- Brief Report
- Open access
- Published:
The polyphenol metabolite urolithin A suppresses myostatin expression and augments glucose uptake in human skeletal muscle cells
Nutrition & Metabolism volume 22, Article number: 12 (2025)
Abstract
Purpose
Polyphenolic plant extracts have demonstrated anti-inflammatory and anti-catabolic effects in vitro, however their meaningful translation into humans remains elusive. Urolithin A (UA), a gut-derived metabolite of ellagitannins, has shown promise for improving muscle function and metabolic health in rodent models. This study aimed to explore the impact of UA on insulin and anabolic sensitivity in human skeletal muscle cells.
Methods
Primary human myogenic cultures were derived from skeletal muscle biopsies of eight healthy adults. After differentiation, myotubes were treated with 0.002, 1 and 50 µM UA or vehicle for 24 h. Cell viability was assessed using a resazurin assay. Basal and insulin-stimulated glucose uptake was measured using tritiated deoxy-D-glucose, whilst amino acid-stimulated protein synthesis was estimated using the surface sensing of translation (SuNSET) technique. Expression of myostatin and glucose transporters was quantified via real-time PCR.
Results
UA treatment at ≤ 50 µM did not compromise cell viability. Treatment with 50 µM UA enhanced both basal- and insulin-stimulated glucose uptake by 21% (P < 0.05) and 24% (P < 0.01), respectively, compared to vehicle and was accompanied by a 1.8-fold upregulation of GLUT4 expression (P < 0.01). 50 µM UA reduced myostatin (MSTN) expression by 14% (P < 0.01) but did not alter amino acid-stimulated global cell protein synthesis.
Conclusion
This study provides evidence of UA’s metabolic benefits in primary human myotubes, notably improving basal- and insulin-stimulated glucose uptake and supressing MSTN expression. These findings suggest UA could be an effective nutraceutical for mitigating insulin resistance and warrants further investigation.
Introduction
According to estimates from The World Health Organization (WHO), non-communicable diseases (NCD) account for 74% of all deaths globally; cardiovascular diseases, cancers, chronic respiratory diseases and diabetes collectively comprising over 80% of all premature NCD deaths [1]. Raised blood pressure overweight/obesity, hyperglycaemia and hyperlipidaemia are the major physiological risk factors for these most common NCDs. Commonly underlying such diseases is a characteristic, subclinical, systemic inflammation, which is associated with a deleterious metabolic cascade involving cellular oxidative stress, atherosclerotic processes and insulin resistance [2]. The identification and exploration of so-called nutraceuticals – naturally-occurring, bioactive compounds with purported health benefits beyond the provision of energy substrates – is growing industry and field of research interested in the prevention of NCD.
Of particular interest, plants produce a class of phytochemicals called polyphenols, some of which are purported to confer health benefits, particularly with regard to NCD and the pathophysiological process of ageing [3]. Certain polyphenolic plant extracts have demonstrated anti-inflammatory and anti-catabolic effects on skeletal muscle cells in both in vitro and in vivo in rodent models [4,5,6,7], however their meaningful translation into humans remains elusive [8]. Amongst the most studied polyphenolic compounds are ellagitannins (ETs) and ellagic acid (EA), which are present in fruits, nuts and seeds, such as pomegranates and walnuts. ETs and EA are metabolised by the microbiota of the gut, with ETs first being hydrolysed into EAs in the upper gastrointestinal tract by tannases [9]. Whilst ETs and EA can be readily obtained from the diet, they exhibit poor pharmacokinetic profiles, low water solubility and intestinal permeability, and a short plasma half-life due to rapid elimination by first-pass metabolism [10,11,12,13,14]. EA is further metabolised by microflora of the large intestine to form a group of readily absorbed and biologically active compounds termed urolithins; the most intensively investigated being Urolithin A (UA) [15]. However, only ~ 40% of individuals demonstrate significant ability to convert ETs and EAs from dietary sources, such as pomegranate juice, into circulating UA, demonstrating the requirement to consider direct supplementation [16]. Indeed, in humans, acute supplementation with 500 mg UA was found to provide > 6-fold greater exposure to circulating UA than pomegranate juice [16].
In skeletal muscle culture, c. elegans and rodent models of ageing, treatment with UA has been shown to increase muscle function, angiogenesis, respiratory and exercise capacity, which have been attributed to enhanced mitophagy and mitochondrial functioning [17,18,19]. Additionally, studies have shown beneficial effects of UA on glucose tolerance and insulin sensitivity in rodent models of obesity and ageing [20,21,22]. In human trials, UA supplementation (500 or 1000 mg·day− 1) demonstrated a favourable safety profile in older adults, with significant biological availability and upregulated skeletal muscle mitochondrial gene expression [23, 24]. Myostatin, which principally serves to negatively regulate skeletal muscle growth, has been implicated in the metabolic dysregulation associated with pathophysiological ageing in humans [25] and positively associated with insulin resistance [26]; pertinently, its suppression has been repeatedly demonstrated through the actions of numerous polyphenols [8]. To date, however, the effects of UA on insulin and anabolic sensitivity in human skeletal muscle have not been investigated. This gap is important to address, since metabolic dysfunction in skeletal muscle contributes to the aetiology of sarcopenia and thus may explain the mechanisms by which UA supplementation in humans contributes to its beneficial effects in healthy middle-aged and older adults [23, 24, 27, 28]. Therefore, the aim of the present study was to establish whether acute treatment of primary human myotubes with UA would confer positive changes in insulin and anabolic sensitivity.
Methods
Primary cell culture and urolithin A treatments
Primary human myogenic cultures were established from skeletal muscle biopsy samples of eight healthy adults (4 male, 4 female) of varying age and BMI (mean [SD] age: 47 [± 26]; BMI: 27.1 [± 5.8] kg·m− 2). Following approval by the University of Nottingham Faculty of Medicine & Health Sciences Research Ethics Committee (reference No 143–1811) in accordance with the Code of Ethics of the World Medical Association (Declaration of Helsinki), written informed consent was obtained from all participants. Following this, they undertook a medical screening (comprising of medical history, health questionnaires, 12-lead electrocardiogram, blood pressure, weight and height measurements, and blood sample for routine chemistry and haematology) prior to obtaining the muscle sample. Myogenic cultures were proliferated and subsequently differentiated into myotubes, as described previously [25, 29]. Cultures were passaged no more than twice to best preserve skeletal muscle and donor phenotype. After 6–8 days of differentiation, myotubes were incubated for 24 h with 0.002, 1 and 50 µM UA (lyophilised UA provided by Amazentis SA, Lausanne, CH) or a matched vehicle treatment (0.1% DMSO). Viability of myotubes after UA treatment was assessed via resazurin assay, using UA concentrations from 0.002 to 100 µM.
Insulin and anabolic sensitivity
Basal and insulin-stimulated (100 nM) glucose uptake was assessed in myotubes using nominally radio-labelled (tritiated) deoxy-D-glucose (DOG) (2-[1,2–3 H (N)]; PerkinElmer, MA, USA), as previously described [30], with glucose uptake normalised against total cell protein content and converted to absolute glucose uptake rate (pmol·mg− 1·min− 1).
Changes in amino acid stimulated global myotube protein synthesis were estimated using the surface sensing of translation (SuNSET) technique [31], which measures the incorporation of the amino-nucleoside antibiotic puromycin into newly synthesised muscle proteins. Cells were stimulated for 3 h with 2 mM L-leucine and 5 mM D-glucose and for the final 30 min incubated with 1µM puromycin dihydrochloride, as previously described [25]. Briefly, equal amounts of total protein (determined via Pierce bicinchoninic acid assay) were separated on 12% SDS-PAGE gels, transferred onto PVDF membranes, incubated with anti-puromycin antibody (clone 12D10; Millipore, US) and visualized using chemiluminescence. Following immunodetection, membranes were stained with 0.1% Coomassie and digitally imaged. Total lane intensity was measured by densitometry and relative protein synthetic activity was estimated as the ratio of puromycin-labelled protein intensity to total protein (Coomassie) intensity.
mRNA expression
Myotubes were harvested using Tri-Reagent™ (ThermoFisher Scientific), prior to RNA extraction and cDNA synthesis using Invitrogen SuperScript™ in accordance with manufacturer’s guidelines. Gene expression was assessed via real time PCR (qPCR) with TaqMan probes (custom designed or commercially available pre-designed assay kits; Supplementary Table 1) on a StepOnePlus™ system (Applied Biosystems, CA, US). For the analysis of relative mRNA expression, genes of interest were normalised against the geometric mean of Hydroxymethylbilane Synthase (HMBS) and Ribosomal Protein Lateral Stalk Subunit P0 (RPLP0) expression, which demonstrated high stability within and between experiments and donors under these experimental conditions.
Statistical analysis and terminology to describe Biological and Technical Replication
All statistical analysis was performed in GraphPad Prism (Version 9.2). Descriptive data are presented as mean ± SD, whilst experimental data are presented as mean ± SEM. Assumptions of normality were tested via Shapiro-Wilk test, whereafter analysis of variance (ANOVA) with repeated measures, was performed. Significant (P < 0.05) post-hoc comparisons are presented as mean difference [95% CI], where relevant. All reported N values on each figure are the number of independent donor experiments, whereas the number of in vitro treatment replicates performed within each donor are reported in the legend, where relevant.
Results
Effect of urolithin A on the cell viability of human myogenic culture
Following differentiation, myotubes were either untreated (vehicle control), or treated with UA concentrations between 0.002 and 100 µM for a period of 24 h. No statistically significant loss of cell viability was observed with 24 h treatment with UA concentrations ≤ 50 µM, whereas at a concentration of 100 µM, there was a ~ 40% significant decline in cell viability (Fig. 1A).
Effect of 24 h UA exposure on cell viability, glucose uptake and amino-acid stimulated protein synthesis. (A) Fully differentiated human myotubes were incubated with either vehicle (0.1% DMSO), a positive control (10% DMSO), or varying concentrations of UA for 24 h. Viability was assayed fluorometrically vis Resazurin assay. Data are presented as Mean ± SEM of fluorescence intensity background (n = 3 replicate wells per treatment). (B) Basal (white bars) and insulin-stimulated (100 nM; grey bars) glucose uptake was measured using [3H] 2-DOG, following serum and glucose starvation. Glucose uptake was measured in pmol·mg− 1 protein·min− 1, however, to account for absolute differences in glucose uptake rate between myotubes from independent donor repeats. (C) Amino-acid stimulated protein synthesis was measured using the incorporation of puromycin (1µM) into newly synthesised muscle proteins following serum starvation and subsequent glucose (5mM) and leucine (2 mM) incubation. Proteins were separated via SDS-PAGE and probed with an anti-puromycin antibody to label newly synthesised peptides. Data for (B) and (C) are presented as Mean ± SEM of fold-change from Vehicle Basal for each donor repeat. Individual data points for each donor are the mean of 4–6 replicate wells for each treatment. Significant differences between treatments, as indicated: *P < 0.05, **P < 0.01
Effects of urolithin A on insulin and anabolic sensitivity
Mixed Effects ANOVA revealed significant treatment effects of both UA (P = 0.043; \(\:{\eta\:}_{P}^{2}\) = 0.410) and insulin (P = 0.008; \(\:{\eta\:}_{P}^{2}\) = 0.781) on glucose uptake, but not an interaction effect between the two (Fig. 1B). Post-hoc comparisons revealed statistically significant increases in basal (Mean Difference [95% CI]: 0.21 [0.01, 0.41]-fold greater; P < 0.05) and insulin-stimulated (Mean Difference: 0.24 [0.04, 0.43]-fold greater; P < 0.01) glucose uptake after treatment with 50 µM UA only, compared to Vehicle. In determining whether UA affected anabolic sensitivity, we observed no effect of UA treatment on amino acid-stimulated global cell protein synthesis (Fig. 1C).
Effects of urolithin A on the mRNA expression of myostatin, glucose transporters and autophagy
Repeated measures one-way ANOVA found a significant effect of UA treatment on the relative mRNA expression of MSTN (P = 0.031; \(\:{\eta\:}_{P}^{2}\) = 0.382), with significant suppression (14%) after treatment with 50 µM UA compared to Vehicle (Mean Difference [95% CI]: -0.210 [-0.031, -0.390] a.u.; P < 0.01; Fig. 2A). A statistically significant effect of UA treatment on GLUT4 (P = 0.016; \(\:{\eta\:}_{P}^{2}\) = 0.427) (Fig. 2B), but not GLUT1 (Fig. 2C), gene expression was also found, with GLUT4 expression being 1.8-fold greater with 50 µM UA vs. Vehicle (Mean Difference [95% CI]: 0.788 [0.080, 1.496] a.u.; P < 0.01). Data on expression of genes involved in cell autophagy, mitophagy and mitochondrial function are included in Fig. 3. One-way ANOVA revealed a significant effect of UA treatment on the relative mRNA expression of MAP1LC3B only (P = 0.028; \(\:{\eta\:}_{P}^{2}\) = 0.506), which was driven by upregulation (20%) with 50 µM UA compared to Vehicle (Fig. 3A).
Effect of 24 h UA exposure on MSTN (A), GLUT4 (B) and GLUT1 (C) mRNA expression. Repeated measures one-way ANOVA revealed a significant effect of UA on MSTN (A) and GLUT4 (B), but no effect on GLUT1 (C) mRNA expression. Data presented as Mean ± SEM of gene of interest expression, relative to the geometric mean of HMBS and RPLP0. Significant differences between treatments, as indicated: *P < 0.05
Effect of 24 h UA exposure on MAP1LC3B (A), NDUFA1 (B), PARK2 (C), SQSTM (D) and ESSRA (E) mRNA expression. Repeated measures one-way ANOVA revealed a significant effect of UA on MAP1LC3B mRNA expression only. Data presented as Mean ± SEM; n = 7 independent donors. Significant differences between treatments, as indicated: *P < 0.05
Discussion
To our awareness, the present study is the first to directly assess the effects of UA on indices of insulin and anabolic sensitivity in primary human myotubes. Expanding upon previous findings in non-human myogenic cultures and animal models, we found that acute (24 h) treatment of primary human myotubes with 50 µM UA markedly improved both basal- and insulin-stimulated glucose uptake, which was associated with significant upregulation of GLUT4 mRNA expression. Whilst we did not observe an acute improvement in global amino-acid stimulated cell protein synthesis, we did find a significant UA-induced reduction (14%) in the mRNA expression of MSTN, which is a potent negative regulator of skeletal muscle mass and development, which could prove beneficial in the context of chronic UA supplementation.
In the present study, treatment of myotubes with 50 µM UA elicited a 21% increase in basal glucose uptake, which was preserved but not further enhanced in the insulin-stimulated state. This suggests that UA has comparable potential to confer beneficial effects in skeletal muscle glucose uptake in both the post-absorptive and post-prandial state. Mechanistically, the increased glucose uptake observed with UA may be attributable to upregulation of GLUT4; treatment with 50 µM UA in our study induced an almost 2-fold increase in GLUT4 mRNA expression, which may explain the observed increases in glucose handling in both the basal and insulin-stimulated states. In accordance, recent observations in rat-derived L6 myogenic cultures have shown enhanced basal glucose uptake via increased recruitment of GLUT4 to the plasma membrane and activation of PI3K/Akt- and AMPK-dependent pathways in response to treatment with 100 and 200 µM but not 50 µM UA [32]. While GLUT4 abundance in primary human myotubes is low, it is still significantly recruited to the plasma membrane upon insulin stimulation [33] and is translocated in C2C12 cells upon stimulation with low doses of EA [34]. This observation therefore warrants further investigation under in vivo conditions.
In the context of the established propensity for UA to promote mitochondrial function and biogenesis in skeletal muscle [17, 23], the increased capacity for myotubes to uptake glucose in the present study may contribute to an increased capacity for oxidative phosphorylation, which is perturbed in insulin resistant individuals [35, 36]. Supporting this hypothesis, it has previously been demonstrated that chronic UA administration upregulates the activity of mitochondrial respiratory complexes I and II in the skeletal muscle of muscular dystrophic or HFD-fed mice [17, 19]. In the postprandial state, as much as 50% of the glucose that is taken up by skeletal muscle undergoes oxidation, demonstrating the importance of this mechanism in glucose homeostasis [37]. To that effect, glucose is first metabolised to pyruvate via glycolysis and can subsequently enter the mitochondria for further metabolism within the TCA cycle. Thus, it is plausible that the observed enhancement of glucose uptake serves the UA‑mediated increased capacity of the mitochondria, which expands upon the previously demonstrated UA-induced upregulation of basal oxygen consumption in C2C12 myotubes [19] and whole-body oxygen consumption in mice [21]. The significant upregulation of MAPLC3B (a key gene in cell autophagy) observed in the current study further support this notion. Interestingly, UA can activate AMPK [38], a common regulator of both autophagy and GLUT4 expression [39, 40], which may provide the mechanism by which UA exerts its dual effects on glucose uptake and mitochondrial function.
Another novel finding of this study was that UA treatment reduced MSTN mRNA expression by ~ 14%. Given myostatin’s involvement in the inhibition of muscle growth and regeneration [41,42,43] and the elevated MSTN mRNA expression reported in older adults with obesity [25], even modest sustained suppression of myostatin may present a promising avenue for nutraceutical targeted mitigation of sarcopenic obesity. Indeed, myostatin is considered a highly druggable target and the present study demonstrates a potential novel nutritional approach for its modulation that negates prior issues of poor ET/EA bioavailability.
Despite the suppression of MSTN mRNA expression, we didn’t observe any changes in amino acid-stimulated myotube protein synthesis, as indicated by the incorporation of the tyrosyl-tRNA analogue Puromycin into newly synthesised peptides. This is consistent with a study of C2C12 myotubes by Rodriguez and colleagues who reported that 24 h treatment with 15 µM UA did not affect myotube protein synthesis, and suggests an uncoupling of insulin and anabolic resistance in the UA-mediated suppression of myostatin in human skeletal muscle cells [44]. Therefore, longer duration of treatment with UA may be required to investigate the effects of sustained myostatin suppression on protein synthesis.
Limitations
In these experiments, myotubes were treated with so-called ‘parent UA’. It must be considered that parent UA is not the dominant form within the circulation. Rather, its phase II metabolites (predominantly UA-glucuronide and UA-sulfate) abound in far greater concentrations [23]. There is some evidence that UA conjugates have shown lower biological potency in vitro, compared to parent UA, on the bioactive anti-proliferation effect [13]. Future research must seek to better understand the role of conjugation in relation to UA effects on skeletal muscle biology. Furthermore, reflecting the time and resource intensive nature of primary human myogenic tissue culture, these experiments employed modest numbers of independent repeats, limiting extrapolation to the wider population. Nevertheless, the presence of significant effects in cultures from a relatively heterogenous sample of donors reinforces the robustness of these results and lays the ground for future studies in humans. Finally, the molecular events studied in the present study were limited to mRNA measurements, which may not always predict changes in protein levels [45]. However, correlations between mRNA and protein levels have been observed in human cell lines [46] and this relationship can be enhanced by applying gene-specific, cell type independent RNA-to-protein conversion factors [47].
Conclusions
This study provides the first direct evidence of metabolically beneficial effects of Urolithin A in primary human skeletal muscle cells. It was demonstrated that acute (24Â h) UA treatment improves both basal and insulin-stimulated glucose uptake in human skeletal muscle cells, and that these effects may be mediated by the upregulation of GLUT4 expression. Additionally, it was revealed that UA exerts a suppressive effect on the expression of MSTN in human skeletal muscle cells; while this did not translate to enhanced muscle protein synthesis in the acute setting, it could feasibly be beneficial in the context of chronic supplementation. Together these findings suggest plausible translation into human studies of obesity and ageing, where these effects, should be investigated further.
Data availability
No datasets were generated or analysed during the current study.
References
Organization WH. NONCOMMUNICABLE DISEASES COUNTRY PROFILES 2018. 2018.
Koch W. Dietary polyphenols-important non-nutrients in the Prevention of Chronic Noncommunicable diseases. A systematic review. Nutrients, 2019. 11(5).
Fraga CG, et al. The effects of polyphenols and other bioactives on human health. Food Funct. 2019;10(2):514–28.
Rodriguez J et al. Pomegranate extract prevents skeletal muscle of mice against wasting induced by acute TNF-α injection. Mol Nutr Food Res, 2017. 61(4).
Meador BM, et al. The Green Tea Polyphenol Epigallocatechin-3-Gallate (EGCg) attenuates skeletal muscle atrophy in a rat model of Sarcopenia. J Frailty Aging. 2015;4(4):209–15.
Salucci S, Falcieri E. Polyphenols and their potential role in preventing skeletal muscle atrophy. Nutr Res. 2020;74:10–22.
Francaux M, Deldicque L. Using polyphenol derivatives to prevent muscle wasting. Curr Opin Clin Nutr Metabolic Care. 2018;21(3):159–63.
Nikawa T, Ulla A, Sakakibara I. Polyphenols and their effects on muscle atrophy and muscle health. Molecules. 2021;26(16):4887.
TomĂ¡s-BarberĂ¡n FA et al. Urolithins, the rescue of old metabolites to understand a new concept: Metabotypes as a nexus among phenolic metabolism, microbiota dysbiosis, and host health status. Mol Nutr Food Res, 2017. 61(1).
Ceci C, et al. Strategies to improve ellagic acid bioavailability: from natural or semisynthetic derivatives to nanotechnological approaches based on innovative carriers. Nanotechnology. 2020;31(38):382001.
Seeram NP, et al. Pomegranate juice ellagitannin metabolites are present in human plasma and some persist in urine for up to 48 hours. J Nutr. 2006;136(10):2481–5.
Kang I, et al. Improvements in Metabolic Health with Consumption of Ellagic Acid and subsequent Conversion into urolithins: evidence and mechanisms. Adv Nutr. 2016;7(5):961–72.
GonzĂ¡lez-SarrĂas A, et al. Identifying the limits for ellagic acid bioavailability: a crossover pharmacokinetic study in healthy volunteers after consumption of pomegranate extracts. J Funct Foods. 2015;19:225–35.
Bala I, et al. Analytical methods for assay of ellagic acid and its solubility studies. J Pharm Biomed Anal. 2006;40(1):206–10.
EspĂn JC, et al. Biological significance of urolithins, the gut microbial ellagic acid-derived metabolites: the evidence so far. Evid Based Complement Alternat Med. 2013;2013:p270418.
Singh A, 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. 2022;76(2):297–308.
Luan P, et al. Urolithin A improves muscle function by inducing mitophagy in muscular dystrophy. Sci Transl Med. 2021;13(588):eabb0319.
Ghosh N, et al. Urolithin A augments angiogenic pathways in skeletal muscle by bolstering NAD(+) and SIRT1. Sci Rep. 2020;10(1):20184.
Ryu D, et al. Urolithin A induces mitophagy and prolongs lifespan in C. Elegans and increases muscle function in rodents. Nat Med. 2016;22(8):879–88.
Toney AM, et al. Urolithin A, a gut metabolite, improves insulin sensitivity through augmentation of mitochondrial function and Biogenesis. Obesity. 2019;27(4):612–20.
Xia B, et al. Urolithin A exerts antiobesity effects through enhancing adipose tissue thermogenesis in mice. PLoS Biol. 2020;18(3):e3000688.
Yang J, et al. Ellagic acid and its Microbial Metabolite Urolithin A Alleviate Diet-Induced insulin resistance in mice. Volume 64. Molecular Nutrition & Food Research; 2020. p. 2000091. 19.
Andreux PA, et al. The mitophagy activator urolithin A is safe and induces a molecular signature of improved mitochondrial and cellular health in humans. Nat Metabolism. 2019;1(6):595–603.
Liu S, et al. Effect of Urolithin A supplementation on Muscle Endurance and Mitochondrial Health in older adults: a Randomized Clinical Trial. JAMA Netw Open. 2022;5(1):e2144279–2144279.
Wilhelmsen A, et al. Skeletal muscle myostatin mRNA expression is upregulated in aged human adults with excess adiposity but is not associated with insulin resistance and ageing. Geroscience. 2024;46(2):2033–49.
Hittel DS, et al. Increased secretion and expression of myostatin in skeletal muscle from extremely obese women. Diabetes. 2009;58(1):30–8.
Coen PM, et al. Mitochondria as a target for mitigating Sarcopenia. Frontiers in Physiology; 2019. p. 9.
Singh A, et al. Urolithin A improves muscle strength, exercise performance, and biomarkers of mitochondrial health in a randomized trial in middle-aged adults. Cell Rep Med. 2022;3(5):100633.
O’Leary MF, et al. IL-15 promotes human myogenesis and mitigates the detrimental effects of TNFalpha on myotube development. Sci Rep. 2017;7(1):12997.
Nicholson T, et al. Vaspin promotes insulin sensitivity of elderly muscle and is upregulated in obesity. J Endocrinol, 2019:JOE-18-0528.R3. https://doiorg.publicaciones.saludcastillayleon.es/10.1530/JOE-18-0528.
Schmidt EK, et al. SUnSET, a nonradioactive method to monitor protein synthesis. Nat Methods. 2009;6(4):275–7.
Kondo S, et al. Antidiabetic effect of Urolithin A in cultured L6 myotubes and type 2 Diabetic Model KK-A(y)/Ta mice with glucose intolerance. Curr Issues Mol Biol. 2024;46(2):1078–90.
Al-Khalili L, et al. Insulin action in cultured human skeletal muscle cells during differentiation: assessment of cell surface GLUT4 and GLUT1 content. Cell Mol Life Sci. 2003;60(5):991–8.
Poulose N, et al. Ellagic acid stimulates glucose transport in adipocytes and muscles through AMPK mediated pathway. Journal of Diabetes & Metabolism. 2011;2(7). https://doiorg.publicaciones.saludcastillayleon.es/10.4172/2155-6156.1000149.
Petersen KF, et al. Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. N Engl J Med. 2004;350(7):664–71.
Sergi D, et al. Mitochondrial (dys)function and insulin resistance: from pathophysiological molecular mechanisms to the impact of diet. Front Physiol. 2019;10:532. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphys.2019.00532.
Kelley D, et al. Skeletal muscle glycolysis, oxidation, and storage of an oral glucose load. J Clin Investig. 1988;81(5):1563–71.
Gong Z, et al. Urolithin A attenuates memory impairment and neuroinflammation in APP/PS1 mice. J Neuroinflammation. 2019;16(1):62.
Holmes BF, Kurth-Kraczek EJ, Winder WW. Chronic activation of 5’-AMP-activated protein kinase increases GLUT-4, hexokinase, and glycogen in muscle. J Appl Physiol (1985). 1999;87(5):1990–5.
Kim J, et al. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 2011;13(2):132–41.
Wagner KR, et al. Muscle regeneration in the prolonged absence of myostatin. Proc Natl Acad Sci. 2005;102(7):2519–24.
Taylor WE, et al. Myostatin inhibits cell proliferation and protein synthesis in C2C12 muscle cells. Am J Physiology-Endocrinology Metabolism. 2001;280(2):E221–8.
Langley B, et al. Myostatin inhibits myoblast differentiation by Down-regulating MyoD expression. J Biol Chem. 2002;277(51):49831–40.
Rodriguez J, et al. Urolithin B, a newly identified regulator of skeletal muscle mass. J Cachexia Sarcopenia Muscle. 2017;8(4):583–97.
Fortelny N, et al. Can we predict protein from mRNA levels? Nature. 2017;547(7664):E19–20.
Wilhelm M, et al. Mass-spectrometry-based draft of the human proteome. Nature. 2014;509(7502):582–7.
Edfors F, et al. Gene-specific correlation of RNA and protein levels in human cells and tissues. Mol Syst Biol. 2016;12(10):883.
Funding
This work was supported by an MRC PhD studentship grant awarded to KT, SWJ and LGK.
Author information
Authors and Affiliations
Contributions
Conceptualization: KT, SWJ, LGK and AW designed the studies. Funding Acquisition: The MRC PhD studentship grant was awarded to KT, SWJ and LGK. Investigation: AW, LGK, AJB, DD, AMF, SWJ and KT conducted research. Data Curation and Formal Analysis: AW, KT, DD, AMF analysed data and performed statistical analysis. Roles/Writing: AW, LGK, SWJ and KT wrote the original manuscript. Review & Editing: AJB, DD, AMF, all reviewed and approved the manuscript prior to submission.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Conflict of interest
DD and AMF are employees of Amazentis SA, Lausanne, Switzerland. LGK was an employee of Nestlé Health Science, Vevey, Switzerland, during the project, is on the scientific advisory boards of Vital Proteins and NUUN, has participated on advisory boards of Liquid I.V and has received personal fees from RNWY and Nestlé Health Science, and is a board member of Siftlink. The other authors report no potential conflicts of interest.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Wilhelmsen, A., Karagounis, L.G., Bennett, A.J. et al. The polyphenol metabolite urolithin A suppresses myostatin expression and augments glucose uptake in human skeletal muscle cells. Nutr Metab (Lond) 22, 12 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12986-025-00909-0
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12986-025-00909-0