全氟辛烷磺酸(PFOS)对大鼠甲状腺激素的抑制作用及其机理的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
全氟辛烷磺酸(Perfluorooctane sulfonate, PFOS)是全氟表面活性剂的代表。PFOS曾被广泛应用于纺织、造纸、皮革、石油化工表面活性剂、医药品、电子产品等生产和日常生活各个领域。PFOS具有难降解性、远距离传播性、生物富集性及多种生物毒性,2009年5月,PFOS被正式列入《关于持久性有机污染物的斯德哥尔摩公约》。
     甲状腺激素(TH)能够调节哺乳动物各项生理机能,对胚胎及幼龄动物的生长发育(尤其是神经智力发育)至关重要。PFOS的甲状腺毒作用研究是其内分泌研究的重要组成部分,也是PFOS人群健康风险评价的重要内容。本研究旨在阐明:①胚胎期接触PFOS与出生后通过乳汁接触PFOS对幼龄动物甲状腺功能的影响以及影响程度的差别;②PFOS干扰大鼠TH的作用机制。围绕以上两个实验目的,本论文开展了如下研究工作:
     (1)建立交叉哺育(cross-foster)实验动物模型,形成仔鼠胚胎期和出生后均不暴露(对照组)及胚胎期暴露、出生后暴露、胚胎期和出生后均暴露三种不同形式接触PFOS的组别,将胚胎期与出生后暴露PFOS分离开来,考察不同方式接触PFOS对不同发育时期仔鼠甲状腺功能的影响,阐明哪种暴露方式对仔鼠甲状腺功能影响程度较大。实验结果表明,PFOS既可以通过胎盘在胚胎体内积累,又可以通过乳汁在新生幼鼠体内积累;出生前和出生后均暴露PFOS仔鼠在出生后14、21和35天血清总甲状腺素(TT4)水平均显著降低,与同龄对照比较,分别降低了36.7、24.6和37.3%;随体内PFOS浓度逐步增加,出生后暴露PFOS组仔鼠在14、21和35天血清TT4水平比对照分别降低了11.9、28.6和35.9%;尽管仅胚胎期暴露组仔鼠在出生后21和35天血清中PFOS浓度水平显著降低,但二个时间点TT4水平仍然显著性下降,分别低于相应对照组20.3和19.4%。多重比较分析表明,胚胎期暴露PFOS或出生后通过乳汁积累PFOS对出生后14和21仔鼠血清TT4的抑制程度无统计学差异p>0.05)。
     (2)考察了不同暴露方式下,PFOS对仔鼠甲状腺滤泡形态以及肝脏组织中与TH代谢、转运、细胞作用等过程相关基因表达的影响。实验结果表明,甲状腺滤泡腔各形态学参数未发生显著性改变p>0.05),PFOS未对仔鼠甲状腺滤泡形态产生明显影响;各TH相关基因中,只有TH结合蛋白TTR,在出生前、后均暴露组的出生后21天仔鼠肝脏中表达显著增强,比对照组TTR表达提高了50%。
     (3)建立91天亚慢性PFOS染毒实验模型,形成1.7、5.0和15.0 mg/L三个染毒剂量组,从TH合成和肝脏代谢角度考察PFOS干扰TH的作用机制。实验结果表明,PFOS染毒显著降低了血清TT4水平,并与染毒剂量呈明显剂量效应关系。15.0 mg/L组,TT4浓度水平仅为对照组的20%。PFOS对TH合成的关键酶甲状腺过氧化物酶活性未产生影响。PFOS上调了肝脏尿苷二磷酸葡萄糖醛酸转移酶A1型酶(UGT1A1)mRNA的表达,在中、高剂量组,UGT1A1表达水平约为相应对照组的2.2和2.3倍。PFOS显著降低了肝脏中脱碘酶(DIO1)基因的表达,在中、高剂量组,DIO1表达为相应对照组的86%和44%。相关分析表明,血清TT4水平与肝脏中UGT1A1和DIO1表达相关性良好(r值分别为-0.754和0.565;p值分别为0.002和0.023),提示PFOS可能增强了T4与肝脏UGT结合水平,提高T4代谢,导致血清TT4水平下降。
     (4)建立成年大鼠亚急性暴露PFOS实验模型,经口连续染毒5天,染毒剂量分别为0.2、1.0和3.0 mg/kg,同时设置PTU染毒组(10 mg/kg)和10 mg/kg PTU+3.0 mg/kgPFOS的联合染毒组。从TH肝胆排泄和血清结合蛋白(TTR)角度考察PFOS降低血清TT4的机制。实验结果表明,血清中TT4浓度随PFOS染毒剂量增大而逐步降低,但胆汁中TT4浓度未受PFOS影响。PFOS对血清TTR水平及甲状腺球蛋白(TG)水平无显著影响。3.0 mg/kg PFOS上调了肝脏中负责摄入T4有机阴离子转运蛋白OAPT2的表达,约为对照组的1.48倍;PFOS对负责将肝脏T4排至胆汁的多药耐药蛋白MRP2表达影响显著,MRP2表达在1.0和3.0 mg/kg PFOS处理组为对照组的1.81和1.70倍。同时,血清TT4水平与肝脏MRP2相对表达水平具有良好相关性(r=-0.528;p=0.12)。PFOS和PTU对TH抑制不存在显著的联合效应,PTU对TH的抑制强于PFOS。
     综上研究结果得出,胚胎期暴露PFOS或出生后通过乳汁暴露PFOS对断乳仔鼠血清TT4水平的抑制程度无统计学差异;PFOS未对TH合成过程产生干扰;PFOS主要通过增强肝脏对T4摄入和排出,增强T4葡萄糖醛酸化,使得T4的肝脏代谢增强和肝胆排泄提高,进而抑制血清T4水平。PFOS与经典的抗甲状腺肿药物PTU的甲状腺毒作用机制存在本质区别。
Perfluorooctane sulfonate (PFOS) is an environmentally persistent organic pollutant (POPs), which was widely used in industrial and consumer applications. Related animal experiments suggest that PFOS is associated with hepatotoxicity, carcinogenicity, reproductive and developmental effects, immunotoxicity neuroendocrine and neurobehavioural effects as well as hormone disruption. In May 2009, PFOS was added to the Stockholm Convention on POPs.
     Thyroid hormone is essential for the normal physiological function of virtually all tissues in mammals, playing important roles in regulating metabolism, growth and development, especially for the development of central nervous system and brain function. The present paper investigated that whether prenatal or postnatal exposure to PFOS alone can disturb the thyroid function in rat pups, and if so, which kind of exposure is a major cause of thyroid disruption. Besides, mechanisms of action by which PFOS interferes with thyroid systems in rats were also investigated. The present study comprise of several parts as follows:
     (1) A cross-foster animal model was established, yield the following four groups: unexposed control (CC), prenatal exposure (TC), postnatal exposure (CT), and prenatal+ postnatal exposure (TT). Serum and liver PFOS concentrations as well as serum TH levels in rat pups at the age of various postnatal days 0,7,14,21 or 35 under different conditions of PFOS exposure were evaluated. The occurrence of PFOS transfer from dams to pups, both via placenta and milk, was observed. Neither total T3 (TT3) nor rT3 in pups was affected by PFOS exposure. Gestational exposure to PFOS alone (TC) significantly (p< 0.05) decreased total T4 (TT4) level in pups on PNDs 21 and 35,20.3 and 19.4% lower than the control on the same PND, respectively. Having postnatal accumulation of PFOS, TT4 levels were 11.9,28.6, and 35.9% lower than controls on PNDs 14,21 and 35, respectively. No significant difference in TT4 level (p> 0.05) was observed between TC and CT on PND 21.
     (2) Thyroid gland colloid histomorphology paremeters and hepatic expression of genes involved in TH transport, metabolism and receptors were also investigated in rat pups at the age of various postnatal days under different conditions of PFOS exposure. The results demonstrate that none of the thyroid gland colloid histomorphology paremeters, including colloid area, longest axis, shortest axis, roundness and aspect ratio, was affected by PFOS. None of the selected TH related transcripts was affected by PFOS in pups on PND 0. Only transcript level of transthyretin, TH binding protein, in group TT significantly increased to 150% of the control on PND 21.
     (3) A subchronic PFOS exposure animal model was established. Male rats were exposed to 1.7,5.0 and 15.0 mg/L of PFOS in drinking water for 91 consecutive days. Thyroid and liver were removed for the measurement of the endpoints closely related to TH biosynthesis and metabolism following PFOS exposure. The results showed that serum TT4 level decreased significantly at all applied dosages. Hepatic UGT1A1 mRNA but not UGT1A6 was up-regulated at 5.0 and 15.0 mg/L of PFOS. Correlation coefficients revealed that hepatic UGT1A1 mRNA expression correlated with TT4 level (r=-0.754; p= 0.002). Treatment with PFOS lowered hepatic DIO1 mRNA at 15.0 mg/L but increased thyroidal DIO1 mRNA dose-dependently. The activity of TPO was not affected by PFOS. NIS and TSHR mRNA in thyroid increased slightly but not significantly. These results indicate that increased hepatic T4 glucuronidation via UGT1A1 and increased thyroidal conversion of T4 to T3 via DIO1 were in part responsible for PFOS-induced hypothyroxinemia in rats.
     (4) A 5 day PFOS exposure was to further identify the major factors contributing to PFOS-induced TH decrease in rats.The consequence of exposure to PTU was also analyzed as a positive control of TH inhibition. The animals were given either (1) vehicle; (2) PFOS; (3) PTU; or (4) PTU+PFOS once a day by gavage for 5 consecutive days. Parameters including contents of TT4 and TT3 in both serum and bile, serum concentrations of transthyretin and thyroglobulin, as well as transcripts of transporters involved in hepatic uptake and efflux of T4 were determined in control and PFOS exposed groups. TT4 and TT3 were also analyzed in PTU and PTU+PFOS groups to reflect the different hormone effects between PFOS, PTU, and PFOS+PTU. Results showed that serum TT4 and TT3 decreased, while bile TT4 and TT3 remained stable following PFOS exposure. Exposure to 3.0 mg/kg of PFOS enhanced hepatic organic anions transporter OATP2 mRNA expression (1.43 times of control). Treatment with PFOS increased hepatic expression of multidrug resistance-associated protein MRP2, approximately 1.80 and 1.69 times of control in 1.0 and 3.0 mg/kg groups, respectively. Spearman's correlation coefficients revealed that MRP2 mRNA expression correlated well with serum TT4 level (r=-0.528,p= 0.012). Serum thyroglobulin and transthyretin levels remained stable. In addition to serum TT4, PFOS and PTU groups demonstrated significantly difference with each other with respect to serum TT3, bile TT4, and bile TT3. No significant differences of TT4 and TT3 in both serum and bile were observed between PTU and PTU+PFOS (p> 0.05).
     In conclusion, gestational and lactational exposure to PFOS depresses TT4 in weaned rat to a similar extent. PFOS has little effects on the synthesis of TH. PFOS-induced TT4 deficiency is mainly due to increased hepatic uptake of T4 and enhanced hepatic metabolism and excretion of T4, which is probably different from the classic goitrogen, PTU.
引文
[1]Kavlock RJ, Daston GP, DeRosa C, et al. Research needs for the risk assessment of health and environmental effects of endocrine disruptors:a report of the U.S. EPA-sponsored workshop. Environmental Health Perspectives 1996,104 Suppl 4:715-740.
    [2]Crisp TM, Clegg ED, Cooper RL, et al. Environmental endocrine disruption:an effects assessment and analysis. Environmental Health Perspectives 1998,106 Suppl 1:11-56.
    [3]Brucker-Davis F. Effects of environmental synthetic chemicals on thyroid function. Thyroid 1998,8 (9): 827-856.
    [4]Guillette LJ, Jr., Gross TS, Masson GR, et al. Developmental abnormalities of the gonad and abnormal sex hormone concentrations in juvenile alligators from contaminated and control lakes in Florida. Environmental Health Perspectives 1994,102 (8):680-688.
    [5]Parks LG, Lambright CS, Orlando EF, et al. Masculinization of female mosquitofish in Kraft mill effluent-contaminated Fenholloway River water is associated with androgen receptor agonist activity. Toxicological Sciences 2001,62 (2):257-267.
    [6]Schroder-van der Elst JP, van der Heide D, Romijn JA, et al. Differential effects of natural flavonoids on growth and iodide content in a human Na+/(?) symporter-transfected follicular thyroid carcinoma cell line. European Journal of Endocrinology 2004,150 (4):557-564.
    [7]Ferreira AC, Lisboa PC, Oliveira KJ, et al. Inhibition of thyroid type 1 deiodinase activity by flavonoids. Food and Chemical Toxicology 2002,40 (7):913-917.
    [8]Gaitan E. Flavonoids and the thyroid. Nutrition 1996,12 (2):127-129.
    [9]van der Heide D, Kastelijn J, Schroder-van der Elst JP. Flavonoids and thyroid disease. Biofactors 2003, 19(3-4):113-119.
    [10]Renner R. Growing concern over perfluorinated chemicals. Environmental Science and Technology 2001,35(7):154-160.
    [11]Seacat AM, Thomford PJ, Hansen KJ, et al. Subchronic toxicity studies on perfluorooctanesulfonate potassium salt in cynomolgus monkeys. Toxicological Sciences 2002,68 (1):249-264.
    [12]Organization for Economic Co-operation and Development.2002. Co-operation on existing chemicals: hazard assessment of perfluorooctane sulfonate (PFOS) and its salts. U.S. Environmental Protection Agency docket AR-226-1140. Paris, France. Available:http://www.oecd.org/dataoecd/23/18/2382880. pdf.
    [13]Beach SA, Newsted JL, Coady K, et al. Ecotoxicological evaluation of perfluorooctanesulfonate (PFOS). Reviews of Environment Contamination and Toxicology 2006,186:133-174.
    [14]Nakayama S, Harada K, Inoue K, et al. Distributions of perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) in Japan and their toxicities. Environmental Science 2005,12 (6): 293-313.
    [15]Jahnke A, Berger U, Ebinghaus R, et al. Latitudinal gradient of airborne polyfluorinated alkyl substances in the marine atmosphere between Germany and South Africa (53 degrees N-33 degrees S). Environmental Science and Technology 2007,41 (9):3055-3061.
    [16]Young CJ, Furdui VI, Franklin J, et al. Perfluorinated acids in Arctic snow:new evidence for atmospheric formation. Environmental Science and Technology 2007,41 (10):3455-3461.
    [17]Liu W, Jin Y, Quan X, et al. Perfluorosulfonates and perfluorocarboxylates in snow and rain in Dalian, China. Environment International 2009,35 (4):737-742.
    [18]Jin YH, Liu W, Sato I, et al. PFOS and PFOA in environmental and tap water in China. Chemosphere 2009,77 (5):605-611.
    [19]Bao J, Jin Y, Liu W, et al. Perfluorinated compounds in sediments from the Daliao River system of northeast China. Chemosphere 2009,77 (5):652-657.
    [20]Yamashita N, Kannan K, Taniyasu S, et al. A global survey of perfluorinated acids in oceans. Marine Pollution Bulletin 2005,51 (8-12):658-668.
    [21]Ju X, Jin Y, Sasaki K, et al. Perfluorinated surfactants in surface, subsurface water and microlayer from Dalian Coastal waters in China. Environmental Science and Technology 2008,42 (10): 3538-3542.
    [22]Moriwaki H, Takatah Y, Arakawa R. Concentrations of perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) in vacuum cleaner dust collected in Japanese homes. Journal of Environmental Monitoring 2003,5 (5):753-757.
    [23]de Vos MG, Huijbregts MA, van den Heuvel-Greve MJ, et al. Accumulation of perfluorooctane sulfonate (PFOS) in the food chain of the Western Scheldt estuary:Comparing field measurements with kinetic modeling. Chemosphere 2008,70 (10):1766-1773.
    [24]Han X, Snow TA, Kemper RA, et al. Binding of perfluorooctanoic acid to rat and human plasma proteins. Chemical Research in Toxicology 2003,16 (6):775-781.
    [25]Jones PD, Hu W, De Coen W, et al. Binding of perfluorinated fatty acids to serum proteins. Environmental Toxicology and Chemistry 2003,22 (11):2639-2649.
    [26]Ericson I, Marti-Cid R, Nadal M, et al. Human exposure to perfluorinated chemicals through the diet: intake of perfluorinated compounds in foods from the Catalan (Spain) market. Journal of Agricultural and Food Chemistry 2008,56 (5):1787-1794.
    [27]Giesy JP, Kannan K. Global distribution of perfluorooctane sulfonate in wildlife. Environmental Science and Technology 2001,35 (7):1339-1342.
    [28]Olivero-Verbel J, Tao L, Johnson-Restrepo B, et al. Perfluorooctanesulfonate and related fluorochemicals in biological samples from the north coast of Colombia. Environmental Pollution 2006,142 (2):367-372.
    [29]Verreault J, Houde M, Gabrielsen GW, et al. Perfluorinated alkyl substances in plasma, liver, brain, and eggs of glaucous gulls (Larus hyperboreus) from the Norwegian arctic. Environmental Science and Technology 2005,39 (19):7439-7445.
    [30]Butenhoff JL, Olsen GW, Pfahles-Hutchens A. The applicability of biomonitoring data for perfluorooctanesulfonate to the environmental public health continuum. Environmental Health Perspectives 2006,114 (11):1776-1782.
    [31]Fromme H, Tittlemier SA, Volkel W, et al. Perfluorinated compounds-exposure assessment for the general population in Western countries. International Journal of Hygiene and Environmental Health 2009,212 (3):239-270.
    [32]Tittlemier SA, Pepper K, Seymour C, et al. Dietary exposure of Canadians to perfluorinated carboxylates and perfluorooctane sulfonate via consumption of meat, fish, fast foods, and food items prepared in their packaging. Journal of Agricultural and Food Chemistry 2007,55 (8):3203-3210.
    [33]Halldorsson TI, Fei C, Olsen J, et al. Dietary predictors of perfluorinated chemicals:a study from the Danish National Birth Cohort. Environmental Science and Technology 2008,42 (23):8971-8977.
    [34]Olsen GW, Church TR, Hansen KJ, et al. Quantitative evaluation of perfluorooctanesulfonate (PFOS) and other fluorochemicals in the serum of children. Journal of Child Health 2004,2:53-76.
    [35]Olsen GW, Church TR, Larson EB, et al. Serum concentrations of perfluorooctanesulfonate and other fluorochemicals in an elderly population from Seattle, Washington. Chemosphere 2004,54 (11): 1599-1611.
    [36]Olsen GW, Church TR, Miller JP, et al. Perfluorooctanesulfonate and other fluorochemicals in the serum of American Red Cross adult blood donors. Environmental Health Perspectives 2003,111 (16): 1892-1901.
    [37]Harada K, Saito N, Inoue K, et al. The influence of time, sex and geographic factors on levels of perfluorooctanesulfonate and perfluorooctanoate in human serum over the last 25 years. Journal of Occupational Health 2004,46:141-147.
    [38]Yeung LW, So MK, Jiang G, et al. Perfluorooctanesulfonate and related fluorochemicals in human blood samples from China. Environmental Science and Technology 2006,40 (3):715-720.
    [39]Jin Y, Saito N, Harada KH, et al. Historical trends in human serum levels of perfluorooctanoate and perfluorooctane sulfonate in Shenyang, China. The Tohoku Journal of Experimental Medicine 2007, 212(1):63-70.
    [40]Lau C, Anitole K, Hodes C, et al. Perfluoroalkyl acids:a review of monitoring and toxicological findings. Toxicological Sciences 2007,99 (2):366-394.
    [41]Olsen GW, Huang HY, Helzlsouer KJ, et al. Historical comparison of perfluorooctanesulfonate, perfluorooctanoate, and other fluorochemicals in human blood. Environmental Health Perspectives 2005,113 (5):539-545.
    [42]Kubwabo C, Vais N, Benoit FM. A pilot study on the determination of perfluorooctanesulfonate and other perfluorinated compounds in blood of Canadians. Journal of Environmental Monitoring 2004,6 (6):540-545.
    [43]Calafat AM, Wong LY, Kuklenyik Z, et al. Polyfluoroalkyl chemicals in the U.S. population:data from the National Health and Nutrition Examination Survey (NHANES) 2003-2004 and comparisons with NHANES 1999-2000. Environmental Health Perspectives 2007,115(11):1596-1602.
    [44]Olsen GW, Mair DC, Reagen WK, et al. Preliminary evidence of a decline in perfluorooctanesulfonate (PFOS) and perfluorooctanoate (PFOA) concentrations in American Red Cross blood donors. Chemosphere 2007,68 (1):105-111.
    [45]Inoue K, Okada F, Ito R, et al. Determination of perfluorooctane sulfonate, perfluorooctanoate and perfluorooctane sulfonylamide in human plasma by column-switching liquid chromatography-electrospray mass spectrometry coupled with solid-phase extraction. Biomedical and Life Sciences 2004,810(1):49-56.
    [46]Kannan K, Corsolini S, Falandysz J, et al. Perfluorooctanesulfonate and related fluorochemicals in human blood from several countries. Environmental Science and Technology 2004,38 (17): 4489-4495.
    [47]Fromme H, Schlummer M, Moller A, et al. Exposure of an adult population to perfluorinated substances using duplicate diet portions and biomonitoring data. Environmental Science and Technology 2007,41 (22):7928-7933.
    [48]Holzer J, Midasch O, Rauchfuss K, et al. Biomonitoring of perfluorinated compounds in children and adults exposed to perfluorooctanoate-contaminated drinking water. Environmental Health Perspectives 2008,116 (5):651-657.
    [49]Olsen GW, Logan PW, Hansen KJ, et al. An occupational exposure assessment of a perfluorooctanesulfonyl fluoride production site:biomonitoring. American Industrial Hygiene Association 2003,64 (5):651-659.
    [50]Olsen GW, Burris JM, Burlew MM, et al. Epidemiologic assessment of worker serum perfluorooctanesulfonate (PFOS) and perfluorooctanoate (PFOA) concentrations and medical surveillance examinations. Journal of Occupational Environmental Medicine 2003,45 (3):260-270.
    [51]Jin Y, Liu X, Li T, et al. Status of perfluorochemicals in adult serum and umbilical blood in Shenyang. Wei Sheng Yan Jiu 2004,33 (4):481-483.
    [52]Inoue K, Okada F, Ito R, et al. Perfluorooctane sulfonate (PFOS) and related perfluorinated compounds in human maternal and cord blood samples:assessment of PFOS exposure in a susceptible population during pregnancy. Environmental Health Perspectives 2004,112 (11):1204-1207.
    [53]Apelberg BJ, Witter FR, Herbstman JB, et al. Cord serum concentrations of perfluorooctane sulfonate (PFOS) and perfluorooctanoate (PFOA) in relation to weight and size at birth. Environmental Health Perspectives 2007,115 (11):1670-1676.
    [54]Tao L, Kannan K, Wong CM, et al. Perfluorinated compounds in human milk from Massachusetts, U.S.A. Environmental Science and Technology 2008,42 (8):3096-3101.
    [55]So MK, Yamashita N, Taniyasu S, et al. Health risks in infants associated with exposure to perfluorinated compounds in human breast milk from Zhoushan, China. Environmental Science and Technology 2006,40 (9):2924-2929.
    [56]Volkel W, Genzel-Boroviczeny O, Demmelmair H, et al. Perfluorooctane sulphonate (PFOS) and perfluorooctanoic acid (PFOA) in human breast milk:results of a pilot study. International Journal of Hygiene and Environmental Health 2008,211 (3-4):440-446.
    [57]Karrman A, Ericson I, van Bavel B, et al. Exposure of perfluorinated chemicals through lactation: levels of matched human milk and serum and a temporal trend,1996-2004, in Sweden. Environmental Health Perspectives 2007,115 (2):226-230.
    [58]Midasch O, Drexler H, Hart N, et al. Transplacental exposure of neonates to perfluorooctanesulfonate and perfluorooctanoate:a pilot study. International Archives of Occupational and Environmental Health 2007,80 (7):643-648.
    [59]Fei C, McLaughlin JK, Tarone RE, et al. Fetal growth indicators and perfluorinated chemicals:a study in the Danish National Birth Cohort. American Journal of Epidemiology 2008,168 (1):66-72.
    [60]Apelberg BJ, Goldman LR, Calafat AM, et al. Determinants of fetal exposure to polyfluoroalkyl compounds in Baltimore, Maryland. Environmental Science and Technology 2007,41 (11):3891-3897.
    [61]Karrman, A.; Domingo, J. L.; Llebaria, X.; Nadal, M.; Bigas, E.; van Bavel, B.; Lindstrom, G., Biomonitoring perfluorinated compounds in Catalonia, Spain:concentrations and trends in human liver and milk samples. Environmental Science and Polluiont Reseach International 2009,17, (3),750-758.
    [62]Luebker DJ, Hansen KJ, Bass NM, et al. Interactions of fluorochemicals with rat liver fatty acid-binding protein. Toxicology 2002,176(3):175-185.
    [63]Giesy JP, Kannan K. Perfluorochemical surfactants in the environment. Environmental Science and Technology 2002,36 (7):146-152.
    [64]Cui L, Liao CY, Zhou QF, et al. Excretion of PFOA and PFOS in male rats during a subchronic exposure. Archives of Environment Contamination and Toxicology,58 (1):205-213.
    [65]Newsted JL, Beach SA, Gallagher SP, et al. Pharmacokinetics and acute lethality of perfluorooctanesulfonate (PFOS) to juvenile mallard and northern bobwhite. Archives of Environment Contamination and Toxicology 2006,50 (3):411-420.
    [66]Seacat AM, Thomford PJ, Hansen KJ, et al. Sub-chronic dietary toxicity of potassium perfluorooctanesulfonate in rats. Toxicology 2003,183 (1-3):117-131.
    [67]Berthiaume J, Wallace KB. Perfluorooctanoate, perflourooctanesulfonate, and N-ethyl perfluorooctanesulfonamido ethanol; peroxisome proliferation and mitochondrial biogenesis. Toxicology Letters 2002,129 (1-2):23-32.
    [68]Hu W, Jones PD, Upham BL, et al. Inhibition of gap junctional intercellular communication by perfluorinated compounds in rat liver and dolphin kidney epithelial cell lines in vitro and Sprague-Dawley rats in vivo. Toxicological Sciences 2002,68 (2):429-436.
    [69]Lau C, Thibodeaux JR, Hanson RG, et al. Exposure to perfluorooctane sulfonate during pregnancy in rat and mouse. Ⅱ:postnatal evaluation. Toxicological Sciences 2003,74 (2):382-392.
    [70]Thibodeaux JR, Hanson RG, Rogers JM, et al. Exposure to perfluorooctane sulfonate during pregnancy in rat and mouse. I:maternal and prenatal evaluations. Toxicological Sciences 2003,74 (2): 369-381.
    [71]Luebker DJ, York RG, Hansen KJ, et al. Neonatal mortality from in utero exposure to perfluorooctanesulfonate (PFOS) in Sprague-Dawley rats:dose-response, and biochemical and pharamacokinetic parameters. Toxicology 2005,215 (1-2):149-169.
    [72]Yu WG, Liu W, Jin YH. Effects of Perfluorooctane Sulfonate on Rat Thyroid Hormone Biosynthesis and Metabolism. Environmental Toxicology and Chemistry 2009,28 (5):990-996.
    [73]Griffith FD, Long JE. Animal toxicity studies with ammonium perfluorooctanoate. American Industrial Hygiene Association Journal 1980,41 (8):576-583.
    [74]Case MT, York RG, Christian MS. Rat and rabbit oral developmental toxicology studies with two perfluorinated compounds. International Journal of Toxicology 2001,20 (2):101-109.
    [75]Luebker DJ, Case MT, York RG, et al. Two-generation reproduction and cross-foster studies of perfluorooctanesulfonate (PFOS) in rats. Toxicology 2005,215 (1-2):126-148.
    [76]Grasty RC, Wolf DC, Grey BE, et al. Prenatal window of susceptibility to perfluorooctane sulfonate-induced neonatal mortality in the Sprague-Dawley rat. Birth Defects Research Part B Developmental and Reproductive Toxicology 2003,68 (6):465-471.
    [77]Grasty RC, Bjork JA, Wallace KB, et al. Effects of prenatal perfluorooctane sulfonate (PFOS) exposure on lung maturation in the perinatal rat. Birth Defects Research Part B Developmental and Reproductive Toxicology 2005,74 (5):405-416.
    [78]Fei C, McLaughlin JK, Lipworth L, et al. Prenatal exposure to perfluorooctanoate (PFOA) and perfluorooctanesulfonate (PFOS) and maternally reported developmental milestones in infancy. Environmental Health Perspectives 2008,116 (10):1391-1395.
    [79]刘冰,于麒麟,金一和等.全氟辛烷磺酸对大鼠海马神经细胞内钙离子浓度的影响.毒理学杂志2005,(S1):225-226.
    [80]Kanzaki M, Zhang YQ, Mashima H, et al. Translocation of a calcium-permeable cation channel induced by insulin-like growth factor-I. Nature Cell Biology 1999,1 (3):165-170.
    [81]Harada KH, Ishii TM, Takatsuka K, et al. Effects of perfluorooctane sulfonate on action potentials and currents in cultured rat cerebellar Purkinje cells. Biochemical and Biophysical Research Communications 2006,351 (1):240-245.
    [82]Liu X, Liu W, Jin Y, et al. Effect of gestational and lactational exposure to perfluorooctanesulfonate on calcium-dependent signaling molecules gene expression in rats'hippocampus. Archives of Toxicology 2010, DOI:10.1007/s00204-010-0517-9.
    [83]Yang Q, Xie Y, Depierre JW. Effects of peroxisome proliferators on the thymus and spleen of mice. Clinical and Experimental Immunology 2000,122 (2):219-226.
    [84]Yang Q, Xie Y, Alexson SE, et al. Involvement of the peroxisome proliferator-activated receptor alpha in the immunomodulation caused by peroxisome proliferators in mice. Biochemical Pharmacology 2002,63(10):1893-1900.
    [85]Zheng L, Dong GH, Jin YH, et al. Immunotoxic changes associated with a 7-day oral exposure to perfluorooctanesulfonate (PFOS) in adult male C57BL/6 mice. Archives of Toxicology 2009,83 (7): 679-689.
    [86]Kannan K, Perrotta E, Thomas NJ. Association between perfluorinated compounds and pathological conditions in southern sea otters. Environmental Science and Technology 2006,40 (16):4943-4948.
    [87]Xie Y, Yang Q, Nelson BD, et al. Characterization of the adipose tissue atrophy induced by peroxisome proliferators in mice. Lipids 2002,37 (2):139-146.
    [88]Alexander BH, Olsen GW, Burris JM, et al. Mortality of employees of a perfluorooctanesulphonyl fluoride manufacturing facility. Occupational and Environmenal Medicine 2003,60 (10):722-729.
    [89]张颖花,范轶欧,麻懿馨等.全氟辛烷磺酸对大鼠血清T3,T4和TSH的影响.中国公共卫生2005,21(6):707.
    [90]Ribes D, Fuentes S, Torrente M, et al. Combined effects of perfluorooctane sulfonate (PFOS) and maternal restraint stress on hypothalamus adrenal axis (HPA) function in the offspring of mice. Toxicology and Applied Pharmacology 2009,243 (1):13-18.
    [91]Austin ME, Kasturi BS, Barber M, et al. Neuroendocrine effects of perfluorooctane sulfonate in rats. Environmental Health Perspectives 2003,111 (12):1485-1489.
    [92]Jernbro S, Rocha PS, Keiter S, et al. Perfluorooctane sulfonate increases the genotoxicity of cyclophosphamide in the micronucleus assay with V79 cells. Further proof of alterations in cell membrane properties caused by PFOS. Environmental Science and Pollution Research International 2007,14 (2):85-87.
    [93]Hu W, Jones PD, DeCoen W, et al. Alterations in cell membrane properties caused by perfluorinated compounds. Comparative Biochemistry and Physiology Part C:Toxicology and Pharmacology 2003, 135(1):77-88.
    [94]陈晓丽,洪亮,王贺珍.欧盟PFOS指令对我国纺织业和皮革行业的影响及对策.Standard Science2009,2009(10):84-87.
    [95]Schussler GC. The thyroxine-binding proteins. Thyroid 2000,10 (2):141-149.
    [96]Klaassen CD. Toxicology:The Basic Science of Poisons.2001.
    [97]Mackenzie PI, Bock KW, Burchell B, et al. Nomenclature update for the mammalian UDP glycosyltransferase (UGT) gene superfamily. Pharmacogenet Genomics 2005,15 (10):677-685.
    [98]You L. Steroid hormone biotransformation and xenobiotic induction of hepatic steroid metabolizing enzymes. Chemico-Biological Interactions 2004,147 (3):233-246.
    [99]Bianco AC, Salvatore D, Gereben B, et al. Biochemistry, cellular and molecular biology, and physiological roles of the iodothyronine selenodeiodinases. Endocrine Reviews 2002,23 (1):38-89.
    [100]Yen PM. Physiological and molecular basis of thyroid hormone action. Physiological Reviews 2001, 81 (3):1097-1142.
    [101]Boas M, Feldt-Rasmussen U, Skakkebaek NE, et al. Environmental chemicals and thyroid function. European Journal of Endocrinology 2006,154 (5):599-611.
    [102]Zoeller RT, Crofton KM. Mode of action:developmental thyroid hormone insufficiency-neurological abnormalities resulting from exposure to propylthiouracil. Critical Reviews in Toxicology 2005,35 (8-9):771-781.
    [103]Koibuchi N, Chin WW. Thyroid hormone action and brain development. Trends in Endocrinology and Metabolism 2000,11 (4):123-128.
    [104]Sher ES, Xu XM, Adams PM, et al. The effects of thyroid hormone level and action in developing brain:are these targets for the actions of polychlorinated biphenyls and dioxins? Toxicology and Industrial Health 1998,14 (1-2):121-158.
    [105]Bernal J. Action of thyroid hormone in brain. Journal of Endocrinological Investigation 2002,25 (3): 268-288.
    [106]de Escobar GM, Obregon MJ, del Rey FE. Maternal thyroid hormones early in pregnancy and fetal brain development. Best Practice and Research Clinical Endocrinology and Metabolism 2004,18 (2): 225-248.
    [107]Wiaderna D, Tomas T. Assessment of long-term effects of exposure to toluene based on the analysis of selected behavioral responses with particular reference to the ability to trigger behavioral hypersensitivity in rats. International Journal of Occupational Medicine and Environmental Health 2002,15 (3):239-245.
    [108]Sanchez-Santed F, Canadas F, Flores P, et al. Long-term functional neurotoxicity of paraoxon and chlorpyrifos:behavioural and pharmacological evidence. Neuro-toxicology and Teratology 2004,26 (2):305-317.
    [109]Capen CC. Correlation of mechanistic data and histopathology in the evaluation of selected toxic endpoints of the endocrine system. Toxicology Letters 1998,102:405-409.
    [110]Shi Y-B, Ritchie JWA, Taylor PM. Complex regulation of thyroid hormone action:multiple opportunities for pharmacological intervention. Pharmacology & Therapeutics 2002,94 (3):235-251.
    [111]Wolff J. Perchlorate and the thyroid gland. Pharmacological Reviews 1998,50 (1):89-105.
    [112]Divi RL, Doerge DR. Inhibition of thyroid peroxidase by dietary flavonoids. Chemical Research in Toxicology 1996,9(1):16-23.
    [113]Curran PG, DeGroot LJ. The effect of hepatic enzyme-inducing drugs on thyroid hormones and the thyroid gland. Endocrine Reviews 1991,12 (2):135-150.
    [114]Kretschmer XC, Baldwin WS. CAR and PXR:xenosensors of endocrine disrupters? Chemico-Biological Interactions 2005,155 (3):111-128.
    [115]Van den Berg M, Birnbaum LS, Denison M, et al. The 2005 World Health Organization reevaluation of human and Mammalian toxic equivalency factors for dioxins and dioxin-like compounds. Toxicological Sciences 2006,93 (2):223-241.
    [116]Tabb MM, Blumberg B. New modes of action for endocrine-disrupting chemicals. Molecular Endocrinology 2006,20 (3):475-482.
    [117]Qatanani M, Zhang J, Moore DD. Role of the constitutive androstane receptor in xenobiotic-induced thyroid hormone metabolism. Endocrinology 2005,146 (3):995-1002.
    [118]Vansell NR, Klaassen CD. Increase in rat liver UDP-glucuronosyltransferase mRNA by microsomal enzyme inducers that enhance thyroid hormone glucuronidation. Drug Metabolism and Disposition 2002,30 (3):240-246.
    [119]McClain RM, Levin AA, Posch R, et al. The effect of phenobarbital on the metabolism and excretion of thyroxine in rats. Toxicology and Applied Pharmacology 1989,99 (2):216-228.
    [120]Barter RA, Klaassen CD. Reduction of thyroid hormone levels and alteration of thyroid function by four representative UDP-glucuronosyltransferase inducers in rats. Toxicology and Applied Pharmacology 1994,128 (1):9-17.
    [121]Liu J, Liu Y, Barter RA, et al. Alteration of thyroid homeostasis by UDP-glucuronosyltransferase inducers in rats:a dose-response study. Journal of Pharmacology and Experimental Therapeutics 1995, 273 (2):977-985.
    [122]Kohrle J. Iodothyronine deiodinases. Methods in Enzymology 2002,347:125-167.
    [123]Capen CC. Mechanistic data and risk assessment of selected toxic end points of the thyroid gland. Toxicologic Pathology 1997,25 (1):39-48.
    [124]Schmutzler C, Hamann I, Hofmann PJ, et al. Endocrine active compounds affect thyrotropin and thyroid hormone levels in serum as well as endpoints of thyroid hormone action in liver, heart and kidney. Toxicology 2004,205 (1-2):95-102.
    [125]Meerts IA, van Zanden JJ, Luijks EA, et al. Potent competitive interactions of some brominated flame retardants and related compounds with human transthyretin in vitro. Toxicological Sciences 2000,56(1):95-104.
    [126]Hamers T, Kamstra JH, Sonneveld E, et al. In vitro profiling of the endocrine-disrupting potency of brominated flame retardants. Toxicological Sciences 2006,92 (1):157-173.
    [127]Wong H, Lehman-McKeeman LD, Grubb MF, et al. Increased hepatobiliary clearance of unconjugated thyroxine determines DMP 904-induced alterations in thyroid hormone homeostasis in rats. Toxicological Sciences 2005,84 (2):232-242.
    [128]Saghir SA, Charles GD, Bartels MJ, et al. Mechanism of trifluralin-induced thyroid tumors in rats. Toxicology Letters 2008,180 (1):38-45.
    [129]Mitchell AM, Tom M, Mortimer RH. Thyroid hormone export from cells:contribution of P-glycoprotein. Journal of Endocrinology 2005,185 (1):93-98.
    [130]Richardson VM, Staskal DF, Ross DG, et al. Possible mechanisms of thyroid hormone disruption in mice by BDE 47, a major polybrominated diphenyl ether congener. Toxicology and Applied Pharmacology 2008,226 (3):244-250.
    [131]Brouwer A, Morse DC, Lans MC, et al. Interactions of persistent environmental organohalogens with the thyroid hormone system:mechanisms and possible consequences for animal and human health. Toxicology and Industrial Health 1998,14 (1-2):59-84.
    [132]da Costa VM, Moreira DG, Rosenthal D. Thyroid function and aging:gender-related differences. Journal of Endocrinology 2001,171 (1):193-198.
    [133]St Germain DL, Galton VA. Comparative study of pituitary-thyroid hormone economy in fasting and hypothyroid rats. Journal of Clinical Investigation 1985,75 (2):679-688.
    [134]Santini F, Vitti P, Ceccarini G, et al. In vitro assay of thyroid disruptors affecting TSH-stimulated adenylate cyclase activity. Journal of Endocrinology Investigation 2003,26 (10):950-955.
    [135]Breous E, Wenzel A, Loos U. The promoter of the human sodium/iodide symporter responds to certain phthalate plasticisers. Molecular and Cell Endocrinology 2005,244 (1-2):75-78.
    [136]Schuur AG, Legger FF, van Meeteren ME, et al. In vitro inhibition of thyroid hormone sulfation by hydroxylated metabolites of halogenated aromatic hydrocarbons. Chemical Research in Toxicology 1998,11 (9):1075-1081.
    [137]Schmutzler C, Hamann I, Hofmann PJ, et al. Endocrine active compounds affect thyrotropin and thyroid hormone levels in serum as well as endpoints of thyroid hormone action in liver, heart and kidney. Toxicology 2004,205 (1-2):95-102.
    [138]Cho SD, Kim JH, Kim DY, et al. Pre-validation study for OECD enhanced test guideline 407 protocol by gavage for 4 weeks using propylthiouracil and tamoxifen. Toxicology Letters 2003,144 (2):195-204.
    [139]Purkey HE, Palaninathan SK, Kent KC, et al. Hydroxylated polychlorinated biphenyls selectively bind transthyretin in blood and inhibit amyloidogenesis:rationalizing rodent PCB toxicity. Chemistry and Biology 2004,11 (12):1719-1728.
    [140]Yamauchi K, Ishihara A, Fukazawa H, et al. Competitive interactions of chlorinated phenol compounds with 3,3',5-triiodothyronine binding to transthyretin:detection of possible thyroid-disrupting chemicals in environmental waste water. Toxicology and Applied Pharmacology 2003,187(2):110-117.
    [141]Kudo Y, Yamauchi K. In vitro and in vivo analysis of the thyroid disrupting activities of phenolic and phenol compounds in Xenopus laevis. Toxicological Sciences 2005,84 (1):29-37.
    [142]van den Berg KJ. Interaction of chlorinated phenols with thyroxine binding sites of human transthyretin, albumin and thyroid binding globulin. Chemico-Biological Interactions 1990,76 (1): 63-75.
    [143]Shimada N, Yamauchi K. Characteristics of 3,5,3'-triiodothyronine (T3)-uptake system of tadpole red blood cells:effect of endocrine-disrupting chemicals on cellular T3 response. Journal of Endocrinology 2004,183 (3):627-637.
    [144]Kitamura S, Kato T, Iida M, et al. Anti-thyroid hormonal activity of tetrabromobisphenol A, a flame retardant, and related compounds:Affinity to the mammalian thyroid hormone receptor, and effect on tadpole metamorphosis. Life Sciences 2005,76 (14):1589-1601.
    [145]Zoeller RT, Bansal R, Parris C. Bisphenol-A, an environmental contaminant that acts as a thyroid hormone receptor antagonist in vitro, increases serum thyroxine, and alters RC3/neurogranin expression in the developing rat brain. Endocrinology 2005,146 (2):607-612.
    [146]Miyazaki W, Iwasaki T, Takeshita A, et al. Polychlorinated biphenyls suppress thyroid hormone receptor-mediated transcription through a novel mechanism. Journal of Biological Chemistry 2004, 279(18):18195-18202.
    [147]Loaiza-Perez AI, Seisdedos MT, Kleiman de Pisarev DL, et al. Hexachlorobenzene, a dioxin-type compound, increases malic enzyme gene transcription through a mechanism involving the thyroid hormone response element. Endocrinology 1999,140 (9):4142-4151.
    [148]Alvarez L, Hernandez S, Martinez-de-Mena R, et al. The role of type I and type Ⅱ 5'deiodinases on hexachlorobenzene-induced alteration of the hormonal thyroid status. Toxicology 2005,207 (3): 349-362.
    [149]Raasmaja A, Viluksela M, Rozman KK. Decreased liver type I 5'-deiodinase and increased brown adipose tissue type II 5'-deiodinase activity in 2,3,7,8-tetrachlorobibenzo-p-dioxin (TCDD)-treated Long-Evans rats. Toxicology 1996,114 (3):199-205.
    [150]Zhou LX, Dehal SS, Kupfer D, et al. Cytochrome P450 catalyzed covalent binding of methoxychlor to rat hepatic, microsomal iodothyronine 5'-monodeiodinase, type I:does exposure to methoxychlor disrupt thyroid hormone metabolism? Archives of Biochemistry and Biophysics 1995,322 (2): 390-394.
    [151]Nishimura N, Miyabara Y, Sato M, et al. Immunohistochemical localization of thyroid stimulating hormone induced by a low oral dose of 2,3,7,8-tetrachlorodibenzo-p-dioxin in female Sprague-Dawley rats. Toxicology 2002,171 (2-3):73-82.
    [152]Nishimura N, Yonemoto J, Miyabara Y, et al. Rat thyroid hyperplasia induced by gestational and lactational exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Endocrinology 2003,144 (5):2075-2083.
    [153]Schuur AG, Brouwer A, Bergman A, et al. Inhibition of thyroid hormone sulfation by hydroxylated metabolites of polychlorinated biphenyls. Chemico-Biological Interactions 1998,109 (1-3):293-297.
    [154]Gutshall DM, Pilcher GD, Langley AE. Mechanism of the serum thyroid hormone lowering effect of perfluoro-n-decanoic acid (PFDA) in rats. Journal of Toxicology and Environment Health 1989,28 (1): 53-65.
    [155]Butenhoff JL, Ehresman DJ, Chang SC, et al. Gestational and lactational exposure to potassium perfluorooctanesulfonate (K+PFOS) in rats:Developmental neurotoxicity. Reproductive Toxicology 2009,27 (3-4):319-330.
    [156]Chang SC, Ehresman DJ, Bjork JA, et al. Gestational and lactational exposure to potassium perfluorooctanesulfonate (K+PFOS) in rats:toxicokinetics, thyroid hormone status, and related gene expression. Reproductive Toxicology 2009,27 (3-4):387-399.
    [157]Zoeller RT, Tan SW, Tyl RW. General background on the hypothalamic-pituitary-thyroid (HPT) axis. Critical Reviews in Toxicology 2007,37 (1-2):11-53.
    [158]Chang SC, Thibodeaux JR, Eastvold ML, et al. Thyroid hormone status and pituitary function in adult rats given oral doses of perfluorooctanesulfonate (PFOS). Toxicology 2008,243 (3):330-339.
    [159]Dallaire R, Dewailly E, Pereg D, et al. Thyroid function and plasma concentrations of polyhalogenated compounds in Inuit adults. Environmental Health Perspectives 2009,117 (9): 1380-1386.
    [160]Du Y, Shi X, Liu C, et al. Chronic effects of water-borne PFOS exposure on growth, survival and hepatotoxicity in zebrafish:a partial life-cycle test. Chemosphere 2009,74 (5):723-729.
    [161]Shi X, Du Y, Lam PK, et al. Developmental toxicity and alteration of gene expression in zebrafish embryos exposed to PFOS. Toxicology and Applied Pharmacology 2008,230 (1):23-32.
    [162]Shi X, Liu C, Wu G, et al. Waterborne exposure to PFOS causes disruption of the hypothalamus-pituitary-thyroid axis in zebrafish larvae. Chemosphere 2009,77 (7):1010-1018.
    [163]Monroy R, Morrison K, Teo K, et al. Serum levels of perfluoroalkyl compounds in human maternal and umbilical cord blood samples. Environmental Research 2008,108 (1):56-62.
    [164]Volkel W, Genzel-Boroviczeny O, Demmelmair H, et al. Perfluorooctane sulphonate (PFOS) and perfluorooctanoic acid (PFOA) in human breast milk:Results of a pilot study. International Journal of Hygiene and Environmental Health 2008,211 (3-4):440-446.
    [165]Porterfield SP, Hendrich CE. The role of thyroid hormones in prenatal and neonatal neurological development-current perspectives. Endocrine Reviews 1993,14 (1):94-106.
    [166]Galton VA. The roles of the iodothyronine deiodinases in mammalian development. Thyroid 2005, 15 (8):823-834.
    [167]Haddow JE, Palomaki GE, Allan WC, et al. Maternal thyroid deficiency during pregnancy and subsequent neuropsychological development of the child. The New England Journal of Medicine 1999, 341 (8):549-555.
    [168]Hansen KJ, Clemen LA, Ellefson ME, et al. Compound-specific, quantitative characterization of organic fluorochemicals in biological matrices. Environmental Science and Technology 2001,35 (4): 766-770.
    [169]Belknap WM, Balistreri WF, Suchy FJ, et al. Physiologic cholestasis Ⅱ:serum bile acid levels reflect the development of the enterohepatic circulation in rats. Hepatology 1981,1 (6):613-616.
    [170]Arbuckle TE. Are there sex and gender differences in acute exposure to chemicals in the same setting? Environmental Research 2006,101 (2):195-204.
    [171]Kuklenyik Z, Reich JA, Tully JS, et al. Automated solid-phase extraction and measurement of perfluorinated organic acids and amides in human serum and milk. Environmental Science and Technology 2004,38(13):3698-3704.
    [172]Fromme H, Tittlemier SA, Volkel W, et al. Perfluorinated compounds-exposure assessment for the general population in Western countries. International Journal of Hygiene and Environmental Health 2009,212 (3):239-270.
    [173]Yamada T, Kunimatsu T, Miyata K, et al. Enhanced rat Hershberger assay appears reliable for detection of not only (anti-)androgenic chemicals but also thyroid hormone modulators. Toxicological Sciences 2004,79 (1):64-74.
    [174]Crofton KM, Kodavanti PR, Derr-Yellin EC, et al. PCBs, thyroid hormones, and ototoxicity in rats: cross-fostering experiments demonstrate the impact of postnatal lactation exposure. Toxicological Sciences 2000,57 (1):131-140.
    [175]Morreale de Escobar G, Obregon MJ, Ruiz de Ona C, et al. Transfer of thyroxine from the mother to the rat fetus near term:effects on brain 3,5,3'-triiodothyronine deficiency. Endocrinology 1988,122 (4):1521-1531.
    [176]Zoeller RT, Crofton KM. Thyroid hormone action in fetal brain development and potential for disruption by environmental chemicals. Neurotoxicology 2000,21 (6):935-945.
    [177]Burrow GN, Fisher DA, Larsen PR. Maternal and fetal thyroid function. The New England Journal of Medicine 1994,331 (16):1072-1078.
    [178]Wade MG, Parent S, Finnson KW, et al. Thyroid toxicity due to subchronic exposure to a complex mixture of 16 organochlorines, lead, and cadmium. Toxicological Sciences 2002,67 (2):207-218.
    [179]Livak KJ, Schmittgen TD. Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2-△△CT Method. Methods 2001,25 (4):402-408.
    [180]王宇,张素才,程薇波等.丙硫氧嘧啶致甲状腺组织增生的时效关系研究.卫生研究2009,38(1):15-18.
    [181]房辉,陈贯一,王素莉等.碘缺乏与碘过多大鼠甲状腺定量形态学研究.中国地方病杂志2000,19(3):161-163.
    [182]叶振坤,孙毅娜,林来详等.高剂量碘酸钾射入后大鼠甲状腺定量形态学研究.中国体式学与图像分析2009,14(1):16-20.
    [183]宝生物有限公司.Real Time PCR实验及解析方法.
    [184]Hagenaars A, Knapen D, Meyer IJ, et al. Toxicity evaluation of perfluorooctane sulfonate (PFOS) in the liver of common carp (Cyprinus carpio). Aquatic Toxicology 2008,88 (3):155-163.
    [185]Weiss JM, Andersson PL, Lamoree MH, et al. Competitive binding of poly-and perfluorinated compounds to the thyroid hormone transport protein transthyretin. Toxicological Sciences 2009,109 (2):206-216.
    [186]Power DM, Elias NP, Richardson SJ, et al. Evolution of the thyroid hormone-binding protein, transthyretin. General and Comparative Endocrinology 2000,119 (3):241-255.
    [187]Sousa JC, de Escobar GM, Oliveira P, et al. Transthyretin is not necessary for thyroid hormone metabolism in conditions of increased hormone demand. Journal of Endocrinology 2005,187 (2): 257-266.
    [188]McClain RM. Mechanistic considerations for the relevance of animal data on thyroid neoplasia to human risk assessment. Mutation Research 1995,333 (1-2):131-142.
    [189]Cavalieri RR. Iodine metabolism and thyroid physiology:current concepts. Thyroid 1997,7 (2): 177-181.
    [190]Visser TJ. Pathways of thyroid hormone metabolism. Acta Med Austriaca 1996,23 (1-2):10-16.
    [191]Hess SY, Zimmermann MB, Arnold M, et al. Iron deficiency anemia reduces thyroid peroxidase activity in rats. Journal of Nutrition 2002,132 (7):1951-1955.
    [192]Zhou T, Ross DG, DeVito MJ, et al. Effects of short-term in vivo exposure to polybrominated diphenyl ethers on thyroid hormones and hepatic enzyme activities in weanling rats. Toxicological Sciences 2001,61 (1):76-82.
    [193]Bock KW, Gschaidmeier H, Heel H, et al. Functions and transcriptional regulation of PAH-inducible human UDP-glucuronosyltransferases. Drug Metabolism Reviews 1999,31 (2):411-422.
    [194]Shipley JM, Hurst CH, Tanaka SS, et al. trans-activation of PPARa and induction of PPARα target genes by perfluorooctane-based chemicals. Toxicological Sciences 2004,80 (1):151-160.
    [195]Takacs ML, Abbott BD. Activation of mouse and human peroxisome proliferator-activated receptors (α,β/δ, γ) by perfluorooctanoic acid and perfluorooctane sulfonate. Toxicological Sciences 2007,95 (1):108-117.
    [196]Cai Y, Nelson BD, Li R, et al. Thyromimetic action of the peroxisome proliferators clofibrate, perfluorooctanoic acid, and acetylsalicylic acid includes changes in mRNA levels for certain genes involved in mitochondrial biogenesis. Archives of Biochemistry and Biophysics 1996,325 (1): 107-112.
    [197]Gonzalez FJ, Peters JM, Cattley RC. Mechanism of action of the nongenotoxic peroxisome proliferators:role of the peroxisome proliferator-activator receptor alpha. Journal of National Cancer Institute 1998,90 (22):1702-1709.
    [198]Corton JC, Anderson SP, Stauber A. Central role of peroxisome proliferator-activated receptors in the actions of peroxisome proliferators. Annual Review of Pharmacology and Toxicology 2000,40: 491-518.
    [199]Peters JM, Cattley RC, Gonzalez FJ. Role of PPAR alpha in the mechanism of action of the nongenotoxic carcinogen and peroxisome proliferator Wy-14,643. Carcinogenesis 1997,18 (11): 2029-2033.
    [200]Palmer CN, Hsu MH, Griffin KJ, et al. Peroxisome proliferator activated receptor-alpha expression in human liver. Molecular Pharmacology 1998,53 (1):14-22.
    [201]Zhou J, Zhang J, Xie W. Xenobiotic nuclear receptor-mediated regulation of UDP-glucuronosyl-transferases. Current Drug Metabolism 2005,6 (4):289-298.
    [202]Osabe M, Sugatani J, Fukuyama T, et al. Expression of hepatic UDP-glucuronosyltransferase 1A1 and 1A6 correlated with increased expression of the nuclear constitutive androstane receptor and peroxisome proliferator-activated receptor alpha in male rats fed a high-fat and high-sucrose diet. Drug Metabolism and Disposition 2008,36 (2):294-302.
    [203]Chanoine JP, Braverman LE, Farwell AP, et al. The thyroid gland is a major source of circulating T3 in the rat. Journal of Clinical Investigation 1993,91:2709-2713.
    [204]Escobar-Morreale HF, Obregon MJ, Hernandez A, et al. Regulation of iodothyronine deiodinase activity as studied in thyroidectomized rats infused with thyroxine or triiodothyronine. Endocrinology 1997,138 (6):2559-2568.
    [205]Klammer H, Schlecht C, Wuttke W, et al. Effects of a 5-day treatment with the UV-filter octyl-methoxycinnamate (OMC) on the function of the hypothalamo-pituitary-thyroid function in rats. Toxicology 2007,238 (2-3):192-199.
    [206]Pazos-Moura CC, Moura EG, Dorris ML, et al. Effect of iodine deficiency and cold exposure on thyroxine 5'-deiodinase activity in various rat tissues. American Journal of Physiology 1991,260 (2): 175-182.
    [207]Abend SL, Fang SL, Alex S, et al. Rapid alteration in circulating free thyroxine modulates pituitary type Ⅱ 5'deiodinase and basal thyrotropin secretion in the rat. Journal of Clinical Investigation 1991, 88 (3):898-903.
    [208]Hood A, Liu J, Klaassen CD. Effects of phenobarbital, pregnenolone-16alpha-carbonitrile, and propylthiouracil on thyroid follicular cell proliferation. Toxicological Sciences 1999,50 (1):45-53.
    [209]Schuur AG, Boekhorst FM, Brouwer A, et al. Extrathyroidal effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on thyroid hormone turnover in male Sprague-Dawley rats. Endocrinology 1997,138 (9): 3727-3734.
    [210]Vansell NR, Klaassen CD. Increased biliary excretion of thyroxine by microsomal enzyme inducers. Toxicology and Applied Pharmacology 2001,176 (3):187-194.
    [211]Schmutzler C, Gotthardt I, Hofmann PJ, et al. Endocrine disruptors and the thyroid gland-a combined in vitro and in vivo analysis of potential new biomarkers. Environmental Health Perspectives 2007,115 Suppl 1:77-83.
    [212]Kato Y, Suzuki H, Ikushiro S, et al. Decrease in serum thyroxine level by phenobarbital in rats is not necessarily dependent on increase in hepatic UDP-glucuronosyltransferase. Drug Metabolism and Disposition 2005,33 (11):1608-1612.
    [213]Weiss JM, Andersson PL, Lamoree MH, et al. Competitive Binding of Poly-and Perfluorinated Compounds to the Thyroid Hormone Transport Protein Transthyretin. Toxicological Sciences 2009, 109 (2):206-212.
    [214]Friesema EC, Jansen J, Milici C, et al. Thyroid hormone transporters. Vitamins and Hormones 2005, 70:137-167.
    [215]Hagenbuch B. Cellular entry of thyroid hormones by organic anion transporting polypeptides. Best Practice and Research Clinical Endocrinology and Metabolism 2007,21 (2):209-221.
    [216]Visser WE, Jansen J, Friesema EC, et al. Novel pathogenic mechanism suggested by ex vivo analysis of MCT8 (SLC16A2) mutations. Human Mutation 2009,30 (1):29-38.
    [217]Martin MT, Brennan RJ, Hu W, et al. Toxicogenomic study of triazole fungicides and perfluoroalkyl acids in rat livers predicts toxicity and categorizes chemicals based on mechanisms of toxicity. Toxicological Sciences 2007,97 (2):595-613.
    [218]Abe T, Kakyo M, Sakagami H, et al. Molecular characterization and tissue distribution of a new organic anion transporter subtype (oatp3) that transports thyroid hormones and taurocholate and comparison with oatp2. Journal of Biological Chemistry 1998,273 (35):22395-22401.
    [219]Cattori V, Hagenbuch B, Hagenbuch N, et al. Identification of organic anion transporting polypeptide 4 (Oatp4) as a major full-length isoform of the liver-specific transporter-1 (rlst-1) in rat liver. FEBS Letters 2000,474 (2-3):242-245.
    [220]Friesema EC, Docter R, Moerings EP, et al. Identification of thyroid hormone transporters. Biochemical and Biophysical Research Communications 1999,254 (2):497-501.
    [221]Ribeiro RC, Cavalieri RR, Lomri N, et al. Thyroid hormone export regulates cellular hormone content and response. Journal of Biological Chemistry 1996,271 (29):17147-17151.
    [222]Kipp H, Arias IM. Trafficking of canalicular ABC transporters in hepatocytes. Annual Review of Physiology 2002,64:595-608.
    [223]Czarnywojtek A, Krysinska I, Lacka K, et al. A study of thyroglobulin concentration in the thyroid and serum of patients with different thyroid disorders. Archivum Immunologiae et Therapiae Experimentalis (Warsz).2002,50 (2):143-148.
    [224]Yu WG, Liu W, Jin YH, et al. Prenatal and Postnatal Impact of Perfluorooctane Sulfonate (PFOS) on Rat Development:A Cross-Foster Study on Chemical Burden and Thyroid Hormone System. Environmental Science and Technology 2009,43 (21):8416-8422.
    [225]Takayama S, Aihara K, Onodera T, et al. Antithyroid effects of propylthiouracil and sulfamonomethoxine in rats and monkeys. Toxicology and Applied Pharmacology 1986,82 (2): 191-199.

© 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