You are on page 1of 7

46 Review TRENDS in Molecular Medicine Vol.9 No.

2 February 2003

Influenza-virus-induced signaling
cascades: targets for antiviral therapy?
Stephan Ludwig1, Oliver Planz2, Stephan Pleschka3 and Thorsten Wolff4
1
Institute of Molecular Medicine, Heinrich Heine University, Universitätsstr. 1, D-40225 Düsseldorf, Germany
2
Institute of Immunology, Bundesforschungsanstalt für Viruserkrankungen der Tiere, Paul Ehrlich Str. 28, D-72076 Tübingen,
Germany
3
Institute of Virology (FB10), Justus Liebig University, Frankfurter Str. 107, D-35392 Giessen, Germany
4
Robert Koch Institute, Nordufer 20, D-13353 Berlin, Germany

Influenza viruses continue to pose a severe threat immunoprophylaxis against influenza has several limi-
worldwide, causing thousands of deaths and an enor- tations in practice: (1) vaccination rates in the population
mous economic loss every year. The major problem in are too low to produce a herd immunity; (2) the efficacy of
fighting influenza is the high genetic variability of the vaccination is reduced in high-risk groups, such as the
virus, resulting in the rapid formation of variants that elderly and in immunocompromised individuals; (3) the
escape the acquired immunity against previous virus constant antigenic drift demands ongoing development of
strains, or have resistance to antiviral agents. Every new vaccines for each season; and (4) there is a constant
virus depends on its host cell and, hence, cellular func- risk of new viruses or shift variants emerging, against
tions that are essential for viral replication might be which even the most recent vaccines are ineffective. Thus,
suitable targets for antiviral therapy. As a result, intra- there is a clear need for potent antiviral therapies.
cellular signaling cascades induced by the virus, in par- The clinically approved anti-influenza drugs available
ticular mitogen-activated protein kinase pathways, today are directed against essential viral protein functions
have recently come into focus. (Table 1). Adamantane compounds, such as amantadine
and rimantadine, have a type-specific inhibitory effect on
Influenza is a highly contagious, acute respiratory disease
that affects all age groups and can occur repeatedly in any
particular individual. The etiological agent of the disease, (a) (b)
the influenza virus, is responsible for an average of vRNA Segments Gene products
114 000 hospitalizations and 20 000 death each year in
PB2 PB2
the USA alone [1]. Epidemics occur almost every year, PB1 PB1
PA
PA
PB1-F2
owing to an antigenic drift in two viral surface glyco-
proteins, hemagglutinin (HA) and neuraminidase (NA) HA HA

(Fig. 1). In recent history, highly pathogenic strains of the NP NP


NA NA
influenza A virus have emerged in an unpredictable
manner, causing pandemics such as the one in 1918 that
caused the death of 20 –40 million people worldwide [2].
Pandemic strains of the virus usually possess antigeni-
cally different, novel glycoproteins, resulting from anti- M M1
M2
genic shift, and hence the available vaccines are NS NS1
NEP/NS2
ineffective.
TRENDS in Molecular Medicine
Strains of avian influenza viruses can also appear in
humans, as happened in Hong Kong in 1997 [3]. These
viruses had an extremely high virulence in humans, Fig. 1. The influenza A virus particle. (a) An electrophoretic separation of the influ-
enza virus genome, which consists of eight RNA segments of negative polarity
killing six out of 18 infected individuals. Fortunately, they (reviewed in [49]). Each segment is named according to the viral protein(s) it
did not acquire the ability to spread in the human encodes. (b) The location of the different structural proteins within the virus par-
population, although it is possible that a novel virus strain ticle. The nucleoprotein (NP) (green) associates with each of the eight viral RNA
(vRNA) segments to form ribonucleoprotein (RNP) complexes. The subunits of the
emerging in the future could have such a capability. viral RNA-dependent RNA-polymerase complex PB1, PB2 and PA (red) are also
associated with the RNPs. The PB1 segment of many, but not all, influenza A virus
strains also contains a þ 1 reading frame encoding the recently discovered PB1-F2
Antiviral agents against influenza virus: the state of the protein [50]. The viral glycoproteins, hemagglutinin (HA) (blue) and neuraminidase
art (NA) (yellow), are exposed on the virus surface as trimers and tetramers, respect-
The most commonly employed and efficient way of ively. Matrix-protein-1 (M1) (purple) is derived from a colinear mRNA of segment 7
and forms the inner layer of the virion. A spliced mRNA of the same segment gives
controlling influenza is by vaccination. However, rise to the transmembrane M2 protein (orange), which is a pH-dependent ion chan-
nel. Alternative splicing of segment 8 yields the non-structural protein 1 (NS1) and
Corresponding author: Stephan Ludwig (stephan.ludwig@uni-duesseldorf.de). the nuclear export protein (NEP; also known as NS2) (grey).

http://tmm.trends.com 1471-4914/03/$ - see front matter q 2003 Elsevier Science Ltd. All rights reserved. PII: S1471-4914(02)00010-2
Review TRENDS in Molecular Medicine Vol.9 No.2 February 2003 47

Table 1. Inhibitors of influenza virus propagation


Inhibitor Action Affected step in viral replication
Inhibitors that target a viral protein or function
Sialic-acid analogs Block the receptor-binding pocket of Attachment of the virion to cell surface
hemagglutinin (HA) receptors
Quinone- and hydroquinone-derivatives Inhibit the low-pH-dependent Acquirement of the fusion-active
conformational change of HA conformation of HA
Anionic polymers Prevent the hydrophobic membrane Insertion of the fusion peptide into the cell
attachment membrane
M2 inhibitorsa (amantadine and Inhibit early Hþ influx into the virion and late Early release of the viral genome into the
rimantadine) Hþ efflux from transport vesicles cytoplasm and late induction of HA
fusion conformation
Inhibitors of the viral RNA-polymerase Inhibit the endonucleolytic (‘cap-snatching’) Transcription of viral mRNAs
subunit, PB2 activity of PB2
Neuraminidase inhibitorsa (e.g. zanamivir, Inhibit the cleavage of the glycosidic bond of Detachment of the virion from the cell
oseltamivir) terminal sialic-acid residues on receptor
determinants
Synthetic capped RNA and Inhibit viral polymerase activities Transcription and replication of the viral
phosphorothioate oligonucleotides genome

Inhibitors of host-cell functions


Protein kinase C (PKC) inhibitorsb (e.g. Block the PKC activity required for a late Entry of the virion by endocytosis
bisindolylmaleimide) endosomal process (in particular the
PKCbII isoform)
Protease inhibitors (exogenous and Inhibit proteolytic cleavage of HA Processing of the HA0 precursor into HA1
endogenous) and HA2
Hþ-ATPase inhibitors Inhibit the acidification of endocytotic pH-dependent activation of the HA fusion
vesicles activity
Ribavarin Inhibits cellular synthesis of the 7mGPPP- Transcription of viral mRNAs
cap of mRNAs
MAPK/ERK kinase (MEK) inhibitorsb Inhibit the Raf –MEK –ERK signaling pathway Nuclear-export-protein-mediated export of
(e.g. U0126) that is required for nuclear export. ribonucleoproteins
Abbreviations: ERK, extracellular-signal-regulated kinase; MAPK, mitogen-activated protein kinase.
a
Inhibitors approved for clinical use.
b
Novel signaling inhibitors effective in preclinical studies.

type-A but not on type-B influenza viruses (reviewed in adapt to, and should affect replication independent of the
[4]). These agent block the ion-channel activity of the viral type, strain and antigenic properties of the invading
M2 protein (Fig. 1), which is mainly required during virus influenza virus.
entry in the early phase of the replication cycle (Fig. 2).
However, amantadine-sensitive influenza A virus strains Intracellular signaling cascades and influenza virus
rapidly develop resistance in vitro and in vivo [5]. infection
Zanamivir and oseltamivir, two products of modern Cell-fate decisions in response to extracellular agents,
drug design, block the receptor-destroying activity of the including pathogenic invaders, are commonly mediated by
viral NA protein, thereby preventing viral release from the phosphorylation-regulated signaling cascades that trans-
cell membrane [6]. Both drugs efficiently inhibited influenza duce signals into stimulus-specific actions, such as changes
virus A and B strains in clinical studies [7,8], and are in gene expression patterns.
approved and commercially available in many countries. Influenza viruses induce expression of a variety of
However, escape from the selective pressure of NA inhibitors cytokine and chemokine genes, such as interferons IFN-a
has been observed in cell culture and in patients, as a result and IFN-b, tumor-necrosis-factor-a (TNF-a), interleukins
of various mutations, although on a much smaller scale than IL-1b, IL-6 and IL-8, monocyte chemo-attractant protein 1
with adamantane compounds [9,10]. (MCP-1) and RANTES (reviewed in [11,12]). In a recent
Ongoing studies are attempting to find new anti- transcriptional-profiling study, 84 out of an array of 13 000
influenza drugs that target the virus replication cycle genes were shown to be deregulated in response to infec-
at many different steps (Table 1) [4], but none has yet tion in a lung epithelial cell line [13], suggesting that the
been approved for clinical use. All these approaches activity of many intracellular signaling mediators might
have the major disadvantage that they attack a viral be altered. Signaling processes can be initiated by the cell
function and/or structure. Hence, even though strongly as a defense against the invader, but can also be used by
conserved elements will not change easily, the virus will the virus to support replication.
eventually adapt to and escapes from the selective pressure
exerted by the drug. Protein kinase C: a regulator of influenza virus entry
As a result, specific cellular functions essential for virus The protein kinase C (PKC) superfamily consists of at
replication have attracted increasing interest, because a least 12 different isoforms, which carry out diverse
missing cellular function is more difficult for the virus to regulatory roles in cellular processes by linking into
http://tmm.trends.com
48 Review TRENDS in Molecular Medicine Vol.9 No.2 February 2003

(a) Adsorption
(i) Budding

Bisindolyl- (h) Packaging ?


maleimide I
Post-translational
Raf
processing
MEK U0126
(f) Translation
PKCβII (b) Endocytosis
ERK

(c) Fusion and


(c) release
(e) (g) RNP
mRNA export

vRNA (–)
(d) Import
cRNA (+) Nucleus

TRENDS in Molecular Medicine

Fig. 2. The lifecycle of the influenza A virus. The virus first binds to sialic-acid-containing receptors (dark blue) on the cell surface via the hemagglutinin protein (a). It then
enters the cell by endocytosis (b) and the viral RNA (vRNA) is released by fusion of the viral and endosomal membranes (c). The viral genome is transported into the
nucleus (d) where transcription and genome amplification take place (e). Viral transcripts are translated in the cytoplasm (f). Late in the infection cycle the viral genome is
transported out of the nucleus in the form of ribonucleoprotein (RNP) complexes (g). RNPs are packaged (h) and progeny virions are then released from the cell surface (i).
The replication cycle is completed within 6–16 h, depending on the particular virus strain and the cell type. Endocytosis of viral particles appears to be controlled by the
protein-kinase-C isoform, PKCbII, and RNP export is dependent upon the Raf –MEK –ERK cascade. Thus, specific inhibition of PKC or of the Raf– MEK– ERK pathway by
bisindolylmaleimides or by the MEK inhibitor U0126, respectively, results in impaired virus propagation.

several downstream signaling pathways [14]. Based on expression [20]. In this way, MAPK cascades regulate
the action of two protein-kinase inhibitors, H7 and numerous cellular responses, such as proliferation and
staurosporine, it has been suggested that PKCs have a differentiation, and also cell activation and immune
role in the entry process of several enveloped viruses response [21]. Four different members of the MAPK
[15]. The influenza virus and the viral HA can rapidly family, which are organized in separate cascades,
activate PKCs upon binding to host-cell surface have been identified to date (Fig. 3) [20]. Three of
receptors (Fig. 3) [16 – 18]. Furthermore, the PKC these, extracellular-signal-regulated kinase (ERK), JUN
inhibitor, bisindolylmaleimide I, has been shown to N-terminal kinase (JNK), and p38 have been reported to
prevent influenza virus entry and subsequent infection be activated upon infection by an influenza virus [22 – 24]
in a dose-dependent and reversible manner [19]. It is and there is also evidence of viral activation of ERK5 (also
likely that the PKCbII isoform performs this function; known as big mitogen-activated protein kinase) (S. Ludwig
overexpression of a phosphorylation-deficient dominant- et al., unpublished).
negative mutant form of PKCbII showed that it is a Recent work has helped to clarify the functions of the
regulator of late endosomal sorting, and hence blocks p38, JNK and ERK signaling pathways. Using specific
virus entry at the level of late endosomes (G. Whittaker, kinase inhibitors, p38 and JNK but not ERK have been
pers. commun.) (Fig. 3). Therefore, a specific pharma- linked with virus-induced expression of RANTES, a
cological interference with PKCbII might help to chemokine involved in the attraction of eosinophils during
prevent influenza virus infections at the most effective an inflammatory response (Fig. 3) [22]. The importance of
level: the initial entry of virions into their target the JNK subgroup of MAPKs in influenza virus infection
cells (Fig. 2). was further suggested by the observation that activator-
protein-1 (AP-1) transcription factors are activated early
Mitogen-activated protein kinase cascades and influenza in productively infected cells [23]; AP-1 factors include
virus infection c-Jun and activating-transcription-factor-2 (ATF-2), whose
Mitogen-activated protein kinase (MAPK) cascades are transcriptional activity is potentiated by phosphorylation
important signal transducers in the conversion of a by JNK [25]. Both c-Jun and ATF-2 are phosphorylated
variety of extracellular signals into changes in gene upon influenza virus infection [23,26]. Furthermore,
http://tmm.trends.com
Review TRENDS in Molecular Medicine Vol.9 No.2 February 2003 49

NA HA Influenza virus infection

dsRNA
Extracellular
TLR-3 Intracellular
Intracellular ?
NP
RNA accumulation
M1 HA (dsRNA)
PKR Raf
?
PKCβII
MKK4 MKK7 MKK6 MEK5 MEK1/2
IKK

Endocytosis P P P P P P P P
P T PY
ΙκBα ΙκBα T GY T EY T EY
p50–p65 JNK p38 ERK5 ERK

P P
p50–p65 ATF-2 c-Jun
? RNP export
NF-κB AP-1
TNF-α expression IFN-β expression IFN-β expression RANTES Nucleus
IL-1 expression IL-8 expression RANTES expression expression
TRENDS in Molecular Medicine

Fig. 3. Influenza-virus-induced signaling processes and their functions in the infected host cell. Induction of signaling by influenza viruses can be divided in early events,
such as binding of the virus, hemagglutanin (HA), neuraminidase (NA) or free double-strand RNA (dsRNA) to cellular receptors, and late events, which commonly require
productive virus replication. Entry of the virus into the cell requires the activation of the protein kinase C bII (PKCbII) isoform. Intracellular expression of the viral proteins
HA, nucleoprotein (NP) and matrix-protein-1 (M1), or accumulation of viral RNA species, leads to activation of nuclear factor kB (NF-kB) (made up of subunits p50 and p65)
and upregulated expression of several antiviral cytokines, such as interferon-b (IFN-b) and interleukin-8 (IL-8). In addition, four members of the mitogen-activated protein
kinase (MAPK) family – JUN N-terminal kinase (JNK), p38, extracellular-signal-regulated kinase (ERK) and ERK5 – are also induced in response to infection by the influenza
virus, by phosphorylation at specific threonine and tyrosine residues. JNK and p38 mediate antiviral signaling via the transcription factor AP-1, whereas ERK signaling is
proviral, inducing export of viral ribonucleoprotein (RNP) from the nucleus. Abbreviations: IkB, inhibitor of NF-kB; IKK, IkB inhibitor; MEK, MAPK/ERK kinase; MKK, MAPK
kinase; P, phosphate; PKR, protein kinase R; TLR-3, Toll-like receptor 3; TNF-a, tumor-necrosis-factor-a.

activation of JNK was observed using different virus NEP [24]. Thus, in contrast to the JNK cascade,
strains in a variety of permissive cell lines [22,23]; it activation of the Raf – MEK – ERK pathway is required
required productive replication and was induced by the for efficient virus growth. The replication of other
accumulating RNA made by the viral polymerase. Two viruses, such as Borna-disease virus [28], Visna virus
MAPK kinases, MKK4 and MKK7, have been identified as [29] or Coxsackie B3 virus [30], is also impaired by
upstream activators in the viral context. The AP-1 factors, MEK inhibition. Interestingly, MEK inhibitors meet
c-Jun and ATF-2, are crucial for the expression of two further criteria of a potential anti-influenza agent:
interferon-b (IFN-b), a potent antiviral cytokine [27], (1) the inhibitors were not toxic in a variety of cell
and hence inhibition of the JNK signaling cascade by types tested [24,28] and (2) there is no tendency
dominant-negative mutant forms of MKK7, JNK or c-Jun towards induction of the formation of resistant virus
during a virus infection resulted in impaired transcription variants. Hence, the Raf – MEK – ERK pathway appears
from the IFN-b promoter and an enhancement of virus to be a suitable target for antiviral intervention.
production. Thus, the JNK pathway appears to be a crucial
mediator of the antiviral response to an influenza virus IKK –NF-kB pathway
infection [23]. Activation of the transcription factor, nuclear factor kB
Interestingly, suppression of ERK using U0126 [a (NF-kB), is a hallmark of most infections by viral
specific inhibitor of the upstream MAPK/ERK kinase pathogens [31], including influenza viruses (reviewed
(MEK)], or using a dominant-negative mutant form of in [11,12]). Induction of NF-kB by a virus involves
ERK or the MEK activator Raf, results in a strong activation of IKK [inhibitor of NF-kB (IkB) kinase].
impairment of virus propagation [24]. At a molecular Isolated components of the influenza virus, such as
level, it is the nuclear export of viral ribonucleoprotein double-strand RNA [32] or overexpressed viral HA,
complexes that is affected by inhibition of the ERK nucleoprotein (NP) or matrix-protein-1 (M1) [33], can
signaling pathway (Fig. 2), probably as a result of also activate IKK (Fig. 3). Gene expression of many
impaired activity of the viral nuclear export protein, pro-inflammatory or antiviral cytokines, such as IFN-b
http://tmm.trends.com
50 Review TRENDS in Molecular Medicine Vol.9 No.2 February 2003

and tumor-necrosis-factor-a (TNF-a), is controlled by dsRNA is the best characterized of the viral signaling
NF-kB and, therefore, it was suggested that IKK and inducers in terms of its cellular receptors (Fig. 3). At least
NF-kB are essential components in the innate immune two cellular gene products bind to dsRNA and induce
response to virus infections [32]. In accordance with activation of NF-kB: the IFN-inducible RNA-dependent
this idea, influenza-virus-induced activity of the IFN-b protein kinase R (PKR) [27] and the Toll-like receptor 3
promoter is strongly impaired in cells expressing (TLR-3) [40]. PKR acts as an intracellular sensor, whereas
transdominant-negative mutants of IKK2 or IkBa TLR-3 is thought to detect extracellular dsRNA species
[34]. However, recent experiments have shown that released from dying cells [40]. Extracellular RNAs have
titers of influenza viruses grown in cells with impaired been detected as relatively stable structures in the super-
NF-kB signaling were dramatically lower than those natants of influenza-virus-infected cells [41]. Thus, the cell
from wild-type cells (S. Ludwig et al., unpublished). has developed a two-step method of dsRNA sensing: TLR-3
This suggests that NF-kB activity is also required for confers immediate protective signaling initiated from the
efficient virus replication, probably by regulating outside, which does not require virus entry, whereas PKR
expression of an as yet unidentified proviral cellular senses the dsRNA produced at later stages within the
protein. infected cell (Fig. 3).

Influenza viral inducers or inhibitors of signaling The influenza non-structural-protein-1: a universal


A specific pharmacological interference with virus-induced antagonist of dsRNA-induced signaling processes
signaling could be achieved by blocking the interaction of Many of the signaling functions in a virus-infected cell,
the inducing viral component with its cellular receptor. especially those induced by dsRNA, are thought to be
Unfortunately, little is known about the molecular inter- part of an antiviral program and, hence, it is not
actions of influenza viral inducers with cellular sensors, surprising that influenza viruses have evolved strate-
although several influenza virus proteins have been gies to counteract this response. For example, recent
reported to exhibit signal-inducing capacity. It was noted analyses have shown that a major in vivo function of
more than 20 years ago that influenza strains differ in the RNA-binding influenza non-structural-protein-1
their ability to induce IFN-a and IFN-b, and that this (NS1) is to antagonize the antiviral type-I-IFN system
correlates with the NA activity of these strains [35]. It was [42]. Strikingly, deletion of the coding region of NS1 in
subsequently shown that treatment of cells with recombin- a recombinant virus resulted in a complete loss of
ant NA, but not HA, induces the production of IL-1 and virulence in wild-type mice [43]. However, this delNS1
TNF-a in murine macrophages (Fig. 3) [36]. In another mutant virus could still kill mice with genetic knock-
study, NA was shown to convert transforming-growth- outs of either the STAT1 (signal-transducer-and-
factor-b (TGF-b) from the latent to the active form to an activator-of-transcription-1) protein (which transduces
extent sufficient to induce TGF-b-dependent apoptosis IFN-mediated signals to the transcriptional machinery)
[37]. Hence, although the exact molecular mechanisms [43], or PKR (whose expression is upregulated by IFN)
and the pathways involved in NA-induced signaling [44]. Moreover, multicyclic replication of the mutant
remain unknown, both direct and indirect events might virus was strongly attenuated in IFN-competent but
be involved. not in IFN-deficient tissue-culture cells. Further
HA can stimulate signaling from both inside the cell comparisons of wild-type and delNS1 viruses revealed
and at the cell surface (Fig. 3): binding of recombinant that NS1 inhibits activation of the gene encoding
HA to cell-surface receptors leads to a rapid induction IFN-b, which is controlled by the transcriptional
of PKC signaling [17], whereas overexpression of HA activators NF-kB, interferon-regulatory-factor-3, and
inside the cell leads to the activation of NF-kB [38], ATF-2/c-Jun [26,34,45]. Significantly, induction of the
probably via an endoplasmic-reticulum (ER)-overload IFN-b gene in the absence of NS1 correlates with a
mechanism. Interestingly, NF-kB can also be activated strong upregulation of the three dsRNA-responsive
by expression of M1 and NP, which are not processed transcription factors, demonstrating that NS1 inter-
in the ER [33]. This suggests that there might be feres with upstream signaling events. The inhibitory
different, and as yet unidentified, cellular sensors for activity of NS1 was shown to be dependent on its
all three proteins. Induction of IKK and NF-kB by HA, RNA-binding domain, and it is thought that NS1
NP and M1 appears to be specific because MAPK sequesters intracellular dsRNA produced during viral
signaling pathways, and transcription factors from the replication, thereby denying access of the antiviral
AP-1 and Ets families, are not activated [33]. enzyme PKR and of other cellular dsRNA-sensor
The most potent viral signaling component, however, is proteins. Inhibition of the processing of residual IFN
probably dsRNA. It is generally believed that most RNA mRNAs or IFN-induced mRNAs by the NS1 protein
viruses produce dsRNA-like intermediates and that these might further contribute to its antagonistic activity
represent a shared molecular pattern that triggers [46]. However, influenza patients actually secrete some
antiviral signaling within the cell [39]. Indeed, dsRNA IFN-a [47], and some IFN-dependent genes are still
induces activation of pathways that regulate the type-I-IFN activated upon infection by the NS1-expressing wild-
response and expression of other antiviral cytokines. type virus [13]. Therefore, the IFN-antagonistic activ-
Several signaling pathways are involved, including the ity of NS1 might not be considered an absolute but
IKK –NF-kB pathway, and both the JNK and p38 MAPK rather a relative, exhaustible function that boosts viral
cascades [32]. replication in the early phase of infection.
http://tmm.trends.com
Review TRENDS in Molecular Medicine Vol.9 No.2 February 2003 51

In conclusion, the NS1 protein is a well-characterized Acknowledgements


example of an IFN-antagonistic protein, and has a This work was supported by the Deutsche Forschungsgemeinschaft (grant
Lu477/4 – 3 to S.L.) and by the Fonds der Chemischen Industrie.
crucial role in viral virulence. Further support for this
role of the NS1 protein came from the finding that the
References
high virulence of the avian H5N1 influenza viruses 1 Simonsen, L. et al. (1997) The impact of influenza epidemics on
that were transmitted to humans in Hong Kong in mortality: introducing a severity index. Am. J. Public Health 87,
1997 was caused largely by a specific amino-acid 1944– 1950
position in the NS1 protein [48]. These viruses were 2 Taubenberger, J.K. et al. (2000) The 1918 influenza virus: a killer
comes into view. Virology 274, 241 – 245
particularly resistant to the antiviral action of IFNs
3 Hatta, M. and Kawaoka, Y. (2002) The continued pandemic threat
and TNF-a, and this resistance could be transmitted to posed by avian influenza viruses in Hong Kong. Trends Microbiol. 10,
other strains of the virus by transfer of the H5N1-virus 340 – 344
NS gene segment [48]. 4 Luscher-Mattli, M. (2000) Influenza chemotherapy: a review of the
Hence, strengthening the innate antiviral response by present state of art and of new drugs in development. Arch. Virol. 145,
2233– 2248
using pharmacological measures to interfere with the 5 Fleming, D.M. (2001) Managing influenza: amantadine, rimantadine
NS1-mediated block of antiviral signaling might be a and beyond. Int. J. Clin. Pract. 55, 189 – 195
legitimate strategy for restraining the replication and 6 Gubareva, L.V. et al. (2000) Influenza virus neuraminidase inhibitors.
spread of influenza viruses. Lancet 355, 827 – 835
7 Dreitlein, W.B. et al. (2001) Zanamivir and oseltamivir: two new
options for the treatment and prevention of influenza. Clin. Ther. 23,
Concluding remarks 327 – 355
Influenza virus infections impose a considerable socio- 8 McClellan, K. and Perry, C.M. (2001) Oseltamivir: a review of its use in
economic burden upon society, despite annual vaccin- influenza. Drugs 61, 263– 283
ation campaigns. Therefore, efficient antiviral therapy is 9 Gubareva, L.V. et al. (1998) Evidence for zanamivir resistance in an
immunocompromised child infected with influenza B virus. J. Infect.
required for control of the virus. Current anti-influenza Dis. 178, 1257– 1262
drugs target viral components (Table 1) and are there- 10 Gubareva, L.V. et al. (2002) Detection of influenza virus resistance to
fore prone to compensatory viral escape mechanisms. neuraminidase inhibitors by an enzyme inhibition assay. Antiviral
However, blocking specific host-cell functions that are Res. 53, 47 – 61
11 Ludwig, S. et al. (1999) A fatal relationship – influenza virus
required for viral replication, but not for short-term cell interactions with the host cell. Viral Immunol. 12, 175 – 196
survival, might reduce the frequency at which resistant 12 Julkunen, I. et al. (2000) Inflammatory responses in influenza A virus
variants arise. infection. Vaccine 19, S32– S37
A variety of cellular signaling cascades are activated 13 Geiss, G.K. et al. (2002) Cellular transcriptional profiling in influenza
A virus-infected lung epithelial cells: the role of the nonstructural NS1
by influenza virus infection. Some of these pathways
protein in the evasion of the host innate defense and its potential
are purely antiviral, whereas others appear to support contribution to pandemic influenza. Proc. Natl. Acad. Sci. U. S. A. 99,
virus replication. Hence, it should be possible to inter- 10736 – 10741
fere with virus replication either by enhancing anti- 14 Toker, A. (1998) Signaling through protein kinase C. Front. Biosci. 3,
viral signaling or by inhibiting proviral signaling, D1134– D1147
15 Constantinescu, S.N. et al. (1991) Effects of protein kinase C inhibitors
especially if this intervention is well tolerated in the on viral entry and infectivity. FEBS Lett. 292, 31 – 33
normal cell. 16 Rott, O. et al. (1995) B cell superstimulatory influenza virus
The suitability of this concept has so far been best (H2-subtype) induces B cell proliferation by a PKC-activating,
proven for the Raf– MEK– ERK signaling cascade. Block- Ca2þ-independent mechanism. J. Immunol. 154, 2092 – 2103
17 Arora, D.J. and Gasse, N. (1998) Influenza virus hemagglutinin
ing this cascade using dominant-negative mutants or a
stimulates the protein kinase C activity of human polymorphonuclear
specific inhibitor resulted in a strong impairment of virus leucocytes. Arch. Virol. 143, 2029 – 2037
propagation, and no severe cytotoxicity or tendency to 18 Kunzelmann, K. et al. (2000) Influenza virus inhibits amiloride-
induce resistant variants was detected. Although target- sensitive Naþ channels in respiratory epithelia. Proc. Natl. Acad. Sci.
ing of a cellular factor will always raise concerns about the U. S. A. 97, 10282 – 10287
19 Root, C.N. et al. (2000) Entry of influenza viruses into cells is inhibited
side effects of a drug, it appears that local administration by a highly specific protein kinase C inhibitor. J. Gen. Virol. 81,
of an agent, such as a MEK inhibitor, to the primary site of 2697– 2705
influenza virus infection, the lung, is well tolerated. Here, 20 Widmann, C. et al. (1999) Mitogen-activated protein kinase: conserva-
the drug primarily affects differentiated lung epithelial tion of a three-kinase module from yeast to human. Physiol. Rev. 79,
143– 180
cells, in which a proliferative signaling cascade like 21 Dong, C. et al. (2002) MAP kinases in the immune response. Annu. Rev.
Raf – MEK–ERK might be dispensable. Using this Immunol. 20, 55 – 72
approach, we have recently demonstrated in a mouse 22 Kujime, K. et al. (2000) p38 mitogen-activated protein kinase and
infection model that local administration of the MEK c-jun-NH2-terminal kinase regulate RANTES production by influenza
virus-infected human bronchial epithelial cells. J. Immunol. 164,
inhibitor U0126 reduces virus titers in the lung (S. Ludwig
3222– 3228
et al., unpublished). 23 Ludwig, S. et al. (2001) Influenza virus-induced AP-1-dependent gene
In addition to the Raf – MEK – ERK cascade, other expression requires activation of the JNK signaling pathway. J. Biol.
signaling pathways and mediators could be possible Chem. 276, 10990 – 10998
targets, including PKCs, other MAPK pathways and the 24 Pleschka, S. et al. (2001) Influenza virus propagation is impaired by
inhibition of the Raf/MEK/ERK signalling cascade. Nat. Cell Biol. 3,
IKK – NF-kB module, or the apoptotic cascades, which 301– 305
are suspected to have some proviral activity in infected 25 Karin, M. et al. (1997) AP-1 function and regulation. Curr. Opin. Cell
cells. Biol. 9, 240– 246
http://tmm.trends.com
52 Review TRENDS in Molecular Medicine Vol.9 No.2 February 2003

26 Ludwig, S. et al. (2002) The influenza A virus NS1 protein inhibits 39 Majde, J.A. (2000) Viral double-stranded RNA, cytokines, and the flu.
activation of Jun N-terminal kinase (JNK) and AP-1 transcription J. Interferon Cytokine Res. 20, 259 – 272
factors. J. Virol. 76, 11166 – 11171 40 Alexopoulou, L. et al. (2001) Recognition of double-stranded RNA
27 Stark, G.R. et al. (1998) How cells respond to interferons. Annu. Rev. and activation of NF-kB by Toll-like receptor 3. Nature 413,
Biochem. 67, 227– 264 732 – 738
28 Planz, O. et al. (2001) MEK-specific inhibitor U0126 blocks spread of 41 Majde, J.A. et al. (1998) Spontaneous release of stable viral double-
Borna disease virus in cultured cells. J. Virol. 75, 4871– 4877 stranded RNA into the extracellular medium by influenza virus-
29 Barber, S.A. et al. (2002) Visna virus-induced activation of MAPK is infected MDCK epithelial cells: implications for the viral acute phase
required for virus replication and correlates with virus-induced response. Arch. Virol. 143, 2371– 2380
neuropathology. J. Virol. 76, 817– 828 42 Garcia-Sastre, A. (2001) Inhibition of interferon-mediated antiviral
30 Luo, H. et al. (2002) Coxsackievirus B3 replication is reduced by responses by influenza A viruses and other negative-strand RNA
inhibition of the extracellular signal-regulated kinase (ERK) signaling viruses. Virology 279, 375– 384
pathway. J. Virol. 76, 3365– 3373 43 Garcia-Sastre, A. et al. (1998) Influenza A virus lacking the NS1
31 Hiscott, J. et al. (2001) Hostile takeovers: viral appropriation of the gene replicates in interferon-deficient systems. Virology 252,
NF-kB pathway. J. Clin. Invest. 107, 143 – 151 324 – 330
44 Bergmann, M. et al. (2000) Influenza virus NS1 protein counter-
32 Chu, W.M. et al. (1999) JNK2 and IKKb are required for activating the
acts PKR-mediated inhibition of replication. J. Virol. 74,
innate response to viral infection. Immunity 11, 721– 731
6203 – 6206
33 Flory, E. et al. (2000) Influenza virus-induced NF-kB-dependent gene
45 Talon, J. et al. (2000) Activation of interferon regulatory factor 3 is
expression is mediated by overexpression of viral proteins and involves
inhibited by the influenza A virus NS1 protein. J. Virol. 74,
oxidative radicals and activation of IkB kinase. J. Biol. Chem. 275,
7989 – 7996
8307 – 8314
46 Kim, M.J. et al. (2002) Human influenza viruses activate an interferon-
34 Wang, X. et al. (2000) Influenza A virus NS1 protein prevents
independent transcription of cellular antiviral genes: Outcome with
activation of NF-kB and induction of a/b interferon. J. Virol. 74,
influenza A virus is unique. Proc. Natl. Acad. Sci. U. S. A. 99,
11566– 11573
10096 – 10101
35 Chomik, M. (1981) Interferon induction by influenza virus: signifi- 47 Hayden, F.G. et al. (1998) Local and systemic cytokine responses
cance of neuraminidase. Arch. Immunol. Ther. Exp. 29, 109– 114 during experimental human influenza A virus infection. Relation
36 Houde, M. and Arora, D.J. (1990) Stimulation of tumor necrosis factor to symptom formation and host defense. J. Clin. Invest. 101,
secretion by purified influenza virus neuraminidase. Cell. Immunol. 643 – 649
129, 104 – 111 48 Heui-Seo, S. et al. (2002) Lethal H5N1 influenza viruses escape host
37 Schultz-Cherry, S. and Hinshaw, V.S. (1996) Influenza virus neur- anti-viral cytokine responses. Nat. Med. 8, 950– 954
aminidase activates latent transforming growth factor b. J. Virol. 70, 49 Lamb, R.A. and Krug, R.M. (2001) Orthomyxoviridae: the viruses and
8624 – 8629 their replication. In Virology (Fields, B.N., ed.), pp. 1487 – 1531,
38 Pahl, H.L. and Baeuerle, P.A. (1995) Expression of influenza virus Lippincott – Raven
hemagglutinin activates transcription factor NF-kB. J. Virol. 69, 50 Chen, W. et al. (2001) A novel influenza A virus mitochondrial protein
1480 – 1484 that induces cell death. Nat. Med. 7, 1306 – 1312

The BioMedNet Magazine


The new online-only BioMedNet Magazine contains a range of topical articles currently available in
Current Opinion and Trends journals, and offers the latest information and observations of direct and
vital interest to researchers.

You can elect to receive the BioMedNet Magazine delivered directly to your e-mail address,
for a regular and convenient survey of what’s happening outside your lab,
your department, or your speciality.

Issue-by-issue, the BioMedNet Magazine provides an array of some of the finest material available on
BioMedNet, dealing with matters of daily importance: careers, funding policies, current controversy and
changing regulations in the practice of research.

Don’t miss out – register now at http://news.bmn.com/magazine

http://tmm.trends.com

You might also like