Elsevier

Brain Research

Volume 1750, 1 January 2021, 147156
Brain Research

Research report
Deficiency of NEAT1 prevented MPP+-induced inflammatory response, oxidative stress and apoptosis in dopaminergic SK-N-SH neuroblastoma cells via miR-1277-5p/ARHGAP26 axis

https://doi.org/10.1016/j.brainres.2020.147156Get rights and content

Highlights

  • NEAT1 was highly induced in response to MPP+ in SK-N-SH cells.

  • Deficiency of NEAT1 suppressed MPP+-induced apoptosis, inflammatory response and oxidative stress in SK-N-SH cells.

  • NEAT1 knockdown downregulated ARHGAP26 via targeting miR-1277-5p.

  • Overexpression of miR-1277-5p triggered similar effect to NEAT1 deficiency in MPP+-induced SK-N-SH cells.

Abstract

Noncoding RNAs including long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) have been documented to play prominent role in neurodegenerative diseases including Parkinson’s disease (PD). This study intended to investigate the role of lncRNA nuclear enriched assembly transcript 1 (NEAT1) in MPP+-induced PD model in dopaminergic neuronblastoma SK-N-SH cells, as well as its mechanism through sponging miRNA (miR)-1277-5p. Real-time PCR and western blotting revealed that NEAT1 and ARHGAP26 were upregulated, and miR-1277-5p was downregulated in MPP+-treated SK-N-SH cells in a certain of concentration- and time- dependent manner. MPP+ induced apoptosis in SK-N-SH cells, as evidenced by decreased cell viability and Bcl-2 expression, and elevated apoptosis rate and levels of Bax and cleaved caspase-3, which were examined by MTT assay, flow cytometry and western blotting. Moreover, commercial assay kits indicated that inflammatory response and oxidative stress were provoked in response to MPP+, due to promoted contents of interleukin (IL)-6, IL-1β, tumor necrosis factor-α, malondialdehyde, and lactate dehydrogenase, accompanied with suppressed superoxide dismutase and glutathione peroxidase levels. Notably, MPP+-induced apoptosis, inflammatory response and oxidative stress in SK-N-SH cells were mitigated by NEAT1 knockdown and/or miR-1277-5p overexpression. Moreover, silencing of miR-1277-5p could abrogate the suppression of NEAT1 deficiency on MPP+-induced cell injury. Similarly, upregulating miR-1277-5p-elicited neuroprotection in MPP+-induced SK-N-SH cells was reversed by ARHGAP26 restoration. Dual-luciferase reporter assay demonstrated a direct interaction between miR-1277-5p and NEAT1 or ARHGAP26. Collectively, NEAT1 upregulation might contribute to MPP+-induced neuron injury via NEAT1-miR-1277-5p-ARHGAP26 competing endogenous RNAs (ceRNAs) pathway.

Introduction

Parkinson’s disease (PD), the second most common neurodegenerative disorder after alzheimer's disease (Poewe et al., 2017). The incidence of PD is rapidly increased with age in China (Li et al., 2019), making PD as one main threat in aged people. Although the etiology of PD is still unclear, genetic factors and environmental factors (such as toxicants) have been declared to evoke the development of PD (Dunn et al., 2019). The common etiopathogenesis of PD is ascribed to the loss of dopaminergic neurons in substantia nigra pars compacta, and the accumulation of Lewy bodies in the brain (Poewe et al., 2017). Extensive evidences have indicated that inflammatory damage and oxidative injury are responsible to neurodegeneration and neuroinflammation in PD (Anderson et al., 2018, Puspita et al., 2017). N-methyl-4-phenylpyridinium (MPP+) is the active metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a kind of neurotoxin that selectively destroys dopaminergic neurons, eventually leading to the occurrence of PD (Jiang et al., 2019b). Therefore, clarifying the mechanism of MPP+-induced PD is important to clarify the pathogenesis of PD.

Noncoding RNAs including long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) are abnormally expressed in neurodevelopment and neurodegenerative diseases including PD (Majidinia et al., 2016, Oe et al., 2019). Growing evidences support a key role of lncRNAs and miRNAs in dopaminergic dysfunction in neurological pathologies, such as PD (Carrick et al., 2016). LncRNAs are longer than 200 nucleotides (nt), and are structurally similar to message RNAs (mRNAs), even though lncRNAs have little protein coding capacity (Lyu et al., 2019). Newest researches on the pathogenesis of diseases have elucidated that lncRNAs are emerging targets for diagnosis and drug treatment of PD (Lyu et al., 2019), and that the role of several lncRNAs have already been illuminated in the pathogenesis of PD (Fan et al., 2019, Majidinia et al., 2016). However, the research on contribution of lncRNAs in PD remains infant. LncRNA nuclear enriched assembly transcript 1 (NEAT1) is aberrantly-mostly upregulated in non-cancerous pathological conditions, including neurodegenerative diseases (Prinz et al., 2019). Furthermore, the de novo investigations have disclosed the close relationship among NEAT1, inflammatory response (Xie et al., 2019, Zhang et al., 2019b) and oxidative stress (Simchovitz et al., 2019, Sunwoo et al., 2017). Here, we intended to explore the role of NEAT1 in inflammatory injury and oxidative damage in PD.

MiRNAs are a class of transcripts (19–24 nt) that control the expression of multiple mRNAs at post-transcriptional level (Majidinia et al., 2016). Numerous documents have expounded the prominent role of miRNAs in PD (Goh et al., 2019), and cell-free miRNAs are considered as potential biomarkers for central nervous system disorders including PD (Doxakis, 2020, van den Berg et al., 2020). MiRNA (miR)-1277-5p is associated with accurate molecular classification of different cancers, such as kidney cancer, colorectal cancer, hepatocellular carcinoma (Cao et al., 2019, Motieghader et al., 2017, Youssef et al., 2011). Nevertheless, the function of miR-1277-5p is not well understood presently. Thus, we wondered the role of miR-1277-5p in MPP+-induced dopaminergic neurons.

The presented study focused on the expression of NEAT1 and miR-1277-5p in MPP+-induced model of PD in dopaminergic neuroblastoma SK-N-SH cells. Furthermore, the role of both was also investigated in inflammatory injury and oxidative damage in response to MPP+. Notably, the molecular mechanism of NEAT1 was further clarified through functioning as a competing endogenous RNA (ceRNA) of miR-1277-5p to regulate Rho GTPase-activating protein 26 (ARHGAP26, also known as GRAF), a negative regulator of the Rho family (Wang et al., 2013).

Section snippets

Expression of NEAT1 was upregulated in cell model of PD

An in vitro model of PD was established in dopaminergic neuroblastoma SK-N-SH cells treated with MPP+. As shown in Fig. 1A and 1B, expression level of NEAT1 was gradually increased in SK-N-SH cells after treatment of 0.25–1 mM MPP+ for 24 h and 1 mM MPP+ for 12–48 h. Thus, we considered that NEAT1 was upregulated in MPP+-induced SK-N-SH cells in a certain of concentration- and time-dependent manner, suggesting a potential role of NEAT1 in neuronal cell injury in PD. And, 1 mM MPP+ treatment for

Discussion

In this study, we proposed that NEAT1 might contribute to dopaminergic neuroblastoma SK-N-SH cell injury by regulating miR-1277-5p and ARHGAP26 to modulate cell viability, apoptosis, inflammation and oxidative stresses, suggesting aNEAT1-miR-1277-5p-ARHGAP26 ceRNA pathway underlying the pathogenesis of PD (Fig. 7).

NEAT1 was overexpressed in both PD cell model and drug-inducible neuroprotection against oxidative damage (Simchovitz et al., 2019). This pointed out a complicated involvement of

Cell model of PD

The dopaminergic neuroblastoma cell lines, SK-N-SH (no. 86012802) and SH-SY5Y (no. 94030304; differentiated from SK-N-SH) were from European Collection of Authenticated Cell Cultures (Public Health England, UK), and MPP+ (no. D048) was from Sigma-Aldrich (St. Louis, MO, USA). For establishment of cell model of PD, SK-N-SH cells were treated with 0.25, 0.5 and 1 mM of MPP+ for 0–48 h in Dulbecco’s modified Eagle’s medium (Hyclone, Logan, Utah, USA) plus 10% fetal bovine serum (Hyclone) in a

CRediT authorship contribution statement

Shufang Zhou: Validation, Investigation, Writing - original draft, Writing - review & editing, Project administration. Dan Zhang: Conceptualization, Methodology, Writing - original draft, Writing - review & editing. Junnan Guo: Conceptualization, Methodology, Writing - original draft, Writing - review & editing. Zhenzhen Chen: Formal analysis, Data curation, Software. Yong Chen: Formal analysis, Data curation, Validation, Investigation. Junshi Zhang: Validation, Investigation.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References (41)

  • X. Cao et al.

    MicroRNA-1277 Inhibits Proliferation and Migration of Hepatocellular Carcinoma HepG2 Cells by Targeting and Suppressing BMP4 Expression and Reflects the Significant Indicative Role in Hepatocellular Carcinoma Pathology and Diagnosis After Magnetic Resonance Imaging Assessment

    Oncol. Res.

    (2019)
  • W.T. Carrick et al.

    Noncoding RNA regulation of dopamine signaling in diseases of the central nervous system

    Front. Mol. Biosci.

    (2016)
  • D.-D. Chen et al.

    NEAT1 contributes to ox‐LDL‐induced inflammation and oxidative stress in macrophages through inhibiting miR‐128

    J. Cell. Biochem.

    (2019)
  • H. Chen et al.

    LncRNA-NEAT1 from the competing endogenous RNA network promotes cardioprotective efficacy of mesenchymal stem cell-derived exosomes induced by macrophage migration inhibitory factor via the miR-142-3p/FOXO1 signaling pathway

    Stem. Cell Res. Ther.

    (2020)
  • Y. Chen et al.

    Long non-coding RNA NEAT1 plays an important role in sepsis-induced acute kidney injury by targeting miR-204 and modulating the NF-kappaB pathway

    Int Immunopharmacol

    (2018)
  • Y. Fan et al.

    Dysregulated Long Non-coding RNAs in Parkinson's Disease Contribute to the Apoptosis of Human Neuroblastoma Cells

    Front. Neurosci.

    (2019)
  • S.Y. Goh et al.

    Role of MicroRNAs in Parkinson's Disease

    Int. J. Mol. Sci.

    (2019)
  • D. Han et al.

    YY1-induced upregulation of lncRNA NEAT1 contributes to OGD/R injury-induced inflammatory response in cerebral microglial cells via Wnt/beta-catenin signaling pathway

    Vitro Cell Dev. Biol. Anim.

    (2019)
  • S. Jarius et al.

    Two new cases of anti-Ca (anti-ARHGAP26/GRAF) autoantibody-associated cerebellar ataxia

    J. Neuroinflammation

    (2013)
  • F. Jiang et al.

    Identification of potential diagnostic biomarkers for Parkinson's disease

    FEBS Open Bio

    (2019)
  • Cited by (35)

    • LncRNA DLX6-AS1 promotes microglial inflammatory response in Parkinson's disease by regulating the miR-223–3p/NRP1 axis

      2022, Behavioural Brain Research
      Citation Excerpt :

      LncRNAs have been accepted to function in human diseases by serving as competing endogenous RNAs (ceRNAs) by binding to microRNAs (miRNAs) and indirectly interacting with mRNAs [10]. For instance, lncRNA NEAT1 upregulation might contribute to MPTP-induced neuron injury via the NEAT1/miR-1277–5p/ARHGAP26 ceRNA pathway [11]. miRNAs are tiny non-coding RNAs, about 23 nt in size, which play a crucial role in gene regulation by binding to three untranslated regions (3’UTR) of mRNA [12].

    • Current and Future Perspectives of Noncoding RNAs in Brain Function and Neuropsychiatric Disease

      2022, Biological Psychiatry
      Citation Excerpt :

      Malat1 regulates a conserved set of synaptic plasticity–associated genes and thereby alters dendritic spine density (57). Neat1, an lncRNA involved in nuclear organization via phase separation of paraspeckles (58,59), is involved in the response to oxidative stress in dopaminergic neurons (60), and Meg3 modulates the degree of AMPA receptor expression of the neuronal cell surface (61). The lncRNA GM12371 is involved in transcriptional regulation of synapse-associated genes in the hippocampus and is activated by the cAMP-PKA signaling cascade (19).

    View all citing articles on Scopus
    View full text