中文题名: | 杂色曲霉素和桔青霉素的细胞毒性和对脂质代谢影响的研究 |
姓名: | |
学号: | 2018108035 |
保密级别: | 公开 |
论文语种: | chi |
学科代码: | 083201 |
学科名称: | 工学 - 食品科学与工程(可授工学、农学学位) - 食品科学 |
学生类型: | 硕士 |
学位: | 工程硕士 |
学校: | 南京农业大学 |
院系: | |
专业: | |
研究方向: | 分子营养与人类健康 |
第一导师姓名: | |
第一导师单位: | |
完成日期: | 2021-06-01 |
答辩日期: | 2021-06-01 |
外文题名: | The study on cytotoxicity and lipid metabolism of sterigmatocystin and citrinin |
中文关键词: | |
外文关键词: | Sterigmatocystin ; Citrinin ; Cytotoxicity ; Lipidomics ; Lipid metabolism |
中文摘要: |
食品安全问题是全球最关注的问题之一,多种食品都很有可能受到真菌的污染。人类主要是通过霉变的食品而接触到真菌毒素。杂色曲霉素(Sterigmatocystin, STC)和桔青霉素(Citrinin, CTN)作为由青霉和曲霉产生的真菌毒素,可广泛共存于霉变的食品中。研究表明,STC具有潜在的肝毒性和致癌性,而CTN可以导致体外和体内的肝毒性。事实上,肝脏是真菌毒素和脂质代谢的主要代谢器官,STC和CTN都与肝毒性相关,然而,STC和CTN的细胞毒性和对脂质代谢的潜在影响仍需进一步研究。本文探究STC和CTN对人肝细胞HL-7702细胞毒性的影响,利用基于HPLC-MS的非靶向脂质组学探究STC和CTN对内源性脂质代谢物的影响,并进一步地探究STC和CTN如何影响肝细胞的脂质代谢。主要的研究内容和结果如下: STC和CTN均剂量依赖性地降低细胞活力,和增加细胞内ROS水平。此外, STC可以触发细胞周期相关基因(如,周期蛋白依赖性激酶2和周期蛋白依赖性激酶4(Cyclin-dependent kinase 2 and Cyclin-dependent kinase 4, CDK2和CDK4)以及细胞周期蛋白依赖性激酶抑制因子(Cyclin dependent kinase inhibitors, CKIs)中的p15INK4b、p16INK4a 、p21Cip1、p27Kip1和p57Kip2(简称p15、p16、p21、p27和 p57))的异常表达,而CTN则导致细胞周期(周期蛋白依赖性激酶1 (Cyclin-dependent kinase 1, CDK1)、p21和p27) 的异常表达。另外,STC和CTN均能时间依赖性地诱导细胞凋亡,并在细胞凋亡的基因水平上得到了验证(BCL2家族蛋白(BCL-2 family proteins, BCL2, Bax, BAK)、半胱天冬氨酸蛋白酶3(Caspase3) 和半胱天冬氨酸蛋白酶3(Caspase8))。 脂质组学的多元统计分析表明,STC和CTN处理后,细胞内脂质代谢物均发生了显著性的变化。STC处理后内源脂质代谢产物,如溶血卵磷脂(Lysophosphatidylcholine, LPC)、溶血磷脂酰乙醇胺( Lysophosphatidylethanolamine, LPE)、溶血磷脂酰(Lysophosphatidylglycerol, LPG)、溶血磷脂酰肌醇(Lysophosphatidylinositol, LPI)、磷脂酸(Phosphatidic acid, PA)、神经酰胺(Ceramides, Cer)、葡糖鞘氨醇(Glucosylsphingosine, CerG2GNAc1)、葡糖鞘氨醇(Glucosylsphingosine, CerG2)、葡糖鞘氨醇(Glucosylsphingosine, CerG3)、胆甾醇酯(Cholesterol ester, ChE)和脂肪酸(Fatty acid, FA)含量降低,而甘油三酯(Triglyceride, TG)和酰基肉碱(Acyl carnitine, AcCa)含量升高;CTN处理后内源脂质代谢产物中LPC、磷脂酰肌醇(Phosphatidylinositol (4,5) bisphosphate, PIP2)、PA、CerG2、CerG2GNAc1和FA含量明显降低。此外,STC处理组中代表性的内源脂质代谢分子,如LPC (16:0)、LPC (16:1)、LPC (18:0)、LPC (18:1)、LPC (18:2)、LPE (16:0)和FA (20:4) 也显著性地下降,而TG (16:0/16:1/16:1) 和 TG (18:1/18:1/18:2)的表达量显著性升高; CTN组中的LPC (16:0)、LPC (16:1)、LPC (17:0)、LPC (18:0)、LPC (18:1)、LPC (18:2)和FA (20:4)也明显地下降。 此外,在基因水平上,STC和CTN均能降低FA合成基因的表达,包括长链酰基-辅酶A合成酶1(Long chain acyl-CoA synthetases1, ACSL1)、脂肪酸去饱和酶1(Fatty acid desaturase1, FADS1 )、长链脂肪酸延长酶5(Long-chain fatty acid elongase 5, ELOVL5)、脂肪酸合酶(Fatty acid synthase, FASN)和固醇调控元件结合蛋白1c(Sterol regulatory element-binding proteins 1c, SREBP1c)。而STC还能增强FA降解相关基因的表达,包括过氧化物酶体增殖物激活受体α(Peroxisome proliferator-activated receptor alpha, PPARα)和肉碱棕榈酰转移酶1α(Carnitine palmitoyl transferase 1al pha, CPT1α),从而降低细胞内FA含量。 综上所述,STC和CTN均能抑制细胞增殖,促进ROS积累,导致细胞周期积聚,诱导细胞凋亡。STC和CTN均导致细胞内脂质代谢的紊乱。STC和CTN都能引起细胞内LPC和FA含量的降低,包括脂质分子 LPC (16:0)、LPC (16:1)、LPC(18:0)、LPC(18:1)、LPC(18:2)和FA(20:4)含量的下降。此外,STC还能诱导细胞内LPE、Cer的降低和TC的升高,以及脂质分子LPE(16:0)的降低、TG (16:0/16:1/16:1) 和 TG (18:1/18:1/18:2)的增强;而CTN还能导致细胞内LPC (17:0)的降低。在基因水平上,STC能通过减弱FA的合成和增强FA的分解而降低细胞内FA的含量,而CTN则主要降低FA的合成而降低细胞内FA的含量。因此,本研究对内源性脂质代谢产物的研究为探究STC和CTN的细胞毒性和对脂质代谢的影响提供了新的见解,从而为进一步研究STC和CTN的细胞毒性和脂质代谢的作用机制奠定基础。 |
外文摘要: |
Food safety is one of the most concerned issues in the world, and many kinds of food are likely to be contaminated by fungi. Humans are exposed to mycotoxins mainly through moldy foods. Sterigmatocystin (STC) and Citrinin (CTN), as mycotoxins produced by Penicillium and Aspergillus, could coexist widely in mildewy foods. Studies had shown that STC have potential hepatotoxicity and carcinogenicity, while CTN could cause hepatotoxicity in vitro and in vivo. The liver is the main metabolic organ for mycotoxin and lipid metabolism, and both STC and CTN are associated with hepatotoxicity. However, the cytotoxicity of STC and CTN and their potential effects on lipid metabolism need further research. Therefore, the effects of STC and CTN on the cytotoxicity of human hepatocyte HL-7702 cells were explored, and the effects of STC and CTN on endogenous lipid metabolites were explored using HPLC-MS based untargeted lipidomics, how STC and CTN affect lipid metabolism of hepatocytes was further explored. The main research contents and results are as follows: STC and CTN all reduced cell viability in a dose-dependent manner, and increased intracellular ROS levels. Additionally, ST C triggered the abnormal expression of cell cycle-related genes, including Cyclin-dependent kinase 2 and Cyclin-dependent kinase 4 (CDK2 and CDK4), and p15INK4b, p16INK4a, p21Cip1, p27Kip1 and p57Kip2 (p15, p16, p21, p27 and p57) of cyclin dependent kinase inhibitors (CKIs), while CTN caused the abnormal expression of cell cycle-related genes (Cyclin-dependent kinase 1 (CDK1), p21 and p27). In addition, STC and CTN all induced caused apoptosis in a time-dependent manner and have been verified at the levels of apoptosis-related genes (BCL-2 family proteins (BCL2, Bax and BAK), Caspase3 and Caspase8). Multivariate statistical analysis of lipidomics showed that the endogenous lipid metabolites changed significantly after STC and CTN treatment. The contents of endogenous lipid metabolites decreased , such as lysophosphatidylcholine (LPC), lysophosphatidylethanolamine (LPE), lysophosphatidylglycerol (LPG), lysophosphatidylinositol (Lysophosphatidylinositol, LPI), Phosphatidic acid (PA), Ceramides (Cer), Glucosylsphingosine (CerG2GNAc1), Glucosylsphingosine (CerG2), Glucosylsphingosine (Glucosylsphingosine, CerG3), Cholesterol ester (ChE) and fatty acid (FA) , whereas the contents of triglyceride (TG) and acyl carnitine (AcCa) increased after STC administration. The contents of LPC, Phosphatidylinositol (4,5) bisphosphate (PIP2), PA, CerG2, CerG2, GNAc1 and FA were also significantly reduced after CTN treatment. As the representative endogenous lipid metabolism molecules, such as LPC (16:0), LPC (16:1), LPC (18:0), LPC (18:1), LPC (18:2), LPE (16:0) and FA (20:4) the contents of these decreased significantly after STC treatment, while TG (16:0/16:1/16:1) and TG (18:1/18:1/18:2) increased dramatically. And the contents of LPC (16:0), LPC (16:1), LPC (17:0), LPC (18:0), LPC (18:1), LPC (18:2) and FA (20:4) also dramatically dropped after CTN-treated groups. In addition, STC and CTN all reduced the expression of synthesis of FA, including long chain acyl-CoA synthetases1 (ACSL1), fatty acid desaturase1 (FADS1), long-chain fatty acid elongase 5 (ELOVL5), fatty acid synthase (FASN), sterol regulatory element-binding proteins 1c (SREBP1c). STC also enhanced the degradation of FA (Peroxisome proliferator-activated receptor alpha, PPARα and Carnitine palmitoyl transferase 1alpha, CPT1α) at the genetic level to reduce the intracellular contents of FA. In summary, STC and CTN all inhibited cell proliferation, promoted ROS accumulation, led to cell cycle accumulation, and induced cell apoptosis. Both STC and CTN disturbed intracellular lipid metabolism. STC and CTN caused the reduction of intracellular LPC and FA, and decreased the contents of LPC (16:0), LPC (16:1), LPC (18:0), LPC (18: 1), LPC (18:2) and FA (20:4). Moreover, STC induced the reducing concentration of intracellular LPE and Cer, and enhancement of intracellular TG, as well as the reduction of LPE (16:0), and the increase of TG (16:0/16:1/16:1) and TG (18:1/18:1/18:2), while CTN declined the contents of intracellular LPC (17:0). In addition, STC reduced the contents of FA by weakening FA synthesis and enhancing the FA degradation, while CTN by reducing the synthesis of FA in the genetic level. Therefore, the study of endogenous lipid metabolites in this study provides new insights for exploring the cytotoxicity of STC and CTN and their effects on lipid metabolism, which lay a foundation for further study on the mechanism of cytotoxicity and lipid metabolism of STC and C TN. |
参考文献: |
[ 1 ] Cabaret O, Puel O, Botterel F, et al. Contribution of uniformly 13C-enriched sterigmatocystin to the study of its pulmonary metabolism [J]. Rapid Commun Mass Spectrom, 2011, 25(19): 2704-2710 [ 2 ] Jahanian E. Mycotoxin-induced toxicity; an updated mini-review on the current concepts [J]. Immunopathologia Persa, 2016, 2(2): [ 3 ] A JP, B JF, B KM. Mycotoxins in food and feed [J]. Advances in Food and Nutrition Research, 2019, 89: 297-345 [ 4 ] Bennett JW, Klich M. Mycotoxins [J]. Clinical Microbiology Reviews, 2003, 16(3): 497-516 [ 5 ] Lger TG, Uar A, Akrolu FP, et al. Genotoxic effects of mycotoxins [J]. Toxicon, 2020: [ 6 ] Versilovskis A, De Saeger S. Sterigmatocystin: occurrence in foodstuffs and analytical methods-an overview [J]. Molecular nutrition & food research, 2010, 54(1): 136-147 [ 7 ] Diaz Nieto CH, Granero AM, Zon MA, et al. Sterigmatocystin: A mycotoxin to be seriously considered [J]. Food Chem Toxicol, 2018, 118: 460-470 [ 8 ] Septien I, Cutuli M, Garcia M, et al. Solubility and stability of sterigmatocystin in different organic solvents [J]. Toxicon : official journal of the International Society on Toxinology, 1993, 31(10): 1337-1340 [ 9 ] Zingales V, Fernandez-Franzon M, Ruiz MJ. Sterigmatocystin: Occurrence, toxicity and molecular mechanisms of action - A review [J]. Food Chem Toxicol, 2020, 146: 111802 [ 10 ] Gruber-Dorninger C, Novak B, Nagl V, et al. Emerging Mycotoxins: Beyond Traditionally Determined Food Contaminants [J]. Journal of Agricultural & Food Chemistry, 2016: 7052 [ 11 ] Rank C, Nielsen KF, Larsen TO, et al. Distribution of sterigmatocystin in filamentous fungi [J]. Fungal Biol, 2011, 115(4-5): 406-420 [ 12 ] Steffen E, Annette L, Dirk S, et al. Occurrence of toxigenic Aspergillus versicolor isolates and sterigmatocystin in carpet dust from damp indoor environments [J]. Applied and environmental microbiology, 2002, 68(8): [ 13 ] T T, K R, T J, et al. Mycotoxins in crude building materials from water-damaged buildings [J]. Applied and environmental microbiology, 2000, 66(5): [ 14 ] Ver?ilovskis A, Saeger SD. Sterigmatocystin: Occurrence in foodstuffs and analytical methods – An overview [J]. Molecular Nutrition & Food Research, 2010, 54(1): [ 15 ] Nieto CHD, Granero AM, Zon MA, et al. Sterigmatocystin: A mycotoxin to be seriously considered [J]. Food and Chemical Toxicology, 2018, 118: [ 16 ] Keller, Nancy, P., et al. Requirement of monooxygenase-mediated steps for sterigmatocystin biosynthesis by Aspergillus [J]. Applied & Environmental Microbiology, 2000, 66(1): 359-359 [ 17 ] Zingales V, Fernández-Franzón M, Ruiz M-J. Sterigmatocystin: Occurrence, toxicity and molecular mechanisms of action – A review [J]. Food and Chemical Toxicology, 146: [ 18 ] N WG. Aflatoxins as risk factors for hepatocellular carcinoma in humans [J]. Cancer research, 1992, 52(7 Suppl): [ 19 ] Wei G, Liping J, Lan G, et al. Sterigmatocystin-induced oxidative DNA damage in human liver-derived cell line through lysosomal damage [J]. Toxicology in vitro : an international journal published in association with BIBRA, 2015, 29(1): [ 20 ] Yang L, Ming D, Genyi Z. Proapoptotic activity of aflatoxin B1 and sterigmatocystin in HepG2 cells [J]. Toxicology reports, 2014, 1: [ 21 ] Wang JS, Groopman JD. DNA damage by mycotoxins [J]. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis, 1999, 424(1-2): 167-181 [ 22 ] Scientific Opinion on the risk for public and animal health related to the presence of sterigmatocystin in food and feed [J]. EFSA Journal, 2013, 11(6): [ 23 ] Zouaoui N, Mallebrera B, Berrada H, et al. Cytotoxic effects induced by patulin, sterigmatocystin and beauvericin on CHO–K1 cells [J]. Food & Chemical Toxicology, 2016: [ 24 ] Gao W, Jiang L, Ge L, et al. Sterigmatocystin-induced oxidative DNA damage in human liver-derived cell line through lysosomal damage [J]. Toxicology in Vitro, 2015, 29(1): 1-7 [ 25 ] Bünger J, Westphal G, Mnnich A, et al. Cytotoxicity of occupationally and environmentally relevant mycotoxins [J]. Toxicology, 2004, 202(3): 199-211 [ 26 ] R S. Induction of DNA single-strand breaks and DNA synthesis inhibition in CHO and AWRF cells after exposure to sterigmatocystin and penicillic acid [J]. Folia Biologica, 1986, 32(6): 406-413 [ 27 ] Sterigmatocystin induces G1 arrest in primary human esophageal epithelial cells but induces G2 arrest in immortalized cells: key mechanistic differences in these two models [J]. Archives of Toxicology, 2015, 89(11): 2015-2025 [ 28 ] Sun X, Zhang X, Wang H, et al. Effects of sterigmatocystin, deoxynivalenol and aflatoxin G1 on apoptosis of human peripheral blood lymphocytes in vitro [J]. Biomedical and environmental sciences : BES, 2002, 15(2): 145-152 [ 29 ] Dubravka R, Daniela J, Andrea HT, et al. Sterigmatocystin moderately induces oxidative stress in male Wistar rats after short-term oral treatment [J]. Mycotoxin Res, 2020, 36(2): 181-191 [ 30 ] Sterigmatocystin moderately induces oxidative stress in male Wistar rats after short-term oral treatment [J]. Mycotoxin Research, 2020, 36(2): 181-191 [ 31 ] Purchase I, van der Watt J. Acute toxicity of sterigmatocystin to rats [J]. Food and cosmetics toxicology, 1969, 7(2): 135-139 [ 32 ] Fujii K, Kurata H, Odashima S, et al. Tumor induction by a single subcutaneous injection of sterigmatocystin in newborn mice [J]. Cancer research, 1976, 36(5): 1615-1618 [ 33 ] Ueda N, Fujie K, Gotoh-Mimura K, et al. Acute cytogenetic effect of sterigmatocystin on rat bone-marrow cells in vivo [J]. Mutation research, 1984, 139(4): 203-206 [ 34 ] Abdel-Wahhab M, Hasan A, Aly S, et al. Adsorption of sterigmatocystin by montmorillonite and inhibition of its genotoxicity in the Nile tilapia fish (Oreachromis nilaticus) [J]. Mutation research, 2005, 582: 20-27 [ 35 ] Schroeder HW, Kelton WH. Production of Sterigmatocystin by Some Species of the Genus Aspergillus and Its Toxicity to Chicken Embryos [J]. Appl Microbiol, 1975, 30(4): 589-591 [ 36 ] Purchase IFH, Watt JJVD. The Acute and Chronic Toxicity of Sterigmatocystin [J]. [ 37 ] Kovalenko AV, Soldatenko NA, Fetisov LN, et al. More accurate determination of the minimum allowable level of sterigmatocystin in piglet feed [J]. Russian Agricultural Sciences, 2011, 37(6): 504-507 [ 38 ] Vesonder R, Horn B. Sterigmatocystin in dairy cattle feed contaminated with Aspergillus versicolor [J]. Applied and environmental microbiology, 1985, 49(1): 234-235 [ 39 ] 张祥宏, 王凤荣, 王俊灵,等. 真菌及其毒素诱发肺癌的动物实验研究 [J]. 北京大学学报(医学版), 2003, (01): 4-6 [ 40 ] 楼建龙,田禾菁,孟昭赫,郭振泉. 胃癌、肝癌高发区和低发区粮食中杂色曲霉素污染量调查 [J]. 卫生研究, 1995, (01): 28-31 [ 41 ] Hu?ana?u C, Sfarti C, Trifan A, et al. [High levels of sterigmatocystin in patients with chronic liver diseases] [J]. Clinica Chimica Acta, 1980, 108(1): 121-128 [ 42 ] Xiaoqin, Cao, Xiaofei, et al. Quantitative determination of carcinogenic mycotoxins in human and animal biological matrices and animal-derived foods using multi-mycotoxin and analyte-specific high performance liquid chromatography-tandem mass spectrometric methods [J]. Journal of Chromatography B Analytical Technologies in the Biomedical & Life Sciences, 2018: [ 43 ] Zsolt R, László S, Anett M, et al. Toxicological and Medical Aspects of Aspergillus-Derived Mycotoxins Entering the Feed and Food Chain [J]. Frontiers in Microbiology, 2020: [ 44 ] H T, J L, C D. [Determination of sterigmatocystin in cancerous tissues, blood and urine in patients with liver and stomach cancer] [J]. Zhonghua yu fang yi xue za zhi [Chinese journal of preventive medicine], 1995, 29(5): [ 45 ] 刘仁荣, 许杨. 桔青霉素简介及其免疫学检测方法研究进展 [J]. 卫生研究, 2004, (01): 124-127 [ 46 ] Naofumi, Kitabatake, Alka, et al. Thermal decomposition and detoxification of citrinin under various moisture conditions [J]. Journal of Agricultural and Food Chemistry, 1991: [ 47 ] Xu BJ, Jia XQ, Gu LJ, et al. Review on the qualitative and quantitative analysis of the mycotoxin citrinin [J]. Food Control, 2006, 17(4): 271-285 [ 48 ] Shu PY, Lin CH. Simple and sensitive determination of citrinin in Monascus by GC-selected ion monitoring mass spectrometry [J]. Analytical Sciences, 2002, 18(3): 283 [ 49 ] Hetherington AC, Raistrick H. Studies in the Biochemistry of Micro-organisms. Part XIV.—On the production and chemical constitution of a new yellow colouring mater, citrinin, produced from glucose by Penicillium [J]. Philosophical Transactions of the Royal Society of London, 1931, 220(2): 269-295 [ 50 ] El-Banna AA, Pitt JI, Leistner L. Production of Mycotoxins by Penicillium Species [J]. Systematic & Applied Microbiology, 1987, 10(1): 42-46 [ 51 ] Blanc PJ, Laussac JP, Bars JL, et al. Charactcrization of Monascidin A from Monascus as Citrinin. International [J]. International Journal of Food Microbiology, 1995, 27(2-3): 201-213 [ 52 ] Filho JWGdO, Islam MT, Ali ES, et al. A comprehensive review on biological properties of citrinin [J]. Food and Chemical Toxicology, 2017, 110(2017): 130-141 [ 53 ] Kiebooms JAL, Huybrechts B, Thiry C, et al. A quantitative UHPLC-MS/MS method for citrinin and ochratoxin A detection in food, feed and red yeast rice food supplements [J]. World Mycotoxin Journal, 2016, 9(3): 1-10 [ 54 ] Liao CD, Chen YC, Lin HY, et al. Incidence of citrinin in red yeast rice and various commercial Monascus products in Taiwan from 2009 to 2012 [J]. Food Control, 2014, 38: 178-183 [ 55 ] Hepatotoxic effect of ochratoxin A and citrinin, alone and in combination, and protective effect of vitamin E: Invitro study in HepG2 cell [J]. Food & Chemical Toxicology, 2015, 83: 151-163 [ 56 ] None. Scientific Opinion on the risks for public and animal health related to the presence of citrinin in food and feed [J]. EFSA Journal, 2012, 10(3): [ 57 ] Speijers GJA, Speijers MHM. Combined toxic effects of mycotoxins [J]. Toxicology Letters, 2004, 153(1): 91-98 [ 58 ] Macholz R. Some Naturally Occurring and Synthetic Food Components, Furocoumarins and Ultraviolet Radiation. IARC Monographs on the Evaluation of the Carcinogenic Risk of Chemicals to Humans. Vol. 40. 444 Seiten. WHO, International Agency for Research on Cancer, Lyon 1986. Preis: 65,—Sw. fr.; 39,— US $ [J]. Food / Nahrung, 1988, 32(2): [ 59 ] Jeswal P, Botany UDo, ) B. Cumulative effect of ochratoxin A and citrinin on induction of hepatorenal carcinogenesis in mice (Mus musculus) [J]. Biomedical Letters, 1995: [ 60 ] F?Llmann W, Behm C, Degen GH. Toxicity of the mycotoxin citrinin and its metabolite dihydrocitrinone and of mixtures of citrinin and ochratoxin A in vitro [J]. Archives of Toxicology, 2014, 88(5): 1097-1107 [ 61 ] Doi K, Uetsuka K. Mechanisms of Mycotoxin-induced Dermal Toxicity and Tumorigenesis Through Oxidative Stress-related Pathways [J]. Journal of Toxicologic Pathology, 2014, 27(1): 1 [ 62 ] Chen CC, Chan WH. Inhibition of Citrinin-Induced Apoptotic Biochemical Signaling in Human Hepatoma G2 Cells by Resveratrol [J]. International Journal of Molecular Sciences, 2009, 10(8): 3338-3357 [ 63 ] Sharath BGR, Tamatam A, Ilaiyaraja N, et al. Pelargonidin Modulates Keap1/Nrf2 Pathway Gene Expression and Ameliorates Citrinin-Induced Oxidative Stress in HepG2 Cells [J]. Frontiers in Pharmacology, 2017, 8: 868- [ 64 ] Knasmüller S, Mersch-Sundermann V, Kevekordes S, et al. Use of human-derived liver cell lines for the detection of environmental and dietary genotoxicants; current state of knowledge [J]. Toxicology, 2004, 198(1-3): 315-328 [ 65 ] Creppy EE, Lorkowski G, Beck G, et al. Combined action of citrinin and ochratoxin A on hepatoma tissue culture cells [J]. Toxicology Letters, 1980, 5(6): 375-380 [ 66 ] Yu FY, Liao YC, Chang CH, et al. Citrinin induces apoptosis in HL-60 cells via activation of the mitochondrial pathway [J]. Toxicology Letters, 2006, 161(2): 143-151 [ 67 ] Chan, Wen-Hsiung. Citrinin induces apoptosis via a mitochondria-dependent pathway and inhibition of survival signals in embryonic stem cells, and causes developmental injury in blastocysts [J]. Biochemical Journal, 2007, 404(2): 317-326 [ 68 ] Ambrose AM, DeEds F. Acute and Subacute Toxicity of Pure Gitrinin [J]. Proceedings of the Society for Experimental Biology and Medicine, 1945, 59(2): [ 69 ] SAKAI, Fuminori. An experimental study on the toxic effect especially on the kidney of "Yellowed Rice" polluted by Penicillium citrinum Thom., as well as of Citrinin, a pigment isolated from the mould [J]. Folia Pharmacologica Japonica, 1955, 51(4): 431-442_431,en427 [ 70 ] Gupta M, Sasmal D, Bandyopadhyay S, et al. Hematological changes produced in mice by ochratoxin A and citrinin [J]. Toxicology, 1983, 26(1): 55-62 [ 71 ] Reddy RV, Mayura K, Hayes AW, et al. Embryocidal, teratogenic and fetotoxic effects of citrinin in rats [J]. Toxicology, 1982, 25(2-3): 151-160 [ 72 ] Krogh P, Hasselager E, Friis P. Studies on fungal nephrotoxicity. 2. Isolation of two nephrotoxic compounds from Penicillium viridicatum Westling: citrinin and oxalic acid [J]. Acta pathologica et microbiologica Scandinavica Section B: Microbiology and immunology, 1970, 78(4): 401-413 [ 73 ] Ambrose AM, Deeds F. Some toxicological and pharmacological properties of citrinin [J]. Journal of Pharmacology & Experimental Therapeutics, 1946, 88(2): 173 [ 74 ] Friis P, Hasselager E, Krogh P. ISOLATION OF CITRININ AND OXALIC ACID FROM PENICILLIUM VIRIDICATUM WESTLING AND THEIR NEPHROTOXICITY IN RATS AND PIGS [J]. Acta Pathologica Microbiologica Scandinavica, 1969, 77(3): [ 75 ] Chagas GM, Oliveira MBM, Campello AP, et al. Mechanism of citrinin‐induced dysfunction of mitochondria. II. Effect on respiration, enzyme activities, and membrane potential of liver mitochondria [J]. Cell Biochemistry & Function, 2010, 10(3): 209-216 [ 76 ] Lurá MC, Fuentes M, Cabagna M, et al. Structural and ultrastructural alterations in BALB/c mice: Effects of Penicillium citrinum metabolites [J]. Mycopathologia, 2004, 158(2): 233-238 [ 77 ] Ali N, Degen GH. Citrinin biomarkers: a review of recent data and application to human exposure assessment [J]. Arch Toxicol, 2019, 93(11): 3057-3066 [ 78 ] Wolfram, F?llmann, Claudia, et al. Toxicity of the mycotoxin citrinin and its metabolite dihydrocitrinone and of mixtures of citrinin and ochratoxin A in vitro [J]. Archives of Toxicology, 2014: [ 79 ] Ali N, Degen GH. Citrinin biomarkers: a review of recent data and application to human exposure assessment [J]. Archives of Toxicology, 2019, 93(11): 3057-3066 [ 80 ] Ali N, Blaszkewicz M, Manirujjama M, et al. Biomonitoring of concurrent exposure to ochratoxin A and citrinin in pregnant women in Bangladesh [J]. Mycotoxin Res, 2016, 32(3): [ 81 ] Kalish BT, Fell GL, Nandivada P, et al. Clinically Relevant Mechanisms of Lipid Synthesis, Transport, and Storage [J]. JPEN J Parenter Enteral Nutr, 2015, 39(1 Suppl): 8S-17S [ 82 ] Nguyen P, Leray V, Diez M, et al. Liver lipid metabolism [J]. J Anim Physiol Anim Nutr (Berl), 2008, 92(3): 272-283 [ 83 ] Hodson L, Gunn PJ. The regulation of hepatic fatty acid synthesis and partitioning: the effect of nutritional state [J]. Nat Rev Endocrinol, 2019, 15(12): 689-700 [ 84 ] Schwartz MW, Seeley RJ, Tschp MH, et al. Cooperation between brain and islet in glucose homeostasis and diabetes [J]. Nature, 2018, 2013年503卷7474期(7474): 59-66页 [ 85 ] Goldstein JL, Brown MS. A Century of Cholesterol and Coronaries: From Plaques to Genes to Statins [J]. Cell, 2015, 161(1): 161-172 [ 86 ] Rosen E, Spiegelman B. What we talk about when we talk about fat [J]. Cell, 2014, 156: 20-44 [ 87 ] Perry RJ, Samuel VT, Petersen KF, et al. The role of hepatic lipids in hepatic insulin resistance and type 2 diabetes [J]. Nature, 2014, 510(7503): 84-91 [ 88 ] Cohen JC, Horton JD, Hobbs HH. Human Fatty Liver Disease: Old Questions and New Insights [J]. Science, 2011, 332(6037): 1519-1523 [ 89 ] Poitelon Y, Kopec AM, Belin S. Myelin Fat Facts: An Overview of Lipids and Fatty Acid Metabolism [J]. Cells, 2020, 9(4): [ 90 ] Grevengoed TJ, Klett EL, Coleman RA. Acyl-CoA metabolism and partitioning [J]. Annu Rev Nutr, 2014, 34: 1-30 [ 91 ] Kanter JE, Tang C, Oram JF, et al. Acyl-CoA synthetase 1 is required for oleate and linoleate mediated inhibition of cholesterol efflux through ATP-binding cassette transporter A1 in macrophages [J]. Biochim Biophys Acta, 2012, 1821(3): 358-364 [ 92 ] Tripathy S, Jump DB. Elovl5 regulates the mTORC2-Akt-FOXO1 pathway by controlling hepatic cis-vaccenic acid synthesis in diet-induced obese mice [J]. J Lipid Res, 2013, 54(1): 71-84 [ 93 ] Lee H, Park WJ. Unsaturated fatty acids, desaturases, and human health [J]. J Med Food, 2014, 17(2): 189-197 [ 94 ] R?hrig F, Schulze A. The multifaceted roles of fatty acid synthesis in cancer [J]. Nature Reviews Cancer, 2016, 16(11): 732-749 [ 95 ] Jones SF, Infante JR. Molecular Pathways: Fatty Acid Synthase [J]. Clinical Cancer Research An Official Journal of the American Association for Cancer Research, 2015, 21(24): 5434 [ 96 ] Cheng C, Geng F, Cheng X, et al. Lipid metabolism reprogramming and its potential targets in cancer [J]. Cancer Commun (Lond), 2018, 38(1): 27 [ 97 ] Bradbury MW. Lipid Metabolism and Liver Inflammation. I. Hepatic fatty acid uptake: possible role in steatosis [J]. American Journal of Physiology Gastrointestinal & Liver Physiology, 2006, 290(2): G194 [ 98 ] Kawano Y, Cohen DE. Mechanisms of hepatic triglyceride accumulation in non-alcoholic fatty liver disease [J]. Journal of Gastroenterology, 2013, 48(4): 434-441 [ 99 ] Chang X, Wang Z, Zhang J, et al. Lipid profiling of the therapeutic effects of berberine in patients with nonalcoholic fatty liver disease [J]. Journal of translational medicine, 2016, 14: 266 [ 100 ] Zechner R, Zimmermann R, Eichmann TO, et al. FAT SIGNALS-lipases and lipolysis in lipid metabolism and signaling [J]. Cell Metab, 2012, 15(3): 279-291 [ 101 ] Schweiger M, Schreiber R, Haemmerle G, et al. Adipose Triglyceride Lipase and Hormone-sensitive Lipase Are the Major Enzymes in Adipose Tissue Triacylglycerol Catabolism [J]. Journal of Biological Chemistry, 2006, 281(52): 40236-40241 [ 102 ] Houten SM, Violante S, Ventura FV, et al. The Biochemistry and Physiology of Mitochondrial Fatty Acid β-Oxidation and Its Genetic Disorders [J]. Annu Rev Physiol, 2016, 78: 23-44 [ 103 ] Bougarne N, Weyers B, Desmet SJ, et al. Molecular Actions of PPARalpha in Lipid Metabolism and Inflammation [J]. Endocr Rev, 2018, 39(5): 760-802 [ 104 ] Muoio DM, Way JM, Tanner CJ, et al. Peroxisome Proliferator-Activated Receptor-α Regulates Fatty Acid Utilization in Primary Human Skeletal Muscle Cells [J]. Diabetes, 2002, 51(4): 901 [ 105 ] Sun WW, Shangguan T, Zhu P, et al. Role of hepatic neuropeptide Y-Y1 receptors in a methionine-choline-deficient model of non-alcoholic steatohepatitis [J]. Life Sciences, 2020, 245: 117356 [ 106 ] Ojuka E, Andrew B, Bezuidenhout N, et al. Measurement of β-oxidation capacity of biological samples by respirometry: a review of principles and substrates [J]. Am J Physiol Endocrinol Metab, 2016, 310(9): E715-723 [ 107 ] Han X, Gross RW. Global analyses of cellular lipidomes directly from crude extracts of biological samples by ESI mass spectrometry: A bridge to lipidomics [J]. Journal of Lipid Research, 2003, 44(6): 1071-1079 [ 108 ] Lagarde M, Gélo?N A, Record M, et al. Lipidomics is emerging [J]. Biochimica Et Biophysica Acta, 2003, 1634(3): 61-61 [ 109 ] Seon, Hwa, Lee, et al. Targeted lipidomics using electron capture atmospheric pressure chemical ionization mass spectrometry [J]. Rapid Communications in Mass Spectrometry, 2003, 17(19): 2168-2176 [ 110 ] Blanksby S, Mitchell T. Advances in mass spectrometry for lipidomics [J]. Annual review of analytical chemistry (Palo Alto, Calif), 2010, 3: 433-465 [ 111 ] Han X, Gross R. Global analyses of cellular lipidomes directly from crude extracts of biological samples by ESI mass spectrometry: a bridge to lipidomics [J]. Journal of lipid research, 2003, 44(6): 1071-1079 [ 112 ] Roberts L, Souza A, Gerszten R, et al. Targeted metabolomics [J]. Current protocols in molecular biology, 2012: Unit 30.32.31-24 [ 113 ] Cajka T, Fiehn O. Toward Merging Untargeted and Targeted Methods in Mass Spectrometry-Based Metabolomics and Lipidomics [J]. Anal Chem, 2016, 88(1): 524-545 [ 114 ] Lee HC, Yokomizo T. Applications of mass spectrometry-based targeted and non-targeted lipidomics [J]. Biochem Biophys Res Commun, 2018, 504(3): 576-581 [ 115 ] A LZ, A BZ, B YZ, et al. Clinical lipidomics in understanding of lung cancer: Opportunity and challenge [J]. Cancer Letters, 2020, 470: 75-83 [ 116 ] Tsugawa H, Arita M, Kanazawa M, et al. MRMPROBS: a data assessment and metabolite identification tool for large-scale multiple reaction monitoring based widely targeted metabolomics [J]. Analytical Chemistry, 2013, 85(10): 5191-5199 [ 117 ] Ismail IT, Showalter MR, Fiehn O. Inborn Errors of Metabolism in the Era of Untargeted Metabolomics and Lipidomics [J]. Metabolites, 2019, 9(10): [ 118 ] Lydic TA, Goo YH. Lipidomics unveils the complexity of the lipidome in metabolic diseases [J]. Clin Transl Med, 2018, 7(1): 4 [ 119 ] Hu T, Zhang JL. Mass-Spectrometry-Based Lipidomics [J]. Journal of Separation Science, 2017, 41(1): [ 120 ] Clugston R, Gao M, Blaner W. The Hepatic Lipidome: A Gateway to Understanding the Pathogenes is of Alcohol-Induced Fatty Liver [J]. Current molecular pharmacology, 2017, 10(3): 195-206 [ 121 ] Zhang C, Wang K, Yang L, et al. Lipid metabolism in inflammation-related diseases [J]. Analyst, 2018, 143(19): 4526-4536 [ 122 ] Han X. Lipidomics for studying metabolism [J]. Nat Rev Endocrinol, 2016, 12(11): 668-679 [ 123 ] Yang L, Li M, Shan Y, et al. Recent advances in lipidomics for disease research [J]. Journal of separation science, 2016, 39(1): 38-50 [ 124 ] Ten Hove M, Pater L, Storm G, et al. The hepatic lipidome: From basic science to clinical translation [J]. Advanced drug delivery reviews, 2020, 159: 180-197 [ 125 ] Gross RW, Han X. Lipidomics in Diabetes and the Metabolic Syndrome [J]. Methods in Enzymology, 2007, 433: 73 [ 126 ] Juergen G, Dominik S, Schwarz PEH, et al. Top-Down Lipidomics Reveals Ether Lipid Deficiency in Blood Plasma of Hypertensive Patients [J]. Plos One, 2009, 4(7): e6261 [ 127 ] Meikle PJ, Christopher MJ. Lipidomics is providing new insight into the metabolic syndrome and its sequelae [J]. Current Opinion in Lipidology, 2011, 22(3): 210-215 [ 128 ] Martel C, Esposti DD, Bouchet A, et al. Non-alcoholic steatohepatitis: new insights from OMICS studies [J]. Curr Pharm Biotechnol, 2012, 13(5): - [ 129 ] Züllig T, Trtzmüller M, Kfeler HC. Lipidomics from sample preparation to data analysis: a primer [J]. Analytical and Bioanalytical Chemistry, 2020, 412(10): 2191-2209 [ 130 ] Yang K, Han X. Lipidomics: Techniques, Applications, and Outcomes Related to Biomedical Sciences [J]. Trends Biochem Sci, 2016, 41(11): 954-969 [ 131 ] Wang M, Wang C, Han X. Selection of internal standards for accurate quantification of complex lipid species in biological extracts by electrospray ionization mass spectrometry—What, how and why? [J]. Mass Spectrometry Reviews, 2016: n/a-n/a [ 132 ] Keereetaweep J, Chapman KD. Lipidomic Analysis of Endocannabinoid Signaling: Targeted Metabolite Identification and Quantification [J]. Neural Plasticity, 2015, 2016(5): 2426398 [ 133 ] Han X, Rozen S, Boyle SH, et al. Metabolomics in Early Alzheimer's Disease: Identification of Altered Plasma Sphingolipidome Using Shotgun Lipidomics [J]. PLoS ONE, 2011, 6(7): e21643- [ 134 ] Merrill AH, Sullards MC, Allegood JC, et al. Sphingolipidomics: high-throughput, structure-specific, and quantitative analysis of sphingolipids by liquid chromatography tandem mass spectrometry [J]. Methods, 2005, 36(2): 207-224 [ 135 ] Jiang X, Cheng H, Yang K, et al. Alkaline methanolysis of lipid extracts extends shotgun lipidomics analyses to the low-abundance regime of cellular sphingolipids [J]. Analytical Biochemistry, 2007, 371(2): 135-145 [ 136 ] Wang M, Wang C, Han RH, et al. Novel Advances in Shotgun Lipidomics for Biology and Medicine [J]. other, 2016, 61: [ 137 ] Wang J, Wang C, Han X. Tutorial on lipidomics [J]. Anal Chim Acta, 2019, 1061: 28-41 [ 138 ] Xianlin, Han, Kui, et al. Multi-dimensional mass spectrometry-based shotgun lipidomics and novel strategies for lipidomic analyses [J]. Mass Spectrometry Reviews, 2011, 31(1): 134-178 [ 139 ] Junot C, Fenaille FO, Colsch B, et al. High resolution mass spectrometry based techniques at the crossroads of metabolic pathways [J]. Mass Spectrometry Reviews, 2015, 33(6): 471-500 [ 140 ] St?Hlman M, Ejsing CS, Tarasov K, et al. High-throughput shotgun lipidomics by quadrupole time-of-flight mass spectrometry [J]. J Chromatogr B Analyt Technol Biomed Life, 2009, 877(26): 2664-2672 [ 141 ] Dennis EA, Deems RA, Harkewicz R, et al. A Mouse Macrophage Lipidome [J]. Journal of Biological Chemistry, 2010, 285(51): 39976-39985 [ 142 ] Fauland A, K?feler H, Tr?tzmüller M, et al. A comprehensive method for lipid profiling by liquid chromatography-ion cyclotron resonance mass spectrometry [J]. Journal of Lipid Research, 2011, 52(12): 2314 [ 143 ] Cajka T, Fiehn O. Comprehensive analysis of lipids in biological systems by liquid chromatography-mass spectrometry [J]. Trends in Analytical Chemistry, 2014, 61: 192-206 [ 144 ] Paglia G, Kliman M, Claude E, et al. Applications of ion-mobility mass spectrometry for lipid analysis [J]. Analytical and Bioanalytical Chemistry, 2015: [ 145 ] Au A. Metabolomics and Lipidomics of Ischemic Stroke [J]. Adv Clin Chem, 2018, 85: 31-69 [ 146 ] Checa A, Bedia C, Jaumot J. Lipidomic data analysis: Tutorial, practical guidelines and applications [J]. Analytica Chimica Acta, 2015, 885: [ 147 ] Wang J, Wang C, Han X. Tutorial on lipidomics [J]. Analytica chimica acta, 2019, 1061: 28-41 [ 148 ] Corey KE, Vuppalanchi R, Wilson LA, et al. NASH resolution is associated with improvements in HDL and triglyceride levels but not improvement in LDL or non-HDL-C levels [J]. Aliment Pharmacol Ther, 2015, 41(3): 301-309 [ 149 ] Hao C, Liu Z, Blum W, et al. Quantification of valproic acid and its metabolite 2-propyl-4-pentenoic acid in human plasma using HPLC-MS/MS [J]. Journal of Chromatography B, 2007, 850(1-2): 206-212 [ 150 ] Ding J, Wang Y, Lin W, et al. A population pharmacokinetic model of valproic acid in pediatric patients with epilepsy: a non-linear pharmacokinetic model based on protein-binding saturation [J]. Clin Pharmacokinet, 2015, 54(3): 305-317 [ 151 ] Blanco-Serrano B, Otero MJ, Santos-Buelga D, et al. Population estimation of valproic acid clearance in adult patients using routine clinical pharmacokinetic data [J]. Biopharm Drug Dispos, 1999, 20(5): 233-240 [ 152 ] Fricke-Galindo I, Jung-Cook H, A LL, et al. Pharmacogenetics of adverse reactions to antiepileptic drugs [J]. Neurologia, 2018, 33(3): 165-176 [ 153 ] Ráduly Z, Szabó L, Madar A, et al. Toxicological and Medical Aspects of Aspergillus-Derived Mycotoxins Entering the Feed and Food Chain [J]. Frontiers in Microbiology, 2020, 10: 2908- [ 154 ] Hu?ana?u C, Sfarti C, Trifan A, et al. [High levels of sterigmatocystin in patients with chronic liver diseases] [J]. Rev Med Chir Soc Med Nat Iasi, 2011, 115(1): 33-37 [ 155 ] Creff J, Besson A. Functional Versatility of the CDK Inhibitor p57(Kip2) [J]. Front Cell Dev Biol, 2020, 8: 584590 [ 156 ] Roy A, Banerjee S. p27 and leukemia: cell cycle and beyond [J]. J Cell Physiol, 2015, 230(3): 504-509 [ 157 ] Hattori H, Kuroda M, Ishida T, et al. Human DNA damage checkpoints and their relevance to soft tissue sarcoma [J]. Pathology International, 2010, 54(1): 26-31 [ 158 ] Schafer, K. A. The cell cycle: a review [J]. Veterinary Pathology, 1998, 35(6): 461-478 [ 159 ] Malumbres M, Barbacid M. Mammalian cyclin-dependent kinases [J]. Trends in biochemical sciences, 2005, 30(11): 630-641 [ 160 ] Roskoski R. Cyclin-dependent protein serine/threonine kinase inhibitors as anticancer drugs [J]. Pharmacological research, 2019, 139: 471-488 [ 161 ] Coffman J. Cell cycle development [J]. Developmental cell, 2004, 6(3): 321-327 [ 162 ] Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation [J]. Development, 2013, 140(15): 3079-3093 [ 163 ] Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm [J]. Nature Reviews Cancer: [ 164 ] Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation [J]. Development (Cambridge, England), 2013, 140(15): 3079-3093 [ 165 ] Malumbres, Marcos. Cyclin-dependent kinases [J]. Chemical Reviews, 2014, 15(6): 122 [ 166 ] Malumbres M, Barbacid M. To cycle or not to cycle: a critical decision in cancer [J]. Nature Reviews Cancer, 2001, 1(3): 222 [ 167 ] Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm [J]. Nat Rev Cancer, 2009, 9(3): 153-166 [ 168 ] William J, Harbour, and, et al. Cdk Phosphorylation Triggers Sequential Intramolecular Interactions that Progressively Block Rb Functions as Cells Move through G1 [J]. Cell, 1999: [ 169 ] Lundberg AS, Weinberg RA. Functional Inactivation of the Retinoblastoma Protein Requires Sequential Modification by at Least Two Distinct Cyclin-cdk Complexes [J]. Molecular & Cellular Biology, 1998, 18(2): 753-761 [ 170 ] Uzma, Asghar, Agnieszka, et al. The history and future of targeting cyclin-dependent kinases in cancer therapy [J]. Nature Reviews Drug Discovery, 2015: [ 171 ] Bertoli C, Skotheim JM, de Bruin RA. Control of cell cycle transcription during G1 and S phases [J]. Nat Rev Mol Cell Biol, 2013, 14(8): 518-528 [ 172 ] Russo GL, Stampone E, Cervellera C, et al. Regulation of p27(Kip1) and p57(Kip2) Functions by Natural Polyphenols [J]. Biomolecules, 2020, 10(9): [ 173 ] Reynisdottir I, Polyak K, Iavarone A, et al. Kip/Cip and Ink4 Cdk inhibitors cooperate to induce cell cycle arrest in response to TGF-beta [J]. Genes & Development, 1995, 9(15): 1831 [ 174 ] Maddika S, Ande SR, Panigrahi S, et al. Cell survival, cell death and cell cycle pathways are interconnected: implications for cancer therapy [J]. Drug Resist Updat, 2007, 10(1-2): 13-29 [ 175 ] Garcia-Gutierrez L, Delgado MD, Leon J. MYC Oncogene Contributions to Release of Cell Cycle Brakes [J]. Genes (Basel), 2019, 10(3): [ 176 ] Cheng M, Sexl V, Sherr CJ, et al. Assembly of cyclin D-dependent kinase and titration of p27Kip1 regulated by mitogen-activated protein kinase kinase(MEK1) [J]. Proceedings of the National Academy of Sciences of the United States of America, 1998, 95(3): 1091-1096 [ 177 ] Roskoski R, Jr. Cyclin-dependent protein serine/threonine kinase inhibitors as anticancer drugs [J]. Pharmacol Res, 2019, 139: 471-488 [ 178 ] Huang S, Wang J, Xing L, et al. Impairment of cell cycle progression by sterigmatocystin in human pulmonary cells in vitro [J]. Food Chem Toxicol, 2014, 66: 89-95 [ 179 ] Jiang X, Wang J, Xing L, et al. Sterigmatocystin-induced checkpoint adaptation depends on Chk1 in immortalized human gastric epithelial cells in vitro [J]. Arch Toxicol, 2017, 91(1): 259-270 [ 180 ] Xing X, Wang J, Xing LX, et al. Involvement of MAPK and PI3K signaling pathway in sterigmatocystin-induced G2 phase arrest in human gastric epithelium cells [J]. Molecular Nutrition & Food Research, 2011: [ 181 ] Zhang D, Cui Y, Shen H, et al. Sterigmatocystin-Induced DNA Damage Triggers G2 Arrest via an ATM/p53-Related Pathway in Human Gastric Epithelium GES-1 Cells In Vitro [J]. Plos One, 2013, 8(5): e65044 [ 182 ] Cui J, Wang J, Huang S, et al. The G2 phase arrest induced by sterigmatocystin is dependent on hMLH1- ERK/p38-p53 pathway in human esophageal epithelium cells in vitro [J]. Food & Chemical Toxicology, 2018, 115: 205-211 [ 183 ] Wang J, Huang S, Xing L, et al. Role of hMLH1 in sterigmatocystin-induced G2 phase arrest in human esophageal epithelial Het-1A cells in vitro [J]. Toxicology Letters, 2013, 217(3): 226-234 [ 184 ] Liu Y, Du M, Zhang G. Proapoptotic activity of aflatoxin B1 and sterigmatocystin in HepG2 cells [J]. Toxicology Reports, 2014, 1(C): [ 185 ] Chang CH, Yu FY, Wu TS, et al. Mycotoxin Citrinin Induced Cell Cycle G2/M Arrest and Numerical Chromosomal Aberration Associated with Disruption of Microtubule Formation in Human Cells [J]. Toxicological Sciences An Official Journal of the Society of Toxicology, 2011, (1): 84 [ 186 ] Liang GA, Hong ZA, Tl A, et al. Molecular signatures of cytotoxic effects in human embryonic kidney 293cells treated with single and mixture of ochratoxin A and citrinin [J]. Food and Chemical Toxicology, 2019, 123: 374-384 [ 187 ] Sharath B, Tamatam A, Ilaiyaraja N, et al. Pelargonidin Modulates Keap1/Nrf2 Pathway Gene Expression and Ameliorates Citrinin-Induced Oxidative Stress in HepG2 Cells [J]. Frontiers in Pharmacology, 2017, 8: 868- [ 188 ] Andrew, H., Wyllie. The genetic regulation of apoptosis [J]. Current Opinion in Genetics & Development, 1995: [ 189 ] Fleisher TA. Apoptosis [J]. Ann Allergy Asthma Immunol, 1997, 78(3): 245-249; quiz 249-250 [ 190 ] Ribeiro DHB, Ferreira FL, Silva VNd, et al. Effects of Aflatoxin B1 and Fumonisin B1 on the Viability and Induction of Apoptosis in Rat Primary Hepatocytes [J]. International Journal of Molecular Sciences, 2010, 11(4): 1944-1955 [ 191 ] Galluzzi L, Vitale I, Aaronson SA, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018 [J]. Cell Death Differ, 2018, 25(3): 486-541 [ 192 ] Abou-Ghali M, Stiban J. Regulation of ceramide channel formation and disassembly: Insights on the initiation of apoptosis [J]. Saudi journal of biological sciences, 2015, 22(6): 760-772 [ 193 ] Ayed-Boussema I, Bouaziz C, Rjiba K, et al. The mycotoxin Zearalenone induces apoptosis in human hepatocytes (HepG2) via p53-dependent mitochondrial signaling pathway [J]. Toxicology in Vitro, 2008, 22(7): 1671-1680 [ 194 ] Cui J, Xing L, Li Z, et al. Ochratoxin A induces G(2) phase arrest in human gastric epithelium GES-1 cells in vitro [J]. Toxicology Letters, 2010, 193(2): 152-158 [ 195 ] Wang K, Lin Z, Zhang H, et al. Investigating proteome and transcriptome response of Cryptococcus podzolicus Y3 to citrinin and the mechanisms involved in its degradation [J]. Food Chemistry, 2019, 283(JUN.15): 345-352 [ 196 ] Hanahan D, Weinberg RA. The hallmarks of cancer [J]. 2000, 100(1): 0-70 [ 197 ] Maiuri MC, Zalckvar E, Kimchi A, et al. Self-eating and self-killing: crosstalk between autophagy and apoptosis [J]. Nature Reviews Molecular Cell Biology, 2007, 8(9): 741-752 [ 198 ] Siddiqui WA, Ahad A, Ahsan H. The mystery of BCL2 family: Bcl-2 proteins and apoptosis: an update [J]. Arch Toxicol, 2015, 89(3): 289-317 [ 199 ] Moldoveanu T, Czabotar PE. BAX, BAK, and BOK: A coming of age for the BCL-2 family effector proteins [J]. Cold Spring Harbor perspectives in biology, 2019, 12(4): a036319 [ 200 ] Galluzzi L, Vitale I, Aaronson SA, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018 [J]. Cell Death and Differentiation, 2018: [ 201 ] Brooks C, Dong Z. Regulation of Mitochondrial Morphological Dynamics During Apoptosis by Bcl-2 family proteins: A Key in Bak? [J]. Cell Cycle, 2007, 6(24): 3043-3047 [ 202 ] Cohen GM. Caspases: the executioners of apoptosis [J]. Biochem J, 1997, 326 ( Pt 1)(Pt 1): 1-16 [ 203 ] Julien O, Wells JA. Caspases and their substrates [J]. Cell Death Differ, 2017, 24(8): 1380-1389 [ 204 ] Tummers B, Green DR. Caspase-8: regulating life and death [J]. Immunol Rev, 2017, 277(1): 76-89 [ 205 ] Robertson JD, Orrenius S. Molecular Mechanisms of Apoptosis Induced by Cytotoxic Chemicals [J]. Critical Reviews in Toxicology, 2000, 30(5): 609-627 [ 206 ] de Souza KFS, Tofoli D, Pereira IC, et al. A styrylpyrone dimer isolated from Aniba heringeri causes apoptosis in MDA-MB-231 triple-negative breast cancer cells [J]. Bioorg Med Chem, 2021, 32: 115994 [ 207 ] Ali D, Tripathi A, Al Ali H, et al. ROS-dependent Bax/Bcl2 and caspase 3 pathway-mediated apoptosis induced by zineb in human keratinocyte cells [J]. Onco Targets Ther, 2018, 11: 489-497 [ 208 ] Gomes CC, Bernardes VF, Diniz MG, et al. Anti-apoptotic gene transcription signature of salivary gland neoplasms [J]. BMC Cancer, 2012, 12: 61 [ 209 ] Siqueira EC, Souza FT, Diniz MG, et al. Hsp27 (HSPB1) differential expression in normal salivary glands and pleomorphic adenomas and association with an increased Bcl2/Bax ratio [J]. Tumour Biol, 2015, 36(1): 213-217 [ 210 ] SunXM等, ZhangXH. Effects of Sterigmatocystin, Deoxynivalenol and Aflatoxin G1 on Apoptosis of Human Peripheral Blood Lymphocytes in vitro [J]. 生物医学与环境科学:英文版, 2002, 15(2): 145-152 [ 211 ] A JC, A JW, C SH, et al. Sterigmatocystin induced apoptosis in human pulmonary cells in vitro [J]. Experimental and Toxicologic Pathology, 2017, 69(8): 695-699 [ 212 ] Zingales V, Fernández-Franzón M, Ruiz MJ. The role of mitochondria in sterigmatocystin-induced apoptosis on SH-SY5Y cells [J]. Food and Chemical Toxicology, 2020, 142: 111493 [ 213 ] Chan W. Citrinin induces apoptosis in mouse embryonic stem cells [J]. Iubmb Life, 2010, 60(3): [ 214 ] Gayathri L, Dhivya R, Dhanasekaran D, et al. Hepatotoxic effect of ochratoxin A and citrinin, alone and in combination, and protective effect of vitamin E: In vitro study in HepG2 cell [J]. Food and Chemical Toxicology, 2015: [ 215 ] Chomczynski PA. A reagent for the single-step simultaneous isolation of RNA, DNA and proteins from cell and tissue samples [J]. Biotechniques, 1993, 15(3): 532 [ 216 ] Liu Y, Du M, Zhang G. Proapoptotic activity of aflatoxin B1 and sterigmatocystin in HepG2 cells [J]. Toxicol Rep, 2014, 1: 1076-1086 [ 217 ] Wang X, Xu Y, Song X, et al. Analysis of glycerophospholipid metabolism after exposure to PCB153 in PC12 cells through targeted lipidomics by UHPLC-MS/MS [J]. Ecotoxicology and Environmental Safety, 2018, 169(MAR.): 120-127 [ 218 ] Wang J, Li Z, Chen J, et al. Metabolomic identification of diagnostic plasma biomarkers in humans with chronic heart failure [J]. Molecular Biosystems, 2013, 9(11): 2618-2626 [ 219 ] Gao W, Jiang L, Ge L, et al. Sterigmatocystin-induced oxidative DNA damage in human liver-derived cell line through lysosomal damage [J]. Toxicol In Vitro, 2015, 29(1): 1-7 [ 220 ] Patterson JC, Joughin BA, van de Kooij B, et al. ROS and Oxidative Stress Are Elevated in Mitosis during Asynchronous Cell Cycle Progression and Are Exacerbated by Mitotic Arrest [J]. Cell Syst, 2019, 8(2): 163-167 e162 [ 221 ] Jia, Guo, Chun, et al. MiR-375 induces ROS and apoptosis in ST cells by targeting the HIGD1A gene [J]. Gene, 2018: [ 222 ] Sunaga H, Matsui H, Anjo S, et al. Elongation of Long‐Chain Fatty Acid Family Member 6 (Elovl6)‐Driven Fatty Acid Metabolism Regulates Vascular Smooth Muscle Cell Phenotype Through AMP‐Activated Protein Kinase/Krüppel‐Like Factor 4 (AMPK/KLF4) Signaling [J]. Journal of the American Heart Association: Cardiovascular and Cerebrovascular Disease, 2016, 5(12): [ 223 ] Roskoski R. Cyclin-dependent protein serine/threonine kinase inhibitors as anticancer drugs [J]. Pharmacological Research, 2018: [ 224 ] BAX, BAK, and BOK: A coming of age for the BCL-2 family effector proteins [J]. Cold Spring Harbor perspectives in biology, 2019, 12(4): a036319 [ 225 ] Menendez JA, Lupu R. Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis [J]. Nature Reviews Cancer, 2007, 7(10): 763-777 [ 226 ] Eum JY, Lee JC, Sun SY, et al. Aging-related lipidomic changes in mouse serum, kidney, and heart by nanoflow ultrahigh-performance liquid chromatography-tandem mass spectrometry [J]. Journal of Chromatography A, 2020, 1618: 460849 [ 227 ] Abnormalities in Plasma Phospholipid Fatty Acid Profiles of Patients with Hepatocellular Carcinoma [J]. Lipids, 2015, 50(10): 977-985 [ 228 ] Okita M, Tomioka K, Ota Y, et al. Arachidonic acid in mononuclear cells and its clinical significance in HCV cirrhotic patients [J]. Nutrition, 2003, 19(9): 727-732 [ 229 ] Arain SQ, Talpur FN, Channa NA, et al. Serum lipid profile as a marker of liver impairment in hepatitis B Cirrhosis patients [J]. Lipids in Health & Disease, 2017, 16(1): 51 [ 230 ] Bao Y, Wang S, Yang X, et al. Metabolomic study of the intervention effects of Shuihonghuazi Formula, a Traditional Chinese Medicinal formulae, on hepatocellular carcinoma (HCC) rats using performance HPLC/ESI-TOF-MS [J]. Journal of Ethnopharmacology, 2017, 198: 468-478 [ 231 ] Huang W, Xie P, Cai Z. Lipid metabolism disorders contribute to hepatotoxicity of triclosan in mice [J]. Journal of Hazardous Materials, 2019, 384: 121310 [ 232 ] Kosuke S, Masafumi I, Yasushi K, et al. Lipid profiling of pre-treatment plasma reveals biomarker candidates associated with response rates and hand–foot skin reactions in sorafenib-treated patients [J]. Cancer Chemotherapy and Pharmacology, 2018, 82: [ 233 ] Cassol E, Misra V, Holman A, et al. Plasma metabolomics identifies lipid abnormalities linked to markers of inflammation, microbial translocation, and hepatic function in HIV patients receiving protease inhibitors [J]. Bmc Infectious Diseases, 2013, 13(1): 203-203 [ 234 ] Wang W, Lv J, Nan C, et al. Dysregulated serum metabolites in staging of hepatocellular carcinoma [J]. Clinical Biochemistry, 2018, 61: 7-11 [ 235 ] Rao RP, Vaidyanathan N, Rengasamy M, et al. Sphingolipid Metabolic Pathway: An Overview of Major Roles Played in Human Diseases [J]. Journal of Lipids,2013,(2013-8-4), 2013, 2013: 178910 [ 236 ] Li Z, Guan M, Yu L, et al. Aberrant Lipid Metabolism in Hepatocellular Carcinoma Revealed by Liver Lipidomics [J]. International Journal of Molecular Sciences, 2017, 18(12): 2550 [ 237 ] Murphy RC, Leiker TJ, Barkley RM. Glycerolipid and cholesterol ester analyses in biological samples by mass spectrometry [J]. BBA - Molecular and Cell Biology of Lipids, 2011, 1811(11): 776-783 [ 238 ] Chitraju C, Trotzmuller M, Hartler J, et al. Lipidomic analysis of lipid droplets from murine hepatocytes reveals distinct signatures for nutritional stress [J]. Journal of Lipid Research, 2012, 53(10): 2141-2152 [ 239 ] Donnelly, Kerry, L., et al. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease [J]. Journal of Clinical Investigation, 2005, 115(5): 1343-1351 [ 240 ] Zhang H, Shao X, Zhao H, et al. Integration of Metabolomics and Lipidomics Reveals Metabolic Mechanisms of Triclosan-Induced Toxicity in Human Hepatocytes [J]. Environmental Science and Technology, 2019, 53(9): 5406-5415 [ 241 ] Currie E, Schulze A, Zechner R, et al. Cellular Fatty Acid Metabolism and Cancer [J]. Cell Metabolism, 2013, 18(2): 153-161 [ 242 ] Sebastiano MR, Konstantinidou G. Targeting Long Chain Acyl-CoA Synthetases for Cancer Therapy [J]. International Journal of Molecular Sciences, 2019, 20(15): 3624 [ 243 ] Kanter JE, Tang C, Oram JF, et al. Acyl-CoA synthetase 1 is required for oleate and linoleate mediated inhibition of cholesterol efflux through ATP-binding cassette transporter A1 in macrophages [J]. Biochimica Et Biophysica Acta, 2012, 1821(3): 358-364 [ 244 ] Resveratrol Modulates Desaturase Expression and Fatty Acid Composition of Cultured Hepatocytes [J]. Frontiers in Nutrition, 2018: [ 245 ] Li C, Paliogiannis P, Cigliano A, et al. Pathogenetic, therapeutic, and prognostic role of fatty acid synthase in human hepatocellular carcinoma [J]. Frontiers in Oncology, 2019, In press: [ 246 ] Wang Y, Viscarra J, Kim SJ, et al. Transcriptional regulation of hepatic lipogenesis [J]. Nature Reviews Molecular Cell Biology, 2015, 16(11): 678 [ 247 ] Staels, Bart, Pawlak, et al. Molecular mechanism of PPAR alpha action and its impact on lipid metabolism, inflammation and fibrosis in non-alcoholic fatty liver disease [J]. Journal of Hepatology: The Journal of the European Association for the Study of the Liver, 2015, 62(3): 720-733 [ 248 ] Sharath Babu GR, Anand T, Ilaiyaraja N, et al. Pelargonidin Modulates Keap1/Nrf2 Pathway Gene Expression and Ameliorates Citrinin-Induced Oxidative Stress in HepG2 Cells [J]. Front Pharmacol, 2017, 8: 868 [ 249 ] Siqueira E, Souza F, Diniz MG, et al. Hsp27 (HSPB1) differential expression in normal salivary glands and pleomorphic adenomas and association with an increased Bcl2/Bax ratio [J]. Tumor Biology, 2015: [ 250 ] Kfsds A, Dt B, Icp A, et al. A styrylpyrone dimer isolated from Aniba heringeri causes apoptosis in MDA-MB-231 triple-negative breast cancer cells [J]. Bioorganic & Medicinal Chemistry, 2021: [ 251 ] Gupta L, Guleria A, Rawat A, et al. NMR‐based clinical metabolomics revealed distinctive serum metabolic profiles in patients with spondyloarthritis [J]. Magnetic Resonance in Chemistry, 2020: [ 252 ] Araújo B, Furtado D, Leite F, et al. Metabolic profiling of organic acids in urine samples of Cri Du Chat syndrome individuals by gas chromatography-mass spectrometry [J]. Journal of Chromatography B, 2020, 1153: 122267 [ 253 ] Chong IG, Jun CH. Performance of some variable selection methods when multicollinearity is present [J]. Chemometrics & Intelligent Laboratory Systems, 2005, 78(1-2): 103-112 [ 254 ] Wold S. PLS-regression : a basic tool for chemometrics [J]. Chemometrintellsystem, 2001, 58: [ 255 ] Ren C, Jia L, Zhou J, et al. Lipidomic profiling of plasma samples from patients with mitochondrial disease [J]. Biochemical and Biophysical Research Communications, 2018, 500(2): [ 256 ] Lai-Fang, Zhou, Bo-Wen, et al. Plasma metabolomics profiling for fish maturation in blunt snout bream [J]. Metabolomics, 2017, 13(4): [ 257 ] Wang Z, Fan M, Can Da S D, et al. Cyclin B1/Cdk1 Coordinates Mitochondrial Respiration for Cell-Cycle G2/M Progression [J]. Developmental Cell, 2014, 29(2): 217-232 [ 258 ] Okita M, Tomioka K, Ota Y, et al. Arachidonic acid in mononuclear cells and its clinical significance in HCV cirrhotic patients [J]. Nutrition, 2003, 19(9): 727-732 [ 259 ] Mcclinton S, Moffat L, Horrobin D, et al. Abnormalities of essential fatty acid distribution in the plasma phospholipids of patients with bladder cancer [J]. British Journal of Cancer: [ 260 ] Bao Y, Wang S, Yang X, et al. Metabolomic study of the intervention effects of Shuihonghuazi Formula, a Traditional Chinese Medicinal formulae, on hepatocellular carcinoma (HCC) rats using performance HPLC/ESI-TOF-MS [J]. J Ethnopharmacol, 2017, 198: 468-478 [ 261 ] Lipid metabolism disorders contribute to hepatotoxicity of triclosan in mice [J]. Journal of Hazardous Materials, 2019, 384: 121310 [ 262 ] Li R, Wang Y, Hou B, et al. Lipidomics insight into chronic exposure to ambient air pollution in mice [J]. Environmental Pollution, 2020, 262: 114668 [ 263 ] Liu P, Zhu W, Chen C, et al. The mechanisms of lysophosphatidylcholine in the development of diseases [J]. Life Sciences, 2020, 247: [ 264 ] Wu T, Zheng X, Yang M, et al. Serum lipid alterations identified in chronic hepatitis B, hepatitis B virus-associated cirrhosis and carcinoma patients [J]. Scientific Reports, 2017, 7: 42710 [ 265 ] Li S, Liu H, Jin Y, et al. Metabolomics study of alcohol-induced liver injury and hepatocellular carcinoma xenografts in mice [J]. J Chromatogr B Analyt Technol Biomed Life Sci, 2011, 879(24): 2369-2375 [ 266 ] Morita Y, Sakaguchi T, Ikegami K, et al. Lysophosphatidylcholine acyltransferase 1 altered phospholipid composition and regulated hepatoma progression [J]. Journal of Hepatology, 2013, 59(2): 292-299 [ 267 ] Saito K, Ikeda M, Kojima Y, et al. Lipid profiling of pre-treatment plasma reveals biomarker candidates associated with response rates and hand-foot skin reactions in sorafenib-treated patients [J]. Cancer Chemother Pharmacol, 2018, 82(4): 677-684 [ 268 ] Wu T, Zheng X, Yang M, et al. Serum lipid alterations identified in chronic hepatitis B, hepatitis B virus-associated cirrhosis and carcinoma patients [J]. Sci Rep, 2017, 7: 42710 [ 269 ] Wang B, Zhang J, Chen C, et al. The size of zinc oxide nanoparticles controls its toxicity through impairing autophagic flux in A549 lung epithelial cells [J]. Toxicol Lett, 2018, 285: 51-59 |
中图分类号: | TS2 |
开放日期: | 2021-06-15 |