Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Aug 7.
Published in final edited form as: Curr Top Microbiol Immunol. 2010;342:1–14. doi: 10.1007/82_2010_10

The Varicella-Zoster Virus Genome

Jeffrey I Cohen 1
PMCID: PMC3413377  NIHMSID: NIHMS391459  PMID: 20225013

Abstract

The varicella-zoster virus (VZV) genome contains at least 70 genes, and all but 6 have homologs in herpes simplex virus. Cosmids and BACs corresponding to the VZV parental Oka and vaccine Oka viruses have been used to “knock-out” 34 VZV genes. Seven VZV genes (ORF4, 5, 9, 21, 29, 62, and 68) have been shown to be required for growth in vitro. Recombinant viruses expressing several markers (e.g. beta-galactosidase, green fluorescence protein, luciferase) and several foreign viral genes (from herpes simplex, Epstein-Barr virus, hepatitis B, mumps, HIV and simian immunodeficiency virus) have been constructed. Further studies of the VZV genome, using recombinant viruses, may facilitate the development of safer and more effective VZV vaccines. Furthermore, VZV might be useful as a vaccine vector to immunize against both VZV and other viruses.

1 Genome Structure and Organization

Varicella-zoster virus (VZV) is an alphaherpesvirus that is in the same subfamily as herpes simplex virus (HSV) 1 and 2. VZV is a member of varicellovirus genus, along with equine herpesvirus 1 and 4, pseudorabies virus, and bovine herpesvirus 1 and 5. Ceropithecine herpesvirus 9 (simian varicella virus) is virus most homologous to VZV.

1.1 VZV genome

The complete sequence of the VZV genome was determined by Davison and Scott (1986). The prototype strain, VZV Dumas is 124,884 base pairs in length. The genome consists of a unique long region (UL) bounded by terminal long (TRL) and internal long (IRL) repeats, and a unique short region (US) bounded by internal short (IRS), and terminal short (TRS) repeats (Figure 1). The US region can orientate either of two directions, while the UL region rarely changes its orientation; thus, there are usually two isomers of the genome in infected cells. The genome is linear in virions with an unpaired nucleotide at each end. In VZV-infected cells the ends pair and the genome circularizes.

Figure 1.

Figure 1

Comparison of the VZV and HSV genomes. The VZV genome (first rows) contains unique long (UL) and unique short (US) regions flanked by terminal long (TRL), terminal short (TRS), internal long (IRL), and internal short (IRS) repeats. VZV genes (second rows) and HSV genes (third rows) are shown by numbers. VZV or HSV genes that do not have homologs in the corresponding virus circled. The HSV genome contains UL and US regions, and TRL, TRS, IRL, and IRS repeats (fourth rows). Modified from Cohen 1999, with permission.

The genome has five repeat regions. Repeat region 1 (R1) is located in open reading frame (ORF) 11, R2 is located in ORF14 (glycoprotein C), R3 in ORF22, R4 between ORF62 and the origin of viral replication, and R5 between ORF 60 and 61. The length of the repeat regions varies among different VZV strains and has been used to distinguish the strains.

1.2 VZV genes

1.2.1 VZV immediate-early genes

VZV encodes at least 70 genes, three (ORF62, 63, 64) are which are present in both of the short repeat regions (Cohen et al. 2007b). VZV encodes at least 3 immediate-early (IE) proteins that are located in the tegument of virions and regulate virus transcription (Table 1). IE4 and IE62 transactivate IE, late, and early promoters. IE63 represses several VZV promoters, and inhibits the activity of interferon-alpha (Ambagala et al. 2007), and binds to anti-silencing protein 1 (Ambagala et al. 2009). ORF61 protein, which is not present in the tegument of virions and has not been shown to be an IE gene, activates IE, early, and late viral promoters.

Table 1.

VZV gene products

VZV
gene
HSV-1 homolog VZV protein function
1 None Membrane protein
2 None
3 UL55
4 UL54 (ICP 27) Transactivator, tegument protein, expressed in latency
5 UL53 (gK) gK
6 UL52 (HPC)
7 UL51
8 UL50 Deoxyuridine triphosphatase
9 UL49 (VP22) Tegument protein
9A UL49A Syncytia formation, putative gN
10 UL48 (VP16) Transactivator, tegument protein
11 UL47 (VP13/14)
12 UL46 (VP11/12)
13 None Thymidylate synthetase
14 UL44 (gC) gC
15 UL43
16 UL42 (PPF) Putative small subunit of viral DNA polymerase
17 UL41 (vhs) Induces RNA cleavage
18 UL40 Ribonucleotide reductase, small subunit
19 UL39 Ribonucleotide reductase, large subunit
20 UL38 (VP19C)
21 UL37 Nucleocapsid protein, expressed in latency
22 UL36
23 UL35 (VP26) Capsid assembly
24 UL34
25 UL33
26 UL32
27 UL31
28 UL30 DNA polymerase
29 UL29 (ICP8) ssDNA binding protein, expressed in latency
30 UL28
31 UL27 (gB) gB
32 None Probable substrate for ORF47 kinase
33 UL26 (VP24) Protease
33.5 UL26.5 (VP22) Assembly protein
34 UL25
35 UL24 Cell-to-cell fusion
36 UL23 Thymidine kinase
37 UL22 (gH) gH
38 UL21
39 UL20
40 UL19 (VP5) Major nucleocapsid protein
41 UL18 (VP23)
42/45 UL15
43 UL17
44 UL16
46 UL14
47 UL13 Protein kinase, tegument protein
48 UL12 Putative DNase
49 UL11 Virion protein
50 UL10 (gM) gM
51 UL9 Origin binding protein
52 UL8 (HPC)
53 UL7
54 UL6 Putative portal protein
55 UL5 (HPC)
56 UL4
57 None Cytoplasmic protein
58 UL3
59 UL2 Uracil-DNA glycosylase
60 UL1 (gL) gL, chaperone for gH
61 ICP0 Transactivator, transrepressor
62,71 ICP4 Transactivator, tegument protein, expressed in latency
63,70 US1 (ICP22) Tegument protein, transrepressor, inhibits interferon- alpha, expressed in latency
64,69 US10
65 US9 Virion protein
66 US3 Protein kinase, expressed in latency
67 US7 (gI) gI
68 US8 (gE) gE
S/L None Cytoplasmic protein (also referred to as ORF0)

HPC=helicase-primase complex, PPF=polymerase processivity factor

1.2.2 VZV genes encoding replication proteins

VZV encodes a viral DNA polymerase, likely composed of two subunits (ORF28 and ORF16) that is inhibited by acyclovir. The viral thymidine kinase (ORF36) phosphorylates deoxycytidine, thymidine, and acyclovir. VZV ORF18 and ORF19 encode the small and large subunits of ribonucleotide reductase which convert ribonucleotides to deoxyribonucleotides. VZV encodes at least two DNA binding proteins- ORF29 protein is a single-stranded DNA binding protein, and ORF 51 protein binds to the origin of DNA replication. VZV encodes two protein kinases. ORF47 protein phosphorylates VZV ORF32 protein, IE62, IE63, and glycoprotein I. ORF66 protein phosphorylates IE62 which results inclusion of IE62 into the virion tegument. VZV encodes other enzymes including a dUTPase (ORF8), thymidylate synthetase (ORF13), protease (ORF33), DNase (ORF48), and uracil DNA glycosylase (ORF59).

1.2.3 VZV genes encoding putative late proteins

VZV ORF10 encodes a tegument protein that forms a complex with transcription factors at the ORF62 promoter to activate transcription of ORF62. ORF17 protein induces cleavage of RNA. ORF33.5 encodes the assembly protein which forms a scaffold thought be involved in construction of nucleocapsids. ORF40 encodes the major nucleocapsid protein, while ORF21 also encodes a nucleocapsid protein. ORF54 encodes the putative portal protein which allows viral DNA to enter nucleocapsids.

1.2.4 VZV genes encoding glycoproteins

VZV encodes 7 viral glycoproteins- gB (ORF31), gC (ORF14), gE (OEF68), gH (ORF 37), gI (ORF67), gK (ORF 5), gL (ORF 60), gM (ORF50), and presumably gN (ORF9A). VZV gB, based on homology with HSV gB, is likely critical for entry of virus into cells. gE is binds to a cellular receptor (insulin degrading enzyme [Li et al. 2006]) and gH and gM are important for cell-to-cell spread of virus (Yamagishi et al. 2008). gI facilitates maturation of gE ,and gL is a chaperone for gH. gK may be important for syncytia formation.

2 Comparative genomics of VZV and HSV

VZV and HSV are largely collinear, although the UL region of HSV is orientated in the opposite direction to that of VZV using the standard nomenclature. The VZV US region is much shorter (5.2 kb) than HSV-1 US (13.0 kb) and the VZV TRL and IRL regions are also shorter (0.9 kb) than their HSV-1 counterparts (9.2 kb).

2.1 Core proteins conserved with herpesviruses in other subfamilies

The VZV genome contains about 41 “core genes” that are conserved with each of the three subfamilies of herpesviruses, alphaherpesvirus, betaherpesvirus, and gammaherpesvirus (Davison 1993). Core genes include IE4, the VZV DNA polymerase, helicase-primase components, single-stranded DNA-binding protein, ribonucleotide reductase, uracil-DNA glycosylase, dUTPase, DNase, ORF47 protein kinase, major capsid protein, protease, assembly protein, several tegument proteins, gB, gH, gL, gM, and gN.

2.2 VZV functional and nonfunctional homologs of HSV genes

Several VZV genes can complement their HSV homologs. VZV ORF61 can substitute for HSV ICP0 (Moriuchi et al. 1992) and VZV ORF62 can complement HSV ICP0 (Felser et al. 1988). Although VZV ORF10 can complement the transactivating function of HSV VP16, HSV-1 VP16 is essential for replication of HSV while VZV ORF10 is dispensable (Cohen and Seidel 1994b; Moriuchi et al. 1993). VZV ORF 51 can complement HSV UL9 (Chen et al 1995). In contrast, VZV ORF4 cannot complement HSV ICP27 (Moriuchi et al 1994a).

2.3 VZV genes not conserved with HSV

VZV encodes 6 genes (ORF1,2,13,32,57, and S/L) that are absent in HSV (Figure 1). ORF 13 encodes the viral thymidylate synthetase which has a homolog in herpesvirus saimiri and Kaposi’s sarcoma associated herpesvirus.

2.4 HSV genes not conserved with VZV

HSV encodes 9 genes (UL45, UL56, US2, US5, US6, US11, US12, and LAT) that are absent in VZV (Figure 1). HSV US 12 encodes ICP47, which blocks presentation of MHC class I. HSV US6, US 4, and US5 encode glycoproteins D, G, and J, respectively. VZV gE is the most abundant viral glycoprotein and shares many features of HSV gD, including binding to a cellular receptor which contributes to VZV entry (Li et al. 2007).

3 Mutagenesis with cosmids and BACs

3.1 Mutagenesis using marker rescue

The first genetically engineered mutant of VZV was constructed in 1987 in which the Epstein-Barr virus gp350 gene was inserted into the VZV genome (Lowe et al. 1987). Fibroblasts were cotransfected with VZV viron DNA and a plasmid with EBV gp350 flanked by VZV thymidine kinase sequences and plaques were purified by limiting dilution. While the procedure was successful, the process of plaque purification which requires sequential rounds of sonication is very labor intensive. The same strategy in which VZV virion DNA is contransfected with plasmids with flanking sequences that are homologous to VZV DNA was used to reinsert and thereby “rescue” essential genes into the genome of VZV lacking ORF4 (Cohen et al.. 2005), ORF21 (Xia et al. 2003), and ORF68 (Ali et al. 2009).

3.2 Mutagenesis using cosmids

A cosmid system was first used to generate mutations in the VZV genome in 1993 (Cohen and Seidel 1993). Virion DNA was isolated from the Oka vaccine strain of VZV, the linear DNA was blunted with T4 DNA polymerase, and oligonucleotides containing Not I or Mst II restriction sites were ligated to the DNA. The resulting DNA was cut with Mst II or Not I and four large DNA fragments, which overlap the entire VZV genome were ligated into a cosmid vector that was linearized with Mst II or Not I. Human melanoma cells were transfected with the four cosmids along with a plasmid encoding VZV IE62 which increases the infectivity of viral DNA (Moriuchi et al 1994b). While the cosmids are able to produce infectious virus in the absence of the plasmid, the latter increases the reliability and enhances the efficiency of virus production. Recombinant VZV derived from the cosmids grew to similar titers as the nonrecombinant virus used to generate the cosmids.

Other cosmid systems have also developed to perform VZV mutagenesis. Mallory et al. (1997) used 5 cosmids derived from the Oka vaccine strain to produce recombinant virus and Niizuma et al. (2003) described a cosmid library from the parental Oka vaccine. These cosmids have been used to “knock-out” a large number of VZV genes and to study the phenotype of the resulting mutants in cell culture and rodents.

3.3 Mutagenesis using BACs

Four separate BACs have been constructed for VZV mutagenesis. Three research groups have inserted the parental Oka virus into BACs (Nagaike et al. 2004; Zhang et al. 2007; Tischer et al. 2007) and one group inserted the vaccine Oka virus into a BAC (Yoshii et al. 2007). While each of the recombinant parental Oka viruses derived from BACs grew to titers similar to nonrecombinant parental Oka virus, recombinant virus derived from the vaccine Oka BAC grew to slightly lower titers than the nonrecombinant vaccine Oka virus.

3.4 Results of mutagenesis studies

VZV cosmids and BACs have been used to mutate multiple VZV genes (Table 2). Several VZV genes (ORF4, 5, 9, 21, 29, 62, and 68) have been shown to be essential for replication in cells in vitro. Other VZV genes that have been tested are not required for growth in cell culture, but some are required for growth in certain types of cells in vitro (Cohen and Nguygen 1997), in lymphocytes (Moffat et al. 1998; Song et al. 2000), or in human skin (Moffat et al. 1998, and Chapter 12). Each of the 6 VZV genes which do not have HSV homologs (ORF1, 2, 13, 32, 57, and S/L) are not required for growth in cell culture (Cohen and Seidel 1995; Cohen and Seidel 1993; Cox et al. 1998; Reddy et al. 1998a; Sato et al. 2002b; Zhang et al. 2007).

Table 2.

Deletion and stop codon mutants constructed using cosmids or BACs in VZV and their effect on growth in vitro.

VZV Gene Mutation Growth in Vitro Reference
ORFS/L (ORF0) del impaired Zhang et al. 2007
ORF1 stop no change Cohen and Seidel 1995
del slight reduced Zhang et al. 2007
ORF2 del no change Sato et al. 2002b
del no change Zhang et al. 2007
ORF3 del slight reduced Zhang et al. 2007
ORF4 del essential Cohen et al. 2005
del essential Sato et al. 2003b
del essential Zhang et al. 2007
ORF5 (gK) del essential Mo et al. 1999
ORF8 stop no change Ross et al. 2007
delete+ reduced Ross et al. 2007
ORF9 start essential Tischer et al. 2007
del essential Che et al. 2008
ORF9A stop no change Ross et al. 1997
ORF10 del no change Cohen and Seidel 1994b
del no change Che et al. 2008
ORF11 del no change Che et al. 2008
ORF12 del no change Che et al. 2008
ORF13 stop no change Cohen and Seidel 1993
ORF14 (gC) stop no change Cohen and Seidel 1994a
ORF17 del reduced Sato et al. 2002a
ORF19 del reduced Heineman and Cohen 1994
ORF21 del essential Xia et al. 2003
ORF23 del reduced Chaudhuri et al. 2008
ORF29 del essential Cohen et al. 2007
ORF32 del no change Reddy et al. 1998a
ORF35 del reduced Ito et al. 2005
ORF47 stop no change Heineman and Cohen 1995
ORF49 del reduced Sadaoka et al. 2007
ORF50 del reduced Yamagishi et al. 2008
ORF57 del no change Cox et al. 1998
ORF58 del no change Yoshii et al. 2008
ORF59 del no change Reddy et al. 1998b
ORF61 del reduced Cohen and Nguyen 1998
ORF62 del essential Sato et al. 2003a
ORF63 del essential Sommer et al. 2001
del reduced Cohen et al. 2004
ORF65 del no change Cohen et al. 2001
del no change Niizuma et al. 2003
ORF66 stop no change Heineman et al. 1996
ORF67 (gI) del reduced* or essential Cohen and Nguyen 1997
del reduced Mallory et al. 1997
ORF68 (gE) del essential Mallory et al. 1997
del essential Ali et al. 2009
+

interrupts expression of ORF8 and ORF9A

*

reduced in melanoma cells, essential for Vero cells

Stop=stop codons, del=deletion, start=mutations in start codon

Chimeras have been constructed containing various portions of the parental and vaccine Oka genomes (Zerboni et al. 2005). These chimeras have demonstrated that attenuation of VZV is a multigenic trait due to mutations throughout the genome.

VZV cosmids has been used to prove that mutations in individual genes correspond with resistance to antiviral compounds. Mutagenesis of VZV ORF54 (homologous to the portal protein of HSV) conferred resistance to a thioruea inhibitor compound (Visalli et al. 2003). VZV cosmids have been used to produce virus expressing beta-galactosidase (Cohen et al. 1998), green fluorescence protein (Zerboni et al. 2000, Li et al. 2006), or luciferase (Oliver et al. 2008, Zhang et al. 2007) which have been useful for studies in animals and virus entry in vitro.

Most recombinant viruses have stable genomes with a few exceptions. Attempts to delete or mutate one copy of a gene that is normally present in both of the short repeat regions of the genome often result in recombination with wild-type sequences in both short repeat regions after several rounds of replication (Sommer et al. 2001; Sato et al. 2003a; Oliver et al. 2008). In addition, point mutations that severely impair growth of the virus can sometimes undergo back mutation with reversion to wild-type virus over time (Cohen et al. 1998)

VZV genes thought to be required for cell growth have been proven to essential by growing a virus mutant unable to express the protein in a complementing cell line (Xia et al. 2003) or in cells infected with baculovirus expressing the VZV protein (Ali et al. 2009; Cohen et al. 2005; Cohen et al 2007) and then showing that the virus cannot be grown on non-complementing cells. An alternative approach has been to insert the essential gene elsewhere in the viral genome. This latter method is not ideal, since the gene will not be formally proven to be required for growth. Inefficiency in cosmid transfections or impaired virus growth may be misinterpreted as showing that a virus gene is essential. One VZV gene, ORF63, has been reported to be essential in the absence of a complementation system (Sommer et al 2001), but was actually shown to be non-essential for growth in cell culture (Cohen et al. 2004). The failure to obtain virus in the former study may have been due to the lack of a cotransfected plasmid expressing the VZV IE62 gene to transiently enhance virus growth after cosmid DNA transfection (Baiker et al. 2004).

3.5 VZV as an expression vector

VZV has been used to express a number of foreign viral proteins (Table 3). While these recombinant VZV mutants have been shown to induce immunity to the foreign virus and to protect animals from disease after challenging with the foreign virus in some cases (Heineman et al. 1995; Heineman et al. 2004), in one case the recombinant VZV actually enhanced replication of the foreign virus (Strapans et al. 2004). Thus, it is critical to test such recombinant viruses in animal models whenever possible. Since children are vaccinated with the varicella vaccine to prevent chickenpox, expression of additional viral proteins might allow immunization against other viruses and ultimately reduce the number of vaccines required during childhood.

Table 3.

Recombinant VZV expressing other viral proteins

Viral protein Immunogenicity Reference
EBV gp350 Not reported Lowe et al. l987
Hepatitis B SAg Induced antibody to Hepatitis B S Ag Shiraki et al. 1991
HIV env Induced humoral and cellular immunity to HIV Shiraki et al. 2001
SIV gp160 Induced neutralizing antibody
Enhanced SIV infection in monkeys
Strapans et al. 2004
HSV gD Induced neutralizing antibody to HSV-2
Reduced severity of HSV-2 in guinea pigs
Heineman et al. 1995
Mumps HA-N Induced neutralizing antibody to mumps Somboonthum et al. 2007
HSV gD and gB Induced neutralizing antibody to HSV-2
Reduced severity of HSV-2 in guinea pigs
Heineman et al. 2004

gp350=glycoprotein 350, SAg=surface antigen, gD=glycoprotein Dm HA-N=hemagglutinin-neuraminidase, gB=glycoprotein B

3.6 Use of genetics to develop safer VZV vaccines

The current vaccine is very safe in immunocompetant persons, but it has caused disease in severely immunocompromised persons. Replication-defective vaccines might be developed by growing viruses in complementing cells; however, attempts to produce high titers of cell-free virus using this approach has been unsuccessful (Cohen, unpublished data). An alternative approach is to express an essential gene, such as one expressed during latency, under a different promoter, which may allow high titers of virus during replication, but may impair latency (Cohen et al 2007).

Acknowledgements

I thank the intramural research program of the National Institutes of Allergy and Infectious Diseases for support.

Abbreviations

VZV

varicella-zoster virus

HSV

herpes simplex virus

UL

unique long

TRL

terminal repeat long

IRL

internal repeat long

US

unique short

IRS

internal repeat short

TRS

terminal repeat short

IE

immediate-early

References

  1. Ali MA, Li Q, Fischer ER, Cohen JI. The insulin degrading enzyme binding domain of varicella-zoster virus (VZV) glycoprotein E is important for cell-to-cell spread and VZV infectivity, while a glycoprotein I binding domain is essential for infection. Virology. 2009;386:270–279. doi: 10.1016/j.virol.2009.01.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Ambagala AP, Bosma T, Ali MA, Poustovoitov M, Chen JJ, Gershon MD, Adams PD, Cohen JI. Varicella-zoster virus immediate-early 63 protein interacts with human antisilencing function 1 protein and alters its ability to bind histones H3.1 and H3.3. J Virol. 2009;83:200–209. doi: 10.1128/JVI.00645-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Ambagala AP, Cohen JI. Varicella-Zoster virus IE63, a major viral latency protein, is required to inhibit the alpha interferon-induced antiviral response. J Virol. 2007;81:7844–7851. doi: 10.1128/JVI.00325-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Baiker A, Bagowski C, Ito H, Sommer M, Zerboni L, Fabel K, Hay J, Ruyechan W, Arvin AM. The immediate-early 63 protein of varicella-zoster virus: analysis of functional domains required for replication in vitro and for T-cell and skin tropism in the SCIDhu model in vivo. J Virol. 2004;78:1181–1194. doi: 10.1128/JVI.78.3.1181-1194.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Chaudhuri V, Sommer M, Rajamani J, Zerboni L, Arvin AM. Functions of Varicella-zoster virus ORF23 capsid protein in viral replication and the pathogenesis of skin infection. J Virol. 2008;82:10231–10246. doi: 10.1128/JVI.01890-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Che X, Reichelt M, Sommer MH, Rajamani J, Zerboni L, Arvin AM. Functions of the ORF9-to-ORF12 gene cluster in varicella-zoster virus replication and in the pathogenesis of skin infection. J Virol. 2008;82:5825–5834. doi: 10.1128/JVI.00303-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Chen D, Stabell EC, Olivio PD. Varicella-zoster virus gene 51 complements a herpes simplex virus type I UL9 mutant. J Virol. 1995;69:4515–4518. doi: 10.1128/jvi.69.7.4515-4518.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Cohen JI. Genomic structure and organization of varicella zoster virus. Contrib Microbiol. 1999;3:10–20. doi: 10.1159/000060313. [DOI] [PubMed] [Google Scholar]
  9. Cohen JI, Cox E, Pesnicak L, Srinivas S, Krogmann T. The varicella-zoster virus ORF63 latency-associated protein is critical for establishment of latency. J Virol. 2004;78:1833–11840. doi: 10.1128/JVI.78.21.11833-11840.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Cohen JI, Krogmann T, Pesnicak L, Ali MA. Absence or overexpression of the varicella-zoster virus (VZV) ORF29 latency-associated protein impairs late gene expression and reduces latency in a rodent model. J Virol. 2007a;81:1586–1591. doi: 10.1128/JVI.01220-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Cohen JI, Krogmann T, Ross JP, Pesnicak LP, Prikhod�ko EA. The varicella-zoster virus ORF4 latency associated protein is important for establishment of latency. J Virol. 2005;79:6969–6975. doi: 10.1128/JVI.79.11.6969-6975.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Cohen JI, Nguyen H. Varicella-zoster virus glycoprotein I (gI) is essential for growth of virus in Vero cells. J Virol. 1997;71:6913–6920. doi: 10.1128/jvi.71.9.6913-6920.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Cohen JI, Nguyen H. Varicella-zoster virus ORF61 deletion mutants replicate in cell culture, but a mutant with stop codons in ORF61 reverts to wild-type virus. Virology. 1998;246:306–316. doi: 10.1006/viro.1998.9198. [DOI] [PubMed] [Google Scholar]
  14. Cohen JI, Seidel KE. Generation of varicella-zoster virus (VZV) and viral mutants from cosmid DNAs: VZV thymidylate synthetase is not essential for replication in vitro. Proc Natl Acad Sci USA. 1993;90:7376–7380. doi: 10.1073/pnas.90.15.7376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Cohen JI, Seidel KE. Absence of varicella-zoster virus (VZV) glycoprotein V does not alter growth of VZV in vitro or sensitivity to heparin. J Gen Virol. 1994a;75:3087–3093. doi: 10.1099/0022-1317-75-11-3087. [DOI] [PubMed] [Google Scholar]
  16. Cohen JI, Seidel KE. Varicella-zoster virus (VZV) open reading frame 10 protein, the homolog of the essential herpes simplex virus protein VP16, is dispensable for VZV replication in vitro. J Virol. 1994b;68:7850–7858. doi: 10.1128/jvi.68.12.7850-7858.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Cohen JI, Seidel KE. Varicella-zoster virus open reading frame 1 encodes a membrane protein that is dispensable for growth of VZV in vitro. Virology. 1995;206:835–842. doi: 10.1006/viro.1995.1006. [DOI] [PubMed] [Google Scholar]
  18. Cohen JI, Sato H, Srinivas S, Lekstrom K. The varicella-zoster virus (VZV) ORF65 virion protein is dispensable for replication in cell culture and is phosphorylated by casein kinase II, but not by the VZV protein kinases. Virology. 2001;280:62–71. doi: 10.1006/viro.2000.0741. [DOI] [PubMed] [Google Scholar]
  19. Cohen JI, Straus SE, Arvin AM. Varicella-zoster virus: Replication, pathogenesis, and management. In: Knipe DM, Howley PM, editors. Fields Virology. 5th ed. Philadelphia: Lippincott-Williams & Wilkins; 2007b. [Google Scholar]
  20. Cohen JI, Wang Y, Nussenblatt R, Straus SE, Hooks JJ. Chronic uveitis in guinea pigs infected with varicella-zoster virus expressing Escherichia coli beta-galactosidase. J Infect Dis. 1998;177:293–300. doi: 10.1086/514210. [DOI] [PubMed] [Google Scholar]
  21. Cox E, Reddy S, Iofin I, Cohen J. Varicella-zoster virus ORF57, unlike its pseudorabies virus UL3.5 homolog, is dispensable for replication in cell culture. Virology. 1998;250:205–209. doi: 10.1006/viro.1998.9349. [DOI] [PubMed] [Google Scholar]
  22. Davison AJ. Herpesvirus genes. Rev Med Virol. 1993;3:237–244. [Google Scholar]
  23. Davison AJ, Scott J. The complete DNA sequence of varicella-zoster virus. J Gen Virol. 1986;67:1759–1816. doi: 10.1099/0022-1317-67-9-1759. [DOI] [PubMed] [Google Scholar]
  24. Felser JM, Kinchington PR, Inchauspe G, Straus SE, Ostrove JM. Cell lines containing varicella-zoster virus open reading frame 62 and expressing the "IE" 175 protein complement ICP4 mutants of herpes simplex virus type 1. J Virol. 1988;62:2076–2082. doi: 10.1128/jvi.62.6.2076-2082.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Heineman TC, Cohen JI. Deletion of the varicella-zoster virus large subunit of ribonucleotide reductase impairs the growth of virus in vitro. J. Virol. 1994;68:3317–3323. doi: 10.1128/jvi.68.5.3317-3323.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Heineman TC, Cohen JI. The varicella-zoster virus (VZV) open reading frame 47 (ORF47) protein kinase is dispensable for viral replication and is not required for phosphorylation of ORF63 protein, the VZV homolog of herpes simplex virus ICP22. J Virol. 1995;69:7367–7370. doi: 10.1128/jvi.69.11.7367-7370.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Heineman TC, Connelly BL, Bourne N, Stanberry LR, Cohen JI. Immunization with recombinant varicella-zoster virus expressing herpes simplex virus type 2 glycoprotein D reduces the severity of genital herpes in guinea pigs. J Virol. 1995;69:8109–8113. doi: 10.1128/jvi.69.12.8109-8113.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Heineman TC, Seidel K, Cohen JI. The varicella-zoster virus ORF66 protein induces kinase activity and is dispensable for viral replication. J Virol. 1996;70:7312–7317. doi: 10.1128/jvi.70.10.7312-7317.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Heineman T, Pesnicak L, Ali M, Krogmann T, Krudwig N, Cohen JI. Varicella-zoster virus expressing HSV-2 glycoproteins B and D induces protection against HSV-2 challenge. Vaccine. 2004;22:2558–2565. doi: 10.1016/j.vaccine.2003.12.010. [DOI] [PubMed] [Google Scholar]
  30. Ito H, Sommer MH, Zerboni L, Baiker A, Sato B, Liang R, Hay J, Ruyechan W, Arvin AM. Role of the varicella-zoster virus gene product encoded by open reading frame 35 in viral replication in vitro and in differentiated human skin and T cells in vivo. J Virol. 2005;79:4819–4827. doi: 10.1128/JVI.79.8.4819-4827.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Li Q, Ali MA, Cohen JI. Insulin degrading enzyme is a cellular receptor for varicella-zoster virus infection and for cell-to-cell spread of virus. Cell. 2006;127:305–316. doi: 10.1016/j.cell.2006.08.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Lowe RS, Keller PM, Keech BJ, Davison AJ, Whang Y, Morgan AJ, Kieff E, Ellis RW. Varicella-zoster virus as a live vector for the expression of foreign genes. Proc Natl Acad Sci U S A. 1987;84:3896–3900. doi: 10.1073/pnas.84.11.3896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Mallory S, Sommer M, Arvin AM. Mutational analysis of the role of glycoprotein I in varicella-zoster virus replication and its effects on glycoprotein E conformation and trafficking. J Virol. 1997;71:8279–8288. doi: 10.1128/jvi.71.11.8279-8288.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Mo C, Suen J, Sommer M, Arvin A. Characterization of Varicella-Zoster virus glycoprotein K (open reading frame 5) and its role in virus growth. J Virol. 1999;73:4197–4207. doi: 10.1128/jvi.73.5.4197-4207.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Moffat JF, Zerboni L, Sommer MH, Heineman TC, Cohen JI, Kaneshima H, Arvin AM. The ORF47 and ORF66 putative protein kinases of varicella-zoster virus determine tropism for human T cells and skin in the SCID-hu mouse. Proc Natl Acad Sci U S A. 1999;95:11969–11974. doi: 10.1073/pnas.95.20.11969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Moriuchi H, Moriuchi M, Smith HA, Cohen JI. Varicella-zoster virus open reading frame 4 protein is functionally distinct from and does not complement its herpes simplex virus type 1 homolog ICP27. J Virol. 1994a;68:1987–1992. doi: 10.1128/jvi.68.3.1987-1992.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Moriuchi H, Moriuchi M, Smith HA, Straus SE, Cohen JI. Varicella-zoster virus open reading frame 61 protein is functionally homologous to herpes simplex virus type 1 ICP0. J Virol. 1992;66:7303–7308. doi: 10.1128/jvi.66.12.7303-7308.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Moriuchi H, Moriuchi M, Straus SE, Cohen JI. Varicella-zoster virus open reading frame 10 protein, the herpes simplex virus VP16 homolog, transactivates herpesvirus immediate-early gene promoters. J Virol. 1993;67:2739–2746. doi: 10.1128/jvi.67.5.2739-2746.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Moriuchi M, Moriuchi H, Straus SE, Cohen JI. Varicella-zoster virus (VZV) virion-associated transactivator open reading frame 62 protein enhances the infectivity of VZV DNA. Virology. 1994b;200:297–300. doi: 10.1006/viro.1994.1190. [DOI] [PubMed] [Google Scholar]
  40. Nagaike K, Mori Y, Gomi Y, Yoshii H, Takahashi M, Wagner M, Koszinowski U, Yamanishi K. Cloning of the varicella-zoster virus genome as an infectious bacterial artificial chromosome in Escherichia coli Vaccine. 2004;22:4069–4074. doi: 10.1016/j.vaccine.2004.03.062. [DOI] [PubMed] [Google Scholar]
  41. Niizuma T, Zerboni L, Sommer MH, Ito H, Hinchliffe S, Arvin AM. Construction of varicella-zoster virus recombinants from parent Oka cosmids and demonstration that ORF65 protein is dispensable for infection of human skin and T cells in the SCID-hu mouse model. J Virol. 2003;77:6062–6065. doi: 10.1128/JVI.77.10.6062-6065.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Oliver SL, Zerboni L, Sommer M, Rajamani J, Arvin AM. Development of recombinant varicella-zoster viruses expressing luciferase fusion proteins for live in vivo imaging in human skin and dorsal root ganglia xenografts. J Virol Methods. 2008;154:182–193. doi: 10.1016/j.jviromet.2008.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Reddy SM, Cox E, Iofin I, Soong W, Cohen JI. Varicella-zoster virus (VZV) ORF32 encodes a phosphoprotein that is posttranscriptionally modified by the VZV ORF47 protein kinase. J Virol. 1998a;72:8083–8088. doi: 10.1128/jvi.72.10.8083-8088.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Reddy SM, Williams M, Cohen JI. Expression of a uracil DNA glycosylase (UNG) inhibitor in mammalian cells: varicella-zoster virus can replicate in vitro in the absence of detectable UNG activity. Virology. 1998b;251:393–401. doi: 10.1006/viro.1998.9428. [DOI] [PubMed] [Google Scholar]
  45. Ross J, Williams M, Cohen JI. Disruption of the varicella-zoster virus dUTPase and the adjacent ORF9A gene results in impaired growth and reduced syncytia formation in vitro. Virology. 1997;234:186–195. doi: 10.1006/viro.1997.8652. [DOI] [PubMed] [Google Scholar]
  46. Sadaoka T, Yoshii H, Imazawa T, Yamanishi K, Mori Y. Deletion in open reading frame 49 of varicella-zoster virus reduces virus growth in human malignant melanoma cells but not in human embryonic fibroblasts. J Virol. 2007;81:12654–12665. doi: 10.1128/JVI.01183-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Sato B, Ito H, Hinchliffe S, Sommer MH, Zerboni L, Arvin AM. Mutational analysis of open reading frames 62 and 71, encoding the varicella-zoster virus immediate-early transactivating protein, IE62, and effects on replication in vitro and in skin xenografts in the SCID-hu mouse in vivo. J Virol. 2003a;77:5607–5620. doi: 10.1128/JVI.77.10.5607-5620.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Sato B, Sommer M, Ito H, Arvin AM. Requirement of varicella-zoster virus immediate-early 4 protein for viral replication. J Virol. 2003b;7:12369–12372. doi: 10.1128/JVI.77.22.12369-12372.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Sato H, Callanan LD, Pesnicak L, Krogmann T, Cohen JI. Varicella-zoster virus (VZV) ORF17 protein induces RNA cleavage and is critical for replication of VZV at 37°C, but not 33°C. J Virol. 2002a;76:11012–11023. doi: 10.1128/JVI.76.21.11012-11023.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Sato H, Pesnicak L, Cohen JI. Varicella-zoster virus ORF2 encodes a membrane phosphoprotein that is dispensable for viral replication and for establishment of latency. J Virol. 2002b;76:3575–3578. doi: 10.1128/JVI.76.7.3575-3578.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Shiraki K, Hayakawa Y, Mori H, Namazue J, Takamizawa A, Yoshida I, Yamanishi K, Takahashi M. Development of immunogenic recombinant Oka varicella vaccine expressing hepatitis B virus surface antigen. J Gen Virol. 1991;72:1393–1399. doi: 10.1099/0022-1317-72-6-1393. [DOI] [PubMed] [Google Scholar]
  52. Shiraki K, Sato H, Yoshida Y, Yamamura JI, Tsurita M, Kurokawa M, Kageyama S. Construction of Oka varicella vaccine expressing human immunodeficiency virus env antigen. J Med Virol. 2001;64:89–95. doi: 10.1002/jmv.1022. [DOI] [PubMed] [Google Scholar]
  53. Somboonthum P, Yoshii H, Okamoto S, Koike M, Gomi Y, Uchiyama Y, Takahashi M, Yamanishi K, Mori Y. Generation of a recombinant Oka varicella vaccine expressing mumps virus hemagglutinin-neuraminidase protein as a polyvalent live vaccine. Vaccine. 2007;25:8741–8755. doi: 10.1016/j.vaccine.2007.10.039. [DOI] [PubMed] [Google Scholar]
  54. Sommer MH, Zagha E, Serrano OK, Ku CC, Zerboni L, Baiker A, Santos R, Spengler M, Lynch J, Grose C, Ruyechan W, Hay J, Arvin AM. Mutational analysis of the repeated open reading frames, ORFs 63 and 70 and ORFs 64 and 69, of varicella-zoster virus. J Virol. 2001;75:8224–8239. doi: 10.1128/JVI.75.17.8224-8239.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Soong W, Schultz JC, Patera AC, Sommer MH, Cohen JI. Infection of human T lymphocytes with varicella-zoster virus: an analysis with viral mutants and clinical isolates. J Virol. 2000;74:1864–1870. doi: 10.1128/jvi.74.4.1864-1870.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Strapans SI, Barry AP, Silvestri G, Safrit JT, Kozyr N, Sumpter B, Nguygen H, McClure H, Montefiori D, Cohen JI, Feinberg M. Enhance simian immunodeficiency virus replication and accelerated AIDS in macaques primed to mount a CD4 T cell response to SIV Env. Proc Natl Acad Sci. 2004;101:13026–13031. doi: 10.1073/pnas.0404739101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Tischer BK, Kaufer BB, Sommer M, Wussow F, Arvin AM, Osterrieder N. A self-excisable infectious bacterial artificial chromosome clone of varicella-zoster virus allows analysis of the essential tegument protein encoded by ORF9. J Virol. 2007;81:13200–13208. doi: 10.1128/JVI.01148-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Visalli RJ, Fairhurst J, Srinivas S, Hu W, Feld B, DiGrandi M, Curran K, Ross A, Bloom JD, van Zeijl M, Jones TR, O'Connell J, Cohen JI. Identification of small molecule compounds that selectively inhibit varicella-zoster virus replication. J Virol. 2003;77:2349–2358. doi: 10.1128/JVI.77.4.2349-2358.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Xia D, Srinivas S, Sato H, Pesnicak L, Straus SE, Cohen JI. Varicella-zoster virus ORF21, which is expressed during latency, is essential for virus replication but dispensable for establishment of latency. J Virol. 2003;77:1211–1218. doi: 10.1128/JVI.77.2.1211-1218.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Yamagishi Y, Sadaoka T, Yoshii H, Somboonthum P, Imazawa T, Nagaike K, Ozono K, Yamanishi K, Mori Y. Varicella-zoster virus glycoprotein M homolog is glycosylated, is expressed on the viral envelope, and functions in virus cell-to-cell spread. J Virol. 2008;82:795–804. doi: 10.1128/JVI.01722-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Yoshii H, Sadaoka K, Matsuura M, Nagaike K, Takahashi M, Yamanishi K, Mori Y. Varicella-zoster virus ORF 58 gene is dispensable for viral replication in cell culture. Virol J. 2008;30:5–54. doi: 10.1186/1743-422X-5-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Yoshii H, Somboonthum P, Takahashi M, Yamanishi K, Mori Y. Cloning of full length genome of varicella-zoster virus vaccine strain into a bacterial artificial chromosome and reconstitution of infectious virus. Vaccine. 2007;25:5006–5012. doi: 10.1016/j.vaccine.2007.04.064. [DOI] [PubMed] [Google Scholar]
  63. Zerboni L, Hinchliffe S, Sommer MH, Ito H, Besser J, Stamatis S, Cheng J, Distefano D, Kraiouchkine N, Shaw A, Arvin AM. Analysis of varicella zoster virus attenuation by evaluation of chimeric parent Oka/vaccine Oka recombinant viruses in skin xenografts in the SCIDhu mouse model. Virology. 2005;332:337–346. doi: 10.1016/j.virol.2004.10.047. [DOI] [PubMed] [Google Scholar]
  64. Zerboni L, Sommer M, Ware CF, Arvin AM. Varicella-zoster virus infection of a human CD4-positive T-cell line. Virology. 2000;270:278–285. doi: 10.1006/viro.2000.0304. [DOI] [PubMed] [Google Scholar]
  65. Zhang Z, Rowe J, Wang W, Sommer M, Arvin A, Moffat J, Zhu H. Genetic analysis of varicella-zoster virus ORF0 to ORF4 by use of a novel luciferase bacterial artificial chromosome system. J Virol. 2007;81:9024–9033. doi: 10.1128/JVI.02666-06. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES