Fbxo28

Fbxo28
  • 文章类型: Journal Article
    背景:由于缺乏早期症状,卵巢癌是最常见的妇科恶性肿瘤之一,早期诊断和有限筛查。因此,了解卵巢癌发生和发展的分子机制,确定卵巢癌的早期诊断和临床治疗的基础生物标志物是十分必要的。
    方法:使用Kaplan-Meier生存分析分析FBXO28与卵巢癌预后之间的关系。从癌症基因组图谱(TCGA)获得正常卵巢组织和卵巢肿瘤组织之间FBXO28mRNA表达的差异,和基因型-组织表达(GTEx)队列。免疫组化法检测FBXO28蛋白在卵巢癌组织和正常卵巢组织中的表达。使用蛋白质印迹法测定卵巢癌细胞中FBXO28的表达水平。CCK-8,菌落形成,进行Transwell迁移和侵袭测定以评估细胞增殖和运动性。
    结果:我们发现,在卵巢癌患者中,较高的FBXO28表达水平与不良预后相关。对TCGA和GTEx队列的分析表明,正常卵巢组织样品中的FBXO28mRNA水平低于卵巢癌组织样品。与正常卵巢组织或细胞系相比,FBXO28在卵巢肿瘤组织或肿瘤细胞中表达较年夜。FBXO28的上调促进了生存能力,扩散,卵巢癌细胞的迁移和侵袭。最后,我们证明FBXO28激活了卵巢癌中TGF-beta1/Smad2/3信号通路。
    结论:结论:FBXO28通过上调卵巢癌中TGF-β1/Smad2/3信号通路增强致癌功能.
    BACKGROUND: Ovarian cancer is one of the most common gynecological malignancies due to the lack of early symptoms, early diagnosis and limited screening. Therefore, it is necessary to understand the molecular mechanism underlying the occurrence and progression of ovarian cancer and to identify a basic biomarker for the early diagnosis and clinical treatment of ovarian cancer.
    METHODS: The association between FBXO28 and ovarian cancer prognosis was analyzed using Kaplan‒Meier survival analysis. The difference in FBXO28 mRNA expression between normal ovarian tissues and ovarian tumor tissues was obtained from The Cancer Genome Atlas (TCGA), and Genotype-Tissue Expression (GTEx) cohorts. The expression levels of the FBXO28 protein in ovarian cancer tissues and normal ovarian tissues were measured via immunohistochemical staining. Western blotting was used to determine the level of FBXO28 expression in ovarian cancer cells. The CCK-8, the colony formation, Transwell migration and invasion assays were performed to evaluate cell proliferation and motility.
    RESULTS: We found that a higher expression level of FBXO28 was associated with poor prognosis in ovarian cancer patients. Analysis of the TCGA and GTEx cohorts showed that the FBXO28 mRNA level was lower in normal ovarian tissue samples than in ovarian cancer tissue samples. Compared with that in normal ovarian tissues or cell lines, the expression of FBXO28 was greater in ovarian tumor tissues or tumor cells. The upregulation of FBXO28 promoted the viability, proliferation, migration and invasion of ovarian cancer cells. Finally, we demonstrated that FBXO28 activated the TGF-beta1/Smad2/3 signaling pathway in ovarian cancer.
    CONCLUSIONS: In conclusion, FBXO28 enhanced oncogenic function via upregulation of the TGF-beta1/Smad2/3 signaling pathway in ovarian cancer.
    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
    染色体1q42区域的结构缺失很少。与这种拷贝数变化相关的表型谱是可变的,包括发育迟缓,智力残疾,癫痫发作,和畸形学。这项研究描述了一名患有发育迟缓和大脑异常的患者。G带核型,FISH,SNP寡核苷酸微阵列分析(SOMA),并进行全外显子组测序分析。产前SOMA的产后再分析和后续父母测试显示,患者在1q42.11时父系遗传63kb缺失。我们描述了这个病人的临床特征,提供与1q42.11子带缺失相关的临床表型的洞察。我们的研究提供了新的证据支持FBXO283'UTR区的潜在功能重要性以及FBXO28是染色体1q41q42微缺失综合征发病机理中的关键基因的假设。它还强调了产前和产后微阵列测试之间的不同目标和报告标准。
    Constitutional deletions of chromosome 1q42 region are rare. The phenotype spectrum associated with this copy number change is variable, including developmental delay, intellectual disability, seizures, and dysmorphology. This study describes a patient with developmental delays and brain abnormalities. G-banded karyotype, FISH, SNP oligonucleotide microarray analysis (SOMA), and whole exome sequencing analysis were performed. Postnatal reanalysis of prenatal SOMA and follow-up parental testing revealed a paternally inherited 63 kb deletion at 1q42.11 in the patient. We characterized the clinical features of this patient, providing insight into the clinical phenotype associated with deletions of the 1q42.11 sub-band. Our study provides new evidence supporting the potential functional importance of the FBXO28 3\' UTR region and the hypothesis that FBXO28 is a critical gene in the pathogenesis of chromosome 1q41q42 microdeletion syndrome. It also highlights the different goals and reporting criteria between prenatal and postnatal microarray tests.
    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
    E3连接酶仅F-box蛋白28(FBXO28)属于在肿瘤发展中起关键作用的F-box蛋白家族。然而,FBXO28在胰腺癌(PC)中的潜在功能及其分子机制尚不清楚。在这项研究中,我们研究了FBXO28在PC中的表达及其生物学作用,并探讨了FBXO28介导的增殖机制,入侵,和PC细胞的转移。与癌旁组织和人正常胰腺导管上皮细胞相比,FBXO28在PC组织和细胞系中高表达。FBXO28的高表达与PC患者的生存预后呈负相关。功能试验表明FBXO28促进PC细胞增殖,入侵,和体内外转移。此外,免疫共沉淀-质谱鉴定SMARCC2为FBXO28的靶标;上调SMARCC2可以逆转FBXO28过表达的促增殖作用,入侵,和PC细胞的转移。机械上,FBXO28通过增加SMARCC2泛素化和蛋白质降解抑制翻译后SMARCC2表达。总之,FBXO28在PC中具有潜在作用,可能通过SMARCC2泛素化促进PC进展。因此,FBXO28可能是PC的潜在治疗靶标。
    The E3 ligase F-box only protein 28 (FBXO28) belongs to the F-box family of proteins that play a critical role in tumor development. However, the potential function of FBXO28 in pancreatic cancer (PC) and its molecular mechanism remain unclear. In this study, we examined FBXO28 expression in PC and its biological role and explored the mechanism of FBXO28-mediated proliferation, invasion, and metastasis of PC cells. Compared with paracancerous tissues and human normal pancreatic ductal epithelial cells, FBXO28 was highly expressed in PC tissues and cell lines. High expression of FBXO28 was negatively correlated with the survival prognosis of patients with PC. Functional assays indicated that FBXO28 promoted PC cell proliferation, invasion, and metastasis in vitro and in vivo. Furthermore, immunoprecipitation-mass spectrometry was used to identify SMARCC2 as the target of FBXO28; upregulation of SMARCC2 can reverse the effect of overexpression of FBXO28 on promoting the proliferation, invasion, and metastasis of PC cells. Mechanistically, FBXO28 inhibited SMARCC2 expression in post-translation by increasing SMARCC2 ubiquitination and protein degradation. In conclusion, FBXO28 has a potential role in PC, possibly promoting PC progression through SMARCC2 ubiquitination. Thus, FBXO28 might be a potential treatment target in PC.
    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
    F-box蛋白是SCF(SKP1/CUL1/F-box)E3泛素连接酶复合物的底物识别亚基,一种多组分RING型E3连接酶,通过靶向泛素化的关键调节蛋白参与许多细胞过程的调节。然而,是否以及如何调节F-box蛋白在很大程度上是未知的。在这里,我们报道了FBXO28,一种表征不佳的F-box蛋白,是SCFE3连接酶的新型底物。SCF活化所需的neddylation途径的药物或遗传抑制稳定FBXO28并延长其半衰期。同时,FBXO28经受泛素化,并且基于cullin1的SCF复合物促进FBXO28降解。此外,F-box结构域的缺失稳定了FBXO28,而内源性FBXO28的敲低强烈上调了外源性FBXO28的表达。一起来看,这些数据表明,SCFFBXO28是负责FBXO28自身泛素化和蛋白酶体降解的E3连接酶,为F-box蛋白的上游信号调节提供了新的线索.
    The F-box protein is the substrate recognition subunit of SCF (SKP1/CUL1/F-box) E3 ubiquitin ligase complex, a multicomponent RING-type E3 ligase involved in the regulation of numerous cellular processes by targeting critical regulatory proteins for ubiquitination. However, whether and how F-box proteins are regulated is largely unknown. Here we report that FBXO28, a poorly characterized F-box protein, is a novel substrate of SCF E3 ligase. Pharmaceutical or genetic inhibition of neddylation pathway that is required for the activation of SCF stabilizes FBXO28 and prolongs its half-life. Meanwhile, FBXO28 is subjected to ubiquitination and cullin1-based SCF complex promotes FBXO28 degradation. Moreover, deletion of F-box domain stabilizes FBXO28 and knockdown of endogenous FBXO28 strongly upregulates exogenous FBXO28 expression. Taken together, these data reveal that SCFFBXO28 is the E3 ligase responsible for the self-ubiquitination and proteasomal degradation of FBXO28, providing a new clue for the upstream signaling regulation for F-box proteins.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Case Reports
    最近,染色体1q41-q42缺失与儿童早期可识别的神经发育综合征(OMIM612530)有关。在这个群体中,发育迟缓(DD)的主要表型,智力残疾(ID),癫痫,独特的形态学,磁共振成像/计算机断层扫描的大脑异常已经出现。先前关于在1q41-q42处有从头缺失的患者的报道已导致鉴定出一个进化的最小重叠区域,该区域包括几个潜在的致病基因,包括DISP1,TP53BP2和FBXO28。在最近的一份报告中,我们描述了一组WDR26中具有从头突变的患者,这些患者具有最初在1q41-q42微缺失综合征(MDS)中描述的许多临床特征.这里,我们描述了一个患有难治性癫痫的3岁女孩的新的种系FBXO28移码突变,ID,DD,以及与1q41-q42MDS重叠的其他功能。通过一项家族性全外显子组测序研究,我们确定了从头FBXO28c.972_973delACinsG(p。Arg325GlufsX3)先证者中的移码突变。移码和导致的过早无义突变尚未在任何基因组数据库中报道。这个孩子没有一个大的1q41-q42删除,她也没有携带WDR26突变。我们的病例与先前报道的患者一起,FBXO28也受到影响,但WDR26没有受到影响。这些发现支持以下观点:FBXO28是单基因疾病基因,并有助于1q41-q42基因缺失综合征的复杂神经发育表型。
    Chromosome 1q41-q42 deletions have recently been associated with a recognizable neurodevelopmental syndrome of early childhood (OMIM 612530). Within this group, a predominant phenotype of developmental delay (DD), intellectual disability (ID), epilepsy, distinct dysmorphology, and brain anomalies on magnetic resonance imaging/computed tomography has emerged. Previous reports of patients with de novo deletions at 1q41-q42 have led to the identification of an evolving smallest region of overlap which has included several potentially causal genes including DISP1, TP53BP2, and FBXO28. In a recent report, a cohort of patients with de novo mutations in WDR26 was described that shared many of the clinical features originally described in the 1q41-q42 microdeletion syndrome (MDS). Here, we describe a novel germline FBXO28 frameshift mutation in a 3-year-old girl with intractable epilepsy, ID, DD, and other features which overlap those of the 1q41-q42 MDS. Through a familial whole-exome sequencing study, we identified a de novo FBXO28 c.972_973delACinsG (p.Arg325GlufsX3) frameshift mutation in the proband. The frameshift and resulting premature nonsense mutation have not been reported in any genomic database. This child does not have a large 1q41-q42 deletion, nor does she harbor a WDR26 mutation. Our case joins a previously reported patient also in whom FBXO28 was affected but WDR26 was not. These findings support the idea that FBXO28 is a monogenic disease gene and contributes to the complex neurodevelopmental phenotype of the 1q41-q42 gene deletion syndrome.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Loss of pancreatic β-cell function and/or mass is a central hallmark of all forms of diabetes but its molecular basis is incompletely understood. β-cell apoptosis contributes to the reduced β-cell mass in diabetes. Therefore, the identification of important signaling molecules that promote β-cell survival in diabetes could lead to a promising therapeutic intervention to block β-cell decline during development and progression of diabetes. In the present study, we identified F-box protein 28 (FBXO28), a substrate-recruiting component of the Skp1-Cul1-F-box (SCF) ligase complex, as a regulator of pancreatic β-cell survival. FBXO28 was down-regulated in β-cells and in isolated human islets under diabetic conditions. Consistently, genetic silencing of FBXO28 impaired β-cell survival, and restoration of FBXO28 protected β-cells from the harmful effects of the diabetic milieu. Although FBXO28 expression positively correlated with β-cell transcription factor NEUROD1 and FBXO28 depletion also reduced insulin mRNA expression, neither FBXO28 overexpression nor depletion had any significant impact on insulin content, glucose-stimulated insulin secretion (GSIS) or on other genes involved in glucose sensing and metabolism or on important β-cell transcription factors in isolated human islets. Consistently, FBXO28 overexpression did not further alter insulin content and GSIS in freshly isolated islets from patients with type 2 diabetes (T2D). Our data show that FBXO28 improves pancreatic β-cell survival under diabetogenic conditions without affecting insulin secretion, and its restoration may be a novel therapeutic tool to promote β-cell survival in diabetes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    SCF (Skp1/Cul1/F-box) ubiquitin ligases act as master regulators of cellular homeostasis by targeting key proteins for ubiquitylation. Here, we identified a hitherto uncharacterized F-box protein, FBXO28 that controls MYC-dependent transcription by non-proteolytic ubiquitylation. SCF(FBXO28) activity and stability are regulated during the cell cycle by CDK1/2-mediated phosphorylation of FBXO28, which is required for its efficient ubiquitylation of MYC and downsteam enhancement of the MYC pathway. Depletion of FBXO28 or overexpression of an F-box mutant unable to support MYC ubiquitylation results in an impairment of MYC-driven transcription, transformation and tumourigenesis. Finally, in human breast cancer, high FBXO28 expression and phosphorylation are strong and independent predictors of poor outcome. In conclusion, our data suggest that SCF(FBXO28) plays an important role in transmitting CDK activity to MYC function during the cell cycle, emphasizing the CDK-FBXO28-MYC axis as a potential molecular drug target in MYC-driven cancers, including breast cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号