1
|
Rhee KH, Yang SA, Pyo MC, Lim JM, Lee KW. MiR-155-5p Elevated by Ochratoxin A Induces Intestinal Fibrosis and Epithelial-to-Mesenchymal Transition through TGF-β Regulated Signaling Pathway In Vitro and In Vivo. Toxins (Basel) 2023; 15:473. [PMID: 37505742 PMCID: PMC10467050 DOI: 10.3390/toxins15070473] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
Ochratoxin A (OTA) is a mycotoxin that induces fibrosis and epithelial-to-mesenchymal transitions (EMT) in kidneys and livers. It enters our bodies through food consumption, where it is absorbed in the intestines. However, the impact of OTA on the intestines is yet to be studied. MicroRNA (miRNAs) are small non-coding single-stranded RNAs that block the transcription of specific mRNAs and are, therefore, involved in many biochemical processes. Our findings indicate that OTA can induce EMT and intestinal fibrosis both in vivo and in vitro. This study examines the impact of OTA on intestinal toxicity and the role of miRNAs in this process. Following OTA treatment, miR-155-5p was the most elevated miRNA by next-generation sequencing. Our research showed that OTA increased miR-155-5p levels through transforming growth factor β (TGF-β), leading to the development of intestinal fibrosis and EMT. Additionally, the study identified that the modulation of TGF-β and miR-155-5p by OTA is linked to the inhibition of CCAAT/enhancer-binding protein β (C/EBPβ) and Smad2/3 accumulation in the progression of intestinal fibrosis.
Collapse
Affiliation(s)
| | | | | | | | - Kwang-Won Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (K.H.R.); (S.A.Y.); (M.C.P.); (J.-M.L.)
| |
Collapse
|
2
|
Frangiamone M, Alonso-Garrido M, Font G, Cimbalo A, Manyes L. Pumpkin extract and fermented whey individually and in combination alleviated AFB1- and OTA-induced alterations on neuronal differentiation invitro. Food Chem Toxicol 2022; 164:113011. [PMID: 35447289 DOI: 10.1016/j.fct.2022.113011] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/04/2022] [Accepted: 04/09/2022] [Indexed: 02/06/2023]
Abstract
Food and feed are daily exposed to mycotoxin contamination which effects may be counteracted by functional compounds like carotenoids and fermented whey. Among mycotoxins, the most toxic and studied are aflatoxin B1 (AFB1) and ochratoxin A (OTA), which neurotoxicity is not well reported. Therefore, SH-SY5Y human neuroblastoma cells ongoing differentiation were exposed during 7 days to digested bread extracts contained pumpkin and fermented whey, individually and in combination, along with AFB1 and OTA and their combination, in order to evaluate their presumed effects on neuronal differentiation. The immunofluorescence analysis of βIII-tubulin and dopamine markers pointed to OTA as the most damaging treatment for cell differentiation. Cell cycle analysis reported the highest significant differences for OTA-contained bread compared to the control in phase G0/G1. Lastly, RNA extraction was performed and gene expression was analyzed by qPCR. The selected genes were related to neuronal differentiation and cell cycle. The addition of functional ingredients in breads not only enhancing the expression of neuronal markers, but also induced an overall improvement of gene expression compromised by mycotoxins activity. These data confirm that in vitro neuronal differentiation may be impaired by AFB1 and OTA-exposure, which could be modulated by bioactive compounds naturally found in diet.
Collapse
Affiliation(s)
- Massimo Frangiamone
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, Universitat de València, Vicent Andrés Estellés s/n, 46100, Burjassot, Spain
| | - Manuel Alonso-Garrido
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, Universitat de València, Vicent Andrés Estellés s/n, 46100, Burjassot, Spain
| | - Guillermina Font
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, Universitat de València, Vicent Andrés Estellés s/n, 46100, Burjassot, Spain
| | - Alessandra Cimbalo
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, Universitat de València, Vicent Andrés Estellés s/n, 46100, Burjassot, Spain.
| | - Lara Manyes
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, Universitat de València, Vicent Andrés Estellés s/n, 46100, Burjassot, Spain
| |
Collapse
|
3
|
Malir F, Louda M, Toman J, Ostry V, Pickova D, Pacovsky J, Brodak M, Pfohl-Leszkowicz A. Investigation of ochratoxin A biomarkers in biological materials obtained from patients suffering from renal cell carcinoma. Food Chem Toxicol 2021; 158:112669. [PMID: 34774926 DOI: 10.1016/j.fct.2021.112669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/08/2021] [Accepted: 11/09/2021] [Indexed: 11/24/2022]
Abstract
Ochratoxin A (OTA) exposure can result in chronic renal diseases and cancer. The incidence of kidney, renal pelvis, and ureter malignant neoplasms in the Czech Republic is approximately 29.5 renal tumours per 100,000 inhabitants. The question arises whether mycotoxins are also involved in kidney disease and cancer. A sensitive validated analytical methodology, based on an immunoaffinity clean-up followed by HPLC with fluorescence detection, was developed to explore whether OTA accumulates in clear renal cell carcinoma-adenocarcinoma in Czech patients. Simultaneously, DNA-adducts and OTA metabolites were qualitatively analysed in tissues and urine. OTA was analysed in 33 kidney and tumour samples from 26 men and 7 women collected during nephrectomy from patients of the East Bohemian region from 2015 to 2017. OTA was found in 76% of the analysed samples. Its concentrations ranged from not detectable to 390 ng/kg with a median of 167 ng/kg in kidney samples and from not detectable to 430 ng/kg with a median of 122 ng/kg in tumour samples. Urinary OTA metabolites and DNA adducts were qualitatively analysed for the corresponding 20 patients. The presence of some OTA metabolites such as ochratoxin A hydroquinone and/or decarboxylated ochratoxin A hydroquinone correlate with the presence of OTA-DNA adducts.
Collapse
Affiliation(s)
- Frantisek Malir
- University of Hradec Kralove, Faculty of Science, Department of Biology, Hradec Kralove, Czech Republic.
| | - Miroslav Louda
- Charles University Medical School and Teaching Hospital, Department of Urology, Hradec Kralove, Czech Republic
| | - Jakub Toman
- University of Hradec Kralove, Faculty of Science, Department of Biology, Hradec Kralove, Czech Republic
| | - Vladimir Ostry
- University of Hradec Kralove, Faculty of Science, Department of Biology, Hradec Kralove, Czech Republic; National Institute of Public Health in Prague, Center for Health, Nutrition and Food, Brno, Czech Republic
| | - Darina Pickova
- University of Hradec Kralove, Faculty of Science, Department of Biology, Hradec Kralove, Czech Republic
| | - Jaroslav Pacovsky
- Charles University Medical School and Teaching Hospital, Department of Urology, Hradec Kralove, Czech Republic
| | - Milos Brodak
- Charles University Medical School and Teaching Hospital, Department of Urology, Hradec Kralove, Czech Republic
| | - Annie Pfohl-Leszkowicz
- formerly University of Toulouse, INP/ENSAT Toulouse, Department Bioprocess & Microbial Systems, Laboratory Chemical Engineering, Auzeville - Tolosane, France
| |
Collapse
|
4
|
Ochratoxin A-Induced Nephrotoxicity: Up-to-Date Evidence. Int J Mol Sci 2021; 22:ijms222011237. [PMID: 34681895 PMCID: PMC8539333 DOI: 10.3390/ijms222011237] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 02/07/2023] Open
Abstract
Ochratoxin A (OTA) is a mycotoxin widely found in various foods and feeds that have a deleterious effect on humans and animals. It has been shown that OTA causes multiorgan toxicity, and the kidney is the main target of OTA among them. This present article aims to review recent and latest intracellular molecular interactions and signaling pathways of OTA-induced nephrotoxicity. Pyroptosis, lipotoxicity, organic anionic membrane transporter, autophagy, the ubiquitin-proteasome system, and histone acetyltransferase have been involved in the renal toxicity caused by OTA. Meanwhile, the literature reviewed the alternative or method against OTA toxicity by reducing ROS production, oxidative stress, activating the Nrf2 pathway, through using nanoparticles, a natural flavonoid, and metal supplement. The present review discloses the molecular mechanism of OTA-induced nephrotoxicity, providing opinions and strategies against OTA toxicity.
Collapse
|
5
|
Wang X, Li X, Wang Y, Qin Y, Yan B, Martyniuk CJ. A comprehensive review of strobilurin fungicide toxicity in aquatic species: Emphasis on mode of action from the zebrafish model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 275:116671. [PMID: 33582629 DOI: 10.1016/j.envpol.2021.116671] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/25/2021] [Accepted: 02/02/2021] [Indexed: 06/12/2023]
Abstract
Strobilurins are popular fungicides used in agriculture on a global scale. Due to their widespread use as agrochemicals, they can enter aquatic environments at concentrations that can elicit adverse effects in organisms. This review synthesizes the current state of knowledge regarding the toxic effects of strobilurin fungicides on aquatic species, including algal species, Daphnia magna, and fish species, to determine risk to aquatic organisms and ecosystems. Data show that the toxicities of strobilurins vary widely across aquatic species. Strobilurins bind cytochrome bc1 in mitochondrial complex III in fungi, and as such, research in aquatic species has focused on mitochondria-related endpoints following exposures to strobilurins. In fish, studies into the activities of mitochondrial complexes and the expression of genes involved in the electron transfer chain have been conducted, converging on the theme that mitochondrial complexes and their enzymes are impaired by strobilurins. In general, the order of toxicity of strobilurins for fish species are pyraoxystrobin > pyraclostrobin ≈ trifloxystrobin > picoxystrobin > kresoxim-methyl > fluoxastrobin > azoxystrobin. In addition to mitochondrial toxicity, studies also report genotoxicity, immunotoxicity, cardiotoxicity, neurotoxicity, and endocrine disruption, and each of these events can potentially impact whole organism-level processes such as development, reproduction, and behavior. Screening data from the US Environmental Protection Agency ToxCast database supports the hypothesis that these fungicides may act as endocrine disruptors, and high throughput data suggest estrogen receptor alpha and thyroid hormone receptor beta can be activated by some strobilurins. It is recommended that studies investigate the potential for endocrine disruption by strobilurins more thoroughly in aquatic species. Based on molecular, physiological, and developmental outcomes, a proposed adverse outcome pathway is presented with complex III inhibition in the electron transfer chain as a molecular initiating event. This review comprehensively addresses sub-lethal toxicity mechanisms of strobilurin fungicides, important as the detection of strobilurins in aquatic environments suggests exposure risks in wildlife.
Collapse
Affiliation(s)
- Xiaohong Wang
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, China
| | - Xiaoyu Li
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, China
| | - Yue Wang
- The New Hope Liuhe Co., Ltd., Qingdao, China
| | - Yingju Qin
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, China
| | - Bing Yan
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, China.
| | - Christopher J Martyniuk
- Center for Environmental and Human Toxicology, Department of Physiological Sciences, College of Veterinary Medicine, UF Genetics Institute, Interdisciplinary Program in Biomedical Sciences in Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
6
|
Cordelli E, Bignami M, Pacchierotti F. Comet assay: a versatile but complex tool in genotoxicity testing. Toxicol Res (Camb) 2021; 10:68-78. [PMID: 33613974 PMCID: PMC7885189 DOI: 10.1093/toxres/tfaa093] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/26/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
The comet assay is a versatile method for measuring DNA strand breaks in individual cells. It can also be applied to cells isolated from treated animals. In this review, we highlight advantages and limitations of this in vivo comet assay in a regulatory context. Modified versions of the standard protocol detect oxidized DNA bases and may be used to reveal sites of DNA base loss, DNA interstrand crosslinks, and the extent of DNA damage induced indirectly by reactive oxygen species elicited by chemical-induced oxidative stress. The assay is, however, at best semi-quantitative, and we discuss possible approaches to improving DNA damage quantitation and highlight the necessity of optimizing protocol standardization to enhance the comparability of results between laboratories. As a genotoxicity test in vivo, the in vivo comet assay has the advantage over the better established micronucleus erythrocyte test that it can be applied to any organ, including those that are specific targets of chemical carcinogens or those that are the first sites of contact of ingested or inhaled mutagens. We illustrate this by examples of its use in risk assessment for the food contaminants ochratoxin and furan. We suggest that improved quantitation is required to reveal the full potential of the comet assay and enhance its role in the battery of in vivo approaches to characterize the mechanisms of toxicity and carcinogenicity of chemicals and to aid the determination of safe human exposure limits.
Collapse
Affiliation(s)
- Eugenia Cordelli
- Territorial and Production Systems Sustainability Department, Health Protection Technology Division, ENEA, CR Casaccia, Via Anguillarese 301, Rome 00123, Italy
| | - Margherita Bignami
- Department of Environment and Health, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Francesca Pacchierotti
- Territorial and Production Systems Sustainability Department, Health Protection Technology Division, ENEA, CR Casaccia, Via Anguillarese 301, Rome 00123, Italy
| |
Collapse
|
7
|
Aşcı Çelik D, Gurbuz N, Toğay VA, Özçelik N. Ochratoxin A causes cell cycle arrest in G1 and G1/S phases through p53 in HK-2 cells. Toxicon 2020; 180:11-17. [DOI: 10.1016/j.toxicon.2020.03.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/18/2020] [Accepted: 03/31/2020] [Indexed: 12/20/2022]
|
8
|
EFSA Panel on Contaminants in the Food Chain (CONTAM), Schrenk D, Bodin L, Chipman JK, del Mazo J, Grasl‐Kraupp B, Hogstrand C, Hoogenboom L(R, Leblanc J, Nebbia CS, Nielsen E, Ntzani E, Petersen A, Sand S, Schwerdtle T, Vleminckx C, Wallace H, Alexander J, Dall'Asta C, Mally A, Metzler M, Binaglia M, Horváth Z, Steinkellner H, Bignami M. Risk assessment of ochratoxin A in food. EFSA J 2020; 18:e06113. [PMID: 37649524 PMCID: PMC10464718 DOI: 10.2903/j.efsa.2020.6113] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The European Commission asked EFSA to update their 2006 opinion on ochratoxin A (OTA) in food. OTA is produced by fungi of the genus Aspergillus and Penicillium and found as a contaminant in various foods. OTA causes kidney toxicity in different animal species and kidney tumours in rodents. OTA is genotoxic both in vitro and in vivo; however, the mechanisms of genotoxicity are unclear. Direct and indirect genotoxic and non-genotoxic modes of action might each contribute to tumour formation. Since recent studies have raised uncertainty regarding the mode of action for kidney carcinogenicity, it is inappropriate to establish a health-based guidance value (HBGV) and a margin of exposure (MOE) approach was applied. For the characterisation of non-neoplastic effects, a BMDL 10 of 4.73 μg/kg body weight (bw) per day was calculated from kidney lesions observed in pigs. For characterisation of neoplastic effects, a BMDL 10 of 14.5 μg/kg bw per day was calculated from kidney tumours seen in rats. The estimation of chronic dietary exposure resulted in mean and 95th percentile levels ranging from 0.6 to 17.8 and from 2.4 to 51.7 ng/kg bw per day, respectively. Median OTA exposures in breastfed infants ranged from 1.7 to 2.6 ng/kg bw per day, 95th percentile exposures from 5.6 to 8.5 ng/kg bw per day in average/high breast milk consuming infants, respectively. Comparison of exposures with the BMDL 10 based on the non-neoplastic endpoint resulted in MOEs of more than 200 in most consumer groups, indicating a low health concern with the exception of MOEs for high consumers in the younger age groups, indicating a possible health concern. When compared with the BMDL 10 based on the neoplastic endpoint, MOEs were lower than 10,000 for almost all exposure scenarios, including breastfed infants. This would indicate a possible health concern if genotoxicity is direct. Uncertainty in this assessment is high and risk may be overestimated.
Collapse
|
9
|
Li L, Chen Y, Jiao D, Yang S, Li L, Li P. Protective Effect of Astaxanthin on Ochratoxin A-Induced Kidney Injury to Mice by Regulating Oxidative Stress-Related NRF2/KEAP1 Pathway. Molecules 2020; 25:molecules25061386. [PMID: 32197464 PMCID: PMC7144393 DOI: 10.3390/molecules25061386] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/12/2020] [Accepted: 03/16/2020] [Indexed: 12/20/2022] Open
Abstract
The present study aimed to investigate the effects of astaxanthin (ASX) on ochratoxin A (OTA)-induced renal oxidative stress and its mechanism of action. Serum kidney markers, histomorphology, ultrastructural observation, and oxidative stress indicators were assessed. Meanwhile, quantitative real-time reverse transcription PCR and western blotting detection of NRF2 (encoding nuclear factor, erythroid 2 like) and members of the NRF2/KEAP1 signaling pathway (KEAP1 (encoding Kelch-like ECH-associated protein), NQO1 (encoding NAD(P)H quinone dehydrogenase), HO-1 (encoding heme oxygenase 1), γ-GCS (gamma-glutamylcysteine synthetase), and GSH-Px (glutathione peroxidase 1)) were performed. Compared with the control group, the OTA-treated group showed significantly increased levels of serum UA (uric acid) and BUN (blood urea nitrogen), tubular epithelial cells were swollen and degenerated, and the levels of antioxidant enzymes decreased significantly, and the expression of NRF2 (cytoplasm), NQO1, HO-1, γ-GCS, and GSH-Px decreased significantly. More importantly, after ASX pretreatment, compared with the OTA group, serum markers were decreased, epithelial cells appeared normal; the expression of antioxidant enzymes increased significantly, NQO1, HO-1, γ-GCS and GSH-Px levels increased significantly, and ASX promoted the transfer of NRF2 from the cytoplasm to the nucleus. These results highlight the protective ability of ASX in renal injury caused by OTA exposure, and provide theoretical support for ASX’s role in other mycotoxin-induced damage.
Collapse
Affiliation(s)
| | | | | | - Shuhua Yang
- Correspondence: (S.Y.); (L.L.); (P.L.); Tel./Fax: +86-24-8848-7156 (S.Y., L.L. & P.L.)
| | - Lin Li
- Correspondence: (S.Y.); (L.L.); (P.L.); Tel./Fax: +86-24-8848-7156 (S.Y., L.L. & P.L.)
| | - Peng Li
- Correspondence: (S.Y.); (L.L.); (P.L.); Tel./Fax: +86-24-8848-7156 (S.Y., L.L. & P.L.)
| |
Collapse
|
10
|
Astaxanthin Protects Ochratoxin A-Induced Oxidative Stress and Apoptosis in the Heart via the Nrf2 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7639109. [PMID: 32190177 PMCID: PMC7073479 DOI: 10.1155/2020/7639109] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/18/2019] [Accepted: 12/21/2019] [Indexed: 12/15/2022]
Abstract
This study assessed the protective mechanism of astaxanthin (ASX) against ochratoxin A- (OTA-) induced cardiac injury in mice. Four groups of mice were established: control group (0.1 mL olive oil + 0.1 mL NaHCO2), OTA group (0.1 mL OTA 5 mg/kg body weight), ASX group (0.1 mL ASX 100 mg/kg body weight), and ASX + OTA group (0.1 mL ASX 100 mg/kg body weight, 2 h later, 0.1 mL OTA 5 mg/kg body weight). The test period lasted for 27 days (7 days of dosing, 2 days of rest). Electrocardiogram, body weight, heart weight, tissue pathology, oxidative markers (malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH)), biochemical markers (creatine kinase (CK), creatine kinase isoenzyme (CK-MB), and lactate dehydrogenase (LDH)), electron microscopy, TUNEL, and Western blot tests were used to examine the effects of OTA on myocardial injury and ASX detoxification. The results showed that OTA exposure significantly decreased both body weight and heart weight. OTA induced a decrease in heart rate in mice and decreased tissue concentrations of SOD, CAT, and GSH, while increasing serum concentrations of cardiac enzymes (CK, CK-MB, and LDH) and tissue MDA. ASX improved heart rate, cardiac enzymes, and antioxidant levels in mice. The results of tissue pathology and TUNEL assay showed that ASX protects against OTA-induced myocardial injury. In addition, Western blot results showed that the OTA group upregulated Keap1, Bax, Caspase3, and Caspase9, while it downregulated Nrf2, HO-1, and Bcl-2 protein expression. ASX played a protective role by changing the expression of Keap1, Nrf2, HO-1, Bax, Bcl-2, Caspase3, and Caspase9 proteins. These results indicate that the protective mechanism of ASX on the myocardium works through the Keap1-Nrf2 signaling pathway and mitochondria-mediated apoptosis pathway. This study provides a molecular rationale for the mechanism underlying OTA-induced myocardial injury and the protective effect of ASX on the myocardium.
Collapse
|
11
|
Cimbalo A, Alonso-Garrido M, Font G, Manyes L. Toxicity of mycotoxins in vivo on vertebrate organisms: A review. Food Chem Toxicol 2020; 137:111161. [PMID: 32014537 DOI: 10.1016/j.fct.2020.111161] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/31/2022]
Abstract
Mycotoxins are considered to be a major risk factor affecting human and animal health as they are one of the most dangerous contaminants of food and feed. This review aims to compile the research developed up to date on the toxicological effects that mycotoxins can induce on human health, through the examination of a selected number of studies in vivo. AFB1 shows to be currently the most studied mycotoxin in vivo, followed by DON, ZEA and OTA. Scarce data was found for FBs, PAT, CIT, AOH and Fusarium emerging mycotoxins. The majority of them concerned the investigation of immunotoxicity, whereas the rest consisted in the study of genotoxicity, oxidative stress, hepatotoxicity, cytotoxicity, teratogenicity and neurotoxicity. In order to assess the risk, a wide range of different techniques have been employed across the reviewed studies: qPCR, ELISA, IHC, WB, LC-MS/MS, microscopy, enzymatic assays, microarray and RNA-Seq. In the last decade, the attention has been drawn to immunologic and transcriptomic aspects of mycotoxins' action, confirming their toxicity at molecular level. Even though, more in vivo studies are needed to further investigate their mechanism of action on human health.
Collapse
Affiliation(s)
- A Cimbalo
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Avinguda Vicent Andrés Estellés S/n, 46100, Burjassot, Spain.
| | - M Alonso-Garrido
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Avinguda Vicent Andrés Estellés S/n, 46100, Burjassot, Spain
| | - G Font
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Avinguda Vicent Andrés Estellés S/n, 46100, Burjassot, Spain
| | - L Manyes
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Avinguda Vicent Andrés Estellés S/n, 46100, Burjassot, Spain
| |
Collapse
|
12
|
Transcriptome Analysis of Ochratoxin A-Induced Apoptosis in Differentiated Caco-2 Cells. Toxins (Basel) 2019; 12:toxins12010023. [PMID: 31906179 PMCID: PMC7020595 DOI: 10.3390/toxins12010023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/21/2019] [Accepted: 12/24/2019] [Indexed: 12/20/2022] Open
Abstract
Ochratoxin A (OTA), an important mycotoxin that occurs in food and animal feed, has aroused widespread concern in recent years. Previous studies have indicated that OTA causes nephrotoxicity, hepatotoxicity, genotoxicity, immunotoxicity, cytotoxicity, and neurotoxicity. The intestinal toxicity of OTA has gradually become a focus of research, but the mechanisms underlying this toxicity have not been described. Here, differentiated Caco-2 cells were incubated for 48 h with different concentrations of OTA and transcriptome analysis was used to estimate damage to the intestinal barrier. Gene expression profiling was used to compare the characteristics of differentially expressed genes (DEGs). There were altogether 10,090 DEGs, mainly clustered into two downregulation patterns. The Search Tool for Retrieval of Interacting Genes (STRING), which was used to analyze the protein-protein interaction network, indicated that 24 key enzymes were mostly responsible for regulating cell apoptosis. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis was used to validate eight genes, three of which were key genes (CASP3, CDC25B, and EGR1). The results indicated that OTA dose-dependently induces apoptosis in differentiated Caco-2 cells. Transcriptome analysis showed that the impairment of intestinal function caused by OTA might be partly attributed to apoptosis, which is probably associated with downregulation of murine double minute 2 (MDM2) expression and upregulation of Noxa and caspase 3 (CASP3) expression. This study has highlighted the intestinal toxicity of OTA and provided a genome-wide view of biological responses, which provides a theoretical basis for enterotoxicity and should be useful in establishing a maximum residue limit for OTA.
Collapse
|
13
|
Astaxanthin Protects OTA-Induced Lung Injury in Mice through the Nrf2/NF-κB Pathway. Toxins (Basel) 2019; 11:toxins11090540. [PMID: 31533259 PMCID: PMC6784241 DOI: 10.3390/toxins11090540] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 12/12/2022] Open
Abstract
The aim of this research was to evaluate the potential protective mechanism of astaxanthin (ASTA) against oxidative damage and inflammation caused by ochratoxin (OTA) in mouse lung. We divided mice into a control group (CG), an OTA group (PG), an astaxanthin group (AG), and an OTA+ASTA group (JG). Oxidative indices (malondialdehyde (MDA), total superoxide dismutase (T-SOD), and reduced glutathione (GSH)) and inflammatory markers (interleukin 1β (IL-1β), interleukin 6 (IL-6), and tumor necrosis factor α (TNF-α)) were assayed in the lung, and the lung-weight-to-body-weight ratio was calculated. Apoptosis was detected in pathological sections by the TdT-mediated dUTP nick-end labeling (TUNEL) assay. Oxidative damage and inflammation were detected in the lung of mice after exposure to OTA. Besides, Nrf2- and NF-κB-pathway-associated proteins were detected by Western blot. In contrast with OTA, ASTA significantly raised the expression of Nrf2, HO-1, and MnSOD, while the expression of other proteins (Keap1, TLR4, and NF-κB) was significantly decreased. These results indicate that ASTA exerted protective effects against OTA-induced oxidative damage and inflammation in the lung by regulating the Nrf2 and NF-κB pathways.
Collapse
|
14
|
Yang X, Xu W, Huang K, Zhang B, Wang H, Zhang X, Gong L, Luo Y, He X. Precision toxicology shows that troxerutin alleviates ochratoxin A-induced renal lipotoxicity. FASEB J 2018; 33:2212-2227. [PMID: 30247986 DOI: 10.1096/fj.201800742r] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Lipotoxicity is the most common cause of severe kidney disease, with few treatment options available today. Precision toxicology can improve detection of subtle intracellular changes in response to exogenous substrates; thus, it facilitates in-depth research on bioactive molecules that may interfere with the onset of certain diseases. In the current study, troxerutin significantly relieved nephrotoxicity, increased endurance, and improved systemic energy metabolism and renal inflammation in OTA-induced nephrotic mice. Lipidomics showed that troxerutin effectively reduced the levels of triglycerides, phosphatidylcholines, and phosphatidylethanolamines in nephropathy. The mechanism was partly attributable to troxerutin in alleviating the aberrantly up-regulated expression of sphingomyelinase, the cystic fibrosis transmembrane conductance regulator, and chloride channel 2. Renal tubular epithelial cells, the main site of toxin-induced accumulation of lipids in the kidney, were subjected to transcriptomic profiling, which uncovered several metabolic factors relevant to aberrant lipid and lipoprotein metabolism. Our work provides new insights into the molecular features of toxin-induced lipotoxicity in renal tubular epithelial cells in vivo and demonstrates the function of troxerutin in alleviating OTA-induced nephrosis and associated systemic energy metabolism disorders.-Yang, X., Xu, W., Huang, K., Zhang, B., Wang, H., Zhang, X., Gong, L., Luo, Y., He, X. Precision toxicology shows that troxerutin alleviates ochratoxin A-induced renal lipotoxicity.
Collapse
Affiliation(s)
- Xuan Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Wentao Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Beijing Laboratory for Food Quality and Safety, Beijing, China
| | - Kunlun Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Beijing Laboratory for Food Quality and Safety, Beijing, China
| | - Boyang Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Haomiao Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xueqin Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Lijing Gong
- China Academy of Sport and Health Sciences, Beijing Sport University, Beijing, China
| | - Yunbo Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism-Food Safety, Ministry of Agriculture, China
| | - Xiaoyun He
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Safety Assessment of Genetically Modified Organism-Food Safety, Ministry of Agriculture, China
| |
Collapse
|
15
|
Tao Y, Xie S, Xu F, Liu A, Wang Y, Chen D, Pan Y, Huang L, Peng D, Wang X, Yuan Z. Ochratoxin A: Toxicity, oxidative stress and metabolism. Food Chem Toxicol 2018; 112:320-331. [DOI: 10.1016/j.fct.2018.01.002] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/28/2017] [Accepted: 01/02/2018] [Indexed: 02/06/2023]
|
16
|
Manderville RA, Wetmore SD. Mutagenicity of Ochratoxin A: Role for a Carbon-Linked C8-Deoxyguanosine Adduct? JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:7097-7105. [PMID: 28830149 DOI: 10.1021/acs.jafc.6b03897] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Ochratoxin A (OTA) is a fungal toxin that is considered to be a potent kidney carcinogen in rodent models. The toxin produces double strand breaks and has a propensity for deletions, single-base substitutions, and insertions. The toxin reacts covalently with DNA to afford a C8-2'-deoxyguanosine carbon-linked adduct (OT-dG) as the major lesion in animal tissues. Incorporation of model C-linked C8-aryl-dG adducts into the G3 site of the NarI sequence demonstrates a tendency to induce base substitutions and deletion mutations in primer extension assays using model polymerases. The degree of misincorporation induced by the C-linked C8-dG adducts correlates with an ability to adopt the promutagenic syn conformation within the NarI duplex as predicted by molecular dynamics (MD) simulations. MD simulations of the OT-dG adduct within the NarI duplex predict an even greater degree of conformational flexibility, suggesting enhanced in vitro mutagenicity compared to the simpler model C-linked C8-dG adducts. Together these findings support the role of OT-dG in promoting OTA-mediated mutagenicity and carcinogenicity in animal studies.
Collapse
Affiliation(s)
- Richard A Manderville
- Departments of Chemistry and Toxicology, University of Guelph , Guelph, Ontario, Canada N1G 2W1
| | - Stacey D Wetmore
- Department of Chemistry & Biochemistry, University of Lethbridge , Lethbridge, Alberta, Canada T1K 3M4
| |
Collapse
|
17
|
Nohmi T, Masumura K, Toyoda-Hokaiwado N. Transgenic rat models for mutagenesis and carcinogenesis. Genes Environ 2017; 39:11. [PMID: 28174618 PMCID: PMC5289047 DOI: 10.1186/s41021-016-0072-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 12/08/2016] [Indexed: 01/18/2023] Open
Abstract
Rats are a standard experimental animal for cancer bioassay and toxicological research for chemicals. Although the genetic analyses were behind mice, rats have been more frequently used for toxicological research than mice. This is partly because they live longer than mice and induce a wider variety of tumors, which are morphologically similar to those in humans. The body mass is larger than mice, which enables to take samples from organs for studies on pharmacokinetics or toxicokinetics. In addition, there are a number of chemicals that exhibit marked species differences in the carcinogenicity. These compounds are carcinogenic in rats but not in mice. Such examples are aflatoxin B1 and tamoxifen, both are carcinogenic to humans. Therefore, negative mutagenic/carcinogenic responses in mice do not guarantee that the chemical is not mutagenic/carcinogenic to rats or perhaps to humans. To facilitate research on in vivo mutagenesis and carcinogenesis, several transgenic rat models have been established. In general, the transgenic rats for mutagenesis are treated with chemicals longer than transgenic mice for more exact examination of the relationship between mutagenesis and carcinogenesis. Transgenic rat models for carcinogenesis are engineered mostly to understand mechanisms underlying chemical carcinogenesis. Here, we review papers dealing with the transgenic rat models for mutagenesis and carcinogenesis, and discuss the future perspective.
Collapse
Affiliation(s)
- Takehiko Nohmi
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo, 158-8501 Japan
- Present address: Biological Safety Research Center, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo, 158-8501 Japan
| | - Kenichi Masumura
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo, 158-8501 Japan
| | - Naomi Toyoda-Hokaiwado
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo, 158-8501 Japan
| |
Collapse
|
18
|
Abdel-Wahhab MA, Aljawish A, El-Nekeety AA, Abdel-Aziem SH, Hassan NS. Chitosan nanoparticles plus quercetin suppress the oxidative stress, modulate DNA fragmentation and gene expression in the kidney of rats fed ochratoxin A-contaminated diet. Food Chem Toxicol 2017; 99:209-221. [PMID: 27923682 DOI: 10.1016/j.fct.2016.12.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/29/2016] [Accepted: 12/02/2016] [Indexed: 12/18/2022]
Abstract
This study aimed to evaluate the protective role of chitosan nanoparticles (COS-NPs) singly or plus quercetin (Q) against OTA-induced oxidative stress and renal genotoxicity. Twelve groups of male Sprague-Dawley rats were treated orally for 3 weeks included the control group, animals fed OTA-contaminated diet (3 mg/kg diet); COS-NPs-treated groups at low (140 mg/kg b.w.) or high (280 mg/kg b.w.) dose, Q-treated group (50 mg/kg b.w.), Q plus low or high dose of COS-NPs-treated groups and OTA plus Q and/or COS-NPs at the two tested doses-treated groups. The results indicated that COS-NPs were roughly rod in shape with average particle size of 200 nm and zeta potential 31.4 ± 2.8 mV. Animals fed OTA-contaminated diet showed significant changes in serum biochemical parameters, increase kidney MDA and DNA fragmentation and decreased GPx and SOD gene expression accompanied with severe histological changes. Q and/or COS-NPs at the two tested doses induced significant improvements in all tested parameters and succeeded to overcome these effects especially in the animals treated with Q plus the high dose of COS-NPs. It could be concluded that COS-NPs are promise candidate to enhance the antioxidant effect of Q and protect against the nephrotoxicity of OTA in high endemic areas.
Collapse
Affiliation(s)
- Mosaad A Abdel-Wahhab
- Food Toxicology & Contaminants Dept., National Research Center, Dokki, Cairo, Egypt.
| | - Abdulhadi Aljawish
- Laboratory of Nutrition and Toxicology (NUTox), INSERM UMR 866, Bourgogne University, 1 Esplanade Erasme, 21000 Dijon, France
| | - Aziza A El-Nekeety
- Food Toxicology & Contaminants Dept., National Research Center, Dokki, Cairo, Egypt
| | | | - Nabila S Hassan
- Pathology Dept., National Research Center, Dokki, Cairo, Egypt
| |
Collapse
|
19
|
Heussner A, Paget T. Evaluation of renal in vitro models used in ochratoxin research. WORLD MYCOTOXIN J 2016. [DOI: 10.3920/wmj2015.1975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ochratoxin A (OTA) induces renal carcinomas in rodents with a specific localisation in the S3 segment of proximal tubules and distinct early severe tissue alterations, which have been observed also in other species. Pronounced species- and sex-specific differences in toxicity occur and similar effects cannot be excluded in humans, however precise mechanism(s) remain elusive until today. In such cases, the use of in vitro models for mechanistic investigations can be very useful; in particular if a non-genotoxic mechanism of cancer formation is assumed which include cytotoxic effects. However, potential genotoxic mechanisms can also be investigated in vitro. A crucial issue of in vitro research is the choice of the appropriate cell model. Apparently, the cellular target of OTA is the renal proximal tubular cell; therefore cells from this tissue area are the most reasonable model. Furthermore, cells from affected species should be used and can be compared to cells of human origin. Another important parameter is whether to use primary cultures or to choose a cell line from the huge variety of cell lines available. In any case, important characteristics and quality controls need to be verified beforehand. Therefore, this review discusses the renal in vitro models that have been used for the investigation of renal ochratoxin toxicity. In particular, we discuss the choice of the models and the essential parameters making them suitable models for ochratoxin research together with exemplary results from this research. Furthermore, new promising models such as hTERT-immortalised cells and 3D-cultures are briefly discussed.
Collapse
Affiliation(s)
- A.H. Heussner
- Human and Environmental Toxicology, University of Konstanz, Universitätsstrasse 10, 78457 Konstanz, Germany
- Pharmacy Health and Well-being, University of Sunderland, Sciences Complex, Wharncliffe Street, Sunderland SR1 3SD, United Kingdom
| | - T. Paget
- Pharmacy Health and Well-being, University of Sunderland, Sciences Complex, Wharncliffe Street, Sunderland SR1 3SD, United Kingdom
| |
Collapse
|
20
|
Manderville RA, Wetmore SD. C-Linked 8-aryl guanine nucleobase adducts: biological outcomes and utility as fluorescent probes. Chem Sci 2016; 7:3482-3493. [PMID: 29997840 PMCID: PMC6007177 DOI: 10.1039/c6sc00053c] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/23/2016] [Indexed: 12/18/2022] Open
Abstract
Aryl radical species derived from enzymatic transformations of aromatic mutagens preferentially react at the 8-site of the guanine (G) nucleobase to afford carbon-linked 8arylG adducts. The resulting lesions possess altered biophysical and genetic coding properties compared to the precursor G nucleoside in B-form DNA. Unlike other adducts, these lesions also possess useful fluorescent properties, since direct attachment of the 8aryl ring extends the purine π-system to afford G mimics with red-shifted excitation maxima and emission that can be sensitive to the microenvironment of the 8arylG base within nucleic acid structures. In B-form DNA, 8arylG adducts are disruptive to duplex formation because they prefer to adopt the syn-conformation about the bond connecting the nucleobase to the deoxyribose backbone, which perturbs Watson-Crick (WC) H-bonding with the opposing cytosine (C). Thus, in a B-form duplex, the emissive properties of 8arylG adducts can be employed as a tool to provide insight into adduct conformation, which can be related to their biological outcomes. However, since Gs preferentially adopt the syn-conformation in left-handed Z-DNA and antiparallel G-quadruplex (GQ) structures, 8arylG lesions can be inserted into syn-G positions without disrupting H-bonding interactions. In fact, 8arylG lesions can serve as ideal fluorescent probes in an antiparallel GQ because their emission is sensitive to GQ folding. This perspective outlines recent developments in the biological implications of 8arylG formation together with their utility as fluorescent G analogs for use in DNA-based diagnostic systems.
Collapse
Affiliation(s)
- Richard A Manderville
- Department of Chemistry & Toxicology , University of Guelph , Guelph , ON , Canada N1G 2W1 .
| | - Stacey D Wetmore
- Department of Chemistry & Biochemistry , University of Lethbridge , Lethbridge , AB , Canada T1K 3M4 .
| |
Collapse
|
21
|
Nohmi T. Past, Present and Future Directions of gpt delta Rodent Gene Mutation Assays. Food Saf (Tokyo) 2016; 4:1-13. [PMID: 32231899 PMCID: PMC6989157 DOI: 10.14252/foodsafetyfscj.2015024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 12/21/2015] [Indexed: 01/28/2023] Open
Abstract
Genotoxicity is a critical endpoint of toxicity to regulate environmental chemicals. Genotoxic chemicals are believed to have no thresholds for the action and impose genotoxic risk to humans even at very low doses. Therefore, genotoxic carcinogens, which induce tumors via genotoxic mechanisms, are regulated more strictly than non-genotoxic carcinogens, which induce tumors through non-genotoxic mechanisms such as hormonal effects, cell proliferation and cell toxicity. Although Ames bacterial mutagenicity assay is the gold standard to identify genotoxicity of chemicals, the genotoxicity should be further examined in rodents because Ames positive chemicals are not necessarily genotoxic in vivo. To better evaluate the genotoxicity of chemicals in a whole body system, gene mutation assays with gpt delta transgenic mice and rats have been developed. A feature of the assays is to detect point mutations and deletions by two distinct selection methods, ie, gpt and Spi- assays, respectively. The Spi- assay is unique in that it allows analyses of deletions and complex DNA rearrangements induced by double-strand breaks in DNA. Here, I describe the concept of gpt delta gene mutation assays and the application in food safety research, and discuss future perspectives of genotoxicity assays in vivo.
Collapse
Affiliation(s)
- Takehiko Nohmi
- Biological Safety Research Center, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| |
Collapse
|
22
|
Masumura K, Sakamoto Y, Kumita W, Honma M, Nishikawa A, Nohmi T. Genomic integration of lambda EG10 transgene in gpt delta transgenic rodents. Genes Environ 2015; 37:24. [PMID: 27350819 PMCID: PMC4918054 DOI: 10.1186/s41021-015-0024-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 10/20/2015] [Indexed: 01/25/2023] Open
Abstract
Background Transgenic gpt delta mouse and rat models were developed to perform gpt and Spi− assays for in vivo mutagenicity tests. The animals were established by integration of lambda EG10 phage DNA as a transgene into the genome. The inserted position of the transgene on chromosome was determined by fluorescent in situ hybridization and Southern blot analyses; however, the exact position and sequence of the inserted junction were not known. To identify the site and pattern of genomic integration of the transgene copies, genomic DNAs extracted from C57BL/6J gpt delta mice and F344 gpt delta rats were applied to whole genome sequencing and mate-pair analysis. Results The result confirmed that multi-copy lambda EG10 transgenes are inserted at a single position in the mouse chromosome 17. The junction contains 70 bp of overlapped genomic sequences, and it has short homology at both ends. A copy number analysis suggested that the inserted transgenes may contain 41 head-to-tail junctions and 16 junctions of other types such as rearranged abnormal junctions. It suggested that the number of intact copies could be approximately 40 at maximum. In the F344 gpt delta rats, transgenes are inserted at a single position in the rat chromosome 4. The junction contains no overlapped sequence but 72-kb genomic sequence including one gene was deleted. The inserted transgenes may contain 15 head-to-tail junctions and two rearranged junctions. It suggested that the number of intact copies could be 14 at maximum. One germline base substitution in the gpt gene rescued from gpt delta rats was characterized. Conclusions The exact inserted positions of the lambda EG10 transgene in the genome of gpt delta transgenic rodents were identified. The copy number and arrangement of the transgene were analyzed. PCR primers for quick genotyping of gpt delta mice and rats have been designed. Electronic supplementary material The online version of this article (doi:10.1186/s41021-015-0024-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kenichi Masumura
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501 Japan
| | - Yasuteru Sakamoto
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501 Japan ; Present address: Ajinomoto co., inc., Material Development & Application Labs, Research Institute For Bioscience Products & Fine Chemicals, 1-1 Suzuki-cho Kawasaki-ku, Kawasaki-shi, 210-8681 Kanagawa Japan
| | - Wakako Kumita
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501 Japan
| | - Masamitsu Honma
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501 Japan
| | - Akiyoshi Nishikawa
- Biological Safety Research Center, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501 Japan
| | - Takehiko Nohmi
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501 Japan ; Biological Safety Research Center, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501 Japan
| |
Collapse
|
23
|
González-Arias CA, Benitez-Trinidad AB, Sordo M, Robledo-Marenco L, Medina-Díaz IM, Barrón-Vivanco BS, Marín S, Sanchis V, Ramos AJ, Rojas-García AE. Low doses of ochratoxin A induce micronucleus formation and delay DNA repair in human lymphocytes. Food Chem Toxicol 2015; 74:249-54. [PMID: 25455892 DOI: 10.1016/j.fct.2014.10.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 09/30/2014] [Accepted: 10/08/2014] [Indexed: 01/13/2023]
Abstract
The contamination of food commodities by fungal toxins has attracted great interest because many of these mycotoxins are responsible for different diseases, including cancer and other chronic illnesses. Ochratoxin A (OTA) is a mycotoxin naturally present in food, and long-term exposure to food contaminated with low levels of OTA has been associated with renal cancer. In the present study, the cytotoxicity, cytostaticity, and genotoxicity of OTA (0.075-15 µM) in human lymphocytes were evaluated. A comet assay, a modified comet assay (DNA repair assay), which uses N-hydroxyurea (NHU) to detect non-repaired lesions produced by OTA, and a cytokinesis-blocked micronucleus assay were used. Treatments with OTA were not cytotoxic, but OTA caused a cytostatic effect in human lymphocytes at a concentration of 15 µM. OTA (0.075-5 µM) produced a slight increase in the percentage of DNA in the comets and a delay in the DNA repair capacity of the lymphocytes. Micronucleus (MN) induction was observed at OTA concentrations of 1.5 and 5 µM. Our results indicate that OTA induces DNA stable damage at low doses that are neither cytotoxic nor cytostatic, and OTA delays the DNA repair kinetics. These findings indicate that OTA affects two pivotal events in the carcinogenesis pathway.
Collapse
Affiliation(s)
- Cyndia A González-Arias
- Food Technology Department, Lleida University, UTPV-XaRTA, Agrotecnio Center, Av. Rovira Roure 191, Lleida, 25198, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Bondy GS, Caldwell DS, Aziz SA, Coady LC, Armstrong CL, Curran IHA, Koffman RL, Kapal K, Lefebvre DE, Mehta R. Effects of Chronic Ochratoxin A Exposure on p53 Heterozygous and p53 Homozygous Mice. Toxicol Pathol 2015; 43:715-29. [DOI: 10.1177/0192623314568391] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Exposure to the mycotoxin ochratoxin A (OTA) causes nephropathy in domestic animals and rodents and renal tumors in rodents and poultry. Humans are exposed to OTA by consuming foods made with contaminated cereal grains and other commodities. Management of human health risks due to OTA exposure depends, in part, on establishing a mode of action (MOA) for OTA carcinogenesis. To further investigate OTA’s MOA, p53 heterozygous (p53+/−) and p53 homozygous (p53+/+) mice were exposed to OTA in diet for 26 weeks. The former are susceptible to tumorigenesis upon chronic exposure to genotoxic carcinogens. OTA-induced renal damage but no tumors were observed in either strain, indicating that p53 heterozygosity conferred little additional sensitivity to OTA. Renal changes included dose-dependent increases in cellular proliferation, apoptosis, karyomegaly, and tubular degeneration in proximal tubules, which were consistent with ochratoxicosis. The lowest observed effect level for renal changes in p53+/− and p53+/+ mice was 200 μg OTA/kg bw/day. Based on the lack of tumors and the severity of renal and body weight changes at a maximum tolerated dose, the results were interpreted as suggestive of a primarily nongenotoxic (epigenetic) MOA for OTA carcinogenesis in this mouse model.
Collapse
Affiliation(s)
- Genevieve S. Bondy
- Food Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON, Canada
| | - Donald S. Caldwell
- Food Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON, Canada
| | - Syed A. Aziz
- Food Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON, Canada
| | - Laurie C. Coady
- Food Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON, Canada
| | - Cheryl L. Armstrong
- Food Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON, Canada
| | - Ivan H. A. Curran
- Food Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON, Canada
| | | | - Kamla Kapal
- Food Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON, Canada
| | - David E. Lefebvre
- Food Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON, Canada
| | - Rekha Mehta
- Food Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON, Canada
| |
Collapse
|
25
|
Kuroda K, Hibi D, Ishii Y, Yokoo Y, Takasu S, Kijima A, Matsushita K, Masumura KI, Kodama Y, Yanai T, Sakai H, Nohmi T, Ogawa K, Umemura T. Role of p53 in the progression from ochratoxin A-induced DNA damage to gene mutations in the kidneys of mice. Toxicol Sci 2015; 144:65-76. [PMID: 25636497 DOI: 10.1093/toxsci/kfu267] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Carcinogenic doses of ochratoxin A (OTA) cause increases of mutant frequencies (MFs) of the red/gam gene (Spi(-)) in the kidneys of p53-deficient gpt delta mice, but not in p53-proficient mice. Here, we investigated the role of p53 in the progression from OTA-induced DNA damage to gene mutations. To this end, p53-proficient and -deficient mice were administered 5 mg/kg OTA for 3 days or 4 weeks by gavage. After 3 days of administration, comet assays were performed and there were no differences in the degrees of OTA-induced DNA damage between p53-proficient and -deficient mice. However, the frequencies of γ-H2AX-positive tubular epithelial cells in p53-deficient mice were significantly higher than those in p53-proficient mice, implying that p53 inhibited the progression from DNA damage to DNA double-strand breaks (DSBs). Evaluation of global gene expression and relevant mRNA/protein expression levels demonstrated that OTA increased the expression of Cdkn1a, which encodes the p21 protein, in p53-proficient mice, but not in p53-deficient mice. Moreover, in p53-deficient mice, mRNA levels of cell cycle progression and DSB repair (homologous recombination repair [HR])-related genes were significantly increased. Thus, G1/S arrest due to activation of the p53/p21 pathway may contribute to the prevention of DSBs in p53-proficient mice. In addition, single base deletions/insertions/substitutions were predominant, possibly due to HR. Overall, these results suggested that OTA induced DSBs at the carcinogenic target site in mice and that p53/p21-mediated cell cycle control prevented an increase in the formation of DSBs, leading to gene mutations.
Collapse
Affiliation(s)
- Ken Kuroda
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Daisuke Hibi
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Yuji Ishii
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Yuh Yokoo
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Shinji Takasu
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Aki Kijima
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Kohei Matsushita
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Ken-ichi Masumura
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Yukio Kodama
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Tokuma Yanai
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Hiroki Sakai
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Takehiko Nohmi
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Kumiko Ogawa
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Takashi Umemura
- *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan *Division of Pathology, Division of Genetics and Mutagenesis, Division of Toxicology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193 and Biological Safety Research Center, National Institute of Health Sciences, Tokyo 158-8501, Japan
| |
Collapse
|
26
|
Sproviero M, Verwey AMR, Rankin KM, Witham AA, Soldatov DV, Manderville RA, Fekry MI, Sturla SJ, Sharma P, Wetmore SD. Structural and biochemical impact of C8-aryl-guanine adducts within the NarI recognition DNA sequence: influence of aryl ring size on targeted and semi-targeted mutagenicity. Nucleic Acids Res 2014; 42:13405-21. [PMID: 25361967 PMCID: PMC4245952 DOI: 10.1093/nar/gku1093] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Chemical mutagens with an aromatic ring system may be enzymatically transformed to afford aryl radical species that preferentially react at the C8-site of 2′-deoxyguanosine (dG). The resulting carbon-linked C8-aryl-dG adduct possesses altered biophysical and genetic coding properties compared to the precursor nucleoside. Described herein are structural and in vitro mutagenicity studies of a series of fluorescent C8-aryl-dG analogues that differ in aryl ring size and are representative of authentic DNA adducts. These structural mimics have been inserted into a hotspot sequence for frameshift mutations, namely, the reiterated G3-position of the NarI sequence within 12mer (NarI(12)) and 22mer (NarI(22)) oligonucleotides. In the NarI(12) duplexes, the C8-aryl-dG adducts display a preference for adopting an anti-conformation opposite C, despite the strong syn preference of the free nucleoside. Using the NarI(22) sequence as a template for DNA synthesis in vitro, mutagenicity of the C8-aryl-dG adducts was assayed with representative high-fidelity replicative versus lesion bypass Y-family DNA polymerases, namely, Escherichia coli pol I Klenow fragment exo− (Kf−) and Sulfolobus solfataricus P2 DNA polymerase IV (Dpo4). Our experiments provide a basis for a model involving a two-base slippage and subsequent realignment process to relate the miscoding properties of C-linked C8-aryl-dG adducts with their chemical structures.
Collapse
Affiliation(s)
- Michael Sproviero
- Department of Chemistry and Toxicology, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| | - Anne M R Verwey
- Department of Chemistry and Toxicology, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| | - Katherine M Rankin
- Department of Chemistry and Toxicology, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| | - Aaron A Witham
- Department of Chemistry and Toxicology, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| | - Dmitriy V Soldatov
- Department of Chemistry and Toxicology, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| | - Richard A Manderville
- Department of Chemistry and Toxicology, University of Guelph, Guelph, Ontario, Canada, N1G 2W1
| | - Mostafa I Fekry
- Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zürich, 8032 Zürich, Switzerland Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo 11562, Egypt
| | - Shana J Sturla
- Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zürich, 8032 Zürich, Switzerland
| | - Purshotam Sharma
- Department of Chemistry & Biochemistry, University of Lethbridge, Lethbridge, AB, Canada, T1K 3M4
| | - Stacey D Wetmore
- Department of Chemistry and Toxicology, University of Guelph, Guelph, Ontario, Canada, N1G 2W1 Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zürich, 8032 Zürich, Switzerland Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo 11562, Egypt Department of Chemistry & Biochemistry, University of Lethbridge, Lethbridge, AB, Canada, T1K 3M4
| |
Collapse
|
27
|
Sharma P, Manderville RA, Wetmore SD. Structural and energetic characterization of the major DNA adduct formed from the food mutagen ochratoxin A in the NarI hotspot sequence: influence of adduct ionization on the conformational preferences and implications for the NER propensity. Nucleic Acids Res 2014; 42:11831-45. [PMID: 25217592 PMCID: PMC4191402 DOI: 10.1093/nar/gku821] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The nephrotoxic food mutagen ochratoxin A (OTA) produces DNA adducts in rat kidneys, the major lesion being the C8-linked-2′-deoxyguanosine adduct (OTB-dG). Although research on other adducts stresses the importance of understanding the structure of the associated adducted DNA, site-specific incorporation of OTB-dG into DNA has yet to be attempted. The present work uses a robust computational approach to determine the conformational preferences of OTB-dG in three ionization states at three guanine positions in the NarI recognition sequence opposite cytosine. Representative adducted DNA helices were derived from over 2160 ns of simulation and ranked via free energies. For the first time, a close energetic separation between three distinct conformations is highlighted, which indicates OTA-adducted DNA likely adopts a mixture of conformations regardless of the sequence context. Nevertheless, the preferred conformation depends on the flanking bases and ionization state due to deviations in discrete local interactions at the lesion site. The structural characteristics of the lesion thus discerned have profound implications regarding its repair propensity and mutagenic outcomes, and support recent experiments suggesting the induction of double-strand breaks and deletion mutations upon OTA exposure. This combined structural and energetic characterization of the OTB-dG lesion in DNA will encourage future biochemical experiments on this potentially genotoxic lesion.
Collapse
Affiliation(s)
- Purshotam Sharma
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Alberta, T1K 3M4, Canada
| | - Richard A Manderville
- Department of Chemistry and Toxicology, University of Guelph, Guelph, Ontario, N1G 2W1, Canada
| | - Stacey D Wetmore
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Alberta, T1K 3M4, Canada
| |
Collapse
|
28
|
|