Congenital disorders of glycosylation

先天性糖基化障碍
  • 文章类型: Journal Article
    先天性糖基化障碍(CDG)是一组罕见的,通常是由蛋白质和脂质糖基化紊乱引起的多系统遗传疾病。SSR4-CDG是一种非常罕见的,相对温和的CDG亚型,主要表现在男性身上。它是由SSR4基因的致病变异引起的,它位于X染色体上。SSR4(信号序列受体蛋白4)是转位相关蛋白(TRAP)复合物的一个亚基,蛋白质跨内质网膜易位所需的结构。SSR4的缺乏导致内质网中蛋白质的N-连接糖基化受到干扰。这里,我们回顾了最常见的临床,18个先前发表的个体的生化和遗传特征,并报告了4例新诊断为SSR4-CDG的病例,包括第一个受这种疾病影响的成年人。根据我们的评论,发育迟缓,说话延迟,智力残疾,肌张力减退,小头症和独特的面部特征是SSR4-CDG的关键症状,存在于所有受影响的个体中。尽管这些症状与许多其他神经发育障碍重叠,它们与其他临床特征的结合,并且受影响个体的面部外观非常可区分,使这种疾病成为潜在可识别的CDG类型。其他体征和症状包括无法茁壮成长,喂养困难,结缔组织受累,肠胃问题,骨骼异常,癫痫发作和,在某些情况下,显著的行为异常。由于缺乏对这种罕见疾病的认识,由于生化检测在受影响的个体中可能是正常的,大多数是通过基因研究诊断的,如全外显子组测序。有了这篇文章,我们扩大了SSR4-CDG的表型,包括心脏症状,喉异常,和远端血管扩张.我们还提供对成年早期预后的见解,并提供适当的管理和护理建议。我们强调非常需要因果疗法,以及解决这种疾病的多种症状的有效对症疗法。特别是,行为问题可严重影响SSR4-CDG患者的生活质量,需要特别关注.最后,我们的目标是改善对受影响家庭和治疗医生的指导和教育,并为该疾病的未来研究奠定基础.
    Congenital disorders of glycosylation (CDG) are a group of rare, often multi-systemic genetic disorders that result from disturbed protein and lipid glycosylation. SSR4-CDG is an ultra-rare, comparably mild subtype of CDG, presenting mostly in males. It is caused by pathogenic variants in the SSR4 gene, which is located on the X chromosome. SSR4 (signal sequence receptor protein 4) is a subunit of the translocon-associated protein (TRAP) complex, a structure that is needed for the translocation of proteins across the ER membrane. A deficiency of SSR4 leads to disturbed N-linked glycosylation of proteins in the endoplasmic reticulum. Here, we review the most common clinical, biochemical and genetic features of 18 previously published individuals and report four new cases diagnosed with SSR4-CDG, including the first adult affected by this disorder. Based on our review, developmental delay, speech delay, intellectual disability, muscular hypotonia, microcephaly and distinct facial features are key symptoms of SSR4-CDG that are present in all affected individuals. Although these symptoms overlap with many other neurodevelopmental disorders, their combination with additional clinical features, and a quite distinguishable facial appearance of affected individuals make this disorder a potentially recognizable type of CDG. Additional signs and symptoms include failure to thrive, feeding difficulties, connective tissue involvement, gastrointestinal problems, skeletal abnormalities, seizures and, in some cases, significant behavioral abnormalities. Due to lack of awareness of this rare disorder, and since biochemical testing can be normal in affected individuals, most are diagnosed through genetic studies, such as whole exome sequencing. With this article, we expand the phenotype of SSR4-CDG to include cardiac symptoms, laryngeal abnormalities, and teleangiectasia. We also provide insights into the prognosis into early adulthood and offer recommendations for adequate management and care. We emphasize the great need for causal therapies, as well as effective symptomatic therapies addressing the multitude of symptoms in this disease. In particular, behavioral problems can severely affect quality of life in individuals diagnosed with SSR4-CDG and need special attention. Finally, we aim to improve guidance and education for affected families and treating physicians and create a basis for future research in this disorder.
    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
    ALG13-先天性糖基化障碍(CDG),是由ALG13(OMIM300776)中的致病变体引起的罕见X连接CDG,其影响N连接的糖基化途径。受影响的个体在婴儿期表现为主要的神经系统表现。癫痫痉挛是ALG13-CDG的常见表现症状。其他常见的表型包括发育迟缓,癫痫发作,智力残疾,小头畸形,和低张力。ALG13-CDG的当前管理旨在解决患者的症状。迄今为止,据报道,ALG13-CDG患者不到100人。在这篇文章中,一个国际CDG专家组审查了所有报告的ALG13-CDG患者,并提出了ALG13-CDG的诊断和治疗指南.该指南基于最佳可用数据和专家意见。神经症状在ALG13-CDG的表型中占主导地位,其中癫痫性痉挛被证实是ALG13-CDG最常见的表现症状,与张力减退和发育迟缓有关。我们建议ACTH/泼尼松龙治疗应首先进行试验,其次是vigabatrin,然而,生酮饮食已被证明在ALG13-CDG中具有有希望的结果。为了优化医疗管理,我们还建议早期心脏,胃肠,骨骼,以及受影响患者的行为评估。
    ALG13-Congenital Disorder of Glycosylation (CDG), is a rare X-linked CDG caused by pathogenic variants in ALG13 (OMIM 300776) that affects the N-linked glycosylation pathway. Affected individuals present with a predominantly neurological manifestation during infancy. Epileptic spasms are a common presenting symptom of ALG13-CDG. Other common phenotypes include developmental delay, seizures, intellectual disability, microcephaly, and hypotonia. Current management of ALG13-CDG is targeted to address patients\' symptoms. To date, less than 100 individuals have been reported with ALG13-CDG. In this article, an international group of experts in CDG reviewed all reported individuals affected with ALG13-CDG and suggested diagnostic and management guidelines for ALG13-CDG. The guidelines are based on the best available data and expert opinion. Neurological symptoms dominate the phenotype of ALG13-CDG where epileptic spasm is confirmed to be the most common presenting symptom of ALG13-CDG in association with hypotonia and developmental delay. We propose that ACTH/prednisolone treatment should be trialed first, followed by vigabatrin, however ketogenic diet has been shown to have promising results in ALG13-CDG. In order to optimize medical management, we also suggest early cardiac, gastrointestinal, skeletal, and behavioral assessments in affected patients.
    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
    糖基化是一种关键的翻译后修饰,在几种生物过程中起着关键作用。比如免疫反应。糖基化的改变可以调节各种病理的进程,例如先天性糖基化疾病(CDG),一组160多种罕见和复杂的遗传疾病。虽然糖基化和免疫功能障碍之间的联系已经被认识到,大多数CDG的免疫参与在很大程度上仍未被探索,也知之甚少.在这项研究中,我们提供了主要免疫参与的12CDG的免疫功能障碍和临床表现的最新信息,根据国际免疫学会联盟(IUIS),分为6类先天免疫错误。全面回顾了磷酸核苷酸变位酶2(PMM2)-CDG-最常见的CDG-的免疫参与,强调在婴儿期和儿童期以及携带R141H的基因型中免疫问题的患病率较高。最后,使用PMM2-CDG作为模型,我们指出宿主细胞中异常的糖基化模式与可能有利的与微生物的相互作用之间的联系,这可能解释了对感染的更高敏感性。进一步表征CDG中的免疫病理学和不寻常的宿主-病原体粘附不仅可以改善免疫学护理标准,而且还可以为创新的预防措施和基于聚糖的靶向疗法铺平道路,这些疗法可以改善CDG患者的生活质量。
    Glycosylation is a critical post-translational modification that plays a pivotal role in several biological processes, such as the immune response. Alterations in glycosylation can modulate the course of various pathologies, such as the case of congenital disorders of glycosylation (CDG), a group of more than 160 rare and complex genetic diseases. Although the link between glycosylation and immune dysfunction has already been recognized, the immune involvement in most CDG remains largely unexplored and poorly understood. In this study, we provide an update on the immune dysfunction and clinical manifestations of the 12 CDG with major immune involvement, organized into 6 categories of inborn errors of immunity according to the International Union of Immunological Societies (IUIS). The immune involvement in phosphomannomutase 2 (PMM2)-CDG - the most frequent CDG - was comprehensively reviewed, highlighting a higher prevalence of immune issues during infancy and childhood and in R141H-bearing genotypes. Finally, using PMM2-CDG as a model, we point to links between abnormal glycosylation patterns in host cells and possibly favored interactions with microorganisms that may explain the higher susceptibility to infection. Further characterizing immunopathology and unusual host-pathogen adhesion in CDG can not only improve immunological standards of care but also pave the way for innovative preventive measures and targeted glycan-based therapies that may improve quality of life for people living with CDG.
    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
    甘露糖基-寡糖葡糖苷酶-先天性糖基化障碍(MOGS-CDG)由甘露糖基-寡糖葡糖苷酶(葡糖苷酶I)基因中的双等位基因突变决定。MOGS-CDG是一种罕见的疾病,影响N-Glycan(CDGII型)的加工,其特征是突出的神经系统受累,包括张力减退,发育迟缓,癫痫发作和运动障碍。据我们所知,到目前为止,已经发表了30例MOGS-CDG患者。我们描述了一个孩子,他是MOGS基因中两个新变体的复合杂合。他介绍了早期婴儿发育和癫痫性脑病(EI-DEE),但没有其他特定的全身受累和一线生化发现。除了前面描述的功能,病人出现了Hirschprung病,以前从未在MOGS-CDG患者中报告过。
    Mannosyl-oligosaccharide glucosidase - congenital disorder of glycosylation (MOGS-CDG) is determined by biallelic mutations in the mannosyl-oligosaccharide glucosidase (glucosidase I) gene. MOGS-CDG is a rare disorder affecting the processing of N-Glycans (CDG type II) and is characterized by prominent neurological involvement including hypotonia, developmental delay, seizures and movement disorders. To the best of our knowledge, 30 patients with MOGS-CDG have been published so far. We described a child who is compound heterozygous for two novel variants in the MOGS gene. He presented Early Infantile Developmental and Epileptic Encephalopathy (EI-DEE) in the absence of other specific systemic involvement and unrevealing first-line biochemical findings. In addition to the previously described features, the patient presented a Hirschprung disease, never reported before in individuals with MOGS-CDG.
    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: Review
    背景:NGLY1相关的先天性去糖基化疾病(CDDG1:OMIM#615273)是一种罕见的常染色体隐性遗传疾病,由糖蛋白降解过程中内质网功能受损引起。已经记录了CDDG1患者的神经认知功能障碍;然而,受影响个体的表型恶化仍然难以捉摸。
    方法:一名精神运动发育延迟的日本男孩从5岁开始出现共济失调运动,从12岁开始出现肌阵挛性癫痫发作。食欲减退,运动和认知能力下降在12岁时变得明显.电生理研究确定了肌阵挛性癫痫发作的阵发性放电和巨大的体感诱发电位。Perampanel可有效控制肌阵挛性癫痫发作。外显子组测序显示,该患者在NGLY1,NM_018297.4中携带复合杂合变体:c.857G>A和c.-17_12del,从母亲和父亲那里继承下来,分别。文献综述证实,在28.5%的癫痫患者中观察到肌阵挛性癫痫发作。没有其他患者出现进行性肌阵挛性癫痫或与NGLY1功能丧失相关的认知功能下降。
    结论:我们的数据提供了证据,一组CDDG1患者在长期临床过程中表现出缓慢进行性肌阵挛性癫痫和认知功能下降。
    BACKGROUND: NGLY1-associated congenital disorder of deglycosylation (CDDG1: OMIM #615273) is a rare autosomal recessive disorder caused by a functional impairment of endoplasmic reticulum in degradation of glycoproteins. Neurocognitive dysfunctions have been documented in patients with CDDG1; however, deteriorating phenotypes of affected individuals remain elusive.
    METHODS: A Japanese boy with delayed psychomotor development showed ataxic movements from age 5 years and myoclonic seizures from age 12 years. Appetite loss, motor and cognitive decline became evident at age 12 years. Electrophysiological studies identified paroxysmal discharges on myoclonic seizure and a giant somatosensory evoked potential. Perampanel was effective for controlling myoclonic seizures. Exome sequencing revealed that the patient carried compound heterozygous variants in NGLY1, NM_018297.4: c.857G > A and c.-17_12del, which were inherited from mother and father, respectively. A literature review confirmed that myoclonic seizures were observed in 28.5% of patients with epilepsy. No other patients had progressive myoclonic epilepsy or cognitive decline in association with loss-of-function variations in NGLY1.
    CONCLUSIONS: Our data provides evidence that a group of patients with CDDG1 manifest slowly progressive myoclonic epilepsy and cognitive decline during the long-term clinical course.
    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
    先天性糖基化障碍是由糖蛋白或糖脂的聚糖部分的合成或聚糖与蛋白质和脂质的结合的缺陷引起的具有异质临床表现的多系统疾病。DPAGT1(UDP-GlcNAc:磷酸氢磷N-乙酰葡糖胺-1-磷酸转移酶)是蛋白质N-糖基化所需的氢磷连接寡糖生物合成途径中的起始蛋白。DPAGT1(UDP-GlcNAc:磷酸氢磷N-乙酰葡糖胺-1-磷酸转移酶)基因中的致病变体引起一种罕见类型的先天性糖基化障碍,称为DPAGT1-CDG(以前的CDG-Ij)(OMIM#608093)。这是一种罕见的常染色体隐性遗传疾病或患有先天性肌无力综合征的轻度版本,称为DPAGT1-CMS。严重的病程伴有张力减退,白内障,骨骼畸形,难治性癫痫,智力残疾,全球发育迟缓,在大多数DPAGT1-CDG患者中已经描述了过早死亡.
    我们描述了两名DPAGT1基因变异的患者:一个8个月大的男孩,错觉DPAGT1:c.339T>G(p。Phe113Leu)新变体和一名13岁女性患者,具有复合杂合变体,DPAGT1:c.466C>T(p。Arg156Cys,R156C)和DPAGT1:c.161+5G>A.虽然8个月大的患者在1个月大的时候被诊断出患有先天性白内障,有畸形的发现,癫痫,另一名患者的临床症状出现较晚,但肌肉无力更为突出,行为障碍,畸形的发现,没有癫痫。
    胆碱酯酶抑制剂治疗对肌无力患者有效,支持DPAGT1缺乏作为潜在病因。我们开始对患者进行吡啶斯的明治疗,患者肌肉无力更明显,我们看到了它的好处.我们的目的是呈现我们的患者诊断为DPAGT1-CDG由于不同的变异在同一基因和不同的临床表现,治疗,并将其与文献中的其他患者进行比较。
    UNASSIGNED: Congenital glycosylation disorders are multisystem diseases with heterogeneous clinical manifestations caused by defects in the synthesis of the glycan moiety of glycoproteins or glycolipids or the binding of glycans to proteins and lipids. DPAGT1 (UDP-GlcNAc: dolichol phosphate N-acetylglucosamine-1-phosphotransferase) is an initiating protein in the biosynthetic pathway of dolichol-linked oligosaccharides required for protein N-glycosylation. Pathogenic variants in DPAGT1 (UDP-GlcNAc: dolichol phosphate N-acetylglucosamine-1-phosphotransferase) gene cause a rare type of congenital glycosylation disorder called DPAGT1-CDG (formerly CDG-Ij) (OMIM #608093). It is a rare autosomal recessive disease or a milder version with congenital myasthenic syndrome known as DPAGT1-CMS. A severe disease course with hypotonia, cataracts, skeletal deformities, resistant epilepsy, intellectual disability, global developmental delay, premature death has been described in most patients with DPAGT1-CDG.
    UNASSIGNED: We describe two patients with variants in the DPAGT1 gene: an 8-month-old boy with a homozygous, missense DPAGT1:c.339T>G (p.Phe113Leu) novel variant and a 13-year-old female patient with compound heterozygous variants, DPAGT1:c.466C>T (p.Arg156Cys, R156C) and DPAGT1:c.161+5G>A. While the 8-month-old patient was diagnosed with congenital cataract at the age of 1 month, had dysmorphic findings, and epilepsy, clinical symptoms in the other patient appeared later but with more prominent muscle weakness, behavioral disorder, dysmorphic findings, and no epilepsy.
    UNASSIGNED: Cholinesterase inhibitor therapy was found to be effective in patients against muscle weakness, supporting DPAGT1 deficiency as the underlying etiology. We started pyridostigmine treatment in our patient with more pronounced muscle weakness, and we saw its benefit. We aimed to present our patients diagnosed with DPAGT1-CDG due to different variants in the same gene and different clinical presentations, treatment and to compare them with other patients in 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: Review
    PGAP3的双等位基因致病变异导致一种罕见的糖基磷脂酰肌醇生物发生障碍,PGAP3-CDG。这种多系统状态表现为主要的神经表型,包括发育迟缓,智力残疾,癫痫发作,和高磷血症.这里,我们总结了来自CDG自然史研究的65名个体的表型,其中6名未报告的个体被确诊为PGAP3-CDG.在这种疾病中发现的常见其他特征包括大脑畸形,行为异常,腭裂,和特征性的面部特征。本报告旨在回顾遗传和代谢发现,并描述疾病的表型,同时强调必要的临床方法来改善这种罕见的CDG的管理。
    Biallelic pathogenic variants in PGAP3 cause a rare glycosylphosphatidyl-inositol biogenesis disorder, PGAP3-CDG. This multisystem condition presents with a predominantly neurological phenotype, including developmental delay, intellectual disability, seizures, and hyperphosphatemia. Here, we summarized the phenotype of sixty-five individuals including six unreported individuals from our CDG natural history study with a confirmed PGAP3-CDG diagnosis. Common additional features found in this disorder included brain malformations, behavioral abnormalities, cleft palate, and characteristic facial features. This report aims to review the genetic and metabolic findings and characterize the disease\'s phenotype while highlighting the necessary clinical approach to improve the management of this rare CDG.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Systematic Review
    心力衰竭(HF)是一种进行性慢性疾病,仍然是全球死亡的主要原因。影响超过6400万患者。HF可由心肌病和具有单基因病因的先天性心脏缺陷引起。与心脏缺陷发展有关的基因和单基因疾病的数量不断增加,包括遗传代谢疾病(IMD)。已经报道了几种影响各种代谢途径的IMD表现为心肌病和心脏缺陷。考虑到糖代谢在心脏组织中的关键作用,包括能源生产,核酸合成和糖基化,越来越多的与碳水化合物代谢相关的IMD被描述为心脏表现并不奇怪.在这次系统审查中,我们提供了与呈现心肌病的碳水化合物代谢相关的IMD的全面概述,心律失常性疾病和/或结构性心脏缺陷。我们确定了58个表现为心脏并发症的IMD:糖/糖连接转运蛋白(GLUT3,GLUT10,THTR1)的3个缺陷;戊糖磷酸途径的2个障碍(G6PDH,TALDO);9种糖原代谢疾病(GAA,GBE1,GDE,GYG1,GYS1,LAMP2,RBCK1,PRKAG2,G6PT1);29先天性糖基化疾病(ALG3,ALG6,ALG9,ALG12,ATP6V1A,ATP6V1E1,B3GALTL,B3GAT3,COG1,COG7,DOLK,DPM3,FKRP,FKTN,GMPPB,MPDU1、NPL、PGM1,PIGA,PIGL,PIGN,PIGO,PIGT,PIGV,PMM2,POMT1,POMT2,SRD5A3,XYLT2);15种碳水化合物相关的溶酶体贮积病(CTSA,GBA1,GLA,GLB1,HEXB,IDUA,IDS,SGSH,NAGLU,HGSNAT,GNS,GALNS,ARSB,GUSB,ARSK).通过这项系统评价,我们旨在提高人们对碳水化合物相关IMD的心脏表现的认识,并提请人们注意可能导致心脏并发症的碳水化合物相关致病机制。
    Heart failure (HF) is a progressive chronic disease that remains a primary cause of death worldwide, affecting over 64 million patients. HF can be caused by cardiomyopathies and congenital cardiac defects with monogenic etiology. The number of genes and monogenic disorders linked to development of cardiac defects is constantly growing and includes inherited metabolic disorders (IMDs). Several IMDs affecting various metabolic pathways have been reported presenting cardiomyopathies and cardiac defects. Considering the pivotal role of sugar metabolism in cardiac tissue, including energy production, nucleic acid synthesis and glycosylation, it is not surprising that an increasing number of IMDs linked to carbohydrate metabolism are described with cardiac manifestations. In this systematic review, we offer a comprehensive overview of IMDs linked to carbohydrate metabolism presenting that present with cardiomyopathies, arrhythmogenic disorders and/or structural cardiac defects. We identified 58 IMDs presenting with cardiac complications: 3 defects of sugar/sugar-linked transporters (GLUT3, GLUT10, THTR1); 2 disorders of the pentose phosphate pathway (G6PDH, TALDO); 9 diseases of glycogen metabolism (GAA, GBE1, GDE, GYG1, GYS1, LAMP2, RBCK1, PRKAG2, G6PT1); 29 congenital disorders of glycosylation (ALG3, ALG6, ALG9, ALG12, ATP6V1A, ATP6V1E1, B3GALTL, B3GAT3, COG1, COG7, DOLK, DPM3, FKRP, FKTN, GMPPB, MPDU1, NPL, PGM1, PIGA, PIGL, PIGN, PIGO, PIGT, PIGV, PMM2, POMT1, POMT2, SRD5A3, XYLT2); 15 carbohydrate-linked lysosomal storage diseases (CTSA, GBA1, GLA, GLB1, HEXB, IDUA, IDS, SGSH, NAGLU, HGSNAT, GNS, GALNS, ARSB, GUSB, ARSK). With this systematic review we aim to raise awareness about the cardiac presentations in carbohydrate-linked IMDs and draw attention to carbohydrate-linked pathogenic mechanisms that may underlie cardiac complications.
    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
    背景:与ALG1相关的糖基化先天性疾病(ALG1-CDG)是一种罕见的常染色体隐性遗传疾病。由于ALG1基因致病变异引起的β1,4甘露糖基转移酶的缺乏,聚糖在蛋白质糖基化途径中的组装和加工受损,导致多器官受累的广泛临床谱。为了提高临床医生对其表现和基因型的认识,我们在此报道了一名ALG1基因新变异的新患者,并回顾了研究基因型-表型相关性的文献.
    方法:收集临床特征,和临床外显子组测序用于鉴定致病变异。MutationTaster,PyMol,和FoldX用于预测致病性,蛋白质三维模型分子结构的变化,以及由新颖变体引起的自由能变化。
    结果:先证者是一名13个月大的中国汉族男性,以癫痫发作为特征,精神运动发育延迟,肌张力减退,肝脏和心脏受累。临床外显子组测序显示双等位基因化合物杂合性变异,先前报道的变体c.434G>A(p。G145N,父系)和一个新的变体c.314T>A(p。V105N,母亲)。文献综述发现,在严重的表型中,临床表现的发生率明显高于轻度表型,包括先天性肾病综合征,丙种球蛋白血症,和严重的积水。纯合c.773C>T是与严重表型相关的强致病性变体。当c.773C>T为杂合时,具有另一种变体导致强保守区域内氨基酸取代的患者(c.866A>T,c.1025A>C,c.1182C>G)可能比不太保守区域内的那些引起更严重的表型(c.434G>A,c.450C>G,c.765G>A,c.1287T>A)。c.1129A>G,c.1076C>T,和c.1287T>A更可能与轻度表型相关。疾病表型的评估需要基因型和临床表现的组合。
    结论:本文报道的病例增加了ALG1-CDG中鉴定的突变,对该文献的回顾扩展了对该疾病的表型和基因型谱的研究。
    The congenital disorder of glycosylation associated with ALG1 (ALG1-CDG) is a rare autosomal recessive disease. Due to the deficiency of β1,4 mannosyltransferase caused by pathogenic variants in ALG1 gene, the assembly and processing of glycans in the protein glycosylation pathway are impaired, resulting in a broad clinical spectrum with multi-organ involvement. To raise awareness of clinicians for its manifestations and genotype, we here reported a new patient with a novel variant in ALG1 gene and reviewed the literature to study the genotype-phenotype correlation.
    Clinical characteristics were collected, and clinical exome sequencing was used to identify the causative variants. MutationTaster, PyMol, and FoldX were used to predict the pathogenicity, changes in 3D model molecular structure of protein, and changes of free energy caused by novel variants.
    The proband was a 13-month-old Chinese Han male characterized by epileptic seizures, psychomotor development delay, muscular hypotonia, liver and cardiac involvement. Clinical exome sequencing revealed the biallelic compound heterozygosity variants, a previously reported variant c.434G>A (p.G145N, paternal) and a novel variant c.314T>A (p.V105N, maternal). The literature review found that in severe phenotypes, the incidences of clinical manifestations were significantly higher than that in mild phenotypes, including congenital nephrotic syndrome, agammaglobulinemia, and severe hydrops. Homozygous c.773C>T was a strongly pathogenic variant associated with a severe phenotype. When heterozygous for c.773C>T, patients with another variant leading to substitution in amino acids within the strongly conserved regions (c.866A>T, c.1025A>C, c.1182C>G) may cause a more severe phenotype than those within less-conserved regions (c.434G>A, c.450C>G, c.765G>A, c.1287T>A). c.1129A>G, c.1076C>T, and c.1287T>A were more likely to be associated with a mild phenotype. The assessment of disease phenotypes requires a combination of genotype and clinical manifestations.
    The case reported herein adds to the mutations identified in ALG1-CDG and a review of this literature expands the study of the phenotypic and genotypic spectrum of this disorder.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    目的:天冬酰胺依赖性糖基化11-先天性糖基化障碍(ALG11-CDG)是一种罕见的常染色体隐性N-糖基化缺陷,多系统受累,特别是神经系统症状,如癫痫和神经运动发育迟缓。
    方法:一名31个月大的男性患者入住我们中心,主诉轴性张力减退,耐药性肌阵挛性癫痫发作,小头畸形和耳聋。脑电图(EEG)显示出爆发抑制模式,没有心律失常。基础代谢研究不显著。进行性脑萎缩,在我们的随访过程中拍摄的脑MRI图像中,骨髓溶解不足和call体发育不全是显着特征。通过全外显子组测序(WES)分析发现ALG11基因的复合杂合突变。确定c.476T>C突变是一种新的突变。通过毛细管区带电泳检查碳水化合物缺乏的转铁蛋白(CDT)检测到CDG1型模式。
    结论:在可能存在先天性糖基化缺陷的患者中,建议进行CDT分析等筛查试验.为了在这个罕见的疾病组中发现新的突变,应进行扩展遗传分析。
    OBJECTIVE: Asparagine-dependent glycosylation 11-congenital disorders of glycosylation (ALG11-CDG) is a rare autosomal recessive N-glycosylation defect with multisystem involvement particularly neurological symptoms such as epilepsy and neuromotor developmental delay.
    METHODS: A 31-month-old male patient admitted to our center with complaints of axial hypotonia, drug-resistant myoclonic seizures, microcephaly and deafness. The electroencephalography (EEG) showed a burst-suppression pattern without hypsarrhythmia. Basal metabolic investigations were unremarkable. Progressive cerebral atrophy, hypomyelination and corpus callosum hypoplasia were striking features in brain MRI images taken during our follow-up. Compound heterozygous mutations of the ALG11 gene were found by whole exome sequencing (WES) analysis. It was determined that the c.476T>C mutation is a novel mutation. CDG type 1 pattern was detected with the examination of carbohydrate-deficient transferrin (CDT) by capillary zone electrophoresis.
    CONCLUSIONS: In patients with a possible congenital defect of glycosylation, a screening test such as CDT analysis is suggested. To discover novel mutations in this rare disease group, expanded genetic analysis should be performed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号