题名: | 玉米赤霉烯酮通过雌激素受体1调节山羊子宫内膜细胞增殖的机制研究 |
作者: | |
学号: | 2022107038 |
保密级别: | 保密两年 |
语种: | chi |
学科代码: | 090601 |
学科: | 农学 - 兽医学 - 基础兽医学 |
学生类型: | 硕士 |
学位: | 兽医硕士 |
学校: | 南京农业大学 |
院系: | |
专业: | |
研究方向: | 动物生殖毒理 |
导师姓名: | |
导师单位: | |
完成日期: | 2025-06-01 |
答辩日期: | 2025-05-26 |
外文题名: | Zearalenone Regulates Cell Proliferation of Goat Endometrial Epithelial Cells through Estrogen Receptor 1 |
关键词: | |
外文关键词: | Zearalenone ; Goat endometrial epithelial cells ; Mitochondria ; Cell proliferation ; Pyruvate |
摘要: |
玉米赤霉烯酮(Zearalenone,ZEN)是一种由镰刀菌属真菌产生的霉菌毒素,广泛存在于玉米和小麦等谷物原料及其饲料中。当家畜摄入含有ZEN的饲料后会损伤子宫机能,引起繁殖障碍,不仅造成经济损失还危害人畜健康。报道称,ZEN与内源性雌激素的结构相似,可特异性结合雌激素受体(Estrogen receptors,ERs)激活下游信号通路,但不同ER(ESR1和ESR2)在调节细胞功能及作用机制上存在显著差异。为探究ZEN对子宫内膜细胞(Endometrial Epithelial Cells,EECs)功能的影响及作用机理,本研究以山羊EECs为试验对象,在明确ZEN与哪个ER结合发挥作用的基础上,以线粒体为切入点,系统研究ZEN对EECs能量代谢与增殖能力的影响,并探讨能量代谢对细胞增殖的调节重塑作用。本试验主要分为以下三个部分: 试验Ⅰ 玉米赤霉烯酮通过线粒体ESR1-OMA1通路影响山羊子宫内膜细胞增殖的机制研究 为明析ZEN与哪个ER结合调节EECs增殖,分子对接分析发现,ZEN对ESR1的结合亲和力高于ESR2;分子动力学模拟证实ESR1-ZEN复合物的结合能大于ESR2-ZEN复合物。基因干扰验证检测发现,ZEN处理显著降低了EECs的细胞增殖率,siRNA干扰ESR1表达可显著减缓ZEN对EECs增殖的抑制作用,且增殖标志物PCNA的蛋白表达水平亦有类似变化,而干扰ESR2未显示出类似的恢复效果。流式细胞分析显示,ZEN处理减缓EECs由S期向G2/M期过渡,伴有细胞周期调节因子CCND1蛋白表达上调,CDK4和CDK6蛋白表达下调,这些影响被转染siRNA-ESR1有效缓解,而转染siRNA-ESR2没有产生显著变化。免疫共沉淀(Co-IP)试验证实,ZEN处理显著促进了ESR1-ESR1同型二聚体的形成,而ESR1-ESR2异源二聚体未被有效激活,表明ZEN通过促进ESR1同源二聚化影响山羊EECs增殖和细胞周期进程。 线粒体功能稳态在细胞增殖中具有关键作用。为探讨ZEN结合ESR1对EECs线粒体形态与功能的影响,透射电镜、线粒体荧光探针和线粒体膜电位试剂盒等检测发现,ZEN处理导致线粒体嵴受损,线粒体面积、周长和长径比等功能评价指数降低;线粒体融合蛋白MFN1、MFN2和OPA1的表达量下降,裂变蛋白FIS1的表达上调;这些变化在siRNA-ESR1处理下得以减缓。同时,ZEN处理显著降低了ATP水平、线粒体膜电位和线粒体呼吸链复合物的活性(CI-CV),而上述指标在ESR1表达被抑制的情况下均得到恢复。此外,ZEN处理导致OMA1蛋白表达显著增加,且CO-IP检测证实了ESR1和OMA1相互作用。通过OMA1自裂诱导剂CPX处理,可显著上调ZEN处理下线粒体融合蛋白MFN1、MFN2、OPA1的表达,表明OMA1在调节线粒体融合和裂变中起关键作用。综上所述,ZEN激活ESR1并上调OMA1表达,扰乱山羊EECs线粒体的结构与功能,从而影响细胞的增殖能力及细胞周期进程。 试验Ⅱ 玉米赤霉烯酮通过ESR1-PKM2通路影响山羊子宫内膜细胞丙酮酸产生和增殖的机制研究 线粒体异常可导致代谢功能障碍,影响细胞增殖和细胞周期。为探究ZEN对山羊EECs能量代谢的影响,通过丙酮酸、乳酸含量试剂盒和糖酵解酶活性试剂盒等检测发现,ZEN处理导致EECs及其培养基中丙酮酸和乳酸浓度显著降低,糖酵解关键酶PKM和LDH的活性显著降低,并且关键糖酵解调节因子PKM2和LDHA表达水平显著下调。同时,ZEN处理降低了细胞内NAD+和NADH水平,抑制了线粒体丙酮酸运输蛋白MPC1的表达。这些结果表明,ZEN可能通过减少丙酮酸的产生及其向线粒体的转运来影响山羊EECs的增殖。为探讨ZEN影响丙酮酸水平的机制,使用PKM2抑制剂-Compound 3k和PKM2激活剂-TEPP-46处理EECs发现,Compound 3k降低细胞的增殖率和PCNA蛋白表达水平,并且将细胞周期阻滞在G2/M期,伴随细胞周期调节因子CCND1、CDK4和CDK6表达下降。相反,TEPP-46能够减缓ZEN引起的增殖率和PCNA蛋白表达量下降,缓解ZEN对细胞增殖和细胞周期进程的抑制作用。CO-IP分析确定了ESR1和PKM2之间的相互作用。siRNA-ESR1或TEPP-46和ZEN共处理后,EECs中的丙酮酸和乳酸水平、PKM和LDH的酶活性以及PKM2和LDHA蛋白表达水平升高,表明ESR1-PKM2相互作用在ZEN处理的EECs中调节丙酮酸代谢。同时,siRNA-ESR1或TEPP-46和ZEN共处理后,线粒体碎片化显著减少,线粒体融合蛋白MFN1、MFN2和OPA1表达增加,线粒体裂变基因FIS1的表达下调。此外,ZEN处理显著降低了ATP水平、线粒体膜电位和线粒体呼吸链复合物的活性(CI-CV),这些变化通过siRNA-ESR1或TEPP-46共处理得到缓解。综上所述,ZEN可能通过与ESR1和PKM2的相互作用影响丙酮酸水平,从而影响山羊EECs线粒体形态和功能,进而影响细胞增殖和细胞周期进程。 试验Ⅲ OMA1介导丙酮酸调控山羊子宫内膜细胞线粒体动力学和增殖的机制研究 为探究OMA1参与调节丙酮酸代谢影响山羊EECs线粒体动力学和增殖作用,通过EdU试验和流式细胞等检测发现,OE-OMA1处理导致细胞增殖率和PCNA蛋白表达量降低,G2/M期细胞比例增加,CCND1、CDK4和CDK6的蛋白表达水平降低,而与丙酮酸共处理可减弱这些效应,表明OMA1过表达抑制了丙酮酸对细胞周期的促进作用。线粒体动力学对丙酮酸的有效利用至关重要。为探究OMA1在丙酮酸调节线粒体结构与功能方面的作用,线粒体荧光探针和线粒体膜电位试剂盒等检测发现,过表达OMA1的细胞中线粒体碎片化显著增加并且线粒体面积、周长、纵横比等功能评价指数降低,线粒体融合蛋白MFN1、MFN2和OPA1的表达下调,裂变蛋白FIS1的表达增加,而添加丙酮酸则有效减缓了这些变化。此外,过表达OMA1显著降低了EECs的ATP水平、线粒体膜电位和线粒体呼吸链复合物的活性(CI-CV),这些作用也可通过添加丙酮酸得到缓解。上述结果表明,OMA1参与丙酮酸调节线粒体结构和功能,进而影响山羊EECs增殖和周期进程。 |
外摘要要: |
Zearalenone (Zearalenone, ZEN) is a mycotoxin produced by fungi of the genus Fusarium and is widely present in cereal raw materials such as corn and wheat, as well as their derived feeds. When livestock ingest feed contaminated with ZEN, it can impair uterine function and cause reproductive disorders, resulting in economic losses and posing risks to the health of both humans and animals. It has been reported that ZEN structurally resembles endogenous estrogens and can specifically bind to estrogen receptors (Estrogen receptors, ERs) to activate downstream signaling pathways. However, there are significant differences in cellular functions and regulatory mechanisms between different ERs (ESR1 and ESR2). To explore the effects of ZEN on the function of endometrial epithelial cells (Endometrial Epithelial Cells, EECs) and its underlying mechanisms of action, this study used goat EECs as the experimental model. Based on clarifying which ER ZEN binds to exert its effects, this study systematically investigated the impact of ZEN on the energy metabolism and proliferative capacity of EECs, using mitochondria as the focal point, and explored the regulatory and remodeling effects of energy metabolism on cell proliferation. The main experimental procedures are divided into the following three parts: Experiment Ⅰ Mechanism of zearalenone affecting proliferation of goat endometrial epithelial cells via the mitochondrial ESR1-OMA1 pathway To elucidate which ER ZEN binds to regulate the proliferation of EECs, molecular docking analysis revealed that ZEN has a higher binding affinity for ESR1 than for ESR2. Molecular dynamics simulations further confirmed that the binding energy of the ESR1-ZEN complex is greater than that of the ESR2-ZEN complex. Validation experiments using gene interference demonstrated that treatment with ZEN significantly reduced the proliferation rate of EECs. Interference with ESR1 expression via siRNA significantly reversed this inhibitory effect, and the protein expression level of the proliferation marker PCNA was also restored accordingly. In contrast, interference with ESR2 did not show a similar restorative effect. Flow cytometry analysis indicated that ZEN treatment slowed the transition of EECs from the S phase to the G2/M phase of the cell cycle, accompanied by upregulation of the cell cycle regulatory factor CCND1 protein expression and downregulation of CDK4 and CDK6 expression. These effects were effectively reversed by transfection with siRNA-ESR1, while transfection with siRNA-ESR2 did not result in significant changes. Co-immunoprecipitation (Co-IP) experiments further confirmed that ZEN treatment significantly promoted the formation of ESR1-ESR1 homodimers, while the ESR1-ESR2 heterodimer was not effectively activated. This suggests that ZEN influences the proliferation and cell cycle progression of goat EECs by promoting ESR1 homodimerization. Mitochondrial functional homeostasis plays a crucial role in cell proliferation. To investigate the effects of ZEN on mitochondrial morphology and function in EECs via ESR1, transmission electron microscopy, mitochondrial fluorescent probes, and mitochondrial membrane potential assay kits were employed. These analyses revealed that ZEN treatment led to damage to the mitochondrial cristae, as well as decreased mitochondrial area, perimeter, and aspect ratio, which are indices for evaluating mitochondrial function. The protein expression of mitochondrial fusion genes (MFN1, MFN2, and OPA1) was downregulated, while the protein expression of the fission gene FIS1 was upregulated. These changes were reversed upon interference with ESR1 expression using siRNA-ESR1. Concurrently, ZEN treatment significantly reduced ATP levels, mitochondrial membrane potential, and the activity of mitochondrial respiratory chain complexes (CI-CV). These indices were restored when ESR1 expression was inhibited. Additionally, ZEN treatment resulted in a significant increase in the protein expression of OMA1, and Co-IP assays confirmed the interaction between ESR1 and OMA1. Treatment with the OMA1 auto-cleavage inducer CPX significantly upregulated the expression of mitochondrial fusion proteins (MFN1, MFN2, and OPA1) under ZEN treatment conditions, indicating that OMA1 plays a key role in regulating mitochondrial fusion and fission. In summary, ZEN primarily affects the proliferation capacity and cell cycle progression of goat EECs by activating ESR1 and upregulating OMA1 expression, thereby disrupting the structure and function of mitochondrial in EECs. Experiment II Mechanism of zearalenone affecting pyruvate production and proliferation of goat endometrial epithelial cells via the ESR1-PKM2 pathway Mitochondrial abnormalities can lead to metabolic dysfunction, affecting cell proliferation and the cell cycle. To investigate the effects of ZEN on the energy metabolism of goat EECs, assays using pyruvate and lactate content kits and glycolytic enzyme activity kits revealed that ZEN treatment significantly decreased the concentrations of pyruvate and lactate in both EECs and their culture media, as well as the activities of key glycolytic enzymes (PKM and LDH). Moreover, the expression levels of the key glycolytic regulatory factors PKM2 and LDHA were significantly downregulated. Additionally, ZEN treatment reduced intracellular NAD+ and NADH levels and inhibited the expression of the mitochondrial pyruvate carrier MPC1. These results suggest that ZEN may affect the proliferation of goat EECs by reducing pyruvate production and its transport into mitochondria. To explore the mechanism by which ZEN affects pyruvate levels, EECs were treated with the PKM2 inhibitor Compound 3k and the PKM2 activator TEPP-46. Compound 3k reduced cell proliferation rates and PCNA protein expression levels, arrested the cell cycle at the G2/M phase, and downregulated the expression of cell cycle regulatory factors CCND1, CDK4, and CDK6. In contrast, TEPP-46 reversed the ZEN-induced decreases in proliferation rates and PCNA protein expression, alleviating the inhibitory effects of ZEN on cell proliferation and cell cycle progression. Co-IP analysis confirmed the interaction between ESR1 and PKM2. After co-treatment with siRNA-ESR1 or TEPP-46 and ZEN, pyruvate and lactate levels, as well as the enzyme activities of PKM and LDH and the protein expression levels of PKM2 and LDHA, were all increased in EECs, indicating that the ESR1-PKM2 interaction regulates pyruvate metabolism in ZEN-treated EECs. Concurrently, mitochondrial fragmentation was significantly reduced, and the expression of mitochondrial fusion proteins MFN1, MFN2, and OPA1 was increased, while the expression of the mitochondrial fission gene FIS1 was downregulated. Furthermore, ZEN treatment significantly decreased ATP levels, mitochondrial membrane potential, and the activity of mitochondrial respiratory chain complexes (CI-CV), changes that were alleviated by co-treatment with siRNA-ESR1 or TEPP-46. In summary, ZEN may affect pyruvate levels through interactions with ESR1 and PKM2, thereby influencing mitochondrial morphology and function in goat EECs and subsequently affecting cell proliferation and cell cycle progression. Experiment III Mechanism of OMA1-mediated pyruvate regulation of mitochondrial dynamics and proliferation in goat endometrial epithelial cells To investigate the role of OMA1 in regulating pyruvate-mediated mitochondrial dynamics and proliferation in goat EECs, assays using EdU labeling and flow cytometry were conducted. The results showed that overexpression of OMA1 (OE-OMA1) significantly decreased cell proliferation rates and PCNA protein expression levels, increased the proportion of cells in the G2/M phase, and downregulated the protein expression levels of CCND1, CDK4, and CDK6. However, co-treatment with pyruvate attenuated these effects, indicating that OMA1 overexpression inhibits the pro-proliferative effects of pyruvate on the cell cycle. Mitochondrial dynamics are crucial for the effective utilization of pyruvate. To explore the role of OMA1 in pyruvate-regulated mitochondrial structure and function, mitochondrial fluorescence probes and mitochondrial membrane potential kits were used. Cells overexpressing OMA1 exhibited increased mitochondrial fragmentation, decreased mitochondrial area, perimeter, aspect ratio, and form factor, which are indices for evaluating mitochondrial function. Additionally, the protein expression of mitochondrial fusion genes (MFN1, MFN2, and OPA1) was downregulated, while the fission gene FIS1 was upregulated. These changes were reversed by the addition of pyruvate. Moreover, OE-OMA1 significantly reduced ATP levels, mitochondrial membrane potential, and the activity of mitochondrial respiratory chain complexes (CI-CV), effects that were also alleviated by pyruvate co-treatment. These findings suggest that OMA1 plays a key role in regulating pyruvate-mediated mitochondrial structure and function, thereby influencing the proliferation and cell cycle progression of goat EECs. |
参考文献: |
[1]Adibnia E, Razi M, Malekinejad H. Zearalenone and 17 β-estradiol induced damages in male rats reproduction potential; evidence for ERα and ERβ receptors expression and steroidogenesis[J]. Toxicon. 2016, 120: 133-146. [2]Ahola S, Rivera Mejías P, Hermans S, et al. OMA1-mediated integrated stress response protects against ferroptosis in mitochondrial cardiomyopathy[J]. Cell Metab. 2022, 34: 1875-1891.e1877. [3]Barański W, Gajęcka M, Zielonka Ł, et al. Occurrence of Zearalenone and Its Metabolites in the Blood of High-Yielding Dairy Cows at Selected Collection Sites in Various Disease States[J]. Toxins (Basel). 2021, 13: 446. [4]Belli P, Bellaton C, Durand J, et al. Fetal and neonatal exposure to the mycotoxin zearalenone induces phenotypic alterations in adult rat mammary gland[J]. Food Chem Toxicol. 2010, 48: 2818-2826. [5]Berger T, Esbenshade K L, Diekman M A, et al. Influence of prepubertal consumption of zearalenone on sexual development of boars[J]. J Anim Sci. 1981, 53: 1559-1564. [6]Birsoy K, Wang T, Chen W W, et al. An Essential Role of the Mitochondrial Electron Transport Chain in Cell Proliferation Is to Enable Aspartate Synthesis[J]. Cell. 2015, 162: 540-551. [7]Biscoto G L, Salvato L A, Alvarenga É R, et al. Mycotoxins in Cattle Feed and Feed Ingredients in Brazil: A Five-Year Survey[J]. Toxins (Basel). 2022, 14: 552. [8]Cai P, Feng N, Zheng W, et al. Treatment with, Resveratrol, a SIRT1 Activator, Prevents Zearalenone-Induced Lactic Acid Metabolism Disorder in Rat Sertoli Cells[J]. Molecules. 2019, 24: 2474. [9]Cai P, Feng Z, Feng N, et al. Activated AMPK promoted the decrease of lactate production in rat Sertoli cells exposed to Zearalenone[J]. Ecotoxicol Environ Saf. 2021, 220: 112367. [10]Casasnovas C, Banchs I, Cassereau J, et al. Phenotypic spectrum of MFN2 mutations in the Spanish population[J]. J Med Genet. 2010, 47: 249-256. [11]Chan D C. Mitochondrial Dynamics and Its Involvement in Disease[J]. Annu Rev Pathol. 2020, 15: 235-259. [12]Chen L, Chen D, Pan Y, et al. Inhibition of mitochondrial OMA1 ameliorates osteosarcoma tumorigenesis[J]. Cell Death Dis. 2024, 15: 786. [13]Chen X X, Yang C W, Huang L B, et al. Zearalenone Altered the Serum Hormones, Morphologic and Apoptotic Measurements of Genital Organs in Post-weaning Gilts[J]. Asian-Australas J Anim Sci. 2015, 28: 171-179. [14]Chianese T, Trinchese G, Leandri R, et al. Glyphosate Exposure Induces Cytotoxicity, Mitochondrial Dysfunction and Activation of ERα and ERβ Estrogen Receptors in Human Prostate PNT1A Cells[J]. Int J Mol Sci. 2024, 25: 7039. [15]Chini C C S, Zeidler J D, Kashyap S, et al. Evolving concepts in NAD(+) metabolism[J]. Cell Metab. 2021, 33: 1076-1087. [16]Cuenoud B, Ipek Ö, Shevlyakova M, et al. Brain NAD Is Associated With ATP Energy Production and Membrane Phospholipid Turnover in Humans[J]. Front Aging Neurosci. 2020, 12: 609517. [17]Dai C, Hou M, Yang X, et al. Increased NAD(+) levels protect female mouse reproductive system against zearalenone-impaired glycolysis, lipid metabolism, antioxidant capacity and inflammation[J]. Reprod Toxicol. 2024, 124: 108530. [18] Dänicke S, Swiech E, Buraczewska L, et al. Kinetics and metabolism of zearalenone in young female pigs[J]. J Anim Physiol Anim Nutr (Berl). 2005, 89: 268-276. [19] Dellafiora L, Ruotolo R, Perotti A, et al. Molecular insights on xenoestrogenic potential of zearalenone-14-glucoside through a mixed in vitro/in silico approach[J]. Food and Chemical Toxicology. 2017, 108: 257-266. [20] Devine T L, Rosenkrans C F, Philipp D, et al. Growth, reproductive development, and estrous behavior of beef heifers treated with growth promotants[J]. The Professional Animal Scientist. 2015, 31: 114-119. [21] Díaz-Ramos J, Flores-Flores M, Ayala M E, et al. Impaired serotonin communication during juvenile development in rats diminishes adult sperm quality[J]. Syst Biol Reprod Med. 2018, 64: 340-347. [22] Dietrich D R. Toxicological and pathological applications of proliferating cell nuclear antigen (PCNA), a novel endogenous marker for cell proliferation[J]. Crit Rev Toxicol. 1993, 23: 77-109. [23] Duranova H, Valkova V, Knazicka Z, et al. Mitochondria: A worthwhile object for ultrastructural qualitative characterization and quantification of cells at physiological and pathophysiological states using conventional transmission electron microscopy[J]. Acta Histochemica. 2020, 122: 151646. [24] Fogo G M, Raghunayakula S, Emaus K J, et al. Mitochondrial membrane potential and oxidative stress interact to regulate Oma1-dependent processing of Opa1 and mitochondrial dynamics[J]. Faseb j. 2024, 38: e70066. [25] Gajęcka M, Otrocka-Domagała I, Brzuzan P, et al. Immunohistochemical Expression (IE) of Oestrogen Receptors in the Intestines of Prepubertal Gilts Exposed to Zearalenone[J]. Toxins (Basel). 2023, 15: 122. [26] Gajecka M, Przybylska-Gornowicz B. The low doses effect of experimental zearalenone (ZEN) intoxication on the presence of Ca2+ in selected ovarian cells from pre-pubertal bitches[J]. Pol J Vet Sci. 2012, 15: 711-720. [27] Gajęcka M, Zielonka Ł, Dąbrowski M, et al. The effect of low doses of zearalenone and its metabolites on progesterone and 17β-estradiol concentrations in peripheral blood and body weights of pre-pubertal female Beagle dogs[J]. Toxicon. 2013, 76: 260-269. [28] Gao L, Yang F, Tang D, et al. Mediation of PKM2-dependent glycolytic and non-glycolytic pathways by ENO2 in head and neck cancer development[J]. J Exp Clin Cancer Res. 2023, 42: 1. [29] Gao X, Sun L, Zhang N, et al. Gestational Zearalenone Exposure Causes Reproductive and Developmental Toxicity in Pregnant Rats and Female Offspring[J]. Toxins (Basel). 2017a, 9: 21. [30] Gao X, Sun L, Zhang N, et al. Gestational Zearalenone Exposure Causes Reproductive and Developmental Toxicity in Pregnant Rats and Female Offspring[J]. Toxins (Basel). 2017b, 9: 21. [31] Grgic D, Novak B, Varga E, et al. Estrogen receptor α interaction of zearalenone and its phase I metabolite α-zearalenol in combination with soy isoflavones in hERα-HeLa-9903 cells[J]. Mycotoxin Res. 2024, 40: 97-109. [32] Gurung S, Greening D W, Catt S, et al. Exosomes and soluble secretome from hormone-treated endometrial epithelial cells direct embryo implantation[J]. Mol Hum Reprod. 2020, 26: 510-520. [33] Han X, Huangfu B, Xu T, et al. Research Progress of Safety of Zearalenone: A Review[J]. Toxins (Basel). 2022, 14: 386. [34] Helguero L A, Faulds M H, Gustafsson J A, et al. Estrogen receptors alfa (ERalpha) and beta (ERbeta) differentially regulate proliferation and apoptosis of the normal murine mammary epithelial cell line HC11[J]. Oncogene. 2005, 24: 6605-6616. [35] Hodsdon M E, Ponder J W, Cistola D P. The NMR solution structure of intestinal fatty acid-binding protein complexed with palmitate: application of a novel distance geometry algorithm[J]. J Mol Biol. 1996, 264: 585-602. [36] Hou Y J, Zhu C C, Xu Y X, et al. Zearalenone exposure affects mouse oocyte meiotic maturation and granulosa cell proliferation[J]. Environ Toxicol. 2015, 30: 1226-1233. [37] Hryc C F, Baker M L. AlphaFold2 and CryoEM: Revisiting CryoEM modeling in near-atomic resolution density maps[J]. iScience. 2022, 25: 104496. [38] Hu J, Xu M, Dai Y, et al. Exploration of Bcl-2 family and caspases-dependent apoptotic signaling pathway in Zearalenone-treated mouse endometrial stromal cells[J]. Biochem Biophys Res Commun. 2016, 476: 553-559. [39] Hueza I M, Raspantini P C, Raspantini L E, et al. Zearalenone, an estrogenic mycotoxin, is an immunotoxic compound[J]. Toxins (Basel). 2014, 6: 1080-1095. [40] Ji J, Wang Q, Wu H, et al. Insights into cellular metabolic pathways of the combined toxicity responses of Caco-2 cells exposed to deoxynivalenol, zearalenone and Aflatoxin B(1)[J]. Food Chem Toxicol. 2019, 126: 106-112. [41] Ji J, Zhu P, Blaženović I, et al. Explaining combinatorial effects of mycotoxins Deoxynivalenol and Zearalenone in mice with urinary metabolomic profiling[J]. Sci Rep. 2018, 8: 3762. [42] Jia M, Dahlman-Wright K, Gustafsson J. Estrogen receptor alpha and beta in health and disease[J]. Best Pract Res Clin Endocrinol Metab. 2015, 29: 557-568. [43] Juraschek L M, Kappenberg A, Amelung W. Mycotoxins in soil and environment[J]. Sci Total Environ. 2022, 814: 152425. [44] Karaman E F, Zeybel M, Ozden S. Evaluation of the epigenetic alterations and gene expression levels of HepG2 cells exposed to zearalenone and α-zearalenol[J]. Toxicol Lett. 2020, 326: 52-60. [45] Kharenko O A, Patel R G, Calosing C, et al. Combination of ZEN-3694 with CDK4/6 inhibitors reverses acquired resistance to CDK4/6 inhibitors in ER-positive breast cancer[J]. Cancer Gene Ther. 2022, 29: 859-869. [46] Kinkade C W, Rivera-Núñez Z, Gorcyzca L, et al. Impact of Fusarium-Derived Mycoestrogens on Female Reproduction: A Systematic Review[J]. Toxins (Basel). 2021, 13: 373. [47] Koh E, Kim Y K, Shin D, et al. MPC1 is essential for PGC-1α-induced mitochondrial respiration and biogenesis[J]. Biochem J. 2018, 475: 1687-1699. [48] Koprivica I, Gajić D, Pejnović N, et al. Ethyl Pyruvate Promotes Proliferation of Regulatory T Cells by Increasing Glycolysis[J]. Molecules. 2020, 25: 4112. [49] Koraichi F, Videmann B, Mazallon M, et al. Zearalenone exposure modulates the expression of ABC transporters and nuclear receptors in pregnant rats and fetal liver[J]. Toxicol Lett. 2012, 211: 246-256. [50] Kornfeld O S, Qvit N, Haileselassie B, et al. Interaction of mitochondrial fission factor with dynamin related protein 1 governs physiological mitochondrial function in vivo[J]. Sci Rep. 2018, 8: 14034. [51] Kowalska K, Habrowska-Górczyńska D E, Domińska K, et al. The dose-dependent effect of zearalenone on mitochondrial metabolism, plasma membrane permeabilization and cell cycle in human prostate cancer cell lines[J]. Chemosphere. 2017, 180: 455-466. [52] Kunishige K, Kawate N, Inaba T, et al. Exposure to Zearalenone During Early Pregnancy Causes Estrogenic Multitoxic Effects in Mice[J]. Reprod Sci. 2017, 24: 421-427. [53] Larson-Casey J L, He C, Carter A B. Mitochondrial quality control in pulmonary fibrosis[J]. Redox Biol. 2020, 33: 101426. [54] Lee R, Kim D W, Lee W Y, et al. Zearalenone Induces Apoptosis and Autophagy in a Spermatogonia Cell Line[J]. Toxins (Basel). 2022, 14: 148. [55] Li L, Xiaoxue S, Yuchong Z, et al. Application in photocatalytic degradation of zearalenone based on graphitic carbon nitride[J]. Luminescence. 2022, 37: 190-198. [56] Li R, Andersen C L, Hu L, et al. Dietary exposure to mycotoxin zearalenone (ZEA) during post-implantation adversely affects placental development in mice[J]. Reprod Toxicol. 2019, 85: 42-50. [57] Li Z, Lyu C, Ren Y, et al. Role of TET Dioxygenases and DNA Hydroxymethylation in Bisphenols-Stimulated Proliferation of Breast Cancer Cells[J]. Environ Health Perspect. 2020, 128: 27008. [58] Liang Z, Ren Z, Gao S, et al. Individual and combined effects of deoxynivalenol and zearalenone on mouse kidney[J]. Environ Toxicol Pharmacol. 2015, 40: 686-691. [59] Lijalem Y G, Gab-Allah M A, Yu H, et al. Occurrence of zearalenone and its major metabolites in cereal flour from Korea[J]. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 2023, 40: 675-687. [60] Lunt S Y, Vander Heiden M G. Aerobic glycolysis: meeting the metabolic requirements of cell proliferation[J]. Annu Rev Cell Dev Biol. 2011, 27: 441-464. [61] Ma J, Huang R, Zhang H, et al. The Protective Effect of Quercetin against the Cytotoxicity Induced by Fumonisin B1 in Sertoli Cells[J]. Int J Mol Sci. 2024, 25: 8764. [62] Ma R, Wu Y, Zhai Y, et al. Exogenous pyruvate represses histone gene expression and inhibits cancer cell proliferation via the NAMPT-NAD+-SIRT1 pathway[J]. Nucleic Acids Res. 2019, 47: 11132-11150. [63] Ma Z, Su J, Guo T, et al. Neuromedin B and Its Receptor: Gene Cloning, Tissue Distribution and Expression Levels of the Reproductive Axis in Pigs[J]. PLoS One. 2016, 11: e0151871. [64] Malekinejad H, Maas-Bakker R, Fink-Gremmels J. Species differences in the hepatic biotransformation of zearalenone[J]. Vet J. 2006, 172: 96-102. [65] Martins C, Torres D, Lopes C, et al. Food Consumption Data as a Tool to Estimate Exposure to Mycoestrogens[J]. Toxins (Basel). 2020, 12: 118. [66] Mohammad I, Starskaia I, Nagy T, et al. Estrogen receptor α contributes to T cell-mediated autoimmune inflammation by promoting T cell activation and proliferation[J]. Sci Signal. 2018, 11: eaap9415. [67] Muthulakshmi S, Hamideh P F, Habibi H R, et al. Mycotoxin zearalenone induced gonadal impairment and altered gene expression in the hypothalamic-pituitary-gonadal axis of adult female zebrafish (Danio rerio)[J]. J Appl Toxicol. 2018, 38: 1388-1397. [68] Nie Y, Yan J, Huang X, et al. Dihydrotanshinone I targets ESR1 to induce DNA double-strand breaks and proliferation inhibition in hepatocellular carcinoma[J]. Phytomedicine. 2024, 130: 155767. [69] Nikaido Y, Danbara N, Tsujita-Kyutoku M, et al. Effects of prepubertal exposure to xenoestrogen on development of estrogen target organs in female CD-1 mice[J]. In Vivo. 2005, 19: 487-494. [70] Pack E D, Weiland S, Musser R, et al. Survey of zearalenone and type-B trichothecene mycotoxins in swine feed in the USA[J]. Mycotoxin Res. 2021, 37: 297-313. [71] Pizzo F, Caloni F, Schutz L F, et al. Individual and combined effects of deoxynivalenol and α-zearalenol on cell proliferation and steroidogenesis of granulosa cells in cattle[J]. Environ Toxicol Pharmacol. 2015, 40: 722-728. [72] Prouillac C, Videmann B, Mazallon M, et al. Induction of cells differentiation and ABC transporters expression by a myco-estrogen, zearalenone, in human choriocarcinoma cell line (BeWo)[J]. Toxicology. 2009, 263: 100-107. [73] Purushottam Dharaskar S, Paithankar K, Kanugovi Vijayavittal A, et al. Mitochondrial chaperone, TRAP1 modulates mitochondrial dynamics and promotes tumor metastasis[J]. Mitochondrion. 2020, 54: 92-101. [74] Qi W, Keenan H A, Li Q, et al. Pyruvate kinase M2 activation may protect against the progression of diabetic glomerular pathology and mitochondrial dysfunction[J]. Nat Med. 2017, 23: 753-762. [75] Rai A, Das M, Tripathi A. Occurrence and toxicity of a fusarium mycotoxin, zearalenone[J]. Crit Rev Food Sci Nutr. 2020, 60: 2710-2729. [76] Ranzenigo G, Caloni F, Cremonesi F, et al. Effects of Fusarium mycotoxins on steroid production by porcine granulosa cells[J]. Anim Reprod Sci. 2008, 107: 115-130. [77] Ren R, Guo J, Shi J, et al. PKM2 regulates angiogenesis of VR-EPCs through modulating glycolysis, mitochondrial fission, and fusion[J]. J Cell Physiol. 2020, 235: 6204-6217. [78] Ribas V, Drew B G, Zhou Z, et al. Skeletal muscle action of estrogen receptor α is critical for the maintenance of mitochondrial function and metabolic homeostasis in females[J]. Sci Transl Med. 2016, 8: 334ra354. [79] Ropejko K, Twarużek M. Zearalenone and Its Metabolites—General Overview, Occurrence, and Toxicity[J]. Toxins (Basel). 2021, 13: 35. [80] Ruan Y, Hu J, Che Y, et al. CHCHD2 and CHCHD10 regulate mitochondrial dynamics and integrated stress response[J]. Cell Death Dis. 2022, 13: 156. [81] Salama S A, Mohammad M A, Diaz-Arrastia C R, et al. Estradiol-17β upregulates pyruvate kinase M2 expression to coactivate estrogen receptor-α and to integrate metabolic reprogramming with the mitogenic response in endometrial cells[J]. J Clin Endocrinol Metab. 2014, 99: 3790-3799. [82] Samik A, Safitri E. Mycotoxin binders potential on histological of ovary mice exposed by zearalenone[J]. Vet World. 2017, 10: 353-357. [83] Sanderson T H, Reynolds C A, Kumar R, et al. Molecular mechanisms of ischemia-reperfusion injury in brain: pivotal role of the mitochondrial membrane potential in reactive oxygen species generation[J]. Mol Neurobiol. 2013, 47: 9-23. [84] Savard C, Gawhary S, Boyer A, et al. Assessment of Zearalenone-Induced Cell Survival and of Global Gene Regulation in Mouse TM4 Sertoli Cells[J]. Toxins (Basel). 2022, 14: 98. [85] Savard C, Nogues P, Boyer A, et al. Prevention of deoxynivalenol- and zearalenone-associated oxidative stress does not restore MA-10 Leydig cell functions[J]. Toxicology. 2016, 341-343: 17-27. [86] Seo B J, Yoon S H, Do J T. Mitochondrial Dynamics in Stem Cells and Differentiation[J]. Int J Mol Sci. 2018, 19: 3893. [87] Sheng N, Zhang Z, Zheng H, et al. Scutellarin Rescued Mitochondrial Damage through Ameliorating Mitochondrial Glucose Oxidation via the Pdk-Pdc Axis[J]. Adv Sci (Weinh). 2023, 10: e2303584. [88] Shi W T, Yao C P, Liu W H, et al. An fusaric acid-based CRISPR library screen identifies MDH2 as a broad-spectrum regulator of Fusarium toxin-induced cell death[J]. J Hazard Mater. 2024, 480: 135937. [89] Shi X Y, Wang Z, Liu L, et al. Low concentrations of bisphenol A promote human ovarian cancer cell proliferation and glycolysis-based metabolism through the estrogen receptor-α pathway[J]. Chemosphere. 2017, 185: 361-367. [90] Silva T E S, De Brito D C C, De Sá N a R, et al. Equol: A Microbiota Metabolite Able to Alleviate the Negative Effects of Zearalenone during In Vitro Culture of Ovine Preantral Follicles[J]. Toxins (Basel). 2019, 11: 652. [91] Smith M C, Hymery N, Troadec S, et al. Hepatotoxicity of fusariotoxins, alone and in combination, towards the HepaRG human hepatocyte cell line[J]. Food Chem Toxicol. 2017, 109: 439-451. [92] Song T, Yang W, Huang L, et al. Zearalenone exposure affects the Wnt/β-catenin signaling pathway and related genes of porcine endometrial epithelial cells in vitro[J]. Anim Biosci. 2021, 34: 993-1005. [93] Song T, Zhou X, Ma X, et al. Zearalenone Promotes Uterine Development of Weaned Gilts by Interfering with Serum Hormones and Up-Regulating Expression of Estrogen and Progesterone Receptors[J]. Toxins (Basel). 2022, 14: 732. [94] Stanciu O, Juan C, Berrada H, et al. Study on Trichothecene and Zearalenone Presence in Romanian Wheat Relative to Weather Conditions[J]. Toxins (Basel). 2019, 11: 163. [95] Suski J M, Lebiedzinska M, Bonora M, et al. Relation between mitochondrial membrane potential and ROS formation[J]. Methods Mol Biol. 2012, 810: 183-205. [96] Szabó-Fodor J, Szabó A, Kócsó D, et al. Interaction between the three frequently co-occurring Fusarium mycotoxins in rats[J]. J Anim Physiol Anim Nutr (Berl). 2019, 103: 370-382. [97] Takenaka M, Yamada K, Lu T, et al. Alternative splicing of the pyruvate kinase M gene in a minigene system[J]. Eur J Biochem. 1996, 235: 366-371. [98] Tanwar A K, Dhiman N, Kumar A, et al. Engagement of phytoestrogens in breast cancer suppression: Structural classification and mechanistic approach[J]. Eur J Med Chem. 2021, 213: 113037. [99] Turcotte J C, Hunt P J, Blaustein J D. Estrogenic effects of zearalenone on the expression of progestin receptors and sexual behavior in female rats[J]. Horm Behav. 2005, 47: 178-184. [100] Tyagi J S, Venkitasubramanian T A. The role of glycolysis in aflatoxin biosynthesis[J]. Can J Microbiol. 1981, 27: 1276-1282. [101] Van Der Spoel D, Lindahl E, Hess B, et al. GROMACS: fast, flexible, and free[J]. J Comput Chem. 2005, 26: 1701-1718. [102] Vásquez-Trincado C, García-Carvajal I, Pennanen C, et al. Mitochondrial dynamics, mitophagy and cardiovascular disease[J]. J Physiol. 2016, 594: 509-525. [103] Wai T, García-Prieto J, Baker M J, et al. Imbalanced OPA1 processing and mitochondrial fragmentation cause heart failure in mice[J]. Science. 2015, 350: aad0116. [104] Wan B, Huang L, Jing C, et al. Zearalenone promotes follicle development through activating the SIRT1/PGC-1α signaling pathway in the ovaries of weaned gilts[J]. J Anim Sci. 2022, 100: skac058. [105] Wang D F, Zhang N Y, Peng Y Z, et al. Interaction of zearalenone and soybean isoflavone on the development of reproductive organs, reproductive hormones and estrogen receptor expression in prepubertal gilts[J]. Anim Reprod Sci. 2010, 122: 317-323. [106] Wang T, Ye Y, Ji J, et al. Diet composition affects long-term zearalenone exposure on the gut-blood-liver axis metabolic dysfunction in mice[J]. Ecotoxicol Environ Saf. 2022, 236: 113466. [107] Wang Y, Tan W, Leung L K. Zeranol upregulates corticotropin releasing hormone expression in the placental cell line JEG-3[J]. Toxicol Lett. 2013, 219: 218-222. [108] Warth B, Preindl K, Manser P, et al. Transfer and Metabolism of the Xenoestrogen Zearalenone in Human Perfused Placenta[J]. Environ Health Perspect. 2019, 127: 107004. [109] Wei C C, Yang N C, Huang C W. Zearalenone Induces Dopaminergic Neurodegeneration via DRP-1-Involved Mitochondrial Fragmentation and Apoptosis in a Caenorhabditis elegans Parkinson's Disease Model[J]. J Agric Food Chem. 2021, 69: 12030-12038. [110] Wu L, Qiu L, Zhang H, et al. Optimization for the Production of Deoxynivalenoland Zearalenone by Fusarium graminearum UsingResponse Surface Methodology[J]. Toxins (Basel). 2017, 9: 57. [111] Xie H, Hu J, Xiao C, et al. Exploration of ZEA cytotoxicity to mouse endometrial stromal cells and RNA-seq analysis[J]. J Biochem Mol Toxicol. 2017, 31 (4). [112] Yan R, Wang H, Zhu J, et al. Procyanidins inhibit zearalenone-induced apoptosis and oxidative stress of porcine testis cells through activation of Nrf2 signaling pathway[J]. Food Chem Toxicol. 2022a, 165: 113061. [113] Yan W K, Liu Y N, Song S S, et al. Zearalenone affects the growth of endometriosis via estrogen signaling and inflammatory pathways[J]. Ecotoxicol Environ Saf. 2022b, 241: 113826. [114] Yang F, Li L, Chen K, et al. Melatonin alleviates β-zearalenol and HT-2 toxin-induced apoptosis and oxidative stress in bovine ovarian granulosa cells[J]. Environ Toxicol Pharmacol. 2019, 68: 52-60. [115] Yang L J, Zhou M, Huang L B, et al. Zearalenone-Promoted Follicle Growth through Modulation of Wnt-1/β-Catenin Signaling Pathway and Expression of Estrogen Receptor Genes in Ovaries of Postweaning Piglets[J]. J Agric Food Chem. 2018, 66: 7899-7906. [116] Yang S, Zhang H, Sun F, et al. Metabolic Profile of Zearalenone in Liver Microsomes from Different Species and Its in Vivo Metabolism in Rats and Chickens Using Ultra High-Pressure Liquid Chromatography-Quadrupole/Time-of-Flight Mass Spectrometry[J]. J Agric Food Chem. 2017, 65: 11292-11303. [117] Yi Y, Gao K, Zhang L, et al. Zearalenone Induces MLKL-Dependent Necroptosis in Goat Endometrial Stromal Cells via the Calcium Overload/ROS Pathway[J]. Int J Mol Sci. 2022, 23: 10170. [118] Yi Y, Wan S, Hou Y, et al. Chlorogenic acid rescues zearalenone induced injury to mouse ovarian granulosa cells[J]. Ecotoxicol Environ Saf. 2020, 194: 110401. [119] Yiew N K H, Finck B N. The mitochondrial pyruvate carrier at the crossroads of intermediary metabolism[J]. Am J Physiol Endocrinol Metab. 2022, 323: E33-e52. [120] Zhang C, Li C, Liu K, et al. Characterization of zearalenone-induced hepatotoxicity and its mechanisms by transcriptomics in zebrafish model[J]. Chemosphere. 2022, 309: 136637. [121] Zhang G, Zeng C, Sun X, et al. Zearalenone modulates the function of goat endometrial cells via the mitochondrial quality control system[J]. Faseb j. 2024, 38: e23701. [122] Zhang G L, Sun X F, Feng Y Z, et al. Zearalenone exposure impairs ovarian primordial follicle formation via down-regulation of Lhx8 expression in vitro[J]. Toxicol Appl Pharmacol. 2017, 317: 33-40. [123] Zhang K, Li H, Song Z. Membrane depolarization activates the mitochondrial protease OMA1 by stimulating self-cleavage[J]. EMBO Rep. 2014, 15: 576-585. [124] Zhao L, Zhang L, Xu Z, et al. Occurrence of Aflatoxin B(1), deoxynivalenol and zearalenone in feeds in China during 2018-2020[J]. J Anim Sci Biotechnol. 2021, 12: 74. [125] Zhao Y, Pan Y, Chen M, et al. PKM2 interacts with and phosphorylates PHB2 to sustain mitochondrial quality control against septic cerebral-cardiac injury[J]. Int J Med Sci. 2024, 21: 633-643. [126] Zheng W, Feng N, Wang Y, et al. Effects of zearalenone and its derivatives on the synthesis and secretion of mammalian sex steroid hormones: A review[J]. Food Chem Toxicol. 2019, 126: 262-276. [127] Zheng W, Pan S, Wang G, et al. Zearalenone impairs the male reproductive system functions via inducing structural and functional alterations of sertoli cells[J]. Environ Toxicol Pharmacol. 2016, 42: 146-155. [128] Zhou M, Yang L, Shao M, et al. Effects of Zearalenone Exposure on the TGF-β1/Smad3 Signaling Pathway and the Expression of Proliferation or Apoptosis Related Genes of Post-Weaning Gilts[J]. Toxins (Basel). 2018a, 10: 49. [129] Zhou Z, Moore T M, Drew B G, et al. Estrogen receptor α controls metabolism in white and brown adipocytes by regulating Polg1 and mitochondrial remodeling[J]. Sci Transl Med. 2020, 12: eaax8096. [130] Zhou Z, Ribas V, Rajbhandari P, et al. Estrogen receptor α protects pancreatic β-cells from apoptosis by preserving mitochondrial function and suppressing endoplasmic reticulum stress[J]. J Biol Chem. 2018b, 293: 4735-4751. [131] Zhu L, Yuan H, Guo C, et al. Zearalenone induces apoptosis and necrosis in porcine granulosa cells via a caspase-3- and caspase-9-dependent mitochondrial signaling pathway[J]. J Cell Physiol. 2012, 227: 1814-1820. |
中图分类号: | S85 |
开放日期: | 2027-06-11 |