Abstract

Neuronal miRNA dysregulation may have a role in the pathophysiology of Alzheimer’s disease (AD). miRNA(miR)-124 is largely abundant and a critical player in many neuronal functions. However, the lack of models reliably recapitulating AD pathophysiology hampers our understanding of miR-124’s role in the disease. Using the classical human SH-SY5Y-APP695 Swedish neuroblastoma cells (SH-SWE) and the PSEN1 mutant iPSC-derived neurons (iNEU-PSEN), we observed a sustained upregulation of miR-124/miR-125b/miR-21, but only miR-124 was consistently shuttled into their exosomes. The miR-124 mimic reduced APP gene expression in both AD models. While miR-124 mimic in SH-SWE neurons led to neurite outgrowth, mitochondria activation and small Aβ oligomer reduction, in iNEU-PSEN cells it diminished Tau phosphorylation, whereas miR-124 inhibitor decreased dendritic spine density. In exosomes, cellular transfection with the mimic predominantly downregulated miR-125b/miR-21/miR-146a/miR-155. The miR-124 inhibitor upregulated miR-146a in the two experimental cell models, while it led to distinct miRNA signatures in cells and exosomes. In sum, though miR-124 function may be dependent on the neuronal AD model, data indicate that keeping miR-124 level strictly controlled is crucial for proper neuronal function. Moreover, the iNEU-PSEN cellular model stands out as a useful tool for AD mechanistic studies and perhaps for the development of personalized therapeutic strategies.

Highlights

  • We started by confirming the elevated expression of inflammatory-associated miR-124, miR-21, miR-125b, miR-155 and miR-146a in SH-SWE cells differentiated with retinoic acid (RA) and cultured for 24 h (Figure 1A; at least p < 0.05), as we have previously shown in [15]

  • Given the advantage of induced pluripotent stem cell (iPSC)-derived neurons in better mimicking the Alzheimer’s disease (AD) pathophysiology [39], we evaluated whether similar changes in those inflamma-miRNAs were present in the iPSCderived iNEU-PSEN cells from the AD patient, when compared with data from the healthy iPSC-derived iNEU-WT cells

  • We have found that miR-124, miR-21 and miR-125b were upregulated in SH-SWE cells and their exosomes, and that increased neuronal miR-124 contributed to its increased expression levels in microglial cells, after 48 and 72 h of coculturing, leading to a decrease in the miR-124 target CCAAT/Enhancer-Binding Protein alpha (C/EBP-α) mRNA expression

Read more

Summary

Introduction

By using SH-SY5Y neuroblastoma cells, An et al demonstrated that miR-124 is a potent negative regulator of β-site amyloid precursor protein-cleaving enzyme (BACE1) and that it may be involved in Alzheimer’s disease (AD) pathology, considering its reduced expression in AD sporadic patients [8]. A marked miR-124 upregulation in the hippocampus and temporal cortex was observed in AD patients and in the Tg2576 transgenic mice, where it was associated to an elevation of the amyloid-β (Aβ) peptide 1–42 [9]. These authors identified the miR-124/PTPN1 pathway as a critical mediator of synaptic dysfunction and memory loss in AD. The results about the beneficial or harmful effects of miR-124 expression levels in AD, which may result from the different models, mutations or disease stages used in such studies, are controversial

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

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