Aicardi-Goutieres Syndrome

  • 文章类型: Journal Article
    TREX1基因突变导致Aicardi-Goutières综合征(AGS)1,与一系列自身免疫和神经退行性表现有关。AGS1,最严重的新生儿AGS类型,以异常的神经系统表现为特征,视觉注意力不集中,肝脾肿大,血小板减少症,皮疹,躁动,和发烧。
    本研究描述了来自伊朗家庭的两个受影响的兄弟姐妹,其表型与宫内感染重叠。他们有几乎相似的演讲,包括发育迟缓,小头畸形,没有固定和跟随癫痫发作和相同模式的脑CT扫描受累。根据临床和临床评估,全外显子组测序用于确定致病变异,随后,进行PCR-Sanger测序以指示家族成员中候选变体的分离模式。
    遗传分析揭示了受影响的家族成员中TREX1基因中的一个新的纯合错义变体(c.461A>C;p.D154A)。其他家族成员的Sanger测序显示了预期的接合性。
    这项研究确定了该家族中TREX1基因的一种新突变,并强调了下一代基于测序的技术在早发性脑病患者中获得明确诊断的效率。
    UNASSIGNED: Mutations in the TREX1 gene cause Aicardi-Goutières syndrome (AGS) 1, associated with a spectrum of autoimmune and neurodegenerative manifestations. AGS 1, the most severe neonatal type of AGS, is characterized by abnormal neurologic findings, visual inattention, hepatosplenomegaly, thrombocytopenia, skin rash, restlessness, and fever.
    UNASSIGNED: The present study described two affected siblings from an Iranian family whose phenotypes overlap with intrauterine infections. They had almost similar presentations, including developmental delay, microcephaly, no fix and follow epileptic seizures and the same pattern of brain CT scan involvements. Following clinical and paraclinical assessments, whole-exome sequencing was employed to determine the disease-causing variant, and subsequently, PCR-Sanger sequencing was performed to indicate the segregation pattern of the candidate variant in family members.
    UNASSIGNED: Genetic analysis revealed a novel homozygous missense variant (c.461A>C; p.D154A) in the TREX1 gene in affected family members. Sanger sequencing of other family members showed the expected zygosities.
    UNASSIGNED: This study identifies a novel mutation in the TREX1 gene in this family and highlights the efficiency of next-generation sequencing-based techniques for obtaining a definite diagnosis in patients with early-onset encephalopathy.
    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
    cGAS-STING途径检测胞质DNA并激活导致I型干扰素(IFN)应答的信号级联。内质网(ER)相关的外切核酸酶TREX1通过从细胞质中消除DNA来抑制cGAS-STING。损害TREX1功能的突变与自身炎症性疾病有关,包括系统性红斑狼疮(SLE)和Aicardi-Goutières综合征(AGS)。尽管在调节cGAS-STING和抑制过度炎症中起关键作用,许多疾病相关TREX1突变的影响,特别是核心催化结构域以外的突变,仍然知之甚少.这里,我们描述了一个隐性AGS连锁TREX1P61Q突变发生在特征不佳的多脯氨酸螺旋(PPII)基序内.为了保持其在催化核心或ER靶向基序之外的位置,既不是P61Q突变,也没有聚集脯氨酸到丙氨酸PPII突变,破坏TREX1核酸外切酶活性,亚细胞定位,或过表达系统中的cGAS-STING调节。将靶向突变引入内源性TREX1基因座表明PPII突变使蛋白质不稳定。导致外切核酸酶活性受损和不受限制的cGAS-STING激活。总的来说,这些结果表明,TREX1PPII突变,包括P61Q,通过TREX1失稳损害适当的免疫调节并导致自身免疫性疾病。
    The cGAS-STING pathway detects cytosolic DNA and activates a signaling cascade that results in a type I interferon (IFN) response. The endoplasmic reticulum (ER)-associated exonuclease TREX1 suppresses cGAS-STING by eliminating DNA from the cytosol. Mutations that compromise TREX1 function are linked to autoinflammatory disorders, including systemic lupus erythematosus (SLE) and Aicardi-Goutières syndrome (AGS). Despite key roles in regulating cGAS-STING and suppressing excessive inflammation, the impact of many disease-associated TREX1 mutations-particularly those outside of the core catalytic domains-remains poorly understood. Here, we characterize a recessive AGS-linked TREX1 P61Q mutation occurring within the poorly characterized polyproline helix (PPII) motif. In keeping with its position outside of the catalytic core or ER targeting motifs, neither the P61Q mutation, nor aggregate proline-to-alanine PPII mutation, disrupts TREX1 exonuclease activity, subcellular localization, or cGAS-STING regulation in overexpression systems. Introducing targeted mutations into the endogenous TREX1 locus revealed that PPII mutations destabilize the protein, resulting in impaired exonuclease activity and unrestrained cGAS-STING activation. Overall, these results demonstrate that TREX1 PPII mutations, including P61Q, impair proper immune regulation and lead to autoimmune disease through TREX1 destabilization.
    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
    一名6岁的女性,有Aicardi-Goutières综合征(AGS)的病史,出现了色素沉着和色素沉着的黄斑和斑块,与对称色素沉着症(DSH)一致。以前的基因检查证明是从头的,腺苷脱氨酶作用于RNA1(ADAR)基因的杂合突变。虽然先前已经在ADAR基因突变中描述了AGS和DSH的共同出现,我们的病例强调了这些疾病之间的潜在关联,这可能有助于将来早期诊断AGS.
    A 6-year-old female with a history of Aicardi-Goutières syndrome (AGS) presented to dermatology clinic with hypopigmented and hyperpigmented macules and patches consistent with dyschromatosis symmetrica hereditaria (DSH). Previous genetic workup demonstrated a de novo, heterozygous mutation in the adenosine deaminase acting on RNA 1 (ADAR) gene. While the co-occurrence of AGS and DSH has previously been described in mutations of the ADAR gene, our case highlights the potential association between these disorders that may aid in earlier future diagnosis of AGS.
    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Aicardi-Goutieres综合征(AGS)是一种遗传性疾病,具有多种表现,包括神经系统,免疫学,有时其他系统参与各种组合。考虑到AGS的高度遗传和临床多样性以及RNASEH2复合物在生物系统中的重要性,重要的是采取系统的方法来描述错义突变的遗传诊断和影响。方法对具有AGS临床特征的个体进行临床靶向基因测序,然后进行Sanger验证。迄今为止,所有报告的RNASEH2A错义变体的蛋白质建模研究都是使用免费的Web服务器BioGrid进行的,ShinyGO.使用Pymol可视化蛋白质结构。结果和讨论我们在RNASEH2A中鉴定了新的纯合剪接位点供体变体c.549+1G>T。此外,蛋白质相互作用组研究确定了潜在的遗传相互作用者,包括RNASEH2A,RNASEH2B,TYMS,RNASEH2C,RPA1,ORC3,ORC2,CDC6,PCNA,LIG1,PRIM1,RFC2,DUT,GINS1、MCM7、FEN1、MCM4、GINS2、CDK4和MCM5。使用SHINYGO将鉴定的基因定位到特定途径。DNA复制和细胞周期,中心体周期,复制后修复,核酸和代谢过程,细胞对压力的反应,DNA代谢过程,核酸磷酸二酯键水解,RNA磷酸二酯键水解,和DNA生物合成过程被确定为与优先基因的连接途径。结论总之,复杂的基因型和表型相关性,然后将基因与关键生物学途径联系起来,为了解疾病病理和治疗干预计划开辟了新的途径。
    Background Aicardi-Goutieres syndrome (AGS) is a genetic disorder that has variable manifestations including neurological, immunological, and sometimes other system involvement in various combinations. Considering the high genetic and clinical diversity of AGS and the importance of RNASEH2 complex in the biological system, it is important to take a systematic approach to delineate the genetic diagnosis and impact of missense mutations. Methods Clinical targeted gene sequencing followed by Sanger validation was performed in an individual with the clinical features of AGS. Protein modeling studies of all the reported RNASEH2A missense variants till date were performed using freely available web servers BioGrid, ShinyGO. Protein structures were visualized using Pymol. Results and discussion We identified a novel homozygous splice site donor variant c.549+1G>T in RNASEH2A. Furthermore protein-interactome studies identifiedpotential genetic interactors that include RNASEH2A, RNASEH2B, TYMS, RNASEH2C, RPA1, ORC3, ORC2, CDC6, PCNA, LIG1, PRIM1, RFC2, DUT, GINS1, MCM7, FEN1, MCM4, GINS2, CDK4, and MCM5. Identified genes were mapped to specific pathways using SHINY GO. DNA replication and cell cycle, centrosome cycle, post-replication repair, nucleic acid and metabolic process, cellular response to stress, DNA metabolic process, nucleic acid phosphodiester bond hydrolysis, RNA phosphodiester bond hydrolysis, and DNA biosynthetic process were identified as the linked pathways with the prioritized genes. Conclusion In conclusion, a sophisticated genotype and phenotype correlation followed by linking the genes to the key biological pathways opens new avenues to understand disease pathology and plan for therapeutic interventions.
    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
    Aicardi-Goutieres综合征(AGS)是一种属于I型干扰素病组的炎症性疾病。AGS的临床诊断比较困难,这可能导致高死亡率。总的来说,中国缺乏AGS的大样本研究数据。我们旨在总结中国AGS患者的临床特征,为临床诊断提供线索。
    收集中国AGS患者的遗传和临床特征。实时聚合酶链反应用于检测干扰素刺激基因(ISG)的表达。
    共纳入23例,由7例AGS1与三个主要修复外切核酸酶1突变组成,3具有核糖核酸酶H2亚基B(RNASEH2B)突变的AGS2,3的ASG3与RNASEH2C,1个带有RNASEH2A突变的AGS4,2的AGS6与腺苷脱氨酶作用于RNA1突变,和7的AGS7与干扰素诱导的解旋酶C结构域1突变。3岁前发病占82.6%。神经系统受累最常见(100%),颅内钙化征象主要分布于双侧基底节区,脑白质营养不良,肌张力障碍,癫痫,脑萎缩和吞咽困难.智力残疾,还观察到语言障碍和运动技能障碍.皮肤表现(60.87%)以冻疮样皮疹为主。小头畸形(47.62%)等特征,身材矮小(52.38%),肝功能障碍(42.11%),甲状腺功能异常(46.15%),自身免疫抗体阳性(66.67%),红细胞沉降率升高(53.85%)。2例表型符合系统性红斑狼疮(SLE)诊断标准。记录了一个死亡。ISGs表达升高。
    AGS是一种引起后遗症和死亡的全身性疾病。对于早发性冻疮样皮疹的患者,应考虑AGS的诊断,颅内钙化,脑白质营养不良,肌张力障碍,发育迟缓,自身免疫抗体阳性,和提升的ISG,以及对常规治疗无反应的诊断为不典型表现的SLE患者。重要器官功能的综合评估和对症治疗很重要。
    Aicardi-Goutieres syndrome (AGS) is an inflammatory disorder belonging to the type I interferonopathy group. The clinical diagnosis of AGS is difficult, which can lead to a high mortality rate. Overall, there is a lack of large-sample research data on AGS in China. We aim to summarize the clinical characteristics of Chinese patients with AGS and provide clues for clinical diagnostic.
    The genetic and clinical features of Chinese patients with AGS were collected. Real-time polymerase chain reaction was used to detect expression of interferon-stimulated genes (ISGs).
    A total of 23 cases were included, consisting of 7 cases of AGS1 with three prime repair exonuclease 1 mutations, 3 of AGS2 with ribonuclease H2 subunit B (RNASEH2B) mutations, 3 of ASG3 with RNASEH2C, 1 of AGS4 with RNASEH2A mutations, 2 of AGS6 with adenosine deaminase acting on RNA 1 mutations, and 7 of AGS7 with interferon induced with helicase C domain 1 mutations. Onset before the age of 3 years occurred in 82.6%. Neurologic involvement was most common (100%), including signs of intracranial calcification which mainly distributed in the bilateral basal ganglia, leukodystrophy, dystonia, epilepsy, brain atrophy and dysphagia. Intellectual disability, language disability and motor skill impairment were also observed. Skin manifestations (60.87%) were dominated by a chilblain-like rash. Features such as microcephaly (47.62%), short stature (52.38%), liver dysfunction (42.11%), thyroid dysfunction (46.15%), positive autoimmune antibodies (66.67%), and elevated erythrocyte sedimentation rate (53.85%) were also found. The phenotypes of 2 cases fulfilled the diagnostic criteria for systemic lupus erythaematosus (SLE). One death was recorded. ISGs expression were elevated.
    AGS is a systemic disease that causes sequelae and mortality. A diagnosis of AGS should be considered for patients who have an early onset of chilblain-like rash, intracranial calcification, leukodystrophy, dystonia, developmental delay, positive autoimmune antibodies, and elevated ISGs, and for those diagnosed with SLE with atypical presentation who are nonresponsive to conventional treatments. Comprehensive assessment of vital organ function and symptomatic treatment are important.
    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
    许多全身性自身炎性疾病起因于编码IL-1激活性炎性体或细胞质核酸传感器(包括受体和传感器STING)的基因中的功能获得突变,并导致IL-1和I型干扰素产生增加,分别。在人类疾病中阻断这些途径提供了概念证明,证实了这些细胞因子在疾病发病机理中的重要作用。对这些传感器途径的多层调节的最新见解以及对它们在放大单基因和复杂遗传疾病的疾病发病机理中的作用的见解刺激了针对传感器的新药开发。这篇综述提供了对这些“原型”疾病的发病机理和遗传原因的见解,这些疾病是由IL-1激活性炎性体(炎症)和干扰素激活途径(干扰素病)中的功能获得突变引起的,包括STING相关的血管病变在婴儿期发病,Aicardi-Goutieres综合征,和蛋白酶体相关的自身炎症综合征,它们与病毒传感器STING的激活有关,“自我”核酸代谢,泛素-蛋白酶体系统对“I型干扰素生产”和人类疾病的影响。Janus激酶抑制剂的临床反应和生物标志物变化证实了干扰素的作用,越来越多的具有“干扰素特征”的疾病揭示了主要炎症途径之间的广泛交叉对话。了解这些相互作用有望提供新的工具来应对治疗这些疾病患者的重大临床挑战。
    A number of systemic autoinflammatory diseases arise from gain-of-function mutations in genes encoding IL-1-activating inflammasomes or cytoplasmic nucleic acid sensors including the receptor and sensor STING and result in increased IL-1 and type I interferon production, respectively. Blocking these pathways in human diseases has provided proof-of-concept, confirming the prominent roles of these cytokines in disease pathogenesis. Recent insights into the multilayered regulation of these sensor pathways and insights into their role in amplifying the disease pathogenesis of monogenic and complex genetic diseases spurred new drug development targeting the sensors. This review provides insights into the pathogenesis and genetic causes of these \"prototypic\" diseases caused by gain-of function mutations in IL-1-activating inflammasomes (inflammasomopathies) and in interferon-activating pathways (interferonopathies) including STING-associated vasculopathy with onset in infancy, Aicardi-Goutieres syndrome, and proteasome-associated autoinflammatory syndromes that link activation of the viral sensors STING, \"self\" nucleic acid metabolism, and the ubiquitin-proteasome system to \"type I interferon production\" and human diseases. Clinical responses and biomarker changes to Janus kinase inhibitors confirm a role of interferons, and a growing number of diseases with \"interferon signatures\" unveil extensive cross-talk between major inflammatory pathways. Understanding these interactions promises new tools in tackling the significant clinical challenges in treating patients with these conditions.
    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
    Interferon-induced with helicase C domain 1 (IFIH1) is a cytosolic sensor of dsRNA that induces an anti-viral Type I interferon (IFN) state. A gain-of-function mutation in IFIH1 can cause increased Type I IFN activity and is clinically associated with Aicardi-Goutières syndrome (AGS). AGS is a multisystem disease, characterized as an early-onset progressive encephalopathy with basal ganglia calcification and systemic lupus erythematosus-like features. Gastrointestinal manifestation is rare in AGS patients. We described a 10-year-old female patient with a heterozygous IFIH1 gene mutation who presented with gastrointestinal colitis, cystitis and very severe diarrhea as initial major manifestations of AGS. Proteinuria with high titer of antinuclear antibody and anti-double-stranded DNA was found in this patient. She also had growth retardation and a history of seizures (about two episodes each year) but without attacks until 7 years old. Serum cytokines detected by flow cytometry indicated extremely high level of interleukin 6 (1970.1 pg/ml) and IFN-α (204.1 pg/ml). A contrast-enhanced CT scan of the whole abdomen and an intestinal hydro-MRI indicated that the walls of her stomach, small bowel, colon, and bladder were in various degrees of edema and thickened states. Whole exome sequencing analysis indicated that she harbors an IFIH1 heterozygous mutation (c.2336G > A (p.R779H)) in both blood and intestinal samples. Abundant inflammatory cells infiltration into the intestinal epithelium was observed by immunohistochemical staining. Positive staining of caspase 4 and caspase 5 suggested that the signaling pathway of pyroptosis was involved in the mechanism of intestinal inflammation in AGS. Diarrhea was significantly improved after steroids and intravenous immunoglobulin treatments. Gastrointestinal colitis and cystitis can be rare manifestations of AGS with IFIH1 mutation. Caspase and its related inflammasome pathway may involve in the pathogenesis of AGS.
    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
    BACKGROUND: Aicardi-Goutières (AGS) is a rare immune dysregulated disease due to mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR1, or IFIH1. Clinical features include basal ganglia calcifications, white matter abnormalities, and cerebral atrophy. Severe systemic inflammation and chronic kidney disease (CKD) are extremely rare in AGS. Herein, we report a patient presenting with systemic inflammation and CKD to broaden the clinical phenotype spectrum of the RNASEH2B defect.
    METHODS: All testing and molecular genetic analysis were performed after obtaining the informed consent of the parents. Demographic, clinical, and laboratory findings were abstracted from outpatient and inpatient encounters. Cerebral magnetic resonance imaging (MRI), computed tomography (CT) scans, and renal biopsy histopathology reports were reviewed and summarized. Whole exome sequencing (WES) was performed on peripheral blood cells. After exposure to cGAMP in vitro for 24 h, mRNA expression of 12 IFN-stimulated cytokine genes in PBMCs was assessed. Serum cytokine levels were detected by Milliplex.
    RESULTS: A 11-year-old girl presented with recurrent aseptic fever, arthritis, chilblains, failure to thrive, mild hearing loss, and neurological manifestations. Laboratory and immunologic findings demonstrated lymphopenia, low complement levels, positive autoantibodies, elevated levels of acute-phase reactants and inflammatory cytokines. Cerebral imaging showed cerebral atrophy, white matter abnormalities, and intracranial calcification. Renal biopsy showed glomerular sclerosis in 3 of 14 glomeruli, infiltration of lymphocytes and other mononuclear cells. WES revealed a homozygous and heterozygous mutations in RNASEH2B. Over-expression of IFN-stimulated cytokine genes was observed, including IFI44, IFI27, IFIT1, IFIT2, IFIT3, ISG15, OAS1, and SIGLEC1.
    CONCLUSIONS: To date, only two cases with AGS have been reported to have renal disease. Here, we describe a patient with both homozygous and heterozygous variants in RNASEH2B, presenting with neurological manifestations, persistently systemic autoinflammation, and CKD. CKD has never been reported in patients with AGS due to the RNASEH2B defect.
    BACKGROUND: Not applicable; this was a retrospective study.
    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
    Transposable Elements (TEs) are mobile genetic elements whose sequences constitute nearly half of the human genome. Each TE copy can be present in hundreds to thousands of locations within the genome, complicating the genetic and genomic studies of these highly repetitive sequences. The recent development of better tools for evaluating TE derived sequences in genomic studies has enabled an increasing appreciation for the contribution of TEs to human development and disease. While some TEs have contributed novel and beneficial host functions, this review will summarize the evidence for detrimental TE activity in neurodegenerative disorders. Much of the evidence for pathogenicity implicates endogenous retroviruses (ERVs), a subset of TEs that entered the genome by retroviral infections of germline cells in our evolutionary ancestors and have since been passed down as a substantial fraction of the human genome. Human specific ERVs (HERVs) represent some of the youngest ERVs in the genome, and thus are presumed to retain greater function and resultant pathogenic potential.
    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
    Ribonucleases (RNases) are crucial for degradation of ribosomal RNA (rRNA). RNASET2 as a subtype of RNASEs is a 256 amino acid protein, encoded by RNASET2 gene located on chromosome six. Defective RNASET2 leads to RNASET2-deficient leukoencephalopathy, a rare autosomal recessive neurogenetic disorder with psychomotor delay as its main clinical symptom. The clinical findings can be similar to congenital cytomegalovirus (CMV) infection and Aicardi-Goutieres syndrome (AGS).
    Herein, we presented a patient with motor delay, neurological regression, infrequent seizures and microcephaly at 5 months of age. Brain imaging showed white matter involvement, calcification and anterior temporal cysts. Basic metabolic tests, serum and urine CMV polymerase chain reaction (PCR) were requested. According to clinical and imaging findings, screening of RNASET2 and RMND1 genes were performed. The clinical data and magnetic resonance imaging (MRI) findings of previous reported individuals with RNASET2-deficient leukodystrophy were also reviewed and compared to the findings of our patient.
    Brain MRI findings were suggestive of RNASET2-deficient leukoencephalopathy, AGS and CMV infection. Basic metabolic tests were normal and CMV PCR was negative. Molecular study revealed a novel homozygous variant of c.233C > A; p.Ser78Ter in exon 4 of RNASET2 gene compatible with the diagnosis of RNASET2-deficient leukoencephalopathy.
    RNASET2-deficiency is a possible diagnosis in an infant presented with a static leukoencephalopathy and white matter involvement without megalencephaly. Due to overlapping clinical and radiologic features of RNASET2-deficient leukoencephalopathy, AGS and congenital CMV infections, molecular study as an important and helpful diagnostic tool should be considered to avoid misdiagnosis.
    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