TSHR

TSHR
  • 文章类型: Journal Article
    甲状腺癌是最常见的内分泌肿瘤,分化型甲状腺癌(DTC)占诊断的95%。虽然大多数DTC患者诊断和治疗放射性碘(RAI),高达20%的DTC患者成为RAI难治性(RAI-R)。与RAI-R患者相比,RAI-R患者的生存率显着降低。本研究探讨了[89Zr]Zr-TR1402作为DTC的促甲状腺激素受体(TSHR)靶向PET放射性药物。[89Zr]Zr-TR1402是通过将重组人TSH(rhTSH)类似物TR1402与螯合剂p-SCN-Bn-去铁胺(DFO)以摩尔比为3:1(DFO/TR1402)并用89Zr(t1/2=78.4h,β+=22.7%)。由于TSHR在常见的DTC衍生细胞系中不存在,通过递送含有人TSHR基因全长编码区的慢病毒,通过稳定转导再引入TSHR。在稳定转导的TSHR+和野生型TSHR-DTC细胞系中体外评估受体介导的[89Zr]Zr-TR1402的摄取。在带有TSHR和TSHR-异种移植物的雄性和雌性无胸腺裸鼠注射后第1-3天进行体内PET成像,以及注射后第3天的离体生物分布。TSHR+THJ529T(P<0.0001)和FTC133(P<0.01)细胞对1nM[89Zr]Zr-TR1402的体外摄取显著高于TSHR-THJ529T和FTC133细胞。通过用250nmDFO-TR1402阻断摄取,显示这种摄取在TSHR+THJ529T(P<0.0001)和TSHR+FTC133(P<0.0001)细胞中都是特异性的。体内PET成像显示[89Zr]Zr-TR1402在TSHR+肿瘤中积累,这是第一天最高的。在雄性FTC133异种移植模型中,离体生物分布证实了FTC133+(1.3±0.1%ID/g)和FTC133-(0.8±0.1%ID/g)肿瘤之间的摄取的显著差异(P<0.001)。在男性THJ529T异种移植模型中,在THJ529T+(1.8±0.6%ID/g)和THJ529T-(0.8±0.4%ID/g)肿瘤之间也观察到摄取的显著差异(P<0.05)。[89Zr]Zr-TR1402在表达TSHR的DTC细胞系中的体外和体内积累支持该方法的持续临床前优化。
    Thyroid cancer is the most common endocrine cancer, with differentiated thyroid cancers (DTCs) accounting for 95% of diagnoses. While most DTC patients are diagnosed and treated with radioiodine (RAI), up to 20% of DTC patients become RAI refractory (RAI-R). RAI-R patients have significantly reduced survival rates compared to patients who remain RAI-avid. This study explores [89Zr]Zr-TR1402 as a thyroid-stimulating hormone receptor (TSHR)-targeted PET radiopharmaceutical for DTC. [89Zr]Zr-TR1402 was synthesized with a molar activity of 25.9 MBq/nmol by conjugating recombinant human TSH (rhTSH) analogue TR1402 to chelator p-SCN-Bn-deferoxamine (DFO) in a molar ratio of 3:1 (DFO/TR1402) and radiolabeling with 89Zr (t1/2 = 78.4 h, β+ = 22.7%). As TSHR is absent in commonly available DTC-derived cell lines, TSHR was reintroduced via stable transduction by delivering a lentivirus containing the full-length coding region of the human TSHR gene. Receptor-mediated uptake of [89Zr]Zr-TR1402 was evaluated in vitro in stably transduced TSHR+ and wild-type TSHR- DTC cell lines. In vivo PET imaging was performed on Days 1-3 postinjection in male and female athymic nude mice bearing TSHR+ and TSHR- xenografts, along with ex vivo biodistribution on Day 3 postinjection. In vitro uptake of 1 nM [89Zr]Zr-TR1402 was significantly higher in TSHR+ THJ529T (P < 0.0001) and FTC133 (P < 0.01) cells than in TSHR- THJ529T and FTC133 cells. This uptake was shown to be specific in both TSHR+ THJ529T (P < 0.0001) and TSHR+ FTC133 (P < 0.0001) cells by blocking uptake with 250 nm DFO-TR1402. In vivo PET imaging showed accumulation of [89Zr]Zr-TR1402 in TSHR+ tumors, which was the highest on Day 1. In the male FTC133 xenograft model, ex vivo biodistribution confirmed a significant difference (P < 0.001) in uptake between FTC133+ (1.3 ± 0.1%ID/g) and FTC133- (0.8 ± 0.1%ID/g) tumors. A significant difference (P < 0.05) in uptake was also seen in the male THJ529T xenograft model between THJ529T+ (1.8 ± 0.6%ID/g) and THJ529T- (0.8 ± 0.4%ID/g) tumors. The in vitro and in vivo accumulation of [89Zr]Zr-TR1402 in TSHR-expressing DTC cell lines support the continued preclinical optimization of this approach.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    促甲状腺激素(TSH),从垂体的促甲状腺激素合成和分泌的糖蛋白,由糖蛋白激素共同α亚基(CGA)和特异性β亚基(TSHB)组成。TSH的主要生物学功能是通过激活其同源受体来刺激甲状腺卵泡合成和分泌甲状腺激素。促甲状腺激素受体(TSHR)。在本研究中,分别制备了抗稻田鳗鱼Tshb和Tshr的多克隆抗血清,在mRNA和蛋白水平检测Tshb和Tshr的表达。RT-PCR分析表明,tshbmRNA主要在垂体以及包括卵巢和睾丸在内的一些垂体外组织中表达。TshrmRNA也以组织特异性方式表达,在包括肾脏在内的组织中检测到转录本,子房,和睾丸。垂体中的免疫反应性Tshb信号显示位于腺垂体的内部区域,该区域靠近成年稻田鳗鱼的神经垂体。在孵化时首先观察到稻田鳗鱼幼虫垂体中的Tshb免疫反应细胞。在稻田鳗鱼卵巢和睾丸中也检测到了免疫反应性Tshb和Cga的表达。在卵巢里,免疫反应性Tshb,Cga,在卵母细胞和颗粒细胞中观察到Tshr。在睾丸里,主要在支持细胞中观察到免疫反应性Tsh,而在生殖细胞和体细胞中检测到免疫反应性Cga和Tshr。本研究的结果表明,Tsh可能在卵巢和睾丸局部合成,可能在稻田鳗鱼的性腺发育中起旁分泌和/或自分泌作用。
    Thyroid stimulating hormone (TSH), a glycoprotein synthesized and secreted from thyrotrophs of the pituitary gland, is composed of a glycoprotein hormone common alpha subunit (CGA) and a specific beta subunit (TSHB). The major biological function of TSH is to stimulate thyroidal follicles to synthesize and secrete thyroid hormones through activating its cognate receptor, the thyroid stimulating hormone receptor (TSHR). In the present study, polyclonal antisera against ricefield eel Tshb and Tshr were generated respectively, and the expression of Tshb and Tshr was examined at mRNA and protein levels. RT-PCR analysis showed that tshb mRNA was expressed mainly in the pituitary as well as in some extrapituitary tissues including the ovary and testis. Tshr mRNA was also expressed in a tissue-specific manner, with transcripts detected in tissues including the kidney, ovary, and testis. The immunoreactive Tshb signals in the pituitary were shown to be localized to the inner areas of adenohypophysis which are close to the neurohypophysis of adult ricefield eels. Tshb-immunoreatvie cells in the pituitary of ricefield eel larvae were firstly observed at hatching. The expression of immunoreactive Tshb and Cga was also detected in ricefield eel ovary and testis together with Tshr. In the ovary, immunoreactive Tshb, Cga, and Tshr were observed in oocytes and granulosa cells. In the testis, immunoreactive Tshb was mainly observed in Sertoli cells while immunoreactive Cga and Tshr were detected in germ cells as well as somatic cells. Results of the present study suggest that Tsh may be synthesized both in the ovary and testis locally, which may play paracrine and/or autocrine roles in gonadal development in ricefield eels.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:本研究旨在建立能够代表临床疾病特征的Graves病(GD)伴Graves眼眶病(GO;GDGO)小鼠模型。
    方法:将胰岛素样生长因子1受体(IGF-1R)α亚基(pcDNA3.1/IGF-1Rα)和促甲状腺激素受体(TSHR)A亚基质粒(pcDNA3.1/TSHR-289)的真核表达质粒注射到雌性BALB/c小鼠中,然后立即电穿孔以诱导GDGO模型。根据注射频率(2至4周间隔)和注射质粒类型进行分组:T:pcDNA3.1/TSHR-289(),I:pcDNA3.1/IGF-1Rα(+),或共注射T+I:pcDNA3.1/TSHR-289(+)和pcDNA3.1/IGF-1Rα(+)。血清TSH,T4、TSAb、TSBAb,体重,并评估血糖水平。进行甲状腺99mTcO4-成像和球后磁共振成像(MRI),测量双侧眼肌体积。对相关组织进行免疫组织化学和苏木精-伊红染色,并进行半定量分析。
    结果:T组中总共有60%的小鼠(3/5,一只小鼠死亡)出现GD+GO。在T+I组中,83.3%的小鼠(5/6)发展为GD+GO。I组中的小鼠没有发展GD。与对照组相比,血清T4,TSAb,GD+GO模型组小鼠的TSBAb均有不同程度的升高(P<0.05),血清TSH和体重均显著低于对照组(P<0.05)。GD+GO组小鼠99mTcO4-的甲状腺摄取能力和眼肌体积明显高于对照组(P<0.05)。这些小鼠的甲状腺和球后肌肉显示出不同的炎症浸润和间质肌肉水肿。共注射组GD+GO的严重程度与注射频率无关;然而,与T组相比,共注射小鼠中的GD和眼部体征更严重。
    结论:我们通过重复共注射pcDNA3.1/IGF-1Rα和pcDNA3.1/TSHR-289质粒成功诱导了GD+GO小鼠模型。单独注射pcDNA3.1/IGF-1Rα不能诱导GD。两个质粒的共注射比单独的pcDNA3.1/TSHR-289(+)诱导更严重的GD+GO。
    OBJECTIVE: The study aimed to establish a mouse model of Graves\' disease (GD) with Graves\' orbitopathy (GO; GD + GO) that can represent the clinical disease characteristics.
    METHODS: A eukaryotic expression plasmid of insulin-like growth factor 1 receptor (IGF-1R) α subunit (pcDNA3.1/IGF-1Rα) and a thyrotropin receptor (TSHR) A subunit plasmid (pcDNA3.1/TSHR-289) were injected in female BALB/c mice followed by immediate electroporation to induce a GD + GO model. Grouping was performed according to the frequency of injection (2- to 4-week intervals) and type of injected plasmids: T: pcDNA3.1/TSHR-289( +), I: pcDNA3.1/IGF-1Rα( +), or co-injection T + I: pcDNA3.1/TSHR-289( +) and pcDNA3.1/IGF-1Rα( +). Serum TSH, T4, TSAb, TSBAb, body weight, and blood glucose levels were evaluated. Thyroid 99mTcO4- imaging and retrobulbar magnetic resonance imaging (MRI) were performed, and bilateral eye muscle volumes were measured. Immunohistochemistry and hematoxylin-eosin staining were performed on the relevant tissues, and semi-quantitative analysis was performed.
    RESULTS: A total of 60% of mice (3/5, one mouse died) in the T group developed GD + GO. In the T + I group, 83.3% of mice (5/6) developed GD + GO. Mice in the I group did not develop GD. Compared with the control group, serum T4, TSAb, and TSBAb of the mice in the GD + GO model groups were increased to varying degrees (P < 0.05), and serum TSH and body weight were significantly lower compared to the control group (P < 0.05). The thyroid uptake capacity of 99mTcO4- and the volume of eye muscle of mice in the GD + GO group were significantly higher compared to the control group (P < 0.05). The thyroid and retrobulbar muscles of these mice showed varying inflammatory infiltration and interstitial muscle edema. The severity of GD + GO in the co-injection group was not related to injection frequency; however, GD and ocular signs in co-injection mice were more severe compared to the T group.
    CONCLUSIONS: We successfully induced a GD + GO mouse model by a repeated co-injection of pcDNA3.1/IGF-1Rα and pcDNA3.1/TSHR-289 plasmids. Injection of pcDNA3.1/IGF-1Rα alone failed to induce GD. Co-injection of two plasmids induced more severe GD + GO than pcDNA3.1/TSHR-289( +) alone.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Graves眼病(GO)是Graves病的甲状腺外表现,眼眶成纤维细胞(OFs)被认为是GO发病机制的关键参与者,与眼眶炎症有关,组织重塑,和纤维化。这项研究提供了对GO(GO-OFs)的细胞行为和特征的初步探索,并与来自健康对照的OF(HC-OF)进行比较。结果表明,GO-OFs表现出从组织碎片的延迟迁移,而在GO-OF和HC-OF之间没有观察到细胞增殖的显著差异。Thy-1,TSHR,IGF-1R提示GO和GD之间共有自身抗原和途径,导致炎症和纤维化。细胞因子反应的研究揭示了GO-OFs中透明质酸(HA)和前列腺素E2(PGE2)的分泌升高,强调它们在组织重塑中的作用。这些发现加深了我们对OFs在GO发病机制中的理解,提供潜在的治疗途径。
    Graves\' ophthalmopathy (GO) is an extrathyroidal manifestation of Graves\' disease, Orbital fibroblasts (OFs) are recognized as key players in GO pathogenesis, involved in orbital inflammation, tissue remodeling, and fibrosis. This study offers a primary exploration of cell behavior and characteristics on OFs from GO (GO-OFs), and compared to OFs from healthy control (HC-OFs). Results reveal that GO-OFs exhibit delayed migration from tissue fragments, while no significant difference in cell proliferation is observed between GO-OFs and HC-OFs. Aberrant expression pattern of surface proteins Thy-1, TSHR, and IGF-1R suggests shared autoantigens and pathways between GO and GD, contributing to inflammation and fibrosis. Investigations into cytokine responses unveil elevated secretion of hyaluronic acid (HA) and prostaglandin E2 (PGE2) in GO-OFs, emphasizing their role in tissue remodeling. These findings deepen our understanding of OFs in GO pathogenesis, offering potential therapeutic avenues.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    促甲状腺激素受体(TSHR),aGPCR,是由刺激TSHR抗体引起的自身免疫性甲状腺功能亢进(Graves病)的主要抗原。虽然我们之前已经发布了TSHR的全长模型,包括其富含亮氨酸的结构域(LRD),连接区(LR)和跨膜结构域(TMD),到目前为止,只有用TSHR自身抗体稳定的部分LRD(aa21-261)已经结晶。最近,然而,全长TSHR的cryo-EM结构已经发表,但它们仅包括不完整的LR。我们现在已经使用了低温EM模型,将二硫键添加到LR,并进行更长的(3000ns)分子动力学(MD)模拟,以更新我们以前的整个全长TSHR模型,在存在和不存在TSH配体的情况下。和我们之前的工作一样,新模型嵌入脂质膜中,用水和反离子溶剂化。我们发现,3000ns分子动力学模拟表明,LRD和TMD的结构非常恒定,而LR,通常称为“铰链区”,再次显示出显著的灵活性,形成几个瞬态次级结构元素。新模拟的分析允许详细检查TSH结合对TSHR结构的影响。我们发现TSH的结构稳定作用,包括增加LR的稳定性,通过分析包括氢键在内的几种内在受体特性清楚地证明了这一点,LRD方向的波动,和回转半径。总之,我们能够量化TSHR的灵活性,并显示其在TSH结合后的稳定性增加。这些数据表明配体在指导受体的信号传导结构中的重要作用。
    The receptor for thyroid stimulating hormone (TSHR), a GPCR, is the primary antigen in autoimmune hyperthyroidism (Graves\' disease) caused by stimulating TSHR antibodies. While we have previously published a full length model of the TSHR, including its leucine rich domain (LRD), linker region (LR) and transmembrane domain (TMD), to date, only a partial LRD (aa 21-261) stabilized with TSHR autoantibodies has been crystallized. Recently, however, cryo-EM structures of the full-length TSHR have been published but they include only an incomplete LR. We have now utilized the cryo-EM models, added disulfide bonds to the LR and performed longer (3000 ns) molecular dynamic (MD) simulations to update our previous model of the entire full-length TSHR, with and without the presence of TSH ligand. As in our earlier work, the new model was embedded in a lipid membrane and was solvated with water and counterions. We found that the 3000 ns Molecular Dynamic simulations showed that the structure of the LRD and TMD were remarkably constant while the LR, known more commonly as the \"hinge region\", again showed significant flexibility, forming several transient secondary structural elements. Analysis of the new simulations permitted a detailed examination of the effect of TSH binding on the structure of the TSHR. We found a structure-stabilizing effect of TSH, including increased stability of the LR, which was clearly demonstrated by analyzing several intrinsic receptor properties including hydrogen bonding, fluctuation of the LRD orientation, and radius of gyration. In conclusion, we were able to quantify the flexibility of the TSHR and show its increased stability after TSH binding. These data indicated the important role of ligands in directing the signaling structure of a receptor.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    TSH受体(TSHR)跨膜结构域(TMD)生活在由脂质和水分子组成的质膜中。为了了解TSHR相关水分子的作用,我们使用了TMD的分子动力学模拟,并确定了推定的受体相关跨膜水通道网络。该结果通过具有和不具有TSH配体结合的全长TSHR的扩展模拟得到证实。尽管在通过TSHR蛋白的模拟中观察到的运输时间比通过脂质双层本身慢,但我们发现,通过TSHR穿越的水明显多于通过脂质双层的水,后者与TSH的结合增加了一倍以上。使用大鼠甲状腺细胞(FRTL-5)和钙黄绿素荧光技术,我们测量了主要的甲状腺细胞水转运蛋白水通道蛋白1和4阻断后的细胞体积。TSH显示出影响水运输的剂量依赖性能力,并且在刺激TSHR自身抗体时观察到类似的作用。小分子TSHR激动剂,它们是TMD的变构活化剂,也增强了水的运输,说明了TMD在这一现象中的作用。此外,在TSHRTMD内的两个激活基序中也绘制了水通道通路,提示水运动如何影响受体的激活.在TSH浓度高度可变的诸如甲状腺功能减退症和甲状腺功能亢进的病理生理状况中,TSH的这种作用可能极大地影响甲状腺细胞和TSHR表达的许多其他甲状腺外部位中的水运动,从而影响正常的细胞功能。
    The thyroid-stimulating hormone receptor (TSHR) transmembrane domain (TMD) is found in the plasma membrane and consists of lipids and water molecules. To understand the role of TSHR-associated water molecules, we used molecular dynamic simulations of the TMD and identified a network of putative receptor-associated transmembrane water channels. This result was confirmed with extended simulations of the full-length TSHR with and without TSH ligand binding. While the transport time observed in the simulations via the TSHR protein was slower than via the lipid bilayer itself, we found that significantly more water traversed via the TSHR than via the lipid bilayer, which more than doubled with the binding of TSH. Using rat thyroid cells (FRTL-5) and a calcein fluorescence technique, we measured cell volumes after blockade of aquaporins 1 and 4, the major thyroid cell water transporters. TSH showed a dose-dependent ability to influence water transport, and similar effects were observed with stimulating TSHR autoantibodies. Small molecule TSHR agonists, which are allosteric activators of the TMD, also enhanced water transport, illustrating the role of the TMD in this phenomenon. Furthermore, the water channel pathway was also mapped across 2 activation motifs within the TSHR TMD, suggesting how water movement may influence activation of the receptor. In pathophysiological conditions such as hypothyroidism and hyperthyroidism where TSH concentrations are highly variable, this action of TSH may greatly influence water movement in thyroid cells and many other extrathyroidal sites where the TSHR is expressed, thus affecting normal cellular function.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Graves病(GD)是一种突出的抗体介导的自身免疫性疾病,其特征是靶向促甲状腺激素受体(TSHR)的刺激抗体(TRAb)。靶向和消除产生TRAb的B淋巴细胞对GD具有重要的治疗潜力。在这项研究中,我们设计了一种称为TSHR-CAR-T的新型嵌合抗原受体T细胞(CAR-T)疗法。该CAR-T构建体包含与CD8跨膜和细胞内信号结构域融合的TSH受体的细胞外结构域(4-1BB)。TSHR-CAR-T细胞表现出在体外和体内识别并有效消除产生TRAb的B淋巴细胞的能力。利用这种基于自身抗原的嵌合受体,我们的研究结果表明,TSHR-CAR-T细胞为治疗抗体介导的自身免疫性疾病提供了一种有前途的创新的免疫治疗方法,包括GD。
    Graves\' disease (GD) is a prominent antibody-mediated autoimmune disorder characterized by stimulating antibodies (TRAb) that target the thyroid-stimulating hormone receptor (TSHR). Targeting and eliminating TRAb-producing B lymphocytes hold substantial therapeutic potential for GD. In this study, we engineered a novel chimeric antigen receptor T cell (CAR-T) therapy termed TSHR-CAR-T. This CAR-T construct incorporates the extracellular domain of the TSH receptor fused with the CD8 transmembrane and intracellular signal domain (4-1BB). TSHR-CAR-T cells demonstrated the ability to recognize and effectively eliminate TRAb-producing B lymphocytes both in vitro and in vivo. Leveraging this autoantigen-based chimeric receptor, our findings suggest that TSHR-CAR-T cells offer a promising and innovative immunotherapeutic approach for the treatment of antibody-mediated autoimmune diseases, including GD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Case Reports
    先天性甲状腺功能减退症(CH)是一种常见的异质性内分泌疾病。促甲状腺激素受体基因(TSHR)是与CH相关的主要候选基因之一。研究已经调查了特定临床特征与TSHR变体的存在之间的可能相关性。然而,只有少数报道关注CH患者的长期随访.在这里,我们介绍了一个与CH相关的TSHRp.Arg109Gln和p.Arg450His罕见的复合杂合变体,随访到青春期。患者在新生儿筛查期间血清TSH水平较高。在1月龄时开始口服左旋甲状腺素(l-T4)。超声显示甲状腺形态和血流正常。观察到I-123的摄取减少和高氯酸盐阴性试验。尽管青春期可以稳定地减少l-T4,但仍需要少量的l-T4。病人被诊断为原位,非甲状腺肿,永久CH。他没有非经典的TSH抗性。具有TSHRp.Arg109Gln复合杂合变体的患者表现出具有高TSH水平和正常或略低的fT4水平的永久性CH。在未来,基因型鉴定有助于预测长期预后,减少对详细检查的需求.需要更多的案例研究来确定CH的遗传变异与临床特征之间的关系。
    Congenital hypothyroidism (CH) is a common heterogeneous endocrine disorder. The thyroid-stimulating hormone receptor gene (TSHR) is one of the major candidate genes associated with CH. Studies have investigated the possible correlations between the specific clinical features and the presence of TSHR variants. However, only a few reports have focused on the long-term follow-up of patients with CH. Here we present a case of CH-associated TSHR p.Arg109Gln and p.Arg450His rare compound heterozygous variants, with a follow-up performed until adolescence. The patient had high serum TSH levels during newborn screening. Oral administration of levothyroxine (l-T4) was initiated at 1 month of age. The ultrasonogram revealed normal thyroid morphology and blood flow. Reduced uptake of I-123 and negative perchlorate test was observed. A small amount of l-T4 remained needed although l-T4 could be steadily reduced by puberty. The patient was diagnosed with orthotopic, nongoitrous, and permanent CH. He had no nonclassical TSH resistance. Patients with the TSHR p.Arg109Gln compound heterozygous variant exhibit permanent CH with high TSH levels and normal or slightly lower fT4 levels. In the future, genotype identification could help predict the long-term prognosis and reduce the requirement for detailed examinations. More case studies are needed to determine the relationship between genetic variants and clinical features in CH.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    针对甲状腺蛋白的自身抗体存在于几种甲状腺疾病中。促甲状腺激素受体(TSHR)是一种G蛋白偶联受体(GPCR),与促甲状腺激素(TSH)结合并刺激甲状腺素(T4)和三碘甲状腺原氨酸(T3)的产生。当被抗TSHR自身抗体激动时,甲状腺激素的异常产生可导致Graves病(GD)。在桥本甲状腺炎(HT)中,抗TSHR自身抗体靶向甲状腺进行免疫攻击。为了更好地了解抗TSHR抗体在甲状腺疾病中的作用,我们产生了一组具有多种亲和力的大鼠抗小鼠(M)TSHR单克隆抗体,TSH阻断,和激动剂活性。这些抗体可用于研究小鼠模型中甲状腺疾病的病因和治疗,并作为靶向甲状腺的蛋白质疗法的基础,用于HT或GD中的治疗。
    Autoantibodies against thyroid proteins are present in several thyroid diseases. Thyroid-stimulating hormone receptor (TSHR) is a G-protein-coupled receptor (GPCR) that binds to thyroid-stimulating hormone (TSH) and stimulates production of thyroxine (T4) and triiodothyronine (T3). When agonized by anti-TSHR autoantibodies, aberrant production of thyroid hormone can lead to Graves\' Disease (GD). In Hashimoto\'s thyroiditis (HT), anti-TSHR autoantibodies target the thyroid for immune attack. To better understand the role of anti-TSHR antibodies in thyroid disease, we generated a set of rat antimouse (m)TSHR monoclonal antibodies with a range of affinities, blocking of TSH, and agonist activity. These antibodies could be used to investigate the etiology and therapy of thyroid disease in mouse models and as building blocks in protein therapeutics that target the thyroid for treatment in either HT or GD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:我们研究了甲状腺细针穿刺(FNA)标本中检测到的促甲状腺激素受体(TSHR)突变的临床意义。
    方法:我们机构的病理档案在2018年至2021年之间进行了审查,以使用Thyroseq®分析显示TSHR突变的不确定(BethesdaIII和IV类)标本。
    结果:共2184例诊断为非典型性/意义不明的滤泡性病变(AUS/FLUS),2625例诊断为滤泡性肿瘤/可疑为滤泡性肿瘤(FN/SFN)。共有1735个AUS/FLUS和2339个SFN/FN接受了Thyroseq®分析;69个显示TSHR突变(1.6%,59女,10男,平均年龄:55岁)。10例表现为嗜酸细胞特征。在FNA的1年内,有12例患者接受了放射性核素扫描:11例功能亢进。结节大小与TSH水平弱相关。鉴定了22种不同的TSHR突变(最常见的:M453T)。在5例中发现了第二个突变(EZH1n=2,EIF1AXn=3)。在所有测序读数中,钠碘转运蛋白(NIS)mRNA的表达在0.01%-62.43%的范围内,49例(71%)升高。手术病理随访5例(除1例外,均为良性)。38例(平均:24个月;范围:7-47个月),34个(89.5%)结节保持稳定,3个(8%)结节大小增加。
    结论:甲状腺FNA样本中被分类为不确定的TSHR突变很少见,通常是良性的,并且通常与扫描自主性(如果执行)相关联。
    OBJECTIVE: We investigated the clinical significance of thyroid-stimulating hormone receptor (TSHR) mutations detected in thyroid fine needle aspiration (FNA) specimens.
    METHODS: The pathology archives at our institution were reviewed between 2018 and 2021 for indeterminate (Bethesda category III and IV) specimens with Thyroseq® analysis showing TSHR mutations.
    RESULTS: A total of 2184 cases diagnosed as atypia/follicular lesion of undetermined significance (AUS/FLUS), and 2625 diagnosed as follicular neoplasm/suspicious for follicular neoplasm (FN/SFN) were identified. A total of 1735 AUS/FLUS and 2339 SFN/FN underwent Thyroseq® analysis; 69 showed TSHR mutations (1.6%, 59 female, 10 male, average age: 55 years). Ten cases showed oncocytic features. Twelve patients underwent radionuclide scans within 1 year of FNA:11 were hyperfunctioning. Nodule size and TSH levels were weakly correlated. Twenty-two different TSHR mutations were identified (most common: M453T). A second mutation was found in five cases (EZH1 n = 2, and EIF1AX n = 3). The expression of sodium-iodide transporter (NIS) mRNA was in the range of 0.01%-62.43% out of all sequencing reads, and was elevated in 49 (71%) cases. Surgical pathology follow-up was available in five cases (all benign except one). On follow-up available for 38 cases (mean: 24 months; range: 7-47 months), 34 (89.5%) nodules remained stable and 3 (8%) increased in size.
    CONCLUSIONS: TSHR mutations in thyroid FNA samples classified as indeterminate are rare, generally benign, and commonly associated with autonomy on scan if performed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号