Ergothioneine as a Natural Antioxidant Against Oxidative Stress-Related Diseases

Tong-Tong Fu, Liang Shen, Tong-Tong Fu, Liang Shen

Abstract

L-Ergothioneine (EGT) is a natural antioxidant derived from microorganisms, especially in edible mushrooms. EGT is found to be highly accumulated in tissues that are susceptible to oxidative damage, and it has attracted extensive attention due to its powerful antioxidant activity and the tight relationships of this natural product with various oxidative stress-related diseases. Herein, we 1) introduce the biological source and in vivo distribution of EGT; 2) review the currently available evidence concerning the relationships of EGT with diabetes, ischemia-reperfusion injury-related diseases like cardiovascular diseases and liver diseases, neurodegenerative diseases, and other diseases pathogenically associated with oxidative stress; 3) summarize the potential action mechanisms of EGT against these diseases; 4) discuss the advantages of EGT over other antioxidants; and 5) also propose several future research perspectives for EGT. These may help to promote the future application of this attractive natural antioxidant.

Keywords: L-ergothioneine; action mechanism; antioxidant; disease; oxidative stress.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Fu and Shen.

Figures

FIGURE 1
FIGURE 1
Antioxidant properties and potential action mechanisms of ergothioneine. (A). Chemical structure of ergothioneine; (B). The main antioxidant properties and cytoprotective effects of ergothioneine; (C). Potential mechanisms underlying the relationships between ergothioneine and oxidative stress-related disease conditions.

References

    1. Akanmu D., Cecchini R., Aruoma O. I., Halliwell B. (1991). The Antioxidant Action of Ergothioneine. Arch. Biochem. Biophys. 288, 10–16. 10.1016/0003-9861(91)90158-f
    1. Albiero M., Poncina N., Tjwa M., Ciciliot S., Menegazzo L., Ceolotto G., et al. (2014). Diabetes Causes Bone Marrow Autonomic Neuropathy and Impairs Stem Cell Mobilization via Dysregulated p66Shc and Sirt1. Diabetes 63, 1353–1365. 10.2337/db13-0894
    1. Arduini A., Eddy L., Hochstein P. (1990). The Reduction of Ferryl Myoglobin by Ergothioneine: a Novel Function for Ergothioneine. Arch. Biochem. Biophys. 281, 41–43. 10.1016/0003-9861(90)90410-z
    1. Arduini A., Mancinelli G., Radatti G. L., Hochstein P., Cadenas E. (1992). Possible Mechanism of Inhibition of Nitrite-Induced Oxidation of Oxyhemoglobin by Ergothioneine and Uric Acid. Arch. Biochem. Biophys. 294, 398–402. 10.1016/0003-9861(92)90702-x
    1. Aruoma O. I., Whiteman M., England T. G., Halliwell B. (1997). Antioxidant Action of Ergothioneine: Assessment of its Ability to Scavenge Peroxynitrite. Biochem. Biophys. Res. Commun. 231, 389–391. 10.1006/bbrc.1997.6109
    1. Asahi T., Wu X., Shimoda H., Hisaka S., Harada E., Kanno T., et al. (2016). A Mushroom-Derived Amino Acid, Ergothioneine, Is a Potential Inhibitor of Inflammation-Related DNA Halogenation. Biosci. Biotechnol. Biochem. 80, 313–317. 10.1080/09168451.2015.1083396
    1. Astwood E. B., Stanley M. M. (1947). Antithyroid Activity of Ergothioneine in Man. Lancet 2, 905–907. 10.1016/s0140-6736(47)90914-8
    1. Bedirli A., Sakrak O., Muhtaroglu S., Soyuer I., Guler I., Riza Erdogan A., et al. (2004). Ergothioneine Pretreatment Protects the Liver from Ischemia-Reperfusion Injury Caused by Increasing Hepatic Heat Shock Protein 70. J. Surg. Res. 122, 96–102. 10.1016/j.jss.2004.06.016
    1. Borodina I., Kenny L. C., McCarthy C. M., Paramasivan K., Pretorius E., Roberts T. J., et al. (2020). The Biology of Ergothioneine, an Antioxidant Nutraceutical. Nutr. Res. Rev. 33, 190–217. 10.1017/S0954422419000301
    1. Brown E. G., Goldman S. M. (2020). Modulation of the Microbiome in Parkinson's Disease: Diet, Drug, Stool Transplant, and beyond. Neurotherapeutics 17, 1406–1417. 10.1007/s13311-020-00942-2
    1. Brummel M. C. (1989). In Search of a Physiological Function for L-Ergothioneine--II. Med. Hypotheses. 30, 39–48. 10.1016/0306-9877(89)90123-0
    1. Calvo M. S., Mehrotra A., Beelman R. B., Nadkarni G., Wang L., Cai W., et al. (2016). A Retrospective Study in Adults with Metabolic Syndrome: Diabetic Risk Factor Response to Daily Consumption of Agaricus Bisporus (White Button Mushrooms). Plant Foods Hum. Nutr. 71, 245–251. 10.1007/s11130-016-0552-7
    1. Chaudière J., Ferrari-Iliou R. (1999). Intracellular Antioxidants: from Chemical to Biochemical Mechanisms. Food Chem. Toxicol. 37, 949–962. 10.1016/s0278-6915(99)00090-3
    1. Cheah I. K., Feng L., Tang R. M. Y., Lim K. H. C., Halliwell B. (2016a). Ergothioneine Levels in an Elderly Population Decrease with Age and Incidence of Cognitive Decline; a Risk Factor for Neurodegeneration? Biochem. Biophys. Res. Commun. 478, 162–167. 10.1016/j.bbrc.2016.07.074
    1. Cheah I. K., Halliwell B. (2020). Could Ergothioneine Aid in the Treatment of Coronavirus Patients? Antioxidants (Basel) 9, 595. 10.3390/antiox9070595
    1. Cheah I. K., Halliwell B. (2021). Ergothioneine, Recent Developments. Redox Biol. 42, 101868. 10.1016/j.redox.2021.101868
    1. Cheah I. K., Halliwell B. (2012). Ergothioneine; Antioxidant Potential, Physiological Function and Role in Disease. Biochim. Biophys. Acta 1822, 784–793. 10.1016/j.bbadis.2011.09.017
    1. Cheah I. K., Tang R., Ye P., Yew T. S., Lim K. H., Halliwell B. (2016b). Liver Ergothioneine Accumulation in a guinea Pig Model of Non-alcoholic Fatty Liver Disease. A Possible Mechanism of Defence? Free Radic. Res. 50, 14–25. 10.3109/10715762.2015.1099642
    1. Cheah I. K., Tang R. M., Yew T. S., Lim K. H., Halliwell B. (2017). Administration of Pure Ergothioneine to Healthy Human Subjects: Uptake, Metabolism, and Effects on Biomarkers of Oxidative Damage and Inflammation. Antioxid. Redox Signal. 26, 193–206. 10.1089/ars.2016.6778
    1. Chen S.-Y., Ho K.-J., Hsieh Y.-J., Wang L.-T., Mau J.-L. (2012). Contents of Lovastatin, γ-aminobutyric Acid and Ergothioneine in Mushroom Fruiting Bodies and Mycelia. Lwt 47, 274–278. 10.1016/j.lwt.2012.01.019
    1. Colognato R., Laurenza I., Fontana I., Coppedé F., Siciliano G., Coecke S., et al. (2006). Modulation of Hydrogen Peroxide-Induced DNA Damage, MAPKs Activation and Cell Death in PC12 by Ergothioneine. Clin. Nutr. 25, 135–145. 10.1016/j.clnu.2005.10.005
    1. D'Onofrio N., Servillo L., Giovane A., Casale R., Vitiello M., Marfella R., et al. (2016). Ergothioneine Oxidation in the protection against High-Glucose Induced Endothelial Senescence: Involvement of SIRT1 and SIRT6. Free Radic. Biol. Med. 96, 211–222. 10.1016/j.freeradbiomed.2016.04.013
    1. Deiana M., Rosa A., Casu V., Piga R., Assunta Dessì M., Aruoma O. I. (2004). L-ergothioneine Modulates Oxidative Damage in the Kidney and Liver of Rats In Vivo: Studies upon the Profile of Polyunsaturated Fatty Acids. Clin. Nutr. 23, 183–193. 10.1016/S0261-5614(03)00108-0
    1. Dransfeld O., Gehrmann T., Köhrer K., Kircheis G., Holneicher C., Häussinger D., et al. (2005). Oligonucleotide Microarray Analysis of Differential Transporter Regulation in the Regenerating Rat Liver. Liver Int. 25, 1243–1258. 10.1111/j.1478-3231.2005.01158.x
    1. Dubost N., Ou B., Beelman R. (2007). Quantification of Polyphenols and Ergothioneine in Cultivated Mushrooms and Correlation to Total Antioxidant Capacity. Food Chem. 105, 727–735. 10.1016/j.foodchem.2007.01.030
    1. Ey J., Schömig E., Taubert D. (2007). Dietary Sources and Antioxidant Effects of Ergothioneine. J. Agric. Food Chem. 55, 6466–6474. 10.1021/jf071328f
    1. Félétou M., Vanhoutte P. M. (2006). Endothelial Dysfunction: a Multifaceted Disorder (The Wiggers Award Lecture). Am. J. Physiology-Heart Circulatory Physiol. 291, H985–H1002. 10.1152/ajpheart.00292.2006
    1. Feng L., Cheah I. K., Ng M. M., Li J., Chan S. M., Lim S. L., et al. (2019). The Association between Mushroom Consumption and Mild Cognitive Impairment: A Community-Based Cross-Sectional Study in Singapore. J. Alzheimers. Dis. 68, 197–203. 10.3233/JAD-180959
    1. Forster R., Spézia F., Papineau D., Sabadie C., Erdelmeier I., Moutet M., et al. (2015). Reproductive Safety Evaluation of L-Ergothioneine. Food Chem. Toxicol. 80, 85–91. 10.1016/j.fct.2015.02.019
    1. CiToxLAB France (2012). “2-week Preliminary Toxicity Study by the Oral Route (Dietary Admixture) in Rats,” in Laboratory Study Number: 39240 TSR. Evreux, France.
    1. CiToxLAB France (2013a). Acute oral Toxicity Study in Rats “Acute Toxic Class Method”. Laboratory Study Number:39664 TAR. Evreux, France.
    1. CiToxLAB France (2013b). Combined 13-Week Repeated Dose Toxicity Study With the Reproduction/Developmental Toxicity Screening Test by Oral Route (Dietary Admixture) in Rats. Laboratory Study Number: 39241 RSR. Evreux, France.
    1. Franzoni F., Colognato R., Galetta F., Laurenza I., Barsotti M., Di Stefano R., et al. (2006). An In Vitro Study on the Free Radical Scavenging Capacity of Ergothioneine: Comparison with Reduced Glutathione, Uric Acid and Trolox. Biomed. Pharmacother. 60, 453–457. 10.1016/j.biopha.2006.07.015
    1. Gamage A. M., Liao C., Cheah I. K., Chen Y., Lim D. R. X., Ku J. W. K., et al. (2018). The Proteobacterial Species Burkholderia Pseudomallei Produces Ergothioneine, Which Enhances Virulence in Mammalian Infection. FASEB J. 32, fj201800716. 10.1096/fj.201800716
    1. Genghof D. S. (1970). Biosynthesis of Ergothioneine and Hercynine by Fungi and Actinomycetales. J. Bacteriol. 103, 475–478. 10.1128/jb.103.2.475-478.1970
    1. Genghof D. S., Vandamme O. (1964). Biosynthesis of Ergothioneine and Hercynine by Mycobacteria. J. Bacteriol. 87, 852–862. 10.1128/jb.87.4.852-862.1964
    1. Gokce G., Arun M. Z. (2014). Ergothioneine Produces Relaxation in Isolated Rat Aorta by Inactivating Superoxide Anion. Eur. Rev. Med. Pharmacol. Sci. 18, 3339–3345.
    1. Grillo C. A., Piroli G. G., Rosell D. R., Hoskin E. K., Mcewen B. S., Reagan L. P. (2003). Region Specific Increases in Oxidative Stress and Superoxide Dismutase in the hippocampus of Diabetic Rats Subjected to Stress. Neuroscience 121, 133–140. 10.1016/s0306-4522(03)00343-9
    1. Gründemann D., Harlfinger S., Golz S., Geerts A., Lazar A., Berkels R., et al. (2005). Discovery of the Ergothioneine Transporter. Proc. Natl. Acad. Sci. U S A. 102, 5256–5261. 10.1073/pnas.0408624102
    1. Gründemann D. (2012). The Ergothioneine Transporter Controls and Indicates Ergothioneine Activity - A Review. Prev. Med. 54, S71–S74. 10.1016/j.ypmed.2011.12.001
    1. Halliwell B., Cheah I. K., Drum C. L. (2016). Ergothioneine, an Adaptive Antioxidant for the protection of Injured Tissues? A Hypothesis. Biochem. Biophys. Res. Commun. 470, 245–250. 10.1016/j.bbrc.2015.12.124
    1. Halliwell B., Cheah I. K., Tang R. M. Y. (2018). Ergothioneine - a Diet-Derived Antioxidant with Therapeutic Potential. FEBS Lett. 592, 3357–3366. 10.1002/1873-3468.13123
    1. Han Y., Tang X., Zhang Y., Hu X., Ren L. J. (2021). The Current Status of Biotechnological Production and the Application of a Novel Antioxidant Ergothioneine. Crit. Rev. Biotechnol. 41, 580–593. 10.1080/07388551.2020.1869692
    1. Hand C. E., Taylor N. J., Honek J. F. (2005). Ab Initio studies of the Properties of Intracellular Thiols Ergothioneine and Ovothiol. Bioorg. Med. Chem. Lett. 15, 1357–1360. 10.1016/j.bmcl.2005.01.014
    1. Hanlon D. P. (1971). Interaction of Ergothioneine with Metal Ions and Metalloenzymes. J. Med. Chem. 14, 1084–1087. 10.1021/jm00293a017
    1. Heath H. (1953). The Metabolism of 35S-Labelled 2-thiolhistidine and Ergothioneine in the Rat. Biochem. J. 54, 689–694. 10.1042/bj0540689
    1. Hu W., Song H., Sae Her A., Bak D. W., Naowarojna N., Elliott S. J., et al. (2014). Bioinformatic and Biochemical Characterizations of C-S Bond Formation and Cleavage Enzymes in the Fungus Neurospora Crassa Ergothioneine Biosynthetic Pathway. Org. Lett. 16, 5382–5385. 10.1021/ol502596z
    1. Hussain T., Tan B., Yin Y., Blachier F., Tossou M. C., Rahu N. (2016). Oxidative Stress and Inflammation: What Polyphenols Can Do for Us? Oxid. Med. Cel Longev. 2016, 7432797. 10.1155/2016/7432797
    1. Ito T., Kato M., Tsuchida H., Harada E., Niwa T., Osawa T. (2011). Ergothioneine as an Anti-Oxidative/Anti-inflammatory Component in Several Edible Mushrooms. Fstr 17, 103–110. 10.3136/fstr.17.103
    1. Jang J. H., Aruoma O. I., Jen L. S., Chung H. Y., Surh Y. J. (2004). Ergothioneine Rescues PC12 Cells from Beta-Amyloid-Induced Apoptotic Death. Free Radic. Biol. Med. 36, 288–299. 10.1016/j.freeradbiomed.2003.11.005
    1. Jong N. N., Nakanishi T., Liu J. J., Tamai I., McKeage M. J. (2011). Oxaliplatin Transport Mediated by Organic Cation/carnitine Transporters OCTN1 and OCTN2 in Overexpressing Human Embryonic Kidney 293 Cells and Rat Dorsal Root Ganglion Neurons. J. Pharmacol. Exp. Ther. 338, 537–547. 10.1124/jpet.111.181297
    1. Kalaras M. D., Richie J. P., Calcagnotto A., Beelman R. B. (2017). Mushrooms: A Rich Source of the Antioxidants Ergothioneine and Glutathione. Food Chem. 233, 429–433. 10.1016/j.foodchem.2017.04.109
    1. Kaneko I., Takeuchi Y., Yamaoka Y., Tanaka Y., Fukuda T., Fukumori Y., et al. (1980). Quantitative Determination of Ergothioneine in Plasma and Tissues by TLC-Densitometry. Chem. Pharm. Bull. (Tokyo) 28, 3093–3097. 10.1248/cpb.28.3093
    1. Kato Y., Kubo Y., Iwata D., Kato S., Sudo T., Sugiura T., et al. (2010). Gene Knockout and Metabolome Analysis of Carnitine/organic Cation Transporter OCTN1. Pharm. Res. 27, 832–840. 10.1007/s11095-010-0076-z
    1. Kawano H., Higuchi F., Mayumi T., Hama T. (1982a). Studies on Ergothioneine. VII. Some Effects on Ergothioneine on Glycolytic Metabolism in Red Blood Cells from Rats. Chem. Pharm. Bull. (Tokyo) 30, 2611–2613. 10.1248/cpb.30.2611
    1. Kawano H., Murata H., Iriguchi S., Mayumi T., Hama T. (1983). Studies on Ergothioneine. XI. Inhibitory Effect on Lipid Peroxide Formation in Mouse Liver. Chem. Pharm. Bull. (Tokyo) 31, 1682–1687. 10.1248/cpb.31.1682
    1. Kawano H., Otani M., Takeyama K., Kawai Y., Mayumi T., Hama T. (1982b). Studies on Ergothioneine. VI. Distribution and Fluctuations of Ergothioneine in Rats. Chem. Pharm. Bull. (Tokyo) 30, 1760–1765. 10.1248/cpb.30.1760
    1. Kerley R. N., McCarthy C., Kell D. B., Kenny L. C. (2018). The Potential Therapeutic Effects of Ergothioneine in Pre-eclampsia. Free Radic. Biol. Med. 117, 145–157. 10.1016/j.freeradbiomed.2017.12.030
    1. Kumosani T. A. (2001). L-ergothioneine Level in Red Blood Cells of Healthy Human Males in the Western Province of Saudi Arabia. Exp. Mol. Med. 33, 20–22. 10.1038/emm.2001.4
    1. Lai Y., Xue J., Liu C. W., Gao B., Chi L., Tu P., et al. (2019). Serum Metabolomics Identifies Altered Bioenergetics, Signaling Cascades in Parallel with Exposome Markers in Crohn's Disease. Molecules 24, 449. 10.3390/molecules24030449
    1. Lam-Sidun D., Peters K. M., Borradaile N. M. (2021). Mushroom-Derived Medicine? Preclinical Studies Suggest Potential Benefits of Ergothioneine for Cardiometabolic Health. Int. J. Mol. Sci. 22, 3246. 10.3390/ijms22063246
    1. Lamhonwah A. M., Hawkins C. E., Tam C., Wong J., Mai L., Tein I. (2008). Expression Patterns of the Organic Cation/carnitine Transporter Family in Adult Murine Brain. Brain Dev. 30, 31–42. 10.1016/j.braindev.2007.05.005
    1. Lamhonwah A. M., Tein I. (2006). Novel Localization of OCTN1, an Organic Cation/carnitine Transporter, to Mammalian Mitochondria. Biochem. Biophys. Res. Commun. 345, 1315–1325. 10.1016/j.bbrc.2006.05.026
    1. Lavelle A., Sokol H. (2020). Gut Microbiota-Derived Metabolites as Key Actors in Inflammatory Bowel Disease. Nat. Rev. Gastroenterol. Hepatol. 17, 223–237. 10.1038/s41575-019-0258-z
    1. Liao C., Seebeck F. P. (2017). Convergent Evolution of Ergothioneine Biosynthesis in Cyanobacteria. Chembiochem 18, 2115–2118. 10.1002/cbic.201700354
    1. Libby P., Ridker P. M., Hansson G. K. (2011). Progress and Challenges in Translating the Biology of Atherosclerosis. Nature 473, 317–325. 10.1038/nature10146
    1. Lo Y. C., Lin S. Y., Ulziijargal E., Chen S. Y., Chien R. C., Tzou Y. J., et al. (2012). Comparative Study of Contents of Several Bioactive Components in Fruiting Bodies and Mycelia of Culinary-Medicinal Mushrooms. Int. J. Med. Mushrooms. 14, 357–363. 10.1615/intjmedmushr.v14.i4.30
    1. Mackenzie J. B., Mackenzie C. G. (1957). The Effect of Age, Sex, and Androgen on Blood Ergothioneine. J. Biol. Chem. 225, 651–657. 10.1016/s0021-9258(18)64864-1
    1. Maeda T., Hirayama M., Kobayashi D., Miyazawa K., Tamai I. (2007). Mechanism of the Regulation of Organic Cation/carnitine Transporter 1 (SLC22A4) by Rheumatoid Arthritis-Associated Transcriptional Factor RUNX1 and Inflammatory Cytokines. Drug Metab. Dispos. 35, 394–401. 10.1124/dmd.106.012112
    1. Marone P. A., Trampota J., Weisman S. (2016). A Safety Evaluation of a Nature-Identical L-Ergothioneine in Sprague Dawley Rats. Int. J. Toxicol. 35, 568–583. 10.1177/1091581816653375
    1. Martel J., Ojcius D. M., Chang C. J., Lin C. S., Lu C. C., Ko Y. F., et al. (2017). Anti-obesogenic and Antidiabetic Effects of Plants and Mushrooms. Nat. Rev. Endocrinol. 13, 149–160. 10.1038/nrendo.2016.142
    1. Mayumi T., Kawano H., Sakamoto Y., Suehisa E., Kawai Y., Hama T. (1978). Studies on Ergothioneine. V. Determination by High Performance Liquid Chromatography and Application to Metabolic Research. Chem. Pharm. Bull. (Tokyo) 26, 3772–3778. 10.1248/cpb.26.3772
    1. McCarthy C. M., Kenny L. C. (2016). Mitochondrial [dys]function; Culprit in Pre-eclampsia? Clin. Sci. (Lond) 130, 1179–1184. 10.1042/CS20160103
    1. Melville D. B., Horner W. H., Lubschez R. (1954). Tissue Ergothioneine. J. Biol. Chem. 206, 221–228. 10.1016/s0021-9258(18)71313-6
    1. Muszyńska B., Grzywacz-Kisielewska A., Kała K., Gdula-Argasińska J. (2018). Anti-inflammatory Properties of Edible Mushrooms: A Review. Food Chem. 243, 373–381. 10.1016/j.foodchem.2017.09.149
    1. Nakamichi N., Nakayama K., Ishimoto T., Masuo Y., Wakayama T., Sekiguchi H., et al. (2016). Food-derived Hydrophilic Antioxidant Ergothioneine Is Distributed to the Brain and Exerts Antidepressant Effect in Mice. Brain Behav. 6, e00477. 10.1002/brb3.477
    1. Nakamura T., Yoshida K., Yabuuchi H., Maeda T., Tamai I. (2008). Functional Characterization of Ergothioneine Transport by Rat Organic Cation/carnitine Transporter Octn1 (Slc22a4). Biol. Pharm. Bull. 31, 1580–1584. 10.1248/bpb.31.1580
    1. Narainsamy K., Farci S., Braun E., Junot C., Cassier-Chauvat C., Chauvat F. (2016). Oxidative-stress Detoxification and Signalling in Cyanobacteria: the Crucial Glutathione Synthesis Pathway Supports the Production of Ergothioneine and Ophthalmate. Mol. Microbiol. 100, 15–24. 10.1111/mmi.13296
    1. Nielsen T. K., Højgaard M., Andersen J. T., Poulsen H. E., Lykkesfeldt J., Mikines K. J. (2015). Elimination of Ascorbic Acid after High-Dose Infusion in Prostate Cancer Patients: a Pharmacokinetic Evaluation. Basic Clin. Pharmacol. Toxicol. 116, 343–348. 10.1111/bcpt.12323
    1. Nikodemus D., Lazic D., Bach M., Bauer T., Pfeiffer C., Wiltzer L., et al. (2011). Paramount Levels of Ergothioneine Transporter SLC22A4 mRNA in Boar Seminal Vesicles and Cross-Species Analysis of Ergothioneine and Glutathione in Seminal Plasma. J. Physiol. Pharmacol. 62, 411–419.
    1. Paul B. D., Snyder S. H. (2010). The Unusual Amino Acid L-Ergothioneine Is a Physiologic Cytoprotectant. Cell Death Differ 17, 1134–1140. 10.1038/cdd.2009.163
    1. Petermann I., Triggs C. M., Huebner C., Han D. Y., Gearry R. B., Barclay M. L., et al. (2009). Mushroom Intolerance: a Novel Diet-Gene Interaction in Crohn's Disease. Br. J. Nutr. 102, 506–508. 10.1017/S0007114509276446
    1. Pfeiffer C., Bach M., Bauer T., Campos da Ponte J., Schömig E., Gründemann D. (2015). Knockout of the Ergothioneine Transporter ETT in Zebrafish Results in Increased 8-oxoguanine Levels. Free Radic. Biol. Med. 83, 178–185. 10.1016/j.freeradbiomed.2015.02.026
    1. Pfeiffer C., Bauer T., Surek B., Schömig E., Gründemann D. (2011). Cyanobacteria Produce High Levels of Ergothioneine. Food Chem. 129, 1766–1769. 10.1016/j.foodchem.2011.06.047
    1. Rajesh A. S., Dash B. P. (2018). “Ergothionine: It's Chemistry and Biological Significance,” in Recent Advances On Molecules Of Chemical And Biological Importance (A Ugc Sponsored National Seminar), Municipal College, Rourkela, February 6, 2018.
    1. Ravi R., Somani S. M., Rybak L. P. (1995). Mechanism of Cisplatin Ototoxicity: Antioxidant System. Pharmacol. Toxicol. 76, 386–394. 10.1111/j.1600-0773.1995.tb00167.x
    1. Rougee M., Bensasson R. V., Land E. J., Pariente R. (1988). Deactivation of Singlet Molecular Oxygen by Thiols and Related Compounds, Possible Protectors against Skin Photosensitivity. Photochem. Photobiol. 47, 485–489. 10.1111/j.1751-1097.1988.tb08835.x
    1. Sakrak O., Kerem M., Bedirli A., Pasaoglu H., Akyurek N., Ofluoglu E., et al. (2008). Ergothioneine Modulates Proinflammatory Cytokines and Heat Shock Protein 70 in Mesenteric Ischemia and Reperfusion Injury. J. Surg. Res. 144, 36–42. 10.1016/j.jss.2007.04.020
    1. Sao Emani C., Gallant J. L., Wiid I. J., Baker B. (2019). The Role of Low Molecular Weight Thiols in Mycobacterium tuberculosis. Tuberculosis (Edinb) 116, 44–55. 10.1016/j.tube.2019.04.003
    1. Sao Emani C., Williams M. J., Wiid I. J., Hiten N. F., Viljoen A. J., Pietersen R. D., et al. (2013). Ergothioneine Is a Secreted Antioxidant in Mycobacterium Smegmatis. Antimicrob. Agents Chemother. 57, 3202–3207. 10.1128/AAC.02572-12
    1. Schauss A. G., Béres E., Vértesi A., Frank Z., Pasics I., Endres J., et al. (2011). The Effect of Ergothioneine on Clastogenic Potential and Mutagenic Activity: Genotoxicity Evaluation. Int. J. Toxicol. 30, 405–409. 10.1177/1091581811405856
    1. Schubert D., Behl C., Lesley R., Brack A., Dargusch R., Sagara Y., et al. (1995). Amyloid Peptides Are Toxic via a Common Oxidative Mechanism. Proc. Natl. Acad. Sci. U S A. 92, 1989–1993. 10.1073/pnas.92.6.1989
    1. Servillo L., D'Onofrio N., Casale R., Cautela D., Giovane A., Castaldo D., et al. (2017b). Ergothioneine Products Derived by Superoxide Oxidation in Endothelial Cells Exposed to High-Glucose. Free Radic. Biol. Med. 108, 8–18. 10.1016/j.freeradbiomed.2017.03.009
    1. Servillo L., DʼOnofrio N., Balestrieri M. L. (2017a). Ergothioneine Antioxidant Function: From Chemistry to Cardiovascular Therapeutic Potential. J. Cardiovasc. Pharmacol. 69, 183–191. 10.1097/FJC.0000000000000464
    1. Shen L., Ji H. F. (2019). Bidirectional Interactions between Dietary Curcumin and Gut Microbiota. Crit. Rev. Food Sci. Nutr. 59, 2896–2902. 10.1080/10408398.2018.1478388
    1. Shen L., Liu L., Ji H. F. (2017). Alzheimer's Disease Histological and Behavioral Manifestations in Transgenic Mice Correlate with Specific Gut Microbiome State. J. Alzheimers Dis. 56, 385–390. 10.3233/JAD-160884
    1. Shimizu T., Masuo Y., Takahashi S., Nakamichi N., Kato Y. (2015). Organic Cation Transporter Octn1-Mediated Uptake of Food-Derived Antioxidant Ergothioneine into Infiltrating Macrophages during Intestinal Inflammation in Mice. Drug Metab. Pharmacokinet. 30, 231–239. 10.1016/j.dmpk.2015.02.003
    1. Shires T. K., Brummel M. C., Pulido J. S., Stegink L. D. (1997). Ergothioneine Distribution in Bovine and Porcine Ocular Tissues. Comp. Biochem. Physiol. C Pharmacol. Toxicol. Endocrinol. 117, 117–120. 10.1016/s0742-8413(96)00223-x
    1. Smith E., Ottosson F., Hellstrand S., Ericson U., Orho-Melander M., Fernandez C., et al. (2020). Ergothioneine Is Associated with Reduced Mortality and Decreased Risk of Cardiovascular Disease. Heart 106, 691–697. 10.1136/heartjnl-2019-315485
    1. Song T. Y., Chen C. L., Liao J. W., Ou H. C., Tsai M. S. (2010). Ergothioneine Protects against Neuronal Injury Induced by Cisplatin Both In Vitro and In Vivo . Food Chem. Toxicol. 48, 3492–3499. 10.1016/j.fct.2010.09.030
    1. Song T. Y., Yang N. C., Chen C. L., Thi T. L. V. (20172017). Protective Effects and Possible Mechanisms of Ergothioneine and Hispidin against Methylglyoxal-Induced Injuries in Rat Pheochromocytoma Cells. Oxid. Med. Cel Longev. 2017, 4824371. 10.1155/2017/4824371
    1. Sotgia S., Zinellu A., Mangoni A. A., Pintus G., Attia J., Carru C., et al. (2014). Clinical and Biochemical Correlates of Serum L-Ergothioneine Concentrations in Community-Dwelling Middle-Aged and Older Adults. Plos. One. 9, e84918. 10.1371/journal.pone.0084918
    1. Spencer K. T., Lindower P. D., Buettner G. R., Kerber R. E. (1998). Transition Metal Chelators Reduce Directly Measured Myocardial Free Radical Production during Reperfusion. J. Cardiovasc. Pharmacol. 32, 343–348. 10.1097/00005344-199809000-00002
    1. Spicer S. S., Wooley J. G., Kessler V. (1951). Ergothioneine Depletion in Rabbit Erythrocytes and its Effect on Methemoglobin Formation and Reversion. Proc. Soc. Exp. Biol. Med. 77, 418–420. 10.3181/00379727-77-18799
    1. Stoffels C., Oumari M., Perrou A., Termath A., Schlundt W., Schmalz H. G., et al. (2017). Ergothioneine Stands Out from Hercynine in the Reaction with Singlet Oxygen: Resistance to Glutathione and TRIS in the Generation of Specific Products Indicates High Reactivity. Free Radic. Biol. Med. 113, 385–394. 10.1016/j.freeradbiomed.2017.10.372
    1. Sugiura T., Kato S., Shimizu T., Wakayama T., Nakamichi N., Kubo Y., et al. (2010). Functional Expression of Carnitine/organic Cation Transporter OCTN1/SLC22A4 in Mouse Small Intestine and Liver. Drug Metab. Dispos. 38, 1665–1672. 10.1124/dmd.110.032763
    1. Sun W. L., Li X. Y., Dou H. Y., Wang X. D., Li J. D., Shen L., et al. (2021). Myricetin Supplementation Decreases Hepatic Lipid Synthesis and Inflammation by Modulating Gut Microbiota. Cell Rep 36, 109641. 10.1016/j.celrep.2021.109641
    1. Tang R. M. Y., Cheah I. K., Yew T. S. K., Halliwell B. (2018). Distribution and Accumulation of Dietary Ergothioneine and its Metabolites in Mouse Tissues. Sci. Rep. 8, 1601. 10.1038/s41598-018-20021-z
    1. Tanret C. (1909). A New Base Taken from rye Ergot, Ergothioneine. Ann. Chim. Phys. 18, 114–124.
    1. Taubert D., Jung N., Goeser T., Schömig E. (2009). Increased Ergothioneine Tissue Concentrations in Carriers of the Crohn's Disease Risk-Associated 503F Variant of the Organic Cation Transporter OCTN1. Gut 58, 312–314. 10.1136/gut.2008.164418
    1. Taubert D., Lazar A., Grimberg G., Jung N., Rubbert A., Delank K. S., et al. (2006). Association of Rheumatoid Arthritis with Ergothioneine Levels in Red Blood Cells: a Case Control Study. J. Rheumatol. 33, 2139–2145.
    1. Troy L., McFarland K., Littman-Power S., Kelly B. J., Walpole E. T., Wyld D., et al. (2000). Cisplatin-based Therapy: a Neurological and Neuropsychological Review. Psychooncology 9, 29–39. 10.1002/(sici)1099-1611(200001/02)9:1<29:aid-pon428>;2-z
    1. Tschirka J., Kreisor M., Betz J., Gründemann D. (2018). Substrate Selectivity Check of the Ergothioneine Transporter. Drug Metab. Dispos. 46, 779–785. 10.1124/dmd.118.080440
    1. Tsiantas K., Tsiaka T., Koutrotsios G., Siapi E., Zervakis G. I., Kalogeropoulos N., et al. (2021). On the Identification and Quantification of Ergothioneine and Lovastatin in Various Mushroom Species: Assets and Challenges of Different Analytical Approaches. Molecules 26, 1832. 10.3390/molecules26071832
    1. Turck D., Turck D., Bresson J. L., Burlingame B., Dean T., Fairweather-Tait S., et al. (2017). Statement on the Safety of Synthetic L-Ergothioneine as a Novel Food - Supplementary Dietary Exposure and Safety Assessment for Infants and Young Children, Pregnant and Breastfeeding Women. EFSA J. 15, e05060. 10.2903/j.efsa.2017.5060
    1. Turck D., Turck D., Bresson J. L., Burlingame B., Dean T., Fairweather‐Tait S., et al. (2016). Safety of Synthetic L‐ergothioneine (Ergoneine) as a Novel Food Pursuant to Regulation (EC) No 258/97. Efs2 14, e04629. 10.2903/j.efsa.2016.4629
    1. Weigand-Heller A. J., Kris-Etherton P. M., Beelman R. B. (2012). The Bioavailability of Ergothioneine from Mushrooms (Agaricus Bisporus) and the Acute Effects on Antioxidant Capacity and Biomarkers of Inflammation. Prev. Med. 54, S75–S78. 10.1016/j.ypmed.2011.12.028
    1. Williamson R. D., McCarthy F. P., Manna S., Groarke E., Kell D. B., Kenny L. C., et al. (2020). L-(+)-Ergothioneine Significantly Improves the Clinical Characteristics of Preeclampsia in the Reduced Uterine Perfusion Pressure Rat Model. Hypertension 75, 561–568. 10.1161/HYPERTENSIONAHA.119.13929
    1. Wong K. H., Ng C. C., Kanagasabapathy G., Yow Y. Y., Sabaratnam V. (2017). An Overview of Culinary and Medicinal Mushrooms in Neurodegeneration and Neurotrauma Research. Int. J. Med. Mushrooms. 19, 191–202. 10.1615/IntJMedMushrooms.v19.i3.10
    1. Xiao L., Zhao L., Li T., Hartle D. K., Aruoma O. I., Taylor E. W. (2006). Activity of the Dietary Antioxidant Ergothioneine in a Virus Gene-Based Assay for Inhibitors of HIV Transcription. Biofactors 27, 157–165. 10.1002/biof.5520270114
    1. Yang N. C., Lin H. C., Wu J. H., Ou H. C., Chai Y. C., Tseng C. Y., et al. (2012). Ergothioneine Protects against Neuronal Injury Induced by β-amyloid in Mice. Food Chem. Toxicol. 50, 3902–3911. 10.1016/j.fct.2012.08.021
    1. Zhou T., Prather E. R., Garrison D. E., Zuo L. (2018). Interplay between ROS and Antioxidants during Ischemia-Reperfusion Injuries in Cardiac and Skeletal Muscle. Int. J. Mol. Sci. 19, 417. 10.3390/ijms19020417
    1. Zhu B. Z., Mao L., Fan R. M., Zhu J. G., Zhang Y. N., Wang J., 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, 30–34. 10.1021/tx100214t

Source: PubMed

3
Abonnieren