Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

This article was downloaded by: [Australian Catholic University]

On: 21 August 2011, At: 13:55


Publisher: Taylor & Francis
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered
office: Mortimer House, 37-41 Mortimer Street, London W1T 3JH, UK

Toxicological & Environmental


Chemistry
Publication details, including instructions for authors and
subscription information:
http://www.tandfonline.com/loi/gtec20

Dioxin alters inflammatory responses to


lipopolysaccharide
a a a
Jesus Olivero-Verbel , Robert A. Roth & Patricia E. Ganey
a
Department of Pharmacology and Toxicology, Center for
Integrative Toxicology, Michigan State University, East Lansing, MI
48824, USA

Available online: 11 May 2011

To cite this article: Jesus Olivero-Verbel, Robert A. Roth & Patricia E. Ganey (2011): Dioxin alters
inflammatory responses to lipopolysaccharide, Toxicological & Environmental Chemistry, 93:6,
1180-1194

To link to this article: http://dx.doi.org/10.1080/02772248.2011.577554

PLEASE SCROLL DOWN FOR ARTICLE

Full terms and conditions of use: http://www.tandfonline.com/page/terms-and-


conditions

This article may be used for research, teaching and private study purposes. Any
substantial or systematic reproduction, re-distribution, re-selling, loan, sub-licensing,
systematic supply or distribution in any form to anyone is expressly forbidden.
The publisher does not give any warranty express or implied or make any representation
that the contents will be complete or accurate or up to date. The accuracy of any
instructions, formulae and drug doses should be independently verified with primary
sources. The publisher shall not be liable for any loss, actions, claims, proceedings,
demand or costs or damages whatsoever or howsoever caused arising directly or
indirectly in connection with or arising out of the use of this material.
Toxicological & Environmental Chemistry
Vol. 93, No. 6, July 2011, 1180–1194

Dioxin alters inflammatory responses to lipopolysaccharide


Jesus Olivero-Verbel, Robert A. Roth and Patricia E. Ganey*

Department of Pharmacology and Toxicology, Center for Integrative Toxicology, Michigan State
University, East Lansing, MI 48824, USA
(Received 25 November 2010; final version received 30 March 2011)
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has well characterized effects on


specific immune responses, but the effects on the innate immune system are less
understood. The effect of TCDD on inflammatory responses induced by
lipopolysaccharide (LPS) was evaluated in C57BL/6J female mice. Mice were
treated with 30 mg kg1 TCDD or vehicle once, p.o., and 4 days later, animals
received LPS (0.05  107 EU kg1, i.p.) or vehicle. Inflammatory mediators and
the liver injury marker, alanine aminotransferase (ALT), were measured, and liver
histology was evaluated. TCDD-treated animals had higher plasma ALT activity
than vehicle-treated animals, but the effect was mild and time-dependent. Few
changes in liver histopathology were observed, mainly represented in greater
steatosis in TCDD/LPS-treated mice compared to mice treated with LPS or
TCDD alone. LPS produced a time-dependent increase in the plasma concen-
trations of interleukins (IL)-6, -10, and -12 and interferon (IFN)- , tumor
necrosis factor (TNF)- , and monocyte chemoattractant protein (MCP)-1. With
the exception of IL-12, concentrations of each of these mediators were higher in
plasma of mice co-treated with TCDD and LPS compared to either agent alone.
The dose–response curve for the concentration of IL-6 in plasma suggested that
dioxin increased the potency of LPS to cause the release of this cytokine but not
the maximal response. Co-treatment with TCDD and LPS also led to greater
expression of mRNA for IL-10 and IFN- compared to either TCDD or LPS
alone. These results suggest that TCDD changes the inflammatory cytokine
profile induced by LPS and that LPS enhances the hepatic steatotic response
to TCDD.
Keywords: inflammation; TCDD; endotoxin; cytokines

Introduction
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is one of the most potent environmental
pollutants. TCDD may be found as a contaminant in the ecosystem as a byproduct of
waste incineration, combustion, and industrial processes, among other sources (Kulkarni,
Crespo, and Afonso 2008; Xu et al. 2009). In addition, the excessive production of
electronic waste has recently increased awareness about TCDD-like compounds not only
for the environment but also for potential human exposure (Chan et al. 2007; Chatterjee
2007). The primary adverse effects due to TCDD identified in humans include chloracne
(Pape and Stahlmann 2007), central neurotoxicity, and hepatotoxicity (Pelclová et al.
2006). Most of these effects depend on the activation of the aryl hydrocarbon receptor

*Corresponding author. Email: ganey@msu.edu

ISSN 0277–2248 print/ISSN 1029–0486 online


ß 2011 Taylor & Francis
DOI: 10.1080/02772248.2011.577554
http://www.informaworld.com
Toxicological & Environmental Chemistry 1181

(AhR), which acts as a transcription factor. TCDD binds to AhR, activating signal
transduction networks that trigger changes in the expression of target genes involved in
cell stress responses (Matsumura and Vogel 2006), matrix metabolism (Hillegass et al.
2006), lipid and glucose metabolism (Sato et al. 2008), growth regulation (Patel et al.
2006), and reproduction (Miyamoto 2004), among many other responses.
There is considerable evidence of crosstalk between TCDD-activated signaling
networks and those elicited by chemical stressors or pathogens (Khan et al. 2007; Neff-
LaFord et al. 2007). The findings with pathogens support the notion that processes evoked
by infection may be impacted by TCDD exposure (Ilbäck and Friman 2007). During
infection, inflammation is a common response to pathogens. For example, the cell walls of
Gram-negative bacteria contain lipopolysaccharide (LPS), a macromolecule that induces
inflammation by modulating signaling pathways that lead to activation of the innate
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

immune system, including changes in cytokine expression, neutrophil infiltration into


tissues, and other biochemical responses (Mayeux 1997; Yamasawa, Ishii, and Kitamura
1999; Allcock et al. 2001; Rose, Banerjee and Ramaiah 2007). Both virus- and bacteria-
induced inflammatory processes were reported to be affected by TCDD (Sugita-Konishi
et al. 2003; Teske et al. 2008). In addition, inflammation resulting from xenobiotic
exposure, such as that observed in a mouse model of atopic dermatitis (Ito et al. 2008),
may also be targeted by this pollutant. Interestingly, biochemical changes that were
observed in the liver after exposure to TCDD (e.g., lipid accumulation) were also found
after treatment with LPS (Ohhira et al. 2007; Sato et al. 2008), suggesting that these agents
might share cellular effectors. Accordingly, the objective of this study was to explore the
interactions between LPS-induced inflammation and TCDD exposure.

Materials and methods


Animals
Female, C57BL/6J mice, 9–11 weeks old, weighing between 19 and 21 g, were housed, five
animals per cage, at 24 C  2 C, 50%  10% relative humidity, and a 12-h light/dark
cycle. Mice were acclimated for at least 1 week before use. Animals were fed standard
chow (Rodent chow/Tek 8640, Harlan Teklad, Madison, WI) and spring water ad libitum.
All experiments were performed in accordance with the Guide for the Care and Use of
Laboratory Animals as adopted by the United States National Institutes of Health.

Experimental protocol
Mice were treated with sesame oil vehicle or 30 mg kg1 TCDD (499%, purchased from
Supelco, Bellefonte, PA) by oral gavage (p.o.). Four days later, saline or LPS (0.01–
3.2 [  107] endotoxin units (EU)/kg, lot 024K4067, derived from Escherichia coli serotype
055:B5, Sigma Aldrich, St Louis, MO) was administered by i.p. injection. These doses were
selected based upon results of preliminary experiments and on published data (North et al.
2009). At various times afterwards, animals were anesthetized with sodium pentobarbital
(50 mg kg1 i.p.). Blood for preparation of plasma was collected via the caudal vena cava
into tubes containing sodium citrate (final concentration 0.76%). Livers were removed
surgically for histopathological studies. Sections of the middle and left lateral liver lobes
were isolated and fixed in 10% neutral buffered formalin, and, 24 h later, preserved in 70%
ethanol. Additional sections were frozen immediately in liquid nitrogen and stored at
80 C. These experiments were repeated twice with similar results.
1182 J. Olivero-Verbel et al.

Enzyme and cytokine analysis


Plasma alanine aminotransferase (ALT) activity (a marker of hepatocellular damage) was
measured at 37 C using a kinetic enzymatic assay (Infinity kits; Melbourne, Australia).
Plasma cytokine concentrations were measured using a cytometric bead array assay
(BD Biosciences, San Jose, CA).

Histological examination
Hepatic tissue fixed in 10% neutral buffered formalin was embedded in paraffin, sectioned
at 8 mm, and stained with hematoxylin and eosin (H&E) using standard histological
techniques. Frozen sections stained with oil Red O were used to detect hepatocellular lipid
accumulation.
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

Quantification of hepatic neutrophil infiltration


Liver sections prepared as described above were deparaffinized and rehydrated, and then
stained using an anti-neutrophil Ig isolated from the serum of rabbits immunized with rat
neutrophils as described by Hewett et al. (1992). After incubation with the primary
antibody, tissue sections were incubated with alkaline phosphatase-conjugated goat-anti-
rabbit IgG (Vector Laboratories, Burlingame, CA) and the Fast-Red chromogen. Sections
were counterstained using Gills Hematoxylin II and then mounted. For each animal,
stained cells with nuclear morphology characteristic of neutrophils were quantified. These
cells were counted in at least 15 randomly selected, high-power (400) microscopic fields
(HPF) from the middle and left hepatic lobes. For each specimen, collected data
represented the average number of cells per HPF.

RNA isolation and reverse transcription-polymerase chain reaction


Total RNA was obtained from liver sections using MagMax–96 total RNA isolation kit
(Applied Biosystems, Foster City, CA).
For real-time polymerase chain reaction (PCR), reverse transcription of RNA samples
was performed using the Superscript kit from Invitrogen (Carlsbad, CA). Real-time PCR
analysis of cDNA was performed at 95–60 C for 40 cycles in a StepOne real time PCR
System (Applied Biosystems, Foster City, California) following the manufacturer’s
instructions, using Taqman Fast Universal PCR Master Mix as well as primers from
Applied Biosystems. Also, -actin mRNA was used as the housekeeping gene, and
quantification was performed using calibration curves for each target. Results for target
genes were normalized to -actin, and relative expression was calculated using threshold
cycle (Ct) values.

Statistical analysis
Data are expressed as the mean  SEM. Statistical analysis for comparisons between
treatment groups was performed using one-way analysis of variance (ANOVA). Multiple
comparisons were performed using the Tukey–Kramer method. p-Values less than 0.05
were considered statistically significant. All calculations were performed using SigmaStat
Software (Aspire Software International, VA).
Toxicological & Environmental Chemistry 1183
40,000 3500 600
Vehicle/Saline Vehicle/Saline Vehicle/Saline
IL-6 3000 * IL-10 IL-12
* TCDD/Saline TCDD/Saline 500 TCDD/Saline
* Vehicle/LPS Vehicle/LPS Vehicle/LPS
30,000
)

)
)
2500
–1

–1
–1
TCDD/LPS TCDD/LPS TCDD/LPS
400
IL-6 (pg mL

IL-12 (pg mL
IL-10 (pg mL
2000
20,000 300
1500
200
1000
10,000
500 100

0 0 0
0 4 8 12 16 20 24 0 4 8 12 16 20 24 0 4 8 12 16 20 24
Hours after LPS Hours after LPS Hours after LPS

2500 3500 50,000


Vehicle/Saline * Vehicle/Saline
* IFN-g Vehicle/Saline
3000 TNF-a MCP-1 TCDD/Saline
TCDD/Saline * TCDD/Saline
2000 40,000
)

)
–1

Vehicle/LPS Vehicle/LPS

)
–1
Vehicle/LPS

–1
2500
IFN-g (pg mL

TCDD/LPS TCDD/LPS

TNF-a (pg mL

MCP-1 (pg mL
TCDD/LPS
1500 2000 30,000

1000 1500 20,000


1000
500 10,000
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

500
0 0 0
0 4 8 12 16 20 24 0 4 8 12 16 20 24 0 4 8 12 16 20 24
Hours after LPS Hours after LPS Hours after LPS

Figure 1. Concentrations of cytokines/chemokines in plasma of mice treated with TCDD and/or


LPS. Mice received a single administration of TCDD (30 mg kg1) or its vehicle, p.o., and 4 days later
animals were given a single dose of LPS (0.05 x 107 EU kg1, i.p.) or its saline vehicle. Mice were
anesthetized before or 2,4,6,8,12, or 24 h after administration of LPS/saline. Plasma was collected for
determination of the concentrations of IL-6, IL-10, IFN- , TNF- , MCP-1, and IL-12. Values are
means  SEM; n ¼ 5. In most panels, symbols for Vehicle/Saline-treated mice are obscured by
symbols for TCDD/Saline-treated mice.
*Significantly different from Vehicle þ LPS.

Results
Inflammation in TCDD/LPS-treated mice
The time course of changes in plasma concentrations of cytokines was determined
(Figure 1). The concentration of interleukin (IL)-6 in plasma of vehicle-treated mice was
small and did not change significantly over 24 h. Similarly, treatment with TCDD alone
did not markedly affect the plasma concentration of IL-6. LPS produced an increase in
plasma IL-6 concentration by 2 h that returned to baseline by 8 h. In TCDD/LPS-treated
mice, the peak concentration of IL-6 was three-fold greater than that seen with LPS alone.
A similar pattern was observed for IL-10, interferon (IFN)- , and tumor necrosis factor
(TNF)- . TCDD also augmented the LPS-induced rise in monocyte chemoattractant
protein (MCP)-1, but the magnitude of this effect was small compared to the other
mediators. TCDD did not markedly affect the LPS-mediated increase in IL-12.
LPS produced a dose-dependent rise in the concentration of IL-6 in plasma at 4 h
(Figure 2). Pretreatment with TCDD resulted in a shift of this dose–response relationship
to the left, with significant differences observed at LPS doses of 0.05  107 and
0.2  107 EU kg1 LPS.

Cytokine/chemokine mRNA expression in liver


TCDD alone did not markedly alter the levels of mRNA for IL-6, IL-10, IL-12, IFN- ,
TNF- , or MCP-1 (Figure 3). LPS increased the hepatic expression of all measured
cytokines/chemokines. TCDD augmented the LPS-dependent enhancement of expression
of IL-10 and IFN- mRNAs.
1184 J. Olivero-Verbel et al.
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

Figure 2. Dose–response relationship for plasma concentration of IL-6 in mice treated with TCDD
and/or LPS. Mice received a single administration of TCDD (30 mg kg1) or its vehicle, p.o., and 4
days later they were given saline or a single administration of LPS (0.01–3.2 x 107 EU kg1, i.p.). Mice
were anesthetized and plasma taken 4 h after administration of LPS. Values are means  SEM; n ¼ 5.
*Significantly different from vehicle at the same dose of LPS.
IL-6 mRNA (Relative expression)

IL-12 mRNA (Relative expression)


60
IL-10 mRNA (Relative expression)

14 300
Vehicle *
IL-6 Vehicle IL-10 ** IL-12
50 TCDD 12 Vehicle *
TCDD 250
TCDD
40 * 10
200
*
8 *
30 150
6
20 100
4
10 50
2
0 0 0
Saline LPS Saline LPS Saline LPS
MCP-1 mRNA (Relative expression)
TNF-α mRNA (Relative expression)
IFN-γ mRNA (Relative expression)

100 120
Vehicle Vehicle *
Vehicle *
30 TCDD IFN-g TNF-a 100 MCP-1 *
** TCDD * TCDD
80
80
20 60
60
40
40
10
20 20

0 0 0
Saline LPS Saline LPS Saline LPS

Figure 3. mRNA expression of cytokines in livers from mice treated with LPS and/or TCDD. Mice
were treated as described in the legend to Figure 1, and livers were collected for measurement of
mRNA by RT-PCR immediately before or 2 h after LPS administration. For calculation of relative
changes, the average expression for the vehicle group before treatment with LPS (time 0) was set as 1.
Values are means  SEM from at least three animals/group.
*Significantly different from respective group treated with saline.
**Significantly different from the respective group not exposed to TCDD.

Liver damage, hepatic histology, and neutrophil accumulation


ALT activity was significantly greater in plasma from TCDD- or TCDD/LPS-treated mice
compared to controls at several time points over the 24 h administration of LPS (Table 1).
At 8 h, mice treated with TCDD/LPS displayed greater ALT activity in plasma compared
to those treated with TCDD only. Livers of mice treated with vehicle appeared normal
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

Table 1. Time course of ALT activity in plasma of mice treated with TCDD and/or LPS.

Plasma ALT activity (U/L)

Hours after LPS administration

Treatment 0 2 4 8 12 24

Veh/Sal 27.5  1.4 (12) 44.5  7.4 (5) 44.5  3.1 (9) 31.3  2.3 (9) 35.7  1.8 (5) 55.2  3.9 (5)
TCDD/Sal 102.8  10.7* (14) 71.5  14.0 (6) 92.7  14.6* (8) 52.9  6.8 (7) 63.4  3.7 (6) 79.0  6.6 (6)
Veh/LPS NA 44.2  4.0 (5) 58.7  6.8 (9) 53.4  5.5 (9) 47.4  6.3 (7) 110.3  31.2 (7)
TCDD/LPS NA 94.9  7.8* (7) 108.4  15.1* (7) 84.7  14.8** (7) 80.2  10.5* (8) 95.8  8.1 (5)

Notes: Mice received a single administration of TCDD (30 mg kg1) or its vehicle, p.o., and 4 days later were given a single dose of LPS
(0.05  107 EU kg1, i.v.) or its saline vehicle. Mice were anesthetized immediately before (time 0) or 2, 4, 8, 12, or 24 h after administration of LPS or its
vehicle, and plasma was collected for determination of the activity of ALT. Data were analyzed within time by one-way ANOVA. Numbers in parentheses
represent n for each group.
*Significantly different from respective group in the absence of TCDD treatment.
**Significantly different from respective group in the absence of LPS treatment.
Toxicological & Environmental Chemistry
1185
1186 J. Olivero-Verbel et al.

histologically (Figure 4). Intracytoplasmic lipid droplets were prominent in livers from
animals treated only with TCDD, and occasional foci of inflammatory cells (mononuclear
and neutrophils) were observed in isolated foci throughout the liver sections. Livers of
mice treated only with LPS had abundant neutrophils scattered throughout the lobules
(not in isolated foci) but no necrosis. In livers of mice co-treated with TCDD and LPS,
steatosis was prominent, and neutrophils appeared in large foci of inflammatory cells.
The number of neutrophils in liver was quantified (Figure 5). LPS administration
produced an increase in the number of hepatic neutrophils by 2 h that was sustained for at
least 12 h. Except for a small reduction at 2 h, TCDD did not markedly affect this response
to LPS.
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

Lipid accumulation in liver tissue


In the absence of TCDD or LPS, little lipid was detected in liver sections (Figure 6). Mice
treated 4 days earlier with TCDD had detectable oil Red O staining in their livers before
treatment with LPS. Treatment with LPS alone induced some lipid deposition.
Co-treatment with TCDD and LPS resulted in pronounced hepatocellular lipid accumu-
lation, with somewhat larger lipid droplets.

Discussion
AhR agonists such as dioxins modulate diverse biochemical and physiological processes
including oxidative stress, metabolism, circadian rhythm, cell proliferation and differen-
tiation, tumor formation, and teratogenesis (Barouki, Coumoul and Fernandez-Salguero
2007; Lin et al. 2007; Shimba and Watabe 2009). In this study, the effects of TCDD on
LPS-induced inflammation were investigated in vivo, showing evidence of synergistic
effects of TCDD on LPS-induced cytokine production.
Co-treatment of mice with TCDD and LPS led to greater protein expression of several
cytokines/chemokines compared to treatment with either agent alone (Figures 1 and 2).
Interactions between TCDD and LPS have been studied both in vitro and in vivo, and
diverse results were noted. For instance, in LPS-sensitized RAW 267.4 cells, a murine
macrophage cell line, TCDD amplified mRNA expression of IL-6 and -1 , but not TNF-
(Pestka and Zhou 2006). On the other hand, TCDD reduced LPS-stimulated IL-6 mRNA
and protein expression in a bone marrow stromal cell line (Jensen et al. 2003).
Furthermore, TCDD exerted no marked effect on IL-6 or IL-8 production induced by
LPS or granulocyte colony-stimulating factor in human airway epithelial cells, alveolar
macrophages, or peripheral blood leukocytes (Lang et al. 1998). In vivo studies showed
that TCDD alters the LPS-suppressed IgM antibody-forming cell number (North et al.
2009), as well as modulates caspase activity in LPS-treated animals independently from
NFkappaB (Patterson, Stachlewitz, and Germolec 2003) and increased lethality from LPS
(Rosenthal et al. 1989). Thus, the effect of interaction of TCDD and LPS in vitro and
in vivo varies with cell type and with treatment conditions.
Despite significant increases in plasma cytokine concentrations, TCDD led to
enhanced mRNA expression in liver only for IFN- and IL-10. One explanation for
this is that the cytokines for which hepatic mRNA expression was not raised are produced
in other tissues. For instance, IL-6 and TNF- are also produced in adipose tissue (Ishii
and Yoshida 2010; Tilg 2010). Another possibility is that cytokines are produced by
nonparenchymal cells in the liver, and the contribution of these cells to total mRNA
Toxicological & Environmental Chemistry 1187

(A)
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

(B)

(C)

(D)

Figure 4. Histopathology and neutrophil accumulation in livers of mice treated with TCDD and/or
LPS. Mice were treated as described in the legend to Figure 1. Liver samples were taken before
(A:Veh, B:TCDD) or 2 h after (C:Veh/LPS, D:TCDD/LPS) LPS treatment. Livers were stained for
neutrophils as described in ‘‘Materials and methods’’, and these cells appear red in the
photomicrographs. Sections on the left were photographed at a magnification of 600 . Sections
on the right were photographed at a magnification of 200 and are presented to show the
distribution of neutrophils and lesions.
1188 J. Olivero-Verbel et al.

20
Vehicle
TCDD

Hepatic Neutrophils/HPF
15

10
*
5

0
0 2 12 24
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

Hours after LPS

Figure 5. Quantification of neutrophil accumulation in livers from mice treated with TCDD and/or
LPS. Mice were treated as described in the legend to Figure 1, and liver samples were taken before
(time 0) or 2, 12, or 24 h after LPS treatment. Neutrophils were quantified as described in Materials
and methods. Values are means  SEM from at least three animals/group.
*Significantly different from vehicle at the same time.

Veh/Sal Veh/LPS

TCDD/Sal TCDD/LPS

Figure 6. Lipid accumulation in livers of mice treated with TCDD and/or LPS. Mice were treated as
described in the legend to Figure 1. Liver samples were taken 24 h after LPS treatment, and lipid
deposition was detected by staining with oil Red O as described in Materials and methods (Original
magnification 200).

expression is not sufficient to detect these changes. In addition, LPS-stimulated TNF-


release might be mediated by the action of TNF- converting enzyme (TACE) to cleave
pro-TNF from cell surfaces (Mohler et al. 1994; Horiuchi et al. 2007). It is also possible
that TCDD elevated plasma TNF- concentration by this mechanism, which would not
necessitate enhanced transcription.
Toxicological & Environmental Chemistry 1189

Treatment of rats in vivo with TCDD led to greater nitric oxide and TNF- production
in response to LPS (Glover et al. 2000); the latter result is similar to results presented here
for mice (Figure 1). In some studies in which LPS was co-administered with other agents
with resultant elevation in plasma concentrations of proinflammatory cytokines in excess
of those produced by LPS alone, animals developed liver damage (Barton et al. 2001; Yee,
Ganey, and Roth 2003; Shaw, Ganey, and Roth 2009). This is in contrast to findings
presented here for TCDD in which co-treatment led to larger foci of inflammatory cells
and greater lipid accumulation, but no frank injury. In other studies, co-treatment of mice
with TCDD and LPS led to greater serum ALT activity and increased hepatic apoptosis
compared to treatment with either agent alone, but these effects were not observed until 10
and 14 days after exposure, respectively (Patterson, Stachlewitz, and Germolec 2003). In
addition, the dose of TCDD used in that study was greater (100 mg kg1) than the one
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

used here.
Earlier studies suggested that TCDD alone has proinflammatory properties. For
example, in human macrophages, TCDD induced several cytokine and chemokine genes in
a manner dependent on RelB (Vogel, Sciullo, and Matsumura 2007), a subunit of
NF-kappa B that physically interacts with AhR (Vogel et al. 2007b). TCDD enhanced the
hepatic expression of MCP-1 and keratinocyte chemoattractant (KC) in mice within 1 day
of treatment (Vogel et al. 2007a). The rise was sustained for 10 days and accompanied by
an elevation in expression of a marker for macrophages at day 7. Moreover, at least in the
case of KC, a functional AhR was necessary for the induction. Furthermore, Clark and
Taylor (1994) proposed that some of the toxicity of TCDD might be attributed to TNF-
production. These results are in contrast to data presented here demonstrating that 4 days
after treatment, TCDD alone did not alter hepatic gene expression or plasma protein
concentration for a number of cytokines (Figures 1 and 3). Others also observed a lack of
induction of proinflammatory mediators after treatment with TCDD alone (Glover et al.
2000; Patterson, Stachlewitz, and Germolec 2003).
TCDD augmented the plasma levels of IL-6, IL-10, and IFN- induced by LPS. Nandi
et al. (2010) reported that IL-6 can attenuate liver injury induced by LPS, probably by
modulating the release of IL-10, a potent anti-inflammatory cytokine. This might explain
the lack of frank hepatic necrosis in TCDD/LPS-treated mice. However, Gomez et al.
(2009) linked IL-6 to tissue injury, although its role may be insult-specific. Regarding
IFN- , the effects of TCDD on LPS-induced expression are similar to those observed for
ovalbumin (Nohara et al. 2009), an effect attributed to AhR activation on T-cells.
Inflammation is associated with accumulation of inflammatory cells, particularly
neutrophils, in tissue (Alves-Filho et al. 2008). Unlike the effect to increase plasma
concentrations of cytokines, TCDD produced a mild decrease in neutrophil accumulation
in the livers 2 h after LPS injection. This effect might be the result of neutrophil migration
toward large foci of inflammatory cells, decreasing the apparent number of cells per field.
Vorderstrasse and Lawrence (2006) showed that treatment of mice with TCDD prior to
intranasal infection with Streptococcus pneumoniae increased survival but decreased the
number of neutrophils in the lungs. In this study, large foci of inflammatory cells
(mononuclear cells and neutrophils) were observed in livers of co-treated mice. These
mixed cell infiltrations, were also found in immature, ovariectomized mice treated with
larger doses of TCDD (Kopec et al. 2008), and were linked with changes in expression of
immune signaling-related genes (Swindell, Johnston, and Gudjonsson 2010).
As shown here, liver steatosis induced by TCDD increased after treatment with LPS.
Hepatic lipid accumulation is a hallmark of TCDD exposure (Chang et al. 2005). Steatosis
has been considered as a predisposing factor for more severe inflammation-related
1190 J. Olivero-Verbel et al.

pathological conditions such as fibrosis (Yilmaz et al. 2007) and cirrhosis (Reddy and Rao
2006; Björnsson and Angulo 2007). Steatosis has also been linked to minor increases in
serum activity of ALT in animal models and humans (Guzzaloni et al. 2000), and this
might explain the small increases in plasma ALT activity observed in mice treated with
TCDD in the studies presented here (Table 1). Moreover, TNF- induces hepatic steatosis
by enhancing gene expression of sterol regulatory element binding protein-1c (SREBP-1c)
(Endo et al. 2007), and TCDD augmented the rise in TNF- plasma concentration induced
by LPS.
Evidence is accumulating that agonists of the AhR, such as TCDD (this study),
3-methylcholanthrene (Sekine et al. 2009), benzo[a]pyrene (Andreou et al. 2004), and
indirubin-30 -monoxime (Springs and Rice 2006), alter expression of genes and proteins
involved in inflammation-related processes. These effects might be important in the
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

initiation or progression of chemically or biologically induced pathological conditions


involving AhR, as explored for airborne particulate-induced carcinogenesis (Matsumoto
et al. 2007), chemically induced atopic dermatitis (Ito et al. 2008), infections by viruses
(Bohn et al. 2005; Lawrence 2007; Neff-LaFord et al. 2007), and several bacteria such as
Listeria monocytogenes (Shi et al. 2007) and S. pneumoniae (Vorderstrasse and Lawrence
2006). Data presented here support this notion and demonstrate that a potent agonist of
the AhR modulates inflammatory processes mediated by LPS, both at molecular and
cellular levels.

Acknowledgments
This study was supported by the National Institutes of Health, National Institute of Environmental
Health Sciences [Grant ES004911]. Jesus Olivero-Verbel was supported in part by the Postdoctoral
Training Program of Colciencias (2006–2007), Bogotá, Colombia, and the University of Cartagena,
Cartagena, Colombia.

References

Allcock, G.H., M. Allegra, R.J. Flower, and M. Perretti. 2001. Neutrophil accumulation induced by
bacterial lipopolysaccharide: Effects of dexamethasone and annexin 1. Clinical and Experimental
Immunology 123: 62–7.
Alves-Filho, J.C., A. de Freitas, F. Spiller, F.O. Souto, and F.Q. Cunha. 2008. The role of
neutrophils in severe sepsis. Shock 30: 3–9.
Andreou, V., M. D’Addario, R. Zohar, B. Sukhu, R.F. Casper, R.P. Ellen, and H.C. Tenenbaum.
2004. Inhibition of osteogenesis in vitro by a cigarette smoke-associated hydrocarbon combined
with Porphyromonas gingivalis lipopolysaccharide: Reversal by resveratrol. Journal of
Periodontology 75: 939–48.
Barouki, R., X. Coumoul, and P.M. Fernandez-Salguero. 2007. The aryl hydrocarbon receptor more
than a xenobiotic-interacting protein. FEBS Letters 581: 3608–15.
Barton, C.C., E.X. Barton, P.E. Ganey, S.L. Kunkel, and R.A. Roth. 2001. Bacterial lipopolysac-
charide enhances aflatoxin B1 hepatotoxicity in rats by a mechanism that depends on tumor
necrosis factor alpha. Hepatology 33: 66–73.
Björnsson, E., and P. Angulo. 2007. Hepatitis C and steatosis. Archives of Medical Research 38:
621–7.
Bohn, A.A., K.S. Harrod, S. Teske, and B.P. Lawrence. 2005. Increased mortality associated with
TCDD exposure in mice infected with influenza A virus is not due to severity of lung injury or
alterations in Clara cell protein content. Chemico-Biological Interactions 155: 181–90.
Toxicological & Environmental Chemistry 1191

Chan, J.K., G.H. Xing, Y. Xu, Y. Liang, L.X. Chen, S.C. Wu, C.K. Wong, C.K. Leung, and
M.H. Wong. 2007. Body loadings and health risk assessment of polychlorinated dibenzo-
p-dioxins and dibenzofurans at an intensive electronic waste recycling site in China.
Environmental Science and Technology 41: 7668–74.
Chang, H., Y.J. Wang, L.W. Chang, and P. Lin. 2005. A histochemical and pathological study on
the interrelationship between TCDD-induced AhR expression, AhR activation, and hepatotox-
icity in mice. Journal of Toxicology and Environmental Health Part A 68: 1567–79.
Chatterjee, R. 2007. E-waste recycling spews dioxins into the air. Environmental Science and
Technology 41: 5577.
Clark, G.C., and M.J. Taylor. 1994. Tumor necrosis factor involvement in the toxicity of TCDD:
The role of endotoxin in the response. Experimental and Clinical Immunogenetics 11: 136–41.
Endo, M., T. Masaki, M. Seike, and H. Yoshimatsu. 2007. TNF-alpha induces hepatic steatosis in
mice by enhancing gene expression of sterol regulatory element binding protein-1c (SREBP-1c).
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

Experimental Biology and Medicine (Maywood) 232: 614–21.


Glover, R.E., D.R. Germolec, R. Patterson, N.J. Walker, G.W. Lucier, and R.P Mason. 2000.
Endotoxin (lipopolysaccharide)-induced nitric oxide production in 2,3,7,8-tetrachlorodibenzo-p-
dioxin-treated Fischer rats: Detection of nitrosyl hemoproteins by EPR spectroscopy. Chemical
Research in Toxicology 13: 1051–5.
Gomez, C.R., V. Nomellini, H. Baila, K. Oshima, and E.J. Kovacs. 2009. Comparison of the effects
of aging and IL-6 on the hepatic inflammatory response in two models of systemic injury: Scald
injury versus i.p. LPS administration. Shock 31: 178–84.
Guzzaloni, G., G. Grugni, A. Minocci, D. Moro, and F. Morabito. 2000. Liver steatosis in juvenile
obesity: Correlations with lipid profile, hepatic biochemical parameters and glycemic and
insulinemic responses to an oral glucose tolerance test. International Journal of Obesity and
Related Metabolic Disorders 24: 772–6.
Hewett, J.A., A.E. Schultze, S. VanCise, and R.A. Roth. 1992. Neutrophil depletion protects against
liver injury from bacterial endotoxin. Laboratory Investigation 66: 347–61.
Hillegass, J.M., K.A. Murphy, C.M. Villano, and L.A. White. 2006. The impact of aryl hydrocarbon
receptor signaling on matrix metabolism: Implications for development and disease. Biological
Chemistry 387: 1159–73.
Horiuchi, K., T. Kimura, T. Miyamoto, H. Takaishi, Y. Okada, Y. Toyama, and C.P. Blobel. 2007.
TNF-alpha-converting enzyme (TACE/ADAM17) inactivation in mouse myeloid cells prevents
lethality from endotoxin shock. Journal of Immunological Studies 179: 2686–9.
Ilbäck, N.G., and G. Friman. 2007. Interactions among infections, nutrients and xenobiotics.
Critical Reviews in Food Science and Nutrition 47: 499–519.
Ishii, H., and M. Yoshida. 2010. Inflammatory cytokines. Nippon Rinsho 68: 819–22.
Ito, T., K. Inouye, K. Nohara, C. Tohyama, and H. Fujimaki. 2008. TCDD exposure exacerbates
atopic dermatitis-related inflammation in NC/Nga mice. Toxicology Letters 177: 31–7.
Jensen, B.A., R.J. Leeman, J.J. Schlezinger, and D.H. Sherr. 2003. Aryl hydrocarbon receptor
(AhR) agonists suppress interleukin-6 expression by bone marrow stromal cells: An immunotox-
icology study. Environmental Health 2: 16–28.
Khan, S., S. Liu, M. Stoner, and S. Safe. 2007. Cobaltous chloride and hypoxia inhibit aryl
hydrocarbon receptor-mediated responses in breast cancer cells. Toxicology and Applied
Pharmacology 223: 28–38.
Kopec, A.K., D.R. Boverhof, L.D. Burgoon, D. Ibrahim-Aibo, J.R. Harkema, C. Tashiro, B.
Chittim, and T.R. Zacharewski. 2008. Comparative toxicogenomic examination of the hepatic
effects of PCB126 and TCDD in immature, ovariectomized C57BL/6 mice. Toxicology Science
102: 61–75.
Kulkarni, P.S., J.G. Crespo, and C.A. Afonso. 2008. Dioxins sources and current remediation
technologies–A review. Environment International 34: 139–53.
Lang, D.S., S. Becker, R.B. Devlin, and H.S. Koren. 1998. Cell-specific differences in the
susceptibility of potential cellular targets of human origin derived from blood and lung following
1192 J. Olivero-Verbel et al.

treatment with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Cell Biology and Toxicology 14:


23–38.
Lawrence, B.P. 2007. Environmental toxins as modulators of antiviral immune responses. Viral
Immunology 20: 231–42.
Lin, P.H., C.H. Lin, C.C. Huang, M.C. Chuang, and P. Lin. 2007. 2,3,7,8-Tetrachlorodibenzo-p-
dioxin (TCDD) induces oxidative stress, DNA strand breaks, and poly(ADP-ribose) polymerase-1
activation in human breast carcinoma cell lines. Toxicology Letters 172: 146–58.
Matsumoto, Y., F. Ide, R. Kishi, T. Akutagawa, S. Sakai, M. Nakamura, T. Ishikawa, Y. Fujii-
Kuriyama, and Y. Nakatsuru. 2007. Aryl hydrocarbon receptor plays a significant role in
mediating airborne particulate-induced carcinogenesis in mice. Environmental Science and
Technology 41: 3775–80.
Matsumura, F., and C.F. Vogel. 2006. Evidence supporting the hypothesis that one of the main
functions of the aryl hydrocarbon receptor is mediation of cell stress responses. Biological
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

Chemistry 387: 1189–94.


Mayeux, P.R. 1997. Pathobiology of lipopolysaccharide. Journal of Toxicology and Environmental
Health 51: 415–35.
Miyamoto, K. 2004. Effects of dioxin on gene expression in female reproductive system in the rat.
Environmental Science 11: 47–55.
Mohler, K.M., P.R. Sleath, J.N. Fitzner, D.P. Cerretti, M. Alderson, S.S. Kerwar, D.S. Torrance,
et al. 1994. Protection against a lethal dose of endotoxin by an inhibitor of tumour necrosis factor
processing. Nature 370: 218–20.
Nandi, D., M.K. Mishra, A. Basu, and B. Bishayi. 2010. Protective effects of interleukin-6 in
lipopolysaccharide (LPS)-induced experimental endotoxemia are linked to alteration in hepatic
anti-oxidant enzymes and endogenous cytokines. Immunobiology 215: 443–51.
Neff-LaFord, H., S. Teske, T.P. Bushnell, and B.P. Lawrence. 2007. Aryl hydrocarbon receptor
activation during influenza virus infection unveils a novel pathway of IFN-gamma production by
phagocytic cells. Journal of Immunology 179: 247–55.
Nohara, K., T. Suzuki, K. Ao, H. Murai, Y. Miyamoto, K. Inouye, X. Pan, H. Motohashi, Y. Fujii-
Kuriyama, M. Yamamoto, and C. Tohyama. 2009. Constitutively active aryl hydrocarbon
receptor expressed in T cells increases immunization-induced IFN-gamma production in mice but
does not suppress T(h)2-cytokine production or antibody production. International Immunology
21: 769–77.
North, C.M., R.B. Crawford, H. Lu, and N.E. Kaminski. 2009. Simultaneous in vivo time course
and dose response evaluation for TCDD-induced impairment of the LPS-stimulated primary IgM
response. Toxicological Sciences 112: 123–32.
Ohhira, M., W. Motomura, M. Fukuda, T. Yoshizaki, N. Takahashi, S. Tanno, N. Wakamiya, Y.
Kohgo, S. Kumei, and T. Okumura. 2007. Lipopolysaccharide induces adipose differentiation-
related protein expression and lipid accumulation in the liver through inhibition of fatty acid
oxidation in mice. Journal of Gastroenterology 42: 969–78.
Pape, F., and R. Stahlmann. 2007. Chloracne after exposure to dioxin. Medizinische Monatsschrift
für Pharmazeuten 30: 206–12.
Patel, R.D., D.J. Kim, J.M. Peters, and G.H. Perdew. 2006. The aryl hydrocarbon receptor directly
regulates expression of the potent mitogen epiregulin. Toxicological Sciences 89: 75–82.
Patterson, R.M., R. Stachlewitz, and D. Germolec. 2003. Induction of apoptosis by 2,3,7,8-
tetrachlorodibenzo-p-dioxin following endotoxin exposure. Toxicology and Applied Pharmacology
190: 120–34.
Pelclová, D., P. Urban, J. Preiss, E. Lukás, Z. Fenclová, T. Navrátil, Z. Dubská, and Z. Senholdová.
2006. Adverse health effects in humans exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD).
Reviews on Environmental Health 21: 119–38.
Pestka, J., and H.R. Zhou. 2006. Toll-like receptor priming sensitizes macrophages to
proinflammatory cytokine gene induction by deoxynivalenol and other toxicants. Toxicological
Sciences 92: 445–55.
Toxicological & Environmental Chemistry 1193

Reddy, J.K., and M.S. Rao. 2006. Lipid metabolism and liver inflammation. II. Fatty liver disease
and fatty acid oxidation. American Journal of Physiology Gastrointestinal and Liver Physiology
290: G852–8.
Rose, R., A. Banerjee, and S.K. Ramaiah. 2007. Characterization of a lipopolysaccharide mediated
neutrophilic hepatitis model in Sprague-Dawley rats. Journal of Applied Toxicology 27: 602–11.
Rosenthal, G.J., E. Lebetkin, J.E. Thigpen, R. Wilson, A.N. Tucker, and M.I. Luster. 1989.
Characteristics of 2,3,7,8-tetrachlorodibenzo-p-dioxin induced endotoxin hypersensitivity:
Association with hepatotoxicity. Toxicology 56: 239–51.
Sato, S., H. Shirakawa, S. Tomita, Y. Ohsaki, K. Haketa, O. Tooi, N. Santo, et al. 2008. Low-dose
dioxins alter gene expression related to cholesterol biosynthesis, lipogenesis, and glucose
metabolism through the aryl hydrocarbon receptor-mediated pathway in mouse liver.
Toxicology and Applied Pharmacology 229: 10–19.
Sekine, H., J. Mimura, M. Oshima, H. Okawa, J. Kanno, K. Igarashi, F.J. Gonzalez, T. Ikuta,
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

K. Kawajiri, and Y. Fujii-Kuriyama. 2009. Hypersensitivity of aryl hydrocarbon receptor-


deficient mice to lipopolysaccharide-induced septic shock. Molecular Cell Biology 29: 6391–400.
Shaw, P.J., P.E. Ganey, and R.A. Roth. 2009. Tumor necrosis factor alpha is a proximal mediator of
synergistic hepatotoxicity from trovafloxacin/lipopolysaccharide coexposure. Journal of
Pharmacology and Experimental Therapeutics 328: 62–8.
Shi, L.Z., N.G. Faith, Y. Nakayama, M. Suresh, H. Steinberg, and C.J. Czuprynski. 2007. The aryl
hydrocarbon receptor is required for optimal resistance to Listeria monocytogenes infection in
mice. Journal of Immunology 179: 6952–62.
Shimba, S., and Y. Watabe. 2009. Crosstalk between the AHR signaling pathway and circadian
rhythm. Biochemistry and Pharmacology 77: 560–5.
Springs, A.E., and C.D. Rice. 2006. The effects of indirubin-30 -monoxime, a novel AhR ligand, on
stress and toxicity-related gene/protein expression in human U937 cells undergoing differentiation
and activation. Jouranl of Immunotoxicology 3: 1–10.
Sugita-Konishi, Y., K. Kobayashi, H. Naito, K. Miura, and Y. Suzuki. 2003. Effect of lactational
exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin on the susceptibility to Listeria infection.
Bioscience, Biotechnology and Biochemistry 67: 89–93.
Swindell, W.R., A. Johnston, and J.E. Gudjonsson. 2010. Transcriptional profiles of leukocyte
populations provide a tool for interpreting gene expression patterns associated with high fat diet in
mice. PLoS One 5: e11861.
Teske, S., A.A. Bohn, J.P. Hogaboam, and B.P. Lawrence. 2008. Aryl hydrocarbon receptor targets
pathways extrinsic to bone marrow cells to enhance neutrophil recruitment during influenza virus
infection. Toxicological Sciences 102: 89–99.
Tilg, H. 2010. The role of cytokines in non-alcoholic fatty liver disease. Digestive Diseases 28:
179–85.
Vogel, C.F., N. Nishimura, E. Sciullo, P. Wong, W. Li, and F. Matsumura. 2007a. Modulation of
the chemokines KC and MCP-1 by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in mice. Archives
of Biochemistry and Biophysics 461: 169–75.
Vogel, C.F., E. Sciullo, W. Li, P. Wong, G. Lazennec, and F. Matsumura. 2007b. RelB, a new
partner of aryl hydrocarbon receptor-mediated transcription. Molecular Endocrinology 12:
2941–55.
Vogel, C.F., E. Sciullo, and F. Matsumura. 2007. Involvement of RelB in aryl hydrocarbon receptor-
mediated induction of chemokines. Biochemical and Biophysical Research Communications 363:
722–6.
Vorderstrasse, B.A., and B.P. Lawrence. 2006. Protection against lethal challenge with Streptococcus
pneumoniae is conferred by aryl hydrocarbon receptor activation but is not associated with an
enhanced inflammatory response. Infection and Immunity 74: 5679–86.
Xu, M.X., J.H. Yan, S.Y. Lu, X.D. Li, T. Chen, M.J. Ni, H.F. Dai, F. Wang, and K.F. Cen. 2009.
Concentrations, profiles, and sources of atmospheric PCDD/Fs near a municipal solid waste
incinerator in Eastern China. Environmental Science and Technology 43: 1023–9.
1194 J. Olivero-Verbel et al.

Yamasawa, H., Y. Ishii, and S. Kitamura. 1999. Cytokine-induced neutrophil chemoattractant in a


rat model of lipopolysaccharide-induced acute lung injury. Inflammation 23: 263–74.
Yee, S.B., P.E. Ganey, and R.A. Roth. 2003. The role of Kupffer cells and TNF-alpha in
monocrotaline and bacterial lipopolysaccharide-induced liver injury. Toxicological Sciences 71:
124–32.
Yilmaz, N., M.L. Shiffman, R.T. Stravitz, R.K. Sterling, V.A. Luketic, A.J. Sanyal, A.S. Mills, et al.
2007. A prospective evaluation of fibrosis progression in patients with recurrent hepatitis C virus
following liver transplantation. Liver Transplantation 13: 975–83.
Downloaded by [Australian Catholic University] at 13:55 21 August 2011

You might also like