Deep muscle-proteomic analysis of freeze-dried human muscle biopsies reveals fiber type-specific adaptations to exercise training

A S Deshmukh, D E Steenberg, M Hostrup, J B Birk, J K Larsen, A Santos, R Kjøbsted, J R Hingst, C C Schéele, M Murgia, B Kiens, E A Richter, M Mann, J F P Wojtaszewski, A S Deshmukh, D E Steenberg, M Hostrup, J B Birk, J K Larsen, A Santos, R Kjøbsted, J R Hingst, C C Schéele, M Murgia, B Kiens, E A Richter, M Mann, J F P Wojtaszewski

Abstract

Skeletal muscle conveys several of the health-promoting effects of exercise; yet the underlying mechanisms are not fully elucidated. Studying skeletal muscle is challenging due to its different fiber types and the presence of non-muscle cells. This can be circumvented by isolation of single muscle fibers. Here, we develop a workflow enabling proteomics analysis of pools of isolated muscle fibers from freeze-dried human muscle biopsies. We identify more than 4000 proteins in slow- and fast-twitch muscle fibers. Exercise training alters expression of 237 and 172 proteins in slow- and fast-twitch muscle fibers, respectively. Interestingly, expression levels of secreted proteins and proteins involved in transcription, mitochondrial metabolism, Ca2+ signaling, and fat and glucose metabolism adapts to training in a fiber type-specific manner. Our data provide a resource to elucidate molecular mechanisms underlying muscle function and health, and our workflow allows fiber type-specific proteomic analyses of snap-frozen non-embedded human muscle biopsies.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Workflow for isolation of muscle…
Fig. 1. Workflow for isolation of muscle fibers and proteomic analysis.
a Single fibers were dissected from freeze-dried muscle biopsies before and after exercise training and fibers were dissolved in lysis buffer. b Fiber-type determination by dot-blotting. Top left: loading scheme of two identical membranes incubated in either MHC I (top right) or MHC II antibody (bottom left). m =  missing fibers, not recognized by MHC I or II antibody; c = contaminated fibers (or hybrid fibers) were discarded. Fibers were pooled according to fiber type and the pools were validated by dot-blotting (bottom right). c Peak rate of oxygen consumption (VO2peak) and insulin-stimulated leg glucose uptake (mean last 40 min of a 120 min insulin clamp) before (PRE) and after (POST) training. LLM lean leg mass. Glycogen content in whole-muscle homogenate (total) (n = 5) and in slow- and fast-twitch fiber pools (n = 4). AU arbitrary units, d.w. dry weight. d Proteomic analysis of purified muscle fibers and primary human muscle cells. Protein digestion by LysC and trypsin followed by optimized LC-MS/MS and computational analysis using MaxQuant with the “match between runs” feature. e Number of proteins and unique peptides identified in muscle fibers and cells and Venn diagram displaying number of common and unique peptides to LysC and trypsin fraction. f Unique peptide-based abundance of MHC isoforms in fiber pools PRE training (n = 5). g Unique peptide-based abundance of AMPK subunits (n = 5). Data in Fig. 1c are shown as means. P values by two-tailed paired t-test are indicated in Fig. 1c.
Fig. 2. Quantitative proteome differences between slow-…
Fig. 2. Quantitative proteome differences between slow- and fast-twitch muscle fibers.
a Principal component analysis (PCA) separating slow- and fast-twitch fibers in two groups. b PCA loading displaying selected examples of the most abundant proteins. c Volcano plot comparing protein abundance in slow- and fast-twitch muscle fibers. Differently regulated proteins (significance score ≤0.05; paired linear model with Xiao correction) are marked in blue (high in fast-twitch fiber) and red (high in slow-twitch fiber). d Hierarchical clustering of significantly different proteins (left) and Fischer exact test for enrichment analysis of significantly different proteins (right). e Graphical representation of a myofibril. f Comparative protein abundance of Terminal cisternae, sarcoplasmic reticulum (SR), T-tubule, and Ca2+ channel complex in slow- and fast-twitch fibers. g Major organelle protein composition in slow- and fast-twitch muscle fibers. Data in f, g are shown as median. P values by two-tailed paired t-test are indicated in f, g.
Fig. 3. Proteomic responses of slow- and…
Fig. 3. Proteomic responses of slow- and fast-twitch muscle fibers to exercise training.
a Volcano plot comparing protein abundance in slow- muscle fibers after exercise training. Differently regulated proteins (significance score ≤0.05; paired linear model with Xiao correction) are marked in red (high after exercise training) and green (low after exercise training). b Volcano plot comparing protein abundance in fast-twitch muscle fibers after exercise training. Differently regulated proteins (significance score ≤0.05; paired linear model with Xiao correction) are marked in blue (high after exercise training) and green (low after exercise training). c MHC composition in whole-muscle lysate PRE and POST training (n = 5). d Overlap of significantly different proteins between slow- and fast-twitch fibers. ClueGO-enriched network of exercise training-regulated proteins in slow- (e) and fast-twitch (f) fibers. The network represents clusters of related Gene Ontology terms enriched as nodes and connected if they annotate common proteins. For visualization purposes, clusters are labeled with the most informative term; node size represents the term enrichment significance and nodes.
Fig. 4. Slow- and fast-twitch muscle fibers…
Fig. 4. Slow- and fast-twitch muscle fibers adapt differentially to exercise training.
a Diagram displaying proteins that are commonly and exclusively expressed in slow- and fast-twitch muscle fibers + the number of exercise training-regulated proteins in the exclusive population. b Volcano plot comparing fold change (POST vs. PRE) between slow- and fast-twitch muscle fibers. Differently regulated proteins (significance score ≤0.05; paired linear model with Xiao correction) are marked in blue (change in fast > slow-twitch fibers) and red (change in slow > fast-twitch fibers). c Scatter plot comparing differences in the log2-changes (POST vs. PRE) between slow- and fast-twitch fibers. The graph includes 131 fiber-type-specific (interaction) significant proteins with examples of few protein categories. d Overlap of exercise training-regulated proteins between whole-muscle lysate and slow-twitch muscle fibers. e Overlap of exercise training-regulated proteins between whole-muscle lysate and fast-twitch muscle fibers.
Fig. 5. Fiber-type-specific effects of exercise training…
Fig. 5. Fiber-type-specific effects of exercise training on capacity to increase mitochondrial content.
a Schematic representation of the steps involved in increasing mitochondrial content. b Percentage protein abundance of TFAM, c proteins involved in mitochondrial protein translation, d mitochondrial DNA-encoded proteins, e proteins involved in cytoplasmic protein translation, f transit peptide-associated proteins, g TIM TOM subunits. Data are shown as median (n = 5). P values by two-way RM ANOVA followed by Tukey’s post hoc test are indicated in Fig. 5b–g. “*” denotes a training effect and “†” denotes a difference between fiber types. Lines indicate main effects.
Fig. 6. Fiber-type-specific regulation of glucose metabolism…
Fig. 6. Fiber-type-specific regulation of glucose metabolism in response to exercise training.
a Schematic representation of glucose metabolism in skeletal muscle. b Percentage protein abundance of glycolytic enzymes, c glucose-6-phosphate dehydrogenase, d enzymes involved in glycogen metabolism, e lactic acid metabolism, f glycerol-3P shuttle, g malate-aspartate shuttle, h pyruvate dehydrogenase complex, i TCA cycle, and j OXPHOS (n = 5). P values by two-way RM ANOVA followed by Tukey’s post hoc test are indicated in Fig. 6b–j. “*” denotes a training effect and “†” denotes a difference between fiber types. Lines indicate main effects. 5× and 10× = multiplied by 5 and 10, respectively.

References

    1. Saltin B, et al. Physical training and glucose tolerance in middle-aged men with chemical diabetes. Diabetes. 1979;28:30–32.
    1. Dela F, et al. Insulin-stimulated muscle glucose clearance in patients with NIDDM: effects of one-legged physical training. Diabetes. 1995;44:1010–1020.
    1. Deshmukh AS, et al. Deep proteomics of mouse skeletal muscle enables quantitation of protein isoforms, metabolic pathways, and transcription factors. Mol. Cell. Proteomics. 2015;14:841–853.
    1. Schiaffino S, Reggiani C. Fiber types in mammalian skeletal muscles. Physiol. Rev. 2011;91:1447–1531.
    1. Zierath JR, Hawley JA. Skeletal muscle fiber type: influence on contractile and metabolic properties. PLoS Biol. 2004;2:1523–1527.
    1. Aebersold R, Mann M. Mass-spectrometric exploration of proteome structure and function. Nature. 2016;537:347–355.
    1. Hoffman NJ, et al. Global phosphoproteomic analysis of human skeletal muscle reveals a network of exercise-regulated kinases and AMPK substrates. Cell Metab. 2015;22:922–935.
    1. Schmalbruch H, Hellhammer U. The number of nuclei in adult rat muscles with special reference to satellite cells. Anat. Rec. 1977;189:169–175.
    1. Murgia M, et al. Single muscle fiber proteomics reveals unexpected mitochondrial specialization. EMBO Rep. 2015;16:387–395.
    1. Murgia M, et al. Single muscle fiber proteomics reveals fiber-type-specific features of human muscle aging. Cell Rep. 2017;19:2396–2409.
    1. Holloway, K. V. et al. Proteomic investigation of changes in human vastus lateralis muscle in response to interval-exercise training. Proteomics10.1002/pmic.200900068 (2009).
    1. Egan B, et al. 2-D DIGE analysis of the mitochondrial proteome from human skeletal muscle reveals time course-dependent remodelling in response to 14 consecutive days of endurance exercise training. Proteomics. 2011;11:1413–1428.
    1. Hostrup M, Onslev J, Jacobson GA, Wilson R, Bangsbo J. Chronic β2-adrenoceptor agonist treatment alters muscle proteome and functional adaptations induced by high intensity training in young men. J. Physiol. 2018;596:231–252.
    1. Oertzen-Hagemann V, et al. Effects of 12 weeks of hypertrophy resistance exercise training combined with collagen peptide supplementation on the skeletal muscle proteome in recreationally active men. Nutrients. 2019;11:1–18.
    1. Güttsches AK, et al. Proteomics of rimmed vacuoles define new risk allele in inclusion body myositis. Ann. Neurol. 2017;81:227–239.
    1. Kley RA, et al. A combined laser microdissection and mass spectrometry approach reveals new disease relevant proteins accumulating in aggregates of filaminopathy patients. Mol. Cell. Proteomics. 2013;12:215–227.
    1. Winter L, et al. Mutant desmin substantially perturbs mitochondrial morphology, function and maintenance in skeletal muscle tissue. Acta Neuropathol. 2016;132:453–473.
    1. Steenberg DE, et al. Exercise training reduces the insulin-sensitizing effect of a single bout of exercise in human skeletal muscle. J. Physiol. 2019;597:89–103.
    1. Cox J, et al. Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ. Mol. Cell. Proteomics. 2014;13:2513–2526.
    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. Wiśniewski JR, Mann M. Consecutive proteolytic digestion in an enzyme reactor increases depth of proteomic and phosphoproteomic analysis. Anal. Chem. 2012;84:2631–2637.
    1. Scheltema RA, et al. The Q Exactive HF, a benchtop mass spectrometer with a pre-filter, high-performance quadrupole and an ultra-high-field orbitrap analyzer. Mol. Cell. Proteomics. 2014;13:3698–3708.
    1. Swaney DL, Wenger CD, Coon JJ. Value of using multiple proteases for large-scale mass spectrometry-based proteomics. J. Proteome Res. 2010;9:1323–1329.
    1. Guo X, Trudgian DC, Lemoff A, Yadavalli S, Mirzaei H. Confetti: a multiprotease map of the HeLa proteome for comprehensive proteomics. Mol. Cell. Proteomics. 2014;13:1573–1584.
    1. Harrison BC, Allen DL, Leinwand LA. IIb or not IIb? Regulation of myosin heavy chain gene expression in mice and men. Skelet. Muscle. 2011;1:1–9.
    1. Kjøbsted R, et al. AMPK in skeletal muscle function and metabolism. FASEB J. 2018;32:1741–1777.
    1. Kristensen DE, et al. Human muscle fibre type-specific regulation of AMPK and downstream targets by exercise. J. Physiol. 2015;593:2053–2069.
    1. Xiao Y, et al. A novel significance score for gene selection and ranking. Bioinformatics. 2014;30:801–807.
    1. Sun GD, et al. The endoplasmic reticulum stress-inducible protein Niban regulates eIF2α and S6K1/4E-BP1 phosphorylation. Biochem. Biophys. Res. Commun. 2007;360:181–187.
    1. Sharma P, Shathasivam T, Ignatchenko V, Kislinger T, Gramolini AO. Identification of an FHL1 protein complex containing ACTN1, ACTN4, and PDLIM1 using affinity purifications and MS-based protein-protein interaction analysis. Mol. Biosyst. 2011;7:1185–1196.
    1. Domenighetti AA, et al. Loss of FHL1 induces an age-dependent skeletal muscle myopathy associated with myofibrillar and intermyofibrillar disorganization in mice. Hum. Mol. Genet. 2014;23:209–225.
    1. Ciciliot S, Rossi AC, Dyar KA, Blaauw B, Schiaffino S. Muscle type and fiber type specificity in muscle wasting. Int. J. Biochem. Cell Biol. 2013;45:2191–2199.
    1. Baker SK. Molecular clues into the pathogenesis of statin-mediated muscle toxicity. Muscle Nerve. 2005;31:572–580.
    1. Allen DG, Lamb GD, Westerblad H. Skeletal muscle fatigue: cellular mechanisms. Physiol. Rev. 2008;88:287–332.
    1. Lamboley CR, Murphy RM, Mckenna MJ, Lamb GD. Sarcoplasmic reticulum Ca2+ uptake and leak properties, and SERCA isoform expression, in type I and type II fibres of human skeletal muscle. J. Physiol. 2014;592:1381–1395.
    1. Snijders T, et al. Satellite cells in human skeletal muscle plasticity. Front. Physiol. 2015;6:1–21.
    1. Albers PH, et al. Human muscle fiber type specific insulin signaling—impact of obesity and type 2 diabetes. Diabetes. 2014;64:485–497.
    1. Serrano AL, et al. Calcineurin controls nerve activity-dependent specification of slow skeletal muscle fibers but not muscle growth. Proc. Natl Acad. Sci. USA. 2001;98:13108–13113.
    1. Baughman JM, et al. Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporter. Nature. 2011;476:341–345.
    1. Rizzuto R, De Stefani D, Raffaello A, Mammucari C. Mitochondria as sensors and regulators of calcium signalling. Nat. Rev. Mol. Cell Biol. 2012;13:566–578.
    1. Bindea G, et al. ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics. 2009;25:1091–1093.
    1. Egan B, Zierath JR. Exercise metabolism and the molecular regulation of skeletal muscle adaptation. Cell Metab. 2013;17:162–184.
    1. Murphy RM, Lamb GD. Important considerations for protein analyses using antibody based techniques: down-sizing western blotting up-sizes outcomes. J. Physiol. 2013;591:5823–5831.
    1. Bensaad K, et al. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell. 2006;126:107–120.
    1. Pedersen BK, Febbraio MA. Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nat. Rev. Endocrinol. 2012;8:457–465.
    1. Meri S, et al. Human protectin (CD59), an 18,000-20,000 MW complement lysis restricting factor, inhibits C5b-8 catalysed insertion of C9 into lipid bilayers. Immunology. 1990;71:1–9.
    1. Gonçalves FGP, et al. Immunohistological analysis of CD59 and membrane attack complex of complement in muscle in juvenile dermatomyositis. J. Rheumatol. 2002;29:1301–1307.
    1. Perry BD, et al. Dissociation between short-term unloading and resistance training effects on skeletal muscle Na+,K+-ATPase, muscle function, and fatigue in humans. J. Appl. Physiol. 2016;121:1074–1086.
    1. Skovgaard C, et al. Effect of speed endurance training and reduced training volume on running economy and single muscle fiber adaptations in trained runners. Physiol. Rep. 2018;6:e13601.
    1. Wyckelsma VL, et al. Intense interval training in healthy older adults increases skeletal muscle [3H]ouabain-binding site content and elevates Na+,K+-ATPase α2 isoform abundance in Type II fibers. Physiol. Rep. 2017;5:e13219.
    1. Christiansen D, et al. Cycling with blood flow restriction improves performance and muscle K + regulation and alters the effect of anti-oxidant infusion in humans. J. Physiol. 2019;597:2421–2444.
    1. MacInnis MJ, et al. Superior mitochondrial adaptations in human skeletal muscle after interval compared to continuous single-leg cycling matched for total work. J. Physiol. 2017;595:2955–2968.
    1. Lanza IR, Nair KS. Muscle mitochondrial changes with aging and exercise. Am. J. Clin. Nutr. 2009;89:467S–471S.
    1. Christensen PM, et al. Unchanged content of oxidative enzymes in fast-twitch muscle fibers and VO2 kinetics after intensified training in trained cyclists. Physiol. Rep. 2015;3:e12428.
    1. Hood DA, Tryon LD, Carter HN, Kim Y, Chen CCW. Unravelling the mechanisms regulating muscle mitochondrial biogenesis. Biochem. J. 2016;473:2295–2314.
    1. Pfanner N, Meijer M. Mitochondrial biogenesis: the Tom and Tim machine. Curr. Biol. 1997;7:R100–R103.
    1. Bengtsson J, Gustafsson T, Widegren U, Jansson E, Sundberg CJ. Mitochondrial transcription factor A and respiratory complex IV increase in response to exercise training in humans. Pflug. Arch. Eur. J. Physiol. 2001;443:61–66.
    1. Anderson S, et al. Sequence and organization of the human mitochondrial genome. Nature. 1981;290:457–465.
    1. Richter EA, Hargreaves M. Exercise, GLUT4, and skeletal muscle glucose uptake. Physiol. Rev. 2013;93:993–1017.
    1. Kletzien RF, Harris PK, Foellmi LA. Glucose-6-phosphate dehydrogenase: a ‘housekeeping’ enzyme subject to tissue-specific regulation by hormones, nutrients, and oxidant stress. FASEB J. 1994;8:174–181.
    1. Lee-Young RS, et al. Glucose-6-phosphate dehydrogenase contributes to the regulation of glucose uptake in skeletal muscle. Mol. Metab. 2016;5:1083–1091.
    1. Lomako J, Lomako WM, Whelan WJ. Glycogenin: the primer for mammalian and yeast glycogen synthesis. Biochim. Biophys. Acta. 2004;1673:45–55.
    1. Testoni G, et al. Lack of glycogenin causes glycogen accumulation and muscle function impairment. Cell Metab. 2017;26:256–266.e4.
    1. Schantz PG, Sjoberg B, Svedenhag J. Malate-aspartate and alpha-glycerophosphate shuttle enzyme levels in human skeletal muscle: methodological considerations and effect of endurance training. Acta Physiol. Scand. 1986;128:397–407.
    1. LeBlanc PJ, Peters SJ, Tunstall RJ, Cameron-Smith D, Heigenhauser GJF. Effects of aerobic training on pyruvate dehydrogenase and pyruvate dehydrogenase kinase in human skeletal muscle. J. Physiol. 2004;557:559–570.
    1. Henriksen TI, et al. Dysregulation of a novel miR-23b/27b-p53 axis impairs muscle stem cell differentiation of humans with type 2 diabetes. Mol. Metab. 2017;6:770–779.
    1. Ladner CL, Yang J, Turner RJ, Edwards RA. Visible fluorescent detection of proteins in polyacrylamide gels without staining. Anal. Biochem. 2004;326:13–20.
    1. Kohn, T. A. & Myburgh, K. H. Electrophoretic separation of human skeletal muscle myosin heavy chain isoforms: the importance of reducing agents. J. Physiol. Sci. 56, 355–360 (2006).
    1. Rappsilber J, Ishihama Y, Mann M. Stop And Go Extraction tips for matrix-assisted laser desorption/ionization, nanoelectrospray, and LC/MS sample pretreatment in proteomics. Anal. Chem. 2003;75:663–670.
    1. Tyanova S, et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods. 2016;13:731–740.
    1. Shannon P, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13:2498–2504.
    1. Clark MG, et al. Blood flow and muscle metabolism: a focus on insulin action. Am. J. Physiol. Endocrinol. Metab. 2003;284:E241–E258.

Source: PubMed

3
Sottoscrivi