Open Access

Role of hypoxia‑inducible factor‑1α in autophagic cell death in microglial cells induced by hypoxia

  • Authors:
    • Xintao Wang
    • Jun Ma
    • Qiang Fu
    • Lei Zhu
    • Zhiling Zhang
    • Fan Zhang
    • Nan Lu
    • Aimin Chen
  • View Affiliations

  • Published online on: March 1, 2017     https://doi.org/10.3892/mmr.2017.6277
  • Pages: 2097-2105
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Microglial cells are phagocytic cells of the central nervous system (CNS) and have been proposed to be a primary component of the innate immune response and maintain efficient CNS homeostasis. Microglial cells are activated during various phases of tissue repair and participate in various pathological conditions in the CNS. Following spinal cord injury (SCI), anoxemia is a key problem that results in tissue destruction. Hypoxia‑inducible factor 1‑α (HIF‑1α) may protect hypoxic cells from apoptosis or necrosis under ischemic and anoxic conditions. However, numerous studies have revealed that hypoxia upregulates HIF‑1α expression leading to the death of microglial cells. The present study investigated the alterations in HIF‑1α expression levels and the mechanism of autophagic cell death mediated by HIF‑1α in microglial cells induced by hypoxia. Hypoxia was demonstrated to induce HIF‑1α expression and autophagic cell death in microglial cells. Enhanced autophagy reduced cell death during the initial stages by restraining the functions of autophagy‑associated genes (microtubule‑associated protein 1A/1B‑light chain 3 phosphatidylethanolamine conjugate and Beclin‑1) and modulating the expression of inflammatory cytokines (tumor necrosis factor‑α and interleukin‑1β). Target value was determined by Cell Counting Kit 8 and cell death by flow cytometry. Transmission electron microscopy, immunohistochemical staining, reverse transcription‑quantitative polymerase chain reaction, western blotting, and ELISA were used for further analysis. However, increased expression of HIF‑1α induced cell death and autophagic cell death in microglial cells. Furthermore, the effects of the HIF‑1α inhibitor 2‑methoxyestradiol and HIF‑1α small interfering RNA on the death and autophagy of microglial cells in vitro were investigated. These investigations revealed the suppression of autophagy, the decrease of cell viability and the increase of inflammatory cytokines results from HIF‑1α inhibition or HIF‑1α silencing. In conclusion, the results indicated that appropriate expression of HIF‑1α can ameliorate autophagic cell death of microglial cells associated with hypoxia, and may provide a novel therapeutic approach for SCI associated with microglial cell activation.
View Figures
View References

Related Articles

Journal Cover

April-2017
Volume 15 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang X, Ma J, Fu Q, Zhu L, Zhang Z, Zhang F, Lu N and Chen A: Role of hypoxia‑inducible factor‑1α in autophagic cell death in microglial cells induced by hypoxia. Mol Med Rep 15: 2097-2105, 2017
APA
Wang, X., Ma, J., Fu, Q., Zhu, L., Zhang, Z., Zhang, F. ... Chen, A. (2017). Role of hypoxia‑inducible factor‑1α in autophagic cell death in microglial cells induced by hypoxia. Molecular Medicine Reports, 15, 2097-2105. https://doi.org/10.3892/mmr.2017.6277
MLA
Wang, X., Ma, J., Fu, Q., Zhu, L., Zhang, Z., Zhang, F., Lu, N., Chen, A."Role of hypoxia‑inducible factor‑1α in autophagic cell death in microglial cells induced by hypoxia". Molecular Medicine Reports 15.4 (2017): 2097-2105.
Chicago
Wang, X., Ma, J., Fu, Q., Zhu, L., Zhang, Z., Zhang, F., Lu, N., Chen, A."Role of hypoxia‑inducible factor‑1α in autophagic cell death in microglial cells induced by hypoxia". Molecular Medicine Reports 15, no. 4 (2017): 2097-2105. https://doi.org/10.3892/mmr.2017.6277