Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Jan 24;8(1):33.
doi: 10.1038/s41420-022-00823-x.

MicroRNA-181a-2-3p shuttled by mesenchymal stem cell-secreted extracellular vesicles inhibits oxidative stress in Parkinson's disease by inhibiting EGR1 and NOX4

Affiliations

MicroRNA-181a-2-3p shuttled by mesenchymal stem cell-secreted extracellular vesicles inhibits oxidative stress in Parkinson's disease by inhibiting EGR1 and NOX4

Jianjun Ma et al. Cell Death Discov. .

Abstract

The current study investigated the physiological mechanisms by which extracellular vesicle (EV)-encapsulated miR-181a-2-3p derived from mesenchymal stem cells (MSCs) might mediate oxidative stress (OS) in Parkinson's disease (PD). First, 6-hydroxydopamine (6-OHDA)-induced PD cell and mouse models were established, after which miR-181a-2-3p, EGR1, and NOX4 expression patterns were determined in SH-SY5Y cells and substantia nigra (SN) of PD mice. Next, the binding affinity among miR-181a-2-3p, EGR1, and NOX4 was identified using multiple assays. Gain- or loss-of-function experiments were further adopted to detect SH-SY5Y cell proliferation and apoptosis and to measure the levels of SOD, MDA, and ROS. Finally, the effects of miR-181a-2-3p from MSC-derived EVs in PD mouse models were also explored. It was found that miR-181a-2-3p was poorly expressed in 6-OHDA-induced SH-SY5Y cells, whereas miR-181a-2-3p from MSCs could be transferred into SH-SY5Y cells via EVs. In addition, miR-181a-2-3p could target and inhibit EGR1, which promoted the expression of NOX4. The aforementioned miR-181a-2-3p shuttled by MSC-derived EVs facilitated SH-SY5Y proliferation and SOD levels, but suppressed apoptosis and MDA and ROS levels by regulating EGR1 via inhibition of NOX4/p38 MAPK, so as to repress OS of PD. Furthermore, in PD mice, miR-181a-2-3p was carried by EVs from MSCs to alleviate apoptosis of dopamine neurons and OS, accompanied by increased expressions of α-syn and decreased 4-HNE in SN tissues. Collectively, our findings revealed that MSC-derived EV-loaded miR-181a-2-3p downregulated EGR1 to inhibit OS via the NOX4/p38 MAPK axis in PD.

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1. miR-181a–2–3p inhibits apoptosis and OS of SH-SY5Y cells.
A The box plot of miR-181a–2–3p in GSE16658, the blue box on the left indicated expression of normal samples, and the red box on the right indicates miR-181a–2–3p expression in PD patients. B Radioactive dopamine-uptake test. C Cell viability in SH-SY5Y cells treated with 6-OHDA determined with MTT assay. D Intracellular oxidation level evaluated after treatment with different drug concentrations. E LDH-release assay. F miR-181a-2-3p expression detected in SH-SY5Y cells treated with 6-OHDA for 24 h. G miR-181a–2–3p expression detected in 6-OHDA-induced SH-SY5Y cells using RT-qPCR. H SH-SY5Y cell proliferation detected using CCK-8 assay after 6-OHDA treatment. I Apoptosis of SH-SY5Y cells treated with 6-OHDA detected using flow cytometry. J SOD level in SH-SY5Y cells treated with 6-OHDA detected using SOD kit. K MDA level in SH-SY5Y cells treated with 6-OHDA detected using MDA kit. L ROS levels in SH-SY5Y cells treated with 6-OHDA detected using DCFH-DA fluorescent staining. *p < 0.05, **p < 0.01, ***p < 0.001 vs. control or SH-SY5Y cells without treatment. Cell experiments were repeated three times independently.
Fig. 2
Fig. 2. miR-181a–2–3p could be carried to SH-SY5Y cells through MSC–EVs.
A Abundance of miR-181a–2–3p in MSCs and MSC-derived EVs. B miR-181a–2–3p expression by RT-qPCR. C EVs observed under the TEM. D EV diameter detected using dynamic light scattering. E EV surface markers detected using Western blot. F Uptake of MSC–EV by SH-SY5Y cells detected using EV PKH67 tracing method. G Cy3-labeled miR-181a–2–3p entered SH-SY5Y cells via MSC–EV (200×). H miR-181a–2–3p expression in the SH-SY5Y cells. *p < 0.05 vs. SH-SY5Y blank, ***p < 0.001, NS insignificant difference. Cell experiments were repeated three times independently.
Fig. 3
Fig. 3. MSC–EVs carrying miR-181a–2–3p inhibit apoptosis and OS of SH-SY5Y cells.
A miR-181a–2–3p expression determined by RT-qPCR. B Cell proliferation in SH-SY5Y cells co-incubated with MSC–EVs. C Apoptosis of SH-SY5Y cells co-incubated with MSC–EVs. D SOD level in SH-SY5Y cells co-incubated with MSC–EVs using SOD kit. E MDA level in SH-SY5Y cells co-incubated with MSC–EVs using MDA kit. F ROS levels in SH-SY5Y cells co-incubated with MSC-EVs using DCFH-DA fluorescent staining. *p < 0.05 vs. MSC–EV–NC mimic or SH-SY5Y cells without treatment, #p < 0.05 vs. 6-OHDA-induced SH-SY5Y cells; $p < 0.05 vs. SH-SY5Y cells co-incubated with MSC–EV–NC mimic. Cell experiments were repeated three times independently.
Fig. 4
Fig. 4. miR-181a–2–3p targets EGR1.
A The downstream genes of miR-181a–2–3p predicted using DIANA TOOLS and mircroRNA and Venn map of the human transcription factor obtained from hTFtarget, Cistrome and JASPAR. B The interaction network of proteins of 9 important transcription factors constructed by String. Red represents the higher core degree, and blue represents the lower core degree. C The binding sites between miR-181a–2–3p and EGR1 predicted using DIANA TOOL. D The binding of miR-181a–2–3p to EGR1 using dual-luciferase assay. E EGR1 mRNA level in SH-SY5Y cells treated with 6-OHDA determined using RT-qPCR. F Western blot analysis of EGR1 protein level in SH-SY5Y cells treated with 6-OHDA. G EGR1 mRNA level in SH-SY5Y cells treated with miR-181a–2–3p mimic or inhibitor determined using RT-qPCR. H Western blot analysis of EGR1 protein level in SH-SY5Y cells treated with miR-181a-2-3p mimic or inhibitor. *p < 0.05 vs. NC mimic or *p < 0.05, **p < 0.01, ***p < 0.001 vs. SH-SY5Y cells without treatment; #p < 0.05 vs. NC inhibitor. Cell experiments were repeated three times independently.
Fig. 5
Fig. 5. miR-181a–2–3p suppresses apoptosis and OS of SH-SY5Y cells by inhibiting EGR1 expression.
A The knockdown effect of the two interference sequences of EGR1. B Expression of miR-181a–2–3p and EGR1 in SH-SY5Y cells after different treatments. C Cell proliferation in SH-SY5Y cells after different treatments. D Apoptosis of SH-SY5Y cells after different treatments. E SOD level in SH-SY5Y cells after different treatments detected using SOD kit. F MDA level in SH-SY5Y cells after different treatments using MDA kit. G ROS levels in SH-SY5Y cells after different treatments detected using DCFH-DA fluorescent staining. *p < 0.05 vs. si-NC, NC inhibitor + si-NC, or cells without treatment, #p < 0.05 vs. NC inhibitor + si-EGR1 or NC inhibitor + si-NC; $p < 0.05 vs. NC inhibitor + si-EGR1. Cell experiments were repeated three times independently.
Fig. 6
Fig. 6. EGR1 promotes apoptosis and OS of SH-SY5Y cells by regulating NOX4.
A Significant co-expression of EGR1 and NOX4 analyzed using MEM. B EGR1 enriched in the NOX4 promoter region verified by ChIP assay. C NOX4 mRNA level in SH-SY5Y cells treated with 6-OHDA at different concentrations detected using RT-qPCR. D NOX4 protein level in SH-SY5Y cells treated with 6-OHDA at different concentrations. E The knockdown effect of the two interference sequences of NOX4. F Expression of EGR1 and NOX4 in SH-SY5Y cells after different treatments measured using RT-qPCR. G Cell proliferation in SH-SY5Y cells after different treatments. H Cell apoptosis in SH-SY5Y cells after different treatments. I SOD level in SH-SY5Y cells after different treatments. J MDA level in SH-SY5Y cells after different treatments. K ROS levels in SH-SY5Y cells after different treatments detected using DCFH-DA fluorescent staining. *p < 0.05 vs. si-NC, oe-NC + si-NC, or SH-SY5Y cells without treatment, #p < 0.05 vs. oe-NC + si-NOX4, oe-NC + si-NC; $p < 0.05 vs. oe-NC + si-NOX4. Cell experiments were repeated three times independently.
Fig. 7
Fig. 7. p38 MAPK pathway mediates NOX4 to promote apoptosis and OS of SH-SY5Y cells.
A MEM analysis of NOX4 and p38 MAPK significant co-expression. B The levels of NOX4, p–p38, and p38 in SH-SY5Y cells after different treatments measured by Western blot assay. C Cell proliferation in SH-SY5Y cells after different treatments. D Cell apoptosis in SH-SY5Y cells after different treatments. E SOD level in SH-SY5Y cells after different treatments. F MDA level in SH-SY5Y cells after different treatments. G ROS levels in SH-SY5Y cells after different treatments detected using DCFH-DA fluorescent staining. *p < 0.05 vs. SH-SY5Y cells without treatment, #p < 0.05 vs. oe-NC + DMSO; $p < 0.05 vs. oe-NOX4 + DMSO. Cell experiments were repeated three times independently.
Fig. 8
Fig. 8. miR-181a–2–3p inhibits apoptosis and OS of SH-SY5Y cells by regulating the EGR1/NOX4 axis.
A miR-181a–2–3p, EGR1, and NOX4 expression in SH-SY5Y cells treated with 6-OHDA in combination with different plasmids determined using RT-qPCR. B Levels of p–p38 and p38 in SH-SY5Y cells treated with 6-OHDA in combination with different plasmids determined using Western blot analysis. C Cell proliferation in SH-SY5Y cells treated with 6-OHDA in combination with different plasmids. D Cell apoptosis in SH-SY5Y cells treated with 6-OHDA in combination with different plasmids. E SOD level in SH-SY5Y cells treated with 6-OHDA in combination with different plasmids detected using SOD kit. F MDA level in SH-SY5Y cells treated with 6-OHDA in combination with different plasmids detected using MDA kit. G ROS levels in SH-SY5Y cells treated with 6-OHDA in combination with different plasmids detected using DCFH-DA fluorescent staining. C, E, F, and G, *p < 0.05 vs. NC mimic + oe-NC or SH-SY5Y cells without treatment, #p < 0.05 vs. miR-181a–2–3p mimic + oe-NC or NC mimic + oe-NC; $p < 0.05 vs. miR-181a–2–3p mimic + oe-NC. Cell experiments were repeated three times independently.
Fig. 9
Fig. 9. MSC–EVs carrying miR-181a–2–3p inhibit neuronal loss and OS injury in the SN area of PD mice.
A The PKH67-labeled EVs were injected intravenously into mice. Colocalization of EVs (green) with dopaminergic neurons in the SN tissues (the left panel). α-SYN expression (red) determined by immunofluorescence. DAPI (blue) located in the nucleus (scale bar = 15 μm). B miR-181a–2–3p expression in SN tissues detected using RT-qPCR. C EGR1, NOX4, p–p38, and p38 levels in SN tissues detected using Western blot analysis. D Pathological changes of SN tissues determined with H&E staining. The arrow represents the lost neurons (200 ×). E APO-induced asymmetric rotation in PD mice injected with EVs after 8 weeks. F TH expression in mouse SN tissues detected by immunohistochemical staining. G TH expression in mouse SN tissues detected using Western blot analysis. H OS marker 4-HNE in mouse SN tissues. I The dopaminergic neuron apoptosis in SN tissues detected by immunofluorescence staining. J The motor ability of mice detected by roller test. K The motor ability of mice determined by rod-climbing pole test *p < 0.05 vs. sham-operated mice, #p < 0.05 vs. saline; $p < 0.05 vs. MSC–EV–NC mimic.
Fig. 10
Fig. 10
MSC–EVs carrying miR-181a–2–3p into SH-SY5Y cells inhibit EGR1 to suppress NOX4 transcription and activation of p38 MAPK pathway, thereby repressing the apoptosis and OS of SH-SY5Y cells in PD.

References

    1. Bi XA, Wu H, Xie Y, Zhang L, Luo X, Fu Y et al. The exploration of Parkinson’s disease: a multi-modal data analysis of resting functional magnetic resonance imaging and gene data. Brain Imaging Behav. 2020;15:1986–96. - PubMed
    1. Wu DM, Han XR, Wen X, Wang S, Fan SH, Zhuang J, et al. Salidroside protection against oxidative stress injury through the Wnt/beta-catenin signaling pathway in rats with Parkinson’s disease. Cell Physiol. Biochem. 2018;46:1793–806. doi: 10.1159/000489365. - DOI - PubMed
    1. Puspita L, Chung SY, Shim JW. Oxidative stress and cellular pathologies in Parkinson’s disease. Mol. Brain. 2017;10:53. doi: 10.1186/s13041-017-0340-9. - DOI - PMC - PubMed
    1. Morry J, Ngamcherdtrakul W, Yantasee W. Oxidative stress in cancer and fibrosis: Opportunity for therapeutic intervention with antioxidant compounds, enzymes, and nanoparticles. Redox Biol. 2017;11:240–53. doi: 10.1016/j.redox.2016.12.011. - DOI - PMC - PubMed
    1. Glavaski-Joksimovic A, Bohn MC. Mesenchymal stem cells and neuroregeneration in Parkinson’s disease. Exp Neurol. 2013;247:25–38. doi: 10.1016/j.expneurol.2013.03.016. - DOI - PubMed