genetic modifiers

遗传修饰剂
  • 文章类型: Journal Article
    镰状细胞病(SCD)在临床上表现为多种并发症。Stroke,缺血性和出血性,以及无声的白质变化,发生在相对较高的患病率。了解中风最可能发生的原因和原因对于有效预防和治疗SCD患者至关重要。基因研究,包括全基因组和外显子组关联研究(GWAS和EWAS),已经发现了几个关键的修饰物,这些修饰物通过包括血红蛋白F(HbF)调节在内的机制与SCD中的卒中/卒中风险增加相关,炎症,细胞粘附,内皮破裂,和溶血。我们对迄今为止最清楚地证明了关联的修饰语进行了综述。需要更多的研究来验证其他潜在的多态性并识别新的多态性。在临床护理中纳入以基因为中心的筛查可以提供更有针对性的途径,更有效,在这一人群中预防中风的毒性较低。本综述的数据将用于告知国际血红蛋白病研究网络(INHERENT)联盟进行的初始GWAS。
    Sickle cell disease (SCD) clinically manifests itself with a myriad of complications. Stroke, both ischemic and hemorrhagic, as well as silent white matter changes, occurs at a relatively high prevalence. Understanding why and in whom stroke is most likely to occur is critical to the effective prevention and treatment of individuals with SCD. Genetic studies, including genome- and exome-wide association studies (GWAS and EWAS), have found several key modifiers associated with increased stroke/stroke risk in SCD via mechanisms including Hemoglobin F (HbF) modulation, inflammation, cellular adhesion, endothelial disruption, and hemolysis. We present a review on the modifiers that have most clearly demonstrated an association to date. More studies are needed to validate other potential polymorphisms and identify new ones. Incorporating gene-focused screenings in clinical care could provide avenues for more targeted, more effective, and less toxic prevention of stroke in this population. The data from this review will be used to inform the initial GWAS performed by the International Hemoglobinopathy Research Network (INHERENT) consortium.
    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:认知功能减退是帕金森病(PD)最常见的非运动症状,而其生理机制仍然知之甚少。遗传因素是PD患者认知下降异质性的基本决定因素。然而,潜在的遗传背景研究仍然较少。
    方法:为了探索导致PD认知能力下降的遗传决定因素,我们使用Cox比例风险模型在450名中国PD患者的纵向队列中进行了全基因组生存分析,并进一步探讨了目标变体的功能效应。此外,我们通过纳入临床特征和多基因风险评分(PRS),构建了一个临床-遗传模型,以预测PD的认知功能下降.
    结果:对该队列进行了平均5.25(SE=2.46)年的随访,95例认知障碍事件。我们确定了rs75819919(DPP6)基因座与加速认知下降之间的显着关联(P=8.63E-09,β=1.74,SE=0.30)。双荧光素酶报告基因分析提示该基因座可能参与DPP6表达的调节。使用英国生物银行的数据集,我们发现rs75819919与一般人群的认知能力相关.PRS的并入增加了模型的可预测性,在1,000次迭代中通过5倍交叉验证实现75.6%的平均AUC。
    结论:这些发现提高了目前对PD认知障碍遗传病因的认识,并提供了一个新的靶点DPP6来探索治疗方案。我们的结果还证明了开发临床遗传模型以识别易患认知障碍的患者的潜力,从而提供个性化的临床指导。
    BACKGROUND: Cognitive decline is among the most common non-motor symptoms in Parkinson\'s disease (PD), while its physiological mechanisms remain poorly understood. Genetic factors constituted a fundamental determinant in the heterogeneity of cognitive decline among PD patients. However, the underlying genetic background was still less studied.
    METHODS: To explore the genetic determinants contributing to cognitive decline in PD, we performed genome-wide survival analysis using a Cox proportional hazards model in a longitudinal cohort of 450 Chinese patients with PD, and further explored the functional effect of the target variant. Additionally, we built a clinical-genetic model by incorporating clinical characteristics and polygenic risk score (PRS) to predict cognitive decline in PD.
    RESULTS: The cohort was followed up for an average of 5.25 (SE = 2.46) years, with 95 incidents of cognitive impairment. We identified significant association between locus rs75819919 (DPP6) and accelerated cognitive decline (p = 8.63E-09, beta = 1.74, SE = 0.30). Dual-luciferase reporter assay suggested this locus might be involved in the regulation of DPP6 expression. Using data set from the UK Biobank, we identified rs75819919 was associated with cognitive performance in the general population. Incorporation of PRS increased the model\'s predictability, achieving an average AUC of 75.6% through fivefold cross-validation in 1 000 iterations.
    CONCLUSIONS: These findings improve the current understanding of the genetic etiology of cognitive impairment in PD, and provide a novel target DPP6 to explore therapeutic options. Our results also demonstrate the potential to develop clinical-genetic model to identify patients susceptible to cognitive impairment and thus provide personalized clinical guidance.
    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
    X连锁肌张力障碍-帕金森病(XDP)是一种神经退行性运动障碍,主要影响成年菲律宾男性。它是由包含六核苷酸重复的TAF1中的创始人逆转录转座子插入引起的,其数量在患者中不同,并与发病年龄(AAO)和其他临床参数相关.最近的一项工作已经确定了XDP中与年龄相关的外显率的其他遗传修饰因子,照亮DNA错配修复基因MSH3和PMS2。尽管有X连锁隐性遗传,一小部分患者是女性,通过纯合性等各种机制表现疾病,不平衡的X染色体失活,或者非整倍性。这里,我们总结和讨论XDP的临床和遗传方面,由于看似同质的患者群体中细微的遗传差异,因此关注可变的疾病表达能力。
    X-linked dystonia-parkinsonism (XDP) is a neurodegenerative movement disorder that primarily affects adult Filipino men. It is caused by a founder retrotransposon insertion in TAF1 that contains a hexanucleotide repeat, the number of which differs among the patients and correlates with the age at disease onset (AAO) and other clinical parameters. A recent work has identified additional genetic modifiers of age-associated penetrance in XDP, bringing to light the DNA mismatch repair genes MSH3 and PMS2. Despite X-linked recessive inheritance, a minor subset of patients are female, manifesting the disease via various mechanisms such as homozygosity, imbalanced X-chromosome inactivation, or aneuploidy. Here, we summarize and discuss clinical and genetic aspects of XDP, with a focus on variable disease expressivity as a consequence of subtle genetic differences within a seemingly homogenous population of 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
    富含亮氨酸的重复激酶2(LRRK2)的突变是显性遗传性帕金森病(PD)的最常见原因。LRRK2突变,其中P.G2019S是最常见的,以降低的外显率遗传。有趣的是,与LRRK2G2019S相关的疾病风险可根据携带者的种族背景而显著变化.虽然这表明LRRK2-PD外显率定义中存在遗传成分,只有少数变异被证明能改变LRRK2突变患者的发病年龄,控制从健康状态到疾病状态转变的确切细胞途径目前仍然难以捉摸。鉴于这种知识差距,最近的研究还探索了环境和生活方式因素作为LRRK2-PD的潜在修饰剂。在这篇文章中,我们(i)描述了LRRK2突变携带者的临床特征,(ii)回顾与LRRK2-PD发病相关的已知基因,(iii)总结LRRK2的细胞功能,特别强调潜在的外显率相关分子机制。本节涵盖LRRK2参与RabGTP酶和免疫信号以及线粒体稳态和动力学的调节。此外,我们探索了有关(iv)生活方式和(v)可能影响LRRK2突变外显率的环境因素的文献,以期进一步进行暴露组学研究。最后,基于这个全面的概述,我们提出了体内潜在的未来,体外和计算机模拟研究可以更好地了解LRRK2突变个体中触发PD的过程。
    Mutations in Leucine-rich repeat kinase 2 (LRRK2) are the most frequent cause of dominantly inherited Parkinson\'s disease (PD). LRRK2 mutations, among which p.G2019S is the most frequent, are inherited with reduced penetrance. Interestingly, the disease risk associated with LRRK2 G2019S can vary dramatically depending on the ethnic background of the carrier. While this would suggest a genetic component in the definition of LRRK2-PD penetrance, only few variants have been shown to modify the age at onset of patients harbouring LRRK2 mutations, and the exact cellular pathways controlling the transition from a healthy to a diseased state currently remain elusive. In light of this knowledge gap, recent studies also explored environmental and lifestyle factors as potential modifiers of LRRK2-PD. In this article, we (i) describe the clinical characteristics of LRRK2 mutation carriers, (ii) review known genes linked to LRRK2-PD onset and (iii) summarize the cellular functions of LRRK2 with particular emphasis on potential penetrance-related molecular mechanisms. This section covers LRRK2\'s involvement in Rab GTPase and immune signalling as well as in the regulation of mitochondrial homeostasis and dynamics. Additionally, we explored the literature with regard to (iv) lifestyle and (v) environmental factors that may influence the penetrance of LRRK2 mutations, with a view towards further exposomics studies. Finally, based on this comprehensive overview, we propose potential future in vivo, in vitro and in silico studies that could provide a better understanding of the processes triggering PD in individuals with LRRK2 mutations.
    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
    运动障碍包括临床上,病理上,与外显率降低相关的遗传异质性疾病组。Penetrance是指当存在特定基因型时发生临床状况的可能性。通过识别可能阻止个体发展疾病的遗传因素,阐明外显率降低的原因可能有助于更个性化的医疗。因此,在这种方法中,病人的材料成为不可替代的资源。首先需要鉴定疾病的遗传修饰剂,然后阐明提供整个遗传背景的内源性人细胞模型中的潜在机制。
    Movement disorders comprise a clinically, pathologically, and genetically heterogeneous group of diseases associated with the phenomenon of reduced penetrance. Penetrance refers to the likelihood that a clinical condition will occur when a particular genotype is present. Elucidating the cause of reduced penetrance may contribute to more personalized medicine by identifying genetic factors that may prevent individuals from developing disease. Therefore, patient material becomes an irreplaceable resource in this approach. It is needed to identify genetic modifiers of the disease in the first place and to subsequently elucidate underlying mechanisms in endogenous human cell models that provide the entire genetic background.
    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
    亨廷顿病(HD)的致病突变,亨廷顿基因(HTT)第一个外显子中扩展的三核苷酸重复序列是天然多态性的,并且不可避免地与39CAG重复以上的疾病症状相关。尽管HD的对症药物治疗可以改善受影响患者的运动和非运动症状,这些药物不能阻止正在进行的神经变性和疾病的进展,导致严重的运动和认知障碍和死亡。迄今为止,仍然迫切需要开发有效的疾病改善疗法来减缓甚至阻止HD的进展.直接干预疾病根源的能力日益增强,即HTT转录和其mRNA的翻译,因此有必要尽可能准确地了解HD的发病机制。除了长期假定的多谷氨酰胺扩增的突变HTT蛋白的毒性外,越来越多的证据表明,含CAG重复序列的RNA也可能直接参与毒性.最近的研究已经确定了顺式(DNA修复基因)和反式(CAA中断的丢失/重复)作用变体作为发病年龄(AO)和疾病进展的主要调节剂。越来越广泛的数据表明,躯体不稳定作为AO的驱动因素以及疾病进展和严重程度,不仅在HD中,而且在其他多聚谷氨酰胺疾病中。因此,重复DNA序列的体细胞扩增对于促进各自的重复长度达到导致三核苷酸疾病的明显神经退行性症状的阈值可能是必不可少的。这些发现支持体细胞扩张作为HD和相关重复扩张障碍的潜在治疗靶标。
    The causative mutation for Huntington disease (HD), an expanded trinucleotide repeat sequence in the first exon of the huntingtin gene (HTT) is naturally polymorphic and inevitably associated with disease symptoms above 39 CAG repeats. Although symptomatic medical therapies for HD can improve the motor and non-motor symptoms for affected patients, these drugs do not stop the ongoing neurodegeneration and progression of the disease, which results in severe motor and cognitive disability and death. To date, there is still an urgent need for the development of effective disease-modifying therapies to slow or even stop the progression of HD. The increasing ability to intervene directly at the roots of the disease, namely HTT transcription and translation of its mRNA, makes it necessary to understand the pathogenesis of HD as precisely as possible. In addition to the long-postulated toxicity of the polyglutamine-expanded mutant HTT protein, there is increasing evidence that the CAG repeat-containing RNA might also be directly involved in toxicity. Recent studies have identified cis- (DNA repair genes) and trans- (loss/duplication of CAA interruption) acting variants as major modifiers of age at onset (AO) and disease progression. More and more extensive data indicate that somatic instability functions as a driver for AO as well as disease progression and severity, not only in HD but also in other polyglutamine diseases. Thus, somatic expansions of repetitive DNA sequences may be essential to promote respective repeat lengths to reach a threshold leading to the overt neurodegenerative symptoms of trinucleotide diseases. These findings support somatic expansion as a potential therapeutic target in HD and related repeat expansion disorders.
    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
    X连锁肌张力障碍-帕金森病(XDP)是一种成人发作的神经退行性运动障碍,由TAF1基因内含子中的创始人逆转录转座子插入引起。该插入包含多态性六核苷酸重复序列(CCCTCT)n,其长度与发病年龄(AAO)和其他临床参数呈负相关,将XDP与重复扩张障碍对齐。然而,可以想象许多其他致病机制在XDP中起作用,表明与其他重复疾病相比,(CCCTCT)n重复可能不是实际(或唯一)的疾病原因。这里,我们总结并讨论了XDP的遗传和分子方面,强调六核苷酸重复序列在年龄相关疾病外显率和表达能力中的作用。
    X-linked dystonia-parkinsonism (XDP) is an adult-onset neurodegenerative movement disorder, caused by a founder retrotransposon insertion in an intron of the TAF1 gene. This insertion contains a polymorphic hexanucleotide repeat (CCCTCT)n, the length of which inversely correlates with the age at disease onset (AAO) and other clinical parameters, aligning XDP with repeat expansion disorders. Nevertheless, many other pathogenic mechanisms are conceivably at play in XDP, indicating that in contrast to other repeat disorders, the (CCCTCT)n repeat may not be the actual (or only) disease cause. Here, we summarize and discuss genetic and molecular aspects of XDP, highlighting the role of the hexanucleotide repeat in age-related disease penetrance and expressivity.
    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
    LMNA基因编码的A型层蛋白中的突变可导致1B型肢带肌营养不良(LGMD1B)。这种疾病表现为近端骨骼肌的虚弱和消瘦,并且具有不同的发病年龄和疾病严重程度。这种变异性归因于个体之间的遗传背景差异;然而,这些变体还没有被很好地表征。为了识别这些变体,我们调查了一个多代家族,其中受影响的个体被诊断为LGMD1B.LGMD1B在该家族中的主要遗传原因是激活隐蔽剪接位点的显性突变,导致成熟mRNA中五个核苷酸的缺失。这导致帧移位和平移的过早停止。家庭成员的骨骼肌活检显示出不同严重程度的营养不良特征,一些家庭成员的肌肉纤维具有核心,肌节中断的区域,线粒体很少,通常不与LGMD1B相关。使用全基因组测序(WGS),我们确定了21种DNA序列变异,这些变异与具有更深刻的营养不良特征和肌核的家族成员分离。这些包括含有卷曲螺旋结构域的蛋白质78(CCDC78)中相对常见的变体。该变体被优先考虑,因为CCDC78中的另一个突变会导致常染色体显性中央核型肌病-4,除了中央定位的细胞核外,还会导致核心。因此,我们分析了家族成员的肌肉活检结果,发现同时具有LMNA突变和CCDC78变异体的家族成员含有同时积累CCDC78和RyR1的肌核.在仅具有LMNA突变的受影响较小的家族成员中,不存在含有错误定位的CCDC78和RyR1的肌核。一起来看,我们的研究结果表明,CCDC78中一个相对常见的变异体可以与LMNA突变相结合,赋予深重的肌肉病理学,并解释骨骼肌疾病表型的变异性.
    Mutations in the LMNA gene-encoding A-type lamins can cause Limb-Girdle muscular dystrophy Type 1B (LGMD1B). This disease presents with weakness and wasting of the proximal skeletal muscles and has a variable age of onset and disease severity. This variability has been attributed to genetic background differences among individuals; however, such variants have not been well characterized. To identify such variants, we investigated a multigeneration family in which affected individuals are diagnosed with LGMD1B. The primary genetic cause of LGMD1B in this family is a dominant mutation that activates a cryptic splice site, leading to a five-nucleotide deletion in the mature mRNA. This results in a frame shift and a premature stop in translation. Skeletal muscle biopsies from the family members showed dystrophic features of variable severity, with the muscle fibers of some family members possessing cores, regions of sarcomeric disruption, and a paucity of mitochondria, not commonly associated with LGMD1B. Using whole genome sequencing (WGS), we identified 21 DNA sequence variants that segregate with the family members possessing more profound dystrophic features and muscle cores. These include a relatively common variant in coiled-coil domain containing protein 78 (CCDC78). This variant was given priority because another mutation in CCDC78 causes autosomal dominant centronuclear myopathy-4, which causes cores in addition to centrally positioned nuclei. Therefore, we analyzed muscle biopsies from family members and discovered that those with both the LMNA mutation and the CCDC78 variant contain muscle cores that accumulated both CCDC78 and RyR1. Muscle cores containing mislocalized CCDC78 and RyR1 were absent in the less profoundly affected family members possessing only the LMNA mutation. Taken together, our findings suggest that a relatively common variant in CCDC78 can impart profound muscle pathology in combination with a LMNA mutation and accounts for variability in skeletal muscle disease phenotypes.
    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
    Niemann-Pick疾病C1型(NPC1)是一种溶酶体疾病,原因是细胞内胆固醇转运出内溶酶体区室。.在具有相同NPC1基因型的个体中观察到明显的异质性,因此表明修饰基因的显着影响。先前的工作表明,在NPC1小鼠模型中,SOAT1活性的降低降低了疾病的严重程度。因此,我们假设与SOAT1表达降低相关的多态性可能影响NPC1表型.作为自然历史试验的一部分,对117名NPC1个体进行了表型分析和基因组测序。表型包括确定疾病严重程度和疾病负担。显著的临床异质性存在于NPC1I1061T变体纯合的个体和兄弟姐妹中。SOAT1多态性分析,rs1044925(A>C),显示C等位基因与神经系统发病的较早年龄显着相关。C等位基因可能与较高的年化严重度指数评分以及肝病和癫痫发作的频率增加有关。与SOAT1表达降低相关的多态性似乎是NPC1表型的遗传修饰。该发现与显示Npc1-/-:Soat1-/-小鼠中表型严重性降低的先前数据一致,并且支持研究SOAT1抑制剂作为NPC1的潜在疗法的潜力的努力。
    Niemann-Pick disease type C1 (NPC1) is a lysosomal disorder due to impaired intracellular cholesterol transport out of the endolysosomal compartment.. Marked heterogeneity has been observed in individuals with the same NPC1 genotype, thus suggesting a significant effect of modifier genes. Prior work demonstrated that decreased SOAT1 activity decreased disease severity in an NPC1 mouse model. Thus, we hypothesized that a polymorphism associated with decreased SOAT1 expression might influence the NPC1 phenotype. Phenotyping and genomic sequencing of 117 individuals with NPC1 was performed as part of a Natural History trial. Phenotyping included determination of disease severity and disease burden. Significant clinical heterogeneity is present in individuals homozygous for the NPC1I1061T variant and in siblings. Analysis of the SOAT1 polymorphism, rs1044925 (A>C), showed a significant association of the C-allele with earlier age of neurological onset. The C-allele may be associated with a higher Annualized Severity Index Score as well as increased frequency of liver disease and seizures. A polymorphism associated with decreased expression of SOAT1 appears to be a genetic modifier of the NPC1 phenotype. This finding is consistent with prior data showing decreased phenotypic severity in Npc1-/-:Soat1-/- mice and supports efforts to investigate the potential of SOAT1 inhibitors as a potential therapy for NPC1.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    Introduction. During the development of the SARS-CoV-2 pandemic in Antioquia, we experienced epidemiological peaks related to the α, ɣ, β, ƛ, and δ variants. δ had the highest incidence and prevalence. This lineage is of concern due to its clinical manifestations and epidemiological characteristics. A total of 253 δ sublineages have been reported in the PANGOLIN database. The sublineage identification through genomic analysis has made it possible to trace their evolution and propagation. Objective. To characterize the genetic diversity of the different SARS-CoV-2 δ sublineages in Antioquia and to describe its prevalence. Materials and methods. We collected sociodemographic information from 2,675 samples, and obtained 1,115 genomes from the GISAID database between July 12th, 2021, and January 18th, 2022. From the analyzed genomes, 515 were selected because of their high coverage values (>90%) to perform phylogenetic analysis and to infer allele frequencies of mutations of interest. Results. We characterized 24 sublineages. The most prevalent was AY.25. Mutations of interest as L452R, P681R, and P681H were identified in this sublineage, comprising a frequency close to 0.99. Conclusions. This study identified that the AY.25 sublineage has a transmission advantage compared to the other δ sublineages. This attribute may be related to the presence of the L452R and P681R mutations associated in other studies with higher evasion of the immune system and less efficacy of drugs against SARS-CoV-2.
    Introducción. Durante el desarrollo de la pandemia por SARS-CoV-2 en Antioquia se presentaron picos epidemiológicos relacionados con las variantes α, ɣ, β, ƛ y δ, donde δ tuvo la mayor incidencia y prevalencia. Este linaje se considera una variante de preocupación dadas las manifestaciones clínicas que desencadena y sus características epidemiológicas. Se han informado 253 sublinajes δ en la base de datos PANGOLIN. La identificación de estos sublinajes mediante análisis genómico ha permitido rastrear su evolución y propagación. Objetivo. Caracterizar la diversidad genética de los diferentes sublinajes δ de SARSCoV-2 en Antioquia y determinar su prevalencia. Materiales y métodos. Se recopiló información sociodemográfica de 2.675 muestras y de 1.115 genomas del repositorio GISAID entre el 12 de julio de 2021 y el 18 de enero de 2022. Se seleccionaron 501 por su alto porcentaje de cobertura (>90 %) para realizar análisis filogenéticos e inferencia de frecuencias alélicas de mutaciones de interés. Resultados. Se caracterizaron 24 sublinajes donde el más prevalente fue AY.25. En este sublinaje se identificaron mutaciones de interés como L452R, P681R y P681H, que comprendían una frecuencia cercana a 0,99. Conclusiones. Este estudio permitió identificar que el sublinaje AY.25 tiene una ventaja de transmisión en comparación con los otros sublinajes δ. Esto puede estar relacionado con la presencia de las mutaciones L452R y P681R que en otros estudios se han visto asociadas con una mayor transmisibilidad, evasión del sistema inmunitario y menor eficacia de los medicamentos contra SARS-CoV-2.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号