NPRL2

NPRL2
  • 文章类型: Journal Article
    已知氮通透酶调节因子样2(NPRL2/TUSC4)在不同类型的癌症中发挥肿瘤抑制和致癌作用。这表明它的行为是依赖于上下文的。这里,我们描述了NPRL2在恶性转化的支气管上皮细胞中的分子和功能作用。要做到这一点,我们在致癌HRas转导和恶性转化的人支气管上皮(BEAS2B)中耗尽NPRL2,Ras-AI-T2细胞。有趣的是,这些细胞中NPRL2的耗尽诱导mTORC1下游信号的激活,抑制自噬,以及体内外Ras-AI-T2细胞增殖受损。这些结果表明NPRL2是致癌HRas诱导的细胞转化所必需的。NPRL2的消耗增加了DNA损伤标记γH2AX的水平,细胞周期抑制剂p21和p27,以及凋亡标志物裂解-PARP。这些NPRL2耗尽的细胞首先在G1和G2积累,后来表现出有丝分裂突变的迹象,这意味着NPRL2耗竭可能对致癌HRas转化细胞有害。此外,NPRL2耗竭降低了Ras-AI-T2细胞中热休克因子1/热休克元素和NRF2/抗氧化剂反应元件指导的荧光素酶报告活性,表明NPRL2耗竭导致致癌HRas转化细胞中两个关键细胞保护过程的抑制。总的来说,我们的数据表明,致癌HRas转导和恶性转化的细胞可能依赖于NPRL2的存活和增殖,和NPRL2的耗尽也诱导这些细胞中的应激状态。
    Nitrogen permease regulator-like 2 (NPRL2/TUSC4) is known to exert both tumor-suppressing and oncogenic effects in different types of cancers, suggesting that its actions are context dependent. Here, we delineated the molecular and functional effects of NPRL2 in malignantly transformed bronchial epithelial cells. To do so, we depleted NPRL2 in oncogenic HRas-transduced and malignantly transformed human bronchial epithelial (BEAS2B), Ras-AI-T2 cells. Intriguingly, depletion of NPRL2 in these cells induced activation of mTORC1 downstream signaling, inhibited autophagy, and impaired Ras-AI-T2 cell proliferation both in vitro and in vivo. These results suggest that NPRL2 is required for oncogenic HRas-induced cell transformation. Depletion of NPRL2 increased levels of the DNA damage marker γH2AX, the cell cycle inhibitors p21 and p27, and the apoptosis marker cleaved-PARP. These NPRL2-depleted cells first accumulated at G1 and G2, and later exhibited signs of mitotic catastrophe, which implied that NPRL2 depletion may be detrimental to oncogenic HRas-transformed cells. Additionally, NPRL2 depletion reduced heat shock factor 1/heat shock element- and NRF2/antioxidant response element-directed luciferase reporter activities in Ras-AI-T2 cells, indicating that NPRL2 depletion led to the suppression of two key cytoprotective processes in oncogenic HRas-transformed cells. Overall, our data suggest that oncogenic HRas-transduced and malignantly transformed cells may depend on NPRL2 for survival and proliferation, and depletion of NPRL2 also induces a stressed state in these cells.
    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
    GATOR1(GAP活性TOwardRag1)是一种进化保守的GTP酶激活蛋白复合物,可控制mTORC1(哺乳动物雷帕霉素靶标复合物1)对细胞中氨基酸可用性的反应。GATOR1亚基的基因突变,NPRL2(氮通透酶调节因子样2),NPRL3(氮通透酶调节因子样3),和DEPDC5(含有5个DEP域),与人类癫痫有关;然而,这些突变对GATOR1功能和mTORC1调控的具体影响尚不清楚.在这里,我们报道了GATOR1的NPRL2亚基,NPRL2-L105P,-T110S,和-D214H,增加细胞中基础mTORC1信号转导。值得注意的是,我们发现NPRL2-L105P是一种功能缺失突变,它破坏了蛋白质与NPRL3和DEPDC5的相互作用,损害了GATOR1复合物的组装,甚至在氨基酸剥夺条件下也能产生高mTORC1活性.此外,我们的研究表明,GATOR1复合物对于mTORC1的快速和强大抑制是必需的,以响应生长因子的戒断或磷脂酰肌醇-3激酶(PI3K)的药理抑制。在没有GATOR1复合体的情况下,细胞难以抑制PI3K依赖性的mTORC1抑制,允许持续翻译和限制TFEB的核定位,由mTORC1调节的转录因子。总的来说,我们的结果表明,NPRL2中的癫痫相关突变可以阻断GATOR1复合物的组装,并在存在或不存在氨基酸的情况下,限制典型PI3K依赖性生长因子信号对mTORC1的适当调节.
    GATOR1 (GAP Activity TOward Rag 1) is an evolutionarily conserved GTPase-activating protein complex that controls the activity of mTORC1 (mammalian Target Of Rapamycin Complex 1) in response to amino acid availability in cells. Genetic mutations in the GATOR1 subunits, NPRL2 (nitrogen permease regulator-like 2), NPRL3 (nitrogen permease regulator-like 3), and DEPDC5 (DEP domain containing 5), have been associated with epilepsy in humans; however, the specific effects of these mutations on GATOR1 function and mTORC1 regulation are not well understood. Herein, we report that epilepsy-linked mutations in the NPRL2 subunit of GATOR1, NPRL2-L105P, -T110S, and -D214H, increase basal mTORC1 signal transduction in cells. Notably, we show that NPRL2-L105P is a loss-of-function mutation that disrupts protein interactions with NPRL3 and DEPDC5, impairing GATOR1 complex assembly and resulting in high mTORC1 activity even under conditions of amino acid deprivation. Furthermore, our studies reveal that the GATOR1 complex is necessary for the rapid and robust inhibition of mTORC1 in response to growth factor withdrawal or pharmacological inhibition of phosphatidylinositol-3 kinase (PI3K). In the absence of the GATOR1 complex, cells are refractory to PI3K-dependent inhibition of mTORC1, permitting sustained translation and restricting the nuclear localization of TFEB, a transcription factor regulated by mTORC1. Collectively, our results show that epilepsy-linked mutations in NPRL2 can block GATOR1 complex assembly and restrict the appropriate regulation of mTORC1 by canonical PI3K-dependent growth factor signaling in the presence or absence of amino acids.
    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
    蛋氨酸是决定mTORC1活化的多种营养素输入的重要分支。在没有蛋氨酸的情况下,SAMTOR结合GATOR1并抑制mTORC1信号传导。然而,mTORC1在蛋氨酸刺激下是如何被激活的仍然是很大程度上难以捉摸的。这里,我们报道,PRMT1通过利用SAM作为基于酶活性调节mTORC1信号传导的辅因子来感知甲硫氨酸/SAM.在蛋氨酸充足的条件下,胞质升高的SAM从GATOR1释放SAMT1,这赋予PRMT1与GATOR1的关联。随后,SAM负载的PRMT1使GATOR1的催化亚基NPRL2甲基化,从而抑制其GAP活性并导致mTORC1活化。值得注意的是,PRMT1的遗传或药理学抑制阻碍mTORC1的肝蛋氨酸传感,并改善老年小鼠的胰岛素敏感性,建立PRMT1介导的蛋氨酸传感在生理水平上的作用。因此,PRMT1与SAMTOR协调以形成mTORC1信号传导的甲硫氨酸传感装置。
    Methionine is an essential branch of diverse nutrient inputs that dictate mTORC1 activation. In the absence of methionine, SAMTOR binds to GATOR1 and inhibits mTORC1 signaling. However, how mTORC1 is activated upon methionine stimulation remains largely elusive. Here, we report that PRMT1 senses methionine/SAM by utilizing SAM as a cofactor for an enzymatic activity-based regulation of mTORC1 signaling. Under methionine-sufficient conditions, elevated cytosolic SAM releases SAMTOR from GATOR1, which confers the association of PRMT1 with GATOR1. Subsequently, SAM-loaded PRMT1 methylates NPRL2, the catalytic subunit of GATOR1, thereby suppressing its GAP activity and leading to mTORC1 activation. Notably, genetic or pharmacological inhibition of PRMT1 impedes hepatic methionine sensing by mTORC1 and improves insulin sensitivity in aged mice, establishing the role of PRMT1-mediated methionine sensing at physiological levels. Thus, PRMT1 coordinates with SAMTOR to form the methionine-sensing apparatus of mTORC1 signaling.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    背景:NPRL2相关癫痫,由NPRL2基因的致病性种系变异引起,是一种新发现的儿童癫痫,与mTORC1信号增强有关。然而,NPRL2变体的表型和基因型仍然知之甚少.这里,我们总结了NPRL2相关癫痫的表型和基因型之间的关联.
    方法:对4例中国儿童癫痫患者进行回顾性分析,原因是通过全外显子组测序(WES)鉴定出可能的致病性NPRL2变异。系统回顾了以前关于NPRL2相关癫痫患者的报道。
    结果:我们的一位患者出现局灶性癫痫,累及中央区,应与具有中央颞部尖峰的自限性癫痫(SeLECTS)区分开。四个新的可能致病性NPRL2变体由两个无义变体组成,一个移码变体,和一个拷贝数变体(CNV)。生物信息学分析显示,这两种无义变体高度保守,并导致蛋白质结构发生变化。包括我们的四个案例,迄今为止,共发现33例NPRL2相关癫痫患者.最常见的表现是局灶性癫痫(70%),包括睡眠相关的运动过度癫痫(SHE),颞叶癫痫(TLE),额叶癫痫(FLE)。婴儿癫痫性痉挛综合征(IESS)也是NPRL2相关癫痫的显着特征。皮质发育畸形(MCD,8/20),尤其是局灶性皮质发育不良(FCD,6/20),是常见的神经影像学异常。报告的NPRL2变体的三分之二是功能丧失(LoF)(14/21)。在这些突变中,c.100C>T(p。Arg34*)和c.314T>C(p。Leu105Pro)已在两个家庭中被检测到(可能是由于创始人的影响)。
    结论:NPRL2相关癫痫表现出高度的表型和基因型异质性。我们的研究扩展了NPRL2相关癫痫的基因型谱,涉及中央区的局灶性癫痫的表型应与SeLECTS明确区分,对临床诊断具有参考价值。
    BACKGROUND: NPRL2-related epilepsy, caused by pathogenic germline variants of the NPRL2 gene, is a newly discovered childhood epilepsy linked to enhanced mTORC1 signalling. However, the phenotype and genotype of NPRL2 variants are still poorly understood. Here, we summarize the association between the phenotype and genotype of NPRL2-related epilepsy.
    METHODS: A retrospective analysis was conducted for four Chinese children with epilepsy due to likely pathogenic NPRL2 variants identified through whole-exome sequencing (WES). Previous reports of patients with NPRL2-related epilepsy were reviewed systematically.
    RESULTS: One of our patients presented focal epilepsy involving the central region, which should be distinguished from self-limited epilepsy with centrotemporal spikes (SeLECTS). The four novel likely pathogenic NPRL2 variants consisted of two nonsense variants, one frameshift variant, and one copy number variant (CNV). Bioinformatics analysis revealed the two nonsense variants to be highly conserved and cause alterations in protein structure. Including our four cases, a total of 33 patients with NPRL2-related epilepsy have been identified to date. The most common presentation is focal epilepsy (70%), including sleep-related hypermotor epilepsy (SHE), temporal lobe epilepsy (TLE), and frontal lobe epilepsy (FLE). Infantile epileptic spasms syndrome (IESS) is also a notable feature of NPRL2-related epilepsy. Malformations of cortical development (MCD, 8/20), especially focal cortical dysplasia (FCD, 6/20), are common neuroimaging abnormalities. Two-thirds of the NPRL2 variants reported are loss of function (LoF) (14/21). Among these mutations, c.100C>T (p.Arg34*) and c.314T>C (p.Leu105Pro) have been detected in two families (likely due to a founder effect).
    CONCLUSIONS: NPRL2-related epilepsy shows high phenotypic and genotypic heterogeneity. Our study expands the genotype spectrum of NPRL2-related epilepsy, and the phenotype of focal epilepsy involving the central region should be clearly distinguished with SeLECTS, with reference value for clinical diagnosis.
    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
    抑癌基因(TSGs)在肿瘤发生和耐药中起着至关重要的作用。我们基于TCGA数据库分析了3p21.3抑癌基因簇中包含的8个基因介导的透明细胞肾细胞癌(ccRCC)的亚型及其对TME细胞浸润的影响。通过主成分分析建立风险评分模型。通过蛋白质-蛋白质相互作用(PPI)分析选择hub基因NPRL2。通过CCK-8、集落形成试验验证NPRL2对ccRCC舒尼替尼敏感性的影响,伤口愈合试验,transwell试验和异种移植肿瘤模型。蛋白质印迹法检测蛋白质表达的变化。我们发现8个TSG在ccRCC样本中均有差异表达,这可以将ccRCC分为两个亚型。所构建的风险评分模型可以预测ccRCC患者的预后和药物敏感性。是ccRCC的独立预后因素。NPRL2过表达促进细胞凋亡,抑制EMT,降低PI3K/AKT/mTOR信号通路的磷酸化抑制其活性,并促进舒尼替尼对ccRCC细胞的敏感性。总的来说,我们的发现增加了对ccRCC中TSG的理解,提示NPRL2作为TSG可以增强舒尼替尼对ccRCC细胞的敏感性。
    Tumor suppressor genes (TSGs) play a crucial role in tumorigenesis and drug resistance. We analyzed the subtypes of clear cell renal cell carcinoma (ccRCC) mediated by 8 genes contained in the 3p21.3 tumor suppressor gene cluster and their effects on TME cell infiltration based on the TCGA database. The risk score model was established by principal component analysis. The hub gene NPRL2 was selected by protein-protein interactions (PPI) analysis. The effect of NPRL2 on sunitinib sensitivity of ccRCC was verified by using CCK-8, colony formation assay, wound healing assay, transwell assay and xenograft tumor model. Changes in protein expression were detected by Western blotting. We found that 8 TSGs were all differentially expressed in ccRCC samples, which could divide ccRCC into two subtypes. The constructed risk score model could predict the prognosis and drug sensitivity of ccRCC patients, and was an independent prognostic factor for ccRCC. Over-expression of NPRL2 promoted apoptosis, inhibited EMT, decreased the phosphorylation of the PI3K/AKT/mTOR signaling pathway to inhibit its activity, and promoted the sensitivity of sunitinib to ccRCC cells. Collectively, our findings increased the understanding of TSGs in ccRCC, suggesting that NPRL2 as a TSG could enhance sunitinib sensitivity to ccRCC cells.
    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
    Variants in GATOR1 genes are well established in focal epilepsy syndromes. A strong association of GATOR1 variants with drug-resistant epilepsy as well as an increased risk of sudden unexplained death in epilepsy warrants developing strategies to facilitate the identification of patients who could potentially benefit from genetic testing and precision medicine. We aimed to determine the yield of GATOR1 gene sequencing in patients with focal epilepsy typically referred for genetic testing, establish novel GATOR1 variants and determine clinical, electroencephalographic, and radiological characteristics of variant carriers.
    Ninety-six patients with clinical suspicion of genetic focal epilepsy with previous comprehensive diagnostic epilepsy evaluation in The Neurology Clinic, University Clinical Center of Serbia, were included in the study. Sequencing was performed using a custom gene panel encompassing DEPDC5, NPRL2, and NPRL3. Variants of interest (VOI) were classified according to criteria proposed by the American College of Medical Genetics and the Association for Molecular Pathology.
    Four previously unreported VOI in 4/96 (4.2%) patients were found in our cohort. Three likely pathogenic variants were determined in 3/96 (3.1%) patients, one frameshift variant in DEPDC5 in a patient with nonlesional frontal lobe epilepsy, one splicogenic DEPDC5 variant in a patient with nonlesional posterior quadrant epilepsy, and one frameshift variant in NPRL2 in a patient with temporal lobe epilepsy associated with hippocampal sclerosis. Only one VOI, a missense variant in NPRL3, found in 1/96 (1.1%) patients, was classified as a variant of unknown significance.
    GATOR1 gene sequencing was diagnostic in 3.1% of our cohort and revealed three novel likely pathogenic variants, including a previously unreported association of temporal lobe epilepsy with hippocampal sclerosis with an NPRL2 variant. Further research is essential for a better understanding of the clinical scope of GATOR1 gene-associated epilepsy.
    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
    GATOR2-GATOR1信号轴对于氨基酸依赖性mTORC1激活是必需的。然而,GATOR2复合物的分子功能仍然未知。这里,我们报道了Mios环域的破坏,WDR24或WDR59完全阻止氨基酸介导的mTORC1激活。机械上,通过与WDR59和WDR24的环域相互作用,Mios的环域充当维持GATOR2完整性的枢纽,其破坏导致WDR24的自我泛素化。生理学上,亮氨酸刺激使Sestrin2从WDR24的环域解离,并赋予其UBE2D3和随后的NPRL2泛素化的可用性,从而导致GATOR2介导的GATOR1失活。因此,WDR24消融或环缺失可防止mTORC1激活,导致小鼠E10.5严重的生长缺陷和胚胎致死率。因此,我们的发现表明,环域对于GATOR2将氨基酸可用性传递给mTORC1至关重要,并进一步揭示了胚胎发育过程中营养感知的必要性。
    The GATOR2-GATOR1 signaling axis is essential for amino-acid-dependent mTORC1 activation. However, the molecular function of the GATOR2 complex remains unknown. Here, we report that disruption of the Ring domains of Mios, WDR24, or WDR59 completely impedes amino-acid-mediated mTORC1 activation. Mechanistically, via interacting with Ring domains of WDR59 and WDR24, the Ring domain of Mios acts as a hub to maintain GATOR2 integrity, disruption of which leads to self-ubiquitination of WDR24. Physiologically, leucine stimulation dissociates Sestrin2 from the Ring domain of WDR24 and confers its availability to UBE2D3 and subsequent ubiquitination of NPRL2, contributing to GATOR2-mediated GATOR1 inactivation. As such, WDR24 ablation or Ring deletion prevents mTORC1 activation, leading to severe growth defects and embryonic lethality at E10.5 in mice. Hence, our findings demonstrate that Ring domains are essential for GATOR2 to transmit amino acid availability to mTORC1 and further reveal the essentiality of nutrient sensing during embryonic 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
    UNASSIGNED:胃腺癌(STAD)是胃癌的一种主要类型,具有很高的发病率和死亡率。NPRL2,候选抑癌基因,已被证明在各种类型的癌症中具有抗癌作用。因此,对STAD中NPRL2的综合分析可能为胃癌的治疗提供潜在的预后指标和临床靶点.
    UNASSIGNED:根据人类蛋白质图谱的数据分析了基因组表达和甲基化,基因表达Omnibus和Oncomine数据库。使用Kaplan-Meier方法进行生存分析,使用来自癌症基因组图谱数据库的数据。使用在线免疫细胞丰度标识符进行免疫相关性分析和对免疫疗法的反应预测。共表达分析,在R,通过逆概率治疗加权方法平衡临床协变量。
    UNASSIGNED:NPRL2在STAD中异常下调(P<0.05)。生存分析强调了NPRL2表达与患者临床结局之间的正相关(P<0.05)。基于共表达分析,我们发现NPRL2可能参与上皮-间质转化,胃癌干细胞,STAD对化疗药物的反应性(P<0.05)。此外,功能聚类分析显示NPRL2参与mTOR信号通路,自噬,和氨基酸饥饿反应(调整P<0.05)。此外,在STAD中,NPRL2与肿瘤浸润免疫细胞呈负相关,而与免疫治疗生物标志物呈正相关(P<0.05)。同时,预计NPRL2高表达的患者对免疫治疗的反应更好(P<0.05).最后,基于NPRL2相关基因构建的预后模型可以预测STAD患者的预后(AUC=0.641),风险评分是STAD患者的独立预后因素(HR=4.855,95%CI:2.683~8.785,P<0.001)。
    UNASSIGNED:本研究对NPRL2在STAD中的作用和潜在机制进行了全面分析,表明NPRL2是STAD患者生存和免疫治疗反应预测的潜在生物标志物。
    UNASSIGNED: Stomach adenocarcinoma (STAD) is a major type of gastric cancer with high morbidity and mortality. NPRL2, a candidate cancer suppressor gene, has been shown to have anti-cancer effects in various types of cancers. Therefore, comprehensive analyses of NPRL2 in STAD may provide a potential prognostic marker and clinical target for the management of gastric cancer.
    UNASSIGNED: Genomic expression and methylation were analysed based on data from the Human Protein Atlas, Gene Expression Omnibus and Oncomine database. Survival analyses were conducted with the Kaplan-Meier method, using data from The Cancer Genome Atlas database. Immune correlation analyses and prediction of response to immunotherapy were performed using the online Immune Cell Abundance Identifier. Co-expression analyses, functional clustering analyses and construction of a prognostic risk model were conducted in R, with the clinical covariates balanced by the inverse probability treatment weighting method.
    UNASSIGNED: NPRL2 was abnormally downregulated in STAD (P<0.05). Survival analysis highlighted a positive association between the expression of NPRL2 and clinical outcomes for patients (P<0.05). Based on co-expression analyses, we found that NPRL2 may be involved in epithelial-mesenchymal transition, gastric cancer stem cells, and responsiveness to chemotherapeutic agents in STAD (P<0.05). Furthermore, functional clustering analysis revealed that NPRL2 was involved in the mTOR signalling pathway, autophagy, and the amino acid starvation response (adjust P<0.05). In addition, NPRL2 was negatively associated with tumour-infiltrating immune cells while positively associated with immunotherapeutic biomarkers in STAD (P<0.05). Meanwhile, patients with high NPRL2 expression were predicted to have a better response to immunotherapy (P<0.05). Finally, a prognostic model constructed based on NPRL2-related genes could predict the prognosis of STAD patients (AUC =0.641), and the risk score was an independent prognostic factor for STAD patients (HR =4.855, 95% CI: 2.683-8.785, P<0.001).
    UNASSIGNED: The present study provided a comprehensive analysis of the role and potential mechanisms of NPRL2 in STAD, suggesting that NPRL2 is a potential biomarker for the survival and prediction of immunotherapy response in STAD.
    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
    氮通透酶调节因子样2(NPRL2)的基因突变与广泛的家族性局灶性癫痫有关,自闭症,癫痫患者突然意外死亡(SUDEP),但NPRL2对这些影响的作用机制尚不清楚。NPRL2是针对Rags1(GATOR1)复合物的GAP活性的必需亚基,当细胞内氨基酸含量较低时,它充当哺乳动物雷帕霉素复合物1(mTORC1)激酶的负调节因子。这里,我们发现NPRL2在小鼠兴奋性谷氨酸能神经元中的表达缺失在死亡前引起癫痫发作,与癫痫患者的SUDEP一致。此外,NPRL2表达缺失会增加mTORC1依赖性信号转导,并显著改变脑内氨基酸稳态.NPRL2的缺失减少了树突分支并增加了神经元中的电刺激动作电位(AP)的强度。增加的AP强度与癫痫相关的表达升高一致,NPRL2缺陷脑中的电压门控钠通道。NPRL2在原代神经元中的靶向缺失增加钠通道Scn1A的表达,而用称为雷帕霉素的药理学mTORC1抑制剂治疗可防止Scn1A上调。这些研究证明了NPRL2和mTORC1信号在钠通道调节中的新作用,会导致癫痫发作和早期致死。
    Genetic mutations in nitrogen permease regulator-like 2 (NPRL2) are associated with a wide spectrum of familial focal epilepsies, autism, and sudden unexpected death of epileptics (SUDEP), but the mechanisms by which NPRL2 contributes to these effects are not well known. NPRL2 is a requisite subunit of the GAP activity toward Rags 1 (GATOR1) complex, which functions as a negative regulator of mammalian target of rapamycin complex 1 (mTORC1) kinase when intracellular amino acids are low. Here, we show that loss of NPRL2 expression in mouse excitatory glutamatergic neurons causes seizures before death, consistent with SUDEP in humans with epilepsy. Additionally, the absence of NPRL2 expression increases mTORC1-dependent signal transduction and significantly alters amino acid homeostasis in the brain. Loss of NPRL2 reduces dendritic branching and increases the strength of electrically stimulated action potentials (APs) in neurons. The increased AP strength is consistent with elevated expression of epilepsy-linked, voltage-gated sodium channels in the NPRL2-deficient brain. Targeted deletion of NPRL2 in primary neurons increases the expression of sodium channel Scn1A, whereas treatment with the pharmacological mTORC1 inhibitor called rapamycin prevents Scn1A upregulation. These studies demonstrate a novel role of NPRL2 and mTORC1 signaling in the regulation of sodium channels, which can contribute to seizures and early lethality.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    氮通透酶调节因子样2(NPRL2)基因相关癫痫的表型在临床上表现为一系列癫痫综合征,包括家族性局灶性癫痫伴可变病灶(FFEVF),睡眠相关的运动过度癫痫(SHE),颞叶癫痫(TLE),额叶癫痫(FLE),和婴儿痉挛(IS)。NPRL2变体的表型和基因型之间的关联尚未被广泛研究。本研究旨在探讨NPRL2相关癫痫的表型和基因型谱。这里,我们介绍了两例NPRL2相关癫痫的临床病例,并讨论了其特点,诊断,和现有文献中的治疗过程。通过下一代测序鉴定出两种新的可能致病的NPRL2变体。包括一个剪接突变(c.933-1G>A),和一个移码突变(c.257delG)。文献复习结果显示,共有20例NPRL2相关癫痫患者的突变多是错义遗传。这些结果表明,对于有家族史的患者,应考虑局灶性癫痫NPRL2基因突变的可能性。携带不同NPRL2变异体的患者有不同的临床表现。我们的研究扩展了NPRL2的基因型谱,并表明NPRL2变体的类型可能为预后评估提供重要信息。
    The phenotype of nitrogen permease regulator-like 2 (NPRL2) gene-related epilepsy clinically manifests as a range of epilepsy syndromes, including familial focal epilepsy with variable foci (FFEVF), sleep-related hypermotor epilepsy (SHE), temporal lobe epilepsy (TLE), frontal lobe epilepsy (FLE), and infantile spasms (IS). The association between phenotype and genotype of NPRL2 variants has not been widely explored. This study aimed to explore the phenotype and genotype spectrum of NPRL2-related epilepsy. Here, we presented two clinical cases with NPRL2-related epilepsy, and discussed the characteristics, diagnosis, and treatment processes in the context of existing literature. Two novel NPRL2 likely pathogenic variants were identified by next-generation sequencing, including one splicing mutation (c.933-1G>A), and one frameshift mutation (c.257delG). The results of literature review showed that there were a total of 20 patients with NPRL2-related epilepsy whose mutations were mostly missense and hereditary. These findings indicate that the possibility of NPRL2 gene mutations in focal epilepsy should be considered for patients with family history, and that patients carrying different NPRL2 variants have different clinical manifestations. Our study expanded the genotype spectrum of NPRL2 and suggested that the type of NPRL2 variants might provide important information for the prognosis evaluation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号