ArticlePDF Available

Metallothionein ameliorates airway epithelial apoptosis upon particulate matter exposure: role of oxidative stress and ion homeostasis

Authors:
  • Renmin hospital of wuhan univeisity, China, Wuhan

Abstract

To investigate the mechanism underlying particulate matter (PM) exposure-induced oxidative stress and potential rescue strategies against pulmonary damage in this context. A combination of omics technology and bioinformatic analysis were used to uncover mechanisms underlying cellular responses to PM exposure in human bronchial epithelia (HBE) cells and imply the potential rescue. Our results implicated that oxidative stress, metal ion homeostasis, and apoptosis were the major cellular responses to PM exposure in HBE cells. PM exposure disrupted oxidative phosphorylation (OXPHOS)-related gene expressions in HBE cells. Rescuing the expression of these genes with supplemental coenzyme Q10 (Co Q10) inhibited reactive oxygen species (ROS) generation; however, it only partially protected HBEs against PM exposure-induced apoptosis. Further, metallothionein (MT)-encoding genes associated with metal ion homeostasis were significantly induced in HBE cells, which was transcriptionally regulated by specificity protein 1 (SP1). SP1 knock-down (KD) aggravated PM-induced apoptosis in HBE cells, suggesting it plays a role in MT induction. Subsequent studies corroborated the protective role of MT by showing that exogenous MT supplement demonstrated effective protection against PM-induced oxidative stress and apoptosis in HBE cells. Importantly, exogenous MT supplement was shown to reduce ROS generation and apoptosis in airway epithelia in both HBE cells and a PM-inhaled murine model. This study demonstrates that the impact of MT on airway epithelia by suppressing oxidative stress and maintaining metal ion homeostasis is beneficial in attenuating damage to pulmonary cells undergoing PM exposure.
Lietal. Current Medicine (2024) 3:9
https://doi.org/10.1007/s44194-024-00036-7
ORIGINAL RESEARCH Open Access
© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which
permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the
original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or
other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line
to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this
licence, visit http://creativecommons.org/licenses/by/4.0/.
Current Medicine
Metallothionein ameliorates airway
epithelial apoptosis uponparticulate matter
exposure: role ofoxidative stress andion
homeostasis
Bin Li1†, Nannan Huang1,2†, Shengnan Wei2, Qingtao Meng1, Shenshen Wu1, Michael Aschner3*,
Xiaobo Li1,2,6* and Rui Chen1,4,5*
Abstract
Purpose To investigate the mechanism underlying particulate matter (PM) exposure-induced oxidative stress
and potential rescue strategies against pulmonary damage in this context.
Methods A combination of omics technology and bioinformatic analysis were used to uncover mechanisms under-
lying cellular responses to PM exposure in human bronchial epithelia (HBE) cells and imply the potential rescue.
Results Our results implicated that oxidative stress, metal ion homeostasis, and apoptosis were the major cellular
responses to PM exposure in HBE cells. PM exposure disrupted oxidative phosphorylation (OXPHOS)-related gene
expressions in HBE cells. Rescuing the expression of these genes with supplemental coenzyme Q10 (Co Q10) inhib-
ited reactive oxygen species (ROS) generation; however, it only partially protected HBEs against PM exposure-induced
apoptosis. Further, metallothionein (MT)-encoding genes associated with metal ion homeostasis were significantly
induced in HBE cells, which was transcriptionally regulated by specificity protein 1 (SP1). SP1 knock-down (KD) aggra-
vated PM-induced apoptosis in HBE cells, suggesting it plays a role in MT induction. Subsequent studies corroborated
the protective role of MT by showing that exogenous MT supplement demonstrated effective protection against PM-
induced oxidative stress and apoptosis in HBE cells. Importantly, exogenous MT supplement was shown to reduce
ROS generation and apoptosis in airway epithelia in both HBE cells and a PM-inhaled murine model.
Conclusion This study demonstrates that the impact of MT on airway epithelia by suppressing oxidative stress
and maintaining metal ion homeostasis is beneficial in attenuating damage to pulmonary cells undergoing PM
exposure.
Keywords Particulate matter, Oxidative stress, Ion homeostasis, Metallothionein
Bin Li and Nannan Huang contributed equally to this work.
*Correspondence:
Michael Aschner
michael.aschner@einsteinmed.edu
Xiaobo Li
101011116@seu.edu.cn
Rui Chen
ruichen@ccmu.edu.cn
Full list of author information is available at the end of the article
Page 2 of 16
Lietal. Current Medicine (2024) 3:9
1 Introduction
Urban particulate matter (PM) in ambient air is a het-
erogeneous mixture of particle sizes and chemicals. PM
typically originates from mobile and industrial fossil fuel
combustion, and its specific constituents and sources
have not been fully elucidated. e adverse health effects
of PM exposure have been associated with respiratory-
related morbidity and mortality (Li et al. 2018; Zhang
2021; Bowe etal. 2019). Airway inflammation, systemic
inflammation, and oxidative stress have been implicated
in experimental animals or longitudinal panel studies
(Feng etal. 2023; Xu etal. 2022).
e respiratory track epithelial cells are at the interface
between the airspace and the internal milieu, acting as
a physical and biochemical barrier to ensure transition
between these distinct compartments. Airway epithe-
lia cells can be activated by a host of stimuli, including
air pollutants (Fu etal. 2019; Barbier etal. 2023). In the
context of activation, epithelial cells provide an adap-
tive response to these stimuli, which is important for the
elimination and wound healing, and the generation of a
systemic response (Crotty Alexander etal. 2018; Gang-
war etal. 2020). erefore, airway epithelia are the first
line of defense against air pollutants, as well as the pri-
mary target of inhaled PM in organisms.
Numerous studies have focused on the mechanisms
underlying PM exposure-induced adverse health effects.
Overall, these studies corroborate that oxidative stress
mediates, at least in part, PM-induced pulmonary toxic-
ity (Gangwar et al. 2020; Lan etal. 2021). Nonetheless,
gaps in knowledge delineating the precise role of ROS in
air pollution-mediated pathologies have yet to be fully
characterized (Gangwar et al. 2020). Furthermore, with
consistent and inevitable air pollution exposure, addi-
tional studies investigating novel efficient therapeutic
interventions are urgently required (Whyand etal. 2018).
In the present study, human bronchial epithelial cells
(HBE) and mice were exposed to various concentra-
tions and doses, respectively, of PM. Omics technology
and biological assays were used to explore the molecular
pathways underlying pulmonary toxicity of PM. e cel-
lular source of ROS generation and underlying mecha-
nisms upon PM exposure was delineated. Importantly,
our findings identified a putative rescue strategy against
PM-induced toxicity, shedding novel insight on interven-
tion strategies and the putative role of MT in rescue from
PM-induced pulmonary damage.
2 Material andmethods
2.1 Particulate matter (PM)
Urban Particulate Matter (Standard Reference Material
1648a) were purchased from National Institute of Stand-
ards and Technology, USA. e components of SRM
1648a were introduced by a research group from NIST
(Schantz etal. 2012).
2.2 Cell culture
e human bronchial epithelial cells (HBE, American
Type Culture Collection) were maintained in Dulbecco’s
modified Eagle’s medium (DMED) at 37°C in 5% CO2.
e culture medium was supplemented with 10% (v/v)
fetal bovine serum (FBS), penicillin (100 U/mL), strepto-
mycin (100μg/mL).
2.3 Cellular viability
Cellular viability was evaluated with a Cell Counting
Kit-8 (CCK-8, Nanjing Jiancheng Bioengineering Insti-
tute, China). According to the reconciliation between
invitro and invivo doses of PM (Li etal. 2003), PM-
induced biological effects at the dose range of 0.2–20 μg/
cm2 invitro were equal to that at the dose of 75 μg/m3
over a 24-h period which is PM2.5 limit recommended in
China. HBE cells were plated at a density of 1 × 104 per
well in a 96-well plate and then exposed to 100 μL PM
mixture (0, 12.5, 25, 50, 100, 250, 500, and 1, 000 μg/
mL) with three biological replicates for each concentra-
tion, equal to PM (0, 0.16, 0.33, 0.65, 1.3, 3.25, 6.5, 13 μg/
cm2) for 24 h. Cell viability affected by PM were moni-
tored every 24 h up to 3 days. en 10 μL CCK-8 solution
was added to each well, the cells were incubated for 4 h
at 37°C, and the absorbance was determined at 450 nm.
2.4 RNA microarray andgene expression analysis
HBE cells were seeded in 10 cm culture dishes and
exposed to 0, 100, and 500 μg/mL PM, respectively,
with three biological replicates. Total cellular RNA was
extracted after 24 h treatment with the TRIZOL rea-
gent (Invitrogen, US), according to the manufacturer’s
instructions (Agilent Technologies, Santa Clara, CA, US),
and subjected to microarray assay. e labeled cRNAs
were hybridized onto a Human LncRNA Array v3.0
(8 × 60K; Arraystar) chip, which is designed for 26, 109
coding genes. e arrays were scanned with an Agilent
G2505C scanner and the density of fluorescence was ana-
lyzed with Agilent Feature Extraction software (version
11.0.1.1). Quantile normalization and subsequent data
processing were performed with GeneSpringGx v12.0
software package (Agilent Technologies). An absolute
fold change (FC) of 1.5 or more and p = 0.05 were set as
threshold to evaluate the significance of gene expression
differences of raw data.
2.5 Functional group analysis
e DAVID 6.7 (Database for Annotation, Visualiza-
tion and Integrated Discovery, https:// david. ncifc rf.
gov/) functional annotation tool was used to denote the
Page 3 of 16
Lietal. Current Medicine (2024) 3:9
significance of gene ontology (GO) term enrichment
in the differentially expressed mRNAs (p-value was set
less than 0.1) based on a background of homo sapiens
genome.
2.6 Animal treatments
Male C57BL/6 mice (20–22 g), were purchased from
GemPharmatech Co. Ltd. (China) and housed in a spe-
cific pathogen-free (SPF) animal facility, in which the
room temperature was maintained at 22.5 C with 12h
light/dark cycle. Mice were housed six per polycarbonate
cage on corncob bedding with adlibitum access to food
and water. Mice received standard rodent chow (Jiangsu
Xietong Pharmaceutical Bio-engineering Co., Ltd.,
China) sterilized by cobalt (Co) 60 radiation and auto-
claved water adlibitum. Animals were treated humanely,
maintained, and used in accordance with Guidelines of
Committee on Animal Use and Care of Southeast Uni-
versity (20190226014).
Mice were divided into six groups (n = 6/group),
namely filtered air (FA)/Vehicle, PM (300μg/m3)/Vehicle,
PM (600μg/m3)/Vehicle, FA/metallothionein (MT), PM
(300μg/m3)/MT, and PM (600μg/m3)/MT. Exposure was
carried out as previously described (Li etal. 2021; Meng
etal. 2022). In brief, mice were placed in three stainless-
steel Hinners-type whole-body inhalation chambers; two
chambers were ventilated with PM, and the remaining
one with HEPA-filtered clean air at the same flow rate as
the PM exposure chamber. Mice were exposed for 2h per
day for 30 consecutive days. MT were administrated at
a dose of 2.5mg/kg, i.p., 1h before PM exposure every
3days. Control mice received equal volume of 0.9% saline
injection.
Mice were deeply euthanized under isoflurane anesthe-
sia 1h after the end of PM exposure on the 30th day. A
portion of the left lung tissue from each mouse was fixed
in 4% paraformaldehyde (PFA), and the remainder of the
lung tissues were stored in liquid nitrogen.
2.7 RNA isolation andquantitative real‑time PCR assay
HBE cells were exposed to 0, 50, 100, 250, or 500μg/mL
PM for 24h. Next, cells were trypsinized and collected.
Total RNA was extracted with the GenEluteTM Mam-
malian Total RNA Miniprep Kit (Sigma, US). cDNA
synthesis for coding genes was performed with 1μg of
total RNA according to the manufacturer’s instruction
(Toyobo, Tokyo, Japan).
e mRNA levels for target genes were determined by
reverse transcription of total RNA followed by quantita-
tive real-time PCR analysis (qRT-PCR) on a Quant Studio
6 Flex system (Applied Biosystems, Life Technologies,
US) with SYBR PCR Master Mix reagent kits (Toyobo,
Japan). Primer sequences are shown in Table S1. All
experiments were independently performed in triplicate.
e mRNA levels for the indicated gene are relative to
cyclophilin A (CycA).
2.8 JC‑1 staining
Changes in mitochondrial membrane potential (Δψm)
in pulmonary cells were determined with a commercial
5,50,6,6-tetrachloro-1,1,3,3-tetraethyl-benzimidazolyl-
carbocyanine iodide (JC-1) kit (C2003S, Beyotime, China).
HBE cells were exposed to PM for 24 h, added with JC-1
staining working solution, and incubated at 37°C for 20
min. After the incubation, HBE cells were washed twice
with JC-1 staining buffer and evaluated under a fluores-
cence microscope (Olympus, Japan) to examine green and
red fluorescence, the relative fluorescent intensities were
determined by the ratio of green/red fluorescent intensity
with Photoshop software.
2.9 ROS generation
ROS levels in HBE cells and murine lung tissues were
determined with a commercial ROS assay kit (S0033S,
Beyotime Biotechnology, China). After exposure to 0,
50, 100, 250, or 500μg/mL PM for 24h, HBE cells were
washed with PBS. en 2’, 7’-dichlorofluorescein-diace-
tate (DCFH-DA) probe was added at a final concentra-
tion of 10μmol/L and incubated at 37°C for 30min to
determine the content of intracellular ROS.
Paraffin-fixed lung tissue sections were incubated with
DCFH-DA at 37°C for 20 min. e sections were covered
with a mounting solution containing DAPI (DAPI Fluo-
romount-G, SouthernBiotech, USA), and then observed
under a fluorescence microscope (Axio Imager M2, Zeiss,
Germany). ree non-overlapping fields containing at
least one small airway at low-power field (100 ×) of each
section were randomly selected, the relative fluorescent
(See figure on next page.)
Fig. 1 PM exposure alters mRNA expression profiles in HBE cells. a HBE cellular viability was inhibited in a concentration-depended manner
following PM exposure. (n = 3/group, one-way ANOVA for each time point). b Volcano plots showed the differentially expressed genes
following 100 or 500 μg/mL PM exposure for 24 h with a cut-off as fold change (FC) < 1.5 and p-value > 0.05 (n = 3/group). c Venn diagram showed
the differentially expressed genes in 100 or 500 μg/mL PM-exposed HBE cells. d and e GO terms (biological function, BP) of differential expressed
genes of 100 or 500 μg/mL PM-exposed HBE cells. f GO terms (BP) of differential expressed genes overlapped in 100 and 500 μg/mL PM-exposed
HBE cells. g Schematic of PM exposure resulted cellular damages in HBE cells
Page 4 of 16
Lietal. Current Medicine (2024) 3:9
Fig. 1 (See legend on previous page.)
Page 5 of 16
Lietal. Current Medicine (2024) 3:9
intensities were determined by the ratio of green/DAPI
fluorescent intensity with Photoshop software.
2.10 ATP levels
ATP levels were measured with a luciferase ATP assay
kit (Beyotime, China). HBE cells were exposed to 0, 50,
100, 250, or 500μg/mL PM for 24h and added with 200
μL of lysis buffer. Cells were collected and centrifuged at
12,000rpm for 5 min at 4°C. e luminescence of the
supernatant was assayed with a luminometer (Berthold
Detection System, Pforzheim, Germany).
2.11 Cytochrome c contents
After exposure to 0, 50, 100, 250, or 500μg/mL PM for
24h, HBE cells were washed with PBS. Mitochondrial
and cytosolic proteins of HBE cells were isolated by a
Mitochondria/Cytosol Fractionation Kit (Beyotime,
China) and subsequently quantitated using the Brad-
ford method. e levels of cytochrome c were estimated
according to the ELISA kit procedures (R&D Systems,
U.S.).
2.12 Cell apoptosis analysis
PM exposure-induced cellular apoptosis was analyzed
by flow cytometry with an Annexin V-FITC Apoptosis
Detection Kit (KeyGEN BioTECH, China). Briefly, after
exposure to 0, 50, 100, 250, or 500μg/mL PM for 24h,
HBE cells were harvested and washed twice with PBS,
followed by centrifugation at 1, 000rpm for 5min. en
the pellets were resuspended in 500 μL binding buffer
and incubated with 5 μL FITC-conjugated annexin V and
5 μL PI for 15min at room temperature in the dark. e
samples were analyzed by FACS Calibur Flow Cytometry
(BD Biosciences, US).
2.13 ELISA assays
HBE cells were exposed to 0, 50, 100, 250, or 500 μg/
mL PM for 24h. Protein content was measured by the
Bradford method. HBE cells were incubated with Ac-
DEVD-pNA, Ac-IETD-pNA, Ac-LEHD-pNA, which are
substrate peptides of caspase-3, 8, and 9, respectively
(Beyotime Institute of Bio-technology, China) for 2h at
37 °C. Release of p-nitroanilide (pNA) from substrates
was measured at 405nm by a microplate reader (Molecu-
lar Devices, U.S.)
2.14 ChIP assay
ChIP was performed using the ChIP-IT Express Mag-
netic assay kit (Active Motif, USA). HBE cells were
exposed to 0, 100, or 500μg/mL PM for 24h, and fixed
with 4% formaldehyde for 10min. e samples were then
centrifuged at 2, 000g for 2min and washed once with
cold PBS. e ChIP reaction antibody was a normal rab-
bit IgG (NI01, EMD Chemicals, Inc., Gibbstown, NJ) and
an anti-SP1 (1:500 dilution; ab231778, Abcam, USA).
Precipitated genomic DNA was analyzed by quantitative
PCR in triplicate measurements for each sample using
appropriate primers (TableS2).
2.15 Western blot
HBE cells were exposed to 0, 100, or 500μg/mL PM for
24h, and total proteins were extracted by a total pro-
tein extraction kit (Life Technologies, USA) from cells.
A total of 20µg of protein was subjected to electropho-
resis. Blots were then incubated with primary antibodies
against SP1 (1: 1000 dilution; ab231778, Abcam, USA) for
48h, follow by incubation with HRP-conjugated second-
ary antibodies for 1h. α-tubulin (1:5000 dilution; ab7291,
Abcam, USA) was used as a loading control. e immu-
noreactive signals were visualized with a Super ECL Plus
Kit (Yeasen, China).
2.16 TUNEL staining
Apoptotic cells in lung tissues were evaluated with ter-
minal-deoxynucleoitidyl transferase mediated nick end
labeling (TUNEL) staining by a Roche InSitu Cell Death
Detection Kit (Roche, U.S.). e nuclear stained areas
(depicted in dark brown) were identified as TUNEL-pos-
itive cell. e proportion of TUNEL-positive cells of an
airway were estimated by two experienced histologists
who were blinded to experimental design. ree to five
non-overlapping airways in each section were counted in
high-power fields (HPFS, × 400 magnification) and ana-
lyzed. e airways with the maximum number of positive
cells were selected for analysis (Li etal. 2017).
Fig. 2 Low-concentration of PM exposure induces oxidative stress in HBE cells. a Venn diagram showed the differentially expressed genes
and potential cellular responses to PM exposure in HBE cells. b Chord diagram of mitochondria-related GO terms and involved genes. c Heatmap
of mitochondria-related genes in HBE cells following PM exposure for 24 h by microarray. d qRT-PCR assays of mitochondria-related genes
in HBE cells following PM exposure for 24 h (n = 9/group, one-way ANO VA). e JC-1 staining of HBE cells following PM exposure for 24 h (n = 3/
group, one-way ANOVA). f ATP production in HBE cells following PM exposure for 24 h (n = 3/group, one -way ANOVA). g Contents of cytosol
and mitochondrial cytochrome c in HBE cells following PM exposure for 24 h (n = 3/group, one-way ANOVA). h ROS generation in HBE cells
following PM exposure for 24 h (n = 3/group, one-way ANO VA). i Schematic of the cellular responses to low-dose PM exposure in HBE cells. *
p < 0.05, ** p < 0.01, *** p < 0.001, compared to the PM (0 μg/mL) group
(See figure on next page.)
Page 6 of 16
Lietal. Current Medicine (2024) 3:9
Fig. 2 (See legend on previous page.)
Page 7 of 16
Lietal. Current Medicine (2024) 3:9
2.17 Short interfering RNA (siRNA) construction andcell
transfection
To knockdown the expression of SP1 and MT1F, SP1-
or MT1F- targeting siRNA and scrambled negative
control (NC) sequences were designed and synthe-
sized by GenePharma (Shanghai, China). HBE cells
were seeded in six-well plates and transfected with siR-
NAs using Lipofectamine 2000 Transfection Reagent
Fig. 3 High-concentration PM exposure leads to apoptosis in HBE cells. a Chord diagram of apoptotic GO terms and involved genes in 500 μg/mL
PM-exposed HBE cells. b Heatmap of apoptosis-related genes in HBE cells following PM exposure for 24 h. c qRT-PCR assays of apoptosis-related
gene expression levels in HBE cells following PM exposure for 24 h (n = 9/group, one-way ANOVA). d Cellular apoptosis induced by PM exposure
in HBE cells (n = 3/group, one-way ANOVA). e Levels of caspase-3, -8, 9 in HBE cells following PM exposure (n = 3/group, one-way ANOVA). f
Schematic of the cellular responses to high-dose PM exposure in HBE cells. * p < 0.05, ** p < 0.01, *** p < 0.001, compared to the PM (0 μg/mL) group
Page 8 of 16
Lietal. Current Medicine (2024) 3:9
(Invitrogen, USA) combined with Opti-MEM (Gibco,
USA) in line with the manufacturer’s protocol. Forty-
eight hours after transfection, knockdown efficiency was
verified by qRT-PCR or WB. e sequences of siRNAs
are listed in TableS3.
2.18 Coenzyme Q10 (Co Q10) supplementation
HBE cells were treated with Co Q10 (2.5 or 5.0 μM,
C9538, Merck, Germany) for 6h, then exposed to 0, 100,
or 500μg/mL PM for 24h. Co Q10 was dissolved in etha-
nol as a stock solution. e stock solution was incubated
at 37 for 15min prior to addition to the HBE cells. Fol-
lowing evaluation of gene expression levels, a single dose
of 5.0μM Co Q10 supplement was used for ROS genera-
tion, caspase-3, -8, -9 ELISA assay, and apoptosis detec-
tions in HBE cells. e control group were supplemented
with an equal volume of ethanol as in the 5.0μM Co Q10
group.
2.19 MT supplementation
HBE cells were treated with 1μg/mL MT (rabbit liver,
ALX-202–070, ENZO, USA)) dissolved in 0.9% saline for
6 h (Nygaard etal. 2015; Leung etal. 2018), then exposed
to 0, 100, or 500 μg/mL PM for 24 h. Gene expression
levels, ROS generation, caspase-3, -8, -9 ELISA assay, and
apoptosis were evaluated in HBE cells as described above.
2.20 Histopathological analysis
Lung tissues were fixed in PFA for 24h at 4°C, embed-
ded in paraffin, serially sectioned (5μm) and mounted
on silane-covered slides. e sections selected from each
mouse were stained after dewaxing with hematoxylin
and eosin (H&E) and evaluated under a light microscope
(400 ×) to examine tissue histology. Images were scanned
with the slide scanner Panoramic SCAN (3DHISTECH,
Hungary) to obtain a whole slide image. Histology of
murine lung tissues were evaluated by an experienced
histologist who was blinded to experimental design.
2.21 Statistical analysis
Data are shown as means ± SD, unless indicated oth-
erwise. For the comparison among multiple groups,
one-way or two-way analysis of variance (ANOVA) fol-
lowed with Dunnett’s multiple comparisons was used, as
indicated in the figure legends. e results of qRT-PCR
were analyzed by 2ΔΔCt method. Statistical significance
was set at p < 0.05.
3 Results
3.1 mRNA expression proles inHBE cells were altered
byPM exposure
e cellular viability of HBE cells was inhibited in a
concentration-dependent manner following 24, 48, or
72 h PM exposure. e significant decrease in cellular
viability (80.21 ± 7.63%, 74.62 ± 8.82%, 70.90 ± 7.61%,
and 47.65 ± 3.56%, relative to the control) occurred
in HBE cells following 100, 250, 500, and 1000 μg/mL
PM exposure for 24 h, respectively (Fig.1a). Two doses
(100 and 500 μg/mL) of PM exposure, which resulted
in cellular viability reduction of 20% and 30%, respec-
tively, relative to the control, were chosen for the mRNA
microarray assays. A total of 140 and 758 up-regulated
genes, and 42 and 927 down-regulated genes were by
100 or 500 μg/mL in PM-exposed HBE cells for 24 h,
respectively, with a cut-off of > 1.5-fold change (FC) and
p-value < 0.05 (Fig. 1b). Venn diagram demonstrated
the number of differentially expressed genes in 100 and
500 μg/mL PM-exposed groups (Fig.1c). Gene ontol-
ogy (GO) of differentially expressed genes suggested
multiple enrichments related to mitochondrial oxida-
tive phosphorylation in the 100 μg/mL PM-exposed
HBE cells (Fig.1d and TableS4); and apoptosis in 500
μg/mL PM-exposed HBE cells (Fig.1e and Table S5).
As for overlapped genes between 100 and 500 μg/
mL PM-exposed groups (including 63 differentially
expressed genes), they were highly enriched in ion
homeostasis-related terms (Fig.1f and TableS6). ere-
fore, we hypothesized that PM exposure exerted effects
on HBE cells in a concentration-dependent manner.
At the relatively low concentration, PM exposure trig-
gered oxidative stress; with increased concentration,
ion homeostasis was disrupted, eventually resulting in
apoptosis in HBE cells (Fig.1g).
Fig. 4 Disruption of ion homeostasis contributes to apoptosis in HBE cells following PM exposure. a Chord diagram of ion homeostasis GO terms
and involved genes in 500 μg/mL PM-exposed HBE cells. b Heatmap of ion homeostasis-related genes in HBE cells following PM exposure for 24
h by microarray. c qRT-PCR assays of ion homeostasis-related genes in HBE cells following PM exposure for 24 h (n = 9/group, one-way ANOVA).
d qRT-PCR assays of MT-encoding genes in HBE cells following PM exposure for 24 h (n = 9/group, one-way ANOVA). e Schematic of transcription
factor SP1 binding to the promoter regions of MT-encoding genes and Expression levels of SP1 in PM-exposed HBE cells for 24 h (n = 3/group,
one-way ANOVA). f ChIP assays verified that SP1 binding promoted MT1X, MT1F, and MT1G transcription ((n = 6/group, t-test). g SP1 knockdown
inhibited expression of MT1F (n = 9/group, one-way ANOVA). h Apoptosis of HBE cells (control or SP1 KD) following PM exposure (n = 3/group,
two-way ANOVA). i Schematic of the cellular responses to PM exposure in HBE cells. * p < 0.05, ** p < 0.01, *** p < 0.001, compared to the PM (0 μg/
mL) group
(See figure on next page.)
Page 9 of 16
Lietal. Current Medicine (2024) 3:9
Fig. 4 (See legend on previous page.)
Page 10 of 16
Lietal. Current Medicine (2024) 3:9
3.2 PM exposure triggered oxidative stress bytargeting
phosphorylation (OXPHOS) inHBE cells
To confirm our hypothesis that relatively low concentra-
tion of PM triggered oxidative stress in HBE cells (Fig.2a),
the expression levels of 5 genes involved in mitochondrial
function and OXPHOS (Fig. 2b and c) were validated.
Consistent to the microarray data (Fig. 2c), expression
levels of NDUFC1, NDUFB1, NDUFB2, and ATP5E were
inhibited following 24 h PM exposure, and the expres-
sion levels of UQCR10 were significantly increased at 50
and 100 μg/mL groups, while decreased at 500 μg/mL
group (Fig.2d) compared to the control. Mitochondrial
functions were consistently disrupted by showing signifi-
cant collapse of the mitochondrial membrane potential
(Fig.2e), decreased ATP content (Fig.2f), increased cyto-
sol cytochrome c levels, and decreased mitochondrial
cytochrome c levels (Fig.2g) in HBE cells following PM
exposure relative to the control. PM exposure also sig-
nificantly induced ROS levels in HBE cells. Among all the
groups, the maximal ROS generation was observed in the
100 μg/mL PM-treated group (Fig.2h). erefore, it was
confirmed that HBE cells responds to relatively low PM
exposure by induction of oxidative stress (Fig.2i).
3.3 PM exposure resulted inapoptosis inHBE cells
To further confirm our hypothesis that high concen-
tration of PM exposure resulted in apoptosis in HBE
cells (Fig.3a), the expression levels of apoptosis-related
genes were validated by qRT-PCR assays. Consistent
to the microarray data (Fig.3b), 8 out of 10 apoptosis-
involved genes were significantly induced in HBE cells
following 24 h 500 μg/mL PM exposure (Fig.3c). e
flowcytometric assays showed significantly increased
apoptosis in 250 and 500 μg/mL PM-exposed HBE
cells for 24 h, relative to the control (Fig.3d). Accord-
ing to previous study, Caspase-related apoptosis can
proceed in intrinsic or extrinsic pathways (Shalini etal.
2015). In intrinsic apoptosis pathway, death-inducing
stimulus, such as ROS and DNA damage, lead to acti-
vation of apoptosome following Caspase-9 cleavage
and Caspase-3 activation. As for extrinsic apoptosis,
it depends on complex formation by death-domain-
containing proteins which activated Caspase-8 leading
to apoptosis. In Fig.3e, we identified the activation of
mitochondria-dependent (Caspase-9) and non-mito-
chondria-dependent (Caspase-8) apoptosis pathways
in HBE cells following low-concentration (50 and 100
μg/mL) and high-concentration (250 and 500 μg/mL)
PM exposure respectively. Our results indicated two
different pathways of PM-induced apoptosis in a dose-
dependent manner. ese results suggested that rela-
tively high dose of PM exposure damaged HBE cells by
inducing apoptosis (Fig.3f).
3.4 MT‑encoding genes consistently responded toPM
exposure inHBE cells
Bioinformatic analysis showed that ion homeostasis-
involved enrichments played a vital role in response to
PM exposure in HBE cells (Fig.4a). Among the 63 over-
lapped differentially expressed genes between 100 and
500 PM-exposed groups, a total of 7 genes were signifi-
cantly induced with twofold change compared to the
control (Fig.4b). Four of the 7 genes, including MAGOH,
NR1I2, CAPRIN2, and SAA1, were ion homeostasis GO
enrichments-involved genes and their expression levels
were significantly higher in PM-exposed groups com-
pared to the control (Fig. 4c). e remainder 3 genes,
including MT1F, MT1G, and MT1X, were MT-encoding
genes.
MT is a well-known antioxidant and its transcrip-
tions can be rapidly induced by stimuli, such as oxidative
stress and exogenous metals exposure (Andrews 2000).
Expression levels of MT-encoding genes were induced
in HBE cells following PM exposure (Fig.4d). Next, we
investigated their up-stream drivers. SP1 was predicted
to be a transcription factor binding to the promoter
region of MT1F, MT1X, and MT1G and its expression
levels were induced in HBE cells following PM expo-
sure (Fig.4e). ChIP assays showed that binding of SP1 to
the promoter regions of MT1F, MT1X, and MT1G was
enhanced in the PM-exposed group compared to the
control (Fig.4f).
Whether increased expression levels of MT-encoding
genes in HBE cells following PM exposure have bene-
ficial or deleterious role has yet to be determined. We
(See figure on next page.)
Fig. 5 MT efficiently protects HBE cells against PM exposure-induced apoptosis. a qRT-PCR assays of UQCR10 expression in HBE cells following PM
exposure with or without Co Q10 supplement (n = 9/group, two-way ANOVA). b ROS generation in HBE cells following PM exposure with or without
Co Q10 supplement (n = 3/group, two-way ANOVA). c Apoptosis n HBE cells following PM exposure with or without Co Q10 supplement (n = 3/
group, two-way ANOVA). d Schematic of the Co Q10 rescue against PM exposure-induced oxidative stress in HBE cells. e qRT-PCR assays of MT1X
and MT1G expression in HBE cells following PM exposure with or without MT supplement (n = 9/group, two-way ANOVA). f ROS generation in HBE
cells following PM exposure with or without MT supplement (n = 3/group, two-way ANOVA). g Apoptosis n HBE cells following PM exposure
with or without MT supplement (n = 3/group, two-way ANOVA). h Schematic of the MT rescue against PM exposure-induced apoptosis in HBE cells.
* p < 0.05, ** p < 0.01, *** p < 0.001, compared to the PM (0 μg/mL) group
Page 11 of 16
Lietal. Current Medicine (2024) 3:9
Fig. 5 (See legend on previous page.)
Page 12 of 16
Lietal. Current Medicine (2024) 3:9
constructed SP1 knock-down (KD) HBE cells and the
expression of SP1 were ablated in HBE cells following
PM exposure (Fig. S1a and Fig.4g) significantly attenu-
ated the induction of MT1F, MT1X, and MT1G expres-
sion levels in HBE cells following PM exposure (Fig.
S1b); whereas, expression levels of MAGOH, NR1I2,
CAPRIN2 were not transcriptionally regulated by SP1
in HBE cells following both 100 and 500 μg/mL PM
exposure; expression levels of CAPRIN2 and SAA1 were
significantly decreased in SP1-KD group only follow-
ing 500 μg/mL PM exposure. (Fig. S1c). Furthermore,
SP1 KD enhanced PM exposure-induced apoptosis in
HBE cells compared to the control (Fig.4h). As shown
in Fig.4d, the expression levels of MT1F were increased
in a dose-dependent manner following PM exposure,
we thus chose to knock down the expression of MT1F
(Fig. S1d) in HBE cells as an example to assess whether
MT-encoding genes act a protect role on PM-induced
apoptosis. Indeed, we observed high apoptosis level in
HBE cells following 24 h 100 and 500 μg/mL PM expo-
sure in MT1F KD group (Fig. S1e). erefore, the induc-
tion of MT-encoding genes was protective merely based
on the increase of PM-induced apoptosis caused by SP1
KD. ese results corroborated our hypothesis that with
the increased PM exposure levels, oxidative stress, ion
homeostasis disruption, and apoptosis were sequen-
tially induced in HBE cells (Fig.4i).
3.5 MT eectively protected HBE cells againstPM
exposure‑induced cellular damages
We further explored the potential rescue to PM expo-
sure-induced damage in HBE cells. CoQ10 is an elec-
tron carrier in the mitochondrial respiratory chain,
an antioxidant, and anti-apoptotic factor (Emma etal.
2016). Here, Co Q10 supplement (5.0 μM) rescued the
expression of UQCR10 to control levels in HBE cells fol-
lowing 100 μg/mL PM exposure (Fig.5a), but failed to
rescue the expression of NDUFB1, NDUFB2, NDUFC1,
and ATP5E (Fig. S2). Co Q10 supplement significantly
reduced ROS generation in 100 and 500 μg/mL PM-
exposed cells (Fig. 5b). However, Co Q10 supplement
only partially inhibited the levels of Caspase-3 and -9
in HBE cells (Fig. S3), and slightly attenuated apopto-
sis in 500 μg/mL PM-exposed HBE cells (Fig.5c). us,
though Co Q10 effectively inhibited ROS generation, it
was not an ideal rescue reagent against PM exposure
induced apoptosis in HBE cells (Fig.5d).
Since induction of MT-encoding genes effectively
inhibited PM exposure-induced apoptosis, we tested
the effects of MT supplement on HBE cells. Following
exogeneous MT supplement, the expressions of MT1F,
MT1X, and MT1G in PM-exposed HBE cells were sta-
tistically indistinguishable from control levels (Fig.5e
and Fig. S4a). The expressions of mitochondria-related
genes were not affected by MT supplement (Fig. S4b
to f). MT displayed anti-oxidative efficacy by inhibit-
ing ROS generation (Fig.5f), fully inhibited Caspase-3,
-8 and -9 induction (Fig. S5), and reduced apoptotic
proportions in PM-exposed HBE cells compared to the
control (Fig.5g and h).
3.6 MT supplement attenuated pulmonary injuries
inPM‑exposed mice
The representative image from FA/vehicle group
showed normal morphology of small airway, which
was composed of a single layer of cuboidal epithelia.
However, in the PM-exposed murine lung, hyperplasia
of the small airway epithelia was evident, which was
characterized by increased layers of surface respira-
tory epithelial cells and desquamation into the airway
lumen (L) (Fig. 6a). Images of PM/MT groups dem-
onstrated that MT supplement effectively reversed
the disruption in airway epithelium (Fig.6a). We next
determined ROS generation in murine lung tissues. As
expected, increased ROS generation in airway epithe-
lia were showed in the PM-exposed murine lungs, and
MT supplement significantly inhibited ROS generation
to statistically indistinguishable levels from the control
(Fig.6b and c). Finally, TUNEL assays were performed
to evaluate the apoptosis in airway epithelia, which
consistently corroborated ROS generation (Fig. 6d).
Taken together, our study suggested MT as a potential
rescue against PM exposure-induced pulmonary inju-
ries both invitro and invivo (Fig.6e).
4 Discussion
In the present study, we characterized the effects of PM
exposure on the bronchial epithelia. Mitochondrial dys-
function, disruption in ion homeostasis, and increased
Fig. 6 MT supplement attenuates bronchial epithelial oxidative stress and apoptosis in mice following PM exposure. a Representative images
of small airways in mice following PM exposure by H&E staining on lung tissue sections. L: lumen of airway b & c: Representative images of ROS
generation in murine lungs following PM exposure (n = 12/group, two-way ANOVA). d & e Representative images of TUNEL staining on lung tissue
sections in mice following PM exposure (n = 12/group, two-way ANOVA). f Schematic of MT as a potential rescue against PM exposure-induced
pulmonary injuries both in vitro and in vivo. * p < 0.05, ** p < 0.01, *** p < 0.001, compared to the PM (0 μg/mL) group
(See figure on next page.)
Page 13 of 16
Lietal. Current Medicine (2024) 3:9
Fig. 6 (See legend on previous page.)
Page 14 of 16
Lietal. Current Medicine (2024) 3:9
oxidative stress contributed apoptosis in response to PM
exposure. Notably, MT afforded anti-oxidative and anti-
apoptosis efficacy, effectively rescuing bronchial epithe-
lial cells from the damage induced by PM exposure in
both mammalian cells and animals.
Cumulative evidence supports a role for oxidative
stress as a critical pathway in response to PM exposure
(Yue etal. 2019; Ge etal. 2020; He etal. 2017). Endog-
enous ROS is generated from diverse sources, including
mitochondrial respiratory chain, NADPH oxidases, nitric
oxide synthases, and cytochrome P450 (Nathan and
Cunningham-Bussel 2013; Wende et al. 2016). Consist-
ent with previous studies, our results demonstrated that
PM exposure resulted in disturbance in the mitochon-
drial respiratory chains. OXPHOS dysfunction results in
two major effects, namely, reduced ATP production and
increased ROS production (Emma et al. 2016). In the
respiratory chain, the electron carrier Co Q10 shuttles
electrons from complexes I and II to complex III. Mild
Co Q10 deficiency has been associated with increased
ROS production, meanwhile, ATP production was not
significantly changed (Quinzii et al. 2010). Co Q10 is
also a well-known antioxidant and anti-apoptotic factor
(Emma etal. 2016). In the present study, CoQ10 sup-
plement partially ameliorated ROS generation, however
it failed to rescue apoptosis in HBE cells following PM
exposure. We thus concluded that CoQ10 acted as a gen-
eral anti-oxidant, but not a specific rescue to PM expo-
sure-induced cellular apoptosis.
Oxidative stress has been intensively associated with
disruption of ion homeostasis, especially the metal ion
homeostasis. Garza-Lombó et al. reviewed the role of
redox signaling played in the cellular toxicity linked to
xenobiotic metal exposure (Garza-Lombo et al. 2018).
ough most of these studies have been conducted in
the nervous system, they emphasized that disruption in
homeostasis of essential metals and exposure to xenobi-
otic metals altered the cellular redox status and signaling.
Metals are important toxic components of PM (Li etal.
2016). ese metal components caused a cellular redox
imbalance through cumulative formation of ROS, yet, via
distinct mechanisms (Goncalves etal. 2021; Valko etal.
2016; Ying etal. 2021; Skalny etal. 2021). erefore, over-
loading of metals from PM exposure is one of the major
triggers to oxidative stress invivo. Besides, literatures also
support the idea that ion homeostasis could be disturbed
by PM exposure. Water-soluble ionic components of PM
can easily dissolve in the wet alveolar wall and affect the
alveolar cells, which directly leads to imbalanced ion
homeostasis (Park etal. 2022). Air pollution, for example,
may reduce sodium excretion, which enhances the risk
of hypertension (Kwak and Kim 2023). e exposure of
PM2.5 disrupts intracellular iron leading to cell ferroptosis
in endothelial cells (Wang and Tang 2019). On the other
hand, PM-exposure induces the aberrant expression of
ion channel-related genes, which may cause disruption of
ion homeostasis, leading to arrhythmia-like cardiotoxic-
ity in zebrafish embryos (Park etal. 2023).
Previous studies have documented the contributions of
several antioxidant systems to the regulation of ROS and
maintenance of optimal redox status, including super-
oxide dismutases, catalases and the enzymes of the glu-
tathione redox cycle (Nathan and Cunningham-Bussel
2013). Compared with these well-known antioxidants,
MT have been considered a more effective antioxidant
against metal-associated oxidative stress (Carocho etal.
2018; Yu etal. 2023). e highly conserved number and
position of cysteine residues inherent to MT enable
them to bind physiological and xenobiotic heavy met-
als, as well as to reduce reactive oxygen species (Chen
etal. 2024). As a stress-induced protein, MT have been
reportedly involved in the underlying mechanisms to
maintain metal homeostasis in cells (Witkowska et al.
2021). Corroborating to our results, it has been reported
that MT are suitable candidates for exobiotic metal
exposure, for their intriguing feature of high binding
potential for various heavy metals, and their expressions
are markedly induced during exposure to environmen-
tal insults (Mehta etal. 2016). MT in humans contains
four subfamilies, among which MT1 is widely expressed
in tissue to balance homeostasis. Typically, MT1G can
scavenge ROS through enhancing activity of NRF2/
ARE signaling without changes in free iron abundance;
although MT1F and MT1X have been linked to cell pro-
liferation, cell cycle, and apoptosis, the role of MT1F
and MT1X involve in ROS was still unknown (Pavic
etal. 2019; Rodrigo etal. 2020). Our results confirmed
the protective role of MT-code gene, especially MT1F, in
PM-exposure induced oxidative stress, ion homeostasis
disruption, and apoptosis in HBE cells.
5 Conclusion
In summary, metal ion homeostasis and oxidative stress
are major up-stream triggers of apoptosis in airway
epithelia. In this context, herein, we show that induc-
tion of metal-binding antioxidants, MT, may serve as
to offset and protect airway epithelia from the adverse
effects of PM exposure.
Supplementary Information
The online version contains supplementary material available at https:// doi.
org/ 10. 1007/ s44194- 024- 00036-7.
Supplementary Material 1. Appendix A. Supplementary data. Supplemen-
tary figures 1 to 5, and supplementary tables 1 to 5.
Page 15 of 16
Lietal. Current Medicine (2024) 3:9
Acknowledgements
Not applicable
Authors’ contributions
Conceptualization, X.L., R.C.; Funding Acquisition, X.L., R.C.; Investigation, B.L.,
N.H., S.W., Q.M., S.W. Supervision, M.A., R.C.; Writing Original Draft, Review, and
Editing, X.L., M.A. All Authors revised and approved the final version of the
manuscript.
Funding
This work was financially supported by the National Natural Science Founda-
tion of China (81973084), Guangdong Provincial Natural Science Foundation
Team Project (2018B030312005).
Availability of data and materials
The mRNA microarray dataset supporting the conclusions of this article are
available in the GEO repository; the accession number is GSE138870.
Declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
Author Rui Chen is a member of the Editorial Board for Current Medicine. The
paper was handled by the other Editor and has undergone rigorous peer
review process. Author Rui Chen was not involved in the journal’s review of, or
decisions related to, this manuscript.
Author details
1 School of Public Health, Capital Medical University, Beijing 100069, China.
2 Key Laboratory of Environmental Medicine Engineering, Ministry of Educa-
tion, School of Public Health, Southeast University, Nanjing, China. 3 Depart-
ment of Molecular Pharmacology, Albert Einstein College of Medicine,
Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY 10461, USA. 4 Advanced
Innovation Center for Human Brain Protection, Capital Medical University, Bei-
jing 100069, China. 5 Institute for Chemical Carcinogenesis, Guangzhou Medi-
cal University, Guangzhou 511436, China. 6 School of Public Health, Southeast
University, Nanjing 210009, China.
Received: 10 February 2024 Accepted: 27 August 2024
References
Andrews GK. Regulation of metallothionein gene expression by oxidative
stress and metal ions. Biochem Pharmacol. 2000;59(1):95–104.
Barbier E, Carpentier J, Simonin O, et al. Oxidative stress and inflammation
induced by air pollution-derived PM2.5 persist in the lungs of mice after
cessation of their sub-chronic exposure. Environ Int. 2023;181:108248.
Bowe B, Xie Y, Yan Y, et al. Burden of cause-specific mortality associated
with PM2.5 air pollution in the United States. JAMA Netw Open.
2019;2(11):e1915834.
Carocho M, Ferreira I, Morales P, et al. Antioxidants and prooxidants: effects on
health and aging. Oxid Med Cell Longev. 2018;2018:1472708.
Chen YZ, Zhao J, Ye H, et al. Beneficial impact of cardiac heavy metal scavenger
metallothionein in sepsis-provoked cardiac anomalies dependent upon
regulation of endoplasmic reticulum stress and ferroptosis but not
autophagy. Life Sci. 2024;336:122291.
Crotty Alexander LE, Drummond CA, Hepokoski M, et al. Chronic inhalation
of e-cigarette vapor containing nicotine disrupts airway barrier function
and induces systemic inflammation and multiorgan fibrosis in mice. Am J
Physiol Regul Integr Comp Physiol. 2018;314(6):R834–847.
Emma F, Montini G, Parikh SM, et al. Mitochondrial dysfunction in
inherited renal disease and acute kidney injury. Nat Rev Nephrol.
2016;12(5):267–80.
Feng SL, Huang FF, Zhang YQ, et al. The pathophysiological and molecular
mechanisms of atmospheric PM2.5 affecting cardiovascular health: a
review. Ecotoxicol Environ Saf. 2023;249:114444.
Fu Y, Lu R, Cui J, et al. Inhibition of ATP citrate lyase (ACLY) protects airway epi-
thelia from PM2.5-induced epithelial-mesenchymal transition. Ecotoxicol
Environ Saf. 2019;167:309–16.
Gangwar RS, Bevan GH, Palanivel R, et al. Oxidative stress pathways of air pollu-
tion mediated toxicity: recent insights. Redox Biol. 2020;34:101545.
Garza-Lombo C, Posadas Y, Quintanar L, et al. Neurotoxicity linked to dysfunc-
tional metal ion homeostasis and xenobiotic metal exposure: redox sign-
aling and oxidative stress. Antioxid Redox Signal. 2018;28(18):1669–703.
Ge C, Tan J, Zhong S, et al. Nrf2 mitigates prolonged PM2.5 exposure-triggered
liver inflammation by positively regulating SIKE activity: protection by
Juglanin. Redox Biol. 2020;36:101645.
Goncalves J, Moog S, Morin A, et al. Loss of SDHB promotes dysregulated iron
homeostasis, oxidative stress, and sensitivity to ascorbate. Cancer Res.
2021;81(13):3480–94.
He M, Ichinose T, Yoshida S, et al. PM2.5-induced lung inflammation in mice:
differences of inflammatory response in macrophages and type II alveolar
cells. J Appl Toxicol. 2017;37(10):1203–18.
Kwak JH, Kim HJ. Alleviating air pollutant-associated hypertension by potas-
sium intake in Korean adults: a cross-sectional study from the 2012–2016
Korea National Health and Nutrition Examination Survey. Environ Sci
Pollut Res. 2023;30(29):73881–9.
Lan Y, Ng CT, Ong CN, et al. Transcriptomic analysis identifies dysregulated
genes and functional networks in human small airway epithelial cells
exposed to ambient PM2.5. Ecotoxicol Environ Saf. 2021;208:111702.
Leung JYK, Bennett WR, King AE, et al. The impact of metallothionein-II on
microglial response to tumor necrosis factor-alpha (TNFalpha) and
downstream effects on neuronal regeneration. J Neuroinflammation.
2018;15(1):56.
Li N, Hao M, Phalen RF, et al. Particulate air pollutants and asthma. A paradigm
for the role of oxidative stress in PM-induced adverse health effects. Clin
Immunol. 2003;109(3):250–65.
Li X, Lv Y, Hao J, et al. Role of microRNA-4516 involved autophagy
associated with exposure to fine particulate matter. Oncotarget.
2016;7(29):45385–97.
Li X, Yang H, Wu S, et al. Suppression of PTPN6 exacerbates aluminum oxide
nanoparticle-induced COPD-like lesions in mice through activation of
STAT pathway. Part Fibre Toxicol. 2017;14(1):53.
Li T, Zhang Y, Wang J, et al. All-cause mortality risk associated with long-term
exposure to ambient PM2.5 in China: a cohort study. Lancet Public
Health. 2018;3(10):e470–477.
Li B, Huang N, Wei S, et al. lncRNA TUG1 as a ceRNA promotes PM exposure-
induced airway hyper-reactivity. J Hazard Mater. 2021;416:125878.
Mehta J, Bhardwaj SK, Bhardwaj N, et al. Progress in the biosensing techniques
for trace-level heavy metals. Biotechnol Adv. 2016;34(1):47–60.
Meng Q, Li B, Huang N, et al. Folic acid targets splenic extramedullary
hemopoiesis to attenuate carbon black-induced coagulation-thrombosis
potential. J Hazard Mater. 2022;424(Pt B):127354.
Nathan C, Cunningham-Bussel A. Beyond oxidative stress: an immunologist’s
guide to reactive oxygen species. Nat Rev Immunol. 2013;13(5):349–61.
Nygaard SB, Lund NS, Larsen A, et al. Exogenous metallothionein potenti-
ates the insulin response at normal glucose concentrations in INS-1E
beta-cells without disturbing intracellular ZnT8 expression. Basic Clin
Pharmacol Toxicol. 2015;116(2):173–7.
Park S, Ku J, Lee SM, et al. Potential toxicity of inorganic ions in particulate mat-
ter: ion permeation in lung and disruption of cell metabolism. Sci Total
Environ. 2022;824:153818.
Park KH, Choi YJ, Min WK, et al. Particulate matter induces arrhythmia-like
cardiotoxicity in zebrafish embryos by altering the expression levels of
cardiac development- and ion channel-related genes. Ecotoxicol Environ
Saf. 2023;263:115201.
Pavic M, Turcic P, Ljubojevic M. Forgotten partners and function regulators of
inducible metallothioneins. Arh Hig Rada Toksikol. 2019;70(4):256–64.
Quinzii CM, Lopez LC, Gilkerson RW, et al. Reactive oxygen species, oxidative
stress, and cell death correlate with level of CoQ10 deficiency. FASEB J.
2010;24(10):3733–43.
Rodrigo MAM, Jimemez AMJ, Haddad Y, et al. Metallothionein isoforms as
double agents - Their roles in carcinogenesis, cancer progression and
chemoresistance. Drug Resist Updat. 2020;52:100691.
Page 16 of 16
Lietal. Current Medicine (2024) 3:9
Schantz MM, McGaw E, Wise SA. Pressurized liquid extraction of diesel and air
particulate standard reference materials: effect of extraction temperature
and pressure. Anal Chem. 2012;84(19):8222–31.
Shalini S, Dorstyn L, Dawar S, et al. Old, new and emerging functions of cas-
pases. Cell Death Differ. 2015;22(4):526–39.
Skalny AV, Aschner M, Jiang Y, et al. Molecular mechanisms of aluminum
neurotoxicity: update on adverse effects and therapeutic strategies. Adv
Neurotoxicol. 2021;5:1–34.
Valko M, Jomova K, Rhodes CJ, et al. Redox- and non-redox-metal-induced
formation of free radicals and their role in human disease. Arch Toxicol.
2016;90(1):1–37.
Wang Y, Tang M. PM2.5 induces ferroptosis in human endothelial cells through
iron overload and redox imbalance. Environ Pollut. 2019;254:112937.
Wende AR, Young ME, Chatham J, et al. Redox biology and the interface
between bioenergetics, autophagy and circadian control of metabolism.
Free Radic Biol Med. 2016;100:94–107.
Whyand T, Hurst JR, Beckles M, et al. Pollution and respiratory disease: can diet
or supplements help? A review. Respir Res. 2018;19(1):79.
Witkowska D, Slowik J, Chilicka K. Heavy metals and human health: possible
exposure pathways and the competition for protein binding sites. Mol-
ecules. 2021;26(19):6060.
Xu RR, Li ZG, Zhu XJ, et al. Acute effects of exposure to fine particulate matter
and ozone on lung function, inflammation and oxidative stress in healthy
adults. Ecotoxicol Environ Saf. 2022;243:114013.
Ying JF, Lu ZB, Fu LQ, et al. The role of iron homeostasis and iron-mediated ROS
in cancer. Am J Cancer Res. 2021;11(5):1895–1912.
Yu Y, Li Y, Tian XH, et al. Pb exposure causes non-linear accumulation of Pb in
D. melanogaster controlled by metallothionein B and exerts ecological
effects. Sci Total Environ. 2023;900:165680.
Yue W, Tong L, Liu X, et al. Short term Pm2.5 exposure caused a robust lung
inflammation, vascular remodeling, and exacerbated transition from
left ventricular failure to right ventricular hypertrophy. Redox Biol.
2019;22:101161.
Zhang Y. All-cause mortality risk and attributable deaths associated with long-
term exposure to ambient PM2.5 in Chinese adults. Environ Sci Technol.
2021;55(9):6116–27.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in pub-
lished maps and institutional affiliations.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Air pollution may increase the risk of hypertension (HTN) by increasing oxidative stress and inflammation, and reducing sodium excretion. Potassium intake may reduce the risk of HTN through sodium excretion and reduce inflammation and oxidative stress. This study is aimed at investigating the association between air pollutants and HTN and whether these associations differ based on the potassium intake data of Korean adults from the 2012–2016 Korean National Health and Nutrition Examination Survey (KNHANES). This cross-sectional study used data from KNHANES (2012–2016) combined with annual air pollutant data from the Ministry of Environment using administrative units. We included 15,373 adults who responded to the semi-food frequency questionnaire. Associations between ambient PM10, SO2, NO2, CO, and O3 with HTN according to potassium intake were assessed using the survey logistic regression model for complex samples analysis. After adjusting for potential covariates such as, age, sex, education level, smoking status, family income, alcohol consumption, body mass index (BMI), exercise status, and survey year, as the air pollutant score considering the five air pollutants increased (severe air pollution), the prevalence of HTN increased in a dose-dependent manner (p for trend < 0.001). Meanwhile, in the adults with higher potassium intake and who were exposed to lowest levels of air pollutants score (score = 0), the ORs of HTN were significantly lower (OR = 0.56, 95% CI: 0.32–0.97). In conclusion, our study suggests that exposure to air pollutants may increase the prevalence of HTN among Korean adults. However, high potassium intake may help prevent HTN caused by air pollutants.
Article
Full-text available
Heavy metals enter the human body through the gastrointestinal tract, skin, or via inhalation. Toxic metals have proven to be a major threat to human health, mostly because of their ability to cause membrane and DNA damage, and to perturb protein function and enzyme activity. These metals disturb native proteins’ functions by binding to free thiols or other functional groups, catalyzing the oxidation of amino acid side chains, perturbing protein folding, and/or displacing essential metal ions in enzymes. The review shows the physiological and biochemical effects of selected toxic metals interactions with proteins and enzymes. As environmental contamination by heavy metals is one of the most significant global problems, some detoxification strategies are also mentioned.
Article
Pb pollution can harm human health and the ecosystem. Therefore, it is worthwhile to study the metabolic processes of heavy metals in individual bodies and their influence on ecological systems. In this work, we analyzed the genetic responses and physiological changes of D. melanogaster which took diets exposed to different doses of Pb using transcriptomic analysis, ICP-MS, and various other physiological methods. We found that the Pb accumulated in D. melanogaster in a nonlinear pattern with the increase of Pb content in food. Metallothioneins (Mtns), especially the MtnB directly affects the accumulation and excretion of metal Pb in D. melanogaster, and causes the nonlinear accumulation. Metal regulatory transcription factor-1 (MTF-1) is involved in the regulation of Pb-induced high expressions of Mtns. Furthermore, an interaction between the metal metabolism pathway and xenobiotic response pathway leads to the cross-tolerances of Pb-exposed D. melanogaster to insecticides and other toxins. The oxidative stress induced by Pb toxicity may be the bridge between them. Our findings provide a physiological and molecular genetic basis for further study of the accumulation and metabolism of Pb in D. melanogaster.
Article
Air pollution is a risk factor that increases cardiovascular morbidity and mortality. In this study, we investigated the cardiotoxicity of particulate matter (PM) exposure using a zebrafish embryo model. We found that PM exposure induced cardiotoxicity, such as arrhythmia, during cardiac development. PM exposure caused cardiotoxicity by altering the expression levels of cardiac development (T-box transcription factor 20, natriuretic peptide A, and GATA-binding protein 4)- and ion-channel (scn5lab, kcnq1, kcnh2a/b, and kcnh6a/b)-related genes. In conclusion, this study showed that PM induces the aberrant expression of cardiac development- and ion channel-related genes, leading to arrhythmia-like cardiotoxicity in zebrafish embryos. Our study provides a foundation for further research on the molecular and genetic mechanisms of cardiotoxicity induced by PM exposure.
Article
Background Exposure to ambient fine particulate matter (PM2.5, with aerodynamic diameter less than 2.5 µm) is a leading environmental risk factor for global cardiovascular health concern. Objective To provide a roadmap for those new to this field, we reviewed the new insights into the pathophysiological and cellular/molecular mechanisms of PM2.5 responsible for cardiovascular health. Main findings: PM2.5 is able to disrupt multiple physiological barriers integrity and translocate into the systemic circulation and get access to a range of secondary target organs. An ever-growing body of epidemiological and controlled exposure studies has evidenced a causal relationship between PM2.5 exposure and cardiovascular morbidity and mortality. A variety of cellular and molecular biology mechanisms responsible for the detrimental cardiovascular outcomes attributable to PM2.5 exposure have been described, including metabolic activation, oxidative stress, genotoxicity, inflammation, dysregulation of Ca²⁺ signaling, disturbance of autophagy, and induction of apoptosis, by which PM2.5 exposure impacts the functions and fates of multiple target cells in cardiovascular system or related organs and further alters a series of pathophysiological processes, such as cardiac autonomic nervous system imbalance, increasing blood pressure, metabolic disorder, accelerated atherosclerosis and plaque vulnerability, platelet aggregation and thrombosis, and disruption in cardiac structure and function, ultimately leading to cardiovascular events and death. Therein, oxidative stress and inflammation were suggested to play pivotal roles in those pathophysiological processes. Conclusion Those biology mechanisms have deepen insights into the etiology, course, prevention and treatment of this public health concern, although the underlying mechanisms have not yet been entirely clarified.
Article
Both fine particulate matter (PM2.5) and ozone (O3) may have adverse effects on human health. However, previous studies on the effects of air pollutants mainly have focused on susceptible population, and evidence on healthy young adults is limited. We aimed to examine the associations of the two main air pollutants (PM2.5 and O3) with lung function, inflammation and oxidative stress in healthy young adults. We recruited 30 healthy young adults for a longitudinal panel study in Beijing and implemented health examination seven times, including lung function (FEV1 and PEF) and biomarkers of inflammation and oxidative stress (i.e. C-reactive protein, CRP; interleukin-6, IL-6; malondialdehyde, MDA) from December 2019 to May 2021. Hourly ambient air pollutants data were obtained from the closest air quality monitoring station. Linear mixed-effect model was applied to explore the associations between air pollutants and lung function, inflammation and oxidative stress. We observed higher PM2.5 exposure was associated with decrement in lung function and increment in CRP and MDA. Each 10 μg/m³ increase in PM2.5 (lag 2 day) is associated with a 17.06 ml (95% CI: −31.53, −2.58) decrease in FEV1, 46.34 ml/s (95% CI: −76.41, −16.27) decrease in PEF and increments of 2.86% (95% CI: 1.47%, 4.27%) in CRP, 1.63% (95% CI: 0.14%, 3.14%) in MDA respectively. However, there is no significant association between ozone exposure and health indicators. The study suggested that short-term exposure to PM2.5 may decrease lung function and induce inflammation and oxidative stress in healthy adults, but there is no association between O3 and each outcome.
Article
Exposure to ambient particulate matter (PM) is associated with adverse health effects. Yet, due to the complexity of its chemical composition, the molecular effects of PM exposure and the mechanism of PM-mediated toxicity remain largely unknown. Here, we show that water-soluble inorganics such as nitrate and sulfate ions, rather than PM itself, rapidly penetrate the lung surfactant barrier to the alveolar region and perturb gene expression in the lungs. Through high-throughput sequencing of lung adenocarcinoma cells, we find that exposure to nitrate and sulfate ions activates the cholesterol biosynthetic metabolism and induces the expression of genes related to tumorigenesis. Transcriptome analysis of mouse lungs exposed to nitrate/sulfate aerosols reveals interferon gamma-associated immune response. Interestingly, we find that exposure to a nitrate/sulfate mixture leads to a unique gene expression pattern that is not observed when nitrate or sulfate is treated alone. Our work suggests that the water-soluble ions are a potential source of PM-mediated toxicity and provides a roadmap to unveil the molecular mechanism of health hazards from PM exposure.
Article
Due to its wide applications in tire and rubber products, carbon black (CB) implicates concerns on its safety during production, collection, and handling. Here we report that exposure CB, increases coagulation-thrombosis potential in a splenic extramedullary hemopoiesis (EMH)-dependent manner. Adult C57BL/6 mice are kept in whole-body inhalation chambers, and exposed to filtered room air (FRA) or CB for 28 consecutive days. CB exposure resulted in splenic EMH characterized with platelet precursor cells, megakaryocytes (MKs), hyperplasia and enhanced in vivo blood coagulation ability. Metabolomics analysis suggests significant enhance in PGE2 production but reduction in folic acid (FA) levels in murine serum following CB exposure. Mechanistically, activation of COX-dependent PGE2 production promotes IL-6 expression in splenic macrophages, which subsequently results in splenic EMH and increased platelet counts in circulation. Administration of FA protects the mice against CB-induced splenic EMH through inhibiting prostaglandin-endoperoxide synthase 2 (Ptgs2 or Cox2) and prostaglandin E synthase (Ptges) expression in splenic macrophages, eventually recover the coagulation capacity to normal level. The results strongly suggest the involvement of splenic EMH in response to CB exposure and subsequently increased coagulation-thrombosis potential. Supplementation with FA may be a candidate to prevent thrombosis potential attributable to CB exposure.