Skip to main content
Log in

Expression of the Mas receptor is upregulated in skeletal muscle wasting

  • Original Paper
  • Published:
Histochemistry and Cell Biology Aims and scope Submit manuscript

Abstract

Skeletal muscle atrophy during sepsis, immobilization, and chronic diseases is characterized by an increase in expression and activity of the muscle-specific ubiquitin 3 ligases atrogin-1 and MuRF-1. The classical renin–angiotensin system (RAS), by high level of circulating angiotensin II (AngII) is directly involved in skeletal muscle wasting associated with cardiac and renal failure. Ang (1–7), a peptide belonging to the non-classical RAS system, produces effects that are opposite to AngII. The actions of Ang (1–7) are mediated by its binding and signalling through the Mas receptor. Our purpose is to assess the effects of atrophic stimuli AngII, lipopolysaccharide (LPS), and immobilization on the expression of the Mas receptor in skeletal muscle. For that we used gastrocnemius and tibialis anterior muscles of C57BL10 mice treated with AngII, LPS or subjected to unilateral hindlimb immobilization by casting. In addition, we used C2C12 myotubes incubated with AngII or LPS. We evaluated Mas expression by quantitative real-time PCR, Western blot immunohistochemical analysis. Skeletal muscle atrophy was corroborated by the expression of atrogin-1 and MuRF-1 and the fibre diameter. Our results show that Mas receptor expression was increased by AngII or LPS in vitro and in vivo, and upregulated by immobilization. The increase of the Mas expression was concomitantly with the upregulation of atrogin-1 and MuRF-1 and the reduction of the fibre diameter. These results from studies in vitro and in vivo demonstrate for the first time that the Mas receptor is increased under atrophic stimulus and suggest that the non-classical RAS system could have an important role in muscle wasting.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  • Acuna MJ et al (2014) Restoration of muscle strength in dystrophic muscle by angiotensin-1–7 through inhibition of TGF-beta signalling. Hum Mol Genet 23:1237–1249

    Article  CAS  PubMed  Google Scholar 

  • Agarwal R (2003) Proinflammatory effects of oxidative stress in chronic kidney disease: role of additional angiotensin II blockade. Am J Physiol Renal Physiol 284:F863–F869

    CAS  PubMed  Google Scholar 

  • Agarwal R, Campbell RC, Warnock DG (2004) Oxidative stress in hypertension and chronic kidney disease: role of angiotensin II. Semin Nephrol 24:101–114

    Article  CAS  PubMed  Google Scholar 

  • Antoniu SA (2008) Targeting the angiotensin pathway in idiopathic pulmonary fibrosis. Expert Opin Ther Targets 12:1587–1590

    Article  CAS  PubMed  Google Scholar 

  • Benter IF, Ferrario CM, Morris M, Diz DI (1995) Antihypertensive actions of angiotensin-(1–7) in spontaneously hypertensive rats. Am J Physiol 269:H313–H319

    CAS  PubMed  Google Scholar 

  • Bodine SC et al (2001) Identification of ubiquitin ligases required for skeletal muscle atrophy. Science 294:1704–1708

    Article  CAS  PubMed  Google Scholar 

  • Borge BA, Kalland KH, Olsen S, Bletsa A, Berggreen E, Wiig H (2009) Cytokines are produced locally by myocytes in rat skeletal muscle during endotoxemia. Am J Physiol Heart Circ Physiol 296:H735–H744

    Article  CAS  PubMed  Google Scholar 

  • Brecher P (1996) Angiotensin II and cardiac fibrosis. Trends Cardiovasc Med 6:193–198

    Article  CAS  PubMed  Google Scholar 

  • Bruells CS et al (2013) Prolonged mechanical ventilation alters the expression pattern of angio-neogenetic factors in a pre-clinical rat model. PLoS ONE 8:e70524

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Cabello-Verrugio C, Acuna MJ, Morales MG, Becerra A, Simon F, Brandan E (2011) Fibrotic response induced by angiotensin-II requires NAD(P)H oxidase-induced reactive oxygen species (ROS) in skeletal muscle cells. Biochem Biophys Res Commun 410:665–670

    Article  CAS  PubMed  Google Scholar 

  • Cabello-Verrugio C, Cordova G, Salas JD (2012a) Angiotensin II: role in skeletal muscle atrophy. Curr Protein Pept Sci 13:560–569

    Article  CAS  PubMed  Google Scholar 

  • Cabello-Verrugio C, Morales MG, Cabrera D, Vio CP, Brandan E (2012b) Angiotensin II receptor type 1 blockade decreases CTGF/CCN2-mediated damage and fibrosis in normal and dystrophic skeletal muscles. J Cell Mol Med 16:752–764

    Article  CAS  PubMed  Google Scholar 

  • Cao L, Xun J, Jiang X, Tan R (2013) Propofol up-regulates Mas receptor expression in dorsal root ganglion neurons. Pharmazie 68:677–680

    CAS  PubMed  Google Scholar 

  • Casselbrant A, Kostic S, Lonroth H (2014) The muscular expression of RAS in patients with achalasia. J Renin Angiotensin Aldosterone Syst. doi:10.1177/1470320313498294

  • Chappell MC (2007) Emerging evidence for a functional angiotensin-converting enzyme 2–angiotensin-(1–7)–MAS receptor axis: more than regulation of blood pressure? Hypertension 50:596–599

    Article  CAS  PubMed  Google Scholar 

  • Chen Z, Tang Y, Yang Z, Liu S, Liu Y, Li Y, He W (2013) Endothelin-1 downregulates Mas receptor expression in human cardiomyocytes. Mol Med Rep 8:871–876

    CAS  PubMed  Google Scholar 

  • Dias-Peixoto MF et al (2012) The cardiac expression of Mas receptor is responsive to different physiological and pathological stimuli. Peptides 35:196–201

    Article  CAS  PubMed  Google Scholar 

  • Doyle A, Zhang G, Abdel Fattah EA, Eissa NT, Li YP (2011) Toll-like receptor 4 mediates lipopolysaccharide-induced muscle catabolism via coordinate activation of ubiquitin–proteasome and autophagy–lysosome pathways. FASEB J 25:99–110

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Echeverria-Rodriguez O, Del Valle-Mondragon L, Hong E (2013) Angiotensin 1–7 improves insulin sensitivity by increasing skeletal muscle glucose uptake in vivo. Peptides 51:26–30

    Article  PubMed  Google Scholar 

  • Eley HL, Russell ST, Tisdale MJ (2008) Attenuation of depression of muscle protein synthesis induced by lipopolysaccharide, tumor necrosis factor, and angiotensin II by beta-hydroxy-beta-methylbutyrate. Am J Physiol Endocrinol Metab 295:E1409–E1416

    Article  CAS  PubMed  Google Scholar 

  • El-Hashim AZ, Renno WM, Raghupathy R, Abduo HT, Akhtar S, Benter IF (2012) Angiotensin-(1–7) inhibits allergic inflammation, via the MAS1 receptor, through suppression of ERK1/2- and NF-kappaB-dependent pathways. Br J Pharmacol 166:1964–1976

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Ender SA, Dallmer A, Lassig F, Lendeckel U, Wolke C (2014) Expression and function of the ACE2/angiotensin(1–7)/Mas axis in osteosarcoma cell lines U-2 OS and MNNG-HOS. Mol Med Rep 10:804–810. doi:10.3892/mmr.2014.2266

  • Ferrario CM, Trask AJ, Jessup JA (2005) Advances in biochemical and functional roles of angiotensin-converting enzyme 2 and angiotensin-(1–7) in regulation of cardiovascular function. Am J Physiol Heart Circ Physiol 289:H2281–H2290

    Article  CAS  PubMed  Google Scholar 

  • Ferreira AJ, Bader M, Santos RA (2012a) Therapeutic targeting of the angiotensin-converting enzyme 2/angiotensin-(1–7)/Mas cascade in the renin–angiotensin system: a patent review. Expert Opin Ther Pat 22:567–574

    Article  CAS  PubMed  Google Scholar 

  • Ferreira AJ, Murca TM, Fraga-Silva RA, Castro CH, Raizada MK, Santos RA (2012b) New cardiovascular and pulmonary therapeutic strategies based on the angiotensin-converting enzyme 2/angiotensin-(1–7)/mas receptor axis Int. J Hypertens 2012:147825

    Google Scholar 

  • Filho AG et al (2008) Selective increase of angiotensin(1–7) and its receptor in hearts of spontaneously hypertensive rats subjected to physical training. Exp Physiol 93:589–598

    PubMed  Google Scholar 

  • Foletta VC, White LJ, Larsen AE, Leger B, Russell AP (2011) The role and regulation of MAFbx/atrogin-1 and MuRF1 in skeletal muscle atrophy. Pflugers Arch 461:325–335

    Article  CAS  PubMed  Google Scholar 

  • Fyhrquist F, Saijonmaa O (2008) Renin–angiotensin system revisited. J Intern Med 264:224–236

    Article  CAS  PubMed  Google Scholar 

  • Gava E et al (2009) Angiotensin-(1–7) activates a tyrosine phosphatase and inhibits glucose-induced signalling in proximal tubular cells. Nephrol Dial Transplant 24:1766–1773

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Glass DJ (2003) Molecular mechanisms modulating muscle mass. Trends Mol Med 9:344–350

    Article  CAS  PubMed  Google Scholar 

  • Glass DJ (2005) Skeletal muscle hypertrophy and atrophy signaling pathways. Int J Biochem Cell Biol 37:1974–1984

    Article  CAS  PubMed  Google Scholar 

  • Gomes MD, Lecker SH, Jagoe RT, Navon A, Goldberg AL (2001) Atrogin-1, a muscle-specific F-box protein highly expressed during muscle atrophy. Proc Natl Acad Sci U S A 98:14440–14445

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Gomes ER et al (2010) Angiotensin-(1–7) prevents cardiomyocyte pathological remodeling through a nitric oxide/guanosine 3′,5′-cyclic monophosphate-dependent pathway. Hypertension 55:153–160

    Article  CAS  PubMed  Google Scholar 

  • Grobe JL, Mecca AP, Mao H, Katovich MJ (2006) Chronic angiotensin-(1–7) prevents cardiac fibrosis in DOCA-salt model of hypertension. Am J Physiol Heart Circ Physiol 290:H2417–H2423

    Article  CAS  PubMed  Google Scholar 

  • Gumucio JP, Mendias CL (2013) Atrogin-1, MuRF-1, and sarcopenia. Endocrine 43:12–21

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Iwai M, Horiuchi M (2009) Devil and angel in the renin–angiotensin system: ACE–angiotensin II–AT1 receptor axis vs. ACE2–angiotensin-(1–7)–Mas receptor axis. Hypertens Res 32:533–536

    Article  CAS  PubMed  Google Scholar 

  • Iwata M, Cowling RT, Gurantz D, Moore C, Zhang S, Yuan JX, Greenberg BH (2005) Angiotensin-(1–7) binds to specific receptors on cardiac fibroblasts to initiate antifibrotic and antitrophic effects. Am J Physiol Heart Circ Physiol 289:H2356–H2363

    Article  CAS  PubMed  Google Scholar 

  • Jiang T, Gao L, Guo J, Lu J, Wang Y, Zhang Y (2012) Suppressing inflammation by inhibiting the NF-kappaB pathway contributes to the neuroprotective effect of angiotensin-(1–7) in rats with permanent cerebral ischaemia. Br J Pharmacol 167:1520–1532

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Kim J et al (2009) p38 MAPK participates in muscle-specific RING finger 1-mediated atrophy in cast-immobilized rat gastrocnemius muscle. Korean J Physiol Pharmacol 13:491–496

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Kondo H, Nakagaki I, Sasaki S, Hori S, Itokawa Y (1993) Mechanism of oxidative stress in skeletal muscle atrophied by immobilization. Am J Physiol 265:E839–E844

    CAS  PubMed  Google Scholar 

  • Kondo H, Nishino K, Itokawa Y (1994) Hydroxyl radical generation in skeletal muscle atrophied by immobilization. FEBS Lett 349:169–172

    Article  CAS  PubMed  Google Scholar 

  • Lu J et al (2013) The expression of angiotensin-converting enzyme 2–angiotensin-(1–7)–Mas receptor axis are upregulated after acute cerebral ischemic stroke in rats. Neuropeptides 47:289–295

    Article  CAS  PubMed  Google Scholar 

  • Lubel JS, Herath CB, Burrell LM, Angus PW (2008) Liver disease and the renin–angiotensin system: recent discoveries and clinical implications. J Gastroenterol Hepatol 23:1327–1338

    Article  CAS  PubMed  Google Scholar 

  • Madaro L, Smeriglio P, Molinaro M, Bouché M (2008) Unilateral immobilization: a simple model of limb atrophy in mice. Basic Appl Myol 18:149–153

    Google Scholar 

  • Mancini GB, Khalil N (2005) Angiotensin II type 1 receptor blocker inhibits pulmonary injury. Clin Invest Med 28:118–126

    CAS  PubMed  Google Scholar 

  • Marangoni RA, Carmona AK, Passaglia RC, Nigro D, Fortes ZB, de Carvalho MH (2006) Role of the kallikrein–kinin system in Ang-(1–7)-induced vasodilation in mesenteric arterioles of Wistar rats studied in vivo-in situ. Peptides 27:1770–1775

    Article  CAS  PubMed  Google Scholar 

  • Marshall AC, Shaltout HA, Nautiyal M, Rose JC, Chappell MC, Diz DI (2013) Fetal betamethasone exposure attenuates angiotensin-(1–7)–Mas receptor expression in the dorsal medulla of adult sheep. Peptides 44:25–31

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • McClung JM, Judge AR, Powers SK, Yan Z (2010) p38 MAPK links oxidative stress to autophagy-related gene expression in cachectic muscle wasting. Am J Physiol Cell Physiol 298:C542–C549

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Metzger R, Bader M, Ludwig T, Berberich C, Bunnemann B, Ganten D (1995) Expression of the mouse and rat mas proto-oncogene in the brain and peripheral tissues. FEBS Lett 357:27–32

    Article  CAS  PubMed  Google Scholar 

  • Morales MG, Cabello-Verrugio C, Santander C, Cabrera D, Goldschmeding R, Brandan E (2011) CTGF/CCN-2 over-expression can directly induce features of skeletal muscle dystrophy. J Pathol 225:490–501

    Article  CAS  PubMed  Google Scholar 

  • Morales MG et al (2012) Angiotensin II-induced pro-fibrotic effects require p38MAPK activity and transforming growth factor beta 1 expression in skeletal muscle cells. Int J Biochem Cell Biol 44:1993–2002

    Article  CAS  PubMed  Google Scholar 

  • Morales MG, Cabrera D, Cespedes C, Vio CP, Vazquez Y, Brandan E, Cabello-Verrugio C (2013a) Inhibition of the angiotensin-converting enzyme decreases skeletal muscle fibrosis in dystrophic mice by a diminution in the expression and activity of connective tissue growth factor (CTGF/CCN-2). Cell Tissue Res 353:173–187

    Article  CAS  PubMed  Google Scholar 

  • Morales MG, Gutierrez J, Cabello-Verrugio C, Cabrera D, Lipson KE, Goldschmeding R, Brandan E (2013b) Reducing CTGF/CCN2 slows down mdx muscle dystrophy and improves cell therapy. Hum Mol Genet 22:4938–4951

    Article  CAS  PubMed  Google Scholar 

  • Morales MG et al (2014) The Ang-(1–7)/Mas-1 axis attenuates the expression and signalling of TGF-beta1 induced by AngII in mouse skeletal muscle. Clin Sci (Lond) 127:251–264

    Article  CAS  Google Scholar 

  • Mouisel E, Vignaud A, Hourde C, Butler-Browne G, Ferry A (2010) Muscle weakness and atrophy are associated with decreased regenerative capacity and changes in mTOR signaling in skeletal muscles of venerable (18–24-month-old) dystrophic mdx mice. Muscle Nerve 41:809–818

    Article  CAS  PubMed  Google Scholar 

  • Munoz MC, Giani JF, Dominici FP (2010) Angiotensin-(1–7) stimulates the phosphorylation of Akt in rat extracardiac tissues in vivo via receptor Mas. Regul Pept 161:1–7

    Article  CAS  PubMed  Google Scholar 

  • Munoz MC, Giani JF, Burghi V, Mayer MA, Carranza A, Taira CA, Dominici FP (2012) The Mas receptor mediates modulation of insulin signaling by angiotensin-(1–7). Regul Pept 177:1–11

    Article  CAS  PubMed  Google Scholar 

  • Painemal P, Acuna MJ, Riquelme C, Brandan E, Cabello-Verrugio C (2013) Transforming growth factor type beta 1 increases the expression of angiotensin II receptor type 2 by a SMAD- and p38 MAPK-dependent mechanism in skeletal muscle. BioFactors 39:467–475

    Article  CAS  PubMed  Google Scholar 

  • Pereira VM, Reis FM, Santos RA, Cassali GD, Santos SH, Honorato-Sampaio K, dos Reis AM (2009) Gonadotropin stimulation increases the expression of angiotensin-(1–7) and MAS receptor in the rat ovary. Reprod Sci 16:1165–1174

    Article  CAS  PubMed  Google Scholar 

  • Powers SK, Kavazis AN, McClung JM (2007) Oxidative stress and disuse muscle atrophy. J Appl Physiol 102:2389–2397

    Article  CAS  PubMed  Google Scholar 

  • Powers SK, Smuder AJ, Judge AR (2012) Oxidative stress and disuse muscle atrophy: cause or consequence? Curr Opin Clin Nutr Metab Care 15:240–245

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Prasannarong M, Santos FR, Henriksen EJ (2012) ANG-(1–7) reduces ANG II-induced insulin resistance by enhancing Akt phosphorylation via a Mas receptor-dependent mechanism in rat skeletal muscle. Biochem Biophys Res Commun 426:369–373

    Article  CAS  PubMed  Google Scholar 

  • Prokop JW et al (2014) MAS promoter regulation: a role for Sry and tyrosine nitration of the KRAB domain of ZNF274 as a feedback mechanism. Clin Sci (Lond) 126:727–738

    Article  CAS  Google Scholar 

  • Rezk BM, Yoshida T, Semprun-Prieto L, Higashi Y, Sukhanov S, Delafontaine P (2012) Angiotensin II infusion induces marked diaphragmatic skeletal muscle atrophy. PLoS ONE 7:e30276

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Sabharwal R, Cicha MZ, Sinisterra RD, De Sousa FB, Santos RA, Chapleau MW (2014) Chronic oral administration of Ang-(1–7) improves skeletal muscle, autonomic and locomotor phenotypes in muscular dystrophy. Clin Sci (Lond) 127:101–109

    Article  CAS  Google Scholar 

  • Sampaio WO, Souza dos Santos RA, Faria-Silva R, da Mata Machado LT, Schiffrin EL, Touyz RM (2007) Angiotensin-(1–7) through receptor Mas mediates endothelial nitric oxide synthase activation via Akt-dependent pathways. Hypertension 49:185–192

    Article  CAS  PubMed  Google Scholar 

  • Sanders PM, Russell ST, Tisdale MJ (2005) Angiotensin II directly induces muscle protein catabolism through the ubiquitin–proteasome proteolytic pathway and may play a role in cancer cachexia. Br J Cancer 93:425–434

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Sandri M et al (2004) Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 117:399–412

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Santos RA et al (2003) Angiotensin-(1–7) is an endogenous ligand for the G protein-coupled receptor Mas. Proc Natl Acad Sci USA 100:8258–8263

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Santos SH et al (2013) Oral Angiotensin-(1–7) prevented obesity and hepatic inflammation by inhibition of resistin/TLR4/MAPK/NF-kappaB in rats fed with high-fat diet. Peptides 46:47–52

    Article  CAS  PubMed  Google Scholar 

  • Semprun-Prieto LC et al (2011) Angiotensin II induced catabolic effect and muscle atrophy are redox dependent. Biochem Biophys Res Commun 409:217–221

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Souza LL, Costa-Neto CM (2012) Angiotensin-(1–7) decreases LPS-induced inflammatory response in macrophages. J Cell Physiol 227:2117–2122

    Article  CAS  PubMed  Google Scholar 

  • Stitt TN et al (2004) The IGF-1/PI3 K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. Mol Cell 14:395–403

    Article  CAS  PubMed  Google Scholar 

  • Sukhanov S, Semprun-Prieto L, Yoshida T, Michael Tabony A, Higashi Y, Galvez S, Delafontaine P (2011) Angiotensin II, oxidative stress and skeletal muscle wasting. Am J Med Sci 342:143–147

    Article  PubMed Central  PubMed  Google Scholar 

  • Tallant EA, Ferrario CM, Gallagher PE (2005) Angiotensin-(1–7) inhibits growth of cardiac myocytes through activation of the mas receptor. Am J Physiol Heart Circ Physiol 289:H1560–H1566

    Article  CAS  PubMed  Google Scholar 

  • Tisdale MJ (2005) The ubiquitin–proteasome pathway as a therapeutic target for muscle wasting. J Support Oncol 3:209–217

    CAS  PubMed  Google Scholar 

  • Villar AJ, Pedersen RA (1994) Parental imprinting of the Mas protooncogene in mouse. Nat Genet 8:373–379

    Article  CAS  PubMed  Google Scholar 

  • Wang Y, Wang J, Liu R, Qi H, Wen Y, Sun F, Yin C (2012) Severe acute pancreatitis is associated with upregulation of the ACE2–angiotensin-(1–7)–Mas axis and promotes increased circulating angiotensin-(1–7). Pancreatology 12:451–457

    Article  CAS  PubMed  Google Scholar 

  • Wang Y et al (2014) TNF-alpha and IL-1beta neutralization ameliorates angiotensin II-induced cardiac damage in male mice. Endocrinology 155:2677–2687. doi:10.1210/en.2013-2065

  • Wong TP, Ho KY, Ng EK, Debnam ES, Leung PS (2012) Upregulation of ACE2–ANG-(1–7)–Mas axis in jejunal enterocytes of type 1 diabetic rats: implications for glucose transport. Am J Physiol Endocrinol Metab 303:E669–E681

    Article  CAS  PubMed  Google Scholar 

  • Yu Z, Li P, Zhang M, Hannink M, Stamler JS, Yan Z (2008) Fiber type-specific nitric oxide protects oxidative myofibers against cachectic stimuli. PLoS ONE 3:e2086

    Article  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgments

This study was supported by research grants from Association-Francaise Contre Les Myopathies AFM 16670 (CCV); FONDECYT 1120380 (CCV), 3130593 (MGM), 1121078 (FS); and the Millennium Institute on Immunology and Immunotherapy, P09-016-F (FS); UNAB-DI-281-13/R (CCV).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Claudio Cabello-Verrugio.

Additional information

María Gabriela Morales and Johanna Abrigo have contributed equally to this work.

Electronic supplementary material

Below is the link to the electronic supplementary material.

418_2014_1275_MOESM1_ESM.tif

Fig. S1: Upregulation of the Mas receptor induced by AngII in C 2 C 12 myotubes is dependent on the AT-1 receptor. C2C12 myotubes from day 5 were pre-incubated with losartan (10 μM) or PD123319 (10 μM) for 1 h prior to the incubation with AngII (500 nM) for 3 h. At the end of this treatment, the mRNA levels of Mas were determined by RT-qPCR using β-actin as the reference gene. The expression was expressed as the fold of induction, normalized to the levels in the control cells. Values correspond to the mean ± SD of three independent experiments (*, P < 0.05 relative to the control cells, #, P < 0.05 relative to AngII-vehicle) (TIFF 219 kb)

418_2014_1275_MOESM2_ESM.tif

Fig. S2: The Mas receptor is increased in the tibialis anterior of AngII-induced skeletal muscle atrophy. Skeletal muscle atrophy was induced in C57BL/10J mice by osmotic infusion of AngII (1 μg/kg/min). (a) Mas receptor was detected by indirect immunohistochemical analysis in cryosections of tibialis anterior muscles from control mice and mice treated with AngII for 14 days. Nuclei were labelled with haematoxylin. The bar corresponds to 50 μm. The images are representative of three independent experiments, using three mice for each experimental condition. (b) Histological analysis of the tibialis anterior of control mice and mice treated with AngII for 14 days. Muscle cross sections were stained with haematoxylin and eosin to visualize muscle architecture. The bar corresponds to 50 μm. The images are representative of three independent experiments, using three mice for each experimental condition. (c) Quantitative analysis of the fibre diameter from three independent experiments using three mice for each experimental condition. The values are expressed as the percentage of the total fibres quantified (*, P < 0.05 relative to the control) (TIFF 8578 kb)

418_2014_1275_MOESM3_ESM.tif

Fig. S3: Upregulation of Mas receptor induced by LPS in C 2 C 12 myotubes is dependent on the TLR-4 receptor. C2C12 myotubes from day 5 were pre-incubated with CLI-095 (5 μM) for 1 h prior to the incubation with LPS (500 ng/ml) for 3 h. At the end of this treatment, mRNA levels of Mas were determined by RT-qPCR using β-actin as the reference gene. The expression was expressed as the fold of induction, normalized to the levels in the control cells. Values correspond to the mean ± SD of three independent experiments (*, P < 0.05 relative to the control cells, #, P < 0.05 relative to LPS-vehicle) (TIFF 208 kb)

418_2014_1275_MOESM4_ESM.tif

Fig. S4: The Mas receptor is upregulated in the tibialis anterior of LPS-induced skeletal muscle atrophy. Skeletal muscle atrophy was induced in C57BL/10J mice by intraperitoneal injection of LPS (1 mg/kg). (a) The Mas receptor was detected by indirect immunohistochemical analysis in cryosections of the tibialis anterior of muscles from control and LPS-treated mice for 14 days. Nuclei were labelled with haematoxylin. The bar corresponds to 50 μm. The images are representative of three independent experiments, using three mice for each experimental condition. (b) Histological analysis of the tibialis anterior of control mice and mice treated with LPS for 14 days. Muscle cross sections were stained with haematoxylin and eosin to visualize muscle architecture. The bar corresponds to 50 μm. The images are representative of three independent experiments, using three mice for each experimental condition. (c) Quantitative analysis of the fibre diameter from three independent experiments using three mice for each experimental condition. The values are expressed as the percentage of the total fibres quantified (*, P < 0.05 relative to the control) (TIFF 8007 kb)

418_2014_1275_MOESM5_ESM.tif

Fig. S5: The Mas receptor is augmented in the tibialis anterior of disuse-induced skeletal muscle atrophy. Skeletal muscle atrophy was induced in C57BL/10J mice by unilateral immobilizated hindlimb by casting. (a) The Mas receptor was detected by indirect immunohistochemical analysis in cryosections of tibialis anterior muscles from non-immobilized hindlimbs and hindlimbs immobilized for 14 days. Nuclei were labelled with haematoxylin. The bar corresponds to 50 μm. The images are representative of three independent experiments, using three mice for each experimental condition. (b) Histological analysis of tibialis anterior non-immobilized hindlimbs and hindlimbs immobilized for 14 days. Muscle cross sections were stained with haematoxylin and eosin to visualize muscle architecture. The bar corresponds to 50 μm. The images are representative of three independent experiments, using three mice for each experimental condition. (c) Quantitative analysis of the fibre diameter from three independent experiments using three mice for each experimental condition. The values are expressed as the percentage of the total fibres quantified (*, P < 0.05 relative to the non-immobilizated muscles) (TIFF 8431 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Morales, M.G., Abrigo, J., Meneses, C. et al. Expression of the Mas receptor is upregulated in skeletal muscle wasting. Histochem Cell Biol 143, 131–141 (2015). https://doi.org/10.1007/s00418-014-1275-1

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00418-014-1275-1

Keywords

Navigation