Sei sulla pagina 1di 7

Interferons and viral infections

Volker Fensterl and Ganes C. Sen*


Department of Molecular Genetics, The Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA

Abstract.
Interferons represent a family of cytokines, which is of IL-12, and its expression is restricted to immune cells such
central importance in the innate immune response to virus as T cells and NK cells. The effectiveness of the interferon
infections. All interferons act as secreted ligands of specific system in counteracting viral infections is reflected by the
cell surface receptors, eliciting the transcription of hundreds multitude of inhibitors of interferon induction or interferon
of interferon-stimulated genes whose protein products have action that are encoded by many viruses, preventing their
antiviral activity, as well as antimicrobial, antiproliferative/ eradication and resulting in the continued coexistence of
antitumor, and immunomodulatory effects. Expression of viruses and vertebrates. The unique biological functions of
type I and III interferons is induced in virtually all cell types interferons have led to their therapeutic use in the
upon recognition of viral molecular patterns, especially treatment of diseases such as hepatitis, multiple sclerosis,
nucleic acids, by cytoplasmic and endosomal receptors, and certain leukemias.
whereas type II interferon is induced by cytokines such as
V
C 2009 International Union of Biochemistry and Molecular Biology, Inc.
Volume 35, Number 1, January/February 2009, Pages 14–20 
E-mail: seng@ccf.org Keywords: Interferon, virus, antiviral, innate immunity

1. The interferon family and three types, based on amino acid sequence homology and
the receptors they use (Table 1): Type I IFN mainly refers to
their receptors the numerous subtypes of IFN-a (13 in human), and a single
In 1957, Isaacs and Lindenmann described a secreted macro- IFN-b, all of which can be induced in most cell types, and
molecule, which was produced by cells after treatment with whose genes are arranged in a cluster on human chromo-
heat-inactivated influenza virus. The molecule was able to some 9. In addition, IFN-x, IFN-e, IFN-j, IFN-s, and IFN-f (¼
‘‘interfere’’ with live virus replication and was therefore limitin) belong to the type I IFNs, but they are expressed
termed interferon [1]. In the following five decades, the with cell-type and species specificity. For example, IFN-s is
human and murine interferon systems have been studied in expressed by trophoblasts and has been found in ruminants
great detail, revealing the mechanisms of transcriptional only, whereas the limitins have been identified only in
induction of interferon (IFN), its mode of action, its antiviral mouse. All type I IFNs use the heterodimeric IFN-a/b recep-
and other effects, the numerous viral countermeasures, and tor (IFNAR). The only member of type II IFNs, IFN-c, is unre-
its benefits in clinical use. lated to type I IFN and engages the heterodimeric IFN-c re-
Interferons are cytokines, which are produced in ceptor (IFNGR); it is mainly produced by activated T cells
response to viral and microbial infections, their most out- and NK cells and its gene is located on human chromosome
standing characteristic being the ability to nonspecifically in- 12 [2,3,5]. The more recently identified group of type III IFNs
hibit viral growth by inducing an ‘‘antiviral state’’ in cells. comprises IFN-k1, 2, and 3, also known as Interleukin-29 (IL-
Interferons consist of 165–208 amino acids in human, and 29), IL-28A, and IL-28B, respectively. They engage the dis-
many are posttranslationally modified by glycosylation [2,3]. tinct heterodimeric IFN-k receptor IL10R2/IFNLR1 (Table 1)
Interferon genes were cloned from mammals, birds, and and their genes are clustered on human chromosome 19.
fish, and there are hints for interferon-like activity in Similarly to type I IFN, their inducibility by virus infection
amphibians [4]. The mammalian interferons are divided into appears to be ubiquitous [6].

*Address for correspondence: Ganes C. Sen, Ph.D., Department of Molecular Genetics/


2. The induction of
NE20, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA. Tel: þ216 444 interferon synthesis
0636; Fax: þ216 444 0513; E-mail: seng@ccf.org.
Received 7 October 2008; accepted 7 November 2008 Viral infections result in the generation of pathogen-associ-
DOI: 10.1002/biof.6
Published online 2009 in Wiley InterScience
ated molecular patterns (PAMP), which, as a means of
(www.interscience.wiley.com) sensing the presence of virus, can be recognized by pattern

14
Table 1
Major components of the IFN signaling system

Type I IFN Type II IFN Type III IFN

Members IFN-a (13 subtypes) IFN-c (active as dimer) IFN-k1 (¼ IL-29)


IFN-b IFN-k2 (¼ IL-28A)
IFN-x IFN-k3 (¼ IL-28B)
IFN-e
IFN-j
IFN-d (pigs)
IFN-s (ruminants)
IFN-f/Limitin (mouse)
Receptors IFNAR1/IFNAR2c IFNGR1/IFNGR2, active as IL10R2 (¼ IL-10Rb)/IFNLR1
heterodimer two heterodimers (¼ IL-28Ra) heterodimer
Receptor-associated kinases JAK1, TYK2 JAK1, JAK2 JAK1, TYK2
Activated transcription factors ISGF3 (STAT1/STAT2/IRF-9) GAF (STAT1 homodimer) ISGF3 (STAT1/STAT2/IRF-9)
DNA binding sites ISRE GAS ISRE

recognition receptors (PRR) on the cell-surface, in the cyto- scription factors IRF-3 and IRF-7 [16–18]. The other signaling
plasm, or in endosomes. Interestingly, in most cases, the branch starting from MAVS involves a TRAF6-containing com-
ligands of these type I IFN-inducing receptors are viral plex, which mediates activation of the three kinases IKK (IjB
nucleic acids such as single-stranded (ss)RNA, double- kinase), p38, and JNK (c-JUN N-terminal kinase), terminally
stranded (ds)DNA or dsRNA, or their synthetic analogs (see resulting in the phosphorylation and activation of transcrip-
Fig. 1) [7,8]. tion factors NF-jB, ATF-2, and c-JUN, respectively [14,15,19].
Figure 2 depicts a simplified view on PAMP-stimulated The endosomal detection or dsRNA requires TLR3 (Fig. 2),
interferon induction with the example of dsRNA-activated whose dimerization allows for recruitment of the cytoplasmic
signaling pathways. DsRNA representing viral RNA genomes adapter protein TRIF (TIR domain-containing adaptor induc-
or viral replication intermediates can be sensed in the cyto- ing IFN-b). The consequent steps are quite similar to the
plasm or in endosomes by the related cytoplasmic RNA heli- RIG-I-dependent pathways described earlier [20,21]. The
cases RIG-I (retinoic acid-induced gene-I) and MDA-5 (mela- dsRNA-activated transcription factors bind to their respective
noma differentiation-associated gene-5) [9–11]. Both RIG-I DNA elements in the promoters of type I and III IFNs as well
and MDA-5 utilize the mitochondrial adaptor protein MAVS as other cytokine/chemokine genes, such as CXCL10 (IP-10)
(also known as IPS-1, VISA, or Cardif ) [12–15], which serves and CCL5 (RANTES), and activate their transcription
as a platform for the initiation of signaling cascades. One [6,22,23].
signaling branch, via a TRAF3-containing complex, activates The induction of IFN-a subtypes requires the inter-
the closely related kinases TBK1 and IKKe, who are crucial feron-induced IRF-7, whereas IFN-b can be induced without
for the induction of type I (and type III) IFN in most cell IRF-7 by the constitutively expressed IRF-3 (in concert with
types, since they phosphorylate and activate the critical tran- NF-jB and ATF-2/c-JUN). Thus, a positive feedback of type I

Fig. 1. Ligands of type I IFN-inducing pattern recognition receptors. Viral pathogen-associated molecular patterns
(PAMP) or their synthetic analogs serve as ligands for toll-like receptors (TLR) in endosomes and on the cell-surface,
and for the cytoplasmic receptors RIG-I, MDA-5, and DAI, resulting in the induction of interferon synthesis.

Interferons and Viral Infections 15


Fig. 2. Pathways of interferon induction and interferon signaling. Signaling events leading to the transcription of
interferon genes, elicited by double-stranded RNA present in the cytoplasm or in endosomes. The secreted type I
interferon subsequently acts via its cell-surface receptor, activating the ‘‘JAK-STAT pathway’’ and inducing the
expression of interferon-stimulated genes (ISG).

IFN induction takes place, for example, in fibroblasts, where 3. Interferon-activated signaling:
IFN-b induces IRF-7 in an autocrine manner, thereby driving
expression of numerous IFN-as [24]. However, the plasmacy-
Janus kinase/signal transducer and
toid dendritic cells (pDC) express IRF-7 constitutively and activator of transcription pathways
are therefore able to immediately express IFN-as upon rec- All interferons exert their effects by binding to specific ubiq-
ognition of viral PAMPs, using TLR7/8 and TLR9 as recep- uitously expressed cell-surface receptors, leading to the acti-
tors. Furthermore, the pDCs are by far the major producer of vation of signaling pathways which target the induction of a
type I IFN in reaction to viral infections in vivo [25]. large number of interferon-stimulated genes (ISG), whose
Type II (IFN-c) production does not occur in direct encoded proteins mediate the antiviral and other effects of
response to viral PAMPs, but is stimulated by IL-12 and IL- interferons. The classical components of the interferon-
18, which are cytokines released by activated macrophages. dependent ‘‘JAK-STAT pathways’’ are listed in Table 1. The
Accordingly, IFN-c production is delayed when compared monomeric type I IFN acts by assembling and engaging the
with type I IFN. IFN-c induction requires transcription factors heterodimeric receptor IFNAR1/IFNAR2c (Fig. 2). The recep-
NF-jB and AP-1 in addition to cell-specific transcription fac- tor-associated Janus tyrosine kinases TYK2 and JAK1 then
tors such as NFAT (nuclear factor of activated T cells), phosphorylate each other as well as the transcription factors
explaining its restriction to NK cells or activated T cells [26]. STAT1 and STAT2 (signal transducer and activator of

16 BioFactors
transcription). The activated STATs form a heterodimer, asso- such as Vesicular stomatitis virus (VSV), Encephalomyocardi-
ciate with IRF-9, and the ternary complex termed ISGF3 (ISG tis virus (EMCV), West Nile virus (WNV), hepatitis C virus
factor 3) translocates into the nucleus and transactivates (HCV), and DNA viruses such as Herpes simplex virus 1
ISGs by binding to the interferon-stimulated response ele- (HSV-1) [35]. Another group of IFN-induced enzymes, which
ment (ISRE) in the promoter of ISGs [27]. According to the needs dsRNA for its activation, are the 20 -50 -Oligoadenylate
current knowledge, type III IFNs, although using a different synthetases which, upon binding dsRNA, generate unique 20 -
receptor (Table 1), activate ISGF3 similarly to type I IFN [6]. 50 -linked AMP-oligomers (2-5A) from ATP. These 2-5As acti-
Type II IFN-mediated STAT1 activation occurs after bind- vate the latent RNase L, which dimerizes and cleaves cellular
ing of the dimeric IFN-c to IFNGR1, resulting in a complex and viral ssRNA, thereby inhibiting protein expression and
that is stabilized by IFNGR2. The associated JAK1 and JAK2 occasionally inducing apoptosis [3,35,38]. The OAS/RNase L
phosphorylate and activate STAT1, which homodimerizes to system is a major antiviral effector against picornaviruses
form the gamma-activated factor (GAF), which in the nucleus (EMCV) and influenza A virus, as well as other RNA viruses.
binds to the gamma-activated site (GAS) in the promoters of Depending on the nature of the virus, its genomic RNA or vi-
ISGs [27]. Since in type I IFN signaling, STAT1 homodimers ral mRNAs are cleaved [38]. Furthermore, RNase L-cleaved
are formed to a minor degree as well, the sets of genes cellular RNAs can amplify interferon induction by serving as
induced by type I or II IFNs overlap partially [28]. In fact, ligands for RIG-I [39]. Nonspecific ssRNA cleavage also
interferon-mediated gene induction is much more complex occurs after induction of ISG20, a 30 -exoribonuclease, which
than initially assumed. The other members of the STAT fam- contributes to inhibition of RNA viruses such as VSV [40]. An
ily, STAT3, 4, 5A/B, and 6, which are predominantly activated additional, nonenzymatic mechanism of translation inhibition
by other cytokines (e.g., IL-4 activates STAT6), are activated is pursued by the ISG56/IFIT family proteins P56 and P54 via
to a minor extent by interferons as well. Furthermore, un- binding to eIF3, with human P56 acting against HCV [41–43].
phosphorylated STATs (U-STAT) play important roles in tran- Another IFN-induced protein, human MxA, accumulates at
scriptional regulation of genes [29–31]. cellular membranes around the nucleus, binds to viral nu-
Importantly, numerous ISRE-driven genes can be cleocapsids, and inhibits viral intracellular trafficking. It is a
directly induced by virus-activated IRF-3 due to sequence- key component in innate defense against orthomyxoviruses
similarity of IRF elements and ISREs, thus providing a such as influenza virus, and additionally, it inhibits Measles
‘‘shortcut’’ to ISG induction as a means of an immediate virus, VSV, Hantavirus, and Semliki Forest virus, but interest-
early antiviral response in infected cells [22,23]. ingly not EMCV. Moreover, it is capable of inhibiting influ-
After their initial discovery as key components of inter- enza virus replication by binding to a viral RNA polymerase
feron signaling, a total of four JAK and seven STAT family subunit [35,44]. ISG15 is conjugated to target proteins by
proteins have been identified and recognized as components interferon-inducible conjugases/ligases, a process similar to
of major signaling pathways used by more than 30 cytokine-, ubiquitin conjugation; however, the effects of ISG15 conjuga-
growth factor-, and peptide receptors [32,33]. tion are still unclear, although they contribute to HSV-1 and
influenza virus resistance [35]. Viperin (CIG5) might interfere
with viral budding of enveloped viruses such as human Cyto-
4. Interferon-stimulated genes as megalovirus (HCMV), HCV, and influenza virus by disrupting
lipid rafts in membranes, but its mode of action remains to
effectors of IFN action be determined [45]. The nucleic acid-editing enzymes APO-
Interferons are defined by their antiviral effectiveness: on BEC3G and -3F, representing deoxycytidine deaminases, in-
one hand, viruses can be eliminated from infected cells, and hibit retroviruses; they act by introducing C to U mutations
on the other hand, uninfected cells can develop an ‘‘antiviral in viral reverse transcribed DNA or by directly interfering
state,’’ both upon interferon exposure. Usually, ISGs do not with reverse transcription [46]. Apart from the many antiviral
have virus-specificity, and therefore, replication of different effectors, signaling components such as RIG-I, TLR3, DAI,
viruses can be inhibited by the actions of the same ISG, of- IRF-7, and STAT1 are upregulated by interferons as well,
ten in concert with several other ISGs. Analyses of antiviral increasing the sensitivity of cells toward PAMPs.
ISG functions include overexpression in infected cell cul- Antibacterial effects of IFN are not clearly defined, but
tures, gene knock-out mouse models, and the identification imply the action of iNOS (inducible nitric oxide synthase)
of virus-encoded inhibitors (see later). especially in macrophages. iNOS catalyzes the oxidation of

The dsRNA-activated protein kinase, PKR, is induced by L-arginine, generating the reactive intermediate NO , which
interferon, although in many cell types, it is constitutively plays important roles in killing of bacteria, for example,
expressed at low levels. Its activation occurs after binding to Mycobacterium and Salmonella [47].
(usually virus-derived) dsRNA, inducing its dimerization and Broader effects of interferons include immunomodula-
autophosphorylation. As a mechanism of action, PKR phos- tory functions. IFN-a/b contributes to DC and macrophage
phorylates translation initiation factor 2a (eIF2a), which activation and the upregulation of MHC I molecules, whereas
results in cellular and viral translation inhibition [34,35]. The IFN-c is important for promoting the transition from innate
protein activator of PKR, PACT, can also activate PKR by to adaptive immune responses, for example, by inducing the
direct binding in response to cellular stress, inducing apo- transcription factors IRF-1 and CIITA, which governs MHC II
ptosis [36,37]. PKR inhibits replication of mostly RNA viruses expression. MHC II allows for antigen presentation to T

Interferons and Viral Infections 17


Table 2
Viral inhibitors of the IFN system

Target level Target Virus (viral protein) Effect

IFN induction dsRNA Vaccinia virus (E3L) sequestration of dsRNA


Influenza A virus (NS1) sequestration of dsRNA
Ebolavirus (VP35) sequestration of dsRNA
MDA-5 Paramyxoviruses (V) binding/inhibition of MDA-5
MAVS Hepatitis C virus (NS3/4A) degradation of MAVS
Hepatitis A virus (3ABC) degradation of MAVS
TRIF Hepatitis C virus (NS3/4A) degradation of TRIF
TBK1 Borna disease virus (P) pseudosubstrate for TBK1
New York-1 virus (Gn) prevents TRAF3 interaction
IKKe, TBK1 Mumps virus, Parainfluenzavirus 5 (V) pseudosubstrates, degradation of IKKe/TBK1
IRF-3 Rotavirus (NSP1) IRF-3 binding/degradation
Human papillomavirus 16 (E6) IRF-3 binding/inhibition
Rabies virus (P) prevents IRF-3 phosphorylation
IFN signaling Secreted IFN Poxviruses (soluble IFN-binding proteins) sequestration of IFN
JAKs Human Cytomegalovirus loss of JAK1, IRF-9
Human papillomavirus 18 (E6) binds/inhibits TYK2
STATs Paramyxoviruses loss/sequestration of STATs
Rabies virus (P) STAT1/2 binding/inhibition
Adenovirus (E1A) loss of STAT1, IRF-9
Human Cytomegalovirus (IE1-72kDa) prevents ISGF3 DNA binding
Ebolavirus inhibits STAT trafficking
ISG protein function PKR Influenza A virus (NS1) binding/inhibition of PKR
Hepatitis C virus (NS5A, E2) binding/inhibition of PKR
Adenovirus (VA-RNA) inhibits dimerization of PKR
ISG15 Influenza B virus (NS1) inhibits ISG15 conjugation
Crimean Congo hemorrhagic virus (L) de-ISG15ylation of proteins

helper cells, whose differentiation to Th1 cells is driven by role for TLR3 and interferon signaling in the central nervous
IFN-c. At the same time, IFN-c represents the marker cyto- system [57].
kine of activated Th1 cells during the adaptive immune
response. Furthermore, IFN-c regulates NK and B cell func-
tions as well as macrophage activation, the latter playing
critical roles in the microbicidal effects of IFN-c (rather than
5. Viral evasion strategies
IFN-a/b) [48,49]. The importance of the interferon system is reflected by the
Important insights into both the function and specific- large number of inhibitors or modulators encoded by most,
ity of many components of the interferon system described if not all, viruses. Table 2 gives a few examples of the diver-
earlier were obtained from murine gene knock-out studies. sity of viral inhibitors; for a comprehensive review, the
Usually, a gene deficiency for a critical component renders reader is referred to ref. [45]. The inhibitory mechanisms
the animal very susceptible to virus infections, increasing include interruptions on the levels of IFN induction, IFN sig-
morbidity and/or mortality, as seen with IFN-b, IFNAR, naling, and ISG protein functions. Induction can be inhibited
IFNGR, MAVS, IRF-3, or IRF-7 knock-out mice [50–54]. Simi- by sequestration or masking of dsRNA, binding to MDA-5,
larly, naturally occurring, rare mutations in human genes can degradation of MAVS or TRIF, forming pseudosubstrates for
result in functional deficiency of the gene. For instance, the kinases TBK1/IKKe, or by interfering with IRF-3 function
TYK2 deficiency leads to enhanced susceptibility to infection directly. Similarly, IFN signaling can be inhibited by viruses
by viruses, fungi, and mycobacteria [55]. Interestingly, mono- by inducing degradation of JAK-STAT pathway components or
genic deficiencies were found to be linked to development by neutralizing IFNs with soluble viral IFN receptors. Several
of Herpes simplex virus encephalitis (HSE), as seen in viral inhibitors even target ISG proteins such as PKR [45,58–
patients with deficiencies for STAT1 or UNC93B1, which is a 61]. Although most viruses encode one or more inhibitory
protein critically involved in directing TLR3/7/9 to endolyso- proteins, they can still induce interferon and can be inhibited
somes for signaling [56]. Indeed, HSE and human TLR3 defi- by interferon treatment, emphasizing the evolutionary bal-
ciency could be connected as well, pointing to a protective ance between the virus and the host.

18 BioFactors
[10] Yoneyama, M. and Fujita, T. (2007) RIG-I family RNA helicases: cytoplas-
6. Clinical use of interferons mic sensor for antiviral innate immunity. Cytokine Growth Factor Rev.
The antiviral and immunomodulatory effects of interferons 18, 545–551.
[11] Yoneyama, M., Kikuchi, M., Natsukawa, T., Shinobu, N., Imaizumi, T.,
have led to their use as therapeutics. Treatments of a num-
Miyagishi, M., Taira, K., Akira, S., and Fujita, T. (2004) The RNA helicase
ber of viral and nonviral diseases currently include recombi- RIG-I has an essential function in double-stranded RNA-induced innate
nant interferons. IFN-a2 is administered to patients chroni- antiviral responses. Nat. Immunol. 5, 730–737.
cally infected with hepatitis C virus and hepatitis B virus. [12] Kawai, T., Takahashi, K., Sato, S., Coban, C., Kumar, H., Kato, H., Ishii,
IFN-b is commonly used in treatment of relapsing-remitting K. J., Takeuchi, O., and Akira, S. (2005) IPS-1, an adaptor triggering
multiple sclerosis (MS) because of its beneficial effects. The RIG-I- and Mda5-mediated type I interferon induction. Nat. Immunol. 6,
981–988.
pathogenesis of MS remains largely unknown, and so are [13] Meylan, E., Curran, J., Hofmann, K., Moradpour, D., Binder, M., Bar-
the reasons for the effectiveness of IFN-b [62,63]. Proapop- tenschlager, R., and Tschopp, J. (2005) Cardif is an adaptor protein in
totic and cytostatic functions have been ascribed to interfer- the RIG-I antiviral pathway and is targeted by hepatitis C virus. Nature
ons, leading to apoptosis of certain tumor cell types and 437, 1167–1172.
size reduction of tumors, as well as inhibition of angiogene- [14] Seth, R. B., Sun, L., Ea, C. K., and Chen, Z. J. (2005) Identification and
characterization of MAVS, a mitochondrial antiviral signaling protein
sis in vivo [63]. The underlying mechanisms are not fully that activates NF-kappaB and IRF 3. Cell 122, 669–682.
understood. For example, PKR and the apoptosis-inducing [15] Xu, L. G., Wang, Y. Y., Han, K. J., Li, L. Y., Zhai, Z., and Shu, H. B. (2005)
ligand TRAIL have been implicated in apoptotic responses to VISA is an adapter protein required for virus-triggered IFN-beta signal-
interferons, whereas IFI-16, a member of the p200 family of ing. Mol. Cell 19, 727–740.
ISGs, has been related to antiproliferative effects, as it inter- [16] Saha, S. K., Pietras, E. M., He, J. Q., Kang, J. R., Liu, S. Y., Oganesyan,
G., Shahangian, A., Zarnegar, B., Shiba, T. L., Wang, Y., and Cheng, G.
acts with several transcription factors, such as p53, pRb, and
(2006) Regulation of antiviral responses by a direct and specific interac-
E2F, and inhibits their functions [45,64]. Accordingly, interfer- tion between TRAF3 and Cardif. EMBO J. 25, 3257–3263.
ons have been used for the treatment of several types of [17] Fitzgerald, K. A., McWhirter, S. M., Faia, K. L., Rowe, D. C., Latz, E.,
cancer such as certain leukemias, lymphoma, malignant mel- Golenbock, D. T., Coyle, A. J., Liao, S. M., and Maniatis, T. (2003) IKKep-
anoma, multiple myeloma, renal cell carcinoma, and Kaposi’s silon and TBK1 are essential components of the IRF3 signaling pathway.
Nat. Immunol. 4, 491–496.
sarcoma [62,65].
[18] Sharma, S., tenOever, B. R., Grandvaux, N., Zhou, G. P., Lin, R., and His-
cott, J. (2003) Triggering the interferon antiviral response through an
IKK-related pathway. Science 300, 1148–1151.
7. Conclusions [19] Munshi, N., Yie, Y., Merika, M., Senger, K., Lomvardas, S., Agalioti, T.,
and Thanos, D. (1999) The IFN-beta enhancer: a paradigm for under-
Interferons are essential for the immediate nonspecific host standing activation and repression of inducible gene expression. Cold
defense against many viruses and promote the onset of an Spring Harb. Symp. Quant. Biol. 64, 149–159.
adaptive immune response. Their antiviral, immunomodula- [20] Alexopoulou, L., Holt, A. C., Medzhitov, R., and Flavell, R. A. (2001) Rec-
ognition of double-stranded RNA and activation of NF-kappaB by toll-
tory, and antitumor effects make them effective therapeutics
like receptor 3. Nature 413, 732–738.
for clinical use; however, future research will be needed to [21] Vercammen, E., Staal, J., and Beyaert, R. (2008) Sensing of viral infec-
help understand the underlying mechanisms of their effects tion and activation of innate immunity by toll-like receptor 3. Clin.
and side-effects in order to take full advantage of their ther- Microbiol. Rev. 21, 13–25.
apeutic potential. [22] Andersen, J., VanScoy, S., Cheng, T. F., Gomez, D., and Reich, N. C.
(2008) IRF-3-dependent and augmented target genes during viral infec-
tion. Genes Immun. 9, 168–175.
[23] Elco, C. P., Guenther, J. M., Williams, B. R., and Sen, G. C. (2005) Analy-
References sis of genes induced by Sendai virus infection of mutant cell lines
[1] Isaacs, A. and Lindenmann, J. (1957) Virus interference. I. The inter- reveals essential roles of interferon regulatory factor 3, NF-kappaB,
feron. Proc. R. Soc. Lond. B Biol. Sci. 147, 258–267. and interferon but not toll-like receptor 3. J. Virol. 79, 3920–3929.
[2] Takaoka, A. and Yanai, H. (2006) Interferon signalling network in innate [24] Honda, K., Takaoka, A., and Taniguchi, T. (2006) Type I interferon [cor-
defence. Cell Microbiol. 8, 907–922. rected] gene induction by the interferon regulatory factor family of
transcription factors. Immunity 25, 349–360.
[3] Samuel, C. E. (2001) Antiviral actions of interferons. Clin. Microbiol.
[25] Gilliet, M., Cao, W., and Liu, Y. J. (2008) Plasmacytoid dendritic cells:
Rev. 14, 778–809.
sensing nucleic acids in viral infection and autoimmune diseases. Nat.
[4] Schultz, U., Kaspers, B., and Staeheli, P. (2004) The interferon system Rev. Immunol. 8, 594–606.
of non-mammalian vertebrates. Dev. Comp. Immunol. 28, 499–508. [26] Malmgaard, L. (2004) Induction and regulation of IFNs during viral
[5] Pestka, S. (2007) The interferons: 50 years after their discovery, there infections. J. Interferon Cytokine Res. 24, 439–454.
is much more to learn. J. Biol. Chem. 282, 20047–20051. [27] Platanias, L. C. (2005) Mechanisms of type-I- and type-II-interferon-
[6] Uze, G. and Monneron, D. (2007) IL-28 and IL-29: newcomers to the mediated signalling. Nat. Rev. Immunol. 5, 375–386.
interferon family. Biochimie 89, 729–734. [28] Der, S. D., Zhou, A., Williams, B. R., and Silverman, R. H. (1998) Identi-
fication of genes differentially regulated by interferon alpha, beta, or
[7] Koyama, S., Ishii, K. J., Coban, C., and Akira, S. (2008) Innate immune
gamma using oligonucleotide arrays. Proc. Natl. Acad. Sci. USA 95,
response to viral infection. Cytokine 43, 336–341.
15623–15628.
[8] Pichlmair, A. and Reis e Sousa, C. (2007) Innate recognition of viruses. [29] van Boxel-Dezaire, A. H., Rani, M. R., and Stark, G. R. (2006) Complex
Immunity 27, 370–383. modulation of cell type-specific signaling in response to type I interfer-
[9] Andrejeva, J., Childs, K. S., Young, D. F., Carlos, T. S., Stock, N., Good- ons. Immunity 25, 361–372.
bourn, S., and Randall, R. E. (2004) The V proteins of paramyxoviruses [30] van Boxel-Dezaire, A. H. and Stark, G. R. (2007) Cell type-specific sig-
bind the IFN-inducible RNA helicase, mda-5, and inhibit its activation of naling in response to interferon-gamma. Curr. Top. Microbiol. Immunol.
the IFN-beta promoter. Proc. Natl. Acad. Sci. USA 101, 17264–17269. 316, 119–154.

Interferons and Viral Infections 19


[31] Yang, J. and Stark, G. R. (2008) Roles of unphosphorylated STATs in sig- [52] Muller, U., Steinhoff, U., Reis, L. F., Hemmi, S., Pavlovic, J., Zinkernagel,
naling. Cell Res. 18, 443–451. R. M., and Aguet, M. (1994) Functional role of type I and type II inter-
[32] Levy, D. E. and Darnell, J. E. Jr. (2002) Stats: transcriptional control and ferons in antiviral defense. Science 264, 1918–1921.
biological impact. Nat. Rev. Mol. Cell Biol. 3, 651–662. [53] Sato, M., Suemori, H., Hata, N., Asagiri, M., Ogasawara, K., Nakao, K.,
[33] Murray, P. J. (2007) The JAK-STAT signaling pathway: input and output Nakaya, T., Katsuki, M., Noguchi, S., Tanaka, N., and Taniguchi, T.
integration. J. Immunol. 178, 2623–2629. (2000) Distinct and essential roles of transcription factors IRF-3 and
[34] Garcia, M. A., Gil, J., Ventoso, I., Guerra, S., Domingo, E., Rivas, C., and IRF-7 in response to viruses for IFN-alpha/beta gene induction. Immu-
Esteban, M. (2006) Impact of protein kinase PKR in cell biology: from nity 13, 539–548.
antiviral to antiproliferative action. Microbiol. Mol. Biol. Rev. 70, [54] Sun, Q., Sun, L., Liu, H. H., Chen, X., Seth, R. B., Forman, J., and Chen,
1032–1060. Z. J. (2006) The specific and essential role of MAVS in antiviral innate
[35] Sadler, A. J. and Williams, B. R. (2008) Interferon-inducible antiviral immune responses. Immunity 24, 633–642.
effectors. Nat. Rev. Immunol. 8, 559–568.
[55] Minegishi, Y., Saito, M., Morio, T., Watanabe, K., Agematsu, K., Tsu-
[36] Li, S., Peters, G. A., Ding, K., Zhang, X., Qin, J., and Sen, G. C. (2006)
chiya, S., Takada, H., Hara, T., Kawamura, N., Ariga, T., Kaneko, H.,
Molecular basis for PKR activation by PACT or dsRNA. Proc. Natl. Acad.
Kondo, N., Tsuge, I., Yachie, A., Sakiyama, Y., Iwata, T., Bessho, F.,
Sci. USA 103, 10005–10010.
Ohishi, T., Joh, K., Imai, K., Kogawa, K., Shinohara, M., Fujieda, M.,
[37] Patel, R. C. and Sen, G. C. (1998) PACT, a protein activator of the inter-
Wakiguchi, H., Pasic, S., Abinun, M., Ochs, H. D., Renner, E. D., Jansson,
feron-induced protein kinase, PKR. EMBO J. 17, 4379–4390.
A., Belohradsky, B. H., Metin, A., Shimizu, N., Mizutani, S., Miyawaki,
[38] Silverman, R. H. (2007) Viral encounters with 20 ,50 -oligoadenylate syn-
T., Nonoyama, S., and Karasuyama, H. (2006) Human tyrosine kinase 2
thetase and RNase L during the interferon antiviral response. J. Virol.
deficiency reveals its requisite roles in multiple cytokine signals
81, 12720–12729.
involved in innate and acquired immunity. Immunity 25, 745–755.
[39] Malathi, K., Dong, B., Gale, M. Jr., and Silverman, R. H. (2007) Small
self-RNA generated by RNase L amplifies antiviral innate immunity. [56] Jouanguy, E., Zhang, S. Y., Chapgier, A., Sancho-Shimizu, V., Puel, A.,
Nature 448, 816–819. Picard, C., Boisson-Dupuis, S., Abel, L., and Casanova, J. L. (2007)
[40] Espert, L., Degols, G., Gongora, C., Blondel, D., Williams, B. R., Silver- Human primary immunodeficiencies of type I interferons. Biochimie 89,
man, R. H., and Mechti, N. (2003) ISG20, a new interferon-induced 878–883.
RNase specific for single-stranded RNA, defines an alternative antiviral [57] Zhang, S. Y., Jouanguy, E., Ugolini, S., Smahi, A., Elain, G., Romero, P.,
pathway against RNA genomic viruses. J. Biol. Chem. 278, 16151–16158. Segal, D., Sancho-Shimizu, V., Lorenzo, L., Puel, A., Picard, C., Chapgier,
[41] White, C. L. and Sen, G. C. (2009) Interferons and antiviral action. InCel- A., Plancoulaine, S., Titeux, M., Cognet, C., von Bernuth, H., Ku, C. L.,
lular Signaling and Innate Immune Responses to RNA Virus Infections, Casrouge, A., Zhang, X. X., Barreiro, L., Leonard, J., Hamilton, C., Lebon,
(Brasier, A. R.,Garcia-Sastre, A., andLemon, S. M., eds.). pp.91–106, P., Heron, B., Vallee, L., Quintana-Murci, L., Hovnanian, A., Rozenberg,
ASM Press,Washington, DC. F., Vivier, E., Geissmann, F., Tardieu, M., Abel, L., and Casanova, J. L.
[42] Terenzi, F., Hui, D. J., Merrick, W. C., and Sen, G. C. (2006) Distinct (2007) TLR3 deficiency in patients with herpes simplex encephalitis.
induction patterns and functions of two closely related interferon-in- Science 317, 1522–1527.
ducible human genes, ISG54 and ISG56. J. Biol. Chem. 281, [58] Alff, P. J., Sen, N., Gorbunova, E., Gavrilovskaya, I. N., and Mackow, E.
34064–34071. R. (2008) The NY-1 hantavirus Gn cytoplasmic tail coprecipitates TRAF3
[43] Wang, C., Pflugheber, J., Sumpter, R. Jr., Sodora, D. L., Hui, D., Sen, G. and inhibits cellular interferon responses by disrupting TBK1-TRAF3
C., and Gale, M. Jr. (2003) Alpha interferon induces distinct transla- complex formation. J. Virol. 82, 9115–9122.
tional control programs to suppress hepatitis C virus RNA replication. [59] Lu, L. L., Puri, M., Horvath, C. M., and Sen, G. C. (2008) Select para-
J. Virol. 77, 3898–3912. myxoviral V proteins inhibit IRF3 activation by acting as alternative sub-
[44] Haller, O., Kochs, G., and Weber, F. (2007) Interferon, Mx, and viral strates for inhibitor of kappaB kinase epsilon (IKKe)/TBK1. J. Biol.
countermeasures. Cytokine Growth Factor Rev. 18, 425–433. Chem. 283, 14269–14276.
[45] Randall, R. E. and Goodbourn, S. (2008) Interferons and viruses: an [60] Fensterl, V., Grotheer, D., Berk, I., Schlemminger, S., Vallbracht, A., and
interplay between induction, signalling, antiviral responses and virus Dotzauer, A. (2005) Hepatitis A virus suppresses RIG-I-mediated IRF-3
countermeasures. J. Gen. Virol. 89, 1–47. activation to block induction of beta interferon. J. Virol. 79,
[46] Aguiar, R. S. and Peterlin, B. M. (2008) APOBEC3 proteins and reverse 10968–10977.
transcription. Virus Res. 134, 74–85. [61] Yang, Y., Liang, Y., Qu, L., Chen, Z., Yi, M., Li, K., and Lemon, S. M.
[47] Chakravortty, D. and Hensel, M. (2003) Inducible nitric oxide synthase (2007) Disruption of innate immunity due to mitochondrial targeting of
and control of intracellular bacterial pathogens. Microbes Infect. 5, a picornaviral protease precursor. Proc. Natl. Acad. Sci. USA 104,
621–627. 7253–7258.
[48] Biron, C. A. (2001) Interferons alpha and beta as immune regulators—a
[62] Borden, E. C., Sen, G. C., Uze, G., Silverman, R. H., Ransohoff, R. M.,
new look. Immunity 14, 661–664.
Foster, G. R., and Stark, G. R. (2007) Interferons at age 50: past, cur-
[49] Schroder, K., Hertzog, P. J., Ravasi, T., and Hume, D. A. (2004) Inter-
rent and future impact on biomedicine. Nat. Rev. Drug Discov. 6,
feron-gamma: an overview of signals, mechanisms and functions.
975–990.
J. Leukoc. Biol. 75, 163–189.
[50] Honda, K., Yanai, H., Negishi, H., Asagiri, M., Sato, M., Mizutani, T., Shi- [63] Friedman, R. M. (2008) Clinical uses of interferons. Br. J. Clin. Pharma-
mada, N., Ohba, Y., Takaoka, A., Yoshida, N., and Taniguchi, T. (2005) col. 65, 158–162.
IRF-7 is the master regulator of type-I interferon-dependent immune [64] Chawla-Sarkar, M., Lindner, D. J., Liu, Y. F., Williams, B. R., Sen, G. C.,
responses. Nature 434, 772–777. Silverman, R. H., and Borden, E. C. (2003) Apoptosis and interferons:
[51] Deonarain, R., Alcami, A., Alexiou, M., Dallman, M. J., Gewert, D. R., role of interferon-stimulated genes as mediators of apoptosis. Apopto-
and Porter, A. C. (2000) Impaired antiviral response and alpha/beta sis 8, 237–249.
interferon induction in mice lacking beta interferon. J. Virol. 74, [65] Parmar, S. and Platanias, L. C. (2003) Interferons: mechanisms of
3404–3409. action and clinical applications. Curr. Opin. Oncol. 15, 431–449.

20 BioFactors

Potrebbero piacerti anche