Large Neutral Amino Acid-Transporter 1

大型中性氨基酸转运蛋白 1
  • 文章类型: Journal Article
    系统性硬化症(SSc)是一种慢性自身免疫性结缔组织疾病。血管损害是SSc的重要特点之一,影响疾病的进展和预后。MiR-126-3p是调节血管结构和功能的重要microRNA(miRNA),可以通过外泌体运输。然而,miR-126-3p在SSc血管损伤中的作用尚不清楚.因此,我们专注于miR-126-3p与SSc血管损伤之间的联系,以及研究miR-126-3p在SSc血管损伤中的潜在作用。首先,这项研究成功地从临床血浆样本中提取了细胞外囊泡,并对其中的外泌体进行了表征。然后,我们通过在线数据库预测并筛选了哺乳动物/机制雷帕霉素靶蛋白(mTOR)的靶途径和miR-126-3p的靶基因SLC7A5。接下来,我们构建了SSc小鼠用于体内研究。结果显示血浆外泌体中miR-126-3p的表达降低,而SLC7A5表达式,自噬,和脂质过氧化在主动脉中增加。荧光素酶报告基因测定表明miR-126-3p可以与SLC7A5结合,导致其表达降低。体外实验表明,外泌体miR-126-3p可以被人脐静脉内皮细胞(HUVEC)内化。miR-126-3p组表现出增强的细胞活力和试管形成能力,随着血管形成标志物CD31的表达增加。此外,miR-126-3p下调HUVECs中SLC7A5和LC3的蛋白表达,在上调mTOR蛋白表达的同时,P62,PPARγ,和CPT-1。然而,miR-126-3p对HUVECs的作用被mTOR抑制剂抵消,被mTOR激活剂增强.结果表明,外泌体miR-126-3p有可能通过调节HUVECs中的SLC7A5/mTOR信号通路来保护SSc免受血管损伤。
    Systemic sclerosis (SSc) is a chronic autoimmune connective tissue disease. Vascular damage is one of the important features of SSc, which affects the progression and prognosis of the disease. MiR-126-3p is an important microRNA (miRNA) that regulates vascular structure and function, which can be transported through exosomes. However, the role of miR-126-3p in vascular damage in SSc is still unclear. Therefore, we focused on the connection between miR-126-3p and vascular damage in SSc, as well as investigated the potential role of miR-126-3p in vascular damage in SSc. First, this study successfully extracted extracellular vesicles from clinical plasma samples and characterized the exosomes within them. Then, we predicted and screened the target pathway mammalian/mechanistic target of rapamycin (mTOR) and the target gene SLC7A5 of miR-126-3p through online databases. Next, we constructed SSc mice for in vivo studies. The results showed that the expression of miR-126-3p was decreased in the plasma exosomes, while the SLC7A5 expression, autophagy, and lipid peroxidation were increased in the aorta. Luciferase reporter gene assays demonstrated that miR-126-3p can bind to SLC7A5, resulting in a decrease in its expression. In vitro experiments have shown that exosomal miR-126-3p can be internalized by human umbilical vein endothelial cells (HUVECs). The miR-126-3p group exhibited enhanced cell viability and tube formation ability, along with increased expression of the vascular formation marker CD31. Additionally, miR-126-3p downregulated the protein expression of SLC7A5 and LC3 in HUVECs, while upregulating the protein expression of mTOR, P62, PPARγ, and CPT-1. However, the effects of miR-126-3p on HUVECs were counteracted by mTOR inhibitors and enhanced by mTOR activators. The results indicated that exosomal miR-126-3p has the potential to protect against vascular injury in SSc by regulating the SLC7A5/mTOR signalling pathway in HUVECs.
    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
    侵袭性自然杀伤细胞白血病(ANKL)是一种罕见的血液恶性肿瘤,具有暴发性临床病程。我们先前的研究表明,ANKL细胞主要在肝窦中增殖,并且强烈依赖于转铁蛋白的补充。此外,我们证明了肝脏驻留的ANKL细胞对PPMX-T003敏感,PPMX-T003是一种抗人转铁蛋白受体1抑制性抗体,而脾脏驻留的ANKL细胞对转铁蛋白受体1抑制具有抗性。然而,调节ANKL细胞铁依赖性的微环境因素尚不清楚.在这项研究中,我们首先揭示了PPMX-T003的抗肿瘤作用以DNA复制依赖性方式的DNA双链断裂为特征,类似于传统的细胞毒性剂。我们还发现,通过LAT1的细胞外氨基酸流入刺激了对PPMX-T003的敏感性。一起来看,我们发现通过LAT1的细胞外氨基酸流入量是通过调节其mTOR/Myc活性决定ANKL细胞铁依赖性的关键环境因素,这为肝脏和脾脏驻留的ANKL细胞对PPMX-T003的不同敏感性提供了很好的解释,因为肝窦含有从肠道吸收的丰富氨基酸。
    Aggressive natural killer cell leukemia (ANKL) is a rare hematological malignancy with a fulminant clinical course. Our previous study revealed that ANKL cells proliferate predominantly in the liver sinusoids and strongly depend on transferrin supplementation. In addition, we demonstrated that liver-resident ANKL cells are sensitive to PPMX-T003, an anti-human transferrin receptor 1 inhibitory antibody, whereas spleen-resident ANKL cells are resistant to transferrin receptor 1 inhibition. However, the microenvironmental factors that regulate the iron dependency of ANKL cells remain unclear. In this study, we first revealed that the anti-neoplastic effect of PPMX-T003 was characterized by DNA double-strand breaks in a DNA replication-dependent manner, similar to conventional cytotoxic agents. We also found that the influx of extracellular amino acids via LAT1 stimulated sensitivity to PPMX-T003. Taken together, we discovered that the amount of extracellular amino acid influx through LAT1 was the key environmental factor determining the iron dependency of ANKL cells via adjustment of their mTOR/Myc activity, which provides a good explanation for the different sensitivity to PPMX-T003 between liver- and spleen-resident ANKL cells, as the liver sinusoid contains abundant amino acids absorbed from the gut.
    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
    上呼吸道鳞状细胞癌(UASCC)是一种常见的侵袭性恶性肿瘤,几乎没有有效的治疗选择。这里,我们研究了这种癌症的氨基酸代谢,令人惊讶的是,UASCC在所有人类癌症中表现出最高的蛋氨酸水平,由其运输车LAT1驱动。我们表明LAT1在UASCC中也以最高水平表达,由UASCC特异性启动子和增强子转录激活,它们由SCC主调节剂TP63/KLF5/SREBF1直接共同调节。出乎意料的是,无偏生物信息学筛选将EZH2鉴定为LAT1-蛋氨酸途径下游的最重要靶标,直接将蛋氨酸代谢与表观基因组重编程联系起来。重要的是,这种级联反应对于UASCC患者来源的肿瘤类器官的存活和增殖是必不可少的。此外,LAT1表达与细胞对LAT1-甲硫氨酸-EZH2轴抑制的敏感性密切相关。值得注意的是,这种独特的LAT1-蛋氨酸-EZH2级联反应可以通过体内药理学方法或饮食干预有效靶向。总之,这项工作映射了表观基因组重编程与蛋氨酸代谢之间的独特机制串扰,确立了其在UASCC生物学中的生物学意义,并确定了一种独特的肿瘤特异性脆弱性,可以在药理和饮食上利用。
    Upper aerodigestive squamous cell carcinoma (UASCC) is a common and aggressive malignancy with few effective therapeutic options. Here, we investigate amino acid metabolism in this cancer, surprisingly noting that UASCC exhibits the highest methionine level across all human cancers, driven by its transporter LAT1. We show that LAT1 is also expressed at the highest level in UASCC, transcriptionally activated by UASCC-specific promoter and enhancers, which are directly coregulated by SCC master regulators TP63/KLF5/SREBF1. Unexpectedly, unbiased bioinformatic screen identifies EZH2 as the most significant target downstream of the LAT1-methionine pathway, directly linking methionine metabolism to epigenomic reprogramming. Importantly, this cascade is indispensable for the survival and proliferation of UASCC patient-derived tumor organoids. In addition, LAT1 expression is closely associated with cellular sensitivity to inhibition of the LAT1-methionine-EZH2 axis. Notably, this unique LAT1-methionine-EZH2 cascade can be targeted effectively by either pharmacological approaches or dietary intervention in vivo. In summary, this work maps a unique mechanistic cross talk between epigenomic reprogramming with methionine metabolism, establishes its biological significance in the biology of UASCC, and identifies a unique tumor-specific vulnerability which can be exploited both pharmacologically and dietarily.
    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
    乳腺癌是世界范围内最常见的恶性肿瘤之一。SLC7A2在多种癌症中异常表达。然而,其在三阴性乳腺癌(TNBC)中的潜力仍不清楚。目的探讨SLC7A2在TNBC中的作用及其分子机制。通过RT-qPCR检测mRNA的表达。蛋白质印迹法检测蛋白质表达。使用FISH测定法测定ACOX1和TCF1的共定位。使用体外组蛋白巴豆化测定进行组蛋白巴豆化。使用CCK-8和流式细胞术测定进行功能分析。进行异种移植物测定以进一步验证SLC7A2在TNBC中的作用。使用免疫组织化学检测CD8A表达。我们发现SLC7A2在TNBC肿瘤中下调。低水平与晚期分期和淋巴结转移有关。SLC7A2表达与CD8A呈正相关。SLC7A2介导的赖氨酸分解代谢驱动CD8+T细胞的活化。此外,SLC7A2通过上调ACOX1促进组蛋白巴豆化。它还促进ACOX1和TCF1之间的相互作用,从而促进抗肿瘤T细胞免疫。此外,SLC7A2的过表达激活CD8+T细胞并增强体内抗PD-1疗法的化学敏感性。总之,SLC7A2可能通过激活抗肿瘤免疫而在TNBC中发挥抗肿瘤基因的作用,提示SLC7A2/ACOX1/TCF1信号传导是一种有前途的治疗策略。
    Breast cancer is one of the most common malignant tumors worldwide. SLC7A2 is abnormally expressed in multiple cancers. However, its potential in triple negative breast cancer (TNBC) is still unclear. The purpose of this study was to investigate the roles of SLC7A2 and its underlying molecular mechanisms in TNBC. mRNA expression was detected by RT-qPCR. Protein expression was detected by western blot. Co-localization of ACOX1 and TCF1 was determined using FISH assay. Histone crotonylation was performed using in vitro histone crotonylation assay. Functional analysis was performed using CCK-8 and flow cytometry assays. Xenograft assay was conducted to further verify the role of SLC7A2 in TNBC. CD8A expression was detected using immunohistochemistry. We found that SLC7A2 is downregulated in TNBC tumors. Low levels are associated with advanced stages and lymph node metastasis. SLC7A2 expression is positively correlated with CD8A. SLC7A2-mediated lysine catabolism drives the activation of CD8+ T cells. Moreover, SLC7A2 promotes histone crotonylation via upregulating ACOX1. It also promotes interaction between ACOX1 and TCF1, thus promoting antitumor T cell immunity. Additionally, overexpression of SLC7A2 activates CD8+ T cells and enhances the chemosensitivity of anti-PD-1 therapies in vivo. In conclusion, SLC7A2 may function as an antitumor gene in TNBC by activating antitumor immunity, suggesting SLC7A2/ACOX1/TCF1 signaling as a promising therapeutic strategy.
    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
    血管生成,从预先存在的脉管系统中生长新血管,对新器官系统的发展至关重要,但是血管生成的转录控制仍未完全了解。在这里,我们表明FOXC1对于视网膜血管生成是必不可少的。Foxc1的内皮细胞(EC)特异性丢失会损害视网膜血管的生长以及Slc3a2和Slc7a5的表达,Slc3a2和Slc7a5编码异二聚体CD98(LAT1/4F2hc)氨基酸转运蛋白,并调节必需氨基酸的细胞内转运和哺乳动物靶蛋白的激活雷帕霉素(mTOR)。EC-Foxc1缺乏会降低mTOR活性,而mTOR激动剂MHY-1485的给药挽救了受干扰的视网膜血管生成。在氧诱导的视网膜病变模型中,EC-Foxc1表达是视网膜血运重建和新生血管簇消退所必需的。Foxc1对于周细胞也是不可或缺的,视网膜血管生成过程中血-视网膜屏障的关键组成部分。我们的发现确立了FOXC1作为视网膜血管的关键调节因子,并确定了治疗视网膜血管疾病的治疗靶点。
    Angiogenesis, the growth of new blood vessels from pre-existing vasculature, is essential for the development of new organ systems, but transcriptional control of angiogenesis remains incompletely understood. Here we show that FOXC1 is essential for retinal angiogenesis. Endothelial cell (EC)-specific loss of Foxc1 impairs retinal vascular growth and expression of Slc3a2 and Slc7a5, which encode the heterodimeric CD98 (LAT1/4F2hc) amino acid transporter and regulate the intracellular transport of essential amino acids and activation of the mammalian target of rapamycin (mTOR). EC-Foxc1 deficiency diminishes mTOR activity, while administration of the mTOR agonist MHY-1485 rescues perturbed retinal angiogenesis. EC-Foxc1 expression is required for retinal revascularization and resolution of neovascular tufts in a model of oxygen-induced retinopathy. Foxc1 is also indispensable for pericytes, a critical component of the blood-retina barrier during retinal angiogenesis. Our findings establish FOXC1 as a crucial regulator of retinal vessels and identify therapeutic targets for treating retinal vascular disease.
    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
    我们以前的研究表明,SLC7A1在上皮性卵巢癌(EOC)肿瘤细胞中的上调显着增加癌细胞的增殖,迁移,和顺铂耐药;然而,SLC7A1在EOC中起作用的分子机制尚不清楚。在后来的研究中,我们发现SLC7A1在高级别浆液性卵巢癌(HGSOC)的间质部分也高表达,但是在间质中这种高表达的意义尚不清楚。这里,我们通过免疫组织化学显示SLC7A1在HGSOC中的间质高表达。富含癌相关成纤维细胞(CAF)的SLC7A1被TGF-β1上调。Transwell分析,划痕试验,cck8实验和细胞粘附实验显示SLC7A1在CAFs中高表达促进肿瘤细胞侵袭,体外迁移和转移。通过RNA测序和蛋白质印迹法验证了SLC7A1对卵巢癌(OC)中MAPK和EMT通路蛋白的影响。SLC7A1在OC中的过表达与MAPK/ERK通路和EMT有关。总的来说,在HGSOC,过表达SLC7A1的CAFs支持肿瘤细胞的迁移和侵袭;SLC7A1在卵巢癌中高表达,并参与MAPK信号通路中的ERK磷酸化和EMT信号传导。这表明SLC7A1可能是OC转移的潜在治疗靶标。
    Our previous studies have shown that upregulation of SLC7A1 in epithelial ovarian cancer (EOC) tumor cells significantly increases cancer cell proliferation, migration, and cisplatin resistance; however, the molecular mechanism by which SLC7A1 functions in EOC remains unknown. In later studies, we found that SLC7A1 is also highly expressed in the interstitial portion of high-grade serous ovarian cancer (HGSOC), but the significance of this high expression in the interstitial remains unclear. Here, we showed the Interstitial high expression of SLC7A1 in HGSOC by immunohistochemistry. SLC7A1 enriched in cancer-associated fibroblasts (CAFs) was upregulated by TGF-β1. Transwell assay, scratch assay, cck8 assay and cell adhesion assay showed that SLC7A1 highly expressed in CAFs promoted tumor cells invasion, migration and metastasis in vitro. The effect of SLC7A1 on MAPK and EMT pathway proteins in ovarian cancer (OC) was verified by RNA sequencing and western blotting. Overexpression of SLC7A1 in OC is involved in MAPK/ ERK pathway and EMT. In general, in HGSOC, CAFs overexpressing SLC7A1 supported the migration and invasion of tumor cells; SLC7A1 is highly expressed in ovarian cancer and is involved in ERK phosphorylation and EMT signaling in MAPK signaling pathway. This suggests that SLC7A1 may be a potential therapeutic target for OC metastasis.
    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
    O-([18F]氟乙基)-1-酪氨酸([18F]FET)通过LAT1和可能的其他氨基酸转运蛋白被主动转运到大脑和癌细胞中。通过正电子发射断层扫描(PET)实现脑肿瘤成像。然而,在存在竞争性氨基酸的情况下,该探针的肿瘤递送可能受到对LAT1的相对低亲和力的限制。本工作的目的是评估元取代的[18F]FET模拟m-[18F]FET和甲酯[18F]FET-OMe,它们旨在通过改变物理化学来改善肿瘤的输送,药代动力学,和/或运输属性。两种示踪剂均可在66-90分钟内以41-56%的良好放射化学产率制备。以[18F]FET作为参考示踪剂的临床前评估表明,U87胶质母细胞瘤细胞对[18F]FET-OMe的体外摄取减少,对体内肿瘤成像没有优势。相比之下,m-[18F]FET在原位成胶质细胞瘤模型中显示出显著改善的体外摄取和加速的体内肿瘤积累。因此,我们的工作确定m-[18F]FET是[18F]FET的有希望的替代脑肿瘤成像,值得就其运输特性和体内生物分布进行进一步评估。
    O-([18F]Fluoroethyl)-l-tyrosine ([18F]FET) is actively transported into the brain and cancer cells by LAT1 and possibly other amino acid transporters, which enables brain tumor imaging by positron emission tomography (PET). However, tumor delivery of this probe in the presence of competing amino acids may be limited by a relatively low affinity for LAT1. The aim of the present work was to evaluate the meta-substituted [18F]FET analog m-[18F]FET and the methyl ester [18F]FET-OMe, which were designed to improve tumor delivery by altering the physicochemical, pharmacokinetic, and/or transport properties. Both tracers could be prepared with good radiochemical yields of 41-56% within 66-90 min. Preclinical evaluation with [18F]FET as a reference tracer demonstrated reduced in vitro uptake of [18F]FET-OMe by U87 glioblastoma cells and no advantage for in vivo tumor imaging. In contrast, m-[18F]FET showed significantly improved in vitro uptake and accelerated in vivo tumor accumulation in an orthotopic glioblastoma model. As such, our work identifies m-[18F]FET as a promising alternative to [18F]FET for brain tumor imaging that deserves further evaluation with regard to its transport properties and in vivo biodistribution.
    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
    在早期癌症中过度表达的蛋白质可用作早期诊断和预后标志物以及癌症治疗的靶标。在这项研究中,我们检测了必需氨基酸载体SLC7A5(LAT1,CD98或4F2轻链)在两个注释良好的早期结直肠癌患者和106例晚期结直肠癌患者的癌组织中的表达.免疫组织化学显示SLC7A5在早期病例的72.0%和晚期病例的56.6%中过度表达。SLC7A5表达不受患者性别影响,年龄,location,或不匹配修复状态,尽管在粘液性分化或淋巴血管浸润的肿瘤中似乎不太普遍。统计分析显示SLC7A5过表达与早期癌症而非晚期癌症的总生存期和无病生存期之间呈正相关。TCGA和CPTAC结直肠癌队列的共表达分析确定了与SLC7A5正相关的基因转录物网络,其异二聚体伴侣SLC3A2具有最高的共表达得分。网络分析发现SLC7A网络与ncRNA如tRNA加工和有丝分裂细胞周期显著相关。由于SLC7A5也是NK等活化淋巴细胞的标志物,T,B淋巴细胞,SLC7A5在早期结直肠癌中的过表达可能会引发强烈的抗肿瘤免疫反应,从而导致更好的临床结果。总的来说,我们的研究提供了SLC7A5表达差异及其对早期结直肠癌预后价值的明确证据,尽管目前对其在结直肠肿瘤发生和癌症免疫中的功能的理解相当有限,有待进一步表征。
    Proteins overexpressed in early-stage cancers may serve as early diagnosis and prognosis markers as well as targets for cancer therapies. In this study, we examined the expression of an essential amino acid carrier SLC7A5 (LAT1, CD98, or 4F2 light chain) in cancer tissue from two well-annotated cohorts of 575 cases of early-stage and 106 cases of late-stage colorectal cancer patients. Immunohistochemistry showed SLC7A5 overexpression in 72.0% of early-stage and 56.6% of late-stage cases. SLC7A5 expression was not influenced by patient gender, age, location, or mismatch repair status, although it appeared to be slightly less prevalent in tumors of mucinous differentiation or with lymphovascular invasion. Statistical analyses revealed a positive correlation between SLC7A5 overexpression and both overall survival and disease-free survival in early-stage but not late-stage cancers. Co-expression analyses of the TCGA and CPTAC colorectal cancer cohorts identified a network of gene transcripts positively related to SLC7A5, with its heterodimer partner SLC3A2 having the highest co-expression score. Network analysis uncovered the SLC7A network to be significantly associated with ncRNA such as tRNA processing and the mitotic cell cycle. Since SLC7A5 is also a marker of activated lymphocytes such as NK, T, and B lymphocytes, SLC7A5 overexpression in early colorectal cancers might trigger a strong anti-tumor immune response which could results in better clinical outcome. Overall, our study provides clear evidence of differential SLC7A5 expression and its prognostic value for early-stage colorectal cancer, although the understanding of its functions in colorectal tumorigenesis and cancer immunity is currently rather limited and awaits further characterization.
    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
    LAT1-4F2hc复合物(SLC7A5-SLC3A2)促进必需氨基酸的摄取,激素和药物。其功能障碍与许多癌症和免疫/神经障碍有关。这里,我们应用基于天然质谱(MS)的方法来提供超二聚体形成的证据(LAT1-4F2hc)2。当与脂质组学结合时,和定点诱变,我们在两个LAT1亚基的界面和C末端发现了四个内源性磷脂酰乙醇胺(PE)分子。我们发现,界面PE结合受4F2hc-R183调节,对于调节相邻LAT1-C187的棕榈酰化至关重要。将天然MS与质量测光(MP)相结合,我们发现超级二聚化对pH敏感,并由4F2hc亚基上的复合N-聚糖调节。我们进一步验证了LAT1-4F2hc在质膜和溶酶体上的动态组装。我们的结果一起将PTM和脂质结合与LAT1-4F2hc超二聚体的调节和定位联系起来。
    The LAT1-4F2hc complex (SLC7A5-SLC3A2) facilitates uptake of essential amino acids, hormones and drugs. Its dysfunction is associated with many cancers and immune/neurological disorders. Here, we apply native mass spectrometry (MS)-based approaches to provide evidence of super-dimer formation (LAT1-4F2hc)2. When combined with lipidomics, and site-directed mutagenesis, we discover four endogenous phosphatidylethanolamine (PE) molecules at the interface and C-terminus of both LAT1 subunits. We find that interfacial PE binding is regulated by 4F2hc-R183 and is critical for regulation of palmitoylation on neighbouring LAT1-C187. Combining native MS with mass photometry (MP), we reveal that super-dimerization is sensitive to pH, and modulated by complex N-glycans on the 4F2hc subunit. We further validate the dynamic assemblies of LAT1-4F2hc on plasma membrane and in the lysosome. Together our results link PTM and lipid binding with regulation and localisation of the LAT1-4F2hc super-dimer.
    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
    甲基汞是一种环境污染的有机金属化合物,具有神经毒性,如在水峰病患者中观察到的。甲基汞损害水晶病人的周围神经,对感觉神经造成的损害比运动神经更大。周围神经由三种细胞类型组成:背根神经节(DRG)细胞,前角细胞(AHC),和施万细胞。在这项研究中,我们比较了培养的这三种来自大鼠的细胞类型对甲基汞细胞毒性的敏感性,细胞内汞的积累,L型氨基酸转运蛋白1(LAT1)的表达,将甲基汞输送到细胞中,多药耐药相关蛋白2(MRP2)的表达,它将甲基汞-谷胱甘肽缀合物输送到细胞外空间。在检查的细胞中,我们发现DRG细胞对甲基汞最敏感,细胞内汞积累明显较高。与AHC和雪旺氏细胞相比,DRG细胞中LAT1的组成水平较高,MRP2的组成水平较低。此外,由甲基汞引起的细胞生存力下降被LAT1抑制剂显著降低,JPH203,或siRNA介导的LAT1敲低。另一方面,MRP2抑制剂,MK571显着加剧了甲基汞引起的细胞活力降低。我们的结果为水晶病患者周围神经的感觉神经主要损伤提供了细胞基础。
    Methylmercury is an environmental polluting organometallic compound that exhibits neurotoxicity, as observed in Minamata disease patients. Methylmercury damages peripheral nerves in Minamata patients, causing more damage to sensory nerves than motor nerves. Peripheral nerves are composed of three cell types: dorsal root ganglion (DRG) cells, anterior horn cells (AHCs), and Schwann cells. In this study, we compared cultured these three cell types derived from the rat for susceptibility to methylmercury cytotoxicity, intracellular accumulation of mercury, expression of L-type amino acid transporter 1 (LAT1), which transports methylmercury into cells, and expression of multidrug resistance-associated protein 2 (MRP2), which transports methylmercury-glutathione conjugates into the extracellular space. Of the cells examined, we found that DRG cells were the most susceptible to methylmercury with markedly higher intracellular accumulation of mercury. The constitutive level of LAT1 was higher and that of MRP2 lower in DRG cells compared with those in AHC and Schwann cells. Additionally, decreased cell viability caused by methylmercury was significantly reduced by either the LAT1 inhibitor, JPH203, or siRNA-mediated knockdown of LAT1. On the other hand, an MRP2 inhibitor, MK571, significantly intensified the decrease in the cell viability caused by methylmercury. Our results provide a cellular basis for sensory neve predominant injury in the peripheral nerves of Minamata disease patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号