National Academies Press: OpenBook

Live Variola Virus: Considerations for Continuing Research (2009)

Chapter: 3 Comparative Poxvirology

« Previous: 2 Overview of Smallpox and Its Surveillance and Control
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 27
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 28
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 29
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 30
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 31
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 32
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 33
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 34
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 35
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 36
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 37
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 38
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 39
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 40
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 41
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 42
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 43
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 44
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 45
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 46
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 47
Suggested Citation:"3 Comparative Poxvirology." Institute of Medicine. 2009. Live Variola Virus: Considerations for Continuing Research. Washington, DC: The National Academies Press. doi: 10.17226/12616.
×
Page 48

Below is the uncorrected machine-read text of this chapter, intended to provide our own search engines and external engines with highly rich, chapter-representative searchable text of each book. Because it is UNCORRECTED material, please consider the following text as a useful but insufficient proxy for the authoritative book pages.

3 Comparative Poxvirology A comprehensive discussion of variola virus must include comparison with other orthopoxviruses, as the similarities and differences between variola and members of its family help elucidate how variola causes disease and how it is modulated by subsequent host responses. In particular, variola virus’s unique adaptation to a single host (while other orthopoxviruses readily infect multiple mammalian species) and its ability to induce unusu- ally severe disease hint at a complex relationship between host and pathogen that may not be as easily explored using other orthopoxviruses. Variola virus also is not a single virus whose strains are identical, and the implications of this diversity could yield further information not just about the host, but also about mechanisms of antiviral therapeutic action, vaccine efficacy, and rapid diagnostic capabilities. Taking advantage of insights from comparative virology is particularly important because experiments with live variola virus must focus on critical questions. Knowledge of the related poxviruses can inform the design and refinement of experiments for which live variola virus is necessary. In the last decade, technological advances and the development of molecular techniques have made it possible to gain a deeper understanding of the general mechanisms involved in poxvirus replication, the host response, and the ways in which these pathogens have adapted to their hosts that is pertinent as background for considering the scientific needs for variola virus. POXVIRUS Taxonomy Viruses with shared characteristics are grouped into taxonomic cat- egories, including those of the Poxviridae family. Variola and the other 27

28 LIVE VARIOLA VIRUS members of Poxviridae are among the largest and most complex known viruses. Their genome is a single linear, double-stranded DNA molecule between 130 and 360 kilobase pairs (kbp) in size, and encodes on aver- age approximately 150 proteins. The family Poxviridae is subdivided into two subfamilies based on the restriction of their host range to vertebrates ( ­ Chordopoxvirinae) or invertebrates (Entomopoxvirinae), and these sub- families are further subdivided into genera of viruses that are genetically related and share aspects of nucleotide composition, host range, and morphology. Chordopoxvirinae consists of eight genera: Orthopoxvirus, Parapoxvirus, Avipoxvirus, Capripoxvirus, Leporipoxvirus, Suipoxvirus, Molluscipoxvirus, and Yatapoxvirus (Moss, 2007). The Orthopoxvirus genus includes many of the known poxviruses that naturally infect mam- mals, including vaccinia, the prototypical poxvirus, as well as two of the three poxviruses that have most commonly infected humans, variola and monkeypox virus; the third, molluscum contagiosum virus, is a member of Molluscipoxvirus. Other members of Chordopoxvirinae, including cow- pox virus (genus Orthopoxvirus) and orf virus (genus Parapoxvirus, most commonly found in sheep and goats) are less common disease agents of humans. Table 3-1 lists the poxviruses that affect humans, along with their reservoir hosts, other infected hosts, and geographic distribution. POXVirus structure Poxviruses were first visualized by electron microscopy (EM) in 1938 (Biel and Gelderblom, 1999). The large virus particles (approximately 240 nm × 300 nm for orthopoxviruses) appear brick-shaped under standard EM, with internal structures resembling a dumbbell-shaped core and two lateral bodies (see Figure 3-1). From the late 1940s through the end of the smallpox eradication era, EM was used to diagnose smallpox and to differen- tiate between variola and varicella zoster virus (VZV), which causes chicken­ pox (Biel and Gelderblom, 1999). The distinctive morphology observed by EM can still be a first step in diagnosis of poxviruses. During the outbreak of monkeypox in the United States in 2003, the first realization that the etiologic agent was an orthopoxvirus occurred when brick-shaped virions were visual- ized in a clinical specimen by EM (Reed et al., 2004). However, it is important to recognize that the orthopoxviruses that infect humans, including variola, vaccinia, and monkeypox, cannot be differentiated by traditional EM alone because the virion structure is highly conserved among orthopoxviruses. POXVirus genomics Advances in genomic sequencing and computational molecular biology have provided new insights into the relatedness and evolutionary history

COMPARATIVE POXVIROLOGY 29 TABLE 3-1  Poxviruses That Infect Humans Reservoir Other Infected Geographic Genus Virus Hosts Hosts Distribution Orthopoxvirus Cowpox Bank voles, Humans, cats, Europe, western long-tailed field cattle, zoo Africa mice animals Monkeypox Unknown, Humans, Western and likely rodents monkeys, zoo central Africa animals, prairie dogs Vaccinia ? Humans, ? rabbits, cattle, river buffalo Variola Humans None Eradicated (formerly worldwide) Parapoxvirus Bovine papular Cattle (beef) Humans Worldwide stomatitis Orf Sheep, goats Humans, Worldwide ruminants Pseudocowpox Cattle (dairy) Humans Worldwide Sealpox Seals Humans Worldwide Yatapoxvirus Tanapox Humans Eastern and central Africa Yabapox ? Primates Humans Western Africa Molluscipoxvirus Molluscum Humans None Worldwide contagiosum of poxviruses. This information helps place variola in its evolutionary con- text and points to significant genetic differences between variola and other orthopoxviruses. Poxvirus genes are usually nonoverlapping but closely spaced, and are arranged in blocks such that genes in the outer quadrants of the genome are transcribed toward the end of the genome in closest proximity, while genes in the central quadrants are transcribed toward the center of the genome. An analysis of 21 Poxviridae complete genome sequences in 2003 revealed a common set of 49 genes and an additional 41 genes shared by the chordopoxviruses (Upton et al., 2003). These families of shared genes encode proteins involved in basic functions such as DNA replication, transcription, and virion assembly, and are located toward the central region of the genome. In contrast, genes that are virus- or host- specific tend to be located toward the genome termini, and encode factors

30 LIVE VARIOLA VIRUS FIGURE 3-1  Electron microscopy of orthopoxvirus structure.

COMPARATIVE POXVIROLOGY 31 involved in subversion of host defenses and immune responses. These v ­ irulence-associated genes are believed to have been acquired more recently by the virus as it adapted to the host species (Lefkowitz et al., 2006). To date, all poxvirus genomes that have been studied have been found to have inverted terminal repeats (ITRs) at both ends of the genome (Garon et al., 1978; see Figure 3-2). Contemporary taxonomic approaches combine sequence-based phylo­ genetic and character trait analyses. This is the case for the poxviruses (Lefkowitz et al., 2006). Alignments of concatenated orthologous pro- tein sequences from the poxviruses have led to reconsideration of genus assignments for some members and genus interrelationships. Gene loss, fragmentation, and duplication all appear to have played important roles in poxvirus evolution, with subsequent restriction of virus host range. Complete genome sequences and comparative analysis have suggested the basis for differences in virulence among strains of the same orthopoxvirus species, as illustrated by monkeypox virus (MPXV). In recent years, it has become increasingly well recognized that west African strains of MPXV are less virulent than central African (Congo basin) strains, despite roughly similar degrees of host exposure in these two regions of the continent. The genome sequences of three west African MPXV strains were found to be more closely related to each other (0.01–0.07 percent difference) than to the previously sequenced strain from the Democratic Republic of the Congo (0.55–0.56 percent difference) (Chen et al., 2005). Of note, five putative virulence-associated genes contained significant deletions or fragmenta- Central conserved region (essential functions for viral replication) Variable left end Variable right end ITR ITR ~ 200,000 base pairs ~ 200 genes FIGURE 3-2  Internal terminal repeats at both ends of the poxvirus genome. Figure 3-2

32 LIVE VARIOLA VIRUS tion in the three west African strains as compared with the central African strain, including the gene encoding the MPXV ortholog of the vaccinia complement-binding protein (VCP-MPXV). On the basis of this sequence analysis and subsequent assessment of the expressed protein, VCP-MPXV is hypothesized to play a key role in the virulence of west African MPXV strains. More genomic information about variola virus isolates, coupled with clinical data on disease severity in the cases from which the virus was recovered, has the potential to elucidate factors influencing the virulence of variola. VirAL life cycle Variola virus, like all orthopoxviruses, replicates solely in the cyto- plasm of infected cells (see Figure 3-3). Most of what is known about the orthopoxvirus life cycle has been learned from extensive study of vaccinia virus (Moss, 2007), which is closely related to variola virus. Vaccinia has been the prototypic model for experimental analysis of the orthopox viral life cycle in vitro. Not only is analysis of the vaccinia life cycle relevant for understanding variola, but because smallpox vaccines are made from vac- cinia, this information can be used to design safer alternatives. There are several key points to be kept in mind as the viral life cycle is reviewed. First, progression through this life cycle involves the action of a significant number of virally encoded products, which are largely conserved throughout the Orthopoxvirus genus but do exhibit strain-specific varia- tions. Subtle features of the variola proteome are likely to be important in this regard. Second, there is increasing evidence—particularly for the pro- cess of transcription—that host proteins participate in the viral life cycle. This participation of host proteins is likely to be a significant determinant of species specificity and may contribute to the narrow host range of variola. Third, the progression of the viral life cycle occurs within the context of the cell, and there is growing evidence that intracellular structures (e.g., cytoskeleton, membranes) are vital for efficient viral replication. How viral proteins interact with these cellular structures is likely to be strain- and species-specific, and this is a relatively understudied area. Fourth, the viral proteins that mediate the progression of the viral life cycle represent the key pool of targets for antiviral therapy. Viral Entry There is good evidence that mature virions (MV) form weak attach- ments with glycosaminoglycans and laminins on the cell surface; various proteins within the virion membrane have been shown to be responsible for these interactions ������������������������������������������������������� (Chung et al., 1998; Carter et al., 2005; Chiu et al.,

core envelope Cytoplasm DNA Attachment Entry Uncoating DNA replication enzymes Transcription Factors WV Nucleus DNA packaging Concatamer resolution EV Wrapping Maturation MV IV Crescents Golgi bodies Virus Factory 33 FIGURE 3-3  Orthopoxvirus replication. Figure 3-3 REV.eps broadside

34 LIVE VARIOLA VIRUS 2007). It is not yet known whether other viral proteins interact strongly or ���������������������������������������������������������������������� specifically with proteinaceous receptors on the plasma membrane. If there are such receptors, however, they must be ubiquitous, since there have been no reports of cell lines that cannot support viral binding or entry. Once bound, virions can enter cells either by direct fusion of the viral and plasma membranes or by uptake of intact virus via macropinocytosis, with subse- quent release of the core from the endosomal compartment (Moss, 2006; Townsley et al., 2006; Mercer and Helenius, 2008) into the cytoplasm. The predominance of one mode of entry or the other differs with different viral strains and cell types, although the determinants of this variability have not been identified. A minority of the virions produced during poxvirus infection mature into enveloped or extracellular virions, also known as EV. EV are MV surrounded by an additional lipid bilayer carrying EV-specific surface pro- teins (Smith and Law, 2004). When EV attach to target cells, this exterior membrane is ruptured in a process known as ligand-dependent dissolution, and the MV found within then enter cells as described above (see also the discussion of morphogenesis and egress below). Gene Expression After delivery into the cytoplasm, the core of the poxvirus particle remains intact, and early gene expression begins (Broyles, 2003; Moss, 2007). Approximately 50 percent of the viral genes are expressed from the genome using the virally encoded and encapsidated transcriptional machinery. This machinery includes a multisubunit RNA polymerase, an RNA Pol accessory protein, an early transcription factor, a capping enzyme and cap modification enzyme, termination and transcript release proteins, and poly A polymerase. This phase in the viral life cycle yields a number of potential targets for antiviral drugs that may be conserved in variola and related poxviruses. The mature transcripts are released from the core and undergo translation on host cell polysomes. The initiation of two subsequent phases of poxvirus gene expression requires the prior onset of DNA replication (see below). Although the mechanism for this dependency is not known, DNA replication has been shown to have a cis-acting effect on the encapsidated viral genome that enables intermediate and late transcription to occur. Intermediate gene expression utilizes viral transcription factors that are expressed as early pro- teins, as well as some host proteins. Late gene expression also uses distinct viral transcription factors, as well as some host factors. Two features that distinguish these postreplicative phases of gene expression are the presence of 5′ polyA heads on the transcripts and imprecise termination of transcrip- tion; the latter feature leads to the presence of long overlapping transcripts,

COMPARATIVE POXVIROLOGY 35 which leads in turn to the presence of dsRNA. The availability of distinct classes of viral mRNA at distinct times after infection is also enhanced by the expression of viral decapping enzymes, which accelerate mRNA turn- over (Parrish and Moss, 2007; Parrish et al., 2007). The cytoplasmic replication cycle of poxviruses means that both transcrip­tion and translation occur in the cytoplasm and can be both tem- porally and physically coupled. Indeed, postreplicative transcription and translation occur within distinct areas of the cytoplasm known as viral factories (Condit, 2007; Katsafanas and Moss, 2007). This compartmen- talization serves to enhance viral protein expression as well as to diminish cellular protein expression, since the translational machinery is depleted in the cytoplasmic areas outside of the viral factories. Genome Replication and Maturation After early gene expression, the core appears to disassemble and the genome is released into the cytoplasm, where it undergoes replication in the viral factories. The proteins that make up the viral replication machinery, all of which are conserved in variola, are expressed early after infection. They include a catalytic DNA polymerase, a dimeric processivity factor (one subunit of which is an enzymatically active uracil DNA glycosylase), an ssDNA binding protein, and a primase/helicase (Moss and De Silva, 2006). A virally encoded protein kinase is also essential for replication; its primary role appears to be to phosphorylate and thus overcome the inhibi- tory action of a cytoplasmic DNA sensor. Other accessory proteins that may be dispensable in tissue culture but essential in vivo are also encoded by the viral genome. The mechanism by which replication initiates is still in question, and there is as yet no clear answer as to whether replication involves only leading strand synthesis or both leading and lagging strand synthesis. Replication does lead to the synthesis of tail/tail concatemers of the genome, which undergo subsequent resolution to mature monomeric genomes. This resolution is accomplished by a virally encoded Holliday junction resolvase and leads to reformation of the unusual telomeres of the viral genome, which are incompletely base-paired hairpins with an A+T content of >95 percent. Replication is robust and occurs from ~3 to 12 hours post-infection, leading to amounts of viral DNA that are estimated to approximate one-third the amount of the cellular DNA content. Morphogenesis and Egress As structural proteins and progeny become available, morphogenesis of nascent virions commences in the cytoplasm (Condit et al., 2006). ­Electron- dense areas of proteins destined for encapsidation are among the first

36 LIVE VARIOLA VIRUS hallmarks of morphogenesis. The appearance of membrane crescents (or cupules in three dimensions) at the periphery of these electron-dense areas is the first sign of membrane biogenesis. As these crescents enlarge and sur- round proteins destined to form the virion core, the curvature of a protein lattice that forms on their external face helps determine the size and shape of the immature virions (IV). Genome encapsidation is associated with the appearance of a nucleoid within these IV (forming IVN). Maturation of these IVN into infectious MV is accompanied by proteolytic processing of the major core proteins. This maturation is also accompanied by the transition of the oval IV to brick-shaped MV that have a characteristic dumbbell core. The majority of the MV remain within the cell as long as the cell remains intact. A minority, however, are transported to the Golgi ­apparatus (or endosomal compartment), where they become wrapped with two addi- tional lipid bilayers (forming wrapped virions, or WV). These outer enve- lopes contain a group of distinctive viral proteins that are not found in MV. WV traffic to the plasma membrane, where their outermost lipid bilayer fuses with the plasma membrane. This fusion leads to the exocytotic release of EV, which can disassociate from the plasma membrane and mediate dis- tal spread. Alternatively, some EV remain bound to the plasma membrane at the site of egress. The viral proteins that were delivered to the subjacent plasma membrane during fusion activate the formation of actin tails, which then propel the attached EV toward neighboring cells, facilitating efficient proximal spread of the virus. Host specificity and range As noted earlier, humans are the sole host for variola and molluscum contagiosum viruses, and the success of the WHO-led smallpox eradication program was achievable because variola virus has no animal reservoir. In general, poxvirus infections in vertebrate hosts show species ­ specificity; however, zoonotic infections do occasionally occur. The underlying mech- anism for host tropism, which is determined largely by host–pathogen interaction at many levels, is not well understood (McFadden, 2005). The reservoir hosts for monkeypox are rodents and squirrels, but this virus can occasionally cross the species barrier to infect monkeys and humans (Di Giulio and Eckburg, 2004). Poxviruses can bind to and enter a wide range of mammalian cells, but their success in replicating may vary. The ability of poxviruses to replicate and complete their viral life cycle in cells is dependent on many host-related factors, including cell type and species origin, cell cycle status, and intracel- lular signaling events leading to antiviral innate immunity and apoptosis. Identification of host range genes and elucidation of their interactions with

COMPARATIVE POXVIROLOGY 37 host proteins and signaling pathways have shed light on poxvirus host tropism. For example, vaccinia E3L and K3L are host range genes target- ing interferon (IFN)-inducible dsRNA-dependent protein kinase R (PKR) (Langland and Jacobs, 2002). E3L functions by sequestering dsRNA and preventing the activation of PKR, whereas K3L mimics eIF2α and acts as a pseudosubstrate for PKR, preventing phosphorylation of eIF2α and thus inhibition of protein synthesis. Suppression of PKR expression in a non- permissive cell line for an E3L knockout virus (∆E3L) restores viral protein synthesis and viral replication. Viral-induced apoptosis is blocked in PKR- deficient cells as well (Zhang et al., 2008). E3L also blocks the activation of another IFN-inducible protein, 2′–5′ oligoadenylate synthetase (2′–5′ OAS), and the subsequent activation of ribonuclease RNaseL (Rivas et al., 1998). The C-terminal dsRNA-binding domain of E3L is required for host range as well as pathogenesis. The N-terminal Z-DNA-binding domain of E3L is not required for host range, but it is required for pathogenesis in mice (Brandt and Jacobs, 2001). Vaccinia K1L gene is an ankyrin-repeat containing host range protein that is involved in inhibiting IκBα degradation, which prevents activation of the host defense mechanisms of the NFkB pathway. The importance of the capacity to block the host cell response is demonstrated by the arrest of replication of vaccinia mutant strains lacking K1L at the stage of inter­ mediate gene transcription in Chinese hamster ovary cells. The conservation of this function, and hence its likely importance to other orthopoxviruses, was shown by the rescue of this mutant by expression of another ankyrin- repeat containing host range protein from cowpox, CP77 (Ramsey-Ewing and Moss, 1996). Another ankyrin-repeat host range protein is M-T5 of myxoma virus, whose deletion leads to an inability to replicate in rabbit T lymphocytes and reduced virulence in European rabbits (Mossman et al., 1996). M-T5 is required for myxoma replication in certain human tumor cells (Sypula et al., 2004). Induction of IFN production has been shown to contribute to maintaining the species barrier for myxoma virus (Wang et al., 2004). C7L is another host range gene, which may be functionally equiva- lent to K1L. Infection with vaccinia mutant lacking both K1L and C7L is nonpermissive in human and murine cells (Perkus et al., 1990; Oguiura et al., 1993). C7L homologues are present in the genomes of almost all mammallian poxviruses. A recent study by Meng and colleagues (2008) demonstrated that vaccinia C7L homologue, myxoma M62R, or yaba-like disease virus 67R, when reconstructed in a vaccinia mutant lacking K1L and C7L, restored the vaccinia mutant’s ability to replicate in human and murine cells, possibly by suppressing PKR activation. MVA and NYVAC are two candidates for third-generation vaccines against smallpox that have alterations in immunomodulatory and host

38 LIVE VARIOLA VIRUS range genes. MVA is derived from more than 500 serial passages of the vac- cinia virus Ankara in chicken embryo fibroblasts (CEF); as a consequence, it lacks 31 kilobases of its genome (Antoine et al., 1998) (see Chapter 7). MVA does not replicate in most mammalian cells. The exceptions are BHK-21 cells and CEF. NYVAC is generated by deletion of 18 nonessential genes implicated in virulence or host range. It replicates in CEF, Vero cells, and BHK-21 cells (Tartaglia et al., 1992). MVA lacks K1L but retains C7L, whereas NYVAC lacks both K1L and C7L. In Hela cells, the MVA life cycle is blocked at a late stage of viral infection, probably at the assembly of immature virions (Sutter and Moss, 1992; Sancho et al., 2002). MVA with deletion of E3L still replicates in BHK-21 cells, but fails to replicate in CEF. Whereas MVA infection in Hela cells reveals a complete cascade of viral early, intermediate, and late gene transcription, MVA-∆E3L infection produces only early and intermediate transcripts. It is related to the induc- tion of 2′–5′ OAS/RNaseL and PKR (Ludwig et al., 2005, 2006). NYVAC induces apoptosis in Hela cells, which can be prevented through introduc- tion of the C7L gene (Nájera et al., 2006). Host–pathogen interactions Immune Modulation Poxviruses, including variola, encode many genes that are known or predicted to modulate host antiviral responses (Seet et al., 2003). These include secreted viral proteins that bind cytokines, chemokines, and com- plement proteins, as well as intracellular antagonists that block key signal- ing pathways leading to establishment of an antiviral state, apoptosis, or proinflammatory responses. Over the last decade, significant advances have been made in the understanding of poxvirus–host interactions and viral immune modulatory genes. Antiviral innate immunity is critical for the host to contain a viral infection initially and to activate the adaptive immune responses that result in viral clearance. Over the last several years, a number of viral-sensing pathways have been discovered in the host cell, including toll-like recep- tors (TLRs), retinoic acid-inducible gene I (RIG-I)-like receptors, nucleo- tide oligomerization domain (NOD)-like receptors (NLRs), and possibly c ­ ytosolic DNA sensors to detect viral nucleic acids and other components (Akira et al., 2006). The induction of type I IFN and proinflammatory cytokines and chemokines in various cell types in response to viral patho- gens, including dendritic cells, macrophages, epithelial cells, and fibroblasts, leads to the further recruitment of other immune cells and the development of adaptive immunity. Both functional type I and type II IFN systems are required for protection against vaccinia infection. For example, mice that

COMPARATIVE POXVIROLOGY 39 have targeted deletions in type I and type II IFN receptors show increased susceptibility to vaccinia infection (Müller et al., 1994). Although not studied for variola, IFNs can be predicted to be critical for the outcome of smallpox based on their fundamental contributions in other systemic viral infections. Through interactions with their hosts, poxviruses have evolved to p ­ ossess many mechanisms for antagonizing the production and actions of IFNs and proinflammatory cytokines and chemokines. Since the capacity of variola to block these responses is likely to be a key factor in its exceptional virulence for the human host, information about these mechanisms in other orthopoxviruses can be helpful in framing scientific questions about their contribution to the capacity of variola to overwhelm the human host. As an example of such mechanisms, many poxviruses encode IFNα/β-binding proteins and IFN-γ receptor homologs that dampen the effects of these molecules that are involved in countering viral replication and spread within the host (Upton et al., 1991; Symons et al., 1995). Specifically, the vaccinia virus (Western Reserve) B18R gene encodes for an IFNα/β-binding protein and is critical for virulence, as B18R knockout virus is attenuated in both murine intranasal and intracranial infection models (Symons et al., 1995). Similarly, ectromelia virus expresses functional homologs of IFN-γR and IFN-α/βR (Smith and Alcami, 2002). Recombinant ectromelia virus with deletion of the gene encoding type I IFN-binding protein was attenu- ated more than 107-fold compared with wild-type ectromelia virus (Xu et al., 2008). Variola virus also encodes homologs of IFN-γR and IFN-α/βR (Esposito et al., 2006; Li et al., 2007). Tumor necrosis factor (TNF) is a pleiotropic cytokine that mediates inflammation and apoptosis within the host. Many poxviruses, including variola virus, encode TNF receptor homologs (TNFRs), which block the host TNF signaling pathway. Whether they do so by binding TNF or by preventing the oligomerization of TNFR through their preligand assem- bly domains (PLADs) remains controversial (Alejo et al., 2006; Sedger et al., 2006). Myxoma virus lacking the TNF receptor homolog M-T2 has decreased virulence compared with wild-type myxoma (Upton et al., 1991). Whereas cowpox encodes four viral TNFRs, including CrmB (cytokine response modifier B), CrmC, CrmD, and CrmE, variola virus encodes one CrmB-like protein. In addition to its ability to bind to TNF, the variola CrmB-like protein is capable of binding to everal chemokines (Alejo et al., 2006). Poxviruses encode intracellular inhibitors that block caspase 1 ­activity, as well as IL-1β receptor homolog and IL-18-binding protein. IL-1β is another major cytokine mediating acute and chronic inflammation in response to infection and injury. It is produced through processing of its inactive precursor, proIL-1β, by caspase 1 to its active form, p17. Activa-

40 LIVE VARIOLA VIRUS tion of caspase 1 is dependent on activation of a multimolecular complex termed an inflammasome, which can be triggered by various stimuli, includ- ing cytosolic bacterial infections, ATP, and alum. IL-18 is another potent inflammatory cytokine released as a result of the activation of caspase I. Vaccinia virus WR B15 encodes an IL-1β-binding protein. Infection of mice with a B15R deletion mutant induced fever, whereas wild-type vaccinia infection suppressed fever, a host response controlled by the presence of IL-1β (Alcami and Smith, 1996). Both variola virus and monkeypox encode CrmA that inhibits caspase 1, an IL-1β receptor, and an IL-18-binding pro- tein (Seet et al., 2003). Myxoma virus attenuates inflammasome activation through an early viral gene, M13L, which interacts with a critical compo- nent of the inflammasome adapter protein, ASC (Johnston et al., 2005). Poxviruses utilize multiple strategies to evade the chemokine system by encoding chemokine-binding proteins, chemokine receptor homologs, and chemokine receptor antagonists. Chemokines are a large family of small, secreted proteins that mediate the recruitment of immune cells to the sites of injury or infection. Chemokines can be divided into several categories depending on the number and position of the highly conserved cysteine residues near the N-terminus of the protein. The two major chemokine subfamilies are CC and CXC, and the two minor subfamilies are C and CX3C. Chemokines perform their functions by binding to their cognate seven transmembrane G-protein-coupled receptors on the surface of cells (Mantovani et al., 2006). Some chemokines have direct antiviral activ- ity (Nakayama et al., 2006). Myxoma virus M-T7 encodes a low-affinity c ­ hemokine-binding protein that binds not only IFN‑γ, but also a wide range of CXC, CC, and C chemokines. Myxoma M-T1 and vaccinia B29R encode high-affinity ­chemokine-binding proteins that bind to CC chemokine, but not to C, CXC, or CX3C chemokines (Graham et al., 1997; Alcami et al., 1998). Variola virus and monkeypox virus also have high-affinity c ­ hemokine-­binding proteins (Alejo et al., 2006; Jones et al., 2008). The complement system is required for successful host defense against poxviruses. Moulton and colleagues (2008) recently showed that mice deficient in complement are more susceptible to ectromelia (mousepox) infection. Variola, monkeypox, ectromelia, and vaccinia viruses encode complement-binding proteins that block both the classical and alternative pathways of complement activation (Kotwal and Moss, 1988; Rosengard et al., 2002; Liszewski et al., 2006; Parker et al., 2008). Poxviruses also produce intracellular inhibitors to block the induction of type I IFN and proinflammatory cytokines and chemokines. Vaccinia virus A46R and A52R contain a Toll/IL-1 receptor (TIR) domain that can block the recruitment of adaptor molecules to IL-1R or TLR, preventing signaling following ligand–receptor interactions (Bowie et al., 2000). A52R has been found to block multiple TLRs through association with IRAK2

COMPARATIVE POXVIROLOGY 41 and TRAF6. Vaccinia mutant strain ∆A52R is attenuated in a murine intranasal model of infection (Harte et al., 2003). Recently, Vaccinia K7 was found to inhibit TBK1/IKKε-mediated IRF activation induced by TLR and non-TLR pathways (Schröder et al., 2008). Vaccinia E3L encodes a dsRNA-binding protein that can inhibit IRF3, IRF7, and NF-κB pathways, in addition to IFN-inducible genes (Smith et al., 2001; Xiang et al., 2002; Deng et al., 2006; Langland et al., 2006; Guerra et al., 2008) that would otherwise be activated following vaccinia infection. E3L also functions to subvert cytosolic RNA-sensing pathway mediated by MAVS and IRF3 in keratinocytes (DiPerna et al., 2004; Deng et al., 2008). Vaccinia N1L and K1L have been shown to block NF-κB signaling (Shisler and Jin, 2004). In addition, vaccinia VH1 gene encodes a phosphatase that blocks the activa- tion of IFN-induced activation of STAT-1 (Najarro et al., 2001). Apoptosis is an effective host mechanism for containing viral infections through programmed death of infected cells. Poxviruses have evolved to evade this defense mechanism through encoding of anti-apoptotic mol- ecules. The IFN-inducible PKR promotes apoptosis through inhibition of protein synthesis, which can be counteracted by vaccinia E3L and K3L (Chang et al., 1992); variola virus encodes E3L and K3L homologs. Myx- oma M11L, a virulence gene, encodes a mitochondria-targeted molecule that prevents apoptosis (Everett et al., 2000), while molluscum contagiosum virus encodes two genes, MC159 and MC160, to block the activation of initiator caspase, caspase 8 (Shisler and Moss, 2001). CrmA, a member of the serine protease inhibitor family, first identified as an inhibitor for IL-1β- converting enzyme (caspase 1), also inhibits caspase 8 and blocks apoptosis induced by various factors (Tewari and Dixit, 1995). Adaptive Immune Response Smallpox was eradicated prior to the development of modern quantita- tive cellular assays that measure virus-specific T cell numbers and function. Likewise, because of the conditions associated with smallpox outbreaks, monitoring humoral immune responses was usually difficult. Although neutralizing antibodies were measured, other more rapid and quantitative approaches, such as enzyme-linked immunosorbent assays (ELISAs), were not well established. As a result, knowledge about smallpox immunity is limited, and concepts of smallpox immunity are based on indirect information that is available about the kinetics, magnitude, and duration of ­orthopoxvirus- specific immunity derived from analysis of vaccinia-specific T cell and anti- body responses elicited by immunization with vaccinia virus. Since vaccinia replication is well controlled while variola is often life-­threatening, how well the host response to vaccinia mimics that induced by variola is not clear (see also Chapter 7). Nevertheless, these vaccinia responses define a protective

42 LIVE VARIOLA VIRUS cross-reactive response that is relevant for vaccine design and may suggest the characteristics of those responses that protected smallpox survivors from reinfection. Following vaccinia inoculation, antiviral T cell responses are difficult to detect at 1 week, but then rapidly expand and peak within approximately 2 weeks of infection (Miller et al., 2008). Antiviral antibody responses are slightly delayed in comparison with the T cell responses and generally peak within 2–3 weeks of inoculation. After the vaccinia lesion at the site of inoculation has resolved, antiviral T cell and antibody responses decline rapidly before reaching a more long-lived plateau phase in which immunological memory is maintained for decades. During this memory phase, vaccinia-specific CD4+ and CD8+ T cell responses decline slowly, with an estimated half-life of 8–15 years (Crotty et al., 2003; Hammarlund et al., 2003), whereas antiviral antibody responses are more stable, with an estimated half-life of 92 years (Amanna et al., 2007). No specific immunological correlate predicts protection against small- pox or any of the other orthopoxviruses. Animal model experiments indi- cate that the induction of immunity to proteins present in the EV form is essential for complete protection against challenge (Kaufman et al., 2008). Results of studies in nonhuman primates indicate that vaccine-mediated immunity against lethal monkeypox challenge is due to the presence of neu- tralizing antibodies (Edghill-Smith et al., 2005). Earlier studies in humans also demonstrated that smallpox patients who developed higher antibody responses during acute smallpox infection had a lower mortality rate than those who mounted weaker antibody responses (Slifka, 2004). However, these individuals could also have had a poor virus-specific T cell response. Administration of high-dose convalescent serum to smallpox patients appeared to protect against lethal smallpox infection in uncontrolled clini- cal studies (Slifka, 2004). It is likely that preexisting antiviral antibodies provide a first line of protection against infection with orthopoxviruses, whereas antiviral T cells, along with inhibitory antibodies, are needed if the virus overcomes this barrier and gains entry into the host. During primary infection, effective induction of both T cell and B cell responses is probably necessary to prevent the virus from causing fatal complications and to clear infectious virus from the host (Slifka, 2004). References Akira, S., S. Uematsu, and O. Takeuchi. 2006. Pathogen recognition and innate immunity. Cell 124:783–801. Alcami, A., and G. L. Smith. 1996. A mechanism for the inhibition of fever by a virus. Proceedings of the National Academy of Sciences of the United States of America 93:11029–11034.

COMPARATIVE POXVIROLOGY 43 Alcami, A., J. A. Symons, P. D. Collins, T. J. Williams, and G. L. Smith. 1998. Blockade of chemokine activity by a soluble chemokine binding protein from vaccinia virus. Journal of Immunology 160:624–633. Alejo, A., M. B. Ruiz-Argüello, Y. Ho, V. P. Smith, M. Saraiva, and A. Alcami. 2006. A c ­ hemokine-binding domain in the tumor necrosis factor receptor from variola (smallpox) virus. Proceedings of the National Academy of Sciences of the United States of America 103:5995–6000. Amanna, I. J., N. E. Carlson, and M. K. Slifka. 2007. Duration of humoral immunity to ­common viral and vaccine antigens. New England Journal of Medicine 357:1903–1915. Antoine, G., F. Scheiflinger, F. Dorner, and F. G. Falkner. 1998. The complete genomic ­sequence of the modified vaccinia Ankara strain: Comparison with other ­orthopoxviruses. ­Virology 244(2):365–396. Biel, S. S., and H. R. Gelderblom. 1999. Diagnostic electron microscopy is still a timely and rewarding method. Journal of Clinical Virology 13:105–119. Bowie, A., E. Kiss-Toth, J. A. Symons, G. L. Smith, S. K. Dower, and L. A. J. O’Neill. 2000. A46R and A52R from vaccinia virus are antagonists of host IL-1 and toll-like ­receptor signaling. Proceedings of the National Academy of Sciences of the United States of America 97:10162–10167. Brandt, T. A., and B. L. Jacobs. 2001. Both carboxyl- and amino-terminal domains of the vaccinia virus interferon resistance gene, E3L, are required for pathogenesis in a mouse model. Journal of Virology 75:850–856. Broyles, S. S. 2003. Vaccinia virus transcription. Journal of General Virology 84:2293–2303. Carter, G. C., M. Law, M. Hollinshead, and G. L. Smith. 2005. Entry of the vaccinia virus intracellular mature virion and its interactions with glycosaminoglycans. Journal of General Virology 86:1279–1290. Chang, H. W., J. C. Watson, and B. L. Jacobs. 1992. The E3L gene of vaccinia virus encodes an inhibitor of the interferon-induced, double-stranded RNA-dependent protein kinase. Proceedings of the National Academy of Sciences of the United States of America 89(11):4825–4829. Chen, N., G. Li, M. K. Liszewski, J. P. Atkinson, P. B. Jahrling, Z. Feng, J. Schriewer, C. Buck, C. Wang, E. J. Lefkowitz, J. J. Esposito, T. Harms, I. K. Damon, R. L. Roper, C. Upton, and R. M. Buller. 2005. Virulence differences between monkeypox virus isolates from West Africa and the Congo basin. Virology 340:46–63. Chiu, W. L., C. L. Lin, M. H. Yang, D. L. Tzou, and W. Chang. 2007. Vaccinia virus 4c (A26L) protein on intracellular mature virus binds to the extracellular cellular matrix laminin. Journal of Virology 81:2149–2157. Chung, C. S., J. C. Hsiao, Y. S. Chang, and W. Chang. 1998. A27L protein mediates vaccinia virus interaction with cell surface heparan sulfate. Journal of Virology 72:1577–1585. Condit, R. C. 2007. Vaccinia, Inc.—Probing the functional substructure of poxviral replication factories. Cell Host and Microbe 2:205–207. Condit, R. C., N. Moussatche, and P. Traktman. 2006. In a nutshell: Structure and assembly of the vaccinia virion. Advances in Virus Research 66:31–124. Crotty, S., P. Felgner, H. Davies, J. Glidewell, L. Villarreal, and R. Ahmed. 2003. Cutting edge: Long-term B cell memory in humans after smallpox vaccination. Journal of Immunology 171:4969–4973. Deng, L., P. Dai, W. Ding, R. D. Granstein, and S. Shuman. 2006. Vaccinia virus infection a ­ ttenuates innate immune responses and antigen presentation by epidermal dendritic cells. Journal of Virology 80:9977–9987.

44 LIVE VARIOLA VIRUS Deng, L., P. Dai, T. Parikh, H. Cao, V. Bhjoy, Q. Sun, Z. J. Chen, T. Merghoub, A. Houghton, and S. Shuman. 2008. Vaccinia virus subverts a mitochondrial antiviral signaling protein- dependent innate immune response in keratinocytes through its dsRNA binding protein E3. Journal of Virology 82:10735–10746. Di Giulio, D. B., and P. B. Eckburg. 2004. Human Monkeypox: An emerging zoonosis. The Lancet Infectious Diseases 4(1):15–25. DiPerna, G., J. Stack, A. G. Bowie, A. Boyd, G. Kotwal, A. Zhang, S. Arvikar, E. Latz, K. A. Fitzgerald, and W. L. Marshall. 2004. Poxvirus protein N1L targets the I-κB kinase com- plex, inhibits signaling to NF-κB by the tumor necrosis factor superfamily of receptors, and inhibits NF-κB and IRF3 signaling by toll-like receptors. The Journal of Biological Chemistry 279:36570–36578. Edghill-Smith, Y., H. Golding, J. Manischewitz, L. R. King, D. Scott, M. Bray, A. Nalca, J. W. Hooper, C. A. Whitehouse, J. E. Schmitz, K. A. Reimann, and G. Franchini. 2005. Smallpox vaccine-induced antibodies are necessary and sufficient for protection against monkeypox virus. Nature Medicine 11:740–747. Esposito, J. J., S. A. Sammons, A. M. Frace, J. D. Osborne, M. Olsen-Rasmussen, M. Zhang, D. Govil, I. K. Damon, R. Kline, M. Laker, Y. Li, G. L. Smith, H. Meyer, J. W. LeDuc, and R. M. Wohlhueter. 2006. Genome sequence diversity and clues to the evolution of variola (smallpox) virus. Science 313(5788)807–812. Everett, H., M. Barry, S. F. Lee, X. Sun, K. Granham, J. Stone, R. C. Bleakley, and G. M ­ cFadden. 2000. M11: A novel mitochondria-localized protein of myxoma virus that blocks apoptosis of infected leukocytes. Journal of Experimental Medicine 191(9): 1487-1498. Garon, C.F., Barbosa, E., and Moss B. 1978. Visualization of an inverted terminal repetition in vaccinia virus DNA. Proceedings of the National Academy of Sciences of the United States of America 75(10):4863-4867. Graham, K. A., A. S. Lalani, J. L. Macen, T. L. Ness, M. Barry, L. Y. Liu, A. Lucas, I. Clark-Lewis, R. W. Moyer, and G. McFadden. 1997. The T1/35kDa family of poxvirus- secreted proteins bind chemokines and modulate leukocyte influx into virus–infected tissues. Virology 229:12–24. Guerra, S., A. Cáceres, K.-P. Knobeloch, I. Horak, and M. Esteban. 2008. Vaccinia virus E3 protein prevents the antiviral action of ISG15. PLos Pathogen 4:e1000096. Hammarlund, E., M. W. Lewis, S. G. Hansen, L. I. Strelow, J. A. Nelson, G. J. Sexton, J. M. Hanifin, and M. K. Slifka. 2003. Duration of antiviral immunity after smallpox vaccina- tion. Nature Medicine 9:1131–1137. Harte, M. T., I. R. Haga, G. Maloney, P. Gray, P. C. Reading, N. W. Bartlett, G. L. Smith, A. Bowie, and L. A. J. O’Neill. 2003. The poxvirus protein A52R targets toll-like receptor signaling to suppress host defense. Journal of Experimental Medicine 197:343–351. Johnston, J. B., J. W. Barrett, SH Nazarian, M. Goodwin, D. Ricuttio, G. Wang, and G. M ­ cFadden. 2005. A poxvirus-encoded pyrin domain protein interacts with ASC-1 to inhibit host inflammatory and apoptotic responses to infection. Immunity 23:587–598. Jones, J. M., I. Messauodi, R. D. Estep, B. Orzechowska, and S.W. Wong. 2008. Monkeypox virus viral chemokine inhibitor (MPV vCCI), a potent inhibitor of rhesus macrophage inflammatory protein-1. Cytokine 43:220-228. Katsafanas, G. C., and B. Moss. 2007. Colocalization of transcription and translation within cytoplasmic poxvirus factories coordinates viral expression and subjugates host func- tions. Cell Host and Microbe 2:221–228.

COMPARATIVE POXVIROLOGY 45 Kaufman, D. R., J. Goudsmit, L. Holterman, B. A. Ewald, M. Denholtz, C. Devoy, A. Giri, L. E. Grandpre, J. M. Heraud, G. Franchini, M. S. Seaman, M. J. Havenga, and D. H. Barouch. 2008. Differential antigen requirements for protection against systemic and intranasal vaccinia virus challenges in mice. Journal of Virology 82(14):6829–6837. Kotwal, G. J., and B. Moss. 1988. Vaccinia virus encodes a secretory polypeptide structurally related to complement control proteins. Nature 335:176-178. Langland, J. O., and B. L. Jacobs. 2002. The role of the PKR-inhibitory genes, E3L and K3L, in determining vaccinia virus host range. Virology 299:133–141. Langland, J.������������������������������������������������������������������������������� O., J.������������������������������������������������������������������������ ������������������������������������������������������������������������������ C. Kash, V. Carter, M.������������������������������������������������� ����������������������������������������������������������������������� J. Thomas, M.����������������������������������� ������������������������������������������������ G. Katze, and B.������������������ ���������������������������������� L. Jacobs. 2006. ����������������� Suppression of proinflammatory signal transduction and gene expression by the dual nucleic acid binding domains of the vaccinia virus E3L proteins. Journal of Virology 80:10083–10095. Lefkowitz, E. J., C. Wang, and C. Upton. 2006. Poxviruses: Past, present and future. Virus Research 117:105–118. Li, Y., D. S. Carroll, S. N. Gardner, M. C. Walsh, E. A. Vitalis, and I. K. Damon. 2007. On the origin of smallpox: Correlating variola phylogenics with historical smallpox records. Proceedings of the National Academy of Sciences of the United States of America 104(40):15787–15792. Liszewski, M. K., Leung, M. K., Hauhart, R., Buller, R. M., Bertram, P., Wang, X., Rosengard, A. M., Kotwal, G. J., and J.P. Atkinson. ���������������������������������������������� 2006. Structure and regulatory profile of the monkeypox inhibitor of complement: comparison to homologs in vaccinia and variola and evidence for dimer formation. Journal of Immunology 176: 3725–3734. Ludwig, H., J. Mages, C. Staib, M. H. Lehmann, R. Lang, and G. Sutter. 2005. Role of viral factor E3L in modified vaccinia virus ankara infection of human HeLa cells: Regulation of the virus life cycle and identification of differentially expressed host genes. Journal of Virology 79(4):2584–2596. Ludwig, H., Y. Suezer, Z. Waibler, U. Kalinke, B. S. Schnierle, and G. Sutter. 2006. Double- stranded RNA-binding protein E3 controls translation of viral intermediate RNA, mark- ing an essential step in the life cycle of modified vaccinia virus Ankara. The Journal of General Virology 87(Pt. 5):1145–1155. Mantovani, A., R. Bonecchi, and M. Locati. 2006. Tuning inflammation and immunity by chemokine sequestration: Decoys and more. Nature Reviews Immunology 6:907–918. McFadden, G. 2005. Poxvirus tropism. Nature Reviews. Microbiology 3(3):201–213. Mercer, J., and A. Helenius. 2008. Vaccinia virus uses macropinocytosis and apoptotic mim- icry to enter host cells. Science 320:531–535. Meng, X., J. Chao, and Y. Xiang. 2008. Identification from diverse mammalian poxviruses of host-range regulatory genes functioning equivalently to vaccinia virus C7L. Virology 372(2):372–383. Miller, J. D., R. G. van der Most, R. S. Akondy, J. T. Glidewell, S. Albott, D. Masopust, K. Murali-Krishna, P. L. Mahar, S. Edupuganti, S. Lalor, S. Germon, C. Del Rio, M. J. M ­ ulligan, S. I. Staprans, J. D. Altman, M. B. Feinberg, and R. Ahmed. 2008. Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. ­ mmunity 28:710–722. I Moss, B. 2006. Poxvirus entry and membrane fusion. Virology 344:48–54. Moss, B. M. 2007. Poxviridae: The viruses and their replication. In Fields’ virology, 5th edi- tion, edited by B. N. Fields, D. M. Knipe, P. M. Howley, and D. E. Griffin. Philadelphia, PA: Lippincott Williams & Wilkins. Pp. 2906–2946.

46 LIVE VARIOLA VIRUS Moss, B., and F. S. De Silva. 2006. Poxvirus. In DNA replication and human disease, edited by M. DePamphilis. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press. Pp. 707–727. Mossman, K., S. F. Lee, M. Barry, L. Boshkov, and G. McFadden. 1996. Disruption of M-T5, a novel myxoma virus gene member of the poxvirus host range superfamily, results in dramatic attenuation of myxomatosis in infected European rabbits. Journal of Virology 70:4394–4410. Moulton, E. A., J. P. Atkinson, and R. M. Buller. 2008. Surviving mousepox infection requires the complement system. PLos Pathogen 4(12):e1000249. Müller, U., U. Steinhoff, L.������������������������������������������������������������������ ����������������������������������������������������������������� F. Reis, S. Hemmi, J. Pavlovic, R.������������������������������� ������������������������������ M. Zinkernagel, and M. Aguet. 1994. Functional role of type I and type II interferons in antiviral defense. Science 264:1918–1921. Najarro, P., P. Traktman, and J. A. Lewis. 2001. Vaccinia virus blocks gamma interferon s ­ ignal transduction: Viral VH1 phosphatase reverses Stat1 activation. Journal of ­Virology 75:3185–3196. Nájera, J. L., C. E. Gómez, E. Domingo-Gil, M. M. Gherardi, and M. Esteban. 2006. Cellular and biochemical differences between two attenuated poxvirus vaccine candidates (MVA and NYVAC) and role of the C7L gene. Journal of Virology 80(12):6033–6047. Nakayama, T., J. Shirane, K. Hieshima, M. Shibano, M. Watanabe, Z. Jin, D. Nagakubo, T. Saito, Y. Shimomura, and O. Yoshie. 2006. Novel antiviral activity of chemokines. Virology 350:484–492. Oguiura, N., D. Spehner, and R. Drillien. 1993. Detection of a protein encoded by the vaccinia virus C7L open reading frame and study of its effect on virus multiplication in different cell lines. The Journal of General Virology 74(Pt. 7):1409–1413. Parker, A. K., W. M. Yokoyama, J. A. Corbett, N. Chen, and R. M. Buller. 2008. Primary naive and interleukin-2-activated natural killer cells do not support efficient ectromelia virus replication. Journal of General Virology 89:751-759. Parrish, S., and B. Moss. 2007. Characterization of a second vaccinia virus mRNA-decapping enzyme conserved in poxviruses. Journal of Virology 81:12973–12978. Parrish, S., W. Resch, and B. Moss. 2007. Vaccinia virus D10 protein has mRNA decapping activity, providing a mechanism for control of host and viral gene expression. Proceedings of the National Academy of Sciences of the United States of America 104:2139–2144. Perkus, M. E., S. J. Goebel, S. W. Davis, G. P. Johnson, K. Limbach, E. K. Norton, and E. Paoletti. 1990. Vaccinia virus host range genes. Virology 179(1):276–286. Ramsey-Ewing, A. L., and B. Moss. 1996. Complementation of a vaccinia virus host-range K1L gene deletion by the non-homologous CP77 gene. Virology 222:75–86. Reed, K. D., J. W. Melski, M. B. Graham, R. L. Regnery, M. J. Sotir, M. V. Wegner, J. J. K ­ azmierczak, E. J. Stratman, Y. Li, J. A. Fairley, G. R. Swain, V. A. Olson, E. K. Sargent, S. C. Kehl, M. A. Frace, R. Kline, S. L. Foldy, J. P. Davis, and I. K. Damon. 2004. The detection of monkeypox in humans in the Western Hemisphere. New England Journal of Medicine 350:342–350. Rivas, C., J. Gil, Z. Mĕlková, M. Esteban, and M. Díaz-Guerra. 1998. Vaccinia virus E3L protein is an inhibitor of the interferon(i.f.n.)-induced 2-5A synthetase enzyme. Virology 1243:406–414. Rosengard, A.M., Liu, Y., Nie, Z., and R. Jimenez. 2002. Variola virus immune evasion d ­ esign: expression of a highly efficient inhibitor of human complement. Proceedings of the ­National Academy of Sciences of the United States of America 99:8808–8813. Sancho, M. C., S. Schleich, G. Griffiths, and J. Krijnse-Locker. 2002. The block in assembly ���������������������������� of modified vaccinia virus Ankara in HeLa cells reveals new insights into vaccinia virus morphogenesis. Journal of Virology 76(16):8318–8334.

COMPARATIVE POXVIROLOGY 47 Schröder, M., M. Barab, and A. G. Bowie. 2008. Viral targeting of DEAD box protein 3 r ­ eveals its role in TBK1/IKKε-mediated IRF activation. EMBO 27:2147–2157. Sedger, L. M., S. R. Osvath, X. M. Xu, G. Li, F. K. Chan, J. W. Barrett, and G. ­McFadden. 2006. Poxvirus tumor necrosis factor receptor (TNFR)-like T2 proteins contain a con- served preligand assembly domain that inhibits cellular TNFR1-induced cell death. Journal of Virology 80(18):9300–9309. Seet, B.��������������������������������������������������������������������������������������� �������������������������������������������������������������������������������������� T., J.�������������������������������������������������������������������������������� ������������������������������������������������������������������������������� B. Johnston, C.���������������������������������������������������������������� ��������������������������������������������������������������� R. Brunetti, J.������������������������������������������������ ����������������������������������������������� W. Barrett, H. Everett, C. Cameron, J. Sypula, S.������������������������������������������������������������������������������ ����������������������������������������������������������������������������� H. Nazarian, A. Lucas, and G. McFadden. 2003. Poxviruses and immune evasion. ­ nnual Review of Immunology 21:377–423. A Shisler, J. L., and X. L. Jin. 2004. The vaccinia virus K1L gene product inhibits host NF-κB activation by preventing IκBα degradation. Journal of Virology 78:3553–3560. Shisler, J.L., and B. Moss. 2001. Molluscum contagiosum virus inhibitors of apoptosis: The MC159 v-FLIP protein blocks Fas-induced activation of procaspases and degradation of the related MC160 protein. Virology 282:14–25. Slifka, M. K. 2004. Immunological memory to viral infection. Current Opinion in ­Immunology 16:443–450. Smith, E.��������������������������������������������������������������������������������� �������������������������������������������������������������������������������� J., I. Marié, A. Prakash, A. García-Sastre, and D.������������������������������ ����������������������������� E. Levy. 2001. IRF3 and IRF7 phosphorylation in virus-infected cells does not require double-stranded RNA-dependent protein kinase R or Ikappa B kinase but is blocked by Vaccinia virus E3L protein. Journal of Biological Chemistry 276:8951–8957. Smith, G. L. and Law, M. 2004. The Exit of Vaccinia Virus From Infected Cells. Virus ­ esearch 106(2): 189–197. R Smith, V. P., and A. Alcami. 2002. Inhibition of interferons by ectromelia virus. Journal of Virology 76:1124–1134. Sutter, G., and B. Moss. 1992. Nonreplicating vaccinia vector efficiently expresses recombinant genes. Proceedings of the National Academy of Sciences of the United States of America 89(22):10847–10851. Symons, J. A., A. Alcami, and G. L. Smith. 1995. Vaccinai virus encodes a soluble type I inter- feron receptor of novel structure and broad species specificity. Cell 81:551–560. Sypula, J., F. Wang, Y. Ma, J. Bell, and G. McFadden. 2004. Myxoma virus tropism in human tumor cells. Gene Therapy & Molecular Biology 8:103–114. Tartaglia, J., M. E. Perkus, J. Taylor, E. K. Norton, J. C. Audonnet, W. I. Cox, S. W. Davis, J. van der Hoeven, B. Meignier, and M. Riviere. 1992. NYVAC: A highly attenuated strain of vaccinia virus. Virology 188(1):217–232. Tewari, M., and V. M. Dixit. 1995. Fas- and tumor necrosis factor-induced apoptosis is inhibited by the poxvirus crmA gene product. Journal of Biological Chemistry 270:3255–3260. Townsley, A. C., A. S. Weisberg, T. R. Wagenaar, and B. Moss. 2006. Vaccinia virus entry into cells via a low-pH-dependent endosomal pathway. Journal of Virology 80:8899–8908. Upton, C., J. L. Macen, M. Schreiber, and G. McFadden. 1991. Myxoma virus expresses a secreted protein with homology to the tumor necrosis factor receptor gene family that contributes a viral virulence. Virology 184:370–382. Upton, C., S. Slack, A. L. Hunter, A. Ehlers, and R. L. Roper. 2003. Poxvirus orthologous clusters: Toward defining the minimum essential poxvirus genome. Journal of Virology 77:7590–7600. Wang, F., Y. Ma, J. W. Barrett, X. Gao, J. Loh, E. Barton, H. W. Virgin, and G. McFadden. 2004. Disruption of ERK-dependent type I interferon induction breaks the myxoma virus species barrier. Nature Immunology 5:1266–1274. Xiang, Y., R.���������������������������������������������������������������������������������� C. Condit, S. Vijaysri, B.������������������������������������������������������� ��������������������������������������������������������������������������������� L. Jacobs, B.����������������������������������������� ������������������������������������������������������ R. Williams, and R.��������������������� ���������������������������������������� H. Silverman. 2002. �������������������� Blockade of interferon induction and action by the E3L double-stranded RNA binding proteins of vaccinia virus. Journal of Virology 76:5251–5259.

48 LIVE VARIOLA VIRUS Xu, R.-H., M. Cohen, Y. Tang, E. Lazear, J. C. Whitbeck, R J. Eisenberg, G. H. Cohen, and L. J. Sigal. 2008. The orthopoxvirus type I IFN binding protein is essential for virulence and an effective target for vaccination. Journal of Experimental Medicine 205:981–992. Zhang, P., B. L. Jacobs, and C. E. Samuel. 2008. Loss of protein kinase PKR expression in human Hela cells complements the vaccinia virus E3L deletion mutant phenotype by restoration of viral protein synthesis. Journal of Virology 82:840–848.

Next: 4 Animal Models Using Variola and Other Orthopoxviruses »
Live Variola Virus: Considerations for Continuing Research Get This Book
×
 Live Variola Virus: Considerations for Continuing Research
Buy Paperback | $54.00
MyNAP members save 10% online.
Login or Register to save!
Download Free PDF

Smallpox was a devastating disease that decimated human populations for centuries, and its eradication in 1980 was a monumental achievement for the global health community. Since then the remaining known strains of its causative agent, variola virus, have been contained in two World Health Organization (WHO)-approved repositories.

In 1999, the World Health Assembly (WHA) debated the issue of destroying these remaining strains. Arguments were presented on the need to retain the live virus for use in additional important research, and the decision to destroy the virus was deferred until this research could be completed. In that same year, the Institute of Medicine (IOM) convened a consensus committee to explore scientific needs for the live virus.

In the ten years since the first IOM report, the scientific, political, and regulatory environments have changed. In this new climate, the IOM was once again tasked to consider scientific needs for live variola virus. The committee evaluated the scientific need for live variola virus in four areas: development of therapeutics, development of vaccines, genomic analysis, and discovery research.

READ FREE ONLINE

  1. ×

    Welcome to OpenBook!

    You're looking at OpenBook, NAP.edu's online reading room since 1999. Based on feedback from you, our users, we've made some improvements that make it easier than ever to read thousands of publications on our website.

    Do you want to take a quick tour of the OpenBook's features?

    No Thanks Take a Tour »
  2. ×

    Show this book's table of contents, where you can jump to any chapter by name.

    « Back Next »
  3. ×

    ...or use these buttons to go back to the previous chapter or skip to the next one.

    « Back Next »
  4. ×

    Jump up to the previous page or down to the next one. Also, you can type in a page number and press Enter to go directly to that page in the book.

    « Back Next »
  5. ×

    To search the entire text of this book, type in your search term here and press Enter.

    « Back Next »
  6. ×

    Share a link to this book page on your preferred social network or via email.

    « Back Next »
  7. ×

    View our suggested citation for this chapter.

    « Back Next »
  8. ×

    Ready to take your reading offline? Click here to buy this book in print or download it as a free PDF, if available.

    « Back Next »
Stay Connected!