SlideShare a Scribd company logo
1 of 9
Download to read offline
Veterinary Immunology and Immunopathology 158 (2014) 53–61
Contents lists available at ScienceDirect
Veterinary Immunology and Immunopathology
journal homepage: www.elsevier.com/locate/vetimm
Research paper
Early inflammatory response to the saponin adjuvant
Matrix-M in the pig
Caroline Fossuma,∗
, Bernt Hjertnera
, Viktor Ahlberga
,
Wasin Charerntantanakula,b
, Kathy McIntoshc
, Lisbeth Fuxlera
,
Navisraj Balagunaseelana
, Per Wallgrend
, Karin Lövgren Bengtssone
a
Department of Biomedicine and Veterinary Public Health, Section for Immunology, Swedish University of Agricultural Sciences, P.O.
Box 588, SE-751 23 Uppsala, Sweden
b
Research Laboratory for Immunity Enhancement in Humans and Domestic Animals Maejo University, Chiang Mai 50290, Thailand
c
Department of Veterinary Microbiology, University of Saskatchewan, Western College of Veterinary Medicine, Saskatoon, Canada
d
National Veterinary Institute, SVA, SE-751 89 Uppsala, Sweden
e
Isconova AB, Kungsgatan 109, SE-753 18 Uppsala, Sweden
a r t i c l e i n f o
Article history:
Received 3 December 2012
Received in revised form 20 July 2013
Accepted 23 July 2013
Keywords:
Matrix-M
ISCOM
Pig
Transcriptional
NETs
IRGs
Adjuvant
a b s t r a c t
The early inflammatory response to Matrix-M was evaluated in pigs. Adverse reactions
measured as body temperature, appetite, activity level and reaction at the site of injection
were not observed after s.c. injection with three doses of the adjuvant (75, 100 or 150 ␮g)
into one week old piglets. Analyses of the immediate cytokine response of PBMC after
in vitro exposure to Matrix-M (AbISCO-100®
) revealed only a low expression of mRNA for
tumour necrosis factor-␣ (p < 0.05) after 6 h incubation. Histological examination revealed
an infiltration of leukocytes, haemorrhage and necrosis in muscle 24 h after i.m. injection of
150 ␮g Matrix-M in pigs aged eleven weeks. At this time, different grades of reactive lym-
phoid hyperplasia were recorded in the draining lymph node that was enlarged in three
of these six pigs injected with Matrix-M. The global transcriptional response at the site of
injection and in the draining lymph node was analyzed using Affymetrix GeneChip Porcine
Genome Array. A significant enrichment of gene signatures for the cell types described as
“myeloid cells” and “plasmacytoid dendritic cells” was observed at the site of injection in
Matrix-M injected pigs compared with pigs injected with saline. A number of genes encod-
ing cytokines/chemokines or their receptors were upregulated at the injection site as well
as in the draining lymph node. In the draining lymph node, a majority of the upregulated
genes were interferon-regulated genes (IRGs). The expression of IFN-␤, but not IFN-␣, was
increased in the draining lymph nodes of a majority of the pigs exposed to Matrix-M. These
IFN-␤ expressing pigs also expressed increased levels of osteopontin (OPN) or stimulator
of interferon genes (STING), two factors known to facilitate the expression of type I IFNs in
response to viral infection. Thus, Matrix-M does not appear to induce any harmful inflam-
matory response in piglets whilst contributing to the innate immunity by activating the
type I IFN system, possibly through several alternative signalling pathways.
© 2013 Elsevier B.V. All rights reserved.
∗ Corresponding author. Tel.: +46 018 4714056; fax: +46 018 4714382.
E-mail address: Caroline.Fossum@slu.se (C. Fossum).
1. Introduction
The immune-stimulating complex (ISCOM) is an
antigen-presenting system in which antigens are incor-
porated into a matrix of the saponin adjuvant Quil
A, formulated into particles together with cholesterol
0165-2427/$ – see front matter © 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.vetimm.2013.07.007
54 C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61
and phospholipid (Morein et al., 1984). The formulation
soon proved useful for a number of membrane-derived
antigens for induction of the protective immunity to
various microorganisms (Morein, 1987, 1988). Electron
microscopy demonstrated the formation of a spherical
cage-like structure, 40 nm in size, regardless of the source
of antigen. A standardized methodology and mixture of
Quil A, cholesterol and phophatidylcholine was established
for incorporation of amphipathic antigens (reviewed in
Lövgren Bengtsson and Morein, 2000). Later, it was realized
that incorporation of antigens into the ISCOM structure
was not necessary for potent immune stimulation (Lövgren
and Morein, 1988; Lövgren Bengtsson and Sjölander, 1996).
Pre-formed ISCOMs without incorporated antigen, called
ISCOM-Matrix and later referred to as Matrix, became used
as stand-alone adjuvant, simply mixed with antigens.
The glycosidic saponins, extracted from the bark of the
tree Quillaja saponaria Molina (Quil A), in free form, have
been used as adjuvant for almost a century (reviewed in
Dalsgaard et al., 1990). Quil A is a potent adjuvant; how-
ever, free saponins have haemolytic activity that may cause
side effects. By formulation of saponins with cholesterol
and phospholipids in ISCOMs or in Matrix particles, the
haemolytic activity is abolished and a more potent and less
reactogenic adjuvant is created. The Matrix made from Quil
A or similar preparations are currently called Matrix-Q.
Biochemical separation techniques revealed that the
lytic and structure forming capacities of Quil A were
mainly restricted to different fractions (Kensil et al., 1991;
Rönnberg et al., 1995) and that various combinations
of these components affected the adjuvant properties
considerably (Johansson and Lövgren-Bengtsson, 1999).
Detailed biochemical and functional characterisations of
the saponin fractions have resulted in a very well-
tolerated Matrix formulation called Matrix-M. Matrix-M
is a mixture of Matrix particles made from two dif-
ferent purified fractions of Quillaja derived saponins.
Matrix-MTM (Isconova AB, Uppsala, Sweden) is a potent
adjuvant that is licensed for several animal vaccines
and now also has entered human clinical trials (Lövgren
Bengtsson et al., 2011). Another similar, albeit differ-
ent product based on the ISCOM technology is the
ISCOMATRIX® adjuvant (CSL; Commonwealth Serum Lab-
oratories, Melbourne, Australia). MatrixTM formulations
are available in various forms and recommended for dif-
ferent species according to their saponin sensitivity and
there are two preparations available for research pur-
poses, AbISCO-100® (Matrix-M type) and AbISCO-300®
(Matrix-Q type).
Attempts to explore the power of Matrix formula-
tions on immune parameters have mainly been carried
out in mice. These studies have revealed a prominent
recruitment and activation of cells in the draining
lymph node/spleen, antigen delivery to dendritic cells
accompanied by cytokine and chemokine production.
This allows for cross-presentation with the subsequent
induction of cytotoxic T cells and a long-lasting antibody
response (Duewell et al., 2011; Morelli et al., 2012;
Reimer et al., 2012). However, which molecular path-
way(s) are activated by the Matrix of ISCOM is still not
clarified.
In the pig, early experimental ISCOM/Matrix-Q vac-
cines focused on Aujeszky’s Disease Virus (Tsuda et al.,
1991; Puentes et al., 1993; Tulman and Garmendia, 1994),
rotavirus (Iosef et al., 2002; Nguyen et al., 2003; González
et al., 2004; Nguyen et al., 2006a; Azevedo et al., 2010), and
the use of virus-like particles in combination with ISCOM-
Matrix (Tulman and Garmendia, 1994). The latter concept
has been used successfully in young pigs, inducing an active
immune response even in the presence of maternal immu-
nity (Nguyen et al., 2006b; McIntosh et al., manuscript) as
also shown for other ISCOM formulations applied in calves
(Hägglund et al., 2004) or mice (Morein et al., 2007).
Taken together, Matrix-M appears as a promising adju-
vant also in the pig where improved vaccines to several
diseases are desirable. The pig is also a valuable model for
studies of adjuvant effects in man because of many simi-
larities between the species in the organization of innate
immune cells and their cytokine production (Auray et al.,
2010; Bertho et al., 2011; Faibairn et al., 2011; Marquet
et al., 2011; Kapetanovic et al., 2012). To reflect the early
inflammatory response to Matrix-M in the pig, results
from in vitro studies using porcine blood mononuclear or
polymorphonuclear cells are reported here together with
results from an in vivo toxicity test carried out in piglets. The
findings are related to the global transcriptional response
to Matrix-M recorded in pigs at the site of injection and in
the draining lymph node (Ahlberg et al., 2012).
2. Materials and methods
2.1. Animals and experimental designs
Five experimental set ups were used to study the early
inflammatory response to the Matrix of ISCOMs in the pig.
An animal toxicity study was carried out in one-week old
PIC-crossbred piglets, housed in a 600 sow commercial
swine farrow-to-finish facility in Saskatchewan, Canada
(Set I). In vitro cellular toxicity studies were performed with
PBMC (Set II) and polymorphonuclear neutrophil leuko-
cytes (PMNL; Set III) purified from the blood of finishing
pigs housed in a specific pathogen free herd (Wallgren et al.,
1999; Swedish Livestock Research Centre, Lövsta-Uppsala,
Sweden). In vitro induced expression of cytokine mRNA (Set
IV) was determined using PBMC obtained from convention-
ally reared pigs at the University Research Station (Funbo
Lövsta, Uppsala, Sweden) whereas in vivo induced expres-
sion of mRNA for interferon-related genes (Set V) was
studied in lymph nodes obtained from 11 weeks old SPF-
pigs (Ahlberg et al., 2012). All procedures were conducted
in accordance with the University of Saskatchewan’s Com-
mittee for Animal Care and Supply (permit #20060004)
and with the Ethical Committee for Animal Experiments,
Uppsala, Sweden.
2.2. In vivo evaluation of Matrix-Q toxicity (Set I)
The potential for adverse reactions was tested by the
s. c. injection of variable doses of Matrix-Q (Isconova AB,
Uppsala, Sweden) into one-week-old piglets. Piglets were
injected with either 75 ␮g (n = 3), 100 ␮g (n = 3), or 150 ␮g
(n = 3) of the Matrix-Q diluted in sterilized 0.01 M PBS to
C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61 55
a final volume of 200 ␮L using a 1 mL-25 gauge needle. A
single injection was administered subcutaneously 1 cm off
the dorsal midline between the shoulders and piglets were
observed 7 times over a period of 30 h for rectal temper-
ature, injection site reactivity (swelling, redness, or pain),
and activity level (active or lethargic).
2.3. In vitro evaluation of Matrix-M toxicity (Set II)
The viability of PBMC was evaluated after 18 h incu-
bation in the presence of 0.3, 1 or 3 ␮g Matrix-M
(AbISCO-100®; Isconova AB) per ml culture medium.
PBMCs were isolated from heparinized blood by density
gradient centrifugation on Ficoll-Paque (Pharmacia, Upp-
sala, Sweden). The cell viability was determined by flow
cytometry (FACSCanto flow cytometer; BD Biosciences,
San Jose, CA) using the Annexin V-FITC Apoptosis Detec-
tion Kit I with Propidium Iodide (PI) staining solution (BD
PharmingenTM, San Jose, CA) as recommended. Parallel cul-
tures with PBMC incubated in growth medium or treated
with UV-light (480 mJ for 90 s) before incubation were
included as negative and positive controls, respectively.
Ten thousand events per sample were collected and the
data were analyzed using the FACSDiva software (v. 5.0.2,
BD Biosciences).
2.4. In vitro effects of Matrix-M on PMNL activity (Set III)
Heparinized blood was diluted with an equal volume of
3% Dextran (T-2000, Pharmacia Biotech, Uppsala, Sweden)
and allowed to sediment for 30 min at RT. The leucocyte
rich fraction was washed once in PBS, resuspended and
layered on a discontinuous gradient of 70% and 80% Per-
coll (GE Healthcare, Uppsala, Sweden). After centrifugation
(300 × g) for 25 min, cells in the two bands generated were
collected, the cell numbers were counted and the purity
was determined on cytospin glasses stained with Diff Quick
(Vector Lab Inc., Burligame, CA). The cells recovered on the
80% Percoll cushion were enriched for PMNL (91.8 ± 6.1%,
n = 20) and used for studies of the formation of neutrophil
extracellular traps (NETs).
The PMNL were washed twice in PBS and resuspended in
RPMI 1640 supplemented with HEPES, l-glutamine antibi-
otics and 2% BSA (Sigma–Aldrich®). Two millilitre cell
suspension (107 neutrophils per ml) was seeded on cov-
erslips in six well plates (Costar®) and incubated for 1 h
at 37 ◦C. Thereafter, 400 ␮l medium (negative control) or
phorbol 12-myristate 13-acetate (PMA; Sigma–Aldrich®)
at a final concentration of 100 nM (positive control) were
added to the wells. The effect of Matrix-M was tested at
the final concentrations of 0.3, 1 and 3 ␮g/ml in parallel
cultures. After 4 h of incubation the medium was care-
fully removed from the wells and 1 ml of 50 nM Sytox
Green (Invitrogen) was added to each well and incu-
bated in the dark for 10 min. Thereafter formaldehyde
(ProlaboTM, Taipan, Malaysia) was added to a final con-
centration of 4% per well and incubated for 30 min before
the coverslip was carefully removed and allowed to dry
before mounted in Vectashield with DAPI (Vector Lab Inc.,
Burligame, CA). The morphology of cell nucleus and extra-
cellular DNA fragments were determined in a fluorescence
microscope (Nikon, Microphot-FX, Nikon Instruments Inc.,
Tokyo, Japan) equipped with a digital camera (Coolpix 990,
Nikon) for documentation.
2.5. In vitro Matrix-M induced expression of cytokine
genes (Set IV)
The effect of Matrix-M on the expression of cytokine
mRNA by porcine PBMC was tested in vitro and com-
pared to that induced by the CpG-ODN 2216 (Cybergene
AB, Huddinge, Sweden) or LPS (Sigma Aldrich, Stein-
heim, Germany), using methods detailed elsewhere (Bolind
Bågenholm, 2009; Wikström et al., 2011). All inducers were
diluted in RPMI 1640 medium (BioWhittaker, Cambrex
Bioscience, Verviers, Belgium) supplemented with 20 mM
HEPES buffer, 2 mM l-glutamine, 200 IU penicillin/ml,
100 ␮g/ml streptomycin, 0.5 ␮M 2-mercaptoethanol and
5% foetal calf serum (Invitrogen, Life Technologies, Carls-
bad, CA, USA). Three ml cell cultures (final concentration
5 × 106 cells per ml) were established in 25 cm2 tissue cul-
ture flasks (Nunc, Roskilde, Denmark) containing Matrix-M
(1 ␮g/ml), LPS (10 ␮g/ml), ODN 2216 (5 ␮g/ml) or only
growth medium. After 6 hours incubation at 37 ◦C, total
RNA was extracted, quality tested and DNA:se treated
before 2 ␮g of RNA was used for cDNA synthesize as
described (Wikström et al., 2011).
Real-time TaqMan PCR was performed for IFN-␣, IFN-
␥, IL-6, IL-10, IL-12p40, IL-1␤, TNF-␣, TGF-␤, CXCR4,
RGS16 and two reference genes, Cyclophilin A (CyA)
and Hypoxanthine-guanine phosphoribosyl transferase
(HPRT), using previously published primers and protocols
(Wikström et al., 2011). The relative expression of target
genes induced by Matrix-M, LPS or ODN 2216 was com-
pared to their expression in PBMC cultured in plain growth
medium according to Livak and Schmittengen (2001), using
Cy A and HPRT as reference genes.
2.6. In vivo transcriptional response to Matrix-M (Set V)
Pigs were injected i.m. with either 150 ␮g Matrix-M
suspended in 1 ml sterile endotoxin-free 0.9% NaCl solu-
tion (n = 6) or just saline (n = 6). Twenty four h after
injection, pigs were necropsied and tissues from the site
of injection and their draining lymph nodes were col-
lected and analyzed for early transcriptional response using
Affymetrix GeneChip® Porcine Genome Array as detailed
in Ahlberg et al. (2012). This was followed up by a more
comprehensive expression analysis focusing on IFN-␣, IFN-
␤, osteopontin (OPN) and stimulator of interferon genes
(STING) in the draining lymph node. Sample preparation,
RNA extraction and analysis, and cDNA construction are
detailed in Ahlberg et al. (2012).
Primer pairs for IFN-␣, IFN-␤, OPN, riboso-
mal protein L32 (RPL32), STING and tyrosine
3-monooxygenase/tryptophan 5-monooxygenase activa-
tion protein (YWHAZ) were chosen from published works
favouring those spanning an intron, and reoptimized in
house (Table 1). SYBR Green PCR was run as described
previously (Hjertner et al., 2013). Using PCR base line
normalized Cq values the expression of IFN-␣, IFN-␤,
OPN and STING was normalized to the geometric average
56 C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61
Table 1
Primer details and gene specific optimized conditions.
Gene Primer sequence Primer location Target sequence Anneal
temp (◦
C)
Primer
conc (nM)
E (%)g
r2
Melt point
(◦
C)
IFN-␣a
F:AGCCTCCTGCACCAGTTCTG 346–365 NM 214393.1 60 500 100 0.997 84.5
R: TCACAGCCAGGATGGAGTCC 469–450
IFN-␤b F: TAGCACTGGCTGGAATGAAACC 288–309 NM 001003923.1 58 400 104 0.993 79.5
R: TCAGGTGAAGAATGGTCATGTCT 427–405
OPNc
F: TTGGACAGCCAAGAGAAGGACAGT 731–754 NM 214023.1 56 300 93 0.997 82.5
R: GCTCATTGCTCCCATCATAGGTCTTG 851–826
RPL32d F: CGGAAGTTTCTGGTACACAATGTAA 249–273 NM 001001636.1 55 300 97 0.997 77
R: TGGAAGAGACGTTGTGAGCAA 342–322
STINGe
F: TTACATCGGGTACCTGCGGC 489–508 NM 001142838.1 56 500 101 0.992 82
R: CCGAGTACGTTCTTGTGGCG 572–553
YWHAZf F: ATTGGGTCTGGCCCTTAACT 961–980 XM 001927228.4 58 400 101 0.997 78.5
R: GCGTGCTGTCTTTGTATGACTC 1106–1085
a
Wikström et al. (2011).
b
Lin et al. (2013).
c
White et al. (2005).
d
Dawson et al. (2004).
e
Xie et al. (2010).
f
Uddin et al. (2011).
g
Efficiency from serial dilutions of reference cDNA.
of RPL32 and YWHAZ (Vandesompele et al., 2002), and
calibrated to the average expression in all six control pigs.
2.7. Statistical analyses
Statistical differences in mRNA expression for IFN-␣,
IFN-␤, STING and OPN between pigs injected with Matrix-
M or saline were analyzed using the non-parametric
Mann–Whitney test whereas statistical differences for var-
ious in vitro treatments were determined using the paired
Student’s t-test (GraphPad Prism version 5.0 for Mac OS X,
GraphPad Software, San Diego, CA). p-Values ≤ 0.05 were
regarded as significant. For all statistical analyses detailed
in Ahlberg et al. (2012), q-values, i.e., p-values corrected for
multiple testing were used.
3. Results and discussion
3.1. In vivo evaluation of Matrix-Q toxicity (Set I)
The safety of saponin-based Matrix adjuvants in pigs
was studied with Matrix-Q in one-week-old piglets. No
adverse reactions were observed at the injection site
nor was the activity level of any piglet during the 30 h
period after injection with the Matrix-Q affected. The
expected normal rectal temperature of a suckling piglet
>24 h of age is 39.2 ± 0.3 ◦C (Straw et al., 1999). Taking
into consideration the baseline rectal temperature of each
piglet (temperature prior to injection), one piglet at 30 h
post-injection recorded a low grade fever of 39.6 ◦C (a tem-
perature greater than 0.3 ◦C above its baseline temperature
and greater than 39.5 ◦C). However, this piglet received the
lowest dose of Matrix-Q of 75 ␮g.
3.2. In vitro evaluation of Matrix-M toxicity (Set II)
The effect of Matrix-M on cell survival was tested
in vitro using PBMC obtained from five pigs (Fig. 1). After
18 h of culture the proportion of Annexin labelled cells
M
edium
0.3
M
atrix
1
M
atrix
3
M
atrix
U
V
0
5
10
15
20
Percentage
a
M
edium
0.3
M
atrix
1
M
atrix
3
M
atrix
U
V
0
20
40
60
80
100
Percentage
b
Fig. 1. Percentage of apoptotic (a) or dead (b) porcine PBMC after 18 h of
incubation in the presence of 0.3, 1 or 3 ␮g per ml of Matrix-M. The cor-
responding figures for PBMC incubated in medium or UV-treated before
incubation are included as negative and positive controls, respectively.
C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61 57
that were negative for PI was in mean 12.6 ± 4.6% for
cells incubated in the presence of Matrix-M, regardless
of concentration. This was similar to cells incubated in
growth medium (11.7 ± 2.7%) but higher than for those
that were UV-treated (6.7 ± 3.5%) prior to incubation. A
great proportion (74.5 ± 16.4%) of the UV-treated PBMC
was however stained with both PI and Annexin indicat-
ing a low cell survival. The corresponding figure for PBMC
grown in various concentrations of Matrix M (12.2 ± 3.7%)
was again very similar to that for PBMC grown in culture
medium (12.6 ± 2.3%). Thus, Matrix M did not induce apo-
ptosis or increase cell death during the present culture
conditions.
3.3. In vitro effects of Matrix-M on PMNL activity (Set III)
The ability of Matrix-M to induce formation of NETs was
tested in vitro using porcine PMNL obtained from pigs in a
SPF herd. The number of PMNL recovered per ml blood was
low due to the high health status, and prior activation of
them in vivo was less likely than for PMNL obtained from
conventionally reared pigs. In accordance, no signs of acti-
vation were evident after 4 h incubation in plain growth
medium (Fig. 3a) whereas a high proportion of neutrophils
with a condensed nucleus, showing signs of NETosis and
release of NETs was observed among the neutrophils incu-
bated with PMA (Fig. 3b and c).
The effect of Matrix-M on the neutrophils was less
evident. In the cultures supplemented with 0.3 or 1.0 ␮g
Matrix-M per ml (Fig. 3d and e) the nucleus of the neu-
trophils showed the same shape as those incubated in plain
growth medium. Among neutrophils cultured in the pres-
ence of 3 ␮g Matrix-M per ml (Fig. 3f) a slightly higher
proportion of the neutrophils had a condensed nuclei but
no NET formation was detected. Prolonged incubation, up
to 16 h, affected the survival of the neutrophils regardless
of culture set up. Still, no NET formation was observed in
cultures supplemented with Matrix-M or in the cultures
with plain growth medium. Instead, the nuclei of PMNL
grown over night in the presence of Matrix-M were dis-
integrated into small Sytox-stained fragments dispersed in
considerably swelled cells or around disrupted cells, giving
the impression of a pyroptotic cell death. The activation
of cathepsin genes (CTSB, CTSD, CTSH, CTSS and CTSZ) at
the injection site and the up regulation of the genes for
IL-1␤ and IL-18 (Table 5, Ahlberg et al., 2012) agree with
pyroptosis (Labbé and Saleh, 2011).
3.4. Early cytokine response of PBMC exposed to
Matrix-M in vitro (Set IV)
Analyses of the immediate cytokine response of PBMC
after in vitro exposure to Matrix-M revealed a low level
induction of TNF-␣ mRNA (p < 0.05) after 6 h incubation.
The mRNA expression for the other cytokines analyzed
did not differ from that of PBMC cultured in plain growth
medium. In comparison, mRNA specific for IL-1␤, IL-6 and
IL-10 (p < 0.001), and IL-12p40 and TNF-␣ (p < 0.01) was
induced after 6 h exposure of the PBMC to LPS, and IFN-
␣ mRNA (p < 0.01) was induced after 6 h in the presence
of the ODN 2216. Thus, Matrix-M did not evoke any strong
Table 2
Significant differences in relative mRNA expression determined by qPCR.
RNA was isolated after 6 h culture of porcine PBMC in the presence
of Matrix-M (1 ␮g/ml), LPS (10 ␮g/ml), ODN 2216 (5 ␮g/ml) or in plain
growth medium.
Target gene Matrix-M vs.
medium (p
value)
LPS vs. medium
(p value)
ODN 2216 vs.
medium (p
value)
IFN-␣ ns * **
IFN-␥ ns ns *
IL-6 ns *** *
IL-10 ns *** *
IL-1␤ ns ***
ns
TNF-␣ * ** *
TGF-␤ ns *
ns
IL-12p40 ns ** *
CXCR4 ns ns **
RGS16 ns ns ns
ns: not significant.
*
p ≤ 0.05.
**
p ≤ 0.01.
***
p ≤ 0.001.
immediate pro-inflammatory cytokine response in porcine
PBMC when tested in vitro. These results are in agreement
with the absence of fever and local reactions in the tox-
icity tests but caution should be taken regarding the fact
that these studies were performed in cultures of purified
lymphoid cells, devoid of PMNL and possibly also lacking
important soluble factors produced by other cells.
3.5. Global transcriptional response to Matrix-M at the
site of administration and in draining lymph node (Set V)
In order to better reflect the immune stimulatory prop-
erties of Matrix-M the global transcriptional response was
analyzed following i.m. administration of 150 ␮g Matrix-M
in pigs aged eleven weeks (Ahlberg et al., 2012). Histo-
logical examination revealed an infiltration of leukocytes,
haemorrhage and necrosis in muscle after 24 h. At this
time, different grades of reactive lymphoid hyperplasia
were recorded in the draining lymph node that showed a
Fig. 2. Interferon-regulated genes (IRGs) upregulated at the injection site
(n = 36) and/or draining lymph node (n = 38) 24 h after Matrix-M admin-
istration. The ten most up-regulated IRGs in each group are listed. IRGs
were defined by the database Interferome (http://www.interferome.org).
58 C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61
moderate enlargement in three of the six pigs injected with
Matrix-M. These observations (Table 2 and Fig. 1; Ahlberg
et al., 2012) were well in line with the absence of visi-
ble signs of inflammation when pigs were injected with
a similar dose of Matrix-Q and the marginal indication of
a pro-inflammatory cytokine response in porcine PBMC
exposed to Matrix-M in vitro.
As reported in Ahlberg et al. (2012), an inflammatory
response to Matrix-M at the injection site was evident
from the enrichment of genes in gene ontology (GO)
terms such as ‘immune response’ (q = 1.4E−10), ‘response
to wounding’ (q = 1.4E−9), ‘defence response’ (q = 5.1E−9),
‘inflammatory response’ (q = 7.4E−9), ‘positive regula-
tion of immune system process’ (q = 2.1E−4) and ‘innate
immune response’ (q = 7.2E−4). The corresponding analy-
sis of gene regulation in the draining lymph node (Ahlberg
et al., 2012) revealed an enrichment of up regulated genes
in the GO-terms ‘immune response’ (q = 9.9E−6), ‘response
to virus’ (q = 4.6E−4), ‘defence response’ (q = 2.3E−3), ‘reg-
ulation of cell proliferation’ (q = 1.1E−2) and ‘behaviour’
(q = 4.8E−2).
Cell migration patterns induced by Matrix-M were
characterized by enrichment for “myeloid cells” and in
particular by plasmacytoid dendritic cells (pDC). This
profile, together with the prominent up regulation of a
considerable number of interferon regulated genes (IRGs),
both at the site of injection and in the draining lymph node
(Fig. 2) initiated studies on possible mechanisms for induc-
tion of IFN production by Matrix-M. Because the genes
for two cytosolic RNA-sensing receptors (RIG-1 and MDA5)
both were upregulated as well as MyD88 that mediates
signalling via TLR7 and TLR9, it was tempting to specu-
late that nucleic acid played a part in the induction of
IRGs.
In patients with the autoimmune disorder systemic
lupus erythematosus (SLE), self-DNA released from PMNL
in the form of NETs can trigger pDC to produce IFN-␣
(Lande et al., 2011). In that case autoantibodies to DNA
are thought to mediate the uptake and delivery of DNA to
endosomal TLR9. One of the advantages with the ISCOMs
is their ability to deliver antigen to the cytosol allowing
presentation of antigen on MHC class I molecules and the
induction of a cytotoxic T cell response (Takahashi et al.,
1990). Therefore, Matrix-M could in theory mediate trans-
port of any associated nucleic acid over cell membrane(s)
and thereby initiate an IFN-response. The studies of PMNL
exposed to Matrix-M in vitro (Set III) did however not sup-
port the hypothesis that Matrix-M induced NET formation,
although a prominent influx of neutrophils were recorded
after in vivo administration.
Fig. 3. Porcine neutrophilic granulocytes incubated for 4 h in plain growth medium (a), 100 nM PMA, 20× magnification (b), 100 nM PMA, 40× magnification
(c) or 0.3 ␮g (d), 1 ␮g (e), 3 ␮g (f) Matrix-M per ml.
C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61 59
IFN-αα
1 2 3 4 5 6 7 8 9 10 11 12
0.0
0.5
1.0
1.5
2.0
Relativeexpression
IFN-ββ
1 2 3 4 5 6 7 8 9 10 11 12
0
10
20
30
40
OPN
1 2 3 4 5 6 7 8 9 10 11 12
0
5
10
15
Matrix Saline
Relativeexpression
STING
1 2 3 4 5 6 7 8 9 10 11 12
0
1
2
3
4
Matrix Saline
Fig. 4. Relative expression of IFN-␣, IFN-␤, OPN and STING in the iliac lymph node of pigs exposed to Matrix-M (nos. 1–6) or saline (nos. 7–12). The
expression was normalized to the geometric average of RPL32 and YWHAZ and calibrated to the average expression of all six control pigs. The difference
in expression between Matrix-M treated and saline treated groups was statistically significant (p < 0.05) for IFN-␤ and OPN.
3.6. In vivo effects of Matrix-M on IFN-related responses
(Set V)
The indicated IFN response in pigs early after injection
with Matrix-M was followed up with an analysis of the
expression of IFN-␣ and -␤, as well as OPN (SPP1) and
STING in the draining lymph nodes of pigs that received
Matrix-M (n = 6) or just saline (n = 6). OPN and STING have
recently been identified as two modulators acting on two
different pathways that signal the expression of IFN-␣/␤
in response to viral infections as reviewed by Levy et al.
(2011). OPN, which had more than a hundred-fold increase
in expression at the site of injection (Ahlberg et al., 2012)
is an essential factor for endosomal TLR7/9 dependent
induction of IFN-␣/␤ expression via the adaptor molecule
MyD88 (Shinohara et al., 2006). STING is essential for
cytoplasmic foreign DNA sensor signalling, as well as RIG-
like receptor (RLR) signalling, acting through recruitment
of TBK1 (Ishikawa et al., 2009). Furthermore, recently a
new innate detection mechanism involving STING/TBK1
but independent of TLR and RLR pathways was described,
which was triggered by virus-cell membrane fusion only
(Holm et al., 2012).
Transcripts from IFN-␣, IFN-␤, OPN and STING could
be detected in the lymph node of all pigs except for two
control pigs, which had no detectable IFN-␤ expression.
In this case a Cq value of 40 was assigned. The relative
expression of IFN-␣ was always less than two with no
difference in expression between Matrix M-treated and
control pigs (Fig. 4). However, four pigs (nos. 1, 2, 3 and 6)
out of the six Matrix-M-treated pigs showed elevated IFN-
␤ levels (p < 0.05) compared to the control group (Fig. 4).
In three of these pigs (nos. 1, 3 and 6) the relative expres-
sion of OPN was increased. The expression of OPN in the
Matrix-M treated group of pigs was significantly (p < 0.05)
higher than in the control group (Fig. 4). The fourth pig with
elevated IFN-␤ expression (no. 2) had an OPN expression
level comparable to the control pigs but in this pig the rela-
tive expression of STING was increased threefold. However,
overall STING expression showed only a small increase in
two of the Matrix-M treated pigs and as a group, this was
not significantly different to the control group. Thus, four
pigs treated with Matrix-M showed an increase in IFN-␤
expression and these pigs also expressed an elevated level
of OPN and/or STING. For three of these pigs (nos. 1, 2, and 3)
the sampled draining lymph node was enlarged at autopsy,
further indicating a potent immune activation.
4. Conclusion
The adjuvant Matrix-M induces an inflammatory
response characterized by a rapid influx of neutrophilic
granulocytes and activation of genes regulating early
inflammation and other immune response genes in
60 C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61
the pig. Functional annotation analysis and gene set
enrichment analysis specified a significant enrichment of
“myeloid cell” and pDC. The latter suggestion is well in
line with the distinct interferon-regulated gene profile
recorded both at the site of injection and in the draining
lymph node. In accordance the relative expression of
IFN-␤ was increased in the draining lymph node 24 h after
injection with Matrix-M. As the IFN-␤ expression was mir-
rored by an increased expression of OPN and STING both
the endosomal TLR7/TLR9 pathway including MyD88/OPN
and the cytosolic STING/TBK1 pathways could be involved
in Matrix-M mediated IFN activation, although the former
pathway seems like the preferred one. The Matrix com-
ponent inducing the interferon response still has to be
identified but the present Matrix-M formulation was well
tolerated in young piglets, signifying that the magnitude of
the inflammation was appropriate to initiate an immune
response without causing any adverse side reactions.
Acknowledgements
We are thankful for the excellent technical assistance of
the staff of the Prairie Diagnostic Services and the Prairie
Swine Centre of Saskatoon, Canada and by Ulla Schmidt
at the University Research Station, Funbo Lövsta, Upp-
sala, Sweden. Funding for this project was provided by
the EU 6th framework programme Food-CT-2004-513928,
Control of Porcine Circovirus Diseases (PCVDs): Towards
Improved Food Quality and Safety and by the Swedish
Research Council for Environment, Agricultural and Spatial
Planning (FORMAS).
References
Ahlberg, V., Lövgren Bengtsson, K., Wallgren, P., Fossum, C., 2012. Global
transcriptional response to ISCOM-Matrix adjuvant at the site of
administration and in the draining lymph node early after intramus-
cular injection in pigs. Dev. Comp. Immunol. 38, 17–26.
Auray, G., Facci, M.R., van Kessel, J., Buchanan, R., Babiuk, L.A., Gerdts,
V., 2010. Differential activation and maturation of two porcine DC
populations following TLR ligand stimulation. Mol. Immunol. 47,
2103–2111.
Azevedo, M.S., Gonzalez, A.M., Yuan, L., Jeong, K.I., Iosef, C., Van Nguyen,
T., Lövgren-Bengtsson, K., Morein, B., Saif, L.J., 2010. An oral versus
intranasal prime/boost regimen using attenuated human rotavirus
or VP2 and VP6 virus-like particles with immunostimulating com-
plexes influences protection and antibody-secreting cell responses to
rotavirus in a neonatal gnotobiotic pig model. Clin. Vaccine Immunol.
17, 420–428.
Bertho, N., Marquet, F., Pascale, F., Kang, C., Bonneau, M., Schwartz-
Cornil, I., 2011. Steady state pig dendritic cells migrating in skin
draining pseudo-afferent lymph are semi-mature. Vet. Immunol.
Immunopathol. 144, 430–436.
Bolind Bågenholm, A., 2009. Early immune response to and adjuvant
(AbISCO-100®
) tested in porcine peripheral blood mononuclear cells.
http://stud.epsilon.slu.se
Dalsgaard, K., Hilgers, L., Trouve, G., 1990. Classical and new approaches
to adjuvant use in domestic food animals. Adv. Vet. Sci. Comp. Med.
35, 121–159.
Dawson, H.D., Royaee, A.R., Nishi, S., Kuhar, D., Schnitzlein, W.M., Zucker-
mann Jr., F., Urban, J., Lunney, J.K., 2004. Identification of key immune
mediators regulating T helper 1 responses in swine. Vet. Immunol.
Immunopathol. 100, 105–111.
Duewell, P., Kisser, U., Heckelsmiller, K., Hoves, S., Stoitzner, P., Koernig, S.,
Morelli, A.B., Clausen, B.E., Dauer, M., Eigler, A., Anz, D., Bourquin, C.,
Maraskovsky, E., Endres, S., Schnurr, M., 2011. ISCOMATRIX adjuvant
combines immune activation with antigen delivery to dendritic cells
in vivo leading to effective cross-priming of CD8+ T cells. J. Immunol.
187, 55–63.
Fairbairn, L., Kapetanovic, R., Sester, D.P., Hume, D.A., 2011. The mononu-
clear phagocyte system of the pig as a model for understanding human
innate immunity and disease. J. Leukoc. Biol. 89, 855–871.
González, A.M., Nguyen, T.V., Azevedo, M.S., Jeong, K., Agarib, F., Iosef, C.,
Chang, K., Lövgren-Bengtsson, K., Morein, B., Saif, L.J., 2004. Antibody
responses to human rotavirus (HRV) in gnotobiotic pigs following a
new prime/boost vaccine strategy using oral attenuated HRV prim-
ing and intranasal VP2/6 rotavirus-like particle (VLP) boosting with
ISCOM. Clin. Exp. Immunol. 135, 361–372.
Hjertner, B., Olofsson, K.M., Lindberg, R., Fuxler, L., Fossum, C.,
2013. Expression of reference genes and T helper 17 asso-
ciated cytokine genes in the equine intestinal tract. Vet. J.,
http://dx.doi.org/10.1016/j.tvjl.2013.05.020 [Epub ahead of print].
Holm, C.K., Jensen, S.B., Jakobsen, M.R., Cheshenko, N., Horan, K.A., Moeller,
H.B., Gonzalez-Dosal, R., Rasmusse, S.B., Christensen, M.H., Yarovin-
sky, T.O., Rixon, F.J., Herold, B.C., Fitzgerald, K.A., Paludan, S.R., 2012.
Virus-cell fusion as a trigger of innate immunity dependent on the
adaptor STING. Nat. Immunol. 13, 343–737.
Hägglund, S., Hu, K.F., Larsen, L.E., Hakhverdyan, M., Valarcher, J.F., Taylor,
G., Morein, B., Belák, S., Alenius, S., 2004. Bovine respiratory syncy-
tial virus ISCOMs—protection in the presence of maternal antibodies.
Vaccine 23, 646–655.
Iosef, C., Van Nguyen, T., Jeong, K., Bengtsson, K., Morein, B., Kim, Y., Chang,
K.O., Azevedo, M.S., Yuan, L., Nielsen, P., Saif, L.J., 2002. Systemic and
intestinal antibody secreting cell responses and protection in gnotobi-
otic pigs immunized orally with attenuated Wa human rotavirus and
Wa 2/6-rotavirus-like-particles associated with immunostimulating
complexes. Vaccine 20, 1741–1753.
Ishikawa, H., Ma, Z., Barber, G.N., 2009. STING regulates intracellular DNA-
mediated, type I interferon-dependent innate immunity. Nature 461,
788–792.
Johansson, M., Lövgren-Bengtsson, K., 1999. Icoms with different quil-
laja saponin components differ in their immunomodulating activities.
Vaccine 17, 2894–2900.
Kapetanovic, R., Fairbairn, L., Beraldi, D., Sester, D.P., Archibald, A.L., Tug-
gle, C.K., Hume, D.A., 2012. Pig bone marrow-derived macrophages
resemble human macrophages in their response to bacterial
lipopolysaccharide. J. Immunol. 188, 3382–3394.
Kensil, C.R., Patel, U., Lennick, M., Marciani, D., 1991. Separation and char-
acterization of saponins with adjuvant activity from Quillaja saponaria
Molina cortex. J. Immunol. 146, 431–437.
Labbé, K., Saleh, M., 2011. Pyroptosis A Caspase-1-dependent pro-
grammed cells death and a barrier to infection. In: Couillin, I., Pétrilli,
V., Martinon, F. (Eds.), The Inflammasome, Progress in Inflammation
Research. Springer, Basel AG, pp. 17–36.
Lande, R., Ganguly, D., Facchinetti, V., Frasca, L., Conrad, C., Gregorio,
J., Meller, S., Chamilos, G., Sebasigari, R., Riccieri, V., Bassett, R.,
Amuro, H., Fukuhara, S., Ito, T., Liu, Y.J., Gilliet, M., 2011. Neutrophils
activate plasmacytoid dendritic cells by releasing self-DNA-peptide
complexes in systemic lupus erythematosus. Sci. Transl. Med. 3,
73ra19.
Levy, D.E., Marié, I.J., Durbin, J.E., 2011. Induction and function of type I
and III interferon in response to viral infection. Curr. Opin. Virol. 1,
476–486.
Lin, W., Qiu, Z., Liu, Q., Cui, S., 2013. Interferon induction and suppres-
sion in swine testicle cells by porcine parvovirus and its proteins. Vet.
Microbiol. 163, 157–161.
Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data
using real-time quantitative PCR and the 2(−Delta Delta C(T)) method.
Methods 25, 402–408.
Lövgren Bengtsson, K., Morein, B., 2000. The ISCOMTM
technology. In:
O’Hagan, D.T. (Ed.), Methods in Molecular Medicine, Vaccine Adju-
vant: Preparation Methods and Research Protocols. Humana Press,
Inc., Totowa, NJ, pp. 239–258.
Lövgren, K., Morein, B., 1988. The requirement of lipids for the formation of
immunostimulating complexes (iscoms). Biotechnol. Appl. Biochem.
10, 161–172.
Lövgren Bengtsson, K., Morein, B., Osterhaus, A.D., 2011. ISCOM
technology-based Matrix MTM
adjuvant: success in future vaccines
relies on formulation. Expert Rev. Vaccines 10, 401–403.
Lövgren Bengtsson, K., Sjölander, A., 1996. Adjuvant activity of iscoms;
effect of ratio and co-incorporation of antigen and adjuvant. Vaccine
14, 753–760.
Marquet, F., Bonneau, M., Pascale, F., Urien, C., Kang, C., Schwartz-Cornil,
I., Bertho, N., 2011. Characterization of dendritic cells subpopula-
tions in skin and afferent lymph in the swine model. PLoS ONE 6,
e16320.
Morein, B., Sundquist, B., Höglund, S., Dalsgaard, K., Osterhaus, A., 1984.
Iscom a novel structure for antigenic presentation of membrane pro-
teins from enveloped viruses. Nature 308, 457–460.
C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61 61
Morein, B., 1987. Potentiation of the immune response by immunization
with antigens in defined multimeric physical forms. Vet. Immunol.
Immunopathol. 17, 153–159.
Morein, B., 1988. The iscom antigen-presenting system. Nature 332,
287–288.
Morein, B., Blomqvist, G., Hu, K., 2007. Immune responsiveness in the
neonatal period. J. Comp. Pathol. 137, S27–S31.
Morelli, A.B., Becher, D., Koernig, S., Silva, A., Drane, D., Maraskovsky,
E., 2012. ISCOMATRIX: a novel adjuvant for use in prophylactic and
therapeutic vaccines against infectious diseases. J. Med. Microbiol. 61,
935–943.
Nguyen, T.V., Iosef, C., Jeong, K., Kim, Y., Chang, K.O., Lövgren-Bengtsson, K.,
Morein, B., Azevedo, M.S., Lewis, P., Nielsen, P., Yuan, L., 2003. Vaccine
21, 4059–4070.
Nguyen, T.V., Yuan, L., Azevedo, M.S., Jeong, K.I., Gonzalez, A.M., Iosef,
C., Lövgren-Bengtsson, K., Morein, B., Lewis, P., Saif, L.J., 2006a. High
titers of circulating maternal antibodies suppress effector and mem-
ory B-cell responses induced by an attenuated rotavirus priming and
rotavirus-like particle-immunostimulating complex boosting vaccine
regimen. Clin. Vaccine Immunol. 13, 475–485.
Nguyen, T.V., Yuan, L., Azevedo, M.S., Jeong, K.I., Gonzalez, A.M., Iosef, C.,
Lövgren-Bengtsson, K., Morein, B., Lewis, P., Saif, L.J., 2006b. Low titer
maternal antibodies can both enhance and suppress B cell responses to
a combined live attenuated human rotavirus and VLP-ISCOM vaccine.
Vaccine 24, 2302–2316.
Puentes, E., Cancio, E., Eiras, A., Nores, M.V., Aguilera, A., Regueiro, B.J.,
Seoane, R., 1993. Efficacy of various non-oily adjuvants in immuniza-
tion against the Aujeszky’s disease (pseudorabies) virus. Zentralbl.
Veterinarmed. B 40, 353–365.
Reimer, J.M., Karlsson, K.H., Lövgren-Bengtsson, K., Magnusson, S.E.,
Fuentes, A., Stertman, L., 2012. Matrix-MTM
adjuvant induces local
recruitment, activation and maturation of central immune cells in
absence of antigen. PLoS ONE 7, e41451.
Rönnberg, B., Fekadu, M., Morein, B., 1995. Adjuvant activity of non-toxic
Quillaja saponaria Molina components for use in ISCOM matrix. Vac-
cine 13, 1375–1382.
Straw, B.E., DAllaire, S., Mengeling, W.L., Taylor, D.J., 1999. Diseases of
Swine, 8th ed. Iowa State University Press, Ames, IA.
Shinohara, M.L., Lu, L., Bu, J., Werneck, M.B., Kobayashi, K.S., Glimcher, L.H.,
Cantor, H., 2006. Osteopontin expression is essential for interferon-
alpha production by plasmacytoid dendritic cells. Nat. Immunol. 7,
498–506.
Takahashi, H., Takeshita, T., Morein, B., Putney, S., Germain, R.N., Berzofsky,
J.A., 1990. Induction of CD8+ cytotoxic T cells by immunization with
purified HIV-1 envelope protein in ISCOMs. Nature 344, 873–875.
Tsuda, T., Sugimura, T., Murakami, Y., 1991. Evaluation of glycoprotein
gII ISCOMs subunit vaccine for pseudorabies in pig. Vaccine 9, 648–
652.
Tulman, E.R., Garmendia, A.E., 1994. Delivery of pseudorabies virus
envelope antigens enclosed in immunostimulating complexes
(ISCOMs): elicitation of neutralizing antibody and lymphoprolif-
erative responses in swine and protection in mice. Vaccine 12,
1349–1354.
Uddin, M.J., Cinar, M.U., Tesfaye, D., Looft, C., Tholen, E., Schellander, K.,
2011. Age-related changes in relative expression stability of com-
monly used housekeeping genes in selected porcine tissues. BMC Res.
Notes 4, 1–13.
Vandesompele, J., De Preter, K., Pattyn, F., Poppe, B., Van Roy, N., De Paepe,
A., Speleman, F., 2002. Accurate normalization of real-time quantita-
tive RT-PCR data by geometric averaging of multiple internal control
genes. Genome Biol., 3, research0034.1–0034.11.
Wallgren, P., Segall, T., Pedersen Mörner, A., Gunnarsson, A., 1999. Exper-
imental infections with Actinobacillus pleuropneumoniae in pigs—I.
Comparison of five different parenteral antibiotic treatments. Zen-
tralbl. Veterinarmed. B 46, 249–260.
White, F.J., Ross, J.W., Joyce, M.M., Geisert, R.D., Burghardt, R.C., Johnson,
G.A., 2005. Steroid regulation of cell specific secreted phosphopro-
tein 1 (Osteopontin) expression in the pregnant porcine uterus. Biol.
Reprod. 73, 1294–1301.
Wikström, F.H., Fossum, C., Fuxler, L., Kruse, R., Lövgren, T., 2011. Cytokine
induction by immunostimulatory DNA in porcine PBMC is impaired
by a hairpin forming sequence motif from the genome of Porcine Cir-
covirus type 2 (PCV2). Vet. Immunol. Immunopathol. 139, 156–166.
Xie, L., Liu, M., Fang, L., Su, X., Cai, K., Wang, D., Xiao, S., 2010. Molecu-
lar cloning and functional characterization of porcine stimulator of
interferon genes (STING). Dev. Comp. Immunol. 34, 847–854.

More Related Content

What's hot

(Carvalho, 2012) Development of production and purification of PspA3
(Carvalho, 2012) Development of production and purification of PspA3(Carvalho, 2012) Development of production and purification of PspA3
(Carvalho, 2012) Development of production and purification of PspA3
Rimenys Junior Carvalho
 
art_10.1007_s00262-015-1719-z (2)
art_10.1007_s00262-015-1719-z (2)art_10.1007_s00262-015-1719-z (2)
art_10.1007_s00262-015-1719-z (2)
Lior Lalush
 
IJAR-9267 BM AFCs Published in 1 4 2016
IJAR-9267 BM AFCs Published in 1 4 2016IJAR-9267 BM AFCs Published in 1 4 2016
IJAR-9267 BM AFCs Published in 1 4 2016
mohamed galal
 
Chitin gama delta linfocitos
Chitin gama delta linfocitosChitin gama delta linfocitos
Chitin gama delta linfocitos
UFRJ
 
Paracrine TGF_IL2 paper
Paracrine TGF_IL2 paperParacrine TGF_IL2 paper
Paracrine TGF_IL2 paper
Michael McHugh
 
J American Science
J American ScienceJ American Science
J American Science
1611974
 
Yamamoto Obiltoxaximab Prevents Disseminated Bacillus anthracis Infection 2016
Yamamoto Obiltoxaximab Prevents Disseminated Bacillus anthracis Infection 2016Yamamoto Obiltoxaximab Prevents Disseminated Bacillus anthracis Infection 2016
Yamamoto Obiltoxaximab Prevents Disseminated Bacillus anthracis Infection 2016
Annette Shadiack
 
Yamamoto Efficacy Projection of Obiltoxaximab for Treatment of Inhalational 2016
Yamamoto Efficacy Projection of Obiltoxaximab for Treatment of Inhalational 2016Yamamoto Efficacy Projection of Obiltoxaximab for Treatment of Inhalational 2016
Yamamoto Efficacy Projection of Obiltoxaximab for Treatment of Inhalational 2016
Annette Shadiack
 

What's hot (20)

(Carvalho, 2012) Development of production and purification of PspA3
(Carvalho, 2012) Development of production and purification of PspA3(Carvalho, 2012) Development of production and purification of PspA3
(Carvalho, 2012) Development of production and purification of PspA3
 
What is the latest fashion-Genetic Engineering
What is  the latest fashion-Genetic EngineeringWhat is  the latest fashion-Genetic Engineering
What is the latest fashion-Genetic Engineering
 
Oncotarget Paper
Oncotarget PaperOncotarget Paper
Oncotarget Paper
 
art_10.1007_s00262-015-1719-z (2)
art_10.1007_s00262-015-1719-z (2)art_10.1007_s00262-015-1719-z (2)
art_10.1007_s00262-015-1719-z (2)
 
IJAR-9267 BM AFCs Published in 1 4 2016
IJAR-9267 BM AFCs Published in 1 4 2016IJAR-9267 BM AFCs Published in 1 4 2016
IJAR-9267 BM AFCs Published in 1 4 2016
 
Chitin gama delta linfocitos
Chitin gama delta linfocitosChitin gama delta linfocitos
Chitin gama delta linfocitos
 
5
55
5
 
Evaluating Ozoralizumab (ATN-203) as a Novel Biotherapeutic Agent for the Tre...
Evaluating Ozoralizumab (ATN-203) as a Novel Biotherapeutic Agent for the Tre...Evaluating Ozoralizumab (ATN-203) as a Novel Biotherapeutic Agent for the Tre...
Evaluating Ozoralizumab (ATN-203) as a Novel Biotherapeutic Agent for the Tre...
 
191 195
191 195191 195
191 195
 
Paracrine TGF_IL2 paper
Paracrine TGF_IL2 paperParacrine TGF_IL2 paper
Paracrine TGF_IL2 paper
 
J American Science
J American ScienceJ American Science
J American Science
 
The Role of Transgenic Animals in Biomedical Research
The Role of Transgenic Animals in Biomedical Research The Role of Transgenic Animals in Biomedical Research
The Role of Transgenic Animals in Biomedical Research
 
Yamamoto Obiltoxaximab Prevents Disseminated Bacillus anthracis Infection 2016
Yamamoto Obiltoxaximab Prevents Disseminated Bacillus anthracis Infection 2016Yamamoto Obiltoxaximab Prevents Disseminated Bacillus anthracis Infection 2016
Yamamoto Obiltoxaximab Prevents Disseminated Bacillus anthracis Infection 2016
 
Poster pdf
Poster pdfPoster pdf
Poster pdf
 
Transgenic
TransgenicTransgenic
Transgenic
 
Yamamoto Efficacy Projection of Obiltoxaximab for Treatment of Inhalational 2016
Yamamoto Efficacy Projection of Obiltoxaximab for Treatment of Inhalational 2016Yamamoto Efficacy Projection of Obiltoxaximab for Treatment of Inhalational 2016
Yamamoto Efficacy Projection of Obiltoxaximab for Treatment of Inhalational 2016
 
PAPER1
PAPER1PAPER1
PAPER1
 
Transgenic animals
Transgenic animalsTransgenic animals
Transgenic animals
 
Drug Discovery Project Presentation
Drug Discovery Project Presentation Drug Discovery Project Presentation
Drug Discovery Project Presentation
 
Ab
AbAb
Ab
 

Viewers also liked (10)

KFS GROUP Introduction
KFS GROUP Introduction KFS GROUP Introduction
KFS GROUP Introduction
 
Pengaruh Infeksi Odontogen terhadap Timbulnya Urtikaria
Pengaruh Infeksi Odontogen terhadap Timbulnya UrtikariaPengaruh Infeksi Odontogen terhadap Timbulnya Urtikaria
Pengaruh Infeksi Odontogen terhadap Timbulnya Urtikaria
 
Allegra
AllegraAllegra
Allegra
 
TANu, Klout et SSI, boostez votre CV avec des indicateurs numériques
TANu, Klout et SSI, boostez votre CV avec des indicateurs numériquesTANu, Klout et SSI, boostez votre CV avec des indicateurs numériques
TANu, Klout et SSI, boostez votre CV avec des indicateurs numériques
 
M1 PART-A
M1 PART-AM1 PART-A
M1 PART-A
 
soldadura
soldadurasoldadura
soldadura
 
CAIXA - fisl 2011
CAIXA -  fisl 2011CAIXA -  fisl 2011
CAIXA - fisl 2011
 
El Barcelonès
El BarcelonèsEl Barcelonès
El Barcelonès
 
CLASIFICACIÓN DE LOS CONTRATOS (PARTE UNO)
CLASIFICACIÓN DE LOS CONTRATOS (PARTE UNO)CLASIFICACIÓN DE LOS CONTRATOS (PARTE UNO)
CLASIFICACIÓN DE LOS CONTRATOS (PARTE UNO)
 
Lamark (1)
Lamark (1)Lamark (1)
Lamark (1)
 

Similar to Paper 2 Navisraj

Life Sci.J
Life Sci.JLife Sci.J
Life Sci.J
1611974
 
PlantEtMIC1_authors_copy
PlantEtMIC1_authors_copyPlantEtMIC1_authors_copy
PlantEtMIC1_authors_copy
Kota Sathish
 
Immunohistological study
Immunohistological studyImmunohistological study
Immunohistological study
Nasrin Lucky
 
IBMX part I -rabbit reproduction Theriogenology 2010 (1)
IBMX part I -rabbit reproduction Theriogenology 2010 (1)IBMX part I -rabbit reproduction Theriogenology 2010 (1)
IBMX part I -rabbit reproduction Theriogenology 2010 (1)
ELENI FOTOPOULOU
 
The molecular characterisation of Escherichia coli K1 isolated from neonatal ...
The molecular characterisation of Escherichia coli K1 isolated from neonatal ...The molecular characterisation of Escherichia coli K1 isolated from neonatal ...
The molecular characterisation of Escherichia coli K1 isolated from neonatal ...
Pauline Ogrodzki
 
A panel of recombinant monoclonal antibodies against zebrafish
A panel of recombinant monoclonal antibodies against zebrafishA panel of recombinant monoclonal antibodies against zebrafish
A panel of recombinant monoclonal antibodies against zebrafish
Shahnaz Yusaf
 
The role of surface charge of ISCOMATRIX nanoparticles on the type of immune ...
The role of surface charge of ISCOMATRIX nanoparticles on the type of immune ...The role of surface charge of ISCOMATRIX nanoparticles on the type of immune ...
The role of surface charge of ISCOMATRIX nanoparticles on the type of immune ...
Nanomedicine Journal (NMJ)
 
Micro-emulsion Methods paper
Micro-emulsion Methods paperMicro-emulsion Methods paper
Micro-emulsion Methods paper
Michael Weiner
 
Draft Genome Sequence of an Enterobacter Species Associated with Illnesses an...
Draft Genome Sequence of an Enterobacter Species Associated with Illnesses an...Draft Genome Sequence of an Enterobacter Species Associated with Illnesses an...
Draft Genome Sequence of an Enterobacter Species Associated with Illnesses an...
Pauline Ogrodzki
 

Similar to Paper 2 Navisraj (20)

Abstract conference mbsmb 2009
Abstract conference mbsmb 2009Abstract conference mbsmb 2009
Abstract conference mbsmb 2009
 
Presentation.pptx
Presentation.pptxPresentation.pptx
Presentation.pptx
 
Barkai2013
Barkai2013Barkai2013
Barkai2013
 
Life Sci.J
Life Sci.JLife Sci.J
Life Sci.J
 
PlantEtMIC1_authors_copy
PlantEtMIC1_authors_copyPlantEtMIC1_authors_copy
PlantEtMIC1_authors_copy
 
My CV_final
My CV_finalMy CV_final
My CV_final
 
Immunohistological study
Immunohistological studyImmunohistological study
Immunohistological study
 
IBMX part I -rabbit reproduction Theriogenology 2010 (1)
IBMX part I -rabbit reproduction Theriogenology 2010 (1)IBMX part I -rabbit reproduction Theriogenology 2010 (1)
IBMX part I -rabbit reproduction Theriogenology 2010 (1)
 
FMDV
FMDVFMDV
FMDV
 
The molecular characterisation of Escherichia coli K1 isolated from neonatal ...
The molecular characterisation of Escherichia coli K1 isolated from neonatal ...The molecular characterisation of Escherichia coli K1 isolated from neonatal ...
The molecular characterisation of Escherichia coli K1 isolated from neonatal ...
 
53
5353
53
 
recent development in culture od Cestode
recent development in culture od Cestoderecent development in culture od Cestode
recent development in culture od Cestode
 
Ciprofloxacin resideu and their impact on Biomolecules n eggs of laying hens ...
Ciprofloxacin resideu and their impact on Biomolecules n eggs of laying hens ...Ciprofloxacin resideu and their impact on Biomolecules n eggs of laying hens ...
Ciprofloxacin resideu and their impact on Biomolecules n eggs of laying hens ...
 
Enterocin 55 produced by non rabbit-derived strain Enterococcus faecium EF55 ...
Enterocin 55 produced by non rabbit-derived strain Enterococcus faecium EF55 ...Enterocin 55 produced by non rabbit-derived strain Enterococcus faecium EF55 ...
Enterocin 55 produced by non rabbit-derived strain Enterococcus faecium EF55 ...
 
Muthamilselvan et al-2015-plant_biotechnology_journal
Muthamilselvan et al-2015-plant_biotechnology_journalMuthamilselvan et al-2015-plant_biotechnology_journal
Muthamilselvan et al-2015-plant_biotechnology_journal
 
A panel of recombinant monoclonal antibodies against zebrafish
A panel of recombinant monoclonal antibodies against zebrafishA panel of recombinant monoclonal antibodies against zebrafish
A panel of recombinant monoclonal antibodies against zebrafish
 
The role of surface charge of ISCOMATRIX nanoparticles on the type of immune ...
The role of surface charge of ISCOMATRIX nanoparticles on the type of immune ...The role of surface charge of ISCOMATRIX nanoparticles on the type of immune ...
The role of surface charge of ISCOMATRIX nanoparticles on the type of immune ...
 
Micro-emulsion Methods paper
Micro-emulsion Methods paperMicro-emulsion Methods paper
Micro-emulsion Methods paper
 
Draft Genome Sequence of an Enterobacter Species Associated with Illnesses an...
Draft Genome Sequence of an Enterobacter Species Associated with Illnesses an...Draft Genome Sequence of an Enterobacter Species Associated with Illnesses an...
Draft Genome Sequence of an Enterobacter Species Associated with Illnesses an...
 
Resume - Vasu Ayyappan
Resume - Vasu AyyappanResume - Vasu Ayyappan
Resume - Vasu Ayyappan
 

Paper 2 Navisraj

  • 1. Veterinary Immunology and Immunopathology 158 (2014) 53–61 Contents lists available at ScienceDirect Veterinary Immunology and Immunopathology journal homepage: www.elsevier.com/locate/vetimm Research paper Early inflammatory response to the saponin adjuvant Matrix-M in the pig Caroline Fossuma,∗ , Bernt Hjertnera , Viktor Ahlberga , Wasin Charerntantanakula,b , Kathy McIntoshc , Lisbeth Fuxlera , Navisraj Balagunaseelana , Per Wallgrend , Karin Lövgren Bengtssone a Department of Biomedicine and Veterinary Public Health, Section for Immunology, Swedish University of Agricultural Sciences, P.O. Box 588, SE-751 23 Uppsala, Sweden b Research Laboratory for Immunity Enhancement in Humans and Domestic Animals Maejo University, Chiang Mai 50290, Thailand c Department of Veterinary Microbiology, University of Saskatchewan, Western College of Veterinary Medicine, Saskatoon, Canada d National Veterinary Institute, SVA, SE-751 89 Uppsala, Sweden e Isconova AB, Kungsgatan 109, SE-753 18 Uppsala, Sweden a r t i c l e i n f o Article history: Received 3 December 2012 Received in revised form 20 July 2013 Accepted 23 July 2013 Keywords: Matrix-M ISCOM Pig Transcriptional NETs IRGs Adjuvant a b s t r a c t The early inflammatory response to Matrix-M was evaluated in pigs. Adverse reactions measured as body temperature, appetite, activity level and reaction at the site of injection were not observed after s.c. injection with three doses of the adjuvant (75, 100 or 150 ␮g) into one week old piglets. Analyses of the immediate cytokine response of PBMC after in vitro exposure to Matrix-M (AbISCO-100® ) revealed only a low expression of mRNA for tumour necrosis factor-␣ (p < 0.05) after 6 h incubation. Histological examination revealed an infiltration of leukocytes, haemorrhage and necrosis in muscle 24 h after i.m. injection of 150 ␮g Matrix-M in pigs aged eleven weeks. At this time, different grades of reactive lym- phoid hyperplasia were recorded in the draining lymph node that was enlarged in three of these six pigs injected with Matrix-M. The global transcriptional response at the site of injection and in the draining lymph node was analyzed using Affymetrix GeneChip Porcine Genome Array. A significant enrichment of gene signatures for the cell types described as “myeloid cells” and “plasmacytoid dendritic cells” was observed at the site of injection in Matrix-M injected pigs compared with pigs injected with saline. A number of genes encod- ing cytokines/chemokines or their receptors were upregulated at the injection site as well as in the draining lymph node. In the draining lymph node, a majority of the upregulated genes were interferon-regulated genes (IRGs). The expression of IFN-␤, but not IFN-␣, was increased in the draining lymph nodes of a majority of the pigs exposed to Matrix-M. These IFN-␤ expressing pigs also expressed increased levels of osteopontin (OPN) or stimulator of interferon genes (STING), two factors known to facilitate the expression of type I IFNs in response to viral infection. Thus, Matrix-M does not appear to induce any harmful inflam- matory response in piglets whilst contributing to the innate immunity by activating the type I IFN system, possibly through several alternative signalling pathways. © 2013 Elsevier B.V. All rights reserved. ∗ Corresponding author. Tel.: +46 018 4714056; fax: +46 018 4714382. E-mail address: Caroline.Fossum@slu.se (C. Fossum). 1. Introduction The immune-stimulating complex (ISCOM) is an antigen-presenting system in which antigens are incor- porated into a matrix of the saponin adjuvant Quil A, formulated into particles together with cholesterol 0165-2427/$ – see front matter © 2013 Elsevier B.V. All rights reserved. http://dx.doi.org/10.1016/j.vetimm.2013.07.007
  • 2. 54 C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61 and phospholipid (Morein et al., 1984). The formulation soon proved useful for a number of membrane-derived antigens for induction of the protective immunity to various microorganisms (Morein, 1987, 1988). Electron microscopy demonstrated the formation of a spherical cage-like structure, 40 nm in size, regardless of the source of antigen. A standardized methodology and mixture of Quil A, cholesterol and phophatidylcholine was established for incorporation of amphipathic antigens (reviewed in Lövgren Bengtsson and Morein, 2000). Later, it was realized that incorporation of antigens into the ISCOM structure was not necessary for potent immune stimulation (Lövgren and Morein, 1988; Lövgren Bengtsson and Sjölander, 1996). Pre-formed ISCOMs without incorporated antigen, called ISCOM-Matrix and later referred to as Matrix, became used as stand-alone adjuvant, simply mixed with antigens. The glycosidic saponins, extracted from the bark of the tree Quillaja saponaria Molina (Quil A), in free form, have been used as adjuvant for almost a century (reviewed in Dalsgaard et al., 1990). Quil A is a potent adjuvant; how- ever, free saponins have haemolytic activity that may cause side effects. By formulation of saponins with cholesterol and phospholipids in ISCOMs or in Matrix particles, the haemolytic activity is abolished and a more potent and less reactogenic adjuvant is created. The Matrix made from Quil A or similar preparations are currently called Matrix-Q. Biochemical separation techniques revealed that the lytic and structure forming capacities of Quil A were mainly restricted to different fractions (Kensil et al., 1991; Rönnberg et al., 1995) and that various combinations of these components affected the adjuvant properties considerably (Johansson and Lövgren-Bengtsson, 1999). Detailed biochemical and functional characterisations of the saponin fractions have resulted in a very well- tolerated Matrix formulation called Matrix-M. Matrix-M is a mixture of Matrix particles made from two dif- ferent purified fractions of Quillaja derived saponins. Matrix-MTM (Isconova AB, Uppsala, Sweden) is a potent adjuvant that is licensed for several animal vaccines and now also has entered human clinical trials (Lövgren Bengtsson et al., 2011). Another similar, albeit differ- ent product based on the ISCOM technology is the ISCOMATRIX® adjuvant (CSL; Commonwealth Serum Lab- oratories, Melbourne, Australia). MatrixTM formulations are available in various forms and recommended for dif- ferent species according to their saponin sensitivity and there are two preparations available for research pur- poses, AbISCO-100® (Matrix-M type) and AbISCO-300® (Matrix-Q type). Attempts to explore the power of Matrix formula- tions on immune parameters have mainly been carried out in mice. These studies have revealed a prominent recruitment and activation of cells in the draining lymph node/spleen, antigen delivery to dendritic cells accompanied by cytokine and chemokine production. This allows for cross-presentation with the subsequent induction of cytotoxic T cells and a long-lasting antibody response (Duewell et al., 2011; Morelli et al., 2012; Reimer et al., 2012). However, which molecular path- way(s) are activated by the Matrix of ISCOM is still not clarified. In the pig, early experimental ISCOM/Matrix-Q vac- cines focused on Aujeszky’s Disease Virus (Tsuda et al., 1991; Puentes et al., 1993; Tulman and Garmendia, 1994), rotavirus (Iosef et al., 2002; Nguyen et al., 2003; González et al., 2004; Nguyen et al., 2006a; Azevedo et al., 2010), and the use of virus-like particles in combination with ISCOM- Matrix (Tulman and Garmendia, 1994). The latter concept has been used successfully in young pigs, inducing an active immune response even in the presence of maternal immu- nity (Nguyen et al., 2006b; McIntosh et al., manuscript) as also shown for other ISCOM formulations applied in calves (Hägglund et al., 2004) or mice (Morein et al., 2007). Taken together, Matrix-M appears as a promising adju- vant also in the pig where improved vaccines to several diseases are desirable. The pig is also a valuable model for studies of adjuvant effects in man because of many simi- larities between the species in the organization of innate immune cells and their cytokine production (Auray et al., 2010; Bertho et al., 2011; Faibairn et al., 2011; Marquet et al., 2011; Kapetanovic et al., 2012). To reflect the early inflammatory response to Matrix-M in the pig, results from in vitro studies using porcine blood mononuclear or polymorphonuclear cells are reported here together with results from an in vivo toxicity test carried out in piglets. The findings are related to the global transcriptional response to Matrix-M recorded in pigs at the site of injection and in the draining lymph node (Ahlberg et al., 2012). 2. Materials and methods 2.1. Animals and experimental designs Five experimental set ups were used to study the early inflammatory response to the Matrix of ISCOMs in the pig. An animal toxicity study was carried out in one-week old PIC-crossbred piglets, housed in a 600 sow commercial swine farrow-to-finish facility in Saskatchewan, Canada (Set I). In vitro cellular toxicity studies were performed with PBMC (Set II) and polymorphonuclear neutrophil leuko- cytes (PMNL; Set III) purified from the blood of finishing pigs housed in a specific pathogen free herd (Wallgren et al., 1999; Swedish Livestock Research Centre, Lövsta-Uppsala, Sweden). In vitro induced expression of cytokine mRNA (Set IV) was determined using PBMC obtained from convention- ally reared pigs at the University Research Station (Funbo Lövsta, Uppsala, Sweden) whereas in vivo induced expres- sion of mRNA for interferon-related genes (Set V) was studied in lymph nodes obtained from 11 weeks old SPF- pigs (Ahlberg et al., 2012). All procedures were conducted in accordance with the University of Saskatchewan’s Com- mittee for Animal Care and Supply (permit #20060004) and with the Ethical Committee for Animal Experiments, Uppsala, Sweden. 2.2. In vivo evaluation of Matrix-Q toxicity (Set I) The potential for adverse reactions was tested by the s. c. injection of variable doses of Matrix-Q (Isconova AB, Uppsala, Sweden) into one-week-old piglets. Piglets were injected with either 75 ␮g (n = 3), 100 ␮g (n = 3), or 150 ␮g (n = 3) of the Matrix-Q diluted in sterilized 0.01 M PBS to
  • 3. C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61 55 a final volume of 200 ␮L using a 1 mL-25 gauge needle. A single injection was administered subcutaneously 1 cm off the dorsal midline between the shoulders and piglets were observed 7 times over a period of 30 h for rectal temper- ature, injection site reactivity (swelling, redness, or pain), and activity level (active or lethargic). 2.3. In vitro evaluation of Matrix-M toxicity (Set II) The viability of PBMC was evaluated after 18 h incu- bation in the presence of 0.3, 1 or 3 ␮g Matrix-M (AbISCO-100®; Isconova AB) per ml culture medium. PBMCs were isolated from heparinized blood by density gradient centrifugation on Ficoll-Paque (Pharmacia, Upp- sala, Sweden). The cell viability was determined by flow cytometry (FACSCanto flow cytometer; BD Biosciences, San Jose, CA) using the Annexin V-FITC Apoptosis Detec- tion Kit I with Propidium Iodide (PI) staining solution (BD PharmingenTM, San Jose, CA) as recommended. Parallel cul- tures with PBMC incubated in growth medium or treated with UV-light (480 mJ for 90 s) before incubation were included as negative and positive controls, respectively. Ten thousand events per sample were collected and the data were analyzed using the FACSDiva software (v. 5.0.2, BD Biosciences). 2.4. In vitro effects of Matrix-M on PMNL activity (Set III) Heparinized blood was diluted with an equal volume of 3% Dextran (T-2000, Pharmacia Biotech, Uppsala, Sweden) and allowed to sediment for 30 min at RT. The leucocyte rich fraction was washed once in PBS, resuspended and layered on a discontinuous gradient of 70% and 80% Per- coll (GE Healthcare, Uppsala, Sweden). After centrifugation (300 × g) for 25 min, cells in the two bands generated were collected, the cell numbers were counted and the purity was determined on cytospin glasses stained with Diff Quick (Vector Lab Inc., Burligame, CA). The cells recovered on the 80% Percoll cushion were enriched for PMNL (91.8 ± 6.1%, n = 20) and used for studies of the formation of neutrophil extracellular traps (NETs). The PMNL were washed twice in PBS and resuspended in RPMI 1640 supplemented with HEPES, l-glutamine antibi- otics and 2% BSA (Sigma–Aldrich®). Two millilitre cell suspension (107 neutrophils per ml) was seeded on cov- erslips in six well plates (Costar®) and incubated for 1 h at 37 ◦C. Thereafter, 400 ␮l medium (negative control) or phorbol 12-myristate 13-acetate (PMA; Sigma–Aldrich®) at a final concentration of 100 nM (positive control) were added to the wells. The effect of Matrix-M was tested at the final concentrations of 0.3, 1 and 3 ␮g/ml in parallel cultures. After 4 h of incubation the medium was care- fully removed from the wells and 1 ml of 50 nM Sytox Green (Invitrogen) was added to each well and incu- bated in the dark for 10 min. Thereafter formaldehyde (ProlaboTM, Taipan, Malaysia) was added to a final con- centration of 4% per well and incubated for 30 min before the coverslip was carefully removed and allowed to dry before mounted in Vectashield with DAPI (Vector Lab Inc., Burligame, CA). The morphology of cell nucleus and extra- cellular DNA fragments were determined in a fluorescence microscope (Nikon, Microphot-FX, Nikon Instruments Inc., Tokyo, Japan) equipped with a digital camera (Coolpix 990, Nikon) for documentation. 2.5. In vitro Matrix-M induced expression of cytokine genes (Set IV) The effect of Matrix-M on the expression of cytokine mRNA by porcine PBMC was tested in vitro and com- pared to that induced by the CpG-ODN 2216 (Cybergene AB, Huddinge, Sweden) or LPS (Sigma Aldrich, Stein- heim, Germany), using methods detailed elsewhere (Bolind Bågenholm, 2009; Wikström et al., 2011). All inducers were diluted in RPMI 1640 medium (BioWhittaker, Cambrex Bioscience, Verviers, Belgium) supplemented with 20 mM HEPES buffer, 2 mM l-glutamine, 200 IU penicillin/ml, 100 ␮g/ml streptomycin, 0.5 ␮M 2-mercaptoethanol and 5% foetal calf serum (Invitrogen, Life Technologies, Carls- bad, CA, USA). Three ml cell cultures (final concentration 5 × 106 cells per ml) were established in 25 cm2 tissue cul- ture flasks (Nunc, Roskilde, Denmark) containing Matrix-M (1 ␮g/ml), LPS (10 ␮g/ml), ODN 2216 (5 ␮g/ml) or only growth medium. After 6 hours incubation at 37 ◦C, total RNA was extracted, quality tested and DNA:se treated before 2 ␮g of RNA was used for cDNA synthesize as described (Wikström et al., 2011). Real-time TaqMan PCR was performed for IFN-␣, IFN- ␥, IL-6, IL-10, IL-12p40, IL-1␤, TNF-␣, TGF-␤, CXCR4, RGS16 and two reference genes, Cyclophilin A (CyA) and Hypoxanthine-guanine phosphoribosyl transferase (HPRT), using previously published primers and protocols (Wikström et al., 2011). The relative expression of target genes induced by Matrix-M, LPS or ODN 2216 was com- pared to their expression in PBMC cultured in plain growth medium according to Livak and Schmittengen (2001), using Cy A and HPRT as reference genes. 2.6. In vivo transcriptional response to Matrix-M (Set V) Pigs were injected i.m. with either 150 ␮g Matrix-M suspended in 1 ml sterile endotoxin-free 0.9% NaCl solu- tion (n = 6) or just saline (n = 6). Twenty four h after injection, pigs were necropsied and tissues from the site of injection and their draining lymph nodes were col- lected and analyzed for early transcriptional response using Affymetrix GeneChip® Porcine Genome Array as detailed in Ahlberg et al. (2012). This was followed up by a more comprehensive expression analysis focusing on IFN-␣, IFN- ␤, osteopontin (OPN) and stimulator of interferon genes (STING) in the draining lymph node. Sample preparation, RNA extraction and analysis, and cDNA construction are detailed in Ahlberg et al. (2012). Primer pairs for IFN-␣, IFN-␤, OPN, riboso- mal protein L32 (RPL32), STING and tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activa- tion protein (YWHAZ) were chosen from published works favouring those spanning an intron, and reoptimized in house (Table 1). SYBR Green PCR was run as described previously (Hjertner et al., 2013). Using PCR base line normalized Cq values the expression of IFN-␣, IFN-␤, OPN and STING was normalized to the geometric average
  • 4. 56 C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61 Table 1 Primer details and gene specific optimized conditions. Gene Primer sequence Primer location Target sequence Anneal temp (◦ C) Primer conc (nM) E (%)g r2 Melt point (◦ C) IFN-␣a F:AGCCTCCTGCACCAGTTCTG 346–365 NM 214393.1 60 500 100 0.997 84.5 R: TCACAGCCAGGATGGAGTCC 469–450 IFN-␤b F: TAGCACTGGCTGGAATGAAACC 288–309 NM 001003923.1 58 400 104 0.993 79.5 R: TCAGGTGAAGAATGGTCATGTCT 427–405 OPNc F: TTGGACAGCCAAGAGAAGGACAGT 731–754 NM 214023.1 56 300 93 0.997 82.5 R: GCTCATTGCTCCCATCATAGGTCTTG 851–826 RPL32d F: CGGAAGTTTCTGGTACACAATGTAA 249–273 NM 001001636.1 55 300 97 0.997 77 R: TGGAAGAGACGTTGTGAGCAA 342–322 STINGe F: TTACATCGGGTACCTGCGGC 489–508 NM 001142838.1 56 500 101 0.992 82 R: CCGAGTACGTTCTTGTGGCG 572–553 YWHAZf F: ATTGGGTCTGGCCCTTAACT 961–980 XM 001927228.4 58 400 101 0.997 78.5 R: GCGTGCTGTCTTTGTATGACTC 1106–1085 a Wikström et al. (2011). b Lin et al. (2013). c White et al. (2005). d Dawson et al. (2004). e Xie et al. (2010). f Uddin et al. (2011). g Efficiency from serial dilutions of reference cDNA. of RPL32 and YWHAZ (Vandesompele et al., 2002), and calibrated to the average expression in all six control pigs. 2.7. Statistical analyses Statistical differences in mRNA expression for IFN-␣, IFN-␤, STING and OPN between pigs injected with Matrix- M or saline were analyzed using the non-parametric Mann–Whitney test whereas statistical differences for var- ious in vitro treatments were determined using the paired Student’s t-test (GraphPad Prism version 5.0 for Mac OS X, GraphPad Software, San Diego, CA). p-Values ≤ 0.05 were regarded as significant. For all statistical analyses detailed in Ahlberg et al. (2012), q-values, i.e., p-values corrected for multiple testing were used. 3. Results and discussion 3.1. In vivo evaluation of Matrix-Q toxicity (Set I) The safety of saponin-based Matrix adjuvants in pigs was studied with Matrix-Q in one-week-old piglets. No adverse reactions were observed at the injection site nor was the activity level of any piglet during the 30 h period after injection with the Matrix-Q affected. The expected normal rectal temperature of a suckling piglet >24 h of age is 39.2 ± 0.3 ◦C (Straw et al., 1999). Taking into consideration the baseline rectal temperature of each piglet (temperature prior to injection), one piglet at 30 h post-injection recorded a low grade fever of 39.6 ◦C (a tem- perature greater than 0.3 ◦C above its baseline temperature and greater than 39.5 ◦C). However, this piglet received the lowest dose of Matrix-Q of 75 ␮g. 3.2. In vitro evaluation of Matrix-M toxicity (Set II) The effect of Matrix-M on cell survival was tested in vitro using PBMC obtained from five pigs (Fig. 1). After 18 h of culture the proportion of Annexin labelled cells M edium 0.3 M atrix 1 M atrix 3 M atrix U V 0 5 10 15 20 Percentage a M edium 0.3 M atrix 1 M atrix 3 M atrix U V 0 20 40 60 80 100 Percentage b Fig. 1. Percentage of apoptotic (a) or dead (b) porcine PBMC after 18 h of incubation in the presence of 0.3, 1 or 3 ␮g per ml of Matrix-M. The cor- responding figures for PBMC incubated in medium or UV-treated before incubation are included as negative and positive controls, respectively.
  • 5. C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61 57 that were negative for PI was in mean 12.6 ± 4.6% for cells incubated in the presence of Matrix-M, regardless of concentration. This was similar to cells incubated in growth medium (11.7 ± 2.7%) but higher than for those that were UV-treated (6.7 ± 3.5%) prior to incubation. A great proportion (74.5 ± 16.4%) of the UV-treated PBMC was however stained with both PI and Annexin indicat- ing a low cell survival. The corresponding figure for PBMC grown in various concentrations of Matrix M (12.2 ± 3.7%) was again very similar to that for PBMC grown in culture medium (12.6 ± 2.3%). Thus, Matrix M did not induce apo- ptosis or increase cell death during the present culture conditions. 3.3. In vitro effects of Matrix-M on PMNL activity (Set III) The ability of Matrix-M to induce formation of NETs was tested in vitro using porcine PMNL obtained from pigs in a SPF herd. The number of PMNL recovered per ml blood was low due to the high health status, and prior activation of them in vivo was less likely than for PMNL obtained from conventionally reared pigs. In accordance, no signs of acti- vation were evident after 4 h incubation in plain growth medium (Fig. 3a) whereas a high proportion of neutrophils with a condensed nucleus, showing signs of NETosis and release of NETs was observed among the neutrophils incu- bated with PMA (Fig. 3b and c). The effect of Matrix-M on the neutrophils was less evident. In the cultures supplemented with 0.3 or 1.0 ␮g Matrix-M per ml (Fig. 3d and e) the nucleus of the neu- trophils showed the same shape as those incubated in plain growth medium. Among neutrophils cultured in the pres- ence of 3 ␮g Matrix-M per ml (Fig. 3f) a slightly higher proportion of the neutrophils had a condensed nuclei but no NET formation was detected. Prolonged incubation, up to 16 h, affected the survival of the neutrophils regardless of culture set up. Still, no NET formation was observed in cultures supplemented with Matrix-M or in the cultures with plain growth medium. Instead, the nuclei of PMNL grown over night in the presence of Matrix-M were dis- integrated into small Sytox-stained fragments dispersed in considerably swelled cells or around disrupted cells, giving the impression of a pyroptotic cell death. The activation of cathepsin genes (CTSB, CTSD, CTSH, CTSS and CTSZ) at the injection site and the up regulation of the genes for IL-1␤ and IL-18 (Table 5, Ahlberg et al., 2012) agree with pyroptosis (Labbé and Saleh, 2011). 3.4. Early cytokine response of PBMC exposed to Matrix-M in vitro (Set IV) Analyses of the immediate cytokine response of PBMC after in vitro exposure to Matrix-M revealed a low level induction of TNF-␣ mRNA (p < 0.05) after 6 h incubation. The mRNA expression for the other cytokines analyzed did not differ from that of PBMC cultured in plain growth medium. In comparison, mRNA specific for IL-1␤, IL-6 and IL-10 (p < 0.001), and IL-12p40 and TNF-␣ (p < 0.01) was induced after 6 h exposure of the PBMC to LPS, and IFN- ␣ mRNA (p < 0.01) was induced after 6 h in the presence of the ODN 2216. Thus, Matrix-M did not evoke any strong Table 2 Significant differences in relative mRNA expression determined by qPCR. RNA was isolated after 6 h culture of porcine PBMC in the presence of Matrix-M (1 ␮g/ml), LPS (10 ␮g/ml), ODN 2216 (5 ␮g/ml) or in plain growth medium. Target gene Matrix-M vs. medium (p value) LPS vs. medium (p value) ODN 2216 vs. medium (p value) IFN-␣ ns * ** IFN-␥ ns ns * IL-6 ns *** * IL-10 ns *** * IL-1␤ ns *** ns TNF-␣ * ** * TGF-␤ ns * ns IL-12p40 ns ** * CXCR4 ns ns ** RGS16 ns ns ns ns: not significant. * p ≤ 0.05. ** p ≤ 0.01. *** p ≤ 0.001. immediate pro-inflammatory cytokine response in porcine PBMC when tested in vitro. These results are in agreement with the absence of fever and local reactions in the tox- icity tests but caution should be taken regarding the fact that these studies were performed in cultures of purified lymphoid cells, devoid of PMNL and possibly also lacking important soluble factors produced by other cells. 3.5. Global transcriptional response to Matrix-M at the site of administration and in draining lymph node (Set V) In order to better reflect the immune stimulatory prop- erties of Matrix-M the global transcriptional response was analyzed following i.m. administration of 150 ␮g Matrix-M in pigs aged eleven weeks (Ahlberg et al., 2012). Histo- logical examination revealed an infiltration of leukocytes, haemorrhage and necrosis in muscle after 24 h. At this time, different grades of reactive lymphoid hyperplasia were recorded in the draining lymph node that showed a Fig. 2. Interferon-regulated genes (IRGs) upregulated at the injection site (n = 36) and/or draining lymph node (n = 38) 24 h after Matrix-M admin- istration. The ten most up-regulated IRGs in each group are listed. IRGs were defined by the database Interferome (http://www.interferome.org).
  • 6. 58 C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61 moderate enlargement in three of the six pigs injected with Matrix-M. These observations (Table 2 and Fig. 1; Ahlberg et al., 2012) were well in line with the absence of visi- ble signs of inflammation when pigs were injected with a similar dose of Matrix-Q and the marginal indication of a pro-inflammatory cytokine response in porcine PBMC exposed to Matrix-M in vitro. As reported in Ahlberg et al. (2012), an inflammatory response to Matrix-M at the injection site was evident from the enrichment of genes in gene ontology (GO) terms such as ‘immune response’ (q = 1.4E−10), ‘response to wounding’ (q = 1.4E−9), ‘defence response’ (q = 5.1E−9), ‘inflammatory response’ (q = 7.4E−9), ‘positive regula- tion of immune system process’ (q = 2.1E−4) and ‘innate immune response’ (q = 7.2E−4). The corresponding analy- sis of gene regulation in the draining lymph node (Ahlberg et al., 2012) revealed an enrichment of up regulated genes in the GO-terms ‘immune response’ (q = 9.9E−6), ‘response to virus’ (q = 4.6E−4), ‘defence response’ (q = 2.3E−3), ‘reg- ulation of cell proliferation’ (q = 1.1E−2) and ‘behaviour’ (q = 4.8E−2). Cell migration patterns induced by Matrix-M were characterized by enrichment for “myeloid cells” and in particular by plasmacytoid dendritic cells (pDC). This profile, together with the prominent up regulation of a considerable number of interferon regulated genes (IRGs), both at the site of injection and in the draining lymph node (Fig. 2) initiated studies on possible mechanisms for induc- tion of IFN production by Matrix-M. Because the genes for two cytosolic RNA-sensing receptors (RIG-1 and MDA5) both were upregulated as well as MyD88 that mediates signalling via TLR7 and TLR9, it was tempting to specu- late that nucleic acid played a part in the induction of IRGs. In patients with the autoimmune disorder systemic lupus erythematosus (SLE), self-DNA released from PMNL in the form of NETs can trigger pDC to produce IFN-␣ (Lande et al., 2011). In that case autoantibodies to DNA are thought to mediate the uptake and delivery of DNA to endosomal TLR9. One of the advantages with the ISCOMs is their ability to deliver antigen to the cytosol allowing presentation of antigen on MHC class I molecules and the induction of a cytotoxic T cell response (Takahashi et al., 1990). Therefore, Matrix-M could in theory mediate trans- port of any associated nucleic acid over cell membrane(s) and thereby initiate an IFN-response. The studies of PMNL exposed to Matrix-M in vitro (Set III) did however not sup- port the hypothesis that Matrix-M induced NET formation, although a prominent influx of neutrophils were recorded after in vivo administration. Fig. 3. Porcine neutrophilic granulocytes incubated for 4 h in plain growth medium (a), 100 nM PMA, 20× magnification (b), 100 nM PMA, 40× magnification (c) or 0.3 ␮g (d), 1 ␮g (e), 3 ␮g (f) Matrix-M per ml.
  • 7. C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61 59 IFN-αα 1 2 3 4 5 6 7 8 9 10 11 12 0.0 0.5 1.0 1.5 2.0 Relativeexpression IFN-ββ 1 2 3 4 5 6 7 8 9 10 11 12 0 10 20 30 40 OPN 1 2 3 4 5 6 7 8 9 10 11 12 0 5 10 15 Matrix Saline Relativeexpression STING 1 2 3 4 5 6 7 8 9 10 11 12 0 1 2 3 4 Matrix Saline Fig. 4. Relative expression of IFN-␣, IFN-␤, OPN and STING in the iliac lymph node of pigs exposed to Matrix-M (nos. 1–6) or saline (nos. 7–12). The expression was normalized to the geometric average of RPL32 and YWHAZ and calibrated to the average expression of all six control pigs. The difference in expression between Matrix-M treated and saline treated groups was statistically significant (p < 0.05) for IFN-␤ and OPN. 3.6. In vivo effects of Matrix-M on IFN-related responses (Set V) The indicated IFN response in pigs early after injection with Matrix-M was followed up with an analysis of the expression of IFN-␣ and -␤, as well as OPN (SPP1) and STING in the draining lymph nodes of pigs that received Matrix-M (n = 6) or just saline (n = 6). OPN and STING have recently been identified as two modulators acting on two different pathways that signal the expression of IFN-␣/␤ in response to viral infections as reviewed by Levy et al. (2011). OPN, which had more than a hundred-fold increase in expression at the site of injection (Ahlberg et al., 2012) is an essential factor for endosomal TLR7/9 dependent induction of IFN-␣/␤ expression via the adaptor molecule MyD88 (Shinohara et al., 2006). STING is essential for cytoplasmic foreign DNA sensor signalling, as well as RIG- like receptor (RLR) signalling, acting through recruitment of TBK1 (Ishikawa et al., 2009). Furthermore, recently a new innate detection mechanism involving STING/TBK1 but independent of TLR and RLR pathways was described, which was triggered by virus-cell membrane fusion only (Holm et al., 2012). Transcripts from IFN-␣, IFN-␤, OPN and STING could be detected in the lymph node of all pigs except for two control pigs, which had no detectable IFN-␤ expression. In this case a Cq value of 40 was assigned. The relative expression of IFN-␣ was always less than two with no difference in expression between Matrix M-treated and control pigs (Fig. 4). However, four pigs (nos. 1, 2, 3 and 6) out of the six Matrix-M-treated pigs showed elevated IFN- ␤ levels (p < 0.05) compared to the control group (Fig. 4). In three of these pigs (nos. 1, 3 and 6) the relative expres- sion of OPN was increased. The expression of OPN in the Matrix-M treated group of pigs was significantly (p < 0.05) higher than in the control group (Fig. 4). The fourth pig with elevated IFN-␤ expression (no. 2) had an OPN expression level comparable to the control pigs but in this pig the rela- tive expression of STING was increased threefold. However, overall STING expression showed only a small increase in two of the Matrix-M treated pigs and as a group, this was not significantly different to the control group. Thus, four pigs treated with Matrix-M showed an increase in IFN-␤ expression and these pigs also expressed an elevated level of OPN and/or STING. For three of these pigs (nos. 1, 2, and 3) the sampled draining lymph node was enlarged at autopsy, further indicating a potent immune activation. 4. Conclusion The adjuvant Matrix-M induces an inflammatory response characterized by a rapid influx of neutrophilic granulocytes and activation of genes regulating early inflammation and other immune response genes in
  • 8. 60 C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61 the pig. Functional annotation analysis and gene set enrichment analysis specified a significant enrichment of “myeloid cell” and pDC. The latter suggestion is well in line with the distinct interferon-regulated gene profile recorded both at the site of injection and in the draining lymph node. In accordance the relative expression of IFN-␤ was increased in the draining lymph node 24 h after injection with Matrix-M. As the IFN-␤ expression was mir- rored by an increased expression of OPN and STING both the endosomal TLR7/TLR9 pathway including MyD88/OPN and the cytosolic STING/TBK1 pathways could be involved in Matrix-M mediated IFN activation, although the former pathway seems like the preferred one. The Matrix com- ponent inducing the interferon response still has to be identified but the present Matrix-M formulation was well tolerated in young piglets, signifying that the magnitude of the inflammation was appropriate to initiate an immune response without causing any adverse side reactions. Acknowledgements We are thankful for the excellent technical assistance of the staff of the Prairie Diagnostic Services and the Prairie Swine Centre of Saskatoon, Canada and by Ulla Schmidt at the University Research Station, Funbo Lövsta, Upp- sala, Sweden. Funding for this project was provided by the EU 6th framework programme Food-CT-2004-513928, Control of Porcine Circovirus Diseases (PCVDs): Towards Improved Food Quality and Safety and by the Swedish Research Council for Environment, Agricultural and Spatial Planning (FORMAS). References Ahlberg, V., Lövgren Bengtsson, K., Wallgren, P., Fossum, C., 2012. Global transcriptional response to ISCOM-Matrix adjuvant at the site of administration and in the draining lymph node early after intramus- cular injection in pigs. Dev. Comp. Immunol. 38, 17–26. Auray, G., Facci, M.R., van Kessel, J., Buchanan, R., Babiuk, L.A., Gerdts, V., 2010. Differential activation and maturation of two porcine DC populations following TLR ligand stimulation. Mol. Immunol. 47, 2103–2111. Azevedo, M.S., Gonzalez, A.M., Yuan, L., Jeong, K.I., Iosef, C., Van Nguyen, T., Lövgren-Bengtsson, K., Morein, B., Saif, L.J., 2010. An oral versus intranasal prime/boost regimen using attenuated human rotavirus or VP2 and VP6 virus-like particles with immunostimulating com- plexes influences protection and antibody-secreting cell responses to rotavirus in a neonatal gnotobiotic pig model. Clin. Vaccine Immunol. 17, 420–428. Bertho, N., Marquet, F., Pascale, F., Kang, C., Bonneau, M., Schwartz- Cornil, I., 2011. Steady state pig dendritic cells migrating in skin draining pseudo-afferent lymph are semi-mature. Vet. Immunol. Immunopathol. 144, 430–436. Bolind Bågenholm, A., 2009. Early immune response to and adjuvant (AbISCO-100® ) tested in porcine peripheral blood mononuclear cells. http://stud.epsilon.slu.se Dalsgaard, K., Hilgers, L., Trouve, G., 1990. Classical and new approaches to adjuvant use in domestic food animals. Adv. Vet. Sci. Comp. Med. 35, 121–159. Dawson, H.D., Royaee, A.R., Nishi, S., Kuhar, D., Schnitzlein, W.M., Zucker- mann Jr., F., Urban, J., Lunney, J.K., 2004. Identification of key immune mediators regulating T helper 1 responses in swine. Vet. Immunol. Immunopathol. 100, 105–111. Duewell, P., Kisser, U., Heckelsmiller, K., Hoves, S., Stoitzner, P., Koernig, S., Morelli, A.B., Clausen, B.E., Dauer, M., Eigler, A., Anz, D., Bourquin, C., Maraskovsky, E., Endres, S., Schnurr, M., 2011. ISCOMATRIX adjuvant combines immune activation with antigen delivery to dendritic cells in vivo leading to effective cross-priming of CD8+ T cells. J. Immunol. 187, 55–63. Fairbairn, L., Kapetanovic, R., Sester, D.P., Hume, D.A., 2011. The mononu- clear phagocyte system of the pig as a model for understanding human innate immunity and disease. J. Leukoc. Biol. 89, 855–871. González, A.M., Nguyen, T.V., Azevedo, M.S., Jeong, K., Agarib, F., Iosef, C., Chang, K., Lövgren-Bengtsson, K., Morein, B., Saif, L.J., 2004. Antibody responses to human rotavirus (HRV) in gnotobiotic pigs following a new prime/boost vaccine strategy using oral attenuated HRV prim- ing and intranasal VP2/6 rotavirus-like particle (VLP) boosting with ISCOM. Clin. Exp. Immunol. 135, 361–372. Hjertner, B., Olofsson, K.M., Lindberg, R., Fuxler, L., Fossum, C., 2013. Expression of reference genes and T helper 17 asso- ciated cytokine genes in the equine intestinal tract. Vet. J., http://dx.doi.org/10.1016/j.tvjl.2013.05.020 [Epub ahead of print]. Holm, C.K., Jensen, S.B., Jakobsen, M.R., Cheshenko, N., Horan, K.A., Moeller, H.B., Gonzalez-Dosal, R., Rasmusse, S.B., Christensen, M.H., Yarovin- sky, T.O., Rixon, F.J., Herold, B.C., Fitzgerald, K.A., Paludan, S.R., 2012. Virus-cell fusion as a trigger of innate immunity dependent on the adaptor STING. Nat. Immunol. 13, 343–737. Hägglund, S., Hu, K.F., Larsen, L.E., Hakhverdyan, M., Valarcher, J.F., Taylor, G., Morein, B., Belák, S., Alenius, S., 2004. Bovine respiratory syncy- tial virus ISCOMs—protection in the presence of maternal antibodies. Vaccine 23, 646–655. Iosef, C., Van Nguyen, T., Jeong, K., Bengtsson, K., Morein, B., Kim, Y., Chang, K.O., Azevedo, M.S., Yuan, L., Nielsen, P., Saif, L.J., 2002. Systemic and intestinal antibody secreting cell responses and protection in gnotobi- otic pigs immunized orally with attenuated Wa human rotavirus and Wa 2/6-rotavirus-like-particles associated with immunostimulating complexes. Vaccine 20, 1741–1753. Ishikawa, H., Ma, Z., Barber, G.N., 2009. STING regulates intracellular DNA- mediated, type I interferon-dependent innate immunity. Nature 461, 788–792. Johansson, M., Lövgren-Bengtsson, K., 1999. Icoms with different quil- laja saponin components differ in their immunomodulating activities. Vaccine 17, 2894–2900. Kapetanovic, R., Fairbairn, L., Beraldi, D., Sester, D.P., Archibald, A.L., Tug- gle, C.K., Hume, D.A., 2012. Pig bone marrow-derived macrophages resemble human macrophages in their response to bacterial lipopolysaccharide. J. Immunol. 188, 3382–3394. Kensil, C.R., Patel, U., Lennick, M., Marciani, D., 1991. Separation and char- acterization of saponins with adjuvant activity from Quillaja saponaria Molina cortex. J. Immunol. 146, 431–437. Labbé, K., Saleh, M., 2011. Pyroptosis A Caspase-1-dependent pro- grammed cells death and a barrier to infection. In: Couillin, I., Pétrilli, V., Martinon, F. (Eds.), The Inflammasome, Progress in Inflammation Research. Springer, Basel AG, pp. 17–36. Lande, R., Ganguly, D., Facchinetti, V., Frasca, L., Conrad, C., Gregorio, J., Meller, S., Chamilos, G., Sebasigari, R., Riccieri, V., Bassett, R., Amuro, H., Fukuhara, S., Ito, T., Liu, Y.J., Gilliet, M., 2011. Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus. Sci. Transl. Med. 3, 73ra19. Levy, D.E., Marié, I.J., Durbin, J.E., 2011. Induction and function of type I and III interferon in response to viral infection. Curr. Opin. Virol. 1, 476–486. Lin, W., Qiu, Z., Liu, Q., Cui, S., 2013. Interferon induction and suppres- sion in swine testicle cells by porcine parvovirus and its proteins. Vet. Microbiol. 163, 157–161. Livak, K.J., Schmittgen, T.D., 2001. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods 25, 402–408. Lövgren Bengtsson, K., Morein, B., 2000. The ISCOMTM technology. In: O’Hagan, D.T. (Ed.), Methods in Molecular Medicine, Vaccine Adju- vant: Preparation Methods and Research Protocols. Humana Press, Inc., Totowa, NJ, pp. 239–258. Lövgren, K., Morein, B., 1988. The requirement of lipids for the formation of immunostimulating complexes (iscoms). Biotechnol. Appl. Biochem. 10, 161–172. Lövgren Bengtsson, K., Morein, B., Osterhaus, A.D., 2011. ISCOM technology-based Matrix MTM adjuvant: success in future vaccines relies on formulation. Expert Rev. Vaccines 10, 401–403. Lövgren Bengtsson, K., Sjölander, A., 1996. Adjuvant activity of iscoms; effect of ratio and co-incorporation of antigen and adjuvant. Vaccine 14, 753–760. Marquet, F., Bonneau, M., Pascale, F., Urien, C., Kang, C., Schwartz-Cornil, I., Bertho, N., 2011. Characterization of dendritic cells subpopula- tions in skin and afferent lymph in the swine model. PLoS ONE 6, e16320. Morein, B., Sundquist, B., Höglund, S., Dalsgaard, K., Osterhaus, A., 1984. Iscom a novel structure for antigenic presentation of membrane pro- teins from enveloped viruses. Nature 308, 457–460.
  • 9. C. Fossum et al. / Veterinary Immunology and Immunopathology 158 (2014) 53–61 61 Morein, B., 1987. Potentiation of the immune response by immunization with antigens in defined multimeric physical forms. Vet. Immunol. Immunopathol. 17, 153–159. Morein, B., 1988. The iscom antigen-presenting system. Nature 332, 287–288. Morein, B., Blomqvist, G., Hu, K., 2007. Immune responsiveness in the neonatal period. J. Comp. Pathol. 137, S27–S31. Morelli, A.B., Becher, D., Koernig, S., Silva, A., Drane, D., Maraskovsky, E., 2012. ISCOMATRIX: a novel adjuvant for use in prophylactic and therapeutic vaccines against infectious diseases. J. Med. Microbiol. 61, 935–943. Nguyen, T.V., Iosef, C., Jeong, K., Kim, Y., Chang, K.O., Lövgren-Bengtsson, K., Morein, B., Azevedo, M.S., Lewis, P., Nielsen, P., Yuan, L., 2003. Vaccine 21, 4059–4070. Nguyen, T.V., Yuan, L., Azevedo, M.S., Jeong, K.I., Gonzalez, A.M., Iosef, C., Lövgren-Bengtsson, K., Morein, B., Lewis, P., Saif, L.J., 2006a. High titers of circulating maternal antibodies suppress effector and mem- ory B-cell responses induced by an attenuated rotavirus priming and rotavirus-like particle-immunostimulating complex boosting vaccine regimen. Clin. Vaccine Immunol. 13, 475–485. Nguyen, T.V., Yuan, L., Azevedo, M.S., Jeong, K.I., Gonzalez, A.M., Iosef, C., Lövgren-Bengtsson, K., Morein, B., Lewis, P., Saif, L.J., 2006b. Low titer maternal antibodies can both enhance and suppress B cell responses to a combined live attenuated human rotavirus and VLP-ISCOM vaccine. Vaccine 24, 2302–2316. Puentes, E., Cancio, E., Eiras, A., Nores, M.V., Aguilera, A., Regueiro, B.J., Seoane, R., 1993. Efficacy of various non-oily adjuvants in immuniza- tion against the Aujeszky’s disease (pseudorabies) virus. Zentralbl. Veterinarmed. B 40, 353–365. Reimer, J.M., Karlsson, K.H., Lövgren-Bengtsson, K., Magnusson, S.E., Fuentes, A., Stertman, L., 2012. Matrix-MTM adjuvant induces local recruitment, activation and maturation of central immune cells in absence of antigen. PLoS ONE 7, e41451. Rönnberg, B., Fekadu, M., Morein, B., 1995. Adjuvant activity of non-toxic Quillaja saponaria Molina components for use in ISCOM matrix. Vac- cine 13, 1375–1382. Straw, B.E., DAllaire, S., Mengeling, W.L., Taylor, D.J., 1999. Diseases of Swine, 8th ed. Iowa State University Press, Ames, IA. Shinohara, M.L., Lu, L., Bu, J., Werneck, M.B., Kobayashi, K.S., Glimcher, L.H., Cantor, H., 2006. Osteopontin expression is essential for interferon- alpha production by plasmacytoid dendritic cells. Nat. Immunol. 7, 498–506. Takahashi, H., Takeshita, T., Morein, B., Putney, S., Germain, R.N., Berzofsky, J.A., 1990. Induction of CD8+ cytotoxic T cells by immunization with purified HIV-1 envelope protein in ISCOMs. Nature 344, 873–875. Tsuda, T., Sugimura, T., Murakami, Y., 1991. Evaluation of glycoprotein gII ISCOMs subunit vaccine for pseudorabies in pig. Vaccine 9, 648– 652. Tulman, E.R., Garmendia, A.E., 1994. Delivery of pseudorabies virus envelope antigens enclosed in immunostimulating complexes (ISCOMs): elicitation of neutralizing antibody and lymphoprolif- erative responses in swine and protection in mice. Vaccine 12, 1349–1354. Uddin, M.J., Cinar, M.U., Tesfaye, D., Looft, C., Tholen, E., Schellander, K., 2011. Age-related changes in relative expression stability of com- monly used housekeeping genes in selected porcine tissues. BMC Res. Notes 4, 1–13. Vandesompele, J., De Preter, K., Pattyn, F., Poppe, B., Van Roy, N., De Paepe, A., Speleman, F., 2002. Accurate normalization of real-time quantita- tive RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol., 3, research0034.1–0034.11. Wallgren, P., Segall, T., Pedersen Mörner, A., Gunnarsson, A., 1999. Exper- imental infections with Actinobacillus pleuropneumoniae in pigs—I. Comparison of five different parenteral antibiotic treatments. Zen- tralbl. Veterinarmed. B 46, 249–260. White, F.J., Ross, J.W., Joyce, M.M., Geisert, R.D., Burghardt, R.C., Johnson, G.A., 2005. Steroid regulation of cell specific secreted phosphopro- tein 1 (Osteopontin) expression in the pregnant porcine uterus. Biol. Reprod. 73, 1294–1301. Wikström, F.H., Fossum, C., Fuxler, L., Kruse, R., Lövgren, T., 2011. Cytokine induction by immunostimulatory DNA in porcine PBMC is impaired by a hairpin forming sequence motif from the genome of Porcine Cir- covirus type 2 (PCV2). Vet. Immunol. Immunopathol. 139, 156–166. Xie, L., Liu, M., Fang, L., Su, X., Cai, K., Wang, D., Xiao, S., 2010. Molecu- lar cloning and functional characterization of porcine stimulator of interferon genes (STING). Dev. Comp. Immunol. 34, 847–854.