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
    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: English Abstract
    OBJECTIVE: To identify the characteristics of pain syndrome in patients with schwannomas depending on genetic predisposition.
    METHODS: The study included 46 patients with peripheral, spinal and intracranial schwannomas, corresponding to the schwannomatosis phenotype according to the 2022 clinical criteria. All patients underwent sequencing of the LZRT1, Nf2 and SMARCB1 and a copy number study in the NF2.
    RESULTS: The most severe widespread pain was observed in patients with pathogenic LZRT1 variants, while patients with mosaic variants may not even have local tumor-related pain. Patients with SMARCB1variants may have no pain or have localized pain that responds well to surgical treatment.
    CONCLUSIONS: Further studies of the molecular features of schwannomatosis and driver mutations in the pathogenesis of pain are necessary to improve the effectiveness of pain therapy in this group of patients. Schwannomatosis is a disease from the group of neurofibromatosis, manifested by the development of multiple schwannomas. Neuropathic pain is one of the main symptoms characteristic of peripheral schwannomas, however, the severity and prevalence of the pain syndrome does not always correlate with the location of the tumors. According to modern concepts, the key factors influencing the characteristics of the pain syndrome are the target gene and the type of pathogenic variant. The most severe widespread pain is observed in patients with pathogenic variants in the LZRT1 gene, while patients with mosaic variants may not even have local pain associated with tumors. Patients with variants in SMARCB1 may have no pain or localized pain that responds well to surgical treatment.
    UNASSIGNED: Выявить особенности болевого синдрома у пациентов со шванномами в зависимости от генетической предрасположенности.
    UNASSIGNED: В исследование включены 46 пациентов с периферическими, спинальными и интракраниальными шванномами, соответствующие фенотипу шванноматозов согласно клиническим критериям 2022 г. Всем пациентам проводилось секвенирование генов LZRT1, Nf2 и SMARCB1 и исследование количества копий в гене NF2.
    UNASSIGNED: Наиболее тяжелая распространенная боль отмечалась у пациентов с патогенными вариантами в гене LZRT1, при этом у пациентов с мозаичными вариантами могло не быть даже локальной боли, связанной с новообразованиями. У пациентов с вариантами в SMARCB1 могло не быть боли или выявлялась локальная боль, хорошо реагирующая на хирургическое лечение.
    UNASSIGNED: Дальнейшие исследования молекулярных особенностей шванноматозов и драйверных мутаций в патогенезе боли необходимы для повышения эффективности терапии боли в данной группе пациентов.
    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
    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
    在中枢神经系统中,少突胶质细胞(OLs)形成髓鞘依赖于肌动蛋白细胞骨架聚合向解聚的转变。触发这种开关的分子机制尚未阐明。这里,我们确定P21激活的激酶1(PAK1)是OLs中肌动蛋白解聚的主要调节因子。我们的结果表明,PAK1以激酶抑制的形式在OLs中积累,触发肌动蛋白拆卸和,因此,髓鞘膜扩张.值得注意的是,PAK1结合配偶体的蛋白质组学分析能够鉴定NF2/Merlin作为其内源性抑制剂。我们的发现表明,在OLs中Nf2敲低导致PAK1激活,肌动蛋白聚合,和髓磷脂膜扩张的减少。通过用PAK1抑制剂治疗来挽救这种效应。我们还提供证据表明,少突胶质细胞中特定的Pak1功能丧失会刺激体内髓鞘的增厚。总的来说,我们的数据表明,PAK1和NF2/Merlin对OL的肌动蛋白细胞骨架的拮抗作用对于正常的髓磷脂形成至关重要。这些发现在脱髓鞘疾病和神经发育障碍中具有广泛的机制和治疗意义。
    In the central nervous system, the formation of myelin by oligodendrocytes (OLs) relies on the switch from the polymerization of the actin cytoskeleton to its depolymerization. The molecular mechanisms that trigger this switch have yet to be elucidated. Here, we identified P21-activated kinase 1 (PAK1) as a major regulator of actin depolymerization in OLs. Our results demonstrate that PAK1 accumulates in OLs in a kinase-inhibited form, triggering actin disassembly and, consequently, myelin membrane expansion. Remarkably, proteomic analysis of PAK1 binding partners enabled the identification of NF2/Merlin as its endogenous inhibitor. Our findings indicate that Nf2 knockdown in OLs results in PAK1 activation, actin polymerization, and a reduction in OL myelin membrane expansion. This effect is rescued by treatment with a PAK1 inhibitor. We also provide evidence that the specific Pak1 loss-of-function in oligodendroglia stimulates the thickening of myelin sheaths in vivo. Overall, our data indicate that the antagonistic actions of PAK1 and NF2/Merlin on the actin cytoskeleton of the OLs are critical for proper myelin formation. These findings have broad mechanistic and therapeutic implications in demyelinating diseases and neurodevelopmental disorders.
    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
    心肌细胞分化涉及通过BMP(骨形态发生蛋白)/Wnt信号通路的调节逐步清除抑制因子和命运限制调节因子。然而,特定发育信号通路的机制以及如何消除监管障碍仍不清楚.
    我们使用肌球蛋白重链6(MYH6)-GFP(绿色荧光蛋白)报告系统进行了全基因组CRISPR筛选,以揭示人胚胎干细胞中心肌细胞规格的必需调节因子。在对25个候选物进行独立的二级sgRNA验证后,我们鉴定了NF2(神经纤维蛋白2),膜蛋白-ezrin-radixin样(MERLIN)肿瘤抑制因子,作为早期心肌细胞谱系规范的上游驱动因素。使用CRISPR-Cas9产生独立的单克隆NF2敲除,并且通过跨分化时间点的大量RNA测序和蛋白质表达分析来推断细胞状态。通过使用体外二维微图案化的gapstruoid模型评估末端谱系分化,三系分化,和心肌细胞分化。使用定点诱变评估了NF2与其相互作用伴侣的蛋白质相互作用和翻译后修饰,共免疫沉淀,和邻近连接测定。
    来自NF2无效细胞的转录调节和轨迹推断揭示了心肌细胞身份的丧失和非中胚层身份的获得。NF2敲除细胞中早期中胚层谱系抑制因子SOX2的持续升高和晚期抗心调节因子CDX2和MSX1的上调反映了NF2在消除调节路障中的必要作用。此外,我们发现NF2和AMOT(血管生成素)在中内胚层形成过程中与YAP(Yes相关蛋白)协同结合,从而防止YAP激活,独立于经典MST(哺乳动物无菌20样丝氨酸-苏氨酸蛋白激酶)-LATS(大肿瘤抑制因子丝氨酸-苏氨酸蛋白激酶)信号传导。机械上,通过野生型和NF2丝氨酸-518磷酸根拯救心肌细胞谱系的身份,但不是NF2FERM(ezrin-radixin-meosin同源蛋白)域蓝盒突变体,证明在粘附连接处AMOT-NF2-YAP支架复合物的关键FERM结构域依赖性形成是早期心肌细胞谱系分化所必需的。
    这些结果通过隔离YAP的抑制作用并缓解心肌细胞途径的调节障碍,为NF2在早期上皮-间质转化过程中的重要作用提供了机械见解。
    UNASSIGNED: Cardiomyocyte differentiation involves a stepwise clearance of repressors and fate-restricting regulators through the modulation of BMP (bone morphogenic protein)/Wnt-signaling pathways. However, the mechanisms and how regulatory roadblocks are removed with specific developmental signaling pathways remain unclear.
    UNASSIGNED: We conducted a genome-wide CRISPR screen to uncover essential regulators of cardiomyocyte specification in human embryonic stem cells using a myosin heavy chain 6 (MYH6)-GFP (green fluorescence protein) reporter system. After an independent secondary single guide ribonucleic acid validation of 25 candidates, we identified NF2 (neurofibromin 2), a moesin-ezrin-radixin like (MERLIN) tumor suppressor, as an upstream driver of early cardiomyocyte lineage specification. Independent monoclonal NF2 knockouts were generated using CRISPR-Cas9, and cell states were inferred through bulk RNA sequencing and protein expression analysis across differentiation time points. Terminal lineage differentiation was assessed by using an in vitro 2-dimensional-micropatterned gastruloid model, trilineage differentiation, and cardiomyocyte differentiation. Protein interaction and post-translation modification of NF2 with its interacting partners were assessed using site-directed mutagenesis, coimmunoprecipitation, and proximity ligation assays.
    UNASSIGNED: Transcriptional regulation and trajectory inference from NF2-null cells reveal the loss of cardiomyocyte identity and the acquisition of nonmesodermal identity. Sustained elevation of early mesoderm lineage repressor SOX2 and upregulation of late anticardiac regulators CDX2 and MSX1 in NF2 knockout cells reflect a necessary role for NF2 in removing regulatory roadblocks. Furthermore, we found that NF2 and AMOT (angiomotin) cooperatively bind to YAP (yes-associated protein) during mesendoderm formation, thereby preventing YAP activation, independent of canonical MST (mammalian sterile 20-like serine-threonine protein kinase)-LATS (large tumor suppressor serine-threonine protein kinase) signaling. Mechanistically, cardiomyocyte lineage identity was rescued by wild-type and NF2 serine-518 phosphomutants, but not NF2 FERM (ezrin-radixin-meosin homology protein) domain blue-box mutants, demonstrating that the critical FERM domain-dependent formation of the AMOT-NF2-YAP scaffold complex at the adherens junction is required for early cardiomyocyte lineage differentiation.
    UNASSIGNED: These results provide mechanistic insight into the essential role of NF2 during early epithelial-mesenchymal transition by sequestering the repressive effect of YAP and relieving regulatory roadblocks en route to cardiomyocytes.
    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
    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)

公众号