Neurofibromin 2

  • 文章类型: Journal Article
    2型神经纤维瘤病(NF2)基因,已知编码肿瘤抑制蛋白Merlin,是研究肿瘤发生和相关细胞过程的核心。这篇综述全面考察了NF2/Merlin的多方面作用,详细说明其结构特征,功能多样性,并参与各种信号通路,如Wnt/β-catenin,河马,TGF-β,RTKs,mTOR,缺口,还有刺猬.这些途径对细胞生长至关重要,扩散,和差异化。NF2突变与神经鞘瘤的发展特别相关,脑膜瘤,室管膜瘤,尽管这些特定细胞类型中肿瘤形成的确切机制仍不清楚。此外,这篇综述探讨了梅林在胚胎发育中的作用,强调NF2缺乏引起的严重发育缺陷和胚胎致死性。还讨论了针对这些遗传畸变的潜在治疗策略,强调mTOR的抑制剂,HDAC,和VEGF作为有希望的治疗途径。当前知识的综合强调了正在进行的研究的必要性,以阐明NF2/Merlin的详细机制并制定有效的治疗策略。最终旨在改善NF2突变个体的预后和生活质量。
    The neurofibromatosis type 2 (NF2) gene, known for encoding the tumor suppressor protein Merlin, is central to the study of tumorigenesis and associated cellular processes. This review comprehensively examines the multifaceted role of NF2/Merlin, detailing its structural characteristics, functional diversity, and involvement in various signaling pathways such as Wnt/β-catenin, Hippo, TGF-β, RTKs, mTOR, Notch, and Hedgehog. These pathways are crucial for cellular growth, proliferation, and differentiation. NF2 mutations are specifically linked to the development of schwannomas, meningiomas, and ependymomas, although the precise mechanisms of tumor formation in these specific cell types remain unclear. Additionally, the review explores Merlin\'s role in embryogenesis, highlighting the severe developmental defects and embryonic lethality caused by NF2 deficiency. The potential therapeutic strategies targeting these genetic aberrations are also discussed, emphasizing inhibitors of mTOR, HDAC, and VEGF as promising avenues for treatment. This synthesis of current knowledge underscores the necessity for ongoing research to elucidate the detailed mechanisms of NF2/Merlin and develop effective therapeutic strategies, ultimately aiming to improve the prognosis and quality of life for individuals with NF2 mutations.
    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
    脑膜瘤是最常见的原发性颅内肿瘤,占所有神经系统肿瘤的近30%。大约一半的脑膜瘤患者表现出神经纤维蛋白2(NF2)基因失活。这里,NF2显示与IOMM-Lee中的内质网(ER)钙(Ca2)通道肌醇1,4,5-三磷酸受体1(IP3R1)相互作用,高度恶性脑膜瘤细胞系,NF2的F1子域在这种相互作用中起着关键作用。功能分析表明,NF2通过与IP3R1结合促进IP3R(Ser1756)的磷酸化和IP3R介导的内质网(ER)Ca2释放,从而导致Ca2依赖性细胞凋亡。NF2基因敲除降低Ca2+释放,促进细胞凋亡抵抗,通过野生型NF2过表达而不是通过F1亚结构域缺失截短过表达来拯救。在小鼠模型中进一步研究了NF2缺陷对肿瘤发展的影响。NF2基因敲除或突变引起的NF2表达水平降低影响IP3R通道的活性,减少Ca2+依赖性细胞凋亡,从而促进肿瘤的发展。我们阐明了NF2和IP3R1的相互作用模式,揭示了NF2调节IP3R1介导的Ca2+释放的分子机制,并阐明了脑膜瘤相关NF2变异的新致病机制。我们的研究拓宽了目前对NF2生物学功能的认识,为NF2相关脑膜瘤的药物筛选提供了思路。
    Meningiomas are the most common primary intracranial tumors and account for nearly 30% of all nervous system tumors. Approximately half of meningioma patients exhibit neurofibromin 2 (NF2) gene inactivation. Here, NF2 was shown to interact with the endoplasmic reticulum (ER) calcium (Ca2+) channel inositol 1,4,5-trisphosphate receptor 1 (IP3R1) in IOMM-Lee, a high-grade malignant meningioma cell line, and the F1 subdomain of NF2 plays a critical role in this interaction. Functional assays indicated that NF2 promotes the phosphorylation of IP3R (Ser 1756) and IP3R-mediated endoplasmic reticulum (ER) Ca2+ release by binding to IP3R1, which results in Ca2+-dependent apoptosis. Knockout of NF2 decreased Ca2+ release and promoted resistance to apoptosis, which was rescued by wild-type NF2 overexpression but not by F1 subdomain deletion truncation overexpression. The effects of NF2 defects on the development of tumors were further studied in mouse models. The decreased expression level of NF2 caused by NF2 gene knockout or mutation affects the activity of the IP3R channel, which reduces Ca2+-dependent apoptosis, thereby promoting the development of tumors. We elucidated the interaction patterns of NF2 and IP3R1, revealed the molecular mechanism through which NF2 regulates IP3R1-mediated Ca2+ release, and elucidated the new pathogenic mechanism of meningioma-related NF2 variants. Our study broadens the current understanding of the biological function of NF2 and provides ideas for drug screening of NF2-associated meningioma.
    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
    背景:程序性死亡配体1(PD-L1)表达是一种免疫逃避机制,已在许多肿瘤中得到证实,并且通常与不良预后相关。多年来,抗PD-L1药物作为新型抗癌治疗药物,在众多恶性肿瘤中诱导持久的肿瘤消退,已引起人们的关注.它们可能是2型神经纤维瘤病(NF2)患者的新治疗选择。
    目的:本研究的目的是检测NF2相关脑膜瘤中PD-L1的表达,探讨PD-L1下调对肿瘤细胞特性和T细胞功能的影响,并探讨调节PD-L1表达的可能通路,进一步剖析NF2肿瘤免疫抑制的可能机制,为NF2患者提供新的治疗选择。
    结果:PD-L1在NF2相关脑膜瘤中异质表达。NF2相关脑膜瘤细胞PD-L1敲低后,肿瘤细胞增殖被显著抑制,细胞凋亡率升高。当T细胞与siPD-L1转染的NF2相关脑膜瘤细胞共培养时,CD69在CD4+和CD8+T细胞上的表达被部分逆转,CD8+T细胞对siPD-L1转染肿瘤细胞的杀伤能力部分恢复。结果还显示PI3K-AKT-mTOR通路调节PD-L1的表达,mTOR抑制剂雷帕霉素快速且持续地抑制PD-L1表达。体内实验结果表明,抗PD-L1抗体可能与mTOR抑制剂在减少肿瘤细胞增殖方面具有协同作用,并且减少的PD-L1表达可能有助于抗肿瘤功效。
    结论:靶向PD-L1可能有助于NF2相关脑膜瘤恢复肿瘤浸润淋巴细胞的功能,诱导细胞凋亡,抑制肿瘤增殖。剖析PD-L1驱动的NF2相关脑膜瘤的肿瘤发生的机制将有助于提高我们对肿瘤进展的潜在机制的理解,并有助于进一步完善当前疗法以改善NF2患者的治疗。
    BACKGROUND: Programmed death-ligand 1 (PD-L1) expression is an immune evasion mechanism that has been demonstrated in many tumors and is commonly associated with a poor prognosis. Over the years, anti-PD-L1 agents have gained attention as novel anticancer therapeutics that induce durable tumor regression in numerous malignancies. They may be a new treatment choice for neurofibromatosis type 2 (NF2) patients.
    OBJECTIVE: The aims of this study were to detect the expression of PD-L1 in NF2-associated meningiomas, explore the effect of PD-L1 downregulation on tumor cell characteristics and T-cell functions, and investigate the possible pathways that regulate PD-L1 expression to further dissect the possible mechanism of immune suppression in NF2 tumors and to provide new treatment options for NF2 patients.
    RESULTS: PD-L1 is heterogeneously expressed in NF2-associated meningiomas. After PD-L1 knockdown in NF2-associated meningioma cells, tumor cell proliferation was significantly inhibited, and the apoptosis rate was elevated. When T cells were cocultured with siPD-L1-transfected NF2-associated meningioma cells, the expression of CD69 on both CD4+ and CD8+ T cells was partly reversed, and the capacity of CD8+ T cells to kill siPD-L1-transfected tumor cells was partly restored. Results also showed that the PI3K-AKT-mTOR pathway regulates PD-L1 expression, and the mTOR inhibitor rapamycin rapidly and persistently suppresses PD-L1 expression. In vivo experimental results suggested that anti-PD-L1 antibody may have a synergetic effect with the mTOR inhibitor in reducing tumor cell proliferation and that reduced PD-L1 expression could contribute to antitumor efficacy.
    CONCLUSIONS: Targeting PD-L1 could be helpful for restoring the function of tumor-infiltrating lymphocytes and inducing apoptosis to inhibit tumor proliferation in NF2-associated meningiomas. Dissecting the mechanisms of the PD-L1-driven tumorigenesis of NF2-associated meningioma will help to improve our understanding of the mechanisms underlying tumor progression and could facilitate further refinement of current therapies to improve the treatment of NF2 patients.
    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
    昆虫翅膀的发育是一个迷人而复杂的过程,涉及通过细胞增殖和凋亡调节翅膀大小。在这项研究中,我们发现Ter94是一种AAA-ATP酶,对于适当的机翼大小至关重要,这取决于其ATPase活性。Ter94的缺失能够抑制Hippo靶基因。当Ter94耗尽时,它导致机翼尺寸减小和细胞凋亡增加,可以通过抑制河马途径来拯救。生化实验表明,Ter94与Mer相互结合,河马途径的一个关键上游部分,破坏了它与Ex和Kib的互动.这种破坏阻止了Ex-Mer-Kib复合体的形成,最终导致河马途径失活并促进机翼的正常发育。最后,我们展示了hVCP,Ter94的人类同源物能够代替Ter94调节果蝇的翅膀大小,强调它们的功能保护。总之,Ter94通过干扰Ex-Mer-Kib复合体在调节机翼大小方面发挥着积极作用,这导致河马途径的抑制。
    Insect wing development is a fascinating and intricate process that involves the regulation of wing size through cell proliferation and apoptosis. In this study, we find that Ter94, an AAA-ATPase, is essential for proper wing size dependently on its ATPase activity. Loss of Ter94 enables the suppression of Hippo target genes. When Ter94 is depleted, it results in reduced wing size and increased apoptosis, which can be rescued by inhibiting the Hippo pathway. Biochemical experiments reveal that Ter94 reciprocally binds to Mer, a critical upstream component of the Hippo pathway, and disrupts its interaction with Ex and Kib. This disruption prevents the formation of the Ex-Mer-Kib complex, ultimately leading to the inactivation of the Hippo pathway and promoting proper wing development. Finally, we show that hVCP, the human homolog of Ter94, is able to substitute for Ter94 in modulating Drosophila wing size, underscoring their functional conservation. In conclusion, Ter94 plays a positive role in regulating wing size by interfering with the Ex-Mer-Kib complex, which results in the suppression of the Hippo pathway.
    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
    作为Hippo信号通路的输出效应子,TEAD转录因子和共激活因子YAP在促进细胞增殖和器官大小方面发挥关键作用。已显示肿瘤抑制因子NF2激活LATS1/2激酶并与Hippo途径相互作用以抑制YAP-TEAD复合物。然而,NF2是否以及如何直接调节TEAD仍然未知。我们确定了NF2和TEAD4之间的直接联系和物理相互作用。NF2通过其FERM结构域和C末端尾部与TEAD4相互作用,并独立于LATS1/2和YAP降低TEAD4的蛋白质稳定性。此外,NF2抑制TEAD4棕榈酰化并诱导TEAD4的细胞质易位,导致TEAD4的泛素化和功能障碍。此外,与TEAD4的相互作用是NF2功能抑制细胞增殖所必需的。这些发现揭示了NF2作为转录因子TEAD的结合伴侣和抑制剂的意想不到的作用,揭示了NF2如何通过Hippo信号级联作为肿瘤抑制因子的替代机制。
    As an output effector of the Hippo signaling pathway, the TEAD transcription factor and co-activator YAP play crucial functions in promoting cell proliferation and organ size. The tumor suppressor NF2 has been shown to activate LATS1/2 kinases and interplay with the Hippo pathway to suppress the YAP-TEAD complex. However, whether and how NF2 could directly regulate TEAD remains unknown. We identified a direct link and physical interaction between NF2 and TEAD4. NF2 interacted with TEAD4 through its FERM domain and C-terminal tail and decreased the protein stability of TEAD4 independently of LATS1/2 and YAP. Furthermore, NF2 inhibited TEAD4 palmitoylation and induced the cytoplasmic translocation of TEAD4, resulting in ubiquitination and dysfunction of TEAD4. Moreover, the interaction with TEAD4 is required for NF2 function to suppress cell proliferation. These findings reveal an unanticipated role of NF2 as a binding partner and inhibitor of the transcription factor TEAD, shedding light on an alternative mechanism of how NF2 functions as a tumor suppressor through the Hippo signaling cascade.
    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
    长非编码RNA小核仁RNA宿主基因5(SNHG5)是在各种人类癌症中发现的癌基因。然而,目前尚不清楚SNHG5在激活肝星状细胞(HSC)和肝纤维化中的作用。在这项研究中,发现SNHG5在体外活化的HSC和体内从纤维化肝分离的原代HSC中上调,SNHG5的抑制抑制了HSC的活化。值得注意的是,神经纤维蛋白2(NF2),河马信号的主要激活器,参与SNHG5对HSC激活的影响。进一步证实了SNHG5与NF2蛋白之间的相互作用,阻止两者的联合使用可以有效阻断SNHG5抑制对EMT过程和Hippo信号传导的影响。此外,在慢性乙型肝炎患者中发现较高的SNHG5,并与纤维化阶段相关。总之,我们证明SNHG5可以通过调节NF2和Hippo途径作为激活的HSC调节因子。
    Long noncoding RNA small nucleolar RNA host gene 5 (SNHG5) is an oncogene found in various human cancers. However, it is unclear what role SNHG5 plays in activating hepatic stellate cells (HSCs) and liver fibrosis. In this study, SNHG5 was found to be upregulated in activated HSCs in vitro and in primary HSCs isolated from fibrotic liver in vivo, and inhibition of SNHG5 suppressed HSC activation. Notably, Neurofibromin 2 (NF2), the main activator for Hippo signalling, was involved in the effects of SNHG5 on HSC activation. The interaction between SNHG5 and NF2 protein was further confirmed, and preventing the combination of the two could effectively block the effects of SNHG5 inhibition on EMT process and Hippo signaling. Additionally, higher SNHG5 was found in chronic hepatitis B patients and associated with the fibrosis stage. Altogether, we demonstrate that SNHG5 could serve as an activated HSCs regulator via regulating NF2 and Hippo pathway.
    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:枕骨大孔(FM)脑膜瘤构成了重大的手术挑战,并且具有很高的发病率和死亡率。本研究旨在调查FM脑膜瘤中临床可行突变的分布,并确定与特定突变谱相关的临床特征。
    方法:作者对来自三个国际机构的62个FM脑膜瘤进行了靶向下一代测序,涵盖所有相关脑膜瘤基因(AKT1、KLF4、NF2、POLR2A、PIK3CA,SMO,TERT启动子,和TRAF7)。患有辐射诱导的脑膜瘤或2型神经纤维瘤病(NF2)的患者被排除在研究之外。此外,患者和肿瘤特征,包括年龄,性别,放射学特征,和肿瘤的位置,进行回顾性收集和评估。
    结果:研究队列包括46名女性和16名男性患者。在58例患者(93.5%)中检测到有临床意义的驱动突变。最常见的改变是TRAF7突变(26,41.9%),其次是AKT1E17K突变(19,30.6%)。两种突变均与相对于脑干的前外侧肿瘤位置显着相关(p=0.0078)。11例(17.7%)存在NF2突变,与肿瘤后部位置相关。与TRAF7和AKT1E17K突变的肿瘤相反。FM脑膜瘤的其他常见突变包括POLR2A突变(8,12.9%;6POLR2AQ403K和2POLR2AH439_L440del),KLF4K409Q突变(7,11.3%),和PIK3CA突变(4,6.5%;2个PIK3CAH1047R和2个PIK3CAE545K)。POLR2A和KLF4突变仅发生在女性患者中,与特定肿瘤位置没有显着关联。所有携带AKT1E17K和POLR2A突变的肿瘤均显示脑膜上皮组织学。十个肿瘤显示肿瘤内钙化,与AKT1突变型FM脑膜瘤相比,NF2突变型的频率明显更高(p=0.047)。
    结论:这些发现为FM脑膜瘤的分子遗传学和临床病理特征提供了重要的见解。与肿瘤位置相关的特定遗传改变的鉴定,volume,钙化,组织学,诊断时的性别可能会对未来的个性化治疗策略产生影响。
    OBJECTIVE: Foramen magnum (FM) meningiomas pose significant surgical challenges and have high morbidity and mortality rates. This study aimed to investigate the distribution of clinically actionable mutations in FM meningiomas and identify clinical characteristics associated with specific mutational profiles.
    METHODS: The authors conducted targeted next-generation sequencing of 62 FM meningiomas from three international institutions, covering all relevant meningioma genes (AKT1, KLF4, NF2, POLR2A, PIK3CA, SMO, TERT promoter, and TRAF7). Patients with a radiation-induced meningioma or neurofibromatosis type 2 (NF2) were excluded from the study. Additionally, patient and tumor characteristics, including age, sex, radiological features, and tumor location, were retrospectively collected and evaluated.
    RESULTS: The study cohort consisted of 46 female and 16 male patients. Clinically significant driver mutations were detected in 58 patients (93.5%). The most commonly observed alteration was TRAF7 mutations (26, 41.9%), followed by AKT1E17K mutations (19, 30.6%). Both mutations were significantly associated with an anterolateral tumor location relative to the brainstem (p = 0.0078). NF2 mutations were present in 11 cases (17.7%) and were associated with posterior tumor location, in contrast to tumors with TRAF7 and AKT1E17K mutations. Other common mutations in FM meningiomas included POLR2A mutations (8, 12.9%; 6 POLR2AQ403K and 2 POLR2AH439_L440del), KLF4K409Q mutations (7, 11.3%), and PIK3CA mutations (4, 6.5%; 2 PIK3CAH1047R and 2 PIK3CAE545K). POLR2A and KLF4 mutations exclusively occurred in female patients and showed no significant association with specific tumor locations. All tumors harboring AKT1E17K and POLR2A mutations displayed meningothelial histology. Ten tumors exhibited intratumoral calcification, which was significantly more frequent in NF2-mutant compared with AKT1-mutant FM meningiomas (p = 0.047).
    CONCLUSIONS: These findings provide important insights into the molecular genetics and clinicopathological characteristics of FM meningiomas. The identification of specific genetic alterations associated with tumor location, volume, calcification, histology, and sex at diagnosis may have implications for personalized treatment strategies in the future.
    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
    创伤性脑损伤(TBI)是世界范围内的常见疾病,具有很高的死亡率和致残率。除了主要的机械损伤,与TBI相关的继发性损伤也可以引起许多病理变化,比如脑水肿,神经细胞凋亡,和神经炎症,这进一步加剧了神经功能障碍,甚至导致因原发性损伤而死亡。其中,神经元凋亡是损伤的关键环节。黑皮质素-1受体(MC1R)是一种G蛋白偶联受体,属于黑皮质素受体家族。研究表明,MC1R的激活抑制氧化应激和细胞凋亡,并赋予对各种神经系统疾病的神经保护作用。Merlin是NF2基因的蛋白质产物,在小鼠的中枢神经系统(CNS)中广泛表达,老鼠,和人类。研究表明Merlin与MC1R有关。在这项研究中,我们探讨了MC1R的抗凋亡作用和潜在机制。通过控制皮质冲击建立TBI大鼠模型。使用MC1R特异性激动剂Nle4-D-Phe7-α-黑素细胞(NDP-MSH)和抑制剂MSG-606来探索TBI后MC1R和Merlin的作用,并研究相关机制。结果表明,TBI后MC1R和Merlin的表达水平上调,MC1R的激活促进了Merlin的表达。Further,我们发现MC1R激活能显著改善TBI诱导的大鼠神经功能障碍,减轻脑水肿和神经元凋亡。机械上,其神经保护功能和抗凋亡作用部分与MC1R激活有关。总之,我们证明,TBI后MC1R的激活可能通过上调Merlin的表达来抑制细胞凋亡并赋予神经保护作用。
    Traumatic brain injury (TBI) is a common disease worldwide with high mortality and disability rates. Besides the primary mechanical injury, the secondary injury associated with TBI can also induce numerous pathological changes, such as brain edema, nerve apoptosis, and neuroinflammation, which further aggravates neurological dysfunction and even causes the death due to the primary injury. Among them, neuronal apoptosis is a key link in the injury. Melanocortin-1 receptor (MC1R) is a G protein coupled receptor, belonging to the melanocortin receptor family. Studies have shown that activation of MC1R inhibits oxidative stress and apoptosis, and confers neuroprotective effects against various neurological diseases. Merlin is a protein product of the NF2 gene, which is widely expressed in the central nervous system (CNS) of mice, rats, and humans. Studies have indicated that Merlin is associated with MC1R. In this study, we explored the anti-apoptotic effects and potential mechanisms of MC1R. A rat model of TBI was established through controlled cortical impact. The MC1R-specific agonist Nle4-D-Phe7-α-Melanocyte (NDP-MSH) and the inhibitor MSG-606 were employed to explore the effects of MC1R and Merlin following TBI and investigated the associated mechanisms. The results showed that the expression levels of MC1R and Merlin were upregulated after TBI, and activation of MC1R promoted Merlin expression. Further, we found that MC1R activation significantly improved neurological dysfunction and reduced brain edema and neuronal apoptosis induced by TBI in rats. Mechanistically, its neuroprotective function and anti-apoptotic were partly associated with MC1R activation. In conclusion, we demonstrated that MC1R activation after TBI may inhibit apoptosis and confer neuroprotection by upregulating the expression of Merlin.
    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
    据报道,中性粒细胞和中性粒细胞胞外诱捕网(NETs)与肿瘤的发展有关,但是确切的作用和具体的机制仍然知之甚少,尤其是三阴性乳腺癌(TNBC)。在这项研究中,我们的结果表明,在TNBC组织中的NETs形成高于非TNBC组织,NETs的形成与肿瘤大小明显相关,TNBC患者的ki67水平和淋巴结转移。随后的体内实验表明,抑制NETs可以抑制TNBC肿瘤的生长和肺转移。进一步的体外实验发现,NETs对TNBC细胞的致癌功能可能取决于TLR9的表达。我们还发现,术后发热的TNBC患者外周血中性粒细胞易形成NETs,并可增强TNBC细胞的增殖和侵袭能力。机械上,我们发现,NETs可以与TLR9相互作用以减少Merlin磷酸化,这有助于TNBC细胞铁凋亡抗性。我们的工作为NETs促进TNBC进展和阻断NETs关键调节剂的机制提供了新的见解,这可能是TNBC的有前途的治疗策略。
    Neutrophil and neutrophil extracellular traps (NETs) were reported to be associated with tumor development, but the exact role and concrete mechanisms are still poorly understood, especially in triple negative breast cancer (TNBC). In this study, our results exhibited that NETs formation in TNBC tissues was higher than that in non-TNBC tissues, and NETs formation was distinctly correlated with tumor size, ki67 level and lymph node metastasis in TNBC patients. Subsequent in vivo experiments demonstrated that NETs inhibition could suppress TNBC tumor growth and lung metastasis. Further in vitro experiments uncovered that oncogenic function of NETs on TNBC cells were possibly dependent on TLR9 expression. We also found that neutrophils from peripheral blood of TNBC patients with postoperative fever were prone to form NETs and could enhance the proliferation and invasion of TNBC cells. Mechanistically, we revealed that NETs could interact with TLR9 to decrease Merlin phosphorylation which contributed to TNBC cell ferroptosis resistance. Our work provides a novel insight into the mechanism of NETs promoting TNBC progression and blocking the key modulator of NETs might be a promising therapeutic strategy in TNBC.
    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
    丙型肝炎病毒(HCV)感染导致肝细胞脂质代谢异常,导致肝脏脂肪变性甚至肝细胞癌。HCV非结构蛋白4B(NS4B)已被报道诱导脂肪生成,但潜在的机制尚不清楚。在这项研究中,进行蛋白质印迹以研究NS4B蛋白水平对Hippo和AKT信号通路的关键效应子的影响。Yes相关蛋白(YAP)和膜蛋白-ezrin-radixin样蛋白(Merlin)是Hippo途径的效应子。NS4B下调Merlin和磷酸化YAP(p-YAP)蛋白的表达,同时增加关键AKT途径蛋白p-AKT和NF-κB的表达。通过观察Merlin过表达或沉默时AKT通路蛋白的水平,已确定Merlin介导AKT途径。我们认为HCVNS4B可能通过抑制Hippo途径介导AKT信号通路。在过表达NS4B的Huh7.5细胞中观察到脂滴,当梅林沉默时,它们的数量显著增加。NS4B的过表达和Merlin沉默增强了固醇调节元件结合蛋白(SREBPs)的表达,已被证明是控制脂肪酸合成的关键调节因素。NS4B和Merlin沉默也增强了肝癌细胞的体外增殖能力。总之,NS4B通过Hippo-YAP途径对AKT信号传导途径的作用诱导脂肪生成,从而在HCV相关疾病的发病机理中发挥重要作用。
    Hepatitis C virus (HCV) infection causes abnormal lipid metabolism in hepatocytes, which leads to hepatic steatosis and even hepatocellular carcinoma. HCV nonstructural protein 4B (NS4B) has been reported to induce lipogenesis, but the underlying mechanism is unclear. In this study, western blots were performed to investigate the effect of NS4B protein levels on key effectors of the Hippo and AKT signaling pathways. Yes-associated protein (YAP) and moesin-ezrin-radixin-like protein (Merlin) are effectors of the Hippo pathway. NS4B downregulated Merlin and phosphorylated YAP (p-YAP) protein expression while increasing the expression of the key AKT pathway proteins p-AKT and NF-κB. By observing the levels of AKT pathway proteins when Merlin was overexpressed or silenced, it was determined that Merlin mediates the AKT pathway. We suggest that HCV NS4B may mediate the AKT signaling pathway by inhibiting the Hippo pathway. Lipid droplets were observed in Huh7.5 cells overexpressing NS4B, and they increased significantly in number when Merlin was silenced. Overexpression of NS4B and Merlin silencing enhanced the expression of sterol regulatory element binding proteins (SREBPs), which have been demonstrated to be key regulatory factors controlling fatty acid synthesis. NS4B and Merlin silencing also enhanced the in vitro proliferative capacity of hepatocellular carcinoma cells. In conclusion, NS4B induces lipogenesis via the effect of the Hippo-YAP pathway on the AKT signaling pathway and thereby plays a significant role in the pathogenesis of HCV-associated diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号