LRRK2 G2019S

  • 文章类型: Journal Article
    小胶质细胞对于维持大脑健康和神经元功能至关重要。这里,我们报道小胶质细胞在生理和病理条件下使用隧道纳米管(TNTs)与神经元建立联系。这些TNT促进了细胞器的快速交换,囊泡,和蛋白质。在神经退行性疾病如帕金森病和阿尔茨海默病,α-突触核蛋白(α-syn)和tau的毒性聚集体在神经元内积累。我们的研究表明,小胶质细胞使用TNTs从这些聚集体中提取神经元,恢复神经元健康。此外,小胶质细胞与负重的神经元共享其健康的线粒体,减少氧化应激和正常化基因表达。在TNT形成之前用抗霉素A破坏线粒体功能消除了这种神经保护。此外,将神经元与小胶质细胞共培养并促进TNT形成可以挽救由α-syn或tau聚集体引起的神经元活性抑制。值得注意的是,TNT介导的聚集体转移在携带Lrrk22(Gly2019Ser)或Trem2(T66M)和(R47H)突变的小胶质细胞中受损,提示这些基因变异在神经退行性疾病的病理学中的作用。
    Microglia are crucial for maintaining brain health and neuron function. Here, we report that microglia establish connections with neurons using tunneling nanotubes (TNTs) in both physiological and pathological conditions. These TNTs facilitate the rapid exchange of organelles, vesicles, and proteins. In neurodegenerative diseases like Parkinson\'s and Alzheimer\'s disease, toxic aggregates of alpha-synuclein (α-syn) and tau accumulate within neurons. Our research demonstrates that microglia use TNTs to extract neurons from these aggregates, restoring neuronal health. Additionally, microglia share their healthy mitochondria with burdened neurons, reducing oxidative stress and normalizing gene expression. Disrupting mitochondrial function with antimycin A before TNT formation eliminates this neuroprotection. Moreover, co-culturing neurons with microglia and promoting TNT formation rescues suppressed neuronal activity caused by α-syn or tau aggregates. Notably, TNT-mediated aggregate transfer is compromised in microglia carrying Lrrk22(Gly2019Ser) or Trem2(T66M) and (R47H) mutations, suggesting a role in the pathology of these gene variants in neurodegenerative diseases.
    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
    帕金森病(Parkinson’sdisease,PD)是一种进行性迟发性神经退行性疾病,可导致身体和认知功能下降。富含亮氨酸的重复激酶2(LRRK2)的突变是PD的最常见的遗传原因。LRRK2是一种复杂的支架蛋白,在多种分子途径中具有已知的调节作用。LRRK2调节途径的两个突出实例是无翼/Int(Wnt)和活化T细胞的核因子(NFAT)信号传导。两者都被描述为免疫和神经系统发育以及成熟的关键调节因子。本研究的目的是建立LRRK2在脑内Wnt和NFAT信号传导中的生理和致病作用,以及非经典Wnt/钙途径的潜在贡献。在LRRK2G2019S突变体敲入(KI)和LRRK2敲除(KO)雄性和雌性小鼠中定量体内脑Wnt和NFATc1信号传导活性,并在28周内重复测量,采用慢病毒荧光素酶生物传感器,并使用混合效应模型进行分析。要建立空间分辨率,我们调查了组织,和来自不同大脑区域的原代神经元细胞培养物,结合了荧光素酶信号传导活性,免疫组织化学,qPCR和蛋白质印迹测定。结果通过非配对t检验与Welch's校正或双向方差分析与事后校正。LRRK2KO和LRRK2G2019SKI小鼠体内Wnt信号活性显著增加~3倍,对男性的影响(〜四倍)比女性(〜两倍)更明显。在LRRK2G2019SKI小鼠中,NFATc1信号传导降低约0.5倍。脑组织分析显示Wnt和NFAT信号传导成分的区域特异性表达变化。这些作用主要在LRRK2KI小鼠的纹状体和大脑皮层的蛋白质水平上观察到。原代神经元细胞培养分析显示在基础和刺激条件下Wnt和NFATc1信号传导的显著基因型依赖性改变。Wnt和NFATc1信号分别主要在皮质和海马神经元中失调。我们的研究进一步基于LRRK2作为Wnt和NFAT信号蛋白的知识。我们在LRRK2PD的神经元模型中发现了复杂的变化,表明突变体LRRK2在NFAT失调中的作用,以及规范和非规范Wnt信号。
    Parkinson\'s disease (PD) is a progressive late-onset neurodegenerative disease leading to physical and cognitive decline. Mutations of leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of PD. LRRK2 is a complex scaffolding protein with known regulatory roles in multiple molecular pathways. Two prominent examples of LRRK2-modulated pathways are Wingless/Int (Wnt) and nuclear factor of activated T-cells (NFAT) signaling. Both are well described key regulators of immune and nervous system development as well as maturation. The aim of this study was to establish the physiological and pathogenic role of LRRK2 in Wnt and NFAT signaling in the brain, as well as the potential contribution of the non-canonical Wnt/Calcium pathway. In vivo cerebral Wnt and NFATc1 signaling activity was quantified in LRRK2 G2019S mutant knock-in (KI) and LRRK2 knockout (KO) male and female mice with repeated measures over 28 weeks, employing lentiviral luciferase biosensors, and analyzed using a mixed-effect model. To establish spatial resolution, we investigated tissues, and primary neuronal cell cultures from different brain regions combining luciferase signaling activity, immunohistochemistry, qPCR and western blot assays. Results were analyzed by unpaired t-test with Welch\'s correction or 2-way ANOVA with post hoc corrections. In vivo Wnt signaling activity in LRRK2 KO and LRRK2 G2019S KI mice was increased significantly ~ threefold, with a more pronounced effect in males (~ fourfold) than females (~ twofold). NFATc1 signaling was reduced ~ 0.5-fold in LRRK2 G2019S KI mice. Brain tissue analysis showed region-specific expression changes in Wnt and NFAT signaling components. These effects were predominantly observed at the protein level in the striatum and cerebral cortex of LRRK2 KI mice. Primary neuronal cell culture analysis showed significant genotype-dependent alterations in Wnt and NFATc1 signaling under basal and stimulated conditions. Wnt and NFATc1 signaling was primarily dysregulated in cortical and hippocampal neurons respectively. Our study further built on knowledge of LRRK2 as a Wnt and NFAT signaling protein. We identified complex changes in neuronal models of LRRK2 PD, suggesting a role for mutant LRRK2 in the dysregulation of NFAT, and canonical and non-canonical Wnt signaling.
    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
    LRRK2G2019S变异体是单基因帕金森病(PD)的最常见原因;然而,关于外显率的问题仍然存在,临床表型和携带者的自然史。我们在大量1286名基因分型的LRRK2G2019S携带者和109154名对照者中进行了3.5年的前瞻性纵向在线研究,有和没有PD,从23andMe研究队列招募。我们每6个月收集自我报告的运动和非运动症状,以及人口统计,家族史和环境危险因素。在随访中发现了PD(表型转化者)的事件病例。我们使用加速故障时间模型确定了PD的终生风险,并探讨了多基因风险对外显率的影响。我们还计算了23andMe数据库中所有LRRK2G2019S携带者的遗传祖先,并确定了世界上携带者频率最高的地区。我们观察到,尽管疾病持续时间长1年(P=0.016),患有PD的LRRK2G2019S携带者具有相似的运动症状负担,但包括认知障碍在内的非运动症状明显减少,REM睡眠行为障碍(RBD)和睡眠障碍(所有P值≤0.0002)。到80岁时,G2019S携带者中PD的累积发生率为49%。与非携带者相比,G2019S携带者患PD的风险是10倍。这在G2019S携带者中上升到27倍的风险,PD多基因风险评分在前25%,而非携带者在后25%。除了确定北非和阿什肯纳齐人血统的古代建国事件外,我们的遗传祖先分析推断G2019S变体后来被引入美洲的西班牙殖民地.我们的结果表明,LRRK2G2019SPD似乎是一种缓慢进展的主要运动亚型PD,其患病率较低,RBD和认知障碍。这表明当前的前驱标准,基于特发性PD,可能缺乏检测G2019S载波中LRRK2PD早期阶段的敏感性。我们表明,多基因负担可能有助于LRRK2G2019S携带者群体中PD的发展。总的来说,这些结果应有助于支持即将进行的LRRK2抑制剂早期疾病试验的筛查计划和候选富集策略.
    The LRRK2 G2019S variant is the most common cause of monogenic Parkinson\'s disease (PD); however, questions remain regarding the penetrance, clinical phenotype and natural history of carriers. We performed a 3.5-year prospective longitudinal online study in a large number of 1286 genotyped LRRK2 G2019S carriers and 109 154 controls, with and without PD, recruited from the 23andMe Research Cohort. We collected self-reported motor and non-motor symptoms every 6 months, as well as demographics, family histories and environmental risk factors. Incident cases of PD (phenoconverters) were identified at follow-up. We determined lifetime risk of PD using accelerated failure time modelling and explored the impact of polygenic risk on penetrance. We also computed the genetic ancestry of all LRRK2 G2019S carriers in the 23andMe database and identified regions of the world where carrier frequencies are highest. We observed that despite a 1 year longer disease duration (P = 0.016), LRRK2 G2019S carriers with PD had similar burden of motor symptoms, yet significantly fewer non-motor symptoms including cognitive difficulties, REM sleep behaviour disorder (RBD) and hyposmia (all P-values ≤ 0.0002). The cumulative incidence of PD in G2019S carriers by age 80 was 49%. G2019S carriers had a 10-fold risk of developing PD versus non-carriers. This rose to a 27-fold risk in G2019S carriers with a PD polygenic risk score in the top 25% versus non-carriers in the bottom 25%. In addition to identifying ancient founding events in people of North African and Ashkenazi descent, our genetic ancestry analyses infer that the G2019S variant was later introduced to Spanish colonial territories in the Americas. Our results suggest LRRK2 G2019S PD appears to be a slowly progressive predominantly motor subtype of PD with a lower prevalence of hyposmia, RBD and cognitive impairment. This suggests that the current prodromal criteria, which are based on idiopathic PD, may lack sensitivity to detect the early phases of LRRK2 PD in G2019S carriers. We show that polygenic burden may contribute to the development of PD in the LRRK2 G2019S carrier population. Collectively, the results should help support screening programmes and candidate enrichment strategies for upcoming trials of LRRK2 inhibitors in early-stage disease.
    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:脑损伤已被认为是神经退行性疾病的危险因素。因此,大脑内在修复损伤能力的缺陷可能导致损伤的累积和脑功能的进行性丧失。LRRK2(富含亮氨酸重复序列激酶2)中的G2019S(GS)突变是帕金森病(PD)中最普遍的遗传改变。这里,我们试图研究这种LRRK2-GS突变如何影响受损大脑的修复.
    方法:立体定位注射ATP致脑损伤,与损伤相关的分子模式(DAMP)组件,进入野生型(WT)和LRRK2-GS小鼠的纹状体。LRRK2-GS突变对脑损伤和损伤恢复的影响通过分析神经元的分子和细胞行为来检测,星形胶质细胞,和单核细胞。
    结果:受损的神经元表达骨桥蛋白(OPN),与大脑修复有关的因素。ATP诱导的损伤后,单核细胞进入受伤的大脑,通过激活炎性体和随后的焦亡,吞噬受损的神经元并产生含有OPN的外泌体样囊泡(EV)。电动汽车生产后,神经元和星形胶质细胞过程向受伤的核心延伸。在LRRK2-GS小鼠中,与WT小鼠相比,OPN表达和单核细胞焦凋亡降低,导致含OPN的EV释放减少,神经元和星形胶质细胞过程的伸长减弱。此外,从受伤的LRRK2-GS大脑制备的外泌体诱导神经突生长的效率低于受伤的WT大脑。
    结论:LRRK2-GS突变通过减少OPN的表达和减少单核细胞释放含OPN的EV来延迟受损大脑的修复。这些发现表明,LRRK2-GS突变可能通过延迟脑损伤的修复来促进PD的发展。
    BACKGROUND: Brain injury has been suggested as a risk factor for neurodegenerative diseases. Accordingly, defects in the brain\'s intrinsic capacity to repair injury may result in the accumulation of damage and a progressive loss of brain function. The G2019S (GS) mutation in LRRK2 (leucine rich repeat kinase 2) is the most prevalent genetic alteration in Parkinson\'s disease (PD). Here, we sought to investigate how this LRRK2-GS mutation affects repair of the injured brain.
    METHODS: Brain injury was induced by stereotaxic injection of ATP, a damage-associated molecular pattern (DAMP) component, into the striatum of wild-type (WT) and LRRK2-GS mice. Effects of the LRRK2-GS mutation on brain injury and the recovery from injury were examined by analyzing the molecular and cellular behavior of neurons, astrocytes, and monocytes.
    RESULTS: Damaged neurons express osteopontin (OPN), a factor associated with brain repair. Following ATP-induced damage, monocytes entered injured brains, phagocytosing damaged neurons and producing exosome-like vesicles (EVs) containing OPN through activation of the inflammasome and subsequent pyroptosis. Following EV production, neurons and astrocytes processes elongated towards injured cores. In LRRK2-GS mice, OPN expression and monocytic pyroptosis were decreased compared with that in WT mice, resulting in diminished release of OPN-containing EVs and attenuated elongation of neuron and astrocyte processes. In addition, exosomes prepared from injured LRRK2-GS brains induced neurite outgrowth less efficiently than those from injured WT brains.
    CONCLUSIONS: The LRRK2-GS mutation delays repair of injured brains through reduced expression of OPN and diminished release of OPN-containing EVs from monocytes. These findings suggest that the LRRK2-GS mutation may promote the development of PD by delaying the repair of brain injury.
    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
    富含亮氨酸的重复激酶2(LRRK2)是属于ROCO蛋白家族的丝氨酸-苏氨酸蛋白激酶。在LRRK2的激酶域中,称为LRRK2G2019S的点突变已成为与帕金森氏病相关的最普遍的变体。最近的临床研究表明,G2019S携带者患癌症的风险较高,包括结肠癌.尽管有这样的观察,LRRK2G2019S与结肠癌相关的潜在机制仍然难以捉摸.在这项研究中,采用结肠炎相关癌症(CAC)模型和LRRK2G2019S敲入(KI)小鼠模型,我们证明LRRK2G2019S促进结肠癌的发病机制,以KI小鼠肿瘤数量和大小增加为特征。此外,LRRK2G2019S增强肿瘤微环境内的肠上皮细胞增殖和炎症。机械上,KI小鼠对DSS诱导的结肠炎表现出更高的易感性,抑制LRRK2激酶活性改善结肠炎严重程度和CAC进展。我们的研究还显示,在结肠炎模型中,LRRK2G2019S促进炎症小体激活并加剧肠上皮坏死。值得注意的是,GSDMD抑制剂减轻LRRK2G2019SKI小鼠的结肠炎。一起来看,我们的发现提供了实验证据,表明LRRK2中激酶活性的增加促进了结直肠肿瘤发生,表明LRRK2是结肠癌患者表现出高LRRK2激酶活性的潜在治疗靶标。
    Leucine-rich repeat kinase 2 (LRRK2) is a serine-threonine protein kinase belonging to the ROCO protein family. Within the kinase domain of LRRK2, a point mutation known as LRRK2 G2019S has emerged as the most prevalent variant associated with Parkinson\'s disease. Recent clinical studies have indicated that G2019S carriers have an elevated risk of cancers, including colon cancer. Despite this observation, the underlying mechanisms linking LRRK2 G2019S to colon cancer remain elusive. In this study, employing a colitis-associated cancer (CAC) model and LRRK2 G2019S knock-in (KI) mouse model, we demonstrate that LRRK2 G2019S promotes the pathogenesis of colon cancer, characterized by increased tumor number and size in KI mice. Furthermore, LRRK2 G2019S enhances intestinal epithelial cell proliferation and inflammation within the tumor microenvironment. Mechanistically, KI mice exhibit heightened susceptibility to DSS-induced colitis, with inhibition of LRRK2 kinase activity ameliorating colitis severity and CAC progression. Our investigation also reveals that LRRK2 G2019S promotes inflammasome activation and exacerbates gut epithelium necrosis in the colitis model. Notably, GSDMD inhibitors attenuate colitis in LRRK2 G2019S KI mice. Taken together, our findings offer experimental evidence indicating that the gain-of-kinase activity in LRRK2 promotes colorectal tumorigenesis, suggesting LRRK2 as a potential therapeutic target in colon cancer patients exhibiting hyper LRRK2 kinase activity.
    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
    多重假说认为帕金森病(PD)的发病机制需要遗传、环境和老龄化。富含亮氨酸的重复激酶2(LRRK2)的突变被认为是PD的危险因素,与散发性和家族性PD密切相关,可以调节多种细胞通路和过程。越来越多的研究表明,神经胶质过度激活介导的神经炎症参与了多巴胺能神经元的变性和PD的发病机制。LRRK2对慢性炎症的调节至关重要,尤其是中枢神经系统。综述了LRRK2在小胶质细胞和星形胶质细胞上的新发展,探索他们潜在的治疗靶点,以便在PD中提供新的见解。关键信息:关于本主题的已知内容LRRK2的G2019S突变现在被认为是PD中的致病性突变。以往的研讨集中在神经元与LRRK2G2019S之间的关系。本研究增加了神经胶质细胞LRRK2G2019S介导的神经炎症影响PD的进展和发展,必须注意LRRK2G2019S在PD中神经胶质细胞中的作用。这项研究如何影响研究,实践或政策从神经胶质细胞LRRK2G2019S的角度开发抗炎药物是治疗PD的新方向。
    The multiple hypothesis holds that the pathogenesis of Parkinson\'s disease (PD) requires many factors such as heredity, environment and ageing. Mutations in Leucine-rich repeat kinase 2 (LRRK2) are recognized the risk factors of PD, and closely related to sporadic and familial PD and can regulate a variety of cellular pathways and processes. An Increasing number of studies has shown that glial hyperactivation-mediated neuroinflammation participates in dopaminergic neuron degeneration and pathogenesis of PD. LRRK2 is essential to the regulaton of chronic inflammation, especially for the central nervous system. The review spotlights on the novel development of LRRK2 on microglia and astrocytes, and explore their potential therapeutic targets, in order to provide a new insights in PD. Key messages: What is already known on this topic The G2019S mutation of LRRK2 is now recognised as a pathogenic mutation in PD. Previous studies have focused on the relationship between neurons and LRRK2 G2019S. What this study adds Neuroinflammation mediated by LRRK2 G2019S of glial cells affects the progress and development of PD and attention must be paid to the role of LRRK2 G2019S in glial cells in PD. How this study might affect research, practice or policy Developing anti-inflammatory drugs from the perspective of LRRK2 G2019S of glial cells is a new direction for the treatment of 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
    编码富含亮氨酸的重复序列激酶2(LRRK2)的LRRK2的基因突变,与帕金森病(PD)最常见的单基因形式之一有关。然而,Gly2019Ser(G2019S)突变的潜在效应子仍然未知.在这项研究中,作者研究了LRRK2G2019S对诱导多能干细胞(iPSC)诱导的多巴胺神经元内质网(ER)应激的影响,并探索了小鼠模型的潜在治疗靶点。这些发现表明LRRK2G2019S显著促进神经元和小鼠的ER应激。有趣的是,抑制LRRK2活性可以改善突变引起的内质网应激。此外,LRRK2突变可通过与血小板反应蛋白-1/转化生长因子β1(THBS1/TGF-β1)直接相互作用来诱导内质网应激。抑制LRRK2激酶活性可有效抑制内质网应激和THBS1/TGF-β1的表达。敲除THBS1可以通过与TGF-β1和LRRK2突变引起的行为负担相互作用来挽救ER应激,而抑制TGF-β1具有类似的效果。总的来说,已证明LRRK2突变通过直接与THBS1/TGF-β1相互作用来促进ER应激,从而导致PD中的神经死亡。这些发现为PD的发病机理提供了有价值的见解,突出潜在的诊断标志物和治疗靶点。
    The gene mutations of LRRK2, which encodes leucine-rich repeat kinase 2 (LRRK2), are associated with one of the most prevalent monogenic forms of Parkinson\'s disease (PD). However, the potential effectors of the Gly2019Ser (G2019S) mutation remain unknown. In this study, the authors investigate the effects of LRRK2 G2019S on endoplasmic reticulum (ER) stress in induced pluripotent stem cell (iPSC)-induced dopamine neurons and explore potential therapeutic targets in mice model. These findings demonstrate that LRRK2 G2019S significantly promotes ER stress in neurons and mice. Interestingly, inhibiting LRRK2 activity can ameliorate ER stress induced by the mutation. Moreover, LRRK2 mutation can induce ER stress by directly interacting with thrombospondin-1/transforming growth factor beta1 (THBS1/TGF-β1). Inhibition of LRRK2 kinase activity can effectively suppress ER stress and the expression of THBS1/TGF-β1. Knocking down THBS1 can rescue ER stress by interacting with TGF-β1 and behavior burden caused by the LRRK2 mutation, while suppression of TGF-β1 has a similar effect. Overall, it is demonstrated that the LRRK2 mutation promotes ER stress by directly interacting with THBS1/TGF-β1, leading to neural death in PD. These findings provide valuable insights into the pathogenesis of PD, highlighting potential diagnostic markers and therapeutic targets.
    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
    富含亮氨酸重复序列激酶2(LRRK2)基因突变是一种与帕金森病(PD)相关的常染色体显性突变。在LRRK2基因突变中,LRRK2G2019S突变经常参与PD的发病。目前,多种基因校正工具,如锌指核酸酶(ZFN),辅助依赖性腺病毒载体(HDAdV),基于细菌人工染色体的同源重组(基于BAC的HR)系统,和CRISPR/Cas9同源定向修复(HDR)或腺嘌呤碱基编辑器(ABE)用于基因组编辑。每当新的基因治疗工具出现时,LRRK2G2019S突变的基因校正已被应用,主要应用于诱导多能干细胞(LRRK2G2019S-突变体iPSCs)。这里,我们全面介绍了ZFN等每种可编程核酸酶的原理和方法,CRISPR/Cas9-HDR或ABE应用于LRRK2G2019S,以及用作非可编程核酸酶系统的基于HDAdV或BAC的HR系统。
    Leucine-rich repeat kinase 2 (LRRK2) gene mutation is an autosomal dominant mutation associated with Parkinson\'s disease (PD). Among LRRK2 gene mutations, the LRRK2 G2019S mutation is frequently involved in PD onset. Currently, diverse gene correction tools such as zinc finger nucleases (ZFNs), helper-dependent adenoviral vector (HDAdV), the bacterial artificial chromosome-based homologous recombination (BAC-based HR) system, and CRISPR/Cas9-homology-directed repair (HDR) or adenine base editor (ABE) are used in genome editing. Gene correction of the LRRK2 G2019S mutation has been applied whenever new gene therapy tools emerge, being mainly applied to induced pluripotent stem cells (LRRK2 G2019S-mutant iPSCs). Here, we comprehensively introduce the principles and methods of each programmable nuclease such as ZFN, CRISPR/Cas9-HDR or ABE applied to LRRK2 G2019S, as well as those of HDAdV or BAC-based HR systems used as nonprogrammable nuclease systems.
    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
    Mutations in the leucine-rich repeat kinase 2 (LRRK2) are the most common genetic determinants of Parkinson\'s disease (PD), with the G2019S accounting for about 3% of PD cases. LRRK2 regulates various cellular processes, including vesicle trafficking that is crucial for receptor localization at the plasma membrane. In this study, induced pluripotent stem cells derived from 2 PD patients bearing the G2019S LRRK2 kinase activating mutation were used to generate neuronal cultures enriched in dopaminergic neurons. The results show that mutant LRRK2 prevents the membrane localization of both the dopamine D3 receptors (D3R) and the nicotinic acetylcholine receptors (nAChR) and the formation of the D3R-nAChR heteromer, a molecular unit crucial for promoting neuronal homeostasis and preserving dopaminergic neuron health. Interestingly, D3R and nAChR as well as the corresponding heteromer membrane localization were rescued by inhibiting the abnormally increased kinase activity. Thus, the altered membrane localization of the D3R-nAChR heteromer associated with mutation in LRRK2 might represent a pre-degenerative feature of dopaminergic neurons contributing to the special vulnerability of this neuronal population.
    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: Journal Article
    帕金森病(PD)的诊断是在运动症状发生后开始的,比如静止的震颤,刚性,和运动迟缓.根据以前的报道,非运动症状,尤其是胃肠功能紊乱,可能是PD患者的早期生物标志物,因为这些症状比运动症状更早出现。然而,将PD连接到肠道在方法上具有挑战性。因此,我们从PD患者体内体外产生人肠器官,并从老年转基因小鼠体内产生小鼠小肠器官。两种肠道类器官(IOs)都含有人类LRRK2G2019S突变,这是家族性和散发性PD最常见的遗传原因。通过对这两种类型的IO进行全面的基因组比较,我们确定了一个特定的基因,即,易洛魁同源异型盒蛋白2(IRX2),不仅在人多能干细胞(PSC)衍生的神经外胚层球中,而且在含有多巴胺能神经元的人PSC衍生的神经元细胞中显示了PD相关的表达模式。我们期望我们使用各种细胞类型的方法为研究多器官在PD病理生理学中的作用以及开发PD诊断标志物提供了一种新颖的技术方法。
    The diagnosis of Parkinson\'s disease (PD) is initiated after the occurrence of motor symptoms, such as resting tremors, rigidity, and bradykinesia. According to previous reports, non-motor symptoms, notably gastrointestinal dysfunction, could potentially be early biomarkers in PD patients as such symptoms occur earlier than motor symptoms. However, connecting PD to the intestine is methodologically challenging. Thus, we generated in vitro human intestinal organoids from PD patients and ex vivo mouse small intestinal organoids from aged transgenic mice. Both intestinal organoids (IOs) contained the human LRRK2 G2019S mutation, which is the most frequent genetic cause of familial and sporadic PD. By conducting comprehensive genomic comparisons with these two types of IOs, we determined that a particular gene, namely, Iroquois homeobox protein 2 (IRX2), showed PD-related expression patterns not only in human pluripotent stem cell (PSC)-derived neuroectodermal spheres but also in human PSC-derived neuronal cells containing dopaminergic neurons. We expected that our approach of using various cell types presented a novel technical method for studying the effects of multi-organs in PD pathophysiology as well as for the development of diagnostic markers for PD.
    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