RAGE in the pathophysiology of skeletal muscle

Francesca Riuzzi, Guglielmo Sorci, Roberta Sagheddu, Sara Chiappalupi, Laura Salvadori, Rosario Donato, Francesca Riuzzi, Guglielmo Sorci, Roberta Sagheddu, Sara Chiappalupi, Laura Salvadori, Rosario Donato

Abstract

Emerging evidence suggests that the signalling of the Receptor for Advanced Glycation End products (RAGE) is critical for skeletal muscle physiology controlling both the activity of muscle precursors during skeletal muscle development and the correct time of muscle regeneration after acute injury. On the other hand, the aberrant re-expression/activity of RAGE in adult skeletal muscle is a hallmark of muscle wasting that occurs in response to ageing, genetic disorders, inflammatory conditions, cancer, and metabolic alterations. In this review, we discuss the mechanisms of action and the ligands of RAGE involved in myoblast differentiation, muscle regeneration, and muscle pathological conditions. We highlight potential therapeutic strategies for targeting RAGE to improve skeletal muscle function.

Keywords: AGEs; HMGB1; RAGE; S100B; muscle wasting; myogenesis.

© 2018 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of the Society on Sarcopenia, Cachexia and Wasting Disorders.

Figures

Figure 1
Figure 1
Schematic representation of fl‐RAGE structure and its of ligands. The dominat negative isoform of RAGE (RAGEΔcyto) lacking of cytoplasmatic tail is reported.
Figure 2
Figure 2
Activation of RAGE signalling. Preassembly of RAGE into dimers and multimers is necessary for RAGE to bind ligands and activate downstream signalling pathways. Upon ligand binding, the RAGE's intracellular domain changes its conformation allowing the interaction with intracellular partners. RAGE signalling involves GTPases and kinases that activate transcription factors. RAGE activation leads to changes in gene expression and altered cellular function including migration, survival, cytokine production, inflammation, proliferation, differentation, and upregulation of RAGE expression itself.
Figure 3
Figure 3
Schematics of S100B/RAGE and HMGB1/RAGE interactions in low‐density (LD) and high‐density (HD) myoblasts. In LD myoblast cultures, interaction of S100B or HMBG1 with RAGE results in the simultaneous stimulation of proliferation and activation of the myogenic programme (left). In HD myoblast cultures, HMGB1‐activated RAGE promotes myoblast differentiation, apoptosis, and myocyte fusion into myofibres (right). HMGB1‐dependent RAGE activation induces p38 MAPK‐dependent expression of myogenin, which upregulates MyoD and represses PAX7 expression, leading to the reduction of proliferation and terminal differentiation. In HD condition, S100B binds bFGF, and the S100B/bFGF complex crosslinks RAGE and FGFR1 on apposed cells, blocking RAGE signalling and enhancing FGFR1 signalling.
Figure 4
Figure 4
Role of RAGE in acute or chronic muscle injury. Upon acute skeletal muscle injury, re‐expressed RAGE in activated satellite cells results in their proliferation and differentiation ultimately leading to regeneration. Under the action of S100B, RAGE signalling promotes proliferation and simultaneously activates the myogenic programme. RAGE activated by HMGB1 causes proliferation arrest and stimulates terminal differentiation of myoblasts into myocytes that build up new myofibres and/or repair damaged myofibres. RAGE‐mediated signalling in infiltrating macrophages modulates the inflammatory response in injured muscles, contributing significantly to accelerate regeneration. S100B/RAGE axis also promotes macrophage infiltration of acutely damaged skeletal muscle and macrophage polarization to M2 phenotype (left). On the contrary, in the course of muscle diseases characterized by chronic inflammation, high glucose conditions, or oxidative stress, an accumulation of RAGE ligands occurs in the serum and damaged myofibres. The overexpressed and overstimulated RAGE amplifies the inflammatory response by stimulating proinflammatory cytokines secretion, induces apoptosis of myocytes, and causes lethal accumulation of oxidative stress contributing to chronic myofibre damage (right).
Figure 5
Figure 5
Schematic representation of the effects of enforced expression of RAGE in TE671 ERMS cells. RAGE engaged by HMGB1 stimulates myogenic differentiation through activation of MKK6/p38 MAPK and myotube hypertrophy through PI3‐K/Akt. HMGB1/RAGE‐dependent activation of p38 MAPK also causes inactivation of ERK1/2 and JNK with consequent inhibition of proliferation and decrease in cell survival. Induction of RAGE in ERMS cells also results in the expression of myogenin and MyoD so as to cause repression of PAX7 expression and PAX7 proteasomal degradation, ultimately leading to reduction of proliferation, enhancement of apoptosis, and myogenic differentiation. Finally, the reduction of Pax7 expression by the RAGE/myogenin axis causes a reduction of cell migration and invasiveness in ERMS.

References

    1. Frontera WR, Ochala J. Skeletal muscle: a brief review of structure and function. Calcif Tissue Int 2015;96:183–195.
    1. Schnyder S, Handschin C. Skeletal muscle as an endocrine organ: PGC‐1α, myokines and exercise. Bone 2015;80:115–125.
    1. Bonaldo P, Sandri M. Cellular and molecular mechanisms of muscle atrophy. Dis Model Mech 2013;6:25–39.
    1. Schiaffino S, Reggiani C. Fiber types in mammalian skeletal muscles. Physiol Rev 2011;91:1447–1531.
    1. Neeper M, Schmidt AM, Brett J, Yan SD, Wang F, Pan YC, et al. Cloning and expression of a cell surface receptor for advanced glycosylation end products of proteins. J Biol Chem 1992;267:14998–15004.
    1. Bierhaus A, Humpert PM, Morcos M, Wendt T, Chavakis T, Arnold B, et al. Understanding RAGE, the receptor for advanced glycation end products. J Mol Med 2005;83:876–886.
    1. Xie J, Méndez JD, Méndez‐valenzuela V. Cellular signalling of the receptor for advanced glycation end products (RAGE). Cell Signal 2013;25:2185–2197.
    1. Riuzzi F, Sorci G, Donato R. The amphoterin (HMGB1)/Receptor for Advanced Glycation End Products (RAGE) pair modulates myoblast proliferation, apoptosis, adhesiveness, migration, and invasiveness: functional inactivation of RAGE in L6 myoblasts results in tumor formation in vivo. J Biol Chem 2006;281:8242–8253.
    1. Riuzzi F, Beccafico S, Sagheddu R, Chiappalupi S, Giambanco I, Bereshchenko O, et al. Levels of S100B protein drive the reparative process in acute muscle injury and muscular dystrophy. Sci Rep 2017;7:12537.
    1. Halaby DM, Mornon JPE. The immunoglobulin superfamily: an insight on its tissular, species, and functional diversity. J Mol Evol 1998;46:389–400.
    1. Hudson BI, Carter AM, Harja E, Harja E, Kalea AZ, Arriero M, et al. Identification, classification, and expression of RAGE gene splice variants. FASEB J 2008;22:1572–1580.
    1. Sugaya K, Fukagawa T, Matsumoto K, Mita K, Takahashi E, Ando A, et al. Three genes in the human MHC class III region near the junction with the class II: gene for receptor of advanced glycosylation end products, PBX2 homeobox gene and a notch homolog, human counterpart of mouse mammary tumor gene int‐3. Genomics 1994;23:408–419.
    1. Sessa L, Gatti E, Zeni F, Antonelli A, Catucci A, Koch M, et al. The receptor for advanced glycation end‐products (RAGE) is only present in mammals, and belongs to a family of cell adhesion molecules (CAMs). PLoS One 2014;9:e86903.
    1. Schmidt AM, Yan SD, Yan SF, Stern DM. The multiligand receptor RAGE as a progression factor amplifying immune and inflammatory responses. J Clin Invest 2001;108:949–955.
    1. Yatime L, Betzer C, Jensen RK, Mortensen S, Jensen PH, Andersen GR. The structure of the RAGE:S100A6 complex reveals a unique mode of homodimerization for S100 proteins. Structure 2016;24:2043–2052.
    1. Leclerc E, Fritz G, Weibel M, Heizmann CW, Galichet A. S100B and S100A6 differentially modulate cell survival by interacting with distinct RAGE (receptor for advanced glycation end products) immunoglobulin domains. J Biol Chem 2007;282:31317–31331.
    1. Turovskaya O, Foell D, Sinha P, Vogl T, Newlin R, Nayak J, et al. RAGE, carboxylated glycans and S100A8/A9 play essential roles in colitis‐associated carcinogenesis. Carcinogenesis 2008;29:2035–2043.
    1. Srikrishna G, Huttunen HJ, Johansson L, Weigle B, Yamaguchi Y, Rauvala H, et al. N ‐Glycans on the receptor for advanced glycation end products influence amphoterin binding and neurite outgrowth. J Neurochem 2002;80:998–1008.
    1. Lee EJ, Park JH. Receptor for Advanced Glycation Endproducts (RAGE), its ligands, and soluble RAGE: potential biomarkers for diagnosis and therapeutic targets for human renal diseases. Genomics Inform 2013;11:224.
    1. Hofmann MA, Drury S, Fu C, Qu W, Taguchi A, Lu Y, Avila C, Kambham N, Bierhaus A, Nawroth P, Neurath MF, Slattery T, Beach D, McClary J, Nagashima M, Morser J, Stern D, Schmidt AM. RAGE mediates a novel proinflammatory axis: a central cell surface receptor for S100/calgranulin polypeptides. Cell. 1999. 25;97:889–901
    1. Raucci A, Cugusi S, Antonelli A, Barabino SM, Monti L, Bierhaus A, et al. A soluble form of the receptor for advanced glycation endproducts (RAGE) is produced by proteolytic cleavage of the membrane‐bound form by the sheddase a disintegrin and metalloprotease 10 (ADAM10). FASEB J 2008;22:3716–3727.
    1. Schmidt AM. Isolation and characterization of two binding proteins for advanced glycosylation end products from bovine lung which are present on the endothelial cell surface. J Biol Chem 1992;267:14987–14997.
    1. Schmidt AM, Hori O, Cao R, Yan SD, Brett J, Wautier JL, et al. RAGE: a novel cellular receptor for advanced glycation end products. Diabetes 1996;45:S77–S80.
    1. Thornalley PJ, Battah S, Ahmed N, Karachalias N, Agalou S, Babaei‐Jadidi R, et al. Quantitative screening of advanced glycation endproducts in cellular and extracellular proteins by tandem mass spectrometry. Biochem J 2003;375:581–592.
    1. Xue J, Rai V, Singer D, Chabierski S, Xie J, Reverdatto S, et al. Advanced glycation end product recognition by the receptor for AGEs. Structure 2011;19:722–732.
    1. Xue J, Ray R, Singer D, Böhme D, Burz DS, Rai V, et al. The receptor for advanced glycation end products (RAGE) specifically recognizes methylglyoxal‐derived AGEs. Biochemistry 2014;53:3327–3335.
    1. Fatchiyah F, Hardiyanti F, Widodo N. Selective inhibition on RAGE‐binding AGEs required by bioactive peptide alpha‐S2 case in protein from goat Ethawah breed milk: study of biological modeling. Acta Inform Med 2015;23:90–96.
    1. Yan SD, Chen X, Fu J, Chen M, Zhu H, Roher A, et al. RAGE and amyloid‐beta peptide neurotoxicity in Alzheimer's disease. Nature 1996;382:685–691.
    1. Rouhiainen A, Kuja‐Panula J, Wilkman E, Pakkanen J, Stenfors J, Tuominen RK, et al. Regulation of monocyte migration by amphoterin (HMGB1). Blood 2004;104:1174–1182.
    1. Yamamoto Y, Yamamoto H. Controlling the receptor for advanced glycation end‐products to conquer diabetic vascular complications. J Diabetes Investig 2012;3:107–114.
    1. Hori O, Brett J, Slattery T, Cao R, Zhang J, Chen JX, et al. The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. Mediation of neurite outgrowth and co‐expression of rage and amphoterin in the developing nervous system. J Biol Chem 1995;270:25752–25761.
    1. Chavakis T, Bierhaus A, Al‐Fakhri N, Schneider D, Witte S, Linn T, et al. The pattern recognition receptor (RAGE) is a counterreceptor for leukocyte integrins: a novel pathway for inflammatory cell recruitment. J Exp Med 2003;198:1507–1515.
    1. Yamamoto Y, Harashima A, Saito H, Tsuneyama K, Munesue S, Motoyoshi S, et al. Septic shock is associated with receptor for advanced glycation end products ligation of LPS. J Immunol 2011;186:3248–3257.
    1. Sirois CM, Jin T, Miller AL, Bertheloot D, Nakamura H, Horvath GL, et al. RAGE is a nucleic acid receptor that promotes inflammatory responses to DNA. J Exp Med 2013;210:2447–2463.
    1. Sasaki N, Takeuchi M, Chowei H, Kikuchi S, Hayashi Y, Nakano N, et al. Advanced glycation end products (AGE) and their receptor (RAGE) in the brain of patients with Creutzfeldt–Jakob disease with prion plaques. Neurosci Lett 2002;326:117–120.
    1. Sorci G, Riuzzi F, Giambanco I, Donato R. RAGE in tissue homeostasis, repair and regeneration. Biochim Biophys Acta ‐ Mol Cell Res 2013;1833:101–109.
    1. Chavakis T, Bierhaus A, Nawroth PP. RAGE (receptor for advanced glycation end products): a central player in the inflammatory response. Microbes Infect 2004;6:1219–1225
    1. Kim S, Kim SY, Pribis JP, Lotze M, Mollen KP, Shapiro R, et al. Signaling of high mobility group box 1 (HMGB1) through toll‐like receptor 4 in macrophages requires CD14. Mol Med 2013;19:88–98.
    1. Brett J, Schmidt AM, Yan SD, Zou YS, Weidman E, Pinsky D, et al. Survey of the distribution of a newly characterized receptor for advanced glycation end products in tissues. Am J Pathol 1993;143:1699–1712.
    1. Sorci G, Giovannini G, Riuzzi F, Bonifazi P, Zelante T, Zagarella S, et al. The danger signal S100B integrates pathogen‐ and danger‐sensing pathways to restrain inflammation. PLoS Pathog 2011;7:e1001315.
    1. Fritz G. RAGE: a single receptor fits multiple ligands. Trends Biochem Sci 2011;36:625–632.
    1. Hudson BI, Lippman ME. Targeting RAGE signaling in inflammatory disease. Annu Rev Med 2018;69:349–364.
    1. Sakatani S, Yamada K, Homma C, Munesue S, Yamamoto Y, Yamamoto H, et al. Deletion of RAGE causes hyperactivity and increased sensitivity to auditory stimuli in mice. PLoS One 2009;4:e8309.
    1. Kierdorf K, Fritz G. RAGE regulation and signaling in inflammation and beyond. J Leukoc Biol 2013;94:55–68.
    1. Kang R, Tang D, Schapiro NE, Loux T, Livesey KM, Billiar TR, et al. The HMGB1/RAGE inflammatory pathway promotes pancreatic tumor growth by regulating mitochondrial bioenergetics. Oncogene 2014;33:567–577.
    1. Morales ME, Rojas RA, Monasterio AV, González BI, Figueroa CI, Manques MB, et al. Expression of RAGE in Helicobacter pylori infested gastric biopsies. Rev Med Chil 2013;141:1240–1248.
    1. Karimi J, Goodarzi MT, Tavilani H, Khodadadi I, Amiri I. Increased receptor for advanced glycation end products in spermatozoa of diabetic men and its association with sperm nuclear DNA fragmentation. Andrologia 2012;44:280–286.
    1. Stopper H, Schinzel R, Sebekova K, Heidland A. Genotoxicity of advanced glycation end products in mammalian cells. Cancer Lett 2003;190:151–156.
    1. Mallidis C, Agbaje IM, Rogers DA, Glenn JV, Pringle R, Atkinson AB, et al. Advanced glycation end products accumulate in the reproductive tract of men with diabetes. Int J Androl 2009;32:295–305.
    1. Kumar V, Fleming T, Terjung S, Gorzelanny C, Gebhardt C, Agrawal R, et al. Homeostatic nuclear RAGE–ATM interaction is essential for efficient DNA repair. Nucleic Acids Res 2017;45:10595–10613.
    1. Koch M, Chitayat S, Dattilo BM, Schiefner A, Diez J, Chazin WJ, et al. Structural basis for ligand recognition and activation of RAGE. Structure 2010;18:1342–1352.
    1. Yatime L, Andersen GR. Structural insights into the oligomerization mode of the human receptor for advanced glycation end‐products. FEBS J 2013;280:6556–6568.
    1. Zong H, Madden A, Ward M, Mooney MH, Elliott CT, Stitt AW. Homodimerization is essential for the receptor for advanced glycation end products (RAGE)‐mediated signal transduction. J Biol Chem 2010;285:23137–23146.
    1. Sakaguchi M, Murata H, Yamamoto K, Ono T, Sakaguchi Y, Motoyama A, et al. TIRAP, an adaptor protein for TLR2/4, transduces a signal from RAGE phosphorylated upon ligand binding. PLoS One 2011;6:e23132.
    1. Sakaguchi M, Kinoshita R, Putranto EW, Ruma IMW, Sumardika IW, Youyi C, et al. Signal diversity of receptor for advanced glycation end products. Acta Med Okayama 2017;71:459–465.
    1. Hudson BI, Kalea AZ, Del Mar Arriero M, Harja E, Boulanger E, D'Agati V, et al. Interaction of the RAGE cytoplasmic domain with diaphanous‐1 is required for ligand‐stimulated cellular migration through activation of Rac1 and Cdc42. J Biol Chem 2008;283:34457–34468.
    1. Bianchi R, Kastrisianaki E, Giambanco I, Donato R. S100B protein stimulates microglia migration via RAGE‐dependent up‐regulation of chemokine expression and release. J Biol Chem 2011;286:7214–7226.
    1. Yamamoto K, Murata H, Putranto EW, Kataoka K, Motoyama A, Hibino T, et al. DOCK7 is a critical regulator of the RAGE‐Cdc42 signaling axis that induces formation of dendritic pseudopodia in human cancer cells. Oncol Rep 2013;29:1073–1079.
    1. Sakaguchi M, Murata H, Aoyama Y, Hibino T, Putranto EW, Ruma IM, et al. DNAX‐activating protein 10 (DAP10) membrane adaptor associates with receptor for advanced glycation end products (RAGE) and modulates the RAGE‐triggered signaling pathway in human keratinocytes. J Biol Chem 2014;289:23389–23402.
    1. lowik A, Merres J, Elfgen A, Jansen S, Mohr F, Wruck CJ, et al. Involvement of formyl peptide receptors in receptor for advanced glycation end products (RAGE)‐and amyloid beta 1‐42‐induced signal transduction in glial cells. Mol Neurodegener 2012;7:55.
    1. Ichiki T, Koga T, Okuno T, Saeki K, Yamamoto Y, Yamamoto H, et al. Modulation of leukotriene B4 receptor 1 signaling by receptor for advanced glycation end products (RAGE). FASEB J 2016;30:1811–1822.
    1. Li JH, Wang W, Huang XR, Oldfield M, Schmidt AM, Cooper ME, et al. Advanced glycation end products induce tubular epithelial‐myofibroblast transition through the RAGE‐ERK1/2 MAP kinase signaling pathway. Am J Pathol 2004;164:1389–1397.
    1. Sorci G, Riuzzi F, Arcuri C, Giambanco I, Donato R. Amphoterin stimulates myogenesis and counteracts the antimyogenic factors basic fibroblast growth factor and S100B via RAGE binding. Mol Cell Biol 2004;24:4880–4894.
    1. Buckingham M, Bajard L, Chang T, Buckingham M, Bajard L, Chang T, et al. The formation of skeletal muscle: from somite to limb. J Anat 2003;202:59–68.
    1. Chargé SBP, Rudnicki MA. Cellular and molecular regulation of muscle regeneration. Physiol Rev 2004;84:209–238.
    1. Dumont NA, Bentzinger CF, Sincennes MC, Rudnicki MA. Satellite cells and skeletal muscle regeneration. Compr Physiol 2015;5:1027–1059.
    1. Hernández‐Hernández JM, García‐González EG, Brun CE, Rudnicki MA. The myogenic regulatory factors, determinants of muscle development, cell identity and regeneration. Semin Cell Dev Biol 2017;72:10–18.
    1. Zammit PS. Function of the myogenic regulatory factors Myf5, MyoD, Myogenin and MRF4 in skeletal muscle, satellite cells and regenerative myogenesis. Semin Cell Dev Biol 2017;72:19–32.
    1. Zammit PS. Pax7 and myogenic progression in skeletal muscle satellite cells. J Cell Sci 2006;119:1824–1832.
    1. Seale P, Sabourin LA, Girgis‐Gabardo A, Mansouri A, Gruss P, Rudnicki MA. Pax7 is required for the specification of myogenic satellite cells. Cell 2000;102:777–786.
    1. Olguin HC, Olwin BB. Pax‐7 up‐regulation inhibits myogenesis and cell cycle progression in satellite cells: a potential mechanism for self‐renewal. Dev Biol 2004;275:375–388.
    1. Zammit PS, Golding JP, Nagata Y, Hudon V, Partridge TA, Beauchamp JR. Muscle satellite cells adopt divergent fates: a mechanism for self‐renewal? J Cell Biol 2004;166:347–357.
    1. Chen JF, Tao Y, Li J, Deng Z, Yan Z, Xiao X, et al. microRNA‐1 and microRNA‐206 regulate skeletal muscle satellite cell proliferation and differentiation by repressing Pax7. J Cell Biol 2010;190:867–879.
    1. Dey BK, Gagan J, Dutta A. miR‐206 and ‐486 induce myoblast differentiation by downregulating Pax7. Mol Cell Biol 2011;31:203–214.
    1. Olguin HC, Yang Z, Tapscott SJ, Olwin BB. Reciprocal inhibition between Pax7 and muscle regulatory factors modulates myogenic cell fate determination. J Cell Biol 2007;177:769–779.
    1. Kuang S, Chargé SB, Seale P, Huh M, Rudnicki MA. Distinct roles for Pax7 and Pax3 in adult regenerative myogenesis. J Cell Biol 2006;172:103–113.
    1. Fidziańska A, Goebel HH. Human ontogenesis. 3. Cell death in fetal muscle. Acta Neuropathol 1991;81:572–577.
    1. Riuzzi F, Sorci G, Sagheddu R, Donato R. HMGB1‐RAGE regulates muscle satellite cell homeostasis through p38‐MAPK‐ and myogenin‐dependent repression of Pax7 transcription. J Cell Sci 2012;125:1440–1454.
    1. Riuzzi F, Sorci G, Beccafico S, Donato R. S100B engages RAGE or bFGF/FGFR1 in myoblasts depending on its own concentration and myoblast density. Implications for muscle regeneration. PLoS One 2012;7:e28700.
    1. Lee J, Hong F, Kwon S, Kim SS, Kim DO, Kang HS, et al. Activation of p38 MAPK induces cell cycle arrest via inhibition of Raf/ERK pathway during muscle differentiation. Biochem Biophys Res Commun 2002;298:765–771.
    1. Chen X, Li Y. Role of matrix metalloproteinases in skeletal muscle: migration, differentiation, regeneration and fibrosis. Cell Adh Migr 2009;3:337–341.
    1. Sorci G, Riuzzi F, Arcuri C, Bianchi R, Brozzi F, Tubaro C, et al. The many faces of S100B protein: when an extracellular factor inactivates its own receptor and activates another one. Ital J Anat Embryol 2010;115:147–151.
    1. Dattilo BM, Fritz G, Leclerc E, Vander KCW, Heizmann CW, Chazin WJ. The extracellular region of the receptor for advanced glycation end products is composed of two independent structural units. Biochemistry 2007;46:6957–6970.
    1. Charlton CA, Mohler WA, Blau HM. Neural cell adhesion molecule (NCAM) and myoblast fusion. Dev Biol 2000;221:112–119.
    1. Lyons GE, Moore R, Yahara O, Buckingham ME, Walsh FS. Expression of NCAM isoforms during skeletal myogenesis in the mouse embryo. Dev Dyn 1992;194:94–104.
    1. Dormoy‐Raclet V, Cammas A, Celona B, Lian XJ, van der Giessen K, Zivojnovic M, et al. HuR and miR‐1192 regulate myogenesis by modulating the translation of HMGB1 mRNA. Nat Commun 2013;4:2388.
    1. Rigamonti E, Zordan P, Sciorati C, Rovere‐Querini P, Brunelli S. Macrophage plasticity in skeletal muscle repair. Biomed Res Int 2014;2014:560629.
    1. Novak ML, Koh TJ. Phenotypic transitions of macrophages orchestrate tissue repair. Am J Pathol 2013;183:1352–1363.
    1. Zhang C, Wang C, Li Y, Miwa T, Liu C, Cui W, Song WC, Du J. Complement C3a signaling facilitates skeletal muscle regeneration by regulating monocyte function and trafficking. Nat Commun 2017;8:2078
    1. De Mori R, Straino S, Di Carlo A, Mangoni A, Pompilio G, Palumbo R, et al. Multiple effects of high mobility group box protein 1 in skeletal muscle regeneration. Arterioscler Thromb Vasc Biol 2007;27:2377–2383.
    1. Sachdev U, Cui X, Tzeng E. HMGB1 and TLR4 mediate skeletal muscle recovery in a murine model of hindlimb ischemia. J Vasc Surg 2013;58:460–469.
    1. Schiraldi M, Raucci A, Muñoz LM, Livoti E, Celona B, Venereau E, et al. HMGB1 promotes recruitment of inflammatory cells to damaged tissues by forming a complex with CXCL12 and signaling via CXCR4. J Exp Med 2012;209:551–563.
    1. Tirone M, Tran NL, Ceriotti C, Gorzanelli A, Canepari M, Bottinelli R, et al. High mobility group box 1 orchestrates tissue regeneration via CXCR4. J Exp Med 2018;215:303–318.
    1. Dutt V, Gupta S, Dabur R, Injeti E, Mittal A. Skeletal muscle atrophy: potential therapeutic agents and their mechanisms of action. Pharmacol Res 2015;99:86–100.
    1. Sandri M. Protein breakdown in muscle wasting: role of autophagy‐lysosome and ubiquitin‐proteasome. Int J Biochem Cell Biol 2013;45:2121–2129.
    1. Glass DJ. Skeletal muscle hypertrophy and atrophy signaling pathways. Int J Biochem Cell Biol 2005;37:1974–1984.
    1. Fukada SI. The roles of muscle stem cells in muscle injury, atrophy and hypertrophy. J Biochem 2018;163:353–358.
    1. McKenna CF, Fry CS. Altered satellite cell dynamics accompany skeletal muscle atrophy during chronic illness, disuse, and aging. Curr Opin Clin Nutr Metab Care 2017;20:447–452.
    1. He WA, Berardi E, Cardillo VM, Acharyya S, Aulino P, Thomas‐Ahner J, et al. NF‐κB‐mediated Pax7 dysregulation in the muscle microenvironment promotes cancer cachexia. J Clin Invest 2013;123:4821–4835.
    1. Latres E, Amini AR, Amini AA, Griffiths J, Martin FJ, Wei Y, 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. J Biol Chem 2005;280:2737–2744.
    1. Cohen S, Brault JJ, Gygi SP, Glass DJ, Valenzuela DM, Gartner C, et al. During muscle atrophy, thick, but not thin, filament components are degraded by MuRF1‐dependent ubiquitylation. J Cell Biol 2009;185:1083–1095.
    1. Milan G, Romanello V, Pescatore F, Armani A, Paik JH, Frasson L, et al. Regulation of autophagy and the ubiquitin‐proteasome system by the FoxO transcriptional network during muscle atrophy. Nat Commun 2015;6:6670.
    1. Li YP, Schwartz RJ, Waddell ID, Holloway BR, Reid MB. Skeletal muscle myocytes undergo protein loss and reactive oxygen‐mediated NF‐kappaB activation in response to tumor necrosis factor alpha. FASEB J 1998;12:871–880.
    1. Ding H, Zhang G, Sin KW, Liu Z, Lin RK, Li M, et al. Activin A induces skeletal muscle catabolism via p38β mitogen‐activated protein kinase. J Cachexia Sarcopenia Muscle 2017;8:202–212.
    1. Clavel S, Siffroi‐Fernandez S, Coldefy AS, Boulukos K, Pisani DF, Dérijard B. Regulation of the intracellular localization of Foxo3a by stress‐activated protein kinase signaling pathways in skeletal muscle cells. Mol Cell Biol 2010;30:470–480.
    1. Andersen H, Schmitz O, Nielsen S. Decreased isometric muscle strength after acute hyperglycaemia in type 1 diabetic patients. Diabet Med 2005;22:1401–1407.
    1. Umegaki H. Sarcopenia and frailty in older patients with diabetes mellitus. Geriatr Gerontol Int 2016;16:293–299.
    1. Aguiari P, Leo S, Zavan B, Vindigni V, Rimessi A, Bianchi K, et al. High glucose induces adipogenic differentiation of muscle‐derived stem cells. Proc Natl Acad Sci U S A 2008;105:1226–1231.
    1. Nguyen MH, Cheng M, Koh TJ. Impaired muscle regeneration in ob/ob and db/db mice. ScientificWorldJournal 2011;11:1525–1535.
    1. Snow LM, Fugere NA, Thompson LV. Advanced glycation end‐product accumulation and associated protein modification in type II skeletal muscle with aging. J Gerontol A Biol Sci Med Sci 2007;62:1204–1210.
    1. Snow LM, Thompson LV. Influence of insulin and muscle fiber type in nepsilon‐(carboxymethyl)‐lysine accumulation in soleus muscle of rats with streptozotocin‐induced diabetes mellitus. Pathobiology 2009;76:227–234.
    1. Chiu CY, Yang RS, Sheu ML, Chan DC, Yang TH, Tsai KS, et al. Advanced glycation end‐products induce skeletal muscle atrophy and dysfunction in diabetic mice via a RAGE‐mediated, AMPK‐down‐regulated, Akt pathway. J Pathol 2016;238:470–482.
    1. Howard AC, McNeil AK, Xiong F, Xiong WC, McNeil PL. A novel cellular defect in diabetes: membrane repair failure. Diabetes 2011;60:3034–3043.
    1. Nagai R, Murray DB, Metz TO, Baynes JW. Chelation: a fundamental mechanism of action of AGE inhibitors, AGE breakers, and other inhibitors of diabetes complications. Diabetes 2012;61:549–559.
    1. Boden G. Obesity, insulin resistance and free fatty acids. Curr Opin Endocrinol Diabetes Obes 2011;18:139–143.
    1. Samuel VT, Shulman GI. Mechanisms for insulin resistance: common threads and missing links. Cell 2012;148:852–871.
    1. Guo Q, Mori T, Jiang Y, Hu C, Osaki Y, Yoneki Y, et al. Methylglyoxal contributes to the development of insulin resistance and salt sensitivity in Sprague‐Dawley rats. J Hypertens 2009;27:1664–1671.
    1. Wang X, Jia X, Chang T, Desai K, Wu L. Attenuation of hypertension development by scavenging methylglyoxal in fructose‐treated rats. J Hypertens 2008;26:765–772.
    1. Li W, Xu H, Hu Y, He P, Ni Z, Xu H, et al. Edaravone protected human brain microvascular endothelial cells from methylglyoxal‐induced injury by inhibiting AGEs/RAGE/oxidative stress. PLoS One 2013;8:e76025.
    1. Berlanga J, Cibrian D, Guillén I, Freyre F, Alba JS, Lopez‐Saura P, et al. Methylglyoxal administration induces diabetes‐like microvascular changes and perturbs the healing process of cutaneous wounds. Clin Sci (Lond) 2005;109:83–95.
    1. Yao D, Brownlee M. Hyperglycemia‐induced reactive oxygen species increase expression of the receptor for advanced glycation end products (RAGE) and RAGE ligands. Diabetes 2010;59:249–255.
    1. Ma H, Li SY, Xu P, Babcock SA, Dolence EK, Brownlee M, et al. Advanced glycation endproduct (AGE) accumulation and AGE receptor (RAGE) up‐regulation contribute to the onset of diabetic cardiomyopathy. J Cell Mol Med 2009;13:1751–1764.
    1. Bourajjaj M, Stehouwer CD, van Hinsbergh VW, Schalkwijk CG. Role of methylglyoxal adducts in the development of vascular complications in diabetes mellitus. Biochem Soc Trans 2003;31:1400–1402.
    1. Uribarri J, Cai W, Ramdas M, Goodman S, Pyzik R, Chen X, et al. Restriction of advanced glycation end products improves insulin resistance in human type 2 diabetes: potential role of AGER1 and SIRT1. Diabetes Care 2011;34:1610–1616.
    1. Aftab MF, Afridi SK, Ghaffar S, Murtaza M, Khan M, Karim A, Khan KM, Waraich RS. A bis‐Schiff base of isatin improves methylglyoxal mediated insulin resistance in skeletal muscle cells. Arch Pharm Res. 2015;Oct 30.
    1. Cassese A, Esposito I, Fiory F, Barbagallo AP, Paturzo F, Mirra P, et al. In skeletal muscle advanced glycation end products (AGEs) inhibit insulin action and induce the formation of multimolecular complexes including the receptor for AGEs. J Biol Chem 2008;283:36088–36099.
    1. He C, Sabol J, Mitsuhashi T, Vlassara H. Dietary glycotoxins: inhibition of reactive products by aminoguanidine facilitates renal clearance and reduces tissue sequestration. Diabetes 1999;48:1308–1315.
    1. Hofmann SM, Dong HJ, Li Z, Cai W, Altomonte J, Thung SN, et al. Improved insulin sensitivity is associated with restricted intake of dietary glycoxidation products in the db/db mouse. Diabetes 2002;51:2082–2089.
    1. Vlassara H. The AGE‐receptor in the pathogenesis of diabetic complications. Diabetes Metab Res Rev 2001;17:436–443.
    1. Miele C, Riboulet A, Maitan MA, Oriente F, Romano C, Formisano P, et al. Human glycated albumin affects glucose metabolism in L6 skeletal muscle cells by impairing insulin‐induced insulin receptor substrate (IRS) signaling through a protein kinase C alpha‐mediated mechanism. J Biol Chem 2003;278:47376–47387.
    1. de la Maza MP, Uribarri J, Olivares D, Hirsch S, Leiva L, Barrera G, et al. Weight increase is associated with skeletal muscle immunostaining for advanced glycation end products, receptor for advanced glycation end products, and oxidation injury. Rejuvenation Res 2008;11:1041–1048.
    1. Melton LJ 3rd, Khosla S, Riggs BL. Epidemiology of sarcopenia. Mayo Clin Proc 2000; Suppl:S10‐2; discussion S12–3.
    1. Sakuma K, Aoi W, Yamaguchi A. Molecular mechanism of sarcopenia and cachexia: recent research advances. Pflugers Arch 2017;469:573–591.
    1. Walston JD. Sarcopenia in older adults. Curr Opin Rheumatol 2012;24:623–627.
    1. Bernet JD, Doles JD, Hall JK, Kelly Tanaka K, Carter TA, Olwin BB. p38 MAPK signaling underlies a cell‐autonomous loss of stem cell self‐renewal in skeletal muscle of aged mice. Nat Med 2014;20:265–271.
    1. Brack AS, Rando TA. Intrinsic changes and extrinsic influences of myogenic stem cell function during aging. Stem Cell Rev 2007;3:226–237.
    1. Cosgrove BD, Gilbert PM, Porpiglia E, Mourkioti F, Lee SP, Corbel SY, et al. Rejuvenation of the muscle stem cell population restores strength to injured aged muscles. Nat Med 2014;20:255–264.
    1. Sousa‐Victor P, Gutarra S, García‐Prat L, Rodriguez‐Ubreva J, Ortet L, Ruiz‐Bonilla V, et al. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature 2014;506:316–321.
    1. Beccafico S, Riuzzi F, Puglielli C, Mancinelli R, Fulle S, Sorci G, et al. Human muscle satellite cells show age‐related differential expression of S100B protein and RAGE. Age (Dordr) 2011;33:523–541.
    1. Tubaro C, Arcuri C, Giambanco I, Donato R. S100B protein in myoblasts modulates myogenic differentiation via NF‐kappaB‐dependent inhibition of MyoD expression. J Cell Physiol 2010;223:270–282.
    1. Morozzi G, Beccafico S, Bianchi R, Riuzzi F, Bellezza I, Giambanco I, et al. Oxidative stress‐induced S100B accumulation converts myoblasts into brown adipocytes via an NF‐κB/YY1/miR‐133 axis and NF‐κB/YY1/BMP‐7 axis. Cell Death Differ 2017;24:2077–2088.
    1. Grillari J, Katinger H, Voglauer R. Aging and the ubiquitinome: traditional and non‐traditional functions of ubiquitin in aging cells and tissues. Exp Gerontol 2006;41:1067–1079.
    1. Dalal M, Ferrucci L, Sun K, Beck J, Fried LP, Semba RD. Elevated serum advanced glycation end products and poor grip strength in older community‐dwelling women. J Gerontol A Biol Sci Med Sci 2009;64:132–137.
    1. Haus JM, Carrithers JA, Trappe SW, Trappe TA. Collagen, cross‐linking, and advanced glycation end products in aging human skeletal muscle. J Appl Physiol 2007;103:2068–2076.
    1. Semba RD, Nicklett EJ, Ferrucci L. Does accumulation of advanced glycation end products contribute to the aging phenotype? J Gerontol A Biol Sci Med Sci 2010;65:963–975.
    1. Finsterer J, Löscher WN, Wanschitz J, Quasthoff S, Grisold W. Secondary myopathy due to systemic diseases. Acta Neurol Scand 2016;134:388–402.
    1. Pfeffer G, Chinnery PF. Diagnosis and treatment of mitochondrial myopathies. Ann Med 2013;45:4–16.
    1. Mercuri E, Muntoni F. Muscular dystrophies. Lancet 2013;381:845–860.
    1. Haslbeck KM, Friess U, Schleicher ED, Bierhaus A, Nawroth PP, Kirchner A, et al. The RAGE pathway in inflammatory myopathies and limb girdle muscular dystrophy. Acta Neuropathol 2005;110:247–254.
    1. Macaione V, Aguennouz M, Rodolico C, Mazzeo A, Patti A, Cannistraci E, et al. RAGE‐NF‐kappaB pathway activation in response to oxidative stress in facioscapulohumeral muscular dystrophy. Acta Neurol Scand 2007;115:115–121.
    1. Wautier MP, Chappey O, Corda S, Stern DM, Schmidt AM, Wautier JL. Activation of NADPH oxidase by AGE links oxidant stress to altered gene expression via RAGE. Am J Physiol Endocrinol Metab 2001;280:E685–E694.
    1. Winokur ST, Barrett K, Martin JH, Forrester JR, Simon M, Tawil R, et al. Facioscapulohumeral muscular dystrophy (FSHD) myoblasts demonstrate increased susceptibility to oxidative stress. Neuromuscul Disord 2003;13:322–333.
    1. Mandel DE, Malemud CJ, Askari AD. Idiopathic inflammatory myopathies: a review of the classification and impact of pathogenesis. Int J Mol Sci 2017;18:1084.
    1. Ulfgren AK, Grundtman C, Borg K, Alexanderson H, Andersson U, Harris HE, et al. Down‐regulation of the aberrant expression of the inflammation mediator high mobility group box chromosomal protein 1 in muscle tissue of patients with polymyositis and dermatomyositis treated with corticosteroids. Arthritis Rheum 2004;50:1586–1594.
    1. Askanas V, Engel WK, Nogalska A. Pathogenic considerations in sporadic inclusion‐body myositis, a degenerative muscle disease associated with aging and abnormalities of myoproteostasis. J Neuropathol Exp Neurol 2012;71:680–693.
    1. Muth IE, Zschüntzsch J, Kleinschnitz K, Wrede A, Gerhardt E, Balcarek P, et al. HMGB1 and RAGE in skeletal muscle inflammation: implications for protein accumulation in inclusion body myositis. Exp Neurol 2015;271:189–197.
    1. Yan SS, Chen D, Yan S, Guo L, Du H, Chen JX. RAGE is a key cellular target for Abeta‐induced perturbation in Alzheimer's disease. Front Biosci (Schol Ed) 2012;4:240–250.
    1. Rüster M, Franke S, Späth M, Pongratz DE, Stein G, Hein GE. Detection of elevated N epsilon‐carboxymethyllysine levels in muscular tissue and in serum of patients with fibromyalgia. Scand J Rheumatol 2005;34:460–463.
    1. Phillips WD, Vincent A. Pathogenesis of myasthenia gravis: update on disease types, models, and mechanisms. F1000Res 2016;5:1513.
    1. Uzawa A, Kanai T, Kawaguchi N, Oda F, Himuro K, Kuwabara S. Changes in inflammatory cytokine networks in myasthenia gravis. Sci Rep 2016;6:25886.
    1. Molin CJ, Westerberg E, Punga AR. Profile of upregulated inflammatory proteins in sera of Myasthenia gravis patients. Sci Rep 2017;7:39716.
    1. Donato R, Cannon BR, Sorci G, Riuzzi F, Hsu K, Weber DJ, et al. Functions of S100 proteins. Curr Mol Med 2013;13:24–57.
    1. Luo Y, Yoneda J, Ohmori H, Sasaki T, Shimbo K, Eto S, et al. Cancer usurps skeletal muscle as an energy repository. Cancer Res 2014;74:330–340.
    1. Pappo AS, Dirksen U. Rhabdomyosarcoma, Ewing sarcoma, and other round cell sarcomas. J Clin Oncol 2018;36:168–179.
    1. Saab R, Spunt SL, Skapek SX. Myogenesis and rhabdomyosarcoma the Jekyll and Hyde of skeletal muscle. Curr Top Dev Biol 2011;94:197–234.
    1. Hettmer S, Wagers AJ. Muscling in: uncovering the origins of rhabdomyosarcoma. Nat Med 2010;16:171–173.
    1. Calhabeu F, Hayashi S, Morgan JE, Relaix F, Zammit PS. Alveolar rhabdomyosarcoma‐associated proteins PAX3/FOXO1A and PAX7/FOXO1A suppress the transcriptional activity of MyoD‐target genes in muscle stem cells. Oncogene 2013;32:651–662.
    1. Blake J, Ziman MR. Aberrant PAX3 and PAX7 expression. A link to the metastatic potential of embryonal rhabdomyosarcoma and cutaneous malignant melanoma? Histol Histopathol 2003;18:529–539.
    1. Chiappalupi S, Riuzzi F, Fulle S, Donato R, Sorci G. Defective RAGE activity in embryonal rhabdomyosarcoma cells results in high PAX7 levels that sustain migration and invasiveness. Carcinogenesis 2014;35:2382–2392.
    1. Riuzzi F, Sorci G, Sagheddu R, Sidoni A, Alaggio R, Ninfo V, et al. RAGE signaling deficiency in rhabdomyosarcoma cells causes upregulation of PAX7 and uncontrolled proliferation. J Cell Sci 2014;127:1699–1711.
    1. Riuzzi F, Sorci G, Donato R. RAGE expression in rhabdomyosarcoma cells results in myogenic differentiation and reduced proliferation, migration, invasiveness, and tumor growth. Am J Pathol 2007;171:947–961.
    1. Wegorzewska M, Krauss RS, Kang JS. Overexpression of the immunoglobulin superfamily members CDO and BOC enhances differentiation of the human rhabdomyosarcoma cell line RD. Mol Carcinog 2003;37:1–4.
    1. Sparvero LJ, Asafu‐Adjei D, Kang R, Tang D, Amin N, Im J, et al. RAGE (receptor for advanced glycation endproducts), RAGE ligands, and their role in cancer and inflammation. J Transl Med 2009;7:17.
    1. Bartling B, Hofmann HS, Weigle B, Silber RE, Simm A. Down‐regulation of the receptor for advanced glycation end‐products (RAGE) supports non‐small cell lung carcinoma. Carcinogenesis 2005;26:293–301.
    1. Tateno T, Ueno S, Hiwatashi K, Matsumoto M, Okumura H, Setoyama T, et al. Expression of receptor for advanced glycation end products (RAGE) is related to prognosis in patients with esophageal squamous cell carcinoma. Ann Surg Oncol 2009;16:440–446.
    1. Bongarzone S, Savickas V, Luzi F, Gee AD. Targeting the receptor for advanced glycation endproducts (RAGE): a medicinal chemistry perspective. J Med Chem 2017;60:7213–7232.
    1. Sabbagh MN, Agro A, Bell J, Aisen PS, Schweizer E, Galasko D. PF‐04494700, an oral inhibitor of receptor for advanced glycation end products (RAGE), in Alzheimer disease. Alzheimer Dis Assoc Disord 2011;25:206–212.
    1. Galasko D, Bell J, Mancuso JY, Kupiec JW, Sabbagh MN, van Dyck C, Thomas RG, Aisen PS; Alzheimer's Disease Cooperative Study. Clinical trial of an inhibitor of RAGE–Aβ interactions in Alzheimer disease. Neurology 2014;82:1536–1542
    1. Burstein AH, Grimes I, Galasko DR, Aisen PS, Sabbagh M, Mjalli AM. Effect of TTP488 in patients with mild to moderate Alzheimer's disease. BMC Neurol 2014;14:12.
    1. Han YT, Choi GI, Son D, Kim NJ, Yun H, Lee S, et al. Ligand‐based design, synthesis, and biological evaluation of 2‐aminopyrimidines, a novel series of receptor for advanced glycation end products (RAGE) inhibitors. J Med Chem 2012;55:9120–9135.
    1. Lee YS, Kim H, Kim YH, Roh EJ, Han H, Shin KJ. Synthesis and structure‐activity relationships of tri‐substituted thiazoles as RAGE antagonists for the treatment of Alzheimer's disease. Bioorg Med Chem Lett 2012;22:7555–7561.
    1. Deane R, Singh I, Sagare AP, Bell RD, Ross NT, LaRue B, et al. A multimodal RAGE‐specific inhibitor reduces amyloid β‐mediated brain disorder in a mouse model of Alzheimer disease. J Clin Invest 2012;122:1377–1392.
    1. Hong Y, Shen C, Yin Q, Sun M, Ma Y, Liu X. Effects of RAGE‐specific inhibitor FPS‐ZM1 on Amyloid‐β metabolism and AGEs‐induced inflammation and oxidative stress in rat hippocampus. Neurochem Res 2016;41:1192–1199.
    1. Wang D, Liu K, Wake H, Teshigawara K, Mori S, Nishibori M. Anti‐high mobility group box‐1 (HMGB1) antibody inhibits hemorrhage‐induced brain injury and improved neurological deficits in rats. Sci Rep 2017;7:46243.
    1. Manigrasso MB, Pan J, Rai V, Zhang J, Reverdatto S, Quadri N, et al. Small molecule inhibition of ligand‐stimulated RAGE‐DIAPH1 signal transduction. Sci Rep 2016;6:22450.
    1. López‐Díez R, Rastrojo A, Villate O, Aguado B. Complex tissue‐specific patterns and distribution of multiple RAGE splice variants in different mammals. Genome Biol Evol 2013;5:2420–2435.
    1. Ding Q, Keller JN. Splice variants of the receptor for advanced glycosylation end products (RAGE) in human brain. Neurosci Lett 2005;373:67–72.
    1. Chuah YK, Basir R, Talib H, Tie TH, Nordin N. Receptor for advanced glycation end products and its involvement in inflammatory diseases. Int J Inflam 2013;2013:403460.
    1. Riehl A, Németh J, Angel P, Hess J. The receptor RAGE: bridging inflammation and cancer. Cell Commun Signal 2009;7:12.
    1. Bierhaus A, Stern DM, Nawroth PP. RAGE in inflammation: a new therapeutic target? Curr Opin Investig Drugs 2006;7:985–991.
    1. Kokkola R, Andersson A, Mullins G, Ostberg T, Treutiger CJ, Arnold B, et al. RAGE is the major receptor for the proinflammatory activity of HMGB1 in rodent macrophages. Scand J Immunol 2005;61:1–9.
    1. Donato R, Sorci G, Riuzzi F, Arcuri C, Bianchi R, Brozzi F, et al. S100B's double life: intracellular regulator and extracellular signal. Biochim Biophys Acta 2009;1793:1008–1022.
    1. Huttunen HJ, Kuja‐Panula J, Sorci G, Agneletti AL, Donato R, Rauvala H. Coregulation of neurite outgrowth and cell survival by amphoterin and S100 proteins through receptor for advanced glycation end products (RAGE) activation. J Biol Chem 2000;275:40096–40105.
    1. Businaro R, Leone S, Fabrizi C, Sorci G, Donato R, Lauro GM, et al. S100B protects LAN‐5 neuroblastoma cells against Abeta amyloid‐induced neurotoxicity via RAGE engagement at low doses but increases Abeta amyloid neurotoxicity at high doses. J Neurosci Res 2006;83:897–906.
    1. Jin X, Yao T, Zhou Z, Zhu J, Zhang S, Hu W, et al. Advanced glycation end products enhance macrophages polarization into M1 phenotype through activating RAGE/NF‐κB pathway. Biomed Res Int 2015;732450.
    1. Malik P, Chaudhry N, Mittal R, Mukherjee TK. Role of receptor for advanced glycation end products in the complication and progression of various types of cancers. Biochim Biophys Acta 2015;1850:1898–1904.
    1. Ahmad S, Khan H, Siddiqui Z, Khan MY, Rehman S, Shahab U, et al. AGEs, RAGEs and s‐RAGE; friend or foe for cancer. Semin Cancer Biol 2018;49:44–55.
    1. Logsdon CD, Fuentes MK, Huang EH, Arumugam T. RAGE and RAGE ligands in cancer. Curr Mol Med 2007;7:777–789.
    1. Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol 2010;28:367–388.
    1. Yu YX, Pan WC, Cheng YF. Silencing of advanced glycosylation and glycosylation and product‐specific receptor (RAGE) inhibits the metastasis and growth of non‐small cell lung cancer. Am J Transl Res 2017;9:2760–2774.
    1. Landesberg R, Woo V, Huang L, Cozin M, Lu Y, Bailey C, et al. The expression of the receptor for glycation endproducts (RAGE) in oral squamous cell carcinomas. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2008;105:617–624.
    1. von Haehling S, Morley JE, Coats AJS, Anker SD. Ethical guidelines for publishing in the Journal of Cachexia, Sarcopenia and Muscle: update 2017. J Cachexia Sarcopenia Muscle 2017;8:1081–1083.

Source: PubMed

3
Abonner