Skip to main content

Advertisement

Log in

LncRNA NKILA Exacerbates Alzheimer’s Disease Progression by Regulating the FOXA1-Mediated Transcription of TNFAIP1

  • Original Paper
  • Published:
Neurochemical Research Aims and scope Submit manuscript

Abstract

Alzheimer’s disease (AD) is one of the most common neurodegenerative diseases in the world, which seriously affects AD patients’ life quality. Recently, long non-coding RNAs (lncRNAs) have been reported to play a key role in AD pathogenesis, however, the specific mechanism remains unclear. Herein, we aimed to investigate the role of lncRNA NKILA in AD. The learning and memory performance of rats from streptozotocin (STZ)-treated or other treated groups were tested by Morris water maze test. Relative levels of genes and proteins were measured using RT-qPCR and Western blotting. Mitochondrial membrane potential was tested by JC-1 staining. Levels of ROS, SOD, MDA, GSH-Px, and LDH were measured using corresponding commercial kits. Apoptosis was evaluated by TUNEL staining or Flow cytometry assay. RNA Immunoprecipitation (RIP), RNA pulldown, Chromatin immunoprecipitation (ChIP), and dual-luciferase reporter assays were utilized to test the interaction between indicated molecules. STZ treatment caused learning and memory impairment in rats and oxidative stress damage in SH-SY5Y cells. LncRNA NKILA was found to be elevated in the hippocampal tissues of rats and SH-SY5Y cells after STZ exposure. Knockdown of lncRNA NKILA alleviated STZ-induced neuronal damage. Furthermore, lncRNA NKILA could bind to ELAVL1, which regulate the stability of FOXA1 mRNA. Moreover, TNFAIP1 transcription process was controlled by FOXA1, which targeted the promoter of TNFAIP1. In vivo results demonstrated that lncRNA NKILA accelerated STZ-induced neuronal damage and oxidative stress by FOXA1/TNFAIP1 axis. Our findings indicated that knockdown of lncRNA NKILA inhibited the neuronal damage and oxidative stress induced by STZ through the FOXA1/TNFAIP1 axis, thereby alleviating the development of AD, revealing a promising therapeutic axis for AD treatment.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Data Availability

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

  1. Soria Lopez JA, Gonzalez HM, Leger GC (2019) Alzheimer’s disease. Handb Clin Neurol 167:231–255

    Article  PubMed  Google Scholar 

  2. Zhang XX et al (2021) The epidemiology of Alzheimer’s disease modifiable risk factors and prevention. J Prev Alzheimers Dis 8(3):313–321

    PubMed  Google Scholar 

  3. Cummings JL, Tong G, Ballard C (2019) Treatment combinations for Alzheimer’s disease: current and future pharmacotherapy options. J Alzheimers Dis 67(3):779–794

    Article  PubMed  PubMed Central  Google Scholar 

  4. Parkhurst CN et al (2013) Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155(7):1596–1609

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Chen Z, Zhong C (2014) Oxidative stress in Alzheimer’s disease. Neurosci Bull 30(2):271–281

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Wang X et al (2014) Oxidative stress and mitochondrial dysfunction in Alzheimer’s disease. Biochim Biophys Acta 1842(8):1240–1247

    Article  CAS  PubMed  Google Scholar 

  7. Rizza S et al (2021) Alterations in Rev-ERBalpha/BMAL1 ratio and glycated hemoglobin in rotating shift workers: the EuRhythDia study. Acta Diabetol 58(8):1111–1117

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zhang L et al (2019) Silencing of long noncoding RNA SOX21-AS1 relieves neuronal oxidative stress Injury in mice with Alzheimer’s disease by upregulating FZD3/5 via the wnt signaling pathway. Mol Neurobiol 56(5):3522–3537

    Article  CAS  PubMed  Google Scholar 

  9. Wu W et al (2018) LncRNA NKILA suppresses TGF-beta-induced epithelial-mesenchymal transition by blocking NF-kappaB signaling in breast cancer. Int J Cancer 143(9):2213–2224

    Article  CAS  PubMed  Google Scholar 

  10. Gao YX et al (2020) Effects of lncRNA MALAT1 and lncRNA NKILA on proliferation, invasion and apoptosis of retinoblastoma. Eur Rev Med Pharmacol Sci 24(16):8296–8307

    PubMed  Google Scholar 

  11. Zhang GD et al (2019) LncRNA NKILA inhibits invasion and migration of osteosarcoma cells via NF-kappaB/Snail signaling pathway. Eur Rev Med Pharmacol Sci 23(10):4118–4125

    PubMed  Google Scholar 

  12. Wang M et al (2018) LncRNA NKILA upregulation mediates oxygen glucose deprivation/re-oxygenation-induced neuronal cell death by inhibiting NF-kappaB signaling. Biochem Biophys Res Commun 503(4):2524–2530

    Article  CAS  PubMed  Google Scholar 

  13. Maziuk B, Ballance HI, Wolozin B (2017) Dysregulation of RNA binding protein aggregation in neurodegenerative disorders. Front Mol Neurosci 10:89

    Article  PubMed  PubMed Central  Google Scholar 

  14. Brinegar AE, Cooper TA (2016) Roles for RNA-binding proteins in development and disease. Brain Res 1647:1–8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Schultz CW et al (2020) Understanding and targeting the disease-related RNA binding protein human antigen R (HuR). Wiley Interdiscip Rev RNA 11(3):e1581

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Yang F et al (2019) Circ-HuR suppresses HuR expression and gastric cancer progression by inhibiting CNBP transactivation. Mol Cancer 18(1):158

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hu YP et al (2019) LncRNA-HGBC stabilized by HuR promotes gallbladder cancer progression by regulating miR-502-3p/SET/AKT axis. Mol Cancer 18(1):167

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Palomo-Irigoyen M et al (2020) HuR/ELAVL1 drives malignant peripheral nerve sheath tumor growth and metastasis. J Clin Invest 130(7):3848–3864

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhang Q et al (2021) LncRNA HOTAIR promotes neuronal damage through facilitating NLRP3 mediated-pyroptosis activation in Parkinson’s disease via regulation of miR-326/ELAVL1 Axis. Cell Mol Neurobiol 41(8):1773–1786

    Article  CAS  PubMed  Google Scholar 

  20. Yang F et al (2020) Identification of key regulatory genes and pathways in prefrontal cortex of Alzheimer’s disease. Interdiscip Sci 12(1):90–98

    Article  PubMed  Google Scholar 

  21. Friedman JR, Kaestner KH (2006) The Foxa family of transcription factors in development and metabolism. Cell Mol Life Sci 63(19–20):2317–2328

    Article  CAS  PubMed  Google Scholar 

  22. Cong L et al (2021) Up-regulated microRNA-132 reduces the cognition-damaging effect of sevoflurane on Alzheimer’s disease rats by inhibiting FOXA1. Genomics 113(6):3644–3652

    Article  CAS  PubMed  Google Scholar 

  23. Zhou J et al (2005) Cloning of two rat PDIP1 related genes and their interactions with proliferating cell nuclear antigen. J Exp Zool A Comp Exp Biol 303(3):227–240

    Article  PubMed  Google Scholar 

  24. Yang L et al (2010) CK2 phosphorylates TNFAIP1 to affect its subcellular localization and interaction with PCNA. Mol Biol Rep 37(6):2967–2973

    Article  CAS  PubMed  Google Scholar 

  25. Kim DM et al (2009) RhoB induces apoptosis via direct interaction with TNFAIP1 in HeLa cells. Int J Cancer 125(11):2520–2527

    Article  CAS  PubMed  Google Scholar 

  26. Link CD et al (2003) Gene expression analysis in a transgenic Caenorhabditis elegans Alzheimer’s disease model. Neurobiol Aging 24(3):397–413

    Article  CAS  PubMed  Google Scholar 

  27. Liu N et al (2016) TNFAIP1 contributes to the neurotoxicity induced by Abeta25-35 in Neuro2a cells. BMC Neurosci 17(1):51

    Article  PubMed  PubMed Central  Google Scholar 

  28. Dong LX et al (2021) LncRNA NEAT1 promotes Alzheimer’s disease by down regulating micro-27a-3p. Am J Transl Res 13(8):8885–8896

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhang J, Wang R (2021) Deregulated lncRNA MAGI2-AS3 in Alzheimer’s disease attenuates amyloid-beta induced neurotoxicity and neuroinflammation by sponging miR-374b-5p. Exp Gerontol 144:111180

    Article  CAS  PubMed  Google Scholar 

  30. Yi J et al (2019) Upregulation of the lncRNA MEG3 improves cognitive impairment, alleviates neuronal damage, and inhibits activation of astrocytes in hippocampus tissues in Alzheimer’s disease through inactivating the PI3K/Akt signaling pathway. J Cell Biochem 120(10):18053–18065

    Article  CAS  PubMed  Google Scholar 

  31. Zhou Q et al (2018) NKILA inhibition protects retinal pigment epithelium cells from hypoxia by facilitating NFkappaB activation. Biochem Biophys Res Commun 503(4):3134–3141

    Article  CAS  PubMed  Google Scholar 

  32. Ebert MS, Neilson JR, Sharp PA (2007) MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nat Methods 4(9):721–726

    Article  CAS  PubMed  Google Scholar 

  33. Tay Y, Rinn J, Pandolfi PP (2014) The multilayered complexity of ceRNA crosstalk and competition. Nature 505(7483):344–352

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Haq S et al (2019) The stability and oncogenic function of LIN28A are regulated by USP28. Biochim Biophys Acta Mol Basis Dis 1865(3):599–610

    Article  CAS  PubMed  Google Scholar 

  35. Mang Y et al (2017) Long noncoding RNA NEAT1 promotes cell proliferation and invasion by regulating hnRNP A2 expression in hepatocellular carcinoma cells. Onco Targets Ther 10:1003–1016

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Skliris A et al (2015) Neuroprotection requires the functions of the RNA-binding protein HuR. Cell Death Differ 22(5):703–718

    Article  CAS  PubMed  Google Scholar 

  37. Taube JH et al (2010) Foxa1 functions as a pioneer transcription factor at transposable elements to activate afp during differentiation of embryonic stem cells. J Biol Chem 285(21):16135–16144

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Pristera A et al (2015) Transcription factors FOXA1 and FOXA2 maintain dopaminergic neuronal properties and control feeding behavior in adult mice. Proc Natl Acad Sci USA 112(35):E4929–E4938

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Xiao Y et al (2021) TNFAIP1 is upregulated in APP/PS1 mice and promotes apoptosis in SH-SY5Y cells by binding to RhoB. J Mol Neurosci 71(6):1221–1233

    Article  CAS  PubMed  Google Scholar 

  40. He D, Tan J, Zhang J (2017) miR-137 attenuates Abeta-induced neurotoxicity through inactivation of NF-kappaB pathway by targeting TNFAIP1 in Neuro2a cells. Biochem Biophys Res Commun 490(3):941–947

    Article  CAS  PubMed  Google Scholar 

  41. Yi J et al (2020) TNFAIP1 mediates Formaldehyde-Induced neurotoxicity by inhibiting the Akt/CREB pathway in N2a cells. Neurotox Res 38(1):184–198

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work was supported by National Natural Science Foundation of China (82074380, 82074503), Discipline Innovation Team of Shaanxi University of Chinese Medicine (2019-QN05), Key research and development project of Shaanxi Province (2020SF-336, 2023-YBSF-615), Key scientific research project of Shaanxi Provincial Department of Education (22JS010), Scientific research Project of Traditional Chinese Medicine Administration of Shaanxi Province (SZY-KJCYC-2023-064), Key research and development plan of Xianyang City (L2022ZDYFSF055), School-level Scientific Research Project of Shaanxi University of Chinese medicine (2020GP41, 2021GP01).

Author information

Authors and Affiliations

Authors

Contributions

Conception of manuscript: all authors. Manuscript design and writing: YZ, YW, YW. Manuscript reviewing and editing: all authors. All authors read and agreed to the final version of the manuscript.

Corresponding authors

Correspondence to Yanjin Su, Yongchang Diwu or Qi Zhang.

Ethics declarations

Competing Interests

The authors declare no competing interests.

Ethical Approval

This study protocol was reviewed and approved by Shaanxi University of Chinese Medicine.

Consent to Participate

This study protocol was reviewed and approved by Shaanxi University of Chinese Medicine.

Consent for Publication

The informed consent was obtained from study participants.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, Y., Wang, Y., Wang, Y. et al. LncRNA NKILA Exacerbates Alzheimer’s Disease Progression by Regulating the FOXA1-Mediated Transcription of TNFAIP1. Neurochem Res 48, 2895–2910 (2023). https://doi.org/10.1007/s11064-023-03944-6

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11064-023-03944-6

Keywords

Navigation