Neurofibromin 2

  • 文章类型: Journal Article
    神经纤维瘤病2型是一种遗传性疾病,导致神经鞘瘤的形成和进行性生长,室管膜瘤,和/或脑膜瘤。NF2基因编码Merlin蛋白,它将细胞皮质元件连接到肌动蛋白细胞骨架,并调节许多关键酶,包括I组p21活化激酶(PAKs),Hippo途径激酶LATS,和mTORC。当Merlin突变或缺失时,PAK1和PAK2直接结合Merlin并传递增殖和存活信号,单独抑制第1组PAK还不足以在体内完全阻止NF2缺陷型脑膜瘤或神经鞘瘤的生长,提示需要第二种途径抑制剂。由于Hippo途径在NF2缺陷细胞中也被激活,最近已经以YAP-TEAD结合抑制剂的形式开发了几种Hippo途径的抑制剂。这些抑制剂防止促增殖和抗凋亡Hippo途径效应物的活化。在这项研究中,我们显示PAK抑制减缓细胞增殖,而TEAD抑制促进凋亡细胞死亡。最后,我们证明了PAK和TEAD抑制剂组合在几种NF2缺陷型神经鞘瘤细胞系中的疗效.
    Neurofibromatosis type 2 is a genetic disorder that results in the formation and progressive growth of schwannomas, ependymomas, and/or meningiomas. The NF2 gene encodes the Merlin protein, which links cell cortical elements to the actin cytoskeleton and regulates a number of key enzymes including Group I p21-activated kinases (PAKs), the Hippo-pathway kinase LATS, and mTORC. While PAK1 and PAK2 directly bind Merlin and transmit proliferation and survival signals when Merlin is mutated or absent, inhibition of Group 1 PAKs alone has not proven sufficient to completely stop the growth of NF2-deficient meningiomas or schwannomas in vivo, suggesting the need for a second pathway inhibitor. As the Hippo pathway is also activated in NF2-deficient cells, several inhibitors of the Hippo pathway have recently been developed in the form of YAP-TEAD binding inhibitors. These inhibitors prevent activation of pro-proliferation and anti-apoptotic Hippo pathway effectors. In this study, we show that PAK inhibition slows cell proliferation while TEAD inhibition promotes apoptotic cell death. Finally, we demonstrate the efficacy of PAK and TEAD inhibitor combinations in several NF2-deficient Schwannoma cell lines.
    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
    NF2相关神经鞘瘤病(NF2)是一种遗传综合征,其特征是神经系统良性肿瘤的生长,尤其是双侧前庭神经鞘瘤,脑膜瘤,和室管膜瘤.这篇综述巩固了目前关于NF2综合征的知识,强调与同名基因突变相关的分子病理学,NF2基因,以及其产品随后的功能障碍,Merlin蛋白.梅林,肿瘤抑制剂,整合了多个调节细胞接触的信号通路,扩散,和运动性,从而影响肿瘤生长。梅林的消失破坏了这些途径,导致肿瘤发生。我们讨论了另外两种可能与NF2缺乏症以及Merlin相关的蛋白质的作用:Yes相关蛋白1(YAP),这可能会促进肿瘤生长,和Raf激酶抑制蛋白(RKIP),这似乎抑制了肿瘤的发展。此外,这篇综述讨论了各种治疗方法的疗效,例如靶向特定途径或抑制NF2缺乏引起的新形态蛋白-蛋白相互作用的分子疗法。本概述不仅扩展了对NF2病理生理学的基本理解,而且还探讨了影响NF2综合征临床方法的新型治疗靶标的潜力。
    NF2-related schwannomatosis (NF2) is a genetic syndrome characterized by the growth of benign tumors in the nervous system, particularly bilateral vestibular schwannomas, meningiomas, and ependymomas. This review consolidates the current knowledge on NF2 syndrome, emphasizing the molecular pathology associated with the mutations in the gene of the same name, the NF2 gene, and the subsequent dysfunction of its product, the Merlin protein. Merlin, a tumor suppressor, integrates multiple signaling pathways that regulate cell contact, proliferation, and motility, thereby influencing tumor growth. The loss of Merlin disrupts these pathways, leading to tumorigenesis. We discuss the roles of another two proteins potentially associated with NF2 deficiency as well as Merlin: Yes-associated protein 1 (YAP), which may promote tumor growth, and Raf kinase inhibitory protein (RKIP), which appears to suppress tumor development. Additionally, this review discusses the efficacy of various treatments, such as molecular therapies that target specific pathways or inhibit neomorphic protein-protein interaction caused by NF2 deficiency. This overview not only expands on the fundamental understanding of NF2 pathophysiology but also explores the potential of novel therapeutic targets that affect the clinical approach to NF2 syndrome.
    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
    暂无摘要。
    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
    II型神经纤维瘤病(NFII)是由NF2基因缺失引起的遗传病,导致YAP/TAZ途径的激活和复发性雪旺细胞肿瘤,以及脑膜瘤和室管膜瘤.不幸的是,NFII的药理学选择很少。这里,我们进行全基因组CRISPR/Cas9筛选,以搜索合成致死基因,当被抑制时,导致NF2突变雪旺氏细胞而非NF2野生型细胞死亡。我们确定ACSL3和G6PD是NF2的两个合成致死伴侣,两者都参与脂质生物发生和细胞氧化还原。我们发现NF2突变体雪旺氏细胞比对照细胞更氧化,部分是由于参与NADPH生成的基因如ME1的表达降低。由于G6PD和ME1冗余地产生胞质NADPH,缺乏任何一种都与细胞活力相容,但不是两者都下调。由于G6PD的遗传缺陷在人群中是耐受的,G6PD可能是NFII的良好药理靶点。
    Neurofibromatosis Type II (NFII) is a genetic condition caused by loss of the NF2 gene, resulting in activation of the YAP/TAZ pathway and recurrent Schwann cell tumors, as well as meningiomas and ependymomas. Unfortunately, few pharmacological options are available for NFII. Here, we undertake a genome-wide CRISPR/Cas9 screen to search for synthetic-lethal genes that, when inhibited, cause death of NF2 mutant Schwann cells but not NF2 wildtype cells. We identify ACSL3 and G6PD as two synthetic-lethal partners for NF2, both involved in lipid biogenesis and cellular redox. We find that NF2 mutant Schwann cells are more oxidized than control cells, in part due to reduced expression of genes involved in NADPH generation such as ME1. Since G6PD and ME1 redundantly generate cytosolic NADPH, lack of either one is compatible with cell viability, but not down-regulation of both. Since genetic deficiency for G6PD is tolerated in the human population, G6PD could be a good pharmacological target for NFII.
    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
    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
    实体肿瘤可以普遍逃避增殖接触抑制(CIP),一种机制,当细胞与细胞接触时停止细胞增殖。梅林,ERM样蛋白质,至关重要地调节CIP,并且在各种癌症中经常失活,表明其在癌症生物学中作为肿瘤抑制因子的意义。尽管对Merlin在癌症中的作用进行了广泛的调查,它缺乏内在的催化活性和频繁的构象变化使其研究具有挑战性。为了应对这一挑战,我们利用创新的荧光素酶技术来创建和验证NanoBiT分裂荧光素酶生物传感器系统,其中Merlin被克隆在NanoLuc荧光素酶的两个分裂成分(LgBiT和SmBiT)之间。该系统能够在体外和活细胞内精确定量Merlin的构象和活性。这种生物传感器显着增强了对Merlin分子功能的研究,作为探索其对CIP和肿瘤发生的贡献的有力工具。
    Solid tumours can universally evade contact inhibition of proliferation (CIP), a mechanism halting cell proliferation when cell-cell contact occurs. Merlin, an ERM-like protein, crucially regulates CIP and is frequently deactivated in various cancers, indicating its significance as a tumour suppressor in cancer biology. Despite extensive investigations into Merlin\'s role in cancer, its lack of intrinsic catalytic activity and frequent conformation changes have made it notoriously challenging to study. To address this challenge, we harnessed innovative luciferase technologies to create and validate a NanoBiT split-luciferase biosensor system in which Merlin is cloned between two split components (LgBiT and SmBiT) of NanoLuc luciferase. This system enables precise quantification of Merlin\'s conformation and activity both in vitro and within living cells. This biosensor significantly enhances the study of Merlin\'s molecular functions, serving as a potent tool for exploring its contributions to CIP and tumorigenesis.
    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
    2型神经纤维瘤病(NF2)相关神经鞘瘤病是一种遗传性疾病,可导致多种类型的神经系统肿瘤的发展。原发性和诊断性肿瘤类型是双侧前庭神经鞘瘤。NF2没有治愈或药物治疗。推荐的治疗方法包括手术切除和放疗,这两种情况都会使患者出现严重的神经功能缺损或增加未来恶性肿瘤的风险。我们先前的先导高通量药物筛选的结果基于小鼠merlin缺陷雪旺氏细胞(MD-SC)的生存力丧失,将磷酸肌醇3-激酶(PI3K)抑制剂鉴定为强候选物。在这里,我们使用新型人类神经鞘瘤模型细胞进行组合药物筛选。我们确定了一种I类PI3K抑制剂,pictilisib和p21活化激酶(PAK)抑制剂,由于在细胞活力测定中的高协同作用,PF-3758309作为顶部组合。在原位同种异体移植小鼠模型中,单一和组合疗法均显著降低小鼠MD-SC的生长。抑制剂组合促进小鼠merlin缺陷型施万(MD-SC)细胞中的细胞周期停滞和凋亡,并促进人MD-SC中的细胞周期停滞。这项研究确定了PI3K和PAK途径作为NF2相关神经鞘瘤病联合药物治疗的潜在靶标。
    Neurofibromatosis Type 2 (NF2)-related schwannomatosis is a genetic disorder that causes development of multiple types of nervous system tumors. The primary and diagnostic tumor type is bilateral vestibular schwannoma. There is no cure or drug therapy for NF2. Recommended treatments include surgical resection and radiation, both of which can leave patients with severe neurological deficits or increase the risk of future malignant tumors. Results of our previous pilot high-throughput drug screen identified phosphoinositide 3-kinase (PI3K) inhibitors as strong candidates based on loss of viability of mouse merlin-deficient Schwann cells (MD-SCs). Here we used novel human schwannoma model cells to conduct combination drug screens. We identified a class I PI3K inhibitor, pictilisib and p21 activated kinase (PAK) inhibitor, PF-3758309 as the top combination due to high synergy in cell viability assays. Both single and combination therapies significantly reduced growth of mouse MD-SCs in an orthotopic allograft mouse model. The inhibitor combination promoted cell cycle arrest and apoptosis in mouse merlin-deficient Schwann (MD-SCs) cells and cell cycle arrest in human MD-SCs. This study identifies the PI3K and PAK pathways as potential targets for combination drug treatment of NF2-related schwannomatosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号