Aicardi–Goutières syndrome

Aicardi - Gouti è res 综合征
  • 文章类型: Case Reports
    背景:Aicardi-Goutières综合征(AGS)是一种罕见的以小头畸形为特征的遗传性疾病,白质病变,大量颅内钙化,冻疮皮肤损伤和脑脊液(CSF)中高水平的干扰素-α(IFN-α)。然而,据报道,眼部受累的频率明显较低。
    方法:我们介绍一例新生儿肥厚不足,小头畸形,冻伤样皮肤损伤,血小板减少症,肝酶升高和肝脾肿大。脑部磁共振成像(MRI)显示多个钙化灶,白质变化,脑萎缩,和心室系统的萎缩性扩张。炎症参数没有升高,排除了感染性病因。相反,检测到血清中IFN-α水平升高。根据相关临床症状,成像和测试结果,怀疑诊断为AGS.基因检测显示有两种致病突变,c.490C>T和c.222del(新突变),在三主要修复核酸外切酶1(TREX1)基因中,确认AGS类型1(AGS1)。对10个月大的儿童进行眼科检查,发现瞳孔对光的反应受损,右眼有Haab线的角膜雾霾(RE),双眼(OU)的浅色视神经盘和神经病变。RE的眼内压(IOP)为51mmHg,左眼(LE)为49mmHg。闪光视觉诱发电位(FVEP)在LE中显示出高达125%的延长的P2潜伏期,并降低了高达10%OU的幅度。这个女孩被诊断患有先天性青光眼,并通过小梁切除术和OU基底虹膜切除术来治疗,导致在没有任何低血压滴眼液的情况下,RE的IOP降低并稳定至12mmHg,LE的IOP稳定至10mmHg。
    结论:我们介绍了临床特征,电生理和成像发现,以及AGS1患者的基因检测结果。我们的病例有助于TREX1中致病性c.490C>T和c.222del突变的扩展眼科受累。
    BACKGROUND: Aicardi-Goutières syndrome (AGS) is a rare genetic disorder characterized by microcephaly, white matter lesions, numerous intracranial calcifications, chilblain skin lesions and high levels of interferon-α (IFN-α) in the cerebrospinal fluid (CSF). However, ocular involvement is reported significantly less frequently.
    METHODS: We present a case of a neonate with hypotrophy, microcephaly, frostbite-like skin lesions, thrombocytopenia, elevated liver enzymes and hepatosplenomegaly. Magnetic resonance imaging (MRI) of the brain showed multiple foci of calcification, white matter changes, cerebral atrophy, and atrophic dilatation of the ventricular system. The inflammatory parameters were not elevated, and the infectious etiology was excluded. Instead, elevated levels of IFN-α in the serum were detected. Based on the related clinical symptoms, imaging and test findings, the diagnosis of AGS was suspected. Genetic testing revealed two pathogenic mutations, c.490C>T and c.222del (novel mutation), in the three prime repair exonuclease 1 (TREX1) gene, confirming AGS type 1 (AGS1). An ophthalmologic examination of the child at 10 months of age revealed an impaired pupillary response to light, a corneal haze with Haab lines in the right eye (RE), pale optic nerve discs and neuropathy in both eyes (OU). The intraocular pressure (IOP) was 51 mmHg in the RE and 49 in the left eye (LE). The flash visual evoked potential (FVEP) showed prolonged P2 latencies of up to 125% in the LE and reduced amplitudes of up to approximately 10% OU. This girl was diagnosed with congenital glaucoma, and it was managed with a trabeculectomy with a basal iridectomy of OU, resulting in a reduction and stabilization in the IOP to 12 mmHg in the RE and 10 mmHg in the LE without any hypotensive eyedrops.
    CONCLUSIONS: We present the clinical characteristics, electrophysiological and imaging findings, as well as the genetic test results of a patient with AGS1. Our case contributes to the extended ophthalmic involvement of the pathogenic c.490C>T and c.222del mutations in TREX1.
    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(干扰素基因的环GMP-AMP合酶(cGAS)-刺激物(STING))轴将DNA损伤和细胞应激与I型干扰素(IFN)信号整合在一起,以促进炎症应激反应的转录变化。cGAS-STING途径以双链DNA的形式响应胞质DNA,微核,和长散布的核元素1(L1)反元素。L1逆转录元件是一类自我繁殖的非长末端重复转座子,其在哺乳动物基因组中保持高度活性。L1逆转录元件正在成为cGAS-STING和IFN信号的重要诱导剂,在几种疾病中经常失调,包括癌症.cGAS-STING和L1活性的关键阻遏物是核酸外切酶三主要修复核酸外切酶1(TREX1),和TREX1的损失促进L1的积累。此外,L1失调是通过cGAS-STING慢性诱导I型IFN信号传导的疾病中的共同主题,比如Aicardi-Goutières综合征,范可尼贫血,和皮肌炎.尽管TREX1在四足动物物种中高度保守,存在抑制L1逆转位的其他抑制蛋白。这些抑制基因在突变时通常与特征在于与高cGAS-STING活性和L1表达水平升高相关的未经检查的炎症的疾病相关。在这次审查中,我们讨论了这些相互关联的L1抑制途径及其在cGAS-STING和炎症疾病中的作用。
    The cGAS-STING (cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)) axis integrates DNA damage and cellular stress with type I interferon (IFN) signalling to facilitate transcriptional changes underlying inflammatory stress responses. The cGAS-STING pathway responds to cytosolic DNA in the form of double-stranded DNA, micronuclei, and long interspersed nuclear element 1 (L1) retroelements. L1 retroelements are a class of self-propagating non-long terminal repeat transposons that have remained highly active in mammalian genomes. L1 retroelements are emerging as important inducers of cGAS-STING and IFN signalling, which are often dysregulated in several diseases, including cancer. A key repressor of cGAS-STING and L1 activity is the exonuclease three prime repair exonuclease 1 (TREX1), and loss of TREX1 promotes the accumulation of L1. In addition, L1 dysregulation is a common theme among diseases with chronic induction of type I IFN signalling through cGAS-STING, such as Aicardi-Goutières syndrome, Fanconi anemia, and dermatomyositis. Although TREX1 is highly conserved in tetrapod species, other suppressor proteins exist that inhibit L1 retrotransposition. These suppressor genes when mutated are often associated with diseases characterized by unchecked inflammation that is associated with high cGAS-STING activity and elevated levels of L1 expression. In this review, we discuss these interconnected pathways of L1 suppression and their role in the regulation of cGAS-STING and inflammation in 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
    目的:报告Aicardi-Goutières综合征的一系列非典型表现。
    方法:临床,神经影像学,和遗传数据。
    结果:我们报告了一系列6名无关患者(5名男性)的亚急性发育里程碑丧失,锥体的迹象,和沟通能力的回归,发病年龄从7到20个月不等,在4到24周后达到最低点。在随访中,能力的丧失显着改善,他们仍然有残余的痉挛和构音障碍,但保留了认知功能。所有患者均在疾病发作前发生免疫或发热疾病。两名患者的脑脊液正常,在四个,出现边缘或轻度淋巴细胞增多。脑部CT扫描显示,六名患者中有一名患有细微的基底神经节钙化。脑MRI显示5例患者的脑白质不对称信号异常,累及半卵卵中心,其中1例弥漫性白质异常,对比增强。诊断并治疗了4例急性脱髓鞘性脑脊髓炎(ADEM)。随访1年或1年以上(平均7年),脑影像学明显改善,但患者仍有残余痉挛和构音障碍,无认知障碍。首次事件发生四年后,一名患者发生脱髓鞘复发。在所有患者中进行了全外显子组测序(WES):其中四名在RNASEH2B中发现了双等位基因致病性变体(三个纯合p.Ala177Thr和一个复合杂合p.Ala177Thr/p。Gln58*)和其中两个RNASEH2A中的相同纯合有害变体(p。Ala249Val)。
    结论:本报告扩大了AGS的表型,包括亚急性发育消退,并有部分临床和神经影像学改善。这些临床特征可能被误诊为ADEM。
    OBJECTIVE: To report a series of atypical presentations of Aicardi-Goutières syndrome.
    METHODS: Clinical, neuroimaging, and genetic data.
    RESULTS: We report a series of six unrelated patients (five males) with a subacute loss of developmental milestones, pyramidal signs, and regression of communication abilities, with onset at ages ranging from 7 to 20 months, reaching a nadir after 4 to 24 weeks. A remarkable improvement of lost abilities occurred in the follow-up, and they remained with residual spasticity and dysarthria but preserved cognitive function. Immunization or febrile illness occurred before disease onset in all patients. CSF was normal in two patients, and in four, borderline or mild lymphocytosis was present. A brain CT scan disclosed a subtle basal ganglia calcification in one of six patients. Brain MRI showed asymmetric signal abnormalities of white matter with centrum semi-ovale involvement in five patients and a diffuse white matter abnormality with contrast enhancement in one. Four patients were diagnosed and treated for acute demyelinating encephalomyelitis (ADEM). Brain imaging was markedly improved with one year or more of follow-up (average of 7 years), but patients remained with residual spasticity and dysarthria without cognitive impairment. Demyelination relapse occurred in a single patient four years after the first event. Whole-exome sequencing (WES) was performed in all patients: four of them disclosed biallelic pathogenic variants in RNASEH2B (three homozygous p.Ala177Thr and one compound heterozygous p.Ala177Thr/p.Gln58*) and in two of them the same homozygous deleterious variants in RNASEH2A (p.Ala249Val).
    CONCLUSIONS: This report expands the phenotype of AGS to include subacute developmental regression with partial clinical and neuroimaging improvement. Those clinical features might be misdiagnosed as ADEM.
    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-Goutières综合征(AGS)是一种罕见的,常染色体隐性遗传,遗传性神经退行性疾病.它的特点主要是早发性进行性脑病,伴随着脑脊液中干扰素-α水平的增加。植入前遗传测试(PGT)是一种程序,可用于在分析活检细胞后选择未受影响的胚胎进行转移,这可以防止有风险的夫妇面临终止妊娠的风险。
    方法:基于Trio的全外显子组测序,核型分析和染色体微阵列分析用于确定该家族的致病突变。为了阻止疾病的遗传,使用多个退火和基于循环的扩增循环对活检滋养外胚层细胞进行全基因组扩增.使用Sanger测序和基于下一代测序(NGS)的单核苷酸多态性(SNP)单倍型分析来检测基因突变的状态。还进行了拷贝数变异(CNV)分析以防止胚胎染色体异常。进行产前诊断以验证PGT结果。
    结果:在致AGS的先证者中发现了一种新的TREX1基因复合杂合突变。对胞浆内单精子注射后形成的总共3个囊胚进行了活检。经过基因分析,在TREX1中携带杂合突变且无CNV的胚胎被转移。一个健康的婴儿在第38周出生,产前诊断结果证实了PGT的准确性。
    结论:在这项研究中,我们在TREX1中发现了两个新的致病突变,这在以前没有报道过.我们的研究扩展了TREX1基因的突变谱,有助于AGS的分子诊断和遗传咨询。我们的结果表明,将基于NGS的PGT-MSNP单倍分型与侵入性产前诊断相结合是阻断AGS传播的有效方法,可用于预防其他单基因疾病。
    BACKGROUND: Aicardi-Goutières syndrome (AGS) is a rare, autosomal recessive, hereditary neurodegenerative disorder. It is characterized mainly by early onset progressive encephalopathy, concomitant with an increase in interferon-α levels in the cerebrospinal fluid. Preimplantation genetic testing (PGT) is a procedure that could be used to choose unaffected embryos for transfer after analysis of biopsied cells, which prevents at-risk couples from facing the risk of pregnancy termination.
    METHODS: Trio-based whole exome sequencing, karyotyping and chromosomal microarray analysis were used to determine the pathogenic mutations for the family. To block the inheritance of the disease, multiple annealing and looping-based amplification cycles was used for whole genome amplification of the biopsied trophectoderm cells. Sanger sequencing and next-generation sequencing (NGS)-based single nucleotide polymorphism (SNP) haplotyping were used to detect the state of the gene mutations. Copy number variation (CNV) analysis was also carried out to prevent embryonic chromosomal abnormalities. Prenatal diagnosis was preformed to verify the PGT outcomes.
    RESULTS: A novel compound heterozygous mutation in TREX1 gene was found in the proband causing AGS. A total of 3 blastocysts formed after intracytoplasmic sperm injection were biopsied. After genetic analyses, an embryo harbored a heterozygous mutation in TREX1 and without CNV was transferred. A healthy baby was born at 38th weeks and prenatal diagnosis results confirmed the accuracy of PGT.
    CONCLUSIONS: In this study, we identified two novel pathogenic mutations in TREX1, which has not been previously reported. Our study extends the mutation spectrum of TREX1 gene and contributes to the molecular diagnosis as well as genetic counseling for AGS. Our results demonstrated that combining NGS-based SNP haplotyping for PGT-M with invasive prenatal diagnosis is an effective approach to block the transmission of AGS and could be applied to prevent other monogenic diseases.
    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
    Aicardi-Goutières综合征(AGS)是一种常染色体隐性遗传疾病,模拟先天性病毒感染,主要影响大脑,免疫系统,和皮肤。主要临床症状是亚急性发作的严重脑病,表现为易怒,能力的丧失,头部生长放缓,和营养不良。AGS中的动脉病是一种罕见的表现,通常与SAMHD1基因突变有关。我们提出了一个罕见的3岁男性由于未能茁壮成长的案例,全球发育迟缓,小头畸形,视力差,上肢和下肢痉挛,胃食管反流病(GERD),在出生后的早期生活中,颅内大,中动脉的早期狭窄病变伴有缺血性后遗症。进行基因检测证实纯合基因突变,SAMHD1与AGS类型5相关联。通过查阅现有文献,我们仅发现1例患者在6个月后诊断为动脉病变.
    Aicardi-Goutières syndrome (AGS) is an autosomal recessive disease that mimics congenital viral infection and mainly affects the brain, immune system, and skin. The dominant clinical symptom is the subacute onset of severe encephalopathy, which manifests as irritability, loss of ability, slowing of head growth, and poor nutrition. Arteriopathy in AGS is an uncommon manifestation usually associated with mutations in the SAMHD1 gene. We present a rare case of a 3-year-old male due to failure to thrive, global developmental delay, microcephaly, poor vision, upper and lower limbs spasticity, and gastroesophageal reflux disease (GERD), who harbored early stenotic lesions of the large and medium intracranial arteries with ischemic sequelae in the early postnatal life. Performed genetic testing confirmed homozygous gene mutation, SAMHD1 associated with AGS type 5. By reviewing the available literature, we were able to find only one patient whose arterial lesions were diagnosed after 6 months.
    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-Goutières综合征(AGS)是一种遗传决定的疾病,具有可变的表型。自从AGS的原始描述以来,基因测序技术的进步导致与AGS基因相关的表型谱显著扩大,新的临床图片已经超越了经典演示。这篇综述的目的是对AGS的临床谱进行全面分析,并报告目前可用的治疗方法和新的免疫抑制策略。
    方法:从数据库中收集文献综述和原始研究文章,包括PubMed和ClinicalTrials.gov.包括有关AGS的相关文章。
    结果:神经系统受累无疑是AGS患者死亡和发病的主要原因。然而,其他临床表现,比如冻疮,肝脾肿大,和血液学紊乱,可能导致诊断,并大大影响这些患者的预后和整体生活质量。AGS的治疗方法仅限于针对特定症状的干预措施和多种合并症的管理。然而,在理解AGS发病机制方面取得的进展可以开辟新的更有效的治疗方法.
    结论:由于干扰素产生上调导致的先天免疫过度激活在AGS中起关键作用,导致多器官损伤,主要累及中枢神经系统。迄今为止,对于AGS没有特异性和有效的治疗方法。专门针对干扰素途径的新药可能给AGS患者带来新的希望。
    Aicardi-Goutières syndrome (AGS) is a genetically determined disorder with a variable phenotype. Since the original description of AGS, advances in gene sequencing techniques have resulted in a significant broadening of the phenotypic spectrum associated with AGS genes, and new clinical pictures have emerged beyond the classic presentation. The aim of this review is to provide a comprehensive analysis of the clinical spectrum of AGS and report currently available treatments and new immunosuppressive strategies.
    Literature reviews and original research articles were collected from databases, including PubMed and ClinicalTrials.gov. Relevant articles about AGS were included.
    The involvement of the nervous system certainly represents the major cause of mortality and morbidity in AGS patients. However, other clinical manifestations, such as chilblains, hepatosplenomegaly, and hematological disturbances, may lead to the diagnosis and considerably impact the prognosis and overall quality of life of these patients. Therapeutic approaches of AGS are limited to interventions aimed at specific symptoms and the management of multiple comorbidities. However, advances in understanding the pathogenesis of AGS could open new and more effective therapies.
    The over-activation of innate immunity due to upregulated interferon production plays a critical role in AGS, leading to multi-organ damage with the main involvement of the central nervous system. To date, there is no specific and effective treatment for AGS. New drugs specifically targeting the interferon pathway may bring new hope to AGS patients.
    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
    作用于RNA1的腺苷脱氨酶(ADAR1)是一种RNA编辑酶,其催化双链RNA(dsRNA)中的腺苷到肌苷的转化。在哺乳动物中,ADAR1由两种同工型组成:核短p110同工型和细胞质长p150同工型。而两种同工型都含有右手dsRNA结合和脱氨酶结构域,ADAR1p150具有与左手dsRNA结合的Zα结构域,称为Z-RNA。内源性dsRNAs作为非自身的MDA5传感导致I型干扰素(IFN)刺激基因的诱导,但最近的研究表明,ADAR1p150介导的RNA编辑,但ADAR1p110不能阻止这种MDA5介导的感知。存在ADAR1p150特异性RNA编辑位点,并且这种特异性至少需要Zα结构域-Z-RNA相互作用。ADAR1基因突变导致Aicardi-Goutières综合征(AGS),婴儿脑病与I型干扰素过度生产。在Adar1基因的Zα结构域插入点突变诱导小鼠AGS样脑病,这是通过同时删除MDA5来拯救的。这一发现表明受损的ADAR1p150介导的RNA编辑是由ADAR1突变引起的AGS的潜在机制。ADAR1p150还可以防止ZBP1对内源性Z-RNA的感应,导致细胞程序性死亡,通过Zα结构域及其RNA编辑活性。此外,ADAR1阻止内源性右手dsRNA的PKR传感,导致转化关闭和增长停滞。因此,ADAR1充当调控枢纽,阻断内源性dsRNAs作为非自身的多个传感蛋白,无论是在RNA编辑依赖和独立的方式,并且是疾病的潜在治疗目标,尤其是癌症。
    Adenosine deaminase acting on RNA 1 (ADAR1) is an RNA-editing enzyme that catalyzes adenosine-to-inosine conversions in double-stranded RNAs (dsRNAs). In mammals, ADAR1 is composed of two isoforms: a nuclear short p110 isoform and a cytoplasmic long p150 isoform. Whereas both isoforms contain right-handed dsRNA-binding and deaminase domains, ADAR1 p150 harbors a Zα domain that binds to left-handed dsRNAs, termed Z-RNAs. Myeloma differentiation-associated gene 5 (MDA5) sensing of endogenous dsRNAs as non-self leads to the induction of type I interferon (IFN)-stimulated genes, but recent studies revealed that ADAR1 p150-mediated RNA editing, but not ADAR1 p110, prevents this MDA5-mediated sensing. ADAR1 p150-specific RNA-editing sites are present and at least a Zα domain-Z-RNA interaction is required for this specificity. Mutations in the ADAR1 gene cause Aicardi-Goutières syndrome (AGS), an infant encephalopathy with type I IFN overproduction. Insertion of a point mutation in the Zα domain of the Adar1 gene induces AGS-like encephalopathy in mice, which is rescued by concurrent deletion of MDA5. This finding indicates that impaired ADAR1 p150-mediated RNA-editing is a mechanism underlying AGS caused by an ADAR1 mutation. ADAR1 p150 also prevents ZBP1 sensing of endogenous Z-RNA, which leads to programmed cell death, via the Zα domain and its RNA-editing activity. Furthermore, ADAR1 prevents protein kinase R (PKR) sensing of endogenous right-handed dsRNAs, which leads to translational shutdown and growth arrest. Thus, ADAR1 acts as a regulatory hub that blocks sensing of endogenous dsRNAs as non-self by multiple sensor proteins, both in RNA editing-dependent and -independent manners, and is a potential therapeutic target for diseases, especially cancer.
    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
    Aicardi-Goutières综合征(AGS)是一种罕见的脑病,其特征是神经系统和免疫学特征。线粒体功能障碍可能导致线粒体DNA(mtDNA)释放和随之而来的免疫系统激活。我们通过研究RNASEH2B和RNASEH2A基因突变的患者,研究了线粒体和mtDNA在AGS发病机制中的作用。使用来自RNASEH2A-和RNASEH2B-突变患者的淋巴母细胞样细胞系(LCL)和健康对照。透射电子显微镜(TEM)和流式细胞术用于评估形态学改变,活性氧(ROS)的产生和线粒体膜电位的变化。海马分析仪用于研究代谢改变,通过免疫荧光评估mtDNA氧化和VDAC1寡聚化。Western印迹和RT-qPCR用于定量mtTFA蛋白和mtDNA释放。在两个突变的LCL中观察到线粒体的形态学改变,主要在RNASEH2ALCLs中发现生理膜电位的损失。RNASEH2BLCL中的ROS产生和8-氧代鸟嘌呤水平增加。此外,VDAC1信号增加,表明线粒体孔的形成可能决定mtDNA的释放。的确,在RNASEH2BLCLs中发现较高的细胞质mtDNA水平。代谢改变证实了两种LCL中的线粒体损伤。数据强调了AGS患者的线粒体改变,表明在AGS发病机理中起关键作用。
    Aicardi-Goutières syndrome (AGS) is a rare encephalopathy characterized by neurological and immunological features. Mitochondrial dysfunctions may lead to mitochondrial DNA (mtDNA) release and consequent immune system activation. We investigated the role of mitochondria and mtDNA in AGS pathogenesis by studying patients mutated in RNASEH2B and RNASEH2A genes. Lymphoblastoid cell lines (LCLs) from RNASEH2A- and RNASEH2B-mutated patients and healthy control were used. Transmission Electron Microscopy (TEM) and flow cytometry were used to assess morphological alterations, reactive oxygen species (ROS) production and mitochondrial membrane potential variations. Seahorse Analyzer was used to investigate metabolic alterations, and mtDNA oxidation and VDAC1 oligomerization were assessed by immunofluorescence. Western blot and RT-qPCR were used to quantify mtTFA protein and mtDNA release. Morphological alterations of mitochondria were observed in both mutated LCLs, and loss of physiological membrane potential was mainly identified in RNASEH2A LCLs. ROS production and 8-oxoGuanine levels were increased in RNASEH2B LCLs. Additionally, the VDAC1 signal was increased, suggesting a mitochondrial pore formation possibly determining mtDNA release. Indeed, higher cytoplasmic mtDNA levels were found in RNASEH2B LCLs. Metabolic alterations confirmed mitochondrial damage in both LCLs. Data highlighted mitochondrial alterations in AGS patients\' LCLs suggesting a pivotal role in AGS 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
    核酸是先天免疫的强大触发因素,可以采用Z构象,不寻常的左旋双螺旋结构.这里,我们研究了腺苷脱氨酶ADAR1对Z-RNA识别的生物学功能,该突变导致Aicardi-Goutières综合征.Adar1mZα/mZα小鼠,在Z-核酸结合(Zα)结构域中带有两个消除Z-RNA结合的点突变,在多个器官中显示I型干扰素(IFNs)的自发诱导,包括肺部,其中基质细胞和造血细胞均显示IFN刺激基因(ISG)诱导。肺嗜中性粒细胞表达由转录因子IRF3诱导的ISG,表明嗜中性粒细胞在这种IFN应答中的启动作用。Adar1mZα/mZα小鼠的IFN应答需要衔接子MAVS,涉及胞质RNA传感。腺苷到肌苷的变化富含转座因子,并揭示了在编辑RNA子集时ADAR1的Zα结构域的特定需求。因此,Z构象的内源性RNA具有被ADAR1抑制的免疫刺激潜能,与人类自身炎性疾病相关.
    Nucleic acids are powerful triggers of innate immunity and can adopt the Z-conformation, an unusual left-handed double helix. Here, we studied the biological function(s) of Z-RNA recognition by the adenosine deaminase ADAR1, mutations in which cause Aicardi-Goutières syndrome. Adar1mZα/mZα mice, bearing two point mutations in the Z-nucleic acid binding (Zα) domain that abolish Z-RNA binding, displayed spontaneous induction of type I interferons (IFNs) in multiple organs, including in the lung, where both stromal and hematopoietic cells showed IFN-stimulated gene (ISG) induction. Lung neutrophils expressed ISGs induced by the transcription factor IRF3, indicating an initiating role for neutrophils in this IFN response. The IFN response in Adar1mZα/mZα mice required the adaptor MAVS, implicating cytosolic RNA sensing. Adenosine-to-inosine changes were enriched in transposable elements and revealed a specific requirement of ADAR1\'s Zα domain in editing of a subset of RNAs. Thus, endogenous RNAs in Z-conformation have immunostimulatory potential curtailed by ADAR1, with relevance to autoinflammatory disease in humans.
    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
    黑色素瘤分化相关蛋白5(MDA5)是由IFIH1在人类中编码的关键RIG-I样受体RNA解旋酶。IFIH1中的单核苷酸多态性导致致命的遗传疾病,如Aicardi-Goutières综合征和Singleton-Merten综合征,以及人类患I型糖尿病的风险增加。在这项研究中,我们选择了MDA5蛋白的四个不同的氨基酸替换负责遗传性疾病:MDA5L372F,MDA5A452T,MDA5R779H,和MDA5R822Q,并使用分子动力学模拟分析了它们的结构和功能关系。我们的结果表明,突变的复合物比野生型MDA5相对更稳定。回转半径,相互作用能,和氢键内分析表明突变复合物相对于野生型的稳定性,特别是MDA5L372F和MDA5R822Q。野生型和突变体复合物表现出的显性运动差异很大。此外,野生型和突变体复合物的中间性中心性显示了信号内传播的共享残基。观察到的结果表明,突变导致功能的获得,正如以前的研究报告,由于突变复合物中RNA和MDA5之间的相互作用能和稳定性增加。这些发现有望加深我们对MDA5变体的理解,并可能有助于开发针对这些疾病的相关疗法。
    Melanoma differentiation-associated protein 5 (MDA5) is a crucial RIG-I-like receptor RNA helicase enzyme encoded by IFIH1 in humans. Single nucleotide polymorphisms in the IFIH1 results in fatal genetic disorders such as Aicardi-Goutières syndrome and Singleton-Merten syndrome, and in increased risk of type I diabetes in humans. In this study, we chose four different amino acid substitutions of the MDA5 protein responsible for genetic disorders: MDA5L372F, MDA5A452T, MDA5R779H, and MDA5R822Q and analyzed their structural and functional relationships using molecular dynamic simulations. Our results suggest that the mutated complexes are relatively more stable than the wild-type MDA5. The radius of gyration, interaction energies, and intra-hydrogen bond analysis indicated the stability of mutated complexes over the wild type, especially MDA5L372F and MDA5R822Q. The dominant motions exhibited by the wild-type and mutant complexes varied significantly. Moreover, the betweenness centrality of the wild-type and mutant complexes showed shared residues for intra-signal propagation. The observed results indicate that the mutations lead to a gain of function, as reported in previous studies, due to increased interaction energies and stability between RNA and MDA5 in mutated complexes. These findings are expected to deepen our understanding of MDA5 variants and may assist in the development of relevant therapeutics against the disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号