Cytokine & Growth Factor Reviews 13 (2002) 441–454
Survey
The role of IFN-␥ in immune responses to viral infections of the central nervous system David A. Chesler a , Carol Shoshkes Reiss a,b,c,d,∗ a
Department of Biology, New York University, 1009 Main Building, 100 Washington Square East, New York, NY 10003, USA Center for Neural Science, New York University, 1009 Main Building, 100 Washington Square East, New York, NY 10003, USA c Department of Microbiology and Kaplan Comprehensive Cancer Center, New York University School of Medicine, 550 First Ave, New York, NY 10016, USA d Department of Microbiology, Mount Sinai School of Medicine, One Gustave Levy Place, New York, NY 10029, USA b
Abstract Interferon (IFN)-␥, is not only a marker of TH 1 CD4, CD8 and natural killer (NK) cells, it is also a critical antiviral mediator which is central to the elimination of viruses from the CNS. In this review, we describe IFN-␥, its receptor, signal transduction from receptor engagement, and antiviral downstream mediators. We demonstrate that although neurons are post-mitotic and non-renewing, they respond to IFN-␥ in a fashion similar to peripheral fibroblasts or lymphocytes. We have illustrated this review with details about studies on the role(s) of IFN-␥ in the pathogenesis of measles virus (MV), herpes simplex virus (HSV) type 1, and vesicular stomatitis virus (VSV) infections of the CNS. For VSV infection, IFN-␥ signals through Jaks 1 and 2 and STAT1 to activate (interferon regulatory factor) IRF-1; although viral protein synthesis is inhibited, PKR is not a critical mediator in the antiviral response to VSV in murine neurons. In contrast, induction of nitric oxide synthase (NOS) type 1 and its production of nitric oxide is essential in the elimination of viruses from neurons. © 2002 Elsevier Science Ltd. All rights reserved. Keywords: Interferon-␥; Neuron; Virus; Antiviral; Nitric oxide
Contents 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2. Molecular biology of IFN-␥ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.1. IFN-␥ expression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.2. IFN-␥ receptor expression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.3. IFN-␥ intracellular signaling. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3. Nitric oxide synthase (NOS) and nitric oxide (NO) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4. Interferon-␥ initiated antiviral responses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.1. Mx . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.2. RNAseL/2 -5 OAS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.3. PKR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.4. NOS and NO . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.5. Undefined and poorly characterized interferon-γ -mediated antiviral responses . . . . . . . . . . . . . .
442 442 442 442 442 443 444 445 445 445 446 446
Abbreviations: 2 -5 OAS, 2 -5 -oligoadenylate synthetase; 2-AP, 2-aminopurine; 7-NI, 7-nitroindazole; C/EBP-, CCAAT/enhancer-binding protein-; CD, cluster of differentiation; CMV, cytomegalovirus; CNS, central nervous system; CREB, cAMP/Ca2+ response element-binding protein; CIITA, class II transactivator; dsRNA, double-stranded RNA; EAE, experimental autoimmune encephalomyelitis; eIF2␣, eukaryotic initiation factor-2␣; GAS, ␥-activation sequence; GATE, ␥-IFN-activated transcriptional element; GFAP, glial fibrillary acidic protein; GTPase, guanine triphosphate nuclease; HIV, human immunodeficiency virus; HSV, herpes simplex virus; IFN, interferon; IL, interleukin; IP-10, interferon-inducible protein of 10 kDa; IRF, interferon regulatory factor; JAK, Janus kinase; LPS, lipopolysaccharide; MAPK, mitogen-activated protein kinase; MHC, major histocompatibility complex; MS, multiple sclerosis; MV, measles virus; NK, natural killer; NO, nitric oxide; NOS, nitric oxide synthase; NS1, non-structural protein 1; ONOO• , peroxynitrite; PKR, dsRNA protein kinase; RNA, ribonucleic acid; RNaseL, ribonucleic acid nuclease L; SSPE, subacute sclerosing panencephalitis; STAT, signal transducer and activation of transcription; TH 1, type 1 helper cell; TNF-␣, tumor necrosis factor-␣; viperin, virus inhibitory protein, endoplasmic reticulum associated, interferon-inducible; VSV, vesicular stomatitis virus ∗ Corresponding author. Tel.: +1-212-998-8269; fax: +1-212-995-4015. E-mail address:
[email protected] (C.S. Reiss). 1359-6101/02/$ – see front matter © 2002 Elsevier Science Ltd. All rights reserved. PII: S 1 3 5 9 - 6 1 0 1 ( 0 2 ) 0 0 0 4 4 - 8
442
D.A. Chesler, C.S. Reiss / Cytokine & Growth Factor Reviews 13 (2002) 441–454
5. Viral infections in the CNS and the effects of IFN-γ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.1. Vesicular stomatitis virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.2. Measles virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.3. Herpes simplex virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgements. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
1. Introduction During viral infections in the periphery as well as in the CNS, the goal of the host immune response is to prevent virus replication and, failing that, to eliminate virus-infected cells. In the periphery, this is accomplished mainly through the targeted destruction of virally infected cells by NK cells, CD8+ T-cells, and CD4+ T-cells and much later, the production of neutralizing antibody [1]. Because of the fundamental danger to the host of destruction of non-renewing neurons, non-cytolytic mechanisms which eliminate virus are critical for CNS infections [2,3].
2. Molecular biology of IFN-␥ 2.1. IFN-γ expression The IFN family of pro-inflammatory cytokines, first described 30 years ago because of their antiviral properties [4,5], are categorized into two distinct groups based primarily on their protein sequences, cellular sources, and use of distinct receptors. Type I IFNs, IFN-␣, -, and -, are expressed by both leukocytes and fibroblasts largely as a direct result of viral infection [6]. While IFN-␣ and - are found across many species including humans and mice, IFN- expression is restricted to humans [6]. An additional type I IFN, IFN-, was identified in ruminants. Rather than possessing antiviral properties, IFN- acts as an early signal in ruminant pregnancy [7–11]. In contrast, type II family of IFNs, contains a single member, IFN-␥ [12,13]. IFN-␥ is a biologically active non-covalently linked 34 kDa homodimer secreted primarily by NK) cells, TH 1 CD4+ T-cells, and CD8+ T-cells. IFN-␥ mediates a wide range of immunomodulatory effects on both innate and acquired immunity [12]. IFN-␥ has been shown to upregulate major histocompatibility complex (MHC)-I and -II expression, activate macrophages/microglia in an antigen-specific fashion, act as a cytostatic agent on numerous normal and oncogenic cell lines, and induce several IFN-inducible antiviral mechanisms [12]. Although IFN-␥ expression within the CNS has been traditionally attributed to infiltrating NK cells and CD4+ T-cells, reports from the literature indicate that there may be endogenous sources of expression. Of particular interest are reports of an IFN-␥-like product produced by neu-
446 446 448 448 449 449 449
rons [14–17]. These studies identified a 60 kDa unglycosylated protein termed neuronal-IFN-␥ (N-IFN-␥) by its cross-reactivity with several antibodies specific for IFN-␥ [14,15,17,18]. Purified N-IFN-␥ was found to have bioactivity similar to recombinant IFN-␥ including MHC-I and -II induction on macrophages and potent proliferative stimulation of the intracellular pathogen Trypanosoma brucei brucei [17]. N-IFN-␥ is expressed in IFN-␥−/− mice and is induced during VSV encephalitis (Hodges and Reiss, New York University). 2.2. IFN-γ receptor expression The IFN-␥ receptor (IFN-␥R) is composed of two subunits, an ␣ (IFN-␥R1 or IFN-␥R␣) and  chain (IFN-␥R2 or IFN-␥R), that oligomerize into a heterotetramer consisting of two ␣ and two  chains upon binding of its ligand [12]. While the IFN-␥R has been previously considered to be ubiquitously expressed on all nucleated cells with constitutive levels of the IFN-␥R␣ and regulated expression of the IFN-␥R [12], Robertson et al. [19] have revealed a restricted pattern of IFN-␥R immunoreactivity in the CNS. In examining IFN-␥R expression in neuronal processes, and therefore, regions where IFN-␥ could affect neurons directly, their study found constitutive expression of IFN-␥R in the normal rat brain is limited primarily to the olfactory bulb, limbic system, hypothalamic structures, and brain stem [19]; though in contrast to other studies [20,21], Robertson et al. [19] failed to find IFN-␥R expression in glial components. Using the upregulation of Fos expression as marker for IFN-␥-mediated activation, intracerebral injection resulted in increased Fos immunoreactivity predominantly in areas co-localizing with IFN-␥R expression. It would appear based on the neuronal distribution of IFN-␥R expression and IFN-␥-mediated induction of Fos in the CNS that IFN-␥ serves to play a role early in the immune response to limit the spread of an intracellular pathogen such as a virus. 2.3. IFN-γ intracellular signaling IFN-␥ signal transduction is made up of primary and secondary responses. STAT1 [12], mediates the primary signaling response. After ligand engagement of the IFN-␥R, STAT1 is phosphorylated by JAK1 and JAK2, dimerizes, and subsequently translocates to the nucleus where it
D.A. Chesler, C.S. Reiss / Cytokine & Growth Factor Reviews 13 (2002) 441–454
443
Fig. 1. IFN-␥ receptor and intracellular signaling cascade. Cytokine-binding induces the formation of the heterotetrameric receptor complex, activating the JAK/STAT kinase cascade ultimately regulating gene expression in the nucleus.
induces transcription of a subset of IFN-␥-inducible genes (Fig. 1). Included in that subset of genes are additional transcription factors such as interferon regulatory factor (IRF)-1 and class II transactivator (CIITA), which are responsible for mediating secondary IFN-␥ responses including the upregulation of MHC-I and -II, expression of the chemokine interferon-inducible protein of 10 kDa (IP-10), and antiviral mechanisms [22]. Ancillary signaling pathways have also been described for IFN-␥. IFN-␥ has been found to induce the phosphorylation of p42/p44 MAPK (ERK1/2) through Jak1 and Raf [23]; activation of Raf leads to cytoskeletal plasticity, frequently seen as morphological changes in IFN-treated cells. In addition, IFN-␥ has been found to regulate inducible genes such as p48/IRF-9 through a novel cis-acting promoter element termed the ␥-IFN-activated transcriptional element (GATE) [24,25] by the CCAAT/enhancer-binding protein- (C/EBP-) [24–26]. The large majority of IFN-␥-inducible responses are attributed to STAT1-dependent mechanisms though recent reports have demonstrated a number of STAT1-independent responses. A subset of IFN-␥-inducible genes are regulated in the absence of STAT1. In some of these instances, gene regulation is identical in WT and STAT1−/− cells though interestingly in others, STAT1 was found to be a repressor of gene expression [26–28].
3. Nitric oxide synthase (NOS) and nitric oxide (NO) Nitric oxide (NO) is a small, reactive, highly diffusible gas which is important in many aspects of physiology and antiviral responses [29,30]. NO is formed by the NOS-mediated conversion of l-arginine to l-citrulline (Fig. 2). Three isoforms of NOS encoded by distinct genes have been identified. All three isoforms possess interaction domains for calmodulin, NADPH, flavin adenine dinucleotide, flavin mononucleotides, and an N-terminal heme site [30]. Table 1 provides a brief summary of the properties of the three NOS isoforms. The neuronal isoform (nNOS, bNOS, NOS-1) is constitutively expressed. It exists as a cytosolic homodimer and its activity is dependent on calcium and calmodulin [31]. Though it is constitutively expressed, its expression level can be modulated under many circumstances including in response to cytokine treatment [32] and following ischemic/reperfusion injury [33]. NOS-1 expression throughout the CNS is involved in many physiologic processes including long-term potentiation [34–36], nocioception [37,38], and neurotransmitter release [39]. NOS-2 (macNOS, iNOS) is not expressed constitutively but is rapidly induced by IFN-␥, TNF-␣, IL-12, and LPS. Like NOS-1, it exists as a cytosolic heterodimer [31] but in contrast to NOS-1, is Ca2+ -independent and found constitu-
444
D.A. Chesler, C.S. Reiss / Cytokine & Growth Factor Reviews 13 (2002) 441–454
Fig. 2. Synthesis of nitric oxide from l-arginine and the production of reactive by-products. NOS catalyzes the conversion of l-arginine to NO and l-citrulline. NO acts through subsequent reactions to form reactive by-products. Table 1 NOS isoform properties and expression profiles in the CNS Isoform
CNS expression
Regulation and activity
NOS-1 (nNOS or bNOS)
Neurons
NOS-2 (macNOS or iNOS)
Microglia, astrocytes, macrophages
NOS-3 (eNOS)
Endothelium, astrocytes, neurons
Ca2+ -dependent, constitutively expressed and stimuli modulated (IFN-␥, IL-12, TNF-␣, Ca2+ ), low capacity for NO production Ca2+ -independent, non-constitutively expressed; inducible by IL-12, IFN-␥, TNF-␣, LPS and others; high capacity for NO production Ca2+ -dependent, constitutively expressed and stimuli modulated by cytokines and estrogens, low capacity for NO production
tively associated with calmodulin [31]. Its CNS expression is restricted to microglia and astrocytes along with infiltrating, inflammatory macrophages [40–43]. NOS-2 has the capacity to produce large bursts of NO that greatly exceed the amount produced by either of the two other NOS isoforms [44,45]. The endothelial isoform of NOS (NOS-3, eNOS) like NOS-1, is constitutively expressed and calcium and calmodulin dependent. It is expressed by most endothelial cells and a subset of neurons in the CNS [46]. Astrocytes also have the capacity to express NOS-3 [47]. NOS-3-dependent production of NO has been shown to have a central role in the regulation of blood pressure [48]. NO participates in numerous, complex biological activities, the best known of which is its ability to induce the relaxation of vascular smooth muscle cells [49]. NO has the ability to interact both directly with targets and indirectly through the formation of reactive intermediates. The free radical, NO• , can directly nitrosylate thiol groups of cysteine residues leading to numerous products. Under biologically relevant conditions, NO can react with molecular oxygen (O2 ), superoxide anions (O2 − ), and transition metals to form reactive nitrogen species (NOx ), highly reactive peroxynitrite (ONOO• ), and metal adducts, respectively. Through direct action, or the formation of these reactive intermediates, NO is involved in a wide range of biologic activities both normal and pathologic. NO has been demonstrated to be a key component of host defenses against pathogens including bacteria, fungi, viruses, and protozoan
parasites [29,50–54]. Excessive production of NO has been associated with disease pathology in amyolaterotrophic sclerosis (ALS) [55], Huntington’s chorea, Alzheimer’s disease and cerebral ischemia [56]. NO has also been implicated as a contributor to pathology in multiple sclerosis though this finding is still controversial [57].
4. Interferon-␥ initiated antiviral responses Work published previously by the Reiss laboratory has shown that IFN-␥ treatment can reduce viral titers in infected neuronal cells in vitro by up to 100-fold compared with untreated cells demonstrating its potent antiviral activity [32]. This effect is not restricted solely to VSV but can be demonstrated in CNS infections with other viruses including Sindbis virus, polio virus, influenza virus [32,58], and measles virus (MV) [15,59]. In the case of other viruses such as herpes simplex virus type 1 (HSV-1), IFN-␥ has been shown to limit transynaptic transmission [60], prevent viral reactivation [61], and inhibit virally induced apoptosis [62]. The literature on the role of IFN-␥ and IFN-␥R in viral infection is summarized in Table 2. Several IFN-␥ regulated antiviral mechanisms have been defined and described. In mice the most prominent mechanisms include Mx, ribonucleic acid nuclease (RNase)L, 2 -5 -oligoadenylate synthetase (2 -5 OAS), double-stranded (ds)RNA protein kinase (PKR), and NOS [63–66]. Important in considering the contribution and effectiveness of these
D.A. Chesler, C.S. Reiss / Cytokine & Growth Factor Reviews 13 (2002) 441–454
445
Table 2 Consequences of IFN-␥ and IFN-␥R deficiencies on viral infections of the CNS Virus
IFN-␥−/−
IFN-␥R−/−
HSV
Increased morbidity and mortality compared with WT [61,62,172–174]
VSV
No differences compared with WT [176] (Hodges and Chen, New York University) Increased morbidity and mortality [15,59] No differences compared with WT [177,178] Increased susceptibility to infection [180]
Increased morbidity and mortality compared with both WT and IFN-␥−/− [61,173], increased complications in patients with IFN-␥R−/− immunodeficiency [175] No differences compared with WT (Hodges, Chen, and Reiss, New York University). – Increased susceptibility to infection compared with WT [179] No initial difference compared to WT though increased late pathology [181]
Measles virus LCMV Theiler’s murine encephalitis virus
Table 3 IFN-␥-induced antiviral mechanisms Antiviral protein
Mechanism of action
Susceptible viruses
Mx RNaseL/2 -5 OAS
Inhibition of transcription of viral RNA Synthesis of oligoadenylate chains by 2 -5 OAS in response to dsRNA which activates RNaseL to degrade ssRNAs halting virus replication Phosphorylation of eIF2␣ in response to dsRNA resulting in inhibition of protein synthesis Targeted nitrosylation of cysteine and tyrosine residues and/or disruption of sulfhydryl-bonds in viral proteins
Measles [15,166]; influenza [182,183]; VSV [184] ECMV [185]; HIV [186]; vaccinia virus [187]
PKR NOS
ECMV [188]; HIV [189]; VSV? [190,191] VSV [149]; HSV-1 [149]; HIV [192]; EBV [193]; Coxsackie [194]; vaccinia [158]
different antiviral mechanisms in the IFN response is that none of them act ubiquitously. Specifically, the contribution of IFN-mediated antiviral responses, is pathogen-specific (Table 3). What might be effective against one virus will not necessarily have similar or comparable effects against another. This is not surprising considering the range of DNA and RNA virus-host cell replication patterns involved as well as viral evasion proteins.
a hallmark of many viral infections [72–77]. 2 -5 OAS synthesizes oligoadenylate chains in response to dsRNA binding acting as an activator of RNaseL. Binding of dsRNA by 2 -5 OAS in turn activates RNaseL to target and degrade single-stranded RNA species inhibiting viral replication [78]. This effect is exemplified during infections with encephalomyocarditis virus (ECMV) [79], HIV [80], and vaccinia virus [81].
4.1. Mx
4.3. PKR
Mx proteins are IFN-inducible, large GTPases belonging to the dynamin superfamily [63]. Depending on the species, they are localized to either the cytosol or nucleus [67]. The antiviral action of Mx proteins varies greatly depending on the virus being investigated. For example, the murine Mx1 protein is a nuclear localized protein that efficiently inhibits the replication of influenza A and other members of the orthomyxovirus family by blocking primary transcription [68,69]. In contrast, the human MxA protein also inhibits Influenza A replication though at a later stage in the replicative cycle [69]. Mx has also been demonstrated to inhibit VSV replication at the transcriptional level in both human and murine models [70,71]. The known genes encoding Mx proteins are absent in most commonly used laboratory mouse strains such as BALB/c, B6 and C3H, but is present in the A2G strain [64].
Perhaps one of the best-studied antiviral enzymes, PKR, an interferon-inducible serine/threonine kinase, is ubiquitously expressed. Activated by autophosphorylation in the presence of dsRNA, PKR exerts its antiviral effects primarily through the phosphorylation of one its many target substrates, the ␣-subunit of the eukaryotic initiation factor 2␣ (eIF2␣) [66]. This phosphorylation leads to the sequestration of eIF2B and the global inhibition of protein synthesis [66]. PKR has been demonstrated in vitro to be important in the inhibition of encephalomyocarditis virus [82], HIV [83], and VSV replication [84–87]. Highlighting the importance of PKR is the evolution of IFN evasion mechanisms in a wide range of viruses that are aimed at circumventing PKR action. The influenza A protein, NS1, interacts with PKR to prevent its activation and action [88]. In hepatitis C virus infections, similar effects are found; the NS5A and the E2 protein have been implicated as inhibitors of PKR [89,90]. Vaccinia virus has also developed a mechanism for the inhibition of PKR through the virally encoded E3L and K3L proteins [73,91]. Deletion of the E3L protein results IFN susceptibility during
4.2. RNAseL/2 -5 OAS Expression of RNaseL and 2 -5 OAS is regulated by IFN-␥ and activated in response to the presence of dsRNA,
446
D.A. Chesler, C.S. Reiss / Cytokine & Growth Factor Reviews 13 (2002) 441–454
vaccinia virus infection [92]. A PKR-antagonistic function has not been attributed to any of the gene products encoded by VSV. 4.4. NOS and NO NOS and its product, NO, and its reaction product, ONOO− , have been implicated as important antiviral mechanisms for a wide range of viruses. NO has been found to be an effective antiviral molecule in studies using DNA and RNA viruses belonging to the Picornaviridae, Flaviviridae, Coronaviridae, Rhabdoviridae, Reoviridae, Retroviridae, Parvoviridae, Herpesviridae, and Poxviridae families demonstrating its broad spectrum of effectiveness [54,93]. Several mechanisms have been proposed for the antiviral properties of NO. The predominant mechanism underlying the antiviral properties of NO is the nitrosylation of viral proteins during infection [93–95]. NO chemically modifies cysteine and tyrosine residues presumably disrupting protein function either through the impairment of folding and/or the disruption of sulfhydryl-bonds within the protein. Nitrosylation by ONOO• does not appear to be random but rather to target-specific viral proteins. In Coxsackie virus infection, a member of Picornaviridae, nitrosylation of catalytic residues in its two cysteine proteases leads to the interruption of its life cycle [96–98]. Similar effects have been attributed to the nitrosylation of the HIV-1 aspartyl protease [99]. The reverse transcriptase of HIV-1 has also been identified as a target of NO action [100]. Putative targets of NO action have also been identified in vaccinia virus [101], ectromelia virus [102], Epstein-Barr virus [98], VSV [103], and HSV-1 [104]. 4.5. Undefined and poorly characterized interferon-γ -mediated antiviral responses Though a substantial body of research exists characterizing the roles and contributions of the IFN-mediated antiviral responses described above, a number of recent studies have hinted at additional, previously unidentified mechanisms of inhibition. A systematic examination of the known IFN induced antiviral mediators (Mx, PKR, RNAseL, and NO) during murine cytomegalovirus (CMV) infection of bone marrow macrophages revealed one such mechanism [105]. IFN-␥ has been found to inhibit early events in the replication of murine CMV demonstrated by the reduction in immediate early (IE) gene expression [106]. These results demonstrated that IFN-␥ is able to inhibit the replication of murine CMV and IE gene expression through a cis-acting, IFN-␥R- and STAT1-dependent mechanism that was independent of PKR, RNAseL, Mx and NO. Using triply-deficient (PKR−/− , RNAseL−/− , Mx−/− ) mice and primary dendritic cell cultures, Ryman et al. [107] found that the IFN-inducible antiviral response to Sindbis virus infection was left largely intact.
One protein, recently implicated as an IFN-inducible protein, is vig1/cig5 or viperin (virus inhibitory protein, endoplasmic reticulum associated, interferon-inducible) [108,109]. Boudinot et al. [108] demonstrated the induction of vig1/cig5 during VSV and pseudorabies virus infection of murine dendritic cells. VSV infection caused the direct induction of cig5, in contrast to pseudorabies virus infection, which required an intact IFN response for induction. Evidence for the antiviral properties of cig5 came from the studies of Chin and Cresswell [109] in which over-expression of the viperin protein inhibited the productive infection of fibroblasts with human CMV. It is unclear how viperin may act to inhibit viral replication but the observation of its close association with the endoplasmic reticulum and Golgi complex suggest it might function through impeding the trafficking of viral proteins during replication.
5. Viral infections in the CNS and the effects of IFN-␥ While a growing knowledge of the mechanisms through which CNS infections are dealt with immunologically, a greater appreciation for the consequences of viral infections in the CNS is evolving. A large number of laboratories are focusing their research on both the inflammatory mediators involved in immune responses to, and the consequences of viral infections within the CNS. Attention has been directed to measles virus, which rarely can produce subacute sclerosing panencephalitis (SSPE), a frequently fatal syndrome characterized by chronic inflammation, deterioration of intellectual capacity and motor degeneration [110,111]. Through the infection and super activation of microglia, HIV infection has been found to cause AIDS associated dementia in about a third of HIV patients [112–114]. Borna virus infection has also been associated with behavioral and cognitive disruption in rats although this still remains controversial in human disease [115,116]. In addition to damage done to the CNS as a direct consequence of CNS infections, several investigators have worked to establish causal links between viral infections and autoimmune disease in the CNS using model systems. Semliki forest virus and Theiler’s murine encephalomyelitis virus have been studied extensively as viral inducers of experimental autoimmune encephalomyelitis, a murine model for the study of multiple sclerosis [117–120]. In spite of the fact no direct causal link has yet been shown between viral infections and multiple sclerosis, many experimental findings point in this direction (for recent reviews on viruses in autoimmune disease, see [121,122]). 5.1. Vesicular stomatitis virus The main focus of the research performed in our laboratory is to understand and characterize immune responses in the CNS during acute viral encephalitis. To accomplish
D.A. Chesler, C.S. Reiss / Cytokine & Growth Factor Reviews 13 (2002) 441–454
this, the vesicular stomatitis virus (VSV) is being used. VSV, is an enveloped, negative-sense, single-stranded RNA virus belonging to the family Rhabdoviridae [123]. The genome of VSV encodes five proteins: a variable glycoprotein, a matrix protein, a nucleoprotein, a large protein, and two phosphoproteins in overlapping reading frames [123]. Two principle serotypes have been identified for VSV: New Jersey and Indiana [124]. Cows and horses are the major natural hosts of VSV, and it is transmitted primarily through arthropod vectors such as the sandfly. In its natural host, infection is mild, characterized by vesicular lesions in the oral cavity. A wide range of studies have been carried out using VSV as a model system including: viral assembly [125], endogenous antigen presentation by MHC-II [126], viral entry [127], membrane fusion [128], defective interfering viral particles [129,130], viral immunity [131,132], and viral inhibition by interferons [32]. Intranasal instillation with VSV has been shown to lead to lethal infection of the CNS [133], which has allowed its use as a model for studies of neurotropic viral infection [131,134–140]. Intranasal infection of mice with VSV results in the initial infection of the olfactory neuro-epithelium [141]. Within 12–24 h, the virus is transmitted to neurons within the olfactory bulb, and the infection spreads caudally through the CNS ultimately reaching the spinal cord [131,135,137]. During infection, the respiratory epithelium is spared and virus cannot be isolated from nearby peripheral neurons such as the trigeminal ganglion [141]. Mice surviving the infection completely clear the virus from the CNS by day 12 post-infection [131]. We have found that infection with VSV activates and requires both innate and acquired immune responses. In characterizing the contribution of T-cell-mediated mechanisms, antibody depletion of CD4+ or CD8+ was found to increase severity of infection compared with control animals [136]. Infection of athymic (nu/nu) mice demonstrated 10-fold greater viral titers and increased rates of mortality compared to their euthymic littermates [136]. In addition, BALB/c-H-2dm2 (dm2) mice, which lack the MHC-I restricting H-2Ld molecule, were found to mount an effective CD4+ antiviral response [131,142]. The dm2 mice were more resistant to viral challenge than BALB/c control mice indicating that CD4+ T-cells are sufficient for a response to VSV infection. Characterization of kinetics of the immune response to VSV demonstrated an absence of B-cells within the CNS until after the infection was cleared [138–140,143]. While antibody production contributes to immunity against re-challenge with VSV, it is unlikely to be important in control of the initial infection. Furthermore, we have established that TH 1-like responses, characterized by the production and action of cytokines such as IL-12 and IFN-␥, are advantageous to host recovery [144] (for a review of the role of IL-12 in VSV encephalitis, see [145]). Current work in our laboratory is aimed at understanding the role of IFN-␥ during viral infection and eluci-
447
dating the mechanisms through which IFN-␥ establishes an antiviral state against VSV infection in neurons. In neuroblastoma cells and primary neurons, we have found that IFN-␥ can rapidly induce cellular changes resulting in a significant inhibition of VSV replication. Though IFN-␥ is a potent inducer of an antiviral response in this system, it is not a requirement for host-survival. Mice deficient in either IFN-␥ or IFN-␥R are equally susceptible to VSV encephalitis as their syngenic wild-type controls illustrating the redundancy of the inflammatory immune response [146] (Hodges, Chen, and Reiss, New York University). Both in vitro and in vivo, shown through the use of specific NOS-1 inhibitors such as 7-nitroindazole (7-NI), a selective NOS-1 inhibitor [32,147], and NOS-1-deficient animals, NO-mediated antiviral effects are central to the clearance of VSV infection in the CNS [32,140]. This presents an interesting finding because of the limited capacity for NO production by NOS-1. In contrast to the inducible isoform of NOS (NOS-2), which can produce M quantities of NO [45], NOS-1 is capable of only producing nM amounts of NO [148], generally held to be insufficient in immune responses. What appears to allow for this antiviral effect is the upregulation of NOS-1 expression following IFN-␥ treatment (data not shown). The increases in NOS-1 expression observed correlate closely with increases in NO production and increased inhibition of VSV replication (data not shown) [149]. The question remains of how IFN-␥ is able to effect these changes. Increasing intervals of IFN-␥ treatment result in greater inhibition of VSV replication. This effect correlates well with the observed increases in NOS-1 protein expression. Protein synthesis experiments demonstrate that short periods of IFN-␥ exposure induce changes in NOS-1 synthesis and increase the stability of the NOS-1 protein. Accordingly, short intervals of IFN-␥ are sufficient to establish an antiviral state. IFN-␥ signaling can be divided into primary and secondary events. Initial signaling from the IFN-␥ receptor takes place through Janus kinases and STAT1 [22,150]. Activated STAT1 can in turn translocate to the nucleus resulting in the transcription of IFN-inducible genes including secondary signaling components such as the IRF-1, which is itself a transcription factor [151]. In addition, IFN-␥ has been shown to activate ERK1/2 through a Jak1-dependent mechanism and C/EBP- [25,26,152,153]. As previously described in a number of cell types, IFN-␥ signaling in neurons also occurs through the phosphorylation of STAT1 and/or ERK along with the induction of IRF-1 (unpublished observation). Sequence analysis of available murine NOS-1 promoter sequences has revealed several putative GAS and IRF-1-binding sequences. This suggests that IFN regulation of NOS-1 and ultimately the antiviral state against VSV would be dependent on the transcriptional regulation of NOS-1 (Fig. 3) [154,155]. Unlike the majority of genes known to be IFN-␥-inducible, NOS-1 is regulated post-transcriptionally through increases in both protein synthesis and stability (data not shown). Two possibilities are
448
D.A. Chesler, C.S. Reiss / Cytokine & Growth Factor Reviews 13 (2002) 441–454
Fig. 3. Putative IRF-1 and STAT-binding sites in the NOS1 promoter. Structure of NOS-1 exons 1a and 1b and sequence 5 . Genbank Accession #AF135825. Using the MatInspector Software Package [154], a putative IRF-1-binding site (GAAAAATGAGAGC) was identified at −2078 → −2066 and a putative STAT half-site (TTTCCAGAA) at −368 → −360. Above the gene schematic is a scale identifying the locations of features of interest based upon the submitted GENBANK sequence.
readily presented by this finding. On the one hand, this could indicate a novel mechanism for IFN-␥-mediated gene regulation suggesting the action of undefined signaling pathways in neurons. On the other hand, this might indicate that there is a missing component to the observed response. An intermediate induced by IFN-␥ may in turn act to regulate the increased expression of NOS-1. Kinetics studies unfortunately do not clarify the situation. Rapid changes in neurons caused by IFN-␥ induces the synthesis of NOS-1 as well as the observed antiviral effect. Supporting the latter hypothesis, is in vitro evidence using STAT1−/− primary neurons, demonstrating a requirement for STAT1 in the IFN-␥-mediated antiviral response to VSV infection. 5.2. Measles virus Measles virus, a paramyxovirus, is considered in most cases, a relatively harmless childhood disease preventable by proper and timely vaccination. In very rare cases however, MV can gain access to the CNS resulting in a range of severe neurologic complications. Among these complications are acute disseminated encephalomyelitis, a monophasic autoimmune disease with immune responses against myelin basic protein similar to that documented in mice with experimental autoimmune encephalomyelitis [156], measles inclusion body encephalitis (MIBE), and SSPE [157,158]. Both MIBE and SSPE are progressive diseases due to persistent CNS infection, neither of which is currently treatable or curable. Though, a large body of literature exists characterizing the clinical findings and significance of the neurologic complications of MV infection, little is known about the immune response to CNS MV infection. Clinical observations indicate that T-cell-mediated immunity is crucial in patients recovering from acute infection. Both CD4+ and CD8+ T-cell clones recognizing MV epitopes can be isolated from patients [159,160]. Furthermore, individuals with suppressed
T-cell function are more susceptible to complications [161]. Studies in murine models of MV infection of the CNS using mouse adapted MV strains, or more recently using human CD46 (the co-receptor for MV) transgenic mice, have shown a requirement in vivo for CD4+ but not CD8+ T-cells in clearance of MV from infected neurons, a result similar to that found with VSV infection [59,162,163]. Unlike VSV infection however, IFN-␥ is required in mice to control and clear MV from infected neurons. BALB/c mice treated with an anti-IFN-␥ neutralizing antibody are more susceptible to MV encephalitis compared with controls [59]. Furthermore, human CD46+ transgenic mice deficient in IFN-␥ (IFN-␥−/− ) show similar results with significant increases in morbidity and mortality compared with IFN-␥-competent CD46+ controls [15]. The mechanisms through which MV replication is inhibited are not clear although evidence indicates a requirement for MxA in the IFN-␥-induced antiviral response in mice [164–166]. 5.3. Herpes simplex virus HSV-1 and -2 are large DNA viruses both of which are highly prevalent in the human population. Whereas initial infection with HSV-1 principally occurs in early childhood, HSV-2 infection is primarily spread through sexual contact. In contrast to both MV and VSV infection which are characterized as being acute infections (the exception being persistent MV infection leading to disorders such as SSPE); HSV infection is a life-long state in which latency is established in ganglia following an initial acute disease. In both childhood and adult HSV infection disease is characterized by recurrences or outbreaks which resolve in a timely manner but have little risk of mortality due to the focused nature of the infection. Neonatal infection with HSV, on the other hand, is almost always symptomatic and frequently lethal. The lethality of neonatal infection is attributed to encephalitis or disseminated infection involving multiple
D.A. Chesler, C.S. Reiss / Cytokine & Growth Factor Reviews 13 (2002) 441–454
organ systems including the CNS [167,168]. With few exceptions, patients surviving neonatal HSV encephalitis demonstrate severe neurologic deficits [169]. IFN-␥ has the ability to affect HSV infected neurons in a number of ways. In vitro replication of HSV-1 in neuroblastoma cells is inhibited by IFN-␥ through a nitric oxide-independent mechanism [32]. Mice expressing IFN-␥ through a rhodopsin promoter driven transgene are less susceptible to lethal HSV-1 encephalitis despite an increase in inflammation due to the expression of IFN-␥ [170,171]. The demonstrated increase in survival in IFN-␥ transgenic mice was suggested to be due in part to an upregulation of the anti-apoptotic gene, Bcl2, which promoted cell survival compared to HSV infected wild-type animals [62]. Consistent results were found in IFN-␥−/− mice exhibiting greater numbers of apoptotic neurons and decreased levels of Bcl2 expression [172]. IFN-␥ has also been implicated in having a role in limiting the axonal transmission of HSV from infected neurons to epidermal cells [60] and importantly, IFN-␥ has been found to inhibit the reactivation of latent HSV in the trigeminal ganglion [5,61].
6. Conclusions IFN-␥ is critical for antiviral responses not only in the periphery, but also in the CNS. The mechanisms of action are tailored to the infection and range from Mx in myxovirus (influenza) infections to PKR in hepatitis C virus infection to induction of NOS for VSV and Coxsackie virus infections. HSV-1 latency may also be maintained in the presence of IFN-␥. In the CNS, especially with respect to infections of neurons (which are non-renewable), non-cytolytic effector mechanisms which eliminate intracellular viruses are essential to preserve neuron function. IFN-␥, as well as inflammatory cytokines (IL-12 and TNF-␣) elicit the innate antiviral responses in neurons. We have shown in neurons both in vitro and in vivo, the signaling and regulation of NOS-1 by IFN-␥ and its role in VSV encephalitis in this review.
Acknowledgements We would like to thank Jane McCutcheon (New York College of Dentistry), Adolfo Garc´ıa-Sastre, Jan Vilcek and David Levy (Mt. Sinai NYU Medical Center) for their helpful comments, feedback, and technical assistance. Funding was provided by NS39746 and DC03536 to CSR. References [1] Janeway CA, Travers P, Walport M, Capra JD. Immunobiology: the immune system in health and disease. New York: Garland; 1999. [2] Guidotti LG, Chisari FV. Noncytolytic control of viral infections by the innate and adaptive immune response. Annu Rev Immunol 2001;19:65–91.
449
[3] Reiss CS, Chesler DA, Hodges J, Ireland DDC, Chen N. Protective and pathological immune responses in the CNS. Current topics in microbiology and immunology. Berlin: Springer; 2002. [4] Samuel CE. Antiviral actions of interferons. Clin Microbiol Rev 2001;14:778–809 [Table of contents]. [5] Liu T, Khanna KM, Carriere BN, Hendricks RL. Gamma interferon can prevent herpes simplex virus type 1 reactivation from latency in sensory neurons. J Virol 2001;75:11178–84. [6] Adolf GR. Human interferon omega—a review. Mult Scler 1995;1(Suppl 1):S44–7. [7] Imakawa K, Anthony RV, Kazemi M, Marotti KR, Polites HG, Roberts RM. Interferon-like sequence of ovine trophoblast protein secreted by embryonic trophectoderm. Nature 1987;330:377–9. [8] Pontzer CH, Torres BA, Vallet JL, Bazer FW, Johnson HM. Antiviral activity of the pregnancy recognition hormone ovine trophoblast protein-1. Biochem Biophys Res Commun 1988;152:801–7. [9] Imakawa K, Roberts RM. Interferons and maternal recognition of pregnancy. Prog Clin Biol Res 1989;294:347–58. [10] Roberts RM. Conceptus interferons and maternal recognition of pregnancy. Biol Reprod 1989;40:449–52. [11] Roberts RM. A role for interferons in early pregnancy. BioEssays 1991;13:121–6. [12] Farrar MA, Schreiber RD. The molecular cell biology of interferon-gamma and its receptor. Annu Rev Immunol 1993;11:571–611. [13] Jeffrey IW, Kadereit S, Meurs EF, Metzger T, Bachmann M, Schwemmle M, et al. Nuclear localization of the interferon-inducible protein kinase PKR in human cells and transfected mouse cells. Exp Cell Res 1995;218:17–27. [14] Ljungdahl A, Olsson T, Van der Meide PH, Holmdahl R, Klareskog L, Hojeberg B. Interferon-gamma-like immunoreactivity in certain neurons of the central and peripheral nervous system. J Neurosci Res 1989;24:451–6. [15] Patterson CE, Lawrence DM, Echols LA, Rall GF. Immune-mediated protection from measles virus-induced central nervous system disease is noncytolytic and gamma interferon dependent. J Virol 2002;76:4497–506. [16] Bentivoglio M, Florenzano F, Peng ZC, Kristensson K. Neuronal IFN-gamma in tuberomammillary neurones. NeuroReport 1994;5:2413–6. [17] Olsson T, Kelic S, Edlund C, Bakhiet M, Hojeberg B, van der Meide PH, et al. Neuronal interferon-gamma immunoreactive molecule: bioactivities and purification. Eur J Immunol 1994;24:308–14. [18] Kiefer R, Haas CA, Kreutzberg GW. Gamma interferon-like immunoreactive material in rat neurons: evidence against a close relationship to gamma interferon. Neuroscience 1991;45:551–60. [19] Robertson B, Kong G, Peng Z, Bentivoglio M, Kristensson K. Interferon-gamma-responsive neuronal sites in the normal rat brain: receptor protein distribution and cell activation revealed by Fos induction. Brain Res Bull 2000;52:61–74. [20] Rubio N, de Felipe C. Demonstration of the presence of a specific interferon-gamma receptor on murine astrocyte cell surface. J Neuroimmunol 1991;35:111–7. [21] Torres C, Aranguez I, Rubio N. Expression of interferon-gamma receptors on murine oligodendrocytes and its regulation by cytokines and mitogens. Immunology 1995;86:250–5. [22] Bach EA, Aguet M, Schreiber RD. The IFN gamma receptor: a paradigm for cytokine receptor signaling. Annu Rev Immunol 1997;15:563–91. [23] Sakatsume M, Stancato LF, David M, Silvennoinen O, Saharinen P, Pierce J, et al. Interferon gamma activation of Raf-1 is Jak1-dependent and p21ras- independent. J Biol Chem 1998;273:3021–6. [24] Weihua X, Kolla V, Kalvakolanu DV. Interferon gamma-induced transcription of the murine ISGF3gamma (p48) gene is mediated by novel factors. Proc Natl Acad Sci USA 1997;94:103–8.
450
D.A. Chesler, C.S. Reiss / Cytokine & Growth Factor Reviews 13 (2002) 441–454
[25] Roy SK, Wachira SJ, Weihua X, Hu J, Kalvakolanu DV. CCAAT/enhancer-binding protein-beta regulates interferon-induced transcription through a novel element. J Biol Chem 2000;275:12626–32. [26] Gil MP, Bohn E, O’Guin AK, Ramana CV, Levine B, Stark GR, et al. Biologic consequences of Stat1-independent IFN signaling. Proc Natl Acad Sci USA 2001;98:6680–5. [27] Ramana CV, Grammatikakis N, Chernov M, Nguyen H, Goh KC, Williams BR, et al. Regulation of c-myc expression by IFN-gamma through Stat1-dependent and -independent pathways. EMBO J 2000;19:263–72. [28] Ramana CV, Gil MP, Han Y, Ransohoff RM, Schreiber RD, Stark GR. Stat1-independent regulation of gene expression in response to IFN- gamma. Proc Natl Acad Sci USA 2001;98:6674–9. [29] Karupiah G, Xie QW, Buller RM, Nathan C, Duarte C, MacMicking JD. Inhibition of viral replication by interferon-gamma-induced nitric oxide synthase. Science 1993;261:1445–8. [30] Marletta MA. Nitric oxide synthase structure and mechanism. J Biol Chem 1993;268:12231–4. [31] Marletta MA. Nitric oxide synthase: aspects concerning structure and catalysis. Cell 1994;78:927–30. [32] Komatsu T, Bi Z, Reiss CS. Interferon-gamma induced type I nitric oxide synthase activity inhibits viral replication in neurons. J Neuroimmunol 1996;68:101–8. [33] Zhang ZG, Chopp M, Gautam S, Zaloga C, Zhang RL, Schmidt HH, et al. Upregulation of neuronal nitric oxide synthase and mRNA, and selective sparing of nitric oxide synthase-containing neurons after focal cerebral ischemia in rat. Brain Res 1994;654:85–95. [34] Doyle C, Holscher C, Rowan MJ, Anwyl R. The selective neuronal NO synthase inhibitor 7-nitro-indazole blocks both long-term potentiation and depotentiation of field EPSPs in rat hippocampal CA1 in vivo. J Neurosci 1996;16:418–24. [35] Son H, Hawkins RD, Martin K, Kiebler M, Huang PL, Fishman MC, et al. Long-term potentiation is reduced in mice that are doubly mutant in endothelial and neuronal nitric oxide synthase. Cell 1996;87:1015–23. [36] Malen PL, Chapman PF. Nitric oxide facilitates long-term potentiation, but not long-term depression. J Neurosci 1997;17:2645–51. [37] Moore PK, Babbedge RC, Wallace P, Gaffen ZA, Hart SL. 7-Nitro indazole, an inhibitor of nitric oxide synthase, exhibits antinociceptive activity in the mouse without increasing blood pressure. Br J Pharmacol 1993;108:296–7. [38] Kindgen-Milles D, Arndt JO. Nitric oxide as a chemical link in the generation of pain from veins in humans. Pain 1996;64:139–42. [39] Haley JE. Gases as neurotransmitters. Essays Biochem 1998;33:79– 91. [40] Zielasek J, Tausch M, Toyka KV, Hartung HP. Production of nitrite by neonatal rat microglial cells/brain macrophages. Cell Immunol 1992;141:111–20. [41] Merrill JE, Ignarro LJ, Sherman MP, Melinek J, Lane TE. Microglial cell cytotoxicity of oligodendrocytes is mediated through nitric oxide. J Immunol 1993;151:2132–41. [42] Galea E, Reis DJ, Feinstein DL. Cloning and expression of inducible nitric oxide synthase from rat astrocytes. J Neurosci Res 1994;37:406–14. [43] Amin AR, Attur M, Vyas P, Leszczynska-Piziak J, Levartovsky D, Rediske J, et al. Expression of nitric oxide synthase in human peripheral blood mononuclear cells and neutrophils. J Inflamm 1995-1996;47:190–205. [44] Nathan CF, Hibbs Jr JB. Role of nitric oxide synthesis in macrophage antimicrobial activity. Curr Opin Immunol 1991;3:65– 70. [45] Lancaster J, Stuehr DJ. The intracellular reactions of nitric oxide in the immune system and its enzymatic synthesis. Nitric oxide: principles and actions. California: Academic Press; 1996. p. 139–76.
[46] Dinerman JL, Dawson TM, Schell MJ, Snowman A, Snyder SH. Endothelial nitric oxide synthase localized to hippocampal pyramidal cells: implications for synaptic plasticity. Proc Natl Acad Sci USA 1994;91:4214–8. [47] Barna M, Komatsu T, Reiss CS. Activation of type III nitric oxide synthase in astrocytes following a neurotropic viral infection. Virology 1996;223:331–43. [48] Furchgott RF, Vanhoutte PM. Endothelium-derived relaxing and contracting factors. FASEB J 1989;3:2007–18. [49] Furchgott RF, Cherry PD, Zawadzki JV, Jothianandan D. Endothelial cells as mediators of vasodilation of arteries. J Cardiovasc Pharmacol 1984;6(Suppl 2):S336–43. [50] Lee SC, Dickson DW, Brosnan CF, Casadevall A. Human astrocytes inhibit Cryptococcus neoformans growth by a nitric oxide-mediated mechanism. J Exp Med 1994;180:365–9. [51] Seguin MC, Klotz FW, Schneider I, Weir JP, Goodbary M, Slayter M, et al. Induction of nitric oxide synthase protects against malaria in mice exposed to irradiated Plasmodium berghei infected mosquitoes: involvement of interferon gamma and CD8+ T cells. J Exp Med 1994;180:353–8. [52] Harris N, Buller RM, Karupiah G. Gamma interferon-induced, nitric oxide-mediated inhibition of vaccinia virus replication. J Virol 1995;69:910–5. [53] Karupiah G, Harris N. Inhibition of viral replication by nitric oxide and its reversal by ferrous sulfate and tricarboxylic acid cycle metabolites. J Exp Med 1995;181:2171–9. [54] Reiss CS, Komatsu T. Does nitric oxide play a critical role in viral infections? J Virol 1998;72:4547–51. [55] Anneser JM, Cookson MR, Ince PG, Shaw PJ, Borasio GD. Glial cells of the spinal cord and subcortical white matter up-regulate neuronal nitric oxide synthase in sporadic amyotrophic lateral sclerosis. Exp Neurol 2001;171:418–21. [56] Meldrum B. Pathophysiology of cerebral ischemia and trauma in relation to possible therapeutic approaches. Prog Clin Biol Res 1990;361:275–90. [57] Liu JS, Zhao ML, Brosnan CF, Lee SC. Expression of inducible nitric oxide synthase and nitrotyrosine in multiple sclerosis lesions. Am J Pathol 2001;158:2057–66. [58] Binder GK, Griffin DE. Interferon-gamma-mediated site-specific clearance of alphavirus from CNS neurons. Science 2001;293:303– 6. [59] Finke D, Brinckmann UG, ter Meulen V, Liebert UG. Gamma interferon is a major mediator of antiviral defense in experimental measles virus-induced encephalitis. J Virol 1995;69:5469–74. [60] Mikloska Z, Cunningham AL. Alpha and gamma interferons inhibit herpes simplex virus type 1 infection and spread in epidermal cells after axonal transmission. J Virol 2001;75:11821–6. [61] Cantin E, Tanamachi B, Openshaw H. Role for gamma interferon in control of herpes simplex virus type 1 reactivation. J Virol 1999;73:3418–23. [62] Geiger KD, Gurushanthaiah D, Howes EL, Lewandowski GA, Reed JC, Bloom FE, et al. Cytokine-mediated survival from lethal herpes simplex virus infection: role of programmed neuronal death. Proc Natl Acad Sci USA 1995;92:3411–5. [63] Arnheiter H, Haller O. Mx gene control of interferon action: different kinetics of the antiviral state against influenza virus and vesicular stomatitis virus. J Virol 1983;47:626–30. [64] Horisberger MA, Staeheli P, Haller O. Interferon induces a unique protein in mouse cells bearing a gene for resistance to influenza virus. Proc Natl Acad Sci USA 1983;80:1910–4. [65] Castelli JC, Hassel BA, Wood KA, Li XL, Amemiya K, Dalakas MC, et al. A study of the interferon antiviral mechanism: apoptosis activation by the 2–5A system. J Exp Med 1997;186:967–72. [66] Clemens MJ, Elia A. The double-stranded RNA-dependent protein kinase PKR: structure and function. J Interferon Cytokine Res 1997;17:503–24.
D.A. Chesler, C.S. Reiss / Cytokine & Growth Factor Reviews 13 (2002) 441–454 [67] Julkunen I, Sareneva T, Pirhonen J, Ronni T, Melen K, Matikainen S. Molecular pathogenesis of influenza A virus infection and virus-induced regulation of cytokine gene expression. Cytokine Growth Factor Rev 2001;12:171–80. [68] Krug RM, Shaw M, Broni B, Shapiro G, Haller O. Inhibition of influenza viral mRNA synthesis in cells expressing the interferon-induced Mx gene product. J Virol 1985;56:201–6. [69] Pavlovic J, Haller O, Staeheli P. Human and mouse Mx proteins inhibit different steps of the influenza virus multiplication cycle. J Virol 1992;66:2564–9. [70] Arnheiter H, Meier E. Mx proteins: antiviral proteins by chance or by necessity? New Biol 1990;2:851–7. [71] Staeheli P, Pavlovic J. Inhibition of vesicular stomatitis virus mRNA synthesis by human MxA protein. J Virol 1991;65:4498–501. [72] Samuel CE. Interferon-induced proteins and their mechanisms of action. Hokkaido Igaku Zasshi 1994;69:1339–47. [73] Jacobs BL, Langland JO. When two strands are better than one: the mediators and modulators of the cellular responses to double-stranded RNA. Virology 1996;219:339–49. [74] Zhong Z, Wen Z, Darnell JE. Stat3 and Stat4: members of the family of signal transducers and activators of transcription. Proc Natl Acad Sci USA 1994;91:4806–10. [75] Heath VL, Showe L, Crain C, Barrat FJ, Trinchieri G, O’Garra A. Cutting edge: ectopic expression of the IL-12 receptor-beta 2 in developing and committed Th2 cells does not affect the production of IL- 4 or induce the production of IFN-gamma. J Immunol 2000;164:2861–5. [76] Nakahira M, Tomura M, Iwasaki M, Ahn HJ, Bian Y, Hamaoka T, et al. An absolute requirement for STAT4 and a role for IFN-gamma as an amplifying factor in IL-12 induction of the functional IL-18 receptor complex. J Immunol 2001;167:1306–12. [77] Sinigaglia F, D’Ambrosio D, Panina-Bordignon P, Rogge L. Regulation of the IL-12/IL-12R axis: a critical step in T-helper cell differentiation and effector function. Immunol Rev 1999;170:65–72. [78] Rebouillat D, Hovanessian AG. The human 2 ,5 -oligoadenylate synthetase family: interferon-induced proteins with unique enzymatic properties. J Interferon Cytokine Res 1999;19:295–308. [79] Li XL, Blackford JA, Hassel BA. RNaseL mediates the antiviral effect of interferon through a selective reduction in viral RNA during encephalomyocarditis virus infection. J Virol 1998;72:2752–9. [80] Maitra RK, Silverman RH. Regulation of human immunodeficiency virus replication by 2 ,5 -oligoadenylate-dependent RNaseL. J Virol 1998;72:1146–52. [81] Diaz-Guerra M, Rivas C, Esteban M. Inducible expression of the 2-5A synthetase/RNase L system results in inhibition of vaccinia virus replication. Virology 1997;227:220–8. [82] Iordanov MS, Paranjape JM, Zhou A, Wong J, Williams BR, Meurs EF, et al. Activation of p38 mitogen-activated protein kinase and c-Jun NH(2)- terminal kinase by double-stranded RNA and encephalomyocarditis virus: involvement of RNase L, protein kinase R, and alternative pathways. Mol Cell Biol 2000;20:617–27. [83] Carpick BW, Graziano V, Schneider D, Maitra RK, Lee X, Williams BR. Characterization of the solution complex between the interferon- induced, double-stranded RNA-activated protein kinase and HIV-I trans- activating region RNA. J Biol Chem 1997;272:9510–6. [84] Lee SB, Bablanian R, Esteban M. Regulated expression of the interferon-induced protein kinase p68 (PKR) by vaccinia virus recombinants inhibits the replication of vesicular stomatitis virus but not that of poliovirus. J Interferon Cytokine Res 1996;16:1073–8. [85] Balachandran S, Roberts PC, Brown LE, Truong H, Pattnaik AK, Archer DR, et al. Essential role for the dsRNA-dependent protein kinase PKR in innate immunity to viral infection. Immunity 2000;13:129–41. [86] Stojdl DF, Abraham N, Knowles S, Marius R, Brasey A, Lichty BD, et al. The murine double-stranded RNA-dependent protein kinase PKR is required for resistance to vesicular stomatitis virus. J Virol 2000;74:9580–5.
451
[87] Durbin RK, Mertz SE, Koromilas AE, Durbin JE. PKR protection against intranasal vesicular stomatitis virus infection is mouse strain-dependent. Viral Immunol 2002;15:41–51. [88] Tan SL, Katze MG. Biochemical and genetic evidence for complex formation between the influenza A virus NS1 protein and the interferon-induced PKR protein kinase. J Interferon Cytokine Res 1998;18:757–66. [89] Gale MJ, Korth MJ, Katze MG. Repression of the PKR protein kinase by the hepatitis C virus NS5A protein: a potential mechanism of interferon resistance. Clin Diagn Virol 1998;10:157–62. [90] Taylor DR, Shi ST, Romano PR, Barber GN, Lai MM. Inhibition of the interferon-inducible protein kinase PKR by HCV E2 protein. Science 1999;285:107–10. [91] Sharp TV, Moonan F, Romashko A, Joshi B, Barber GN, Jagus R. The vaccinia virus E3L gene product interacts with both the regulatory and the substrate binding regions of PKR: implications for PKR autoregulation. Virology 1998;250:302–15. [92] Beattie E, Denzler KL, Tartaglia J, Perkus ME, Paoletti E, Jacobs BL. Reversal of the interferon-sensitive phenotype of a vaccinia virus lacking E3L by expression of the reovirus S4 gene. J Virol 1995;69:499–505. [93] Colasanti M, Persichini T, Venturini G, Ascenzi P. S-Nitrosylation of viral proteins: molecular bases for antiviral effect of nitric oxide. IUBMB Life 1999;48:25–31. [94] Komatsu T, Srivastava N, Revzin M, Ireland DD, Chesler DA, Reiss CS. Mechanisms of cytokine-mediated inhibition of viral replication. Virology 1999;259:334–41. [95] Bogdan C. Nitric oxide and the regulation of gene expression. Trends Cell Biol 2001;11:66–75. [96] Lowenstein CJ, Hill SL, Lafond-Walker A, Wu J, Allen G, Landavere M. Nitric oxide inhibits viral replication in murine myocarditis. J Clin Invest 1996;97:1837–43. [97] Zaragoza C, Ocampo CJ, Saura M, McMillan A, Lowenstein CJ. Nitric oxide inhibition of Coxsackie virus replication in vitro. J Clin Invest 1997;100:1760–7. [98] Mannick JB, Asano K, Izumi K, Kieff E, Stamler JS. Nitric oxide produced by human B lymphocytes inhibits apoptosis and Epstein-Barr virus reactivation. Cell 1994;79:1137–46. [99] Persichini T, Colasanti M, Lauro GM, Ascenzi P. Cysteine nitrosylation inactivates the HIV-1 protease. Biochem Biophys Res Commun 1998;250:575–6. [100] Persichini T, Colasanti M, Fraziano M, Colizzi V, Medana C, Polticelli F. Nitric oxide inhibits the HIV-1 reverse transcriptase activity. Biochem Biophys Res Commun 1999;258:624–7. [101] Lepoivre M, Raddassi K, Oswald I, Tenu JP, Lemaire G. Antiproliferative effects of NO synthase products. Res Immunol 1991;142:580–3 [Discussion 591-2]. [102] Mannick JB. The antiviral role of nitric oxide. Res Immunol 1995;146:693–7. [103] Coll JM. The glycoprotein G of rhabdoviruses. Arch Virol 1995;140:827–51. [104] Laquerre S, Anderson DB, Argnani R, Glorioso JC. Herpes simplex virus type 1 glycoprotein B requires a cysteine residue at position 633 for folding, processing, and incorporation into mature infectious virus particles. J Virol 1998;72:4940–9. [105] Presti RM, Popkin DL, Connick M, Paetzold S, Virgin IV HW. Novel cell type-specific antiviral mechanism of interferon gamma action in macrophages. J Exp Med 2001;193:483–96. [106] Gribaudo G, Ravaglia S, Caliendo A, Cavallo R, Gariglio M, Martinotti MG, et al. Interferons inhibit onset of murine cytomegalovirus immediate-early gene transcription. Virology 1993;197:303–11. [107] Ryman KD, White LJ, Johnson RE, Klimstra WB. Effects of PKR/RNase L-dependent and alternative antiviral pathways upon alphavirus replication and pathogenesis. Viral Immunol 2002;15:53– 76.
452
D.A. Chesler, C.S. Reiss / Cytokine & Growth Factor Reviews 13 (2002) 441–454
[108] Boudinot P, Riffault S, Salhi S, Carrat C, Sedlik C, Mahmoudi N, et al. Vesicular stomatitis virus and pseudorabies virus induce a vig1/cig5 homologue in mouse dendritic cells via different pathways. J Gen Virol 2000;81:2675–82. [109] Chin KC, Cresswell P. Viperin (cig5), an IFN-inducible antiviral protein directly induced by human cytomegalovirus. Proc Natl Acad Sci USA 2001;98:15125–30. [110] Schneider-Schaulies S, ter Meulen V. Pathogenic aspects of measles virus infections. Arch Virol Suppl 1999;15:139–58. [111] Schneider-Schaulies J, ter Meulen V, Schneider-Schaulies S. Measles virus interactions with cellular receptors: consequences for viral pathogenesis. J Neurovirol 2001;7:391–9. [112] Kolson DL, Lavi E, Gonzalez-Scarano F. The effects of human immunodeficiency virus in the central nervous system. Adv Virus Res 1998;50:1–47. [113] Tardieu M. HIV-1-related central nervous system diseases. Curr Opin Neurol 1999;12:377–81. [114] Krebs FC, Ross H, McAllister J, Wigdahl B. HIV-1-associated central nervous system dysfunction. Adv Pharmacol 2000;49:315– 85. [115] Gies U, Gorcs TJ, Mulder J, Planz O, Stitz L, Bilzer T, et al. Cortical cholinergic decline parallels the progression of Borna virus encephalitis. NeuroReport 2001;12:3767–72. [116] Solbrig MV, Koob GF, Fallon JH, Reid S, Lipkin WI. Prefrontal cortex dysfunction in Borna disease virus (BDV)-infected rats. Biol Psychiatry 1996;40:629–36. [117] Morris MM, Dyson H, Baker D, Harbige LS, Fazakerley JK, Amor S. Characterization of the cellular and cytokine response in the central nervous system following Semliki forest virus infection. J Neuroimmunol 1997;74:185–97. [118] Mokhtarian F, Zhang Z, Shi Y, Gonzales E, Sobel RA. Molecular mimicry between a viral peptide and a myelin oligodendrocyte glycoprotein peptide induces autoimmune demyelinating disease in mice. J Neuroimmunol 1999;95:43–54. [119] Miller SD, Eagar TN. Functional role of epitope spreading in the chronic pathogenesis of autoimmune and virus-induced demyelinating diseases. Adv Exp Med Biol 2001;490:99–107. [120] Miller SD, Katz-Levy Y, Neville KL, Vanderlugt CL. Virus-induced autoimmunity: epitope spreading to myelin autoepitopes in Theiler’s virus infection of the central nervous system. Adv Virus Res 2001;56:199–217. [121] Cermelli C, Jacobson S. Viruses and multiple sclerosis. Viral Immunol 2000;13:255–67. [122] Rouse BT, Deshpande S. Viruses and autoimmunity: an affair but not a marriage contract. Rev Med Virol 2002;12:107–13. [123] Wagner RR. Rhabdovirus biology and infection: an overview. In: Wagner RR, editor. The rhabdovirus. New York: Plenum Press; 1987. p. 9–74. [124] Clewley JP, Bishop DH, Kang CY, Coffin J, Schnitzlein WM, Reichmann ME, et al. Oligonucleotide fingerprints of RNA species obtained from rhabdoviruses belonging to the vesicular stomatitis virus subgroup. J Virol 1977;23:152–66. [125] Moyer SA, Smallwood-Kentro S, Haddad A, Prevec L. Assembly and transcription of synthetic vesicular stomatitis virus nucleocapsids. J Virol 1991;65:2170–8. [126] Cao BN, Huneycutt BS, Gapud CP, Arceci RJ, Reiss CS. Lymphokine expression profile of resting and stimulated CD4+ CTL clones specific for the glycoprotein of vesicular stomatitis virus. Cell Immunol 1993;146:147–56. [127] Fuller S, von Bonsdorff CH, Simons K. Vesicular stomatitis virus infects and matures only through the basolateral surface of the polarized epithelial cell line, MDCK. Cell 1984;38:65–77. [128] Bundo-Morita K, Gibson S, Lenard J. Radiation inactivation analysis of fusion and hemolysis by vesicular stomatitis virus. Virology 1988;163:622–4. [129] Huang AS, Baltimore D. Defective viral particles and viral disease processes. Nature 1970;226:325–7.
[130] Browning MJ, Huneycutt BS, Huang AS, Reiss CS. Replication-defective viruses modulate immune responses. J Immunol 1991;147:2685–91. [131] Forger JM, Bronson RT, Huang AS, Reiss CS. Murine infection by vesicular stomatitis virus: initial characterization of the H-2d system. J Virol 1991;65:4950–8. [132] Bachmann MF, Kundig TM, Kalberer CP, Hengartner H, Zinkernagel RM. Formalin inactivation of vesicular stomatitis virus impairs T-cell- but not T-help-independent B-cell responses. J Virol 1993;67:3917–22. [133] Sabin AB, Olitsky PK. Influence of host factors on the neuroinvasiveness of vesicular stomatitis virus. Effect of age on the invasion of the brain by virus instilled in the nose. J Exp Med 1937;66:15. [134] Cave DR, Hagen FS, Palma EL, Huang AS. Detection of vesicular stomatitis virus RNA and its defective-interfering particles in individual mouse brains. J Virol 1984;50:86–91. [135] Lundh B, Kristensson K, Norrby E. Selective infections of olfactory and respiratory epithelium by vesicular stomatitis and Sendai viruses. Neuropathol Appl Neurobiol 1987;13:111–22. [136] Huneycutt BS, Bi Z, Aoki CJ, Reiss CS. Central neuropathogenesis of vesicular stomatitis virus infection of immunodeficient mice. J Virol 1993;67:6698–706. [137] Huneycutt BS, Plakhov IV, Shusterman Z, Bartido SM, Huang A, Reiss CS. Distribution of vesicular stomatitis virus proteins in the brains of BALB/c mice following intranasal inoculation: an immunohistochemical analysis. Brain Res 1994;635:81–95. [138] Bi Z, Barna M, Komatsu T, Reiss CS. Vesicular stomatitis virus infection of the central nervous system activates both innate and acquired immunity. J Virol 1995;69:6466–72. [139] Barna M, Komatsu T, Bi Z, Reiss CS. Sex differences in susceptibility to viral infection of the central nervous system. J Neuroimmunol 1996;67:31–9. [140] Komatsu T, Barna M, Reiss CS. Interleukin-12 promotes recovery from viral encephalitis. Viral Immunol 1997;10:35–47. [141] Plakhov IV, Arlund EE, Aoki C, Reiss CS. The earliest events in vesicular stomatitis virus infection of the murine olfactory neuroepithelium and entry of the central nervous system. Virology 1995;209:257–62. [142] Browning MJ, Huang AS, Reiss CS. Cytolytic T lymphocytes from the BALB/c-H-2dm2 mouse recognize the vesicular stomatitis virus glycoprotein and are restricted by class II MHC antigens. J Immunol 1990;145:985–94. [143] Christian AY, Barna M, Bi Z, Reiss CS. Host immune response to vesicular stomatitis virus infection of the central nervous system in C57BL/6 mice. Viral Immunol 1996;9:195–205. [144] Bi Z, Quandt P, Komatsu T, Barna M, Reiss CS. IL-12 promotes enhanced recovery from vesicular stomatitis virus infection of the central nervous system. J Immunol 1995;155:5684–9. [145] Komatsu T, Ireland DD, Reiss CS. IL-12 and viral infections. Cytokine Growth Factor Rev 1998;9:277–85. [146] Komatsu T, Reiss CS. IFN-gamma is not required in the IL-12 response to vesicular stomatitis virus infection of the olfactory bulb. J Immunol 1997;159:3444–52. [147] Komatsu T, Ireland DD, Chen N, Reiss CS. Neuronal expression of NOS-1 is required for host recovery from viral encephalitis. Virology 1999;258:389–95. [148] Huang PL, Dawson TM, Bredt DS, Snyder SH, Fishman MC. Targeted disruption of the neuronal nitric oxide synthase gene. Cell 1993;75:1273–86. [149] Komatsu T, Bi Z, Reiss CS. Interferon-gamma induced type I nitric oxide synthase activity inhibits viral replication in neurons. J Neuroimmunol 1996;68:101–8. [150] Levy DE. The house that Jak/Stat built. Cytokine Growth Factor Rev 1997;8:81–90. [151] Taniguchi T, Ogasawara K, Takaoka A, Tanaka N. IRF family of transcription factors as regulators of host defense. Annu Rev Immunol 2001;19:623–55.
D.A. Chesler, C.S. Reiss / Cytokine & Growth Factor Reviews 13 (2002) 441–454 [152] Nishiya T, Uehara T, Edamatsu H, Kaziro Y, Itoh H, Nomura Y. Activation of Stat1 and subsequent transcription of inducible nitric oxide synthase gene in C6 glioma cells is independent of interferongamma-induced MAPK activation that is mediated by p21ras. FEBS Lett 1997;408:33–8. [153] Hu J, Roy SK, Shapiro PS, Rodig SR, Reddy SP, Platanias LC, et al. ERK1 and ERK2 activate CCAAAT/enhancer-binding protein-beta-dependent gene transcription in response to interferon-gamma. J Biol Chem 2001;276:287–97. [154] Quandt K, Frech K, Karas H, Wingender E, Werner T. MatInd and MatInspector: new fast and versatile tools for detection of consensus matches in nucleotide sequence data. Nucl Acids Res 1995;23:4878–84. [155] Sasaki M, Gonzalez-Zulueta M, Huang H, Herring WJ, Ahn S, Ginty DD, et al. Dynamic regulation of neuronal NO synthase transcription by calcium influx through a CREB family transcription factor-dependent mechanism. Proc Natl Acad Sci USA 2000;97:8617–22. [156] Johnson RT, Griffin DE, Hirsch RL, Wolinsky JS, Roedenbeck S, Lindo de Soriano I, et al. Measles encephalomyelitis—clinical and immunologic studies. N Engl J Med 1984;310:137–41. [157] Jabbour JT, Duenas DA, Sever JL, Krebs HM, Horta-Barbosa L. Epidemiology of subacute sclerosing panencephalitis (SSPE). A report of the SSPE registry. JAMA 1972;220:959–62. [158] Detels R, Brody JA, McNew J, Edgar AH. Further epidemiological studies of subacute sclerosing panencephalitis. Lancet 1973;2: 11–4. [159] van Binnendijk RS, Poelen MC, de Vries P, Voorma HO, Osterhaus AD, Uytdehaag FG. Measles virus-specific human T cell clones. Characterization of specificity and function of CD4+ helper/cytotoxic and CD8+ cytotoxic T cell clones. J Immunol 1989;142:2847–54. [160] van Binnendijk RS, Poelen MC, Kuijpers KC, Osterhaus AD, Uytdehaag FG. The predominance of CD8+ T cells after infection with measles virus suggests a role for CD8+ class I MHC-restricted cytotoxic T lymphocytes (CTL) in recovery from measles. Clonal analyses of human CD8+ class I MHC-restricted CTL. J Immunol 1990;144:2394–9. [161] Nahmias A, Griffith JD, Salisbury C, Yoshida K. Thymic aplasia with lymphopenia, plasma cells, and normal immunoglobulins. Relation to measles virus infection. JAMA 1967;201:729–35. [162] Niewiesk S, Brinckmann U, Bankamp B, Sirak S, Liebert UG, ter Meulen V. Susceptibility to measles virus-induced encephalitis in mice correlates with impaired antigen presentation to cytotoxic T lymphocytes. J Virol 1993;67:75–81. [163] Finke D, Liebert UG. CD4+ T cells are essential in overcoming experimental murine measles encephalitis. Immunology 1994;83:184–9. [164] Schnorr JJ, Schneider-Schaulies S, Simon-Jodicke A, Pavlovic J, Horisberger MA, ter Meulen V. MxA-dependent inhibition of measles virus glycoprotein synthesis in a stably transfected human monocytic cell line. J Virol 1993;67:4760–8. [165] Schneider-Schaulies S, Schneider-Schaulies J, Schuster A, Bayer M, Pavlovic J, ter Meulen V. Cell type-specific MxA-mediated inhibition of measles virus transcription in human brain cells. J Virol 1994;68:6910–7. [166] Pavlovic J, Arzet HA, Hefti HP, Frese M, Rost D, Ernst B, et al. Enhanced virus resistance of transgenic mice expressing the human MxA protein. J Virol 1995;69:4506–10. [167] Nahmias AJ, Josey WE, Naib ZM, Luce CF, Duffey A. Antibodies to herpes virus hominis type 1. Am J Epidemiol 1970;91:539– 46. [168] Whitley RJ, Soong SJ, Hirsch MS, Karchmer AW, Dolin R, Galasso G, et al. Herpes simplex encephalitis: vidarabine therapy and diagnostic problems. N Engl J Med 1981;304:313–8. [169] Whitley RJ, Nahmias AJ. Therapeutic challenges of neonatal herpes simplex virus infection. Scand J Infect Dis Suppl 1985;47:97– 106.
453
[170] Geiger K, Howes EL, Sarvetnick N. Ectopic expression of gamma interferon in the eye protects transgenic mice from intraocular herpes simplex virus type 1 infections. JVirol 1994;68:5556–67. [171] Geiger K, Howes E, Gallina M, Huang XJ, Travis GH, Sarvetnick N. Transgenic mice expressing IFN-gamma in the retina develop inflammation of the eye and photoreceptor loss. Invest Ophthalmol Vis Sci 1994;35:2667–81. [172] Geiger KD, Nash TC, Sawyer S, Krahl T, Patstone G, Reed JC, et al. Interferon-gamma protects against herpes simplex virus type 1-mediated neuronal death. Virology 1997;238:189–97. [173] Cantin EM, Hinton DR, Chen J, Openshaw H. Gamma interferon expression during acute and latent nervous system infection by herpes simplex virus type 1. J Virol 1995;69:4898–905. [174] Ellison AR, Yang L, Voytek C, Margolis TP. Establishment of latent herpes simplex virus type 1 infection in resistant, sensitive, and immunodeficient mouse strains. Virology 2000;268:17–28. [175] Dorman SE, Uzel G, Roesler J, Bradley JS, Bastian J, Billman G, et al. Viral infections in interferon-gamma receptor deficiency. J Pediatr 1999;135:640–3. [176] Komatsu T, Reiss CS. IFN-gamma is not required in the IL-12 response to vesicular stomatitis virus infection of the olfactory bulb. J Immunol 1997;159:3444–52. [177] Lohman BL, Welsh RM. Apoptotic regulation of T cells and absence of immune deficiency in virus-infected gamma interferon receptor knockout mice. J Virol 1998;72:7815–21. [178] Nansen A, Christensen JP, Ropke C, Marker O, Scheynius A, Thomsen AR. Role of interferon-gamma in the pathogenesis of LCMV-induced meningitis: unimpaired leucocyte recruitment, but deficient macrophage activation in interferon-gamma knock-out mice. J Neuroimmunol 1998;86:202–12. [179] Muller U, Steinhoff U, Reis LF, Hemmi S, Pavlovic J, Zinkernagel RM, et al. Functional role of type I and type II interferons in antiviral defense. Science 1994;264:1918–19121. [180] Rodriguez M, Pavelko K, Coffman RL. Gamma interferon is critical for resistance to Theiler’s virus-induced demyelination. J Virol 1995;69:7286–90. [181] Fiette L, Aubert C, Muller U, Huang S, Aguet M, Brahic M, et al. Theiler’s virus infection of 129Sv mice that lack the interferon alpha/beta or interferon gamma receptors. J Exp Med 1995;181:2069–76. [182] Krug RM, Shaw M, Broni B, Shapiro G, Haller O. Inhibition of influenza viral mRNA synthesis in cells expressing the interferon-induced Mx gene product. J Virol 1985;56:201–6. [183] Pavlovic J, Haller O, Staeheli P. Human and mouse Mx proteins inhibit different steps of the influenza virus multiplication cycle. J Virol 1992;66:2564–9. [184] Staeheli P, Pavlovic J. Inhibition of vesicular stomatitis virus mRNA synthesis by human MxA protein. J Virol 1991;65:4498–501. [185] Theerasurakarn S, Ubol S. Apoptosis induction in brain during the fixed strain of rabies virus infection correlates with onset and severity of illness. J Neurovirol 1998;4:407–14. [186] Maitra RK, Silverman RH. Regulation of human immunodeficiency virus replication by 2 ,5 -oligoadenylate-dependent RNase L. J Virol 1998;72:146–52. [187] Diaz-Guerra M, Rivas C, Esteban M. Inducible expression of the 2-5A synthetase/RNase L system results in inhibition of vaccinia virus replication. Virology 1997;227:220–8. [188] Iordanov MS, Paranjape JM, Zhou A, Wong J, Williams BR, Meurs EF. Activation of p38 mitogen-activated protein kinase and c-Jun NH(2)- terminal kinase by double-stranded RNA and encephalomyocarditis virus: involvement of RNase L, protein kinase R, and alternative pathways. Mol Cell Biol 2000;20:617–27. [189] Carpick BW, Graziano V, Schneider D, Maitra RK, Lee X, Williams BR. Characterization of the solution complex between the interferon- induced, double-stranded RNA-activated protein kinase and HIV-I trans- activating region RNA. J Biol Chem 1997;272:9510–6.
454
D.A. Chesler, C.S. Reiss / Cytokine & Growth Factor Reviews 13 (2002) 441–454
[190] Lee SB, Bablanian R, Esteban M. Regulated expression of the interferon-induced protein kinase p68 (PKR) by vaccinia virus recombinants inhibits the replication of vesicular stomatitis virus but not that of poliovirus. J Interferon Cytokine Res 1996;16:1073–8. [191] Khabar KS, Dhalla M, Siddiqui Y, Zhou A, Al-Ahdal MN, Der SD, et al. Effect of deficiency of the double-stranded RNA-dependent protein kinase, PKR, on antiviral resistance in the presence or absence of ribonuclease L: HSV-1 replication is particularly sensitive to deficiency of the major IFN-mediated enzymes. J Interferon Cytokine Res 2000;20:653–9.
[192] Persichini T, Colasanti M, Lauro GM, Ascenzi P. Cysteine nitrosylation inactivates the HIV-1 protease. Biochem Biophys Res Commun 1998;250:575–6. [193] Mannick JB, Asano K, Izumi K, Kieff E, Stamler JS. Nitric oxide produced by human B lymphocytes inhibits apoptosis and Epstein-Barr virus reactivation. Cell 1994;79:1137–46. [194] Lowenstein CJ, Hill SL, Lafond-Walker A, Wu J, Allen G, Landavere M. Nitric oxide inhibits viral replication in murine myocarditis. J Clin Invest 1996;97:1837–43.