1,068
Views
0
CrossRef citations to date
0
Altmetric
Research Paper

Simultaneous loss of TSC1 and DEPDC5 in skeletal and cardiac muscles produces early-onset myopathy and cardiac dysfunction associated with oxidative damage and SQSTM1/p62 accumulation

ORCID Icon, , , , ORCID Icon, , , ORCID Icon & show all
Pages 2303-2322 | Received 16 Jun 2020, Accepted 06 Dec 2021, Published online: 29 Dec 2021

References

  • Cornu M, Albert V, Hall MN. mTOR in aging, metabolism, and cancer. Curr Opin Genet Dev. 2013;23(1):53–62.
  • Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes Dev. 2004;18:1926–1945.
  • Dunlop EA, Tee AR. mTOR and autophagy: a dynamic relationship governed by nutrients and energy. Semin Cell Dev Biol. 2014;36:121–129.
  • Bentzinger CF, Romanino K, Cloetta D, et al. Skeletal muscle-specific ablation of raptor, but not of rictor, causes metabolic changes and results in muscle dystrophy. Cell Metab. 2008;8:411–424.
  • Zhang Q, Duplany A, Moncollin V, et al. Lack of muscle mTOR kinase activity causes early onset myopathy and compromises whole-body homeostasis. J Cachexia Sarcopenia Muscle. 2019;10:35–53.
  • Castets P, Lin S, Rion N, et al. Sustained activation of mTORC1 in skeletal muscle inhibits constitutive and starvation-induced autophagy and causes a severe, late-onset myopathy. Cell Metab. 2013;17:731–744.
  • Baraldo M, Geremia A, Pirazzini M, et al. Skeletal muscle mTORC1 regulates neuromuscular junction stability. J Cachexia Sarcopenia Muscle. 2020;11:208–225.
  • Bodine SC, Stitt TN, Gonzalez M, et al. Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol. 2001;3:1014–1019.
  • Pallafacchina G, Calabria E, Serrano AL, et al. A protein kinase B-dependent and rapamycin-sensitive pathway controls skeletal muscle growth but not fiber type specification. Proc Natl Acad Sci U S A. 2002;99:9213–9218.
  • Goodman CA, Frey JW, Mabrey DM, et al. The role of skeletal muscle mTOR in the regulation of mechanical load-induced growth. J Physiol. 2011;589(22):5485–5501. DOI:10.1113/jphysiol.2011.218255.
  • Kim J, Guan KL. mTOR as a central hub of nutrient signalling and cell growth. Nat Cell Biol. 2019;21:63–71.
  • Huang J, Manning BD. The TSC1-TSC2 complex: a molecular switchboard controlling cell growth. Biochem J. 2008;412:179–190.
  • Dokudovskaya S, Rout MP. SEA you later alli-GATOR–a dynamic regulator of the TORC1 stress response pathway. J Cell Sci. 2015;128:2219–2228.
  • Tang H, Inoki K, Brooks SV, et al. mTORC1 underlies age-related muscle fiber damage and loss by inducing oxidative stress and catabolism. Aging Cell. 2019;18:e12943.
  • Dutchak PA, Estill-Terpack SJ, Plec AA, et al. Loss of a negative regulator of mTORC1 induces aerobic glycolysis and altered fiber composition in skeletal muscle. Cell Rep. 2018;23:1907–1914.
  • Graber TG, Fry CS, Brightwell CR, et al. Skeletal muscle-specific knockout of DEP domain containing 5 protein increases mTORC1 signaling, muscle cell hypertrophy, and mitochondrial respiration. J Biol Chem. 2019;294:4091–4102.
  • Bruning JC, Michael MD, Winnay JN, et al. A muscle-specific insulin receptor knockout exhibits features of the metabolic syndrome of NIDDM without altering glucose tolerance. Mol Cell. 1998;2:559–569.
  • Soliman GA, Acosta-Jaquez HA, Dunlop EA, et al. mTOR Ser-2481 autophosphorylation monitors mTORC-specific catalytic activity and clarifies rapamycin mechanism of action. J Biol Chem. 2010;285:7866–7879.
  • Bodine SC, Latres E, Baumhueter S, et al. Identification of ubiquitin ligases required for skeletal muscle atrophy. Science. 2001;294:1704–1708.
  • Lee JH, Budanov AV, Park EJ, et al. Sestrin as a feedback inhibitor of TOR that prevents age-related pathologies. Science. 2010;327:1223–1228.
  • Kim M, Kowalsky AH, Lee JH. Sestrins in physiological stress responses. Annu Rev Physiol. 2021;83:381–403.
  • Asp M, Bergenstrahle J, Lundeberg J. Spatially resolved transcriptomes-next generation tools for tissue exploration. Bioessays. 2020;42:e1900221.
  • Shah OJ, Wang Z, Hunter T. Inappropriate activation of the TSC/Rheb/mTOR/S6K cassette induces IRS1/2 depletion, insulin resistance, and cell survival deficiencies. Curr Biol. 2004;14:1650–1656.
  • Zhao J, Brault JJ, Schild A, et al. FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells. Cell Metab. 2007;6:472–483.
  • Glass DJ. Molecular mechanisms modulating muscle mass. Trends Mol Med. 2003;9:344–350.
  • Glass DJ. Signaling pathways perturbing muscle mass. Curr Opin Clin Nutr Metab Care. 2010;13:225–229.
  • Egan DF, Shackelford DB, Mihaylova MM, et al. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science. 2011;331:456–461.
  • Kim J, Kundu M, Viollet B, et al. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 2011;13:132–141.
  • Buchan JR, Parker R. Eukaryotic stress granules: the ins and outs of translation. Mol Cell. 2009;36:932–941.
  • Wang B, Maxwell BA, Joo JH, et al. ULK1 and ULK2 regulate stress granule disassembly through phosphorylation and activation of VCP/p97. Mol Cell. 2019;74:742–57 e8.
  • Zhao H, Kalivendi S, Zhang H, et al. Superoxide reacts with hydroethidine but forms a fluorescent product that is distinctly different from ethidium: potential implications in intracellular fluorescence detection of superoxide. Free Radic Biol Med. 2003;34:1359–1368.
  • Carnio S, LoVerso F, Baraibar MA, et al. Autophagy impairment in muscle induces neuromuscular junction degeneration and precocious aging. Cell Rep. 2014;8:1509–1521.
  • Thiemermann C. Membrane-permeable radical scavengers (tempol) for shock, ischemia-reperfusion injury, and inflammation. Crit Care Med. 2003;31:S76–84.
  • Muscoli C, Cuzzocrea S, Riley DP, et al. On the selectivity of superoxide dismutase mimetics and its importance in pharmacological studies. Br J Pharmacol. 2003;140:445–460.
  • Khan NA, Nikkanen J, Yatsuga S, et al. mTORC1 regulates mitochondrial integrated stress response and mitochondrial myopathy progression. Cell Metab. 2017;26:419–28 e5.
  • Joseph GA, Wang SX, Jacobs CE, et al. Partial Inhibition of mTORC1 in aged rats counteracts the decline in muscle mass and reverses molecular signaling associated with Sarcopenia. Mol Cell Biol. 2019;39: ;e00141–19.
  • Bielas J, Herbst A, Widjaja K, et al. Long term rapamycin treatment improves mitochondrial DNA quality in aging mice. Exp Gerontol. 2018;106:125–131.
  • Park JM, Jung CH, Seo M, et al. The ULK1 complex mediates MTORC1 signaling to the autophagy initiation machinery via binding and phosphorylating ATG14. Autophagy. 2016;12:547–564.
  • Russell RC, Tian Y, Yuan H, et al. ULK1 induces autophagy by phosphorylating Beclin-1 and activating VPS34 lipid kinase. Nat Cell Biol. 2013;15:741–750.
  • Ho A, Cho CS, Namkoong S, et al. Biochemical basis of sestrin physiological activities. Trends Biochem Sci. 2016;41:621–632.
  • Hardie DG, Ross FA, Hawley SA. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol. 2012;13:251–262.
  • Egan D, Kim J, Shaw RJ, et al. The autophagy initiating kinase ULK1 is regulated via opposing phosphorylation by AMPK and mTOR. Autophagy. 2011;7:643–644.
  • Yu H, Zhou X, Wang Y, et al. Nicotinamide N-methyltransferase inhibits autophagy induced by oxidative stress through suppressing the AMPK pathway in breast cancer cells. Cancer Cell Int. 2020;20:191.
  • Joshi A, Iyengar R, Joo JH, et al. Nuclear ULK1 promotes cell death in response to oxidative stress through PARP1. Cell Death Differ. 2016;23:216–230.
  • Ro SH, Semple IA, Park H, et al. Sestrin2 Promotes Unc-51-like Kinase 1 (ULK1)-mediated phosphorylation of p62/sequestosome-1. FEBS J. 2014;281:3816–3827.
  • Mammucari C, Milan G, Romanello V, et al. FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab. 2007;6:458–471.
  • Milan G, Romanello V, Pescatore F, et al. Regulation of autophagy and the ubiquitin-proteasome system by the FoxO transcriptional network during muscle atrophy. Nat Commun. 2015;6:6670.
  • Tezze C, Romanello V, Desbats MA, et al. Age-associated loss of OPA1 in muscle impacts muscle mass, metabolic homeostasis, systemic inflammation, and epithelial senescence. Cell Metab. 2017;25:1374–89 e6.
  • Segatto M, Fittipaldi R, Pin F, et al. Epigenetic targeting of bromodomain protein BRD4 counteracts cancer cachexia and prolongs survival. Nat Commun. 2017;8:1707.
  • Komatsu M, Kurokawa H, Waguri S, et al. The selective autophagy substrate p62 activates the stress responsive transcription factor Nrf2 through inactivation of Keap1. Nat Cell Biol. 2010;12:213–223.
  • Zid BM, Rogers AN, Katewa SD, et al. 4E-BP extends lifespan upon dietary restriction by enhancing mitochondrial activity in Drosophila. Cell. 2009;139:149–160.
  • Bartolome A, Garcia-Aguilar A, Asahara SI, et al. MTORC1 regulates both general autophagy and mitophagy induction after oxidative phosphorylation uncoupling. Mol Cell Biol. 2017;37:e00441–17.
  • Tsang CK, Chen M, Cheng X, et al. SOD1 phosphorylation by mTORC1 couples nutrient sensing and redox regulation. Mol Cell. 2018;70:502–15 e8.
  • Ferreira LF, Laitano O. Regulation of NADPH oxidases in skeletal muscle. Free Radic Biol Med. 2016;98:18–28.
  • Cho CS, Kowalsky AH, Namkoong S, et al. Concurrent activation of growth factor and nutrient arms of mTORC1 induces oxidative liver injury. Cell Discov. 2019;5:60.
  • Neuman NA, Henske EP. Non-canonical functions of the tuberous sclerosis complex-Rheb signalling axis. EMBO Mol Med. 2011;3:189–200.
  • Kim YC, Park HW, Sciarretta S, et al. Rag GTPases are cardioprotective by regulating lysosomal function. Nat Commun. 2014;5:4241.
  • Lustgarten MS, Jang YC, Liu Y, et al. Conditional knockout of Mn-SOD targeted to type IIB skeletal muscle fibers increases oxidative stress and is sufficient to alter aerobic exercise capacity. Am J Physiol Cell Physiol. 2009;297:C1520–32.
  • Kim M, Sujkowski A, Namkoong S, et al. Sestrins are evolutionarily conserved mediators of exercise benefits. Nat Commun. 2020;11:190.
  • Seligman AM, Karnovsky MJ, Wasserkrug HL, et al. Nondroplet ultrastructural demonstration of cytochrome oxidase activity with a polymerizing osmiophilic reagent, diaminobenzidine (DAB). J Cell Biol. 1968;38:1–14.
  • Loughlin M. Muscle biopsy. A laboratory investigation. Oxford, Boston: Butterworth-Heinemann; 1993.
  • Sheehan D, Hrapchak B. Histotechnology. 2nd ed. Columbus: Batelle Press; 1987.
  • Dobin A, Davis CA, Schlesinger F, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.
  • Trapnell C, Roberts A, Goff L, et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc. 2012;7:562–578.
  • Li R, Huang Y, Semple I, et al. Cardioprotective roles of sestrin 1 and sestrin 2 against doxorubicin cardiotoxicity. Am J Physiol Heart Circ Physiol. 2019;317:H39–H48.
  • Buras ED, Converso-Baran K, Davis CS, et al. Fibro-Adipogenic remodeling of the diaphragm in obesity-associated respiratory dysfunction. Diabetes. 2019;68:45–56.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.