Lot—”2:. :. E... 4.5.; I 4 ‘1... .31. . .5 .1 c 3.... 771. www‘a .. .w: .m‘ a. #1.: .— c .. . L432. t» » ‘5 1:. . 1!. . €be 1‘? ‘1 5.11.1. ‘ .3. 3:33..“ 2am“. .215 , . 4?? L .anu ML“. {.kmmn.‘ YE. I5... xx! All . .I: ‘ hvafifimmmv‘ v“— I. I...“ \..v.i‘.Q “I, 3.!aivku... ; x91ls§ .1 Q): r Ii 47‘; I a i: 3.1.1.? , . .3144. .IV‘GL...“ :‘lot. . . I. I:.J.\ 1...! u. 1.. .33. J t 1. s37! . 3... its. . a. Itx.ll.kiA! illicit. 5.4)}! fl}... .9 , t .i . .u.3.....«..«.2€a..vr.u. ... .51. .1... .. .. m i: .11... A 1.51. . 1 it w Ear...“ N 9 N o LIBRARY Michigan State University This is to certify that the dissertation entitled THE COMPLETE AND ANNOTATED GENOMIC SEQUENCE OF FELINE HERPESVIRUS 1 (FHV-1) AND AN INFECTIOUS BAC CLONE: A PLATFORM FOR STUDIES OF TARGETED MUTANTS BY RECOMBINEERING presented by Shih-Han Tai has been accepted towards fulfillment of the requirements for the Comparative Medicine and Ph. D. degree in Integrative Biology /‘/ /(_ (:3 OAQ‘WW" / Major Vrofessor’s Signature / /”r,r:)\Z f)2(W/Cs Date MSU is an Affinnative Action/Equal Opportunity Employer PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 5/08 K2IProj/AccsPresIClRCIDatoDueindd THE COMPLETE AND ANNOTATED GENOMIC SEQUENCE OF FELINE HERPESVIRUS l (FHV-l) AND AN INFECTIOUS BAC CLONE: A PLATFORM FOR STUDIES OF TARGETED MUTAN TS BY RECOMBINEERING By Shih-Han Tai A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILSOPHY Comparative Medicine and Integrative Biology 2010 ABSTRACT THE COMPLETE AND ANNOTATED GENOMIC SEQUENCE OF FELINE HERPESVIRUS 1 (FHV-l) AND AN INFECTIOUS BAC CLONE: A PLATFORM FOR STUDIES OF TARGETED MUTANTS BY RECOMBINEERING By Shih-Han Tai Infection with feline herpesvirus-l (F HV-l) is a major cause of upper respiratory and ocular disease in Felidae. For the first time, the complete genomic sequence of FHV-l was determined and annotated. Complete genomic sequences were derived fi'om both purified virion DNA and an infectious and virulent FHV-l BAC clone. The FHV -1 genome is 135,797 bp in size with an overall G+C content of 45%. A total of 78 open reading frames were predicted, encoding 74 distinct proteins. The gene arrangement is collinear with the majority of other sequenced varicelloviruses. A bacterial artificial chromosome (BAC) clone of FHV-l that contains the entire FHV-l genome and has the BAC vector inserted at the 5’-end of the UL was constructed and characterized in vitro and in vivo. The virus regenerated from the BAC was very similar to the parental C-27 strain in vitro in terms of plaque morphology and growth characteristics, and highly virulent in cats in a preliminary in vivo study. Using the latest recombination-mediated genetic engineering (recombineering) techniques, FHV-l mutants lacking the entire open reading frame encoding glycoprotein C (gC) or E (gE) were constructed based on the FHV-l BAC clone. The gC' FHV-l mutant virus produced primary plaques that enlarged slowly and had relatively few secondary plaques compared to the wild type virus. Analysis on grth kinetics showed that the gC' mutant had a growth titer reduced by approximately 6,000 — 9,000 folds compared to the wild-type virus titer, and the titer of intracellular virus was 3.5 — 10 folds higher than the extracellular virus titer. Taken together, the results show that the gC- FHV-l mutant is more cell-associated compared to the wild- type virus. Thus, FHV-l gC, like is the case in other varicelloviruses, plays a significant role in initial attachment/penetration, replication, and egress of FHV-l. In contrast, the gE' FHV—l mutant had single-step growth kinetics that were indistinguishable from those of the F HVlABAC, and grew to a titer that was approximately 10-fold lower than that of the FHVlABAC in the multi-step growth kinetics. These results suggest that virus egress, and most likely cell-to-cell spread as shown in other alphaherpesviruses, were affected by gE deletion, while virus entry and replication were not. A gC‘gE- and an U83 protein kinase (PK)' mutant of FHV-l were also generated, and generation of a gE-PK' mutant are currently in progress. Both the gC- gE_ and the PK- mutants demonstrated reduced growth rates in CRFK cells. The actual effect of these deletions on growth properties needs to be further characterized. The infectious FHV-l BAC clone and the complete and annotated FHV-l genomic sequence form a very suitable starting platform for mutagenesis aiming at functional studies of viral genes or vaccine development. To my family iv ACKNOWLEDGEMENTS I am extremely grateful to my advisor, Dr. Roger K. Maes, for his advice, help, and encouragement during my graduate study. I am very thankful to the members of my guidance committee, Drs. Hans Cheng, John Kruger, Annabel Wise, and Vilma Yuzbasiyan-Gurkan, for their guidance, encouragement, and support. I am especially thankful to Dr. Masahiro Niikura for guiding me through the process of BAC cloning, sequencing, and mutagenesis of the FHV-l genome, and sharing with me his research and life experience. I would like to thank Dr. Taejoong Kim for helpful advice on the recombineering process, Drs. Henry Hunt and Ian York for generously providing materials and advice, and Dr. Amalfitano for the use of ultracentn'fuge. I would like to thank Dr. Jeff Landgraf, Kevin Carr, Colleen Curry, and Shari Tjugum-Holland at the Research Technology Supporting Facility, for oligonucleotide synthesis, 454 sequencing and Newbler assembly. Special thanks to Tom Goodwill and Laurie Molitor at USDA-ARS Avian Disease and Oncology Laboratory for their help on Sanger sequencing. I am gratefiil to Michelle Brown, Janean DeVaul, Michael Engstrom, and Crystal Passmore for technical assistance, Janice F orcier for assistance in the in vivo experiment, and Denise Arnold and Victoria Hoelzer-Maddox for their help in administrative matters. It was a great pleasure to work among such excellent faculty members, researchers, and supporting staff while working in Dr. Cheng’s laboratory in the USDA- ARS Avian Disease and Oncology Laboratory and Dr. Maes’s laboratory in the Diagnostic Center for Population and Animal Health. Finally, I would like to thank my fellow graduate students in and out of the Comparative Medicine and Integrative Biology Graduate Program for their friendship and advice. I am very thankful to my parents for their support, and to Ai-Ting for her company and support over these years. vi TABLE OF CONTENTS LIST OF TABLES ........................................................................................................... ix LIST OF FIGURES ........................................................................................................... x LIST OF ABBREVIATIONS .......................................................................................... xi INTRODUCTION ............................................................................................................. 1 CHAPTER 1 A REVIEW OF FELINE HERPESVIRUS TYPE 1 (FHV-l) .................. 3 1.1 Classification ................................................................................................... 4 1.2 Virus Characteristics ....................................................................................... 4 1.2.1 Morphology .......................................................................................... 4 1.2.2 Host range and Cross-reactivity ........................................................... 5 1.2.3 Genome ................................................................................................. 5 1.2.4 Genes .................................................................................................... 6 1.3 Replication ..................................................................................................... 10 1.3.1 Attachment and Entry ......................................................................... 10 1.3.2 Gene Expression and DNA Replication ............................................. 11 1.3.3 Virion Assembly and Egress .............................................................. 12 1.4 Latency .......................................................................................................... 12 1.5 Clinical Manifestations and Pathogenesis ..................................................... 14 1.6 Diagnosis ....................................................................................................... 15 1.7 Treatment and Control ................................................................................... 16 1.7.1 Supportive Treatment ......................................................................... 16 1.7.2 Antiviral Therapy ............................................................................... 17 1.8 Immunity and Vaccination ............................................................................ 19 1.9 In Vitro Models of FHV-l Infection ............................................................. 22 1.10 Conclusions ................................................................................................... 24 1.11 References ..................................................................................................... 25 CHAPTER 2 BACTERIAL ARTIFICIAL CHROMOSOME (BAC) AND RECOMBINATION-MEDIATED GENETIC ENGINEERING (RECOMBINEERING) ........................................................................... 33 2.1 BAC Cloning ................................................................................................. 34 2.2 Recombineering ............................................................................................. 36 2.3 References ..................................................................................................... 39 CHAPTER 3 COMPLETE GENOMIC SEQUENCE AND AN INFECTIOUS BAC CLONE OF FELINE HERPESVIRUS 1 (FHV-l) ................................. 42 Abstract .................................................................................................................. 44 Introduction ............................................................................................................ 45 Results .................................................................................................................... 48 Discussion .............................................................................................................. 57 vii Materials and Methods ........................................................................................... 61 Acknowledgements ................................................................................................ 68 References .............................................................................................................. 69 Tables ..................................................................................................................... 74 Figures and Figure Legends ................................................................................... 83 CHAPTER 4 GENERATION AND IN VITRO CHARACTERIZATION OF F ELINE HERPESVIRUS 1 (FHV -1) MUTANT S LACKIN G GLYCOPROTEINS C (gC) AND E (gE) ............................................... 95 Abstract .................................................................................................................. 97 Introduction ............................................................................................................ 98 Materials and Methods ......................................................................................... 102 Results .................................................................................................................. 107 Discussion ............................................................................................................ 109 Acknowledgements .............................................................................................. 1 13 References ............................................................................................................ l 15 Tables ................................................................................................................... 120 Figures and Figure Legends ................................................................................. 121 CHAPTER 5 GENERATION OF gC’gE', US3 PROTEIN KINASE (PK): AND PK-gE- MUTANTS OF FELINE HERPESVIRUS l (FHV-l) .......... 130 5.1 Introduction ................................................................................................. 131 5.2 Materials and Methods ................................................................................ 132 5.3 Results and Discussions .............................................................................. 134 5.4 References ................................................................................................... 136 CHAPTER 6 CONCLUSIONS AND FUTURE DIRECTIONS ................................ 138 6.1 Conclusions ................................................................................................. 139 6.1.1 The FHV-l BAC Clone .................................................................... 139 6.1.2 The Complete Sequence of the FHV -1 Genome .............................. 139 6.1.3 FHV-l Mutants Lacking Glycoprotein C (gC) or E (gE) ................. 139 6.1.4 Other Deletion Mutants .................................................................... 140 6.2 Future Directions ......................................................................................... 140 6.2.1 Transcriptional & translational analyses .......................................... 140 6.2.2 Testing of the mutants in other feline cells ...................................... 141 6.2.3 In vivo testing of the F HV-l mutants ............................................... 141 6.2.4 A gC-complementing Cell Line ....................................................... 142 6.2.5 DIVA ELISA .................................................................................... 143 6.2.6 Further engineering of the gE- and gC- mutants ............................. 144 6.2.7 Molecular aspects of gC ......................................................................... APPENDICIES .............................................................................................................. 145 Appendix A ......................................................................................................... 145 Appendix B ..................................................................................................... 146 viii LIST OF TABLES Table 3.1 ......................................................................................................................... 74 Table 3.2 ......................................................................................................................... 76 Table 3.3 ......................................................................................................................... 77 Table 3.4 ......................................................................................................................... 80 Table 3.5 ......................................................................................................................... 81 Table 3.6 ......................................................................................................................... 82 Table 4.1 ....................................................................................................................... 120 Table 5.1 ....................................................................................................................... 133 Table 5.2 ....................................................................................................................... 135 Table 6.1 ....................................................................................................................... 141 ix LIST OF FIGURES Figure 3.1 ........................................................................................................................ 83 Figure 3.2 ........................................................................................................................ 85 Figure 3.3 ........................................................................................................................ 87 Figure 3.4 ........................................................................................................................ 89 Figure 3.5 ........................................................................................................................ 93 Figure 3.6 ........................................................................................................................ 94 Figure 4.1* .................................................................................................................... 121 Figure 4.2* .................................................................................................................... 123 Figure 4.3 ...................................................................................................................... 124 Figure 4.4 ...................................................................................................................... 125 Figure 4.5* .................................................................................................................... 126 Figure 4.6* .................................................................................................................... 127 Figure 4.7* .................................................................................................................... 128 Figure 4.8* .................................................................................................................... 129 *Images in the dissertation are presented in color. BAC BHV- l BHV-S CRFK DIVA EHV— l EHV-4 ELISA EMEM EGFP FBS FCE FCV F CWF-4 FHV-l LIST OF ABBREVIATIONS bacterial artificial chromosome bovine herpesvirus type 1 (bovine herpesvirus 1) bovine herpesvirus type 5 (bovine herpesvirus 5) Crandell-Reese feline kidney cells differentiation between infected and vaccinated animals equine herpesvirus type 1 (equid herpesvirus 1) equine herpesvirus type 4 (equid herpesvirus 4) enzyme-linked immunosorbent assay Eagle’s minimum essential medium enhanced green fluorescence protein fetal bovine serum feline corneal epithelial cells feline calicivirus felis catus whole fetus cells feline herpesvirus type 1 (felid herpesvirus 1) glycoprotein B glycoprotein C glycoprotein D glycoprotein E glycoprotein G glycoprotein H glycoprotein 1 xi gN HSV— 1 HSV—2 MDV ORF PCR PK PRV Recombineering RT-PCR TK VI VN VZV glycoprotein J glycoprotein K glycoprotein L glycoprotein M glycoprotein N herpes simplex virus type 1 (human herpesvirus 1) herpes simplex virus type 2 (human herpesvirus 2) Marek’s disease virus (gallid herpesvirus 2) open reading frame polymerase chain reaction protein kinase pseudorabies virus (suid herpesvirus 1) recombination-mediated genetic engineering reverse transcription-polymerase chain reaction thymidine kinase virus isolation virus neutralization varicella-zoster virus (human herpesvirus 3) xii INTRODUCTION Feline herpesvirus type 1 (FHV-l) is a major cause of upper respiratory infection in cats worldwide. In. recent years, its importance in ocular and chronic respiratory disease has also been increasingly recognized. According to the 2007 US. Pet Ownership & Demographics Sourcebook, there were 81,721,000 cats in the US. Various studies have shown that 50-90% of cats have serological evidence of exposure to the virus, more than 80% of infected cats remain latently infected for life, and ~45% of latently infected individuals shed virus throughout life. Consequently, the economic losses associated with F HV -1 infection are significant. Despite its high prevalence and worldwide distribution, our knowledge of F HV-l at the molecular level is limited. Further, vaccines currently available protect cats from disease, but not from infection and subsequent latency. There is a need for a safer and more effective vaccine. The goal of the research presented in this dissertation was to obtain an overview of the molecular composition of FHV—l, develop tools for fimctional studies on viral genes and virulence factors, and construct potential vaccine candidates. Chapter 1 provides a literature review of F HV-l. Chapter 2 introduces the state-of-the-art techniques that were essential for this research, including bacterial artificial chromosome (BAC) cloning and recombination-mediated genetic engineering (recombineering). Chapter 3 is a manuscript submitted to Virology, describing cloning of the entire FHV- l genome as a BAC, in vitro and in vivo characterization of this BAC clone, and sequencing and annotation of the entire FHV-l genome. Chapter 4 describes the construction and in vitro characterization of F HV—l mutants lacking glycoprotein C or E based on this FHV-l BAC clone, also in manuscript format. Chapter 5 summarizes the generation and preliminary characterization of other deletion mutants of FHV-l, including a gC-gE- mutant, a US3- mutant, and a US3-gE- mutant. Chapter 6 concludes the dissertation and points out potential directions for future research. CHAPTER 1 A REVIEW OF F ELINE HERPESVIRUS TYPE 1 (F HV-l) l . 1 Classification Felid herpesvirus 1 (FeHV-l, commonly known as feline rhinotracheitis virus, feline herpesvirus type 1, or FHV-l) is currently classified under the Order Herpesvirales, Family Herpesviridae, Subfamily Alphaherpesvirinae, genus Varicelloviruses (Davison et al., 2009). Prototype viruses for the Alphaherpesvirinae and Varicelloviruses are human herpesvirus 1 (HHV-l, commonly known as herpes simplex virus type 1, or HSV-l) and 3 (HHV-3, commonly known as varicella zoster virus, or VZV), respectively. Other varicelloviruses of veterinary importance that will be frequently mentioned in this and the following chapters include bovine herpesvirus 1 (BoHV-l or BHV-l) and 5 (BoHV-5 or BHV-S), equid herpesvirus 1 (commonly known as equine herpesvirus type 1, or EHV-l) and 4 (commonly known as equine herpesvirus type 4, or EHV-4), suid herpesvirus 1 (SuHV -1, commonly known as pseudorabies virus, or PRV). Other alphaherpesviruses of human or veterinary medical importance include human herpesvirus 2 (HHV-2, commonly known as herpes simplex virus type 2, or HSV-2) and gallid herpesvirus 2 (GaHV-2, commonly known as Marek’s disease virus, or MDV). 1.2 Virus Characteristics 1.2.1 Morphology FHV -1 particles are 120-180 nm in diameter. Herpesvirus particles consist of four elements: (a) an electron-opaque core containing the double-stranded viral DNA genome, (b) an icosahedral capsid surrounding the core, (c) a largely unstructured proteinacous layer called the tegument that surrounds the capsid, and (d) an outer lipid bilayer envelope surrounding the tegument layer and exhibiting glycoprotein spikes on its surface (Flint et al., 2004; Murphy et al., 1999; Pellett and Roizman, 2007; Roizman et al., 2007). 1.2.2 Host range and cross-reactivity Compared to some alphaherpesviruses, e. g., HSV—l and -2, FHV-l is considered to have a narrow host range. FHV-l infects only members of the Family F elidae (Povey, 1979). In vitro, FHV-l also replicates only in cells of feline origin. Crandell-Reese feline kidney (CRFK) cells are the cell line routinely used for propagation of FHV-l. F elis catus whole fetus (FCWF-4) cells is another cell line that can be used. In addition, feline alveolar macrophages, alveolar pneumocytes, CD4+ T-lymphoblastoid cells (MYA-l and FL74 cells), and feline corneal epithelial (FCE) cells, have been shown to be suitable for FHV-l propagation (Sandmeyer eta1., 2005a; Spatz, 1993). FHV-l, canid herpesvirus 1 (CaHV -1, commonly known as canine herpesvirus type 1, or CHV-l), and phocid herpesvirus 1 (PhHV-l, seal herpesvirus) are closely related genetically and antigenetically. Cross-protection between these viruses has been reported (Xuan et al., 1992). 1.2.3 Genome As a member of the Herpesviridae, the FHV—l genome consists of a single linear molecule of double-stranded DNA. FHV-l genomes of two different strains have been mapped using restriction enzymes. Rota et a1. (1986) first constructed a SalI map of the genome of C—27 strain and determined that it was ~l34 kb in size. Grail et al. (1991) mapped the B927 strain and determined that the genome was ~126 kb in size. Both studies found that the genome organization of FHV-l is similar to that of other varicelloviruses. The genome consists of two segments of unique sequences called Unique Long (UL) and Unique Short (Us). The Us region is flanked by a pair of identical but inverted sequences, known as Internal Repeat Short (IRS) and Terminal Repeat Short (TRS) (Figure 3.1). 1.2.4 Genes One characteristics of herpesviruses is that they carry a large array of enzymes involved in nucleic acid metabolism (e.g., thymidine kinase, thymidylate synthetase, dUTPase, ribonucleotide reductase), DNA synthesis (e.g., DNA polymerase, helicase, primase), and processing of proteins (e.g., protein kinases), although the exact array of enzymes may vary from one herpesvirus to another (Pellett and Roizman, 2007). Alphaherpesvirus genomes typically posess 65-80 open reading frames (Alba et al., 2001). FHV-l has been shown to contain 23 virion-associated proteins (Fargeaud et al., 1984). Eight glycoproteins have been identified, designated as gB, gC, gD, gE, gG, gH, g1, and gL. There are only limited studies on the genes encoded by FHV-l. Most of them are focused on envelope glycoproteins, most likely because they are predicted to play a role in inducing host immunity, thus, have potential for vaccine development. Studies on the functions of the first seven glycoproteins were reviewed by Maeda et a1. (1998). It is clear from this review that much more work is needed to define their actual fimctions and roles in viral pathogenesis and immunity. HSV gB contains two or three transmembrane segments and is essential for entry by fission of the envelope with the plasma membrane. Virions lacking gB egress the cell but are not infectious. The gB protein binds heparan sulfate (Roizman et al., 2007). Using heparin-affinity column chromatography, Maeda et al. (1997a) showed that FHV- 1 gB weakly binds to heparin. gC There have been very limited studies on gC of FHV-l. Willemse et al. (1994) first determined a partial sequence of gC in 1994. They also found that the adjacent UL45 gene can be co-transcribed with gC. The complete sequence of FHV-l gC was later determined by Maeda et al. (1997b). Based on the amino acid sequence deduced from the nucleotide sequence, they predicted that gC is a membrane glycoprotein containing a characteristic N-terminal hydrophobic signal sequence, nine potential N-linked glycosylation sites, and C-terminal transmembrane and cytoplasmic domains. Maeda et al. (1997a) further demonstrated that gC is the major heparin-binding glycoprotein involved in the initial step in virus adsorption to cells as observed in ng of other herpesviruses. In addition, they found that gC can agglutinate murine red blood cells, and that infection of FHV-l is inhibited by addition of soluble heparin in cells cultures. Some field isolates of F HV-l have been found to have genetic rearrangements in the N- terminal region of gC, however, the function of gC did not seem to be affected (Hamano et al., 2004). gC homologues have been extensively studied in several alphaherpesviruses. gC homologues are non-essential for herpesvirus replication in vitro, but they mediate several important biological functions. First of all, gC is involved in the initial step of viral attachment by interacting with heparan sulfate on the cell surface, as demonstrated in HSV-l, PRV, BHV-l, and EHV-l (Herold et al., 1991; Mettenleiter et al., 1990; Okazaki et al., 1991; Osterrieder, 1999). gC deficient mutants attach to cells with reduced efficiency (Osterrieder, 1999). Secondly, ng of HSV-1 and -2 can bind the complement component C3b (Frink et al., 1983; Lubinski et al., 1999). Binding of this complement factor may protect herpesvirus-infected cells fiom complement-mediated lysis (Fries et al., 1986). Viruses lacking complement-binding domains are less virulent than wild-type virus (Frink et al., 1983; Herold et al., 1991; Lubinski et al., 1999). The gC of FHV-l has been shown to be the dominant heparin-binding glycoprotein that mediates the initial stage of viral adsorption, as observed in other herpesviruses (Maeda et al., 1998). However, it remains to be determined whether FHV-l gC protects virus- infected cells from complement-mediated lysis. gD HSV gD is a multifunctional protein with the following properties: it interacts with three cellular receptors for entry — HveA, nectinl, and a modified heparin sulfate and, hence, defines viral tropism. 0n receptor binding, an ensuing change in conformation exposes profusion domains that enable firsogenic glycoprotein gB, gH, and gL to complete fusion of the envelope with the plasma membrane. The N-terminal domain may be replaced with ligands for entry via other receptors. gD protects the cell from apoptosis induced by AgD mutants. Antiapoptotic activity is mediated by the mannose-phosphate receptor. AgD mutants grown in cells ectopically expressing gD produce Virions that exit the cell, but are not infectious (Brunetti et al., 1998; Cocchi et al., 1998; Geraghty et al., 1998; Reynolds et al., 2001; Roizman et al., 2007). gE and g1 In our laboratory, it has previously been shown that gE and g1 are virulence factors (Kruger et al., 1996; Sussman et al., 1995). Unlike most commercial vaccines, an experimental F HV-l vaccine with deletions in gE and g1 was safe and efficacious via the oronasal route, and was able to highly reduce field virus latency loads in cats that were first vaccinated then exposed one month later to a high dose of field virus. Both glycoproteins have been shown to be nonessential for viral replication in vitro (Jacobs, 1994; Sussman et al., 1995; Willemse et al., 1996). gG Glycoprotein G (gG) homologues have been described in several alphaherpesviruses as a minor non-essential glycoprotein (Baranowski et al., 1996). Based on the viral species, gG has been reported either as a structural or a non-structural protein. The protein encoded by FHV-l gG gene exists as two different forms, a membrane-anchored form and a secreted form. The latter is generated by proteolytic cleavage of the former (Drummer et al., 1998). A recent study by Costes et al. demonstrated that FHV-l gG belongs to a newly discovered viral chemokine-binding protein (vCKBP) family, binding with high afiinity to a broad spectrum of chemokines. Both the secreted form and the membrane-anchored form of gG expressed at the surface of virus-infected cells can bind chemokines. In addition, this study also showed that the expression of a secreted vCKBP activity is a general property of field strains (Costes et al., 2006). Although gG is not essential for virus growth, gG mutants of several alphaherpesviruses have attenuated virulence and, thus, suggested as vaccine candidates. These include PRV (Demmin et al., 2001), avian infectious laryngotracheitis virus (Devlin et al., 2007), EHV-l and EHV-4 (Huang et al., 2005). l .3 Replication All alphaherpesviruses are believed to follow a similar replication pattern as HSV-1, which has been extensively studied. The replication cycle is summarized below (Flint et al., 2004; Pellett and Roizman, 2007; Roizman et al., 2007). 1.3.1 Attachement and Entry The replication cycle begins with virion attachment to the cell surface via binding of gC and gB to the heparin sulfate or chondroitin sulfate proteoglycans on the cell surface. Following the initial attachment, gD initiates fusion of viral envelope and cell membrane, mediated by gB, gD, gH, and gL. As a result of membrane firsion, viral nucleocapsid is released into the cytoplasm. It attaches to microtubules and is transported to the nucleus. Also released into the cytoplasm are the tegument proteins. 10 1.3.2 Gene Expression and DNA Replication The expression of three classes of genes (immediate-early, early, and late) in a temporal order is a characteristic of alphaherpesvirus replication. After viral DNA is released into the nucleus via the nuclear pore, VP16 interacts with host transcription proteins to initiate the transcription of immediate-early genes by host RNA polymerase II. Immediate-early gene mRNAs are transported to the cytoplasm and translated. The immediate-early proteins are transported back to the nucleus, activate transcription of early genes, and down-regulate transcription of immediate-early genes. Early gene mRNAs are also transported to the cytoplasm and translated. Early proteins are primarily involved in DNA replication and nucleotide metabolism. Subsequently, viral DNA synthesis is initiated from viral origins of replication. The replication of genomic DNA is often referred to as a rolling circle model, in which DNA replication and recombination produce long, concatemeric DNA, from a circularized template. The concatemeric DNA serves as a template for late gene expression. Late mRN As are transported to the cytoplasm and translated. Late proteins are primarily virion structural proteins and additional proteins needed for virus assembly and particle egress. Envelope glycoproteins are also late proteins that are made on and inserted into membranes of the rough endoplasmic reticulum. The precursor glycoproteins are transported to the Golgi apparatus for glycosylation, further modification and processing. Mature glycoproteins are transported to the plasma membrane of the infected cell. 11 1.3.3 Virion Assembly and Egress Newly replicated viral DNA is packaged into nucleocapsids in the nucleus. DNA- containing nucleocapsids, surrounded by some tegument proteins, bud from the inner nuclear membrane into the perinuclear lumen, acquiring a preliminary envelope that contains precursor viral membrane proteins. This process is known as primary envelopment. Immature enveloped Virions then fuse with the outer nuclear membrane from within, and release the nucleocapsid into the cytoplasm. The nucleocapsids are transported to and bud into the late Golgi-endosome compartment, acquiring an envelope containing mature viral envelope proteins and the complete tegument layer. The process is known as secondary envelopment. The enveloped virus particle then buds into a vesicle that is transported to the plasma membrane for release by exocytosis. 1.4 Latency Lifelong latency following the acute phase of the disease is a hallmark of herpesvirus infections. During the latent stage, the viral genome persists in neural tissues but infectious virus is not produced. The trigeminal ganglion is considered a primary site of latency for F HV —1, although recent studies implied other tissues as potential sites (Jacobi, 2008). When the latent state is established, viral DNA can circularize and persist in the nucleus as an episome. Lytic gene expression is repressed, while the latency-associated transcript (LAT) is expressed, which yields several RNA species by splicing. These multiple species are collectively referred to as LATs. Low- level or sporadic transcription of immediate-early and early genes can occur, but is not 12 sufficient to initiate a productive infection. No infectious Virions can be detected in the ganglia during latent infection. The LAT RNA is spliced, and a stable intron in the form of a lan'at, called the 2-kb LAT, is produced in the nucleus. The spliced LAT mRNA is transported to the cytoplasm where several small ORFs may be translated into proteins. The function of LAT RN As and the production of LAT proteins are still controversial. Different biological stresses, or the administration of corticosteroids, can induce the necessary biochemical stimuli in latently infected cells that lead to renewed production of progeny Virions. Infectious virus is carried by anterograde axonal transport to peripheral tissues, usually to cells at or near the site of initial infection, and is a potential source of viral transmission (Pellett and Roizman, 2007; Roizman et al., 2007). Depending on several factors, including the status of host immune system, the reactivation may be asymptomatic or lead to a recurrent disease, which can vary considerably in severity. Severe keratitis can also result from reactivation. The role of reactivation in the epidemiology is directly related to the frequency by which it takes place. Some herpesviruses, including F HV-l, reactivate much more easily than others from the latent state, both under natural and experimental conditions. The ease by which latent FHV-l is reactivated is an important element in the justification of FHV-l infection of cats as a natural host model to study the molecular pathogenesis of herpesvirus latency and approaches to prevent it. 13 1.5 Clinical Manifestations and Pathogenesis Following entry via the oronasal route, F HV-l replicates extensively in the mucosae of the upper respiratory tract. Pathologic examination reveals necrosis of epithelia of the nasal cavity, pharynx, epiglottis, tonsils, larynx, and trachea. In extreme cases and in young kittens, there can be an extensive rhinotracheitis and an associated bronchopneumonia. Clinically, F HV-l infection manifests itself as a sudden onset of sneezing, coughing, serous nasal and ocular discharge which can progress to mucopurulent discharges, frothy salivation, dyspnea, anorexia, weight loss, and fever after an incubation period of 24-48 hours. Conjunctivitis and keratitis are also common. Occasionally there may be ulcers on the tongue. Infection in cats over six months of age is likely to result in mild or subclinical infection. The mortality can reach 50% in kittens, since the virus tends to generalize in this age group. Exposure of pregnant queens can lead to abortion, but infection with FHV-l is not a common cause of abortion in cats. The main reason is that viremia is low, because the natural temperature sensitivity of FHV-l favors replication in the upper respiratory tract, which is below body temperature. In addition, there is no evidence that the virus crosses the placenta and fatally infects fetuses, and virus has not been isolated from placentas or aborted fetuses; abortion, when it occurs, is thought to be secondary to fever and toxemia. Neurological disorders are rare but have been observed clinically. In recent years, the importance of FHV-l in ocular disease has been increasingly recognized. FHV-l is the most common cause of conjunctivitis in cats (Nasisse, 1990). Epithelial keratitis commonly occurs during acute infection in young cats and resolves spontaneously in most cases (N asisse, 1995). In adult cats, reactivation of latent virus 14 can result in corneal ulceration, accompanied by a varying degree of conjunctivitis (Stiles, 2000). Since herpetic stromal keratitis caused by HSV-1 is the leading cause of infectious blindness in the industrialized countries, ocular infection of FHV-l in cats is considered a very good natural host model. 1.6 Diagnosis Clinically, the acute disease of FHV-l infection is very similar to that caused by feline calicivirus (FCV). Profuse frothy salivation and corneal ulcers suggest feline herpesvirus infection, whereas ulcers of the tongue, palate, and pharynx are encountered more fi'equently in calicivirus infections. In our laboratory, commonly performed diagnostics for FHV-l infection includes virus isolation (V I), polymerase chain reaction (PCR) assays, virus neutralization (VN) tests, and direct fluorescent antibody (FA) staining. VI detect infectious virus and has been the gold standard for the diagnosis of alphaherpesvirus infections (Maggs, 2005). It is performed by grinding tissue samples or elute the virus from swabs in Borvarnick’s buffer, passing through 0.45 pm filters, and incubate on CRFK monolayers in a 24-well plate or 25-cm2 flask. The cell culture is observed for the development of characteristic cytopathic effect caused by F HV-l. Compared to VI, PCR is more sensitive and efficient. Multiple PCR assays have been described for use in the detection of FHV-l DNA (Jacobi, 2008). The TaqMan based real-time PCR assay described by Vogtlin et al. (2002) targeting the gB gene is routinely used in our laboratory for diagnosis of F HV-l infection. It can be converted 15 into a quantitative assay by incorporating a standard curve, consisting of a serial dilution of FHV-l DNA whose genomic copy number had been determined. Our result showed that the detection limit of this assay is approximately 10 copies of FHV-l genome (unpublished data). A quantitative SYBR-Green based real-time PCR assay targeting gE and g1 genes was also developed in our laboratory. The detection limit for this assay is also approximately 10 copies (unpublished data). An internal control can be spiked in the reaction to determine whether a negative reaction was caused by inhibitory factors in the sample (unpublished data). Virus neutralizing antibody titers are determined by VN tests, which are commonly used to detect prior infection of FHV-l in cats. However, the presence of neutralizing antibody in serum does not necessarily correlate with clinical disease (Dawson et al., 1998). Feline herpesvirus-specific proteins on conjunctival or corneal smears or biopsies can be detected by direct FA staining. Polyclonal fluorescein conjugated antibody binds to FHV-l epitopes on the cell surface and can be visualized by fluorescent microscopy. 1.7 Treatment and Control 1.7.1 Supportive Treatment The European Advisory Board on Cat Diseases (ABCD) recently published a guideline for management of this disease (Thiry et al., 2009). Like many viral infections, supportive treatment is advised. To prevent secondary bacterial infections, broad- 16 spectrum antibiotics that achieve good penetration into the respiratory tract should be given in all acute cases. Food intake is extremely important. Food should be highly palatable and flavorful, since many cats will not eat because of loss of their sense of smell or the presence of ulcers in the oral cavity. Appetite stimulants (eg, cyproheptadine) may be used. If the cat does not eat for more than 3 days, a feeding tube should be placed. In cats with severe clinical signs, restoration of fluids, electrolytes and acid-base balance is required, preferably by intravenous administration. Nasal decongestants and mucolytic drugs have been suggested to help clear airways. Nebulization with saline can be used to ease dehydration of the airways. Eye drops or ointrnents can be administered several times a day. 1.7.2 Antiviral Therapy Nucleoside analogue antivirals have been widely used to treat HSV and VZV infections. Generally speaking, in infected cells, these nucleoside analogues are converted into triphosphates by viral thymidine kinase and other host enzymes, and competitively inhibit viral DNA polymerase, preventing DNA chain elongation (Snoeck, 2000). As a result, viral DNA synthesis, and hence viral replication, are disrupted. However, use of these agents against F HV -1 infection has been largely limited to topical administration. First generation nucleoside analogues, including acyclovir and its pro-drug valacyclovir, have little efficacy against F HV-l in vitro and moderate effect in vivo. More importantly, they produce serious side effects in cats, including myelosuppression, hepatotoxicity, and nephrotoxicity, when administered systemically at therapeutic levels (Nasisse et al., 1997; Owens et al., 1996). According to the 17 European ABCD’s guideline (Thiry et al., 2009), trifluridine is the topical treatment of choice in cats with ocular F HV-l manifestations. Acyclovir, ganciclovir, and idoxuridine are also suggested for topical use in the guideline. Other drugs that have been proposed for the treatment of FHV—l ocular infections include bromovinyldeoxyuridine, cidofovir, fameiclovir, HPMA (N-[2-hydroxypropyl] methacrylamide), penciclovir, ribavirin, valaciclovir, vidarabine, foscamet and lactoferrin. It was noted that, except for acyclovir, there is a lack of controlled in vivo efficacy study for these agents in the literature (Thiry et al., 2009). The efficacy of topical application of cidofovir on primary ocular FHV -1 infection was recently demonstrated (F ontenelle et al., 2008). F amciclovir, which is converted into penciclovir triphosphate by viral thymidine kinase in alphaherpesvirus-infected cells, was recently shown to be safe and efficacious when administered orally in treating ocular signs, cutaneous disease, and rhinosinusitis induced by FHV-l infection in 10 cats (Malik et al., 2009). Although it was not a controlled study, this is probably the first antiviral shown to be safe for systemic administration in cats. L-lysine is an antagonist of arginine, the latter has been shown to be essential for human herpes simplex virus and FHV-l proteosynthesis (Maggs et al., 2003). Treatment with L-lysine therefore decreases viral replication and has been shown to have some inhibitory effect against both human herpesvirus and FHV-l infection (Maggs et al., 2000; Maggs et al., 2003; Stiles et al., 2002). Oral supplementation with L-lysine reduces the severity of experimentally induced F HV-l conjunctivitis (Stiles et al., 2002) and ocular virus shedding associated with reactivation of latent infection (Maggs et al., 2003). It was suggested for use early in acute disease or as a means of 18 reducing the severity of disease and virus shedding at times of stress (Gaskell et al., 2007). Feline interferon (IFN) 0) and human IFN-a can be given systemically along with L-lysine to reduce clinical disease (Thiry et al., 2009). 1.8 Immunity and Vaccination Primary FHV-l infection induces both humoral and cellular immune responses. It is likely that virus neutralizing antibodies recognize incoming virus during the acute phase of infection, and contributes to antibody-dependent cellular cytotoxicity and antibody-induced complement lysis (Wardley et al., 1976). The immunity induced by natural F HV-l infection protects cats from the disease, but not from infection. In fact, the protection becomes incomplete six months after the primary infection (Gaskell and Povey, 1977; Walton and Gillespie, 1970), and mild clinical signs have been observed following re-infection as soon as 150 days after the primary infection (Thiry et al., 2009). Virus neutralizing antibody titers are generally low and in some cases undetectable after primary infection, although after further exposure to virus, they tend to rise to more moderate levels and thereafter remain reasonably stable (Gaskell et al., 2007; Gaskell and Povey, 1979). Since FHV-l is a pathogen of the respiratory tract, mucosal immune responses are important (Lappin et al., 2006). During the first weeks of life, maternally derived antibodies protect kittens against disease, although the exact time of protection varies considerably among individuals. Generally speaking, maternally derived antibodies persist for 2 to 10 weeks, with mean titers falling below detectable levels (< 1:2) by nine weeks of age (Gaskell and Povey, l9 1982). More recent studies found that about 25% of the kittens have an antibody level of < 1:4 at only 6 weeks of age. It should also be noted that kittens with low levels of maternally derived antibodies are not necessarily protected from subclinical infection and latency (Gaskell and Povey, 1982), and that these kittens may respond to early vaccination (Dawson et al., 2001). On the other hand, in some individuals, maternally derived antibodies may still be at interfering levels at 12—14 weeks of age (Dawson et al., 2001; Gaskell et al., 2007). The European ABCD recommends two vaccine injections, at 9 and 12 weeks of age, followed by yearly boosters (Thiry et a1, 2009). The American Association of Feline Practitioners Feline Vaccine Advisory Panel advices that the primary series of vaccination should begin as early as 6 weeks of age, then one dose every 3 to 4 weeks until 16 weeks of age, followed by 2 doses 3 to 4 weeks apart. A single dose is given 1 year following the last dose of the primary series, then one dose every 3 years (Richards et al., 2006). FHV-l vaccines protect cats against disease but not against infection and, as a consequence, latency. An approximately 90% reduction in clinical scores to experimental challenge has been achieved with vaccination, and virus excretion is reduced (Gaskell etal., 2007). There is complete cross-protection among FHV-l strains, as they all belong to the same single serotype. All current commercial FHV-l vaccines are multivalent, most commonly include FCV and feline panleukopenia virus components, and are collectively called feline viral rhinotracheitis, calicivirus, and panleukopenia (F VRCP) vaccines. Of the three vaccine components, the protection induced against FHV -1 is generally the least effective (Lappin et al., 2002; Scott and Geissinger, 1999). 20 A number of modified live and killed FVRCP vaccines for parenteral administration are available in the United States (Lappin et al., 2006; Richards et al., 2006). Modified-live vaccines are routinely used, but they have residual virulence and may induce clinical signs if administered incorrectly (e.g. by accidental aerosolisation or spillage on the skin) (Kruger et al., 1996). Killed virus vaccines are generally preferred for use in pregnant queens (and only if absolutely necessary) and in F eLV- or FIV-infected cats (Richards et al., 2006). In addition to vaccines labeled for systemic immunization, an intranasal multivalent vaccine including an FHV-l component is commercially available. Testing under experimental conditions showed that this vaccine (Feline UltraNasal FVRC or F VRCP Vaccine, Heska Corporation, Loveland, CO) was safe and induced protection against the clinical signs of field virus exposure within a week post-vaccination (Lappin et al., 2006). With systemically administered vaccine this would take 2-3 weeks (Lappin et al., 2009). However, it is unclear whether the new, vaccine reduces the length and level of field virus shedding. It is also unclear whether the new vaccine prevents latent infection. Several F HV-l deletion and/or insertion mutants have been constructed as potential vaccine candidates, some of which have incorporated other genes including the FCV capsid gene (Cole et al., 1990; Mishima et al., 2001; Mishima et al., 2002; Wardley et al., 1992; Yokoyarna, 1995, 1996a, 1996b, 1998). A gE—gl deletion mutant was previously developed in our laboratory (Kruger et al., 1996; Sussman et al., 1997; Sussman et al., 1995). Williamse et a1. (1996) developed a gI insertional mutant, which also appeared to affect the transcription pattern and expression of the upstream gene gD. 21 Williamse et al. (1994) also developed an FHV-l mutant by inserting B-galactosidase marker gene into UL45, adjacent to the gC gene. Yokoyama et al. (1996c, 1998) developed a TK- recombinant FHV-l vaccine. In general, these deletion/insertional mutants are less virulent for cats and offer good protection against disease, especially via the oronasal route. One reason that none has so far been marketed is probably because the protection offered is not superior to conventionally attenuated vaccines (Gaskell et al., 2007). 1.9 In vitro models of FHV-l infection FHV-l infection in cats is an excellent natural model for herpes simplex virus infection in humans, especially for ocular diseases induced by HSV-1 and -2. Other herpesvirus infections in large animals, including EHV -1, BHV -1, and PRV, have also been used extensively to study common aspects of herpesvirus biology. However, animal experiments in these species are expensive, labor-intensive, and sometimes of ethical concerns. In recent years, several in vitro systems have been developed as substitutes for in vivo animal experiments. Tracheal organ cultures have been used in several species to study physiology and infectious diseases, having the advantage over cell cultures of more closely mimicking the natural situation (Cook et al., 1976; Dhinakar Raj and Jones, 1996). This technique has only been used in one comparative study involving FHV-l and FCV, which showed that viral titers peaked later in tracheal cultures than in cell monolayers (Milek et al., 22 1976). Leeming et al. (2006) established feline tracheal organ cultures as an in vitro system for the examination of functional and morphological effects of FHV-l on the respiratory epithelium. Based upon daily assessment of cilia movement and tissue morphology, the respiratory epithelium remains viable in culture for at least 120 hours, allowing sufficient viral replication and development of lesions. Therefore, aspects of FHV-l infection in respiratory epithelium can now be studied in a model that closely mimics natural infection in airways. Recently, ex vivo cultures of nasal respiratory, nasopharygeal, and tracheal mucosa from equines have been established, the explants were maintained in vitro for up to 96 hours on fine-meshed gauze at an air-liquid interface, mimicking the air-liquid interface found in the respiratory tract of the living animal (V andekerckhove et al., 2009). Ex vivo cultures of porcine nasal mucosa have also been established and maintained for up to 60 hours post-sampling (Glorieux et al., 2007). Further, a quantitative analysis system was established to study the kinetics of horizontal and vertical spread of PRV, including crossing of the basement membrane, in the porcine nasal mucosa explant system. This system was subsequently used to examine different historical PRV strains isolated between the 19608 and 2000, and found them behave differently in the respiratory nasal mucosa (Glorieux et al., 2009). Ocular infection of F HV-l in cats is an excellent natural model for ocular diseases induced by HSV-l and -2. Conjunctival and corneal epithelial tissues are the primary targets for FHV-l in ocular infections (Bistner et al., 1971; Nasisse et al., 1989). A primary culture of PCB cells has been established and maintained in heavily-supplied medium for 6 passages, and CPE of FHV-l in these cells has been characterized 23 (Sandmeyer et al., 2005a). FCE cells have also been used to evaluate the effects of interferon-alpha and cidofovir on cell viability and replication of FHV-l (Sandmeyer et al., 2005b; Sandmeyer et al., 2005c). 1.10 Conclusions In summary, FHV-l has long been causing a widespread and very important respiratory disease in cats. Ocular lesions are also an important aspect of FHV-l infections, like is the case for HSV-1 in humans. Expanding our limited knowledge of FHV-l, particularly in the regards of gene contents and functions, molecular pathogenesis, mechanism of latent infection, and host-virus interactions, will not only benefit cats, but also contribute to the shared aspects of herpesvirus biology. Furthermore, F HV-l infection in cats has potential as a natural host system for the study of immunity against herpesvirus infections, as well as the study of antivirals and new therapeutic approaches to treatment of herpesvirus infections. 24 1.11 References Alba, M.M., Das, R., Orengo, C.A., Kellam, P., 2001. Genomewide function conservation and phylogeny in the Herpesviridae. Genome Res 11, 43-54. Baranowski, E., Keil, G., Lyaku, J., Rijsewijk, F.A., van Oirschot, J.T., Pastoret, P.P., Thiry, E., 1996. Structural and functional analysis of bovine herpesvirus 1 minor glycoproteins. Vet Microbiol 53, 91-101. Bistner, S.I., Carlson, J.H., Shively, J.N., Scott, F.W., 1971. Ocular manifestations of feline herpesvirus infection. J Am Vet Med Assoc 159, 1223-37. Brunetti, C.R., Dingwell, K.S., Wale, C., Graham, F.L., Johnson, DC, 1998. Herpes simplex virus gD and Virions accumulate in endosomes by mannose 6-phosphate- dependent and -independent mechanisms. J Virol 72, 3330-9. Cocchi, F., Menotti, L., Mirandola, P., Lopez, M., Campadelli-Fiume, G., 1998. The ectodomain of a novel member of the immunoglobulin subfamily related to the poliovirus receptor has the attributes of a bona fide receptor for herpes simplex virus types 1 and 2 in human cells. J Virol 72, 9992-10002. Cole, G.E., Stacy-Phipps, S., Nunberg, J .H., 1990. Recombinant feline herpesviruses expressing feline leukemia virus envelope and gag proteins. J Virol 64, 4930-8. Cook, J.K., Darbyshire, J.H., Peters, R.W., 1976. The use of chicken tracheal organ cultures for the isolation and assay of avian infectious bronchitis virus. Arch Virol 50, 109-18. Costes, B., Thirion, M., Dewals, B., Mast, J ., Ackermann, M., Markine-Goriaynoff, N., Gillet, L., Vanderplasschen, A., 2006. Felid herpesvirus 1 glycoprotein G is a structural protein that mediates the binding of chemokines on the viral envelope. Microbes Infect 8, 2657-67. Davison, A.J., Eberle, R., Ehlers, B., Hayward, G.S., McGeoch, D.J., Minson, A.C., Pellett, P.E., Roizman, B., Studdert, M.J., Thiry, E., 2009. The order Herpesvirales. Arch Virol 154, 171-7. Dawson, D.A., Carman, J., Collins, J., Hill, 8., Lappin, M.R., 1998. Enzyme-linked immunosorbent assay for detection of feline herpesvirus 1 IgG in serum, aqueous humor, and cerebrospinal fluid. J Vet Diagn Invest 10, 315-9. Dawson, 8., Willoughby, K., Gaskell, R.M., Wood, G., Chalmers, W.S., 2001. A field trial to assess the effect of vaccination against feline herpesvirus, feline calicivirus and feline panleucopenia virus in 6-week-old kittens. J Feline Med Surg 3, 17-22. 25 Demmin, G.L., Clase, A.C., Randall, J .A., Enquist, L.W., Banfield, B.W., 2001. Insertions in the gG gene of pseudorabies virus reduce expression of the upstream Us3 protein and inhibit cell-to-cell spread of virus infection. J Virol 75, 10856-69. Devlin, J.M., Browning, G.F., Hartley, C.A., Gilkerson, J.R., 2007. Glycoprotein G deficient infectious laryngotracheitis virus is a candidate attenuated vaccine. Vaccine 25, 3561-6. Dhinakar Raj, G., Jones, R.C., 1996. Protectotypic differentiation of avian infectious bronchitis viruses using an in vitro challenge model. Vet Microbiol 53, 239-52. Drummer, H.E., Studdert, M.J., Crabb, BS, 1998. Equine herpesvirus-4 glycoprotein G is secreted as a disulphide-linked homodimer and is present as two homodimeric species in the virion. J Gen Virol 79, 1205-13. Fargeaud, D., Jeannin, C.B., Kato, F., Chappuis, G., 1984. Biochemical study of the Feline Herpesvirus 1. Identification of glycoproteins by affinity. Arch Virol 80, 69-82. Flint, S.J., Enquist, L.W., Racaniello, V.R., Skalka, A.M., 2004. Principles of virology: molecular biology, pathogenesis, and control of animal viruses, second ed. ASM Press, Washington. Fontenelle, J .P., Powell, C.C., Veir, J .K., Radecki, S.V., Lappin, M.R., 2008. Effect of topical ophthalmic application of cidofovir on experimentally induced primary ocular feline herpesvirus-1 infection in cats. Am J Vet Res 69, 289-93. Fries, L.F., Friedman, H.M., Cohen, G.H., Eisenberg, R.J., Hammer, C.H., Frank, M.M., 1986. Glycoprotein C of herpes simplex virus 1 is an inhibitor of the complement cascade. J Irnmunol 137, 1636-41. Frink, R.J., Eisenberg, R., Cohen, G., Wagner, E.K., 1983. Detailed analysis of the portion of the herpes simplex virus type 1 genome encoding glycoprotein C. J Virol 45, 634-47. Gaskell, R., Dawson, 8., Radford, A., Thiry, E., 2007. Feline herpesvirus. Vet Res 38, 337-54. Gaskell, R.M., Povey, R.C., 1977. Experimental induction of feline viral rhinotracheitis virus re-excretion in F VR-recovered cats. Vet Rec 100, 128-33. Gaskell, R.M., Povey, R.C., 1979. The dose response of cats to experimental infection with feline viral rhinotracheitis virus. J Comp Pathol 89, 179-91. Gaskell, R.M., Povey, R.C., 1982. Transmission of feline viral rhinotracheitis. Vet Rec 111, 359-62. 26 Geraghty, R.J., Krummenacher, C., Cohen, G.H., Eisenberg, R.J., Spear, P.G., 1998. Entry of alphaherpesviruses mediated by poliovirus receptor-related protein 1 and poliovirus receptor. Science 280, 1618-20. Glorieux, S., Favoreel, H.W., Meesen, G., de Vos, W., Van den Broeck, W., Nauwynck, H.J., 2009. Different replication characteristics of historical pseudorabies virus strains in porcine respiratory nasal mucosa explants. Vet Microbiol 136, 341 -6. Glorieux, S., Van den Broeck, W., van der Meulen, K.M., Van Reeth, K., Favoreel, H.W., Nauwynck, H.J., 2007. In vitro culture of porcine respiratory nasal mucosa explants for studying the interaction of porcine viruses with the respiratory tract. J Virol Methods 142, 105-12. Grail, A., Harbour, D.A., Chia, W., 1991. Restriction endonuclease mapping of the genome of feline herpesvirus type 1. Arch Virol 116, 209-20. Hamano, M., Maeda, K., Mizukoshi, F ., Mochizuki, M., Tohya, Y., Akashi, H., Kai, K., 2004. Genetic rearrangements in the gC gene of the feline herpesvirus type 1. Virus Genes 28, 55-60. Herold, B.C., WuDunn, D., Soltys, N., Spear, P.G., 1991. Glycoprotein C of herpes simplex virus type 1 plays a principal role in the adsorption of virus to cells and in infectivity. J Virol 65, 1090-8. Huang, J ., Hartley, C.A., Ficorilli, N.P., Crabb, B.S., Studdert, M.J., 2005. Glycoprotein G deletion mutants of equine herpesvirus 1 (EHVl; equine abortion virus) and EHV4 (equine rhinopneumonitis virus). Arch Virol 150, 2583-92. Jacobi, S., 2008. Molecular pathology of FHV-l infection within feline cornea, conjunctiva, lacrimal gland, nictitans gland, trigeminal ganglion and ciliary ganglion, M.S. thesis, Michigan State University. Jacobs, L., 1994. Glycoprotein E of pseudorabies virus and homologous proteins in other alphaherpesvirinae. Arch Virol 137, 209-28. Kruger, J.M., Sussman, M.D., Maes, R.K., 1996. Glycoproteins g1 and gE of feline herpesvirus-1 are virulence genes: safety and efficacy of a gl-gE deletion mutant in the natural host. Virology 220, 299-308. Lappin, M.R., Andrews, J ., Simpson, D., Jensen, W.A., 2002. Use of serologic tests to predict resistance to feline herpesvirus 1, feline calicivirus, and feline parvovirus infection in cats. J Am Vet Med Assoc 220, 38-42. Lappin, M.R., Sebring, R.W., Porter, M., Radecki, S.J., Veir, J., 2006. Effects of a single dose of an intranasal feline herpesvirus 1, calicivirus, and panleukopenia 27 vaccine on clinical signs and virus shedding afier challenge with virulent feline herpesvirus 1. J Feline Med Surg 8, 158-63. Lappin, M.R., Veir, J ., Hawley, J ., 2009. Feline panleukopenia virus, feline herpesvirus- 1, and feline calicivirus antibody responses in seronegative specific pathogen-free cats after a single administration of two different modified live FVRCP vaccines. J Feline Med Surg 11, 159-62. Leeming, G., Meli, M.L., Cripps, P., Vaughan-Thomas, A., Lutz, H., Gaskell, R., Kipar, A., 2006. Tracheal organ cultures as a useful tool to study Felid herpesvirus 1 infection in respiratory epithelium. J Virol Methods 138, 191-5. Lubinski, J., Wang, L., Mastellos, D., Sahu, A., Lambris, J.D., Friedman, H.M., 1999. In vivo role of complement-interacting domains of herpes simplex virus type 1 glycoprotein gC. J Exp Med 190, 1637-46. Maeda, K., Horimoto, T., Mikami, T., 1998. Properties and functions of feline herpesvirus type 1 glycoproteins. J Vet Med Sci 60, 881-8. Maeda, K., Yokoyama, N., Fujita, K., Maejima, M., Mikami, T., 1997a. Heparin- binding activity of feline herpesvirus type 1 glycoproteins. Virus Res 52, 169-76. Maeda, K., Yokoyama, N., Fujita, K., Mikami, T., 1997b. Identification and characterization of the feline herpesvirus type 1 glycoprotein C gene. Virus Genes 14, 105-9. Maggs, D.J., 2005. Update on pathogenesis, diagnosis, and treatment of feline herpesvirus type 1. Clin Tech Small Anim Pract 20, 94-101. Maggs, D.J., Collins, B.K., Thorne, J.G., Nasisse, MP, 2000. Effects of L-lysine and L-arginine on in vitro replication of feline herpesvirus type-1. Am J Vet Res 61, 1474-8. Maggs, D.J., Nasisse, M.P., Kass, RH, 2003. Efficacy of oral supplementation with L- lysine in cats latently infected with feline herpesvirus. Am J Vet Res 64, 37-42. Meme R., Lessels, N.S., Webb, 5., Meek, M., Graham, P.G., Vitale, c., Norris, J.M., Power, H., 2009. Treatment of feline herpesvirus-l associated disease in cats with fameiclovir and related drugs. J Feline Med Surg 11, 40-8. Mettenleiter, T.C., Zsak, L., Zuckermann, F ., Sugg, N., Kern, H., Ben-Porat, T., 1990. Interaction of glycoprotein gHI with a cellular heparinlike substance mediates adsorption of pseudorabies virus. J Virol 64, 278-86. Milek, M., Wooley, R.E., Blue, J .L., 1976. Replication of feline herpesvirus and feline calicivirus in cell and organ cultures. Am J Vet Res 37, 723-4. 28 Mishima, M., Xuan, X., Nishikawa, Y., Makala, L., Yokoyama, N., Nagasawa, H., Mikami, T., 2001. Construction of recombinant feline herpesvirus type 1 expressing Toxoplasma gondii surface antigen 1. Mol Biochem Parasitol 117, 103-6. Mishima, M., Xuan, X., Yokoyama, N., Igarashi, I., Fujisaki, K., Nagasawa, H., Mikami, T., 2002. Recombinant feline herpesvirus type 1 expressing Toxoplasma gondi ROP2 antigen inducible protective immunity in cats. Parasitol Res 88, 144- 9. Murphy, F.A., Gibbs, E.P.J., Horzinek, M.C., Studdert, M.J., 1999. Veterinary virology, third ed. Academic Press, San Diego. Nasisse, MP, 1990. Feline herpesvirus ocular disease. Vet Clin North Am Small Anim Pract 20, 667-80. Nasisse, MP, 1995. Diagnosis and treatment of conjunctival and corneal diseases. Vet Q 17 Suppl 1, S42-3. Nasisse, M.P., Dorman, D.C., Jamison, K.C., Weigler, B.J., Hawkins, E.C., Stevens, J .B., 1997. Effects of valacyclovir in cats infected with feline herpesvirus 1. Am J Vet Res 58, 1141-4. Nasisse, M.P., Guy, J.S., Davidson, M.G., Sussman, W.A., Fairley, N.M., 1989. Experimental ocular herpesvirus infection in the cat. Sites of virus replication, clinical features and effects of corticosteroid administration. Invest Ophthahnol Vis Sci 30, 1758-68. Okazaki, K., Matsuzaki, T., Sugahara, Y., Okada, J ., Hasebe, M., Iwamura, Y., Ohnishi, M., Kanno, T., Shimizu, M., Honda, E., et al., 1991. BHV-l adsorption is mediated by the interaction of glycoprotein gIII with heparinlike moiety on the cell surface. Virology 181, 666-70. Osterrieder, N., 1999. Construction and characterization of an equine herpesvirus 1 glycoprotein C negative mutant. Virus Res 59, 165-77. Owens, J .G., Nasisse, M.P., Tadepalli, S.M., Dorman, DC, 1996. Pharmacokinetics of acyclovir in the cat. J Vet Pharrnacol Ther 19, 488-90. Pellett, P.E., Roizman, B., 2007. The family Herpesviridae: a brief introduction, in: Knipe, D.M., Howley, P.M. (Eds.), Fields Virology. Lippincott Williams & Wilkins, Philadelphia, pp. 2479-99. Povey, R.C., 1979. A review of feline viral rhinotracheitis (feline herpesvirus I infection). Comp Irnmunol Microbiol Infect Dis 2, 373-87. 29 Reynolds, A.E., Ryckrnan, B.J., Baines, J.D., Zhou, Y., Liang, L., Roller, R.J., 2001. U(L)31 and U(L)34 proteins of herpes simplex virus type 1 form a complex that accumulates at the nuclear rim and is required for envelopment of nucleocapsids. J Virol 75, 8803-17. Richards, J.R., Elston, T.H., Ford, R.B., Gaskell, R.M., Hartrnann, K., Hurley, K.F., Lappin, M.R., Levy, J.K., Rodan, I., Scherk, M., Schultz, R.D., Sparkes, A.H., 2006. The 2006 American Association of Feline Practitioners Feline Vaccine Advisory Panel report. J Am Vet Med Assoc 229, 1405-41. Roizman, B., Knipe, D.M., Whitley, R.J., 2007. Herpes simplex viruses, in: Knipe, D.M., Howley, P.M. (Eds.), Fields Virology. Lippincott Williams & Wilkins, Philadelphia, pp. 2502-601. Rota, F.A., Maes, R.K., Ruyechan, W.T., 1986. Physical characterization of the genome of feline herpesvirus-l. Virology 154, 168-79. Sandmeyer, L.S., Keller, C.B., Bienzle, D., 2005a. Culture of feline corneal epithelial cells and infection with feline herpesvirus-l as an investigative tool. Am J Vet Res 66, 205-9. Sandmeyer, L.S., Keller, C.B., Bienzle, D., 2005b. Effects of cidofovir on cell death and replication of feline herpesvirus-l in cultured feline corneal epithelial cells. Am J Vet Res 66, 217-22. Sandmeyer, L.S., Keller, C.B., Bienzle, D., 2005c. Effects of interferon-alpha on cytopathic changes and titers for feline herpesvirus-1 in primary cultures of feline corneal epithelial cells. Am J Vet Res 66, 210-6. Scott, F.W., Geissinger, C.M., 1999. Long-term immunity in cats vaccinated with an inactivated trivalent vaccine. Am J Vet Res 60, 652-8. Snoeck, R., 2000. Antiviral therapy of herpes simplex. Int J Antimicrob Agents 16, 157- 9. Spatz, S.J., 1993. Genetic Analysis of the Genes Encoding the Major Glycoproteins of Feline Herpesvirus Type 1, PhD. thesis, Michigan State University. Stiles, J ., 2000. Feline herpesvirus. Vet Clin North Am Small Anim Pract 30, 1001-14. Stiles, J., Townsend, W.M., Rogers, Q.R., Krohne, S.G., 2002. Effect of oral administration of L-lysine on conjunctivitis caused by feline herpesvirus in cats. Am J Vet Res 63, 99-103. 30 Sussman, M.D., Maes, R.K., Kruger, J.M., 1997. Vaccination of cats for feline rhinotracheitis results in a quantitative reduction of virulent feline herpesvirus-1 latency load after challenge. Virology 228, 379-82. Sussman, M.D., Maes, R.K., Kruger, J.M., Spatz, S.J., Venta, P.J., 1995. A feline herpesvirus-1 recombinant with a deletion in the genes for glycoproteins g1 and gE is effective as a vaccine for feline rhinotracheitis. Virology 214, 12-20. Thiry, E., Addie, D., Belak, S., Boucraut-Baralon, C., Egberink, H., Frymus, T., Gruffydd-Jones, T., Hartmann, K., Hosie, M.J., Lloret, A., Lutz, H., Marsilio, F., Pennisi, M.G., Radford, A.D., Truyen, U., Horzinek, M.C., 2009. Feline herpesvirus infection. ABCD guidelines on prevention and management. J Feline Med Surg 11, 547-55. Vandekerckhove, A., Glorieux, S., Broeck, W.V., Gryspeerdt, A., van der Meulen, K.M., Nauwynck, H.J., 2009. In vitro culture of equine respiratory mucosa explants. VetJ 181, 280-7. Vogtlin, A., Fraefel, C., Albini, S., Leutenegger, C.M., Schraner, E., Spiess, B., Lutz, H., Ackermann, M., 2002. Quantification of feline herpesvirus 1 DNA in ocular fluid samples of clinically diseased cats by real-time TaqMan PCR. J Clin Microbiol 40, 519-23. Walton, T.E., Gillespie, J.H., 1970. Feline viruses. VII. Immunity to the feline herpesvirus in kittens inoculated experimentally by the aerosol method. Cornell Vet 60, 232-9. Wardley, R.C., Berlinski, P.J., Thomsen, D.R., Meyer, A.L., Post, LE, 1992. The use of feline herpesvirus and baculovirus as vaccine vectors for the gag and env genes of feline leukaemia virus. J Gen Virol 73, 1811-8. Wardley, R.C., Rouse, B.T., Babiuk, L.A., 1976. Observations on recovery mechanisms from feline viral rhinotracheitis. Can J Comp Med 40, 257-64. Willemse, M.J., Chalmers, W.S., Cronenberg, A.M., Pfundt, R., Strijdveen, I.G., Sonderrneijer, P.J., 1994. The gene downstream of the gC homologue in feline herpes virus type 1 is involved in the expression of virulence. J Gen Virol 75, 3107-16. Willemse, M.J., Chalmers, W.S., Sondermeijer, P.J., 1996. In vivo properties of a feline herpesvirus type 1 mutant carrying a lacZ insertion at the g1 locus of the unique short segment. Vaccine 14, 1-5. Xuan, X., Horimoto, T., Limcumpao, J .A., Tohya, Y., Takahashi, E., Mikami, T., 1992. Glycoprotein-specific immune response in canine herpesvirus infection. Arch Virol 122, 359-65. 31 Yokoyama, N., Fujita, K., Damiani, A., Sato, E., Kurosawa, K., Miyazawa, T., Ishiguro, S., Mochizuki, M., Maeda, K., Mikami, T., 1998. Further development of a recombinant feline herpesvirus type 1 vector expressing feline calicivirus immunogenic antigen. J Vet Med Sci 60, 717-23. Yokoyama, N., Maeda, K., Fujita, K., Ishiguro, S., Sagawa, T., Mochizuki, M., Tohya, Y., Mikami, T., 1996a. Vaccine efficacy of recombinant feline herpesvirus type 1 expressing immunogenic proteins of feline calicivirus in cats. Arch Virol 141, 2339-51. Yokoyama, N., Maeda, K., Kawaguchi, Y., Ono, M., Tohya, Y., Mikami, T., 1995. Construction of the recombinant feline herpesvirus type I deleted thymidine kinase gene. J Vet Med Sci 57, 709-14. Yokoyama, N., Maeda, K., Tohya, Y., Kawaguchi, Y., Fujita, K., Mikami, T., 1996b. Recombinant feline herpesvirus type 1 expressing immunogenic proteins inducible virus neutralizing antibody against feline calicivirus in cats. Vaccine 14, 1657-63. Yokoyama, N., Maeda, K., Tohya, Y., Kawaguchi, Y., Shin, Y.S., Ono, M., Ishiguro, S., Fujikawa, Y., Mikami, T., 1996c. Pathogenicity and vaccine efficacy of a thymidine kinase-deficient mutant of feline herpesvirus type 1 in cats. Arch Virol 141, 481-94. 32 CHAPTER 2 BACTERIAL ARTIFICIAL CHROMOSOME (BAC) AND RECOMBINATION—MEDIATED GENETIC ENGINEERING (RECOMBINEERIN G) 33 2.1 BAC Cloning Traditionally, functional studies of viral genes have been achieved by creating mutant alleles and screening for phenotypic alterations, an approach known as reverse genetics. Because the natural mutation rate in DNA viruses sUch as herpesviruses is low, experimental generation of viral mutants is necessary. However, the fact that the frequency of the experimentally induced mutation must be controllable, in order to prevent accumulation of multiple mutations, makes the process inefficient. In addition, to map these mutations, time-consuming and labor-intensive procedures are required (Schaffer, 1975; Schaffer et al., 1984). Advances in molecular cloning technologies facilitated functional Studies at the level of isolated genes. Viral genes and genomic fragments can be cloned, then manipulated in vitro and reintroduced into the viral genome. Nevertheless, due to the limitations in the capacity of vectors, this method allowed only step-by-step analysis for large and complex genomes. In the late 1980s herpesvirus genomes were subcloned as a set of overlapping cosmid clones in Escherichia coli. After co-transfecting cells with the cosmid set, infectious virus can be reconstituted by multiple homologous recombinations via the overlapping sequences (van Zijl et al., 1988). Mutations can be introduced into one of the fiagrnents. Because recombination procedures in cells cannot be controlled, mutagenesis is often associated with unwanted and illegitimate mutations and extensive post-mutagenesis analysis of the isolated mutants is therefore required. Null mutants of an essential gene can only be propagated in cell lines that provide the gene product in trans (DeLuca and Schaffer, 1985). In addition, the successful use of these strategies is determined by the replication efficiency of the virus under study. The 34 plentiful selection of mutants available for herpes simplex viruses is in contrast to the paucity of mutants of other herpesviruses that remain cell associated, that replicate slowly or that have the propensity to enter the lytic cycle only under specific conditions. Yeast artificial chromosomes (YACs) were the first cloning vectors for large genomic DNA fiagments. Unfortunately, YACs also caused difficulties, such as frequent spontaneous rearrangements and insert instability, and there can be substantial contamination of purified YACs with yeast DNA (Ramsay, 1994; Schalkwyk et al., 1995) Bacterial artificial chromosomes (BACs) are single copy F-factor-based plasmid vectors that can stably hold 300 kb or more of foreign DNA (Shizuya et al., 1992). BACs have several advantages over the other methods. First of all, BACs are much more stable than other vectors, because the strict control of the F-factor replicon maintains a single copy of the BAC per bacterial cell. This reduces the risk of otherwise frequent recombination events via repetitive DNA elements present in the DNA inserts (Kimman et al., 1994; Shizuya et al., 1992). The capacity and stability of BACs enables the cloning of an entire herpesvirus genome into a single plasmid. Secondly, for subsequent fimctional genetics study, once a viral genome is cloned into a BAC, it can be manipulated within E. coli. Utilizing prokaryotic recombinases, such as recA, recE, recT (Horsburgh et al., 1999; Link et al., 1997; Narayanan et al., 1999) or the mini- lambda system (Costantino and Court, 2003; Court et al., 2003), site-specific mutations can be introduced, theoretically anywhere in the viral genome. All mutagenesis steps can be controlled and analyzed in E. coli, and the manipulated viral genome can be stably maintained in the E. coli. This is in contrast to the other methods, where the 35 recombination takes place in eukaryotic cells and the analyses can only start after the virus has been reconstituted and isolated. Unwanted additional changes that may have occurred in the viral genome, such as deletions, rearrangements or illegitimate recombinations frequently can only be observed after considerable expense of time and effort. Finally, it is safer to work with herpesviruses when the viral genome is maintained as a BAC. These properties have made BACs the vectors of choice for the cloning of eukaryotic genome libraries, and have attracted herpesvirologists as well. Several herpesvirus genomes of medical and veterinary importance have been cloned in BAC since the first infectious BAC of herpesvirus reported in 1997 (Messerle et al., 1997), including herpes simplex virus (Saeki et al., 1998; Stavropoulos and Strathdee, 1998), Epstein-Barr virus (Delecluse et al., 1998), human cytomegalovirus (Borst et al., 1999; Marchini et al., 2001; Yu et al., 2002), pseudorabies virus (Smith and Enquist, 1999), equine herpesvirus 1 (Rudolph et al., 2002), and Marek’s disease virus Wiikura et al., 2006; Schumacher et al., 2000). 2.2 Recombineering Recombineering is a powerful method for fast and efficient manipulation of the BAC. (See Figure 2 for overview.) It allows DNA cloned in E. coli to be modified via lambda (k) Red-mediated homologous recombination, obviating the need for restriction enzymes and DNA ligases. Specific bacterial strains have been constructed for this purpose (Lee et al., 2001; Warming et al., 2005; Y‘u et al., 2000). A defective I. prophage (mini- it) is inserted into their bacterial genome and encodes three genes that 36 make recombineering possible: exo, bet and gam. Exo is a 5'-3' exonuclease that creates single-stranded overhangs on linear DNA introduced into the bacteria. Bet protects these overhangs and assists in the subsequent recombination process. Garn prevents degradation of linear DNA by inhibiting E. coli RecBCD protein. Exo, bet, and gam are transcribed from the XPL promoter. This promoter is repressed by the temperature- sensitive repressor c1857 at 32°C and derepressed (the repressor is inactive) at 42°C. When bacteria containing mini-X prophage are kept at 32°C, no recombination proteins are produced. However, after 15 minutes of heat shock at 42°C, sufficient amounts of recombination proteins are produced. Linear DNA (PCR product, oligonucleotide, etc.) with sufficient homology in the 5' and 3' ends to a target DNA molecule already present in the bacteria (plasmid, BAC, or the bacterial genome itself) can be introduced into heat-shocked and electrocompetent bacteria using electroporation. The introduced DNA will now be modified by Exo and Bet and undergo homologous recombination with the target molecule. In our case, once a viral genome is cloned into a BAC, it can be easily and efficiently manipulated within E. coli. Utilizing recombineering techniques, site- specific mutations can be introduced anywhere in the viral genome, provided that the genomic sequence is known. All mutagenesis steps can be strictly controlled and analyzed in E. coli, and the manipulated viral genome can be stably maintained in the E. coli. This is in contrast to the other methods, where the recombination takes place in eukaryotic cells and the analyses can only start after the virus has been reconstituted and isolated. Unwanted additional changes that may have occurred in the viral genome, such as deletions, rearrangements or illegitimate recombinations frequently can only be observed after considerable expense of time and effort. 37 In 2006, Tischer et al. published a technique called two-step Red-mediated recombination, which further improves the efficiency of recombineering. This technique is suitable for site-specific deletion, insertion, and point mutation. An overview of this technique is provided in Figure l of Chapter 4. 38 2.3 References Borst, E.M., Hahn, G., Koszinowski, U.H., Messerle, M., 1999. Cloning of the human cytomegalovirus (HCMV) genome as an infectious bacterial artificial chromosome in Escherichia coli: a new approach for construction of HCMV mutants. J Virol 73, 8320-9. Costantino, N., Court, D.L., 2003. Enhanced levels of lambda Red-mediated recombinants in mismatch repair mutants. Proc Natl Acad Sci U S A 100, 15748- 53. Court, D.L., Swaminathan, 8., Yu, D., Wilson, H., Baker, T., Bubunenko, M., Sawitzke, J ., Sharan, SK, 2003. Mini-lambda: a tractable system for chromosome and BAC engineering. Gene 315, 63-9. Delecluse, H.J., Hilsendegen, T., Pich, D., Zeidler, R., Hammerschmidt, W., 1998. Propagation and recovery of intact, infectious Epstein-Barr virus from prokaryotic to human cells. Proc Natl Acad Sci U S A 95, 8245-50. DeLuca, N.A., Schaffer, P.A., 1985. Activation of immediate-early, early, and late promoters by temperature-sensitive and wild-type forms of herpes simplex virus type 1 protein ICP4. Mol Cell Biol 5, 1997-2008. Horsburgh, B.C., Hubinette, M.M., Qiang, D., MacDonald, M.L., Tufaro, F., 1999. Allele replacement: an application that permits rapid manipulation of herpes simplex virus type 1 genomes. Gene Ther 6, 922-30. Kimman, T.G., De Wind, N., De Bruin, T., de Visser, Y., Voermans, J., 1994. Inactivation of glycoprotein gE and thymidine kinase or the US3-encoded protein kinase synergistically decreases in vivo replication of pseudorabies virus and the induction of protective immunity. Virology 205, 511-8. Lee, E.C., Yu, D., Martinez de Velasco, J., Tessarollo, L., Swing, D.A., Court, D.L., Jenkins, N.A., Copeland, N.G., 2001. A highly efficient Escherichia coli-based chromosome engineering system adapted for recombinogenic targeting and subcloning of BAC DNA. Genomics 73, 56-65. Link, A.J., Phillips, D., Church, G.M., 1997. Methods for generating precise deletions and insertions in the genome of wild-type Escherichia coli: application to open reading frame characterization. J Bacteriol 179, 6228-37. Marchini, A., Liu, H., Zhu, H., 2001. Human cytomegalovirus with IE2 (UL122) deleted fails to express early lytic genes. J Virol 75, 1870-8. 39 Messerle, M., Crnkovic, I., Hammerschmidt, W., Ziegler, H., Koszinowski, U.H., 1997. Cloning and mutagenesis of a herpesvirus genome as an infectious bacterial artificial chromosome. Proc Natl Acad Sci U S A 94, 14759-63. Narayanan, K., Williamson, R., Zhang, Y., Stewart, A.F., Ioannou, P.A., 1999. Efficient and precise engineering of a 200 kb beta-globin human/bacterial artificial chromosome in E. coli DHlOB using an inducible homologous recombination system. Gene Ther 6, 442-7. Niikura, M., Dodgson, J ., Cheng, H., 2006. Direct evidence of host genome acquisition by the alphaherpesvirus Marek's disease virus. Arch Virol 151, 537-49. Ramsay, M., 1994. Yeast artificial chromosome cloning. Mol Biotechnol 1, 181-201. Rudolph, J ., O'Callaghan, D.J., Osterrieder, N., 2002. Cloning of the genomes of equine herpesvirus type 1 (EHV-l) strains KyA and racLll as bacterial artificial chromosomes (BAC). J Vet Med B Infect Dis Vet Public Health 49, 31-6. Saeki, Y., Ichikawa, T., Saeki, A., Chiocca, E.A., Tobler, K., Ackermann, M., Breakefield, X.O., Fraefel, C., 1998. Herpes simplex virus type 1 DNA amplified as bacterial artificial chromosome in Escherichia coli: rescue of replication- competent virus progeny and packaging of amplicon vectors. Hum Gene Ther 9, 2787-94. Schaffer, P.A., 1975. Temperature-sensitive mutants of herpesviruses. Curr Top Microbiol Irnmunol 70, 51-100. Schaffer, P.A., Weller, S.K., Pancake, B.A., Coen, D.M., 1984. Genetics of herpes simplex virus. J Invest Derrnatol 83, 42S-47S. Schalkwyk, L.C., Francis, F ., Lehrach, H., 1995. Techniques in mammalian genome mapping. Curr Opin Biotechnol 6, 37-43. Schumacher, D., Tischer, B.K., Fuchs, W., Osterrieder, N., 2000. Reconstitution of Marek's disease virus serotype 1 (MDV-1) from DNA cloned as a bacterial artificial chromosome and characterization of a glycoprotein B-negative MDV-1 mutant. J Virol 74, 11088-98. Shizuya, H., Birren, 8., Kim, U.J., Mancino, V., Slepak, T., Tachiiri, Y., Simon, M., 1992. Cloning and stable maintenance of 300-kilobase-pair fragments of human DNA in Escherichia coli using an F -factor-based vector. Proc Natl Acad Sci U S A 89, 8794-7. Smith, G.A., Enquist, L.W., 1999. Construction and transposon mutagenesis in Escherichia coli of a full-length infectious clone of pseudorabies virus, an alphaherpesvirus. J Virol 73, 6405-14. 40 Stavropoulos, T.A., Strathdee, C.A., 1998. An enhanced packaging system for helper- dependent herpes simplex virus vectors. J Virol 72, 7137-43. Tischer, B.K., von Einem, J ., Kaufer, B., Osterrieder, N., 2006. Two-step red-mediated recombination for versatile high-efficiency markerless DNA manipulation in Escherichia coli. Biotechniques 40, 191-7. van Zijl, M., Quint, W., Briaire, J., de Rover, T., Gielkens, A., Berns, A., 1988. ' Regeneration of herpesviruses from molecularly cloned subgenomic fragments. J Virol 62, 2191-5. Warming, 8., Costantino, N., Court, D.L., Jenkins, N.A., Copeland, N.G., 2005. Simple and highly efficient BAC recombineering using galK selection. Nucleic Acids Res 33, e36. Yu, D., Ellis, H.M., Lee, E.C., Jenkins, N.A., Copeland, N.G., Court, D.L., 2000. An efficient recombination system for chromosome engineering in Escherichia coli. Proc Natl Acad Sci U S A 97, 5978-83. Yu, D., Smith, G.A., Enquist, L.W., Shenk, T., 2002. Construction of a self-excisable bacterial artificial chromosome containing the human cytomegalovirus genome and mutagenesis of the diploid TRL/IRL13 gene. J Virol 76, 2316-28. 41 CHAPTER 3 COMPLETE GENOMIC SEQUENCE AND AN INFECTIOUS BAC CLONE OF F ELIN E HERPESVIRUS-l (FHV-l) Tai, S.H., Niikura, M., Cheng, H.H., Kruger, J .M., Wise, A.G., Maes, R.K., 2009. Complete genomic sequence and an infectious BAC clone of feline herpesvirus-1 (FHV-l), submitted to: Virology. 42 Complete Genomic Sequence and an Infectious BAC Clone of Feline Herpesvirus-1 (FHV-l) S. H. Sheldon Taia, Masahiro Niikurab’c’l, Hans H. Chenga’c, John M. Krugera’d, Annabel G. Wisee, Roger K. Maesa’b’e’* a Graduate Program in Comparative Medicine and Integrative Biology, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA b Department of Microbiology and Molecular Genetics, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA c United States Department of Agriculture, Agricultural Research Service, Avian Disease and Oncology Laboratory, 3606 East Mount Hope Road, East Lansing, MI 48823, USA d Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA c Diagnostic Center for Population and Animal Health, College of Veterinary Medicine, Michigan State University, 4125 Beaumont Road, Lansing, MI 48910, USA * Corresponding author. Diagnostic Center for Population and Animal Health, College of Veterinary Medicine, Michigan State University, 4125 Beaumont Road, Lansing, MI 48910, USA. Fax: +1 517 432 6527. E-mail address: Maes@dcpah.msu.edu (R.K. Maes). 43 1 Present address: Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, V5A IS6, Canada. ABSTRACT Infection with feline herpesvirus-1 (FHV-l) is a major cause of upper respiratory and ocular disease in Felidae. We report the first complete genomic sequence of FHV-l, as well as the construction and characterization of a bacterial artificial chromosome (BAC) clone of FHV-l, which contains the entire FHV-l genome and has the BAC vector inserted at the left-hand side (5’-end) of the UL region. Complete genomic sequences were derived from both the FHV-l BAC clone and purified virion DNA. The FHV-l genome is 135,797 bp in size with an overall G+C content of 45%. A total of 78 open reading frames were predicted, encoding. 74 distinct proteins. The gene arrangement is collinear with that of most sequenced varicelloviruses. The virus regenerated from the BAC was very similar to the parental C-27 strain in vitro in terms of plaque morphology and growth . characteristics, and highly virulent in cats in a preliminary in vivo study. Keywords: Feline herpesvirus; FHV-l; genomic sequence; infectious BAC clone INTRODUCTION Feline herpesvirus-1 (FHV-l) is a significant viral pathogen of Felidae, first isolated in 1957 by Crandell and Maurer (1958). FHV-l accounts for approximately 50% of all diagnosed viral upper respiratory infections in cats and is also a significant cause of oeular lesions (Nasisse, 1990). The pathobiology of F HV-l has been reviewed (Gaskell, 2007; Maggs, 2005; Stiles, 2003). Briefly, following entry via the oronasal route, FHV-l replicates extensively in the mucosae of the upper respiratory tract resulting in high fever, depression, anorexia, sneezing, conjunctivitis, keratitis, and ocular and nasal discharge. The acute phase of the disease is followed by life-long latency, a hallmark of herpesvirus infections. During the latent stage, viral DNA appears to persist mainly in sensory ganglia. Different biological stresses, or administration of corticosteroids, can induce the necessary biochemical stimuli in latently infected cells that lead to renewed production of infectious virus, which can travel to the periphery and is a potential source of viral transmission. In addition to the primary disease, FHV-l is also a contributor to feline chronic rhinosinusitis (Johnson and Maggs, 2005). Despite its high morbidity and worldwide distribution, currently available vaccines cannot totally protect cats from field virus infection and, as a consequence, from field virus latency (Gaskell and Povey, 1979; Tharn and Studdert, 1987). In addition, some commercial vaccines are avirulent when administered subcutaneously, but virulent when administered oronasally (Kruger et al., 1996; Povey, 1979). Since reactivation of latent FHV-l is common, vaccinated cats may act as 45 asymptomatic carriers and contribute to the spread of virulent virus (Gaskell and Povey, 1982) Our knowledge of FHV -1 at the molecular level is still limited. F HV-l is a member of Herpesviridae family, Alphaherpesvirinae subfamily, genus Varicellovirus (Davison et al., 2009). All FHV-l isolates appear to be relatively similar, and belong to one serotype. The FHV-l genome, mapped by restriction endonuclease digestion techniques, is approximately 134 kb, and has a typical type D structure similar to other varicelloviruses (Figure 3.1). The double-stranded DNA genome consists of two segments of unique sequences, known as Unique Long (UL) and Unique Short (U s)- The Us region is flanked by a pair of identical but inverted repeat sequences, known as Terminal Repeat Short (TRS) and Inverted Repeat Short (IRS) (Grail et al., 1991; Rota et al., 1986). Alphaherpesviruses usually encode 65-80 open reading flames (ORFs) (Alba et al., 2001). FHV-l has been shown to contain 23 virion-associated proteins (Fargeaud et al., 1984). Eight glycoproteins have previously been identified and designated as gB, gC, gD, gE, gG, gH, g1 and gL. Knowledge about the fimctions of these glycoproteins has been reviewed by Maeda et al. (1998) though it is clear that much more work is needed to define their actual functions and roles in viral pathogenesis and immunity. In recent years, many herpesvirus genomes of human and veterinary medical importance have been completely sequenced and annotated, among them herpes simplex virus types 1 (HSV-1) (McGeoch et al., 1988) and 2 (HSV-2) (Dolan et al., 1998), varicella-zoster virus (V ZV) (Davison and Scott, 1986), bovine herpesvirus types 1 (BHV-l) (Schwyzer and Ackermann, 1996) and 5 (BHV-S) (Delhon et al., 2003), equine herpesvirus types 1 (EHV-l) (Telford et al., 1992) and 4 (EHV-4) (Telford et al., 1998), Marek’s disease 46 virus (MDV) (Lee et al., 2000; Tulman et al., 2000), and pseudorabies virus (PRV) (Klupp et al., 2004). Prior to this report, only portions of the FHV-l genomic sequence were available. I Bacterial artificial chromosomes (BACs) are single copy F-factor-based plasmid vectors that can stably hold 300 kb or more of foreign DNA (Shizuya et al., 1992). BACs have advantages over other vectors including higher cloning capacity, greater stability in E. coli, and the efficiency of manipulation. Because of these advantages, BACs have been widely used for cloning of entire herpesvirus genomes, and the resulting clones have been shown to be infectious. Herpesviruses that have been cloned as BACs include HSV-l (Saeki et al., 1998; Stavropoulos and Strathdee, 1998), Epstein-Barr virus (Delecluse et al., 1998), human cytomegalovirus (Borst et al., 1999; Marchini etal., 2001; Yu et al., 2002), PRV (Smith and Enquist, 1999), EHV-l (Hansen et al., 2006; Rudolph et al., 2002), MDV (Schumacher et al., 2000; Niikura et al., 2006), canine herpesvirus (Arii et al., 2006; Strive et al., 2006), and FHV—l (Costes etal., 2006). In this report, we present the first complete and annotated genomic sequence of FHV-l, as well as a BAC clone that contains the entire FHV-l genome, and is infectious both in vitro and in vivo. 47 RESULTS Cloning of the FHV-l genome as a BAC The C-27 strain F HV-l genome was cloned as a BAC. U54, which encodes gG, was selected as the target site for the BAC cassette insertion because it has been shown to be non-essential for virus growth in several alphaherpesviruses (Balan et al., 1994; Baranowski et al., 1996) including FHV-l (Costes et al., 2006). Analyses later revealed that the BAC vector was in fact inserted at the genomic termini, or between UL and TRS in the circular DNA (Figure 3.1). In addition, an unknown sequence of ~2.7 kb in size was present immediately upstream of the BAC vector. Restriction pattern analysis was used to examine the overall integrity of the FHV- l genome in the BAC clone. The SalI digestion pattern of the BAC clone was very similar to that of the parent strain (Figure 3.2). In addition to the restriction pattern analysis, PCR assays were used to examine the insertion of the BAC vector. Using primers targeting different BAC vector components (Table 3.1), including the enhanced green fluorescence protein (EGFP) gene (RM0901 and RM0944, or RM0945 and RM0911), the chloramphenicol resistance gene (RM0955 and RM0956), and the bacterial origin of replication (RM0957 and RM0958), PCR assays confirmed that the BAC vector was inserted into the FHV-l genome (data not shown). However, PCR primers targeted to the expected junction between the BAC vector and the viral sequence (RM0910 and RM0959) failed, and primers RM0988 and RM0989 amplified the full length of an intact US4. These results 48 suggested that the BAC was inserted into the FHV-l genome, but not into US4 as intended. Sequencing of the FHV-l genome Using “454” next generation sequencing technology, complete genome sequences were generated de novo for both the viral genomic DNA and BAC clone, and then compared to one another. The sequence assembly of the BAC clone represented 97.62% of the entire FHV-l genome with an average read depth of 33x. The assembly of the viral DNA represented 97.52% of the entire genome with an average read depth of 25x. To complete the sequences, gaps were filled by primer walking, using both pure viral DNA and BAC DNA as template. Three major gaps were present in both assemblies. Gap no. 1 represents the junction of UL and IRS, gap no. 2 was located inside both IRS and TRS, and gap no. 3 contained the genomic termini, or the junction of TRS and UL when the genome is circularized. Based on the S011 restriction pattern, the estimated sizes for gaps l, 2, and 3 were 1.0 kb, 0.6 kb, and 1.0 kb, respectively. Attempts to close these gaps by primer walking were partially successfirl. Gaps 1 and 3 were completely closed, and the sequence lengths were as predicted. Gap 2 could not be fully sequenced. Primer walking results revealed that this region contains repetitive units of very high G+C content (77%). Use of additional sequencing chemistry (i.e., dGTP BigDye Terminator) failed to sequence through this gap. The remaining ~314 bp lacking sequence was tentatively filled with the same repetitive unit found at both ends of the gap. No ORFs were found inside or spanning these repeats. 49 When sequences derived flom the viral genome and BAC were compared to each other, they were completely identical, except for the insertion of the BAC vector between UL and TRS in the BAC clone, an addition of a 2.7-kb fragment (Contig 3) in the BAC clone, and three aforementioned gaps whose sequence could only be determined flom the BAC clone. A BLASTN search against the non-redundant nucleotide collection database nr/nt (Altschul et al., 1997) found that Contig 3 did not resemble any known herpesvirus sequence. A BLAST search in the WGS Contigs database of the cat genome project (http://www.ncbi.nlm.nih.gov/projects/genome/seq/BlastGen/BlastGen.cgi?pid=10703) revealed that a 219-bp portion near the right hand side (5’-end) of Contig 3 is highly similar to a genomic region of cat DNA. PCR primers RM1025 and RM1026 (Table 3.1) targeting sequences near the left hand side (3’-end) of Contig 3 successfully amplified the same 225-bp sequence flom the DNA of uninfected Crandell-Reese feline kidney (CRFK) cells (data not shown). Therefore, it is very likely that Contig 3 is a part of the cat genome, presumably acquired by the virus during the recombination process in CRFK cells. The complete, FHV-l genomic sequence was compared to all FHV-l DNA sequences in GenBank that contained at least one complete gene. The results are presented in Table 3.2. The DNA sequences were highly similar across strains. The differences observed were mostly limited to single bases. It was noted that the copy number of repetitive sequences can vary between strains, or even within the same strain. The complete DNA sequences of the FHV-l genome and the FHV-l BAC clone were deposited in GenBank under accessions F J478159 and GU250525 respectively. 50 Structure of the FHV-l Genome The FHV-l genome is 135,797 bp in length. The overall G+C content of FHV-l genome is 45%. The genome consists of a 106,369 bp UL and a 8,424 bp Us region, with the former being flanked by an inverted 33-bp sequence, and the latter being flanked by IRS and TRs elements of 10,469 bp each. ORF finding, gene content, and gene arrangement To identify all proteins encoded in the genome, we employed two methods. First, ORFs encoding proteins of Z 60 amino acids with a methionine start codon were evaluated for coding potential by searching for homologs in other alphaherpesviruses. Homology searches were conducted using BLASTX on the non-redundant protein sequence database nr (Altschul et al., 1997). Other criteria used included compact gene arrangements on both strands with little gene overlap. To find novel genes in the FHV-l genome ab initio and as a second approach to verify the annotation, the sequence was submitted to GeneMarkS (Besemer et al., 2001). GeneMarkS predicted 10 fewer genes, namely UL3, ULI 1, UL24, UL26.5, UL43, UL53, U52, US6, US8.5, and US9. However, these genes are consistently present among varicelloviruses (Tables 3.3 and 3.4), and were easily identified using BLASTX, except for US2. Despite the low degree of sequence similarity, US2 was also predicted based on relative position similar to other varicelloviruses. Table 3.3 lists all FHV-l ORFs predicted in the genomic sequence and summarizes the characteristics of the predicted gene products. The arrangement of these 51 ORFs in the FHV-l genome is shown in Figure 3.3. Seventy-eight ORFs were predicted in the FHV-l genome, encoding 74 different proteins, as genes encoding ICP4, U81, and US10 proteins are found twice, once in the IRS and once in the TRS, and UL15 consists of two ORFs. Seventy-two ORFs are present as single copies with 64 located in the UL, seven located entirely in the Us, and one that initiates in the Us and ends in the TRS. Three ORFs are located entirely in the repeat region, each present once in IRS and once in TRS. All ORF start locations were assumed to be the first possible ATG. The name of each protein was given based on its homology to HSV-1 and VZV genes. The predicted properties and functions assigned to each predicted gene were based on those assigned to other varicelloviruses by Refseq entries in the NCBI database. In addition to the ORFs listed in Table 3.3, 53 ORFs with a coding capacity of more than 60 amino acids were identified: 25 were found on the top strand and 28 were on the bottom strand. However, searches for cellular or viral homologs of these ORFs failed to find any significant match, and none of these ORFs were considered as strong candidates for new genes. Table 3.3 also lists the amino acid sequence similarity of each protein to their counterparts in the other sequenced varicelloviruses. All F HV-l gene products showed some degree of homology to the gene products of the other varicelloviruses. Not surprisingly, genes involved in nucleotide metabolism, DNA replication and packaging were among the most conserved. Glycoprotein genes were less conserved, although gB did show a high degree of similarity to gB of the other varicelloviruses. FHV-l proteins were most similar to homologues of EHV-l and EHV-4, averaging 49.4% and 49.2% animo acid identity, respectively. Phylogenetic relationships between FHV-l and other 52 varicelloviruses were examined using single glycoprotein and concatenated amino acid sequence alignments (McGeoch et al., 2000). The results are presented in Figure 3.4 and showed that FHV-l is most closely related to EHV-l and EHV-4. The arrangement of FHV-l genes is collinear with VZV, BHV-l, BHV-5, EHV-l and EHV-4. The gene content is highly similar to those of varicelloviruses, especially EHV-l and -4, with a few exceptions (Table 3.4). FHV-l lacks the homologs of V67 and US5, while both are present in EHV-l and -4. V67 was found in EHV-l, EHV-4, BHV -1, BHV-S, and VZV, but is absent in PRV. US5, encoding g], was found only in EHV-l and -4. None of the genes identified were unique to FHV-l. Polyadenylation signals and splice sites The PolyADQ program (Tabaska and Zhang, 1999) was used to search for all potential polyadenylation signals in the FHV-l genome. The search found 323 sites; each was given a score between 0 and 1. In the PRV study (Klupp et al., 2004), experimental results were used to determine a cut off score. Since there were very few, if any, data for the mapping of F HV-l transcripts, every signal was located in the complete genomic sequence initially. Many signals did not associate with any upstream gene, while very often multiple signals were found for a given ORF or a set of coterrninal transcripts. The polyadenylation signals for each gene were annotated based on the assumption that each transcript either ends near the termination codon or is a member of a 3’-coterminal family. The ones that are closest to the stop codon and have the highest scores were annotated. 53 UL15 is made up of two exons and is well conserved among herpesviruses. The region between and including the two exons of UL15 were submitted to the Neural Network Splice Site Prediction program conditioned for human splice site recognition (Reese et al., 1997). The predicted donor site with the highest score (0.98) located 39 bp upstream flom the end of the first ORF of UL15. The predicted acceptor site with the highest score (0.99) located between the ORF of ULI 7 and the second ORF of UL15. The spliced mRNA would encode a protein of 734 amino acids. Origins of replication and tandem repeats The origins of replication of herpesviruses are characterized by the presence of an AT-rich palindromic sequence. EHV -1, EHV-4, and PRV have at least 3 origins of DNA replication: one copy of OriL found in UL, between ULZI and UL22, and two copies of OriS found in IRS and TRS (Klupp et al., 2004; Telford et al., 1992; Telford et al., 1998). In contrast, VZV lacks an OriL (Davison and Scott, 1986). In the FHV-l genome, the OriS palindrome was found in IRS and TRS. However, no sequence that resembles an OriL was found in the region between UL21 and UL22. Comparison of the DNA sequences in this region between the BAC clone and wild type C-27 strain (data not shown) has ruled out the possibility that a similar palindrome might have been mis- assembled or deleted during cloning. Using the Tandem Repeats Finder program (Benson, 1999), eight different types of tandem repeat elements at 16 locations in the FHV -1 genome were identified (Table 3.5). One tandem repeat element was found in UL, one in Us, and six in IRS/TRS. Four tandem repeat elements were located near genomic termini. In addition, the Tandem 54 Repeats Finder found an imperfect 78-mer repeat in the ORF of UL44. The two repeat units have a 91% match with each other. Reconstitution of virus particles flom BAC clone and excision of the BAC vector Virus particles were reconstituted flom the BAC by transfecting CRFK cells with BAC DNA. The reconstituted virus, with the BAC vector in its genome, produced fluorescent plaques in CRFK monolayers (data not shown). To reduce possible effects the BAC vector might have on viral growth characteristics and virulence, CRFK cells were co-transfected with BAC DNA and pcDNA-Cre. pcDNA-Cre expresses Cre protein, which specifically recognizes the loxP sites flanking the BAC cassette and EGFP gene, excises them, and re-ligates the DNA leaving a single loxP site (Figure 3.1). After plaque purification, no fluorescent plaques were found. The excision of BAC vector was also verified by PCR and sequence analysis (data not shown). The BAC- excised FHV-l BAC clone, FHVlABAC, was used for subsequent in vitro and in vivo characterizations. In vitro characterization of F HVlABAC To define the in vitro grth characteristics of the BAC-derived virus, plaque morphologies and multiple-step growth curves for the C-27 strain and FHVlABAC were compared. Plaques produced by the C-27 strain and F HVlABAC virus are morphologically undistinguishable flom each other. The average size of 100 plaques was determined for each virus strain. The mean plaque diameter of the FHVIABAC virus was 101% of the C-27 parent strain and not significantly different (p = 0.740). 55 Multi-step growth curves were constructed for both the FHVlABAC virus and the C-27 parent strain (Figure 3.5). At 48 and 72 hours p.i., the FHVlABAC virus grew to titers 0.5 - 1 logs lower than those of the parent strain. Analysis of the growth curves by ANOVA indicated that the differences at 48 hours (p = 0.003) and 72 hours p.i. (p = 0.013) were statistically significant. Preliminary in vivo characterization In order to investigate possible attenuation resulting flom BAC cloning, a preliminary challenge experiment was carried out using four specific-pathogen-flee (SPF) cats. Two cats were inoculated with the F HVlABAC virus, one cat with the C-27 strain (positive control), and another cat was inoculated with cell culture medium (negative control). The results are summarized in Table 3.6 and Figure 3.6. The negative control cat did not show any clinical signs throughout the study. The cats inoculated with the C-27 strain and FHVlABAC virus all developed similar clinical signs. The cumulative clinical scores of the FHVlABAC-infected cats and the cat inoculated with the parent strain were comparable. Virus shedding was detected by virus isolation in the cats inoculated with the C-27 strain and the FHV lABAC virus on days 3, 6, and 9 p.i. Virus neutralization tests demonstrated that the positive control cat and the FHVIABAC virus-infected cats all showed seroconversion on day 14 p.i. The neutralizing antibody titers of the FHVlABAC virus-infected cats and the cat inoculated with the parent strain were very similar. 56 DISCUSSION We report here the first BAC clone containing the entire C-27 strain FHV-1 genome that exhibits very high virulence in preliminary in vivo inoculation experiments. The restriction pattern of this BAC clone was nearly identical to that of the parent C-27 strain with all exceptions being related to the insertion of the BAC vector or circularization of the genome (Figure 3.2). The size of the genome, derived flom both the sequence assembly and the restriction pattern analysis, is consistent with the literature (Rota et al., 1986). An unexpected 2.7-kb cat DNA was inserted along with the BAC at the genomic termini. A possible explanation is the fact that herpesviruses can acquire parts of host genomes, through an unknown mechanism, during virus replication (Niikura et al., 2006). The BAC clone-derived FHV-l virus was characterized in vitro by observing its plaque morphology and comparative growth curve analysis. Plaques produced by the BAC clone-derived virus were identical in size to those produced by the parent C-27 strain. The comparative growth curve analysis also showed that the BAC clone-derived virus can grow to > 107 TCleo/ml, which was 0.5-1.0 logs lower than its parent strain (Figure 3.5). Although the BAC vector was excised flom the genome through Cre-loxP recombination, parts of the recombination arms, as well as the cellular sequence still remained. The presence of these sequences, totaling 3.5 kb, might have subtle influences on virus growth. 57 In a limited in vivo study, the BAC clone-derived virus showed very similar pathogenicity in the infected cats. The delayed onset of clinical signs and the longer persistence could be related to the slightly slower replication of the BAC-derived virus, as shown in the in vitro characterization. However, since the number of cats used was very small, it could also be just a difference in susceptibility between individuals. Although the number was very small, the fact that both of the cats developed symptoms similar to the wild-type strain suggested that the BAC-derived virus is infectious, and its virulence appears to be similar to that of the parent strain. Moreover, in previous studies, it was shown that the symptoms induced by FHV-l in experimentally infected cats are quite consistent (Kruger et al., 1996; Sussman et al., 1995). The fact that this BAC- derived virus behaves very similarly to its parent strain both in vitro and in vivo makes it an excellent starting platform for defining the function of individual viral genes, especially those mediating the virulence of FHV-l. We also present the first complete and annotated sequence of the FHV-1 genome, and a complete overview of the molecular composition of FHV-1. Prior to this study, sequencing efforts were focused on smaller flagrnents of the genome or individual genes flom various strains. A firll list of FHV-l genes completely sequenced prior to this report is included in Table 3.2. Envelope glycoprotein genes drew the most interest; 8 out of 11 had been completely sequenced. The sequence presented in this report was obtained by sequencing both the FHV-l DNA genome and the FHV-l BAC clone using automated high-throughput pyrosequencing. This system is rapid, provides high read depth, does not require any subcloning, and is more economical than traditional Sanger sequencing. One 58 disadvantage of this method is its short read length. Compared to 500-1000 base read lengths of Sanger sequencing, the read lengths of pyrosequencing averaged 100 bases. Recently, however, the read length has been improved to ~500 bases. The short read length poses difficulties for assembly of repetitive sequences, such as the “a” sequences at the genomic termini. The “a” sequences, which consists complex tandem repeats, could not be assembled, and had to be resolved by primer walking. Another inherent problem of pyrosequencing is the difficulty in determining the number of incorporated nucleotides in homopolymeric regions of 5 or more nucleotides (Ronaghi and Elahi, 2002). Therefore, despite the high redundancy, there is a slight possibility that the lengths of such repeats in this genomic sequence are not entirely accurate. The physical structure of the FHV-l genome was first mapped in 1986 when Rota et al. reported a SalI restriction map of the C-27 strain (Rota et al., 1986). In 1991, Grail et al. mapped another strain of F HV-l, B927 (Grail et al., 1991). The complete genomic sequence showed that the gene arrangement in the F HV -1 genome is collinear with that of many varicelloviruses, including BHV-l, BHV-5, EHV-l, EHV-4, and VZV. The shuffling of gene blocks found in the PRV genome did not appear in the FHV-1 genome of this strain. A search for SalI sites in our genomic sequence revealed that the Sall map of C-27 is very similar to that of the B927 strain, with only a few differences. The region in the B927 strain where there is a 16.5-kb SalI flagment contains a 13.5- and a 13.6-kb flagment in the C-27 strain. This could explain why the predicted length of the B927 genome was ~9 kb shorter than our sequence. The second and fourth flagrnents in the C-27 strain seem to have exchanged their locations in the B927 strain, implying a possible re-arrangement of the genes, as seen in the PRV genome (Klupp et al., 2004). 59 Most of the FHV-1 ORFs were identified by homology search except for US2 and US26.5. Due to its low similarity to the counterparts in the other varicelloviruses, USZ was annotated based on its relative position in the genome, which was similar to the other varicelloviruses. UL26.5 is in the same reading flame as UL26, but use alternative start codons. It was also annotated based on its relative position. The ab initio gene finding program did not identify new genes unique to FHV- 1. A few alphaherpesviruses, including HSV-1, HSV-2, BHV-l, and PRV, have evolved genomes with a relatively high G+C content. In these genomes, the third codon position is particularly biased towards G or C, since it is the most flexible concerning the amino acid encoded. In PRV, all functional ORFs could be easily identified by screening for ORFs with a high G+C content on the third nucleotide position of codons (Klupp et al., 2004). The FHV-l genome has not acquired this characteristic. The average G+C percentage of FHV-l genome was 45%, one of the lowest among sequenced varicelloviruses. Therefore, this method was not applicable for FHV-1. The number of tandem repeats found in the FHV-1 genome is far lower than in other varicelloviruses. Due to the short read length of the pyrosequencing, it is possible that multiple copies of the same repetitive unit were assembled into far fewer copies. However, many of the reiterated elements in varicellovirus genomes are shorter than 50 bp, which means in the sequence assembly there should still be at least two copies, if they are present, hence detectable by the Tandem Repeats Finder program. Therefore, it is more likely that the FHV-1 genome does not have as many tandem repeat elements. In other alphaherpesvirus genomes, tandem repeats are flequently found between two polyadenylation signals flom convergent transcripts, or within 1 kb flom the genomic 60 termini. Based on the location of the repeats, it was hypothesized that they play possible roles in insulation against accidental read-through by RNA polymerase into the oppositely transcribed gene, as well as in the process of genome circulization (Klupp et al,2004) In conclusion, the F HV-l BAC clone we report contains the entire FHV-1 genome, the BAC clone-derived virus grows to slightly lower titers in vitro compared to the C-27 parent strain, and its plaque morphology is not significantly altered. Preliminary indications are that its in vivo virulence is also similar to that of the C-27 parent strain. Recent advances in recombineering (recombination-mediated genetic engineering) techniques have made it easier to introduce site-specific mutations into a BAC clone (Tischer et al., 2006). In addition, random libraries of herpesvirus BAC mutants can be generated by the use of transposon-mediated insertion mutagenesis (Brune et al., 1999). These characteristics, along with the availability of the complete genome sequence, will make the BAC clone an excellent starting platform for future mutagenesis-based fimctional studies of viral genes and ultimately for future vaccine developments. MATERIALS AND METHODS Cells and Viruses CRFK cells (ATCC, Manassas, VA) were cultured in Eagle’s Minimum Essential Medium (EMEM) containing 10% fetal bovine serum (FBS) and 10 ug/ml of 61 ciprofloxacin. The FHV-l prototype strain C-27 (ATCC, Manassas, VA) was propagated in the CRFK cells and used as wild-type virus throughout this study. The purified FHV-1 Virions were prepared by pelleting the supernatant through a 20% potassium tartrate cushion at 24,000 x g for 2 hours at 4°C. Plasmids and Vectors The 0.6-kb upstream homologous region, which includes the 3’-end of US3 gene, the intergenic region of US3 and US4, and the 5’-end of US4 (gG) gene, was amplified flom purified FHV-1 genomic DNA by PCR using the Extended High Fidelity PCR System (Roche Applied Science, Indianapolis, IN) with the following primers: RM0874, which contains a MIuI site at the 5’-end; and RM0875, which contains a SwaI site, a BerI site, and a loxP site at the 5’-end (Table 3.1). The PCR product was cloned into a pCRH vector (Invitrogen, Carlsbad, CA), resulting in pCRII-UgG. The 1.0-kb downstream homologous region, which includes the intergenic region of US4 (gG) and US6 (gD), and the 5’-end of US6 (gD) gene, was amplified flom purified FHV-l genomic DNA by PCR using the Extended High Fidelity PCR System with the following primers: RM0876, which contains a HindIII site, a SalI site, and a loxP site at the 5’-end; and RM0877, which contains a MluI site at the 5’-end (Table 3.1). The PCR product was also cloned into a pCRII vector, resulting in pCRII-DgG. pCRH-EGFPmZ was constructed as follows. The vector pEGFP-Nl (Clontech Laboratories, Mountain View, CA) was digested by XhoI and SalI and re-ligated, eliminating X7201, SacI, HindIII, and SaII sites. The 1.6-kb region containing the modified EGFP expression cassette, which includes the CMV immediate early promoter, 62 the EGFP ORF, and SV40 early mRNA polyadenylationsignal, was amplified from the modified pEGFP-Nl by PCR using Extended High Fidelity PCR System with the following primers: RM0872, which contains a BszWI site at the 5’-end; and RM0873, which contains a Sad and SalI site at the 5’-end (Table 3.1). The PCR product was cloned into a pCRII vector, resulting in pCRII-EGFPmZ. pGEM3Zf-DUgG2, which contains two homologous regions and two loxP sites, was constructed by ligation of the following flagrnents: the MIuI-SwaI flagment of pCRII-UgG; the HindIII-Mlul flagment of pCRH-DgG; and the pGEM-3Zf vector (Promega, Madison, WI), digested with HindIII and SmaI. pGEM3Zf-DUgGEGFP2, which contains two homologous regions, two loxP sites, and a EGFP expression cassette, was constructed as follows: pCRII-EGFPm2 was digested with BsiWI and Sacl, and the fragment containing EGFP expression cassette was ligated with pGEM3Zf-DugG2, also digested with BsNVI and SacI. To target the BAC cassette insertion site to US4, the BAC vector pBAC04, which contains the BAC cassette, two (upstream and downstream) homologous recombination arms between two loxP sites and an EGFP expression cassette, was constructed as follows. The BAC vector pBeloBACll (Invitrogen, Carlsbad, CA) was digested with SalI and dephosphorylated; the 6.4-kb flagment was purified and ligated with the SalI flagment of pGEM3Zf-DUgGEGFP2. To construct pcDNA-Cre, a plasmid that expresses Cre recombinase, the entire ORF of Cre was excised by KpnI and SmaI digestions flom pGIKS-Cre (ATCC, Manassas, VA) and ligated with pcDNA3.1 vector (Invitrogen, Carlsbad, CA), digested with KpnI and EcoRV. 63 All the plasmid constructs were verified by both restriction digestion and sequencing. pBAC04 was transformed into the E. coli strain DHlOB (Invitrogen, Carlsbad, CA). All the other constructs were transformed into the E. coli strains TOP10 or DHSa (both flom Invitrogen). The transformants were plated on selective agar that contained 75 ug/ml of ampicillin or 34 ug/ml of chloramphenicol. DNA Extraction Pure viral genomic DNA was prepared as follows: Purified Virions were incubated in lysis buffer (0.1 M Tris-Cl, pH8.0, 1 mM EDTA, 1% SDS) with 50 ug/ml proteinase K at 37°C. DNA was subsequently extracted with phenol-chloroform- isoamylalcohol (25:24: 1), and precipitated with 100% ethanol. Care was used to avoid shearing the DNA. High copy number plasmids were extracted using the Plasmid Mini Kit (Qiagen, Valencia, CA). Small-scale BAC DNA purifications were carried out using the alkaline lysis method (Sambrook and Russell, 2001). Large-scale and high-purity BAC DNA purifications were carried out using the Large Construct Kit (Qiagen, Valencia, CA) following the manufacturer’s instructions. BAC Cloning The strategy used for cloning FHV-1 genome as a BAC exploits spontaneous homologous recombinations in the transfected CRFK cells, and was similar to those used in previous reports (Costes et al., 2006; Niikura et al., 2006). To reduce possible sequence alterations during in vitro passage in cell culture, low-passage C-27 strain virus (P4) was used for BAC cloning. Purified FHV-1 genomic DNA and pBAC04 64 were co-transfected into CRFK cells using Lipofectamine 2000 (Invitrogen, Carlsbad, CA) according to the manufacturer’s instructions. The supernatant of co-transfected cells was collected for plaque assay and plaques were examined for EGFP expression. One virus clone that consistently produced fluorescent plaques was obtained. After two times of plaque purification, CRFK cells were inoculated with this virus clone, and the circular form replication intermediate of the virus genome was extracted from the cells using the method of Hirt (1967) and transformed into E. coli DHlOB cells. Colonies growing on the chloramphenicol plate were examined for the presence of F HV-l genome and BAC vector by restriction pattern analysis and PCR. A clone that had a SalI pattern representative of most bacterial clones and most similar to that of the C-27 strain was used for all subsequent studies. Reconstitution of infectious virus flom BAC The CRFK cells were co-transfected with 1 pg of the BAC DNA and 1 ug of pcDNA-Cre using Lipofectamine 2000 (Invitrogen, Carlsbad, CA) according to the manufacturer’s instructions. Sequencing, sequence assembly and gap closure The majority of the sequence was determined by shotgun sequencing, using the high-throughput pyrosequencing instrument Genome Sequencer 20 (Roche Applied Science, Indianapolis, IN) at the Genomics Core of Michigan State University’s Research Technology Support Facility (RTSF). The reads were assembled using the Newbler assembly program (Roche Applied Science, Indianapolis, IN) at the 65 Bioinforrnatics Core of RTSF. Gaps were closed by primer walking. Gap closure was carried out using BigDye Terminator 3.0 and dGTP BigDye Terminator chemistries and Gene Analyzer 3100 (Applied Biosystems, Foster City, CA). ORF finding and sequence analysis To identify the genes encoded in the FHV-1 genome, all potential ORFs with a minimum length of 60 codons and a methionine as start codon were analyzed for homology to known proteins using BLASTX against non-redundant protein database (Altschul et al., 1997). To find novel genes in the FHV-l genome ab initio, and as a second approach to verify the annotation, the sequence was submitted to GeneMarkS (Besemer et al., 2001). To search for polyadenylation signals, the FHV-l genome sequence was submitted to PolyADQ, a eukaryotic polyadenylation signal search engine (Tabaska and Zhang, 1999). The Neural Network Splice Site Prediction program conditioned for human splice site recognition (Reese et al., 1997) was used to identify possible splice donor and acceptor sites of UL15. The Tandem Repeats Finder program (Benson, 1999) was used to search for tandem repeats. Phylogenetic analyses were carried out using the MEGA4 software (Tamura et al., 2007). Plaque Morphology One hundred fifty TCID50 units of the virus were inoculated on CRFK monolayers. Afier one-hour adsorption, the diluted viruses were removed, and flesh grth medium added. The cells were then incubated at 37°C in an atmosphere of 5% C02. After 40 hours of incubation, 100 plaques produced by each virus were 66 photographed and the diameters determined using the QCapture software (QIrnaging, Surrey, BC, Canada). The measurements were analyzed by AN OVA. Multiple-step Growth Curve The wild type virus and the BAC clone were inoculated on CRFK monolayers in triplicate at an moi. of 0.01. After one-hour adsorption, the diluted viruses were removed and the cells overlaid with flesh medium. The first supernatant samples were collected at this time as 0 hour p.i. The monolayers were then incubated at 37°C in an atmosphere of 5% C02. Supematants were collected at 0, 6, 24, 48, and 72 hours p.i. and stored at -80°C until titration. The mean titers of each time point were analyzed by ANOVA. Cats Four 12-week old, female SPF cats were used (Liberty Research, Waverly, NY). Cats were housed in individual cages in rooms with controlled temperature, humidity, and lighting. They were fed a combination of dry and moist diets. Each group of cats was housed in a separate Biocontainment Level-2 room. All cats were acclimated for 13 days before virus exposure. Two cats were inoculated oronasally with 2 x 105 TCID50 of the FHVlABAC. The other two cats were either inoculated oronasally with 2 x 105 TCID50 of the C-27 strain wild type virus (positive control) or Eagle’s Minimum Essential Medium (negative control). The titers were re-checked prior to inoculation. Clinical signs induced by inoculation of the viruses were scored as described in the USDA Supplemental Assay Method 311 (U .S. Department of Agriculture, Animal and 67 Plant Health Inspection Service, National Animal Veterinary Services Laboratory, 1985; Table 3.1). Oral swabs were collected flom each cat at days 0, 3, 6, 9, 14, and 21 p.i. for virus isolation. Serum samples were collected flom each cat at days 0, 14, and 21 p.i. for virus neutralization testing. The cat study was reviewed and approved by the Institutional Animal Care and Use Committee at Michigan State University. ACKNOWLEDGEMENTS This research was funded by the Companion Animal Fund and the Center for Feline Health and Well-Being at Michigan State University’s College of Veterinary Medicine. We would like to thank Dr. Jeff Landgraf, Kevin Carr, Colleen Curry, and Shari Tjugum-Holland at the Research Technology Supporting Facility at Michigan State University, for nucleotide synthesis, pyrosequencing and Newbler assembly, all of which were invaluable for the completion of this project. We would also like to thank Torn Goodwill and Laurie Molitor for their excellent technical assistance in Sanger sequencing, Dr. Henry Hunt for supply of materials used in the BAC cloning, Janice Forcier for assistance in the in vivo experiment, Crystal Passmore for assistance in the VN tests, Dr. Patricia Schenck for advice on statistical tests, and Dr. Vilma Yuzbasiyan- Gurkan for helpful discussions and critical reading of the manuscript. 68 REFERENCES Alba, M.M., Das, R., Orengo, C.A., Kellam, P., 2001. Genomewide function conservation and phylogeny in the Herpesviridae. Genome Res. 11, 43-54. Altschul, S.F., Madden, T.L., Schaffer, A.A., Zhang, J ., Zhang, 2., Miller, W., Lipman, D.J., 1997. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 25, 3389-3402. Arii, J., Hushur, O., Kato, K., Kawaguchi, Y., Tohya, Y., Akashi, H., 2006. Construction of an infectious clone of canine herpesvirus genome as a bacterial artificial chromosome. Microbes Infect. 8, 1054-1063. Balan, P., Davis-Poynter, N., Bell, S., Atkinson, H., Browne, H., Minson, T., 1994. An analysis of the in vitro and in vivo phenotypes of mutants of herpes simplex virus type 1 lacking glycoproteins gG, gE, gI or the putative gJ. J. Gen. Virol. 75, 1245- 1258. Baranowski, E., Keil, G., Lyaku, J., Rijsewijk, F.A., van Oirschot, J.T., Pastoret, P.P., Thiry, E., 1996. Structural and functional analysis of bovine herpesvirus 1 minor glycoproteins. Vet. Microbiol. 53, 91-101 . Benson, G., 1999. Tandem repeats finder: a program to analyze DNA sequences. Nucleic Acids Res. 27, 573-580. Besemer, J ., Lomsadze, A., Borodovsky, M., 2001. GeneMarkS: a self-training method for prediction of gene starts in microbial genomes. Implications for finding sequence motifs in regulatory regions. Nucleic Acids Res. 29, 2607-2618. Borst, E.M., Hahn, G., Koszinowski, U.H., Messerle, M., 1999. Cloning of the human cytomegalovirus (HCMV) genome as an infectious bacterial artificial chromosome in Escherichia coli: a new approach for construction of HCMV mutants. J. Virol. 73, 8320-8329. Brune, W., Menard, C., Hobom, U., Odenbreit, S., Messerle, M., Koszinowski, U.H., 1999. Rapid identification of essential and nonessential herpesvirus genes by direct transposon mutagenesis. Nat. Biotechnol. 17, 360-364. Czostes, B., Thirion, M., Dewals, B., Mast, J ., Ackermann, M., Markine-Goriaynoff, N., Gillet, L., Vanderplasschen, A., 2006. Felid herpesvirus 1 glycoprotein G is a structural protein that mediates the binding of chemokines on the viral envelope. Microbes Infect. 8, 2657-2667. 69 Crandell, R.A., Maurer, RD, 1958. Isolation of a feline virus associated with intranuclear inclusion bodies. Proc. Soc. Exp. Biol. Med. 97, 487-490. Davison, A.J., Eberle, R., Ehlers, B., Hayward, G.S., McGeoch, D.J., Minson, A.C., Pellett, P.E., Roizman, B., Studdert, M.J., Thiry, E., 2009. The order Herpesvirales. Arch Virol 154, l71-7. Davison, A.J., Scott, J .E., 1986. The complete DNA sequence of varicella-zoster virus. J. Gen. Virol. 67, 1759-1816. Delecluse, H.J., Hilsendegen, T., Pich, D., Zeidler, R., Hammerschmidt, W., 1998. Propagation and recovery of intact, infectious Epstein-Barr virus flom prokaryotic to human cells. Proc. Natl. Acad. Sci. U. S. A. 95, 8245-8250. Delhon, G., Moraes, M.P., Lu, 2., Afonso, C.L., Flores, E.F., Weiblen, R., Kutish, G.F., Rock, D.L., 2003. Genome of bovine herpesvirus 5. J. Virol. 77, 10339-10347. Dolan, A., Jamieson, F.E., Cunningham, C., Barnett, B.C., McGeoch, D.J., 1998. The genome sequence of herpes simplex virus type 2. J. Virol. 72, 2010-2021. F argeaud, D., Jeannin, C.B., Kato, F., Chappuis, G., 1984. Biochemical study of the Feline Herpesvirus 1. Identification of glycoproteins by affinity. Arch. Virol. 80, 69-82. Gaskell, R., Dawson, 8., Radford, A., Thiry, E., 2007. Feline herpesvirus. Vet. Res. 38, 337-354. Gaskell, R.M., Povey, R.C., 1979. Feline viral rhinotracheitis: sites of virus replication and persistence in acutely and persistently infected cats. Res. Vet. Sci. 27, 167- 174. Gaskell, R.M., Povey, R.C., 1982. Transmission of feline viral rhinotracheitis. Vet. Rec. 111, 359-362. Grail, A., Harbour, D.A., Chia, W., 1991. Restriction endonuclease mapping of the genome of feline herpesvirus type 1. Arch. Virol. 116, 209-220. Hansen, K., Napier, I., Koen, M., Bradford, S., Messerle, M., Bell, E., Seshadri, L., Stokes, H.W., Birch, D., Whalley, J .M., 2006. In vitro transposon mutagenesis of an equine herpesvirus 1 genome cloned as a bacterial artificial chromosome. Arch. Virol. 151, 2389-2405. Hirt, B., 1967. Selective extraction of polyoma DNA flom infected mouse cell cultures. J. Mol. Biol. 26, 365-369. 70 101 M N] Pc R: R0 Johnson, L.R., Maggs, D.J., 2005. Feline herpesvirus type-l transcription is associated with increased nasal cytokine gene transcription in cats. Vet. Microbiol. 108, 225- 233. Klupp, B.G., Hengartner, C.J., Mettenleiter, T.C., Enquist, L.W., 2004. Complete, annotated sequence of the pseudorabies virus genome. J. Virol. 78, 424-440. Kruger, J.M., Sussman, M.D., Maes, R.K., 1996. Glycoproteins g1 and gE of feline herpesvirus-1 are virulence genes: safety and efficacy of a gl-gE deletion mutant in the natural host. Virology 220, 299-308. Lee, L.F., Wu, P., Sui, D., Ren, D., Kamil, J., Kung, H.J., Witter, R.L., 2000. The complete unique long sequence and the overall genomic organization of the GA strain of Marek's disease virus. Proc. Natl. Acad. Sci. U. S. A. 97, 6091-6. Maeda, K., Horimoto, T., Mikami, T., 1998. Properties and functions of feline herpesvirus type 1 glycoproteins. J. Vet. Med. Sci. 60, 881-888. Maggs, D.J., 2005. Update on pathogenesis, diagnosis, and treatment of feline herpesvirus type 1. Clin. Tech. Small Anim. Pract. 20, 94-101. Marchini, A., Liu, H., Zhu, H., 2001. Human cytomegalovirus with IE-2 (UL122) deleted fails to express early lytic genes. J. Virol. 75, 1870-1878. McGeoch, D.J., Dahymple, M.A., Davison, A.J., Dolan, A., Frame, M.C., McNab, D., Perry, L.J., Scott, J .E., Taylor, R, 1988. The complete DNA sequence of the long unique region in the genome of herpes simplex virus type 1. J. Gen. Virol. 69, 1531-1574. McGeoch, D.J., Dolan, A., Ralph, A.C., 2000. Toward a comprehensive phylogeny for mammalian and avian herpesviruses. J. Virol. 74, 10401-6. Nasisse, MP, 1990. Feline herpesvirus ocular disease. Vet. Clin. North Am. Small Anim. Pract. 20, 667-680. Niikura, M., Dodgson, J ., Cheng, H., 2006. Direct evidence of host genome acquisition by the alphaherpesvirus Marek's disease virus. Arch. Virol. 151, 537-549. Povey, R.C., 1979. A review of feline viral rhinotracheitis (feline herpesvirus 1 infection). Comp. Immunol. Microbiol. Infect. Dis. 2, 373-387. Reese, M.G., Eeckman, F.H., Kulp, D., Haussler, D., 1997. Improved splice site detection in Genie. J. Comput. Biol. 4, 311-323. Ronaghi, M., Elahi, E., 2002. Discovery of single nucleotide polymorphisms and mutations by pyrosequencing. Comp. Funct. Genomics. 3, 51-56. 71 Rota, P.A., Maes, R.K., Ruyechan, W.T., 1986. Physical characterization of the genome of feline herpesvirus-l. Virology 154, 168-179. Rudolph, J ., O'Callaghan, D.J., Osterrieder, N., 2002. Cloning of the genomes of equine herpesvirus type 1 (EHV-l) strains KyA and racLll as bacterial artificial chromosomes (BAC). J. Vet. Med. B Infect. Dis. Vet. Public Health 49, 31-36. Saeki, Y., Ichikawa, T., Saeki, A., Chiocca, E.A., Tobler, K., Ackermann, M., Breakefield, X.O., Fraefel, C., 1998. Herpes simplex virus type 1 DNA amplified as bacterial artificial chromosome in Escherichia coli: rescue of replication- competent virus progeny and packaging of amplicon vectors. Hum. Gene Ther. 9, 2787-2794. Sambrook, J., Russell, D.W., 2001. Molecular Cloning: A Laboratory Manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York. Schumacher, D., Tischer, B.K., Fuchs, W., Osterrieder, N., 2000. Reconstitution of Marek's disease virus serotype 1 (MDV-1) flom DNA cloned as a bacterial artificial chromosome and characterization of a glycoprotein B-negative MDV-l mutant. J. Virol. 74, 11088-11098. Schwyzer, M., Ackermann, M., 1996. Molecular virology of ruminant herpesviruses. Vet. Microbiol. 53, 17-29. Shizuya, H., Birren, B., Kim, U.J., Mancino, V., Slepak, T., Tachiiri, Y., Simon, M., 1992. Cloning and stable maintenance of 300-kilobase-pair flagments of human DNA in Escherichia coli using an F-factor-based vector. Proc. Natl. Acad. Sci. U. S. A. 89, 8794-8797. Smith, G.A., Enquist, L.W., 1999. Construction and transposon mutagenesis in Escherichia coli of a full-length infectious clone of pseudorabies virus, an alphaherpesvirus. J. Virol. 73, 6405-6414. Stavropoulos, T.A., Strathdee, C.A., 1998. An enhanced packaging system for helper- dependent herpes simplex virus vectors. J. Virol. 72, 7137-7143. Stiles, J., 2003. Feline herpesvirus. Clin. Tech. Small Anim. Pract. 18, 178-185. Strive, T., Hardy, C.M., French, N., Wright, J.D., Nagaraja, N., Reubel, G.H., 2006. Development of canine herpesvirus based antifertility vaccines for foxes using bacterial artificial chromosomes. Vaccine 24, 980-988. Sussman, M.D., Maes, R.K., Kruger, J.M., Spatz, S.J., Venta, P.J., 1995. A feline herpesvirus-l recombinant with a deletion in the genes for glycoproteins g1 and gE is effective as a vaccine for feline rhinotracheitis. Virology 214, 12-20. 72 Tabaska, J .E., Zhang, M.Q., 1999. Detection of polyadenylation signals in human DNA sequences. Gene 231, 77-86. Tamura, K., Dudley, J ., Nei, M., Kumar, S., 2007. MEGA4: Molecular Evolutionary Genetics Analysis (MEGA) software version 4.0. Mol. Biol. Evol. 24, 1596-1599. Telford, E.A., Watson, M.S., McBride, K., Davison, A.J., 1992. The DNA sequence of equine herpesvirus-1. Virology 189, 304-316. Telford, E.A., Watson, M.S., Perry, J., Cullinane, A.A., Davison, A.J., 1998. The DNA sequence of equine herpesvirus-4. J. Gen. Virol. 79, 1197-1203. Tham, K.M., Studdert, M.J., 1987. Clinical and immunological responses of cats to feline herpesvirus type 1 infection. Vet. Rec. 120, 321-326. Tischer, B.K., von Einem, J ., Kaufer, B., Osterrieder, N., 2006. Two-step red-mediated recombination for versatile high-efficiency markerless DNA manipulation in Escherichia coli. Biotechniques 40, 191-197. Tulman, E.R., Afonso, C.L., Lu, Z., Zsak, L., Rock, D.L., Kutish, G.F., 2000. The genome of a very virulent Marek's disease virus. J. Virol. 74, 7980-7988. Yu, D., Smith, G.A., Enquist, L.W., Shenk, T., 2002. Construction of a self-excisable bacterial artificial chromosome containing the human cytomegalovirus genome and mutagenesis of the diploid TRL/IRL13 gene. J. Virol. 76, 2316-2328. 73 TABLES Table 3.1 List of primers used in this study Primer Sequence 45' - 3) BAC Construction RM0872 QQTAQ GTAGTTATTAATAGTAATCAATTACGG RM0873 QTQQAC GAGCTCACGCCTTAAGATACATTGATGAG RM0874 ACGCGTGAATI'CCCAGGCGACCCGAC RM0875 ATTTAAAT CGTACGA TAACTTCG TA TAA TG TA TGC TA TA C GAA G TTA TTATCTCTGGTGATCTTGAAGAG RM0876 AAGCTT GTCGACA TAA C ITC G TA TAG CA TA CA TTA TA C GAA G TTA TTAAACTGATTAAATTTAATTAAAG RM0877 MGAGATAGTTGTACCATAAAATCTG Sequence verification of the transfer plasmid BA C 04 RM0898 CAA'ITTCACACAG GAAACAG RM0899 ACGGATTTTTAAGCACACCA RMOQOO TATTCGAAC CATACAACTATC RM0903 AAGGCGATTAAGTTGGGTA RM0904 GTTGGGGTCTTTGCTCAGGG RM0905 AAGAAGTCGTG CTGCTTCATG RM0906 GGAGTTGTTACGACATTITGG RM0907 TTACTATGGGAACATACGTC PCR Assays RM0901 ATACGATTGTATGTCAAAATAC RM091O AACGGAGTAACCTCGGTGTG RM091 1 AATGCTTAATGAATTACAACAG RM0944 GCGTTACTATGGGAACATAC RM0945 ATCAGCCATACCACATTTGTAG RM0955 ACCAATTCTCATGTTTGACAGC RM0956 ATCGGCACGTAAGAGGTTCC RM0957 TGGACAGAACAACCTAATGAAC RM0958 ATGACAGATCCATGTGAAGTG RM0959 TAATCACGAG CTCCATCAAGGC RM0988 ATTACGTITATTGGACCCGAG RM0989 TGAGACTTTAATTAAATI'TAATCAG RM1 025 ACTTCTTTTCTACTTGCCCTGG RM1026 TAACCCCAGCTTAAACAGAAC Restriction sites are underlined; loxP sites are in italic. 74 Table 3.1, continued Primer Sequence (5‘ — 3’) Gap closure by primer walking RM1023 RM1024 RM1027 Rm1028 RM1037 RM1038 RM1039 RM1040 RM1041 GTS565 GT8569 RM1240 RM1242 RM1243 RM1244 RM1245 RM1246 AATATCACCCCGGTAAACGACAC ACTTCCTGGAATAATGCACATC GTT'ITI'GTTAATACTTCTGTGGAT AGTATATATAAACCCTACCTACG ACACCTCTCGCTTCTCCTCTGAC TCAGAGACGAAACTGCTCGCATAG AATAGTTAT'IT'I'I'ATCTCACATGC CCTCCGCGGGTTCTAAAACTTG GAGGACGCGGAGAGTGGATGGTTC CATATTCAAATCTGGCCTAAT AGATTTGAATATTTACTGATGTGA TCCTTATTAGGCCAGATTGAATATG TCTACATATGAATAGTAGATTTGAATA'ITTACT TATTTACTGATGTGATAACTCGATCC GGGGGAAAAAAATTGAGTTGGAGCTC TGAGTTGGAGCTCAGTCTCCATATACG CATATACGGAGGTGGACAGTGTAG Restriction sites are underlined; loxP sites are in italic. 75 Table 3.2 Comparison of the complete FHV-l genomic sequence with sequences available in GenBank X98449 . Encoded genes Strain or Sequence . . . Sequence Am“ N°' isolate length (nt) “’“h mp'ete w'th '"gggnp'ete identity (%) ”“95 D14563 62620 3.038 UL45 UL46. UL44 99.9 GM036023 N/A 10,803 UL45. UL44. UL46. UL39 99.6 UL43, UL42. UL41, UL40 Ax018721 N/A 2,956 UL45. UL44 UL46. UL43 99.9 AR018958, N/A 1,007 UL45 UL46. UL44 99.9 158415 086616 07301 2.337 UL44 UL45 99.8 AX244515. N/A 1,602 UL44 — 99.9 80396132 M006454 8927 5,587 UL40. UL39 UL41, UL38 99.4 N224971 8927 4.848 UL30 UL31, UL29 99.5 866371 C-27 3,340 UL27 UL28 99.5 849775 C7301 3.240 UL27 UL28 99.8 AX24451 1. N/A 2,829 UL27 — 99.7 BD396130 GM981 1 19 N/A 2,847 UL27 — 99.8 E12463 N/A 1,619 UL23 UL24. UL22 99.9 M26660 UCD 1.619 UL23 UL24. UL22 100.0 864566 C7301 3.746 ‘ UL22 UL23. UL21 100.0 A8112595 00-035 3.007 UL17 UL16 100.0 AF022391 C-27 4.918 UL2. UL1. ICPO UL3 99.4 21-mer repeat: >11 vs 11 copies DQ452613 Taichung 1 882 UL2 — 99.7 D30766 C7301 6.175 ICPO — 100.0 20-mer repeat: 20.1 vs 6.1 copies AY740677 8927 3.127 U84 U83, U86 99.9 16-mer repeat 3 vs 12 copies 872415 C-27 6.208 U84. U86, U87. U83. U88.5 99.6 16-mer mpeat: 13 vs U88 12 copies D3076? C7301 2.317 U86 U84, U87 100.0 16-mer repeat: 7 vs 12 copies 042113 62620 8.276 U86, U87. U88. U84 99.9 16-mer repeat: 9 vs 12 U885. U89. copies U810. U81 AX244521. N/A 1,122 US6 — 100.0 80396135 GM981 120 N/A 1,125 U86 — 99.7 A37005. N/A 6,154 U87, U88, — 99.9 AR281844 U885. U89. U810. U81 X98448 8927 1.155 U87 — 99.7 8927 1,599 U88 — 100.0 76 Table 3.3 Predicted FHV-l ORFs 77 Protein Length aa identity (%) Predicted function or (aa) EHV-1 EHV-4 8HV-1 8HV-5 PRV vzv property UL56 203 22.9 23.2 N/A N/A 10.5 19.7 membrane protein UL56 V1 144 27.0 23.9 N/A N/A NlA 15.3 membrane protein V1 myristyiated tegument CIRC 278 32.0 31.7 31.5 31.5 N/A 26.1 protein CIRC UL55 209 47.5 46.0 N/A N/A NlA 33.9 nuclear protein UL55 multifunctional expression UL54 438 40.7 41.8 42.1 42.5 42.1 30.8 regulator UL53 344 44.9 46.1 34.0 33.2 34.3 42.6 envelope glycoprotein K UL52 1,062 46.1 47.3 40.5 43.0 43.9 39.9 gjgfiifte‘pmase pnmase UL51 242 46.4 46.6 38.7 37.8 41.6 38.6 tegument protein UL51 deoxyuridine UL50 325 38.8 38.8 32.8 34.1 36.7 27.7 tripho s ph at a s e UL49.5 95 40.0 41.1 31.5 35.2 35.2 32.2 envelope glycoprotein N UL49 345 26.2 26.8 26.7 23.6 27.3 19.9 tegument protein VP22 transactivating tegument UL48 447 54.6 54.4 44.0 43.7 46.6 42.6 protein VP16 UL47 858 39.3 38.4 29.5 30.8 31.6 19.5 tegument protein VP13I14 UL46 720 38.8 38.8 31.0 30.7 28.5 27.9 tegument protein VP11/12 UL45 193 28.3 28.3 N/A N/A N/A N/A membrane protein UL45 UL44 534 32.1 30.9 29.0 28.2 20.0 28.3 envelope glycoprotein C UL43 415 40.3 39.4 27.3 26.7 22.5 24.1 envelope protein UL43 DNA polymerase UL42 395 38.7 39.8 30.5 28.7 27.5 24.1 p r oce s sivity subunit tegument host shutoff UL41 453 59.6 59.4 52.4 52.6 56.6 40.8 protein ribonucleotide reductase UL40 331 67.0 66.2 65.6 68.3 70.3 64.4 subunit 2 UL39 786 64.6 64.7 57.9 56.5 61.2 52.9 gfigflflgfiwde '°°”°t°°° UL38 463 50.6 50.9 45.8 44.2 48.2 40.6 capsid triplex subunit 1 UL37 1,027 43.9 43.8 34.0 33.5 37.2 32.2 tegument protein UL37 UL36 3,033 42.5 42.6 33.9 34.2 34.9 32.3 large tegument protein UL35 108 46.7 43.9 50.0 49.1 54.0 36.8 small capsid protein 01.34 271 49.8 48.5 44.4 42.0 44.2 44.1 33;?" °9'°°° m°"'°'°”° UL33 123 46.3 45.0 49.1 49.5 45.0 39.0 Bfgpad‘aging °’°t°i" UL32 582 54.3 53.8 48.8 49.6 53.4 52.6 Bfgpawaging °'°‘°i" nuclear egress lamina UL31 337 63.4 62.5 54.1 52.8 61.0 55.3 protein UL30 1.205 64.9 65.1 59.0 59.4 62.7 56.7 Elifirpitolymerase °°‘°'V"° Table 3.3, continued protein Length 33 identity (%) Predicted function or (88) EHV-1 EHV-4 8HV-1 env-5 PRV vzv property single-stranded DNA- UL29 1,200 66.2 66.2 57.7 57.5 59.9 54.4 binding protein DNA packaging terminase UL28 779 57.8 59.0 51.3 51.6 53.8 50.5 subunit2 UL27 948 58.2 59.1 59.2 59.1 59.4 53.2 envelope glycoprotein B V32 130 47.2 43.0 N/A N/A N/A 36.9 protein V32 UL26.5 294 37.3 36.0 21.4 23.2 16.2 26.0 capsid scaffold protein capsid maturation UL26 598 49.1 47.3 39.6 40.0 42.0 34.4 protease DNA acka in te ument UL25 623 56.1 55.1 52.0 52.0 49.7 49.2 ”0,6,5" UL295 9 9 UL24 260 48.6 50.2 38.1 37.6 55.6 36.2 nuclear protein UL24 UL23 343 47.4 48.0 37.9 36.9 47.3 35.3 thymidine kinase UL22 821 33.8 33.1 30.0 29.5 27.4 29.0 envelope glycoprotein H UL21 527 47.5 46.9 35.2 36.3 40.4 39.0 tegument protein UL21 UL20 229 45.9 43.2 31.6 30.7 26.7 33.2 envelope protein UL20 UL19 1,377 72.8 72.2 65.9 66.3 68.1 60.0 major capsid protein UL18 312 64.1 64.1 59.8 61.1 58.8 55.3 capsid triplex subunit 2 DNA packaging tegument UL17 691 51.2 50.8 41.8 41.4 44.4 37.0 protein UL17 UL16 363 52.1 51.0 44.0 45.3 39.1 37.3 tegument protein UL16 DNA packaging terminase UL15 734 71.3 71.0 63.6 63.5 61.3 61.3 subunit1 UL14 322 34.5 35.4 32.5 32.7 42.7 31.6 tegument protein UL14 tegument serine/threonine UL13 606 49.2 49.0 36.8 36.0 31.9 35.5 protein kinase UL12 544 59.9 60.4 41.0 41.2 47.1 37.6 deoxyribonuclease myristylated tegument UL11 76 36.1 35.6 33.8 34.7 25.4 36.1 protein UL10 422 40.7 40.5 36.7 37.0 39.6 33.2 envelope glyCOprotein M DNA replication origin- UL9 860 58.5 58.4 48.8 51.5 49.8 46.5 binding helicase UL8 782 46.0 45.2 36.8 36.5 36.2 36.6 heiicase-primase subunit UL7 296 46.1 44.9 36.9 38.5 40.4 37.3 tegument protein UL7 UL6 717 57.9 58.7 50.9 53.5 60.1 48.2 capsid portal protein UL5 663 70.7 70.0 64.9 65.0 60.5 66.1 gjgfiifte'i’mase "9"“39 UL4 236 43.6 42.9 36.4 37.8 32.4 26.7 nuclear protein UL4 UL3.5 181 21.9 21.4 18.3 16.3 18.0 22.5 protein V57 UL3 201 67.7 69.5 54.5 51.0 44.4 43.0 nuclear protein UL3 UL2 293 49.8 49.8 50.0 48.1 46.2 43.4 uracil-DNA glycosylase UL1 147 28.6 28.8 25.4 27.3 24.6 27.1 envelope glycoprotein L ICPO 498 23.4 22.0 21.4 20.1 18.9 14.2 ubiquitin E3 ligase ICPO 78 Table 3.3, continued protein Length 33 identity (%) Predicted function or (88) EHV—1 EHV-4 BHV-1 8HV-5 PRV vzv property ICP4 1,396 42.7 43.2 39.6 40.7 40.2 35.4 :‘Cagjmp‘ma' regu'am' US1 334 54.8 53.4 44.3 43.5 33.5 31.2 regulatory protein ICP22 U810 213 45.0 44.5 N/A NIA N/A 27.1 virion protein U810 U82 102 12.7 11.8 10.8 11.8 13.7 N/A virion protein U82 serine/threonine protein US3 351 54.4 51.6 41.9 41.0 46.1 38.2 kinase U83 U84 434 36.3 36.3 28.7 28.4 27.0 NIA envelope glycoprotein G U86 374 24.9 25.1 30.1 30.2 26.3 NIA envelope glycoprotein D US? 384 35.4 36.7 23.6 23.4 22.9 23.3 envelope glycoprotein I U88 532 45.9 45.4 30.8 29.8 22.9 25.2 envelope glycoprotein E US8A 100 13.0 14.6 NIA NIA NIA NIA membrane protein U88A U89 152 30.1 35.2 25.9 26.3 35.8 26.7 membrane protein U89 U810 213 virion protein U810 U81 334 regulatory protein ICP22 transcriptional regulator ICP4 1.398 ICP4 79 Table 3.4 Differences in gene composition among sequenced Varicellovirus genomes Predicted Function or Protein FHV-1 EHV-1 EHV-4 8HV-1 8HV-5 PRV VZV P r o perty UL56 + + + — — + + membrane protein UL56 V1 + + + — — — + membrane protein V1 m 'st ated te ument C'RC + + + + + ‘ + prbi'eiri'CIRc g UL55 + + + - — — + nuclear protein UL55 VZV ORF 13 — - - - — — + thymidylate synthase UL45 + + + — — — - membrane protein UL45 V32 + + + — — — + protein V32 V67 — + + + + — + virion protein V67 U810 + + + — — — + vin’on protein U810 US4 + + + + + + — envelope glycoprotein G U85 — + + — — — - envelope glycoprotein J US6 + + + + + + — envelope glycoprotein D US8A + + + - — — membrane protein U88A 8O Table 3.5 Tandem repeats within the FHV-1 genome Repeat unit Total length Location size 1132) Copy no. Sequence (bp) Note 121 — 497 17 22.2 tggagtctaggtgtggg 377 UL / Gap3 106.506 - 106.736 21 11 ggcctaataaggaaggggagg 231 Re, / Gap1 (135.497 - 135.727) (TRs I Gap3) 106.734 — 106,949 30 7.2 tctggcggtttgtgggttggcatattcaaa 216 Rs / Gap1 (135,284 — 135.499) (TRs / Gap3) 106,963 - 107,066 30 3.5 104 IRS I Gap1 (135.167 - 135,270) (T Rs I Gap3) 1 10.463 — 110,501 18 2.2 tggagcgacgctcactga 39 RS / ICP4 (131.732 — 131.770) (TRs / ICP4) 1 11,766 — 1 11,877 20 6.1 accttcgctcctcccctcgt 122 IR; / Between ICP4 and U81 (130.346 - 130.467) (TFLc. / Between ICP4 and U81) 112,613 - 112.814 53 3.8 aggttggaagccatgttgttccggttgcac 202 IR; / Between ICP4 and U81 atctaatctacatgaaagtggga (129.419 - 129.620) (T Re I Between ICP4 and U81) 112.851 - 113.459 26 23.4 gggggatcgagggggggcagagggga 609 Re. I Gap2 (128.774 - 129.382) (T Fig / Gap2) 120.219 — 120,410 16 12 W 192 Us / Between U84 and U86 81 Table 3.6 Cumulative clinical score, virus shedding pattern, and virus neutralizing antibody titers of SPF cats inoculated with the C-27 parent strain, the FHVlABAC strain, or mock-inoculated with cell culture medium Days AJJ5 AJCZ AJC3 AJDB p.i. (C-27) (FHV1A8AC) LFHV1ABAQ (EMEM) Vl VN VI VN VI VN VI VN 0 — <4 - <4 - <4 — <4 3 + ND + ND + ND — ND 6 + ND + ND + ND — ND 9 + ND + ND + ND — ND 14 — 64 - 32 - 32 — <4 21 - 64 — 32 - 64 — <4 Cumulative clinical score 22 22 26 0 VI: virus isolation; VN: virus neutralization; ND: not determined. 82 FIGURES AND FIGURE LEGENDS Figure 3.1 Diagrams showing (A) structure of FHV-1 genome, (B) location of the BAC cassette and cellular sequence insertion, (C) the composition of the BAC cassette, and (D) the FHV-l BAC clone after excision of the BAC cassette. The diagram is not to scale. 83 mEE canes—=89. 5:6 \ / \ ElxlU a mucczuum L23=ou 3.» 6x3 EB Eu c323E82 cgunsnEouo. _stna _Stma 28 ES Ema may. 95 m5! nEOM can 0:! ea. es. 00:0360m \Lm_:__ou 0mm: .5 FigureZ3J 84 Figure 3.2 Three bacterial clones of FHV—1 BAC (lanes 1-3) and C-27 parent strain genomic DNA (lane 4) were treated overnight with restriction endonuclease SalI at 37°C. The DNA flagrnents were resolved and visualized using a 0.7% agarose gel stained with ethidium bromide. Based on sequence analysis, the linear FHV-1 C-27 strain genome contains the following SalI flagrnents. A: 16,123 bp; B: 14,011 bp; C: 13,648 bp; D: 13,466 bp; E: 11,730 bp; F: 10,121 bp; G: 9,559 bp; H: 9,119 bp; 1: 8,564 bp; J: 6,977 bp; K: 6,701 bp; L: 4,951; M: 3,996 bp; N: 2,587; 0: 1,724 bp; P: 769 bp; Q: 677 bp. The 414-bp R flagment and two 330-bp S flagrnents were not visible in this picture. G and M flagments that contained genomic termini (marked by diamonds in lane 4) would have been ligated when the genome circularized, but were instead resolved as three flagrnents because of the introduction of additional SaII sites by the BAC insertion. The first two of these flagrnents, 9,445 and 9,171 bp in size (arrows), form a triple band with the H fragment. The other flagment was 6,384-bp in size (arrows) The size of the K fiagrnent was found to be variable among bacterial clones (arrowheads). Position of 1 kb DNA ladders are indicated at the left side. 85 12K 11K 10K— 9K . 8K 7K 6K6 ~ 5K 4K“- 3K 2K 1.6K Figure 3.2 86 ' . , 994...: I it" m3: 433 m3: 8.15 ~m> A A613...... mg: amt—D 0.3: ~33 5.5 Figure 3.3 Predicted FHV-l gene arrangement. 87 A (am: i T T 9 I it 2': «do. 55 o5: own 3: Km: om: oooeor ooommv oooomp oooowp oooomp ooovmw ooowwr 883 BE. V , 5: I A E ' T T '. mm: 5: .3.0. odO. «.5 88: 88:. 89.: 88:- 88: .ooomop--- 889 89.9 ooomow mm. V ' I A .I 35 S: . m5 . . :5 , 2.5 V ti! .f T t 2 m4: 3: m4: 3: 3: o5: N5: I T T 25 t 'I 3.5 2.5 2.5 9.5 2.5 8.5 3.5 8.5 Figure 3.3, continued 88 FHV-1 100 EHV-1 100 —— EHV-4 PRV 100 .__... BHV-5 VZV l———-l 0.05 B UL53 (9K) 95 FHV-1 VZV »— EHV-1 100 ._ EHV-4 PRV 100 BHV-5 i———l 0.1 UL49.5 (gN) FHV-1 56 — EHV-1 _ 100 ._ EHV-4 PRV 100 ——- BHV-5 VZV 0 1 Figure 3.4 Phylogenetic analyses based on concatenated amino acid sequence alignment of conserved genes (A) and individual glycoprotein amino acid sequence alignments (B). Trees were generated using the neighbor-joining method. Bootstrap values (1,000 replicates) are given for each branch. Scale bars represent number of amino acid substitutions per site. 89 UL44 (gC) FHV-1 43 r —— EHV-1 100 —— EHV-4 100 — BHV-1 BHV-5 3“ PRV vzv 1—1 0.1 UL27 (gB) FHV-1 94 EHV-1 F 100 EHV-4 PRV 97 .— 8HV-1 100 -— BHV-5 vzv 0.05 UL22 (gH) FHV-1 91 EHV-1 99 100 EHV-4 vzv PRV BHV-1 100 — BHV-5 I——| 0.1 Figure 3.4, continued. 90 UL10 (9M) 50 FHV-1 [— EHV-1 100 EHV-4 PRV 72 BHV-1 100 . BHV-5 VZV 0 1 UL1(gL) 57 FHV-1 BHV-1 32, 100 BHV-5 EHV-1 100 EHV-4 VZV PRV l-——l 0.1 US4 (gG) 85 FHV-1 EHV-1 100 EHV-4 PRV Bl-N-1 100 BHV-5 l-——-l 0.1 Figure 3.4, continued. 91 U86 (gD) 63 FHV'1 PRV BHV-1 100 .— BHV-5 EHV-1 100 .— EHV4 F—-l 0.1 US7 (gl) FHV 1 91 EHV-1 100 —— EHV4 7" PRV 97 BHV-1 100 BHV-5 VZV ' 0.2 T US3 (95) -1 100 FHV EHV-1 100 -— EHV-4 VZV PRV 95 ‘ BHV-1 100 BHV-5 l——-l 0.1 Figure 3.4, continued. 92 Time (Hr) ; T . e . r i C CD 66 N <0 ID V to N ‘- O (“rm/090131 501) 1311.1. Figure 3.5 Multi-step grth curve of the parent wild type strain C-27 (grey) and the BAC-derived virus (black). The wild type virus and the FHVlABAC were inoculated on CRFK monolayers at an moi. of 0.01, and supernatants collected and titrated at 0, 6, 24, 48, and 72 hours post inoculation (p.i.). Error bars at each data point represent :l:1 standard deviation. 93 14 13 12 10 ”1 __- __- 1 I- $093309 Days Post Inoculation __- __- ___-- ---_ 6 5),“! £010) 9,0400 19,0410) 23% 266 265 233 Figure 3.6 Durations of clinical signs and virus shedding in cats inoculated with the C- 27 parent strain (Cat AJJ5) or the FHVlABAC virus (Cats AJC2 and AJC3). The Gantt chart shows the time flame during which a specific clinical sign or virus shedding persisted. The cat number is shown next to its respective bar. A bar is shown only if a clinical sign was observed or virus shedding occurred. 94 CHAPTER 4 GENERATION AND IN VITRO CHARACTERIZATION OF FELINE HERPESVIRUS 1 (FHV-1) MUTAN TS LACKING GLYCOPROTEIN S C (gC) AND E (gE) Tai, S.H., Cheng, H.H., Niikura, M., Kim, T., Engstrom, M.D., Maes, R.K. Generation and in vitro characterization of feline herpesvirus 1 (FHV-1) mutants lacking glycoproteins C (gC) and E (gE). In preparation for submission. 95 Generation and in vitro characterization of feline herpesvirus 1 (FHV-1) mutants lacking glycoproteins C (gC) and E (gE) S. H. Sheldon Taia, Hans H. Chen 5|"), Masahiro Niikurab’l, Taejoong Kimb’z, Michael g D. Engstromc, Roger K. Maesa’c’d’* a Graduate Program in Comparative Medicine and Integrative Biology, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA b Avian Disease and Oncology Laboratory, Agricultural Research Service, United States Department of Agriculture, 3606 East Mount Hope Road, East Lansing, MI 48823, USA c Department of Microbiology and Molecular Genetics, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA d Diagnostic Center for Population and Animal Health, College of Veterinary Medicine, Michigan State University, 4125 Beaumont Road, Lansing, MI 48910, USA * Corresponding author. Diagnostic Center for P0pulation and Animal Health, College of Veterinary Medicine, Michigan State University, 4125 Beaumont Road, Lansing, MI 48910, USA. Fax: +1 517 432 6527. E-mail address: Maes@dcpah.msu.edu (R.K. Mass). 1 Present address: Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, V5A 186, Canada. 96 2 Present address: Poultry Diagnostic and Research Center, University of Georgia, Athens, GA 30602, USA. ABSTRACT Feline herpesvirus 1 (FHV-1) mutants lacking the entire open reading frames encoding glycoprotein C (gC) or E (gE) were constructed, based on a FHV-1 BAC clone. The gC' FHV-1 mutant virus produced primary plaques that enlarged slowly and had relatively few secondary plaques compared to the wild type virus. Analysis on multi-step growth kinetics showed that the mutant virus grew to titers that were reduced by approximately 6,000 — 9,000 folds, compared with the wild-type virus. Analysis of the mutant virus on single-step growth kinetics showed that the titer of intracellular virus was 3.5 - 10 folds higher than the extracellular virus titer. Taken together, the results indicate that the mutant virus is more cell-associated compared to the wild-type virus. Thus, FHV-1 gC, like is the case in other varicelloviruses, plays a significant role in initial attachment/penetration, replication, and egress of FHV-1. In contrast, the gE- FHV-1 mutant had single-step growth kinetics that were indistinguishable flom those of the FHVlABAC, and grew to a titer that was approximately 10-fold lower than that of the FHVlABAC in the multi-step growth kinetics. These results suggested that virus egress, and most likely cell-to-cell spread as shown in other alphaherpesviruses, were affected by gE deletion, while virus entry and replication were not. 97 Keywords: Feline herpesvirus 1, FHV-1, glycoprotein C, glycoprotein E, mutant, infectious BAC clone, virulence INTRODUCTION Feline herpesvirus 1 (FHV-l) is an important viral pathogen of Felidae and has a world-wide distribution. Infection with FHV-l not only accounts for approximately 50% of all diagnosed viral upper respiratory infections, but is also a significant cause of ocular diseases in cats (Nasisse, 1990). Pathobiology and clinical manifestations associated with FHV-1 have recently been reviewed (Gaskell, 2007; Maggs, 2005; Stiles, 2000). Primary infection, acquired via the oronasal route, results in fever, sneezing, ocular and nasal discharge, conjunctivitis, and keratitis. Like herpesvirus infections in other species, the acute phase of the disease is followed by lifelong latency. During the latent stage, the FHV-1 genome persists in neural tissues but infectious virus is not produced. Different biological stresses, or administration of corticosteroids, can induce the necessary biochemical stimuli in latently infected cells that lead to renewed production of infectious virus, which then travel to the periphery and is a potential source of viral transmission. Despite the clinical significance and high prevalence of FHV-1 infection, currently available vaccines can not totally protect cats flom field virus infection and, as a consequence, flom field virus latency (Gaskell, 1993; Harbour et al., 1991; Nasisse et al., 1997; Tham and Studdert, 1987). Furthermore, most vaccines are safe only when administered subcutaneously (Kruger et. al., 1996). 98 FHV-1 has a double-stranded DNA genome and a genomic organization similar to that of other varicelloviruses of the Alphaherpesvirus subfamily (Grail et al., 1991; Rota et al., 1986), which includes the prototype varicella zoster virus (VZV), bovine herpesvirus 1 (BHV-l), bovine herpesvirus 5 (BHV-S), equine herpesvirus 1 (EHV—l), equine herpesvirus 4 (EHV-4), and pseudorabies virus (PRV), among others. The complete 135,797-bp sequence of the FHV-1 genome has been determined and 74 encoding proteins have been identified, including 11 glycoproteins (Tai et al., submitted). FHV-1 has been shown to contain 23 virion-associated proteins (Fargeaud etal., 1984). The functions of seven glycoproteins, gB, gC, gD, gE, gG, gH and g1, had been reviewed by Maeda et al. (1997). Glycoprotein C (gC) homologues have been extensively studied in several alphaherpesviruses. gC homologues are non-essential for herpesvirus replication in vitro, but they mediate several important biological functions. gC is involved in the initial step of viral attachment by interacting with heparan sulfate on the cell surface, as demonstrated for herpes simplex virus-1 (HSV-1) , PRV, BHV-l, and EHV-l (Herold et al., 1991; Mettenleiter et al., 1990; Okazaki et al., 1991; Osterrieder, 1999). gC deficient mutants attach to cells with reduced efficiency (Osterrieder, 1999). ng of HSV-1 and -2 can bind the complement component C3b (Geraghty et al., 1998; Lubinski et al., 1999). Binding of this complement factor may protect herpesvirus- infected cells flom complement-mediated lysis (Fries et al., 1986). Viruses lacking complement-binding domains are less virulent than wild-type virus (Frink et al., 1983; Herold et al., 1991; Lubinski et al., 1999). The gC of FHV-1 has been shown to be the dominant heparin-binding glycoprotein that mediates the initial stage of viral adsorption, 99 as observed in other herpesviruses (Maeda et al., 1997). However, it remains to be determined whether FHV-1 gC protect virus-infected cells flom complement-mediated lysis. A deficiency of gC is commonly seen in strains of herpesviruses attenuated by serial passage in cell cultures. The PRV Bartha vaccine strain carries mutations within the gC gene that include an alteration in the amino terminal signal sequence, resulting in inefficient intracellular translocation and incorporation of this protein into the viral envelope (Robbins et al., 1989). The VZV vaccine strain Oka is defective in gC expression compared to wild type VZV strains (Kinchington et al., 1990). In addition, a genetically engineered TK-gC- vaccine strain has been licensed for use in control of pseudorabies (Kit, 1990). A sensitive and highly specific ELISA kit was developed for use in conjunction with this vaccine to distinguish non-infected and/or vaccinated pigs flom those infected with PRV field strains (Kit et al., 1990). Glycoprotein E (gE) is a virulence factor of FHV-1. In alphaherpesviruses, glycoprotein E (gE) and glycoprotein I (g1) form a heterodimer that fimctions in cell-to- cell spread of the virus and spread of infection throughout the host nervous system, which is ultimately the cause of neurovirulence. Generally, alphaherpesvirus mutants that lack these glycoproteins are replication-competent in cell culture but produce smaller plaques, due to reduced capacity for cell-to-cell spread. gE/gI deletion mutants of herpes simplex virus type 1 (HSV-1) and other alphaherpesviruses such as pseudorabies virus (PRV) and bovine herpesvirus 1 (BHV-l), show impaired ability to spread flom cell to cell (Balan et al., 1994; Dingwell et al., 1994; Dingwell and Johnson, 1998; Otsuka and Xuan, 1996; Zuckermann et al., 1988). The gE/gI heterodimer 100 appears to play an even greater role in the spread of varicella-zoster virus (V ZV) (Frink et al., 1983; Mallory et al., 1997; Mallory et al., 1998) and in Marek’s disease virus (MDV), in which the gE/gl heterodimer has been found to actually be essential for grth in cultured cells (Schumacher et al., 2001). Previous studies have shown that gE/gI complex of FHV-l is not essential for virus growth, although virulence is significantly reduced and the virus produces smaller plaques when the gE/gI genes are deleted (Sussman et al., 1995). The FHV-l gE/gI mutant, when administered via the oronasal route, can protect cats flom clinical signs and significantly reduce viral loads in subsequent challenge with a high dose of field virus. However, this mutant at higher dose levels still retained partial virulence as it can produce mild clinical signs in a dose- dependent manner (Kruger et al., 1996; Sussman et al., 1997). Bacterial artificial chromosome (BAC) cloning and recombination-mediated genetic engineering (recombineering) are two state-of-the-art techniques to facilitate the mutagenesis of herpesviruses. BACs are single copy F-factor-based plasmid vectors which can stably hold 300 kb or more of foreign DNA (Shizuya et al., 1992). The BACs’ larger capacity and greater stability over the other vectors have made BAC the vector of choice for the cloning of entire herpesvirus genomes. Many alphaherpesvirus genomes have been cloned as BACs since the first infectious BAC of a herpesvirus was reported in 1997 (Messerle et al., 1997). These include HSV (Saeki et al., 1998; Stavropoulos and Strathdee, 1998), PRV (Smith and Enquist, 1999), EHV-l (Rudolph et al., 2002), and MDV (Niikura et al., 2006; Schumacher et al., 2000). Recently, we have constructed a BAC clone that contains the entire FHV-1 genome, and virus derived flom this BAC clone has been shown to possess in vitro and in vivo 101 characteristics very similar to the prototype C-27 strain (Tai et al., submitted). This FHV-1 BAC clone is therefore a suitable starting platform for characterizing virulence factors and development of vaccine candidates. Recombineering is a powerfirl method for fast and efficient manipulation of the BAC. It allows DNA cloned in E. coli to be modified via lambda (it) Red-mediated homologous recombination, obviating the need for restriction enzymes and DNA ligases. Site-specific and “scarless” deletions, insertions, and point mutations can be introduced efliciently using a recombineering procedure developed by Tischer et al. (2006), known as two-step Red-mediated recombination (See Figure 4.1 for an overview.) In this report, we present the construction and in vitro characterization of the first FHV-1 gC-deletion mutant, as well as a gE-deletion mutant. Both were constructed based on an infectious FHV-1 BAC clone by the two-step Red-mediated recombination approach. MATERIALS AND METHODS Cells and Viruses Crandell-Reese feline kidney (CRFK) cells (ATCC CCL-94, Manassas, VA) were cultured in Eagle’s Minimum Essential Medium (EMEM) containing 10% fetal bovine serum (FBS) and 10 ug/ml of ciprofloxacin. The FHV-l prototype strain C-27 (ATCC 102 VR-636, Manassas, VA) was propagated in the CRFK cells and used as the wild-type virus throughout this study. Construction of Mutant FHV-1 BACs “Scarless” deletions of the entire open reading flame (ORF) of gC or gE were introduced into the FHV-l BAC clone (Tai et al., submitted) using the two-step Red- mediated recombination method described by Tischer et a1. (2006) and illustrated in Figure 4.1. First, a mutating DNA fragment that contained 50-bp of upstream recombination arm, 50-bp of downstream recombination arm, kanamycin resistance gene expression cassette (kan), and a restriction site of homing endonuclease I-SceI was generated by PCR using the Extended High Fidelity PCR System (Roche Applied Science, Indianapolis, IN). The 25 pl reaction consisted of 1x Expand High Fidelity buffer, 1.5 mM of MgC12, 200uM of each dNTP, 600 nM of each dUL44F and dUL44R primer (Integrated DNA Technologies, Coralville, IA) (Table 1), 2 ng of pEPkan-S as template, and 1.4 U of Expand High Fidelity enzyme mix. PCR amplification was carried out in a Thermal Cycler 2720 (Applied Biosystems, Foster City, CA). The program started with an initial denaturation step at 94°C for 2 minutes, followed by 40 cycles of 94°C for 1 minute, 55°C for 1 minute, and 68°C for 1 minute. Following the last elongation step at 68°C for 10 minutes, the template plasmid was degraded by adding 40 U of Dpnl (New England BioLabs, Ipswich, MA) to the PCR reaction and incubating at 37°C for 1 hour. The 1,145-bp amplicon was resolved in a 1% agarose gel and purified using the QIAquick Gel Extraction Kit (Qiagen, Valencia, CA). The gel- purified DNA was then transformed into E. coli SW105 cells (Warming et al., 2005) 103 containing the FHV-1 BAC clone, which was induced for the expression of recombination genes exo, bet and gam at 42°C for 15 min. Candidate colonies grown on LB agar plates containing 50 pg/ml of chloramphenicol and 75 pg/ml of kanamycin were checked by PCR using primers RM1206-RM1207 and RM1208-RM1209 (Table 1), and restriction pattern analysis. Positive clones were further engineered to remove the kan insertion. The second recombination was carried out by inducing the expression of recombination genes at 42°C for 15 min and the expression of I-SceI by 1% of arabinose. The successfully engineered gC- and gE-deleted FHV-1 BAC clones were identified based on the restriction pattern, results of colony PCR, and sequence analysis, and designated as FHV lBACAUL44 and FHVlBACAUS8, respectively. Reconstitution of Infectious Viruses flom BACs To reconstitute mutant FHV-l Virions flom the mutant BAC clones and excise the BAC cassette flom the viral genome, CRFK cells were co—transfected with either FHVlBACAUL44 or FHVlBACAUSS DNA, and pcDNA-Cre plasmid using Lipofectamine 2000 (Invitrogen, Carlsbad, CA) (Tai et al., submitted). Briefly, 300,000 CRFK cells were seeded in each well of a 24-well plate and incubated overnight at 37°C in an atmosphere of 5% C02 until the cultures reached ~95% confluence. To form DNA-Lipofectamine 2000 complexes, 1 pg of either FHVlBACAUL44 DNA or FHVlBACAUS8 DNA, and 1 pg of pcDNA-Cre plasmid were diluted in 50 pl of EMEM without antibiotics or FBS, and combined with 3 pl of Lipofectamine 2000, also diluted in 50 pl of EMEM without antibiotics or F BS. The cell culture medium was replaced with 100 pl of each DNA-Lipofectamine 2000 complex and 500 pl of flesh 104 EMEM containing 10 pg/ml of ciprofloxacine and 10% PBS. After 5 — 7 days of incubation, the supernatants were harvested and progeny virus that produced non- fluorescent plaques was plaque-purified before being propagated in CRFK cells. Reconstituted gC-deleted virus that had the BAC cassette removed thru Cre-loxP recombination was designated as FHVlAgC. Reconstituted gE-deleted virus that had the BAC cassette removed was designated as FHVlAgE. The removal of BAC cassette was verified by PCR using primhers RM1101 and RM1102 (Table 4.1) and direct sequencing of the purified PCR product. Deletions of gC and gE was verified again by PCR and direct sequencing of the purified PCR product as described above. Plaque Morphology and Plaque Size Analysis Fifty TCID50 of the viruses to be tested were inoculated on confluent CRFK monolayers in a 6-well plate. After a one-hour adsorption at room temperature, the inoculum was removed, and flesh growth medium was added. The cells were then incubated at 37°C in an atmosphere of 5% C02 and observed daily for 5 days. Plaques produced by each virus were photographed using a QImaging camera (QImaging, Surrey, BC, Canada). To perform plaque size analysis, 150 TCID50 of the viruses to be tested were inoculated on confluent CRFK monolayers grown in a 6-well plate. After a one-hour adsorption at room temperature, the inoculum was removed, and flesh growth medium containing 0.8% carboxymethylcellulose was added. The cells were then incubated at 37°C in an atmosphere of 5% C02. After 5 days of incubation, the infected monolayers were fixed and plaques were stained using fluorescein—conjugated anti-FHV-l 105 antibodies. One hundred plaques produced by each virus were photographed and the diameters were determined using a QImaging camera and the QCapture Pro software (QImaging, Surrey, BC, Canada). ANOVA was used to assess whether differences in plaque size were statistically significant. Growth Kinetics Single-step growth curves were constructed as follows: monolayers of CRFK cells were infected in triplicate with the C-27 strain or the deletion mutants at a multiplicity of infection (MOI) of 3. After an adsorption period of 1 hour, cells were washed with 0.1 M phosphate buffered saline, pH 7.2 (PBS), overlaid with growth medium, and incubated at 37°C in an atmosphere of 5% C02. The cell culture supernatant and infected cells were harvested separately at successive intervals between 4 and 48 hours post inoculation (p.i.) and the amount of infectious virus was titrated on CRFK cells, as described previously (Sussman et al, 1995). Multi-step grth curves were constructed by infection of CRFK monolayers in triplicate with the C-27 strain or the deletion mutants at an MOI of 0.01. After an adsorption period of 1 hour, cells were washed with PBS, overlaid with growth medium, and incubated at 37°C in an atmosphere of 5% C02. Culture supernatants and infected cells were harvested separately at successive intervals between 6 and 120 hours p.i. The amount of infectious virus was titrated on CRFK cells as described previously. 106 RESULTS Construction of Mutant FHV-1 BACs Using two-step Red-mediated recombination techniques (Tischer et al., 2006), FHV-1 mutants lacking the entire ORF of gC or gE were generated, based on the F HV- ] BAC clone previously constructed in our laboratory (Tai et al., submitted). The mutagenesis of the BAC clone was confirmed by colony PCR (data not shown), restriction pattern analysis (Figure 4.2), and sequence analysis (data not shown). Reconstitution of Infectious Virus flom BACs The removal of the BAC vector in the reconstituted FHVlAgC and FHVlAgC viruses was verified by the loss of fluorescence resulting flom the EGFP gene in the BAC vector, PCR and direct sequencing of purified PCR products. The deletions of gC and gE were also verified by PCR (Figure 4.3) and direct sequencing of purified PCR products (data not shown). Plaque Morphology and Plaque Size Analysis Figure 4.4 shows the development of plaques produced by the FHVlAgE and the parent C-27 strain on CRFK monolayers without carboxymethycellulose overlay. On day l p.i., plaques produced by both FHVlAgC and C-27 parent strain were very small. After day 2 p.i., FHVlAgC plaques expanded gradually, while C-27 strain plaques spread rapidly. Overall, plaques produced by F HVlAgC developed slower and remained focal, while the FHV-1 C-27 strain produced extensive CPE on day 4 p.i. 107 Relatively few secondary plaques were produced by FHVlAgC compared to the C-27 strain. Preliminary plaque size analysis did not show a significant difference between the FHVlAgC and the C-27 strains. Growth Kinetics Single-step (Figures 4.5 and 4.7) and multiple-step (Figures 4.6 and 4.8) growth curves were constructed for FHV-l C-27 strain, FHVlABAC, FHVlAgC, and FHVlAgE. FHV lABAC grows to a titer that is approximately 2 — 12 fold lower than the C-27 strain. When inoculated at an MOI of 3 on CRFK monolayers, both the wild type and the gC-deletion mutant started to replicate extensively in CRFK cells after 8 hours post inoculation (Figure 4.5). Compared to the wild type, the gC-deletion mutant replicated much less efficiently and released fewer Virions into the medium. In case of the wild type, the amount of extracellular virus surpassed the amount of intracellular virus between 20 and 24 hours p.i. In contrast, the amount of extracellular virus of the gC- deletion mutant never surpassed the intracellular virus. For both viruses, CPE was nearly complete at 24 hours p.i., all the cells were showing CPE. The amount of intracellular viruses remained constant, and the amount of extracellular viruses continued to increase. In contrast, the gE-deletion mutant has a grth curve very similar to FHVlABAC (Figure 4.7). When inoculated at an MOI of 0.01 on CRFK monolayers, the wild type virus started to replicate extensively in CRFK cells between 6 and 16 hours p.i. (Figure 4.6). 108 The amount of the intracellular virus reached a plateau at 48 hours p.i. The amount of extracellular virus surpassed intracellular virus between 48 and 72 hours p.i., and reached a plateau at 72 hours p.i. After 96 hours p.i., cell lysis was extensive and the virus titer started to decrease. The amounts of extracellular and intracellular virus of the gC-deletion mutant started to increase between 16 and 24 hours p.i., and were very similar between 24 and 72 hours p.i. The gC-deletion mutant also reached a plateau at 72 hours p.i., but the titer was 6,000 — 9,000 fold lower than the wild type titer. The gE- deletion mutant grew to a titer that was approximately 10-fold lower than the FHVlABAC (Figure 4.8). DISCUSSION Previous work in our laboratory involved cloning of the entire FHV-1 genome as an infectious BAC. Virus generated flom this BAC clone was shown to behave very similarly to the parent C-27 strain, both in vitro and in vivo (Tai et al., submitted). Therefore, it was considered to be a suitable starting platform for mutagenesis and virulence studies. Based on this BAC clone, we constructed FHV-1 mutants in which the entire ORF of gC or gE was deleted. The FHV-1 gC-deletion mutant had in vitro characteristics that suggested attenuation. The results of this study suggested that gC could play a role at three different stages of viral infection: attachment/penetration, replication, and/or egress. 109 FHV-1’s attachment/penetration abilities were impaired by gC deletion. In plaque assays without carboxymethylcellulose overlay, the gC-deletion mutant produces relatively few secondary plaques, while the wild type produces many secondary plaques and spreads to the entire monolayer very quickly. Adsorption conditions can influence virus titer. When the inocula were not removed flom the monolayers after an hour of adsorption, essentially allowing longer time for adsorption, the titer of combined intracellular and extracellular virus stock of the F HV -1 gC-deletion mutant could reach 105 TCleo/ml, about 10 times higher than as shown in Figure 4.6. A stock of 106 TCIDso/ml could be grown by co-cultivation of uninfected and FHV lAgC-infected CRFK cells (data not shown). It is well-documented that herpesvirus gC orthologues are the major protein that confers the initial attachment of the virion to the host cell, by electrostatically binding to heparin- and chondroitin-like glycosaminoglycans on the cell surface (Herold et al., 1991; Mettenleiter et al., 1990; Okazaki et al., 1991; Osterrieder, 1999). This binding step can be blocked with soluble heparin (Okazaki et al., 1991). In addition to attachment, PRV gC was shown to play a role in penetration, independently flom its attachment firnction (Mettenleiter, 1989; Rue and Ryan, 2002). A gC-deficient mutant of EHV-l, a very close relative of FHV-1, attaches to cells with reduced efficiency, and penetration assays showed that this mutant’s penetration ability was impaired in primary equine cell culture, and severely impaired in a rabbit cell line (Osterrieder, 1999). The FHV-1 gC-deletion mutant grew to a lower titer and showed a lower growth rate, as shown in the single-step (Figure 4.5) and multi-step (Figure 4.6) growth curves. This observation is not consistent with all studies in other herpesviruses. An EHV-l gC- 110 negative mutant has been shown to have a 5- to 10-fold reduction in titer when grown in rabbit and equine cell lines, and a 48- to 210-fold reduction when grown in primary equine cells (Osterrieder, 1999). gC-negative mutants of HSV-1 and PRV grow to titers that are only reduced by approximately lO-fold in cultured cells (Osterrieder, 1999). In contrast, deletion of gC in BHV-l did not change its growth kinetics significantly in a bovine kidney cell line (Liman etal., 2000), and gC-negative VZV Oka strain exhibited accelerated and more efficient growth in cultured melanoma cells compared to the wild- type Oka strain (Cohen and Seidel, 1994). The reason for FHVlAgC’s lower titer and slower grth may be due to reduced ability of the mutant virus to attach, thus resulting in lower virus load compared to the wild type. It is possible that a higher titer can be reached by inoculating the cells with more virions, as done in the studies of EHV -1 and BHV -1, which used 5 M01 to construct single-step grth curves. However, in order to carry out an inoculation at 5 M01, pelleting of FHVlAgC virions would have been necessary. In addition, different cell type could affect the virus’s growth, as shown in the EHV-l study. This issue can be addressed by using different types of cells, such as feline corneal epithelial cells (Sandmeyer et al., 2005) and Felis catus whole fetus cells (Jacobse-Geels and Horzinek, 1983; Pedersen et al., 1981). It is also possible that FHV-1 gC plays a role in virus replication. In BHV-l, gC has been shown to be important for maintaining the efficacy of viral replication in the natural host (Liang et al., 1992). Yet another reason for the reduced growth could be the reduced ability of the mutant virus to egress. The gC-deletion mutant is more cell-associated than the wild type, suggesting a possible impairment in viral egress. As demonstrated in single-step lll growth curve (Figure 4.5), which characterizes virus replication in individual cells, most of the gC-deleted mutant virus remained in the cell. Specifically, the titer of intracellular virus was 3.5 — 10 times higher than the extracellular virus. This is in contrast to the wild type, where the titer of extracellular virus can be ~29-fold higher than the intracellular virus. This finding is less obvious in the multi-step grth curves. Herpesvirus gC is not considered to have a major role in viral egress or release. Nonetheless, it was noted that the reduction in titer of the gC- mutant of EHV-l in primary equine cells was partially due to impairment in viral egress (Osterrieder, 1999). The limited ability of the FHV-1 gC- mutant to spread in cultured epithelial CRFK cells, as demonstrated in Figure 4.4, suggest that when administered via the natural oronasal route in vivo, it could spread slower on the mucosal epithelial cells at the primary infection site, allowing more time for the immune system to respond, and hence induce a stronger immune response. In addition, it is known that herpesvirus gC plays an important role in immune evasion. gC inhibits the activation of the complement cascade by binding to complement component C3b, and by blocking the binding of properdin and C5 to C3b (Frink et al., 1983; Lubinski et al., 1999, Roizman et al., 2007). It has been shown in HSV that gC-negative mutants are more easily inactivated by complement than the wild-type (Hidaka et al., 1991). Therefore, by deleting F HV-l gC, it is likely that the virus will induce a stronger immune response and fewer clinical signs. Although in vivo experiments are necessary to substantiate this hypothesis, it is reasonable to assume that the F HV-l gC- mutant will show reduced virulence in the natural host. 112 The fact that single-step grth property of the gE-deletion mutant was practically indistinguishable flom that of the FHVlABAC (Figure 4.6) and the multi- step growth curve showed reduced titer suggests that virus egress but not entry and replication was affected by gE deletion. This observation is consistent with previous studies that showed gE plays an important role in cell-to-cell spread (Balan etal., 1994; Dingwell et al., 1994; Dingwell and Johnson, 1998; Otsuka and Xuan, 1996; Zuckermann et al., 1988). In summary, the gC- and gE- mutants have potential as vaccine candidates that are safer and more immunogenic than existing vaccines. An additional advantage of these mutants over current commercially available vaccines is that they can be serologically differentiated flom field virus. A distinct advantage of the recombineering-based approach to create deletion is that additional deletions could be made in either one of these mutants, should it be evident flom future in vivo experiments that they still have residual virulence. ACKNOWLEDGEMENTS Funding for this research was provided by Michigan State University College of Veterinary Medicine’s Feline Health Endowment and Center for Feline Health and Well-Being. The authors thank Biological Resources Branch at National Cancer Institute for providing E. coli SW105 cells, and Dr. Nikolaus Osterrieder for generously 113 providing plasmids which were essential for two-step Red-mediated recombination. The authors also thank Laurie Molitor for excellent technical assistance. 114 REFERENCES Balan, P., Davis-Poynter, N., Bell, S. Atkinson, H., Browne, H., Minson, T., 1994. An analysis of the in vitro and in vivo phenotypes of mutants of herpes simplex virus type 1 lacking glycoproteins gG, gE, g1 or the putative gJ. J Gen Virol 75, 1245- 58. Cohen, J.I., Nguyen, H., 1997. Varicella-zoster virus glycoprotein I is essential for growth of virus in Vero cells. J Virol 71, 6913-20. Cohen, J .I., Seidel, KB, 1994. Absence of varicella-zoster virus (VZV) glycoprotein V does not alter growth of VZV in vitro or sensitivity to heparin. J Gen Virol 75, 3087-93. Dingwell, K.S., Brunetti, C.R., Hendricks, R.L., Tang, Q., Tang, M., Rainbow, A.J., Johnson, DC, 1994. Herpes simplex virus glycoproteins E and I facilitate cell-to- cell spread in vivo and across junctions of cultured cells. J Virol 68, 834-45. Dingwell, KS, and Johnson, DC, 1998. The herpes simplex virus gE-gI complex facilitates cell-to-cell spread and binds to components of cell junctions. J Virol 72, 8933-42. Fargeaud, D., Jeannin, C.B., Kato, F., Chappuis, G., 1984. Biochemical study of the Feline Herpesvirus 1. Identification of glycoproteins by affinity. Arch Virol 80, 69-82. Fries, L.F., Friedman, H.M., Cohen, G.H., Eisenberg, R.J., Hammer, C.H., Frank, M.M., 1986. Glycoprotein C of herpes simplex virus 1 is an inhibitor of the complement cascade. J Irnmunol 137, 1636-41. Frink, R.J., Eisenberg, R., Cohen, G., Wagner, E.K., 1983. Detailed analysis of the portion of the herpes simplex virus type 1 genome encoding glycoprotein C. J Virol 45, 634-47. Gaskell, R.M., 1993. Upper respiratory disease in the cat (including chlamydia): control and prevention. Feline Pract 21, 29-34. Grail, A., Harbour, D.A., Chia, W., 1991. Restriction endonuclease mapping of the genome of feline herpesvirus type 1. Arch Virol 116, 209-20. Harbour, D.A., Howard, P.E., Gaskell, R.M., 1991. Isolation of feline calicivirus and feline herpesvirus flom domestic cats 1980 to 1989. Vet Rec 128, 77-80. 115 Herold, B.C., WuDunn, D., Soltys, N., Spear, P.G., 1991. Glycoprotein C of herpes simplex virus type 1 plays a principal role in the adsorption of virus to cells and in infectivity. J Virol 65, 1090-8. Hidaka, Y., Sakai, Y., Toh, Y., Mori, R., 1991. Glycoprotein C of herpes simplex virus type 1 is essential for the virus to evade antibody-independent complement- mediated virus inactivation and lysis of virus-infected cells. J Gen Virol 72, 915- 21. Jacobse-Geels, H.E., Horzinek, M.C., 1983. Expression of feline infectious peritonitis coronavirus antigens on the surface of feline macrophage-like cells. J Gen Virol 64 (Pt 9), 1859-66. Kinchington, P.R., Ling, P., Pensiero, M., Moss, B., Ruyechan, W.T., Hay, J., 1990. The glycoprotein products of varicella-zoster virus gene 14 and their defective accumulation in a vaccine strain (Oka). J Virol 64, 4540-8. Kit, S., 1990. Genetically engineered vaccines for control of Aujeszky's disease (pseudorabies). Vaccine 8, 420-4. Kit, S., Awaya, Y., Otsuka, H., Kit, M., 1990. Blocking ELISA to distinguish pseudorabies virus-infected pigs flom those vaccinated with a glycoprotein gIII deletion mutant. J Vet Diagn Invest 2, 14-23. Kruger, J M., Sussman, M.D., and Maes R.K., 1996. Glycoproteins g1 and gE of feline herpesvirus-1 are virulence genes: safety and efficacy of a gI-gE deletion mutant in the natural host. Virology 220, 299-308. Liang, X., Babiuk, L.A., Zamb, T.J., 1992. An in vivo study of a glycoprotein gIII- negative bovine herpesvirus 1 (BHV-l) mutant expressing beta-galactosidase: evaluation of the role of gIII in virus infectivity and its use as a vector for mucosal immunization. Virology 189, 629-39. Liman, A., Engels, M., Meyer, G., Ackermann, M., 2000. Glycoprotein C of bovine herpesvirus 5 (BHV-S) confers a distinct heparin-binding phenotype to BHV-l. Arch Virol 145, 2047-59.Lubinski, J ., Wang, L., Mastellos, D., Sahu, A., Lambris, J.D., Friedman, H.M., 1999. In vivo role of complement-interacting domains of herpes simplex virus type 1 glycoprotein gC. J Exp Med 190, 1637-46. Lubinski, J., Wang, L., Mastellos, D., Sahu, A., Lambris, J.D., Friedman, H.M., 1999. In vivo role of complement-interacting domains of herpes simplex virus type 1 glycoprotein gC. J Exp Med 190, 1637-46. Maeda, K., Horimoto, T., Mikami, T., 1998. Properties and firnctions of feline herpesvirus type 1 glycoproteins. J Vet Med Sci 60, 881-8. 116 Maeda, K., Yokoyama, N., Fujita, K., Maejima, M., Mikami, T., 1997. Heparin-binding activity of feline herpesvirus type 1 glycoproteins. Virus Res 52, 169-76. Mallory, S., Sommer, M., Arvin, A.M., 1998. Analysis of the glycoproteins I and E of varicella-zoster virus (V ZV) using deletional mutations of VZV cosmids. J Infect Dis 178 Suppl 1, S22-6. Mallory, S., Sommer, M., Arvin, A.M., 1997. Mutational analysis of the role of glycoprotein I in varicella-zoster virus replication and its effects on glycoprotein E conformation and trafficking. J Virol 71, 8279-88. Messerle, M., Crnkovic, I., Hammerschmidt, W., Ziegler, H., Koszinowski, U.H., 1997. Cloning and mutagenesis of a herpesvirus genome as an infectious bacterial artificial chromosome. Proc Natl Acad Sci U S A 94, 14759-63. Mettenleiter, T.C., 1989. Glycoprotein gIII deletion mutants of pseudorabies virus are impaired in virus entry. Virology 171, 623-5. Mettenleiter, T.C., Zsak, L., Zuckermann, F ., Sugg, N., Kern, H., Ben-Porat, T., 1990. Interaction of glycoprotein gIII with a cellular heparinlike substance mediates adsorption of pseudorabies virus. J Virol 64, 278-86. Mettenleiter, T.C., 1989. Glycoprotein gIII deletion mutants of pseudorabies virus are impaired in virus entry. Virology 171, 623-5. Okazaki, K., Matsuzaki, T., Sugahara, Y., Okada, J ., Hasebe, M., Iwamura, Y., Ohnishi, M., Kanno, T., Shimizu, M., Honda, E., et al., 1991. BHV-l adsorption is mediated by the interaction of glycoprotein gIII with heparinlike moiety on the cell surface. Virology 181, 666-70. Osterrieder, N., 1999. Construction and characterization of an equine herpesvirus 1 glycoprotein C negative mutant. Virus Res 59, 165-77. Otsuka, H., Xuan, X., 1996. Construction of bovine herpesvirus-1 (BHV-l) recombinants which express pseudorabies virus (PRV) glycoproteins gB, gC, gD, and gE. Arch Virol 141, 57-71. Pedersen, N.C., Boyle, J .F., Floyd, K., 1981. Infection studies in kittens, using feline infectious peritonitis virus propagated in cell culture. Am J Vet Res 42, 363-7. Robbins, A.K., Ryan, J .P., Whealy, M.E., Enquist, L.W., 1989. The gene encoding the gIII envelope protein of pseudorabies virus vaccine strain Bartha contains a mutation affecting protein localization. J Virol 63, 250-8. 117 Roizman, B., Knipe, D.M., Whitley, R.J., 2007. Herpes simplex viruses, in: Knipe, D.M., Howley, P.M. (Eds), Fields Virology. Lippincott Williams & Wilkins, Philadelphia, pp. 2502-601. Rota, P.A., Macs, R.K., Ruyechan, W.T., 1986. Physical characterization of the genome of feline herpesvirus-1. Virology 154, 168-79. Rudolph, J ., O'Callaghan, D.J., Osterrieder, N., 2002. Cloning of the genomes of equine herpesvirus type 1 (EHV-l) strains KyA and racLll as bacterial artificial chromosomes (BAC). J Vet Med B Infect Dis Vet Public Health 49, 31-6. Rue, C.A., Ryan, P., 2002. Characterization of pseudorabies virus glycoprotein C attachment to heparan sulfate proteoglycans. J Gen Virol 83, 301-9. Sandmeyer, L.S., Keller, C.B., Bienzle, D., 2005. Culture of feline corneal epithelial cells and infection with feline herpesvirus-l as an investigative tool. Am J Vet Res 66, 205-9. Saeki, Y., Ichikawa, T., Saeki, A., Chiocca, E.A., Tobler, K., Ackermann, M., Breakefield, X.O., F raefel, C., 1998. Herpes simplex virus type 1 DNA amplified as bacterial artificial chromosome in Escherichia coli: rescue of replication- competent virus progeny and packaging of amplicon vectors. Hum Gene Ther 9, 2787-94. Schumacher, D., Tischer, B.K., Reddy, S.M., Osterrieder, N., 2001. Glycoproteins E and I of Marek's disease virus serotype 1 are essential for virus grth in cultured cells. J Virol 75, 11307-18. Schumacher, D., Tischer, B.K., Reddy, S.M., Osterrieder, N., 2001. Glycoproteins E and I of Marek's disease virus serotype 1 are essential for virus growth in cultured cells. J Virol 75, 11307-18. Shizuya, H., Birren, B., Kim, U.J., Mancino, V., Slepak, T., Tachiiri, Y., Simon, M., 1992. Cloning and stable maintenance of 300-kilobase-pair flagrnents of human DNA in Escherichia coli using an F -factor-based vector. Proc Natl Acad Sci U S A 89, 8794-7. Smith, G.A., Enquist, L.W., 1999. Construction and transposon mutagenesis in Escherichia coli of a full-length infectious clone of pseudorabies virus, an alphaherpesvirus. J Virol 73, 6405-14. Stavropoulos, T.A., Strathdee, C.A., 1998. An enhanced packaging system for helper- dependent herpes simplex virus vectors. J Virol 72, 7137-43. 118 Sussman, M.D., Macs, R.K., Kruger, J .M., 1997. Vaccination of cats for feline rhinotracheitis results in a quantitative reduction of virulent feline herpesvirus-1 latency load after challenge. Virology 228, 379-82. Sussman, M.D., Maes, R.K., Kruger, J.M., Spatz, S.J., Venta, P.J., 1995. A feline herpesvirus-1 recombinant with a deletion in the genes for glycoproteins g1 and gE is effective as a vaccine for feline rhinotracheitis. Virology 214, 12-20. Tai, S.H., Niikura, M., Cheng, H.H., Kruger, J.M., Wise, A.G., Maes, R.K. Complete sequence of the feline herpesvirus type 1 (FHV-1) genome derived flom a firlly virulent BAC clone. Submitted. Tham, K.M., Studdert, M.J., 1987. Clinical and immunological responses of cats to feline herpesvirus type 1 infection. Vet Rec 120, 321-6. Warming, S., Costantino, N., Court, D.L., Jenkins, N.A., Copeland, N.G., 2005. Simple and highly efficient BAC recombineering using galK selection. Nucleic Acids Res 33, e36. Weigler, B.J., Guy, J .S., Nasisse, M.P., Hancock, S.I., Sherry, B., 1997. Effect of a live attenuated intranasal vaccine on latency and shedding of feline herpesvirus 1 in domestic cats. Arch Virol 142, 2389-400. Zuckermann, F.A., Mettenleiter, T..C., Schreurs, C., Sugg, N., Ben-Porat, T., 1988. Complex between glycoproteins gI and gp63 of pseudorabies virus: its effect on virus replication. J Virol 62, 4622-6. 119 TABLES Table 4.1 Primers used in this study Primer Sequence (5’ — 3’) Used for dUL44F taagtagtcccgcgagacgatttacatcccgggatcaccaacaa Generation of gC- tctgchGGATGACGACGATAAGTAGGG mutating flagrnent dUL44R ataaccgctgaaacacgggtgataagfltttatacgaactataag ggcgcagattgttggtgatcccgggatgtaaatcgtctcgcggga ctacttaCAACCAATTAACCAATTCTGATTAG dUS3F agacgtagggcaggtttctgttatttaggaagtgtatacgtttggct Generation of gE- aatAGGATGACGACGATAAGTAGGG mutating flagment dUS3R ctctgggtgaacctttagagtggttataactttacgaagatggttgc gggattagccaaacggmcacuccmaamcagawcctgccct acggctCAACCAATTAACCAATTCTGATTAG RM1188 AGTCGAAATCACAGGCAGTG Differentiation of gC- RM1189 TGGAGTCGGT'I'TATATCCATAC mutant and wild type RM94 GGTCATGTGTAATGTTGACG Differentiation of gE- RMl 193 ATACAATATACGCGTTTGACG mutant and wild type RM1206 GCGGTTGTGTCTAATAACTG Confirmation of kan RM1207 GCTTCCCATACAATCGATAGAT insertion in gC (5’-end) RM1208 GATTTTGATGACGAGCGTAAT Confirmation of kan RM1209 CGGTTTATATCCATACGAATGA insertion in gC (3’-end) RM1210 CGCATAGTGGGTGGTGACCTAA Confirmation of kan RM1211 ATCATTGGCAACGCTACCTTT insertion in gE (5’-end) RM1212 TTGCCATTCTCACCGGATTCA Confirmation of kan RM1213 CCCGGATCGACTAGTGTGAAC insertion in gE (3’-end) RM1101 TGCCCCTCCCCCGTTCATGCTGTG Confirmation of the RM1102 TGGAGCCTGTTTCCGATTCTGTGT excision of BAC cassette Sequences in upper-case, nucleotides annealing to template; lower-case, hanging nucleotides; italic, upstream homologous recombination arm; underlined, downstream homologous recombination arm. 120 FIGURES AND FIGURE LEGENDS Figure 4.1 An overview of the two-step Red-recombination approach (Tischer et al., 2006) used to construct the gC-deleted FHV-1 BAC. Diagrams at the left column depict events at cellular level, while the right column depicts events at DNA level. Recombination arms with the same sequence are shown by a line in the same color (red and blue). Kan: kanamycin resistance gene expression cassette; S: I-SceI recognition site; arrows: priming sites of RM1188 and RM1189. 1: E. coli SW105 cells carrying FHV-l BAC were induced for expression of recombination proteins, which were encoded in mini-lambda DNA. 2: Cells flom step 1 were transformed with a mutating DNA flagment produced by PCR. 2A: Homologous recombination results in the replacement of the gC gene by kan. 3: Cells flom step 2 were transformed with pBAD- I-Scel, a plasmid that carries endonuclease I-Scel. 4: Expression of I-Scel was induced by arabinose. 5: Cells were induced for expression of recombination proteins. 5A: BAC DNA was excised by I-Scel, and kan was removed by the second homologous recombination, resulted in a scarless gC deletion. 6: The gC-deleted FHV-1 BAC was isolated flom E. coli. 121 mini-7. 119021113 gimme FHV-1 BAC E. CO/i/ gigao target SW105 gene 1. Induction at 42°C 2A- ” . ”“8“"9 mm FHV-1 PCR fragment UL43 ' UL45 BAC ~ . §-— Mutating 4425.. 2. Electroporation kan PCRf'agme'“ O pBADrlrSQeI A ‘ ' 4." Odo UL43 UL45 0?) 4'. 3. Electroporation 5A. i_‘, ._ UL43 UL45 I 4. Induction with arabinose l UL45 [ 5. Induction at 42°C . @J FHV-1 BAC [6. BAC isolation @ ”“5 gC‘ mutant 0 Figure 4.1 122 2K 1.6K Figure 4.2 SalI flagments of purified FHV-l BAC (A), FHVlBACAUL44 (B), and FHVlBACAUS8 (C) were resolved in a 0.7% agarose gel and visualized by ethidium bromide staining. DNA fragments of 10,120 bp and 769 bp (pink arrows) merged into a 10,120 bp fragment because of gC deletion. DNA flagrnents of 14,020 bp (blue arrows) became 12,417 bp because of gE deletion. Sizes of the ladder are indicated in bp. 123 Figure 4.3 (A) PCRs carried out using primers RMl 188 and RM1189 were resolved in a 1% agarose gel and visualized by ethidium bromide staining. The 1,879-bp fragment was amplified flom the FHV-l C-27 strain (Lane 1), and the 274-bp flagment was amplified from FHVlAgC (Lane 2). (B) PCRs carried out using primers RM94 and RM1193 were resolved in a 1% agarose gel and visualized by ethidium bromide staining. The 1,867-bp fragment was amplified flom the FHV-l C-27 strain (Lane 2), and the 268—bp fragment was amplified flom FHVlAgC (Lane 1). Sizes of the ladders are indicated in bp. 124 Figure 4.4 Development of one FHVlAgC plaque (right panel) and one C-27 strain plaque (left panel) over a period of 5 days. Magnification, 10X. 125 _ «I ... I . .3 i , T I A _ I." _ ‘3’ ‘—0— FHV-1 C-27 extracellular ‘-r— FHV-1 C-27 intracellular . + FHV1AgC extracellular —r *— FHV1AgC intracellular - FHV1ABAC extracellular ‘ ~ FHV1ABAC intracellular I Log Titer ncrcsolml) A 0 7 ' ‘ ' i r 0 10 20 30 40 50 Time (hours p.|.) Figure 4.5 Single-step growth kinetics of the FHV-1 C-27 strain, FHV IABAC, and the FHVlAgC. The CRFK cells were infected with the respective virus strain at an MOI of 3. At the indicated times intracellular and extracellular virus were harvested and titrated on CRFK cells. All growth kinetics was performed in triplicate except for intracellular and extracellular FHV1 ABAC (1 repeat). Error bars represent :l:l standard deviation. 126 ‘—¢— FHV—1 C-27 extracellular i—o— FHV-1 c-27 intracellular l i -— FHV1AgC extracellular i_D_ Fl-N1AgC intracellular ‘ Fl-N1ABAC extracelluliag Log Tlter (T CIDsoIml) 0 24 48 72 96 120 Time (hours p.|.) Figure 4.6 Multi-step growth kinetics of the FHV-1 C—27 strain, FHVlABAC, and the FHV1 AgC. The CRFK cells were infected with the respective virus at an MOI of 0.01. At the indicated times intracellular virus (open symbols) and extracellular virus (filled symbols) were harvested and titrated on CRFK cells. All growth kinetics was performed in triplicate. Error bars represent :tl standard deviation. 127 E —+— Fl-N-1 C-27 extracellular 3 -°- FHV-1 C-27 intracellular g --1-— FHV1ABAC extracelular 5 r FHV1ABAC intracellular i: + FHV1AgE extracelular _6'.’ ,,*ET_EW1A9§l9£@QflIar 0 10 20 30 40 50 Time (hours p.|.) Figure 4.7 Single-step growth kinetics of the FHV-l C-27 strain, FHVIABAC, and the FHVlAgE. The CRFK cells were infected with the respective virus strain at an MOI of 3. At the indicated times intracellular and extracellular virus were harvested and titrated on CRFK cells. Growth kinetics for FHV-1 C-27 strain was performed in triplicate. Growth kinetics for FHVIABAC and F HVlAgE is shown in 1 repeat. Error bars represent i1 standard deviation. 128 —o— Fl-N-1 C-27 extracellular —o- Fl-N-1 C-27 intracellular a. Fl-N1ABAC extracellular «__Lwizlggexrragelular, Log Titer (1'6ngde 0 24 48 72 96 1 20 Time (hours p.i.) Figure 4.8 Multi-step growth kinetics of the FHV-1 C-27 strain, FHVIABAC, and the FHV lAgE. The CRFK cells were infected with the respective virus at an MOI of 0.01. At the indicated times intracellular virus (open symbols) and extracellular virus (filled symbols) were harvested and titrated on CRF K cells. All growth kinetics was performed in triplicate. Error bars represent :l:1 standard deviation. 129 CHAPTER 5 GENERATION OF gC-gE-, US3 PROTEIN KINASE (PK): AND PK-gE- MUTANTS OF F ELINE HERPESVIRUS 1 (F HV—l) 130 5. 1 Introduction In addition to the glycoprotein C (gC) and E (gE) deleted FHV-l mutants described in Chapter 4, additional FHV-l genes were selected, individually or in combination, as targets for mutagenesis. As reviewed in Chapters 1 and 4, glycoprotein E (gE) is a virulence factor of FHV-1. The gE/gI heterodimer functions in cell-to-cell spread of the virus and spread of infection throughout the host nervous system, ultimately contributing to neurovirulence. A partial gE/ g1 deletion mutant previously generated in our laboratory using traditional methods produced smaller plaques and exhibited significantly reduced virulence (Sussman et al., 1995). This FHV-l gE/gI mutant, when administered via the oronasal route, can protect cats from clinical signs and significantly reduce viral loads in subsequent challenge with a high dose of field virus (Kruger et al., 1996; Sussman et al., 1997). However, this mutant at higher dose levels still retained partial virulence as it can produce mild clinical signs. We hypothesize that further attenuation can be achieved by deletion of additional virulence factors, resulting in a vaccine candidate that is safer and more efficacious. The US3 gene of FHV-1 encodes a serine/threonine protein kinase (PK), and its amino acid sequence is conserved in the subfamily Alphaherpesvirinae (Frame et al., 1987; McGeoch and Davison, 1986; Purves et al., 1987). Possible functions of PK include blocking of apoptosis induced by both viral and cellular proteins (Leopardi et al., 1997; Munger et al.,. 2001; Munger and Roizman, 2001; Ogg et al., 2004), regulation of the nuclear egress of progeny nucleocapsids (Reynolds et al., 2001; Reynolds et al., 2002), and control of the morphology of infected cells (Kato et al., 2008; Murata et al., 131 2002). Kimman et al. demonstrated that 3 PK' mutant of pseudorabies virus (PRV) has strongly reduced virulence, and animals inoculated with PK-gE- PRV mutant and subsequently challenged with wild-type virus has reduced virus shedding (Kimman et aL,1994) We expect that single deletion of PK will attenuate FHV-1, and an additional gC or PK deletion in the gE-deleted FHV-1 mutant will further attenuate the virus, and thus improve its safety as a vaccine. 5.2 Materials and Methods Cells, Viruses, Plasmids, and DNA Preparation The methodologies are presented in Chapters 3 and 4. Construction of Mutant Viruses FHV-l PK- mutant was constructed using the FHV-l BAC clone described in Chapter 3. FHV-1 gC-gE', and PK'gE' mutants were constructed using the FHV-l gE' BAC clone described in Chapter 4. Site-specific and “scarless” deletions of F HV -1 open reading frames (ORFs) UL44 (encoding gC) and U58 (encoding gE) were carried out using the two-step Red-mediated recombineering methods developed by Tischer et al. (2006), as described in Chapter 4. Briefly, mutating DNA fragments were generated by PCR using the primers dUL44F/dUL44R (for gC deletion), dUSSF/dUSSR (for gE deletion), or dUS3F/dUS3R (for PK deletion) (Table 5.1). In the first recombination, 132 these PCR mutating fragments replaced target gene with kanamycin resistance gene (kan), an I-SceI recognition site, and an additional 50-bp upstream recombination arm. The entire ORF of gC and gE was deleted. Because part of the US3 gene was present twice in the FHV-l I BAC (Chapter 3), only the first 793 hp at the N-terminal of the entire 1,056-bp ORF were deleted. The kan, I-SceI site, and additional recombination arm were subsequently removed in the second recombination, resulting in a scarless deletion. Mutant FHV-1 virions were reconstituted from the mutated BAC as described in Chapters 3 and 4. PCR assays were designed for differentiation of deletion mutants and wild type virus throughout the entire process (Table 5.1). Table 5.1 Primers used in this study Primer Sequence (5’ - 3’) Use dUL44F taagtagtcccgcgagacgatttacatcccgggatcaccaacaat Generation of gC- ctgchGGATGACGACGATAAGTAGGG mutating fragment dUL44R ataaccgctgaaacacggttatgataagtaatttatacgaactata agagcgcagattgtjggtgatcccgggatgtafltcgtctcgcggga WWgtagttaCAACCAATTAACCAATTCTGATTAG dUS3F agacgtagggcaggtttctgttatttaggaagtgtatacgtttggcta Generation of gE- atAGGATGACGACGATAAGTAGGG mutating fragment dUS3R ctctgggtgaacctttagagtggttataactttacgaagatggttgcg 33W ggCAACCAATTAACCAATTCTGATTAG RM1188 AGTCGAAATCACAGGCAGTG Differentiation of gC- RM1189 TGGAGTCGGTTTATATCCATAC mutant and wild type RM1343 AGGCACTCAGTGGGCCAAAGT Differentiation of PK- RM1346 AGGCTGTCTTACACATGAGGCA mutant and wild type Sequence in upper-case, nucleotides annealing to template; lower-case, hanging nucleotides; italic, upstream homologous recombination arm; underlined, downstream homologous recombination arm. 133 5.3 Results and Discussions Current progress of the construction and in vitro characterization of FHV-1 mutants are summarized in Table 5.2. Construction and in vitro characterization of FHV-l gC- and gE_ mutants has been completed and reported in Chapter 4. FHV-1 gC' and gE- mutant viruses that carry the BAC vector and EGFP marker was also generated, but not yet characterized. Construction of PK’ and gC'gE- mutant viruses, with and without the BAC vector, has been completed. Preliminary findings shows that these mutant FHV-1 viruses grew poorer in CRFK cells compared to the gC- mutant, and needed to be propagated by co-cultivating infected and uninfected cells. The US3 gene in a gE' FHV-l BAC clone has been replaced by kan, resulting in a gE'PK"kan+ BAC clone. To obtain scarless deletion mutant viruses, either with or without the BAC vector and EGFP marker (i.e., gE‘PK'BAC+ or gE'PK'BAC‘), kan will need to be removed by the second Red-mediated recombination. 134 Table 5.2 Current progress of the construction and in vitro characterization of FHV-1 mutants. Construction In vitro characterization First Second Propaga- Multi- Single- Plaque Virus recom- recom- tron Of step step size bination bination mutant gr OWth gr OWth analysis vrrus curve curve gC'BAC+ o o o o O o gC‘BAC' O O O O O I gE-BAC+ o o o o o o gE-BAC' O O O O G O 1 PK'BAC+ o o o o o o .- PK-BAC' O O O 1 O O : gC-gE'BAC+ O O O O O O j gC‘gE‘BAC‘ o o o o o o - gE'PK'BAc+ o c o O o o l gE'PK—BAC' O C O O O O 0: completed, 1: in progress, 0: to be done. 135 5.4 Reference Frame, M.C., Purves, F.C., McGeoch, D.J., Marsden, H.S., Leader, DR, 1987. Identification of the herpes simplex virus protein kinase as the product of viral gene US3. J Gen Virol 68, 2699-704. Kato, A., Tanaka, M., Yamamoto, M., Asai, R., Sata, T., Nishiyama, Y., Kawaguchi, Y., 2008. Identification of a physiological phosphorylation site of the herpes simplex virus l-encoded protein kinase US3 which regulates its optimal catalytic activity in vitro and influences its function in infected cells. J Virol 82, 6172—89. Leopardi, R., Van Sant, C., Roizman, B., 1997. The herpes simplex virus 1 protein kinase US3 is required for protection from apoptosis induced by the virus. Proc Natl Acad Sci U S A 94, 7891-6. Kimman, T.G., De Wind, N., De Bruin, T., de Visser, Y., Voermans, J., 1994. Inactivation of glycoprotein gE and thymidine kinase or the US3-encoded protein kinase synergistically decreases in vivo replication of pseudorabies virus and the induction of protective immunity. Virology 205, 511-8. Kruger, J.M., Sussman, M.D., Maes, R.K., 1996. Glycoproteins g1 and gE of feline herpesvirus-l are virulence genes: safety and efficacy of a gl-gE deletion mutant in the natural host. Virology 220, 299-308. McGeoch, D.J., Davison, A.J., 1986. Alphaherpesviruses possess a gene homologous to the protein kinase gene family of eukaryotes and retroviruses. Nucleic Acids Res 14, 1765-77. Munger, J ., Chee, A.V., Roizman, B., 2001. The U(S)3 protein kinase blocks apoptosis induced by the d120 mutant of herpes simplex virus 1 at a premitochondrial stage. J Virol 75, 5491-7. Munger, J., Roizman, B., 2001. The US3 protein kinase of herpes simplex virus 1 mediates the posttranslational modification of BAD and prevents BAD-induced programmed cell death in the absence of other viral proteins. Proc Natl Acad Sci U S A 98,10410-5. Murata, T., Goshima, F., Nishizawa, Y., Daikoku, T., Takakuwa, H., Ohtsuka, K., Yoshikawa, T., Nishiyama, Y., 2002. Phosphorylation of cytokeratin 17 by herpes simplex virus type 2 US3 protein kinase. Microbiol Immunol 46, 707-19. Ogg, P.D., McDonell, P.J., Ryckman, B.J., Knudson, C.M., Roller, R.J., 2004. The HSV-1 US3 protein kinase is sufficient to block apoptosis induced by overexpression of a variety of Bel-2 family members. Virology 319, 212-24. 136 Purves, F.C., Longnecker, R.M., Leader, D.P., Roizman, B., 1987. Herpes simplex virus 1 protein kinase is encoded by open reading frame US3 which is not essential for virus growth in cell culture. J Virol 61, 2896-901. Reynolds, A.E., Ryckman, B.J., Baines, J.D., Zhou, Y., Liang, L., Roller, R.J., 2001. U(L)3l and U(L)34 proteins of herpes simplex virus type 1 form a complex that accumulates at the nuclear rim and is required for envelopment of nucleocapsids. J Virol 75, 8803-17. Reynolds, A.E., Wills, B.G., Roller, R.J., Ryckman, B.J., Baines, J.D., 2002. Ultrastructural localization of the herpes simplex virus type 1 UL31, UL34, and US3 proteins suggests specific roles in primary envelopment and egress of nucleocapsids. J Virol 76, 8939-52. Sussman, M.D., Maes, R.K., Kruger, J.M., 1997. Vaccination of cats for feline rhinotracheitis results in a quantitative reduction of virulent feline herpesvirus-1 latency load after challenge. Virology 228, 379-82. Sussman, M.D., Macs, R.K., Kruger, J.M., Spatz, S.J., Venta, P.J., 1995. A feline herpesvirus-l recombinant with a deletion in the genes for glycoproteins g1 and gE is effective as a vaccine for feline rhinotracheitis. Virology 214, 12-20. Tischer, B.K., von Einem, J ., Kaufer, B., Osterrieder, N., 2006. Two-step red-mediated recombination for versatile high-efficiency markerless DNA manipulation in Escherichia coli. Biotechniques 40, 191-7. 137 CHAPTER 6 CONCLUSIONS AND FUTURE DIRECTIONS 138 6. 1 Conclusions 6.1.1 The FHV-l BAC Clone The entire FHV-1 genome was cloned as a BAC and transformed into E. coli for maintenance and fiiture manipulation. The virus derived from this BAC clone grew to a high titer in cell culture, produced plaques that are similar in size to those produced by the C-27 parent strain, and is virulent in vivo. Therefore, this FHV-1 BAC clone is a very suitable starting platform for studies on viral virulence, as well as development of future vaccine candidates. 6.1.2 The Complete Sequence of the FHV-1 Genome Complete genomic sequences were derived from both the FHV-1 BAC clone and purified virion DNA. The FHV-l genome is 135,797 bp in size with an overall G+C content of 45%. A total of 78 open reading frames were predicted, encoding 74 distinct proteins. The gene arrangement is collinear with that of most sequenced varicelloviruses. 6.1.3 F HV-l Mutants Lacking Glycoprotein C (gC) or E (gE) FHV-1 mutants lacking the entire open reading flames encoding gC or gE were constructed, based on the FHV-1 BAC clone. Results of in vitro characterization indicate that gC plays a role in initial attachment/penetration, replication, and egress of FHV-1. The gC- mutant is more cell-associated and has a reduced in growth rate. In contrast, the gE' FHV-l grew to a high titer in cell culture. Results of in vitro characterization suggest that virus egress, and most likely cell-to-cell spread were affected by gE deletion, while virus entry and replication were not. Both mutants are 139 likely to be attenuated, and further studies are necessary to evaluate their in vivo characteristics and potential as vaccine candidates. 6.1.4 Other Deletion Mutants A gC’gE‘, an US3 protein kinase (PK)_, and a gE—PK"l105°F 3 each day Conjunctivitis Serous discharge only 1 to 3 1 >4 2 Mucopurulent discharge 1 to 2 2 3 to 5 4 a6 6 Rhinitis Serous discharge only 1 to 3 1 >4 2 Mucopurulent discharge 1 to 2 2 3 to 5 4 36 6 Sneezing 1 each day Dyspnea Audible rales 2 each day Coughing 2 each day Open mouth breathing 3 each day Depression Anorexia 1 each day Dehydration 1 to 2 3 23 4 Hypothermia <99°F 2 each day Oral ulcers (linguinal or oral mucosa) 1 ulcer <4 mm 1 to 5 2 6 to 9 3 310 4 Multiple ulcers <4 mm 1 to 4 3 5 to 8 5 >9 7 Ulcer or ulcers >4 mm 1 to 4 5 5 to 8 7 29 9 Salivating 1 each day Extenal ulcers (lip or nares) Nonbleeding ulcer 4 Bleeding ulcer 6 Death 15 ‘ Sussman, M.D., Maes, R.K., Kruger, J.M., Spatz, S.J., Venta, P.J., 1995. A feline herpesvirus-l recombinant with a deletion in the genes for glycoproteins g1 and gE is effective as a vaccine for feline rhinotracheitis. Virology 214, 12-20. USDA Supplemental Assay Method 311 (US. Department of Agriculture, Animal and Plant Health Inspection Service, National Animal Veterinary Services Laboratory, 1985.) 145 Appendix B Clinical scores recorded in the pilot in vivo study (Chapter 3) Cat Identification: AJ03 Group: Negative Control Color: Orange Sex: F Age: 11-13 weeks Scoring: Janice Quail Cement mmmmm mmmmmafimmmm 0 09/04/07 N N N N N 1 09/05/07 N N N N N N N 2 09/06/07 N N N N N N N 3 09/07/07 N N N N N N N 4 09/08/07 N N N N N N N 5 09/09/07 N N N N N N N 6 09/10/07 N N N N N N N 7 09/11/07 N N N N N N N 6 09/12/07 N N N N N N N 9 09/13/07 N N N N N N N 10 09/14/07 N N N N N N N 11 09/15/07 N N N N N N N 12 09/16/07 N N N N N N N 13 09/17/07 N N N N N N N 14 09/16/07 N N N N N N N Score - 0 0 0 0 - — 0 Nasal Qtai External 122: max Discharge Salazar/m 1119mm: ulceration lmtratiszn Alumna 12mm 19mm 0 N N N N N Y 36. 6 97. 9 1 N N N N N Y - - 2 N N N N N Y — - 3 N N N N N Y - - 4 N N N N N Y - — 5 N N N N N Y - - 6 N N N N N Y — — 7 N N N N N Y - - 8 N N N N N Y - - 9 N N N N N Y — — 10 N N N N N Y — — 11 N N N N N Y - - 12 N N N N N Y - - 13 N N N N N Y - — 14 N N N N N Y — - Score 0 0 0 0 0 0 — 0 Total Score = 0 Y: yes; N: no; S: serous; MP: mucopurulent; —: not applicable. 146 Appendix B, continued Cat Identification: AJ03 Group: Negative Control Color: Orange Sex: F Age: 11-13 weeks Scoring: Sheldon Qnen Qnmeel MenthBLennzuleenniannuler QAXQateSneezeQnennBeleeflre'fmeneeleneeimmlme 0 09/04/07 N N N N N 1 09/05/07 N N N N N N N 2 09/06/07 N N N N N N N 3 09/07/07 N N N N N N N 4 09/08/07 N N N N N N N 5 09/09/07 N N N N N N N 6 09/10/07 N N N N N N N 7 09/11/07 N N N N N N N 8 09/12/07 N N N N N N N 9 09/13/07 N N N N N N N 10 09/14/07 N N N N N N N 11 09/15/07 N N N N N N N 12 09l16/07 N N N N N N N 13 09/17/07 N N N N N N N 14 09I18/07 N N N N N N N Score — 0 0 0 0 - — 0 Nasal Qrel External 122 nnx Qienlleme Salli/alien uleeLallnn itinerarinn Malina Anmiite Ielnnlm Iemnifl N N N N 36 6 97.9 1 N N N N N Y 37.3 99.1 2 N N N N N Y — — 3 N N N N N Y - — 4 N N N N N Y — - 5 N N N N N Y — — 6 N N N N N Y — - 7 N N N N N Y - — a N N N N N Y - — 9 N N N N N Y — — 10 N N N N N Y - - 11 N N N N N Y - - 12 N N N N N Y - - 13 N N N N N Y - - 14 N N N N N Y - - Score 0 0 O O O 0 — 0 Total Score = 0 Y: yes; N: no; S: serous; MP: mucopurulent; —: not applicable. 147 Appendix B, continued Cat Identification: AJCZ Group: BAC Color: Grey Tabby Sex: F Age: 11-13 weeks Scoring: Janice Qnen Corneal Merlinmannzmnelenflnder QMQetefimezeQnuehBeheflreehenaethneniMQieeneme 0 09/04/07 N N N N N N 1 09/05/07 N N N N N N N 2 09/06/07 N N N N N N N 3 09/07/07 N N N N N N N 4 09/08/07 N N N N N N N 5 09/09/07 N N N N N N S 6 09/10/07 Y N N N N N S 7 09/11/07 Y N Y N N N S 8 09/12/07 Y Y Y N N N S 9 09/13/07 Y N Y N N N N 10 09/14/07 N N Y SLIGHT N N N N 11 09/15/07 N N Y SLIGHT N N N N 12 09/16/07 N N N N N N N 13 09/17/07 Y N Y SLIGHT N N N N 14 09/18/07 N N N N N N N Score - 5 2 12 0 - — 2 Diesel 913! Enemel De; 12A! Discharge W Uleeratien uleeralinn lmlretinn Annette Iemnfl). Iemnlfl 0 N N N N 37. 2 99.0 1 N N N N N Y 37.1 98.8 2 N N N N N Y 33.1 91.6 3 N N N N N Y 37.0 98.6 4 N N N N N Y 37.8 100.0 5 N N N N N Y 38.0 100.4 6 N N N N N Y 37.0 98.6 7 S N N N Y SLIGHT Y 37.5 99.5 8 N N N N N Y SLIGHT 37.0 98.6 9 S N N N Y SLIGHT N 37.6 99.7 10 N N N N N Y 37.1 98.8 11 N N N N N Y 37.0 98.6 12 S N N N N Y 37.6 99.7 13 N N N N N Y 37.0 98.6 14 N N N N N Y 37.7 99.9 Score 1 0 0 0 3 2 - 0 Total Score = 27 Y: yes; N: no; S: serous; MP: mucopurulent; —: not applicable. 148 Appendix B, continued Cat Identification: AJCZ Group: BAC Color: Grey Tabby Sex: F Age: 11-13 weeks Scoring: Sheldon an Semeal Mnethfllennzttleenalengnular DAYQateSneezeSnuehBaleeEreathenaethnanlMDIeename 09/04I07 N N N 1 09/05/07 N N N N N N N 2 09/06/07 N N N N N N N 3 09I07/07 N N N N N N N 4 09/08/07 N N N N N N N 5 09/09/07 N N N N N N S 6 09/10/07 Y N N N N N S 7 09l1 1/07 Y Y N N N N S 8 09l12/O7 Y Y N N Y N S 9 09/13/07 Y N N N N N N 10 09/14/07 N N N N N N N 1 1 09I15/07 N N N N N N N 12 09I16/07 N N N N N N N 13 09/17/07 Y N N N N N N 14 09/18/07 N N N N N N N Score — 5 4 0 0 - - 2 Qrel External De; DA! WWWWMMMM N N N N 37. 2 99. 0 1 N N N N N Y 37.3 99.1 2 N N N N N Y 38.4 101.1 3 N N N N N Y 37.0 98.6 4 N N N N N Y 37.8 100.0 5 N N N N N Y 38.0 100.4 6 N N N N N Y 37.2 99.0 7 S N N N Y Y 37.5 99.5 8 N N N N N N 37.0 98.6 9 N N N N N Y 37.6 99.7 10 S N N N N Y 37.1 98.8 1 1 S N N N N Y 37.5 99.5 12 S N N N N Y 37.0 98.6 13 N N N N N Y 37.0 98.6 14 N N N N N Y 37.7 99.9 Score 2 0 0 0 3 1 - 0 Total Score = 17 Y: yes; N: no; S: serous; MP: mucopurulent; —: not applicable. 149 Appendix B, continued Cat identification: AJCS Group: BAC Color: Brown Grey Tabby Sex: F Age: 11-13 weeks Scorinnganice Qnen Semeal Mnutn Slenn; weenainn Qallar 12A! Date Sneeze Senna Bales SLeNatn enaem lQnaeInaQieenarne 0 09/04/07 N N N N N 1 09/05/07 N N N N N N N 2 09/06/07 N N N N N N N 3 09/07/07 N N N N N N N 4 09/08/07 N N N N N N N 5 09/09/07 Y N N N N N N 6 09/10/07 N N N N Y N N 7 09/11/07 Y N Y N Y N S 8 09/12/07 Y N Y N Y N S 9 09/13/07 N Y Y N Y N S 10 09/14/07 Y Y Y N N N S 11 09/15/07 Y N Y SLIGHT N N N S 12 09/16/07 Y N Y SLIGHT N N N S 13 09/17/07 Y N Y SLIGHT N N N MP? 14 09/18/07 N N N N N N S Score — 7 4 14 0 - — 2 Nasal Qrel Extemal Qt 12A! Discharge Sallxatine Mineratinn tlleetattnn hieratinn Annette Iean’Qt Iemnlfl N N N N 37. 2 99. o 1 N N N N N Y 37.3 99.1 2 N N N N N Y 37.4 99.3 3 N N N N N Y 36.0 96.8 4 N N Y N N Y 38.6 101.5 5 N N Y N N Y 38.0 100.4 6 N N Y N N Y 38.3 100.9 7 S N N N Y SLIGHT Y 37.0 98.6 8 S N Y N Y N 37.4 99.3 9 S N N N Y SLIGHT N 37.7 99.9 10 MP N N N Y N 36.3 97.3 11 S N N N Y SLIGHT Y 37.2 99.0 12 S N N N N Y 36.8 98.2 13 N N N N N Y 37.5 99.5 14 S N N N N Y 37.0 98.6 Score 2 0 2 0 4 3 — 0 Total Score = 38 Y: yes; N: no; S: serous; MP: mucopurulent; —: not applicable. 150 Appendix B, continued Cat identification: AJC3 Group: BAC Color: Brown Grey Tabby Sex: F Age: 11-13 weeks Scoring: Sheldon Qnen Semen! Mnlltn Eleni]; weenatnn inlar 0A1 Date Sneeze Seven Bales Steam enaem thenim Discharge 09/04/07 09/05/07 09/06/07 09/07/07 09/08/07 09/09/07 09/10/07 09/11/07 09/12/07 09/13/07 09/14/07 09/15/07 09/16/07 09/17/07 09/18/07 uz<<<