1. Academic Validation
  2. Acute Methylmercury Exposure and the Hypoxia-Inducible Factor-1α Signaling Pathway under Normoxic Conditions in the Rat Brain and Astrocytes in Vitro

Acute Methylmercury Exposure and the Hypoxia-Inducible Factor-1α Signaling Pathway under Normoxic Conditions in the Rat Brain and Astrocytes in Vitro

  • Environ Health Perspect. 2019 Dec;127(12):127006. doi: 10.1289/EHP5139.
Jie Chang 1 Bobo Yang 1 Yun Zhou 1 Changsheng Yin 1 2 Tingting Liu 1 Hai Qian 1 Guangwei Xing 1 Suhua Wang 1 Fang Li 1 Yubin Zhang 3 Da Chen 4 Michael Aschner 5 Rongzhu Lu 1 6
Affiliations

Affiliations

  • 1 Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, China.
  • 2 Institute of Life Sciences, Jiangsu University, Zhenjiang, China.
  • 3 Department of Occupational Health and Toxicology, School of Public Health, Fudan University, Shanghai, China.
  • 4 School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, China.
  • 5 Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA.
  • 6 Center for Experimental Research, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, China.
Abstract

Background: As a ubiquitous environmental pollutant, methylmercury (MeHg) induces toxic effects in the nervous system, one of its main targets. However, the exact mechanisms of its neurotoxicity have not been fully elucidated. Hypoxia-inducible factor-1α (HIF-1α), a transcription factor, plays a crucial role in adaptive and cytoprotective responses in cells and is involved in cell survival, proliferation, Apoptosis, inflammation, angiogenesis, glucose metabolism, erythropoiesis, and other physiological activities.

Objectives: The aim of this study was to explore the role of HIF-1α in response to acute MeHg exposure in rat brain and primary cultured astrocytes to improve understanding of the mechanisms of MeHg-induced neurotoxicity and the development of effective neuroprotective strategies.

Methods: Primary rat astrocytes were treated with MeHg (0-10μM) for 0.5h. Cell Proliferation and Cytotoxicity were assessed with a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl diphenyltetrazolium bromide (MTT) assay and a Lactate Dehydrogenase (LDH) release assay, respectively. Reactive Oxygen Species (ROS) levels were analyzed to assess the level of oxidative stress using 2',7'-dichlorofluorescin diacetate (DCFH-DA) fluorescence. HIF-1α, and its downstream proteins, glucose transporter 1 (GLUT-1), erythropoietin (EPO), and vascular endothelial growth factor A (VEGF-A) were analyzed by means of Western blotting. Real-Time PCR was used to detect the expression of HIF-1α mRNA. Pretreatment with protein synthesis inhibitor (CHX), Proteasome Inhibitor (MG132), or proline hydroxylase inhibitor (DHB) were applied to explore the possible mechanisms of HIF-1α inhibition by MeHg. To investigate the role of HIF-1α in MeHg-induced neurotoxicity, cobalt chloride (CoCl2), 2-methoxyestradiol (2-MeOE2), small interfering RNA (siRNA) transfection and adenovirus overexpression were used. Pretreatment with N-acetyl-L-cysteine (NAC) and vitamin E (Trolox) were used to investigate the putative role of oxidative stress in MeHg-induced alterations in HIF-1α levels. The expression of HIF-1α and related downstream proteins was detected in adult rat brain exposed to MeHg (0-10mg/kg) for 0.5h in vivo.

Results: MeHg caused lower cell proliferation and higher cytotoxicity in primary rat astrocytes in a time- and concentration-dependent manner. In comparison with the control cells, exposure to 10μM MeHg for 0.5h significantly inhibited the expression of astrocytic HIF-1α, and the downstream genes GLUT-1, EPO, and VEGF-A (p<0.05), in the absence of a significant decrease in HIF-1α mRNA levels. When protein synthesis was inhibited by CHX, MeHg promoted the degradation rate of HIF-1α. MG132 and DHB significantly blocked the MeHg-induced decrease in HIF-1α expression (p<0.05). Overexpression of HIF-1α significantly attenuated the decline in MeHg-induced cell proliferation, whereas the inhibition of HIF-1α significantly increased the decline in cell proliferation (p<0.05). NAC and Trolox, two established antioxidants, reversed the MeHg-induced decline in HIF-1α protein levels and the decrease in cell proliferation (p<0.05). MeHg suppressed the expression of HIF-1α and related downstream target proteins in adult rat brain.

Discussion: MeHg induced a significant reduction in HIF-1α protein by activating proline hydroxylase (PHD) and the ubiquitin Proteasome system (UPS) in primary rat astrocytes. Additionally, ROS scavenging by antioxidants played a neuroprotective role via increasing HIF-1α expression in response to MeHg toxicity. Moreover, we established that up-regulation of HIF-1α might serve to mitigate the acute toxicity of MeHg in astrocytes, affording a novel therapeutic target for future exploration. https://doi.org/10.1289/EHP5139.

Figures
Products