Human skeletal muscle mitochondrial dynamics in relation to oxidative capacity and insulin sensitivity

Alexandre Houzelle, Johanna A Jörgensen, Gert Schaart, Sabine Daemen, Nynke van Polanen, Ciarán E Fealy, Matthijs K C Hesselink, Patrick Schrauwen, Joris Hoeks, Alexandre Houzelle, Johanna A Jörgensen, Gert Schaart, Sabine Daemen, Nynke van Polanen, Ciarán E Fealy, Matthijs K C Hesselink, Patrick Schrauwen, Joris Hoeks

Abstract

Aims/hypothesis: Mitochondria operate in networks, adapting to external stresses and changes in cellular metabolic demand and are subject to various quality control mechanisms. On the basis of these traits, we here hypothesise that the regulation of mitochondrial networks in skeletal muscle is hampered in humans with compromised oxidative capacity and insulin sensitivity.

Methods: In a cross-sectional design, we compared four groups of participants (selected from previous studies) ranging in aerobic capacity and insulin sensitivity, i.e. participants with type 2 diabetes (n = 11), obese participants without diabetes (n = 12), lean individuals (n = 10) and endurance-trained athletes (n = 12); basal, overnight fasted muscle biopsies were newly analysed for the current study and we compared the levels of essential mitochondrial dynamics and quality control regulatory proteins in skeletal muscle tissue.

Results: Type 2 diabetes patients and obese participants were older than lean participants and athletes (58.6 ± 4.0 and 56.7 ± 7.2 vs 21.8 ± 2.5 and 25.1 ± 4.3 years, p < 0.001, respectively) and displayed a higher BMI (32.4 ± 3.7 and 31.0 ± 3.7 vs 22.1 ± 1.8 and 21.0 ± 1.5 kg/m2, p < 0.001, respectively) than lean individuals and endurance-trained athletes. Fission protein 1 (FIS1) and optic atrophy protein 1 (OPA1) protein content was highest in muscle from athletes and lowest in participants with type 2 diabetes and obesity, respectively (FIS1: 1.86 ± 0.79 vs 0.79 ± 0.51 AU, p = 0.002; and OPA1: 1.55 ± 0.64 vs 0.76 ± 0.52 AU, p = 0.014), which coincided with mitochondrial network fragmentation in individuals with type 2 diabetes, as assessed by confocal microscopy in a subset of type 2 diabetes patients vs endurance-trained athletes (n = 6). Furthermore, lean individuals and athletes displayed a mitonuclear protein balance that was different from obese participants and those with type 2 diabetes. Mitonuclear protein balance also associated with heat shock protein 60 (HSP60) protein levels, which were higher in athletes when compared with participants with obesity (p = 0.048) and type 2 diabetes (p = 0.002), indicative for activation of the mitochondrial unfolded protein response. Finally, OPA1, FIS1 and HSP60 correlated positively with aerobic capacity (r = 0.48, p = 0.0001; r = 0.55, p < 0.001 and r = 0.61, p < 0.0001, respectively) and insulin sensitivity (r = 0.40, p = 0.008; r = 0.44, p = 0.003 and r = 0.48, p = 0.001, respectively).

Conclusions/interpretation: Collectively, our data suggest that mitochondrial dynamics and quality control in skeletal muscle are linked to oxidative capacity in humans, which may play a role in the maintenance of muscle insulin sensitivity. CLINICAL TRIAL REGISTRY: numbers NCT00943059, NCT01298375 and NL1888 Graphical abstract.

Keywords: Fission, FIS1; Fusion; HSP60; Insulin sensitivity; Mitochondria; OPA1; Oxidative phosphorylation; Skeletal muscle.

Figures

https://www.ncbi.nlm.nih.gov/pmc/articles/instance/7801361/bin/125_2020_5335_Figa_HTML.jpg
Graphical abstract
Fig. 1
Fig. 1
Type 2 diabetic individuals show lower expression of proteins involved in mitochondrial dynamics. Representative western blots (a) and associated quantifications (bf) of mitochondrial dynamics regulatory proteins (MFN1, MFN2, OPA1, DRP1 and FIS1) in human skeletal muscle biopsies. T2DM, participants with type 2 diabetes (n = 11); O, obese individuals without type 2 diabetes (n=12); L, lean individuals (n=10); A, endurance-trained athletes (n=12). Values are normalised to Revert staining and expressed as mean ± SD. *p<0.05, **p<0.01
Fig. 2
Fig. 2
Type 2 diabetic individuals display a more fragmented skeletal muscle mitochondrial network. Representative confocal images of the mitochondrial network in type I muscle fibres. The cellular membrane was stained in green using laminin as a marker, in red the mitochondrial network is visualised using TOMM20 as a marker. In the overview images (a, b and e, f) scale bar, 20 μm; in the zoomed images (c, d and g, h) scale bar, 5 μm. (ad) Representative overview and zoomed images of two separate individuals with type 2 diabetes. (eh) Representative overview and zoomed images of two individual trained athletes. Note the longitudinal pattern of interconnected mitochondria alongside the myofibrils (yellow arrows) and the cross-striated pattern reflecting mitochondria near the Z-line (blue arrows) in the athletes whereas this pattern is virtually absent in the type 2 diabetic participants and the networks present a more punctate dot-like pattern, reflecting disconnected mitochondria
Fig. 3
Fig. 3
Type 2 diabetic individuals show lower mitochondrial content. Representative blots (a) and associated quantifications (bg) of mitochondrial density markers VDAC1 and OXPHOS complex I (CI-NDUFB8), complex II (CII-SDHB), complex III (CIII-UQCRC2), complex IV (CIV-COX II) and complex V (CV-ATP5A) in human skeletal muscle. T2DM, participants with type 2 diabetes (n=11); O, obese individuals without type 2 diabetes (n=12); L, lean individuals (n=10); A, endurance-trained athletes (n=12). Values are normalised to Revert staining and expressed as mean ± SD. OXPHOS complexes are expressed as fold change, with T2DM set to 1. *p<0.05, **p<0.01, ***p<0.001
Fig. 4
Fig. 4
Mitonuclear protein balance differs in lean individuals and athletes vs obese and type 2 diabetic participants. Representative blots (a) and associated quantifications of (b) the nuclear-encoded complex IV (CIV) subunit COX4I1, (c) the mtDNA-encoded complex IV subunit COX II and (d) mitonuclear protein balance (COX4I1/COX II) in human skeletal muscle. Both COX4I1 and COX II were detected on the same blots. T2DM, participants with type 2 diabetes (n=11); O, obese individuals without type 2 diabetes (n=12); L, lean individuals (n=10); A, endurance-trained athletes (n=12). Values are expressed as mean fold change ± SD, with T2DM set to 1. *p<0.05, **p<0.01, ***p<0.001, †p=0.06
Fig. 5
Fig. 5
Endurance-trained athletes show highest protein levels of the mitochondrial chaperone HSP60. Representative western blot analysis (a) and quantification of (b) mitochondrial chaperone HSP60 and (d, e) mito-/autophagy-related proteins in human skeletal muscle biopsies. (c) Mitonuclear protein balance (COX4I1/COX II) is associated with HSP60 protein levels. T2DM, participants with type 2 diabetes (n=11); O, obese individuals without type 2 diabetes (n=12); L, lean individuals (n=10); A, endurance-trained athletes (n=12). Values are normalised to Revert staining and expressed as mean ± SD. *p<0.05, **p<0.01. Correlations are computed with Pearson’s r
Fig. 6
Fig. 6
Proteins involved in mitochondrial dynamics and quality control positively correlate with metabolic variables in human skeletal muscle. Association between protein expression levels of OPA1, FIS1 and HSP60, normalised to Revert staining, with V˙O2max (ac) and insulin sensitivity (df). Correlations are computed with Pearson’s r

References

    1. Cade WT. The manifold role of the mitochondria in skeletal muscle insulin resistance. Curr Opin Clin Nutr Metab Care. 2018;21:267–272. doi: 10.1097/MCO.0000000000000480.
    1. Di Meo S, Iossa S, Venditti P. Skeletal muscle insulin resistance: role of mitochondria and other ROS sources. J Endocrinol. 2017;233:R15–R42. doi: 10.1530/JOE-16-0598.
    1. Hoeks J, Schrauwen P. Muscle mitochondria and insulin resistance: a human perspective. Trends Endocrinol Metab. 2012;23:444–450. doi: 10.1016/j.tem.2012.05.007.
    1. Hancock CR, Han DH, Chen M, et al. High-fat diets cause insulin resistance despite an increase in muscle mitochondria. Proc Natl Acad Sci U S A. 2008;105:7815–7820. doi: 10.1073/pnas.0802057105.
    1. Holloszy JO. “Deficiency” of mitochondria in muscle does not cause insulin resistance. Diabetes. 2013;62:1036–1040. doi: 10.2337/db12-1107.
    1. Fisher-Wellman KH, Weber TM, Cathey BL, et al. Mitochondrial respiratory capacity and content are normal in young insulin-resistant obese humans. Diabetes. 2014;63:132–141. doi: 10.2337/db13-0940.
    1. Benard G, Bellance N, James D, et al. Mitochondrial bioenergetics and structural network organization. J Cell Sci. 2007;120:838–848. doi: 10.1242/jcs.03381.
    1. Detmer SA, Chan DC. Functions and dysfunctions of mitochondrial dynamics. Nat Rev Mol Cell Biol. 2007;8:870–879. doi: 10.1038/nrm2275.
    1. Liesa M, Shirihai OS. Mitochondrial dynamics in the regulation of nutrient utilization and energy expenditure. Cell Metab. 2013;17:491–506. doi: 10.1016/j.cmet.2013.03.002.
    1. Liesa M, Palacin M, Zorzano A. Mitochondrial dynamics in mammalian health and disease. Physiol Rev. 2009;89:799–845. doi: 10.1152/physrev.00030.2008.
    1. Johri A, Beal MF. Mitochondrial dysfunction in neurodegenerative diseases. J Pharmacol Exp Ther. 2012;342:619–630. doi: 10.1124/jpet.112.192138.
    1. Fealy CE, Mulya A, Axelrod CL, Kirwan JP. Mitochondrial dynamics in skeletal muscle insulin resistance and type 2 diabetes. Transl Res. 2018;202:69–82. doi: 10.1016/j.trsl.2018.07.011.
    1. Fealy CE, Mulya A, Lai N, Kirwan JP. Exercise training decreases activation of the mitochondrial fission protein dynamin-related protein-1 in insulin-resistant human skeletal muscle. J Appl Physiol (1985) 2014;117:239–245. doi: 10.1152/japplphysiol.01064.2013.
    1. Sacks J, Mulya A, Fealy CE, et al. Effect of Roux-en-Y gastric bypass on liver mitochondrial dynamics in a rat model of obesity. Physiol Rep. 2018;6(4):e13600. doi: 10.14814/phy2.13600.
    1. Vincent AE, White K, Davey T, et al. Quantitative 3D Mapping of the Human Skeletal Muscle Mitochondrial Network. Cell Rep. 2019;26:996–1009. doi: 10.1016/j.celrep.2019.01.010.
    1. Dahl R, Larsen S, Dohlmann TL, et al. Three-dimensional reconstruction of the human skeletal muscle mitochondrial network as a tool to assess mitochondrial content and structural organization. Acta Physiol (Oxford) 2015;213:145–155. doi: 10.1111/apha.12289.
    1. Kristensen MD, Petersen SM, Moller KE, et al. Obesity leads to impairments in the morphology and organization of human skeletal muscle lipid droplets and mitochondrial networks, which are resolved with gastric bypass surgery-induced improvements in insulin sensitivity. Acta Physiol (Oxford) 2018;224:e13100. doi: 10.1111/apha.13100.
    1. Jovaisaite V, Mouchiroud L, Auwerx J. The mitochondrial unfolded protein response, a conserved stress response pathway with implications in health and disease. J Exp Biol. 2014;217:137–143. doi: 10.1242/jeb.090738.
    1. Aldridge JE, Horibe T, Hoogenraad NJ. Discovery of genes activated by the mitochondrial unfolded protein response (mtUPR) and cognate promoter elements. PLoS One. 2007;2:e874. doi: 10.1371/journal.pone.0000874.
    1. Ashrafi G, Schwarz TL. The pathways of mitophagy for quality control and clearance of mitochondria. Cell Death Differ. 2013;20:31–42. doi: 10.1038/cdd.2012.81.
    1. Kleinridders A, Lauritzen HP, Ussar S, et al. Leptin regulation of Hsp60 impacts hypothalamic insulin signaling. J Clin Invest. 2013;123:4667–4680. doi: 10.1172/JCI67615.
    1. Yi HS, Chang JY, Shong M. The mitochondrial unfolded protein response and mitohormesis: a perspective on metabolic diseases. J Mol Endocrinol. 2018;61:R91–R105. doi: 10.1530/JME-18-0005.
    1. Meex RC, Schrauwen-Hinderling VB, Moonen-Kornips E, et al. Restoration of muscle mitochondrial function and metabolic flexibility in type 2 diabetes by exercise training is paralleled by increased myocellular fat storage and improved insulin sensitivity. Diabetes. 2010;59:572–579. doi: 10.2337/db09-1322.
    1. Phielix E, Meex R, Ouwens DM, et al. High oxidative capacity due to chronic exercise training attenuates lipid-induced insulin resistance. Diabetes. 2012;61:2472–2478. doi: 10.2337/db11-1832.
    1. van de Weijer T, Phielix E, Bilet L, et al. Evidence for a direct effect of the NAD+ precursor acipimox on muscle mitochondrial function in humans. Diabetes. 2015;64:1193–1201. doi: 10.2337/db14-0667.
    1. Vosselman MJ, Hoeks J, Brans B, et al. Low brown adipose tissue activity in endurance-trained compared with lean sedentary men. Int J Obes. 2015;39:1696–1702. doi: 10.1038/ijo.2015.130.
    1. Siri WE (1993) Body composition from fluid spaces and density: analysis of methods. 1961. Nutrition 9:480–491; discussion 480, 492
    1. Hawley JA, Noakes TD. Peak power output predicts maximal oxygen uptake and performance time in trained cyclists. Eur J Appl Physiol Occup Physiol. 1992;65:79–83. doi: 10.1007/BF01466278.
    1. DeFronzo RA, Tobin JD, Andres R. Glucose clamp technique: a method for quantifying insulin secretion and resistance. Am J Phys. 1979;237:E214–E223.
    1. Phielix E, Meex R, Moonen-Kornips E, Hesselink MK, Schrauwen P. Exercise training increases mitochondrial content and ex vivo mitochondrial function similarly in patients with type 2 diabetes and in control individuals. Diabetologia. 2010;53:1714–1721. doi: 10.1007/s00125-010-1764-2.
    1. Daemen S, Gemmink A, Brouwers B, et al. Distinct lipid droplet characteristics and distribution unmask the apparent contradiction of the athlete’s paradox. Mol Metab. 2018;17:71–81. doi: 10.1016/j.molmet.2018.08.004.
    1. Houtkooper RH, Mouchiroud L, Ryu D, et al. Mitonuclear protein imbalance as a conserved longevity mechanism. Nature. 2013;497:451–457. doi: 10.1038/nature12188.
    1. Mouchiroud L, Houtkooper RH, Moullan N, et al. The NAD(+)/Sirtuin Pathway Modulates Longevity through Activation of Mitochondrial UPR and FOXO Signaling. Cell. 2013;154:430–441. doi: 10.1016/j.cell.2013.06.016.
    1. Jheng HF, Tsai PJ, Guo SM, et al. Mitochondrial fission contributes to mitochondrial dysfunction and insulin resistance in skeletal muscle. Mol Cell Biol. 2012;32:309–319. doi: 10.1128/MCB.05603-11.
    1. Li J, Kanasaki M, Xu L, et al. A ketogenic amino acid rich diet benefits mitochondrial homeostasis by altering the AKT/4EBP1 and autophagy signaling pathways in the gastrocnemius and soleus. Biochim Biophys Acta Gen Subj. 2018;1862:1547–1555. doi: 10.1016/j.bbagen.2018.03.013.
    1. Joseph AM, Joanisse DR, Baillot RG, Hood DA. Mitochondrial dysregulation in the pathogenesis of diabetes: potential for mitochondrial biogenesis-mediated interventions. Exp Diabetes Res. 2012;2012:642038. doi: 10.1155/2012/642038.
    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 (Oxford) 2019;225:e13216. doi: 10.1111/apha.13216.
    1. Yu R, Jin SB, Lendahl U, Nister M, Zhao J (2019) Human Fis1 regulates mitochondrial dynamics through inhibition of the fusion machinery. EMBO J 38. 10.1002/ped4.12164
    1. Pei S, Minhajuddin M, Adane B et al (2018) AMPK/FIS1-Mediated Mitophagy Is Required for Self-Renewal of Human AML Stem Cells. Cell Stem Cell 23:86–100.e106. 10.1016/j.stem.2018.05.021
    1. Xian H, Yang Q, Xiao L, Shen HM, Liou YC. STX17 dynamically regulated by Fis1 induces mitophagy via hierarchical macroautophagic mechanism. Nat Commun. 2019;10:2059. doi: 10.1038/s41467-019-10096-1.
    1. Liu R, Jin P, Yu L, et al. Impaired mitochondrial dynamics and bioenergetics in diabetic skeletal muscle. PLoS One. 2014;9:e92810. doi: 10.1371/journal.pone.0092810.
    1. Bach D, Naon D, Pich S, et al. Expression of Mfn2, the Charcot-Marie-tooth neuropathy type 2A gene, in human skeletal muscle: effects of type 2 diabetes, obesity, weight loss, and the regulatory role of tumor necrosis factor alpha and interleukin-6. Diabetes. 2005;54:2685–2693. doi: 10.2337/diabetes.54.9.2685.
    1. Mishra P, Varuzhanyan G, Pham AH, Chan DC. Mitochondrial Dynamics is a Distinguishing Feature of Skeletal Muscle Fiber Types and Regulates Organellar Compartmentalization. Cell Metab. 2015;22:1033–1044. doi: 10.1016/j.cmet.2015.09.027.
    1. Kelley DE, He J, Menshikova EV, Ritov VB. Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes. 2002;51:2944–2950. doi: 10.2337/diabetes.51.10.2944.
    1. Barone R, Macaluso F, Sangiorgi C, et al. Skeletal muscle Heat shock protein 60 increases after endurance training and induces peroxisome proliferator-activated receptor gamma coactivator 1 alpha1 expression. Sci Rep. 2016;6:19781. doi: 10.1038/srep19781.
    1. Arribat Y, Broskey NT, Greggio C et al (2018) Distinct patterns of skeletal muscle mitochondria fusion, fission and mitophagy upon duration of exercise training. Acta Physiol (Oxf) e13179

Source: PubMed

3
Abonneren