...
首页> 外文期刊>PLoS Genetics >Spermatogenesis Studies Reveal a Distinct Nonsense-Mediated mRNA Decay (NMD) Mechanism for mRNAs with Long 3′UTRs
【24h】

Spermatogenesis Studies Reveal a Distinct Nonsense-Mediated mRNA Decay (NMD) Mechanism for mRNAs with Long 3′UTRs

机译:精子发生研究揭示了具有长3'UTR的MRNA的不同的废话介导的mRNA腐烂(NMD)机制

获取原文

摘要

Extensive alternative splicing and polyadenylation of pre-mRNAs not only expands the protein coding potential of our genomes but also generates a wealth of mRNA isoforms with different 3′ untranslated regions (UTRs) [ 1 , 2 ]. Since 3′UTRs are major regulators of mRNA stability, localization, and translation, the tissue-specific, developmentally regulated, and stress-induced generation of alternative 3′UTRs greatly contributes to the posttranscriptional regulation of gene expression. It is an intriguing observation that mRNAs with the longest 3′UTRs are predominately present in the brain, whereas the testis is enriched in mRNA isoforms with shorter 3′UTRs [ 3 , 4 ]. In general, mRNAs with shorter 3′UTRs tend to be more stable because (i) they contain fewer binding sites for decay-inducing miRNAs or RNA-binding proteins (RBPs) and (ii) long 3′UTRs can trigger nonsense-mediated mRNA decay (NMD) [ 1 , 5 ]. The term NMD was initially coined to describe the accelerated degradation observed for mRNAs with nonsense mutations that prematurely truncate the open reading frame (ORF) [ 6 ]. NMD serves an important cellular quality control function by reducing the production of potentially deleterious C-terminally truncated proteins. However, genome-wide studies uncovered that beyond degrading aberrant mRNAs harboring premature translation termination codons (PTCs), NMD also targets many “normal” mRNAs encoding apparently full-length functional proteins, suggesting a broader biological function in posttranscriptional gene regulation [ 5 , 7 ]. The three conserved core factors (UPF1, UPF2, and UPF3) and additional metazoan-specific proteins are required for NMD in mammalian cells and, although the exact molecular mechanism of NMD is not known, inefficient or aberrant translation termination seems to be a key trigger for NMD. It has been empirically found that exon–exon junctions located >50 nucleotides downstream of the termination codon often trigger NMD, which is typically the case in aberrant PTC-containing transcripts, and that long 3′UTRs can also elicit NMD, a feature found in many of the PTC-free NMD targets [ 8 – 10 ]. Whether these two groups of NMD substrates are recognized and degraded by a common mechanism or whether they employ mechanistically distinct branches of NMD is a major unresolved question in the field. Two new studies on mouse spermatogenesis [ 11 , 12 ], both published in this issue of PLOS Genetics , now shed some unexpected new light on this question. During spermatogenesis, spermatogonia differentiate into spermatocytes, which undergo meiosis and postmeiotically develop into round and, further, elongated spermatids ( Fig 1 ). 10.1371/journal.pgen.1005979.g001 Fig 1 Illustration depicting mammalian spermatogenesis and the effect of a UPF2 or TDRD6 knockout on two different types of NMD-targeted mRNAs. Precursors of chromatid bodies (CBs: red) begin to form in late-stage spermatocytes at the end of meiosis and condense into the typical CB structure in early round spermatids. CBs are enriched in different classes of RNA, NMD factors, and additional proteins, including TDRD6 and MHV. Both papers report that the NMD factors UPF1 and UPF2 are highly expressed in postmeiotic spermatocytes and spermatids, where they are found in germ cell-specific perinuclear structures called chromatin bodies (CBs). CBs consist of RNA, many RNA-binding proteins, helicases, and several members of the TUDOR-domain protein family (TDRDs); furthermore, CBs are best known for their role in piRNA biogenesis [ 13 , 14 ]. A conditional UPF2 knockout in spermatogonia led to infertile mice with small testes [ 11 ], resembling the “Sertoli-only syndrome” in humans; ablation of TDRD6, which disrupts CB formation and arrests spermatogenesis, gave a similar phenotype. Transcriptome profilings of UPF2 or TDRD6 knockout spermatocytes and round spermatids revealed in both cases a strong enrichment of transcripts with long 3′UTRs among the up-regulated RNAs, while the classical NMD targets (defined by the presence of an exon–exon junction >50 nucleotides downstream of the termination codon) were largely unaffected [ 11 , 12 ]. The UPF2 knockout data is reminiscent of a previous study showing that NMD induced by the presence of exon junction complex (EJC) factors eIF4A3, Y14, and MAGOH downstream of the PTC does not require UPF2 [ 15 ]. Together with the results from Bao and colleagues [ 11 ], this finding provides evidence for the existence of mechanistically different modes of NMD acting on different types of transcripts and, in particular, strongly suggests a UPF2-independent route of NMD for transcripts with EJCs in the 3′UTR. Because UPF2 is thought to play a crucial role in NMD by promoting the SMG1-mediated phosphorylation of UPF1 [ 5 ], it remains, however, unclear how UPF1 phosphorylation could be achieved in the postulated UPF2-independent NMD mode. That the TDRD6 knockout affects the transcriptome in the same way as the UPF2
机译:前MRNA的广泛的替代剪接和多腺苷酸不仅扩大了我们基因组的蛋白质编码势,而且还产生了具有不同3'未翻译区(UTRS)[1,2]的MRNA同种型。由于3'UTR是MRNA稳定性,本地化和翻译的主要调节因子,因此组织特异性,发育调节和应力诱导的替代3'UTR的产生极大地有助于基因表达的前剖析调节。它是一种有趣的观察,即具有最长的3'UTRS的MRNA主要存在于大脑中,而睾丸在具有较短的3'UTRS [3,4]的mRNA同种型中。通常,具有较短3'UTR的MRNA趋于更稳定,因为(i)含有较少的结合位点,用于衰减诱导的miRNA或RNA结合蛋白(RBPS)和(II)长3'UTRS可以引发胡说八道介导的mRNA衰减(NMD)[1,5]。术语NMD最初注入,以描述用于MRNA的加速降解与过早截断开放阅读框(ORF)[6]的无意义突变。 NMD通过减少潜在有害的C末端截短的蛋白质的生产提供了重要的细胞质量控制功能。然而,基因组研究发现,除了降低的异常MRNA患有过早翻译终止密码子(PTCS),NMD还靶向许多“正常”MRNA编码明显全长官能蛋白,这表明术后基因调节中的更广泛的生物学功能[5,7 ]。在哺乳动物细胞中NMD需要三个保守的核心因子(UPF1,UPF2和UPF3)和另外的甲卓特异性蛋白质,但NMD的确切分子机制尚不清楚,效率低或异常翻译终止似乎是关键触发对于NMD。经验证明,终止密码子下游的外显子外显子连接常常触发NMD,其通常是含异常PTC的转录物的情况,并且长3'UTR也可以引发NMD,其中包含NMD许多无铅NMD靶标​​[8 - 10]。这两组NMD基底是否被共同机制识别和降解,或者它们是否采用纽姆的机械主义不同分支是该领域的主要未解决的问题。两项关于小鼠精子发生的新研究[11,12],都在这个问题上发表的PLOS遗传学,现在在这个问题上揭示了一些意想不到的新光。在精子发生期间,精子寄生虫分为精子胶质细胞,其经过分数突和后生物地发展成圆形,另外,细长的精子(图1)。 10.1371 / journal.pgen1005979.g001图1图1插图描绘了哺乳动物精子发生和UPF2或TDRD6敲除在两种不同类型的NMD靶向MRNA上的影响。染色体体的前体(CBS:RED)开始在减数分裂结束时在后期精子细胞中形成,并凝结到早期精子的典型CB结构中。 CBS在不同类别的RNA,NMD因子和其他蛋白质中富集,包括TDRD6和MHV。两篇论文都报告说,NMD因子UPF1和UPF2在后霉菌精胶质细胞和精子中高度表达,在那里它们存在于称为染色质体(CBS)的胚芽细胞特异性蠕变结构中。 CBS由RNA,许多RNA结合蛋白,螺旋酶和巢域蛋白质家族(TDDD)的几个成员组成;此外,CBS最闻名于它们在PiRNA生物发生中的作用[13,14]。精子寄生虫的条件UPF2敲除导致具有小型睾丸的不孕小鼠[11],类似于人类的“唯一综合征”;消融TDRD6,其破坏CB形成和捕获精子发生,得到了类似的表型。在两种情况下,UPF2或TDRD6敲除精胶质细胞和圆形精子的转录组分布在上调的RNA中具有长3'UTR的转录物的强烈富集,而经典的NMD靶标​​(通过外显子连接> 50的存在定义在终止密码子下游的核苷酸在很大程度上不受影响[11,12]。 UPF2敲除数据使得先前的研究表明,通过PTC下游的外显子结络合物(EJC)因子EIF4A3,Y14和MAGOH诱导的NMD不需要UPF2 [15]。与BaO和同事的结果一起[11],该发现提供了存在于作用于不同类型的转录物的机械主义不同模式的证据,并且特别强烈地表明EJC的转录物的UPF2独立的NMD途径3'UTR。由于UPF2通过促进UPF1的SMG1介导的SMG1介导的磷酸化而在NMD中发挥至关重要的作用,但仍不清楚在假设的UPF2独立的NMD模式下可以实现UPF1磷酸化。 TDRD6敲除以与UPF2相同的方式影响转录组

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号