Haploinsufficiency

单倍功能不全
  • 文章类型: Journal Article
    目的:PURA基因中的罕见遗传变异导致PURA相关神经发育障碍(PURA-NDD),以新生儿异常和发育迟缓为特征。使用全基因组DNA甲基化分析对患有PURA变异的患者,我们的目标是建立PURA-NDD特异性甲基化谱,并提供对PURA-NDD分子基础的进一步见解。
    方法:纳入了23名携带PURA变异体的个体(包括12名未发表的个体)。我们对17名PURA-NDD个体进行了IlluminaInfiniumEPIC微阵列分析。进行体外实验以检查PURA变体如何影响Pur-α表达。
    结果:本研究描述了12名新发现的患者的其他表型。全基因组DNA甲基化分析揭示了PURA-NDD独特的甲基化谱,建立的分类器可以对不确定意义的PURA变体进行重新分类。带有PURAhapolocomfortness和错义变异的患者具有可比的DNA甲基化倾向,表达这些PURA变体的细胞显示出一致的Pur-α下调,表明单倍体不足机制。
    结论:PURA-NDD患者表现出特定的epi特征,这有可能帮助鉴定和诊断PURA-NDD患者,并为进一步的功能调查提供启示。
    OBJECTIVE: Rare genetic variants in the PURA gene cause the PURA-related neurodevelopmental disorder (PURA-NDD), characterized by neonatal abnormalities and developmental delay. Using genome-wide DNA methylation analysis on patients with PURA variants, we aim to establish a PURA-NDD-specific methylation profile and provide further insights on the molecular basis of the PURA-NDD.
    METHODS: Twenty three individuals (including 12 unpublished) carrying PURA variants were enrolled. We conducted the Illumina Infinium EPIC microarray analysis in 17 PURA-NDD individuals. In vitro experiments were performed to examine how PURA variants affect Pur-a expression.
    RESULTS: Additional phenotypes in 12 newly identified patients were described in this study. Genome-wide DNA methylation analysis unveiled distinctive methylation profiles to PURA-NDD, and the established classifier can reclassify PURA variants of uncertain significance. Patients bearing PURA hapoloinsufficient and missense variants have comparable DNA methylation profiles, and cells expressing these PURA variants showed consistent Pur-a downregulation, suggesting a haploinsufficiency mechanism.
    CONCLUSIONS: Patients with PURA-NDD exhibit a specific episignature, which has potential to aid identification and diagnosis of PURA-NDD patients and offer implications for further functional investigations.
    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
    SRCAP基因的突变是在自闭症谱系障碍(ASD)中鉴定的遗传改变之一。然而,致病机制尚不清楚。在这项研究中,我们证明Srcap+/-小鼠在社会新颖性反应中表现出缺陷,以及增加的重复行为,焦虑,学习和记忆障碍。值得注意的是,在这些小鼠的脾后皮质(RSC)和齿状回(DG)中观察到小白蛋白阳性神经元的减少。通过RNA测序,我们在Srcap+/-小鼠中鉴定了27个ASD相关基因的失调。具体来说,我们发现Srcap通过H2A调节Satb2的表达。z在启动子中。通过在新生Srcap/-小鼠中眶后注射腺相关病毒(AAV)-Satb2的治疗干预导致神经发育和ASD样异常的改善。此外,Satb2仅在青春期小鼠的RSC中的表达纠正了社交新颖性损害。这些结果强调了Srcap在神经发育中的关键作用,通过调节Satb2,为ASD的病理生理学提供有价值的见解。
    Mutations in the SRCAP gene are among the genetic alterations identified in autism spectrum disorders (ASD). However, the pathogenic mechanisms remain unclear. In this study, we demonstrate that Srcap+/- mice manifest deficits in social novelty response, as well as increased repetitive behaviors, anxiety, and impairments in learning and memory. Notably, a reduction in parvalbumin-positive neurons is observed in the retrosplenial cortex (RSC) and dentate gyrus (DG) of these mice. Through RNA sequencing, we identify dysregulation in 27 ASD-related genes in Srcap+/- mice. Specifically, we find that Srcap regulates expression of Satb2 via H2A.z in the promoter. Therapeutic intervention via retro-orbital injection of adeno-associated virus (AAV)-Satb2 in neonatal Srcap+/- mice leads to amelioration of the neurodevelopmental and ASD-like abnormalities. Furthermore, the expression of Satb2 only in the RSC of adolescent mice rectifies social novelty impairments. These results underscore the pivotal role of Srcap in neurodevelopment, by regulating Satb2, providing valuable insights for the pathophysiology of ASD.
    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
    许多类型的肿瘤细胞改变代谢途径以满足其对增殖或应激适应的能量和生物合成需求。在本期《细胞》中,Kong等人。发现糖酵解代谢产物甲基乙二醛通过诱导BRCA2蛋白水解导致癌症相关的突变单碱基取代特征,导致BRCA2的功能单倍体不足。
    Many types of tumor cells alter metabolic pathways to meet their energy and biosynthetic demands for proliferation or stress adaptation. In this issue of Cell, Kong et al. find that the glycolytic metabolite methylglyoxal causes cancer-associated mutant single-base substitution features by inducing BRCA2 proteolysis, leading to functional haploinsufficiency of BRCA2.
    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
    Na+/K+-ATP酶(NKA)的功能障碍已在各种神经退行性疾病中被证明。然而,NKAα1在帕金森病(PD)中的具体作用尚不完全清楚。在这次调查中,我们利用NKAα1单倍体功能不全(NKAα1/-)小鼠来探讨NKAα1对1-甲基-4-苯基-1,2,3,6-四氢吡啶(MPTP)诱导的多巴胺能(DA)神经变性的影响。我们的发现表明,与暴露于MPTP的对照组相比,NKAα1/-小鼠表现出DA神经元的高度丧失和更明显的运动功能障碍。有趣的是,这种现象与体内和体外铁凋亡的激活和线粒体自噬的受损同时发生。为了研究NKAα1在PD中的作用和潜在机制,我们用了DR-Ab,靶向NKAα亚基的DR区的抗体。我们的研究表明,DR-Ab的施用有效地恢复了NKAa1的膜丰度,从而减轻了MPTP诱导的DA神经元丢失并随后改善了行为缺陷。机械上,DR-Ab增强了表面NKAα1/SLC7A11复合物的形成,抑制SLC7A11依赖性铁性凋亡。此外,DR-Ab破坏了NKAα1和Parkin之间的胞浆相互作用,促进Parkin向线粒体的易位并增强线粒体自噬的过程。总之,本研究确立了NKAα1作为铁凋亡和线粒体自噬的关键调节因子,确定其DR区作为PD的有希望的治疗靶标。
    Dysfunction of the Na+/K+-ATPase (NKA) has been documented in various neurodegenerative diseases, yet the specific role of NKAα1 in Parkinson\'s disease (PD) remains incompletely understood. In this investigation, we utilized NKAα1 haploinsufficiency (NKAα1+/-) mice to probe the influence of NKAα1 on dopaminergic (DA) neurodegeneration induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Our findings reveal that NKAα1+/- mice displayed a heightened loss of DA neurons and more pronounced motor dysfunction compared to the control group when exposed to MPTP. Intriguingly, this phenomenon coincided with the activation of ferroptosis and impaired mitophagy both in vivo and in vitro. To scrutinize the role and underlying mechanism of NKAα1 in PD, we employed DR-Ab, an antibody targeting the DR-region of the NKA α subunit. Our study demonstrates that the administration of DR-Ab effectively reinstated the membrane abundance of NKAα1, thereby mitigating MPTP-induced DA neuron loss and subsequent improvement in behavioral deficit. Mechanistically, DR-Ab heightened the formation of the surface NKAα1/SLC7A11 complex, inhibiting SLC7A11-dependent ferroptosis. Moreover, DR-Ab disrupted the cytosolic interaction between NKAα1 and Parkin, facilitating the translocation of Parkin to mitochondria and enhancing the process of mitophagy. In conclusion, this study establishes NKAα1 as a key regulator of ferroptosis and mitophagy, identifying its DR-region as a promising therapeutic target for PD.
    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:孤立性生长激素缺乏症(IGHD)是一种罕见的遗传异质性疾病,主要由GH1和GH释放激素受体(GHRHR)突变引起。这项研究的目的是确定IGHD中国男孩的分子病因。
    方法:全外显子组测序,进行sanger测序和生物信息学分析以筛选候选突变.候选突变对基因表达的影响,通过体外分析进一步评估细胞内定位和蛋白质功能。
    结果:GHRH基因中的一种新的杂合移码突变(c.91dupC,p.R31Pfs*98)在临床诊断为患有IGHD的中国男孩中鉴定。该突变在多个公共数据库中不存在,使用计算机预测被认为是有害的,保守分析和三维同源建模。此外,突变型GHRH的mRNA和蛋白表达水平显著高于野生型GHRH(p<0.05)。此外,突变GHRH显示细胞质内的异常积累,与野生型GHRH相比,人表达GHRHR的垂体GH3细胞刺激GH分泌和cAMP积累的能力明显降低(p<0.05)。
    结论:我们的研究发现了一个中国男孩IGHD中GHRH的功能缺失突变,并为GHRH单倍体功能不足引起的IGHD发病机制提供了新的见解。
    BACKGROUND: Isolated growth hormone deficiency (IGHD) is a rare genetically heterogeneous disorder caused primarily by mutations in GH1 and GH releasing hormone receptor (GHRHR). The aim of this study was to identify the molecular etiology of a Chinese boy with IGHD.
    METHODS: Whole-exome sequencing, sanger sequencing and bioinformatic analysis were performed to screen for candidate mutations. The impacts of candidate mutation on gene expression, intracellular localization and protein function were further evaluated by in vitro assays.
    RESULTS: A novel heterozygous frameshift mutation in the GHRH gene (c.91dupC, p.R31Pfs*98) was identified in a Chinese boy clinically diagnosed as having IGHD. The mutation was absent in multiple public databases, and considered as deleterious using in silico prediction, conservative analysis and three-dimensional homology modeling. Furthermore, mRNA and protein expression levels of mutant GHRH were significantly increased than wild-type GHRH (p < 0.05). Moreover, mutant GHRH showed an aberrant accumulation within the cytoplasm, and obviously reduced ability to stimulate GH secretion and cAMP accumulation in human GHRHR-expressing pituitary GH3 cells compared to wild-type GHRH (p < 0.05).
    CONCLUSIONS: Our study discovered the first loss-of function mutation of GHRH in a Chinese boy with IGHD and provided new insights on IGHD pathogenesis caused by GHRH haploinsufficiency.
    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
    年龄相关的B细胞(ABCs)在感染过程中积累,老化,和自身免疫,有助于狼疮的发病机制。在这项研究中,我们筛选了驱动ABC形成的转录因子,发现锌指E盒结合同源盒2(ZEB2)是体外人和小鼠ABC分化所必需的。在ZEB2单倍体不足的个体中和在B细胞中缺乏Zeb2的小鼠中,ABCs减少。在Toll样受体7(TLR7)驱动的狼疮小鼠中,ZEB2对于ABC形成和自身免疫病理学至关重要。ZEB2结合+20-kb肌细胞增强因子2b(Mef2b)的内含子增强子,抑制MEF2B介导的生发中心B细胞分化并促进ABC形成。ZEB2还靶向对ABC规范和功能重要的基因,包括Itgax.ZEB2驱动的ABC分化需要JAK-STAT(Janus激酶-信号转导和转录激活因子),用JAK1/3抑制剂治疗可减少自身免疫小鼠和患者的ABC积累。因此,ZEB2是B细胞自身免疫的驱动因素。
    Age-associated B cells (ABCs) accumulate during infection, aging, and autoimmunity, contributing to lupus pathogenesis. In this study, we screened for transcription factors driving ABC formation and found that zinc finger E-box binding homeobox 2 (ZEB2) is required for human and mouse ABC differentiation in vitro. ABCs are reduced in ZEB2 haploinsufficient individuals and in mice lacking Zeb2 in B cells. In mice with toll-like receptor 7 (TLR7)-driven lupus, ZEB2 is essential for ABC formation and autoimmune pathology. ZEB2 binds to +20-kb myocyte enhancer factor 2b (Mef2b)\'s intronic enhancer, repressing MEF2B-mediated germinal center B cell differentiation and promoting ABC formation. ZEB2 also targets genes important for ABC specification and function, including Itgax. ZEB2-driven ABC differentiation requires JAK-STAT (Janus kinase-signal transducer and activator of transcription), and treatment with JAK1/3 inhibitor reduces ABC accumulation in autoimmune mice and patients. Thus, ZEB2 emerges as a driver of B cell autoimmunity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Letter
    目的:线粒体稳态对于维持氧化还原平衡至关重要。除了规范的自噬,Rab9依赖性选择性线粒体自噬是代谢性心肌病的重要机制。这里,我们旨在研究选择性线粒体自噬和Beclin1单倍体功能不全(Beclin1+/-)在高脂饮食(HFD)诱导的代谢性心肌病中的作用.
    结果:WT小鼠24周HFD糖耐量和心肌细胞收缩受损,两者都在Beclin1+/-小鼠中获救。Beclin1单倍体功能不全对常规自噬介质(ATG5,LC3II/LC3I)影响不大,但进一步上调Rab9表达,替代自噬的标记,响应HFD挑战。此外,选择性自噬或Beclin1单倍体功能不全的抑制均可消除棕榈酸诱导的心肌细胞收缩异常。体外,棕榈酸过度激活线粒体自噬,导致H9C2细胞中线粒体含量降低。这些畸变在缺乏替代性自噬的细胞中得到缓解,但在缺乏常规自噬的细胞中没有得到缓解。机械上,HFD促进了ROS的生产,激活的Rab9依赖性选择性线粒体自噬,并抑制线粒体生物合成。Beclin1+/-拯救HFD诱导的ROS溢出,线粒体生物发生损伤,并阻止Rab9从细胞质转移到线粒体,从而抑制Rab9介导的线粒体自噬过度激活。
    方法:第一次,本研究提示,持续的选择性线粒体自噬加重了慢性HFD诱导的心功能不全,并支持Beclin1单倍体功能不全在代谢性心肌病中的保护作用.这为基于靶标的药理学干预提供了额外的证据。
    结论:Beclin1单倍体功能不全通过抑制Rab9依赖的选择性线粒体自噬和ROS的产生来保护HFD引起的心功能不全,同时促进线粒体生物发生。调节Beclin1表达有望预防慢性HFD相关心肌病。
    暂无摘要。
    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
    A20单倍功能不全(HA20)是一种罕见的单基因疾病,由位于染色体6q2.3的肿瘤坏死因子α诱导蛋白3(TNFAIP3)基因的杂合功能丧失突变引起。大多数HA20病例中的大多数致病突变包含单核苷酸变异,小插入,或TNFAIP3中的缺失,导致提前终止密码子,并随后破坏其抗炎作用。零星报道了大量缺失。HA20患者在儿童早期可能存在多种自身炎症和自身免疫特征;然而,新生儿发病的病例很少见。这里,我们描述了1例中国新生儿,其表现为6号染色体5.15Mb间质缺失引起的伴随炎症和其他综合征表现;这些缺失影响TNFAIP3.一起来看,数据扩展了HA20的临床和遗传谱。
    Haploinsufficiency of A20 (HA20) is a rare monogenic disease caused by heterozygous loss-of-function mutations in the tumor necrosis factor alpha-induced protein 3 (TNFAIP3) gene located on chromosome 6q23.3. The majority of disease-causing mutations in most cases of HA20 comprise single nucleotide variations, small insertions, or deletions in TNFAIP3, which result in a premature termination codon and subsequent disruption of its anti-inflammatory role. Large deletions have been reported sporadically. HA20 patients may present with a variety of autoinflammatory and autoimmune features during early childhood; however, cases with neonatal onset are rare. Here, we describe a Chinese neonate presenting with concomitant inflammatory and other syndromic manifestations caused by a 5.15 Mb interstitial deletion in chromosome 6; these deletions affect TNFAIP3. Taken together, the data extend the clinical and genetic spectra of HA20.
    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
    创伤性脑损伤(TBI),特别是当中度到重度和重复性时,是几种进行性神经退行性疾病的强烈环境风险因素。已经报道了在TBI和TBI相关的神经退行性疾病中的反式反应DNA结合蛋白43(TDP-43)的错误定位和沉积。有人假设轴突病理学,TBI后的早期事件,可能促进TDP-43失调,并作为神经退行性过程的触发因素。我们试图确定阻断前变性Sarm1(无菌α和含有1个TIR基序)轴突死亡途径是否会减弱TBI后的TDP-43病理。我们对111个雄性Sarm1野生型,半合子,和敲除小鼠中度至重度重复性TBI(rTBI)使用先前建立的损伤范例。我们进行了连续的神经系统评估,然后进行了组织学分析(NeuN,MBP,Iba-1,GFAP,rTBI后1个月,pTDP-43和AT8)。Sarm1基因的遗传消融减弱了磷酸化TDP-43(pTDP-43)的表达和错位以及pTau的积累。此外,Sarm1基因敲除小鼠皮质神经元和轴突完整性显著改善,功能缺陷,并改善rTBI后的总生存率。相比之下,去除一个延迟的Sarm1等位基因,但并没有阻止,神经功能缺损和神经轴突丢失。然而,Sarm1单倍体不足的小鼠显示出较少的小胶质细胞增生,pTDP-43病理学,和与野生型小鼠相比时的pTau积累。这些数据表明,Sarm1介导的前变性途径有助于rTBI的发病机理,包括pTDP-43的病理性积累。这表明抗Sarm1疗法是在中度至重度rTBI后保持神经功能的可行方法。
    Traumatic brain injury (TBI), particularly when moderate-to-severe and repetitive, is a strong environmental risk factor for several progressive neurodegenerative disorders. Mislocalization and deposition of transactive response DNA binding protein 43 (TDP-43) has been reported in both TBI and TBI-associated neurodegenerative diseases. It has been hypothesized that axonal pathology, an early event after TBI, may promote TDP-43 dysregulation and serve as a trigger for neurodegenerative processes. We sought to determine whether blocking the prodegenerative Sarm1 (sterile alpha and TIR motif containing 1) axon death pathway attenuates TDP-43 pathology after TBI. We subjected 111 male Sarm1 wild type, hemizygous, and knockout mice to moderate-to-severe repetitive TBI (rTBI) using a previously established injury paradigm. We conducted serial neurological assessments followed by histological analyses (NeuN, MBP, Iba-1, GFAP, pTDP-43, and AT8) at 1 month after rTBI. Genetic ablation of the Sarm1 gene attenuated the expression and mislocalization of phosphorylated TDP-43 (pTDP-43) and accumulation of pTau. In addition, Sarm1 knockout mice had significantly improved cortical neuronal and axonal integrity, functional deficits, and improved overall survival after rTBI. In contrast, removal of one Sarm1 allele delayed, but did not prevent, neurological deficits and neuroaxonal loss. Nevertheless, Sarm1 haploinsufficient mice showed significantly less microgliosis, pTDP-43 pathology, and pTau accumulation when compared to wild type mice. These data indicate that the Sarm1-mediated prodegenerative pathway contributes to pathogenesis in rTBI including the pathological accumulation of pTDP-43. This suggests that anti-Sarm1 therapeutics are a viable approach for preserving neurological function after moderate-to-severe rTBI.
    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
    不孕症影响全世界的夫妇。过早卵巢功能不全(POI)是指40岁之前卵巢功能丧失,是导致不孕的一个因素。一些案例研究报道了具有杂合EIF4ENIF1(4E-T)突变的家庭中显性遗传POI症状。然而,在动物模型中很少研究EIF4ENIF1单倍体功能不全的影响以揭示与不孕症相关的潜在分子变化。这里,我们证明Eif4enif1单倍体不足导致小鼠不育,损害卵母细胞成熟并部分阻止早期胚胎发育。使用双重测序,我们观察到Eif4enif1单倍体不足显著改变小鼠卵母细胞的转录组和翻译组,由此,我们进一步揭示了Eif4enif1缺陷卵母细胞中卵母细胞线粒体过度融合和线粒体相关核糖核蛋白结构域分布的改变。这项研究为临床生育失败的分子机制提供了新的见解,并为寻求新的治疗目标以解决不孕症提供了新的途径。
    Infertility affects couples worldwide. Premature ovarian insufficiency (POI) refers to loss of ovarian function before 40 years of age and is a contributing factor to infertility. Several case studies have reported dominant-inherited POI symptoms in families with heterozygous EIF4ENIF1 (4E-T) mutations. However, the effects of EIF4ENIF1 haploinsufficiency have rarely been studied in animal models to reveal the underlying molecular changes related to infertility. Here, we demonstrate that Eif4enif1 haploinsufficiency causes mouse subfertility, impairs oocyte maturation and partially arrests early embryonic development. Using dual-omic sequencing, we observed that Eif4enif1 haploinsufficiency significantly altered both transcriptome and translatome in mouse oocytes, by which we further revealed oocyte mitochondrial hyperfusion and mitochondria-associated ribonucleoprotein domain distribution alteration in Eif4enif1-deficient oocytes. This study provides new insights into the molecular mechanisms underlying clinical fertility failure and new avenues to pursue new therapeutic targets to address infertility.
    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