三聚氰胺单独或与三聚氰酸协同对雄性小鼠血睾屏障的影响
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
2008年中国婴幼儿因食用非法掺杂三聚氰胺(melamine, MA)的配方奶粉爆发尿结石,该事件引起了世人对三聚氰胺或与其同系物三聚氰酸(cyanuric acid, CA)协同作用CMA+CA, MC)对人和动物健康影响的广泛关注。过去大量的研究都集中在MA及其同系物对泌尿系统的毒性作用上,对其他系统的毒性作用研究报道还很少见。本研究采用组织细胞病理学和超微病理学观察技术、免疫组化、Western Blot技术、细胞培养和流式细胞术等方法,在原有研究的基础上,以血睾屏障和支持细胞为切入点,从毒理病理学角度观察研究了MA及其同系物对小鼠生殖系统的毒性作用,在整体水平、组织学水平、细胞学水平、超微水平和分子水平上多角度、系统地揭示了MA对小鼠生殖系统的毒性作用及其机理。研究结果如下:
     1.体内试验
     以8周龄雄性昆明小鼠为试验对象,设立对照组,MA组和MC组,其中MA组和MC组分别设立低、中、高三个剂量组,MA剂量分别是30、140和7OOmg/kg.bw/d, MC剂量分别是15(15MA+15CA).70(70MA+70CA)和350(350MA+350CA) mg/kg.bw/d, MC由相同剂量的MA和CA混合。采用灌胃的方式染毒,连续3天,染毒后第1天(Post1d)和第5天(Post5d)剖检取材,评价MA和MC的雄性生殖毒性效应,观察血睾屏障结构的变化,探讨它们对睾丸毒性作用的靶细胞。
     通过临床观察、血清学分析、剖检及组织病理学观察和精子指标检测可见,Post1d时,与对照组相比,MC各剂量组体重增长均显著下降(P<0.05或P<0.01),中、高剂量组小鼠出现明显的临床症状;高剂量组睾酮水平显著下降(P<0.01),睾丸和附睾脏器指数显著下降(P<0.05或P<0.01)。所有MA和MC组小鼠的睾丸和附睾组织出现了明显的病理学损伤,呈现剂量相关性,病变主要表现为生精细胞脱落和生精小管上皮空泡化,附睾输出管中充满多量脱落的生精上皮细胞,严重者出现少精或无精;所有处理组精子畸形率均显著升高(P<0.05或P<0.01)。到Post5d时,所有处理组精子畸形率仍然是显著升高(P<0.05或P<0.01),其它检测指标显示睾丸毒性程度减轻。以上结果表明,MA和MC对睾丸生精上皮具有直接的损伤作用,从而导致精子质量下降。
     利用TUNEL染色观察睾丸细胞凋亡情况,并用免疫组化和Western Blot定位和定量检测了Fas/Fas-L蛋白在睾丸组织中的表达变化。结果可见:Postld时,与对照组相比,处理组睾丸生精细胞出现明显凋亡,MA低剂量组和MC各剂量组睾丸凋亡指数显著升高(P<0.05或P<0.01),Fas和Fas-L分别在生精细胞和支持细胞中有不同程度的表达;Post5d时,与对照组相比,MA高剂量组和MC中、高剂量组凋亡指数仍显著升高(P<0.05或P<0.01),MA与MC高剂量组Fas和Fas-L均在生精细胞中出现强阳性表达。上述结果提示,MA和MC可以促进小鼠睾丸的生精细胞凋亡。
     利用透射电镜观察血睾屏障超微结构变化,并用免疫组化和Western Blot定位和定量检测了各层血睾屏障结构中关键蛋白的表达变化。结果发现:Postld时,与对照组相比,所有MA与MC组的小鼠血睾屏障各层超微结构出现了明显的损伤,主要表现有:毛细血管内皮细胞、肌样细胞和支持细胞胞浆中出现大量空泡,支持细胞间的紧密连接出现明显裂隙;支持细胞中的波形蛋白出现崩解,表达量明显下降(P<0.05或P<0.01),睾丸组织中β-actin蛋白表达量不同程度出现显著下降(P<0.05或P<0.01)。Post5d,各处理组血睾屏障的超微结构损伤仍很严重,波形蛋白表达量仍下降(P<0.05或P<0.01)。以上结果表明,MA和MC可以破坏小鼠睾丸血睾屏障结构,导致支持细胞骨架结构崩解,说明血睾屏障结构的破坏在MA和MC引发的雄性生殖毒性作用中发挥了关键的作用。
     2.体外试验
     以小鼠TM4支持细胞株为试验对象,采用MTT、透射电镜、流式细胞仪、Western Blot等方法,观察不同浓度MA(0-1000ug/mL)对支持细胞的毒性作用。结果可见,TM4细胞在50ug/mL及以上的MA作用24h后,细胞存活率显著下降(P<0.05或P<0.01);在30-500ug/mL的MA作用24h后,细胞的超微结构出现了明显损伤,高浓度MA促进细胞凋亡(P<0.05或P<0.01);在50ug/mL MA分别作用0、1、3、6、24h后,波形蛋白随着时问延长表达量下降。以上结果表明低浓度的MA即可对TM4支持细胞产生明显的损伤,提示支持细胞可能是MA生殖毒性作用的重要靶点细胞。
     上述体内和体外试验观察研究结果表明,在本试验剂量条件下MA单独和MC混合灌服均可引起雄性小鼠睾丸组织明显的损伤,精子畸形率升高,造成损伤的主要机制可能是由于MA和MC靶向于支持细胞破坏血睾屏障结构,继而引起各级生精细胞发生变性坏死、凋亡加剧,以致精子生成障碍、发育畸形。
In2008in China, the occurrence of an outbreak of urinary stones in infants draw critical attention to the toxicity of melamine (MA) or the mixture of MA and cyanuric acid (CA) in human and animals.Until now, a large number of researches focused on the toxic effects of MA and its analoges on the urinary organs. In this study, based on our prelimitary results, we focused on the blood-testis barrier and Sertoli cells to investigate the toxic effects of MA and its analogues on male reproductive system on different levels with methods of histochemical and ultrastructural pathology,immunohistochemical staining, Western blot and flow cytometry. The research contents are as follows:
     1. In vivo study
     Eight-week-old male Kunming mice were administered either melamine (MA,30,140, or700mg/kg/day), a melamine and cyanuric acid mixture (MC, each at15,70, or350mg/kg/day), or vehicle (control) for3consecutive days. Testicular toxicity was evaluated on days1(Post Id) and5(Post5d) after the final exposure. Furthermore, the structure of blood-testis barrier and possible cellular target for MA and MC was investigated.
     With the methods of clinical observation, serum chemistry analysis、autopsy、histopathological examination and sperm detection, on Post1d, we found that the body weight gain of the MC-treated mice significantly decreased (P<0.05or P<0.01), with obvious clinical sighs in the higher groups. In the highest MC group, the serum T level significantly decreased(P<0.01), and the organ indexes of testis and epididymis decreased (P<0.05or P<0.01). In both MA and MC groups, the testes and epididymis tissue showed dose-related histopathological lesions, and the characterize changes were germ cell exfoliation and tubular vacuolation. Additionally, the sperm abnormality were significantly increased in all treated groups (P<0.05or P<0.01). On Post5d, the sperm abnormality were still significantly increased in all treated groups (P<0.05or P<0.01).The other indexes indicated that the degree of testicular injury became slighter at this time point. These results suggested that MA and MC had direct toxicity on the testicular epithelium and caused decrease of sperm quality.
     We detected the apoptosis and the protein expression of Fas/Fas-L in the testes with TUNEL staining, immunohistochemistry staining and Western Blot. On post Id, the results showed that the germ cells exhibited apoptosis in the testes among all treated groups, and apoptotic index was significantly increased in low MA group and all MC groups(P<0.05or P<0.01). The Fas and Fas-L distributed in the germ cells and Sertoli cells, respectively. On post5d, apoptotic index in the highest MA group and the highest two MC groups(P<0.05or P<0.01). In the highest MC group, the Fas and Fas-L protein showed strong expression in germ cells. These results suggested that MA and MC could induce germ cell apoptosis, and the Fas/Fas-L system may involved the apoptotic process.
     The structure of blood-testis barrier and related protein was evaluated with TEM, immunohistochemistry staining and Western Blot. On Post Id, the results showed that structure of the blood-testis barrier was damaged dose dependently. The vimentin filament in Sertoli cell collapsed in all treated groups. The expression of vimentin and β-actin protein decreased with difference degree among the treated groups (P<0.05or P<0.01). On Post5d, the lesions of the blood-testis barrier structure was still serious in the treated groups, with decreased expression of vimentin protein(P<0.05or P<0.01). These results suggested that MA and MC could disrupt the blood-testis barrier and decreased cytoskeleton protein expression. Sertoli cells appear to be a cellular target for MA and MC.
     2. In vitro study
     With the methods of MTT, TEM, Flow cytometry and Western Blot, we evaluated the cytotoxicity of MA (0-1000ug/mL) on TM4Sertoli cells. When cells were treated with MA at50ug/mL or above for24h, the cell viability was significantly decreased(P<0.05or P<0.01). When cells were treated with MA at range of30to500ug/mL for24h, the ultrastructure was obviously damaged, and high concentration of MA induced significant apoptosis(P<0.05or P<0.01). When cells were treated with MA at50ug/mL MA for0,1,3,6or24h, the expression of vimentin protein decreased with time. It can be seen that MA even at low concentration could induce obvious injury in TM4cells, suggesting that Sertoli cells maybe sensitive to MA toxicity.
     Taken together, the in vivo and in vitro studies showed that oral exposure to MA and MC in male Kumming mice could produce obvious testicular toxicity including germ cell sloughing and apoptosis and high rate of sperm abnormality. MC exhibited much more toxicity than MA. The damage of blood testis barrier and Sertoli cell play a critical role in the MA-and MC-induced male reproductive toxicity.
引文
[1]王延吉.化工产品手册有机化工原料.北京:化学工业出版社,2004.
    [2]Jutzi, K., A.M. Cook, and R. Hutter. The degradative pathway of the s-triazine melamine. The steps to ring cleavage. Biochem J,1982,208(3):679-84.
    [3]Shelton, D.R. Metabolism of Melamine by Klebsiella terragena. Appl Environ Microbiol,1997, 63(7):2832-5.
    [4]Canelli, E. Chemical, bacteriological, and toxicological properties of cyanuric acid and chlorinated isocyanurates as applied to swimming pool disinfection:a review. Am J Public Health,1974,64(2):155-62.
    [5]Barbee, S. Metabolism and disposition of sodium cyanurate. Toxicologist,1983,3:80.
    [6]Barbee,S. Metabolism and disposition of sodium cyanurate in the dog. Toxicologist,1984,4:92.
    [7]NTP. Carcinogenesis bioassay of melamine (CAS No.108-78-1) in F344/N rats and B6C3F1 mice (feed study). Research Triangle Park,NC, and Bethesda,MD,United States Department of Health and Human Services, Public Heath Service, National Institutes of Health,National Toxicology Program(NTP Technical Report Series No.245;NTP-81-86; NIH Publication No.83-2501;http://ntp.niehs.nih.gov/ntp/htdocs/LT-rpts/tr245.pdf.1983.
    [8]Heck, H.D. and R.W. Tyl. The induction of bladder stones by terephthalie acid, dimethyl terephthalate, and melamine (2,4,6-triamino-s-triazine) and its relevance to risk assessment. Regul Toxicol Pharmacol,1985,5(3):294-313.
    [9]朱金凤.三聚氰胺和三聚氰酸对小鼠泌尿系统毒性作用的研究[硕士学位论文].北京:中国农业大学,2011.
    [10]IUCLID. International Uniform Chemical Information Database. European Commission (http://iuclid.echa.europa.eu/).2000.
    [11]Bingham E, B Cohrssen, CH Powell. Patty's toxicology.5th ed. Vol.IV.Hydrocarbons/organic nitrogen compounds.New York,NY,John Wiley&Sons.2001.
    [12]Clark R. Melamine crystalluria in sheep. Journal of the South African Veterinary Medical Association,1966,37:349-351.
    [13]MacKenzie HI, van Rensburg IBJ. Ammelide and ammeline as nonprotein nitrogen supplements for sheep. Journal of the South African Veterinary Medical Association,1968,39: 41-45.
    [14]陈杰,黄绍棠,章再婷,等.三聚氰胺在猪体内的药物残留研究.饲料与畜牧,2009,18(8):53-55.
    [15]黄小燕.三聚氰胺对断奶仔猪的毒性效应和血浆清楚规律的研究[D].雅安:四川农业大学,2009.
    [16]林祥梅,王建峰,贾广乐,等.三聚氰胺的毒性研究.毒理学杂志,2008,22(3):216-218.
    [17]刘海燕,张伟, 薛敏,等.三聚氰胺对吉富罗非鱼的急性毒性研究.河北师范大学学报, 2009,33(5):659-665.
    [18]燕磊,郭吉原,孙作为,等.饲喂不同含量三聚氰胺日粮后肉鸭组织中三聚氰胺的残留量分析.中国家禽,2009,31(5):16-19.
    [19]张小燕,施祖灏,陆俊贤,等.三聚氰胺对黄羽肉鸡生长性能和血液生化指标的影响.中国家禽,2010,31(21):25-28.
    [20]高春起,武书庚,齐广海,等.三聚氰胺对蛋鸭产蛋性能、血清指标和肝肾组织结构的影响.中国农业科学,2010,43(5):p.1050-1056.
    [21]Skinner, C.G, J.D. Thomas, and J.D. Osterloh, Melamine toxicity. J Med Toxicol,2010,6(1): 50-5.
    [22]Ai K, Y Liu, L Lu, et al.Hydrogen-Bonding Recognition-Induced Color Change of Gold Nanoparticles for Visual Detection of Melamine in Raw Milk and Infant Formula. Journal of the American Chemical Society,2009,131(27):9496-9497.
    [23]Ranganathan A, V.R., Rpedireddi, P.V., Rao CNR. Hydrothermal synthesis of organic channel structures:1:1 hydrogen-bonded adducts of melamine with cyanuric and trithiocyanuric acids. J Am Chem Soc,1999,121(8):1752-1753.
    [24]Archer, EA, H Gong, MJ Krische. Hydrogen bonding in noncovalent synthesis:selectivity and the directed organization of molecular strands. Tetrahedron,2001,57(7):1139-1159.
    [25]Perdigao Luis M. A., NR Champness, PH Beton. Surface self-assembly of the cyanuric acid-melamine hydrogen bonded network. Chem Commun (Camb),2006,5:538-540.
    [26]Whitesides GM, JP Mathias, CT Seto. Molecular self-assembly and nanochemistry:a chemical strategy for the synthesis of nanostructures. Science,1991,254(5036):1312-1319.
    [27]Whitesides GM, EE Simanek, JP Mathias, et al. Noncovalent synthesis:using physical organic chemistry to make aggregates. Acc Chem Res,1995,28(1):37-44.
    [28]Zhang, Q.X., G Yang, J Li, et al. Melamine induces sperm DNA damage and abnormality, but not genetic toxicity. Regul Toxicol Pharmacol,2011,60(1):144-50.
    [29]Yin, R.H., XZ Wang, WL Bai, et al. The reproductive toxicity of melamine in the absence and presence of cyanuric acid in male mice. Res Vet Sci,2013,94(3):p.618-27.
    [30]田永刚,张迎梅.低剂量三聚氰胺对小鼠精子质量影响的研究.四川动物,2010.3:466-468.
    [31]尤华.三聚氰胺和三聚氰酸对雄性小鼠生殖系统毒性作用的研究[博士学位论文].北京:中国农业大学,2012.
    [32]常玲玲,朱金凤,岳卓,等.三聚氰胺对小鼠急性毒性试验中消化系统重要组织器官损伤的病理学观察.中国兽医科学,2011,10:1056-1060.
    [33]Yang, J., L An, Y Yao, et al. Melamine impairs spatial cognition and hippocampal synaptic plasticity by presynaptic inhibition of glutamatergic transmission in infant rats. Toxicology, 2011.289(2-3):167-74.
    [34]Zhou, W., Y Jiang, H Shi, et al. The characteristics of immune system changes in children who ingested melamine-contaminated powdered formula in China. Int J Environ Health Res.20(4): 289-97.
    [35]WHO.Sodium dichloroisocyanurate. In:Safety evaluation of certain food additives and contaminants. Prepared by the sixty-first meeting of the Joint FAO/WHO Expert Committee on Food Additives. Geneva, World Health Organization (WHO) Food Additives Series, No.52; http://whqlibdoc.who.int/publications/2004/924166052X.pdf),2004.
    [36]OECD, Screening Information Dataset(SIDS) for isocyanuric acid.CAS No.108-80-5.1999.
    [37]Rajasekaran D.13-week toxicity study in rats. Unpublished report from the International Research and Development Corporation, San Francisco Medical Center, San Francisco, CA. Submitted to WHO by the Industry Ad Hoc Committee on Isocyanurates (Report No.167-151).1981.
    [38]Serota D, K Hepner, R Alsaker, et al.104-week oncogenicity study in mice:monosodium cyanurate. Project No.2169-101. Unpublished report from Hazleton Laboratories. Submitted to WHO by the Industry Ad Hoc Committee on Isocyanurates,1986.
    [39]Cohen SM, L McKinney, BM Wagner, et al. An evaluation of the long-term toxicity/carcinogenicity of sodium isocyanurate. Unpublished report to Occidental Chemical Corporation,25 February 1999, provided to United States Environmental Protection Agency and assigned MRID No.44834403.
    [40]Aldridge D, JL Schardin, M Blair, et al. Three generation reproductive study in the rat with sodium salt of cyan uric acid (striazinetriol). Unpublished report from the International Research and Development Corporation.,1985. Submitted to WHO by the Industry Ad Hoc Committee on Isocyanurates (Report No.497-001):667-674.
    [41]Wheeler AG, SJ Barbee, BG Hammond, et al. Three generation reproduction study in rats administered cyanurate. Toxicologist,1985,5:189.
    [42]Hammond BG, SJ Barbee, AG Wheeler, et al.Absence of mutagenic activity for monosodium cyanurate. Fundam Appl Toxicol,1985,5(4):655-64.
    [43]Brown, C.A., KS Jeong, RH Poppenga, et al. Outbreaks of renal failure associated with melamine and cyanuric acid in dogs and cats in 2004 and 2007. J Vet Diagn Invest,2007,19(5): 525-31.
    [44]Puschner, B., RH Poppenga, LJ. Lowenstine, et al. Assessment of melamine and cyanuric acid toxicity in cats. J Vet Diagn Invest,2007,19(6):616-24.
    [45]Dobson, R.L., S Motlagh, M Quijano, et al. Identification and characterization of toxicity of contaminants in pet food leading to an outbreak of renal toxicity in cats and dogs. Toxicol Sci, 2008,106(1):251-62.
    [46]Jeong, W.I., SH Do, DH Jeong, et al. Canine renal failure syndrome in three dogs. J Vet Sci, 2006,7(3):299-301.
    [47]Gonzalez, J., B Puschner, V Perez, et al. Nephrotoxicosis in Iberian piglets subsequent to exposure to melamine and derivatives in Spain between 2003 and 2006. J Vet Diagn Invest, 2009,21(4):558-63.
    [48]Xie, G., X Zheng, X Qi, et al. Metabonomic evaluation of melamine-induced acute renal toxicity in rats. J Proteome Res,2009,9(1):125-33.
    [49]Stine, C.B., R Reimschuessel, CM Gieseker, et al.A No Observable Adverse Effects Level (NOAEL) for pigs fed melamine and cyanuric acid. Regul Toxicol Pharmacol,2011,60(3):363-72.
    [50]Wang, L, XM Ding, KY Zhang, et al. Toxicity of cyanuric acid to broilers on hepatic and renal health with and without melamine. Hum Exp Toxicol,2012,31(2):166-73.
    [51]Reimschuessel, R., E. Evans, W. C. Andersen, et al. Residue depletion of melamine and cyanuric acid in catfish and rainbow trout following oral administration. J Vet Pharmacol Ther,2010,33(2):172-82.
    [52]Kobayashi, T., A Okada, Y Fujii, et al.The mechanism of renal stone formation and renal failure induced by administration of melamine and cyanuric acid. Urol Res,2010,38(2):117-25.
    [53]Sprando, R.L., R Reimschuessel, CB Stine, et al. Timing and route of exposure affects crystal formation in melamine and cyanuric exposed male and female rats:gavage vs. feeding. Food Chem Toxicol,2012,50(12):4389-97.
    [54]Kim, C.W., JW Yun, IH Bae, et al.Determination of spatial distribution of melamine-cyanuric acid crystals in rat.kidney tissue by histology and imaging matrix-assisted laser desorption/ionization quadrupole time-of-flight mass spectrometry. Chem Res Toxicol,2009,23(1):220-7.
    [55]康友敏,张健,李健,等.哺乳动物血-睾屏障的研究进展.中国兽医学报,2002,22(4):418-420.
    [56]成令忠.现代组织学.上海科学技术文献出版社,2003,969-979.
    [57]Mruk, D.D. and C.Y. Cheng. Sertoli-Sertoli and Sertoli-germ cell interactions and their significance in germ cell movement in the seminiferous epithelium during spermatogenesis. Endocr Rev,2004.25(5):747-806.
    [58]Cheng, C.Y. and D.D. Mruk. The blood-testis barrier and its implications for male contraception. Pharmacol Rev,2012,64(1):16-64.
    [59]Yan, H.H., D.D. Mruk, and C.Y. Cheng. Junction restructuring and spermatogenesis:the biology, regulation, and implication in male contraceptive development. Curr Top Dev Biol, 2008,80:57-92.
    [60]Fiorini, C., A Tilloy-Ellul, S Chevalier, et al.Sertoli cell junctional proteins as early targets for different classes of reproductive toxicants. Reprod Toxicol,2004,18(3):413-21.
    [61]Okumura, K., I.P. Lee, and R.L. Dixon.Permeability of selected drugs and chemicals across the blood-testis barrier of the rat. Journal of Pharmacology and Experimental Therapeutics,1975, 194(1):89-95.
    [62]Elkin, N.D., J.A. Piner,and R.M. Sharpe, et al.Toxicant-induced leakage of germ cell-specific proteins from seminiferous tubules in the rat:Relationship to blood-testis barrier integrity and prospects for biomonitoring. Toxicological Sciences,2010,117(2):439-448.
    [63]Wang, X.W., GR Ding, CH Shi, et al. Mechanisms involved in the blood-testis barrier increased permeability induced by EMP. Toxicology,2010,276(1):58-63.
    [64]Pereira, M.L. and Garcia e Costa. The blood-testis barrier as a target of some chemotherapeutic agents. Chemotherapy,2007,53(6):446-448.
    [65]Wolski, K.M., D.D. Mruk, and D.F. Cameron. The Sertoli-Spermatid Junctional Complex Adhesion Strength Is Affected In Vitro by Adjudin. Journal of andrology,2006,27(6):790-794.
    [66]Siu, E.R., DD Mruk, CS Porto, et al. Cadmium-induced testicular injury. Toxicol Appl Pharmacol,2009,238(3):240-9.
    [67]Boekelheide,K., KJ Johnson, J Heindel, Sertoli cell toxicants. In:Korach, K. (Ed.), Reproductive and Developmental Toxicology. Marcel Dekker, New York.,1998.
    [68]Benoff, S., A. Jacob, and I.R. Hurley.Male infertility and environmental exposure to lead and cadmium. Hum Reprod Update,2000,6(2):107-21.
    [69]Parizek, J. THE THIRD OLIVER BIRD LECTURE STERILIZATION OF THE MALE BY CADMIUM SALTS. Journal of Reproduction and Fertility,1960,1(3):294-309.
    [70]Parizek, J. THE PECULIAR TOXICITY OF CADMIUM DURING PREGNANCY—AN EXPERIMENTALTOXAEMIA OF PREGNANCY'INDUCED BY CADMIUM SALTS. Journal of Reproduction and Fertility,1965,9(1):111-112.
    [71]Johnson, M. The effect of cadmium chloride on the blood-testis barrier of the guinea-pig. Journal of Reproduction and Fertility,1969.19(3):551-553.
    [72]Setchell, B. and G. Waites. Changes in the permeability of the testicular capillaries and of the'blood-testis barrier'after injection of cadmium chloride in the rat. Journal of Endocrinology,1970,47(1):81-86.
    [73]Lui, W.Y., CH Wong, DD Mruk, et al. TGF-P3 regulates the blood-testis barrier dynamics via the p38 mitogen activated protein (MAP) kinase pathway:an in vivo study. Endocrinology, 2003,144(4):1139-1142.
    [74]Dalgaard, M., C Nellemann, HR Lam, et al. The acute effects of mono (2-ethylhexyl) phthalate (MEHP) on testes of prepubertal Wistar rats. Toxicology letters,2001,122(1):69-79.
    [75]Moffit, J.S., BH Bryant, SJ Hallet, et al. Dose-dependent effects of sertoli cell toxicants 2, 5-hexanedione, carbendazim, and mono-(2-ethylhexyl) phthalate in adult rat testis. Toxicologic pathology,2007,35(5):719-727.
    [76]Yao, P.L., Y.C. Lin, and J.H. Richburg. Mono-(2-ethylhexyl) phthalate-induced disruption of junctional complexes in the seminiferous epithelium of the rodent testis is mediated by MMP2. Biology of reproduction,2010,82(3):516-527.
    [77]Oakberg, E.F. A description of spermiogenesis in the mouse and its use in analysis of the cycle of the seminiferous epithelium and germ cell renewal. American Journal of Anatomy,1956, 99(3):391-413.
    [78]Hess, R.A. Quantitative and qualitative characteristics of the stages and transitions in the cycle of the rat seminiferous epithelium:light microscopic observations of perfusion-fixed and plastic-embedded testes. Biology of reproduction,1990,43(3):525-542.
    [79]Hellsten, E., DJ Bernard, JW Owens, et al. Sertoli cell vacuolization and abnormal germ cell adhesion in mice deficient in an inositol polyphosphate 5-phosphatase. Biol Reprod,2002,66(5): 1522-30.
    [80]Lanning, L.L., DM Creasy, RE Chapin, et al. Recommended approaches for the evaluation of testicular and epididymal toxicity. Toxicologic pathology,2002,30(4):507-520.
    [81]Linder, R.E., LF Strader, VL Slott, et al. Endpoints of spermatotoxicity in the rat after short duration exposures to fourteen reproductive toxicants. Reproductive Toxicology,1992,6(6): 491-505.
    [82]Richburg, J.H. The relevance of spontaneous-and chemically-induced alterations in testicular germ cell apoptosis to toxicology. Toxicology letters,2000,112:79-86.
    [83]Lee, J., JH Richburg, EB Shipp, et al. The Fas System, a Regulator of Testicular Germ Cell Apoptosis, Is Differentially Up-Regulated in Sertoli Cell Versus Germ Cell Injury of the Testis 1. Endocrinology,1999,140(2):852-858.
    [84]Lee, J., JH Richburg, SC Younkin, et al. The Fas System Is a Key Regulator of Germ Cell Apoptosis in the Testis 1. Endocrinology,1997,138(5):2081-2088.
    [85]Schmelz, H.U., M Abend, K Kraft, et al. Fas/Fas ligand system and apoptosis induction in testicular carcinoma. Cancer,2002,95(1):73-81.
    [86]Koji, T., Y Hishikawa, H Ando, et al. Expression of Fas and Fas ligand in normal and ischemia-reperfusion testes:involvement of the Fas system in the induction of germ cell apoptosis in the damaged mouse testis. Biology of reproduction,2001,64(3):946-954.
    [87]Pentikainen V, E.K., Dunkel L. Fas regulates germ cell apoptosis in the human testis in vitro. American Journal of Physiology-Endocrinology and Metabolism,1999,276(2):E310-E316.
    [88]Theas, S., C. Rival, and L. Lustig. Germ cell apoptosis in autoimmune orchitis:involvement of the Fas-FasL system. American Journal of Reproductive Immunology,2003,50(2):166-176.
    [89]Ichimura, T., M. Kawamura, and A. Mitani. Co-localized expression of FasL, Fas, Caspase-3 and apoptotic DNA fragmentation in mouse testis after oral exposure to di (2-ethylhexyl) phthalate. Toxicology,2003,194(1):35-42.
    [90]D'Abrizio, P., E Baldini, PF Russoet, et al.Ontogenesis and cell specific localization of Fas ligand expression in the rat testis. International journal of andrology,2004,27(5):304-310.
    [91]Holash, J.A., SI Harik, G Perry, et al. Barrier properties of testis microvessels. Proceedings of the National Academy of Sciences,1993,90(23):11069-11073.
    [92]Harrison, R. The distribution of the vasal and cremasteric arteries to the testis and their functional importance. Journal of anatomy,1949,83(Pt 3):267.
    [93]Clegg, E. and I. Carr. Changes in the blood vessels of the rat testis and epididymis produced by cadmium chloride. The Journal of pathology and bacteriology,1967,94(2):317-322.
    [94]Skinner, M., P. Tung, and I. Fritz. Cooperativity between Sertoli cells and testicular peri tubular cells in the production and deposition of extracellular matrix components. The Journal of cell biology,1985,100(6):1941-1947.
    [95]Maekawa, M., K. Kamimura, and T. Nagano. Peritubular myoid cells in the testis:their structure and function. Archives of histology and cytology,1996,59(1):1-13.
    [96]Zhang, Y., L Lin, ZW Liu, et al. Disruption effects of monophthalate exposures on inter-Sertoli tight junction in a two-compartment culture model. Environmental toxicology,2008,23(3): 302-308.
    [97]Boekelheide, K., M.D. Neely, and T.M. Sioussat. The Sertoli cell cytoskeleton:a target for toxicant-induced germ cell loss. Toxicol Appl Pharmacol,1989,101(3):373-89.
    [98]Aumuller G, S.M., W Krause, et al.Distribution of vimentin-type intermediate filaments in Sertoli cells of the human testis, normal and pathologic. Anatomy and embryology,1988, 178(2):129-136.
    [99]Nieminen, M., T Henttinen, M Merinen, et al. Vimentin function in lymphocyte adhesion and transcellular migration. Nature cell biology,2006,8(2):156-162.
    [100]Zamoner, A., P Pierozan, LF Vidal, et al. Vimentin phosphorylation as a target of cell signaling mechanisms induced by la,25-dihydroxyvitamin D< sub> 3 in immature rat testes. Steroids,2008,73(14):1400-1408.
    [101]Upadhyay R, D'Souza R, Sonawane S, et al. Altered phosphorylation and distribution status of vimentin in rat seminiferous epithelium following 17β-estradiol treatment. Histochemistry and cell biology,2011,136(5):543-555.
    [102]Erkekoglu P, Zeybek N D, Giray B, et al.The effects of di (2-ethylhexyl) phthalate exposure and selenium nutrition on sertoli cell vimentin structure and germ-cell apoptosis in rat testis. Archives of environmental contamination and toxicology,2012,62(3):539-547.
    [103]Madekurozwa M C. Post-hatch Changes in the Immunoexpression of Desmin, Smooth Muscle Actin and Vimentin in the Testicular Capsule and Interstitial Tissue of the Japanese quail (Coturnix coturnix japonica). Anatomia, histologia, embryologia,2013,42(5):369-378.
    [104]Alam M S, Kurohmaru M. Disruption of Sertoli cell vimentin filaments in prepubertal rats:An acute effect of butylparaben< i> in vivo and< i> in vitro. Acta histochemica,2014.
    [105]ZHANG, L. and J.Z. Wei. Injuries of Cadmium Chloride on Ultrastructure of Rat Testis. Reproduction & Contraception,2006,9:002.
    [106]LIAO, X.G., Z SHI, C ZOU, et al. Effect of the cadmium on the expression of vimentin & E-cadherin in sertoli cell and the ultrastructure of blood-testis barrier in rats. Journal of Chongqing Medical University,2011,7:013.
    [107]Zhihu, S., L Xiaogang, Z Cong, et al. Protective effect of astragalus mongholicus against cadmium toxicity on the expression of vimentin & e-cadherin and the ultrastructure of sertoli cells in rats. Chinese Journal of Histochemistry and Cytochemistry,2012,3:016.
    [108]Choong G, Liu Y, Templeton D M. Interplay of Calcium and Cadmium in Mediating Cadmium Toxicity. Chemico-biological interactions,2014,211,54-65.
    [109]Wang Y, Yang C, Mao K, et al. Cellular localization of NLRP3 inflammasome. Protein & cell, 2013,4(6):425-431.
    [110]Hussein S, El-Sakhawy M, El-Saba A A, et al. Histological and Immunohistochemical studies of the effect of vitamin C and Nigella sativa on the palate of albino mice's offspring after cadmium exposure. Life Science Journal,2013,10(1).
    [411]Li, MW, DD Mruk, WM Lee, et al. Disruption of the blood-testis barrier integrity by bisphenol A in vitro:is this a suitable model for studying blood-testis barrier dynamics? The international journal of biochemistry & cell biology,2009,41(11):302-2314.
    [112]Salian, S., T. Doshi, and G. Vanage. Neonatal exposure of male rats to Bisphenol A impairs fertility and expression of sertoli cell junctional proteins in the testis. Toxicology,2009,265(1): 56-67.
    [113]Higaki S, Koyama Y, Shimada M, et al. Response to fish specific reproductive hormones and endocrine disrupting chemicals of a Sertoli cell line expressing endogenous receptors from an endemic cyprinid Gnathopogon caerulescens. General and comparative endocrinology, 2013,191,65-73.
    [114]Wan H T, Mruk D D, Wong C K C, et al. Perfluorooctanesulfonate (PFOS) Perturbs Male Rat Sertoli Cell Blood-Testis Barrier Function by Affecting F-Actin Organization via p-FAK-Tyr407:An in Vitro Study. Endocrinology,2013,155(1):249-262.
    [115]Ge L C, Chen Z J, Liu H Y, et al. Involvement of activating ERK1/2 through G protein coupled receptor 30 and estrogen receptor α/β in low doses of bisphenol A promoting growth of Sertoli TM4 cells. Toxicology letters,2014,226(1):81-89.
    [116]Chang L, She R, Ma L, et al. Acute testicular toxicity induced by melamine alone or a mixture of melamine and cyanuric acid in mice. Reproductive Toxicology,2014,46:1-11.
    [117]Monsees, T.K., M Franz, S Gebhardt, et al. Sertoli cells as a target for reproductive hazards. Andrologia,2000,32(4-5):239-246.
    [118]Fiorini, C., A Tilloy-Ellul, S Chevalier, et al. Sertoli cell junctional proteins as early targets for different classes of reproductive toxicants. Reproductive Toxicology,2004,18(3):413-421.
    [119]Xiao X, Mruk D D, Tang E I, et al. Environmental toxicants perturb human Sertoli cell adhesive function via changes in F-actin organization mediated by actin regulatory proteins. Human Reproduction,2014:deu011.
    [120]Guerrero-Bosagna C, Savenkova M, Haque M M, et al. Environmentally induced epigenetic transgenerational inheritance of altered Sertoli cell transcriptome and epigenome:molecular etiology of male infertility. PloS one,2013,8(3):e59922.
    [121]Piersma A H, Bosgra S, van Duursen M B M, et al. Evaluation of an alternative< i> in vitro test battery for detecting reproductive toxicants. Reproductive Toxicology,2013,38: 53-64.
    [122]de Liz Oliveira Cavalli V L, Cattani D, Heinz Rieg C E, et al. Roundup disrupts male reproductive functions by triggering calcium-mediated cell death in rat testis and Sertoli cells. Free Radical Biology and Medicine,2013,65:335-346.
    [123]Richburg, J.H. and K. Boekelheide. Mono-(2-ethylhexyl) phthalate rapidly alters both Sertoli cell vimentin filaments and germ cell apoptosis in young rat testes. Toxicology and Applied Pharmacology,1996,137(1):42-50.
    [124]Boekelheide, K., SL Fleming, KJ Johnson, et al.Role of Sertoli cells in injury-associated testicular germ cell apoptosis. Proceedings of the Society for Experimental Biology and Medicine,2000,225(2):105-115.
    [125]Wang, Z.Q., Y Watanabe, A Toki, et al.Altered distribution of Sertoli cell vimentin and increased apoptosis in cryptorchid rats. Journal of pediatric surgery,2002,37(4):648-652.
    [126]Guttenbach, M., C Steinlein, W Engel, et al. Cytogenetic characterization of the TM4 mouse Sertoli cell line. I. Conventional banding techniques, FISH and SKY. Cytogenetic and Genome Research,2001,94(1-2):71-78.
    [127]Kheradmand F, Nourmohammadi I, Ahmadi-Faghih M A, et al. Zinc and low-dose of cadmium protect sertoli cells against toxic-dose of cadmium:The role of metallothionein. Iranian Journal of Reproductive Medicine,2013,11(6).
    [128]Liu X, Nie S, Huang D, et al. Effects of nonylphenol exposure on expression of cell receptors and secretory function in mouse Sertoli TM4 cells. Environmental toxicology and pharmacology,2014,37(2):608-616.
    [129]Sato Y, Yoshida K, Nozawa S, et al. Establishment of adult mouse Sertoli cell lines by using the starvation method. Reproduction,2013,145(5):505-516.
    [130]Ge L C, Chen Z J, Liu H Y, et al. Involvement of activating ERK1/2 through G protein coupled receptor 30 and estrogen receptor α/β in low doses of bisphenol A promoting growth of Sertoli TM4 cells. Toxicology letters,2014,226(1):81-89.
    [131]Yang Y, Cao M, Wang Q, et al. The effects of 2-aminoethoxydiphenyl borate and diphenylboronic anhydride on gap junctions composed of connexin43 in TM4 sertoli cells. Biological and Pharmaceutical Bulletin,2011,34(9):1390.
    [132]Hanelt, M., M. Gareis, and B. Kollarczik. Cytotoxicity of mycotoxins evaluated by the MTT-cell culture assay. Mycopathologia,1994,128(3):167-174.
    [133]Hamid, R., Y Rotshteyn, L Rabadi, et al.Comparison of alamar blue and MTT assays for high through-put screening. Toxicology in vitro,2004,18(5):703-710.
    [134]Buch K, Peters T, Nawroth T, et al. Determination of cell survival after irradiation via clonogenic assay versus multiple MTT Assay-A comparative study. Radiation oncology,2012, 7(1):1-6.
    [135]Ciofani G, Danti S, D'Alessandro D, et al. Assessing cytotoxicity of boron nitride nanotubes: interference with the MTT assay. Biochemical and biophysical research communications,2010, 394(2):405-411.
    [136]van Meerloo J, Kaspers G J L, Cloos J. Cell sensitivity assays:the MTT assay[M]//Cancer Cell Culture. Humana Press,2011:237-245.
    [137]Wong, C.H., DD Mruk, W Lui, et al.Regulation of blood-testis barrier dynamics:an in vivo study. Journal of cell science,2004,117(5):783-798.
    [138]Wang, X.W., GR Ding, CH Shi, et al.Mechanisms involved in the blood testis barrier increased permeability induced by EMP. Toxicology,2010,276(1):58-63.
    [139]Vogl, A.W., K..S. Vaid, and J.A. Guttman. The Sertoli cell cytoskeleton.Molecular mechanisms in spermatogenesis,2008,636,186-211.
    [140]Lie P P Y, Cheng C Y, Mruk D D. Signalling pathways regulating the blood-testis barrier. The international journal of biochemistry & cell biology,2013,45(3):621-625.
    [141]Wong E W P, Cheng C Y. NCI domain of collagen α3 (Ⅳ) derived from the basement membrane regulates Sertoli cell blood-testis barrier dynamics. Spermatogenesis,2013,3(2).

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700