alpha Karyopherins

α 核动力素
  • 文章类型: Journal Article
    背景:肝细胞癌(HCC)是一种以缺氧环境而闻名的高度恶性肿瘤,这有助于对抗癌药物索拉非尼(SF)的抗性。解决HCC中的SF耐药性需要创新的策略来改善肿瘤氧合并有效地提供治疗。
    结果:在我们的研究中,我们探讨了KPNA4在介导缺氧诱导的肝癌SF抵抗中的作用。我们开发了能够携带氧气的血红蛋白纳米簇(Hb-NC),负载吲哚菁绿(ICG)和SF,名为HPRG@SF。体外,HPRG@SF靶向肝癌细胞,缓解缺氧,抑制KPNA4表达,并增强了PDT对缺氧的细胞毒性,SF抗性HCC细胞。体内实验支持这些发现,表明HPRG@SF有效改善了肿瘤微环境内的氧合,并通过联合光动力疗法(PDT)抵抗了SF抵抗。
    结论:Hb-NC与ICG和SF的组合,形成HPRG@SF,提出了通过改善缺氧和使用PDT来克服肝细胞癌耐药性的有效策略。这种方法不仅针对耐药性背后的低氧条件,而且还提供了协同抗癌作用。强调其临床应用于治疗耐药肝癌的潜力。
    BACKGROUND: Hepatocellular carcinoma (HCC) is a highly malignant tumor known for its hypoxic environment, which contributes to resistance against the anticancer drug Sorafenib (SF). Addressing SF resistance in HCC requires innovative strategies to improve tumor oxygenation and effectively deliver therapeutics.
    RESULTS: In our study, we explored the role of KPNA4 in mediating hypoxia-induced SF resistance in HCC. We developed hemoglobin nanoclusters (Hb-NCs) capable of carrying oxygen, loaded with indocyanine green (ICG) and SF, named HPRG@SF. In vitro, HPRG@SF targeted HCC cells, alleviated hypoxia, suppressed KPNA4 expression, and enhanced the cytotoxicity of PDT against hypoxic, SF-resistant HCC cells. In vivo experiments supported these findings, showing that HPRG@SF effectively improved the oxygenation within the tumor microenvironment and countered SF resistance through combined photodynamic therapy (PDT).
    CONCLUSIONS: The combination of Hb-NCs with ICG and SF, forming HPRG@SF, presents a potent strategy to overcome drug resistance in hepatocellular carcinoma by improving hypoxia and employing PDT. This approach not only targets the hypoxic conditions that underlie resistance but also provides a synergistic anticancer effect, highlighting its potential for clinical applications in treating resistant HCC.
    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
    RNA结合蛋白(RBP)在基因组调控中起关键作用。在这项研究中,我们探索了KPNA2的潜在功能,KPNA2是RBP家族的重要成员,在胃癌(GC)中选择性剪接(AS)的调控。我们通过RNA测序(RNA-seq)和改进的RNA免疫沉淀测序(iRIP-seq)分析了KPNA2在调节差异表达和AS中的作用。临床标本用于分析KPNA2表达与临床病理特征之间的关联。CCK8测定,进行了transwell测定和伤口愈合测定,以探索KPNA2/WDR62对GC细胞进展的影响。KPNA2在GC细胞和组织中高表达,并与淋巴结转移有关。KPNA2促进了扩散,GC细胞的迁移和侵袭,主要调节外显子跳跃,替代3的剪接位点(A3SS),替代5个剪接位点(A5SS),和盒式磁带外显子。我们进一步揭示了KPNA2参与了与细胞增殖相关的生物学过程。而GC中的免疫应答则经由过程转录调控。此外,KPNA2优先结合内含子区域。值得注意的是,KPNA2调节WDR62的A3SSAS模式,WDR62的上调逆转了KPNA2下调诱导的GC细胞增殖抑制,移民和入侵。最后,我们发现免疫相关分子的AS可以被KPNA2调节。总的来说,我们的研究结果首次证明,KPNA2在GC中作为致癌剪接因子起作用,调节AS和GC相关基因的差异表达,KPNA2可能是GC治疗的潜在靶标。
    RNA-binding proteins (RBPs) play critical roles in genome regulation. In this study, we explored the latent function of KPNA2, which is an essential member of the RBP family, in the regulation of alternative splicing (AS) in gastric cancer (GC). We analyzed the role of KPNA2 in regulating differential expression and AS via RNA sequencing (RNA-seq) and improved RNA immunoprecipitation sequencing (iRIP-seq). Clinical specimens were used to analyze the associations between KPNA2 expression and clinicopathological characteristics. CCK8 assays, transwell assays and wound healing assays were performed to explore the effect of KPNA2/WDR62 on GC cell progression. KPNA2 was shown to be highly expressed in GC cells and tissues and associated with lymph node metastases. KPNA2 promoted the proliferation, migration and invasion of GC cells and primarily regulated exon skipping, alternative 3\'s splice sites (A3SSs), alternative 5\' splice sites (A5SSs), and cassette exons. We further revealed that KPNA2 participated in biological processes related to cell proliferation, and the immune response in GC via the regulation of transcription. In addition, KPNA2 preferentially bound to intron regions. Notably, KPNA2 regulated the A3SS AS mode of WDR62, and upregulation of WDR62 reversed the KPNA2 downregulation-induced inhibition of GC cell proliferation, migration and invasion. Finally, we discovered that the AS of immune-related molecules could be regulated by KPNA2. Overall, our results demonstrated for the first time that KPNA2 functions as an oncogenic splicing factor in GC that regulated the AS and differential expression of GC-related genes, and KPNA2 may be a potential target for GC treatment.
    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:胃复春(WFC)是一种常用于治疗萎缩性胃炎和肠上皮化生的中药。直到日期,其对胃癌(GC)的抗肿瘤作用及其潜在机制仍未阐明。
    目的:我们的目的是研究WFC是否可以抑制胃癌细胞的恶性,并剖析其分子基础和相关的分子和细胞特征。
    方法:用WFC处理胃癌细胞株和正常胃上皮细胞。CCK8测定,caspase-3活性测定,粘附试验,microRNA数据库分析,转染,RT-PCR,西方印迹,信号通路分析,和体内GC模型用于检查胃癌细胞特征的变化以及WFC作用的分子机制。
    结果:在这里,我们提供的数据表明WFC抑制了GC的恶性细胞表型,并且这种抑制作用是通过下调致癌KPNA2的表达而介导的。此外,WFC通过miR-26a-5p介导的基因沉默和丝裂原活化蛋白激酶(MAPK)的失活磷酸化动力学下调KPNA2。在动物模型中进一步证实了WFC对胃癌细胞行为的抑制作用。
    结论:因此,WFC可通过降低KPNA2水平对GC细胞的恶性程度产生抑制作用。此外,WFC诱导的miR-26a-5p拯救和MAPK通路失活导致KPNA2表达下调。因此,我们的研究结果表明WFC是一种潜在的抗GC治疗方案.
    BACKGROUND: Weifuchun (WFC) is a Traditional Chinese Medicine commonly used for treating atrophic gastritis and intestinal metaplasia. Till date, its antitumor effect on gastric cancer (GC) and the underlying mechanisms of the effect remains unelucidated.
    OBJECTIVE: We aim to investigate if WFC can suppress the malignancy of stomach cancer cells and dissect the molecular basis and the associated molecular and cellular features.
    METHODS: Stomach cancer cell lines and normal gastric epithelial cells were treated with WFC. CCK8 assay, caspase-3 activity assay, adhesion assay, microRNA database analysis, transfection, RT-PCR, Western Blotting, signaling pathway analysis, and in vivo GC model were employed to examine the changes in the features of the gastric cancer cells and the molecular mechanisms of the effect of WFC.
    RESULTS: Here we present data demonstrating that WFC suppresses the malignant cellular phenotypes of GC and this inhibitory effect is mediated by downregulating the expression of oncogenic KPNA2. Furthermore, WFC downregulates KPNA2 through miR-26a-5p-mediated gene silencing and the deactivated phosphorylation dynamics of mitogen-activated protein kinase (MAPK). The suppressive effect of WFC on stomach cancer cell behavior was further confirmed in animal model.
    CONCLUSIONS: Therefore, WFC can exert inhibitory effect on the malignancy of GC cells by reducing the levels of KPNA2. Moreover, the miR-26a-5p rescue and the deactivation MAPK pathway induced by WFC result in the downregulation of KPNA2 expression. Thus, our findings suggest WFC as a potential treatment option against GC.
    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
    Poxviridae的正痘病毒(OPXV)属包括人类病原体天花病毒(VARV),猴痘病毒(MPXV),痘苗病毒(VACV),和一些人畜共患病毒。VACV的许多Bcl-2样蛋白参与逃避宿主先天免疫。然而,在其他OPXVs中,很少有工作致力于其直系同源物的进化和功能。这里,我们发现由P2L基因编码的MPXV蛋白P2,和来自其他OPXV的P2直系同源物,例如VACV蛋白N2,定位于细胞核并拮抗干扰素(IFN)的产生。例外的是缺少核定位信号(NLS)的骆驼痘病毒(CMLV)和taterapox病毒(TATV)中的截短的P2直向同源物。机械上,MPXVP2的NLS与核蛋白α-2(KPNA2)相互作用以促进P2核易位,并竞争性抑制KPNA2介导的IRF3核易位和下游IFN的产生。在P2或直系同源物中NLS的缺失显着增强IRF3核易位和先天免疫反应,从而减少病毒复制。此外,在VACV中从N2中缺失NLS减弱了小鼠中的病毒复制和毒力。这些数据表明,NLS介导的P2易位对于P2诱导的先天免疫抑制至关重要。我们的发现有助于深入了解OPXVP2直向同源物在先天免疫逃避中的机制。
    The Orthopoxvirus (OPXV) genus of the Poxviridae includes human pathogens variola virus (VARV), monkeypox virus (MPXV), vaccinia virus (VACV), and a number of zoonotic viruses. A number of Bcl-2-like proteins of VACV are involved in escaping the host innate immunity. However, little work has been devoted to the evolution and function of their orthologues in other OPXVs. Here, we found that MPXV protein P2, encoded by the P2L gene, and P2 orthologues from other OPXVs, such as VACV protein N2, localize to the nucleus and antagonize interferon (IFN) production. Exceptions to this were the truncated P2 orthologues in camelpox virus (CMLV) and taterapox virus (TATV) that lacked the nuclear localization signal (NLS). Mechanistically, the NLS of MPXV P2 interacted with karyopherin α-2 (KPNA2) to facilitate P2 nuclear translocation, and competitively inhibited KPNA2-mediated IRF3 nuclear translocation and downstream IFN production. Deletion of the NLS in P2 or orthologues significantly enhanced IRF3 nuclear translocation and innate immune responses, thereby reducing viral replication. Moreover, deletion of NLS from N2 in VACV attenuated viral replication and virulence in mice. These data demonstrate that the NLS-mediated translocation of P2 is critical for P2-induced inhibition of innate immunity. Our findings contribute to an in-depth understanding of the mechanisms of OPXV P2 orthologue in innate immune evasion.
    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
    委内瑞拉马脑炎病毒(VEEV)是一种高毒力病原体,其衣壳蛋白的核定位信号(NLS)序列与宿主importin-α转运蛋白结合并阻断核输入。我们研究了两个小配体的分子机制,称为I1和I2,干扰VEEV的NLS肽与importin-α蛋白的结合。为此,我们进行了全原子复制交换分子动力学模拟,探索了VEEV核心NLS肽和I1或I2配体与importin-α主要NLS结合位点的竞争性结合。作为参考,我们用之前的模拟,检查了coreNLS肽或抑制剂与importin-α的非竞争性结合。我们发现这两种抑制剂都完全消除了核心NLS肽的天然结合,迫使其在importin-α主要NLS结合位点内采用多种非天然松散结合的姿势。两种抑制剂主要通过掩蔽其氨基酸而不是与其竞争结合importin-α来使天然coreNLS结合不稳定。因为I2与I1相反,结合位点外定位在主要NLS结合位点的边缘,与I1相比,它抑制较少的coreNLS天然结合相互作用。结构分析得到了在有或没有来自抑制剂的竞争的情况下与importin-α结合的核NLS肽的自由能的计算的支持。具体来说,这两种抑制剂都降低了核心NLS结合的自由能增益,I1造成的损失明显大于I2。为了测试我们的模拟,我们进行了AlphaScreen实验,测量两种抑制剂的IC50值.与计算机模拟结果一致,发现I1的IC50值低于I2的IC50值。我们假设I1和I2配体的抑制作用可能对VEEV衣壳蛋白的NLS具有特异性。
    Venezuelan equine encephalitis virus (VEEV) is a highly virulent pathogen whose nuclear localization signal (NLS) sequence from capsid protein binds to the host importin-α transport protein and blocks nuclear import. We studied the molecular mechanisms by which two small ligands, termed I1 and I2, interfere with the binding of VEEV\'s NLS peptide to importin-α protein. To this end, we performed all-atom replica exchange molecular dynamics simulations probing the competitive binding of the VEEV coreNLS peptide and I1 or I2 ligand to the importin-α major NLS binding site. As a reference, we used our previous simulations, which examined noncompetitive binding of the coreNLS peptide or the inhibitors to importin-α. We found that both inhibitors completely abrogate the native binding of the coreNLS peptide, forcing it to adopt a manifold of nonnative loosely bound poses within the importin-α major NLS binding site. Both inhibitors primarily destabilize the native coreNLS binding by masking its amino acids rather than competing with it for binding to importin-α. Because I2, in contrast to I1, binds off-site localizing on the edge of the major NLS binding site, it inhibits fewer coreNLS native binding interactions than I1. Structural analysis is supported by computations of the free energies of the coreNLS peptide binding to importin-α with or without competition from the inhibitors. Specifically, both inhibitors reduce the free energy gain from coreNLS binding, with I1 causing significantly larger loss than I2. To test our simulations, we performed AlphaScreen experiments measuring IC50 values for both inhibitors. Consistent with in silico results, the IC50 value for I1 was found to be lower than that for I2. We hypothesize that the inhibitory action of I1 and I2 ligands might be specific to the NLS from VEEV\'s capsid protein.
    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
    猪deltacoronavirus(PDCoV)是一种肠道致病性冠状病毒,可引起仔猪急性和严重的水样腹泻,并具有跨物种传播的能力,对养猪生产和公众健康造成极大威胁。干扰素(IFN)介导的信号转导是病毒与宿主相互作用的重要组成部分,在调节病毒感染中起着至关重要的作用。以前的研究表明,冠状病毒编码的多功能病毒蛋白通过各种方式拮抗IFN的产生。然而,这些病毒蛋白在调节IFN介导的信号通路中的功能在很大程度上是未知的。在这项研究中,我们证明PDCoV及其编码的核衣壳(N)蛋白拮抗I型IFN介导的JAK-STAT信号通路。我们确定PDCoV感染刺激但延迟了IFN刺激基因(ISG)的产生。此外,PDCoV通过靶向STAT1和ISGF3形成的核易位抑制JAK-STAT信号转导。进一步的证据表明,PDCoVN是通过经由其C末端结构域靶向STAT1核易位而参与抑制I型IFN信号传导的必需蛋白。机械上,PDCoVN通过与STAT1相互作用并随后抑制其核易位来靶向STAT1。此外,PDCoVN通过溶酶体途径特异性靶向KPNA2降解抑制STAT1核易位,从而抑制JAK-STAT信号通路下游传感器的激活。一起来看,我们的结果揭示了PDCoVN干扰宿主抗病毒反应的新机制。IMPORTANCEP猪deltacronovirus(PDCoV)是一种新型肠致病性冠状病毒,日益受到关注,严重威胁养猪业和公众健康。了解PDCoV在感染过程中逃避宿主防御的潜在机制对于开发针对PDCoV的靶向药物和有效疫苗至关重要。这项研究表明,PDCoV及其编码的核衣壳(N)蛋白通过靶向STAT1拮抗I型干扰素信号,STAT1是JAK-STAT信号通路中的关键信号传感器。进一步的实验表明,PDCoVN介导的STAT1核易位抑制涉及KPNA2的降解,溶酶体在KPNA2降解中起作用。这项研究为PDCoVN在JAK-STAT信号通路中的调控提供了新的见解,并揭示了PDCoV逃避宿主抗病毒反应的新机制。新发现可能指导我们发现新的治疗靶点并开发用于PDCoV感染的减毒活疫苗。
    Porcine deltacoronavirus (PDCoV) is an enteric pathogenic coronavirus that causes acute and severe watery diarrhea in piglets and has the ability of cross-species transmission, posing a great threat to swine production and public health. The interferon (IFN)-mediated signal transduction represents an important component of virus-host interactions and plays an essential role in regulating viral infection. Previous studies have suggested that multifunctional viral proteins encoded by coronaviruses antagonize the production of IFN via various means. However, the function of these viral proteins in regulating IFN-mediated signaling pathways is largely unknown. In this study, we demonstrated that PDCoV and its encoded nucleocapsid (N) protein antagonize type I IFN-mediated JAK-STAT signaling pathway. We identified that PDCoV infection stimulated but delayed the production of IFN-stimulated genes (ISGs). In addition, PDCoV inhibited JAK-STAT signal transduction by targeting the nuclear translocation of STAT1 and ISGF3 formation. Further evidence showed that PDCoV N is the essential protein involved in the inhibition of type I IFN signaling by targeting STAT1 nuclear translocation via its C-terminal domain. Mechanistically, PDCoV N targets STAT1 by interacting with it and subsequently inhibiting its nuclear translocation. Furthermore, PDCoV N inhibits STAT1 nuclear translocation by specifically targeting KPNA2 degradation through the lysosomal pathway, thereby inhibiting the activation of downstream sensors in the JAK-STAT signaling pathway. Taken together, our results reveal a novel mechanism by which PDCoV N interferes with the host antiviral response.IMPORTANCEPorcine deltacoronavirus (PDCoV) is a novel enteropathogenic coronavirus that receives increased attention and seriously threatens the pig industry and public health. Understanding the underlying mechanism of PDCoV evading the host defense during infection is essential for developing targeted drugs and effective vaccines against PDCoV. This study demonstrated that PDCoV and its encoded nucleocapsid (N) protein antagonize type I interferon signaling by targeting STAT1, which is a crucial signal sensor in the JAK-STAT signaling pathway. Further experiments suggested that PDCoV N-mediated inhibition of the STAT1 nuclear translocation involves the degradation of KPNA2, and the lysosome plays a role in KPNA2 degradation. This study provides new insights into the regulation of PDCoV N in the JAK-STAT signaling pathway and reveals a novel mechanism by which PDCoV evades the host antiviral response. The novel findings may guide us to discover new therapeutic targets and develop live attenuated vaccines for PDCoV infection.
    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
    在这个问题上,Yang等人的发现。(https://doi.org/10.1083/jcb.202308013)折叠YAP1的WW域和其他蛋白质结合Impa引入了一类新的球状NLS,与广泛研究的线性NLS基序形成对比。这一发现强调了重要蛋白在识别其货物蛋白方面的多功能性。
    In this issue, the discovery by Yang et al. (https://doi.org/10.1083/jcb.202308013) that folded WW domains of YAP1 and other proteins bind to Impα introduces a new class of globular NLS, contrasting with the extensively studied linear NLS motifs. This finding underscores the versatility of importins in recognizing their cargo proteins.
    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
    蛋白质的核转运在介导从卵到胚胎的过渡中起着重要作用,并且不同的核动力蛋白参与了这一过程。这里,我们研究了KPNA2缺乏对小鼠植入前胚胎发育的影响。KPNA2的丢失导致在2细胞阶段完全停滞,胚胎表现出无法激活其胚胎基因组以及核酶2的严重干扰的核易位。我们的发现将KPNA2定义为新的母体效应基因。
    The nuclear transport of proteins plays an important role in mediating the transition from egg to embryo and distinct karyopherins have been implicated in this process. Here, we studied the impact of KPNA2 deficiency on preimplantation embryo development in mice. Loss of KPNA2 results in complete arrest at the 2cell stage and embryos exhibit the inability to activate their embryonic genome as well as a severely disturbed nuclear translocation of Nucleoplasmin 2. Our findings define KPNA2 as a new maternal effect gene.
    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
    口腔鳞状细胞癌(OSCC)是一种常见的恶性肿瘤。Importin7(IPO7)负责RNA和蛋白质的核质运输,它已被证实与人类癌症的发展有关。本研究旨在探讨IPO7在OSCC中的作用及机制。通过RT-qPCR测定组织和细胞中的IPO7表达。细胞增殖性,迁徙,并通过transwell试验和集落形成试验检测侵袭能力。建立小鼠异种移植模型以评估肿瘤生长。通过蛋白质印迹和免疫荧光(IF)通过细胞中的LC3水平估计自噬。Westernblot用于检测PERK/EIF2AK3/ATF4途径中的关键蛋白,以评估内质网应激(ERS)。通过GST下拉测定和Co-IP测定测试IPO7和同源盒A10(HOXA10)的相互作用。使用ChIP测定和荧光素酶报告基因测定来确定HOXA10和EIF2AK3的组合。我们证明IPO7在OSCC组织和细胞中上调,它的耗竭减少了细胞增殖,迁移,侵袭和肿瘤生长。此外,发现细胞中的LC3表达通过IPO7敲低而降低。IPO7通过激活自噬促进OSCC肿瘤转移。此外,我们发现IPO7可以通过激活PERK/ATF4通路来调节ERS。EIF2AK3上调可促进细胞自噬。此外,IPO7被证明可促进HOXA10在细胞中的核转位。EIF2AK3启动子可以与HOXA10结合。挽救试验证实HOXA10上调可以逆转IPO7沉默对OSCC进展的影响。IPO7可以增强增殖,迁移,入侵,和自噬通过HOXA10的核转位和EIF2AK3/ATF4通路的激活在OSCC中。
    Oral squamous cell carcinoma (OSCC) is a common malignant tumor. Importin7 (IPO7) is responsible for nucleoplasmic transport of RNAs and proteins, and it has been confirmed to be involved in the development of human cancers. This study aimed to explore the function and mechanism of IPO7 in OSCC. IPO7 expression in tissues and cells was determined by RT-qPCR. Cell proliferative, migratory, and invasive capabilities were detected through transwell assay and colony formation assay. Mice xenograft models were established for evaluating tumor growth. Autophagy was estimated by the LC3 levels in cells through western blot and immunofluorescence (IF). Western blot was utilized to detect the key proteins in PERK/EIF2AK3/ATF4 pathway for assessing the endoplasmic reticulum stress (ERS). The interaction of IPO7 and homeobox A10 (HOXA10) was tested by GST pull-down assay and Co-IP assay. ChIP assay and luciferase reporter assay were utilized to determine the combination of HOXA10 and EIF2AK3. We proved that IPO7 was upregulated in OSCC tissues and cells, and its depletion reduced cell proliferation, migration, invasion and tumor growth. Furthermore, LC3 expression in cells was found to be reduced by IPO7 knockdown. IPO7 promoted OSCC tumor metastasis by activating autophagy. Additionally, we discovered that IPO7 could regulate ERS by activating the PERK/ATF4 pathway. EIF2AK3 upregulation can promote cell autophagy. Furthermore, IPO7 was proven to promote nuclear translocation of HOXA10 in cells. EIF2AK3 promoter can bind to HOXA10. Rescue assay confirmed that HOXA10 upregulation can reverse the effect of IPO7 silencing on OSCC progression. IPO7 can enhance proliferation, migration, invasion, and autophagy by nuclear translocation of HOXA10 and the activation of EIF2AK3/ATF4 pathway in OSCC.
    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
    干细胞维持和分化可以通过干细胞及其后代中转录因子的差异活性来调节。为了使这些因素活跃起来,它们需要从细胞质中的合成位点运输到细胞核中。对参与核输入的因子的组织特异性要求是调节干细胞分化的潜在机制。我们已经对果蝇中的雄性不育输入蛋白α1(Dα1)无效等位基因进行了表征,发现Dα1是维持睾丸生态位中的种系干细胞(GSC)所必需的。GSC的丢失可以通过Dα1在种系内的异位表达来挽救,但动物仍然不育,表明Dα1在精子发生中的第二个作用。GSC中Dα1显性阴性转基因的表达证实了在GSC维持中对Dα1的功能要求,但在分化精原细胞中转基因的表达未表现出表明Dα1在GSC中的特定作用的表型。我们的数据表明,Dα1被用作GSC内的调节蛋白,以促进维持干细胞库的蛋白的核输入。
    Stem cell maintenance and differentiation can be regulated via the differential activity of transcription factors within stem cells and their progeny. For these factors to be active, they need to be transported from their site of synthesis in the cytoplasm into the nucleus. A tissue-specific requirement for factors involved in nuclear importation is a potential mechanism to regulate stem cell differentiation. We have undertaken a characterization of male sterile importin alpha 1 (Dα1) null alleles in Drosophila and found that Dα1 is required for maintaining germline stem cells (GSCs) in the testis niche. The loss of GSCs can be rescued by ectopic expression of Dα1 within the germline but the animals are still infertile, indicating a second role for Dα1 in spermatogenesis. Expression of a Dα1 dominant negative transgene in GSCs confirmed a functional requirement for Dα1 in GSC maintenance but expression of the transgene in differentiating spermatogonia did not exhibit a phenotype indicating a specific role for Dα1 within GSCs. Our data indicate that Dα1 is utilized as a regulatory protein within GSCs to facilitate nuclear importation of proteins that maintain the stem cell pool.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号