Pharmacological Effects of Urolithin A and Its Role in Muscle Health and Performance: Current Knowledge and Prospects
Abstract
:1. Introduction
2. Urolithin A’s Sources in the Diet
3. Metabolism and Bioavailability of Urolithin A in the Body
3.1. Gut Microbiota Metabolism
3.2. Absorption, Distribution, Metabolism, and Excretion
3.3. Factors Influencing the Absorption and Distribution of Urolithin A
3.3.1. Dietary Components
3.3.2. Composition and Activity of the Gut Microbiota
3.3.3. Individual Variations
3.3.4. Drug Interactions
4. Pharmacological Effects of Urolithin A
4.1. Activation of Mitochondrial Autophagy and Regeneration
4.2. Antioxidant Activity
4.3. Regulation of Cell Cycle and Cell Apoptosis
4.4. Influences on Metabolic Regulation
5. The Effects of Urolithin A on Muscle Health
5.1. The Antioxidant Activity of Urolithin A
5.2. The Anti-Inflammatory Effects of Urolithin A
5.3. The Role of Urolithin A in Promoting Mitochondrial Function and Muscle Energy Metabolism
6. The Effects of Urolithin A on Muscle Performance
6.1. The Potential Effects of Urolithin A on Endurance and Anti-Fatigue Capacity
6.2. The Effects of Urolithin A on Muscle Hypertrophy and Maintenance of Muscle Mass
7. The Signaling Pathways and Mechanisms of Action of Urolithin A in Muscle
7.1. The Interaction of Urolithin A with Key Signaling Pathways Involved in Muscle Health and Performance
7.1.1. AMPK Pathway
7.1.2. mTOR Pathway
7.1.3. NF-κB Pathway
7.1.4. PGC-1α Pathway
7.2. The Regulatory Effects of Urolithin A on Muscle Protein Synthesis and Degradation
7.2.1. FoxO Family
7.2.2. Ubiquitin-Proteasome System
7.2.3. mTORC1 and Atrogin-1/MuRF1
8. Clinical Research Limitations and Future Perspectives
9. Future Research Directions and Potential Applications of Urolithin A in Exercise Science
10. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Sinclair, J.R. Importance of a One Health approach in advancing global health security and the Sustainable Development Goals. Rev. Sci. Tech. OIE 2019, 38, 145–154. [Google Scholar] [CrossRef] [PubMed]
- Burke, L.M. Nutritional approaches to counter performance constraints in high-level sports competition. Exp. Physiol. 2021, 106, 2304–2323. [Google Scholar] [CrossRef] [PubMed]
- Forbes, S.C.; Little, J.P.; Candow, D.G. Exercise and nutritional interventions for improving aging muscle health. Endocrine 2012, 42, 29–38. [Google Scholar] [CrossRef] [PubMed]
- Sui, S.X.; Williams, L.J.; Holloway-Kew, K.L.; Hyde, N.K.; Pasco, J.A. Skeletal Muscle Health and Cognitive Function: A Narrative Review. Int. J. Mol. Sci. 2020, 22, 255. [Google Scholar] [CrossRef] [PubMed]
- Seeram, N.P.; Aronson, W.J.; Zhang, Y.; Henning, S.M.; Moro, A.; Lee, R.P.; Sartippour, M.; Harris, D.M.; Rettig, M.; Suchard, M.A.; et al. Pomegranate ellagitannin-derived metabolites inhibit prostate cancer growth and localize to the mouse prostate gland. J. Agric. Food Chem. 2007, 55, 7732–7737. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Qiu, Z.; Zhou, B.; Liu, C.; Ruan, J.; Yan, Q.; Liao, J.; Zhu, F. In vitro antiproliferative and antioxidant effects of urolithin A, the colonic metabolite of ellagic acid, on hepatocellular carcinomas HepG2 cells. Toxicol. Vitr. 2015, 29, 1107–1115. [Google Scholar] [CrossRef] [PubMed]
- D’Amico, D.; Andreux, P.A.; Valdes, P.; Singh, A.; Rinsch, C.; Auwerx, J. Impact of the Natural Compound Urolithin A on Health, Disease, and Aging. Trends Mol. Med. 2021, 27, 687–699. [Google Scholar] [CrossRef]
- Andreux, P.A.; Blanco-Bose, W.; Ryu, D.; Burdet, F.; Ibberson, M.; Aebischer, P.; Auwerx, J.; Singh, A.; Rinsch, C. The mitophagy activator urolithin A is safe and induces a molecular signature of improved mitochondrial and cellular health in humans. Nat. Metab. 2019, 1, 595–603. [Google Scholar] [CrossRef]
- Liu, S.; D’Amico, D.; Shankland, E.; Bhayana, S.; Garcia, J.M.; Aebischer, P.; Rinsch, C.; Singh, A.; Marcinek, D.J. Effect of Urolithin A Supplementation on Muscle Endurance and Mitochondrial Health in Older Adults: A Randomized Clinical Trial. JAMA Netw. Open 2022, 5, e2144279. [Google Scholar] [CrossRef] [PubMed]
- Singh, A.; D’Amico, D.; Andreux, P.A.; Fouassier, A.M.; Blanco-Bose, W.; Evans, M.; Aebischer, P.; Auwerx, J.; Rinsch, C. 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, 100633. [Google Scholar] [CrossRef]
- Stevens, J.F.; Maier, C.S. The Chemistry of Gut Microbial Metabolism of Polyphenols. Phytochem. Rev. 2016, 15, 425–444. [Google Scholar] [CrossRef] [PubMed]
- Lu, C.; Li, X.; Gao, Z.; Song, Y.; Shen, Y. Urolithins and intestinal health. Drug Discov. Ther. 2022, 16, 105–111. [Google Scholar] [CrossRef]
- Larrosa, M.; Garcia-Conesa, M.T.; Espin, J.C.; Tomas-Barberan, F.A. Ellagitannins, ellagic acid and vascular health. Mol. Asp. Med. 2010, 31, 513–539. [Google Scholar] [CrossRef]
- Espin, J.C.; Larrosa, M.; Garcia-Conesa, M.T.; Tomas-Barberan, F. Biological significance of urolithins, the gut microbial ellagic Acid-derived metabolites: The evidence so far. Evid. Based Compl. Alt. 2013, 2013, 270418. [Google Scholar] [CrossRef] [PubMed]
- Miller, K.; Feucht, W.; Schmid, M. Bioactive Compounds of Strawberry and Blueberry and Their Potential Health Effects Based on Human Intervention Studies: A Brief Overview. Nutrients 2019, 11, 1510. [Google Scholar] [CrossRef] [PubMed]
- Chen, P.; Guo, Z.; Chen, F.; Wu, Y.; Zhou, B. Recent Advances and Perspectives on the Health Benefits of Urolithin B, A Bioactive Natural Product Derived from Ellagitannins. Front. Pharmacol. 2022, 13, 917266. [Google Scholar] [CrossRef]
- Tomas-Barberan, F.A.; Gonzalez-Sarrias, A.; Garcia-Villalba, R.; Nunez-Sanchez, M.A.; Selma, M.V.; Garcia-Conesa, M.T.; Espin, J.C. 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, 1500901. [Google Scholar] [CrossRef]
- Selma, M.V.; Beltran, D.; Garcia-Villalba, R.; Espin, J.C.; Tomas-Barberan, F.A. Description of urolithin production capacity from ellagic acid of two human intestinal Gordonibacter species. Food Funct. 2014, 5, 1779–1784. [Google Scholar] [CrossRef] [PubMed]
- Selma, M.V.; Tomas-Barberan, F.A.; Beltran, D.; Garcia-Villalba, R.; Espin, J.C. Gordonibacter urolithinfaciens sp. nov., a urolithin-producing bacterium isolated from the human gut. Int. J. Syst. Evol. Micr. 2014, 64, 2346–2352. [Google Scholar] [CrossRef] [PubMed]
- Selma, M.V.; Beltran, D.; Luna, M.C.; Romo-Vaquero, M.; Garcia-Villalba, R.; Mira, A.; Espin, J.C.; Tomas-Barberan, F.A. Isolation of Human Intestinal Bacteria Capable of Producing the Bioactive Metabolite Isourolithin A from Ellagic Acid. Front. Microbiol. 2017, 8, 1521. [Google Scholar] [CrossRef]
- Beltran, D.; Romo-Vaquero, M.; Espin, J.C.; Tomas-Barberan, F.A.; Selma, M.V. Ellagibacter isourolithinifaciens gen. nov., sp. nov., a new member of the family Eggerthellaceae, isolated from human gut. Int. J. Syst. Evol. Micr. 2018, 68, 1707–1712. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Fang, Y.; Yang, G.; Hou, X.; Hai, Y.; Xia, M.; He, F.; Zhao, Y.; Liu, S. Isolation and characterization of a novel human intestinal Enterococcus faecium FUA027 capable of producing urolithin A from ellagic acid. Front. Nutr. 2022, 9, 1039697. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M.; Cui, S.; Mao, B.; Zhang, Q.; Zhao, J.; Zhang, H.; Tang, X.; Chen, W. Ellagic acid and intestinal microflora metabolite urolithin A: A review on its sources, metabolic distribution, health benefits, and biotransformation. Crit. Rev. Food Sci. 2022, 63, 6900–6922. [Google Scholar] [CrossRef]
- Garcia-Villalba, R.; Gimenez-Bastida, J.A.; Cortes-Martin, A.; Avila-Galvez, M.A.; Tomas-Barberan, F.A.; Selma, M.V.; Espin, J.C.; Gonzalez-Sarrias, A. Urolithins: A Comprehensive Update on their Metabolism, Bioactivity, and Associated Gut Microbiota. Mol. Nutr. Food Res. 2022, 66, e2101019. [Google Scholar] [CrossRef]
- Cerda, B.; Periago, P.; Espin, J.C.; Tomas-Barberan, F.A. Identification of urolithin a as a metabolite produced by human colon microflora from ellagic acid and related compounds. J. Agric. Food Chem. 2005, 53, 5571–5576. [Google Scholar] [CrossRef] [PubMed]
- Larrosa, M.; Gonzalez-Sarrias, A.; Garcia-Conesa, M.T.; Tomas-Barberan, F.A.; Espin, J.C. Urolithins, ellagic acid-derived metabolites produced by human colonic microflora, exhibit estrogenic and antiestrogenic activities. J. Agric. Food Chem. 2006, 54, 1611–1620. [Google Scholar] [CrossRef] [PubMed]
- Savi, M.; Bocchi, L.; Mena, P.; Dall’Asta, M.; Crozier, A.; Brighenti, F.; Stilli, D.; Del, R.D. In vivo administration of urolithin A and B prevents the occurrence of cardiac dysfunction in streptozotocin-induced diabetic rats. Cardiovasc. Diabetol. 2017, 16, 80. [Google Scholar] [CrossRef]
- Heilman, J.; Andreux, P.; Tran, N.; Rinsch, C.; Blanco-Bose, W. Safety assessment of Urolithin A, a metabolite produced by the human gut microbiota upon dietary intake of plant derived ellagitannins and ellagic acid. Food Chem. Toxicol. 2017, 108, 289–297. [Google Scholar] [CrossRef] [PubMed]
- Milala, J.; Kosmala, M.; Karlinska, E.; Juskiewicz, J.; Zdunczyk, Z.; Fotschki, B. Ellagitannins from Strawberries with Different Degrees of Polymerization Showed Different Metabolism through Gastrointestinal Tract of Rats. J. Agric. Food Chem. 2017, 65, 10738–10748. [Google Scholar] [CrossRef] [PubMed]
- Piwowarski, J.P.; Stanislawska, I.; Granica, S.; Stefanska, J.; Kiss, A.K. Phase II Conjugates of Urolithins Isolated from Human Urine and Potential Role of beta-Glucuronidases in Their Disposition. Drug Metab. Dispos. 2017, 45, 657–665. [Google Scholar] [CrossRef]
- Yaskolka, M.A.; Tuohy, K.; von Bergen, M.; Krajmalnik-Brown, R.; Heinig, U.; Zelicha, H.; Tsaban, G.; Rinott, E.; Kaplan, A.; Aharoni, A.; et al. The Metabolomic-Gut-Clinical Axis of Mankai Plant-Derived Dietary Polyphenols. Nutrients 2021, 13, 1866. [Google Scholar] [CrossRef]
- Cortes-Martin, A.; Selma, M.V.; Tomas-Barberan, F.A.; Gonzalez-Sarrias, A.; Espin, J.C. Where to Look into the Puzzle of Polyphenols and Health? The Postbiotics and Gut Microbiota Associated with Human Metabotypes. Mol. Nutr. Food Res. 2020, 64, e1900952. [Google Scholar] [CrossRef] [PubMed]
- Iglesias-Aguirre, C.E.; Cortes-Martin, A.; Avila-Galvez, M.A.; Gimenez-Bastida, J.A.; Selma, M.V.; Gonzalez-Sarrias, A.; Espin, J.C. Main drivers of (poly)phenol effects on human health: Metabolite production and/or gut microbiota-associated metabotypes? Food Funct. 2021, 12, 10324–10355. [Google Scholar] [CrossRef] [PubMed]
- Cortes-Martin, A.; Colmenarejo, G.; Selma, M.V.; Espin, J.C. Genetic Polymorphisms, Mediterranean Diet and Microbiota-Associated Urolithin Metabotypes can Predict Obesity in Childhood-Adolescence. Sci. Rep. 2020, 10, 7850. [Google Scholar] [CrossRef]
- Bahar, M.A.; Setiawan, D.; Hak, E.; Wilffert, B. Pharmacogenetics of drug-drug interaction and drug-drug-gene interaction: A systematic review on CYP2C9, CYP2C19 and CYP2D6. Pharmacogenomics 2017, 18, 701–739. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Xu, X.; Badawy, S.; Ihsan, A.; Liu, Z.; Xie, C.; Wang, X.; Tao, Y. A Review: Effects of Macrolides on CYP450 Enzymes. Curr. Drug Metab. 2020, 21, 928–937. [Google Scholar] [CrossRef] [PubMed]
- Niemi, M.; Backman, J.T.; Fromm, M.F.; Neuvonen, P.J.; Kivisto, K.T. Pharmacokinetic interactions with rifampicin: Clinical relevance. Clin. Pharmacokinet. 2003, 42, 819–850. [Google Scholar] [CrossRef]
- Klainer, A.S. Clindamycin. Med. Clin. N. Am. 1987, 71, 1169–1175. [Google Scholar] [CrossRef] [PubMed]
- Bingol, B.; Sheng, M. Mechanisms of mitophagy: PINK1, Parkin, USP30 and beyond. Free Radic. Bio Med. 2016, 100, 210–222. [Google Scholar] [CrossRef] [PubMed]
- Sliter, D.A.; Martinez, J.; Hao, L.; Chen, X.; Sun, N.; Fischer, T.D.; Burman, J.L.; Li, Y.; Zhang, Z.; Narendra, D.P.; et al. Parkin and PINK1 mitigate STING-induced inflammation. Nature 2018, 561, 258–262. [Google Scholar] [CrossRef]
- Quinn, P.; Moreira, P.I.; Ambrosio, A.F.; Alves, C.H. PINK1/PARKIN signalling in neurodegeneration and neuroinflammation. Acta Neuropathol. Com. 2020, 8, 189. [Google Scholar] [CrossRef] [PubMed]
- Huang, J.R.; Zhang, M.H.; Chen, Y.J.; Sun, Y.L.; Gao, Z.M.; Li, Z.J.; Zhang, G.P.; Qin, Y.; Dai, X.Y.; Yu, X.Y.; et al. Urolithin A ameliorates obesity-induced metabolic cardiomyopathy in mice via mitophagy activation. Acta Pharmacol. Sin. 2023, 44, 321–331. [Google Scholar] [CrossRef]
- Hassanein, E.; Sayed, A.M.; Hussein, O.E.; Mahmoud, A.M. Coumarins as Modulators of the Keap1/Nrf2/ARE Signaling Pathway. Oxid. Med. Cell Longev. 2020, 2020, 1675957. [Google Scholar] [CrossRef]
- Boakye, Y.D.; Groyer, L.; Heiss, E.H. An increased autophagic flux contributes to the anti-inflammatory potential of urolithin A in macrophages. BBA Gen. Subj. 2018, 1862, 61–70. [Google Scholar] [CrossRef] [PubMed]
- Morgan, M.J.; Liu, Z.G. Crosstalk of reactive oxygen species and NF-kappaB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef] [PubMed]
- Esselun, C.; Theyssen, E.; Eckert, G.P. Effects of Urolithin A on Mitochondrial Parameters in a Cellular Model of Early Alzheimer Disease. Int. J. Mol. Sci. 2021, 22, 8333. [Google Scholar] [CrossRef]
- Vicinanza, R.; Zhang, Y.; Henning, S.M.; Heber, D. Pomegranate Juice Metabolites, Ellagic Acid and Urolithin A, Synergistically Inhibit Androgen-Independent Prostate Cancer Cell Growth via Distinct Effects on Cell Cycle Control and Apoptosis. Evid. Based Compl. Alt. 2013, 2013, 247504. [Google Scholar] [CrossRef] [PubMed]
- Sanchez-Gonzalez, C.; Ciudad, C.J.; Izquierdo-Pulido, M.; Noe, V. Urolithin A causes p21 up-regulation in prostate cancer cells. Eur. J. Nutr. 2016, 55, 1099–1112. [Google Scholar] [CrossRef]
- El-Wetidy, M.S.; Ahmad, R.; Rady, I.; Helal, H.; Rady, M.I.; Vaali-Mohammed, M.A.; Al-Khayal, K.; Traiki, T.B.; Abdulla, M.H. Urolithin A induces cell cycle arrest and apoptosis by inhibiting Bcl-2, increasing p53-p21 proteins and reactive oxygen species production in colorectal cancer cells. Cell Stress. Chaperon. 2021, 26, 473–493. [Google Scholar] [CrossRef] [PubMed]
- Wei, W.; Peng, C.; Gu, R.; Yan, X.; Ye, J.; Xu, Z.; Sheng, X.; Huang, G.; Guo, Y. Urolithin A attenuates RANKL-induced osteoclastogenesis by co-regulating the p38 MAPK and Nrf2 signaling pathway. Eur. J. Pharmacol. 2022, 921, 174865. [Google Scholar] [CrossRef] [PubMed]
- Lin, S.C.; Hardie, D.G. AMPK: Sensing Glucose as well as Cellular Energy Status. Cell Metab. 2018, 27, 299–313. [Google Scholar] [CrossRef]
- Kim, J.; Yang, G.; Kim, Y.; Kim, J.; Ha, J. AMPK activators: Mechanisms of action and physiological activities. Exp. Mol. Med. 2016, 48, e224. [Google Scholar] [CrossRef] [PubMed]
- Han, Q.A.; Yan, C.; Wang, L.; Li, G.; Xu, Y.; Xia, X. Urolithin A attenuates ox-LDL-induced endothelial dysfunction partly by modulating microRNA-27 and ERK/PPAR-gamma pathway. Mol. Nutr. Food Res. 2016, 60, 1933–1943. [Google Scholar] [CrossRef] [PubMed]
- Janani, C.; Ranjitha, K.B. PPAR gamma gene—A review. Diabetes Metab. Synd. 2015, 9, 46–50. [Google Scholar] [CrossRef] [PubMed]
- Sies, H. Oxidative stress: A concept in redox biology and medicine. Redox Biol. 2015, 4, 180–183. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, N.H.; Tran, G.B.; Nguyen, C.T. Anti-oxidative effects of superoxide dismutase 3 on inflammatory diseases. J. Mol. Med. 2020, 98, 59–69. [Google Scholar] [CrossRef] [PubMed]
- Powers, S.K.; Deminice, R.; Ozdemir, M.; Yoshihara, T.; Bomkamp, M.P.; Hyatt, H. Exercise-induced oxidative stress: Friend or foe? J. Sport. Health Sci. 2020, 9, 415–425. [Google Scholar] [CrossRef] [PubMed]
- Toney, A.M.; Fox, D.; Chaidez, V.; Ramer-Tait, A.E.; Chung, S. Immunomodulatory Role of Urolithin A on Metabolic Diseases. Biomedicines 2021, 9, 192. [Google Scholar] [CrossRef]
- D’Amico, D.; Olmer, M.; Fouassier, A.M.; Valdes, P.; Andreux, P.A.; Rinsch, C.; Lotz, M. Urolithin A improves mitochondrial health, reduces cartilage degeneration, and alleviates pain in osteoarthritis. Aging Cell 2022, 21, e13662. [Google Scholar] [CrossRef]
- Halling, J.F.; Pilegaard, H. PGC-1alpha-mediated regulation of mitochondrial function and physiological implications. Appl. Physiol. Nutr. Metab. 2020, 45, 927–936. [Google Scholar] [CrossRef]
- Rius-Perez, S.; Torres-Cuevas, I.; Millan, I.; Ortega, A.L.; Perez, S. PGC-1alpha, Inflammation, and Oxidative Stress: An Integrative View in Metabolism. Oxid. Med. Cell Longev. 2020, 2020, 1452696. [Google Scholar] [CrossRef]
- Rajendran, M.; Dane, E.; Conley, J.; Tantama, M. Imaging Adenosine Triphosphate (ATP). Biol. Bull. 2016, 231, 73–84. [Google Scholar] [CrossRef]
- Chen, X.; Xue, Y.; Jia, G.; Zhao, H.; Liu, G.; Huang, Z. Antifatigue effect of naringin on improving antioxidant capacity and mitochondrial function and preventing muscle damage. Exp. Biol. Med. 2022, 247, 1776–1784. [Google Scholar] [CrossRef] [PubMed]
- Schoenfeld, B.J. The mechanisms of muscle hypertrophy and their application to resistance training. J. Strength. Cond. Res. 2010, 24, 2857–2872. [Google Scholar] [CrossRef]
- Jayatunga, D.; Hone, E.; Khaira, H.; Lunelli, T.; Singh, H.; Guillemin, G.J.; Fernando, B.; Garg, M.L.; Verdile, G.; Martins, R.N. Therapeutic Potential of Mitophagy-Inducing Microflora Metabolite, Urolithin A for Alzheimer’s Disease. Nutrients 2021, 13, 3744. [Google Scholar] [CrossRef] [PubMed]
- Li, K.; Xiao, Y.; Bian, J.; Han, L.; He, C.; El-Omar, E.; Gong, L.; Wang, M. Ameliorative Effects of Gut Microbial Metabolite Urolithin A on Pancreatic Diseases. Nutrients 2022, 14, 2549. [Google Scholar] [CrossRef] [PubMed]
- Huang, W.C.; Liou, C.J.; Shen, S.C.; Hu, S.; Chao, J.C.; Hsiao, C.Y.; Wu, S.J. Urolithin A Inactivation of TLR3/TRIF Signaling to Block the NF-kappaB/STAT1 Axis Reduces Inflammation and Enhances Antioxidant Defense in Poly(I:C)-Induced RAW264.7 Cells. Int. J. Mol. Sci. 2022, 23, 4697. [Google Scholar] [CrossRef] [PubMed]
- Tow, W.K.; Chee, P.Y.; Sundralingam, U.; Palanisamy, U.D. The Therapeutic Relevance of Urolithins, Intestinal Metabolites of Ellagitannin-Rich Food: A Systematic Review of In Vivo Studies. Nutrients 2022, 14, 3494. [Google Scholar] [CrossRef] [PubMed]
- Luo, C.; Wei, X.; Song, J.; Xu, X.; Huang, H.; Fan, S.; Zhang, D.; Han, L.; Lin, J. Interactions between Gut Microbiota and Polyphenols: New Insights into the Treatment of Fatigue. Molecules 2022, 27, 7377. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Proud, C.G. The mTOR pathway in the control of protein synthesis. Physiology 2006, 21, 362–369. [Google Scholar] [CrossRef]
- Ersahin, T.; Tuncbag, N.; Cetin-Atalay, R. The PI3K/AKT/mTOR interactive pathway. Mol. Biosyst. 2015, 11, 1946–1954. [Google Scholar] [CrossRef]
- Kim, Y.C.; Guan, K.L. mTOR: A pharmacologic target for autophagy regulation. J. Clin. Investig. 2015, 125, 25–32. [Google Scholar] [CrossRef] [PubMed]
- Mehra, S.; Srinivasan, S.; Singh, S.; Zhou, Z.; Garrido, V.; Silva, I.C.; Totiger, T.M.; Dosch, A.R.; Dai, X.; Dawra, R.K.; et al. Urolithin A attenuates severity of chronic pancreatitis associated with continued alcohol intake by inhibiting PI3K/AKT/mTOR signaling. Am. J. Physiol. Gastr. Liver Physiol. 2022, 323, G375–G386. [Google Scholar] [CrossRef] [PubMed]
- Chen, P.; Chen, F.; Lei, J.; Li, Q.; Zhou, B. Activation of the miR-34a-Mediated SIRT1/mTOR Signaling Pathway by Urolithin A Attenuates D-Galactose-Induced Brain Aging in Mice. Neurotherapeutics 2019, 16, 1269–1282. [Google Scholar] [CrossRef]
- Totiger, T.M.; Srinivasan, S.; Jala, V.R.; Lamichhane, P.; Dosch, A.R.; Gaidarski, A.R.; Joshi, C.; Rangappa, S.; Castellanos, J.; Vemula, P.K.; et al. Urolithin A, a Novel Natural Compound to Target PI3K/AKT/mTOR Pathway in Pancreatic Cancer. Mol. Cancer Ther. 2019, 18, 301–311. [Google Scholar] [CrossRef] [PubMed]
- Lawrence, T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb. Perspect. Biol. 2009, 1, a1651. [Google Scholar] [CrossRef] [PubMed]
- Hayden, M.S.; Ghosh, S. NF-kappaB in immunobiology. Cell Res. 2011, 21, 223–244. [Google Scholar] [CrossRef]
- Fu, X.; Gong, L.F.; Wu, Y.F.; Lin, Z.; Jiang, B.J.; Wu, L.; Yu, K.H. Urolithin A targets the PI3K/Akt/NF-kappaB pathways and prevents IL-1beta-induced inflammatory response in human osteoarthritis: In vitro and in vivo studies. Food Funct. 2019, 10, 6135–6146. [Google Scholar] [CrossRef]
- Tao, H.; Li, W.; Zhang, W.; Yang, C.; Zhang, C.; Liang, X.; Yin, J.; Bai, J.; Ge, G.; Zhang, H.; et al. Urolithin A suppresses RANKL-induced osteoclastogenesis and postmenopausal osteoporosis by, suppresses inflammation and downstream NF-kappaB activated pyroptosis pathways. Pharmacol. Res. 2021, 174, 105967. [Google Scholar] [CrossRef]
- Ghosh, N.; Das, A.; Biswas, N.; Gnyawali, S.; Singh, K.; Gorain, M.; Polcyn, C.; Khanna, S.; Roy, S.; Sen, C.K. Urolithin A augments angiogenic pathways in skeletal muscle by bolstering NAD(+) and SIRT1. Sci. Rep. 2020, 10, 20184. [Google Scholar] [CrossRef] [PubMed]
- Shi, P.Z.; Wang, J.W.; Wang, P.C.; Han, B.; Lu, X.H.; Ren, Y.X.; Feng, X.M.; Cheng, X.F.; Zhang, L. Urolithin a alleviates oxidative stress-induced senescence in nucleus pulposus-derived mesenchymal stem cells through SIRT1/PGC-1alpha pathway. World J. Stem Cells 2021, 13, 1928–1946. [Google Scholar] [CrossRef]
- Liu, J.; Jiang, J.; Qiu, J.; Wang, L.; Zhuo, J.; Wang, B.; Sun, D.; Yu, S.; Lou, H. Urolithin A protects dopaminergic neurons in experimental models of Parkinson’s disease by promoting mitochondrial biogenesis through the SIRT1/PGC-1alpha signaling pathway. Food Funct. 2022, 13, 375–385. [Google Scholar] [CrossRef] [PubMed]
- Chan, M.C.; Arany, Z. The many roles of PGC-1alpha in muscle—Recent developments. Metabolism 2014, 63, 441–451. [Google Scholar] [CrossRef] [PubMed]
- Martins, R.; Lithgow, G.J.; Link, W. Long live FOXO: Unraveling the role of FOXO proteins in aging and longevity. Aging Cell 2016, 15, 196–207. [Google Scholar] [CrossRef]
- Link, W. Introduction to FOXO Biology. In FOXO Transcription Factors: Methods and Protocols; Springer: Berlin/Heidelberg, Germany, 2019; Volume 1890, pp. 1–9. [Google Scholar] [CrossRef]
- Fang, E.F.; Hou, Y.; Palikaras, K.; Adriaanse, B.A.; Kerr, J.S.; Yang, B.; Lautrup, S.; Hasan-Olive, M.M.; Caponio, D.; Dan, X.; et al. Mitophagy inhibits amyloid-beta and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat. Neurosci. 2019, 22, 401–412. [Google Scholar] [CrossRef] [PubMed]
- Nandi, D.; Tahiliani, P.; Kumar, A.; Chandu, D. The ubiquitin-proteasome system. J. Biosci. 2006, 31, 137–155. [Google Scholar] [CrossRef]
- Khalil, R. Ubiquitin-Proteasome Pathway and Muscle Atrophy. Adv. Exp. Med. Biol. 2018, 1088, 235–248. [Google Scholar] [CrossRef]
- Xiong, Y.; Yu, C.; Zhang, Q. Ubiquitin-Proteasome System-Regulated Protein Degradation in Spermatogenesis. Cells 2022, 11, 1058. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez, J.; Caille, O.; Ferreira, D.; Francaux, M. Pomegranate extract prevents skeletal muscle of mice against wasting induced by acute TNF-alpha injection. Mol. Nutr. Food Res. 2017, 61, 1600169. [Google Scholar] [CrossRef]
- Gumucio, J.P.; Mendias, C.L. Atrogin-1, MuRF-1, and sarcopenia. Endocrine 2013, 43, 12–21. [Google Scholar] [CrossRef]
- De Naeyer, H.; Lamon, S.; Russell, A.P.; Everaert, I.; De Spaey, A.; Vanheel, B.; Taes, Y.; Derave, W. Androgenic and estrogenic regulation of Atrogin-1, MuRF1 and myostatin expression in different muscle types of male mice. Eur. J. Appl. Physiol. 2014, 114, 751–761. [Google Scholar] [CrossRef] [PubMed]
- Condon, K.J.; Sabatini, D.M. Nutrient regulation of mTORC1 at a glance. J. Cell Sci. 2019, 132, jcs222570. [Google Scholar] [CrossRef] [PubMed]
- Szwed, A.; Kim, E.; Jacinto, E. Regulation and metabolic functions of mTORC1 and mTORC2. Physiol. Rev. 2021, 101, 1371–1426. [Google Scholar] [CrossRef] [PubMed]
- McCarty, M.F. Nutraceutical and Dietary Strategies for Up-Regulating Macroautophagy. Int. J. Mol. Sci. 2022, 23, 2054. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Zhao, H.; Song, G.; Zhu, H.; Qian, H.; Pan, X.; Song, X.; Xie, Y.; Liu, C. Pharmacological Effects of Urolithin A and Its Role in Muscle Health and Performance: Current Knowledge and Prospects. Nutrients 2023, 15, 4441. https://doi.org/10.3390/nu15204441
Zhao H, Song G, Zhu H, Qian H, Pan X, Song X, Xie Y, Liu C. Pharmacological Effects of Urolithin A and Its Role in Muscle Health and Performance: Current Knowledge and Prospects. Nutrients. 2023; 15(20):4441. https://doi.org/10.3390/nu15204441
Chicago/Turabian StyleZhao, Haotian, Ge Song, Hongkang Zhu, He Qian, Xinliang Pan, Xiaoneng Song, Yijie Xie, and Chang Liu. 2023. "Pharmacological Effects of Urolithin A and Its Role in Muscle Health and Performance: Current Knowledge and Prospects" Nutrients 15, no. 20: 4441. https://doi.org/10.3390/nu15204441
APA StyleZhao, H., Song, G., Zhu, H., Qian, H., Pan, X., Song, X., Xie, Y., & Liu, C. (2023). Pharmacological Effects of Urolithin A and Its Role in Muscle Health and Performance: Current Knowledge and Prospects. Nutrients, 15(20), 4441. https://doi.org/10.3390/nu15204441