1
|
Hirozawa MT, Ono MA, de Souza Suguiura IM, Garcia S, Bordini JG, Amador IR, Hirooka EY, Ono EYS. Limosilactobacillus reuteri as sustainable biological control agent against toxigenic Fusarium verticillioides. Braz J Microbiol 2023; 54:2219-2226. [PMID: 37531006 PMCID: PMC10484862 DOI: 10.1007/s42770-023-01081-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 07/23/2023] [Indexed: 08/03/2023] Open
Abstract
Corn contamination with Fusarium verticillioides (Sacc.) Nirenberg is a worldwide problem that affects yield and grain quality resulting in severe economic losses and implications for food safety. Control of F. verticillioides is a challenge, but lactic acid bacteria (LAB) has high potential as a biological control agent. In this study, the antifungal effect of Limosilactobacillus reuteri (formerly Lactobacillus reuteri) LR-92 against F. verticillioides 97L was investigated. Cell-free supernatant (CFS) from L. reuteri showed concentration-dependent fungicidal and fungistatic activity against F. verticillioides 97L. The antifungal compounds from CFS showed heat stability and pH dependence, and antifungal activity was not affected by treatment with proteolytic enzymes. High-performance liquid chromatography analysis indicated that L. reuteri LR-92 produces lactic and acetic acids. After liquid-liquid extraction, electrospray ionization mass spectrometry analysis of the active ethyl acetate fraction containing antifungal compounds revealed the production of 3-phenyllactic acid, cyclo-(L-Pro-L-Leu), cyclo-(L-Pro-L-Phe), and cyclo-(L-Phe-trans-4-OH-L-Pro). L. reuteri LR-92 has potential as a biocontrol agent for F. verticillioides and contributes to food safety.
Collapse
Affiliation(s)
- Melissa Tiemi Hirozawa
- State University of Londrina, Department of Biochemistry and Biotechnology, P.O. Box 10, 011, 86057-970, Londrina, Paraná, Brazil
| | - Mario Augusto Ono
- State University of Londrina, Department of Pathological Sciences, P.O. Box 10, 011, 86057-970, Londrina, Paraná, Brazil
| | | | - Sandra Garcia
- State University of Londrina, Department of Food Science and Technology, P.O. Box 10, 011, 86057-970, Londrina, Paraná, Brazil
| | - Jaqueline Gozzi Bordini
- State University of Londrina, Department of Biochemistry and Biotechnology, P.O. Box 10, 011, 86057-970, Londrina, Paraná, Brazil
| | - Ismael Rodrigues Amador
- State University of Londrina, Department of Biochemistry and Biotechnology, P.O. Box 10, 011, 86057-970, Londrina, Paraná, Brazil
| | - Elisa Yoko Hirooka
- State University of Londrina, Department of Food Science and Technology, P.O. Box 10, 011, 86057-970, Londrina, Paraná, Brazil
| | - Elisabete Yurie Sataque Ono
- State University of Londrina, Department of Biochemistry and Biotechnology, P.O. Box 10, 011, 86057-970, Londrina, Paraná, Brazil.
| |
Collapse
|
2
|
Yan B, Fung K, Ye S, Lai PM, Wei YX, Sze KH, Yang D, Gao P, Kao RYT. Linoleic acid metabolism activation in macrophages promotes the clearing of intracellular Staphylococcus aureus. Chem Sci 2022; 13:12445-12460. [PMID: 36382278 PMCID: PMC9629105 DOI: 10.1039/d2sc04307f] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/05/2022] [Indexed: 09/02/2023] Open
Abstract
Multidrug-resistant bacterial pathogens pose an increasing threat to human health. Certain bacteria, such as Staphylococcus aureus, are able to survive within professional phagocytes to escape the bactericidal effects of antibiotics and evade killing by immune cells, potentially leading to chronic or persistent infections. By investigating the macrophage response to S. aureus infection, we may devise a strategy to prime the innate immune system to eliminate the infected bacteria. Here we applied untargeted tandem mass spectrometry to characterize the lipidome alteration in S. aureus infected J774A.1 macrophage cells at multiple time points. Linoleic acid (LA) metabolism and sphingolipid metabolism pathways were found to be two major perturbed pathways upon S. aureus infection. The subsequent validation has shown that sphingolipid metabolism suppression impaired macrophage phagocytosis and enhanced intracellular bacteria survival. Meanwhile LA metabolism activation significantly reduced intracellular S. aureus survival without affecting the phagocytic capacity of the macrophage. Furthermore, exogenous LA treatment also exhibited significant bacterial load reduction in multiple organs in a mouse bacteremia model. Two mechanisms are proposed to be involved in this progress: exogenous LA supplement increases downstream metabolites that partially contribute to LA's capacity of intracellular bacteria-killing and LA induces intracellular reactive oxygen species (ROS) generation through an electron transport chain pathway in multiple immune cell lines, which further increases the capacity of killing intracellular bacteria. Collectively, our findings not only have characterized specific lipid pathways associated with the function of macrophages but also demonstrated that exogenous LA addition may activate lipid modulator-mediated innate immunity as a potential therapy for bacterial infections.
Collapse
Affiliation(s)
- Bingpeng Yan
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong 21 Sassoon Road Pokfulam Hong Kong China
| | - Kingchun Fung
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong 21 Sassoon Road Pokfulam Hong Kong China
| | - Sen Ye
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong Pokfulam Road Hong Kong P. R. China
| | - Pok-Man Lai
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong 21 Sassoon Road Pokfulam Hong Kong China
| | - Yuan Xin Wei
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong 21 Sassoon Road Pokfulam Hong Kong China
| | - Kong-Hung Sze
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong 21 Sassoon Road Pokfulam Hong Kong China
| | - Dan Yang
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong Pokfulam Road Hong Kong P. R. China
- Laboratory of Chemical Biology and Molecular Medicine, School of Life Sciences, Westlake University Hangzhou Zhejiang P. R. China
| | - Peng Gao
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong 21 Sassoon Road Pokfulam Hong Kong China
| | - Richard Yi-Tsun Kao
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong 21 Sassoon Road Pokfulam Hong Kong China
| |
Collapse
|
3
|
Li T, Huang S, Wang J, Yin P, Liu H, Sun C. Alginate oligosaccharides protect against fumonisin B1-induced intestinal damage via promoting gut microbiota homeostasis. Food Res Int 2022; 152:110927. [PMID: 35181098 DOI: 10.1016/j.foodres.2021.110927] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 12/10/2021] [Accepted: 12/20/2021] [Indexed: 11/26/2022]
Abstract
Fumonisin B1 (FB1), one of the most common mycotoxins contaminating feed and food, has been shown to induce intestinal barrier degradation. However, its role on gut microbiota in this process is still unclear. Alginate oligosaccharides (AOS) have been reported to exert their anti-inflammatory and anti-apoptotic function partially via modulation the gut microbiota. However, little is known about the beneficial effect of AOS on gut microbiota upon FB1 exposure. Results show that FB1 degraded intestinal epithelial barrier function as evidenced by increased pathological epithelial cell shedding, reduced the number of goblet cells, and promoted intestinal cell apoptosis. Markedly, FB1 disturbed the cecal and fecal microbiota composition. FB1 increased the level of Lactobacillus and decreased the relative abundance of beneficial microbes. FB1 largely inhibited the production of short chain fatty acids (SCFAs). AOS greatly ameliorated FB1-induced intestinal damage, inflammation, and oxidative stress (eg., T-SOD and MDA). AOS alleviated gut microbial dysbiosis by promoting the growth of beneficial microbes such as Roseburia, Bifidobacterium, and Akkermansia, and increasing SCFAs production upon FB1 exposure. Moreover, the correlation analysis showed that FB1- and AOS-treated gut microbiota alteration is closely associated with the change of intestinal phenotype. We have thus provided a novel insight into the protective role of AOS on FB1-induced gut microbial dysbiosis.
Collapse
Affiliation(s)
- Tiantian Li
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Jun Wang
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Peng Yin
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Hujun Liu
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Changpo Sun
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China; Standards and Quality Center of National Food and Strategic Reserves Administration, China.
| |
Collapse
|
4
|
Wang Y, Xu Y, Ju JQ, Liu JC, Sun SC. Fumonisin B1 exposure deteriorates oocyte quality by inducing organelle dysfunction and DNA damage in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 223:112598. [PMID: 34388657 DOI: 10.1016/j.ecoenv.2021.112598] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/29/2021] [Accepted: 08/01/2021] [Indexed: 06/13/2023]
Abstract
Oocyte quality is critical for fertilization and early embryo development. Fumonisin B1 (FB1) is a Fusarium mycotoxin and it is commonly found in contaminated food and feedstuff, posing a potential health hazard to both animals and human. FB1 is reported to have hepatotoxicity, neurotoxicity, nephrotoxicity, immunotoxicity and embryotoxicity. However, the effects of FB1 on mouse oocyte quality are still unknown. Here, we explored the toxic effects and potential mechanisms of FB1 on oocyte maturation quality in mice. FB1 exposure inhibited the first polar body extrusion at concentrations of 30 μM and 50 μM, which further induced oocyte meiotic arrest. Besides, disrupted spindle structure was found in oocytes after FB1 exposure. Our results also showed that FB1 exposure impaired mitochondria dysfunction, which further induced oxidative stress and early apoptosis. In addition, we reported that FB1 exposure induced the accumulation of lysosome and occurrence of autophagy. Aberrant ER distribution and ER stress were also found in FB1-exposed oocytes. Moreover, DNA damage was also observed. These results together suggested that FB1 exposure affected oocyte quality by destroying spindle structure, leading to mitochondria, lysosome and ER dysfunction, which further induced oxidative stress, apoptosis, autophagy and DNA damage in mouse oocytes.
Collapse
Affiliation(s)
- Yue Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yao Xu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jia-Qian Ju
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jing-Cai Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
5
|
Research Progress on Fumonisin B1 Contamination and Toxicity: A Review. Molecules 2021; 26:molecules26175238. [PMID: 34500671 PMCID: PMC8434385 DOI: 10.3390/molecules26175238] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 12/19/2022] Open
Abstract
Fumonisin B1 (FB1), belonging to the member of fumonisins, is one of the most toxic mycotoxins produced mainly by Fusarium proliferatum and Fusarium verticillioide. FB1 has caused extensive contamination worldwide, mainly in corn, rice, wheat, and their products, while it also poses a health risk and is toxic to animals and human. It has been shown to cause oxidative stress, endoplasmic reticulum stress, cellular autophagy, and apoptosis. This review focuses on the current stage of FB1 contamination, its toxic effects of acute toxicity, immunotoxicity, organ toxicity, and reproductive toxicity on animals and humans. The potential toxic mechanisms of FB1 are discussed. One of the main aims of the work is to provide a reliable reference strategy for understanding the occurrence and toxicity of FB1.
Collapse
|
6
|
Arumugam T, Ghazi T, Chuturgoon A. Fumonisin B 1 Epigenetically Regulates PTEN Expression and Modulates DNA Damage Checkpoint Regulation in HepG2 Liver Cells. Toxins (Basel) 2020; 12:toxins12100625. [PMID: 33007920 PMCID: PMC7601513 DOI: 10.3390/toxins12100625] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Fumonisin B1 (FB1), a Fusarium-produced mycotoxin, is found in various foods and feeds. It is a well-known liver carcinogen in experimental animals; however, its role in genotoxicity is controversial. The current study investigated FB1-triggered changes in the epigenetic regulation of PTEN and determined its effect on DNA damage checkpoint regulation in human liver hepatoma G2 (HepG2) cells. Following treatment with FB1 (IC50: 200 µM; 24 h), the expression of miR-30c, KDM5B, PTEN, H3K4me3, PI3K, AKT, p-ser473-AKT, CHK1, and p-ser280-CHK1 was measured using qPCR and/or Western blot. H3K4me3 enrichment at the PTEN promoter region was assayed via a ChIP assay and DNA damage was determined using an ELISA. FB1 induced oxidative DNA damage. Total KDM5B expression was reduced, which subsequently increased the total H3K4me3 and the enrichment of H3K4me3 at PTEN promoters. Increased H3K4me3 induced an increase in PTEN transcript levels. However, miR-30c inhibited PTEN translation. Thus, PI3K/AKT signaling was activated, inhibiting CHK1 activity via phosphorylation of its serine 280 residue preventing the repair of damaged DNA. In conclusion, FB1 epigenetically modulates the PTEN/PI3K/AKT signaling cascade, preventing DNA damage checkpoint regulation, and induces significant DNA damage.
Collapse
|
7
|
Toxicological effects of fumonisin B1 in combination with other Fusarium toxins. Food Chem Toxicol 2018; 121:483-494. [DOI: 10.1016/j.fct.2018.09.043] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 12/29/2022]
|
8
|
The molecular mechanism of cell cycle arrest in the Bursa of Fabricius in chick exposed to Aflatoxin B 1. Sci Rep 2018; 8:1770. [PMID: 29379099 PMCID: PMC5789014 DOI: 10.1038/s41598-018-20164-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 01/15/2018] [Indexed: 12/12/2022] Open
Abstract
Aflatoxin B1 shows potent hepatotoxic, carcinogenic, genotoxic, immunotoxic potential in humans and many species of animals. The aim of this study was to clarify the underlying mechanism of G0G1 phase and G2M phase arrest of cell cycle in the bursa of Fabricius in broilers exposed to dietary AFB1. 144 one-day-old healthy Cobb broilers were randomly divided into two groups and fed on control diet and 0.6 mg·Kg−1 AFB1 diet for 3 weeks. Histological observation showed that AFB1 induced the increase of nuclear debris and vacuoles in lymphoid follicle of BF. Results of flow cytometry studies showed that bursal cells arrested in G2M phase at 7 days of age and blocked in G0G1 phase at 14 and 21 days of age following exposure to AFB1. The qRT-PCR analysis indicated that cell cycle arrested in G2M phase via ATM-Chk2-cdc25-cyclin B/cdc2 pathway, and blocked in G0G1 phase through ATM-Chk2-cdc25-cyclin D/CDK6 pathway and ATM-Chk2-p21-cyclin D/CDK6 route. In a word, our results provided new insights that AFB1 diet induced G2M and G0G1 phase blockage of BF cells in different periods, and different pathways were activated in different arrested cell cycle phase.
Collapse
|
9
|
Khan RB, Phulukdaree A, Chuturgoon AA. Concentration-dependent effect of fumonisin B1 on apoptosis in oesophageal cancer cells. Hum Exp Toxicol 2017; 37:762-771. [DOI: 10.1177/0960327117735570] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The geographical distribution of oesophageal cancer is linked to the exposure of fumonisin B1 (FB1), a mycotoxin produced by fungi that contaminates staple food worldwide. Non-genotoxic carcinogens like FB1 disturb homeostasis through increased cell proliferation or suppression of apoptosis. This study investigated the involvement of FB1 (0–20 μM) in spindle-shaped N-cadherin (+) CD45 (−) osteoblastic (SNO) cell death. Cell viability and death were assessed using the MTS and Annexin V-Fluos assays, respectively. Caspase activities were determined luminometrically and the comet assay assessed DNA damage. Induction of oxoguanine glycosylase 1 (OGG1) was measured using quantitative Polymerase Chain Reaction (qPCR), while cleaved poly (ADP-ribose) polymerase 1 (PARP-1) and Bax were determined by western blotting. Cell viability and PARP-1 cleavage were not affected by 1.25 μM FB1, but phosphatidylserine externalization, Bax protein expression, caspase activity, comet tail length and OGG1 transcripts were increased. The reduced cell viability in 10 μM FB1-treated cells was accompanied by corresponding increases in externalized phosphatidylserine, Bax, caspase-3/7 activity and cleaved PARP-1. The OGG1 transcripts were not significantly increased, but comet tails were increased. Bax, caspase-3/7 activities and cleaved PARP-1 were inhibited at 20 μM FB1. In addition, the OGG1 transcript levels were decreased ( p < 0.0001) along with comet lengths ( p < 0.0001). This study showed that FB1-induced apoptosis in SNO cells may be caspase-dependent or caspase-independent; the pathway used depends on the exposure concentration.
Collapse
Affiliation(s)
- RB Khan
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - A Phulukdaree
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - AA Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
10
|
Mary VS, Arias SL, Otaiza SN, Velez PA, Rubinstein HR, Theumer MG. The aflatoxin B 1 -fumonisin B 1 toxicity in BRL-3A hepatocytes is associated to induction of cytochrome P450 activity and arachidonic acid metabolism. ENVIRONMENTAL TOXICOLOGY 2017; 32:1711-1724. [PMID: 28181396 DOI: 10.1002/tox.22395] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 01/12/2017] [Accepted: 01/15/2017] [Indexed: 06/06/2023]
Abstract
Human oral exposure to aflatoxin B1 (AFB1 ) and fumonisin B1 (FB1 ) is associated with increased hepatocellular carcinoma. Although evidence suggested interactive AFB1 -FB1 hepatotoxicity, the underlying mechanisms remain mostly unidentified. This work was aimed at evaluating the possible AFB1 -FB1 interplay to induce genetic and cell cycle toxicities in BRL-3A rat hepatocytes, reactive oxygen species (ROS) involvement, and the AFB1 metabolizing pathways cytochrome P450 (CYP) and arachidonic acid (ArAc) metabolism as ROS contributors. Flow cytometry of stained BRL-3A hepatocytes was used to study the cell cycle (propidium iodide), ROS intracellular production (DCFH-DA, HE, DAF-2 DA), and phospholipase A activity (staining with bis-BODIPY FL C11-PC). The CYP1A activity was assessed by the 7-ethoxyresorufin-O-deethylase (EROD) assay. Despite a 48-h exposure to FB1 (30 μM) not being genotoxic, the AFB1 (20 μM)-induced micronucleus frequency was overcome by the AFB1 -FB1 mixture (MIX), presumably showing toxin interaction. The mycotoxins blocked G1/S-phase, but only MIX caused cell death. Overall, the oxidative stress led these alterations as the pretreatment with N-acetyl-l-cysteine reduced such toxic effects. While AFB1 had a major input to the MIX pro-oxidant activity, with CYP and ArAc metabolism being ROS contributors, these pathways were not involved in the FB1 -elicited weak oxidative stress. The MIX-induced micronucleus frequency in N-acetyl-l-cysteine pretreated cells was greater than that caused by AFB1 without antioxidants, suggesting enhanced AFB1 direct genotoxicity probably owing to the higher CYP activity and ArAc metabolism found in MIX. The metabolic pathways modulation by AFB1 -FB1 mixtures could raise its hepatocarcinogenic properties.
Collapse
Affiliation(s)
- Verónica S Mary
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), UNC, CONICET, Córdoba, X5000HUA, Argentina
| | - Silvina L Arias
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), UNC, CONICET, Córdoba, X5000HUA, Argentina
| | - Santiago N Otaiza
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), UNC, CONICET, Córdoba, X5000HUA, Argentina
| | - Pilar A Velez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), UNC, CONICET, Córdoba, X5000HUA, Argentina
| | - Héctor R Rubinstein
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), UNC, CONICET, Córdoba, X5000HUA, Argentina
| | - Martín G Theumer
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), UNC, CONICET, Córdoba, X5000HUA, Argentina
| |
Collapse
|
11
|
Zhang H, Zhang L, Diao X, Li N, Liu C. Toxicity of the mycotoxin fumonisin B 1 on the insect Sf9 cell line. Toxicon 2017; 129:20-27. [DOI: 10.1016/j.toxicon.2017.01.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 01/21/2017] [Accepted: 01/23/2017] [Indexed: 12/19/2022]
|
12
|
Sancak D, Ozden S. Global histone modifications in Fumonisin B1 exposure in rat kidney epithelial cells. Toxicol In Vitro 2015. [PMID: 26208285 DOI: 10.1016/j.tiv.2015.07.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Fumonisin B1 (FB1) is a Fusarium mycotoxin frequently occurring in maize-based food and feed. Although the effects of FB1 on sphingolipid metabolism are clear, little is known about early molecular changes associated with FB1 carcinogenicity. It has been shown that FB1 disrupts DNA methylation and chromatin modifications in HepG2 cells. We investigated dose- and time-dependent effects of FB1 in global histone modifications such as histone H3 lysine 9 di-, trimethylation (H3K9me2/me3), histone H3 lysine 4 trimethylation (H3K4me3), histone H4 lysine 20 trimethylation (H4K20me3), histone H3 lysine 9 acetylation (H3K9ac) and the enzymes involved in these mechanisms in rat kidney epithelial cells (NRK-52E). The increased levels of global H3K9me2/me3 were observed in FB1 treated cells, while the global levels of H4K20me3 and H3K9ac were decreased. FB1 caused some changes on the activities of H3K9 histone methyltransferase (HMT) and histone acetyltransferase (HAT) at high concentrations in NRK-52E cells. Further, the effects of trichostatin A (TSA), a histone deacetylase inhibitor, were investigated in NRK-52E cells. TSA was found to cause an increase on H3K9ac levels as expected. In this study we suggest that FB1 may disrupt epigenetic events by altering global histone modifications, introducing a novel aspect on the potential mechanism of FB1 carcinogenesis.
Collapse
Affiliation(s)
- Duygu Sancak
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University, 34116 Beyazit, Istanbul, Turkey
| | - Sibel Ozden
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Istanbul University, 34116 Beyazit, Istanbul, Turkey.
| |
Collapse
|
13
|
Wang SK, Wang TT, Huang GL, Shi RF, Yang LG, Sun GJ. Stimulation of the proliferation of human normal esophageal epithelial cells by fumonisin B 1 and its mechanism. Exp Ther Med 2013; 7:55-60. [PMID: 24348764 PMCID: PMC3860871 DOI: 10.3892/etm.2013.1364] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 10/16/2013] [Indexed: 11/06/2022] Open
Abstract
Previous epidemiological studies have demonstrated a correlation between fumonisin B1 (FB1) and human esophageal cancer in China, Iran and South Africa. The purpose of this study was to investigate the effects of FB1 on the proliferation, cell-cycle and apoptosis of normal human esophageal epithelial cells (HEECs) and to explore the molecular mechanisms of these effects. The proliferation of HEECs treated with FB1 was assessed using a colorimetric assay, while analyses of the cell cycle and apoptosis were performed using flow cytometry and the measurement of the protein expressions of genes associated with the cell cycle was conducted using western blotting. The results showed that FB1 stimulated the proliferation of HEECs, decreased the percentage of cells in the G0/G1 phase and reduced apoptosis. The western blotting results showed that FB1 significantly increased the protein expression of cyclin D1 and significantly decreased the protein expression of cyclin E, p21 and p27. The results indicated that FB1 stimulated the proliferation of HEECs by affecting the cell cycle and apoptosis. This mechanism was associated with changes in cyclin D1, cyclin E, p21 and p27 expression.
Collapse
Affiliation(s)
- Shao-Kang Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China ; Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Ting-Ting Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China ; Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Gui-Ling Huang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China ; Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Ruo-Fu Shi
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China ; Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Li-Gang Yang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China ; Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Gui-Ju Sun
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China ; Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|