Reactive oxygen species in skeletal muscle signaling

Elena Barbieri, Piero Sestili, Elena Barbieri, Piero Sestili

Abstract

Generation of reactive oxygen species (ROS) is a ubiquitous phenomenon in eukaryotic cells' life. Up to the 1990s of the past century, ROS have been solely considered as toxic species resulting in oxidative stress, pathogenesis and aging. However, there is now clear evidence that ROS are not merely toxic species but also-within certain concentrations-useful signaling molecules regulating physiological processes. During intense skeletal muscle contractile activity myotubes' mitochondria generate high ROS flows: this renders skeletal muscle a tissue where ROS hold a particular relevance. According to their hormetic nature, in muscles ROS may trigger different signaling pathways leading to diverging responses, from adaptation to cell death. Whether a "positive" or "negative" response will prevail depends on many variables such as, among others, the site of ROS production, the persistence of ROS flow or target cells' antioxidant status. In this light, a specific threshold of physiological ROS concentrations above which ROS exert negative, toxic effects is hard to determine, and the concept of "physiologically compatible" levels of ROS would better fit with such a dynamic scenario. In this review these concepts will be discussed along with the most relevant signaling pathways triggered and/or affected by ROS in skeletal muscle.

Figures

Figure 1
Figure 1
Major signaling pathways triggered and/or affected by ROS in skeletal muscle. Low levels of ROS activate specific key signaling molecules such as PGC-1α, AMPK, and MAPK, which control cellular mechanisms for muscle adaptation (oxidative metabolism, mitochondrial biogenesis, and mitochondrial functionality) as well as antioxidant enzymes that function as backregulators of intracellular ROS levels. Slight ROS accumulation also inhibits PPases and promotes the phosphorylation state of many proteins involved in the muscle signaling responses. Moreover, low levels of ROS play an important role in inducing upregulation of growth factors such as IGF-1, which has beneficial effects in muscle protein balance, supports oxidative metabolism, and contributes to the development of an oxidant-resistant phenotype, therefore preventing oxidative damage and chronic diseases. Thus, low levels of ROS elicit positive effects on physiological muscle responses. By contrast high levels of ROS cause functional oxidative damages of proteins, lipids, nucleic acids and cell components, induce a significant rise of intracellular [Ca2+], and promote signaling cascades for apoptosis or autophagy via NF-κB or FoxO paths. For these reasons high ROS levels are reputed to act as etiological, or at least exacerbating factors in muscle atrophy, sarcopenia, wasting, and chronic-/aging-related muscle diseases and myopathies. Depending on their level/persistence, ROS may also turn the same process from “physiologic” into “pathologic”, as in the case of inflammation.

References

    1. Murphy ME, Kehrer JP. Activities of antioxidant enzymes in muscle, liver and lung of chickens with inherited muscular dystrophy. Biochemical and Biophysical Research Communications. 1986;134(2):550–556.
    1. Tidball JG. Inflammatory processes in muscle injury and repair. American Journal of Physiology—Regulatory Integrative and Comparative Physiology. 2005;288(2):R345–R353.
    1. Adhihetty PJ, Irrcher I, Joseph AM, Ljubicic V, Hood DA. Plasticity of skeletal muscle mitochondria in response to contractile activity. Experimental Physiology. 2003;88(1):99–107.
    1. Rochard P, Rodier A, Casas F, et al. Mitochondrial activity is involved in the regulation of myoblast differentiation through myogenin expression and activity of myogenic factors. Journal of Biological Chemistry. 2000;275(4):2733–2744.
    1. Sestili P, Barbieri E, Martinelli C, et al. Creatine supplementation prevents the inhibition of myogenic differentiation in oxidatively injured C2C12 murine myoblasts. Molecular Nutrition and Food Research. 2009;53(9):1187–1204.
    1. Davies KJA, Quintanilha AT, Brooks GA, Packer L. Free radicals and tissue damage produced by exercise. Biochemical and Biophysical Research Communications. 1982;107(4):1198–1205.
    1. Koren A, Sauber C, Sentjurc M, Schara M. Free radicals in tetanic activity of isolated skeletal muscle. Comparative Biochemistry and Physiology—B Biochemistry and Molecular Biology. 1983;74(3):633–635.
    1. Jackson MJ, Pye D, Palomero J. The production of reactive oxygen and nitrogen species by skeletal muscle. Journal of Applied Physiology. 2007;102(4):1664–1670.
    1. Langen RCJ, Schols AMWJ, Kelders MCJM, van der Velden JLJ, Wouters EFM, Janssen-Heininger YMW. Tumor necrosis factor-α inhibits myogenesis through redox-dependent and -independent pathways. American Journal of Physiology—Cell Physiology. 2002;283(3):C714–C721.
    1. Boveris A, Chance B. The mitochondrial generation of hydrogen peroxide. General properties and effect of hyperbaric oxygen. Biochemical Journal. 1973;134(3):707–716.
    1. Loschen G, Azzi A, Richter C, Flohe L. Superoxide radicals as precursors of mitochondrial hydrogen peroxide. FEBS Letters. 1974;42(1):68–72.
    1. Barja G. Mitochondrial oxygen radical generation and leak: sites of production in states 4 and 3, organ specificity, and relation to aging and longevity. Journal of Bioenergetics and Biomembranes. 1999;31(4):347–366.
    1. Muller FL, Liu Y, Van Remmen H. Complex III releases superoxide to both sides of the inner mitochondrial membrane. Journal of Biological Chemistry. 2004;279(47):49064–49073.
    1. Kanter M. Free radicals, exercise and antioxidant supplementation. Proceedings of the Nutrition Society. 1998;57(1):9–13.
    1. Urso ML, Clarkson PM. Oxidative stress, exercise, and antioxidant supplementation. Toxicology. 2003;189(1-2):41–54.
    1. St-Pierre J, Buckingham JA, Roebuck SJ, Brand MD. Topology of superoxide production from different sites in the mitochondrial electron transport chain. Journal of Biological Chemistry. 2002;277(47):44784–44790.
    1. Powers SK, Nelson WB, Hudson MB. Exercise-induced oxidative stress in humans: cause and consequences. Free Radical Biology and Medicine. 2011;51(5):942–950.
    1. Brand MD, Affourtit C, Esteves TC, et al. Mitochondrial superoxide: production, biological effects, and activation of uncoupling proteins. Free Radical Biology and Medicine. 2004;37(6):755–767.
    1. Brand MD, Esteves TC. Physiological functions of the mitochondrial uncoupling proteins UCP2 and UCP3. Cell Metabolism. 2005;2(2):85–93.
    1. Adhihetty PJ, Ljubicic V, Menzies KJ, Hood DA. Differential susceptibility of subsarcolemmal and intermyofibrillar mitochondria to apoptotic stimuli. American Journal of Physiology—Cell Physiology. 2005;289(4):C994–C1001.
    1. Di Meo S, Venditti P. Mitochondria in exercise-induced oxidative stress. Biological Signals and Receptors. 2001;10(1-2):125–140.
    1. Herrero A, Barja G. ADP-Regulation of mitochondrial free radical production is different with complex I- or complex II-linked substrates: implications for the exercise paradox and brain hypermetabolism. Journal of Bioenergetics and Biomembranes. 1997;29(3):241–249.
    1. Kozlov AV, Szalay L, Umar F, et al. Skeletal muscles, heart, and lung are the main sources of oxygen radicals in old rats. Biochimica et Biophysica Acta—Molecular Basis of Disease. 2005;1740(3):382–389.
    1. Piao YJ, Seo YH, Hong F, et al. Nox 2 stimulates muscle differentiation via NF-κB/iNOS pathway. Free Radical Biology and Medicine. 2005;38(8):989–1001.
    1. Powers SK, Jackson MJ. Exercise-induced oxidative stress: cellular mechanisms and impact on muscle force production. Physiological Reviews. 2008;88(4):1243–1276.
    1. Zuo L, Christofi FL, Wright VP, Bao S, Clanton TL. Lipoxygenase-dependent superoxide release in skeletal muscle. Journal of Applied Physiology. 2004;97(2):661–668.
    1. Zhao X, Bey EA, Wientjes FB, Cathcart MK. Cytosolic phospholipase A2 (cPLA2) regulation of human monocyte NADPH oxidase activity: cPLA2 affects translocation but not phosphorylation of p67phox and p47phox. Journal of Biological Chemistry. 2002;277(28):25385–25392.
    1. Nethery D, Callahan LA, Stofan D, Mattera R, DiMarco A, Supinski G. PLA2 dependence of diaphragm mitochondrial formation of reactive oxygen species. Journal of Applied Physiology. 2000;89(1):72–80.
    1. Gong MC, Arbogast S, Guo Z, Mathenia J, Su W, Reid MB. Calcium-independent phospholipase A2 modulates cytosolic oxidant activity and contractile function in murine skeletal muscle cells. Journal of Applied Physiology. 2006;100(2):399–405.
    1. Nethery D, Stofan D, Callahan L, DiMarco A, Supinski G. Formation of reactive oxygen species by the contracting diaphragm is PLA2 dependent. Journal of Applied Physiology. 1999;87(2):792–800.
    1. Gomez-Cabrera MC, Borrás C, Pallardo FV, Sastre J, Ji LL, Viña J. Decreasing xanthine oxidase-mediated oxidative stress prevents useful cellular adaptations to exercise in rats. Journal of Physiology. 2005;567, part 1:113–120.
    1. Gómez-Cabrera MC, Pallardó FV, Sastre J, Viña J, Garcia-del-Moral L. Allopurinol and markers of muscle damage among participants in the Tour de France. Journal of the American Medical Association. 2003;289(19):2503–2504.
    1. Baudry N, Laemmel E, Vicaut E. In vivo reactive oxygen species production induced by ischemia in muscle arterioles of mice: involvement of xanthine oxidase and mitochondria. American Journal of Physiology—Heart and Circulatory Physiology. 2008;294(2):H821–H828.
    1. Li W, Brakefield D, Pan Y, Hunter D, Myckatyn TM, Parsadanian A. Muscle-derived but not centrally derived transgene GDNF is neuroprotective in G93A-SOD1 mouse model of ALS. Experimental Neurology. 2007;203(2):457–471.
    1. Ji LL. Antioxidant signaling in skeletal muscle: a brief review. Experimental Gerontology. 2007;42(7):582–593.
    1. Pedersen BK, Ostrowski K, Rohde T, Bruunsgaard H. The cytokine response to strenuous exercise. Canadian Journal of Physiology and Pharmacology. 1998;76(5):505–511.
    1. Gutteridge JMC, Halliwell B. Free radicals and antioxidants in the year 2000. A historical look to the future. Annals of the New York Academy of Sciences. 2000;899:136–147.
    1. Veal EA, Day AM, Morgan BA. Hydrogen peroxide sensing and signaling. Molecular Cell. 2007;26(1):1–14.
    1. Altun M, Edström E, Spooner E, et al. Iron load and redox stress in skeletal muscle of aged rats. Muscle and Nerve. 2007;36(2):223–233.
    1. Gomez-Cabrera MC, Domenech E, Viña J. Moderate exercise is an antioxidant: upregulation of antioxidant genes by training. Free Radical Biology and Medicine. 2008;44(2):126–131.
    1. Schreck R, Baeuerle A. A role for oxygen radicals as second messengers. Trends in Cell Biology. 1991;1(2-3):39–42.
    1. Bensaad K, Cheung EC, Vousden KH. Modulation of intracellular ROS levels by TIGAR controls autophagy. EMBO Journal. 2009;28(19):3015–3026.
    1. Bensaad K, Tsuruta A, Selak MA, et al. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell. 2006;126(1):107–120.
    1. Sestili P, Martinelli C, Colombo E, et al. Creatine as an antioxidant. Amino Acids. 2011;40(5):1385–1396.
    1. Wallimann T, Tokarska-Schlattner M, Schlattner U. The creatine kinase system and pleiotropic effects of creatine. Amino Acids. 2011;40(5):1271–1296.
    1. Young JF, Larsen LB, Malmendal A, et al. Creatine-induced activation of antioxidative defence in myotube cultures revealed by explorative NMR-based metabonomics and proteomics. Journal of the International Society of Sports Nutrition. 2010;7(1, article 9)
    1. Zong H, Ren JM, Young LH, et al. AMP kinase is required for mitochondrial biogenesis in skeletal muscle in response to chronic energy deprivation. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(25):15983–15987.
    1. Ceddia RB, Sweeney G. Creatine supplementation increases glucose oxidation and AMPK phosphorylation and reduces lactate production in L6 rat skeletal muscle cells. Journal of Physiology. 2004;555, part 2:409–421.
    1. Guidi C, Potenza L, Sestili P, et al. Differential effect of creatine on oxidatively-injured mitochondrial and nuclear DNA. Biochimica et Biophysica Acta—General Subjects. 2008;1780(1):16–26.
    1. Lawler JM, Barnes WS, Wu G, Song W, Demaree S. Direct antioxidant properties of creatine. Biochemical and Biophysical Research Communications. 2002;290(1):47–52.
    1. Gordon A, Hultman E, Kaijser L, et al. Creatine supplementation in chronic heart failure increases skeletal muscle creatine phosphate and muscle performance. Cardiovascular Research. 1995;30(3):413–418.
    1. Matthews RT, Ferrante RJ, Klivenyi P, et al. Creatine and cyclocreatine attenuate MPTP neurotoxicity. Experimental Neurology. 1999;157(1):142–149.
    1. Mazzini L, Balzarini C, Colombo R, et al. Effects of creatine supplementation on exercise performance and muscular strength in amyotrophic lateral sclerosis: preliminary results. Journal of the Neurological Sciences. 2001;191(1-2):139–144.
    1. Stout JR, Eckerson JM, May E, Coulter C, Bradley-Popovich GE. Effects of resistance exercise and creatine supplementation on myasthenia gravis: a case study. Medicine and Science in Sports and Exercise. 2001;33(6):869–872.
    1. Tarnopolsky MA, Mahoney DJ, Vajsar J, et al. Creatine monohydrate enhances strength and body composition in Duchenne muscular dystrophy. Neurology. 2004;62(10):1771–1777.
    1. Vorgerd M, Grehl T, Jäger M, et al. Creatine therapy in myophosphorylase deficiency (McArdle disease): a placebo-controlled crossover trial. Archives of Neurology. 2000;57(7):956–963.
    1. McGinley C, Shafat A, Donnelly AE. Does antioxidant vitamin supplementation protect against muscle damage? Sports Medicine. 2009;39(12):1011–1032.
    1. Nakazato K, Ochi E, Waga T. Dietary apple polyphenols have preventive effects against lengthening contraction-induced muscle injuries. Molecular Nutrition and Food Research. 2010;54(3):364–372.
    1. Strobel NA, Peake JM, Matsumoto A, Marsh SA, Coombes JS, Wadley GD. Antioxidant supplementation reduces skeletal muscle mitochondrial biogenesis. Medicine and Science in Sports and Exercise. 2010;43(6):1017–1024.
    1. Ames BN, Shigenaga MK, Hagen TM. Oxidants, antioxidants, and the degenerative diseases of aging. Proceedings of the National Academy of Sciences of the United States of America. 1993;90(17):7915–7922.
    1. Yu BP, Chung HY. Adaptive mechanisms to oxidative stress during aging. Mechanisms of Ageing and Development. 2006;127(5):436–443.
    1. Ji LL. Exercise at old age: does it increase or alleviate oxidative stress? Annals of the New York Academy of Sciences. 2001;928:236–247.
    1. McArdle A, Vasilaki A, Jackson M. Exercise and skeletal muscle ageing: cellular and molecular mechanisms. Ageing Research Reviews. 2002;1(1):79–93.
    1. Ji LL, Dillon D, Wu E. Alteration of antioxidant enzymes with aging in rat skeletal muscle and liver. American Journal of Physiology. 1990;258(4, part 2):R918–R923.
    1. Luhtala TA, Roecker EB, Pugh T, Feuers RJ, Weindruch R. Dietary restriction attenuates age-related increases in rat skeletal muscle antioxidant enzyme activities. Journals of Gerontology. 1994;49(5):B231–B238.
    1. Hollander J, Bejma J, Ookawara T, Ohno H, Ji LL. Superoxide dismutase gene expression in skeletal muscle: fiber-specific effect of age. Mechanisms of Ageing and Development. 2000;116(1):33–45.
    1. Oh-Ishi S, Toshinai K, Kizaki T, et al. Effects of aging and/or training on antioxidant enzyme system in diaphragm of mice. Respiration Physiology. 1996;105(3):195–202.
    1. Ji LL. Exercise-induced modulation of antioxidant defense. Annals of the New York Academy of Sciences. 2002;959:82–92.
    1. Irrcher I, Ljubicic V, Hood DA. Interactions between ROS and AMP kinase activity in the regulation of PGC-1α transcription in skeletal muscle cells. American Journal of Physiology—Cell Physiology. 2009;296(1):C116–C123.
    1. Jackson MJ. Skeletal muscle aging: role of reactive oxygen species. Critical Care Medicine. 2009;37(10):S368–S371.
    1. Musarò A, Fulle S, Fanò G. Oxidative stress and muscle homeostasis. Current Opinion in Clinical Nutrition and Metabolic Care. 2010;13(3):236–242.
    1. Mammucari C, Rizzuto R. Signaling pathways in mitochondrial dysfunction and aging. Mechanisms of Ageing and Development. 2010;131(7-8):536–543.
    1. Marzetti E, Hwang JCY, Lees HA, et al. Mitochondrial death effectors: relevance to sarcopenia and disuse muscle atrophy. Biochimica et Biophysica Acta—General Subjects. 2010;1800(3):235–244.
    1. Hood DA, Irrcher I, Ljubicic V, Joseph AM. Coordination of metabolic plasticity in skeletal muscle. Journal of Experimental Biology. 2006;209, part 12:2265–2275.
    1. Choi SC, Befroy DE, Codella R, et al. Paradoxical effects of increased expression of PGC-1α on muscle mitochondrial function and insulin-stimulated muscle glucose metabolism. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(50):19926–19931.
    1. Koves TR, Li P, An J, et al. Peroxisome proliferator-activated receptor-γ co-activator 1α-mediated metabolic remodeling of skeletal myocytes mimics exercise training and reverses lipid-induced mitochondrial inefficiency. Journal of Biological Chemistry. 2005;280(39):33588–33598.
    1. Liang H, Ward WF. PGC-1α: a key regulator of energy metabolism. American Journal of Physiology—Advances in Physiology Education. 2006;30(4):145–151.
    1. Michael LF, Wu Z, Cheatham RB, et al. Restoration of insulin-sensitive glucose transporter (GLUT4) gene expression in muscle cells by the transcriptional coactivator PGC-1. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(7):3820–3825.
    1. Uguccioni G, Hood DA. The importance of PGC-1α in contractile activity-induced mitochondrial adaptations. American Journal of Physiology—Endocrinology and Metabolism. 2011;300(2):E361–E371.
    1. Kelly DP, Scarpulla RC. Transcriptional regulatory circuits controlling mitochondrial biogenesis and function. Genes and Development. 2004;18(4):357–368.
    1. Barbieri E, Battistelli M, Casadei L, et al. Morphofunctional and biochemical approaches for studying mitochondrial changes during myoblasts differentiation. Journal of Aging Research. 2011;2011:16 pages. Article ID 845379.
    1. Wu H, Kanatous SB, Thurmond FA, et al. Regulation of mitochondrial biogenesis in skeletal muscle by caMK. Science. 2002;296(5566):349–352.
    1. Irrcher I, Ljubicic V, Kirwan AF, Hood DA. AMP-activated protein kinase-regulated activation of the PGC-1alpha promoter in skeletal muscle cells. PLoS ONE. 2008;3(10) Article ID e3614.
    1. Akimoto T, Pohnert SC, Li P, et al. Exercise stimulates Pgc-1α transcription in skeletal muscle through activation of the p38 MAPK pathway. Journal of Biological Chemistry. 2005;280(20):19587–19593.
    1. Kang C, O’Moore KM, Dickman JR, Ji LL. Exercise activation of muscle peroxisome proliferator-activated receptor-γ coactivator-1α signaling is redox sensitive. Free Radical Biology and Medicine. 2009;47(10):1394–1400.
    1. Spiegelman BM. Transcriptional control of mitochondrial energy metabolism through the PGC1 coactivators. Novartis Foundation Symposium. 2007;287:60–63.
    1. Scherz-Shouval R, Shvets E, Fass E, Shorer H, Gil L, Elazar Z. Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO Journal. 2007;26(7):1749–1760.
    1. Kroemer G, Levine B. Autophagic cell death: the story of a misnomer. Nature Reviews Molecular Cell Biology. 2008;9(12):1004–1010.
    1. Singh R, Cuervo AM. Autophagy in the cellular energetic balance. Cell Metabolism. 2011;13(5):495–504.
    1. Kirisako T, Ichimura Y, Okada H, et al. The reversible modification regulates the membrane-binding state of Apg8/Aut7 essential for autophagy and the cytoplasm to vacuole targeting pathway. Journal of Cell Biology. 2000;151(2):263–275.
    1. Scherz-Shouval R, Shvets E, Elazar Z. Oxidation as a post-translational modification that regulates autophagy. Autophagy. 2007;3(4):371–373.
    1. Chen Y, McMillan-Ward E, Kong J, Israels SJ, Gibson SB. Oxidative stress induces autophagic cell death independent of apoptosis in transformed and cancer cells. Cell Death and Differentiation. 2008;15(1):171–182.
    1. Dobrowolny G, Aucello M, Rizzuto E, et al. Skeletal muscle is a primary target of SOD1G93A-mediated toxicity. Cell Metabolism. 2008;8(5):425–436.
    1. Aucello M, Dobrowolny G, Musarò A. Localized accumulation of oxidative stress causes muscle atrophy through activation of an autophagic pathway. Autophagy. 2009;5(4):527–529.
    1. Mammucari C, Milan G, Romanello V, et al. FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metabolism. 2007;6(6):458–471.
    1. Sandri M. Autophagy in health and disease. 3. Involvement of autophagy in muscle atrophy. American Journal of Physiology—Cell Physiology. 2010;298(6):C1291–C1297.
    1. 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 Metabolism. 2007;6(6):472–483.
    1. Bar-Shai M, Carmeli E, Reznick AZ. The role of NF-κB in protein breakdown in immobilization, aging, and exercise: from basic processes to promotion of health. Annals of the New York Academy of Sciences. 2005;1057:431–447.
    1. Deval C, Mordier S, Obled C, et al. Identification of cathepsin L as a differentially expressed message associated with skeletal muscle wasting. Biochemical Journal. 2001;360, part 1:143–150.
    1. Lemasters JJ. Selective mitochondrial autophagy, or mitophagy, as a targeted defense against oxidative stress, mitochondrial dysfunction, and aging. Rejuvenation Research. 2005;8(1):3–5.
    1. Masiero E, Agatea L, Mammucari C, et al. Autophagy is required to maintain muscle mass. Cell Metabolism. 2009;10(6):507–515.
    1. Tanida I, Ueno T, Kominami E. LC3 conjugation system in mammalian autophagy. International Journal of Biochemistry and Cell Biology. 2004;36(12):2503–2518.
    1. Primeau AJ, Adhihetty PJ, Hood DA. Apoptosis in heart and skeletal muscle. Canadian Journal of Applied Physiology. 2002;27(4):349–395.
    1. Circu ML, Aw TY. Reactive oxygen species, cellular redox systems, and apoptosis. Free Radical Biology and Medicine. 2010;48(6):749–762.
    1. Dupont-Versteegden EE, Strotman BA, Gurley CM, et al. Nuclear translocation of EndoG at the initiation of disuse muscle atrophy and apoptosis is specific to myonuclei. American Journal of Physiology—Regulatory Integrative and Comparative Physiology. 2006;291(6):R1730–R1740.
    1. Madesh M, Hajnóczky G. VDAC-dependent permeabilization of the outer mitochondrial membrane by superoxide induces rapid and massive cytochrome c release. Journal of Cell Biology. 2001;155(6):1003–1015.
    1. Tomasello F, Messina A, Lartigue L, et al. Outer membrane VDAC1 controls permeability transition of the inner mitochondrial membrane in cellulo during stress-induced apoptosis. Cell Research. 2009;19(12):1363–1376.
    1. Hildeman DA, Mitchell T, Aronowt B, Wojciechowski S, Kappler J, Marrack P. Control of Bcl-2 expression by reactive oxygen species. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(25):15035–15040.
    1. Azad N, Iyer A, Vallyathan V, et al. Role of oxidative/nitrosative stress-mediated Bcl-2 regulation in apoptosis and malignant transformation. Annals of the New York Academy of Sciences. 2010;1203:1–6.
    1. Giorgio M, Migliaccio E, Orsini F, et al. Electron transfer between cytochrome c and p66Shc generates reactive oxygen species that trigger mitochondrial apoptosis. Cell. 2005;122(2):221–233.
    1. Pinton P, Rimessi A, Marchi S, et al. Protein kinase C β and prolyl isomerase 1 regulate mitochondrial effects of the life-span determinant p66Shc. Science. 2007;315(5812):659–663.
    1. Dhalla NS, Elmoselhi AB, Hata T, Makino N. Status of myocardial antioxidants in ischemia-reperfusion injury. Cardiovascular Research. 2000;47(3):446–456.
    1. Pellegrino MA, Desaphy J-F, Brocca L, Pierno S, Camerino DC, Bottinelli R. Redox homeostasis, oxidative stress and disuse muscle atrophy. Journal of Physiology. 2011;589(9):2147–2160.
    1. Adhihetty PJ, Taivassalo T, Haller RG, Walkinshaw DR, Hood DA. The effect of training on the expression of mitochondrial biogenesis- and apoptosis-related proteins in skeletal muscle of patients with mtDNA defects. American Journal of Physiology—Endocrinology and Metabolism. 2007;293(3):E672–E680.
    1. Forcales SV, Puri PL. Signaling to the chromatin during skeletal myogenesis: novel targets for pharmacological modulation of gene expression. Seminars in Cell and Developmental Biology. 2005;16(4-5):596–611.
    1. Messina S, Altavilla D, Aguennouz M, et al. Lipid peroxidation inhibition blunts nuclear factor-κB activation, reduces skeletal muscle degeneration, and enhances muscle function in mdx mice. American Journal of Pathology. 2006;168(3):918–926.
    1. Toscano A, Messina S, Campo GM, et al. Oxidative stress in myotonic dystrophy type 1. Free Radical Research. 2005;39(7):771–776.
    1. Fulle S, Protasi F, Di Tano G, et al. The contribution of reactive oxygen species to sarcopenia and muscle ageing. Experimental Gerontology. 2004;39(1):17–24.
    1. Buck M, Chojkier M. Muscle wasting and dedifferentiation induced by oxidative stress in a murine model of cachexia is prevented by inhibitors of nitric oxide synthesis and antioxidants. EMBO Journal. 1996;15(8):1753–1765.
    1. Ardite E, Barbera JA, Roca J, Fernández-Checa JC. Glutathione depletion impairs myogenic differentiation of murine skeletal muscle C2C12 cells through sustained NF-κB activation. American Journal of Pathology. 2004;165(3):719–728.
    1. Dedieu S, Mazères G, Cottin P, Brustis JJ. Involvement of myogenic regulator factors during fusion in the cell line C2C12. International Journal of Developmental Biology. 2002;46(2):235–241.
    1. Ishibashi J, Perry RL, Asakura A, Rudnicki MA. MyoD induces myogenic differentiation through cooperation of its NH 2- and COOH-terminal regions. Journal of Cell Biology. 2005;171(3):471–482.
    1. Louis M, Van Beneden R, Dehoux M, Thissen JP, Francaux M. Creatine increases IGF-I and myogenic regulatory factor mRNA in C 2C12 cells. FEBS Letters. 2004;557(1–3):243–247.
    1. Pawlikowska P, Gajkowska B, Hocquette JF, Orzechowski A. Not only insulin stimulates mitochondriogenesis in muscle cells, but mitochondria are also essential for insulin-mediated myogenesis. Cell Proliferation. 2006;39(2):127–145.
    1. Thaloor D, Miller KJ, Gephart J, Mitchell PO, Pavlath GK. Systemic administration of the NF-κB inhibitor curcumin stimulates muscle regeneration after traumatic injury. American Journal of Physiology—Cell Physiology. 1999;277, part 1(2):C320–C329.
    1. Coletti D, Moresi V, Adamo S, Molinaro M, Sassoon D. Tumor necrosis factor-α gene transfer induces cachexia and inhibits muscle regeneration. Genesis. 2005;43(3):120–128.
    1. Mourkioti F, Kratsios P, Luedde T, et al. Targeted ablation of IKK2 improves skeletal muscle strength, maintains mass, and promotes regeneration. Journal of Clinical Investigation. 2006;116(11):2945–2954.
    1. Goichberg P, Shtutman M, Ben-Ze’ev A, Geiger B. Recruitment of β-catenin to cadherin-mediated intercellular adhesions is involved in myogenic induction. Journal of Cell Science. 2001;114, part 7:1309–1319.
    1. Li YP, Schwartz RJ, Waddell ID, Holloway BR, Reid MB. Skeletal muscle myocytes undergo protein loss and reactive oxygen- mediated NF-κB activation in response to tumor necrosis factor α . FASEB Journal. 1998;12(10):871–880.
    1. Langen RCJ, Schols AMWJ, Kelders MCJM, Wouters EFM, Janssen-Heininger YMW. Inflammatory cytokines inhibit myogenic differentiation through activation of nuclear factor-κB. FASEB Journal. 2001;15(7):1169–1180.
    1. Guttridge DC, Albanese C, Reuther JY, Pestell RG, Baldwin AS. NF-κB controls cell growth and differentiation through transcriptional regulation of cyclin D1. Molecular and Cellular Biology. 1999;19(8):5785–5799.
    1. Guttridge DC, Mayo MW, Madrid LV, Wang CY, Baldwin AS., Jr. NF-κB-induced loss of MyoD messenger RNA: possible role in muscle decay and cachexia. Science. 2000;289(5488):2363–2365.
    1. Parker MH, Perry RLS, Fauteux MC, Berkes CA, Rudnicki MA. MyoD synergizes with the E-protein HEBβ to induce myogenic differentiation. Molecular and Cellular Biology. 2006;26(15):5771–5783.
    1. Wang H, Hertlein E, Bakkar N, et al. NF-κB regulation of YY1 inhibits skeletal myogenesis through transcriptional silencing of myofibrillar genes. Molecular and Cellular Biology. 2007;27(12):4374–4387.
    1. Lee KH, Kim DG, Shin NY, et al. NF-κB-dependent expression of nitric oxide synthase is required for membrane fusion of chick embryonic myoblasts. Biochemical Journal. 1997;324, part 1:237–242.
    1. Kaliman P, Canicio J, Testar X, Palacín M, Zorzano A. Insulin-like growth factor-II, phosphatidylinositol 3-kinase, nuclear factor-κB and inducible nitric-oxide synthase define a common myogenic signaling pathway. Journal of Biological Chemistry. 1999;274(25):17437–17444.
    1. Lee S, Tak E, Lee J, et al. Mitochondrial H2O2 generated from electron transport chain complex i stimulates muscle differentiation. Cell Research. 2011;21(5):817–834.
    1. Migliaccio E, Giogio M, Mele S, et al. The p66(shc) adaptor protein controls oxidative stress response and life span in mammals. Nature. 1999;402(6759):309–313.
    1. Trinei M, Giorgio M, Cicalese A, et al. A p53-p66Shc signalling pathway controls intracellular redox status, levels of oxidation-damaged DNA and oxidative stress-induced apoptosis. Oncogene. 2002;21(24):3872–3878.
    1. Francia P, Delli Gatti C, Bachschmid M, et al. Deletion of p66shc gene protects against age-related endothelial dysfunction. Circulation. 2004;110(18):2889–2895.
    1. Napoli C, Martin-Padura I, de Nigris F, et al. Deletion of the p66Shc longevity gene reduces systemic and tissue oxidative stress, vascular cell apoptosis, and early atherogenesis in mice fed a high-fat diet. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(4):2112–2116.
    1. Zaccagnini G, Martelli F, Fasanaro P, et al. p66ShcA modulates tissue response to hindlimb ischemia. Circulation. 2004;109(23):2917–2923.
    1. Zaccagnini G, Martelli F, Magenta A, et al. p66ShcA and oxidative stress modulate myogenic differentiation and skeletal muscle regeneration after hind limb ischemia. Journal of Biological Chemistry. 2007;282(43):31453–31459.
    1. Fulco M, Schiltz RL, Iezzi S, et al. Sir2 regulates skeletal muscle differentiation as a potential sensor of the redox state. Molecular Cell. 2003;12(1):51–62.
    1. Clempus RE, Griendling KK. Reactive oxygen species signaling in vascular smooth muscle cells. Cardiovascular Research. 2006;71(2):216–225.
    1. Martindale JL, Holbrook NJ. Cellular response to oxidative stress: signaling for suicide and survival. Journal of Cellular Physiology. 2002;192(1):1–15.
    1. Hye SP, Seung HL, Park D, et al. Sequential activation of phosphatidylinositol 3-kinase, βPix, Rac1, and Nox1 in growth factor-induced production of H2O2 . Molecular and Cellular Biology. 2004;24(10):4384–4394.
    1. Arthur PG, Grounds MD, Shavlakadze T. Oxidative stress as a therapeutic target during muscle wasting: considering the complex interactions. Current Opinion in Clinical Nutrition and Metabolic Care. 2008;11(4):408–416.
    1. Moylan JS, Reid MB. Oxidative stress, chronic disease, and muscle wasting. Muscle and Nerve. 2007;35(4):411–429.
    1. Kokoszko A, Dabrowski J, Lewiński A, Karbownik-Lewińska M. Protective effects of GH and IGF-I against iron-induced lipid peroxidation in vivo. Experimental and Toxicologic Pathology. 2008;60(6):453–458.
    1. Yang SY, Hoy M, Fuller B, Sales KM, Seifalian AM, Winslet MC. Pretreatment with insulin-like growth factor i protects skeletal muscle cells against oxidative damage via PI3K/Akt and ERK1/2 MAPK pathways. Laboratory Investigation. 2010;90(3):391–401.
    1. Wallis M. New insulin-like growth factor (IGF)-precursor sequences from mammalian genomes: the molecular evolution of IGFs and associated peptides in primates. Growth Hormone and IGF Research. 2009;19(1):12–23.
    1. Barton ER, Demeo J, Lei H. The insulin-like growth factor (IGF)-I E-peptides are required for isoform-specific gene expression and muscle hypertrophy after local IGF-I production. Journal of Applied Physiology. 2010;108(5):1069–1076.
    1. Dobrowolny G, Giacinti C, Pelosi L, et al. Muscle expression of a local Igf-1 isoform protects motor neurons in an ALS mouse model. Journal of Cell Biology. 2005;168(2):193–199.
    1. Musaró A, Giacinti C, Borsellino G, et al. Stem cell-mediated muscle regeneration is enhanced by local isoform of insulin-like growth factor 1. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(5):1206–1210.
    1. Kravchenko IV, Furalyov VA, Lisitsina ES, Popov VO. Stimulation of mechano-growth factor expression by second messengers. Archives of Biochemistry and Biophysics. 2011;507(2):323–331.
    1. Satoh A, Brace CS, Ben-Josef G, et al. SIRT1 promotes the central adaptive response to diet restriction through activation of the dorsomedial and lateral nuclei of the hypothalamus. Journal of Neuroscience. 2010;30(30):10220–10232.
    1. Berg U, Bang P. Exercise and circulating insulin-like growth factor I. Hormone Research. 2004;62(1):50–58.
    1. Eppler E, Zapf J, Bailer N, Falkmer UG, Reinecke M. IGF-I in human breast cancer: low differentiation stage is associated with decreased IGF-I content. European Journal of Endocrinology. 2002;146(6):813–821.
    1. Philip Karl J, Alemany JA, Koenig C, et al. Diet, body composition, and physical fitness influences on IGF-I bioactivity in women. Growth Hormone and IGF Research. 2009;19(6):491–496.
    1. Papaconstantinou J. Insulin/IGF-1 and ROS signaling pathway cross-talk in aging and longevity determination. Molecular and Cellular Endocrinology. 2009;299(1):89–100.
    1. Bashan N, Kovsan J, Kachko I, Ovadia H, Rudich A. Positive and negative regulation of insulin signaling by reactive oxygen and nitrogen species. Physiological Reviews. 2009;89(1):27–71.
    1. Coderre L, Kandror KV, Vallega G, Pilch PF. Identification and characterization of an exercise-sensitive pool of glucose transporters in skeletal muscle. Journal of Biological Chemistry. 1995;270(46):27584–27588.
    1. Lund S, Holman GD, Schmitz O, Pedersen O. Contraction stimulates translocation of glucose transporter GLUT4 in skeletal muscle through a mechanism distinct from that of insulin. Proceedings of the National Academy of Sciences of the United States of America. 1995;92(13):5817–5821.
    1. Katz A, Broberg S, Sahlin K, Wahren J. Leg glucose uptake during maximal dynamic exercise in humans. American Journal of Physiology. 1986;251(1, part 1):E65–E70.
    1. Higaki Y, Mikami T, Fujii N, et al. Oxidative stress stimulates skeletal muscle glucose uptake through a phosphatidylinositol 3-kinase-dependent pathway. American Journal of Physiology—Endocrinology and Metabolism. 2008;294(5):E889–E897.
    1. Sandström ME, Zhang SJ, Bruton J, et al. Role of reactive oxygen species in contraction-mediated glucose transport in mouse skeletal muscle. Journal of Physiology. 2006;575, part 1:251–262.
    1. Hardie DG, Sakamoto K. AMPK: a key sensor of fuel and energy status in skeletal muscle. Physiology. 2006;21(1):48–60.
    1. Hayashi T, Hirshman MF, Kurth EJ, Winder WW, Goodyear LJ. Evidence for 5’AMP-activated protein kinase mediation of the effect of muscle contraction on glucose transport. Diabetes. 1998;47(8):1369–1373.
    1. Holloszy JO. A forty-year memoir of research on the regulation of glucose transport into muscle. American Journal of Physiology—Endocrinology and Metabolism. 2003;284(3):E453–E467.
    1. Kurth-Kraczek EJ, Hirshman MF, Goodyear LJ, Winder WW. 5’ AMP-activated protein kinase activation causes GLUT4 translocation in skeletal muscle. Diabetes. 1999;48(8):1667–1671.
    1. Fujii N, Jessen N, Goodyear LJ. AMP-activated protein kinase and the regulation of glucose transport. American Journal of Physiology—Endocrinology and Metabolism. 2006;291(5):E867–E877.
    1. Katz A. Modulation of glucose transport in skeletal muscle by reactive oxygen species. Journal of Applied Physiology. 2007;102(4):1671–1676.
    1. Latres E, Amini AR, Amini AA, et al. Insulin-like growth factor-1 (IGF-1) inversely regulates atrophy-induced genes via the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway. Journal of Biological Chemistry. 2005;280(4):2737–2744.
    1. Solomon AM, Bouloux PMG. Modifying muscle mass—The endocrine perspective. Journal of Endocrinology. 2006;191(2):349–360.
    1. Delafontaine P, Ku L. Reactive oxygen species stimulate insulin-like growth factor I synthesis in vascular smooth muscle cells. Cardiovascular Research. 1997;33(1):216–222.
    1. Delafontaine P, Song YH, Li Y. Expression, regulation, and function of IGF-1, IGF-1R, and IGF-1 binding proteins in blood vessels. Arteriosclerosis, Thrombosis and Vascular Biology. 2004;24(3):435–444.
    1. Handayaningsih A-E, Iguchi G, Fukuoka H, et al. Reactive oxygen species play an essential role in IGF-I signaling and IGF-I-induced myocyte hypertrophy in C2C12 myocytes. Endocrinology. 2011;152(3):912–921.
    1. Diaz PT, She ZW, Davis WB, Clanton TL. Hydroxylation of salicylate by the in vitro diaphragm: evidence for hydroxyl radical production during fatigue. Journal of Applied Physiology. 1993;75(2):540–545.
    1. Reid MB, Haack KE, Franchek KM, Valberg PA, Kobzik L, West MS. Reactive oxygen in skeletal muscle. I. Intracellular oxidant kinetics and fatigue in vitro. Journal of Applied Physiology. 1992;73(5):1797–1804.
    1. Zuo L, Christofi FL, Wright VP, et al. Intra- and extracellular measurement of reactive oxygen species produced during heat stress in diaphragm muscle. American Journal of Physiology—Cell Physiology. 2000;279(4):C1058–C1066.
    1. Falk DJ, DeRuisseau KC, Van Gammeren DL, Deering MA, Kavazis AN, Powers SK. Mechanical ventilation promotes redox status alterations in the diaphragm. Journal of Applied Physiology. 2006;101(4):1017–1024.
    1. Zuo L, Clanton TL. Reactive oxygen species formation in the transition to hypoxia in skeletal muscle. American Journal of Physiology—Cell Physiology. 2005;289(1):C207–C216.
    1. Martins AS, Shkryl VM, Nowycky MC, Shirokova N. Reactive oxygen species contribute to Ca2+ signals produced by osmotic stress in mouse skeletal muscle fibres. Journal of Physiology. 2008;586(1):197–210.
    1. Cheng W, Li B, Kajstura J, et al. Stretch-induced programmed myocyte cell death. Journal of Clinical Investigation. 1995;96(5):2247–2259.
    1. Espinosa A, García A, Härtel S, Hidalgo C, Jaimovich E. NADPH oxidase and hydrogen peroxide mediate insulin-induced calcium increase in skeletal muscle cells. Journal of Biological Chemistry. 2009;284(4):2568–2575.
    1. Goossens V, Grooten J, De Vos K, Fiers W. Direct evidence for tumor necrosis factor-induced mitochondrial reactive oxygen intermediates and their involvement in cytotoxicity. Proceedings of the National Academy of Sciences of the United States of America. 1995;92(18):8115–8119.
    1. Thannickal VJ, Fanburg BL. Reactive oxygen species in cell signaling. American Journal of Physiology—Lung Cellular and Molecular Physiology. 2000;279(6):L1005–L1028.
    1. Felty Q, Xiong WC, Sun D, et al. Estrogen-induced mitochondrial reactive oxygen species as signal-transducing messengers. Biochemistry. 2005;44(18):6900–6909.
    1. Zhang A, Jia Z, Guo X, Yang T. Aldosterone induces epithelial-mesenchymal transition via ROS of mitochondrial origin. American Journal of Physiology—Renal Physiology. 2007;293(3):F723–F731.
    1. Wright VP, Reiser PJ, Clanton TL. Redox modulation of global phosphatase activity and protein phosphorylation in intact skeletal muscle. Journal of Physiology. 2009;587, part 23:5767–5781.
    1. Hecht D, Zick Y. Selective inhibition of protein tyrosine phosphatase activities by H2O2 and vanadate in vitro. Biochemical and Biophysical Research Communications. 1992;188(2):773–779.
    1. Heffetz D, Bushkin I, Dror R, Zick Y. The insulinomimetic agents H2O2 and vanadate stimulate protein tyrosine phosphorylation in intact cells. Journal of Biological Chemistry. 1990;265(5):2896–2902.
    1. Agbas A, Hui D, Wang X, Tek V, Zaidi A, Michaelis EK. Activation of brain calcineurin (Cn) by Cu-Zn superoxide dismutase (SOD1) depends on direct SOD1-Cn protein interactions occurring in vitro and in vivo. Biochemical Journal. 2007;405(1):51–59.
    1. Namgaladze D, Hofer HW, Ullrich V. Redox control of calcineurin by targeting the binuclear Fe2+-Zn2+ center at the enzyme active site. Journal of Biological Chemistry. 2002;277(8):5962–5969.
    1. Namgaladze D, Shcherbyna I, Kienhöfer J, Hofer HW, Ullrich V. Superoxide targets calcineurin signaling in vascular endothelium. Biochemical and Biophysical Research Communications. 2005;334(4):1061–1067.
    1. Powers SK, Kavazis AN, DeRuisseau KC. Mechanisms of disuse muscle atrophy: role of oxidative stress. American Journal of Physiology—Regulatory Integrative and Comparative Physiology. 2005;288(2):R337–R344.
    1. Powers SK, Kavazis AN, McClung JM. Oxidative stress and disuse muscle atrophy. Journal of Applied Physiology. 2007;102(6):2389–2397.
    1. Hamilton KL, Staib JL, Phillips T, Hess A, Lennon SL, Powers SK. Exercise, antioxidants, and HSP72: protection against myocardial ischemia/reperfusion. Free Radical Biology and Medicine. 2003;34(7):800–809.
    1. Ristow M, Zarse K, Oberbach A, et al. Antioxidants prevent health-promoting effects of physical exercise in humans. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(21):8665–8670.

Source: PubMed

3
Suscribir