ArticlePDF Available

Abstract and Figures

Hepatic fibrosis, featured by accumulation of excessive extracellular matrix in liver tissues, is associated with metabolic disease and cancer. Inhalation exposure to airborne particulate matter in fine ranges (PM2.5) correlates with pulmonary dysfunction, cardiovascular disease, and metabolic syndrome. In this study, we investigated the effect and mechanism of PM2.5 exposure on hepatic fibrogenesis. Both inhalation exposure of mice and in vitro exposure of specialized cells to PM2.5 were performed to elucidate the effect of PM2.5 exposure on hepatic fibrosis. Histological examinations, gene expression analyses, and genetic animal models were utilized to determine the effect and mechanism by which PM2.5 exposure promotes hepatic fibrosis. Inhalation exposure to concentrated ambient PM2.5 induces hepatic fibrosis in mice under the normal chow or high-fat diet. Mice after PM2.5 exposure displayed increased expression of collagens in liver tissues. Exposure to PM2.5 led to activation of the transforming growth factor β (TGFβ)-SMAD3 signaling, suppression of peroxisome proliferator-activated receptor γ (PPARγ), and expression of collagens in hepatic stellate cells. NADPH oxidase plays a critical role in PM2.5-induced liver fibrogenesis. Exposure to PM2.5 exerts discernible effects on promoting hepatic fibrogenesis. NADPH oxidase mediates the effects of PM2.5 exposure on promoting hepatic fibrosis. Copyright © 2015. Published by Elsevier B.V.
PM 2.5 exposure induces hepatic fibrosis in mouse liver. (A and B) Sirius-red staining of hepatic collagen deposition (A) and Masson's trichrome staining of collagen fiber (B) in formalin-fixed liver tissue sections from C57BL/6 mice under the normal chow (NC) or high-fat diet (HFD) exposed to FA or PM 2.5 for 10 weeks. Magnifications: 200Â. The arrows point out areas of hepatic fibrosis. (C) Immunohistochemical (IHC) staining of the hepatic stellate cell surface marker a-SMA in the liver tissue sections from the NC or HFD fed mice exposed to FA or PM 2.5 for 10 weeks. Magnifications: 200Â. (D) Hepatic fibrosis grades of the mice exposed to PM 2.5 or FA for 10 weeks. Hepatic fibrosis grades were determined based on the histological analyses of Sirius-red staining of collagens, according to the Scheuer scoring system for fibrosis and cirrhosis [16,17]. Data are shown as mean ± SEM (n = 8 FA-or 9 PM 2.5-exposed animals). (E) Immunoblotting analysis of collagen I (Col1) in the liver tissue from the mice exposed to PM 2.5 or FA. The values below gel images represent quantification of Col1 protein signal intensities after normalization to those of b-actin. (F) Serum levels of activated TGFb1 (determined by ELISA) in the mice exposed to PM 2.5 or FA. For A-B, each bar denotes the mean ± SEM (n = 3). (G) Quantitative real-time PCR (qRT-PCR) analysis of expression levels of the Tgfb1 mRNA in the livers of the mice exposed to PM 2.5 or FA for 10 weeks. Fold changes of mRNA levels were shown by comparing to the FA-exposed control mice. From B-G, * p <0.05; ** p <0.01. (This figure appears in colour on the web.)
… 
Exposure to PM 2.5 stimulates TGFb signaling in macrophages and collagen production in HSC. (A) Levels of secreted TGFb1 in primary hepatocytes, RAW264.7 cells, and LX-2 cells upon the challenge of PM 2.5 (5 lg/ml) for different time intervals as indicated. The levels of TGFb1 in the culture medium were determined by ELISA and presented after normalization to cell numbers. Each time point represents the mean ± SEM (n = 3 biological replicates). (B) qRT-PCR analysis of expression levels of the Tgfb1, Tgfb2, and Tgfb3 mRNAs in RAW264.7 cells challenged with PM 2.5 for different time intervals as indicated. Fold changes of mRNA levels were shown by comparing to the vehicle (PBS)-treated controls. Each bar represents the mean ± SEM (n = 3 biological replicates). (C) Immunoblotting analysis of total and phosphorylated SMAD3 in LX-2 cells cultured in the conditioned medium from RAW264.7 cells exposed to PM 2.5 for 12 or 24 h. The graph beside the images showed fold changes of phosphorylated SMAD3 levels after normalization to total SMAD3 levels. Each bar denotes the mean ± SEM (n = 3 biological replicates). (D) qRT-PCR analysis of expression levels of the Collagen 1a1 (Col1a1) and Collagen 4a4 (Col4a4) mRNA in LX-2 cells cultured in the conditioned medium from RAW264.7 cells exposed to PM 2.5 for 12 or 24 h. LX-2 cells were incubated with the conditioned medium for 36 h before subjected to the real-time RT-PCR analysis. Fold changes of mRNA are shown by comparing to the vehicle control. Each bar denotes the mean ± SEM (n = 3 biological replicates). (E) Immunoblotting analysis of Col1 protein levels in LX-2 cells cultured in the conditioned medium from RAW264.7 cells exposed to PM 2.5 for 12 or 24 h. The graph beside the images showed fold changes of collagen protein levels after normalization to those of Tubulin. Each bar denotes the mean ± SEM (n = 3 biological replicates). From A–E, * p <0.05; ** p <0.01. (F) Immunofluorescent analysis of Col1 production in the LX-2 cells cultured in the conditioned medium from RAW264.7 cells exposed to PM 2.5 for 10 h. LX-2 cells were incubated with the conditioned medium for 36 h before subjected to the immunofluorecent analysis. LX-2 cells were cultured in the conditioned medium from RAW264.7 cells exposed to PBS (vehicle) as the control. The nucleus was stained with DAPI. The experiments were repeated three times, and representative images were shown. (This figure appears in colour on the web.)
… 
Hepatic fibrosis under PM 2.5 exposure relies on NOX activity. (A) Histological analysis of hepatic collagen deposition (Sirius-red staining) in formalin-fixed liver tissue sections from p47phox À/À and wild-type control mice exposed to FA or PM 2.5 for 10 weeks. Magnifications: 200Â. The arrows point out areas of hepatic fibrosis. (B) Hepatic fibrosis grades of the p47phox À/À and control mice exposed to PM 2.5 or FA. Hepatic fibrosis grades were determined based on the histological analyses of Sirius-red staining of collagens, according to the Scheuer scoring system for fibrosis and cirrhosis [16,17]. Data are shown as mean ± SEM (n = 4 animals per group). (C) qRT-PCR analysis of expression levels of Col1 mRNA in p47phox À/À and control mice exposed to FA or PM 2.5 for 10 weeks. Fold changes of mRNA are shown by normalizing mRNA levels to that of the FA-exposed control animals. Each bar denotes the mean ± SEM (n = 3 animals per group). (D) DHE staining of ROS signals in the liver tissue sections from the mice exposed to FA or PM 2.5. The oxidative red fluorescence was detected by a Zeiss fluorescence microscope. Magnification: 400Â. (E) Quantification of DHE-stained ROS signals in the PM 2.5-and FA-exposed p47phox À/À and control mice. DHE signals were quantified by counting the number of positive stained nuclei in 8 random fields. Microscopic interference contrast was used to exclude positive signals from non-cell origin. The percentages of DHE-positive nuclei (compared to total nuclei) were shown. Data are shown as mean ± SEM (n = 4 animals per group). * p <0.05. (F) Oil-red O staining of hepatic lipid droplets in the livers of p47phox À/À and wild-type control mice exposed to FA or PM 2.5 for 10 weeks (magnifications: 200Â). (G) qRT-PCR analysis of expression levels of PPARc mRNA in the liver tissues of p47phox À/À and control mice exposed to FA or PM 2.5 for 10 weeks. Fold changes of mRNA are shown by normalizing mRNA levels to that of the FA-exposed control animals. Each bar denotes the mean ± SEM (n = 4 animals per group). For B, C, E, and G, * p <0.05. (This figure appears in colour on the web.)
… 
Content may be subject to copyright.
Exposure to fine airborne particulate matters induces hepatic
fibrosis in murine models
Ze Zheng
1
, Xuebao Zhang
1
, Jiemei Wang
1
, Aditya Dandekar
2
, Hyunbae Kim
1
, Yining Qiu
1
,
Xiaohua Xu
4
, Yuqi Cui
3
, Aixia Wang
3,4
, Lung Chi Chen
5
, Sanjay Rajagopalan
3,
, Qinghua Sun
3,4
,
Kezhong Zhang
1,2,
1
Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA;
2
Department of
Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
3
Division of Cardiovascular Medicine,
Davis Heart & Lung Research Institute, College of Medicine, Ohio State University, Columbus, OH 43210, USA;
4
Division of Environmental Health Sciences, College of Public Health, Ohio State University, Columbus, OH 43210, USA;
5
Department of Environmental Medicine, New York University, Tuxedo, NY 10987, USA
Background & Aims: Hepatic fibrosis, featured by the accumula-
tion of excessive extracellular matrix in liver tissue, is associated
with metabolic disease and cancer. Inhalation exposure to air-
borne particulate matter in fine ranges (PM
2.5
) correlates with
pulmonary dysfunction, cardiovascular disease, and metabolic
syndrome. In this study, we investigated the effect and mecha-
nism of PM
2.5
exposure on hepatic fibrogenesis.
Methods: Both inhalation exposure of mice and in vitro exposure
of specialized cells to PM
2.5
were performed to elucidate the
effect of PM
2.5
exposure on hepatic fibrosis. Histological examina-
tions, gene expression analyses, and genetic animal models were
utilized to determine the effect and mechanism by which PM
2.5
exposure promotes hepatic fibrosis.
Results: Inhalation exposure to concentrated ambient PM
2.5
induces hepatic fibrosis in mice under the normal chow or
high-fat diet. Mice after PM
2.5
exposure displayed increased
expression of collagens in liver tissues. Exposure to PM
2.5
led to
activation of the transforming growth factor b-SMAD3 signaling,
suppression of peroxisome proliferator-activated receptor
c
, and
expression of collagens in hepatic stellate cells. NADPH oxidase
plays a critical role in PM
2.5
-induced liver fibrogenesis.
Conclusions: Exposure to PM
2.5
exerts discernible effects on pro-
moting hepatic fibrogenesis. NADPH oxidase mediates the effects
of PM
2.5
exposure on promoting hepatic fibrosis.
Ó2015 Published by Elsevier B.V. on behalf of the European
Association for the Study of the Liver.
Introduction
Recent studies indicated that exposure to fine ambient particu-
late matter (aerodynamic diameter <2.5
l
m, PM
2.5
) is a risk fac-
tor for pulmonary and cardiovascular diseases as well as
metabolic syndrome [1–3]. Traffic-related airborne PM
2.5
is a
complex mixture of particles and gases from gasoline and diesel
engines, together with dust from wear of road surfaces, tires, and
brakes [4,5]. Airborne PM
2.5
demonstrates an incremental capac-
ity to penetrate into the distal airway units and potentially enter
the systemic circulation with diminishing sizes. It has been sug-
gested that the cytotoxic effects of PM
2.5
are more associated
with PM
2.5
as a complex other than single or a few components
of PM
2.5
particles [6]. The particle sizes, charges, and combined
effects of individual components of PM
2.5
are all crucial to the
adverse health impact of PM
2.5
exposure. Studies from our group
and others suggested that PM
2.5
exposure triggers a variety of
maladaptive signaling pathways in the lung, blood vessels, liver,
and adipose tissues that are associated with endoplasmic reticu-
lum (ER) stress, oxidative stress, and inflammatory responses
[1,7–12]. Moreover, we recently demonstrated an important find-
ing that inhalation exposure to PM
2.5
causes a non-alcoholic
steatohepatitis (NASH)-like phenotype and depletion of hepatic
glycogen storage in animals [1]. Through both in vivo and
in vitro analyses, we revealed the signaling pathways through
which PM
2.5
exposure promotes NASH-associated activities and
impairment of hepatic glucose metabolism. We identified the dis-
ruption of hepatic lipid/glucose homeostasis, lobular and portal
inflammation, as well as mild hepatic steatosis in the liver of
the mice exposed to PM
2.5
for 10 weeks [1]. However, the pro-
nounced effect of PM
2.5
exposure on modulating hepatic path-
ways associated with liver fibrogenesis has not been
characterized.
Liver fibrosis and cirrhosis are the advanced stages of chronic
liver injuries caused by chronic hepatitis viral infection, obesity,
alcoholism, or autoimmune diseases. Recent studies showed that
PM
2.5
exposure activates Kupffer cells in murine liver tissues,
indicating that PM
2.5
represents a risk factor for NAFLD progres-
sion [1,10,13]. In this study, we used a ‘‘real-world’’ PM
2.5
Journal of Hepatology 2015 vol. 63 j1397–1404
Keywords: Air pollution; Hepatic fibrosis; PM
2.5
.
Received 9 September 2014; received in revised form 7 July 2015; accepted 16 July
2015; available online 26 July 2015
Corresponding author. Address: 540 E. Canfield Avenue, Detroit, MI 48201, USA.
Tel.: +1 313 577 2669; fax: +1 313 577 5218.
E-mail address: kzhang@med.wayne.edu (K. Zhang).
Current address: Division of Cardiology, University of Maryland School of
Medicine, Baltimore, MD 21201, USA.
Abbreviations: PM, ambient particulate matter; PM
2.5
, PM with aerodynamic
diameter less than 2.5
l
m; FA, filtered air; OASIS, Ohio’s Air Pollution Exposure
System for the Interrogation of Systemic Effects; PPAR, peroxisome
proliferator-activated receptor; TGFb, transforming growth factor b;HSC,
hepatic stellate cells; p47phox, Neutrophil cytosolic factor 1; NOX, NADPH
peroxidase; ROS, reactive oxygen species.
Research Article
exposure system, ‘‘Ohio’s Air Pollution Exposure System for the
Interrogation of Systemic Effects (OASIS)’’, to perform
whole-body exposure to mice of environmentally relevant
PM
2.5
. We demonstrate that exposure to PM
2.5
causes a dis-
cernible phenotype of hepatic fibrosis in animals. Through both
in vivo and in vitro analyses, we reveal the signaling pathways
through which PM
2.5
exposure promotes hepatic
fibrogenesis-associated activities. The information from this work
has important implications in the understanding and treatment
of air pollution-induced liver diseases.
Materials and methods
Animal experiments
C57BL/6 male mice of six-weeks-old were purchased from the Jackson Laborato-
ries (Bar Harbor, ME), and were equilibrated for two weeks prior to experimental
enrollment. The mice were housed in cages with regular chow or a high-fat diet
(Teklad TD 88137, 42% calories from fat) in an Association for Assessment and
Accreditation of Laboratory Animal Care-accredited animal housing facility. All
the animal experiments were approved by the Ohio State University and the
Wayne State University IACUC committee and carried out under the institutional
guidelines for ethical animal use.
Exposure of animals to ambient PM
2.5
Mice were exposed to concentrated ambient PM
2.5
or filtered air (FA) in the OASIS
in Columbus, OH, where most of the PM
2.5
component is attributed to long-range
transport [10]. The concentrated PM
2.5
was generated using a versatile aerosol
concentration enrichment system (VACES) as we described previously [14]. Mice
under the normal chow or high-fat diet were exposed to concentrated PM
2.5
for
six hours per day, five days per week for 10 weeks or 9 months [1,10]. The control
(FA) mice were exposed to an identical protocol with the exception of a
high-efficiency particulate-air filter positioned in the inlet valve to remove all
of the PM
2.5
in the filtered air stream. Mice deficient in the cytosolic subunit of
the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (p47phox
/
)
and wild-type control mice of C57BL/6 strain background (both from Jackson
Laboratories) were exposure to FA or PM
2.5
beginning at the age of three weeks
for a duration of 10 weeks.
Histological scoring for hepatic fibrosis
Paraffin-embedded mouse liver tissue sections (5
l
m) were subjected to
Sirius-red or Masson’s trichrome staining for hepatic fibrosis. The histological
analysis of liver fibrosis were as described previously [15,16]. Each section was
examined by a specialist who was blinded to the sample information. Hepatic
fibrosis were scored according to the modified Scheuer scoring system for fibrosis
and cirrhosis [16,17]. The fibrosis stage scores were based on the 0–4 stage sys-
tem: 0, none; 1, zone 3 perisinusoidal fibrosis; 2, zone 3 perisinusoidal fibrosis
plus portal fibrosis; 3, perisinusoidal fibrosis, portal fibrosis, plus bridging fibro-
sis; and 4, cirrhosis.
Statistics
Experimental results are shown as mean ± SEM (for variation between animals or
experiments). All in vitro experiments were repeated with biological triplicates at
least three times independently. The data were analyzed and compared by paired,
two-tailed Student’s ttests. Multiple comparisons were compared with ANOVA
and proceeded by ad hoc statistical test when necessary. Statistical tests with
p<0.05 were considered significant.
For a full description of materials and methods used in this work, see Supple-
mentary data.
β-actin
0
1
2
3
4
5
6
0
20
40
60
80
100
120
140
160
180
200
*
0.0
0.5
1.0
1.5
2.0
2.5
3.0
0.0
0.2
0.4
0.6
0.8
1.0
1.2
1.4
1.6
1.8
FA
MPAF 2.5
** *
Fibrosis scores
(arbitrary units)
ABC
FA
α-SMA IHC
FA FA
Col1
1 2 3 1 2 3
FA
0
20
40
60
80
100
120
140
160 **
Activated TGFβ1 in mouse
serum (ng/ml)
FA
*
TGFβ1 mRNA in mouse liver
(fold changes)
FA
FA
D
NC
HFD
NC
HFD
NC
HFD
FA
Masson’s trichromeSirius-red
FA
Normal chow
Normal chow HFD
0.0
0.2
0.4
0.6
0.8
1.0
1.2
1.4
1.6
1.8
2.0
Normal chow Normal chow
EFG
Col1/actin
0.09
0.06
0.07
0.14
0.19
0.41
0.01
0.02
0.02
1.01
0.71
0.95
HFD
PM 2.5
PM 2.5 PM 2.5
1 2 3 1 2 3
PM 2.5 PM 2.5 PM 2.5 PM 2.5
HFD HFD
PM 2.5 PM 2.5 PM 2.5
Fig. 1. PM
2.5
exposure induces hepatic fibrosis in mouse liver. (A and B) Sirius-red staining of hepatic collagen deposition (A) and Masson’s trichrome staining of collagen
fiber (B) in formalin-fixed liver tissue sections from C57BL/6 mice under the normal chow (NC) or high-fat diet (HFD) exposed to FA or PM
2.5
for 10 weeks. Magnifications:
200. The arrows point out areas of hepatic fibrosis. (C) Immunohistochemical (IHC) staining of the hepatic stellate cell surface marker
a
-SMA in the liver tissue sections
from the NC or HFD fed mice exposed to FA or PM
2.5
for 10 weeks. Magnifications: 200. (D) Hepatic fibrosis grades of the mice exposed to PM
2.5
or FA for 10 weeks.
Hepatic fibrosis grades were determined based on the histological analyses of Sirius-red staining of collagens, according to the Scheuer scoring system for fibrosis and
cirrhosis [16,17]. Data are shown as mean ± SEM (n = 8 FA- or 9 PM
2.5
-exposed animals). (E) Immunoblotting analysis of collagen I (Col1) in the liver tissue from the mice
exposed to PM
2.5
or FA. The values below gel images represent quantification of Col1 protein signal intensities after normalization to those of b-actin. (F) Serum levels of
activated TGFb1 (determined by ELISA) in the mice exposed to PM
2.5
or FA. For A-B, each bar denotes the mean ± SEM (n = 3). (G) Quantitative real-time PCR (qRT-PCR)
analysis of expression levels of the Tgfb1mRNA in the livers of the mice exposed to PM
2.5
or FA for 10 weeks. Fold changes of mRNA levels were shown by comparing to the
FA-exposed control mice. From B-G,
*
p<0.05;
**
p<0.01. (This figure appears in colour on the web.)
Research Article
1398 Journal of Hepatology 2015 vol. 63 j1397–1404
Results
Inhalation exposure to PM
2.5
induces discernible hepatic fibrosis in
mice
To elucidate in vivo effects of PM
2.5
exposure, male C57BL/6 mice
on the normal chow or high-fat diet were exposed to concen-
trated ambient PM
2.5
or FA in exposure chambers of OASIS
located at Columbus, USA, where most of the PM
2.5
is attributed
to long-range transport [1,10] (Supplementary Fig. 1). OASIS is a
VACES through which fine and ultrafine particles are concen-
trated and exposed to the animals in the chambers [10,18].It
has been demonstrated that the distribution and size of concen-
trated PM
2.5
collected from the exposure chamber air truly reflect
that of non-concentrated PM
2.5
present in the ambient air
[14,19]. At the same exposure site, animals were exposed to
PM
2.5
or FA for 10 weeks or 9 months in two different time peri-
ods. During the first exposure period, the ambient mean daily
PM
2.5
concentration at the study site was 6.5
l
g/m
3
, while the
mean concentration of PM
2.5
in the exposure chamber was
74.6
l
g/m
3
[1]. During the second exposure period, the ambient
mean daily PM
2.5
concentration at the site was 15.8
l
g/m
3
, and
the mean concentration of PM
2.5
in the exposure chamber was
111.0
l
g/m
3
[20]. Previously, we reported that the elemental
composition, as measured by energy-dispersive X-ray fluores-
cence (ED-XRF) analysis, include alkali metals, alkaline earth
metals, transition metals, poor metals, non-metals, metalloid,
and halogens [10].
To evaluate effects of PM
2.5
exposure on liver fibrosis, we first
performed histological analyses with liver tissue sections of the
mice exposed to PM
2.5
or FA for 10 weeks. Sirius-red and Mas-
son’s trichrome staining of collagen deposition revealed signifi-
cant perisinusoidal fibrosis in the liver of the mice under
normal chow diet exposed to PM
2.5
(Fig. 1A, B). The fibrosis
grades of the PM
2.5
-exposed mice were significantly higher than
that of the FA-exposed mice (Fig. 1D). We evaluated production
of collagen proteins in the livers of the mice under PM
2.5
expo-
sure. Levels of collagen I, the major collagen protein in liver extra-
cellular matrix, were significantly increased in the livers of the
PM
2.5
-exposed mice (Fig. 1E). These results indicate a discernible
effect of PM
2.5
exposure on promoting hepatic fibrosis. Further-
more, we evaluated hepatic fibrosis in the PM
2.5
-exposed animals
under the high-fat diet. The grades of hepatic fibrosis in the ani-
mals that were simultaneously fed the high-fat diet and exposed
to PM
2.5
for 10 weeks were significantly higher than that of
FA-exposed control animals (Fig. 1A, B, D, E). Notably, the
high-fat fed, PM
2.5
-exposed animals displayed advanced stages
of hepatic fibrosis, including portal fibrosis and bridging fibrosis,
as shown by histological analysis of collagen disposition (Fig. 1A),
suggesting that PM
2.5
exposure may interact with the high-fat
diet to exacerbate its effect in promoting fibrogenesis. Addition-
ally, we examined hepatic fibrosis in the animals under the nor-
mal chow diet exposed to PM
2.5
for 9 months. Interestingly, the
hepatic fibrosis grades in the animals after 9 months PM
2.5
expo-
sure were only insignificantly higher than those of the
FA-exposed animals (Supplementary Fig. 2A, B). This observation
suggests that the animals may possess adaptation mechanisms to
attenuate the effect of a single environmental risk factor during
the chronic PM
2.5
exposure.
Next, we examined expression of
a
-smooth muscle actin
(
a
-SMA), a prominent marker for activation of hepatic stellate
cells (HSC) and fibrosis progression [21], in the livers of PM
2.5
-
or FA-exposed mice under the normal chow or high-fat diet.
Immunohistochemical analysis showed markedly increased
a
-SMA staining in the mouse liver tissues upon PM
2.5
exposure,
suggesting a strong effect of PM
2.5
exposure on HSC activation
(Fig. 1C). In hepatic fibrosis, the release of latent transforming
growth factor b(TGFb) stimulates HSC to produce collagens, a
key event of hepatic fibrogenesis [22,23]. Enzyme-linked
immunosorbent assay (ELISA) indicated that serum levels of acti-
vated TGFb1 in the mice exposed to PM
2.5
were significantly
higher than those of the FA-exposed mice (Fig. 1F). Gene expres-
sion analysis confirmed higher expression levels of Tgfb1mRNA
in the livers of mice exposed to PM
2.5
(Fig. 1G). Together, these
results implicate that TGFb, the inflammatory trigger of hepatic
fibrosis, is upregulated upon PM
2.5
challenge.
Exposure to PM
2.5
stimulates TGFbrelease by macrophages and
collagen production by HSC
In the liver, fibrogenesis is a coordinated process that involves
multiple specialized cell types, including Kupffer cells (resident
macrophages), Ito cells (quiescent adipocytes), HSC, and extracel-
lular matrix [22]. Upon liver injuries or chronic stress, Ito cells
can be activated and differentiate into myofibroblastic HSC with
decreased peroxisome proliferator-activated receptor
c
(PPAR
c
)
expression and fat-storing function [24,25]. In this process, TGFb
produced by inflammatory cells facilitates the differentiation of
myofibroblastic HSC, which is specialized in producing extracel-
lular matrix proteins, especially collagens, to promote hepatic
fibrogenesis [22,23]. It has been reported that three major liver
cell types, Kupffer cells, HSC, and hepatocytes, can produce TGFb
in liver pathogenesis [26,27]. Previously we demonstrated that
PM
2.5
exposure activates Kupffer cells in animal models [1].
Numbers of activated macrophages and induction of
macrophage-associated inflammatory cytokine genes, including
TGFb1,IL1b,IL6,TNF
a
,IL8,CCL2,CCL3, and Gro1, were increased
in the liver of PM
2.5
-exposed mice (Supplementary Fig. 3A–D).
To define the target liver cell types that produce TGFbunder
PM
2.5
exposure, we exposed mouse primary hepatocytes, macro-
phage cell line RAW264.7, and LX-2, a human HSC cell line that
retains the key features of HSC and has been used to study hep-
atic fibrosis and liver disease [28],toPM
2.5
particles. In response
to PM
2.5
challenge, levels of activated TGFbin the culture med-
ium from RAW264.7 cells, but not hepatocytes or LX-2 cells, were
increased in a time-dependent manner (Fig. 2A). Gene expression
analysis confirmed that expression of the mRNAs encoding Tgfb1,
Tgfb2, and Tgfb3in RAW264.7 cells was significantly increased
upon PM
2.5
exposure (Fig. 2B). These results suggest that macro-
phage is the major cell type that produces TGFbunder PM
2.5
exposure.
We further assessed the effects of PM
2.5
on
fibrogenesis-associated signaling in HSC. LX-2 cells were cultured
in the conditioned medium from the macrophage cell line
RAW264.7 exposed to PM
2.5
or vehicle for 12 and 24 h, respec-
tively. Upon PM
2.5
exposure, phosphorylation of SMAD3, a key
mediator of TGFb-triggered fibrotic response [29], was signifi-
cantly increased in LX-2 cells in a time-dependent manner
(Fig. 2C). Correlated to the phosphorylation of SMAD3, expression
levels of both mRNAs and proteins of collagen I and
a
-SMA were
increased in LX-2 cells incubated with the conditioned media
from PM
2.5
-exposed macrophages (Fig. 2D, E; Supplementary
JOURNAL OF HEPATOLOGY
Journal of Hepatology 2015 vol. 63 j1397–1404 1399
Fig. 5). Additionally, we visualized production of collagen I and
collagen IV in LX-2 cells upon PM
2.5
challenge through
immunofluorescent analysis. LX-2 cells cultured in the condi-
tioned medium from PM
2.5
-exposed RAW264.7 cells produced
much more collagen I and collagen IV proteins than those cul-
tured in the control medium (Fig. 2F; Supplementary Fig. 4), thus
confirming the effect of PM
2.5
exposure on collagen production
by HSC. Furthermore, to verify whether the PM
2.5
-induced colla-
gen production from HSC depends on TGFbsignaling, we incu-
bated LX2 cells with the specific TGF receptor (TGFR) inhibitor
SB431542 prior to the culture with the conditioned medium from
PM
2.5
- or vehicle-exposed RAW264.7 cells. Under PM
2.5
-exposed
conditioned medium, pre-treatment of SB431542 significantly
reduced both mRNA and protein levels of collagen I in LX2 cells
(Supplementary Fig. 6), suggesting a crucial role of TGFbsignaling
in PM
2.5
-induced hepatic fibrogenesis as well as the effectiveness
of TGFbreceptor antagonist SB431542 in suppressing
PM
2.5
-induced hepatic fibrosis.
Exposure to PM
2.5
represses PPAR
c
in HSC
It is known that expression of PPAR
c
in HSC is closely associated
with fibrosis in the liver under injuries or chronic stress [24,25].
The decrease in expression of PPAR
c
enables the quiescent adipo-
cytes (Ito cells) to be activated and further differentiated into
myofibroblastic HSC [24,25]. Previously, we showed that inhala-
tion exposure to PM
2.5
reduces expression of PPAR
c
and PPAR
a
in
liver tissues [1]. However, we have not determined whether
PM
2.5
exposure modulates PPAR
c
expression in stellate cells. To
determine whether PM
2.5
exposure can modulate PPAR
c
expres-
sion in HSC, we examined levels of PPAR
c
in the HSC cell line
LX-2 upon PM
2.5
challenge. LX-2 cells were cultured in the condi-
tioned medium from macrophage cell line RAW264.7 exposed to
PM
2.5
for 12 or 24 h. Upon exposure to the conditioned medium,
both protein and mRNA levels of PPAR
c
were gradually decreased
in LX-2 cells in a manner that depends on the PM
2.5
exposure
time of RAW264.7 cells (Fig. 3A, B).
Pioglitazone is a commonly-used PPAR
c
agonist to prevent
inflammation, fibrosis, and insulin resistance in type II diabetes
patient by increasing PPAR
c
levels [30]. To test whether pioglita-
zone can prevent PM
2.5
-triggered downregulation of PPAR
c
in
HSC, LX-2 cells were incubated with pioglitazone for 48 h
followed by incubation with the conditioned medium from
PM
2.5
-exposed RAW264.7 cells. The pre-treatment of pioglitazone
at the concentration of 1
l
M can partially rescue the downregu-
lation of PPAR
c
1and PPAR
c
2mRNA expression in the LX-2 cells
0.0
0.5
1.0
1.5
2.0
2.5
3.0
3.5
4.0
4.5
0
100
200
300
400
500
600
700
800
900
Primary hepatocytes
Raw264.7
LX2
0
1
2
3
4
5
6
7
8
9
0
50
100
150
200
250
300
ABC
Veh 12 h 24 h
p-SMAD3
SMAD3
β-actin
Tgfβ1 Tgfβ2 Tgfβ3
*
***
**
**
**
n.s.
**
*
Ctl
12 h
24 h
24 h 2 h 8 h 24 h
Activated mouse TGFβ
1
(pg/ml)
Vehicle
DAPI
Col1
Merged
D
*
*
*
Col1α1 Col4α4
Veh
12 h
24 h
Col1
Tubulin
Veh 12 h 24 h
E
mRNA levels in RAW264.7
cells (fold changes)
Veh 12 h 24 h
Ctl
Collagen I in LX2 cells
(% of control)
*
0
20
40
60
80
100
120
140
160
FA 12 h 24 h
p-Smad3 / Smad3 in LX2 cells
(% of control)
Collagen mRNA levels in LX2
cells (fold change)
PM 2.5
n.s.
PM 2.5
PM 2.5
PM 2.5
PM 2.5
PM 2.5
PM 2.5
PM 2.5
n.s.
PM 2.5
PM 2.5
n.s.
F
Fig. 2. Exposure to PM
2.5
stimulates TGFbsignaling in macrophages and collagen production in HSC. (A) Levels of secreted TGFb1 in primary hepatocytes, RAW264.7
cells, and LX-2 cells upon the challenge of PM
2.5
(5
l
g/ml) for different time intervals as indicated. The levels of TGFb1 in the culture medium were determined by ELISA and
presented after normalization to cell numbers. Each time point represents the mean ± SEM (n = 3 biological replicates). (B) qRT-PCR analysis of expression levels of the
Tgfb1,Tgfb2, and Tgfb3mRNAs in RAW264.7 cells challenged with PM
2.5
for different time intervals as indicated. Fold changes of mRNA levels were shown by comparing to
the vehicle (PBS)-treated controls. Each bar represents the mean ± SEM (n = 3 biological replicates). (C) Immunoblotting analysis of total and phosphorylated SMAD3 in LX-2
cells cultured in the conditioned medium from RAW264.7 cells exposed to PM
2.5
for 12 or 24 h. The graph beside the images showed fold changes of phosphorylated SMAD3
levels after normalization to total SMAD3 levels. Each bar denotes the mean ± SEM (n = 3 biological replicates). (D) qRT-PCR analysis of expression levels of the Collagen 1
a
1
(Col1
a
1) and Collagen 4
a
4(Col4
a
4) mRNA in LX-2 cells cultured in the conditioned medium from RAW264.7 cells exposed to PM
2.5
for 12 or 24 h. LX-2 cells were incubated
with the conditioned medium for 36 h before subjected to the real-time RT-PCR analysis. Fold changes of mRNA are shown by comparing to the vehicle control. Each bar
denotes the mean ± SEM (n = 3 biological replicates). (E) Immunoblotting analysis of Col1 protein levels in LX-2 cells cultured in the conditioned medium from RAW264.7
cells exposed to PM
2.5
for 12 or 24 h. The graph beside the images showed fold changes of collagen protein levels after normalization to those of Tubulin. Each bar denotes
the mean ± SEM (n = 3 biological replicates). From A–E,
*
p<0.05;
**
p<0.01. (F) Immunofluorescent analysis of Col1 production in the LX-2 cells cultured in the conditioned
medium from RAW264.7 cells exposed to PM
2.5
for 10 h. LX-2 cells were incubated with the conditioned medium for 36 h before subjected to the immunofluorecent
analysis. LX-2 cells were cultured in the conditioned medium from RAW264.7 cells exposed to PBS (vehicle) as the control. The nucleus was stained with DAPI. The
experiments were repeated three times, and representative images were shown. (This figure appears in colour on the web.)
Research Article
1400 Journal of Hepatology 2015 vol. 63 j1397–1404
cultured in the conditioned media from PM
2.5
-exposed RAW264.7
cells (Fig. 3C, D). Interestingly, when the concentration of pioglita-
zone was increased to 2.5
l
M, the pioglitazone pre-treatment was
effective in increasing expression of PPAR
c
2, but not PPAR
c
1,in
LX-2 cells. Immunoblotting analysis confirmed that pioglitazone
treatment at the concentration of 1
l
M can rescue the downreg-
ulation of PPAR
c
protein levels by PM
2.5
in LX-2 cells (Fig. 3E).
These results imply the potential role of the PPAR
c
antagonist
pioglitazone in preventing PM
2.5
-induced hepatic fibrogenesis
by attenuating PM
2.5
-induced PPAR
c
downregulation.
PM
2.5
-induced hepatic fibrosis relies on NADPH oxidase
We explored potential involvements of cell stress sensors in
PM
2.5
-induced hepatic fibrosis. It is known that NADPH oxidase
(NOX) generates large amounts of superoxide anions in phago-
cytes and plays a key role in immune defense [31]. Kupffer cells
highly express NOX and generate high amounts of reactive oxy-
gen species (ROS) in response to early liver injuries [32]. Upon
cellular stress, neutrophil cytosolic factor 1 (NCF1 or p47phox),
a key regulatory subunit of NOX, is phosphorylated and translo-
cate to the cell membrane to form the active NOX. To assess
the involvement of NADPH oxidase in experimental liver fibrosis,
we exposed p47phox
/
and wild-type control mice to PM
2.5
or
FA for 10 weeks. Upon PM
2.5
exposure, p47phox
/
mice exhib-
ited significantly attenuated liver fibrosis, compared to the
wild-type counterparts (Fig. 4A, B), suggesting that NOX plays a
major role in mediating the action of PM
2.5
exposure in hepatic
fibrogenesis. Consistent with the hepatic fibrosis staining result,
expression levels of collagen 1 mRNA in the liver tissues of the
p47phox
/
mice were dramatically decreased, compared to that
of the control mice, under PM
2.5
exposure (Fig. 4C).
Next, we examined production of ROS, an important trigger of
hepatic fibrosis [33], in the liver tissues of p47phox
/
and control
mice after PM
2.5
exposure. While inhalation exposure to PM
2.5
significantly induced ROS in the liver tissues of the wild-type
control mice, PM
2.5
-triggered ROS production was significantly
reduced in the p47phox
/
mice (Fig. 4D, E). Additionally, we
examined hepatic steatosis in the p47phox
/
and wild-type con-
trol mice after PM
2.5
exposure (Fig. 4F). Consistent with our early
finding, PM
2.5
exposure lead to hepatic lipid accumulation in the
wild-type mice. However, hepatic steatosis was relieved in the
PM
2.5
-exposed p47phox
/
mice, compared to that of wild-type
mice under PM
2.5
exposure (Fig. 4F), suggesting a critical role of
NOX in mediating PM
2.5
-induced hepatic steatosis. Furthermore,
we examined expression of PPAR
c
and TGFb1 in the livers of
p47phox
/
and control mice under PM
2.5
exposure. While
PM
2.5
exposure repressed expression of PPAR
c
mRNA in the
wild-type mice, expression of PPAR
c
in the p47phox
/
mouse liv-
ers was not suppressed by PM
2.5
exposure (Fig. 4G). Indeed, the
levels of PPAR
c
mRNA were even increased in the livers of
PM
2.5
-exposed p47phox
/
mice, compared to the PM
2.5
-exposed
wild-type mice or FA-exposed p47phox
/
mice. Moreover, PM
2.5
exposure was not able to increase expression of Tgfb1mRNA in
the p47phox
/
liver (Supplementary Fig. 7). Together, these
results suggested that NOX plays an important role in mediating
the action of PM
2.5
exposure in suppressing PPAR
c
and stimulat-
ing TGFb1 expression in the liver.
Discussion
In this study, we demonstrate that inhalation exposure to PM
2.5
represents a significant risk factor to hepatic fibrosis. Previously,
β-actin
PPARγ
Ctl 12 h 24 h
0
20
40
60
80
100
120
A
**
0.0
0.2
0.4
0.6
0.8
1.0
1.2
Veh
E
**
PPARγ
Tubulin
Vehicle
B
*
**
0
20
40
60
80
100
120
140
160
PPARγ levels in LX2 cells
(fold change %)
Veh
PPARγ protein in LX2
cells (fold change %)
PPARγ1mRNA in LX2 cells
(fold change)
0
1
2
3
4
5
6
7
8DMSO
1 μM Pio
2.5 μM Pio
**
0.0
0.5
1.0
1.5
2.0
2.5
3.0
3.5
4.0
4.5
5.0 DMSO
1 μM Pio
2.5 μM Pio
*
*
PPARγ1 mRNA levels in
LX2 cells (fold change)
PPAR
γ2 mRNA levels in
LX2 cells (fold change)
PBS PBS
12 h
24 h
PM 2.5
PM 2.5 PM 2.5
PM 2.5
PM 2.5
PM 2.5
PM 2.5
n.s.
n.s.
Veh
12 h
24 h
n.s.
n.s.
n.s. n.s.
DMSO
1 μM Pio
2.5 μM Pio
DMSO
1 μM Pio
2.5 μM Pio
CD
DMSO
1 μM Pio
2.5 μM Pio
Fig. 3. PM
2.5
exposure downregulates expression of PPAR
c
in HSC. (A)
Quantitative real-time PCR analysis of expression levels of PPAR
c
1mRNA in LX-
2 cells cultured in the conditioned medium from RAW264.7 cells exposed to PM
2.5
for 12 or 24 h. Fold changes of mRNA are shown by comparing to the vehicle
control. Each bar denotes the mean ± SEM (n = 3 biological replicates). (B)
Immunoblotting analysis of PPAR
c
levels in LX-2 cells cultured in the conditioned
media from RAW264.7 cells exposed to PM
2.5
for 12 or 24 h. LX-2 cells were
incubated with the conditioned medium for 36 h before subjected to immunoblot-
ting analysis. LX-2 cells were cultured in the conditioned medium from RAW264.7
cells exposed to PBS for 24 h as the control. The graph beside the images shows
fold changes of PPAR
c
protein levels in LX-2 cells. The fold changes of PPAR
c
in
PM
2.5
-exposed LX-2 cells were determined by normalizing to the PPAR
c
signal
intensities in PM
2.5
-exposed LX-2 cells to that of vehicle-exposed LX-2 cells. (C-E)
Pre-treatment of pioglitazone (Pio) prevents PM
2.5
-induced downregulation of
PPAR
c
in HSC. LX-2 cells were cultured in the presence of Pio at the concentration
of 1 or 2.5
l
M for 48 h before they were incubated with the conditioned medium
from RAW264.7 cell exposed to PM
2.5
(50
l
g/ml) for 14 h. LX-2 were incubated
with the conditioned medium for 36 h in the presence of Pio, and then subjected to
extraction of RNAs and proteins for qRT-PCR analyses of PPAR
c
1and PPAR
c
2mRNA
levels (C and D) and Western blot analysis of PPAR
c
protein levels (E). LX-2 cells
treated with vehicles (DMSO, the vehicle for Pio treatment; and PBS, the vehicle for
PM
2.5
). The fold changes of PPAR
c
mRNA levels in PM
2.5
- and/or Pio-treated cells
were determined by normalizing to PPAR
c
levels in vehicle-treated cells. Each bar
denotes the mean ± SEM (n = 3 biological replicates). The graph beside the images
(E) showed fold changes of PPAR
c
levels in LX-2 cells. The fold changes of PPAR
c
in
the Pio-treated LX2 cells were determined by comparing the normalized PPAR
c
signal intensities in the Pio-treated LX2 cells to that in the vehicle-treated LX2
cells (100%).
*
p60.05,
**
p60.01.
JOURNAL OF HEPATOLOGY
Journal of Hepatology 2015 vol. 63 j1397–1404 1401
we revealed important roles of ER stress, oxidative stress, and
inflammation in liver pathogenesis under PM
2.5
exposure [1,10].
Our works demonstrated that animals under PM
2.5
exposure dis-
played a NASH-like phenotype, reduction of hepatic glycogen
storage, and hepatic insulin resistance under normal chow diet
[1]. The current work extends our prior observations on the link
between air pollution exposure and liver pathogenesis. While
PM
2.5
exposure alone can cause mild but discernable hepatic
fibrosis, PM
2.5
can interact with another health risk factor, the
high-fat diet, to facilitate advanced stages of hepatic fibrosis.
Hepatic inflammation is a major contributing factor to the
development of liver fibrosis. Our study showed that inhalation
exposure to PM
2.5
induces TGFbproduction in mouse liver, pre-
sumably from Kupffer cells, which subsequently stimulates
SMAD3 signaling and collagen expression in HSCs (Figs. 1 and
2). As suggested by our previous studies, systemic inflammation
through circulating inflammatory cytokines, macrophage or neu-
trophil infiltration, and direct stimulation of Kupffer cells by
PM
2.5
particles delivered to the liver can all potentially promote
hepatic inflammation [1,10]. Therefore, systemic inflammation
and infiltrated leukocytes, in addition to Kupffer cells, may also
be involved in facilitating hepatic fibrosis through the
TGFb-SMAD3-collagen regulatory axis. Another important event
linking to hepatic fibrosis is the downregulation of PPAR
c
by
PM
2.5
exposure in HSC. In the liver, PPAR
c
plays important roles
in anti-inflammatory response and energy homeostasis
associated with HSC, Kupffer cells, and hepatocytes [34].In
particular, the differentiation of HSCs from Ito cells is inversely
correlated with PPAR
c
levels [24]. Therefore, the downregulation
of PPAR
c
in HSC could be important to the progression of hepatic
fibrogenesis in PM
2.5
-exposed mice (Fig. 3). Additionally, hepatic
steatosis developed under PM
2.5
exposure, partially due to
repression of PPAR
a
and PPAR
c
[1], may also contribute to the
progression of liver fibrosis. Based on these scenarios, delineating
which inflammatory pathways, either systemic or local, are
relatively important in promoting hepatic fibrogenesis, and
defining the extent to which hepatic inflammation or steatosis
contributes to liver fibrosis under PM
2.5
exposure are interesting
subjects to be investigated in the future.
Our study demonstrated that PM
2.5
-induced hepatic steatosis
relies on NOX activity. We previously reported a critical role of
NOX in mediating oxidative stress, as p47phox
/
animals
appeared to be protective from PM
2.5
-mediated insulin resistance
and inflammation [20,35]. Studies from other groups showed that
NOX is critically involved in hepatic fibrosis induced by angioten-
sin II-, bile duct ligation-, or methionine-choline-deficient diet
[36,37]. Our work with PM
2.5
exposure provides new evidence
that NOX is essential for hepatic fibrosis under real-world envi-
ronmental stress. NOX generates large amounts of ROS in phago-
cytic cells, and ROS is known to play an important role in hepatic
fibrosis [31,32]. Under PM
2.5
exposure, p47phox
/
mice dis-
played significantly reduced ROS production and hepatic fibrosis
FA
A
B
D
CG
WT
FA
*
0.0
0.5
1.0
1.5
2.0
2.5
3.0
3.5
0.0
0.4
0.8
1.2
1.6
2.0
Col1 mRNA levels
(fold changes)
PPARγ1 mRNA levels
(fold changes)
*
FA FA FA FA
FA
E
F
PM 2.5 PM 2.5 PM 2.5 PM 2.5
PM 2.5
PM 2.5 PM 2.5
*
*
p47phox -/- WT
p47phox -/- WT
p47phox -/-
WT p47phox-/- WT
p47phox-/- WT
p47phox-/- WT
p47phox-/-
FA FA PM 2.5 PM 2.5 FA FA PM 2.5 PM 2.5
0.0
0.5
1.0
1.5
2.0
0
50
100
150
DHE fluorescence
(arbitrary unit)
Fibrosis score
arbitrary units
Fig. 4. Hepatic fibrosis under PM
2.5
exposure relies on NOX activity. (A) Histological analysis of hepatic collagen deposition (Sirius-red staining) in formalin-fixed liver
tissue sections from p47phox
/
and wild-type control mice exposed to FA or PM
2.5
for 10 weeks. Magnifications: 200. The arrows point out areas of hepatic fibrosis. (B)
Hepatic fibrosis grades of the p47phox
/
and control mice exposed to PM
2.5
or FA. Hepatic fibrosis grades were determined based on the histological analyses of Sirius-red
staining of collagens, according to the Scheuer scoring system for fibrosis and cirrhosis [16,17]. Data are shown as mean ± SEM (n = 4 animals per group). (C) qRT-PCR
analysis of expression levels of Col1 mRNA in p47phox
/
and control mice exposed to FA or PM
2.5
for 10 weeks. Fold changes of mRNA are shown by normalizing mRNA
levels to that of the FA-exposed control animals. Each bar denotes the mean ± SEM (n = 3 animals per group). (D) DHE staining of ROS signals in the liver tissue sections from
the mice exposed to FA or PM
2.5
. The oxidative red fluorescence was detected by a Zeiss fluorescence microscope. Magnification : 400. (E) Quantification of DHE-stained
ROS signals in the PM
2.5
- and FA-exposed p47phox
/
and control mice. DHE signals were quantified by counting the number of positive stained nuclei in 8 random fields.
Microscopic interference contrast was used to exclude positive signals from non-cell origin. The percentages of DHE-positive nuclei (compared to total nuclei) were shown.
Data are shown as mean ± SEM (n = 4 animals per group).
*
p<0.05. (F) Oil-red O staining of hepatic lipid droplets in the livers of p47phox
/
and wild-type control mice
exposed to FA or PM
2.5
for 10 weeks (magnifications: 200). (G) qRT-PCR analysis of expression levels of PPAR
c
mRNA in the liver tissues of p47phox
/
and control mice
exposed to FA or PM
2.5
for 10 weeks. Fold changes of mRNA are shown by normalizing mRNA levels to that of the FA-exposed control animals. Each bar denotes the
mean ± SEM (n = 4 animals per group). For B, C, E, and G,
*
p<0.05. (This figure appears in colour on the web.)
Research Article
1402 Journal of Hepatology 2015 vol. 63 j1397–1404
(Fig. 4). Importantly, PM
2.5
-induced downregulation of PPAR
c
and upregulation of TGFb1 were attenuated in the livers of
p47phox
/
mice (Fig. 4G; Supplementary Fig. 7), suggesting that
NOX plays a major role in mediating PM
2.5
-triggered fibrogenesis
through modulating PPAR
c
and TGFbsignaling.
The significance of our study needs to be placed in the context
of real-world PM
2.5
exposure. The PM
2.5
concentration in the ani-
mal exposure chambers was approximately 10 times that of daily
ambient PM
2.5
concentration in most US and European cities. In
the developing countries, such as China, India, and Latin Ameri-
can, where the daily PM
2.5
levels range from 100 to 200
l
g/m
3
(approximately 10–20-fold higher than those in major U.S. cities)
[38], the detrimental effects of PM
2.5
exposure on public health
have been grossly underestimated. Even in the U.S. or European
countries, the prevalence of metabolic syndrome was increased
with increasing PM
2.5
concentrations, as evidenced by a 1%
increase in diabetes prevalence seen with a 10
l
g/m
3
increase
in PM
2.5
exposure [39]. Therefore, the PM
2.5
levels at high ranges,
as obtained through our experimental expose system, not only
recapitulate true air pollution environment in the developing
countries but also can be translated into cumulative exposures
in the U.S. or European countries. Our studies suggest that the
liver is an important target organ and a key player in pathophys-
iology under PM
2.5
exposure. This finding has important implica-
tions in the medical care from the prevention to treatment of
systemic diseases associated with air pollution.
Conflict of interest
The authors who have taken part in this study declared that they
do not have anything to disclose regarding funding or conflict of
interest with respect to this manuscript.
Authors’ contributions
Study concept and design: K.Z., Z.Z., Q.S.; acquisition of data,
analysis and interpretation of data: Z.Z., K.Z., Q.S., X.Z., J.W.,
A.D., H.K., Y.Q., A.W.; drafting of the manuscript: K.Z., Z.Z.; critical
revision of the manuscript for important intellectual content:
K.Z., Z.Z., Q.S.; statistical analysis: Z.Z., X.Z., J.W., A.D., X.X., Q.S.;
obtained funding: Z.Z., Q.S.; administrative, technical, or material
support: X.X., Y.C., A.W., L.C.C., S.R.; study supervision: K.Z., Q.S.
Acknowledgement
Portions of this work were supported by National Institutes of
Health (NIH) grants DK090313 and ES017829 to KZ, American
Heart Association Grants 0635423Z and 09GRNT2280479 to KZ,
NIH grant ES018900 to QS, and NIH grants R01ES019616,
R01ES017290, and R01ES015146 to SR. We thank Dr. Scott Fried-
man for kindly providing LX-2 cells and Dr. Todd Leff for provid-
ing pioglitazone.
Supplementary data
Supplementary data associated with this article can be found, in
the online version, at http://dx.doi.org/10.1016/j.jhep.2015.07.
020.
References
[1] Zheng Z, Xu X, Zhang X, Wang A, Zhang C, Huttemann M, et al. Exposure to
ambient particulate matter induces a NASH-like phenotype and impairs
hepatic glucose metabolism in an animal model. J Hepatol
2013;58:148–154.
[2] Liu C, Xu X, Bai Y, Wang TY, Rao X, Wang A, et al. Air pollution-mediated
susceptibility to inflammation and insulin resistance: influence of CCR2
pathways in mice. Environ Health Perspect 2014;122:17–26.
[3] Sun L, Liu C, Xu X, Ying Z, Maiseyeu A, Wang A, et al. Ambient fine particulate
matter and ozone exposures induce inflammation in epicardial and
perirenal adipose tissues in rats fed a high fructose diet. Part Fibre Toxicol
2013;10:43.
[4] Alfaro-Moreno E, Martinez L, Garcia-Cuellar C, Bonner JC, Murray JC, et al.
Biologic effects induced in vitro by PM10 from three different zones of
Mexico City. Environ Health Perspect 2002;110:715–720.
[5] Soukup JM, Becker S. Human alveolar macrophage responses to air pollution
particulates are associated with insoluble components of coarse mate-
rial, including particulate endotoxin. Toxicol Appl Pharmacol 2001;171:
20–26.
[6] Brook RD, Franklin B, Cascio W, Hong Y, Howard G, Lipsett M, et al. Air
pollution and cardiovascular disease: a statement for healthcare profession-
als from the Expert Panel on Population and Prevention Science of the
American Heart Association. Circulation 2004;109:2655–2671.
[7] Xu X, Liu C, Xu Z, Tzan K, Wang A, Rajagopalan S, et al. Altered adipocyte
progenitor population and adipose-related gene profile in adipose tissue by
long-term high-fat diet in mice. Life Sci 2012;90:1001–1009.
[8] Kampfrath T, Maiseyeu A, Ying Z, Shah Z, Deiuliis JA, Xu X, et al. Chronic fine
particulate matter exposure induces systemic vascular dysfunction via
NADPH oxidase and TLR4 pathways. Circ Res 2011;108:716–726.
[9] Wang G, Rajagopalan S, Sun Q, Zhang K. Real-world exposure of airborne
particulate matter triggers oxidative stress in an animal model. Int J Physiol
Pathophysiol Pharmacol 2010;2:64–68.
[10] Laing S, Wang G, Briazova T, Zhang C, Wang A, Zheng Z, et al. Airborne
particulate matter selectively activates endoplasmic reticulum stress
response in the lung and liver tissues. Am J Physiol Cell Physiol
2010;299:C736–C749.
[11] Mendez R, Zheng Z, Fan Z, Rajagopalan S, Sun Q, Zhang K. Exposure to fine
airborne particulate matter induces macrophage infiltration, unfolded
protein response, and lipid deposition in white adipose tissue. Am J Transl
Res 2013;5:224–234.
[12] Chen LC, Quan C, Hwang JS, Jin X, Li Q, Zhong M, et al. Atherosclerosis lesion
progression during inhalation exposure to environmental tobacco smoke: a
comparison to concentrated ambient air fine particles exposure. Inhal
Toxicol 2010;22:449–459.
[13] Tan HH, Fiel MI, Sun Q, Guo J, Gordon RE, Chen LC, et al. Kupffer cell
activation by ambient air particulate matter exposure may exacerbate non-
alcoholic fatty liver disease. J Immunotoxicol 2009;6:266–275.
[14] Maciejczyk P, Zhong M, Li Q, Xiong J, Nadziejko C, Chen LC. Effects of
subchronic exposures to concentrated ambient particles (CAPs) in mice. II.
The design of a CAPs exposure system for biometric telemetry monitoring.
Inhal Toxicol 2005;17:189–197.
[15] Zheng Z, Zhang C, Zhang K. Measurement of ER stress response and
inflammation in the mouse model of nonalcoholic fatty liver disease.
Methods Enzymol 2011;489:329–348.
[16] Brunt EM. Nonalcoholic steatohepatitis: definition and pathology. Semin
Liver Dis 2001;21:3–16.
[17] Kleiner DE, Brunt EM, Van Natta M, Behling C, Contos MJ, Cummings OW,
et al. Design and validation of a histological scoring system for nonalcoholic
fatty liver disease. Hepatology 2005;41:1313–1321.
[18] Ying Z, Yue P, Xu X, Zhong M, Sun Q, Mikolaj M, et al. Air pollution and
cardiac remodeling: a role for RhoA/Rho-kinase. Am J Physiol Heart Circ
Physiol 2009;296:H1540–H1550.
[19] Chen LC, Nadziejko C. Effects of subchronic exposures to concentrated
ambient particles (CAPs) in mice. V. CAPs exacerbate aortic plaque devel-
opment in hyperlipidemic mice. Inhal Toxicol 2005;17:217–224.
[20] Xu X, Yavar Z, Verdin M, Ying Z, Mihai G, Kampfrath T, et al. Effect of early
particulate air pollution exposure on obesity in mice: role of p47phox.
Arterioscler Thromb Vasc Biol 2010;30:2518–2527.
[21] Akpolat N, Yahsi S, Godekmerdan A, Yalniz M, Demirbag K. The value of
alpha-SMA in the evaluation of hepatic fibrosis severity in hepatitis B
infection and cirrhosis development: a histopathological and immunohis-
tochemical study. Histopathology 2005;47:276–280.
[22] Safadi R, Friedman SL. Hepatic fibrosis–role of hepatic stellate cell activation.
MedGenMed 2002;4:27.
JOURNAL OF HEPATOLOGY
Journal of Hepatology 2015 vol. 63 j1397–1404 1403
[23] Roth S, Gong W, Gressner AM. Expression of different isoforms of TGF-beta
and the latent TGF-beta binding protein (LTBP) by rat Kupffer cells. J Hepatol
1998;29:915–922.
[24] Miyahara T, Schrum L, Rippe R, Xiong S, Yee Jr HF, Motomura K, et al.
Peroxisome proliferator-activated receptors and hepatic stellate cell activa-
tion. J Biol Chem 2000;275:35715–35722.
[25] Hautekeete ML, Geerts A. The hepatic stellate (Ito) cell: its role in human
liver disease. Virchows Arch 1997;430:195–207.
[26] Ankoma-Sey V. Hepatic regeneration-revisiting the Myth of prometheus.
News Physiol Sci 1999;14:149–155.
[27] Sakata R, Ueno T, Nakamura T, Ueno H, Sata M. Mechanical stretch induces
TGF-beta synthesis in hepatic stellate cells. Eur J Clin Invest
2004;34:129–136.
[28] Xu L, Hui AY, Albanis E, Arthur MJ, O’Byrne SM, Blaner WS, et al. Human
hepatic stellate cell lines, LX-1 and LX-2: new tools for analysis of hepatic
fibrosis. Gut 2005;54:142–151.
[29] Flanders KC. Smad3 as a mediator of the fibrotic response. Int J Exp Pathol
2004;85:47–64.
[30] Gillies PS, Dunn CJ. Pioglitazone. Drugs 2000;60:333–343, Discussion 344–
335.
[31] Babior BM, Lambeth JD, Nauseef W. The neutrophil NADPH oxidase. Arch
Biochem Biophys 2002;397:342–344.
[32] Wheeler MD, Kono H, Yin M, Nakagami M, Uesugi T, Arteel GE, et al. The role
of Kupffer cell oxidant production in early ethanol-induced liver disease.
Free Radic Biol Med 2001;31:1544–1549.
[33] Parola M, Robino G. Oxidative stress-related molecules and liver fibrosis. J
Hepatol 2001;35:297–306.
[34] Feige JN, Gelman L, Michalik L, Desvergne B, Wahli W. From molecular
action to physiological outputs: peroxisome proliferator-activated receptors
are nuclear receptors at the crossroads of key cellular functions. Prog Lipid
Res 2006;45:120–159.
[35] Ying Z, Kampfrath T, Thurston G, Farrar B, Lippmann M, Wang A, et al.
Ambient particulates alter vascular function through induction of reactive
oxygen and nitrogen species. Toxicol Sci 2009;111:80–88.
[36] Bataller R, Schwabe RF, Choi YH, Yang L, Paik YH, Lindquist J, et al. NADPH
oxidase signal transduces angiotensin II in hepatic stellate cells and is
critical in hepatic fibrosis. J Clin Invest 2003;112:1383–1394.
[37] De Minicis S, Seki E, Paik YH, Osterreicher CH, Kodama Y, Kluwe J, et al. Role
and cellular source of nicotinamide adenine dinucleotide phosphate oxidase
in hepatic fibrosis. Hepatology 2010;52:1420–1430.
[38] Tao J, Gao J, Zhang L, Zhang R, Che H, Zhang Z, et al. PM2.5 pollution in a
megacity of southwest China: source apportionment and implication. Atmos
Chem Phys 2014;14:20.
[39] Pearson JF, Bachireddy C, Shyamprasad S, Goldfine AB, Brownstein JS.
Association between fine particulate matter and diabetes prevalence in the
U.S. Diabetes Care 2010;33:2196–2201.
Research Article
1404 Journal of Hepatology 2015 vol. 63 j1397–1404
... Growing evidence suggests that liver regeneration is governed by a complex regulatory network involving various molecules, such as growth factors, transcription factors, mRNA, miRNA, lncRNA, circRNA, DNA methylation, RNA methylation, and histone modi cations [4]. miRNAs do not function in isolation during liver regeneration but interact with multiple signaling pathways and gene regulatory networks. ...
... The Wnt/β-catenin signaling pathway plays a crucial role in initiating liver regeneration, with miRNAs regulating several key members of this pathway. Research indicates that miR-214 can inhibit liver regeneration by downregulating β-catenin expression [4], while miR-375 enhances the Wnt/β-catenin signaling pathway by regulating Frizzled-8, thereby promoting hepatocyte proliferation [15]. ...
Preprint
Full-text available
Background: Hepatocellular carcinoma (HCC) is one of the most common and aggressive malignant tumors. Partial hepatectomy (PHx) is currently the primary treatment for HCC, but many patients suffer from poor liver reserve function and insufficient remaining liver volume, limiting the liver's regenerative capacity. Therefore, this study aims to explore the mechanisms of miRNA and mRNA in liver regeneration through high-throughput sequencing. Methods: A rat model of 70% hepatectomy was used, and physiological indicators related to liver regeneration were assessed on days 3, 7, and 14 post-surgery. Small RNA sequencing and transcriptome analysis were conducted to evaluate the miRNA and mRNA expression profiles at different stages of regeneration. Bioinformatics tools were used to identify differentially expressed genes, construct miRNA-mRNA regulatory networks, and protein-protein interaction (PPI) networks, to identify key regulatory molecules. Results: The rat liver regeneration model was successfully established, and the body weight and liver regeneration rate data on days 3, 7, and 14 indicated a smooth regeneration process. Small RNA sequencing and transcriptome analysis identified 395 known miRNAs and 299 precursor miRNAs. Differential expression analysis revealed dynamic expression patterns of multiple miRNAs and mRNAs during liver regeneration. The miRNA-mRNA regulatory network showed interactions between 17 differentially expressed miRNAs and 31 differentially expressed mRNAs involved in liver regeneration. Conclusion: This study, through small RNA sequencing and transcriptome analysis, revealed key regulatory roles of miRNAs in various signaling pathways during liver regeneration. The constructed miRNA-mRNA regulatory network further elucidates the molecular mechanisms of liver regeneration. The results demonstrate the complex regulatory roles of miRNAs in promoting hepatocyte proliferation, inhibiting apoptosis, and regulating multiple key signaling pathways, providing new insights into the understanding of liver regeneration mechanisms.
... A comprehensive cross-sectional analysis involving 90,086 participants in southwest China demonstrated a positive association between prolonged exposure to environmental particulate matter and metabolically associated fatty liver disease. Additionally, animal studies have identified a steatohepatitis-like phenotype and liver fibrosis associated with such exposures, reinforcing the broader implications of particulate pollutants on liver health (20,21). According to a cross-sectional study conducted in the United States, hospitalized patients with higher ambient PM 2.5 exposure were more likely to develop non-alcoholic fatty liver disease (22). ...
Article
Full-text available
Objectives Increasing concern about air pollution’s impact on public health underscores the need to understand its effects on non-neoplastic digestive system diseases (NNDSD). This study explores the link between air pollution and NNDSD in China. Methods We conducted a national cross-sectional study using 2015 data from the China Health and Retirement Longitudinal Study (CHARLS), involving 13,046 Chinese adults aged 45 and above from 28 provinces. Satellite-based spatiotemporal models estimated participants’ exposure to ambient particulate matter (3-year average). An analysis of logistic regression models was conducted to estimate the association between air pollutants [particulate matter with a diameter ≤ 2.5 μm (PM2.5) or ≤10 μm (PM10), sulfur dioxide (SO2), nitrogen dioxide (NO2), ozone (O3), and carbon monoxide (CO)] and NNDSD. Interaction analyses were conducted to examine potential modifiers of these associations. Results The prevalence of NNDSD among participants was 26.29%. After adjusted for multivariate factors, we observed a 6% [odd ratio (OR) = 1.06, 95% confidence interval (CI): 0.94, 1.19], 23% (OR = 1.23, 95% CI: 1.09, 1.38), 26% (OR = 1.26, 95% CI: 1.12, 1.41), 30% (OR = 1.30, 95% CI: 1.16, 1.46), 13% (OR = 1.13, 95% CI: 1.01, 1.27) and 27% (OR = 1.27, 95% CI: 1.13, 1.43) increase in NNDSD risk with an interquartile range increase in PM2.5 (23.36 μg/m³), PM10 (50.33 μg/m³), SO2 (17.27 μg/m³), NO2 (14.75 μg/m³), O3 (10.80 μg/m³), and CO (0.42 mg/m³), respectively. Interaction analyses showed that PM2.5, SO2, and O3 had stronger effects on NNDSD risk among older adults, highly educated individuals, smokers, and married people, respectively. Conclusion This study demonstrates that long-term exposure to PM2.5, PM10, SO2, NO2, O3, and CO is positively associated with NNDSD risk in Chinese adults aged 45 and above. Implementing intervention strategies to enhance air quality is essential for reducing the burden of NNDSD.
... The link between PM2.5 exposure and the incidence of liver disease was studied in different meta-analysis [20,21]. Additionally, PM2.5 exposure would not only damage the liver but also lead to lipid metabolic abnormalities reported by some toxicological studies [22,23]. The odd ratio of developing hepatic lipid metabolic disease increases by 1.3% with the increase in 10 μg/m 3 rise in PM2.5 pollutant level [24]. ...
Article
Full-text available
Fine particulate matter (PM2.5) pollution’s harmful effects on the environment and human health have gained global attention. After entering the liver via the bloodstream, these small particles could disrupt hepatic lipid metabolism and even cause liver damage, but the underlying mechanisms remain unclear. To investigate the impact of PM2.5 on the liver and metabolism, ApoE⁻/⁻ mice were used and then exposed to PM2.5 to establish a hyperlipidemic model. Our findings revealed the involvement of oxidative stress in this model, which was accompanied by elevated expression of inflammatory markers and enhanced GSDMD-mediated pyroptosis activation. Our results further show that PM2.5 exposure causes significant alterations in key metabolic hepatic proteins. Inhibiting pyroptosis with disulfiram (DSF) balances hepatic lipid-protein levels and decreases hepatic injury, suggesting its potential to protect the liver from PM2.5 exposure. N-acetyl-L-cysteine (NAC) treatment reversed PM2.5-induced oxidative stress, inhibiting pyroptosis and improving hepatocyte function, as demonstrated by the restored balance of hepatic proteins. Our investigation establishes a critical link between PM2.5 exposure and liver dysfunction through oxidative stress and pyroptosis pathways while demonstrating the therapeutic potential of DSF and NAC in protecting against PM2.5-induced metabolic disorders and hepatic injury. These insights provide a foundation for developing targeted interventions to address air pollution-related liver diseases, especially in vulnerable high-lipid populations.
Article
Full-text available
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent chronic liver condition marked by excessive lipid accumulation in hepatic tissue. This disorder can lead to a range of pathological outcomes, including metabolic dysfunction-associated steatohepatitis (MASH) and cirrhosis. Despite extensive research, the molecular mechanisms driving MASLD initiation and progression remain incompletely understood. Oxidative stress and lipid peroxidation are pivotal in the “multiple parallel hit model”, contributing to hepatic cell death and tissue damage. Gut microbiota plays a substantial role in modulating hepatic oxidative stress through multiple pathways: impairing the intestinal barrier, which results in bacterial translocation and chronic hepatic inflammation; modifying bile acid structure, which impacts signaling cascades involved in lipidic metabolism; influencing hepatocytes’ ferroptosis, a form of programmed cell death; regulating trimethylamine N-oxide (TMAO) metabolism; and activating platelet function, both recently identified as pathogenetic factors in MASH progression. Moreover, various exogenous factors impact gut microbiota and its involvement in MASLD-related oxidative stress, such as air pollution, physical activity, cigarette smoke, alcohol, and dietary patterns. This manuscript aims to provide a state-of-the-art overview focused on the intricate interplay between gut microbiota, lipid peroxidation, and MASLD pathogenesis, offering insights into potential strategies to prevent disease progression and its associated complications.
Article
Inhalation exposure to airborne fine particulate matter (aerodynamic diameter < 2.5 µm, PM 2.5 ) is known to cause metabolic dysfunction-associated steatohepatitis (MASH) and the associated metabolic syndrome. Hepatic lipid accumulation and inflammation are the key characteristics of MASH. However, the mechanism by which PM 2.5 exposure induces lipid accumulation and inflammation in the liver remains to be further elucidated. In this study, we revealed that inhalation exposure to PM 2.5 induces nitrosative stress in mouse livers by suppressing hepatic S-nitrosoglutathione reductase (GSNOR) activities, which leads to S-nitrosylation modification of the primary unfolded protein response (UPR) transducer IRE1α, an endoplasmic reticulum (ER)-resident protein kinase and endoribonuclease (RNase). S-nitrosylation suppresses RNase activity of IRE1α and subsequently decreases IRE1α-mediated splicing of the mRNA encoding X-box binding protein 1 (Xbp1) and IRE1α-dependent degradation of select microRNAs (miRNAs), including miR-200 family, miR-34, miR-223, miR-155, and miR-146, in the livers of the mice exposed to PM 2.5 . Elevation of IRE1α-target miRNAs, due to impaired IRE1α RNase activity by PM 2.5 -triggered S-nitrosylation, leads to decreased expression of the major regulators of fatty acid oxidation, lipolysis and anti-inflammatory response, including XBP1, SIRT1, PPARα, and PPARγ, in the liver, which account at least partially for hepatic lipid accumulation and inflammation in mice exposed to airborne PM 2.5 . In summary, our study revealed a novel pathway by which PM 2.5 causes cytotoxicity and promotes MASH-like phenotypes through inducing hepatic nitrosative stress and S-nitrosylation of the primary UPR transducer and subsequent elevation of select miRNAs involved in metabolism and inflammation in the liver.
Article
Background and Aims To investigate the association between air pollution and hepatocellular carcinoma (HCC) in chronic hepatitis B (CHB) patients treated with nucleotide/nucleoside analogues. Methods We enrolled 1298 CHB patients treated with nucleotide/nucleoside analogues and analysed the incidence and risk factors for HCC. Daily estimates of air pollutants were estimated since the previous year from the enrolment date. Results The annual incidence of HCC was 2.1/100 person‐years after a follow‐up period of over 4840.5 person‐years. Factors with the strongest association with HCC development were liver cirrhosis (hazard ratio [HR]/95% confidence interval [CI]: 3.00/1.55–5.81; p = 0.001), male sex (2.98/1.51–5.90; p = 0.02), body mass index (1.11/1.04–1.18; p = 0.002) and age (1.06/1.04–1.09; p < 0.001). Among patients with cirrhosis, the factors associated with HCC development were male sex (HR/95% CI: 2.10/1.00–4.25; p = 0.04) and NO 2 (per one‐unit increment, parts per billion; 1.07/1.01–1.13; p = 0.01). Moreover, patients with the highest quartile of annual NO 2 exposure had more than a three‐fold risk of HCC than those with the lowest quartile of annual exposure (HR/95% CI: 3.26/1.34–7.93; p = 0.01). Among patients without cirrhosis, the strongest factors associated with HCC development were male sex (HR/95% CI: 5.86/1.79–19.23; p = 0.004), age (1.12/1.07–1.17; p < 0.001) and platelet count (0.99/0.98–1.00; p = 0.04). Conclusions Air pollution influences HCC development in CHB patients who receive nucleotide/nucleoside analogue therapy. Long‐term NO 2 exposure might accelerate HCC development in CHB patients with cirrhosis receiving nucleotide/nucleoside analogue treatment.
Article
The association between the stress defense system and exposure to fine particulate matter (PM 2.5 ) is a hot topic in the field of environmental health. PM 2.5 pollution is an increasingly serious issue, and its impact on health cannot be ignored. The stress defense system is an important biological mechanism for maintaining cell and internal environment homeostasis, playing a crucial role in PM 2.5 ‐induced damage and diseases. The association between PM 2.5 exposure and activation of the stress defense system has been reported. Moderate PM 2.5 exposure rapidly mobilizes the stress defense system, while excessive PM 2.5 exposure may exceed its compensatory and coping abilities, resulting in system imbalance and dysfunction that triggers pathological changes in cells and tissues, thereby increasing the risk of chronic diseases, such as respiratory diseases, cardiovascular diseases, and cancer. This detailed review focuses on the composition, function, and regulatory mechanisms of the antioxidant defense system, autophagy system, ubiquitin–proteasome system, and inflammatory response system, which are all components of the stress defiance system. In particular, the influence of PM 2.5 exposure on each of these defense systems and their roles in responding to PM 2.5 ‐induced damage was investigated to provide an in‐depth understanding of the pathogenesis of PM 2.5 exposure, accurately assess potential hazards, and formulate prevention and intervention strategies for health damage caused by PM 2.5 exposure.
Article
Full-text available
Daily PM2.5 (aerosol particles with an aerodynamic diameter of less than 2.5 μm) samples were collected at an urban site in Chengdu, an inland megacity in southwest China, during four 1-month periods in 2011, with each period in a different season. Samples were subject to chemical analysis for various chemical components ranging from major water-soluble ions, organic carbon (OC), element carbon (EC), trace elements to biomass burning tracers, anhydrosugar levoglucosan (LG), and mannosan (MN). Two models, the ISORROPIA II thermodynamic equilibrium model and the positive matrix factorization (PMF) model, were applied to explore the likely chemical forms of ionic constituents and to apportion sources for PM2.5. Distinctive seasonal patterns of PM2.5 and associated main chemical components were identified and could be explained by varying emission sources and meteorological conditions. PM2.5 showed a typical seasonality of waxing in winter and waning in summer, with an annual mean of 119 μg m−3. Mineral soil concentrations increased in spring, whereas biomass burning species elevated in autumn and winter. Six major source factors were identified to have contributed to PM2.5 using the PMF model. These were secondary inorganic aerosols, coal combustion, biomass burning, iron and steel manufacturing, Mo-related industries, and soil dust, and they contributed 37 ± 18, 20 ± 12, 11 ± 10, 11 ± 9, 11 ± 9, and 10 ± 12%, respectively, to PM2.5 masses on annual average, while exhibiting large seasonal variability. On annual average, the unknown emission sources that were not identified by the PMF model contributed 1 ± 11% to the measured PM2.5 mass. Various chemical tracers were used for validating PMF performance. Antimony (Sb) was suggested to be a suitable tracer of coal combustion in Chengdu. Results of LG and MN helped constrain the biomass burning sources, with wood burning dominating in winter and agricultural waste burning dominating in autumn. Excessive Fe (Ex-Fe), defined as the excessive portion in measured Fe that cannot be sustained by mineral dust, is corroborated to be a straightforward useful tracer of iron and steel manufacturing pollution. In Chengdu, Mo / Ni mass ratios were persistently higher than unity, and considerably distinct from those usually observed in ambient airs. V / Ni ratios averaged only 0.7. Results revealed that heavy oil fuel combustion should not be a vital anthropogenic source, and additional anthropogenic sources for Mo are yet to be identified. Overall, the emission sources identified in Chengdu could be dominated by local sources located in the vicinity of Sichuan, a result different from those found in Beijing and Shanghai, wherein cross-boundary transport is significant in contributing pronounced PM2.5. These results provided implications for PM2.5 control strategies.
Article
Full-text available
Epidemiologic and experimental studies support an association between fine ambient particulate matter < 2.5 µm (PM2.5) exposure and insulin resistance (IR). A role for innate immune cell activation has been suggested in the pathogenesis of these effects. To evaluate the role of CC-chemokine receptor 2 (CCR2) in PM2.5-mediated inflammation and IR. Wild-type C57BL/6 and CCR2(-/-) male mice were fed a high-fat diet and assigned to concentrated ambient PM2.5 or filtered air for 17 weeks via a whole body exposure system. Glucose tolerance and insulin sensitivity were evaluated. At euthanasia, blood, spleen and visceral adipose tissue (VAT) were collected to measure inflammatory cells using flow cytometry. Standard immunoblots, immnunohistochemical methods and quantitative PCR were used to assess pathways of interest involving insulin signaling, inflammation, lipid and glucose metabolism in various organs. Vascular function was assessed with myography. PM2.5 exposure resulted in whole body IR and increased hepatic lipid accumulation in the liver which was attenuated in CCR2(-/-) mice by inhibiting SREBP1c mediated transcriptional programming, decreasing fatty acid uptake and suppressing p38 MAPK activity. CCR2(-/-) restored abnormal phosphorylation levels of AKT, AMPK in VAT and adipose tissue macrophage content. However, the impaired whole body glucose tolerance and reduced GLUT-4 in skeletal muscle in response to PM2.5 was not corrected by CCR2 deficiency. PM2.5 mediates IR by regulating VAT inflammation, hepatic lipid metabolism and glucose utilization in skeletal muscle via both CCR2-dependent and independent pathways. These findings provide new mechanistic links between air pollution and metabolic abnormalities underlying IR.
Article
Full-text available
Inflammation and oxidative stress play critical roles in the pathogenesis of inhaled air pollutant-mediated metabolic disease. Inflammation in the adipose tissues niches are widely believed to exert important effects on organ dysfunction. Recent data from both human and animal models suggest a role for inflammation and oxidative stress in epicardial adipose tissue (EAT) as a risk factor for the development of cardiovascular disease. We hypothesized that inhalational exposure to concentrated ambient fine particulates (CAPs) and ozone (O3) exaggerates inflammation and oxidative stress in EAT and perirenal adipose tissue (PAT). Eight- week-old Male Sprague--Dawley rats were fed a normal diet (ND) or high fructose diet (HFr) for 8 weeks, and then exposed to ambient AIR, CAPs at a mean of 356 mug/m3, O3 at 0.485 ppm, or CAPs (441 mug/m3) + O3 (0.497 ppm) in Dearborn, MI, 8 hours/day, 5 days/week, for 9 days over 2 weeks. EAT and PAT showed whitish color in gross, and less mitochondria, higher mRNA expression of white adipose specific and lower brown adipose specific genes than in brown adipose tissues. Exposure to CAPs and O3 resulted in the increase of macrophage infiltration in both EAT and PAT of HFr groups. Proinflammatory genes of Tnf-alpha, Mcp-1 and leptin were significantly upregulated while IL-10 and adiponectin, known as antiinflammatory genes, were reduced after the exposures. CAPs and O3 exposures also induced an increase in inducible nitric oxide synthase (iNOS) protein expression, and decrease in mitochondrial area in EAT and PAT. We also found significant increases in macrophages of HFr-O3 rats. The synergetic interaction of HFr and dirty air exposure on the inflammation was found in most of the experiments. Surprisingly, exposure to CAPs or O3 induced more significant inflammation and oxidative stress than co-exposure of CAPs and O3 in EAT and PAT. EAT and PAT are both white adipose tissues. Short-term exposure to CAPs and O3, especially with high fructose diet, induced inflammation and oxidative stress in EAT and PAT in rats. These findings may provide a link between air-pollution exposure and accelerated susceptibility to cardiovascular disease and metabolic complications.
Article
Full-text available
Recent epidemiological studies have suggested a link between exposure to ambient air-pollution and susceptibility to metabolic disorders such as Type II diabetes mellitus. Previously, we provided evidence that both short- and long-term exposure to concentrated ambient particulate matter with aerodynamic diameter <2.5 μm (PM2.5) induces multiple abnormalities associated with the pathogenesis of Type II diabetes mellitus, including insulin resistance, visceral adipose inflammation, brown adipose mitochondrial adipose changes, and hepatic endoplasmic reticulum (ER) stress. In this report, we show that chronic inhalation exposure to PM2.5 (10 months exposure) induces macrophage infiltration and Unfolded Protein Response (UPR), an intracellular stress signaling that regulates cell metabolism and survival, in mouse white adipose tissue in vivo. Gene expression studies suggested that PM2.5 exposure induces two distinct UPR signaling pathways mediated through the UPR transducer inositol-requiring 1α (IRE1α): 1) ER-associated Degradation (ERAD) of unfolded or misfolded proteins, and 2) Regulated IRE1-dependent Decay (RIDD) of mRNAs. Along with the induction of the UPR pathways and macrophage infiltration, expression of genes involved in lipogenesis, adipocyte differentiation, and lipid droplet formation was increased in the adipose tissue of the mice exposed to PM2.5. In vitro study confirmed that PM2.5 can trigger phosphorylation of the UPR transducer IRE1α and activation of macrophages. These results provide novel insights into PM2.5-triggered cell stress response in adipose tissue and increase our understanding of pathophysiological effects of particulate air pollution on the development of metabolic disorders.
Article
Full-text available
Background & aims: Air pollution is a global challenge to public health. Epidemiological studies have linked exposure to ambient particulate matter with aerodynamic diameters<2.5 μm (PM(2.5)) to the development of metabolic diseases. In this study, we investigated the effect of PM(2.5) exposure on liver pathogenesis and the mechanism by which ambient PM(2.5) modulates hepatic pathways and glucose homeostasis. Methods: Using "Ohio's Air Pollution Exposure System for the Interrogation of Systemic Effects (OASIS)-1", we performed whole-body exposure of mice to concentrated ambient PM(2.5) for 3 or 10 weeks. Histological analyses, metabolic studies, as well as gene expression and molecular signal transduction analyses were performed to determine the effects and mechanisms by which PM(2.5) exposure promotes liver pathogenesis. Results: Mice exposed to PM(2.5) for 10 weeks developed a non-alcoholic steatohepatitis (NASH)-like phenotype, characterized by hepatic steatosis, inflammation, and fibrosis. After PM(2.5) exposure, mice displayed impaired hepatic glycogen storage, glucose intolerance, and insulin resistance. Further investigation revealed that exposure to PM(2.5) led to activation of inflammatory response pathways mediated through c-Jun N-terminal kinase (JNK), nuclear factor kappa B (NF-κB), and Toll-like receptor 4 (TLR4), but suppression of the insulin receptor substrate 1 (IRS1)-mediated signaling. Moreover, PM(2.5) exposure repressed expression of the peroxisome proliferator-activated receptor (PPAR)γ and PPARα in the liver. Conclusions: Our study suggests that PM(2.5) exposure represents a significant "hit" that triggers a NASH-like phenotype and impairs hepatic glucose metabolism. The information from this work has important implications in our understanding of air pollution-associated metabolic disorders.
Article
Pioglitazone is an orally administered insulin sensitising thiazolidinedione agent that has been developed for the treatment of type 2 diabetes mellitus. ▴ Pioglitazone activates the nuclear peroxisome proliferator activated receptor-γ (PPAR-γ), which leads to the increased transcription of various proteins regulating glucose and lipid metabolism. These proteins amplify the post-receptor actions of insulin in the liver and peripheral tissues, which leads to improved glycaemic control with no increase in the endogenous secretion of insulin. ▴ In placebo-controlled clinical trials, monotherapy with pioglitazone 15 to 45 mg/day has been shown to decrease blood glycosylated haemoglobin (HbA1c) levels in patients with type 2 diabetes mellitus. ▴ The addition of pioglitazone 30 mg/day to preexisting therapy with metformin, or of pioglitazone 15 or 30 mg/day to sulphonylurea, insulin or voglibose therapy, has been shown to decrease HbA1c and fasting blood glucose levels significantly in patients with poorly controlled type 2 diabetes mellitus. ▴ Pioglitazone has also been associated with improvements in serum lipid profiles in randomised placebo-controlled clinical studies. ▴ The drug has been well tolerated by adult patients of all ages in clinical studies. Oedema has been reported with monotherapy, and pooled data have shown hypoglycaemia in 2 to 15% of patients after the addition of pioglitazone to sulphonylurea or insulin treatment. There have been no reports of hepatotoxicity.
Article
Epidemiological studies have shown a strong link between air pollution and the increase of cardio-pulmonary mortality and morbidity. In particular, inhaled airborne particulate matter (PM) exposure is closely associated with the pathogenesis of air pollution-induced systemic diseases. In this study, we exposed C57BIV6 mice to environmentally relevant PM in fine and ultra fine ranges (diameter < 2.5 μm, PM(2.5)) using a "real-world" airborne PM exposure system. We investigated the pathophysiologic impact of PM(2.5) exposure in the animal model and in cultured primary pulmonary macrophages. We demonstrated that PM(2.5) exposure increased the production of reactive oxygen species (ROS) in blood vessels in vivo. Furthermore, in vitro PM(2.5) exposure experiment suggested that PM(2.5) could trigger oxidative stress response, reflected by an increased expression of the anti-oxidative stress enzymes superoxide dismutase-1 (SOD-1) and heme oxygenase-1(HO-1), in mouse primary macrophages. Together, the results obtained through our "real-world" PM exposure approach demonstrated the pathophysiologic effect of ambient PM(2.5) exposure on triggering oxidative stress in the specialized organ and cell type of an animal model. Our results and approach will be informative for the research in air pollution-associated physiology and pathology.
Article
Chronic exposure to ambient air-borne particulate matter of < 2.5 μm (PM₂.₅) increases cardiovascular risk. The mechanisms by which inhaled ambient particles are sensed and how these effects are systemically transduced remain elusive. To investigate the molecular mechanisms by which PM₂.₅ mediates inflammatory responses in a mouse model of chronic exposure. Here, we show that chronic exposure to ambient PM₂.₅ promotes Ly6C(high) inflammatory monocyte egress from bone-marrow and mediates their entry into tissue niches where they generate reactive oxygen species via NADPH oxidase. Toll-like receptor (TLR)4 and Nox2 (gp91(phox)) deficiency prevented monocyte NADPH oxidase activation in response to PM₂.₅ and was associated with restoration of systemic vascular dysfunction. TLR4 activation appeared to be a prerequisite for NAPDH oxidase activation as evidenced by reduced p47(phox) phosphorylation in TLR4 deficient animals. PM₂.₅ exposure markedly increased oxidized phospholipid derivatives of 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphorylcholine (oxPAPC) in bronchioalveolar lavage fluid. Correspondingly, exposure of bone marrow-derived macrophages to oxPAPC but not PAPC recapitulated effects of chronic PM₂.₅ exposure, whereas TLR4 deficiency attenuated this response. Taken together, our findings suggest that PM₂.₅ triggers an increase in oxidized phospholipids in lungs that then mediates a systemic cellular inflammatory response through TLR4/NADPH oxidase-dependent mechanisms.