中国神经再生研究(英文版) ›› 2024, Vol. 19 ›› Issue (11): 2532-2542.doi: 10.4103/1673-5374.393102

• 原著:神经损伤修复保护与再生 • 上一篇    下一篇

miR-128 通过调节 PPARγ 依赖性通路发挥G9a 抑制的神经保护作用

  

  • 出版日期:2024-11-15 发布日期:2024-03-29
  • 基金资助:
    研究受经济、工业和竞争力部(国家研究机构)和欧洲区域发展基金 (MINECO-FEDER),加泰罗尼亚政府。Secretaria d’Universitats i Recerca del Departament d’Empresai Coneixement de la Generalitat de Catalunya 2021;阿尔茨海默病协会研究奖学金 (AARF-21-848511) ;FI-SDUR 奖学金的支持

Neuroprotective effects of G9a inhibition through modulation of peroxisome-proliferator activator receptor gamma-dependent pathways by miR-128

Aina Bellver-Sanchis1, Pedro A. Ávila-López2, Iva Tic1, David Valle-García3, †, Marta Ribalta-Vilella1, Luis Labrador4, #br# Deb Ranjan Banerjee5, Ana Guerrero1, Gemma Casadesus4, Coralie Poulard6, 7, 8, Mercè Pallàs1, 9, Christian Griñán-Ferré1, 9, *#br#   

  1. 1Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, Barcelona, Spain; 2Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; 3Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca, Mexico; 4Department of Pharmacology and Therapeutics, Health Science Center-University of Florida, Gainesville, FL, USA; 5Department of Chemistry, National Institute of Technology Durgapur, M G Avenue, Durgapur, West Bengal, India; 6Cancer Research Cancer Lyon, Université de Lyon, Lyon, France; 7Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France; 8CNRS UMR5286, Centre de Recherche en Cancérlogie de Lyon, Lyon, France; 9Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
    †Current address: Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City, Mexico
  • Online:2024-11-15 Published:2024-03-29
  • Contact: Christian Griñán-Ferré, PhD, Christian.grinan@ub.edu.
  • Supported by:
    This study was supported by the Ministerio de Economía, Industria y Competitividad (Agencia Estatal de Investigación, AEI; to CGF and MP) and Fondo Europeo de Desarrollo Regional (MINECO-FEDER) (PID2022-139016OA-I00, PDC2022-133441-I00; to CGF and MP), Generalitat de Catalunya (2021 SGR 00357; to CGF and MP). This study was co-financed by Secretaria d’Universitats i Recerca del Departament d’Empresai Coneixement de la Generalitat de Catalunya 2021 (Llavor 00086; to CGF). AG is the recipient of an Alzheimer’s Association Research Fellowship (AARF-21-848511). ABS acknowledges the Agència de Gestió d’Ajuts Universitaris i de Recerca (AGAUR) for her FI-SDUR fellowship (2021FISDU 00182).

摘要:

已有研究证实,阿尔茨海默病中存在组蛋白-赖氨酸 N-甲基转移酶 G9a 的失调,并且与慢性炎症和氧化应激水平的增加相关。同样,microRNAs 也参与了许多生物过程和疾病,在发病机制中发挥着关键作用,尤其是在阿尔茨海默病等多因素疾病中。因此,实验旨在提供有关 G9a,miRNAs,氧化应激和神经炎症之间相互联系的见解。为了更好地了解 G9a 的生物学特性,实验比较了阿尔茨海默病 SAMP8 对照组小鼠和用 G9a 抑制剂 UNC0642 治疗的 SAMP8 小鼠的全局 microRNA 表达。结果发现 G9a 抑制剂治疗后miR-128 表达下调,而 miR-128 与过氧化物酶体-增殖激活剂受体γ(PPARG)mRNA 的 3′非翻译区(3′-UTR)结合。因此,使用 G9a 抑制剂治疗的 SAMP8 组 Pparg 基因表达水平高于 SAMP8 对照组。实验还观察到 G9a 抑制剂对氧化应激反应的调节可能主要是由 Pparg 驱动的。为了证实这些抗氧化作用,实验用过氧化氢作为氧化损伤处理原代神经元细胞培养物。在这种情况下,用 G9a 抑制剂处理可提高细胞存活率和抗氧化酶。此外,G9a 抑制剂对 PPARγ 的上调还能增加 DNA 损伤反应和细胞凋亡相关基因的表达。实验还发现 PPARγ/AMPK 轴可以部分解释自噬标记表达的调控。最后,PPARγ/GADD45α可能有助于增强G9a抑制后的突触可塑性和神经发生。总之,药理抑制 G9a 起到的神经保护效应至少部分是由于 miR-128 对 PPARγ 依赖性通路的调节作用。

https://orcid.org/0000-0002-5424-9130 (Christian Griñán-Ferré)

Abstract: Dysregulation of G9a, a histone-lysine N-methyltransferase, has been observed in Alzheimer’s disease and has been correlated with increased levels of chronic inflammation and oxidative stress. Likewise, microRNAs are involved in many biological processes and diseases playing a key role in pathogenesis, especially in multifactorial diseases such as Alzheimer’s disease. Therefore, our aim has been to provide partial insights into the interconnection between G9a, microRNAs, oxidative stress, and neuroinflammation. To better understand the biology of G9a, we compared the global microRNA expression between senescence-accelerated mouse-prone 8 (SAMP8) control mice and SAMP8 treated with G9a inhibitor UNC0642. We found a downregulation of miR-128 after a G9a inhibition treatment, which interestingly binds to the 3′ untranslated region (3′-UTR) of peroxisome-proliferator activator receptor γ (PPARG) mRNA. Accordingly, Pparg gene expression levels were higher in the SAMP8 group treated with G9a inhibitor than in the SAMP8 control group. We also observed modulation of oxidative stress responses might be mainly driven Pparg after G9a inhibitor. To confirm these antioxidant effects, we treated primary neuron cell cultures with hydrogen peroxide as an oxidative insult. In this setting, treatment with G9a inhibitor increases both cell survival and antioxidant enzymes. Moreover, up-regulation of PPARγ by G9a inhibitor could also increase the expression of genes involved in DNA damage responses and apoptosis. In addition, we also described that the PPARγ/AMPK axis partially explains the regulation of autophagy markers expression. Finally, PPARγ/GADD45α potentially contributes to enhancing synaptic plasticity and neurogenesis after G9a inhibition. Altogether, we propose that pharmacological inhibition of G9a leads to a neuroprotective effect that could be due, at least in part, by the modulation of PPARγ-dependent pathways by miR-128.

Key words: aging, cognitive decline, epigenetics, G9a inhibition, microRNAs, miR-128, peroxisome-proliferator activator receptor γ (PPARγ), PPARG, SAMP8