You are on page 1of 12

Molecular Immunology 55 (2013) 200–211

Contents lists available at SciVerse ScienceDirect

Molecular Immunology
journal homepage: www.elsevier.com/locate/molimm

Review: Influenza virus in pigs


Elisa Crisci a,1 , Tufária Mussá a,1 , Lorenzo Fraile b , Maria Montoya a,c,∗
a
Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Spain
b
Universitat de Lleida, Lleida, Spain
c
Institut de Recerca i Tecnologia Agroalimentàries (IRTA), Barcelona, Spain

a r t i c l e i n f o a b s t r a c t

Article history: Influenza virus disease still remains one of the major threats to human health, involving a wide range
Received 8 December 2012 of animal species and pigs play an important role in influenza ecology. Pigs were labeled as “mixing
Received in revised form 23 February 2013 vessels” since they are susceptible to infection with avian, human and swine influenza viruses and genetic
Accepted 25 February 2013
reassortment between these viruses can occur. After the H1N1 influenza pandemic of 2009 with a swine
Available online 21 March 2013
origin virus, the most recent research in “influenzology” is directed at improving knowledge of porcine
influenza virus infection. This tendency is probably due to the fact that domestic pigs are closely related to
Keywords:
humans and represent an excellent animal model to study various microbial infectious diseases. In spite
Pig
Influenza virus
of the role of the pig in influenza virus ecology, swine immune responses against influenza viruses are
Immune responses not fully understood. Considering these premises, the aim of this review is to focus on the in vitro studies
performed with porcine cells and influenza virus and on the immune responses of pigs against human,
avian and swine influenza viruses in vivo. The increased acceptance of pigs as suitable and valuable models
in the scientific community may stimulate the development of new tools to assess porcine immune
responses, paving the way for their consideration as the future “gold standard” large-animal model in
immunology.
© 2013 Elsevier Ltd. All rights reserved.

1. Introduction The viral envelope, derived from the host plasma membrane, con-
sists of a lipid bilayer containing transmembrane proteins (HA and
1.1. Influenza virus and pigs NA) and matrix proteins (M1 and M2). HA, is the major enve-
lope protein forming projection like spikes on the virion surface
Since the “obscuri coeli influentia”, defined by the ancient as the (Webster et al., 1992), it provides the receptor-binding site and
“astral influence” during influenza virus (IV) illness, to today, much elicits neutralizing antibodies. HA binds to host cell receptors that
progress has been made with regard to the knowledge of this dis- contain terminal ␣-2,6-linked or ␣-2,3-linked sialic acid (␣-2,6-SA
ease. Nevertheless, IV disease still remains one of the major threats or ␣-2,3-SA) moieties. Cleavage of HA is essential for fusion and
to human health, and involves a wide range of animal species. virus infectivity. NA removes the cell surface receptor (sialic acid);
Influenza viruses (IVs) are enveloped, single stranded RNA it is critical for release of virus particles from the cell surface and
viruses of the family Orthomyxoviridae. This family comprises dif- spread of virus. M2, an ion channel, is crucial during uncoating
ferent genera, in particular influenza A, B and C viruses. Influenza A for dissociating the virus ribonucleocapsids (vRNP) from M1 in the
viruses are further classified into subtypes based on the antigenic- early phase of the infectious cycle. The viral core consists of helical
ity of their hemagglutinin (HA) and neuraminidase (NA) molecules. vRNP containing vRNA (negative stranded) and nucleoprotein (NP)
Currently, 17 HA (H1–H17) (H17 recently described in Tong et al. along with minor amounts of the nuclear export protein (NEP) (also
(2012)) and 9 NA subtypes (N1–N9) are known. called non-structural protein NS2) and three polymerase proteins
Influenza A and B viruses possess segmented genomes of (PB1, PB2, and PA) which form the viral RNA polymerase complex
eight single-stranded negative-sense RNA molecules that typically (Arranz et al., 2012; Nayak et al., 2004, 2009; Palese and Shaw,
encode 11 or 12 viral proteins (Medina and Garcia-Sastre, 2011). 2007).
IVs are thought to infect different animal species and pigs
(Sus scrofa) are one of the natural hosts of these viruses. Swine
influenza is a highly important respiratory swine disease and the
∗ Corresponding author at: Centre de Recerca en Sanitat Animal (CReSA),
main causative viruses are H1N1, H1N2 and H3N2 subtypes (Brown,
UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, 08193 Bellaterra (Cer-
danyola del Vallès), Spain. Tel.: +34 935814562; fax: +34 935814490.
2012), which are antigenically related to human IVs. Pigs are sus-
E-mail address: maria.montoya@cresa.uab.es (M. Montoya). ceptible to infection with avian and human IVs, and are supposed
1
These authors contributed equally to this paper. to play an important role in human influenza ecology. In fact,

0161-5890/$ – see front matter © 2013 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.molimm.2013.02.008
E. Crisci et al. / Molecular Immunology 55 (2013) 200–211 201

genetic reassortment between human and/or avian and/or swine interaction between IV and the respiratory epithelium. In vitro stud-
IVs can occur. The potential to generate novel IVs has resulted in ies with progenitors or differentiated respiratory epithelial cells are
swine being labeled “mixing vessels”. There are three facts sup- possible, as are air–liquid interface culture or explant culture or ex
porting the mixing vessel hypothesis: (1) swine are susceptible to vivo organ culture. Nowadays, the latest tendency is to use differ-
avian and human viruses; (2) reassortment of swine/avian/human ent porcine cell systems for studying IV infection, probably because
viruses occurs in pigs and (3) pigs can transmit reassortant IVs to swine-origin cells are considered a more suitable tool to investigate
humans (Ma et al., 2009). However, infection of pigs could result in the host cell contributions to pathogenesis in pigs.
viruses gaining the adaptations necessary to either maintain infec- In the case of differentiated airway epithelial cells from pigs,
tion within pigs, thereby providing a further reservoir for human infection studies with IVs have been reported with explant cul-
infection, or provide the mutations required to allow transmission tures from different parts of the respiratory tract (Van Poucke et al.,
in humans, leading to establishment of this virus in the human 2010). These explants are viable for four days and different IVs were
population (Londt et al., 2012). used in this system (Table 1). The infectivity of aIVs was shown
Other animal models, such as ferrets and guinea pigs, have been to be lower in the upper respiratory tract, while the pattern of
successfully used for influenza virus studies (Belser et al., 2011; huIVs more closely resembled that of swIVs. Compared to swIVs
Sun et al., 2010), but after the H1N1 influenza pandemic of 2009 (A and huIVs, replication of the aIVs was limited in all cultures but
(H1N1)pdm/2009) with a swine origin virus (Garten et al., 2009), most strikingly in nasal and tracheal explants (Van Poucke et al.,
much effort has been made to improve the knowledge of IV infec- 2010).
tion in pigs. Various studies have demonstrated the pathogenesis Nunes et al. also described an ex vivo organ culture (EVOC) sys-
and transmission of A (H1N1)pdm/2009 IV in swine (Brookes et al., tem using porcine trachea (Nunes et al., 2010). In this study the
2010; Busquets et al., 2010; Lange et al., 2009) and further studies EVOC system was maintained for 7 days and the ciliary move-
have been performed on pathogenesis and epidemiology of other ment exhibited a gradual decrease after 4 days. They challenged
IVs in pigs (De Vleeschauwer and Van Reeth, 2010; Khatri et al., the EVOC with swIV H1N1 (Table 1) and the system showed his-
2010; Kitikoon et al., 2012; Zhu et al., 2011). In this way, the recent tological changes similar to in vivo IV infection and supported
studies in “influenzology” have been directed at swine IV infec- productive viral replication over multiple cycles of infection with
tion. This tendency is probably due to the fact that domestic pigs dose-dependent kinetics (Nunes et al., 2010). Recently, Londt et al.
are closely related to humans anatomically, genetically and physio- (2012) used tracheal, bronchi and lung explants and tested dif-
logically, and represent an excellent animal model to study various ferent IV subtypes (Table 1). Pandemic viruses and HPAI H5N1
microbial infectious diseases. Indeed, experiments in pigs are much subtype were able to infect pig ex vivo organ culture, and the
more likely to be predictive of therapeutic treatments in humans detection of virus was higher in the tissues of the lower respira-
than experiments in rodents. A recent review (Meurens et al., 2012) tory tract (i.e. lungs). Endemic swIV was able to replicate to higher
highlighted the numerous advantages of the pig model for infec- levels in trachea when compared to avian and pandemic viruses.
tious disease research and vaccine development. Based on the same A(H1N1)pdm/2009 and aIVs appeared to replicate preferentially
subtypes that infect pigs and human and the similarities between in the bronchi and lung tissue explants. This may be due to the
clinical diseases, pathogenesis and tissue tropism of IV infections increase in ␣2–3 receptor density or the number of susceptible
in pigs and humans, pig are ideal experimental animals to study cells in the lower respiratory tract. Thus, also this report supports
different aspects of influenza infections (Meurens et al., 2012). the use of ex vivo explants for screening viruses for their ability to
In spite of the role of the pig in IV ecology, swine immune infect pigs prior to in vivo studies (Londt et al., 2012).
responses against IVs are not fully understood. Taking into account An interesting new culture system for porcine differentiated
these premises, the aim of this review is to focus on the immune respiratory epithelial cells, precision-cut lung slices (PCLS) was also
responses of pigs against swine (swIV), human (huIV) and avian set up. Punyadarsaniya et al. (2011) used this culture system to
(aIV) influenza viruses. compare the infection of respiratory epithelial cells by swine IV and
two LPAI aIVs (Table 1). Interestingly, this system is much more sus-
2. In vitro interaction between influenza viruses and the ceptible to aIV H9N2 than to the H7N7 subtype and swIV growth
porcine immune system was faster than aIVs. The ciliary activity was maintained for up to
48 h post infection (h pi) with all the three viruses and ciliostasis
IVs are thought to infect mainly the respiratory-tract epithelial appeared to be dependent on the amount of the virus generated
cells of animals and humans in vivo. IVs are able to infect a variety of during the infection. The results have shown that all the viruses
cells in vitro, including the Madin-Darby canine kidney (MDCK) cell were able to infect ciliated epithelial cells and swIV also infected
line (Tobita et al., 1975), the African Green Monkey Kidney (VERO) mucus-producing cells (Punyadarsaniya et al., 2011).
cell line (Genzel et al., 2010), human monocytes (Bussfeld et al., Olsen’s group also described two different types of porcine
1998) and human monocyte-derived macrophages (Hofmann et al., airway epithelial culture for studying IV infection. Recently, dif-
1997). ferentiated swine airway epithelial (SAE) cell cultures were grown
Several in vitro studies have investigated the interaction at an air–liquid interface (ALI) varying amounts of retinoic acid and
between IV and different systems of mammalian cells. These epidermal growth factor and infected with reverse genetics(rg)-
reports mainly focused attention on the binding ability of the virus created viruses from triple reassortant rH3N2 and human-lineage
and on the pathogenesis, but few data have been published on the swine IV (Table 1). The expression of ␣2–3 and ␣2–6-linked sialic
immune responses elicited after the IV-cell interaction. Here, we acid on SAE cultures is similar to that in the swine lower trachea. IVs
review the in vitro studies performed with different IVs and porcine replicated successfully in these multi-layered cultures, even in the
cell systems. Other studies have been performed using the porcine absence of exogenous trypsin (Bateman et al., 2013). Infection and
system considering the co-infection of IV and other pathogens. replication characteristic of the IVs used were similar to results pre-
These data are out of the scope of this review. viously seen in vivo and in monolayer of primary swine respiratory
epithelial cells (SRECs) in vitro (Busch et al., 2008).
2.1. Epithelial cells Another fascinating recent report has shown the possibility of
isolating and using stem/progenitor epithelial cells from the lungs
Epithelial cells are the first host defence barrier and the primary of pigs for in vitro studies (Khatri et al., 2012). These Oct4+ SSEA-
target for IVs; for this reason different studies have investigated the 1+ cells have extensive self-renewal potential and the capacity to
202 E. Crisci et al. / Molecular Immunology 55 (2013) 200–211

Table 1
In vitro studies on influenza viruses and porcine cell systems.

Cells Culture cell system/tissue Viruses References

Epithelial cells Explant culture/ swIV Van Poucke et al. (2010)


Nasal respiratory mucosa Sw/Belgium/1/98 (H1N1)
Trachea Sw/Flanders/1/98(H3N2)
Bronchi Sw/Gent/7625/99 (H1N2)
Lung huIV
A/New Caledonia/20/99 (H1N1)
A/Panama/2007/99 (H3N2)
LP aIV
Duck/Italy/1447/05 (H1N1)
Mallard/Alberta/279/98 (H3N8)
Duck/Belgium/06936/05 (H4N6)
Chicken/Belgium/150/99 (H5N2)
Chicken/Italy/1067/V99 (H7N1)
Mallard/Italy/3401/2005 (H5N1)
Explant culture/ HP aIV Londt et al. (2012)
Trachea A/chicken/Netherlands/3219-3/03 (H7N7)
Bronchi A/turkey/Turkey/1/2005 (H5N1)
Lung LPAI aIV
A/chicken/England/4054/06 (H7N3)
huIV
A/England/195/09 (H1N1)
A/California/7/09 (H1N1)
swIV
A/swine/England/195852/92 (H1N1)
Ex vivo organ culture (EVOC) system/Trachea swIV Nunes et al. (2010)
A/swine/England/453/06 (H1N1)
Differentiated respiratory epithelial cells, swIV Punyadarsaniya et al. (2011)
precision-cut lung slices (PCLS)/ A/sw/Bissendorf/IDT1864/2003 (H3N2)
Lung LP aIV
A/duck/Potsdam/15/80 (H7N7)
A/chicken/Saudi Arabia/CP7/98 (H9N2)
Progenitor epithelial cells swIV Sw/OH/07, H1N1 Khatri and Saif (2011)
Type II pneumocytes/ huIV Hu/OH/06, H1N1
Bone Marrow aIV Ch/NY/95, H7N2
Stem/progenitor epithelial cells swIV Sw/OH/07 H1N1 Khatri et al. (2012)
Type I and II pneumocytes/ huIV Hu/OH/06, H1N1
Lung aIV Ch/NY/95, H7N2
Differentiated swine airway epithelial (SAE) swIV H3N2 Bateman et al. (2013)
cells, multi-layered culture/Distal trachea rg (A/Swine/Minnesota/593/99)
rg (A/swine/Ontario/00130/97)
Monolayer of primary swine respiratory swIV H3N2 Busch et al. (2008)
epithelial cells (SRECs)/ rg (A/Swine/Minnesota/593/99)
Distal tracheal-proximal bronchi rg (A/swine/Ontario/00130/97)
A/Swine/Iowa/533/99
A/Swine/Iowa/569/99
A/Swine/Nebraska/209/98
A/Swine/CO/1/77
Intestinal epithelial cell line (SD-PJEC) 25 influenza virus isolates of human, swine or Sun et al. (2012)
subclone of IPEC-J2/ avian origin
Jejunal epithelia of a neonatal unsuckled piglet
Newborn swine kidney (NSK, BS CL 177)/ LP aIV Lombardo et al. (2012)
Kidney A/Mallard/Italy/3401/05 (H5N1)
A/Turkey/Italy/2962/V03 (H7N3)
A/Turkey/Wisconsin/66 (H9N2)
HP aIV
A/Chicken/Italy/8/A98 (H5N2)
swIV
A/Swine/Italy/25674/09 (H1N1)
A/Swine/Italy/290271/09 (H1N1)
A/Swine/BS2156 (H1N2)
A/Swine/Oedenrode/1/96 (H3N2)

Stromal cells Mesenchymal stromal cells (MSCs) from bone A/Sw/OH/24366 (H1N1) Khatri and Saif (2011)
marrow A/Hu/OH/K1130/06 (H1N1)

Macrophages M/BAL A/Hu/Sydney/5/97 (H3N2) Seo et al. (2004)


A/New Caledonia/1/99 (H1N1)
B/Shichuan/99
M/BAL swIV H1N2 (not specified) Kim et al. (2009)
3D/4 spontaneously transformed line/ A/Hu/Nanjing/108/2009 (H1N1) Gao et al. (2012)
Myelomonocytic cell line alveolar M
E. Crisci et al. / Molecular Immunology 55 (2013) 200–211 203

Table 1 (Continued )

Cells Culture cell system/tissue Viruses References

Dendritic cells cDCs (BMDCs)/ swIV A/Swine/Spain/SF32071/2007 (H3N2) Mussá et al. (2011, accepted for
Bone Marrow huIV A/Catalonia/63/2009 (H1N1) publication)
LP aIV A/Anas
plathyrhynchos/Spain/1877/2009 (H7N2)
HP aIV A/Chicken/Italy/5093/1999 (H7N1)
cDCs/ aIV A/chicken/Yamaguchi/7/04 (H5N1) Ocana-Macchi et al. (2012)
Bone Marrow swIV A/swine/Belzig/2/01 (H1N1) Bel et al. (2011)
pDCs/ HP aIV H5N1
Blood PBMCs A/turkey/Turkey/05
A/Cygnus/Italy/742/06
A/mallard/Italy/835/06
HP aIV H7N1
A/turkey/Italy/4580/98
A/ostrich/Italy/2332/00
LP aIV H7N1
A/turkey/Italy/3675/99
huIV
A/New Caledonia/20/99 (H1N1)
A/Wisconsin/67/05 (H3N2)
A/Hong Kong/8/68 (H3N2)
swIV
A/swine/Iowa/1976/31 (H1N1)
A/swine/Belgium/1/98 (H1N1)
A/swine/Flanders/1/98 (H3N2)

undergo differentiation to alveolar type I- and type II-like pneu- to prevent the infection of respiratory epithelial cells. Therefore,
mocytes. These cells express sialic acid receptors and supported innate immune cells like macrophages, neutrophils, dendritic
the active replication of IVs (Table 1), which was accompanied by cells and natural killer cells are recruited with the objective of
cell lysis. The data provided new insights into the pathogenesis of controlling virus replication and dissemination and to orchestrate
IV, suggesting that the infection and lysis of lung progenitor cells by the immune response. These cells secrete different types of chem-
IV is responsible for the loss of lung functions, delayed lung repair ical mediators such as cytokines that will activate the T cells and
and delayed epithelial regeneration after IV infection (Khatri et al., induce their differentiation into memory cells or elicit an adaptive
2012). The authors also studied the permissiveness of mesenchi- response by humoral (virus specific antibodies) and cellular immu-
mal stromal cells and their cytokine production after swine and nity (T cells) (Fig. 1). In the following section we will consider each
human H1N1 infection (Table 1). The findings demonstrated that component of the porcine innate immunity separately.
mesenchimal stromal cells are susceptible to IVs and the infection
resulted in cell lysis and in the production of TNF-␣ and IL-6 (Khatri 2.2.1. Collectins
and Saif, 2011). Surfactant protein D (SP-D) belongs to the family of C-type
Lombardo et al. (2012) compared different mammalian and lectins (collectins) which are important effectors of innate immune
avian cell lines in their ability to support different IV subtypes system. Hillaire et al. (2011) evaluated the potential of recombi-
(Table 1). The most interesting finding was that newborn swine kid- nant porcine SP-D as an antiviral agent against influenza A viruses
ney (NSK, BS CL 177) permitted viral isolation of aIVs, either LPAI in vitro. Thirty IVs (Table 1) from bird, pig and human origins were
or HPAI strains, from target tissues of infected chicken. In addition, tested for their sensitivity to recombinant SP-D. They showed that
NSK cells appeared more susceptible than MDCK cells for primary porcine SP-D bind HA, inhibited NA activity and was more potent
isolation from clinical specimens. These cells also allowed serial than recombinant human SP-D. Porcine SP-D was active against a
virus cultivation of both aIVs and swIVs, yielding high quantities of broad range of influenza A virus strains and neutralized a variety of
IV mainly for HPAI (Lombardo et al., 2012). H1N1 and H3N2 viruses, including pandemic H1N1 viruses. Finally,
Finally, the porcine intestinal epithelial cell line (SD-PJEC) was SP-D prevented attachment of human seasonal H1N1 and H3N2 to
characterized for IV production (Sun et al., 2012). The SD-PJEC cell receptor on epithelial cells of the upper respiratory tract (Hillaire
line is a subclone of the IPEC-J2, which is a non-transformed, non- et al., 2011).
tumorigenic small intestinal epithelial cell line originally derived
from jejunal epithelia of a neonatal, unsuckled piglet (Rhoads et al., 2.2.2. Macrophages
1994). Sun et al. (2012) demonstrated that SD-PJEC preferentially Upon infection of the alveoli, alveolar macrophages (M)
expresses ␣2–6 receptors and 25 IV isolates were tested (Table 1). become activated and phagocytose (apoptotic) IV infected cells
This cell line was permissive to infection with human, swine and limiting viral spread in pigs (Kim et al., 2008). Seo et al. (2004) iso-
some avian IVs, but poorly supported the growth of human-origin lated alveolar M in bronco-alveolar lavage (BAL) of post-mortem
influenza B viruses. The authors suggested this system as an appro- pigs. Thus, M that constitutively reside in the respiratory tract of
priate cellular model for swine-origin IV, since then to date, findings pigs, were infected with huIVs (Table 1) and the infectivity titers
concerning the pathogenic mechanism of IV within the digestive of the viruses were similar. The inflammatory cytokines IL-1␤, IL-6
system have been limited due to a lack of a good in vitro cellular and TNF-␣ were induced by alveolar M and of these, TNF-␣ was
system (Sun et al., 2012). the most highly induced. H3N2 huIV induced significantly greater
expression of TNF-␣ than did H1N1 or influenza B virus. TNF-␣
2.2. Innate immune system protein increased as the duration of infection increased and alve-
olar M did not undergo apoptosis when infected with IVs. Taken
The innate immune system forms the first line of defence together, these findings suggest that alveolar M are involved in
against IV infection (Fig. 1). It consists of a plethora of components, the pathogenesis of IV infection and in the associated immune
such as mucus, collectins and acute phase proteins, which aim response (Seo et al., 2004).
204 E. Crisci et al. / Molecular Immunology 55 (2013) 200–211

Fig. 1. Immunity during influenza virus infection. The innate immune system forms the first line of defence against influenza infection. It consists of plethora of components,
such as mucus, collectins and acute phase proteins, which aim to prevent infection of respiratory epithelial cells. Therefore, rapid innate immune cells like macrophages,
dendritic cells, natural killer and  T cells are recruited with the objective of controlling and blocking virus replication and dissemination and to orchestrate the immune
response. These cells secrete different types of chemical mediators such as cytokines that will activate the T cells and induces their differentiation or elicit an adaptive
response with the production of specific IV-antibodies responses. This figure is mainly based on human and murine studies.

Similar results in M culture supernatant from M infected present the IV derived antigen through the MHC to T cells and
with swIV H1N2 were shown by Kim et al. (2009). Significant activate them (GeurtsvanKessel and Lambrecht, 2008). Among the
differences in TNF-␣ concentration between swIV-infected and various other activities of DCs in IV infection, they can also exert
uninfected alveolar M were detected at different h pi, with a peak cytolytic activity and contribute to the formation of bronchus asso-
at 36 h pi. These results suggested that TNF-␣ may be an impor- ciated lymphoid tissue (BALT) (GeurtsvanKessel and Lambrecht,
tant mediator in the pathophysiology of swIV infection (Kim et al., 2008; Kreijtz et al., 2011). Although DCs have an important role
2009). during IV infection, few studies have investigated the interaction
Another in vitro study used 3D/4 cells, a spontaneoulsy- between IV and porcine DCs.
transformed line of swine M (ATCC), infected with a pandemic Our group described for the first time the interaction between
H1N1 virus (Table 1) (Gao et al., 2012). This report demonstrated porcine bone marrow-derived DCs (poBMDCs) (cDCs) and swIV
that A (H1N1)pdm/2009 retains the ability to infect and replicate H3N2 (Table 1) in vitro. Infection of poBMDCs resulted in struc-
in swine M, inducing a typical cytopathic effect (16 h pi) and ture resembling IV inside vesicles and also free in the cytoplasm
destroying the cell monolayer (32 h pi). This study also examined of the cells. Viral progeny was undetectable in the supernatant but
the pattern of cytokine responses in pH1N1-infected swine M by limited replication was detected in the first 8 h pi. However, viral
real time RT-PCR. IL-6 and IL-8 levels were up-regulated at 16 h particles from infected-poBMDCs were able to induce cytopathic
and level of IL-8 continued to rise up at 36 h pi. Robust induction of effect in susceptible cells only when cell-to-cell interaction was
antiviral IFN-␤ and TNF family members, which may be attributable favored (Mussá et al., 2011). Additionally Mussá et al. (accepted for
to cell death, was observed. FasL and TNF-␣ remained undetectable, publication), observed that similarly to the swIV H3N2, porcine DCs
while the TNF-related apoptosis-inducing ligand (TRAIL) seemed also supported a limited replication of other IVs (Table 1) during
to be the most abundant one before infection. FasL and TNF-␣ the first 8 h pi, without release of infectious progeny. These viruses
were induced most robustly, but TRAIL was only mildly induced also similarly modulated the expression of NF-␬B, TGF-␤ and IL-
in response to infection. The level of IL-1␤ remained unchanged 10 genes. However, they induced different kinetics and levels of
throughout the infection (different from Barbe et al., 2011; Van inflammatory cytokines. Infection of poBMDCs with swIV induced
Reeth, 2000), indicating that IL-6 and IL-8, as well as TNF-␣ were a peak of IFN-␣ secretion at 24 h pi, whereas with the others the
the main pro-inflammatory cytokines up-regulated. The authors production of IFN-␣ was not detected. SwIV and HPAI induced more
also observed the induction of RIG-1 and MDA-5, which appeared TNF-␣ compared to huIV and LPAI. SwIV, LPAI and HPAI induced an
to be suppressed completely by inhibitors of ERK1/2 or JNK1/2. This increase of IL-12 from 16 to 24 h pi and all viruses used induced
indicated that the induction of RIG-1 or MDA-5 depends on the IL-18 secretion in a time-dependent manner.
activation of ERK1/2 and JNK1/2 in pig M (Gao et al., 2012). Summerfield’s group also used GM-CSF derived DCs and other
avian and porcine IVs (Table 1). They also generated recombinant
2.2.3. Dendritic cells reassortants by reverse genetics to elucidate the role of the sin-
Conventional dendritic cells (cDCs), situated underneath the air- gle gene segments in the activation of cDCs. The highest IFN type I
way epithelium barrier and above the basal membrane, monitor responses were achieved by the porcine virus reassortant contain-
the airway lumen via their dendrites that are extended through the ing the avian polymerase gene PB2. This finding was not due to
tight junctions between the airway epithelial cells (Kreijtz et al., the differential tropism since all viruses infected DCs equally (and
2011). cDCs can detect and opsonise (neutralize) virions and apop- also porcine PK-15 epithelial cells) and infectivity was independent
totic bodies from infected cells and can also be infected themselves. of the HA expressed by the virus. All viruses induced MHC-II, but
Upon entry of the virion into the cells, the cDCs migrate via the porcine H1N1 expressing the avian viral PB2 induced more promi-
afferent lymphatic system to the draining lymph node. Here they nent nuclear NF-kB translocation compared to its parent virus.
E. Crisci et al. / Molecular Immunology 55 (2013) 200–211 205

Therefore, for porcine DCs, PB2 was defined as an important viral Lung viral load Cytokines APPs
element controlling IFN type I. While all viruses had a comparable IFN- T cells Lymphocytes IV-specific Abs
ability to infect DCs, to initiate replication and to activate the cells

Magnitude of infection/responses
in terms of MHC-II induction, only those expressing PB2 derived
from H5N1 were unable to prevent IFN type I induction; however
no viral progeny was detected (Ocana-Macchi et al., 2012).
Plasmacytoid DCs (pDCs), a subset of the DC family, are highly
specialized in sensing viruses and readily secreting IFN-␣ after
exposure to swIV (Calzada-Nova et al., 2009). Therefore, consid-
ering the distinct features of pDCs and the crucial role of IFN in
fighting virus infections, Bel et al. (2011) analyzed the interac-
tions of different influenza A viruses (Table 1) isolated from avian,
human and swine with pDCs obtained from pigs. Their results
demonstrated that porcine pDCs can produce high levels of IFN-␣ in
response to all tested strains, with subtype-specific differences in a
0 1 2 3 4 5 6 7 8 9 10 11 12 13 14
virus-dose dependent manner. High levels of IFN-␣ were detected
upon live, chemically inactivated or UV-inactivated virus stimula- Days after infection
tion. In contrast, heat-inactivated virus failed to induce a response.
Fig. 2. Swine influenza virus infection in pigs. General kinetics of lung viral load,
The observation that chemical and UV-inactivation did not abol- acute phase proteins (APPs), cytokines (e.g. TNF-␣, IL-6, and IL-8) and different
ish IFN-␣ release indicated that non-infectious particles are also immune system components (lymphocyte, IFN-␥ producing cells and influenza
stimulatory for pDCs. Additionally, these treatments did not abol- virus (IV)-specific antibodies) during infection with different swine influenza viruses
ish hemagglutination, suggesting that the integrity of HA and its (H1N1 and H3N2) in pigs. The magnitude of responses is related to the days after
infection (0–14).
binding function are necessary to induce IFN-␣ responses in pDCs.
At low viral doses, H5N1 and H7N1 avian viruses, are more efficient
in infecting pDCs when compared to human H1N1, and at inducing 3.1. Acute phase proteins
the secretion of IFN-␣ (Bel et al., 2011).
The acute phase response is a nonspecific early response of
the organism caused by various stimuli. This reaction is mediated
2.2.4. Natural killer cells
by pro-inflammatory cytokines and involves local and systemic
NK cells are important effector cells of the innate immune
reactions, including changes in the concentration of acute phase
response. They can recognize antibody-bound IV infected cells and
proteins (APPs) (Heegaard et al., 1998; Pomorska-Mol et al.,
lyse these cells, a process called antibody dependent cell cyto-
2012a,b). APPs in swine are C-reactive protein (CRP), haptoglobin
toxicity (ADCC). These cells can recognize IV-infected cells with
(Hp), serum amyloid A (SAA) and pig major acute phase protein
their cytotoxicity receptors NKp44 and NKp46. Upon binding to
(Pig-MAP) and only a few studies have investigated the role of APPs
the influenza virus HA the receptors trigger the NK cells to lyse
in pigs during IV infection.
the infected cell (Mandelboim et al., 2001). It has been suggested
An increase of CRP and Hp concentrations in pigs was reported
that invariant NKT (iNKT) cells stimulate the induction of cellular
by Brookes et al. (2010) during acute IV infection caused by pan-
immunity and regulate infection induced pathology (Paget et al.,
demic H1N1 2009 virus (Table 2). The CRP levels peaked at 4 days
2011). An NK specific monoclonal antibody from pigs was recently
post infection (dpi)/days post contact (dpc), while Hp responses
reported (Mair et al., 2012) and iNKT cells were identified in pig
were more protracted, peaking at 9–11 dpi/dpc in both infected and
lungs (Renukaradhya et al., 2011). With the identification of NK cell
in-contact pigs. CRP responses were higher in infected pigs than in
markers in pigs, many studies on the role of these cells in influenza
pigs at first-fourth transmission cycles (TC1-4), and were more ele-
infected pigs will probably be addressed in the future.
vated in the infected and TC1 animals than the later transmission
cycles (Brookes et al., 2010).
3. In vivo immune responses during influenza infection in Barbe et al. (2011) also described in intratracheal swH1N1
pigs (A/Swine/Belgium/1/98) infected pigs that CRP, Hp, lipopolysaccha-
ride (LPS)-binding protein (LBP) peaked in broncho-alveolar lavage
The three swIV subtypes (H1N1, H1N2 and H3N2) have been fluid (BALF) at 2 dpi (CRP, Hp) or 30 dpi (LBP) and then decreased.
associated with disease in swine. Subclinical infections are also While the concentration of CRP and Hp peaked at 48 h in serum, the
very common and many, if not most pigs become infected with one level of LBP remained relatively constant (LBP increased in BALF and
or more IV subtypes without ever showing clinical signs (Charley not in serum). APPs reached higher levels in serum than in BALF and
et al., 2006; Van Reeth, 2007). Serological profiles after consecutive did not directly correlate with viral load. Given the small number of
experimental infections of pigs with European H1N1, H3N2, and pigs, APPs concentration post infection did not differ significantly
H1N2 swIV have been described by Van Reeth et al. (2006). The from control values (Barbe et al., 2011).
disease caused by IVs in pigs is essentially similar to that recorded A recent study of APP responses after subclinical intranasal
in human, with low mortality but high morbidity (Brown, 2000; infection with swine H1N1 (A/Sw/Poland/KPR9/2004) in pigs
Fouchier et al., 2003). showed that in this model Hp and SAA were significantly induced
Pigs are susceptible to infection with IV from both avian and from 1 to 2 dpi, before specific Abs in serum were present
human origin with occasional transmission of virus to human. (Pomorska-Mol et al., 2012b) (Fig. 2). Although the concentration
Examples are the pandemic of 1957, 1968 and the 2009 pandemic of CRP and Pig-MAP remained generally unchanged to the end of
which were caused by a reassortant influenza (Fouchier et al., 2003) the study, in half of the infected pigs, the concentration of CRP
that had been generated in pigs (Garten et al., 2009). tended to increase at 1 dpi (Pomorska-Mol et al., 2012b). A similar
The purpose of this section is to summarize the immune study was also performed by the same group using H1N2 subtype
responses during an IV infection in swine considering pigs exper- (Pomorska-Mol et al., 2012a) (Table 2). In infected pigs concentra-
imentally or naturally infected with human, avian or porcine tion of CRP, Hp, SAA increased significantly at the time when the
subtypes of IV (swIVs summarized in Fig. 2). most amount of virus was shed (from 1 to 3 dpi) (Fig. 2) and the
206 E. Crisci et al. / Molecular Immunology 55 (2013) 200–211

Table 2
In vivo studies on influenza viruses and pigs.

Type Virus subtype identification Route of inoculation Age of pigs References

SwIV H1N1 A/Sw/Belgium/1/83 Intratracheal 3-week-old Van Reeth et al. (1998)


Cesarian
Clostrum-deprived
H1N1 A/Sw/Belgium/1/98 Intratracheal 10- to 14-week-old Van Reeth et al. (2002a,b)
H3N2 A/Sw/Flanders/1/98
H1N2 A/Sw/Gent/7625/99
H1N1 A/Sw/Indiana/1726/88 Intranasal 8-week-old Larsen et al. (2000)
H1N1 A/Sw/Best/96 Culture supernatant 10-week-old Heinen et al. (2001)
H3N2 A/Sw/Oedenrode/96 aereosola
H3N2 A/Sw/Neth/St.Oedenrode/96 Culture supernatant SPF Heinen et al. (2000)
aereosola 10-week-old

H1N1 A/Sw/Neth/Best/96 Culture supernatant 7- to 15-week-old Loeffen et al. (2003)


aereosola
H1N1 A/Swine/IA/40776/92 Lung homogenate 5-week-old Kim et al. (2006)
H3N2 A/Swine/IA/41305/98 nebulization
H1N2 A/Swine/Korea/S5/05 Intranasal 6-week-old Jo et al. (2007)
H1N1 A/Swine/IA/40776/92 Intratracheal 5-week-old Kitikoon et al. (2008)
H1N2 A/Swine/Wisconsin/R33f/01
H3N2 A/Swine/Texas/4199-2/98
H1N1 A/Swine/IA/35233/99
H1N2 A/Swine/Indiana/9K035/99
H3N2 A/Swine/Wisconsin/R7c/01
H1N1 A/Sw/Finistere Intranasal 8-week-old Ferrari et al. (2008)
Intrapharingeal
H1N1 A/Sw/OH/24366/07 Intranasal 5-week-old Khatri et al. (2010)
Intratracheal
H1N2 A/Sw/Grandstedt/IDT3475/2004 Intranasal 5-week-old Pomorska-Mol et al. (2012a,b)
SwIV (rH3N2p) A/Swine/Illinois/A01201606/2011 Intranasal 3-week-old Kitikoon et al. (2012)
HuIV (H3N2-TRIG) A/Swine/Pennsylvania/62170-1/2010 Intratracheal
HuIV H3N2 A/Indiana/08/2011
HuIV H3N2 A/Sidney/5/97 Intranasal 4-week-old Seo et al. (2004)
H1N1 A/New Caledonia/1/99
Influenza B/Shichuan/99
H1N1 A/New Caledonia/20/99 Intranasal 6-week-old Kim et al. (2008)
H1N1 1918/rec Intranasal 4-week-old Weingartl et al. (2009)
Intratracheal
H1N1 A/Regensburg/D6/09 Intranasal 10-week-old Lange et al. (2009)
H1N1 A/California/04/09 Intranasal 8-week-old SPF miniature Itoh et al. (2009)
pigs Brookes et al. (2010)
H1N1 A/California/07/09 4- to 5-week-old
aIV 42 strains of IV (38 aIV) Intranasal 50- to 80-day-old Kida et al. (1994)
aIV subtypes:
H2N2, H2N8, H2N9,H3N2,H3N6,
H3N8,H4N5,H4N6,H4N8,H4N9,H5N2,H5N3,H6N1,
H6N2,H7N1, H7N2,H7N3,H8N4,H9N2,H9N5,
H10N1,H10N3,H10N8,H10N9,H11N9,H12N5,H13N6
aIV H5N1 A/Ck/Vt/C-58/04 Intranasal 4-week-old Choi et al. (2005)
HPAI H5N1 A/Dk/Th/D4AT/04
H5N1 A/Gs/Th/G7CS/04
H5N1 A/Chicken/Yamaguchi/7/04 Intranasal 8-week-old Isoda et al. (2006)
H5N1 A/Duck/Yokohama/aq-10/03 Miniature pigs
H5N1 R
(A/Duck/Mongolia/54/01-A/Duck/Mongolia/47/01)
H5N1 A/Chicken/Indonesia/7/03 Intranasal 2- to 3-week-old Lipatov et al. (2008)
H5N1 A/Whooper swan/Mongolia/244/05
H5N1 A/Muscovy duck/Vietnam/209/05
H5N1 A/goose/Hubei/ZFE/2004 Intranasal 4-week-old Li et al. (2008)

Intranasal route mimics natural infection, whereas intratracheal challenge is a better way to investigate the pathogenesis.
a
Aereosol produced by nebulization of 2 ml culture supernatant using airbrush device (Badger).

level of Pig-MAP remained unchanged during the study. They found development of the active immunity after an infection with a
a significant positive correlation between maximum concentration homologous or heterologous virus. Loeffen et al. (2003) used
of SAA in serum and lung scores, suggesting SAA as a useful indi- homologous infection with H1N1 SwIV (Table 2) and showed
cator in experimental infection or as a marker for disease severity that MDA protected piglets against the clinical consequences
(Pomorska-Mol et al., 2012a). of a primary IV, but that this protection was not complete.
Piglets with and without MDA were protected against clinical
3.2. Swine influenza virus consequences of a secondary infection; but pigs with MDA devel-
oped weaker immunity than other animals without MDA. This
3.2.1. Role of maternal antibodies study opened the way to consider the role of MDA in IV infec-
Few studies have been performed to determine the role of tion and overall growth performance from weaning to slaughter
maternally derived antibodies (MDA) against IV in the clini- showed a trend in favor of pigs without MDA (Loeffen et al.,
cal protection of piglets and particularly their effect on the 2003).
E. Crisci et al. / Molecular Immunology 55 (2013) 200–211 207

Concerning the presence of MDA and the use of vaccination, These studies were complemented by Kim et al. (2006))who
Kitikoon et al. (2006) have shown that MDA suppressed serum Ab described Ab responses to specific viral proteins in pigs infected
responses and the induction of SwIV-specific memory T cells fol- with H1N1 and H3N2 swIV (Table 2). Both IgM and IgG against HA,
lowing the administration of a bivalent inactivated vaccine in pigs. NP, NS1 and NS2 were detected by 7 dpi. IgG against these pro-
Enhancement of lung pneumonia was observed in immunized pigs teins were still detectable at 28 dpi. In contrast, IgG against NA and
in the presence of MDA when challenged with a heterologous H1N1 M1 were not detected until 14 dpi and no IgM against these pro-
virus (Kitikoon et al., 2006). These authors also tested levels of M2e- teins was detected. This study was useful in determining that the
specific Abs in pigs that were positive for SwIV-specific MDA. These NP protein was a suitable antigen for a subtype-unrestricted sero-
Abs were low and did not appear to be correlated with the higher logic assay, whereas NS1 was for serologic assays that differentiates
HI Ab titers. Variation in the SwIV exposure profile of the sow may between infected and vaccinated pigs (Kim et al., 2006). Taking
account for the difference in M2e-specific Ab levels of the MDA+ into account humoral responses, the responses to the extracellu-
pigs (Kitikoon et al., 2008). Finally, when compared to the previ- lar region of M2 (M2e) and M1 were also investigated (Kitikoon
ous study with H1N1, similar results have also been observed with et al., 2008). In this study, pigs were tested with different swIV
H3N2 based vaccines using homologous and heterologous infection (Table 2) and the primary objective was to characterize the M2-
(Vincent et al., 2012). specific antibody response. Low M2 antibody responses occurred
following primary infection and the antibody titers did not increase
3.2.2. H1N1 and H3N2 influenza viruses following homologous challenge, but did increase following het-
The first article regarding pigs that demonstrated the produc- erologous challenge. When seroconversion occurred, the infection
tion of TNF-␣ and IL-1 in the BAL of pigs was published in the late dose had a major effect on HA, M2e but not the M1 antibody level
90s by Van Reeth et al. (1998) and later reviewed in (Van Reeth, (Kitikoon et al., 2008).
2000; Van Reeth et al., 2002b). Biological active IFN-␣, TNF-␣ and Another experimental design used swIV H1N1 (Table 2) in con-
IL-1 were detected in the BALF of pigs inoculated with H1N1 swIV ventional pigs (Khatri et al., 2010). In this study, they focused on
(Table 2). Cytokine titers and lung virus titers were significantly the host mucosal immune responses and they were the first to
higher 18–24 h pi than at 48–72 h pi. All the cytokines were posi- demonstrate the phenotype of different immune cells associated
tively correlated with a 3- to 4-fold increase in BAL cell numbers and with innate, adaptive, and regulatory immune responses during
with a drastic neutrophil infiltration. In addition, cytokine produc- the acute phase of the disease, in particular at the site of viral
tion coincided with the onset of general and respiratory symptoms replication. Infected pigs showed: (1) higher frequency of CD8+
of influenza and with the development of a necrotizing bronchop- cells in BAL, tonsil, lung and tracheobronchial lymph nodes (TBLN),
neumonia (Van Reeth et al., 1998). This study was complemented indicating the proliferation of IV-specific CD8+ cells; (2) increased
by a follow up article from the same group reporting conventional frequency of activated CD4+ cells associated with enhanced gen-
pigs inoculated with all the circulating subtypes of swIV (Table 2) eration of swIV-specific mucosal IgA and an increased frequency
(Van Reeth et al., 2002a). The major finding in this last study was of activated CD8+ cells in lungs; (3) higher frequencies of CD4+
that as previously seen, IFN-␣, TNF-␣ and also IL-6 correlated with CD8+ cells in BAL, TBLN and tonsils; (4) NK cells in lungs, BAL and
virus titer in the lungs (Fig. 2) and symptom severity, while there PBMCs decreased; (5) enrichment of the DC fraction and larger
was not a clear correlation for IL-1. They suggested that the discrep- amounts of IL-12 in lungs; (6) increased amount of IFN-␥ in lungs
ancy in IL-1 between the two studies may be due to differences and TBLN; (7) increase of ␥␦ T cells in BAL and lung; 8) increase
in the sanitary status of the pigs. They also assessed for the first of Treg, IL-10 and TGF-␤ in lungs. In conclusion they detected
time IL-8 in swIV-infected pigs. IL-8 correlated with virus titers innate, proinflammatory, Th1, Th2 and Th3 cytokines, as well as
and appeared to be induced by swIV infection. These two studies swIV-specific IgA in lungs of infected pigs. Production of IFN-␥ by
argued for the role of IFN-␣, IL-6 and TNF-␣ in swIV pathogenesis lymphocytes of the TBLN was also detected. Higher frequencies
(Van Reeth et al., 1998, 2002a). Another study highlighted the role of CTLs ␥␦ T cells, DCs, activated T cells, CD4+ and CD8+ T cells
of IFN-␣ in IL-6 and IL-12 induction in swIV infection using IFN-␣ were detected in the infected-pig lungs. Concomitantly, higher fre-
neutralizing Abs (Barbe et al., 2009). quencies of the immunosuppressive T regulatory cells were also
In 2000, a more detailed investigation on systemic and mucosal detected in the infected pigs. This study highlighted the striking
responses was performed during H1N1 swIV infection in pigs by similarities between the pathogenesis of swIV and the pandemic
Olsen’s group (Larsen et al., 2000) (Table 2). Virus-specific serum H1N1 virus in humans, suggesting the pig as a useful model for
IgG, IgA and HI titers all peaked 2–3 weeks after the primary pathogenesis studies (Khatri et al., 2010).
infection and did not substantially increase after re-challenge. As In the same way, Heinen et al. (2001) previously described the
expected, the predominant virus-specific isotype in serum was IgG primary infection of pigs with swIV H1N1 and H3N2 (Table 2),
whereas IgA was the predominant isotype in upper and lower respi- focusing on cellular immunity. An acute inflammatory response
ratory tract. Nevertheless, pigs also responded with virus-specific characterized by a massive increase in neutrophils from 6 to 40% (2
IgG in both upper (nasal washes) and lower (BAL) airways. Results dpi) of all leukocytes in the lungs was observed. From 8 dpi the % of
of Ab-secreting cell ELISPOT assays demonstrated that the numbers lymphocytes in the lungs was higher than controls and remained
of both IgG and IgA secreting cells in the nasal mucosa were higher elevated until 42 dpi for both subtypes. Additionally after H3N2
than in any other tissue examined. IFN-␥ secreting cells (Fig. 2), as infection different events occurred: (a) an increase in the percent-
a measure of T-cell responses, were predominantly localized to the age of Th cells starting at 2 dpi, (b) peak in the percentage of Th
spleen and tracheo-bronchial lymph nodes and peaked on day 21 cells between 4 and 11 dpi, (c) a massive infiltration of CTLs on 8
after primary infection (Larsen et al., 2000). Similar results were dpi, leading to a decrease in the percentage of NKs, (d) the tem-
described by Ferrari et al. (2008) with swIV H1N1 (Table 2). Virus porary disappearance of the CTLs from lung around 11 dpi and (e)
was isolated from 1 to 5 dpi only after the first inoculation and a long-lived increase in the percentage of CTLs in the lungs and
cell-mediated response showed an increase of T lymphocyte pro- the decrease in NK to the pre-infection level. The same sequence
liferation from 14 to 20 dpi after the first infection (Fig. 2). Further was also observed after H1N1 infection, but it developed faster
enhancement, was observed even earlier (7 days) during the sec- than after H3N2 infection. Finally, they demonstrated the existence
ond infection and a plateau phase at high values until the end of the of a clear heterosubtypic immunity (Het-I) in pigs, that partially
experiment (47 dpi). IFN-␥ producing cells increased in spleen and protected H1N1-infected pigs from H3N2 secondary infection and
in peripheral blood from 14 to 40 dpi (Fig. 2) (Ferrari et al., 2008). they suggested that CD8+ T cells have a role in Het-I (Heinen et al.,
208 E. Crisci et al. / Molecular Immunology 55 (2013) 200–211

2001). The same authors (Heinen et al., 2000), in a previous study, (Seo et al., 2004). Alveolar M infected with human H3N2 (Table 2)
described the systemic and mucosal Ab responses in serum, BAL and showed greater expression of TNF-␣ than did alveolar M infected
nasal lavage fluid (NLF). The Ab response to infection was character- with human H1N1 or influenza B virus (Table 2) and they did not
ized by high IgA titers in BAL and NLF. Moreover, substantial titers undergo apoptosis when infected. IL-8 and IL-1 were not induced
of IgG1 were detected in BAL and less in NLF. The specific activity in alveolar M in vivo and TNF-␣ peaked at 5 dpi without a strong
of IgA was much higher in NLF and BAL than in serum. The specific correlation with viral titers (peak 3 dpi) (Seo et al., 2004). Succes-
activity of IgG1 was higher in BALF than in serum, suggesting a local sively, Kim et al. demonstrated that alveolar M are essential for
production of this isotype in the lung also. IgA was the predominant controlling human H1N1 (Table 2) in pigs. In fact, in vivo deple-
Ig in NLF and IgG in BAL. The appearance of influenza-specific IgM tion of alveolar M results in 40% mortality when pigs are infected
between 4 and 7 dpi coincided with the clearance of virus from the with currently circulating human H1N1, while none of the infected
oropharyngeal tract. The authors suggested that IgM are respon- control pigs dies. Additionally, more severe respiratory signs were
sible for viral clearance after a primary infection, but in contrast found in depleted pigs and TNF-␣ was significantly lower and IL-10
to IgA, IgM probably has a minor role in long lasting immunity to higher in depleted animals than controls. Lower Ab titers and CD8+
secondary infection (Heinen et al., 2000). T cells expressing IFN-␥ were detectable in depleted pigs (Kim et al.,
With regard to H3N2 infection, one of the most recent articles 2008).
describes infection in pigs with the novel porcine H3N2 reassor- Human 1918 pandemic IV was tested in pigs; the virus was not
tant viruses (H3N2-TRIG and rH3N2p) and a human H3N2 virus lethal, in contrast to other species (mice, ferrets and macaques), and
(Table 2) (Kitikoon et al., 2012). This study did not detect an increase pigs did not develop severe respiratory distress (Table 2) (Weingartl
in virulence in human H3N2 or rH3N2p viruses compared to the et al., 2009). In this study virus was isolated at 3 and 5 dpi, and
endemic H3N2-TRIG virus and the Abs specific to the H3N2-TRIG piglets developed antibodies at 7 dpi, but a profound investigation
virus cross-reacted to both reassortant H3N2 viruses. No further on immune responses was not performed (Weingartl et al., 2009).
immunological studies were performed during these infections. The first work with the recent A(H1N1)pdm/2009 was per-
In conclusion swIV H1N1 and H3N2 infections showed similar formed by Lange et al. (2009) (Table 2). NP- and H1-specific Abs
patterns in various studies: (1) a rapid increase of cytokines corre- could be detected 7 dpi and CD4+ T cells became activated imme-
lated with viral lung titers and APPs in the early stage; (2) the peak diately after the infection. Both CD4+ and CD8+ T lymphocytes
of humoral responses after 14 dpi; and (3) the beginning of cellu- expanded from 3 to 7 dpi coinciding with clinical signs (Lange
lar responses at the site of infection after 7 dpi. Some discrepancy et al., 2009). Subsequently, Brookes et al. (2010) used a similar
between the different systems may be related to the animals and virus (Table 2) that in infected pigs produced clinical signs, viral
the experimental design used. pathogenesis restricted to the respiratory tract, infection dynamics
consistent with endemic pig strains, virus transmissibility between
3.2.3. H1N2 influenza viruses pigs and virus-host adaptation events (Brookes et al., 2010). In
This subtype kindled little interest in swine research and the miniature pigs, the pandemic virus did not cause any signs and
first work on porcine H1N2 pathogenesis was performed by Jo titers were detected in lungs on 3 dpi (Itoh et al., 2009) (Table 2).
et al. (2007) (Table 2). In this work high levels of IL-1, IL-8 and In summary, pigs are susceptible to different types of huIV and
TNF-␣ were detected in lungs of infected pigs, while the induced alveolar M have been shown to have an important role during this
amount of IL-4, IL-10 and IFN-␥ was similar in infected and control infection. Further studies have to be performed to full understand
pigs. Among the cytokines tested TNF-␣ was the highest induced the immune responses during huIV infection in pigs.
cytokine and peaked at 5 dpi, after the beginning of virus shedding
by nasal discharge (3 dpi) (Jo et al., 2007). 3.4. Avian influenza virus
Subsequently, Kim et al. (2009) described the same cytokines
in BAL of 3-week-old pigs intranasally inoculated with swIV H1N2. Pigs are clearly susceptible to infection with both LPAI and HPAI
In BAL the TNF-␣ concentration was maximal at 1 dpi and declined viruses and LPAI viruses have occasionally been isolated from pigs
markedly by 3 dpi. The body temperatures were correlated with the in the field (Table IV (Van Reeth, 2007)). Most of these viruses have
levels of TNF-␣ in BAL and the pulmonary lesion scores correlated an H1 or H3, HA subtype that are usually restricted to birds and
with TNF-␣ positive cells by IHQ (Kim et al., 2009). can also cross the species barrier to pigs. Serological studies in Asia
The most recent study was performed by Pomorska-Mol et al. have shown evidence for infections of pigs with avian H4, H5 and
(2012a) (Table 2), focusing attention on the APPs and the immune H9 viruses (Van Reeth, 2007). Few studies have been performed on
responses. In this study Abs against swIV H1N2 in the serum of the avian IV infection in pigs and most of them have been focused on
infected pigs were detected from 7 dpi and a specific T cell response the pathogenesis and transmission of HPAI H5N1, since this virus
was observed from 5 dpi. Starting from 7 dpi a significant antigen- infected humans causing high mortality (Choi et al., 2005; Isoda
specific response was seen in all infected animals. A significant et al., 2006; Lipatov et al., 2008). In this section we summarize the
drop in lymphocyte numbers and an increase in medium-sized most important studies performed with aIV in pigs.
cell counts with no accompanying leucopenia were observed from One of the first experimental studies in pigs with different aIV
3 to 7 dpi in all infected animals. The mean number of lympho- was performed by Kida et al. (1994) in 1994. In this work pigs were
cytes did not return to the mean pre-inoculated concentration and inoculated with 38 LPAI aIVs (Table 2), mainly of duck origin, and
the mean number of medium-sized cells returned to the normal 27 with non-human-type HA. At least one strain of each HA sub-
concentration at 10 dpi (Pomorska-Mol et al., 2012a). type replicated in the porcine respiratory tract for 5–7 days and
In conclusion, immune responses to swIV H1N2 infection seem most of them were detected in nasal swabs and induced a sero-
to be slightly delayed compared to the other H1N1 and H3N2 sub- logical response. Sera of infected pigs showed high Ab titers in
types. ELISA and neutralization tests, but did not inhibit haemagglutina-
tion of homologous viruses. Co-infection of pigs with a swIV and
3.3. Human influenza virus with an aIV able to replicate in these animals generating reassor-
tant viruses. Thus, these results indicated that aIV with or without
The first study in vivo that demonstrated that huIV can infect non-human-type HA can be transmitted to pigs (Kida et al., 1994).
alveolar porcine M that constitutively reside in the respiratory Most recent studies have focused attention on H5N1 aIV to
tract, without causing apoptosis in pigs, was published in 2004 determine whether this virus associated with human infections had
E. Crisci et al. / Molecular Immunology 55 (2013) 200–211 209

been transmitted to pigs. Choi et al. (2005) studied different 2004 assess porcine immune responses, which will facilitate the future
Asian H5N1 viruses (Table 2) in pigs, revealing that all viruses tested development of pigs as the “gold standard” large-animal model in
replicated in the swine respiratory tract but none were transmit- immunology, and more precisely for IV immunology.
ted to contact pigs. Virus titers from nasal swabs peaked on day
2 pi and low titers were detected in the liver of two of the four Funding
pigs tested. Their findings indicated that pigs can be infected with
highly lethal Asian H5N1 viruses but that these viruses are not Our work in this field was funded by the project AGL2010-
readily transmitted between pigs under experimental conditions 22200-C02-01 of Spanish Ministry of Science and Innovation.
(Choi et al., 2005). In the same way, a follow up paper, assessed HPAI
H5N1 viruses (Table 2) in different species of birds and mammals
Conflict of interests
(Isoda et al., 2006). In this study the authors showed that miniature
pigs were resistant to infection with the viruses tested (Isoda et al.,
The authors declare no financial conflict of interests.
2006). Finally, Lipatov et al. (2008) tested other HPAI H5N1 viruses
(Table 2) in pigs. Strains were different from the other studies,
but also in this case pigs demonstrated low susceptibility to infec- Acknowledgement
tion with HPAI H5N1 viruses. Inoculation of pigs with these viruses
resulted in asymptomatic to mild symptomatic infection restricted We want to thank Davide Boscolo for Fig. 1 (www.
to the respiratory tract, mainly to the lungs, and tonsils (Lipatov davideboscolo.org).
et al., 2008). This was also confirmed by recent surveillance stud-
ies that observed that H5N1 aIV was circulating in pigs in Indonesia References
without causing disease. In this study the authors reported that one
isolate recognized human-type receptors, meaning that the H5N1 Aigner, B., Renner, S., Kessler, B., Klymiuk, N., Kurome, M., Wunsch, A., Wolf, E.,
2010. Transgenic pigs as models for translational biomedical research. Journal
can replicate for long periods undetected and facilitate the adapt- of Molecular Medicine (Berlin) 88, 653–664.
ion of aIV to mammalian host (Nidom et al., 2010). Li et al. (2008) Arranz, R., Coloma, R., Chichon, F.J., Conesa, J.J., Carrascosa, J.L., Valpuesta, J.M., Ortin,
describe H5N1 infection (Table 2) in different species, and in this J., Martin-Benito, J., 2012. The structure of native influenza virion ribonucleo-
proteins. Science 338 (December (6114)), 1634–1637.
experimental infection pigs showed “flu-like” clinical symptoms. Barbe, F., Atanasova, K., Van Reeth, K., 2011. Cytokines and acute phase proteins
Animals developed severe respiratory symptoms within 5 dpi and associated with acute swine influenza infection in pigs. Veterinary Journal 187,
a continuous fever of over 40 ◦ C for 5–7 days. Specific Abs against 48–53.
Barbe, F., Saelens, X., Braeckmans, D., Lefevre, F., Reeth, K.V., 2009. Role of IFN-
the virus were detected at 28 and 45 dpi (Li et al., 2008).
alpha during the acute stage of a swine influenza virus infection. Research in
In recent years porcine research with aIVs were directed mainly Veterinary Science 88, 172–178.
toward the role of the cross-protection between different IV. Two Bateman, A.C., Karasin, A.I., Olsen, C.W., 2013. Differentiated swine airway epithelial
studies performed by Van Reeth’s group showed how prior infec- cell cultures for the investigation of influenza A virus infection and replication.
Influenza and Other Respiratory Viruses 7 (March (2)), 139–150.
tion with swIV may be a total or partial barrier to infection with aIVs Bel, M., Ocana-Macchi, M., Liniger, M., McCullough, K.C., Matrosovich, M., Sum-
(De Vleeschauwer and Van Reeth, 2010; Van Reeth et al., 2009). merfield, A., 2011. Efficient sensing of avian influenza viruses by porcine
In conclusion, pigs are susceptible to different aIVs but H5 plasmacytoid dendritic cells. Viruses 3, 312–330.
Belser, J.A., Katz, J.M., Tumpey, T.M., 2011. The ferret as a model organism to study
subtype infections are in general abortive and not able to be trans- influenza A virus infection. Disease Models and Mechanisms 4, 575–579.
mitted between pigs. Thus, the risk of aIV infection is mainly Brookes, S.M., Nunez, A., Choudhury, B., Matrosovich, M., Essen, S.C., Clifford, D.,
related to the possibility of reassortment and not to the morbid- Slomka, M.J., Kuntz-Simon, G., Garcon, F., Nash, B., Hanna, A., Heegaard, P.M.,
Queguiner, S., Chiapponi, C., Bublot, M., Garcia, J.M., Gardner, R., Foni, E., Loeffen,
ity/transmissibility. Further studies have to be performed to clarify W., Larsen, L., Van Reeth, K., Banks, J., Irvine, R.M., Brown, I.H., 2010. Replication,
the transmission between pigs using different avian subtypes. pathogenesis and transmission of pandemic (H1N1) 2009 virus in non-immune
pigs. PLoS ONE 5, e9068.
Brown, I.H., 2000. The epidemiology and evolution of influenza viruses in pigs. Vet-
erinary Microbiology 74, 29–46.
4. Conclusions Brown, I.H., 2012. History and epidemiology of swine influenza in Europe. Current
Topics in Microbiology and Immunology January (Epub ahead of print).
Busch, M.G., Bateman, A.C., Landolt, G.A., Karasin, A.I., Brockman-Schneider, R.A.,
Influenza virus represents one of the human pathogen for
Gern, J.E., Suresh, M., Olsen, C.W., 2008. Identification of amino acids in the
which the porcine model has been successfully used, improving HA of H3 influenza viruses that determine infectivity levels in primary swine
the knowledge in this infectious human disease. Pigs are well respiratory epithelial cells. Virus Research 133, 269–279.
accepted as “mixing vessels” for IVs and their role in the emer- Busquets, N., Segales, J., Cordoba, L., Mussá, T., Crisci, E., Martin-Valls, G.E., Simon-
Grife, M., Perez-Simo, M., Perez-Maillo, M., Nunez, J.I., Abad, F.X., Fraile, L., Pina,
gence and epidemiology of influenza pandemic is well documented. S., Majo, N., Bensaid, A., Domingo, M., Montoya, M., 2010. Experimental infection
For this reason, new strategies are gaining attention in porcine with H1N1 European swine influenza virus protects pigs from an infection with
vaccination to develop future IV vaccine such as live-attenuated, the 2009 pandemic H1N1 human influenza virus. Veterinary Research 41, 74.
Bussfeld, D., Kaufmann, A., Meyer, R.G., Gemsa, D., Sprenger, H., 1998. Differential
subunit, vectored and DNA vaccines. This topic was extensively mononuclear leukocyte attracting chemokine production after stimulation with
discussed in a recent review by Van Reeth and Ma (2012). There- active and inactivated influenza A virus. Cellular Immunology 186, 1–7.
fore, immunological features associated with influenza infection Calzada-Nova, G., Schnitzlein, W., Husmann, R., Zuckermann, F.A., 2009. Characteri-
zation of the cytokine and maturation responses of pure populations of porcine
are now of great interest in influenza research for the similari- plasmacytoid dendritic cells to porcine viruses and toll-like receptor agonists.
ties with human immunity. An exhaustive recent review (Meurens Veterinary Immunology and Immunopathology 135, 20–33.
et al., 2012) highlighted the importance of the pig model for Charley, B., Riffault, S., Van Reeth, K., 2006. Porcine innate and adaptative immune
responses to influenza and coronavirus infections. Annals of the New York
human infectious disease research and vaccine development, and
Academy of Sciences 1081, 130–136.
humanized piglets have been described as “stand-ins” for micro- Choi, Y.K., Nguyen, T.D., Ozaki, H., Webby, R.J., Puthavathana, P., Buranathal, C.,
biome studies (Hvistendahl, 2012). Additionally, pig-to-primate Chaisingh, A., Auewarakul, P., Hanh, N.T., Ma, S.K., Hui, P.Y., Guan, Y., Peiris,
J.S., Webster, R.G., 2005. Studies of H5N1 influenza virus infection of pigs by
organ xenotransplantation have been described (Ibrahim et al.,
using viruses isolated in Vietnam and Thailand in 2004. Journal of Virology 79,
2006) and transgenic pigs have been proposed as a model for 10821–10825.
translational biomedical research (Aigner et al., 2010) or develop- De Vleeschauwer, A., Van Reeth, K., 2010. Prior infection of pigs with swine influenza
mental immunology (Rothkotter et al., 2002). Thus, the increased viruses is a barrier to infection with avian influenza viruses. Veterinary Micro-
biology 15, 340–345.
acceptance of pigs as a suitable and valuable model in the scien- Ferrari, M., Candotti, P., Lombardi, G., Amadori, M., Dotti, S., Guana, S., Petrini,
tific community may stimulate the development of new tools to S., 2008. A comparison of the humoral and cell-mediated response of pigs
210 E. Crisci et al. / Molecular Immunology 55 (2013) 200–211

experimentally infected with either influenza or PRRS viruses. Veterinary Kim, W.I., Wu, W.H., Janke, B., Yoon, K.J., 2006. Characterization of the humoral
Research Communications 32 (Suppl. 1), S199–S201. immune response of experimentally infected and vaccinated pigs to swine
Fouchier, R.A., Osterhaus, A.D., Brown, I.H., 2003. Animal influenza virus surveillance. influenza viral proteins. Archives of Virology 151, 23–36.
Vaccine 21, 1754–1757. Kitikoon, P., Nilubol, D., Erickson, B.J., Janke, B.H., Hoover, T.C., Sornsen, S.A., Thacker,
Gao, W., Sun, W., Qu, B., Cardona, C.J., Powell, K., Wegner, M., Shi, Y., Xing, Z., 2012. E.L., 2006. The immune response and maternal antibody interference to a
Distinct regulation of host responses by ERK and JNK MAP kinases in swine heterologous H1N1 swine influenza virus infection following vaccination. Vet-
macrophages infected with pandemic (H1N1) 2009 influenza virus. PLoS ONE 7, erinary Immunology and Immunopathology 112, 117–128.
e30328. Kitikoon, P., Strait, E.L., Thacker, E.L., 2008. The antibody responses to swine influenza
Garten, R.J., Davis, C.T., Russell, C.A., Shu, B., Lindstrom, S., Balish, A., Sessions, W.M., virus (SIV) recombinant matrix 1 (rM1), matrix 2 (M2), and hemagglutinin
Xu, X., Skepner, E., Deyde, V., Okomo-Adhiambo, M., Gubareva, L., Barnes, J., (HA) proteins in pigs with different SIV exposure. Veterinary Microbiology 126,
Smith, C.B., Emery, S.L., Hillman, M.J., Rivailler, P., Smagala, J., de Graaf, M., Burke, 51–62.
D.F., Fouchier, R.A., Pappas, C., Alpuche-Aranda, C.M., Lopez-Gatell, H., Olivera, Kitikoon, P., Vincent, A.L., Gauger, P.C., Schlink, S.N., Bayles, D.O., Gramer, M.R., Dar-
H., Lopez, I., Myers, C.A., Faix, D., Blair, P.J., Yu, C., Keene, K.M., Dotson Jr., P.D., nell, D., Webby, R.J., Lager, K.M., Swenson, S.L., Klimov, A., 2012. Pathogenicity
Boxrud, D., Sambol, A.R., Abid, S.H., St George, K., Bannerman, T., Moore, A.L., and transmission in pigs of the novel A(H3N2)v influenza virus isolated from
Stringer, D.J., Blevins, P., Demmler-Harrison, G.J., Ginsberg, M., Kriner, P., Water- humans and characterization of swine H3N2 viruses isolated in 2010-2011.
man, S., Smole, S., Guevara, H.F., Belongia, E.A., Clark, P.A., Beatrice, S.T., Donis, Journal of Virology 86, 6804–6814.
R., Katz, J., Finelli, L., Bridges, C.B., Shaw, M., Jernigan, D.B., Uyeki, T.M., Smith, Kreijtz, J.H., Fouchier, R.A., Rimmelzwaan, G.F., 2011. Immune responses to influenza
D.J., Klimov, A.I., Cox, N.J., 2009. Antigenic and genetic characteristics of swine- virus infection. Virus Research 162, 19–30.
origin 2009 A(H1N1) influenza viruses circulating in humans. Science 325, Lange, E., Kalthoff, D., Blohm, U., Teifke, J.P., Breithaupt, A., Maresch, C., Starick, E.,
197–201. Fereidouni, S., Hoffmann, B., Mettenleiter, T.C., Beer, M., Vahlenkamp, T.W., 2009.
Genzel, Y., Dietzsch, C., Rapp, E., Schwarzer, J., Reichl, U., 2010. MDCK and Vero cells Pathogenesis and transmission of the novel swine-origin influenza virus A/H1N1
for influenza virus vaccine production: a one-to-one comparison up to lab-scale after experimental infection of pigs. Journal of General Virology 90, 2119–2123.
bioreactor cultivation. Applied Microbiology and Biotechnology 88, 461–475. Larsen, D.L., Karasin, A., Zuckermann, F., Olsen, C.W., 2000. Systemic and mucosal
GeurtsvanKessel, C.H., Lambrecht, B.N., 2008. Division of labor between dendritic immune responses to H1N1 influenza virus infection in pigs. Veterinary Micro-
cell subsets of the lung. Mucosal Immunology 1, 442–450. biology 74, 117–131.
Heegaard, P.M., Klausen, J., Nielsen, J.P., Gonzalez-Ramon, N., Pineiro, M., Lamp- Li, X., Jin, M., Yu, Z., Zhang, A., Chen, H., Qian, P., 2008. Pathogenicity of a goose isolate
reave, F., Alava, M.A., 1998. The porcine acute phase response to infection with of highly pathogenic H5N1 influenza A virus for chickens, mice, and pigs. Acta
Actinobacillus pleuropneumoniae. Haptoglobin, C-reactive protein, major acute Virologica 52, 41–46.
phase protein and serum amyloid A protein are sensitive indicators of infection. Lipatov, A.S., Kwon, Y.K., Sarmento, L.V., Lager, K.M., Spackman, E., Suarez, D.L.,
Comparative Biochemistry and Physiology. Part B: Biochemistry and Molecular Swayne, D.E., 2008. Domestic pigs have low susceptibility to H5N1 highly
Biology 119, 365–373. pathogenic avian influenza viruses. PLoS Pathogens 4, e1000102.
Heinen, P.P., de Boer-Luijtze, E.A., Bianchi, A.T., 2001. Respiratory and systemic Loeffen, W.L., Heinen, P.P., Bianchi, A.T., Hunneman, W.A., Verheijden, J.H., 2003.
humoral and cellular immune responses of pigs to a heterosubtypic influenza A Effect of maternally derived antibodies on the clinical signs and immune
virus infection. Journal of General Virology 82, 2697–2707. response in pigs after primary and secondary infection with an influenza H1N1
Heinen, P.P., van Nieuwstadt, A.P., Pol, J.M., de Boer-Luijtze, E.A., van Oirschot, J.T., virus. Veterinary Immunology and Immunopathology 92, 23–35.
Bianchi, A.T., 2000. Systemic and mucosal isotype-specific antibody responses Lombardo, T., Dotti, S., Renzi, S., Ferrari, M., 2012. Susceptibility of different cell lines
in pigs to experimental influenza virus infection. Viral Immunology 13, to Avian and Swine Influenza viruses. Journal of Virological Methods 185, 82–88.
237–247. Londt, B.Z., Brookes, S.M., Nash, B.J., Nunez, A., Stagg, D.A., Brown, I.H., 2012. The
Hillaire, M.L., van Eijk, M., van Trierum, S.E., van Riel, D., Saelens, X., Romijn, R.A., infectivity of pandemic 2009 H1N1 and avian influenza viruses for pigs: an
Hemrika, W., Fouchier, R.A., Kuiken, T., Osterhaus, A.D., Haagsman, H.P., Rim- assessment by ex vivo respiratory tract organ culture. Influenza and Other Respi-
melzwaan, G.F., 2011. Assessment of the antiviral properties of recombinant ratory Viruses June, http://dx.doi.org/10.1111/j. 1750-2659.2012.00397.x (Epub
porcine SP-D against various influenza A viruses in vitro. PLoS ONE 6, e25005. ahead of print).
Hofmann, P., Sprenger, H., Kaufmann, A., Bender, A., Hasse, C., Nain, M., Gemsa, D., Ma, W., Lager, K.M., Vincent, A.L., Janke, B.H., Gramer, M.R., Richt, J.A., 2009. The role
1997. Susceptibility of mononuclear phagocytes to influenza A virus infection of swine in the generation of novel influenza viruses. Zoonoses Public Health 56,
and possible role in the antiviral response. Journal of Leukocyte Biology 61, 326–337.
408–414. Mair, K.H., Essler, S.E., Patzl, M., Storset, A.K., Saalmuller, A., Gerner, W., 2012. NKp46
Hvistendahl, M., 2012. Pigs as stand-ins for microbiome studies. Science 336, 1250. expression discriminates porcine NK cells with different functional properties.
Ibrahim, Z., Busch, J., Awwad, M., Wagner, R., Wells, K., Cooper, D.K., 2006. Selected European Journal of Immunology 42, 1261–1271.
physiologic compatibilities and incompatibilities between human and porcine Mandelboim, O., Lieberman, N., Lev, M., Paul, L., Arnon, T.I., Bushkin, Y., Davis, D.M.,
organ systems. Xenotransplantation 13, 488–499. Strominger, J.L., Yewdell, J.W., Porgador, A., 2001. Recognition of haemagglu-
Isoda, N., Sakoda, Y., Kishida, N., Bai, G.R., Matsuda, K., Umemura, T., Kida, tinins on virus-infected cells by NKp46 activates lysis by human NK cells. Nature
H., 2006. Pathogenicity of a highly pathogenic avian influenza virus, 409, 1055–1060.
A/chicken/Yamaguchi/7/04 (H5N1) in different species of birds and mammals. Medina, R.A., Garcia-Sastre, A., 2011. Influenza A viruses: new research develop-
Archives of Virology 151, 1267–1279. ments. Nature Reviews Microbiology 9, 590–603.
Itoh, Y., Shinya, K., Kiso, M., Watanabe, T., Sakoda, Y., Hatta, M., Muramoto, Y., Meurens, F., Summerfield, A., Nauwynck, H., Saif, L., Gerdts, V., 2012. The pig: a model
Tamura, D., Sakai-Tagawa, Y., Noda, T., Sakabe, S., Imai, M., Hatta, Y., Watan- for human infectious diseases. Trends in Microbiology 20, 50–57.
abe, S., Li, C., Yamada, S., Fujii, K., Murakami, S., Imai, H., Kakugawa, S., Ito, M., Mussá, T., Ballester, M., Silva-Campa, E., Baratelli, M., Busquets, N., Lecours, M.P.,
Takano, R., Iwatsuki-Horimoto, K., Shimojima, M., Horimoto, T., Goto, H., Taka- Dominguez, J., Amadori, M., Fraile, L., Hernández, J., Montoya, M. Infection by
hashi, K., Makino, A., Ishigaki, H., Nakayama, M., Okamatsu, M., Warshauer, D., swine, human or avian influenza viruses differentially activates porcine den-
Shult, P.A., Saito, R., Suzuki, H., Furuta, Y., Yamashita, M., Mitamura, K., Nakano, dritic cells cytokine profile. Veterinary Immunology and Immunopathology,
K., Nakamura, M., Brockman-Schneider, R., Mitamura, H., Yamazaki, M., Sug- accepted for publication.
aya, N., Suresh, M., Ozawa, M., Neumann, G., Gern, J., Kida, H., Ogasawara, K., Mussá, T., Rodriguez-Carino, C., Pujol, M., Cordoba, L., Busquets, N., Crisci, E.,
Kawaoka, Y., 2009. In vitro and in vivo characterization of new swine-origin Dominguez, J., Fraile, L., Montoya, M., 2011. Interaction of porcine conventional
H1N1 influenza viruses. Nature 460, 1021–1025. dendritic cells with swine influenza virus. Virology 420, 125–134.
Jo, S.K., Kim, H.S., Cho, S.W., Seo, S.H., 2007. Pathogenesis and inflammatory Nayak, D.P., Balogun, R.A., Yamada, H., Zhou, Z.H., Barman, S., 2009. Influenza virus
responses of swine H1N2 influenza viruses in pigs. Virus Research 129, 64–70. morphogenesis and budding. Virus Research 143, 147–161.
Khatri, M., Dwivedi, V., Krakowka, S., Manickam, C., Ali, A., Wang, L., Qin, Z., Nayak, D.P., Hui, E.K., Barman, S., 2004. Assembly and budding of influenza virus.
Renukaradhya, G.J., Lee, C.W., 2010. Swine influenza H1N1 virus induces acute Virus Research 106, 147–165.
inflammatory immune responses in pig lungs: a potential animal model for Nidom, C.A., Takano, R., Yamada, S., Sakai-Tagawa, Y., Daulay, S., Aswadi, D., Suzuki,
human H1N1 influenza virus. Journal of Virology 84, 11210–11218. T., Suzuki, Y., Shinya, K., Iwatsuki-Horimoto, K., Muramoto, Y., Kawaoka, Y., 2010.
Khatri, M., Goyal, S.M., Saif, Y.M., 2012. Oct4+ stem/progenitor Swine lung epithe- Influenza A (H5N1) viruses from pigs, Indonesia. Emerging Infectious Diseases
lial cells are targets for influenza virus replication. Journal of Virology 86, 16, 1515–1523.
6427–6433. Nunes, S.F., Murcia, P.R., Tiley, L.S., Brown, I.H., Tucker, A.W., Maskell, D.J., Wood,
Khatri, M., Saif, Y.M., 2011. Epithelial cells derived from swine bone marrow express J.L., 2010. An ex vivo swine tracheal organ culture for the study of influenza
stem cell markers and support influenza virus replication in vitro. PLoS One 6, infection. Influenza and Other Respiratory Viruses 4, 7–15.
e29567, http://dx.doi.org/10.1371/journal.pone.0029567. Ocana-Macchi, M., Ricklin, M.E., Python, S., Monika, G.A., Stech, J., Stech, O., Summer-
Kida, H., Ito, T., Yasuda, J., Shimizu, Y., Itakura, C., Shortridge, K.F., Kawaoka, Y., Web- field, A., 2012. Avian influenza A virus PB2 promotes interferon type I inducing
ster, R.G., 1994. Potential for transmission of avian influenza viruses to pigs. properties of a swine strain in porcine dendritic cells. Virology 427, 1–9.
Journal of General Virology 75 (Pt 9), 2183–2188. Paget, C., Ivanov, S., Fontaine, J., Blanc, F., Pichavant, M., Renneson, J., Bialecki, E.,
Kim, B., Ahn, K.K., Ha, Y., Lee, Y.H., Kim, D., Lim, J.H., Kim, S.H., Kim, M.Y., Cho, K.D., Lee, Pothlichet, J., Vendeville, C., Barba-Spaeth, G., Huerre, M.R., Faveeuw, C., Si-Tahar,
B.H., Chae, C., 2009. Association of tumor necrosis factor-alpha with fever and M., Trottein, F., 2011. Potential role of invariant NKT cells in the control of pul-
pulmonary lesion score in pigs experimentally infected with swine influenza monary inflammation and CD8+ T cell response during acute influenza A virus
virus subtype H1N2. Journal of Veterinary Medical Science 71, 611–616. H3N2 pneumonia. Journal of Immunology 186, 5590–5602.
Kim, H.M., Lee, Y.W., Lee, K.J., Kim, H.S., Cho, S.W., van Rooijen, N., Guan, Y., Seo, S.H., Palese, P., Shaw, M.L., 2007. Orthomyxoviridae: the viruses and their replication. In:
2008. Alveolar macrophages are indispensable for controlling influenza viruses Knipe, D.M., Howley, P.M. (Eds.), Fields Virology. Lippincott Williams & Wilkins,
in lungs of pigs. Journal of Virology 82, 4265–4274. Philadelphia, pp. 1647–1689.
E. Crisci et al. / Molecular Immunology 55 (2013) 200–211 211

Pomorska-Mol, M., Markowska-Daniel, I., Kwit, K., 2012a. Immune and acute phase Van Reeth, K., 2000. Cytokines in the pathogenesis of influenza. Veterinary Micro-
response in pigs experimentally infected with H1N2 swine influenza virus. FEMS biology 74, 109–116.
Immunology and Medical Microbiology 66, 334–342. Van Reeth, K., 2007. Avian and swine influenza viruses: our current understanding
Pomorska-Mol, M., Markowska-Daniel, I., Pejsak, Z., 2012b. Acute phase protein of the zoonotic risk. Veterinary Research 38, 243–260.
response during subclinical infection of pigs with H1N1 swine influenza virus. Van Reeth, K., Braeckmans, D., Cox, E., Van Borm, S., van den Berg, T., Goddeeris, B.,
Veterinary Microbiology 159, 499–503. De Vleeschauwer, A., 2009. Prior infection with an H1N1 swine influenza virus
Punyadarsaniya, D., Liang, C.H., Winter, C., Petersen, H., Rautenschlein, S., Hennig- partially protects pigs against a low pathogenic H5N1 avian influenza virus.
Pauka, I., Schwegmann-Wessels, C., Wu, C.Y., Wong, C.H., Herrler, G., 2011. Vaccine 27, 6330–6339.
Infection of differentiated porcine airway epithelial cells by influenza virus: dif- Van Reeth, K., Labarque, G., Pensaert, M., 2006. Serological profiles after consecutive
ferential susceptibility to infection by porcine and avian viruses. PLoS ONE 6, experimental infections of pigs with European H1N1, H3N2, and H1N2 swine
e28429. influenza viruses. Viral Immunology 19, 373–382.
Renukaradhya, G.J., Manickam, C., Khatri, M., Rauf, A., Li, X., Tsuji, M., Rajashekara, G., Van Reeth, K., Ma, W., 2012. Swine influenza virus vaccines: to change or not to
Dwivedi, V., 2011. Functional invariant NKT cells in pig lungs regulate the airway change-that’s the question. Current Topics in Microbiology and Immunology
hyperreactivity: a potential animal model. Journal of Clinical Immunology 31, September (Epub ahead of print).
228–239. Van Reeth, K., Nauwynck, H., Pensaert, M., 1998. Bronchoalveolar interferon-alpha,
Rhoads, J.M., Chen, W., Chu, P., Berschneider, H.M., Argenzio, R.A., Paradiso, A.M., tumor necrosis factor-alpha, interleukin-1, and inflammation during acute
1994. l-Glutamine and l-asparagine stimulate Na+ –H+ exchange in porcine jeju- influenza in pigs: a possible model for humans? Journal of Infectious Diseases
nal enterocytes. American Journal of Physiology 266, G828–G838. 177, 1076–1079.
Rothkotter, H.J., Sowa, E., Pabst, R., 2002. The pig as a model of developmental Van Reeth, K., Van Gucht, S., Pensaert, M., 2002a. Correlations between lung
immunology. Human and Experimental Toxicology 21, 533–536. proinflammatory cytokine levels, virus replication, and disease after swine
Seo, S.H., Webby, R., Webster, R.G., 2004. No apoptotic deaths and different levels influenza virus challenge of vaccination-immune pigs. Viral Immunology 15,
of inductions of inflammatory cytokines in alveolar macrophages infected with 583–594.
influenza viruses. Virology 329, 270–279. Van Reeth, K., Van Gucht, S., Pensaert, M., 2002b. In vivo studies on cytokine
Sun, Y., Bi, Y., Pu, J., Hu, Y., Wang, J., Gao, H., Liu, L., Xu, Q., Tan, Y., Liu, M., Guo, X., involvement during acute viral respiratory disease of swine: trouble-
Yang, H., Liu, J., 2010. Guinea pig model for evaluating the potential public health some but rewarding. Veterinary Immunology and Immunopathology 87,
risk of swine and avian influenza viruses. PLoS ONE 5, e15537. 161–168.
Sun, Z., Huber, V.C., McCormick, K., Kaushik, R.S., Boon, A.C., Zhu, L., Hause, B., Webby, Vincent, A.L., Ma, W., Lager, K.M., Richt, J.A., Janke, B.H., Sandbulte, M.R., Gauger,
R.J., Fang, Y., 2012. Characterization of a porcine intestinal epithelial cell line for P.C., Loving, C.L., Webby, R.J., Garcia-Sastre, A., 2012. Live attenuated influenza
influenza virus production. Journal of General Virology 93, 2008–2016. vaccine provides superior protection from heterologous infection in pigs with
Tobita, K., Sugiura, A., Enomote, C., Furuyama, M., 1975. Plaque assay and primary maternal antibodies without inducing vaccine-associated enhanced respiratory
isolation of influenza A viruses in an established line of canine kidney cells disease. Journal of Virology 86, 10597–10605.
(MDCK) in the presence of trypsin. Medical Microbiology and Immunology 162, Webster, R.G., Bean, W.J., Gorman, O.T., Chambers, T.M., Kawaoka, Y., 1992.
9–14. Evolution and ecology of influenza A viruses. Microbiological Reviews 56,
Tong, S., Li, Y., Rivailler, P., Conrardy, C., Castillo, D.A., Chen, L.M., Recuenco, S., Ellison, 152–179.
J.A., Davis, C.T., York, I.A., Turmelle, A.S., Moran, D., Rogers, S., Shi, M., Tao, Y., Weil, Weingartl, H.M., Albrecht, R.A., Lager, K.M., Babiuk, S., Marszal, P., Neufeld, J.,
M.R., Tang, K., Rowe, L.A., Sammons, S., Xu, X., Frace, M., Lindblade, K.A., Cox, N.J., Embury-Hyatt, C., Lekcharoensuk, P., Tumpey, T.M., Garcia-Sastre, A., Richt, J.A.,
Anderson, L.J., Rupprecht, C.E., Donis, R.O., 2012. A distinct lineage of influenza A 2009. Experimental infection of pigs with the human 1918 pandemic influenza
virus from bats. Proceedings of the National Academy of Sciences of the United virus. Journal of Virology 83, 4287–4296.
States of America 109, 4269–4274. Zhu, H., Zhou, B., Fan, X., Lam, T.T., Wang, J., Chen, A., Chen, X., Chen, H., Webster,
Van Poucke, S.G., Nicholls, J.M., Nauwynck, H.J., Van Reeth, K., 2010. Replication of R.G., Webby, R., Peiris, J.S., Smith, D.K., Guan, Y., 2011. Novel reassortment of
avian, human and swine influenza viruses in porcine respiratory explants and Eurasian avian-like and pandemic/2009 influenza viruses in swine: infectious
association with sialic acid distribution. Virology Journal 7, 38. potential for humans. Journal of Virology 85, 10432–10439.

You might also like