Deficient nitric oxide signalling impairs skeletal muscle growth and performance: involvement of mitochondrial dysregulation

Clara De Palma, Federica Morisi, Sarah Pambianco, Emma Assi, Thierry Touvier, Stefania Russo, Cristiana Perrotta, Vanina Romanello, Silvia Carnio, Valentina Cappello, Paolo Pellegrino, Claudia Moscheni, Maria Teresa Bassi, Marco Sandri, Davide Cervia, Emilio Clementi, Clara De Palma, Federica Morisi, Sarah Pambianco, Emma Assi, Thierry Touvier, Stefania Russo, Cristiana Perrotta, Vanina Romanello, Silvia Carnio, Valentina Cappello, Paolo Pellegrino, Claudia Moscheni, Maria Teresa Bassi, Marco Sandri, Davide Cervia, Emilio Clementi

Abstract

Background: Nitric oxide (NO), generated in skeletal muscle mostly by the neuronal NO synthases (nNOSμ), has profound effects on both mitochondrial bioenergetics and muscle development and function. The importance of NO for muscle repair emerges from the observation that nNOS signalling is defective in many genetically diverse skeletal muscle diseases in which muscle repair is dysregulated. How the effects of NO/nNOSμ on mitochondria impact on muscle function, however, has not been investigated yet.

Methods: In this study we have examined the relationship between the NO system, mitochondrial structure/activity and skeletal muscle phenotype/growth/functions using a mouse model in which nNOSμ is absent. Also, NO-induced effects and the NO pathway were dissected in myogenic precursor cells.

Results: We show that nNOSμ deficiency in mouse skeletal muscle leads to altered mitochondrial bioenergetics and network remodelling, and increased mitochondrial unfolded protein response (UPR(mt)) and autophagy. The absence of nNOSμ is also accompanied by an altered mitochondrial homeostasis in myogenic precursor cells with a decrease in the number of myonuclei per fibre and impaired muscle development at early stages of perinatal growth. No alterations were observed, however, in the overall resting muscle structure, apart from a reduced specific muscle mass and cross sectional areas of the myofibres. Investigating the molecular mechanisms we found that nNOSμ deficiency was associated with an inhibition of the Akt-mammalian target of rapamycin pathway. Concomitantly, the Akt-FoxO3-mitochondrial E3 ubiquitin protein ligase 1 (Mul-1) axis was also dysregulated. In particular, inhibition of nNOS/NO/cyclic guanosine monophosphate (cGMP)/cGMP-dependent-protein kinases induced the transcriptional activity of FoxO3 and increased Mul-1 expression. nNOSμ deficiency was also accompanied by functional changes in muscle with reduced muscle force, decreased resistance to fatigue and increased degeneration/damage post-exercise.

Conclusions: Our results indicate that nNOSμ/NO is required to regulate key homeostatic mechanisms in skeletal muscle, namely mitochondrial bioenergetics and network remodelling, UPR(mt) and autophagy. These events are likely associated with nNOSμ-dependent impairments of muscle fibre growth resulting in a deficit of muscle performance.

Keywords: Akt-FoxO3-Mul-1 axis; Akt-mTOR pathway; Autophagy; Fibre growth; Mitochondrial bioenergetics; Mitochondrial network; Muscle exercise; Muscle structure; Nitric oxide synthase and signalling; Unfolded protein response.

Figures

Figure 1
Figure 1
Mitochondrial metabolism is impaired in skeletal muscles of NOS1-/- mice. Fibres were isolated from different muscles of wild-type and NOS1-/- mice at P120. (A) Mitochondrial membrane potential measured in fibres isolated from flexor digitorum brevis muscles, loaded with TMRM and treated with 5 μM oligomycin (Olm) or 4 μM FCCP. TMRM staining was monitored in six to ten fibres obtained from at least three different animals per experimental group. Data are expressed by setting the initial value as 1. (B) ATP production on mitochondria isolated from tibialis anterior and diaphragm muscles, at 10 minutes after substrate addition. Data are expressed by setting the initial value as 1. (C-D) Oxygen consumption on fibres isolated from tibialis anterior and diaphragm muscles, supplied with specific CI, CII and CIV mitochondrial complex substrates, as indicated in the Methods. (E) Quantitative analysis of the mtDNA copy number. Data are expressed by normalizing mtDNA values versus nuclear DNA. Each histogram represents the data obtained from at least five different animals per experimental group. * P <0.05 and ** P <0.01 versus the respective wild-type control.
Figure 2
Figure 2
Mitochondrial morphology, UPRmtand autophagy in skeletal muscles of NOS1-/- mice.Tibialis anterior muscles were isolated from wild-type and NOS1-/- mice at P120. (A)In vivo imaging of the mitochondrial network by two-photon confocal microscopy. Muscles were transfected with the mitochondrially targeted red fluorescent protein pDsRed2-Mito. The images are representative of results obtained from at least five different animals per experimental group. Scale bar: 10 μm. (B) TEM images detecting the presence of abnormal, enlarged subsarcolemmal mitochondria (asterisks) or autophagic vacuoles (arrowheads) in NOS1-/- muscles. The inset depicts a multivesicular body in NOS1-/- fibres taken at higher magnification. The images are representative of results obtained from at least three different animals per experimental group. (C-D) Subsarcolemmal mitochondrial ultrastructure analysis by TEM. Data represent the quantification of the mitochondrial area and morphometric analysis of mitochondrial cristae complexity. Each histogram represents the data obtained from at least three different animals per experimental group. * P <0.05 and ** P <0.01 versus the respective wild-type control. (E) Western blot analysis of HSP60 and ClpP expression. Actin was used as the internal standard. The image is representative of results obtained from at least five to seven different animals per experimental group. (F)In vivo imaging of autophagosome formation by two-photon confocal microscopy. Muscles were transfected with YFP-LC3. The images are representative of results obtained from at least five different animals per experimental group. Scale bar: 10 μm. (G) Western blot analysis of LC3 lipidation. Actin was used as the internal standard. The image is representative of results obtained from at least 10 different animals per experimental group.
Figure 3
Figure 3
NO signalling, UPRmt, and autophagy on myogenic precursor cells. Cells were differentiated for six hours in the absence (control) or in the presence of L-NAME (6 mM), ODQ (10 μM), KT5823 (1 μM), L-NAME + DETA-NO (80 μM), and ODQ +8 Br-cGMP (2.5 mM). (A) Western blot analysis of HSP60 and ClpP expression. Actin was used as the internal standard. (B) Confocal microscopy imaging of cells transfected with YFP-LC3. Mitochondrial morphology was detected by mitochondrial matrix-specific protein cyclophillin D (CypD) staining. Scale Bar: 10 μm. Images are representative of at least three to five independent experiments..
Figure 4
Figure 4
NO signalling and autophagic pathway on myogenic precursor cells. (A) Western blot analysis of LC3 lipidation in cells differentiated for six hours in the absence or in the presence of L-NAME (6 mM), ODQ (10 μM), KT5823 (1 μM), L-NAME + DETA-NO (80 μM), and ODQ +8 Br-cGMP (2.5 mM). Actin was used as the internal standard. Image is representative of at least five independent experiments. (B) qPCR analysis of mRNA levels for p62, Bnip3 and Atg4 in cells differentiated for six hours in the absence (control) or in the presence of L-NAME ODQ, and KT5823. Values are expressed as the fold change over control. Each histogram represents the data obtained from at least five independent experiments. * P <0.05 versus respective control.
Figure 5
Figure 5
NO signalling, FoxO3, and ubiquitin ligases. Western blot analysis of phosphorylated FoxO3 levels (pFoxO3) or mitochondrial ubiquitin ligase Mul-1 expression in tibialis anterior(A-B) and diaphragm (C-D) of wild-type and NOS1-/- mice at P120. FoxO3 or actin were used as the internal standard. The images are representative of results obtained from at least four to ten different animals per experimental group. (E) Western blot analysis of Mul-1 expression in myogenic precursor cells differentiated in the absence or in the presence of L-NAME (6 mM), ODQ (10 μM), KT5823 (1 μM), L-NAME + DETA-NO (80 μM) and ODQ +8 Br-cGMP (2.5 mM). Actin was used as the internal standard. The image is representative of at least five independent experiments. qPCR analysis of mRNA levels for atrogin-1, muRF1 and MUSA1 in tibialis anterior(F) and diaphragm (G) muscles of wild-type and NOS1-/- mice at P120. Values are expressed as the fold change over wild-type. Each histogram represents the data obtained from at least five to eight different animals per experimental group. * P <0.05 versus the respective wild-type control.
Figure 6
Figure 6
Skeletal muscle phenotype of wild-type and NOS1-/- mice at P120. (A) Weight of tibialis anterior, gastrocnemius, soleus, and extensor digitorum longus (EDL) muscles. The muscle size is relative to body weight. Each histogram represents the data obtained from at least 10 different animals per experimental group. (B) The number of myofibres in tibialis anterior. Each histogram represents the data obtained from at least four to five different animals per experimental group. Laminin staining of tibialis anterior(C-E) and diaphragm (F-H) muscles. (C, F) Immunohistochemical images. Scale bar: 100 μm. (D, G) Representative distribution of CSA values. (E, H) Quantification of CSA. Images and quantifications represent the data obtained from at least four to seven different animals per experimental group. *P <0.05, **P <0.01, and ***P <0.001 versus the respective wild-type control.
Figure 7
Figure 7
Skeletal muscle phenotype of wild-type and NOS1-/- mice at P10. (A-C) Laminin staining of hind limb muscles. (A) Immunohistochemical images. Scale bar: 100 μm. (B) Representative distribution of CSA values. (C) Quantification of CSA. Images and quantifications represent the data obtained from at least five different animals per experimental group. (D) Number of myonuclei per fibre in hind limb muscles. Each histogram represents the data obtained from at least three different animals per experimental group. (E) Western blot analysis of myosin (MF20) and MyoD expression in myogenic precursor cells isolated from wild-type and NOS1-/- mice and differentiated for increasing times. Calnexin was used as the internal standard. Images are representative of at least three independent experiments. (F) Confocal microscopy imaging of myogenic precursor cells isolated from wild-type and NOS1-/- mice and differentiated for 48 hours. Mitochondrial morphology was detected by mitochondrial matrix-specific protein cyclophillin D staining. Scale Bar: 10 μm. Images are representative of at least three independent experiments. * P <0.05 versus the respective wild-type control.
Figure 8
Figure 8
Skeletal muscle phenotype of wild-type and NOS1-/- mice at P30. (A-C) Laminin staining of tibialis anterior muscles. (A) Immunohistochemical images. Scale bar: 100 μm. (B) Representative distribution of CSA values. (C) Quantification of CSA. Images and quantifications represent the data obtained from at least five different animals per experimental group. Western blot analysis in tibialis anterior: (D) phosphorylated S6, 4E-BP1 and Akt levels, (E) phosphorylated FoxO3 levels or mitochondrial ubiquitin ligase Mul-1 expression. S6, 4E-BP1, Akt, FoxO3 or actin were used as the internal standard. The images are representative of results obtained from at least four different animals per experimental group. *P <0.05 versus the respective wild-type control.
Figure 9
Figure 9
Skeletal muscle function in wild-type and NOS1-/- mice. (A) WBT measurements determined by dividing the average of the top ten or top five forward pulling tensions, respectively, by the body weight. (B) Running distance calculated during one bout of exhaustive treadmill running (day 1) and after repeated challenges (days 2 and 3). (C) Treadmill runtime to exhaustion calculated as the averages obtained at day 1 to 3. Each histogram represents the data obtained from at least four to five different animals per experimental group. (D) TEM analysis performed in tibialis anterior muscles of both unchallenged (no run) and challenged (exhaustive running) mice. The images are representative of results obtained from at least three different animals per experimental group. (E) EBD uptake in tibialis anterior muscles after the treadmill running. Scale Bar: 100 μm. The images are representative of results obtained from at least four different animals per experimental group. (F) CK serum levels (units per litre) of mice after treadmill running. Each histogram represents the data obtained from at least four different animals per experimental group. *P <0.05, **P <0.01 and ***P <0.001 versus the respective wild-type control. WBT and treadmill running were performed on animals at P120.

References

    1. Alderton WK, Cooper CE, Knowles RG. Nitric oxide synthases: structure, function and inhibition. Biochem J. 2001;357:593–615. doi: 10.1042/0264-6021:3570593.
    1. Stamler JS, Meissner G. Physiology of nitric oxide in skeletal muscle. Physiol Rev. 2001;81:209–237.
    1. Brenman JE, Chao DS, Xia H, Aldape K, Bredt DS. Nitric oxide synthase complexed with dystrophin and absent from skeletal muscle sarcolemma in Duchenne muscular dystrophy. Cell. 1995;82:743–752. doi: 10.1016/0092-8674(95)90471-9.
    1. Rubinstein I, Abassi Z, Coleman R, Milman F, Winaver J, Better OS. Involvement of nitric oxide system in experimental muscle crush injury. J Clin Invest. 1998;101:1325–1333. doi: 10.1172/JCI810.
    1. De Palma C, Clementi E. Nitric oxide in myogenesis and therapeutic muscle repair. Mol Neurobiol. 2012;46:682–692. doi: 10.1007/s12035-012-8311-8.
    1. McConell GK, Rattigan S, Lee-Young RS, Wadley GD, Merry TL. Skeletal muscle nitric oxide signaling and exercise: a focus on glucose metabolism. Am J Physiol Endocrinol Metab. 2012;303:E301–307. doi: 10.1152/ajpendo.00667.2011.
    1. Eu JP, Hare JM, Hess DT, Skaf M, Sun J, Cardenas-Navina I, Sun QA, Dewhirst M, Meissner G, Stamler JS. Concerted regulation of skeletal muscle contractility by oxygen tension and endogenous nitric oxide. Proc Natl Acad Sci U S A. 2003;100:15229–15234. doi: 10.1073/pnas.2433468100.
    1. Buono R, Vantaggiato C, Pisa V, Azzoni E, Bassi MT, Brunelli S, Sciorati C, Clementi E. Nitric oxide sustains long-term skeletal muscle regeneration by regulating fate of satellite cells via signaling pathways requiring Vangl2 and cyclic GMP. Stem Cells. 2012;30:197–209. doi: 10.1002/stem.783.
    1. Cordani N, Pisa V, Pozzi L, Sciorati C, Clementi E. Nitric oxide controls fat deposition in dystrophic skeletal muscle by regulating fibro-adipogenic precursor differentiation. Stem Cells. 2014;32:874–885. doi: 10.1002/stem.1587.
    1. Chao DS, Gorospe JR, Brenman JE, Rafael JA, Peters MF, Froehner SC, Hoffman EP, Chamberlain JS, Bredt DS. Selective loss of sarcolemmal nitric oxide synthase in Becker muscular dystrophy. J Exp Med. 1996;184:609–618. doi: 10.1084/jem.184.2.609.
    1. Crosbie RH, Barresi R, Campbell KP. Loss of sarcolemma nNOS in sarcoglycan-deficient muscle. FASEB J. 2002;16:1786–1791. doi: 10.1096/fj.02-0519com.
    1. Kobayashi YM, Rader EP, Crawford RW, Iyengar NK, Thedens DR, Faulkner JA, Parikh SV, Weiss RM, Chamberlain JS, Moore SA, Campbell KP. Sarcolemma-localized nNOS is required to maintain activity after mild exercise. Nature. 2008;456:511–515. doi: 10.1038/nature07414.
    1. Gucuyener K, Ergenekon E, Erbas D, Pinarli G, Serdaroglu A. The serum nitric oxide levels in patients with Duchenne muscular dystrophy. Brain Dev. 2000;22:181–183. doi: 10.1016/S0387-7604(00)00106-6.
    1. Wehling M, Spencer MJ, Tidball JG. A nitric oxide synthase transgene ameliorates muscular dystrophy in mdx mice. J Cell Biol. 2001;155:123–131. doi: 10.1083/jcb.200105110.
    1. Wehling-Henricks M, Oltmann M, Rinaldi C, Myung KH, Tidball JG. Loss of positive allosteric interactions between neuronal nitric oxide synthase and phosphofructokinase contributes to defects in glycolysis and increased fatigability in muscular dystrophy. Hum Mol Genet. 2009;18:3439–3451. doi: 10.1093/hmg/ddp288.
    1. Brunelli S, Sciorati C, D'Antona G, Innocenzi A, Covarello D, Galvez BG, Perrotta C, Monopoli A, Sanvito F, Bottinelli R, Ongini E, Cossu G, Clementi E. Nitric oxide release combined with nonsteroidal antiinflammatory activity prevents muscular dystrophy pathology and enhances stem cell therapy. Proc Natl Acad Sci U S A. 2007;104:264–269. doi: 10.1073/pnas.0608277104.
    1. Deponti D, Francois S, Baesso S, Sciorati C, Innocenzi A, Broccoli V, Muscatelli F, Meneveri R, Clementi E, Cossu G, Brunelli S. Necdin mediates skeletal muscle regeneration by promoting myoblast survival and differentiation. J Cell Biol. 2007;179:305–319. doi: 10.1083/jcb.200701027.
    1. Sciorati C, Buono R, Azzoni E, Casati S, Ciuffreda P, D'Angelo G, Cattaneo D, Brunelli S, Clementi E. Co-administration of ibuprofen and nitric oxide is an effective experimental therapy for muscular dystrophy, with immediate applicability to humans. Br J Pharmacol. 2010;160:1550–1560. doi: 10.1111/j.1476-5381.2010.00809.x.
    1. D'Angelo MG, Gandossini S, Martinelli Boneschi F, Sciorati C, Bonato S, Brighina E, Comi GP, Turconi AC, Magri F, Stefanoni G, Brunelli S, Bresolin N, Cattaneo D, Clementi E. Nitric oxide donor and non steroidal anti inflammatory drugs as a therapy for muscular dystrophies: evidence from a safety study with pilot efficacy measures in adult dystrophic patients. Pharmacol Res. 2012;65:472–479. doi: 10.1016/j.phrs.2012.01.006.
    1. Cossu MV, Cattaneo D, Fucile S, Pellegrino PM, Baldelli S, Cozzi V, Capetti A, Clementi E. Combined isosorbide dinitrate and ibuprofen as a novel therapy for muscular dystrophies: evidence from Phase I studies in healthy volunteers. Drug Des Devel Ther. 2014;8:411–419.
    1. Tengan CH, Rodrigues GS, Godinho RO. Nitric oxide in skeletal muscle: role on mitochondrial biogenesis and function. Int J Mol Sci. 2012;13:17160–17184. doi: 10.3390/ijms131217160.
    1. Nisoli E, Falcone S, Tonello C, Cozzi V, Palomba L, Fiorani M, Pisconti A, Brunelli S, Cardile A, Francolini M, Cantoni O, Carruba MO, Moncada S, Clementi E. Mitochondrial biogenesis by NO yields functionally active mitochondria in mammals. Proc Natl Acad Sci U S A. 2004;101:16507–16512. doi: 10.1073/pnas.0405432101.
    1. Clementi E, Nisoli E. Nitric oxide and mitochondrial biogenesis: a key to long-term regulation of cellular metabolism. Comp Biochem Physiol A Mol Integr Physiol. 2005;142:102–110. doi: 10.1016/j.cbpb.2005.04.022.
    1. Cleeter MW, Cooper JM, Darleyusmar VM, Moncada S, Schapira AH. Reversible inhibition of cytochrome-C-oxidase, the terminal enzyme of the mitochondrial respiratory-chain, by nitric-oxide - implications for neurodegenerative diseases. FEBS Lett. 1994;345:50–54. doi: 10.1016/0014-5793(94)00424-2.
    1. De Palma C, Falcone S, Pisoni S, Cipolat S, Panzeri C, Pambianco S, Pisconti A, Allevi R, Bassi MT, Cossu G, Pozzan T, Moncada S, Scorrano L, Brunelli S, Clementi E. Nitric oxide inhibition of Drp1-mediated mitochondrial fission is critical for myogenic differentiation. Cell Death Differ. 2010;17:1684–1696. doi: 10.1038/cdd.2010.48.
    1. Russell AP, Foletta VC, Snow RJ, Wadley GD. Skeletal muscle mitochondria: A major player in exercise, health and disease. Biochim Biophys Acta. 1840;2014:1276–1284.
    1. Wagatsuma A, Sakuma K. Mitochondria as a potential regulator of myogenesis. ScientificWorldJournal. 2013;2013:593267. doi: 10.1155/2013/593267.
    1. Eisner V, Lenaers G, Hajnoczky G. Mitochondrial fusion is frequent in skeletal muscle and supports excitation-contraction coupling. J Cell Biol. 2014;205:179–195. doi: 10.1083/jcb.201312066.
    1. Romanello V, Guadagnin E, Gomes L, Roder I, Sandri C, Petersen Y, Milan G, Masiero E, Del Piccolo P, Foretz M, Scorrano L, Rudolf R, Sandri M. Mitochondrial fission and remodelling contributes to muscle atrophy. EMBO J. 2010;29:1774–1785. doi: 10.1038/emboj.2010.60.
    1. Gomes LC, Di Benedetto G, Scorrano L. During autophagy mitochondria elongate, are spared from degradation and sustain cell viability. Nat Cell Biol. 2011;13:589–598. doi: 10.1038/ncb2220.
    1. Sandri M, Coletto L, Grumati P, Bonaldo P. Misregulation of autophagy and protein degradation systems in myopathies and muscular dystrophies. J Cell Sci. 2013;126:5325–5333. doi: 10.1242/jcs.114041.
    1. Grumati P, Coletto L, Sandri M, Bonaldo P. Autophagy induction rescues muscular dystrophy. Autophagy. 2011;7:426–428. doi: 10.4161/auto.7.4.14392.
    1. Grumati P, Coletto L, Sabatelli P, Cescon M, Angelin A, Bertaggia E, Blaauw B, Urciuolo A, Tiepolo T, Merlini L, Maraldi NM, Bernardi P, Sandri M, Bonaldo P. Autophagy is defective in collagen VI muscular dystrophies, and its reactivation rescues myofiber degeneration. Nat Med. 2010;16:1313–1320. doi: 10.1038/nm.2247.
    1. Haynes CM, Ron D. The mitochondrial UPR - protecting organelle protein homeostasis. J Cell Sci. 2010;123:3849–3855. doi: 10.1242/jcs.075119.
    1. Acosta-Alvear D, Zhou Y, Blais A, Tsikitis M, Lents NH, Arias C, Lennon CJ, Kluger Y, Dynlacht BD. XBP1 controls diverse cell type- and condition-specific transcriptional regulatory networks. Mol Cell. 2007;27:53–66. doi: 10.1016/j.molcel.2007.06.011.
    1. Iwawaki T, Akai R, Kohno K, Miura M. A transgenic mouse model for monitoring endoplasmic reticulum stress. Nat Med. 2004;10:98–102. doi: 10.1038/nm970.
    1. Percival JM, Anderson KN, Huang P, Adams ME, Froehner SC. Golgi and sarcolemmal neuronal NOS differentially regulate contraction-induced fatigue and vasoconstriction in exercising mouse skeletal muscle. J Clin Invest. 2010;120:816–826. doi: 10.1172/JCI40736.
    1. Mammucari C, Milan G, Romanello V, Masiero E, Rudolf R, Del Piccolo P, Burden SJ, Di Lisi R, Sandri C, Zhao J, Goldberg AL, Schiaffino S, Sandri M. FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab. 2007;6:458–471. doi: 10.1016/j.cmet.2007.11.001.
    1. Cervia D, Garcia-Gil M, Simonetti E, Di Giuseppe G, Guella G, Bagnoli P, Dini F. Molecular mechanisms of euplotin C-induced apoptosis: involvement of mitochondrial dysfunction, oxidative stress and proteases. Apoptosis. 2007;12:1349–1363. doi: 10.1007/s10495-007-0075-7.
    1. Kuznetsov AV, Veksler V, Gellerich FN, Saks V, Margreiter R, Kunz WS. Analysis of mitochondrial function in situ in permeabilized muscle fibers, tissues and cells. Nat Protoc. 2008;3:965–976. doi: 10.1038/nprot.2008.61.
    1. Votion DM, Gnaiger E, Lemieux H, Mouithys-Mickalad A, Serteyn D. Physical fitness and mitochondrial respiratory capacity in horse skeletal muscle. PLoS One. 2012;7:e34890. doi: 10.1371/journal.pone.0034890.
    1. Jacobs RA, Boushel R, Wright-Paradis C, Calbet JA, Robach P, Gnaiger E, Lundby C. Mitochondrial function in human skeletal muscle following high-altitude exposure. Exp Physiol. 2013;98:245–255. doi: 10.1113/expphysiol.2012.066092.
    1. Perrotta C, Buldorini M, Assi E, Cazzato D, De Palma C, Clementi E, Cervia D. The thyroid hormone triiodothyronine controls macrophage maturation and functions: protective role during inflammation. Am J Pathol. 2014;184:230–247. doi: 10.1016/j.ajpath.2013.10.006.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2-DDCT Method. Methods. 2001;25:402–408. doi: 10.1006/meth.2001.1262.
    1. Mouchiroud L, Houtkooper RH, Moullan N, Katsyuba E, Ryu D, Canto C, Mottis A, Jo YS, Viswanathan M, Schoonjans K, Guarente L, Auwerx J. 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. Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh K, Schiaffino S, Lecker SH, Goldberg AL. Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell. 2004;117:399–412. doi: 10.1016/S0092-8674(04)00400-3.
    1. De Palma C, Morisi F, Cheli S, Pambianco S, Cappello V, Vezzoli M, Rovere-Querini P, Moggio M, Ripolone M, Francolini M, Sandri M, Clementi E. Autophagy as a new therapeutic target in Duchenne muscular dystrophy. Cell Death Dis. 2012;3:e418. doi: 10.1038/cddis.2012.159.
    1. Francolini M, Brunelli G, Cambianica I, Barlati S, Barbon A, La Via L, Guarneri B, Boroni F, Lanzillotta A, Baiguera C, Ettorre M, Buffelli M, Spano P, Clementi F, Pizzi M. Glutamatergic reinnervation and assembly of glutamatergic synapses in adult rat skeletal muscle occurs at cholinergic endplates. J Neuropathol Exp Neurol. 2009;68:1103–1115. doi: 10.1097/NEN.0b013e3181b7bfc8.
    1. Bizzozero L, Cazzato D, Cervia D, Assi E, Simbari F, Pagni F, De Palma C, Monno A, Verdelli C, Querini PR, Russo V, Clementi E, Perrotta C. Acid sphingomyelinase determines melanoma progression and metastatic behaviour via the microphtalmia-associated transcription factor signalling pathway. Cell Death Differ. 2014;21:507–520. doi: 10.1038/cdd.2013.173.
    1. Armani C, Catalani E, Balbarini A, Bagnoli P, Cervia D. Expression, pharmacology, and functional role of somatostatin receptor subtypes 1 and 2 in human macrophages. J Leukoc Biol. 2007;81:845–855. doi: 10.1189/jlb.0606417.
    1. Sciorati C, Touvier T, Buono R, Pessina P, Francois S, Perrotta C, Meneveri R, Clementi E, Brunelli S. Necdin is expressed in cachectic skeletal muscle to protect fibers from tumor-induced wasting. J Cell Sci. 2009;122:1119–1125. doi: 10.1242/jcs.041665.
    1. Carlson CG, Rutter J, Bledsoe C, Singh R, Hoff H, Bruemmer K, Sesti J, Gatti F, Berge J, McCarthy L. A simple protocol for assessing inter-trial and inter-examiner reliability for two noninvasive measures of limb muscle strength. J Neurosci Methods. 2010;186:226–230. doi: 10.1016/j.jneumeth.2009.11.006.
    1. George Carlson C, Bruemmer K, Sesti J, Stefanski C, Curtis H, Ucran J, Lachey J, Seehra JS. Soluble activin receptor type IIB increases forward pulling tension in the mdx mouse. Muscle Nerve. 2011;43:694–699. doi: 10.1002/mus.21944.
    1. Wu J, Ruas JL, Estall JL, Rasbach KA, Choi JH, Ye L, Bostrom P, Tyra HM, Crawford RW, Campbell KP, Rutkowski DT, Kaufman RJ, Spiegelman BM. The unfolded protein response mediates adaptation to exercise in skeletal muscle through a PGC-1alpha/ATF6alpha complex. Cell Metab. 2011;13:160–169. doi: 10.1016/j.cmet.2011.01.003.
    1. Sandri M. Autophagy in skeletal muscle. FEBS Lett. 2010;584:1411–1416. doi: 10.1016/j.febslet.2010.01.056.
    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. Zhao Q, Wang J, Levichkin IV, Stasinopoulos S, Ryan MT, Hoogenraad NJ. A mitochondrial specific stress response in mammalian cells. EMBO J. 2002;21:4411–4419. doi: 10.1093/emboj/cdf445.
    1. Ju JS, Varadhachary AS, Miller SE, Weihl CC. Quantitation of "autophagic flux" in mature skeletal muscle. Autophagy. 2010;6:929–935. doi: 10.4161/auto.6.7.12785.
    1. Fader CM, Colombo MI. Autophagy and multivesicular bodies: two closely related partners. Cell Death Differ. 2009;16:70–78. doi: 10.1038/cdd.2008.168.
    1. Francis SH, Busch JL, Corbin JD, Sibley D. cGMP-dependent protein kinases and cGMP phosphodiesterases in nitric oxide and cGMP action. Pharmacol Rev. 2010;62:525–563. doi: 10.1124/pr.110.002907.
    1. De Palma C, Di Paola R, Perrotta C, Mazzon E, Cattaneo D, Trabucchi E, Cuzzocrea S, Clementi E. Ibuprofen-arginine generates nitric oxide and has enhanced anti-inflammatory effects. Pharmacol Res. 2009;60:221–228. doi: 10.1016/j.phrs.2009.06.002.
    1. Cazzato D, Assi E, Moscheni C, Brunelli S, De Palma C, Cervia D, Perrotta C, Clementi E. Nitric oxide drives embryonic myogenesis in chicken through the upregulation of myogenic differentiation factors. Exp Cell Res. 2014;320:269–280. doi: 10.1016/j.yexcr.2013.11.006.
    1. Perrotta C, Bizzozero L, Falcone S, Rovere-Querini P, Prinetti A, Schuchman EH, Sonnino S, Manfredi AA, Clementi E. Nitric oxide boosts chemoimmunotherapy via inhibition of acid sphingomyelinase in a mouse model of melanoma. Cancer Res. 2007;67:7559–7564. doi: 10.1158/0008-5472.CAN-07-0309.
    1. Paolucci C, Rovere P, De Nadai C, Manfredi AA, Clementi E. Nitric oxide inhibits the tumor necrosis factor alpha -regulated endocytosis of human dendritic cells in a cyclic GMP-dependent way. J Biol Chem. 2000;275:19638–19644. doi: 10.1074/jbc.M000511200.
    1. Falcone S, Perrotta C, De Palma C, Pisconti A, Sciorati C, Capobianco A, Rovere-Querini P, Manfredi AA, Clementi E. Activation of acid sphingomyelinase and its inhibition by the nitric oxide/cyclic guanosine 3',5'-monophosphate pathway: key events in Escherichia coli-elicited apoptosis of dendritic cells. J Immunol. 2004;173:4452–4463. doi: 10.4049/jimmunol.173.7.4452.
    1. Clementi E, Sciorati C, Nistico G. Growth factor-induced Ca2+ responses are differentially modulated by nitric oxide via activation of a cyclic GMP-dependent pathway. Mol Pharmacol. 1995;48:1068–1077.
    1. Mehrpour M, Esclatine A, Beau I, Codogno P. Overview of macroautophagy regulation in mammalian cells. Cell Res. 2010;20:748–762. doi: 10.1038/cr.2010.82.
    1. Lokireddy S, Wijesoma IW, Teng S, Bonala S, Gluckman PD, McFarlane C, Sharma M, Kambadur R. The ubiquitin ligase Mul1 induces mitophagy in skeletal muscle in response to muscle-wasting stimuli. Cell Metab. 2012;16:613–624. doi: 10.1016/j.cmet.2012.10.005.
    1. Schiaffino S, Dyar KA, Ciciliot S, Blaauw B, Sandri M. Mechanisms regulating skeletal muscle growth and atrophy. FEBS J. 2013;280:4294–4314. doi: 10.1111/febs.12253.
    1. Sartori R, Schirwis E, Blaauw B, Bortolanza S, Zhao J, Enzo E, Stantzou A, Mouisel E, Toniolo L, Ferry A, Stricker S, Goldberg AL, Dupont S, Piccolo S, Amthor H, Sandri M. BMP signaling controls muscle mass. Nat Genet. 2013;45:1309–1318. doi: 10.1038/ng.2772.
    1. Percival JM, Anderson KN, Gregorevic P, Chamberlain JS, Froehner SC. Functional deficits in nNOSmu-deficient skeletal muscle: myopathy in nNOS knockout mice. PLoS One. 2008;3:e3387. doi: 10.1371/journal.pone.0003387.
    1. Froehner SC, Reed SM, Anderson KN, Huang PL, Percival JM: Loss of nNOS inhibits compensatory muscle hypertrophy and exacerbates inflammation and eccentric contraction-induced damage in mdx mice.Hum Mol Genet, in press.
    1. Pallafacchina G, Blaauw B, Schiaffino S. Role of satellite cells in muscle growth and maintenance of muscle mass. Nutr Metab Cardiovasc Dis. 2013;23(Suppl 1):S12–18. doi: 10.1016/j.numecd.2012.02.002.
    1. Sandri M, Barberi L, Bijlsma AY, Blaauw B, Dyar KA, Milan G, Mammucari C, Meskers CG, Pallafacchina G, Paoli A, Pion D, Roceri M, Romanello V, Serrano AL, Toniolo L, Larsson L, Maier AB, Munoz-Canoves P, Musaro A, Pende M, Reggiani C, Rizzuto R, Schiaffino S. Signalling pathways regulating muscle mass in ageing skeletal muscle: the role of the IGF1-Akt-mTOR-FoxO pathway. Biogerontology. 2013;14:303–323. doi: 10.1007/s10522-013-9432-9.
    1. Suzuki N, Motohashi N, Uezumi A, Fukada S, Yoshimura T, Itoyama Y, Aoki M, Miyagoe-Suzuki Y, Takeda S. NO production results in suspension-induced muscle atrophy through dislocation of neuronal NOS. J Clin Invest. 2007;117:2468–2476. doi: 10.1172/JCI30654.
    1. Clementi E, Brown GC, Foxwell N, Moncada S. On the mechanism by which vascular endothelial cells regulate their oxygen consumption. Proc Natl Acad Sci U S A. 1999;96:1559–1562. doi: 10.1073/pnas.96.4.1559.
    1. Nisoli E, Clementi E, Paolucci C, Cozzi V, Tonello C, Sciorati C, Bracale R, Valerio A, Francolini M, Moncada S, Carruba MO. Mitochondrial biogenesis in mammals: the role of endogenous nitric oxide. Science. 2003;299:896–899. doi: 10.1126/science.1079368.
    1. Gross SS, Wolin MS. Nitric oxide: pathophysiological mechanisms. Annu Rev Physiol. 1995;57:737–769. doi: 10.1146/annurev.ph.57.030195.003513.
    1. Pellegrino MW, Nargund AM, Haynes CM. Signaling the mitochondrial unfolded protein response. Biochim Biophys Acta. 1833;2013:410–416.
    1. Church JE, Gehrig SM, Chee A, Naim T, Trieu J, McConell GK, Lynch GS. Early functional muscle regeneration after myotoxic injury in mice is unaffected by nNOS absence. Am J Physiol Regul Integr Comp Physiol. 2011;301:R1358–1366. doi: 10.1152/ajpregu.00096.2011.
    1. Samengo G, Avik A, Fedor B, Whittaker D, Myung KH, Wehling-Henricks M, Tidball JG. Age-related loss of nitric oxide synthase in skeletal muscle causes reductions in calpain S-nitrosylation that increase myofibril degradation and sarcopenia. Aging Cell. 2012;11:1036–1045. doi: 10.1111/acel.12003.
    1. Ito N, Ruegg UT, Kudo A, Miyagoe-Suzuki Y, Takeda S. Activation of calcium signaling through Trpv1 by nNOS and peroxynitrite as a key trigger of skeletal muscle hypertrophy. Nat Med. 2013;19:101–106. doi: 10.1038/nm.3019.
    1. Li D, Yue Y, Lai Y, Hakim CH, Duan D. Nitrosative stress elicited by nNOSmicro delocalization inhibits muscle force in dystrophin-null mice. J Pathol. 2011;223:88–98. doi: 10.1002/path.2799.
    1. Nguyen HX, Tidball JG. Expression of a muscle-specific, nitric oxide synthase transgene prevents muscle membrane injury and reduces muscle inflammation during modified muscle use in mice. J Physiol. 2003;550:347–356. doi: 10.1113/jphysiol.2003.040907.
    1. Anderson JE. A role for nitric oxide in muscle repair: nitric oxide-mediated activation of muscle satellite cells. Mol Biol Cell. 2000;11:1859–1874. doi: 10.1091/mbc.11.5.1859.
    1. Baum O, Vieregge M, Koch P, Gul S, Hahn S, Huber-Abel FA, Pries AR, Hoppeler H. Phenotype of capillaries in skeletal muscle of nNOS-knockout mice. Am J Physiol Regul Integr Comp Physiol. 2013;304:R1175–1182. doi: 10.1152/ajpregu.00434.2012.
    1. Thomas GD. Functional muscle ischemia in Duchenne and Becker muscular dystrophy. Front Physiol. 2013;4:381. doi: 10.3389/fphys.2013.00381.
    1. Bredt DS. NO skeletal muscle derived relaxing factor in Duchenne muscular dystrophy. Proc Natl Acad Sci U S A. 1998;95:14592–14593. doi: 10.1073/pnas.95.25.14592.
    1. Thomas GD, Sander M, Lau KS, Huang PL, Stull JT, Victor RG. Impaired metabolic modulation of alpha-adrenergic vasoconstriction in dystrophin-deficient skeletal muscle. Proc Natl Acad Sci U S A. 1998;95:15090–15095. doi: 10.1073/pnas.95.25.15090.
    1. Shibata K, Shimokawa H, Yanagihara N, Otsuji Y, Tsutsui M. Nitric oxide synthases and heart failure - lessons from genetically manipulated mice. J UOEH. 2013;35:147–158. doi: 10.7888/juoeh.35.147.
    1. Lu XM, Zhang GX, Yu YQ, Kimura S, Nishiyama A, Matsuyoshi H, Shimizu J, Takaki M. The opposite roles of nNOS in cardiac ischemia-reperfusion-induced injury and in ischemia preconditioning-induced cardioprotection in mice. J Physiol Sci. 2009;59:253–262. doi: 10.1007/s12576-009-0030-1.
    1. Chao DS, Silvagno F, Bredt DS. Muscular dystrophy in mdx mice despite lack of neuronal nitric oxide synthase. J Neurochem. 1998;71:784–789. doi: 10.1046/j.1471-4159.1998.71020784.x.
    1. Crosbie RH, Straub V, Yun HY, Lee JC, Rafael JA, Chamberlain JS, Dawson VL, Dawson TM. Campbell KP: mdx muscle pathology is independent of nNOS perturbation. Hum Mol Genet. 1998;7:823–829. doi: 10.1093/hmg/7.5.823.

Source: PubMed

3
Iratkozz fel