7 figures, 1 table and 3 additional files

Figures

Figure 1 with 1 supplement
Generation of mice with muscle-specific deletion of Mpc1 (MPC SkmKO).

(A) Scheme illustrating generation of the muscle-specific Mpc1 null allele. (B) Representative western blots of MPC1 and MPC2 protein abundance in mouse tissues. Loading was normalized to total protein. A reference protein is not shown because of lack of an equally expressed protein across different tissues (age 21 weeks; n = 3, littermates; TA, tibialis anterior; BAT, brown adipose tissue; WAT, white adipose tissue). (C14C-pyruvate uptake by muscle mitochondria isolated from WT and MPC SkmKO mice (age 13 weeks, n = 6, four littermates + 2 non-littermates, two-tailed t-test). (D, E) Pyruvate- (D) and glutamate-driven (E) respiration by muscle mitochondria isolated from WT and MPC SkmKO mice. Experimental media contained 1 mM malate and 10 mM pyruvate or 10 mM glutamate (age 16 weeks; n = 6, four littermates + 2 non-littermates; two-tailed t-test; FCCP, trifluoromethoxy carbonylcyanide phenylhydrazone; CHC, 4-alpha-hydroxycinnamatic acid; Rot., rotenone). (F) Representative western blots of components of electron transport chain (ETC) complexes I-V, MPC1, VDAC, and HSP90 proteins in TA muscle from WT and MPC SkmKO mice (age 21 weeks, n = 6, littermates). Data presented as mean ± SEM (*p<0.05, **p<0.01).

https://doi.org/10.7554/eLife.45873.002
Figure 1—figure supplement 1
Loss of Mpc1 but not Mpc2 transcript in MPC skmKO skeletal muscle.

Relative Mpc1 and Mpc2 transcript abundance in the TA muscle of WT and MPC SkmKO mice, normalized to U36b4. (n = 5-6 littermates, age = 16 weeks, two tailed t-test). Data are presented as mean +/- SEM (*p<0.05, **p<0.01, ***p<0.001).

https://doi.org/10.7554/eLife.45873.003
Figure 2 with 1 supplement
Basic characterization of MPC SkmKO mice.

(A) Grip strength measured by full force on a triangular bar of WT and MPC SkmKO mice (age 15 weeks, n = 8, seven littermates + 1 non-littermate, two-tailed t-test). (B) Exercise tolerance of WT and MPC SkmKO mice measured by running duration and speed at exhaustion on a rodent treadmill where belt velocity was incrementally increased (age 14 weeks, n = 6, littermates, two-tailed t-test). (C) Body weight (BW)-normalized daily food intake of WT and MPC SkmKO mice (age 15 weeks, n = 8, seven littermates + 1 non-littermate, two-tailed t-test). (D - F) Oxygen consumption (VO2) (D), Respiratory exchange ratio (RER) (E), and voluntary locomotion (beam breaks) (F) of WT and MPC SkmKO mice (age 12 weeks, n = 8, littermates, 60 min rolling averages analyzed by two-tailed t-test). Black dots indicate points where significant differences were detected and volcano plots show the distribution of p-values plotted by the direction of change. Data presented as mean ± SEM (*p<0.05 and as indicated on volcano plots).

https://doi.org/10.7554/eLife.45873.004
Figure 2—figure supplement 1
Skeletal muscle specific force production and glycogen content are unchanged in MPC SkmKO mice.

(A) Specific tetanic force generated by the EDL muscle ex vivo of WT and MPC SkmKO mice. (n = 5 littermates, age = 45 weeks, two tailed t-test). (B) Glycogen content in quadriceps muscle of WT and MPC SkmKO mice. (n = 8, 7 littermates one non-littermates, age = 24 weeks, two tailed t-test). Data are presented as mean ± SEM (*p<0.05, **p<0.01, ***p<0.001).

https://doi.org/10.7554/eLife.45873.005
Figure 3 with 2 supplements
Leanness, increased fatty acid oxidation, and altered systemic glucose metabolism.

(A-B) Total fat (A) and lean (B) mass of live WT and MPC SkmKO mice measured by NMR (ages 9–17 weeks, n = 6 littermates, two-tailed t-test). (C) Ratio of muscle 3H-triolein uptake and partitioning into aqueous and organic fractions (age 16 weeks, n = 7, littermates, two-tailed t-test). (D) Oxidation of 14C-palmitate by incubated EDL muscles to CO2 (complete) and acid soluble metabolites (ASM) (age 13 weeks, before MPC SkmKO leanness, n = 6, littermates, two-tailed t-test; EDL, extensor digitorum longus). (E, F) Blood glucose (E) and lactate (F) levels during glucose tolerance tests with 7 week-old (young) WT and MPC SkmKO mice (n=17, 15 littermates, 2 non-littermates, two-tailed t-test). (G, H) Blood glucose (G) and lactate (H) levels during glucose tolerance tests with 36 week-old (old) WT and MPC SkmKO mice (n=8, littermates, two-tailed t-test). Data presented as mean ± SEM (*p<0.05, **p<0.01).

https://doi.org/10.7554/eLife.45873.006
Figure 3—source data 1

Serum parameters of 12 hr fasted and 30 min post refed WT and MPC SkmKO mice.

(n = 8, littermates, age = 15 weeks, two way ANOVA). Data are presented as mean ± SEM (**p<0.01, ***p<0.001).

https://doi.org/10.7554/eLife.45873.009
Figure 3—figure supplement 1
Fat mass loss and lean mass retention in MPC SkmKO mice.

(A) Body composition of WT and MPC SkmKO mice at 40 weeks of age. (n = 5 littermates, two tailed t-test). (B) Adipose depot weights of WT and MPC SkmKO mice at 40 weeks of age. (n = 5 littermates, two tailed t-test). (C) Muscle mass/Tibia length of WT and MPC SkmKO hindlimb and forelimb muscles at 40 weeks of age. (n = 5 littermates; two tailed t-test; Quads, quadriceps; TA, tibialis anterior; EDL, extensor digitorum longus). (D) Grip strength of WT and MPC SkmKO mice at 40 weeks of age. (n = 5 littermates, two tailed t-test). (E) Serum FGF21 levels in WT and MPC SkmKO mice. (n = 6 littermates, age = 21 weeks, two tailed t-test) Data are presented as mean ± SEM (*p<0.05, **p<0.01, ***p<0.001).

https://doi.org/10.7554/eLife.45873.007
Figure 3—figure supplement 2
Fat mass loss in female MPC SkmKO mice and not Myogenin-CRE/+ mice.

(A) Body composition of WT and MPC SkmKO female mice at 28 weeks of age (n = 10–13 littermates, two tailed t-test). (B) Body composition of WT and Myogenin-CRE/+mice at 21 weeks of age (n = 8 littermates, two tailed t-test) Data are presented as mean ± SEM (*p<0.05, **p<0.01, ***p<0.001).

https://doi.org/10.7554/eLife.45873.008
Figure 4 with 2 supplements
Hyperinsulinemic-euglycemic clamps.

Clamps were performed on 5 hr fasted, unrestrained, and conscious WT and MPC SkmKO mice. After the basal sampling period, whole body glucose flux was traced by infusion of 2.5 mU/kg/min insulin and of D-[3 H3]-glucose at time t = 0. At 45 min prior to clamp conclusion, [1-14C]−2-deoxy-D-glucose was infused over 5 min in a single bolus. Tissue samples were collected at clamp conclusion (ages 24–29 weeks, n = 6–8, six littermates + 2 non-littermates, two tailed t-test). (A - D) Blood glucose levels (A), glucose infusion rate (GIR) (B), appearance rate (Ra) (C), and disposal rate (Rd) (D) (ages 24–29 weeks, n = 6–8, six littermates + 2 non-littermates, two tailed t-test). (E, F) Tissue [1-14C]−2-deoxy-D-glucose uptake (E) and blood lactate levels (F) during the steady-state portion of the clamp (ages 24–29 weeks; n = 6–8; six littermates + 2 non-littermates; two tailed t-test; WAT, white adipose tissue, BAT, brown adipose tissue, EDL, extensor digitorum longus, Gastroc, gastrocnemius). Data presented as mean ± SEM (*p<0.05, **p<0.01).

https://doi.org/10.7554/eLife.45873.010
Figure 4—figure supplement 1
Basal and steady state plasma insulin concentrations during hyperinsulinemic-euglycemic clamps.

Insulin levels measured during basal and steady state phases of hyperinsulinemic-euglycemic clamps in WT and MPC SkmKO mice (ages 24–29 weeks, n = 6–8, six littermates + 2 non-littermates, two tailed t-test). Data are presented as mean ± SEM.

https://doi.org/10.7554/eLife.45873.011
Figure 4—figure supplement 2
AKT, AMPK, and PDH phosphorylation in gastrocnemius muscle collected after hyper-insulinemic euglycemic clamps.

(A) Representative western blot and quantification of total and phosphorylated (S473) AKT following hyperinsulinemic euglycemia clamps in gastroc tissue of WT and MPC SkmKO mice. GAPDH is shown as loading control (ages 24-29 weeks, n=6-8, 6 littermates + 2 non-littermates, two tailed t-test). (B) Representative western blot and quantification of total and phosphorylated (Thr172) AMPK following hyperinsulinemic euglycemia clamps in gastroc tissue of WT and MPC SkmKO mice. GAPDH is shown as loading control (ages 24-29 weeks, n=6-8, 6 littermates + 2 non-littermates, two tailed t-test). (C) Representative western blot and quantification of total and phosphorylated (S232) PDH E1 following hyperinsulinemic euglycemia clamps in gastroc tissue of WT and MPC SkmKO mice. GAPDH is shown as loading control (ages 24-29 weeks, n=6-8, 6 littermates + 2 non-littermates, two tailed t-test). Data are presented as mean ± SEM (*p<0.05).

https://doi.org/10.7554/eLife.45873.012
Figure 5 with 3 supplements
Mechanisms of metabolic adaptation.

(A) Tibialis anterior (TA) muscle force production during 0.5 Hz isometric contraction by in situ peroneal nerve stimulation of live anesthetized WT and MPC SkmKO mice (age 12 weeks, n = 5–6, littermates, 30 s rolling averages analyzed by two-tailed t-test). (B) Relative metabolite abundance of pyruvate, lactate, NADH, and NAD+ in sham-treated (-) and contracted (+) TA muscles of WT and MPC SkmKO mice (age 12 weeks, n = 5–6, littermates, two-way ANOVA). (C) Relative metabolite abundance of isocitrate +citrate (Iso)Citrate, malate, succinate, and α-ketoglutarate in sham-treated (-) and contracted (+) TA muscles of WT and MPC SkmKO mice (age 12 weeks, n = 5–6, littermates, two-way ANOVA). (D) Relative metabolite abundance of acetylcarnitine, butyryl-L-carnitine, octanoyl-L-carnitine, and carnitine in sham-treated (-) and contracted (+) TA muscles of WT and MPC SkmKO mice (age 12 weeks, n = 5–6, littermates, two-way ANOVA). (E) Citrate 13C enrichment (13C labeled/non-labeled citrate) in TA muscles 65 min after intraperitoneal U13C-glucose injection of WT and MPC SkmKO mice (age 12 weeks, n = 8, seven littermates + 1 non-littermate, two-way ANOVA). (F) Malonyl-CoA levels in quadriceps muscles of WT and MPC SkmKO mice (age 11 weeks, n = 6, littermates, two tailed student's t test). (G-H) Relative metabolite abundance of glutamate and glutamine (G) and alanine and aspartate (H) in sham-treated (-) and contracted (+) TA muscles of WT and MPC SkmKO mice (age 12 weeks, n = 5–6, littermates, two-way ANOVA). (I) Ex vivo pyruvate-driven, ADP-stimulated respiration of permeabilized mouse soleus muscle treated with UK5099, β-chloro-alanine (β-Cl-A), rotenone (Rot), and rescued with succinate (age 11 weeks, n = 6, littermates, two-tailed t-test). Data are presented as mean ± SEM (*p<0.05, **p<0.01, ***p<0.001, placement on x-axis signifies genotype main effect).

https://doi.org/10.7554/eLife.45873.013
Figure 5—figure supplement 1
TA muscle force production during 1 Hz in situ contraction and metabolomics principal component (PCA) analysis of 0.5 Hz contracted TA muscles after in situ contraction.

(A) Normalized real-time force produced during 1 Hz isometric twitches of the TA muscle of WT and MPC SkmKO mice (n = 6–7, littermates, age = 12 weeks, 30 s rolling averages analyzed by two tailed t-test). (B) Principal component analysis (PCA) score plot of the first and second principal components (PCs) from 61 metabolites. (n = 5–6, littermates; age = 12 weeks; KO, MPC SkmKO; Con, contracted; Non-Con, non-contracted). Data are presented as mean ± SEM (*p<0.05, **p<0.01, ***p<0.001).

https://doi.org/10.7554/eLife.45873.014
Figure 5—figure supplement 2
Transcript abundance of key fatty acid oxidation enzymes is not changed in MPC SkmKO skeletal muscle.

(A-E) Relative transcript abundance in the TA muscle of WT and MPC SkmKO mice of Cpt1b (A), Echs1 (B), Hadha (C), Mpc1 (D), Mpc2 (E) normalized to U36b4 (n = 6, littermates, age = 11 weeks, two tailed t-test). Data are presented as mean ± SEM ***p<0.001).

https://doi.org/10.7554/eLife.45873.015
Figure 5—figure supplement 3
Increased liver citrate 13C enrichment in MPC SkmKO mice following intraperitoneal injection of U13C-glucose.

Liver citrate 13C enrichment (13C labeled/non-labeled citrate) in liver of WT and MPC SkmKO mice 65 min after U13C-glucose injection (n = 8, 7 littermates one non-littermate, age = 12 weeks, two tailed t-test). Data are presented as mean ± SEM (*p<0.05).

https://doi.org/10.7554/eLife.45873.016
Figure 6 with 2 supplements
Protection and recovery from high fat diet-induced obesity.

(A) Schema illustrating the time course of high fat diet (HFD) feeding and a switch to synthetic normal fat control diet (NFD). (B, C, and D) Body weight (B), fat mass (C), and lean mass (D) of WT and MPC SkmKO mice measured by NMR during HFD feeding of WT and MPC SkmKO mice (ages 9–25 weeks, n = 7–8, littermates, two-tailed t-test). (E, F, and G) Body weight (E), fat mass (F), and lean mass (G) of WT and MPC SkmKO mice measured by NMR during post-HFD, normal fat synthetic control diet (NFD) feeding of WT and MPC SkmKO mice (ages 58–71 weeks, n = 5–6, littermates, two-tailed t-test). (H, I) Fasted and refed blood glucose (H) and serum insulin (I) levels at ends of HFD and post-HFD, NFD treatments of WT and MPC SkmKO mice (ages 58 and 71 weeks, n = 5–6, littermates, two way ANOVA). (J) Schema illustrating the time course of mouse HFD treatment, tamoxifen injection for acute muscle-specific Mpc1 deletion (MPC iSkmKO), and switch to normal chow diet (NCD) feeding. (K, L, and M) Body weight (K), fat mass (L), and lean mass (M) measured by NMR from end of 5 days tamoxifen administration, during 2 weeks continued HFD, and during 10 weeks post-HFD, NCD feeding of WT and MPC iSkmKO mice (ages 22–34 weeks, n = 6–7, littermates, two-tailed t-test). Data are presented as mean ± SEM (*p<0.05, **p<0.01, ***p<0.001).

https://doi.org/10.7554/eLife.45873.017
Figure 6—source data 1

Serum Parameters of 12 hr fasted and 30 min post refed WT and MPC SkmKO mice after 48 weeks of HFD and after 14 weeks of NFD recovery.

(n = 8, littermates, age 58 and 71 weeks, two way ANOVA). Data are presented as mean ± SEM (*p<0.05, **p<0.01, ***p<0.001).

https://doi.org/10.7554/eLife.45873.020
Figure 6—figure supplement 1
WT and MPC SkmKO mouse physiological parameters during high fat diet (HFD) and after return to normal fat diet (NFD) feeding.

(A) Body weight of WT and MPC SkmKO mice on HFD (n = 7–8, littermates, age 9–58 weeks, two tailed t-test). (B) Daily food intake by WT and MPC SkmKO mice on HFD, normalized to body weight (BW) (n = 5–6, littermates, age = 30 weeks, two tailed t-test). (C) Daily food intake by WT and MPC SkmKO mice on NFD, normalized to body weight (BW) (n = 5–6, littermates, age = 61 weeks, two tailed t-test). Data are presented as mean ± SEM.

https://doi.org/10.7554/eLife.45873.018
Figure 6—figure supplement 2
Physiological parameters in MPC iSkmKO mice.

(A) Relative western blot of Mpc1 and Mpc2 protein abundance in TA muscles of WT and MPC iSkmKO mice. Loading was normalized to total protein. Tubulin was used as a reference loading control. (n = 8, littermates, age = 36 weeks, two tailed t-test). (B) Adipose depot weight in WT and MPC iSkmKO mice (n = 6–7, littermates, age = 36 weeks, two tailed t-test). (C) Muscle mass/Tibia length of WT and MPC iSkmKO hindlimb muscles (n = 6–7, littermates, age = 36 weeks, two tailed t-test). (D) Daily food intake by WT and MPC iSkmKO mice on chow diet, normalized to body weight (BW) (n = 6–7, littermates, age = 25 weeks, two tailed t-test). Data are presented as mean ± SEM (**p<0.01).

https://doi.org/10.7554/eLife.45873.019
Skeletal muscle MPC disruption drives Cori cycling and fatty acid oxidation.

Skeletal muscle MPC disruption (MPC SkmKO) impairs glycolytically generated pyruvate entry into skeletal muscle mitochondria, thereby increasing conversion of pyruvate to lactate and consequent skeletal muscle lactate excretion. Increased skeletal muscle lactate excretion drives hepatic gluconeogenesis that re-supplies glucose to skeletal muscle. Thus, skeletal muscle MPC disruption increases Cori Cycling. The Cori Cycle is energetically futile because each round produces two skeletal muscle ATP molecules and consumes six liver ATP equivalents, for a net whole-body consumption of 4 ATP equivalents. Because hepatic gluconeogenesis is energetically supported by fatty acid oxidation and muscle MPC disruption increases muscle fatty acid oxidation, futile Cori Cycling is energetically supported by fatty acid oxidation. Together, increased energy expenditure and fatty acid oxidation contribute to leanness arising from skeletal muscle MPC disruption.

https://doi.org/10.7554/eLife.45873.021

Tables

Key resources table
Reagent
type (species)
or resource
DesignationSource or
reference
IdentifiersAdditional
information
Genetic reagent (Mus musculus)Mpc1flox/flox, C57BL/6JGray et al., 2015
Genetic reagent (Mus musculus)Myogenin-Cre, C57BL/6JLi et al., 2005Gift from Dr. Eric Olsen
Genetic reagent (Mus musculus)HSA-MerCreMer, C57BL/6JMcCarthy et al., 2012Gift from Dr. Karyn Esser
AntibodyRabbit monoclonal anti-MPC1ProteintechS4154-2Gift from Dr. Brian Finck; (1:1000)
AntibodyRabbit monoclonal anti-MPC2 (D4I7G)Cell Signaling Technology#46141, RRID:AB_2799295(1:1000)
AntibodyRabbit monoclonal anti-VDAC (D73D12)Cell Signaling Technology#4661, RRID:AB_10557420(1:1000)
AntibodyMouse monoclonal anti-Actin (AC-15)Sigma#A5441, RRID:AB_476744(1:10000)
AntibodyRabbit monoclonal anti-Tubulin (DM1A)Cell Signaling Technology#3873S, RRID:AB_1904178(1:1000)
AntibodyRabbit monoclonal anti-HSP90Cell Signaling#4874, RRID:AB_2121214(1:1000)
AntibodyTotal OXPHOS cocktailAbcamab110413, RRID:AB_2629281(1:1000)
AntibodyMouse monoclonal anti-AMPKα (F6)Cell Signaling Technology#2793, RRID:AB_915794(1:1000)
AntibodyRabbit monoclonal anti-pAMPKα (Thr172) (40H9)Cell Signaling Technology#2535, RRID:AB_331250(1:1000)
AntibodyRabbit monoclonal anti-AKT (pan) (11E7)Cell Signaling Technology#4685, RRID:AB_2225340(1:1000)
AntibodyRabbit polyclonal anti-pAKT (Ser473)Cell Signaling Technology#9271, RRID:AB_329825(1:1000)
AntibodyMouse monoclonal anti-PDH-E1α (D6)Santa Cruz Biotechnology#SC-377092, RRID:AB_2716767(1:1000)
AntibodyRabbit polyclonal anti-pPDH-E1α (Ser232)Millipore Sigma#AP1063, RRID:AB_10616070(1:1000)
AntibodyRabbit monoclonal anti-GAPDH (D16H11)Cell Signaling Technology#5174, RRID:AB_10622025(1:20000)
AntibodyGoat anti-Mouse Dylight 800ThermoFisherSA5-10176, RRID:AB_2556756(1:10000)
AntibodyDonkey anti-Rabbit DyLight 680ThermoFisherSA5-10042, RRID:AB_2556622(1:5000)
AntibodyGoat anti-Rabbit DyLight 800ThermoFisher#35571, RRID:AB_614947(1:10000)
Sequence-based reagent36b4Forward: 5'-CGTCCTCGTTGGAGTGACAReverse: 5'-CGGTGCGTCAGGGATTG
Sequence-based reagentMpc1Forward: 5'-AACTACGAGATGAGTAAGCGGCReverse: 5'-GTGTTTTCCCTTCAGCACGAC
Sequence-based reagentMpc2Forward: 5'-CCGCTTTACAACCACCCGGCAReverse: 5'-CAGCACACACCAATCCCCATTTCA
Sequence-based reagentCpt1bForward: 5'-GGTCCCATAAGAAACAAGACCTCCReverse: 5'-CAGAAAGTACCTCAGCCAGGAAAG
Sequence-based reagentHadhaForward: 5'-TGGATGTGGATGACATTGCTReverse: 5'-GGGGAAGAGTATCGGCTAGG
Sequence-based reagentEchs1Forward: 5'-CTTCACTGTAAGGGCAGGTGReverse: 5'-CTTGAGTTGGGAATCAGCAG
Commercial assay or kitFGF21 ELISA kitThermoFisherNC9903102
Commercial assay or kitGlucose Assay KitSigma-AldrichHK20
Commercial assay or kitHigh-Capacity cDNA Reverse Transcription kitApplied Biosystems4368814
Commercial assay or kitInfinity Cholesterol ReagentThermo ScientificTR13421
Commercial assay or kitInfinity Triglyceride ReagentThermo ScientificTR22421
Commercial assay or kitLeptin ELISA kitR and D SystemsMOB00
Commercial assay or kitSerum Ketone KitWako Diagnostics415–73301, 411–73401, 412–73791
Commercial assay or kitSerum NEFA kitWako Diagnostics999–34691, 995–034791, 991–34891, 993–35191, 276–76491
Commercial assay or kitUltra-sensitive Mouse Insulin ELISA kitCrystal Chem90080
Commercial assay or kitInsulin chemiluminescence ELISAAmerican Laboroatory Products#80-INSMR-CH01
Chemical compound, drug[1–14C]−2-deoxy-D-glucosePerkin ElmerNEC495001MCSBF3
Chemical compound, drugα-Cyano-4-hydroxycinnamic acid (CHC)Sigma-Aldrich476870
Chemical compound, drug13C3-Sodium Pyruvate (99%)Cambridge Isotope Laboratories142014-11-17
Chemical compound, drug13C3-Sodium-L-Lactate (98%)Cambridge Isotope LaboratoriesCLM-1579
Chemical compound, drug14C-Palmitic acidPerkin ElmerNEC075H250UC
Chemical compound, drug14C-Sodium PyruvatePerkin ElmerNEC256050UC
Chemical compound, drugAntimycin ASigma-AldrichA8674
Chemical compound, drugD-[3-3H]-glucosePerkin ElmerNET331C001MC SBF3
Chemical compound, drugInsulinNovo NordiskNovolin R
Chemical compound, drugDeoxy-D-glucose 2-[1,2-3H(N)]Perkin ElmerNET328A001MC
Chemical compound, drugEnsureAbbottVanilla-57g
Chemical compound, drugFCCPSigma-AldrichC2920
Chemical compound, drugHigh Fat Diet 60%Kcal from fat (HFD)Research Diets IncD12492
Chemical compound, drugLow Fat Diet 10% Kcal from fat (control diet, NFD)Research Diets IncD12450J
Chemical compound, drugNormal Chow/Teklad Global Soy Protein-Free Extruded Rodent Diet Irradiated (NCD)Envigo2920X
Chemical compound, drugOligomycin ASigma-Aldrich75351–5 MG
Chemical compound, drugRotenoneSigma-AldrichR8875-1G
Chemical compound, drugTamoxifenSigma-AldrichT5648-1G
Chemical compound, drugTriolein, [9,10-3H(N)]Perkin ElmerNET431001MC
Chemical compound, drugUK-5099ThermoFisher418610
Chemical compound, drugAcetyl coenzyme A sodium saltSigma-AldrichA2056-10MG
Chemical compound, drug3H-Acetyl-CoAPerkin ElmerNET290050UC
Chemical compound, drugMalonyl coenzyme A lithium saltSigma-AldrichM4263-5MG
Chemical compound, drugPurified chicken fatty acid synthaseDavid Thomson Lab, BYU
Chemical compound, drugProtease ArrestG Biosciences786–437
OtherAccupulserWorld Precision InstrumentA310 Accupulser
OtherBody Composition AnalyzerBrukerLF50-BCA Analyzer
OtherCallipersMSC Industrial Supply35518166
OtherGlucose MeterLifeScanOnetouch ulta mini
OtherGlucose StripsDiabetic ExpressFeb-67
OtherHeadstageAxon InstrumentsCV 203BV
OtherIntegrating patch clampAxon InstrumentsAxopatch 200B
OtherLactate MeterNova BiomedicalLactate plus meter
OtherLactate StripsNova BiomedicalNC0071872
OtherLow noise data acquisition systemAxon InstrumentsAxon digidata 1550
OtherLow noise data acquisition systemAxon InstrumentsAxon digidata 1440A
OtherMicrovette blood collection tubesSarstedt IncNC9141704
OtherOhaus Hand-Held ScalesThermoFisherS65222
OtherPromethion CagesSable Systems InternationalPromethion Line
OtherRodent TreadmillColumbus InstrumentsExer 3/6 Treadmill
OtherStimulus IsolaterWorld Precision InstrumentStimulus Isolater
OtherWestern Blot ImagerLi-CorOdyssey CLx
OtherPlatformAurora Scientific809B
OtherForce tranducerAurora Scientific305C
OtherRefridgerated/Heated bath circulatorThermo Fisher6200 R20F
OtherXF96 4-port FluxPak with PET MicroplatesSeahorse Bioscience102416–100
Software/toolSigmaplotSigmaplotRRID:SCR_003210
Software/toolExcelMicrosoftRRID:SCR_016137

Additional files

Supplementary file 1

TA muscle metabolomic profiles after sham and in situ contraction. n = 5–6, littermates, age = 12 weeks, two way ANOVA.

https://doi.org/10.7554/eLife.45873.022
Supplementary file 2

Percent isotopologue distribution of TCA cycle intermediates in muscle and liver 65 min after U13C-labeled glucose injection of WT and MPC SkmKO mice.

(n = 8, 7 littermates one non-littermate, age = 12 weeks, two tailed t-test) Data are presented as mean ± SEM (*p<0.05, **p<0.01, ***p<0.001).

https://doi.org/10.7554/eLife.45873.023
Transparent reporting form
https://doi.org/10.7554/eLife.45873.024

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Arpit Sharma
  2. Lalita Oonthonpan
  3. Ryan D Sheldon
  4. Adam J Rauckhorst
  5. Zhiyong Zhu
  6. Sean C Tompkins
  7. Kevin Cho
  8. Wojciech J Grzesik
  9. Lawrence R Gray
  10. Diego A Scerbo
  11. Alvin D Pewa
  12. Emily M Cushing
  13. Michael C Dyle
  14. James E Cox
  15. Chris Adams
  16. Brandon S Davies
  17. Richard K Shields
  18. Andrew W Norris
  19. Gary Patti
  20. Leonid V Zingman
  21. Eric B Taylor
(2019)
Impaired skeletal muscle mitochondrial pyruvate uptake rewires glucose metabolism to drive whole-body leanness
eLife 8:e45873.
https://doi.org/10.7554/eLife.45873