Effect of Urolithin A Supplementation on Muscle Endurance and Mitochondrial Health in Older Adults: A Randomized Clinical Trial

Sophia Liu, Davide D'Amico, Eric Shankland, Saakshi Bhayana, Jose M Garcia, Patrick Aebischer, Chris Rinsch, Anurag Singh, David J Marcinek, Sophia Liu, Davide D'Amico, Eric Shankland, Saakshi Bhayana, Jose M Garcia, Patrick Aebischer, Chris Rinsch, Anurag Singh, David J Marcinek

Abstract

Importance: Aging is associated with a decline in mitochondrial function and reduced exercise capacity. Urolithin A is a natural gut microbiome-derived food metabolite that has been shown to stimulate mitophagy and improve muscle function in older animals and to induce mitochondrial gene expression in older humans.

Objective: To investigate whether oral administration of urolithin A improved the 6-minute walk distance, muscle endurance in hand and leg muscles, and biomarkers associated with mitochondrial and cellular health.

Design, setting, and participants: This double-blind, placebo-controlled randomized clinical trial in adults aged 65 to 90 years was conducted at a medical center and a cancer research center in Seattle, Washington, from March 1, 2018, to July 30, 2020. Muscle fatigue tests and plasma analysis of biomarkers were assessed at baseline, 2 months, and 4 months. Six-minute walk distance and maximal ATP production were assessed using magnetic resonance spectroscopy at baseline and at the end of study at 4 months. The analysis used an intention-to-treat approach.

Interventions: Participants were randomized to receive daily oral supplementation with either 1000 mg urolithin A or placebo for 4 months.

Main outcomes and measures: The primary end point was change from baseline in the 6-minute walk distance and change from baseline to 4 months in maximal ATP production in the hand skeletal muscle. The secondary end points were change in muscle endurance of 2 skeletal muscles (tibialis anterior [TA] in the leg and first dorsal interosseus [FDI] in the hand). Cellular health biomarkers were investigated via plasma metabolomics. Adverse events were recorded and compared between the 2 groups during the intervention period.

Results: A total of 66 participants were randomized to either the urolithin A (n = 33) or the placebo (n = 33) intervention group. These participants had a mean (SD) age of 71.7 (4.94) years, were predominantly women (50 [75.8%]), and were all White individuals. Urolithin A, compared with placebo, significantly improved muscle endurance (ie, increase in the number of muscle contractions until fatigue from baseline) in the FDI and TA at 2 months (urolithin A: FDI, 95.3 [115.5] and TA, 41.4 [65.5]; placebo: FDI, 11.6 [147.4] and TA, 5.7 [127.1]). Plasma levels of several acylcarnitines, ceramides, and C-reactive protein were decreased by urolithin A, compared with placebo, at 4 months (baseline vs 4 mo: urolithin A, 2.14 [2.15] vs 2.07 [1.46]; placebo, 2.17 [2.52] vs 2.65 [1.86]). The mean (SD) increase from baseline in the 6-minute walk distance was 60.8 (67.2) m in the urolithin A group and 42.5 (73.3) m in the placebo group. The mean (SD) change from baseline to 4 months in maximal ATP production in the FDI was 0.07 (0.23) mM/s in the urolithin A group and 0.06 (0.20) mM/s in the placebo group; for the TA, it was -0.03 (0.10) mM/s in the urolithin A group and 0.03 (0.10) mM/s in the placebo group. These results showed no significant improvement with urolithin A supplementation compared with placebo. No statistical differences in adverse events were observed between the 2 groups.

Conclusions and relevance: This randomized clinical trial found that urolithin A supplementation was safe and well tolerated in the assessed population. Although the improvements in the 6-minute walk distance and maximal ATP production in the hand muscle were not significant in the urolithin A group vs the placebo group, long-term urolithin A supplementation was beneficial for muscle endurance and plasma biomarkers, suggesting that urolithin A may counteract age-associated muscle decline; however, future work is needed to confirm this finding.

Trial registration: ClinicalTrials.gov Identifier: NCT03283462.

Conflict of interest statement

Conflict of Interest Disclosures: Dr Liu reported receiving grants from drugs at no cost for research use from Stealth BioTherapeutics and supplements at no cost for research use from AstaReal outside the submitted work. Dr D'Amico reported being an employee of Amazentis SA during the conduct of the study and receiving personal fees from Amazentis SA outside the submitted work. Dr Shankland reported receiving drugs at no cost for research use from Stealth BioTherapeutics and supplements at no cost for research use from AstaReal outside the submitted work. Ms Bhayana reported receiving drugs at no cost for research use from Stealth BioTherapeutics and supplements at no cost for research use from AstaReal outside the submitted work. Dr Garcia reported being a full-time employee of the US government during the conduct of the study. Dr Aebischer reported being the chair of the board and a shareholder of Amazentis SA during the conduct of the study. Dr Rinsch reported being the founder, the chief executive officer, and a board member of Amazentis SA during the conduct of the study; holding a patent for US 9,872,850 issued, a patent for US 10,028,932 issued, a patent for US 10,485,782 issued, a patent for US 11,020,373 issued, a patent for US 10,906,883 issued, a patent for US 20180256538 pending, and a patent for US 20180256539 pending. Dr Singh reported being an employee of Amazentis SA during the conduct of the study and holding a patent for US 20180256538 pending and a patent for US 20180256539 pending. Dr Marcinek reported receiving drugs at no cost for research use from Stealth BioTherapeutics, grants from Boehringer Ingelheim, and supplements at no cost for research use from AstaReal outside the submitted work.

Figures

Figure 1.. CONSORT Diagram of Participant Flow…
Figure 1.. CONSORT Diagram of Participant Flow Through the Study
Figure 2.. Effect of Urolithin A Supplementation…
Figure 2.. Effect of Urolithin A Supplementation on the 6-Minute Walk Distance and Maximal Adenosine Triphosphate (ATP) Production in Hand Muscles and Muscle Endurance
Data were analyzed using an analysis of covariance with 95% CIs for treatment differences. The center horizontal line represents the mean and the top and bottom lines represent the SEM. FDI indicates first dorsal interosseus; TA, tibialis anterior. aP <.01 with post hoc 2-sided Wilcoxon 2-sample test. bP = .05. cP = .07 with post hoc 2-sided Wilcoxon 2-sample test.
Figure 3.. Effect of Urolithin A on…
Figure 3.. Effect of Urolithin A on Biomarkers of Mitochondrial Health and Inflammation
A-F, Data represent geometric mean (95% CI). A 2-way repeated-measures analysis of variance was used. G, Data represent mean (95% CI). An analysis of covariance model was used. BL indicates baseline; CRP indicates C-reactive protein. Light gray circles indicate change in plasma levels in the control group at 2 months; dark blue circles, change in plasma levels in the control group at 4 months; orange circles, change in plasma levels in the urolithin A group at 2 months; bright blue circles, change in plasma levels in the urolithin A group at 4 months. aP <.05. b.05 is less than P < .10.

References

    1. Atella V, Piano Mortari A, Kopinska J, et al. . Trends in age-related disease burden and healthcare utilization. Aging Cell. 2019;18(1):e12861. doi:10.1111/acel.12861
    1. Fulmer T, Reuben DB, Auerbach J, Fick DM, Galambos C, Johnson KS. Actualizing better health and health care for older adults. Health Aff (Millwood). 2021;40(2):219-225. doi:10.1377/hlthaff.2020.01470
    1. von Haehling S, Morley JE, Anker SD. An overview of sarcopenia: facts and numbers on prevalence and clinical impact. J Cachexia Sarcopenia Muscle. 2010;1(2):129-133. doi:10.1007/s13539-010-0014-2
    1. Landi F, Liperoti R, Fusco D, et al. . Prevalence and risk factors of sarcopenia among nursing home older residents. J Gerontol A Biol Sci Med Sci. 2012;67(1):48-55. doi:10.1093/gerona/glr035
    1. Moon HE, Paek SH. Mitochondrial dysfunction in Parkinson’s disease. Exp Neurobiol. 2015;24(2):103-116. doi:10.5607/en.2015.24.2.103
    1. Coen PM, Jubrias SA, Distefano G, et al. . Skeletal muscle mitochondrial energetics are associated with maximal aerobic capacity and walking speed in older adults. J Gerontol A Biol Sci Med Sci. 2013;68(4):447-455. doi:10.1093/gerona/gls196
    1. Conley KE, Jubrias SA, Esselman PC. Oxidative capacity and ageing in human muscle. J Physiol. 2000;526(pt 1):203-210. doi:10.1111/j.1469-7793.2000.t01-1-00203.x
    1. Short KR, Bigelow ML, Kahl J, et al. . Decline in skeletal muscle mitochondrial function with aging in humans. Proc Natl Acad Sci U S A. 2005;102(15):5618-5623. doi:10.1073/pnas.0501559102
    1. Santanasto AJ, Glynn NW, Jubrias SA, et al. . Skeletal muscle mitochondrial function and fatigability in older adults. J Gerontol A Biol Sci Med Sci. 2015;70(11):1379-1385. doi:10.1093/gerona/glu134
    1. Fivenson EM, Lautrup S, Sun N, et al. . Mitophagy in neurodegeneration and aging. Neurochem Int. 2017;109:202-209. doi:10.1016/j.neuint.2017.02.007
    1. Shi R, Guberman M, Kirshenbaum LA. Mitochondrial quality control: the role of mitophagy in aging. Trends Cardiovasc Med. 2018;28(4):246-260. doi:10.1016/j.tcm.2017.11.008
    1. D’Amico D, Andreux PA, Valdés P, Singh A, Rinsch C, Auwerx J. Impact of the natural compound urolithin A on health, disease, and aging. Trends Mol Med. 2021;27(7):687-699. doi:10.1016/j.molmed.2021.04.009
    1. Cerdá B, Periago P, Espín JC, Tomás-Barberán FA. Identification of urolithin A as a metabolite produced by human colon microflora from ellagic acid and related compounds. J Agric Food Chem. 2005;53(14):5571-5576. doi:10.1021/jf050384i
    1. Singh A, D’Amico D, Andreux PA, et al. . Direct supplementation with urolithin A overcomes limitations of dietary exposure and gut microbiome variability in healthy adults to achieve consistent levels across the population. Eur J Clin Nutr. 2021. doi:10.1038/s41430-021-00950-1
    1. Ryu D, Mouchiroud L, Andreux PA, et al. . Urolithin A induces mitophagy and prolongs lifespan in C. elegans and increases muscle function in rodents. Nat Med. 2016;22(8):879-888. doi:10.1038/nm.4132
    1. Luan P, D’Amico D, Andreux PA, et al. . Urolithin A improves muscle function by inducing mitophagy in muscular dystrophy. Sci Transl Med. 2021;13(588):eabb0319. doi:10.1126/scitranslmed.abb0319
    1. Andreux PA, Blanco-Bose W, Ryu D, et al. . The mitophagy activator urolithin A is safe and induces a molecular signature of improved mitochondrial and cellular health in humans. Nat Metab. 2019;1(6):595-603. doi:10.1038/s42255-019-0073-4
    1. World Medical Association . World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310(20):2191-2194. doi:10.1001/jama.2013.281053
    1. Liu SZ, Valencia AP, VanDoren MP, et al. . Astaxanthin supplementation enhances metabolic adaptation with aerobic training in the elderly. Physiol Rep. 2021;9(11):e14887. doi:10.14814/phy2.14887
    1. Roshanravan B, Liu SZ, Ali AS, et al. . In vivo mitochondrial ATP production is improved in older adult skeletal muscle after a single dose of elamipretide in a randomized trial. PLoS One. 2021;16(7):e0253849. doi:10.1371/journal.pone.0253849
    1. Jubrias SA, Crowther GJ, Shankland EG, Gronka RK, Conley KE. Acidosis inhibits oxidative phosphorylation in contracting human skeletal muscle in vivo. J Physiol. 2003;553(pt 2):589-599. doi:10.1113/jphysiol.2003.045872
    1. Steffen TM, Hacker TA, Mollinger L. Age- and gender-related test performance in community-dwelling elderly people: six-minute walk test, Berg balance scale, timed up & go test, and gait speeds. Phys Ther. 2002;82(2):128-137. doi:10.1093/ptj/82.2.128
    1. Bohannon RW, Crouch R. Minimal clinically important difference for change in 6-minute walk test distance of adults with pathology: a systematic review. J Eval Clin Pract. 2017;23(2):377-381. doi:10.1111/jep.12629
    1. Johnson MA, Polgar J, Weightman D, Appleton D. Data on the distribution of fibre types in thirty-six human muscles: an autopsy study. J Neurol Sci. 1973;18(1):111-129. doi:10.1016/0022-510X(73)90023-3
    1. Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci. 2014;69(suppl 1):S4-S9. doi:10.1093/gerona/glu057
    1. Hilvo M, Meikle PJ, Pedersen ER, et al. . Development and validation of a ceramide- and phospholipid-based cardiovascular risk estimation score for coronary artery disease patients. Eur Heart J. 2020;41(3):371-380.
    1. Kurz J, Parnham MJ, Geisslinger G, Schiffmann S. Ceramides as novel disease biomarkers. Trends Mol Med. 2019;25(1):20-32. doi:10.1016/j.molmed.2018.10.009
    1. Lassale C, Batty GD, Steptoe A, et al. . Association of 10-year C-reactive protein trajectories with markers of healthy aging: findings from the English Longitudinal Study of Aging. J Gerontol A Biol Sci Med Sci. 2019;74(2):195-203. doi:10.1093/gerona/gly028

Source: PubMed

3
購読する