Endocrinol Metab.  2021 Jun;36(3):478-490. 10.3803/EnM.2021.1081.

Receptor-Mediated Muscle Homeostasis as a Target for Sarcopenia Therapeutics

Affiliations
  • 1Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
  • 2Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Korea
  • 3Aventi Inc., Daejeon, Korea

Abstract

Sarcopenia is a disease characterized by age-related decline of skeletal muscle mass and function. The molecular mechanisms of the pathophysiology of sarcopenia form a complex network due to the involvement of multiple interconnected signaling pathways. Therefore, signaling receptors are major targets in pharmacological strategies in general. To provide a rationale for pharmacological interventions for sarcopenia, we herein describe several druggable signaling receptors based on their role in skeletal muscle homeostasis and changes in their activity with aging. A brief overview is presented of the efficacy of corresponding drug candidates under clinical trials. Strategies targeting the androgen receptor, vitamin D receptor, Insulin-like growth factor-1 receptor, and ghrelin receptor primarily focus on promoting anabolic action using natural ligands or mimetics. Strategies involving activin receptors and angiotensin receptors focus on inhibiting catabolic action. This review may help to select specific targets or combinations of targets in the future.

Keyword

Sarcopenia; Muscle, skeletal; Aging; Drug therapy; Receptors, cell surface; Receptors, cytoplasmic and nuclear; Signal transduction; Molecular mechanisms of pharmacological action; Clinical trial

Figure

  • Fig. 1. Cell signaling pathways and pharmacological strategies for sarcopenia. GHSR, growth hormone secretagogues receptor or ghrelin receptor; IGF-1R, insulin-like growth factor 1 receptor; ACE, angiotensin-converting enzyme; ActRIIB, activin type II receptor; Ang1, angiotensin I; Ang2, angiotensin II; AGTR1, angiotensin II receptor type 1; SARM, selective androgen receptor modulator; AR, androgen receptor; VDR, vitamin D receptor; mTOR, mammalian target of the rapamycin; FOXO, forkhead box O.

  • Fig. 2. Distribution of target families of small-molecule drugs in sarcopenia. Related data are described on ClinicalTrials.gov, and the figure includes data up to March 2021. IS, indoxyl sulfate; XO, xanthine oxidase.

  • Fig. 3. The current status of articles on sarcopenia and cell signaling receptors. A literature search was conducted in PubMed (keyword: sarcopenia, receptor, and/or related-term) and the relevant data extend up to March 2021. IGF-1, insulin-like growth factor 1; IL-15, interleukin 15.


Reference

1. Cruz-Jentoft AJ, Sayer AA. Sarcopenia. Lancet. 2019; 393:2636–46.
Article
2. Cao L, Morley JE. Sarcopenia is recognized as an independent condition by an International Classification of Disease, Tenth Revision, Clinical Modification (ICD-10-CM) code. J Am Med Dir Assoc. 2016; 17:675–7.
Article
3. Chen LK, Woo J, Assantachai P, Auyeung TW, Chou MY, Iijima K, et al. Asian Working Group for Sarcopenia: 2019 consensus update on sarcopenia diagnosis and treatment. J Am Med Dir Assoc. 2020; 21:300–7.
Article
4. Cruz-Jentoft AJ, Landi F, Schneider SM, Zuniga C, Arai H, Boirie Y, et al. Prevalence of and interventions for sarcopenia in ageing adults: a systematic review: report of the International Sarcopenia Initiative (EWGSOP and IWGS). Age Ageing. 2014; 43:748–59.
Article
5. Cruz-Jentoft AJ, Bahat G, Bauer J, Boirie Y, Bruyere O, Cederholm T, et al. Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing. 2019; 48:16–31.
Article
6. Chen LK, Liu LK, Woo J, Assantachai P, Auyeung TW, Bahyah KS, et al. Sarcopenia in Asia: consensus report of the Asian Working Group for Sarcopenia. J Am Med Dir Assoc. 2014; 15:95–101.
Article
7. Dent E, Morley JE, Cruz-Jentoft AJ, Arai H, Kritchevsky SB, Guralnik J, et al. International Clinical Practice Guidelines for Sarcopenia (ICFSR): screening, diagnosis and management. J Nutr Health Aging. 2018; 22:1148–61.
Article
8. Kwak JY, Kwon KS. Pharmacological interventions for treatment of sarcopenia: current status of drug development for sarcopenia. Ann Geriatr Med Res. 2019; 23:98–104.
Article
9. Dao T, Green AE, Kim YA, Bae SJ, Ha KT, Gariani K, et al. Sarcopenia and muscle aging: a brief overview. Endocrinol Metab (Seoul). 2020; 35:716–32.
Article
10. Ceglia L, Niramitmahapanya S, da Silva Morais M, Rivas DA, Harris SS, Bischoff-Ferrari H, et al. A randomized study on the effect of vitamin D3 supplementation on skeletal muscle morphology and vitamin D receptor concentration in older women. J Clin Endocrinol Metab. 2013; 98:E1927–35.
11. Snyder PJ, Bhasin S, Cunningham GR, Matsumoto AM, Stephens-Shields AJ, Cauley JA, et al. Lessons from the testosterone trials. Endocr Rev. 2018; 39:369–86.
Article
12. Becker C, Lord SR, Studenski SA, Warden SJ, Fielding RA, Recknor CP, et al. Myostatin antibody (LY2495655) in older weak fallers: a proof-of-concept, randomised, phase 2 trial. Lancet Diabetes Endocrinol. 2015; 3:948–57.
Article
13. Rooks D, Praestgaard J, Hariry S, Laurent D, Petricoul O, Perry RG, et al. Treatment of sarcopenia with bimagrumab: results from a phase ii, randomized, controlled, proof-of-concept study. J Am Geriatr Soc. 2017; 65:1988–95.
Article
14. Rooks D, Swan T, Goswami B, Filosa LA, Bunte O, Panchaud N, et al. Bimagrumab vs optimized standard of care for treatment of sarcopenia in community-dwelling older adults: a randomized clinical trial. JAMA Netw Open. 2020; 3:e2020836.
15. Lee SM, Lee SH, Jung Y, Lee Y, Yoon JH, Choi JY, et al. FABP3-mediated membrane lipid saturation alters fluidity and induces ER stress in skeletal muscle with aging. Nat Commun. 2020; 11:5661.
Article
16. Kalyani RR, Corriere M, Ferrucci L. Age-related and disease-related muscle loss: the effect of diabetes, obesity, and other diseases. Lancet Diabetes Endocrinol. 2014; 2:819–29.
Article
17. Jaitovich A, Barreiro E. Skeletal muscle dysfunction in chronic obstructive pulmonary disease: what we know and can do for our patients. Am J Respir Crit Care Med. 2018; 198:175–86.
Article
18. Furrer R, Handschin C. Muscle wasting diseases: novel targets and treatments. Annu Rev Pharmacol Toxicol. 2019; 59:315–39.
Article
19. Schiaffino S, Dyar KA, Ciciliot S, Blaauw B, Sandri M. Mechanisms regulating skeletal muscle growth and atrophy. FEBS J. 2013; 280:4294–314.
Article
20. Campins L, Camps M, Riera A, Pleguezuelos E, Yebenes JC, Serra-Prat M. Oral drugs related with muscle wasting and sarcopenia. A review. Pharmacology. 2017; 99:1–8.
Article
21. Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R, et al. Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol. 2001; 3:1014–9.
Article
22. From the American Association of Neurological Surgeons (AANS); American Society of Neuroradiology (ASNR); Cardiovascular and Interventional Radiology Society of Europe (CIRSE); Canadian Interventional Radiology Association (CIRA); Congress of Neurological Surgeons (CNS); European Society of Minimally Invasive Neurological Therapy (ESMINT), et al. Multisociety consensus quality improvement revised consensus statement for endovascular therapy of acute ischemic stroke. Int J Stroke. 2018; 13:612–32.
23. Ali S, Garcia JM. Sarcopenia, cachexia and aging: diagnosis, mechanisms and therapeutic options: a mini-review. Gerontology. 2014; 60:294–305.
Article
24. von Haehling S, Steinbeck L, Doehner W, Springer J, Anker SD. Muscle wasting in heart failure: an overview. Int J Biochem Cell Biol. 2013; 45:2257–65.
Article
25. Wiedmer P, Jung T, Castro JP, Pomatto LC, Sun PY, Davies KJ, et al. Sarcopenia: molecular mechanisms and open questions. Ageing Res Rev. 2021; 65:101200.
26. Mankhong S, Kim S, Moon S, Kwak HB, Park DH, Kang JH. Experimental models of sarcopenia: bridging molecular mechanism and therapeutic strategy. Cells. 2020; 9:1385.
Article
27. Suvarna BS. Drug-receptor interactions. Kathmandu Univ Med J (KUMJ). 2011; 9:203–7.
28. Santos R, Ursu O, Gaulton A, Bento AP, Donadi RS, Bologa CG, et al. A comprehensive map of molecular drug targets. Nat Rev Drug Discov. 2017; 16:19–34.
Article
29. Waller DG, Sampson AP. Medical pharmacology and therapeutics. 5th ed. Edinburgh: Elsevier;2018. Chapter 1, Principles of pharmacology and mechanisms of drug action. p. 3–31.
30. Panyam J, Labhasetwar V. Targeting intracellular targets. Curr Drug Deliv. 2004; 1:235–47.
Article
31. Lounsbury K. Pharmacology. San Diego: Academic Press;2009. Chapter 6, Signal transduction and second messengers. p. 103–12.
32. Tamai I, Tsuji A. Transporter-mediated permeation of drugs across the blood-brain barrier. J Pharm Sci. 2000; 89:1371–88.
Article
33. Bareford LM, Swaan PW. Endocytic mechanisms for targeted drug delivery. Adv Drug Deliv Rev. 2007; 59:748–58.
Article
34. Adcock IM. Molecular mechanisms of glucocorticosteroid actions. Pulm Pharmacol Ther. 2000; 13:115–26.
Article
35. Tien AH, Sadar MD. Keys to unlock androgen receptor translocation. J Biol Chem. 2019; 294:8711–2.
Article
36. Chen Y, Zajac JD, MacLean HE. Androgen regulation of satellite cell function. J Endocrinol. 2005; 186:21–31.
Article
37. Hobbs CJ, Plymate SR, Rosen CJ, Adler RA. Testosterone administration increases insulin-like growth factor-I levels in normal men. J Clin Endocrinol Metab. 1993; 77:776–9.
Article
38. Morley JE. Pharmacologic options for the treatment of sarcopenia. Calcif Tissue Int. 2016; 98:319–33.
Article
39. Griggs RC, Kingston W, Jozefowicz RF, Herr BE, Forbes G, Halliday D. Effect of testosterone on muscle mass and muscle protein synthesis. J Appl Physiol (1985). 1989; 66:498–503.
Article
40. Serra C, Tangherlini F, Rudy S, Lee D, Toraldo G, Sandor NL, et al. Testosterone improves the regeneration of old and young mouse skeletal muscle. J Gerontol A Biol Sci Med Sci. 2013; 68:17–26.
Article
41. Grech A, Breck J, Heidelbaugh J. Adverse effects of testosterone replacement therapy: an update on the evidence and controversy. Ther Adv Drug Saf. 2014; 5:190–200.
Article
42. Coviello AD, Kaplan B, Lakshman KM, Chen T, Singh AB, Bhasin S. Effects of graded doses of testosterone on erythropoiesis in healthy young and older men. J Clin Endocrinol Metab. 2008; 93:914–9.
Article
43. Tan RS, Salazar JA. Risks of testosterone replacement therapy in ageing men. Expert Opin Drug Saf. 2004; 3:599–606.
Article
44. Hackett GI. Testosterone replacement therapy and mortality in older men. Drug Saf. 2016; 39:117–30.
Article
45. Davis MP, Panikkar R. Sarcopenia associated with chemotherapy and targeted agents for cancer therapy. Ann Palliat Med. 2019; 8:86–101.
Article
46. Narayanan R, Coss CC, Dalton JT. Development of selective androgen receptor modulators (SARMs). Mol Cell Endocrinol. 2018; 465:134–42.
Article
47. Srinath R, Dobs A. Enobosarm (GTx-024, S-22): a potential treatment for cachexia. Future Oncol. 2014; 10:187–94.
Article
48. Dobs AS, Boccia RV, Croot CC, Gabrail NY, Dalton JT, Hancock ML, et al. Effects of enobosarm on muscle wasting and physical function in patients with cancer: a doubleblind, randomised controlled phase 2 trial. Lancet Oncol. 2013; 14:335–45.
Article
49. Crawford J, Prado CM, Johnston MA, Gralla RJ, Taylor RP, Hancock ML, et al. Study design and rationale for the phase 3 clinical development program of enobosarm, a selective androgen receptor modulator, for the prevention and treatment of muscle wasting in cancer patients (POWER trials). Curr Oncol Rep. 2016; 18:37.
Article
50. Papanicolaou DA, Ather SN, Zhu H, Zhou Y, Lutkiewicz J, Scott BB, et al. A phase IIA randomized, placebo-controlled clinical trial to study the efficacy and safety of the selective androgen receptor modulator (SARM), MK-0773 in female participants with sarcopenia. J Nutr Health Aging. 2013; 17:533–43.
Article
51. Wagatsuma A, Sakuma K. Vitamin D signaling in myogenesis: potential for treatment of sarcopenia. Biomed Res Int. 2014; 2014:121254.
Article
52. Wang TT, Tavera-Mendoza LE, Laperriere D, Libby E, MacLeod NB, Nagai Y, et al. Large-scale in silico and microarray-based identification of direct 1,25-dihydroxyvitamin D3 target genes. Mol Endocrinol. 2005; 19:2685–95.
Article
53. Bischoff-Ferrari HA, Borchers M, Gudat F, Durmuller U, Stahelin HB, Dick W. Vitamin D receptor expression in human muscle tissue decreases with age. J Bone Miner Res. 2004; 19:265–9.
Article
54. Tan LJ, Liu SL, Lei SF, Papasian CJ, Deng HW. Molecular genetic studies of gene identification for sarcopenia. Hum Genet. 2012; 131:1–31.
Article
55. Bhat M, Kalam R, Qadri SS, Madabushi S, Ismail A. Vitamin D deficiency-induced muscle wasting occurs through the ubiquitin proteasome pathway and is partially corrected by calcium in male rats. Endocrinology. 2013; 154:4018–29.
Article
56. Girgis CM, Clifton-Bligh RJ, Hamrick MW, Holick MF, Gunton JE. The roles of vitamin D in skeletal muscle: form, function, and metabolism. Endocr Rev. 2013; 34:33–83.
Article
57. Endo I, Inoue D, Mitsui T, Umaki Y, Akaike M, Yoshizawa T, et al. Deletion of vitamin D receptor gene in mice results in abnormal skeletal muscle development with deregulated expression of myoregulatory transcription factors. Endocrinology. 2003; 144:5138–44.
Article
58. Girgis CM, Cha KM, So B, Tsang M, Chen J, Houweling PJ, et al. Mice with myocyte deletion of vitamin D receptor have sarcopenia and impaired muscle function. J Cachexia Sarcopenia Muscle. 2019; 10:1228–40.
Article
59. Beaudart C, Buckinx F, Rabenda V, Gillain S, Cavalier E, Slomian J, et al. The effects of vitamin D on skeletal muscle strength, muscle mass, and muscle power: a systematic review and meta-analysis of randomized controlled trials. J Clin Endocrinol Metab. 2014; 99:4336–45.
Article
60. Bian A, Ma Y, Zhou X, Guo Y, Wang W, Zhang Y, et al. Association between sarcopenia and levels of growth hormone and insulin-like growth factor-1 in the elderly. BMC Musculoskelet Disord. 2020; 21:214.
Article
61. Musaro A, McCullagh K, Paul A, Houghton L, Dobrowolny G, Molinaro M, et al. Localized Igf-1 transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle. Nat Genet. 2001; 27:195–200.
Article
62. Yoshida T, Delafontaine P. Mechanisms of IGF-1-mediated regulation of skeletal muscle hypertrophy and atrophy. Cells. 2020; 9:1970.
Article
63. Kwak JY, Hwang H, Kim SK, Choi JY, Lee SM, Bang H, et al. Prediction of sarcopenia using a combination of multiple serum biomarkers. Sci Rep. 2018; 8:8574.
Article
64. Schakman O, Gilson H, de Coninck V, Lause P, Verniers J, Havaux X, et al. Insulin-like growth factor-I gene transfer by electroporation prevents skeletal muscle atrophy in glucocorticoid-treated rats. Endocrinology. 2005; 146:1789–97.
Article
65. Shavlakadze T, White J, Hoh JF, Rosenthal N, Grounds MD. Targeted expression of insulin-like growth factor-I reduces early myofiber necrosis in dystrophic mdx mice. Mol Ther. 2004; 10:829–43.
Article
66. Park S, Brisson BK, Liu M, Spinazzola JM, Barton ER. Mature IGF-I excels in promoting functional muscle recovery from disuse atrophy compared with pro-IGF-IA. J Appl Physiol (1985). 2014; 116:797–806.
Article
67. Barton-Davis ER, Shoturma DI, Musaro A, Rosenthal N, Sweeney HL. Viral mediated expression of insulin-like growth factor I blocks the aging-related loss of skeletal muscle function. Proc Natl Acad Sci U S A. 1998; 95:15603–7.
Article
68. Anderson LJ, Tamayose JM, Garcia JM. Use of growth hormone, IGF-I, and insulin for anabolic purpose: pharmacological basis, methods of detection, and adverse effects. Mol Cell Endocrinol. 2018; 464:65–74.
Article
69. Williams RM, McDonald A, O’Savage M, Dunger DB. Mecasermin rinfabate: rhIGF-I/rhIGFBP-3 complex: iPLEX. Expert Opin Drug Metab Toxicol. 2008; 4:311–24.
70. Sullivan DH, Carter WJ, Warr WR, Williams LH. Side effects resulting from the use of growth hormone and insulinlike growth factor-I as combined therapy to frail elderly patients. J Gerontol A Biol Sci Med Sci. 1998; 53:M183–7.
Article
71. Laron Z. The essential role of IGF-I: lessons from the longterm study and treatment of children and adults with Laron syndrome. J Clin Endocrinol Metab. 1999; 84:4397–404.
Article
72. Major JM, Laughlin GA, Kritz-Silverstein D, Wingard DL, Barrett-Connor E. Insulin-like growth factor-I and cancer mortality in older men. J Clin Endocrinol Metab. 2010; 95:1054–9.
Article
73. Liew WK, Kang PB. Recent developments in the treatment of Duchenne muscular dystrophy and spinal muscular atrophy. Ther Adv Neurol Disord. 2013; 6:147–60.
Article
74. Gnanapavan S, Kola B, Bustin SA, Morris DG, McGee P, Fairclough P, et al. The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in humans. J Clin Endocrinol Metab. 2002; 87:2988.
Article
75. Guan XM, Yu H, Palyha OC, McKee KK, Feighner SD, Sirinathsinghji DJ, et al. Distribution of mRNA encoding the growth hormone secretagogue receptor in brain and peripheral tissues. Brain Res Mol Brain Res. 1997; 48:23–9.
Article
76. Gaskin FS, Farr SA, Banks WA, Kumar VB, Morley JE. Ghrelin-induced feeding is dependent on nitric oxide. Peptides. 2003; 24:913–8.
Article
77. Chen JA, Splenser A, Guillory B, Luo J, Mendiratta M, Belinova B, et al. Ghrelin prevents tumour- and cisplatininduced muscle wasting: characterization of multiple mechanisms involved. J Cachexia Sarcopenia Muscle. 2015; 6:132–43.
Article
78. Collden G, Tschop MH, Muller TD. Therapeutic potential of targeting the ghrelin pathway. Int J Mol Sci. 2017; 18:798.
Article
79. Wu CS, Wei Q, Wang H, Kim DM, Balderas M, Wu G, et al. Protective effects of ghrelin on fasting-induced muscle atrophy in aging mice. J Gerontol A Biol Sci Med Sci. 2020; 75:621–30.
Article
80. Nass R, Pezzoli SS, Oliveri MC, Patrie JT, Harrell FE Jr, Clasey JL, et al. Effects of an oral ghrelin mimetic on body composition and clinical outcomes in healthy older adults: a randomized trial. Ann Intern Med. 2008; 149:601–11.
Article
81. Takayama K, Katakami N, Yokoyama T, Atagi S, Yoshimori K, Kagamu H, et al. Anamorelin (ONO-7643) in Japanese patients with non-small cell lung cancer and cachexia: results of a randomized phase 2 trial. Support Care Cancer. 2016; 24:3495–505.
Article
82. Gordon KJ, Blobe GC. Role of transforming growth factorbeta superfamily signaling pathways in human disease. Biochim Biophys Acta. 2008; 1782:197–228.
Article
83. Rooks DS, Laurent D, Praestgaard J, Rasmussen S, Bartlett M, Tanko LB. Effect of bimagrumab on thigh muscle volume and composition in men with casting-induced atrophy. J Cachexia Sarcopenia Muscle. 2017; 8:727–34.
Article
84. Elkina Y, von Haehling S, Anker SD, Springer J. The role of myostatin in muscle wasting: an overview. J Cachexia Sarcopenia Muscle. 2011; 2:143–51.
Article
85. Park HM. Current status of sarcopenia in Korea: a focus on Korean geripausal women. Ann Geriatr Med Res. 2018; 22:52–61.
Article
86. Sartori R, Gregorevic P, Sandri M. TGFβ and BMP signaling in skeletal muscle: potential significance for muscle-related disease. Trends Endocrinol Metab. 2014; 25:464–71.
Article
87. Amirouche A, Durieux AC, Banzet S, Koulmann N, Bonnefoy R, Mouret C, et al. Down-regulation of Akt/mammalian target of rapamycin signaling pathway in response to myostatin overexpression in skeletal muscle. Endocrinology. 2009; 150:286–94.
Article
88. Chen JL, Colgan TD, Walton KL, Gregorevic P, Harrison CA. The TGF-β signalling network in muscle development, adaptation and disease. Adv Exp Med Biol. 2016; 900:97–131.
Article
89. McPherron AC, Lee SJ. Double muscling in cattle due to mutations in the myostatin gene. Proc Natl Acad Sci U S A. 1997; 94:12457–61.
90. Thomas M, Langley B, Berry C, Sharma M, Kirk S, Bass J, et al. Myostatin, a negative regulator of muscle growth, functions by inhibiting myoblast proliferation. J Biol Chem. 2000; 275:40235–43.
Article
91. Rios R, Carneiro I, Arce VM, Devesa J. Myostatin is an inhibitor of myogenic differentiation. Am J Physiol Cell Physiol. 2002; 282:C993–9.
92. Wagner KR, McPherron AC, Winik N, Lee SJ. Loss of myostatin attenuates severity of muscular dystrophy in mdx mice. Ann Neurol. 2002; 52:832–6.
93. Lee SJ, Huynh TV, Lee YS, Sebald SM, Wilcox-Adelman SA, Iwamori N, et al. Role of satellite cells versus myofibers in muscle hypertrophy induced by inhibition of the myostatin/activin signaling pathway. Proc Natl Acad Sci U S A. 2012; 109:E2353–60.
Article
94. Schuelke M, Wagner KR, Stolz LE, Hubner C, Riebel T, Komen W, et al. Myostatin mutation associated with gross muscle hypertrophy in a child. N Engl J Med. 2004; 350:2682–8.
Article
95. Singh P, Rong H, Gordi T, Bosley J, Bhattacharya I. Translational pharmacokinetic/pharmacodynamic analysis of MYO-029 antibody for muscular dystrophy. Clin Transl Sci. 2016; 9:302–10.
Article
96. Suh J, Lee YS. Myostatin inhibitors: panacea or predicament for musculoskeletal disorders? J Bone Metab. 2020; 27:151–65.
Article
97. Suh J, Kim NK, Lee SH, Eom JH, Lee Y, Park JC, et al. GDF11 promotes osteogenesis as opposed to MSTN, and follistatin, a MSTN/GDF11 inhibitor, increases muscle mass but weakens bone. Proc Natl Acad Sci U S A. 2020; 117:4910–20.
Article
98. Campbell C, McMillan HJ, Mah JK, Tarnopolsky M, Selby K, McClure T, et al. Myostatin inhibitor ACE-031 treatment of ambulatory boys with Duchenne muscular dystrophy: results of a randomized, placebo-controlled clinical trial. Muscle Nerve. 2017; 55:458–64.
Article
99. Pirruccello-Straub M, Jackson J, Wawersik S, Webster MT, Salta L, Long K, et al. Blocking extracellular activation of myostatin as a strategy for treating muscle wasting. Sci Rep. 2018; 8:2292.
Article
100. Benigni A, Cassis P, Remuzzi G. Angiotensin II revisited: new roles in inflammation, immunology and aging. EMBO Mol Med. 2010; 2:247–57.
Article
101. Benigni A, Corna D, Zoja C, Sonzogni A, Latini R, Salio M, et al. Disruption of the Ang II type 1 receptor promotes longevity in mice. J Clin Invest. 2009; 119:524–30.
Article
102. Yabumoto C, Akazawa H, Yamamoto R, Yano M, Kudo-Sakamoto Y, Sumida T, et al. Angiotensin II receptor blockade promotes repair of skeletal muscle through down-regulation of aging-promoting C1q expression. Sci Rep. 2015; 5:14453.
Article
103. Heisterberg MF, Andersen JL, Schjerling P, Bulow J, Lauersen JB, Roeber HL, et al. Effect of losartan on the acute response of human elderly skeletal muscle to exercise. Med Sci Sports Exerc. 2018; 50:225–35.
Article
104. Burks TN, Andres-Mateos E, Marx R, Mejias R, Van Erp C, Simmers JL, et al. Losartan restores skeletal muscle remodeling and protects against disuse atrophy in sarcopenia. Sci Transl Med. 2011; 3:82. ra37.
Article
105. Kamo T, Akazawa H, Komuro I. Pleiotropic effects of angiotensin II receptor signaling in cardiovascular homeostasis and aging. Int Heart J. 2015; 56:249–54.
Article
106. Burton LA, Sumukadas D. Optimal management of sarcopenia. Clin Interv Aging. 2010; 5:217–28.
107. Maggio M, Ceda GP, Lauretani F, Pahor M, Bandinelli S, Najjar SS, et al. Relation of angiotensin-converting enzyme inhibitor treatment to insulin-like growth factor-1 serum levels in subjects >65 years of age (the InCHIANTI study). Am J Cardiol. 2006; 97:1525–9.
Article
108. Hutcheon SD, Gillespie ND, Crombie IK, Struthers AD, McMurdo ME. Perindopril improves six minute walking distance in older patients with left ventricular systolic dysfunction: a randomised double blind placebo controlled trial. Heart. 2002; 88:373–7.
Article
109. Peters R, Beckett N, Burch L, de Vernejoul MC, Liu L, Duggan J, et al. The effect of treatment based on a diuretic (indapamide) +/− ACE inhibitor (perindopril) on fractures in the Hypertension in the Very Elderly Trial (HYVET). Age Ageing. 2010; 39:609–16.
Article
110. Elbaz M, Yanay N, Aga-Mizrachi S, Brunschwig Z, Kassis I, Ettinger K, et al. Losartan, a therapeutic candidate in congenital muscular dystrophy: studies in the dy(2J)/dy(2J) mouse. Ann Neurol. 2012; 71:699–708.
Article
111. Kim M, Won CW. Sarcopenia is associated with cognitive impairment mainly due to slow gait speed: results from the Korean Frailty and Aging Cohort Study (KFACS). Int J Environ Res Public Health. 2019; 16:1491.
Article
112. Aguirre F, Abrigo J, Gonzalez F, Gonzalez A, Simon F, Cabello-Verrugio C. Protective effect of angiotensin 1-7 on sarcopenia induced by chronic liver disease in mice. Int J Mol Sci. 2020; 21:3891.
Article
113. Capogrosso RF, Mantuano P, Uaesoontrachoon K, Cozzoli A, Giustino A, Dow T, et al. Ryanodine channel complex stabilizer compound S48168/ARM210 as a disease modifier in dystrophin-deficient mdx mice: proof-of-concept study and independent validation of efficacy. FASEB J. 2018; 32:1025–43.
114. Ebner DC, Bialek P, El-Kattan AF, Ambler CM, Tu M. Strategies for skeletal muscle targeting in drug discovery. Curr Pharm Des. 2015; 21:1327–36.
Article
115. Rong S, Wang L, Peng Z, Liao Y, Li D, Yang X, et al. The mechanisms and treatments for sarcopenia: could exosomes be a perspective research strategy in the future? J Cachexia Sarcopenia Muscle. 2020; 11:348–65.
Article
116. Li Y, Chen M, Zhao Y, Li M, Qin Y, Cheng S, et al. Advance in drug delivery for ageing skeletal muscle. Front Pharmacol. 2020; 11:1016.
Article
117. Raimondo TM, Mooney DJ. Functional muscle recovery with nanoparticle-directed M2 macrophage polarization in mice. Proc Natl Acad Sci U S A. 2018; 115:10648–53.
Article
118. Ran N, Gao X, Dong X, Li J, Lin C, Geng M, et al. Effects of exosome-mediated delivery of myostatin propeptide on functional recovery of mdx mice. Biomaterials. 2020; 236:119826.
Article
Full Text Links
  • ENM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr