ArticlePDF AvailableLiterature Review

Pathogenesis of emerging avian influenza viruses in mammals and the host innate immune response

Authors:

Abstract and Figures

Influenza A viruses of avian origin represent an emerging threat to human health as the progenitors of the next influenza pandemic. In recent years, highly pathogenic avian influenza H5N1 viruses have caused unprecedented epizootics on three continents and rare but highly fatal disease among humans exposed to diseased birds. Avian viruses of the H7 and H9 subtypes have also infected humans but generally resulted in far milder disease, yet they too should be considered as possible pandemic threats. Influenza virus infection elicits a complex network of host immune responses that, in uncomplicated influenza, results in effective control of the virus and the development of long-term memory responses. However, fatal avian H5N1 virus infection in both humans and experimental mammalian models is characterized by a high viral load in the respiratory tract, peripheral leukopenia and lymphopenia, a massive infiltration of macrophages into the lung, and dysregulation of cytokine and chemokine responses. This review focuses on avian influenza viruses as a pandemic threat, their induction of host innate immune responses in mammalian species, and the contribution of these responses to the disease process.
Content may be subject to copyright.
Pathogenesis of emerging avian
influenza viruses in mammals and
the host innate immune response
Summary: Influenza A vir uses of avian origin represent an emerging threat
to human health as the progenitors of the next influenza pandemic. In
recent years, highly pathogenic avian influenza H5N1 viruses have caused
unprecedented epizootics on three continents and rare but highly fatal
disease among humans exposed to diseased birds. Avian viruses of the H7
and H9 subtypes have also infected humans but generally resulted in far
milder disease, yet they too should be considered as possible pandemic
threats. Influenza virus infection elicits a complex network of host
immune responses that, in uncomplicated influenza, results in effective
control of the virus and the development of long-term memory responses.
However, fatal avian H5N1 virus infection in both humans and experi-
mental mammalian models is characterized by a high viral load in the
respiratory tract, peripheral leukopenia and lymphopenia, a massive
infiltration of macrophages into the lung, and dysregulation of cytokine
and chemokine responses. This review focuses on avian influenza viruses
as a pandemic threat, their induction of host innate immune responses in
mammalian species, and the contribution of these responses to the disease
process.
Keywords: avian influenza virus, pathogenesis, innate immunity
Introduction
Influenza has long been recognized as a significant public
health problem that presents a considerable economic burden
on society due to both epidemics, local or regional outbreaks,
and pandemics, epidemics on a global scale. Seasonal epi-
demics of influenza cause 4300 000 deaths annually around
the world. In the United States alone, an average of 36 000
influenza-related deaths and over 200 000 hospitalizations
occur annually due to seasonal influenza, primarily among
persons aged 65 years and over (1). In the 20th century, three
novel influenza strains emerged to cause the pandemics of
1918, 1957, and 1968. These emerging viruses differed
substantially in their overall mortality rates and apparent
virulence for humans, the 1918 pandemic being the most
severe with overall estimated numbers of deaths worldwide
ranging from 20 to 100 million (2). Although pandemics
result in excess deaths in people of all ages, a large proportion
Taronna R. Maines
Kristy J. Szretter
Lucy Perrone
Jessica A. Belser
Rick A. Bright
Hui Zeng
Terrence M. Tumpey
Jacqueline M. Katz
Immunological Reviews 2008
Vol. 225: 68–84
Printed in Singapore. All rights reserved
r2008 The Authors
Journal compilation r2008 Blackwell Munksgaard
Immunological Reviews
0105-2896
Authors’ address
Taronna R. Maines
1
, Kristy J. Szretter
,1
, Lucy Perrone
1
, Jessica A. Belser
1
,
Rick A. Bright
w,1
, Hui Zeng
1
, Terrence M. Tumpey
1
, Jacqueline M. Katz
1
1
Influenza Division, National Center for Immunization
and Respiratory Diseases, Centers for Disease Control &
Prevention, Atlanta, GA, USA.
Correspondence to:
Jacqueline Katz
1600 Clifton Road, MS-G16
Atlanta, GA 30333, USA
Tel.: 11 404 639 4966
Fax: 11 404 639 2350
e-mail: jkatz@cdc.gov
Acknowledgement
The findings and conclusions in this report are those of the
authors and do not necessarily represent the views of the
Centers for Disease Control and Prevention.
Present Address: Washington University School of
Medicine St Louis, MO, USA.
w
Present Address: Vaccine Development Global Program,
PATH Washington, DC, USA.
68 r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008
of deaths in the early pandemic periods occurs among those
o65 years of age (3). The unusually high rate of mortality
among younger, healthy adults in 1918–1919, remains an
enigmatic property of this deadliest of influenza viruses.
Whether severe disease and death in younger adults due to
emerging influenza A viruses are a result of inherent virulence
of the virus, a robust or exacerbated host immune response, a
level of protective immunity in older adults, or combination of
these factors remains unknown.
Since late 2003, highly pathogenic avian influenza (HPAI)
H5N1 viruses have caused unprecedented outbreaks in poultry
in over 60 countries, broadened their host range in birds and
mammals, and as of June 2008, have caused over 380 human
H5N1 virus infections with an overall fatality rate of over 60%
(4). The exceptional virulence of this virus in humans,
together with its ongoing genetic evolution in birds, identifies
this emerging influenza virus as a significant pandemic threat.
Nevertheless, two other avian influenza subtypes have emerged
to infect humans that also warrant our attention. Avian H9N2
viruses are known to infect swine and humans, are endemic
among poultry in which they may not cause overt disease, and
in some cases, possess altered receptor-binding properties that
may promote their ability to infect and spread in humans (5,
6). Likewise, contemporary North American H7N2 viruses
recently have been shown to possess altered receptor-binding
properties that may enhance their ability to infect and spread
among humans (7). Here we review the emergence of avian
influenza viruses as a pandemic threat, their induction of host
innate immune responses in mammalian species, and the
contribution of these responses to the disease process.
Influenza virus overview
Influenza viruses belong to the Orthomyxoviridae family and are
divided into three genera, influenzavirus A,influenzavirus B, and
influenzavirus C (8); however, only influenza A and B viruses
cause epidemic human disease. Only influenza A viruses cause
pandemics due to the availability of a diverse reservoir of virus
subtypes that exist in nature, and as such, they are the focus of
this review. Influenza A viruses are subtyped based on their two
surface glycoproteins, hemagglutinin (HA) and neuraminidase
(NA), and currently, 16 known types of HA and 9 known types
of NA have been isolated from aquatic birds, the natural
reservoir for all influenza viruses (9, 10). Various subtypes
infect a number of mammalian species, but stable lineages have
evolved only in humans, pigs, and horses. Mutations accumu-
late in the surface glycoproteins of influenza viruses that enable
the viruses to evade the host immune response. This process,
known as antigenic drift, is the cause of annual epidemics of
influenza and is the reason for yearly updating of annual
vaccines (11). Because influenza viruses have a segmented
genome, they have the unique capability of undergoing
reassortment with other influenza viruses to generate entirely
novel viruses that are antigenically distinct; this process is
called antigenic shift. If a viable virus with a novel HA is
generated from this process that has the ability to cause disease
and spread efficiently among humans that lack immunity to the
novel HA, a pandemic could occur.
Influenza A virus structure
Influenza A viruses have a single-stranded, negative-sense RNA
genome made up of eight gene segments encoding 10–11
proteins (12, 13). Virions are enveloped and have two surface
glycoproteins, HA and NA that are responsible for virus
attachment and release from host cells, respectively, and are
the principal targets of the host antibody response. Each gene
segment is associated with three polymerase proteins (PB2,
PB1, and PA) and a nucleoprotein (NP), which together form
the eight viral ribonucleoprotein (vRNP) complexes in each
virion (14). The polymerase complex functions as an RNA-
dependent RNA polymerase and mediates the nuclear transport
of the vRNPs facilitating viral transcription (15). The recently
identified PB1-F2 protein, encoded in the 11 reading frame of
the PB1 gene segment of a majority of influenza A viruses,
localizes to the mitochondria and is involved in the induction
of apoptosis within host cells (13). PB1 functions in transcrip-
tion initiation and elongation (16), PB2 is involved in acquir-
ing cap structures from cellular mRNAs (17), and PA has been
recently implicated in virus assembly and release (18, 19). The
NP protein is an integral structural component of vRNPs and is
important for viral RNA transcription and assembly (20). The
M gene encodes the M1 matrix protein, which is abundant in
infected cells and forms a matrix on the interior side of the
virion lipid envelope (21). The M2 protein is derived by
alternative splicing of the M gene mRNA, forms ion channels
within the viral envelope, and functions by acidifying the
virion and facilitating virion uncoating (22). The NS gene
encodes NS1, which has multiple functions including obstruc-
tion of the host antiviral response (23, 24), and NS2, also
known as nuclear export protein (NEP), which is expressed by
alternative mRNA splicing and is important for nuclear export
of vRNPs (25).
Infection and replication
In aquatic birds, replication of influenza viruses occurs primar-
ily in the intestinal tract facilitating virus spread among avian
Maines et al Influenza virus pathogenesis and innate immunity
r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008 69
species via fecal–oral transmission (26). Most infections
in aquatic birds are asymptomatic; however, certain viruses
within the H5 and H7 subtype have been associated with severe
disease and mortality in domestic poultry. Avian influenza
viruses are considered to be of either high or low pathogenicity
(HPAI or LPAI) based on their intravenous pathogenicity index
(27). Cleavage of the HA is an essential step during infection
but the amino acids around the HA cleavage site determines the
susceptibility of the HA to host proteases and has been
identified as a determinant of tissue tropism for influenza
viruses (reviewed in 28). LPAI and human influenza viruses
have a cleavage site that is cleaved by host trypsin-like proteases
that limit the virus to tissues of the respiratory tract. However,
HPAI viruses possess additional basic amino acids within the
HA cleavage site that make the HA susceptible to a wide
range of proteases and allows the virus to replicate outside
of the respiratory tract. Sporadic infections with avian
influenza viruses have been reported in several mammalian
species but stable lineages have emerged only in humans,
pigs, horses, and more recently, dogs (29). Furthermore,
only three subtypes have been documented to cause wide-
spread and sustained disease in humans (H1N1, H3N2, and
H2N2) (10).
Influenza virus infection occurs after the virus attaches to
sialic acid (SA)-terminated glycans on the surface of host cells
via the receptor binding domain of the HA surface
glycoprotein. The binding preference of the HA for particular
SA moieties is an important host range determinant. Human-
adapted influenza viruses preferentially bind to a terminal SA
linked to galactose by an a2,6 linkage (a2,6 SA), a major
glycan of human respiratory epithelia, whereas avian influenza
viruses preferentially bind SA in an a2,3 linkage with galactose
(a2,3 SA), which are abundant in the respiratory and intestinal
tracts of aquatic birds (30–33). After attachment, the virus
can enter the cell either by clathrin-mediated or a clathrin- and
caveolin-independent endocytic pathway (34, 35). At low pH
within the endosome, the HA undergoes a conformational
change that results in fusion of the viral and endosomal
lipid membranes releasing the viral RNPs into the cytoplasm
of the host cell (36). Viral RNPs are transported to the nucleus
for vRNA replication and mRNA transcription using cellular
50-methylated cap structures (m
7
GpppX
m
), which are more
readily available in the nucleus because NS1 blocks cellular
mRNA transport to the cytoplasm but viral mRNAs are
transported to the cytoplasm for translation (37). M1, NP,
and NS2 are transported back to the nucleus for translocation of
vRNAs to the cytoplasm for virion assembly (38, 39). HA, NA,
and M2 proteins migrate to the apical cell membrane and are
poised for virion budding (40). The NA is required for release
of budding virions by cleaving the SA receptors.
Influenza pandemics of the 20th century
Although influenza pandemics have likely occurred at varying
intervals in human history, only the pandemics of the 20th
century have been studied in detail. The pandemic H1N1 virus
of 1918 may have been derived from a virus wholly of avian
origin with the accumulation of multiple adaptive mutation
that allowed it to cause disease and spread among humans,
although the absolute origin and adaptive host remain
unknown (41–43). The H2N2 pandemic in 1957 arose when
a circulating human influenza virus acquired the H2, N2, and
PB1 genes from an avian influenza virus, and the H3N2
pandemic in 1968 occurred after a circulating human influenza
virus acquired the H3 and PB1 genes from an avian influenza
virus (44). Although the factors allowing an influenza virus to
acquire pandemic capability are poorly understood, the accu-
mulation of human host-specific adaptive mutations is critical.
Previous pandemic viruses crossed the species barrier after
acquiring mutations that changed the binding preference of the
HA from avian-like, a2,3 SA, to human-like, a2,6 SA (31, 32,
45, 46). Some subtypes of avian influenza viruses have caused
limited human infections, but none have acquired the capacity
for efficient and sustained transmission among humans, a key
property of a pandemic virus.
Human and avian influenza virus infection in humans
Seasonal influenza virus infections can vary in severity from an
asymptomatic infection to a severe febrile illness characterized
by headache, sore throat, cough, runny nose, muscle aches,
fatigue, and in some cases gastrointestinal symptoms, a more
pronounced symptom of children, lasting a week or more
(47). Complications of influenza virus infection arise primarily
in the very young or elderly and can involve the exacerbation of
underlying health conditions such as chronic pulmonary and
cardiopulmonary diseases and can progress to viral or, more
commonly, secondary bacterial pneumonia (1, 48). In primary
influenza virus pneumonia, infection occurs in the alveolar
epithelial lining which undergoes necrosis and desquamation.
Alveolar macrophages are present in large numbers and in early
stages, neutrophils predominate the infiltrating leukocytes,
while in later stages, lymphocytes and plasma cells predomi-
nate (49).
Proinflammatory cytokines including type I interferon (IFN-
ab), interleukin-1a(IL-1a) and b, IL-6, IL-8, and tumor
necrosis factor-a(TNF-a) are detected in the respiratory tract
in the acute phase of influenza virus infection in humans at the
Maines et al Influenza virus pathogenesis and innate immunity
70 r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008
time of peak illness (50). Levels of T-cell regulatory cytokines
IFN-gand IL-10 are also transiently elevated in infected
individuals, while the increased expression of chemokines,
such macrophage inflammatory protein-1a(MIP-1a), MIP-
1b, and monocyte chemotactic protein-1 (MCP-1), is
consistent with their role in the recruitment of lymphocytes
and monocytes to the infected respiratory epithelium (51). In
human volunteer challenge studies, increased levels of the IL-6
and IFN-acorrelate with the magnitude and time course of
virus replication and symptoms, particularly fever, in infected
individuals (50, 52). Although these local cytokine responses
in human influenza have been evaluated experimentally using
challenge strains of modest virulence for obvious ethical
reasons, these studies nevertheless provide insight into the
dynamics of the host innate response in uncomplicated
infections.
Avian influenza viruses of the H7, H9, and H5 subtypes have
been responsible for human infections (Table 1). Historically,
H7 human infections were largely limited to laboratory and
occupational exposures (53–58) but more recently have
resulted from exposure to infected poultry during outbreak
responses. In 2003 in the Netherlands, an outbreak of highly
pathogenic H7N7 virus resulted in 89 reported infections
among veterinarians and individuals involved in poultry
culling operations and a few family members of cullers who
had no direct exposure to infected poultry (59). The majority
of patients presented with conjunctivitis, a typical clinical sign
of H7 human infection, although respiratory illness was seen in
a few individuals, and in one, the disease progressed to a fatal
pneumonia with acute respiratory distress syndrome (60, 61).
In recent years, repeated outbreaks of both HPAI and LPAI have
occurred in Europe and North America that have been
associated with rare infections among humans exposed to
infected poultry, primarily resulting in conjunctivitis or
relatively mild respiratory illness (59, 61–65). Low
pathogenic H9N2 avian influenza viruses have caused a small
number of documented human infections in mainland China
and Hong Kong, SAR, primarily in young children who
experience mild influenza-like illness and fully recovered
(66–68).
HPAI of the H5N1 subtype have caused the most numerous
and severe human infections of any of the avian influenza
viruses. HPAI H5N1 viruses were first recognized to infect and
cause fatal disease in humans in 1997 after Hong Kong live bird
markets experienced widespread outbreaks of disease in
multiple poultry species. The 18 confirmed human cases, six
of them fatal, resulted from exposure to infected birds and/or
the contaminated environment at the live bird markets
(69–71); the outbreak was ended by the slaughter of all
poultry in Hong Kong. Two additional human cases, a father
and son, were reported in Hong Kong, SAR in 2003 (72). The
father eventually succumbed to infection, but the son survived.
Since late 2003, over 380 human infections of H5N1 virus
have been confirmed in 15 countries in Asia, Africa, and
Europe, with about 60% of those being fatal (4). The majority
of human infections have been the result of exposure to H5N1
virus-infected poultry, but occasional family clusters have
provided evidence for limited person-to-person transmission.
The viruses isolated to date from humans are wholly avian in
origin and lack the ability for sustained transmission in
humans. Clusters of H5N1 virus disease among blood relatives
have repeatedly raised the question as to whether there is a host
genetic component that influences the susceptibility and/or
severity of disease in humans, although in many cases,
epidemiologic evidence for similar environmental exposure
among family members cannot be ruled out (73, 74).
In general, patients infected with H5N1 viruses presented
with fever, respiratory symptoms including cough and
shortness of breath (60, 75–77), gastrointestinal symptoms
including diarrhea (78, 79), and in one pediatric case,
neurological complications. Leukopenia and lymphopenia
have been associated with a poor prognosis (4, 79). Severe
cases were characterized by pneumonia, multi-organ failure,
and in some cases acute respiratory distress syndrome and
death (60, 76, 77). The average incubation period is 2–5 days,
and for the fatal cases, death occurred approximately 9 days
Ta b l e 1 . Avian influenza virus subtypes that have caused human infections
Subtype Year Location
Cases Fatalities
H7N7 1996, 2003 UK, NL 90 1
H5N1 1997 HK 18 6
H9N2 1999, 2003, 2007 CN, HK 9 0
H7N2 2002, 2003, 2007 US, UK 6 0
H5N1 2003–2008 HK, CN, TH, VN, KH, ID, AZ, DJ, EG, IQ,TR, LA, MM, NG, PK, BD 367 237
H7N3 2004 CA 2 0
Country codes for the United Kingdom, the Netherlands, Hong Kong Special Administrative Region, China, the United States, Thailand, Vietnam,
Cambodia, Indonesia, Azerbaijan, Djibouti, Egypt, Iraq, Turkey, Laos, Myanmar, Nigeria, Pakistan, Bangladesh, and Canada.
Maines et al Influenza virus pathogenesis and innate immunity
r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008 71
after symptom onset, a more rapid progression to fatal disease
than seen in the first human cases in 1997 (4, 71). In contrast
to the older age for fatal disease with seasonal influenza viruses,
the highest case fatality rate among H5N1-infected patients is
among individuals 10–19 years of age. Although few in
number, post-mortem examinations conducted among H5N1
virus victims have revealed diffuse alveolar damage with
hyaline membrane formation, interstitial lymphocytic
infiltrates, bronchiolitis, pulmonary congestion, and
hemorrhage; alveolar damage with macrophage, neutrophil,
and activated lymphocyte involvement was noted in patients
who died acutely, within 2 weeks of symptom onset. Apoptosis
in alveolar cells and infiltrating leukocytes and lymphocyte
depletion in lymphoid organs have been detected (80).
Hemophagocytosis, a disorder characterized by excessive
activation of mononuclear phagocytosis of erythrocytes,
platelets, and leukocytes, and one which is thought to be
cytokine-driven, has been observed among the limited H5N1
fatal case studies (72, 81–83). Fatal H5N1 virus disease was
associated with high viral load in the pharynx, more
pronounced cytokine dysregulation, and more frequent
detection of viral RNA in plasma compared with non-fatal
H5N1 cases (83). There are several reports of extrapulmonary
detection of viral RNA in intestine or fecal specimens and
cerebrospinal fluid, but more data are needed to confirm
dissemination of H5N1 viruses in humans. Patients with
severe H5N1 virus infections were found to have elevated
serum IFN-g, sIL-2R, IL-6, IFN-inducible protein-10 (IP-10),
and MCP-1 compared with uncomplicated influenza virus
infections (72, 82, 83).
Emerging threat
Human infection with avian influenza viruses remains a rare
occurrence, even for H5N1 viruses. However, the ongoing
endemicity of avian influenza viruses among some domestic
poultry populations provides increasing opportunities for
human exposure and infection. Together with the continuous
genetic evolution of avian influenza viruses in the avian host,
the possibility that one of these novel viruses may adapt to the
humans and result in a pandemic strain is ever-present. Figure 1
depicts how avian influenza virus could acquire pandemic
capabilities by one or more mechanisms. The results of human
Fig. 1. Generation of a pandemic influenza virus. Pandemic influenza A viruses may arise by one of two general mechanisms. One possible
mechanism is through a reassortment event in a mammalian host as postulated for the H2N2 and H3N2 pandemics (155). Given that some circulating
avian influenza viruses are capable of directly infecting multiple mammalian species, including humans, a reassortment event could occur in any of
these mammalian hosts that are simultaneously infected with both a human and an avian influenza virus. Alternatively, an avian influenza virus could
acquire pandemic capability by adapting to humans without a reassortment event but with the gradual accumulation of genetic mutations, as is
speculated to have been the case for the 1918 pandemic virus. All previous pandemic strains adapted to humans after acquiring mutations in the HA that
changed the receptor specificity of the virus from an avian-like to a human-like receptor binding preference. Compensatory changes in the NA are likely
also necessary. The precise mutations required to switch HA receptor specificity of the previous pandemic viruses is now known, but even with this
information, the mutations that would confer human-like receptor binding properties on currently circulating avian strains remains incomplete (156).
Maines et al Influenza virus pathogenesis and innate immunity
72 r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008
and avian virus reassortment studies suggest that genetic changes
in addition to those that confer human-like receptor binding are
likely to be required for H5N1 viruses and potentially other
avian strains to acquire pandemic capability (84). Identifying
this constellation of genetic factors as well as a better under-
standing of mechanisms of influenza pathogenesis and transmis-
sion and identification of effective prevention strategies will be
paramount in our preparedness for the next pandemic.
Mammalian models of influenza pathogenesis
Because there is only limited information on the mechanisms
of influenza pathogenesis of emerging avian viruses and the
role of host innate immune response in virulence in humans,
animal models are necessary to identify virus and host deter-
minants of disease. Multiple mammalian models have been
used to investigate avian influenza virus pathogenesis (85–88),
with each animal system contributing valuable information.
Our laboratory has used inbred mice and outbred ferrets as
mammalian models to better understand the virus host rela-
tionship of potential emerging pandemic viruses. Ferrets are
widely recognized to be an excellent small animal model of
influenza, as they are susceptible to human influenza and avian
influenza viruses and exhibit disease signs and a level of disease
severity resembling that seen in humans. Ferrets, like humans,
have a predominance of a2,6 SA receptors on upper respiratory
tract tissue (89, 90). Furthermore, van Riel et al. (91) demon-
strated the H5N1 virus displayed a binding pattern to respira-
tory tract epithelial tissues of ferrets that was most similar to
human tissue, binding primarily to type II pneumocytes,
macrophages, and non-ciliated cuboidal epithelial cells in the
alveoli. Infection of ferrets with seasonal human influenza
H1N1 or H3N2 viruses causes fever and mild respiratory
illness, with the animals recovering fully in 7–10 days (64,
87, 92, 93). In contrast, infection of this host with some HPAI
H5N1 or H7N7 viruses results in a severe and in some cases
fatal disease (87, 94–96).
We observed a notable difference in the severity and kinetics
of disease in ferrets caused by highly virulent H5N1 viruses
isolated from humans in 1997 and those isolated after 2004
(84, 95). H5N1 viruses isolated from humans in 1997
(A/HK/486/97 and A/HK/483/97) caused substantial
weight loss, lethargy, respiratory signs, neurological signs,
and secondary bacterial infections in some animals typically
during the second week of illness and was lethal in
approximately 35% of ferrets. In contrast, H5N1 viruses
isolated from humans during 2004 (e.g. A/Thailand/16/04,
A/Vietnam/1203/04) caused a disease characterized by
extreme weight loss, more severe lethargy, transient
lymphopenia, and respiratory signs and were lethal in 100%
of ferrets within the first week of infection. These results
recapitulated the contracted period of severe illness and
overall higher rate of mortality observed with more recent
human H5N1 virus infections (97) compared with those
documented in 1997 and confirmed that the ferret is an
appropriate model for studies of avian influenza virus
pathogenicity in mammals. However, a current limitation of
this model is the lack of ferret-specific immunological reagents
and genomic sequence information, which has hindered the
analysis of the innate immune response in this model. Recently,
a panel of ferret cytokine genes was cloned and sequenced and
a semi-quantitative real time polymerase chain reaction (PCR)
assay was established (98) to identify immune correlates of
disease severity in animals infected with seasonal human
influenza viruses (93). Cytokine mRNA levels were measured
in nasal washes collected from ferrets during the first 4 days
after intranasal infection. A rapid and strong induction of
IFN-a, IFN-g, and TNF-awas observed in nasal washes of
ferrets with mild disease, while ferrets with severe disease,
characterized by increased lung pathology and delayed viral
clearance compared with mild disease, had a reduced level of
IFN-aand expression of IFN-gand TNF-awas delayed. IL-6
was only detected in ferrets exhibiting severe disease, whereas
IL-8 expression was detected in ferrets with mild disease. These
results suggest less virulent influenza viruses elicit a stronger
innate immune response in ferrets, whereas infection with
more virulent strains led to a weaker host response. As more
ferret gene sequence information and ferret-specific antibodies
become available (99, 100), additional immunological
questions regarding host response to influenza virus infection
can be addressed. In the meantime, the inbred mouse remains
our primary tool for such analyses.
The murine model has been used extensively to identify
virulence determinants of influenza viruses. Although most
human influenza A viruses require prior adaptation in mice
before they will result in a productive infection, avian influenza
viruses will replicate and cause disease in mice without prior
adaptation. This outcome is most likely due, at least in part, to
the predominance of SAa2,3 (receptor preferred by avian
viruses) present in murine respiratory tissues (101). As is seen
in humans, onset of symptoms, lung pathology, and cytokine
production are temporally related to virus replication
(102–105). Differential pathogenicities of H5 avian influenza
viruses have been demonstrated in mice. A disease of low
pathogenicity (LP) is generally characterized by a non-lethal
infection that is restricted to the respiratory tract and is cleared
Maines et al Influenza virus pathogenesis and innate immunity
r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008 73
within a week, whereas a high pathogenicity (HP) phenotype
in mice is characterized by a lethal infection, systemic
replication, and peripheral blood lymphopenia (85, 95,
106–108). Viruses of the HP phenotype were at least 1000-
fold more lethal for mice than their LP counterparts (106, 109).
A similar dichotomy of pathogenicity phenotypes in mice were
observed among viruses of the H7 phenotype (96). Two HPAI
H7N1 viruses isolated from Italy in 2000 were also shown to be
neurovirulent in mice (110). However, H9N2 viruses exhibited
a low virulence phenotype in mice, although they replicated
efficiently and to high titers in the lungs of infected mice (111).
A summary of the relative levels of virulence exhibited in mice
and ferrets compared with humans is presented in Ta b l e 2 . Based
on the overall similarities between the relative virulence of
avian influenza viruses observed in humans and recapitulated in
the mouse model, we felt that the further use of inbred mouse
model for investigation of the role of the innate immune
response in the pathogenesis of these emerging viruses with
pandemic potential was warranted.
Molecular determinants of virulence
Pathogenesis studies in animal models are critical for the
identification of virulence determinants of disease to better
understand the disease caused by influenza viruses observed in
humans. Multiple molecular determinants affecting virus viru-
lence have been identified. In mammalian models, the viru-
lence of H5N1 viruses is not always dictated by the multi-basic
cleavage site of the HA, indicating that additional virulence
determinants are involved (94, 95).
The polymerase complex (PB2, PB1, and PA) has been
identified as a virulence determinant of H5N1 viruses,
although phenotypes vary among animal models. The
presence of a Lys at 627 or an Asn at 701 in the PB2 protein of
H5N1 viruses confers a virulent phenotype in inbreed mice but
not in ferrets (94, 95, 112, 113) and does not always correlate
with severe disease in humans (83). Additional studies have
also implicated the polymerase complex in HPAI virus
virulence and virus adaptation to mammalian hosts (114,
115) and identified efficient polymerase activity as a virulence
marker. The PB1-F2 protein targets alveolar macrophages and is
associated with secondary bacterial infections and virulence in
mice (13, 116–120). The NS1 protein of influenza viruses acts
as a virulence determinant by disrupting the IFN response of
hosts and preventing an antiviral state in infected cells from
being achieved (23, 24). The NS proteins of 1997 H5N1
viruses were shown to upregulate the expression of
proinflammatory cytokines in mice and pigs, directly
contributing to the virulent phenotype observed (121, 122).
HPAI virus virulence is dictated by multiple determinants
encoded on multiple gene segments. Continued characterization
of the molecular basis of influenza virus pathogenicity and the
identification of key host factors in mammals is essential to
identify better ways to prevent disease in humans.
Innate immunity to avian influenza viruses
Cellular infiltration into the H5N1 virus-infected lung
When administered at high infectious dose [100–1000 50%
mouse infectious dose (MID
50
)] both HP and LP H5N1 viruses
Ta b l e 2 . Comparison of mouse and ferret models and human disease outcomes for avian influenza virus subtypes
Subtype Avian Pathotype Case, Outcome Human Disease Mice (LD
50
)
Ferret
(Lethality %) References
H5N1 (1997)
w
HPAI 13/Male Fatal Respiratory, lymphopenia, ARDS 1.6 33 (71, 95)
H5N1 (1997)
z
HPAI 5/Female Survived Respiratory, gastrointestinal, lymphocytosis 47 36 (71, 95, 106)
H5N1 (2004)
HPAI 58/Female Fatal Respiratory, Renal, lymphopenia, ARDS 5.5 0 (95)
H5N1 (2004)
z
HPAI 7/Male Fatal Respiratory, gastrointestinal, lymphpenia, ARDS 1.7 100 (4, 95)
H7N7 (2003)
k
HPAI 57/Male Fatal ARDS 2.5 67 (59, 61, 96, 157)
H7N7 (2003)

HPAI Adult Survived Conjunctivitis 47 0 (59, 61, 96, 158)
H7N2 (2002-3)
ww
LPAI Adult/Male Survived Respiratory 47 0 (96, 159, 160)
H7N3 (2004)
zz
HPAI/LPAI 45/Male Survived Respiratory 47 0 (7, 62, 63, 96)
H9N2 (1999)
‰‰
LPAI 4/Female Survived Respiratory 48 0 (111, 161)
Expressed as the log
10
EID
50
required to give 1 LD
50
.AnLD
50
o3 is considered highly pathogenic.
w
A/HongKong/483/97.
z
A/HongKong/486/97.
A/Thailand/SP83/04.
z
A/Thailand/16/04.
k
A/the Netherlands/219/03.

A/the Netherlands/230/03 and A/the Netherlands/33/03.
ww
A/New York/107/03.
zz
A/Canada/504/04.
‰‰
A/Hong Kong/1073/99.
Maines et al Influenza virus pathogenesis and innate immunity
74 r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008
replicate extensively in the lungs of infected BALB/c mice.
However, compared with the LP virus, the HP virus elicited a
rapid and massive depletion of peripheral lymphocytes (80%
drop in lymphocyte numbers on day 4 post-infection), deple-
tion of CD4
1
and CD8
1
T cell in lung and lymphoid tissue as
well as depletion of CD4CD8 double positive thymocytes on
day 6 post-infection (107). Interestingly, the HP virus repli-
cated to substantial titers in the thymus and thymic involution
was observed (95, 123), suggesting that subsets of T cells may
support replication of HP H5N1 strains in vivo. More recent
studies from this laboratory by Perrone and colleagues (127)
have compared two H5N1 viruses isolated from humans in
2004 (A/Thailand/16/2004 and A/Thailand/SP83/2004),
which exhibited differential virulence in BALB/c mice and
ferrets (95). Using a sub-lethal dose (approximately 1–10
MID
50
), infection of mice permitted characterization of the
phenotype and extended kinetics of leukocyte infiltration into
the infected lung. Using this approach, macrophages and
neutrophils were shown to be the predominant cell types
infiltrating the lung early after infection and peaking around
day 7 post-infection; significantly higher numbers of these two
cell types were detected in HP virus-infected lungs compared
with LP virus infection, which also correlated in this compar-
ison with the reduced lung viral titers achieved by the LP virus
compared with the HP strain. In contrast, no significant
differences in the percentages of dendritic cells (DCs), CD4
1
,
and CD8
1
T cells in the lungs between HP and LP virus
infections were observed.
Proinflammatory cytokine and chemokine response in
H5N1 virus-infected mice
Following primary infection of mice with mouse-adapted
strains of influenza A viruses, multiple proinflammatory cyto-
kines are produced in the lungs, including IL-1b, TNF-a, and
IFN-g(102, 103, 105, 124). The differential pathogenicity
phenotypes exhibited by HPAI H5N1 viruses in mice prompted
us to examine further the virus–host interactions in this system.
We first addressed the level of proinflammatory cytokine and
chemokines in the lungs and brains of mice infected with a
high dose of prototype H5N1 viruses isolated from humans in
1997 that exhibited either a HP or LP phenotype in BALB/c or
C57Bl/6 mice (107, 123). Elevated levels of IL-1b, TNF-a,
IFN-g, IL-6, MIP-1a, MIP-2, and RANTES (regulated upon
activation, normal T-cell expressed, and presumably secreted)
proteins were detected 3–5 days after infection with either HP
or LP virus (107, Szretter et al., unpublished data) (Fig. 2).
However, by day 5–6 post-infection, concentrations
of IL-1b, IFN-g, and MIP-1awere significantly lower in HP
virus-infected mice than in mice infected with the LP virus. In
contrast, levels of IL-6 were higher in the lungs of mice
infected with the HP virus, whereas levels of TNF-awere
comparable for the two viruses. Recently, acute lung injury in
H5N1 virus-infected mice was shown to be associated with
elevated IL-6 expression and mediated by TLR4 (Toll-like
receptor 4)-TRIF (TIR-domain-containing adapter-inducing
IFN-b)-TRAF6 (TNF receptor-associated factor)-NF-kB
(nuclear factor-kB) signaling (125). Because HP viruses but
not LP viruses administered intranasally at lethal doses spread
to the brain, we also assessed the inflammatory cytokine/
chemokine response in this organ. As expected, there was no
significant production of any of the cytokines or chemokines
tested in mice infected with the LP virus. In contrast, mice
infected with HP virus exhibited significantly elevated levels of
IL-1b, TNF-a, IFN-g, IL-6, MIP-2, and MIP-1aby day 6 post-
infection, a time point that immediately preceded the death of
the mice. The local synthesis of proinflammatory cytokines,
such as TNF-aor IL-1, within the brain can lead to anorexia,
weight loss, and death (126), possibly contributing to H5N1
virus pathogenesis in the murine model.
In studies which have used sublethal intranasal infection of
mice with even more virulent H5N1 strains isolated from
humans in 2004, elevated levels of MCP-1, MIP-1a,KC
(murine IL-8), IL-1a, IL-6, and IFN-gwere detected on day 4
post-infection, which coincided with peak lung cellularity. In a
side-by-side comparison, the HP 2004 H5N1 virus strain
consistently induced levels of proinflammatory cytokines and
chemokines that significantly surpassed those induced by the
1918 H1N1 pandemic virus, despite a comparable level of
virus replication (127). Contemporary H5N1 viruses are
approximately 50 times more lethal for BALB/c mice than the
1918 pandemic virus (95, 128). Early and elevated production
of TNF-aand type I IFNs in the lungs of mice is also a notable
feature of infection with a highly virulent H7N7 virus (96).
Role of cytokine and chemokine responses in H5N1 virus
pathogenesis in mice
To more clearly define the role of certain cytokines or
chemokines implicated in recovery from influenza infection
and/or contribution to severity of illness, we used mice with
targeted gene disruptions to investigate the consequences of
signaling by IL-6, IL-1b, TNF-a, and MIP-1a, because differ-
ential expression of these cytokines and chemokine were
observed in the murine H5N1 virus disease model. Using this
approach, we observed that despite the strong lung IL-6
production in response to H5N1 virus infection among wild-
type mice, the disruption of IL-6 expression did not
Maines et al Influenza virus pathogenesis and innate immunity
r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008 75
Fig. 2. Determination of proinflammatory cytokine levels in the lungs and brains of BALB/c mice infected intranasally with 1000 MID
50
of
avian H5N1 viruses isolated from humans and exhibiting a HP (filled bars) or LP (shaded bars) phenotype. Tissues were collected at the indicated
times, and clarified tissue homogenized were assayed for levels of the indicated cytokines or chemokine by ELISA. Bars represent the mean protein
concentration (in pg/ml) SD. Constitutive levels of cytokine/chemokine in normal mice are indicated by the dotted lines.
P0.05, HP versus LP
group.
Maines et al Influenza virus pathogenesis and innate immunity
76 r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008
appreciably affect the kinetics of virus replication and outcome
of infection with HP and LP H5N1 viruses that show different
virulence phenotypes in wildtype mice. This may be due, in
part, to the redundancy of the biological effects of IL-6 in
mammalian systems. Nevertheless, these findings were consis-
tent with those of Kozak et al. (129), who observed no
significant difference in weight loss between IL-6-deficient
and wildtype mice infected with mouse-adapted influenza A
virus. Similarly, no apparent role for MIP-1achemokine
expression in the H5N1 virus-induced disease process was
identified. Previous studies with mouse-adapted human influ-
enza viruses suggested that MIP-1acontributed to the severity
of the lung inflammatory response and pathology, and viral
clearance, and may contribute to recruitment of virus-specific
CD8
1
T cells to the infected lung (130, 131). Our results
suggest that MIP-1aexpression is not critical for virus recovery
from LP H5N1 virus infection, but it remains possible that in
the MIP-1a-deficient mouse, other chemokines with over-
lapping functions may act in a compensatory manner. In
studies where signaling through multiple chemokines was
assessed using mice deficient in the chemokine receptor
CCR5 (receptor for MIP-1a, MIP-1ba, and RANTES), an
earlier, robust macrophage infiltration into the influenza
virus-infected lung was associated with increased mortality
(132). In contrast, mice deficient in the chemokine receptor
CCR2 (receptor for MCP-1a) demonstrated delayed macro-
phage recruitment and increased survival (132). These results
highlight the overall consequences for the outcome of influen-
za infection when the balance of the infiltrating monocyte/
macrophage population is altered.
In lethal high dose infections with the HP H5N1 virus in
mice, we observed previously a significant reduction in IL-1b
production in the lung at later times during infection.
Following infection with the LP H5N1 virus, IL-1R-deficient
mice lost significantly more weight, exhibited delayed
clearance of virus and had a greater proportion of animals
succumb to infection compared with their immunocompetent
counterparts. These results suggest that IL-1 is important for
the induction of effective adaptive immune responses against
H5N1 viruses that aid in viral clearance and that the reduced
IL-1bproduction observed in HP H5N1-infected wildtype
mice may contribute to rapid and lethal disease outcome.
Other studies with mouse-adapted influenza virus have shown
a more modest role for IL-1bin virus clearance and recovery,
with diminished or altered antigen-specific T-cell responses
compared with wildtype mice (124, 133).
Both HP and LP H5N1 viruses induced elevated amounts of
TNF-afollowing productive replication of virus in the lungs of
mice. HP H5N1 virus-infected mice that lacked the TNF
receptor1 (TNFR1) exhibited a significant delay in weight
loss, a marker of morbidity, but only a modest delay in time to
death and no effect on virus replication or clearance, lung
histopathology or systemic spread. Similar results were
obtained when TNF-awas neutralized by treatment with anti-
TNF-aneutralizing antibody. An interesting finding in TNFR1
deficient mice infected with HP H5N1 virus was that thymic
involution, observed in wildtype BALB/c and C57Bl/6x129
mice, was substantially diminished, and the loss of total
thymocytes was reduced in TNFR1-deficient mice compared
with wildtype control mice. Because TNF-aand related TNF-
superfamily members including TNF-related apoptosis-
inducing ligand (TRAIL) are known to mediate apoptosis
of T cells, and in particular, thymocytes (134, 135), our
results suggest that the thymic depletion observed in immuno-
competent H5N1 virus-infected mice is primarily due to the
production of increased levels of TNF-ain response to H5N1
infection, rather than a direct cytolytic effect of the virus itself.
In summary, these results suggest that TNF-amay contribute to
early H5N1 virus disease severity, whereas IL-1 may play a role
in viral clearance late in H5N1 virus infection in the murine
model.
Control of H5N1 virus infection in mice by type I interferon
Type I interferons play a critical role in innate resistance to
influenza virus infection and the induction of adaptive immu-
nity effector responses. Viral RNA products generated during
infection are recognized by Toll-like receptors (TLRs) and RIG-
I like receptors (RLR) to initiate the interferon response.
Airway epithelial cells recognize the double-strand RNA and/
or 50-triphosphate ssRNA via retinoic acid-inducible gene I
(RIG-I), a cytosolic RNA helicase, resulting in production of
type-I IFN through an adapter protein IPS-1 (136). The
interferon signaling can also be initiated by engagement of
TLR3 to dsRNA in airway epithelial cells (137). In plasmacy-
toid DCs, ssRNA is recognized by TLR7 and its adaptor MyD88,
triggering type I interferon response (138). Production of type
I interferon leads to the induction of interferon-stimulated
genes (ISGs) with antiviral activities mediated through the JAK-
STAT signaling pathway. Evasion of the host innate immunity,
including the type I interferon response, has been proposed as
a mechanism of virulence of avian H5N1 viruses in mammals
including humans. Influenza virus NS1 protein inhibits the
RIG-I induction of interferon-b(139, 140). Avian H5N1
viruses induced elevated amounts of IFN-ab following pro-
ductive replication of the virus in the lungs of inbred mice
(K. J. Szretter et al. unpublished data). Although inbred mouse
Maines et al Influenza virus pathogenesis and innate immunity
r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008 77
strains lack a functional Mx1 protein, an ISG product which
confers resistance to orthomyxoviruses, other interferon-in-
duced antiviral mediators are expressed which can relay the
effects of the type I interferon response to influenza infection in
these model systems. IFN-abR-deficient mice infected with
either HP or LP H5N1 virus had a more rapid time-to-death
and increased viral replication in extrapulmonary organs as
compared with immunocompetent mice (Szretter et al., un-
published data). Furthermore, H5N1/1997 and H5N1/2004
viruses were found to be sensitive to the antiviral effects of
interferon in vitro in that replication of either HP or LP virus was
significantly reduced in murine lung epithelial cells (LA-4)
pretreated with either IFN-aor IFN-b. Therefore, in the mouse
model in which avian H5N1 viruses exhibit either high or low
virulence, the type I interferon response contributes to early
containment of H5N1 virus in vivo and control of replication in
vitro. These results are consistent with studies using a virulent,
mouse-adapted virus, in which mice that are unable to respond
to IFN-ab experienced greater mortality and virus dissemina-
tion (141). These results further highlight the fact that the
outcome of influenza infection depends not only on the dose
and relative virulence of the virus, but also on host-dependent
factors.
Cytokine and chemokine expression in human cells in
response to avian influenza virus infection
Because innate immune responses are inherently host specific,
it is important to assess the relative induction of cytokine and
chemokines by avian influenza viruses in human primary cell
cultures and/or cell lines in order to establish correlating
phenotypes related to the virulence of these viruses in vivo.
Identifying signature phenotypes of virulence in in vitro systems
may be a means with which to rapidly assess the potential
virulence of emerging influenza viruses for humans, as well as
improve our understanding of the contribution of human
genetics to the severity of influenza disease in general. In
human primary alveolar (type II pneumocytes) and bronchial
epithelial cells (NHBE), H5N1 viruses from 1997 and 2004
induced higher levels of IP-10, IFN-b, RANTES, and IL-6
mRNA expression compared with a human H1N1 virus
(142), although all viruses replicated to comparable levels in
these two cell types. Production of IP-10, IL-6, and RANTES
protein was also significantly higher in H5N1 virus-infected
cultures versus human H1N1 virus-infected cultures, but
production of IFN-bwas below detectable limits for all viruses
up to 24 h post-infection. Interestingly, the 2004 H5N1 viruses
were more potent inducers of IP-10 than the1997 H5N1 virus;
high levels of IP-10, a macrophage chemoattractant, were also
detected in humans with severe or fatal H5N1 virus disease
(72, 83). Because primary human cell cultures may vary
considerably in terms of SA expression and support for virus
replication, our laboratory has used polarized human bronchial
epithelial (Calu-3) cell cultures, which express SA receptors
recognized by both human and avian viruses, for the relative
comparison of cytokine induction by human and avian influ-
enza viruses. In studies focusing on the type I interferon
response, a highly virulent 2004 H5N1 virus was found to
trigger a delayed and weaker IFN-bresponse, weaker JAK-STAT
pathway activation, and ISGs induction compared with a
contemporary human H3N2 influenza virus (143). In particu-
lar, comparison of H5N1 viruses which exhibited HP and LP
pathogenicity phenotypes in mice and ferrets also displayed
differential induction of IFN-b, with the HP strain exhibiting a
stronger dampening of the early IFN-bresposne in Calu-3 cells
but higher levels of TNF-aexpression (143, H. Zeng et al.,
unpublished data). Therefore, an H5N1 virus which exhibits
heightened virulence in mammalian models demonstrated a
better ability to attenuate the host IFN response in human
respiratory epithelial cells, compared with less virulent avian or
human viruses.
In ongoing studies, we are evaluating cytokine expression in
Calu-3 cells in response to HPAI H7 subtype viruses, to
determine whether features of human cytokine induction are
common for highly virulent avian viruses in respiratory
epithelial cultures that support their replication. Interestingly,
we found that HPAI H7 viruses exhibited a profoundly reduced
IFN-bresponse in Calu-3 cells, a response even lower than levels
induced by an HPAI H5N1 virus (Belser et al., unpublished data).
This delayed and/or weakened type I interferon response in
respiratory epithelial cells infected with highly virulent H5 and
H7 viruses suggests that one mechanism of severe disease may
be due to suppression of the early innate immune response in
cells in which primary virus replication takes place. The
pandemic 1918 virus was recently reported to cause a similar
downregulation of the type I interferon response in infected
non-human primates (144). Studies are underway to determine
whether the NS1 protein, a known viral antagonist of the host
interferon response, contributes to this human response to
highly pathogenic avian strains.
Because macrophage infiltration into infected lungs has been
implicated in the immunopathology of H5N1 virus infection
in both humans and a murine model, investigators have used
cultures of primary human monocyte-derived macrophages to
better understand their ability to express cytokines and
chemokines in response to emerging avian influenza viruses.
Cheung et al. (145) demonstrated that H5N1 viruses from
Maines et al Influenza virus pathogenesis and innate immunity
78 r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008
1997 were more potent inducers of IFN-band TNF-agene
expression than were human H3N2 or H1N1 influenza viruses.
In particular, monocyte-derived macrophages infected with a
1997 H5N1 virus produced TNF-ain excess of that stimulated
by LPS treatment. High levels of TNF-awere also induced by a
LPAI H9N2 virus which possessed essentially similar internal
genes to the 1997 H5N1 viruses, including the non-structural
gene which was implicated in contributing to the abundant
TNF-aproduction. Furthermore, infection of monocyte-
derived macrophages with 1997 H5N1 viruses or the LPAI
H9N2 virus, induced high levels of another TNF family
member, TRAIL which contributed to the apoptosis of a co-
cultured Jurkat T cell line (146). Induction of TRAIL by the
avian H7 influenza subtype has also been implicated in efficient
replication of the virus in epithelial cells (147). Human
peripheral blood mononuclear cell-derived macrophages
supported productive and efficient replication of a 2004 HP
H5N1 virus which induced significantly higher levels
of cytokines and chemokines including IL-1b, IL-6, IL-12,
TNF-a, MCP-1, and MIP-1bcompared with a LP strain (127).
High levels of IFN-awere also detected following infection of
human monocyte-derived macrphages with 1997 H5N1
strain, but in comparison, highly pathogenic viruses of the H7
subtype, exhibit reduced and/or suppressed production of
IFN-ain this cell type (Belser et al., unpublished data). In
addition to macrophages, human monocyte-derived and
myeloid DCs are susceptible to infection with H5N1 viruses
(127, 148).
Immunomodulators as interventions for H5N1 virus
infection
Human infections with currently circulating H5N1 viruses are
frequently fatal and intervention strategies are few. Suscept-
ibility to one of two classes of influenza antivirals, the
adamantane M2 channel blockers, varies widely among the
genetically diverse clades and subclades of H5N1 viruses in
circulation (97) and emergence of resistant strains during
treatment with the NA inhibitor, oseltamivir, has been
reported (149). Late initiation of antiviral therapy, due to
delayed presentation of patients or diagnosis of H5N1 virus
infection, further limits efficacy. The general understanding
that H5N1 viruses induce hypercytokinemia in severely ill
patients has raised the question as to whether the use of
modulators of proinflammatory cytokine responses may repre-
sent a beneficial therapeutic strategy. However, early clinical use
of corticosteroids to reduce inflammatory responses in H5N1
virus-infected patients proved to be detrimental for disease
outcome (97). Treatment of mice with a gluticocorticoid
cytokine inhibitors failed to reduce the lethality in mice
infected with a highly virulent 2004 H5N1 (150). In contrast,
gemfibrozil, a commonly prescribed lipid-lowering drug,
which has also been shown to inhibit proinflammatory cyto-
kines, was shown to significantly increase survival in mice
infected with an H2N2 virus, similar to that which caused the
1957 pandemic, but of more modest virulence than the avian
H5N1 viruses (151). Cholesterol-lowering statins, which ex-
hibit non-specific anti-inflammatory activities, have been pro-
posed as another possible class of drugs which could be
efficacious in the treatment for human H5N1 virus infection
(152). Preliminary studies conducted in this laboratory with
two FDA-approved statin drugs have shown no benefit to
survival or have seen no effect, and found only modest delay
in morbidity (as measured by weight loss), and no effect on
survival, viral replication or spread in our murine model of
H5N1 virus disease (K. J. Szretter et al., unpublished data).
Although we observed no improvement in the outcome of
disease, we also noted no enhancement of disease caused by
global suppression of the immune response during H5N1 virus
infection. A recent report suggested that a combination therapy
which included a NA inhibitor, celecoxib, a non-steroidal anti-
inflammatory, and mesalazine, an anti-inflammatory aminosa-
licylate, significantly improved survival in mice infected with
highly virulent H5N1 virus (153). While these studies have
taken a rather non-specific approach to moderating the cyto-
kine response to H5N1 viruses, a more targeted approach,
focusing on the type I interferon response, has yielded
promising results. Prophylactic treatment of mice expressing
the Mx1 gene with human IFN-abefore infection with a highly
lethal dose of H5N1 virus significantly reduced viral titers in
the lung, prevented viral spread and protected mice from fatal
disease (154). It will be interesting to determine whether
therapeutic treatment with interferon is also beneficial in this
model system. Furthermore, these results suggest that addi-
tional knowledge of the contribution of cytokines in the
immunopathology of fatal H5N1 virus disease will provide a
more rational and targeted approach in the development of
intervention strategies that can enhance patient survival.
Concluding remarks
Another influenza pandemic will certainly occur. However,
answers to the critical questions of when, where, and what
virus will emerge continue to evade us. Since 2005, avian
H5N1 viruses have evolved into multiple phylogenetically and
largely geographically distinct groups, but only some of these
Maines et al Influenza virus pathogenesis and innate immunity
r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008 79
have caused human disease (4). This massive geographic,
genetic, and antigenic diversity among avian H5N1 viruses
has heightened concern that these viruses may be poised to
cause the next pandemic and have complicated international
public health efforts to develop effective prevention and
control strategies. Avian viruses of the H7 and H9 subtypes
have also demonstrated changes in receptor binding that
potentially move them one step closer to a pandemic pheno-
type. Concerns that the next pandemic may be a return to the
H2 subtype are also warranted, because approximately two-
thirds of the today’s global population were born after 1967,
the last time an H2N2 virus circulated in humans, and have no
specific B cell immunity to the H2 HA. A better understanding
of the complex interplay between the virus and the host and the
consequences for disease outcome, will help us develop
improved methods to identify rapidly the potential for heigh-
tened virulence of an emerging influenza virus, identify a
possible genetic basis for disease, and develop appropriate
intervention strategies.
The human and animal studies reviewed here suggest that that
severe and fatal H5N1 virus disease is a consequence of highly
efficient replication and preferred tropism, at least in humans, for
type II alveolar epithelial cells and alveolar macrophages in the
lower airways, followed by a dysregulated host inflammatory
response. Whether the high efficiency of replication by H5N1
viruses observed in vivo and in vitro is due to molecular
determinants in polymerase gene(s) that enhance inherent
replication efficiency of the virus, or those in other genes that
may allow the virus to overcome or delay early host control via
the type I interferon response, or a combination of both, requires
further study. Efficient and rapid viral replication in respiratory
epithelial cells and alveolar macrophages would enhance
expression of chemokines and cytokines, in a virus-dependent
manner, and promote cellular infiltration of macrophages,
neutrophils and lymphocytes and their activation in the infected
lung. A second wave of cytokines released from infiltrating,
macrophages undergoing virus-induced apoptosis would further
amplify the inflammatory response, leading to high
concentrations of cytokines in the circulation which may have
destructive, multi-organ consequences. Several lines of evidence
suggestthatlymphocytes,particularlyTcells,aredepletedfrom
respiratory tissues, and peripheral blood in H5N1 virus infection,
possibly due to apoptosis induced by macrophage-derived TNF
family proteins. The role of CD8
1
T cells in release of cytokines
and immunopathology, bears further attention, but their lack of
accumulation at least in the murine H5N1 virus-infected lung, is
consistent with the observed uncontrolled replication of HP
H5N1 viruses. Although multiple studies suggest that
proinflammatory cytokine responses correlate with disease
severity, the use of gene knockout mice failed to place any single
cytokine in a key role in fatal H5N1 virus disease. Strikingly, the
HP H5N1 viruses exhibited greater virulence in mice, and
produced significantly higher levels of cytokines and
chemokines in mouse lungs and human macrophages than the
reconstructed 1918 virus, the so-called ‘mother of all pandemics’
(42). Further studies in mice deficient in multiple cytokines or
cytokine receptors may overcome the redundancies in the
cytokine network and identify more pronounced effects on
disease outcome. Such studies should provide further insight
and a basis for the rational design of intervention strategies that
target specific host responses for the amelioration of severe
influenza disease caused by emerging or remerging viruses.
References
1. Thompson WW, et al. Mortality associated
with influenza and respiratory syncytial
virus in the United States. JAMA
2003;289:179–186.
2. Johnson NP, Mueller J. Updating the
accounts: global mortality of the
1918–1920 ‘‘Spanish’ influenza
pandemic. Bull History Med
2002;76:105–115.
3. Simonsen L, Clarke MJ, Schonberger LB,
Arden NH, Cox NJ, Fukuda K. Pandemic
versus epidemic influenza mortality:
a pattern of changing age distribution.
J Infect Dis 1998;178:53–60.
4. WHO. Cumulative Number of Confirmed
Human Cases of Avian Influenza A/(H5N1)
Reported to WHO. Geneva: WHO, 2008.
5. Matrosovich MN, Krauss S, Webster RG.
H9N2 influenza A viruses from poultry
in Asia have human virus-like receptor
specificity. Virology 2001;281:
156–162.
6. Gambarian AS, Iamnikova SS, L’Vov DK,
Robertson JS, Webster RG, Matrosovich MN.
Differences in receptor specificity between
the influenza A viruses isolated from the
duck, chicken, and human. Molekuliarnaia
Biol 2002;36:542–549.
7. Belser JA, et al. Contemporary North
American influenza H7 viruses possess
human receptor specificity: implications for
virus transmissibility. Proc Natl Acad Sci
USA 2008;105:7558–7563.
8. Cox NJ, Subbarao K. Global epidemiology of
influenza: past and present. Ann Rev Med
2000;51:407–421.
9. Fouchier RA, et al. Characterization of a
novel influenza A virus hemagglutinin sub-
type (H16) obtained from black-headed
gulls. J Virol 2005;79:2814–2822.
10. Webster RG, Bean WJ, Gorman OT, Cham-
bers TM, Kawaoka Y. Evolution and ecology
of influenza A viruses. Microbiol Rev
1992;56:152–179.
11. Wright PF, Webster RG. Orthomyxoviruses.
In: Knipe DM, ed. Fields Virology, 4th edn.
Philadelphia: Lippincott-Raven,
2001:1533–1579.
12. McGeoch D, Fellner P, Newton C. Influenza
virus genome consists of eight distinct RNA
species. Proc Natl Acad Sci USA
1976;73:3045–3049.
13. Chen W, et al. A novel influenza A virus
mitochondrial protein that induces cell
death. Nat Med 2001;7:1306–1312.
14. Hsu MT, Parvin JD, Gupta S, Krystal M,
Palese P. Genomic RNAs of influenza viruses
Maines et al Influenza virus pathogenesis and innate immunity
80 r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008
are held in a circular conformation in vir-
ions and in infected cells by a terminal
panhandle. Proc Natl Acad Sci USA
1987;84:8140–8144.
15. Akkina RK, Chambers TM, Londo DR, Nayak
DP. Intracellular localization of the viral
polymerase proteins in cells infected with
influenza virus and cells expressing PB1
protein from cloned cDNA. J Virol
1987;61:2217–2224.
16. Braam J, Ulmanen I, Krug RM. Molecular
model of a eucaryotic transcription com-
plex: functions and movements of influenza
P proteins during capped RNA-primed
transcription. Cell 1983;34:609–618.
17. Nakagawa Y, et al. The RNA polymerase PB2
subunit is not required for replication of the
influenza virus genome but is involved in
capped mRNA synthesis. J Virol
1995;69:728–733.
18. Lamb RA, Krug RM. Orthomyxoviridae: the
viruses and their replication. In: Knipe DM,
ed. Field’s Virology, 4th edn. Philadelphia:
Lippincott-Raven, 2001:1487–1531.
19. Regan JF, Liang Y, Parslow TG. Defective
assembly of influenza A virus due to a
mutation in the polymerase subunit PA.
J Virol 2006;80:252–261.
20. Baudin F, Bach C, Cusack S, Ruigrok RW.
Structure of influenza virus RNP. I. Influenza
virus nucleoprotein melts secondary struc-
ture in panhandle RNA and exposes the
bases to the solvent. EMBO J 1994;13:
3158–3165.
21. Fujiyoshi Y, Kume NP, Sakata K, Sato SB. Fine
structure of influenza A virus observed by
electron cryo-microscopy. EMBO J
1994;13:318–326.
22. Pinto LH, Lamb RA. The M2 proton chan-
nels of influenza A and B viruses. J Biol
Chem 2006;281:8997–9000.
23. Garcia-Sastre A. Inhibition of interferon-
mediated antiviral responses by influenza A
viruses and other negative-strand RNA
viruses. Virology 2001;279:375–384.
24. Krug RM, Yuan W, Noah DL, Latham AG.
Intracellular warfare between human influ-
enza viruses and human cells: the roles of
the viral NS1 protein. Virology 2003;309:
181–189.
25. Inglis SC, Barrett T, Brown CM, Almond JW.
The smallest genome RNA segment of in-
fluenza virus contains two genes that may
overlap. Proc Natl Acad Sci USA 1979;76:
3790–3794.
26. Webster RG, Yakhno M, Hinshaw VS, Bean
WJ, Murti KG. Intestinal influenza: replica-
tion and characterization of influenza
viruses in ducks. Virology 1978;84:
268–278.
27. WHO Manual on Animal Influenza
Diagnosis and Surveillance. Geneva: WHO,
2004:62–63.
28. Horimoto T, Kawaoka Y. Pandemic threat
posed by avian influenza A viruses. Clin
Microbiol Rev 2001;14:129–149.
29. Payungporn S, et al. Influenza A virus
(H3N8) in dogs with respiratory disease,
Florida. Emerg Infect Dis 2008;14:
902–908.
30. Ito T, et al. Molecular basis for the genera-
tion in pigs of influenza A viruses with
pandemic potential. J Virol 1998;72:
7367–7373.
31. Matrosovich M, et al. Early alterations of the
receptor-binding properties of H1, H2, and
H3 avian influenza virus hemagglutinins
after their introduction into mammals. J
Virol 2000;74:8502–8512.
32. Connor RJ, Kawaoka Y, Webster RG, Paulson
JC. Receptor specificity in human, avian, and
equine H2 and H3 influenza virus isolates.
Virology 1994;205:17–23.
33. Shinya K, Kawaoka Y. Influenza virus recep-
tors in the human airway. Uirusu 2006;56:
85–89.
34. Rust MJ, Lakadamyali M, Zhang F, Zhuang X.
Assembly of endocytic machinery around
individual influenza viruses during viral
entry. Nat Struct Mol Biol 2004;11:
567–573.
35. Lakadamyali M, Rust MJ, Zhuang X. Endo-
cytosis of influenza vir uses. Microbes In-
fect/Inst Pasteur 2004;6:929–936.
36. Pinto LH, Holsinger LJ, Lamb RA. Influenza
virus M2 protein has ion channel activity.
Cell 1992;69:517–528.
37. Plotch SJ, Bouloy M, Ulmanen I, Krug RM. A
unique cap (m7GpppXm)-dependent influ-
enza virion endonuclease cleaves capped
RNAs to generate the primers that initiate
viral RNA transcription. Cell 1981;23:
847–858.
38. Martin K, Helenius A. Nuclear transport of
influenza virus ribonucleoproteins: the viral
matrix protein (M1) promotes export and
inhibits import. Cell 1991;67:117–130.
39. O’Neill RE, Talon J, Palese P. The influenza
virus NEP (NS2 protein) mediates the
nuclear export of viral ribonucleoproteins.
EMBO J 1998;17:288–296.
40. Lamb RA, Takeda M. Death by influenza
virus protein. Nat Med 2001;7:1286–1288.
41. Taubenberger JK, Reid AH, Lourens RM,
Wang R, Jin G, Fanning TG. Characterization
of the 1918 influenza vir us polymerase
genes. Nature 2005;437:889–893.
42. Taubenberger JK, Morens DM. 1918 influ-
enza: the mother of all pandemics. Emerg
Infect Dis 2006;12:15–22.
43. Reid AH, Taubenberger JK, Fanning TG.
Evidence of an absence: the genetic origins
of the 1918 pandemic influenza virus. Nat
Rev 2004;2:909–914.
44. Kawaoka Y, Krauss S, Webster RG. Avian-to-
human transmission of the PB1 gene of
influenza A viruses in the 1957 and 1968
pandemics. J Virol 1989;63:4603–4608.
45. Naeve CW, Hinshaw VS, Webster RG.
Mutations in the hemagglutinin receptor-
binding site can change the biological
properties of an influenza virus. J Virol
1984;51:567–569.
46. Glaser L, et al. A single amino acid substitu-
tion in 1918 influenza virus hemagglutinin
changes receptor binding specificity. J Virol
2005;79:11533–11536.
47. Monto AS, Gravenstein S, Elliott M, Colopy
M, Schweinle J. Clinical signs and symptoms
predicting influenza infection. Arch Int Med
2000;160:3243–3247.
48. Nicholson KG. Clinical features of influenza.
Semin Respir Infect 1992;7:26–37.
49. Taubenberger JK, Morens DM. The pathol-
ogy of influenza virus infections. Ann Rev
Pathol 2008;3:499–522.
50. Hayden FG, Fritz R, Lobo MC, Alvord W,
Strober W, Straus SE. Local and systemic
cytokine responses during experimental
human influenza A virus infection.
Relation to symptom formation and
host defense. J Clin Invest 1998;
101:643–649.
51. Fritz RS, et al. Nasal cytokine and chemo-
kine responses in experimental influenza A
virus infection: results of a placebo-con-
trolled trial of intravenous zanamivir treat-
ment. J Infect Dis 1999;180:586–593.
52. Skoner DP, Gentile DA, Patel A, Doyle WJ.
Evidence for cytokine mediation of disease
expression in adults experimentally infected
with influenza A virus. J Infect Dis 1999;
180:10–14.
53. DeLay PD, Casey HL, Tubiash HS. Compara-
tive study of fowl plague virus and a virus
isolated from man. Public Health Rep
1967;82:615–620.
54. Campbell CH, Webster RG, Breese SS Jr. Fowl
plague virus from man. J Infect Dis
1970;122:513–516.
55. Taylor HR, Turner AJ. A case report of fowl
plague keratoconjunctivitis. Br J Ophthalmol
1977;61:86–88.
56. Webster RG, Geraci J, Petursson G, Skirnis-
son K. Conjunctivitis in human beings
caused by influenza A virus of seals. N Engl J
Med 1981;304:911.
57. Kurtz J, Manvell RJ, Banks J. Avian influenza
virus isolated from a woman with conjunc-
tivitis. Lancet 1996;348:901–902.
58. Banks J, Speidel E, Alexander DJ. Character-
isation of an avian influenza A virus isolated
from a human is an intermediate host
necessary for the emergence of pandemic
influenza viruses? Arch Virol 1998;143:
781–787.
59. Koopmans M, et al. Transmission of H7N7
avian influenza A vir us to human beings
during a large outbreak in commercial
Maines et al Influenza virus pathogenesis and innate immunity
r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008 81
poultry farms in the Netherlands. Lancet
2004;363:587–593.
60. Chotpitayasunondh T, et al. Human disease
from influenza A (H5N1), Thailand,
2004. Emerg Infect Dis 2005;11:201–209.
61. Fouchier RA, et al. Avian influenza A virus
(H7N7) associated with human conjuncti-
vitis and a fatal case of acute respiratory
distress syndrome. Proc Natl Acad Sci USA
2004;101:1356–1361.
62. Tweed SA, et al. Human illness from avian
influenza H7N3, British Columbia. Emerg
Infect Dis 2004;10:2196–2199.
63. Hirst M, et al. Novel avian influenza H7N3
strain outbreak, British Columbia. Emerg
Infect Dis 2004;10:2192–2195.
64. Herlocher ML, et al. Influenza viruses resis-
tant to the antiviral drug oseltamivir: trans-
mission studies in ferrets. J Infect Dis
2004;190:1627–1630.
65. Avian influenza A/(H7N2) outbreak in the
United Kingdom. Euro Surveill. 2007;
12:E0705312.
66. Lin YP, et al. Avian-to-human transmission
of H9N2 subtype influenza A viruses: rela-
tionship between H9N2 and H5N1 human
isolates. Proc Natl Acad Sci USA 2000;97:
9654–9658.
67. Butt KM, et al. Human infection with an
avian H9N2 influenza A virus in Hong Kong
in 2003. J Clin Microbiol 2005;43:
5760–5767.
68. Guo Y, Li J, Cheng X. Discovery of men
infected by avian influenza A (H9N2) virus.
Chin J Exp Clin Virol 1999;13:105–108.
69. Claas EC, et al. Human influenza A H5N1
virus related to a highly pathogenic avian
influenza virus. Lancet 1998;351:
472–477.
70. Subbarao K, et al. Characterization of an
avian influenza A (H5N1) virus isolated
from a child with a fatal respiratory illness.
Science (New York, NY) 1998;279:
393–396.
71. Yuen KY, et al. Clinical features and rapid
viral diagnosis of human disease associated
with avian influenza A H5N1 virus. Lancet
1998;351:467–471.
72. Peiris JS, et al. Re-emergence of fatal human
influenza A subtype H5N1 disease. Lancet
2004;363:617–619.
73. Kandun IN, et al. Three Indonesian clusters
of H5N1 virus infection in 2005. N Engl J
Med 2006;355:2186–2194.
74. Olsen SJ, et al. Family clustering of avian
influenza A (H5N1). Emerg Infect Dis
2005;11:1799–1801.
75. Avian influenza virus A (H10N7) circulating
among humans in Egypt. EID Weekly
Updates. 2004; 2.
76. Tran TH, et al. Avian influenza A (H5N1) in
10 patients in Vietnam. N Engl J Med
2004;350:1179–1188.
77. Beigel JH, et al. Avian influenza A (H5N1)
infection in humans. N Engl J Med
2005;353:1374–1385.
78. Apisarnthanarak A, et al. Atypical avian
influenza (H5N1). Emerg Infect Dis
2004;10:1321–1324.
79. de Jong MD, et al. Fatal avian influenza A
(H5N1) in a child presenting with diarrhea
followed by coma. N Engl J Med
2005;352:686–691.
80. Uiprasertkul M, et al. Apoptosis and patho-
genesis of avian influenza A (H5N1) virus in
humans. Emerg Infect Dis
2007;13:708–712.
81. Fisman DN. Hemophagocytic syndromes
and infection. Emerg Infect Dis
2000;6:601–608.
82. To KF, et al. Pathology of fatal human
infection associated with avian influenza
A H5N1 virus. J Med Virol
2001;63:242–246.
83. de Jong MD, et al. Fatal outcome of human
influenza A (H5N1) is associated with high
viral load and hypercytokinemia. Nat Med
2006;12:1203–1207.
84. Maines TR, et al. Lack of transmission of
H5N1 avian-human reassortant influenza
viruses in a ferret model. Proc Natl Acad Sci
USA 2006;103:12121–12126.
85. Gao P, et al. Biological heterogeneity, in-
cluding systemic replication in mice, of
H5N1 influenza A virus isolates from hu-
mans in Hong Kong. J Virol
1999;73:3184–3189.
86. Rimmelzwaan GF, Kuiken T, van Ameron-
gen G, Bestebroer TM, Fouchier RA,
Osterhaus AD. Pathogenesis of influenza
A (H5N1) virus infection in a
primate model. J Virol 2001;75:
6687–6691.
87. Zitzow LA, Rowe T, Morken T, Shieh WJ,
Zaki S, Katz JM. Pathogenesis of avian influ-
enza A (H5N1) viruses in ferrets. J Virol
2002;76:4420–4429.
88. Kuiken T, et al. Avian H5N1 influenza in
cats. Science (New York, NY)
2004;306:241.
89. Baum LG, Paulson JC. Sialyloligosaccharides
of the respiratory epithelium in the selection
of human influenza virus receptor specifi-
city. Acta Histochem 1990;40
(Suppl.):35–38.
90. Leigh MW, Connor RJ, Kelm S, Baum LG,
Paulson JC. Receptor specificity of influenza
virus influences severity of illness in ferrets.
Vaccine 1995;13:1468–1473.
91. van Riel D, et al. H5N1 virus attachment to
lower respiratory tract. Science (New York,
NY) 2006;312:399.
92. Herlocher ML, et al. Ferrets as a transmission
model for influenza: sequence changes in
HA1 of type A (H3N2) virus. J Infect Dis
2001;184:542–546.
93. Svitek N, Rudd PA, Obojes K, Pillet S, von
Messling V. Severe seasonal influenza in
ferrets correlates with reduced interferon
and increased IL-6 induction. Virology
2008;376:53–59.
94. Govorkova EA, et al. Lethality to ferrets of
H5N1 influenza viruses isolated from hu-
mans and poultry in 2004. J Virol
2005;79:2191–2198.
95. Maines TR, et al. Avian influenza (H5N1)
viruses isolated from humans in Asia in
2004 exhibit increased virulence in mam-
mals. J Virol 2005;79:11788–11800.
96. Belser JA, et al. Pathogenesis of avian influ-
enza (H7) virus infection in mice and
ferrets: enhanced virulence of Eurasian
H7N7 viruses isolated from humans. J Virol
2007;81:11139–11147.
97. Abdel-Ghafar AN, et al. Update on avian
influenza A (H5N1) virus infection in
humans. N Engl J Med 2008;358:
261–273.
98. Svitek N, von Messling V. Early cytokine
mRNA expression profiles predict Morbilli-
virus disease outcome in ferrets. Virology
2007;362:404–410.
99. Ochi A, et al. Cloning, expression and
immunoassay detection of ferret IFN-gam-
ma. Dev Comp Immunol 2008;32:
890–897.
100. Danesh A, et al. Cloning, expression and
characterization of ferret CXCL10. Mol Im-
munol 2008;45:1288–1297.
101. Ibricevic A, et al. Influenza virus receptor
specificity and cell tropism in mouse and
human airway epithelial cells. J Virol
2006;80:7469–7480.
102. Conn CA, McClellan JL, Maassab HF, Smitka
CW, Majde JA, Kluger MJ. Cytokines and
the acute phase response to influenza
virus in mice. Am J Physiol
1995;268:R78–R84.
103. Hennet T, Ziltener HJ, Frei K, Peterhans E. A
kinetic study of immune mediators in the
lungs of mice infected with influenza A
virus. J Immunol 1992;149:932–939.
104. Peper RL, Van Campen H. Tumor necrosis
factor as a mediator of inflammation in
influenza A viral pneumonia. Microbial
Pathogen 1995;19:175–183.
105. Vacheron F, Rudent A, Perin S, Labarre C,
Quero AM, Guenounou M. Production of
interleukin 1 and tumour necrosis factor
activities in bronchoalveolar washings
following infection of mice by influenza
virus. J Gen Virol 1990;71 (Part
2):477–479.
106. Lu X, Tumpey TM, Morken T, Zaki SR,
Cox NJ, Katz JM. A mouse model for
the evaluation of pathogenesis and immu-
nity to influenza A (H5N1) viruses
isolated from humans. J Virol
1999;73:5903–5911.
Maines et al Influenza virus pathogenesis and innate immunity
82 r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008
107. Tumpey TM, Lu X, Morken T, Zaki SR, Katz
JM. Depletion of lymphocytes and dimin-
ished cytokine production in mice infected
with a highly virulent influenza A (H5N1)
virus isolated from humans. J Virol
2000;74:6105–6116.
108. Katz JM, Lu X, Frace AM, Morken T,
Zaki SR, Tumpey TM. Pathogenesis of
and immunity to avian influenza A H5
viruses. Biomed Pharmacother
2000;54:178–187.
109. Katz JM, Lu X, Tumpey TM, Smith CB,
Shaw MW, Subbarao K. Molecular correlates
of influenza A H5N1 virus pathogenesis
in mice. J Virol 2000;74:10807–10810.
110. Rigoni M, et al. Pneumo- and neurotropism
of avian origin Italian highly pathogenic
avian influenza H7N1 isolates in experi-
mentally infected mice. Virology
2007;364:28–35.
111. Lu X, Renshaw M, Tumpey TM, Kelly GD,
Hu-Primmer J, Katz JM. Immunity to influ-
enza A H9N2 viruses induced by infection
and vaccination. J Virol 2001;75:
4896–4901.
112. Hatta M, Gao P, Halfmann P, Kawaoka Y.
Molecular basis for high virulence of Hong
Kong H5N1 influenza A viruses. Science
(New York, NY) 2001;293:1840–1842.
113. Li Z, et al. Molecular basis of replication of
duck H5N1 influenza vir uses in a mamma-
lian mouse model. J Virol 2005;79:
12058–12064.
114. Salomon R, et al. The polymerase complex
genes contribute to the high virulence of the
human H5N1 influenza virus isolate A/
Vietnam/1203/04. J Exp Med 2006;203:
689–697.
115. Gabriel G, Dauber B, Wolff T, Planz O, Klenk
HD, Stech J. The viral polymerase mediates
adaptation of an avian influenza virus to a
mammalian host. Proc Natl Acad Sci USA
2005;102:18590–18595.
116. Coleman JR. The PB1-F2 protein of Influen-
za A virus: increasing pathogenicity by dis-
rupting alveolar macrophages. Virol J
2007;4:9.
117. Zamarin D, Garcia-Sastre A, Xiao X, Wang R,
Palese P. Influenza virus PB1-F2 protein in-
duces cell death through mitochondrial ANT3
and VDAC1. PLoS Pathogens 2005;1:e4.
118. Zamarin D, Ortigoza MB, Palese P. Influenza
A virus PB1-F2 protein contributes to viral
pathogenesis in mice. J Virol
2006;80:7976–7983.
119. McAuley JL, et al. Expression of the 1918
influenza A vir us PB1-F2 enhances the
pathogenesis of viral and secondary bacter-
ial pneumonia. Cell Host Microbe
2007;2:240–249.
120. Conenello GM, Zamarin D, Perrone LA,
Tumpey T, Palese P. A single mutation in the
PB1-F2 of H5N1 (HK/97) and 1918 influ-
enza A vir uses contributes to increased
virulence. PLoS Pathogens 2007;3:e141.
121. Seo SH, Hoffmann E, Webster RG. Lethal
H5N1 influenza viruses escape host anti-
viral cytokine responses. Nat Med
2002;8:950–954.
122. Lipatov AS, et al. Pathogenesis of Hong
Kong H5N1 influenza virus NS gene reas-
sortants in mice: the role of cytokines and
B- and T-cell responses. J Gen Virol
2005;86:1121–1130.
123. Szretter KJ, et al. Role of host cytokine
responses in the pathogenesis of avian
H5N1 influenza viruses in mice. J Virol
2007;81:2736–2744.
124. Kozak W, Zheng H, Conn CA, Soszynski D,
van der Ploeg LH, Kluger MJ. Thermal
and behavioral effects of lipopolysaccharide
and influenza in interleukin-1 beta-
deficient mice. Am J Physiol
1995;269:R969–R977.
125. Imai Y, et al. Identification of oxidative stress
and Toll-like receptor 4 signaling as a key
pathway of acute lung injury. Cell
2008;133:235–249.
126. Rothwell NJ. Annual review prize lecture
cytokines killers in the brain? J Physiol
1999;514 (Part 1):3–17.
127. Perrone LA, Plowden JK, Garcia-Sastre A,
Katz JM, Tumpey TM. H5N1 and 1918
pandemic influenza virus infection results in
early and excessive infiltration of macro-
phages and neutrophils in the lungs of mice.
PLoS Pathogens 2008;4:e1000115.
128. Tumpey TM, et al. Characterization of the
reconstructed 1918 Spanish influenza pan-
demic virus. Science (New York, NY)
2005;310:77–80.
129. Kozak W, Poli V, Soszynski D, Conn CA, Leon
LR, Kluger MJ. Sickness behavior in mice
deficient in interleukin-6 during turpentine
abscess and influenza pneumonitis. Am J
Physiol 1997;272:R621–R630.
130. Jones E, Price DA, Dahm-Vicker M, Cerun-
dolo V, Klenerman P, Gallimore A. The
influence of macrophage inflammatory
protein-1alpha on protective immunity
mediated by antiviral cytotoxic T cells. Im-
munology 2003;109:68–75.
131. Cook DN, et al. Requirement of MIP-1 alpha
for an inflammatory response to viral infec-
tion. Science (New York, NY)
1995;269:1583–1585.
132. Dawson TC, Beck MA, Kuziel WA, Hender-
son F, Maeda N. Contrasting effects of CCR5
and CCR2 deficiency in the pulmonary
inflammatory response to influenza A vir us.
Am J Pathol 2000;156:1951–1959.
133. Schmitz N, Kurrer M, Bachmann MF, Kopf
M. Interleukin-1 is responsible for acute
lung immunopathology but increases survi-
val of respiratory influenza virus infection. J
Virol 2005;79:6441–6448.
134. Simon AK, et al. Tumor necrosis factor-
related apoptosis-inducing ligand in T cell
development: sensitivity of human thymo-
cytes. Proc Natl Acad Sci USA
2001;98:5158–5163.
135. Wang J, et al. The critical role of LIGHT,
a TNF family member, in T cell develop-
ment. J Immunol 2001;167:5099–5105.
136. Yoneyama M, Fujita T. RIG-I family RNA
helicases: cytoplasmic sensor for antiviral
innate immunity. Cytokine Growth Factor
Rev 2007;18:545–551.
137. Guillot L, et al. Involvement of toll-like
receptor 3 in the immune response of lung
epithelial cells to double-stranded RNA and
influenza A virus. J Biol Chem 2005;280:
5571–5580.
138. Diebold SS, Kaisho T, Hemmi H, Akira S, Reis
e Sousa C. Innate antiviral responses by
means of TLR7-mediated recognition of sin-
gle-stranded RNA. Science (New York, NY)
2004;303:1529–1531.
139. Mibayashi M, Martinez-Sobrido L, Loo YM,
Cardenas WB, Gale M Jr., Garcia-Sastre A.
Inhibition of retinoic acid-inducible gene
I-mediated induction of beta interferon by
the NS1 protein of influenza A vir us. J Virol
2007;81:514–524.
140. Guo Z, et al. NS1 protein of influenza A virus
inhibits the function of intracytoplasmic
pathogen sensor, RIG-I. Am J Respir Cell
Mol Biol 2007;36:263–269.
141. Garcia-Sastre A, et al. The role of interferon
in influenza virus tissue tropism. J Virol
1998;72:8550–8558.
142. Chan MC, et al. Proinflammatory cytokine
responses induced by influenza A (H5N1)
viruses in primary human alveolar and
bronchial epithelial cells. Respir Res
2005;6:135.
143. Zeng H, et al. Highly pathogenic avian
influenza H5N1 viruses elicit an attenuated
type I interferon response in polarized hu-
man bronchial epithelial cells. J Virol
2007;81:12439–12449.
144. Kobasa D, et al. Aberrant innate immune
response in lethal infection of macaques
with the 1918 influenza virus. Nature
2007;445:319–323.
145. Cheung CY, et al. Induction of proinflam-
matory cytokines in human macrophages
by influenza A (H5N1) viruses: a
mechanism for the unusual severity of
human disease? Lancet
2002;360:1831–1837.
146. Zhou J, Law HK, Cheung CY, Ng IH, Peiris
JS, Lau YL. Functional tumor necrosis factor-
related apoptosis-inducing ligand produc-
tion by avian influenza virus-infected
macrophages. J Infect Dis 2006;193:
945–953.
147. Wurzer WJ, et al. NF-kappaB-dependent
induction of tumor necrosis factor-related
Maines et al Influenza virus pathogenesis and innate immunity
r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008 83
apoptosis-inducing ligand (TRAIL) and Fas/
FasL is crucial for efficient influenza virus
propagation. J Biol Chem 2004;279:
30931–30937.
148. Thitithanyanont A, et al. High susceptibil-
ity of human dendritic cells to avian influ-
enza H5N1 virus infection and protection
by IFN-alpha and TLRligands. J Immunol
2007;179:5220–5227.
149. de Jong MD, et al. Oseltamivir resistance
during treatment of influenza A (H5N1)
infection. N Engl J Med
2005;353:2667–2672.
150. Salomon R, Hoffmann E, Webster RG. In-
hibition of the cytokine response does not
protect against lethal H5N1 influenza infec-
tion. Proc Natl Acad Sci USA 2007;104:
12479–12481.
151. Budd A, et al. Increased survival after gem-
fibrozil treatment of severe mouse influen-
za. Antimicrob Agents Chemother
2007;51:2965–2968.
152. Rainsford KD. Influenza (‘‘Bird Flu’’),
inflammation and anti-inflammatory/
analgesic drugs. Inflammopharmacology
2006;14:2–9.
153. Zheng BJ, et al. Delayed antiviral plus
immunomodulator treatment still
reduces mortality in mice infected by high
inoculum of influenza A/H5N1 virus.
Proc Natl Acad Sci USA 2008;
105:8091–8096.
154. Tumpey TM, et al. A two-amino acid
change in the hemagglutinin of the 1918
influenza virus abolishes transmission.
Science (New York, NY) 2007;315:
655–659.
155. Shortridge KF, Stuart-Harris CH. An influ-
enza epicentre? Lancet 1982;2:812–813.
156. Stevens J, Blixt O, Tumpey TM, Taubenber-
ger JK, Paulson JC, Wilson IA. Structure
and receptor specificity of the hemaggluti-
nin from an H5N1 influenza virus. Science
(New York, NY) 2006;312:404–410.
157. de Wit E, et al. Protection of mice
against lethal infection with highly
pathogenic H7N7 influenza A virus by
using a recombinant low-pathogenicity
vaccine strain. J Virol 2005;79:
12401–12407.
158. Munster VJ, et al. The molecular basis
of the pathogenicity of the Dutch highly
pathogenic human influenza A H7N7
viruses.
J Infect Dis 2007;196:258–265.
159. CDC. Update: influenza activity United
States and worldwide, 2003–04 season, and
composition of the 2004–05 influenza
vaccine. Morb Mortal Wkly Rep
2004;53:547–552.
160. CDC. Update: influenza activity United
States, 2003–04 season. Morb Mortal Wkly
Rep 2004;53:284–287.
161. Peiris M, et al. Human infection with
influenza H9N2. Lancet 1999;354:
916–917.
Maines et al Influenza virus pathogenesis and innate immunity
84 r2008 The Authors Journal compilation r2008 Blackwell Munksgaard Immunological Reviews 225/2008
... The acute phase of infection is characterized by a multifocal destruction of the epithelium with cellular desquamation to the luminal space, which may often lead to patches along the mucosa with few or no epithelial cells over the basement membrane. It is also associated with the presence of oedema, vascular congestion, hyperaemia and infiltration of inflammatory cells into the submucosa underlying the epithelial layer [120,121]. ...
... In addition to being important natural hosts of influenza A viruses, pigs constitute a valuable animal model for the study of influenza infection [101,121], as virus may be transmitted experimentally to in-contact animals and they can exhibit clinical signs, histopathology and cytokine expression profiles comparable to influenza infection in other animal species [35,101,120,121,123]. ...
... In addition to being important natural hosts of influenza A viruses, pigs constitute a valuable animal model for the study of influenza infection [101,121], as virus may be transmitted experimentally to in-contact animals and they can exhibit clinical signs, histopathology and cytokine expression profiles comparable to influenza infection in other animal species [35,101,120,121,123]. ...
Thesis
Full-text available
Ex vivo organ cultures (EVOC) of tracheal explants with an air interface system have been successfully developed and used in the study of both human and animal respiratory pathogens. Such systems reproduce, to a great extent, the physiological conditions and the cellular complexity of the respiratory mucosa of the host in a highly controlled setting. In this study, we developed an EVOC system using pig trachea to quantify the infection dynamics of influenza A viruses. Pig tracheal explants maintained the structure and function present in the living host preserving the intricate complexity of the respiratory mucosa during the first four days of the organ culture. Inoculation with the swine virus A/swine/England/435/06 (H1N1) resulted in productive viral infection in a dose-dependent fashion. Infected explants showed histopathology of influenza infection. Viral titres peaked at day 2 post-inoculation and ciliary activity was abrogated shortly after. Explants collected from different sections of the trachea showed significant differences in the production of infectious virus. The host innate immunity induction was evaluated by quantifying the mRNA levels of the cytokines IFN-β, IFN-α, TNF-α, IL-1α, IL-6 and the chemokine IL-8 during influenza infection of tracheal explant. In uninfected explants, IFN-β IFN-α and TNF-α mRNA expression levels did not change while cytokines associated with the inflammatory response - IL-1α, IL-6 and IL-8 - were significantly elevated throughout the organ culture. In influenza-infected explants the elevation in IFN-β expression appeared to be the predominant early response to influenza infection. IFN-α and TNF-α levels were not significantly altered whereas IL-1α, IL-6 and IL-8 expression showed a marked decrease in expression when the viral genome copy numbers peaked. Productive infection was also observed in explants infected with the 2009 influenza pandemic virus A/England/195/09 (H1N1) but not with laboratory-adapted strain A/Puerto Rico/8/34 (H1N1) and not related with the induction of the innate immune system in tracheal explants.
... Juveniles/young individuals might be more susceptible due to their naive immune system, but adults displayed increased inflammatory responses, leading to higher disease severity in older individuals (e.g. Rioux et al., 2021;Gholipour et al., 2017;Maines et al., 2008). Different exposure risks and the likelihood of co-infections might exist across age groups, and behavioural differences may make infected or clinically ill adult mammals less likely to be encountered (Elsmo et al., 2023). ...
... This is yet to be demonstrated, as experiments are restricted to a few combinations of subtypes/strains and mammal species, mostly ferrets (Mustela furo) (but see Xiang et al., 2020;Lina et al., 2019;Root et al., 2016;Romero Tejada et al., 2015;Root et al., 2014). Ferrets are recognised as an excellent model of influenza as they are susceptible to human and avian influenza viruses, exhibit disease signs and severity resembling those in humans, and these are dependent on host factors such as obesity, pregnancy, immune status, age, and viral strain (Cáceres et al., 2021;Rioux et al., 2021;Maines et al., 2008;Maher and DeStefano, 2004). Yet, they cannot represent the diversity of species infected in the wild. ...
Article
Full-text available
Avian influenza (AI) is an infectious viral disease of birds, including domestic poultry, which has been causing outbreaks worldwide, leading to several millions of dead wild birds and culled poultry. AI is mainly found in birds, but recently, there was an increase in reported infections in mammals, ranging from no symptoms to mass mortality events and some human cases. Epidemiologically of great concern, evidence of mammalian adaptations have been found, but the transmission routes and pathogenesis in mammals are still to be defined. Hence, it is paramount to address all facets of AI viruses epidemiology, including investigating taxa not customarily thought to be involved in the transmission and/or trafficking of AI, such as wild mammals. The scope of this report was to assess the role of mammals in AI epidemiology, virology and pathology, i.e. AI maintenance, reservoir role, immunity, role of mammals in a potential pandemic. To do so, we performed an all-encompassing review of the literature on the topic with a two-fold approach: a systematic review of the published AI cases in wild mammals and a narrative approach to provide an expert opinion on the role of mammals in AI spread. The final number of peer-reviewed papers included in the systematic literature review was 76, resulting in 120 unique infection records with AI in wild mammal species. The most represented taxa were included in the order Carnivora. The risk of infection was identified mainly as predation (or feeding) upon infected birds or contact with avian species. Evidence of mammal-to-mammal transmission in the wild is only circumstantial and yet to be confirmed. Cases of AI from the systematic review of experimental findings were discussed concerning epidemiology, pathology and virology. Knowledge gaps and potential pandemic drivers were identified. In summary, although a greater number of infections in wild mammals have been reported, there is no hard evidence for sustained mammal-to-mammal transmission in the wild. The factors contributing to the increased number of infections found in wild carnivores are not clear yet, but the unprecedented global spread of highly pathogenic avian influenza (HPAI) viruses creates ample opportunities for intense, mostly alimentary, contact between infected wild birds and carnivores. Close surveillance of circulating strains and continued assessment of new epidemiological situations are crucial to quickly identify strains with enhanced mammalian fitness.
... According to Hethcote, deterministic epidemiological modeling started early in the twentieth century for studying the dynamics of measles epidemics. Although much of today's focus on epidemiological modeling is on non-infectious disease (the continued importance of infectious diseases in developing countries, as well as emerging or reemerging diseases in developed countries, has sustained a substantial need for and interest in mathematical modeling of infectious diseases (Hethcote, 2000;Maines et al., 2008). Some basic concepts in epidemiology that are carried in epizootiology include basic classes and flow patterns in epidemiological models such as SEIR, where S is susceptible individuals, E is those exposed, I those infected, and R recovered (immune individuals), as well as the threshold concept, i.e., that the density of susceptible individuals must exceed a critical value for an epidemic outbreak to occur. ...
Chapter
Full-text available
... Moreover, another study underscored the significant role of macrophages in the pathogenicity of the influenza A virus. (Maines et al., 2008). Given the established link between TNF-α and macrophages, we propose that ALA's prophylactic properties might influence macrophage activity. ...
... In contrast to a previous study on IAV-induced senescence (12), we observed lower viral loads in the aged pulmonary tissue for all the IAV strains and SARS-CoV-2 isolates tested. Nevertheless, using comprehensive approaches to analyse autopsy-derived lung specimens of patients with COVID-19 in contrast to age- Several studies have demonstrated that H5N1 infection leads to a more robust induction of cytokines and chemokines compared to H1N1 (88)(89)(90)(91)(92). Furthermore, H5N1 infected humans present unusually high serum concentrations of pro-inflammatory cytokines and chemokines, which are believed to contribute to disease severity (57)(58)(59)93). Similarly, characteristic features of severe COVID-19 rely in the excessive release of pro-inflammatory cytokines, a so-called "cytokine storm", and an immune dysregulation, specifically, a massive immune stimulation resulting in a macrophage activation syndrome, aberrant neutrophil activation, or T cell hyperreactivity. ...
Preprint
Full-text available
The recent coronavirus disease 2019 (COVID-19) outbreak revealed the susceptibility of elderly patients to respiratory virus infections, showing cell senescence or subclinical persistent inflammatory profiles and favouring the development of severe pneumonia. In our study, we evaluated the potential influence of lung aging on the efficiency of replication of influenza A virus (IAV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV 2), as well as determined the pro-inflammatory and antiviral responses of the distal lung tissue. Using precision-cut lung slices (PCLS) from donors of different ages, we found that pandemic H1N1 and avian H5N1 IAV replicated in the lung parenchyma with high efficacy. In contrast to these IAV strains, SARS-CoV-2 early isolate and Delta variant of concern (VOC) replicated less efficiently in PCLS. Interestingly, both viruses showed reduced replication in PCLS from older compared to younger donors, suggesting that aged lung tissue represents a sub optimal environment for viral replication. Regardless of the age-dependent viral loads, PCLS responded to infection with both viruses by an induction of IL-6 and IP-10/CXCL10 mRNAs, being highest for H5N1. Finally, while SARS-CoV-2 infection was not causing detectable cell death, IAV infection caused significant cytotoxicity and induced significant early interferon responses. In summary, our findings suggest that aged lung tissue might not favour viral dissemination, pointing to a determinant role of dysregulated immune mechanisms in the development of severe disease.
... The innate immune system is the first line of defense against influenza virus infection consisting of physical barriers and innate cellular immune responses. During influenza virus infection, epithelial cells on the mucosa are infected and subsequently macrophages may be the cells that first responded to them immunologically and play a crucial role in virus resistance (Maines et al., 2008). Infection of respiratory epithelial cells with IAV is initiated by the binding of cell surface salivary acids to the viral HA proteins (Wiley and Skehel, 1987;Skehel and Wiley, 2000). ...
Article
Full-text available
Influenza viruses are one of the major causes of human respiratory infections and the newly emerging and re-emerging strains of influenza virus are the cause of seasonal epidemics and occasional pandemics, resulting in a huge threat to global public health systems. As one of the early immune cells can rapidly recognize and respond to influenza viruses in the respiratory, lung macrophages play an important role in controlling the severity of influenza disease by limiting viral replication, modulating the local inflammatory response, and initiating subsequent adaptive immune responses. However, influenza virus reproduction in macrophages is both strain- and macrophage type-dependent, and ineffective replication of some viral strains in mouse macrophages has been observed. This review discusses the function of lung macrophages in influenza virus infection in order to better understand the pathogenesis of the influenza virus.
... Previously, mathematical models have investigated human inflammation following physical injury [19][20][21], cancer [22,23], lung infections [10], rheumatoid arthritis [24,25], atherosclerosis [26], H. pylori [27], diabetes [28], and nonspecific pathogens [29][30][31][32][33][34][35]. Specifically, mathematical modeling of viral infections with pathogens such as influenza [36][37][38][39][40], hepatitis B [41][42][43][44], SARS-CoV [45,46], and SARS-CoV-2 [47][48][49][50][51][52][53][54][55][56] have investigated the role of immune responses in disease resolution or inflammation. Some of these studies highlighted the importance of cytokine production and neutrophil function in modulating disease pathogenesis. ...
Article
Full-text available
Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) results in varied clinical outcomes, with virus-induced chronic inflammation and tissue injury being associated with enhanced disease pathogenesis. To determine the role of tissue damage on immune populations recruitment and function, a mathematical model of innate immunity following SARS-CoV-2 infection has been proposed. The model was fitted to published longitudinal immune marker data from patients with mild and severe COVID-19 disease and key parameters were estimated for each clinical outcome. Analytical, bifurcation, and numerical investigations were conducted to determine the effect of parameters and initial conditions on long-term dynamics. The results were used to suggest changes needed to achieve immune resolution.
... The results demonstrate that the HP virus is more capable of infecting susceptible cells and reducing interferon response from infected cells. The results are supported by a variety of experimental studies where enhanced infection and replication rates [28,29] and attenuated interferon production rates [3,9,12,13,[30][31][32][33][34] are evidenced as possible explanations to high viral pathogenicity. Fig 3D shows that the rate of infection-induced macrophage recruitment s V is lower for the HP strain (97.4% of the ratio samples are less than 1), suggesting that a high recruitment rate is not the cause for the observed high level of macrophages during the HP infection seen in Fig 2. Instead, our model result indicates that the high level of macrophages is due to a higher number of infected cells which activate more macrophages. ...
Article
Full-text available
Epidemiological and clinical evidence indicates that humans infected with the 1918 pandemic H1N1 influenza virus and highly pathogenic avian H5N1 influenza viruses often displayed severe lung pathology. High viral load and extensive infiltration of macrophages are the hallmarks of highly pathogenic (HP) influenza viral infections. However, it remains unclear what biological mechanisms primarily determine the observed difference in the kinetics of viral load and macrophages between HP and low pathogenic (LP) viral infections, and how the mechanistic differences are associated with viral pathogenicity. In this study, we develop a mathematical model of viral dynamics that includes the dynamics of different macrophage populations and interferon. We fit the model to in vivo kinetic data of viral load and macrophage level from BALB/c mice infected with an HP or LP strain of H1N1/H5N1 virus using Bayesian inference. Our primary finding is that HP viruses have a higher viral infection rate, a lower interferon production rate and a lower macrophage recruitment rate compared to LP viruses, which are strongly associated with more severe tissue damage (quantified by a higher percentage of epithelial cell loss). We also quantify the relative contribution of macrophages to viral clearance and find that macrophages do not play a dominant role in the direct clearance of free viruses although their role in mediating immune responses such as interferon production is crucial. Our work provides new insight into the mechanisms that convey the observed difference in viral and macrophage kinetics between HP and LP infections and establishes an improved model-fitting framework to enhance the analysis of new data on viral pathogenicity.
Article
Full-text available
Acute respiratory distress syndrome is an acute respiratory failure caused by cytokine storms; highly pathogenic influenza A virus infection can induce cytokine storms. The innate immune response is vital in this cytokine storm, acting by activating the transcription factor NF-κB. Tissue injury releases a danger-associated molecular pattern that provides positive feedback for NF-κB activation. Exogenous mesenchymal stem cells can also modulate immune responses by producing potent immunosuppressive substances, such as prostaglandin E2. Prostaglandin E2 is a critical mediator that regulates various physiological and pathological processes through autocrine or paracrine mechanisms. Activation of prostaglandin E2 results in the accumulation of unphosphory-lated β-catenin in the cytoplasm, which subsequently reaches the nucleus to inhibit the transcription factor NF-κB. The inhibition of NF-κB by β-catenin is a mechanism that reduces inflammation.
Article
We consider a hierarchy of ordinary differential equation models that describe the within-host viral kinetics of influenza infections: the IR model explicitly accounts for an immune response to the virus, while the simpler, target-cell limited TEIV and TV models do not. We show that when the IR model is fitted to pooled experimental murine data of the viral load, fraction of dead cells, and immune response levels, its parameters values can be determined. However, if, as is common, only viral load data are available, we can estimate parameters of the TEIV and TV models but not the IR model. These results are substantiated by a structural and practical identifiability analysis. We then use the IR model to generate synthetic data representing infections in hosts whose immune responses differ. We fit the TV model to these synthetic datasets and show that it can reproduce the characteristic exponential increase and decay of viral load generated by the IR model. Furthermore, the values of the fitted parameters of the TV model can be mapped from the immune response parameters in the IR model. We conclude that, if only viral load data are available, a simple target-cell limited model can reproduce influenza infection dynamics and distinguish between hosts with differing immune responses.
Article
Full-text available
Avian influenza A (H5N1) viruses cause severe disease in humans, but the basis for their virulence remains unclear. In vitro and animal studies indicate that high and disseminated viral replication is important for disease pathogenesis. Laboratory experiments suggest that virus-induced cytokine dysregulation may contribute to disease severity. To assess the relevance of these findings for human disease, we performed virological and immunological studies in 18 individuals with H5N1 and 8 individuals infected with human influenza virus subtypes. Influenza H5N1 infection in humans is characterized by high pharyngeal virus loads and frequent detection of viral RNA in rectum and blood. Viral RNA in blood was present only in fatal H5N1 cases and was associated with higher pharyngeal viral loads. We observed low peripheral blood T-lymphocyte counts and high chemokine and cytokine levels in H5N1-infected individuals, particularly in those who died, and these correlated with pharyngeal viral loads. Genetic characterization of H5N1 viruses revealed mutations in the viral polymerase complex associated with mammalian adaptation and virulence. Our observations indicate that high viral load, and the resulting intense inflammatory responses, are central to influenza H5N1 pathogenesis. The focus of clinical management should be on preventing this intense cytokine response, by early diagnosis and effective antiviral treatment.
Article
During 1997 in Hong Kong, 18 human cases of respiratory illness, including 6 fatalities, were caused by highly pathogenic avian influenza A (H5N1) viruses. Since H5 viruses had previously been isolated only from avian species, the outbreak raised questions about the ability of these viruses to cause severe disease and death in humans. To better understand the pathogenesis and immunity to these viruses, we have used the BALB/c mouse model. Four H5N1 viruses replicated equally well in the lungs of mice without prior adaptation but differed in lethality for mice. H5N1 viruses that were highly lethal for mice were detected in multiple organs, including the brain. This is the first demonstration of an influenza A virus that replicates systemically in a mammalian species and is neurotropic without prior adaptation. The mouse model was also used to evaluate a strategy of vaccination against the highly pathogenic avian H5N1 viruses, using an inactivated vaccine prepared from nonpathogenic A/Duck/Singapore-Q/F119-3/97 (H5N3) virus that was antigenically related to the human H5N1 viruses. Mice administered vaccine intramuscularly, with or without alum, were completely protected from lethal challenge with H5N1 virus. Protection from infection was also observed in 70% of animals administered vaccine alone and 100% of mice administered vaccine with alum. The protective effect of vaccination correlated with the level of virus-specific serum antibody. These results suggests a strategy of vaccine preparedness for rapid intervention in future influenza pandemics that uses antigenically related nonpathogenic viruses as vaccine candidates.
Article
Background: In 1997, highly pathogenic avian influenza A H5N1 viruses caused 18 human cases of respiratory illness and six deaths in Hong Kong. The genes of the H5N1 viruses isolated from humans were derived from avian influenza viruses, with no evidence of reassortment with human influenza viruses. The molecular determinants of human pathogenesis were not evident. Methods: The BALB/c mouse was used as a mammalian model to evaluate the biological and molecular basis of human H5N1 virus pathogenesis. Molecular correlates of H5N1 virus pathogenicity for mice were sought by analyzing the nucleotide sequence of human H5N1 viruses. Results: Based on high and low lethality for mice, the pathogenicity phenotype of 15 human H5N1 viruses was characterized; nine viruses displayed a high, five a low, and one an intermediate pathogenicity phenotype. H5N1 viruses with a high pathogenicity phenotype replicated in multiple solid organs and caused depletion of peripheral blood leukocytes. Sequence analysis determined that five specific amino acids in four proteins correlated with pathogenicity in mice. Conclusions: Alone or in combination, these specific residues are the likely determinants of virulence of human H5N1 influenza viruses in this mammalian model.
Article
Several cases of influenza-like-illness (ILI) and/or conjunctivitis in humans have been linked to an outbreak of avian influenza in poultry at a smallholding near Corwen in northern Wales in the United Kingdom (UK).
Article
METHODS Animak. Breeder mice deficient in the IL- 6 gene (IL- 6 -/-) were obtained from Dr. Valeria Poli (Istituto di Ricerche di Biologia Molecolare IRBM P. Angeletti, Rome, Italy) and were bred in our barrier facility. The generation of IL- 6 -deficient mice by gene targeting ...
Article
The influenza virus M2 protein was expressed in Xenopus laevis oocytes and shown to have an associated ion channel activity selective for monovalent ions. The anti-influenza virus drug amantadine hydrochloride significantly attenuated the inward current induced by hyperpolarization of oocyte membranes. Mutations in the M2 membrane-spanning domain that confer viral resistance to amantadine produced currents that were resistant to the drug. Analysis of the currents of these altered M2 proteins suggests that the channel pore is formed by the transmembrane domain of the M2 protein. The wild-type M2 channel was found to be regulated by pH. The wild-type M2 ion channel activity is proposed to have a pivotal role in the biology of influenza virus infection.
Article
We determined the origin and evolutionary pathways of the PB1 genes of influenza A viruses responsible for the 1957 and 1968 human pandemics and obtained information on the variable or conserved region of the PB1 protein. The evolutionary tree constructed from nucleotide sequences suggested the following: (i) the PB1 gene of the 1957 human pandemic strain, A/Singapore/1/57 (H2N2), was probably introduced from avian species and was maintained in humans until 1968; (ii) in the 1968 pandemic strain, A/NT/60/68 (H3N2), the PB1 gene was not derived from the previously circulating virus in humans but probably from another avian virus; and (iii) a current human H3N2 virus inherited the PB1 gene from an A/NT/60/68-like virus. Nucleotide sequence analysis also showed that the avian PB1 gene was introduced into pigs. Hence, transmission of the PB1 gene from avian to mammalian species is a relatively frequent event. Comparative analysis of deduced amino acid sequences disclosed highly conserved regions in PB1 proteins, which may be key structures required for PB1 activities.