Elsevier

Environmental Research

Volume 177, October 2019, 108579
Environmental Research

Oxidative stress and related gene expression effects of cyfluthrin in human neuroblastoma SH-SY5Y cells: Protective effect of melatonin

https://doi.org/10.1016/j.envres.2019.108579Get rights and content

Highlights

  • . Cyfluthrin neurotoxicity in human neuroblastoma SH-SY5Y cells.

  • . Cyfluthrin decreased MTT cell viability.

  • . Cyfluthrin increased MDA, NO and ROS production and decreased NQO1 activity.

  • . Cyfluthrin affected oxidative stress signaling pathways.

  • . Melatonin showed effective protection against cyfluthrin-induced oxidative stress.

Abstract

This study was designed to assess oxidative stress induction in human neuroblastoma SH-SY5Y cells in response to cyfluthrin exposure. Cell viability MTT assay was carried out to assess cyfluthrin cytotoxicity; IC30 and IC50 values for cyfluthrin were calculated to be 4.81 ± 0.92 μM and 19.39 ± 3.44 μM, respectively. Cyfluthrin induced a significant increase in ROS generation, lipid peroxides measured as malondialdehyde (MDA) and nitric oxide (NO) production and a significant decrease in NQO1 activity. The antioxidant activity of melatonin (MEL), Trolox, N-acetylcysteine (NAC) and Sylibin against cyfluthrin-induced oxidative stress was examined. Cyfluthrin increased significantly gene expressions of apoptosis, proinflammation and oxidative stress (Bax, Bcl-2, Casp-3, BNIP3, AKT1, p53, APAF1, NFκB1, TNFα and Nrf2) mediators. In the most genes, the mRNA levels induced by cyfluthrin were partially reduced by MEL (1 μM). Cyfluthrin effects on gene expression profiling of oxidative stress pathway by Real-Time PCR array analysis showed that of the 84 genes examined, (fold change > 1.5) changes in mRNA levels were detected in 31 genes: 13 upregulated and 18 down-regulated. A fold change >3.0 fold was observed on upregulated CYBB, DUOX1, DUOX2, AOX1, BNIP3, HSPA1A, NOS2, and NQO1 genes. The greater fold change reversion (2.5 fold) by MEL (1 μM) was observed on cyfluthrin-upregulated CYBB, AOX1, BNIP3 and NOS2 genes. These results demonstrated that oxidative stress is a key element in cyfluthrin induced neurotoxicity as well as MEL may play a role in reducing cyfluthrin-induced oxidative stress.

Introduction

Pyrethroids are a class of synthetic, neurotoxic, insecticides that are structurally based on purified extracts (pyrethrins) of Chrysanthemum sp. flowers. Characteristically, pyrethroids are described as phenoxybenzoic and cyclopropane moieties joined by an ester bond. Pyrethroids are frequently grouped according to the absence (Type I) or presence (Type II) of a cyano group at the α-carbon of the phenoxybenzoic constituent. The principal target site for pyrethroids is defined as the voltage-dependent sodium channel in the neuronal membrane leading to depolarization and hyperexcitation of the nervous system (Narahashi et al., 1998; Soderlund et al., 2002). This group of compounds has also been shown to act on isoforms of voltage sensitive calcium channels (Hildebrand et al., 2004), thereby contributing to the release of neurotransmitters and hence leading to pyrethroid induced toxicity (Symington and Clark, 2005). Pyrethroids are classified as Type I or Type II based on their chemical structure and also based on the toxic syndrome than they produce in rodents. Type I pyrethroids are associated to hyper-excitation and fine tremors (T-syndrome), and Type II pyrethroids are associated to a more complex syndrome, including clonic seizures (choreoathetosis) and salivation (CS syndrome) (Verschoyle and Aldridge, 1980; Aldridge, 1990). Some pyrethroids produce tremors and salivation, classified as the intermediate TS-syndrome (Soderlund et al., 2002; Soderlund, 2012). Types I and II pyrethroids are extensively applied to control pests in residential and agricultural settings, to treat head lice and scabies in humans and fleas in pets, for public health vector control, and for disinfection of commercial aircrafts (Anadón et al., 2009, Anadón et al., 2013a; USEPA, 2013, 2017). Data from in vivo and in vitro studies have shown that pyrethroids undergo extensive metabolism by carboxylesterases and CYP (Godin et al., 2007; Scollon et al., 2009). However, pyrethroids could interact with the normal metabolism of drugs and xenobiotics and some pyrethroids are found to induce CYP enzyme activities (Yang et al., 2009; Anadón et al., 2013b; Martínez et al., 2018). Moreover, there is evidence suggesting that the detoxifying enzymes of pyrethroids are present at lower levels during fetal and early postnatal development than they are later in life (Cantalamessa, 1993; Sheets, 2000). Previous experiments showed that the toxicity of pyrethroids increases as the age of the animal decreases, so that neonates appear highly sensitive to pyrethroid exposure (Cantalamessa, 1993). A comprehensive review summarizing the existing 22 studies of the developmental neurotoxicity of pyrethroids suggests that pyrethroids may exert developmental neurotoxicity (Shafer et al., 2005). Developmental neurotoxicity involves alterations in behavior, neurohistology, neurochemistry and/or dysmorphology of the central nervous system occurring in the adult age, as a result of neonatal exposure to pyrethroids. Learning and memory impairments in pyrethroid-treated rats and neurobehavioral changes could also be attributed to increased oxidative stress (Liu et al., 2003; Ansari et al., 2012a, b; Nasuti et al., 2007, 2013; Gargouri et al., 2018). Induction of oxidative stress is an important mechanism in pesticide induced toxicities, being the major endpoints damage to DNA, proteins and membrane lipids (Banerjee et al., 2001; Rehman et al., 2006; Tiwari et al., 2010; Romero et al., 2012, 2017). Because, the increased use of pyrethroids has made human exposure almost inevitable, further studies are necessary to assess the neurotoxicity of pyrethroids. It is accepted that animal testing should be reduced, refined or replaced as far as it is practicably possible. Alternative in vitro testing strategies are required, in particular for screening, as part of a tired testing scheme, and routine testing (Anadón et al., 2013c).

Cyfluthrin, a commonly used Type II pyrethroid, was selected for the current investigation. Cyfluthrin, a mixture of four diastereoisomeric pairs of enantiomers (I, II, III, IV) was first registered for use in the United States in 1987 (USEPA, 1987), frequently it is used in veterinary medicine, agriculture against grasshoppers and pests, industrial and residential settings, and public health and, in some countries for the protection of stored products (Ritter and Chappel, 1997; FAO, 1999; Surgan et al., 2002). Despite beneficial roles in agricultural and household products, recent studies in Wistar rats showed that oral exposure to cyfluthrin induced hepatic and renal CYP2E, CYP1A and CYP4A subfamilies, and also increased the β-oxidation of palmitoyl-coenzyme A and carnitine acetyltransferase activity, supporting cyfluthrin classification as a peroxisome proliferator with possible implications in oxidative stress (Anadón et al., 2013b). Also, alteration of GPx and AchE activities in liver and kidney of Wistar rats following intraperitoneal treatment of cyfluthrin has been described (Yilmaz et al., 2015). Cyfluthrin is readily absorbed by the oral route, enters the brain, accumulates in significant quantity and therefore it is appropriate that cyfluthrin affects the CNS function of the non-target organisms (Rodríguez et al., 2018). Because cyfluthrin is one of the more used pyrethroid insecticides worldwide (Surgan et al., 2002), and studies describing mechanisms underlying neurotoxicity of this insecticide (Mense et al., 2006; Eraslan et al., 2007; Rodríguez et al., 2016) are limited, in the present study the possible link between oxidative stress pathways and cyfluthrin neurotoxicity, was investigated. This in vitro study was undertaken (i) to characterize the concentration-dependent cytotoxicity of cyfluthrin using cell viability MTT assay and to determine the protective role of selected antioxidant substances (MEL, Trolox, NAC and Sylibin) on MTT reduction, lipid peroxidation, NO and ROS production and NQO1 activity and (ii) to evaluate gene expressions of apoptosis, proinflammation and oxidative stress (Bax, Bcl-2, Casp-3, BNIP3, AKT1, p53, APAF1, NFκB1, TNFα and Nrf2) mediators as well as to examine by Real-Time PCR array the expression of key genes related to oxidative stress after cyfluthrin and cyfluthrin plus MEL exposure. In the present study, human dopaminergic neuroblastoma cell line (SH-SY5Y) was used as an in vitro model to determine cytotoxic mechanisms of cyfluthrin. The human dopaminergic neuroblastoma cell line, SH-SY5Y, is a commonly used cell line in studies related to neurotoxicity, oxidative stress, and neurodegenerative diseases (Pahlman, 1990; Krishna et al., 2014).

Section snippets

Chemicals and reagents

The test substance, cyfluthrin [cyano (4-fluoro-3-phenoxyphenyl) methyl-3-(2,2-dichloroethenyl)-2,2- dimethylcyclopropanecarboxylate] was provided by Bayer AG (Wuppertal-Elberfeld, Germany) (CAS no: 68359-37-5), ≥97.5% purity, molecular weight 434.3 g/mol.

The compounds 2′,7′-dichlorofluorescin diacetate (DCFH), 3-[4,5 dimethylthiazol-2-yl]-2,5-diphenyl-tetrazolium bromide (MTT), melatonin (N-acetyl-5-methoxytryptamine) (MEL), N-acetyl-cysteine (NAC), Trolox

Cyfluthrin effect on SH-SY5Y cell viability (MTT assay)

In order to evaluate cell survival we used MTT assay. As shown in Fig. 1A, the difference between data of vehicle-treated cells (0.1% DMSO) and control cells was not statistically significant. A 24 h incubation period with cyfluthrin at increasing concentrations (0.01–25 μM) reduced cell viability in a concentration-dependent manner compared with vehicle-treated cells (negative control) (Fig. 1A). The IC30 and IC50 values for cyfluthrin was calculated to be 4.81 ± 0.92 μM and 19.39 ± 3.44 μM,

Discussion

In the environment, individuals are exposed to a multitude of environmental toxicants. Increased production and application of pyrethroid insecticides in recent years have raised serious concerns on potential risks of exposure. Cyfluthrin is a Type II synthetic pyrethroid with a broad spectrum of insecticidal and acaricidal activity used to control wide range of insect pests in a variety of applications (Anadón et al., 2013a). This compound, alike other synthetic pyrethroids Type II, is a

Conclusion

In conclusion, our work includes new data suggesting that oxidative stress mechanisms play a fundamental role in the toxicity induced by the insecticide pyrethroid cyfluthrin. Our work adds information on the possible involvement of several genes mainly AKT1, BNIP3, Nrf2, CYBB, DUOX1, DUOX2, AOX1, and NOS2 in the cyfluthrin toxicity in neuronal cells. This investigation provides a list of genes and pathways for further detailed studies and provides a framework for the observation of possible

Conflicts of interest

The authors declare that there are no conflicts of interest.

Acknowledgments

This work was supported by Project Ref. RTA2015-00010-C03-03 from Ministerio de Economía, Industria y Competitividad, Spain.

References (96)

  • Q. Lu et al.

    Deltamethrin toxicity: a review of oxidative stress and metabolism

    Environ. Res.

    (2019)
  • K. Maeda et al.

    Aldehyde oxidase 1 gene is regulated by Nrf2 pathway

    Gene

    (2012)
  • M.A. Martínez et al.

    Pyrethroid insecticide lambda-cyhalothrin induces hepatic cytochrome P450 enzymes, oxidative stress and apoptosis in rats

    Sci. Total Environ.

    (2018)
  • A.L. McCormack et al.

    Environmental risk factors and Parkinson's disease: selective degeneration of nigral dopaminergic neurons caused by the herbicide paraquat

    Neurobiol. Dis.

    (2002)
  • M.J. Morgan et al.

    Reactive oxygen species in TNFα-induced signaling and cell death

    Mol. Cells

    (2010)
  • C. Nasuti et al.

    Dopaminergic system modulation, behavioral changes, and oxidative stress after neonatal administration of pyrethroids

    Toxicology

    (2007)
  • C. Nasuti et al.

    Effects of early life permethrin exposure on spatial working memory and on monoamine levels in different brain areas of pre-senescent rats

    Toxicology

    (2013)
  • K.M. Nicholson et al.

    The protein kinase B/Akt signalling pathway in human malignancy

    Cell. Signal.

    (2002)
  • K. Prabhakaran et al.

    Upregulation of BNIP3 and translocation to mitochondria mediates cyanide-induced apoptosis in cortical cells

    Neuroscience

    (2007)
  • C. Ramakers et al.

    Assumption-free analysis of quantitative real-time polymerase chain reaction (PCR) data

    Neurosci. Lett.

    (2003)
  • H. Rehman et al.

    The modulatory effect of deltamethrin on antioxidants in mice

    Clin. Chim. Acta

    (2006)
  • J.L. Rodríguez et al.

    Effects of exposure to pyrethroid cyfluthrin on serotonin and dopamine levels in brain regions of male rats

    Environ. Res.

    (2016)
  • J.L. Rodríguez et al.

    Bioavailability and nervous tissue distribution of pyrethroid insecticide cyfluthrin in rats

    Food Chem. Toxicol.

    (2018)
  • A. Romero et al.

    Cytotoxicity induced by deltamethrin and its metabolites in SH-SY5Y cells can be differentially prevented by selected antioxidants

    Toxicol. In Vitro

    (2012)
  • A. Romero et al.

    Evidence for dose-additive effects of a Type II pyrethroid mixture in vitro assessment

    Environ. Res.

    (2015)
  • D. Ross et al.

    NAD(P)H: quinone oxidoreductase 1 (NQO1): chemoprotection, bioactivation, gene regulation and genetic polymorphisms

    Chem. Biol. Interact.

    (2000)
  • T. Seredenina et al.

    Evaluation of NADPH oxidases as drug targets in a mouse model of familial amyotrophic lateral sclerosis

    Free Radic. Biol. Med.

    (2016)
  • J. Schumann et al.

    Silibinin protects mice from T cell-dependent liver injury

    J. Hepatol.

    (2003)
  • D.M. Soderlund et al.

    Mechanisms of pyrethroid neurotoxicity: implications for cumulative risk assessment

    Toxicology

    (2002)
  • S.B. Symington et al.

    Action of deltamethrin on voltage-sensitive calcium channels in rat brain

    Pestic. Biochem. Physiol.

    (2005)
  • H. Wang et al.

    Quantifying cellular oxidative stress by dichlorofluorescein assay using microplate reader

    Free Radical Biol. Med.

    (1999)
  • L. Xin et al.

    Development of stable HSPA1A promoter-driven luciferase reporter HepG2 cells for assessing the toxicity of organic pollutants present in air

    Cell Stress Chaperones

    (2012)
  • D. Yang et al.

    Pyrethroid insecticides: isoform dependent hydrolysis induction of cytochrome P450 3A4 and evidence on the involvement of the pregnane X receptor

    Toxicol. Appl. Pharmacol.

    (2009)
  • Y. Yang et al.

    An overview of the molecular mechanisms and novel roles of Nrf2 in neurodegenerative disorders

    Cytokine Growth Factor Rev.

    (2015)
  • F. Yin et al.

    Silibinin: a novel inhibitor of Aβ aggregation

    Neurochem. Int.

    (2011)
  • Y.H. Yook et al.

    Nitric oxide induces BNIP3 expression that causes cell death in macrophages

    Biochem. Biophys. Res. Commun.

    (2004)
  • W.,N. Aldridge

    An assessment of the toxicological properties of pyrethroids and their neurotoxicity

    Crit. Rev. Toxicol.

    (1990)
  • A. Anadón et al.

    Pyrethrins and synthetic pyrethroids: use in veterinary medicine

  • A. Anadón et al.

    The role of in vitro methods as alternatives to animals in toxicity testing

    Expert Opin. Drug Metabol. Toxicol.

    (2013)
  • R.W. Ansari et al.

    Cholinergic dysfunctions and enhanced oxidative stress in the neurobehavioral toxicity of lambda-cyhalothrin in developing rats

    Neurotox. Res.

    (2012)
  • B.D. Banerjee et al.

    Pesticide-induced oxidative stress: perspectives and trends

    Rev. Environ. Health

    (2001)
  • F. Bauche et al.

    In vitro regulation an inducible-type NO synthase in the rat seminiferous tubule cells

    Biol. Reprod.

    (1998)
  • K. Bedard et al.

    The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology

    Physiol. Rev.

    (2007)
  • F. Cantalamessa

    Acute toxicity of two pirethroid, permethrin, and cypermethrin in neonatal and adult rats

    Arch. Toxicol.

    (1993)
  • T.T. Cao et al.

    Increased nuclear factor-erythroid 2 p45-related factor 2 activity protects SH-SY5Y cells against oxidative damage

    J. Neurochem.

    (2005)
  • G. Cardamone et al.

    Genetic association and altered gene expression of CYBB in multiple sclerosis patients

    Biomedicines

    (2018)
  • L.C. Chen et al.

    Ferrous citrate up-regulates the NOS2 through nuclear translocation of NFkB induced by free radicals generation in mouse cerebral endothelial cells

    PLoS One

    (2012)
  • E.Y. Choi et al.

    Melatonin inhibits Prevotella intermedia lipopolysaccharide-induced production of nitric oxide and interleukin-6 in murine macrophages by suppressing NF-κB and STAT-1 activity

    J. Pineal Res.

    (2011)
  • Cited by (26)

    • A systematic review of mechanistic studies on the relationship between pesticide exposure and cancer induction

      2022, Toxicology and Applied Pharmacology
      Citation Excerpt :

      More than that, both apoptosis and autophagy gene expression (BCL2, ATP6V1G2, CASP9, FAS, SPATA2, GADD45A, SYCP2, ATG7, SNCA, NFKB1, ULK1, and JPH3) had been raised >3-fold after pyrethroids (alpha-cypermethrin) exposure (Romero et al., 2017). Also, pyrethroids insecticides (cyfluthrin and toosendanin), OC pesticides (endosulfan), biopesticides (spinosad), rotenone, and glyphosate-based herbicides under the brand name Roundup increased apoptosis by regulating the expression of Bax, BCL-2, casp-3, BNTP3, akt1, p53, APAF1 genes (Martínez et al., 2019; Xu et al., 2018; Hao et al., 2019; Zhang et al., 2019; Gao et al., 2019; Xiao et al., 2020). p53 protein activates in stress situations like DNA damage and oxidative stress.

    • The SH-SY5Y human neuroblastoma cell line, a relevant in vitro cell model for investigating neurotoxicology in human: Focus on organic pollutants

      2022, NeuroToxicology
      Citation Excerpt :

      Similarly to OCs and OPs, oxidative stress represents the main mechanism of cytotoxicity for a variety of pyrethroids (Ding et al., 2017; Martínez et al., 2019; Martínez et al., 2020a; Romero et al., 2012; Romero et al., 2015; Romero et al., 2017) (Supplementary Table 2). This oxidative stress leads to increased expression of apoptotic-related genes, oxidative stress response genes and neuroinflammation-related genes such as BAX, Bcl-2, Bcl-2 interacting protein 3 (BNIP3), AKT serine/threonine kinase 1 (AKT1), apoptotic peptidase activating factor 1 (APAF1), nuclear factor kappa B subunit 1 (NFKB1), TNF-α, Nrf2, caspase-3 (CASP3) and tumor protein 53 (P53) in cells treated with cyfluthrin and cypermethrin (Martínez et al., 2019). The two main metabolites of deltamethrin, 2′-OH-deltamethrin and 4′-OH-deltamethrin show higher cytotoxicity and stronger effect on lipid peroxidation and NO production on SH-SY5Y than deltamethrin (Romero et al., 2012).

    View all citing articles on Scopus
    View full text