Hostname: page-component-54dcc4c588-xh45t Total loading time: 0 Render date: 2025-10-08T07:25:23.246Z Has data issue: false hasContentIssue false

Ergothioneine for cognitive health, longevity and healthy ageing: where are we now?

Published online by Cambridge University Press:  19 September 2025

Linda S. May-Zhang*
Affiliation:
Blue California, Rancho Santa Margarita, California, USA
Irwin K. Cheah
Affiliation:
Department of Biochemistry, Yong Loo Lin School of Medicine, and Centre for Life Sciences, National University of Singapore, Singapore
Ian T. Zajac
Affiliation:
College of Education, Psychology and Social Work, Flinders University, Adelaide, Australia
Emily Brindal
Affiliation:
Health & Biosecurity, Commonwealth Scientific and Industrial Research Organization, Adelaide, Australia
Naomi Kakoschke
Affiliation:
Health & Biosecurity, Commonwealth Scientific and Industrial Research Organization, Adelaide, Australia
*
Corresponding author: Linda S. May-Zhang; Email: linda.mayzhang@bluecal-ingredients.com

Abstract

As the global population ages, the prevalence of cognitive decline is rising, creating urgent demand for proactive strategies that support brain health and healthy ageing. Ergothioneine, a unique dietary amino-thione absorbed via the OCTN1 transporter, has recently gained attention for its potential as a neuroprotective, longevity-promoting compound. This review synthesizes growing evidence from observational, interventional and mechanistic studies. Observational data consistently associate low blood ergothioneine levels with cognitive impairment, neurodegenerative diseases, cardiovascular disorders, frailty and mortality. Interventional trials in older adults suggest that ergothioneine supplementation may improve cognition, memory, sleep quality and stabilize neurodegeneration biomarkers, with no safety concerns at doses up to 25 mg/day. Mechanistic studies reveal that ergothioneine acts through multiple pathways: mitigating oxidative stress, reducing neuroinflammation, preserving mitochondrial function and potentially modulating neurogenesis and NAD+ metabolism, although some mechanisms require further investigation. Beyond cognition, ergothioneine shows promise in supporting other physiological systems relevant to ageing, including cardiovascular, metabolic, gut, eye, auditory, liver, kidney, immune, skin and lung health. Together, current evidence positions ergothioneine as a promising nutritional intervention for promoting cognitive resilience and systemic health in ageing, although larger, long-term interventional trials are needed to confirm causality and optimize use.

Information

Type
Conference on Food for all: Promoting Equity, Diversity and Inclusion in Nutrition
Copyright
© The Author(s), 2025. Published by Cambridge University Press on behalf of The Nutrition Society

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

Article purchase

Temporarily unavailable

References

Lipnicki, DM, Crawford, JD, Dutta, R et al. (2017) Age-related cognitive decline and associations with sex, education and apolipoprotein E genotype across ethnocultural groups and geographic regions: a collaborative cohort study. PLoS Med 14, e1002261.Google Scholar
Foster, TC, DeFazio, RA, Bizon, JL. (2012) Characterizing cognitive aging of spatial and contextual memory in animal models. Front Aging Neurosci 4. doi: 10.3389/fnagi.2012.00012.Google Scholar
Zheng, D, Sun, H, Dong, X et al. (2014) Executive dysfunction and gray matter atrophy in amnestic mild cognitive impairment. Neurobiol Aging 35, 548555.Google Scholar
Deary, IJ, Corley, J, Gow, AJ et al. (2009) Age-associated cognitive decline. Br Med Bull 92, 135152.Google Scholar
Livingston, G, Huntley, J, Liu, KY et al. (2024) Dementia prevention, intervention, and care: 2024 report of the Lancet standing Commission. Lancet 404, 572628.Google Scholar
Norton, S, Matthews, FE, Barnes, DE et al. (2014) Potential for primary prevention of Alzheimer’s disease: an analysis of population-based data. Lancet Neurol 13(8), 788794.Google Scholar
Kolappa, K, Seeher, K. (2022) Optimizing brain health across the life course: WHO position paper. World Health Organization. ISBN: 9789240054561 https://www.who.int/publications/i/item/9789240054561 Google Scholar
Scarmeas, N, Stern, Y, Tang, M et al. (2006) Mediterranean diet and risk for Alzheimer’s disease. Ann Neurol 59, 912921.Google Scholar
Martínez-Lapiscina, EH, Clavero, P, Toledo, E et al. (2013) Mediterranean diet improves cognition: the PREDIMED-NAVARRA randomised trial. J Neurol Neurosurg Psychiatry 84, 13181325.Google Scholar
Smith, PJ, Blumenthal, JA, Babyak, MA et al. (2010) Effects of the dietary approaches to stop hypertension diet, exercise, and caloric restriction on neurocognition in overweight adults with high blood pressure. Hypertension 55, 13311338.Google Scholar
van Soest, AP, Beers, S, van de Rest, O et al. (2014) The Mediterranean-dietary approaches to stop hypertension intervention for neurodegenerative delay (MIND) diet for the aging brain: a systematic review. Adv Nutr 15, 100184.Google Scholar
Chen, KH, Ho, MH, Wang, CS et al. (2023) Effect of dietary patterns on cognitive functions of older adults: a systematic review and meta-analysis of randomized controlled trials. Arch Gerontol Geriatr 110, 104967.Google Scholar
Tingö, L, Bergh, C, Rode, J et al. (2024) The effect of whole-diet interventions on memory and cognitive function in healthy older adults – a systematic review. Adv Nutr 15, 100291.Google Scholar
Landi, F, Calvani, R, Tosato, M et al. (2016) Anorexia of aging: risk factors, consequences, and potential treatments. Nutrients 8, 69.Google Scholar
Gómez-Pinilla, F. (2008) Brain foods: the effects of nutrients on brain function. Nat Rev Neurosci 9, 568578.Google Scholar
Gutierrez, L, Folch, A, Rojas, M et al. (2021) Effects of nutrition on cognitive function in adults with or without cognitive impairment: a systematic review of randomized controlled clinical trials. Nutrients 13, 3728.Google Scholar
Fekete, M, Lehoczki, A, Tarantini, S et al. (2023) Improving cognitive function with nutritional supplements in aging: a comprehensive narrative review of clinical studies investigating the effects of vitamins, minerals, antioxidants, and other dietary supplements. Nutrients 15, 5116.Google Scholar
Vauzour, D, Scholey, A, White, DJ et al. (2023) A combined DHA-rich fish oil and cocoa flavanols intervention does not improve cognition or brain structure in older adults with memory complaints: results from the CANN randomized, controlled parallel-design study. Am J Clin Nutr 118, 369381.Google Scholar
Baleztena, J, Ruiz-Canela, M, Sayon-Orea, C et al. (2018) Association between cognitive function and supplementation with omega-3 PUFAs and other nutrients in ≥ 75 years old patients: a randomized multicenter study. PLoS One 13, e0193568.Google Scholar
Dangour, AD, Allen, E, Elbourne, D et al. (2010) Effect of 2-y n−3 long-chain polyunsaturated fatty acid supplementation on cognitive function in older people: a randomized, double-blind, controlled trial. Am J Clin Nutr 91, 17251732.Google Scholar
Gründemann, D, Harlfinger, S, Golz, S et al. (2005) Discovery of the ergothioneine transporter. Proc Nati Acad Sci 102, 52565261.Google Scholar
Motohashi, N, Mori, I & Sugiura, Y. (1976) 13C-nuclear magnetic resonance and Raman spectroscopic studies on ionization and mercury complex of ergothioneine. Chem Pharm Bull 24, 17371741.Google Scholar
Yadan, J. (2022) Matching chemical properties to molecular biological activities opens a new perspective on l-ergothioneine. FEBS Lett 596, 12991312.Google Scholar
Cheah, IK, Tang, RMY, Yew, TSZ et al. (2017) Administration of pure ergothioneine to healthy human subjects: uptake, metabolism, and effects on biomarkers of oxidative damage and inflammation. Antioxid Redox Signal 26, 193206.Google Scholar
Halliwell, B & Cheah, I. (2024) Are age-related neurodegenerative diseases caused by a lack of the diet-derived compound ergothioneine? Free Radic Biol Med 217, 6067.Google Scholar
Halliwell, B, Tang, RMY & Cheah, IK. (2023) Diet-derived antioxidants: the special case of ergothioneine. Annu Rev Food Sci Technol 14, 323345.Google Scholar
Fromentin, S, Forslund, SK, Chechi, K et al. (2022) Microbiome and metabolome features of the cardiometabolic disease spectrum. Nat Med 28, 303314.Google Scholar
Sebastiani, P, Monti, S, Lustgarten, MS et al. (2024) Metabolite signatures of chronological age, aging, survival, and longevity. Cell Rep 43, 114913.Google Scholar
Kameda, M, Teruya, T, Yanagida, M et al. (2020) Frailty markers comprise blood metabolites involved in antioxidation, cognition, and mobility. Proc Natl Acad Sci 117, 94839489.Google Scholar
Yau, YF, Cheah, IK, Mahendran, R et al. (2024) Investigating the efficacy of ergothioneine to delay cognitive decline in mild cognitively impaired subjects: a pilot study. J Alzheimer’s Disease 102, 841854.Google Scholar
Zajac, IT, Kakoschke, N, Kuhn-Sherlock, B et al. (2025) The effect of ergothioneine supplementation on cognitive function, memory, and sleep in older adults with subjective memory complaints: a randomized placebo-controlled trial. Nutraceuticals 5, 15.Google Scholar
Katsube, M, Ishimoto, T, Fukushima, Y et al. (2024) Ergothioneine promotes longevity and healthy aging in male mice. GeroScience 46, 38893909.Google Scholar
Okumura, H, Araragi, Y, Nishioka, K et al. (2025) Estimation and validation of an effective ergothioneine dose for improved sleep quality using physiologically based pharmacokinetic model. Food Sci. Nutr 13, e70382.Google Scholar
Paul, BD & Snyder, SH. (2010) The unusual amino acid L-ergothioneine is a physiologic cytoprotectant. Cell Death Differ 17, 11341140.Google Scholar
Ames, BN. (2018) Prolonging healthy aging: longevity vitamins and proteins. Proc Natl Acad Sci 115, 1083610844.Google Scholar
Chen, L, Qiu, R, Wang, B et al. (2024) Investigating the association between inflammation mediated by mushroom consumption and mild cognitive impairment in Chinese older adults. Food Funct 15, 53435351.Google Scholar
Yan, Y, Li, B, Li, F et al. (2023) The relationship between mushroom consumption and cognitive performance among middle-aged and older adults: a cross-sectional study. Food Funct 14, 76637671.Google Scholar
Feng, L, Cheah, IKM, Ng, MMX et al. (2019) The association between mushroom consumption and mild cognitive impairment: a community-based cross-sectional study in singapore. J Alzheimer’s Disease 68, 197203.Google Scholar
Aoki, S, Yamagishi, K, Maruyama, K et al. (2024) Mushroom intake and risk of incident disabling dementia: the Circulatory Risk in Communities Study (CIRCS). Br J Nutr 131, 16411647.Google Scholar
Zhang, S, Tomata, Y, Sugiyama, K et al. (2017) Mushroom consumption and incident dementia in elderly Japanese: the Ohsaki cohort 2006 study. J Am Geriatr Soc 65, 14621469.Google Scholar
Ba, DM, Gao, X, Al-Shaar, L et al. (2021) Mushroom intake and depression: a population-based study using data from the US National Health and Nutrition Examination Survey (NHANES), 2005–2016. J Affect Disord 294, 686692.Google Scholar
Ba, DM, Ssentongo, P, Pelucchi, C et al. (2023) Mushroom consumption and risk of gastric cancer: a pooled analysis within the stomach cancer pooling project and a combined meta-analysis with other observational studies. European J Cancer Prevention 32, 222228.Google Scholar
Ba, DM, Gao, X, Muscat, J et al. (2021) Association of mushroom consumption with all-cause and cause-specific mortality among American adults: prospective cohort study findings from NHANES III. Nutr J 20, 38.Google Scholar
Cha, S, Bell, L & Williams, CM. (2024) The relationship between mushroom intake and cognitive performance: an epidemiological study in the european investigation of cancer—Norfolk Cohort (EPIC-Norfolk). Nutrients 16, 353.Google Scholar
Ba, DM, Gao, X, Al-Shaar, L et al. (2022) Mushroom intake and cognitive performance among US older adults: the National Health and Nutrition Examination Survey, 2011–2014. Br J Nutr 128, 22412248.Google Scholar
Li, N, Li, H, Liu, Z et al. (2023) Unveiling the therapeutic potentials of mushroom bioactive compounds in Alzheimer’s Disease. Foods 12, 2972.Google Scholar
Uffelman, CN, Doenges, KA, Armstrong, ML et al. (2023) Metabolomics profiling of white button, crimini, portabella, lion’s mane, maitake, oyster, and shiitake mushrooms using untargeted metabolomics and targeted amino acid analysis. Foods 12, 2985.Google Scholar
Kalaras, MD, Richie, JP, Calcagnotto, A, et al. (2017) Mushrooms: a rich source of the antioxidants ergothioneine and glutathione. Food Chem 233, 429433.Google Scholar
Ey, J, Schömig, E & Taubert, D. (2007) Dietary sources and antioxidant effects of ergothioneine. J Agric Food Chem 55, 64666474.Google Scholar
Chen, F, Wang, B, Sun, X et al. (2024) Ergothioneine improves cognitive function by ameliorating mitochondrial damage and decreasing neuroinflammation in a d-galactose-induced aging model. Food Funct 15, 1168611696.Google Scholar
Cheah, IK, Feng, L, Tang, RMY et al. (2016) Ergothioneine levels in an elderly population decrease with age and incidence of cognitive decline; a risk factor for neurodegeneration? Biochem Biophys Res Commun 478, 162167.Google Scholar
González-Domínguez, R, Castellano-Escuder, P, Carmona, F et al. (2021) Food and microbiota metabolites associate with cognitive decline in older subjects: a 12-year prospective study. Mol Nutr Food Res 65, e2100606.Google Scholar
Wu, LY, Kan, CN, Cheah, IK et al. (2022) Low plasma ergothioneine predicts cognitive and functional decline in an elderly cohort attending memory clinics. Antioxidants (Basel) 11, 1717.Google Scholar
Teruya, T, Chen, YJ, Fukuji, Y et al. (2021) Whole-blood metabolomics of dementia patients reveal classes of disease-linked metabolites. Proc Natl Acad Sci 118, e2022857118.Google Scholar
Wu, LY, Cheah, IK, Chong, JR et al. (2021) Low plasma ergothioneine levels are associated with neurodegeneration and cerebrovascular disease in dementia. Free Radic Biol Med 177, 201211.Google Scholar
Hatano, T, Saiki, S, Okuzumi, A et al. (2016) Identification of novel biomarkers for Parkinson’s disease by metabolomic technologies. J Neurol Neurosurg Psychiatry 87, 295301.Google Scholar
Novotny, BC, Fernandez, MV, Wang, C et al. (2023) Metabolomic and lipidomic signatures in autosomal dominant and late-onset Alzheimer’s disease brains. Alzheimer’s Dementia 19, 17851799.Google Scholar
Cao, D, Zhang, Y, Zhang, S et al. (2024). Risk of Alzheimer’s disease and genetically predicted levels of 1400 plasma metabolites: a Mendelian randomization study. Sci Rep 14, 26078.Google Scholar
Sotgia, S, Mangoni, AA, Zoroddu, S et al. (2024) Higher scores of the kessler psychological distress scale (k10) are associated with lower serum ergothioneine and higher serum asymmetric dimethyl-l-arginine concentrations in a cohort of middle-aged and older adults. Clin Nutr ESPEN 64, 107113.Google Scholar
Liu, J & Pan, R. (2024) Genetic liability to human serum metabolites is causally linked to telomere length: insights from genome-wide Mendelian randomization and metabolic pathways analysis. Front Nutr 11. doi: 10.3389/fnut.2024.1458442.Google Scholar
Xu, R, Liu, S, Li, L-Y et al. (2024) Exploring the causal association between serum metabolites and erectile dysfunction: a bidirectional Mendelian randomisation study. Int J Impot Res 37601611. doi: 10.1038/s41443-024-00926-2.Google Scholar
Ahmad, S, Wu, T, Arnold, M et al. (2024) The blood metabolome of cognitive function and brain health in middle-aged adults – influences of genes, gut microbiome, and exposome. MedRxiv. doi: 10.1101/2024.12.16.24317793.Google Scholar
Kenny, LC, Brown, LW, Ortea, P et al. (2023) Relationship between the concentration of ergothioneine in plasma and the likelihood of developing pre-eclampsia. Biosci Rep 43. doi: 10.1042/BSR20230160.Google Scholar
Nierenberg, JL, He, J, Li, C et al. (2020) Serum metabolites associate with physical performance among middle-aged adults: evidence from the bogalusa heart study. Aging 12, 1191411941.Google Scholar
Smith, E, Ottosson, F, Hellstrand, S et al. (2020) Ergothioneine is associated with reduced mortality and decreased risk of cardiovascular disease. Heart 106, 691697.Google Scholar
Beelman, RB, Kalaras, MD, Phillips, AT et al. (2020) Is ergothioneine a ‘longevity vitamin’ limited in the American diet? J Nutr Sci 9, e52.Google Scholar
Solano-Aguilar, G, Matuszek, G, Matthan, NR et al. (2024) Differential regulation of brain microvessel transcriptome and brain metabolome by western and heart-healthy dietary patterns in Ossabaw pigs. Sci Rep 14, 29621.Google Scholar
Matsuda, Y, Ozawa, N, Shinozaki, T et al. (2020) Ergothioneine, a metabolite of the gut bacterium Lactobacillus reuteri, protects against stress-induced sleep disturbances. Transl Psychiatry 10, 170.Google Scholar
Nakamichi, N, Nakayama, K, Ishimoto, T et al. (2016) Food-derived hydrophilic antioxidant ergothioneine is distributed to the brain and exerts antidepressant effect in mice. Brain Behav 6. doi: 10.1002/brb3.477.Google Scholar
Katsube, M, Watanabe, H, Suzuki, K et al. (2022) Food-derived antioxidant ergothioneine improves sleep difficulties in humans. J Funct Foods 95, 105165.Google Scholar
Zhu, D, Wu, JZ, Griffin, P et al. (2025) Metabolomics biomarkers of frailty: a longitudinal study of aging female and male mice. npj Aging 11. doi: 10.1038/s41514-025-00237-w.Google Scholar
Cheah, IK, Lee, JZ, Tang, RMY et al. (2022) Does Lactobacillus reuteri influence ergothioneine levels in the human body? FEBS Lett 596, 12411251.Google Scholar
Intili, G, Paladino, L, Rappa, F et al. (2023) From dysbiosis to neurodegenerative diseases through different communication pathways: an overview. Biol (Basel) 12, 195.Google Scholar
Turck, D, Bresson, J, Burlingame, B et al. (2016) Safety of synthetic l-ergothioneine (Ergoneine®) as a novel food pursuant to Regulation (EC) No 258/97. EFSA J 14, e04629. doi: 10.2903/j.efsa.2016.4629.Google Scholar
Adams, M & Keefe, DM. Agency Response Letter GRAS Notice No. GRN 000736 - FDA has no questions. https://www.fda.gov/media/113846/download Google Scholar
Fennema, D, Phillips, IR & Shephard, EA. (2016) Trimethylamine and Trimethylamine N-Oxide, a flavin-containing Monooxygenase 3 (FMO3)-Mediated Host-Microbiome Metabolic axis implicated in health and disease. Drug Metab Dispos 44, 18391850.Google Scholar
Monti, JM, Moulton, CJ & Cohen, NJ. (2015) The role of nutrition on cognition and brain health in ageing: a targeted approach. Nutr Res Rev 28, 167180.Google Scholar
Toh, DSL, Cheung, FSG, Murray, M et al. (2013) Functional analysis of novel variants in the organic cation/ergothioneine transporter 1 identified in Singapore populations. Mol Pharm 10, 25092516.Google Scholar
Morris, MC. (2011) A potential design flaw of randomized trials of vitamin supplements. JAMA 305, 13481349.Google Scholar
Scarmeas, N, Anastasiou, CA & Yannakoulia, M. (2018) Nutrition and prevention of cognitive impairment. Lancet Neurol 17, 10061015.Google Scholar
Macready, AL, Butler, LT, Kennedy, OB et al. (2010) Cognitive tests used in chronic adult human randomised controlled trial micronutrient and phytochemical intervention studies. Nutr Res Rev 23, 200229.Google Scholar
Schöll, M, Verberk, IMW, del Campo, M et al. (2024) Challenges in the practical implementation of blood biomarkers for Alzheimer’s disease. Lancet Healthy Longev 5, 100630.Google Scholar
Jessen, F. (2010) Prediction of Dementia by subjective memory impairment. Arch Gen Psychiatry 67, 414422.Google Scholar
Ferry, M, Coley, N, Andrieu, S et al. (2013) How to design nutritional intervention trials to slow cognitive decline in apparently healthy populations and apply for efficacy claims: a statement from the international academy on nutrition and aging task force. J Nutr Health Aging 17, 619623.Google Scholar
Ishimoto, T & Kato, Y. (2022) Ergothioneine in the brain. FEBS Lett 596, 12901298.Google Scholar
Galluccio, M, Tripicchio, M & Pochini, L. (2024) The human OCTN sub-family: gene and protein structure, expression, and regulation. Int J Mol Sci 25, 8743.Google Scholar
Behof, WJ, Whitmore, CA, Haynes, JR et al. (2022) Improved synthesis of an ergothioneine PET radioligand for imaging oxidative stress in Alzheimer’s disease. FEBS Lett 596, 12791289.Google Scholar
Zhu, BZ, Mao, L, Fan, RM et al. (2011) Ergothioneine prevents copper-induced oxidative damage to dna and protein by forming a redox-inactive ergothioneine−copper complex. Chem Res Toxicol 24, 3034.Google Scholar
Akanmu, D, Cecchini, R, Aruoma, OI et al. (1991) The antioxidant action of ergothioneine. Arch Biochem Biophys 288, 1016.Google Scholar
Samuel, P, Tsapekos, M, de Pedro, N, et al. (2022) Ergothioneine mitigates telomere shortening under oxidative stress conditions. J Diet Suppl 19, 212225.Google Scholar
Song, TY, Chen, CL, Liao, JW et al. (2010) Ergothioneine protects against neuronal injury induced by cisplatin both in vitro and in vivo. Food Chem Toxicol 48, 34923499.Google Scholar
Petrovic, D, Slade, L, Paikopoulos, Y et al. (2025) Ergothioneine improves healthspan of aged animals by enhancing cGPDH activity through CSE-dependent persulfidation. Cell Metab 37, 542556.Google Scholar
Yuzawa, S, Nakashio, M, Ichimura, S et al. (2024) Ergothioneine prevents neuronal cell death caused by the neurotoxin 6-hydroxydopamine. Cells 13, 230.Google Scholar
Leow, DMK, Cheah, IKM, Chen, L et al. (2024) Ergothioneine-mediated neuroprotection of human iPSC-derived dopaminergic neurons. Antioxidants (Basel) 13, 693.Google Scholar
Meng, X, Meng, H, Hou, S et al. (2024) L-Ergothioneine ameliorates MPTP-induced Parkinson’s disease in C57BL/6J mice by activating DJ-1. Food Sci Hum Wellness 14, 9250068.Google Scholar
Nakamichi, N, Nakao, S, Nishiyama, M et al. (2020) Oral administration of the food-derived hydrophilic antioxidant ergothioneine enhances object recognition memory in mice. Curr Mol Pharmacol 14, 220233.Google Scholar
Zhang, W, Xiao, D, Mao, Q et al. (2023) Role of neuroinflammation in neurodegeneration development. Signal Transduct Target Ther 8, 267.Google Scholar
Ishimoto, T, Nakamichi, N, Nishijima, H et al. (2018) Carnitine/organic cation transporter OCTN1 negatively regulates activation in murine cultured microglial cells. Neurochem Res 43, 116128.Google Scholar
Navarro, A, Boveris, A. (2010) Brain mitochondrial dysfunction in aging, neurodegeneration and Parkinson’s disease. Front Aging Neurosci 2. doi: 10.3389/fnagi.2010.00034.Google Scholar
Leow, DMK, Cheah, IKM, Fong, ZWJ et al. (2023) Protective effect of ergothioneine against 7-ketocholesterol-induced mitochondrial damage in hCMEC/D3 human brain endothelial cells. Int J Mol Sci 24, 5498.Google Scholar
Fong, ZW, Tang, RMY, Cheah, IKM et al. (2024) Ergothioneine and mitochondria: an important protective mechanism? Biochem Biophys Res Commun 726, 150269.Google Scholar
Ishimoto, T, Masuo, Y, Kato, Y et al. (2019) Ergothioneine-induced neuronal differentiation is mediated through activation of S6K1 and neurotrophin 4/5-TrkB signaling in murine neural stem cells. Cell Signal 53, 269280.Google Scholar
Shi, C, Asaba, S, Nakamura, S et al. (2025) Ergothioneine stimulates Ca 2+ -mediated brain-derived neurotrophic factor expression in NE-4C nerve cells. ACS Omega 10, 70047012.Google Scholar
Cheah, IK, Ng, L, Ng, L et al. (2019) Inhibition of amyloid-induced toxicity by ergothioneine in a transgenic Caenorhabditis elegans model. FEBS Lett 593, 21392150.Google Scholar
Gao, W, Wang, Y, Wang, F et al. (2025) Ergothioneine exerts neuroprotective effects in Parkinson’s disease: targeting α-synuclein aggregation and oxidative stress. Food Res Int 201, 115590.Google Scholar
Jang, JH, Aruoma, OI, Jen, LS et al. (2004) Ergothioneine rescues PC12 cells from β-amyloid-induced apoptotic death. Free Radic Biol Med 36, 288299.Google Scholar
Yang, NC, Lin, HC, Wu, JH et al. (2012) Ergothioneine protects against neuronal injury induced by β-amyloid in mice. Food Chem Toxicol 50(11), 39023911.Google Scholar
Vignane, T, Hugo, M, Hoffmann, C et al. (2023) Protein thiol alterations drive aberrant phase separation in aging. BioRxiv. doi: 10.1101/2023.11.07.566021.Google Scholar
Petrovic, D, Kouroussis, E, Vignane, T et al. (2021) The role of protein persulfidation in brain aging and neurodegeneration. Front Aging Neurosci 13. doi: 10.3389/fnagi.2021.674135 Google Scholar
Sprenger, HG, Mittenbühler, MJ, Sun, Y et al. (2025) Ergothioneine controls mitochondrial function and exercise performance via direct activation of MPST. Cell Metab 37, 857869.Google Scholar
Lautrup, S, Sinclair, DA, Mattson, MP et al. (2019) NAD+ in brain aging and neurodegenerative disorders. Cell Metab 30, 630655.Google Scholar
Lu, Y, Yang, J, Wu, Q et al. (2025) The Role and molecular pathways of SIRT6 in senescence and age-related diseases. Adv Biol 9, e2400469.Google Scholar
D’Onofrio, N, Servillo, L, Giovane, A et al. (2016) Ergothioneine oxidation in the protection against high-glucose induced endothelial senescence: involvement of SIRT1 and SIRT6. Free Radic Biol Med 96, 211222.Google Scholar
Wang, Z, Ma, J, Miao, Z et al. (2023) Ergothioneine inhibits the progression of osteoarthritis via the Sirt6/NF-κB axis both in vitro and in vivo. Int Immunopharmacol 119, 110211.Google Scholar
Pan, HY, Ye, ZW, Zheng, QW et al. (2022) Ergothioneine exhibits longevity-extension effect in Drosophila melanogaster via regulation of cholinergic neurotransmission, tyrosine metabolism, and fatty acid oxidation. Food Funct 13, 227241.Google Scholar
Fovet, T, Guilhot, C, Delobel, P et al. (2022) Ergothioneine improves aerobic performance without any negative effect on early muscle recovery signaling in response to acute exercise. Front Physiol 9, 13:834597.Google Scholar
Pan, H, Zheng, Q, Zou, Y et al. (2024). Geroprotection from ergothioneine treatment in Drosophila melanogaster by improving intestinal barrier and activation of intestinal autophagy. Food Sci Hum Wellness 13, 34343446.Google Scholar
Pang, L, Wang, T, Liao, Q et al. (2022). Protective role of ergothioneine isolated from Pleurotus ostreatus against dextran sulfate sodium-induced ulcerative colitis in rat model. J Food Sci 87, 415426.Google Scholar
Shimizu, T, Masuo, Y, Takahashi, S et al. (2015). Organic cation transporter Octn1-mediated uptake of food-derived antioxidant ergothioneine into infiltrating macrophages during intestinal inflammation in mice. Drug Metab Pharmacokinet 30, 231239.Google Scholar
Gao, Y, Zhou, B, Zhang, H et al. (2022) l-Ergothioneine exhibits protective effects against dextran sulfate sodium-induced colitis in mice. ACS Omega 7, 2155421565.Google Scholar
Liu, Y, Wang, C, Liu, R et al. (2023) Adhesive ergothioneine hyaluronate gel protects against radiation gastroenteritis by alleviating apoptosis, inflammation, and gut microbiota dysbiosis. ACS Appl Mater Interfaces 15, 1983319846.Google Scholar
Mao, Y, Xie, Z, Zhang, X et al. (2024) Ergothioneine ameliorates liver fibrosis by inhibiting glycerophospholipids metabolism and TGF-β/Smads signaling pathway: based on metabonomics and network pharmacology. J Appl Toxicol 45(3), 361530. doi: 10.1002/jat.4728.Google Scholar
Xiong, K, Guo, H, Jia, Y et al. (2024) Supplement of food functional factor ergothioneine can effectively prevent liver injury in mice. Food Biosci 57, 103530 Google Scholar
Lv, X, Nie, C, Shi, Y et al. (2024) Ergothioneine ameliorates metabolic dysfunction-Associated Steatotic Liver Disease (MASLD) by enhancing autophagy, inhibiting oxidative damage and inflammation. Lipids Health Dis 23, 395.Google Scholar
Zhao, W, Wu, F, Hu, R et al. (2025) The antioxidant ergothioneine alleviates cisplatin-induced hearing loss through the Nrf2 pathway. Antioxid Redox Signal 42, 97114.Google Scholar
Bauer, MA, Bazard, P, Acosta, AA et al. (2024) L-Ergothioneine slows the progression of age-related hearing loss in CBA/CaJ mice. Hear Res 446, 109004.Google Scholar
Kim, EC, Toyono, T, Berlinicke, CA et al. (2017) Screening and characterization of drugs that protect corneal endothelial cells against unfolded protein response and oxidative stress. Invest. Opthalmology Vis. Science 58, 892900.Google Scholar
Gu, S, Wu, S, Lin, Z et al. (2024) Screening and evaluation of antioxidants for retinal pigment epithelial cell protection: L-ergothioneine as a novel therapeutic candidate through NRF2 activation. Exp Eye Res 242, 109862.Google Scholar
Evans, VJ, Wu, X, Tran, KK et al. (2024) Impact of aging and ergothioneine pre-treatment on naphthalene toxicity in lung. Toxicol Lett 397, 89102.Google Scholar
Iqbal, S, Jabeen, F, Aslam, N et al. (2023) Anti-EMT properties of ergothioneine attenuate lipopolysaccharide-induced oxidative stress-mediated acute lung injury via modulating TGF-β/smad/snail signaling pathway. Hum Exp Toxicol 42. doi: 10.1177/09603271231178015.Google Scholar
Repine, JE, Elkins, ND. (2012) Effect of ergothioneine on acute lung injury and inflammation in cytokine insufflated rats. Prev Med (Baltim) 54, S7982.Google Scholar
Rahman, I, Gilmour, PS, Jimenez, LA et al. (2003) Ergothioneine inhibits oxidative stress- and TNF-α-induced NF-κB activation and interleukin-8 release in alveolar epithelial cells. Biochem Biophys Res Commun 302, 860864.Google Scholar
Dare, A, Elrashedy, AA, Channa, ML et al. (2022) Cardioprotective effects and in-silico antioxidant mechanism of L-ergothioneine in experimental type-2 diabetic rats. Cardiovasc Hematol Agents Med Chem 20, 133147.Google Scholar
Duan, R, Pan, H, Li, D et al. (2023) Ergothioneine improves myocardial remodeling and heart function after acute myocardial infarction via S-glutathionylation through the NF-ĸB dependent Wnt5a-sFlt-1 pathway. Eur J Pharmacol 950, 175759.Google Scholar
Cheah, IK, Halliwell, B. (2020) Could ergothioneine aid in the treatment of coronavirus patients? Antioxidants (Basel) 9, 595.Google Scholar
Li, A, Liu, Y, Yu, P et al. (2025) Ergothioneine attenuates psoriasis symptoms through modulation of M1/M2 macrophage polarisation via the NF-κB/JAK-STAT3 pathway. Front Pharmacol 16. doi: 10.3389/fphar.2025.1521743.Google Scholar
Yoshida, S, Shime, H, Funami, K et al. (2017) The anti-oxidant ergothioneine augments the immunomodulatory function of tlr agonists by direct action on macrophages PLoS One 12, e0169360.Google Scholar
Williamson, RD, McCarthy, FP, Manna, S et al. (2020) L-(+)-ergothioneine significantly improves the clinical characteristics of preeclampsia in the reduced uterine perfusion pressure rat model. Hypertension 75, 561568.Google Scholar
Morillon, AC, Williamson, RD, Baker, PN et al. (2020) Effect of L-ergothioneine on the metabolic plasma profile of the RUPP rat model of pre-eclampsia. PLoS One 15, e0230977.Google Scholar
McElwain, CJ, Musumeci, A, Manna, S et al. (2024) L-ergothioneine reduces mitochondrial-driven NLRP3 activation in gestational diabetes mellitus. J Reprod Immunol 161, 104171.Google Scholar
Guo, Z, Huang, Y, Wang, X et al. (2025) Ergothioneine alleviates osteoporosis via the ROS-MAPK signaling axis. Bone 197, 117496.Google Scholar
Sakata, S, Kunimatsu, R & Tanimoto, K. (2024) Protective effect of ergothioneine against oxidative stress-induced chondrocyte death. Antioxidants (Basel) 13, 800.Google Scholar
Benson, KF, Ager, DM, Landes, B et al. (2012) Improvement of joint range of motion (ROM) and reduction of chronic pain after consumption of an ergothioneine-containing nutritional supplement. Prev Med (Baltim) 54, S839.Google Scholar
Tsay, GJ, Lin, SY, Li, CY et al. (2021) Comparison of single and combined use of ergothioneine, ferulic acid, and glutathione as antioxidants for the prevention of ultraviolet B radiation-induced photoaging damage in human skin fibroblasts. Processes 9, 1204.Google Scholar
Ko, HJ, Kim, J, Ahn, M et al. (2021) Ergothioneine alleviates senescence of fibroblasts induced by UVB damage of keratinocytes via activation of the Nrf2/HO-1 pathway and HSP70 in keratinocytes. Exp Cell Res 400, 112516.Google Scholar
Hseu, YC, Vudhya Gowrisankar, Y, Chen, XZ et al. (2020) The antiaging activity of ergothioneine in UVA-irradiated human dermal fibroblasts via the inhibition of the AP-1 pathway and the activation of Nrf2-mediated antioxidant genes. Oxid Med Cell Longev 2020, 113.Google Scholar
Hseu, YC, Lo, HW, Korivi, M et al. (2015) Dermato-protective properties of ergothioneine through induction of Nrf2/ARE-mediated antioxidant genes in UVA-irradiated Human keratinocytes. Free Radic Biol Med 86, 102117.Google Scholar
Bazela, K, Solyga-Zurek, A, Debowska, R et al. (2014) L-ergothioneine protects skin cells against UV-induced damage-a preliminary study. Cosmetics 1, 5160.Google Scholar
Markova, NG, Karaman-Jurukovska, N, Dong, KK et al. (2009) Skin cells and tissue are capable of using l-ergothioneine as an integral component of their antioxidant defense system. Free Radic Biol Med 46, 11681176.Google Scholar
Botta, C, Di Giorgio, C, Sabatier, AS et al. (2008). Genotoxicity of visible light (400-800 nm) and photoprotection assessment of ectoin, l-ergothioneine and mannitol and four sunscreens. J Photochem Photobiol B 91, 2434.Google Scholar
Dong, KK, Damaghi, N, Kibitel, J et al. (2017) A comparison of the relative antioxidant potency of L-ergothioneine and idebenone. J Cosmet Dermatol 6, 183188.Google Scholar
Bartosz, G, Pieńkowska, N, Sadowska-Bartosz, I. (2024) Effect of selected antioxidants on the in vitro aging of human fibroblasts. Int J Mol Sci 25, 1529.Google Scholar
Hanayama, M, Mori, K, Ishimoto, T et al. (2024) Effects of an ergothioneine-rich Pleurotus sp. on skin moisturizing functions and facial conditions: a randomized, double-blind, placebo-controlled trial. Front Med (Lausanne) 11, 1396783.Google Scholar
Tian, X, Guo, J, Gu, C, et al. (2024) Ergothioneine–sodium hyaluronate dressing: a promising approach for protecting against radiation-induced skin injury. ACS Appl Mater Interfaces 16, 2991729929.Google Scholar
Wang, H, Wang, Y, Li, X et al. (2025) Ergothioneine regulates the paracrine of dermal papilla cells through SIRT1/Nrf2 pathway to antagonize oxidative stress and natural hair follicle aging. Free Radic Biol Med 237, 570584.Google Scholar
Thomas, TA, Francis, RO, Zimring, JC et al. (2024) The role of ergothioneine in red blood cell biology: a review and perspective. Antioxidants (Basel) 13, 717.Google Scholar
Deiana, M, Rosa, A, Casu, V et al. (2004) Ergothioneine modulates oxidative damage in the kidney and liver of rats in vivo: studies upon the profile of polyunsaturated fatty acids. Clin Nutrition 23, 183193.Google Scholar
Salama, SA, Abd-Allah, GM, Mohamadin, AM et al. (2021) Ergothioneine mitigates cisplatin-evoked nephrotoxicity via targeting Nrf2, NF-κB, and apoptotic signaling and inhibiting γ-glutamyl transpeptidase. Life Sci 278, 119572.Google Scholar
Shinozaki, Y, Furuichi, K, Toyama, T et al. (2017) Impairment of the carnitine/organic cation transporter 1–ergothioneine axis is mediated by intestinal transporter dysfunction in chronic kidney disease. Kidney Int 92, 13561369.Google Scholar
Suba, JK, Keo, LS & Sirich, TL. (2024) Depletion by hemodialysis of the antioxidant ergothioneine. Kidney360 6, 265271.Google Scholar
Bartman, S, Coppotelli, G & Ross, JM. (2024) Mitochondrial dysfunction: a key player in brain aging and diseases. Curr Issues Mol Biol 46, 19872026.Google Scholar
Yang, J, Luo, J, Tian, X et al. (2024) Progress in understanding oxidative stress, aging, and aging-related diseases. Antioxidants (Basel) 13, 394.Google Scholar
Maldonado, E, Morales-Pison, S, Urbina, F et al. (2023) Aging hallmarks and the role of oxidative stress. Antioxidants (Basel) 12, 651.Google Scholar
Pratti, PG, Pinho, J da S Junior, Petito, N de L et al. (2024) Effect of hot air-drying and pasteurization on ergothioneine content in edible mushrooms. J Food Compos Anal 126, 105865.Google Scholar
Nguyen, TH, Nagasaka, R & Ohshima, T. (2012) Effects of extraction solvents, cooking procedures and storage conditions on the contents of ergothioneine and phenolic compounds and antioxidative capacity of the cultivated mushroom Flammulina velutipes. Int J Food Sci Technol 47, 11931205.Google Scholar
Yadav, H, Jaldhi, Bhardwaj R et al. (2023) Unveiling the role of gut-brain axis in regulating neurodegenerative diseases: a comprehensive review. Life Sci 330, 122022.Google Scholar