Muscle-specific Drp1 overexpression impairs skeletal muscle growth via translational attenuation

T Touvier, C De Palma, E Rigamonti, A Scagliola, E Incerti, L Mazelin, J-L Thomas, M D'Antonio, L Politi, L Schaeffer, E Clementi, S Brunelli, T Touvier, C De Palma, E Rigamonti, A Scagliola, E Incerti, L Mazelin, J-L Thomas, M D'Antonio, L Politi, L Schaeffer, E Clementi, S Brunelli

Abstract

Mitochondrial fission and fusion are essential processes in the maintenance of the skeletal muscle function. The contribution of these processes to muscle development has not been properly investigated in vivo because of the early lethality of the models generated so far. To define the role of mitochondrial fission in muscle development and repair, we have generated a transgenic mouse line that overexpresses the fission-inducing protein Drp1 specifically in skeletal muscle. These mice displayed a drastic impairment in postnatal muscle growth, with reorganisation of the mitochondrial network and reduction of mtDNA quantity, without the deficiency of mitochondrial bioenergetics. Importantly we found that Drp1 overexpression activates the stress-induced PKR/eIF2α/Fgf21 pathway thus leading to an attenuated protein synthesis and downregulation of the growth hormone pathway. These results reveal for the first time how mitochondrial network dynamics influence muscle growth and shed light on aspects of muscle physiology relevant in human muscle pathologies.

Figures

Figure 1
Figure 1
Mice overexpressing Drp1 (Drp/MC) in muscle have a severe reduction of muscle mass. (a) Transgene construct. (b) Left: Drp1 and Myc proteins were analysed by western blot in EDL and soleus muscles and liver of WT and transgenic animals, right: Drp1 expression in mitochondrial extract was analysed by western blot. Cyclophilin D (CypD) is used as a loading control. (c) Mitofusin 1 (Mfn1), Mitofusin 2 (Mfn2) and Opa1 expression was analysed by western blot. Quantification is given in the right chart (n=4 animals per genotype). (d) Growth curve of WT and Drp/MC animals (n=5 animals per group). (e) Pictures of WT and Drp/MC Tibialis anterior (TA), EDL and soleus muscles. (f) Speed and distance performed by P200 WT and transgenic mice on treadmill tests evaluating power and resistance (n=7 animals per genotype). Error bars represent S.E.M. ***P<0.001 versus wt
Figure 2
Figure 2
Muscle-specific Drp1 overexpression impairs postnatal muscle growth. (a, b, c) Representative images of immunofluorescence on TA sections from P1 (a), P7 (b) and P25 (c) mice by using a laminin-specific antibody. Original magnification x20, scale bar 50um. Frequency histogram showing the distribution of myofibre CSA in WT and Drp/MC TA muscles from P1 (a), P7 (b) and P25 (c) mice. Per condition, at least 500 fibres were counted. Mean myofibre CSA in WT and Drp/MC from P1 (a), P7 (b) and P25 (c) mice (n≥3 animals per genotype). (d) Representative sections of WT and Drp/MC TA muscles from P100 mice stained with H&E. Representative images of immunofluorescence on TA sections by using a laminin-specific antibody. Frequency histogram showing the distribution of myofibre CSA in WT and Drp/MC TA muscles from P100 mice. Mean myofibre CSA in WT and Drp/MC (n=3 animals per genotype). (e) Mean number of myofibres muscle (n=3 animals per genotype). (f) Mean number of myonuclei and Pax7 positive cells in WT and Drp/MC single fibres (n=4 preparations per genotype). Original magnification x20, scale bar: 50 μm. Error bars represent S.E.M. **P<0.01; *P<0.05 versus WT
Figure 3
Figure 3
Muscle-specific Drp1 overexpression induces a drastic remodelling of mitochondrial network. (a) SDH staining of transversal sections from WT and Drp/MC from P1, P7, P25 TA muscles. Original magnification × 20, scale bar: 50 μm. (b) SDH staining of transversal and longitudinal sections from WT and Drp/MC P100 TA muscle. Low magnification, × 4; high magnification, × 40. (c) Representative confocal images of mitochondrial network in PhAM and Drp/MC/PhAM TA myofibres from P100 animals. Transversal and longitudinal optical sections are also shown. Original magnification x40, scale bar: 10 μm. (d) Western blot analysis of Tim23 and Cyclophilin D (CypD) protein expression levels in mitochondrial protein lysates from quadriceps of adult WT and Drp/MC mice. Gapdh was used as internal control. (e) Electron microscopy analysis of TA muscles from P100 Drp/MC and WT mice. Arrows point to swollen mithocondria, with none or fewer than normal cristae. (f) Western blot analysis of Opa1 GTPase activity after pull-down experiments using GTP-conjugated beads
Figure 4
Figure 4
Mitochondrial bioenergetics remained unchanged but mtDNA content is reduced and a mtUPR response is activated in Drp/MC muscle. (a) Fibres were loaded with the mitochondrial potentiometric dye TMRM (25 nM) and the fluorescence was recorded for 40 min. The arrows indicate the time of addition of oligomycin (Oligo; 5 μM) and FCCP (4 μM). (b) Mitochondrial respiration in permeabilised TA myofibres was measured by using high-sensitivity respirometer (n=8 animals per genotype); complex I (CI), complex II (CII) and complex IV (CIV). (c) Isolated mitochondria from TA muscle were incubated with luciferin-luciferase and the ATP generated through oxidative phosphorylation was measured (n=9 animals per genotype). (d) Mitochondrial DNA (mtDNA) content in TA muscle from P7 and P100 animals (n=8 per genotype). (e) RT-qPCR analysis of mtDNA and nuclear DNA encoded respiratory chain subunits in P100 quadriceps muscle. (f) RT-qPCR analysis of Chop mRNA levels in quadriceps from P7, P25 and P100 WT and Drp/MC mice (n=8 per genotype). (g) Western blot analysis of Hsp60 and ClpP protein expression levels in quadriceps from adult WT and Drp/MC mice (n=8 per genotype). Actin was used as control. (h) RT-qPCR analysis of Atpif1 mRNA levels in quadriceps from P100 WT and Drp/MC mice (n=8 per genotype). (i) Western blot analysis of ATPIF1 protein expression levels in quadriceps from adult WT and Drp/MC mice (n=8 per genotype). Tim23 and Complex II (CII) were used as control. Error bars represent S.E.M. *P<0.05, **P<0.01, ***P<0.001 versus WT; ##P<0.01, ###P<0.001 versus P7 mice
Figure 5
Figure 5
eIF2α/Atf4 pathway is activated in Drp/MC muscle. (a) Polysome profile in quadriceps from young (P40) WT and Drp/MC mice. Lysates were analysed by centrifugation in a 10–50% sucrose gradient, and the profiles were measured by absorbance at 254 nm. The panel is representative of three independent experiments. (b) Representative western blot analysis of phosphorylated and total eIF2α protein expression levels in quadriceps from P7, P25 and adult (P100) WT and Drp/MC mice. Quantification is given in the right chart (n=5 per genotype). (c) Western blot analysis of phosphorylated PKR protein levels after immunoprecipitation of total PKR protein from gastrocnemius protein lysate of adult (P100) WT and Drp/MC mice (n=3 per genotype). (d) Atf4 mRNA distribution in the fractions collected from the sucrose gradients was determined by RT-qPCR. Percentage of Atf4 transcripts present in polysomal fractions (6–12) (n=3 per genotype). (e) RT-qPCR analysis of representative stress-response genes mRNA levels in quadriceps from adult WT and Drp/MC mice (n=8 per genotype). (f) Plasma levels of Fgf21 in adult WT and Drp/MC mice (n=11 per genotype). (g) RT-qPCR analysis of Cish mRNA levels in quadriceps from adult WT and Drp/MC mice 4 h after GH treatment (1.5 mg/kg i.p., n=7 per group). Error bars represent S.E.M. *P<0.05, **P<0.01, ***P<0.001 versus WT

References

    1. 1Pham AH, McCaffery JM, Chan DC. Mouse lines with photo-activatable mitochondria to study mitochondrial dynamics. Genesis 2012; 50: 833–843.
    1. 2Wagatsuma A, Sakuma K. Mitochondria as a potential regulator of myogenesis. Sci World J 2013; 2013: 1–9.
    1. 3Chen H, Vermulst M, Wang YE, Chomyn A, Prolla TA, McCaffery JM et al. Mitochondrial fusion is required for mtDNA stability in skeletal muscle and tolerance of mtDNA mutations. Cell 2010; 141: 280–289.
    1. 4Wagatsuma A, Kotake N, Yamada S. Muscle regeneration occurs to coincide with mitochondrial biogenesis. Mol Cell Biochem 2011; 349: 139–147.
    1. 5Bach D, Pich S, Soriano FX, Vega N, Baumgartner B, Oriola J et al. Mitofusin-2 determines mitochondrial network architecture and mitochondrial metabolism. A novel regulatory mechanism altered in obesity. J Biol Chem 2003; 278: 17190–17197.
    1. 6De Palma C, Falcone S, Pisoni S, Cipolat S, Panzeri C, Pambianco S et al. Nitric oxide inhibition of Drp1-mediated mitochondrial fission is critical for myogenic differentiation. Cell Death Differ 2010; 17: 1684–1696.
    1. 7Chan DC. Fusion and fission: interlinked processes critical for mitochondrial health. Annu Rev Genet 2012; 46: 265–287.
    1. 8Otera H, Ishihara N, Mihara K. New insights into the function and regulation of mitochondrial fission. Biochim Biophys Acta 2013; 1833: 1256–1268.
    1. 9Delettre C, Lenaers G, Griffoin JM, Gigarel N, Lorenzo C, Belenguer P et al. Nuclear gene OPA1, encoding a mitochondrial dynamin-related protein, is mutated in dominant optic atrophy. Nat Genet 2000; 26: 207–210.
    1. 10Waterham HR, Koster J, van Roermund CW, Mooyer PA, Wanders RJ, Leonard JV et al. A lethal defect of mitochondrial and peroxisomal fission. N Engl J Med 2007; 356: 1736–1741.
    1. 11Züchner S, Mersiyanova IV, Muglia M, Bissar-Tadmouri N, Rochelle J, Dadali EL et al. Mutations in the mitochondrial GTPase mitofusin 2 cause Charcot-Marie-Tooth neuropathy type 2 A. Nat Genet 2004; 36: 449–451.
    1. 12Lokireddy S, Wijesoma IW, Bonala S, Wei M, Sze SK, McFarlane C et al. Myostatin is a novel tumoral factor that induces cancer cachexia. Biochem J 2012; 446: 23–36.
    1. 13Jheng HF, Tsai PJ, Guo SM, Kuo LH, Chang CS, Su IJ et al. Mitochondrial fission contributes to mitochondrial dysfunction and insulin resistance in skeletal muscle. Mol Cell Biol 2011; 32: 309–319.
    1. 14Liu R, Jin P, Liqun Yu, Wang Y, Han L, Shi T et al. Impaired mitochondrial dynamics and bioenergetics in diabetic skeletal muscle. PLoS One 2014; 9: e92810.
    1. 15Eisner V, Lenaers G, Hajnóczky G. Mitochondrial fusion is frequent in skeletal muscle and supports excitation-contraction coupling. J Cell Biol 2014; 205: 179–195.
    1. 16Romanello V, Guadagnin E, Gomes L, Roder I, Sandri C, Petersen Y et al. Mitochondrial fission and remodelling contributes to muscle atrophy. EMBO J 2010; 29: 1774–1785.
    1. 17Kim B, Kim JS, Yoon Y, Santiago MC, Brown MD, Park JY et al. Inhibition of Drp1-dependent mitochondrial division impairs myogenic differentiation. Am J Physiol 2013; 305: R927–R938.
    1. 18Kanisicak O, Mendez JJ, Yamamoto S, Yamamoto M, Goldhamer DJ. Progenitors of skeletal muscle satellite cells express the muscle determination gene, MyoD. Dev Biol 2009; 332: 131–141.
    1. 19Reeds PJ, Fiorotto ML. Growth in perspective. Proc Nutr Soc 2007; 49: 411–420.
    1. 20Irwin WA, Bergamin N, Sabatelli P, Reggiani C, Megighian A, Merlini L et al. Mitochondrial dysfunction and apoptosis in myopathic mice with collagen VI deficiency. Nat Genet 2003; 35: 367–371.
    1. 21Martinus RD, Garth GP, Webster TL, Cartwright P, Naylor DJ, Høj PB et al. Selective induction of mitochondrial chaperones in response to loss of the mitochondrial genome. Eur J Biochem 1996; 240: 98–103.
    1. 22Horibe T, Hoogenraad NJ. The chop gene contains an element for the positive regulation of the mitochondrial unfolded protein response. PLoS One 2007; 2: e835.
    1. 23Chen WW, Birsoy K, Mihaylova MM, Snitkin H, Stasinski I, Yucel B et al. Inhibition of ATPIF1 ameliorates severe mitochondrial respiratory chain dysfunction in mammalian cells. Cell Rep 2014; 7: 27–34.
    1. 24Wek RC, Jiang HY, Anthony TG. Coping with stress: eIF2 kinases and translational control. Biochem Soc Trans 2006; 34: 7–11.
    1. 25Rath E, Berger E, Messlik A, Nunes T, Liu B, Kim SC et al. Induction of dsRNA-activated protein kinase links mitochondrial unfolded protein response to the pathogenesis of intestinal inflammation. Gut 2012; 61: 1269–1278.
    1. 26Palam LR, Baird TD, Wek RC. Phosphorylation of eIF2 facilitates ribosomal bypass of an inhibitory upstream ORF to enhance CHOP translation. J Biol Chem 2011; 286: 10939–10949.
    1. 27Ebert SM, Monteys AM, Fox DK, Bongers KS, Shields BE, Malmberg SE et al. The transcription factor ATF4 promotes skeletal myofiber atrophy during fasting. Mol Endocrinol 2010; 24: 790–799.
    1. 28Suomalainen A, Elo JM, Pietiläinen KH, Hakonen AH, Sevastianova K, Korpela M et al. FGF-21 as a biomarker for muscle-manifesting mitochondrial respiratory chain deficiencies: a diagnostic study. Lancet Neurol 2011; 10: 806–818.
    1. 29Wu S., Grunwald T, Kharitonenkov A, Dam J, Jockers R, De Luca F. et al. Increased expression of fibroblast growth factor 21 (FGF21) during chronic undernutrition causes growth hormone insensitivity in chondrocytes by inducing leptin receptor overlapping transcript (LEPROT) and leptin receptor overlapping transcript-like 1 (LEPROTL1) expression. J Biol Chem 2013; 288: 27375–27383.
    1. 30Ebert SM, Dyle MC, Kunkel SD, Bullard SA, Bongers KS, Fox DK et al. Stress-induced skeletal muscle Gadd45a expression reprograms myonuclei and causes muscle atrophy. J Biol Chem 2012; 287: 27290–27301.
    1. 31Inagaki T, Dyle MC, Kunkel SD, Bullard SA, Bongers KS, Fox DK et al. Inhibition of growth hormone signaling by the fasting-induced hormone FGF21. Cell Metab 2008; 8: 77–83.
    1. 32Touvier T, Conte-Auriol F, Briand O, Cudejko C, Paumelle R, Caron S et al. LEPROT and LEPROTL1 cooperatively decrease hepatic growth hormone action in mice. J Clin Invest 2009; 119: 3830–3838.
    1. 33Ogata T, Yamasaki Y. Ultra-high-resolution scanning electron microscopy of mitochondria and sarcoplasmic reticulum arrangement in human red, white, and intermediate muscle fibers. Anat Rec 1997; 248: 214–223.
    1. 34Kasahara A, Cipolat S, Chen Y, Dorn GW, Scorrano L. Mitochondrial fusion directs cardiomyocyte differentiation via calcineurin and Notch signaling. Science 2013; 342: 734–737.
    1. 35Frieden M, James D, Castelbou C, Danckaert A, Martinou JC, Demaurex N et al. Ca2+ Homeostasis during mitochondrial fragmentation and perinuclear clustering induced by hFis1. J Biol Chem 2004; 279: 22704–22714.
    1. 36Al-Mehdi AB, Pastukh VM, Swiger BM, Reed DJ, Patel MR et al. Perinuclear mitochondrial clustering creates an oxidant-rich nuclear domain required for hypoxia-induced transcription. Sci Signal 2012; 5: ra47.
    1. 37Elachouri G, Vidoni S, Zanna C, Pattyn A, Boukhaddaoui H, Gaget K et al. OPA1 links human mitochondrial genome maintenance to mtDNA replication and distribution. Genome Res 2011; 21: 12–20.
    1. 38Alter J, Bengal E. Stress-induced C/EBP homology protein (CHOP) represses MyoD transcription to delay myoblast differentiation. PLoS One 2011; 6: e29498.
    1. 39Muñoz JP, Zorzano A. Mfn2 modulates the unfolded protein response. Cell Cycle 2014; 13: 0–1.
    1. 40Ngoh GA, Papanicolaou KN, Walsh K. Loss of Mitofusin 2 promotes endoplasmic reticulum stress. J Biol Chem 2012; 287: 20321–20332.
    1. 41Eley HL, Russell ST, Tisdale MJ. Attenuation of muscle atrophy in a murine model of cachexia by inhibition of the dsRNA-dependent protein kinase. Br J Cancer 2007; 96: 1216–1222.
    1. 42Eley HL, Tisdale MJ. Skeletal muscle atrophy, a link between depression of protein synthesis and increase in degradation. J Biol Chem 2007; 282: 7087–7097.
    1. 43Russell ST, Rajani S, Dhadda RS, Tisdale MJ. Mechanism of induction of muscle protein loss by hyperglycaemia. Exp Cell Res 2009; 315: 16–25.
    1. 44Tyynismaa H, Carroll CJ, Raimundo N, Ahola-Erkkilä S, Wenz T, Ruhanen H et al. Mitochondrial myopathy induces a starvation-like response. Hum Mol Genet 2010; 19: 3948–3958.
    1. 45Kopchick JJ, Laron Z. Is the Laron mouse an accurate model of Laron syndrome? Mol Genet Metab 1999; 68: 232–236.
    1. 46Badaloni A, Bonanomi D, Albieri I, Givogri I, Bongarzone E, Valtorta F et al. Transgenic mice expressing a dual, CRE-inducible reporter for the analysis of axon guidance and synaptogenesis. Genesis 2007; 45: 405–412.
    1. 47Rigamonti E, Touvier T, Clementi E, Manfredi AA, Brunelli S, Rovere-Querini P et al. Requirement of inducible nitric oxide synthase for skeletal muscle regeneration after acute damage. J Immunol 2013; 190: 1767–1777.
    1. 48D'Antonio M, Musner N, Scapin C, Ungaro D, Del Carro U, Ron D et al. Resetting translational homeostasis restores myelination in Charcot-Marie-Tooth disease type 1B mice. J Exp Med 2013; 210: 821–838.
    1. 49Frezza C, Cipolat S, Scorrano L. Organelle isolation: functional mitochondria from mouse liver, muscle and cultured fibroblasts. Nat Prot 2007; 2: 287–295.
    1. 50Taillandier D, Aurousseau E, Combaret L, Guezennec CY, Attaix D. Regulation of proteolysis during reloading of the unweighted soleus muscle. Int J Biochem Cell Biol 2003; 35: 665–675.

Source: PubMed

3
Iratkozz fel