Melatonin inhibits Benzo(a)pyrene-Induced apoptosis through activation of the Mir-34a/Sirt1/autophagy pathway in mouse liver

https://doi.org/10.1016/j.ecoenv.2020.110556Get rights and content

Highlights

  • Benzo(a)pyrene can induce hepatotoxicity through increased oxidative stress and apoptosis markers.

  • Benzo(a)pyrene exposure increased hepatic miR-34a and decreased Sirt1 which reversed by melatonin.

  • Melatonin reduced the liver toxic effect of Benzo(a)pyrene by decreasing oxidative stress and apoptosis markers.

Abstract

Benzo(a)pyrene (BaP), an important environmental pollutant, is produced as the result of incomplete combustion of organic materials in many industries and food cooking process. It has been purposed that BaP induces hepatotoxicity through oxidative stress and apoptosis. Several studies have shown that melatonin can protect against chemical-induced apoptosis through autophagy pathway. In this study, we assessed the modulating effect of melatonin, a well-known antioxidant, on BaP-induced hepatotoxicity through induction of autophagy. Thirty male mice were treated daily for 28 consecutive days. BaP (75 mg/kg; oral gavage) and melatonin (10 and 20 mg/kg, i.p.) were administered to mice. The liver histopathology and the levels of apoptosis and autophagy proteins as well as the expression of miR-34a were determined. The BaP exposure induced severe liver histological injury and markedly enhanced AST, ALT and MDA level. Also, apoptosis proteins and hepatic miR-34a expression increased. However, the level of Sirt1 and autophagy markers such as LC3 II/I ratio and Beclin-1 reduced. The co-administration of melatonin reversed all changes caused by BaP. In summary, melatonin appears to be effective in BaP-induced hepatotoxicity maybe through the miR-34a/Sirt1/autophagy molecular pathway.

Introduction

Benzo(a)pyrene (BaP), a polycyclic aromatic hydrocarbon, is formed from the incomplete combustion of organic matter at temperatures between 300° and 600 °C. The ubiquitous compound is found in coal tar, tobacco smoke and many foods, especially grilled meats. The dietary intake of BaP per day by the general population is different in various communities. For example, the daily intake of BaP in the USA and Korea is estimated to be 2.2 μg/day and 124.55 ng/day, respectively, whereas an inhabitant of East Germany could approximately consume 1.0–3.3 μg/day of Bap (Fritz, 1972; Hattemer-Frey and Travis, 1991; Lee and Shim, 2007). The epoxide metabolites (BPDE) of BaP can bind to DNA, form DNA adduct and finally, caused to mutation (Shiizaki et al., 2013). For this reason, it is listed as a group 1 carcinogen by the International Agency for Research on Cancer (IARC) (Baird et al., 2005; Phillips and Venitt, 2012). BaP can be absorbed through oral, inhalation, and/or dermal exposure (Ramesh et al., 2001). After absorption, BaP is distributed primarily into the liver, kidneys, and the urinary bladder (Yamazaki et al., 1987). The primary site of BaP toxicity and metabolism is the liver which contains the majority of the enzymes required for bioactivation of BaP (Delgado-Roche et al., 2019).

As part of the BaP metabolism process, reactive oxygen species (ROS) are generated resulting in oxidative damage of intracellular macromolecules such as DNA and proteins, alterations in cellular structures and cell cycle progression (Guo et al., 2015). Oxidative stress and mitochondrial dysfunction are important mechanisms involved in BaP-induced hepatotoxicity (Ji et al., 2016), ultimately leading to cytotoxicity and ROS-mediated apoptotic cell death (Guo et al., 2015). BaP also is capable of inducing apoptosis via other mechanisms including DNA damage-related p53 pathway (Gao et al., 2011) and/or covalently metabolic intermediates-nuclear DNA binding (Stolpmann et al., 2012). Oral exposure to BaP has been reported to active a p53 DNA-damage response, which involves angiogenesis, induction of apoptosis, and growth signals (Labib et al., 2012).

Apoptosis or programmed cell death is a highly regulated process that results in cell death with energy consumption (Elmore, 2007). Caspases and the Bcl-2 family are important proteins, which are essential to activate apoptosis pathway (Cory and Adams, 2002; Pop and Salvesen, 2009). In addition, Bnip3 (BCL2/adenovirus E1B 19 kDa protein-interacting protein 3) is an apoptotic Bcl-2 protein (Zhang and Ney, 2009), which can modulate the permeability state of the outer mitochondrial membrane resulting in increased ROS production, mitochondrial depolarization and cytochrome c release. Bnip3 is a pro-apoptotic protein which can induce cell death and apoptosis (Burton and Gibson, 2009). Previous studies have shown BaP-induced apoptosis in different organs such as liver, lung and brain (He et al., 2016; Qin et al., 2015; Sakthivel et al., 2019). It has been demonstrated that BaP-induced apoptosis could regulate by several natural compounds including catechin and resveratrol (Banerjee et al., 2016; Shahid et al., 2016).

Melatonin (5 methoxy-N-acetyltryptamine) is an endogenous hormone synthesized from the amino acid tryptophan and circulating melatonin is produced primarily by the pineal gland at night (Tan et al., 2015). The melatonin synthesis and secretion is enhanced by darkness while inhibited by light. Melatonin is involved in the regulation of circadian rhythms such as the neuroendocrine rhythm, the sleep-wake rhythm, and the body temperature cycle (Karasek and Winczyk, 2006; Tordjman et al., 2017). The effects of melatonin on blood pressure (Scheer et al., 2004), autonomic cardiovascular regulation (Nishiyama et al., 2001), retinal function (Iuvone et al., 2005), and regulation of the immune system (Carrillo-Vico et al., 2005) have been reported. In addition, melatonin has several pharmacological activities such as hepatoprotective (Oleshchuk et al., 2019; Sheen et al., 2016; Tunon et al., 2011), neuroprotective (Alghamdi, 2018), gastroprotective (Brzozowski et al., 2005), and anti-cancer effects (Asghari et al., 2017). Furthermore, melatonin has been shown to exhibit a potent antioxidant activity through its ability to scavenge free radical oxygen species, an indirect effect that results in reduction of nitric oxide formation or induction of antioxidant enzymes, thereby protecting cells and tissues against oxidative stress damage (Galano et al., 2013; Reiter et al., 2016). Other studies have proposed that melatonin has a protective role through modulation of inflammation (Mauriz et al., 2013), apoptosis (Bizzarri et al., 2013; Tunon et al., 2011), and autophagy (Roohbakhsh et al., 2018; San-Miguel et al., 2015; Stacchiotti et al., 2019).

Autophagy, a highly conserved catabolic process, allows the orderly degradation and recycling of cellular components. As part of the autophagy process of supporting cellular homeostasis, the process responds to different conditions including starvation, hypoxia, and oxidative stress (Hashemzaei et al., 2017). Autophagy contributes to degradation of cellular content by the lysosomes which involves removal of cytosolic components, misfolded protein aggregates and damaged or ectopic organelles (e.g., mitochondria and endoplasmic reticulum) (Barangi et al., 2019). Generally, autophagy is considered a protector and survival response to starvation or a toxic environment insult and is critical to normal cell physiology. However, sometimes, dysregulated autophagy switches to autophagic (non-apoptotic) programmed cell death (Kroemer and Levine, 2008). Melatonin has been reported to attenuate neuronal apoptosis markers or to increase basal autophagy proteins as a protective pathway in peripheral sciatic nerves and dorsal root ganglion in oxaliplatin-administered rats (Areti et al., 2017). In contrast, pre-ischemia melatonin treatment has showed anti-fibrogenic effects against CCl4-induced fibrosis through inhibition of autophagy (San-Miguel et al., 2015).

The autophagy pathway is mediated by the autophagy-related (Atg) genes. Microtubule associated protein 1 light chain 3 (LC3; mammalian homologues of yeast Atg8) and Beclin-1 (mammalian homologues of yeast Atg6) are two such autophagic proteins that have been reported to contribute to the induction and regulation of autophagy and autophagosome formation (Petibone et al., 2017). Moreover, Beclin-1 interacts with the anti-apoptotic Bcl-2 family and thus regulates autophagy. It has been demonstrated that the Bcl-2/Beclin-1 complex plays a key role in the crosstalk between autophagy and apoptosis (Wirawan et al., 2010). On the other hand, autophagy can be regulated by other factors such as transcription factor activity like NF-κB and Sirtuin activity (Wang et al., 2019).

Sirtuins, a nicotinamide adenine dinucleotide (NAD+)-dependent protein, contains either mono-ADP-ribosyltransferase activity or deacylase activity, including deacetylase, desuccinylase, demalonylase, demyristoylase, and/or depalmitoylase activity (Mayo et al., 2017). Sirtuin 1 (silent information regulator 1; Sirt1), one of seven isoforms, occurs both in the cytosol and nucleus of mammals (Tanno et al., 2007). Sirt1 plays a pivotal role in antioxidant defense (Hariharan et al., 2010), inflammation (Carloni et al., 2016), autophagy (Jang et al., 2012), and apoptosis (Zhang et al., 2016). It was reported that melatonin enhanced Sirt1 and autophagy proteins, but reduced inflammation and apoptosis markers in brain injury (Carloni et al., 2016). Recently, Ren et al. illustrated that melatonin could ameliorate hepatocellular damage induced by chronic intermittent hypoxia via activating Sirt1-mediated autophagy pathway (Ren et al., 2019).

Sirt1 expression in various cellular processes is modulated by microRNAs (miRNAs), small endogenous and highly conserved noncoding RNAs (~22 nucleotide in length) (Karbasforooshan et al., 2018). MicroRNAs modulate post-transcriptional gene expression by targeting mRNA and suppressing protein expression (Razavi-Azarkhiavi et al., 2017). Among the various miRNAs, miR-34a is known to inhibit the expression of Sirt1 protein in different tissues under various pathophysiological processes (Carloni et al., 2016; Karbasforooshan and Karimi, 2018). MiR-34a inhibits Sirt1 expression and increases the acetylation of p53; ultimately leading to apoptosis in human colon cancer cells (Yamakuchi et al., 2008). Melatonin has been reported to trigger autophagy through upregulating Sirt1 and downregulating miR-34a in non-alcoholic fatty liver disease (Stacchiotti et al., 2019) and in a neonatal brain inflammation model (Carloni et al., 2016).

Induction of oxidative stress, followed by apoptosis, is an important mechanism in BaP-induced hepatotoxicity. Since no information was found on the protective effect of melatonin as well as autophagy pathway on BaP-induced hepatotoxicity, we undertook a study to investigate the interaction between melatonin and the miR-34a/Sirt1/autophagy pathway as related to BaP-induced hepatotoxicity. The study investigated whether melatonin had a protective effect against liver injury in BaP treated mice and whether the attenuate BaP-induced oxidative stress and apoptosis were mediated through the miR-34a/Sirt1/autophagy pathway.

Section snippets

Chemicals and reagents

Benzo(a)pyrene (C20H12, >96% purity; sc-257,130) and melatonin (C13H16N2O2, >99% purity; sc-207848) were purchased from the Santa Cruze Biotechnology Company, USA; potassium chloride (KCl), phosphoric acid, and thiobarbituric acid (TBA) were purchased from the Merck Company, Germany; 5, 5′-Dithiobis 2-nitrobenzoic acid (DTNB) and trichloroacetic acid (TCA) were purchased from the Sigma Company, Germany. All of used antibodies were purchased from Cell Signaling Company, USA.

Animals

Thirty male Razi mice

BaP and melatonin effect on the liver enzymes activity

The results in Fig. 1 show that after BaP treatment, the serum AST and ALT activity (IU/L) significantly increased compared to controls (P < 0.0001). Melatonin at a dose of 10 mg/kg did not decrease these elevated levels; however, 20 mg/kg of melatonin statistically attenuated the AST and ALT activity comparing to the BaP treated group (P < 0.001). There were no significant changes in the activity of AST and ALT in mice administered only melatonin compared to control group.

BaP and melatonin effect on MDA and GSH content in mouse liver

MDA is a marker of

Discussion

The current study investigated whether melatonin had the potential to reduce hepatotoxicity arising from oxidative injury and apoptosis induced in mice by BaP through the miR34a/Sirt1/autophagy signaling pathway. BaP (75 mg/kg, orally, 28 days) increased apoptosis following enhancement of ROS production and thus, induced hepatotoxicity in mice, which was confirmed by assessment of oxidative stress markers and histopathology. Moreover, BaP could reduce autophagy markers and miR-34a expression

Conclusion

In conclusion, the enhancement of oxidative stress and apoptosis as well as the reduction of autophagy were shown to be involved in the BaP-induced hepatotoxicity in mice. BaP increased the MDA level and induced apoptosis proteins (Bax/Bcl-2 ratio, Bnip3, and caspase-3) and the miR-34a expression, while decreasing the level of autophagy markers (LC3 II/I ratio, Beclin-1, and Sirt1). Most importantly, this study provides evidence for the hepatoprotective effects of melatonin in BaP-related liver

CRediT authorship contribution statement

Samira Barangi: Data curation, Writing - original draft. Soghra Mehri: Conceptualization, Methodology. Zahra Moosavi: Investigation. A. Wallace Hayesd: Writing - review & editing, Validation. Russel J. Reiter: Writing - review & editing, Validation. Daniel P. Cardinali: Writing - review & editing, Validation. Gholamreza Karimi: Conceptualization, Methodology.

Declaration of competing interest

The Authors declare that they have no conflicts of interest to disclose.

Acknowledgements

Authors are grateful to the Vice Chancellor of Research, Mashhad University of Medical Sciences, Mashhad, Iran for financial support.

References (95)

  • P.M. Iuvone

    Circadian clocks, clock networks, arylalkylamine N-acetyltransferase, and melatonin in the retina

    Prog. Retin. Eye Res.

    (2005)
  • S.Y. Jang

    Nicotinamide-induced mitophagy: event mediated by high NAD+/NADH ratio and SIRT1 protein activation

    J. Biol. Chem.

    (2012)
  • X. Ji

    Depletion of mitochondrial enzyme system in liver, lung, brain, stomach and kidney induced by benzo(a)pyrene

    Environ. Toxicol. Pharmacol.

    (2016)
  • H. Jin

    Melatonin protects endothelial progenitor cells against AGE-induced apoptosis via autophagy flux stimulation and promotes wound healing in diabetic mice

    Exp. Mol. Med.

    (2018)
  • H. Karbasforooshan et al.

    The role of SIRT1 in diabetic retinopathy

    Biomed. Pharmacother.

    (2018)
  • H. Karbasforooshan

    SIRT1 and microRNAs: the role in breast, lung and prostate cancers

    Exp. Cell Res.

    (2018)
  • H.J. Kim

    Carbon monoxide protects against hepatic ischemia/reperfusion injury by modulating the miR-34a/SIRT1 pathway

    Biochim. Biophys. Acta

    (2015)
  • M. Kumar

    Understanding the role of 3-O-Acetyl-11-keto-beta-boswellic acid in conditions of oxidative-stress mediated hepatic dysfunction during benzo(a)pyrene induced toxicity

    Food Chem. Toxicol.

    (2017)
  • Y. Lin

    Berberine protects against ischemia/reperfusion injury after orthotopic liver transplantation via activating Sirt1/FoxO3alpha induced autophagy

    Biochem. Biophys. Res. Commun.

    (2017)
  • J.Q. Ma

    Ganoderma applanatum terpenes protect mouse liver against benzo(alpha)pyren-induced oxidative stress and inflammation

    Environ. Toxicol. Pharmacol.

    (2011)
  • M.S. Moron

    Levels of glutathione, glutathione reductase and glutathione S-transferase activities in rat lung and liver

    Biochim. Biophys. Acta

    (1979)
  • E. Murawska-Cialowicz

    Effects of melatonin on lipid peroxidation and antioxidative enzyme activities in the liver, kidneys and brain of rats administered with benzo(a)pyrene

    Exp. Toxicol. Pathol.

    (2011)
  • H. Ohkawa

    Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction

    Anal. Biochem.

    (1979)
  • C. Pop et al.

    Human caspases: activation, specificity, and regulation

    J. Biol. Chem.

    (2009)
  • G. Qin

    Sulfur dioxide and benzo(a)pyrene trigger apoptotic and anti-apoptotic signals at different post-exposure times in mouse liver

    Chemosphere

    (2015)
  • A. Ramesh

    Metabolism, bioavailability, and toxicokinetics of benzo(alpha)pyrene in F-344 rats following oral administration

    Exp. Toxicol. Pathol.

    (2001)
  • A. Roohbakhsh

    Melatonin as an endogenous regulator of diseases: the role of autophagy

    Pharmacol. Res.

    (2018)
  • A. Shahid

    Modulatory effects of catechin hydrate against genotoxicity, oxidative stress, inflammation and apoptosis induced by benzo(a)pyrene in mice

    Food Chem. Toxicol.

    (2016)
  • K. Shiizaki

    Dioxin suppresses benzo[a]pyrene-induced mutations and DNA adduct formation through cytochrome P450 1A1 induction and (+/-)-anti-benzo[a]pyrene-7,8-diol-9,10-epoxide inactivation in human hepatoma cells

    Mutat. Res.

    (2013)
  • M. Tanno

    Nucleocytoplasmic shuttling of the NAD+-dependent histone deacetylase SIRT1

    J. Biol. Chem.

    (2007)
  • X. Wang

    Astragaloside IV inhibits glucose-induced epithelial-mesenchymal transition of podocytes through autophagy enhancement via the SIRT-NF-kappaB p65 axis

    Sci. Rep.

    (2019)
  • W. Zhang

    Dioscin alleviates dimethylnitrosamine-induced acute liver injury through regulating apoptosis, oxidative stress and inflammation

    Environ. Toxicol. Pharmacol.

    (2016)
  • H. Zou

    Autophagy and gap junctional intercellular communication inhibition are involved in cadmium-induced apoptosis in rat liver cells

    Biochem. Biophys. Res. Commun.

    (2015)
  • B.S. Alghamdi

    The neuroprotective role of melatonin in neurological disorders

    J. Neurosci. Res.

    (2018)
  • A. Areti

    Melatonin prevents mitochondrial dysfunction and promotes neuroprotection by inducing autophagy during oxaliplatin-evoked peripheral neuropathy

    J. Pineal Res.

    (2017)
  • W.M. Baird

    Carcinogenic polycyclic aromatic hydrocarbon-DNA adducts and mechanism of action

    Environ. Mol. Mutagen.

    (2005)
  • M. Bizzarri

    Molecular mechanisms of the pro-apoptotic actions of melatonin in cancer: a review

    Expert Opin. Ther. Targets

    (2013)
  • T. Brzozowski

    Importance of the pineal gland, endogenous prostaglandins and sensory nerves in the gastroprotective actions of central and peripheral melatonin against stress-induced damage

    J. Pineal Res.

    (2005)
  • T.R. Burton et al.

    The role of Bcl-2 family member BNIP3 in cell death and disease: NIPping at the heels of cell death

    Cell Death Differ.

    (2009)
  • Z. Cao

    Melatonin alleviates cadmium-induced liver injury by inhibiting the TXNIP-NLRP3 inflammasome

    J. Pineal Res.

    (2017)
  • S. Carloni

    Melatonin modulates neonatal brain inflammation through endoplasmic reticulum stress, autophagy, and miR-34a/silent information regulator 1 pathway

    J. Pineal Res.

    (2016)
  • A. Carrillo-Vico

    A review of the multiple actions of melatonin on the immune system

    Endocrine

    (2005)
  • S. Cory et al.

    The Bcl2 family: regulators of the cellular life-or-death switch

    Nat. Rev. Canc.

    (2002)
  • D.N. Das

    Bacopa monnieri-induced protective autophagy inhibits benzo[a]pyrene-mediated apoptosis

    Phytother Res.

    (2016)
  • L. Delgado-Roche

    Chemoprotective effects of Ulva lactuca (green seaweed) aqueous-ethanolic extract against subchronic exposure to benzo(a)pyrene by CYP1A1 inhibition in mice

    Phytother Res.

    (2019)
  • S. Elmore

    Apoptosis: a review of programmed cell death

    Toxicol. Pathol.

    (2007)
  • W. Fritz

    [Extent and source of contamination of food with carcinogenic hydrocarbons]

    Nahrung

    (1972)
  • Cited by (0)

    View full text