Está en la página 1de 17

Review

Veterinary Pathology
2014, Vol 51(2) 410-426
Influenza A Virus Infections in Swine: ª The Author(s) 2013
Reprints and permission:
sagepub.com/journalsPermissions.nav
Pathogenesis and Diagnosis DOI: 10.1177/0300985813513043
vet.sagepub.com

B. H. Janke1

Abstract
Influenza has been recognized as a respiratory disease in swine since its first appearance concurrent with the 1918 ‘‘Spanish flu’’
human pandemic. All influenza viruses of significance in swine are type A, subtype H1N1, H1N2, or H3N2 viruses. Influenza
viruses infect epithelial cells lining the surface of the respiratory tract, inducing prominent necrotizing bronchitis and
bronchiolitis and variable interstitial pneumonia. Cell death is due to direct virus infection and to insult directed by leukocytes
and cytokines of the innate immune system. The most virulent viruses consistently express the following characteristics of
infection: (1) higher or more prolonged virus replication, (2) excessive cytokine induction, and (3) replication in the lower
respiratory tract. Nearly all the viral proteins contribute to virulence. Pigs are susceptible to infection with both human and
avian viruses, which often results in gene reassortment between these viruses and endemic swine viruses. The receptors on the
epithelial cells lining the respiratory tract are major determinants of infection by influenza viruses from other hosts. The
polymerases, especially PB2, also influence cross-species infection. Methods of diagnosis and characterization of influenza
viruses that infect swine have improved over the years, driven both by the availability of new technologies and by the necessity
of keeping up with changes in the virus. Testing of oral fluids from pigs for virus and antibody is a recent development that
allows efficient sampling of large numbers of animals.

Keywords
influenza virus, swine, pathogenesis, cytokines, apoptosis, virulence, cross-species infection, hemagglutinin, receptors, poly-
merases, diagnosis

The evolution and epidemiology of influenza A virus (IAV) Pathogenesis of Infection


infections in swine have been integrally tied to the segmented
Although the genetic makeup of influenza viruses in swine var-
nature of the genome of the virus, which facilitates gene
ies, the clinical disease and lesions induced by the classic H1N1
reassortment, and to the adaptability of these genes through
swine influenza virus,52,53,101,139,158,166 avian-like H1N1 viruses
mutation, which sometimes facilitates replication in a new
host.8,19,77,85,89 Influenza was first recognized as a respiratory adapted to swine,9,24,29 and various H1N1,21,84,134,155,158,159
H1N2,29,56,155,158 H3N2,76,118 and H2N387 reassortants (includ-
disease entity in swine during the 1918 human Spanish flu pan-
ing the recent 2009 human pandemic H1N178,156 and subsequent
demic.67 The virus was not isolated until 1931, when nasal dis-
reassortants15,63) are essentially the same. The gross and micro-
charge from pigs was inoculated into ferrets and subsequently
scopic lesions, clinical signs, virus distribution in tissues, and
into embryonated chicken eggs.129 In 1933, a similar virus was
virus shedding patterns have been described with variable
recovered from humans with this technique.131 The viruses
emphasis in the above studies and in other reports on isolates
were essentially identical and served as the prototype for type
of interest, in comparison studies, and in vaccine trials. Two
A subtype H1N1 influenza viruses.
All influenza viruses of significance that infect swine are of the earliest studies, one employing histopathology and immu-
nofluorescence,101 the other histopathology and ultrastructural
type A viruses, defined by the highly conserved internal
nucleoproteins and matrix proteins. Influenza A virus subtypes
are based on the external hemagglutinin (HA) and neuramini- 1
Veterinary Diagnostic Laboratory, Department of Veterinary Diagnostic and
dase (NA) proteins. The predominant viruses in swine popula- Production Animal Medicine, College of Veterinary Medicine, Iowa State
tions in different parts of the world may have genes of different University, Ames, IA, USA
origin, even though the subtypes may be similar.10,73,171 The
original classic H1N1 swine influenza virus, which remained Corresponding Author:
B. H. Janke, DVM, PhD, Veterinary Diagnostic Laboratory, Department of
the predominant influenza virus in North American swine Veterinary Diagnostic and Production Animal Medicine, College of Veterinary
populations for nearly 70 years, has been almost completely Medicine, Iowa State University, Ames, IA 50011, USA.
displaced by subsequent reassortant viruses.157 Email: bhjanke@iastate.edu
Janke 411

characterization,166 still provide some of the most detailed histo- tightly packed tall columnar cells, resulting in an irregular hyper-
pathologic descriptions. plastic appearance, often by 72 hours PI (Fig. 3). In severely
In experimentally infected pigs inoculated with a sufficient damaged airways, proliferation of fibroblasts from exposed
dose of challenge virus (2 ml of 106 egg infectious dose 50 subepithelial lamina propria may result in formation of endo-
[EID50] or tissue culture infectious dose 50 [TCID50] intratra- bronchial polyps or incomplete splitting of repairing airways
cheally) and in the individual pig with natural infection, the (bronchiolitis obliterans) (Fig. 4). Such damage is most likely
course of infection is very short (5–7 days) with minimal virus to occur in the smaller airways.
detected after this time. In the immunofluorescent study,101 the In both field cases and experimental challenge studies, the
first evidence of virus infection was a pale fluorescence in the sizes of airways that are affected may vary. In the author’s
nucleus of bronchial epithelial cells by 2 hours postinoculation experience, the largest noncartilaginous airways (primary
(PI). By 4 hours PI, virus antigen was abundant in both nucleus bronchioles) are most consistently and most severely affected,
and cytoplasm of infected cells. In the ultrastructural study,166 likely due to the dynamics of airflow that affect droplet suspen-
virus was observed budding from the surface of type II pneu- sion and deposition or possibly due to receptor distribution.
mocytes as early as 5 hours PI. In a more recent ultrastructural There is some variation between cases, and the larger lobar or
study on a differentiated human airway epithelial cell line, vir- segmental bronchi may be spared or only minimally affected,
ions in high numbers were budding from cell surfaces at 24 or conversely, only these larger airways may be damaged. Like-
hours PI, predominantly from the tips of microvilli of both wise, the smallest terminal or respiratory bronchioles also are
ciliated and secretory cell types.69 Although virus has been frequently spared but in severe extensive infections may exhibit
detected in mediastinal lymph nodes, blood, and other tissues the characteristic epithelial necrosis. An early immunofluores-
such as brain in a few studies in swine,9,24,77 productive influ- cent study on sequentially sampled pigs described infection as
enza virus replication with consequent tissue damage is limited progressive, beginning in the small bronchi followed by distal
in this species to the respiratory tract. spread to terminal bronchioles and alveoli as well as proximally
Because the virus enters the lung through the airways, the to bronchi and the trachea,101 which may suggest that some of
most consistent macroscopic manifestation of influenza virus the variation in lesion distribution is a function of time after
infection is cranioventral bronchopneumonia that affects a infection or of the position of the lobule in question along the
variable percentage of the lung. In milder infections, small infected branch of the respiratory tree.
clusters of medium to dark red lobules are located in the cranial Simultaneous to the onset and progression of bronchial and
and middle lung lobes and occasionally in cranioventral por- bronchiolar damage and inflammation, degenerative and inflam-
tions of the caudal lobes and in accessory lobes (Fig. 1). In matory lesions develop in the alveoli.101,166 During the first
some pigs, these foci of consolidation are somewhat linear and 24 hours PI, alveolar walls, primarily peribronchiolar in location,
confluent and situated in the hilar region at the junction of the are widened by vascular congestion and interstitial edema.
lung lobes. In more severe or extensive infections, most or all Endothelial cells lining the capillaries are swollen, and neutro-
of the cranial and middle lung lobes and cranioventral portions phils accumulate in capillary lumens and marginate against
of the caudal lobes may be consolidated. Less frequently, vessel walls. Type II pneumocytes become swollen. Neutrophils,
severe acute infection presents as a diffusely congested lung macrophages, and lymphocytes lightly infiltrate the alveolar
with prominent interlobular edema and extensive foam in the interstitium and migrate into alveolar lumens. As in the airways,
bronchi and trachea. Such cases likely represent an exaggerated neutrophils predominate early in infection, but subsequently
cytokine response, and in such lungs, the characteristic cranio- (by 48 hours PI), the alveolar luminal exudate becomes pre-
ventral lobular consolidation is obscured by the more diffuse dominantly mononuclear. These cells appear to be a mixture
interstitial pneumonia and edema. of sloughed swollen pneumocytes and macrophages. By close
In swine, influenza viruses primarily infect the epithelial cells examination, necrosis of constituent cells may be identified
lining the surface of the respiratory tract, from nasal mucosa to in alveolar walls, but the septa usually remain intact. In severe
alveoli, but virus also has been detected in the glandular epithe- infections, necrotic cellular debris accumulates in alveolar
lial cells associated with the larger airways. The hallmark micro- lumens, or rarely, the necrotic debris and exudative proteins
scopic lesion is necrotizing bronchitis and bronchiolitis. During form hyaline membranes. In such cases, alveolar and interlobu-
the first 24 hours PI, neutrophils begin to accumulate in the vas- lar edema is usually evident. Swollen alveolar epithelial cells
culature adjacent to bronchioles, and light loose lymphocytic lining the lumen add to the thickness of the alveolar septa. In
cuffs appear around the airways, sometimes before epithelial some infections, in contrast to the alveolar lesions described
damage is evident. Necrosis and sloughing of airway epithelial above, alveoli may be affected only minimally.
cells and neutrophil transmigration into airway lumens occur Consistent with the multifocal macroscopic lesions in the
at 24 to 48 hours PI (Fig. 2). By 48 hours PI, affected airways lung, lobules within the same histopathologic section of lung
are lined by flattened epithelial layers, and neutrophils in the may differ in the degree of involvement, and unaffected lobules
luminal debris have been replaced or are accompanied by macro- often sit adjacent to severely affected lobules. This likely
phages. Lymphocyte accumulation around airways and adjacent reflects irregular distribution of virus through the branches
vessels becomes more prominent. Continuing in repair, the of the respiratory tract via inhalation of aerosolized virus.
epithelial cells lining the airways proliferate and mature into Experimentally, the virus titer in the inoculum and the route
412 Veterinary Pathology 51(2)

Figure 1. Lung in situ, 4-week-old pig, 5 days after experimental intratracheal inoculation with triple reassortant H3N2 swine influenza virus.
Multifocal lobular consolidation in cranial and middle lung lobes is evident. Figure 2. Lung, pig, field case. The acute phase of influenza A virus
infection is characterized by necrosis and sloughing of bronchiolar epithelial cells and flattening of the remaining cells lining the airway. The lumen
contains clusters of neutrophils and sloughed epithelial cells. Light peribronchiolar lymphocyte infiltration is evident. Hematoxylin and eosin
(HE). Figure 3. Lung, pig, field case. The subacute phase of influenza A virus infection is characterized by an irregular hyperplastic epithelial
layer lining the airway. The lumen contains mucus and a few neutrophils and macrophages. Transmigration of the epithelial layer by lymphocytes
and a few neutrophil clusters is evident. HE. Figure 4. Lung, pig, field case. A small bronchiole is disrupted by an endobronchiolar polyp (bronch-
iolitis obliterans), a sequel sometimes observed following severe infection with influenza A virus. HE.

of inoculation can affect lesion development.24,76 Inducing infection, airways disrupted by bronchiolitis obliterans may
clinical disease and significant lung lesions can be difficult provide evidence of earlier infection.
with intranasal inoculation as pigs may swallow much of the
inoculum, reducing the volume of inoculum that reaches the
lung in contrast to intratracheal inoculation. If the virus dose is Mechanisms of Cell Injury
too light, lesions may be minimal but multifocally asynchronous Research into the molecular mechanisms of influenza-induced
in development. The latter issue can interfere with studies that cell injury has been conducted primarily in vitro in cell lines of
evaluate lesion progression in sequential necropsies. nonswine origin with viruses not isolated from swine. Although
Subsequently, the epithelial hyperplasia resolves and all that in vitro studies have their limitations because of the artificial
remain are peribronchiolar lymphocytic cuffing and partially nature of the medium, control of variables is more readily
atelectatic alveoli with variable leukocyte populations. Although accomplished than in the in vivo studies. The segmented gen-
somewhat dependent on the extent of damage, lungs return ome of influenza viruses and the ability to isolate the effect
to normal by 2 weeks PI. However, in pigs that survive severe of specific proteins through generation of virus with the desired
Janke 413

genes by reverse genetics facilitate these studies. Commonal- induced more cellular apoptosis in vitro than less virulent
ities in mechanisms of infection likely exist regardless of the viruses of other subtypes.75
source of virus or host involved. Thus, in attempting to under-
stand pathogenesis of influenza virus infection at the cellular
level and the resulting disease in swine (or in other species), the
results of such studies warrant consideration. Findings suggest
Immunopathology
necrosis, apoptosis, and cytokines of the innate immune Numerous studies have described the cellular and cytokine
response all contribute to the destruction of influenza virus– response to influenza virus infection as measured in vivo in var-
infected cells. ious animal models and in vitro in cell cultures.11,14,54,55,59,126,148
Contradictory results from various studies on the interactions
of components of the inflammatory response suggest that
Necrosis the severity of tissue damage is determined by the balance
Much of the damage to the respiratory tract by influenza virus between proinflammatory and anti-inflammatory cytokine
can be attributed to direct viral destruction of infected cells.55,126 activity. An in-depth discussion of the complex multitude of
Influenza virus infection induces cytolysis of infected cells molecules involved in the innate immune response to influ-
through suppression of host cell gene expression and protein enza virus infection is beyond the scope of this review, but
synthesis. Viral polymerases bind to host cell polymerase tran- an excellent review article on immunopathology has recently
scripts and cleave host messenger RNAs (mRNAs) (50 cap been published.23 A brief summary of the mechanisms of
removal), which then degrade. Viral NS1 protein inhibits export destruction of influenza virus–infected cells by the innate
of host mRNAs from the nucleus, which greatly reduces transla- immune response is given below.
tion in the cytoplasm. Blockage of host cell protein synthesis, in Type 1 interferon (IFN), predominantly INF-a, is generated
concert with initiation of viral protein synthesis and virus parti- within a few hours by infected respiratory epithelial cells to
cle budding from cytoplasmic membranes, results in cytolytic inhibit viral replication and infection of adjacent cells31 and
death within 20 to 40 hours of infection. Necrosis is character- to initiate proinflammatory cytokine/chemokine responses.107
ized by disruption of cell membranes and swelling of cytoplasm Surface proteins expressed by the infected cells serve as signals
and mitochondria, followed by disintegration of organelles and for their destruction by cells of the innate immune system
total cell lysis. Cell death by necrosis stimulates a strong inflam- (killer cells, neutrophils, macrophages) resident in nearby tis-
matory response through cytokine induction. sues. Neutrophils destroy virus-infected cells through genera-
tion of oxidases and engulf debris from cells killed by virus
but also may damage tissue by release of other enzymes (eg,
Apoptosis elastase, myeloperoxidase).51,61
Cell death induced by influenza virus infection also occurs The leukocytic cells also effect destruction of virus-infected
through apoptosis by both extrinsic and intrinsic pathways.46 cells during the early innate immune response through genera-
Apoptosis has been most thoroughly studied in established cell tion of a variety of cytokines,* which also recruit additional
lines of both respiratory and nonrespiratory origin74,75,126 but leukocytes to the site of injury. The primary proinflammatory
has also been detected in respiratory tract cells collected fol- cytokines are tumor necrosis factor (TNF)–a, interleukin
lowing in vivo infection.98,145 Viral components that have been (IL)–1, and IL-6. TNF-a and IL-1 are strong upregulators of
most strongly implicated in the induction and regulation of leukocyte adhesion molecules on endothelial cells. TNF-a also
apoptosis include nonstructural proteins NS1 and PB1-F2 and increases microvascular permeability in the lung. IFN-a, TNF-
the ion channel structural protein M2.46,74,75 NS1 appears capa- a, and IL-1, IL-6, and IL-8 can be detected in bronchoalveolar
ble of exerting either proapoptotic or antiapoptotic influences, lavage fluid (BALF) of influenza virus-infected pigs, reaching
depending on intracellular conditions.46,74,75 Some studies sug- peak levels within 18 to 24 hours PI, followed by a drop to
gest viral neuraminidase99,124 and M1 proteins176 and single- or insignificant levels by 48 to 72 hours PI. This wave corre-
double-stranded RNA intermediates3 also influence apoptosis. sponds to a similar rise and fall in neutrophil numbers in BALF,
Inconsistent results between different studies with regard to the virus titers in lung, lesions of acute necrotizing bronchiolitis,
induction of necrosis vs apoptosis appear to be related to the and clinical illness. By 48 hours, macrophages have replaced
cell types studied and the viruses used. neutrophils as the predominant inflammatory cell in the
Although apoptosis has been proposed as a host defense lung.149
mechanism that reduces virus spread through rapid death of Cytokines released by innate immune cells also have effects
virus-infected cells and dampens inflammation through phago- that reach beyond the localized cellular environment. TNF-a
cytosis of apoptotic cell debris, recent studies suggest viruses has other well-described actions in the lung, including
can use the process to their advantage. Viruses may enhance increased bronchoconstriction and bronchial hyperreactivity,
their infectivity by inhibiting caspase activity until sufficient as well as changes in mucus and surfactant production. IFN-
replication of viral proteins has been completed.46,79,94,168
Contrarily, in a case of Highly Pathogenic Avian Influenza
(HPAI) zoonosis, an H5N1 virus isolated from a human fatality *References 36, 51, 61, 62, 109, 116, 144.
414 Veterinary Pathology 51(2)

a and IL-6 also have systemic effects such as induction of fever speculate that the potential may exist for similar vaccine-
and production of acute phase proteins by the liver.4,116 associated disease enhancement in humans.33
Although endothelial cells have not been considered targets of
influenza virus infection, recent studies suggest that endothelial
cells in the lung are significant modulators of both intracellular
and cytokine aspects of the acute response.34,107,138 These studies
Virulence
have been conducted primarily in mouse tissues and human In discussions on the pathogenesis of disease induced by influ-
endothelial cell lines using human viruses considered highly viru- enza virus infections, the question soon arises whether viruses
lent but not necessarily pandemic viruses. Various cytokines and vary in inherent virulence capabilities. The clinical signs asso-
cell surface signals play significant roles in the endothelial ciated with natural influenza virus infections in both swine and
damage. Endothelial cells variably exhibit necrosis or apoptosis, humans can vary considerably, from mild respiratory illness to
but the predominant change is loss of intercellular junctions, severe dyspnea followed shortly by death. Secondary bacterial
which results in vascular leakage.2 When prominent, this leakage infection can contribute to more severe pneumonia and higher
corresponds to extremely high cytokine levels in the tissues mortality rates in both species. Rapidly spreading epidemics,
(cytokine storm).162 Although some studies have described rec- sometimes accompanied by higher mortality rates, usually sig-
eptors for influenza virus on the surface of human endothelial nal introduction of a new virus strain against which the popu-
cells170 and virus infection of avian28,65 and human endothelial lation has little immune protection. However, some influenza
cells,175 these studies suggest the endothelial cell damage is due viral infections without bacterial augmentation and some epi-
to cytokines. Influenza virus infection of endothelial cells in demics with excessively high morbidity and/or mortality rates
swine lungs has not been documented. suggest the capacity for increased virulence may reside within
In summary, injury and destruction of the cells lining the some viruses. In swine, only very occasional influenza field
respiratory tract by influenza virus is effected by both direct virus outbreaks present with high mortality that appears to be
infection and by the cytokines of the innate immune response. The effected by virus alone.86 In humans, viruses responsible for
most severe infections are characterized by high cytokine levels pandemics usually elicit higher morbidity rates (likely reflec-
in the tissues. In humans, therapy directed toward important com- tive of greater transmissibility) and/or greater severity of clin-
ponents of the innate immune response may be as effective as ical disease (likely reflective of higher virus replication rates)
antiviral products in alleviating severe lung damage.107 than that observed with most seasonal influenzas, but overall
mortality rates may or may not be higher. Depending on the
pandemic, the incidence of infection may be higher in certain
Immune-Mediated Enhancement of Disease age segments of the population.
In an experimental trial evaluating the effect of an inactivated Pathologic studies on tissues from natural infections reveal
H1N1 vaccine on subsequent challenge with a heterologous some differences. In lungs of pigs submitted from high mortal-
virus, several pigs developed lung lesions dramatically more ity field cases, diffuse noncollapse and prominent interlobular
severe than that induced in nonvaccinated control pigs. The edema may mask the characteristic cranioventral consolida-
enhanced lesions were produced in a second trial with the same tion. Microscopically, the extent of airway involvement (the
vaccine and challenge virus and in additional trials with differ- number of affected airways, both larger and smaller, within
ent vaccines and challenge viruses.32,33,64,157 each lobule) and the severity of alveolar inflammation (wide-
The enhanced lesions were characterized macroscopically spread prominence of pneumocyte swelling, increased numbers
by involvement of a greater percentage of lung and microsco- of infiltrating cells in alveolar walls and lumens, and abun-
pically by more severe necrotizing and proliferative bronchio- dance of proteinaceous fluid in alveolar lumens) are the most
litis, more prominent peribronchiolar lymphocytic cuffing, and remarkable differences from routine infections. In humans,
severe lymphohistiocytic interstitial pneumonia. There was microscopic lesions induced by the most virulent influenza
more intense lymphocyte infiltration into the epithelial layer viruses, the 1918 H1N1 and HPAI H5N1 viruses, have been
and subepithelial lamina propria of the large- to medium- described in some studies as not differing markedly from non-
sized airways and suppurative bronchitis and bronchiolitis of pandemic infections but with the most significant differences
greater severity than that observed in the nonvaccinated, chal- involving the degree of inflammatory changes at the alveolar
lenged pigs. The epithelial lining of the trachea was also more level.137 The alveolar inflammation described in these studies
frequently disrupted and disorganized, and the subepithelial resembles that observed in severe swine cases.86 Others have
inflammation was more intense in the vaccinated pigs. described the lesions induced by the 1918 and 2009 pandemic
The antigenic relationship between vaccine virus and chal- H1N1 viruses and the HPAI H5N1 virus as involving both the
lenge virus or other factors that might contribute to this upper and lower portions of the respiratory tract, whereas a sea-
enhanced inflammatory response have not been determined. sonal H3N2 virus infection induced lesions primarily limited to
Such overzealous reactions likely occur in field situations, but the upper tract.42 Thus, results of pathologic examination of
the frequency of concurrent multipathogen infections that lungs of both naturally infected swine and humans suggest
result in more severe pneumonia makes identification of such viruses that induce inflammation in the lower respiratory tract
cases of immune-augmented disease difficult. The researchers cause the most severe disease.
Janke 415

In vivo challenge studies also have been used in the search for the early innate immune response appears to be responsible for
more virulent strains. With swine-origin viruses, comparative both of these characteristics.70
challenge studies conducted in pigs have not provided definitive Numerous studies, using both in vivo animal models and in
results. Attempts to experimentally reproduce severe disease vitro cell culture systems, have investigated the viral proteins
with isolates from high mortality field cases have not been suc- as the basis for these differences in virulence. Parameters eval-
cessful.86 Pigs challenged with viruses adapted to or endemic in uated in animal studies include severity of clinical disease or
swine usually develop only mild clinical disease and similar lesions and virus titers or cytokine levels resulting from infec-
lesions of mild to moderate severity. Although some studies tion. Although evaluation of virulence in the in vitro systems
describe more extensive lung involvement, comparison of is not technically plausible, increased production of virus or
results from different researchers must consider the impact of cytokines (if the cell type is capable of such production) is
factors that may vary between studies, such as virus titer in interpreted in these studies to be evidence for possible contri-
inoculum, method of restraint and route of inoculation, age of bution to virulence. These studies often use viruses generated
pigs, and so on.24,76,118 In most comparative studies with ende- by reverse genetics to evaluate the effect of gene replacement
mic viruses run concurrently under the same protocols, lesion or gene mutation from a more or less virulent strain on an
differences have not tended to be dramatic. However, measure- otherwise identical virus.
ment of quantifiable parameters such as fever, cytokine levels in Many such studies report findings that run counter to earlier
lung or BALF, and virus titers in nasal secretions, lung tissue, or work or reach opposite or different conclusions when initial
BALF can supply additional data that often correlate well with studies are extended to other viruses, hosts, or experimental
gross and microscopic lung scores. Such studies provide further protocols. Nearly all viral proteins have been implicated as
evidence of possible virulence differences between viruses. virulence factors, either individually or in concert with other
Some studies suggest that the triple reassortant viruses may be proteins.17,88,123 Such studies emphasize the polygenic nature
more virulent than the classic H1N1 viruses,158 especially those of virulence attributes in influenza viruses, the complexity of
reassortants that have been recovered from circumstances in the interactions between viral proteins, and the importance of
which both pigs and humans were infected.159 differences between hosts (and tissues within hosts) in the dis-
Pigs also appear to be relatively resistant to infection with ease process. Several recent articles have summarized current
viruses that cause disease in other host species. Infection of pigs knowledge of how viral proteins affect virulence.6,70,143 The
with low-pathogenicity avian influenza (LPAI) viruses or seaso- reported roles for various specific viral proteins in virulence are
nal human influenza viruses usually results in limited viral repli- described below.
cation with mild clinical disease and lesions, unless the virus
adapts through genetic mutation.77 Even inoculation with viruses
such as the 1918 human pandemic H1N1 virus and HPAI H5N1
Hemagglutinin
that induce severe disease in other mammals (humans, primates, The hemagglutinin (HA) is a protein protruding from the sur-
ferrets, and mice) only results in disease of similar or less sever- face of the virus that is responsible for attachment of virus to
ity than that induced by endemic swine viruses.82,164 host cells, internalization of virus, and intracellular fusion of
With viruses associated with human disease, challenge stud- viral capsid with endosomal membranes. In avian species, the
ies seeking to identify or characterize virulent strains have of cleavage site in the HA plays an important role in virulence,
necessity been conducted in animal models, primarily mice, which is manifested as systemic infection. The single basic
ferrets, and macaques, and in the in vitro studies in influenza- amino acid adjacent to the cleavage site in LPAI viruses is sus-
susceptible cell lines. Mice have been used extensively to study ceptible to proteases that are limited to the gastrointestinal and
influenza viruses but are not a natural host for these viruses; respiratory tracts. The H5 (and H7) in HPAI viruses has a
human viruses do not cause disease in mice unless first adapted sequence of multiple basic amino acids that allows cleavage
through serial passage.6 Ferrets are a better model for human by furin-like proteases that are present in many tissues, thus
influenza infections.5,7,153 Pigs probably would be an even closer permitting lethal systemic infection.135
model,59 but the logistics and cost of maintaining swine for Influenza viruses that naturally infect mammalian hosts do
experimental studies precludes their use in many research insti- not have a multibasic cleavage site, and systemic infection is
tutions. In mouse and ferret models, clear differences in lesion not a normal characteristic of influenza virus infection in mam-
severity between the HPAI H5N1 or 1918 human pandemic mals. Artificially inserting a multiple basic amino acid
H1N1viruses and the seasonal viruses are evident. The severity sequence by reverse genetics into the HA of mildly virulent
of disease induced in ferrets by the 2009 human pandemic H1N1 viruses does not result in increased virulence or systemic infec-
virus is intermediate.42,146 tion in ferrets.122 However, the HPAI H5N1 virus, which does
In summary from these studies, influenza viruses that are have a multibasic cleavage site, can be lethal for mice and fer-
more virulent in humans consistently express 2 characteristics rets in which it induces increased cytokine levels and virus
of infection: (1) higher or more prolonged levels of virus repli- titers and results in systemic infection.136 This does not occur
cation and (2) induction of a strong proinflammatory response in pigs,82 which may reflect a difference in tissue proteases
(cytokine storm), a significant result of which is increased per- in this species. The HA from the 1918 human pandemic
meability of alveolar vasculature.70,81,113,162 Dysregulation of H1N1virus, which does not have a multibasic cleavage site but
416 Veterinary Pathology 51(2)

still appears to be highly cleavable, also confers increased temperature sensitivity (ie, increased replication rates at the war-
virulence to reassortants as reflected in increased virus titers mer body temperatures of birds). Lysine at this site (627K) favors
and inflammation in mice and human airway cells.66,111 Thus, adaptation to and increased virulence in mammalian hosts, which
no consistent association has been established between a have cooler temperatures in the upper respiratory tract.90 Another
sequence of multiple basic amino acids in the HA and virulence mutation in PB2 that favors mammalian host infection is D701N
of viruses possessing such a sequence for mammalian species. (aspartic acid to asparagine at position 701), as manifested by
increased virus replication.30 In larger studies using single and
multiple gene reassortants, it became obvious that interaction
Neuraminidase between the polymerases was as important as the individual poly-
Neuraminidase (NA) is an external protein that prevents aggre- merases. Compatibility of the polymerases with each other as well
gation of viral progeny upon release from infected cells. as with the other viral proteins appeared to be critical.17,88,133
Balanced function of HA and NA results in the most efficient Surprisingly, the pandemic 2009 H1N1 virus, which was
virus replication, and alterations in the NA protein can interfere more virulent than most seasonal influenza viruses for humans
with this balance, resulting in decreased virulence. In studies of and in experimental mammalian hosts, did not have the reputed
HPAI H5N1 in mice, changes in the length and glycosylation of virulence marker mutations in its PB2 protein.45 When the
NA influenced virulence.93 Some NA proteins inherently molecular mutations in PB2 suggested to be virulence markers
appear to contribute to greater virulence. In a study with reas- for avian viruses were introduced individually into the avian-
sortant 1918 human pandemic virus and contemporary H1N1 origin PB2 of the pandemic 2009 H1N1 virus, virulence was
viruses, NA from the 1918 human pandemic H1N1 virus con- not altered.45 In summary, the polymerases appear to influence
tributed to high virus replication rates in both mouse lungs and virulence through control of protein synthesis processes that
human airway cells.111 In studies of reassortants between pan- favor virus production over cell maintenance functions, but
demic 2009 H1N1 and seasonal human viruses inoculated into sequence variations between polymerases will likely preclude
ferrets, reassortants containing the H3N2 NA were more viru- identification of definitive virulence markers for all viruses.
lent, whereas other reassortants were attenuated.123 Some stud-
ies suggest the NA protein also contributes to apoptosis.99,124
PB1-F2
The PB1-F2 protein is a nonstructural protein that is expressed
Polymerases during infection as an alternative open reading frame of the
The polymerases direct cell processes toward virus replication PB1 gene.72,130,151 Within host cells, the protein associates most
over cell maintenance functions. Within the virus, a trimeric extensively with mitochondria but has also been detected in both
polymerase gene complex is bound with the nucleoprotein cytoplasm and nucleus.18,94 The results of multiple in vitro and
attached to each genome segment. After virus entry into the cell in vivo studies in various cell lines and experimental animals
and release from the endosome, the viral ribonucleoproteins suggest the protein contributes to virulence through several func-
and template RNA are transported to the nucleus, where viral tions: (1) apoptosis induction, primarily in cells of the innate
mRNA is transcribed by the polymerases (RNA-dependent immune system; (2) suppression of early interferon response
RNA polymerase). PB1 is the catalyst, and PB2 removes 50 by infected cells; (3) increased viral replication rates or delayed
caps from host pre-mRNA molecules (cap-snatching), assisted viral clearance; and (4) increased inflammation in host tissues.
by the endonuclease PA, for use as primers for viral mRNA Nearly all of these functions require cooperative interaction of
production. These viral mRNAs are moved to the cytoplasm for the PB1-F2 with other cellular or virus-induced proteins.41,80,169
translation of viral proteins and for additional rounds of tran- Some effects of the protein involve dampening of the host
scription.6,12 It is to the advantage of the virus to maintain the response to infection. Localization of the protein to mitochondria
function of cellular mRNA synthesis. The polymerases control in macrophages results in mitochondrial membrane depolariza-
the steps of production of viral components, and increased rates tion and permeabilization, initiating death of these cells via the
of activity could result in increased viral replication.6 intrinsic apoptosis pathway.18,172 Studies also have demonstrated
Reverse genetics techniques have been used extensively to suppression of interferon induction by PB1-F2 in both epithelial
study the individual polymerases and to compare the virulence and immune cells in the in vitro and in vivo models.22,151
of different gene reassortmentsy and point mutations within spe- Other studies have focused on the effect of the protein on
cific polymerases.12,83,177,178 All 3 polymerases have some effect virus multiplication and host tissue response. Co-localization
on virulence in various studies, including PA47,133,178 and PB1,133 of the protein with PB1 in the nucleus leads to increased poly-
but PB2 protein seems to most significantly alter pathogeni- merase activity and likely consequent increased viral replica-
city.88,133,177 The amino acid at position 627 of the PB2 protein tion.94 In a mouse model, insertion of PB1-F2 from the 1918
influences virulence and host preference. A glutamic acid at this H1N1 pandemic virus into a laboratory-adapted virus resulted
site (627E) favors replication in avian hosts, possibly through in a higher viral production rate per cell and a higher infected
cell rate.130 Contrarily, in another mouse model study, virus
replication rates were not increased, but virus clearance was
y
References 6, 17, 45, 47, 84, 88, 123, 133, 143. delayed.173 Studies in mice have associated PB1-F2 with
Janke 417

increased cytokine concentrations in BALF, infiltration of subtypes that have successfully adapted to mammalian host
alveolar walls and lumens with neutrophils and macrophages, species originated from this virus gene pool.163 Recently, an
and deposition of fibrin and necrotic cell debris in alveoli.95 influenza virus expressing a novel 17thHA was detected in
Although the PB1-F2 protein appears to contribute to viru- bats.141 The other genes in this virus also were widely diver-
lence in many studies, the length (full vs truncated) of the pro- gent from other type A viruses. Until this discovery, all influ-
tein and association with virulence are not consistent for all enza viruses endemic in mammalian host species, if traced to
influenza viruses.44,96,121 The protein is expressed at full length their source, appeared to be the result of crossover by avian
(87–90 aa) by most avian influenza viruses, including H5N1 viruses. The genes in influenza viruses that currently circulate
HPAI viruses, and by the 1918 (H1N1), 1957 (H2N2), and in swine populations originated from avian viruses that directly
1968 (H3N2) human pandemic viruses, but in truncated form infected pigs or that adapted first to humans before contributing
in many viruses from mammalian hosts, including the pan- genes to swine viruses.39,77,87 Domestic avian species are
demic 2009 H1N1 virus. Exceptions to these generalizations infected by a larger subset of the subtypes that infect wild birds,
abound.72,143,174 Insertion of genes for the full-length protein but routine infection of humans and swine is limited to only a
into viruses with truncated forms does not necessarily enhance few subtypes. Subtypes H1N1, H3N2, and H1N2 routinely
virulence, calling into question the protein’s role as a virulence infect pigs. Humans are infected with these same subtypes but
factor. Similarly in swine, most but not all swine triple reassor- historically also were infected with an H2 subtype, the 1957
tant H3N2 viruses express full-length PB1-F2 proteins, but in H2N2 pandemic virus (Asian flu). No infection of swine with
vitro and in vivo studies have not consistently associated the an H2 subtype virus had been documented until the report of
presence of these proteins with increased virulence.114,115 the one localized H2N3 outbreak in the Midwestern United
States in 2006.87
NS1 A point of frequent discussion is whether swine must serve
as the intermediate host for infection of humans with avian
The NS1 protein is not a structural part of the virion and is pres- viruses, particularly the pandemic viruses, a question that
ent only within the cytosol of infected cells. The protein contri- goes back to the origin of the classic swine influenza virus and
butes to virulence by interference with the host antiviral the 1918 Spanish flu pandemic. Early studies that reported
response, including IFN-a/b production and expression of other experimental infection of pigs with nearly all avian HA sub-
cytokines such as TNF-a and IL-6.6,132 Both proapoptotic and types60 and identified receptors for both avian and human
antiapoptotic activities have been documented for NS1.46,74,75 viruses in the trachea of swine led to speculation that pigs
Inhibiting apoptosis of cells that produce viral progeny and pro- likely serve as the ‘‘mixing vessel’’ from which pandemic
moting apoptosis of cells of the innate immune system that human influenza viruses arise.50 However, there is no evi-
attack infected cells would be beneficial for the virus.152 dence that pigs were previously infected with the subtypes
In conclusion, influenza viruses evaluated under a variety of responsible for the 1918, 1957, and 1968 pandemics but rather
experimental protocols do appear to express a range of virulence that, with the exception of the 1918 virus, the pandemic
or pathogenic capabilities. Identification of specific virulence viruses were reassortants of avian viruses and the preexisting
determinants has proven to be difficult because of polygenic viruses circulating in human populations. Recent receptor
contributions to pathogenicity. The polymerase proteins effect studies on the respiratory tracts of humans and swine have
alterations in virulence through misdirection of host cell protein revealed very similar distribution patterns, suggesting that
synthetic processes to viral protein production. Viral proteins avian viruses could just as readily infect the human respira-
NS1 and PB1-F2 contribute to virulence through inhibition in tory tract as the swine respiratory tract, negating the need of
innate immune responses of the host. The roles of HA and NA the pig as an intermediate host.142,147
proteins as virulence attributes in the infection of mammalian Avian and human viruses do not readily infect swine,77 but
species are less clear, but balanced function of these 2 proteins infection of pigs with intact avian and human viruses has been
is necessary for efficient virus multiplication. Thus, not all reported.77,85 The presence of avian- and human-origin genes
HA-NA pairings that emerge from reassortment will likely result in the triple reassortants that comprise most of the influenza
in viruses of equivalent virulence. viruses currently circulating in swine also provides evidence
for such infections.73,157,171 These triple reassortant viruses
with the cassette of internal protein genes from human-,
Cross-Species Infections avian-, and swine-origin viruses appear to have an increased
Although only tangentially related to pathogenesis of infection capability of adding genes from viruses currently circulating
or virulence, infection of swine with viruses originating from in human populations. Since 1998, when the triple reassortants
other species has been a major factor in the evolution of ende- were first identified, the incidence of new reassortant viruses that
mic strains within swine populations. In any discussion of contain genes from human viruses appearing in swine populations
influenza in swine, the question of what to expect from has increased. These new reassortant viruses cause significant
cross-species infection receives consideration. problems for the swine industry by becoming endemic, adding
Wild waterfowl serve as the reservoir of nearly all known to the antigenic diversity of influenza viruses against which pigs
subtypes of influenza A viruses (16 HA, 9 NA), and the few need protection.103
418 Veterinary Pathology 51(2)

Infection of humans with swine viruses also has been doc- lines,48,50,92,105,128,140 and fluorescein isothiocyanate–labeled
umented,z including more than 70 cases since 1958, with 41 inactivated viruses on sections of formalin-fixed, paraffin-
cases in the United States alone from 1990 to 2009. Viruses that embedded tissues.26,150 Information derived through use of the
apparently reassorted in pigs and infected humans have been various methods has tended to be in agreement, but conflicting
documented.13 The pandemic 2009 H1N1 virus, the initial or confusing results are sometimes reported, and initial ‘‘facts’’
source of which is still unknown, is the most recent well- may actually be more complicated.91,106 Early studies tried to
known example.37 Interestingly, in different locations in define the association of receptors with specific cell types
the world, this virus first caused widespread clinical disease in (ciliated, nonciliated, goblet, type I pneumocyte, type II pneu-
the human population and only subsequently was detected in mocyte), with inconsistent results. Later studies revealed that
swine populations (in which it promptly reassorted with endemic viruses infect more than one type of cell, that some cells
swine viruses154). This pandemic virus highlights the possibility express more than one type of receptor,48,68,140 and that after
of ‘‘reverse zoonosis’’ that exists where humans and swine virus entry, cells may cease to express receptors.142 In the in
closely interact.125 Serologic surveys have been conducted that vitro studies, removal of sialic acid residues with neuramini-
suggest people who work with swine are more frequently dase did not prevent infection,38,43,140 and virus infection of
infected with swine viruses than people who do not.40,108 Results cells with few or no recognized receptors has been reported.106
of these surveys should be interpreted with caution because of In 2 studies, virus attachment did not always correlate with effi-
possible partial cross-reactivity in the serologic assays between cient virus replication.20,119 These studies may suggest a core-
human and swine viruses of the same subtype. Thus, the inci- ceptor is necessary for virus entry, or at least caution is
dence of human infection with swine viruses is unknown. warranted against overinterpretation of binding patterns to sia-
Studies have been conducted to determine the factors that lic acids.35 Studies that include discussion of problems with the
favor movement of intact virus across species and reassort- use of lectins48 and some of the discrepancies in these stud-
ment.77,87 Two factors influencing cross-species infection have ies106 have been published.
received the most focus in swine studies: (1) the relationship Results of more recent studies suggest that other factors
between viral HA and the host cell receptors to which the beyond the type of sialic acid linkage also influence attach-
HA attaches and (2) the polymerase gene complex, the viral ment: conformation of the glycan, the structure and length of
proteins responsible for usurping cellular processes for virus the oligosaccharide chain underlying the sialic acid, the density
replication. of receptors on the surface of the cell, the avidity of the binding
between HA and receptor, the balance between HA binding
avidity and neuraminidase activity, and temperature sensitiv-
Hemagglutinin and Host Cell Receptors ity.49,161 Mutations at HA amino acids 226, 228, and 190 influ-
The presence or absence of specific cell receptors dictates which ence host receptor specificity.104
hosts can be successfully infected by a given virus, and the dis- Several recent studies have provided excellent detail on re-
tribution of those receptors in the respiratory tract may influence ceptor distribution in the pig respiratory tract.102,147,150 Although
extent of infection and thus secondarily the severity of disease. early studies identified both a2,3 and a2,6 receptors on tracheal
Gene reassortment that results in emergence of a virus with a epithelial cells, setting the stage for establishing the pig’s reputa-
new combination of genes can occur only in a host species with tion as ‘‘a mixing vessel,’’ the more recent studies indicate that
receptors for the viruses that contributed genes to the new virus. very few swine tracheal cells express a2,3 receptors. Receptor
The molecular structure of the HA selects the type of cells distribution in human and swine respiratory tracts is very similar,
within the host that are infected and thus strongly influences the suggesting that swine are no more likely than humans to be suc-
host range. The HA occurs as a trimer molecule projecting cessfully infected by avian viruses and thus are not a necessary
from the surface of the influenza virion with a receptor pocket intermediate host for avian-to-human infections. The ferret
at the distal end that binds to sialic acids linked in either a2,3 or respiratory tract is also very similar to that of human and swine,
a2,6 conformation to the terminal galactose of oligosaccharide supporting the use of this species as a laboratory model. Contra-
chains on glycoproteins or glycolipids that project from the rily, receptor distribution in the mouse, which is not a natural host
host cell surface.112 Avian viruses attach preferentially to for influenza virus, is quite dissimilar. Very few a2,6 receptors
a2,3 sialic acids, whereas mammalian-adapted viruses prefer are present at any level of the mouse respiratory tract.
to attach to a2,6 sialic acids. In the pig, epithelial cells lining the nasal passages, trachea,
The relative distribution of these receptors in avian and and bronchi almost exclusively express a2,6 receptors; only a
mammalian tissues and the binding of viruses recovered from very few scattered cells with a2,3 receptors were identified
different hosts to these receptors have been studied by a variety in the upper airways in these studies. At the level of the termi-
of techniques: lectin immunohistochemistry on frozen or nal bronchioles, the number of cells expressing a2,3 receptors
formalin-fixed tissue sections, ex vivo biopsy tissues, live begins to increase. At the level of respiratory bronchioles and
cells derived from respiratory tracts or respiratory cell alveoli, both a2,6 and a2,3 receptors can be identified,
although the percentages of each and the types of cells expres-
sing these receptors vary with the study. In summary, a2,6
z
References 1, 63, 71, 77, 100, 127, 159, 167. receptors are present in abundance throughout the porcine
Janke 419

respiratory tract, but a2,3 receptors are limited to the small In investigation of host range, reduced virus replication
bronchioles and alveoli and are of similar or lesser abundance in a second host may not necessarily be the result of dif-
than a2,6 receptors in this portion of the tract. Interestingly, ferences in cell tropism but could involve differing ability
many cells other than the airway epithelial cells also appear to efficiently use the protein synthetic machinery or an
to express sialic acid receptors, including peribronchial muco- incompatibility with other viral proteins. As an example,
sal gland epithelial cells, subepithelial connective tissue cells, in a study that compared cell tropism, replication, and
and endothelial cells.170 cytokine induction by HPAI H5N1, pandemic 2009 H1N1,
The placement of these receptors in various portions of the human seasonal H1N1 and H3N2, LPAI H12N5, and classic
respiratory tract (and other tissues) dictates to a large degree the swine H1N1 viruses in human lung explants, all viruses
distribution of infection and severity of disease in the individ- replicated only in type II pneumocytes, but only the HPAI
ual animal of a given host as well as the likelihood of transmis- H5N1 and seasonal viruses replicated to high titer. The
sion between individuals of the same species and between avian viruses (HPAI and LPAI) stimulated high cytokine
species. Thus, a preponderance of receptors for a virus in the responses, whereas the seasonal viruses and the pandemic
smaller bronchioles and alveoli will predispose a host to devel- 2009 H1N1 stimulated only weak responses. The classic
oping severe disease (pneumonia) rather than bronchitis, but swine virus replicated poorly and stimulated only a weak
transmission of virus between individuals may be limited. Con- cytokine response.165
versely, if most receptors for a particular virus are in the upper Evidence is strongest for a major role of the PB2 polymerase
respiratory tract and larger airways, virus shedding and thus protein in host range.129 One of the most consistent results of
transmission between individuals may be facilitated, but the studies indicates a glutamic acid (E) at amino acid 627 favors
disease induced will be relatively milder (bronchitis). infectivity of avian species, whereas lysine at this position
(627K) favors infectivity of mammalian species.129 Passage
of avian viruses in mammalian hosts will result in selection
Polymerases and Other Viral Proteins of E627K mutants. Another potential determinant of host range
After hemagglutinins and receptors, the polymerases have occurs at amino acid 701 of PB2, with aspartic acid favoring
received the most study directed toward defining determinants avian hosts and asparagine favoring mammalian hosts. In other
of host range. These studies have been conducted in the in vitro studies, most of the viral proteins have been shown to have
systems and in various experimental hosts (mice, primates, fer- some influence on host range.16,58,77,97,104
rets) but infrequently in pigs. As with much of the basic
research on virulence factors, inconsistent results in studies
on host range conducted in different experimental hosts pre- Diagnosis of Influenza Virus Infections in
cludes definitive extrapolation of observations to other host Swine
species until supported by additional evidence. Methods of diagnosis of influenza virus infections have
The techniques that employ viruses generated by reverse improved over the years with change being driven by both the
genetics to investigate virulence also have been used to provide availability of new technologies and the necessity of keeping
information about host range, and results should be interpreted up with changes in the virus. A recent well-referenced review
with caution. A gene substitution or mutation in a given virus article describes the various tests used in the diagnosis of influ-
that results in reduced replication in the same host or in vitro sys- enza virus infection in swine and addresses attributes and ratio-
tem could be interpreted to demonstrate attenuation of the viru- nale behind influenza surveillance systems.25
lence capability of that virus. However, a similar gene change (in
the same virus or in another virus) that results in reduced repli-
cation when inoculated into a different host could be due to
Pig Selection
attenuation of the virulence of the virus or be a reflection of host Diagnosis of influenza virus infection in swine is most effec-
range, that is, a lesser ability to replicate in the second host, tively accomplished by sampling multiple pigs rather than
regardless of the gene change. Particularly in the investigation individual animals. This approach may be critical because,
of the effect of a specific gene change in additional viruses or within a group of affected pigs at any given point in time, not
in other hosts, sufficient control groups must be included to sup- all pigs will be at the same point in the course of infection.
port interpretation of results and conclusions. Herein lies the Sampling multiple animals increases the chances of catching
problem with studies seeking to clarify viral virulence factors some pigs at the peak of virus shedding or with maximal viral
that affect severity of disease in humans since inoculation of titers in lung tissues. Given the short course of infection by
humans with potentially virulent viruses is not morally or ethi- this virus, selection of the right pig for individual sampling
cally justifiable. The use of cell culture systems derived from cannot be emphasized enough. Such pigs have high fevers
humans is as close to the actual host as such studies can get even (105 F) and clear nasal discharge and exhibit lethargy, reluc-
though results may not accurately reflect human infection. Like- tance to move, and dyspnea or hyperpnea. Although cough is
wise, results of studies on swine viruses that are conducted in a clinical hallmark of influenza virus infection, pigs at this
mice or other laboratory animals or in nonswine cell culture sys- stage of disease will already have passed the point of highest
tems may not reflect what happens in the pig. virus load in the lungs.
420 Veterinary Pathology 51(2)

Samples of Choice cytoplasm and nucleus, and in many sections, the nuclear stain-
ing predominates. This probably reflects the fact that during viral
Although samples to be used for diagnostic efforts may involve
morphogenesis, nucleoprotein is formed within the nucleus.55
collection from individual animals, these samples often are
In situ hybridization methods applied to tissue sections also
analyzed in pools. Antemortem sampling involves collection
have been described.57 This technique is not routinely used in
of nasal secretions on synthetic swabs and suspension of the
US veterinary diagnostic laboratories.
fluid in saline or similar physiologic solution. Postmortem sam-
Commercially available antigen-detection enzyme-linked
ples include collection of bronchial swabs or bronchoalveolar
immunosorbent assay (ELISA) kits developed for diagnosis
lavage fluid flushed from the intact respiratory tract and fresh
of human and avian influenza virus infections are used in some
and formalin-fixed portions of lung with characteristic lesions.
laboratories to detect the virus in nasal or bronchial swabs or
bronchoalveolar lavage fluids. These tests can be used to detect
Histopathology endemic swine influenza viruses because the antibodies in the
assays are directed against conserved type A antigens in matrix
Although a variety of assays are available, histopathologic proteins or nucleoproteins.
examination of lung tissue remains a valuable tool in the diagno- Virus isolation can be conducted in embryonated chicken
sis of influenza infection in swine: (1) the acute lesions are eggs or in a number of established cell lines. Before the develop-
nearly pathognomonic for this virus, and (2) lesions induced ment of polymerase chain reaction (PCR) methodologies, virus
by the virus will remain after the virus is gone, although resol- growth in allantoic fluids was monitored by HA tests. Currently,
ving lesions become less definitive. No other commonly encoun- PCR techniques are used to detect and characterize influenza
tered respiratory pathogen in swine induces widespread viruses isolated in eggs or cell cultures. The isolation procedures
necrotizing bronchitis and bronchiolitis. Even severe purulent are more frequently used for recovery of virus for autogenous
bronchopneumonia will not cause similar damage to the airways. vaccine production or for amplification of virus titer to facilitate
Severe porcine circovirus type 2 (PCV2) infections may subtype determination than for routine diagnosis.
result in necrosis and fibrosis of medium to small airways that PCR techniques are currently used routinely for diagnosis
can cause confusion with influenza infection; however, other because large numbers of samples can be handled efficiently
features of that virus infection differ. In PCV2-infected lungs, and because the method is least affected by sample quality.
the damage to airways may be ‘‘one-sided’’ and not involve the Either in separate assays or as a multiplex PCR, genes for the
entire circumference of the cross section, and the fibrosis may conserved nucleoproteins or matrix proteins are detected as a
extend irregularly into the surrounding interstitium. Peri- screening test followed by assays for the HA and NA subtypes.
bronchiolar inflammation in PCV2 infection also tends to be
more histiocytic than lymphocytic.
Resolving influenza infections have lesions that resemble Serology
Mycoplasma hyopneumoniae infection, and additional testing The hemagglutination inhibition (HI) test has served as the
may be required to differentiate these possibilities. Airways are standard serologic assay for the detection of antibody against
lined by tightly packed, somewhat irregular, hyperplastic influenza viruses in swine. ELISAs have been developed, some
columnar epithelial cells and surrounded by prominent lym- of which are available as commercial kits, to remove the sub-
phocytic cuffs. A subtle difference sometimes evident is early jectivity of the HI test and to facilitate testing of large numbers
nodular hyperplasia of bronchial-associated lymphoid tissue of samples. However, because of the variety of reassortant
associated with the medium to larger airways (in addition to the influenza viruses circulating in swine populations and the anti-
lymphocytic cuffing) in the M. hyopneumoniae–infected lungs, genic variation within subtypes, and because of the widespread
whereas the lymphocytic infiltration is relatively evenly dis- use of commercial and autogenous vaccines, diagnosis of influ-
tributed around airways in resolving influenza infections. enza infection through the use of serologic assays has become
much more difficult. The ultimate result is a much reduced
ability to detect antibody against many reassortants.
Methods to Detect Influenza Virus To circumvent this problem due to antigenic variability, a
The direct or indirect fluorescent antibody test using hyperim- blocking ELISA has been developed that will detect antibody
mune serum (polyclonal antibody) to detect virus antigen in fro- against the nucleoprotein (NP), a highly conserved internal pro-
zen lung sections was the primary assay employed in veterinary tein in all type A influenza viruses. If results of the blocking
diagnostic laboratories for many years. The staining techniques ELISA suggest recent influenza infection, additional HI serologic
could also be used on cell culture monolayers used for virus assays can be conducted with a panel of standard virus strains. An
isolation. ELISA also has been developed that will detect antibody against
Immunohistochemistry applied to sections of formalin-fixed the NS1 protein. This protein is not present in intact virions in
tissue is now used more frequently to detect virus in tissue sec- killed vaccines but is produced only in infected cells. Thus, anti-
tions. The preferred antibody is a monoclonal antibody against body against NS1 will be stimulated in infected pigs but not in
the conserved type A nucleoprotein because it will detect influ- vaccinated pigs, an example of a naturally occurring DIVA (Dif-
enza virus of all subtypes.160 Virus antigen is visualized in both ferentiating Infected from Vaccinated Animals) vaccine.
Janke 421

More Than Detection—Virus Characterization large percentage of the pigs in a pen will take a turn at the rope
(S. T. Millman, personal communication, 2012). After approx-
Because of the propensity of influenza viruses for reassortment,
imately 10 to 20 minutes, the rope can be removed and the sal-
mere detection of influenza virus may not provide sufficient
iva wrung out into a container. Contaminating feed and fecal
information for full evaluation of the field situation and manage-
debris are removed by centrifugation and the supernatant pro-
ment decisions. Determination of H and N subtypes is routine in
cessed for virus detection.
many veterinary diagnostic laboratories, but a subtype descrip-
Virus is detected by PCR techniques, although determi-
tion by itself provides little information about the origin of the
nation of subtype may be less successful in some samples.
genes that make up an influenza virus or the possible source
Virus isolation has proven to be more difficult. Substances
of infection. Determining the full or partial nucleic acid
are present in oral fluids that may interfere with detection
sequence of the HA1 portion of the hemagglutinin protein allows
by PCR, neutralize live viruses, or are toxic to cell lines.
preparation of dendograms, which compare the current virus iso-
Studies are being conducted to remove or counteract these
late with well-characterized viruses endemic in swine popula-
substances and improve the efficiency of assays performed
tions, as well as prior isolates from the same operation. If this
on oral fluids.
historic information includes prior isolates from the same site
Oral fluids are not currently used for influenza serology, but
or production unit as well as different segments of the pig flow
testing will likely be available soon. Research studies indicate
in the same operation, this information may suggest whether the
that the commercial avian influenza multispecies ELISA
currently detected infection is due to reappearance of the same
(FlockChek; IDEXX Laboratories, Westbrook, ME) will detect
virus from that site or elsewhere in the operation, or the introduc-
antibody against a variety of influenza viruses infecting pigs,
tion of a new virus from the outside. The first possibilities may
including the 2009 pandemic H1N1.21,110 Antibody can be
suggest a need for changes in management and biosecurity mea-
detected in saliva as early as 5 days PI. Antibody induced by
sures within the operation. The latter possibility may be even
vaccination also can be detected. Because antibody titers in
more critical if pigs will now be exposed to an antigenically dif-
oral secretions are not as high as in serum, some alterations
ferent virus and additional reassortant viruses appear.
in methodology may be necessary for optimum results.
Although methodologies exist for characterization/compar-
Swine influenza has been a recognized respiratory disease in
ison of all genes, characterization efforts on routine cases usu-
pigs for more than 90 years, and over those years, the etiology
ally are limited to the HA gene. An exception would be the
and pathogenesis of infection have been determined and step-
development of PCR assays for the M gene that were used for
wise facilitated by advances in research technologies that allow
surveillance purposes to identify the potential entrance of the
us to probe further into the details of infection. The virus has
2009 H1N1 virus into swine populations.140 Research studies
continued to change, most significantly by the introduction of
in many veterinary diagnostic laboratories focus on character-
genes from influenza viruses from other hosts, to the extent that
ization of other genes in field isolates with the objective of
‘‘influenza A virus infection of swine’’ better describes this dis-
identifying new reassortants or reassortment trends.
ease than ‘‘swine influenza.’’ These changes in the virus, as
well as significant changes in swine production operations,
have compelled veterinary diagnostic laboratories to develop
Oral Fluids improved and more flexible diagnostic methods.
A relatively new approach to diagnosis involves collection of
oral fluids from groups or pens of pigs to detect virus shedding
Declaration of Conflicting Interests
in oral secretions, a method particularly valuable in detecting a
virus with such a short duration of infection in the individual The author(s) declared no potential conflicts of interest with respect to
the research, authorship, and/or publication of this article.
pig.27,110,117,120 More pigs can be sampled easily and with less
expense, without the need to identify individual pigs early in
infection. This method has a very high probability of detecting Funding
virus when only 10% to 20% of the pigs in the group are shed- The author(s) received no financial support for the research, author-
ding virus, and thus this approach can be used to suggest the ship, and/or publication of this article.
cause of current illness. Alternatively, through repeated sam-
pling of a single group of pigs as they move through various
growth stages or through sampling of sequential groups of pigs References
at each stage (or a particular stage), the behavior of a virus 1. Alexander DJ, Brown IH. Recent zoonoses caused by influenza
within a large operation may be better defined. A viruses. Rev Sci Tech. 2000;19(1):197–225.
Oral fluids are collected by hanging absorbent thick cotton 2. Armstrong SM, Wang C, Tigdi J, et al. Influenza infects lung
ropes on the sides of pens. As naturally curious creatures, pigs microvascular endothelium leading to microvascular leak: role
will quickly investigate the new object in their environment by of apoptosis and claudin-5. PLoS One. 2012;7(10):e47323.
chewing on the rope. As social creatures, pigs will join their 3. Balachandran S, Roberts PC, Brown LE, et al. Essential role for
penmates in examining the rope. Within a short period, the rope the dsRNA-dependent protein kinase PKR in innate immunity
will be soaked with saliva. Videotaping has revealed that a to viral infection. Immunity. 2000;13(1):129–141.
422 Veterinary Pathology 51(2)

4. Barbe F, Saelens X, Braeckmans D, et al. Role of IFN-alpha dur- 22. Conenello GM, Tisoncik JR, Rosenzweig E, et al. A single N66S
ing the acute stage of a swine influenza virus infection. Res Vet mutation in the PB1-F2 protein of influenza A virus increases
Sci. 2010;88(1):172–178. virulence by inhibiting the early interferon response in vivo.
5. Barman S, Krylov PS, Fabrizio TP, et al. Pathogenicity and trans- J Virol. 2011;85(2):652–662.
missibility of North American triple reassortant swine influenza 23. Damjanovic D, Small CL, Jeyananthan M, et al. Immunopathol-
A viruses in ferrets. PLoS Pathog. 2012;8(7):e1002791. ogy in influenza virus infection: uncoupling the friend from foe.
6. Basler CF, Aguilar PV. Progress in identifying virulence determi- Clin Immunol. 2012;144(1):57–69.
nants of the 1918 H1N1 and the Southeast Asian H5N1 influenza 24. De Vleeschauwer A, Atanasova K, Van Borm S, et al. Compara-
A viruses. Antiviral Res. 2008;79(3):166–178. tive pathogenesis of an avian H5N2 and a swine H1N1 influenza
7. Belser JA, Gustin KM, Maines TR, et al. Pathogenesis and trans- virus in pigs. PLoS One. 2009;4(8):e6662.
mission of triple-reassortant swine H1N1 influenza viruses isolated 25. Detmer S, Gramer M, Goyal S, et al. Diagnostics and surveillance
before the 2009 H1N1 pandemic. J Virol. 2011;85(4):1563–1572. for swine influenza. Curr Top Microbiol Immunol. 2013;370:
8. Brown IH. The epidemiology and evolution of influenza viruses 85–112.
in pigs. Vet Microbiol. 2000;74(1–2):29–46. 26. Detmer SE, Gramer MR, Goyal SM, et al. In vitro characteriza-
9. Brown IH, Done SH, Spencer YI, et al. Pathogenicity of a swine tion of influenza A virus attachment in the upper and lower
influenza H1N1 virus antigenically distinguishable from classical respiratory tracts of pigs. Vet Pathol. 2013;50(4):648–658.
and European strains. Vet Rec. 1993;132(24):598–602. 27. Detmer SE, Patnayak DP, Jiang Y, et al. Detection of influenza A
10. Brown IH, Harris PA, McCauley JW, et al. Multiple genetic reas- virus in porcine oral fluid samples. J Vet Diagn Invest. 2011;
sortment of avian and human influenza A viruses in European 23(2):241–247.
pigs, resulting in the emergence of an H1N2 virus of novel geno- 28. Feldmann A, Schafer MK, Garten W, et al. Targeted infection of
type. J Gen Virol. 1998;79(pt 12):2947–2955. endothelial cells by avian influenza virus A/FPV/Rostock/34
11. Bruder D, Srikiatkhachorn A, Enelow RI. Cellular immunity and (H7N1) in chicken embryos. J Virol. 2000;74(17):8018–8027.
lung injury in respiratory virus infection. Viral Immunol. 2006; 29. Ferrari M, Borghetti P, Foni E, et al. Pathogenesis and subsequent
19(2):147–155. cross-protection of influenza virus infection in pigs sustained by
12. Bussey KA, Bousse TL, Desmet EA, et al. PB2 residue 271 plays an H1N2 strain. Zoonoses Public Health. 2010;57(4):273–280.
a key role in enhanced polymerase activity of influenza A viruses 30. Gabriel G, Herwig A, Klenk H-D. Interaction of polymerase sub-
in mammalian host cells. J Virol. 2010;84(9):4395–4406. unit PB2 and NP with importin-a1 is a determinant of host range
13. Castrucci MR, Donatelli I, Sidoli L, et al. Genetic reassort- of influenza A virus. PLoS Pathog. 2008;4(2):e11.
ment between avian and human influenza A viruses in Italian 31. Garcia-Sastre A, Biron CA. Type 1 interferons and the virus-host
pigs. Virology. 1993;193(1):503–506. relationship: a lesson in detente. Science. 2006;312(5775):879–882.
14. Charley B, Riffault S, Van Reeth K. Porcine innate and adaptative 32. Gauger PC, Vincent AL, Loving CL, et al. Kinetics of lung lesion
immune responses to influenza and coronavirus infections. Ann N development and pro-inflammatory cytokine response in pigs
Y Acad Sci. 2006;1081:130–136. with vaccine-associated enhanced respiratory disease induced
15. Charoenvisal N, Keawcharoen J, Sreta D, et al. Experimental by challenge with pandemic (2009) A/H1N1 influenza virus. Vet
infection with a Thai reassortant swine influenza virus of pan- Pathol. 2012;49(6):900–912.
demic H1N1 origin induced disease. Virol J. 2013;10:88. 33. Gauger PC, Vincent AL, Loving CL, et al. Enhanced pneumonia
16. Chen GW, Chang SC, Mok CK, et al. Genomic signatures of and disease in pigs vaccinated with an inactivated human-like
human versus avian influenza A viruses. Emerg Infect Dis. (delta-cluster) H1N2 vaccine and challenged with pandemic
2006;12(9):1353–1360. 2009 H1N1 influenza virus. Vaccine. 2011;29(15):2712–2719.
17. Chen L-M, Davis CT, Zhou H, et al. Genetic compatibility and 34. Gautam M. Endothelial cells as regulators of cytokine storms dur-
virulence of reassortants derived from contemporary avian ing influenza infection. Thorax. 2011;67(7):617.
H5N1 and human H3N2 influenza A viruses. PLoS Pathog. 35. Ge S, Wang Z. An overview of influenza A virus receptors. Crit
2008;4(5):e1000072. Rev Microbiol. 37(2):157–165.
18. Chen W, Calvo PA, Malide D, et al. A novel influenza A virus 36. Geiler J, Michaelis M, Sithisarn P, et al. Comparison of pro-
mitochondrial protein that induces cell death. Nat Med. 2001; inflammatory cytokine expression and cellular signal transduction
7(12):1306–1312. in human macrophages infected with different influenza A
19. Chen Y, Zhang J, Qiao C, et al. Co-circulation of pandemic 2009 viruses. Med Microbiol Immunol. 2011;200(1):53–60.
H1N1, classical swine H1N1 and avian-like swine H1N1 influenza 37. Girard MP, Tam JS, Assossou OM, et al. The 2009 A (H1N1) influ-
viruses in pigs in China. Infect Genet Evol. 2013;13:331–338. enza virus pandemic: a review. Vaccine. 2010;28(31):4895–4902.
20. Chu VC, Whittaker GR. Influenza virus entry and infection 38. Glaser L, Conenello G, Paulson J, et al. Effective replication of
require host cell N-linked glycoprotein. Proc Natl Acad Sci human influenza viruses in mice lacking a major alpha 2,6 sialyl-
U S A. 2004;101(52):18153–18158. transferase. Virus Res. 2007;126(1–2):9–18.
21. Ciacci-Zanella JR, Vincent AL, Prickett JR, et al. Detection of anti– 39. Gorman OT, Bean WJ, Kawaoka Y, et al. Evolution of influenza
influenza A nucleoprotein antibodies in pigs using a commercial A virus nucleoprotein genes: implications for the origins of H1N1
influenza epitope-blocking enzyme-linked immunosorbent assay human and classical swine viruses. J Virol. 1991;65(7):
developed for avian species. J Vet Diagn Invest. 2010;22(1):3–9. 3704–3714.
Janke 423

40. Gray GC, McCarthy T, Capuano AW, et al. Swine workers and 58. Karasin AI, West K, Carman S, et al. Characterization of avian
swine influenza virus infections. Emerg Infect Dis. 2007;13(12): H3N3 and H1N1 influenza A viruses isolated from pigs in
1871–1878. Canada. J Clin Microbiol. 2004;42(9):4349–4354.
41. Guan ZH, Zhang ML, Hou PL, et al. Identification of cellular pro- 59. Khatri M, Dwivedi V, Krakowka S, et al. Swine influenza H1N1
teins interacting with influenza A virus PB1-F2 protein. Acta virus induces acute inflammatory immune responses in pig lungs:
Virol. 2012;56(3):199–207. a potential animal model for human H1N1 influenza virus.
42. Guarner J, Falcon-Escobedo R. Comparison of the pathology J Virol. 2010;84(21):11210–11218.
caused by H1N1, H5N1, and H3N2 influenza viruses. Arch Med 60. Kida H, Ito T, Yasuda J, et al. Potential for transmission of avian
Res. 2009;40(8):655–661. influenza viruses to pigs. J Gen Virol. 1994;75(pt 9):2183–2188.
43. Guo Y, Rumschlag-Booms E, Wang J, et al. Analysis of 61. Kim B, Shin JH, Han K, et al. Expression of myeloperoxidase in
hemagglutinin-mediated entry tropism of H5N1 avian influenza. swine influenza virus (SIV)–infected neutrophils in lungs from
Virol J. 2009;6(1):39. pigs experimentally infected with SIV subtype H1N2. Vet Res
44. Hai R, Schmolke M, Varga ZT, et al. PB1-F2 expression Commun. 2011;35(7):469–475.
by the 2009 pandemic H1N1 influenza virus has minimal 62. Kim HM, Lee YW, Lee KJ, et al. Alveolar macrophages are indis-
impact on virulence in animal models. J Virol. 2010;84(9): pensable for controlling influenza viruses in lungs of pigs. J Virol.
4442–4450. 2008;82(9):4265–4274.
45. Herfst S, Chutinimitkul S, Ye J, et al. Introduction of virulence 63. Kitikoon P, Vincent AL, Gauger PC, et al. Pathogenicity and
markers in PB2 of pandemic swine-origin influenza virus does not transmission in pigs of the novel A(H3N2)v influenza virus iso-
result in enhanced virulence or transmission. J Virol. 2010;84(8): lated from humans and characterization of swine H3N2 viruses
3752–3758. isolated in 2010–2011. J Virol. 2012;86(12):6804–6814.
46. Herold S, Ludwig S, Pleschka S, et al. Apoptosis signaling in 64. Kitikoon P, Vincent AL, Janke BH, et al. Swine influenza matrix
influenza virus propagation, innate host defense, and lung injury. 2 (M2) protein contributes to protection against infection with dif-
J Leukoc Biol. 2012;92(1):75–82. ferent H1 swine influenza virus (SIV) isolates. Vaccine. 2009;
47. Hsieh EF, Lin SJ, Mok CK, et al. Altered pathogenicity for seaso- 28(2):523–531.
nal influenza virus by single reassortment of the RNP genes 65. Klenk HD. Infection of the endothelium by influenza viruses.
derived from the 2009 pandemic influenza virus. J Infect Dis. Thromb Haemost. 2005;94(2):262–265.
2011;204(6):864–872. 66. Kobasa D, Takada A, Shinya K, et al. Enhanced virulence of
48. Ibricevic A. Influenza virus receptor specificity and cell tropism influenza A viruses with the haemagglutinin of the 1918 pan-
in mouse and human airway epithelial cells. J Virol. 2006; demic virus. Nature. 2004;431(7009):703–707.
80(15):7469. 67. Koen J. A practical method for field diagnosis of swine disease.
49. Imai M, Kawaoka Y. The role of receptor binding specificity in Am J Vet Med. 1919;14:468–470.
interspecies transmission of influenza viruses. Curr Opin Virol. 68. Kogure T, Suzuki T, Takahashi T, et al. Human trachea primary
2012;2(2):160–167. epithelial cells express both sialyl(a2-3)Gal receptor for human
50. Ito T, Couceiro JN, Kelm S, et al. Molecular basis for the gener- parainfluenza virus type 1 and avian influenza viruses, and sia-
ation in pigs of influenza A viruses with pandemic potential. lyl(a2,6)Gal receptor for human influenza viruses. Glycoconju-
J Virol. 1998;72(9):7367–7373. gate J. 2006;23(1–2):101–106.
51. Ivan FX, Tan KS, Phoon MC, et al. Neutrophils infected with 69. Kolesnikova L, Heck S, Matrosovich T, et al. Influenza virus bud-
highly virulent influenza H3N2 virus exhibit augmented early cell ding from the tips of cellular microvilli in differentiated human
death and rapid induction of type I interferon signaling pathways airway epithelial cells. J Gen Virol. 2013;94(pt 5):971–976.
[published online November 27, 2012]. Genomics. doi:10.1016/j. 70. Korteweg C, Gu J. Pathology, molecular biology, and pathogen-
ygeno.2012.11.008. esis of avian influenza A (H5N1) infection in humans. Am J
52. Janke BH. Classic swine influenza. Large Anim Pract. 1998;19: Pathol. 2008;172(5):1155–1170.
24–29. 71. Krueger WS, Gray GC. Swine influenza virus infections in man.
53. Janke BH. Clinicopathological features of swine influenza. Curr Curr Top Microbiol Immunol. 2013;370:201–225.
Top Microbiol Immunol. 2013;370:69–83. 72. Krumbholz A, Philipps A, Oehring H, et al. Current knowledge on
54. Jo SK, Kim HS, Cho SW, et al. Pathogenesis and inflammatory PB1-F2 of influenza A viruses. Med Microbiol Immunol. 2011;
responses of swine H1N2 influenza viruses in pigs. Virus Res. 200(2):69–75.
2007;129(1):64–70. 73. Kuntz-Simon G, Madec F. Genetic and antigenic evolution of
55. Julkunen I, Sareneva T, Pirhonen J, et al. Molecular pathogenesis swine influenza viruses in Europe and evaluation of their zoonotic
of influenza A virus infection and virus-induced regulation of potential. Zoonoses Public Health. 2009;56(6–7):310–325.
cytokine gene expression. Cytokine Growth Factor Rev. 2001; 74. Lam WY, Tang JW, Yeung AC, et al. Avian influenza virus A/
12(2–3):171–180. HK/483/97(H5N1) NS1 protein induces apoptosis in human air-
56. Jung K, Ha Y, Chae C. Pathogenesis of swine influenza virus subtype way epithelial cells. J Virol. 2008;82(6):2741–2751.
H1N2 infection in pigs. J Comp Pathol. 2005;132(2–3):179–184. 75. Lam WY, Yeung AC, Chan PK. Apoptosis, cytokine and chemo-
57. Jung T, Choi C, Chae C. Localization of swine influenza virus in kine induction by non-structural 1 (NS1) proteins encoded by dif-
naturally infected pigs. Vet Pathol Online. 2002;39(1):10–16. ferent influenza subtypes. Virol J. 2011;8:554.
424 Veterinary Pathology 51(2)

76. Landolt GA, Karasin AI, Phillips L, et al. Comparison of the 94. Mazur I, Anhlan D, Mitzner D, et al. The proapoptotic influenza A
pathogenesis of two genetically different H3N2 influenza A virus protein PB1-F2 regulates viral polymerase activity by interac-
viruses in pigs. J Clin Microbiol. 2003;41(5):1936–1941. tion with the PB1 protein. Cell Microbiol. 2008;10(5):1140–1152.
77. Landolt GA, Olsen CW. Up to new tricks—a review of cross- 95. McAuley JL, Chipuk JE, Boyd KL, et al. PB1-F2 proteins from
species transmission of influenza A viruses. Anim Health Res Rev. H5N1 and 20 century pandemic influenza viruses cause immu-
2007;8(1):1–21. nopathology. PLoS Pathog. 2010;6(7):e1001014.
78. Lange E, Kalthoff D, Blohm U, et al. Pathogenesis and transmis- 96. McAuley JL, Zhang K, McCullers JA. The effects of influenza A
sion of the novel swine-origin influenza virus A/H1N1 after experi- virus PB1-F2 protein on polymerase activity are strain specific
mental infection of pigs. J Gen Virol. 2009;90(pt 9):2119–2123. and do not impact pathogenesis. J Virol. 2010;84(1):558–564.
79. Lawen A. Apoptosis-an introduction. Bioessays. 2003;25(9): 97. Mehle A, Doudna JA. Adaptive strategies of the influenza virus
888–896. polymerase for replication in humans [published online Decem-
80. Le Goffic R, Leymarie O, Chevalier C, et al. Transcriptomic analysis ber 7, 2009]. Proc Natl Acad Sci U S A. doi:10.1073/pnas.
of host immune and cell death responses associated with the influ- 0911915106.
enza A virus PB1-F2 protein. PLoS Pathog. 2011;7(8):e1002202. 98. Mori I, Komatsu T, Takeuchi K, et al. In vivo induction of apop-
81. Lee DC, Law AH, Hui K, et al. Interferon dysregulation and virus- tosis by influenza virus. J Gen Virol. 1995;76(pt 11):2869–2873.
induced cell death in avian influenza H5N1 virus infections. Hong 99. Morris SJ, Price GE, Barnett JM, et al. Role of neuraminidase in
Kong Med J. 2012;18(suppl 2):12–16. influenza virus–induced apoptosis. J Gen Virol. 1999;80(pt 1):
82. Lipatov AS, Kwon YK, Sarmento LV, et al. Domestic pigs have 137–146.
low susceptibility to H5N1 highly pathogenic avian influenza 100. Myers KP, Olsen CW, Gray GC. Cases of swine influenza in
viruses. PLoS Pathog. 2008;4(7):e1000102. humans: a review of the literature. Clin Infect Dis. 2007;44(8):
83. Liu Q, Qiao C, Marjuki H, et al. Combination of PB2 271A and 1084–1088.
SR polymorphism at positions 590/591 is critical for viral replica- 101. Nayak DP, Twiehaus MJ, Kelley GW, et al. Immunocytologic
tion and virulence of swine influenza virus in cultured cells and in and histopathologic development of experimental swine influ-
vivo. J Virol. 2012;86(2):1233–1237. enza infection in pigs. Am J Vet Res. 1965;26(115):1271–1283.
84. Ma W, Brenner D, Wang Z, et al. The NS segment of an H5N1 102. Nelli RK, Kuchipudi SV, White GA, et al. Comparative distribu-
highly pathogenic avian influenza virus (HPAIV) is sufficient to tion of human and avian type sialic acid influenza receptors in
alter replication efficiency, cell tropism, and host range of an the pig. BMC Vet Res. 2010;6:4.
H7N1 HPAIV. J Virol. 2010;84(4):2122–2133. 103. Nelson MI, Gramer MR, Vincent AL, et al. Global transmission
85. Ma W, Lager KM, Vincent AL, et al. The role of swine in the gen- of influenza viruses from humans to swine. J Gen Virol. 2012;
eration of novel influenza viruses. Zoonoses Public Health. 2009; 93(pt 10):2195–2203.
56(6–7):326–337. 104. Neumann G, Kawaoka Y. Host range restriction and pathogeni-
86. Ma W, Vincent A, Lager K, et al. Identification and characteriza- city in the context of influenza pandemic. Emerg Infect Dis.
tion of a highly virulent triple reassortant H1N1 swine influenza 2006;12(6):881–886.
virus in the United States. Virus Genes. 2010;40(1):28–36. 105. Nicholls JM, Chan MC, Chan WY, et al. Tropism of avian influ-
87. Ma W, Vincent AL, Gramer MR, et al. Identification of H2N3 enza A (H5N1) in the upper and lower respiratory tract. Nat
influenza A viruses from swine in the United States. Proc Natl Med. 2007;13(2):147–149.
Acad Sci U S A. 2007;104(52):20949–20954. 106. Nicholls JM, Chan RW, Russell RJ, et al. Evolving complexities
88. Maines TR, Chen LM, Belser JA, et al. Multiple genes contribute to of influenza virus and its receptors. Trends Microbiol. 2008;16(4):
the virulent phenotype observed in ferrets of an H5N1 influenza virus 149–157.
isolated from Thailand in 2004. Virology. 2011;413(2):226–230. 107. Oldstone MB, Teijaro JR, Walsh KB, et al. Dissecting influenza
89. Marozin S, Gregory V, Cameron K, et al. Antigenic and genetic virus pathogenesis uncovers a novel chemical approach to com-
diversity among swine influenza A H1N1 and H1N2 viruses in bat the infection. Virology. 2013;435(1):92–101.
Europe. J Gen Virol. 2002;83(pt 4):735–745. 108. Olsen CW, Brammer L, Easterday BC, et al. Serologic evidence
90. Massin P, van der Werf S, Naffakh N. Residue 627 of PB2 is a of H1 swine Influenza virus infection in swine farm residents
determinant of cold sensitivity in RNA replication of avian influ- and employees. Emerg Infect Dis. 2002;8(8):814–819.
enza viruses. J Virol. 2001;75(11):5398–5404. 109. Owen RE, Yamada E, Thompson CI, et al. Alterations in recep-
91. Matrosovich M, Stech J, Klenk HD. Influenza receptors, poly- tor binding properties of recent human influenza H3N2 viruses
merase and host range. Rev Sci Tech. 2009;28(1):203–217. are associated with reduced natural killer cell lysis of infected
92. Matrosovich MN, Matrosovich TY, Gray T, et al. Human and cells. J Virol. 2007;81(20):11170–11178.
avian influenza viruses target different cell types in cultures of 110. Panyasing Y, Goodell CK, Wang C, et al. Detection of influenza
human airway epithelium. Proc Natl Acad Sci U S A. 2004; A virus nucleoprotein antibodies in oral fluid specimens from
101(13):4620–4624. pigs infected under experimental conditions using a blocking
93. Matsuoka Y, Swayne DE, Thomas C, et al. Neuraminidase stalk ELISA [published online October 9, 2012]. Transbound Emerg
length and additional glycosylation of the hemagglutinin influ- Dis. doi:10.1111/tbed.12019.
ence the virulence of influenza H5N1 viruses for mice. J Virol. 111. Pappas C, Aguilar PV, Basler CF, et al. Single gene reassortants
2009;83(9):4704–4708. identify a critical role for PB1, HA, and NA in the high virulence
Janke 425

of the 1918 pandemic influenza virus. Proc Natl Acad Sci U S A. 128. Shinya K, Ebina M, Yamada S, et al. Avian flu: influenza virus
2008;105(8):3064–3069. receptors in the human airway. Nature. 2006;440(7083):435–436.
112. Paulson JC. Interactions of animal viruses with cell surface 129. Shope RE.Swine influenza, III: filtration experiments and etiol-
receptors. Receptors. 1985;2:131–219. ogy. J Exp Med. 1931;54(3):373–385.
113. Peiris JS, Hui KP, Yen HL. Host response to influenza virus: 130. Smith AM, Adler FR, McAuley JL, et al. Effect of 1918 PB1-F2
protection versus immunopathology. Curr Opin Immunol. 2010; expression on influenza A virus infection kinetics. PLoS Comput
22(4):475–481. Biol. 2011;7(2):e1001081.
114. Pena L, Vincent AL, Loving CL, et al. Strain-dependent effects 131. Smith W, Andrewes CH, Laidlaw PP. A virus obtained from
of PB1-F2 of triple-reassortant H3N2 influenza viruses in swine. influenza patients. Lancet. 1933;222(5732):66–68.
J Gen Virol. 2012;93(pt 10):2204–2214. 132. Solorzano A, Webby RJ, Lager KM, et al. Mutations in the NS1
115. Pena L, Vincent AL, Loving CL, et al. Restored PB1-F2 in the protein of swine influenza virus impair anti-interferon activity
2009 pandemic H1N1 influenza virus has minimal effects in and confer attenuation in pigs. J Virol. 2005;79(12):7535–7543.
swine. J Virol. 2012;86(10):5523–5532. 133. Song MS. Virulence and genetic compatibility of polymerase
116. Pomorska-Mol M, Markowska-Daniel I, Kwit K. Immune and reassortant viruses derived from the pandemic (H1N1) 2009
acute phase response in pigs experimentally infected with influenza virus and circulating influenza A viruses. J Virol.
H1N2 swine influenza virus. FEMS Immunol Med Microbiol. 2011;85(13):6275.
2012;66(3):334–342. 134. Sreta D, Paphavasit T, Kedkovid R, et al. Pathogenesis of swine
117. Prickett JR, Zimmerman JJ. The development of oral fluid-based influenza virus (Thai Isolates) in weaning pigs. Paper presented
diagnostics and applications in veterinary medicine. Anim at: 15th Congress of the Federation of Asian Veterinary Associa-
Health Res Rev. 2010;11(2):207–216. tions, Fava-Oie Joint Symposium on Emerging Diseases; Octo-
118. Richt JA, Lager KM, Janke BH, et al. Pathogenic and antigenic ber 27–30, 2008; Bangkok, Thailand.
properties of phylogenetically distinct reassortant H3N2 swine 135. Steinhauer DA. Role of hemagglutinin cleavage for the patho-
influenza viruses cocirculating in the United States. J Clin genicity of influenza virus. Virology. 1999;258(1):1–20.
Microbiol. 2003;41(7):3198–3205. 136. Suguitan AL Jr, Matsuoka Y, Lau YF, et al. The multibasic clea-
119. Rimmelzwaan GF, Nieuwkoop NJ, de Mutsert G, et al. Attach- vage site of the hemagglutinin of highly pathogenic A/Vietnam/
ment of infectious influenza A viruses of various subtypes to live 1203/2004 (H5N1) avian influenza virus acts as a virulence fac-
mammalian and avian cells as measured by flow cytometry. tor in a host-specific manner in mammals. J Virol. 2012;86(5):
Virus Res. 2007;129(2):175–181. 2706–2714.
120. Romagosa A, Gramer M, Joo HS, et al. Sensitivity of oral fluids 137. Taubenberger JK, Morens DM. The pathology of influenza virus
for detecting influenza A virus in populations of vaccinated and infections. Annu Rev Pathol. 2008;3:499–522.
non-vaccinated pigs. Influenza Other Respir Viruses. 2012;6(2): 138. Teijaro JR, Walsh KB, Cahalan S, et al. Endothelial cells are
110–118. central orchestrators of cytokine amplification during influenza
121. Schmolke M, Manicassamy B, Pena L, et al. Differential contri- virus infection. Cell. 2011;146(6):980–991.
bution of PB1-F2 to the virulence of highly pathogenic H5N1 139. Thacker EL, Thacker BJ, Janke BH. Interaction between Myco-
influenza A virus in mammalian and avian species. PLoS plasma hyopneumoniae and swine influenza virus. J Clin Micro-
Pathog. 2011;7(8):e1002186. biol. 2001;39(7):2525–2530.
122. Schrauwen EJ, Bestebroer TM, Munster VJ, et al. Insertion of a 140. Thompson CI, Barclay WS, Zambon MC, et al. Infection of
multibasic cleavage site in the haemagglutinin of human influ- human airway epithelium by human and avian strains of influ-
enza H3N2 virus does not increase pathogenicity in ferrets. enza a virus. J Virol. 2006;80(16):8060–8068.
J Gen Virol. 2011;92(pt 6):1410–1415. 141. Tong S, Li Y, Rivailler P, et al. A distinct lineage of influenza A virus
123. Schrauwen EJ, Herfst S, Chutinimitkul S, et al. Possible from bats. Proc Natl Acad Sci U S A. 2012;109(11):4269–4274.
increased pathogenicity of pandemic (H1N1) 2009 influenza 142. Trebbien R, Larsen LE, Viuff BM. Distribution of sialic acid
virus upon reassortment. Emerg Infect Dis. 2011;17(2):200–208. receptors and influenza A virus of avian and swine origin in
124. Schultz-Cherry S, Hinshaw VS. Influenza virus neuraminidase experimentally infected pigs. Virol J. 2011;8:434.
activates latent transforming growth factor beta. J Virol. 1996; 143. Tscherne DM, Garcia-Sastre A. Virulence determinants of pan-
70(12):8624–8629. demic influenza viruses. J Clin Invest. 2011;121(1):6–13.
125. Scotch M, Brownstein JS, Vegso S, et al. Human vs. animal out- 144. Tumpey TM, Garcia-Sastre A, Taubenberger JK, et al. Patho-
breaks of the 2009 swine-origin H1N1 influenza A epidemic. genicity of influenza viruses with genes from the 1918 pandemic
Ecohealth. 2011;8(3):376–380. virus: functional roles of alveolar macrophages and neutrophils
126. Seo SH, Webby R, Webster RG. No apoptotic deaths and differ- in limiting virus replication and mortality in mice. J Virol.
ent levels of inductions of inflammatory cytokines in alveolar 2005;79(23):14933–14944.
macrophages infected with influenza viruses. Virology. 2004; 145. Uiprasertkul M, Kitphati R, Puthavathana P, et al. Apoptosis and
329(2):270–279. pathogenesis of avian influenza A (H5N1) virus in humans.
127. Shinde V, Bridges CB, Uyeki TM, et al. Triple-reassortant swine Emerg Infect Dis. 2007;13(5):708–712.
influenza A (H1) in humans in the United States, 2005–2009. 146. van den Brand JM, Stittelaar KJ, van Amerongen G, et al. Sever-
N Engl J Med. 2009;360(25):2616–2625. ity of pneumonia due to new H1N1 influenza virus in ferrets is
426 Veterinary Pathology 51(2)

intermediate between that due to seasonal H1N1 virus and 162. Wang S, Le TQ, Kurihara N, et al. Influenza virus-cytokine-
highly pathogenic avian influenza H5N1 virus. J Infect Dis. protease cycle in the pathogenesis of vascular hyperpermeability
2010;201(7):993–999. in severe influenza. J Infect Dis. 2010;202(7):991–1001.
147. Van Poucke SG, Nicholls JM, Nauwynck HJ, et al. Replica- 163. Webster RG, Bean WJ, Gorman OT, et al. Evolution and
tion of avian, human and swine influenza viruses in porcine ecology of influenza A viruses. Microbiol Rev. 1992;56(1):
respiratory explants and association with sialic acid distribu- 152–179.
tion. Virol J. 2010;7:38. 164. Weingartl HM, Albrecht RA, Lager KM, et al. Experimental
148. Van Reeth K. In vivo studies on cytokine involvement during infection of pigs with the human 1918 pandemic influenza virus.
acute viral respiratory disease of swine: troublesome but reward- J Virol. 2009;83(9):4287–4296.
ing. Vet Immunol Immunopathol. 2002;87(3):161. 165. Weinheimer VK, Becher A, Tonnies M, et al. Influenza A
149. Van Reeth K, Van Gucht S, Pensaert M. Correlations between viruses target type II pneumocytes in the human lung. J Infect
lung proinflammatory cytokine levels, virus replication, and dis- Dis. 2012;206(11):1685–1694.
ease after swine influenza virus challenge of vaccination- 166. Winkler GC, Cheville NF. Ultrastructural morphometric investi-
immune pigs. Viral Immunol. 2002;15(4):583–594. gation of early lesions in the pulmonary alveolar region of pigs
150. Van Riel D. Human and avian influenza viruses target different during experimental swine influenza infection. Am J Pathol.
cells in the lower respiratory tract of humans and other mam- 1986;122(3):541–552.
mals. Am J Pathol. 2007;171(4):1215. 167. Wong KK, Greenbaum A, Moll ME, et al. Outbreak of influenza
151. Varga ZT, Palese P. The influenza A virus protein PB1-F2: kill- A (H3N2) variant virus infection among attendees of an agricul-
ing two birds with one stone? Virulence. 2011;2(6):542–546. tural fair, Pennsylvania, USA, 2011. Emerg Infect Dis. 2012;
152. Varga ZT, Ramos I, Hai R, et al. The influenza virus protein PB1- 18(12):1937–1944.
F2 inhibits the induction of type I interferon at the level of the 168. Wurzer WJ, Ehrhardt C, Pleschka S, et al. NF-kB–depen-
MAVS adaptor protein. PLoS Pathog. 2011;7(6):e1002067. dent induction of tumor necrosis factor–related apoptosis-
153. Veldhuis Kroeze EJ, van Amerongen G, Dijkshoorn ML, et al. inducing ligand (TRAIL) and Fas/FasL is crucial for effi-
Pulmonary pathology of pandemic influenza A/H1N1 virus cient influenza virus propagation. J Biol Chem. 2004;
(2009)–infected ferrets upon longitudinal evaluation by com- 279(30):30931–30937.
puted tomography. J Gen Virol. 2011;92(pt 8):1854–1858. 169. Yamada H, Chounan R, Higashi Y, et al. Mitochondrial targeting
154. Vijaykrishna D, Poon LL, Zhu HC, et al. Reassortment of pan- sequence of the influenza A virus PB1-F2 protein and its func-
demic H1N1/2009 influenza A virus in swine. Science. 2010; tion in mitochondria. FEBS Lett. 2004;578(3):331–336.
328(5985):1529. 170. Yao L, Korteweg C, Hsueh W, et al. Avian influenza receptor
155. Vincent A, Ma W, Lager K, et al. Characterization of a newly expression in H5N1-infected and noninfected human tissues.
emerged genetic cluster of H1N1 and H1N2 swine influenza FASEB J. 2008;22(3):733–740.
virus in the United States. Virus Genes. 2009;39(2):176–185. 171. Yu H, Hua RH, Zhang Q, et al. Genetic evolution of swine influ-
156. Vincent AL, Lager KM, Faaberg KS, et al. Experimental inocu- enza A (H3N2) viruses in China from 1970 to 2006. J Clin
lation of pigs with pandemic H1N1 2009 virus and HI cross- Microbiol. 2008;46(3):1067–1075.
reactivity with contemporary swine influenza virus antisera. 172. Zamarin D, Garcia-Sastre A, Xiao X, et al. Influenza virus PB1-
Influenza Other Respi Viruses. 2010;4(2):53–60. F2 protein induces cell death through mitochondrial ANT3 and
157. Vincent AL, Lager KM, Janke BH, et al. Failure of protection VDAC1. PLoS Pathog. 2005;1(1):e4.
and enhanced pneumonia with a US H1N2 swine influenza virus 173. Zamarin D, Ortigoza MB, Palese P. Influenza A virus PB1-F2
in pigs vaccinated with an inactivated classical swine H1N1 vac- protein contributes to viral pathogenesis in mice. J Virol.
cine. Vet Microbiol. 2008;126(4):310–323. 2006;80(16):7976–7983.
158. Vincent AL, Lager KM, Ma W, et al. Evaluation of hemaggluti- 174. Zell R, Krumbholz A, Eitner A, et al. Prevalence of PB1-F2 of
nin subtype 1 swine influenza viruses from the United States. Vet influenza A viruses. J Gen Virol. 2007;88(pt 2):536–546.
Microbiol. 2006;118(3–4):212–222. 175. Zhilinskaia IN, Azarenok AA, Il’inskaia EV, et al. Influenza
159. Vincent AL, Swenson SL, Lager KM, et al. Characterization of virus reproduction in the endothelial cells of human blood ves-
an influenza A virus isolated from pigs during an outbreak of sels. Vopr Virusol. 2012;57(2):20–23.
respiratory disease in swine and people during a county fair in 176. Zhirnov OP, Ksenofontov AL, Kuzmina SG, et al. Interaction of
the United States. Vet Microbiol. 2009;137(1–2):51–59. influenza A virus M1 matrix protein with caspases. Biochemistry
160. Vincent LL, Janke BH, Paul PS, et al. A monoclonal-antibody– (Mosc). 2002;67(5):534–539.
based immunohistochemical method for the detection of swine 177. Zhou B, Li Y, Halpin R, et al. PB2 residue 158 is A pathogenic
influenza virus in formalin-fixed, paraffin-embedded tissues. determinant of pandemic H1N1 and H5 influenza A viruses in
J Vet Diagn Invest. 1997;9(2):191–195. mice. J Virol. 2011;85(1):357–365.
161. Viswanathan K, Chandrasekaran A, Srinivasan A, et al. Glycans 178. Zhu W, Zhu Y, Qin K, et al. Mutations in polymerase genes
as receptors for influenza pathogenesis. Glycoconjugate J. 2010; enhanced the virulence of 2009 pandemic H1N1 influenza virus
27(6):561–570. in mice. PLoS One. 2012;7(3):e33383.

También podría gustarte