16
Targeted Metabolomics Connects Thioredoxin-interacting Protein (TXNIP) to Mitochondrial Fuel Selection and Regulation of Specific Oxidoreductase Enzymes in Skeletal Muscle * Received for publication, August 21, 2013, and in revised form, January 27, 2014 Published, JBC Papers in Press, January 30, 2014, DOI 10.1074/jbc.M113.511535 Karen L. DeBalsi ‡§ , Kari E. Wong , Timothy R. Koves ‡¶ , Dorothy H. Slentz , Sarah E. Seiler ‡§ , April H. Wittmann , Olga R. Ilkayeva , Robert D. Stevens , Christopher G. R. Perry , Daniel S. Lark , Simon T. Hui**, Luke Szweda ‡‡ , P. Darrell Neufer , and Deborah M. Muoio ‡§¶1 From the Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and § Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27704, the East Carolina Diabetes and Obesity Institute and Department of Physiology, East Carolina University, Greenville, North Carolina 27858, the **Department of Medicine, Division of Cardiology, University of California at Los Angeles, Los Angeles, California 90095, and the ‡‡ Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104 Background: Thioredoxin-interacting protein (TXNIP) is a redox sensor that opposes glucose uptake and glycolytic metabolism. Results: TXNIP-deficient skeletal muscles lose capacity for ketone and branched chain amino acid oxidation due to deficits in specific mitochondrial dehydrogenases. Conclusion: TXNIP permits muscle use of alternative respiratory fuels during glucose deprivation. Significance: Dysregulation of TXNIP might contribute to aberrant fuel selection in the context of metabolic disease. Thioredoxin-interacting protein (TXNIP) is an -arrestin family member involved in redox sensing and metabolic control. Growing evidence links TXNIP to mitochondrial function, but the molecular nature of this relationship has remained poorly defined. Herein, we employed targeted metabolomics and com- prehensive bioenergetic analyses to evaluate oxidative metabo- lism and respiratory kinetics in mouse models of total body (TKO) and skeletal muscle-specific (TXNIP SKM/ ) Txnip defi- ciency. Compared with littermate controls, both TKO and TXNIP SKM/ mice had reduced exercise tolerance in associa- tion with muscle-specific impairments in substrate oxidation. Oxidative insufficiencies in TXNIP null muscles were not due to perturbations in mitochondrial mass, the electron transport chain, or emission of reactive oxygen species. Instead, metabolic profiling analyses led to the discovery that TXNIP deficiency causes marked deficits in enzymes required for catabolism of branched chain amino acids, ketones, and lactate, along with more modest reductions in enzymes of -oxidation and the tricar- boxylic acid cycle. The decrements in enzyme activity were accom- panied by comparable deficits in protein abundance without changes in mRNA expression, implying dysregulation of protein synthesis or stability. Considering that TXNIP expression increases in response to starvation, diabetes, and exercise, these findings point to a novel role for TXNIP in coordinating mitochon- drial fuel switching in response to nutrient availability. Thioredoxin-interacting protein (TXNIP), 2 also known as vitamin D 3 up-regulated protein 1 (1) and thioredoxin-binding protein 2 (2), is a ubiquitously expressed 46-kDa protein that binds to the catalytic center of thioredoxin (TXN), an antioxi- dant protein that regulates the reversible oxidation/reduction of protein thiols/disulfides (3, 4). Thus, TXNIP serves as a neg- ative regulator of TXN activity and sensitizes the cell to oxida- tive stress (2). Structurally, TXNIP belongs to the -arrestin superfamily of proteins that are characterized by a two-lobe, immunoglobulin-like -strand sandwich motif (5). Recent findings suggest that at least some biological functions of TXNIP are mediated via the arrestin domain rather than its redox-sensitive cysteine residues (6). Evidence that TXNIP plays a critical role in metabolic regu- lation first came to light when the hyperlipidemic phenotype of the HcB-19/Dem (HcB-19) mouse strain was linked to a non- sense mutation in the Txnip gene, resulting in complete loss of the protein. In addition to hypertriglyceridemia, hypercholes- terolemia, and increased secretion of triglyceride-rich lipopro- teins (3), HcB-19 mice have abnormally high levels of free fatty acids and ketones during food restriction, suggesting perturba- tions in either the production or clearance of these fuels (3, 7–9). Many of the same metabolic features were recapitulated * This work was supported, in whole or in part, by National Institutes of Health Grants P01DK058398 (to D. M. M.) and R01DK080339 (to S. H.) from the United States Public Health Service and an American Heart Association Mid-Atlantic Affiliate Predoctoral Fellowship 10PRE4140033 (to K. L. D.). 1 To whom correspondence should be addressed: Independence Park Facil- ity, 4321 Medical Park Dr., Suite 200, Durham, NC 27704. Tel.: 919-479-2328; Fax: 919-477-0632; E-mail: [email protected]. 2 The abbreviations used are: TXNIP, thioredoxin interacting protein; BCAA, branched chain amino acid; BCKAD, branched chain ketoacid dehydro- genase; BDH1, -hydroxybutyrate dehydrogenase; EDL, extensor digito- rum longus; ETC, electron transport chain; ICD2, NADP -dependent isoci- trate dehydrogenase; ICD3, NAD -dependent isocitrate dehydrogenase; RER, respiratory exchange ratio; SCHAD, short chain hydroxyacyl-CoA dehydrogenase; TXN, thioredoxin; TKO, TXNIP whole body knockout mice; TXNIP SKM/ , skeletal muscle-specific knockout mice; TXNIP fl/fl , TXNIP floxed control mice; CPT, carnitine palmitoyltransferase; LDH, lactate dehydrogenase. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 289, NO. 12, pp. 8106 –8120, March 21, 2014 © 2014 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A. 8106 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 12 • MARCH 21, 2014 by guest on October 19, 2020 http://www.jbc.org/ Downloaded from

TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

Targeted Metabolomics Connects Thioredoxin-interactingProtein (TXNIP) to Mitochondrial Fuel Selection andRegulation of Specific Oxidoreductase Enzymes in SkeletalMuscle*

Received for publication, August 21, 2013, and in revised form, January 27, 2014 Published, JBC Papers in Press, January 30, 2014, DOI 10.1074/jbc.M113.511535

Karen L. DeBalsi‡§, Kari E. Wong‡, Timothy R. Koves‡¶, Dorothy H. Slentz‡, Sarah E. Seiler‡§, April H. Wittmann‡,Olga R. Ilkayeva‡, Robert D. Stevens‡, Christopher G. R. Perry�, Daniel S. Lark�, Simon T. Hui**, Luke Szweda‡‡,P. Darrell Neufer�, and Deborah M. Muoio‡§¶1

From the ‡Sarah W. Stedman Nutrition and Metabolism Center, Departments of ¶Medicine and §Pharmacology and CancerBiology, Duke University, Durham, North Carolina 27704, the �East Carolina Diabetes and Obesity Institute and Department ofPhysiology, East Carolina University, Greenville, North Carolina 27858, the **Department of Medicine, Division of Cardiology,University of California at Los Angeles, Los Angeles, California 90095, and the ‡‡Oklahoma Medical Research Foundation,Oklahoma City, Oklahoma 73104

Background: Thioredoxin-interacting protein (TXNIP) is a redox sensor that opposes glucose uptake and glycolyticmetabolism.Results: TXNIP-deficient skeletal muscles lose capacity for ketone and branched chain amino acid oxidation due to deficits inspecific mitochondrial dehydrogenases.Conclusion: TXNIP permits muscle use of alternative respiratory fuels during glucose deprivation.Significance: Dysregulation of TXNIP might contribute to aberrant fuel selection in the context of metabolic disease.

Thioredoxin-interacting protein (TXNIP) is an �-arrestinfamily member involved in redox sensing and metabolic control.Growing evidence links TXNIP to mitochondrial function, butthe molecular nature of this relationship has remained poorlydefined. Herein, we employed targeted metabolomics and com-prehensive bioenergetic analyses to evaluate oxidative metabo-lism and respiratory kinetics in mouse models of total body(TKO) and skeletal muscle-specific (TXNIPSKM�/�) Txnip defi-ciency. Compared with littermate controls, both TKO andTXNIPSKM�/� mice had reduced exercise tolerance in associa-tion with muscle-specific impairments in substrate oxidation.Oxidative insufficiencies in TXNIP null muscles were not due toperturbations in mitochondrial mass, the electron transportchain, or emission of reactive oxygen species. Instead, metabolicprofiling analyses led to the discovery that TXNIP deficiencycauses marked deficits in enzymes required for catabolism ofbranched chain amino acids, ketones, and lactate, along withmore modest reductions in enzymes of �-oxidation and the tricar-boxylic acid cycle. The decrements in enzyme activity were accom-panied by comparable deficits in protein abundance withoutchanges in mRNA expression, implying dysregulation of proteinsynthesis or stability. Considering that TXNIP expressionincreases in response to starvation, diabetes, and exercise, thesefindings point to a novel role for TXNIP in coordinating mitochon-drial fuel switching in response to nutrient availability.

Thioredoxin-interacting protein (TXNIP),2 also known asvitamin D3 up-regulated protein 1 (1) and thioredoxin-bindingprotein 2 (2), is a ubiquitously expressed 46-kDa protein thatbinds to the catalytic center of thioredoxin (TXN), an antioxi-dant protein that regulates the reversible oxidation/reductionof protein thiols/disulfides (3, 4). Thus, TXNIP serves as a neg-ative regulator of TXN activity and sensitizes the cell to oxida-tive stress (2). Structurally, TXNIP belongs to the �-arrestinsuperfamily of proteins that are characterized by a two-lobe,immunoglobulin-like �-strand sandwich motif (5). Recentfindings suggest that at least some biological functions ofTXNIP are mediated via the arrestin domain rather than itsredox-sensitive cysteine residues (6).

Evidence that TXNIP plays a critical role in metabolic regu-lation first came to light when the hyperlipidemic phenotype ofthe HcB-19/Dem (HcB-19) mouse strain was linked to a non-sense mutation in the Txnip gene, resulting in complete loss ofthe protein. In addition to hypertriglyceridemia, hypercholes-terolemia, and increased secretion of triglyceride-rich lipopro-teins (3), HcB-19 mice have abnormally high levels of free fattyacids and ketones during food restriction, suggesting perturba-tions in either the production or clearance of these fuels (3,7–9). Many of the same metabolic features were recapitulated

* This work was supported, in whole or in part, by National Institutes of HealthGrants P01DK058398 (to D. M. M.) and R01DK080339 (to S. H.) from theUnited States Public Health Service and an American Heart AssociationMid-Atlantic Affiliate Predoctoral Fellowship 10PRE4140033 (to K. L. D.).

1 To whom correspondence should be addressed: Independence Park Facil-ity, 4321 Medical Park Dr., Suite 200, Durham, NC 27704. Tel.: 919-479-2328;Fax: 919-477-0632; E-mail: [email protected].

2 The abbreviations used are: TXNIP, thioredoxin interacting protein; BCAA,branched chain amino acid; BCKAD, branched chain ketoacid dehydro-genase; BDH1, �-hydroxybutyrate dehydrogenase; EDL, extensor digito-rum longus; ETC, electron transport chain; ICD2, NADP�-dependent isoci-trate dehydrogenase; ICD3, NAD�-dependent isocitrate dehydrogenase;RER, respiratory exchange ratio; SCHAD, short chain hydroxyacyl-CoAdehydrogenase; TXN, thioredoxin; TKO, TXNIP whole body knockout mice;TXNIPSKM�/�, skeletal muscle-specific knockout mice; TXNIPfl/fl, TXNIPfloxed control mice; CPT, carnitine palmitoyltransferase; LDH, lactatedehydrogenase.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 289, NO. 12, pp. 8106 –8120, March 21, 2014© 2014 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A.

8106 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 12 • MARCH 21, 2014

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 2: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

in three additional mouse models with targeted disruptions inthe Txnip gene (10 –13).

In addition to regulating lipid metabolism, strong evidencehas established a central role for TXNIP in cellular glucosehomeostasis. Numerous studies in cultured cells have shownthat overexpression of TXNIP lowers glucose uptake, whereassilencing of the gene imparts the opposite effect (14). Likewise,each of the foregoing mouse models of TXNIP deficiency ischaracterized by fasting-induced hypoglycemia, improved glu-cose tolerance, enhanced insulin sensitivity, and increased glu-cose transport into peripheral tissues (10 –13). Notably, thisinsulin-sensitive phenotype is maintained even when TXNIPnull animals are fed a high fat diet (10, 11) or bred into theobese, ob/ob background (15), raising interest in this protein asa potential antidiabetic drug target.

The molecular mechanisms underlying the profound meta-bolic consequences of TXNIP deficiency appear to be highlycomplex and tissue dependent. Previous studies have impli-cated perturbations in pancreatic insulin secretion (8), hepaticglucose output (13), Akt signaling (11), glucose transporter 1trafficking (16), NLRP3 inflammasome activation (17), and oxi-dative stress (18). There is growing suspicion that at least someof these outcomes are secondary to mitochondrial dysfunctionand impaired oxidative metabolism. For example, oxidationrates of 14C-labeled glucose, fatty acids, and ketones are lowerin isolated skeletal muscles from TXNIP knock-out (TKO)mice compared with their littermate controls (11), and pyru-vate oxidation is disrupted in heart mitochondria from TKOmice (19). Most notably, in both cell culture and animal models,deletion of Txnip results in robust increases in lactate produc-tion (3, 9, 11, 13, 14, 19), a strong biomarker of oxidativeinsufficiency.

Whereas the foregoing observations have fueled widespreadspeculation surrounding a critical role for this protein in regu-lating mitochondrial function, rigorous experimental evidenceto support this presumption is sparse. The purpose of the pres-ent study was to further delineate the connection betweenTXNIP and oxidative metabolism in skeletal muscle and liver.To this end, we employed targeted metabolomics and compre-hensive bioenergetic analyses to evaluate oxidative metabolismand respiratory kinetics in mouse models of total body and skel-etal muscle-specific Txnip deficiency. In aggregate, our resultsshow that TXNIP is not required for normal functioning of theelectron transport chain (ETC), but is essential for maintainingskeletal muscle protein abundance of specific mitochondrialdehydrogenase enzymes that permit switching from glucose toalternative fuels. These findings shed new insight into the phys-iological function of TXNIP and the metabolic turmoil causedby its deficiency.

EXPERIMENTAL PROCEDURES

Animal Studies—Animal studies were approved by the DukeUniversity Institutional Animal Care and Use Committee. Gen-eration of the total body Txnip knock-out (TKO) and Txniphomozygous floxed control (TXNIPfl/fl) mice has been previ-ously described (11). Skeletal muscle-specific knock-out mice(TXNIPSKM�/�) were generated by breeding TXNIPfl/fl micewith transgenic mice expressing Cre recombinase under con-

trol of the mouse myogenin promoter and MEF2C (myo-CreTg/0), which express Cre recombinase exclusively in skeletalmuscle but not heart. All lines were backcrossed �6 genera-tions to the C57BL/6J background. Male mice were used for allstudies. Animals were housed in a temperature-controlledenvironment with a 12-h light:12-h dark cycle and allowed adlibitum access to standard chow (Purina Rodent Chow no.5015, Purina Mills, St. Louis, MO) and water. On the day ofsacrifice, food was removed from cages 5– 6 h prior to harvest-ing tissues and blood.

Metabolic Phenotype Parameters—For glucose tolerancetests, mice were fasted 6 h prior to an intraperitoneal injectionof 1.75 g/kg of glucose and blood glucose and lactate levels weremeasured from the tail using a glucometer (BD Medical, Frank-lin Lakes, NJ) and the Lactate Plus analyzer (Nova Biomedical,Waltham, MA). EDTA plasma collected at 9, 12, and 18 weeksof age were stored at �20 °C prior to analysis of triglycerides(Sigma, 337-B), non-esterified fatty acids (Wako Chemicals,Richmond, VA), and ketone bodies (acetone, acetoacetate, and�-hydroxybutyrate) (Hitachi 911 clinical chemistry analyzer).Plasma lactate assay was assayed spectrophotometrically (20) at340 nm using a SpectroMax spectrophotometer (MolecularDevices, Silicon Valley, CA).

Exercise Testing—Mice were habituated for three consecu-tive days prior to exercise testing by exposing them to a station-ary treadmill set at a 10% incline for 5 min, followed by 1-minintervals at speeds of 6, 9, and 12 m/min. The high-intensitytreadmill test began with a 5-min 0-m/min acclimation periodfollowed by a speed of 15 m/min, which was then increased atincrements of 3 m/min every 3 min to a maximal pace of 30m/min. Mice ran to exhaustion, defined as 10 consecutive secondson the shock grid. Peak VO2 and the respiratory exchange ratio(RER) were measured by indirect calorimetry using the Com-prehensive Laboratory Animal Monitoring System (CLAMS)attached to an enclosed treadmill (Columbus Instruments,Columbus, OH). Endurance exercise was conducted on an openfield six-lane treadmill set at a 10% incline. Following a 5-min 0m/min acclimation period, the speed was raised to 6 m/min andincreased by 2 m/min every 5 min until exhaustion.

Mitochondrial Isolation—Mitochondria from gastrocnemiusmuscles were isolated by differential centrifugation using one oftwo methods. For respiratory function and enzyme assays, weused a protease digestion method involving 2 min exposure to0.5 mg/ml of Type XXIV protease (Sigma) (20). Liver mito-chondria and muscle mitochondria used for Western blot anal-yses were harvested using a mechanical preparation devoid ofproteases. Tissues were homogenized in KMEM buffer (100mM KCl, 50 mM MOPS, 1 mM EGTA, 5 mM MgSO4, 1 mM ATP,pH, 7.4) with a Teflon pestle and centrifuged at 500 � g toremove cell debris. The supernatants were then centrifuged at10,000 � g to isolate mitochondria, followed by three slowercentrifugations (7,000 � g, 7,000 � g, and 3,500 � g) for wash-ing. The mitochondrial pellet was resuspended in 250 �l ofKMEM or SET buffer (250 mM sucrose, 1 mM EDTA, 10 mM

Tris-HCl, 2 mM ATP, pH 7.4). The protein concentration wasassayed using the BCA kit (Sigma).

Substrate Oxidation—Fatty acid and pyruvate oxidationrates were determined by measuring the production of

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

MARCH 21, 2014 • VOLUME 289 • NUMBER 12 JOURNAL OF BIOLOGICAL CHEMISTRY 8107

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 3: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

[14C]CO2 from either 0.625 �Ci/ml of [1-14C]palmitate (100�M palmitate bound to 0.15% BSA) or 0.625 �Ci/ml of[2-14C]pyruvate (1 mM pyruvate with 0.15% BSA) (21). Leucineoxidation rates were determined by measuring the productionof [14C]CO2 from 2.5 �Ci/ml of [U-14C]leucine (100 �M). Iso-lated mitochondria were resuspended to 0.8 �g/�l in SETbuffer and transferred to incubation buffer (pH 7.8) consistingof 62.5 mM sucrose, 12.5 mM potassium phosphate, 100 mM

potassium chloride, 1.25 mM magnesium chloride, 2.5 mM car-nitine, 0.25 mM malic acid, 1.25 mM DTT, 2 mM NAD�, 2 mM

ATP, 10 mM Tris-HCl, 0.5 mM CoA, 0.1 mM pyroxidal phos-phate, 0.4 mM thiamine pyrophosphate, and 0.5 mM �-keto-glutarate. Added to the final volume of incubation buffer wereappropriate volumes of lipoyl dehydrogenase (5 units/ml) andbranched-chain amino acid (BCAA) transaminase, where 1unit converts 1.0 �mol of leucine per minute to 2-ketoisocap-roate at 37 °C in the presence of �-ketoglutarate and pyrox-idal phosphate. The incubation buffer was kept in a shakingwater bath at 37 °C for at least 3 h before use to allow forconversion of the radiolabeled leucine to its labeled interme-diate, 2-ketoisocaproate.

Isolated Muscle Incubations—[1-14C]Palmitate oxidationand [U-14C]leucine oxidation in intact soleus and extensor digi-torum longus (EDL) muscles were conducted as described inRefs. 20 and 22. A similar procedure was used for measuring[U-14C]glucose oxidation in low calcium (Krebs Henseleit)KHB buffer containing 5 mM glucose, 0.5 mM carnitine, and12.5 mM HEPES � 100 nM insulin (23). Incubation media wascollected for the spectrophotometric assessment of lactate pro-duction via the reduction of NAD� at 340 nm.

Isolated Mitochondrial Oxygen Consumption—Mitochon-dria were resuspended at 0.5 mg/ml in MiR05 respiration media(24) using BD Oxygen Biosensor plates (BD Biosciences, MA)as described in Ref. 20.

Preparation of Permeabilized Muscle Fibers, MitochondrialRespiration, and Mitochondrial H2O2 emission in Permeabi-lized Fiber Bundles—These techniques were adapted from pre-vious methods (25, 26) and as described in Refs. 27 and 28.

Metabolic Profiling—Tissue and plasma metabolites weremeasured using tandem mass spectrometry (29).

Enzyme Activity Assays—Citrate synthase was assessed usinga colorimetric assay (20). Short chain hydroxyl-acyl-CoA dehy-drogenase (SCHAD) activity was measured for 10 min at 37 °Cin a reaction buffer (pH 6.5) containing 100 mM potassiumphosphate, 0.1 mM EDTA, and 0.3 mg/ml of essential fatty acid-free BSA using a modified protocol (30, 31). The rate of disap-pearance of 0.1 mM NADH at 340 nm upon the addition of 50�M acetoacetyl-CoA substrate was determined over the linearrange of the reaction and normalized to the molar extinctioncoefficient for NADH (6.22 mM�1 cm�1) and protein content.The �-hydroxybutyrate dehydrogenase (BDH1) activity assaywas adapted from Refs. 32 and 33 and measured the reductionof NAD� at 37 °C over 15 min following the addition of 20 �M

3-hydroxybutyrate to 200 �g of mitochondria in BDH1 activitybuffer (50 mM Tris-HCl, 2 mM NAD�, 20 mM sodium hydroxy-butyrate, 1 mM KCN, 0.4 �g/ml of rotenone, and 0.4 mg/mlBSA). Isocitrate dehydrogenase (ICD3) (NAD�-dependent)and ICD2 (NADP�-dependent) activities were measured over

10 min (340 nm, 30 °C) following the addition of a mixture of 10mM citrate and 1.5 mM isocitrate to 150 �g of mitochondria inpre-warmed (37 °C) 70 mM Tris-HCl, pH 7.2, 2 mM NAD� orNADP�, 2 mM ADP, 8 mM MgCl2, 1 mM MnCl2, and 5 �l/ml ofantimycin A (5% in EtOH) (34, 35). Succinate dehydrogenasewas assayed by first activating the enzyme by incubating 5–10�g of mitochondria in 100 mM potassium phosphate buffer (pH7.4) with 20 mM succinate for 5 min at 37 °C followed by theaddition of 2 mM KCN, 200 �M 2,6-dichloroindophenol, and 1.6mM phenazine methosulfate (35). Succinate dehydrogenasewas measured spectrophotometrically (600 nm, 30 °C) basedupon the reduction of 2,6-dichlorophenolindophenol (� � 21mM�1 cm�1) by phenazine methosulfate over 7 min. Branchedchain amino acid dehydrogenase (BCKAD) and pyruvate dehy-drogenase activity rates were determined by measuring the pro-duction of [14C]CO2 from either 1.25 �Ci/ml of [1-14C]leucine(100 �M) or 0.625 �Ci/ml of [1-14C]pyruvate (1 mM with 0.15%BSA), respectively, using the protocols for leucine and pyruvateoxidation in isolated mitochondria. Reduced (GSH) and oxi-dized (GSSG) glutathione were measured using the Biotech�GSH/GSSG-412 assay kit (OxisResearch, Portland, OR) opti-mized for tissues.

Analysis of mRNA Expression—Total RNA was isolated frommixed gastrocnemius muscle using the total RNA isolation kit(Qiagen). RNA quality and quantity were determined with aNanoDrop 8000 (Thermo Scientific). cDNA was synthesizedfrom 1 �g of RNA using the IScript cDNA synthesis kit (Bio-Rad) in a 20-�l reaction volume and diluted 5-fold for subse-quent RT-PCR. mRNA abundance was assayed by real-timequantitative PCR using a Prism 7000 with TaqMan real-timemaster mix and pre-designed/pre-validated FAM-labeledAssays-on-Demand (Applied Biosystems, Foster City, CA).Data were normalized using values from a duplexed reactionusing a VIC-labeled 18 S endogenous control (AppliedBiosystems).

Mitochondrial DNA Content—Total DNA was extractedfrom mixed gastrocnemius muscle using phenol/chloroform(25:4) followed by ethanol precipitation (36). The ratio of mito-chondrial DNA to nuclear DNA was measured using real-timequantitative PCR by calculating the ratio of the fold-changefrom the ��Ct of a mitochondrial encoded gene (COX1) and anuclear encoded gene (�-actin).

Protein Expression—Gastrocnemius muscle lysates were pre-pared using protease and phosphatase inhibitor mixtures and0.001 �g/�l of microcystin (Sigma). Protein concentrationswere determined using a BCA Assay (Sigma) and separatedon 4 –15% Criterion TGX Precast gels (Bio-Rad). Blots wereblocked with 0.45% fish gelatin for 1 h then exposed to pri-mary antibody to detect citrate synthase (Alpha DiagnosticsInternational, San Antonio, TX), oxidative phosphorylation(OXPHOS) complexes (MitoSciences), BDH1, ICD3 (Abcam,Cambridge, UK), BCKAD E1� subunit (generously provided byDr. Chris Lynch), and TXNIP (generously provided by Dr.Roger Davis). MemCode (Pierce) protein staining served as aloading control for each blot with band intensity being quanti-fied using ImageQuant software (Amersham Biosciences). Pro-teins were visualized with secondary antibodies labeled withIRDye infrared dyes using an Odyssey CLx Infrared Imaging

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

8108 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 12 • MARCH 21, 2014

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 4: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

System and Image Studio software (LI-COR Biosciences, Lin-coln, NE). The muscle (A) and heart (B) subunits of the lactatedehydrogenase (LDH) complex were measured colorimetri-cally by loading 30 �g of gastrocnemius muscle protein onto a7.5% native polyacrylamide gel (Bio-Rad). Following electro-phoresis, the gel was stained in 30 ml of staining solution contain-ing 0.1 M sodium lactate, 1.5 mM NAD, 0.1 M Tris-HCl (pH 8.6), 10mM NaCl, 5 mM MgCl2, 0.03 mg/ml of phenazine methosulfate,and 0.25 mg/ml nitro blue tetrazolium. Band intensity was quan-tified using Image Lab 4.1 software (Bio-Rad).

Statistical Analysis—Data are expressed as mean � S.E.Results were analyzed by Student’s t test unless otherwise indi-cated in the figure legends. A p value less than or equal to 0.05was considered statistically significant.

RESULTS

TXNIP Deficiency Disrupts Whole Body Energy Metabolism—Previous studies examining the metabolic consequences oftotal body Txnip deficiency were performed in the context ofprolonged food deprivation (18 –36 h) (3, 9, 11, 12). To avoidthe complexities of the murine starvation response, we exam-ined metabolic regulation during a 5– 6-h fast. In the morningafter overnight ad libitum feeding and throughout 6 h of foodwithdrawal, Txnip knock-out (TKO) mice had lower blood glu-cose levels and higher blood lactate (Fig. 1A). In line with earlierreports, when challenged with an intraperitoneal injection ofglucose TKO mice exhibited exquisite glucose tolerance (Fig.1B), with a trend toward lower insulin levels (Fig. 1C) despiteincreased body weight (Fig. 1D) and higher circulating levels oflipid-derived fuels such as triglycerides, non-esterified fattyacids, and ketones (Fig. 1E). Because TXNIP is expressed abun-dantly in skeletal muscle (Fig. 1F), the principal contributor tocirculating lactate, we proceeded to examine muscle glucosemetabolism ex vivo. In isolated soleus muscles from TKO com-pared with WT mice, lactate production was increased in boththe basal and insulin-stimulated states, whereas glycogen syn-thesis was elevated only in the latter condition (Fig. 1, G and H).Using the Lox-P/Cre strategy described under “ExperimentalProcedures,” we generated mice lacking Txnip specifically inskeletal muscle (TXNIPSKM�/�) but retaining expression inheart (Fig. 1I). The metabolic phenotype of these mice wasremarkably similar to the TKO line (Fig. 1, J–M), suggestingderangements in skeletal muscle energy metabolism contributesubstantially to the phenotype of the TKO model. In summary,these findings are consistent with the idea that genetic ablation ofTXNIP promotes glucose uptake and glycolytic metabolism inmuscle.

TXNIP Deficiency Compromises Exercise Tolerance—Thehigher glycolytic rates in TKO mice could be due to oxidativedeficits, as previously suggested (11). Surprisingly, however,when whole body energy metabolism was monitored by indi-rect calorimetry during a 72-h period we were unable to detectgenotype differences in RER, heat production, and oxygen con-sumption (data not shown). Physical activity and food con-sumption were also similar between groups. To further explorethe connection between TXNIP and oxidative capacity in thecontext of increased energetic demand we challenged the micewith two distinct exercise tests. During a graded, high intensity

regimen, peak VO2 obtained by TKO mice was 13% lower thanthat measured in their WT counterparts (Fig. 2A), whereas RERwas elevated throughout the test, indicative of a substrate shifttoward carbohydrate oxidation (Fig. 2B). Because fatty acid oxi-dation becomes an essential source of energy during prolongedexercise, we next subjected the animals to a graded, mid-inten-sity endurance run using an open-air treadmill. Exercise endur-ance was diminished in TKO mice relative to the WT controls,evidenced by reduced time (78.0 � 2.19 versus 84.8 � 1.28 min)and distance (1774 � 81.8 versus 2035 � 52.3 m) to exhaustion(Fig. 2, C and D). Circulating levels of �-hydroxybutyrate mea-sured immediately after exercise were elevated in TKO mice,consistent with impaired ketone uptake and/or catabolism (Fig.2E). Importantly, TXNIPSKM�/� mice exhibited a similar exer-cise intolerant phenotype during both treadmill tests (Fig. 2,F–J), again implicating muscle as a major site of metabolic dys-regulation in the TKO mice. Although these results clearlypoint to a role for TXNIP in regulating muscle substrate selec-tion, both loss-of-function models maintained considerableoxidative reserve, arguing against a severe defect in mitochon-drial respiratory function.

TXNIP Deficiency Diminishes Substrate Oxidation in Musclebut Not Liver Mitochondria—We next sought to pinpoint thespecific sites of oxidative dysfunction in the TXNIP null condi-tion. Because TXNIP has been implicated in regulating energymetabolism in both skeletal muscle and liver, TKO mice wereused to permit direct comparisons between these two tissues.Measurement of substrate oxidation in isolated soleus and EDLmuscles revealed marked genotype-dependent deficits in glu-cose, fatty acid, and amino acid catabolism (Fig. 3, A–C). Nota-bly, whereas TXNIP deficiency lowered glucose and fatty acidoxidation 25–30% in both soleus and EDL, oxidation rates ofthe BCAA, leucine, were diminished 68% in soleus and 87% inEDL, as compared with WT muscles. Experiments in isolatedmitochondria from TKO gastrocnemius muscles producedsimilar results, again showing moderate declines in pyruvateand palmitate oxidation (Fig. 3, D and E) and a much morestriking 75% deficit in leucine oxidation (Fig. 3F). By contrast,oxidation rates of all three substrates were normal in liver mito-chondria harvested from TKO compared with WT mice (Fig. 3,G–I). Thus metabolic insufficiencies measured at the wholebody level appeared to be due in large part to remodeling of themuscle oxidative machinery.

TXNIP Deficiency Does Not Affect Mitochondrial Content orRespiratory Function—We questioned whether the foregoingdeficits in oxidative metabolism were related to changes inmitochondrial mass and/or respiratory function. Contrary tothis possibility, muscle quantities of mitochondrial DNA, cit-rate synthase protein content, and enzyme activity (not shown),as well as protein abundance of several constituents of the ETC(Fig. 3, J and K), were similar between genotypes. Comprehen-sive evaluation of respiratory function using isolated mitochon-dria also proved unremarkable (not shown). More rigorousassessment of respiratory kinetics using permeabilized fiberbundles in combination with a high resolution respirometrysystem revealed only modest abnormalities in ETC function(Fig. 3L). ADP-stimulated Vmax in the presence of saturatingglutamate-malate concentrations was 20% lower in the TKO

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

MARCH 21, 2014 • VOLUME 289 • NUMBER 12 JOURNAL OF BIOLOGICAL CHEMISTRY 8109

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 5: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

mice, implying a minor deficit in maximal rates of ATP synthe-sis at Complex V. When substrate kinetics were analyzed atmore physiological, submaximal concentrations, we identifieda subtle shift toward increased sensitivity (lower Km) to gluta-mate (Fig. 3M), suggesting enhanced use of this fuel to provide

NADH to Complex I. These findings argue against a majorimpingement at the level of the ETC.

TXNIP Deficiency Disrupts Redox Balance without AffectingMitochondrial Reactive Oxygen Species Emission—BecauseTXNIP is best known for its role in redox regulation, we sought

FIGURE 1. TXNIP deficiency alters whole body energy metabolism. TKO mice and WT littermates were fed a standard chow diet and metabolic parameterswere measured at 18 –22 weeks unless otherwise noted. A, blood glucose and lactate levels were measured 0 – 6 h following food withdrawal (n � 6). B, glucosetolerance tests were performed at 12 weeks of age using 1.75 g/kg of glucose administered 6 h after food withdrawal (n � 12). C, serum insulin was measuredat 0 and 15 min. D, body weight at 22 weeks. E, plasma triglycerides (TAG), non-esterified fatty acids (NEFA), and �-hydroxybutyrate (BHB) were measured 5 hafter food withdrawal at 9 –12 weeks of age (n � 6 – 8). F, TXNIP mRNA tissue distribution was measured by real-time quantitative PCR and normalized to 18Sas an endogenous control gene (n � 5). Lactate production (G) and rates of [14C]glucose (H) incorporation into glycogen were measured in isolated soleusmuscles incubated 1 h � 100 nM insulin. I, TXNIP mRNA expression in skeletal muscle and heart from TXNIPSKM�/� and TXNIPfl/fl mice (n � 3). Metabolicparameters of TXNIPSKM�/� mice and TXNIPfl/fl littermate controls were measured at 12–14 weeks of age. J, blood glucose and lactate levels (n � 5). K, glucosetolerance tests (n � 6). L, body weight at sacrifice. M, plasma metabolites (n � 9). Data are mean � S.E. and results were analyzed by Student’s t test (*, p � 0.05and **, p � 0.001).

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

8110 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 12 • MARCH 21, 2014

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 6: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

to determine whether the unique form of mitochondrial dys-function in muscles from the TKO mice might be related tochanges in redox balance. To this end, we measured a series ofmetabolites that report on shifts in cellular energy charge andoxidative stress. Interestingly, despite high rates of glycolysisand lactate production, muscle concentrations of NADH (Fig.4A) were unchanged in the TKO mice. By contrast, levels ofNADPH, which can also arise from glucose metabolism via thepentose-phosphate shunt, were elevated 2.5-fold in the TKOmice (Fig. 4B), resulting in a striking decrease in the NADH/NADPH ratio (Fig. 4C). Conversely, TXNIP deficiency resultedin a 25% decline in muscle concentrations of reduced GSH and

a 33% increase in the oxidized form, GSSG (Fig. 4, D–F).Because GSH participates in antioxidant defense, we questionedwhether the alterations in redox balance in TKO muscles wererelated to increased mitochondrial production of reactive oxygenspecies. On the contrary, when assayed in permeabilized fiber bun-dles, rates of succinate-supported mitochondrial H2O2 emissiontrended lower in the TKO group (Fig. 4G). Thus, the metabolicdefects in TKO muscles did not appear to result from mitochon-drial-derived oxidative stress.

Targeted Metabolomics Revealed Several Muscle-specificRoadblocks in Mitochondrial Carbon Flux—To gain furtherinsight into the oxidative lesions arising from TXNIP deficiency

FIGURE 2. TXNIP deficiency decreases exercise tolerance. TKO mice and WT littermates were subjected to two different treadmill exercises at 12–15 weeksof age. Indirect calorimetry was used to measure peak VO2 (A) and the RER (VCO2/VO2) (B) as a function of workload during a graded, high intensity regimen.Exercise endurance was evaluated using an open-air treadmill and a mid-intensity regimen. Performance was assessed by measuring time (min) (C) anddistance (meters) (D) to exhaustion. E, plasma �-hydroxybutyrate was measured prior to and immediately after endurance exercise. Data are expressed asmean � S.E. from 7 to 8 animals per group. The same tests were administered to TXNIPSKM�/� mice and TXNIPfl/fl littermate controls at 22–24 weeks of age. F,peak VO2. G, RER. H, time; and I, distance to exhaustion. J, plasma �-hydroxybutyrate concentrations. Data are expressed as mean � S.E. from 5 to 7 animals pergroup and results were analyzed by Student’s t test (*, p � 0.05 and **, p � 0.001). Panels B and G were evaluated using a one-factor analysis of variance and posthoc analysis to determine differences between groups.

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

MARCH 21, 2014 • VOLUME 289 • NUMBER 12 JOURNAL OF BIOLOGICAL CHEMISTRY 8111

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 7: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

we used a targeted metabolomics approach to capture a snap-shot of intermediary metabolism in vivo. Because mitochon-drial degradation of carbon fuels results in the production ofmultiple acyl-CoA intermediates, we expected muscle quanti-ties of these molecules to change in association with TXNIP

expression. A survey of acyl-CoAs in gastrocnemius musclefrom TKO compared with WT mice revealed a 35% reductionin acetyl-CoA (Fig. 5A), the universal end product of carbohy-drate, fat, and protein catabolism. Additionally, the leucine-derived metabolite, isovaleryl-CoA, was 65% lower in TKO

FIGURE 3. Substrate oxidation and respiratory function in skeletal muscle and liver mitochondria. Substrate oxidation assays were performed in tissuesand mitochondria from TKO mice and WT littermates harvested 5– 6 h after food withdrawal. A, oxidation of [U-14C]glucose to CO2 was measured in isolatedsoleus muscles during a 2-h incubation in modified KHB buffer containing 5 mM glucose � 100 nM insulin. Oxidation of 500 �M [1-14C]palmitate (B) or 200 �M

[U-14C]leucine (C) to CO2 was measured in isolated soleus and EDL muscles during a 2-h incubation in modified KHB (n � 6). Isolated mitochondria fromgastrocnemius muscles were used to measure substrate oxidation to CO2 in the presence of 1 mM [2-14C]pyruvate (D), 100 �M [1-14C]palmitate (E), or 100 �M

[U-14C]leucine (F) for 30 min (n � 11–12). Isolated mitochondria from liver was used to measure substrate oxidation to CO2 in the presence of 1 mM [2-14C]py-ruvate (G), 100 �M [1-14C]palmitate (H), or 100 �M [U-14C]leucine (I) for 30 min (n � 6 –12). Gastrocnemius muscles from TKO mice and WT littermate controlswere used for Western blot of complexes I-V of the ETC (J) and quantified results (K) were normalized to MemCode staining to control for loading. Respiratoryfunction in situ was measured using permeabilized fiber bundles under state 3 respiration conditions in the presence of saturating (L) or increasing (M) substrateconcentrations to determine substrate sensitivity (Km). Data are expressed as mean � S.E. and results were analyzed by Student’s t test (*, p � 0.05 and **, p � 0.001).

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

8112 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 12 • MARCH 21, 2014

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 8: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

mice (Fig. 5A), and a similar trend was evident for several evenchain, fatty acid-derived species (Fig. 5A). By virtue of the mito-chondrial carnitine acyltransferase enzymes, the foregoingacyl-CoA intermediates give rise to their corresponding acyl-carnitine conjugates, which also report on substrate flux at spe-cific mitochondrial enzymes. Whereas muscle levels of acetyl-CoA-derived acetylcarnitine (C2) were unaffected by genotype(Fig. 5B), most odd chain/amino acid-derived acylcarnitines, aswell as several medium and long chain/fatty acid-derived spe-cies, were reduced with TXNIP deficiency (Fig. 5B). Long chainacylcarnitines are produced by the carnitine palmitoyltrans-ferase enzymes (CPT1 and CPT2), which reside in the outer andinner mitochondrial membranes, respectively, and catalyze theinitial steps in �-oxidation. Although a reduction in total longchain acylcarnitines (Fig. 5B) is consistent with diminishedCPT1 activity, tissue concentrations of malonyl-CoA, a potentglucose-derived inhibitor of CPT1 (37), were similar betweengenotypes (Fig. 5A). Therefore, reduced lipid catabolism inTKO mice did not appear to result from malonyl-CoA-medi-ated inhibition of fat oxidation.

In contrast to many other even chain species, levels of 3-hy-droxybutyrylcarnitine (C4OH) were robustly elevated in themuscles of TKO mice (Fig. 5B). Because the C4OH metabolitecan arise from the ketone intermediate, 3-hydroxybutyryl-CoA(38), this finding is consistent with a severe bottleneck in ketol-ysis. Muscle concentrations of several glucose-derived organicacids and amino acids (including glycine, alanine, and lactate)were elevated in the TXNIP null condition, whereas intermedi-ates directly involved in anaplerotic entry of carbons into thetricarboxylic acid (TCA) cycle (such as aspartate, glutamate,�-ketoglutarate, and fumarate) were diminished (Fig. 5, C andD). These results are consistent with perturbations in glycolysisand TCA cycle flux and might reflect increased traffickingthrough the malate/aspartate and glutamate/aspartate shuttles.Although muscle concentrations of the BCAAs (valine, leucine,and isoleucine) were similar between groups (Fig. 5D), circulat-

ing levels of these metabolites were elevated in TKO mice (Fig.5E), consistent with a defect in BCAA catabolism. Whereas themetabolite signature in skeletal muscle was clearly distinguish-able between genotypes, the only differentially abundantmetabolites in TKO liver were the lower levels of long chainacylcarnitines (Fig. 6, A–C), which again occurred independentof changes in malonyl-CoA (not shown). In aggregate, theseresults add further evidence that TXNIP plays a key role inmaintaining oxidative metabolism at sites upstream of the ETC.

TXNIP Deficiency Results in Muscle-specific Deficits in Mul-tiple Dehydrogenase Enzymes—The foregoing static metabolicprofiles (Fig. 5, A–E), taken together with the substrate oxida-tion presented in Fig. 3, A–F, pointed toward several specificmitochondrial oxidoreductase enzymes responsible for degrad-ing BCAAs, ketones, and fatty acids, including BCKAD, BDH1,and SCHAD. As predicted, the activities of these three enzymeswere diminished 65, 47, and 15%, respectively, in mitochon-dria from TKO compared with WT muscles (Table 1). Like-wise, activity of the NAD-dependent TCA cycle enzyme,ICD3, was 27% lower in muscles from TXNIP KO comparedwith WT mice (Table 1). The impact of TXNIP deficiency onmitochondrial dehydrogenase enzymes was not universal, asthe activities of succinate dehydrogenase (SDH), pyruvatedehydrogenase (PDH), and NADPH�-dependent ICD2,were similar between genotypes.

We proceeded to determine whether the decline in the afore-mentioned enzyme activities was related to changes in geneand/or protein expression. Whereas mRNA expression of theseenzymes was similar between TKO and WT mice (Fig. 7A),Western blot analyses revealed marked deficits at the level ofprotein expression, as quantities of the BCKAD E1� subunit,BDH1, and ICD3 were decreased 77, 65, and 22%, respectively(Fig. 7B). These changes in protein abundance were remarkablysimilar in muscles from TXNIPSKM�/� mice (Fig. 7C) and mir-rored the degree to which oxidation rates of each respectivesubstrate were diminished in the TXNIP null condition. By

FIGURE 4. Redox imbalance in TXNIP-deficient skeletal muscles. Skeletal muscles from TKO mice and WT littermate controls were harvested 5 h after foodwithdrawal and immediately flash frozen in liquid N2 for subsequent assessment of redox metabolites. Gastrocnemius muscles were used to measure NADH(A), NADPH (B), and the NADH/NADPH (C) ratio. Quadriceps muscles were used to measure reduced glutathione (GSH) (D), oxidized glutathione (GSSG) (E), andthe GSH/GSSG (F) ratio. G, mitochondrial potential for producing reactive oxygen species was assessed in permeabilized fiber bundles by measuring succinate-supported H2O2 emission rates under state 4 conditions. Data are expressed as mean � S.E. from 5 to 6 animals per group and results were analyzed byStudent’s t test (*, p � 0.05).

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

MARCH 21, 2014 • VOLUME 289 • NUMBER 12 JOURNAL OF BIOLOGICAL CHEMISTRY 8113

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 9: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

FIGURE 5. Targeted metabolic profiling of skeletal muscle and serum. Gastrocnemius muscles and serum from TKO mice and WT littermate controls wereharvested 5– 6 h following food withdrawal at 18 weeks of age. Tissues were immediately flash frozen in liquid N2 and subsequently processed for massspectrometry-based measurement of acyl-CoAs (A), acylcarnitines (B), organic acids (C), amino acids (D), and serum amino acids (E). Data are expressed asmean � S.E. from 5 to 7 animals per group. Results were analyzed by Student’s t test (*, p � 0.05 and §, p � 0.10).

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

8114 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 12 • MARCH 21, 2014

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 10: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

contrast, expression levels of the same three proteins wereunchanged in liver of TKO mice as compared with their controllittermates (not shown). Considering that TXNIP deficiencyincreased circulating levels and muscle production of lactate(Fig. 1, A and G), we examined muscle isoenzyme distributionof LDH, a tetrameric complex consisting of M (muscle) and H(heart) subunits (39). Based on its subunit composition, thecomplexes are referred to as LDH1 (H4), LDH2 (H3M1), LDH3(H2M2), LDH4 (H1M3), or LDH5 (M4). As suspected, native gel

electrophoresis revealed a pronounced shift toward an isoen-zyme composition depleted of LDH1 and enriched in LDH5 inTXNIP null muscles (Fig. 7, D–F), whereas mRNA abundanceof the Ldh5 and Ldh1 genes were unchanged (Fig. 7G). Similarresults were found in muscles from TXNIPSKM�/� mice (Fig. 7,H–K). The LDH1 isozyme favors conversion of lactate to pyru-vate, whereas the opposite is true of LDH5 (39) fitting with themetabolic phenotype of the Txnip null animals. Last, becauseTXNIP appeared to be essential for maximum oxidation of leu-cine, ketones, and lactate, we questioned whether TXNIP istranscriptionally up-regulated in response to physiologicalstresses known to increase muscle reliance on these alternativefuels. Indeed, overnight fasting, genetic diabetes, and prolongedexercise increased Txnip mRNA levels in rodent skeletal mus-cles (Fig. 7, L–O). In summary, these findings establish a tissue-specific role for TXNIP in preserving mitochondrial capacity toswitch substrates during glucose deprivation.

DISCUSSION

TXNIP is a multifaceted molecule that sits at the crossroad ofredox sensing, oxidative stress, and metabolic control (3, 7–14).Despite numerous reports showing that TXNIP deficiencycauses profound derangements in energy metabolism, themolecular mechanisms underlying these consequences remain

FIGURE 6. Targeted metabolic profiling of liver. Liver from TKO mice and WT littermate controls were harvested 5– 6 h following food withdrawal at 18 weeksof age. Specimens were immediately flash frozen in liquid N2 and subsequently processed for mass spectrometry-based measurement of acylcarnitines (A),organic acids (B), and amino acids (C). Data are mean � S.E. from 6 animals per group and results were analyzed by Student’s t test (*, p � 0.05 and §, p � 0.10).

TABLE 1Enzyme activity in isolated mitochondriaData are mean � S.E. (n � 5 or 6 per group).

EnzymeActivity

ChangeaWT KO

nmol/mg protein/h %BCKAD 66.7 (�1.42) 23.7 (�0.73)b 2 65%BDH1 47.9 (�5.34) 24.8 (�2.52)b 2 47%SCHAD 902 (�39.9) 770 (�40.5)c 2 15%ICD3 129.3 (�9.55) 94.8 (�7.02)b 2 27%SDH 889 (�47.7) 817 (�25.4) �PDH 1229 (�28.0) 1240 (�29.6) �ICD2 215 (�9.1) 204 (�12.1) �

a The � and2 indicate percent change in the TXNIP KO relative to the WTcontrol.

b Results were analyzed by Student’s t test (p � 0.001).c Results were analyzed by Student’s t test (p � 0.05).

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

MARCH 21, 2014 • VOLUME 289 • NUMBER 12 JOURNAL OF BIOLOGICAL CHEMISTRY 8115

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 11: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

8116 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 12 • MARCH 21, 2014

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 12: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

murky. Previous reports have suggested that loss of TXNIPleads to respiratory dysfunction that in turn gives rise to aerobicglycolysis, also known as the Warburg effect (8, 9, 11–13). Here,we explored the connection between TXNIP and mitochon-drial performance in skeletal muscle and liver, two organs prin-cipally responsible for whole body glucose and lipid homeosta-sis. We report several new and enlightening observations thatadd to our current physiological understanding of this protein.First, TXNIP deficiency in skeletal muscle resulted in dimin-ished oxidation of all major substrates, leading to reduced exer-cise tolerance. Second, the oxidative insufficiencies in TXNIPnull muscles were not due to reductions in mitochondrial mass,impaired ETC function, or increased mitochondrial reactiveoxygen species production; but instead were attributed todecreased protein abundance of specific mitochondrial dehy-drogenase enzymes. Last, the consequences of TXNIP defi-ciency were tissue-dependent, as the striking metabolic deficitsevident in muscle mitochondria of TKO mice were absent inliver.

Despite marked changes in circulating levels of several met-abolic fuels, whole body energy expenditure and substrateselection were unchanged in TKO mice. This observationaligns with an earlier report (10) and fits with the finding thatTXNIP deficiency did not affect substrate oxidation rates inisolated mitochondria from liver, an organ that contributessubstantially to resting RER. By contrast, genotype-dependentdifferences in the muscle-fueled selection were revealed duringa treadmill challenge. The higher RER in the TKO mice under-going exercise agreed with the lower rates of fatty acid andamino acid oxidation measured in isolated muscles and isolatedmitochondria. Although exercise tolerance was diminished inboth TKO and TXNIPSKM�/� mice, these animals were stillcapable of reaching and maintaining relatively high workloadsduring an acute treadmill test. This outcome was inconsistentwith severe respiratory defects. Indeed, subsequent experi-ments showed that mitochondrial content and ETC functionwere essentially normal in TXNIP null skeletal muscles.

Despite high rates of glycolysis in the TXNIP null muscles,NADH levels were unchanged. This result adds further supportto the notion that ETC function is not a limiting factor in theTKO mice, and raises the possibility that enhanced transfer ofglucose-derived NADH to complex I via the malate/aspartateshuttle (Fig. 8) might compensate for deficiencies in fatty acidand amino acid catabolism during exercise. Additionally, thisfinding conflicts with the idea that NADH accumulation inTKO muscles leads to inactivation of PTEN, a redox-sensitivephosphatidylinositol 3-phosphatase that lowers cellular levelsof (3,4,5)-trisphosphate thereby dampening Akt phosphoryla-tion and insulin signaling (11). Because high NADH levels have

been shown to block TXN/NADP(H)-dependent activation ofPTEN, investigators suggested that a rise in the cellular NADH/NADPH ratio, secondary to respiratory dysfunction, might leadto constitutive oxidation and inactivation of PTEN, thus ampli-fying Akt phosphorylation. Although the latter outcomes wereclearly apparent in TKO muscles (11), NADH and NADPHwere not measured. In the current study TXNIP deficiency didnot affect NADH levels but instead caused a 2-fold rise inNADPH, possibly resulting from increased glucose transportand subsequent metabolism in the pentose phosphate pathway(40). NADPH provides reducing power for regenerating GSHand TXN. Surprisingly, however, elevated NADPH levels in theTKO muscles were accompanied by lower GSH concentrations.Although the reason for this disconnect is uncertain, we suspectthat substrate-driven pressure on membrane-associated NADPHoxidase (NOX), which generates superoxide, might give rise tonon-mitochondrial H2O2 production, thereby increasingdemands on the GSH oxidant scavenging system. Interestingly,NOX-derived reactive oxygen species has been shown to targetand inactivate PTEN as part of a feed-forward mechanism thatboosts insulin action (41).

To our knowledge, this investigation was the first to applycomprehensive metabolic profiling tools to the TXNIP nullmodels. When the targeted metabolomics analyses were com-bined with results of the foregoing substrate oxidation and res-piratory function assays, a clearer picture of TXNIP deficiencybegan to take shape (Fig. 8). First, the tissue-specific role of thisprotein in regulating mitochondrial metabolism was furtherunderscored, as the only commonality between the muscle andliver was the long chain acylcarnitine profile. In both tissues, thedecline in long chain acylcarnitines suggested a decrease ineither CPT1 activity or substrate delivery. Because neitherCPT1 protein abundance nor malonyl-CoA levels were affectedby genotype, we suspect that loss of TXNIP impedes fatty acidtransport into tissues via a yet unknown mechanism. Second,the reduction in skeletal muscle acetyl-CoA levels was consis-tent with catabolic defects upstream of the ETC, and both theacyl-CoA and acylcarnitine profiles pointed to major defects inthe pathways of amino acid and ketone degradation. Most nota-bly, muscle content of isovaleryl-CoA (C5) as well as each of theodd-chain acylcarnitines were robustly decreased in the TKOgroup, whereas circulating BCAA levels were elevated. Consid-ering that isovaleryl-CoA is produced by BCKAD, and becausethis metabolic profile resembles BCKAD deficiency in humansand mice (42), attention was drawn to this complex.

Also quite striking in the TKO mice was the marked rise inintramuscular levels of �-hydroxybutyrylcarnitine. This metabo-lite derives from 3-hydroxyacyl-CoA, a product of ketone metab-olism and a substrate of the SCHAD enzyme (43, 44). Addition-

FIGURE 7. Expression of mitochondrial dehydrogenase enzymes in skeletal muscle and liver. Tissues were harvested 5– 6 h following food withdrawal at18 –22 weeks of age. Skeletal muscles from TKO mice and littermate controls were used for quantitative RT-PCR analysis of mRNAs encoding (A) bckad e1�,bdh1, and icd3 normalized to 18 S, and Western blot analysis of the BCKAD E1A, BDH1, and ICD3 proteins (B) normalized to Memcode staining. Expression of thesame proteins was measured in gastrocnemius muscles (C) from TXNIPSKM�/� mice and TXNIPfl/fl controls. Skeletal muscles from TKO mice and littermatecontrols were used for: D, an LDH isozyme activity gel with WT heart shown as a control; E, quantification of results; F, calculation of the LDH5:LDH1 ratio; andG, quantitative RT-PCR analysis of mRNAs encoding ldh5 and ldh1. The same analyses were performed using gastrocnemius muscles from TXNIPSKM�/� miceand TXNIPfl/fl mice (H–K). Quantitative RT-PCR analysis of mRNA encoding txnip normalized to 18 S was measured in skeletal muscles from fed and fasted (12h) C57BL/6J mice (L), fasted (12 h) and refed (3 h) C57BL/6J mice (M), Zucker Diabetic Fatty (ZDF) rats, and lean controls (O) C57BL/6J mice (N) at rest and 10 min,3 h, or 24 h after a 90-min graded treadmill exercise bout (*, p � 0.05 versus rest). Data are mean � S.E. (n � 5 per group) and results were analyzed by Student’st test (*, p � 0.05 and **, p � 0.001).

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

MARCH 21, 2014 • VOLUME 289 • NUMBER 12 JOURNAL OF BIOLOGICAL CHEMISTRY 8117

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 13: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

ally, the decline in muscle levels of �-KG suggested a potentialimpingement in TCA cycle flux at the ICD3 step. Collectively,these signatures led to the discovery that TXNIP deficiency inmuscle results in a near complete loss of both the BCKAD E1Asubunit and BDH1 protein, with more modest declines in pro-tein abundance of SCHAD and ICD3. These findings help toexplain the elevated circulating levels of ketones, fatty acids,and amino acids in the TXNIP-deficient models as well as theirintolerance to prolonged fasting (12) and endurance exercise.

Another important finding of this study was the TXNIP-de-pendent shift in muscle abundance of the LDH1 and LDH5complexes. This result could explain why the knock-out micehave such high levels of circulating lactate despite normal res-piratory function. Recent studies have shown that permeabi-lized myofibers and isolated muscle mitochondria oxidize lac-tate in a NAD-dependent manner (45), and confocal imaging ofimmunolabeled fibers revealed significant amounts of LDHlocalized to both the outer mitochondrial membrane as well asthe intermembrane space (46). Exercise training-induced adap-tations in the LDH complex, which favor increased LDH1 activ-ity, are thought to play an important role in enhancing pyruvateoxidation and mitigating blood lactate levels during physicalactivity (47). Results herein suggest that TXNIP might contrib-ute to exercise-induced remodeling of the complex by promot-ing synthesis and/or stability of the LDH M subunit.

These findings imply that the redox environment plays akey role in modulating protein abundance of BDH1, BCKAD,

and LDH1, all of which produce NADH. The mechanism(s)by which TXNIP protects specific oxidoreductase enzymesremains unknown, although our data point to regulation at thelevel of protein synthesis and/or stability. TXNIP can formintermolecular disulfide bonds with reduced thioredoxin aswell as other proteins containing a CXXC motif. Emerging evi-dence suggests that TXNIP is present and functionally active inthe mitochondrial matrix (19, 48). Moreover, in INS1 cells,recombinant TXNIP was shown to translocate to the mito-chondria in response to oxidative stress, which in turn activatedthe apoptosis signaling kinase ASK1 by disrupting its interac-tion with mitochondrial localized TXN2 (48). Additionally,TXN has been shown to bind directly to various 2-oxoaciddehydrogenase complexes, which appears to modulate enzymeactivity in vitro (49, 50). Thus, the absence of TXNIP might alterinteractions between TXN2 and other redox-sensitive mito-chondrial proteins.

Last, we consider the physiological relevance of our findingsin the settings of feast and famine. Transcriptional induction ofTXNIP is typically viewed as a negative feedback response toexcessive glucose exposure, mediated via the activation of car-bohydrate-responsive transcription factors, ChREBP andMondoA (51). Our finding that skeletal muscle mRNA expres-sion of TXNIP is up-regulated during nutritional states charac-terized by intramuscular glucose insufficiency, such as fasting,severe diabetes, and prolonged exercise, supports evidence thatthis protein can be regulated by nutrient signals other than

FIGURE 8. TXNIP deficiency disrupts mitochondrial fuel selection. During periods of carbohydrate deprivation, up-regulation of TXNIP serves to limitglucose transport into skeletal muscle and promote mitochondrial oxidation of alternative fuels. TXNIP deficiency enhances glucose uptake and flux throughboth glycolysis and the pentose phosphate pathway. Despite this shift to glycolytic metabolism, respiratory function of the ETC remains intact, therebypermitting transfer of glucose-derived NADH to complex I via the malate/aspartate shuttle. Diminished protein expression and activities of specific mitochon-drial dehydrogenase enzymes lowers muscle capacity for oxidizing lactate, fatty acid, amino acid, and ketones, thus disrupting adaptive mitochondrial fuelswitching in response to starvation and exercise. Key: metabolites affected by TXNIP deficiency are indicated in red (increased) and blue (decreased). Abbre-viations: AcAc, acetoacetate; �-KG, �-ketoglutarate dehydrogenase; AST, apartate aminotransferase; BHB, �-hydroxybutyrate; BDH1, �-hydroxybutyrate dehy-drogenase; CACT, carnitine acylcarnitine translocase; CPT1 and CPT2, carnitine palmitoyltransferase 1 and 2; DHAP, dihydroxyacetone phosphate; G-6-P,glucose 6-phosphate; GDH, glutamate dehydrogenase; GPx, glutathione peroxidase; ICD3, NAD�-dependent isocitrate dehydrogenase; LCAC, long chainacylcarnitine; LCFA-CoA, long chain fatty acid acyl-CoA; LDH1, lactate dehydrogenase 1; LDH5, lactate dehydrogenase 5; MDH, malate dehydrogenase; NOX,NADPH oxidase; PDH, pyruvate dehydrogenase; PPP, pentose phosphate pathway.

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

8118 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 12 • MARCH 21, 2014

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 14: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

glycolytic intermediates (14). Prominent among these is insu-lin, as TXNIP was identified as one of the most potently sup-pressed skeletal muscle mRNAs in healthy human individualssubjected to a hyperinsulinemic-euglycemic clamp (14). Takentogether, these results imply that insulin-mediated suppressionof TXNIP in the refed state serves not only to disinhibit glucoseuptake, but also to restrict ketone and BCAA oxidation andpromote net protein synthesis. Conversely, during periods ofprolonged glucose restriction or chronic exercise training, skel-etal muscles are reprogrammed to switch energy currency fromglucose to fatty acids, ketones, and amino acids. This occurs atthe level of substrate supply as well as increased enzyme activ-ities and protein abundance (52–54) and serves as a fundamen-tal survival mechanism that preserves glucose for the brain.

Whereas glucose sparing favors survival during times of fam-ine, elevated TXNIP expression in nutrient replete muscles ofobese and diabetic individuals might contribute to glucose dis-use. For example, obesity and diabetes are associated with ele-vated circulating levels of fatty acids, ketones, and BCAA, aswell as hyperglycemia (55, 56). A large body of evidence linksthe development of insulin resistance to heightened competi-tion between glucose and lipid-derived substrates (57). Morerecent studies implicate a similar role for BCAAs (56, 58),which can fuel the TCA cycle by providing both acetyl-CoA andanaplerotic carbons thereby rendering glucose superfluous.Based on these findings, we speculate that inappropriate up-regu-lation of TXNIP in the context of overnutrition exacerbates sub-strate competition and contributes to nutrient-induced mito-chondrial stress and/or redox imbalance (59). In summary, thiswork identifies a novel role for TXNIP in coordinating fuel selec-tion in response to nutrient availability and furthers implicates thisprotein as a potential antidiabetic drug target.

Acknowledgments—We acknowledge the late Dr. Roger Davis for gen-erating the mouse lines used in these experiments and for his manyintellectual contributions to this field. We thank Dr. Chris Lynch forproviding the BCKAD antibody.

REFERENCES1. Chen, K. S., and DeLuca, H. F. (1994) Isolation and characterization of a

novel cDNA from HL-60 cells treated with 1,25-dihydroxyvitamin D3.Biochim. Biophys. Acta 1219, 26 –32

2. Nishiyama, A., Matsui, M., Iwata, S., Hirota, K., Masutani, H., Nakamura,H., Takagi, Y., Sono, H., Gon, Y., and Yodoi, J. (1999) Identification ofthioredoxin-binding protein-2/vitamin D3 up-regulated protein 1 as anegative regulator of thioredoxin function and expression. J. Biol. Chem.274, 21645–21650

3. Bodnar, J. S., Chatterjee, A., Castellani, L. W., Ross, D. A., Ohmen, J.,Cavalcoli, J., Wu, C., Dains, K. M., Catanese, J., Chu, M., Sheth, S. S.,Charugundla, K., Demant, P., West, D. B., de Jong, P., and Lusis, A. J.(2002) Positional cloning of the combined hyperlipidemia gene Hyplip1.Nat. Genet. 30, 110 –116

4. Nordberg, J., and Arnér, E. S. (2001) Reactive oxygen species, antioxidants,and the mammalian thioredoxin system. Free Radic. Biol. Med. 31,1287–1312

5. Alvarez, C. E. (2008) On the origins of arrestin and rhodopsin. BMC Evol.Biol. 8, 222

6. Spindel, O. N., World, C., and Berk, B. C. (2012) Thioredoxin interactingprotein. Redox dependent and independent regulatory mechanisms. An-tioxid. Redox Signal. 16, 587–596

7. Donnelly, K. L., Margosian, M. R., Sheth, S. S., Lusis, A. J., and Parks, E. J.(2004) Increased lipogenesis and fatty acid reesterification contribute tohepatic triacylglycerol stores in hyperlipidemic Txnip�/� mice. J. Nutr.134, 1475–1480

8. Hui, T. Y., Sheth, S. S., Diffley, J. M., Potter, D. W., Lusis, A. J., Attie, A. D.,and Davis, R. A. (2004) Mice lacking thioredoxin-interacting protein pro-vide evidence linking cellular redox state to appropriate response to nu-tritional signals. J. Biol. Chem. 279, 24387–24393

9. Sheth, S. S., Castellani, L. W., Chari, S., Wagg, C., Thipphavong, C. K.,Bodnar, J. S., Tontonoz, P., Attie, A. D., Lopaschuk, G. D., and Lusis, A. J.(2005) Thioredoxin-interacting protein deficiency disrupts the fasting-feeding metabolic transition. J. Lipid Res. 46, 123–134

10. Chutkow, W. A., Birkenfeld, A. L., Brown, J. D., Lee, H. Y., Frederick,D. W., Yoshioka, J., Patwari, P., Kursawe, R., Cushman, S. W., Plutzky, J.,Shulman, G. I., Samuel, V. T., and Lee, R. T. (2010) Deletion of the �-ar-restin protein Txnip in mice promotes adiposity and adipogenesis whilepreserving insulin sensitivity. Diabetes 59, 1424 –1434

11. Hui, S. T., Andres, A. M., Miller, A. K., Spann, N. J., Potter, D. W., Post,N. M., Chen, A. Z., Sachithanantham, S., Jung, D. Y., Kim, J. K., and Davis,R. A. (2008) Txnip balances metabolic and growth signaling via PTENdisulfide reduction. Proc. Natl. Acad. Sci. U.S.A. 105, 3921–3926

12. Oka, S., Liu, W., Masutani, H., Hirata, H., Shinkai, Y., Yamada, S., Yoshida,T., Nakamura, H., and Yodoi, J. (2006) Impaired fatty acid utilization inthioredoxin binding protein-2 (TBP-2)-deficient mice. A unique animalmodel of Reye syndrome. FASEB J. 20, 121–123

13. Chutkow, W. A., Patwari, P., Yoshioka, J., and Lee, R. T. (2008) Thiore-doxin-interacting protein (Txnip) is a critical regulator of hepatic glucoseproduction. J. Biol. Chem. 283, 2397–2406

14. Parikh, H., Carlsson, E., Chutkow, W. A., Johansson, L. E., Storgaard, H.,Poulsen, P., Saxena, R., Ladd, C., Schulze, P. C., Mazzini, M. J., Jensen, C. B.,Krook, A., Björnholm, M., Tornqvist, H., Zierath, J. R., Ridderstråle, M., Alt-shuler, D., Lee, R. T., Vaag, A., Groop, L. C., and Mootha, V. K. (2007) TXNIPregulates peripheral glucose metabolism in humans. PLoS Med 4, e158

15. Yoshihara, E., Fujimoto, S., Inagaki, N., Okawa, K., Masaki, S., Yodoi, J.,and Masutani, H. (2010) Disruption of TBP-2 ameliorates insulin sensi-tivity and secretion without affecting obesity. Nat. Commun. 1, 127

16. Wu, N., Zheng, B., Shaywitz, A., Dagon, Y., Tower, C., Bellinger, G., Shen,C. H., Wen, J., Asara, J., McGraw, T. E., Kahn, B. B., and Cantley, L. C.(2013) AMPK-dependent degradation of TXNIP upon energy stress leadsto enhanced glucose uptake via GLUT1. Mol. Cell 49, 1167–1175

17. Zhou, R., Tardivel, A., Thorens, B., Choi, I., and Tschopp, J. (2010) Thi-oredoxin-interacting protein links oxidative stress to inflammasome acti-vation. Nat. Immunol. 11, 136 –140

18. Schulze, P. C., Yoshioka, J., Takahashi, T., He, Z., King, G. L., and Lee, R. T.(2004) Hyperglycemia promotes oxidative stress through inhibition ofthioredoxin function by thioredoxin-interacting protein. J. Biol. Chem.279, 30369 –30374

19. Yoshioka, J., Chutkow, W. A., Lee, S., Kim, J. B., Yan, J., Tian, R., Lindsey,M. L., Feener, E. P., Seidman, C. E., Seidman, J. G., and Lee, R. T. (2012)Deletion of thioredoxin-interacting protein in mice impairs mitochon-drial function but protects the myocardium from ischemia-reperfusioninjury. J. Clin. Invest. 122, 267–279

20. Muoio, D. M., Noland, R. C., Kovalik, J. P., Seiler, S. E., Davies, M. N.,DeBalsi, K. L., Ilkayeva, O. R., Stevens, R. D., Kheterpal, I., Zhang, J., Cov-ington, J. D., Bajpeyi, S., Ravussin, E., Kraus, W., Koves, T. R., and Mynatt,R. L. (2012) Muscle-specific deletion of carnitine acetyltransferase com-promises glucose tolerance and metabolic flexibility. Cell Metab. 15,764 –777

21. Kim, J. Y., Koves, T. R., Yu, G. S., Gulick, T., Cortright, R. N., Dohm, G. L.,and Muoio, D. M. (2002) Evidence of a malonyl-CoA-insensitive carnitinepalmitoyltransferase I activity in red skeletal muscle. Am. J. Physiol. Endo-crinol. Metab. 282, E1014 –1022

22. Muoio, D. M., Dohm, G. L., Tapscott, E. B., and Coleman, R. A. (1999)Leptin opposes insulin’s effects on fatty acid partitioning in muscles iso-lated from obese ob/ob mice. Am. J. Physiol. 276, E913–921

23. Dohm, G. L., Tapscott, E. B., Pories, W. J., Dabbs, D. J., Flickinger, E. G.,Meelheim, D., Fushiki, T., Atkinson, S. M., Elton, C. W., and Caro, J. F.(1988) An in vitro human muscle preparation suitable for metabolic stud-

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

MARCH 21, 2014 • VOLUME 289 • NUMBER 12 JOURNAL OF BIOLOGICAL CHEMISTRY 8119

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 15: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

ies. Decreased insulin stimulation of glucose transport in muscle frommorbidly obese and diabetic subjects. J. Clin. Invest. 82, 486 – 494

24. Boushel, R., Gnaiger, E., Schjerling, P., Skovbro, M., Kraunsøe, R., andDela, F. (2007) Patients with type 2 diabetes have normal mitochondrialfunction in skeletal muscle. Diabetologia 50, 790 –796

25. Kuznetsov, A. V., Tiivel, T., Sikk, P., Kaambre, T., Kay, L., Daneshrad, Z.,Rossi, A., Kadaja, L., Peet, N., Seppet, E., and Saks, V. A. (1996) Strikingdifferences between the kinetics of regulation of respiration by ADP inslow-twitch and fast-twitch muscles in vivo. Eur. J. Biochem. 241,909 –915

26. Tonkonogi, M., Fernström, M., Walsh, B., Ji, L. L., Rooyackers, O., Ham-marqvist, F., Wernerman, J., and Sahlin, K. (2003) Reduced oxidativepower but unchanged antioxidative capacity in skeletal muscle from agedhumans. Pflugers Arch. 446, 261–269

27. Perry, C. G., Kane, D. A., Lin, C. T., Kozy, R., Cathey, B. L., Lark, D. S.,Kane, C. L., Brophy, P. M., Gavin, T. P., Anderson, E. J., and Neufer, P. D.(2011) Inhibiting myosin-ATPase reveals a dynamic range of mitochon-drial respiratory control in skeletal muscle. Biochem. J. 437, 215–222

28. Anderson, E. J., Lustig, M. E., Boyle, K. E., Woodlief, T. L., Kane, D. A., Lin,C. T., Price, J. W., 3rd, Kang, L., Rabinovitch, P. S., Szeto, H. H., Houmard, J. A.,Cortright, R. N., Wasserman, D. H., and Neufer, P. D. (2009) MitochondrialH2O2 emission and cellular redox state link excess fat intake to insulin resis-tance in both rodents and humans. J. Clin. Invest. 119, 573–581

29. Koves, T. R., Ussher, J. R., Noland, R. C., Slentz, D., Mosedale, M., Ilkayeva,O., Bain, J., Stevens, R., Dyck, J. R., Newgard, C. B., Lopaschuk, G. D., andMuoio, D. M. (2008) Mitochondrial overload and incomplete fatty acidoxidation contribute to skeletal muscle insulin resistance. Cell Metab. 7,45–56

30. Binstock, J. F., and Schulz, H. (1981) Fatty acid oxidation complex fromEscherichia coli. Methods Enzymol. 71, 403– 411

31. Hale, D. E., Cornell, J. E., and Bennett, M. J. (1997) Stability of long-chainand short-chain 3-hydroxyacyl-CoA dehydrogenase activity in postmor-tem liver. Clin. Chem. 43, 273–278

32. Grinblat, L., Pacheco Bolaños, L. F., and Stoppani, A. O. (1986) Decreasedrate of ketone-body oxidation and decreased activity of D3-hydroxybu-tyrate dehydrogenase and succinyl-CoA:3-oxo-acid CoA-transferase inheart mitochondria of diabetic rats. Biochem. J. 240, 49 –56

33. Lehninger, A. L., Sudduth, H. C., and Wise, J. B. (1960) D-�-Hydroxybu-tyric dehydrogenase of mitochondria. J. Biol. Chem. 235, 2450 –2455

34. Reisch, A. S., and Elpeleg, O. (2007) Biochemical assays for mitochondrialactivity. Assays of TCA cycle enzymes and PDHc. Methods Cell Biol. 80,199 –222

35. Yarian, C. S., Toroser, D., and Sohal, R. S. (2006) Aconitase is the mainfunctional target of aging in the citric acid cycle of kidney mitochondriafrom mice. Mech. Ageing Dev. 127, 79 – 84

36. Guo, W., Jiang, L., Bhasin, S., Khan, S. M., and Swerdlow, R. H. (2009)DNA extraction procedures meaningfully influence qPCR-based mtDNAcopy number determination. Mitochondrion 9, 261–265

37. McGarry, J. D., Leatherman, G. F., and Foster, D. W. (1978) Carnitinepalmitoyltransferase I. The site of inhibition of hepatic fatty acid oxidationby malonyl-CoA. J. Biol. Chem. 253, 4128 – 4136

38. Soeters, M. R., Serlie, M. J., Sauerwein, H. P., Duran, M., Ruiter, J. P., Kulik,W., Ackermans, M. T., Minkler, P. E., Hoppel, C. L., Wanders, R. J., andHouten, S. M. (2012) Characterization of D3-hydroxybutyrylcarnitine (ke-tocarnitine). An identified ketosis-induced metabolite. Metabolism 61,966 –973

39. Dawson, D. M., Goodfriend, T. L., and Kaplan, N. O. (1964) Lactic dehy-drogenases. Functions of the two types rates of synthesis of the two majorforms can be correlated with metabolic differentiation. Science 143,929 –933

40. Stanton, R. C. (2012) Glucose-6-phosphate dehydrogenase, NADPH, andcell survival. IUBMB Life 64, 362–369

41. Seo, J. H., Ahn, Y., Lee, S. R., Yeol Yeo, C., and Chung Hur, K. (2005) Themajor target of the endogenously generated reactive oxygen species inresponse to insulin stimulation is phosphatase and tensin homolog and

not phosphoinositide 3-kinase (PI-3 kinase) in the PI-3 kinase/Akt path-way. Mol. Biol. Cell 16, 348 –357

42. Chuang, D. T., and Shih, V. E. (2001) Maple syrup urine disease (branched-chain ketoaciduria). in The Metabolic and Molecular Bases of Inherited Dis-ease (Scriver, C. R., ed.) 8th Ed., Vol. 2, pp. 1971–2005, McGraw-Hill, NewYork

43. Rinaldo, P., Cowan, T. M., and Matern, D. (2008) Acylcarnitine profileanalysis. Genet. Med. 10, 151–156

44. Koeberl, D. D., Young, S. P., Gregersen, N. S., Vockley, J., Smith, W. E.,Benjamin, D. K., Jr., An, Y., Weavil, S. D., Chaing, S. H., Bali, D., McDonald,M. T., Kishnani, P. S., Chen, Y. T., and Millington, D. S. (2003) Raredisorders of metabolism with elevated butyryl- and isobutyryl-carnitinedetected by tandem mass spectrometry newborn screening. Pediatr. Res.54, 219 –223

45. Jacobs, R. A., Meinild, A. K., Nordsborg, N. B., and Lundby, C. (2013)Lactate oxidation in human skeletal muscle mitochondria. Am. J. Physiol.Endocrinol. Metab. 304, E686 – 694

46. Elustondo, P. A., White, A. E., Hughes, M. E., Brebner, K., Pavlov, E., andKane, D. A. (2013) Physical and functional association of lactate dehydro-genase (LDH) with skeletal muscle mitochondria. J. Biol. Chem. 288,25309 –25317

47. Summermatter, S., Santos, G., Pérez-Schindler, J., and Handschin, C.(2013) Skeletal muscle PGC-1� controls whole-body lactate homeostasisthrough estrogen-related receptor �-dependent activation of LDH B andrepression of LDH A. Proc. Natl. Acad. Sci. U.S.A. 110, 8738 – 8743

48. Saxena, G., Chen, J., and Shalev, A. (2010) Intracellular shuttling and mi-tochondrial function of thioredoxin-interacting protein. J. Biol. Chem.285, 3997– 4005

49. Bunik, V., Raddatz, G., Lemaire, S., Meyer, Y., Jacquot, J. P., and Biss-wanger, H. (1999) Interaction of thioredoxins with target proteins. Role ofparticular structural elements and electrostatic properties of thioredoxinsin their interplay with 2-oxoacid dehydrogenase complexes. Protein Sci. 8,65–74

50. Bunik, V. I. (2003) 2-Oxo acid dehydrogenase complexes in redox regula-tion. Eur. J. Biochem. 270, 1036 –1042

51. Stoltzman, C. A., Peterson, C. W., Breen, K. T., Muoio, D. M., Billin, A. N.,and Ayer, D. E. (2008) Glucose sensing by MondoA:Mlx complexes. A rolefor hexokinases and direct regulation of thioredoxin-interacting proteinexpression. Proc. Natl. Acad. Sci. U.S.A. 105, 6912– 6917

52. Fujii, H., Shimomura, Y., Tokuyama, K., and Suzuki, M. (1994) Modula-tion of branched-chain 2-oxo acid dehydrogenase complex activity in ratskeletal muscle by endurance training. Biochim. Biophys. Acta 1199,130 –136

53. Holecek, M. (2001) Effect of starvation on branched-chain �-keto aciddehydrogenase activity in rat heart and skeletal muscle. Physiol. Res. 50,19 –24

54. Shimomura, Y., Fujii, H., Suzuki, M., Murakami, T., Fujitsuka, N., andNakai, N. (1995) Branched-chain �-keto acid dehydrogenase complex inrat skeletal muscle. Regulation of the activity and gene expression by nu-trition and physical exercise. J. Nutr. 125, 1762S-1765S

55. Muoio, D. M., and Newgard, C. B. (2008) Mechanisms of disease. Molec-ular and metabolic mechanisms of insulin resistance and �-cell failure intype 2 diabetes. Nat. Rev. Mol. Cell Biol. 9, 193–205

56. Newgard, C. B. (2012) Interplay between lipids and branched-chain aminoacids in development of insulin resistance. Cell Metab. 15, 606 – 614

57. Hue, L., and Taegtmeyer, H. (2009) The Randle cycle revisited. A new headfor an old hat. Am. J. Physiol. Endocrinol. Metab. 297, E578 –591

58. Wang, T. J., Larson, M. G., Vasan, R. S., Cheng, S., Rhee, E. P., McCabe, E.,Lewis, G. D., Fox, C. S., Jacques, P. F., Fernandez, C., O’Donnell, C. J., Carr,S. A., Mootha, V. K., Florez, J. C., Souza, A., Melander, O., Clish, C. B., andGerszten, R. E. (2011) Metabolite profiles and the risk of developing dia-betes. Nat. Med. 17, 448 – 453

59. Muoio, D. M., and Neufer, P. D. (2012) Lipid-induced mitochondrialstress and insulin action in muscle. Cell Metab 15, 595– 605

TXNIP Coordinates Mitochondrial Fuel Switching in Skeletal Muscle

8120 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 12 • MARCH 21, 2014

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 16: TargetedMetabolomicsConnectsThioredoxin-interacting ... · [14C]CO 2 from either 0.625 Ci/ml of [1- 14C]palmitate (100 M palmitate bound to 0.15% BSA) or 0.625 Ci/ml of [2-14C]pyruvate(1mM

MuoioPerry, Daniel S. Lark, Simon T. Hui, Luke Szweda, P. Darrell Neufer and Deborah M.

Seiler, April H. Wittmann, Olga R. Ilkayeva, Robert D. Stevens, Christopher G. R. Karen L. DeBalsi, Kari E. Wong, Timothy R. Koves, Dorothy H. Slentz, Sarah E.

in Skeletal MuscleMitochondrial Fuel Selection and Regulation of Specific Oxidoreductase Enzymes

Targeted Metabolomics Connects Thioredoxin-interacting Protein (TXNIP) to

doi: 10.1074/jbc.M113.511535 originally published online January 30, 20142014, 289:8106-8120.J. Biol. Chem. 

  10.1074/jbc.M113.511535Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

  http://www.jbc.org/content/289/12/8106.full.html#ref-list-1

This article cites 59 references, 20 of which can be accessed free at

by guest on October 19, 2020

http://ww

w.jbc.org/

Dow

nloaded from