Elsevier

NeuroToxicology

Volume 85, July 2021, Pages 133-144
NeuroToxicology

Arsenic induces autophagy-dependent apoptosis via Akt inactivation and AMPK activation signaling pathways leading to neuronal cell death

https://doi.org/10.1016/j.neuro.2021.05.008Get rights and content

Highlights

  • As3+ induced a concomitant activation of apoptosis and autophagy in neuronal cell death.

  • Akt inactivation-regulated autophagy was involved in As3+-induced neuronal apoptosis.

  • AMPK activation played an important role in the As3+-triggered neuronal cell autophagy contributing to apoptosis.

Abstract

Inorganic arsenic (As3+), a well-known worldwide industrial and environmental pollutant, has been linked to neurodegenerative disorders (NDs). Autophagy plays an important role in controlling neuronal cell survival/death. However, limited information is available regarding the toxicological mechanism at the interplay between autophagy and As3+-induced neurotoxicity. The present study found that As3+ exposure induced a concomitant activation of apoptosis and autophagy in Neuro-2a cells, which was accompanied with the increase of phosphatidylserine exposure on outer membrane leaflets and apoptotic cell population, and the activation of caspase-3, -7, and PARP as well as the elevation of protein expressions of LC3-II, Atg-5, and Beclin-1, and the accumulation of autophagosome. Pretreatment of cells with autophagy inhibitor 3-MA, but not that of Z-VAD-FMK (a pan-caspase inhibitor), effectively prevented the As3+-induced autophagic and apoptotic responses, indicating that As3+-triggered autophagy was contributing to neuronal cell apoptosis. Furthermore, As3+ exposure evoked the dephosphorylation of Akt. Pretreatment with SC79, an Akt activator, could significantly attenuated As3+-induced Akt inactivation as well as autophagic and apoptotic events. Expectedly, inhibition of Akt signaling with LY294002 obviously enhanced As3+-triggered autophagy and apoptosis. Exposure to As3+ also dramatically increased the phosphorylation level of AMPKα. Pretreatment of AMPK inhibitor (Compound C) could markedly abrogate the As3+-induced phosphorylated AMPKα expression, and autophagy and apoptosis activation. Taken together, these results indicated that As3+ exerted its cytotoxicity in neuronal cells via the Akt inactivation/AMPK activation downstream-regulated autophagy-dependent apoptosis pathways, which ultimately lead to cell death. Our findings suggest that the regulation of Akt/AMPK signals may be a promising intervention to against As3+-induced neurotoxicity and NDs.

Introduction

Arsenic, the 33rd element of the Periodic Table of chemical elements, is a toxic metalloid and ubiquitous in the environment, which exists in air, water and soil (Mandal and Suzuki, 2002). Chronic exposure to As is still a global health problem in human (Ratnaike, 2003). Arsenic possesses both organic and inorganic forms that the inorganic form of arsenic such as trivalent arsenic (As3+), which is predominant in surface and groundwater reservoirs, is more toxic than the organic form (Gupta and Chatterjee, 2017). As3+ contamination supplied by natural deposits or industrial pollution is being reported from various countries, including India, Bangladesh, Myanmar, Mexico, the United States, and others (Camacho et al., 2011; Mochizuki et al., 2019; Natasha et al., 2020). Unfortunately, As3+ exposure via contaminated food chain, particularly at a nonlethal level in drinking water consumed over a period of time, is the major source of exposure to arsenic in human, resulting in the manifestations of toxicity in almost all of systems of the body (Mitra et al., 2020). Epidemiological studies have indicated a higher correlation between As3+ exposure and the development of neurodegenerative disorders (NDs), which could detect the higher levels of As3+ in the plasma and cerebrospinal fluid in mental health burden, Alzheimer's disease (AD), and Parkinson's disease (PD), leading to the neurobehavioral impairments and neuronal cell degeneration and death (Gong and O’Bryant, 2010; Miguel et al., 2015; Mitra et al., 2020; Mochizuki et al., 2019). In the experimental in vivo models, As3+ caused the serious neurological and neurobehavioral disorders as well as the neuronal cell apoptosis (Lu et al., 2014; Prakash et al., 2016; Wang et al., 2015a,b,c; Yen et al., 2011). Studies of Pellacani and Costa (2018) and Zhang et al. (2016) have also reported that exposure to toxic metals (such as MeHg, Pb, Cu, Cd, and Mn) can induce autophagy and neurotoxicity, which are correlated with the development of NDs. A few studies have indicated that As3+ exposure can induce autophagy in developing mouse brain, accompanied with the disappearances of axons, irregular shrinkage of cells, and karyolysis, pyknosis, and loss of neurons (Manthari et al., 2018a; and 2018b). However, limited information is available regarding the toxicological mechanism at the interplay between autophagy and As3+-induced neurotoxicity/neuronal cell apoptosis.

Autophagy is also known as type II programmed cell death, which involves a sequential set of programs including double membrane formation, elongation, vesicle maturation and finally delivery of the targeted materials to the lysosome (Ghavami et al., 2014; Gump and Thorburn, 2011). Autophagy plays a crucial role in maintaining cellular homeostasis and physiological processes that protects cells from environmental or toxic stress-induced insults, such as aggregated and misfolded proteins and damaged organelles, which induces cellular stress, failure, and death (Doherty and Baehrecke, 2018; Schneider and Cuervo, 2014). Autophagy occurs in almost all cell types and is useful to maintain cellular homeostasis, allowing cellular differentiation, growth control, cell defense, tissue remodeling, and adaptation for adverse environments (Doherty and Baehrecke, 2018; Pellacani and Costa, 2018). Both pro-survival and pro-death roles of autophagy have been proposed in the pathophysiology in many human diseases, including cancer, metabolic dysfunction, and NDs (Choi et al., 2013; Das et al., 2012). Growing studies have implicated that disrupted/defected autophagy is one of the factors contributing to mammalian cell death (particularly in neuronal cells), pointing to autophagic dysfunction as a potential pathogenesis in NDs (Martinez-Vicente, 2015; Nixon and Yang, 2012; Schneider and Cuervo, 2014). For examples, the abundant autophagosome vesicles have been found in AD brains and autophagosome-like structures have been observed in the substantia nigra (SN) neurons of PD patients (Anglade et al., 1997; Boland et al., 2008). Recently, the alteration in autophagy process upon exposure to environmental pollutants/chemicals-induced neurotoxicity has been indicated to associate with neurodegeneration (Manthari et al., 2018b; Song et al., 2019).

Akt (protein kinase B), a serine/threonine protein kinase, plays an essential role in the regulation of multiple cellular functions including cell differentiation, proliferation, survival, and apoptosis (Franke et al., 2003). The regulation/dysregulation mechanism of Akt signaling is an important factor involved in several life-threatening diseases, including cancer, diabetes, and NDs (Xu et al., 2020). Studies have shown that Akt activity and Akt levels are decreased in the brains of AD and PD patients, linking Akt signal and NDs development (Anglade et al., 1997; Liu et al., 2011; Malagelada et al., 2008). On the other hand, adenosine monophosphate-activated protein kinase (AMPK), a highly conserved serine/threonine protein kinase, is considered to be an importantly intracellular sensor and regulator of metabolic homeostasis (Spasic et al., 2009). In neurons, AMPK plays a critical role in the energy and functional maintenance, survival, and apoptosis (Chen et al., 2010; Spasic et al., 2009). Over-expression of AMPK signaling has been detected in several brain diseases, including NDs, suggesting a correlation between AMPK activation and NDs (Domise and Vingtdeux, 2016). In ischemic brain injury, the AMPK activation-related neuronal cell death has been detected, which could be obviously attenuated by the inhibition of AMPK activity (Li et al., 2010; Nakatsu et al., 2008; Xu et al., 2014). The increasing studies have shown that the diminished Akt phosphorylation and/or the activation of AMPK signal contribute to neuronal cell apoptosis by exposure to chemicals (Chung et al., 2019; Eom et al., 2016; Xu et al., 2014). Furthermore, it has also been reported that Akt inhibits autophagy and AMPK stimulates apoptotic and autophagic signals under the pathological processes of cell damage (Maiese, 2016).

Even though both Akt and AMPK signals have been explored in chemicals-induced neuronal autophagic/apoptotic cell death (He et al., 2017; Zhao et al., 2019), the roles of both signals in As3+-induced neurotoxicity linked to autophagy/apoptosis pathway are still unclear. In this study, we aimed to investigate how As3+ influences both Akt and AMPK-regulated autophagy and apoptosis pathways contributing to neuronal cell death.

Section snippets

Materials

Unless specified, otherwise, all chemicals (including As2O3 (As3+)) and laboratory plastic wares were purchased from Sigma-Aldrich (St. Louis, MO, USA) and Falcon Labware (Bectone-Diskinson, Franlin Lakes, NJ, USA), respectively. Dulbecco’s modified Eagle’s medium (DMEM), fetal bovine serum (FBS), and antibiotics were purchased from Gibco/Invitrogen (Thermo Fisher Scientific, Waltham, MA, USA). Mouse- or rabbit- monoclonal antibodies specific for caspase-3, -7, PARP, LC3-I/II, Atg-5, beclin-1,

Inorganic arsenic (As3+) induces concentration- and time-dependent apoptosis in Neuro-2a cells

We first examined the cytotoxic effect of As3+ in Neuro-2a cells by MTT assay. Treatment of cells with 1–10 μM As3+ for 24 h significantly reduced the number of viable cells in a concentration-dependent manner (1 μM, 93.7 ± 2.5 % of control; 3 μM, 75.2 ± 6.1 % of control; 5 μM, 50.1 ± 3.62 % of control; 7 μM, 38.7 ± 5.4 % of control; 10 μM, 22.4 ± 0.9 % of control) (Fig. 1A), which was consistency with FDA staining assay. The median effective concentration (EC50) of As3+ in Neuro-2a cells was

Discussion

The present study demonstrated for the first time that As3+ induced autophagy-dependent apoptosis-triggered cell death in neuronal cells, which could be significantly reversed by autophagy inhibitor 3-MA. Furthermore, As3+ was capable of inducing the inactivation of Akt and the activation of AMPK signals, which linked to autophagy and apoptosis responses in neuronal cells. These findings highlight that Akt/AMPK signals-regulated autophagy-dependent apoptotic mechanism is involved in the As3+

CRediT authorship contribution statement

All authors approved the final version to be published. Study conception and design: Kuan-I Lee and Ya-Wen Chen. Acquisition of data, and writing original draft preparation: Shih-Chang Fu, Jhe-Wei Lin, Jui-Ming Liu, Shing-Hwa Liu, and Kai-Min Fang. Analysis and interpretation of data: Shih-Chang Fu, Jhe-Wei Lin, Shing-Hwa Liu, Ren-Jun Hsu, and Chun-Fa Huang. Provided reagents and technical support: Jui-Ming Liu, Chin-Chuan Su, and Chin-Ching Wu. Wrote, reviewed, and edited the manuscript:

Declaration of Competing Interest

The authors report no declarations of interest.

Acknowledgments

This work was supported by the grants from the Ministry of Science and Technology, Taiwan (MOST 109-2320-B-039-039; MOST 108-2320-B-039-025; 104-2815-C-039-007-B), the Buddhist Tzuchi Medical Foundation of the Taichung Tzu chi Hospital, Taiwan (TTCRD 108-09), the Changhua Christian Hospital, Taiwan (110-CCH-IRP-057), the Taoyuan General Hospital, Ministry of Health and Welfare, Taiwan (Grants No. 107012; 109018), and the China Medical University, Taiwan (CMU109-S-40).

References (85)

  • X. He et al.

    An autophagic mechanism is involved in the 6-hydroxydopamine-induced neurotoxicity in vivo

    Toxicol. Lett.

    (2017)
  • C.F. Huang et al.

    Roles of ERK/Akt signals in mitochondria-dependent and endoplasmic reticulum stress-triggered neuronal cell apoptosis induced by 4-methyl-2,4-bis(4-hydroxyphenyl)pent-1-ene, a major active metabolite of bisphenol A

    Toxicology

    (2021)
  • P. Jiang et al.

    Adenosine monophosphate-activated protein kinase overactivation leads to accumulation of alpha-synuclein oligomers and decrease of neurites

    Neurobiol. Aging

    (2013)
  • K.I. Lee et al.

    Silica nanoparticles induce caspase-dependent apoptosis through reactive oxygen species-activated endoplasmic reticulum stress pathway in neuronal cells

    Toxicol. In Vitro

    (2020)
  • J. Li et al.

    Adenosine monophosphate activated protein kinase inhibition is protective in both sexes after experimental stroke

    Neurosci. Lett.

    (2010)
  • T.H. Lu et al.

    Arsenic induces reactive oxygen species-caused neuronal cell apoptosis through JNK/ERK-mediated mitochondria-dependent and GRP 78/CHOP-regulated pathways

    Toxicol. Lett.

    (2014)
  • B.K. Mandal et al.

    Arsenic round the world: a review

    Talanta

    (2002)
  • M. Martinez-Vicente

    Autophagy in neurodegenerative diseases: from pathogenic dysfunction to therapeutic modulation

    Semin. Cell Dev. Biol.

    (2015)
  • Y. Nakatsu et al.

    Activation of AMP-activated protein kinase by tributyltin induces neuronal cell death

    Toxicol. Appl. Pharmacol.

    (2008)
  • C. Pellacani et al.

    Role of autophagy in environmental neurotoxicity

    Environ. Pollut.

    (2018)
  • M.M. Rahman et al.

    Ameliorative effects of selenium on arsenic-induced cytotoxicity in PC12cells via modulating autophagy/apoptosis

    Chemosphere

    (2018)
  • R. Roy et al.

    Zinc oxide nanoparticles induce apoptosis by enhancement of autophagy via PI3K/Akt/mTOR inhibition

    Toxicol. Lett.

    (2014)
  • J.L. Schneider et al.

    Autophagy and human disease: emerging themes

    Curr. Opin. Genet. Dev.

    (2014)
  • Y.C. Teng et al.

    Role of autophagy in arsenite-induced neurotoxicity: the involvement of alpha-synuclein

    Toxicol. Lett.

    (2015)
  • G. Tovilovic et al.

    Arylpiperazine-mediated activation of Akt protects SH-SY5Y neuroblastoma cells from 6-hydroxydopamine-induced apoptotic and autophagic death

    Neuropharmacology

    (2013)
  • L. Wang et al.

    Conditional inactivation of Akt three isoforms causes tau hyperphosphorylation in the brain

    Mol. Neurodegener.

    (2015)
  • Y. Xu et al.

    Activation of AMPK and inactivation of Akt result in suppression of mTOR-mediated S6K1 and 4E-BP1 pathways leading to neuronal cell death in in vitro models of Parkinson’s disease

    Cell. Signal.

    (2014)
  • L. Xu et al.

    Role of autophagy in sevoflurane-induced neurotoxicity in neonatal rat hippocampal cells

    Brain Res. Bull.

    (2018)
  • H. Zhang et al.

    Cadmium results in accumulation of autophagosomes-dependent apoptosis through activating Akt-impaired autophagic flux in neuronal cells

    Cell. Signal.

    (2019)
  • M. Zhao et al.

    Mitochondrial calcium dysfunction contributes to autophagic cell death induced by MPP(+) via AMPK pathway

    Biochem. Biophys. Res. Commun.

    (2019)
  • P. Anglade et al.

    Apoptosis and autophagy in nigral neurons of patients with Parkinson’s disease

    Histol. Histopathol.

    (1997)
  • F. Arnoldi et al.

    Rotavirus increases levels of lipidated LC3 supporting accumulation of infectious progeny virus without inducing autophagosome formation

    PLoS One

    (2014)
  • A.M. Belle et al.

    Evaluation of in vitro neuronal platforms as surrogates for in vivo whole brain systems

    Sci. Rep.

    (2018)
  • B. Boland et al.

    Autophagy induction and autophagosome clearance in neurons: relationship to autophagic pathology in Alzheimer’s disease

    J. Neurosci.

    (2008)
  • Y. Chen et al.

    Antidiabetic drug metformin (GlucophageR) increases biogenesis of Alzheimer’s amyloid peptides via up-regulating BACE1 transcription

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • P. Chen et al.

    Metals and neurodegeneration

    F1000 Faculty Rev.

    (2016)
  • H. Chi et al.

    Neuronal cell death mechanisms in major neurodegenerative diseases

    Int. J. Mol. Sci.

    (2018)
  • A.M. Choi et al.

    Autophagy in human health and disease

    N. Engl. J. Med.

    (2013)
  • G. Das et al.

    Regulation and function of autophagy during cell survival and cell death

    Cold Spring Harb. Perspect. Biol.

    (2012)
  • M. Degtyarev et al.

    Akt inhibition promotes autophagy and sensitizes PTEN-null tumors to lysosomotropic agents

    J. Cell Biol.

    (2008)
  • J. Doherty et al.

    Life, death and autophagy

    Nat. Cell Biol.

    (2018)
  • M. Domise et al.

    AMPK in neurodegenerative diseases

    Exp. Suppl.

    (2016)
  • Cited by (0)

    1

    These authors contributed equally to this study.

    View full text