Abstract

IntroductionThe extracellular deposition of β-amyloid (Aβ) is a pathological hallmark in Alzheimer's disease (AD), which induces microglial activation in the pathology of AD. The expression of serine/threonine-protein kinase 2 (SRPK2) is increased in the brain tissues of patients with AD. In this study, we examined the effect of SRPK2 in the activation of microglia.MethodsMicroglia (BV2) cells were cultured and the expression of SRPK2 was enhanced by transfection of SRPK2 recombinant vectors or knockdown by SRPK2 small interfering RNA (siRNA). The cells were stimulated by lipopolysaccharide (LPS) + interferon-γ (IFN-γ) or Aβ in vitro, generating inflammatory cytokines [tumor necrosis factor-α (TNF-α), interleukin (IL)-10, and IL-6], which were investigated by real-time quantitative PCR (qPCR) and ELISA. The proliferation ability of the BV2 cells with/without SRPK2 expression was evaluated by WST-1 under pressure in the presence of Aβ. The effects of SRPK2 on microglia polarization were evaluated by investigating the expression of CD16/32 and CD206 by western blot and the expression of ionized calcium-binding adapter molecule-1 (IBA-1) and arginase-1 (Arg-1) by immunofluorescence. Hippocampal cells HT-22 were cultured with a BV2 cell (with/without SRPK2 expression)-derived medium stimulated by Aβ or LPS + IFN-γ, prior to the evaluation of HT-22 cytotoxicity by assessment of cell viability. Possible relationships between Akt and SRPK2 in the BV2 cells were investigated by western blot.ResultsThe expression of SRPK2 was related to the phenotype polarization changes of microglia with increased expression of CD16/32 and IBA-1. The expression of proinflammatory cytokines IL-6 and TNF-α was increased, whereas the expression of anti-inflammatory cytokine IL-10 was decreased in the BV2 cells with SRPK2 overexpression. Moreover, with the expression enhancement of SRPK2, the BV2 cells had a higher proliferation rate. Aβ treatment can promote SRPK2 expression in BV2 cells. Aβ or LPS + IFN-γ promoted the production of cytokines IL-6 and TNF-α but decreased cytokine IL-10 in the BV2 cells. SRPK2 deficiency alleviated the cytotoxic effects of Aβ or LPS + IFN-γ exposed microglia on HT22 cells. In addition, the activated Akt pathway promoted the expression of SRPK2 in the BV2 cells.ConclusionOur data have found that enhanced SRPK2 expression contributed to the proinflammatory activation of microglia. Thus, SRPK2 may be a key modulating pathway of inflammatory mediators in AD pathology.

Highlights

  • The extracellular deposition of β-amyloid (Aβ) is a pathological hallmark in Alzheimer’s disease (AD), which induces microglial activation in the pathology of AD

  • Morphology analysis showed that enhanced expression of serine/threonine-protein kinase 2 (SRPK2) produced a higher percentage of ameboid microglia than that in the controls while blocking the expression of SRPK2 by small interfering RNA (siRNA) resulted in a decreased percentage of ameboid microglia (Figures 1G,H)

  • This study shows that SRPK2 is involved in microglia-mediated neurotoxicity induced by Aβ

Read more

Summary

Introduction

The extracellular deposition of β-amyloid (Aβ) is a pathological hallmark in Alzheimer’s disease (AD), which induces microglial activation in the pathology of AD. The expression of serine/threonine-protein kinase 2 (SRPK2) is increased in the brain tissues of patients with AD. We examined the effect of SRPK2 in the activation of microglia. Alzheimer’s disease (AD), which is pathologically characterized by brain tissue atrophy, amyloid plaque formation [β-amyloid (Aβ)], neurofibrillary tangles (mainly tau protein) formation, neuronal synapse loss, etc., was first described by the German pathologist Alzheimer in 1906. Neuroinflammation has been identified to play an important role in the development of AD (Bagyinszky et al, 2017). The role of glial cell activation, especially microglia cells, in neuroinflammation has been widely confirmed (Kim et al, 2020). The role of microglia has been identified in multiple neurodegenerative diseases including AD pathogenesis (Maragakis and Rothstein, 2006; Ransohoff and Perry, 2009; Glass et al, 2010)

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call