deep intronic variant

深内含子变体
  • 文章类型: Journal Article
    使用next-gene-ration测序(NGS)技术的胚系DNA检测已成为诊断遗传性疾病的分析标准,包括癌症.它的使用越来越多,对正确的样品识别提出了很高的要求,优先变体的独立确认,以及它们的功能和临床解释。为了简化这些流程,我们使用相同的捕获组CZECANCA引入了基于DNA和RNA捕获的并行NGS,通常用于遗传性癌症易感性的DNA分析。这里,我们介绍了RNA样品处理的分析工作流程及其分析和诊断性能。并行DNA/RNA分析通过计算亲缘系数可以进行可靠的样品鉴定。基于RNA捕获的方法将大多数临床相关癌症易感性基因的转录靶标富集到允许分析鉴定的DNA变体对mRNA加工的影响的程度。通过比较面板和全外显子组RNA富集,我们证明了组织特异性基因表达模式与捕获组无关。此外,技术重复证实了测试RNA分析的高可重复性。我们得出的结论是,使用相同基因组的平行DNA/RNANGS是一种稳健且具有成本效益的诊断策略。在我们的设置中,它允许使用NextSeq500/550MidOutputKitv2.5(150个周期)在单次运行中对48个DNA/RNA对进行常规分析,并具有足够的覆盖率,以分析226种癌症易感性和候选基因。这种方法可以取代费力的桑格确认测序,增加测试周转时间,降低分析成本,并通过分析变体对mRNA加工的影响来改善对变体影响的解释。
    Germline DNA testing using the next-gene-ration sequencing (NGS) technology has become the analytical standard for the diagnostics of hereditary diseases, including cancer. Its increasing use places high demands on correct sample identification, independent confirmation of prioritized variants, and their functional and clinical interpretation. To streamline these processes, we introduced parallel DNA and RNA capture-based NGS using identical capture panel CZECANCA, which is routinely used for DNA analysis of hereditary cancer predisposition. Here, we present the analytical workflow for RNA sample processing and its analytical and diagnostic performance. Parallel DNA/RNA analysis allowed credible sample identification by calculating the kinship coefficient. The RNA capture-based approach enriched transcriptional targets for the majority of clinically relevant cancer predisposition genes to a degree that allowed analysis of the effect of identified DNA variants on mRNA processing. By comparing the panel and whole-exome RNA enrichment, we demonstrated that the tissue-specific gene expression pattern is independent of the capture panel. Moreover, technical replicates confirmed high reproducibility of the tested RNA analysis. We concluded that parallel DNA/RNA NGS using the identical gene panel is a robust and cost-effective diagnostic strategy. In our setting, it allows routine analysis of 48 DNA/RNA pairs using NextSeq 500/550 Mid Output Kit v2.5 (150 cycles) in a single run with sufficient coverage to analyse 226 cancer predisposition and candidate ge-nes. This approach can replace laborious Sanger confirmatory sequencing, increase testing turnaround, reduce analysis costs, and improve interpretation of the impact of variants by analysing their effect on mRNA processing.
    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: Case Reports
    大多数致病性DMD变体可通过标准的肌营养不良基因检测来检测和解释。然而,大约1~3%的肌营养不良患者在标准基因检测后仍然没有可检测到的DMD变异,最有可能是由于结构染色体重排和/或深内含子假外显子激活变异。这里,我们报道了一名疑似诊断为Becker型肌营养不良症(BMD)的男孩,该男孩在基于外显子DNA的标准基因检测后仍未发现DMD变异.在男孩中进行了肌营养不良蛋白mRNA研究和基因组Sanger测序,其次是在硅剪接分析。我们成功地在DMD基因中检测到一种新的深层内含子致病变异(c.2380+3317A>T),因此,通过增强隐蔽的供体剪接位点,导致新的肌营养不良蛋白假外显子激活。因此,该患者被遗传诊断为BMD。我们的病例报告进一步强调了深层内含子区域内致病剪接变异在遗传未诊断的肌营养不良蛋白病中的重要作用。
    Most pathogenic DMD variants are detectable and interpretable by standard genetic testing for dystrophinopthies. However, approximately 1∼3% of dystrophinopthies patients still do not have a detectable DMD variant after standard genetic testing, most likely due to structural chromosome rearrangements and/or deep intronic pseudoexon-activating variants. Here, we report on a boy with a suspected diagnosis of Becker muscular dystrophy (BMD) who remained without a detectable DMD variant after exonic DNA-based standard genetic testing. Dystrophin mRNA studies and genomic Sanger sequencing were performed in the boy, followed by in silico splicing analyses. We successfully detected a novel deep intronic disease-causing variant in the DMD gene (c.2380 + 3317A > T), which consequently resulting in a new dystrophin pseudoexon activation through the enhancement of a cryptic donor splice site. The patient was therefore genetically diagnosed with BMD. Our case report further emphasizes the significant role of disease-causing splicing variants within deep intronic regions in genetically undiagnosed dystrophinopathies.
    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
    ATP7A的遗传变异与一系列X连锁疾病有关。按照严重性的降序,这些是门克斯病,枕角综合征,和X连锁远端脊髓性肌萎缩症。经过30年的诊断调查,我们在一个主要受骨骼表型影响程度不同的家族中的四名男性中发现了一个深内含子ATP7A变体,以弯曲长骨为特征,活动受限的肘关节经常脱臼,粗糙的卷发,慢性腹泻,和运动协调困难。在临床重新评估后,对来自GenomicsEngland100,000基因组项目的全基因组测序数据进行分析,确定了一个深内含子ATP7A变体,SpliceAI预测具有适度的剪接效果。使用微型基因剪接测定法,我们确定内含子变体导致异常剪接。患者cDNA的Sanger测序显示ATP7A转录本具有外显子5跳跃,或包含一个新的内含子4伪外显子。在这两种情况下,预测会导致过早终止的移码。使用qPCR测定法对ATP7AmRNA转录本进行定量表明,大多数转录本(86.1%)具有非规范剪接,68.0%有外显子5跳跃,18.1%的人以小说伪外显子为特色。我们建议受影响的男性中表型的变异性是剪接的随机效应所致。这种深内含子变体,导致ATP7A剪接异常,扩展了对ATP7A相关疾病谱内含子变异的理解。
    Genetic variants in ATP7A are associated with a spectrum of X-linked disorders. In descending order of severity, these are Menkes disease, occipital horn syndrome, and X-linked distal spinal muscular atrophy. After 30 years of diagnostic investigation, we identified a deep intronic ATP7A variant in four males from a family affected to variable degrees by a predominantly skeletal phenotype, featuring bowing of long bones, elbow joints with restricted mobility which dislocate frequently, coarse curly hair, chronic diarrhoea, and motor coordination difficulties. Analysis of whole genome sequencing data from the Genomics England 100,000 Genomes Project following clinical re-evaluation identified a deep intronic ATP7A variant, which was predicted by SpliceAI to have a modest splicing effect. Using a mini-gene splicing assay, we determined that the intronic variant results in aberrant splicing. Sanger sequencing of patient cDNA revealed ATP7A transcripts with exon 5 skipping, or inclusion of a novel intron 4 pseudoexon. In both instances, frameshift leading to premature termination are predicted. Quantification of ATP7A mRNA transcripts using a qPCR assay indicated that the majority of transcripts (86.1 %) have non-canonical splicing, with 68.0 % featuring exon 5 skipping, and 18.1 % featuring the novel pseudoexon. We suggest that the variability of the phenotypes within the affected males results from the stochastic effects of splicing. This deep intronic variant, resulting in aberrant ATP7A splicing, expands the understanding of intronic variation on the ATP7A-related disease spectrum.
    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
    丙酸血症(PA)是由PCCA或PCCB变异引起的常染色体隐性代谢紊乱,丙酰辅酶A羧化酶(PCC)的两个亚基。某些氨基酸和奇数链脂肪酸的分解代谢需要PCC。在缺席的情况下,积累的有毒代谢物引起代谢性酸中毒,神经症状,多器官功能障碍和可能的死亡。PA的临床表现是高度可变的,典型的发病在新生儿或婴儿早期。我们遇到了两个家庭,他们的孩子被诊断患有PA。外显子组测序(ES)未能确定致病变异,我们进行了基因组测序(GS),证明了深内含子PCCB变体的纯合性。RNA分析确定该变体产生具有过早终止密码子的假外显子。父母是变异携带者,尽管其中三个由于第二个等位基因上常见的大良性内含子缺失而显示假纯合性。父母假定的纯合性值得特别注意,因为它一开始掩盖了致病变异,这只能通过RNA研究解决。到达快速诊断,无论是生化还是遗传,在指导救生护理方面至关重要,结束诊断冒险,并允许在随后的怀孕中进行家庭产前检查。这项研究证明了综合遗传研究在达到诊断的能力,利用GS和RNA分析克服ES限制并定义致病性。重要的是,它强调在分析基因组数据时应该考虑内含子缺失,这样假纯合性就不会被误解为真纯合性,和致病变异不会被错误标记为良性。
    Propionic acidemia (PA) is an autosomal recessive metabolic disorder caused by variants in PCCA or PCCB, both sub-units of the propionyl-CoA carboxylase (PCC) enzyme. PCC is required for the catabolism of certain amino acids and odd-chain fatty acids. In its absence, the accumulated toxic metabolites cause metabolic acidosis, neurologic symptoms, multi-organ dysfunction and possible death. The clinical presentation of PA is highly variable, with typical onset in the neonatal or early infantile period. We encountered two families, whose children were diagnosed with PA. Exome sequencing (ES) failed to identify a pathogenic variant, and we proceeded with genome sequencing (GS), demonstrating homozygosity to a deep intronic PCCB variant. RNA analysis established that this variant creates a pseudoexon with a premature stop codon. The parents are variant carriers, though three of them display pseudo-homozygosity due to a common large benign intronic deletion on the second allele. The parental presumed homozygosity merits special attention, as it masked the causative variant at first, which was resolved only by RNA studies. Arriving at a rapid diagnosis, whether biochemical or genetic, can be crucial in directing lifesaving care, concluding the diagnostic odyssey, and allowing the family prenatal testing in subsequent pregnancies. This study demonstrates the power of integrative genetic studies in reaching a diagnosis, utilizing GS and RNA analysis to overcome ES limitations and define pathogenicity. Importantly, it highlights that intronic deletions should be taken into consideration when analyzing genomic data, so that pseudo-homozygosity would not be misinterpreted as true homozygosity, and pathogenic variants will not be mislabeled as benign.
    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
    无眼症和小眼症(A/M)是最严重的先天性发育性眼部疾病。尽管基因组筛选技术取得了进步,超过一半的A/M患者没有接受分子诊断。我们包括来自巴基斯坦队列和未知分子基础的七个受A/M影响的近亲家庭。进行了FOXE3的单基因检测,然后对未解决的先证者进行基因组测序,以便为这些家庭建立遗传诊断。所有七个家庭都接受了基因诊断。鉴定的变体都是纯合的,分类为(可能)致病性,并存在于A/M相关基因中。靶向FOXE3测序揭示了四个家族中的两个先前报道的致病性FOXE3变体。在剩下的家庭里,基因组测序揭示了一种已知的致病性PXDN变体,VSX2中的一个新的13bp缺失,以及PXDN中的一个新的深内含子剪接变体。对显示严重剪接缺陷的PXDN剪接变体进行体外剪接测定。我们的研究证实了基因组测序作为A/M感染个体的诊断工具的实用性。此外,在PXDN中鉴定一种新的深层内含子致病变异体,突出了非编码变异体在A/M-疾病中的作用以及基因组测序对鉴定这类变异体的价值.
    Anophthalmia and microphthalmia (A/M) are among the most severe congenital developmental eye disorders. Despite the advancements in genome screening technologies, more than half of A/M patients do not receive a molecular diagnosis. We included seven consanguineous families affected with A/M from Pakistani cohort and an unknown molecular basis. Single gene testing of FOXE3 was performed, followed by genome sequencing for unsolved probands in order to establish a genetic diagnosis for these families. All seven families were provided with a genetic diagnosis. The identified variants were all homozygous, classified as (likely) pathogenic and present in an A/M-associated gene. Targeted FOXE3 sequencing revealed two previously reported pathogenic FOXE3 variants in four families. In the remaining families, genome sequencing revealed a known pathogenic PXDN variant, a novel 13bp deletion in VSX2, and one novel deep intronic splice variant in PXDN. An in vitro splice assay was performed for the PXDN splice variant which revealed a severe splicing defect. Our study confirmed the utility of genome sequencing as a diagnostic tool for A/M-affected individuals. Furthermore, the identification of a novel deep intronic pathogenic variant in PXDN highlights the role of non-coding variants in A/M-disorders and the value of genome sequencing for the identification of this type of variants.
    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
    罕见的常染色体显性遗传性脑疾病DLG4相关的突触病是由DLG4的从头变异引起的(编码PSD-95),其中大部分被预测为蛋白质截短。除了剪接位点变异,预测许多同义和错义的DLG4变体通过改变RNA剪接发挥其作用,尽管没有功能RNA研究,这些变体的致病性尚不确定。这里,我们描述了一个使用全基因组测序鉴定的具有深内含子DLG4变体(c.2105+235C>T)的小男孩。通过对来自外周血的RNA进行逆转录PCR,我们证明了DLG4mRNA表达在血液中是可检测的,并且深内含子变体产生了两种替代的DLG4转录本,其中之一包括假外显子。两种替代转录本都是框架外的,并预测会导致蛋白质截短,从而建立先证者的遗传诊断。这增加了有关深层内含子变异的致病潜力的证据,并强调了功能研究的重要性。即使在报告的组织特异性基因表达可能暗示其他情况的情况下。
    The rare autosomal dominant brain disorder DLG4-related synaptopathy is caused by de novo variants in DLG4 (encoding PSD-95), the majority of which are predicted to be protein-truncating. In addition to splice site variants, a number of synonymous and missense DLG4 variants are predicted to exert their effect through altered RNA splicing, although the pathogenicity of these variants is uncertain without functional RNA studies. Here, we describe a young boy with a deep intronic DLG4 variant (c.2105+235C>T) identified using whole genome sequencing. By using reverse-transcription PCR on RNA derived from peripheral blood, we demonstrate that DLG4 mRNA expression is detectable in blood and the deep intronic variant gives rise to two alternative DLG4 transcripts, one of which includes a pseudoexon. Both alternative transcripts are out-of-frame and predicted to result in protein-truncation, thereby establishing the genetic diagnosis for the proband. This adds to the evidence concerning the pathogenic potential of deep intronic variants and underlines the importance of functional studies, even in cases where reported tissue-specific gene expression might suggest otherwise.
    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:法布里病是由GLA基因变异导致的α-半乳糖苷酶A(GLA)活性不足引起的X连锁溶酶体贮积症,导致鞘糖脂积累和危及生命,多器官并发症。已经报道了大约50种变体导致GLA中的剪接异常。大多数是在规范的剪接位点内发现的,它们是高度保守的GT和AG剪接受体和供体二核苷酸,而三分之一位于规范剪接位点之外,很难解释它们的致病性。在这项研究中,我们的目的是研究位于GLA基因中非经典剪接位点的变异的遗传致病性。
    方法:13种变体,包括四个深内含子变体,选自人类基因变异数据库专业人员。我们进行了体外剪接测定以鉴定变体中的剪接异常。
    结果:GLA中所有候选的非规范剪接位点变异体均导致异常剪接。此外,除了一个变种是蛋白质截短的.四个深层内含子变异产生异常转录本,包括一个神秘的外显子,以及正常的成绩单,每个的比例以细胞特异性的方式不同。
    结论:使用体外剪接试验验证剪接效应对于确认致病性和确定与临床表型的关联是有用的。
    BACKGROUND: Fabry disease is an X-linked lysosomal storage disorder caused by insufficient α-galactosidase A (GLA) activity resulting from variants in the GLA gene, which leads to glycosphingolipid accumulation and life-threatening, multi-organ complications. Approximately 50 variants have been reported that cause splicing abnormalities in GLA. Most were found within canonical splice sites, which are highly conserved GT and AG splice acceptor and donor dinucleotides, whereas one-third were located outside canonical splice sites, making it difficult to interpret their pathogenicity. In this study, we aimed to investigate the genetic pathogenicity of variants located in non-canonical splice sites within the GLA gene.
    METHODS: 13 variants, including four deep intronic variants, were selected from the Human Gene Variant Database Professional. We performed an in vitro splicing assay to identify splicing abnormalities in the variants.
    RESULTS: All candidate non-canonical splice site variants in GLA caused aberrant splicing. Additionally, all but one variant was protein-truncating. The four deep intronic variants generated abnormal transcripts, including a cryptic exon, as well as normal transcripts, with the proportion of each differing in a cell-specific manner.
    CONCLUSIONS: Validation of splicing effects using an in vitro splicing assay is useful for confirming pathogenicity and determining associations with clinical phenotypes.
    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:苯丙酮尿症(PKU)是由PAH变异引起的常染色体隐性遗传先天性代谢紊乱。以前,在Sanger测序和多重连接依赖性探针扩增后,约5%的PKU患者仍未确诊.迄今为止,据报道,在100多个疾病相关基因中,有越来越多的致病性深层内含子变异.
    方法:在本研究中,我们对无明确基因诊断的PKU患者进行了PAH的全长测序,以研究PAH的深层内含子变异.
    结果:我们确定了五个深层内含子变体(c.1199502A>T,c.1065+241C>A,c.706+368T>C,c.706+531>C,和c.706+608A>C)。其中,c.1199502A>T变异在中国PKU中发现频率高,可能是热点PAH变异。c.706+531T>C和c.706+608A>C是扩展PAH的深内含子变体谱的两个新变体。
    结论:深度内含子变异致病性分析可进一步提高PKU患者的基因诊断。计算机预测和小基因分析是研究深层内含子变异的功能和影响的有力方法。全长基因扩增后的靶向测序是一种经济有效的检测小片段基因深层内含子变异的工具。
    Phenylketonuria (PKU) is an autosomal recessive congenital metabolic disorder caused by PAH variants. Previously, approximately 5% of PKU patients remained undiagnosed after Sanger sequencing and multiplex ligation-dependent probe amplification. To date, increasing numbers of pathogenic deep intronic variants have been reported in more than 100 disease-associated genes.
    In this study, we performed full-length sequencing of PAH to investigate the deep intronic variants in PAH of PKU patients without definite genetic diagnosis.
    We identified five deep intronic variants (c.1199+502A>T, c.1065+241C>A, c.706+368T>C, c.706+531>C, and c.706+608A>C). Of these, the c.1199+502A>T variant was found at high frequency and may be a hotspot PAH variant in Chinese PKU. c.706+531T>C and c.706+608A>C are two novel variants that extend the deep intronic variant spectrum of PAH.
    Deep intronic variant pathogenicity analysis can further improve the genetic diagnosis of PKU patients. In silico prediction and minigene analysis are powerful approaches for studying the functions and effects of deep intronic variants. Targeted sequencing after full-length gene amplification is an economical and effective tool for the detection of deep intron variation in genes with small fragments.
    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
    EMC1编码内质网(ER)膜蛋白复合物(EMC)的亚基1,参与膜蛋白生物合成的跨膜结构域插入酶。EMC1的变异被描述为全球发育迟缓的原因,低张力,皮质视觉障碍,通常,MRI扫描的脑萎缩。我们报告了一个患有严重的整体发育迟缓和进行性小脑萎缩的个体,其中外显子组测序鉴定了EMC1内含子3中的杂合必需剪接位点变异(NM_015047.3:c.287-1G>A)。全基因组测序(WGS)鉴定了EMC1的内含子20中的深层内含子变体(NM_015047.3:c.2588-771C>G),通过计算机程序很难预测该变体会破坏mRNA前剪接。逆转录PCR(RT-PCR)揭示了与c.2588-771C>G变体相关的假外显子的随机激活和c.287-1G>A变体引起的错误剪接。该案例强调了WGS和RNA研究在识别和评估深层内含子变体的可能致病性方面的实用性,并扩展了EMC1相关疾病的基因型和表型谱。
    EMC1 encodes subunit 1 of the endoplasmic reticulum (ER) membrane protein complex (EMC), a transmembrane domain insertase involved in membrane protein biosynthesis. Variants in EMC1 are described as a cause of global developmental delay, hypotonia, cortical visual impairment, and commonly, cerebral atrophy on MRI scan. We report an individual with severe global developmental delay and progressive cerebellar atrophy in whom exome sequencing identified a heterozygous essential splice-site variant in intron-3 of EMC1 (NM_015047.3:c.287-1G>A). Whole genome sequencing (WGS) identified a deep intronic variant in intron-20 of EMC1 (NM_015047.3:c.2588-771C>G) that was poorly predicted by in silico programs to disrupt pre-mRNA splicing. Reverse Transcription-PCR (RT-PCR) revealed stochastic activation of a pseudo-exon associated with the c.2588-771C>G variant and mis-splicing arising from the c.287-1G>A variant. This case highlights the utility of WGS and RNA studies to identify and assess likely pathogenicity of deep intronic variants and expands the genotypic and phenotypic spectrum of EMC1-related disorders.
    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
    这项研究的目的是检测KIF11相关视网膜病变患者的遗传性缺失,并描述其临床和遗传特征。我们招募了来自7个无关家庭的10名个体,这些个体在KIF11中具有致病性单等位基因变体。所有受试者均接受眼科评估和眼外表型评估,以及使用下一代测序的全面分子遗传分析。进行小基因测定以观察一种新的深内含子变体(DIV)和一种新的同义变体对前mRNA剪接的影响。我们在七个家系中检测到KIF11的6种新的不同致病变体。共分离分析和超深度测序结果表明,5个变体在5个家族(71%)中从头产生。功能验证显示,同义变体导致外显子跳跃,而DIV会导致假外显子(PE)包含在内。患者的表型表现出高度差异,两个家庭表现出不完整的外显率。观察所有患者的眼部表现和特征性面部特征,以及七名患者的小头畸形,五名患者的智力残疾,还有一个病人的淋巴水肿.KIF11相关视网膜病变的关键视网膜特征是视网膜褶皱,牵引性视网膜脱离,和脉络膜视网膜发育不良.所有七个先证者的视觉检测都比其他受影响的家庭成员更严重。我们的发现拓宽了KIF11变体的遗传谱。DIV解释了KIF11相关视网膜病变的罕见未解决病例。患者表现出可变的表型表现力和不完全的外显率,表明基因分析对KIF11相关视网膜病变患者的重要性。
    The purpose of this study was to detect the missing heritability of patients with KIF11-related retinopathy and to describe their clinical and genetic characteristics. We enrolled 10 individuals from 7 unrelated families harboring a pathogenic monoallelic variant in KIF11. All subjects underwent ophthalmic assessment and extraocular phenotype evaluations, as well as comprehensive molecular genetic analyses using next-generation sequencing. Minigene assays were performed to observe the effects of one novel deep intron variant (DIV) and one novel synonymous variant on pre-mRNA splicing. We detected 6 novel different disease-causing variants of KIF11 in the seven pedigrees. Co-segregation analysis and ultra-deep sequencing results indicated that 5 variants arose de novo in 5 families (71%). Functional validation revealed that the synonymous variant leads to an exon skip, while the DIV causes a pseudoexon (PE) inclusion. The patients presented with high variations in their phenotype, and two families exhibited incomplete penetrance. Ocular manifestations and characteristic facial features were observed in all patients, as well as microcephaly in seven patients, intellectual disability in five patients, and lymphedema in one patient. The key retinal features for KIF11-related retinopathy were retinal folds, tractional retinal detachment, and chorioretinal dysplasia. All seven probands had more severe visual detects than other affected family members. Our findings widen the genetic spectrum of KIF11 variants. DIV explained rare unresolved cases with KIF11-related retinopathy. The patients displayed a variable phenotype expressivity and incomplete penetrance, indicating the importance of genetic analysis for patients with KIF11-related retinopathy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号