1
|
Sato M, Torigoe D, Kinoshita Y, Cyuman M, Toda C, Sato M, Ikeda K, Kadomatsu T, Horiguchi H, Morinaga J, Fukami H, Sugizaki T, Miyata K, Kusaba R, Okadome Y, Matsunaga E, Node K, Oike Y. Long-term intake of Tamogi-take mushroom (Pleurotus cornucopiae) mitigates age-related cardiovascular dysfunction and extends healthy life expectancy. NPJ AGING 2025; 11:1. [PMID: 39779757 PMCID: PMC11711650 DOI: 10.1038/s41514-024-00191-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
Age-related declines in cardiac function and exercise tolerance interfere with healthy living and decrease healthy life expectancy in older individuals. Tamogi-take mushrooms (Pleurotus cornucopiae) are known to contain high levels of Ergothioneine (EGT), an antioxidant with potential health benefits. In this study, we assessed the possibility that long-term consumption of Tamogi-take mushrooms might attenuate age-related decline in cardiac and vascular endothelial function in mice. We found that long-term intake of Tamogi-take mushrooms significantly maintained cardiac and vascular endothelial function and improved exercise tolerance in mice. Long-term mushroom consumption also increased levels of Nrf2 (Nuclear factor E2-related factor 2) protein in heart tissues and increased translation of HO-1 (Heme Oxygenase 1) proteins, which have antioxidant effects in heart and aortic tissues. Finally, long-term Tamogi-take mushroom consumption inhibited ROS accumulation with aging and reduced expression of inflammatory biomarkers. We conclude that ingestion of Tamogi-take mushrooms could serve as a dietary intervention to promote cardiovascular health, support healthy aging and slow the progression of age-related diseases.
Collapse
Affiliation(s)
- Michio Sato
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Cardiovascular Medicine, School of Medicine, Saga University, Saga, Japan
- Division of Kumamoto Mouse Clinic (KMC), Kumamoto University, Kumamoto, Japan
| | - Daisuke Torigoe
- Division of Laboratory Animal Science, Institute of Resource Development and Analysis (IRDA), Kumamoto University, Kumamoto, Japan
| | - Yuya Kinoshita
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Momoka Cyuman
- Division of Laboratory Animal Science, Institute of Resource Development and Analysis (IRDA), Kumamoto University, Kumamoto, Japan
| | - Chitoku Toda
- Department of Neuroscience for Metabolic Control, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaru Sato
- Laboratory of Biomolecule Analysis, Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
| | - Kazutaka Ikeda
- Laboratory of Biomolecule Analysis, Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
- Laboratory of Omics and Informatics, Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | | | - Haruki Horiguchi
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Aging and Geriatric Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Jun Morinaga
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
- Department of Disease Genome Epidemiology, Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kumamoto, Japan
| | - Hirotaka Fukami
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Taichi Sugizaki
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Keishi Miyata
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Ryoko Kusaba
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Yusuke Okadome
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Eiji Matsunaga
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Koichi Node
- Department of Cardiovascular Medicine, School of Medicine, Saga University, Saga, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan.
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
- Division of Laboratory Animal Science, Institute of Resource Development and Analysis (IRDA), Kumamoto University, Kumamoto, Japan.
- Laboratory of Biomolecule Analysis, Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan.
| |
Collapse
|
2
|
Zhou Y, Zhang Y, Jin S, Lv J, Li M, Feng N. The gut microbiota derived metabolite trimethylamine N-oxide: Its important role in cancer and other diseases. Biomed Pharmacother 2024; 177:117031. [PMID: 38925016 DOI: 10.1016/j.biopha.2024.117031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024] Open
Abstract
An expanding body of research indicates a correlation between the gut microbiota and various diseases. Metabolites produced by the gut microbiota act as mediators between the gut microbiota and the host, interacting with multiple systems in the human body to regulate physiological or pathological functions. However, further investigation is still required to elucidate the underlying mechanisms. One such metabolite involved in choline metabolism by gut microbes is trimethylamine (TMA), which can traverse the intestinal epithelial barrier and enter the bloodstream, ultimately reaching the liver where it undergoes oxidation catalyzed by flavin-containing monooxygenase 3 (FMO3) to form trimethylamine N-oxide (TMAO). While some TMAO is eliminated through renal excretion, remaining amounts circulate in the bloodstream, leading to systemic inflammation, endoplasmic reticulum (ER) stress, mitochondrial stress, and disruption of normal physiological functions in humans. As a representative microbial metabolite originating from the gut, TMAO has significant potential both as a biomarker for monitoring disease occurrence and progression and for tailoring personalized treatment strategies for patients. This review provides an extensive overview of TMAO sources and its metabolism in human blood, as well as its impact on several major human diseases. Additionally, we explore the latest research areas related to TMAO along with future directions.
Collapse
Affiliation(s)
- Yuhua Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yuwei Zhang
- Nantong University Medical School, Nantong, China
| | - Shengkai Jin
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jing Lv
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Menglu Li
- Department of Urology, Jiangnan University Medical Center, Wuxi, China.
| | - Ninghan Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China; Nantong University Medical School, Nantong, China; Department of Urology, Jiangnan University Medical Center, Wuxi, China.
| |
Collapse
|
3
|
Chen M, Yi Y, Chen B, Zhang H, Dong M, Yuan L, Zhou H, Jiang H, Ma Z. Metformin inhibits OCTN1- and OCTN2-mediated hepatic accumulation of doxorubicin and alleviates its hepatotoxicity in mice. Toxicology 2024; 503:153757. [PMID: 38364893 DOI: 10.1016/j.tox.2024.153757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/18/2024]
Abstract
Doxorubicin (DOX) is a widely used antitumor agent; however, its clinical application is limited by dose-related organ damage. Because organic cation/carnitine transporters (OCTN1 and OCTN2), which are critical for DOX uptake, are highly expressed in hepatocytes, we aimed to elucidate the role of these transporters in hepatic DOX uptake. The results indicated that inhibitors and RNA interference both significantly reduced DOX accumulation in HepG2 and HepaRG cells, suggesting that OCTN1/2 contribute substantially to DOX uptake by hepatocytes. To determine whether metformin (MET, an inhibitor of OCTN1 and OCTN2) ameliorates DOX-induced hepatotoxicity, we conducted in vitro and in vivo studies. MET (1-100 μM) inhibited DOX (500 nM) accumulation and cytotoxicity in vitro in a concentration-dependent manner. Furthermore, intravenous MET administration at 250 or 500 mg/kg or by gavage at 50, 100, or 200 mg/kg reduced DOX (8 mg/kg) accumulation in a dose-dependent manner in the mouse liver and attenuated the release of alanine aminotransferase, aspartate aminotransferase, and carboxylesterase 1. Additionally, MET reduced the distribution of DOX in the heart, liver, and kidney and enhanced the urinary elimination of DOX; however, it did not increase the nephric toxicity of DOX. In conclusion, our study demonstrated that MET alleviates DOX hepatotoxicity by inhibiting OCTN1- and OCTN2-mediated DOX uptake in vitro (mouse hepatocytes and HepaRG or HepG2 cells) and in mice.
Collapse
Affiliation(s)
- Mingyang Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yaodong Yi
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Binxin Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Hengbin Zhang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Minlei Dong
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Luexiang Yuan
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hui Zhou
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Jinhua Institute of Zhejiang University, Jinhua, China
| | - Huidi Jiang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Jinhua Institute of Zhejiang University, Jinhua, China.
| | - Zhiyuan Ma
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| |
Collapse
|
4
|
Shu G, Lei X, Li G, Zhang T, Wang C, Song A, Yu H, Wang X, Deng X. Ergothioneine suppresses hepatic stellate cell activation via promoting Foxa3-dependent potentiation of the Hint1/Smad7 cascade and improves CCl 4-induced liver fibrosis in mice. Food Funct 2023; 14:10591-10604. [PMID: 37955610 DOI: 10.1039/d3fo03643j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Ergothioneine (EGT) is a bioactive compound derived from certain edible mushrooms. The activation of hepatic stellate cells (HSCs) is critically involved in the etiology of liver fibrosis (LF). Here, we report that in LX-2 HSCs, EGT upregulates the expression of Hint1 and Smad7 and suppresses their activation provoked by TGFβ1. The EGT-triggered inhibition of HSC activation is abolished by knocking down the expression of Hint1. Overexpression of Hint1 increases Smad7 and represses TGFβ1-provoked activation of LX-2 HSCs. In silico predictions unveiled that in the promoter region of the human Hint1 gene, there are two conserved cis-acting elements that have the potential to interact with the transcription factor Foxa3 termed hFBS1 and hFBS2, respectively. The knockdown of Foxa3 obviously declined Hint1 expression at both mRNA and protein levels. Transfection of Foxa3 or EGT treatment increased the activity of the luciferase reporter driven by the Hint1 promoter in an hFBS2-dependent manner. The knockdown of Foxa3 eliminated EGT-mediated upregulation of Hint1 promoter activity. Additionally, EGT triggered the nuclear translocation of Foxa3 without obviously affecting its expression level. Molecular docking analysis showed that EGT has the potential to directly interact with the Foxa3 protein. Moreover, Foxa3 played a critical role in EGT-mediated hepatoprotection. EGT modulated the Foxa3/Hint1/Smad7 signaling in mouse primary HSCs and inhibited their activation. The gavage of EGT considerably relieved CCl4-induced LF in mice. Our data provide new insights into the anti-LF activity of EGT. Mechanistically, EGT triggers the nuclear translocation of Foxa3 in HSCs, which promotes Hint1 transcription and subsequently elevates Smad7.
Collapse
Affiliation(s)
- Guangwen Shu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| | - Xiao Lei
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| | - Guangqiong Li
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| | - Tiantian Zhang
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| | - Chuo Wang
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| | - Anning Song
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| | - Huifan Yu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiaoming Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Xukun Deng
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Hubei, China.
| |
Collapse
|
5
|
Liu Y, Wang C, Liu R, Zhao M, Ding X, Zhang T, He R, Zhu S, Dong X, Xie J, Gu Z, Zhao Y. Adhesive Ergothioneine Hyaluronate Gel Protects against Radiation Gastroenteritis by Alleviating Apoptosis, Inflammation, and Gut Microbiota Dysbiosis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:19833-19846. [PMID: 37052616 DOI: 10.1021/acsami.2c23142] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Radiation gastroenteritis represents one of the most prevalent and hazardous complications of abdominopelvic radiotherapy, which not only severely reduces patients' life quality but also restricts radiotherapy efficacy. However, there is currently no clinically available oral radioprotector for this threatening disease due to its complex pathogenesis and the harsh gastrointestinal environment. To this end, this study developed a facile but effective oral radioprotector, ergothioneine hyaluronate (EGT@HA) gel, protecting against radiation gastroenteritis by synergistically regulating oxidative stress, inflammation, and gut microbiota. In vitro and cellular experiments verified the chemical stability and free radical scavenging ability of EGT and its favorable cellular radioprotective efficacy by inhibiting intracellular reactive oxidative species (ROS) generation, DNA damage, mitochondrial damage, and apoptosis. At the in vivo level, EGT@HA with prolonged gastrointestinal residence mitigated radiation-induced gastrointestinal tissue injury, apoptosis, neutrophil infiltration, and gut flora dysbiosis. For the first time, this work investigated the protective effects of EGT@HA gel on radiation gastroenteritis, which not only hastens the advancement of the novel gastrointestinal radioprotector but also provides a valuable gastrointestinal radioprotection paradigm by synergistically modulating oxidative stress, inflammation, and gut microbiota disturbance.
Collapse
Affiliation(s)
- Yaping Liu
- The First Affiliated Hospital of University of Science and Technology of China, Hefei 230001, Anhui, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Chengyan Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Ruixue Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Maoru Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Xuefeng Ding
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Tingjun Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Rendong He
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Xinghua Dong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Jiani Xie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
- China School of Food and Biological Engineering, Chengdu University, Chengdu 610106, Sichuan, China
| | - Zhanjun Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
6
|
Safe and Effective Antioxidant: The Biological Mechanism and Potential Pathways of Ergothioneine in the Skin. Molecules 2023; 28:molecules28041648. [PMID: 36838636 PMCID: PMC9967237 DOI: 10.3390/molecules28041648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/02/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023] Open
Abstract
Ergothioneine, a sulfur-containing micromolecular histidine derivative, has attracted increasing attention from scholars since it was confirmed in the human body. In the human body, ergothioneine is transported and accumulated specifically through OCTN-1, especially in the mitochondria and nucleus, suggesting that it can target damaged cells and tissues as an antioxidant. It shows excellent antioxidant, anti-inflammatory effects, and anti-aging properties, and inhibits melanin production. It is a mega antioxidant that may participate in the antioxidant network system and promote the reducing glutathione regeneration cycle. This review summarizes studies on the antioxidant effects of ergothioneine on various free radicals in vitro to date and systematically introduces its biological activities and potential mechanisms, mostly in dermatology. Additionally, the application of ergothioneine in cosmetics is briefly summarized. Lastly, we propose some problems that require solutions to understand the mechanism of action of ergothioneine. We believe that ergothioneine has good prospects in the food and cosmetics industries, and can thus meet some needs of the health and beauty industry.
Collapse
|
7
|
Abstract
Liver fibrosis has a high incidence worldwide and is the common pathological basis of many chronic liver diseases. Liver fibrosis is caused by the excessive deposition of extracellular matrix and concomitant collagen accumulation in livers and can lead to the development of liver cirrhosis and even liver cancer. A large number of studies have provided evidence that liver fibrosis can be blocked or even reversed by appropriate medical interventions. However, the antifibrosis drugs with ideal clinical efficacy are still insufficient. The edible plant-derived natural compounds have been reported to exert effective antifibrotic effects with few side-effects, representing a kind of promising source for the treatment of liver fibrosis. In this article, we reviewed the current progress of the natural compounds derived from dietary plants in the treatment of liver fibrosis, including phenolic compounds (capsaicin, chlorogenic acid, curcumin, ellagic acid, epigallocatechin-3-gallate, resveratrol, sinapic acid, syringic acid, vanillic acid and vitamin E), flavonoid compounds (genistein, hesperidin, hesperetin, naringenin, naringin and quercetin), sulfur-containing compounds (S-allylcysteine, ergothioneine, lipoic acid and sulforaphane) and other compounds (betaine, caffeine, cucurbitacin B, lycopene, α-mangostin, γ-mangostin, ursolic acid, vitamin C and yangonin). The pharmacological effects and related mechanisms of these compounds in in-vivo and in-vitro models of liver fibrosis are focused.
Collapse
|
8
|
Abstract
Significance: Ergothioneine (ET) is an unusual sulfur-containing amino acid derived from histidine, acquired predominantly from food. Its depletion is associated with deleterious consequences in response to stress stimuli in cell culture models, prompting us to classify it as a vitamin in 2010, which was later supported by in vivo studies. ET is obtained from a variety of foods and is taken up by a selective transporter. ET possesses antioxidant and anti-inflammatory properties that confer cytoprotection. ET crosses the blood-brain barrier and has been reported to have beneficial effects in the brain. In this study, we discuss the cytoprotective and neuroprotective properties of ET, which may be harnessed for combating neurodegeneration and decline during aging. Recent Advances: The designation of ET as a stress vitamin is gaining momentum, opening a new field of investigation involving small molecules that are essential for optimal physiological functioning and maintenance of health span. Critical Issues: Although ET was discovered more than a century ago, its physiological functions are still being elucidated, especially in the brain. As ET is present in most foods, toxicity associated with its deprivation has been difficult to assess. Future Directions: Using genetically engineered cells and mice, it may now be possible to elucidate roles of ET. This coupled with advances in genomics and metabolomics may lead to identification of ET function. As ET is a stable antioxidant with anti-inflammatory properties, whose levels decline during aging, supplementing ET in the diet or consuming an ET-rich diet may prove beneficial. Antioxid. Redox Signal. 36, 1306-1317.
Collapse
Affiliation(s)
- Bindu D Paul
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of The Solomon H. Snyder Department of Neuroscience, and Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Iciek M, Bilska-Wilkosz A, Kozdrowicki M, Górny M. Reactive Sulfur Compounds in the Fight against COVID-19. Antioxidants (Basel) 2022; 11:antiox11061053. [PMID: 35739949 PMCID: PMC9220020 DOI: 10.3390/antiox11061053] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/20/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
The SARS-CoV-2 coronavirus pandemic outbreak in 2019 resulted in the need to search for an effective and safe strategy for treating infected patients, relieving symptoms, and preventing severe disease. SARS-CoV-2 is an RNA virus that can cause acute respiratory failure and thrombosis, as well as impair circulatory system function. Permanent damage to the heart muscle or other cardiovascular disorders may occur during or after the infection. The severe course of the disease is associated with the release of large amounts of pro-inflammatory cytokines. Due to their documented anti-inflammatory, antioxidant, and antiviral effects, reactive sulfur compounds, including hydrogen sulfide (H2S), lipoic acid (LA), N-acetylcysteine (NAC), glutathione (GSH), and some other lesser-known sulfur compounds, have attracted the interest of scientists for the treatment and prevention of the adverse effects of diseases caused by SARS-CoV-2. This article reviews current knowledge about various endogenous or exogenous reactive sulfur compounds and discusses the possibility, or in some cases the results, of their use in the treatment or prophylaxis of COVID-19.
Collapse
|
10
|
Gründemann D, Hartmann L, Flögel S. The Ergothioneine Transporter (ETT): Substrates and Locations, an Inventory. FEBS Lett 2021; 596:1252-1269. [PMID: 34958679 DOI: 10.1002/1873-3468.14269] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 11/07/2022]
Abstract
In all vertebrates including mammals, the ergothioneine transporter ETT (obsolete name OCTN1; human gene symbol SLC22A4) is a powerful and highly specific transporter for the uptake of ergothioneine (ET). ETT is not expressed ubiquitously and only cells with high ETT cell-surface levels can accumulate ET to high concentration. Without ETT, there is no uptake because the plasma membrane is essentially impermeable to this hydrophilic zwitterion. Here, we review the substrate specificity and localization of ETT, which is prominently expressed in neutrophils, monocytes/macrophages, and developing erythrocytes. Most sites of strong expression are conserved across species, but there are also major differences. In particular, we critically analyze the evidence for the expression of ETT in the brain as well as recent data suggesting that the transporter SLC22A15 may transport also ET. We conclude that, to date, ETT remains the only well-defined biomarker for intracellular ET activity. In humans, the ability to take up, distribute, and retain ET depends principally on this transporter.
Collapse
Affiliation(s)
- Dirk Gründemann
- Department of Pharmacology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Gleueler Straße 24, 50931, Cologne, Germany
| | - Lea Hartmann
- Department of Pharmacology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Gleueler Straße 24, 50931, Cologne, Germany
| | - Svenja Flögel
- Department of Pharmacology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Gleueler Straße 24, 50931, Cologne, Germany
| |
Collapse
|
11
|
Yadan JC. Matching chemical properties to molecular biological activities opens a new perspective on L-ergothioneine. FEBS Lett 2021; 596:1299-1312. [PMID: 34928499 DOI: 10.1002/1873-3468.14264] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 11/10/2022]
Abstract
L-ergothioneine is a low-molecular weight natural product, the chemical structure of which comprises oxygen-, nitrogen- and sulfur-containing functional groups. This gives L-ergothioneine specific physicochemical properties and allows to better understanding its chemical reactivity, which is primarily due to the 2-thio-imidazole group. Here, I review how different modes of chemical reactivity account for the reported molecular biological activities of L-ergothioneine. By matching the physicochemical properties to the biological properties of L-ergothioneine, a new perspective of the function and the mode of action of this enigmatic molecule emerges into the limelight.
Collapse
|
12
|
Dare A, Channa ML, Nadar A. L-ergothioneine and metformin alleviates liver injury in experimental type-2 diabetic rats via reduction of oxidative stress, inflammation, and hypertriglyceridemia. Can J Physiol Pharmacol 2021; 99:1137-1147. [PMID: 34582252 DOI: 10.1139/cjpp-2021-0247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Type-2 diabetes (T2D) is associated with liver toxicity. L-ergothioneine (L-egt) has been reported to reduce toxicity in tissues exposed to injury, while metformin is commonly prescribed to manage T2D. Hence, this study evaluates the hepatoprotective role of L-egt, with or without metformin, in T2D male rats. A total of 36 adult male Sprague-Dawley rats were randomly divided into non-diabetic (n = 12) and diabetic (n = 24) groups. After induction of diabetes, animals were divided into six groups (n = 6) and treated orally either with deionized water, L-egt (35 mg/kg bodyweight (bwt)), metformin (500 mg/kg bwt), or a combination of L-egt and metformin for 7 weeks. Body weight and blood glucose were monitored during the experiment. Thereafter, animals were euthanized and liver tissue was excised for biochemical, ELISA, real-time quantitative PCR, and histopathological analysis. L-egt with or without metformin reduced liver hypertrophy, liver injury, triglycerides, oxidative stress, and inflammation. Also, L-egt normalized mRNA expression of SREBP-1c, fatty acid synthase, nuclear factor kappa B, transforming growth factor β1, nuclear factor erythroid 2-related factor 2, and sirtuin-1 in diabetic rats. Furthermore, co-administration of L-egt with metformin to diabetic rats reduced blood glucose and insulin resistance. These results provide support to the therapeutic benefits of L-egt in the management of liver complications associated with T2D.
Collapse
Affiliation(s)
- Ayobami Dare
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban X54001, South Africa
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban X54001, South Africa
| | - Mahendra L Channa
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban X54001, South Africa
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban X54001, South Africa
| | - Anand Nadar
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban X54001, South Africa
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban X54001, South Africa
| |
Collapse
|
13
|
Tian X, Cioccoloni G, Sier JH, Naseem KM, Thorne JL, Moore JB. Ergothioneine supplementation in people with metabolic syndrome (ErgMS): protocol for a randomised, double-blind, placebo-controlled pilot study. Pilot Feasibility Stud 2021; 7:193. [PMID: 34715934 PMCID: PMC8555363 DOI: 10.1186/s40814-021-00929-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 10/12/2021] [Indexed: 02/06/2023] Open
Abstract
Background Ergothioneine is a naturally occurring metabolite of histidine found in many foods and in high amounts in mushrooms. In vivo, ergothioneine acts as an antioxidant and is widely distributed in most mammalian tissues. While ergothioneine is sold as a dietary supplement for its antioxidant and anti-inflammatory properties, to date there are no published intervention trials examining its health benefits in humans. The aim of this work was to develop a study protocol for a pilot interventional trial that will establish the primary and secondary outcomes, and the power required, for a definitive randomised controlled trial to test the hypothesis that ergothioneine supplementation is beneficial for people with metabolic syndrome. Methods We have designed the ErgMS study as a single-centre, randomised, double-blind, placebo-controlled, 3-arm parallel, pilot intervention trial, which aims to supplement participants with either placebo, 5 or 30 mg/day ergothioneine for 12 weeks. Measurements of metabolic syndrome risk factors, serum markers of oxidative stress (lipid peroxidation), inflammation, blood platelet function and liver function will take place at baseline, and after 6 weeks and 12 weeks of supplementation. In addition, we will examine if there are any changes in the serum metabolome in response to ergothioneine supplementation. Linear regression and two-way ANOVA will be utilised to analyse the association between ergothioneine and measured variables. Discussion The ErgMS study will be the first study to address the question does ergothioneine supplementation have health benefits for people with metabolic syndrome. Study results will provide preliminary data as to which dose may improve inflammatory markers in adults with metabolic syndrome and will inform dose and primary outcome selection for a definitive randomised controlled trial. Trial registration ISRCTN, ISRCTN25890011 Registered February 10th, 2021 Supplementary Information The online version contains supplementary material available at 10.1186/s40814-021-00929-6.
Collapse
Affiliation(s)
- Xiaoying Tian
- School of Food Science & Nutrition, University of Leeds, Leeds, LS2 9JT, UK
| | - Giorgia Cioccoloni
- School of Food Science & Nutrition, University of Leeds, Leeds, LS2 9JT, UK
| | - Joanna H Sier
- School of Food Science & Nutrition, University of Leeds, Leeds, LS2 9JT, UK
| | - Khalid M Naseem
- Leeds Institute of Cardiovascular & Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - James L Thorne
- School of Food Science & Nutrition, University of Leeds, Leeds, LS2 9JT, UK
| | - J Bernadette Moore
- School of Food Science & Nutrition, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
14
|
Lam-Sidun D, Peters KM, Borradaile NM. Mushroom-Derived Medicine? Preclinical Studies Suggest Potential Benefits of Ergothioneine for Cardiometabolic Health. Int J Mol Sci 2021; 22:ijms22063246. [PMID: 33806754 PMCID: PMC8004618 DOI: 10.3390/ijms22063246] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/18/2022] Open
Abstract
Medicinal use of mushrooms has been documented since ancient times, and in the modern world, mushrooms have a longstanding history of use in Eastern medicine. Recent interest in plant-based diets in Westernized countries has brought increasing attention to the use of mushrooms and mushroom-derived compounds in the prevention and treatment of chronic diseases. Edible mushrooms are the most abundant food sources of the modified amino acid, ergothioneine. This compound has been shown to accumulate in almost all cells and tissues, but preferentially in those exposed to oxidative stress and injury. The demonstrated cytoprotectant effect of ergothioneine has led many to suggest a potential therapeutic role for this compound in chronic conditions that involve ongoing oxidative stress and inflammation, including cardiovascular and metabolic diseases. However, the in vivo effects of ergothioneine and its underlying therapeutic mechanisms in the whole organism are not as clear. Moreover, there are no well-defined, clinical prevention and intervention trials of ergothioneine in chronic disease. This review highlights the cellular and molecular mechanisms of action of ergothioneine and its potential as a Traditional, Complementary and Alternative Medicine for the promotion of cardiometabolic health and the management of the most common manifestations of cardiometabolic disease.
Collapse
|
15
|
Cheah IK, Halliwell B. Ergothioneine, recent developments. Redox Biol 2021; 42:101868. [PMID: 33558182 PMCID: PMC8113028 DOI: 10.1016/j.redox.2021.101868] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/14/2021] [Accepted: 01/14/2021] [Indexed: 02/06/2023] Open
Abstract
There has been a recent surge of interest in the unique low molecular weight dietary thiol/thione, ergothioneine. This compound can accumulate at high levels in the body from diet and may play important physiological roles in human health and development, and possibly in prevention and treatment of disease. Blood levels of ergothioneine decline with age and onset of various diseases. Here we highlight recent advances in our knowledge of ergothioneine.
Collapse
Affiliation(s)
- Irwin K Cheah
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117596, Singapore; Life Science Institute, Neurobiology Programme, Centre for Life Sciences, National University of Singapore, 117456, Singapore
| | - Barry Halliwell
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117596, Singapore; Life Science Institute, Neurobiology Programme, Centre for Life Sciences, National University of Singapore, 117456, Singapore.
| |
Collapse
|
16
|
Organic Cation Transporters in Human Physiology, Pharmacology, and Toxicology. Int J Mol Sci 2020; 21:ijms21217890. [PMID: 33114309 PMCID: PMC7660683 DOI: 10.3390/ijms21217890] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/13/2022] Open
Abstract
Individual cells and epithelia control the chemical exchange with the surrounding environment by the fine-tuned expression, localization, and function of an array of transmembrane proteins that dictate the selective permeability of the lipid bilayer to small molecules, as actual gatekeepers to the interface with the extracellular space. Among the variety of channels, transporters, and pumps that localize to cell membrane, organic cation transporters (OCTs) are considered to be extremely relevant in the transport across the plasma membrane of the majority of the endogenous substances and drugs that are positively charged near or at physiological pH. In humans, the following six organic cation transporters have been characterized in regards to their respective substrates, all belonging to the solute carrier 22 (SLC22) family: the organic cation transporters 1, 2, and 3 (OCT1–3); the organic cation/carnitine transporter novel 1 and 2 (OCTN1 and N2); and the organic cation transporter 6 (OCT6). OCTs are highly expressed on the plasma membrane of polarized epithelia, thus, playing a key role in intestinal absorption and renal reabsorption of nutrients (e.g., choline and carnitine), in the elimination of waste products (e.g., trimethylamine and trimethylamine N-oxide), and in the kinetic profile and therapeutic index of several drugs (e.g., metformin and platinum derivatives). As part of the Special Issue Physiology, Biochemistry, and Pharmacology of Transporters for Organic Cations, this article critically presents the physio-pathological, pharmacological, and toxicological roles of OCTs in the tissues in which they are primarily expressed.
Collapse
|
17
|
Cheah IK, Halliwell B. Could Ergothioneine Aid in the Treatment of Coronavirus Patients? Antioxidants (Basel) 2020; 9:E595. [PMID: 32646061 PMCID: PMC7402156 DOI: 10.3390/antiox9070595] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/25/2020] [Accepted: 07/02/2020] [Indexed: 01/08/2023] Open
Abstract
Infection with SARS-CoV-2 causes the coronavirus infectious disease 2019 (COVID-19), a pandemic that has, at present, infected more than 11 million people globally. Some COVID-19 patients develop a severe and critical illness, spurred on by excessive inflammation that can lead to respiratory or multiorgan failure. Numerous studies have established the unique array of cytoprotective properties of the dietary amino acid ergothioneine. Based on studies in a range of in vitro and in vivo models, ergothioneine has exhibited the ability to modulate inflammation, scavenge free radicals, protect against acute respiratory distress syndrome, prevent endothelial dysfunction, protect against ischemia and reperfusion injury, protect against neuronal damage, counteract iron dysregulation, hinder lung and liver fibrosis, and mitigate damage to the lungs, kidneys, liver, gastrointestinal tract, and testis, amongst many others. When compiled, this evidence suggests that ergothioneine has a potential application in the treatment of the underlying pathology of COVID-19. We propose that ergothioneine could be used as a therapeutic to reduce the severity and mortality of COVID-19, especially in the elderly and those with underlying health conditions. This review presents evidence to support that proposal.
Collapse
Affiliation(s)
- Irwin K. Cheah
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore;
- Life Science Institute, Neurobiology Programme, Centre for Life Sciences, National University of Singapore, Singapore 117456, Singapore
| | - Barry Halliwell
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore;
- Life Science Institute, Neurobiology Programme, Centre for Life Sciences, National University of Singapore, Singapore 117456, Singapore
| |
Collapse
|
18
|
Natural Sulfur-Containing Compounds: An Alternative Therapeutic Strategy against Liver Fibrosis. Cells 2019; 8:cells8111356. [PMID: 31671675 PMCID: PMC6929087 DOI: 10.3390/cells8111356] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/25/2019] [Accepted: 10/26/2019] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis is a pathophysiologic process involving the accumulation of extracellular matrix proteins as collagen deposition. Advanced liver fibrosis can evolve in cirrhosis, portal hypertension and often requires liver transplantation. At the cellular level, hepatic fibrosis involves the activation of hepatic stellate cells and their transdifferentiation into myofibroblasts. Numerous pro-fibrogenic mediators including the transforming growth factor-β1, the platelet-derived growth factor, endothelin-1, toll-like receptor 4, and reactive oxygen species are key players in this process. Knowledge of the cellular and molecular mechanisms underlying hepatic fibrosis development need to be extended to find novel therapeutic strategies. Antifibrotic therapies aim to inhibit the accumulation of fibrogenic cells and/or prevent the deposition of extracellular matrix proteins. Natural products from terrestrial and marine sources, including sulfur-containing compounds, exhibit promising activities for the treatment of fibrotic pathology. Although many therapeutic interventions are effective in experimental models of liver fibrosis, their efficacy and safety in humans are largely unknown. This review aims to provide a reference collection on experimentally tested natural anti-fibrotic compounds, with particular attention on sulfur-containing molecules. Their chemical structure, sources, mode of action, molecular targets, and pharmacological activity in the treatment of liver disease will be discussed.
Collapse
|
19
|
van der Hoek SA, Darbani B, Zugaj KE, Prabhala BK, Biron MB, Randelovic M, Medina JB, Kell DB, Borodina I. Engineering the Yeast Saccharomyces cerevisiae for the Production of L-(+)-Ergothioneine. Front Bioeng Biotechnol 2019; 7:262. [PMID: 31681742 PMCID: PMC6797849 DOI: 10.3389/fbioe.2019.00262] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/26/2019] [Indexed: 01/08/2023] Open
Abstract
L-(+)-Ergothioneine (ERG) is an unusual, naturally occurring antioxidant nutraceutical that has been shown to help reduce cellular oxidative damage. Humans do not biosynthesise ERG, but acquire it from their diet; it exploits a specific transporter (SLC22A4) for its uptake. ERG is considered to be a nutraceutical and possible vitamin that is involved in the maintenance of health, and seems to be at too low a concentration in several diseases in vivo. Ergothioneine is thus a potentially useful dietary supplement. Present methods of commercial production rely on extraction from natural sources or on chemical synthesis. Here we describe the engineering of the baker's yeast Saccharomyces cerevisiae to produce ergothioneine by fermentation in defined media. After integrating combinations of ERG biosynthetic pathways from different organisms, we screened yeast strains for their production of ERG. The highest-producing strain was also engineered with known ergothioneine transporters. The effect of amino acid supplementation of the medium was investigated and the nitrogen metabolism of S. cerevisiae was altered by knock-out of TOR1 or YIH1. We also optimized the media composition using fractional factorial methods. Our optimal strategy led to a titer of 598 ± 18 mg/L ergothioneine in fed-batch culture in 1 L bioreactors. Because S. cerevisiae is a GRAS ("generally recognized as safe") organism that is widely used for nutraceutical production, this work provides a promising process for the biosynthetic production of ERG.
Collapse
Affiliation(s)
- Steven A. van der Hoek
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Behrooz Darbani
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Karolina E. Zugaj
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Bala Krishna Prabhala
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mathias Bernfried Biron
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Milica Randelovic
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Jacqueline B. Medina
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Douglas B. Kell
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Irina Borodina
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
20
|
Maurer A, Leisinger F, Lim D, Seebeck FP. Structure and Mechanism of Ergothionase fromTreponema denticola. Chemistry 2019; 25:10298-10303. [DOI: 10.1002/chem.201901866] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/21/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Alice Maurer
- Department for ChemistryUniversity of Basel Mattenstrasse 24a Basel 4002 Switzerland
| | - Florian Leisinger
- Department for ChemistryUniversity of Basel Mattenstrasse 24a Basel 4002 Switzerland
| | - David Lim
- Department for ChemistryUniversity of Basel Mattenstrasse 24a Basel 4002 Switzerland
| | - Florian P. Seebeck
- Department for ChemistryUniversity of Basel Mattenstrasse 24a Basel 4002 Switzerland
| |
Collapse
|
21
|
Antifibrotic Effect of Marine Ovothiol in an In Vivo Model of Liver Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:5045734. [PMID: 30647809 PMCID: PMC6311726 DOI: 10.1155/2018/5045734] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 09/18/2018] [Indexed: 12/13/2022]
Abstract
Liver fibrosis is a complex process caused by chronic hepatic injury, which leads to an excessive increase in extracellular matrix protein accumulation and fibrogenesis. Several natural products, including sulfur-containing compounds, have been investigated for their antifibrotic effects; however, the molecular mechanisms underpinning their action are partially still obscure. In this study, we have investigated for the first time the effect of ovothiol A, π-methyl-5-thiohistidine, isolated from sea urchin eggs on an in vivo murine model of liver fibrosis. Mice were intraperitoneally injected with carbon tetrachloride (CCl4) to induce liver fibrosis and treated with ovothiol A at the dose of 50 mg/kg 3 times a week for 2 months. Treatment with ovothiol A caused a significant reduction of collagen fibers as observed by histopathological changes and serum parameters compared to mice treated with control solution. This antifibrotic effect was associated to the decrease of fibrogenic markers involved in liver fibrosis progression, such as the transforming growth factor (TGF-β), the α-smooth muscle actin (α-SMA), and the tissue metalloproteinases inhibitor (TIMP-1). Finally, we provided evidence that the attenuation of liver fibrosis by ovothiol A treatment can be regulated by the expression and activity of the membrane-bound γ-glutamyl-transpeptidase (GGT), which is a key player in maintaining intracellular redox homoeostasis. Overall, these findings indicate that ovothiol A has significant antifibrotic properties and can be considered as a new marine drug or dietary supplement in potential therapeutic strategies for the treatment of liver fibrosis.
Collapse
|
22
|
Combination Metabolomics Approach for Identifying Endogenous Substrates of Carnitine/Organic Cation Transporter OCTN1. Pharm Res 2018; 35:224. [PMID: 30280275 DOI: 10.1007/s11095-018-2507-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 09/18/2018] [Indexed: 01/02/2023]
Abstract
PURPOSE Solute carrier SLC22A4 encodes the carnitine/organic cation transporter OCTN1 and is associated with inflammatory bowel disease, although little is known about how this gene is linked to pathogenesis. The aim of the present study was to identify endogenous substrates that are associated with gastrointestinal inflammation. METHODS HEK293/OCTN1 and mock cells were incubated with colon extracts isolated from dextran sodium sulfate-induced colitis mice; the subsequent cell lysates were mixed with the amino group selective reagent 3-aminopyridyl-N-hydroxysuccinimidyl carbamate (APDS), to selectively label OCTN1 substrates. Precursor ion scanning against the fragment ion of APDS was then used to identify candidate OCTN1 substrates. RESULTS Over 10,000 peaks were detected by precursor ion scanning; m/z 342 had a higher signal in HEK293/OCTN1 compared to mock cells. This peak was detected as a divalent ion that contained four APDS-derived fragments and was identified as spermine. Spermine concentration in peripheral blood mononuclear cells from octn1 gene knockout mice (octn1-/-) was significantly lower than in wild-type mice. Lipopolysaccharide-induced gene expression of inflammatory cytokines in peritoneal macrophages from octn1-/- mice was lower than in wild-type mice. CONCLUSIONS The combination metabolomics approach can provide a novel tool to identify endogenous substrates of OCTN1.
Collapse
|
23
|
Halliwell B, Cheah IK, Tang RMY. Ergothioneine - a diet-derived antioxidant with therapeutic potential. FEBS Lett 2018; 592:3357-3366. [PMID: 29851075 DOI: 10.1002/1873-3468.13123] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 05/22/2018] [Indexed: 12/21/2022]
Abstract
Ergothioneine is a thiol/thione molecule synthesised only by some fungi and bacteria. Nonetheless, it is avidly taken up from the diet by humans and other animals through a transporter, OCTN1, and accumulates to high levels in certain tissues. Ergothioneine is not rapidly metabolised, or excreted in urine and is present in many, if not all, human tissues and body fluids. Ergothioneine has powerful antioxidant and cytoprotective properties in vitro and there is evidence that the body may concentrate it at sites of tissue injury by raising OCTN1 levels. Decreased blood and/or plasma levels of ergothioneine have been observed in some diseases, suggesting that a deficiency could be relevant to the disease onset or progression. This brief Review explores the possible roles of ergothioneine in human health and disease.
Collapse
Affiliation(s)
- Barry Halliwell
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
| | - Irwin K Cheah
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
| | - Richard M Y Tang
- National University of Singapore Graduate School for Integrative Sciences and Engineering, Singapore, Singapore
| |
Collapse
|
24
|
Mede R, Hoffmann P, Klein M, Görls H, Schmitt M, Neugebauer U, Gessner G, Heinemann SH, Popp J, Westerhausen M. A Water-Soluble Mn(CO)3-Based and Non-Toxic PhotoCORM for Administration of Carbon Monoxide Inside of Cells. Z Anorg Allg Chem 2017. [DOI: 10.1002/zaac.201700349] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Ralf Mede
- Institute of Inorganic and Analytical Chemistry; Friedrich Schiller University; Humboldtstraße 8 07743 Jena Germany
| | - Patrick Hoffmann
- Center for Sepsis Control and Care (CSCC); Jena University Hospital; Am Klinikum 1 07747 Jena Germany
- Leibniz Institute of Photonic Technology (Leibniz IPHT); Albert-Einstein-Straße 9 07745 Jena Germany
| | - Moritz Klein
- Leibniz Institute of Photonic Technology (Leibniz IPHT); Albert-Einstein-Straße 9 07745 Jena Germany
- Institute of Physical Chemistry; Friedrich Schiller University; Helmholtzweg 4 07743 Jena Germany
| | - Helmar Görls
- Institute of Inorganic and Analytical Chemistry; Friedrich Schiller University; Humboldtstraße 8 07743 Jena Germany
| | - Michael Schmitt
- Leibniz Institute of Photonic Technology (Leibniz IPHT); Albert-Einstein-Straße 9 07745 Jena Germany
- Institute of Physical Chemistry; Friedrich Schiller University; Helmholtzweg 4 07743 Jena Germany
| | - Ute Neugebauer
- Center for Sepsis Control and Care (CSCC); Jena University Hospital; Am Klinikum 1 07747 Jena Germany
- Leibniz Institute of Photonic Technology (Leibniz IPHT); Albert-Einstein-Straße 9 07745 Jena Germany
- Institute of Physical Chemistry; Friedrich Schiller University; Helmholtzweg 4 07743 Jena Germany
| | - Guido Gessner
- Center for Molecular Biomedicine (CMB); Department of Biophysics; Friedrich Schiller University Jena and Jena University Hospital; Hans-Knöll-Straße 2 07745 Jena Germany
| | - Stefan H. Heinemann
- Center for Molecular Biomedicine (CMB); Department of Biophysics; Friedrich Schiller University Jena and Jena University Hospital; Hans-Knöll-Straße 2 07745 Jena Germany
| | - Jürgen Popp
- Center for Sepsis Control and Care (CSCC); Jena University Hospital; Am Klinikum 1 07747 Jena Germany
- Leibniz Institute of Photonic Technology (Leibniz IPHT); Albert-Einstein-Straße 9 07745 Jena Germany
- Institute of Physical Chemistry; Friedrich Schiller University; Helmholtzweg 4 07743 Jena Germany
| | - Matthias Westerhausen
- Institute of Inorganic and Analytical Chemistry; Friedrich Schiller University; Humboldtstraße 8 07743 Jena Germany
| |
Collapse
|
25
|
Impairment of the carnitine/organic cation transporter 1-ergothioneine axis is mediated by intestinal transporter dysfunction in chronic kidney disease. Kidney Int 2017; 92:1356-1369. [PMID: 28754554 DOI: 10.1016/j.kint.2017.04.032] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 04/03/2017] [Accepted: 04/18/2017] [Indexed: 01/01/2023]
Abstract
Carnitine/organic cation transporter 1 (OCTN1) is a specific transporter of the food-derived antioxidant ergothioneine. Ergothioneine is absorbed by intestinal OCTN1, distributed through the bloodstream, and incorporated into each organ by OCTN1. OCTN1 expression is upregulated in injured tissues, and promotes ergothioneine uptake to reduce further damage caused by oxidative stress. However, the role of the OCTN1-ergothioneine axis in kidney-intestine cross-talk and chronic kidney disease (CKD) progression remains unclear. Here we assessed ergothioneine uptake via intestinal OCTN1 and confirmed the expression of OCTN1. The ability of OCTN1 to absorb ergothioneine was diminished in mice with CKD. In combination with OCTN1 dysfunction, OCTN1 localization on the intestinal apical cellular membrane was disturbed in mice with CKD. Proteomic analysis, RT-PCR, Western blotting, and immunohistochemistry revealed that PDZ (PSD95, Dlg, and ZO1), a PDZK1 domain-containing protein that regulates the localization of transporters, was decreased in mice with CKD. Decreased intestinal ergothioneine uptake from food decreased ergothioneine levels in the blood of mice with CKD. Despite increased OCTN1 expression and ergothioneine uptake into the kidneys of mice with CKD, ergothioneine levels did not increase. To identify the role of the OCTN1-ergothioneine axis in CKD, we evaluated kidney damage and oxidative stress in OCTN1-knockout mice with CKD and found that kidney fibrosis worsened. Oxidative stress indicators were increased in OCTN1-knockout mice. Moreover, ergothioneine levels in the blood of patients with CKD decreased, which were restored after kidney transplantation. Thus, a novel inter-organ interaction mediated by transporters is associated with CKD progression.
Collapse
|