用户名: 密码: 验证码:
碘对甲状腺滤泡功能的调控作用及分子机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
碘对甲状腺功能的影响表现为循环碘及甲状腺内部有机碘储备状况改变了甲状腺自身的碘代谢行为及其对垂体TSH的敏感性。这种作用不依赖于垂体TSH,其目的是维持合适碘池、稳定甲状腺功能,属于甲状腺功能的自我调控作用。
     甲状腺激素的合成与分泌是一个连续而复杂的生理过程,主要涉及钠共转运体(NIS)、甲状腺髓过氧化物酶(TPO)、甲状腺球蛋白(TG)、及促甲状腺素受体(TSHr)等甲状腺特异蛋白及这些蛋白表达的主要转录调控因子TTF-1、TTF-2、PAX8的表达及活性。甲状腺内外碘的变化,通过各种复杂的机制,最终导致甲状腺转录因子及甲状腺特异蛋白的变化,实现甲状腺功能的自我调控。
     甲状腺功能调控的研究,需选择合适的实验模型。动物水平的研究由于影响因素众多,不易阐明某种因素的具体作用及机制;体外条件下,多数甲状腺细胞,呈单层细胞生长,无法形成滤泡结构,缺乏完整的激素合成、储备及分泌功能,均不是甲状腺功能调控研究的最佳选择。体外重构甲状腺滤泡,克服了上述二种模型的不足,既脱离了体内复杂的调控环境,又兼备完整的功能,是甲状腺功能调控研究理想实验模型。
     我们探索体外重构猪甲状腺滤泡的诱导方法,从甲状腺滤泡的形态、功能及甲状腺特异蛋白表达三方面证实体外重构猪甲状腺滤泡模型构建成功。在此基础上,利用Wenstern-blolt、RealTimeRT-PCR、ELISA、RIA等方法及免疫荧光、激光共聚焦显微镜观察、创新的甲状腺滤泡腔TG提取等技术,对高碘(HI)及低碘(LI)下甲状腺特异蛋白,甲状腺转录因子、甲状腺激素分泌、滤泡腔TG的碘化度进行深入研究,探讨碘对甲状腺功能的调控作用及分子机制,现将结果总结如下:
     一.猪体外重构甲状腺滤泡的构建及鉴定:
     1.猪体外重构甲状腺滤泡的构建方法:
     依照下列要点:①猪甲状腺低胰蛋白酶(0.1%)消化。②震荡法分散细胞。③1.5-3X106/L高密度、均匀接种。④接种头3天时在培养基中加入1mIU/mlTSH及15%的胎牛血清。⑤滤泡形成后改胎牛血清为10%小牛血清。经过3天的培养,即可形成了大量球状或半球状的重构甲状腺滤泡。
     2.体外重构甲状腺滤泡的形态学鉴定:
     用普通倒置显微镜可以观察到形成的圆圈样结构及细胞团,直径约10-100um;相差倒置显微镜下,圆圈样结构为半球状滤泡的贴壁面,穹窿顶由细胞构成;免疫荧光染色,利用激光共聚焦显微镜的“光学切片”作用下可以清晰地观察到细胞团是由周边的甲状腺细胞及内部的滤泡腔构成。
     3.体外重构甲状腺滤泡的功能学鉴定:
     ①重构滤泡组上清液中有FT3、FT4的分泌,滤泡腔有TG的储备,而单层细胞组几乎未能检测到FT3、FT4分泌及TG储备。
     ②重构滤泡组:高碘干预甲状腺特异蛋白NIS、TG、TPO、TSHr及甲状腺转录因子TTF-1、PAX8mRNA的表达高于低碘干预组。单层细胞组:高碘干预组上述指标表达均低于低碘干预组。
     二.以体外重构猪甲状腺滤泡为实验模型,研究碘对甲状腺功能的调控作用:
     1.高碘干预组甲状腺激素合成相关基因NIS、TPO、TGmRNA表达显著高于低碘组。
     2.高碘干预组培养上清液中FT3、FT4分泌显著多于低碘组。
     三.碘对甲状腺功能调控作用的分子机制研究:
     1.高碘干预组TSHr、TTF-1、PAX8mRNA表达及蛋白合成显著高于低碘组。
     2.①高碘干预组甲状腺滤泡腔TG碘化度高于低碘组;
     ②高碘+ MMI(甲硫咪唑)组较单纯高碘组滤泡腔TG的碘化度显著下降;
     ③高碘组TSHr、TTF-1、PAX8mRNA表达及蛋白合成较低碘组及高碘+MMI显著上调。
     ④高碘+MMI组较MMI+高碘组TSHr、TTF-1、PAX8mRNA表达有升高的趋势。
     (高碘+MMI组:先高碘干预36h,再加入MMI;MMI+高碘组:先MMI干预36h,再加入高碘.即高碘、MMI干预顺序不同。)
     3.①PKA阻断组:TTF-1、TSHrmRNA表达及蛋白合成较对照组显著上调。
     ②IP3阻断组:TTF-1、TSHrmRNA表达及蛋白合成较对照组显著上调。
     ③PKA阻断组及IP3阻断组:NIS的表达较对照组上调。
     ④PKA阻断组:NIS2的表达较对照组上调;IP3阻断组:NIS2的表达较对照组下调。
     结论:
     1.保持细胞膜的完整性、高密度均匀接种、增加细胞的接触、粘附、聚集的机会和能力,是体外重构甲状腺滤泡形成的关键;避免使用胎牛血清是防止甲状腺滤泡结构蜕变为单层结构,维持甲状腺滤泡形态的重要因素。
     2.体外重构甲状腺滤泡脱离了体内复杂环境,又具备完整的甲状腺功能,是甲状腺功能调控作用及机制研究的理想实验模型,可用于观察甲状腺滤泡形成过程,探讨甲状腺滤泡结构、滤泡内容物对甲状腺功能的调控作用。
     3.高碘组甲状腺激素合成相关基因NIS、TG、TPO高表达,培养上清液FT3、FT4高分泌,甲状腺激素的合成及分泌功能处于活跃状态,在滤泡层面上,碘对甲状腺激素合成及分泌功能具有促进作用。
     4.碘离子、MMI影响滤泡腔TG的碘化,其本身对TSHr及TTF-1的表达无直接作用;TSH/TSHr信号的CAMP/AKP及磷脂酶C/IP3途径对TTF-1、TSHr的表达亦无上调作用,TTF-1、TSHr高表达是滤泡腔TG高碘化的直接结果。
     5.碘离子提高了滤泡腔TG的碘化度,通过调控TTF-1,进而上调TSHr的表达,增强了甲状腺滤泡对中枢TSH/TSHr信号的敏感性,是其功能自我调控的重要途径之一。
     6. TG高碘化时,TSHr表达上调,滤泡对中枢信号敏感,促进了NIS、TPO、TG的表达及激素的分泌。是其功能活跃的原因之一。
     7.TSH/TSHr信号的不同传导途径对NIS及NIS2具有不同的调控作用,提示在NISmRNA剪接水平上中枢信号参与NIS不同亚型表达的调控。
     综上,我们发现:在重构滤泡这一层面上,碘影响滤泡腔TG的碘化度,通过调控TTF-1,进而上调TSHr的表达,改变了滤泡对中枢TSH/TSHr信号的敏感性。TG的碘化程度不同,滤泡对中枢TSH/TSHr信号敏感性存在差异,是甲状腺适应内外碘环境的变化,实现功能自我调控的重要途径之一;TG高碘化时,TSHr表达上调,滤泡对中枢信号敏感,促进了NIS、TPO、TG的表达及甲状腺激素的分泌。在滤泡水平上,碘通过上述机制有效促进甲状腺激素的合成与分泌。
     近10余来,高碘相关的甲状腺功能异常、自身免疫性甲状腺炎及甲状腺肿瘤发病率呈升高趋势。碘如何调控甲状腺功能,目前知之甚少,我们的工作为该领域的研究提供了科学实用的实验模型、拓宽了研究思路,奠定了一定的方法学基础,对探讨甲状腺疾病的发病机制及科学制定补碘策略具有重要理论和现实意义。
The reserve of organic thyroid iodine and its circulation change the metabolism of thyroid and its sensitivity to pituitary TSH, which reflects the effects of iodine on thyroid function and is independent of pituitary TSH. This mechanism is a part of the self-regulation of thyroid function, which is conducive to keeping suitable iodine content and the stability of thyroid function.
     The synthesis and secretion of thyroid hormone are durative and complicated physiological process which relate to the expressions of thyroid-specific proteins including the sodium/iodide cotransporter (NIS), thyroid peroxidase (TPO), thyroglobulin (Tg) and thyroid stimulating hormone receptor (TSHr) as well as their transcriptional regulators, such as TTF1, TTF-2 and PAX8. The changes of internal and external iodine concentration bring about the alteration of the expressions of thyroid transcriptional factors and thyroid-specific proteins through various complicated mechanisms, which finally results in the self-regulation of thyroid function.
     The proper experimental model is a key point for the research on the regulation of thyroid function. Research in animals is affected by many factors and is unable to elucidate the detailed functions and mechanisms of some element; most thyroid cells cultured in vitro grow as a monolayer and cannot form the follicle structure, they do not possess the ability of hormonal synthesis, reserve and secretion, which are unsuitable for research on thyroid functional regulation. The recombinant thyroid follicle (RTF) is an ideal experimental model for thyroid functional regulation because it both breaks from the complex regulatory conditions in vivo and possesses the integral functions.
     We probed the methods of inducing swine recombinant thyroid follicle in vitro, and the RTF model was confirmed to be successful through identifying the morphology and function of thyroid follicle and the expression of thyroid-specific proteins. A series of methods including Western-blotting, realtime RT-PCR, ELISA, RIA, immunofluorescence, laser scanning confocal microscope and the techniques for extraction of thyroid follicular TG were employed to investigate the secretion of thyroid hormone, iodinated extent of follicular TG and the expressions of thyroid-specific proteins and thyroid transcriptional factors under the stress of high or low concentration of iodine. We aimed to elucidate the molecular mechanisms of iodine-regulated thyroid function. The results are as follows:
     (一)Establishment and identification of swine recombinant thyroid follicle
     ①The methods of establishing swine recombinant thyroid follicle in vitro are briefly introduced as follows: thyroid tissues of swine were digested with 0.1% typsin and vibrated to disperse cells. Cells were maintained in RPMI-1640 medium supplemented with 15% fetal calf serum containing 1 mIU/mlTSH at the density of 1.5-3x106/L and the next day 10% calf serum replaced the fetal calf serum. After cultured 3 days thyroid cells accumulated together to form recombinant thyroid follicle.
     ②Morphological identification of recombinant thyroid follicle Thyroid cells accumulated together to form a circular structure with diameter of 10 to 100 um under the inverted microscope. Observation of phase-contrast microscope revealed the circular structure was the surface of hemisphere follicle closing to the coverslip and the fornix was established with lots of cells. Laser confocal scanning microscope observation revealed that the cell aggregate consisted of thyroid cells and the inner follicle, which could be clearly visualized after immunofluorescence staining.
     ③Functional identification of recombinant thyroid follicle
     In the supernate of RTF group FT3 and FT4 were secreted and TG was reserved in the follicle, while all of them were not detected in the supernate of MC group.
     ④Under the stress of high concentration of iodine the expressions of thyroid-specific proteins, NIS, TG, TPO, and TSHr, and the mRNA levels of thyroid transcriptional factors, TTF-1 and PAX8 were all up-regulated in RTF group, whose levels were all down-regulated in MC group.
     (二)Study the role of iodine on the regulation of thyroid function based on the in vitro experimental model of swine recombinant thyroid follicle
     ①The mRNA levels of NIS, TPO and TG, which were related to the synthesis of thyroid hormone, were all up-regulated in HI group, while were down-regulated in LI group.
     ②The secretion of FT3 and FT4 were enhanced in the supernate of HI group medium, while were significantly decreased in LI group supernate.
     (三)The molecular mechanism of iodine on the regulation of thyroid function
     ①In group HI, the mRNA levels of TSHr, TTF-1 and PAX8 were all up-regulated as well as their protein synthesis.
     ②The iodinated extent of follicular TG in group HI was relative higher than that in other groups, and it was decreased in group“HI+MMI”; when compared with“MMI+HI”group the iodinated extent of TG in group“HI+MMI”significantly rose.
     (“MMI+HI”group and“HI+MMI”group were dealt with a different procedure; cells in“HI+MMI”group were first treated with high concentration of iodine for 36 h and then MMI while cells in group“MMI+HI”were treated with a reverse sequence.)
     When follicular TG was highly iodinated, the mRNA levels of TSHr, TTF-1 and PAX8 were markedly improved as well as their protein synthesis.
     ④The mRNA levels of TTF-1 and TSHr in group of H89 (inhibitor of PKA) and LY294002 (inhibitor of IP3) were both significantly enhanced as well as their protein synthesis;
     ⑤The expression of NIS in group of H89 (inhibitor of PKA) was down-regulated while NIS2 (the subtype of NIS) was up-regulated; while in group of LY294002 the expression of NIS and NIS2 were up-regulated and down-regulated, respectively.
     Conclusions:
     1. Keeping the integrity of cell membrane, seeding at high density and increasing the opportunities of cell aggregation, adhesion and contact were the key points of successfully establishing recombinant thyroid follicle in vitro; one very important element was avoid using fetal bovine serum which could drive cells forming thyroid follicle to be monolayer growth.
     2. Recombinant thyroid follicle was an ideal experimental model for the research on the mechanism of regulation of thyroid function, because it both broke away from the complex environment in vivo and had integrated thyroid function. Therefore, the recombinant thyroid follicle could be used for observing the process of the production of thyroid follicle and exploring the regulatory roles of follicular structure and substances on thyroid function.
     3. The expressions of NIS, TG and TPO, which were all related to the synthesis of thyroid hormone, were overexpressed; the secretions of FT3 and FT4 in the supernate of culture medium were enhanced, indicating the recombinant thyroid follicle was undergoing vibrant hormonal synthesis and secretion. All evidences above proved that iodine promoted the synthesis and secretion of thyroid hormone at follicular level.
     4. Iodine and MMI affected the iodination of follicular TG and they had no direct effects on the expressions of TSHr and TTF-1; the CAMP/AKP and C/IP3 pathways, which belonged to TSH/TSHr signal, did not up-regulate the expressions of TTF-1 and TSHr, indicating highly iodinated TG directly resulted in the overexpressions of TTF-1 and TSHr.
     5. Iodine improved the iodinated extent of follicular TG and increased the expression level of TSHr through regulating TTF-1, which finally resulted in promoting the sensitivity of thyroid follicle to central TSH/TSHr signal. This is one of the important pathways for thyroid functional self-regulation.
     6. When TG was highly iodinated, the expressions of TSHr, NIS, TPO and TG was enhanced as well as the hormone secretion, and follicle was sensitive to central signal, which was one of the reasons for the vibrant thyroid functions.
     7. Different signal transduction pathways of TSH/TSHr signal played different regulatory roles on the expressions of NIS and NIS2, indicating central signals regulated the expression of different subtype of NIS at the level of mRNA splicing.
     In summary, we found iodine affected the iodinated extent of follicular TG from the aspect of follicle, and promoted the expression of TSHr through regulating TTF-1, which finally changed the sensitivity of follicle to central TSH/TSHr signal. TG, which was iodinated on different extent, led to the different sensitivity of follicle to central TSH/TSHr signal, this was one of the important pathways for thyroid adapting itself to different concentration of iodine and achieving functional self-regulation. The highly iodinated TG enhanced the expression of TSHr, drove follicle to be sensitive to central signal, and promoted the secretion of thyroid hormone and the expression of NIS, TPO and TG. Taken follicle as a research model, iodine efficiently expedited the synthesis and secretion of thyroid hormone through the mechanisms mentioned above.
     Over the past 10 years, the incidences of thyroid dysfunction, autoimmune thyroiditis and thyroid cancer that were all high iodine-related diseases obviously increased. At present little is known about how iodine regulates thyroid functions. Our work offered methodology basis and directions for the research on this area and had important theoretical and practical significance for exploring the pathogenesis of iodine-related thyroid diseases and making the scientific strategies for iodine supplement.
引文
1 Perez Perez F. [Comparative study of iodine and thyroid gland in biological processes: development and reproduction]. An R Acad Nac Med (Madr), 2000,117:395-425.
    2 Braverman LE, Roti E. Effects of iodine on thyroid function. Acta Med Austriaca, 1996,23:4-9.
    3 Vassart G, Dumont JE. The thyrotropin receptor and the regulation of thyrocyte function and growth. Endocr Rev, 1992,13:596-611.
    4 Morton ME, Chaikoff IL, Rosenfeld S. Inhibiting effect of inorganic iodide on the formation in vitro of thyroxine and diiodotyrosine by surviving thyroid tissue. J Biol Chem, 1944:154:381-387.
    5 Espinoza CR, Schmitt TL, Loos U. Thyroid transcription factor 1 and Pax8 synergistically activate the promoter of the human thyroglobulin gene. J Mol Endocrinol, 2001,27:59-67.
    6 WOLFF J, CHAIKOFF IL. Plasma inorganic iodide as a homeostatic regulator of thyroid function. J Biol Chem, 1948,174:555-64.
    7 WOLFF J, CHAIKOFF IL, et al. The temporary nature of the inhibitory action of excess iodine on organic iodine synthesis in the normal thyroid. Endocrinology, 1949,45:504-13, illust.
    8 BRAVERMAN LE, INGBAR SH. CHANGES IN THYROIDAL FUNCTION DURING ADAPTATION TO LARGE DOSES OF IODIDE. J Clin Invest, 1963,42:1216-31.
    9 Christophe D, Cabrer B, Bacolla A, et al. An unusually long poly(purine)-poly(pyrimidine) sequence is located upstream from the human thyroglobulin gene. Nucleic Acids Res, 1985,13:5127-44.
    10 Ikuyama S, Ohe K, Takayanagi R, et al. Cloning and characterization of the4.2 kb region of the rat thyrotropin receptor promoter. Endocr J, 1997,44:247-56.
    11 Damante G, Di Lauro R. Thyroid-specific gene expression. Biochim Biophys Acta, 1994,1218:255-66.
    12 Dai G, Levy O, Carrasco N. Cloning and characterization of the thyroid iodide transporter. Nature, 1996,379:458-60.
    13 Eng PH, Cardona GR, Fang SL, et al. Escape from the acute Wolff-Chaikoff effect is associated with a decrease in thyroid sodium/iodide symporter messenger ribonucleic acid and protein. Endocrinology, 1999,140:3404-10.
    14 Uyttersprot N, Pelgrims N, Carrasco N, et al. Moderate doses of iodide in vivo inhibit cell proliferation and the expression of thyroperoxidase and Na+/I- symporter mRNAs in dog thyroid. Mol Cell Endocrinol, 1997,131:195-203.
    15 Cardoso LC, Martins DC, Campos DV, et al. Effect of iodine or iopanoic acid on thyroid Ca2+/NADPH-dependent H2O2-generating activity and thyroperoxidase in toxic diffuse goiters. Eur J Endocrinol, 2002,147:293-8.
    16 Pregliasco L, Bocanera L, Krawiec L, et al. Effects of iodide on thyroglobulin biosynthesis in FRTL-5 cells. Thyroid, 1996,6:319-23.
    17 Ferreira AC, Lima LP, Araujo RL, et al. Rapid regulation of thyroid sodium-iodide symporter activity by thyrotrophin and iodine. J Endocrinol, 2005,184:69-76.
    18 Grollman EF, Smolar A, Ommaya A, et al. Iodine suppression of iodide uptake in FRTL-5 thyroid cells. Endocrinology, 1986,118:2477-82.
    19 Suzuki K, Kohn LD. Differential regulation of apical and basal iodide transporters in the thyroid by thyroglobulin. J Endocrinol, 2006,189:247-55.
    20 Noguchi Y, Harii N, Giuliani C, et al. Thyroglobulin (Tg) induces thyroid cell growth in a concentration-specific manner by a mechanism other than thyrotropin/cAMP stimulation. Biochem Biophys Res Commun, 2010,391:890-4.
    21 Deshpande V, Venkatesh SG. Thyroglobulin, the prothyroid hormone: chemistry, synthesis and degradation. Biochim Biophys Acta, 1999,1430:157-78.
    22 Kimura T, Van Keymeulen A, Golstein J, et al. Regulation of thyroid cell proliferation by TSH and other factors: a critical evaluation of in vitro models. Endocr Rev, 2001,22:631-56.
    23 Bernier-Valentin F, Trouttet-Masson S, Rabilloud R, et al. Three-dimensional organization of thyroid cells into follicle structures is a pivotal factor in the control of sodium/iodide symporter expression. Endocrinology, 2006,147:2035-42.
    24 Suzuki K, Mori A, Lavaroni S, et al. In vivo expression of thyroid transcription factor-1 RNA and its relation to thyroid function and follicular heterogeneity: identification of follicular thyroglobulin as a feedback suppressor of thyroid transcription factor-1 RNA levels and thyroglobulin synthesis. Thyroid, 1999,9:319-31.
    25 Suzuki K, Mori A, Saito J, et al. Follicular thyroglobulin suppresses iodide uptake by suppressing expression of the sodium/iodide symporter gene. Endocrinology, 1999,140:5422-30.
    26 Suzuki K, Mori A, Lavaroni S, et al. Thyroglobulin regulates follicular function and heterogeneity by suppressing thyroid-specific gene expression. Biochimie, 1999,81:329-40.
    27 Altmann A, Schulz RB, Glensch G, et al. Effects of Pax8 and TTF-1 thyroid transcription factor gene transfer in hepatoma cells: imaging of functional protein-protein interaction and iodide uptake. J Nucl Med, 2005,46:831-9.
    28 Ulianich L, Suzuki K, Mori A, et al. Follicular thyroglobulin (TG) suppression of thyroid-restricted genes involves the apical membrane asialoglycoprotein receptor and TG phosphorylation. J Biol Chem, 1999,274:25099-107.
    29 Faggiano A, Coulot J, Bellon N, et al. Age-dependent variation of follicularsize and expression of iodine transporters in human thyroid tissue. J Nucl Med, 2004,45:232-7.
    30 Ambesi-Impiombato FS, Parks LA, Coon HG. Culture of hormone-dependent functional epithelial cells from rat thyroids. Proc Natl Acad Sci U S A, 1980,77:3455-9.
    31 Takasu N, Ohno S, Komiya I, et al. Requirements of follicle structure for thyroid hormone synthesis; cytoskeletons and iodine metabolism in polarized monolayer cells on collagen gel and in double layered, follicle-forming cells. Endocrinology, 1992,131:1143-8.
    32 Mauchamp J, Mirrione A, Alquier C, et al. Follicle-like structure and polarized monolayer: role of the extracellular matrix on thyroid cell organization in primary culture. Biol Cell, 1998,90:369-80.
    33 Dunn JT. Correcting iodine deficiency is more than just spreading around a lot of iodine. Thyroid, 2001,11:363-4.
    34 Delange F, Burgi H, Chen ZP, et al. World status of monitoring iodine deficiency disorders control programs. Thyroid, 2002,12:915-24.
    35 Dunn JT. Guarding our nation's thyroid health. J Clin Endocrinol Metab, 2002,87:486-8.
    36 Teng W, Shan Z, Teng X, et al. Effect of iodine intake on thyroid diseases in China. N Engl J Med, 2006,354:2783-93.
    37 Laurberg P, Bulow Pedersen I, Knudsen N, et al. Environmental iodine intake affects the type of nonmalignant thyroid disease. Thyroid, 2001,11:457-69.
    38 Yoshida A, Taniguchi S, Hisatome I, et al. Pendrin is an iodide-specific apical porter responsible for iodide efflux from thyroid cells. J Clin Endocrinol Metab, 2002,87:3356-61.
    39 Nilsson M, Bjorkman U, Ekholm R, et al. Iodide transport in primary cultured thyroid follicle cells: evidence of a TSH-regulated channel mediating iodide efflux selectively across the apical domain of the plasmamembrane. Eur J Cell Biol, 1990,52:270-81.
    40 Toda S, Sugihara H. Reconstruction of thyroid follicles from isolated porcine follicle cells in three-dimensional collagen gel culture. Endocrinology, 1990,126:2027-34.
    41 Fayet G, Hovsepian S. Reconstruction of the thyroid follicle with long-term cultured cells. Thyroidology, 1991,3:123-5.
    42 Kogai T, Curcio F, Hyman S, et al. Induction of follicle formation in long-term cultured normal human thyroid cells treated with thyrotropin stimulates iodide uptake but not sodium/iodide symporter messenger RNA and protein expression. J Endocrinol, 2000,167:125-35.
    43 Hoshi N, Kusakabe T, Taylor BJ, et al. Side population cells in the mouse thyroid exhibit stem/progenitor cell-like characteristics. Endocrinology, 2007,148:4251-8.
    44 Goto K, Hosaka S, Ishii H, et al. Follicle-like hemicysts reorganized in monolayer culture of rat thyroid cells in the medium supplemented with isologous serum. Tohoku J Exp Med, 1991,164:223-30.
    45 Tognella C, Marti U, Peter HJ, et al. Follicle-forming cat thyroid cell lines synthesizing extracellular matrix and basal membrane components: a new tool for the study of thyroidal morphogenesis. J Endocrinol, 1999,163:505-14.
    46 Nakazato M, Chung HK, Ulianich L, et al. Thyroglobulin repression of thyroid transcription factor 1 (TTF-1) gene expression is mediated by decreased DNA binding of nuclear factor I proteins which control constitutive TTF-1 expression. Mol Cell Biol, 2000,20:8499-512.
    47 Kohn LD, Suzuki K, Nakazato M, et al. Effects of thyroglobulin and pendrin on iodide flux through the thyrocyte. Trends Endocrinol Metab, 2001,12:10-6.
    48 Doganay L, Puyan FO, Oz F, et al. Regenerative hyperplasia of follicular epithelium in chronic lymphocytic thyroiditis. Appl Immunohistochem MolMorphol, 2005,13:353-7.
    49 Dugrillon A. Iodolactones and iodoaldehydes--mediators of iodine in thyroid autoregulation. Exp Clin Endocrinol Diabetes, 1996,104 Suppl 4:41-5.
    50 Filetti S, Rapoport B. Evidence that organic iodine attenuates the adenosine 3',5'-monophosphate response to thyrotropin stimulation in thyroid tissue by an action at or near the adenylate cyclase catalytic unit. Endocrinology, 1983,113:1608-15.
    51 Suzuki K, Lavaroni S, Mori A, et al. Autoregulation of thyroid-specific gene transcription by thyroglobulin. Proc Natl Acad Sci U S A, 1998,95:8251-6.
    52 Eng PH, Cardona GR, Previti MC, et al. Regulation of the sodium iodide symporter by iodide in FRTL-5 cells. Eur J Endocrinol, 2001,144:139-44.
    53 Mahmoud I, Colin I, Many MC, et al. Direct toxic effect of iodide in excess on iodine-deficient thyroid glands: epithelial necrosis and inflammation associated with lipofuscin accumulation. Exp Mol Pathol, 1986,44:259-71.
    54 Becks GP, Eggo MC, Burrow GN. Regulation of differentiated thyroid function by iodide: preferential inhibitory effect of excess iodide on thyroid hormone secretion in sheep thyroid cell cultures. Endocrinology, 1987,120:2569-75.
    55 Spitzweg C, Joba W, Morris JC, et al. Regulation of sodium iodide symporter gene expression in FRTL-5 rat thyroid cells. Thyroid, 1999,9:821-30.
    56 Scipioni A, Ferretti E, Soda G, et al. hNIS protein in thyroid: the iodine supply influences its expression and localization. Thyroid, 2007,17:613-8.
    57 Leoni SG, Galante PA, Ricarte-Filho JC, et al. Differential gene expression analysis of iodide-treated rat thyroid follicular cell line PCCl3. Genomics, 2008,91:356-66.
    58 Shimura H, Shimura Y, Ohmori M, et al. Single strand DNA-binding proteins and thyroid transcription factor-1 conjointly regulate thyrotropin receptor gene expression. Mol Endocrinol, 1995,9:527-39.
    59 Schmitt TL, Espinoza CR, Loos U. Transcriptional regulation of the humansodium/iodide symporter gene by Pax8 and TTF-1. Exp Clin Endocrinol Diabetes, 2001,109:27-31.
    60 Ohno M, Zannini M, Levy O, et al. The paired-domain transcription factor Pax8 binds to the upstream enhancer of the rat sodium/iodide symporter gene and participates in both thyroid-specific and cyclic-AMP-dependent transcription. Mol Cell Biol, 1999,19:2051-60.
    61 Altmann A, Schulz RB, Glensch G, et al. Effects of Pax8 and TTF-1 thyroid transcription factor gene transfer in hepatoma cells: imaging of functional protein-protein interaction and iodide uptake. J Nucl Med, 2005,46:831-9.
    62 Di Palma T, Nitsch R, Mascia A, et al. The paired domain-containing factor Pax8 and the homeodomain-containing factor TTF-1 directly interact and synergistically activate transcription. J Biol Chem, 2003,278:3395-402.
    63 Presta I, Arturi F, Ferretti E, et al. Recovery of NIS expression in thyroid cancer cells by overexpression of Pax8 gene. BMC Cancer, 2005,5:80.
    64 Yen PM. Physiological and molecular basis of thyroid hormone action. Physiol Rev, 2001,81:1097-142.
    65 Sandler B, Webb P, Apriletti JW, et al. Thyroxine-thyroid hormone receptor interactions. J Biol Chem, 2004,279:55801-8.
    66 Gutekunst R, Smolarek H, Hasenpusch U, et al. Goitre epidemiology: thyroid volume, iodine excretion, thyroglobulin and thyrotropin in Germany and Sweden. Acta Endocrinol (Copenh), 1986,112:494-501.
    67 Meinhold H, Campos-Barros A, Behne D. Effects of selenium and iodine deficiency on iodothyronine deiodinases in brain, thyroid and peripheral tissue. Acta Med Austriaca, 1992,19 Suppl 1:8-12.
    68 Stubner D, Gartner R, Greil W, et al. Hypertrophy and hyperplasia during goitre growth and involution in rats--separate bioeffects of TSH and iodine. Acta Endocrinol (Copenh), 1987,116:537-48.
    69 Pisarev MA. Thyroid autoregulation. J Endocrinol Invest, 1985,8:475-84.
    70 Capen CC. Mechanisms of chemical injury of thyroid gland. Prog Clin BiolRes, 1994,387:173-91.
    71 Dohan O, De la Vieja A, Paroder V, et al. The sodium/iodide Symporter (NIS): characterization, regulation, and medical significance. Endocr Rev, 2003,24:48-77.
    72 Lacroix L, Pourcher T, Magnon C, et al. Expression of the apical iodide transporter in human thyroid tissues: a comparison study with other iodide transporters. J Clin Endocrinol Metab, 2004,89:1423-8.
    73 Selmi-Ruby S, Watrin C, Trouttet-Masson S, et al. The porcine sodium/iodide symporter gene exhibits an uncommon expression pattern related to the use of alternative splice sites not present in the human or murine species. Endocrinology, 2003,144:1074-85.
    74 Corvilain B, Collyn L, van Sande J, et al. Stimulation by iodide of H(2)O(2) generation in thyroid slices from several species. Am J Physiol Endocrinol Metab, 2000,278:E692-9.
    75 Theodoropoulou A, Vagenakis AG, Makri M, et al. Thyroid hormone synthesis and secretion in humans after 80 milligrams of iodine for 15 days and subsequent withdrawal. J Clin Endocrinol Metab, 2007,92:212-4.
    76 Pitsiavas V, Smerdely P, Li M, et al. Amiodarone induces a different pattern of ultrastructural change in the thyroid to iodine excess alone in both the BB/W rat and the Wistar rat. Eur J Endocrinol, 1997,137:89-98.
    77 Schmitt TL, Espinoza CR, Loos U. Transcriptional regulation of the human sodium/iodide symporter gene by Pax8 and TTF-1. Exp Clin Endocrinol Diabetes, 2001,109:27-31.
    78 Riesco G, Taurog A, Larsen R, et al. Acute and chronic responses to iodine deficiency in rats. Endocrinology, 1977,100:303-13.
    79 Pisarev MA, Thomasz L, Juvenal GJ. Role of transforming growth factor beta in the regulation of thyroid function and growth. Thyroid, 2009,19:881-92.
    80 Taurog A. Thyroid peroxidase-catalyzed iodination of thyroglobulin; inhibition by excess iodide. Arch Biochem Biophys, 1970,139:212-20.
    81 Thomasz L, Oglio R, Rivandeira DT, et al. Inhibition of goiter growth and of cyclic AMP formation in rat thyroid by 2-iodohexadecanal. Mol Cell Endocrinol, 2010,317:141-7.
    82 Cochaux P, Van Sande J, Swillens S, et al. Iodide-induced inhibition of adenylate cyclase activity in horse and dog thyroid. Eur J Biochem, 1987,170:435-42.
    83 Panneels V, Van den Bergen H, Jacoby C, et al. Inhibition of H2O2 production by iodoaldehydes in cultured dog thyroid cells. Mol Cell Endocrinol, 1994,102:167-76.
    84 Bruno R, Ferretti E, Tosi E, et al. Modulation of thyroid-specific gene expression in normal and nodular human thyroid tissues from adults: an in vivo effect of thyrotropin. J Clin Endocrinol Metab, 2005,90:5692-7.
    85 Postiglione MP, Parlato R, Rodriguez-Mallon A, et al. Role of the thyroid-stimulating hormone receptor signaling in development and differentiation of the thyroid gland. Proc Natl Acad Sci U S A, 2002,99:15462-7.
    86 Marians RC, Ng L, Blair HC, et al. Defining thyrotropin-dependent and -independent steps of thyroid hormone synthesis by using thyrotropin receptor-null mice. Proc Natl Acad Sci U S A, 2002,99:15776-81.
    87 Suh JM, Song JH, Kim DW, et al. Regulation of the phosphatidylinositol 3-kinase, Akt/protein kinase B, FRAP/mammalian target of rapamycin, and ribosomal S6 kinase 1 signaling pathways by thyroid-stimulating hormone (TSH) and stimulating type TSH receptor antibodies in the thyroid gland. J Biol Chem, 2003,278:21960-71.
    88 Mirebeau-Prunier D, Guyetant S, Rodien P, et al. Decreased expression of thyrotropin receptor gene suggests a high-risk subgroup for oncocytic adenoma. Eur J Endocrinol, 2004,150:269-76.
    89 Ohmori M, Ohta M, Shimura H, et al. Cloning of the single strand DNA-binding protein important for maximal expression and thyrotropin(TSH)-induced negative regulation of the TSH receptor. Mol Endocrinol, 1996,10:1407-24.
    90 Medina DL, Suzuki K, Pietrarelli M, et al. Role of insulin and serum on thyrotropin regulation of thyroid transcription factor-1 and pax-8 genes expression in FRTL-5 thyroid cells. Thyroid, 2000,10:295-303.
    91 Perrone L, Pasca di Magliano M, Zannini M, et al. The thyroid transcription factor 2 (TTF-2) is a promoter-specific DNA-binding independent transcriptional repressor. Biochem Biophys Res Commun, 2000,275:203-8.
    92 Whitley RJ, Ain KB. Thyroglobulin: a specific serum marker for the management of thyroid carcinoma. Clin Lab Med, 2004,24:29-47.
    93 Harish K. Thyroglobulin: current status in differentiated thyroid carcinoma (review). Endocr Regul, 2006,40:53-67.
    94 Dunn JT, Dunn AD. Update on intrathyroidal iodine metabolism. Thyroid, 2001,11:407-14.
    95 Dunn JT, Anderson PC, Fox JW, et al. The sites of thyroid hormone formation in rabbit thyroglobulin. J Biol Chem, 1987,262:16948-52.
    96 Everett LA, Glaser B, Beck JC, et al. Pendred syndrome is caused by mutations in a putative sulphate transporter gene (PDS). Nat Genet, 1997,17:411-22.
    97 Herzog V, Berndorfer U, Saber Y. Isolation of insoluble secretory product from bovine thyroid: extracellular storage of thyroglobulin in covalently cross-linked form. J Cell Biol, 1992,118:1071-83.
    98 Baudry N, Lejeune PJ, Delom F, et al. Role of multimerized porcine thyroglobulin in iodine storage. Biochem Biophys Res Commun, 1998,242:292-6.
    99 Roti E, Uberti ED. Iodine excess and hyperthyroidism. Thyroid, 2001,11:493-500.
    100 Giraud A, Siffroi S, Lanet J, et al. Binding and internalization of thyroglobulin: selectivity, pH dependence, and lack of tissue specificity.Endocrinology, 1997,138:2325-32.
    101 Lemansky P, Herzog V. Endocytosis of thyroglobulin is not mediated by mannose-6-phosphate receptors in thyrocytes. Evidence for low-affinity-binding sites operating in the uptake of thyroglobulin. Eur J Biochem, 1992,209:111-9.
    102 Vuchak LA, Tsygankova OM, Prendergast GV, et al. Protein kinase A and B-Raf mediate extracellular signal-regulated kinase activation by thyrotropin. Mol Pharmacol, 2009,76:1123-9.
    103 Van Heuverswyn B, Leriche A, Van Sande J, et al. Transcriptional control of thyroglobulin gene expression by cyclic AMP. FEBS Lett, 1985,188:192-6.
    104 Pratt MA, Eggo MC, Bachrach LK, et al. Regulation of thyroperoxidase, thyroglobulin and iodide levels in sheep thyroid cells by TSH, tumor promoters and epidermal growth factor. Biochimie, 1989,71:227-35.
    105 Kim WB, Lewis CJ, McCall KD, et al. Overexpression of Wnt-1 in thyrocytes enhances cellular growth but suppresses transcription of the thyroperoxidase gene via different signaling mechanisms. J Endocrinol, 2007,193:93-106.
    106 Nagayama Y, Yamashita S, Hirayu H, et al. Regulation of thyroid peroxidase and thyroglobulin gene expression by thyrotropin in cultured human thyroid cells. J Clin Endocrinol Metab, 1989,68:1155-9.
    107 Gerard CM, Lefort A, Christophe D, et al. Control of thyroperoxidase and thyroglobulin transcription by cAMP: evidence for distinct regulatory mechanisms. Mol Endocrinol, 1989,3:2110-8.
    108 van den Hove MF, Croizet-Berger K, Tyteca D, et al. Thyrotropin activates guanosine 5'-diphosphate/guanosine 5'-triphosphate exchange on the rate-limiting endocytic catalyst, Rab5a, in human thyrocytes in vivo and in vitro. J Clin Endocrinol Metab, 2007,92:2803-10.
    109 Cauvi D, Venot N, Nlend MC, et al. Thyrotropin and iodide regulate sulfate concentration in thyroid cells. Relationship to thyroglobulin sulfation. Can J Physiol Pharmacol, 2003,81:1131-8.
    110 Muscella A, Marsigliante S, Verri T, et al. PKC-epsilon-dependent cytosol-to-membrane translocation of pendrin in rat thyroid PC Cl3 cells. J Cell Physiol, 2008,217:103-12.
    111 Chung JK. Sodium iodide symporter: its role in nuclear medicine. J Nucl Med, 2002,43:1188-200.
    112 Riesco-Eizaguirre G, Santisteban P. A perspective view of sodium iodide symporter research and its clinical implications. Eur J Endocrinol, 2006,155:495-512.
    113 Riedel C, Levy O, Carrasco N. Post-transcriptional regulation of the sodium/iodide symporter by thyrotropin. J Biol Chem, 2001,276:21458-63.
    114 Kogai T, Endo T, Saito T, et al. Regulation by thyroid-stimulating hormone of sodium/iodide symporter gene expression and protein levels in FRTL-5 cells. Endocrinology, 1997,138:2227-32.
    115 Kogai T, Sajid-Crockett S, Newmarch LS, et al. Phosphoinositide-3-kinase inhibition induces sodium/iodide symporter expression in rat thyroid cells and human papillary thyroid cancer cells. J Endocrinol, 2008,199:243-52.
    116 Nguyen LQ, Kopp P, Martinson F, et al. A dominant negative CREB (cAMP response element-binding protein) isoform inhibits thyrocyte growth, thyroid-specific gene expression, differentiation, and function. Mol Endocrinol, 2000,14:1448-61.
    117 Leal AL, Pantaleao TU, Moreira DG, et al. Hypothyroidism and hyperthyroidism modulates Ras-MAPK intracellular pathway in rat thyroids. Endocrine, 2007,31:174-8.
    118 De Gregorio G, Coppa A, Cosentino C, et al. The p85 regulatory subunit of PI3K mediates TSH-cAMP-PKA growth and survival signals. Oncogene, 2007,26:2039-47.
    119 Brewer C, Yeager N, Di Cristofano A. Thyroid-stimulating hormone initiated proliferative signals converge in vivo on the mTOR kinase without activating AKT. Cancer Res, 2007,67:8002-6.
    120 Shimura H, Okajima F, Ikuyama S, et al. Thyroid-specific expression and cyclic adenosine 3',5'-monophosphate autoregulation of the thyrotropin receptor gene involves thyroid transcription factor-1. Mol Endocrinol, 1994,8:1049-69.
    121 Mascia A, Nitsch L, Di Lauro R, et al. Hormonal control of the transcription factor Pax8 and its role in the regulation of thyroglobulin gene expression in thyroid cells. J Endocrinol, 2002,172:163-76.
    122 Saito T, Endo T, Nakazato M, et al. Thyroid-stimulating hormone-induced down-regulation of thyroid transcription factor 1 in rat thyroid FRTL-5 cells. Endocrinology, 1997,138:602-6.
    123 Lonigro R, Donnini D, Fabbro D, et al. Thyroid-specific gene expression is differentially influenced by intracellular glutathione level in FRTL-5 cells. Endocrinology, 2000,141:901-9.
    124 Kambe F, Nomura Y, Okamoto T, et al. Redox regulation of thyroid-transcription factors, Pax-8 and TTF-1, is involved in their increased DNA-binding activities by thyrotropin in rat thyroid FRTL-5 cells. Mol Endocrinol, 1996,10:801-12.
    125 Tell G, Pines A, Pandolfi M, et al. APE/Ref-1 is controlled by both redox and cAMP-dependent mechanisms in rat thyroid cells. Horm Metab Res, 2002,34:303-10.
    126 Di Palma T, D'Andrea B, Liguori GL, et al. TAZ is a coactivator for Pax8 and TTF-1, two transcription factors involved in thyroid differentiation. Exp Cell Res, 2009,315:162-75.
    127 Esposito C, Miccadei S, Saiardi A, et al. PAX 8 activates the enhancer of the human thyroperoxidase gene. Biochem J, 1998,331 ( Pt 1):37-40.
    128 Ohmori M, Endo T, Harii N, et al. A novel thyroid transcription factor is essential for thyrotropin-induced up-regulation of Na+/I- symporter gene expression. Mol Endocrinol, 1998,12:727-36.
    129 Taki K, Kogai T, Kanamoto Y, et al. A thyroid-specific far-upstreamenhancer in the human sodium/iodide symporter gene requires Pax-8 binding and cyclic adenosine 3',5'-monophosphate response element-like sequence binding proteins for full activity and is differentially regulated in normal and thyroid cancer cells. Mol Endocrinol, 2002,16:2266-82.
    130 Miccadei S, De Leo R, Zammarchi E, et al. The synergistic activity of thyroid transcription factor 1 and Pax 8 relies on the promoter/enhancer interplay. Mol Endocrinol, 2002,16:837-46.
    131 Damante G. Thyroid defects due to Pax8 gene mutations. Eur J Endocrinol, 1998,139:563-6.
    132 Pasca di Magliano M, Di Lauro R, Zannini M. Pax8 has a key role in thyroid cell differentiation. Proc Natl Acad Sci U S A, 2000,97:13144-9.
    133 Trueba SS, Auge J, Mattei G, et al. PAX8, TITF1, and FOXE1 gene expression patterns during human development: new insights into human thyroid development and thyroid dysgenesis-associated malformations. J Clin Endocrinol Metab, 2005,90:455-62.
    1 Bruno R, Ferretti E, Tosi E, et al. Modulation of thyroid-specific gene expression in normal and nodular human thyroid tissues from adults: an in vivo effect of thyrotropin. J Clin Endocrinol Metab, 2005,90:5692-7.
    2 Braverman LE, Roti E. Effects of iodine on thyroid function. Acta Med Austriaca, 1996,23:4-9.
    3 Vassart G, Dumont JE. The thyrotropin receptor and the regulation of thyrocyte function and growth. Endocr Rev, 1992,13:596-611.
    4 Morton ME, Chaikoff IL, Rosenfeld S. Inhibiting effect of inorganic iodide on the formation in vitro of thyroxine and diiodotyrosine by surviving thyroid tissue. J Biol Chem, 1944:154:381-387.
    5 Gutekunst R, Smolarek H, Hasenpusch U, et al. Goitre epidemiology: thyroid volume, iodine excretion, thyroglobulin and thyrotropin in Germany and Sweden. Acta Endocrinol (Copenh), 1986,112:494-501.
    6 Meinhold H, Campos-Barros A, Behne D. Effects of selenium and iodine deficiency on iodothyronine deiodinases in brain, thyroid and peripheral tissue. Acta Med Austriaca, 1992,19 Suppl 1:8-12.
    7 Stubner D, Gartner R, Greil W, et al. Hypertrophy and hyperplasia during goitre growth and involution in rats--separate bioeffects of TSH and iodine. Acta Endocrinol (Copenh), 1987,116:537-48.
    8 Scipioni A, Ferretti E, Soda G, et al. hNIS protein in thyroid: the iodine supply influences its expression and localization. Thyroid, 2007,17:613-8.
    9 Pisarev MA. Thyroid autoregulation. J Endocrinol Invest, 1985,8:475-84.
    10 Capen CC. Mechanisms of chemical injury of thyroid gland. Prog Clin Biol Res, 1994,387:173-91.
    11 Dohan O, De la Vieja A, Paroder V, et al. The sodium/iodide Symporter (NIS): characterization, regulation, and medical significance. Endocr Rev, 2003,24:48-77.
    12 Lacroix L, Pourcher T, Magnon C, et al. Expression of the apical iodide transporter in human thyroid tissues: a comparison study with other iodide transporters. J Clin Endocrinol Metab, 2004,89:1423-8.
    13 Spitzweg C, Joba W, Morris JC, et al. Regulation of sodium iodide symporter gene expression in FRTL-5 rat thyroid cells. Thyroid, 1999,9:821-30.
    14 Uyttersprot N, Pelgrims N, Carrasco N, et al. Moderate doses of iodide in vivo inhibit cell proliferation and the expression of thyroperoxidase and Na+/I- symporter mRNAs in dog thyroid. Mol Cell Endocrinol, 1997,131:195-203.
    15 Espinoza CR, Schmitt TL, Loos U. Thyroid transcription factor 1 and Pax8 synergistically activate the promoter of the human thyroglobulin gene. J Mol Endocrinol, 2001,27:59-67.
    16 WOLFF J, CHAIKOFF IL. Plasma inorganic iodide as a homeostatic regulator of thyroid function. J Biol Chem, 1948,174:555-64.
    17 WOLFF J, CHAIKOFF IL, et al. The temporary nature of the inhibitory action of excess iodine on organic iodine synthesis in the normal thyroid. Endocrinology, 1949,45:504-13, illust.
    18 Corvilain B, Collyn L, van Sande J, et al. Stimulation by iodide of H(2)O(2) generation in thyroid slices from several species. Am J Physiol Endocrinol Metab, 2000,278:E692-9.
    19 BRAVERMAN LE, INGBAR SH. CHANGES IN THYROIDAL FUNCTION DURING ADAPTATION TO LARGE DOSES OF IODIDE. J Clin Invest, 1963,42:1216-31.
    20 Leoni SG, Galante PA, Ricarte-Filho JC, et al. Differential gene expression analysis of iodide-treated rat thyroid follicular cell line PCCl3. Genomics, 2008,91:356-66.
    21 Theodoropoulou A, Vagenakis AG, Makri M, et al. Thyroid hormone synthesis and secretion in humans after 80 milligrams of iodine for 15 days and subsequent withdrawal. J Clin Endocrinol Metab, 2007,92:212-4.
    22 Eng PH, Cardona GR, Fang SL, et al. Escape from the acute Wolff-Chaikoff effect is associated with a decrease in thyroid sodium/iodide symporter messenger ribonucleic acid and protein. Endocrinology, 1999,140:3404-10.
    23 Cardoso LC, Martins DC, Campos DV, et al. Effect of iodine or iopanoic acid on thyroid Ca2+/NADPH-dependent H2O2-generating activity and thyroperoxidase in toxic diffuse goiters. Eur J Endocrinol, 2002,147:293-8.
    24 Pregliasco L, Bocanera L, Krawiec L, et al. Effects of iodide on thyroglobulin biosynthesis in FRTL-5 cells. Thyroid, 1996,6:319-23.
    25 Pitsiavas V, Smerdely P, Li M, et al. Amiodarone induces a different pattern of ultrastructural change in the thyroid to iodine excess alone in both the BB/W rat and the Wistar rat. Eur J Endocrinol, 1997,137:89-98.
    26 Schmitt TL, Espinoza CR, Loos U. Transcriptional regulation of the human sodium/iodide symporter gene by Pax8 and TTF-1. Exp Clin Endocrinol Diabetes, 2001,109:27-31.
    27 Riesco G, Taurog A, Larsen R, et al. Acute and chronic responses to iodine deficiency in rats. Endocrinology, 1977,100:303-13.
    28 Pisarev MA, Thomasz L, Juvenal GJ. Role of transforming growth factor beta in the regulation of thyroid function and growth. Thyroid, 2009,19:881-92.
    29 Ferreira AC, Lima LP, Araujo RL, et al. Rapid regulation of thyroid sodium-iodide symporter activity by thyrotrophin and iodine. J Endocrinol, 2005,184:69-76.
    30 Grollman EF, Smolar A, Ommaya A, et al. Iodine suppression of iodide uptake in FRTL-5 thyroid cells. Endocrinology, 1986,118:2477-82.
    31 Taurog A. Thyroid peroxidase-catalyzed iodination of thyroglobulin; inhibition by excess iodide. Arch Biochem Biophys, 1970,139:212-20.
    32 Thomasz L, Oglio R, Rivandeira DT, et al. Inhibition of goiter growth and ofcyclic AMP formation in rat thyroid by 2-iodohexadecanal. Mol Cell Endocrinol, 2010,317:141-7.
    33 Filetti S, Rapoport B. Evidence that organic iodine attenuates the adenosine 3',5'-monophosphate response to thyrotropin stimulation in thyroid tissue by an action at or near the adenylate cyclase catalytic unit. Endocrinology, 1983,113:1608-15.
    34 Cochaux P, Van Sande J, Swillens S, et al. Iodide-induced inhibition of adenylate cyclase activity in horse and dog thyroid. Eur J Biochem, 1987,170:435-42.
    35 Dugrillon A. Iodolactones and iodoaldehydes--mediators of iodine in thyroid autoregulation. Exp Clin Endocrinol Diabetes, 1996,104 Suppl 4:41-5.
    36 Panneels V, Van den Bergen H, Jacoby C, et al. Inhibition of H2O2 production by iodoaldehydes in cultured dog thyroid cells. Mol Cell Endocrinol, 1994,102:167-76.
    37 Suzuki K, Kohn LD. Differential regulation of apical and basal iodide transporters in the thyroid by thyroglobulin. J Endocrinol, 2006,189:247-55.
    38 Suzuki K, Mori A, Lavaroni S, et al. In vivo expression of thyroid transcription factor-1 RNA and its relation to thyroid function and follicular heterogeneity: identification of follicular thyroglobulin as a feedback suppressor of thyroid transcription factor-1 RNA levels and thyroglobulin synthesis. Thyroid, 1999,9:319-31.
    39 Suzuki K, Mori A, Saito J, et al. Follicular thyroglobulin suppresses iodide uptake by suppressing expression of the sodium/iodide symporter gene. Endocrinology, 1999,140:5422-30.
    40 Herzog V, Berndorfer U, Saber Y. Isolation of insoluble secretory product from bovine thyroid: extracellular storage of thyroglobulin in covalently cross-linked form. J Cell Biol, 1992,118:1071-83.
    41 Baudry N, Lejeune PJ, Delom F, et al. Role of multimerized porcine thyroglobulin in iodine storage. Biochem Biophys Res Commun,1998,242:292-6.
    42 Roti E, Uberti ED. Iodine excess and hyperthyroidism. Thyroid, 2001,11:493-500.
    43 Vuchak LA, Tsygankova OM, Prendergast GV, et al. Protein kinase A and B-Raf mediate extracellular signal-regulated kinase activation by thyrotropin. Mol Pharmacol, 2009,76:1123-9.
    44 Mirebeau-Prunier D, Guyetant S, Rodien P, et al. Decreased expression of thyrotropin receptor gene suggests a high-risk subgroup for oncocytic adenoma. Eur J Endocrinol, 2004,150:269-76.
    45 Van Heuverswyn B, Leriche A, Van Sande J, et al. Transcriptional control of thyroglobulin gene expression by cyclic AMP. FEBS Lett, 1985,188:192-6.
    46 Pratt MA, Eggo MC, Bachrach LK, et al. Regulation of thyroperoxidase, thyroglobulin and iodide levels in sheep thyroid cells by TSH, tumor promoters and epidermal growth factor. Biochimie, 1989,71:227-35.
    47 Kim WB, Lewis CJ, McCall KD, et al. Overexpression of Wnt-1 in thyrocytes enhances cellular growth but suppresses transcription of the thyroperoxidase gene via different signaling mechanisms. J Endocrinol, 2007,193:93-106.
    48 Nagayama Y, Yamashita S, Hirayu H, et al. Regulation of thyroid peroxidase and thyroglobulin gene expression by thyrotropin in cultured human thyroid cells. J Clin Endocrinol Metab, 1989,68:1155-9.
    49 Gerard CM, Lefort A, Christophe D, et al. Control of thyroperoxidase and thyroglobulin transcription by cAMP: evidence for distinct regulatory mechanisms. Mol Endocrinol, 1989,3:2110-8.
    50 van den Hove MF, Croizet-Berger K, Tyteca D, et al. Thyrotropin activates guanosine 5'-diphosphate/guanosine 5'-triphosphate exchange on the rate-limiting endocytic catalyst, Rab5a, in human thyrocytes in vivo and in vitro. J Clin Endocrinol Metab, 2007,92:2803-10.
    51 Cauvi D, Venot N, Nlend MC, et al. Thyrotropin and iodide regulate sulfate concentration in thyroid cells. Relationship to thyroglobulin sulfation. Can JPhysiol Pharmacol, 2003,81:1131-8.
    52 Muscella A, Marsigliante S, Verri T, et al. PKC-epsilon-dependent cytosol-to-membrane translocation of pendrin in rat thyroid PC Cl3 cells. J Cell Physiol, 2008,217:103-12.
    53 Altmann A, Schulz RB, Glensch G, et al. Effects of Pax8 and TTF-1 thyroid transcription factor gene transfer in hepatoma cells: imaging of functional protein-protein interaction and iodide uptake. J Nucl Med, 2005,46:831-9.
    54 Chung JK. Sodium iodide symporter: its role in nuclear medicine. J Nucl Med, 2002,43:1188-200.
    55 Riesco-Eizaguirre G, Santisteban P. A perspective view of sodium iodide symporter research and its clinical implications. Eur J Endocrinol, 2006,155:495-512.
    56 Riedel C, Levy O, Carrasco N. Post-transcriptional regulation of the sodium/iodide symporter by thyrotropin. J Biol Chem, 2001,276:21458-63.
    57 Kogai T, Endo T, Saito T, et al. Regulation by thyroid-stimulating hormone of sodium/iodide symporter gene expression and protein levels in FRTL-5 cells. Endocrinology, 1997,138:2227-32.
    58 Kogai T, Sajid-Crockett S, Newmarch LS, et al. Phosphoinositide-3-kinase inhibition induces sodium/iodide symporter expression in rat thyroid cells and human papillary thyroid cancer cells. J Endocrinol, 2008,199:243-52.
    59 Nguyen LQ, Kopp P, Martinson F, et al. A dominant negative CREB (cAMP response element-binding protein) isoform inhibits thyrocyte growth, thyroid-specific gene expression, differentiation, and function. Mol Endocrinol, 2000,14:1448-61.
    60 Postiglione MP, Parlato R, Rodriguez-Mallon A, et al. Role of the thyroid-stimulating hormone receptor signaling in development and differentiation of the thyroid gland. Proc Natl Acad Sci U S A, 2002,99:15462-7.
    61 Leal AL, Pantaleao TU, Moreira DG, et al. Hypothyroidism andhyperthyroidism modulates Ras-MAPK intracellular pathway in rat thyroids. Endocrine, 2007,31:174-8.
    62 De Gregorio G, Coppa A, Cosentino C, et al. The p85 regulatory subunit of PI3K mediates TSH-cAMP-PKA growth and survival signals. Oncogene, 2007,26:2039-47.
    63 Brewer C, Yeager N, Di Cristofano A. Thyroid-stimulating hormone initiated proliferative signals converge in vivo on the mTOR kinase without activating AKT. Cancer Res, 2007,67:8002-6.
    64 Shimura H, Shimura Y, Ohmori M, et al. Single strand DNA-binding proteins and thyroid transcription factor-1 conjointly regulate thyrotropin receptor gene expression. Mol Endocrinol, 1995,9:527-39.
    65 Shimura H, Okajima F, Ikuyama S, et al. Thyroid-specific expression and cyclic adenosine 3',5'-monophosphate autoregulation of the thyrotropin receptor gene involves thyroid transcription factor-1. Mol Endocrinol, 1994,8:1049-69.
    66 Medina DL, Suzuki K, Pietrarelli M, et al. Role of insulin and serum on thyrotropin regulation of thyroid transcription factor-1 and pax-8 genes expression in FRTL-5 thyroid cells. Thyroid, 2000,10:295-303.
    67 Perrone L, Pasca di Magliano M, Zannini M, et al. The thyroid transcription factor 2 (TTF-2) is a promoter-specific DNA-binding independent transcriptional repressor. Biochem Biophys Res Commun, 2000,275:203-8.
    68 Ohmori M, Ohta M, Shimura H, et al. Cloning of the single strand DNA-binding protein important for maximal expression and thyrotropin (TSH)-induced negative regulation of the TSH receptor. Mol Endocrinol, 1996,10:1407-24.
    69 Mascia A, Nitsch L, Di Lauro R, et al. Hormonal control of the transcription factor Pax8 and its role in the regulation of thyroglobulin gene expression in thyroid cells. J Endocrinol, 2002,172:163-76.
    70 Saito T, Endo T, Nakazato M, et al. Thyroid-stimulating hormone-induceddown-regulation of thyroid transcription factor 1 in rat thyroid FRTL-5 cells. Endocrinology, 1997,138:602-6.
    71 Presta I, Arturi F, Ferretti E, et al. Recovery of NIS expression in thyroid cancer cells by overexpression of Pax8 gene. BMC Cancer, 2005,5:80.
    72 Lonigro R, Donnini D, Fabbro D, et al. Thyroid-specific gene expression is differentially influenced by intracellular glutathione level in FRTL-5 cells. Endocrinology, 2000,141:901-9.
    73 Kambe F, Nomura Y, Okamoto T, et al. Redox regulation of thyroid-transcription factors, Pax-8 and TTF-1, is involved in their increased DNA-binding activities by thyrotropin in rat thyroid FRTL-5 cells. Mol Endocrinol, 1996,10:801-12.
    74 Tell G, Pines A, Pandolfi M, et al. APE/Ref-1 is controlled by both redox and cAMP-dependent mechanisms in rat thyroid cells. Horm Metab Res, 2002,34:303-10.
    75 Di Palma T, D'Andrea B, Liguori GL, et al. TAZ is a coactivator for Pax8 and TTF-1, two transcription factors involved in thyroid differentiation. Exp Cell Res, 2009,315:162-75.
    76 Di Palma T, Nitsch R, Mascia A, et al. The paired domain-containing factor Pax8 and the homeodomain-containing factor TTF-1 directly interact and synergistically activate transcription. J Biol Chem, 2003,278:3395-402.
    77 Esposito C, Miccadei S, Saiardi A, et al. PAX 8 activates the enhancer of the human thyroperoxidase gene. Biochem J, 1998,331 ( Pt 1):37-40.
    78 Ohmori M, Endo T, Harii N, et al. A novel thyroid transcription factor is essential for thyrotropin-induced up-regulation of Na+/I- symporter gene expression. Mol Endocrinol, 1998,12:727-36.
    79 Taki K, Kogai T, Kanamoto Y, et al. A thyroid-specific far-upstream enhancer in the human sodium/iodide symporter gene requires Pax-8 binding and cyclic adenosine 3',5'-monophosphate response element-like sequence binding proteins for full activity and is differentially regulated in normal andthyroid cancer cells. Mol Endocrinol, 2002,16:2266-82.
    80 Miccadei S, De Leo R, Zammarchi E, et al. The synergistic activity of thyroid transcription factor 1 and Pax 8 relies on the promoter/enhancer interplay. Mol Endocrinol, 2002,16:837-46.
    81 Perez Perez F. [Comparative study of iodine and thyroid gland in biological processes: development and reproduction]. An R Acad Nac Med (Madr), 2000,117:395-425.
    82 Yoshida A, Taniguchi S, Hisatome I, et al. Pendrin is an iodide-specific apical porter responsible for iodide efflux from thyroid cells. J Clin Endocrinol Metab, 2002,87:3356-61.
    83 Noguchi Y, Harii N, Giuliani C, et al. Thyroglobulin (Tg) induces thyroid cell growth in a concentration-specific manner by a mechanism other than thyrotropin/cAMP stimulation. Biochem Biophys Res Commun, 2010,391:890-4.
    84 Ambesi-Impiombato FS, Parks LA, Coon HG. Culture of hormone-dependent functional epithelial cells from rat thyroids. Proc Natl Acad Sci U S A, 1980,77:3455-9.
    85 Takasu N, Ohno S, Komiya I, et al. Requirements of follicle structure for thyroid hormone synthesis; cytoskeletons and iodine metabolism in polarized monolayer cells on collagen gel and in double layered, follicle-forming cells. Endocrinology, 1992,131:1143-8.
    86 Mauchamp J, Mirrione A, Alquier C, et al. Follicle-like structure and polarized monolayer: role of the extracellular matrix on thyroid cell organization in primary culture. Biol Cell, 1998,90:369-80.
    87 Nilsson M, Bjorkman U, Ekholm R, et al. Iodide transport in primary cultured thyroid follicle cells: evidence of a TSH-regulated channel mediating iodide efflux selectively across the apical domain of the plasma membrane. Eur J Cell Biol, 1990,52:270-81.
    88 Toda S, Sugihara H. Reconstruction of thyroid follicles from isolated porcinefollicle cells in three-dimensional collagen gel culture. Endocrinology, 1990,126:2027-34.
    89 Fayet G, Hovsepian S. Reconstruction of the thyroid follicle with long-term cultured cells. Thyroidology, 1991,3:123-5.
    90 Kogai T, Curcio F, Hyman S, et al. Induction of follicle formation in long-term cultured normal human thyroid cells treated with thyrotropin stimulates iodide uptake but not sodium/iodide symporter messenger RNA and protein expression. J Endocrinol, 2000,167:125-35.
    91 Hoshi N, Kusakabe T, Taylor BJ, et al. Side population cells in the mouse thyroid exhibit stem/progenitor cell-like characteristics. Endocrinology, 2007,148:4251-8.
    92 Goto K, Hosaka S, Ishii H, et al. Follicle-like hemicysts reorganized in monolayer culture of rat thyroid cells in the medium supplemented with isologous serum. Tohoku J Exp Med, 1991,164:223-30.
    93 Tognella C, Marti U, Peter HJ, et al. Follicle-forming cat thyroid cell lines synthesizing extracellular matrix and basal membrane components: a new tool for the study of thyroidal morphogenesis. J Endocrinol, 1999,163:505-14.
    94 Bernier-Valentin F, Trouttet-Masson S, Rabilloud R, et al. Three-dimensional organization of thyroid cells into follicle structures is a pivotal factor in the control of sodium/iodide symporter expression. Endocrinology, 2006,147:2035-42.

© 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