1
|
Ranjbar S, Mohammadi P, Pashaei S, Sadeghi M, Mehrabi M, Shabani S, Ebrahimi A, Brühl AB, Khodarahmi R, Brand S. Effect of Aflatoxin B1 on the Nervous System: A Systematic Review and Network Analysis Highlighting Alzheimer's Disease. BIOLOGY 2025; 14:436. [PMID: 40282301 PMCID: PMC12024953 DOI: 10.3390/biology14040436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/13/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
Exposure to aflatoxin (AF) triggers the production of inflammatory molecules and free radicals, leading to chronic inflammation, cancer, and neurodegenerative diseases. This systematic review evaluated the effects of AFB1 on the nervous system, particularly focusing on Alzheimer's disease (AD). A comprehensive search was conducted in Scopus, Cochrane Library, PubMed, and Web of Science databases up to 1 June 2024, without restrictions. From 993 records retrieved, 16 articles were included in the systematic review. AFB1 participates in various biochemical processes and pathological conditions. The study highlights that AFB1 contributes to AD by inducing DNA damage, oxidative stress, and endoplasmic reticulum (ER) stress, impairing DNA repair mechanisms. This results in neuronal damage, cognitive decline, and neurodegeneration. AFB1 also affects key signaling pathways, reduces sodium-potassium pump activity, and disrupts cell cycle regulation involving p53, leading to neurotoxicity, inflammation, and the formation of amyloid-beta (Aβ) plaques and neurofibrillary tangles. Additionally, network analysis revealed 309 genes associated with AD, inflammation, angiopathy, and aflatoxin B1 (AFB1). Among these, ESR1 exhibited the highest number of direct connections to other nodes within the network. The gene TP53 played a pivotal role in mediating communication among genes, while the EP300 gene significantly influenced the overall network structure. Additionally, KEGG enrichment analysis demonstrated that these 309 genes are substantially involved in pathways related to cancer, the FoxO signaling pathway, apoptosis, and AD. In summary, the study highlights that AFB1 causes DNA damage and stress, leading to cognitive decline and neurodegeneration. It disrupts signaling pathways, damages neurons, and affects DNA repair, contributing to neurotoxicity and inflammation. PROSPERO registration number: CRD420250651007.
Collapse
Affiliation(s)
- Samira Ranjbar
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
| | - Somayeh Pashaei
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran
| | - Masoud Sadeghi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
| | - Masomeh Mehrabi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
| | - Sasan Shabani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
| | - Ali Ebrahimi
- Dermatology Department, Hajdaie Dermatology Clinic, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran
| | - Annette B. Brühl
- Center for Affective, Stress and Sleep Disorders, Psychiatric Clinics, University of Basel, 4002 Basel, Switzerland;
| | - Reza Khodarahmi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
- Department of Pharmacognosy and Biotechnology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran
| | - Serge Brand
- Center for Affective, Stress and Sleep Disorders, Psychiatric Clinics, University of Basel, 4002 Basel, Switzerland;
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
- Substance Abuse Prevention Research Center, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
- Division of Sport Science and Psychosocial Health, Department of Sport, Exercise and Health, University of Basel, 4002 Basel, Switzerland
- School of Medicine, Tehran University of Medical Sciences, Tehran 1416753955, Iran
- Center for Disaster Psychiatry and Disaster Psychology, Center of Competence for Disaster Medicine, Swiss Armed Forces, 4002 Basel, Switzerland
| |
Collapse
|
2
|
de Sá SVM, Faria MA, Fernandes JO, Cunha SC. Investigating the individual and mixture cytotoxicity of co-occurring aflatoxin B1, enniatin B, and sterigmatocystin on gastric, intestinal, hepatic, and renal cellular models. Food Chem Toxicol 2024; 188:114640. [PMID: 38583501 DOI: 10.1016/j.fct.2024.114640] [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: 02/02/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/09/2024]
Abstract
This study investigates the individual and combined effects of the mycotoxins, Aflatoxin B1 (AFB1), Enniatin B (ENNB) and Sterigmatocystin (STG), on the cellular viability of gastric (NCI-N87), intestinal (Caco-2), hepatic (Hep-G2) and renal (Hek-293) cells, shedding light on synergistic or antagonistic effects using a constant ratio combination design proposed by Chou-Talalay. These toxins are prevalent in cereal-based foods, frequently consumed by children which raises concerns about their exposure to these mycotoxins. This population is particularly vulnerable to the effects of these toxins due to their underdeveloped organs and incompletely structured physiological processes. Results showed that ENB was the most toxic of the three mycotoxins across all cell lines, while STG and AFB1 showed lower toxicity. The combination of ENNB + STG was found to be the most potent in terms of binary mixtures. In regard to ternary combinations, Caco-2 cells are more sensitive to the tested mycotoxins, whereas NCI-N87 cells show lower levels of cell damage. Worrying dose reduction values (>10-fold) were found for ENNB in binary and ternary combinations at low exposure levels. These findings are significant for establishing initial reference values, which play a pivotal role in estimating reference doses that are subsequently incorporated into the broader risk assessment process.
Collapse
Affiliation(s)
- Soraia V M de Sá
- LAQV-REQUIMTE, Laboratory of Bromatology and Hydrology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Miguel A Faria
- LAQV-REQUIMTE, Laboratory of Bromatology and Hydrology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - José O Fernandes
- LAQV-REQUIMTE, Laboratory of Bromatology and Hydrology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Sara C Cunha
- LAQV-REQUIMTE, Laboratory of Bromatology and Hydrology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| |
Collapse
|
3
|
Yang B, Li X. Unveiling the hub genes associated with aflatoxin B 1-induced hepatotoxicity in chicken. ENVIRONMENTAL RESEARCH 2023; 239:117294. [PMID: 37832762 DOI: 10.1016/j.envres.2023.117294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/22/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023]
Abstract
Aflatoxin B1 (AFB1), a ubiquitous and toxic mycotoxin in human food and animal feedstuff, can impair the function and health of some organs, especially the liver. However, the knowledge about the potential mechanisms of AFB1-induced hepatotoxicity in chickens is limited. Therefore, we analyzed the gene expression data of chicken embryo primary hepatocytes (CEPHs) treated with and without AFB1 at the dose of 0.1 μg/mL which were cultured at 37 °C in Medium 199 (Life Technologies, Shanghai, China) with 5.0% CO2 for 48 h. Totally 1,711 differentially expressed genes (DEGs) were identified, in which 1,170 and 541 genes were up- and down-regulated in AFB1-administrated CEPHs compared to the control, respectively. Biological process analysis suggested that these DEGs might take part in angiogenesis, cell adhesion, immune response, cell differentiation, inflammatory response, cell migration regulation, and blood coagulation. Signaling pathways analysis revealed that these DEGs were mainly linked to metabolic pathways, MAPK, TLR2, and actin cytoskeleton regulation pathways. Moreover, the hub genes, including GYS2, NR1H4, ALDH8A1, and ANGPTL3, might participate in AFB1-induced hepatotoxicity. Taken together, our study offers a new insight into the mechanisms of the AFB1-induced hepatotoxicity.
Collapse
Affiliation(s)
- Bing Yang
- Anhui Key Laboratory of Poultry Infectious Disease Prevention and Control, Anhui Science and Technology University, Chuzhou, 233100, China
| | - Xiaofeng Li
- Anhui Key Laboratory of Poultry Infectious Disease Prevention and Control, Anhui Science and Technology University, Chuzhou, 233100, China.
| |
Collapse
|
4
|
Chen X, F. Abdallah M, Chen X, Rajkovic A. Current Knowledge of Individual and Combined Toxicities of Aflatoxin B1 and Fumonisin B1 In Vitro. Toxins (Basel) 2023; 15:653. [PMID: 37999516 PMCID: PMC10674195 DOI: 10.3390/toxins15110653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/15/2023] [Accepted: 09/05/2023] [Indexed: 11/25/2023] Open
Abstract
Mycotoxins are considered the most threating natural contaminants in food. Among these mycotoxins, aflatoxin B1 (AFB1) and fumonisin B1 (FB1) are the most prominent fungal metabolites that represent high food safety risks, due to their widespread co-occurrence in several food commodities, and their profound toxic effects on humans. Considering the ethical and more humane animal research, the 3Rs (replacement, reduction, and refinement) principle has been promoted in the last few years. Therefore, this review aims to summarize the research studies conducted up to date on the toxicological effects that AFB1 and FB1 can induce on human health, through the examination of a selected number of in vitro studies. Although the impact of both toxins, as well as their combination, were investigated in different cell lines, the majority of the work was carried out in hepatic cell lines, especially HepG2, owing to the contaminants' liver toxicity. In all the reviewed studies, AFB1 and FB1 could invoke, after short-term exposure, cell apoptosis, by inducing several pathways (oxidative stress, the mitochondrial pathway, ER stress, the Fas/FasL signaling pathway, and the TNF-α signal pathway). Among these pathways, mitochondria are the primary target of both toxins. The interaction of AFB1 and FB1, whether additive, synergistic, or antagonistic, depends to great extent on FB1/AFB1 ratio. However, it is generally manifested synergistically, via the induction of oxidative stress and mitochondria dysfunction, through the expression of the Bcl-2 family and p53 proteins. Therefore, AFB1 and FB1 mixture may enhance more in vitro toxic effects, and carry a higher significant risk factor, than the individual presence of each toxin.
Collapse
Affiliation(s)
- Xiangrong Chen
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (M.F.A.); (A.R.)
| | - Mohamed F. Abdallah
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (M.F.A.); (A.R.)
| | - Xiangfeng Chen
- Shandong Analysis and Test Centre, Qilu University of Technology (Shandong Academy of Science), Jinan 250014, China;
| | - Andreja Rajkovic
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium; (M.F.A.); (A.R.)
| |
Collapse
|
5
|
Song Z, Wang X, Chen P, Wang Z, Ma X. A gold nanoflower based dual mode aptasensor for aflatoxin B 1 detection using SERS and fluorescence effect simultaneously. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 301:122963. [PMID: 37302200 DOI: 10.1016/j.saa.2023.122963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/27/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
Aflatoxin B1 (AFB1) is usually the major aflatoxin produced by toxigenic strains and has been identified the most potent natural carcinogen. Here, a SERS/fluorescence dual-mode nanosensor has been designed while gold nanoflowers (AuNFs) was used as substrate for the detection of AFB1. AuNFs exhibited excellent SERS enhancement effect as well as the good fluorescence quenching effect which made the dual signal detection possible. First, the surface of AuNFs was modified with AFB1 aptamer via Au-SH group. Then, the complementary sequence functionalized with Cy5 (the signal molecule) was attached to AuNFs based on the base complementary pairing principle. On this case, Cy5 was close to AuNFs, the SERS intensity was greatly enhanced and the fluorescence intensity was quenched. After incubation with AFB1, the aptamer was preferentially combined to its target AFB1. Thus, the complementary sequence detached from AuNFs which caused the SERS intensity of Cy5 decreased while its fluorescence effect recovered. Then, the quantitative detection was realized with two optical properties. The LOD was calculated to be 0.03 ng/mL. It was a convenient and fast detection method which expanded the application of nanomaterials based multi-signal simultaneous detection.
Collapse
Affiliation(s)
- Zhiyi Song
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China; Collaborative innovation center of food safety and quality control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Xinyi Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China; Collaborative innovation center of food safety and quality control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Peifang Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China; Collaborative innovation center of food safety and quality control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Zhouping Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China; Collaborative innovation center of food safety and quality control in Jiangsu Province, Jiangnan University, Wuxi 214122, China
| | - Xiaoyuan Ma
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi 214122, China; Collaborative innovation center of food safety and quality control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
6
|
Aslanli A, Domnin M, Stepanov N, Efremenko E. Synergistic Antimicrobial Action of Lactoferrin-Derived Peptides and Quorum Quenching Enzymes. Int J Mol Sci 2023; 24:3566. [PMID: 36834977 PMCID: PMC9965131 DOI: 10.3390/ijms24043566] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/13/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Combined use of various antimicrobial peptides (AMPs) with enzymes that hydrolyze the signaling molecules of the resistance mechanism of various microorganisms, quorum sensing (QS), to obtain effective antimicrobials is one of the leading approaches in solving the antimicrobial resistance problem. Our study investigates the lactoferrin-derived AMPs, lactoferricin (Lfcin), lactoferampin and Lf(1-11), as potential partners for combination with enzymes hydrolyzing lactone-containing QS molecules, the hexahistidine-containing organophosphorus hydrolase (His6-OPH) and penicillin acylase, to obtain effective antimicrobial agents with a scope of practical application. The possibility of the effective combination of selected AMPs and enzymes was first investigated in silico using molecular docking method. Based on the computationally obtained results, His6-OPH/Lfcin combination was selected as the most suitable for further research. The study of physical-chemical characteristics of His6-OPH/Lfcin combination revealed the stabilization of enzymatic activity. A notable increase in the catalytic efficiency of action of His6-OPH in combination with Lfcin in the hydrolysis of paraoxon, N-(3-oxo-dodecanoyl)-homoserine lactone and zearalenone used as substrates was established. Antimicrobial efficiency of His6-OPH/Lfcin combination was determined against various microorganisms (bacteria and yeasts) and its improvement was observed as compared to AMP without enzyme. Thus, our findings demonstrate that His6-OPH/Lfcin combination is a promising antimicrobial agent for practical application.
Collapse
Affiliation(s)
| | | | | | - Elena Efremenko
- Chemical Faculty, Lomonosov Moscow State University, Lenin Hills 1/3, 119991 Moscow, Russia
| |
Collapse
|
7
|
Zhang M, Li Q, Wang J, Sun J, Xiang Y, Jin X. Aflatoxin B1 disrupts the intestinal barrier integrity by reducing junction protein and promoting apoptosis in pigs and mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 247:114250. [PMID: 36334341 DOI: 10.1016/j.ecoenv.2022.114250] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 06/16/2023]
Abstract
With the growing diversity and complexity of diet, animals and humans are at risk of exposure to aflatoxin B1 (AFB1), which is a well-known contaminant in the food chain that causes various toxicological effects. The intestine acts as the first barrier against external contaminants, but the effect of AFB1 on intestinal barrier has not been determined. This study aimed to evaluate AFB1 on the intestinal barrier function in vitro and in vivo. In vitro, porcine jejunal epithelial cells (IPEC-J2) were treated with increasing concentrations of AFB1 (10-60 mg/L). In vivo, Kunming (KM) mice were used as controls or gavaged with 1% dimethyl sulfoxide (110 mg/kg b.w.) and AFB1 (0.3 mg/kg b.w.) for 28 days. In IPEC-J2 cells, the cell viability decreased with increasing mycotoxin concentrations, and the viability of IPEC-J2 cells decreased significantly (P < 0.05) when the AFB1 concentrations were greater than 30 mg/L. In addition, quantitative real-time PCR, Western blot analysis, and immunofluorescence results show that AFB1 can downregulate the tight junction proteins and increase the expression levels of Caspase-3 and the ratio of Bax/Bcl-2, suggesting that AFB1 was cytotoxic to IPEC-J2. In vivo, the ratio of villus height to crypt depth, the intestinal wall thickness, the number of intestinal villus per 1000 µm in the jejunum, the expression levels of ZO-1, Claudin-3, Occludin, MUC2, and Caspase-3, and the ratio of Bax/Bcl-2 were significantly affected in mice exposed to AFB1. In vitro and in vivo results showed that the effects of exposure to AFB1 on the intestinal function in the jejunum of KM mice and in the IPEC-J2 was similar, suggesting that AFB1 may adversely affect animal intestine.
Collapse
Affiliation(s)
- Man Zhang
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, Henan, China
| | - Qinghao Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Jun Wang
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, Henan, China
| | - Juan Sun
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Yuqiang Xiang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China
| | - Xin Jin
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan, China.
| |
Collapse
|
8
|
Cytotoxicity of Mycotoxins and Their Combinations on Different Cell Lines: A Review. Toxins (Basel) 2022; 14:toxins14040244. [PMID: 35448853 PMCID: PMC9031280 DOI: 10.3390/toxins14040244] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 12/11/2022] Open
Abstract
Mycotoxins are secondary metabolites of molds and mainly produced by species of the genera Aspergillus, Penicillium and Fusarium. They can be synthesized on the field, during harvest as well as during storage. They are fairly stable compounds and difficult to remove. Among several hundreds of mycotoxins, according to the WHO, ochratoxin A, aflatoxins, zearalenone, deoxynivalenol, patulin, fumonisins as well as T-2 and HT-2 toxins deserve special attention. Cytotoxicity is one of the most important adverse properties of mycotoxins and is generally assessed via the MTT assay, the neutral red assay, the LDH assay, the CCK-8 assay and the ATP test in different cell lines. The apoptotic cell ratio is mainly assessed via flow cytometry. Aside from the assessment of the toxicity of individual mycotoxins, it is important to determine the cytotoxicity of mycotoxin combinations. Such combinations often exhibit stronger cytotoxicity than individual mycotoxins. The cytotoxicity of different mycotoxins often depends on the cell line used in the experiment and is frequently time- and dose-dependent. A major drawback of assessing mycotoxin cytotoxicity in cell lines is the lack of interaction typical for complex organisms (for example, immune responses).
Collapse
|
9
|
Kurup AH, Patras A, Pendyala B, Vergne MJ, Bansode RR. Evaluation of Ultraviolet-Light (UV-A) Emitting Diodes Technology on the Reduction of Spiked Aflatoxin B1 and Aflatoxin M1 in Whole Milk. FOOD BIOPROCESS TECH 2022. [DOI: 10.1007/s11947-021-02731-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
10
|
Yang B, Li L, Geng H, Wang G, Zhang C, Yang S, Zhao Y, Xing F, Liu Y. Detoxification of aflatoxin B1 by H2SO3 during maize wet processing, and toxicity assessment of the transformation product of aflatoxin B1. Food Control 2022. [DOI: 10.1016/j.foodcont.2021.108444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
11
|
The Protective Effects of Lactoferrin on Aflatoxin M1-Induced Compromised Intestinal Integrity. Int J Mol Sci 2021; 23:ijms23010289. [PMID: 35008712 PMCID: PMC8745159 DOI: 10.3390/ijms23010289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/15/2021] [Accepted: 12/18/2021] [Indexed: 11/17/2022] Open
Abstract
Aflatoxin M1 (AFM1), the only toxin with maximum residue levels in milk, has adverse effects on the intestinal barrier, resulting in intestinal inflammatory disease. Lactoferrin (LF), one of the important bioactive proteins in milk, performs multiple biological functions, but knowledge of the protective effects of LF on the compromised intestinal barrier induced by AFM1 has not been investigated. In the present study, results using Balb/C mice and differentiated Caco-2 cells showed that LF intervention decreased AFM1-induced increased intestinal permeability, improved the protein expression of claudin-3, occludin and ZO-1, and repaired the injured intestinal barrier. The transcriptome and proteome were used to clarify the underlying mechanisms. It was found that LF reduced the intestinal barrier dysfunction caused by AFM1 and was associated with intestinal cell survival related pathways, such as cell cycle, apoptosis and MAPK signaling pathway and intestinal integrity related pathways including endocytosis, tight junction, adherens junction and gap junction. The cross-omics analysis suggested that insulin receptor (INSR), cytoplasmic FMR1 interacting protein 2 (CYFIP2), dedicator of cytokinesis 1 (DOCK1) and ribonucleotide reductase regulatory subunit M2 (RRM2) were the potential key regulators as LF repaired the compromised intestinal barrier. These findings indicated that LF may be an alternative treatment for the compromised intestinal barrier induced by AFM1.
Collapse
|
12
|
Lactoferrin Alleviated AFM1-Induced Apoptosis in Intestinal NCM 460 Cells through the Autophagy Pathway. Foods 2021; 11:foods11010023. [PMID: 35010149 PMCID: PMC8750231 DOI: 10.3390/foods11010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/25/2022] Open
Abstract
Aflatoxin M1 (AFM1) is the only mycotoxin with maximum residue limit in milk, which may result in serious human diseases. On the contrary, lactoferrin (Lf) is an active protein with multiple functions. Studies have confirmed that Lf has a powerful potential to protect the intestines, but the influence of Lf on mycotoxins is not clear. This study aims to explore whether Lf can protect the cytotoxicity induced by AFM1, and determine the underlying mechanisms in human normal colonic epithelial NCM460 cells. The results indicated that AFM1 decreased the cell viability, and increased the levels of apoptosis and autophagy of NCM460 cells. Lf can alleviate the cytotoxicity induced by AFM1 through enhancing cell viability, significantly down-regulated the expression of apoptotic genes and proteins (BAX, caspase3, caspase9, caspase3, and caspase9), and regulated the gene and protein expression of autophagy factors (Atg5, Atg7, Atg12, Beclin1, ULK1, ULK2, LC3, and p62). Furthermore, interference of the key gene Atg5 of autophagy can reduce AFM1-induced apoptosis, which is consistent with the role of Lf, implying that Lf may protect AFM1-induced intestinal injury by inhibiting excessive autophagy-mediated apoptosis. Taken together, our data indicated that Lf has a mitigating effect on apoptosis induced by AFM1 through the autophagy pathway.
Collapse
|
13
|
Li B, Zhang B, Liu X, Zheng Y, Han K, Liu H, Wu C, Li J, Fan S, Peng W, Zhang F, Liu X. The effect of lactoferrin in aging: role and potential. Food Funct 2021; 13:501-513. [PMID: 34928288 DOI: 10.1039/d1fo02750f] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Aging is frequently accompanied by various types of physiological deterioration, which increases the risk of human pathologies. Global public health efforts to increase human lifespan have increasingly focused on lowering the risk of aging-related diseases, such as diabetes, neurodegenerative diseases, cardiovascular disease, and cancers. Dietary intervention is a promising approach to maintaining human health during aging. Lactoferrin (LF) is known for its physiologically pleiotropic properties. Anti-aging interventions of LF have proven to be safe and effective for various pharmacological activities, such as anti-oxidation, anti-cellular senescence, anti-inflammation, and anti-carcinogenic. Moreover, LF has a pivotal role in modulating the major signaling pathways that influence the longevity of organisms. Thus, LF is expected to be able to attenuate the process of aging and greatly ameliorate its effects.
Collapse
Affiliation(s)
- Bing Li
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Bo Zhang
- Henan Key Laboratory of Rare Earth Functional Materials, The Key Laboratory of Rare Earth Functional Materials and Applications, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Xudong Liu
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Yidan Zheng
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Kuntong Han
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Henan Liu
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Changjing Wu
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Jin Li
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Shuhua Fan
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Weifeng Peng
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Fuli Zhang
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Xiaomeng Liu
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| |
Collapse
|
14
|
Cao X, Liu C, Zhang M, Bi R, Fu M, Korik E, Chen J, Gao J, Semak I, Liu J. Bovine lactoferrin and Lentinus edodes mycelia polysaccharide complex: The formation and the activity to protect islet β cells. Int J Biol Macromol 2021; 191:811-820. [PMID: 34592222 DOI: 10.1016/j.ijbiomac.2021.09.143] [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: 07/12/2021] [Revised: 09/10/2021] [Accepted: 09/18/2021] [Indexed: 11/20/2022]
Abstract
The formation of complexes may be used for the development of delivery systems in foods field. The aim of this study was to explore the interaction mechanism between Lentinus edodes mycelia polysaccharide (LMP) and bovine lactoferrin (BLF), and the activity of LMP-BLF complex to inhibit oxidative stress in islet β cells. The interaction mechanisms of LMP with BLF were investigated with multi-spectroscopic techniques. The multi-spectroscopic analysis result showed that LMP bound with BLF by van der Waals force and hydrogen bond. The quenching mechanism of BLF with LMP was static quenching. Cell viability, reactive oxygen species (ROS) level, apoptosis and the related signaling pathways were detected with thiazolyl blue tetrazolium bromide (MTT) assay, 2,7-Dichlorofluorescin diacetate (DCFH-DA) staining, Hoechst 33258 staining and Western blot methods respectively. The complex alleviated apoptosis induced by hydrogen peroxide (H2O2), and inhibited oxidative stress via MAPK pathways in MIN6 cells. In addition, the complex was able to promote glucose uptake in HepG2 cells. These results will broaden our understanding of LMP-BLF complexes and the applications of polysaccharide-protein complexes in the foods field.
Collapse
Affiliation(s)
- Xiangyu Cao
- Department of Biological Sciences, School of life Science, Liaoning University, Chongshan Road 66, Shenyang 110036, PR China
| | - Chengying Liu
- Department of Biological Sciences, School of life Science, Liaoning University, Chongshan Road 66, Shenyang 110036, PR China
| | - Mengmeng Zhang
- Department of Biochemistry Belarusian State University, Nezavisimisty Ave., 4, BSU, The Faculty of Biology, 220030 Minsk, Belarus
| | - Ruochen Bi
- Department of Biological Sciences, School of life Science, Liaoning University, Chongshan Road 66, Shenyang 110036, PR China
| | - Mingyang Fu
- Department of Biological Sciences, School of life Science, Liaoning University, Chongshan Road 66, Shenyang 110036, PR China
| | - Elena Korik
- Department of Biochemistry Belarusian State University, Nezavisimisty Ave., 4, BSU, The Faculty of Biology, 220030 Minsk, Belarus
| | - Jiahe Chen
- Department of Biological Sciences, School of life Science, Liaoning University, Chongshan Road 66, Shenyang 110036, PR China
| | - Jianyun Gao
- Department of Biological Sciences, School of life Science, Liaoning University, Chongshan Road 66, Shenyang 110036, PR China
| | - Igor Semak
- Department of Biochemistry Belarusian State University, Nezavisimisty Ave., 4, BSU, The Faculty of Biology, 220030 Minsk, Belarus.
| | - Jianli Liu
- Department of Biological Sciences, School of life Science, Liaoning University, Chongshan Road 66, Shenyang 110036, PR China.
| |
Collapse
|
15
|
Jahangiri-Dehaghani F, Zare H, Shekari Z. Encapsulation of hemin in Fe-based metal-organic frameworks and its application for the direct determination of aflatoxin M1. WORLD MYCOTOXIN J 2021. [DOI: 10.3920/wmj2020.2578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A label-free electrochemical aptasensor was constructed for the sensitive and selective determination of AFM1. For preparation of the aptasensor, the AFM1 aptamer was immobilised on the surface of a glassy carbon electrode modified with hemin encapsulated in Fe-based metal-organic frameworks (hemin@Fe-MIL-101). The morphology and the structure of Fe-MIL-101 and hemin@Fe-MIL-101 were evaluated by scanning electron microscopy, Fourier-transform infrared spectroscopy, X-ray powder diffraction and Brunauer-Emmett-Teller-N2 sorption methods. Electrochemical impedance spectroscopy and cyclic voltammetry were performed to monitor the fabrication process of the electrochemical aptasensor. The electrochemical reduction current of hemin encapsulated in Fe-MIL-101 serves as a signal for the quantitative determination of AFM1. Differential pulse voltammetry was done to determine the AFM1 concentration in the linear range of 1.0×10-1-100.0 ng/ml. The detection limit of AFM1 was estimated to be 4.6×10-2 ng/ml. Finally, the fabricated aptasensor was applied to determine AFM1 in raw and boiled milk samples.
Collapse
Affiliation(s)
| | - H.R. Zare
- Department of Chemistry, Faculty of Science, Yazd University, Yazd, 89195-741, Iran
| | - Z. Shekari
- Department of Chemistry, Faculty of Science, Yazd University, Yazd, 89195-741, Iran
| |
Collapse
|
16
|
Wu K, Jia S, Zhang J, Zhang C, Wang S, Rajput SA, Sun L, Qi D. Transcriptomics and flow cytometry reveals the cytotoxicity of aflatoxin B 1 and aflatoxin M 1 in bovine mammary epithelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 209:111823. [PMID: 33360594 DOI: 10.1016/j.ecoenv.2020.111823] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/13/2020] [Accepted: 12/16/2020] [Indexed: 06/12/2023]
Abstract
Aflatoxin is a known mycotoxin that pollutes various grains widely in the environment. Aflatoxin B1 (AFB1) and Aflatoxin M1 (AFM1) have been shown to induce cytotoxicity in many cells, yet their effects on mammary epithelial cells remain unclear. In this study, we examined the toxicity and the effects of AFB1 and AFM1 on bovine mammary epithelial cells (BME cells). The cells were treated with AFB1 or AFM1 at a concentration of 0-10 mg/L for 24 or 48 h, followed by cytotoxicity assays, flow cytometry, and transcriptomics. Our results demonstrated that AFB1 and AFM1 induced cell proliferation inhibition, apoptosis and cell cycle arrest. However, the level of intracellular reactive oxygen species has no significant difference. The RNA-Seq results also showed that AFB1 and AFM1 changed many related gene expressions like apoptosis and oxidative stress, cycle, junction, and signaling pathway. Taken together, AFB1 and AFM1 were found to affect cytotoxicity and related gene changes in BME cells. Notably, this study reported that 2 mg/L of AFB1 and AFM1 affected the expression of methylation-related genes, and ultimately altered the rate of m6A methylation in RNA. It may provide a potential direction for toxins to indirectly regulate gene expression by affecting RNA methylation modification. Our research provides some novel insights and data about AFB1 and AFM1 toxicity in BME cells.
Collapse
Affiliation(s)
- Kuntan Wu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Sifan Jia
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jiacai Zhang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Cong Zhang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Shuai Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Shahid Ali Rajput
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Lvhui Sun
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Desheng Qi
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
17
|
Hua Z, Liu R, Chen Y, Liu G, Li C, Song Y, Cao Z, Li W, Li W, Lu C, Liu Y. Contamination of Aflatoxins Induces Severe Hepatotoxicity Through Multiple Mechanisms. Front Pharmacol 2021; 11:605823. [PMID: 33505311 PMCID: PMC7830880 DOI: 10.3389/fphar.2020.605823] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
Aflatoxins (AFs) are commonly contaminating mycotoxins in foods and medicinal materials. Since they were first discovered to cause “turkey X” disease in the United Kingdom in the early 1960s, the extreme toxicity of AFs in the human liver received serious attention. The liver is the major target organ where AFs are metabolized and converted into extremely toxic forms to engender hepatotoxicity. AFs influence mitochondrial respiratory function and destroy normal mitochondrial structure. AFs initiate damage to mitochondria and subsequent oxidative stress. AFs block cellular survival pathways, such as autophagy that eliminates impaired cellular structures and the antioxidant system that copes with oxidative stress, which may underlie their high toxicities. AFs induce cell death via intrinsic and extrinsic apoptosis pathways and influence the cell cycle and growth via microribonucleic acids (miRNAs). Furthermore, AFs induce the hepatic local inflammatory microenvironment to exacerbate hepatotoxicity via upregulation of NF-κB signaling pathway and inflammasome assembly in the presence of Kupffer cells (liver innate immunocytes). This review addresses the mechanisms of AFs-induced hepatotoxicity from various aspects and provides background knowledge to better understand AFs-related hepatoxic diseases.
Collapse
Affiliation(s)
- Zhenglai Hua
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Rui Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Youwen Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Guangzhi Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chenxi Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yurong Song
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiwen Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wen Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Weifeng Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
18
|
The Compromised Intestinal Barrier Induced by Mycotoxins. Toxins (Basel) 2020; 12:toxins12100619. [PMID: 32998222 PMCID: PMC7600953 DOI: 10.3390/toxins12100619] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/23/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
Mycotoxins are fungal metabolites that occur in human foods and animal feeds, potentially threatening human and animal health. The intestine is considered as the first barrier against these external contaminants, and it consists of interconnected physical, chemical, immunological, and microbial barriers. In this context, based on in vitro, ex vivo, and in vivo models, we summarize the literature for compromised intestinal barrier issues caused by various mycotoxins, and we reviewed events related to disrupted intestinal integrity (physical barrier), thinned mucus layer (chemical barrier), imbalanced inflammatory factors (immunological barrier), and dysfunctional bacterial homeostasis (microbial barrier). We also provide important information on deoxynivalenol, a leading mycotoxin implicated in intestinal dysfunction, and other adverse intestinal effects induced by other mycotoxins, including aflatoxins and ochratoxin A. In addition, intestinal perturbations caused by mycotoxins may also contribute to the development of mycotoxicosis, including human chronic intestinal inflammatory diseases. Therefore, we provide a clear understanding of compromised intestinal barrier induced by mycotoxins, with a view to potentially develop innovative strategies to prevent and treat mycotoxicosis. In addition, because of increased combinatorial interactions between mycotoxins, we explore the interactive effects of multiple mycotoxins in this review.
Collapse
|
19
|
Bigot J, Guillot L, Guitard J, Ruffin M, Corvol H, Balloy V, Hennequin C. Bronchial Epithelial Cells on the Front Line to Fight Lung Infection-Causing Aspergillus fumigatus. Front Immunol 2020; 11:1041. [PMID: 32528481 PMCID: PMC7257779 DOI: 10.3389/fimmu.2020.01041] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/30/2020] [Indexed: 12/18/2022] Open
Abstract
Aspergillus fumigatus is an environmental filamentous fungus that can be pathogenic for humans, wherein it is responsible for a large variety of clinical forms ranging from allergic diseases to life-threatening disseminated infections. The contamination occurs by inhalation of conidia present in the air, and the first encounter of this fungus in the human host is most likely with the bronchial epithelial cells. Although alveolar macrophages have been widely studied in the Aspergillus–lung interaction, increasing evidence suggests that bronchial epithelium plays a key role in responding to the fungus. This review focuses on the innate immune response of the bronchial epithelial cells against A. fumigatus, the predominant pathogenic species. We have also detailed the molecular interactants and the effects of the different modes of interaction between these cells and the fungus.
Collapse
Affiliation(s)
- Jeanne Bigot
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service de Parasitologie-Mycologie, Paris, France
| | - Loïc Guillot
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France
| | - Juliette Guitard
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service de Parasitologie-Mycologie, Paris, France
| | - Manon Ruffin
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France
| | - Harriet Corvol
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Trousseau, Service de Pneumologie Pédiatrique, Paris, France
| | - Viviane Balloy
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France
| | - Christophe Hennequin
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service de Parasitologie-Mycologie, Paris, France
| |
Collapse
|
20
|
Arantes MR, Peijnenburg A, Hendriksen PJM, Stoopen G, Almeida TS, Souza TM, Farias DF, Carvalho AFU, Rocha TM, Leal LKAM, Vasconcelos IM, Oliveira JTA. In vitro toxicological characterisation of the antifungal compound soybean toxin (SBTX). Toxicol In Vitro 2020; 65:104824. [PMID: 32165152 DOI: 10.1016/j.tiv.2020.104824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 02/13/2020] [Accepted: 03/08/2020] [Indexed: 12/18/2022]
Abstract
Soybean toxin (SBTX) is a protein isolated from soybean seeds and composed of two polypeptide subunits (17 and 27 kDa). SBTX has in vitro activity against phytopathogenic fungi such as Cercospora sojina, Aspergillus niger, and Penicillium herguei, and yeasts like Candida albicans, C. parapsilosis, Kluyveromyces marxiannus, and Pichia membranifaciens. The present study aimed to analyze in vitro whether SBTX causes any side effects on non-target bacterial and mammalian cells that could impede its potential use as a novel antifungal agent. SBTX at 100 μg/mL and 200 μg/mL did not hinder the growth of the bacteria Salmonella enterica (subspecies enterica serovar choleraesuis), Bacillus subtilis (subspecies spizizenii) and Staphylococcus aureus. Moreover, SBTX at concentrations up to 500 μg/mL did not significantly affect the viability of erythrocytes, neutrophils, and human intestinal Caco-2 cells. To study whether SBTX could induce relevant alterations in gene expression, in vitro DNA microarray experiments were conducted in which differentiated Caco-2 cells were exposed for 24 h to 100 μg/mL or 200 μg/mL SBTX. SBTX up-regulated genes involved in cell cycle and immune response pathways, but down-regulated genes that play a role in cholesterol biosynthesis and platelet degranulation pathways. Thus, although SBTX did not affect bacteria, nor induced cytotoxity in mammalian cells, it affected some biological pathways in the human Caco-2 cell line that warrants further investigation.
Collapse
Affiliation(s)
- Mariana Reis Arantes
- Department of Biochemistry and Molecular Biology, Federal University of Ceara, 60020-181 Fortaleza, CE, Brazil
| | - Ad Peijnenburg
- WFSR, Wageningen University and Research Centre, P.O. Box 230, 6700 AE Wageningen, the Netherlands.
| | - Peter J M Hendriksen
- WFSR, Wageningen University and Research Centre, P.O. Box 230, 6700 AE Wageningen, the Netherlands.
| | - Geert Stoopen
- WFSR, Wageningen University and Research Centre, P.O. Box 230, 6700 AE Wageningen, the Netherlands.
| | - Thiago Silva Almeida
- Department of Biochemistry and Molecular Biology, Federal University of Ceara, 60020-181 Fortaleza, CE, Brazil
| | - Terezinha Maria Souza
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht 6229, ER, the Netherlands.
| | - Davi Felipe Farias
- Department of Molecular Biology, Federal University of Paraíba, 58051-900 Joao Pessoa, PB, Brazil.
| | | | | | | | - Ilka Maria Vasconcelos
- Department of Biochemistry and Molecular Biology, Federal University of Ceara, 60020-181 Fortaleza, CE, Brazil.
| | - Jose Tadeu Abreu Oliveira
- Department of Biochemistry and Molecular Biology, Federal University of Ceara, 60020-181 Fortaleza, CE, Brazil.
| |
Collapse
|
21
|
Peles F, Sipos P, Győri Z, Pfliegler WP, Giacometti F, Serraino A, Pagliuca G, Gazzotti T, Pócsi I. Adverse Effects, Transformation and Channeling of Aflatoxins Into Food Raw Materials in Livestock. Front Microbiol 2019; 10:2861. [PMID: 31921041 PMCID: PMC6917664 DOI: 10.3389/fmicb.2019.02861] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 11/26/2019] [Indexed: 01/18/2023] Open
Abstract
Aflatoxins are wide-spread harmful carcinogenic secondary metabolites produced by Aspergillus species, which cause serious feed and food contaminations and affect farm animals deleteriously with acute or chronic manifestations of mycotoxicoses. On farm, both pre-harvest and post-harvest strategies are applied to minimize the risk of aflatoxin contaminations in feeds. The great economic losses attributable to mycotoxin contaminations have initiated a plethora of research projects to develop new, effective technologies to prevent the highly toxic effects of these secondary metabolites on domestic animals and also to block the carry-over of these mycotoxins to humans through the food chain. Among other areas, this review summarizes the latest findings on the effects of silage production technologies and silage microbiota on aflatoxins, and it also discusses the current applications of probiotic organisms and microbial products in feeding technologies. After ingesting contaminated foodstuffs, aflatoxins are metabolized and biotransformed differently in various animals depending on their inherent and acquired physiological properties. These mycotoxins may cause primary aflatoxicoses with versatile, species-specific adverse effects, which are also dependent on the susceptibility of individual animals within a species, and will be a function of the dose and duration of aflatoxin exposures. The transfer of these undesired compounds from contaminated feed into food of animal origin and the aflatoxin residues present in foods become an additional risk to human health, leading to secondary aflatoxicoses. Considering the biological transformation of aflatoxins in livestock, this review summarizes (i) the metabolism of aflatoxins in different animal species, (ii) the deleterious effects of the mycotoxins and their derivatives on the animals, and (iii) the major risks to animal health in terms of the symptoms and consequences of acute or chronic aflatoxicoses, animal welfare and productivity. Furthermore, we traced the transformation and channeling of Aspergillus-derived mycotoxins into food raw materials, particularly in the case of aflatoxin contaminated milk, which represents the major route of human exposure among animal-derived foods. The early and reliable detection of aflatoxins in feed, forage and primary commodities is an increasingly important issue and, therefore, the newly developed, easy-to-use qualitative and quantitative aflatoxin analytical methods are also summarized in the review.
Collapse
Affiliation(s)
- Ferenc Peles
- Institute of Food Science, Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Debrecen, Hungary
| | - Péter Sipos
- Institute of Nutrition, University of Debrecen, Debrecen, Hungary
| | - Zoltán Győri
- Institute of Nutrition, University of Debrecen, Debrecen, Hungary
| | - Walter P. Pfliegler
- Department of Molecular Biotechnology and Microbiology, Institute of Biotechnology, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| | - Federica Giacometti
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Andrea Serraino
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Giampiero Pagliuca
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Teresa Gazzotti
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - István Pócsi
- Department of Molecular Biotechnology and Microbiology, Institute of Biotechnology, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
22
|
Lepanto MS, Rosa L, Cutone A, Scotti MJ, Conte AL, Marazzato M, Zagaglia C, Longhi C, Berlutti F, Musci G, Valenti P, Conte MP. Bovine Lactoferrin Pre-Treatment Induces Intracellular Killing of AIEC LF82 and Reduces Bacteria-Induced DNA Damage in Differentiated Human Enterocytes. Int J Mol Sci 2019; 20:ijms20225666. [PMID: 31726759 PMCID: PMC6888356 DOI: 10.3390/ijms20225666] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 11/10/2019] [Indexed: 12/17/2022] Open
Abstract
LF82, a prototype of adherent-invasive E. coli (AIEC), is able to adhere to, invade, survive and replicate into intestinal epithelial cells. LF82 is able to enhance either its adhesion and invasion by up-regulating carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM-6), the main cell surface molecule for bacterial adhesion, and its intracellular survival by inducing host DNA damage, thus blocking the cellular cycle. Lactoferrin (Lf) is a multifunctional cationic glycoprotein of natural immunity, exerting an anti-invasive activity against LF82 when added to Caco-2 cells at the moment of infection. Here, the infection of 12 h Lf pre-treated Caco-2 cells was carried out at a time of 0 or 3 or 10 h after Lf removal from culture medium. The effect of Lf pre-treatment on LF82 invasiveness, survival, cell DNA damage, CEACAM-6 expression, apoptosis induction, as well as on Lf subcellular localization, has been evaluated. Lf, even if removed from culture medium, reduced LF82 invasion and survival as well as bacteria-induced DNA damage in Caco-2 cells independently from induction of apoptosis, modulation of CEACAM-6 expression and Lf sub-cellular localization. At our knowledge, this is the first study showing that the sole Lf pre-treatment can activate protective intracellular pathways, reducing LF82 invasiveness, intracellular survival and cell-DNA damages.
Collapse
Affiliation(s)
- Maria Stefania Lepanto
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (M.J.S.); (A.L.C.); (M.M.); (C.Z.); (C.L.); (F.B.); (P.V.)
| | - Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (M.J.S.); (A.L.C.); (M.M.); (C.Z.); (C.L.); (F.B.); (P.V.)
| | - Antimo Cutone
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (A.C.); (G.M.)
| | - Mellani Jinnett Scotti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (M.J.S.); (A.L.C.); (M.M.); (C.Z.); (C.L.); (F.B.); (P.V.)
| | - Antonietta Lucia Conte
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (M.J.S.); (A.L.C.); (M.M.); (C.Z.); (C.L.); (F.B.); (P.V.)
| | - Massimiliano Marazzato
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (M.J.S.); (A.L.C.); (M.M.); (C.Z.); (C.L.); (F.B.); (P.V.)
| | - Carlo Zagaglia
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (M.J.S.); (A.L.C.); (M.M.); (C.Z.); (C.L.); (F.B.); (P.V.)
| | - Catia Longhi
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (M.J.S.); (A.L.C.); (M.M.); (C.Z.); (C.L.); (F.B.); (P.V.)
| | - Francesca Berlutti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (M.J.S.); (A.L.C.); (M.M.); (C.Z.); (C.L.); (F.B.); (P.V.)
| | - Giovanni Musci
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (A.C.); (G.M.)
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (M.J.S.); (A.L.C.); (M.M.); (C.Z.); (C.L.); (F.B.); (P.V.)
| | - Maria Pia Conte
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (M.J.S.); (A.L.C.); (M.M.); (C.Z.); (C.L.); (F.B.); (P.V.)
- Correspondence:
| |
Collapse
|
23
|
Taranu I, Marin DE, Palade M, Pistol GC, Chedea VS, Gras MA, Rotar C. Assessment of the efficacy of a grape seed waste in counteracting the changes induced by aflatoxin B1 contaminated diet on performance, plasma, liver and intestinal tissues of pigs after weaning. Toxicon 2019; 162:24-31. [PMID: 30849456 DOI: 10.1016/j.toxicon.2019.02.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 02/26/2019] [Accepted: 02/28/2019] [Indexed: 01/24/2023]
Abstract
The aim of this study was to investigate the potential of a grape seed byproduct to mitigate the harmful damage produced by aflatoxin B1 (AFB1) at systemic level in plasma and liver as well as at local level in the gastrointestinal tract in weaned piglets. Twenty four crossbred pigs (TOPIG) were randomly assigned to one of four experimental diets: 1)- control diet (normal compound feed for starter piglets without mycotoxin), 2)- AFB1 diet (compound feed contaminated with 320 ppb pure AFB1), 3)- GS diet (compound feed including 8% of grape seed meal), 4)- AFB1+GS diet (compound feed containing 8% of grape seed meal contaminated with 320 ppb AFB1) for 30 days. The results showed that pigs fed AFB1 diet had altered performance (-25.1%), increased the thiobarbituric substances (TBARS) concentration wile reduced total antioxidant capacity and activity of antioxidant enzymes (CAT, SOD and GPx) in plasma and organs. AFB1 produced a dual effect on inflammatory response by increasing the level of pro-inflammatory cytokines in liver and colon and decreasing these cytokines in duodenum. The inclusion of grape seed in the diet of AFB1 intoxicated pigs enhanced the antioxidant enzymes activity, decreased the pro-inflammatory cytokines and TBARS level and ameliorated the growth performance of AFB1-treated animals. These findings suggest that grape waste is a promising feed source in counteracting the harmful effect of aflatoxin B1.
Collapse
Affiliation(s)
- Ionelia Taranu
- Laboratory of Animal Biology, National Institute for Research and Development for Biology and Animal Nutrition, Calea Bucuresti No. 1, Balotesti, Ilfov, 077015, Romania.
| | - Daniela Eliza Marin
- Laboratory of Animal Biology, National Institute for Research and Development for Biology and Animal Nutrition, Calea Bucuresti No. 1, Balotesti, Ilfov, 077015, Romania
| | - Mihai Palade
- Laboratory of Animal Biology, National Institute for Research and Development for Biology and Animal Nutrition, Calea Bucuresti No. 1, Balotesti, Ilfov, 077015, Romania
| | - Gina Cecilia Pistol
- Laboratory of Animal Biology, National Institute for Research and Development for Biology and Animal Nutrition, Calea Bucuresti No. 1, Balotesti, Ilfov, 077015, Romania
| | - Veronica Sanda Chedea
- Laboratory of Animal Biology, National Institute for Research and Development for Biology and Animal Nutrition, Calea Bucuresti No. 1, Balotesti, Ilfov, 077015, Romania
| | - Mihail Alexandru Gras
- Laboratory of Animal Biology, National Institute for Research and Development for Biology and Animal Nutrition, Calea Bucuresti No. 1, Balotesti, Ilfov, 077015, Romania
| | - Catalin Rotar
- Laboratory of Animal Biology, National Institute for Research and Development for Biology and Animal Nutrition, Calea Bucuresti No. 1, Balotesti, Ilfov, 077015, Romania
| |
Collapse
|
24
|
Marchese S, Sorice A, Ariano A, Florio S, Budillon A, Costantini S, Severino L. Evaluation of Aflatoxin M1 Effects on the Metabolomic and Cytokinomic Profiling of a Hepatoblastoma Cell Line. Toxins (Basel) 2018; 10:E436. [PMID: 30373285 PMCID: PMC6265880 DOI: 10.3390/toxins10110436] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022] Open
Abstract
Hepatoblastoma incidence has been associated with different environmental factors even if no data are reported about a correlation between aflatoxin exposure and hepatoblastoma initiation. Considering that hepatoblastoma develops in infants and children and aflatoxin M1 (AFM1), the aflatoxin B1 (AFB1) hydroxylated metabolite, can be present in mothers' milk and in marketed milk products, in this study we decided to test the effects of AFM1 on a hepatoblastoma cell line (HepG2). Firstly, we evaluated the effects of AFM1 on the cell viability, apoptosis, cell cycle, and metabolomic and cytokinomic profile of HepG2 cells after treatment. AFM1 induced: (1) a decrease of HepG2 cell viability, reaching IC50 at 9 µM; (2) the blocking of the cell cycle in the G0/G1 phase; (3) the decrease of formiate levels and incremented level of some amino acids and metabolites in HepG2 cells after treatment; and (4) the increase of the concentration of three pro-inflammatory cytokines, IL-6, IL-8, and TNF-α, and the decrease of the anti-inflammatory interleukin, IL-4. Our results show that AFM1 inhibited the growth of HepG2 cells, inducing both a modulation of the lipidic, glycolytic, and amino acid metabolism and an increase of the inflammatory status of these cells.
Collapse
Affiliation(s)
- Silvia Marchese
- Unità di Farmacologia e Tossicologia-Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli "Federico II", 80138 Napoli, Italy.
| | - Angela Sorice
- Unità di Farmacologia Sperimentale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy.
| | - Andrea Ariano
- Unità di Farmacologia e Tossicologia-Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli "Federico II", 80138 Napoli, Italy.
| | - Salvatore Florio
- Unità di Farmacologia e Tossicologia-Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli "Federico II", 80138 Napoli, Italy.
| | - Alfredo Budillon
- Unità di Farmacologia Sperimentale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy.
| | - Susan Costantini
- Unità di Farmacologia Sperimentale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy.
| | - Lorella Severino
- Unità di Farmacologia e Tossicologia-Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli "Federico II", 80138 Napoli, Italy.
| |
Collapse
|