GPR4 Knockout Improves the Neurotoxin-Induced, Caspase-Dependent Mitochondrial Apoptosis of the Dopaminergic Neuronal Cell

In Parkinson’s disease, mitochondrial oxidative stress-mediated apoptosis is a major cause of dopaminergic neuronal loss in the substantia nigra (SN). G protein-coupled receptor 4 (GPR4), previously recognised as an orphan G protein coupled-receptor (GPCR), has recently been claimed as a member of t...

Full description

Bibliographic Details
Main Authors: Md Ezazul Haque, Mahbuba Akther, Shofiul Azam, Dong-Kug Choi, In-Su Kim
Format: Article
Language:English
Published: MDPI AG 2020-10-01
Series:International Journal of Molecular Sciences
Subjects:
Online Access:https://www.mdpi.com/1422-0067/21/20/7517
_version_ 1797551251641073664
author Md Ezazul Haque
Mahbuba Akther
Shofiul Azam
Dong-Kug Choi
In-Su Kim
author_facet Md Ezazul Haque
Mahbuba Akther
Shofiul Azam
Dong-Kug Choi
In-Su Kim
author_sort Md Ezazul Haque
collection DOAJ
description In Parkinson’s disease, mitochondrial oxidative stress-mediated apoptosis is a major cause of dopaminergic neuronal loss in the substantia nigra (SN). G protein-coupled receptor 4 (GPR4), previously recognised as an orphan G protein coupled-receptor (GPCR), has recently been claimed as a member of the group of proton-activated GPCRs. Its activity in neuronal apoptosis, however, remains undefined. In this study, we investigated the role of GPR4 in the 1-methyl-4-phenylpyridinium ion (MPP<sup>+</sup>) and hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>)-treated apoptotic cell death of stably GPR4-overexpressing and stably GPR4-knockout human neuroblastoma SH-SY5Y cells. In GPR4-OE cells, MPP<sup>+</sup> and H<sub>2</sub>O<sub>2</sub> were found to significantly increase the expression levels of both mRNA and proteins of the pro-apoptotic Bcl-2-associated X protein (Bax) genes, while they decreased the anti-apoptotic B-cell lymphoma 2 (Bcl-2) genes. In addition, MPP<sup>+</sup> treatment activated Caspase-3, leading to the cleavage of poly (ADP-ribose) polymerase (PARP) and decreasing the mitochondrial membrane potential (ΔΨm) in GPR4-OE cells. In contrast, H<sub>2</sub>O<sub>2</sub> treatment significantly increased the intracellular calcium ions (Ca<sup>2+</sup>) and reactive oxygen species (ROS) in GPR4-OE cells. Further, chemical inhibition by NE52-QQ57, a selective antagonist of GPR4, and knockout of GPR4 by clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 decreased the Bax/Bcl-2 ratio and ROS generation, and stabilised the ΔΨm, thus protecting the SH-SY5Y cells from MPP<sup>+</sup>- or H<sub>2</sub>O<sub>2</sub>-induced apoptotic cell death. Moreover, the knockout of GPR4 decreased the proteolytic degradation of phosphatidylinositol biphosphate (PIP<sub>2</sub>) and subsequent release of the endoplasmic reticulum (ER)-stored Ca<sup>2+</sup> in the cytosol. Our results suggest that the pharmacological inhibition or genetic deletion of GPR4 improves the neurotoxin-induced caspase-dependent mitochondrial apoptotic pathway, possibly through the modulation of PIP<sub>2</sub> degradation-mediated calcium signalling. Therefore, GPR4 presents a potential therapeutic target for neurodegenerative disorders such as Parkinson’s disease.
first_indexed 2024-03-10T15:41:56Z
format Article
id doaj.art-f57775c1bcdd4c8eba19602779b6e4f2
institution Directory Open Access Journal
issn 1661-6596
1422-0067
language English
last_indexed 2024-03-10T15:41:56Z
publishDate 2020-10-01
publisher MDPI AG
record_format Article
series International Journal of Molecular Sciences
spelling doaj.art-f57775c1bcdd4c8eba19602779b6e4f22023-11-20T16:47:02ZengMDPI AGInternational Journal of Molecular Sciences1661-65961422-00672020-10-012120751710.3390/ijms21207517GPR4 Knockout Improves the Neurotoxin-Induced, Caspase-Dependent Mitochondrial Apoptosis of the Dopaminergic Neuronal CellMd Ezazul Haque0Mahbuba Akther1Shofiul Azam2Dong-Kug Choi3In-Su Kim4Department of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, KoreaDepartment of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, KoreaDepartment of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, KoreaDepartment of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, KoreaDepartment of Biotechnology, Research Institute of Inflammatory Disease (RID), College of Biomedical and Health Science, Konkuk University, Chungju 27478, KoreaIn Parkinson’s disease, mitochondrial oxidative stress-mediated apoptosis is a major cause of dopaminergic neuronal loss in the substantia nigra (SN). G protein-coupled receptor 4 (GPR4), previously recognised as an orphan G protein coupled-receptor (GPCR), has recently been claimed as a member of the group of proton-activated GPCRs. Its activity in neuronal apoptosis, however, remains undefined. In this study, we investigated the role of GPR4 in the 1-methyl-4-phenylpyridinium ion (MPP<sup>+</sup>) and hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>)-treated apoptotic cell death of stably GPR4-overexpressing and stably GPR4-knockout human neuroblastoma SH-SY5Y cells. In GPR4-OE cells, MPP<sup>+</sup> and H<sub>2</sub>O<sub>2</sub> were found to significantly increase the expression levels of both mRNA and proteins of the pro-apoptotic Bcl-2-associated X protein (Bax) genes, while they decreased the anti-apoptotic B-cell lymphoma 2 (Bcl-2) genes. In addition, MPP<sup>+</sup> treatment activated Caspase-3, leading to the cleavage of poly (ADP-ribose) polymerase (PARP) and decreasing the mitochondrial membrane potential (ΔΨm) in GPR4-OE cells. In contrast, H<sub>2</sub>O<sub>2</sub> treatment significantly increased the intracellular calcium ions (Ca<sup>2+</sup>) and reactive oxygen species (ROS) in GPR4-OE cells. Further, chemical inhibition by NE52-QQ57, a selective antagonist of GPR4, and knockout of GPR4 by clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 decreased the Bax/Bcl-2 ratio and ROS generation, and stabilised the ΔΨm, thus protecting the SH-SY5Y cells from MPP<sup>+</sup>- or H<sub>2</sub>O<sub>2</sub>-induced apoptotic cell death. Moreover, the knockout of GPR4 decreased the proteolytic degradation of phosphatidylinositol biphosphate (PIP<sub>2</sub>) and subsequent release of the endoplasmic reticulum (ER)-stored Ca<sup>2+</sup> in the cytosol. Our results suggest that the pharmacological inhibition or genetic deletion of GPR4 improves the neurotoxin-induced caspase-dependent mitochondrial apoptotic pathway, possibly through the modulation of PIP<sub>2</sub> degradation-mediated calcium signalling. Therefore, GPR4 presents a potential therapeutic target for neurodegenerative disorders such as Parkinson’s disease.https://www.mdpi.com/1422-0067/21/20/7517apoptosisneurodegenerationGPR4 receptorMPP<sup>+</sup>Parkinson’s diseaseCRISPR/cas9
spellingShingle Md Ezazul Haque
Mahbuba Akther
Shofiul Azam
Dong-Kug Choi
In-Su Kim
GPR4 Knockout Improves the Neurotoxin-Induced, Caspase-Dependent Mitochondrial Apoptosis of the Dopaminergic Neuronal Cell
International Journal of Molecular Sciences
apoptosis
neurodegeneration
GPR4 receptor
MPP<sup>+</sup>
Parkinson’s disease
CRISPR/cas9
title GPR4 Knockout Improves the Neurotoxin-Induced, Caspase-Dependent Mitochondrial Apoptosis of the Dopaminergic Neuronal Cell
title_full GPR4 Knockout Improves the Neurotoxin-Induced, Caspase-Dependent Mitochondrial Apoptosis of the Dopaminergic Neuronal Cell
title_fullStr GPR4 Knockout Improves the Neurotoxin-Induced, Caspase-Dependent Mitochondrial Apoptosis of the Dopaminergic Neuronal Cell
title_full_unstemmed GPR4 Knockout Improves the Neurotoxin-Induced, Caspase-Dependent Mitochondrial Apoptosis of the Dopaminergic Neuronal Cell
title_short GPR4 Knockout Improves the Neurotoxin-Induced, Caspase-Dependent Mitochondrial Apoptosis of the Dopaminergic Neuronal Cell
title_sort gpr4 knockout improves the neurotoxin induced caspase dependent mitochondrial apoptosis of the dopaminergic neuronal cell
topic apoptosis
neurodegeneration
GPR4 receptor
MPP<sup>+</sup>
Parkinson’s disease
CRISPR/cas9
url https://www.mdpi.com/1422-0067/21/20/7517
work_keys_str_mv AT mdezazulhaque gpr4knockoutimprovestheneurotoxininducedcaspasedependentmitochondrialapoptosisofthedopaminergicneuronalcell
AT mahbubaakther gpr4knockoutimprovestheneurotoxininducedcaspasedependentmitochondrialapoptosisofthedopaminergicneuronalcell
AT shofiulazam gpr4knockoutimprovestheneurotoxininducedcaspasedependentmitochondrialapoptosisofthedopaminergicneuronalcell
AT dongkugchoi gpr4knockoutimprovestheneurotoxininducedcaspasedependentmitochondrialapoptosisofthedopaminergicneuronalcell
AT insukim gpr4knockoutimprovestheneurotoxininducedcaspasedependentmitochondrialapoptosisofthedopaminergicneuronalcell