APPL1

APPL1
  • 文章类型: Journal Article
    背景:缺氧/复氧(H/R)诱导心肌细胞铁性凋亡,心肌缺血/再灌注损伤中的核心重塑事件。甲基转移酶样14(METTL14)是N6-甲基腺苷(m6A)修饰的作者。进行这项研究以破译METTL14在H/R诱导的心肌细胞铁性凋亡中的作用。
    方法:培养小鼠心肌细胞HL-1,并进行H/R处理。H/R处理后的铁死亡程度通过细胞计数试剂盒-8测定来评价,测定试剂盒(ROS/GSH/Fe2+),和蛋白质印迹(GPX4/ACSL4)。METTL14,pri-miR-146a-5p的细胞内表达,miR-146a-5p,或衔接蛋白磷酸酪氨酸与PH结构域和亮氨酸拉链1(APPL1)的相互作用通过实时定量聚合酶链反应或蛋白质印迹进行检查,通过m6A定量分析和RNA免疫沉淀来确定与DiGeorge临界区8(DGCR8)结合的pri-miR-146a-5p和m6A修饰的pri-miR-146a-5p的总m6A水平和表达。miR-146a-5p与APPL1的结合通过双荧光素酶测定来证明。
    结果:H/R处理诱导心肌细胞铁性凋亡(ROS增加,Fe2+,和ACSL4并降低GSH和GPX4)并上调METTL14表达。METTL14敲低减弱H/R诱导的心肌细胞铁凋亡。METTL14通过增加pri-miR-146a-5p上的m6A修饰来诱导DGCR8对pri-miR-146a-5p的识别,这促进pri-miR-146a-5p转化为miR-146a-5p并进一步抑制APPL1转录。miR-146a-5p上调或APPL1下调限制了METTL14下调对H/R诱导的心肌细胞铁性凋亡的抑制作用。
    结论:METTL14通过DGCR8识别和加工pri-miR-146a-5p促进miR-146a-5p表达,抑制APPL1转录并触发H/R诱导的心肌细胞铁凋亡。
    BACKGROUND: Hypoxia/reoxygenation (H/R) induces cardiomyocyte ferroptosis, a core remodeling event in myocardial ischemia/reperfusion injury. Methyltransferase-like 14 (METTL14) emerges as a writer of N6-methyladenosine (m6A) modification. This study was conducted to decipher the role of METTL14 in H/R-induced cardiomyocyte ferroptosis.
    METHODS: Mouse cardiomyocytes HL-1 were cultured and underwent H/R treatment. The degree of ferroptosis after H/R treatment was appraised by the cell counting kit-8 assay, assay kits (ROS/GSH/Fe2+), and Western blotting (GPX4/ACSL4). The intracellular expressions of METTL14, pri-miR-146a-5p, miR-146a-5p, or adaptor protein phosphotyrosine interacting with PH domain and leucine zipper 1 (APPL1) were examined by real-time quantitative polymerase chain reaction or Western blotting, with m6A quantification analysis and RNA immunoprecipitation to determine the total m6A level and the expression of pri-miR-146a-5p bound to DiGeorge critical region 8 (DGCR8) and m6A-modified pri-miR-146a-5p. The binding of miR-146a-5p to APPL1 was testified by the dual-luciferase assay.
    RESULTS: H/R treatment induced cardiomyocyte ferroptosis (increased ROS, Fe2+, and ACSL4 and decreased GSH and GPX4) and upregulated METTL14 expression. METTL14 knockdown attenuated H/R-induced cardiomyocyte ferroptosis. METTL14 induced the recognition of pri-miR-146a-5p by DGCR8 by increasing m6A modification on pri-miR-146a-5p, which promoted the conversion of pri-miR-146a-5p into miR-146a-5p and further repressed APPL1 transcription. miR-146a-5p upregulation or APPL1 downregulation limited the inhibitory effect of METTL14 downregulation on H/R-induced cardiomyocyte ferroptosis.
    CONCLUSIONS: METTL14 promoted miR-146a-5p expression through the recognition and processing of pri-miR-146a-5p by DGCR8, which repressed APPL1 transcription and triggered H/R-induced cardiomyocyte ferroptosis.
    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:骨骼肌是体内最大的组织,它影响运动,新陈代谢和体内平衡。骨骼肌发育包括成肌细胞增殖,融合和分化形成肌管,随后形成成熟的肌肉纤维。这个过程受到一系列分子网络的严格调控。越来越多的证据表明,非编码RNA,特别是microRNAs(miRNAs),在调节骨骼肌生长中起着至关重要的作用。这里,我们发现miR-668-3p在骨骼肌中高表达。
    方法:用miR-668-3p模拟物和/或抑制剂转染增殖和分化的C2C12细胞,并通过RT-qPCR和Western印迹分析评估其靶基因的mRNA和蛋白质水平。通过双荧光素酶测定证实miR-668-3p对Appl的靶向。通过C2C12细胞共转染验证miR-668-3p和Appl的相互依赖性。
    结果:我们的数据显示miR-668-3p可以抑制成肌细胞增殖和成肌分化。与PH结构域和亮氨酸拉链1相互作用的磷酸酪氨酸(Appl1)是miR-668-3p的靶基因,并通过激活p38MAPK通路促进成肌细胞增殖和分化。此外,miR-668-3p对成肌细胞增殖和成肌分化的抑制作用可以通过App1来挽救。
    结论:我们的结果表明miR-668-3p/Appl1/p38MAPK通路调节骨骼肌发育的新机制。
    BACKGROUND: Skeletal muscle is the largest tissue in the body, and it affects motion, metabolism and homeostasis. Skeletal muscle development comprises myoblast proliferation, fusion and differentiation to form myotubes, which subsequently form mature muscle fibres. This process is strictly regulated by a series of molecular networks. Increasing evidence has shown that noncoding RNAs, especially microRNAs (miRNAs), play vital roles in regulating skeletal muscle growth. Here, we showed that miR-668-3p is highly expressed in skeletal muscle.
    METHODS: Proliferating and differentiated C2C12 cells were transfected with miR-668-3p mimics and/or inhibitor, and the mRNA and protein levels of its target gene were evaluated by RT‒qPCR and Western blotting analysis. The targeting of Appl1 by miR-668-3p was confirmed by dual luciferase assay. The interdependence of miR-668-3p and Appl1 was verified by cotransfection of C2C12 cells.
    RESULTS: Our data reveal that miR-668-3p can inhibit myoblast proliferation and myogenic differentiation. Phosphotyrosine interacting with PH domain and leucine zipper 1 (Appl1) is a target gene of miR-668-3p, and it can promote myoblast proliferation and differentiation by activating the p38 MAPK pathway. Furthermore, the inhibitory effect of miR-668-3p on myoblast cell proliferation and myogenic differentiation could be rescued by Appl1.
    CONCLUSIONS: Our results indicate a new mechanism by which the miR-668-3p/Appl1/p38 MAPK pathway regulates skeletal muscle 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
    目的:定义细胞代谢和信号传导的调节因子对于设计肥胖和NAFLD/NASH的新治疗策略至关重要。E3泛素连接酶通过泛素化介导的蛋白质靶标调节来控制多种细胞功能,因此,它们的功能畸变与许多疾病有关。E3连接酶Ube4A与人类肥胖有关,炎症,和癌症。然而,它的体内功能是未知的,并且没有动物模型可用于研究这种新型蛋白质。
    方法:制作了一个Ube4A基因敲除(UKO)全身小鼠模型,并比较了高脂饮食(HFD)喂养的WT和UKO小鼠的各种代谢参数,在他们的肝脏里,脂肪组织,和血清。在HFD喂养的WT和UKO小鼠的肝脏样品中进行脂质组学和RNA-Seq研究。进行蛋白质组学研究以鉴定代谢中Ube4A的靶标。此外,鉴定了Ube4A调节代谢的机制。
    结果:尽管年轻人的体重和组成,食物喂养的WT和UKO小鼠相似,基因敲除表现为轻度高胰岛素血症和胰岛素抵抗.HFD喂养显著增加肥胖,高胰岛素血症,和UKO小鼠两性的胰岛素抵抗。HFD喂养的UKO小鼠的白色和棕色脂肪组织储库具有增加的胰岛素抵抗和炎症以及减少的能量代谢。此外,Ube4A缺失会加剧肝脏脂肪变性,炎症,和HFD喂养小鼠的肝损伤,肝细胞的脂质摄取和脂肪生成增加。急性胰岛素治疗导致饲喂UKO小鼠的肝脏和脂肪组织中胰岛素效应蛋白激酶Akt的激活受损。我们将Akt激活蛋白APPL1鉴定为Ube4A相互作用物。Akt和APPL1的K63连接的泛素化(K63-Ub),已知可促进胰岛素诱导的Akt激活,在UKO小鼠中受损。此外,Ube4AK63-泛素化Akt体外。
    结论:Ube4A是一种新型的肥胖调节因子,胰岛素抵抗,脂肪组织功能障碍和NAFLD,防止其下调可能会改善这些疾病。
    Defining the regulators of cell metabolism and signaling is essential to design new therapeutic strategies in obesity and NAFLD/NASH. E3 ubiquitin ligases control diverse cellular functions by ubiquitination-mediated regulation of protein targets, and thus their functional aberration is associated with many diseases. The E3 ligase Ube4A has been implicated in human obesity, inflammation, and cancer. However, its in vivo function is unknown, and no animal models are available to study this novel protein.
    A whole-body Ube4A knockout (UKO) mouse model was generated, and various metabolic parameters were compared in chow- and high fat diet (HFD)-fed WT and UKO mice, and in their liver, adipose tissue, and serum. Lipidomics and RNA-Seq studies were performed in the liver samples of HFD-fed WT and UKO mice. Proteomic studies were conducted to identify Ube4A\'s targets in metabolism. Furthermore, a mechanism by which Ube4A regulates metabolism was identified.
    Although the body weight and composition of young, chow-fed WT and UKO mice are similar, the knockouts exhibit mild hyperinsulinemia and insulin resistance. HFD feeding substantially augments obesity, hyperinsulinemia, and insulin resistance in both sexes of UKO mice. HFD-fed white and brown adipose tissue depots of UKO mice have increased insulin resistance and inflammation and reduced energy metabolism. Moreover, Ube4A deletion exacerbates hepatic steatosis, inflammation, and liver injury in HFD-fed mice with increased lipid uptake and lipogenesis in hepatocytes. Acute insulin treatment resulted in impaired activation of the insulin effector protein kinase Akt in liver and adipose tissue of chow-fed UKO mice. We identified the Akt activator protein APPL1 as a Ube4A interactor. The K63-linked ubiquitination (K63-Ub) of Akt and APPL1, known to facilitate insulin-induced Akt activation, is impaired in UKO mice. Furthermore, Ube4A K63-ubiquitinates Akt in vitro.
    Ube4A is a novel regulator of obesity, insulin resistance, adipose tissue dysfunction and NAFLD, and preventing its downregulation may ameliorate these diseases.
    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
    前列腺导管内癌(IDCP)的存在与晚期疾病和前列腺腺癌患者的不良预后相关,但准确可靠的疾病严重程度分期仍然具有挑战性.免疫组织化学(IHC)已用于克服评估IDCP形态学的问题,但是目前的标记在表征该病变的复杂生物学方面仅显示出有限的实用性。在一项对被诊断为IDCP的患者队列的回顾性研究中,我们利用IHC对前列腺癌根治性切除术切片和生物标志物组App1,Sortilin和Syndecan-1进行了分析,以解释不同的结构模式,并探讨IDCP发生于高级别浸润性前列腺腺癌逆行扩散的理论.CribriformIDCP显示出强烈的Appl1,Sortilin和Syndecan-1标记模式,而坚实的IDCP架构具有高强度的App1和Syndecan-1标签,但最低限度的Sortilin标签。值得注意的是,IDCP区域的生物标志物组的表达模式与邻近的浸润性前列腺腺癌相似,也与显示神经周围和血管侵犯的前列腺癌相当。TheApp1,Sortilin,IDCP中的Syndecan-1生物标记物组为浸润性前列腺癌逆行扩散到导管/腺泡的模型提供了证据,并支持将IDCP纳入五层Gleason分级系统。
    The presence of intraductal carcinoma of the prostate (IDCP) correlates with late-stage disease and poor outcomes for patients with prostatic adenocarcinoma, but the accurate and reliable staging of disease severity remains challenging. Immunohistochemistry (IHC) has been utilised to overcome problems in assessing IDCP morphology, but the current markers have only demonstrated limited utility in characterising the complex biology of this lesion. In a retrospective study of a cohort of patients who had been diagnosed with IDCP, we utilised IHC on radical prostatectomy sections with a biomarker panel of Appl1, Sortilin and Syndecan-1, to interpret different architectural patterns and to explore the theory that IDCP occurs from retrograde spread of high-grade invasive prostatic adenocarcinoma. Cribriform IDCP displayed strong Appl1, Sortilin and Syndecan-1 labelling patterns, while solid IDCP architecture had high intensity Appl1 and Syndecan-1 labelling, but minimal Sortilin labelling. Notably, the expression pattern of the biomarker panel in regions of IDCP was similar to that of adjacent invasive prostatic adenocarcinoma, and also comparable to prostate cancer showing perineural and vascular invasion. The Appl1, Sortilin, and Syndecan-1 biomarker panel in IDCP provides evidence for the model of retrograde spread of invasive prostatic carcinoma into ducts/acini, and supports the inclusion of IDCP into the five-tier Gleason grading system.
    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
    人骨髓间充质干细胞(MSCs)成脂和成骨分化失衡在骨质疏松症的发病机制中起着重要作用。我们之前的研究证实了衔接子蛋白,磷酸酪氨酸与PH结构域和亮氨酸拉链1(APPL1)相互作用/肌铁蛋白缺乏通过阻断骨质疏松中的自噬通量促进MSCs的成脂分化。然而,APPL1在MSCs成骨分化中的作用尚不清楚。本研究旨在探讨APPL1在骨质疏松骨髓间充质干细胞成骨分化中的作用及其调控机制。在这项研究中,我们证明了APPL1在骨质疏松症患者和骨质疏松症小鼠中的表达下调。临床骨质疏松的严重程度与骨髓间充质干细胞APPL1的表达呈负相关。我们发现APPL1在体外和体内正向调节MSCs的成骨分化。此外,RNA测序显示MGP的表达,骨钙蛋白/基质Gla家族成员,APPL1敲低后显著上调。机械上,我们的研究表明,减少的APPL1通过促进基质Gla蛋白表达来破坏骨质疏松中的BMP2通路,从而损害间充质干细胞的成骨分化。我们还评估了APPL1在骨质疏松症小鼠模型中促进成骨的意义。这些结果表明APPL1可能是诊断和治疗骨质疏松症的重要靶点。
    An imbalance of human mesenchymal stem cells (MSCs) adipogenic and osteogenic differentiation plays an important role in the pathogenesis of osteoporosis. Our previous study verified that Adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1 (APPL1)/myoferlin deficiency promotes adipogenic differentiation of MSCs by blocking autophagic flux in osteoporosis. However, the function of APPL1 in the osteogenic differentiation of MSCs remains unclear. This study aimed to investigate the role of APPL1 in the osteogenic differentiation of MSCs in osteoporosis and the underlying regulatory mechanism. In this study, we demonstrated the downregulation of APPL1 expression in patients with osteoporosis and osteoporosis mice. The severity of clinical osteoporosis was negatively correlated with the expression of APPL1 in bone marrow MSCs. We found that APPL1 positively regulates the osteogenic differentiation of MSCs in vitro and in vivo. Moreover, RNA sequencing showed that the expression of MGP, an osteocalcin/matrix Gla family member, was significantly upregulated after APPL1 knockdown. Mechanistically, our study showed that reduced APPL1 impaired the osteogenic differentiation of mesenchymal stem cells by facilitating Matrix Gla protein expression to disrupt the BMP2 pathway in osteoporosis. We also evaluated the significance of APPL1 in promoting osteogenesis in a mouse model of osteoporosis. These results suggest that APPL1 may be an important target for the diagnosis and treatment of osteoporosis.
    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:腹内侧前额叶皮层(vmPFC)已被视为存储和回忆灭绝记忆的场所。然而,这个过程背后的突触和细胞机制仍然难以捉摸。
    方法:我们结合转基因小鼠,电生理记录,活性依赖性细胞标记,和化学遗传学操作来分析衔接蛋白APPL1在vmPFC中的作用,以进行恐惧灭绝检索。
    结果:我们发现组成型和条件性APPL1敲除都会降低vmPFC中NMDA受体(NMDAR)的功能,并损害恐惧消退的恢复。此外,APPL1在灭绝恢复过程中经历了核移位。阻断APPL1核质易位可减少NMDAR电流并破坏灭绝恢复。我们进一步确定了前额叶神经元集合,对于存储灭绝记忆既必要又足够。该集合中的诱导型APPL1敲除消除了NMDAR依赖性突触增强作用,并破坏了灭绝恢复,同时,这种合奏的化学遗传激活可以挽救受损的行为。
    结论:因此,我们的结果表明,前额叶神经元集合存储灭绝记忆,和APPL1信号传导支持这些神经元通过控制NMDAR依赖性增强来检索灭绝记忆。
    The ventromedial prefrontal cortex has been viewed as a locus for storage and recall of extinction memory. However, the synaptic and cellular mechanisms underlying these processes remain elusive.
    We combined transgenic mice, electrophysiological recording, activity-dependent cell labeling, and chemogenetic manipulation to analyze the role of adaptor protein APPL1 in the ventromedial prefrontal cortex in fear extinction retrieval.
    We found that both constitutive and conditional APPL1 knockout decreased NMDA receptor (NMDAR) function in the ventromedial prefrontal cortex and impaired fear extinction retrieval. Moreover, APPL1 undergoes nuclear translocation during extinction retrieval. Blocking APPL1 nucleocytoplasmic translocation reduced NMDAR currents and disrupted extinction retrieval. We also identified a prefrontal neuronal ensemble that is both necessary and sufficient for the storage of extinction memory. Inducible APPL1 knockout in this ensemble abolished NMDAR-dependent synaptic potentiation and disrupted extinction retrieval, while chemogenetic activation of this ensemble simultaneously rescued the impaired behaviors.
    Our results indicate that a prefrontal neuronal ensemble stores extinction memory, and APPL1 signaling supports these neurons in retrieving extinction memory by controlling NMDAR-dependent potentiation.
    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
    已经证明APPL1(衔接蛋白,与PH结构域和亮氨酸拉链1相互作用的磷酸酪氨酸)参与了几种与生长相关的信号传导途径的调节,因此与某些癌症的发展和进展密切相关。二烯丙基三硫(DAT),一种来源于大蒜的生物活性化合物,通过干扰促生存信号通路对各种人类癌细胞发挥选择性细胞毒性。然而,DAT是否以及如何影响人肝细胞癌(HCC)细胞的存活仍不清楚。在这里,我们检验了APPL1参与DAT诱导的HCCHepG2细胞毒性的假设。我们发现,在DAT处理的HepG2细胞中,APPL1的Lys63(K63)连接的多泛素化显着降低,而丝氨酸残基上APPL1的磷酸化保持不变。与野生型APPL1相比,APPL1K63R突变体的过表达显著增加了细胞凋亡,减轻了细胞存活,随着STAT3,Akt,Erk1/2此外,DAT施用显著降低细胞内TNF受体相关因子6(TRAF6)的蛋白质水平。TRAF6的遗传抑制降低了APPL1的K63连接的聚泛素化。此外,野生型APPL1的过表达大大减弱了DAT对HepG2细胞的细胞毒性影响。一起来看,这些结果表明,APPL1聚泛素化可能通过调节STAT3、Akt、和Erk1/2通路。
    It has been demonstrated that APPL1 (adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1) is involved in the regulation of several growth-related signaling pathways and thus closely associated with the development and progression of some cancers. Diallyl trisulfide (DAT), a garlic-derived bioactive compound, exerts selective cytotoxicity to various human cancer cells through interfering with pro-survival signaling pathways. However, whether and how DAT affects survival of human hepatocellular carcinoma (HCC) cells remain unclear. Herein, we tested the hypothesis of the involvement of APPL1 in DAT-induced cytotoxicity in HCC HepG2 cells. We found that Lys 63 (K63)-linked polyubiquitination of APPL1 was significantly decreased whereas phosphorylation of APPL1 at serine residues remained unchanged in DAT-treated HepG2 cells. Compared with wild-type APPL1, overexpression of APPL1 K63R mutant dramatically increased cell apoptosis and mitigated cell survival, along with a reduction of phosphorylation of STAT3, Akt, and Erk1/2. In addition, DAT administration markedly reduced protein levels of intracellular TNF receptor-associated factor 6 (TRAF6). Genetic inhibition of TRAF6 decreased K63-linked polyubiquitination of APPL1. Moreover, the cytotoxicity impacts of DAT on HepG2 cells were greatly attenuated by overexpression of wild-type APPL1. Taken together, these results suggest that APPL1 polyubiquitination probably mediates the inhibitory effects of DAT on survival of HepG2 cells by modulating STAT3, Akt, and Erk1/2 pathways.
    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
    前列腺癌患者的诊断和评估取决于组织病理学的准确解释和分级。然而,形态学不一定反映前列腺癌疾病进展中发生的复杂生物学变化,和目前的生物标志物在患者评估中的临床应用有限。这项研究旨在通过区分特定的病理特征来开发准确定义前列腺癌生物学的生物标志物,从而为患者的准确Gleason分级提供可靠的病理学解释。询问在线基因表达数据库并鉴定前列腺癌的致病途径。该途径中关键基因的蛋白质表达,包括含有pleckstrin同源(PH)结构域的衔接蛋白,磷酸酪氨酸结合(PTB)结构域,和亮氨酸拉链基序1(App1),在29例前列腺癌患者的初步研究中,通过免疫组织化学(IHC)检查了Sortilin和Syndecan-1,使用针对独特表位设计的单克隆抗体。App1,Sortilin,首先在112例患者样本的组织微阵列队列中评估了Syndecan-1的表达,证明单克隆抗体清楚地说明了腺体的形态。为了确定一种新颖的IHC辅助解释的影响(Appl1,Sortilin,和Syndecan-1标记为Gleason分级的面板),与标准苏木精和曙红(H&E)格里森等级分配相比,对包含114例患者的根治性前列腺切除术样本队列进行了评估.与H&E相比,生物标志物组的实用性降低了前列腺癌组织形态学解释的主观性,提高了病理学评估的可靠性,导致41%的患者样本的Gleason等级重新分布。重要的是,对于模棱两可的IHC辅助标记和H&E染色结果,在重新审查H&E组织切片时,可以更准确地应用癌症形态学解释.这项研究通过提出三种生物标志物的新型组合来解决前列腺癌病理学领域的关键问题,并具有通过标准化组织形态学解释来改变临床病理学实践的潜力。
    Diagnosis and assessment of patients with prostate cancer is dependent on accurate interpretation and grading of histopathology. However, morphology does not necessarily reflect the complex biological changes occurring in prostate cancer disease progression, and current biomarkers have demonstrated limited clinical utility in patient assessment. This study aimed to develop biomarkers that accurately define prostate cancer biology by distinguishing specific pathological features that enable reliable interpretation of pathology for accurate Gleason grading of patients. Online gene expression databases were interrogated and a pathogenic pathway for prostate cancer was identified. The protein expression of key genes in the pathway, including adaptor protein containing a pleckstrin homology (PH) domain, phosphotyrosine-binding (PTB) domain, and leucine zipper motif 1 (Appl1), Sortilin and Syndecan-1, was examined by immunohistochemistry (IHC) in a pilot study of 29 patients with prostate cancer, using monoclonal antibodies designed against unique epitopes. Appl1, Sortilin, and Syndecan-1 expression was first assessed in a tissue microarray cohort of 112 patient samples, demonstrating that the monoclonal antibodies clearly illustrate gland morphologies. To determine the impact of a novel IHC-assisted interpretation (the utility of Appl1, Sortilin, and Syndecan-1 labelling as a panel) of Gleason grading, versus standard haematoxylin and eosin (H&E) Gleason grade assignment, a radical prostatectomy sample cohort comprising 114 patients was assessed. In comparison to H&E, the utility of the biomarker panel reduced subjectivity in interpretation of prostate cancer tissue morphology and improved the reliability of pathology assessment, resulting in Gleason grade redistribution for 41% of patient samples. Importantly, for equivocal IHC-assisted labelling and H&E staining results, the cancer morphology interpretation could be more accurately applied upon re-review of the H&E tissue sections. This study addresses a key issue in the field of prostate cancer pathology by presenting a novel combination of three biomarkers and has the potential to transform clinical pathology practice by standardising the interpretation of the tissue morphology.
    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
    人骨髓间充质干细胞(hMSCs)成脂和成骨分化的失衡在骨质疏松的发病机制中至关重要。迫切需要阐明潜在的机制。APPL1,脂联素受体的衔接蛋白,最近被证明与骨量密切相关。然而,APPL1在骨质疏松hMSC分化失衡中的作用尚不清楚.因此,我们旨在探讨APPL1改变骨质疏松中hMSCs成脂分化的机制.这里,我们发现APPL1在老年骨质疏松症患者和小鼠骨质疏松症模型中表达下调。APPL1在体内和体外负调控hMSC成脂分化。机械上,通过增强泛素化介导的Myoferlin降解,下调APPL1表达增加了hMSCs成脂分化过程中溶酶体功能障碍的风险。溶酶体功能障碍通过抑制自噬体降解抑制自噬通量并促进hMSC向脂肪细胞系分化。我们的发现表明APPL1/Myoferlin下调通过抑制自噬通量促进hMSCs成脂分化,进一步损害骨质疏松症中hMSCs成脂和成骨分化的平衡;APPL1/Myoferlin轴可能是骨质疏松症的有希望的诊断和治疗靶标。
    An imbalance of human mesenchymal stem cells (hMSCs) adipogenic and osteogenic differentiation is crucial in the pathogenesis of osteoporosis, and elucidation of the underlying mechanism is urgently needed. APPL1, an adaptor protein of the adiponectin receptor, was recently shown to be closely related to bone mass. However, the role of APPL1 in the imbalance of hMSC differentiation in osteoporosis is unclear. Therefore, we aimed to explore the mechanisms by which APPL1 alters hMSCs adipogenic differentiation in osteoporosis. Here, we found that APPL1 expression was downregulated in elderly patients with osteoporosis and in mouse osteoporosis model. APPL1 negatively regulated hMSC adipogenic differentiation in vivo and in vitro. Mechanistically, by enhancing ubiquitination-mediated Myoferlin degradation, downregulated APPL1 expression increased the risk of lysosome dysfunction during hMSCs adipogenic differentiation. Lysosomal dysfunction inhibited autophagy flux by suppressing autophagosome degradation and promoted hMSC differentiation towards the adipocyte lineage. Our findings suggest that APPL1/Myoferlin downregulation promoted hMSCs adipogenic differentiation by inhibiting autophagy flux, further impairing the balance of hMSCs adipogenic and osteogenic differentiation in osteoporosis; the APPL1/ Myoferlin axis may be a promising diagnostic and therapeutic target for osteoporosis.
    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:转化生长因子β(TGFβ)在几种晚期癌症类型中过表达并促进肿瘤进展。我们已经报道了TGFβ受体(TβR)I的胞内结构域(ICD)被癌细胞中的蛋白水解酶切割,然后以依赖于内体衔接蛋白APPL1/2的方式易位到细胞核,驱动侵袭程序。癌细胞如何逃避TGFβ诱导的生长抑制尚不清楚。
    方法:我们进行了微阵列分析,以在去势抵抗的前列腺癌(CRPC)细胞中搜索APPL1/2蛋白调节的基因。我们研究了在不存在外源性TGFβ的情况下,在10%FBS中培养的癌细胞系中TβRI和TRAF6在有丝分裂中的作用。还研究了TRAF6在有丝分裂中AURKB泛素化的分子机制以及癌细胞系和组织微阵列中AURKB-TβRI复合物的形成。
    结果:在有丝分裂和胞质分裂过程中,在CRPC和KELLY神经母细胞瘤细胞的中体中形成AURKB-TβRI复合物。TRAF6诱导AURKB在K85和K87上的多泛素化,在AURKB表面突出以促进其激活。患者肿瘤组织切片中的AURKB-TβRI复合物与前列腺癌的恶性程度相关。
    结论:AURKB-TβRI复合物可能成为有发展为侵袭性PC风险的患者的预后生物标志物。
    背景:瑞典医学研究理事会(2019-01598,ML;2015-02757和2020-01291,CHH),瑞典癌症协会(200964,ML),于默奥大学与Västerbotten地区之间的区域协议(ALF;RV-939377,-967041,-970057,ML)。欧洲研究理事会(787472,CHH)。KAW2019.0345,和肯佩基金会SMK-1866;ML。国家显微镜基础设施(NMIVR-RFI2016-00968)。
    BACKGROUND: Transforming growth factor β (TGFβ) is overexpressed in several advanced cancer types and promotes tumor progression. We have reported that the intracellular domain (ICD) of TGFβ receptor (TβR) I is cleaved by proteolytic enzymes in cancer cells, and then translocated to the nucleus in a manner dependent on the endosomal adaptor proteins APPL1/2, driving an invasiveness program. How cancer cells evade TGFβ-induced growth inhibition is unclear.
    METHODS: We performed microarray analysis to search for genes regulated by APPL1/2 proteins in castration-resistant prostate cancer (CRPC) cells. We investigated the role of TβRI and TRAF6 in mitosis in cancer cell lines cultured in 10% FBS in the absence of exogenous TGFβ. The molecular mechanism of the ubiquitination of AURKB by TRAF6 in mitosis and the formation of AURKB-TβRI complex in cancer cell lines and tissue microarrays was also studied.
    RESULTS: During mitosis and cytokinesis, AURKB-TβRI complexes formed in midbodies in CRPC and KELLY neuroblastoma cells. TRAF6 induced polyubiquitination of AURKB on K85 and K87, protruding on the surface of AURKB to facilitate its activation. AURKB-TβRI complexes in patient\'s tumor tissue sections correlated with the malignancy of prostate cancer.
    CONCLUSIONS: The AURKB-TβRI complex may become a prognostic biomarker for patients with risk of developing aggressive PC.
    BACKGROUND: Swedish Medical Research Council (2019-01598, ML; 2015-02757 and 2020-01291, CHH), the Swedish Cancer Society (20 0964, ML), a regional agreement between Umeå University and Region Västerbotten (ALF; RV-939377, -967041, -970057, ML). The European Research Council (787472, CHH). KAW 2019.0345, and the Kempe Foundation SMK-1866; ML. National Microscopy Infrastructure (NMI VR-RFI 2016-00968).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号