1
|
Lee Y, Cha D, Choi S, Cho J, Lee S. Toxic effects of sterigmatocystin on porcine oocyte maturation and subsequent embryo development. Toxicol Lett 2025; 407:63-72. [PMID: 40154669 DOI: 10.1016/j.toxlet.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/22/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Sterigmatocystin (STE), a precursor of aflatoxin B1, is one of the mycotoxins that easily contaminates feed. Although previous studies have suggested the toxic effects of aflatoxin B1 on oocyte maturation, little attention has been given to the effects of STE. Therefore, we investigated the effects of STE on porcine oocyte maturation. In this study, porcine oocytes were subjected to in vitro maturation supplemented with various concentrations of STE (0, 5, 10, and 25 μM). The results showed that the cumulus cell expansion indexes of all STE-treated groups were significantly decreased compared to the control group, with 10 μM significantly decreasing the transcript expression of cumulus expansion-related genes. Regarding nuclear maturation, metaphase II rates in all STE-treated groups were significantly lower than in the control group, with 10 μM significantly decreasing the transcript expression of oocyte competence-, mitogen-activated protein kinase-, and maturation-promoting factor-related genes. While cleavage rates showed no significant differences, the blastocyst formation rates significantly declined in groups treated with more than 10 μM of STE. Based on these findings, the 10 μM STE group was selected for subsequent experiments. STE supplementation significantly increased reactive oxygen species levels and decreased glutathione levels in oocytes compared to the control group. Furthermore, STE significantly decreased mitochondrial quantity and membrane potential, while increasing the percentage of γ-H2AX-positive oocytes. The number of LC3-positive dots and Annexin-V-positive oocytes was also significantly higher in the STE-treated group than in the control group. In conclusion, STE impairs porcine oocyte maturation and subsequent embryo development by inducing oxidative stress, mitochondrial dysfunction, DNA damage, excessive autophagy, and early apoptosis.
Collapse
Affiliation(s)
- Yumin Lee
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Dabin Cha
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Seunghyun Choi
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jongki Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Sanghoon Lee
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
2
|
Wang Y, Jin Y, Wang Y, Li Y, Wang X, Li Z, Zhou J. Sow reproductive disorders: a key issue affecting the pig industry. Front Vet Sci 2025; 12:1535719. [PMID: 40110434 PMCID: PMC11920173 DOI: 10.3389/fvets.2025.1535719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025] Open
Abstract
Pig farming is essential to global agricultural economies and food security. However, reproductive disorders in sows significantly impact the economic viability and sustainability of the pig industry. These disorders often result from complex interactions between pathogenic and non-pathogenic factors. Preventing abortions is typically more cost-effective than managing and treating them, particularly in intensive pig farming system. This highlights the importance of comprehensively understanding the underlying causes of abortion in sows. This review explores the factors contributing to sow reproductive disorders, including both non-infectious factors (environmental conditions and management practices) and infectious factors (viruses, bacteria, and parasites). We also outline preventive and control strategies, alongside integrated management approaches, by analyzing the underlying causes and pathogenic mechanisms of pregnancy disorders. Overall, implementing the "One Health" concept in large-scale farming provides an effective strategy to reduce the incidence of sow abortion rate, ensure stable livestock production, and maintain a reliable global pork supply.
Collapse
Affiliation(s)
- Yihan Wang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, College of Veterinary Medicine, Lanzhou University, Lanzhou, China
| | - Youshun Jin
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, College of Veterinary Medicine, Lanzhou University, Lanzhou, China
| | - Yanyan Wang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, College of Veterinary Medicine, Lanzhou University, Lanzhou, China
| | - Yunhui Li
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, College of Veterinary Medicine, Lanzhou University, Lanzhou, China
- Animal Pathology Laboratory, College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Xiaoxue Wang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, College of Veterinary Medicine, Lanzhou University, Lanzhou, China
- College of Life Sciences, Yulin University, Yulin, China
| | - Zhaocai Li
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, College of Veterinary Medicine, Lanzhou University, Lanzhou, China
| | - Jizhang Zhou
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, College of Veterinary Medicine, Lanzhou University, Lanzhou, China
| |
Collapse
|
3
|
Lu PS, Sun SC. Mycotoxin toxicity and its alleviation strategy on female mammalian reproduction and fertility. J Adv Res 2025:S2090-1232(25)00041-4. [PMID: 39814223 DOI: 10.1016/j.jare.2025.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/23/2024] [Accepted: 01/12/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Mycotoxin, a secondary metabolite of fungus, found worldwide and concerning in crops and food, causes multiple acute and chronic toxicities. Its toxic profile includes hepatotoxicity, carcinogenicity, teratogenicity, estrogenicity, immunotoxicity, and neurotoxicity, leading to deleterious impact on human and animal health. Emerging evidence suggests that it adversely affects perinatal health and progeny by its ability to cross placental barriers. AIM OF REVIEW Due to its wide occurrence and potential toxicity on reproductive health, it is essential to understand the mechanisms of mycotoxin-related reproductive toxicity. This review summarizes the toxicities and mechanisms of mycotoxin on maternal and offspring reproduction among mammalian species. Approaches for effective mycotoxin alleviation are also discussed, providing strategies against mycotoxin contamination. KEY SCIENTIFIC CONCEPTS OF REVIEW The profound mycotoxin toxicities in female mammalian reproduction affect follicle assembly, embryo development, and fetus growth, thereby decreasing offspring fertility. Factors from endocrine system such as hypothalamic-pituitary-gonadal axis and gut-ovarian axis, placenta ABC transporters, organelle and cytoskeleton dynamics, cell cycle control, genomic stability, and redox homeostasis are found to be closely related to mycotoxin toxicities. Approaches from physical, chemical, biological, and supplementation of natural antioxidants are discussed for the mycotoxin elimination, while their applications are not widespread. Available ways for mycotoxin and its toxicities alleviation need further study. Since a species-, time-, and dose-specific response might exist in mycotoxin toxicities, more consideration should be given to the protocols for mycotoxin toxicity studies, such as experimental animal models, exposure duration, and dosage. Specific mechanism for mycotoxin, especially form a molecular biology perspective, could be investigated with multi-omics technologies and advanced imaging techniques. Mass spectrometry with algorithms may provide more accurate exposure assessments, and it may be further helpful to identify the high-risk individuals in the future.
Collapse
Affiliation(s)
- Ping-Shuang Lu
- 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; Key Laboratory of Research On Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Reproductive Medicine of Guangxi Medical and Health Key Discipline Construction Project, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China.
| |
Collapse
|
4
|
Kalo D, Yaacobi-Artzi S, Manovich S, Michaelov A, Komsky-Elbaz A, Roth Z. Environmental Stress-Induced Alterations in Embryo Developmental Morphokinetics. J Xenobiot 2024; 14:1613-1637. [PMID: 39449428 PMCID: PMC11503402 DOI: 10.3390/jox14040087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/13/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
The association between embryo morphokinetics and its developmental competence is well documented. For instance, early cleaved embryos are more competent in developing to blastocysts, whereas the proportion of abnormally cleaved embryos that further developed to blastocysts is low. Numerous factors, such as the parental age, lifestyle, health, and smoking habits have been reported to affect the embryo morphokinetics and, consequently, its development. However, less is known about the effect of environmental stressors on embryo morphokinetics. The current review discusses the effect of the most concerning environmental stressors on embryo morphokinetics. These stresses include heat stress and human-made chemicals such as phthalates (e.g., bis-(2-ethylhexyl phthalate, dibutyl phthalate, dimethyl phthalate, and their primary metabolites), herbicides (e.g., diaminochlorotriazine, the primary metabolite of atrazine), pharmaceutical compounds (e.g., carbamazepine, nocodazole) and pro-oxidant agents (cumene hydroperoxide, Triton X-100), as well as naturally occurring toxins such as mycotoxin (e.g., aflatoxin B1 and its metabolite, and ochratoxin A). In addition, this review discusses the effect of ionizing or non-ionizing radiation and viral infections (e.g., SARS-CoV-2, papillomavirus). Finally, it points out some potential mechanisms that underlie the impairment of embryo morphokinetics, and it suggests protective compounds, mainly the supplementation of antioxidants to improve the morphokinetics, and consequently, the embryo developmental competence.
Collapse
Affiliation(s)
| | | | | | | | | | - Zvi Roth
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 7610001, Israel; (D.K.)
| |
Collapse
|
5
|
Jalili C, Ranjbar Shamsi R, Amiri B, Kakebaraie S, Jalili F, Nasta TZ. Genotoxic and cytotoxic effects of aflatoxin on the reproductive system: Focus on cell cycle dynamics and apoptosis in testicular tissue. Toxicology 2024; 504:153773. [PMID: 38484789 DOI: 10.1016/j.tox.2024.153773] [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: 01/11/2024] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 03/24/2024]
Abstract
Aflatoxins (AFs) are inevitable environmental contaminants that are detrimental to human and animal health. AFs interfere with metabolic processes, metabolizing into different hydroxylated derivatives in the liver, as well as mechanistically induce ROS accumulation, S-phase arrest, DNA damage, and cell apoptosis. Chronic consumption of aflatoxin-contaminated foods can adversely affect the male reproductive system, cause testicular damage, prevent testosterone synthesis, decline sperm quality, and cause infertility. Oxidative stress is the fundamental pathogenesis of aflatoxin-induced reproductive toxicity. The overproduction of reactive oxygen substances can cause testicular failure and disturb the process of spermatogenesis. Mitochondria are susceptible to being impaired by oxidative stress, and its damage is associated with infertility. AFs also disturb the process of spermatogenesis by disrupting the regulation of genes related to the progression of the cell cycle such as cyclins and inducing genes related to apoptosis, thereby weakening fertility and negatively affecting the testicular endocrine potential by suppressing androgen synthesis. Additionally, AFs downregulate ERα expression, potentially negatively impacting spermatogenesis by enhancing the apoptotic mechanism. In this review, we provide new insights into the genotoxic and cytotoxic effects of AFB1 on the male reproductive system with a focus on the cell cycle and apoptosis destruction of testicular tissue.
Collapse
Affiliation(s)
- Cyrus Jalili
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Islamic Republic of Iran
| | - Rahele Ranjbar Shamsi
- Department of Clinical Biochemistry, Faculty of Veterinary Medicine, Tabriz, Islamic Republic of Iran
| | - Bita Amiri
- Department of Chemistry and Biochemistry, University of Texas at Arlington, TX, USA
| | - Seyran Kakebaraie
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Islamic Republic of Iran
| | - Faramarz Jalili
- School of Health Administration, Faculty of Health, Dalhousie University, Halifax, NS, Canada
| | - Touraj Zamir Nasta
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Islamic Republic of Iran.
| |
Collapse
|
6
|
Yaacobi-Artzi S, Kalo D, Roth Z. Effect of the aflatoxins B1 and M1 on bovine oocyte developmental competence and embryo morphokinetics. Reprod Toxicol 2023; 120:108437. [PMID: 37422138 DOI: 10.1016/j.reprotox.2023.108437] [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: 03/19/2023] [Revised: 06/21/2023] [Accepted: 07/04/2023] [Indexed: 07/10/2023]
Abstract
Aflatoxins are considered as reproductive toxins for mammalian species. Here, we studied the effect of aflatoxin B1 (AFB1) and its metabolite aflatoxin M1 (AFM1) on the development and morphokinetics of bovine embryos. Cumulus oocyte complexes (COCs) were matured with AFB1 (0.032, 0.32, 3.2, 32 µM) or AFM1 (0.015, 0.15, 1.5, 15, 60 nM), then fertilized and the putative zygotes were cultured in an incubator equipped with a time-lapse system. Exposing COCs to 32 µM AFB1 or 60 nM AFM1 reduced the cleavage rate, whereas exposing them to 3.2 or 32 µM AFB1 further reduced the blastocyst formation. A delay was recorded for the first and second cleavages in a dose-dependent manner for both AFB1- and AFM1-treated oocytes. A delay was recorded in the third cleavage in the AFM1-treated group. To explore potential mechanisms, subgroups of COCs were examined for nuclear and cytoplasmic maturation (n = 225; DAPI and FITC-PNA, respectively), and mitochondrial function was examined in a stage-dependent manner. COCs were examined for their oxygen consumption rates (n = 875; Seahorse XFp analyzer) at the end of maturation, MII-stage oocytes were examined for their mitochondrial membrane potential (n = 407; JC1), and putative zygotes were examined using a fluorescent time-lapse system (n = 279; IncuCyte). Exposing COCs to AFB1 (3.2 or 32 µM) impaired oocyte nuclear and cytoplasmic maturation and increased mitochondrial membrane potential in the putative zygotes. These alterations were associated with changes in the expression of mt-ND2 (32 µM AFB1) and STAT3 (all AFM1 concentrations) genes in the blastocyst stage, suggesting a carryover effect from the oocyte to the developing embryos.
Collapse
Affiliation(s)
- Shira Yaacobi-Artzi
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, the Hebrew University, Rehovot 7610000, Israel
| | - Dorit Kalo
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, the Hebrew University, Rehovot 7610000, Israel
| | - Zvi Roth
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, the Hebrew University, Rehovot 7610000, Israel.
| |
Collapse
|
7
|
Huang W, Cao Z, Cui Y, Huo S, Shao B, Song M, Cheng P, Li Y. Lycopene ameliorates aflatoxin B 1-induced testicular lesion by attenuating oxidative stress and mitochondrial damage with Nrf2 activation in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 256:114846. [PMID: 37018856 DOI: 10.1016/j.ecoenv.2023.114846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 06/19/2023]
Abstract
Aflatoxin B1 (AFB1) is an extremely hazardous and unavoidable pollutant for cereals and feedstuff. AFB1 can cause testicular lesion, and how to alleviate its testicular toxicity has received much attention in recent years. Lycopene (LYC), a foodborne nutrient derived from red fruits and vegetables, has protective effects against sperm abnormality and testicular lesions. To confirm the beneficial effects and mechanisms of LYC on AFB1-induced testicular lesion, 48 male mice were exposed to 0.75 mg/kg AFB1 or/and 5 mg/kg LYC for consecutive 30 days. Results demonstrated the LYC significantly restored the lesions of testicular microstructure and ultrastructure, and sperm abnormalities in AFB1-exposed mice. Furthermore, LYC effectively attenuated AFB1-induced oxidative stress and mitochondrial damage, including ameliorative mitochondrial structural, and elevated mitochondrial biogenesis for maintaining mitochondrial function. Meanwhile, LYC resisted AFB1-induced mitochondrial-dependent apoptosis. In addition, LYC promoted nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation, and upregulated the Nrf2 signaling pathway. Collectively, our findings demonstrate LYC ameliorates AFB1-induced testicular lesion by attenuating oxidative stress and mitochondrial damage, which is related to the activation of Nrf2.
Collapse
Affiliation(s)
- Wanyue Huang
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| | - Zheng Cao
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yilong Cui
- College of Animal Science and Technology, Inner Mongolia Minzu University, 028000 Tongliao, China
| | - Siming Huo
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Bing Shao
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Miao Song
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Ping Cheng
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei 230036, China
| | - Yanfei Li
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
8
|
Yu W, Peng Y, Peng X, Li Z, Liu C, Yang L, Gao Y, Liang S, Yuan B, Chen C, Kim NH, Jiang H, Zhang J. 6-Gingerol Improves In Vitro Porcine Embryo Development by Reducing Oxidative Stress. Animals (Basel) 2023; 13:ani13081315. [PMID: 37106877 PMCID: PMC10135256 DOI: 10.3390/ani13081315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/01/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
6-Gingerol, the main active ingredient in ginger, exhibits a variety of biological activities, such as antioxidant, anti-inflammatory, and anticancer activities, and can affect cell development. However, the effects of 6-gingerol on mammalian reproductive processes, especially early embryonic development, are unclear. This study explored whether 6-gingerol can be used to improve the quality of in vitro-cultured porcine embryos. The results showed that 5 μM 6-gingerol significantly increased the blastocyst formation rates of porcine early embryos. 6-Gingerol attenuated intracellular reactive oxygen species accumulation and autophagy, increased intracellular glutathione levels, and increased mitochondrial activity. In addition, 6-gingerol upregulated NANOG, SRY-box transcription factor 2, cytochrome c oxidase subunit II, mechanistic target of rapamycin kinase, and RPTOR independent companion of MTOR complex 2 while downregulating Caspase 3, baculoviral IAP repeat containing 5, autophagy related 12, and Beclin 1. Most importantly, 6-gingerol significantly increased the levels of p-extracellular regulated protein kinase 1/2 while reducing the levels of p-c-Jun N-terminal kinase 1/2/3 and p-p38. These results indicate that 6-gingerol can promote the development of porcine early embryos in vitro.
Collapse
Affiliation(s)
- Wenjie Yu
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, China
| | - Yanxia Peng
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, China
| | - Xinyue Peng
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, China
| | - Ze Li
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, China
| | - Chang Liu
- School of Grains, Jilin Business and Technology College, Changchun 130507, China
| | - Liu Yang
- Tongyu Grassland Management Station, Changchun 137200, China
| | - Yan Gao
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, China
| | - Shuang Liang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, China
| | - Bao Yuan
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, China
| | - Chengzhen Chen
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, China
| | - Nam-Hyung Kim
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, China
- Department of Animal Science, Chungbuk National University, Cheongju 361-763, Chungbuk, Republic of Korea
| | - Hao Jiang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, China
- Department of Animal Science, Chungbuk National University, Cheongju 361-763, Chungbuk, Republic of Korea
| | - Jiabao Zhang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun 130062, China
| |
Collapse
|
9
|
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
|
10
|
Zhang J, Hu S, Zhao C, Zhou Y, Zhang L, Liu H, Zhou P, Li S, Fu L, Zheng Z, Xiang Y, Xu X, Ruan J, Li X, Sun L, Cao G, Zhao S, Wang X, Xie S. Genome-Scale CRISPR Knockout Screening Identifies BACH1 as a Key Regulator of Aflatoxin B 1-Induced Oxidative Damage. Antioxidants (Basel) 2022; 11:antiox11091787. [PMID: 36139865 PMCID: PMC9495794 DOI: 10.3390/antiox11091787] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
Aflatoxin B1 (AFB1) is amongst the mycotoxins commonly affecting human and animal health, raising global food safety and control concerns. The mechanisms underlying AFB1 toxicity are poorly understood. Moreover, antidotes against AFB1 are lacking. Genome-wide CRISPR/Cas9 knockout screening in porcine kidney cells identified the transcription factor BTB and CNC homolog 1 (BACH1) as a gene required for AFB1 toxicity. The inhibition of BACH1 expression in porcine kidney cells and human hepatoma cells resulted in increased resistance to AFB1. BACH1 depletion attenuates AFB1-induced oxidative damage via the upregulation of antioxidant genes. Subsequently, virtual structural screening identified the small molecule 1-Piperazineethanol, α-[(1,3-benzodioxol-5-yloxy)methyl] -4-(2-methoxyphenyl) (M2) as an inhibitor of BACH1. M2 and its analogues inhibited AFB1-induced porcine and human cell death in vitro, while M2 administration significantly improved AFB1-induced symptoms of weight loss and liver injury in vivo. These findings demonstrate that BACH1 plays a central role in AFB1-induced oxidative damage by regulating antioxidant gene expression. We also present a potent candidate small-molecule inhibitor in developing novel treatments for AFB1 toxicity.
Collapse
Affiliation(s)
- Jinfu Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Siyi Hu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China;
| | - Changzhi Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Yuan Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Lu Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Hailong Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Peng Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Sheng Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Liangliang Fu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Zhuqing Zheng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Yue Xiang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Xuewen Xu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Jinxue Ruan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
| | - Xinyun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (G.C.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Lvhui Sun
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (G.C.)
| | - Gang Cao
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (G.C.)
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (G.C.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence: (S.Z.); (X.W.); (S.X.)
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China;
- Correspondence: (S.Z.); (X.W.); (S.X.)
| | - Shengsong Xie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; (J.Z.); (C.Z.); (Y.Z.); (L.Z.); (H.L.); (P.Z.); (S.L.); (L.F.); (Z.Z.); (Y.X.); (X.X.); (J.R.); (X.L.)
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangzhou 510642, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China; (L.S.); (G.C.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence: (S.Z.); (X.W.); (S.X.)
| |
Collapse
|
11
|
Li C, Liu X, Wu J, Ji X, Xu Q. Research progress in toxicological effects and mechanism of aflatoxin B 1 toxin. PeerJ 2022; 10:e13850. [PMID: 35945939 PMCID: PMC9357370 DOI: 10.7717/peerj.13850] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/16/2022] [Indexed: 01/18/2023] Open
Abstract
Fungal contamination of animal feed can severely affect the health of farm animals, and result in considerable economic losses. Certain filamentous fungi or molds produce toxic secondary metabolites known as mycotoxins, of which aflatoxins (AFTs) are considered the most critical dietary risk factor for both humans and animals. AFTs are ubiquitous in the environment, soil, and food crops, and aflatoxin B1(AFB1) has been identified by the World Health Organization (WHO) as one of the most potent natural group 1A carcinogen. We reviewed the literature on the toxic effects of AFB1 in humans and animals along with its toxicokinetic properties. The damage induced by AFB1 in cells and tissues is mainly achieved through cell cycle arrest and inhibition of cell proliferation, and the induction of apoptosis, oxidative stress, endoplasmic reticulum (ER) stress and autophagy. In addition, numerous coding genes and non-coding RNAs have been identified that regulate AFB1 toxicity. This review is a summary of the current research on the complexity of AFB1 toxicity, and provides insights into the molecular mechanisms as well as the phenotypic characteristics.
Collapse
Affiliation(s)
- Congcong Li
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China
| | - Xiangdong Liu
- Huazhong Agricultural University, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Wuhan, Hubei, China
| | - Jiao Wu
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China
| | - Xiangbo Ji
- Henan University of Animal Husbandry and Economy, Henan Key Laboratory of Unconventional Feed Resources Innovative Utilization, Zhengzhou, Henan, China
| | - Qiuliang Xu
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China
| |
Collapse
|
12
|
Effect of Fumonisin B1 on Proliferation and Apoptosis of Intestinal Porcine Epithelial Cells. Toxins (Basel) 2022; 14:toxins14070471. [PMID: 35878209 PMCID: PMC9323054 DOI: 10.3390/toxins14070471] [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: 06/17/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 11/30/2022] Open
Abstract
Fumonisin B1 (FB1), which is a mycotoxin produced by Fusarium moniliforme and Fusarium rotarum, has a number of toxic effects in animals. Moldy feed containing FB1 can damage the intestine. In this study, we used intestinal porcine epithelial cells (IPEC-J2) as an in vitro model to explore the effects of FB1 on cell cycle and apoptosis. The results showed that IPEC-J2 cells treated with 10, 20, and 40 μg/mL FB1 for 48 h experienced different degrees of damage manifested as decreases in cell number and viability, as well as cell shrinkage and floating. In addition, FB1 reduced cell proliferation and the mRNA and protein expression of proliferating cell nuclear antigen (PCNA), cyclin-dependent kinase 2 (CDK2), CDK4, cyclinD1, and cyclinE1. FB1 blocked the cell cycle in the G1 phase. FB1 also induced mitochondrial pathway apoptosis, reduced mitochondrial membrane potential, and promoted mRNA and protein expression of Caspase3, Caspase9, and Bax. The findings suggest that FB1 can induce IPEC-J2 cell damage, block the cell cycle, and promote cell apoptosis.
Collapse
|
13
|
Apigeninidin-rich Sorghum bicolor (L. Moench) extracts suppress A549 cells proliferation and ameliorate toxicity of aflatoxin B1-mediated liver and kidney derangement in rats. Sci Rep 2022; 12:7438. [PMID: 35523904 PMCID: PMC9076626 DOI: 10.1038/s41598-022-10926-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 04/08/2022] [Indexed: 02/07/2023] Open
Abstract
Sorghum bicolor plant has a high abundance of 3-deoxyanthocyanins, flavonoids and other polyphenol compounds that have been shown to offer numerous health benefits. Epidemiological studies have linked increased intake of S. bicolor to reduced risk of certain cancer types, including lung adenocarcinoma. S. bicolor extracts have shown beneficial effects in managing hepatorenal injuries. This study investigated the cytotoxic potential of three apigeninidin-rich extracts of S. bicolor (SBE-05, SBE-06 and SBE-07) against selected cancer cell lines and their ameliorative effect on aflatoxin B1 (AFB1)-mediated hepatorenal derangements in rats. We observed that, among the three potent extracts, SBE-06 more potently and selectively suppressed the growth of lung adenocarcinoma cell line (A549) (IC50 = 6.5 μg/mL). SBE-06 suppressed the expression of STAT3 but increased the expression of caspase 3. In addition, SBE-05, SBE-06 and SBE-07 inhibited oxidative and nitrosative stress, inflammation, and apoptosis and preserved the histoarchitectural networks of the liver and kidney of rats treated with AFB1. These in vitro and in vivo studies indicate the potential of these cheap and readily accessible extracts for cancer therapy and as chemo-preventive agents in preventing aflatoxin-related health issues.
Collapse
|
14
|
Li Q, Zhao T, He H, Robert N, Ding T, Hu X, Zhang T, Pan Y, Cui Y, Yu S. Ascorbic acid protects the toxic effects of aflatoxin B 1 on yak oocyte maturation. Anim Sci J 2022; 93:e13702. [PMID: 35257449 DOI: 10.1111/asj.13702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/26/2022] [Accepted: 02/03/2022] [Indexed: 11/29/2022]
Abstract
High-quality oocytes are a prerequisite for successful fertilization. Mammals feeding on aflatoxin-contaminated feed can cause reproductive toxicity, including follicular atresia, poor oocyte development and maturation, and aberrant epigenetic modifications of oocytes. In addition, the important role of ascorbic acid (AA) in reproductive biology has been confirmed, and AA is widely used as an antioxidant in cell culture. However, the toxic effects of aflatoxin B1 (AFB1 ) on yak oocytes and whether AA has protective effects remain unknown. In this study, we found that exposure to AFB1 impedes meiotic maturation of oocytes, promotes apoptosis by triggering high levels of reactive oxygen species (ROS), and disrupts mitochondrial distribution and actin integrity, resulting in a decrease in the fertilization ability and parthenogenetic development ability of oocytes. In addition, these injuries changed the DNA methylation transferase transcription level of mature oocytes. After adding 50 μg/ml AA, the indices recovered to levels close to those of the control group. The results showed that AA could protect yak oocytes from the toxic effects of AFB1 and improve the quality of oocytes.
Collapse
Affiliation(s)
- Qin Li
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Tian Zhao
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Honghong He
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Niayale Robert
- Laboratory of Animal Anatomy & Tissue Embryology, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Tianyi Ding
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Xuequan Hu
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Tongxiang Zhang
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yangyang Pan
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yan Cui
- Laboratory of Animal Anatomy & Tissue Embryology, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Sijiu Yu
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
15
|
Hajarizadeh A, Eidi A, Arefian E, Tvrda E, Mohammadi-Sangcheshmeh A. Aflatoxin B1 impairs in vitro early developmental competence of ovine oocytes. Theriogenology 2022; 183:53-60. [DOI: 10.1016/j.theriogenology.2022.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 10/19/2022]
|
16
|
In vitro and in vivo evaluation of AFB1 and OTA-toxicity through immunofluorescence and flow cytometry techniques: A systematic review. Food Chem Toxicol 2021; 160:112798. [PMID: 34973406 DOI: 10.1016/j.fct.2021.112798] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/03/2021] [Accepted: 12/24/2021] [Indexed: 01/20/2023]
Abstract
Due to the globalization, mycotoxins have been considered a major risk to human health being the main contaminants of foodstuffs. Among them, AFB1 and OTA are the most toxic and studied. Therefore, the goal of this review is to deepen the knowledge about the toxicological effects that AFB1 and OTA can induce on human health by using flow cytometry and immunofluorescence techniques in vitro and in vivo models. The examination of the selected reports shows that the majority of them are focused on immunotoxicity while the rest are concerned about nephrotoxicity, hepatotoxicity, gastrointestinal toxicity, neurotoxicity, embryotoxicity, reproductive system, breast, esophageal and lung toxicity. In relation to immunofluorescence analysis, biological processes related to AFB1- and OTA-toxicity were evaluated such as inflammation, neuronal differentiation, DNA damage, oxidative stress and cell death. In flow cytometry analysis, a wide range of assays have been performed across the reviewed studies being apoptosis assay, cell cycle analysis and intracellular ROS measurement the most employed. Although, the toxic effects of AFB1 and OTA have been reported, further research is needed to clarify AFB1 and OTA-mechanism of action on human health.
Collapse
|
17
|
Hao Y, Xing M, Gu X. Research Progress on Oxidative Stress and Its Nutritional Regulation Strategies in Pigs. Animals (Basel) 2021; 11:1384. [PMID: 34068057 PMCID: PMC8152462 DOI: 10.3390/ani11051384] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress refers to the dramatic increase in the production of free radicals in human and animal bodies or the decrease in the ability to scavenging free radicals, thus breaking the antioxidation-oxidation balance. Various factors can induce oxidative stress in pig production. Oxidative stress has an important effect on pig performance and healthy growth, and has become one of the important factors restricting pig production. Based on the overview of the generation of oxidative stress, its effects on pigs, and signal transduction pathways, this paper discussed the nutritional measures to alleviate oxidative stress in pigs, in order to provide ideas for the nutritional research of anti-oxidative stress in pigs.
Collapse
Affiliation(s)
| | | | - Xianhong Gu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.H.); (M.X.)
| |
Collapse
|
18
|
Wang X, Sun M, Li J, Song X, He H, Huan Y. Melatonin protects against defects induced by Enniatin B1 during porcine early embryo development. Aging (Albany NY) 2021; 13:5553-5570. [PMID: 33589578 PMCID: PMC7950273 DOI: 10.18632/aging.202484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/30/2020] [Indexed: 12/30/2022]
Abstract
Exogenous factors influence embryo development. Enniatin B1 (EB1), one emerging mycotoxin of Fusarium fungi, can cause damage to cells and mouse blastocysts. However, the toxicity of EB1 on porcine embryo development and whether melatonin can eliminate the detrimental effects of EB1 on embryos remain unclear. Here, this work demonstrated that EB1 significantly decreased the cleavage and blastocyst rates and blastocyst cell number of embryos in a dose and time dependent manner. Further study displayed that EB1 obviously destroyed nuclear remodeling dynamics. Importantly, EB1 triggered embryo apoptosis through downregulating the expression of Sod1,Gpx4, Cat and Bcl2l1 while upregulating the transcription of Bax and Caspase3. Moreover, EB1 significantly disrupted the transcription of Dnmt1, Dnmt3a, Tet1 and Tet3, further leading to incomplete DNA demethylation of CenRep, Oct4, Nanog and Sox2, thus, the expression of Eif1a, Oct4, Nanog and Sox2 remarkably decreased. Whereas EB1-exposed embryos were treated with melatonin, these disorders were obviously ameliorated, and the development ability of embryos was also rescued. In conclusion, EB1 exerted detrimental effects on porcine early embryos, while melatonin effectively rescued EB1-mediated defects in embryos. This work provides a novel insight into the improvement of embryo quality and the promotion of human and animal reproduction.
Collapse
Affiliation(s)
- Xiangyu Wang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, Shandong Province, China
| | - Mingju Sun
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, Shandong Province, China
| | - Jingyu Li
- Chongqing Key Laboratory of Human Embryo Engineering, Chongqing Health Center for Women and Children, Chongqing 400013, China
| | - Xuexiong Song
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, Shandong Province, China
| | - Hongbin He
- College of Life Science, Shandong Normal University, Jinan 250014, Shandong Province, China
| | - Yanjun Huan
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, Shandong Province, China
| |
Collapse
|
19
|
Effect of Triclosan Exposure on Developmental Competence in Parthenogenetic Porcine Embryo during Preimplantation. Int J Mol Sci 2020; 21:ijms21165790. [PMID: 32806749 PMCID: PMC7461051 DOI: 10.3390/ijms21165790] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 01/05/2023] Open
Abstract
Triclosan (TCS) is included in various healthcare products because of its antimicrobial activity; therefore, many humans are exposed to TCS daily. While detrimental effects of TCS exposure have been reported in various species and cell types, the effects of TCS exposure on early embryonic development are largely unknown. The aim of this study was to determine if TCS exerts toxic effects during early embryonic development using porcine parthenogenetic embryos in vitro. Porcine parthenogenetic embryos were cultured in in vitro culture medium with 50 or 100 µM TCS for 6 days. Developmental parameters including cleavage and blastocyst formation rates, developmental kinetics, and the number of blastomeres were assessed. To determine the toxic effects of TCS, apoptosis, oxidative stress, and mitochondrial dysfunction were assessed. TCS exposure resulted in a significant decrease in 2-cell rate and blastocyst formation rate, as well as number of blastomeres, but not in the cleavage rate. TCS also increased the number of apoptotic blastomeres and the production of reactive oxygen species. Finally, TCS treatment resulted in a diffuse distribution of mitochondria and decreased the mitochondrial membrane potential. Our results showed that TCS exposure impaired porcine early embryonic development by inducing DNA damage, oxidative stress, and mitochondrial dysfunction.
Collapse
|
20
|
Toralova T, Kinterova V, Chmelikova E, Kanka J. The neglected part of early embryonic development: maternal protein degradation. Cell Mol Life Sci 2020; 77:3177-3194. [PMID: 32095869 PMCID: PMC11104927 DOI: 10.1007/s00018-020-03482-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 01/24/2020] [Accepted: 02/07/2020] [Indexed: 12/28/2022]
Abstract
The degradation of maternally provided molecules is a very important process during early embryogenesis. However, the vast majority of studies deals with mRNA degradation and protein degradation is only a very little explored process yet. The aim of this article was to summarize current knowledge about the protein degradation during embryogenesis of mammals. In addition to resuming of known data concerning mammalian embryogenesis, we tried to fill the gaps in knowledge by comparison with facts known about protein degradation in early embryos of non-mammalian species. Maternal protein degradation seems to be driven by very strict rules in terms of specificity and timing. The degradation of some maternal proteins is certainly necessary for the normal course of embryonic genome activation (EGA) and several concrete proteins that need to be degraded before major EGA have been already found. Nevertheless, the most important period seems to take place even before preimplantation development-during oocyte maturation. The defects arisen during this period seems to be later irreparable.
Collapse
Affiliation(s)
- Tereza Toralova
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - Veronika Kinterova
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic.
- Department of Veterinary Sciences, Czech University of Life Sciences in Prague, Prague, Czech Republic.
| | - Eva Chmelikova
- Department of Veterinary Sciences, Czech University of Life Sciences in Prague, Prague, Czech Republic
| | - Jiri Kanka
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| |
Collapse
|
21
|
Shin KT, Nie ZW, Zhou W, Zhou D, Kim JY, Ock SA, Niu YJ, Cui XS. Connexin 43 Knockdown Induces Mitochondrial Dysfunction and Affects Early Developmental Competence in Porcine Embryos. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2020; 26:287-296. [PMID: 32036801 DOI: 10.1017/s1431927620000033] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Connexin 43 (CX43) is a component of gap junctions. The lack of functional CX43 induces oxidative stress, autophagy, and apoptosis in somatic cells. However, the role of CX43 in the early development of porcine embryos is still unknown. Thus, the aim of this study was to investigate the role of CX43, and its underlying molecular mechanisms, on the developmental competence of early porcine embryos. We performed CX43 knockdown by microinjecting dsRNA into parthenogenetically activated porcine parthenotes. The blastocyst development rate and the total number of cells in the blastocysts were significantly reduced by CX43 knockdown. Results from FITC-dextran assays showed that CX43 knockdown significantly increased membrane permeability. ZO-1 protein was obliterated in CX43 knockdown blastocysts. Mitochondrial membrane potential and ATP production were significantly reduced following CX43 knockdown. Reactive oxygen species (ROS) levels were significantly increased in the CX43 knockdown group compared to those in control embryos. Moreover, CX43 knockdown induced autophagy and apoptosis. Our findings indicate that CX43 is essential for the development and preimplantation of porcine embryos and maintains mitochondrial function, cell junction structure, and cell homeostasis by regulating membrane permeability, ROS generation, autophagy, and apoptosis in early embryos.
Collapse
Affiliation(s)
- Kyung-Tae Shin
- Department of Animal Sciences, Chungbuk National University, Chungbuk, Cheongju361-763, Republic of Korea
| | - Zheng-Wen Nie
- Department of Animal Sciences, Chungbuk National University, Chungbuk, Cheongju361-763, Republic of Korea
| | - Wenjun Zhou
- Department of Animal Sciences, Chungbuk National University, Chungbuk, Cheongju361-763, Republic of Korea
| | - Dongjie Zhou
- Department of Animal Sciences, Chungbuk National University, Chungbuk, Cheongju361-763, Republic of Korea
| | - Ju-Yeon Kim
- Department of Animal Sciences, Chungbuk National University, Chungbuk, Cheongju361-763, Republic of Korea
| | - Sun A Ock
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Jeonju55365, Republic of Korea
| | - Ying-Jie Niu
- Department of Animal Sciences, Chungbuk National University, Chungbuk, Cheongju361-763, Republic of Korea
| | - Xiang-Shun Cui
- Department of Animal Sciences, Chungbuk National University, Chungbuk, Cheongju361-763, Republic of Korea
| |
Collapse
|
22
|
Schrenk D, Bignami M, Bodin L, Chipman JK, del Mazo J, Grasl‐Kraupp B, Hogstrand C, Hoogenboom L(R, Leblanc J, Nebbia CS, Nielsen E, Ntzani E, Petersen A, Sand S, Schwerdtle T, Vleminckx C, Marko D, Oswald IP, Piersma A, Routledge M, Schlatter J, Baert K, Gergelova P, Wallace H. Risk assessment of aflatoxins in food. EFSA J 2020; 18:e06040. [PMID: 32874256 PMCID: PMC7447885 DOI: 10.2903/j.efsa.2020.6040] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
EFSA was asked to deliver a scientific opinion on the risks to public health related to the presence of aflatoxins in food. The risk assessment was confined to aflatoxin B1 (AFB1), AFB2, AFG1, AFG2 and AFM1. More than 200,000 analytical results on the occurrence of aflatoxins were used in the evaluation. Grains and grain-based products made the largest contribution to the mean chronic dietary exposure to AFB1 in all age classes, while 'liquid milk' and 'fermented milk products' were the main contributors to the AFM1 mean exposure. Aflatoxins are genotoxic and AFB1 can cause hepatocellular carcinomas (HCCs) in humans. The CONTAM Panel selected a benchmark dose lower confidence limit (BMDL) for a benchmark response of 10% of 0.4 μg/kg body weight (bw) per day for the incidence of HCC in male rats following AFB1 exposure to be used in a margin of exposure (MOE) approach. The calculation of a BMDL from the human data was not appropriate; instead, the cancer potencies estimated by the Joint FAO/WHO Expert Committee on Food Additives in 2016 were used. For AFM1, a potency factor of 0.1 relative to AFB1 was used. For AFG1, AFB2 and AFG2, the in vivo data are not sufficient to derive potency factors and equal potency to AFB1 was assumed as in previous assessments. MOE values for AFB1 exposure ranged from 5,000 to 29 and for AFM1 from 100,000 to 508. The calculated MOEs are below 10,000 for AFB1 and also for AFM1 where some surveys, particularly for the younger age groups, have an MOE below 10,000. This raises a health concern. The estimated cancer risks in humans following exposure to AFB1 and AFM1 are in-line with the conclusion drawn from the MOEs. The conclusions also apply to the combined exposure to all five aflatoxins.
Collapse
|
23
|
Huang W, Cao Z, Zhang J, Ji Q, Li Y. Aflatoxin B 1 promotes autophagy associated with oxidative stress-related PI3K/AKT/mTOR signaling pathway in mice testis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 255:113317. [PMID: 31610502 DOI: 10.1016/j.envpol.2019.113317] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/05/2019] [Accepted: 09/27/2019] [Indexed: 06/10/2023]
Abstract
Aflatoxin B1 (AFB1) is a hazard environmental pollutants and the most toxic one of all the aflatoxins. AFB1 can cause a serious impairment to testicular development and spermatogenesis, yet the underlying mechanisms remain inconclusive. Oxidative stress acts as a master mechanism of AFB1 toxicity, and can promote autophagy. Abnormal autophagy resulted in testicular damage and spermatogenesis disorders. The objective of this study was to explore the effect of AFB1 on autophagy in mice testis and its potential mechanisms. In this study, male mice were intragastrically administered with 0, 0.375, 0.75 or 1.5 mg/kg body weight AFB1 for 30 days. We found that AFB1 induced testicular damage, reduced serum testosterone level and impaired sperm quality accompanied with the elevation of oxidative stress and germ cell apoptosis. Interestingly, we observed increasing numbers of autophagosomes in AFB1-exposed mice testis. Meanwhile, AFB1 caused testis abnormal autophagy with the characterization of increased expressions of LC3, Beclin-1, Atg5 and p62. Furthermore, AFB1 downregulated the expressions of PI3K, p-AKT and p-mTOR in mice testis. Taken together, our data indicated AFB1 induced testicular damage and promoted autophagy, which were associated with oxidative stress-related PI3K/AKT/mTOR signaling pathway in mice testis.
Collapse
Affiliation(s)
- Wanyue Huang
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zheng Cao
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jian Zhang
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Qiang Ji
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yanfei Li
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
24
|
Chen X, Li C, Chen Y, Ni C, Chen X, Zhang L, Xu X, Chen M, Ma X, Zhan H, Xu A, Ge R, Guo X. Aflatoxin B1 impairs leydig cells through inhibiting AMPK/mTOR-mediated autophagy flux pathway. CHEMOSPHERE 2019; 233:261-272. [PMID: 31176127 DOI: 10.1016/j.chemosphere.2019.05.273] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/19/2019] [Accepted: 05/29/2019] [Indexed: 06/09/2023]
Abstract
Aflatoxin B1 (AFB1), a potential endocrine disrupter, has been shown to induce hepatotoxicity in animal models, but the effects of AFB1 on Leydig cell function are unclear. In this study, in vivo exposure to AFB1 at 15 and 150 μg/kg/day lowered serum testosterone (T), luteinizing hormone (LH), and follicle-stimulating hormone (FSH) levels, reduced Leydig cell number, and down-regulated the expression of testosterone biosynthesis-related genes. In vitro study showed that AFB1 (10 μM) significantly increased ROS levels, and decreased T production in Leydig cells by suppressing certain T-biosynthesis gene expressions. Moreover, AFB1 induced Leydig cell apoptosis through lowering pAMPK/AMPK ratio and increasing pmTOR/mTOR ratio, and then further up-regulating autophagy and apoptosis proteins, LC3, BECLIN 1, and BAX, as well as down-regulating autophagy flux protein P62 and anti-apoptosis protein BCL-2. AFB1-induced toxicity in Leydig cells was characterized by inhibiting T-biosynthesis gene expression, reducing Leydig cell number, promoting ROS production, and inducing cell apoptosis via suppressing AMPK/mTOR-mediated autophagy flux pathway.
Collapse
Affiliation(s)
- Xianwu Chen
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chao Li
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yong Chen
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chaobo Ni
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiuxiu Chen
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linlei Zhang
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuni Xu
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; The Second Clinical Medical School of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Min Chen
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; The Second Clinical Medical School of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinyi Ma
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; The Second Clinical Medical School of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huilu Zhan
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; The Second Clinical Medical School of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Aoyu Xu
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; The Second Clinical Medical School of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Renshan Ge
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoling Guo
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; The Second Clinical Medical School of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
25
|
Jiang Y, Hansen PJ, Xiao Y, Amaral TF, Vyas D, Adesogan AT. Aflatoxin compromises development of the preimplantation bovine embryo through mechanisms independent of reactive oxygen production. J Dairy Sci 2019; 102:10506-10513. [PMID: 31521360 DOI: 10.3168/jds.2019-16839] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 07/11/2019] [Indexed: 01/12/2023]
Abstract
Aflatoxin is a potent carcinogen often found in animal feedstuffs. Although it reportedly impairs development of the preimplantation pig embryo, it is not known whether it adversely affects development of the preimplantation bovine embryo. We conducted 3 experiments to investigate this possibility and determine whether deleterious effects of aflatoxin were caused by increased production of reactive oxygen species (ROS). Experiments were conducted with embryos produced in vitro and cultured after fertilization with various concentrations of aflatoxin. For experiment 1, embryos were treated with 0 (control), 40, 400, or 4,000 µg/L of aflatoxin B1 (AFB1). Treatment at all concentrations of AFB1 tended to reduce cleavage rate, with the 2 highest concentrations having significant effects. As compared with the control, 40 µg/L AFB1 reduced the percentage of oocytes becoming blastocysts and the percentage of cleaved embryos becoming blastocysts (19.7 vs. 8.1% and 30.3 vs. 14.3%, respectively). Complete inhibition of blastocyst formation occurred at concentrations of 400 and 4,000 µg/L of AFB1. Experiments 2 and 3 involved a 2 × 2 factorial design with effects of AFB1 (0 and 40 µg/L), the antioxidant Trolox (6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid, a water-soluble analog of vitamin E; 0 and 5 µM), and their interaction on production of ROS in putative zygotes (experiment 2) and development to the blastocyst stage (experiment 3). Production of ROS was increased by AFB1, and this effect was reversed by Trolox. However, Trolox did not prevent the reduction in development to the blastocyst stage caused by AFB1. Thus, the anti-developmental effects of AFB1 are not caused solely by increased ROS production. Rather, other underlying mechanisms exist for the adverse effects of aflatoxin on embryonic development. Overall, results indicate the potential for feeding aflatoxin-contaminated feed to cause embryonic loss in cattle.
Collapse
Affiliation(s)
- Y Jiang
- Department of Animal Sciences, University of Florida, Gainesville 32611
| | - P J Hansen
- Department of Animal Sciences, University of Florida, Gainesville 32611
| | - Y Xiao
- Department of Animal Sciences, University of Florida, Gainesville 32611
| | - T F Amaral
- Department of Animal Sciences, University of Florida, Gainesville 32611
| | - D Vyas
- Department of Animal Sciences, University of Florida, Gainesville 32611
| | - A T Adesogan
- Department of Animal Sciences, University of Florida, Gainesville 32611.
| |
Collapse
|