中国神经再生研究(英文版) ›› 2023, Vol. 18 ›› Issue (2): 237-243.doi: 10.4103/1673-5374.346540

• 综述:退行性病与再生 •    下一篇

c-Abl 激酶处于神经退行性疾病中健康突触重塑和突触功能障碍的十字路口

  

  • 出版日期:2023-02-15 发布日期:2022-08-05

c-Abl kinase at the crossroads of healthy synaptic remodeling and synaptic dysfunction in neurodegenerative diseases

Daniela A. Gutiérrez1, América Chandía-Cristi1, María José Yáñez2, Silvana Zanlungo3, Alejandra R. Álvarez1, *   

  1. 1Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Center for Aging and Regeneration (CARE), Millennium Institute on Immunology and Immunotherapy, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile; 2School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Sede Los Leones, Santiago, Chile; 3Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
  • Online:2023-02-15 Published:2022-08-05
  • Contact: Alejandra R. Alvarez, PhD, aalvarez@bio.puc.cl.
  • Supported by:
    This work was supported by Comisión Nacional de Investigación Científica y Tecnológica-Chile: Fondecyt 12011668 (to ARA), Fondecyt 1190334 (to SZ), Fondecyt 11200592 (to MJY), Fondef ID21I10347 (to ARA and SZ), Fondef D10E1077 (to ARA and SZ), CARE-UC AFB 170005 (to ARA), MSCA-RISE-2016-Lysomod-734825 European Union’s Horizon 2020 Research and Innovation Program under the Marie Sklodowska-Curie grant agreement N°953489 (to SZ), Millennium Science Initiative Program - ICN09_016 / ICN 2021_045 (to ARA). 

摘要: https://orcid.org/0000-0002-8129-9280 (Alejandra R. Alvarez)

Abstract: Our ability to learn and remember depends on the active formation, remodeling, and elimination of synapses. Thus, the development and growth of synapses as well as their weakening and elimination are essential for neuronal rewiring. The structural reorganization of synaptic complexes, changes in actin cytoskeleton and organelle dynamics, as well as modulation of gene expression, determine synaptic plasticity. It has been proposed that dysregulation of these key synaptic homeostatic processes underlies the synaptic dysfunction observed in many neurodegenerative diseases. Much is known about downstream signaling of activated N-methyl-D-aspartate and α-amino-3-hydroxy-5-methyl-4-isoazolepropionate receptors; however, other signaling pathways can also contribute to synaptic plasticity and long-lasting changes in learning and memory. The non-receptor tyrosine kinase c-Abl (ABL1) is a key signal transducer of intra and extracellular signals, and it shuttles between the cytoplasm and the nucleus. This review focuses on c-Abl and its synaptic and neuronal functions. Here, we discuss the evidence showing that the activation of c-Abl can be detrimental to neurons, promoting the development of neurodegenerative diseases. Nevertheless, c-Abl activity seems to be in a pivotal balance between healthy synaptic plasticity, regulating dendritic spines remodeling and gene expression after cognitive training, and synaptic dysfunction and loss in neurodegenerative diseases. Thus, c-Abl genetic ablation not only improves learning and memory and modulates the brain genetic program of trained mice, but its absence provides dendritic spines resiliency against damage. Therefore, the present review has been designed to elucidate the common links between c-Abl regulation of structural changes that involve the actin cytoskeleton and organelles dynamics, and the transcriptional program activated during synaptic plasticity. By summarizing the recent discoveries on c-Abl functions, we aim to provide an overview of how its inhibition could be a potentially fruitful treatment to improve degenerative outcomes and delay memory loss.

Key words: actin cytoskeleton, activity-dependent plasticity, Alzheimer’s disease, c-Abl, dendritic spines, learning, synapse, synaptic plasticity, transcription, tyrosine kinase