Abstract

Parkinson’s disease (PD) is a prevalent neurodegenerative disorder with pathological features including death of dopaminergic neurons in the substantia nigra and intraneuronal accumulations of Lewy bodies. As the main component of Lewy bodies, α-synuclein is implicated in PD pathogenesis by aggregation into insoluble filaments. However, the detailed mechanisms underlying α-synuclein induced neurotoxicity in PD are still elusive. MicroRNAs are ~20nt small RNA molecules that fine-tune gene expression at posttranscriptional level. A plethora of miRNAs have been found to be dysregulated in the brain and blood cells of PD patients. Nevertheless, the detailed mechanisms and their in vivo functions in PD still need further investigation. By using Drosophila PD model expressing α-synuclein A30P, we examined brain miRNA expression with high-throughput small RNA sequencing technology. We found that five miRNAs (dme-miR-133-3p, dme-miR-137-3p, dme-miR-13b-3p, dme-miR-932-5p, dme-miR-1008-5p) were upregulated in PD flies. Among them, miR-13b, miR-133, miR-137 are brain enriched and highly conserved from Drosophila to humans. KEGG pathway analysis using DIANA miR-Path demonstrated that neuroactive-ligand receptor interaction pathway was most likely affected by these miRNAs. Interestingly, miR-137 was predicted to regulate most of the identified targets in this pathway, including dopamine receptor (DopR, D2R), γ-aminobutyric acid (GABA) receptor (GABA-B-R1, GABA-B-R3) and N-methyl-D-aspartate (NMDA) receptor (Nmdar2). The validation experiments showed that the expression of miR-137 and its targets was negatively correlated in PD flies. Further experiments using luciferase reporter assay confirmed that miR-137 could act on specific sites in 3’ UTR region of D2R, Nmdar2 and GABA-B-R3, which downregulated significantly in PD flies. Collectively, our findings indicate that α-synuclein could induce the dysregulation of miRNAs, which target neuroactive ligand-receptor interaction pathway in vivo. We believe it will help us further understand the contribution of miRNAs to α-synuclein neurotoxicity and provide new insights into the pathogenesis driving PD.

Highlights

  • Parkinson’s disease (PD) is the second most prevalent neurodegenerative disorder affecting the elderly population [1]

  • Multiplication of α-synuclein or mutations such as A53T, A30P and E46K were found in familial forms PD patients [3,4,5].the detailed mechanisms underlying α-synuclein induced neurotoxicity in PD still need further investigation

  • We found that PD flies exhibited shorter lifespan (Fig 1A) and impaired locomotive ability (Fig 1B) compared with control flies

Read more

Summary

Introduction

Parkinson’s disease (PD) is the second most prevalent neurodegenerative disorder affecting the elderly population [1]. Its predominant pathological features are death of dopaminergic (DA) neurons in the substantia nigra pars compacta and intraneuronal accumulations of Lewy bodies [2]. PD animal models have been established by ectopic expression of human α-synuclein in yeast, Caenorhabditis elegans, Drosophila melanogaster, rat, mouse, and non-human primates [6,7,8,9,10,11]. Drosophila models have been widely used to study neurodegenerative diseases including Alzheimer’s disease (AD), Huntington’s disease (HD) and PD [8, 12, 13]. Panneuronal expression of human wild type and mutant α-synuclein (A53T and A30P) demonstrate adult onset PD pathological features including DA neuronal loss, decreased dopamine level, impaired locomotive ability and shortened lifespan [8, 14, 15]. Drosophila models provide efficient tools for screening genes participate in PD and potential drugs against PD

Methods
Results
Discussion
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