ArticlePDF Available

Interaction between Porcine Reproductive-Respiratory Syndrome Virus and Bacterial Endotoxin in the Lungs of Pigs: Potentiation of Cytokine Production and Respiratory Disease

American Society for Microbiology
Journal of Clinical Microbiology
Authors:

Abstract and Figures

Porcine reproductive-respiratory syndrome virus (PRRSV) is a key agent in multifactorial respiratory disease of swine. Intratracheal administration of bacterial lipopolysaccharides (LPSs) to PRRSV-infected pigs results in markedly enhanced respiratory disease, whereas the inoculation of each component alone results in largely subclinical disease. This study examines whether PRRSV-LPS-induced respiratory disease is associated with the excessive production of proinflammatory cytokines in the lungs. Gnotobiotic pigs were inoculated intratracheally with PRRSV and then with LPS at 3, 5, 7, 10, or 14 days of infection and euthanatized 6 h after LPS inoculation. Controls were inoculated with PRRSV or LPS only or with phosphate-buffered saline. Virus titers, (histo)pathological changes in the lungs, numbers of inflammatory cells, and bioactive tumor necrosis factor alpha (TNF-alpha), interleukin-1 (IL-1), and IL-6 levels in bronchoalveolar lavage fluids were examined. All pigs inoculated with PRRSV-LPS developed severe respiratory disease, whereas the controls that were inoculated with PRRSV or LPS alone did not. PRRSV infection significantly enhanced cytokine production in response to LPS. Peak TNF-alpha, IL-1, and IL-6 titers were 10 to 100 times higher in the PRRSV-LPS-inoculated pigs than in the pigs inoculated with PRRSV or LPS alone; and the titers correlated with the respiratory signs. The levels of neutrophil infiltration and the pathological changes detected in the lungs of PRRSV-LPS-inoculated pigs resembled those detected when the effects of PRRSV and LPS inoculated alone are combined, but with no synergistic effects between PRRSV and LPS. These data demonstrate a synergism between PRRSV and LPS in the induction of proinflammatory cytokines and an association between induction of these cytokines and disease.
Content may be subject to copyright.
JOURNAL OF CLINICAL MICROBIOLOGY, Mar. 2003, p. 960–966 Vol. 41, No. 3
0095-1137/03/$08.000 DOI: 10.1128/JCM.41.3.960–966.2003
Copyright © 2003, American Society for Microbiology. All Rights Reserved.
Interaction between Porcine Reproductive-Respiratory Syndrome
Virus and Bacterial Endotoxin in the Lungs of Pigs: Potentiation
of Cytokine Production and Respiratory Disease
Steven Van Gucht, Kristien Van Reeth,* and Maurice Pensaert
Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, B-9820 Merelbeke, Belgium
Received 1 August 2002/Returned for modification 22 October 2002/Accepted 15 December 2002
Porcine reproductive-respiratory syndrome virus (PRRSV) is a key agent in multifactorial respiratory
disease of swine. Intratracheal administration of bacterial lipopolysaccharides (LPSs) to PRRSV-infected pigs
results in markedly enhanced respiratory disease, whereas the inoculation of each component alone results in
largely subclinical disease. This study examines whether PRRSV-LPS-induced respiratory disease is associated
with the excessive production of proinflammatory cytokines in the lungs. Gnotobiotic pigs were inoculated
intratracheally with PRRSV and then with LPS at 3, 5, 7, 10, or 14 days of infection and euthanatized 6 h after
LPS inoculation. Controls were inoculated with PRRSV or LPS only or with phosphate-buffered saline. Virus
titers, (histo)pathological changes in the lungs, numbers of inflammatory cells, and bioactive tumor necrosis
factor alpha (TNF-), interleukin-1 (IL-1), and IL-6 levels in bronchoalveolar lavage fluids were examined. All
pigs inoculated with PRRSV-LPS developed severe respiratory disease, whereas the controls that were inoc-
ulated with PRRSV or LPS alone did not. PRRSV infection significantly enhanced cytokine production in
response to LPS. Peak TNF-, IL-1, and IL-6 titers were 10 to 100 times higher in the PRRSV-LPS-inoculated
pigs than in the pigs inoculated with PRRSV or LPS alone; and the titers correlated with the respiratory signs.
The levels of neutrophil infiltration and the pathological changes detected in the lungs of PRRSV-LPS-
inoculated pigs resembled those detected when the effects of PRRSV and LPS inoculated alone are combined,
but with no synergistic effects between PRRSV and LPS. These data demonstrate a synergism between PRRSV
and LPS in the induction of proinflammatory cytokines and an association between induction of these
cytokines and disease.
European strains of porcine reproductive-respiratory syn-
drome virus (PRRSV) fail to cause respiratory disease as such.
Nevertheless, PRRSV is considered one of the most important
etiological agents in multifactorial respiratory disease of swine
both in Europe and in the United States (18). Few studies,
however, have been able to reproduce clinical respiratory dis-
ease by experimental inoculation of PRRSV followed by inoc-
ulation of a secondary virus or bacterium (4, 6, 17, 21). Vari-
ations in the severities of clinical signs and a lack of
reproducibility are the main problems with these types of stud-
ies. Even a single experimental infection with respiratory vi-
ruses results in intrinsic variations in virological, inflammatory,
and clinical parameters. Therefore, a second infection may
enhance this variation, as the outcome of the second infection
is in part dependent on that of the first infection.
We have previously developed an alternative dual-inocula-
tion model consisting of a primary inoculation with PRRSV
followed by inoculation of a nonreplicating agent, namely, li-
popolysaccharide (LPS) from Escherichia coli (8). LPS is a
major component of the outer membrane and the main endo-
toxin of gram-negative bacteria. Intratracheal administration
of LPS (20 g/kg of body weight) to PRRSV-infected pigs
resulted in severe respiratory disease, characterized by tachy-
pnea, abdominal breathing, dyspnea, and high fever. In con-
trast, inoculation of PRRSV or LPS alone resulted in subclin-
ical or mild disease. This model proved to be reproducible, in
contrast to the classic dual-infection models consisting of in-
oculation of PRRSV followed by inoculation of a second rep-
licating agent. In addition, we believe that the combination of
PRRSV and LPS has practical relevance. Most pigs become
infected with PRRSV between 4 and 16 weeks of age, and the
virus persists in the lungs for up to 40 days after inoculation (9;
B. Mateusen, D. Maes, H. Nauwynck, B. Balis, M. Verdonck,
and A. de Kruif, Proc. 17th IPVS Congr., vol. 2, p. 240, 2002).
Also, most pigs are exposed to LPS under farm conditions, as
LPS is present in stable dust at concentrations ranging up to
4.9 g/m
3
. Furthermore, LPS is released at high concentrations
in the lungs during pulmonary infections with gram-negative
bacteria (16, 27).
The proinflammatory cytokines interleukin-1 (IL-1), tumor
necrosis factor-(TNF-), and IL-6 are important mediators
of several respiratory diseases. IL-1 and TNF-are among the
first cytokines that are produced in the lungs during an infec-
tion. They cause infiltration and activation of leukocytes in the
lungs, increased microvascular permeability, and pulmonary
dysfunctions (3, 20, 25). IL-1 and TNF-also induce a cascade
of secondary cytokines, such as IL-6. IL-6 is a potent inducer of
acute-phase proteins in the liver (12). Although IL-6 is gener-
ally considered a proinflammatory cytokine, it also has some
anti-inflammatory properties (19). IL-6 can down-regulate the
production of IL-1 and TNF-and suppress their activities by
inducing the production of IL-1 receptor antagonists and sol-
uble TNF-receptors. Furthermore, the production of each of
* Corresponding author. Mailing address: Laboratory of Virology,
Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133,
B-9820 Merelbeke, Belgium. Phone: 00 32 9 264 73 66. Fax: 00 32 9 264
74 95. E-mail: kristien.vanreeth@rug.ac.be.
960
the three cytokines in the lungs has been associated with gen-
eral signs of disease such as fever, depression, and anorexia.
The present study was undertaken to test the hypothesis that
PRRSV-LPS-induced respiratory disease is associated with the
excessive production of proinammatory cytokines in the
lungs. Therefore, we compared the levels of production of
IL-1, TNF-, and IL-6 in the lungs of pigs after dual inocula-
tion of PRRSV and LPS with those after the inoculation of
each agent alone. Correlations between cytokine levels and
respiratory signs, macroscopic and microscopic lung patholo-
gies, and the inltration of inammatory cells in the bronchoal-
veolar spaces were examined.
MATERIALS AND METHODS
Virus and LPS preparations. PRRSV (Lelystad virus strain) (26) was used in
the present study. The virus used for inoculation was at the fth passage in
alveolar macrophages, which had been obtained from 4- to 6-week-old gnotobi-
otic pigs. The inoculation dose was 10
6
50% tissue culture infective doses per pig.
E. coli O111:B4 LPS was obtained from Difco Laboratories (Detroit, Mich.) and
was used at a dose of 20 g/kg of body weight. This dose was based on data from
earlier experiments and was selected because it caused no clinical disease and
minimal IL-1 and TNF-secretion into the lungs (24). Virus and LPS were
diluted in sterile pyrogen-free phosphate-buffered saline (PBS; Gibco, Merel-
beke, Belgium) to obtain a 3-ml inoculum.
Pigs, experimental design, and sampling. Thirty-eight colostrum-deprived pigs
(age, 4 weeks) delivered by cesarean section were used in the study. They were
housed in individual Horsefall-type isolation units with positive-pressure venti-
lation and were fed commercial ultrahigh-temperature-treated cows milk. All
inoculations were performed intratracheally with an 18-gauge needle that was
inserted through the skin cranial to the sternum.
The pigs were allocated to four groups (see Table 1). Fourteen pigs were
inoculated with PRRSV; and 3 (n2),5(n3),7(n6), 10 (n2), or 14
(n1) days later they were inoculated with LPS (PRRSV-LPS group). These
pigs were euthanatized 6 h after the LPS inoculation. This time point was chosen
because previous virus-LPS experiments showed that cytokine production peaks
at 3 to 8 h after the LPS inoculation and declines afterwards (24). Fourteen pigs
were inoculated exclusively with PRRSV and euthanatized at 3 (n3),5(n
3),7(n3), 10 (n4), and 14 (n1) days after inoculation (PRRSV control
group). Five pigs were inoculated exclusively with LPS and euthanatized 6 h later
(LPS control group). Five pigs were mock inoculated with PBS and euthanatized
6 h later (PBS control group). All pigs were clinically monitored until euthanasia.
Samples from the left lung were collected for virological, histopathological,
and standard bacteriological examinations. The right lung was used for lung
lavage by a method described earlier (22). The brochoalveolar lavage (BAL)
uids that were recovered were separated into cells and cell-free uids by
centrifugation (400 g, 10 min, 4°C). For four of the six pigs that were inocu-
lated with PRRSV and then with LPS 7 days later, both the left and right lungs
were used for lung lavage.
Clinical and pathological examinations. Pigs were monitored for clinical signs
daily throughout the experiment and every hour after the LPS inoculation. A
respiratory disease score was attributed to each pig at the time of euthanasia.
Scores ranged from 0 to 4 (0, normal; 1, tachypnea when stressed; 2, tachypnea
at rest; 3, tachypnea and dyspnea at rest; 4, severe tachypnea and dyspnea with
labored, jerky breathing).
Macroscopic lung lesions were evaluated by visual inspection. For histopatho-
logical examination, samples of the cardiac and diaphragmatic lung lobes were
xed in 10% neutral buffered formalin, embedded in parafn, sectioned, and
stained with hematoxylin-eosin.
BAL uid cells were counted in a Tu¨rk chamber, and cytocentrifuge prepa-
rations were stained with Diff-Quik (Baxter, Du¨dingen, Switzerland) to deter-
mine the percentage of neutrophils and mononuclear cells.
Cytokine bioassays. Cell-free BAL uids were concentrated 20 times by dial-
ysis against a 20% (wt/vol) solution of polyethylene glycol (molecular weight,
20,000) and cleared of residual virus by centrifugation at 100,000 gbefore
analysis in bioassays for cytokines. Bioassays for IL-1, IL-6, and TNF-have
been described in detail elsewhere (7, 23).
IL-1 was assayed by determination of its capacity to stimulate proliferation of
D10(N4)M cells in the presence of concanavalin A (grade IV; Sigma, Bornem,
Belgium) and recombinant human IL-2 (Genzyme, Cambridge, Mass.). The
percentage of proliferation was determined by the thiazolyl blue [3-(4,5-dimeth-
ylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] conversion procedure, and op-
tical densities were measured. The number of biological units per milliliter of
BAL uid was determined as the dilution that produced 50% maximal prolifer-
ation. To conrm the specicity of the bioassay, D10 cells were incubated with
monoclonal rat anti-mouse IL-1 receptor type 1 antibodies (Genzyme).
TNF-activity was measured in a cytotoxicity assay with porcine kidney (PK)
(15) subclone 15 cells (a gift from G. Bertoni, Bern, Switzerland) in the presence
of actinomycin D. The plates were stained with crystal violet and read spectro-
photometrically. The number of biological units per milliliter was dened as the
dilution that produced 50% cytotoxicity. Specicity was demonstrated by neu-
tralization of samples with rabbit anti-human TNF-antibodies (Innogenetics,
Zwijnaarde, Belgium).
IL-6 was assayed by determination of its capacity to stimulate proliferation of
B9 cells (a gift from L. A. Aarden, Amsterdam, The Netherlands). Percent
proliferation was determined by the thiazolyl blue [3-(4,5-dimethylthiazol-2-yl)-
2,5-diphenyltetrazolium bromide] conversion procedure, and optical densities
were measured. The number of biological units per milliliter of BAL uid was
determined as the dilution that produced 50% maximal proliferation. To conrm
TABLE 1. Respiratory scores, virus titers, and numbers of inammatory cells in BAL uids
Inoculum No. of
pigs
Time of euthanasia after
inoculation with: Mean respiratory
score
a
Mean virus titer
(log
10
TCID
50b
/g)
Mean no. (10
6
) of BAL cells
PRRSV (days) LPS (h) Neutrophils Mononuclear cells
PBS 5
c
0 Negative 2 2 115 52
PRRSV 3 3 0 4.4 1.1 5 8 154 115
350 5.4 1.3 4 3 117 42
370 6.0 0.6 7 2 216 71
4100 5.7 0.9 38 38 409 219
1140 6.0 3 337
LPS 5 6 0 Negative 303 105 233 60
PRRSV-LPS 2 3 6 2 1.4 6.0 01823 121 22
356 30 5.1 2.2 296 163 208 14
6 7 6 3.2 0.8 5.5 0.3 320 263 275 144
2 10 6 2.5 0.7 5.9 1.3 380 474 486 142
1 14 6 3 5.7 576 483
a
Respiratory scores range from 0 to 4 (see text for the denition of each score).
b
TCID
50
, 50% tissue culture infective dose.
c
, not applicable.
VOL. 41, 2003 PRRSV POTENTIATES ENDOTOXIN RESPIRATORY EFFECTS 961
the specicity of the bioassay, samples were neutralized with goat anti-porcine
IL-6 antibodies (R&D Systems, Abingdon, United Kingdom).
Bioassays were done with twofold dilutions of samples in 96-well microtitra-
tion plates. Laboratory standards were run in each bioassay. Samples were tested
in two or three individual bioassays, and the geometric means were calculated.
Virological and bacteriological examinations. Tissue samples from the dia-
phragmatic lobe of the left lung were used for virological and bacteriological
examinations. PRRSV titrations were performed on alveolar macrophages by
standard methods (26). For bacteriology, samples of lung tissue were plated on
bovine blood agar and cultured aerobically. A nurse colony of coagulase-positive
Staphylococcus species was streaked diagonally on each plate. The plates were
inspected for bacterial growth after 48 and 72 h. The colonies were then iden-
tied by standard techniques.
Statistical analysis. Standard two-sample Mann-Whitney tests were used to
compare respiratory disease scores and cytokine titers. Correlation coefcients
were calculated by the Spearman rank correlation test. Pvalues 0.05 were
considered signicant. Statistical analyses were performed by using SPSS (ver-
sion 6.1) software.
RESULTS
The lungs of all pigs were free of bacteria by culture.
PRRSV titers are presented in Table 1. PRRSV was isolated
from the lungs of all virus-inoculated pigs but not from pigs
inoculated with LPS or PBS only. There were no differences in
virus titers between the pigs inoculated with PRRSV and LPS
combined and the pigs inoculated with PRRSV alone or at the
different time points after inoculation of PRRSV.
Clinical signs. Mean respiratory scores are presented in
Table 1. Pigs that received PBS or LPS only remained asymp-
tomatic. Pigs inoculated with PRRSV only showed no respira-
tory signs at any day after inoculation. They showed mild
anorexia and dullness between 3 and 5 days after inoculation.
In contrast, all pigs inoculated with PRRSV-LPS developed
marked respiratory signs. All pigs were clinically normal before
the LPS inoculation but developed tachypnea, dyspnea with
labored, abdominal breathing, and depression within 1 to 2 h
after LPS. These signs were still present at the time of eutha-
nasia. There were no differences in disease severity among the
pigs inoculated with LPS at 3, 5, 7, 10, or 14 days after inoc-
ulation of PRRSV. Respiratory disease scores were signi-
cantly (P0.05) higher for the group inoculated with PRRSV-
LPS than for any other group.
Macroscopic and microscopic lung pathologies. PBS-treated
control pigs did not have macroscopic or microscopic lung
pathologies (Fig. 1). The lungs of PRRSV-inoculated pigs had
a mottled appearance with multifocal red and tan areas. Mul-
tifocal interstitial pneumonia was found microscopically. Inter-
FIG. 1. Hematoxylin-eosin staining of the lungs of pigs inoculated with PBS only (a), LPS only (b), PRRSV only (10 days after inoculation) (c),
and PRRSV and then LPS over a 10-day interval (d). Interalveolar septal thickening was comparable in pigs inoculated with PRRSV-LPS and pigs
inoculated with PRRSV alone. Magnications, 100.
962 VAN GUCHT ET AL. J. CLIN.MICROBIOL.
alveolar septal thickening with inltration of mononuclear cells
was the major feature and increased from 3 to 14 days after
PRRSV inoculation. Inoculation with LPS only resulted in
milder pneumonic lesions. Macroscopic lesions were charac-
terized by focal areas of atelectasis and interlobular edema.
The characteristic histopathological features were thickening
of the interalveolar septa, although it was less pronounced than
that after inoculation with PRRSV, and bronchiolar inltra-
tion with neutrophils and macrophages. Intra-alveolar edema
and focal transudation of erythrocytes were occasionally seen.
The macroscopic and microscopic lung lesions after
PRRSV-LPS inoculation resembled the combination of the
lesions seen after inoculation of PRRSV or LPS alone. The
lungs were mottled with small red and tan areas and interlob-
ular edema. Microscopically, there was thickening of the inter-
alveolar septa due to an inltration of mononuclear cells and
neutrophils. The degree of septal thickening was comparable
to that seen after inoculation of PRRSV only.
Inltration of inammatory cells. The BAL cells of the
PBS-treated control pigs consisted mainly of mononuclear cells
(mean, 115 10
6
cells) and a few neutrophils (mean, 2 10
6
cells) (Table 1). PRRSV-inoculated pigs showed an inux of
mononuclear cells in the bronchoalveolar spaces, and this in-
ux increased from 3 to 14 days after inoculation. Starting at 7
days after inoculation with PRRSV, the mean number of
mononuclear cells was at least two times higher in PRRSV-
inoculated pigs than in PBS-treated control pigs. The number
of neutrophils in all except one of the PRRSV-inoculated pigs
was comparable to that in PBS-treated control pigs; one
PRRSV-inoculated pig had 91 10
6
neutrophils. The LPS
inoculation induced inltration of both neutrophils (mean, 303
10
6
cells) and mononuclear cells (mean, 233 10
6
cells).
Pigs inoculated with PRRSV-LPS showed an inux of both
mononuclear cells and neutrophils. The amount and kinetics of
mononuclear cell inltration in the pigs inoculated with
PRRSV-LPS were comparable to those in the PRRSV-inocu-
lated control pigs. The neutrophil numbers in the pigs inocu-
lated with PRRSV-LPS, on the other hand, were generally
comparable to those in the LPS-inoculated control pigs. Only
3 of the 14 pigs inoculated with PRRSV-LPS had larger num-
bers of neutrophils (567 10
6
to 786 10
6
) than the LPS-
inoculated control pigs. One pig inoculated with PRRSV-LPS
showed no neutrophil inltration at all (1 10
6
), and three
pigs inoculated with PRRSV-LPS showed only minor neutro-
phil inltration (19 10
6
to 44 10
6
) compared to that
detected in the LPS-inoculated control pigs. Two of these pigs
were inoculated with LPS 3 days after PRRSV inoculation,
which explains the low mean number of neutrophils in this
group.
Biologically active IL-1, TNF-, and IL-6 in BAL uids.
Figure 2 shows the IL-1, TNF-, and IL-6 titers in the BAL
uids of individual pigs after inoculation of PRRSV-LPS,
PRRSV only, and LPS only. PBS-treated control pigs had no
detectable IL-1, TNF-, or IL-6. Ten of 14 PRRSV-inoculated
pigs had elevated IL-1 titers, with the highest titers (183 to 339
U/ml) detected 10 days after inoculation. Only 3 of these 14
pigs (which were euthanatized 7, 10, and 14 days after inocu-
lation, respectively) had detectable TNF-titers (28 to 61
U/ml). Ten pigs had detectable IL-6 titers (61 to 343 U/ml).
LPS inoculation induced the production of all three cytokines
in the lungs. IL-1 (titers, 28 to 1,022 U/ml) and IL-6 (titers,
1,276 to 2,659 U/ml) were detected in all ve pigs, and TNF-
(titers, 28 to 133 U/ml) was detected in three pigs.
Compared to the pigs inoculated with PRRSV and LPS
alone, 10 of 14 pigs inoculated with PRRSV-LPS showed sig-
nicantly (P0.05) increased titers of at least one of the three
cytokines. In nine pigs the titers of IL-1 (titers, 2,172 to 20,480
U/ml), TNF-(titers, 164 to 6,047 U/ml), and IL-6 (titers,
2,511 to 378,724 U/ml) were strongly increased; and in one pig
only the titer of IL-1 (2,840 U/ml) was increased. The highest
cytokine titers were detected in pigs inoculated with LPS 5 to
14 days after the PRRSV inoculation, and they were 10 to 100
times higher than the cytokine titers of the control pigs inoc-
ulated with PRRSV or LPS only. Four pigs inoculated with
FIG. 2. Titers of proinammatory cytokines in BAL uids of
PRRSV-LPS-inoculated pigs and pigs inoculated with PRRSV only or
LPS only. Each dot corresponds to one pig: F, pigs inoculated with
LPS at the indicated day after PRRSV inoculation; E, pigs inoculated
with PRRSV only; , pigs inoculated with LPS only. The dotted line
represents the detection limit.
VOL. 41, 2003 PRRSV POTENTIATES ENDOTOXIN RESPIRATORY EFFECTS 963
PRRSV-LPS, on the other hand, did not show enhanced levels
of cytokine production. These pigs had negligible levels of
TNF-(titers, 20 to 31 U/ml), and the levels of IL-1 (titers,
191 to 1,571 U/ml) and IL-6 (titers, 266 to 2425 U/ml) were
comparable to those for the LPS-treated control pigs.
The left and right lungs of pigs that were inoculated with
PRRSV-LPS and whose lungs were lavaged showed no differ-
ence in cytokine titers or cell counts (P0.05) (data not
shown).
Table 2 presents the correlation between respiratory scores,
cytokine levels, and numbers of inammatory cells in BAL
uids. The levels of all three cytokines were tightly correlated
with each other and with the respiratory scores and the neu-
trophil numbers. There was, however, little correlation be-
tween neutrophil numbers and respiratory scores. The number
of inltrated mononuclear cells did not correlate with cytokine
levels or respiratory scores. The four pigs inoculated with
PRRSV-LPS that did not have increases in cytokine levels also
had lower neutrophil numbers (1 10
6
to 129 10
6
). The
cytokine titers and BAL cell numbers did not correlate with the
virus titers (data not shown).
DISCUSSION
This study demonstrates that a PRRSV infection sensitizes
the lungs for production of proinammatory cytokines upon
exposure to LPS. Moreover, the cytokine titers were tightly
correlated with the appearance of respiratory signs. We have
previously documented a similar phenomenon for another re-
spiratory virus of swine that causes subclinical disease, porcine
respiratory coronavirus (PRCV) (24). Like PRRSV, PRCV
infection enhanced the levels of production of TNF-and IL-1
in response to LPS, and the levels of both cytokines correlated
with the severity of disease. The pathogenesis of PRRSV-LPS-
induced disease appears to be similar to the pathogenesis of
PRCV-LPS-induced disease. As IL-1 and TNF-have over-
lapping effects and potentiate the effects of each other, we
consider them both to be central mediators in virus-LPS-in-
duced disease. IL-6 levels in the lungs of pigs inoculated with
both virus and LPS were assessed for the rst time in the
present study, and they were also found to be markedly en-
hanced. IL-6 is probably induced as a secondary cytokine in
response to IL-1 and TNF-, which may explain the tight
correlation between IL-6 levels and IL-1 and TNF-levels.
Because IL-6 has both pro- and anti-inammatory activities, it
may either contribute to disease or counteract the activities of
IL-1 and TNF-.
We have indications that the tachypnea and dyspnea result-
ing from PRRSV-LPS or PRCV-LPS inoculations are due to a
functional process, such as bronchoconstriction, rather than to
structural lung damage. First, the onset of respiratory signs is
hyperacute. In another study of PRRSV-LPS inoculation, it
was shown that respiratory signs started within 1 h after LPS
inoculation, reached a climax 2 to 4 h later, and were clearly
diminished 12 h later (8). Second, the microscopic lesions in
the lungs of pigs inoculated with PRRSV-LPS and with
PRRSV only did not differ much. Pigs in both groups had
interstitial pneumonia typical of PRRSV infection, and LPS
inoculation had little extra effect. The inoculation with LPS as
such caused a marked increase in the numbers of neutrophils
in BAL uids, but there were no differences in neutrophil
numbers between pigs inoculated with PRRSV-LPS and those
inoculated with LPS alone. Third, it is well known that IL-1
and TNF-can cause bronchial hyperreactivity (2, 15) and
bronchoconstriction (10), leading to asthma-like symptoms.
Moreover, TNF-and IL-1 were shown to act synergistically in
the induction of bronchoconstriction in the rat lung (10).
Therefore, simultaneous overproduction of these cytokines af-
ter PRRSV-LPS inoculation may cause increased and sus-
tained contraction of bronchi, which may explain the acute
respiratory signs.
We cannot explain why four pigs inoculated with PRRSV-
LPS, which showed clear respiratory signs, had only low cyto-
kine titers and negligible neutrophil inltration. There were no
consistent differences in PRRSV titers or the numbers of
mononuclear cells in BAL uids between these and the other
pigs. Because LPS exerts its effect locally, our initial hypothesis
was that the LPS inoculum probably did not reach the right
lung in those pigs and that cytokine production and neutrophil
inltration might have been restricted to the left lung. To test
this hypothesis we lavaged both the left and the right lungs of
four pigs inoculated with PRRSV-LPS. There were no differ-
ences in the levels of cytokine production or neutrophil inl-
tration between the two lung halves. Therefore, it can be as-
sumed that the LPS inoculum is distributed equally between
both lung halves in most pigs. The true reason for the variabil-
ity in cytokine production and neutrophil inltration among
pigs inoculated with PRRSV-LPS is unclear.
There have been few studies on the interactions between
viruses and LPS in vivo. To our knowledge, PRRSV and
PRCV are the rst respiratory viruses shown to act synergis-
tically with LPS in the induction of respiratory disease and
cytokines. Recently, it has been described that systemic infec-
TABLE 2. Correlation coefcients between respiratory scores, cytokine titers, and numbers of inammatory cells in BAL uids
Parameter Correlation with:
Respiratory score IL-1 titer TNF-titer IL-6 titer Neutrophils no. Mononuclear cell no.
Respiratory score 11 0.81 0.70 0.71 0.59 NS
a
IL-1 titer
b
1 0.75 0.85 0.80 0.43
TNF-titer ——1 0.84 0.74 0.40
IL-6 titer ——1 0.84 NS
Neutrophil no. ——— 1 0.61
Mononuclear cell no. ——— 1
a
NS, no signicant correlation (P0.05).
b
, not applicable.
964 VAN GUCHT ET AL. J. CLIN.MICROBIOL.
tion of mice with lymphocytic choriomeningitis virus or vesic-
ular stomatitis virus leads to fatal shock upon intraperitoneal
inoculation of a sublethal dose of LPS (13, 14). It appeared
that the shock syndrome was caused by the overproduction of
TNF-. Mice inoculated with virus-LPS had 3- to 50-fold
higher serum TNF-levels compared to those in the sera of
mice inoculated with LPS alone. By use of knockout mice, it
was demonstrated that virus-induced interferon was responsi-
ble for the increased sensitivity to LPS (5, 13). Both alpha/beta
interferon and gamma interferon were able to sensitize cells to
LPS. It is unlikely, however, that alpha interferon is involved in
the sensitization of PRRSV-infected pigs to LPS, because al-
pha interferon production is minimal during infection with
PRRSV (1, 23).
It remains to be seen whether the PRRSV-induced inltra-
tion of the lungs with mononuclear cells contributes to the
increased responsiveness to LPS. PRRSV induces inltration
of monocytes in the lungs, reaching a peak at 25 days after
inoculation (9). In mice, it was shown that monocytes inltrat-
ing the lungs in response to monocyte chemoattractant protein
type 1 (MCP-1) have increased levels of expression of CD14,
the LPS receptor, and become primed for enhanced TNF-
production in response to LPS (11). It is possible that mono-
cytes attracted to PRRSV are an important source of cytokines
upon LPS exposure and that they are responsible for the en-
hanced cytokine response compared to the response of unin-
fected lungs. In this study, the number of mononuclear cells in
the bronchoalveolar spaces did not correlate with the respira-
tory signs. There are two important considerations in this re-
gard. First, the proles of the cells in the BAL uid of pigs
inoculated with PRRSV-LPS were partly the result of the LPS
inoculation and, as such, did not reect the situation before the
LPS inoculation. Second, we counted the mononuclear cells in
the BAL uids and not in the interstitium, while interstitial
monocytes may be important targets for LPS.
In conclusion, respiratory viruses like PRRSV, which do not
cause respiratory signs on their own, can sensitize the lungs for
the production of proinammatory cytokines and respiratory
signs upon exposure to bacterial endotoxins. This interaction
may be important in the development of multifactorial respi-
ratory disease, as is often seen in the eld.
ACKNOWLEDGMENTS
This work was supported by grant 5772A from the Belgian Ministry
of Agriculture. S.V.G. and K.V.R. are fellows of the Fund for Scientic
Research Flanders (FWO-Vlaanderen).
We thank Lieve Sys, Fernand De Backer, and Chantal Vanmaercke
for excellent technical assistance. We also thank G. Bertoni for pro-
viding recombinant porcine TNF-and PK(15) subclone 15 cells and
L. A. Aarden for providing B9 cells.
REFERENCES
1. Albina, E., C. Carrat, and B. Charley. 1998. Interferon-alpha response to
swine arterivirus (PoAV), the porcine reproductive and respiratory syn-
drome virus. J. Interferon Cytokine Res. 18:485490.
2. Anticevich, S. Z., J. M. Hughes, J. L. Black, and C. L. Armour. 1995.
Induction of human airway hyperresponsiveness by tumor necrosis factor-
alpha. Eur. J. Pharmacol. 284:221225.
3. Bielefeldt-Ohmann, H. 1995. Role of cytokines in the pathogenesis and
treatment of respiratory disease, p. 291332. In M. J. Myers and M. P.
Murtaugh (ed.), Cytokines in animal health and disease. Marcel Dekker,
Inc., New York, N.Y.
4. Brockmeier, S. L., M. V. Palmer, and S. R. Bolin. 2000. Effects of intranasal
inoculation of porcine reproductive and respiratory syndrome virus, Borde-
tella bronchiseptica, or a combination of both organisms in pigs. Am. J. Vet.
Res. 61:892899.
5. Doughty, L. A., K. B. Nguyen, J. E. Durbin, and C. A. Biron. 2001. A role for
IFN-␣␤ in virus infection-induced sensitization to endotoxin. J. Immunol.
166:26582664.
6. Galina, L., C. Pijoan, M. Sitjar, W. T. Christianson, K. Rossow, and J. E.
Collins. 1994. Interaction between Streptococcus suis serotype 2 and porcine
reproductive and respiratory syndrome virus in specic pathogen-free pig-
lets. Vet. Rec. 134:6064.
7. Helle, M., L. Boeije, and L. A. Aarden. 1988. Functional discrimination
between interleukin 6 and interleukin 1. Eur. J. Immunol. 18:15351540.
8. Labarque, G., K. Van Reeth, S. Van Gucht, H. Nauwynck, and M. Pensaert.
2002. Porcine reproductive-respiratory syndrome virus (PRRSV) infection
predisposes pigs for respiratory signs upon exposure to bacterial lipopoly-
saccharide. Vet. Microbiol. 88:112.
9. Labarque, G. G., H. J. Nauwynck, K. Van Reeth, and M. B. Pensaert. 2000.
Effect of cellular changes and onset of humoral immunity on the replication
of porcine reproductive and respiratory syndrome virus in the lungs of pigs.
J. Gen. Virol. 81:13271334.
10. Martin, C., A. Wohlsen, and S. Uhlig. 2001. Changes in airway resistance by
simultaneous exposure to TNF-and IL-1in perfused rat lungs. Am. J.
Physiol. Lung Cell. Mol. Physiol. 280:L595L601.
11. Maus, U., S. Herold, H. Muth, R. Maus, L. Ermert, M. Ermert, N. Weiss-
man, S. Rosseau, W. Seeger, F. Grimminger, and J. Lohmeyer. 2001. Mono-
cytes recruited into the alveolar air space of mice show a monocytic pheno-
type but upregulate CD14. Am. J. Physiol. Lung Cell. Mol. Physiol. 280:L58
L68.
12. Murtaugh, M. P., M. J. Baarsch, Y. Zhou, R. W. Scamurra, and G. Lin. 1996.
Inammatory cytokines in animal health and disease. Vet. Immunol. Immu-
nopathol. 54:4555.
13. Nansen, A., and A. R. Thomsen. 2001. Viral infection causes rapid sensiti-
zation to lipopolysaccharide: central role of IFN-␣␤. J. Immunol. 166:982
988.
14. Nguyen, K. B., and C. A. Biron. 1999. Synergy for cytokine-mediated disease
during concurrent endotoxin and viral challenges: roles for NK and T cell
IFN-production. J. Immunol. 162:52385246.
15. Okada, S., H. Inoue, K. Yamauchi, H. Iijima, Y. Ohkawara, T. Takishima,
and K. Shirato. 1995. Potential role of interleukin-1 in allergen-induced late
asthmatic reactions in guinea pigs: suppressive effect of interleukin-1 recep-
tor antagonist on late asthmatic reaction. J. Allergy Clin. Immunol. 95:1236
1245.
16. Pugin, J., R. Auckenthaler, O. Delaspre, E. Van Gessel, and P. M. Suter.
1992. Rapid diagnosis of gram negative pneumonia by assay of endotoxin in
bronchoalveolar lavage uid. Thorax 47:547549.
17. Thacker, E. L., P. G. Halbur, R. F. Ross, R. Thanawongnuwech, and B. J.
Thacker. 1999. Mycoplasma hyopneumoniae potentiation of porcine repro-
ductive and respiratory syndrome virus-induced pneumonia. J. Clin. Micro-
biol. 37:620627.
18. Thacker, E. L. 2001. Immunology of the porcine respiratory disease complex.
Vet. Clin. N. Am. Food Anim. Pract. 17:551565.
19. Tilg, H., E. Trehu, M. B. Atkins, C. A. Dinarello, and J. W. Mier. 1994.
Interleukin-6 (IL-6) as an anti-inammatory cytokine: induction of circulat-
ing IL-1 receptor antagonist and soluble tumor necrosis factor receptor p55.
Blood 83:113118.
20. Ulich, T. R., L. R. Watson, S. M. Yin, K. Z. Guo, P. Wang, H. Thang, and J.
Del Castillo. 1991. The intratracheal administration of endotoxin and cyto-
kines. I. Characterization of LPS-induced TNF mRNA expression and the
LPS-, IL-1-, and TNF-induced inammatory inltrate. Am. J. Pathol. 138:
14851496.
21. Van Reeth, K., H. J. Nauwynck, and M. B. Pensaert. 1996. Dual infections of
feeder pigs with porcine reproductive and respiratory syndrome virus fol-
lowed by porcine respiratory coronavirus or swine inuenza virus: a clinical
and virological study. Vet. Microbiol. 48:325335.
22. Van Reeth, K., H. Nauwynck, and M. Pensaert. 1998. Broncho-alveolar
interferon-, tumor necrosis factor-, interleukin-1 and inammation during
acute inuenza in pigs: a possible model for humans? J. Infect. Dis. 177:
10761079.
23. Van Reeth, K., G. Labarquel, H. Nauwynck, and M. Pensaert. 1999. Differ-
ential production of proinammatory cytokines in the pig lung during dif-
ferent respiratory virus infections: correlations with pathogenicity. Res. Vet.
Sci. 67:4752.
24. Van Reeth, K., H. Nauwynck, and M. Pensaert. 2000. A potential role for
tumor necrosis factor-in synergy between porcine respiratory coronavirus
and bacterial lipopolysaccharide in the induction of respiratory disease in
pigs. J. Med. Microbiol. 49:613620.
VOL. 41, 2003 PRRSV POTENTIATES ENDOTOXIN RESPIRATORY EFFECTS 965
25. Wang, Z., K. Larsson, L. Palmberg, P. Malmberg, P. Larsson, and L. Lars-
son. 1997. Inhalation of swine dust induces cytokine release in the upper and
lower airways. Eur. Respir. J. 10:381387.
26. Wensvoort, G., C. Terpstra, J. M. A. Pol, E. A. ter Laak, M. Bloemraad, E. P.
de Kluyver, C. Kragten, L. van Buiten, A. den Besten, F. Wagenaar, J. M.
Broekhuijsen, P. L. J. M. Moonen, T. Zetstra, E. A. de Boer, H. J. Tibben,
M. F. de Jong, P. vant Veld, G. J. R. Groenland, J. A. van Gennep, M. T.
Voets, J. H. M. Verheijden, and J. Braamskamp. 1991. Mystery swine disease
in The Netherlands: the isolation of Lelystad virus. Vet. Q. 13:121130.
27. Zhiping, W., P. Malmberg, K. Larsson, L. Larsson, and A. Saraf. 1996.
Exposure to bacteria in swine-house dust and acute inammatory reactions
in humans. Am. J. Respir. Care Med. 154:12611266.
966 VAN GUCHT ET AL. J. CLIN.MICROBIOL.
... The selection of disease-resistant pig phenotypes, differing from the lean type, highly susceptible ones [13]. The high levels of oxidative stress in such pigs [14] are likely to exacerbate the inflammatory responses to infectious and non-infectious stressors and, in particular, the noxious synergism between bacterial LPS and PRRSV infection [15]. This is probably a point of some importance, since LPS can also be inhaled at high concentrations in pig herds [16], and circumstantial evidence on farm showed clinical improvement in PRRSV-infected groups after reduction of animal concentration in outdoor weaning cages. ...
... This is fully in line with in vivo findings of PRRSV infections leading to serious clinical outcomes; these are correlated with enhanced inflammatory cytokine responses, but not with the extent of viral replication [31]. These results hint at a major upregulation of the inflammatory response following high-titered replication of PRRSV in virus-permissive pig macrophages and at a possible synergism with inflammatory stressors such as LPS [15]. Instead, virulent PRRSV strains do not exert this function in non-permissive cells; this makes sense in order to avoid any subsequent restriction of growth in differentiated pig macrophages [11]. ...
Article
Full-text available
The control of porcine reproductive and respiratory syndrome (PRRS) is still a major issue worldwide in the pig farming sector. Despite extensive research efforts and the practical experience gained so far, the syndrome still severely affects farmed pigs worldwide and challenges established beliefs in veterinary virology and immunology. The clinical and economic repercussions of PRRS are based on concomitant, additive features of the virus pathogenicity, host susceptibility, and the influence of environmental, microbial, and non-microbial stressors. This makes a case for integrated, multi-disciplinary research efforts, in which the three types of contributing factors are critically evaluated toward the development of successful disease control strategies. These efforts could be significantly eased by the definition of reliable markers of disease risk and virus pathogenicity. As for the host’s susceptibility to PRRSV infection and disease onset, the roles of both the innate and adaptive immune responses are still ill-defined. In particular, the overt discrepancy between passive and active immunity and the uncertain role of adaptive immunity vis-à-vis established PRRSV infection should prompt the scientific community to develop novel research schemes, in which apparently divergent and contradictory findings could be reconciled and eventually brought into a satisfactory conceptual framework.
... -The selection of disease-resistant pig phenotypes, differing from the lean type, highly susceptible ones [11]. The high levels of oxidative stress in such pigs [12] are likely to exacerbate the inflammatory responses to infectious and non-infectious stressors and, in particular, the noxious synergism between bacterial LPS and PRRSV infection [13]. This is probably a point of some importance, since LPS can be also inhaled at high concentrations in pig herds [14], and circumstantial evidence on farm showed clinical improvement in PRRSV-infected groups after reduction of the animals' concentration in outdoor weaning cages. ...
... This is fully in line with in vivo findings of PRRSV infections leading to serious clinical outcomes: these are correlated with enhanced inflammatory cytokine responses, but not with the extent of viral replication [29]. These results hint at a major up-regulation of the inflammatory response following high-titered replication of PRRSV in virus-permissive pig macrophages and at a possible synergism with inflammatory stressors like LPS [13]. Instead, virulent PRRSV strains do not exert this activity in non-permissive cells; this makes sense in order to avoid any subsequent restriction of growth in differentiated pig macrophages [9]. ...
Preprint
Full-text available
The control of Porcine Reproductive and Respiratory Syndrome (PRRS) is still a major issue worldwide in the pig farming sector. Despite extensive research efforts and the practical experience gained so far, the syndrome still heavily affects farmed pigs worldwide and challenges established beliefs in veterinary virology and immunology. The clinical and economic repercussions of PRRS are based on concomitant, additive features of virus pathogenicity, host susceptibility and influence of environmental, microbial and non-microbial stressors. This makes a case for integrated, multi-disciplinary research efforts in which the three types of contributing factors are critically evaluated toward the development of successful disease control strategies. These could be definitely eased by the definition of reliable markers of disease risk and virus pathogenicity. As for the host’s susceptibility to PRRSV infection and disease onset, the roles of both innate and adaptive immune responses are still ill-defined. In particular, the overt discrepancy between passive and active immunity and the uncertain role of adaptive immunity vis-à-vis an established PRRSV infection should prompt the scientific community to the development of novel research schemes, in which apparently diverging and contradictory findings could be reconciled, and eventually brought to a satisfactory conceptual framework.
... It may increase the intensity of symptoms of mild respiratory viruses and lead to fatal fibrinous pneumonia (32). This synergistic interaction between respiratory viruses and bacterial LPS is examined in different studies (137)(138)(139). ...
Article
The emergence of the novel human coronavirus (SARS-CoV-2) has been one of the most overwhelming human challenges of recent decades. Despite global efforts to stop or slow down the spread of the disease, it has infected millions of people across the world within a short period of time. Research on human coronaviruses has not been received adequate attention and there is currently no specific treatment or vaccine available for coronaviruses in humans. Interestingly, evolution, pathogenesis, and immunogenicity of coronaviruses in livestock and poultry have been described for many years and they are controlled by mass vaccination programs. Moreover, elevating immune responses to coronaviruses, a major hurdle in virulent human-coronaviruses, has been studied by several investigators in cat populations. Indeed, cross-species transmission and tropism of animal coronaviruses have been investigated by several research groups. It is estimated that three out of every four new emerging infectious diseases are zoonotic. Preliminary reports indicate that like other human coronaviruses, SARS-CoV-2 has been transmitted from bats to humans by an intermediate animal host. Hence, interdisciplinary collaborations among veterinary and medical researchers and clinicians, biologists, and environmental scientists are essential for identifying the possible animal reservoir of the viruses in general and discovering the evolutionary pathway of SARS-CoV-2 in particular. This can also result in the designing of effective prevention and control measures against SARS-CoV-2. In this review, advances in the understanding of different features of animal coronaviruses that can facilitate effective measures in dealing with the SARS-CoV-2 pandemic have been discussed.
... The role of TNFα in PRRSV pathology is somewhat contradictory given that in some models there is very little induction [5], while in others show significant expression at certain time points [7]. Expression of TNFα has been positively correlated with severity of porcine respiratory disease [5,31]. IL-8 is a chemotactic, pro-inflammatory cytokine secreted on induction of apoptosis in bronchiolar epithelial cells. ...
Article
Full-text available
Background: The effect of a water-soluble formulation of tylvalosin (Aivlosin® 625 mg/g granules) on disease caused by porcine reproductive and respiratory syndrome virus (PRRSV) and Mycoplasma hyopneumoniae (Mhyop) was investigated in two animal studies. In a PRRSV challenge model in pregnant sows (n = 18), six sows received water medicated at target dose of 5 mg tylvalosin/kg body weight/day from 3 days prior to challenge until the end of gestation. Six sows were left untreated, with a third group remaining untreated and unchallenged. Sows were challenged with PRRSV-2 at approximately 85 days of gestation. Cytokines, viremia, viral shedding, sow reproductive parameters and piglet performance to weaning were evaluated. In a dual infection study (n = 16), piglets were challenged with Mhyop on days 0, 1 and 2, and with PRRSV-1 on day 14 and euthanized on day 24. From day 10 to 20, eight piglets received water medicated at target dose of 20 mg tylvalosin/kg body weight/day and eight piglets were left untreated. Cytokines, viremia, bacteriology and lung lesions were evaluated. Results: In the PRRSV challenge study in pregnant sows, tylvalosin significantly reduced the levels of serum IL-8 (P < 0.001), IL-12 (P = 0.032), TNFα (P < 0.001) and GM-CSF (P = 0.001). IL-8 (P = 0.100) tended to be lower in uterus of tylvalosin sows. All piglets from tylvalosin sows surviving to weaning were PRRSV negative in faecal swabs at weaning compared to 33.3% PRRSV positive piglets from untreated sows (P = 0.08). In the dual challenge study in piglet, tylvalosin reduced serum IL1β, IL-4, IL-6, IL-8, IL-10, IL-12, IL-1α, IL-13, IL-17A, IL-18, GM-CSF, TGFβ1, TNFα, CCL3L1, MIG, PEPCAM-1 (P < 0.001) and increased serum IFNα, IL-1ra and MIP-1b (P < 0.001). In the lungs, tylvalosin reduced IL-8, IL-10 and IL-12 compared to untreated pigs (P < 0.001) and tended to reduce TNFα (P = 0.082). Lung lavage samples from all tylvalosin treated piglets were negative for Mhyop (0 cfu/mL) compared to the untreated piglets which had mean Mhyop counts of 2.68 × 104 cfu/mL (P = 0.023). Conclusion: Overall, tylvalosin reduced both local and systemic proinflammatory cytokines after challenge with respiratory pathogens in sows and in piglets. Tylvalosin was effective in reducing Mhyop recovery from the lungs and may reduce virus shedding in piglets following transplacental PRRSV infection in sows.
... Thus, chronic exposure to high endotoxin (E. coli LPS) concentrations might sensitize the respiratory tract for damage upon exposure to a respiratory virus like IBV and increase disease susceptibility. Yet, lung lesions of Porcine Reproductive-Respiratory Syndrome Virus (PRRSV) infected pigs were little aggravated by subsequent LPS exposure (Van Gucht et al., 2003). Similarly, in the IBV vaccinated group no differences were found between HE and LE broilers for lesions in the trachea or lung. ...
Article
Full-text available
In and around poultry farms, high concentrations of endotoxins are found that have a negative impact on the health of farmers and local residents. However, little is known about the effects of chronic exposure to endotoxins on poultry health. The aim of this study was to identify effects of chronic exposure to airborne endotoxins (E. coli LPS) on the immune system, respiratory tract, disease susceptibility and welfare of broilers. Effects of high (HE) and low endotoxin (LE) concentrations on natural antibody titers (NAb), performance and behavior of broilers were determined. After treatment with a respiratory virus infection, infectious bronchitis virus (IBV), mRNA expression of cytokines and Toll-like receptor (TLR) 4 in the lung, tracheal ciliary activity and lesions in the respiratory tract were measured. Endotoxin affected the immune system and respiratory tract, where HE broilers tended to have lower IgM NAb binding Phosphorylcholine-conjugated to Bovine Serum Albumin, but higher interferon (IFN)-α mRNA expression and more lesions in the nasal tissue compared to LE broilers. Furthermore, HE broilers had higher TLR4 mRNA expression compared to LE broilers. However, endotoxin did not affect NAb levels binding Keyhole Limpet Hemocyanin, IFN-β and interleukin-10 mRNA expression, IBV replication or lesions in the lung and trachea. HE and LE broilers further had similar body weight, but HE broilers showed numerically more passive behavior compared to LE broilers. In conclusion, chronic exposure to high airborne endotoxin concentrations affects components of the immune system and respiratory tract in broilers and could therefore influence disease susceptibility.
... Pulmonary alveolar and/or intravascular macrophages are known as the major target cells for PRRSV in the lungs (58). The studies on PRRSV infection and replication in host alveolar macrophages continue to give conflicting results as to its effect on innate cell activation, antigen presentation, and the triggering of acquired immunity (20,21,23,25,27,59,60). It has been widely demonstrated that PRRSV causes apoptosis in different organs (lungs, testes, lymph nodes, and thymus) as well as in cell lines (5,6,11,22,24,48,51,52). On a morphological basis, the apoptotic cells are assumed to be alveolar and pulmonary intravascular macrophages and mononuclear cells in the alveolar septa. ...
Article
Porcine reproductive and respiratory syndrome virus (PRRSV) induces apoptosis through the activation of death receptors, including cell-surface Fas receptor. The aim of this study was to investigate the impact of intradermal (ID) and intramuscular (IM) vaccination with a commercial PRRSV-modified live vaccine in piglets on Fas-related apoptosis. The study included 104 suckling piglets from a commercial farrow-to-finish pig farm, suffering from positive unstable PRRSV status. Animals were assigned in four groups: group A-Porcilis PRRS ID-vaccinated pigs, group B-Porcilis PRRS IM-vaccinated pigs, group C-Diluvac ID adjuvant-administered pigs, and group D-Diluvac IM adjuvant-administered pigs. Vaccines were administered at 2 weeks of age. Blood samples were collected from the same pigs at 4, 7, and 10 weeks of age. Sera were examined by quantitative real-time reverse transcription-PCR (qRT-PCR) for PRRSV and by ELISA for soluble Fas (sFas). At 4 weeks of age, all groups were negative qRT-PCR for PRRSV; at 7 weeks only group A was negative; and at 10 weeks all groups were positive. sFas was significantly increased in groups C (4 vs. 7, 4 vs. 10, and 7 vs. 10 weeks) and D (7 vs. 10 weeks). Significant differences among groups were noticed only at 10 weeks (A vs. C, A vs. D, B vs. C, B vs. D). A significant positive and moderate correlation between PRRSV viral load and Fas level was observed. In unvaccinated piglets, increased serum sFas levels reveal apoptotic suppression compared with vaccinated piglets. In the latter, vaccine-derived antibodies limit the infection and may attribute to the reduced Fas expression, suggesting a weak induction of lymphocyte-mediated cytotoxicity.
... IFN-α can then induce IL-10 in LPS-stimulated monocytes and CD4+ T cells [54]. Interestingly, there is definitely a synergism in vivo between PRRSV and LPS [55], and an IL-10 response to some PRRSV strains was repeatedly observed in vitro by PRRS-naïve swine PBMC [56,57]. Last but not least, IFN-α and IL-10 can promote the differentiation of Type I T regulatory (Treg) cells [58]. ...
Article
Full-text available
Interferons (IFNs) are a family of cytokines that play a pivotal role in orchestrating the innate immune response during viral infections, thus representing the first line of defense in the host. After binding to their respective receptors, they are able to elicit a plethora of biological activities, by initiating signaling cascades which lead to the transcription of genes involved in antiviral, anti-inflammatory, immunomodulatory and antitumoral effector mechanisms. In hindsight, it is not surprising that viruses have evolved multiple IFN escape strategies toward efficient replication in the host. Hence, in order to achieve insight into preventive and treatment strategies, it is essential to explore the mechanisms underlying the IFN response to viral infections and the constraints thereof. Accordingly, this review is focused on three RNA and three DNA viruses of major importance in the swine farming sector, aiming to provide essential data as to how the IFN system modulates the antiviral immune response, and is affected by diverse, virus-driven, immune escape mechanisms.
... IL1B (IL-1b) and TNF genes were upregulated in the Lena group, unlike the 3249 group. While classical PRRSV strains are reported to show a delayed and weak systemic production of these cytokines (66)(67)(68)(69), this proinflammatory profile seems to be a hallmark of virulent PRRSV strains (24,25,39,58,59), resulting in being useless in controlling viral replication but exacerbating clinical signs and lung damage. ...
Article
Full-text available
Porcine reproductive and respiratory syndrome virus (PRRSV) has evolved to escape the immune surveillance for a survival advantage leading to a strong modulation of host’s immune responses and favoring secondary bacterial infections. However, limited data are available on how the immunological and transcriptional responses elicited by virulent and low virulent PRRSV-1 strains are comparable and how are them conserved along the infection. To explore the kinetic transcriptional signature associated with the modulation of host immune response at lung level a time-series transcriptomic analysis was performed in bronchoalveolar lavage cells upon experimental in vivo infection with two PRRSV-1 strains of different virulence, virulent subtype 3 Lena strain or the low virulent subtype 1 3249 strain. The time-series analysis revealed overlapping patterns of dysregulated genes enriched in T-cell signaling pathways among both virulent and low-virulent strains, highlighting an up-regulation of co-stimulatory and co-inhibitory immune checkpoints which were disclosed as Hub genes. On the other hand, virulent Lena infection induced an early and more marked “negative regulation of immune system process” with an overexpression of co-inhibitory receptors genes related to T-cell and NK cell functions, in association with more severe lung lesion, lung viral load and BAL cell kinetics. These results underline a complex network of molecular mechanisms governing PRRSV-1 immunopathogenesis at lung level, revealing a pivotal role of co-inhibitory and co-stimulatory immune checkpoints in the pulmonary disease, which may have an impact on T-cell activation and related-pathways. These immune checkpoints together with the regulation of cytokine-signaling pathways, modulated in a virulence-dependent fashion, orchestrate an interplay among pro- and anti-inflammatory responses. Importance: Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the major threats to swine health and global production, causing substantial economic losses. We explore the mechanisms involved in the modulation of host immune response at lung level performing a time-series transcriptomic analysis upon experimental infection with two PRRSV-1 strains of different virulence. A complex network of molecular mechanisms was revealed to control the immunopathogenesis of PRRSV-1 infection, highlighting an interplay among pro- and anti-inflammatory responses as a potential mechanism to restrict inflammation-induced lung injury. Moreover, a pivotal role of co-inhibitory and co-stimulatory immune checkpoints was evidenced, which may lead to progressive dysfunction of T cells, impairing viral clearance and leading to persistent infection, favoring as well secondary bacterial infections or viral rebound. Although further studies should be conducted to evaluate the functional role of immune checkpoints in advanced stages of PRRSV infection and explore a possible T-cell exhaustion state.
... Pathogens, particularly of the digestive tract and respiratory tract, are common sources of MAMPs and frequent causes of elevated cytokine levels in pigs (reviewed in [44]). Poor hygiene, dust, LPS, and high levels of ammonia in the air are regularly present in pig houses and lead to activation of the inflammatory cascade via the respiratory tract [72][73][74][75]. Pigs are exposed to massive stressors, including high housing density, lack of pen structure, regrouping, lack of opportunities to eat at the same time, disease, poor housing air, etc. [10,76,77]. ...
Article
Full-text available
Tail biting is a prevalent and undesirable behaviour in pigs and a major source of significant reduction in well-being. However, focusing on biting considers only one part of the solution, because tail damage can be found with a high prevalence without any action by other pigs. The lesions are not limited to the tail but can also be found in the ears, heels, soles, claw coronary bands, teats, navel, vulva, and face. Environmental improvement alone often fails to overcome the problem. This review addresses a new inflammation and necrosis syndrome in swine (SINS). It shows the clinical signs and the frequencies of occurrence in different age groups. It compiles scientific evidence from clinical and histopathological studies in newborn piglets that argue for a primary endogenous aetiology of the disease. Bringing together the findings of a broad body of research, the possible mechanisms leading to the disease are identified and then discussed. This part will especially focus on microbe-associated molecular patterns in the circulation and their role in activating defence mechanisms and inflammation. Finally, the methods are identified to ameliorate the problem by optimizing husbandry and selecting a suitable breeding stock.
Article
Full-text available
The objective of this study was to characterize the enzymatic hydrolysis of lipopolysaccharide (LPS) by wheat phytase and to investigate the effects of wheat phytase-treated LPS on in vitro toxicity, cell viability and release of a pro-inflammatory cytokine, interleukin (IL)-8 by target cells compared with the intact LPS. The phosphatase activity of wheat phytase towards LPS was investigated in the presence or absence of inhibitors such as L-phenylalanine and L-homoarginine. In vitro toxicity of LPS hydrolyzed with wheat phytase in comparison to intact LPS was assessed. Cell viability in human aortic endothelial (HAE) cells exposed to LPS treated with wheat phytase in comparison to intact LPS was measured. The release of IL-8 in human intestinal epithelial cell line, HT-29 cells applied to LPS treated with wheat phytase in comparison to intact LPS was assayed. Wheat phytase hydrolyzed LPS, resulting in a significant release of inorganic phosphate for 1 h (p < 0.05). Furthermore, the degradation of LPS by wheat phytase was nearly unaffected by the addition of L-phenylalanine, the inhibitor of tissue-specific alkaline phosphatase or L-homoarginine, the inhibitor of tissue-non-specific alkaline phosphatase. Wheat phytase effectively reduced the in vitro toxicity of LPS, resulting in a retention of 63% and 54% of its initial toxicity after 1–3 h of the enzyme reaction, respectively (p < 0.05). Intact LPS decreased the cell viability of HAE cells. However, LPS dephosphorylated by wheat phytase counteracted the inhibitory effect on cell viability. LPS treated with wheat phytase decreased IL-8 secretion from intestinal epithelial cell line, HT-29 cell to 14% (p < 0.05) when compared with intact LPS. In conclusion, wheat phytase is a potential therapeutic candidate and prophylactic agent for control of infections induced by pathogenic Gram-negative bacteria and associated LPS-mediated inflammatory diseases in animal husbandry.
Article
Full-text available
Biologically active interferon-α, tumor necrosis factor-α (TNF-α), and interleukin-1 (IL-1) were detected in bronchoalveolar lavage (BAL) fluids of 3-week-old cesarian-derived colostrum-deprived pigs inoculated with H1N1 influenza virus. Cytokine titers and lung virus titers were significantly higher 18–24 h after inoculation than at 48–72 h after inoculation in all 4 litters of pigs examined. All three cytokines were positively correlated with a 3- to 4-fold increase in BAL cell numbers (P < .036) and with a drastic neutrophil infiltration (24%–77% of BAL cells vs. 0–1.5% in controls) (P < .001). In addition, cytokine production coincided with the onset of general and respiratory symptoms of influenza and with the development of a necrotizing bronchopneumonia. This study is the first demonstration of TNF-α and IL-1 in BAL fluids of a natural influenza virus host. It documents that pigs may be a highly valuable experimental model in human influenza virus pneumonia.
Article
Full-text available
Diagnosis of ventilator associated pneumonia can be made by quantitative cultures of bronchoalveolar lavage fluid or of protected specimen brushings, though cultures require 24-48 hours to provide results. In 80% of cases aerobic Gram negative bacteria are the cause. A rapid diagnostic method of assessing the endotoxin content of lavage fluid by Limulus assay is described. Forty samples of lavage fluid were obtained from patients with multiple trauma requiring mechanical ventilation for a prolonged period. Pneumonia was diagnosed on the basis of clinical, radiological, and bacteriological findings, including quantitative cultures of lavage fluid. A relation was observed between the concentration of endotoxin in lavage fluid and the quantity of Gram negative bacteria. The median endotoxin content of lavage fluid in Gram negative bacterial pneumonia was 15 endotoxin units (EU)/ml; the range observed in individual patients was 6 to > 150 EU/ml. In patients with pneumonia due to Gram positive cocci and in non-infected patients the median endotoxin level was 0.17 (range < or = 0.06 to 2) EU/ml. An endotoxin level greater than or equal to 6 EU/ml distinguished patients with Gram negative bacterial pneumonia from colonised patients and from those with pneumonia due to Gram positive cocci. The measurement of endotoxin in lavage fluid is a rapid (less than two hours) and accurate diagnostic method. It should allow specific and early treatment of Gram negative bacterial pneumonia.
Article
Full-text available
Endotoxin (LPS), one of the major proinflammatory constituents of the cell walls of gram-negative bacteria, induces alveolar macrophages to express interleukin-1 (IL-1) and tumor necrosis factor (TNF) messenger RNA (mRNA), peaking at 1 hour in vitro. Intratracheal injection of LPS induces IL-1 and TNF mRNA expression in vivo in whole-lung RNA preparations. Interleukin-1 mRNA is not constitutively detected. In the case of TNF, however, a constitutively-expressed hybridization band is noted at 1.6 kb, whereas the LPS-induced hybridization band is noted at approximately 1.95 kb. Intratracheal injection of LPS induces an intra-alveolar inflammatory reaction composed of a neutrophilic exudate, peaking at 6 to 12 hours, a monocytic exudate peaking at 24 hours, and a lymphocytic exudate peaking at 48 hours, as quantitated by bronchoalveolar lavage. Intratracheal injection of IL-1 recapitulates the kinetics and relative magnitudes of the acute neutrophilic and chronic monocytic and lymphocytic inflammatory sequence. Intratracheal injection of TNF also induces an acute intraalveolar neutrophilic exudate, but TNF is much less potent of an inflammatory stimulus than IL-1. The effects of recombinant IL-1 and TNF are not due to LPS contamination, as shown by abrogation of the cytokines' inflammatory activity by boiling. In conclusion, LPS induces IL-1 and TNF mRNA expression in vitro in alveolar macrophages and in vivo in pulmonary tissue, and intratracheal injection of IL-1 and TNF recapitulates the LPS-induced pulmonary inflammatory sequence, strongly supporting the hypothesis that these cytokines play an important in vivo role in the pathogenesis of gram-negative bacterial pneumonia.
Article
The aim of this study was to investigate whether interleukin (IL)-6 induces the production of IL-1 and tumor necrosis factor (TNF) antagonists. Serial plasma samples were obtained from cancer patients participating in phase I and II trials of recombinant IL-6 administered as a 120-hour continuous intravenous (IV) infusion. Plasma IL-1 receptor antagonist (IL-1Ra) and soluble TNF receptor p55 (TNFsRp55) levels were measured by specific radioimmunoassays (RIAs). IL-1Ra levels increased rapidly, reaching peak values (9.6 +/- 1.7 ng/mL) within 2 to 4 hours of beginning treatment. Thereafter, levels promptly declined, reaching near baseline within 24 hours despite continuation of IL-6. TNFsRp55 plasma levels increased within 4 to 8 hours after initiating treatment and increased progressively throughout the duration of therapy. IL-1 beta and TNF-alpha plasma levels were below the detection limit in all samples tested. Peripheral blood mononuclear cells (PBMC) exposed to IL-6 produced only small amounts (1.56 +/- 0.3 ng/mL) of IL-1Ra, even in the presence of exogenous soluble IL-6 receptor (gp80). TNFsRp55 levels measured in the supernatants of IL-6- stimulated PBMC were below the detection limit of the assay. Macrophages generated by culturing monocytes in granulocyte-macrophage colony-stimulating factor (GM-CSF) were much more responsive to IL-6 than freshly isolated unfractionated or adherent PBMC and synthesized almost as much IL-1Ra when stimulated with IL-6 as with endotoxin. These results suggest that the antinflammatory properties of IL-6 may be due; in part, to the induction of IL-1Ra synthesis and the release of soluble TNF receptors. Our findings also suggest that tissue macrophages may be an important source of IL-6-induced IL-1Ra.
Article
In early 1991, the Dutch pig-industry was struck by the so-called mystery swine disease. Large-scale laboratory investigations were undertaken to search for the etiological agent. We focused on isolating viruses and mycoplasmas, and we tested paired sera of affected sows for antibodies against ten known pig viruses. The mycoplasmas M. hyosynoviae, M. hyopneumoniae, and Acholeplasma laidlawii, and the viruses encephalomyocarditis virus and porcine enterovirus types 2 and 7 were isolated from individual pigs. An unknown agent, however, was isolated from 16 of 20 piglets and from 41 of 63 sows. This agent was characterised as a virus and designated Lelystad virus. No relationship between this virus and other viruses has yet been established. Of 165 sows reportedly afflicted by the disease, 123 (75 per cent) seroconverted to Lelystad virus, whereas less than 10 per cent seroconverted to any of the other virus isolates or to the known viral pathogens. Antibodies directed against Lelystad virus were also found in pigs with mystery swine disease in England, Germany, and in the United States. We conclude that infection with Lelystad virus is the likely cause of mystery swine disease.
Article
An experimental model that demonstrates a mycoplasma species acting to potentiate a, viral pneumonia was developed. Mycoplasma hyopneumoniae, which produces a chronic, lymphohistiocytic bronchopneumonia in pigs, was found to potentiate the severity and the duration of a virus-induced pneumonia in pigs, Pigs were inoculated with M hyopneumoniae 21 days prior to, simultaneously with, or 10 days after inoculation with porcine reproductive and respiratory syndrome virus (PRRSV), which induces an acute interstitial pneumonia in pigs, PRRSV-induced clinical respiratory disease and macroscopic and microscopic pneumonic lesions were more severe and persistent in M. hyopneumoniae-infected pigs. At 28 or 38 days after PRRSV inoculation, nl, hyopneumoniae-infected pigs still exhibited lesions typical of PRRSV-induced pneumonia, whereas the lungs of pigs which had received only PRRSV were essentially normal. On the basis of macroscopic lung lesions, it appears that PRRSV infection did not influence the severity of M hyopneumoniae infection, although microscopic lesions typical of M hyopneumoniae were more se,ere in PRRSV-infected pigs, These results indicate that M: hyopneumoniae infection potentiates PRRSV induced disease and lesions. Most importantly, M hyopneumoniae-infected pigs with minimal to nondetectable mycoplasmal pneumonia lesions manifested significantly increased PRRSV-induced pneumonia lesions compared to pigs infected with PRRSV only, This discovery is important with respect to the control of respiratory disease in pigs and has implications in elucidating the potential contribution of mycoplasmas in the pathogenesis of viral infections of other species, including humans.
Article
Underlying viral infections can heighten sensitivity and worsen cytokine-mediated disease following secondary inflammatory challenges, Mechanisms for this are poorly understood. The impact of the innate response to lymphocytic choriomeningitis virus (LCMV) infection on sensitivity to endotoxin (LPS) was investigated, Compared with uninfected mice, infection with LCMV for 2-days-sensitized mice to LPS by similar to2-fold for lethality and by 2- to 6-fold for serum TNF-alpha levels. Priming for LPS-induced TNF-alpha was also seen with splenic and peritoneal leukocytes isolated from infected mice and challenged with LPS ex vivo, The effect on TNF-alpha production was present in the absence of IFN-gamma, its major producers NK and T cells, and the major pathways for its induction through IL-12 and the signal transducer and activator of transcription 4 (STAT4), and therefore was IFN-gamma independent. ;Early LCMV infection induces high concentrations of the type 1 IFNs, IFN-alpha beta. Administration of recombinant IFN-alpha alone heightened the TNF-alpha response to LPS, Innate IFN-alpha beta and IFN-gamma responses to LCMV exist in a delicate balance. To reduce priming for LPS-induced TNF-alpha during LCMV, deficiencies in both the IFN-alpha beta and IFN-gamma receptors or STATI, a transcription factor downstream to bath IFNs, were required. These data demonstrate that early viral infection can enhance sensitivity to bacterial products, and that this sensitization can occur in part as a result of endogenously expressed IFN-alpha beta. This work also raises issues about potential complications associated with IFN-alpha beta therapies.
Article
We studied the interferon-alpha (IFN-alpha) system in relation to the porcine arterivirus (PoAV), porcine reproductive and respiratory syndrome virus (PRRSV). Recombinant porcine IFN-alpha inhibited the growth of this virus in alveolar macrophage cultures, When pigs were challenged intranasally with PoAV, their serum contained IFN-alpha in relatively low concentrations on the second day after challenge and up to 5 days at the latest. Most animals had no IFN-alpha in their lung secretions, even though PoAV replicates in the respiratory tract. In vitro, PoAV replicates in alveolar macrophages, but neither these nor peripheral blood mononuclear cells (PBMC) produced IFN-alpha in response to infection. This may be because PoAV suppresses IFN-alpha production. When macrophages treated with PoAV were superinfected with swine transmissible gastroenteritis virus (TGEV), a known good inducer of IFN, no IFN-alpha was detected. This suppressive effect was lost when the virus was inactivated by UV light. Our results suggest that downregulation of IFN-alpha production may play an important part in enabling PoAV to replicate in cell cultures and in pigs.
Article
Tumour necrosis factor-α (TNFα) is implicated in the pathogenesis of asthma; however, little is known of its direct effect on smooth muscle reactivity. We investigated the effect of TNFα on the responsiveness of human bronchial tissue to electrical field stimulation in vitro. Incubation of non-sensitized tissue with 1 nM, 3 nM and 10 nM TNFα significantly increased responsiveness to electrical field stimulation (113 ± 8, 110 ± 4 and 112 ± 2% respectively) compared to control (99 ± 2%) (P < 0.05, n = 6). Responses were not increased in sensitized tissue (101 ± 3% versus 105 ± 5%, n = 3, P > 0.05) nor were responses to exogenous acetylcholine (93 ± 4% versus 73 ± 7%, n = 3, P = 0.38). These results show that TNFα causes an increase in responsiveness of human bronchial tissue and that this occurs prejunctionally on the parasympathetic nerve pathway. This is the first report of a cytokine increasing human airway tissue responsiveness.