Structural proteins of baculovirus occlusion bodies and virions 您所在的位置:网站首页 ac142 Structural proteins of baculovirus occlusion bodies and virions

Structural proteins of baculovirus occlusion bodies and virions

2024-07-10 11:51| 来源: 网络整理| 查看: 265

Infectious baculoviruses can be generated from the transfection of naked viral DNA into susceptible insect cells indicating that the transcription of early genes required for initiating the infection is accomplished via the host cell RNA polymerase. Therefore, viral structural proteins, although necessary for protecting the viral genome and initiating infection, very likely are not required for functions such as transcription or DNA replication. Below is an overview of the proteins that have been associated with major baculovirus virion structures. These include occlusion bodies, budded virus (BV) and occlusion derived virus (ODV) envelopes, and proteins that are components of nucleocapsids. Many of the latter category are shared between BV and ODV (1).

Occlusion body evolution

In temperate climates and in many tropical areas where there are wet and dry seasons, insect populations are transitory and expand dramatically during warm, moist periods, and then collapse with the onset of colder temperatures or drought when food sources are reduced or eliminated. In addition, even during seasons optimal for insect growth and reproduction, their populations are normally limited by predators, pathogens, normal temperature cycles, and food sources. However, under certain circumstances, insect populations can expand dramatically when a combination of conditions greatly increases their food supply, facilitates high levels of reproduction, or eliminates predators and pathogens. These cycles of population expansion are well documented for forest insects and may be separated by long periods of time. For example, the Douglas fir tussock moth, a pest of forests in western North America, has cycles of 7 to 10 years, and other insect epizootics are separated by 5 to 40 years (reviewed in (2)). Viruses often play a major role in causing the collapse of these large insect populations.

A major consequence of the cyclic nature of insect populations is that their pathogens are left without hosts either seasonally, or for much longer periods of time. Viruses, in general, have developed several methods to insure their survival until their hosts reappear. If their hosts are seasonal, such as those of insect pathogens, viruses could be present in the eggs or pupae of over wintering insects, or they might persist in alternate hosts, or some might remain stable outside their hosts. Although there is some evidence for baculoviral persistence within host insects (see Chapter 3), stability outside their hosts mediated by their presence within occlusion bodies appears to be a common feature of all baculoviruses and some nudiviruses. By immobilizing the virus within the crystalline protein lattice of the occlusion body, an environment is provided that allows virions to remain viable indefinitely as long as they are protected from extremes of heat and from UV light. In addition, the stability provided by occlusion may be of such a selective advantage that it has apparently been incorporated into the life cycle of three different types of insect viruses; in Reoviridae (cypoviruses, double-stranded RNA viruses with segmented genomes), the Poxviridae (entomopox viruses-dsDNA viruses with cytoplasmic replication), and in the Baculoviridae/Nudiviridae. No primary amino acid sequence relatedness is evident between these different categories of occlusion body proteins and the primary sequence of the occlusion body protein of a dipteran baculovirus appears to be unrelated to other baculovirus polyhedrins (3), and the polyhedrins from some cypoviruses also appear to be only distantly related to each other (4). Similarly, the occlusion body protein of a nudivirus of the pink shrimp, Penaeus monodon, did not appear to be related to baculovirus polyhedrin (5), even though sequence data from other predicted proteins indicated the virus was related to the baculovirus lineage (6). The structure of both cypovirus and baculovirus polyhedrins have been determined, and they appear to be unrelated and, therefore, may be examples of convergent evolution (7, 8), although one report suggests that they may be distantly related (4).

Polyhedra are thought to stabilize baculovirus virions, and in one example it was estimated that only about 0.16% of OpMNPV occlusion bodies remained infectious one year after an epizootic. However, considering that over 3 x 1015 occlusion bodies were estimated to have been produced per hectare during the outbreak, this would still be a substantial number of infectious viruses (reviewed in (2)).

Occlusion body proteins

In addition to virions, occlusion bodies are composed of the matrix that occludes the virions and an outer membrane-like structure on the surface. Several other proteins are also associated with polyhedra (Table 1). The major protein forming occlusion bodies is polyhedrin/granulin.

Table 1

Proteins Associated with Baculovirus Occlusion Bodies

View in own window

AcMNPV orf # and nameDistribution in the BaculoviridaeEffect of Deletion2Ac8 PolyhedrinAll1ViableAc131 Polyhedron envelope/CalyxAll except CuniNPVViableEnhancinA few NPVs and GVsViableAc137 p10Group I/II; some GVsViableAlkaline proteasesNon baculovirus contaminants1

CuniNPV polyhedrin is unrelated to that of other baculoviruses.

2 For details see Chapter 12

Polyhedrin/Granulin. Polyhedrin and granulin are closely related and are the major structural components of occlusion bodies (polyhedrin in NPVs and granulin in GVs). They initially were challenging to characterize because the occlusion bodies are naturally contaminated with a protease (see below) that caused degradation of the protein upon dissolution of polyhedra. However, once it was found that the proteases could be heat inactivated, polyhedrins from several viruses were characterized and were found to contain about 250 aa (30 kDa) and are one of the most conserved baculovirus proteins. They form a crystalline cubic lattice that is interrupted by and surrounds embedded virions (Figure 1). Orthologs of polyhedrin/granulin are found in all baculovirus genomes, except for that of the dipteran virus (CuniNPV). Surprisingly, CuniNPV has an occlusion body protein that appears to be unrelated in primary amino acid sequence to the polyhedrin of other baculoviruses and is about three times as large (3, 9). The polyhedrin of an NPV pathogenic for pink shrimp also appears to be unrelated to that from other baculoviruses in its primary sequence (5). However, it was reported that it did react with antiserum to AcMNPV and TnGV polyhedrin even though at 50 kDa it appears to be twice as large as the insect baculovirus polyhedrins (10).

Figure 1.

Sections of portions of polyhedra showing crystalline structure. Top panel is from an NPV of Pseudohazis eglanterina (western sheep moth). Bottom panel is a higher magnification from an NPV of Nepytia freemani (false hemlock looper). Measurements are (more...)

Despite forming a natural crystal in nature, determining the structure of polyhedrin by crystallography was unsuccessful because it was not possible to recrystallize it from solution. However, advances in crystallography, plus the observation that a single amino acid change (G25D in AcMNPV polyhedrin) resulted in larger than normal crystals (11) allowed the determination of the structure of polyhedrins from two different Alphabaculoviruses (AcMNPV and Wiseana sp NPV) using occlusion bodies produced by viral infection (8) (12). A structure for a granulin from CpGV has also been determined (13). The 30 kDa polyhedrin subunits form trimers that are then arranged into dodecamers (four trimers) via disulfide bonds. This structure interlocks with another dodecamer to form the cubic-shaped unit cell of the crystal (Figure 2). Hydrophobic and salt bridge interactions between the cubes likely form the linkages at the crystal interfaces that are disrupted by the alkaline pH of the insect midgut (see below). Occlusion of virion bundles might displace up to 20,000 polyhedrin subunits and could destabilize the crystals. This possibly led to the evolution of the envelope structure that is thought to stabilize these structures (12). The presence of disulfide bonds may have prevented the attempts to crystallize purified polyhedrin.

Figure 2.

The assembly of polyhedrin into polyhedra. Polyhedrin trimers are depicted as simplified cubic blocks. To clarify interpretation, the edges of the unit cell are shown in gold and a cyan tetrahedron symbolizes the cell center. Within a unit cell, disulphide-linked (more...)

The calyx/polyhedron envelope (PE). The calyx/polyhedron envelope is an electron-dense structure that forms a smooth, seamless surface that surrounds polyhedra. The function of the calyx/PE appears to be to seal the surface of polyhedra and to enhance their stability. In the laboratory when polyhedra are subjected to alkaline treatment, the crystalline lattice is dissolved, but the polyhedron envelope remains as a bag-like structure in which many virions are trapped (Figure 3). Although the calyx/polyhedron envelope was originally found to be composed of carbohydrate (14), a phosphorylated protein component was subsequently identified (15). This protein appears to be an integral component of the calyx/PE (16). Homologs of the PE protein (Ac131) are found in the genomes of all lepidopteran NPVs. It is likely that when polyhedra are ingested by susceptible insects, they are dismantled by a combination of the alkaline pH of the insect midgut and proteinases that are present in the midgut and associated with polyhedra. This combination would likely contribute to the disruption of the polyhedron and polyhedron envelope to facilitate virion release. The PE protein is associated with p10 fibrillar structures (Figure 4), and p10 appears to be required for the proper assembly of the polyhedron envelope (17-20). Polyhedra from viruses with either the p10 or PE gene deleted have a similar appearance; they have a rough pitted surface and the PE appears to be fragmented or absent (Figure 5). In addition, in some viral genomes, genes are present that appear to be fusions of both PE and p10 protein domains, further suggesting a fundamental relationship between these two proteins (see below). A partial structure for the PEP of CpGV has been determined (21).

Figure 3.

Two adjacent dissolved polyhedra showing rod-shaped virions trapped by the collapsed polyhedron envelope. Photo by K. Hughes.

Figure 4.

Fibrous p10-containing material aligned with the calyx/polyhedron envelope. Photo courtesy of G. Williams. From (239), with permission.

Figure5.

Polyhedra from OpMNPV with the polyhedron envelope protein and p10 genes deleted. From (16), with permission.

Polyhedron-associated proteins

Ac68 may be involved in polyhedron morphogenesis. Homologs of Ac68 appear to be present in all baculoviruses. When Ac68 was deleted, no major effects were detected other than a longer lethal time in larvae (22). Similar results were obtained when the ortholog in BmNPV (Bm56) was deleted from a bacmid. However, the polyhedra produced by the mutant BmNPV bacmid were abnormal and lacked virions, suggesting that Bm56 may be involved in polyhedron morphogenesis (23).

P10 (Ac137). Although p10 does not appear to be a major occlusion body protein, it colocalizes with the PE protein and appears to be required for the proper formation of the polyhedron envelope. When p10 is phosphorylated, it becomes associated with microtubules (24) (25). This could be related to the structures it forms, including microtubule-associated filaments and tube-like structures that surround the nuclei of infected cells (26, 27). Deletion of P10 results in polyhedra that resemble those produced by mutants lacking the calyx/polyhedron envelope protein; they are fragile, have a rough surface showing cavities where virions have apparently become dislodged, and often show an incomplete calyx/polyhedron envelope (16, 28, 29) (see Figure 5). Serial block-face scanning electron microscopy (SBFSEM) has been used for the 3-D characterization of p10 structures in AcMNPV infected Tn-368 cells (Figure 6) (30). It forms large vermiform structures that surround nuclei and also structures within nuclei that are associated with ‘electron dense spacers’ which appear to contain Ac131, the polyhedron envelope/calyx associated protein (19). In cultured cells p10 facilitates occlusion body release by nuclear lysis (31) (30) and an increase in infectious virus (28). Homologs of p10 are found in the genomes of all Group I/II NPVs and most GVs, in some instances in multiple copies, e.g., PlxyGV has 3 copies (32). Their phylogeny is complicated somewhat by the fact that some of the GV genes appear to consist of a combination of p10 and polyhedron envelope protein domains (33, 34). A p10 homolog has also been characterized in an entomopox virus (35).

Figure 6.

Whole cell 3D reconstruction of P10 structures in AcMNPV infected Tn-368 cells (30) using serial block-face scanning electron microscopy Left image: 72 hpi with the following structures labeled: virogenic stroma (VS); P10 (nuclear, n; cytoplasmic, c); (more...)

Viral enhancing factors: enhancin. Enhancins are a class of metalloproteinases (36) that are encoded by a few lepidopteran NPVs (e.g., Ld-, Cf-, and MacoNPV) and GVs (e.g., Ag-, As-, Tn-, XcGVs), but not AcMNPV. In one study of TnGV, enhancin was estimated to comprise up to 5% of the mass of occlusion bodies (37). In another virus, LdMNPV, it was found to be associated with ODV envelopes (38). Enhancin genes are often present in multiple copies, e.g., the XecnGV genome has four copies (39). In LdMNPV which encodes two enhancins, deletion of either results in a 2- to 3-fold reduction in potency, whereas deletion of both caused a 12-fold reduction (40). Enhancin is thought to facilitate baculovirus infection by digesting the peritrophic matrix (PM) (see Figure 2, Chapter 3). The PM forms a barrier in insect guts that prevents the ready access of pathogens to the epithelial cells. The PM is rich in chitin and intestinal mucin protein, and enhancins appear to target the degradation of the mucin, thereby facilitating access of virions to the underlying cells (41). Enhancins show sequence homology with high levels of significance (e.g., 3e-29) to predicted proteins of a number of pathogenic bacteria, e.g., Clostridium botulinum, and a variety of Bacillus (e.g., B. anthracis) and Yersinia (e.g., Y. pestis) species. To investigate their function, enhancin homologs from B. cereus, Y. pseudotuberculosis, or TnGV were cloned into a construct of AcMNPV that yielded occluded viruses. Although the LD50 of these constructs was found to be about half of wt, only the construct expressing the TnGV enhancin caused a reduction in survival time. In addition, the bacterial enhancins failed to degrade insect intestinal mucin. It was suggested that the bacterial enhancins may have evolved an activity distinct from their viral homologs (42).

Proteinases. The initial research on occlusion body structure was hindered by the presence of proteinases that degraded the proteins under investigation. These preparations were derived from insect carcasses in various states of disintegration and decay. In hindsight, it is not surprising that these preparations showed proteinase activity. The finding that the proteinases could be heat inactivated (43), led to the ability to purify and eventually sequence a number of polyhedrin proteins before the advent of DNA sequencing technology (44, 45). Subsequently, it was found that the proteinases associated with occlusion bodies had properties similar to enzymes associated with the insect gut (46) and that polyhedra produced in cell culture lacked associated proteinases (47-49). Although many baculoviruses encode a cathepsin-like proteinase, in AcMNPV (Ac127) it was most active under acidic (pH 5) conditions (50). Therefore, the proteinases associated with occlusion bodies are likely a combination of enzymes derived from bacteria, the insect gut, and the virus.

Baculovirus virions: The envelope proteins

For AcMNPV and other relatively well-characterized lepidopteran NPVs, there are two types of virions produced during the virus infection; in insects the infection is initiated by occlusion-derived virus (ODV) that are released into the insect midgut upon dissolution of the occlusion bodies. ODV initiate infection in the midgut epithelium, and the virus propagated in these cells are budded virus (BV) that exit the cells in the direction of the basement membrane and spread the infection throughout the insect. Late in the infection, virions become occluded within the nuclei of infected cells and are released into the environment upon the death and disintegration of the insect. The major difference between BV and ODV is the origin of their envelopes. BVs derive their envelopes as they bud through the host cell plasma membrane that has been modified by viral proteins. In contrast, ODV obtain their envelope in the nucleus and it may be derived from nuclear membranes that are modified with a number of viral proteins. Whereas viral contributions to the BV envelope may be limited to one or two proteins, ODV envelopes are very complex. They appear to contain a number of virally encoded proteins, and in some instances, it is difficult to separate them from capsid proteins.

Cell entry: Baculovirus envelope fusion proteins

Many viruses are surrounded by a lipid envelope and enter cells either by fusion with the cell surface membrane or an endosomal membrane after phagocytosis by a cell. The merging of the membranes is activated by fusion proteins. There are three classes of viral envelope fusion proteins, I, II, and III (reviewed in (51) and baculoviruses employ categories I and III. Class I fusion proteins are activated by cleavage of a trimeric single-chain precursor protein. Cleavage is usually activated by exposure to low pH. Examples are the influenza hemagglutinin (HA), the HIV gp41, and the baculovirus F fusion proteins. Class II fusion proteins are found in small icosahedral viruses such as flaviviruses and alphaviruses and are present in a 1:1 complex with a chaperone protein that inhibits fusion. The inhibition is removed by cleavage of the chaperone. Class III fusion proteins are activated by low pH, but are not cleaved, and are found in rhabdoviruses (e.g. vesicular stomatitis virus - VSV-G), herpes viruses (gB), and some baculoviruses and orthomyxovirues (gp64).

Envelope proteins of budded virions

AcMNPV the most well studied baculovirus may enter cells by two different processes; direct fusion, and by receptor mediated endocytosis (52). Probably the most well characterized baculovirus structural protein is the envelope fusion protein of Group I baculoviruses, GP64 (Class III), because a relatively simple assay for its fusion activity was developed early in its investigation (53). Because of early studies elucidating the importance of this protein for AcMNPV BV infectivity (54, 55), it was unexpected when genome sequence analyses of additional baculoviruses revealed that many of them lacked homologs of the gp64 gene (56, 57). Furthermore, it was determined that these viruses use a different fusion protein called F (Class I) and that homologs of F are retained in gp64-containing viruses (Figure 7). These F homologs in viruses encoding GP64 are inactive as fusion proteins, suggesting that gp64 was obtained by a baculovirus and displaced the fusion function of the F protein, but the F gene was retained. These proteins are described below.

Figure 7.

Distribution of envelope fusion proteins. Group I have homologs of both GP64 and F, but F is not a fusion protein. Group II, GVs and dipteran viruses have homologs of F, whereas the hymenopteran viruses have homologs of neither GP64 nor F. As noted in (more...)

GP64 (Ac128) is present in all Group I NPVs, a single GV, and certain lineages of orthomyxoviruses. GP64 is a fatty acid acylated glycoprotein (58) and a low pH activated envelope fusion protein (59-61) that is one of the three most abundant proteins, along with vp39 and p6.9, found associated with AcMNPV budded virions (62). The presence of the gp64 gene is one of the major distinguishing features of the Group I alphabaculoviruses. Deletion of AcMNPV gp64 results in viruses that replicate in a single cell, but cannot bud out and infect surrounding cells (55, 63). It was originally thought that all Group I viruses use GP64 for the entry of BV into cells, whereas all other baculoviruses lack a gp64 homolog and appear to use the F (Ac23 homolog) protein as their envelope fusion protein except for hymenopteran NPVs, which lack both genes (Figure 7). However, it was subsequently found that Diatraea saccharalis (the sugarcane borer) granulovirus, DisaGV, encodes both F and gp64 fusion protein orthologs (64). This was the first example outside of the Group I nucleopolyhedroviruses where a baculovirus possesses gp64 in addition to the F protein. It is thought that gp64 was incorporated into the Group I viruses in a recombination event (Figure 8) that may have involved up to 11 other additional genes specific to Group I NPVs (Chapter 1, Table 3). After incorporation, it is thought that gp64 displaced the fusion function of the F protein which has been retained but is not an essential gene (65). An AcMNPV bacmid deleted for gp64 and pseudotyped with DisaGV gp64 was infectious although at a lower level than wt AcMNPV. In cultured sf9 cells, it infected about 60% of the cells, vs wt which infected close to 100%. Also, although DisaGV gp64 was fusogenic for Sf9 cells at low pH, it was less efficient than AcMNPV gp64 (66). DisaGV gp64 shows 67-74% identity to many NPV gp64 sequences in genbank. Phylogenetic analysis of DisaGV gp64 suggests that its incorporation into the GV genome is not a recent event (Figure 9) although it does clearly belong to the baculovirus gp64 lineage. It has not been determined if gp64 is the main fusion protein of DisaGV or if its F protein is active and is also involved in fusion.

Figure 8.

A model for the transfer and incorporation of envelope fusion proteins (EFP) between viral families. Orthomyxoviridae: The original orthomyxoviruses likely had gp64 as their EFP because they originated as pathogens of invertebrates at a time predating (more...)

Figure 9.

Phylogeny of the GP64 viral envelope fusion protein in the Baculoviridae and Orthomyxoviridae. This is a combination tree: Neighbor Joining Best Tree with Neighbor Joining Bootstrap (1000 replicates). The numbers in bold indicate Bootstrap values (1000 (more...)

GP64 and the Orthomyxoviridae. In addition to the Group I NPVs, orthologs of gp64 are also found in several genera of the Orthomyxoviridae. Currently, there are 7 genera of Orthomyxoviridae, four that appear to be specific to vertebrates (birds and mammals)(Influenza virus A, B, C, and D), and encode a hemagglutinin (Class I) that facilitates viral attachment to sialic acid residues and fusion. Genera A and B also encode a neuraminidase that digests the receptor and prevents the binding of newly replicated virions to cells, whereas genera C and D encode a combination hemagglutinin-esterase (HE) that has both functions. The Isavirus genus of Orthomyxoviridae (infectious salmon anemia virus -ISAV), a pathogen of Atlantic salmon, appears to encode the attachment and fusion domains on two different molecules. Although evolutionarily distant, structural evidence suggests that the ISAV F protein is a Class I fusion protein related to those from other orthomyxoviruses (67, 68). In contrast, the Quaranjaviruses and Thogotovirus genera lack genes similar to HA and neuraminidase and instead encode an ortholog of baculovirus gp64 (Class III) that is involved in both attachment and fusion. These viruses are associated with ticks and birds, but some have been reported to infect humans. In addition, an unclassified Orthomyxovirus, Sinu virus, associated with mosquitoes also encodes gp64. Primary sequence and structural information indicate that the baculovirus and orthmyxovirus gp64 molecules have a common origin (69). Phylogenetic analysis of gp64 indicates that the Sinu and baculovirus gp64s have a common lineage distinct from the other orthomyxovirus gp64 suggesting that the baculovirus gp64 may have originated from a virus in the Sinu virus lineage (Figure 9). The post-fusion structure of AcMNPV GP64 indicates that six of seven disulfide bonds are conserved between thogotovirus and baculovirus GP64 and one forms an intermolecular bond involved in trimer formation (70). The data indicate that the fusion peptide and receptor binding sites co-localize to a hydrophobic patch located in two loops (L1 and L2) at the tip of the trimer (Figure 10). Further evidence for the involvement of L1 and L2 was subsequently provided in studies that employed alanine scanning mutagenesis (71) (72). A third loop (L3) attached to L2 was not involved in binding or fusion. It was suggested that transient forms of GP64 embed hydrophobic side chains into cell membranes triggering endocytosis independent of specific receptor molecules. This lack of specific receptors (see below) and the affinity of GP64 for cell membranes may explain its ability to facilitate the entry of AcMNPV into a wide variety of different cell types. In addition, it was suggested that the lack of a high affinity receptor is consistent with AcMNPV systemic infections. Since the infection has already been initiated by midgut infection using a mechanism independent of GP64, GP64 has evolved to spread the infection to as many cell types as possible via systemic infection. A comparison of the major structural features of GP64 and F proteins is shown in Figure 10.

Figure 10.

Structure of the baculovirus F (fusion) protein (Ld130) from LdMNPV and AcMNPV GP64. A) Ld130 F protein. Shown is a predicted signal peptide (SP), fusion peptide (FP) and transmembrane domain (TM) including the amino acid coordinates. The cleavage site (more...)

Evolution of Group I (GP64 containing) baculoviruses. As indicated in Chapter 1, Group I baculoviruses appear to have been a lineage that had, in addition to gp64, 11 genes not present in other lepidopteran NPVs. Since many groups of NPVs have sets of genes not present in other lineages, this would not have been extraordinary as such genes might be lost and gained over time. However, the incorporation of gp64 appears to have changed the biology of the lineage enough so that they evolved into a distinct group of viruses (73). In addition, this evolutionary event was recently recapitulated with the demonstration that gp64 could pseudotype and partially rescue a Group I virus deleted for its F protein (74) (75). However, in those experiments, the recombinant virus lacked both the F protein and the other genes unique to the Group I lineage. The latter includes genes encoding the transactivator IE-2 (Ac151) and the global transactivator ortholog among others that could have facilitated the evolution of this lineage after it incorporated gp64 (73).

Fusion protein-F, (Ac23). Although F (Ac23) is not an active envelope fusion protein in AcMNPV, in Group II NPVs, GVs and the dipteran virus (CuniNPV), orthologs of F are likely used as the fusion protein, because all these viruses lack homologs of gp64. An exception appears to be the hymenopteran NPVs that lack homologs of both the F and GP64 proteins (Figure 7) (see discussion in Chapter 3). Several reports have described investigations in cultured cells on AcMNPV in which gp64 was substituted with the F gene from other baculoviruses. In one study, a construct with Ac23 was not completely rescued by Se8 (77), whereas in another, the presence of Ac23 appeared to be required for the elevation of infectivity to near wt levels (78). It was suggested that these contrasting results might be due to the different strength of the promoters used to express Se8 in the two constructs (78). F proteins of Group II NPVs function as low-pH envelope fusion proteins (79) (80) and can also rescue AcMNPV lacking gp64 (77) (81). The F protein of granuloviruses has been implicated as a fusion protein due to their lack of a gp64 ortholog. Although the gene encoding one such protein (PlxyGV orf26) failed to mediate fusion when incorporated into the genome of an AcMNPV mutant bacmid lacking gp64 (77), another from Agrotis segetum GV (Agse orf108) was able to rescue AcMNPV lacking gp64 (82). Other GV F proteins can also substitute for gp64 (83). F proteins appear to be members of a large and diverse family of viral envelope fusion proteins called Class I (see above), reviewed in (70). They are present as homotrimers and are synthesized as a precursor that is cleaved by a furin-like proteinase into two subunits, and near the amino terminus of C-terminal peptide is a hydrophobic fusion peptide (Figure 10). Class I fusion proteins include many other viral fusion proteins such as influenza HA (e.g., see (84, 85)). In addition, orthologs of the baculovirus F gene are also found as the env gene of insect retroviruses (Figure 8) (reviewed in (65) and are also present in some insect genomes (86, 87) (see Chapter 11). With the recent determination of the structure of a variety of viral envelope fusion proteins, when baculovirus F and insect retrovirus env proteins are analyzed using the Hhpred structure prediction program (88), they all have predicted structures similar to the fusion proteins of paramyxoviruses. Some probabilities are quite high; the PxGV or26, 95%; Kanga env 97%; LD130, 92%. Even the nonfunctional Ac23 from the virus that uses gp64 as its fusion protein, scored about 90%. Paramyxoviruses are negative strand RNA viruses and they only seem to be found in vertebrates, although there is one report from a parasitic insect of birds. Although that data might have been due to the insect feeding on an infected host (89). In addition, recent evidence suggests that the F and GP64 categories of fusion proteins may be distantly related (70).

Although inactive as a fusion protein in Group I viruses, the F protein homolog in OpMNPV (Op21) is glycosylated. Similarly, in Helicoverpa armigera NPV (HearNPV) where F is the active fusion protein, it is N-glycosylated at 5 of 6 sites (76). In OpMNPV it was associated with the envelope of BV and with the membranes of OpMNPV infected cells (90). In AcMNPV, the F homolog (Ac23) is also associated with BV membranes and its deletion from the genome results in infectious virus with titers similar to wt in cultured cells, but the time to kill larvae is somewhat extended (91). In addition, antibodies against some selected regions of GP64 appeared to inhibit binding of BV to Sf9 cells of Ac23 deleted virus to a greater extent than wt virus. This was interpreted to suggest that Ac23 may increase the binding of BV to Sf9 receptor molecules (92). It could also indicate that Ac23 is closely associated with GP64 in the virion envelope and when it is absent, the antibodies have greater access to GP64. Proteomic studies found Ac23 to be associated with AcMNPV ODV as was the homolog in CuniNPV (93, 94). This suggests that it is transported to the nuclei of infected cells. The significance of this is not clear. Whereas AcMNPV is able to enter a variety of vertebrate cells, when the gene encoding its envelope fusion protein, gp64, was replaced by F genes from 5 different NPVs, although some replicated well in insect cells, none of these recombinants was capable of entering vertebrate cells (81) suggesting that they have a much greater cell specificity than GP64.

Identification of virion proteins using mass spectrometry and bacmid knockouts: Is a protein really an essential component of a virion’s structure?

A major advance in the enumeration of proteins associated with virion components has involved the use of mass spectrometry. Fractionating BV and ODV into envelope and nucleocapsid fractions and analyzing their content using mass spectrometry in conjunction with data from DNA sequencing, has provided a wealth of information of many proteins associated with these structures. This information can include both host proteins and also modifications of proteins. These studies also identify proteins that would not be predicted to be part of the virion structure (93-95) including a variety of proteins involved in DNA replication and transcription. It is unknown whether the presence of these proteins is adventitious or if they are bona fide structural proteins and play a role in accelerating the initiation of the infection cycle. The presence of proteins, such as DNA polymerase, likely reflects an intimate relationship between DNA replication and packaging, and nucleocapsid assembly and envelopment. In addition, lipids can be ‘sticky,’ and proteins in close proximity during virion assembly could adhere to the envelope and co-purify with ODV. Evidence from proteomic analysis of BV support this theory as they appear to lack most proteins associated with DNA replication and transcription that have been reported from ODV, suggesting that they were stripped off the nucleocapsids as they moved through the cell to the cytoplasmic membrane (62) (96). Furthermore, the facultative association of proteins with BV was demonstrated when it was found that BV can trap baculovirus expressed chloramphenicol acetyl transferase (97). Likewise, some proteins that are present in polyhedra and associated with occluded virions may have a lesser affinity for the envelope and may be lost during the ODV purification process. Proteomic analysis combined with the use of bacmid knockout constructs provides additional information regarding whether the protein is required for virion structure. The bacmid data can provide its own set of complications such as when an observed structural defect may be due to a secondary effect on some other structure. An example is the DNA binding protein DBP (Ac25). It is not associated with virions, but when deleted, virions appear to be structurally defective. Since DBP associates with the virogenic stroma, and when deleted, this structure is absent, it has been suggested that the role of DBP in virion structure is caused by the contributions of DBP to the structure of the virogenic stroma. Without a properly formed virogenic stroma, the assembly of virions is aberrant (98). Consequently, the best data is derived from a combination of proteomic, structural investigations using knock out bacmids, and immunological data that can provide definitive information on the location of a protein. Fortunately, the latter information can be often conveniently derived from the use of epitope tagged repair viruses produced using the bacmid technology. In the following overview, I have concentrated on the proteins for which there is information in addition to that provided by mass spectrometry. Information on AcMNPV proteins, that are not covered below can be found in (1) and Chapter 12.

Host proteins associated with BV envelopes. Evidence for host proteins associated with virions is described at the end of this chapter.

Cell receptors and virus entry for budded virions

Evidence indicates that GP64 is the receptor binding protein of AcMNPV (99). It is also well documented that GP64 can mediate the entry of AcMNPV into a wide variety of vertebrate cell lines (e.g., (100)). However, the identification of the cell receptor for budded virions has remained elusive. In one study, it was suggested that cell surface phospholipids might be involved in the AcMNPV BV (GP64 mediated) entry into vertebrate cells since treating cells with phospholipase C reduced reporter gene expression in cells (101). In addition, acidic phospholipids in giant unilamellar vesicles are required for fusion with AcMNPV BV envelopes (102). A role for membrane spanning heparin sulfate proteoglycans (syndecans) in virus binding has also been proposed for entry into mammalian cells (103). Investigations also have suggested that macropinocytosis, dynamin- and clathrin-dependent endocytosis, and cholesterol in the plasma membrane, all may be involved in the entry of AcMNPV into mammalian cells (103). In mammalian cells, AcMNPV entry is facilitated by low pH. In addition, when the basicity of the basic loop in GP64 was increased, the ability of the virus to enter mammalian cells was elevated. However, this mutant failed to spread between Sf9 cells. Viruses grown in insect cells from different species showed differing efficiency of mammalian cell entry suggesting that some host factors incorporated into the virus might facilitate cell entry (104). As described above, the lack of specific receptors and the affinity of GP64 for cell membranes may explain its ability to facilitate the entry of AcMNPV into a wide variety of different cell types and this might facilitate systemic infections where many different cell types are encountered (104). Other reports indicate that BV of a virus that used an F fusion protein did not have the ability to enter the array of vertebrate cells as AcMNPV. Consequently, it was suggested that the F fusion proteins might use a different receptor from GP64 (105) (106). In contrast, another report described the ability of UV-inactivated HearSNPV expressing either F and GP64 was able to compete for receptors with an F expressing wt virus suggesting that they have similar modes of entry (74). For further information see (1).

Envelope proteins of occlusion-derived virus that are also BV associated

The source and content of the envelope is the major distinguishing feature between BV and ODV. In contrast to BV, where a few virus-encoded proteins have been identified as envelope associated, the ODV envelope is much more complex (Figure 11). There may be five or more such proteins categorized as envelope proteins along with a set of about 9-12 proteins called per os infectivity factors (PIF) (107) (108) (109) that are likely envelope components. Some of these proteins contain an N-terminal hydrophobic sequence in combination with several adjacent positively charged amino acids. These have been predicted to be motifs that target these proteins to intranuclear microvesicles that are the likely precursors from which the envelopes of occluded virus are derived (110). The following proteins (see also Table 2) have been characterized and are likely to be components of ODV envelopes (see also Figure 11).

Figure 11.

Selected structural proteins of ODV. Shown are envelope associated proteins (ODV-E), the PIF proteins (also envelope associated), the tegument protein, gp41, the DNA binding protein, p6.9, and two basal end-associated proteins, pp78/83 and VLF-1. For (more...)

Table 2.

Occlusion Derived Virus Envelope Proteins and Per os infectivity factors

BV/ODV-E26, (Ac16). Homologs of Ac16 are found in lepidopteran Group I NPVs. Evidence for the association of Ac16 with BV and ODV envelopes has been reported for AcMNPV (111) (93) (62). However, in BmNPV, the homolog of AcMNPV Ac16 (Bm8) was not identified as a virion structural protein (112). It was suggested that these conflicting results were due to the different sources of the antibodies used to detect the protein (110). Ac16 interacts with FP25 (Ac61), forms a complex with cellular actin (111), and is palmitoylated (113). A mutant in which AcMNPV orf16 (called DA26) was insertionally inactivated was viable and showed no difference from wt in infections of T. ni or S. frugiperda cells or larvae (114). In another report, a virus deleted for Ac16 infected cells showed a delay in BV production (115). It has also been shown to interact with both IE-1 and IE-0 and may be involved in the regulation of these gene products (116).

ODV-E25, (Ac94). Ac94 is a core gene present in all baculovirus genomes (117). The protein encoded by this gene was originally identified in OpMNPV, and immunogold staining with a specific antibody against Ac94 was localized to ODV envelopes (118). It has also been shown to be associated with BV and ODV of AcMNPV and HearNPV (93, 95) (62) (119) and is highly expressed in midgut cells (120). The hydrophobic N-terminal 24 aa of AcMNPV ODV-E25 appears to be a nuclear targeting signal (121). Ac94 associates with NSF and may be involved in the nuclear entry and egress of BV (122). Deletion of Ac94 resulted in a 100 fold reduction in infectious BV and ODV were not evident and virions were not occluded in polyhedra (123). When ODV-E25 is expressed as an early gene under the IE-1 promoter, it accumulates on the cytoplasmic side of the nuclear membrane rather than within nuclei, and budded virus production is severely reduced. This suggests that it might play a role in the shift from BV to ODV virions. An open reading frame of ODV-E25 encodes a microRNA that down regulates ODV-E25 expression. It was suggested that this might result in a reduction in infectious virus production and be involved in the shift to occluded virus production (124) (125). In addition, expression of Ac94 from the very late polyhedrin or p10 promoter reduced and delayed occlusion body formation suggesting that it may play a role in virion occlusion (126) (127).

ODV-EC43, (Ac109). Ac109 is a core gene present in all baculovirus genomes. Evidence suggests that it is ODV-associated in AcMNPV (93) and Helicoverpa armigera NPV (Ha94=ODV-EC43) (95, 128) and also is BV-associated in AcMNPV (62) (129). Four studies have examined deletions of Ac109 and demonstrate that it is an essential gene and when deleted, DNA replication is not affected. One study reported that deletion of Ac109 resulted in a block in nucleocapsid and polyhedron formation (130). However, the other reports described different results. One indicated that polyhedra and virions were produced by Ac109 deletions, but the virus was not infectious (129). Another study found similar results, but also showed that the nucleocapsids had defects in envelopment and the polyhedra lacked virions (131). A fourth report also described similar findings, but indicated that the BV produced by a Ac109 knockout could enter the cytoplasm, but not nuclei, and also noted that the occlusion bodies lacked virions (132).

ODV-E18, (Ac143). Ac143 is a core gene present in the genomes of all baculoviruses. An antibody generated against an Ac143-GST fusion reacted with a protein of 18 kDa in the ODV envelope fraction and Ac143 was named ODV-E18 (133). Ac143 and its HearNPV homolog were found in surveys of ODV-associated proteins by proteomic analysis (93, 95). Deletion of Ac143 results in single cell infections that produce polyhedra and therefore appears to be essential for BV production (134). In addition to its presence in ODV envelopes, it is also BV associated (62). Hhpred (135) predicts the presence of a transmembrane helix and similarity to a protein translocation complex of bacteria.

Ac144 (ODV-EC27) See below.

Per os infectivity factors (PIF); ODV envelope associated proteins required for midgut infection

Ac22 (pif2), Ac68 (pif6), Ac83(pif8), Ac96 (pif4), Ac108(pif9), Ac110 (pif7), Ac115 (pif3), Ac119 (pif1), Ac138 (p74-pif0), Ac148 (pif5). Per os infectivity factors were originally identified because they were required for infection of insects, but dispensable for infection of cultured cells (107) (108, 109, 136). There are at least 10 pif genes encoded by AcMNPV (Figure 11). Orthologs of PIF genes (except PIF9) are present in all baculovirus genomes and most are also found in genomes of nudiviruses (137) and some are found in other virus pathogenic for invertebrates (see Chapter 1). Three additional possible PIF genes have also been noted (Ac46 (ODV-E66), Ac145, and Ac150) (see below). AcMNPV mutants lacking pif 1, 2, or 3 are not orally infectious for T. ni or S. exigua larvae based on feeding of 10,000 PIB of the deleted virus. They are also not orally infectious for H. virescens, except for the PIF2 mutant that shows limited infectivity. In contrast, injection of 1 pfu of the three deletion mutants into third instar larvae of these three species caused over 80% mortality.

PIF function; binding and entry to midgut cells. Most of the PIF proteins contain a hydrophobic region that is thought to be involved integration into the ODV envelope. Nine of the PIF proteins form a complex that is involved in binding and entry of ODV into midgut cells. It includes three that form a stable core complex (PIFs 1-3) and five that associate with this complex (PIFs -0(p74), -4, -6, -7, and -9) to form a larger complex. In addition, Pif8(Ac83) can bind to the larger 8-subunit complex (reviewed in (108) (109, 136) (Figure 11). PIF1, PIF2, and p74 mediate specific binding of ODV to midgut cells, suggesting that they are directly involved in virus cell interaction as an initial step in infection (138) (138, 139). Although PIF-3 appears to be a component of the PIF complex, evidence suggests that it is not involved in specific binding and its function is not clear (140). PIF-5 is apparently not part of the PIF complex (141). A 35-kDa binding partner for AcMNPV P74 was detected in extracts of brush border membrane vesicles from host larvae (S. exigua), but not from a non-host (H. armigera larvae) (142). The identity of this host protein has not been determined.

Ac83 (VP95)(PIF8); essential for nucleocapsid assembly and is a PIF protein. Ac83 (PIF8) encodes a virion associated protein called VP91 that was originally characterized in OpMNPV (143). It is a core baculovirus gene and is also found in nudiviruses (137) and possibly in several insect genomes, e.g., Anopheles gambiae (E = 5e-04). It is an essential gene because a deletion mutant in BmNPV (Bm69) did not produce BV (144). It was found to associate with the PIF complex (141) and although the complete Pif8 gene is required for viability, when a predicted region containing three zinc fingers was deleted, the virus was not infectious when fed to larvae, but could replicate when injected into the haemocoel confirming that it is a PIF protein (145). Originally the zinc finger region was predicted to contain a chitin binding domain, but evidence indicated that AC83 does not bind to chitin (145). The zinc finger region appears to be involved in the assembly and localization of the PIF complex to ODV envelopes and the binding or entry of ODV into midgut cells (146). It was also determined that the ac83 gene contains a cis-acting nucleotide sequence essential for nucleocapsid assembly and is called the nucleocapsid assembly-essential element (NAE) (147). Therefore, the Ac83 gene appears to encode at least two functions; the zinc fingers are involved in the PIF function, whereas the NAE is a nucleotide sequence involved in nucleocapsid assembly. For more information, see Chapter 5.

Ac46 (ODV-E66), a chondroitinase and a PIF protein. Ac46 is a component of ODV envelopes (148) is the only known viral chondroitinase (149). It interacts with several members of the PIF complex (reviewed in (108)). Homologs of Ac46 are found in the genomes of all Group I NPVs, GVs, and most Group II NPVs, but not in hymenopteran or dipteran viruses. Two copies of the gene are present in some genomes (e.g., SeMNPV). In a study of AcMNPV, an ODV-E66 deletion was observed to kill Plutella xylostella larvae as efficiently as wt when injected into larvae, however when infected per os, the LD50 was1000 fold greater for the mutant than wt virus. Therefore it was suggested that ODV-E66 is a per os infectivity factor (150) and could be designated PIF10. A truncated form of Ac46 lacking the N-terminal 66 amino acids was found to be secreted into the medium by infected cells and had chondroitinase activity (149). Chondroitinases have been shown to regulate cytokine and growth factors and can influence a variety of processes including development, inflammation, and organ morphogenesis. Chondroitin sulfate is present in the peritrophic matrix (PM) of B. mori and it can be digested by Ac46. This has led to the suggestion that Ac46 enhances the primary infection by digesting the chondroitin sulfate in the PM (151). This activity could be the reason Ac46 is a per os infectivity factor. Chondroitin glycosaminoglycans have been found to localize to the apical midgut microvilli of Anopheles gambiae (152) suggesting that if Lepidoptera are similar, this enzyme might also be involved in recognition or entry into midgut cells. Ac46 was previously shown to have hyaluronan lyase activity suggesting that it might be involved in penetrating the extracellular matrix which is composed of hyaluronan (153). However, its activity as a hyaluronan lyase was minimal in the chondroitinase study (149). When the N-terminal 23 amino acids of ODV-E66 are fused to a reporter gene, it is targeted to the nucleus (121). This is consistent it being a protein involved in ODV envelope which develops in association with the nucleus. The crystal structure has been determined (154).

Ac 145 and 150; more possible PIF proteins. These two genes encode small proteins (~9 and 11 kDa, respectively) that are related to one another (23% amino acid sequence identity) and localize to ODV envelopes (155). Close relatives of Ac 145 are found in all baculoviruses except the dipteran NPV. In contrast to Ac145, close relatives of Ac150 are only found in a few NPVs closely related to AcMNPV. Ac145 and 150 are predicted to encode a domain thought to bind to chitin (156). In one study (155), deletion of AcMPNV Ac145 led to a six-fold drop in infectivity in T. ni, but not H. virescens larvae. An effect of deletion of AcMNPV Ac150 was not detected. Deletion of both genes causes a major (39 fold) reduction of infectivity for H. virescens. Injection of BV of the double mutant intrahemocoelically produced the same level of infection as injected wt BV, suggesting that these genes play a role in oral infection. Ac145 and 150 were found to be associated with both BV and ODV (155). In another study (157), occluded virions deleted for Ac150 were found to be significantly less virulent when administered per os than the wt virus in Heliothis virescens, Spodoptera exigua and Trichoplusia ni larvae. Evidence suggested that the mutant had a reduction in its ability to establish primary infections in midgut cells.

Nucleocapsid Structure

Baculovirus nucleocapsids have a defined rod-shaped capsid this is capped by distinct apical and basal structures (Figure 12). Although many proteins appear to be capsid associated, a few appear to be located to end structures. Three of these VP80, VLF-1 and pp78/83 are shown in Figure 11. Although shown located to the basal region, this has not been definitively shown for VP80 or VLF-1.

Figure 12.

Morphology of AcMNPV capsids stained with uranyl acetate. The figure shows the morphology of the two ends, one with a conical shape (arrow). Scale bar, 50 nm .From Au and Pante (241). Reproduced with permission of Elsevier Limited via Copyright (more...)

Essential BV and ODV Nucleocapsid associated proteins encoded by all baculoviruses

As described above many proteins have been associated with BV and ODV nucleocapsids (reviewed in (1) however their role, if any, is unknown. Rather than an exhaustive list of these proteins, in this section I will focus on proteins that evidence suggests are essential components of both BV and ODV nucleocapsids (Table 3). The following are core baculovirus proteins that appear to be nucleocapsid associated.

Table 3

Selected Proteins Associated with Baculovirus Nucleocapsids

VP1054, (Ac54). VP1054 was named for the size of its orf and encodes a protein required for nucleocapsid assembly. A ts mutant failed to produce nucleocapsids at the non-permissive temperature, indicating that it is an essential gene. It is found in both BV and ODV (158) and it interacts with 38K (Ac98) (22). Ac98 is likely to be a phosphatase. VP1054 also associates with BV/ODV-C42 and VP80, but not VP39 suggesting that it is critical for nucleocapsid assembly (159). When the vp1054 gene was deleted from a bacmid, the bacmid was not infectious and nucleocapsids appeared to be replaced with tube-like structures. It was suggested that vp1054 is related to a cellular protein called PURα that binds to purine-rich sequences and may be involved in DNA packaging and it was shown to bind to single stranded DNA or RNA sequences that contained runs of GGN. Therefore, it was suggested that it might interact with the orf1629/p/78/83 (ac9) sequence which encodes a series of prolines and therefore is rich in GGN codons (160).

Ac66. Orthologs of Ac66 are present in all baculovirus genomes. An AcMNPV bacmid deleted for Ac66 was severely compromised and BV titers derived from transfected cells were reduced by over 99% compared to wt. In addition, at low titers the mutant BV appeared to infect single cells and was unable to spread to other cells. Although the nucleocapsids appeared to be normal and had an electron-dense core suggesting that they contained DNA, they appeared to be trapped in the virogenic stroma, indicating that Ac66 was required for the efficient egress of virions from nuclei. The deletion did not affect the levels of DNA replication or polyhedrin transcription, but the production of occlusion bodies was eliminated (161). This suggests that Ac66 is required both for egress of virions from nuclei and also may be involved in the nucleation of polyhedra. Ac66 in BV but not ODV is ubiquitinated by the viral ubiquitin and appears to interact with Ac141 (predicted to be an E3 ubiquitin ligase). It was suggested that this might be a signal for BV egress in contrast to ODV that remain in nuclei (162, 163). For more information see Chapter 12.

VLF-1, (Ac77). The very late factor, VLF-1, is a member of the lambda integrase (Int) family of proteins and was originally identified because it influences the hyperexpression of very late genes (164) possibly by binding to their regulatory regions (165). VLF-1 appears to be a structural protein present in both BV and ODV (166) and is clearly required for the production of nucleocapsids. Bacmid deletion mutants produce tube-like structures that stained with vp39 antiserum suggesting that the lack of VLF1 prevents normal capsid assembly. Bacmids with point mutations in the conserved tyrosine form normal appearing capsids, but are also not infectious. VLF-1 localized to the end regions of nucleocapsids further suggesting that it is a structural protein (167). VLF-1 was also associated with ODV virions as determined by proteomic analysis in AcMNPV and CuniNPV, but not HearNPV (93-95). For additional information, see Chapter 5 and Chapter 12.

GP41, tegument protein, (Ac80). GP41 is modified with O-linked N-acetylglucosamine, and is located between the virion envelope and capsid in a structure called the tegument (168, 169). Homologs are present in all baculovirus genomes. Based on the characterization of a ts mutant, it is an essential gene required for the egress of nucleocapsids from the nucleus (170). Under the restrictive temperature, this mutant produces no BV and infection is limited to single cells. Proteomic studies identified GP41 in both BV and ODV of HearSNPV (119). In addition, it was found that oligomerization of gp41 was required for BV production (171).

VP39, (Ac89). VP39 is thought to be the major capsid protein. It is one of the three most abundant proteins found by proteomic analysis of BV (62). Homologs of vp39 are present in all baculovirus and nudivirus genomes. It has been observed that VP39 interacts with a conserved domain of kinesin 1 and it has been suggested that this interaction is involved in the transport of nucleocapsids destined to become BV to the cell membrane after their assembly in nuclei (172). Based on mutagenic analysis it appears to be required for proper DNA packaging and nucleocapsid assembly (173).

Ac92, (P33), a sulfhydryl oxidase. Ac92 is an essential core gene that encodes a sulfhydryl oxidase, and is likely involved in the production of disulfide bonds in viral proteins (174) (175). It forms a stable complex with the tumor suppressor gene p53 and appears to enhance its apoptotic function (176) (177). P33 was found to be associated with ODV virions by proteomic analysis in several baculoviruses (93-95) (119) and in both AcMNPV BV and ODV in western blot analyses (175) (178). It is unclear whether Ac92 is required for virion structure or is associated with virions because it facilitates disulfide bond formation during virion assembly. Further information on Ac92 is available in Chapters 5, 7, and 12.

Ac98 (38K). Ac98 is encoded by all baculoviruses and is associated with both BV and ODV nucleocapsids. By yeast two-hybrid assays it interacted with VP1054, VP39, VP80, and itself (22). When deleted, tube-like structures devoid of DNA but that stain with vp39 antibody are produced (179). It is related to a set of enzymes including CTD phosphatases and evidence indicates that it dephosphorylates the p6.9 DNA binding protein which allows it to be packaged with the viral DNA (180).

P6.9, (Ac100). P6.9 is a DNA binding protein and one of the three most abundant proteins found in proteomic analysis of BV (62). It is a small (55 aa) arginine/serine/threonine rich protein (181). Homologs appear to be encoded by all baculovirus genomes, but may be difficult to identify in computer analyses because of their small size and repetitive amino acid content. P6.9 was originally identified as a DNA binding protein in a GV (182) and the homolog was subsequently identified from AcMNPV (181). The high concentration of arg and ser/thr residues is similar to protamines that are also small molecules of 44-65 amino acids (183, 184) present in sperm nuclei of many higher eukaryotes and are involved in the production of highly condensed DNA. Arginine has a high affinity for the phosphate backbone of DNA, and the polyarginine tracts in protamines neutralize the phosphodiester backbone, whereas the ser and thr residues interact with other protamine molecules, thereby yielding a neutral, highly compact DNA-protein complex that is biochemically inert. P6.9 localizes to the nuclear matrix during infection (185). Once viral DNA has been delivered to the nucleus, p6.9 is phosphorylated (see below) resulting in both DNA and p6.9 being negatively charged. It is thought that this causes the removal of p6.9 from the viral DNA thereby allowing access to transcription factors (186). AcMNPV has two candidate kinases that could be involved in this process. Protein kinase 1, PK1 (Ac10), is encoded by lepidopteran baculoviruses (GVs and NPVs), whereas PK2 (Ac123) is encoded by only a few baculoviruses closely related to AcMNPV. Neither protein was found to be associated with ODV by mass spectrometry (93-95). However, a kinase was found to be associated with BV and ODV, although its source was not determined (187). P6.9 was found to co-localize with viral DNA and to fractionate with RNA polymerase II at 24 hpi (141). After synthesis, p6.9 is hyperphosphorylated, at least in part by pk-1 and this is associated with high levels of expression of very late genes (188) (189). This occurs immediately upon synthesis and p6.9 is dephosphorylated by the 38k protein (180) before being complexed with DNA (189). Using an AcMNPV bacmid deleted for p6.9, nucleocapsids were not produced although tube-like structures similar to those associated with the deletion of VLF-1 and Ac98 (see above) were observed. The mutant appeared to synthesize normal amounts of DNA, but did not produce infectious virus (190). BmNPV micro RNA-3 (mir-3) appears to regulate, at least in part, the expression of BmNPV p6.9 (191).

BV/ODV-C42, (Ac101). Ac101 is a core gene and encodes a capsid-associated protein found in both BV and ODV (93). In addition, it was reported to interact in a yeast two-hybrid assay and by native gel electrophoresis (192) with the actin nucleation factor pp78/83 (Ac9) (described below) and is required for its transport into nuclei (193). It stabilizes P78/83 by inhibiting its degradation. C42 also interacts with Ac102 and this suppresses ubiquitination of C42 further regulating the stability of P78/83 (194-196). Deletion of Ac101 affected nucleocapsid formation but, although lethal, did not appear to affect DNA synthesis (197).

Ac141 (exon0). Ac141 is conserved in all lepidopteran baculoviruses and is associated with both BV and ODV nucleocapsids (198) (62) and interacts with BV/ODV-C42 (Ac101) and FP25 (Ac61) (199). It appears to be required for the efficient transport of nucleocapsids from nuclei through the cytoplasm (198, 200). It appears to both co-localize with and co-purify with β-tubulin. In addition, inhibitors of microtubules reduced BV production by over 85%. Therefore it has been suggested that the interaction of Ac141 with microtubules might be important in the egress of BV (201). It has also been shown to interact with a conserved domain of kinesin 1, a motor protein involved in transporting cargo along microtubules to the periphery of the cell further supporting a role for microtubules in transport of virions to the cell surface (172, 202). Hhpred (135) predicts with over 90% probability that the C-terminal ~90 amino acids has structural similarity to E3 protein ubiquitin ligase along with several other proteins. Deletion of ac141 and vubi results in single cell infection and BV were not produced. The ubiquitination of Ac141 was essential for optimal production of BV. BV but not ODV nucleocapsids were ubiquitinated by vUbi. The target was Ac66 and it was shown to co-localize with vUbi and Ac141 at the nuclear periphery. It was suggested that the ubiquitination of capsid proteins may be a signal for BV egress from nuclei (162).

Ac142, (p49). Ac142 is encoded by a core gene and is associated with both BV and ODV virions. Deletion of Ac142 appeared to affect nucleocapsid formation but, although lethal, did not appear to affect DNA synthesis (197). Similar results were reported for the homolog in BmNPV (Bm118) (203). Another study describing a different bacmid construct that deleted less of the Ac142 gene showed similar results except that nucleocapsids, although fully formed, were not enveloped in nuclei and were not occluded (204). It is unclear whether the difference in the two studies was due to the removal of a 3' processing signal for the upstream Ac141 gene in the former investigation, or to the presence of a significant portion of the Ac142 orf in the latter study. Proteomic analysis also suggests that Ac142 is ODV-associated in three different viruses (93-95). In addition, it was reported that Ac142 interacts with components of the ESCRT-III complex and may be involved in the release of nucleocapsids at the nuclear membrane (205).

Ac144. Ac144 is a core gene and was originally named ODV-EC27 and it was suggested that it is a cyclin (206). However, another investigation using an HA-tagged Ac144 recombinant virus found that Ac144 was expressed as an ~ 33.5 kDa protein which conforms to the predicted MW (197). In addition, it was found to be BV associated (62, 197). Proteomic analysis suggests that Ac144 is ODV-associated in three different viruses (93-95). A variety of investigations have been conducted on Ac144. Initially, it was confirmed that its transcript initiates at a late promoter element (133). It was reported to interact in a yeast two-hybrid assay with Ac101 described above (also named C42) and with both Ac101 and p78/83 (Ac9) in native gel electrophoresis assays (192). It was also found that deletion of Ac144 resulted in amorphous electron dense structures that stained with vp39-capsid antibodies, but no nucleocapsids were evident. Although lethal, deletion of Ac144 did not appear to affect DNA synthesis (197).

Other structural proteins

The following structural proteins are found in some, but not all baculovirus genomes. This could indicate either that they are not present or have evolved to such an extent that their relatedness can no longer be identified in the genomes in which they are not found.

PP78/83, (Ac9). PP78/83 is a phosphorylated protein that is located at one end of nucleocapsids (207, 208). It is a Wiskott-Aldrich syndrome protein (WASP)-like protein. Such proteins are involved in nuclear actin assembly, and it has been demonstrated that pp78/83 serves this function during AcMNPV infection (209) (210) (see Chapter 3). Homologs of pp78/83 are found in all lepidopteran NPV genomes. It is an essential gene, and because it is located adjacent to the polyhedrin gene, it was originally manipulated via complementation to elevate the frequency of obtaining recombinant baculoviruses at the polyhedrin locus (211).

GP37 (Ac64). Orthologs of gp37 have been found in the genomes of all alpha- and betabaculoviruses (212). In AcMNPV it is expressed as a late gene (213) and is nonessential for replication in cell culture or T. ni larvae (214). The BmNPV homolog (Bm52) was also found to be non essential in BmN cells (144). The gp37 homolog in SpltNPV has been reported to contain chitin binding domains and is capable of binding to chitin (215). The gp37 of CpGV also binds chitin and was able to enhance per os infections (216). Insect proteins, such as the coagulation protein hemolectin, also have chitin-binding domains (217). Whether gp37 somehow inhibits or redirects such pathways remains to be determined. GP37 was reported to be polyhedron associated in AcMNPV and to be N-glycosylated (218). It was also found to be BV associated (62). In OpMNPV infected L. dispar cells, GP37 was found to be an N-glycosylated protein located in cytoplasmic occlusions late in infection (219). In entomopox viruses the gp37 ortholog forms crystallized spindle-like structures. These structures have been suggested to digest the peritrophic matrix (220). Structural analysis of these spindles indicated that they contained a globular domain that is related to lytic polysaccharide monooxygenases of chitinovorous bacteria. It is thought that upon ingestion by the host, the spindles are dissolved and the monooxygenase domain is exposed and can then digest the chitin-rich peritrophic matrix (221) thereby facilitating the passage of virions through the peritrophic matrix and allowing them access to midgut cells. It has been reported that GP37 can degrade the peritrophic matrix and also facilitates the binding of ODV to midgut cells (222). The Spodoptera litura NPV genome was found to contain a gene that is a fusion of ubiquitin and gp37 (for discussion in Chapter 12, see Ac35, ubiquitin) and the protein was associated with the envelopes of BV and ODV (223). For more information see Chapter 12.

Vp80 (Ac104). Orthologs of Ac104 are found in all Group I and II lepidopteran NPV genomes, but not in those of GVs or hymenopteran or dipteran NPVs. It is capsid associated in both OpMNPV (224) and AcMNPV (93, 225, 226) and interacts with 38K (Ac98) (22). Deletion of Vp80 showed that it is an essential gene and resulted in nucleocapsids that were unable to move from the virogenic stroma (227). It appears to localize in nuclei near actin scaffolds that may connect the virogenic stroma to the nuclear envelope. In addition, it co-immunoprecipitates with actin. It also appears to localize to one end of nucleocapsids and contains sequences similar to paramyosin motifs that may be involved in the transport of virions to the periphery of nuclei (227). It forms dimers, contains a C-terminal region that was predicted to contain a basic helix-loop-helix domain, and binds to DNA (228).

Ac109 (ME53). Homologs of ac139 are present in the genomes of all the lepidopteran NPVs and GVs, but have not been reported in hymenopteran or dipteran baculovirus genomes. It is BV and ODV associated (229). One study indicated that AcMNPV deleted for this gene is not viable and fails to replicate its DNA and does not produce nucleocapsids. However, cells transfected with DNA from the mutant showed early stages of cpe, including nuclear enlargement and the formation of granular material in the nucleus (230). This suggests that the mutant is blocked in an early gene function. This is consistent with its original characterization as a major early gene (231). However, another study showed that deletion of Ac139 did not alter DNA replication, but results in a 1000-fold reduction in BV titer. In addition, it was found that it appears to be required both early and late in infection (229). ME53 fused to GFP localized mostly to the cytoplasm early and to nuclei late in infection. However, foci of ME53 were also noted at the cell periphery late in infection and co-localized with gp64 and VP39-capsid and was capsid associated in BV. It was suggested that it may provide a connection between the nucleocapsid and the viral envelope (232).

P24-capsid, (Ac129). Ac129 (P24) was found to be associated with both BV and ODV of AcMNPV and OpMNPV by Western blot and electron microscopic analyses (233). It is likely to be nonessential as interruption of this gene with a transposable element in a strain of AcMNPV has been reported (234, 235) and it can also be deleted from BmNPV with no detectable effects (144). Homologs of Ac129 are present in all Group I /II and GV genomes. The Ac129 homolog was not reported to be associated with ODV of HearNPV (95). It was also reported to be BV associated (62).

Additional virion associated proteins. Many more proteins have been found to be associated with AcMNPV virions. Information on these and summarized in (1) and Chapter 12.

Host proteins and protein modification

Host proteins. Analysis of BV from AcMNPV using mass spectrometry identified 48 virally encoded proteins and 11 host proteins (62). In addition, a variety of host proteins were also associated with preparations of HearSNPV ODV and BV, with 21 and 101 host proteins identified, respectively. Many of these proteins could be simply trapped as the virions are assembled or are present as normal components of the membranes from which their viral envelopes were derived. However, some components could be essential for virion function. One such protein, cyclophilin A was associated with both HearSNPV BV and ODV. Cyclophilin A catalyzes the isomerization of peptide bonds from trans to cis at proline residues, a process that can be important in protein folding. In HIV-1, but not other primate immunodeficiency viruses, cyclophilin A associates with a proline rich region of the GAG polyprotein and is required for the production of infectious virions (236). The 11 host proteins that were identified in AcMNPV BV (62) included cytoplasmic actin and actin depolymerizing factor, perhaps reflecting the role actin plays in BV transport (210). In addition, it was observed that baculoviruses grown in different insect cells show differing efficiencies in mammalian cell entry suggesting that some host factor may facilitate cell entry (104).

Post translational modification of structural proteins. Proteomic analysis using several techniques was applied to both BV and ODV of the HearSNPV (119). They reported N-glycosylated proteins in only BV, but not in ODV. These included F, FGF, V-CATH, ChiA, P26 (=Ac136), and ODV-E18 (=Ac143). One of these, ODV-E18, is present in both BV and ODV. They also observed differing phosphorylation profiles between the two phenotypes with 38 sites in ODV associated proteins, and 4 in BV. Serine was the predominant phosphorylation site (72.1%) followed by Thr (22.3%) and Tyr (4.6%). The phosphorylated proteins (phosphoproteome) of B. mori cells infected with BmNPV has been examined. Many phosphorylated host and viral proteins were identified, some of which were hyperphosphorylated including p6.9 (237).

References1.Blissard GW, Theilmann DA. Baculovirus Entry and Egress from Insect Cells. Annu Rev Virol. 2018;5:113–139. [PubMed: 30004832]2.Evans HF. 1986. Ecology and epizootiology of baculoviruses, p 89-132. In Granados RR, Federici BA (ed), The biology of baculoviruses, vol II. CRC Press, Inc., Boca Raton.3.Perera OP, Valles SM, Green TB, White S, Strong CA, Becnel JJ. Molecular analysis of an occlusion body protein from Culex nigripalpus nucleopolyhedrovirus (CuniNPV). J Invertebr Pathol. 2006;91:35–42. [PubMed: 16376931]4.Ji X, Axford D, Owen R, Evans G, Ginn HM, Sutton G, Stuart DI. Polyhedra structures and the evolution of the insect viruses. J Struct Biol. 2015;192:88–99. [PMC free article: PMC4597613] [PubMed: 26291392]5.Chaivisuthangkura P, Tawilert C, Tejangkura T, Rukpratanporn S, Longyant S, Sithigorngul W, Sithigorngul P. Molecular isolation and characterization of a novel occlusion body protein gene from Penaeus monodon nucleopolyhedrovirus. Virology. 2008;381:261–7. [PubMed: 18829059]6.Wang Y, Jehle JA. Nudiviruses and other large, double-stranded circular DNA viruses of invertebrates: New insights on an old topic. J Invertebr Pathol. 2009;101:187–93. [PubMed: 19460388]7.Coulibaly F, Chiu E, Ikeda K, Gutmann S, Haebel PW, Schulze-Briese C, Mori H, Metcalf P. The molecular organization of cypovirus polyhedra. Nature. 2007;446:97–101. [PubMed: 17330045]8.Coulibaly F, Chiu E, Gutmann S, Rajendran C, Haebel PW, Ikeda K, Mori H, Ward VK, Schulze-Briese C, Metcalf P. The atomic structure of baculovirus polyhedra reveals the independent emergence of infectious crystals in DNA and RNA viruses. Proc Natl Acad Sci U S A. 2009;106:22205–10. [PMC free article: PMC2799703] [PubMed: 20007786]9.Afonso CL, Tulman ER, Lu Z, Balinsky CA, Moser BA, Becnel JJ, Rock DL, Kutish GF. Genome sequence of a baculovirus pathogenic for Culex nigripalpus. J Virol. 2001;75:11157–65. [PMC free article: PMC114695] [PubMed: 11602755]10.Summers MD. 1977. Characterization of shrimp baculovirus. EPA Ecological Research Series:EPA-600/3-77-130.11.Carstens EB, Krebs A, Gallerneault CE. l986. Identification of an amino acid essential to the normal assembly of Autographa californica nuclear polyhedrosis virus polyhedra. J Virol. 58:684–688. [PMC free article: PMC252964] [PubMed: 3528527]12.Ji X, Sutton G, Evans G, Axford D, Owen R, Stuart DI. How baculovirus polyhedra fit square pegs into round holes to robustly package viruses. EMBO J. 2010;29:505–514. [PMC free article: PMC2824454] [PubMed: 19959989]13.Gati C, Oberthuer D, Yefanov O, Bunker RD, Stellato F, Chiu E, Yeh SM, Aquila A, Basu S, Bean R, Beyerlein KR, Botha S, Boutet S, DePonte DP, Doak RB, Fromme R, Galli L, Grotjohann I, James DR, Kupitz C, Lomb L, Messerschmidt M, Nass K, Rendek K, Shoeman RL, Wang D, Weierstall U, White TA, Williams GJ, Zatsepin NA, Fromme P, Spence JC, Goldie KN, Jehle JA, Metcalf P, Barty A, Chapman HN. Atomic structure of granulin determined from native nanocrystalline granulovirus using an X-ray free-electron laser. Proc Natl Acad Sci U S A. 2017;114:2247–2252. [PMC free article: PMC5338516] [PubMed: 28202732]14.Minion FC, Coons LB, Broome JR. Characterization of the polyhedral envelope of the nuclear polyhedrosis virus of Heliothis virescens. J Invertebr Pathol. 1979;34:303–307.15.Whitt MA, Manning JS. A phosphorylated 34-kDa protein and a subpopulation of polyhedrin are thiol-linked to the carbohydrate layer surrounding a baculovirus occlusion body. Virology. 1988;163:33–42. [PubMed: 3279702]16.Gross CH, Russell RLQ, Rohrmann GF. The Orgyia pseudotsugata baculovirus p10 and polyhedron envelope protein genes: analysis of their relative expression levels and role in polyhedron structure. J Gen Virol. 1994;75:1115–1123. [PubMed: 8176372]17.Gombart AF, Pearson MN, Rohrmann GF, Beaudreau GS. A baculovirus polyhedral envelope-associated protein: genetic location, nucleotide sequence, and immunocytochemical characterization. Virology. 1989;169:182–193. [PubMed: 2646825]18.Russell RLQ, Rohrmann GF. A baculovirus polyhedron envelope protein: Immunogold localization in infected cells and mature polyhedra. Virology. 1990;174:177–184. [PubMed: 2403704]19.Russell RLQ, Pearson MN, Rohrmann GF. Immunoelectron microscopic examination of Orgyia pseudotsugata multicapsid nuclear polyhedrosis virus-infected Lymantria dispar cells: time course and localization of major polyhedron-associated proteins. J Gen Virol. 1991;72:275–283. [PubMed: 1993872]20.Lee SY, Poloumienko A, Belfry S, Qu X, Chen W, MacAfee N, Morin B, Lucarotti C, Krause M. A common pathway for p10 and calyx proteins in progressive stages of polyhedron envelope assembly in AcMNPV-infected Spodoptera frugiperda larvae. Arch Virol. 1996;141:1247–58. [PubMed: 8774685]21.Busby JN, K.N.Goldie, Metcalf P. 2009. Structural studies of granulovirus envelope fibres. doi: 10.2210/pdb4YE7/pdb, p]. Protein Data Bank. [CrossRef]22.Wu W, Liang H, Kan J, Liu C, Yuan M, Liang C, Yang K, Pang Y. Autographa californica Multiple Nucleopolyhedrovirus 38K Is a Novel Nucleocapsid Protein That Interacts with VP1054, VP39, VP80 and Itself. J Virol. 2008;82:12356–64. [PMC free article: PMC2593362] [PubMed: 18922869]23.Xu H, Yang Z, Zhao J, Tian C, Ge J, Tang X, Bao Y, Zhang C. Bombyx mori nucleopolyhedrovirus ORF56 encodes an occlusion-derived virus protein and is not essential for budded virus production. J Gen Virol. 2008;89:1212–1219. [PubMed: 18420799]24.Cheley S, Kosik KS, Paskevich P, Bakalis S, Bayley H. Phosphorylated baculovirus p10 is a heat-stable microtubule-associated protein associated with process formation in Sf9 cells. J Cell Sci. 1992;102:739–752. [PubMed: 1331130]25.Raza F, McGouran JF, Kessler BM, Possee RD, King LA. Phosphorylation Induces Structural Changes in the Autographa californica Nucleopolyhedrovirus P10 Protein. J Virol. 2017:91. [PMC free article: PMC5469262] [PubMed: 28424279]26.Carpentier DCJ, Griffiths CM, King LA. The baculovirus P10 protein of Autographa californica nucleopolyhedrovirus forms two distinct cytoskeletal-like structures and associates with polyhedral occlusion bodies during infection. Virology. 2008;371:278–91. [PubMed: 17991504]27.Patmanidi AL, Possee RD, King LA. Formation of P10 tubular structures during AcMNPV infection depends on the integrity of host-cell microtubules. Virology. 2003;317:308–20. [PubMed: 14698669]28.Vlak JM, Klinkenberg FA, Zaal KJM, Usmany M, Klinge-roode EC, Geervliet JBF, Roosien J, Lent JWV. Functional studies on the p10 gene of Autographa californica nuclear polyhedrosis virus using a recombinant expressing a p10-b-galactosidase fusion gene. J Gen Virol. 1988;69:765–776. [PubMed: 3128641]29.Williams GV, Rohel DZ, Kuzio J, Faulkner P. A cytopathological investigation of Autographa californica nuclear polyhedrosis virus p10 gene function using insertion/deletion mutants. J Gen Virol. 1989;70:187–202. [PubMed: 2659726]30.Graves LP, Hughes LC, Irons SL, Possee RD, King LA. In cultured cells the baculovirus P10 protein forms two independent intracellular structures that play separate roles in occlusion body maturation and their release by nuclear disintegration. PLoS Pathog. 2019:15. [PMC free article: PMC6557513] [PubMed: 31181119]31.van Oers MM, Flipsen JTM, Reusken CBEM, Sliwinsky EL, Goldbach RW, Vlak JM. Functional domains of the p10 protein of Autographa californica nuclear polyhedrosis virus. J Gen Virol. 1993;74:563–574. [PubMed: 8468550]32.Hashimoto Y, Hayakawa T, Ueno Y, Fugita T, Sano Y, Matsumoto T. Sequence analysis of the Plutella xylostella granulovirus genome. Virology. 2000;275:358–372. [PubMed: 10998336]33.Escasa SR, Lauzon HA, Mathur AC, Krell PJ, Arif BM. Sequence analysis of the Choristoneura occidentalis granulovirus genome. J Gen Virol. 2006;87:1917–33. [PubMed: 16760394]34.Lange M, Jehle JA. The genome of the Cryptophlebia leucotreta granulovirus. Virology. 2003;317:220–36. [PubMed: 14698662]35.Alaoui-Ismaili MH, Richardson CD. Insect virus proteins (FALPE and p10) self-associate to form filaments in infected cells. J Virol. 1998;72:2213–23. [PMC free article: PMC109518] [PubMed: 9499079]36.Lepore LS, Roelvink PR, Granados RR. Enhancin, the granulosis virus protein that facilitates nucleopolyhedrovirus (NPV) infections, is a metalloprotease. J Invertebr Pathol. 1996;68:131–40. [PubMed: 8858909]37.Hashimoto Y, Cosaro BG, Granados RR. Location and nucleotide sequence of the gene encoding the viral enhancing factor of the Trichoplusia ni granulosis virus. J Gen Virol. 1991;72:2645–2651. [PubMed: 1940861]38.Slavicek JM, Popham HJ. The Lymantria dispar nucleopolyhedrovirus enhancins are components of occlusion-derived virus. J Virol. 2005;79:10578–88. [PMC free article: PMC1182639] [PubMed: 16051850]39.Hayakawa T, Ko R, Okano K, Seong S, Goto C, Maeda S. Sequence analysis of the Xestia c-nigrum granulovirus genome. Virology. 1999;262:277–297. [PubMed: 10502508]40.Popham HJ, Bischoff DS, Slavicek JM. Both Lymantria dispar nucleopolyhedrovirus enhancin genes contribute to viral potency. J Virol. 2001;75:8639–48. [PMC free article: PMC115109] [PubMed: 11507209]41.Wang P, Granados RR. An intestinal mucin is the target substrate for a baculovirus enhancin. Proc Natl Acad Sci U S A. 1997;94:6977–82. [PMC free article: PMC21270] [PubMed: 9192677]42.Galloway CS, Wang P, Winstanley D, Jones IM. Comparison of the bacterial Enhancin-like proteins from Yersinia and Bacillus spp. with a baculovirus Enhancin. J Invertebr Pathol. 2005;90:134–7. [PubMed: 16081094]43.Kozlov EA, Sidorova NM, Serebryani SB. Proteolytic cleavage of polyhedral protein during dissolution of inclusion bodies of the nuclear polyhedrosis viruses of Bombyx mori and Galleria mellonella under alkaline conditions. J Invertebr Pathol. 1975;25:97–101. [PubMed: 1089735]44.Kozlov EA, Levitina TL, Gusak NM, Serebryani SB. Comparison of the amino acid sequences of inclusions body proteins of Bombyx mori, Porthetria dispar, and Galleria mellonella. Biorganic Khimia. 1981;7:1008–1015.45.Serebryani SB, Levitina TL, Kautsman ML, Radavski YL, Gusak NM, Ovander MN, Sucharenko NV, Kozlov EA. The primary structure of the polyhedral protein of nuclear polyhedrosis virus (NPV) of Bombyx mori. J Invertebr Pathol. 1977;30:442–443.46.Rubinstein R, Polson A. Midgut and viral associated proteases of Heliothis armigera. Intervirology. 1983;19:16–25. [PubMed: 6337972]47.McCarthy WJ, DiCapua RA. Characterization of solubilized proteins from tissue culture and host-derived nuclear polyhedra of Lymantria dispar and Autographa californica. Intervirology. 1979;11:174–181. [PubMed: 372133]48.Wood HA. Autographa californica nuclear polyhedrosis virus-induced proteins in tissue culture. Virology. 1980;102:21–27. [PubMed: 18631644]49.Zummer M, Faulkner P. Absence of protease in baculovirus polyhedral bodies propagated in vitro. J Invertebr Pathol. 1979;33:383–384.50.Slack JM, Kuzio J, Faulkner P. Characterization of v-cath, a cathepsin L-like proteinase expressed by the baculovirus Autographa californica multiple nuclear polyhedrosis virus. J Gen Virol. 1995;76:1091–1098. [PubMed: 7730794]51.Harrison SC. Viral membrane fusion. Virology. 2015;479-480:498–507. [PMC free article: PMC4424100] [PubMed: 25866377]52.Dong S, Wang M, Qiu Z, Deng F, Vlak JM, Hu Z, Wang H. Autographa californica multicapsid nucleopolyhedrovirus efficiently infects Sf9 cells and transduces mammalian cells via direct fusion with the plasma membrane at low pH. J Virol. 2010;84:5351–9. [PMC free article: PMC2863812] [PubMed: 20219938]53.Blissard GW, Wenz JR. Baculovirus gp64 envelope glycoprotein is sufficient to mediate pH-dependent membrane fusion. J Virol. 1992;66:6829–6835. [PMC free article: PMC240187] [PubMed: 1404622]54.Oomens AGP, Monsma SA, Blissard GW. The baculovirus gp64 envelope fusion protein: synthesis, oligomerization, and processing. Virology. 1995:592–603. [PubMed: 7778291]55.Monsma SA, Oomens AGP, Blissard GW. The gp64 envelope fusion protein is an essential baculovirus protein required for cell-to-cell transmission of infection. J Virol. 1996;70:4607–4616. [PMC free article: PMC190397] [PubMed: 8676487]56.Kuzio J, Pearson MN, Harwood SH, Funk CJ, Evans JT, Slavicek J, Rohrmann GF. Sequence and analysis of the genome of a baculovirus pathogenic for Lymantria dispar. Virology. 1999;253:17–34. [PubMed: 9887315]57.Ijkel WFJ, van Strien EA, Jeldens JGM, Broer R, Zuidema D, Goldbach RW, Vlak JM. Sequence and organization of the Spodoptera exigua multicapsid nucleopolyhedrovirus genome. J Gen Virol. 1999;80:3289–3304. [PubMed: 10567663]58.Roberts TE, Faulkner P. Fatty acid acylation of the 67K envelope glycoprotein of a baculovirus: Autographa californica nuclear polyhedrosis virus. Virology. 1989;172:377–381. [PubMed: 2672565]59.Hohmann AW, Faulkner P. Monoclonal antibodies to baculovirus structural proteins: determination of specificities by western blot analysis. Virology. 1983;125:432–444. [PubMed: 6340331]60.Whitford M, Stewart S, Kuzio J, Faulkner P. Identification and sequence analysis of a gene encoding gp67 an abundant envelope glycoprotein of the baculovirus, Autographa californica nuclear polyhedrosis virus. J Virol. 1989;63:1393–1399. [PMC free article: PMC247838] [PubMed: 2644449]61.Volkman LE. l986. The 64K envelope protein of budded Autographa californica nuclear polyhedrosis virus. Curr Top Microbiol Immunol. 131:103–118. [PubMed: 3545692]62.Wang R, Deng F, Hou D, Zhao Y, Guo L, Wang H, Hu Z. Proteomics of the Autographa californica Nucleopolyhedrovirus Budded Virions. J Virol. 2010;84:7233–7242. [PMC free article: PMC2898249] [PubMed: 20444894]63.Oomens AG, Blissard GW. Requirement for GP64 to drive efficient budding of Autographa californica multicapsid nucleopolyhedrovirus. Virology. 1999;254:297–314. [PubMed: 9986796]64.Ardisson-Araujo DM, Pereira BT, Melo FL, Ribeiro BM, Bao SN. A betabaculovirus encoding a gp64 homolog. BMC Genomics. 2016;17:94. de AZPM, Moscardi F, Kitajima EW, Sosa-Gomez DR, Wolff JL. [PMC free article: PMC4741009] [PubMed: 26847652]65.Pearson MN, Rohrmann GF. Transfer, incorporation, and substitution of envelope fusion proteins among members of the Baculoviridae, Orthomyxoviridae, and Metaviridae (insect retrovirus) families. J Virol. 2002;76:5301–5304. [PMC free article: PMC137044] [PubMed: 11991958]66.Ardisson-Araujo DM, Melo FL, Clem RJ, Wolff JL, Ribeiro BM. A Betabaculovirus-Encoded gp64 Homolog Codes for a Functional Envelope Fusion Protein. J Virol. 2016;90:1668–72. [PMC free article: PMC4719602] [PubMed: 26537678]67.Cook JD, Soto-Montoya H, Korpela MK, Lee JE. Electrostatic Architecture of the Infectious Salmon Anemia Virus (ISAV) Core Fusion Protein Illustrates a Carboxyl-Carboxylate pH Sensor. J Biol Chem. 2015;290:18495–504. [PMC free article: PMC4513110] [PubMed: 26082488]68.Cook JD, Sultana A, Lee JE. Structure of the infectious salmon anemia virus receptor complex illustrates a unique binding strategy for attachment. Proc Natl Acad Sci U S A. 2017;114:E2929–E2936. [PMC free article: PMC5389325] [PubMed: 28320973]69.Peng R, Zhang S, Cui Y, Shi Y, Gao GF, Qi J. Structures of human-infecting Thogotovirus fusogens support a common ancestor with insect baculovirus. Proc Natl Acad Sci U S A. 2017;114:E8905–E8912. [PMC free article: PMC5651748] [PubMed: 29073031]70.Kadlec J, Loureiro S, Abrescia NG, Stuart DI, Jones IM. The postfusion structure of baculovirus gp64 supports a unified view of viral fusion machines. Nat Struct Mol Biol. 2008;10:1024–30. [PubMed: 18776902]71.Dong S, Blissard GW. Functional analysis of the Autographa californica multiple nucleopolyhedrovirus GP64 terminal fusion loops and interactions with membranes. J Virol. 2012;86:9617–28. [PMC free article: PMC3446601] [PubMed: 22740400]72.Yu Q, Blissard GW, Liu TX, Li Z. Autographa californica multiple nucleopolyhedrovirus GP64 protein: Analysis of domain I and V amino acid interactions and membrane fusion activity. Virology. 2016;488:259–70. [PubMed: 26655244]73.Jiang Y, Deng F, Rayner S, Wang H, Hu Z. Evidence of a major role of GP64 in group I alphabaculovirus evolution. Virus Res. 2009;142:85–91. [PubMed: 19428740]74.Wang M, Yin F, Shen S, Tan Y, Deng F, Vlak JM, Hu Z, Wang H. Partial Functional Rescue of HearNPV Infectivity by Substitution of F Protein with GP64 from AcMNPV. J Virol. 2010;84:11505–14. [PMC free article: PMC2953197] [PubMed: 20739531]75.Wang M, Wang J, Yin F, Tan Y, Deng F, Chen X, Jehle JA, Vlak JM, Hu Z, Wang H. Unraveling the entry mechanism of baculoviruses and its evolutionary implications. J Virol. 2014;88:2301–11. [PMC free article: PMC3911565] [PubMed: 24335309]76.Shen S, Wang M, Li X, Li S, van Oers MM, Vlak JM, Braakman I, Hu Z, Deng F, Wang H. Mutational and functional analysis of N-linked glycosylation of envelope fusion protein F of Helicoverpa armigera nucleopolyhedrovirus. J Gen Virol. 2016;97:988–99. [PubMed: 26769631]77.Lung O, Westenberg M, Vlak JM, Zuidema D, Blissard GW. Pseudotyping Autographa californica multicapsid nucleopolyhedrovirus (AcMNPV): F proteins from group II NPVs are functionally analogous to AcMNPV GP64. J Virol. 2002;76:5729–36. [PMC free article: PMC137061] [PubMed: 11992001]78.Wang M, Tan Y, Yin F, Deng F, Vlak JM, Hu Z, Wang H. The F-like protein Ac23 enhances the infectivity of the budded virus of gp64-null AcMNPV pseudotyped with baculovirus envelope fusion protein F. J Virol. 2008;82:9800–4. [PMC free article: PMC2546973] [PubMed: 18653446]79.Pearson MN, Groten C, Rohrmann GF. Identification of the Lymantria dispar nucleopolyhedrovirus envelope fusion protein provides evidence for a phylogenetic division of the Baculoviridae. J Virol. 2000;74:6126–6131. [PMC free article: PMC112111] [PubMed: 10846096]80.Ijkel WFJ, Westernberg M, Goldbach RW, Blissard GW, Vlak JM, Zuidema D. A novel baclovirus envelope fusion protein with a proprotein convertase cleavage site. Virology. 2000;274:30–41. [PubMed: 11017785]81.Westenberg M, Soedling HM, Hirani N, Nicholson LJ, Mann DA, Dolphin CT. Seamless replacement of Autographa californica multiple nucleopolyhedrovirus gp64 with each of five novel type II alphabaculovirus fusion sequences generates pseudotyped virus that fails to transduce mammalian cells. J Gen Virol. 2012;93:1583–90. [PMC free article: PMC3542734] [PubMed: 22492915]82.Yin F, Wang M, Tan Y, Deng F, Vlak JM, Hu Z, Wang H. A functional F analogue of AcMNPV GP64 from the Agrotis segetum Granulovirus. J Virol. 2008;82:8922–6. [PMC free article: PMC2519692] [PubMed: 18562524]83.Yin F, Wang M, Tan Y, Deng F, Vlak JM, Hu Z, Wang H. 2013. Betabaculovirus F proteins showed different efficiencies when rescuing the infectivity of gp64-null Autographa californica nucleopolyhedrovirus. Virology 436:2013 Feb 5;436(1):59-66. [PubMed: 23245471]84.Eckert DM, Kim PS. Mechanisms of viral membrane fusion and its inhibition. Annu Rev Biochem. 2001;70:777–810. [PubMed: 11395423]85.Skehel J, Wiley D. Receptor binding and membrane fusion in virus entry: the influenza hemagglutinin. Annu Rev Biochem. 2000;69:531–69. [PubMed: 10966468]86.Rohrmann GF, Karplus PA. Relatedness of baculovirus and gypsy retrotransposon envelope proteins. BMC Evol Biol. 2001;1:1. [PMC free article: PMC29073] [PubMed: 11244578]87.Malik HS, Henikoff S. Positive Selection of Iris, a Retroviral Envelope-Derived Host Gene in Drosophila melanogaster. PLoS Genet. 2005;1:429–43. [PMC free article: PMC1262188] [PubMed: 16244705]88.Soding J. Protein homology detection by HMM-HMM comparison. Bioinformatics. 2005;21:951–60. [PubMed: 15531603]89.Gresikova M, Nosek J, Ciampor F, Sekeyova M, Turek R. Isolation of paramyxovirus type 4 from Oeciacus hirundinis bugs. Acta Virol. 1980;24:222–3. [PubMed: 6107039]90.Pearson MN, Russell R, Rohrmann GF. Characterization of a baculovirus encoded protein that is associated with infected-cell membranes and budded virions. Virology. 2001;291:22–31. [PubMed: 11878873]91.Lung OY, Cruz-Alvarez M, Blissard GW. Ac23, an envelope fusion protein homolog in the baculovirus Autographa californica multicapsid nucleopolyhedrovirus, is a viral pathogenicity factor. J Virol. 2003;77:328–39. [PMC free article: PMC140606] [PubMed: 12477838]92.Zhou J, Blissard GW. Identification of a GP64 subdomain involved in receptor binding by budded virions of the baculovirus AcMNPV. J Virol. 2008;82:4449–60. [PMC free article: PMC2293031] [PubMed: 18287233]93.Braunagel SC, Russell WK, Rosas-Acosta G, Russell DH, Summers MD. Determination of the protein composition of the occlusion-derived virus of Autographa californica nucleopolyhedrovirus. Proc Natl Acad Sci U S A. 2003;100:9797–802. [PMC free article: PMC187845] [PubMed: 12904572]94.Perera O, Green TB, Stevens SM Jr, White S, Becnel JJ. Proteins associated with Culex nigripalpus nucleopolyhedrovirus occluded virions. J Virol. 2007;81:4585–90. [PMC free article: PMC1900190] [PubMed: 17301145]95.Deng F, Wang R, Fang M, Jiang Y, Xu X, Wang H, Chen X, Arif BM, Guo L, Wang H, Hu Z. Proteomics analysis of Helicoverpa armigera single nucleocapsid nucleopolyhedrovirus identified two new occlusion-derived virus-associated proteins, HA44 and HA100. J Virol. 2007;81:9377–85. [PMC free article: PMC1951453] [PubMed: 17581982]96.Hou D, Zhang L, Deng F, Fang W, Wang R, Liu X, Guo L, Rayner S, Chen X, Wang H, Hu Z. Comparative Proteomics Reveal Fundamental Structural and Functional Differences between the Two Progeny Phenotypes of a Baculovirus. J Virol. 2013;87:829–39. [PMC free article: PMC3554090] [PubMed: 23115289]97.Carbonell LF, Miller LK. Baculovirus interaction with nontarget organisms: a virus-borne reporter gene is not expressed in two mammalian cell lines. Appl Environ Microbiol. 1987;53:1412–1417. [PMC free article: PMC203885] [PubMed: 3116925]98.Vanarsdall AL, Mikhailov VS, Rohrmann GF. Characterization of a baculovirus lacking the DBP (DNA-binding protein) gene. Virology. 2007;364:475–85. [PMC free article: PMC2697660] [PubMed: 17449080]99.Hefferon KL, Oomens AG, Monsma SA, Finnerty CM, Blissard GW. Host cell receptor binding by baculovirus GP64 and kinetics of virion entry. Virology. 1999;258:455–68. [PubMed: 10366584]100.Hofmann C, Sandig V, Jennings G, Rudolph M, Schlag P, Strauss M. Efficient gene transfer into human hepatocytes by baculovirus vectors. Proc Natl Acad Sci U S A. 1995;92:10099–103. [PMC free article: PMC40743] [PubMed: 7479733]101.Tani H, Nishijima M, Ushijima H, Miyamura T, Matsuura Y. Characterization of cell-surface determinants important for baculovirus infection. Virology. 2001;279:343–53. [PubMed: 11145915]102.Kamiya K, Kobayashi J, Yoshimura T, Tsumoto K. Confocal microscopic observation of fusion between baculovirus budded virus envelopes and single giant unilamellar vesicles. Biochim Biophys Acta. 2010;1798:1625–1631. [PubMed: 20493165]103.Kataoka C, Kaname Y, Taguwa S, Abe T, Fukuhara T, Tani H, Moriishi K, Matsuura Y. Baculovirus GP64-mediated entry into mammalian cells. J Virol. 2012;86:2610–20. [PMC free article: PMC3302255] [PubMed: 22190715]104.O'Flynn NM, Patel A, Kadlec J, Jones IM. Improving promiscuous mammalian cell entry by the baculovirus Autographa californica multiple nuclear polyhedrosis virus. Biosci Rep. 2012;33:23–36. [PMC free article: PMC3522474] [PubMed: 23035899]105.Westenberg M, Uijtdewilligen P, Vlak JM. Baculovirus envelope fusion proteins F and GP64 exploit distinct receptors to gain entry into cultured insect cells. J Gen Virol. 2007;88:3302–6. [PubMed: 18024899]106.Liang C, Song J, Chen X. The GP64 protein of Autographa californica multiple nucleopolyhedrovirus rescues Helicoverpa armigera nucleopolyhedrovirus transduction in mammalian cells. J Gen Virol. 2005;86:1629–35. [PubMed: 15914840]107.Kikhno I, Guitiérrez S, Croizier L, Croizier G, Lopéz-Ferber ML. Characterization of pif, a gene required for the per os infectivity of Spodoptera littoralis nucleopolyhedrovirus. J Gen Virol. 2002;83:3013–3022. [PubMed: 12466478]108.Boogaard B, van Oers MM, van Lent JWM. An Advanced View on Baculovirus per Os Infectivity Factors. Insects. 2018:9. [PMC free article: PMC6164829] [PubMed: 30018247]109.Zheng Q, Shen Y, Kon X, Zhang J, Feng M, Wu X. Protein-protein interactions of the baculovirus per os infectivity factors (PIFs) in the PIF complex. J Gen Virol. 2017;98:853–861. [PubMed: 28141488]110.Braunagel SC, Summers MD. Molecular Biology of the Baculovirus Occlusion-Derived Virus Envelope. Curr Drug Targets. 2007;8:1084–1095. [PubMed: 17979668]111.Beniya H, Braunagel SC, Summers MD. Autographa californica nuclear polyhedrosis virus: subcellular localization and protein trafficking of BV/ODV-E26 to intranuclear membranes and viral envelopes. Virology. 1998;240:64–75. [PubMed: 9448690]112.Imai N, Kurihara M, Matsumoto S, Kang WK. Bombyx mori nucleopolyhedrovirus orf8 encodes a nucleic acid binding protein that colocalizes with IE1 during infection. Arch Virol. 2004;149:1581–94. [PubMed: 15290382]113.Burks JK, Summers MD, Braunagel SC. BV/ODV-E26: a palmitoylated, multifunctional structural protein of Autographa californica nucleopolyhedrovirus. Virology. 2007;361:194–203. [PubMed: 17169392]114.O'Reilly DR, Passarelli AL, Goldman IF, Miller LK. Characterization of the DA26 gene in a hypervariable region of the Autographa californica nuclear polyhedrosis virus genome. J Gen Virol. 1990;71(Pt 5):1029–37. [PubMed: 2189022]115.Nie Y, Theilmann DA. Deletion of AcMNPV AC16 and AC17 results in delayed viral gene expression in budded virus infected cells but not transfected cells. Virology. 2010;404:168–79. [PubMed: 20627351]116.Nie Y, Fang M, Theilmann DA. AcMNPV AC16 (DA26, BV/ODV-E26) regulates the levels of IE0 and IE1 and binds to both proteins via a domain located within the acidic transcriptional activation domain. Virology. 2009;385:484–95. [PubMed: 19150105]117.Yuan M, Huang Z, Wei D, Hu Z, Yang K, Pang Y. Identification of Autographa californica nucleopolyhedrovirus ac93 as a core gene and its requirement for intranuclear microvesicle formation and nuclear egress of nucleocapsids. J Virol. 2011;85:11664–74. [PMC free article: PMC3209287] [PubMed: 21880748]118.Russell RLQ, Rohrmann GF. A 25 kilodalton protein is associated with the envelopes of occluded baculovirus virions. Virology. 1993;195:532–540. [PubMed: 8337828]119.Hou D, Zhang L, Deng F, Fang W, Wang R, Liu X, Guo L, Rayner S, Chen X, Wang H, Hu Z. 2012. Comparative Proteomics Reveal Fundamental Structural and Functional Differences between the Two Progeny Phenotypes of a Baculovirus. J Virol doi:JVI.02329-12 [pii] 10.1128/JVI.02329-12. [PMC free article: PMC3554090] [PubMed: 23115289] [CrossRef]120.Shrestha A, Bao K, Chen YR, Chen W, Wang P, Fei Z, Blissard GW. Global Analysis of Baculovirus Autographa californica Multiple Nucleopolyhedrovirus Gene Expression in the Midgut of the Lepidopteran Host Trichoplusia ni. J Virol. 2018:92. [PMC free article: PMC6232492] [PubMed: 30209166]121.Hong T, Summers M, Braunagel S. N-terminal sequences from Autographa californica nuclear polyhedrosis virus envelope proteins ODV-E66 and ODV-E25 are sufficient to direct reporter proteins to the nuclear envelope, intranuclear microvesicles and the envelope of occlusion derived virus. Proc Natl Acad Sci U S A. 1997;8:4050–4055. [PMC free article: PMC20566] [PubMed: 9108103]122.Guo YJ, Fu SH, Li LL. Autographa californica multiple nucleopolyhedrovirus ac75 is required for egress of nucleocapsids from the nucleus and formation of de novo intranuclear membrane microvesicles. PLoS One. 2017;12:e0185630. [PMC free article: PMC5624611] [PubMed: 28968422]123.Chen L, Hu X, Xiang X, Yu S, Yang R, Wu X. Autographa californica multiple nucleopolyhedrovirus odv-e25 (Ac94) is required for budded virus infectivity and occlusion-derived virus formation. Arch Virol. 2012;157:617–25. [PubMed: 22218963]124.Zhu M, Wang J, Deng R, Xiong P, Liang H, Wang X. A MicroRNA Encoded by Autographa californica Nucleopolyhedrovirus Regulates Expression of Viral Gene ODV-E25. J Virol. 2013;87:13029–34. [PMC free article: PMC3838118] [PubMed: 24027316]125.Zhu M, Wang J, Deng R, Wang X. Functional Regulation of an Autographa californica Nucleopolyhedrovirus-Encoded MicroRNA, AcMNPV-miR-1, in Baculovirus Replication. J Virol. 2016;90:6526–6537. [PMC free article: PMC4936140] [PubMed: 27147751]126.Luo XC, Wang SS, Zhang J, Qian DD, Wang SM, Li LL. Effects of Early or Overexpression of the Multiple Nucleopolyhedrovirus (ODV-E25) on Virus Replication. PLoS One. 2013;8:e65635. [PMC free article: PMC3688825] [PubMed: 23825525]127.Chen L, Yang R, Hu X, Xiang X, Yu S, Wu X. The formation of occlusion-derived virus is affected by the expression level of ODV-E25. Virus Res. 2013;173:404–14. [PubMed: 23298549]128.Fang M, Wang H, Wang H, Yuan L, Chen X, Vlak JM, Hu Z. Open reading frame 94 of Helicoverpa armigera single nucleocapsid nucleopolyhedrovirus encodes a novel conserved occlusion-derived virion protein, ODV-EC43. J Gen Virol. 2003;84:3021–7. [PubMed: 14573807]129.Fang M, Nie Y, Theilmann DA. Deletion of the AcMNPV core gene ac109 results in budded virions that are non-infectious. Virology. 2009;389:66–74. [PubMed: 19411088]130.Lin L, Wang J, Deng R, Ke J, Wu H, Wang X. ac109 is required for the nucleocapsid assembly of Autographa californica multiple nucleopolyhedrovirus. Virus Res. 2009;144:130–5. [PubMed: 19393701]131.Lehiy CJ, Wu W, Berretta MF, Passarelli AL. Autographa californica M nucleopolyhedrovirus open reading frame 109 affects infectious budded virus production and nucleocapsid envelopment in the nucleus of cells. Virology. 2013;435:442–52. [PubMed: 23149091]132.Alfonso V, Maroniche GA, Reca SR, Lopez MG, Del Vas M, Taboga O. AcMNPV Core Gene ac109 Is Required for Budded Virion Transport to the Nucleus and for Occlusion of Viral Progeny. PLoS One. 2012;7:e46146. [PMC free article: PMC3458853] [PubMed: 23049963]133.Braunagel SC, He H, Ramamurthy P, Summers MD. Transcription, translation, and cellular localization of three Autographa californica nuclear polyhedrosis virus structural proteins: ODV-E18, ODV-E35 and ODV-EC27. Virology. 1996;222:100–114. [PubMed: 8806491]134.McCarthy CB, Theilmann DA. AcMNPV ac143(odv-e18) is essential for mediating budded virus production and is the 30th baculovirus core gene. Virology. 2008;375:277–91. [PubMed: 18328526]135.Zimmermann L, Stephens A, Nam SZ, Rau D, Kubler J, Lozajic M, Gabler F, Soding J, Lupas AN, Alva V. A Completely Reimplemented MPI Bioinformatics Toolkit with a New HHpred Server at its Core. J Mol Biol. 2018;430:2237–2243. [PubMed: 29258817]136.Wang X, Shang Y, Chen C, Liu S, Chang M, Zhang N, Hu H, Zhang F, Zhang T, Wang Z, Liu X, Lin Z, Deng F, Wang H, Zou Z, Vlak JM, Wang M, Hu Z. Baculovirus per os Infectivity Factor Complex: Components and Assembly. J Virol 2019 [PMC free article: PMC6401453] [PubMed: 30602603] [CrossRef]137.Wang Y, Kleespies RG, Huger AM, Jehle JA. The genome of Gryllus bimaculatus nudivirus indicates an ancient diversification of baculovirus-related nonoccluded nudiviruses of insects. J Virol. 2007;81:5395–406. [PMC free article: PMC1900193] [PubMed: 17360757]138.Ohkawa T, Washburn JO, Sitapara R, Sid E, Volkman LE. Specific binding of Autographa californica M nucleopolyhedrovirus occlusion-derived virus to midgut cells of Heliothis virescens larvae is mediated by products of pif genes Ac119 and Ac022 but not by Ac115. J Virol. 2005;79:15258–64. [PMC free article: PMC1316039] [PubMed: 16306597]139.Peng K, van Oers MM, Hu Z, van Lent JW, Vlak JM. Baculovirus per os infectivity factors form a complex on the surface of occlusion-derived virus. J Virol. 2010;84:9497–504. [PMC free article: PMC2937639] [PubMed: 20610731]140.Li X, Song J, Jiang T, Liang C, Chen X. The N-terminal hydrophobic sequence of Autographa californica nucleopolyhedrovirus PIF-3 is essential for oral infection. Arch Virol. 2007;152:1851–8. [PubMed: 17585368]141.Peng K, van Lent JW, Boeren S, Fang M, Theilmann DA, Erlandson MA, Vlak JM, van Oers MM. Characterization of novel components of the baculovirus per os infectivity factor complex. J Virol. 2012;86:4981–8. [PMC free article: PMC3347349] [PubMed: 22379094]142.Zhou W, Yao L, Xu H, Yan F, Qi Y. The function of envelope protein p74 from Autographa californica M nucleopolyhedrovirus in primary infection to host. Virus Genes. 2005;30:139–150. [PubMed: 15744572]143.Russell RL, Rohrmann GF. Characterization of P91, a protein associated with virions of an Orgyia pseudotsugata baculovirus. Virology. 1997;233:210–23. [PubMed: 9201231]144.Ono C, Kamagata T, Taka H, Sahara K, Asano S, Bando H. Phenotypic grouping of 141 BmNPVs lacking viral gene sequences. Virus Res. 2012;165:197–206. [PubMed: 22421381]145.Zhu S, Wang W, Wang Y, Yuan M, Yang K. The baculovirus core gene ac83 is required for nucleocapsid assembly and per os infectivity of Autographa californica nucleopolyhedrovirus. J Virol. 2013;87:10573–86. [PMC free article: PMC3807414] [PubMed: 23864639]146.Javed MA, Biswas S, Willis LG, Harris S, Pritchard C, van Oers MM, Donly BC, Erlandson MA, Hegedus DD, Theilmann DA. Autographa californica Multiple Nucleopolyhedrovirus AC83 is a Per Os Infectivity Factor (PIF) Protein Required for Occlusion-Derived Virus (ODV) and Budded Virus Nucleocapsid Assembly as well as Assembly of the PIF Complex in ODV Envelopes. J Virol. 2017;91:02115–16. [PMC free article: PMC5309931] [PubMed: 28031365]147.Huang Z, Pan M, Zhu S, Zhang H, Wu W, Yuan M, Yang K. The Autographa californica Multiple Nucleopolyhedrovirus ac83 Gene Contains a cis-Acting Element That Is Essential for Nucleocapsid Assembly. J Virol. 2017:91. [PMC free article: PMC5309959] [PubMed: 28031366]148.Hong T, Braunagel SC, Summers MD. Transcription, translation, and cellular localization of PDV-E66: A structural protein of the PDV envelope of Autographa californica nuclear polyhedrosis virus. Virology. 1994;204:210–224. [PubMed: 8091653]149.Sugiura N, Setoyama Y, Chiba M, Kimata K, Watanabe H. Baculovirus envelope protein ODV-E66 is a novel chondroitinase with distinct substrate specificity. J Biol Chem. 2011;286:29026–34. [PMC free article: PMC3190710] [PubMed: 21715327]150.Xiang X, Chen L, Hu X, Yu S, Yang R, Wu X. Autographa californica multiple nucleopolyhedrovirus odv-e66 is an essential gene required for oral infectivity. Virus Res. 2011;158:72–8. [PubMed: 21440017]151.Sugiura N, Ikeda M, Shioiri T, Yoshimura M, Kobayashi M, Watanabe H. Chondroitinase from baculovirus Bombyx mori nucleopolyhedrovirus and chondroitin sulfate from silkworm Bombyx mori. Glycobiology. 2013;23:1520–30. [PubMed: 24052236]152.Dinglasan RR, Alaganan A, Ghosh AK, Saito A, van Kuppevelt TH, Jacobs-Lorena M. Plasmodium falciparum ookinetes require mosquito midgut chondroitin sulfate proteoglycans for cell invasion. Proc Natl Acad Sci U S A. 2007;104:15882–7. [PMC free article: PMC2000438] [PubMed: 17873063]153.Vigdorovich V, Miller AD, Strong RK. Ability of hyaluronidase 2 to degrade extracellular hyaluronan is not required for its function as a receptor for jaagsiekte sheep retrovirus. J Virol. 2007;81:3124–9. [PMC free article: PMC1866058] [PubMed: 17229709]154.Kawaguchi Y, Sugiura N, Onishi M, Kimata K, Kimura M, Kakuta Y. Crystallization and X-ray diffraction analysis of chondroitin lyase from baculovirus: envelope protein ODV-E66. Acta Crystallogr Sect F Struct Biol Cryst Commun. 2012;68:190–2. [PMC free article: PMC3274400] [PubMed: 22297996]155.Lapointe R, Popham HJ, Straschil U, Goulding D, O'Reilly DR, Olszewski JA. Characterization of two Autographa californica nucleopolyhedrovirus proteins, Ac145 and Ac150, which affect oral infectivity in a host-dependent manner. J Virol. 2004;78:6439–48. [PMC free article: PMC416519] [PubMed: 15163737]156.Dall D, Luque T, O'Reilly DR. Insect-virus relationships: sifting by informatics. Bioessays. 2001;23:184–193. [PubMed: 11169592]157.Zhang JH, Ohkawa T, Washburn JO, Volkman LE. Effects of Ac150 on virulence and pathogenesis of Autographa californica multiple nucleopolyhedrovirus in noctuid hosts. J Gen Virol. 2005;86:1619–27. [PubMed: 15914839]158.Olszewski J, Miller LK. Identification and characterization of a baculovirus structural protein, VP1054, required for nucleocapsid formation. J Virol. 1997;71:5040–5050. [PMC free article: PMC191737] [PubMed: 9188569]159.Guan Z, Zhong L, Li C, Wu W, Yuan M, Yang K. The Autographa californica Multiple Nucleopolyhedrovirus ac54 Gene Is Crucial for Localization of the Major Capsid Protein VP39 at the Site of Nucleocapsid Assembly. J Virol. 2016;90:4115–4126. [PMC free article: PMC4810565] [PubMed: 26865720]160.Marek M, Romier C, Galibert L, Merten O, van Oers MM. Baculovirus VP1054 is an acquired cellular PURα, a nucleic-acid6n binding protein specific for GGN repeats. J Virol. 2013;87:8465–80. [PMC free article: PMC3719801] [PubMed: 23720732]161.Ke J, Wang J, Deng R, Wang X. Autographa californica multiple nucleopolyhedrovirus ac66 is required for the efficient egress of nucleocapsids from the nucleus, general synthesis of preoccluded virions and occlusion body formation. Virology. 2008;374:421–31. [PubMed: 18241908]162.Biswas S, Willis LG, Fang M, Nie Y, Theilmann DA. Autographa californica Nucleopolyhedrovirus AC141 (Exon0), a Potential E3 Ubiquitin Ligase, Interacts with Viral Ubiquitin and AC66 To Facilitate Nucleocapsid Egress. J Virol. 2018:92. [PMC free article: PMC5774878] [PubMed: 29142135]163.Feng G, Krell PJ. Autographa californica multiple nucleopolyhedrovirus DNA polymerase C terminus is required for nuclear localization and viral DNA replication. J Virol. 2014;88:10918–33. [PMC free article: PMC4178887] [PubMed: 25008932]164.McLachlin JR, Miller LK. Identification and characterization of vlf-1, a baculovirus gene involved in very late gene expression. J Virol. 1994;68:7746–7756. [PMC free article: PMC237236] [PubMed: 7966564]165.Yang S, Miller LK. Activation of baculovirus very late promoters by interaction with very late factor 1. J Virol. 1999;73:3404–9. [PMC free article: PMC104104] [PubMed: 10074194]166.Yang S, Miller LK. Expression and mutational analysis of the baculovirus very late factor 1 (vlf-1) gene. Virology. 1998;245:99–109. [PubMed: 9614871]167.Vanarsdall AL, Okano K, Rohrmann GF. Characterization of the role of VLF-1 in baculovirus capsid structure and DNA processing. J Virol. 2006;80:1724–1733. [PMC free article: PMC1367162] [PubMed: 16439529]168.Whitford M, Faulkner P. A structural polypeptide of the baculovirus Autographa californica nuclear polyhedrosis virus contains 0-linked N-acetylglucosamine. J Virol. 1992;66:3324–3329. [PMC free article: PMC241110] [PubMed: 1583718]169.Whitford M, Faulkner P. Nucleotide sequence and transcriptional analysis of a gene encoding gp41, a structural glycoprotein of the baculovirus Autographa californica nuclear polyhedrosis virus. J Virol. 1992;66:4763–4768. [PMC free article: PMC241303] [PubMed: 1629955]170.Olszewski J, Miller LK. A role for baculovirus GP41 in budded virus production. Virology. 1997;233:292–301. [PubMed: 9217053]171.Li Y, Shen S, Hu L, Deng F, Vlak JM, Hu Z, Wang H, Wang M. The Functional Oligomeric State of Tegument Protein GP41 Is Essential for Baculovirus Budded Virion and Occlusion-Derived Virion Assembly. J Virol. 2018:92. [PMC free article: PMC5974493] [PubMed: 29643237]172.Danquah JO, Botchway S, Jeshtadi A, King LA. Direct interaction of baculovirus capsid proteins VP39 and EXON0 with kinesin-1 in insect cells determined by fluorescence resonance energy transfer-fluorescence lifetime imaging microscopy. J Virol. 2012;86:844–53. [PMC free article: PMC3255796] [PubMed: 22072745]173.Katsuma S, Kokusho R. A Conserved Glycine Residue Is Required for Proper Functioning of a Baculovirus VP39 Protein. J Virol. 2017:91. [PMC free article: PMC5331800] [PubMed: 28077638]174.Long CM, Rohrmann GF, Merrill GF. The conserved baculovirus protein p33 (Ac92) is a flavin adenine dinucleotide-linked sulfhydryl oxidase. Virology. 2009;388:231–5. [PubMed: 19409596]175.Wu W, Passarelli AL. Autographa californica M nucleopolyhedrovirus Ac92 (ORF92, P33) is required for budded virus production and multiply-enveloped occlusion-derived virus formation. J Virol. 2010;84:12351–61. [PMC free article: PMC2976406] [PubMed: 20861245]176.Prikhod'ko GG, Wang Y, Freulich E, Prives C, Miller LK. Baculovirus p33 binds human p53 and enhances p53-mediated apoptosis. J Virol. 1999;73:1227–34. [PMC free article: PMC103944] [PubMed: 9882325]177.Wu W, Clem RJ, Rohrmann GF, Passarelli AL. The baculovirus sulfhydryl oxidase Ac92 (P33) interacts with the Spodoptera frugiperda P53 protein and oxidizes it in vitro. Virology. 2013;447:197–207. [PMC free article: PMC3870475] [PubMed: 24210115]178.Nie Y, Fang M, Theilmann DA. Autographa californica multiple nucleopolyhedrovirus core gene ac92 (p33) is required for the efficient budded virus production. Virology. 2011;409:38–45. [PubMed: 20965540]179.Wu W, Lin T, Pan L, Yu M, Li Z, Pang Y, Yang K. Autographa californica multiple nucleopolyhedrovirus nucleocapsid assembly is interrupted upon deletion of the 38K gene. J Virol. 2006;80:11475–85. [PMC free article: PMC1642600] [PubMed: 16987976]180.Lai Q, Wu W, Li A, Wang W, Yuan M, Yang K. The 38K-Mediated Specific Dephosphorylation of the Viral Core Protein P6.9 Plays an Important Role in the Nucleocapsid Assembly of Autographa californica Multiple Nucleopolyhedrovirus. J Virol. 2018:92. [PMC free article: PMC5899202] [PubMed: 29444944]181.Wilson ME, Mainprize TH, Friesen PD, Miller LK. l987. Location, transcription, and sequence of a baculovirus gene encoding a small arginine-rich polypeptide. J Virol. 61:66l–666. [PMC free article: PMC254004] [PubMed: 3543402]182.Tweeten KA, Bulla LA, Consigli RA. Characterization of an extremely basic protein derived from granulosis virus nucleocapsid. J Virol. 1980;33:866–876. [PMC free article: PMC288612] [PubMed: 16789190]183.Balhorn R. A model for the structure of chromatin in mammalian sperm. J Cell Biol. 1982;93:298–305. [PMC free article: PMC2112839] [PubMed: 7096440]184.Nakano M, Kasai K, Yoshida K, Tanimoto T, Tamaki Y, Tobita T. Conformation of the fowl protamine, galline, and its binding properties to DNA. J Biochem. 1989;105:133–137. [PubMed: 2738040]185.Wilson ME, Price KH. Association of Autographa californica nuclear polyhedrosis virus (AcMNPV) with the nuclear matrix. Virology. 1988;167:233–241. [PubMed: 3055664]186.Wilson ME, Consigli RA. Functions of a protein kinase activity associated with purified capsids of the granulosis virus infecting Plodia interpunctella. Virology. 1985;143:526–535. [PubMed: 18639856]187.Miller LK, Adang MJ, Browne D. Protein kinase activity associated with the extracellular and occluded forms of the baculovirus Autographa californica NPV. J Virol. 1983;46:275–278. [PMC free article: PMC255117] [PubMed: 16789240]188.Li A, Zhao H, Lai Q, Huang Z, Yuan M, Yang K. Posttranslational Modifications of Baculovirus Protamine-Like Protein P6.9 and the Significance of Its Hyperphosphorylation for Viral Very Late Gene Hyperexpression. J Virol. 2015;89:7646–59. [PMC free article: PMC4505634] [PubMed: 25972542]189.Oppenheimer DI, Volkman LE. Proteolysis of p6.9 induced by cytochalasin D in Autographa californica M nuclear polyhedrosis virus-infected cells. Virology. 1995;207:1–11. [PubMed: 7871717]190.Wang M, Tuladhar E, Shen S, Wang H, van Oers MM, Vlak JM, Westenberg M. Specificity of baculovirus P6.9 basic DNA-binding proteins and critical role of the C terminus in virion formation. J Virol. 2010;84:8821–8. [PMC free article: PMC2919018] [PubMed: 20519380]191.Singh CP, Singh J, Nagaraju J. bmnpv-miR-3 facilitates BmNPV infection by modulating the expression of viral P6.9 and other late genes in Bombyx mori. Insect Biochem Mol Biol. 2014;49:59–69. [PubMed: 24698834]192.Braunagel SC, Guidry PA, Rosas-Acosta G, Engelking L, Summers MD. Identification of BV/ODV-C42, an Autographa californica nucleopolyhedrovirus orf101-encoded structural protein detected in infected-cell complexes with ODV-EC27 and p78/83. J Virol. 2001;75:12331–8. [PMC free article: PMC116129] [PubMed: 11711623]193.Wang Y, Wang Q, Liang C, Song J, Li N, Shi H, Chen X. Autographa californica multiple nucleopolyhedrovirus nucleocapsid protein BV/ODV-C42 mediates the nuclear entry of P78/83. J Virol. 2008;82:4554–61. [PMC free article: PMC2293022] [PubMed: 18287235]194.Wang Y, Zhang Y, Han S, Hu X, Zhou Y, Mu J, Pei R, Wu C, Chen X. Identification of a novel regulatory sequence of actin nucleation promoting factor encoded by Autographa californica multiple nucleopolyhedrovirus. J Biol Chem. 2015;290:9533–41. [PMC free article: PMC4392258] [PubMed: 25691574]195.Zhang Y, Hu X, Mu J, Hu Y, Zhou Y, Zhao H, Wu C, Pei R, Chen J, Chen X, Wang Y. Ac102 Participates in Nuclear Actin Polymerization by Modulating BV/ODV-C42 Ubiquitination during Autographa californica Multiple Nucleopolyhedrovirus Infection. J Virol. 2018:92. [PMC free article: PMC5974488] [PubMed: 29618641]196.Hepp SE, Borgo GM, Ticau S, Ohkawa T, Welch MD. Baculovirus AC102 Is a Nucleocapsid Protein That Is Crucial for Nuclear Actin Polymerization and Nucleocapsid Morphogenesis. J Virol. 2018:92. [PMC free article: PMC5952165] [PubMed: 29540600]197.Vanarsdall AL, Pearson MN, Rohrmann GF. Characterization of baculovirus constructs lacking either the Ac 101, Ac 142, or the Ac 144 open reading frame. Virology. 2007;367:187–95. [PMC free article: PMC2077081] [PubMed: 17585983]198.Fang M, Dai X, Theilmann DA. Autographa californica multiple nucleopolyhedrovirus EXON0 (ORF141) is required for efficient egress of nucleocapsids from the nucleus. J Virol. 2007;81:9859–69. [PMC free article: PMC2045402] [PubMed: 17626083]199.Fang M, Nie Y, Dai X, Theilmann DA. Identification of AcMNPV EXON0 (ac141) domains required for efficient production of budded virus, dimerization and association with BV/ODV-C42 and FP25. Virology. 2008;375:265–76. [PubMed: 18313716]200.Dai X, Stewart TM, Pathakamuri JA, Li Q, Theilmann DA. Autographa californica multiple nucleopolyhedrovirus exon0 (orf141), which encodes a RING finger protein, is required for efficient production of budded virus. J Virol. 2004;78:9633–44. [PMC free article: PMC514987] [PubMed: 15331696]201.Fang M, Nie Y, Theilmann DA. AcMNPV EXON0 (AC141) which is required for the efficient egress of budded virus nucleocapsids interacts with beta-tubulin. Virology. 2009;385:496–504. [PubMed: 19155039]202.Biswas S, Blissard GW, Theilmann DA. Trichoplusia ni Kinesin-1 Associates with Autographa californica Multiple Nucleopolyhedrovirus Nucleocapsid Proteins and Is Required for Production of Budded Virus. J Virol. 2016;90:3480–95. [PMC free article: PMC4794668] [PubMed: 26763996]203.Yang ZN, Xu HJ, Park EY, Zhang CX. 2008. Characterization of Bombyx mori nucleopolyhedrovirus with a deletion of bm118. Virus Res. [PubMed: 18462822]204.McCarthy CB, Dai X, Donly C, Theilmann DA. Autographa californica multiple nucleopolyhedrovirus ac142, a core gene that is essential for BV production and ODV envelopment. Virology. 2008;372:325–39. [PubMed: 18045640]205.Yue Q, Yu Q, Yang Q, Xu Y, Guo Y, Blissard GW, Li Z. Distinct Roles of Cellular ESCRT-I and ESCRT-III Proteins in Efficient Entry and Egress of Budded Virions of Autographa californica Multiple Nucleopolyhedrovirus. J Virol. 2018:92. [PMC free article: PMC5730794] [PubMed: 29046462]206.Belyavskyi M, Braunagel SC, Summers MD. The structural protein ODV-EC27 of Autographa californica nucleopolyhedrovirus is a multifunctional viral cyclin. Proc Natl Acad Sci U S A. 1998;95:11205–10. [PMC free article: PMC21620] [PubMed: 9736714]207.Vialard JE, Richardson CD. The 1,629-nucleotide open reading frame located downstream of the Autographa californica nuclear polyhedrosis virus polyhedrin gene encodes a nucleocapsid-associated phosphoprotein. J Virol. 1993;67:5859–5866. [PMC free article: PMC238004] [PubMed: 8371345]208.Russell RLQ, Funk CJ, Rohrmann GF. Association of a baculovirus encoded protein with the capsid basal region. Virology. 1997;227:142–152. [PubMed: 9007067]209.Goley ED, Ohkawa T, Mancuso J, Woodruff JB, D'Alessio JA, Cande WZ, Volkman LE, Welch MD. Dynamic nuclear actin assembly by Arp2/3 complex and a baculovirus WASP-like protein. Science. 2006;314:464–7. [PubMed: 17053146]210.Ohkawa T, Volkman LE, Welch MD. Actin-based motility drives baculovirus transit to the nucleus and cell surface. J Cell Biol. 2010;190:187–95. [PMC free article: PMC2930276] [PubMed: 20660627]211.Kitts PA, Possee RD. A method for producing recombinant baculovirus expression vectors at high frequency. BioTechniques. 1993;14:810–816. [PubMed: 8512707]212.Garavaglia MJ, Miele SA, Iserte JA, Belaich MN, Ghiringhelli PD. Ac53, ac78, ac101 and ac103 are newly discovered core genes in the family Baculoviridae. J Virol. 2012;86:12069–79. [PMC free article: PMC3486445] [PubMed: 22933288]213.Wu J, Miller LK. Sequence, transcription, and translation of a late gene of the Autographa californica nuclear polyhedrosis virus encoding a 34.8 K polypeptide. Journal of General Virology. 1989;70:2449–2459. [PubMed: 2674327]214.Cheng X, Krell P, Arif B. P34.8 (GP37) is not essential for baculovirus replication. J Gen Virol. 2001;82:299–305. [PubMed: 11161266]215.Li Z, Li C, Yang K, Wang L, Yin C, Gong Y, Pang Y. Characterization of a chitin-binding protein GP37 of Spodoptera litura multicapsid nucleopolyhedrovirus. Virus Res. 2003;96:113–22. [PubMed: 12951271]216.Liu X, Ma X, Lei C, Xiao Y, Zhang Z, Sun X. Synergistic effects of Cydia pomonella granulovirus GP37 on the infectivity of nucleopolyhedroviruses and the lethality of Bacillus thuringiensis. Arch Virol. 2011;156:1707–15. [PubMed: 21643992]217.Lemaitre B, Hoffmann J. The host defense of Drosophila melanogaster. Annu Rev Immunol. 2007;25:697–743. [PubMed: 17201680]218.Vialard JE, Yuen L, Richardson CD. Identification and characterization of a baculovirus occlusion body glycoprotein which resembles spheroidin, an entomopoxvirus protein. Journal of Virology. 1990;64:5804–5811. [PMC free article: PMC248735] [PubMed: 2243377]219.Gross CH, Wolgamot GM, Russell RLQ, Pearson MN, Rohrmann GF. A 37 kda glycoprotein from a baculovirus of Orgyia pseudotsugata is localized to cytoplasmic inclusion bodies. Journal of Virology. 1993;67:469–475. [PMC free article: PMC237384] [PubMed: 8380088]220.Mitsuhashi W, Miyamoto K. Disintegration of the peritrophic membrane of silkworm larvae due to spindles of an entomopoxvirus. J Invertebr Pathol. 2003;82:34–40. [PubMed: 12581717]221.Chiu E, Hijnen M, Bunker RD, Boudes M, Rajendran C, Aizel K, Olieric V, Schulze-Briese C, Mitsuhashi W, Young V, Ward VK, Bergoin M, Metcalf P, Coulibaly F. Structural basis for the enhancement of virulence by viral spindles and their in vivo crystallization. Proc Natl Acad Sci U S A. 2015;112:3973–8. [PMC free article: PMC4386404] [PubMed: 25787255]222.Liu X, Fang W, Fan R, Zhang L, Lei C, Zhang J, Nian W, Dou T, An S, Zhou L, Sun X. Granulovirus GP37 Facilitated ODVs Cross Insect Peritrophic Membranes and Fuse with Epithelia. Toxins. 2019:11. [PMC free article: PMC6468389] [PubMed: 30836616]223.Li Z, Gong Y, Yin C, Wang L, Li C, Pang Y. Characterization of a novel ubiquitin-fusion gene Uba256 from Spodoptera litura nucleopolyhedrovirus. Gene. 2003;303:111–9. [PubMed: 12559572]224.Müller R, Pearson MN, Russell RLQ, Rohrmann GF. A capsid-associated protein of the multicapsid nuclear polyhedrosis virus of Orgyia pseudotsugata: Genetic location, sequence, transcriptional mapping, and immunocytochemical characterization. Virology. 1990;176:133–144. [PubMed: 2184573]225.Marek M, van Oers MM, Devaraj FF, Vlak JM, Merten OW. Engineering of baculovirus vectors for the manufacture of virion-free biopharmaceuticals. Biotechnol Bioeng. 2011;108:1056–67. [PubMed: 21449023]226.Lu A, Carstens EB. Nucleotide sequence and transcriptional analysis of the p80 gene of Autographa californica nuclear polyhedrosis virus: a homologue of the Orgyia pseudotsugata nuclear polyhedrosis virus capsid-associated gene. Virology. 1992;190:201–209. [PubMed: 1529529]227.Marek M, Merten OW, Galibert L, Vlak JM, van Oers MM. Baculovirus VP80 protein and the F-actin cytoskeleton interact and connect the viral replication factory with the nuclear periphery. J Virol. 2011;85:5350–62. [PMC free article: PMC3094977] [PubMed: 21450830]228.Marek M, Merten OW, Francis-Devaraj F, Oers MM. Essential C-terminal region of the baculovirus minor capsid protein VP80 binds DNA. J Virol. 2012;86:1728–38. [PMC free article: PMC3264343] [PubMed: 22090126]229.de Jong J, Arif BM, Theilmann DA, Krell PJ. Autographa californica multiple nucleopolyhedrovirus me53 (ac140) is a nonessential gene required for efficient budded-virus production. J Virol. 2009;83:7440–8. [PMC free article: PMC2708630] [PubMed: 19457997]230.Xi Q, Wang J, Deng R, Wang X. Characterization of AcMNPV with a deletion of me53 gene. Virus Genes. 2007;34:223–32. [PubMed: 17096186]231.Knebel-Mörsdorf D, Kremer A, Jahnel F. Baculovirus gene ME53, which contains a putative zinc finger motif, is one of the major early-transcribed genes. J Virol. 1993;67:753–8. [PMC free article: PMC237427] [PubMed: 8093490]232.de Jong J, Theilmann DA, Arif BM, Krell PJ. Immediate-early protein ME53 forms foci and colocalizes with GP64 and the major capsid protein VP39 at the cell membranes of Autographa californica multiple nucleopolyhedrovirus-infected cells. J Virol. 2011;85:9696–707. [PMC free article: PMC3196415] [PubMed: 21775466]233.Wolgamot GM, Gross CH, Russell RLQ, Rohrmann GF. Immunocytochemical characterization of p24, a baculovirus capsid-associated protein. J Gen Virol. 1993;74:103–107. [PubMed: 8423444]234.Gombart AF, Blissard GW, Rohrmann GF. Characterization of the genetic organization of the HindIII-M region of the multicapsid nuclear polyhedrosis virus of Orgyia pseudotsugata reveals major differences among baculoviruses. J Gen Virol. 1989;70:1815–1828. [PubMed: 2661722]235.Schetter C, Oellig C, Doerfler W. An insertion of insect cell DNA in the 81-map-unit segment of Autographa californica nuclear polyhedrosis virus DNA. J Virol. 1990;64:1844–1850. [PMC free article: PMC249327] [PubMed: 2157067]236.Franke EK, Yuan HE, Luban J. Specific incorporation of cyclophilin A into HIV-1 virions. Nature. 1994;372:359–62. [PubMed: 7969494]237.Shobahah J, Xue S, Hu D, Zhao C, Wei M, Quan Y, Yu W. Quantitative phosphoproteome on the silkworm (Bombyx mori) cells infected with baculovirus. Virol J. 2017;14:117. [PMC free article: PMC5477107] [PubMed: 28629377]238.Hughes KM. The macromolecular lattices of polyhedra. J Invertebr Pathol. 1978;31:217–224.239.Rohrmann GF. Baculovirus structural proteins. J Gen Virol. 1992;73:749–761. [PubMed: 1634870]240.Westenberg M, Wang H, IJkel WF, Goldbach RW, Vlak JM, Zuidema D. Furin is involved in baculovirus envelope fusion protein activation. J Virol. 2002;76:178–84. [PMC free article: PMC135720] [PubMed: 11739683]241.Au S, Pante N. Nuclear transport of baculovirus: revealing the nuclear pore complex passage. J Struct Biol. 2012;177:90–8. [PubMed: 22100338]242.Nie Y, Fang M, Erlandson MA, Theilmann DA. Analysis of the autographa californica multiple nucleopolyhedrovirus overlapping gene pair lef3 and ac68 reveals that AC68 is a per os infectivity factor and that LEF3 is critical, but not essential, for virus replication. J Virol. 2012;86:3985–94. [PMC free article: PMC3302525] [PubMed: 22278232]243.Fang M, Nie Y, Harris S, Erlandson MA, Theilmann DA. Autographa californica multiple nucleopolyhedrovirus core gene ac96 encodes a per Os infectivity factor (PIF-4). J Virol. 2009;83:12569–78. [PMC free article: PMC2786767] [PubMed: 19759145]244.Jiantao L, Zhu L, Zhang S, Deng Z, Huang Z, Yuan M, Wu W, Yang K. The Autographa californica multiple nucleopolyhedrovirus ac110 gene encodes a new per os infectivity factor. Virus Res. 2016;221:30–7. [PubMed: 27212681]245.Kuzio J, Jaques R, Faulkner P. Identification of p74, a gene essential for virulence of baculovirus occlusion bodies. Virology. 1989;173:759–763. [PubMed: 2688302]246.Harrison RL, Sparks WO, Bonning BC. Autographa californica multiple nucleopolyhedrovirus ODV-E56 envelope protein is required for oral infectivity and can be substituted functionally by Rachiplusia ou multiple nucleopolyhedrovirus ODV-E56. J Gen Virol. 2010;91:1173–82. [PubMed: 20032203]247.Vanarsdall AL, Okano K, Rohrmann GF. Characterization of a baculovirus with a deletion of vlf-1. Virology. 2004;326:191–201. [PubMed: 15262507]


【本文地址】

公司简介

联系我们

今日新闻

    推荐新闻

    专题文章
      CopyRight 2018-2019 实验室设备网 版权所有