Acute RyR1 Ca2+ leak enhances NADH-linked mitochondrial respiratory capacity

Nadège Zanou, Haikel Dridi, Steven Reiken, Tanes Imamura de Lima, Chris Donnelly, Umberto De Marchi, Manuele Ferrini, Jeremy Vidal, Leah Sittenfeld, Jerome N Feige, Pablo M Garcia-Roves, Isabel C Lopez-Mejia, Andrew R Marks, Johan Auwerx, Bengt Kayser, Nicolas Place, Nadège Zanou, Haikel Dridi, Steven Reiken, Tanes Imamura de Lima, Chris Donnelly, Umberto De Marchi, Manuele Ferrini, Jeremy Vidal, Leah Sittenfeld, Jerome N Feige, Pablo M Garcia-Roves, Isabel C Lopez-Mejia, Andrew R Marks, Johan Auwerx, Bengt Kayser, Nicolas Place

Abstract

Sustained ryanodine receptor (RyR) Ca2+ leak is associated with pathological conditions such as heart failure or skeletal muscle weakness. We report that a single session of sprint interval training (SIT), but not of moderate intensity continuous training (MICT), triggers RyR1 protein oxidation and nitrosylation leading to calstabin1 dissociation in healthy human muscle and in in vitro SIT models (simulated SIT or S-SIT). This is accompanied by decreased sarcoplasmic reticulum Ca2+ content, increased levels of mitochondrial oxidative phosphorylation proteins, supercomplex formation and enhanced NADH-linked mitochondrial respiratory capacity. Mechanistically, (S-)SIT increases mitochondrial Ca2+ uptake in mouse myotubes and muscle fibres, and decreases pyruvate dehydrogenase phosphorylation in human muscle and mouse myotubes. Countering Ca2+ leak or preventing mitochondrial Ca2+ uptake blunts S-SIT-induced adaptations, a result supported by proteomic analyses. Here we show that triggering acute transient Ca2+ leak through RyR1 in healthy muscle may contribute to the multiple health promoting benefits of exercise.

Conflict of interest statement

U.D.M. and J.N.F. are employees of Nestlé Research, which is part of the Société des Produits Nestlé SA. A.R.M. is chair of the SAB and holds stock in ARMGO Pharma Inc., a biotech company developing drugs targeting RyR channel leak. The remaining authors declare no competing interests.

© 2021. The Author(s).

Figures

Fig. 1. One session of SIT induces…
Fig. 1. One session of SIT induces RyR1 post-translational modifications, calstabin1 dissociation from the RyR1 complex and leads to increased OXPHOS protein expression in human muscle.
a Models of MICT (a1) and SIT (a2) in humans. b, c Assessment of b knee extensor maximal voluntary contraction (MVC) force; n = 8 participants per group and c the ratio of electrically evoked forces at 10 and 100 Hz; n = 7 and 8 participants for MICT and SIT, respectively. Two-way ANOVA followed by Sidak’s multiple comparisons test. d Representative immunoblots (IB) of immunoprecipitated (IP) RyR1, RyR1 post-translational modifications and calstabin1 dissociation. DNP (2,4-dinitrophenylhydrazone): RyR1 oxidation. P RyR1 Ser2844: RyR1 phosphorylation at serine 2844. Cys NO: RyR1 nitrosylation. SK SR: skeletal muscle sarcoplasmic reticulum vesicle. SK SR treated with 200 µM H2O2, 250 µM NOC-12 and 5 units PKA per reaction: positive control for RyR1 oxidation and nitrosylation and calstabin1 dissociation. No antibody in IP: negative control. The whole gel and an additional control are shown in Supplementary Fig. 1f. eh Quantification of immunoblots in (d); n = 8 participants per group. Two-way ANOVA followed by Sidak’s multiple comparisons test. i Representative immunoblots of PGC-1α. j Representative immunoblots of mitochondrial OXPHOS proteins. All the cropped parts of OXPHOS proteins are part of the same blot that is shown in Supplementary Fig. 5. k Quantification of PGC-1α proteins in (i) related to GAPDH protein and expressed as % of Pre-MICT; n = 8 participants per group. Two-way ANOVA. lp Quantification of OXPHOS proteins in (j) related to GAPDH protein and expressed as % of Pre-MICT; n = 8 participants per group. Two-way ANOVA, followed by Sidak’s multiple comparisons test (l, m). Data are mean ± SD. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, and $main effect of time. Source data are provided as a Source Data file.
Fig. 2. S-SIT in C2C12 myotubes induces…
Fig. 2. S-SIT in C2C12 myotubes induces RyR1 post-translational modifications, calstabin1 dissociation and decreased SR Ca2+ content.
a S-MICT (a1) and S-SIT (a2) models in C2C12 myotubes. b Metabolomic analysis immediately after stimulation showing changes expressed in % of CTRL values; n = 2 independent biological experiments of six technical replicates each. cf Representative immunoblots (c) and quantifications (df) of RyR1 immunoprecipitation and assessment of RyR1 post-translational modifications immediately after stimulation. DNP (2,4-dinitrophenylhydrazone): RyR1 oxidation. Cys NO: RyR1 nitrosylation. SK SR: skeletal muscle sarcoplasmic reticulum vesicle. SK SR treated with 200 µM H2O2 and 250 µM NOC-12: positive control for RyR1 oxidation, nitrosylation and calstabin1 dissociation. No antibody in IP: negative control; n = 4 independent biological experiments per group. One-way ANOVA followed by Tukey’s multiple comparisons test and Kruskal–Wallis ANOVA, followed by Dunn’s multiple comparison test (e). The whole gel and an additional control are shown in Supplementary Fig. 2g. gh Original recording (g) and quantifications (h) of normalized Fluo-4 fluorescence in response to 2.5 mM caffeine immediately after S-MICT or S-SIT; n = 7 independent biological experiments per group. One-way ANOVA followed by Tukey’s multiple comparisons test. i, j Fluorescence resonance energy-based assessment of SR Ca2+ content (i) and quantifications of D1ER ratio (j) immediately after S-MICT and S-SIT (i). (i1) D1ER plasmid distribution recorded at 535 nm. (i2) Detail of the reticular pattern of the D1ER signal. (i3) Ca2+ distribution pattern is rendered in pseudocolor scale (from dark blue, low Ca2+ level to red, high Ca2+ level, see arrow); n = 3 independent biological experiments per group. One-way ANOVA followed by Tukey’s multiple comparisons test. kl Representative immunoblots (k) and quantification (l) of STIM1 after S-MICT and S-SIT; n = 3 independent biological experiments per group. One-way ANOVA. mo Characterization of store-operated Ca2+ entry (SOCE) in S-MICT and S-SIT myotubes immediately after stimulation. Original recording of SOCE (m); SR Ca2+ store quantifications (n) and SOCE quantifications (o); n = 3 independent biological experiments per group. One-way ANOVA. Data are mean ± SD. *p ≤ 0.05, **p ≤ 0.01 and ***p ≤ 0.001. Source data are provided as a Source Data file.
Fig. 3. S-SIT in C2C12 myotubes induces…
Fig. 3. S-SIT in C2C12 myotubes induces higher mitochondrial protein content, supercomplexes levels and respiratory capacity as compared to MICT, which are blunted by S107-induced RyR1 stabilization.
ad Representative immunoblots (a) and quantifications (bd) of immunoprecipitated RyR1 post-translational modifications 3 h post stimulation without or with S107 treatment for 3 h; n = 4 (CTRL) and 3 (S-MICT, S-SIT) independent biological experiments. One-way ANOVA, followed by Tukey’s multiple comparisons test (c, d). e Quantification of Fluo-4/AM fluorescence ratio 3 h post stimulation without or with S107; n = 4 (CTRL, S-SIT) and 3 (S-SIT S107) independent biological experiments. One-way ANOVA followed by Tukey’s multiple comparisons test. fk Representative immunoblots (f) and quantifications (gk) of mitochondrial OXPHOS proteins 72 h post stimulation without or with S107 treatment for 72 h. Whole OXPHOS blot shown in Supplementary Fig. 6; n = 7 (CTRL), 6 (S-MICT, S-SIT) and 3 (S-MICT S107, S-SIT S107) independent biological experiments. One-way ANOVA followed by Tukey’s multiple comparisons test. lp Immunoblots (l) and quantification (mp) of supercomplexes after stimulations; n = 3 independent biological experiments per group. One-way ANOVA followed by Tukey’s multiple comparisons test (mo). qr MitoTracker red fluorescence (q) and quantification of mitochondrial area (r) 72 h post stimulation without or with S107 treatment; n = 6 independent biological experiments per group. One-way ANOVA with Sidak’s multiple comparisons test. sw O2 flux per mass (pmol/s/mg of protein) 72 h after stimulation and S107 treatment. s MPL: malate pyruvate leak state. t NP: N-linked OXPHOS state with ADP-stimulated. u NSP: N- and S-OXPHOS pathways. v NSE: ET state, noncoupled and w SE, S-pathway; n = 5 (CTRL, S-SIT), 4 (S-MICT) and 3 (S-SIT S107) independent biological experiments. One-way ANOVA with Sidak’s multiple comparisons test. xz NADH (x), NAD (y) concentration and NADH/NAD ratio (z) in S-SIT myotubes without or with S107 treatment for 72 h; n = 6 independent biological experiments per group. Unpaired t test. For S107 groups, 10 µM of S107 treatment was applied immediately after stimulation for the indicated time. Data are mean ± SD. *p ≤ 0.05, **p ≤ 0.01 and ***p ≤ 0.001. Source data are provided as a Source Data file.
Fig. 4. Proteomic analysis showing the global…
Fig. 4. Proteomic analysis showing the global mapping of RyR1 Ca2+ leak on muscle adaptations to S-SIT.
ac Proteomic analysis of protein groups related to Gene Ontology Biological Processes (GoBP), Molecular Function (GoMF) and Cellular Component (GoCC) that are significantly decreased S-SIT after 10 µM S107 treatment for 72 h (the treatment was applied immediately after stimulation). Protein groups exceeding 400 proteins were excluded. The median values of S-SIT − S-SIT S107 difference were calculated, and a score affected to the amplitude of the difference. The positive scores display the pathways significantly inhibited by S107 treatment. n = 5 per group. Benjamini–Hochberg corrected t test. Source data are provided as a Source Data file.
Fig. 5. (S-)SIT-induced leaky RyR1 increases mitochondrial…
Fig. 5. (S-)SIT-induced leaky RyR1 increases mitochondrial Ca2+ uptake and decreases PDH phosphorylation levels in muscle cells.
ae Normalized Rhod-2 fluorescence imaging in C2C12 myotubes. A 300 s time-lapse confocal recording of a bout of slightly modified S-SIT and S-MICT protocols: b S-MICT and c S-SIT. d, e Mitochondrial Ca2+ uptake in S-SIT myotubes pre-treated with 10 µM S107 for 1 h (d) or 20 µM mitoxantrone (MTX) (e). Horizontal black lines indicate the periods of stimulation; vertical red arrows indicate the amplitude of mitochondrial Ca2+ at the end of the recordings. fg Maximal amplitude of normalized Rhod-2 fluorescence during stimulations (f) and 80 s after the end of S-MICT and S-SIT stimulations (g) in (ae); n = 5 (CTRL, S-SIT and S-SIT S107) and 3 (S-MICT, S-SIT MTX) independent biological experiments. One-way ANOVA followed by Tukey’s multiple comparisons test. hj Normalized Rhod-2 fluorescence imaging in mouse FDB intact single muscle fibres. Same protocols as described for (ac), except that the voltage was 40 V. kl Maximal amplitude of normalized Rhod-2 fluorescence in FDB muscle fibres in (hj) during (k) and 80 s after the end (l) of S-MICT and S-SIT stimulations; n = 3 mice per group. One-way ANOVA followed by Tukey’s multiple comparisons test. mn Representative immunoblots (m) and quantification (n) of phosphorylated PDH E1α at serine 293 related to total PDH E1α in myotubes 1 h after stimulation; 10 µM S107 or 20 µM MTX were applied after the stimulation for 1 h when indicated; n = 7 (CTRL, S-MICT, S-SIT and S-SIT S107) and 3 (S-SIT MTX) independent biological experiments. One-way ANOVA followed by Sidak’s multiple comparisons test. o, p Representative immunoblots (o) and quantification (p) of phosphorylated PDH E1α at serine 293 related to total PDH E1α in human muscles; n = 8 participants per group. Two-way ANOVA followed by Sidak’s multiple comparisons test. q Proposed schematic of RyR1 Ca2+ leak-activated mitochondrial PDH E1α dephosphorylation in response to S-SIT. Data are mean ± SD. *p ≤ 0.05, **p ≤ 0.01 and ***p ≤ 0.001. Source data are provided as a Source Data file.
Fig. 6. Mitochondrial Ca 2+ uptake contributes…
Fig. 6. Mitochondrial Ca2+ uptake contributes to mitochondrial adaptations to S-SIT.
a Immunoblots of MCU, RyR1, SERCA1, GAPDH and calstabin1 in si-MCU (siRNAs directed against MCU) compared to si-CTRL (negative control siRNAs) myotubes. b Original recordings of normalized Rhod-2 fluorescence imaging in si-MCU compared to si-CTRL myotubes at 72 h post transfection. c Representative immunoblots of mitochondrial OXPHOS and MCU proteins expression in S-SIT si-MCU myotubes at 72 h post stimulation (cells were transfected immediately after the stimulation with the siRNAs against MCU) compared to S-SIT si-CTRL myotubes (cells were transfected immediately after the stimulation with the negative control siRNAs) and si-CTRL (non-stimulated myotubes transfected with the negative control siRNAs). All the cropped parts of OXPHOS proteins are part of the same blot that is shown in Supplementary Fig. 6. di Quantifications of the immunoblots in (c) related to total protein and expressed as % of si-CTRL; n = 6 independent biological experiments per group. One-way ANOVA followed by Tukey’s multiple comparisons test (d, e, h). Data are mean ± SD. *p ≤ 0.05, **p ≤ 0.01 and ***p ≤ 0.001. Source data are provided as a Source Data file.

References

    1. Flucher BE, et al. Triad formation: organization and function of the sarcoplasmic reticulum calcium release channel and triadin in normal and dysgenic muscle in vitro. J. Cell Biol. 1993;123:1161–1174.
    1. Marks AR, et al. Molecular cloning and characterization of the ryanodine receptor/junctional channel complex cDNA from skeletal muscle sarcoplasmic reticulum. Proc. Natl Acad. Sci. USA. 1989;86:8683–8687.
    1. Awad SS, Lamb HK, Morgan JM, Dunlop W, Gillespie JI. Differential expression of ryanodine receptor RyR2 mRNA in the non-pregnant and pregnant human myometrium. Biochem. J. 1997;322:777–783.
    1. Nakashima Y, et al. Molecular cloning and characterization of a human brain ryanodine receptor. FEBS Lett. 1997;417:157–162.
    1. Zhang L, et al. Functional SNP in the microRNA-367 binding site in the 3’UTR of the calcium channel ryanodine receptor gene 3 (RYR3) affects breast cancer risk and calcification. Proc. Natl Acad. Sci. USA. 2011;108:13653–13658.
    1. Zalk R, et al. Structure of a mammalian ryanodine receptor. Nature. 2015;517:44–49.
    1. Santulli G, Marks AR. Essential roles of intracellular calcium release channels in muscle, brain, metabolism, and aging. Curr. Mol. Pharm. 2015;8:206–222.
    1. Marks AR, Marx SO, Reiken S. Regulation of ryanodine receptors via macromolecular complexes: a novel role for leucine/isoleucine zippers. Trends Cardiovasc. Med. 2002;12:166–170.
    1. Lehnart SE, et al. Phosphodiesterase 4D deficiency in the ryanodine-receptor complex promotes heart failure and arrhythmias. Cell. 2005;123:25–35.
    1. Tilgen N, et al. Identification of four novel mutations in the C-terminal membrane spanning domain of the ryanodine receptor 1: association with central core disease and alteration of calcium homeostasis. Hum. Mol. Genet. 2001;10:2879–2887.
    1. Santulli G, Xie W, Reiken SR, Marks AR. Mitochondrial calcium overload is a key determinant in heart failure. Proc. Natl Acad. Sci. USA. 2015;112:11389–11394.
    1. Bellinger AM, et al. Hypernitrosylated ryanodine receptor calcium release channels are leaky in dystrophic muscle. Nat. Med. 2009;15:325–330.
    1. Waning DL, Guise TA. Cancer-associated muscle weakness: What’s bone got to do with it? Bonekey Rep. 2015;4:691.
    1. Andersson DC, et al. Ryanodine receptor oxidation causes intracellular calcium leak and muscle weakness in aging. Cell Metab. 2011;14:196–207.
    1. Ivarsson N, et al. SR Ca(2+) leak in skeletal muscle fibers acts as an intracellular signal to increase fatigue resistance. J. Gen. Physiol. 2019;151:567–577.
    1. Erikssen G, et al. Changes in physical fitness and changes in mortality. Lancet. 1998;352:759–762.
    1. WHO. WHO guidelines on physical activity and sedentary behaviour. (2020).
    1. Shaw, K., Gennat, H., O’Rourke, P. & Del Mar, C. Exercise for overweight or obesity. Cochrane Database Syst. Rev. 10.1002/14651858.CD003817.pub3 (2006).
    1. Pedersen BK, Saltin B. Exercise as medicine—evidence for prescribing exercise as therapy in 26 different chronic diseases. Scand. J. Med. Sci. Sports. 2015;25(Suppl. 3):1–72.
    1. Bhatia C, Kayser B. Preoperative high-intensity interval training is effective and safe in deconditioned patients with lung cancer: a randomized clinical trial. J. Rehabil. Med. 2019;51:712–718.
    1. Cuddy, T. F., Ramos, J. S. & Dalleck, L. C. Reduced exertion high-intensity interval training is more effective at improving cardiorespiratory fitness and cardiometabolic health than traditional moderate-intensity continuous training. Int. J. Environ. Res. Public Health16, 10.3390/ijerph16030483 (2019).
    1. Robinson MM, et al. Enhanced protein translation underlies improved metabolic and physical adaptations to different exercise training modes in young and old humans. Cell Metab. 2017;25:581–592.
    1. Burgomaster KA, et al. Similar metabolic adaptations during exercise after low volume sprint interval and traditional endurance training in humans. J. Physiol. 2008;586:151–160.
    1. Metcalfe RS, Babraj JA, Fawkner SG, Vollaard NB. Towards the minimal amount of exercise for improving metabolic health: beneficial effects of reduced-exertion high-intensity interval training. Eur. J. Appl. Physiol. 2012;112:2767–2775.
    1. Wright DC, et al. Exercise-induced mitochondrial biogenesis begins before the increase in muscle PGC-1alpha expression. J. Biol. Chem. 2007;282:194–199.
    1. Place N, et al. Ryanodine receptor fragmentation and sarcoplasmic reticulum Ca2+ leak after one session of high-intensity interval exercise. Proc. Natl Acad. Sci. USA. 2015;112:15492–15497.
    1. Marx SO, et al. PKA phosphorylation dissociates FKBP12.6 from the calcium release channel (ryanodine receptor): defective regulation in failing hearts. Cell. 2000;101:365–376.
    1. Schlittler M, et al. Three weeks of sprint interval training improved high-intensity cycling performance and limited ryanodine receptor modifications in recreationally active human subjects. Eur. J. Appl. Physiol. 2019;119:1951–1958.
    1. Westerblad H, Duty S, Allen DG. Intracellular calcium concentration during low-frequency fatigue in isolated single fibers of mouse skeletal muscle. J. Appl. Physiol. 1993;75:382–388.
    1. Granata C, Jamnick NA, Bishop DJ. Training-induced changes in mitochondrial content and respiratory function in human skeletal muscle. Sports Med. 2018;48:1809–1828.
    1. Trewin AJ, et al. Acute HIIE elicits similar changes in human skeletal muscle mitochondrial H2O2 release, respiration, and cell signaling as endurance exercise even with less work. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2018;315:R1003–R1016.
    1. Fiorenza M, et al. High-intensity exercise training enhances mitochondrial oxidative phosphorylation efficiency in a temperature-dependent manner in human skeletal muscle: implications for exercise performance. FASEB J. 2019;33:8976–8989.
    1. Lin J, et al. Transcriptional co-activator PGC-1 alpha drives the formation of slow-twitch muscle fibres. Nature. 2002;418:797–801.
    1. Canto C, et al. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature. 2009;458:1056–1060.
    1. Pilegaard H, Saltin B, Neufer PD. Exercise induces transient transcriptional activation of the PGC-1alpha gene in human skeletal muscle. J. Physiol. 2003;546:851–858.
    1. Mathai AS, Bonen A, Benton CR, Robinson DL, Graham TE. Rapid exercise-induced changes in PGC-1alpha mRNA and protein in human skeletal muscle. J. Appl Physiol. (1985) 2008;105:1098–1105.
    1. Ruas JL, et al. A PGC-1alpha isoform induced by resistance training regulates skeletal muscle hypertrophy. Cell. 2012;151:1319–1331.
    1. Wright DC, Geiger PC, Han DH, Jones TE, Holloszy JO. Calcium induces increases in peroxisome proliferator-activated receptor gamma coactivator-1alpha and mitochondrial biogenesis by a pathway leading to p38 mitogen-activated protein kinase activation. J. Biol. Chem. 2007;282:18793–18799.
    1. Axelrod CL, Fealy CE, Mulya A, Kirwan JP. Exercise training remodels human skeletal muscle mitochondrial fission and fusion machinery towards a pro-elongation phenotype. Acta Physiol. 2019;225:e13216.
    1. Wai T, et al. Imbalanced OPA1 processing and mitochondrial fragmentation cause heart failure in mice. Science. 2015;350:aad0116.
    1. Yang C, Svitkina TM. Ultrastructure and dynamics of the actin-myosin II cytoskeleton during mitochondrial fission. Nat. Cell Biol. 2019;21:603–613.
    1. Huertas JR, Casuso RA, Agustin PH, Cogliati S. Stay fit, stay young: mitochondria in movement: the role of exercise in the new mitochondrial paradigm. Oxid. Med. Cell Longev. 2019;2019:7058350.
    1. Nikolic N, et al. Electrical pulse stimulation of cultured skeletal muscle cells as a model for in vitro exercise—possibilities and limitations. Acta Physiol. 2017;220:310–331.
    1. Burch N, et al. Electric pulse stimulation of cultured murine muscle cells reproduces gene expression changes of trained mouse muscle. PLoS ONE. 2010;5:e10970.
    1. Abdelmoez, A. M. et al. Comparative profiling of skeletal muscle models reveals heterogeneity of transcriptome and metabolism. Am. J. Physiol. Cell Physiol.10.1152/ajpcell.00540.2019 (2019).
    1. Olsson K, et al. Intracellular Ca(2+)-handling differs markedly between intact human muscle fibers and myotubes. Skelet. Muscle. 2015;5:26.
    1. Mihaylova MM, Shaw RJ. The AMPK signalling pathway coordinates cell growth, autophagy and metabolism. Nat. Cell Biol. 2011;13:1016–1023.
    1. Westerblad H, Allen DG. The effects of intracellular injections of phosphate on intracellular calcium and force in single fibres of mouse skeletal muscle. Pflug. Arch. 1996;431:964–970.
    1. Beard NA, Laver DR, Dulhunty AF. Calsequestrin and the calcium release channel of skeletal and cardiac muscle. Prog. Biophys. Mol. Biol. 2004;85:33–69.
    1. De Marchi U, Castelbou C, Demaurex N. Uncoupling protein 3 (UCP3) modulates the activity of Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) by decreasing mitochondrial ATP production. J. Biol. Chem. 2011;286:32533–32541.
    1. Kiviluoto S, et al. STIM1 as a key regulator for Ca2+ homeostasis in skeletal-muscle development and function. Skelet. Muscle. 2011;1:16.
    1. Matecki S, et al. Leaky ryanodine receptors contribute to diaphragmatic weakness during mechanical ventilation. Proc. Natl Acad. Sci. USA. 2016;113:9069–9074.
    1. Reiken S, et al. PKA phosphorylation activates the calcium release channel (ryanodine receptor) in skeletal muscle: defective regulation in heart failure. J. Cell Biol. 2003;160:919–928.
    1. Letts JA, Sazanov LA. Clarifying the supercomplex: the higher-order organization of the mitochondrial electron transport chain. Nat. Struct. Mol. Biol. 2017;24:800–808.
    1. Genova ML, Lenaz G. Functional role of mitochondrial respiratory supercomplexes. Biochim. Biophys. Acta. 2014;1837:427–443.
    1. Greggio C, et al. Enhanced respiratory chain supercomplex formation in response to exercise in human skeletal muscle. Cell Metab. 2017;25:301–311.
    1. Sun, D., Li, B., Qiu, R., Fang, H. & Lyu, J. Cell type-specific modulation of respiratory chain supercomplex organization. Int. J. Mol. Sci. 17, 10.3390/ijms17060926 (2016).
    1. Larsen S, et al. Biomarkers of mitochondrial content in skeletal muscle of healthy young human subjects. J. Physiol. 2012;590:3349–3360.
    1. Canto C, Garcia-Roves PM. High-resolution respirometry for mitochondrial characterization of ex vivo mouse tissues. Curr. Protoc. Mouse Biol. 2015;5:135–153.
    1. Pilegaard H, et al. Influence of pre-exercise muscle glycogen content on exercise-induced transcriptional regulation of metabolic genes. J. Physiol. 2002;541:261–271.
    1. Jannas-Vela S, et al. Assessment of Na+/K+ ATPase activity in small rodent and human skeletal muscle samples. Med. Sci. Sports Exerc. 2019;51:2403–2409.
    1. Mall S, et al. The presence of sarcolipin results in increased heat production by Ca(2+)-ATPase. J. Biol. Chem. 2006;281:36597–36602.
    1. Periasamy M, Kalyanasundaram A. SERCA pump isoforms: their role in calcium transport and disease. Muscle Nerve. 2007;35:430–442.
    1. Bhupathy P, Babu GJ, Periasamy M. Sarcolipin and phospholamban as regulators of cardiac sarcoplasmic reticulum Ca2+ ATPase. J. Mol. Cell. Cardiol. 2007;42:903–911.
    1. Sahoo SK, Shaikh SA, Sopariwala DH, Bal NC, Periasamy M. Sarcolipin protein interaction with sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) is distinct from phospholamban protein, and only sarcolipin can promote uncoupling of the SERCA pump. J. Biol. Chem. 2013;288:6881–6889.
    1. Smith WS, Broadbridge R, East JM, Lee AG. Sarcolipin uncouples hydrolysis of ATP from accumulation of Ca2+ by the Ca2+-ATPase of skeletal-muscle sarcoplasmic reticulum. Biochem. J. 2002;361:277–286.
    1. Clapham DE. Calcium signaling. Cell. 2007;131:1047–1058.
    1. Zanou N, et al. Trpc1 ion channel modulates phosphatidylinositol 3-kinase/Akt pathway during myoblast differentiation and muscle regeneration. J. Biol. Chem. 2012;287:14524–14534.
    1. Dirksen RT. Sarcoplasmic reticulum-mitochondrial through-space coupling in skeletal muscle. Appl. Physiol. Nutr. Metab. 2009;34:389–395.
    1. Shoshan-Barmatz V, Krelin Y, Shteinfer-Kuzmine A. VDAC1 functions in Ca(2+) homeostasis and cell life and death in health and disease. Cell Calcium. 2018;69:81–100.
    1. Mammucari C, Raffaello A, Vecellio Reane D, Rizzuto R. Molecular structure and pathophysiological roles of the mitochondrial calcium uniporter. Biochim. Biophys. Acta. 2016;1863:2457–2464.
    1. Diaz-Vegas AR, et al. Mitochondrial calcium increase induced by RyR1 and IP3R channel activation after membrane depolarization regulates skeletal muscle metabolism. Front. Physiol. 2018;9:791.
    1. Arduino DM, et al. Systematic identification of MCU modulators by orthogonal interspecies chemical screening. Mol. Cell. 2017;67:711–723 e717.
    1. Williams GS, Boyman L, Chikando AC, Khairallah RJ, Lederer WJ. Mitochondrial calcium uptake. Proc. Natl Acad. Sci. USA. 2013;110:10479–10486.
    1. Collins TJ, Lipp P, Berridge MJ, Bootman MD. Mitochondrial Ca(2+) uptake depends on the spatial and temporal profile of cytosolic Ca(2+) signals. J. Biol. Chem. 2001;276:26411–26420.
    1. Santo-Domingo J, Demaurex N. Calcium uptake mechanisms of mitochondria. Biochim. Biophys. Acta. 2010;1797:907–912.
    1. Harris RA, Bowker-Kinley MM, Huang B, Wu P. Regulation of the activity of the pyruvate dehydrogenase complex. Adv. Enzym. Regul. 2002;42:249–259.
    1. Korotchkina LG, Patel MS. Site specificity of four pyruvate dehydrogenase kinase isoenzymes toward the three phosphorylation sites of human pyruvate dehydrogenase. J. Biol. Chem. 2001;276:37223–37229.
    1. Pilegaard H, et al. PDH-E1alpha dephosphorylation and activation in human skeletal muscle during exercise: effect of intralipid infusion. Diabetes. 2006;55:3020–3027.
    1. Howlett RA, et al. Regulation of skeletal muscle glycogen phosphorylase and PDH at varying exercise power outputs. Am. J. Physiol. 1998;275:R418–R425.
    1. Pan X, et al. The physiological role of mitochondrial calcium revealed by mice lacking the mitochondrial calcium uniporter. Nat. Cell Biol. 2013;15:1464–1472.
    1. Lander N, et al. Calcium-sensitive pyruvate dehydrogenase phosphatase is required for energy metabolism, growth, differentiation, and infectivity of Trypanosoma cruzi. J. Biol. Chem. 2018;293:17402–17417.
    1. Ghosh S, et al. An essential role for cardiolipin in the stability and function of the mitochondrial calcium uniporter. Proc. Natl Acad. Sci. USA. 2020;117:16383–16390.
    1. Carreras-Sureda A, et al. Non-canonical function of IRE1alpha determines mitochondria-associated endoplasmic reticulum composition to control calcium transfer and bioenergetics. Nat. Cell Biol. 2019;21:755–767.
    1. Boncompagni S, et al. Mitochondria are linked to calcium stores in striated muscle by developmentally regulated tethering structures. Mol. Biol. Cell. 2009;20:1058–1067.
    1. Jain SS, et al. High-fat diet-induced mitochondrial biogenesis is regulated by mitochondrial-derived reactive oxygen species activation of CaMKII. Diabetes. 2014;63:1907–1913.
    1. Rose AJ, Kiens B, Richter EA. Ca2+-calmodulin-dependent protein kinase expression and signalling in skeletal muscle during exercise. J. Physiol. 2006;574:889–903.
    1. Perry CG, et al. Repeated transient mRNA bursts precede increases in transcriptional and mitochondrial proteins during training in human skeletal muscle. J. Physiol. 2010;588:4795–4810.
    1. Skelly, L. E. et al. Human skeletal muscle fiber type-specific responses to sprint interval and moderate-intensity continuous exercise: acute and training-induced changes. J. Appl. Physiol.10.1152/japplphysiol.00862.2020 (2021).
    1. Granata C, Oliveira RS, Little JP, Renner K, Bishop DJ. Sprint-interval but not continuous exercise increases PGC-1alpha protein content and p53 phosphorylation in nuclear fractions of human skeletal muscle. Sci. Rep. 2017;7:44227.
    1. MacInnis MJ, Gibala MJ. Physiological adaptations to interval training and the role of exercise intensity. J. Physiol. 2017;595:2915–2930.
    1. Gibala MJ, et al. Short-term sprint interval versus traditional endurance training: similar initial adaptations in human skeletal muscle and exercise performance. J. Physiol. 2006;575:901–911.
    1. Flockhart M, et al. Excessive exercise training causes mitochondrial functional impairment and decreases glucose tolerance in healthy volunteers. Cell Metab. 2021;33:957–970 e956.
    1. Magistris MR, et al. Needle muscle biopsy in the investigation of neuromuscular disorders. Muscle Nerve. 1998;21:194–200.
    1. Zanou N, et al. Osmosensation in TRPV2 dominant negative expressing skeletal muscle fibres. J. Physiol. 2015;593:3849–3863.
    1. Palmer AE, Jin C, Reed JC, Tsien RY. Bcl-2-mediated alterations in endoplasmic reticulum Ca2+ analyzed with an improved genetically encoded fluorescent sensor. Proc. Natl Acad. Sci. USA. 2004;101:17404–17409.
    1. Schindelin J, et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods. 2012;9:676–682.
    1. Burbulla, L. F. & Kruger, R. The use of primary human fibroblasts for monitoring mitochondrial phenotypes in the field of Parkinson’s disease. J. Vis. Exp.10.3791/4228 (2012).
    1. Picard M, et al. Mechanical ventilation triggers abnormal mitochondrial dynamics and morphology in the diaphragm. J. Appl. Physiol. 2015;118:1161–1171.
    1. Jha P, Wang X, Auwerx J. Analysis of mitochondrial respiratory chain supercomplexes using blue native polyacrylamide gel electrophoresis (BN-PAGE) Curr. Protoc. Mouse Biol. 2016;6:1–14.
    1. Gallart-Ayala H, et al. A global HILIC-MS approach to measure polar human cerebrospinal fluid metabolome: exploring gender-associated variation in a cohort of elderly cognitively healthy subjects. Anal. Chim. Acta. 2018;1037:327–337.
    1. Kulak NA, Pichler G, Paron I, Nagaraj N, Mann M. Minimal, encapsulated proteomic-sample processing applied to copy-number estimation in eukaryotic cells. Nat. Methods. 2014;11:319–324.
    1. Wisniewski JR, Gaugaz FZ. Fast and sensitive total protein and peptide assays for proteomic analysis. Anal. Chem. 2015;87:4110–4116.
    1. Geiser L, Dayon L, Vaezzadeh AR, Hochstrasser DF. Shotgun proteomics: a relative quantitative approach using off-gel electrophoresis and LC-MS/MS. Methods Mol. Biol. 2011;681:459–472.
    1. Cox J, Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 2008;26:1367–1372.
    1. Cox J, Mann M. Quantitative, high-resolution proteomics for data-driven systems biology. Annu. Rev. Biochem. 2011;80:273–299.
    1. Tyanova S, Temu T, Cox J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat. Protoc. 2016;11:2301–2319.
    1. Hochberg Y, Benjamini Y. More powerfµl procedures for multiple significance testing. Stat. Med. 1990;9:811–818.
    1. Cox J, Mann M. 1D and 2D annotation enrichment: a statistical method integrating quantitative proteomics with complementary high-throughput data. BMC Bioinform. 2012;13(Suppl.):S12.
    1. Perez-Riverol Y, et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 2019;47:D442–D450.

Source: PubMed

3
Subscribe