Molecular Regulators of Muscle Mass and Mitochondrial Remodeling Are Not Influenced by Testosterone Administration in Young Women

Oscar Horwath, Marcus Moberg, Angelica Lindén Hirschberg, Björn Ekblom, William Apró, Oscar Horwath, Marcus Moberg, Angelica Lindén Hirschberg, Björn Ekblom, William Apró

Abstract

Testosterone (T) administration has previously been shown to improve muscle size and oxidative capacity. However, the molecular mechanisms underlying these adaptations in human skeletal muscle remain to be determined. Here, we examined the effect of moderate-dose T administration on molecular regulators of muscle protein turnover and mitochondrial remodeling in muscle samples collected from young women. Forty-eight healthy, physically active, young women (28 ± 4 years) were assigned in a random double-blind fashion to receive either T (10 mg/day) or placebo for 10-weeks. Muscle biopsies collected before and after the intervention period were divided into sub-cellular fractions and total protein levels of molecular regulators of muscle protein turnover and mitochondrial remodeling were analyzed using Western blotting. T administration had no effect on androgen receptor or 5α-reductase levels, nor on proteins involved in the mTORC1-signaling pathway (mTOR, S6K1, eEF2 and RPS6). Neither did it affect the abundance of proteins associated with proteasomal protein degradation (MAFbx, MuRF-1 and UBR5) and autophagy-lysosomal degradation (AMPK, ULK1 and p62). T administration also had no effect on proteins in the mitochondria enriched fraction regulating mitophagy (Beclin, BNIP3, LC3B-I, LC3B-II and LC3B-II/I ratio) and morphology (Mitofilin), and it did not alter the expression of mitochondrial fission- (FIS1 and DRP1) or fusion factors (OPA1 and MFN2). In summary, these data indicate that improvements in muscle size and oxidative capacity in young women in response to moderate-dose T administration cannot be explained by alterations in total expression of molecular factors known to regulate muscle protein turnover or mitochondrial remodeling.

Trial registration: ClinicalTrials.gov NCT03210558.

Keywords: androgen receptor; fission; fusion; mTORC1-signaling; ubiquitin-proteasome system.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Horwath, Moberg, Hirschberg, Ekblom and Apró.

Figures

Figure 1
Figure 1
Protein levels of androgen receptor (A), 5α-reductase (B), mTOR (C), S6K1 (D), eEF2 (E) and RPS6 (F) before (black dots) and after the intervention (red dots). Representative blots for each individual protein target are shown above each graph. Loading control is represented by a band at ~ 95 kDa from the corresponding total protein stain (Memcode™). The values presented are means ± SD and individual data points from 18 and 16 individuals (DF 32) in the placebo group and T group, respectively. The ANOVA revealed a significant main effect of time with respect to changes in protein levels for 5α-reductase and a significant main effect of time and group for RPS6.
Figure 2
Figure 2
Protein levels of MAFbx (A), MuRF-1 (B), UBR5 (C), AMPK (D), ULK1 (E) and p62 (F) before (black dots) and after the intervention (red dots). Representative blots for each individual protein target are shown above each graph. Loading control is represented by a band at ~ 95 kDa from the corresponding total protein stain (Memcode™). The values presented are means ± SD and individual data points from 18 and 16 individuals (DF 32) in the placebo group and T group, respectively.
Figure 3
Figure 3
Protein levels of Beclin (A), BNIP3 (B), LC3B-I (C), LC3B-II (D), LC3B-II/I ratio (E) and Mitofilin (F) in the mitochondrial fraction before (black dots) and after the intervention (red dots), as well as the assessment of mitochondrial fraction purity (G). Representative blots for each individual protein target and Porin are shown above each graph. The values presented are means ± SD and individual data points from 22 and 20 individuals (DF 40) in the placebo group and T group, respectively. The ANOVA revealed a significant main effect of time with respect to changes in protein levels for BNIP3 and LC3B-II. For illustrative purposes, two subjects from the placebo group displaying extreme values were removed from (A) (pre-post; 6.7 to 18.5 and 12.3 to 23.6, respectively) and one subject from the placebo group was removed from (D) (pre-post; 6.1 to 19.4), but these values were included in the statistical analysis.
Figure 4
Figure 4
Protein levels of OPA1 (A), MFN2 (B), FIS1 (C), and DRP1 (D) in the mitochondrial fraction before (black dots) and after the intervention (red dots). Representative blots for each individual protein target and Porin are shown above each graph. The values presented are means ± SD and individual data points from 22 and 20 individuals (DF 40) in the placebo group and T group, respectively. For illustrative purposes, one subject from the placebo group displaying extreme values was removed from (A) (pre-post; 6.1 to 19.4), but these values were included in the statistical analysis.

References

    1. Sinha-Hikim I, Artaza J, Woodhouse L, Gonzalez-Cadavid N, Singh AB, Lee MI, et al. . Testosterone-Induced Increase in Muscle Size in Healthy Young Men Is Associated With Muscle Fiber Hypertrophy. Am J Physiol Endocrinol Metab (2002) 283(1):E154–64. doi: 10.1152/ajpendo.00502.2001
    1. Sinha-Hikim I, Cornford M, Gaytan H, Lee ML, Bhasin S. Effects of Testosterone Supplementation on Skeletal Muscle Fiber Hypertrophy and Satellite Cells in Community-Dwelling Older Men. J Clin Endocrinol Metab (2006) 91(8):3024–33. doi: 10.1210/jc.2006-0357
    1. Bhasin S, Storer TW, Berman N, Callegari C, Clevenger B, Phillips J, et al. . The Effects of Supraphysiologic Doses of Testosterone on Muscle Size and Strength in Normal Men. N Engl J Med (1996) 335(1):1–7. doi: 10.1056/NEJM199607043350101
    1. Hirschberg AL, Elings Knutsson J, Helge T, Godhe M, Ekblom M, Bermon S, et al. . Effects of Moderately Increased Testosterone Concentration on Physical Performance in Young Women: A Double Blind, Randomised, Placebo Controlled Study. Br J Sports Med (2020) 54(10):599–604. doi: 10.1136/bjsports-2018-100525
    1. Huang G, Basaria S, Travison TG, Ho MH, Davda M, Mazer NA, et al. . Testosterone Dose-Response Relationships in Hysterectomized Women With or Without Oophorectomy: Effects on Sexual Function, Body Composition, Muscle Performance and Physical Function in a Randomized Trial. Menopause (2014) 21(6):612–23. doi: 10.1097/GME.0000000000000093
    1. Smith GI, Yoshino J, Reeds DN, Bradley D, Burrows RE, Heisey HD, et al. . Testosterone and Progesterone, But Not Estradiol, Stimulate Muscle Protein Synthesis in Postmenopausal Women. J Clin Endocrinol Metab (2014) 99(1):256–65. doi: 10.1210/jc.2013-2835
    1. Dickinson JM, Fry CS, Drummond MJ, Gundermann DM, Walker DK, Glynn EL, et al. . Mammalian Target of Rapamycin Complex 1 Activation Is Required for the Stimulation of Human Skeletal Muscle Protein Synthesis by Essential Amino Acids. J Nutr (2011) 141(5):856–62. doi: 10.3945/jn.111.139485
    1. Drummond MJ, Fry CS, Glynn EL, Dreyer HC, Dhanani S, Timmerman KL, et al. . Rapamycin Administration in Humans Blocks the Contraction-Induced Increase in Skeletal Muscle Protein Synthesis. J Physiol (2009) 587(Pt 7):1535–46. doi: 10.1113/jphysiol.2008.163816
    1. Rossetti ML, Steiner JL, Gordon BS. Androgen-Mediated Regulation of Skeletal Muscle Protein Balance. Mol Cell Endocrinol (2017) 447:35–44. doi: 10.1016/j.mce.2017.02.031
    1. Basualto-Alarcon C, Jorquera G, Altamirano F, Jaimovich E, Estrada M. Testosterone Signals Through mTOR and Androgen Receptor to Induce Muscle Hypertrophy. Med Sci Sports Exerc (2013) 45(9):1712–20. doi: 10.1249/MSS.0b013e31828cf5f3
    1. Wu Y, Bauman WA, Blitzer RD, Cardozo C. Testosterone-Induced Hypertrophy of L6 Myoblasts Is Dependent Upon Erk and mTOR. Biochem Biophys Res Commun (2010) 400(4):679–83. doi: 10.1016/j.bbrc.2010.08.127
    1. White JP, Gao S, Puppa MJ, Sato S, Welle SL, Carson JA. Testosterone Regulation of Akt/mTORC1/FoxO3a Signaling in Skeletal Muscle. Mol Cell Endocrinol (2013) 365(2):174–86. doi: 10.1016/j.mce.2012.10.019
    1. Howard EE, Margolis LM, Berryman CE, Lieberman HR, Karl JP, Young AJ, et al. . Testosterone Supplementation Up-Regulates Androgen Receptor Expression and Translational Capacity During Severe Energy Deficit. Am J Physiol Endocrinol Metab (2020) 319(4):E678–88. doi: 10.1152/ajpendo.00157.2020
    1. Gharahdaghi N, Rudrappa S, Brook MS, Idris I, Crossland H, Hamrock C, et al. . Testosterone Therapy Induces Molecular Programming Augmenting Physiological Adaptations to Resistance Exercise in Older Men. J Cachexia Sarcopenia Muscle (2019) 10(6):1276–94. doi: 10.1002/jcsm.12472
    1. Brook MS, Wilkinson DJ, Phillips BE, Perez-Schindler J, Philp A, Smith K, et al. . Skeletal Muscle Homeostasis and Plasticity in Youth and Ageing: Impact of Nutrition and Exercise. Acta Physiol (Oxf) (2016) 216(1):15–41. doi: 10.1111/apha.12532
    1. Ferrando AA, Sheffield-Moore M, Wolf SE, Herndon DN, Wolfe RR. Testosterone Administration in Severe Burns Ameliorates Muscle Catabolism. Crit Care Med (2001) 29(10):1936–42. doi: 10.1097/00003246-200110000-00015
    1. Ferrando AA, Sheffield-Moore M, Yeckel CW, Gilkison C, Jiang J, Achacosa A, et al. . Testosterone Administration to Older Men Improves Muscle Function: Molecular and Physiological Mechanisms. Am J Physiol Endocrinol Metab (2002) 282(3):E601–7. doi: 10.1152/ajpendo.00362.2001
    1. Ferrando AA, Sheffield-Moore M, Paddon-Jones D, Wolfe RR, Urban RJ. Differential Anabolic Effects of Testosterone and Amino Acid Feeding in Older Men. J Clin Endocrinol Metab (2003) 88(1):358–62. doi: 10.1210/jc.2002-021041
    1. Sandri M. Protein Breakdown in Muscle Wasting: Role of Autophagy-Lysosome and Ubiquitin-Proteasome. Int J Biochem Cell Biol (2013) 45(10):2121–9. doi: 10.1016/j.biocel.2013.04.023
    1. Bodine SC, Baehr LM. Skeletal Muscle Atrophy and the E3 Ubiquitin Ligases MuRF1 and MAFbx/atrogin-1. Am J Physiol Endocrinol Metab (2014) 307(6):E469–84. doi: 10.1152/ajpendo.00204.2014
    1. Bodine SC, Latres E, Baumhueter S, Lai VK, Nunez L, Clarke BA, et al. . Identification of Ubiquitin Ligases Required for Skeletal Muscle Atrophy. Science (2001) 294(5547):1704–8. doi: 10.1126/science.1065874
    1. Kruse R, Petersson SJ, Christensen LL, Kristensen JM, Sabaratnam R, Ortenblad N, et al. . Effect of Long-Term Testosterone Therapy on Molecular Regulators of Skeletal Muscle Mass and Fibre-Type Distribution in Aging Men With Subnormal Testosterone. Metabolism (2020) 154347. doi: 10.1016/j.metabol.2020.154347
    1. Ferrando AA, Tipton KD, Doyle D, Phillips SM, Cortiella J, Wolfe RR. Testosterone Injection Stimulates Net Protein Synthesis But Not Tissue Amino Acid Transport. Am J Physiol (1998) 275(5):E864–71. doi: 10.1152/ajpendo.1998.275.5.E864
    1. Serra C, Sandor NL, Jang H, Lee D, Toraldo G, Guarneri T, et al. . The Effects of Testosterone Deprivation and Supplementation on Proteasomal and Autophagy Activity in the Skeletal Muscle of the Male Mouse: Differential Effects on High-Androgen Responder and Low-Androgen Responder Muscle Groups. Endocrinology (2013) 154(12):4594–606. doi: 10.1210/en.2013-1004
    1. Steiner JL, Fukuda DH, Rossetti ML, Hoffman JR, Gordon BS. Castration Alters Protein Balance After High-Frequency Muscle Contraction. J Appl Physiol (2017) 122(2):264–72. doi: 10.1152/japplphysiol.00740.2016
    1. Cardinale DA, Horwath O, Elings-Knutsson J, Helge T, Godhe M, Bermon S, et al. . Enhanced Skeletal Muscle Oxidative Capacity and Capillary-To-Fiber Ratio Following Moderately Increased Testosterone Exposure in Young Healthy Women. Front Physiol (2020) 11:585490. doi: 10.3389/fphys.2020.585490
    1. Rossetti ML, Esser KA, Lee C, Tomko RJ, Jr., Eroshkin AM, Gordon BS. Disruptions to the Limb Muscle Core Molecular Clock Coincide With Changes in Mitochondrial Quality Control Following Androgen Depletion. Am J Physiol Endocrinol Metab (2019) 317(4):E631–E45. doi: 10.1152/ajpendo.00177.2019
    1. Rossetti ML, Steiner JL, Gordon BS. Increased Mitochondrial Turnover in the Skeletal Muscle of Fasted, Castrated Mice Is Related to the Magnitude of Autophagy Activation and Muscle Atrophy. Mol Cell Endocrinol (2018) 473:178–85. doi: 10.1016/j.mce.2018.01.017
    1. Guo W, Wong S, Li M, Liang W, Liesa M, Serra C, et al. . Testosterone Plus Low-Intensity Physical Training in Late Life Improves Functional Performance, Skeletal Muscle Mitochondrial Biogenesis, and Mitochondrial Quality Control in Male Mice. PloS One (2012) 7(12):e51180. doi: 10.1371/journal.pone.0051180
    1. Petersson SJ, Christensen LL, Kristensen JM, Kruse R, Andersen M, Hojlund K. Effect of Testosterone on Markers of Mitochondrial Oxidative Phosphorylation and Lipid Metabolism in Muscle of Aging Men With Subnormal Bioavailable Testosterone. Eur J Endocrinol (2014) 171(1):77–88. doi: 10.1530/EJE-14-0006
    1. Horwath O, Apro W, Moberg M, Godhe M, Helge T, Ekblom MM, et al. . Fiber Type-Specific Hypertrophy and Increased Capillarization in Skeletal Muscle Following Testosterone Administration in Young Women. J Appl Physiol (2020) 1985). doi: 10.1152/japplphysiol.00893.2019
    1. Ekblom B. The Muscle Biopsy Technique. Historical and Methodological Considerations. Scand J Med Sci Sports (2017) 27(5):458–61. doi: 10.1111/sms.12808
    1. Schwalm C, Deldicque L, Francaux M. Lack of Activation of Mitophagy During Endurance Exercise in Human. Med Sci Sports Exerc (2017) 49(8):1552–61. doi: 10.1249/MSS.0000000000001256
    1. Kadi F, Bonnerud P, Eriksson A, Thornell LE. The Expression of Androgen Receptors in Human Neck and Limb Muscles: Effects of Training and Self-Administration of Androgenic-Anabolic Steroids. Histochem Cell Biol (2000) 113(1):25–9. doi: 10.1007/s004180050003
    1. Sinha-Hikim I, Taylor WE, Gonzalez-Cadavid NF, Zheng W, Bhasin S. Androgen Receptor in Human Skeletal Muscle and Cultured Muscle Satellite Cells: Up-Regulation by Androgen Treatment. J Clin Endocrinol Metab (2004) 89(10):5245–55. doi: 10.1210/jc.2004-0084
    1. Singh R, Artaza JN, Taylor WE, Gonzalez-Cadavid NF, Bhasin S. Androgens Stimulate Myogenic Differentiation and Inhibit Adipogenesis in C3H 10T1/2 Pluripotent Cells Through an Androgen Receptor-Mediated Pathway. Endocrinology (2003) 144(11):5081–8. doi: 10.1210/en.2003-0741
    1. Morton RW, Sato K, Gallaugher MPB, Oikawa SY, McNicholas PD, Fujita S, et al. . Muscle Androgen Receptor Content But Not Systemic Hormones Is Associated With Resistance Training-Induced Skeletal Muscle Hypertrophy in Healthy, Young Men. Front Physiol (2018) 9:1373. doi: 10.3389/fphys.2018.01373
    1. Ferry A, Schuh M, Parlakian A, Mgrditchian T, Valnaud N, Joanne P, et al. . Myofiber Androgen Receptor Promotes Maximal Mechanical Overload-Induced Muscle Hypertrophy and Fiber Type Transition in Male Mice. Endocrinology (2014) 155(12):4739–48. doi: 10.1210/en.2014-1195
    1. Sheffield-Moore M, Paddon-Jones D, Casperson SL, Gilkison C, Volpi E, Wolf SE, et al. . Androgen Therapy Induces Muscle Protein Anabolism in Older Women. J Clin Endocrinol Metab (2006) 91(10):3844–9. doi: 10.1210/jc.2006-0588
    1. Nicoll JX, Fry AC, Mosier EM. Sex-Based Differences in Resting MAPK, Androgen, and Glucocorticoid Receptor Phosphorylation in Human Skeletal Muscle. Steroids (2019) 141:23–9. doi: 10.1016/j.steroids.2018.11.004
    1. MacLean HE, Chiu WS, Notini AJ, Axell AM, Davey RA, McManus JF, et al. . Impaired Skeletal Muscle Development and Function in Male, But Not Female, Genomic Androgen Receptor Knockout Mice. FASEB J (2008) 22(8):2676–89. doi: 10.1096/fj.08-105726
    1. Lakshman KM, Kaplan B, Travison TG, Basaria S, Knapp PE, Singh AB, et al. . The Effects of Injected Testosterone Dose and Age on the Conversion of Testosterone to Estradiol and Dihydrotestosterone in Young and Older Men. J Clin Endocrinol Metab (2010) 95(8):3955–64. doi: 10.1210/jc.2010-0102
    1. Bhasin S, Travison TG, Storer TW, Lakshman K, Kaushik M, Mazer NA, et al. . Effect of Testosterone Supplementation With and Without a Dual 5alpha-Reductase Inhibitor on Fat-Free Mass in Men With Suppressed Testosterone Production: A Randomized Controlled Trial. JAMA (2012) 307(9):931–9. doi: 10.1001/jama.2012.227
    1. Alexander SE, Pollock AC, Lamon S. The Effect of Sex Hormones on Skeletal Muscle Adaptation in Females. Eur J Sport Sci (2021), 1–11. doi: 10.1080/17461391.2021.1921854
    1. Baehr LM, Tunzi M, Bodine SC. Muscle Hypertrophy Is Associated With Increases in Proteasome Activity That Is Independent of MuRF1 and MAFbx Expression. Front Physiol (2014) 5:69. doi: 10.3389/fphys.2014.00069
    1. Tipton KD, Hamilton DL, Gallagher IJ. Assessing the Role of Muscle Protein Breakdown in Response to Nutrition and Exercise in Humans. Sports Med (2018) 48(Suppl 1):53–64. doi: 10.1007/s40279-017-0845-5
    1. Sandri M. Autophagy in Skeletal Muscle. FEBS Lett (2010) 584(7):1411–6. doi: 10.1016/j.febslet.2010.01.056
    1. Drake JC, Yan Z. Mitophagy in Maintaining Skeletal Muscle Mitochondrial Proteostasis and Metabolic Health With Ageing. J Physiol (2017) 595(20):6391–9. doi: 10.1113/JP274337
    1. Guan Y, Drake JC, Yan Z. Exercise-Induced Mitophagy in Skeletal Muscle and Heart. Exerc Sport Sci Rev (2019) 47(3):151–6. doi: 10.1249/JES.0000000000000192
    1. Rossetti ML, Gordon BS. The Role of Androgens in the Regulation of Muscle Oxidative Capacity Following Aerobic Exercise Training. Appl Physiol Nutr Metab (2017) 42(9):1001–7. doi: 10.1139/apnm-2017-0230
    1. Balan E, Schwalm C, Naslain D, Nielens H, Francaux M, Deldicque L. Regular Endurance Exercise Promotes Fission, Mitophagy, and Oxidative Phosphorylation in Human Skeletal Muscle Independently of Age. Front Physiol (2019) 10:1088. doi: 10.3389/fphys.2019.01088
    1. Mesquita PHC, Lamb DA, Parry HA, Moore JH, Smith MA, Vann CG, et al. . Acute and Chronic Effects of Resistance Training on Skeletal Muscle Markers of Mitochondrial Remodeling in Older Adults. Physiol Rep (2020) 8(15):e14526. doi: 10.14814/phy2.14526

Source: PubMed

3
Subskrybuj