RARS2

RARS2
  • 文章类型: Journal Article
    线粒体精氨酰转移RNA合成酶(RARS2)基因中的双等位基因变体已参与分类为6型小脑小脑发育不全(PCH)和癫痫性脑病的早发性脑病。一个变体(NM_020320.3:c。-2A>G)先前已在PCH家族中鉴定出RARS2基因的启动子和5'UTR。仅检测到该变体对mRNA水平的轻微影响。由于RARS2是非剂量敏感的,这一观察结果对该变异体的致病性尚无定论.我们在这里报告和描述了一个新的患者,在RARS2基因中具有相同的变异,在纯合状态。尽管没有乳酸性酸中毒,但该患者的临床表型与PCH6一致。与先前的研究一致,我们测量了患者成纤维细胞中的RARS2mRNA水平,并检测到与对照组相比部分保留的基因表达。重要的是,该变体位于控制翻译起始的Kozak序列中。因此,我们使用生物信息学方法和蛋白质印迹法研究了对蛋白质翻译的影响.我们在这里展示了这个变体,除了它对转录的影响,也破坏了一致的Kozak序列,并对RARS2蛋白翻译有主要影响。通过对这一额外病例的鉴定和分子后果的表征,我们阐明了这种Kozak变体在PCH和蛋白质合成中的参与。这项工作还指出了位于翻译起始区的变体的致病性预测的当前局限性。
    Bi-allelic variants in the mitochondrial arginyl-transfer RNA synthetase (RARS2) gene have been involved in early-onset encephalopathies classified as pontocerebellar hypoplasia (PCH) type 6 and in epileptic encephalopathy. A variant (NM_020320.3:c.-2A > G) in the promoter and 5\'UTR of the RARS2 gene has been previously identified in a family with PCH. Only a mild impact of this variant on the mRNA level has been detected. As RARS2 is non-dosage-sensitive, this observation is not conclusive in regard of the pathogenicity of the variant.We report and describe here a new patient with the same variant in the RARS2 gene, at the homozygous state. This patient presents with a clinical phenotype consistent with PCH6 although in the absence of lactic acidosis. In agreement with the previous study, we measured RARS2 mRNA levels in patient\'s fibroblasts and detected a partially preserved gene expression compared to control. Importantly, this variant is located in the Kozak sequence that controls translation initiation. Therefore, we investigated the impact on protein translation using a bioinformatic approach and western blotting. We show here that this variant, additionally to its effect on the transcription, also disrupts the consensus Kozak sequence, and has a major impact on RARS2 protein translation. Through the identification of this additional case and the characterization of the molecular consequences, we clarified the involvement of this Kozak variant in PCH and on protein synthesis. This work also points to the current limitation in the pathogenicity prediction of variants located in the translation initiation region.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    前脑小脑发育不全(PCH)是一种异质性神经发育障碍,其特征是脑干和小脑体积减少。前脑小脑发育不全6型(PCH6)是一种与常染色体隐性遗传相关的线粒体疾病,由RARS2基因突变引起。在这个案例报告中,我们描述了一个新的临床表现与一个新的RARS2致病变异。我们在这里报道了2位出现新生儿乳酸性酸中毒的兄弟姐妹,小头畸形,生长迟缓,持续性癫痫发作,和具有先前未定义的RARS2致病变体的胆汁淤积。在我们的文献综述中,自2007年首次发现PCH6中的RARS2致病变异以来,我们评估了16篇出版物报道的34例患者的临床特征和致病变异.检测到两个兄弟姐妹的c.1564G>A(p。Val522Ile),RARS2基因的新型纯合致病变体。影像学显示晚期脑萎缩和小脑发育不全,同时保留了基底神经节和脑桥。在后续行动中,肝功能检查结果和胆汁淤积的升高已经消退,而LDH和GGT的升高持续.两个兄弟姐妹在随访中都表现出小头畸形,并开始癫痫发作。观察到严重的发育迟缓和营养问题,都死于婴儿期.RARS2致病变异是一种线粒体疾病,可导致严重的精神,电机,和发育迟缓,以及预期寿命短。我们的患者是PCH6肝脏受累的首例病例,并且是文献中报道的新型纯合RARS2致病变体。这种额外的表型可以被认为是对文献的有效贡献。
    Pontocerebellar hypoplasia (PCH) is a heterogeneous neurodevelopmental disorder that is characterized by decreased brainstem and cerebellum volume. Pontocerebellar hypoplasia type 6 (PCH6) is a mitochondrial disease associated with autosomal recessive inheritance that results from mutations in the RARS2 gene. In this case report, we describe a new clinical presentation with a novel RARS2 pathogenic variant. We report here on 2 siblings who presented with neonatal lactic acidosis, microcephaly, growth retardation, persistent seizures, and cholestasis with a previously undefined RARS2 pathogenic variant. In our literature review, we evaluated the clinical features and pathogenic variants of 34 patients reported in 16 publications since the initial identification of RARS2 pathogenic variants in PCH6 in 2007. Both siblings were detected with c.1564G>A (p.Val522Ile), a novel homozygous pathogenic variant of the RARS2 gene. Imaging revealed advanced cerebral atrophy and cerebellar hypoplasia, while the basal ganglia and pons were preserved. At follow-up, the elevations in liver function test results and cholestasis had regressed while the LDH and GGT elevations persisted. Both siblings showed microcephaly on follow-up and started to suffer seizures. Severe developmental delay and nutritional problems were observed, and both died in infancy. RARS2 pathogenic variant is a mitochondrial disease that causes severe mental, motor, and developmental retardation, as well as short life expectancy. Our patients are the first cases with liver involvement in PCH6 and a novel homozygous RARS2 pathogenic variant to be reported in the literature. This additional phenotype can be considered as making a valid contribution to the literature.
    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
    前脑小脑发育不全6型(PCH6)是一种早发性脑病,有/没有线粒体呼吸复合物缺陷,由线粒体精氨酰-tRNA合成酶(RARS2)的隐性突变引起。高度异质性的临床表型和许多意义不确定的错义变化使诊断变得困难。PCH6的发病机制尚不清楚。
    评估患者的面部特征。进行基因测试。结构预测基于来自AlphaFold蛋白质结构数据库的模板。在HEK293T细胞中测试突变体RARS2的表达。检测患者来源的诱导多能干细胞(iPSCs)的人线粒体tRNAArg(hmtRNAArg)稳态水平,线粒体呼吸复合体(MRC)活性,耗氧率(OCR),细胞外酸化率(ECAR),线粒体膜电位(MMP),活性氧(ROS)丰度,和凋亡水平。
    三个家系被诊断为由复合杂合RARS2变异引起的PCH6。鉴定了五种RARS2变体:c.3G>C(p。M1?),c.685C>T(p。R229*),c.1060T>A(p。F354I),c.1210A>G(p。M404V),和c.1369G>A(p。G457R)。RARS2c.3G>C破坏的蛋白质表达。RARS2c.685C>T产生了一种截短的蛋白质,缺乏完整的催化核心和反密码子结合结构域。预测RARS2c.1060T>A和c.1369G>A会导致结构异常。患者iPSCs中hmtRNAArg稳态丰度未受影响。线粒体能量代谢正常,包括MRC活动,OCR,ECAR,和MMP,而线粒体相关的细胞特征,包括ROS(P<0.001)和凋亡水平(P<0.001),增加。
    本研究报告了五种RARS2变体,其中c.3G>C和c.1060T>A是新颖的。总结PCH6患者的面部特征将有助于诊断。线粒体能量代谢缺陷可能不是关键点,但是线粒体相关的异常细胞生理学,包括细胞凋亡,可能是潜在的发病机制。
    Pontocerebellar hypoplasia type 6 (PCH6) is an early-onset encephalopathy with/without mitochondrial respiratory complex defects caused by recessive mutations in mitochondrial arginyl-tRNA synthetase (RARS2). Highly heterogeneous clinical phenotypes and numerous missense variations of uncertain significance make diagnosis difficult. Pathogenesis of PCH6 remains unclear.
    Facial characteristics of patients were assessed. Genetic tests were performed. Structure prediction was based on the template from AlphaFold Protein Structure Database. Expression of mutant RARS2 was tested in HEK293T cells. Patient-derived induced pluripotent stem cells (iPSCs) were detected for human mitochondrial tRNAArg (hmtRNAArg) steady-state level, mitochondrial respiratory complex (MRC) activity, oxygen consumption rate (OCR), extracellular acidification rate (ECAR), mitochondrial membrane potential (MMP), reactive oxygen species (ROS) abundance, and apoptosis level.
    The three pedigrees were diagnosed as PCH6 caused by compound heterozygous RARS2 variations. Five RARS2 variants were identified: c.3G>C(p.M1?), c.685C>T(p.R229∗), c.1060T>A(p.F354I), c.1210A>G(p.M404V), and c.1369G>A(p.G457R). RARS2 c.3G>C disrupted protein expression. RARS2 c.685C>T created a truncated protein lacking complete catalytic core and anticodon-binding domain. RARS2 c.1060T>A and c.1369G>A were predicted to cause structural abnormality. The hmtRNAArg steady-state abundance in a patient\'s iPSCs was unaffected. Mitochondrial energy metabolism was normal, including MRC activity, OCR, ECAR, and MMP, while mitochondria-related cellular characteristics, including ROS (P < 0.001) and apoptosis levels (P < 0.001), increased.
    This study reports five RARS2 variations among which c.3G>C and c.1060T>A are novel. Summarized facial features of PCH6 patients will facilitate diagnosis. Defective mitochondrial energy metabolism may not be key points, but mitochondria-related abnormal cellular physiology, including apoptosis, may be an underlying pathogenesis.
    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
    Developmental and epileptic encephalopathies (DEE) encompass rare, sporadic neurodevelopmental disorders and usually with pediatric onset. As these conditions are characterized by marked clinical and genetic heterogeneity, whole-exome sequencing (WES) represents the strategy of choice for the molecular diagnosis. While its usefulness is well established in pediatric DEE cohorts, our study is aimed at assessing the WES feasibility in adult DEE patients who experienced a diagnostic odyssey prior to the advent of this technique. We analyzed exomes from 71 unrelated adult DEE patients, consecutively recruited from an Italian cohort for the EPI25 Project. All patients underwent accurate clinical and electrophysiological characterization. An overwhelming percentage (90.1%) had already undergone negative genetic testing. Variants were classified according to the American College of Medical Genetics and Genomics guidelines. WES disclosed 24 (likely) pathogenic variants among 18 patients in epilepsy-related genes with either autosomal dominant, recessive or X-linked inheritance. Ten of these were novel. We obtained a diagnostic yield of 25.3%, higher among patients with brain malformations, early-onset epilepsy and dysmorphisms. Despite a median diagnostic delay of 38.7 years, WES analysis provided the long-awaited diagnosis for 18 adult patients, which also had an impact on the clinical management of 50% of them.
    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: Case Reports
    Mitochondrial aminoacyl-tRNA synthetases play a major role in protein translation, synthesis, and oxidative phosphorylation. We reviewed all patients diagnosed with mitochondrial aminoacyl-tRNA synthetase deficiencies diagnosed in a single neurometabolic clinic. We report five patients with mitochondrial aminoacyl-tRNA synthetase deficiencies including DARS2, EARS2, PARS2, and RARS2 deficiencies. Siblings with DARS2 deficiency presented with global developmental delay within the first year of life. DARS2, EARS2, PARS2, and RARS2 deficiencies were identified by whole exome sequencing. We report coagulation factor abnormalities in PARS2 deficiency for the first time. We also report symmetric increased signal intensity in globus pallidi in FLAIR images in brain MRI in EARS2 deficiency for the first time. One patient with RARS2 deficiency had compound heterozygous variants in RARS2. One of those variants was an intronic variant. We confirmed the pathogenicity by mRNA studies. Mitochondrial aminoacyl-tRNA synthetase deficiencies are diagnosed by molecular genetic investigations. Clinically available non-invasive biochemical investigations are non-specific for the diagnosis of mitochondrial aminoacyl-tRNA synthetase deficiencies. A combination of brain MRI features and molecular genetic investigations should be undertaken to confirm the diagnosis of mitochondrial aminoacyl-tRNA synthetase deficiencies.
    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: Case Reports
    Pontocerebellar hypoplasia type 6 (PCH6) is a rare infantile-onset progressive encephalopathy caused by biallelic mutations in RARS2 that encodes the mitochondrial arginine-tRNA synthetase enzyme (mtArgRS). The clinical presentation overlaps that of PEHO syndrome (Progressive Encephalopathy with edema, Hypsarrhythmia and Optic atrophy). The proband presented with severe intellectual disability, epilepsy with varying seizure types, optic atrophy, axial hypotonia, acquired microcephaly, dysmorphic features and progressive cerebral and cerebellar atrophy and delayed myelination on MRI. The presentation had resemblance to PEHO syndrome but sequencing of ZNHIT3 did not identify pathogenic variants. Subsequent whole genome sequencing revealed novel compound heterozygous variants in RARS2, a missense variant affecting a highly conserved amino acid and a frameshift variant with consequent degradation of the transcript resulting in decreased mtArgRS protein level confirming the diagnosis of PCH6. Features distinguishing the proband\'s phenotype from PEHO syndrome were later appearance of hypotonia and elevated lactate levels in blood and cerebrospinal fluid. On MRI the proband presented with more severe supratentorial atrophy and lesser degree of abnormal myelination than PEHO syndrome patients. The study highlights the challenges in clinical diagnosis of patients with neonatal and early infantile encephalopathies with overlapping clinical features and brain MRI findings.
    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
    Pontocerebellar hypoplasia type 6 (PCH6) is a rare autosomal recessive disease that occurs due to mutations in the mitochondrial arginyl-tRNA synthetase 2 (RARS2) gene. To the best of our knowledge, 23 cases with relatively complete clinical data have been reported thus far. In the present study, a case with PCH6 caused by novel RARS2 mutations is described, in which distinct magnetic resonance imaging (MRI) features were identified. In addition, 23 PCH6 cases found in the literature were reviewed. Early onset hypotonia (43.48%), epileptic seizures (34.78%), encephalopathy (26.08%) and feeding difficulties (17.39%) were common initial symptoms of PCH6. During disease progression, the patients presented refractory epileptic seizures (94.12%), feeding problems (60.87%), severe developmental delay (100%), microcephaly (88.89%) and hyperlactacidemia (76.47%). The clinical features of the present patient were suggestive of PCH6, with early onset epilepsy, feeding difficulties, severe developmental delay, microcephaly, hearing loss and hyperlactacidemia. According to available MRI data from 20 reported cases with PCH6, the characteristic finding in MRI was pontocerebellar dysplasia or progressive cerebral/pontocerebellar atrophy in 16 cases, while 4 cases did not present pontocerebellar hypoplasia, and no basal ganglia involvement was observed in any of the cases. Distinctive MRI features were also identified in the present case, including pontocerebellar preservation after 1 year of age, as well as a high diffusion-weighted imaging signal suggesting intracellular edema in the cerebellar hemispheres, basal ganglia, thalamus and corpus callosum. Progressive loss of cerebral white matter and cortical volume were common features shared by all patients. In conclusion, in the present study, two novel heterozygous mutations were identified in RARS2, namely c.1718C>T(p.Thr573Ile) and c.991A>G (p.Ile331Val). Thus, the present case enriched the phenotypic and genotypic spectrum of the RARS2 mutations.
    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
    前脑小脑发育不全6型(PCH6)是一种由RARS2基因突变引起的线粒体疾病。RARS2编码线粒体精氨酰转移RNA合成酶,参与线粒体蛋白质翻译的酶。到目前为止,共有来自14个家庭的27名患者被报告。特征性临床特征包括新生儿乳酸性酸中毒,严重脑病,顽固性癫痫发作,喂养问题和严重的发育迟缓。大多数患者表现出典型的神经放射学异常,包括小脑发育不全和进行性桥小脑萎缩。
    我们描述了临床,在RARS2中具有新的纯合突变的2个兄弟姐妹的生化和分子特征。两名患者均出现乳酸性酸中毒。虽然年长的兄弟姐妹有严重的神经症状,伴有小头畸形,癫痫发作和发育迟缓,年龄较小的患者在2个月大时仍无神经系统症状.
    两名儿童的MRI研究均缺乏桥小脑受累。OXPHOS复合蛋白的表达在两个患者中均降低,而耗氧量增加。
    在RARS2突变的患者中,PCH6的特征性神经放射学异常,如疣和小脑发育不全和进行性桥小脑萎缩可能缺失。因此,RARS2测试也应在没有小脑发育不全但其他典型临床症状的患者中进行。
    Pontocerebellar hypoplasia type 6 (PCH6) is a mitochondrial disease caused by mutations in the RARS2 gene. RARS2 encodes mitochondrial arginyl transfer RNA synthetase, an enzyme involved in mitochondrial protein translation. A total of 27 patients from 14 families have been reported so far. Characteristic clinical features comprise neonatal lactic acidosis, severe encephalopathy, intractable seizures, feeding problems and profound developmental delay. Most patients show typical neuroradiologic abnormalities including cerebellar hypoplasia and progressive pontocerebellar atrophy.
    We describe the clinical, biochemical and molecular features of 2 siblings with a novel homozygous mutation in RARS2. Both patients presented neonatally with lactic acidosis. While the older sibling had severe neurological symptoms with microcephaly, seizures and developmental delay, the younger patient was still neurologically asymptomatic at the age of 2 months.
    MRI studies in both children lacked pontocerebellar involvement. The expression of the OXPHOS complex proteins was decreased in both patients, whereas oxygen consumption was increased.
    Characteristic neuroradiological abnormalities of PCH6 such as vermis and cerebellar hypoplasia and progressive pontocerebellar atrophy may be missing in patients with RARS2 mutations. RARS2 testing should therefore also be performed in patients without pontocerebellar hypoplasia but otherwise typical clinical symptoms.
    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
    Intellectual disability is a heterogeneous disease with many genes and mutations influencing the phenotype. Consanguineous families constitute a rich resource for the identification of rare variants causing autosomal recessive disease, due to the effects of inbreeding. Here, we examine three consanguineous Arab families, recruited in a quest to identify novel genes/mutations. All the families had multiple offspring with non-specific intellectual disability. We identified homozygosity (autozygosity) intervals in those families through SNP genotyping and whole exome sequencing, with variants filtered using Ingenuity Variant Analysis (IVA) software. The families showed heterogeneity and novel mutations in three different genes known to be associated with intellectual disability. These mutations were not found in 514 ethnically matched control chromosomes. p.G410C in WWOX, p.H530Y in RARS2, and p.I69F in C10orf2 are novel changes that affect protein function and could give new insights into the development and function of the central nervous system.
    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: Case Reports
    前脑小脑发育不全是一组异质性神经发育障碍,其特征是脑干和小脑体积减少。我们报告了两名男性兄弟姐妹,他们出现了早期婴儿阵挛性癫痫发作,然后在磁共振成像(MRI)上出现了与突出的孤立小脑发育不全/萎缩相关的婴儿痉挛。使用全外显子组测序技术,发现两者都是先前报道的复合杂合子,并且是编码线粒体精氨酰-tRNA合成酶2(RARS2)的基因中的一个新突变。该基因的突变已在桥脑小脑发育不全六型(PCH6)中得到经典描述,以早期(通常是棘手的)癫痫发作为特征的表型,严重的发育迟缓,进行性桥小脑萎缩.PCH6的电临床频谱广泛,包括许多癫痫发作类型:肌阵挛性,广义强直-阵挛性,和局灶性阵挛性癫痫发作.我们的报告扩展了PCH6疾病谱的特征,并将婴儿痉挛表现为相关的电临床表型。
    Pontocerebellar hypoplasia is a group of heterogeneous neurodevelopmental disorders characterized by reduced volume of the brainstem and cerebellum. We report two male siblings who presented with early infantile clonic seizures, and then developed infantile spasms associated with prominent isolated cerebellar hypoplasia/atrophy on magnetic resonance imaging (MRI). Using whole exome sequencing techniques, both were found to be compound heterozygotes for one previously reported and one novel mutation in the gene encoding mitochondrial arginyl-tRNA synthetase 2 (RARS2). Mutations in this gene have been classically described in pontocerebellar hypoplasia type six (PCH6), a phenotype characterized by early (often intractable) seizures, profound developmental delay, and progressive pontocerebellar atrophy. The electroclinical spectrum of PCH6 is broad and includes a number of seizure types: myoclonic, generalized tonic-clonic, and focal clonic seizures. Our report expands the characterization of the PCH6 disease spectrum and presents infantile spasms as an associated electroclinical phenotype.
    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