Congenital disorders of glycosylation

先天性糖基化障碍
  • 文章类型: Journal Article
    N-糖基化在生肌过程中的作用仍然知之甚少。这里,我们评估了衣霉素(TUN)或磷酸核苷酸变位酶2(PMM2)基因敲低对N-糖基化抑制的影响,它编码一种催化N-糖基化途径早期步骤所必需的酶,关于C2C12成肌细胞分化。TUN慢性治疗对WT和MLC/mIgf-1转基因小鼠胫骨前肌(TA)和趾长伸肌(EDL)的影响,过度表达肌肉Igf-1EamRNA亚型,也被调查了。TUN处理和PMM2敲低的C2C12细胞显示ConA减少,PHA-L,与对照组相比,AAL凝集素结合和ER应激相关基因表达(Chop和Hspa5mRNA和s/uXbp1比率)增加。肌源性标记(MyoD,Myogenin,和Mrf4mRNA和MF20蛋白)和肌管形成在TUN处理和PMM2敲低C2C12细胞中均减少。WT和MLC/mIgf-1小鼠的体重和TA体重未通过TUN处理进行修改,而WT(ConA和AAL)和MLC/mIgf-1(ConA)小鼠的TA肌肉中的凝集素结合略有降低。TUN处理后,WT和MLC/mIgf-1小鼠的TA肌肉中ER应激相关基因表达没有变化。TUN治疗降低了肌原蛋白mRNA和增加了atrogen-1mRNA,特别是在WT小鼠的TA肌肉中。最后,由于TA和EDL肌肉中的N-糖基化抑制,IGF-1产生和IGF1R信号通路激活降低.在TUN处理的C2C12成肌细胞中发现IGF1R表达降低,这与较低的IGF-1诱导的IGF1R相关,AKT,与CTR细胞相比,ERK1/2磷酸化。慢性TUN攻击模型可以帮助阐明与异常N-糖基化相关的疾病的分子机制。如先天性糖基化障碍(CDG),影响肌肉和其他组织功能。
    The role of N-glycosylation in the myogenic process remains poorly understood. Here, we evaluated the impact of N-glycosylation inhibition by Tunicamycin (TUN) or by phosphomannomutase 2 (PMM2) gene knockdown, which encodes an enzyme essential for catalyzing an early step of the N-glycosylation pathway, on C2C12 myoblast differentiation. The effect of chronic treatment with TUN on tibialis anterior (TA) and extensor digitorum longus (EDL) muscles of WT and MLC/mIgf-1 transgenic mice, which overexpress muscle Igf-1Ea mRNA isoform, was also investigated. TUN-treated and PMM2 knockdown C2C12 cells showed reduced ConA, PHA-L, and AAL lectin binding and increased ER-stress-related gene expression (Chop and Hspa5 mRNAs and s/uXbp1 ratio) compared to controls. Myogenic markers (MyoD, myogenin, and Mrf4 mRNAs and MF20 protein) and myotube formation were reduced in both TUN-treated and PMM2 knockdown C2C12 cells. Body and TA weight of WT and MLC/mIgf-1 mice were not modified by TUN treatment, while lectin binding slightly decreased in the TA muscle of WT (ConA and AAL) and MLC/mIgf-1 (ConA) mice. The ER-stress-related gene expression did not change in the TA muscle of WT and MLC/mIgf-1 mice after TUN treatment. TUN treatment decreased myogenin mRNA and increased atrogen-1 mRNA, particularly in the TA muscle of WT mice. Finally, the IGF-1 production and IGF1R signaling pathways activation were reduced due to N-glycosylation inhibition in TA and EDL muscles. Decreased IGF1R expression was found in TUN-treated C2C12 myoblasts which was associated with lower IGF-1-induced IGF1R, AKT, and ERK1/2 phosphorylation compared to CTR cells. Chronic TUN-challenge models can help to elucidate the molecular mechanisms through which diseases associated with aberrant N-glycosylation, such as Congenital Disorders of Glycosylation (CDG), affect muscle and other tissue functions.
    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
    目的:我们的报告描述了临床,遗传,在研究的第5年,在先天性糖基化障碍(FCDGC)自然病史队列中纳入分子证实的先天性糖基化障碍(CDG)参与者的生化特征。
    方法:我们将已知或疑似CDG的个体纳入FCDGC自然史研究,对CDG的所有遗传原因进行多中心前瞻性和回顾性自然史研究。我们对2019年10月至2023年11月同意参加FCDGC自然历史研究(5U54NS115198)的确诊CDG参与者的基线研究访问数据进行了横断面分析。
    结果:133名受试者同意FCDGC自然史研究。其中,280个独特的个体具有与CDG的诊断一致的可用遗传数据。这280人在2019年10月8日至2023年11月29日期间被纳入研究。一百四十一名(50.4%)是女性,男性139人(49.6%)。入学时的平均年龄为10.1岁和6.5岁,分别,范围为0.22至71.4年。该队列包括患有N-连接蛋白糖基化疾病的个体(57%),糖基磷脂酰肌醇锚定障碍(GPI锚定)(15%),高尔基稳态疾病,贩运和运输(12%),dolichol代谢紊乱(5%),多种途径障碍(6%),其他(5%)。导致诊断的最常见症状是发育迟缓/残疾(77%)。其次是低张力(56%)和进食困难(42%)。首次相关症状和诊断之间的平均和中位时间为2.7年和0.8年,分别。我们队列中百分之百的人在基线就诊时存在发育差异/残疾,其次是97%的神经系统受累,91%有胃肠道(GI)/肝脏受累,88%有肌肉骨骼受累。在NijmegenProgressionCDG评定量表(NPCRS)上对个体疾病的严重程度进行了评分,其中27%的评分被归类为轻度,44%中度,29%严重。在有N-连接蛋白糖基化缺陷的个体中,83%的数据显示碳水化合物缺乏转铁蛋白(CDT)分析为1型模式,包括82/84例PMM2-CDG患者,6%的2型模式,类型1和类型2模式均为1%,正常或非特异性模式为10%。百分之百的高尔基稳态和贩运缺陷的个体在CDT分析中显示出2型模式,而高尔基运输缺陷在73%的情况下表现出II型模式,7%的1型模式,20%有正常或非特异性模式。使用ACMG标准将记录的大多数变体分类为致病性或可能致病性。对于大多数变体,预测的分子结果是错误的,其次是无义和剪接位点,大多数诊断是以常染色体隐性遗传模式遗传的,但包括所有主要核遗传的疾病。
    结论:FCDGC自然历史研究是建立未来研究的重要资源,改善临床护理,并为临床试验做好准备。这是FCDGC自然史研究的CDG参与者的第一个概述。
    OBJECTIVE: Our report describes clinical, genetic, and biochemical features of participants with a molecularly confirmed congenital disorder of glycosylation (CDG) enrolled in the Frontiers in Congenital Disorders of Glycosylation (FCDGC) Natural History cohort at year 5 of the study.
    METHODS: We enrolled individuals with a known or suspected CDG into the FCDGC Natural History Study, a multicenter prospective and retrospective natural history study of all genetic causes of CDG. We conducted a cross-sectional analysis of baseline study visit data from participants with confirmed CDG who were consented into the FCDGC Natural History Study (5U54NS115198) from October 2019 to November 2023.
    RESULTS: Three hundred thirty-three subjects consented to the FCDGC Natural History Study. Of these, 280 unique individuals had genetic data available that was consistent with a diagnosis of CDG. These 280 individuals were enrolled into the study between October 8, 2019 and November 29, 2023. One hundred forty-one (50.4%) were female, and 139 (49.6%) were male. Mean and median age at enrollment was 10.1 and 6.5 years, respectively, with a range of 0.22 to 71.4 years. The cohort encompassed individuals with disorders of N-linked protein glycosylation (57%), glycosylphosphatidylinositol anchor disorder (GPI anchor) (15%), disorders of Golgi homeostasis, trafficking and transport (12%), dolichol metabolism disorders (5%), disorders of multiple pathways (6%), and other (5%). The most frequent presenting symptom(s) leading to diagnosis were developmental delay/disability (77%), followed by hypotonia (56%) and feeding difficulties (42%). Mean and median time between first related symptom and diagnosis was 2.7 and 0.8 years, respectively. One hundred percent of individuals in our cohort had developmental differences/disabilities at the time of their baseline visit, followed by 97% with neurologic involvement, 91% with gastrointestinal (GI)/liver involvement, and 88% with musculoskeletal involvement. Severity of disease in individuals was scored on the Nijmegen Progression CDG Rating Scale (NPCRS) with 27% of scores categorized as mild, 44% moderate, and 29% severe. Of the individuals with N-linked protein glycosylation defects, 83% of those with data showed a type 1 pattern on carbohydrate deficient transferrin (CDT) analysis including 82/84 individuals with PMM2-CDG, 6% a type 2 pattern, 1% both type 1 and type 2 pattern and 10% a normal or nonspecific pattern. One hundred percent of individuals with Golgi homeostasis and trafficking defects with data showed a type 2 pattern on CDT analysis, while Golgi transport defect showed a type II pattern 73% of the time, a type 1 pattern for 7%, and 20% had a normal or nonspecific pattern. Most of the variants documented were classified as pathogenic or likely pathogenic using ACMG criteria. For the majority of the variants, the predicted molecular consequence was missense followed by nonsense and splice site, and the majority of the diagnoses are inherited in an autosomal recessive pattern but with disorders of all major nuclear inheritance included.
    CONCLUSIONS: The FCDGC Natural History Study serves as an important resource to build future research studies, improve clinical care, and prepare for clinical trial readiness. Herein is the first overview of CDG participants of the FCDGC Natural History Study.
    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:先天性糖基化疾病(CDG)是一组不断扩展的单基因疾病,会破坏糖蛋白和糖脂的生物合成,导致多系统表现。根据糖基化过程的哪个部分受损,将这些病症分为不同的组。CDG的心脏表现可以显着不同,不仅在不同类型之间,而且在具有相同CDG遗传原因的个体之间。心肌病是CDG的重要表型。CDG患者心肌病的临床表现和进展尚未得到很好的表征。这项研究旨在描述各种CDG遗传原因中心肌病的常见模式,并为该患者人群提出基线筛查和随访评估。
    方法:根据对其病历的回顾性回顾,确定了CDG分子确认的患者,这些患者被纳入了先天性糖基化障碍联盟(FCDGC)自然史研究的前瞻性或纪念性研究。所有患者均由FCDGC成员的临床遗传学家在各自的学术中心进行评估。患者接受了心肌病筛查,并回顾性收集了详细的数据。我们分析了他们的临床和分子病史,心脏受累的影像学特征,心肌病的类型,最初出现心肌病的年龄,额外的心脏特征,给予的治疗,和他们的临床结果。
    结果:在截至2023年6月参与FCDGC自然史研究的305名分子确认CDG患者中,有17名个体,九位女性八位男性,同时诊断为心肌病。这些患者大多数被诊断为PMM2-CDG(n=10)。然而,在其他诊断中也观察到心肌病,包括PGM1-CDG(n=3),ALG3-CDG(n=1),DPM1-CDG(n=1),DPAGT1-CDG(n=1),和SSR4-CDG(n=1)。所有PMM2-CDG患者均报告患有肥厚型心肌病。在三名患者中观察到扩张型心肌病,2例PGM1-CDG和1例ALG3-CDG;2例诊断为左心室致密化不全心肌病,1例PGM1-CDG,1例DPAGT1-CDG;2例,一个带有DPM1-CDG,一个带有SSR4-CDG,被诊断为非缺血性心肌病。估计诊断为心肌病的中位年龄为5个月(范围:产前-27岁)。在3例PMM2-CDG患者中观察到心脏改善。五名患者表现出心肌病的进行性过程,而8个人的情况保持不变。6例患者出现心包积液,三名患者表现出心脏压塞。一名SSR4-CDG患者最近被诊断为心肌病;因此,疾病的进展尚未确定。1例PGM1-CDG患者行心脏移植。七名病人死亡,包括五个PMM2-CDG,一个带有DPAGT1-CDG,和一个ALG3-CDG。两名患者死于心包积液引起的心包填塞;其余患者,心肌病不一定是死亡的主要原因.
    结论:在这项回顾性研究中,在~6%的CDG患者中发现了心肌病。值得注意的是,大多数,包括所有带有PMM2-CDG的,表现为肥厚型心肌病。有些病例没有进展,然而,通常观察到心包积液,尤其是在PMM2-CDG患者中,偶尔会升级到危及生命的心包填塞。建议临床医生管理CDG患者,特别是那些带有PMM2-CDG和PGM1-CDG的,警惕心肌病风险和潜在危及生命的心包积液风险。心脏监测,包括超声心动图和心电图,应该在诊断时进行,在最初的五年中,每年,随后每2-3年检查一次,如果直到成年后才出现担忧。随后,建议每五年进行一次心脏常规检查。此外,诊断为心肌病的患者应接受持续的心脏护理,以确保对其病情进行有效的管理和监测。需要进行前瞻性研究以确定CDG中心肌病的真实患病率。
    BACKGROUND: Congenital disorders of glycosylation (CDG) are a continuously expanding group of monogenic disorders that disrupt glycoprotein and glycolipid biosynthesis, leading to multi-systemic manifestations. These disorders are categorized into various groups depending on which part of the glycosylation process is impaired. The cardiac manifestations in CDG can significantly differ, not only across different types but also among individuals with the same genetic cause of CDG. Cardiomyopathy is an important phenotype in CDG. The clinical manifestations and progression of cardiomyopathy in CDG patients have not been well characterized. This study aims to delineate common patterns of cardiomyopathy across a range of genetic causes of CDG and to propose baseline screening and follow-up evaluation for this patient population.
    METHODS: Patients with molecular confirmation of CDG who were enrolled in the prospective or memorial arms of the Frontiers in Congenital Disorders of Glycosylation Consortium (FCDGC) natural history study were ascertained for the presence of cardiomyopathy based on a retrospective review of their medical records. All patients were evaluated by clinical geneticists who are members of FCDGC at their respective academic centers. Patients were screened for cardiomyopathy, and detailed data were retrospectively collected. We analyzed their clinical and molecular history, imaging characteristics of cardiac involvement, type of cardiomyopathy, age at initial presentation of cardiomyopathy, additional cardiac features, the treatments administered, and their clinical outcomes.
    RESULTS: Of the 305 patients with molecularly confirmed CDG participating in the FCDGC natural history study as of June 2023, 17 individuals, nine females and eight males, were identified with concurrent diagnoses of cardiomyopathy. Most of these patients were diagnosed with PMM2-CDG (n = 10). However, cardiomyopathy was also observed in other diagnoses, including PGM1-CDG (n = 3), ALG3-CDG (n = 1), DPM1-CDG (n = 1), DPAGT1-CDG (n = 1), and SSR4-CDG (n = 1). All PMM2-CDG patients were reported to have hypertrophic cardiomyopathy. Dilated cardiomyopathy was observed in three patients, two with PGM1-CDG and one with ALG3-CDG; left ventricular non-compaction cardiomyopathy was diagnosed in two patients, one with PGM1-CDG and one with DPAGT1-CDG; two patients, one with DPM1-CDG and one with SSR4-CDG, were diagnosed with non-ischemic cardiomyopathy. The estimated median age of diagnosis for cardiomyopathy was 5 months (range: prenatal-27 years). Cardiac improvement was observed in three patients with PMM2-CDG. Five patients showed a progressive course of cardiomyopathy, while the condition remained unchanged in eight individuals. Six patients demonstrated pericardial effusion, with three patients exhibiting cardiac tamponade. One patient with SSR4-CDG has been recently diagnosed with cardiomyopathy; thus, the progression of the disease is yet to be determined. One patient with PGM1-CDG underwent cardiac transplantation. Seven patients were deceased, including five with PMM2-CDG, one with DPAGT1-CDG, and one with ALG3-CDG. Two patients died of cardiac tamponade from pericardial effusion; for the remaining patients, cardiomyopathy was not necessarily the primary cause of death.
    CONCLUSIONS: In this retrospective study, cardiomyopathy was identified in ∼6% of patients with CDG. Notably, the majority, including all those with PMM2-CDG, exhibited hypertrophic cardiomyopathy. Some cases did not show progression, yet pericardial effusions were commonly observed, especially in PMM2-CDG patients, occasionally escalating to life-threatening cardiac tamponade. It is recommended that clinicians managing CDG patients, particularly those with PMM2-CDG and PGM1-CDG, be vigilant of the cardiomyopathy risk and risk for potentially life-threatening pericardial effusions. Cardiac surveillance, including an echocardiogram and EKG, should be conducted at the time of diagnosis, annually throughout the first 5 years, followed by check-ups every 2-3 years if no concerns arise until adulthood. Subsequently, routine cardiac examinations every five years are advisable. Additionally, patients with diagnosed cardiomyopathy should receive ongoing cardiac care to ensure the effective management and monitoring of their condition. A prospective study will be required to determine the true prevalence of cardiomyopathy in 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: Journal Article
    随着全外显子组和基因组测序(WES和WGS)在临床实践中的整合,孟德尔疾病的诊断显着进步。然而,WES在变异解释和未发现变异方面的挑战仍然使相当大比例的患者未得到诊断.在这种情况下,整合RNA测序(RNA-seq)改善了诊断工作流程,特别是对于WES不确定的情况。此外,功能研究往往是必要的,以阐明优先变异对基因表达和蛋白质功能的影响。我们的研究集中在三个无关的男性患者(P1-P3)与ATP6AP1-CDG(先天性糖基化障碍),表现为智力残疾和不同程度的肝病,糖基化缺陷,并通过WES进行初步不确定的诊断。随后的RNA-seq在确定P1和P2的潜在遗传原因,检测ATP6AP1表达不足和异常剪接方面至关重要。成纤维细胞中的分子研究证实了这些发现,并分别在P1和P2中鉴定了罕见的内含子变体c.289-233C>T和c.289-289G>A。Trio-WGS还揭示了P3中的变异c.289-289G>A,这在两名患者中是从头变化。在HAP1细胞中表达突变等位基因的功能测定通过再现在患者中观察到的剪接改变证明了这些变体的致病影响。我们的研究强调了RNA-seq和WGS在提高CDG等遗传疾病的诊断率方面的作用。通过确定该X连锁基因中的前两个深层内含子变体,为ATP6AP1-CDG分子碱基提供新的见解。此外,我们的研究强调了整合RNA-seq和WGS的必要性,其次是功能验证,在常规诊断中,对分子病因不明的患者进行全面评估。
    The diagnosis of Mendelian disorders has notably advanced with integration of whole exome and genome sequencing (WES and WGS) in clinical practice. However, challenges in variant interpretation and uncovered variants by WES still leave a substantial percentage of patients undiagnosed. In this context, integrating RNA sequencing (RNA-seq) improves diagnostic workflows, particularly for WES inconclusive cases. Additionally, functional studies are often necessary to elucidate the impact of prioritized variants on gene expression and protein function. Our study focused on three unrelated male patients (P1-P3) with ATP6AP1-CDG (congenital disorder of glycosylation), presenting with intellectual disability and varying degrees of hepatopathy, glycosylation defects, and an initially inconclusive diagnosis through WES. Subsequent RNA-seq was pivotal in identifying the underlying genetic causes in P1 and P2, detecting ATP6AP1 underexpression and aberrant splicing. Molecular studies in fibroblasts confirmed these findings and identified the rare intronic variants c.289-233C > T and c.289-289G > A in P1 and P2, respectively. Trio-WGS also revealed the variant c.289-289G > A in P3, which was a de novo change in both patients. Functional assays expressing the mutant alleles in HAP1 cells demonstrated the pathogenic impact of these variants by reproducing the splicing alterations observed in patients. Our study underscores the role of RNA-seq and WGS in enhancing diagnostic rates for genetic diseases such as CDG, providing new insights into ATP6AP1-CDG molecular bases by identifying the first two deep intronic variants in this X-linked gene. Additionally, our study highlights the need to integrate RNA-seq and WGS, followed by functional validation, in routine diagnostics for a comprehensive evaluation of patients with an unidentified molecular etiology.
    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
    N-乙酰-d-神经氨酸合酶-糖基化先天性疾病(NANS-CDG)是N-乙酰-神经氨酸生物合成途径中罕见的常染色体隐性遗传缺陷。在这里,我们报告首例韩国NANS-CDG患者。一个10岁的男孩被转诊到我们的诊所,因为偶然的影像学发现表明脊椎表骨干发育不良。病人有小头畸形,透明隔腔,出生时的脑室肿大,十年后,身材矮小.他有特发性慢性免疫性血小板减少症病史,中枢肾上腺功能不全,从婴儿期开始甲状腺功能减退.第一次无缘无故的癫痫发作发生在2岁时,他随后因呼吸道感染和顽固性癫痫发作而经常入院。外显子组测序鉴定了未报道的NANS基因的双等位基因变体。NANS-CDG的临床和遗传证实突出了其不断扩大的表型和基因型多样性。
    N-acetyl-d-neuraminic acid synthase-congenital disorder of glycosylation (NANS-CDG) is a rare autosomal recessive defect in the N-acetyl-neuraminic acid biosynthesis pathway. Herein, we report the first Korean NANS-CDG patient. A 10-year-old boy was referred to our clinic because of incidental radiographic findings indicating spondyloepimetaphyseal dysplasia. The patient had microcephaly, cavum septum pellucidum, and ventriculomegaly at birth, and at 10 years, a very short stature. He had a history of idiopathic chronic immune thrombocytopenia, central adrenal insufficiency, and hypothyroidism since infancy. The first unprovoked seizure occurred at the age of 2 years, and he was subsequently admitted to the hospital frequently because of respiratory infections and intractable seizures. Exome sequencing identified unreported biallelic variants of the NANS gene. Clinical and genetic confirmation of NANS-CDG highlights its expanding phenotypic and genotypic diversity.
    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
    Dolichol是N-糖基化的关键脂质,可作为活化糖和新生寡糖的载体。通常认为它是通过酶SRD5A3从聚丙炔醇直接产生的。相反,我们发现dolichol合成需要三步绕道,涉及额外的代谢物,其中SRD5A3仅催化第二个反应。第一步和第三步由DHRSX执行,其基因位于X和Y染色体的伪常染色体区域。因此,我们报道了一种在DHRSX错义变异(DHRSX-CDG)患者中表现为先天性糖基化异常的常染色体隐性遗传病.值得注意的是,DHRSX具有独特的双重底物和辅因子特异性,允许它在两个非连续步骤中充当NAD依赖性脱氢酶和NADPH依赖性还原酶。因此,我们的工作揭示了dolichol生物合成的最终步骤中意想不到的复杂性。此外,我们提供了有关dolichol代谢缺陷导致疾病的机制的见解。
    Dolichol is a lipid critical for N-glycosylation as a carrier for activated sugars and nascent oligosaccharides. It is commonly thought to be directly produced from polyprenol by the enzyme SRD5A3. Instead, we found that dolichol synthesis requires a three-step detour involving additional metabolites, where SRD5A3 catalyzes only the second reaction. The first and third steps are performed by DHRSX, whose gene resides on the pseudoautosomal regions of the X and Y chromosomes. Accordingly, we report a pseudoautosomal-recessive disease presenting as a congenital disorder of glycosylation in patients with missense variants in DHRSX (DHRSX-CDG). Of note, DHRSX has a unique dual substrate and cofactor specificity, allowing it to act as a NAD+-dependent dehydrogenase and as a NADPH-dependent reductase in two non-consecutive steps. Thus, our work reveals unexpected complexity in the terminal steps of dolichol biosynthesis. Furthermore, we provide insights into the mechanism by which dolichol metabolism defects contribute to disease.
    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
    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
    Mabry等人的病例报告。(1970)一个有四个孩子的家庭,组织非特异性碱性磷酸酶升高,癫痫发作和严重的发育障碍,成为具有被称为Mabry综合征的特征的儿童表型的基础。除了改善为患者和家庭提供的服务外,然而,诊断和治疗,和许多其他发育障碍,在大规模平行测序出现之前没有显著变化。随着越来越多具有Mabry综合征特征的患者被发现,外显子组和基因组测序用于鉴定糖磷脂酰肌醇(GPI)生物合成障碍(GPIBDs)为一组先天性糖基化障碍(CDG).磷脂酰肌醇聚糖(PIG)生物合成的双等位基因变体,在Mabry综合征中鉴定出的V型(PIGV)基因成为表型系列中第一个的证据,该系列按发现顺序编号为HPMRS1-6。HPMRS1[MIM:239300]是由双等位基因PIGV变体的遗传产生的表型。同样,HPMRS2(MIM614749),HPMRS5(MIM616025)和HPMRS6(MIM616809)是由PIGO的破坏引起的,PIGW和PIGY基因在内质网中表达。相比之下,HPMRS3(MIM614207)和HPMRS4(MIM615716)由与蛋白质PGAP2(HPMRS3)和PGAP3(HPMRS4)的后附着的破坏产生。GPI生物合成障碍(GPIBDs)目前编号为GPIBD1-21。和Mabry医生一起工作,在2020年,我们能够使用改进的实验室诊断来完成他最初在1970年描述的患者的分子诊断.我们在首次报道的HPMRS患者中鉴定了PGAP2基因的双等位基因变体。我们在吡哆醇治疗癫痫发作的效用以及HPMRS3患者中推定的糖脂储存的证据的背景下,讨论了Mabry综合征指数患者的寿命。从实验室创新的角度来看,这些创新使Mabry博士的患者能够识别HPMRS表型,显然,有必要进行治疗创新,使受发育障碍影响的患者和家庭受益。
    The case report by Mabry et al. (1970) of a family with four children with elevated tissue non-specific alkaline phosphatase, seizures and profound developmental disability, became the basis for phenotyping children with the features that became known as Mabry syndrome. Aside from improvements in the services available to patients and families, however, the diagnosis and treatment of this, and many other developmental disabilities, did not change significantly until the advent of massively parallel sequencing. As more patients with features of the Mabry syndrome were identified, exome and genome sequencing were used to identify the glycophosphatidylinositol (GPI) biosynthesis disorders (GPIBDs) as a group of congenital disorders of glycosylation (CDG). Biallelic variants of the phosphatidylinositol glycan (PIG) biosynthesis, type V (PIGV) gene identified in Mabry syndrome became evidence of the first in a phenotypic series that is numbered HPMRS1-6 in the order of discovery. HPMRS1 [MIM: 239300] is the phenotype resulting from inheritance of biallelic PIGV variants. Similarly, HPMRS2 (MIM 614749), HPMRS5 (MIM 616025) and HPMRS6 (MIM 616809) result from disruption of the PIGO, PIGW and PIGY genes expressed in the endoplasmic reticulum. By contrast, HPMRS3 (MIM 614207) and HPMRS4 (MIM 615716) result from disruption of post attachment to proteins PGAP2 (HPMRS3) and PGAP3 (HPMRS4). The GPI biosynthesis disorders (GPIBDs) are currently numbered GPIBD1-21. Working with Dr. Mabry, in 2020, we were able to use improved laboratory diagnostics to complete the molecular diagnosis of patients he had originally described in 1970. We identified biallelic variants of the PGAP2 gene in the first reported HPMRS patients. We discuss the longevity of the Mabry syndrome index patients in the context of the utility of pyridoxine treatment of seizures and evidence for putative glycolipid storage in patients with HPMRS3. From the perspective of the laboratory innovations made that enabled the identification of the HPMRS phenotype in Dr. Mabry\'s patients, the need for treatment innovations that will benefit patients and families affected by developmental disabilities is clear.
    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
    先天性糖基化疾病(CDG)是增长最快的先天性代谢错误之一,包括超过160描述的疾病到今天。CDG的特征是糖基化过程功能失调,分子缺陷位于胞质溶胶中,内质网,或者高尔基体.根据CDG,N-糖基化,可以影响0-糖基化和/或糖胺聚糖合成。各种蛋白质,脂质,糖基磷脂酰肌醇锚带有聚糖链,对它们的折叠有潜在的影响,瞄准,分泌,稳定性,因此,功能。因此,糖基化缺陷可能会产生多种严重的临床后果。CDG患者通常存在非特异性,多系统综合征,包括神经系统受累,生长延迟,肝病和凝血病。由于CDG是罕见疾病,通常缺乏独特的临床症状,生化和基因检测具有特别重要和互补的诊断作用。这里,在简要介绍了糖基化和CDG之后,我们回顾了CDG生物标志物和相关分析技术的历史和最新发现,特别强调那些在专业临床化学实验室中具有相关用途的人。我们为读者提供了见解和方法,可以帮助他们正确地协助临床医生浏览糖基化障碍的迷宫。
    Congenital disorders of glycosylation (CDG) are one of the fastest growing groups of inborn errors of metabolism, comprising over 160 described diseases to this day. CDG are characterized by a dysfunctional glycosylation process, with molecular defects localized in the cytosol, the endoplasmic reticulum, or the Golgi apparatus. Depending on the CDG, N-glycosylation, O-glycosylation and/or glycosaminoglycan synthesis can be affected. Various proteins, lipids, and glycosylphosphatidylinositol anchors bear glycan chains, with potential impacts on their folding, targeting, secretion, stability, and thus, functionality. Therefore, glycosylation defects can have diverse and serious clinical consequences. CDG patients often present with a non-specific, multisystemic syndrome including neurological involvement, growth delay, hepatopathy and coagulopathy. As CDG are rare diseases, and typically lack distinctive clinical signs, biochemical and genetic testing bear particularly important and complementary diagnostic roles. Here, after a brief introduction on glycosylation and CDG, we review historical and recent findings on CDG biomarkers and associated analytical techniques, with a particular emphasis on those with relevant use in the specialized clinical chemistry laboratory. We provide the reader with insights and methods which may help them properly assist the clinician in navigating the maze of glycosylation disorders.
    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
    O-GlcNAc转移酶基因(OGT)中的致病变体与先天性糖基化障碍(OGT-CDG)有关,表现为可能是神经外胚层起源的智力残疾。为了检验病理与早期胚胎发生过程中的分化缺陷有关的假设,我们开发了一种OGT-CDG诱导的多能干细胞系,以及通过CRISPR/Cas9基因编辑产生的等基因对照.尽管OGT-CDG变体导致OGT和O-GlcNAcase蛋白水平显著降低,分化潜能或干性没有变化.然而,分化为外胚层导致O-GlcNAc稳态的显着差异。进一步分化为神经元干细胞显示患者和对照组之间的形态差异,伴随着O-GlcNAc途径的破坏。这表明O-GlcNAcylation在早期神经外胚层结构中的关键作用,在干细胞分化的早期阶段具有强大的补偿机制。
    Pathogenic variants in the O-GlcNAc transferase gene (OGT) have been associated with a congenital disorder of glycosylation (OGT-CDG), presenting with intellectual disability which may be of neuroectodermal origin. To test the hypothesis that pathology is linked to defects in differentiation during early embryogenesis, we developed an OGT-CDG induced pluripotent stem cell line together with isogenic control generated by CRISPR/Cas9 gene-editing. Although the OGT-CDG variant leads to a significant decrease in OGT and O-GlcNAcase protein levels, there were no changes in differentiation potential or stemness. However, differentiation into ectoderm resulted in significant differences in O-GlcNAc homeostasis. Further differentiation to neuronal stem cells revealed differences in morphology between patient and control lines, accompanied by disruption of the O-GlcNAc pathway. This suggests a critical role for O-GlcNAcylation in early neuroectoderm architecture, with robust compensatory mechanisms in the earliest stages of stem cell differentiation.
    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