Skip to main content
Log in

Potential mechanisms of uremic muscle wasting and the protective role of the mitochondria-targeted antioxidant Mito-TEMPO

  • Nephrology - Original Paper
  • Published:
International Urology and Nephrology Aims and scope Submit manuscript

Abstract

Background

Muscle wasting is common in patients with chronic kidney disease (CKD). Many studies report that mitochondrial dysfunction and endoplasmic reticulum (ER) stress are involved in the development of muscle wasting. However, treatment approaches to protect against muscle wasting are limited. In this study, we investigated the benefits and potential mechanism of Mito-TEMPO, a mitochondria-targeted antioxidant on uremic-induced muscle wasting.

Methods

Mice were randomly divided into four groups as follows: control group, CKD group, CKD + Mito-TEMPO group, and Mito-TEMPO group. Renal injury was assessed by measurement of serum creatinine and BUN along with PAS and Masson’s staining. Bodyweight, gastrocnemius muscle mass, grip strength, and myofiber cross-sectional areas were investigated to evaluate muscle atrophy. Muscle protein synthesis and proteolysis were evaluated by Western blot and real-time PCR. Inflammatory cytokines including TNF-α, IL-6, IL-1β, and MCP-1 were measured by ELISA kits. Oxidative stress markers such as SOD2 activity and MDA level in gastrocnemius muscle tissue were measured by colorimetric assay. Mitochondrial dysfunction was evaluated by transmission electron microscopy and real-time PCR. ER stress was evaluated by Western blot.

Results

Impaired renal function was significantly restored by Mito-TEMPO treatment. Severe muscle atrophy was observed in muscle tissues of CKD mice along with increased inflammatory factors, oxidative stress markers, mitochondrial dysfunction, and ER stress. However, these effects were significantly attenuated with Mito-TEMPO treatment.

Conclusions

Mito-TEMPO improved muscle wasting in CKD mice possibly through alleviating mitochondrial dysfunction and endoplasmic reticulum stress, providing a potential new therapeutic approach for preventing muscle wasting in chronic kidney disease.

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.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Stevens PE, Levin A (2013) Evaluation and management of chronic kidney disease: synopsis of the kidney disease: improving global outcomes 2012 clinical practice guideline. Ann Intern Med 158(11):825–830

    Article  PubMed  Google Scholar 

  2. Hoerger TJ, Simpson SA, Yarnoff BO, Pavkov ME, Rios BN, Saydah SH, Williams DE, Zhuo X (2015) The future burden of CKD in the United States: a simulation model for the CDC CKD initiative. Am J Kidney Dis 65(3):403–411

    Article  PubMed  Google Scholar 

  3. Workeneh BT, Mitch WE (1132S) Review of muscle wasting associated with chronic kidney disease. Am J Clin Nutr 91(4):1128S–1132S

    Article  CAS  PubMed  Google Scholar 

  4. Chao CT, Tang CH, Cheng RW, Wang MY, Hung KY (2017) Protein-energy wasting significantly increases healthcare utilization and costs among patients with chronic kidney disease: a propensity-score matched cohort study. Curr Med Res Opin 33(9):1705–1713

    Article  CAS  PubMed  Google Scholar 

  5. Wang XH, Mitch WE (2014) Mechanisms of muscle wasting in chronic kidney disease. Nat Rev Nephrol 10(9):504–516

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Wang XH, Mitch WE (2013) Muscle wasting from kidney failure-a model for catabolic conditions. Int J Biochem Cell Biol 45(10):2230–2238

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Thomas SS, Mitch WE (2013) Mechanisms stimulating muscle wasting in chronic kidney disease: the roles of the ubiquitin-proteasome system and myostatin. Clin Exp Nephrol 17(2):174–182

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Du J, Hu Z, Mitch WE (2005) Molecular mechanisms activating muscle protein degradation in chronic kidney disease and other catabolic conditions. Eur J Clin Invest 35(3):157–163

    Article  CAS  PubMed  Google Scholar 

  9. Pieczenik SR, Neustadt J (2007) Mitochondrial dysfunction and molecular pathways of disease. Exp Mol Pathol 83(1):84–92

    Article  CAS  PubMed  Google Scholar 

  10. Wang D, Chen J, Liu X, Zheng P, Song G, Yi T, Li S (2017) A Chinese herbal formula, Jian-Pi-Yi-Shen decoction, improves muscle atrophy via regulating mitochondrial quality control process in 5/6 nephrectomised rats. Sci Rep 7(1):9253

    Article  PubMed  PubMed Central  Google Scholar 

  11. Tamaki M, Miyashita K, Wakino S, Mitsuishi M, Hayashi K, Itoh H (2014) Chronic kidney disease reduces muscle mitochondria and exercise endurance and its exacerbation by dietary protein through inactivation of pyruvate dehydrogenase. Kidney Int 85(6):1330–1339

    Article  CAS  PubMed  Google Scholar 

  12. Su Z, Klein JD, Du J, Franch HA, Zhang L, Hassounah F, Hudson MB, Wang XH (2017) Chronic kidney disease induces autophagy leading to dysfunction of mitochondria in skeletal muscle. Am J Physiol Renal Physiol 312(6):F1128–F1140

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Enoki Y, Watanabe H, Arake R, Fujimura R, Ishiodori K, Imafuku T, Nishida K, Sugimoto R, Nagao S, Miyamura S, Ishima Y, Tanaka M, Matsushita K, Komaba H, Fukagawa M, Otagiri M, Maruyama T (2017) Potential therapeutic interventions for chronic kidney disease-associated sarcopenia via indoxyl sulfate-induced mitochondrial dysfunction. J Cachexia Sarcopenia Muscle 8(5):735–747

    Article  PubMed  PubMed Central  Google Scholar 

  14. Cao SS, Kaufman RJ (2014) Endoplasmic reticulum stress and oxidative stress in cell fate decision and human disease. Antioxid Redox Signal 21(3):396–413

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Afroze D, Kumar A (2019) ER stress in skeletal muscle remodeling and myopathies. FEBS J 286(2):379–398

    Article  CAS  PubMed  Google Scholar 

  16. Pluquet O, Pourtier A, Abbadie C (2015) The unfolded protein response and cellular senescence. A review in the theme: cellular mechanisms of endoplasmic reticulum stress signaling in health and disease. Am J Physiol Cell Physiol 308(6):C415–C425

    Article  CAS  PubMed  Google Scholar 

  17. Bohnert KR, McMillan JD, Kumar A (2018) Emerging roles of ER stress and unfolded protein response pathways in skeletal muscle health and disease. J Cell Physiol 233(1):67–78

    Article  CAS  PubMed  Google Scholar 

  18. Reddy SS, Shruthi K, Prabhakar YK, Sailaja G, Reddy GB (2018) Implication of altered ubiquitin-proteasome system and ER stress in the muscle atrophy of diabetic rats. Arch Biochem Biophys 639:16–25

    Article  CAS  PubMed  Google Scholar 

  19. Ma L, Chu W, Chai J, Shen C, Li D, Wang X (2017) ER stress and subsequent activated calpain play a pivotal role in skeletal muscle wasting after severe burn injury. PLoS ONE 12(10):e186128

    Google Scholar 

  20. Paul PK, Bhatnagar S, Mishra V, Srivastava S, Darnay BG, Choi Y, Kumar A (2012) The E3 ubiquitin ligase TRAF6 intercedes in starvation-induced skeletal muscle atrophy through multiple mechanisms. Mol Cell Biol 32(7):1248–1259

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Bohnert KR, Gallot YS, Sato S, Xiong G, Hindi SM, Kumar A (2016) Inhibition of ER stress and unfolding protein response pathways causes skeletal muscle wasting during cancer cachexia. FASEB J 30(9):3053–3068

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Dikalov S (2011) Cross talk between mitochondria and NADPH oxidases. Free Radic Biol Med 51(7):1289–1301

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Cheung WW, Ding W, Gunta SS, Gu Y, Tabakman R, Klapper LN, Gertler A, Mak RH (2014) A pegylated leptin antagonist ameliorates CKD-associated cachexia in mice. J Am Soc Nephrol 25(1):119–128

    Article  CAS  PubMed  Google Scholar 

  24. Ding W, Yang L, Zhang M, Gu Y (2012) Chronic inhibition of nuclear factor kappa B attenuates aldosterone/salt-induced renal injury. Life Sci 90(15–16):600–606

    Article  CAS  PubMed  Google Scholar 

  25. Remuzzi G, Zoja C, Gagliardini E, Corna D, Abbate M, Benigni A (1999) Combining an antiproteinuric approach with mycophenolate mofetil fully suppresses progressive nephropathy of experimental animals. J Am Soc Nephrol 10(7):1542–1549

    CAS  PubMed  Google Scholar 

  26. Takeshita H, Yamamoto K, Nozato S, Inagaki T, Tsuchimochi H, Shirai M, Yamamoto R, Imaizumi Y, Hongyo K, Yokoyama S, Takeda M, Oguro R, Takami Y, Itoh N, Takeya Y, Sugimoto K, Fukada SI, Rakugi H (2017) Modified forelimb grip strength test detects aging-associated physiological decline in skeletal muscle function in male mice. Sci Rep 7:42323

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Fanzani A, Conraads VM, Penna F, Martinet W (2012) Molecular and cellular mechanisms of skeletal muscle atrophy: an update. J Cachexia Sarcopenia Muscle 3(3):163–179

    Article  PubMed  PubMed Central  Google Scholar 

  28. Powers SK, Morton AB, Ahn B, Smuder AJ (2016) Redox control of skeletal muscle atrophy. Free Radic Biol Med 98:208–217

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Abrigo J, Elorza AA, Riedel CA, Vilos C, Simon F, Cabrera D, Estrada L, Cabello-Verrugio C (2018) Role of oxidative stress as key regulator of muscle wasting during cachexia. Oxid Med Cell Longev 2018:2063179

    Article  PubMed  PubMed Central  Google Scholar 

  30. Lee H, Lim Y (2018) Tocotrienol-rich fraction supplementation reduces hyperglycemia-induced skeletal muscle damage through regulation of insulin signaling and oxidative stress in type 2 diabetic mice. J Nutr Biochem 57:77–85

    Article  CAS  PubMed  Google Scholar 

  31. Tang P, Gu Y, Gu JM, Xie ZA, Xu JQ, Zhao XD, Huang KM, Wang JY, Jiang XS, Fan SW, Hu ZJ (2018) Ascorbic acid attenuates multifidus muscles injury and atrophy after posterior lumbar spine surgery by suppressing inflammation and oxidative stress in a rat model. Spine (Phile Pa 1976) 43:1249–1259

    Article  Google Scholar 

  32. Powers SK, Smuder AJ, Judge AR (2012) Oxidative stress and disuse muscle atrophy: cause or consequence? Curr Opin Clin Nutr Metab Care 15(3):240–245

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Choi WH, Son HJ, Jang YJ, Ahn J, Jung CH, Ha TY (2017) Apigenin ameliorates the obesity-induced skeletal muscle atrophy by attenuating mitochondrial dysfunction in the muscle of obese mice. Mol Nutr Food Res. https://doi.org/10.1002/mnfr.201700218

    Article  PubMed  Google Scholar 

  34. Wang D, Wei L, Yang Y, Liu H (2018) Dietary supplementation with ketoacids protects against CKD-induced oxidative damage and mitochondrial dysfunction in skeletal muscle of 5/6 nephrectomised rats. Skelet Muscle 8(1):18

    Article  PubMed  PubMed Central  Google Scholar 

  35. Moungjaroen J, Nimmannit U, Callery PS, Wang L, Azad N, Lipipun V, Chanvorachote P, Rojanasakul Y (2006) Reactive oxygen species mediate caspase activation and apoptosis induced by lipoic acid in human lung epithelial cancer cells through Bcl-2 down-regulation. J Pharmacol Exp Ther 319(3):1062–1069

    Article  CAS  PubMed  Google Scholar 

  36. Eisner V, Csordas G, Hajnoczky G (2013) Interactions between sarco-endoplasmic reticulum and mitochondria in cardiac and skeletal muscle pivotal roles in Ca(2)(+) and reactive oxygen species signaling. J Cell Sci 126(Pt 14):2965–2978

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lee H, Lim JY, Choi SJ (2017) Oleate prevents palmitate-induced atrophy via modulation of mitochondrial ROS production in skeletal myotubes. Oxid Med Cell Longev 2017:2739721

    PubMed  PubMed Central  Google Scholar 

  38. Salazar G, Huang J, Feresin RG, Zhao Y, Griendling KK (2017) Zinc regulates Nox1 expression through a NF-kappaB and mitochondrial ROS dependent mechanism to induce senescence of vascular smooth muscle cells. Free Radic Biol Med 108:225–235

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This study was funded by “The Belt and Road international cooperation project”(18410741500), “interdisciplinary program of Shanghai Jiao Tong University” (YG2017MS07), and “the distinguished Young Scholar of Ninth people’s hospital” (jyyq09201701).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Yingdeng Wang or Wei Ding.

Ethics declarations

Conflict of interest

The authors declare that there are no conflicts of interest.

Ethical approval

All animal experiments were approved by the Animal Care Committee at Shanghai Jiao Tong University and carried out in accordance with institutional guidelines.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, Y., Perumal, E., Bi, X. et al. Potential mechanisms of uremic muscle wasting and the protective role of the mitochondria-targeted antioxidant Mito-TEMPO. Int Urol Nephrol 52, 1551–1561 (2020). https://doi.org/10.1007/s11255-020-02508-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11255-020-02508-9

Keywords

Navigation