ArticlePDF Available

Melatonin Alleviates PM2.5-Induced Hepatic Steatosis and Metabolic-Associated Fatty Liver Disease in ApoE-/- Mice

Wiley
Oxidative Medicine and Cellular Longevity
Authors:

Abstract and Figures

Background: Exposure to fine particulate matter (PM2.5) is associated with the risk of developing metabolic-associated fatty liver disease (MAFLD). Melatonin is the main secreted product of the pineal gland and has been reported to prevent hepatic lipid metabolism disorders. However, it remains uncertain whether melatonin could protect against PM2.5-induced MAFLD. Methods and results: The purpose of our study was to investigate the mitigating effects of melatonin on hepatic fatty degeneration accelerated by PM2.5 in vivo and in vitro. Histopathological analysis and ultrastructural images showed that PM2.5 induced hepatic steatosis and lipid vacuolation in ApoE-/- mice, which could be effectively alleviated by melatonin administration. Increased ROS production and decreased expression of antioxidant enzymes were detected in the PM2.5-treated group, whereas melatonin showed recovery effects after PM2.5-induced oxidative damage in both the liver and L02 cells. Further investigation revealed that PM2.5 induced oxidative stress to activate PTP1B, which in turn had a positive feedback regulation effect on ROS release. When a PTP1B inhibitor or melatonin was administered, SP1/SREBP-1 signalling was effectively suppressed, while Nrf2/Keap1 signalling was activated in the PM2.5-treated groups. Conclusion: Our study is the first to show that melatonin alleviates the disturbance of PM2.5-triggered hepatic steatosis and liver damage by regulating the ROS-mediated PTP1B and Nrf2 signalling pathways in ApoE-/- mice. These results suggest that melatonin administration might be a prospective therapy for the prevention and treatment of MAFLD associated with air pollution.
This content is subject to copyright. Terms and conditions apply.
Research Article
Melatonin Alleviates PM
2.5
-Induced Hepatic Steatosis and
Metabolic-Associated Fatty Liver Disease in ApoE
-/-
Mice
Zhou Du,
1,2
Shuang Liang,
1,2
Yang Li,
1,2
Jingyi Zhang,
1,2
Yang Yu,
1,2
Qing Xu,
3
Zhiwei Sun ,
1,2
and Junchao Duan
1,2
1
Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
2
Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
3
Core Facilities for Electrophysiology, Core Facilities Center, Capital Medical University, Beijing 100069, China
Correspondence should be addressed to Zhiwei Sun; zwsun@ccmu.edu.cn and Junchao Duan; jcduan@ccmu.edu.cn
Received 6 March 2022; Accepted 6 May 2022; Published 8 June 2022
Academic Editor: Reiko Matsui
Copyright © 2022 Zhou Du et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Background. Exposure to ne particulate matter (PM
2.5
) is associated with the risk of developing metabolic-associated fatty liver
disease (MAFLD). Melatonin is the main secreted product of the pineal gland and has been reported to prevent hepatic lipid
metabolism disorders. However, it remains uncertain whether melatonin could protect against PM
2.5
-induced MAFLD.
Methods and Results. The purpose of our study was to investigate the mitigating eects of melatonin on hepatic fatty
degeneration accelerated by PM
2.5
in vivo and in vitro. Histopathological analysis and ultrastructural images showed that
PM
2.5
induced hepatic steatosis and lipid vacuolation in ApoE
-/-
mice, which could be eectively alleviated by melatonin
administration. Increased ROS production and decreased expression of antioxidant enzymes were detected in the PM
2.5
-treated
group, whereas melatonin showed recovery eects after PM
2.5
-induced oxidative damage in both the liver and L02 cells.
Further investigation revealed that PM
2.5
induced oxidative stress to activate PTP1B, which in turn had a positive feedback
regulation eect on ROS release. When a PTP1B inhibitor or melatonin was administered, SP1/SREBP-1 signalling was
eectively suppressed, while Nrf2/Keap1 signalling was activated in the PM
2.5
-treated groups. Conclusion. Our study is the rst
to show that melatonin alleviates the disturbance of PM
2.5
-triggered hepatic steatosis and liver damage by regulating the ROS-
mediated PTP1B and Nrf2 signalling pathways in ApoE
-/-
mice. These results suggest that melatonin administration might be a
prospective therapy for the prevention and treatment of MAFLD associated with air pollution.
1. Introduction
Health risks associated with particulate air pollution have
become a major focus of global concern due to rapid popu-
lation growth, industrialization, and urbanization. Fine par-
ticulate matter at a size of 2.5 μm (PM
2.5
) has been
considered as a strong potential threat to public health that
it can penetrate through the alveoli of lungs into the sys-
temic circulation and accumulate in the liver, kidney, or
brain [13]. Recently, a precise imaging technique was
developed to visualize the deposition of PM
2.5
particles in
the liver through inhalation, providing solid evidence that
the PM
2.5
particles can enter the extrapulmonary organs
[4]. Toxicological studies have demonstrated that the toxic-
ity of PM
2.5
not only induces respiratory and cardiovascular
morbidities but also contributes to other unfavourable out-
comes, such as systemic metabolic disorder, obesity, and
the pathogenesis of metabolic-associated fatty liver disease
(MAFLD) [2, 5], eventually resulting in liver dysfunction
and damage. Consistent with evidence from animal studies,
a prospective cohort study showed that people living in areas
with higher PM
2.5
concentrations had a 34% higher inci-
dence of MAFLD than those living in areas with lower
PM
2.5
concentrations. The hazard ratio (HR) of MAFLD
was 1.06 for every 1 μg/m
3
increase in PM
2.5
[6]. MAFLD
covers a broad spectrum of liver abnormalities from hepatic
steatosis to inammation and has become one of the main
cause of cirrhosis and liver cancer. Its prevalence continues
to progress universally, keeping pace with the obesity epi-
demic, reaching 20%-30% of the total population, 8090%
Hindawi
Oxidative Medicine and Cellular Longevity
Volume 2022, Article ID 8688643, 24 pages
https://doi.org/10.1155/2022/8688643
of obese individuals, and even more subjects with type 2 dia-
betes mellitus (T2DM) [7].
The mechanism for the pathophysiology of MAFLD was
initially explained by the two-hithypothesis. The rst hit is
that insulin resistance leads to enhanced hepatic de novo
lipogenesis and decreased lipolysis. Mice exposed to PM
2.5
have been demonstrated to develop MAFLD, characterized
by changes in liver appearance, extensive distribution of
lipid vacuoles, and balloon-like degeneration within the lob-
ular structure [5, 8]. The accumulation of free fatty acid ux
in hepatic cells further triggers a second hitinvolving oxi-
dative stress and lipid peroxidation [9]. It has also been
reported that PM
2.5
exposure induces excessive oxygen spe-
cies (ROS) production and redox homeostasis disorder [10,
11]. In brief, oxidative stress appears to be an integral mech-
anism that conveys hepatic injury in MAFLD and plays a
well-described role in mediating the toxicity of PM
2.5
[12].
However, the specic mechanism by which PM
2.5
exposure
promotes the risk of oxidative stress-driven MAFLD
remains incompletely understood.
A growing body of evidence in the cellular and molecular
biology of lipid metabolism have shown that protein tyro-
sine phosphatase 1B (PTP1B) is a new activator in the pro-
cess of MAFLD that regulates lipogenesis in the liver [13,
14]. Total PTP1B protein levels were generally upregulated
in liver biopsies from patients with MAFLD [15]. Function-
ally, PTP1B deciency prevents the adverse metabolic eects
of a high-fat diet, including weight gain, increased liver
lipids, and reduced glucose tolerance [16]. PTP1B
-/-
mice
also exhibited downregulation of genes involved in fat pro-
duction, including sterol regulatory element-binding pro-
teins (SREBPs) [17]. Furthermore, SREBP-1 could
extensively aect multiple metabolic steps in the liver and
extranet, thereby regulating the progression of MAFLD
[18]. However, it is still not clear whether PM
2.5
has a tar-
geted regulatory eect on PTP1B.
Melatonin is well-known for its ability to neutralize ROS
and reduce oxidative stress [19]. It upregulates nuclear factor
erythroid 2-related factor 2 (Nrf2) through inhibition of
Kelchlike ECH-associated protein (Keap1) to suppress
oxidative stress in the liver [20]. Investigations have noted
that melatonin critically participates in lipid metabolism
and potentially contributes to the onset and progression of
MAFLD [21, 22]. However, it remains uncertain whether
melatonin could protect against PM
2.5
-induced oxidative
stress in the liver and ameliorate MAFLD.
Compared with the general population, people with obe-
sity, hyperlipidaemia, or abnormal lipid metabolism are
more sensitive to PM
2.5
and have a higher risk of developing
MAFLD [23, 24]. According to a cohort study of full-exome
association of alanine aminotransferase, ApoE was found
closely linked with fatty liver [25], and allele-specic variants
of ApoE were associated with an increased incidence of
MAFLD and obesity [26]. Thus, intense eorts have been
made to investigate MAFLD based on ApoE
-/-
mice
[2730]. Hua et al. demonstrated that naringin administra-
tion improved metabolic parameters in ApoE
-/-
mice, inhib-
ited hepatic steatosis, and reduced hepatic brosis [31].
Stachowicz et al. found that high fat diet resulted in more
exacerbated hepatic steatosis in ApoE
-/-
mice [32]. In this
study, ApoE
-/-
mice were chosen as an animal model to
explore the molecular mechanism of the melatonin-
mediated protective eects against PM
2.5
-induced MAFLD.
We speculated that melatonin may ameliorate PM
2.5
-
induced MAFLD. Our ndings supported this hypothesis
and further indicated that melatonin alleviated the distur-
bance of PM
2.5
-triggered hepatic steatosis and liver damage
by regulating the ROS-mediated PTP1B and Nrf2 signalling
pathways. These results not only provide novel insight into
the underlying molecular mechanism by which PM
2.5
con-
tributes to the pathogenesis of MAFLD but also suggest the
use of melatonin as a potential treatment.
2. Materials and Methods
2.1. Collection and Extraction of PM
2.5
.PM
2.5
was collected
on quartz bre lters with a special sampler (TH-1000C,
Wuhan Tianhong, China) from Capital Medical University
Table 1: The primer lists of realtime PCR.
Primer Forward primer (5-3) Reverse primer (5-3)
mus-SOD AAGGGAGATGTTACAACTCAGG GCTCAGGTTTGTCCAGAAAATG
hsa-SOD CCCGACCTGCCCTACGACTAC AACGCCTCCTGGTACTTCTCCTC
mus-Keap1 GACTGGGTCAAATACGACTGC GAATATCTGCACCAGGTAGTCC
hsa-Keap1 ATTCAGCTGAGTGTTACTACCC CAGCATAGATACAGTTGTGCAG
mus-Nrf2 CAGCCATGACTGATTTAAGCAG CAGCTGCTTGTTTTCGGTATTA
hsa-Nrf2 TCCAAGTCCAGAAGCCAAACTGAC GGAGAGGATGCTGCTGAAGGAATC
mus-SREBP1 GCTACCGGTCTTCTATCAATGA CGCAAGACAGCAGATTTATTCA
hsa-SREBP1 CTGTGTGACCTGCTTCTTGT CTCATGTAGGAACACCCTCC
mus-SP1 GAAGCAGCAGCACAGGCAGTAG GCCAGCAGAGCCAAAGGAGATG
hsa-SP1 TCACTCCATGGATGAAATGACA CAGAGGAGGAAGAGATGATCTG
mus-PP2A AGTTACACTGCTTGTAGCTCTT AACCCATAAACCTGTGTGATCT
hsa-PP2A CGAAGGTGTGAAGGGGAAGAAGC CAGCGTGTTGAGAAGAGCGACTAG
mus-PTP1B GAGAGATCCTGCATTTCCACTA TACTTTCTTGATGTCCACGGAA
hsa-PTP1B CCATTTACCAGGATATCCGACA TGACGTCTCTGTACCTATTTCG
2 Oxidative Medicine and Cellular Longevity
Con
(a)
(b)
(c)
(d)
1.0
(e)
(f) (g) (h)
0.8
0.6
Ratio of liver size to body weight
Oil red O staining area (%)
T-CHO (mmol/g protein)
TAGs (mmol/g protein)
0.4
0.2
0.0
100
80
60
40
20
0
0.25 3
2
1
0
0.20
0.15
0.10
0.05
0.00
Sag
Tra ns
Con
Mel
PM
2.5
PM
2.5
+Mel
Con
Mel
PM
2.5
PM
2.5
+Mel
Con
Mel
PM
2.5
PM
2.5
+Mel
Mel PM2.5 PM2.5+Mel
Con
Mel
PM2.5
PM2.5+Mel
Figure 1: Melatonin improved the increased lipid content and steatosis in the liver induced by PM
2.5
. (a) Ultrasound examination of
livercomparison of liver echo and kidney echo. (b) The ultrastructure of liver tissues via electron microscopy (magnication, 200; scale
bar, 2 μm). (c) Liver sections with haematoxylin and eosin (H&E) staining (magnication, 200 and 400; scale bar, 60 μm and 30 μm). (d)
Liver steatosis assessed by Oil Red O staining (magnication, 200 and 400; scale bar, 60 μm and 30 μm). (e) Liver sag (anterior-posterior
diameter) and liver trans (left-right diameter) measurement to mice weight ratio. (f) The ratio of the Oil Red O-stained area to the total
tissue area. (g) Hepatic total cholesterol lipid levels (mmol/g). (h) Hepatic triacylglycerol lipid levels (mmol/g). Con: animals were treated
with saline; Mel: animals were treated with melatonin; PM
2.5
: animals were treated with PM
2.5
;PM
2.5
+Mel: animals were treated with
melatonin and PM
2.5
. Data are shown as means ± SD.n=612 mice per group. P<0:05 for Con group vs PM
2.5
group and PM
2.5
group vs PM
2.5
+Mel group.
3Oxidative Medicine and Cellular Longevity
Con
DAPIFITCMERGE
Mel PM2.5 PM2.5+Mel
(a)
0.08
0.06
0.04
AOD (pixel)
0.02
0.00
Con
Mel
PM
2.5
PM
2.5
+Mel
⁎⁎
(b)
5
4
3
2
MDA (nmol/mgprrot)
1
0
Con
Mel
PM
2.5
PM
2.5
+Mel
⁎⁎
(c)
70
60
50
40
4-HNE (𝜇mol/L)
Con
Mel
PM2.5
PM2.5+Mel
⁎⁎
(d)
600
400
200
0
GSH-PX (𝜇mol/mgprot)
Con
Mel
PM2.5
PM2.5+Mel
(e)
Figure 2: Continued.
4 Oxidative Medicine and Cellular Longevity
100
80
60
SOD vatality (U/mgprot)
40
20
0
Con
Mel
PM2.5
PM2.5+Mel
⁎⁎
(f)
4
3
2
1
0
mRNA expression
Nrf2
Keap1
SOD
Con
Mel
PM2.5
PM2.5+Mel
#
##
(g)
Con Mel PM2.5 PM2.5+Mel
Nrf2
Keap1
SOD
GAPDH
(h)
1.5
1.0
0.5
0.0
Relative Nrf2 protein levels
Con
Mel
PM
2.5
PM
2.5
+Mel
(i)
2.0
1.5
1.0
0.0
0.5
Relative Keap1 protein levels
Con
Mel
PM
2.5
PM
2.5
+Mel
(j)
Figure 2: Continued.
5Oxidative Medicine and Cellular Longevity
(Beijing, China) for the entire year of 2017. The physico-
chemical characterization of PM
2.5
was described in detail
in our previous study. Tables S1 and S2 show the results of
element analysis. S, Ca, Na, Si, and Fe are the most
abundant elements. Toxic heavy metals (including Mn, Cd,
Cr, Ni, and Sb), toxic nonmetallic elements (As), and
water-soluble ions (NO
3-
, SO4
2-
, and NH4
+
) were detected
in PM
2.5
[33, 34]. Sampled lters were placed in ultrapure
water for 3 hours using an ice-water bath ultrasonic
instrument. Then, freeze-dried samples were irradiated
with ultraviolet light for 2 hours, diluted and mixed with
pure water, and suspended in PM
2.5
by ultrasonication for
30 minutes for later use.
2.2. Animals and Treatments. Seven-week-old male ApoE
-/-
mice (specic-pathogen free) were obtained from the Exper-
imental Laboratory Animal Technology Co., Ltd. (Vital
River, Beijing, China). Animal experimental procedures
were approved by the Experimental Animal Welfare Com-
mittee (Capital Medical University; AEEI-2016-076). All
mice were fed a high-fat diet (0.15% cholesterol and 21%
fat). After acclimatization for one week, a total of 60 mice
were randomly divided into four groups: (i) control group
(Con): animals were treated with saline; (ii) PM
2.5
group
(PM
2.5
): animals were treated with PM
2.5
; (iii) melatonin
group (Mel): animals were treated with melatonin; and (iv)
melatonin and PM
2.5
group (PM
2.5
+Mel): animals were
treated with PM
2.5
and melatonin. Mice were orally gavaged
with melatonin (20 mg/kgbw, in 20~25 μL of 0.5% ethanol
solution) daily and PM
2.5
(5 mg/kgbw, in 20~25 μLof
saline) via intratracheal instillation twice a week for 4 weeks.
The control mice received a corresponding volume of blank
lters eluted with saline by intratracheal instillation. The
vehicle mice were gavaged with the same amount of sterile
water (0.5% ethanol).
According to the concentration and intervention method
of melatonin in previous studies, melatonin (Sigma, USA)
was dissolved in absolute ethanol and diluted in sterile water
to a nal concentration of 0.5% ethanol, with the oral gavage
at a dose of 20 mg/kg/day [35, 36]. The dose of PM
2.5
expo-
sure was based on the respiratory physiological parameters
of mice and the annual mean PM
2.5
concentration (35 μg/
m
3
), according to the WHO air quality guidelines [37].
The respiratory volume of an adult mouse (25 g) is 0.15 mL
at each breath, and the breath rate is 163 times per min,
and respirato ry volume for one day reaches 0.035208 m
3
.
For this reason, the daily exposure of mice was 0:035208
35 μg/m3=1:23228 μg. Based on the body weight of mice
25 g and the extrapolation coecient of species 100, the vol-
ume of intratracheal instillation was 1:23228 μg/25 g 100
=4:93 μg/g. Previous studies have demonstrated that
PM
2.5
at 5 mg/kg can cause varying degrees of organ damage
[38, 39]. Therefore, a dose of 5 mg/kg was selected for animal
modeling.
2.3. Ultrasonic Examination of Liver. Before ultrasound
imaging, the mice were fasted for 12 h, the abdominal
regions were shaved, and then the mice were anaesthetized
with a saturated tribromoethanol solution via intraperito-
neal injection. We acquired transcutaneous ultrasound
images using a Vevo2100 Ultrasonic Doppler System (Fuji-
lm Visual Sonics, US).
2.4. Histopathological Examination. Both haematoxylin-
eosin (H&E) and Oil Red O staining are eective and repro-
ducible methods for quantifying hepatic steatosis [40]. For
1.5
1.0
0.5
0.0
Relative SOD protein levels
Con
Mel
PM2.5
PM2.5+Mel
⁎⁎
(k)
Figure 2: Melatonin improved liver oxidative damage induced by PM
2.5
. (a) Production of ROS detected by the uorescent probe DHE
(magnication, 200; scale bar, 20 μm). (b) Quantitative analysis of ROS production is reected by the mean uorescence intensity as
shown in dierent groups. (c) The level of MDA. (d) The level of 4-HNE. (e) The level of GSH-PX. (f) The vitality of SOD. (g) The
mRNA expression of Nrf2, Keap1, and SOD. (h) Western blotting of Nrf2, Keap-1, and SOD. (i) Protein quantication of Nrf2. (j)
Protein quantication of Keap1. (k) Protein quantication of SOD. All values are presented as the mean ± SD (n=6). P<0:05 for Con
group vs PM
2.5
group and
#
P<0:05 for PM
2.5
group vs PM
2.5
+Mel group.
6 Oxidative Medicine and Cellular Longevity
10
8
Con
Mel
PM2.5
PM2.5+Mel
6
4
2
0
mRNA expression
PTP1B
PP2A
SREBP-1
SP1
#
#
#
#
(a)
Con
PTP1B
PP2A
SP1
GAPDH
PPP2A
PSP1
SREBP1
Mel PM2.5 PM2.5+Mel
(b)
0.5
0.4
0.3
0.2
0.1
0.0
Con
Mel
PM2.5
PM2.5+Mel
Relative PTP1B protein levels
⁎⁎
(c)
0.0
0.5
1.0
1.5
Con
Mel
PM2.5
PM2.5+Mel
Relative PPP2A/PP2A protein levels
⁎⁎
(d)
0.6
0.4
0.2
0.0
Con
Mel
PM2.5
PM2.5+Mel
Relative PSP1/SP1 protein levels
⁎⁎
(e)
0.25
0.20
0.15
0.10
0.05
0.00
Con
Mel
PM2.5
PM2.5+Mel
Relative SREBP1 protein levels
⁎⁎
(f)
Figure 3: Continued.
7Oxidative Medicine and Cellular Longevity
histological examination, liver specimens were xed over-
night with 4% paraformaldehyde and then embedded in par-
an sections (46μm). Tissue sections were counterstained
with H&E. To visualize lipid droplet accumulation, frozen
liver sections (10 μm) were taken, stained with Oil Red O
(0.5%) for 10 min, washed and rinsed with isopropanol,
and counterstained with haematoxylin for a few seconds.
Representative photographs were taken at 200x and 400x
magnication using an in-microscope system. There were
6 samples in each group, and twenty regions were randomly
selected from each separate section. The color pickerin
Image-Pro-Plus was used to select the red fat droplets in
images until all the red fat droplets were marked. Then,
Oil Red O-stained area was measured, and its ratio to the
total tissue area was calculated.
2.5. Ultrastructural Observation by Transmission Electron
Microscopy (TEM). Lipid accumulation in the liver tissue
was observed by transmission electron microscopy. The liver
tissues were immediately placed into 2.5% glutaraldehyde
for 10 min at 4
°
C and then washed with PBS 3 times and
dehydrated. Sample sections (60 nm) were stained on copper
mesh and assessed using TEM (JEM-2100plus).
2.6. Detection of ROS Levels in Liver Tissue. Frozen sections
of the liver were washed, DHE solution (10 μM) was added,
and the sections were incubated at room temperature for
30 min. A confocal microscope (LSCM, TCS SP8 STED,
Germany) was used to capture uorescence images. There
were 6 samples in each group, and 3 visual elds were ran-
domly selected for each sample. Then, the ratio of red area
to total area was statistically analyzed by Image-Pro-Plus.
2.7. Cell Culture and Treatment. The human normal liver
cell line L02 was obtained from Shanghai Institutes for Bio-
logical Sciences (SIBS, China). Cells were cultured in Dul-
beccos modied Eagles medium (DMEM; Corning, USA)
containing 1% penicillin-streptomycin solution and 10%
foetal bovine serum (Corning, USA) at 37
°
C in a humidied
incubator with 5% CO
2
. Palmitic acid (PA) is an inducer for
cell steatosis. For treatment before each experiment, cells
were treated with PA solution dissolved in DMEM for
24 h. When the cell density reached 70%-80%, DMEM
(without serum) containing PM
2.5
or melatonin was added
and then cultured for 24 h. The control group was cultured
in a constant volume of pure medium.
2.8. Assessment of Cytotoxicity. A total of 1×10
4L02 cells
per well were seeded in 96-well culture plates. When the cell
density reached 50%, the L02 cells were exposed to gradient
concentrations of PM
2.5
(0, 12.5, 25, and 50 100 μg/mL), PA
(0, 50, 100, 200, 400, 800, and 1600 μmol/L), and melatonin
(0, 12.5, 25, 50, 100, and 200 μmol/L). According to the pro-
tocols, cell viabilities were measured by Cell Counting Kit-8
(CCK-8, Tongren, Japan), and the absorbance was measured
at 450 nm using a microplate reader (Thermo, USA).
2.9. Biochemical Parameter Analysis. Triacylglycerols
(TAGs), total cholesterol (TC), low-density cholesterol
(LDL-C), high-density cholesterol (HDL-C), glutathione
peroxidase (GSH-Px), superoxide dismutase (SOD), and
malonaldehyde (MDA) levels were measured spectropho-
tometrically according to the instructions of the kit (Nanjing
Jiancheng Institute of Biotechnology, Nanjing, China). Pro-
tein concentration was determined using a BCA protein
assay kit (Dingguo Changsheng Biotech, China). The 4-
hydroxynonenal (4-HNE) activity was determined using a
Hailian Biotechnology Co. Ltd. ELISA (enzyme-linked
immunosorbent assay) kit (Jiangxi, China).
2.10. Cellular BODIPY Staining. BODIPY493/503
(Thermo, USA) is a lipophilic uorescent probe targeting
polar lipids that can be used to label cell neutral lipid con-
tent, especially lipid content localized to lipid droplets. It
was dissolved in anhydrous ethanol to generate a 10 mM
stock solution, which was frozen, dried, and stored away
Con
Mel
PM2.5
PM2.5+Mel
PP2A activity (U/L)
3
2
1
0
(g)
Con
Mel
PM2.5
PM2.5+Mel
SP1 activity (𝜇g/ml)
0.4
0.3
0.2
0.1
0.0
(h)
Figure 3: Melatonin ameliorated abnormal liver lipid metabolism caused by elevated PTP1B expression induced by PM
2.5
. (a) The mRNA
expression of PTP1B, PP2A, SP1, and SREBP-1. (b) Western blotting of PTP1B, PP2A, P-PP2A, SP1, P-SP1, and SREBP-1. (c) Protein
quantication of PTP1B. (d) Protein quantication of P-PP2A/PP2A. (e) Protein quantication of P-SP1/SP1. (f) Protein quantication
of SREBP-1. (g) The activity of PP2A. (h) The activity of SP1. All values are presented as the mean ± SD (n=6). P<0:05 for Con
group vs PM
2.5
group and
#
P<0:05 for PM
2.5
group vs PM
2.5
+Mel group.
8 Oxidative Medicine and Cellular Longevity
PM2.5 concentration (𝜇g/ml)
0 6.25 12.5 25 50 100
0.5
0.6
0.7
0.8
0.9
Cell viability (% of control)
1.0
1.1
(a)
PM2.5 concentration (𝜇g/ml)
0 12.5 25 50 100
0.00
0.01
0.02
0.03
T-CHO (mmol/g protein)
0.04
0.05
(b)
PM2.5 concentration (𝜇g/ml)
0 12.5 25 50 100
0.00
0.05
0.10
0.15
TAGs (mmol/g protein)
0.20
(c)
PM2.5 concentration (𝜇g/ml)
0 12.5 25 50 100
0
2
4
6
8
ROS uorescence intensity
(fold control)
10
(d)
101
0
20
40
60
80
100
Count (%)
0 102103104
FITC-A
0 𝜇g/ml
105106107.2
12.5 𝜇g/ml
25 𝜇g/ml
50 𝜇g/ml
100 𝜇g/ml
(e)
Figure 4: Continued.
9Oxidative Medicine and Cellular Longevity
MERGE FITC DAPI
0 𝜇g/ml 12.5 𝜇g/ml 25 𝜇g/ml 50 𝜇g/ml 100 𝜇g/ml
(f)
PM2.5 concentration (𝜇g/ml)
0 12.5 25 50 100
PTP1B
PP2A
P-PP2A
SP1
P-SP1
SREBP-1
GAPDH
(g)
0
12.5
25
50
100
0.00
PM2.5 concentration
(𝜇g/ml)
0.05
0.10
0.15
0.20
Relative PTP1B protein levels
(h)
0
12.5
25
50
100
0.0
0.5
1.0
1.5
Relative P-PP2A/PP2A
protein levels
PM2.5 concentration
(𝜇g/ml)
(i)
0
12.5
25
50
100
0.0
0.2
0.4
0.6
0.8
1.0
Relative P-SP1/SP1 protein levels
PM2.5 concentration
(𝜇g/ml)
(j)
Figure 4: Continued.
10 Oxidative Medicine and Cellular Longevity
from light. PBS was used to dilute the solution to 10 μM,
which was used for incubation with the cells at room tem-
perature in the dark for 20 min; nally, the cells were
observed via a confocal microscope (LSCM, TCS SP8 STED,
Germany).
2.11. Detection of ROS Levels in L02 Cells. The level of ROS
in L02 cells was analyzed by ow cytometry. After the cells
were infected for 24 h, a 2,7-dichlorouorescein diacetate
(DCFH-DA, Sigma, USA) working solution (10 μM) was
added followed by incubation at 37
°
C for 30 min. The cells
were washed twice with PBS, and ROS levels were deter-
mined by ow cytometry. The single-parameter histograms
were obtained by taking the logarithm of uorescence signal
as abscissa and the number of cells as ordinate, which could
intuitively reect the relative intensity of ROS in living cells.
The average uorescence intensity was the number of cells
divided by the area under each peak. ROS uorescence was
measured with a confocal scanning laser microscope. To
quantify the ROS production in L02 cells treated with
0
12.5
25
50
100
0.00
PM2.5 concentration
(𝜇g/ml)
0.02
0.04
0.06
0.08
0.10
Relative SREBP-1 protein levels
(k)
0
12.5
25
50
100
0.0
0.2
0.4
0.6
0.8
Relative Nrf2 protein levels
PM2.5 concentration
(𝜇g/ml)
(l)
0
12.5
25
50
100
0.0
0.1
0.2
0.3
0.4
Relative Keap1 protein levels
PM2.5 concentration
(𝜇g/ml)
(m)
0
12.5
25
50
100
0.0
0.2
0.4
0.6
0.8
Relative SOD protein levels
PM2.5 concentration
(𝜇g/ml)
(n)
0 12.5 25 50 100
GAPDH
SOD
Keap1
Nrf2
PM2.5 concentration (𝜇g/ml)
(o)
Figure 4: PM
2.5
induced lipid accumulation in hepatocytes. (a) Cell viability. (b) Total cholesterol lipid levels (mmol/g). (c) Triacylglycerol
lipid levels (mmol/g). (d) Representative uorescence intensity images obtained from ow cytometry in L02 cells. (e) Analysis of
uorescence intensity obtained from ow cytometry. (f) Representative confocal images of ROS. (g) Western blotting of PTP1B, PP2A,
P-PP2A, SP1, P-SP1, and SREBP-1. (h) Protein quantication of PTP1B. (i) Protein quantication of P-PP2A/PP2A. (j) Protein
quantication of P-SP1/SP1. (k) Protein quantication of SREBP-1. (l) Protein quantication of Nrf2. (m) Protein quantication of
Keap1. (n) Protein quantication of SOD. (o) Western blotting of Nrf2, Keap1, and SOD. All values are presented as the mean ± SD.P
<0:05.
11Oxidative Medicine and Cellular Longevity
MERGE FITC DAPI
Con NAC PM2.5 PM2.5+NAC
(a)
0
0
20
40
60
80
100
101102103
Count (%)
104105
FITC-A
106107.2
Con
NAC
PM2.5
PM2.5+NAC
(b)
Con
NAC
PM2.5
PM2.5+NAC
ROS uorescence intensity
(fold of control)
0
1
2
3
4
5
⁎⁎
(c)
MERGE FITC DAPI
Con NAC PM2.5 PM2.5+NAC
(d)
GAPDH
SREBP-1
P-SP 1
SP1
P-PP2A
PP2A
PTP1B
Con NAC PM2.5 PM2.5+NAC
(e)
Con
NAC
PM2.5
PM2.5+NAC
0.0
0.2
0.4
0.6
0.8
Relative PTP1B protein levels
⁎⁎
(f)
Figure 5: Continued.
12 Oxidative Medicine and Cellular Longevity
PM
2.5
and/or melatonin, cells were pretreated with the ROS
inhibitor N-acetylcysteine (NAC; Sigma, USA) (1 mM) for
1 h before PM
2.5
and/or melatonin exposure.
DCFH-DA, intracellular reactive oxygen species detec-
tion probe, is a universal indicator of oxidative stress. After
it enters the cell, it is hydrolyzed to produce DCFH. Intracel-
lular reactive oxygen species can oxidize nonuorescent
DCFH to produce uorescent DCF. Intracellular reactive
oxygen species (ROS) levels were obtained by measuring
the uorescence intensity of DCF.
2.12. Real-Time Polymerase Chain Reaction (qPCR). Total
RNA from L02 cells and liver tissue was extracted using TRI-
zolReagent (Thermo, USA). According to the protocol,
the RNA was reverse transcribed into cDNA with Prime-
Script RT Master Mix (Takara, China). SYBR® Premix Ex
TaqII (Takara, China) was used for quantitative PCR on
a Realplex2 (Eppendorf, Germany). The mRNA primers
are listed in Table 1.
2.13. Western Blot Analysis. Protein extracts of mouse liver
tissue and L02 cells were prepared by a Whole Cell Lysis
Assay Kit (Keygen Biotech, China), and the concentrations
were determined by a BCA protein quantitative assay kit
(Dingguo Changsheng Biotech, China). The same amounts
of protein were separated by electrophoresis using 8%12%
SDS-PAGE gels and transferred to suitably sized nitrocellu-
lose membranes (Pall Corp., USA). After blocking with 5%
BSA or 5% skim milk in Tris-buered saline (TBS), the
membranes were incubated with primary antibodies at 4
°
C
overnight, including PTP1B (Abcam, UK), SP1 (Abcam,
UK), P-SP1 (Abcam, UK), SOD (Abcam, UK), Nrf2
(Abcam, UK), Keap1 (Abcam, UK), PP2A (Santa, USA), P-
PP2A (Santa, USA), SREBP-1 (Santa, USA), and GAPDH
(CST, USA). The next day, the membranes were washed
three times with TBST and incubated with an anti-rabbit/
mouse IgG secondary antibody (CST, USA). A LI-COR
Odyssey system (LI-COR Biosciences, USA) was used for
detection of the protein bands, which were quantied using
Image Studio software (NIH, Bethesda, MD).
2.14. The Addition of PTP1B Inhibitor. We dissolve PTP1B
inhibitor PTP1B-IN-1 (MedChemExpress, USA) in DMSO
to prepare a stock solution at a concentration of 20 mM.
Then, L02 cells were treated with PTP1B-IN-1 (5 μg/mL,
10 μg/mL, 20 μg/mL, and 40 μg/mL) diluted with DMEM
for 12 h. After qPCR anal ysis, 10 μg/mL was selected as the
dose of PTP1B-IN-1.
2.15. Statistical Analysis. SPSS 24.0 software was used to ana-
lyze all experimental data. Data are presented as the mean
±SD. Data consistent with a normal distribution and an
Con
NAC
PM2.5
PM2.5+NAC
0
1
2
3
Relative P-PP2A/PP2A
protein levels
⁎⁎
(g)
Con
NAC
PM2.5
PM2.5+NAC
0.0
0.5
1.0
1.5
Relative P-SP1/SP1
protein levels
⁎⁎
(h)
Con
NAC
PM2.5
PM2.5+NAC
0.0
0.1
0.2
0.3
Relative SREBP-1 protein levels
⁎⁎
(i)
Figure 5: PM
2.5
induced lipid accumulation in hepatocytes by increasing ROS levels and PTP1B expression. (a) BODIPY staining of L02
cells (scale bar, 50 μm). (b) Representative uorescence intensity images obtained from ow cytometry in L02 cells. (c) Analysis of
uorescence intensity obtained from ow cytometry. (d) Representative confocal images of ROS. (e) Western blotting of PTP1B, PP2A,
P-PP2A, SP1, P-SP1, and SREBP-1. (f) Protein quantication of PTP1B. (g) Protein quantication of P-PP2A/PP2A. (h) Protein
quantication of P-SP1/SP1. (i) Protein quantication of SREBP-1. All values are presented as the mean ± SD.P<0:05.
13Oxidative Medicine and Cellular Longevity
Melatonin concentration (𝜇mol/L)
0
12.5
25
50
100
200
0.6
0.8
1.0
1.2
1.4
Cell viability (% of control)
(a)
0.7
0.8
1.0
0.9
1.1
1.2
Cell viability (% of control)
Con
Mel
PM2.5
PM2.5+Mel
(b)
Con
Mel
PM2.5
PM2.5+Mel
0.0
0.5
1.0
1.5
2.0
2.5
T-CHO (mmol/g protein)
⁎⁎
(c)
Con
Mel
PM2.5
PM2.5+Mel
0.1
0.3
0.2
0.5
0.4
TAGs (mmol/g protein)
(d)
Con
MERGE FITC DAPI
Mel PM2.5 PM2.5+Mel
(e)
–102.9 0
0
20
40
60
80
100
Count (%)
103104105
FITC-A
106106.8
Con
Mel
PM2.5
PM2.5+Mel
(f)
Figure 6: Continued.
14 Oxidative Medicine and Cellular Longevity
ROS uorescence intensity
(fold of control)
Con
0
1
2
3
4
Mel PM2.5 PM2.5+Mel
⁎⁎
(g)
0
1
2
###
#
3
mRNA expression
4
PTP1B SP1 PP2A SREBP-1
Con
Mel
PM2.5
PM2.5+Mel
(h)
Con
GAPDH
SREBP-1
P-SP1
SP1
P-PP2A
PP2A
PTP1B
Mel PM2.5 PM2.5+Mel
(i)
Con
Mel
PM2.5
PM2.5+Mel
0.0
0.1
0.2
0.3
Relative PTP1B protein levels
⁎⁎
(j)
Con
Mel
PM2.5
PM2.5+Mel
0
2
1
4
3
5
Relative P-PP2A/PP2A
protein levels
⁎⁎
(k)
Con
Mel
PM2.5
PM2.5+Mel
0.0
0.2
0.1
0.3
0.4
0.5
Relative P-SP1/SP1 protein levels
⁎⁎
(l)
Con
Mel
PM2.5
PM2.5+Mel
0.00
0.10
0.05
0.15
0.20
Relative SREBP-1 protein levels
⁎⁎
(m)
Con
Mel
PM2.5
PM2.5+Mel
80
100
120
140
160
PP2A activity (mU/ml)
⁎⁎
(n)
Figure 6: Continued.
15Oxidative Medicine and Cellular Longevity
⁎⁎
Con
Mel
PM2.5
PM2.5+Mel
6
8
10
12
14
SP1 activity (ng/ml)
(o)
0
1
2
mRNA expression
3
Nrf2 Keap1
##
#
SOD
Con
Mel
PM2.5
PM2.5+Mel
(p)
Con
Nrf2
Keap1
SOD
GAPDH
Mel PM2.5 PM2.5+Mel
(q)
Con
Mel
PM2.5
PM2.5+Mel
0.0
0.4
0.2
0.6
0.8
Relative Nrf2 protein levels
(r)
Con
Mel
PM2.5
PM2.5+Mel
0.0
0.4
0.2
0.6
Relative Keap1 protein levels
(s)
Con
Mel
PM2.5
PM2.5+Mel
0.0
0.4
0.2
1.0
0.6
0.8
Relative SOD protein levels
(t)
Figure 6: Melatonin alleviated PM
2.5
-induced oxidative damage and upregulated PTP1B expression in vitro. (a) and (b) Cell ability. (c)
Total cholesterol lipid levels (mmol/g). (d) Triacylglycerol lipid levels (mmol/g). (e) Representative confocal images of ROS. (f)
Representative uorescence intensity images obtained from ow cytometry in L02 cells. (g) Analysis of uorescence intensity obtained
from ow cytometry. (h) The mRNA expression of PTP1B, PP2A, SP1, and SREBP-1. (i) Western blotting of PTP1B, PP2A, P-PP2A,
SP1, P-SP1, and SREBP-1. (j) Protein quantication of PTP1B. (k) Protein quantication of P-PP2A/PP2A. (l) Protein quantication of
P-SP1/SP1. (m) Protein quantication of SREBP-1. (n) The activity of PP2A. (o) The activity of SP1. (p) The mRNA expression of Nrf2,
Keap-1, and SOD. (q) Western blotting of Nrf2, Keap-1, and SOD. (r) Protein quantication of Nrf2. (s) Protein quantication of Keap-
1. (t) Protein quantication of SOD. All values are presented as the mean ± SD.P<0:05 for Con group vs PM
2.5
group and
#
P<0:05
for PM
2.5
group vs PM
2.5
+Mel group.
16 Oxidative Medicine and Cellular Longevity
0
0
PM2.5
Mel
PTP1B-inhibitor
101102103104105106107.2
20 40 60
Count (%)
80 100
−−
−−
−+
++
++++
++
+
+
+
−−
−−
FITC-A
(a)
0
1
2
ROS uorescence intensity
(fold of control)
3
4
PM2.5
Mel
PTP1B-inhibitor
−−
−−
+
++
++++
++
+
+
+
(b)
PM2.5
Mel
PTP1B-inhibitor
PTP1B
PP2A
P-PP2A
SREBP-1
GAPDH
SP1
P-SP 1
−−−−
+
++
++++
++
+
+
+
(c)
0.0
0.2
0.4
0.6
0.8
Relative PTP1B protein levels
1.0
PM2.5
Mel
PTP1B-inhibitor
−−−−
−−
−+
++
++++
++
+++
−−
−−
(d)
Figure 7: Continued.
17Oxidative Medicine and Cellular Longevity
equal variance were tested by one-way ANOVA or two-way
ANOVA. Among them, the data with only one variable of
PM
2.5
adopted one-way ANOVA, and the data with two var-
iables of melatonin and PM
2.5
adopted two-way ANOVA.
The Kruskal-Wallis test was used for nonparametric data.
A value of p<0:05 indicates statistical signicance. Each
experiment was repeated at least three times.
3. Results
3.1. Melatonin Alleviated the PM
2.5
-Induced Fatty Increase
and Steatosis in ApoE
-/-
Mice. To evaluate the eects of
PM
2.5
on liver lipid accumulation in mice, we rst conrmed
that PM
2.5
induced liver changes by ultrasound examination.
The contrast of liver-kidney echo is one of the obvious man-
ifestations of a fatty liver. Compared to the control group,
ultrasonography showed that the echo of the liver paren-
chyma was high and dense, and the contrast sign of the liver
and kidney was positive in the PM
2.5
group, but this expres-
sion was relieved in the melatonin group (Figure 1(a)). The
anterior-posterior diameter and left-right diameter of the
liver can reect changes in liver size. Although these two
indicators did not change signicantly between the PM
2.5
and control groups, there was a signicant dierence
between the PM
2.5
and melatonin groups (Figure 1(e)). Con-
cordant with this, analysis of the body weights, liver weights,
and liver coecient of the mice showed that the liver size of
the PM2.5 group was signicantly higher than that of the
control group, and that melatonin had a slight alleviating
eect on liver weight gain (Supplementary Figure S1E-1G).
Histological examinations of the liver are presented in
Figures 1(b)1(d). Electron microscopy images showed
large lipid droplets, and HE and Oil Red O staining
revealed notably enlarged adipocytes, fatty degeneration,
and specic lipid accumulation in PM
2.5
-treated mice
compared to the control group. However, treatment with
melatonin visibly alleviated these alterations. Next, changes
in lipid content in the liver were examined. The levels of
total cholesterol (TC) and triacylglycerols (TAGs) in the
livers increased in response to PM
2.5
, while melatonin
treatment signicantly decreased lipid levels (Figures 1
(g)and 1(h)). In addition, the Masson staining and qPCR
analysis results of inammatory factors (IL-1, IL-6, and
TNF-α) in liver tissue showed that PM
2.5
could cause liver
injury, and melatonin had a mitigating eect
(Supplementary Figure S1A-1D). Taken together, these
results suggested that PM
2.5
exposure could induce hepatic
lipid metabolism disorders and that melatonin treatment
had a benecial eect on the liver.
3.2. Protective Eects of Melatonin on PM
2.5
-Induced
Oxidative Damage in Liver. Multiple studies have shown
PM2.5
Mel
PTP1B-inhibitor
−−−−
−−
−+
++
++++
++
+++
−−
−−
0.0
0.5
1.0
1.5
2.0
Relative P-PP2A/PP2A
protein levels
(e)
PM2.5
Mel
PTP1B-inhibitor
−−−−
−−
−+
++
++++
++
+++
−−
−−
0.0
0.5
1.0
1.5
Relative P-SP1/SP1
protein levels
(f)
PM2.5
Mel
PTP1B-inhibitor
−−−−
−−
−+
++
++++
++
+++
−−
−−
0.0
Relative SREBP-1
protein levels
0.1
0.2
0.3
0.4
(g)
Figure 7: PTP1B inhibitor preconditioning eliminated lipid dysregulation in hepatocytes caused by PM
2.5
and melatonin intervention. (a)
Representative uorescence intensity images obtained from ow cytometry in L02 cells. (b) Analysis of uorescence intensity obtained from
ow cytometry. (c) Western blotting of PTP1B, PP2A, P-PP2A, SP1, P-SP1, and SREBP-1. (d) Protein quantication of PTP1B. (e) Protein
quantication of P-PP2A/PP2A. (f) Protein quantication of P-SP1/SP1. (g) Protein quantication of SREBP-1. All values are presented as
the mean ± SD.P<0:05.
18 Oxidative Medicine and Cellular Longevity
that PM
2.5
aggravates lipid metabolism disorder by inducing
oxidative stress. To determine the eects of PM
2.5
/melatonin
on ROS production, liver sections were stained with the uo-
rescent probe DHE to evaluate ROS levels. As shown in the
representative uorescence micrographs (red uorescence)
and histogram of ROS relative uorescence density
(Figures 2(a) and 2(b)), PM
2.5
treatment increased ROS gen-
eration, while melatonin supplementation alleviated ROS
generation. MDA and 4-HNE are important indexes of lipid
peroxidation. According to the quantitative analysis, expo-
sure to PM
2.5
resulted in signicantly increased levels of
MDA and 4-HNE, whereas melatonin treatment reversed
these eects (Figures 2(c) and 2(d)). Moreover, the degree
of oxidative stress was detected by examination of GSH-Px
and SOD. As anticipated, PM
2.5
treatment reduced the activ-
ity of GSH-Px and SOD in the liver compared to the control.
However, these two indicators were reversed by melatonin
(Figures 2(e) and 2(f)). Subsequent analysis of the protein
and mRNA expression levels of known indicators of oxida-
tive stress, including Keap1, Nrf2, and SOD, was performed.
In the present study, PM
2.5
exposure decreased Nrf2 and
SOD mRNA expression and increased Keap1 mRNA expres-
sion, and these negative eects were mitigated by melatonin
(Figure 2(g)). Consistently, compared with the control
group, the expression of Nrf2/Keap1 and the SOD protein
in the PM
2.5
group was not signicantly dierent, but there
were signicant changes after melatonin treatment
(Figures 2(h)2(k)). However, this result did not rule out
an impact on their gene expression. Overall, these data sug-
gested that the antioxidative stress eects of melatonin
might protect the liver from PM
2.5
exposure.
3.3. Melatonin Ameliorated Abnormal Liver Lipid
Metabolism and Caused Elevated PTP1B Expression
Induced by PM
2.5
.To identify the potential mechanisms by
which PM
2.5
or melatonin induced gene expression in lipid
accumulation, qPCR analysis of genes related to lipid metab-
olism was examined in liver samples. Analysis of the mRNA
expression levels showed that PM
2.5
exposure was associated
with lipid metabolism disorder, and PTP1B, which is closely
related to MAFLD, was an important upregulated transcrip-
tion factor (Supplementary Figure S2). PTP1B is a key
regulator of the antioxidant system and an activator of
liver adipogenesis. Next, the regulation of downstream
PTP1B on genes related to liver lipid metabolism was
veried by qPCR analysis. As expected, PM
2.5
exposure
signicantly increased the expression of lipid accumulation
markers (PP2A, SP1, and SREBP-1), whereas exogenous
melatonin treatment decreased their levels (Figure 3(a)).
Lipid accumulation plays an important role in the
progression of MAFLD. Additionally, for further
verication, Western blotting was carried out. PM
2.5
administration led to an increase in the accumulation of
PTP1B, P-PP2A/PP2A, P-SP1/SP1, and SREBP-1, and
subsequent analysis showed that melatonin inhibited their
expression (Figures 3(b)3(f)). However, the enzymatic
activities of PP2A and SP1 did not change signicantly in
PM
2.5
-exposed mice (Figures 3(g) and 3(h)). These results
PM2.5
ROS
Melatonin
PP2A PP2A
PTP1B
SP1 SP1
SREBP-1
Normal liver cells
Metabolic-associated fatty liver disease
pY
pY
Nrf2/Keap1
SOD
Lipogenesis / Steatosis
Lipid drops
Figure 8: Schematic of melatonin ameliorating PM
2.5
-induced hepatic lipid accumulation.
19Oxidative Medicine and Cellular Longevity
could be greatly downregulated by melatonin treatment. All
of the above data indicated that melatonin supplementation
reduced adiposity accumulation triggered by PM
2.5
.
3.4. PM
2.5
Exposure Caused Lipid Accumulation in L02 Cells
by Inducing ROS Production. The cytotoxic eects of PM
2.5
on L02 cell viability was assessed by CCK-8 assay. As evi-
denced in Figure 4(a), the viability of L02 cells decreased
with increasing doses of PM
2.5
. Treatment with between 25
and 100 μg/mL PM
2.5
for 24 h showed signicant dierences
compared with untreated cells. To detect the eects of PM
2.5
on lipid synthesis, L02 cells were exposed to various doses of
PM
2.5
(0100 μg/mL) for 24 h. As expected, the contents of
T-CHO and TAGs in L02 cells gradually increased as the
concentration of PM
2.5
increased (Figures 4(b) and 4(c)).
Next, ROS generated by PM
2.5
treatment were detected by
ow cytometry analysis quantication and confocal micros-
copy (Figures 4(d)4(f)). Compared to the control group,
the ROS uorescence intensity was observably increased in
PM
2.5
-treated L02 cells, which occurred in a manner depen-
dent on the PM
2.5
concentration. Then, activation of the
PTP1B pathway was determined after PM
2.5
exposure.
Quantitative measurements of protein expression showed
that P-PP2A/PP2A, P-SP1/SP1, and SREBP-1 expression
was dependent on the PM
2.5
concentration (Figures 4(g)
4(k)). In addition, activation of the Nrf2/Keap1 pathway
was detected by Western blot. Low-dose PM
2.5
induced the
upregulation of Nrf2 and SOD protein expression, while
high-dose PM
2.5
inhibited their expression. The opposite
trend was observed for Keap1 (Figures 4(l)4(o)). Given
the above data, although the eect indexes in the 25 μg/mL
dose group were signicantly dierent, the expression of
the proteins in the PTP1B pathway was signicantly dier-
ent in the 50 μg/mL dose group, and the Nrf2/Keap1 path-
way was inhibited in the 50 μg/mL dose group. For this
reason, the concentration of PM
2.5
(50 μg/mL) was selected
for subsequent experiments.
To obtain more details on the role of oxidative stress in
PM
2.5
-induced liver lipid accumulation, NAC (N-acetylcys-
teine) was added to L02 cells exposed to PM
2.5
(Figure 5).
Representative images of BODIPY staining are shown in
Figure 5(a). NAC treatment signicantly abolished the
PM
2.5
-induced increase in lipid content in L02 cells. Flow
cytometry and confocal microscopy data analysis indicated
that the uorescence intensity of the ROS generated in the
NAC-treated cultures exposed to PM
2.5
(50 μg/mL) was sig-
nicantly less than that in PM
2.5
-exposed cells
(Figures 5(b)5(d)). Additionally, NAC treatment restored
the expression of the proteins PTP1B, P-PP2A/PP2A, P-
SP1/SP1, and SREBP-1 in PM
2.5
-treated L02 cells to levels
comparable with the control (Figures 5(e)5(i)). PM
2.5
expo-
sure initially triggered oxidative stress, which further led to
lipid peroxidation. In short, the eects of PM
2.5
exposure
on lipid accumulation in L02 cells were ROS dependent
and involved PTP1B signalling.
3.5. Melatonin Alleviated PM
2.5
-Induced Oxidative Damage
and Lipid Accumulation In Vitro. To determine the dosage
of melatonin to be used, CCK-8 assays were applied to
examine cell viability. With increasing concentrations of
melatonin, the viability of L02 cells rst increased and then
decreased, and 200 μmol/L melatonin showed a signicant
decrease compared to the control group (Figure 6(a)). More-
over, the eects of PM
2.5
and melatonin on cell viability were
investigated (Figure 6(b)). Finally, 100 μmol/L melatonin
was selected for further experiments. First, the protective
eects of melatonin on lipid accumulation were detected
by measuring the levels of TC and TAGs (Figures 6(c) and
6(d)). The results indicated that melatonin decreased the
lipid levels in PM
2.5
-induced L02 cells compared with
PM
2.5
treatment alone. Flow cytometry and confocal micros-
copy data analysis indicated that treatment with melatonin
could restore ROS to control levels (Figures 6(e)6(g)). Fur-
thermore, melatonin altered the mRNA levels of PTP1B,
PP2A, SP1, and SREBP-1 in the presence of PM
2.5
(Figure 6(h)). Similarly, the levels of the proteins in the
PTP1B pathway showed that melatonin reduced PM
2.5
-
induced lipid accumulation (Figures 6(i)6(m)). Intrigu-
ingly, melatonin restored the PM
2.5
-mediated increase in
PP2A and SP1 activity to a normal level (Figures 6(n) and
6(o)). Moreover, PM
2.5
exposure decreased the Nrf2 and
SOD mRNA expression and increased the Keap1 mRNA
expression, and these negative eects were mitigated by mel-
atonin (Figure 6(p)). Melatonin activation of the Nrf2/Keap1
pathway represented relief of oxidative stress after PM
2.5
treatment. Compared to the control group, Nrf2/Keap1
and SOD protein expression was signicantly aected in
the melatonin group but only slightly changed after PM
2.5
exposure (Figures 6(q)6(t)). It could therefore be concluded
that the relief of oxidative stress after melatonin treatment
could alleviate fat accumulation in L02 liver cells.
3.6. Melatonin Regulated Hepatic Lipid Metabolism through
the PTP1B and Nrf2 Signalling Pathways in PM
2.5
-Treated
L02 Cells. To conrm that PM
2.5
-induced hepatocyte steato-
sis occurred through the upregulation of PTP1B, a specic
inhibitor of PTP1B (10 μg/mL) was used in this study.
Firstly, the dose was determined by PCR to detect the inhib-
itory eect on PTP1B mRNA expression (Supplementary
Figure S3). As shown in Figures 7(a) and 7(b), both
melatonin and the PTP1B inhibitor had signicant ROS
scavenging ability. Compared with PM
2.5
treatment alone,
melatonin treatment decreased the ROS levels by
approximately 40%, and the PTP1B inhibitor decreased the
ROS levels by approximately 80%. The levels of melatonin
and PTP1B inhibitor in PM
2.5
-exposed cells were also
signicantly lower than those in the group treated with
both melatonin and PM
2.5
. Similar to melatonin, the
inhibitor downregulated the expression of PTP1B and its
downstream molecular proteins (PTP1B, P-PP2A/PP2A, P-
SP1/SP1, and SREBP-1) that regulate lipid production. The
synergistic eects of melatonin and the inhibitor were
greater than the eects of melatonin alone (Figures 7(c)
7(g)). Consistent with the results above, PTP1B was the
direct target gene of PM
2.5
-induced oxidative stress, and
inhibition of PTP1B expression downregulated the
expression of its downstream gene SREBP-1 and reduced
ROS production, thus reducing lipid production in L02 cells.
20 Oxidative Medicine and Cellular Longevity
4. Discussion
Environmental PM
2.5
has been recognized as the largest
global threat aecting human health, including the develop-
ment of MAFLD [4]. The pathogenesis and molecular mech-
anisms of PM
2.5
-induced MAFLD have not yet been well
elucidated. In this study, we found that PM
2.5
induced oxi-
dative stress, which activated PTP1B and in turn regulated
ROS release with positive feedback. Moreover, melatonin
alleviated the interference of liver fat metabolism disorder
caused by PM
2.5
through regulation of the ROS-mediated
PTP1B and Nrf2 signalling pathways.
MAFLD is a redenition of NAFLD (nonalcoholic fatty
liver disease). MAFLD represents a general overview of com-
mon liver metabolic disorders (not just nonalcoholics) and
has multiple dominant drivers of subphenotypic response
diseases. It covers more than NAFLD and has more specic
diagnostic criteria [41]. Therefore, the more accurate and
clear terminology of MAFLD was adopted in this work,
which may provide a much wider applicability for our subse-
quent study to explore the toxic mechanism of PM
2.5
. In our
study, ultrasonography showed that in the PM
2.5
exposure
group, the liver had a smooth contour, sharp edges, and
increased parenchymal echo density, and positive contrast
signs were also observed in the liver and kidney (Figure 1).
The hepatorenal index is an important indicator of a fatty
liver [42]. Similarly, the histopathological observation results
(Oil Red O and HE staining) showed obvious fat vacuoles of
varying sizes in the livers of PM
2.5
-treated mice, which pre-
sented with severe steatosis (Figure 1). Exposure to PM
2.5
has been reported to cause systemic IR and increase the
accumulation of hepatic lipids in the liver, which is consis-
tent with our ndings [43]. In addition, we observed
increases in T-CHO and TAGs in mouse livers and human
hepatocytes exposed to PM
2.5
, indicating that the induced
lipid metabolism disorders were severe, as evidenced by the
lipid index (Figures 1 and 4). The liver is a central organ
for lipid homeostasis and energy metabolism [44]. Liver ste-
atosis is caused by an imbalance in lipid homeostasis, where
lipid absorption or de novo fat production exceeds lipid oxi-
dation or output [45]. Here, our results revealed that PM
2.5
exposure induced signicant lipid accumulation in the liver
accompanied by an increase in liver volume, suggesting that
PM
2.5
exposure triggered pathological changes in liver mor-
phology and function.
To date, many studies have demonstrated that MAFLD
is closely related to oxidative stress induced by PM
2.5
expo-
sure [5]. Oxidative stress is caused by the imbalance between
the production of ROS and the ROS scavenging activity [46].
Excessive ROS results in increased adipogenesis and
decreased β-oxidation of fatty acids, which leads to the accu-
mulation of triacylglycerols in hepatocytes [47]. We studied
the induction of ROS generation by PM
2.5
in ApoE
-/-
mice,
and the data revealed that PM
2.5
promoted ROS production,
decreased SOD activity, and induced lipid peroxidation (as
evidenced by the levels of 4-HNE and MDA) (Figure 2).
Consistent with the results of the animal experiments,
PM
2.5
increased intracellular ROS in a dose-dependent man-
ner in L02 cells (Figure 4). Compared with reliable evidence
that PM
2.5
exposure can upregulate ROS pathways, the reg-
ulation and defence of antioxidant mechanisms are relatively
scarce. We further examined the mRNA and protein levels
of antioxidant stress markers, namely, Nrf2/Keap1 and
SOD. The qPCR results showed that PM
2.5
acted by upregu-
lating the Nrf2 inhibitor Keap1. Compared with the control
group, Nrf2 protein expression was slightly downregulated
in the PM
2.5
group, but there was no signicant dierence
(Figure 2). The reason for this result may be that at the tran-
scriptional and translational levels, the course of mRNA
translation into proteins is adjusted by a variety of factors,
which may lead to an inconsistency between mRNA and
protein expression [48]. Interestingly, the expression of
Nrf2 was upregulated in the low-dose PM
2.5
group and
downregulated in the high-dose PM
2.5
group, while the
expression of Keap1 showed the opposite trend (Figure 4).
It may be that the low dose of PM
2.5
induces the bodys
stress response and activates Nrf2. However, with increasing
exposure dose, PM
2.5
inhibits the Nrf2 pathway. Similarly, it
has been found that low concentrations of PM
2.5
slightly
upregulate Nrf2 expression, and subsequently, PM
2.5
treat-
ment dose-dependently decreases Nrf2 expression [10].
These studies have shown that PM
2.5
induces ROS produc-
tion and changes in antioxidant genes, which play vital reg-
ulatory roles in the progression of MAFLD.
ROS overproduction can modulate many cellular events
involved in hepatic lipid metabolism diseases by regulating a
variety of disease-related targets, such as PTP1B [49]. In
addition, PTP1B levels were signicantly elevated in the
hepatocytes of fructose-fed hamsters, HFD-fed mice, and
fatty liver and insulin-resistant animal models [50]. The
overexpression of PTP1B in liver cells increased the expres-
sion level and transcriptional activity of SREBP1, which
resulted in the increased synthesis of liver triacylglycerols
and fatty acids [51]. This was due to the enhanced transcrip-
tional activity of the recombinant SP1 site in the SREBP1
promoter by increasing the activity of PP2A when PTP1B
was overexpressed [52]. This study found that PM
2.5
could
upregulate PTP1B expression by inducing ROS generation,
and the expression level of PTP1B and ROS production
was dose-dependent (Figures 3, 4, and 6). Especially, PM
2.5
and melatonin did not aect the activity of PP2A and SP1
(Figure 3). This may be related to the posttranslational mod-
ication of proteins [53, 54]. NAC preconditioning inhibited
PM
2.5
-induced PTP1B overexpression, suggesting that ROS
plays an important role in PTP1B activation after PM
2.5
exposure (Figure 5). Surprisingly, we found that PTP1B reg-
ulated by PM
2.5
had positive feedback regulation on ROS
release. As shown in Figure 7, pretreatment of L02 cells with
PTP1B inhibitors signicantly reduced ROS production and
subsequently downregulated the PTP1B downstream pro-
teins PP2A, SP1, and SREBP1. Based on previous evidence,
PTP1B KO mice showed decreased ROS production and
lipid peroxidation in the liver [55]. To our knowledge, our
results provide new evidence of the mechanism by which
PM
2.5
exposure promotes the occurrence and development
of MAFLD, demonstrating that PM
2.5
exposure activates
the ROS/PTP1B pathway and that PTP1B regulates ROS
by positive feedback.
21Oxidative Medicine and Cellular Longevity
Many studies have shown that the nocturnal indole
melatonin produced by the pineal gland is eective against
metabolic syndrome [56]. More recently, melatonin has
been shown to reverse the harmful eects of fructose in
the diet, and this animal model modulates metabolic path-
ways such as lipid production, β-oxidation, lipolysis, and
gluconeogenesis [57]. Melatonin may also be ingested in
the liver in a dose-dependent manner through specic cel-
lular and nuclear receptors [58]. The pathogenesis of
MAFLD is complex, but melatonin may be the key to
the treatment of MAFLD. Furthermore, melatonin allevi-
ated hepatic steatosis and lipid accumulation in ApoE
-/-
mice under dierent experimental conditions [59]. A pre-
vious animal study showed that ROS mediated
lipopolysaccharide-induced SREBP-1c activation and lipid
accumulation in the liver. Melatonin might be used as a
pharmacological agent to prevent endotoxin-induced
MAFLD [60]. Liver lipotoxicity is closely related to hepatic
metabolic disorders caused by impaired fatty acid oxida-
tion and increased ROS production [61]. It may be helpful
to study the protective eects of melatonin on PM
2.5
-
induced hepatic fatty degeneration. Therefore, more
research is encouraged to explore this issue. In our study,
as expected, melatonin mitigated steatosis and decreased
the lipid content of the liver during PM
2.5
damage. Both
animal and cell experiments showed that melatonin eec-
tively reduced ROS levels and helped to downregulate
PTP1B and increase Nrf2 expression in PM
2.5
-treated
groups, thereby changing the eects of PM
2.5
exposure
on liver lipid accumulation.
The schematic diagram summarizing these results and
mechanisms shows that PM
2.5
-induced ROS accumulation
simultaneously promotes fat generation signal transduction
by activating the PTP1B-PP2A-SP1-SREBP1 axis and inhi-
biting the Nrf2/Keap1-SOD axis, resulting in lipid accumu-
lation and promotion of the occurrence and development
of MAFLD. Moreover, melatonin plays an antioxidative
stress role and regulates the ROS-mediated PTP1B and
Nrf2 signalling pathways by inhibiting ROS production to
alleviate the harmful eects induced by PM
2.5
(Figure 8). A
comprehensive study of ROS targets could not only provide
insight into the mechanism of PM
2.5
-induced MAFLD but
also give more evidence for the clinical applications of
melatonin.
5. Conclusions
In summary, this study shows that PM
2.5
promoted the
occurrence and development of MAFLD in ApoE
-/-
mice.
Excess accumulation of PM
2.5
-induced ROS could activate
PTP1B, which in turn had a positive feedback regulation
eect on ROS release. Our study is the rst to show that mel-
atonin alleviated the disturbance of PM
2.5
-triggered hepatic
steatosis and liver damage by regulating the ROS-mediated
PTP1B and Nrf2 signalling pathways. These results suggest
that melatonin administration may be a prospective therapy
for the prevention and treatment of MAFLD associated with
air pollution.
Data Availability
Most of data and materials generated or analyzed during this
study are included in this manuscript. Other data are avail-
able from the corresponding authors on reasonable request.
Ethical Approval
The animal experiment in this study was approved by the
Animal Ethics Committee of Capital Medical University
(Ethics No. AEEI-2016-076).
Consent
Not applicable.
Conflicts of Interest
The authors declare they have no conict of interest.
AuthorsContributions
Zhiwei Sun and Junchao Duan conceived and designed the
experiments. Zhou Du, Shuang Liang, Jingyi Zhang, and
Qing Xu performed the experiments. Zhou Du, Yang Li,
Yang Yu, and Qing Xu analyzed the data. Zhiwei Sun and
Junchao Duan contributed reagents/materials/analysis tools.
Zhou Du and Junchao Duan wrote the paper.
Acknowledgments
This work was supported by the National Natural Science
Foundation of China (91943301, 92043301, 81930091, and
81973077), Beijing Natural Science Foundation Program
and Scientic Research Key Program of Beijing Municipal
Commission of Education (KZ202110025040).
Supplementary Materials
Figure S1: liver damage eects of PM
2.5
. Figure S2: mRNA
expression levels associated with PM
2.5
exposure to liver
injury. Figure S3: mRNA expression of PTP1B after treat-
ment with PTP1B inhibitor. Table S1: concentrations of
inorganic elements in PM
2.5
. Table S2: content of water sol-
uble ions in PM
2.5
. Table S3: introduction of genes related to
liver injury caused by PM
2.5
.(Supplementary Materials)
References
[1] H. Wang, X. Shen, J. Liu et al., The eect of exposure time and
concentration of airborne PM
2.5
on lung injury in mice: a tran-
scriptome analysis,Redox biology, vol. 26, article 101264,
2019.
[2] M.-X. Xu, C.-X. Ge, Y.-T. Qin et al., Prolonged PM
2.5
expo-
sure elevates risk of oxidative stress-driven nonalcoholic fatty
liver disease by triggering increase of dyslipidemia,Free Rad-
ical Biology and Medicine, vol. 130, pp. 542556, 2019.
[3] T. Li, R. Hu, Z. Chen et al., Fine particulate matter (PM
2.5
):
the culprit for chronic lung diseases in China,Chronic dis-
eases and translational medicine, vol. 4, no. 3, pp. 176186,
2018.
22 Oxidative Medicine and Cellular Longevity
[4] D. Li, Y. Li, G. Li, Y. Zhang, J. Li, and H. Chen, Fluorescent
reconstitution on deposition of PM
2.5
in lung and extrapul-
monary organs,Proceedings of the National Academy of Sci-
ences of the United States of America, vol. 116, no. 7,
pp. 24882493, 2019.
[5] T. Jian, X. Ding, Y. Wu et al., Hepatoprotective eect of
loquat leaf avonoids in PM
2.5
-induced non-alcoholic fatty
liver disease via regulation of IRs-1/Akt and CYP2E1/JNK
pathways,International journal of molecular sciences,
vol. 19, no. 10, p. 3005, 2018.
[6] S. Sun, Q. Yang, Q. Zhou et al., Long-term exposure to ne
particulate matter and non-alcoholic fatty liver disease: a pro-
spective cohort study,Gut, vol. 71, no. 2, pp. 443445, 2022.
[7] S. Spahis, E. Delvin, J. M. Borys, and E. Levy, Oxidative stress
as a critical factor in nonalcoholic fatty liver disease pathogen-
esis,Antioxidants & redox signaling, vol. 26, no. 10, pp. 519
541, 2017.
[8] S. Ding, C. Yuan, B. Si et al., Combined eects of ambient par-
ticulate matter exposure and a high-fat diet on oxidative stress
and steatohepatitis in mice,PLoS One, vol. 14, no. 3, article
e0214680, 2019.
[9] F. Bessone, M. V. Razori, and M. G. Roma, Molecular path-
ways of nonalcoholic fatty liver disease development and pro-
gression,Cellular and molecular life sciences : CMLS, vol. 76,
no. 1, pp. 99128, 2019.
[10] C. Ge, J. Tan, S. Zhong et al., Nrf2 mitigates prolonged PM
2.5
exposure-triggered liver inammation by positively regulating
SIKE activity: protection by Juglanin,Redox biology, vol. 36,
article 101645, 2020.
[11] J. P. Ribeiro, A. C. Kalb, P. P. Campos et al., Toxicological
eects of particulate matter (PM
2.5
) on rats: bioaccumulation,
antioxidant alterations, lipid damage, and ABC transporter
activity,Chemosphere, vol. 163, pp. 569577, 2016.
[12] M. J. Piao, M. J. Ahn, K. A. Kang et al., Particulate matter 2.5
damages skin cells by inducing oxidative stress, subcellular
organelle dysfunction, and apoptosis,Archives of Toxicology,
vol. 92, no. 6, pp. 20772091, 2018.
[13] L. Chen, X. Zhang, L. Zhang, and D. Zheng, Eect of saxaglip-
tin, a dipeptidyl peptidase 4 inhibitor, on non-alcoholic fatty
liver disease,Diabetes, metabolic syndrome and obesity : tar-
gets and therapy, vol. 13, pp. 35073518, 2020.
[14] A. Vivero, M. Ruz, M. Rivera et al., Zinc supplementation and
strength exercise in rats with type 2 diabetes: Akt and PTP1B
phosphorylation in nonalcoholic fatty liver,Biological trace
element research., vol. 199, no. 6, pp. 22152224, 2021.
[15] Á. González-Rodríguez, M. P. Valdecantos, P. Rada et al.,
Dual role of protein tyrosine phosphatase 1B in the progres-
sion and reversion of non-alcoholic steatohepatitis,Molecular
metabolism, vol. 7, pp. 132146, 2018.
[16] N. Aberdein, R. J. Dambrino, J. M. do Carmo et al., Role
of PTP1B in POMC neurons during chronic high-fat diet:
sex dierences in regulation of liver lipids and glucose toler-
ance,American Journal of Physiology-Regulatory, Integra-
tive and Comparative Physiology, vol. 314, no. 3,
pp. R478R488, 2018.
[17] J. Zhou, K. Huang, and X. G. Lei, Selenium and diabetesEvi-
dence from animal studies,Free radical biology & medicine,
vol. 65, pp. 15481556, 2013.
[18] S. F. Bhat, S. E. Pinney, K. M. Kennedy et al., Exposure to high
fructose corn syrup during adolescence in the mouse alters
hepatic metabolism and the microbiome in a sex-specic man-
ner,The Journal of physiology, vol. 599, no. 5, pp. 14871511,
2021.
[19] B. Bilska, F. Schedel, A. Piotrowska et al., Mitochondrial func-
tion is controlled by melatonin and its metabolites in vitro in
human melanoma cells,Journal of Pineal Research, vol. 70,
no. 3, p. e12728, 2021.
[20] K. H. Jung, S. W. Hong, H. M. Zheng et al., Melatonin ame-
liorates cerulein-induced pancreatitis by the modulation of
nuclear erythroid 2-related factor 2 and nuclear factor-
kappaB in rats,Journal of Pineal Research, vol. 48, no. 3,
pp. 239250, 2010.
[21] J. I. Heo, D. W. Yoon, J. H. Yu et al., Melatonin improves
insulin resistance and hepatic steatosis through attenuation
of alpha-2-HS-glycoprotein,Journal of Pineal Research,
vol. 65, no. 2, article e12493, 2018.
[22] A. Stacchiotti, I. Grossi, R. García-Gómez et al., Melatonin
eects on non-alcoholic fatty liver disease are related to micro-
RNA-34a-5p/Sirt1 axis and autophagy,Cell, vol. 8, no. 9,
p. 1053, 2019.
[23] S. Bose, G. B. Diette, H. Woo et al., Vitamin D status modies
the response to indoor particulate matter in obese urban chil-
dren with asthma,The journal of allergy and clinical immu-
nology In practice, vol. 7, no. 6, pp. 18151822.e2, 2019.
[24] K. D. Lu, P. N. Breysse, G. B. Diette et al., Being overweight
increases susceptibility to indoor pollutants among urban chil-
dren with asthma,The Journal of allergy and clinical immu-
nology, vol. 131, no. 4, pp. 10171023, 2013.
[25] O. Jamialahmadi, R. M. Mancina, E. Ciociola et al., Exome-
wide association study on alanine aminotransferase identies
sequence variants in the GPAM and APOE_associated with
fatty liver disease,Gastroenterology, vol. 160, no. 5,
pp. 16341646.e7, 2021.
[26] N. D. Palmer, B. Kahali, A. Kuppa et al., Allele specic varia-
tion at APOE increases non-alcoholic fatty liver disease and
obesity but decreases risk of Alzheimer's disease and myocar-
dial infarction,Human molecular genetics, vol. 30, no. 15,
pp. 14431456, 2021.
[27] W. C. Huang, J. W. Xu, S. Li, X. E. Ng, and Y. T. Tung, Eects
of exercise on high-fat diet-induced non-alcoholic fatty liver
disease and lipid metabolism in ApoE knockout mice,Nutri-
tion & metabolism, vol. 19, no. 1, p. 10, 2022.
[28] J. H. Dumolt, M. S. Patel, and T. C. Rideout, Gestational
hypercholesterolemia programs hepatic steatosis in a sex-
specic manner in ApoE-decient mice,The Journal of nutri-
tional biochemistry, vol. 101, article 108945, 2022.
[29] S. Liu, R. Weng, X. Gu, L. Li, and Z. Zhong, Association
between apolipoprotein E gene polymorphism and nonalco-
holic fatty liver disease in Southern China: a case-control
study,Journal of clinical laboratory analysis, vol. 35, no. 12,
article e24061, 2021.
[30] J. Xu, Y. Guo, X. Huang et al., Eects of DHA dietary inter-
vention on hepatic lipid metabolism in apolipoprotein E-
decient and C57BL/6J wild-type mice,Biomedicine & phar-
macotherapy = Biomedecine & pharmacotherapie, vol. 144,
article 112329, 2021.
[31] Y. Q. Hua, Y. Zeng, J. Xu, and X. L. Xu, Naringenin alleviates
nonalcoholic steatohepatitis in middle-aged Apoe
/
mice: role
of SIRT1,Phytomedicine : international journal of phytother-
apy and phytopharmacology, vol. 81, article 153412, 2021.
[32] A. Stachowicz, A. Wiśniewska, K. Kuśet al., Diminazene
aceturate stabilizes atherosclerotic plaque and attenuates
23Oxidative Medicine and Cellular Longevity
hepatic steatosis in apoE-knockout mice by inuencing mac-
rophages polarization and taurine biosynthesis,International
journal of molecular sciences, vol. 22, no. 11, p. 5861, 2021.
[33] Y. Zhang, H. Hu, Y. Shi et al.,
1
H NMR-based metabolomics
study on repeat dose toxicity of ne particulate matter in rats
after intratracheal instillation,The Science of the total envi-
ronment, vol. 589, pp. 212221, 2017.
[34] J. Liu, S. Liang, Z. Du et al., PM
2.5
aggravates the lipid accu-
mulation, mitochondrial damage and apoptosis in macro-
phage foam cells,Environmental Pollution, vol. 249,
pp. 482490, 2019.
[35] M. Zhang, J. Lin, S. Wang et al., Melatonin protects against
diabetic cardiomyopathy through Mst1/Sirt3 signaling,Jour-
nal of Pineal Research, vol. 63, no. 2, p. e12418, 2017.
[36] J. Hu, L. Zhang, Y. Yang et al., Melatonin alleviates postin-
farction cardiac remodeling and dysfunction by inhibiting
Mst1,Journal of Pineal Research, vol. 62, no. 1, p. e12368,
2017.
[37] WHO, Air Quality Guidelines: Global Update 2005,in Par-
ticulate Matter, Ozone, Nitrogen Dioxide and Sulfur Dioxide,
World Health Organization, Geneva, Switzerland, 2005.
[38] R. Ning, Y. Li, Z. Du et al., The mitochondria-targeted antiox-
idant MitoQ attenuated PM
2.5
-induced vascular brosis via
regulating mitophagy,Redox biology, vol. 46, article 102113,
2021.
[39] Z. Zhang, S. Hu, P. Fan et al., The roles of liver inammation
and the insulin signaling pathway in PM
2.5
instillation-
induced insulin resistance in Wistar rats,Disease markers,
vol. 2021, Article ID 2821673, 11 pages, 2021.
[40] M. Catta-Preta, L. S. Mendonca, J. Fraulob-Aquino, M. B.
Aguila, and C. A. Mandarim-de-Lacerda, A critical analysis
of three quantitative methods of assessment of hepatic steato-
sis in liver biopsies,Virchows Archiv : an international journal
of pathology, vol. 459, no. 5, pp. 477485, 2011.
[41] M. Eslam, A. J. Sanyal, J. George et al., MAFLD: a consensus-
driven proposed nomenclature for metabolic associated fatty
liver disease,Gastroenterology, vol. 158, no. 7, pp. 1999
2014.e1, 2020.
[42] P. Avramovski, M. Avramovska, Z. Nikleski, B. Ilkovska,
K. Sotiroski, and E. Sikole, The predictive value of the hepa-
torenal index for detection of impaired glucose metabolism
in patients with non-alcoholic fatty liver disease,Indian jour-
nal of gastroenterology : ocial journal of the Indian Society of
Gastroenterology, vol. 39, no. 1, pp. 5059, 2020.
[43] C. Liu, X. Xu, Y. Bai et al., Air pollution-mediated susceptibil-
ity to inammation and insulin resistance: inuence of CCR2
pathways in mice,Environmental Health Perspectives,
vol. 122, no. 1, pp. 1726, 2014.
[44] Z. Wu, H. Ma, L. Wang et al., Tumor suppressor ZHX2
inhibits NAFLD-HCC progression via blocking LPL- medi-
ated lipid uptake,Cell death and dierentiation, vol. 27,
no. 5, pp. 16931708, 2020.
[45] S. L. Friedman, B. A. Neuschwander-Tetri, M. Rinella, and
A. J. Sanyal, Mechanisms of NAFLD development and thera-
peutic strategies,Nature Medicine, vol. 24, no. 7, pp. 908922,
2018.
[46] X. F. Hu, G. Xiang, T. J. Wang et al., Impairment of type H
vessels by NOX2-mediated endothelial oxidative stress: critical
mechanisms and therapeutic targets for bone fragility in
streptozotocin-induced type 1 diabetic mice,Theranostics,
vol. 11, no. 8, pp. 37963812, 2021.
[47] A. Mansouri, C. H. Gattolliat, and T. Asselah, Mitochondrial
dysfunction and signaling in chronic liver diseases,Gastroen-
terology, vol. 155, no. 3, pp. 629647, 2018.
[48] S. Lin, C. Cao, Y. Meng et al., Comprehensive analysis of the
value of RAB family genes in prognosis of breast invasive car-
cinoma,Bioscience Reports, vol. 40, no. 5, 2020.
[49] J. Dong, S. Viswanathan, E. Adami et al., Hepatocyte-specic
IL11 cis-signaling drives lipotoxicity and underlies the transi-
tion from NAFLD to NASH,Nature Communications,
vol. 12, no. 1, p. 66, 2021.
[50] C. Proença, D. Ribeiro, M. Freitas, F. Carvalho, and
E. Fernandes, A comprehensive review on the antidiabetic
activity of avonoids targeting PTP1B and DPP-4: a
structure-activity relationship analysis,Critical reviews in
food science and nutrition., vol. 62, no. 15, pp. 40954151,
2022.
[51] X. Zhou, L. L. Wang, W. J. Tang, and B. Tang, Astragaloside
IV inhibits protein tyrosine phosphatase 1B and improves
insulin resistance in insulin-resistant HepG2 cells and triglyc-
eride accumulation in oleic acid (OA)-treated HepG2 cells,
Journal of Ethnopharmacology, vol. 268, article 113556, 2021.
[52] H. Wu, T. Zhang, F. Pan et al., MicroRNA-206 prevents hepa-
tosteatosis and hyperglycemia by facilitating insulin signaling
and impairing lipogenesis,Journal of Hepatology, vol. 66,
no. 4, pp. 816824, 2017.
[53] N. Zheng and N. Shabek, Ubiquitin ligases: structure, func-
tion, and regulation,Annual Review of Biochemistry, vol. 86,
no. 1, pp. 129157, 2017.
[54] T. Narita, B. T. Weinert, and C. Choudhary, Functions and
mechanisms of non-histone protein acetylation,Nature
reviews Molecular cell biology, vol. 20, no. 3, pp. 156174, 2019.
[55] M. F. Hsu, S. Koike, A. Mello, L. E. Nagy, and F. G. Haj,
Hepatic protein-tyrosine phosphatase 1B disruption and
pharmacological inhibition attenuate ethanol-induced oxida-
tive stress and ameliorate alcoholic liver disease in mice,
Redox biology, vol. 36, article 101658, 2020.
[56] J. Yin, Y. Li, H. Han et al., Melatonin reprogramming of gut
microbiota improves lipid dysmetabolism in high-fat diet-fed
mice,Journal of Pineal Research, vol. 65, no. 4, article
e12524, 2018.
[57] F. J. Valenzuela-Melgarejo, C. Caro-Díaz, and G. Cabello-Guz-
mán, Potential crosstalk between fructose and melatonin: a
new role of melatonininhibiting the metabolic eects of
fructose,International journal of endocrinology, vol. 2018,
Article ID 7515767, 11 pages, 2018.
[58] C. Venegas, J. A. García, C. Doerrier et al., Analysis of the
daily changes of melatonin receptors in the rat liver,Journal
of Pineal Research, vol. 54, no. 3, pp. 313321, 2013.
[59] F. Bonomini, G. Favero, L. F. Rodella, M. H. Moghadasian, and
R. Rezzani, Melatonin modulation of Sirtuin-1 attenuates
liver injury in a hypercholesterolemic mouse model,BioMed
Research International, vol. 2018, Article ID 7968452, 9 pages,
2018.
[60] X. Chen, C. Zhang, M. Zhao et al., Melatonin alleviates
lipopolysaccharide-induced hepatic SREBP-1c activation and
lipid accumulation in mice,Journal of Pineal Research,
vol. 51, no. 4, pp. 416425, 2011.
[61] B. Sun, J. Zhou, Y. Gao et al., Fas-associated factor 1 promotes
hepatic insulin resistance via JNK signaling pathway,Oxida-
tive medicine and cellular longevity, vol. 2021, Article ID
3756925, 10 pages, 2021.
24 Oxidative Medicine and Cellular Longevity
... An epidemiological study reveals that chronic exposure to PM 2.5 increases the incidence of MAFLD in China [8]. Another prospective cohort study indicates that the incidence of MAFLD is 34% higher in people living in areas with high PM 2.5 concentrations than in those with low PM 2.5 concentrations [9]. Consistent with epidemiological evidence, toxicological studies show that exposure to air pollutants can cause liver metabolic disorders and even liver cancer [10,11]. ...
... Consistent with epidemiological evidence, toxicological studies show that exposure to air pollutants can cause liver metabolic disorders and even liver cancer [10,11]. Du et al. found that PM 2.5 can induce hepatic steatosis in ApoE −/− mice [9]. Zheng et al. reported that mice exposed to ambient PM display non-alcoholic steatohepatitis (NASH)-like phenotypes and impaired liver glucose metabolism [12]. ...
Article
Full-text available
Ambient particulate matter (PM2.5) is a potential risk factor for metabolic damage to the liver. Epidemiological studies suggest that elevated PM2.5 concentrations cause changes in hepatic metabolism, but there is a lack of laboratory evidence. Here, we aimed to evaluate the effects of PM2.5 exposure on liver metabolism in C57BL/6j female mice (10 months old) and to explore the mechanisms underlying metabolic alterations and differential gene expressions by combining metabolomics and transcriptomics analyses. The metabolomics results showed that PM2.5 exposure notably affected the metabolism of amino acids and organic acids and caused hepatic lipid and bile acid accumulation. The transcriptomic analyses revealed that PM2.5 exposure led to a series of metabolic pathway abnormalities, including steroid biosynthesis, steroid hormone biosynthesis, primary bile acid biosynthesis, etc. Among them, the changes in the bile acid pathway might be one of the causes of liver damage in mice. In conclusion, this study clarified the changes in liver metabolism in mice caused by PM2.5 exposure through combined transcriptomic and metabolomic analyses, revealed that abnormal bile acid metabolism is the key regulatory mechanism leading to metabolic-associated fatty liver disease (MAFLD) in mice, and provided laboratory evidence for further clarifying the effects of PM2.5 on body metabolism.
... Blood samples were collected by cardiac puncture when the rats were sacrificed to alleviate their suffering [37]. The plasma levels of total cholesterol, aspartate transaminase (AST), and alanine aminotransferase (ALT) were determined by a standard auto-analyzer (Hitachi model 7450, Tokyo, Japan). ...
Article
Full-text available
Prenatal exposure to a high-fat diet (HFD) or microplastics can impact liver fat accumulation in offspring. This study investigates the protective effects of prenatal melatonin on liver injury in male pups resulting from maternal exposure to a HFD and microplastics. Pregnant Sprague-Dawley rats were fed either an HFD or a normal chow diet, with some groups exposed to microplastics alone or in combination with melatonin. Male pups were evaluated on postnatal day 7. Results indicated that pups in the HFD-microplastics group (HFD-Mi) exhibited increased liver lipid accumulation (observed in histological staining), apoptosis (elevated cleaved caspase 3, phospho-AKT, and TUNEL staining), inflammation (higher IL- 6 and TNF-α), and oxidative stress (elevated malondialdehyde). Conversely, melatonin treatment (HFD-Mi + M) significantly reduced these effects, including lipid accumulation, apoptosis, and inflammation, while enhancing antioxidant enzyme glutathione peroxidase activity and improving lipid metabolism (reduced SREBP- 1 expression). These findings suggest that prenatal melatonin mitigates liver injury caused by maternal HFD and microplastics through its anti-inflammatory, antioxidative, and lipid-regulating properties, underscoring its potential hepatoprotective role.
... Third, investigating pharmaceutical interventions for treating air pollution-induced MAFLD may benefit the working population subjected to prolonged ambient air pollution exposure. For instance, a study using ApoE-/-mice demonstrated that melatonin could inhibit the expression of protein tyrosine phosphatase 1B (PTP1B), triggered by PM 2.5 exposure, which is associated with weight gain and increased liver lipids (Du et al., 2022a). Another study suggested that total flavonoids could significantly reduce body weight, the liver-to-body weight ratio, and the surge in blood lipids induced by PM 2.5 (Jian et al., 2018). ...
Article
Full-text available
Background: While evidence suggests that PM2.5 is associated with overall prevalence of Metabolic (dysfunction)-Associated Fatty Liver Disease (MAFLD), effects of comprehensive air pollutant mixture on MAFLD and its subtypes remain unclear. Objective: To investigate individual and joint effects of long-term exposure to comprehensive air pollutant mixture on MAFLD and its subtypes. Methods: Data of 27,699 participants of the Chinese Cohort of Working Adults were analyzed. MAFLD and subtypes, including overweight/obesity, lean, and diabetes MAFLD, were diagnosed according to clinical guidelines. Concentrations of NO3-, SO42-, NH4+, organic matter (OM), black carbon (BC), PM2.5, SO2, NO2, O3 and CO were estimated as a weighted average over participants' residential and work addresses for the three years preceding outcome assessment. Logistic regression and weighted quantile sum regression were used to estimate individual and joint effects of air pollutant mixture on presence of MAFLD. Results: Overall prevalence of MAFLD was 26.6 % with overweight/obesity, lean, and diabetes MAFLD accounting for 92.0 %, 6.4 %, and 1.6 %, respectively. Exposure to SO42-, NO3-, NH4+, BC, PM2.5, NO2, O3and CO was significantly associated with overall MAFLD, overweight/obesity MAFLD, or lean MAFLD in single pollutant models. Joint effects of air pollutant mixture were observed for overall MAFLD (OR = 1.10 [95 % CI: 1.03, 1.17]), overweight/obesity (1.09 [1.02, 1.15]), and lean MAFLD (1.63 [1.28, 2.07]). Contributions of individual air pollutants to joint effects were dominated by CO in overall and overweight/obesity MAFLD (Weights were 42.31 % and 45.87 %, respectively), while SO42- (36.34 %), SO2 (21.00 %) and BC (12.38 %) were more important in lean MAFLD. Being male, aged above 45 years and smoking increased joint effects of air pollutant mixture on overall MAFLD. Conclusions: Air pollutant mixture was associated with MAFLD, particularly the lean MAFLD subtype. CO played a pivotal role in both overall and overweight/obesity MAFLD, whereas SO42- were associated with lean MAFLD.
... A meta-analysis of 16 studies showed that when PM 2.5 increased by 10 µg/m 3 , the risk of liver cancer, liver cirrhosis, and fatty liver disease increased by 23 (95% CI: 14-33%), 17 (6-29%), and 51% (9-108%), respectively [84]. The increased risk of these diseases suggests that air pollution may lead to abnormal liver lipid metabolism, which has also been confirmed in some animal experiments [85][86][87]. Pathological accumulation of triglycerides and other lipids in hepatocytes is a feature of MAFLD [88]. The balance between synthesis and secretion of triglycerides in the liver is very important for the liver to maintain homeostasis of lipid metabolism. ...
Article
Full-text available
Dyslipidemia is a critical factor in the development of atherosclerosis and consequent cardiovascular disease. Numerous pieces of evidence demonstrate the association between air pollution and abnormal blood lipids. Although the results of epidemiological studies on the link between air pollution and blood lipids are unsettled due to different research methods and conditions, most of them corroborate the harmful effects of air pollution on blood lipids. Mechanism studies have revealed that air pollution may affect blood lipids via oxidative stress, inflammation, insulin resistance, mitochondrial dysfunction, and hypothalamic hormone and epigenetic changes. Moreover, there is a risk of metabolic diseases associated with air pollution, including fatty liver disease, diabetes mellitus, and obesity, which are often accompanied by dyslipidemia. Therefore, it is biologically plausible that air pollution affects blood lipids. The overall evidence supports that air pollution has a deleterious effect on blood lipid health. However, further research into susceptibility, indoor air pollution, and gaseous pollutants is required, and the issue of assessing the effects of mixtures of air pollutants remains an obstacle for the future.
... Recent epidemiological studies and animal experiments have confirmed the occurrence of metabolic-associated fatty liver disease (MAFLD) induced by fine particulate matter (PM 2.5 ) exposure. MAFLD is associated with metabolic disorder, hepatic dysfunction, and liver dyslipidemia (Choe et al., 2019;Du et al., 2022b;Li et al., 2020a;Xu et al., 2019). Lipids are essential nutrients that help maintain energy homeostasis and form the constituents of cells and tissues. ...
Article
Background: Nonalcoholic fatty liver disease (NAFLD) is the extremely usual reason of chronic liver disease, extending from simple hepatic steatosis (HS), nonalcoholic steatohepatitis (NASH) to advanced hepatic fibrosis and cirrhosis. Though orlistat is a Food and Drug Administration (FDA) approved drug for long-duration management of obesity, few cases of severe hepatic insult were declared. Melatonin is an efficient antioxidant; it also regulates metabolic processes that lead to fat accumulation and obesity. Aim of the work: The current research aimed to compare the impact of orlistat, melatonin, and their combination on the structural changes of the hepatic tissue of adult male albino rats supplied with high fat diet (HFD). Material and methods: Thirty adult male albino rats divided into five groups. Liver specimens were divided into two parts. One-half was processed to obtain paraffin blocks, and the other half was processed to obtain semithin sections. Morphometric study and statistical analysis were done. Results: Hepatic tissue from the HFD group showed steatosis, ballooning, and inflammation and all these parameters were moderately improved - except for inflammation which worsened with therapy. Combined orlistat and melatonin-treated groups showed marked improvement of all parameters as well as marked improvement in the hepatic fibrosis.Orlistat/Melatonin combination therapy is both safe and effective in comparison to orlistat and melatonin monotherapy.
Article
Aims This study sought to explore the impact of Fusobacterium nucleatum on hepatic steatosis in apolipoprotein E (ApoE) knockout (KO) mice induced by a high‐fat diet (HFD) and elucidate the underlying mechanism. Methods ApoE KO mice, on a HFD, received F. nucleatum oral inoculation every other day. After 24 weeks, body weight, liver weight, and liver index were assessed. Serum biochemistry and pro‐inflammatory factors in serum and liver were analyzed. The histopathology of right maxilla and live were performed. Oil red O, immunohistochemistry, and immunofluorescence staining for the liver were conducted. Myeloperoxidase (MPO) activity, apoptosis, lipid reactive oxygen species (ROS), ROS, lipid peroxides, and hepatic lipids were also evaluated. Liver inflammation, fibrosis, de novo lipogenesis (DNL)‐related molecule, and Nrf2/Keap1‐related signaling molecule gene/protein expression were determined by real‐time PCR (RT‐PCR) and/or Western blot (WB) analysis. Results HFD‐fed ApoE KO mice infected by F. nucleatum demonstrated significant changes, including increased body and liver weight, elevated proinflammatory factors and lipids in serum and liver, as well as neutrophil infiltration, fibrosis, apoptosis, oxidative stress, and lipid peroxidation in the liver. Additionally, F. nucleatum stimulates hepatic lipid accumulation and activates de novo lipogenesis (DNL), while simultaneously suppressing the Nrf2/Keap1 antioxidant pathway. Conclusion In conclusion, our study reveals that oral inoculation of F. nucleatum might promote hepatic steatosis by inhibiting Nrf2/Keap1 pathway.
Article
Background & aims: Recent cross-sectional studies found that exposure to ambient air pollution (AP) was associated with an increased risk of metabolic dysfunction-associated fatty liver disease (MAFLD). The alternation of blood lipids may explain the association, but epidemiological evidence is lacking. We aimed to examine whether and to what extent the association between long-term exposure to AP and incident MAFLD is mediated by blood lipids and dyslipidemia in a prospective cohort. Methods: We included 6350 participants from the China Multi-Ethnic Cohort (CMEC, baseline 2018-2019, follow-up 2020-2021). Three-year average (2016-2018) of AP (PM1, PM2.5, PM10, NO2), blood lipids (TC, LDL-C, HDL-C, TG with their combinations) and incident MAFLD for each individual were assessed chronologically. Linear and logistic regression was used to assess the associations among AP, blood lipids, and MAFLD, and the potential mediation effects of blood lipids were evaluated using causal mediation analysis. Results: A total of 744 participants were newly diagnosed with MAFLD at follow-up. The odds ratios of MAFLD associated with a 10 μm increase in PM1, PM2.5, and PM10 were 1.35 (95 % CI: 1.14, 1.58), 1.34 (1.10, 1.65) and 1.28 (1.14, 1.44), respectively. Blood lipids are important mediators between AP and incident MAFLD. LDL-C (Proportion Mediated: 6.9 %), non-HDL (13.4 %), HDL-C (20.7 %), LDL/HDL (30.1 %), and dyslipidemia (6.5 %) significantly mediated the association between PM2.5 and MAFLD. For PM1, the indirect effects were similar to those for PM2.5, with a larger value for the direct effect, and the mediation proportion by blood lipids was less for NO2. Conclusion: Blood lipids are important mediators between AP and MAFLD, and can explain 5 %-30 % of the association between AP and incident MAFLD, particularly cholesterol-related variables, indicating that AP could lead to MAFLD through the alternation of blood lipids. These findings provided mechanical evidence of AP leading to MAFLD in epidemiological studies.
Article
Full-text available
Background Non-alcoholic fatty liver disease (NAFLD), which is growing more common in the Western world, has become the main cause of chronic liver disease and is strongly associated with metabolism syndromes. NAFLD can indicate a wide spectrum of hepatic pathologies, ranging from simple hepatic steatosis and inflammatory non-alcoholic steatohepatitis to more severe stages of fibrosis and cirrhosis. Moreover, evidence has demonstrated that physical inactivity and westernized dietary habits may facilitate the development of NAFLD. Lipid modulation and metabolism could be important factors in the development of steatosis. Lipid species, characterized using a lipidomic approach with untargeted analysis, could provide potential biomarkers for the pathogenesis of NAFLD or therapeutic applications. Thus, in this study, the effects of exercise on the improvement of NAFLD were further investigated from a lipidomic perspective through the aspects of lipid regulation and metabolism. Methods Wild type (WT) C57BL/6 J and C57BL/6-ApoEem1Narl/Narl mice were assigned to one of four groups: WT mice fed a normal chow diet (CD), apolipoprotein E (ApoE) knockout mice fed a normal CD, ApoE knockout mice fed a high-fat diet (HFD), and ApoE knockout mice fed a HFD and provided with swimming exercise. The treatments (e.g., normal diet, HFD, and exercise) were provided for 12 consecutive weeks before the growth curves, biochemistry, fat composition, pathological syndromes, and lipid profiles were determined. Results Exercise significantly reduced the HFD-induced obesity (weight and fat composition), adipocyte hypertrophy, liver lipid accumulation, and pathological steatosis. In addition, exercise ameliorated HFD-induced steatosis in the process of NAFLD. The lipidomic analysis revealed that the changes in plasma triglyceride (14:0/16:0/22:2), phosphatidic acid (18:0/17:2), and phosphatidylglycerol (16:0/20:2) induced by the administration of the HFD could be reversed significantly by exercise. Conclusions The 12-week regular exercise intervention significantly alleviated HFD-induced NAFLD through modulation of specific lipid species in plasma. This finding could elucidate the lipids effects behind the hepatic pathogenesis with exercise.
Article
Full-text available
To elucidate the mechanism of how the liver participates in PM2.5-caused insulin resistance. A novel Wistar rat model was developed in this study by instilling a suspension of lyophilized PM2.5 sample (2.5 mg/kg, 5 mg/kg, or 10 mg/kg) collected from the atmosphere. Systemic insulin resistance indicators, including serum fasting blood glucose (FBG), fasting insulin (FINS), Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), and hemoglobin A1 (HbA1), were upregulated by the PM2.5 instillation. The area under the curve (AUCglu) calculated by intraperitoneal glucose tolerance testing (IPGTT) was also significantly greater in the PM2.5 instillation groups. Additionally, PM2.5 instillation was found to cause liver damage and inflammation. The serum levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), total bilirubin (TBIL), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) were significantly elevated by PM2.5 instillation. PM2.5 also triggered IL-6 and TNF-α transcription but inhibited mRNA synthesis and suppressed signaling activation of the insulin-phosphoinositide 3-kinase- (PI3K-) Akt-glucose transporter 2 (GLUT2) pathway in the rat liver by reducing the ratio of phosphorylated Akt to phosphorylated insulin receptor substrate 1 (IRS-1). Thus, PM2.5-induced inflammation activation and insulin signaling inhibition in the rat liver contribute to the development of systemic insulin resistance.
Article
Full-text available
Background Apolipoprotein E (ApoE) polymorphisms have been reported to be associated with nonalcoholic fatty liver disease (NAFLD), but the conclusions of studies are inconsistent in different regions. The present study aims to investigate the role of ApoE genotypes on NAFLD in southern China. Methods A total of 1064 subjects including 372 NAFLD patients and 692 controls who attended Meizhou People's Hospital located in southern China from March 1, 2016 to April 30, 2020 were enrolled in this study. The ApoE genotypes were detected and the laboratory parameters were examined. Results Significant differences were observed between NAFLD patients and controls in the prevalence of ε3/ε3 (p < 0.001) and ε3/ε4 (p = 0.004). NAFLD patients presented higher frequency of ε4 allele than controls (p = 0.013). Logistic regression analysis suggested that ε3/ε3 was an independent risk factor (OR: 1.435, 95% CI: 1.084–1.891, p = 0.010), while ε3/ε4 was an independent protective factor (OR: 0.578, 95% CI: 0.404–0.828, p = 0.003) for development of NAFLD. In addition, allele ε4 showed a protective effect on NAFLD with an adjusted OR of 0.588 (95% CI: 0.420–0.824, p = 0.002). Conclusion Our results suggested that ApoE genotype was associated with the development of NAFLD in the population of southern China. Individuals carrying ε3/ε3 were at higher risk of NAFLD, while those carrying ε3/ε4 were at lower risk of NAFLD.
Article
Full-text available
Short-term PM2.5 exposure is related to vascular remodeling and stiffness. Mitochondria-targeted antioxidant MitoQ is reported to improve the occurrence and development of mitochondrial redox-related diseases. At present, there is limited data on whether MitoQ can alleviate the vascular damage caused by PM2.5. Therefore, the current study was aimed to evaluate the protective role of MitoQ on aortic fibrosis induced by PM2.5 exposure. Vascular Doppler ultrasound manifested PM2.5 damaged both vascular function and structure in C57BL/6J mice. Histopathological analysis found that PM2.5 induced aortic fibrosis and disordered elastic fibers, accompanied by collagen I/III deposition and synthetic phenotype remodeling of vascular smooth muscle cells; while these alterations were partially alleviated following MitoQ treatment. We further demonstrated that mitochondrial dysfunction, including mitochondrial reactive oxygen species (ROS) overproduction and activated superoxide dismutase 2 (SOD2) expression, decreased mitochondrial membrane potential (MMP), oxygen consumption rate (OCR), ATP and increased intracellular Ca²⁺, as well as mitochondrial fragmentation caused by increased Drp1 expression and decreased Mfn2 expression, occurred in PM2.5-exposed aorta or human aortic vascular smooth muscle cells (HAVSMCs), which were reversed by MitoQ. Moreover, the enhanced expressions of LC3II/I, p62, PINK1 and Parkin regulated mitophagy in PM2.5-exposed aorta and HAVSMCs were weakened by MitoQ. Transfection with PINK1 siRNA in PM2.5-exposed HAVSMCs further improved the effects of MitoQ on HAVSMCs synthetic phenotype remodeling, mitochondrial fragmentation and mitophagy. In summary, our data demonstrated that MitoQ treatment had a protective role in aortic fibrosis after PM2.5 exposure through mitochondrial quality control, which regulated by mitochondrial ROS/PINK1/Parkin-mediated mitophagy. Our study provides a possible targeted therapy for PM2.5-induced arterial stiffness.
Article
Full-text available
Atherosclerosis and nonalcoholic fatty liver disease are leading causes of morbidity and mortality in the Western countries. The renin–angiotensin system (RAS) with its two main opposing effectors, i.e., angiotensin II (Ang II) and Ang-(1–7), is widely recognized as a major regulator of cardiovascular function and body metabolic processes. Angiotensin-converting enzyme 2 (ACE2) by breaking-down Ang II forms Ang-(1–7) and thus favors Ang-(1–7) actions. Therefore, the aim of our study was to comprehensively evaluate the influence of prolonged treatment with ACE2 activator, diminazene aceturate (DIZE) on the development of atherosclerotic lesions and hepatic steatosis in apoE−/− mice fed a high-fat diet (HFD). We have shown that DIZE stabilized atherosclerotic lesions and attenuated hepatic steatosis in apoE−/− mice fed an HFD. Such effects were associated with decreased total macrophages content and increased α-smooth muscle actin levels in atherosclerotic plaques. Moreover, DIZE changed polarization of macrophages towards increased amount of anti-inflammatory M2 macrophages in the atherosclerotic lesions. Interestingly, the anti-steatotic action of DIZE in the liver was related to the elevated levels of HDL in the plasma, decreased levels of triglycerides, and increased biosynthesis and concentration of taurine in the liver of apoE−/− mice. However, exact molecular mechanisms of both anti-atherosclerotic and anti-steatotic actions of DIZE require further investigations.
Article
Full-text available
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease and is associated with a higher risk of all- cause and cause-specific morbidity and mortality. Animal studies suggest that air pollution may play a role in the development of NAFLD. However, evidence from human studies is limited. Here, we prospectively estimated the association between long-term exposure to fine particulate matter (PM2.5) and risk of NAFLD in 58 026 Taiwan residents who received a standard medical screening programme between 2001 and 2016. We excluded participants with (a) missing values of covariates; (b) excess alcohol intake; (c) liver disease at baseline; (d) NAFLD at baseline and (e) only one medical examination (online supplemental figure S1). The final analytic sample included 35 614 participants for fatty liver index (FLI)-based cohort and 34 741 participants for hepatic steatosis index (HSI)-based cohort.
Article
Full-text available
Melanoma is a leading cause of cancer deaths worldwide. Although immunotherapy has revolutionised the treatment for some patients, resistance towards therapy and unwanted side effects remain a problem for numerous individuals. Broad anti‐cancer activities of melatonin are recognised, however additional investigations still need to be elucidated. Herein, using various human melanoma cell models, we explore in vitro the new insights into the regulation of melanoma by melatonin and its metabolites which possess, on the other side, high safety profiles and biological meaningful. In this study, using melanotic (MNT‐1) and amelanotic (A375, G361, Sk‐Mel‐28) melanoma cell lines, the comparative oncostatic responses, the impact on melanin content (for melanotic MNT‐1 melanoma cells) as well as the mitochondrial function controlled by melatonin, its precursor (serotonin), a kynuric (N¹‐acetyl‐N²‐formyl‐5‐methoxykynuramine, AFMK) and indolic pathway (6‐hydroxymelatonin, 6(OH)MEL and 5‐methoxytryptamine, 5‐MT) metabolites were assessed. Namely, significant disturbances were observed in bioenergetics as follows: (i) uncoupling of oxidative phosphorylation (OXPHOS), (ii) attenuation of glycolysis, (iii) dissipation of mitochondrial transmembrane potential (mtΔΨ) accompanied by (iv) massive generation of reactive oxygen species (ROS), and (v) decrease of glucose uptake. Collectively, these results together with previously published reports provide a new biological potential and make an imperative to consider using melatonin or its metabolites for complementary future treatments of melanoma‐affected patients, however these associations should be additionally investigated in clinical setting.
Article
Maternal hypercholesterolemia (MHC), a pathological condition characterized by an exaggerated rise in maternal serum cholesterol during pregnancy, may influence offspring hepatic lipid metabolism and increase the risk of nonalcoholic fatty liver disease (NAFLD). As NAFLD is characterized by a sexual dimorphic response, we assessed whether early-life exposure to excessive cholesterol influences the development of NAFLD in offspring and whether this occurs in a sex-specific manner. Female apoE−/− mice were randomly assigned to a control (CON) or a high cholesterol (CH; 0.15%) diet prior to breeding. At parturition, a cross-fostering approach was used to establish 3 groups: i) normal cholesterol exposure throughout gestation and lactation (CON-CON); (ii) excessive cholesterol exposure throughout gestation and lactation (CH-CH); and (iii) excessive cholesterol exposure in the gestation period only (CH-CON). Adult male offspring (PND 84) exposed to excessive cholesterol during gestation only (CH-CON) demonstrated hepatic triglyceride (TG) accumulation and reduced lipogenic gene expression. However, male mice with a prolonged cholesterol exposure throughout gestation and lactation (CH-CH) had a similar, but not exacerbated hepatic response. Further, with the exception of higher serum TG in adult CH-CH females, evidence for a programming effect in female offspring was largely absent in comparison with males. These results indicate a sexual dimorphic response with respect to the effect of MHC on later life hepatic steatosis and highlight the gestation period as the most influential malprogramming window for hepatic lipid dysfunction in males.
Article
Lipid metabolic disorder occurs when ApoE gene is deficient. However, the role of Docosahexaenoic acid (DHA) in relieving hepatic lipid metabolic disorder in apolipoprotein E-deficient (ApoE -/-) mice remains unknown. We fed 3-month-old C57BL/6J wild-type (C57 wt) and ApoE -/- mice respectively with normal or DHA fortified diet for 5 months. We found ApoE gene deficiency caused hepatic lipid deposition and increased lipid levels in plasma and liver. Hepatic gene expression of SRB1, CD36 and FABP5 in ApoE -/- mice, protein expression of HMGCR, LRP1 in C57 wt mice and protein expression of LRP1 in ApoE -/- mice increased after DHA intervention. In DHA-fed ApoE -/- mice, LXRα/β and PPARα protein expression down-regulated in cytoplasm, but LXRα/β protein expression up-regulated in nucleus. DHA treatment decreased RXRα and RXRβ expression in C57 wt and ApoE -/- female mice. Deletion of ApoE gene caused lipid metabolism disorder in liver of mice. DHA treatment efficiently meliorated lipid metabolism caused by ApoE deficient through the regulation of gene and protein expressions of molecules involved in liver fatty acids transport and lipid metabolism.
Article
Non-alcoholic fatty liver disease (NAFLD) is a leading cause of chronic liver disease and is highly correlated with metabolic disease. NAFLD results from environmental exposures acting on a susceptible polygenic background. This study performed the largest multiethnic investigation of exonic variation associated with NAFLD and correlated metabolic traits and diseases. An exome array meta-analysis was carried out among eight multiethnic population-based cohorts (n = 16 492) with computed tomography (CT) measured hepatic steatosis. A fixed effects meta-analysis identified five exome-wide significant loci (P < 5.30x10-7); including a novel signal near TOMM40/APOE. Joint analysis of TOMM40/APOE variants revealed the TOMM40 signal was attributed to APOE rs429358-T; APOE rs7412 was not associated with liver attenuation. Moreover, rs429358-T was associated with higher serum alanine aminotransferase, liver steatosis, cirrhosis, triglycerides and obesity; as well as, lower cholesterol and decreased risk of myocardial infarction (MI) and Alzheimer's disease (ad) in phenome-wide association analyses in the Michigan Genomics Initiative, United Kingdom Biobank and/or public datasets. These results implicate APOE in imaging-based identification of NAFLD. This association may or may not translate to non-alcoholic steatohepatitis (NASH); however, these results indicate a significant association with advanced liver disease and hepatic cirrhosis. These findings highlight allelic heterogeneity at the APOE locus and demonstrate an inverse link between NAFLD and ad at the exome level in the largest analysis to date.