Abstract
Apoptosis is associated with virus-induced human diseases of the central nervous system, heart and liver, and causes substantial morbidity and mortality. Although virus-induced apoptosis is well characterized in individual cells in cell culture, virus-induced apoptosis in vivo and the role of apoptosis in virus-induced disease is not well established. This Review focuses on animal models of virus-induced diseases of the central nervous system, heart and liver that provide insights into the role of apoptosis in pathogenesis, the pathways involved and the potential therapeutic implications.
Apoptosis plays an essential part in homeostasis, development and human disease by facilitating the removal of unwanted, damaged or infected cells. However, apoptosis may also have a pathogenic role by contributing to cell death and tissue injury. Apoptotic cell death occurs in a wide range of human viral infections, including infections of the central nervous system (CNS), heart and liver. For example, apoptosis can be detected in the brains of patients with virus-induced CNS disease, including patients with HIV-1-associated dementia1, herpes simplex virus (HSV)2,3 and cytomegalovirus (CMV)3 encephalitis. Furthermore, apoptotic cells can be observed in the diseased human heart during both active and chronic viral myocarditis4, and hepatocyte apoptosis is emerging as an important feature of liver injury in patients with hepatitis B virus (HBV) and hepatitis C virus (HCV) infections5–10.
The morphological and nuclear changes associated with apoptosis are typically caused by the sequential activation of a family of aspartate-specific cysteine proteases called caspases11–13 (BOX 1). Caspase 3, the main executioner caspase, can be activated by the initiator caspase, caspase 8, following activation of cell-surface death receptors in the extrinsic apoptotic pathway (BOX 1; FIG. 1). Alternatively, the intrinsic apoptotic pathway can activate caspase 3 following release of pro-apoptotic factors from the mitochondria and activation of the initiator caspase, caspase 9 (BOX 1; FIG. 1). In some cell types (type I cells), activation of caspase 8 is sufficient for apoptosis. However, in other cell types (type II cells), apoptosis requires intrinsic apoptotic signalling, which can be activated directly or through extrinsic apoptotic signalling through the caspase 8-dependent cleavage of BCL-2 interacting domain death agonist (BID), a member of the BH3-only B-cell lymphoma protein 2 family (BOX 2; FIG. 1). Caspase 3 can also be activated by granzyme B, a serine protease found in cytotoxic T lymphocytes (CTLs) (FIG. 1). Granzyme B can also cleave BID, forming the truncated protein gtBID and resulting in the activation of intrinsic apoptotic signalling14. Granzyme B and extrinsic apoptotic signalling constitute the two major components of CTL-induced cell death.
Box 1.
Caspase activation
Caspases are a class of cysteine-aspartyl proteases that are synthesized as inactive precursor enzymes or proenzymes (see the figure). These proteases typically lie dormant in the healthy cell and, in response to cell-death stimuli, are converted, either by proteolytic cleavage or by recruitment into large complexes, into active enzymes. Once activated, caspases cleave their substrates after conserved aspartate residues. The effects of caspases on cellular substrates bring about the biochemical and morphological features of apoptosis (FIG. 1).
Caspase proenzymes contain three domains: an amino-terminal prodomain; a large subunit that contains the active-site cysteine within a conserved QACXG motif (p20); and a carboxy-terminal small subunit (p10). Two cleavage events at aspartate (Asp) residues are required to activate caspases. The first divides the proenzyme into large and small caspase subunits, and the second removes the N-terminal prodomain. The resulting functional caspase is a tetramer of two large (p20) and two small (p10) subunits.
Caspases are divided into two groups. Initiator caspases (such as caspase 8 and caspase 9) activate the effector caspases and have long prodomains that allow them to interact with death effector domains (DEDs) or caspase recruitment domains (CARDs) in adaptor proteins such as FAS-associated death domain protein (FADD) and apoptotic protease-activating factor 1 (APAF1) (reviewed in REF. 106). Effector caspases (such as caspase 3) function primarily to cause the morphological features of apoptosis. Specifically, activation of caspase 3 leads to the breakdown of several cytoskeletal proteins, cleavage of poly (ADP-ribose) polymerase (PARP) and degradation of ICAD (inhibitor of caspase-activated DNase), thereby releasing CAD, which cleaves cellular DNA. Activation of caspase 3 and PARP cleavage, by western blot and immunohistochemistry, and DNA fragmentation, by gel electrophoresis and TUNEL, are frequently used as apoptosis assays in vivo.
Figure 1. Caspase-dependent apoptosis.
Activation of caspase 3 and caspase-dependent apoptosis are caused by the initiator caspases, caspase 8 and caspase 9, as part of the extrinsic and intrinsic apoptotic signalling pathways, or by granzyme B. In the extrinsic pathway, death ligands, including Fas Ligand (FasL), bind and activate their cognate receptors (Fas). Activated receptors, an adaptor molecule, such as Fas-associated death domain protein (FADD), and pro-caspase 8 then form a death-inducing signalling complex (DISC) in which caspase 8 is activated. Caspase 8 can activate caspase 3 and can cleave the BH3-only protein BCL-2 interacting domain death agonist (BID), forming truncated BID (tBID), which can activate the intrinsic apoptotic pathway. In the intrinsic apoptotic pathway, pro-apoptotic B-cell lymphoma 2 (BCL-2) family members BCL-2 associated × protein (BAX) and BCL-2 antagonist killer 1 (BAK1) form pores in the mitochondrial membrane. Pro-apoptotic mitochondrial factors, including DIABLO (direct inhibitor of apoptosis protein-binding protein with low pI; also known as SMAC) and cytochrome c, are released and contribute to apoptosis by activating caspase 9 or inhibiting cellular inhibitor of apoptosis proteins (IAPs). Granzyme B can directly cleave and activate pro-caspase 3. Granzyme B can also cleave BID, resulting in granzyme tBID, which can activate the intrinsic apoptotic pathway. cFLIP (cellular FLIP/caspase 8 inhibitor protein) can block the activation of caspase 8. Bcl-xL, BCL-extra large.
Box 2. BCL-2 family proteins.
Mitochondrial apoptotic signalling and mitochondrial outer-membrane permeabilization (MOMP) controlled by the B-cell lymphoma 2 (BCL-2) family of proteins, which is divided into three groups based on the presence of BCL-2 homology (BH) domains. Anti-apoptotic BCL-2 family proteins, such as BCL-2, BCL-w (BCL-2 related gene, long isoform), BCL-xL, BCL-extra large., A1 and myeloid cell leukaemia 1 (MCL1), contain BH domains 1–4 and are generally integrated into the outer mitochondrial membrane107. These proteins function to directly bind and inhibit the pro-apoptotic BCL-2 proteins. The pro-apoptotic BCL-2 proteins are functionally divided into two classes. The effector molecules, such as BCL-2 associated × protein (BAX) and BCL-2 antagonist killer 1 (BAK1), contain BH domains 1–3 and permeabilize the outer mitochondrial membrane by creating a proteolipid pore. The BH3-only proteins, such as BCL-2 antagonist of cell death (BAD), BCL-2 interacting domain death agonist (BID), BCL-2 interacting killer (BIK), BCL-2-like protein 11 (BCL2L11; also known as BIM), BCL-2 modifying factor (BMF), BCL-2/adenovirus E1B 19 KD protein-interacting protein 3 (bNIP3), Harakiri (HRK), Noxa NOXA1 and BCL-2-binding component 3 (BBC3; also known as PUMA), function in distinct cellular stress pathways and signal that a stress has occurred through interactions with other BCL-2 family members.
The combined signalling in the BCL-2 family dictates the immediate fate of the cell, and two models have been described. In the rheostat model (see the figure, part a), a basal state is described in which the number of anti- and pro-apoptotic BCL-2 family proteins is equal and tipping this balance dictates cell fate. In the anti-apoptotic protein neutralization model (see the figure, part b), the BH3-only proteins BID, BIM and PUMA engage MOMP because they bind and engage all anti-apoptotic BCL-2 family members. A combination of other BH3-only proteins is required to promote apoptosis because each neutralizes only a subset of anti-apoptotic BCL-2 family proteins (reviewed in REF. 108). JNK, Jun N-terminal kinase.
Our understanding of the potential role of apoptosis in the pathogenesis of human diseases has been facilitated by the availability of animal models. This Review focuses on representative animal models that have been used to determine the role of apoptosis in virus-induced pathogenesis in the CNS, heart and liver (TABLE 1), to define the specific pathways involved and to assess the potential of anti-apoptotic treatment strategies.
Table 1.
Apoptosis in representative animal models of human disease
Virus family | Virus | Human disease | Animal model | Apoptosis | |
---|---|---|---|---|---|
Acute disease | chronic disease | ||||
Central nervous system (CNS) | |||||
Picornaviridae | TMEV | No | Infects neurons, leading to acute CNS disease in mice, which can progress to chronic inflammatory demyelinating disease with a low level of viral persistence in macrophages, astrocytes and oligodendrocytes; serves as a model for multiple sclerosis125–127 | Oligodendrocytes with no evidence of colocalization with virus antigen32 | |
Togaviridae | SINV | SINV is the prototype alphavirus; alphaviruses cause fever, rash, arthritis, meningitis and encephalomyelitis in humans. | Infects neurons, leading to acute CNS disease in mice128; in surviving mice, persistent SINV RNA, but not infectious virus, is associated with neurological damage in the hippocampus64 | Infected neurons33 | Immune-mediated apoptosis of uninfected hippocampal cells64 |
Flaviviridae | WNV | Infection can be associated with a febrile illness that can progress to severe neuroinvasive disease, including encephalitis, meningitis or myelitis | Infects neurons, leading to encephalitis and myelitis15,26,129,130 | Apoptosis in neurons in same areas as viral antigen15,26 | Not applicable |
Reoviridae | Reovirus (type 3) | No | Infects neurons, leading to acute meningoencephalitis or, in newborn mice, acute flaccid paralysis16,35 | Apoptosis of infected neurons16,34,35,131 | Not applicable |
Heart | |||||
Picornaviridae | CVB3 | Myocarditis; early acute disease followed by chronic autoimmune-mediated disease | Acute myocarditis associated with a low level of infected myocytes (1–3%); can be followed by chronic disease in some strains of mice65 | Myocytes and infiltrating mononuclear cells19,45,132 | Massive necrosis and fibrosis owing to cardiac-specific autoimmune responses in the absence of infectious virus65 |
Reoviridae | Reovirus strain 8B | No | Infects myocytes, leading to acute myocarditis in newborn mice133 | Apoptosis of infected myocytes21,134 | Not applicable |
Liver | |||||
Hepadnaviridae | HBV | Chronic hepatitis and hepatocellular carcinoma | Transgenic mice that conditionally express viral proteins28,36 | Not applicable | Apoptosis of hepatocytes that express viral antigen28,36 |
Flaviviridae | HCV | None | None | None | None |
CVB3, coxsackievirus B3; HBV, hepatitis B virus; HCV, hepatitis C virus; SINV, Sindbis virus; TMEV, Theiler’s murine encephalomyelitis virus; WNV, West Nile virus.
Viruses induce apoptosis in vivo
In many experimental animal models of apoptosis there is an association between the severity of apoptosis and the amount of virus or virus antigen, suggesting that apoptosis is a consequence of viral replication.
Apoptosis and viral replication
Peak virus titres in the CNS are associated with the appearance of activated caspase 3 following infection of mice with West Nile virus (WNV)15. Viral titre also correlates with caspase 3-related apoptotic events following virus infection of the CNS (BOX 1). For example, increased viral titre correlates with increased oligonucleosomal DNA laddering following infection with reovirus16 and with TUNEL staining following infection with sindbis virus (SINV)17. Increased apoptosis also parallels viral replication in the hearts of mice infected with coxsackievirus B3 (CVB3)18–20, reovirus21,22, encephalomyocarditis virus23 and murine CMV24, and in the livers of mice infected with herpes simplex virus 1 (HSV-1)25.
Apoptosis and virus antigen
Animal models of virus-induced disease have also shown that there is a temporal and spatial correlation between the appearance of virus antigen and the onset of apoptosis. examples of temporal correlations include time courses of reovirus and WNV infection, which show that apoptosis occurs concurrently with, or slightly after, the appearance of viral antigen16,26. In addition, in the livers of transgenic mice that express HBV and HCV proteins, the expression or recognition of viral antigen triggers apoptosis27,28.
Spatial correlations are revealed by the appearance of apoptotic cell death in the same regions as viral antigen following infection of the CNS with reovirus16,29 and WNV26, and following infection of the heart with reovirus21,30,31 and CVB3. Colocalization of apoptosis and virus antigen in individual cells can also be detected in the CNS following infection with Theiler’s murine encephalomyelitis virus (TMeV)32, SINV33 and reovirus34,35, in the hearts of CVB3-infected mice19 and in the livers of transgenic mice that express HCV and HBV proteins28,36.
Apoptosis in non-infected cells
Non-infected cells also undergo apoptosis in some animal models of virus-induced disease. In these cases, apoptosis typically occurs in proximity to infected cells (bystander apoptosis), as exemplified in the mouse CNS during acute TMeV and reovirus encephalitis and during WNV-induced acute flaccid polio-like paralysis16,32,37. By contrast, following infection of the mouse CNS with Venezuelan equine encephalitis virus (Vee) or the rat CNS with Borna disease virus (BDV), apoptosis occurs in areas that lack viral antigen but do show evidence of astrogliosis and inflammation (in the case of Vee)38 or microglial proliferation (in the case of BDV)39. It has been proposed that virus-induced upregulation of proinflammatory genes, including those that encode inducible nitric oxide synthase (iNOS) and tumour necrosis factor (TNF), may be responsible for apoptosis in areas that do not contain viral antigen38. Similarly, although Murray Valley encephalitis virus infects neurons, most apoptotic cells are infiltrating inflammatory cells, including macrophages, lymphocytes and neutrophils, and the death of these cells may contribute to the eventual death of the host through the release of inflammatory mediators, such as iNOS40,41.
Apoptosis and pathogenicity
In addition to showing that the severity of apoptosis is associated with increased viral titre and that apoptosis commonly occurs in anatomical areas targeted by viral infection, animal models have also revealed a positive correlation between apoptosis and disease severity, suggesting that apoptosis is a pathogenic mechanism in virus-induced disease.
For example, in a mouse model of SINV CNS infection, the rates of both apoptosis and mortality are high in young mice, but not in old mice17,42. The neurovirulence of viral strains also affects the level of apoptosis and mortality. For example, few apoptotic cells are observed in the CNSs of 2-week-old mice following infection with SINV strain 663, and these animals survive infection. By contrast, infection of 2-week-old mice with a more neurovirulent recombinant SINV strain (Te) induces apoptosis in the brains and spinal cords and results in death of the animals33. Similarly, infection of mice with the highly virulent TMeV GDVII strain produces an acute, fatal polioencephalomyelitis that is associated with high levels of neuronal apoptosis, whereas mice infected with the attenuated TMeV DA strain have lower levels of neuronal apoptosis and survive acute infection32. It is thought that differences in the capsid protein VP2 contribute to the differences in apoptosis that are induced by the GDVII and DA TMeV strains43.
Increased apoptosis is also associated with increased severity of CNS disease following infection with WNV, as the brain and spinal cord from non-paralyzed mice showed virtually no apoptosis, whereas significant numbers of apoptotic cells were detected in the brain and spinal cord of paralyzed mice44. Increased apoptosis also correlates with increased severity of disease in CVB3-induced and murine CMV-induced myocarditis in infiltrating mononuclear cells and myocytes18,24,45.
Although apoptosis plays a major part in disease severity in several virus-induced diseases, there are exceptions. For example, an inverse relationship between pathogenicity and apoptosis is usually observed following rabies virus infection of the CNS, indicating that apoptosis is not an essential component of rabies virus-induced CNS disease46–52.
The immune system in virus-induced apoptosis
The evidence indicates that cellular immune responses are not always necessary for the induction of apoptosis in infected cells in the CNS, heart and liver. For example, in vitro studies show that many viruses induce apoptosis following infection of cultured neurons, myocytes and hepatocytes in the absence of immune cells. In addition, in vivo evidence supporting direct viral induction of apoptosis has come from models in which apoptotic injury occurs in the absence of, or prior to, an obvious immune response. For example, apoptosis occurs following infection of the CNS and heart with reovirus16,21,30, and shortly after the induction of protein expression in transgenic mice that conditionally express HCV structural proteins in the liver28 in the presence of little, or no, infiltrating immune cells. This shows that immune cells are not required for apoptosis in these models.
The ability of individual viral proteins to interact with cellular apoptotic signalling molecules also suggests that viruses can directly induce apoptosis without the involvement of immune cells. For example, virus proteins have been directly linked to apoptosis or pro-apoptotic proteins during virus-induced myocarditis. Specifically, the pro-apoptotic protein Siva, which binds to the CVB3 capsid protein VP2 (a determinant of CVB3-induced myocarditis), is strongly upregulated in the same region where apoptosis occurs during acute CVB3-induced myocarditis53. In addition, Reoviruses that are deficient in the expression of the non-structural reovirus protein σ1 small (σ1s) induce less CNS and cardiac apoptosis than parental virus strains31, and a single amino-acid change at position 55 in the SINV e2 glycoprotein results in different levels of CNS apoptosis33. Although it has not yet been shown that σ1s or e2 binds the apoptotic machinery, mutations in these proteins do not seem to alter the host immune response following infection, suggesting that they have a direct effect on cellular apoptotic signalling.
Viral clearance
For most viral infections, the immune system of the host probably induces apoptosis to promote virus clearance. During acute viral infections, the number of virus-specific T cells, including CTLs, increases, resulting in apoptosis of virus-infected cells and viral clearance. For example, the appearance of CD3+ CTLs coincides with a drop in TMeV titre during acute disease, which suggests that these cells are involved in the apoptotic death of TMeV-infected cells and the partial clearance of TMeV from the CNS54. CTLs secrete cytotoxic cytokines, including TNF, and express high levels of surface Fas ligand (FasL) and TRAIL, which can induce apoptosis by interacting with death receptors on target cells and activating the extrinsic apoptotic pathway (FIG. 1). Alternatively, cytotoxic immune cells can induce apoptosis by the action of granzyme B (FIG. 1), which can directly activate caspase 8 and caspase 3, thereby activating apoptosis. Immune-cell-mediated Fas-induced apoptosis thus plays a part in CD8+ CTL-induced death of WNV-infected cells, leading to viral clearance in mice. In these mice, Fas is strongly upregulated in neurons in the same areas where virus antigen is present. In addition, in the generalized lymphoproliferative disease (gld) mouse model, in which mice are defective in the expression of functional FasL, there was an increase in susceptibility to WNV with increased virus burden and delayed clearance. Finally, WNV-primed CD8+ T cells from wild-type, but not of gld, mice limited WNV infection in the CNS and enhanced survival in recipient mice55. Fas-induced apoptosis is not only involved in the action of perforin and granzymes, but also promotes virus clearance by infiltrating T cells that express the γδ T-cell receptor56,57.
Interestingly, transgenic mice that expressed HCV core, e1, e2 and NS2 proteins were resistant to Fas antibody-stimulated apoptosis and lethality, as they inhibited Fas antibody-induced release of cytochrome c and activated caspase 9, and caspase 3 or caspase 7, but not caspase 8 (REF. 58). It has been proposed that the suppression of Fas-mediated death of hepatocytes that express HCV proteins protects these cells from immune-cell-mediated apoptosis and may contribute to the development of persistent infection.
Apoptosis and chronic disease
Activated T cells generated during viral infections produce potentially toxic molecules (discussed above) and must be silenced following virus clearance. This feat is accomplished by a process known as activation-induced cell death, which results in the survival of only a small number of virus-specific T cells that become memory cells. Activation-induced cell death following virus infection is mediated, at least in part, by Fas59, BCL-2-like protein 11 (BCL2L11; also known as BIM)59 and BCL-2-binding component 3 (BBC3; also known as PuMA)60. Furthermore, the expression of both Fas and FasL are induced following T-cell activation61. Defects in activation-induced cell death have been proposed to contribute to the autoimmune-mediated, apoptotic spinal cord injury that occurs during chronic TMeV infection. As a result, inflammation in the brain and grey matter of the spinal cord subsides following acute TMeV CNS infection, and TuNeL- (BOX 1) and virus-positive cells are seldom seen. Instead, TuNeL-positive oligodendrocytes, microglia and macrophages can be observed in the demyelinated white matter. These TuNeL-positive cells localize near antigen-positive cells (macrophages, astrocytes and oligodendrocytes) but do not appear to colocalize in specific cells32,54,62,63.
Autoimmune-mediated apoptosis also has a role in the chronic CNS disease that can follow acute CNS infection with SINV and TMeV. For example, although SINV is cleared from the CNS following acute, non-fatal encephalomyelitis, virus RNA persists. The brains of recovered mice show marked destruction of regions of the hippocampus and extensive loss of brain tissue in the surrounding area that is associated with mononuclear cell infiltration. In these animals, the number of CD4+ T cells and macrophages or microglia that infiltrate the hippocampal gyrus correlates with the number of apoptotic pyramidal neurons. This indicates that in chronic SINV-induced CNS disease, CD4+ cells promote progressive apoptotic neuronal damage in the brain despite clearance of the virus64.
Apoptosis is also an important pathogenic mechanism in the chronic immune-mediated phase of virus-induced myocarditis apoptosis following infection with CVB3. The persistence of apoptosis and inflammatory infiltrates in the severe disease form of CVB3-induced myocarditis is associated with CVB3-stimulated autoimmune T-cell responses to cardiac antigens, resulting in large areas of necrosis and fibrosis19. Induction of autoimmunity depends on CD4+ T helper 1 (TH1) cells, whereas TH2 responses promote disease resistance. It has also been shown that T lymphocytes which express the γδ T-cell receptor selectively induce Fas-mediated apoptosis in TH2 cells and are therefore crucial for the maintenance of a dominant TH1 response45,65.
The immune response is also thought to be involved in the development of chronic hepatitis and hepatocellular carcinoma. In transgenic mice that contain the entire HBV-envelope coding region, HBV-antigen recognition by CD8+ cells27 induces hepatocellular apoptosis by activating FasL-dependent death pathways that are inhibited by the administration of soluble Fas66 and by the neutralization of FasL activity27.
Immune evasion
Unsurprisingly, given the involvement of the immune system in the onset of apoptosis in infected cells, many viruses have developed strategies to avoid host immunity to prevent the apoptotic death of infected cells and thereby increase viral growth. For example, in addition to infecting hepatocytes, HCV can infect and replicate in immune cells, which adversely affects immune-cell function. In transgenic mice with directed expression of the HCV core to T cells, T-cell responses are inhibited, possibly owing to the increased sensitivity of these T cells to Fas-mediated apoptosis. This suggests that, in contrast to the inhibition of Fas-mediated apoptosis in hepatocytes (discussed above), HCV protein expression in mononuclear cells contributes to liver injury by increasing Fas-mediated mononuclear cell apoptosis and may also contribute to persistent infection67. Increased Fas-mediated apoptosis of concanavalin-activated T cells is also observed in transgenic mice with hepatic expression of HCV core, e1 and e2 proteins following adoptive transfer68. Furthermore, FasL expression is increased in these livers, supporting the role of Fas signalling in HCV-mediated T-cell apoptosis. Similarly, evasion of the virus-specific T-cell response by increased lymphocyte apoptosis is also observed following exposure of woodchucks to woodchuck hepatitis virus (WHV) in the woodchuck model of HBV infection. In this case, lymphocytes derived from the early acute period of infection, when non-specific T-cell responses profoundly decline but the WHV-specific response has not yet increased, are significantly more prone to activation-induced death and delay the virus-specific T-cell response69.
The apoptotic death of immune effector cells can also result from the expression of FasL by virus-infected cells. FasL may ‘arm’ the infected cell and allow it to kill infiltrating, uninfected bystander cells, including immune effector cells (which themselves express death receptors). This has been shown following rabies virus infection of the mouse CNS70.
The immune response can also be evaded by inhibiting extrinsic apoptotic signalling in infected cells, as occurs following infection of mice with murine CMV. Four murine CMV genes are involved in regulating murine CMV-induced apoptosis. These include the gene that encodes M36, which interacts and prevents the activation of pro-caspase 8, the initiator caspase involved in extrinsic apoptotic signalling. In vivo, the growth of murine CMV recombinants that lack M36 results in more apoptosis in the liver and reduced replication, a phenotype that can be rescued by expression of dominant-negative Fas-associated death domain protein (FADD), which also blocks caspase 8-dependent apoptosis71.
Interferon signalling
Interferons (IFNs) are secreted cytokines that are part of the innate immune response to virus infection. They elicit distinct antiviral effects and can control most virus infections in the absence of adaptive immunity (BOX 3). IFNs also establish a pro-apoptotic state in cells, which helps to remove infected cells72. Although the molecular basis for the role of IFNs in apoptosis remains unclear, several mechanisms are probably involved (BOX 3). For example, IFN induces the expression of several genes, including TRAIL, and NK cells can use TRAIL to kill virus-infected cells in an IFN-dependent manner73. The IFN-inducible proteins PKR (protein kinase R; also known as eIF2AK2) and 2′-5′-oli-goadenylate synthetase may also affect virus-induced apoptosis74.
Box 3. Interferon signalling.
Type I interferons (IFNs), including IFNα and IFNβ, are induced directly in response to viral infection (reviewed in REF. 74). They act through a common heterodimeric receptor on infected, and neighbouring uninfected, cells to activate a signal-transduction pathway that triggers the transcription of a diverse set of IFN-inducible genes that establish an antiviral response. IFNα and IFNβ also promote the development of the acquired immune response. IFN can be induced in cells following detection of viral proteins and nucleic acid. One of the most efficient inducers of IFNβ is double-stranded RNA (dsRNA), which can be presented to the cell in several ways, including presentation to the outside of the cell; presentation to endosomes by endocytosis of extracellular dsRNA; uncoating of viral particles; and degradation of engulfed apoptotic cells. One of the cellular detectors of dsRNA is TLR3 (see the figure, part,a). dsRNA binds Toll-like receptor 3 (TLR3), which recruits the adaptor protein TIR domain-containing adapter molecule 1 (TICAM1; also known as TRIF). TRIF acts as a scaffold to signalling components that feed into the IFN-regulatory factor 3 (IRF3) or nuclear factor-κB (NF-κB) pathways, resulting in the transcriptional activation of IFNβ. IFNα and IFNβ signalling involves the Janus kinase (JAK) family of cytoplasmic tyrosine kinases and the signal transducers and activators of transcription (STAT) that are phosphorylated by JAKs and bind to DNA (see the figure, part B).
Treatment of cells with IFNα or IFNβ upregulates the expression of hundreds of genes that contain an IFN-stimulated response element. Some of the best-characterized genes include protein kinase R (PKR; also known as EIF2AK2) and 2′-5′-oligoadenylate synthetase (2′5′OAS), which are synthesized in an inactive form and undergo dimerization and activation in response to dsRNA. Activated PKR results in phosphorylation of the α-subunit of eukaryotic translational initiation factor 2 (eIF2α) and halts eukaryotic transcription. Activation of 2′5′OAS activates RNase L, which degrades cellular and viral RNAs. A simplified schematic of IFN induction and IFN signalling pathways is provided in the figure. ISRE, interferon-stimulated response element.
In addition to triggering pro-apoptotic events, IFN signalling may inhibit virus-induced apoptosis by inhibiting viral replication. IFN signalling involves the Janus kinase (JAK) family of cytoplasmic tyrosine kinases and the signal transducers and activators of transcription (STAT), which are phosphorylated by JAKs and bind to DNA. A role for interferon signalling in the prevention of virus-induced apoptosis has been demonstrated following infection of mice with murine norovirus75. In these experiments, infection and virus-induced apoptosis were inhibited in STAT1-deficient mice75. Mice that lack the Stat1 gene are also more susceptible to reovirus CNS infection, suggesting that IFN signalling can prevent reovirus-induced apoptosis76. Differential IFN and JAK or STAT signalling also plays a part in cell type-specific immune responses in reovirus-infected murine cardiac cells77. IFN signalling is activated following the detection of viruses by several viral sensors, including Toll-like receptor 3 (TLR3), which increases the expression of genes that express IFN through the actions of IFN regulatory factor 3 (IRF3) and IRF7 (REF. 74). The role of IFN signalling in the inhibition of viral replication in the CNS and the limitation of virus-induced apoptotic disease has also been shown following WNV infection of mice deficient in TLR3, IRF3 and IRF7 (REFs 78–80).
Taken together, the studies discussed in this section suggest that virus infection or specific viral proteins can induce apoptosis without the involvement of the immune system, but that in the vast majority of cases the immune system plays a part in the onset of apoptosis in infected cells in animal models of virus-induced disease.
Triggering apoptosis
Both the extrinsic and the intrinsic apoptosis pathways, as well as Jun N-terminal kinase (JNK) signalling (BOX 4), have been shown to be involved in the triggering of apoptosis in animal models of virus-induced disease. Studies on this topic have typically been performed in vitro, but in some cases questions regarding specific apoptotic signalling pathways have also been addressed in vivo.
Box 4. C-Jun N-terminal kinases.
Mitogen-activated protein kinases (MAPKs) are important mediators of signal-transduction pathways that serve to coordinate the cellular response to various extracellular stimuli. The MAPK superfamily includes the c-Jun N-terminal kinases (JNKs) (see the figure), which are activated in response to various environmental stresses and inflammatory signals and promote apoptosis and growth inhibition. JNK influences apoptosis by various mechanisms, including cytochrome c release109 and requires the pro-apoptotic B-cell lymphoma 2 (BCL-2) family proteins BCL-2 associated × protein (BAX) and BCL-2 antagonist killer 1 (BAK1) (REF. 110). It has been proposed that JNK influences cytochrome c release in various ways, including post-translational modification of the BH3-only BCL-2 family proteins Bcl-2-like protein 11 (BCL2L11; also known as BIM) (phosphorylation) and BCL-2 interacting domain death agonist (BID) (cleavage to form jBid), by up-regulating BAX and by promoting accumulation of active BAX at the mitochondria109,111–114. In addition, multiple transcription factors (TFs), including Jun, p53 and activating transcription factor 2 (ATF2), are activated by JNK (JNK-dependent TFs), and these TFs may affect apoptosis by upregulating apoptosis-related genes. For example, JNK can trigger extrinsic apoptotic signalling by inducing the upregulation of death receptors and/or their ligands115–121. JNK itself can also be activated by death receptor (DR)-induced apoptotic signalling (see the figure), suggesting that a positive-feedback loop may occur to amplify apoptosis122–124. DIABLO, direct inhibitor of apoptosis protein-binding protein with low pI; FADD, Fas-associated death domain protein; IAP, inhibitor of apoptosis protein; tBID, truncated tBID..
Extrinsic apoptotic signalling
In the extrinsic apoptotic pathway, death ligands, including FasL, TNF and TRAIL, bind and activate their cognate receptors (Fas, TNF receptors and TRAIL receptors, respectively)81–83. Activated receptors, an adaptor molecule, such as FADD or TNF receptor type 1-associated DeATH domain protein (TRADD), and pro-caspase 8 then form a death-inducing signalling complex, in which pro-caspase 8 undergoes autoactivation. This results in activation of the initiator caspase, caspase 8, which can activate caspase 3 to bring about the apoptotic phenotype (FIG. 1). The extrinsic pathway of apoptosis is central to the process of immune-mediated viral clearance, which has already been discussed above. However, in some cases, viral-induced increases in death receptors and their ligands might also trigger apoptosis in infected cells without the involvement of infiltrating immune cells. For example, following reovirus infection, it seems that Fas is upregulated in infected cells, resulting in the activation of caspase 8 without any involvement of the immune system (P.C. and K.L.T., unpublished observations). In addition, in HBV-mediated acute liver failure, the expression of TRAIL in the liver is not dependent on immune cells and is thought to be a direct response of the infected cell84.
Intrinsic apoptotic signalling
In the intrinsic apoptotic pathway, pro-apoptotic members of the B-cell lymphoma 2 (BCL-2) family of proteins (BCL-2 associated × protein (BAX) and BCL-2 antagonist killer 1 (BAK1)) form pores in the outer mitochondrial membrane (BOX 2; FIG. 1). Pro-apoptotic mitochondrial factors, including cytochrome c and DIABLO (direct inhibitor of apoptosis protein-binding protein with low pI; also known as SMAC), are released through these pores and contribute to apoptosis. Binding of cytochrome c and dATP (deoxyadenosine triphosphate) causes the adaptor molecule, apoptotic protease-activating factor 1 (APAF1), to form a large complex called the apoptosome. The apoptosome recruits pro-caspase 9, which is then processed by autocatalysis. DIABLO inhibits cellular inhibitor of apoptosis proteins (IAPs), which normally function to block the activity of caspase 3 and caspase 9 (REFs 85,86).
It has been proposed that cytochrome c release from the mitochondria occurs by a two-step process87. In the first step, cytochrome c is released from the inner mitochondrial membrane, where it is bound by interactions with cardiolipin. Once released, cytochrome c is then free to leave the mitochondria following permeabilization of the outer mitochondrial membrane by pro-apoptotic BCL-2 family proteins (BOX 2). Release of cytochrome c is brought about by oxidation of cardiolipin by reactive oxygen species (ROS), which are generated through the activity of the mitochondrial respiratory chain.
Evidence that the intrinsic pathway of apoptosis can be involved in virus-induced apoptosis in animal models of virus-induced disease comes from studies which show that the upregulation or activation of pro-apoptotic BCL-2 family members is associated with apoptosis. For example, an increase in the amount of pro-apoptotic BAX mRNA correlates strongly with cardiomyocyte apoptosis in some18,88, but not all89, studies of CVB3-induced myocarditis. Furthermore, glutathione levels decrease during CVB3-induced myocarditis, suggesting that ROS are released from the mitochondria (as a by-product of BAX dimerization and activation). In this study, ROS were associated with high mortality and had a role in the pathogenesis of cardiac damage following CVB3 infection88.
Increases in the ratio of BAX (pro-apoptotic) to BCL-2 (anti-apoptotic) and levels of cytosolic cytochrome c are also associated with apoptosis in the livers of animals infected with rabbit haemorrhagic disease virus, again suggesting that the intrinsic apoptotic signalling pathway is involved in virus-induced disease90. In addition, in HCV transgenic mice, HCV core protein localizes to the mitochondria and promotes ROS generation91,92.
JNK signalling
The mitogen-activated protein kinase (MAPK) JNK is also thought to play a part in apoptosis either by post-translational modifications of BCL-2 family proteins or by its ability to activate transcription factors and promote transcription of apoptosis-related genes (BOX 4). Reovirus infection of the mouse CNS leads to the activation of JNK and JuN (also known as mitogen-activated protein kinase 8; MAPK8) in reovirus-infected neurons in regions that colocalize with areas of virus-induced injury and apoptosis, suggesting that JNK contributes to neuronal apoptosis in these animals34. JNK may contribute to extrinsic apoptotic signalling following reovirus infection of the mouse CNS (P.C. and K.L.T., unpublished observations). Alternatively, or additionally, JNK may mediate reovirus-induced apoptosis in the mouse CNS by activating intrinsic apoptotic signalling, as found following reovirus infection of epithelial cell lines93. JNK is activated following infection with various viruses and may therefore have a more widespread role in virus-induced apoptosis. For example, JNK has a role as a pro-apoptotic factor in HSV-1- and HSV-2-induced CNS disease, although precisely how JNK contributes to apoptosis is unclear2,94.
Taken together, the studies discussed in this section show that multiple apoptotic signalling pathways can be induced in animal models of virus-induced disease. Much still remains unknown, however, including the virus-specific and cell-specific nature of the response and the interactions that exist between the various signalling pathways.
Therapeutic implications
In animal models of virus-induced disease, virus-infected cells thus appear to die by apoptosis. This process can facilitate viral clearance, but is also a mechanism of virus-induced tissue injury and resulting disease. The central role of apoptosis in virus-induced tissue injury has prompted studies designed to test the effect of inhibition of apoptosis on both viral growth and the severity of virus-induced disease.
Effect on viral replication
The use of apoptosis as a host antiviral strategy to eliminate virus-infected cells is counteracted by the capacity of many viruses to encode anti-apoptotic proteins that allow them to complete their replication cycles before destruction of the cell95. In these cases, therapeutic strategies designed to inhibit apoptosis might actually enhance viral replication and increase pathogenicity. The host can also use apoptosis to block viral spread. For example, virus-induced neuronal apoptosis of peripheral neuronal nervous-system cells could be a protective host response that blocks transmission of HSV-2 to the CNS96. Again, therapeutic inhibition of apoptosis under these circumstances might be expected to enhance viral spread and pathogenicity. However, viruses have evolved and adapted to use many cellular processes to their advantage and the same might have happened for apoptosis. The apoptotic death of a cell may facilitate virus egress, and chromatin margination may ‘make room’ for viral replication compartments95. In such cases, inhibition of apoptosis would be expected to inhibit viral replication and pathogenesis.
In light of these potentially contradicting effects, it is perhaps not surprising that inhibition of apoptosis has been reported to have mixed effects on viral replication. For example, in a mouse model of WNV, caspase 3−/− mice have less-severe disease and apoptosis than the wild type, but the CNS titres of WNV do not change15. Similarly, the CNS titre of reovirus-infected mice does not change when virus-induced apoptosis and tissue injury are reduced. Consequently, the CNS titre of reovirus-infected mice does not change compared with wild-type controls following pharmacological inhibition of JNK34, following infection with an apoptosis-deficient reovirus strain31 or in mice that do not express the P50 subunit of the transcription factor nuclear factor-κB22. By contrast, apoptosis and disease severity are reduced and viral titre decreases following infection of mice with a recombinant SINV chimera that expresses BCL-2 compared with the virus from which it was derived97. Similarly, virus titre decreases in reovirus-infected hearts of caspase 3−/− mice compared with wild-type mice21. In addition, inhibition of CVB3 replication in vivo with the drug WIN 54954 reduces both viral replication and apoptosis98. These differences are probably due to differences in viral replication strategies that have evolved to enable viruses to survive in the host or by organ-specific differences in antiviral responses.
Effect on severity of disease
Studies in animals have shown that conditions designed to inhibit apoptosis reduce disease severity, even though there were conflicting effects on viral titre15,21,22,31,34,97,98 (discussed above). These studies provide strong evidence that apoptosis is an important mechanism of virus-induced disease and that anti-apoptotic treatments may be beneficial in human virus-induced diseases.
Reports show that caspase 3−/− mice, or mice treated with pharmacological caspase inhibitors, are more resistant to virus-induced disease following infection of the CNS with WNV15, following infection of the heart with reovirus21 (FIG. 2) and following infection with rhesus rotavirus in a mouse model of biliary atresia99. Inhibition of specific apoptotic pathways can also inhibit virus-induced disease. For example, the anti-apoptotic mitochondrial proteins BCL-2 (REF. 97) and peripheral benzodiazepine receptor100 are protective following CNS infection with SINV, and N-acetyl-cysteine90 and astragaloside101, both of which prevent oxidative stress, prevent virus-induced liver and heart injury, respectively. Furthermore, intracerebral injection of recombinant simian virus 40 (SV40) vectors that carry Cu–Zn superoxide dismutase 1 (SOD1) or glutathione peroxidase 1 (GPX1) significantly protected neurons from HIV-1 envelope gp120 (REF. 102), and pharmacological inhibition of JNK signalling, which is upregulated following reovirus infection in vivo (discussed above), and has been shown to be required for reovirus-induced apoptosis in vitro, decreased disease severity following reovirus infection of the mouse CNS34. Anti-FasL antibody prevents hepatocyte apoptosis and proliferation, liver inflammation and the development of hepatocellular carcinoma in transgenic mice that express HBV proteins27. Finally, soluble TRAIL inhibits liver damage by blocking apoptosis in an acute hepatitis model in which BALB/c mice were transfected with pcDNA3-HBV1.1, which contains the entire HBV genome103, and silencing TRADD expression with small-interfering RNA reduces neuronal apoptosis and subsequent microglial and astroglial activation104,105.
Figure 2. Inhibition of caspase 3, apoptosis (caspase 3 activation) and survival in reovirus-infected mice.
Consecutive sections from reovirus-infected hearts were analysed for histological injury (haematoxylin and eosin staining), active caspase 3 (brown diaminobenzadine staining) and virus antigen (fluorescent green staining) in mice treated with the pharmacological caspase inhibitor Q-VD-OPH (a–c) and in non-treated control mice (d–f). Survival curves for reovirus-infected caspase 3−/− mice and wild-type controls are also shown (g). In the caspase 3−/− mice, 37.5% of animals survived, and cardiac tissues from long-term survivors appeared to be normal (54 days post-infection). Figure modified, with permission, from REF. 21 © (2004) American Society for Microbiology.
Concluding remarks
Apoptotic cells occur in the CNS, heart and liver following infection with various viruses or expression of viral proteins. In many cases, these apoptotic cells colocalize to the same areas or specific cells as viral antigen. Apoptosis correlates with disease severity in a number of different animal models of virus-induced disease. Furthermore, several studies have shown that there is a decrease in disease severity following treatment with anti-apoptotic agents. These studies indicate that apoptosis is an important mechanism in viral-induced disease and suggest that apoptosis and apoptotic signalling pathways may provide novel targets for treatment of virus-induced human disease. However, it must be remembered that viruses have different strategies to benefit from or evade apoptosis. Accordingly, it will be important to define the specific role of apoptosis in the pathogenesis of individual viral infections. For example, in some cases, virus-induced chronic disease can be immunologically mediated and result from the inability to terminate the immune response, and therefore inhibition of apoptosis might actually exacerbate disease.
Further work is required to determine whether inhibition of apoptosis ameliorates or worsens virus disease in relevant animal models. Then, for cases in which the inhibition of apoptosis promotes survival or reduces tissue injury, it is crucial to define the apoptotic signalling pathways involved. With this knowledge, we could identify specific targets for potential antiviral therapy.
Acknowledgments
Work in the authors’ laboratory was supported by the National Institutes of Health (grant numbers T32AI07537, 5RO1NS050138 and 1RO1NS051403) and a VA Merit and Career Development Award. K.L.T. is supported by the Reuler–Lewin Family Professorship.
- Extrinsic apoptotic pathway
An apoptotic signalling pathway that is triggered by the binding of ligands to cell-surface death receptors and culminates in the activation of the initiator caspase, caspase 8
- Intrinsic apoptotic pathway
An apoptotic signalling pathway that involves the release of pro-apoptotic factors from the mitochondria and activation of the initiator caspase, caspase 9
- Cytotoxic T lymphocyte
A T lymphocyte that induces the death of pathogen-infected cells
- Oligonucleosomal DNA laddering
A generally accepted biochemical criterion of apoptosis that involves cleavage of DNA into 180 bp fragments by a caspase-activated DNase
- TUNEL
(Terminal deoxynucleotidyl transferase dUTP nick end labelling). A method used to label free ends of DNA, which increase during apoptosis owing to the action of a caspase-activated DNase
- Astrogliosis
An abnormal increase in the number of astrocytes
- Microglial proliferation
An abnormal increase in the number of microglia
- Inducible nitric oxide synthase
An enzyme involved in the generation of nitric oxide, which can result in cell death through apoptosis or necrosis
- Macrophage
A cell within tissues that originates from specific white blood cells called monocytes. Monocytes and macrophages are phagocytes that engulf and then digest cellular debris and pathogens. They also stimulate lymphocytes to respond to the pathogen
- Lymphocyte
A white blood cell from the vertebrate immune system. Lymphocytes include large granular lymphocytes, commonly known as natural killer (NK) cells, and small lymphocytes (T and B cells). NK cells are a part of the innate immune system and help defend the host from tumours and virally infected cells by releasing cytotoxic granules. T cells and B cells are the main cellular components of the adaptive immune response. B lymphocytes respond to pathogens by producing large quantities of antibodies that neutralize foreign objects, such as bacteria and viruses. T cells include helper T cells, which produce cytokines that direct the immune response, and cytotoxic T cells, which induce the death of pathogen-infected cells
- Neutrophil
The most abundant type of white blood cell in humans. Neutrophils are normally found in the bloodstream and form an essential part of the immune system. However, during the acute phase of inflammation, neutrophils migrate towards the site of inflammation, usually as a result of bacterial infection
- Fas ligand
A member of the tumour necrosis factor family of cytokines that triggers apoptosis following the binding of cell-surface receptors
- TRAIL
(Tumour necrosis factor (TNF)-related apoptosis inducing ligand). A member of the TNF family of cytokines that triggers apoptosis following the binding of cell-surface receptors
- Microglia
A type of glial cell that acts as the first and main form of active immune defence in the central nervous system
- Astrocyte
A type of non-neuronal glial cell that performs many functions, including biochemical support of endothelial cells, that form the blood–brain barrier, provision of nutrients to the nervous tissue and aiding the repair and scarring process in the brain
- Reactive oxygen species
(ROs). Highly reactive ions or very small molecules that include oxygen ions, free radicals and peroxides. During oxidative stress, increased ROs levels can result in substantial damage to cell structures
References
- 1.Li W, Galey D, Mattson MP, Nath A. Molecular and cellular mechanisms of neuronal cell death in HIV dementia. Neurotox Res. 2005;8:119–134. doi: 10.1007/BF03033824. [DOI] [PubMed] [Google Scholar]
- 2.Perkins D, Gyure KA, Pereira EF, Aurelian L. Herpes simplex virus type 1-induced encephalitis has an apoptotic component associated with activation of c-Jun N-terminal kinase. J Neurovirol. 2003;9:101–111. doi: 10.1080/13550280390173427. [DOI] [PubMed] [Google Scholar]
- 3.DeBiasi RL, Kleinschmidt-Demasters BK, Richardson-Burns S, Tyler KL. Central nervous system apoptosis in human herpes simplex virus and cytomegalovirus encephalitis. J Infect Dis. 2002;186:1547–1557. doi: 10.1086/345375. [DOI] [PubMed] [Google Scholar]
- 4.Alter P, Jobmann M, Meyer E, Pankuweit S, Maisch B. Apoptosis in myocarditis and dilated cardiomyopathy: does enterovirus genome persistence protect from apoptosis? An endomyocardial biopsy study. Cardiovasc Pathol. 2001;10:229–234. doi: 10.1016/s1054-8807(01)00077-1. [DOI] [PubMed] [Google Scholar]
- 5.Bantel H, Schulze-Osthoff K. Apoptosis in hepatitis C virus infection. Cell Death Differ. 2003;10 (Suppl 1):48–58. doi: 10.1038/sj.cdd.4401119. [DOI] [PubMed] [Google Scholar]
- 6.Calabrese F, et al. Liver cell apoptosis in chronic hepatitis C correlates with histological but not biochemical activity or serum HCV-RNA levels. Hepatology. 2000;31:1153–1159. doi: 10.1053/he.2000.7123. [DOI] [PubMed] [Google Scholar]
- 7.Patel T, Roberts LR, Jones BA, Gores GJ. Dysregulation of apoptosis as a mechanism of liver disease: an overview. Semin Liver Dis. 1998;18:105–114. doi: 10.1055/s-2007-1007147. [DOI] [PubMed] [Google Scholar]
- 8.Rodrigues CM, et al. Apoptotic cell death does not parallel other indicators of liver damage in chronic hepatitis C patients. J Viral Hepat. 2000;7:175–183. doi: 10.1046/j.1365-2893.2000.00219.x. [DOI] [PubMed] [Google Scholar]
- 9.Walsh MJ, et al. Steatosis and liver cell apoptosis in chronic hepatitis C: a mechanism for increased liver injury. Hepatology. 2004;39:1230–1238. doi: 10.1002/hep.20179. [DOI] [PubMed] [Google Scholar]
- 10.Leifeld L, et al. Intrahepatic activation of caspases in human fulminant hepatic failure. Liver Int. 2006;26:872–879. doi: 10.1111/j.1478-3231.2006.01300.x. [DOI] [PubMed] [Google Scholar]
- 11.Nunez G, Benedict MA, Hu Y, Inohara N. Caspases: the proteases of the apoptotic pathway. Oncogene. 1998;17:3237–3245. doi: 10.1038/sj.onc.1202581. [DOI] [PubMed] [Google Scholar]
- 12.Cohen GM. Caspases: the executioners of apoptosis. Biochem J. 1997;326:1–16. doi: 10.1042/bj3260001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Thornberry NA, Lazebnik Y. Caspases: enemies within. Science. 1998;281:1312–1316. doi: 10.1126/science.281.5381.1312. [DOI] [PubMed] [Google Scholar]
- 14.Heibein JA, et al. Granzyme B-mediated cytochrome c release is regulated by the Bcl-2 family members Bid and Bax. J Exp Med. 2000;192:1391–1402. doi: 10.1084/jem.192.10.1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Samuel MA, Morrey JD, Diamond MS. Caspase 3-dependent cell death of neurons contributes to the pathogenesis of West Nile virus encephalitis. J Virol. 2007;81:2614–2623. doi: 10.1128/JVI.02311-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Oberhaus SM, Smith RL, Clayton GH, Dermody TS, Tyler KL. Reovirus infection and tissue injury in the mouse central nervous system are associated with apoptosis. J Virol. 1997;71:2100–2106. doi: 10.1128/jvi.71.3.2100-2106.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Labrada L, Liang XH, Zheng W, Johnston C, Levine B. Age-dependent resistance to lethal alphavirus encephalitis in mice: analysis of gene expression in the central nervous system and identification of a novel interferon-inducible protective gene, mouse ISG12. J Virol. 2002;76:11688–11703. doi: 10.1128/JVI.76.22.11688-11703.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Joo CH, et al. Coxsackievirus B3 induces apoptosis in the early phase of murine myocarditis: a comparative analysis of cardiovirulent and noncardiovirulent strains. Intervirology. 2003;46:135–140. doi: 10.1159/000071453. [DOI] [PubMed] [Google Scholar]
- 19.Saraste A, et al. Cardiomyocyte apoptosis in experimental coxsackievirus B3 myocarditis. Cardiovasc Pathol. 2003;12:255–262. doi: 10.1016/s1054-8807(03)00077-2. [DOI] [PubMed] [Google Scholar]
- 20.Henke A, Huber S, Stelzner A, Whitton JL. The role of CD8+ T lymphocytes in coxsackievirus B3-induced myocarditis. J Virol. 1995;69:6720–6728. doi: 10.1128/jvi.69.11.6720-6728.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.DeBiasi RL, et al. Caspase inhibition protects against reovirus-induced myocardial injury in vitro and in vivo. J Virol. 2004;78:11040–11050. doi: 10.1128/JVI.78.20.11040-11050.2004. This study shows that inhibition of caspase 3 reduces the severity of reovirus-induced cardiac disease and enhances survival in infected mice. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.O’Donnell SM, et al. Organ-specific roles for transcription factor NF-κB in reovirus-induced apoptosis and disease. J Clin Invest. 2005;115:2341–2350. doi: 10.1172/JCI22428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Riedl SJ, et al. Structural basis for the inhibition of caspase-3 by XIAP. Cell. 2001;104:791–800. doi: 10.1016/s0092-8674(01)00274-4. [DOI] [PubMed] [Google Scholar]
- 24.Lenzo JC, Fairweather D, Cull V, Shellam GR, James Lawson CM. Characterisation of murine cytomegalovirus myocarditis: cellular infiltration of the heart and virus persistence. J Mol Cell Cardiol. 2002;34:629–640. doi: 10.1006/jmcc.2002.2003. [DOI] [PubMed] [Google Scholar]
- 25.Irie H, et al. Herpes simplex virus hepatitis in macrophage-depleted mice: the role of massive, apoptotic cell death in pathogenesis. J Gen Virol. 1998;79:1225–1231. doi: 10.1099/0022-1317-79-5-1225. [DOI] [PubMed] [Google Scholar]
- 26.Xiao SY, Guzman H, Zhang H, Travassos da Rosa AP, Tesh RB. West Nile virus infection in the golden hamster (Mesocricetus auratus): a model for West Nile encephalitis. Emerg Infect Dis. 2001;7:714–721. doi: 10.3201/eid0704.010420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Nakamoto Y, et al. Prevention of hepatocellular carcinoma development associated with chronic hepatitis by anti-Fas ligand antibody therapy. J Exp Med. 2002;196:1105–1111. doi: 10.1084/jem.20020633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Tumurbaatar B, Sun Y, Chan T, Sun J. Cre-estrogen receptor-mediated hepatitis C virus structural protein expression in mice. J Virol Methods. 2007;146:5–13. doi: 10.1016/j.jviromet.2007.05.025. This study shows that inducible expression of HCV core proteins induces apoptosis in the hepatocytes of transgenic mice. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Richardson-Burns SM, Tyler KL. Regional differences in viral growth and central nervous system injury correlate with apoptosis. J Virol. 2004;78:5466–5475. doi: 10.1128/JVI.78.10.5466-5475.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.DeBiasi RL, Edelstein CL, Sherry B, Tyler KL. Calpain inhibition protects against virus-induced apoptotic myocardial injury. J Virol. 2001;75:351–361. doi: 10.1128/JVI.75.1.351-361.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Hoyt CC, et al. Nonstructural protein σ1s is a determinant of reovirus virulence and influences the kinetics and severity of apoptosis induction in the heart and central nervous system. J Virol. 2005;79:2743–2753. doi: 10.1128/JVI.79.5.2743-2753.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Tsunoda I, Kurtz CI, Fujinami RS. Apoptosis in acute and chronic central nervous system disease induced by Theiler’s murine encephalomyelitis virus. Virology. 1997;228:388–393. doi: 10.1006/viro.1996.8382. [DOI] [PubMed] [Google Scholar]
- 33.Lewis J, Wesselingh SL, Griffin DE, Hardwick JM. Alphavirus-induced apoptosis in mouse brains correlates with neurovirulence. J Virol. 1996;70:1828–1835. doi: 10.1128/jvi.70.3.1828-1835.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Beckham JD, Goody RJ, Clarke P, Bonny C, Tyler KL. Novel strategy for treatment of viral central nervous system infection by using a cell-permeating inhibitor of c-Jun N-terminal kinase. J Virol. 2007;81:6984–6992. doi: 10.1128/JVI.00467-07. This paper reports that the inhibition of JNK can be used to block neuronal apoptosis, decrease the severity of reovirus-induced CNS disease and enhance survival in infected mice. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Goody RJ, Schittone SA, Tyler KL. Experimental reovirus-induced acute flaccid paralysis and spinal motor neuron cell death. J Neuropathol Exp Neurol. 2008;67:231–239. doi: 10.1097/NEN.0b013e31816564f0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Nakamoto Y, Suda T, Momoi T, Kaneko S. Different procarcinogenic potentials of lymphocyte subsets in a transgenic mouse model of chronic hepatitis B. Cancer Res. 2004;64:3326–3333. doi: 10.1158/0008-5472.can-03-3817. [DOI] [PubMed] [Google Scholar]
- 37.Morrey JD, et al. West Nile virus-induced acute flaccid paralysis is prevented by monoclonal antibody treatment when administered after infection of spinal cord neurons. J Neurovirol. 2008;14:152–163. doi: 10.1080/13550280801958930. This study shows that WNV-induced acute flaccid paralysis in hamsters is due to neuronal infection and apoptosis in the lumbar spinal cord and that treatment with a therapeutic antibody prevents paralysis when administered after WNV infection. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Schoneboom BA, Catlin KM, Marty AM, Grieder FB. Inflammation is a component of neurodegeneration in response to Venezuelan equine encephalitis virus infection in mice. J Neuroimmunol. 2000;109:132–146. doi: 10.1016/s0165-5728(00)00290-3. [DOI] [PubMed] [Google Scholar]
- 39.Weissenbock H, Hornig M, Hickey WF, Lipkin WI. Microglial activation and neuronal apoptosis in Bornavirus infected neonatal Lewis rats. Brain Pathol. 2000;10:260–272. doi: 10.1111/j.1750-3639.2000.tb00259.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Andrews DM, Matthews VB, Sammels LM, Carrello AC, McMinn PC. The severity of murray valley encephalitis in mice is linked to neutrophil infiltration and inducible nitric oxide synthase activity in the central nervous system. J Virol. 1999;73:8781–8790. doi: 10.1128/jvi.73.10.8781-8790.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Matthews V, et al. Morphological features of Murray Valley encephalitis virus infection in the central nervous system of Swiss mice. Int J Exp Pathol. 2000;81:31–40. doi: 10.1046/j.1365-2613.2000.00135.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Taylor RM, Hurlbut HS, Work TH, Kingston JR, Frothingham TE. Sindbis virus: a newly recognized arthropod-transmitted virus. Am J Trop Med Hyg. 1955;4:844–862. doi: 10.4269/ajtmh.1955.4.844. [DOI] [PubMed] [Google Scholar]
- 43.Tsunoda I, Libbey JE, Fujinami RS. TGF-β1 suppresses T cell infiltration and VP2 puff B mutation enhances apoptosis in acute polioencephalitis induced by Theiler’s virus. J Neuroimmunol. 2007;190:80–89. doi: 10.1016/j.jneuroim.2007.07.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Shrestha B, Gottlieb D, Diamond MS. Infection and injury of neurons by West Nile encephalitis virus. J Virol. 2003;77:13203–13213. doi: 10.1128/JVI.77.24.13203-13213.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Huber SA, Mortensen A, Moulton G. Modulation of cytokine expression by CD4+ T cells during coxsackievirus B3 infections of BALB/c mice initiated by cells expressing the γδ+ T-cell receptor. J Virol. 1996;70:3039–3044. doi: 10.1128/jvi.70.5.3039-3044.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Rasalingam P, Rossiter JP, Jackson AC. Recombinant rabies virus vaccine strain SAD-l16 inoculated intracerebrally in young mice produces a severe encephalitis with extensive neuronal apoptosis. Can J Vet Res. 2005;69:100–105. [PMC free article] [PubMed] [Google Scholar]
- 47.Jackson AC, Rasalingam P, Weli SC. Comparative pathogenesis of recombinant rabies vaccine strain SAD-L16 and SAD-D29 with replacement of Arg333 in the glycoprotein after peripheral inoculation of neonatal mice: less neurovirulent strain is a stronger inducer of neuronal apoptosis. Acta Neuropathol. 2006;111:372–378. doi: 10.1007/s00401-005-0006-z. [DOI] [PubMed] [Google Scholar]
- 48.Sarmento L, Tseggai T, Dhingra V, Fu ZF. Rabies virus-induced apoptosis involves caspase-dependent and caspase-independent pathways. Virus Res. 2006;121:144–151. doi: 10.1016/j.virusres.2006.05.002. [DOI] [PubMed] [Google Scholar]
- 49.Reid JE, Jackson AC. Experimental rabies virus infection in Artibeus jamaicensis bats with CVS-24 variants. J Neurovirol. 2001;7:511–517. doi: 10.1080/135502801753248097. [DOI] [PubMed] [Google Scholar]
- 50.Yan X, et al. Silver-haired bat rabies virus variant does not induce apoptosis in the brain of experimentally infected mice. J Neurovirol. 2001;7:518–527. doi: 10.1080/135502801753248105. [DOI] [PubMed] [Google Scholar]
- 51.Park CH, et al. The histopathogenesis of paralytic rabies in six-week-old C57BL/6J mice following inoculation of the CVS-11 strain into the right triceps surae muscle. J Vet Med Sci. 2006;68:589–595. doi: 10.1292/jvms.68.589. [DOI] [PubMed] [Google Scholar]
- 52.Jackson AC, Scott CA, Owen J, Weli SC, Rossiter JP. Therapy with minocycline aggravates experimental rabies in mice. J Virol. 2007;81:6248–6253. doi: 10.1128/JVI.00323-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Henke A, et al. Apoptosis in coxsackievirus B3-caused diseases: interaction between the capsid protein VP2 and the proapoptotic protein siva. J Virol. 2000;74:4284–4290. doi: 10.1128/jvi.74.9.4284-4290.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Oleszak EL, et al. Apoptosis of infiltrating T cells in the central nervous system of mice infected with Theiler’s murine encephalomyelitis virus. Virology. 2003;315:110–123. doi: 10.1016/s0042-6822(03)00517-8. [DOI] [PubMed] [Google Scholar]
- 55.Shrestha B, Diamond MS. Fas ligand interactions contribute to CD8+ T-cell-mediated control of West Nile virus infection in the central nervous system. J Virol. 2007;81:11749–11757. doi: 10.1128/JVI.01136-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wang T, et al. IFN-γ-producing γδ T cells help control murine West Nile virus infection. J Immunol. 2003;171:2524–2531. doi: 10.4049/jimmunol.171.5.2524. [DOI] [PubMed] [Google Scholar]
- 57.Huber SA, Born W, O’Brien R. Dual functions of murine γδ cells in inflammation and autoimmunity in coxsackievirus B3-induced myocarditis: role of Vγ1+ and Vγ4+ cells. Microbes Infect. 2005;7:537–543. doi: 10.1016/j.micinf.2004.12.011. [DOI] [PubMed] [Google Scholar]
- 58.Machida K, et al. Inhibition of cytochrome c release in Fas-mediated signaling pathway in transgenic mice induced to express hepatitis C viral proteins. J Biol Chem. 2001;276:12140–12146. doi: 10.1074/jbc.M010137200. [DOI] [PubMed] [Google Scholar]
- 59.Weant AE, et al. Apoptosis regulators Bim and Fas function concurrently to control autoimmunity and CD8+ T cell contraction. Immunity. 2008;28:218–230. doi: 10.1016/j.immuni.2007.12.014. [DOI] [PubMed] [Google Scholar]
- 60.Fischer SF, Belz GT, Strasser A. BH3-only protein Puma contributes to death of antigen-specific T cells during shutdown of an immune response to acute viral infection. Proc Natl Acad Sci USA. 2008;105:3035–3040. doi: 10.1073/pnas.0706913105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Budd RC. Activation-induced cell death. Curr Opin Immunol. 2001;13:356–362. doi: 10.1016/s0952-7915(00)00227-2. [DOI] [PubMed] [Google Scholar]
- 62.Mohindru M, Kang B, Kim BS. Initial capsid-specific CD4+ T cell responses protect against Theiler’s murine encephalomyelitis virus-induced demyelinating disease. Eur J Immunol. 2006;36:2106–2115. doi: 10.1002/eji.200535785. [DOI] [PubMed] [Google Scholar]
- 63.Palma JP, Yauch RL, Lang S, Kim BS. Potential role of CD4+ T cell-mediated apoptosis of activated astrocytes in Theiler’s virus-induced demyelination. J Immunol. 1999;162:6543–6551. [PubMed] [Google Scholar]
- 64.Kimura T, Griffin DE. Extensive immune-mediated hippocampal damage in mice surviving infection with neuroadapted Sindbis virus. Virology. 2003;311:28–39. doi: 10.1016/s0042-6822(03)00110-7. CD4+ T cells were shown to promote progressive neuronal death and tissue injury following sindbis virus infection, despite clearance of infectious virus. [DOI] [PubMed] [Google Scholar]
- 65.Huber SA, Budd RC, Rossner K, Newell MK. Apoptosis in coxsackievirus B3-induced myocarditis and dilated cardiomyopathy. Ann NY Acad Sci. 1999;887:181–190. doi: 10.1111/j.1749-6632.1999.tb07932.x. [DOI] [PubMed] [Google Scholar]
- 66.Kondo T, Suda T, Fukuyama H, Adachi M, Nagata S. Essential roles of the Fas ligand in the development of hepatitis. Nature Med. 1997;3:409–413. doi: 10.1038/nm0497-409. [DOI] [PubMed] [Google Scholar]
- 67.Soguero C, et al. Hepatitis C virus core protein leads to immune suppression and liver damage in a transgenic murine model. J Virol. 2002;76:9345–9354. doi: 10.1128/JVI.76.18.9345-9354.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Iken K, Huang L, Bekele H, Schmidt EV, Koziel MJ. Apoptosis of activated CD4+ and CD8+ T cells is enhanced by co-culture with hepatocytes expressing hepatitis C virus (HCV) structural proteins through FasL induction. Virology. 2006;346:363–372. doi: 10.1016/j.virol.2005.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Gujar SA, Jenkins AK, Guy CS, Wang J, Michalak TI. Aberrant lymphocyte activation precedes delayed virus-specific T-cell response after both primary infection and secondary exposure to hepadnavirus in the woodchuck model of hepatitis B virus infection. J Virol. 2008;82:6992–7008. doi: 10.1128/JVI.00661-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Baloul L, Camelo S, Lafon M. Up-regulation of Fas ligand (FasL) in the central nervous system: a mechanism of immune evasion by rabies virus. J Neurovirol. 2004;10:372–382. doi: 10.1080/13550280490521122. [DOI] [PubMed] [Google Scholar]
- 71.Cicin-Sain L, et al. Dominant-negative FADD rescues the in vivo fitness of a cytomegalovirus lacking an antiapoptotic viral gene. J Virol. 2008;82:2056–2064. doi: 10.1128/JVI.01803-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Maher SG, Romero-Weaver AL, Scarzello AJ, Gamero AM. Interferon: cellular executioner or white knight? Curr Med Chem. 2007;14:1279–1289. doi: 10.2174/092986707780597907. [DOI] [PubMed] [Google Scholar]
- 73.Sato K, et al. Antiviral response by natural killer cells through TRAIL gene induction by IFN-α/β. Eur J Immunol. 2001;31:3138–3146. doi: 10.1002/1521-4141(200111)31:11<3138::aid-immu3138>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
- 74.Randall RE, Goodbourn S. Interferons and viruses: an interplay between induction, signalling, antiviral responses and virus countermeasures. J Gen Virol. 2008;89:1–47. doi: 10.1099/vir.0.83391-0. [DOI] [PubMed] [Google Scholar]
- 75.Mumphrey SM, et al. Murine norovirus 1 infection is associated with histopathological changes in immunocompetent hosts, but clinical disease is prevented by STAT1-dependent interferon responses. J Virol. 2007;81:3251–3263. doi: 10.1128/JVI.02096-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Goody RJ, Beckham JD, Rubtsova K, Tyler KL. JAK–STAT signaling pathways are activated in the brain following reovirus infection. J Neurovirol. 2007;13:373–383. doi: 10.1080/13550280701344983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Zurney J, Howard KE, Sherry B. Basal expression levels of IFNAR and Jak–STAT components are determinants of cell-type-specific differences in cardiac antiviral responses. J Virol. 2007;81:13668–13680. doi: 10.1128/JVI.01172-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Daffis S, et al. Interferon regulatory factor IRF-7 induces the antiviral alpha interferon response and protects against lethal West Nile virus infection. J Virol. 2008;82:8465–8475. doi: 10.1128/JVI.00918-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Daffis S, Samuel MA, Suthar MS, Gale M, Jr, Diamond MS. Toll-like receptor-3 has a protective role against West Nile virus infection. J Virol. 2008;82:10349–10358. doi: 10.1128/JVI.00935-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Daffis S, Samuel MA, Keller BC, Gale M, Jr, Diamond MS. Cell-specific IRF-3 responses protect against West Nile virus infection by interferon-dependent and -independent mechanisms. PLoS Pathog. 2007;3:e106. doi: 10.1371/journal.ppat.0030106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Thorburn A. Death receptor-induced cell killing. Cell Signal. 2004;16:139–144. doi: 10.1016/j.cellsig.2003.08.007. [DOI] [PubMed] [Google Scholar]
- 82.Schneider P, Tschopp J. Apoptosis induced by death receptors. Pharm Acta Helv. 2000;74:281–286. doi: 10.1016/s0031-6865(99)00038-2. [DOI] [PubMed] [Google Scholar]
- 83.Ware CF, VanArsdale S, VanArsdale TL. Apoptosis mediated by the TNF-related cytokine and receptor families. J Cell Biochem. 1996;60:47–55. doi: 10.1002/(SICI)1097-4644(19960101)60:1%3C47::AID-JCB8%3E3.0.CO;2-3. [DOI] [PubMed] [Google Scholar]
- 84.Mundt B, et al. Involvement of TRAIL and its receptors in viral hepatitis. FASEB J. 2003;17:94–96. doi: 10.1096/fj.02-0537fje. [DOI] [PubMed] [Google Scholar]
- 85.Green DR, Reed JC. Mitochondria and apoptosis. Science. 1998;281:1309–1312. doi: 10.1126/science.281.5381.1309. [DOI] [PubMed] [Google Scholar]
- 86.Green DR. Apoptotic pathways: ten minutes to dead. Cell. 2005;121:671–674. doi: 10.1016/j.cell.2005.05.019. [DOI] [PubMed] [Google Scholar]
- 87.Ott M, Gogvadze V, Orrenius S, Zhivotovsky B. Mitochondria, oxidative stress and cell death. Apoptosis. 2007;12:913–922. doi: 10.1007/s10495-007-0756-2. [DOI] [PubMed] [Google Scholar]
- 88.Kyto V, et al. Glutathione depletion and cardiomyocyte apoptosis in viral myocarditis. Eur J Clin Invest. 2004;34:167–175. doi: 10.1111/j.1365-2362.2004.01313.x. [DOI] [PubMed] [Google Scholar]
- 89.Colston JT, Chandrasekar B, Freeman GL. Expression of apoptosis-related proteins in experimental coxsackievirus myocarditis. Cardiovasc Res. 1998;38:158–168. doi: 10.1016/s0008-6363(97)00323-4. [DOI] [PubMed] [Google Scholar]
- 90.San Miguel B, Alvarez M, Culebras JM, Gonzalez-Gallego J, Tunon MJ. N-acetyl-cysteine protects liver from apoptotic death in an animal model of fulminant hepatic failure. Apoptosis. 2006;11:1945–1957. doi: 10.1007/s10495-006-0090-0. [DOI] [PubMed] [Google Scholar]
- 91.Wang T, Weinman SA. Causes and consequences of mitochondrial reactive oxygen species generation in hepatitis C. J Gastroenterol Hepatol. 2006;21 (Suppl 3):34–37. doi: 10.1111/j.1440-1746.2006.04591.x. [DOI] [PubMed] [Google Scholar]
- 92.Chang ML, et al. Acute expression of hepatitis C core protein in adult mouse liver: mitochondrial stress and apoptosis. Scand J Gastroenterol. 2008;43:747–755. doi: 10.1080/00365520701875987. [DOI] [PubMed] [Google Scholar]
- 93.Clarke P, et al. JNK regulates the release of proapoptotic mitochondrial factors in reovirus-infected cells. J Virol. 2004;78:13132–13138. doi: 10.1128/JVI.78.23.13132-13138.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Mori I, et al. The US3 protein kinase of herpes simplex virus attenuates the activation of the c-Jun N-terminal protein kinase signal transduction pathway in infected piriform cortex neurons of C57BL/6 mice. Neurosci Lett. 2003;351:201–205. doi: 10.1016/j.neulet.2003.08.033. [DOI] [PubMed] [Google Scholar]
- 95.Blaho JA. Virus infection and apoptosis (issue II) an introduction: cheating death or death as a fact of life? Int Rev Immunol. 2004;23:1–6. doi: 10.1080/0883018049026551. [DOI] [PubMed] [Google Scholar]
- 96.Mori I, et al. Herpes simplex virus US3 protein kinase regulates virus-induced apoptosis in olfactory and vomeronasal chemosensory neurons in vivo. Microbes Infect. 2006;8:1806–1812. doi: 10.1016/j.micinf.2006.02.018. [DOI] [PubMed] [Google Scholar]
- 97.Levine B, Goldman JE, Jiang HH, Griffin DE, Hardwick JM. Bc1–2 protects mice against fatal alphavirus encephalitis. Proc Natl Acad Sci USA. 1996;93:4810–4815. doi: 10.1073/pnas.93.10.4810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Kyto V, et al. Cardiomyocyte apoptosis after antiviral WIN 54954 treatment in murine coxsackievirus B3 myocarditis. Scand Cardiovasc J. 2002;36:187–192. doi: 10.1080/cdv.36.3.187.192. [DOI] [PubMed] [Google Scholar]
- 99.Erickson N, et al. Temporal–spatial activation of apoptosis and epithelial injury in murine experimental biliary atresia. Hepatology. 2008;47:1567–1577. doi: 10.1002/hep.22229. [DOI] [PubMed] [Google Scholar]
- 100.Johnston C, Jiang W, Chu T, Levine B. Identification of genes involved in the host response to neurovirulent alphavirus infection. J Virol. 2001;75:10431–10445. doi: 10.1128/JVI.75.21.10431-10445.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Zhang ZC, et al. Effect of astragaloside on cardiomyocyte apoptosis in murine coxsackievirus B3 myocarditis. J Asian Nat Prod Res. 2007;9:145–151. doi: 10.1080/10286020412331286506. [DOI] [PubMed] [Google Scholar]
- 102.Louboutin JP, Agrawal L, Reyes BA, Van Bockstaele EJ, Strayer DS. Protecting neurons from HIV-1 gp120-induced oxidant stress using both localized intracerebral and generalized intraventricular administration of antioxidant enzymes delivered by SV40-derived vectors. Gene Ther. 2007;14:1650–1661. doi: 10.1038/sj.gt.3303030. [DOI] [PubMed] [Google Scholar]
- 103.Liu YG, et al. Blockade of TRAIL pathway ameliorates HBV-induced hepatocyte apoptosis in an acute hepatitis model. Biochem Biophys Res Commun. 2007;352:329–334. doi: 10.1016/j.bbrc.2006.11.024. This study shows that recombinant human soluble death receptor 5 (sDR5) is a potential novel therapeutic drug for patients with fulminant hepatitis. [DOI] [PubMed] [Google Scholar]
- 104.Swarup V, Ghosh J, Das S, Basu A. Tumor necrosis factor receptor-associated death domain mediated neuronal death contributes to the glial activation and subsequent neuroinflammation in Japanese encephalitis. Neurochem Int. 2008;52:1310–1321. doi: 10.1016/j.neuint.2008.01.014. [DOI] [PubMed] [Google Scholar]
- 105.Swarup V, Das S, Ghosh S, Basu A. Tumor necrosis factor receptor-1-induced neuronal death by TRADD contributes to the pathogenesis of Japanese encephalitis. J Neurochem. 2007;103:771–783. doi: 10.1111/j.1471-4159.2007.04790.x. [DOI] [PubMed] [Google Scholar]
- 106.Hunter AM, LaCasse EC, Korneluk RG. The inhibitors of apoptosis (IAPs) as cancer targets. Apoptosis. 2007;12:1543–1568. doi: 10.1007/s10495-007-0087-3. [DOI] [PubMed] [Google Scholar]
- 107.Youle RJ, Strasser A. The BCL-2 protein family: opposing activities that mediate cell death. Nature Rev Mol Cell Biol. 2008;9:47–59. doi: 10.1038/nrm2308. [DOI] [PubMed] [Google Scholar]
- 108.Chipuk JE, Green DR. How do BCL-2 proteins induce mitochondrial outer membrane permeabilization? Trends Cell Biol. 2008;18:157–164. doi: 10.1016/j.tcb.2008.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Tournier C, et al. Requirement of JNK for stress-induced activation of the cytochrome c-mediated death pathway. Science. 2000;288:870–874. doi: 10.1126/science.288.5467.870. [DOI] [PubMed] [Google Scholar]
- 110.Lei K, et al. The Bax subfamily of Bcl2-related proteins is essential for apoptotic signal transduction by c-Jun NH2-terminal kinase. Mol Cell Biol. 2002;22:4929–4942. doi: 10.1128/MCB.22.13.4929-4942.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Lei K, Davis RJ. JNK phosphorylation of Bim-related members of the Bcl2 family induces Bax-dependent apoptosis. Proc Natl Acad Sci USA. 2003;100:2432–2437. doi: 10.1073/pnas.0438011100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Deng Y, Ren X, Yang L, Lin Y, Wu X. A JNK-dependent pathway is required for TNFα-induced apoptosis. Cell. 2003;115:61–70. doi: 10.1016/s0092-8674(03)00757-8. [DOI] [PubMed] [Google Scholar]
- 113.Putcha GV, et al. JNK-mediated BIM phosphorylation potentiates BAX-dependent apoptosis. Neuron. 2003;38:899–914. doi: 10.1016/s0896-6273(03)00355-6. [DOI] [PubMed] [Google Scholar]
- 114.Papadakis ES, et al. The regulation of Bax by c-Jun N-terminal protein kinase (JNK) is a prerequisite to the mitochondrial-induced apoptotic pathway. FEBS Lett. 2006;580:1320–1326. doi: 10.1016/j.febslet.2006.01.053. [DOI] [PubMed] [Google Scholar]
- 115.Hsu WH, et al. Berberine induces apoptosis in SW620 human colonic carcinoma cells through generation of reactive oxygen species and activation of JNK/p38 MAPK and FasL. Arch Toxicol. 2007;81:719–728. doi: 10.1007/s00204-006-0169-y. [DOI] [PubMed] [Google Scholar]
- 116.Koide N, et al. Lipopolysaccharide and interferon-γ enhance Fas-mediated cell death in mouse vascular endothelial cells via augmentation of Fas expression. Clin Exp Immunol. 2007;150:553–560. doi: 10.1111/j.1365-2249.2007.03499.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Guan QH, et al. Neuroprotection against ischemic brain injury by SP600125 via suppressing the extrinsic and intrinsic pathways of apoptosis. Brain Res. 2006;1092:36–46. doi: 10.1016/j.brainres.2006.03.086. [DOI] [PubMed] [Google Scholar]
- 118.Harwood FG, et al. Regulation of FasL by NF-κB and AP-1 in Fas-dependent thymineless death of human colon carcinoma cells. J Biol Chem. 2000;275:10023–10029. doi: 10.1074/jbc.275.14.10023. [DOI] [PubMed] [Google Scholar]
- 119.Takada E, et al. Interferon-β-induced activation of c-Jun NH2-terminal kinase mediates apoptosis through up-regulation of CD95 in CH31 B lymphoma cells. Exp Cell Res. 2005;304:518–530. doi: 10.1016/j.yexcr.2004.11.015. [DOI] [PubMed] [Google Scholar]
- 120.Xu XN, et al. Induction of Fas ligand expression by HIV involves the interaction of Nef with the T cell receptor zeta chain. J Exp Med. 1999;189:1489–1496. doi: 10.1084/jem.189.9.1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Yang SH, et al. Involvement of c-Jun N-terminal kinase in G2/M arrest and FasL-mediated apoptosis induced by a novel indoloquinoline derivative, IQDMA, in K562 cells. Leuk Res. 2007;31:1413–1420. doi: 10.1016/j.leukres.2007.02.014. [DOI] [PubMed] [Google Scholar]
- 122.Urano F, et al. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science. 2000;287:664–666. doi: 10.1126/science.287.5453.664. [DOI] [PubMed] [Google Scholar]
- 123.Liang SH, Zhang W, McGrath BC, Zhang P, Cavener DR. PERK (eIF2α kinase) is required to activate the stress-activated MAPKs and induce the expression of immediate-early genes upon disruption of ER calcium homoeostasis. Biochem J. 2006;393:201–209. doi: 10.1042/BJ20050374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Juo P, et al. FADD is required for multiple signaling events downstream of the receptor Fas. Cell Growth Differ. 1999;10:797–804. [PubMed] [Google Scholar]
- 125.Brahic M, Stroop WG, Baringer JR. Theiler’s virus persists in glial cells during demyelinating disease. Cell. 1981;26:123–128. doi: 10.1016/0092-8674(81)90040-4. [DOI] [PubMed] [Google Scholar]
- 126.Rodriguez M, Oleszak E, Leibowitz J. Theiler’s murine encephalomyelitis: a model of demyelination and persistence of virus. Crit Rev Immunol. 1987;7:325–365. [PubMed] [Google Scholar]
- 127.Theiler M. Spontaneous encephalomyelitis of mice — a new virus disease. Science. 1934;80:122. doi: 10.1126/science.80.2066.122-a. [DOI] [PubMed] [Google Scholar]
- 128.Griffin DE. Neuronal cell death in alphavirus encephalomyelitis. Curr Top Microbiol Immunol. 2005;289:57–77. doi: 10.1007/3-540-27320-4_3. [DOI] [PubMed] [Google Scholar]
- 129.Samuel MA, et al. PKR and RNase L contribute to protection against lethal West Nile Virus infection by controlling early viral spread in the periphery and replication in neurons. J Virol. 2006;80:7009–7019. doi: 10.1128/JVI.00489-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Zhang S, et al. A mutation in the envelope protein fusion loop attenuates mouse neuroinvasiveness of the NY99 strain of West Nile virus. Virology. 2006;353:35–40. doi: 10.1016/j.virol.2006.05.025. [DOI] [PubMed] [Google Scholar]
- 131.Tyler KL, et al. Linkage between reovirus-induced apoptosis and inhibition of cellular DNA synthesis: role of the S1 and M2 genes. J Virol. 1996;70:7984–7991. doi: 10.1128/jvi.70.11.7984-7991.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Huber SA. Coxsackievirus-induced myocarditis is dependent on distinct immunopathogenic responses in different strains of mice. Lab Invest. 1997;76:691–701. [PubMed] [Google Scholar]
- 133.Sherry B. Pathogenesis of reovirus myocarditis. Curr Top Microbiol Immunol. 1998;233:51–66. doi: 10.1007/978-3-642-72095-6_3. [DOI] [PubMed] [Google Scholar]
- 134.Sherry B, Li XY, Tyler KL, Cullen JM, Virgin HW. Lymphocytes protect against and are not required for reovirus-induced myocarditis. J Virol. 1993;67:6119–6124. doi: 10.1128/jvi.67.10.6119-6124.1993. This study shows that humoral immunity and cellular immunity are protective against, and not required for, reovirus-induced myocarditis and reveals that the potential to induce cardiac damage is independent of lymphocyte-based immunity. [DOI] [PMC free article] [PubMed] [Google Scholar]