SLC5A5

SLC5A5
  • 文章类型: Journal Article
    甲状腺乳头状癌(PTC)手术后复发的可能性很高,术后应用促甲状腺激素(TSH)抑制和放射性碘(131I)治疗。本研究探讨了淋巴结转移(LNM)的潜在机制,旨在开发PTC的分化治疗方法。
    本研究通过分析2073例PTC患者的临床资料,探讨PTC淋巴结转移的危险因素。分析了癌症基因组图谱甲状腺癌(TCGA-THCA)和基因表达的基因表达综合(GEO)数据库,以鉴定基因表达与表型之间的相互关系。
    分析临床数据,我们发现男性性别,年龄较小,肿瘤较大,甲状腺外延伸(ETE)是淋巴结转移的危险因素(P<0.05)。相反,甲状腺功能参数,如TSH,FT3,FT4,TSH/FT3和TSH/FT4与LNM无关(P>0.05)。女性TSH水平较高(P<0.05)。基因表达分析显示男性SLC5A5表达下调,年轻的个人,那些有淋巴结转移的人,较低的SLC5A5水平与较差的无病生存率相关(P<0.05)。此外,我们对单细胞RNA测序(scRNA-seq)数据的检查表明,SLC5A5在肿瘤和淋巴结转移样本中的表达降低,与TSHR的表达呈正相关。
    TSH对PTC行为的影响尚不清楚,而依赖于SLC5A5的吸收131I的能力显示出不同性别和年龄的差异。我们得出结论,PTC的术后治疗应考虑性别和年龄引起的差异。
    UNASSIGNED: Thyroid papillary carcinoma (PTC) had a high possibility of recurrence after surgery, and thyroid stimulating hormone (TSH) suppression and radioactive iodine (131I) were used for postoperative therapy. This study explored the potential mechanism of lymph node metastasis (LNM) and aimed to develop differentiated treatments for PTC.
    UNASSIGNED: This study explored the risk factors of lymph node metastasis in PTC by analyzing the clinical information of 2073 cases. The Cancer Genome Atlas Thyroid Cancer (TCGA-THCA) and the Gene Expression Omnibus (GEO) databases of gene expression were analyzed to identify the interrelationships between gene expression to phenotype.
    UNASSIGNED: Analyzing clinical data, we found that male gender, younger age, larger tumor size, and extra-thyroidal extension (ETE) were risk significant risk factors for lymph node metastasis(P<0.05). Conversely, thyroid function parameters such as TSH, FT3, FT4, TSH/FT3, and TSH/FT4 didn\'t correlate with LNM(P>0.05), and TSH levels were observed to be higher in females(P<0.05). Gene expression analysis revealed that SLC5A5 was down-regulated in males, younger individuals, and those with lymph node metastasis, and a lower level of SLC5A5 was associated with a worse disease-free survival(P<0.05). Additionally, our examination of single-cell RNA sequencing (scRNA-seq) data indicated that SLC5A5 expression was reduced in tumors and lymph node metastasis samples, correlating positively with the expression of TSHR.
    UNASSIGNED: The impact of TSH on PTC behavior remained unclear, while the capacity for absorbing 131I in dependence on SLC5A5 showed variations across different genders and ages. We conclude that postoperative treatment of PTC should take into account the differences caused by gender and age.
    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
    溶质载体(SLC)家族是一大类膜转运蛋白。它们的功能障碍在甲状腺癌的发病机制中起着重要作用。最著名的SLC是碘化钠同向转运蛋白(NIS),甲状腺癌中也称为钠/碘共转运蛋白或溶质载体家族5成员5(SLC5A5)。甲状腺癌中NIS的失调是有据可查的。NIS在碘化物摄取中的作用在甲状腺癌的治疗中至关重要,特别是放射性碘化物(RAI)治疗。除了NIS,其他SLC成员可能会影响自噬,扩散,和甲状腺癌细胞的凋亡,表明SLC成员的改变可能会影响甲状腺癌演变过程中的不同细胞事件。SLC成员的表达可能会影响甲状腺对化学物质的摄取,提示靶向SLC成员可能是甲状腺癌的一种有希望的治疗策略.
    The solute carrier (SLC) family is a large group of membrane transport proteins. Their dysfunction plays an important role in the pathogenesis of thyroid cancer. The most well-known SLC is the sodium-iodide symporter (NIS), also known as sodium/iodide co-transporter or solute carrier family 5 member 5 (SLC5A5) in thyroid cancer. The dysregulation of NIS in thyroid cancer is well documented. The role of NIS in the uptake of iodide is critical in the treatment of thyroid cancer, radioactive iodide (RAI) therapy in particular. In addition to NIS, other SLC members may affect the autophagy, proliferation, and apoptosis of thyroid cancer cells, indicating that an alteration in SLC members may affect different cellular events in the evolution of thyroid cancer. The expression of the SLC members may impact the uptake of chemicals by the thyroid, suggesting that targeting SLC members may be a promising therapeutic strategy in thyroid cancer.
    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
    母鸡的产蛋性能在家禽业中具有重要的经济意义。母鸡母鸡的亲代遗传可能导致改善鸡蛋产量的不良选择,是一种受多种遗传因素影响的现象。然而,很少有研究描述育苗鸡卵巢退化的分子机制。这里,我们探索了蛋鸡卵巢卵泡调节的关键基因,使用RNA测序分析鸡和产蛋鸡的小卵泡。测序数据分析揭示了200个基因的差异表达,与细胞活化和代谢相关的生物过程中的主要富集。在这些基因中,我们关注溶质载体家族5成员5(SLC5A5),与肉鸡相比,产卵卵泡中的RNA表达水平明显更高。随后的细胞功能研究通过敲低鸡卵巢卵泡颗粒细胞(GC)中的SLC5A5导致与细胞增殖和类固醇激素合成相关的基因下调,同时促进与细胞凋亡相关的基因表达。这些发现表明SLC5A5缺乏导致增殖抑制,类固醇激素的合成和分泌,并促进鸡GCs的凋亡。我们的研究证明了SLC5A5在鸡GCs的发育和功能中的关键作用,阐明其在鸡卵巢卵泡发育的更广泛背景下的潜在意义,分子标记辅助育种技术为提高鸡的产蛋性能提供了一个有前景的靶标。
    The egg-laying performance of hens holds significant economic importance within the poultry industry. Broody inheritance of the parent stock of chickens can result in poor options for the improvement of egg production, and is a phenomenon influenced by multiple genetic factors. However, few studies have been conducted to delineate the molecular mechanism of ovarian regression in brooding chickens. Here, we explored the pivotal genes responsible for the regulation of ovarian follicles in laying hens, using RNA-sequencing analysis on the small ovarian follicles from broody and laying chickens. Sequencing data analysis revealed the differential expression of 200 genes, with a predominant enrichment in biological processes related to cell activation and metabolism. Among these genes, we focused on solute carrier family 5 member 5 (SLC5A5), which exhibited markedly higher RNA expression levels in follicles from laying compared with broody chickens. Subsequent cellular function studies with knockdown of SLC5A5 in chicken ovarian follicle granulosa cells (GCs) led to the down-regulation of genes associated with cell proliferation and steroid hormone synthesis, and concurrent promotion of gene expression linked to apoptosis. These findings indicated that SLC5A5 deficiency led to the inhibition of proliferation, steroid hormone synthesis and secretion, and promotion of apoptosis in chicken GCs. Our study demonstrated a pivotal role for SLC5A5 in the development and function of chicken GCs, shedding light on its potential significance in the broader context of chicken ovarian follicle development, and providing a prospective target to improve the egg-laying performance of chickens via molecular marker-assisted breeding technology.
    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
    氧气在支持地球上的生命中起着关键作用。它在高等真核生物中特别重要,它作为热力学上有利的末端电子受体促进生物能学,并且在细胞信号传导和发育中具有重要作用。许多人类疾病源于氧气不足或过量。尽管它具有根本性的重要性,我们缺乏在细胞和生物体中以高时空分辨率操纵氧气供应的方法。这里,我们引入了一个遗传系统,补充从ChLorite(SNORCL)释放的氧气,在活细胞中按需局部产生分子氧,通过利用将亚氯酸盐(ClO2-)转化为分子氧(O2)和氯化物(Cl-)的原核亚氯酸盐O2-裂解酶(Cld)。我们表明,活性Cld酶可以靶向人细胞的细胞质或线粒体,并且共表达亚氯酸盐转运蛋白会导致细胞内部产生分子氧,以响应外部添加的亚氯酸盐。这种第一代系统可以对氧气的产生进行精确的时间和空间控制,即时研究应用。在未来,我们预计基于SNORCL的技术将在研究中具有更广泛的应用,生物技术,和医学。
    Oxygen plays a key role in supporting life on our planet. It is particularly important in higher eukaryotes where it boosts bioenergetics as a thermodynamically favorable terminal electron acceptor and has important roles in cell signaling and development. Many human diseases stem from either insufficient or excessive oxygen. Despite its fundamental importance, we lack methods with which to manipulate the supply of oxygen with high spatiotemporal resolution in cells and in organisms. Here, we introduce a genetic system, SupplemeNtal Oxygen Released from ChLorite (SNORCL), for on-demand local generation of molecular oxygen in living cells, by harnessing prokaryotic chlorite O2-lyase (Cld) enzymes that convert chlorite (ClO2-) into molecular oxygen (O2) and chloride (Cl-). We show that active Cld enzymes can be targeted to either the cytosol or mitochondria of human cells, and that coexpressing a chlorite transporter results in molecular oxygen production inside cells in response to externally added chlorite. This first-generation system allows fine temporal and spatial control of oxygen production, with immediate research applications. In the future, we anticipate that technologies based on SNORCL will have additional widespread applications in research, biotechnology, and medicine.
    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
    脊椎动物钠-碘转运体(NIS或SLC5A5)将碘化物转运到合成甲状腺激素的甲状腺滤泡细胞中。SLC5A蛋白家族包括维生素的转运蛋白,矿物,和营养。高氯酸盐破坏SLC5A5功能,一种普遍的环境污染物,导致人类病理,尤其是甲状腺功能减退.高氯酸盐还会破坏模型动物的性发育,包括三松棘鱼(Gasterosteusaculeatus)和斑马鱼(Daniorerio),但其作用机制尚不清楚。为了检验SLC5A5旁系同源物在生殖器官发育所必需的组织中表达的假设,因此,似乎是介导高氯酸盐对性发育影响的候选人,我们首先调查了Slc5a旁系同源物的进化史,以更好地了解该基因家族的潜在功能轨迹.我们确定了相对于钠/胆碱协同转运蛋白(slc5a7)的两个slc5a旁系分子;这些进化枝是钠/碘化物和乳酸协同转运蛋白的NIS进化枝(slc5a5,slc5a6,slc5a8,slc5a8和slc5a12)和SGLTclade,5scl2,sla5和5crencorters。我们还表征了slc5a基因在发育过程中的表达模式。粘回胚胎和早期幼虫在结缔组织中表达NIS进化枝基因,软骨,牙齿,和甲状腺。Stickleback雄性和雌性在性腺中表达slc5a5及其旁系同源物。斑马鱼性别基因分型性腺上的单细胞转录组学(scRNA-seq)表明,表达NIS进化枝的细胞包括生殖细胞(slc5a5,slc5a6a,和slc5a6b)和性腺体细胞(slc5a8l)。这些结果与高氯酸盐通过与生殖组织中的slc5a5或其旁系同源物相互作用而对性发育产生影响的假设一致。这些发现显示slc5基因在stickleback和斑马鱼中的新表达结构域,这表明包括人类在内的脊椎动物的功能相似,并提供候选人来介导高氯酸盐对性发育的影响。
    The vertebrate sodium-iodide symporter (NIS or SLC5A5) transports iodide into the thyroid follicular cells that synthesize thyroid hormone. The SLC5A protein family includes transporters of vitamins, minerals, and nutrients. Disruption of SLC5A5 function by perchlorate, a pervasive environmental contaminant, leads to human pathologies, especially hypothyroidism. Perchlorate also disrupts the sexual development of model animals, including threespine stickleback (Gasterosteus aculeatus) and zebrafish (Danio rerio), but the mechanism of action is unknown. To test the hypothesis that SLC5A5 paralogs are expressed in tissues necessary for the development of reproductive organs, and therefore are plausible candidates to mediate the effects of perchlorate on sexual development, we first investigated the evolutionary history of Slc5a paralogs to better understand potential functional trajectories of the gene family. We identified two clades of slc5a paralogs with respect to an outgroup of sodium/choline cotransporters (slc5a7); these clades are the NIS clade of sodium/iodide and lactate cotransporters (slc5a5, slc5a6, slc5a8, slc5a8, and slc5a12) and the SGLT clade of sodium/glucose cotransporters (slc5a1, slc5a2, slc5a3, slc5a4, slc5a10, and slc5a11). We also characterized expression patterns of slc5a genes during development. Stickleback embryos and early larvae expressed NIS clade genes in connective tissue, cartilage, teeth, and thyroid. Stickleback males and females expressed slc5a5 and its paralogs in gonads. Single-cell transcriptomics (scRNA-seq) on zebrafish sex-genotyped gonads revealed that NIS clade-expressing cells included germ cells (slc5a5, slc5a6a, and slc5a6b) and gonadal soma cells (slc5a8l). These results are consistent with the hypothesis that perchlorate exerts its effects on sexual development by interacting with slc5a5 or its paralogs in reproductive tissues. These findings show novel expression domains of slc5 genes in stickleback and zebrafish, which suggest similar functions across vertebrates including humans, and provide candidates to mediate the effects of perchlorate on sexual development.
    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
    NIS is a potent iodide transporter encoded by the SLC5A5 gene. Its expression is reduced in papillary thyroid carcinoma (PTC). In this study we analyzed the impact of miR-181a-5p on NIS expression in the context of PTC. We used real-time PCR to analyze the expression of SLC5A5 and miR-181a-5p in 49 PTC/normal tissue pairs. Luciferase assays and mutagenesis were performed to confirm direct binding of miR-181a-5p to the 3\'UTR of SLC5A5 and identify the binding site. The impact of modulation of miR-181a-5p using appropriate plasmids on endogenous NIS and radioactive iodine accumulation was verified. We confirmed downregulation of SLC5A5 and concomitant upregulation of miR-181a-5p in PTC. Broadly used algorithms did not predict the binding site of miR-181a-5p in 3\'UTR of SLC5A5, but we identified and confirmed the binding site through mutagenesis using luciferase assays. In MCF7 and HEK293-flhNIS cell lines, transfection with mir-181a-expressing plasmid decreased endogenous SLC5A5, whereas silencing of miR-181a-5p increased it. We observed similar tendencies in protein expression and radioactive iodine accumulation. This study shows for the first time that miR-181a-5p directly regulates SLC5A5 expression in the context of PTC and may decrease efficacy of radioiodine treatment. Accordingly, miR-181a-5p may serve as an emerging target to enhance the efficacy of radioactive iodine therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Defects in the human sodium/iodide symporter (SLC5A5) gene have been reported to be one of the causes of congenital hypothyroidism (CH). We aimed to identify SLC5A5 mutations in Chinese patients with CH and to evaluate the function of the mutation.
    Two hundred and seventy-three patients with primary CH were screened for mutations in SLC5A5 using next-generation sequencing. We investigated the expression and cellular localization of the novel compound heterozygous mutation in SLC5A5. The functional activity of the mutants was further examined in vitro.
    In 273 patients with CH, two previously undescribed pathogenic mutations p.Gly51AlafsTer45 (G51fs) and p.Gly421Arg (G421R) in a compound heterozygous state in SLC5A5 were identified in a pediatric patient. G51fs was located in the first intercellular loop connecting transmembrane segment I and II, whereas G421R was in the transmembrane segment (TMS) XI. G51fs and G421R resulted in a truncated NIS and reduced protein expression, respectively. In vitro experiments further showed that the normal function of iodine transport of sodium-iodide symporter (NIS) mutants was markedly impaired.
    The undescribed compound heterozygous mutation of SLC5A5 was discovered in a Chinese CH patient. The mutation led to significantly reduced NIS expression and impaired iodide transport function accompanied by the impaired location of the NIS on the plasma membrane. Our study thus provides further insights into the roles of SLC5A5 in CH pathogenesis.
    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
    Iodine transportation is an important step in thyroid hormone biosynthesis. Uptake of iodine into the thyroid follicle is mediated mainly by the basolateral sodium-iodide symporter (NIS or solute carrier family 5 member 5: SLC5A5), and iodine efflux across the apical membrane into the follicular lumen is mediated by pendrin (SLC26A4). In addition to these transporters, SLC26A7, which has recently been identified as a causative gene for congenital hypothyroidism, was found to encode a novel apical iodine transporter in the thyroid. Although SLC5A5 and SLC26A4 have been well-characterized, little is known about SLC26A7, including its regulation by TSH, the central hormone regulator of thyroid function. Using rat thyroid FRTL-5 cells, we showed that the mRNA levels of Slc26a7 and Slc26a4, two apical iodine transporters responsible for iodine efflux, were suppressed by TSH, whereas the mRNA level of Slc5a5 was induced. Forskolin and dibutyryl cAMP (dbcAMP) had the same effect as that of TSH on the mRNA levels of these transporters. TSH, forskolin and dbcAMP also had suppressive effects on SLC26A7 promoter activity, as assessed by luciferase reporter gene assays, and protein levels, as determined by Western blot analysis. TSH, forskolin and dbcAMP also induced strong localization of Slc26a7 to the cell membrane according to immunofluorescence staining and confocal laser scanning microscopy. Together, these results suggest that TSH suppresses the expression level of Slc26a7 but induces its accumulation at the cell membrane, where it functions as an iodine transporter.
    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
    The sodium iodide symporter (NIS) is widely used as a reporter gene to noninvasively monitor the biodistribution and durability of vector-mediated gene expression via gamma scintigraphy, single-photon emission computed tomography (SPECT), and positron-emission tomography (PET). However, the approach is limited by background signal due to radiotracer uptake by endogenous NIS-expressing tissues. In this study, using the SPECT tracer pertechnetate (99mTcO4) and the PET tracer tetrafluoroborate (B18F4), in combination with the NIS inhibitor perchlorate, we compared the transport properties of human NIS and minke whale (Balaenoptera acutorostrata scammoni) NIS in vitro and in vivo. Based on its relative resistance to perchlorate, the NIS protein from minke whale appeared to be the superior candidate reporter gene. SPECT and PET imaging studies in nude mice challenged with NIS-encoding adeno-associated virus (AAV)-9 vectors confirmed that minke whale NIS, in contrast to human and endogenous mouse NIS, continues to function as a reliable reporter even when background radiotracer uptake by endogenous NIS is blocked by perchlorate.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Chasing an intriguing biological question on the disparity of sodium iodide symporter (NIS, officially known as SLC5A5) expression and function in the clinical scenario of breast cancer, this study addresses key molecular defects involved. NIS in cancer patients has primarily been recorded to be a cytoplasmic protein, thus limiting the scope for targeted radio-iodine therapy. We developed NIS transgene-overexpressing MCF-7 breast cancer cells, and found a few clonal derivatives that show predominant expression of NIS in the plasma membrane. The majority of clones, however, showed cytosolic NIS expression over long passages. Cells expressing membranous NIS show unperturbed dynamic trafficking of NIS through secretory pathway organelles when compared to cells expressing cytoplasmic NIS or to parental cells. Further, treatment of cells expressing membranous NIS with specific glycosylation inhibitors highlighted the importance of inherent glycosylation processing and an 84 gene signature glycosylation RT-Profiler array revealed that clones expressing NIS in their membrane cluster separately compared to the other cells. We further confirm a role of three differentially expressed genes, i.e. MAN1B1, MAN1A1 and MAN2A1, in regulating NIS localization by RNA interference. Thus, this study shows the important role of mannosidase in N-glycosylation processing in order to correctly traffic NIS to the plasma membrane in breast cancer cells.This article has an associated First Person interview with the first author of the paper.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号