exonic variant

外显子变体
  • 文章类型: Journal Article
    背景:X连锁低磷酸盐血症(XLH,OMIM307800)是一种罕见的由PHEX基因变异体惹起的磷代谢杂乱。仅在DNA水平分析简单分类为错义或无义变体的许多变体。然而,越来越多的证据表明,这些变体中的一些可能会改变前mRNA的剪接,导致疾病。因此,本研究旨在使用生物信息学工具和小基因分析来确定PHEX变异对pre-mRNA剪接的影响。方法:我们分析了被描述为错义或无义变体的PHEX基因中的174个变体。最后,我们使用生物信息学工具选择了8个候选变异体,使用小基因检测系统评估它们对pre-mRNA剪接的影响.PHEX基因的互补DNA(cDNA)序列(RefSeqNM_000444.6)用作DNA变体编号的基础。结果:在八个候选变异中,三个被发现导致异常剪接。变体c.617T>Gp。(Leu206Trp)和c.621T>Ap。(Tyr207*)外显子5改变了前mRNA的剪接,由于外显子5中隐蔽剪接位点的激活,产生了缺少外显子5一部分的异常转录本,而外显子16中的变体c.1700G>Cp。(Arg567Pro)导致内含子16中隐蔽剪接位点的激活,导致内含子16的部分包含。结论:我们的研究采用了小基因系统,在无法获得患者mRNA样本的情况下,具有很大程度的灵活性来评估异常剪接模式,探讨外显子变体对pre-mRNA剪接的影响。根据上述实验结果,我们证明了在mRNA水平分析外显子变异的重要性.
    Background: X-linked hypophosphatemia (XLH, OMIM 307800) is a rare phosphorus metabolism disorder caused by PHEX gene variants. Many variants simply classified as missense or nonsense variants were only analyzed at the DNA level. However, growing evidence indicates that some of these variants may alter pre-mRNA splicing, causing diseases. Therefore, this study aimed to use bioinformatics tools and a minigene assay to ascertain the effects of PHEX variations on pre-mRNA splicing. Methods: We analyzed 174 variants in the PHEX gene described as missense or nonsense variants. Finally, we selected eight candidate variants using bioinformatics tools to evaluate their effects on pre-mRNA splicing using a minigene assay system. The complementary DNA (cDNA) sequence for the PHEX gene (RefSeq NM_000444.6) serves as the basis for DNA variant numbering. Results: Of the eight candidate variants, three were found to cause abnormal splicing. Variants c.617T>G p.(Leu206Trp) and c.621T>A p.(Tyr207*) in exon 5 altered the splicing of pre-mRNA, owing to the activation of a cryptic splice site in exon 5, which produced an aberrant transcript lacking a part of exon 5, whereas variant c.1700G>C p.(Arg567Pro) in exon 16 led to the activation of a cryptic splice site in intron 16, resulting in a partial inclusion of intron 16. Conclusion: Our study employed a minigene system, which has a great degree of flexibility to assess abnormal splicing patterns under the circumstances of patient mRNA samples that are not available, to explore the impact of the exonic variants on pre-mRNA splicing. Based on the aforementioned experimental findings, we demonstrated the importance of analyzing exonic variants at the mRNA level.
    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
    IV型胶原是基底膜的组成部分。COL4A1是编码IV型胶原蛋白的关键基因之一,会导致多种疾病。显然,影响剪接的显著比例的突变可直接引起疾病或促成疾病的易感性或严重性。这里,我们使用生物信息学程序分析了COL4A1基因中描述的外显子突变和内含子突变,并鉴定了可能通过小基因系统改变正常剪接模式的候选突变.我们鉴定了7种通过破坏正常剪接位点诱导剪接改变的变体,创造新的,或改变剪接调节元件。预计这些突变会影响蛋白质功能。我们的结果有助于COL4A1变体的正确分子表征,并可能有助于开发更个性化的治疗方案。
    Type IV collagen is an integral component of basement membranes. Mutations in COL4A1, one of the key genes encoding Type IV collagen, can result in a variety of diseases. It is clear that a significant proportion of mutations that affect splicing can cause disease directly or contribute to the susceptibility or severity of disease. Here, we analyzed exonic mutations and intronic mutations described in the COL4A1 gene using bioinformatics programs and identified candidate mutations that may alter the normal splicing pattern through a minigene system. We identified seven variants that induce splicing alterations by disrupting normal splice sites, creating new ones, or altering splice regulatory elements. These mutations are predicted to impact protein function. Our results help in the correct molecular characterization of variants in COL4A1 and may help develop more personalized treatment options.
    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:X连锁Alport综合征(XLAS)是一种遗传性肾脏疾病,由染色体Xq22上的COL4A5罕见变异引起。许多研究表明,外显子中的单核苷酸变体(SNV)可以通过改变各种剪接调节信号来破坏前mRNA的正常剪接过程。男性XLAS患者具有很强的基因型-表型相关性。证实变异体对剪接的影响有助于预测肾脏预后。本研究旨在探讨单核苷酸取代是否,位于COL4A5基因中外显子5'末端或外显子内部位置的三个碱基内,导致剪接过程异常。
    方法:我们通过生物信息学程序分析了COL4A5基因中以前推测的401个SNV错义和无义变体,并通过小基因测定鉴定了可能影响pre-mRNA剪接的候选变体。
    结果:我们的研究表明8个候选变异中的3个可诱导完全或部分外显子跳跃。变体c.2678G>C和c.2918G>A可能会干扰经典的剪接位点,导致相应的外显子跳跃。变体c.3700C>T可以破坏伴随剪接沉默子序列产生的剪接增强子基序,导致外显子41的跳跃。
    结论:我们的研究表明,两个错义变体位于COL4A5外显子5'末端的第一个核苷酸,一个内部外显子无义变体导致异常剪接。重要的是,这项研究强调了在mRNA水平上评估SNV效应的必要性.
    BACKGROUND: X-linked Alport syndrome (XLAS) is an inherited renal disease caused by rare variants of COL4A5 on chromosome Xq22. Many studies have indicated that single nucleotide variants (SNVs) in exons can disrupt normal splicing process of the pre-mRNA by altering various splicing regulatory signals. The male patients with XLAS have a strong genotype-phenotype correlation. Confirming the effect of variants on splicing can help to predict kidney prognosis. This study aimed to investigate whether single nucleotide substitutions, located within three bases at the 5\' end of the exons or internal position of the exons in COL4A5 gene, cause aberrant splicing process.
    METHODS: We analyzed 401 SNVs previously presumed missense and nonsense variants in COL4A5 gene by bioinformatics programs and identified candidate variants that may affect the splicing of pre-mRNA via minigene assays.
    RESULTS: Our study indicated three of eight candidate variants induced complete or partial exon skipping. Variants c.2678G>C and c.2918G>A probably disturb classic splice sites leading to corresponding exon skipping. Variant c.3700C>T may disrupt splicing enhancer motifs accompanying with generation of splicing silencer sequences resulting in the skipping of exon 41.
    CONCLUSIONS: Our study revealed that two missense variants positioned the first nucleotides of the 5\' end of COL4A5 exons and one internal exonic nonsense variant caused aberrant splicing. Importantly, this study emphasized the necessity of assessing the effects of SNVs at the mRNA level.
    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
    囊肿是一种严重的,由CTNS基因变异引起的单基因全身性疾病。目前,越来越多的证据表明,许多疾病中的外显子变异可以影响pre-mRNA剪接。由于缺乏RNA水平的常规研究,可能低估了CTNS基因外显子变体对剪接调节的影响。这里,我们使用生物信息学工具分析了CTNS基因中的59个外显子变体,并通过小基因分析确定了可能诱导剪接改变的候选变体.我们通过破坏外显子剪接增强子/外显子剪接沉默(ESEs/ESSs)的比例或通过干扰经典剪接位点的识别,鉴定了6种诱导剪接改变的外显子变体,或者两者兼而有之。我们的研究结果有助于对胱氨酸病的变异进行正确的分子表征,并为新兴的治疗方法提供信息。此外,我们的工作表明,计算机模拟和体外分析的结合有助于评估驱动罕见遗传疾病的DNA变异对剪接调节的影响,并将增强变异功能注释的临床效用.
    Cystinosis is a severe, monogenic systemic disease caused by variants in CTNS gene. Currently, there is growing evidence that exonic variants in many diseases can affect pre-mRNA splicing. The impact of CTNS gene exonic variants on splicing regulation may be underestimated due to the lack of routine studies at the RNA level. Here, we analyzed 59 exonic variants in the CTNS gene using bioinformatics tools and identified candidate variants that may induce splicing alterations by minigene assays. We identified six exonic variants that induce splicing alterations by disrupting the ratio of exonic splicing enhancers/exonic splicing silencers (ESEs/ESSs) or by interfering with the recognition of classical splice sites, or both. Our results help in the correct molecular characterization of variants in cystinosis and inform emerging therapies. Furthermore, our work suggests that the combination of in silico and in vitro assays facilitates to assess the effects of DNA variants driving rare genetic diseases on splicing regulation and will enhance the clinical utility of variant functional annotation.
    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:常染色体显性多囊肾病(ADPKD)是一种常见的单基因多系统疾病,主要由PKD1基因或PKD2基因突变引起。越来越多的证据表明,其中一些变体,被描述为误解,文献或数据库中的同义或无义突变,通过影响前mRNA剪接过程可能是有害的。
    结果:本研究旨在结合预测性生物信息学工具和小基因测定确定这些PKD1和PKD2变体对外显子剪接的影响。因此,在19个候选单核苷酸改变中,11个变体分布在PKD1中(c.7866C>A,c.7960A>G,c.7979A>T,c.7987C>T,c.11248C>G,c.11251C>T,c.11257C>G,c.11257C>T,c.11346C>T,和c.11393C>G)和PKD2(c.1480G>T)被鉴定为导致外显子跳跃。
    结论:我们证实了PKD1和PKD2基因中的11个变异体通过干扰经典剪接位点的识别或通过破坏外显子剪接增强子并产生外显子剪接沉默来影响正常剪接。考虑到近年来在PKD1和PKD2基因中鉴定的变异数量的增加,这是迄今为止关于ADPKD相关致病基因中外显子变异体的前mRNA剪接的最全面的研究。这些结果强调了在mRNA水平上评估外显子单核苷酸变体在ADPKD中的作用的重要性。
    BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is a common monogenic multisystem disease caused primarily by mutations in the PKD1 gene or PKD2 gene. There is increasing evidence that some of these variants, which are described as missense, synonymous or nonsense mutations in the literature or databases, may be deleterious by affecting the pre-mRNA splicing process.
    RESULTS: This study aimed to determine the effect of these PKD1 and PKD2 variants on exon splicing combined with predictive bioinformatics tools and minigene assay. As a result, among the 19 candidate single nucleotide alterations, 11 variants distributed in PKD1 (c.7866C > A, c.7960A > G, c.7979A > T, c.7987C > T, c.11248C > G, c.11251C > T, c.11257C > G, c.11257C > T, c.11346C > T, and c.11393C > G) and PKD2 (c.1480G > T) were identified to result in exon skipping.
    CONCLUSIONS: We confirmed that 11 variants in the gene of PKD1 and PKD2 affect normal splicing by interfering the recognition of classical splicing sites or by disrupting exon splicing enhancers and generating exon splicing silencers. This is the most comprehensive study to date on pre-mRNA splicing of exonic variants in ADPKD-associated disease-causing genes in consideration of the increasing number of identified variants in PKD1 and PKD2 gene in recent years. These results emphasize the significance of assessing the effect of exon single nucleotide variants in ADPKD at the mRNA level.
    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:Gitelman综合征(GS)是一种脱盐肾小管疾病,其中大部分是由SLC12A3基因变异引起的,错觉变体占大多数。最近,外显子跳跃的现象,其中变体破坏正常的pre-mRNA剪接,与多种疾病有关。因此,我们假设一定比例的SLC12A3变异体可通过干扰正常剪接过程导致疾病.
    方法:我们使用生物信息学程序分析了342个先前假定的SLC12A3错义变体,并通过小基因测定鉴定了可能改变前mRNA剪接的候选变体。
    结果:我们的研究表明,在十个候选变体中,六个变种(c.602G>A,c.602G>T,c.1667C>T,c.1925G>A,c.2548G>C,和c.2549G>C)通过影响外显子剪接调控元件和/或干扰规范剪接位点而导致完整或不完整的外显子跳跃。
    结论:值得一提的是,这是关于SLC12A3外显子变体的前mRNA剪接的最大研究。此外,我们的研究强调了在GSmRNA水平检测剪接功能的重要性,并表明小基因分析是体外变异体剪接功能分析的有价值的工具.
    Gitelman syndrome (GS) is a type of salt-losing tubular disease, most of which is caused by SLC12A3 gene variants, and missense variants account for the majority. Recently, the phenomenon of exon skipping, in which variants disrupt normal pre-mRNA splicing, has been related to a variety of diseases. Therefore, we hypothesize that a certain proportion of SLC12A3 variants can result in disease via interfering with the normal splicing process.
    We analyzed 342 previously presumed SLC12A3 missense variants using bioinformatics programs and identified candidate variants that may alter the splicing of pre-mRNA through minigene assays.
    Our study revealed that, among ten candidate variants, six variants (c.602G>A, c.602G>T, c.1667C>T, c.1925G>A, c.2548G>C, and c.2549G>C) led to complete or incomplete exon skipping by affecting exonic splicing regulatory elements and/or disturbing canonical splice sites.
    It is worth mentioning that this is the largest study on pre-mRNA splicing of SLC12A3 exonic variants. In addition, our study emphasizes the importance of detecting splicing function at the mRNA level in GS and indicates that minigene analysis is a valuable tool for splicing functional assays of variants in vitro.
    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
    原发性远端肾小管酸中毒(dRTA)是一种罕见的肾小管疾病,与SLC4A1,ATP6V0A4,ATP6V1B1,FOXⅠ1或WDR72基因的变异相关。目前,越来越多的证据表明,所有类型的外显子变体都可以改变剪接调节元件,影响前体信使RNA(pre-mRNA)剪接过程。这项研究旨在确定与dRTA相关的变体对pre-mRNA剪接的影响,并结合预测性生物信息学工具和小基因测定。因此,在15个候选变体中,7个变体分布在SLC4A1中(c.1765C>T,p.Arg589Cys),ATP6V1B1(c.368G>T,p.Gly123Val;c.370C>T,p.Arg124Trp;c.484G>T,p.Glu162*andc.1102G>A,p.Glu368Lys)和ATP6V0A4基因(c.322C>T,p.Gln108*andc.1572G>A,p.Pro524Pro)被鉴定为通过外显子剪接增强剂(ESE)的破坏和外显子剪接消音器的产生而导致完整或不完整的外显子跳跃,或干扰经典拼接部位的识别,或者两者兼而有之。据我们所知,这是对dRTA相关基因外显子变异体的mRNA前剪接的首次研究.这些结果突出了在mRNA水平上评估外显子变体作用的重要性,并表明小基因分析是评估剪接对体外变体作用的有效工具。
    Primary distal renal tubular acidosis (dRTA) is a rare tubular disease associated with variants in SLC4A1, ATP6V0A4, ATP6V1B1, FOXⅠ1, or WDR72 genes. Currently, there is growing evidence that all types of exonic variants can alter splicing regulatory elements, affecting the precursor messenger RNA (pre-mRNA) splicing process. This study was to determine the consequences of variants associated with dRTA on pre-mRNA splicing combined with predictive bioinformatics tools and minigene assay. As a result, among the 15 candidate variants, 7 variants distributed in SLC4A1 (c.1765C>T, p.Arg589Cys), ATP6V1B1 (c.368G>T, p.Gly123Val; c.370C>T, p.Arg124Trp; c.484G>T, p.Glu162* and c.1102G>A, p.Glu368Lys) and ATP6V0A4 genes (c.322C>T, p.Gln108* and c.1572G>A, p.Pro524Pro) were identified to result in complete or incomplete exon skipping by either disruption of exonic splicing enhancers (ESEs) and generation of exonic splicing silencers, or interference with the recognition of the classic splicing site, or both. To our knowledge, this is the first study on pre-mRNA splicing of exonic variants in the dRTA-related genes. These results highlight the importance of assessing the effects of exonic variants at the mRNA level and suggest that minigene analysis is an effective tool for evaluating the effects of splicing on variants in vitro.
    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
    家族性肾性糖尿症是一种由SLC5A2基因变异引起的罕见肾小管疾病。它们中的大多数是外显子变体,并且已被分类为错义变体。然而,越来越多的证据表明,这些变体中的一些可能通过影响pre-mRNA剪接过程而有害.因此,我们假设一定比例的SLC5A2外显子变体可以通过干扰pre-mRNA的正常剪接过程而导致疾病。
    我们使用生物信息学程序分析了77种先前描述的假定的SLC5A2错义变体,并通过小基因测定鉴定了可能改变pre-mRNA剪接的候选变体。
    我们的研究表明7个候选变体中有6个诱导剪接改变。变体c.216C>A,c.294C>A,c.886G>C,c.932A>G和c.92A>G可能会破坏剪接增强子基序并产生剪接沉默子序列,导致外显子3的跳跃。变体c.305C>T和c.1129G>A可能干扰导致外显子跳跃的剪接位点。
    据我们所知,我们报告,第一次,在前mRNA中产生改变的SLC5A2外显子变体。我们的研究加强了在mRNA水平上评估推定点变体的后果的重要性。此外,我们认为小基因功能分析对于评估SLC5A2外显子变异体对无患者RNA样本前mRNA剪接的影响可能是有价值的.
    UNASSIGNED: Familial renal glucosuria is a rare renal tubular disorder caused by SLC5A2 gene variants. Most of them are exonic variants and have been classified as missense variants. However, there is growing evidence that some of these variants can be detrimental by affecting the pre-mRNA splicing process. Therefore, we hypothesize that a certain proportion of SLC5A2 exonic variants can result in disease via interfering with the normal splicing process of the pre-mRNA.
    UNASSIGNED: We used bioinformatics programs to analyze 77 previously described presumed SLC5A2 missense variants and identified candidate variants that may alter the splicing of pre-mRNA through minigene assays.
    UNASSIGNED: Our study indicated six of 7 candidate variants induced splicing alterations. Variants c.216C > A, c.294C > A, c.886G > C, c.932A > G and c.962A > G may disrupt splicing enhancer motifs and generate splicing silencer sequences resulting in the skipping of exon 3. Variants c.305C > T and c.1129G > A probably disturb splice sites leading to exon skipping.
    UNASSIGNED: To our knowledge, we report, for the first time, SLC5A2 exonic variants that produce alterations in pre-mRNA. Our research reinforces the importance of assessing the consequences for putative point variants at the mRNA level. Additionally, we propose that minigenes function analysis may be valuable to evaluate the impact of SLC5A2 exonic variants on pre-mRNA splicing without patients\' RNA samples.
    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