Oxidative stress-mediated epidermal growth factor receptor activation regulates PM2.5-induced over-secretion of pro-inflammatory mediators from human bronchial epithelial cells

https://doi.org/10.1016/j.bbagen.2020.129672Get rights and content

Highlights

  • PM2.5 exposure increased pro-inflammatory mediator expression in BEAS-2B cells.

  • Exposure to PM2.5 elevates intracellular levels of reactive oxygen species.

  • Exposure to PM2.5 induces phosphorylation of the EGFR.

  • Oxidative stress is required for PM2.5-induced EGFR activation.

Abstract

Background

Exposure to PM2.5 has been associated with increased morbidity and mortality of lung diseases although the underlying mechanisms have not been fully uncovered. Airway inflammation is a critical event in the pathogenesis of lung diseases. This study aimed to examine the role of oxidative stress and epidermal growth factor receptor (EGFR) in PM2.5-induced pro-inflammatory response in a human bronchial epithelial cell line, BEAS-2B.

Methods

BEAS-2B cells were exposed to 0, 20, 50, 100 and 150 μg/ml of PM2.5. Secretion of pro-inflammatory mediators including interleukin-6 (IL-6), IL-8 and IL-1β was determined using enzyme linked immunosorbent assay. Levels of intracellular reactive oxygen species (ROS) were determined using flow cytometry. Phosphorylation of the EGFR was examined with immunoblotting.

Results

PM2.5 exposure increased the secretion of IL-6, IL-8, and IL-1β in a concentration-dependent fashion. Moreover, exposure to PM2.5 elevated intracellular levels of ROS, and phosphorylation of the EGFR (Y1068). Pretreatment of BEAS-2B cells with either an antioxidant or a specific EGFR inhibitor significantly reduced PM2.5-induced IL-6, IL-8 and IL-1β secretion, implying that both oxidative stress and EGFR activation were involved in PM2.5-induced pro-inflammatory response. Furthermore, pre-treatment of BEAS-2B cells with an antioxidant significantly blunted PM2.5-induced EGFR activation, suggesting that oxidative stress was required for PM2.5-induced EGFR activation.

Conclusion

PM2.5 exposure induces pro-inflammatory response in human bronchial epithelial cells through oxidative stress-mediated EGFR activation.

Introduction

Air pollution is a complex mixture of heterogeneous components from a wide variety of sources, mainly consisting of particulate matter (PM) and gaseous pollutants. All components of air pollution are harmful to human health, but the most severe effects have been attributed to PM [1]. Though the air quality has been improving in recent years, China is still experiencing severe and persistent air pollution because of rapid urbanization and climate change. Fine PM pollution in winter time is still severe, especially in Northern and Central China [2]. Inhaled PM can induce inflammatory alterations and severe health consequences including pulmonary and cardiovascular effects [1,[3], [4], [5], [6]]. Recent assessment of health outcomes of fine PM has suggested that the fine particles may be more closely associated with adverse respiratory health effects than larger particles [[7], [8], [9]]. Acute and long-term exposure to higher concentrations of ambient airborne fine particles increased morbidity and mortality of pulmonary and cardiovascular diseases [10].

PM can be classified into the following categories based on aerodynamic diameter: coarse PM (2.5–10 μm, PM2.510), fine PM (< 2.5 μm, PM2.5), and ultrafine PM (< 0.1 μm, PM0.1) [11]. The size of PM is directly linked to their potential for causing health problems. Small particles less than 10 μm in diameter pose the greatest problems, because they can get deep into lungs, and some may even get into bloodstream. PM2.5 is capable of reaching alveoli and deposits deeper in the lung while PM10 mostly stops at the upper bronchi [12]. Inhaled PM2.5 can penetrate pulmonary surfactant and attack the underling pulmonary epithelial cells [13]. Accumulated evidence indicates airway epithelium lining the respiratory airways not only acts as a frontline barrier against PM but also is actively involved in innate and acquired immune responses as well as airway inflammation, the latter has been proposed to play a critical role in the pathogenesis of diverse lung diseases [14]. In a recent review [6], we have presented the evidence showing that short- or long-term exposure to PM2.5 is associated with significant increases in hospitalizations, emergency department visits, or the incidence of inflammatory lung diseases including chronic obstructive pulmonary disease (COPD), asthma, and pneumonia, which is supported by the experimental results from toxicological studies with animal and cellular models [6].

Thus far, the exact mechanisms for PM2.5-induced airway inflammation have not been fully elaborated. Previous studies have demonstrated that oxidative stress and epidermal growth factor receptor (EGFR) are involved in airway inflammation induced by environmental insults [[15], [16], [17], [18], [19]]. Oxidative stress refers to the excessive production of ROS (reactive oxygen species) in the cells and tissues and antioxidant system cannot be able to neutralize them. The role of oxidative stress in air pollution-induced adverse health effects including lung inflammation has been acknowledged [20,21]. Evidence shows that inhaled ambient PM adversely affects the bronchial epithelium through various mechanisms including the imposition of oxidative stress, which drives the transcription of pro-inflammatory mediators relevant to asthma and other inflammatory lung diseases [22]. EGFR is a single transmembrane protein that possesses intrinsic tyrosine kinase activity. It can be directly activated or trans-activated in response to a variety of stimuli [23,24]. EGFR signaling plays an important role in the pathogenesis of inflammation through regulation of pro-inflammatory genes [25,26]. The activation of the EGFR signaling in PM- and PM component-treated human bronchial epithelial cells has been detected [27,28]. In this study, we used pro-inflammatory mediators as the biomarker of airway inflammation and determined the role of oxidative stress and EGFR in ambient PM2.5-induced pro-inflammatory response in human bronchial epithelial cells.

Section snippets

Reagents

NAC (N-acetyl-L-cysteine) was obtained from Sigma-Aldrich (St. Louis, USA). PD153035 was ordered from Apex Biotechnology Institute (Taiwan, China). Glass fiber filters were purchased from Pall Corporation (NY, USA). The rabbit antibodies against phospho-EGFR (Y1068) and pan EGFR antibody were obtained from Cell Signaling Technology (MA, USA). Horseradish peroxidase (HRP)-conjugated goat anti-rabbit antibody was obtained from Affinity Biosciences (OH, USA). MTT

Chemical composition of PM2.5

The PM2.5 was collected from Xinxiang, one of the most polluted cities in Central China in winter time. Metal and anion contents of PM2.5 are presented in Table 1. Noticeably, the contents of metals in PM2.5 vary greatly. Fe, Ba, Zn or Al were present at higher contents than other metals. In addition, NO3 and SO42− were detected as the main anions.

PM2.5 exposure increases the secretion of pro-inflammatory mediators

The cytotoxicity of PM2.5 was examined using MTT assay. After exposure of BEAS-2B cells to 20, 50, 100, 200, 300, 400 and 500 μg/ml PM2.5 for 6 h,

Discussion

A large number of studies have shown that PM2.5 air pollution poses adverse effects on human respiratory, cardiovascular and nervous systems [5,30,31]. In recent years, China has experienced serious, persistent and extensive air pollution. A national survey from China has shown that only a small portion of Chinese population live in areas that meet the annual standards of PM2.5 and PM10 from Chinese Ambient Air Quality Standards-Grade II [2]. PM2.5 has been a major air pollutant in urban areas

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

This study was supported by the National Natural Science Foundation of China (81573112; 81373030; 81703182) and Henan University Key Scientific Research Program (17A330004).

References (57)

  • L. Han et al.

    Impact of urbanization level on urban air quality: a case of fine particles (PM(2.5)) in Chinese cities

    Environ. Pollut.

    (2014)
  • M. Donadelli et al.

    Intracellular zinc increase inhibits p53−/− pancreatic adenocarcinoma cell growth by ROS/AIF-mediated apoptosis

    Biochim. Biophys. Acta, Mol. Cell Res.

    (2009)
  • W. Song et al.

    Role of the dissolved zinc ion and reactive oxygen species in cytotoxicity of ZnO nanoparticles

    Toxicol. Lett.

    (2010)
  • W. Wu et al.

    Zinc ions as effectors of environmental oxidative lung injury

    Free Radic. Biol. Med.

    (2013)
  • I.Y. Adamson et al.

    Zinc is the toxic factor in the lung response to an atmospheric particulate sample

    Toxicol. Appl. Pharmacol.

    (2000)
  • M. Sagai et al.

    Biological effects of diesel exhaust particles. I. in vitro production of superoxide and in vivo toxicity in mouse

    Free Radic. Biol. Med.

    (1993)
  • Y. Kumagai et al.

    Generation of reactive oxygen species during interaction of diesel exhaust particle components with NADPH-cytochrome P450 reductase and involvement of the bioactivation in the DNA damage

    Free Radic. Biol. Med.

    (1997)
  • W. Wu et al.

    Src-dependent phosphorylation of the epidermal growth factor receptor on tyrosine 845 is required for zinc-induced Ras activation

    J. Biol. Chem.

    (2002)
  • S.R. Lee et al.

    Reversible inactivation of protein-tyrosine phosphatase 1B in A431 cells stimulated with epidermal growth factor

    J. Biol. Chem.

    (1998)
  • N.K. Tonks

    Redox redux: revisiting PTPs and the control of cell signaling

    Cell.

    (2005)
  • S. Duan et al.

    Mechanism of PM2.5-induced human bronchial epithelial cell toxicity in Central China

    J. Hazard. Mater.

    (2020)
  • P.M. Mannucci et al.

    Effects on health of air pollution: a narrative review

    Intern. Emerg. Med.

    (2015)
  • D.W. Dockery et al.

    An association between air pollution and mortality in six U.S. cities

    N. Engl. J. Med.

    (1993)
  • J.H. Choi et al.

    Comparative study of PM2.5 - and PM10 - induced oxidative stress in rat lung epithelial cells

    J. Vet. Sci.

    (2004)
  • M. Franklin et al.

    Association between PM2.5 and all-cause and specific-cause mortality in 27 US communities

    J. Exp. Sci. Env. Epidemiol.

    (2007)
  • A. Valavanidis et al.

    Airborne particulate matter and human health: toxicological assessment and importance of size and composition of particles for oxidative damage and carcinogenic mechanisms

    J. Env. Sci. Heal. C Env. Carcinog. Ecotoxicol. Rev.

    (2008)
  • G. Hoek et al.

    Long-term air pollution exposure and cardio- respiratory mortality: a review

    Environ. Health

    (2013)
  • J. Øvrevik et al.

    Activation of Proinflammatory responses in cells of the airway mucosa by particulate matter: oxidant- and non-oxidant-mediated triggering mechanisms

    Biomolecules.

    (2015)
  • Cited by (11)

    • Mechanistic toxicity assessment of fine particulate matter emitted from fuel combustion via pathway-based approaches in human cells

      2022, Science of the Total Environment
      Citation Excerpt :

      Previous researches also indicated that PM2.5 exposure caused a decrease in the viability of A549 cells with a time- and concentration-dependent manner (Niu et al., 2017; Wu et al., 2017). Oxidative stress induced by ROS has been confirmed as one of the main toxic mechanisms resulting in the health effects of PM2.5 (Xiang et al., 2016; Wang et al., 2020). To determine the underlying mechanisms of toxicity induced by diesel and petrol FEPMs, intracellular ROS production in A549 cells and JEG-3 cells was measured.

    • The role of PKA/PP2B-mediated Drp1 phosphorylation and the subsequent EGFR inhibition in Cr(VI)-induced premature senescence

      2021, Ecotoxicology and Environmental Safety
      Citation Excerpt :

      We found that Cr(VI)-induced premature senescent hepatocytes showed decreased EGFR expression, and EGFR was activated in lung cancer tissues of non-smokers occupationally exposed to Cr(VI) for 19 years (Kim et al., 2014). The EGFR pathway is controlled by the level of ROS, which can induce EGFR activation (Wang et al., 2020). The activation of EGFR can inhibit premature senescence and promote malignant transformation of cells, and the inhibition of EGFR can promote senescence (Wang et al., 2014).

    View all citing articles on Scopus
    View full text