. 2018;32:1417–1427.
    1. Wang J., Tu J., Cao B., Mu L., Yang X., Cong M. Astrocytic l-lactate signaling facilitates amygdala-anterior cingulate cortex synchrony and decision making in rats. Cell Rep. 2017;21:2407–2418.
    1. Schurr A., Gozal E. Aerobic production and utilization of lactate satisfy increased energy demands upon neuronal activation in hippocampal slices and provide neuroprotection against oxidative stress. Front Pharmacol. 2012;2:96. doi: 10.3389/fphar.2011.00096.
    1. Suzuki A., Stern S.A., Bozdagi O., Huntley G.W., Walker R.H., Magistretti P.J. Astrocyte-neuron lactate transport is required for long-term memory formation. Cell. 2011;144:810–823.
    1. Steinman M.Q., Gao V., Alberini C.M. The role of lactate-mediated metabolic coupling between astrocytes and neurons in long-term memory formation. Front Integr Neurosci. 2016;10:10. doi: 10.3389/fnint.2016.00010.
    1. Shapiro N.I., Howell M.D., Talmor D., Nathanson L.A., Lisbon A., Wolfe R.E. Serum lactate as a predictor of mortality in emergency department patients with infection. Ann Emerg Med. 2005;45:524–528.
    1. Zhang Z., Xu X. Lactate clearance is a useful biomarker for the prediction of all-cause mortality in critically ill patients: a systematic review and meta-analysis*. Crit Care Med. 2014;42:2118–2125.
    1. Messonnier L.A., Emhoff C.A., Fattor J.A., Horning M.A., Carlson T.J., Brooks G.A. Lactate kinetics at the lactate threshold in trained and untrained men. J Appl Physiol (1985) 2013;114:1593–1602.
    1. Rivers E., Nguyen B., Havstad S., Ressler J., Muzzin A., Knoblich B. Early goal-directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med. 2001;345:1368–1377.
    1. Miller B.F., Fattor J.A., Jacobs K.A., Horning M.A., Suh S.H., Navazio F. Metabolic and cardiorespiratory responses to “the lactate clamp”. Am J Physiol Endocrinol Metab. 2002;283:E889–E898.
    1. Marik P., Bellomo R. Lactate clearance as a target of therapy in sepsis: a flawed paradigm. OA Critical Care. 2013;1:3–5.
    1. Marik P., Bellomo R. A rational approach to fluid therapy in sepsis. Br J Anaesth. 2016;116:339–349.
    1. Nalos M., Leverve X., Huang S., Weisbrodt L., Parkin R., Seppelt I. Half-molar sodium lactate infusion improves cardiac performance in acute heart failure: a pilot randomised controlled clinical trial. Crit Care. 2014;18:R48. doi: 10.1186/cc13793.
    1. Sharma A.B., Barlow M.A., Yang S.H., Simpkins J.W., Mallet R.T. Pyruvate enhances neurological recovery following cardiopulmonary arrest and resuscitation. Resuscitation. 2008;76:108–119.
    1. Sharma A.B., Knott E.M., Bi J., Martinez R.R., Sun J., Mallet R.T. Pyruvate improves cardiac electromechanical and metabolic recovery from cardiopulmonary arrest and resuscitation. Resuscitation. 2005;66:71–81.
    1. Oddo M., Levine J.M., Frangos S., Maloney-Wilensky E., Carrera E., Daniel R.T. Brain lactate metabolism in humans with subarachnoid hemorrhage. Stroke. 2012;43:1418–1421.
    1. Wolahan S.M., Mao H.C., Real C., Vespa P.M., Glenn T.C. Lactate supplementation in severe traumatic brain injured adults by primed constant infusion of sodium L-lactate. J Neurosci Res. 2018;96:688–695.
    1. Azevedo J.L., Tietz E., Two-Feathers T., Paull J., Chapman K. Lactate, fructose and glucose oxidation profiles in sports drinks and the effect on exercise performance. PloS One. 2007;2:e927. doi: 10.1371/journal.pone.0000927.
    1. Hofmann P., Pokan R. Value of the application of the heart rate performance curve in sports. Int J Sports Physiol Perform. 2010;5:437–447.
    1. San-Millán I., Brooks G.A. Assessment of metabolic flexibility by means of measuring blood lactate, fat, and carbohydrate oxidation responses to exercise in professional endurance athletes and less-fit individuals. Sports Med. 2018;48:467–479.
    1. Horning MA, Brooks GA, Inventors. Formulation and methods to provide nutrition to human and other patients. US patent 20140120238. May 1, 2014.
    1. Horning MA, Brooks GA, Inventors. Formulations containing labels for medical diagnostics. US patent 09500657. Nov. 22, 2016.
    1. Horning MA, Brooks GA, Inventors. Systems and methods to estimate nutritional needs of human and other patients. US patent 8927490. Jan. 6, 2015.
    1. Mitchell J.H., Sproule B.J., Chapman C.B. The physiological meaning of the maximal oxygen intake test. J Clin Invest. 1958;37:538–547.
    1. Holloszy J.O. Biochemical adaptations in muscle. Effects of exercise on mitochondrial oxygen uptake and respiratory enzyme activity in skeletal muscle. J Biol Chem. 1967;242:2278–2282.
    1. Gladden L.B. Lactate metabolism: a new paradigm for the third millennium. J Physiol. 2004;558:5–30.
    1. Smuder A.J., Morton A.B., Hall S.E., Wiggs M.P., Ahn B., Wawrzyniak N.R. Effects of exercise preconditioning and HSP72 on diaphragm muscle function during mechanical ventilation. J Cachexia Sarcopenia Muscle. 2019;10:767–781.
    1. Dempsey J.A., Rankin J. Physiologic adaptations of gas transport systems to muscular work in health and disease. Am J Phys Med. 1967;46:582–647.
    1. Fahey T.D., Del Valle-Zuris A., Oehlsen G., Trieb M., Seymour J. Pubertal stage differences in hormonal and hematological responses to maximal exercise in males. J Appl Physiol Respir Environ Exerc Physiol. 1979;46:823–827.
    1. Rivera M.A., Fahey T.D. Association between aquaporin-1 and endurance performance: a systematic review. Sports Med Open. 2019;5:40. doi: 10.1186/s40798-019-0213-0.
    1. Fahey T.D., Insel P.M., Roth W.T. 13th ed. McGraw-Hill Higher Education; Mountain View, CA: 2019. Fit and well: core concepts and labs in physical fitness and wellness.
    1. Brooks G.A., Fahey T.D., Baldwin K.M. 5th ed. 1. Kindle Direct Publishing; Lexington, KY: 2019. (Exercise physiology: human bioenergetics and its applications).
    1. Rumsey W.L., Schlosser C., Nuutinen E.M., Robiolio M., Wilson D.F. Cellular energetics and the oxygen dependence of respiration in cardiac myocytes isolated from adult rat. J Biol Chem. 1990;265:15392–15402.
    1. Bendahan D., Chatel B., Jue T. Comparative NMR and NIRS analysis of oxygen dependent metabolism in exercising finger flexor muscles. Am J Physiol Regul Integr Comp Physiol. 2017;313:R740–R753.
    1. Connett R.J., Honig C.R., Gayeski T.E., Brooks G.A. Defining hypoxia: a systems view of VO2, glycolysis, energetics, and intracellular PO2. J Appl Physiol (1985) 1990;68:833–842.
    1. Brooks G.A., Fahey T.D., Baldwin K.M. 5th ed. 2. Kindle Direct Publishing; San Bernardino, CA: 2019. (Exercise physiology: human bioenergetics and its applications).
    1. Saddik M., Gamble J., Witters L.A., Lopaschuk G.D. Acetyl-CoA carboxylase regulation of fatty acid oxidation in the heart. J Biol Chem. 1993;268:25836–25845.

Source: PubMed

3
Subscribe