CPEB2

CPEB2
  • 文章类型: Journal Article
    背景:突触的局部翻译对于快速重塑突触蛋白质组以维持长期可塑性和记忆很重要。虽然记忆相关的局部翻译的调节机制已在突触后/树突区域得到广泛阐明,没有直接证据表明轴突中的RNA结合蛋白(RBP)控制靶特异性mRNA的翻译,从而促进长时程增强(LTP)和记忆.我们先前报道,由胞质聚腺苷酸化元件结合蛋白2(CPEB2)控制的翻译对于突触后可塑性和记忆很重要。这里,我们调查了CPEB2是否调节轴突平移以支持突触前可塑性。
    方法:在具有CPEB2的泛神经元/神经胶质细胞或谷氨酸能神经元特异性敲除的小鼠中进行行为和电生理学评估。电记录海马Schaffer侧支(SC)-CA1和颞氨(TA)-CA1途径,以监测4列高频刺激引起的突触传递和LTP。RNA免疫沉淀,结合生物信息学分析,用于揭示与学习相关的CPEB2结合轴突RNA候选物,通过Western印迹和荧光素酶报告基因检测进一步验证。将表达Cre重组酶的腺相关病毒立体定向递送至TA回路的突触前或突触后区域以消融Cpeb2用于进一步的电生理研究。在微流体平台上培养的生化分离的突触小体和轴突化神经元用于测量轴突蛋白合成和FM4-64FX负载的突触小泡。
    结果:海马CA1神经元的电生理分析检测到CPEB2耗尽的SC和TA传入的异常兴奋性和囊泡释放概率,因此,我们将CPEB2免疫沉淀的转录组与成人皮质中学习诱导的轴突翻译组交叉比较,以鉴定可能受CPEB2调节的轴突靶标.我们验证了Slc17a6,编码囊泡谷氨酸转运蛋白2(VGLUT2),由CPEB2翻译上调。在表达VGLUT2的谷氨酸能神经元中CPEB2的条件性敲除会损害小鼠海马依赖性记忆的巩固。在VGLUT2主导的TA传入中,突触前特异性的Cpeb2消融足以减弱蛋白质合成依赖性LTP。此外,CPEB2缺乏症或环己酰亚胺阻断活性诱导的轴突Slc17a6翻译减少了含VGLUT2的突触小泡的可释放池。
    结论:我们确定了272个CPEB2结合转录本,其轴突翻译在学习后发生改变,并在CPEB2驱动的轴突合成VGLUT2和突触前翻译依赖性LTP之间建立了因果关系。这些发现扩展了我们对突触前室中与记忆相关的翻译控制机制的理解。
    BACKGROUND: Local translation at synapses is important for rapidly remodeling the synaptic proteome to sustain long-term plasticity and memory. While the regulatory mechanisms underlying memory-associated local translation have been widely elucidated in the postsynaptic/dendritic region, there is no direct evidence for which RNA-binding protein (RBP) in axons controls target-specific mRNA translation to promote long-term potentiation (LTP) and memory. We previously reported that translation controlled by cytoplasmic polyadenylation element binding protein 2 (CPEB2) is important for postsynaptic plasticity and memory. Here, we investigated whether CPEB2 regulates axonal translation to support presynaptic plasticity.
    METHODS: Behavioral and electrophysiological assessments were conducted in mice with pan neuron/glia- or glutamatergic neuron-specific knockout of CPEB2. Hippocampal Schaffer collateral (SC)-CA1 and temporoammonic (TA)-CA1 pathways were electro-recorded to monitor synaptic transmission and LTP evoked by 4 trains of high-frequency stimulation. RNA immunoprecipitation, coupled with bioinformatics analysis, were used to unveil CPEB2-binding axonal RNA candidates associated with learning, which were further validated by Western blotting and luciferase reporter assays. Adeno-associated viruses expressing Cre recombinase were stereotaxically delivered to the pre- or post-synaptic region of the TA circuit to ablate Cpeb2 for further electrophysiological investigation. Biochemically isolated synaptosomes and axotomized neurons cultured on a microfluidic platform were applied to measure axonal protein synthesis and FM4-64FX-loaded synaptic vesicles.
    RESULTS: Electrophysiological analysis of hippocampal CA1 neurons detected abnormal excitability and vesicle release probability in CPEB2-depleted SC and TA afferents, so we cross-compared the CPEB2-immunoprecipitated transcriptome with a learning-induced axonal translatome in the adult cortex to identify axonal targets possibly regulated by CPEB2. We validated that Slc17a6, encoding vesicular glutamate transporter 2 (VGLUT2), is translationally upregulated by CPEB2. Conditional knockout of CPEB2 in VGLUT2-expressing glutamatergic neurons impaired consolidation of hippocampus-dependent memory in mice. Presynaptic-specific ablation of Cpeb2 in VGLUT2-dominated TA afferents was sufficient to attenuate protein synthesis-dependent LTP. Moreover, blocking activity-induced axonal Slc17a6 translation by CPEB2 deficiency or cycloheximide diminished the releasable pool of VGLUT2-containing synaptic vesicles.
    CONCLUSIONS: We identified 272 CPEB2-binding transcripts with altered axonal translation post-learning and established a causal link between CPEB2-driven axonal synthesis of VGLUT2 and presynaptic translation-dependent LTP. These findings extend our understanding of memory-related translational control mechanisms in the presynaptic compartment.
    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:滋养细胞功能紊乱是导致子痫前期(PE)的重要因素之一。已发现细胞质聚腺苷酸化元件结合2(CPEB2)在PE患者中差异表达,但它是否通过调节滋养细胞功能介导PE过程尚不清楚。
    方法:采用实时定量PCR检测CPEB2和生长抑素受体3(SSTR3)的表达,Westernblot(WB)和免疫荧光染色。通过CCK-8测定分析细胞功能,EdU分析,流式细胞术和transwell分析。通过WB检测上皮-间质转化(EMT)相关蛋白水平。CPEB2和SSTR3的相互作用通过RIP实验证实,双荧光素酶报告基因测定和PCRpoly(A)尾测定。进行动物实验以探索CPEB2对体内PE进展的影响。大鼠胎盘组织进行H&E染色,免疫组化染色和TUNEL染色。
    结果:CPEB2在PE患者中低表达。CPEB2上调加速滋养细胞增殖,迁移,入侵和EMT,而它的击倒却产生了相反的效果。CPEB2与SSTR3mRNA的3'-UTR中的CPE位点结合,通过减少poly(A)尾巴来抑制SSTR3的翻译。此外,SSTR3过表达抑制滋养层细胞增殖,迁移,入侵和EMT,而它的沉默加速了滋养层细胞的功能。然而,这些影响可以通过CPEB2的上调和敲低来逆转,分别。体内实验,CPEB2过表达减轻组织病理学改变,抑制细胞凋亡,通过降低SSTR3的表达促进PE大鼠胎盘的增殖并增强EMT。
    结论:CPEB2抑制PE进展,通过多腺苷酸化抑制SSTR3翻译促进滋养层细胞功能。
    Trophoblast cell dysfunction is one of the important factors leading to preeclampsia (PE). Cytoplasmic polyadenylation element-binding 2 (CPEB2) has been found to be differentially expressed in PE patients, but whether it mediates PE process by regulating trophoblast cell function is unclear.
    The expression of CPEB2 and somatostatin receptor 3 (SSTR3) was detected by quantitative real-time PCR, Western blot (WB) and immunofluorescence staining. Cell functions were analyzed by CCK-8 assay, EdU assay, flow cytometry and transwell assay. Epithelial-mesenchymal transition (EMT)-related protein levels were detected by WB. The interaction of CPEB2 and SSTR3 was confirmed by RIP assay, dual-luciferase reporter assay and PCR poly(A) tail assay. Animal experiments were performed to explore the effect of CPEB2 on PE progression in vivo, and the placental tissues of rat were used for H&E staining, immunohistochemical staining and TUNEL staining.
    CPEB2 was lowly expressed in PE patients. CPEB2 upregulation accelerated trophoblast cell proliferation, migration, invasion and EMT, while its knockdown had an opposite effect. CPEB2 bound to the CPE site in the 3\'-UTR of SSTR3 mRNA to suppress SSTR3 translation through reducing poly(A) tails. Besides, SSTR3 overexpression suppressed trophoblast cell proliferation, migration, invasion and EMT, while its silencing accelerated trophoblast cell functions. However, these effects could be reversed by CPEB2 upregulation and knockdown, respectively. In vivo experiments, CPEB2 overexpression relieved histopathologic changes, inhibited apoptosis, promoted proliferation and enhanced EMT in the placenta of PE rat by decreasing SSTR3 expression.
    CPEB2 inhibited PE progression, which promoted trophoblast cell functions by inhibiting SSTR3 translation through polyadenylation.
    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:多发性骨髓瘤(MM)的过程是多个基因联合作用的结果。本研究旨在探讨细胞质聚腺苷酸化元件结合蛋白2(CPEB2)在MM进展中的作用及机制。
    方法:通过实时定量PCR和蛋白质印迹分析评估CPEB2和肌动蛋白相关蛋白2/3复合物亚基5(ARPC5)的mRNA和蛋白表达水平。细胞功能通过细胞计数试剂盒8测定,软琼脂集落形成试验,流式细胞术和试管形成测定。采用荧光原位杂交技术分析CPEB2和ARPC5在MM细胞中的共定位。进行放线菌素D处理和环己酰亚胺追踪测定以评估ARPC5的稳定性。通过RNA免疫沉淀实验证实了CPEB2和ARPC5之间的相互作用。
    结果:在MM患者和细胞的CD138+浆细胞中,CPEB2和ARPC5mRNA和蛋白表达水平上调。CPEB2下调降低MM细胞增殖,血管生成,和增加细胞凋亡,而其过度表达则有相反的作用。CPEB2和ARPC5共定位在细胞质中,可以通过介导其mRNA稳定性来正向调节ARPC5的表达。ARPC5过表达逆转了CPEB2敲低对MM进展的抑制作用,敲除也废除了CPEB2促进的MM进展。此外,CPEB2沉默还通过降低ARPC5表达来降低MM肿瘤生长。
    结论:我们的结果表明,CPEB2通过促进ARPC5的mRNA稳定性增加其表达,从而加速MM的恶性进程。
    BACKGROUND: The process of multiple myeloma (MM) is the result of the combined action of multiple genes. This study aims to explore the role and mechanism of cytoplasmic polyadenylation element binding protein2 (CPEB2) in MM progression.
    METHODS: The mRNA and protein expression levels of CPEB2 and actin-related protein 2/3 complex subunit 5 (ARPC5) were assessed by quantitative real-time PCR and western blot analysis. Cell function was determined by cell counting kit 8 assay, soft-agar colony formation assay, flow cytometry and tube formation assay. Fluorescent in situ hybridization assay was used to analyze the co-localization of CPEB2 and ARPC5 in MM cells. Actinomycin D treatment and cycloheximide chase assay were performed to assess the stability of ARPC5. The interaction between CPEB2 and ARPC5 was confirmed by RNA immunoprecipitation assay.
    RESULTS: CPEB2 and ARPC5 mRNA and protein expression levels were upregulated in CD138+ plasma cells from MM patients and cells. CPEB2 downregulation reduced MM cell proliferation, angiogenesis, and increased apoptosis, while its overexpression had an opposite effect. CPEB2 and ARPC5 were co-localized at cell cytoplasm and could positively regulate ARPC5 expression by mediating its mRNA stability. ARPC5 overexpression reversed the suppressive effect of CPEB2 knockdown on MM progression, and it knockdown also abolished CPEB2-promoted MM progression. Besides, CPEB2 silencing also reduced MM tumor growth by decreasing ARPC5 expression.
    CONCLUSIONS: Our results indicated that CPEB2 increased ARPC5 expression through promoting its mRNA stability, thereby accelerating MM malignant process.
    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
    p53在肿瘤抑制中起重要作用。新出现的证据表明,长链非编码RNA(lncRNA)是控制p53信号传导的一类重要调控分子。这里,我们报告说,致癌lncRNAE2F1信使RNA(mRNA)稳定因子(EMS)和p53相互抑制彼此的表达。EMS受p53负调控。作为p53的直接转录抑制靶标,EMS令人惊讶地显示抑制p53表达。EMS与胞质聚腺苷酸化元件结合蛋白2(CPEB2)相关,破坏CPEB2-p53mRNA的相互作用。这种解离减弱CPEB2介导的p53mRNA聚腺苷酸化并抑制p53翻译。功能上,EMS能够发挥其致癌活性,至少部分地,通过CPEB2-p53轴。一起,这些发现揭示了p53和EMS之间的双负反馈回路,通过它可以精确地控制p53。我们的研究还证明了EMS在通过p53的负调控促进肿瘤发生中的关键作用。
    p53 plays a central role in tumor suppression. Emerging evidence suggests long noncoding RNA (lncRNA) as an important class of regulatory molecules that control the p53 signaling. Here, we report that the oncogenic lncRNA E2F1 messenger RNA (mRNA) stabilizing factor (EMS) and p53 mutually repress each other\'s expression. EMS is negatively regulated by p53. As a direct transcriptional repression target of p53, EMS is surprisingly shown to inhibit p53 expression. EMS associates with cytoplasmic polyadenylation element-binding protein 2 (CPEB2) and thus, disrupts the CPEB2-p53 mRNA interaction. This disassociation attenuates CPEB2-mediated p53 mRNA polyadenylation and suppresses p53 translation. Functionally, EMS is able to exert its oncogenic activities, at least partially, via the CPEB2-p53 axis. Together, these findings reveal a double-negative feedback loop between p53 and EMS, through which p53 is finely controlled. Our study also demonstrates a critical role for EMS in promoting tumorigenesis via the negative regulation of p53.
    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
    放射治疗在乳腺癌(BC)的治疗中起着重要的作用,从乳腺中的恶性细胞发展而来。长非编码RNA(lncRNAs)已被报道与人类癌症的放射抗性或放射敏感性有关。其中包括乳腺癌。然而,在BC中尚未研究长的基因间非蛋白质编码RNA0504(LINC00504)。在我们的研究中,从RT-qPCR分析,发现LINC00504在BC细胞中上调。通过进行体外试验,证实LINC00504敲低可增强BC细胞的放射敏感性。LINC00504在BC中的调控机制也通过染色质免疫沉淀(ChIP)得到证实,RNA免疫沉淀(RIP)和荧光素酶报告基因测定。从实验结果来看,我们知道LINC00504的上调是由信号转导和转录激活因子1(STAT1)介导的。此外,LINC00504通过与TATA盒结合蛋白相关因子15(TAF15)结合,稳定了细胞质聚腺苷酸化元件结合蛋白2(CPEB2)的表达。此外,救援试验验证了LINC00504通过上调CPEB2参与调节BC细胞的放射敏感性.总之,我们的研究表明,STAT1可以介导LINC00504,并通过与TAF15结合并稳定CPEB2表达来削弱BC细胞的放射敏感性。
    Radiotherapy plays important roles in the treatment of breast cancer (BC), which develops from malignant cells in the breast. Long non-coding RNAs (lncRNAs) have been reported to be implicated in radio-resistance or radio-sensitivity of human cancer, which includes breast cancer. Nevertheless, long intergenic non-protein coding RNA 0504 (LINC00504) has not been investigated in BC. In our study, from RT-qPCR analysis, LINC00504 was found to be up-regulated in BC cells. By conducting in vitro assays, it was confirmed that the knockdown of LINC00504 could enhance the radio-sensitivity of BC cells. The regulatory mechanism of LINC00504 in BC was also verified by chromatin immunoprecipitation (ChIP), RNA immunoprecipitation (RIP) and luciferase reporter assays. From the experimental results, we knew that the up-regulation of LINC00504 was mediated by signal transducer and activator of transcription 1 (STAT1). Moreover, LINC00504 stabilized the expression of cytoplasmic polyadenylation element-binding protein 2 (CPEB2) via binding to TATA-box binding protein associated factor 15 (TAF15). Furthermore, rescue assays validated that LINC00504 participated in regulating the radio-sensitivity of BC cells via up-regulating CPEB2. In summary, our study disclosed that STAT1 could mediate LINC00504 and weaken the radio-sensitivity of BC cells via binding to TAF15 and stabilizing CPEB2 expression.
    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
    肿瘤抑制因子p53反式激活多个基因的表达以发挥其多方面的功能并最终维持基因组稳定性。因此,癌细胞发展各种机制来减少p53表达并绕过细胞周期检查点。在这项研究中,我们将编码RNA结合蛋白胞质聚腺苷酸化元件结合蛋白2(CPEB2)的基因鉴定为p53靶标.反过来,CPEB2降低p53信使RNA稳定性和翻译以微调p53水平。具体来说,我们显示CPEB2结合p533'-非翻译区的细胞质聚腺苷酸化元件,这种结合需要CPEB2的RNA识别基序和锌指(ZF)结构域。此外,我们发现CPEB2在肾癌组织中上调并促进肾癌细胞增殖和迁移。CPEB2的致癌作用部分依赖于p53的负反馈调节。总的来说,我们确定了p53和CPEB2之间的一个新的调节反馈回路,并证明CPEB2通过失活p53促进肿瘤进展,这表明CPEB2是人类肾癌的一个潜在治疗靶点.
    The tumor suppressor p53 transactivates the expression of multiple genes to exert its multifaceted functions and ultimately maintains genome stability. Thus, cancer cells develop various mechanisms to diminish p53 expression and bypass the cell cycle checkpoint. In this study, we identified the gene encoding RNA-binding protein cytoplasmic polyadenylation element-binding protein 2 (CPEB2) as a p53 target. In turn, CPEB2 decreases p53 messenger RNA stability and translation to fine-tune p53 level. Specifically, we showed that CPEB2 binds the cytoplasmic polyadenylation elements in the p53 3\'-untranslated region, and the RNA recognition motif and zinc finger (ZF) domains of CPEB2 are required for this binding. Furthermore, we found that CPEB2 was upregulated in renal cancer tissues and promotes the renal cancer cell proliferation and migration. The oncogenic effect of CPEB2 is partially dependent on negative feedback regulation of p53. Overall, we identify a novel regulatory feedback loop between p53 and CPEB2 and demonstrate that CPEB2 promotes tumor progression by inactivating p53, suggesting that CPEB2 is a potential therapeutic target in human renal cancer.
    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
    BACKGROUND: Alveologenesis is the final stage of lung development to form air-exchanging units between alveoli and blood vessels. Genetic susceptibility or hyperoxic stress to perturb this complicated process can cause abnormal enlargement of alveoli and lead to bronchopulmonary dysplasia (BPD)-associated emphysema. Platelet-derived growth factor receptor α (PDGFRα) signaling is crucial for alveolar myofibroblast (MYF) proliferation and its deficiency is associated with risk of BPD, but posttranscriptional mechanisms regulating PDGFRα synthesis during lung development remain largely unexplored. Cytoplasmic polyadenylation element-binding protein 2 (CPEB2) is a sequence-specific RNA-binding protein and translational regulator. Because CPEB2-knockout (KO) mice showed emphysematous phenotypes, we investigated how CPEB2-controlled translation affects pulmonary development and function.
    METHODS: Respiratory and pulmonary functions were measured by whole-body and invasive plethysmography. Histological staining and immunohistochemistry were used to analyze morphology, proliferation, apoptosis and cell densities from postnatal to adult lungs. Western blotting, RNA-immunoprecipitation, reporter assay, primary MYF culture and ectopic expression rescue were performed to demonstrate the role of CPEB2 in PDGFRα mRNA translation and MYF proliferation.
    RESULTS: Adult CPEB2-KO mice showed emphysema-like dysfunction. The alveolar structure in CPEB2-deficient lungs appeared normal at birth but became simplified through the alveolar stage of lung development. In CPEB2-null mice, we found reduced proliferation of MYF progenitors during alveolarization, abnormal deposition of elastin and failure of alveolar septum formation, thereby leading to enlarged pulmonary alveoli. We identified that CPEB2 promoted PDGFRα mRNA translation in MYF progenitors and this positive regulation could be disrupted by H2O2, a hyperoxia-mimetic treatment. Moreover, decreased proliferating ability in KO MYFs due to insufficient PDGFRα expression was rescued by ectopic expression of CPEB2, suggesting an important role of CPEB2 in upregulating PDGFRα signaling for pulmonary alveologenesis.
    CONCLUSIONS: CPEB2-controlled translation, in part through promoting PDGFRα expression, is indispensable for lung development and function. Since defective pulmonary PDGFR signaling is a key feature of human BPD, CPEB2 may be a risk factor for BPD.
    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
    妊娠并发症先兆子痫与缺氧应激和滋养细胞分化不足直接相关。缺氧诱导的微小RNA(miRNA),miR-210已被鉴定为先兆子痫胎盘中显著上调的miRNA,和其他细胞类型的证据表明,在缺氧条件下,miR-210和缺氧诱导因子-1α(HIF-1α)之间存在反馈调节。目前尚不清楚miR-210和HIF-1α之间的反馈回路是否以及如何导致先兆子痫的滋养细胞功能障碍。这里,我们证明细胞质多聚腺苷酸化元件结合2(CPEB2)是miR-210在人滋养细胞中的直接靶标.CPEB2可以通过直接结合HIF-1αmRNA3'-非翻译区(UTR)的细胞质多聚腺苷酸化元件(CPE)位点来抑制缺氧诱导的HIF-1α的翻译。缺氧处理后HIF-1α水平的增加可被miR-210抑制剂有效逆转。此外,CPEB2主要在绒毛合胞体滋养层中表达,miR-210对滋养细胞合胞体的抑制可被CPEB2过表达显著挽救。先兆子痫胎盘,CPEB2的表达明显低于正常妊娠对照,miR-210水平异常升高,滋养细胞合胞体有限.研究结果揭示了miR-210和HIF-1α之间的正反馈环,这是由CPEB2介导的人滋养细胞,并证明了在缺氧应激下先兆子痫中滋养细胞融合不足的潜在机制。
    The pregnancy complication preeclampsia is directly associated with hypoxic stress and insufficient trophoblast cell differentiation. The hypoxia-inducible microRNA (miRNA), miR-210, has been identified as a significantly up-regulated miRNA in preeclamptic placenta, and evidence in other cell types has indicated a feedback regulation between miR-210 and hypoxia-inducible factor-1α (HIF-1α) under hypoxic condition. It remains unclear whether and how the feedback loop between miR-210 and HIF-1α may contribute to trophoblast dysfunction in preeclampsia. Here, we proved that cytoplasmic polyadenylation element-binding 2 (CPEB2) was a direct target of miR-210 in human trophoblast. CPEB2 could inhibit the translation of hypoxia-induced HIF-1α via directly binding the cytoplasmic polyadenylation element (CPE) site in the 3\'-untranslated region (UTR) of HIF-1α mRNA. The increase in the HIF-1α level upon hypoxia treatment could be efficiently reversed by miR-210 inhibitor. In addition, CPEB2 was primarily expressed in villous syncytiotrophoblasts, and the suppression of trophoblast cell syncytialization by miR-210 could be significantly rescued by CPEB2 overexpression. In preeclamptic placenta, the expression of CPEB2 was evidently lower than normal pregnant control, and the miR-210 level was aberrantly higher and trophoblast syncytialization was limited. The findings revealed a positive feedback loop between miR-210 and HIF-1α that is mediated by CPEB2 in human trophoblasts, and demonstrated a mechanism underlying the insufficient trophoblast syncytialization in preeclampsia under hypoxic stress.
    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
    BACKGROUND: Over-expression of cyclooxygenase (COX)-2 promotes breast cancer progression by multiple mechanisms, including induction of stem-like cells (SLC). Combined gene expression and microRNA microarray analyses of empty vector vs COX-2- transfected COX-2 low MCF7 breast cancer cell line identified two COX-2-upregulated microRNAs, miR-526b and miR-655, both found to be oncogenic and SLC-promoting. Cytoplasmic Polyadenylation Element-Binding Protein 2 (CPEB2) was the single common target of both microRNAs, the functions of which remain controversial. CPEB2 has multiple isoforms (A-F), and paradoxically, a high B/A ratio was reported to impart anoikis-resistance and metastatic phenotype in triple- negative breast cancer cells. We tested whether CPEB2 is a tumor suppressor in mammary epithelial cells.
    METHODS: We knocked-out CPEB2 in the non-tumorigenic mammary epithelial cell line MCF10A by CRISPR/Cas9-double nickase approach, and knocked-down CPEB2 with siRNAs in the poorly malignant MCF7 cell line, both lines being high CPEB2-expressing. The resultant phenotypes for oncogenity were tested in vitro for both lines and in vivo for CPEB2KO cells. Finally, CPEB2 expression was compared between human breast cancer and non-tumor breast tissues.
    RESULTS: CPEB2 (isoform A) expression was inversely correlated with COX-2 or the above microRNAs in COX-2-divergent breast cancer cell lines. CPEB2KO MCF10A cells exhibited oncogenic properties including increased proliferation, migration, invasion, EMT (decreased E-Cadherin, increased Vimentin, N-Cadherin, SNAI1, and ZEB1) and SLC phenotype (increased tumorsphere formation and SLC marker-expression). Tumor-suppressor p53 protein was shown to be a novel translationally-regulated target of CPEB2, validated with polysome profiling. CPEB2KO, but not wild-type cells produced lung colonies upon intravenous injection and subcutaneous tumors and spontaneous lung metastases upon implantation at mammary sites in NOD/SCID/IL2Rϒ-null mice, identified with HLA immunostaining. Similarly, siRNA-mediated CPEB2 knockdown in MCF7 cells promoted oncogenic properties in vitro. Human breast cancer tissues (n = 105) revealed a lower mRNA expression for CPEB2 isoform A and also a lower A/B isoform ratio than in non-tumour breast tissues (n = 20), suggesting that CPEB2A accounts for the tumor-suppressor functions of CPEB2.
    CONCLUSIONS: CPEB2, presumably the isoform A, plays a key role in suppressing tumorigenesis in mammary epithelial cells by repressing EMT, migration, invasion, proliferation and SLC phenotype, via multiple targets, including a newly-identified translational target p53.
    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
    Oocyte meiotic maturation and embryogenesis are some of the most important physiological processes that occur in organisms, playing crucial roles in the preservation of life in all species. The post-transcriptional regulation of maternal messenger ribonucleic acids (mRNAs) and the post-translational regulation of proteins are critical in the control of oocyte maturation and early embryogenesis. Translational control affects the basic mechanism of protein synthesis, thus, knowledge of the key components included in this machinery is required in order to understand its regulation. Cytoplasmic polyadenylation element binding proteins (CPEBs) bind to the 3\'-end of mRNAs to regulate their localization and translation and are necessary for proper development. In this study we examined the expression pattern of cytoplasmic polyadenylation element binding protein 2 (CPEB2) both on the mRNA (by real-time quantitative reverse transcription polymerase chain reaction, qRT-PCR) and protein (by Western blotting, WB) level, as well as its localization during the meiotic maturation of porcine oocytes and early embryonic development by immunocytochemistry (ICC). For the elucidation of its functions, CPEB2 knockdown by double-strand RNA (dsRNA) was used. We discovered that CPEB2 is expressed during all stages of porcine meiotic maturation and embryonic development. Moreover, we found that it is necessary to enable a high percentage of oocytes to reach the metaphase II (MII) stage, as well as for the production of good-quality parthenogenetic blastocysts.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号