ArticlePDF AvailableLiterature Review

Cytokines Induced During Influenza Virus Infection

Authors:

Abstract and Figures

Influenza A virus is one of the major human pathogens. The influenza infection can pass out without any subclinical symptoms or infestation can appear in upper respiratory tract as well as in lower respiratory tract where it can result in lethal outcome. Both innate and adaptive immune responses are activated shortly after infection providing protection against infection. Many activities of the cells of innate and adaptive immunity are coordinated by cytokines. However, inordinate or disbalanced immune response may result in overproduction of cytokines as well as chemokines what can lead to severe inflammation, including excessive recruitment of neutrophils and mononuclear cells at the site of infection. These may damage lung tissue, reduce respiratory capacity, and cause severe disease and mortality. Recently, the role of cytokines induced by virus infection has been reevaluated. While moderate inflammatory response protects against development of severe illness, the hyper-inflammatory response can elevate the disease progression. In this minireview we summarized the data on cytokines and chemokines induced in the sera of hospitalized patients infected with human and avian high pathogenic influenza viruses and define their possible role in pathogenesis. Interleukine IL-6 and chemokines CCL-2/MCP-1, CCL-4, MIP/1β, CXCL-8/IL-8, CXCL-9/MIG, and CXCL-10/IP-10 are associated with pathogenicity of both avian (H5N1 and H7N9) and human (pdmH1N1 and H3N2) viruses. Chemokines CCL-2/MCP-1, CXCL-8/IL-8, CXCL-9/MIG, and CXCL-10/IP-10 are also connected with mortality. These cytokines may be used as targets for new, more complex therapy in the extenuation of unfavorable effects of hyper inflammatory response.
Content may be subject to copyright.
Send Orders for Print-Reprints and e-prints to reprints@benthamscience.ae
Current Pharmaceutical Design, 2017, 23, 1-7 1
REVIEW ARTICLE
1381-6128/17 $58.00+.00 © 2017 Bentham Science Publishers
Cytokines Induced During Influenza Virus Infection
Tatiana Betáková1,2,*, Anna Kostrábová2, Veronika Lachová1 and Lucia Turianová1
1Biomedical Research Center - Slovak Academy of Sciences, Institute of Virology, Bratislava, Slovak Republic; 2Comenius University
in Bratislava, Faculty o f Natural Sciences, Departm ent of Microbiology and Viro logy, Bratislava, Slovak Republic
A R T I C L E H I S T O R Y
Received: January 27, 2017
Accepted: March 13, 2017
DOI:
10.2174/1381612823666170316123736
Abstract: Influenza A virus is one of the major human pathogens. The influenza infection can pass out without
any subclinical symptoms or infestation can appear in upper respiratory tract as well as in lower respiratory tract
where it can result in lethal outcome.
Both innate and adaptive immune responses are activated shortly after infection providing protection against
infection. Many activities of the cells of innate and adaptive immunity are coordinated by cytokines. However,
inordinate or disbalanced immune response may result in overproduction of cytokines as well as chemokines
which can lead to severe inflammation, including excessive recruitment of neutrophils and mononuclear cells at
the site of infection. These may damage lung tissue, reduce respiratory capacity, and cause severe disease and
mortality. Recently, the role of cytokines induced by virus infection has been reevaluated. While moderate in-
flammatory response protects against development of severe illness, the hyper-inflammatory response can elevate
the disease progression. In this mini-review, we summarized the data on cytokines and chemokines induced in the
sera of hospitalized patients infected with human and avian influenza viruses and define their possible role in
pathogenesis. Interleukin IL-6 and chemokines CCL-2/MCP-1, CCL-4/MIP-1β, CXCL-8/IL-8, CXCL-9/MIG,
and CXCL-10/IP-10 are associated with pathogenicity of both avian (H5N1 and H7N9) and human (pdmH1N1
and H3N2) viruses. Chemokines CCL-2/MCP-1, CXCL-8/IL-8, CXCL-9/MIG, and CXCL-10/IP-10 are also
related with mortality. These cytokines may be used as targets for new, more complex therapy in the extenuation
of unfavorable effects of hyper inflammatory response.
Keywords: Cytokines, chemokines, interferon, influenza virus, interleukin, immunity, pathogenesis, macrophage, NK cell, T cell, B cell.
1. INTRODUCTION
Influenza viruses cau se respirato ry infections and as widespread
pathogens endanger the human population. The severity of influ-
enza infection can range from asymptomatic infection to primary
viral pneumonia and death. Each year, about 500 million people are
infected and about 500 000 people die due to the influenza A virus
infection [1, 2]. Th e genome of viruses consists of eight RNAs with
negative polarity. Influenza A viruses are responsible for annual
epidemics and occasional pandemics. Understanding the immune
response to influenza viruses is crucial for interference during clini-
cal manifestation of influenza infection [3].
The innate immune response represents the first line of defense
against influ enza virus infection. Immune system recognizes the
infected cells by pathogen recognition receptors (PRRs) which are
present on each cell. PRPs recognize foreign molecular patterns
known as pathogen-associated molecular patterns (PAMPs). Upon
PAMP recruitment, PRRs trigger intracellular signaling cascades
inside the cell resulting in the expression of a variety of
proinflammatory molecules, which invite dendritic cells,
macrophages, neutrophils, NK cells, basophils and eosinophils to
the site of infection, induce production of int erferons to secure
antiviral activity, restriction of virus replication, spreading the
virus, and conduct the adaptive immune response [4]. The PRRs
include th e Toll-like receptors (TLR), RIG-like receptors and NOD-
like receptors. The TLRs are expressed in most cells where they are
present within endosomal compartments or on the cell surface [4].
Activation of cytosolic melanoma differentiation- associated gene 5
(MDA-5) or retinoic acid-inducible gen e I (RIG-1) leads to
*Address correspondence to this author at the Biomedical research Center,
Institute of Virology, Dubravska cesta 9, 845 05 Bratislava, Slovak
Republic; Tel: 00421-2-59 302440; Fax: 004 21-2-54774284;
E-mail: virubeta@savba.sk
interaction of the caspase activ ation and recruitment domains
(CARDs) with CARD of the mitochondrial activator of virus sig-
naling (MAVS) protein, when RLRs are the signaling adapter pro-
teins [5, 6].
CARD-CARD interactions activate TBK1 and IKKɛ protein
kinases which are responsible for phosphorylation of the transcrip-
tion factors interferon regulatory factor (IRF ) 3 and nuclear factor
κB (NF-κB). The tumor necrosis factor receptor type 1-associated
death domain (TRADD), Fas-associated protein with death domain
(FADD), receptor interacting protein 1 (RIP1), caspase-8, and
caspase-10 activate IKKα, IKKβ, and IKKγ proteins which create
the IKK comp lex, the fundamental element of th e NF-κB cascade
[4, 5, 7]. A ctivated IRF3 and NF -κB are then transported from the
cytosol into the nucleus where they turn on genes responsible for
regulation of inn ate immunity, including interferons (IFNs) an d
proinflammatory genes. IFNs activate hundreds of interferon-
stimulated genes (ISGs) and ensure antiviral, metabolic regulatory,
cell growth regulatory, and immunomodulatory actions [4, 8].
Uncontrolled inflammatory response to virus in combination
with viral virulence can lead to severe lung devastation [9, 10].
Therefore, it has been broadly suggested that disease severity de-
pends on the amount of cytokines released during a cytokine storm
in response to influenza infection [2, 11].
2. ROLE OF CYTOKINES DURING INFLUENZA VIRUS
INFECTION
Cytokines are a large group of secreted proteins with diverse
structure and functions, which regulate and coordinate many activi-
ties o f the cells of innate and adaptive immunity. Cytokine is
a general name for the group of small proteins that include lym-
phokines (cytokines made by lymphocytes), monokines (cytokines
made by monocytes), chemokines (cytokines with ch emotactic
activities), interleukins (cytokines mad e by one leukocyte and act-
2 Current Pharmaceutical Design, 2017, Vol. 23, No. 00 Betáková et al.
ing on other leukocytes), and interferons (cytokines, which interfere
with viral infection).
Viral antigens are recognized by dendritic cells (DC), macro-
phages, and T cells in subepithelial tissues which produce Th1-
polarized proinflammatory cytokines, IFNγ and tumor necro sis
factor (TNF-α). These cytokines induce production of chemokines
and other molecules responsible for host defense. The presence of
conserved NH2- terminal cysteine residues assigns chemokines into
4 groups: CC, XC, CXC and CX3C [12]. There are 28 known CC
chemokine ligands (CCL1-CCL28) which induce the migration of
monocytes, dendritic cells, T cells and other leukocytes. Monokines
induced by IFN-γ CXCL9/M IG, IFN-inducible protein CXCL10/
IP-10 and IFN-inducible T cell α-chemoatt ractant CXCL11/I-TAC
belong to the group of CXC chemokines [13-15]. The activity of
these chemokin es is closely associated with CXC chemokine recep-
tor 3 (CXCR3) located on the surface of T cells and natural killer
(NK) cells [16]. Fractalkine, the only member of CX3C group, ser-
ves as a chemoattractant as well as an adhesion molecule. The last
subfamily includes two chemokines, lymphotactin a and b,
expressed predominantly in activ ated T cells. All cells of immune
system secrete at least some cytokines with diverse structures and
functions.
2.1. Cytokines Associated with Complicated Influenza in
Humans
Moderate inflammatory response protects against development
of severe illness and hyper-inflammatory response support the
negative disease development. Many studies focusing on influenza
H1N1 pdm09 found out that disease severity was strongly associ-
ated with high level of circulating interleukin IL-6, IL-10, IFN-γ-
induced protein (CXCL10/IP-10) and monocyte chemotactic pro-
tein-1 (CCL2/MCP-1) [17]. In symptomatic individuals, the major
cytokine which undergoes the earliest increase is IL-6. Later during
infection, the level of CXCL10/IP-10 and CCL2/MCP-1 is in-
creased which suggests that these cytokines play important role in
early pathology development. Afterwards, a number of other cyto-
kines, such as T helper type I (Th1) cytokines IFN-γ, IL-15, and
chemoattractant IL-8, begin to be expressed significantly [18, 19].
Inflammation induced by influenza virus is connected to influenza
virus-associated encephalopathy (IE). IL-1β, IL-2, IL-6, IL-7, IL-8,
IL-10, IL-13, granulocyte colony stimulating factor (G-CSF),
granulocyte macrophage colony-stimulating factor (GM-CSF),
TNF-α, metallopeptidase inhibitor 1 (T IMP-1), matrix metallopep-
tidase 9 (MMP-9), sE-selectin, and neutrophil elastase were ele-
vated significantly in sera from patients with uncomplicated influ-
enza and those with IE. N eutrophil elastase, sE-selectin, IL-8, and
IL-13 were significantly increased in IE when compared with un-
complicated influenza [19]. Concentrations of IL-6, IL-10, IL-15,
CXCL8/IL-8, CXCL9/MIG, CXCL10/IP-10, IL-2R, hepatocyte
growth factor (HGF), ST2, CCL2/MCP-1, CCL4/MIP, and soluble
tumor necrosis factor receptors 1 (sTNFR-1) were significantly
higher in patients with severe influenza H1N1 pdm09 virus infec-
tion [18]. Distinct changes for IFN-α2, CCL7/MCP-3, IL-6, vascu-
lar endothelial growth factor A (VEGFA) in nasal washes and
CCL7/MCP-3, IL-10, and CXCL10/IP-10 in peripheral blood of
human patients correlated significantly with disease severity and
did not depend on the virus load [20]. Elevated proinflammatory
cytokines, particularly IL-6, predicted ICU admission and corre-
lated with fever, tachypnoea, deoxygenation, and length of stay
during influenza infections [17, 21, 22].
Elevated levels of CXCL10/IP-10, IL-6, IL-17, and IL-2 were
detected in sera of 16 hospital-admi tted patients infected with
H7N9 virus [23, 24]. Recent studies confirm correlation between
levels of cytokines in serum: macrophage migration inhibitory fac-
tor (MIF), stem cell factor (SCF), CCL2/MCP-1, CCL2/MCP-1,
HGF, and mast/stem cell growth factor beta (SCGF-β) and disease
severity after H7N9 infection [25]. The increased expression of
proteins associated with the recruitment and activation of memory
T cells, such as CXCL11/I-TAC, CXCL10/IP-10, IL-16 and granu-
lysin (GNLY) was detected in n asal washes [14, 26-29].
The levels of CXCL10/IP-10, IL-2, IL-6, IL-17 were elevated
in the individuals infected with influenza H7N9 [23, 30].
High levels of inflammatory cytokines in the peripheral blood,
including CXCL10/IP-10, CCL2/MCP-1, monokine induced by
IFN-γ (CXCL9/MIG) and CXCL8/IL-8 were detected in patients
who died due to avian influenza A virus (H5N1) infection. Other
research findings suggest that modulation of the inflammatory
pathway can be used in adjunctive therapies [3, 31-33]. Influenza
infection with H5N1 virus induces activation of chemoattractants
CXCL10/IP-10, CCL2/MCP-1, CCL4/MIP-1β, CXCL9/MIG, and
CXCL8/IL-8, which are driven by adaptive and innate responses.
The induction of certain proinflamm atory cytokines, such as TNF-α
and chemoattractans CXCL10/IP-10, CCL4/MIP-1β, CCL2/MCP-
1, and CXCL9/MIG is closely bound with pathogenesis of severe
H5N1 and H3N2 influenza infections [31, 32, 34, 35].
Expression of mitogen-activated protein kinases (MAPKs) and
c-Jun N-terminal kinases (JNKs), which play a role in the immune
response to avian and human influenza infection, was reduced in
sera from influenza infected patients versu s healthy controls [24].
2.2. Type I and Type III Interferons Determine the Course of
Infection
Anti-viral protection in mammals is substantially supported by
type I and type III IFNs. Both IFN-α/β and IFN-λs signal through
their receptors and activate the same pathway which results in the
formation of a ternary transcription complex: STAT1, STAT2 and
IFN regulatory factor-9 (IRF9), known as IFN-stimulated gen e
factor 3 (ISGF3) [36-38]. The type I IFNs receptor complex con-
sists of two chains, IFNAR1 (IFN-αR1) and CD118 (IFN-αR2),
type III IFNs heterodimeric receptor contains unique IFNLR1 (IFN-
λR1) and CD210B (IL10R2) chains [36, 39, 40]. The CD210B
serves as the second subunit for IL-10, IL- 22, and IL-26 [36]. The
expression of IFN-λ receptors is restricted to a subset of cells, pri-
marily epithelial cells, keratinocytes, some dendritic cells and neu-
trophil population [38, 41-45]. Activity of type I and type III IFNs
differs in rapidity and specificity. Type I IFNs induce greater and
faster response than type III IFNs. On the other hand, activity of
type III IFNs emerges later, but induction is more permanent [46-
48].
Type I IFNs activate ISGs promoters by binding of IRF and
other transcription factors to interferonstimulated response ele-
ment (ISRE). Antiviral signaling of type III IFNs is connected to
activation of NF-κB and MAPK [49]. The type III IFNs rely
strongly upon both p38 and JNK MAP kinases for gene induction
[46, 50]. Transcription factor Med23 selectively upregulates type
III but not type I IFN signaling, and type III IFNs are specifically
downregulated by ZEB1 [49, 51,52].
The main antiviral action of IFNs lies in inhibition of viral rep-
lication and induction of the immune response elicited by viruses
[53-59]. However, recently, it has been shown that IFNs cause im-
munopathology in some acute influenza virus infections. Type I
IFNs appear to influence the role of several immune functions dur-
ing superinfection. These IFNs have been implicated in the d evel-
opment of Staphylococcus aureus and Streptococcus pneumoniae
superinfection [60, 61]. The IFNs are associated with suppression
of Th17 and production of peptides with antimicrobial activities,
which increase risk for both gram-positive and gram-negative bac-
terial coinfection during influenza infection [61]. The activity of
type I IFNs results in decreased amount of CCL2/MCP-1 and inhi-
bition of IL-17 [62-64]. It seems that the proinflammatory response
initiated by RIG-1 and TLR7 plays positive role in influenza virus
replication. After TLR7 and RIG-1 signaling, monocyte derived
DCs are accumulated in the lungs where they can be infected by the
Cytokines Induced during Influenza Virus Infectio n Current Pharmaceutical Design, 2017, Vol. 23, No. 00 3
virus [65]. The type III IFNs facilitate infection of lower respiratory
tract. In a model of post-influenza acute Staphylococcus aureus
lung infection, wild type mice infected with influenza were more
susceptible to bacterial infection due to production of type III IFNs
in the lungs [66].
2.3. The IFN-γ as a Major Player of Innate and Adaptive
Immunity
The type II IFN (IFN-γ) is expressed by certain cell types,
namely NK, T cells and natural killer T cells (NKTs). The biologi-
cal activity of IFN-γ is induced after binding to its receptor, which
consist s of two ligand binding CD119 (IFN-γR1) chains and two
signal-transducing IFNGR2 (IFN-γR2) chains. The activation of
this signaling pathway involves the binding of IFN -γ to form the
IFN-γR complex [67]. Each IFN-γR subunit binds to JAK1 and
JAK2 complex, where N-terminal domains are activated by tyrosine
phosphorylation [67-69]. Activated JAK1 and JAK2 are responsible
for phosphorylation of the IFN-γR tails. The phosphorylated recep-
tor binds the STAT1 monomers via their src-homology 2 domains.
After phosphorylation by JAKs, STAT1 monomer dissociates from
the receptor and as STAT1:STAT1 homodimer is transported to the
nucleus. Binding of the STA1:STAT1 homodimer with γ-activated
sequence (GAS) elements activates the promoters of IFN-γ respon-
sive genes [68]. IFN-γ can also stimulate STAT3 and STAT5 [70,
71].
The IFN-γ induces the expression of: i) proteins with antiviral
activities (PKR, OAS, and Mx GTPase); ii) enzyme which impairs
translation of viral proteins: dsRNA-specific adenosine deaminase
(ADAR); iii) type I IFNs; and iv) proinflammatory cytokines and
chemokines by endothelial cells and fibroblasts which invite
macrophages, neutrophils and T cells to the site of infection [72].
IFN-γ affects the local cellular response in the respiratory tract as
well as the systemic humoral response to influenza virus infection
[73]. However, IFN-γ deficiency does not influence the elimination
of influenza virus, suggesting that IFN-γ may primarily function to
modulate severity of disease caused by influenza infection [74].
Influenza infection results in serious lung damage facilitated by
CD8+ T cell effector functions. Recently, it was identified that IFN-
γ signaling and STAT1-independen t IFN-γ signaling have impor-
tant roles in regulating CD8+ T cell-mediated acute lung injury
[75]. The IFN-γ regulates local immunity against virus infection by
reducing antigen-specific CD8+ T cells and inhibiting memory pre-
cursor form ation. By this mechanism, IFN-γ reduces th e population
of the memory cells after an influenza virus infection [76]. The
relatively high levels of IFN-γ in the lung following influenza
caused inhibition of MARCO expression, which is essential for
phagocytosis of bacteria. Thus, alveolar macrophagemediated
clearance of S. pneumoniae is significantly inhibited by prior influ-
enza virus infection [77].
CONCLUSION
The exact functions of cytokines in immune response to influ-
enza virus infection are yet to be determined. Recent data proved
that the cytokine levels correlate with disease severity. High ex-
pression of cytokines does not need to depend on virus load [20].
Here, we briefly reviewed the overexpression of cytokines which is
associated with disease severity and mo rtality. Table 1 summarizes
the available data on the principal cell source and cellular targets
and biological effects of these cytokines during common infection
with influenza virus.
We are aware that this table does not provide comprehensive
information about all cytokines enhanced during influenza infec-
tion. The human’s experiments with in fluen za viruses are restricted
and most of the available data were obtained from the patients and
from post mortal analyses. In most studies, the age of patients is
missing. Nevertheless, the level of some cytokines were raised in
children as well as in adults and increased level of these cytokines
always correlated with disease severity [3, 9, 19]. Interleukin IL-6
and chemokines CCL-2/MCP-1, CCL-4/MIP-1β, CXCL-8/IL-8,
CXCL-9/MIG, and CXCL-10/IP-10 are associated with pathogenic-
ity of avian and human viruses (Fig. 1). Interestingly, chemokines
CCL-2/MCP-1, CXCL-8/IL-8, CXCL-9/MIG, and CXCL-10/IP-10
are connected with mortality and these chemokines regulate T cell,
macrophages, neutrophils, basophils and NK cells function and
recruitm ent (Table 1). Expression of these cytokines can be mis-
regulated also by the influenza viruses. The multifunctional non-
structural protein 1 (NS1) suppresses the antiviral host defence by
affecting interferon responses and enhances virus pathogenesis
[78]. Virus replication, pathogenicity and innate immune response
can also be influenced by PB1-F2 and PB2 proteins [79-82]. The
PB1-F2 protein inhibits induction of type I IFNs and NK cells and
influences neutrophil recruitment resulting in increased viral patho-
genesis [83, 84]. The role of viral proteins in immune response is
started to be extensively studied.
Table 1. Selected cytokines associated with severe infection and their primary activities. Cytokines whose overproduction is linked to
mortality are in bold.
Cytokine
Principal Cell source
Cellular targets and biologic effects
Negative role during influenza infection
IL-2
T cells
induces expansion of antigen-specific clones of T
cells; augments cytokine production by T cells and
NK cells; enhances antibody secretion by B cells
H1N1 pdm09 - associated with d isease severity [2] and
encephalopathy [19];
H7N9 - associated with d isease severity [23,24];
IL-6
macrophages, endothe-
lial cells, T cells, B cells
proliferation of antibody producing cells; develop-
ment or inhibition of defined effector T cell popula-
tions;
synthesis of acute-phase proteins
H1N1 pdm09 - associated with d isease severity
[17,18,20] and encephalopathy [19]; pathology devel-
opment [18]; correlates with fever, tachypnoe and
deoxygenation [17,21,22]
H7N9 - associated with d isease severity [23,24];
IL-10
macrophages, mast
cells, B cells, T cells
(regulatory)
downregulates expression of class II MHC and
costimulatory molecules on macrophages and den-
dritic cells;
stimulatory to mast cells and T cells; promotes B
cell maturation
H1N1 pdm09 - associated with d isease severity [17,18,
20] and encephalopathy [19];
4 Current Pharmaceutical Design, 2017, Vol. 23, No. 00 Betáková et al.
(Table 1) Contd....
Cytokine
Principal Cell source
Negative role during influenza infection
IL-13
CD4+ T cells (TH2),
NKT cells, mast cells,
group 2 innate lymphoid
cells
H1N1 pdm09 - associated with encephalopa-
thy [19];
IL-15
macrophages, dendritic
cells,
other cell types
H1N1 pdm09 - associated with d isease sever-
ity [18]
IL-16
CD4+ and CD8+ cells,
epithelial cells
H7N9 - associated with d isease severity [14,
26-29];
IL-17
T cells
H7N9 - associated with d isease severity
[23,24];
SCF
(MGF)
stem cells
H7N9 - associated with disease severity [25]
HGF
stromal cells,
mesenchymal cells
H1N1 pdm09 - associated with d isease sever-
ity [18];
H7N9 - associated with d isease severity [25]
SCGF-β
mast cells,
stem cells
H7N9 - associated with d isease severity [25]
MIF
macrophages, T cells,
dendritic cells, endothe-
lial cells, eosinophils
H7N9 - associated with d isease severity [25]
IFN-α/β
plasmacytoid dendritic
cells,
macrophages, endothe-
lial cells, NK cells, B
and T cells, fibroblasts
H1N1 pdm09 - associated with d isease sever-
ity [20];
suppression of Th17 [61];
decreasing of CCL2 and IL-17 [62-64]
IFN-γ
T cells (TH1, CD8+ T
cells)
NK cells
reduction of CD8+ T cells in lung [75];
reduction of the memory cells population
[76];
inhibition of MARCO expression [77]
IFN-λ
dendritic cells
facilitate infection of lower respiratory tract
by bacteria [66]
TNF-α
macrophages, CD4+ T
cells, NK cells, neutro-
phils, mast cells, eosi-
nophils, neurons
H1N1 pdm09 - associated with encephalopa-
thy [19]
CCL2
(MCP-1)
monocytes, macro-
phages, dendritic cells
H1N1 pdm09 - associated with d isease sever-
ity [17,18]
H3N2 - associated with p athogenicity [31, 32,
34, 35]
H7N9 - associated with d isease severity [25];
H5N1 associated with p athogenicity [31, 32,
34, 35] and mortality [3, 31-33]
Cytokines Induced during Influenza Virus Infectio n Current Pharmaceutical Design, 2017, Vol. 23, No. 00 5
(Table 1) Contd....
Cytokine
Principal Cell source
Cellular targets and biologic effects
Negative role during influenza infection
CCL4
(MIP-1β)
macrophages,
CD8+ T cells
T cells, dendritic cells, monocytes and NK cells
recruitment
H1N1 pdm09 - associated with disease severity [18];
H3N2- associated with pathogenicity [31,32,34,35];
H5N1 - associated with p athogenicity [31,32,34,35]
CCL7
(MCP-3)
macrophages, tumor cell
lines
mixed leukocyte recruitment;
regulates acute neutrophilic lung inflammation
H1N1 pdm09 - associated with d isease severity [20]
CXCL8
(IL-8)
macrophages, epithelial
cells,
endothelial cells
mediator of inflammatory response; attracts neutro-
phils, basophils, and T-cells;
regulates macrophages, endothelial cells and mast
cells function
H1N1 pdm09 - associated with d isease severity [18]
and encephalopathy [19];
H5N1 mortality [3, 31-33]
CXCL9
(MIG)
alveolar macrophages
IFN-induced T cell chemoattractant; affects the
growth, movement or activation state of cells that
participate in immune and inflammatory response
H1N1 pdm09 - associated with d isease severity [18];
H3N2 - associated with p athogenicity [31,32,34,35]
H5N1 associated with p athogenicity [31,32,34,35]
and mortality [3,31-33]
CXCL10
(IP-10)
alveolar macrophages,
monocytes, endothelial
cells, fibroblasts, other
cell types
effector T cell recruitment; chemotactic for T cells,
macrophages and dendritic cells;
stimulation of monocytes, NK cells and T cell mi-
gration, and modulation of adhesion molecule ex-
pression
H1N1 pdm09 - associated with d isease severity [17,
18, 20]; pathology developm ent [18,19];
H3N2 - associated with p athogenicity [31, 32, 34, 35]
H7N9 - associated with d isease severity [14, 23, 24,
26-29];
H5N1 associated with p athogenicity [31,32,34,35]
and mortality [3,31-33]
CXCL11
epithelial cells, thyro-
cytes, peripheral blood
leukocytes
Recruits T cells, NK cells and macrophages to the
site of inflammation
H7N9 - associated with d isease severity [14, 26-29]
Fig. (1). Cytokine associated with severity and mortality of both avian and human influenza viruses. Several different cell types coordinate their effort as
part of immune system during influenza infection. Macrophages, dendritic cells, B cells, T cells NK cells, endothelial cells, mast cells, neutrophils, basophils
and eosinophils have distinct roles in immune system using secreted cytokines.
6 Current Pharmaceutical Design, 2017, Vol. 23, No. 00 Betáková et al.
Better understanding of the interaction between virus, tissue
and immune response may help in better diagnostic and prognoses
during severe acute influenza infection. One of the possible future
approaches to extenuate the unfavorable effects of hyper inflamma-
tory response may also focus on targeted control or reduction of the
level of cytokines which are linked to mortality.
CONFLICT OF INTEREST
The authors confirm that this article content has no conflict of
interest.
ACKNOWLEDGEMENTS
This research was supported by the Slovak Research and De-
velopment Agency, grant No. APVV-0676-12, by the Scientific
Grant Agency of the Slovak Republic VEGA No. 2/0014/16.
REFERECES
[1] Fauci AS. Seasonal and pandemic influenza preparedness: science
and countermeasures. J Infect Dis 2006; 194: S73-6.
[2] Marion T, Elbahesh H, Thomas PG et al. Respiratory Mucosal Pro-
teome Quantification in Human Influenz a Infections. PLoS One
2016; 11(4): e0153674.
[3] McClain MT, Henao R, Williams J et al. Differential evolution of
peripheral cytokine levels in symptomatic and asymptomatic re-
sponses to experimental influenza virus challenge. Clin Exp Immu-
nol 2016; 183: 441-51.
[4] Kell AM, Gale M Jr. RIG-I in RNA virus recognition. Virology
2015; 479-480: 110-21.
[5] Kawai T, Takahash i K, Sato S et al. IPS-1, an adaptor triggering
RIG-I- and Mda5-mediated type I interferon induction. Nat Immu-
nol 2005; 6: 981-8.
[6] Xu LG, Wang YY, Han KJ, Li LY, Zhai Z, Shu HB. VISA is an
adapter protein required for virus-triggered IFN-beta signaling. Mol
Cell 2005; 19: 727-40.
[7] Takahashi K, Kawai T, Kumar H, Sato S, Yonehara S, Akira S.
Roles of caspase-8 and caspase-10 in innate immune responses to
double-stranded RNA.J Immunol 2006; 176: 4520-4.
[8] Chiang JJ, Davis ME, Gack MU. Regulation of RIG-I-like receptor
signaling by host and viral proteins. Cytokine Growth Factor Rev
2014; 25: 491-505.
[9] Kuiken T, Riteau B, Fouchier RA, Rimmelzwaan GF. Pathogenesis
of influenza virus infections: the good, the bad and the ugly. Curr
Opin Virol 2012; 2: 276-86.
[10] Kumar Y, Liang C, Limmon GV, et al. Molecular analysis of serum
and bronchoalveolar lavage in a mouse model of influenza reveals
markers of disease severity that can be clinically useful in humans.
PLoS One 2014; 9: e86912.
[11] Tisoncik JR, Korth MJ, Simmons CP, Farrar J, Martin TR, Katze
MG. Into the eye of the cytokine storm. Microbiol Mol Biol Rev
2012; 76: 16-32.
[12] Abbas AK, Lichtman A H, Pillai S. Cellular and molecular
Immmnology 8th ed. Elsevier, 2 015.
[13] Liao F, Rabin RL, Yannelli JR, Koniaris LG, Vanguri P, Farber JM.
Human Mig chemokine: biochemical and functional characteriza-
tion. J Exp Med 1995; 182: 1301-14.
[14] Cole KE, Strick CA, Paradis TJ, et al. Interferon-indu cible T cell
alpha chemoattractant (I-TAC): a novel non-ELR CXC chemokine
with potent activity on activated T cells through selective high affin-
ity binding to CXCR3. J Exp Med 1998; 187: 2009-21.
[15] Luster AD. The role of chemokines in linking innate and adaptive
immunity. Curr Opin Immunol 2002; 14: 129-35.
[16] Egesten A, Eliasson M, Johansson HM, et al. The CXC chemokine
MIG/CXCL9 is important in innate immunity against Streptococcus
pyogenes. J Infect Dis 2007; 195: 684-93.
[17] Davey RT Jr., Lynfield R, Dwyer DE, et al. The association be-
tween serum biomarkers and disease outcome in influenza A
(H1N1)pdm09 virus infection: results of two international observa-
tional coh ort studies. PLo S One 2013; 8: e57121.
[18] Bradley-Stewart A, Jolly L, Adamson W, et al. Cytokine responses
in patients with mild or severe influenza A(H1N1)pdm09. J Clin Vi-
rol 2013; 58: 100-7.
[19] Sun G, Ota C, Kitaoka S, et al. Elevated serum levels of neutrophil
elastase in patients with in fluen za virus-associated encephalopathy.
J Neurol Sci 2015; 349: 190-5.
[20] Oshansky CM, Gar tland AJ, Wong SS, et al. Mucosal immune
responses predict clinical outcomes during influenza infection inde-
pendently of age and viral load. Am J Respir Crit Care Med 2014;
189: 449-62.
[21] Lee N, Wong CK, Chan PK, et al. Cytokine response patterns in
severe pandemic 2009 H1N1 and seasonal influenza among hospi-
talized adults. PLoS One 2011; 6: e26050.
[22] Yang ZF, Mok CK, Liu XQ, et al. Clinical, virological and immu-
nological features from patients infected with re-emergent avian-
origin human H7N9 influenza disease of varying severity in Guang-
dong province. PLoS One 2015; 10: e0117846.
[23] Chi Y, Zhu Y, Wen T, et al. Cytokine and chemokine levels in
patients infected with the novel avian influenza A (H7N9) virus in
China. J Infect Dis 2013; 208: 1962-7.
[24] Marion T, Elbahesh H, Thomas PG, DeVincenzo JP, Webby R,
Schughart K. Respiratory Mucosal Proteome Quantification in Hu-
man Influenza Infections. PLoS One 2016; 11: e0153674.
[25] Crowley JJ, Zhabotynsky V, Sun W, et al. Analyses of allele-
specific gene expression in highly divergent mouse crosses identi-
fies pervasive allelic imbalance. Nat Genet 2015; 47: 3 53-60.
[26] Qin S, Rottman JB, Myers P, et al. The chemokine receptors
CXCR3 and CCR5 mark subsets of T cells associated with certain
inflammatory reactions. J Clin Invest 1998; 101: 746-54.
[27] Sallusto F, Lanzavecchia A, Mackay CR. Chemokines and
chemokine receptors in T-cell priming and Th1/Th2-mediated re-
sponses. Immunol Today 1998; 19: 568-74.
[28] Cruikshank WW, Long A, Tarpy RE, et al. Early identification of
interleukin-16 (lymphocyte chemoattractant factor) and macrophage
inflammatory protein 1 alpha (MIP1 alpha) in bronchoalveolar lav-
age fluid of antigen-challenged asthmatics. Am J Respir Cell Mol
Biol 1995; 13: 738-47.
[29] Deng A, Chen S, Li Q, Lyu SC, Clayberger C, Krensky AM. Granu-
lysin, a cytolytic molecule, is also a chemoattractant and proinflam-
matory activator. J Immunol 2005; 174: 5243-8.
[30] Wang Z, Zhang A, Wan Y, et al. Early hypercytokinemia is associ-
ated with interferon-induced transmembrane protein-3 dysfunction
and predictive of fatal H7N9 infection. Proc Natl Acad Sci USA
2014; 111: 769-74.
[31] Peiris JS, Yu WC, Leung CW, et al. Re-emergence of fatal human
influenza A subtype H5N1 disease. Lancet 2004; 363: 617-9.
[32] Chan MC, Cheung CY, Chui WH, et al. Pro inflammatory cytokine
responses induced by influenza A (H5N1) viruses in primary human
alveolar and bronchial epithelial cells. Respir Res 2005; 6: 135.
[33] Walsh KB, Teijaro JR, Rosen H, Oldstone MB. Quelling the storm:
utilization of sphingosine-1-phosphate receptor signaling to amelio-
rate influenza virus-induced cytokine storm. Immunol Res 2011; 51:
15-25.
[34] Monteerarat Y, Sakabe S, Ngamurulert S, et al. Induction of TNF-
alpha in human macrophages by avian and human influenza viruses.
Arch Virol 2010; 155: 1273-9.
[35] Aoyagi T, Newstead MW, Zeng X, Kunkel SL, Kaku M, Standiford
TJ. IL-36 receptor deletion attenuates lung injury and decreases
mortality in murine influenza pneumonia. Muco sal Immunol 2016.
doi: 10.1038/mi.2016.107.
[36] Kotenko SV, Gallagher G, Baurin VV, et al. IFN-lambdas mediate
antiviral protection through a distinct class II cytokine receptor
complex. Nat Immunol. 2003; 4: 69-77.
[37] Donnelly RP, Kotenko SV. Interferon-lambda: a new addition to an
old family. J Interferon Cytokine Res 2010; 30: 555-64.
[38] Lin JD, Feng N, Sen A, Balan M, et al. Distinct Roles of Type I and
Type III Interferons in Intestinal Immunity to Homologous and Het-
erologous Rotavirus Infections. PLoS Pathog 2016; 12: e1005600.
[39] Uzé G, Lutfalla G, Gresser I. Genetic transfer of a functional human
interferon alpha receptor into mouse cells: cloning and expression of
its cDNA. Cell 1990; 60: 225-34.
[40] Cleary CM, Donnelly RJ, Soh J, Mariano TM, Pestka S. Knockout
and reconstitution of a functional human type I interferon receptor
complex. J Biol Chem 1994; 269: 18747-9.
[41] Sommereyns C, Paul S, Staeheli P, Michiels T. IFN-lambda (IFN-
lambda) is expressed in a tissue-dependent fashion and primarily
acts on epithelial cells in vivo. PLoS Pathog 2008; 4: e1000017.
Cytokines Induced during Influenza Virus Infectio n Current Pharmaceutical Design, 2017, Vol. 23, No. 00 7
[42] Cohen TS, Prince AS. Bacterial pathogens activate a common in-
flammatory pathway through IFNλ regulation of PDCD4. PLoS
Pathog 2013; 9: e1003682.
[43] Blazek K, Eames HL, Weiss M, et al. IFN-λ resolves inflammation
via suppression of neutrophil infiltration and IL-1β production. J
Exp Med 2015; 212: 845-53.
[44] Lukacikova L, Oveckova I, Betakova T, et al. Antiviral Effect of
Interferon Lambda Against Lymphocytic Choriomeningitis Virus. J
Interferon Cytokine Res 2015; 35: 540-53.
[45] Mahlakõiv T, Hernandez P, Gronke K, Diefenbach A, Staeheli P.
Leukocyte-derived IFN-α/β and epithelial IFN-λ constitute a com-
partmentalized mucosal defense system that restricts enteric virus
infections. PLoS Pathog 2015; 11, e1004782.
[46] Zhou Z, Hamming OJ, Ank N, Paludan SR, Nielsen AL, Hartmann
R. Type III interferon (IFN) induces a type I IFN-like response in a
restricted subset of cells through signaling pathways involving both
the Jak-STAT pathway and the mitogen-activated protein kinases. J
Virol 2007; 81: 7749-58.
[47] Bolen CR, Ding S, Ro bek MD, Kleinstein SH. Dynamic expression
profiling of type I and type III interferon-stimulated hepatocytes re-
veals a stable hierarchy of gene expression. Hepatology 2014; 59:
1262-72.
[48] Jilg N, Lin W, Hong J, et al. Kinetic differences in the induction of
interferon stimulated genes by interferon-α and interleukin 28B are
altered by infection with hepatitis C virus. Hepatology 2014; 59:
1250-61.
[49] Cohen TS, Parker D. Microbial pathogenesis and type III interfer-
ons. Cytokine Growth Factor Rev 2016; 29: 45-51.
[50] Cohen TS, Parker D. Microbial pathogenesis and type III interfer-
ons. Cytokine Growth Factor Rev 2016; 29: 45-51.
[51] Griffiths SJ, Koegl M, Boutell C, et al. A systematic analysis of host
factors reveals a Med23-interferon-λ regulatory axis against herpes
simplex virus type 1 replication. PLoS Pathog. 2013; 9: e1003514.
[52] Swider A, Siegel R, Eskdale J, Gallagher G. Regulation of inter-
feron lambda-1 (IFNL1/IFN-λ1/IL-29) expression in human colon
epithelial cells. Cytokine. 2014; 65: 17-23.
[53] Svetlik ova D, Kabat P, Ohradanova A, Pastorek J, Betakova T.
Influenza A virus replication is inhibited in IFN-λ2 and IFN-λ3
transfected or stimulated cells. Antiviral Res 2010; 88 : 329-33.
[54] Pothlichet J, Meunier I, Davis BK, et al. Type I IFN triggers RIG-
I/TLR3/NLRP3-dependent inflammasome activation in influenza A
virus infected cells. PLoS Pathog 2013; 9 : e1003256.
[55] Bennett AL, Smith DW, Cummins MJ, Jacoby PA, Cummins JM,
Beilharz MW. Low-dose oral interferon alpha as prophylaxis against
viral respiratory illness: a double-blind, parallel controlled trial dur-
ing an influenza pandemic year. Influenza Other Respir Viruses
2013; 7: 854-62.
[56] Svancarova P, Svetlikova D, Betakov a T. Induction of interferon
lambda in influenza A virus infected cells treated with shRNAs
against M1 transcript. Acta Virol 2015; 59: 148-55.
[57] Škorvanová L, Švančarová P, Svetlíková D, Betáková T. Protective
efficacy of IFN-ω AND IFN-λs against influenza viruses in induced
A549 cells. Acta Virol 2015; 59: 413-7.
[58] Svancarova P, Svetlikova D, Betakova T. Synergic and antagonistic
effect of small hairpin RNAs targeting the NS gene of the influenza
A virus in cells and mice. Virus Research 2015; 195: 100-11.
[59] Kim S, Kim MJ, Kim CH, et al. The Superiority of IFN-lambda as a
Therapeutic Candidate to Control Acute Influenza Viral Lung Infec-
tion. Am J Respir Cell Mol Biol 2016 Sep 15.
[60] Shahangian A, Ch ow EK, Tian X, et al. Type I IFNs mediate devel-
opment of postinfluenza bacterial pneumonia in mice J Clin Invest
2009; 119: 1910-1920.
[61] Lee B, Robinson KM, McHugh KJ, et al. Influenza-induced type I
interferon enhances susceptibility to gram-negative and gram-
positive bacterial pneumonia in mice. Am J Physiol Lung Cell Mol
Physiol 2015; 309: L158-67.
[62] S. Nakamura, K.M. Davis, J.N. Weiser Synergistic stimulation of
type I interferons during influenza virus coinfection promotes Strep-
tococcus pneumoniae colonization in mice. J Clin Invest 2011; 121:
3657-65
[63] Li W, Moltedo B, Moran TM. Type I interferon induction during
influenza virus infection increases susceptibility to secondary Strep-
tococcus pneumoniae infection by negative regulation of
gammadelta T cells. J Virol 2012; 86 : 12304-12.
[64] Robinson KM, Lee B, Scheller EV, et al. The role of IL-27 in sus-
ceptibility to post-influenza Staphylococcus aureus pneumonia.
Respir Res 2015; 16: 10.
[65] Pang IK, Pillai PS, Iwasaki A. Efficient influenza A virus replication
in the respiratory tract requires signals from TLR7 and RIG-I. Proc
Natl Acad Sci U S A 2013; 110: 13910-5.
[66] Planet PJ, Parker D, Cohen TS, et al. Interferon-lambda restructures
the nasal microbio me and increases susceptibility to staphylococcus
aureus superinfection. mBio 2016; 7: e01939-115.
[67] van Boxel-Dezaire AH, Stark GR. Cell type-specific signaling in
response to interferon-gamma Curr Top Microbiol Immunol 2007;
316: 119-54.
[68] Igarashin K, Garotta G, Ozmen L, et al. Interferon-gamma induces
tyrosine phosphorylation of interferon-gamma recepto r and regu-
lated association of protein tyrosine kinases, Jak1 and Jak2, with its
receptor. J Biol Chem 1994; 269: 14333-6.
[69] Levy DE, Darnell JE Jr. Stats: transcriptional control and biological
impact Nat Rev Mol Cell Biol 2002; 3: 651-62.
[70] Eilers A, Decker T. Activity of Stat family transcription factors is
developmentally controlled in cells of the macrophage lineage. Im-
munobiology 1995; 193: 328-33.
[71] Kotenko SV, Pestka S. Jak-Stat signal transd uction pathway through
the eyes of cytokine class II receptor complexes. Oncogene 2000;
19: 2557-65.
[72] Roff SR, Noon-Song EN, Yamamoto JK. The Significance of Inter-
feron-γ in HIV-1 Pathogenesis, Therapy, and Prophylaxis. Front
Immunol 2014; 4: 498 .
[73] Baumgarth N, Kelso A. In vivo blockade of gamma interferon af-
fects the influenza virus-induced humoral and the local cellular im-
mune response in lung tissue. J Virol 1996; 70: 4411-8.
[74] Graham MB, Dalton DK, Giltinan D, Braciale VL , Stewart TA,
Braciale TJ. Response to influenza infection in mice with a targeted
disruption in the interferon-γ gene. J Exp Med 1993; 178: 1725-32.
[75] Ramana CV, DeBerge MP, Kumar A, Alia CS, Durbin JE, Enelow
RI. Inflammatory impact of IFN-γ in CD8+ T cell-mediated lung in-
jury is mediated by both Stat1-dependent and -independent path-
ways. Am J Physiol Lung Cell Mol Physiol. 2015; 308: L650-7.
[76] Prabhu N, Ho AW, Wong KH, et al. Gamma interferon regulates
contraction of the influenza virus-specific CD8 T cell response and
limits the size of the memory population. J Viro l. 2013; 87: 12510-
22.
[77] Sun K, Metzger DW. Inh ibition of pulmonary antibacterial defense
by interferon-gamma during recovery from influenza infection. Nat.
Med 2008; 14: 558-4.
[78] Gack MU, Albrecht RA, Urano T, et al. Influenza A virus NS1
targets the ubiquitin ligase TRIM25 to evade recognition by RIG-I.
Cell Host Microbe 2009; 5: 439-449.
[79] Lee J, Henningson J, Ma J, et al. Effects of PB1-F2 on the patho-
genicity of H1N1 swine influenza virus in mice and pigs. J Gen Vi-
rol 2016 doi: 10.1099/jgv.0.000695.
[80] Wang C, Lee HH, Yang ZF, Mok CK, Zhang Z. PB2-Q591K Muta-
tion Determines the Pathogenic ity of Avian H9N2 Influenza Viruses
for Mammalian Species. PLoS One 2016; 11: e0162163.
[81] Prokopyeva EA, Romanovskaya AA, Sharshov KA, et al. Patho-
genicity assessment of wild-type and mouse-adapted influenza
A(H1N1)pdm09 viruses in comparison with highly pathogenic in-
fluenza A(H5N1) virus. Histol Histopathol 2017 Jan 13: 11866. doi:
10.14670/HH-11-866.
[82] Hu M, Yuan S, Zhang K, et al. PB2 substitutions V598T/I increase
the virulence of H7N9 influenza A virus in mammals. Virology
2017; 501: 92-101.
[83] Varga ZT, Ramos I, Hai R et al. The influenza virus protein PB1-F2
inhibits the induction of type I interferon at the level of the MAVS
adaptor protein. PLoS Pathog 2011; 7: e1002067.
[84] Vidy A, Maisonnasse P, Da Costa B, et al. The Influenza Virus
Protein PB1-F2 Increases Viral Pathogenesis through Neutrophil
Recruitment and NK Cells Inhibition. PLoS One 2016; 11:
e0165361.
... Zhang Y et al. [25] found that IL-10 and IFN-γ may be good predictors for RMPP in school-age children. Studies have proposed that cytokines can be used as new therapeutic targets to reduce the damage caused by excessive inflammatory response to the body [26]. Most of these biomarkers (including CCL2) are still under study or are not widely available in clinical practice. ...
... CCL2 is not only a specific parameter of MPP, it is usually an anti-inflammatory value. Betakova T et al. [26] summarized the data of induced cytokines and chemokines in the serum of patients infected with human and avian influenza virus, and determined that they may play a role in the pathogenesis. Chemokines CCL-2, CXCL-8, CXCL-9 and CXCL-10 are related to the pathogenicity and mortality of avian influenza virus. ...
Article
Full-text available
Background There are relatively few studies investigating C-C motif chemokine ligand 2 (CCL2) level in bronchoalveolar lavage fluid (BALF) in children with Mycoplasma pneumoniae pneumonia (MPP), and the relationship between CCL2 level in BALF and refractory mycoplasma pneumoniae pneumonia (RMPP) is unclear. This study aims to explore the relationship between chemokine CCL2 level in BALF and clinical characteristics and clinical outcome in children with MPP. Methods A total of 51 children with confirmed acute MPP and requiring bronchoalveolar lavage in Department of Pediatrics, Huanghe Sanmenxia Hospital and The First Clinical College of Xinxiang Medical University from October 2021 to February 2023 were selected as the study group. And 11 children with bronchial foreign body were selected as the control group. The study group was divided into the non-refractory mycoplasma pneumoniae pneumonia (NRMPP) group and the RMPP group based on the response to treatment. BALF and clinical data of the patients were collected. And CCL2 levels were tested in the patients. Differences in CCL2 level in BALF and clinical characteristics were tested and compared. Results The CCL2 level in BALF of the study group was higher than that of the control group, with significant difference (P < 0.05). With ROC curve, the area under the curve (AUC) of CCL2 in BALF predicting RMPP was 0.94, the cut-off value was 0.645 ng/ml, the sensitivity was 85%, and the specificity was 94%, and the diagnostic value was better than that of serum CRP and LDH. Logistic regression analysis was used to build the RMPP prediction model, and CCL2 showed good predictive value. Conclusion The level of CCL2 in BALF was high in children with MPP and had a high predictive value for RMPP. CCL2 can be used as one of the biomarkers for predicting RMPP.
... Inflammatory chemokines are important inflammatory factors that cause influenza immune pathological damage. They can strongly chemoattract inflammatory cells to accumulate in the lesion, and are positively correlated with the degree of inflammatory damage caused by influenza virus infection [9][10][11]. Studies have found that Escherichia coli or Klebsiella pneumoniae can promote the expression of inflammatory chemokines, thereby aggravating the degree of inflammatory damage [12]. ...
Article
Full-text available
Background Influenza is an acute respiratory infection caused by influenza virus. Maxing Shigan Decoction (MXSGD) is a commonly used traditional Chinese medicine prescription for the prevention and treatment of influenza. However, its mechanism remains unclear. Method The mice model of influenza A virus pneumonia was established by nasal inoculation. After 3 days of intervention, the lung index was calculated, and the pathological changes of lung tissue were detected by HE staining. Firstly, transcriptomics technology was used to analyze the differential genes and important pathways in mouse lung tissue regulated by MXSGD. Then, real-time fluorescent quantitative PCR (RT-PCR) was used to verify the changes in mRNA expression in lung tissues. Finally, intestinal microbiome and intestinal metabolomics were performed to explore the effect of MXSGD on gut microbiota. Results The lung inflammatory cell infiltration in the MXSGD group was significantly reduced (p < 0.05). The results of bioinformatics analysis for transcriptomics results show that these genes are mainly involved in inflammatory factors and inflammation-related signal pathways mediated inflammation biological modules, etc. Intestinal microbiome showed that the intestinal flora Actinobacteriota level and Desulfobacterota level increased in MXSGD group, while Planctomycetota in MXSGD group decreased. Metabolites were mainly involved in primary bile acid biosynthesis, thiamine metabolism, etc. This suggests that MXSGD has a microbial–gut–lung axis regulation effect on mice with influenza A virus pneumonia. Conclusion MXSGD may play an anti-inflammatory and immunoregulatory role by regulating intestinal microbiome and intestinal metabolic small molecules, and ultimately play a role in the treatment of influenza A virus pneumonia.
... The exception to this were myalgia, shivering and asthenia, which were 32%, 28% and 24% more frequent within patients with headache, respectively. These symptoms have been associated with the innate immune response and the release of inflammatory mediators [24,25]. The symptoms that were independently associated with the presence of headache supported the study hypothesis that headache is likely caused by the immune response and the cytokine and interleukin release. ...
Article
Full-text available
Headache is a common symptom of influenza infection; however, its causes and consequences remain uncertain. In this manuscript, we analyzed which demographic and clinical factors were associated with the presence of headache during the course of influenza infection and whether patients with headache had a different prognosis, evaluated by need of hospitalization, sick leave or school absenteeism. The influence study (NCT05704335) was an observational study that analyzed data routinely collected from the Health Sentinel Network between 2010 and 2020. During the study period, 7832 cases were considered, among which, 5275 (67.4%) reported headache. The presence of headache was independently associated with myalgia (2.753; 95%CI: 2.456–3.087, P < 0.001), asthenia (OR: 1.958; 95%CI: 1.732–2.214, P < 0.001), shivering (OR: 1.925; 95%CI: 1.718–2.156, P < 0.001), nasopharyngeal erythema (OR: 1.505; 95%CI: 1.293–1.753, P < 0.001), fever (OR: 1.469; 95%CI: 1.159–1.861; P = 0.001), sudden onset of symptoms (OR: 1.380; 95%CI: 1.120–1.702, p = 0.004), female sex (OR: 1.134; 95%CI: 1.023–1.257, P = 0.018), and gastrointestinal symptoms (OR: 1.169; 95%CI: 1.039–1.315; P = 0.01). Patients with headache had a sex and age adjusted lower odds of being referred to the hospital (OR: 0.463; 95%CI: 0.264–0.812, P = 0.007) and a higher odd of having a sick leave and/or school absenteeism (absenteeism (OR: 1.342; 95%CI: 1.190–1.514, P < 0.001). In conclusion, the presence of headache seems associated with symptoms caused by the innate immune response. These findings support a headache pathophysiology linked with the innate immune response. Due to the potential negative consequences and its treatable nature, clinicians should systematically evaluate it and, whenever necessary, treat it too.
... Significance IPF is a fatal condition of unbalanced immune responses associated with physiological manifestations such as lung mechanical dysfunction and dyspnea. Recent studies suggest that chemokines are important regulators of immune responses and fibrogenesis in the context of pulmonary fibrosis, accompanied by both elevated acute lung injury and infection [19][20][21][22][23], lung immune responses triggered by helminth infections [24], asthma [25], and pulmonary fibrosis [26,27]. Chemokines modulate fibrogenesis, leukocyte and fibrocyte influx, angiogenesis, alternative macrophage activation, and fibroblast collagen production that can sustain IPF development [3,28]. ...
... To expand the understanding of innate immune responses in influenza B virus infection using protein level immunoassays, a custom Luminex panel was designed to detect commonly upregulated cytokines and chemokines in response to infection with influenza and other respiratory viruses [32]. Cytokine and chemokine production was evaluated both at 48 hpi and 96 hpi. ...
Article
Full-text available
Understanding Influenza B virus infections is of critical importance in our efforts to control severe influenza and influenza-related diseases. Until 2020, two genetic lineages of influenza B virus-Yamagata and Victoria-circulated in the population. These lineages are antigenically distinct, but the differences in virus replication or the induction of host cell responses after infection have not been carefully studied. Recent IBV clinical isolates of both lineages were obtained from influenza surveillance efforts of the Johns Hopkins Center of Excellence in Influenza Research and Response and characterized in vitro. B/Victoria and B/Yamagata clinical isolates were recognized less efficiently by serum from influenza-vaccinated individuals in comparison to the vaccine strains. B/Victoria lineages formed smaller plaques on MDCK cells compared to B/Yamagata, but infectious virus production in primary human nasal epithelial cell (hNEC) cultures showed no differences. While ciliated epithelial cells were the dominant cell type infected by both lineages, B/Victoria lineages had a slight preference for MUC5AC-positive cells, and B/Yamagata lineages infected more basal cells. Finally, while both lineages induced a strong interferon response 48 h after infection of hNEC cultures, the B/Victoria lineages showed a much stronger induction of interferon-related signaling pathways compared to B/Yamagata. This demonstrates that the two influenza B virus lineages differ not only in their antigenic structure but also in their ability to induce host innate immune responses.
... Viral infection, especially which from the highly pathogenic influenza virus, leads to a marked increase in pro-inflammatory cytokines that induce the "cytokine storm" hypothesized to be the main cause of host death [36] . Previous studies have shown that mortality from influenza viral infection can be reduced by inhibiting cytokines that regulate inflammatory messages [37] . In addition, the "cytokine storm", characterized by overproduction and dysfunction of inflammatory cytokines, has become an important cause of host death during influenza viral challenge [38] , an effect that is used as a predictor of poor prognosis [39] . ...
Article
Full-text available
Influenza virus is prone to seasonal spread and widespread outbreaks, which pose important challenges to public health security. Therefore, it is important to effectively prevent and treat influenza virus infection. Schisandra polysaccharide (SPJ) is a polysaccharide derived from the fruit of Schisandra chinensis (Turcz.) Baill. In this study, we evaluated the antiviral activity of SPJ in vitro and in vivo, especially against influenza A virus (IAV) infection. By analyzing SPJ structure and monosaccharide composition, the molecular weight of SPJ was determined to be 115.5 KD, and it is composed of galacturonic acid (89.4%), rhamnose (0.8%), galactose (4.4%), arabinose (3.8%) and glucose (1.7%). Immunofluorescence analysis showed that SPJ treatment reduced the positive rate of viral nucleoproteins in cells, indicating that the compound had an inhibitory effect on influenza virus replication. Furthermore, SPJ therapy improved the survival of infected mice. Lung virus titer assays indicated that SPJ treatment significantly reduced viral-loading in the lung tissue of infected mice, and alleviated the pathological damage caused by influenza virus infection. Moreover, SPJ reduced cytokine expression during influenza virus challenge. In conclusion, SPJ has anti-influenza virus effects, and may have potential as an anti-influenza drug candidate in further clinical studies.
Article
Full-text available
Abstract Introduction: In recent years, highly pathogenicity avian influenza (HPAI), especially H5N1, has emerged as a major global health concern due to its potential as a zoonotic disease and its devastating impact on poultry populations. Identifying the molecular mechanisms of response to HPAI infection is critical to control, treatment and prevent the risk of a potential pandemic. Microarray technology is becoming a standard technology used in research laboratories all across the world and it is considered as one of the centers of research in cellular processes related to the level and manner of gene expression, including gene function and cell differentiation mechanisms. By using microarray technology, it is possible to obtain a detailed view of the interaction function of genes while simultaneously studying how the genome is expressed. Using of microarrays provides the analysis of gene expression in response to viral infections such as: influenza, etc., the study of host-pathogen interactions and also the identification of the effectiveness of drugs and vaccines. The aim of this study was to analyze the microarray data of H5N1 avian influenza in order to compare the gene network and analyze the functional pathway in chickens and ducks. Materials and Methods: Data mining and searching of microarray data related to Highly Pathogenic Avian Influenza infection was done in the GEO gene expression database (https://www.ncbi.nlm.nih.gov/geo). The microarray data set with accession number of GSE33389 based on GPL3213 platform was selected which contained lung tissue samples challenged with H5N1 virus in chickens and ducks. Normalization of selected microarray data was done using R software, and samples were grouped in order to compare between infected and control samples. Limma, Biobase and GEOquery software packages in R software were used to determine the expression level of genes and to investigate the differentially expressed genes (DEGs) between healthy and H5N1 influenza virus infected lung tissue samples in chickens and ducks. The criterion for selecting significant DEGs was considered as |logFC|>2 and P-Value<0.05. DAVID online tool (https://david.ncifcrf.gov) was used to investigate biological pathways, structural and functional characteristics of genes with different expression, and functional interpretation of up regulated and down regulated DEGs. It was evaluated and visualized separately based on biological processes (BP), molecular functions (MF) and cellular components (CC). KEGG tool (http://www.genome.jp/kegg) was used to evaluate and study metabolic pathway enrichment. In order to reveal interactions between proteins and analyze them, STRING database and Cytoscape software were used. While using the Cytohubba plugin to identify and display key genes, the main modules affecting the interaction of genes and proteins were also identified by the MCODE plugin. Results and Discussion: Gene expression analysis revealed 2062 and 565 differentially expressed genes between normal and infected tissue in chickens and ducks, respectively (p<0.05 and |logFC|>2). The results of bioinformatics analysis and protein-protein interaction network analysis showed BUB1, NDC80, CDC20, PLK1, PRC1, KIF11 and AURKA genes as hub genes in chicken and also COL6A3, COL3A1, COL4A3, COL18A1, PLOD2, PLOD1 and P4HA2 as highly effective genes in duck (p < 0.05). The results of the ontology comparison of DEGs proved that most of these genes in chickens are involved in the innate immune response and inflammatory resistance of the host, and the most effective genes in ducks play a role in lipid metabolism and energy production to meet the host's resistance to disease. The results of comparative gene network analysis between chickens and ducks are promising to increase our understanding of the host response to H5N1 influenza infection and the factors affecting virus pathogenesis in different bird species. Differentially expressed genes in response to H5N1 infection in chickens and ducks play critical roles in various biological processes, including immune response, inflammation, viral replication, and host-pathogen interactions. In general, gene network analysis showed that chickens and ducks use different genetic strategies to respond to avian influenza virus infection. Conclusion: The present research was conducted with the aim of discovering the response of H5N1 HPAI infection in chickens and ducks through comparative gene network analysis. It is important to note that the gene network analysis presented in this research is an initial step towards discovering the response mode of HPAI (H5N1) infection in chickens and ducks, and further functional studies, validation experiments and integration with other omics data are needed to confirm the role of genes, pathways and hub genes in host response to H5N1 virus. Therefore, the results of comparative gene network analysis in chickens and ducks obtained from this research can provide valuable insight into the underlying molecular mechanisms of host response to H5N1 influenza infection. Thus, by identifying differentially expressed genes, functional modules and hub genes in this research, it can be stated that potential targets for future research have been highlighted to some extent. Undoubtedly, further studies in this field will improve our knowledge about the pathogenesis of avian influenza and will help to develop strategies for effective control and prevention of H5N1 influenza outbreaks. Keywords: High Pathogenicity Avian influenza H5N1, bioinformatics, microarray, gene interaction network and gene ontology
Article
Background Respiratory complications often accompany influenza in patients with chronic obstructive pulmonary disease (COPD). In this retrospective study, we quantified the impact of antiviral therapy on exacerbations, healthcare resource utilization (HRU), and costs in patients with COPD across 5 influenza seasons. Methods Using claims data from US MarketScan® databases, we identified patients with COPD who had an influenza diagnosis during the 2012–2016 influenza seasons. Patients who received a neuraminidase inhibitor within 48 h of diagnosis ( N = 4134) were identified and propensity score–matched 1:1 to a comparator cohort of untreated patients. We determined COPD‐ and pneumonia‐related HRU and costs during month 1, each subsequent quarter, and months 2–13. Results Antiviral‐treated patients had a significantly lower frequency of COPD‐related outcomes than untreated patients during all periods (exacerbations: 10.4% vs 18.2% [month 1] and 17.7% vs 24.2% [months 2–13]; inpatient visit: 2.5% vs 7.9% [month 1] and 3.8% vs 6.7% [months 2–13]; P < 0.0001, all comparisons). Treated patients also had significantly lower outpatient and emergency department (ED) visits beyond month 1. Pneumonia‐related inpatient, ED, and outpatient visits were significantly lower in antiviral‐treated patients than in untreated patients over all periods ( P < 0.0001, all comparisons). In all HRU categories, COPD‐ and pneumonia‐related costs were significantly lower in treated patients over all periods (month‐1 ED visit costs were higher). Conclusions Antiviral treatment in patients with COPD and influenza is associated with significantly lower HRU and costs in the postinfection month and for an entire year following infection compared with untreated patients.
Preprint
Full-text available
Background: Tagetes erecta Linn. (TE) is used to treat cardiovascular, renal, and gastrointestinal diseases. In this study, we aimed to investigate the active compounds and targets of the TE extract in influenza A infections. Method: Active compounds and targets of the TE extract were identified using network pharmacology. Pathway and target studies were conducted using the Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Expression Omnibus (GEO) gene difference, and molecular docking analyses. In vitro validation of the TE extract was performed using antiviral and plaque reduction assays. Results: From 6 compounds, 95 active targets, 887 influenza-associated target genes, and 14 intersecting core targets were identified. A compound–target network was constructed and two compounds (lutein and beta-carotene) were identified. Protein–protein interaction and GEO analysis revealed interleukin (IL)-6 as a hub gene. In vitro experimental results revealed that the TE extract exerted therapeutic rather than protective effects. Conclusions: Overall, we identified the two main active compounds in the TE extract, lutein and beta-carotene, and IL-6 as an important target associated with influenza. Moreover, our findings confirmed the activity of the TE extract against the influenza A virus.
Article
Full-text available
Here we compare the results of pathological and virological examinations of mice experimentally infected with either wild-type or mouse-adapted pandemic A(H1N1) pdm09 viruses and highly pathogenic avian influenza (HPAI) virus A(H5N1). Mice were sacrificed on days 1, 3, 6, and 10 post infection or whenever morbidity was severe enough to justify euthanasia. Morbidity rates were calculated on the basis of clinical signs (weight loss, poor hair coat, hunched posture and paresis); virus-induced disease was characterised by the histopathology of lung; virus dissemination was determined by virus isolation on organ samples of lung, brain, liver, kidney and spleen. All mice infected with mouse-adapted A(H1N1) pdm09 died in the course of the experiment, whereas 20% of animals survived the infection with A(H5N1). Echinocyte formation changed the rheological properties of blood in animals infected with either mouse-adapted A(H1N1) pdm09 or A(H5N1). To sum up, the adaptation of pandemic A(H1N1) pdm09 virus can confer an enhanced virulence similar to or even exceeding that of HPAI A(H5N1) virus.
Article
Full-text available
Although several studies have exploited effects of PB1-F2 in swine influenza viruses, its contribution to the pathogenicity of swine influenza viruses remains unclear. Herein, we investigated effects of PB1-F2 on the pathogenicity of influenza virus using a virulent H1N1 A/swine/Kansas/77778/2007 (KS07) virus, which expresses a full-length PB1-F2, in mice and pigs. Using reverse genetics, we generated the wild type KS07 (KS07_WT), a PB1-F2 knockout mutant (KS07_K/O) and its N66S variant (KS07_N66S). KS07_K/O showed similar pathogenicity in mice as the KS07_WT, whereas KS07_N66S displayed enhanced virulence when compared to the other two viruses. KS07_WT exhibited more efficient replication in lungs and nasal shedding in infected pigs than the other two viruses. Pigs infected with the KS07_WT had higher pulmonary levels of GM-CSF, IFN-γ, IL-6 and IL-8 at 3 and 5 days post-infection, as well as lower levels of IL-2, IL-4 and IL-12 at 1 day post-infection compared to those infected with the KS07_K/O. These results indicate that PB1-F2 modulates KS07 H1N1 virus replication, pathogenicity and innate immune responses in pigs, and the single substitution at position 66 (N/S) in the PB1-F2 plays a critical role in virulence in mice. Taken together, our results provide new insights into the effects of PB1-F2 on the virulence of influenza virus in swine, and support PB1-F2 as a virulence factor of influenza A virus in a strain- and host-dependent manner.
Article
Full-text available
Influenza virus causes a respiratory disease in humans that can progress to lung injury with fatal outcome. The interleukin (IL)-36 cytokines are newly described IL-1 family cytokines that promote inflammatory responses via binding to the IL-36 receptor (IL-36R). The mechanism of expression and the role of IL-36 cytokines are poorly understood. Here, we investigated the role of IL-36 cytokines in modulating the innate inflammatory response during influenza virus-induced pneumonia in mice. The intranasal administration of influenza virus upregulated IL-36α mRNA and protein production in the lungs. In vitro, influenza virus-mediated IL-36α but not IL-36γ is induced and secreted from alveolar epithelial cells (AECs) through both a caspase-1 and caspase-3/7 dependent pathway. IL-36α was detected in microparticles shed from AECs and promoted the production of pro-inflammatory cytokines and chemokines in respiratory cells. IL-36R-deficient mice were protected from influenza virus-induced lung injury and mortality. Decreased mortality was associated with significantly reduced early accumulation of neutrophils and monocytes/macrophages, activation of lymphocytes, production of pro-inflammatory cytokines and chemokines, and permeability of the alveolar-epithelial barrier in despite impaired viral clearance. Taken together, these data indicate that IL-36 ligands exacerbate lung injury during influenza virus infection.Mucosal Immunology advance online publication 14 December 2016; doi:10.1038/mi.2016.107.
Article
Full-text available
The influenza A virus (IAV) PB1-F2 protein is a virulence factor contributing to the pathogenesis observed during IAV infections in mammals. In this study, using a mouse model, we compared the host response associated with PB1-F2 with an early transcriptomic signature that was previously associated with neutrophils and consecutively fatal IAV infections. This allowed us to show that PB1-F2 is partly involved in neutrophil-related mechanisms leading to death. Using neutropenic mice, we confirmed that the harmful effect of PB1-F2 is due to an excessive inflammation mediated by an increased neutrophil mobilization. We identified the downstream effects of this PB1-F2-exacerbated neutrophil recruitment. PB1-F2 had no impact on the lymphocyte recruitment in the airways at day 8 pi. However, functional genomics analysis and flow cytometry in broncho-alveolar lavages at 4 days pi revealed that PB1-F2 induced a NK cells deficiency. Thus, our results identify PB1-F2 as an important immune disruptive factor during the IAV infection.
Article
Full-text available
Background: Influenza A subtype H9N2 is widespread and prevalent in poultry. It has repeatedly transmitted zoonotically to cause mild influenza-like illness in humans and is regarded as a potential pandemic candidate. In additon, the six internal genes of H7N9 and H10N8 viruses which caused infection in human in China as well as some of the highly pathogenic H5N1 strains are origined from H9N2. Previous studies have shown that the mammalian adaptation PB2-Q591K contributes to the pathogenicity of H5N1 and H7N9 viruses. However, the role of the PB2-Q591K mutation in H9N2 subtype is still not well understood. Methods: To define and compare the individual role of PB2-Q591K substitution in the PB2 gene segment of H9N2 in relation to polymerase activity, replication competence and the pathogenicity using in vitro and in vivo models. Results: The PB2-Q591K mutation in H9N2 virus enhanced the polymerase activity and virus replication in human NHBE cells when compared to the wild type strain. Mice infected with the PB2 mutant showed significant weight loss, higher virus replication and immune responses in the lungs. Conclusions: Our evidences suggest that the PB2-Q591K, in addition to the -E627K mutation in H9N2 enhanced the pathogenicity in mammalian host.
Article
Full-text available
Type I (IFN-α/β) and type III (IFN-λ) interferons (IFNs) exert shared antiviral activities through distinct receptors. However, their relative importance for antiviral protection of different organ systems against specific viruses remains to be fully explored. We used mouse strains deficient in type-specific IFN signaling, STAT1 and Rag2 to dissect distinct and overlapping contributions of type I and type III IFNs to protection against homologous murine (EW-RV strain) and heterologous (non-murine) simian (RRV strain) rotavirus infections in suckling mice. Experiments demonstrated that murine EW-RV is insensitive to the action of both types of IFNs, and that timely viral clearance depends upon adaptive immune responses. In contrast, both type I and type III IFNs can control replication of the heterologous simian RRV in the gastrointestinal (GI) tract, and they cooperate to limit extra-intestinal simian RRV replication. Surprisingly, intestinal epithelial cells were sensitive to both IFN types in neonatal mice, although their responsiveness to type I, but not type III IFNs, diminished in adult mice, revealing an unexpected age-dependent change in specific contribution of type I versus type III IFNs to antiviral defenses in the GI tract. Transcriptional analysis revealed that intestinal antiviral responses to RV are triggered through either type of IFN receptor, and are greatly diminished when receptors for both IFN types are lacking. These results also demonstrate a murine host-specific resistance to IFN-mediated antiviral effects by murine EW-RV, but the retention of host efficacy through the cooperative action by type I and type III IFNs in restricting heterologous simian RRV growth and systemic replication in suckling mice. Collectively, our findings revealed a well-orchestrated spatial and temporal tuning of innate antiviral responses in the intestinal tract where two types of IFNs through distinct patterns of their expression and distinct but overlapping sets of target cells coordinately regulate antiviral defenses against heterologous or homologous rotaviruses with substantially different effectiveness.
Article
Full-text available
Respiratory influenza virus infections represent a serious threat to human health. Underlying medical conditions and genetic make-up predispose some influenza patients to more severe forms of disease. To date, only a few studies have been performed in patients to correlate a selected group of cytokines and chemokines with influenza infection. Therefore, we evaluated the potential of a novel multiplex micro-proteomics technology, SOMAscan, to quantify proteins in the respiratory mucosa of influenza A and B infected individuals. The analysis included but was not limited to quantification of cytokines and chemokines detected in previous studies. SOMAscan quantified more than 1,000 secreted proteins in small nasal wash volumes from infected and healthy individuals. Our results illustrate the utility of micro-proteomic technology for analysis of proteins in small volumes of respiratory mucosal samples. Furthermore, when we compared nasal wash samples from influenza-infected patients with viral load ≥ 28 and increased IL-6 and CXCL10 to healthy controls, we identified 162 differentially-expressed proteins between the two groups. This number greatly exceeds the number of DEPs identified in previous studies in human influenza patients. Most of the identified proteins were associated with the host immune response to infection, and changes in protein levels of 151 of the DEPs were significantly correlated with viral load. Most important, SOMAscan identified differentially expressed proteins heretofore not associated with respiratory influenza infection in humans. Our study is the first report for the use of SOMAscan to screen nasal secretions. It establishes a precedent for micro-proteomic quantification of proteins that reflect ongoing response to respiratory infection.
Article
PB2 is one of the subunits of the influenza A virus (IAV) polymerase complex. By bioinformatics analysis we identified PB2 substitutions at positions 389 and 598 among IAV isolates from humans, which might associate with viral pathogenicity. To evaluate the biological significance of these substitutions, PB2-K389R and -V598T/I mutant viruses of avian H7N9 IAVs were generated by reverse genetics. Compared to the wild type, the mutant viruses displayed an enhanced growth capacity in human and mammalian cells. Meanwhile, they presented increased transcription and replication by producing higher levels of viral mRNA, cRNA and vRNA. Minireplicon assays indicated that the polymerase activity was elevated by these substitutions. Notably, the PB2-V598T/I substitutions substantially increased virus replication and virulence in mice. Together, we demonstrated that the substitutions PB2-V598T/I contributed to higher IAV replication and virulence in mammals, which added to the knowledge of IAV virulence determinants and benefited the surveillance of IAVs.
Article
Rationale and objectives: Here we studied the interferon (IFN)-regulated innate immune response against influenza A virus (IAV) infection in the mouse lung and the therapeutic effect of IFN-λ2/3 in acute IAV lung infection. . Methods: For viral infections, IAV (WS/33, H1N1, PR8 H1N1, H5N1) were inoculated into WT mice by intranasal delivery and IAV mRNA level and viral titer were measured. To compare the antiviral effect of IFNs in vivo lung, neutralizing antibodies and recombinant IFNs were used. Measurement and main results: Following intranasal inoculation of IAV into mice, viral infection peaked at 7 days post of infection (dpi) and the IAV titer also reached its peak at this time. We found that IFN-β and IFN-λ2/3 were preferentially induced after IAV infection and the IFN-λ2/3-mediated innate immune response was specifically required for the induction of IFN-stimulated genes (ISGs) transcription in the mouse respiratory tract. Neutralization of secreted IFN-λ2/3 aggravated acute IAV lung infection in mice with intact IFN-β induction; consistent with this finding, the transcription of ISGs was significantly reduced. Intranasal administration of IFN-λ2/3 significantly suppressed various strains of IAV infection including WS/33 (H1N1), PR (H1N1), and H5N1 in the mouse lung and was accompanied by greater up-regulation of ISGs. Conclusion: Our data indicate that the IFN-λ2/3-mediated innate immune response is necessary to protect the lungs from IAV infection and intranasally delivered IFN-λ2/3 has the potential to be a useful therapeutic strategy for treating acute IAV lung infection.
Article
The innate immune system possesses a multitude of pathways to sense and respond to microbial pathogens. One such family are the interferons (IFNs), a family of cytokines that are involved in several cellular functions. Type I IFNs are appreciated to be important in several viral and bacterial diseases, while the recently identified type III IFNs (IFNL1, IFNL2, IFNL3, IFNL4) have been studied primarily in the context of viral infection. Viral and bacterial infections however are not mutually exclusive, and often the presence of a viral pathogen increases the pathogenesis of bacterial infection. The role of type III IFN in bacterial and viral-bacterial co-infections has just begun to be explored. In this mini review we discuss type III IFN signaling and its role in microbial pathogenesis with an emphasis on the work that has been conducted with bacterial pathogens.