SMNΔ7

  • 文章类型: Journal Article
    脊髓性肌萎缩症(SMA)是一种由运动神经元存活蛋白(SMN)缺乏引起的神经退行性疾病。虽然SMA是一种遗传性疾病,环境因素有助于疾病进展。常见的病原体成分如脂多糖(LPS)被认为是炎症的重要贡献者,并且与肌肉萎缩有关。这被认为是SMA的标志。在这项研究中,我们使用SMA的SMNΔ7实验小鼠模型来仔细检查全身LPS给药的效果,强烈的促炎刺激,关于疾病的结果。全身性LPS给药促进中枢神经系统中SMN表达水平的降低,外周淋巴器官,和骨骼肌。此外,与CNS组织相比,外周组织更容易受到LPS诱导的损伤.此外,全身LPS给药导致SMNΔ7小鼠CNS中具有反应性表型的小胶质细胞和星形胶质细胞的显着增加。总之,我们在此首次展示全身LPS给药,尽管它可能不会导致SMA小鼠模型中运动功能缺陷的改变,它可能,然而,导致骨骼肌和中枢神经系统中SMN蛋白表达水平降低,从而促进突触损伤和神经胶质细胞反应表型。
    Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by deficiency of the survival motor neuron (SMN) protein. Although SMA is a genetic disease, environmental factors contribute to disease progression. Common pathogen components such as lipopolysaccharides (LPS) are considered significant contributors to inflammation and have been associated with muscle atrophy, which is considered a hallmark of SMA. In this study, we used the SMNΔ7 experimental mouse model of SMA to scrutinize the effect of systemic LPS administration, a strong pro-inflammatory stimulus, on disease outcome. Systemic LPS administration promoted a reduction in SMN expression levels in CNS, peripheral lymphoid organs, and skeletal muscles. Moreover, peripheral tissues were more vulnerable to LPS-induced damage compared to CNS tissues. Furthermore, systemic LPS administration resulted in a profound increase in microglia and astrocytes with reactive phenotypes in the CNS of SMNΔ7 mice. In conclusion, we hereby show for the first time that systemic LPS administration, although it may not precipitate alterations in terms of deficits of motor functions in a mouse model of SMA, it may, however, lead to a reduction in the SMN protein expression levels in the skeletal muscles and the CNS, thus promoting synapse damage and glial cells\' reactive phenotype.
    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:脊髓性肌萎缩(SMA)是一种常染色体隐性遗传的神经肌肉疾病,影响儿童。它是由存活运动神经元1(SMN1)基因突变或缺失引起的,导致下运动神经元(MN)变性,然后是运动障碍。进行性骨骼肌麻痹和呼吸衰竭。除了现有的治疗方法,一种可能的组合策略可以通过使用脂肪来源的间充质干细胞(ASC)来表示,所述间充质干细胞可以容易地从脂肪组织中大量获得。它们的功效似乎与其旁分泌活性和通过细胞外囊泡(EV)释放的可溶性因子的产生有关。EV是细胞间通讯的重要介质,直径在30至100nm之间。由于其内容的释放,它们在其他神经退行性疾病中的使用显示出神经保护作用,尤其是蛋白质,miRNA和mRNA。
    方法:在本研究中,我们评估了从ASCs(ASC-EV)分离的EV在SMNΔ7小鼠中的作用,严重的SMA模型。为了这个目的,我们在出生后第3天(P3)和第6天通过侧脑室注射对SMA幼崽进行了两次ASC-EV(0.5µg)给药.然后,我们通过从P2到P10的行为测试和P10的组织学分析来评估治疗功效。
    结果:结果显示ASC-EV对疾病进展有积极作用,具有改善的运动性能和治疗动物的脊髓MN变性的显著延迟。ASC-EV还可以减少凋亡激活(裂解的Caspase-3)和调节神经炎症与观察到的减少神经胶质激活在腰脊髓,而在外周水平,ASC-EV只能部分限制肌肉萎缩和纤维神经支配。
    结论:我们的结果可以鼓励使用ASC-EV作为SMA的治疗性组合治疗,绕过有争议的干细胞使用。
    BACKGROUND: Spinal Muscular Atrophy (SMA) is an autosomal-recessive neuromuscular disease affecting children. It is caused by the mutation or deletion of the survival motor neuron 1 (SMN1) gene resulting in lower motor neuron (MN) degeneration followed by motor impairment, progressive skeletal muscle paralysis and respiratory failure. In addition to the already existing therapies, a possible combinatorial strategy could be represented by the use of adipose-derived mesenchymal stem cells (ASCs) that can be obtained easily and in large amounts from adipose tissue. Their efficacy seems to be correlated to their paracrine activity and the production of soluble factors released through extracellular vesicles (EVs). EVs are important mediators of intercellular communication with a diameter between 30 and 100 nm. Their use in other neurodegenerative disorders showed a neuroprotective effect thanks to the release of their content, especially proteins, miRNAs and mRNAs.
    METHODS: In this study, we evaluated the effect of EVs isolated from ASCs (ASC-EVs) in the SMNΔ7 mice, a severe SMA model. With this purpose, we performed two administrations of ASC-EVs (0.5 µg) in SMA pups via intracerebroventricular injections at post-natal day 3 (P3) and P6. We then assessed the treatment efficacy by behavioural test from P2 to P10 and histological analyses at P10.
    RESULTS: The results showed positive effects of ASC-EVs on the disease progression, with improved motor performance and a significant delay in spinal MN degeneration of treated animals. ASC-EVs could also reduce the apoptotic activation (cleaved Caspase-3) and modulate the neuroinflammation with an observed decreased glial activation in lumbar spinal cord, while at peripheral level ASC-EVs could only partially limit the muscular atrophy and fiber denervation.
    CONCLUSIONS: Our results could encourage the use of ASC-EVs as a therapeutic combinatorial treatment for SMA, bypassing the controversial use of stem cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号