‘Ar .1 e. ‘ .4! , an”. _ 3.1:, {iv .. sQ 2.: Luv: 1 .1 I}! z.- .n 1 his .fi! jt.‘i£ .- "h .41....vtila...‘ "Ab|lv|'“ ‘ .9. (pitfall . .l.i\.... (.nfl. ‘1 ‘ l. 455..., Ill. . {.13‘1563‘. . 1. .l n! . .‘ unintuislnHE tliit). . .437. .5 .Jml'ulflii. .41 ”W; it. A. . 4%. may? V . _LlBFtARY MlCtht‘m State University This is to certify that the thesis entitled MOLECULAR PATHOLOGY OF FHV—1 INFECTION WITHIN FELINE CORNEA, CONJUNCTIVA, LACRIMAL GLAND. NICTITANS GLAND. TRIGEMINAL GANGLION AND CILIARY Master of Science GANGLION presented by SUSAN JACOBI has been accepted towards fulfillment of the requirements for the degree in Department of Small Animal Clinical Sciences 4 n QJWV 4M Tm DWI ow: Mspflwo Mag? Professor’s Signatdre (chi/08 Date MSU is an affirmative-action, equal-opportunity employer PLACE IN RETURN BOX to remove this checkout from your record. TO AVOlD FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 5/08 K'lProlecc8PresICIRC/DateDue,indd MOLECULAR PATHOLOGY OF FHV-l INFECTION WITHIN FELINE CORNEA, CONJUNCTIVA, LACRIMAL GLAND, NICTITANS GLAND, TRIGEMINAL GANGLION AND CILIARY GANGLION By Susan Jacobi A THESIS Submitted to Michigan State University in partial fulfillment of the requirements for the degree of MASTER OF SCIENCE Department of Small Animal Clinical Sciences 2008 ABSTRACT MOLECULAR PATHOLOGY OF FHV-l INFECTION WITHIN FELINE CORNEA, CONJUNCTIV A, LACRIMAL GLAND, NICTITAN S GLAND, TRIGEMINAL GANGLION AND CILIARY GANGLION By Susan Jacobi Feline herpesvirus 1 (FHV-l) is one of the major causes of feline upper respiratory tract and ocular disease. Despite intensive research in this area, the pathogenesis of FHV-l infection within ocular and neural tissues remains poorly understood. In this study we utilized virus isolation, a quantitative real-time polymerase chain reaction, histopathologic examination, and in situ hybridization to quantify the amount F HV-l present and evaluate the associated morphological changes within the corneas, conjunctiva, lacrimal glands, nictitans glands, trigeminal ganglia, and ciliary ganglia of experimentally inoculated specific pathogen free cats during both the acute and latent phases of infection. We determined F HV -1 is able to infect not only conjunctiva] epithelium, corneal epithelium, and trigeminal ganglion, but also the ciliary ganglion. The ciliary ganglion may serve as an additional site of latency for FHV-l and could explain the preponderance of ocular lesions noted during recrudescent infections. However additional studies are required to explore this theory. The theory that lacrimal adenitis contributes to the development of KCS was not supported as there was no evidence of FHV-l infection or secondary inflammation within the glandular tissues at any time. TABLE OF CONTENTS LIST OF TABLES ................................................................................................... V LIST OF FIGURES ................................................................................................. VI 1. CHAPTER - FELINE HERPESVIRUS 1 LITERATURE REVIEW .............. 1 1.1 Feline herpesvirus 1 — Classification ..................................................... 1 1.2 Feline herpesvirus 1 — Characteristics .................................................... 1 1.3 Feline herpesvirus 1 — Replication ......................................................... 2 1.4 Feline herpesvirus 1 — Latency .............................................................. 4 1.5 Feline herpesvirus 1 disease - Epidemiology and pathogenesis .............. 6 1.6 Feline herpesvirus 1 disease — Other species .......................................... 9 1.7 Feline herpesvirus 1 disease - Systemic manifestations ........................ 13 1.8 Feline herpesvirus 1 disease - Ocular manifestations ........................... 14 1.8.1 Conjunctivitis .......................................................................... 14 1.8.2 Keratitis ................................................................................... 15 1.8.3 Stromal keratitis ....................................................................... 16 1.8.4 Keratoconjunctivitis sicca ........................................................ 18 1.8.5 Ophthalmia neonatorum and symblepharon ............................. 20 1.8.6 Corneal sequestrum .................................................................. 20 1.8.7 Eosinophilic keratitis ............................................................... 21 1.8.8 Anterior uveitis ........................................................................ 22 1.9 Feline herpesvirus 1 disease — Diagnosis ............................................. 23 1.9.1 Virus isolation ......................................................................... 23 1.9.2 Serology .................................................................................. 24 1.9.3 Fluorescent antibody test .......................................................... 25 1.9.4 PCR assays .............................................................................. 26 1.9.5 In situ Hybridization ................................................................ 28 1.9.6 FHV-l detection in different tissues ......................................... 28 1.10 Feline herpesvirus 1 disease - Treatment and prevention ..................... 31 1.11 Antiviral therapy ................................................................................. 33 1.11.1 Nucleoside analogues ............................................................... 33 1.11.2 Acyclovir ................................................................................. 34 1.11.3 Valacyclovir ............................................................................ 35 1.11.4 Ganciclovir .............................................................................. 35 1.11.5 Penciclovir ............................................................................... 36 1.11.6 Famciclovir .............................................................................. 37 1.11.7 Idoxuridine .............................................................................. 37 1.11.8 Trifluridine .............................................................................. 38 1.11.9 Vidarabine ............................................................................... 38 1.11.10 Cidofovir ................................................................................. 39 1.11.11 Bromovinyldeoxyuridine ......................................................... 39 1.11.12 Ribavirin .................................................................................. 40 1.12 Efficacy of nucleoside analogues against FHV-l ................................. 40 1.13 Non-nucleoside antivirals .................................................................... 42 1.13.1 Foscamet ................................................................................. 42 1.13.2 L-Lysine .................................................................................. 42 iii 1.13.3 Interferons ............................................................................... 45 1.13.4 Bovine lactoferrin .................................................................... 46 1.13.5 Vaccination .............................................................................. 47 1.14 Conclusions ......................................................................................... 50 2. CHAPTER — ANATOMY AND HISTOLOGY OF TISSUES RELEVANT TO THIS STUDY ................................................................... 52 2.1 Cornea ................................................................................................ 52 2.2 Conjunctiva ......................................................................................... 53 2.3 Lacrimal gland .................................................................................... 53 2.4 Nictitans gland .................................................................................... 54 2.5 Trigeminal ganglion ............................................................................ 55 2.6 Ciliary ganglion .................................................................................. 56 3. CHAPTER - HY POTHESES AND SPECIFIC AIMS .................................. 57 3.1 Hypotheses .......................................................................................... 57 3.2 Specific aims ....................................................................................... 57 4. CHAPTER — RESEARCH PROTOCOL, RESULTS, AND DISCUSSION .............................................................................................. 59 4.1 Introduction ......................................................................................... 59 4.2 Materials and Methods ........................................................................ 61 4.2.1 Animals ................................................................................... 61 4.2.2 Inoculation and clinical scoring ................................................ 61 4.2.3 Samples ................................................................................... 62 4.2.4 Virus isolation ......................................................................... 63 4.2.5 Quantitative real time PCR ...................................................... 63 4.3 Results ................................................................................................ 71 4.3.1 Clinical features ....................................................................... 71 4.3.2 Virus isolation ......................................................................... 72 4.3.3 Quantitative real time PCR ...................................................... 73 4.3.4 Histopathology ......................................................................... 74 4.3.5 In situ hybridization ................................................................. 78 4.4 Discussion ........................................................................................... 80 APPENDICES ........................................................................................................ 87 5. APPENDIX A ............................................................................................. 88 6. APPENDIX B .............................................................................................. 89 7. APPENDIX C .............................................................................................. 94 8 APPENDIX D ............................................................................................. 95 9. BIBLIOGRAPHY ...................................................................................... 101 iv Table 4.1. Table 4.2. Table 4.3. Table 4.4. Table 5.1. Table 7.1. Table 7.2. Table 7.3. Table 8.1. Table 8.2. Table 8.3. Table 8.4. Table 8.5. Table 8.6. LIST OF TABLES Clinical scores ......................................................................................... 72 Summarized results of virus isolation (positive/total) .............................. 73 Summarized results of quantitative real time PCR (positive/total) ........... 74 Summarized results of in situ hybridization ............................................. 78 USDA Scoring Method for FHV-l .......................................................... 88 Virus isolation day 6 post inoculation ...................................................... 94 Virus isolation day 10 post inoculation .................................................... 94 Virus isolation day 30 post inoculation .................................................... 94 PCR results of cornea (Copy no. / 100 ng DNA) ..................................... 95 PCR results of conjunctiva (Copy no. / 100 ng DNA) ............................. 96 PCR results of lacrimal gland (Copy no. / 100 ng DNA) ......................... 97 PCR results of nictitans gland (Copy no. / 100 ng DNA) ......................... 98 PCR results of trigeminal ganglia (Copy no. / 100 ng DNA) ................... 99 PCR results of ciliary ganglia (Copy no. / 20 ng DNA) ......................... 100 LIST OF FIGURES Images in this thesis are presented in color. Figure 4.1. Standard melt curve chart. ..................................................................... 65 Figure 4.2. Standard melt peak chart ........................................................................ 65 Figure 4.3. Standard amplification chart, 10 fold dilutions of 106 genomic copies of FHV-l, triplicate. .......................................................................... 66 Figure 4.4. PCR standard curve. .............................................................................. 66 Figure 4.5. FHV-l pellet pre-treated with different protocols ................................... 68 Figure 4.6. FHV-l pellet pre-treated via micowaving for 10 minutes using an antibody dilution of 1:250. ........................................................................... 69 Figure 4.7. Conjunctiva] sample from cat # 12. (HE) ............................................... 76 Figure 4.8. Corneal sample from cat # 9. (HE) ......................................................... 76 Figure 4.9. Lacrimal gland sample from cat # 2. (HE) ............................................. 77 Figure 4.10. Trigeminal ganglion sample from cat # 1. (HE) ................................... 77 Figure 4.11. In situ hybridization of the conjunctiva] sample from cat # 12 .............. 79 Figure 4.12. In situ hybridization of the corneal sample from cat # 4. ...................... 79 Figure 6.1. Results of the clinical examination: sneezing. ........................................ 89 Figure 6.2. Results of the clinical examination: elevated body temperature. ............. 89 Figure 6.3. Results of the clinical examination: dehydration. ................................... 90 Figure 6.4. Results of the clinical examination: anorexia. ........................................ 90 Figure 6.5. Results of the clinical examination: ptyalism. ........................................ 91 Figure 6.6. Results of the clinical examination: oral ulceration. ............................... 91 Figure 6.7. Results of the clinical examination: nasal discharge. .............................. 92 Figure 6.8. Results of the clinical examination: blepharospasm ................................ 92 Figure 6.9. Results of the clinical examination: ocular discharge. ............................ 93 Figure 6.10. Results of the clinical examination: conjunctivitis ................................ 93 1. CHAPTER - FELINE HERPESVIRUS 1 LITERATURE REVIEW 1.1 Feline herpesvirus 1 — Classification The family of the herpesviridae is comprised of different subfamilies: alphaherpesvirinae, betaherpesvirinae, gammaherpesvirinae, and an unidentified subfamily.l Herpesvirz'dae are enveloped viruses that have an icosahedral capsid containing a linear double stranded DNA.1 Feline herpesvirus 1 (FHV-l) has been classified as a member of the varicellovirus genus of the subfamily alphaherpesvirz’nae.2'5 Prototypical members of this subfamily are human herpesvirus-1 (herpes simplex virus, HSV-l, genus simplexvirus), and human varicella-zoster virus (VZV, genus varicellovirus).1 Alphaherpesvirinae relevant to the veterinary field include equine herpesvirus 1 and 4 (EHV-l and 4), bovine herpesvirus-1 (BHV-l), canine herpesvirus-1 (CHV-l), pseudorabies virus (PRV, PHV-l), and gallid herpesvirus-2 (GHV-Z, Marek’s disease virus).6’7 Feline herpesvirus-1 is closely related genetically and antigenetically to CHV-l and phocid (seal) herpesvirus-1 (PhV-l), and cross-protection between feline and PhV-l has been reportedg'l6 1.2 Feline herpesvirus 1 - Characteristics The size of feline herpesvirus particles is 120-180 nm in diameter.17 Like most alphaherpesviruses, FHV-l is composed of four major structural elements including an electron-opaque core, an icosahedral capsid, a tegument, and an envelope."7 A single molecule of linear double stranded DNA is wrapped around the fibrous spool- 1ike core, which is anchored inside the capsid.1 The capsid is a structural characteristic of all herpesviruses, 100 nm in diameter, and consists of 150 hexameric and 12 - r,4,7,1 . - pentamenc capsomers. 7 The nucleocapsrd is surrounded by an electron dense 1,4,7 amorphous material, called the tegument. The outer layer of the virion is formed by a fragile lipoprotein envelope, which carries viral glycoproteins.l The physical structure of the FHV -1 genome is similar to the genomes of other alphahennesvirinae.18 Rota et a1., first characterized the genome of FHV-l strain C-27, the strain utilized in the present study, and determined that FHV-l DNA was approximately 134 kb and was composed of a long (L) and a short (S) segment. The long segment (U1) was 104 kb in size and was composed of unique DNA.18 The adjacent S segment was approximately 30 kb in size and contained a central portion of unique DNA (Us) which was approximately 8kb in size.18 The Us region was bounded by inverted repeat sequences which were 11 kb in size.18 Later Grail et a1., determined the size of the genome of F HV-l strain B-927 to be 126 kb, composed of a long unique region of 99 kb and a short region of 27 kb containing a unique sequence of 8-9 kb flanked by inverted repeats of 7-8.5 kb.19 1.3 Feline herpesvirus 1 — Replication The replication cycle of HSV-1 has been studied extensively. Most other alpha-herpesvirina follow a similar pattern. The different phases of replication are initiation, early gene expression, DNA replication, late gene expression, assembly, and release. 1’7’20 Epithelial cells of the nasal mucosa, conjunctiva, tonsils, and nasal turbinates are the target for primary infection by F HV -1 .21 During the initiation phase, multiple proteins participate in the attachment of the virus to the host cell.1 Three viral glycoproteins, gB, gC, and gD, are essential for HSV-l infection.l An FHV-l gB homolog has been identified and sequenced.22 Via gB and gC the virion binds to heparin sulfate or chondroitin sulfate proteoglycans of the extracellular matrix of the host cell.l To facilitate cellular entry, gD binds to a cellular co-receptor."20 This 2 contact initiates fusion of viral and plasma membranes.1 Viral nucleocapsids then attach to microtubules and are transported to the nucleus.23 The viral nucleocapsid docks at the nuclear pore, releasing viral DNA into the nucleus. L7 Gene expression of the FHV-l genome is believed to be similar to that of HSV-1 in human cells.“25 VP16, a tegument protein, interacts with host transcription proteins to stimulate transcription of immediate-early genes by host RNA polymerase II."26 Immediate-early mRNAs are spliced and transported to the cytoplasm where they are translated. The immediate-early proteins (or-proteins) are transported to the nucleus where they activate transcription of early genes and regulate transcription of immediate-early genes.l Early gene transcripts are transported to the cytoplasm. The early proteins (B-proteins) fimction primarily in DNA replication and production of substrates for DNA synthesis. Some fl-proteins are transported to the nucleus, while others function in the cytoplasm.1 DNA replication and recombination produces long, concatemeric DNA, the template for late gene expression. Late mRNAs are transported to the cytoplasm where they are translated.1 Late proteins (“y-proteins) function primarily as viral structural proteins or participate in virus assembly and particle egress. The precursor glycoproteins are also transported to the Golgi apparatus for further modification and processing. Mature glycoproteins are transported to the plasma membrane of the infected cell. 'y-proteins are transported to the nucleus for assembly of the nucleocapsid and DNA packaging. Newly replicated DNA is packaged into nucleocapsids. ' DNA containing nucleocapsids, together with some tegument proteins, bud from the inner nuclear membrane into the perinuclear lumen, acquiring an envelope.1 These immature enveloped virions fuse with the outer nuclear membrane, releasing the nucleocapsid into the cytoplasm.1 The structure is then transported to a late trans- Golgi compartment or endosome that contains mature viral membrane proteins.1 Here the particles acquire an envelope containing mature viral envelope proteins and the complete tegument layer.1 The enveloped virus particle then buds into a vesicle that is transported to the plasma membrane for release by exocytosis.1 Enveloped viruses tend to spread from cell to cell, which accounts for the formation of plaques and . . . . . . 27-29 syncytra seen during vrrus rsolatron 1n cell culture. 1.4 Feline herpesvirus 1 -— Latency Feline herpesvirus-1, similar to other herpesviruses can establish latency, a phase where replication is limited. The virus may become latent for the lifetime of the host. Three general properties characterize latent infections: expression of productive cycle viral genes is absent or inefficient; immune detection of the cell harbouring the latent genome is reduced or eliminated; and the viral genome persists intact so that at some later time a productive acute infection can be initiated.1 In the latent state, the genome can be maintained as a non-replicating chromosome in a non-dividing cell such as a neuron (e.g., HSV-l, VZV); become an autonomous, self replicating chromosome in a dividing cell (e.g., Epstein-Barr virus), or be integrated into a host chromosome (e.g., adeno-associated virus).1 During latency, a balance among the regulators of viral and cellular gene expression must be maintained. Therefore only a restricted set of viral gene products is produced.l For (Jr-herpesviruses it was generally believed that sensory neurons within ganglia were the primary site of latency.30 However, more recent studies have shown herpes simplex latency within the autonomic pathways of mice.31 The trigeminal ganglion is considered a primary site of latency following ocular, oral, or intranasal infection.32’33 Once latency is established, viral DNA persists in neurons for the life of 4 an infected host.30 It is unclear if latency induces neurologic injury.3'0 However, it has been shown that during reactivation from a latent infection, inflammation and destruction of neurons and surrounding tissue can occur.“35 In neurons latently infected with HSV-l the only abundantly transcribed gene is the latency-associated transcript (LAT).36’37 Therefore, LATs provide molecular markers for latently infected cells.3°’38 The length of the primary LAT transcripts is 8.3 to 8.5 “3.39,“, LAT overlaps the viral genes for ICPO and ICP34.5 in an anti-sense direction.”37 It has been suggested that LAT is a regulatory RNA that represses expression of ICPO.37 Although the LATs are first detectable at the peak of an acute infection, they are present at much higher levels during latency with high levels in 37““ Several studies suggested that LATs play a role in trigeminal ganglia. establishment of latency. It has been demonstrated that HSV-l LATs enhances establishment of latency in mice.42’43 Furthermore, it has been found, that LATs repress productive viral gene expression in trigeminal ganglia of mice during acute infection.“45 Although LATs are not mandatory for the virus to establish latency, viral mutants that are unable to express LATs are reactivation deficient“6 Therefore LATs may play a role in reactivation from latency. Different studies using various LAT mutants have shown that LATs increase the induced and spontaneous reactivation phenotypes in the rabbit ocular model“’48 and the induced reactivation phenotype in mice.42’49'52 LATs also have anti-apoptotic features and maintain latency by promoting the survival of infected neurons.53’54 Gupta et al.55 showed that microRNA encoded by the HSV-l LAT gene confers resistance to apoptosis. The anti-apoptotic activity of LATs is important during the latency-reactivation cycle. Because infected neurons would normally be eliminated by cytotoxic T cells, which kill their target cells in part by apoptosis, high levels of LAT within latently infected neurons may prevent their elimination.56 A functional relationship between the ability of LATs to promote cell survival and their ability to enhance spontaneous reactivation has been demonstrated.57 Primary and recrudescent herpetic disease is more prevalent among immune- compromised individuals.58 The fact that viral reactivation is associated with immune suppression and the persistence of T cells and cytokines in latently infected human trigeminal ganglia suggest that T cells play a role in control of the virus at the site of latency.5860 Theil et al.59 found T cells positive for CD3 and CD8 markers as well as CD68 positive macrophages in 30 of 42 trigeminal ganglia from 21 healthy humans. The presence of these cells correlated constantly with the occurrence of HSV-1 LATs. Uninfected trigeminal ganglia showed no immune cell infiltration. Quantitative real time PCR revealed an elevated cytokine/chemokine expression (IFN-y, TNF-or, RANTES, and IP-lO) in the latently infected trigeminal ganglia demonstrating that latent herpesviral infection in humans is accompanied by a chronic inflammatory process, but without neuronal destruction. It seems likely that the immune response maintains viral latency and inhibits viral reactivation. 1.5 Feline herpesvirus 1 disease - Epidemiology and pathogenesis Feline herpesvirus-1 is also known as feline rhinotracheitis virus. Feline viral rhinotracheitis was first recognized in 1957 in 5 to lO-week-old farm kittens that were being raised in a laboratoryzw Since then it has been recognized as a disease with world-wide distribution within the cat population. Epidemiologic studies indicate that as many as 50-75% of cats have serologic evidence of exposure to the virus.(’2’63 Other studies estimate that over 90% of cats are sero-positive for the virus;64 at least 80% of infected individuals remain latently infected for life; and approximately 45% of latently infected cats shed virus throughout life.65 Reactivation of virus may occur spontaneously but is most likely after stress.66 Re-excretion of F HV-l could be stimulated in more than 50% of cats recovered from F HV-l infection through systemic administration of corticosteroids or a change in housing.67 Detectable viral shedding does not immediately follow the stress.66 A lag phase of 4-11 days (mean 7.2 days) precedes the shedding of infectious virus.65 The period of shedding can range in duration from 1-13 days (mean 6.5 days).65'67 “”68 although in some cases carriers show Shedding carriers are mainly asymptomatic, recrudescence of mild clinical signs while they are shedding.69 During episodes of viral shedding infectious virus is present within oro-nasal and conjunctival secretions and serves as source of infection to other cats.”67 The enveloped virion is relatively fragile and very susceptible to heat, drying, or disinfectants.7°’7| At room temperature FHV-l can only persist in the enviromnent for 18-24 hours in relatively dry conditions.7°‘72’73 Transmission of FHV-] between cats occurs via direct contact, fomites, or macrodroplets which can be sneezed about 1.3 m.74 Management of infected cats within a hospital situation requires appropriate separation of the cats and routine hygiene practices for personnel and facilities.75 The virus survives less than one hour in Ophthalmic solutions used for routine diagnostics testing, such as fluorescein stain and topical ocular anesthetics. 76 However, the virus may persist for up to 5 days in eye wash solutions.76 Young cats are most susceptible to primary infection, especially around 8-12 weeks of age when maternal antibodies wane.70 Cats with underlying disease (feline leukaemia virus (FeLV), feline immunodeficiency virus (F IV)) are also very susceptible to FHV-l infection.77 Due to limited ability of FHV-l to replicate at temperatures of 37 °C or higher, the primary sites of replication are the cooler epithelial surfaces.70 The natural routes of infection are via nasal, oral, and conjunctival mucous membranes.“’70 In the acute phase of infection virus replication occurs predominantly in the mucous membranes of the nasal septum, turbinates, nasopharynx, and tonsils; other tissues including conjunctivae, mandibular lymph nodes and the upper trachea are also often involved.66’78’79 The infection is most commonly restricted to superficial epithelial tissue, which may include the corneal epithelium-’3'80 Since viral replication is usually restricted to areas of lower body temperature, viremia is rare. It has only been occasionally reported and the virus appears to be associated with peripheral blood leucocytes.79’81 Generalized disease is predominantly seen in debilitated animals or neonatal kittens.70‘82’83 As the virus replicates its cytopathic effect creates areas of multifocal epithelial necrosis leading to inflammation with neutrophilic infiltration and fibrin - 27,6l,6 ,7 .72,79 exudation. 6 0 Secondary bacterial infection often worsens the condition.70 Incubation periods can range from 2-10 days.27 The clinical signs usually resolve within 10-14 days.27 Detection of infectious virus is possible as early as 24 h post infection via oropharyngeal and nasal swabs. The virus generally persists for 1-3 weeks, although detection of viral DNA by PCR might be possible for longer.”85 In severe cases osteolytic changes within the turbinate can be caused by the virus.66 Following intravenous inoculation of young kittens a predilection of the virus for regions of skeletal growth, including the turbinates has been shown.86 Experimentally it also has been shown that intravaginal instillation of the virus in pregnant queens can lead to vaginitis and congenitally infected kittens.87 Intravenous inoculation of pregnant queens led to transplacental infection and abortion.88 1.6 Feline herpesvirus 1 disease — Other species Feline herpesvirus 1 has been described to possess a very narrow host range.89 However, FHV-l can exist within other non-domestic felids or even non-felidae as well. In 1971 Thomson et al.90 reported the isolation of a herpesvirus from three Cheetahs (Acinonyx jubatus) living at the Lion Park at Warragamba, New South Wales. Two of the cats developed conjunctivitis and rhinitis.90 The third cat showed no clinical symptoms and it was suggested that this animal may have been a carrier.90 Virus was isolated from the eyes, nose, and mouth, but not from the rectum. The isolates were identified a herpesvirus by their production of cytopathic effects, including giant cells and Cowdry type A inclusion bodies, and their appearance on electron microscopy.90 In 1984 a disease outbreak occurred at the St. Louis Zoological Park, affecting two of three queen-reared Cheetah cub siblings.91 The cubs had been vaccinated with a killed virus product at three-week intervals from ages 3.5 to 18 weeks.91 By the age of three months they developed chronic, multifocal, ulcerative cutaneous lesions, which were refractory to treatment.” Excisional biopsies suggested a diagnosis of eosinophilic granuloma, but intranuclear inclusions characteristic of herpesvirus were also found.g1 Herpesvirus was also found in these samples by transmission electron microscopy. Virus isolation revealed a virus that induced cytopathology similar to that of alphaherpesviruses. The presence of a herpesvirus was suggested, which was named cheetah herpesvirus (ChHV).91 In order to compare this virus with FHV-l, further tests (dot blot hybridization, restriction endonuclease digested DNA electrophoretic profiles, antigenic comparison by serum neutralization and western immunoblotting analysis, SDS polyacrylamide gel electrophoresis comparison of viral 9 polypeptides) revealed that the ChHV isolate was closely related to FHV -1.91 However, at least two detectable differences in the Kpn I digestion patterns were considered to be significant because of the remarkable stability of FHV-l and lack of restriction fragment length polymorphism.92 Whether or not ChHV is truly different from FHV-l remains to be investigated. In 1977 Boever et a].93 reported viral rhinotracheitis in 4 of 5 clouded leopards (Felis nebulosa) housed at the St. Louis Zoo. The leopards had clinical signs which consisted of serous nasal and ocular discharge and sneezing. Two of the cats also had ulcerative glossitis. One pregnant animal developed vaginitis and aborted. Nasal and rectal swabs were obtained for virus isolation from three severely affected animals that died due to progression of their illness. A cytopathogenic agent, which was identified as feline rhinotracheitis virus, was isolated from the nasal swab of one animal.93 Truyen et a]. in 199094 reported an acute outbreak of encephalomyelitis in 10 lions from a safari park in Germany. Three lions were euthanized and different diagnostic tests (histopathology, virus isolation, serum neutralization) were performed. Apart from mild swelling and reddening of the tonsils, alterations were mainly found in the brain. The pathological diagnosis was non-suppurative polioencephalomyelitis with demyelinization of the spinal cord. In the nuclei of some glial cells small intranuclear, acidophilic, round and ovoid structures suggestive of intranuclear inclusion bodies were found. Virus isolated from the tonsils of one animal demonstrated cytopathic effects. The cytopathic agent was serologically characterized as FHV -1. However, FHV-l was not isolated from any brain tissues. Ultimately it was concluded that FHV-l was unlikely to be the cause of the encephalitis outbreak.94 10 Surveys of antibody titers in free—ranging lions in Africa revealed a seroprevalence for FHV—l of 67% and 91% in Etosha National Park and Kruger National Park, respectively.”96 Antibodies to FHV -1 were found in 309/310 samples in lions living within Serengeti National Park and Ngorongoro Crater.97 In 1991, Eberle et a].98 evaluated sera from 19 free-ranging bobcats (Lynx rufus) to determine the seroprevalence of FHV-l via an enzyme linked immunosorbent assay (ELISA). All samples were negative suggesting that FHV-l was uncommon or not present at all in this species. Four free-ranging bobcats were captured, and ELISA for anti- FHV -1 antibodies and attempts at virus isolation from oropharyngeal swabs were negative. The cats were inoculated by instilling 200 u] of the viral suspension in each nostril. None of the animals developed clinical signs of disease. However, virus was isolated from the oropharynx and nostrils on days 3-4, 7- 8, and 14-15 post-inoculation. Although the bobcats did produce anti-FHV-l IgG, titers were generally lower than those seen in domestic cats.98 The experiment demonstrated that bobcats are susceptible to FHV-l infection, but clinical signs of disease do not accompany the infections. Paul-Murphy et al."9 demonstrated a seroprevalence of 19% for FHV-l in 58 mountain lions (F elis concolor) in California. In 1999, Pratelli et a].100 reported an outbreak of respiratory tract disease characterized by sneezing, nasal and ocular mucopurulent discharge, anorexia and prostration in three tigers and one leopard from the Fasano Zoo in southern Italy. Nasal swabs were obtained from all four animals for virus isolation. Characteristic cytopathic effects and intranuclear inclusions were observed. Virus neutralization tests using a goat antiserum, monospecific to FHV-l, identified the isolates as F HV -1 strains. Restriction patterns were compared with those of a vaccine] and a field strain of FHV-l. Restriction patterns obtained with PstI and BamHI revealed that the 11 genomes of the four isolates, the field strain, and the vaccination strain were identical. It could not be concluded if the disease was caused by a FHV-l strain reactivated in a wild felid or by a strain transmitted by domestic cats.100 Nakamura et al.101 investigated the seroprevalence of F HV-l in domestic cats (F elis catus) and leopard cats (F elis bengalensis) in Vietnam. All cats were considered to be unvaccinated. In the Hanoi area only 1.4% (1/69) of domestic cats were positive for FHV-l antibodies, compared to 44% (22/50) in the Ho Chi Minh City area. Of the tested leopard cats, 11.1% (1/9) were positive in the Hanoi area, whereas 50% (2/4) were positive in the Ho Chi Minh City area. The authors suggested that the higher cat population density in Ho Chi Minh City area explained in part the higher prevalence of HIV-1. Everrnann et al.102 isolated a herpesvirus from dogs with diarrhea. Virus neutralization tests revealed a relation of the isolated viruses to F HV-l. Rota et 31.103 compared five herpesvirus isolates from these dogs to the DNA and polypeptide patterns of FHV-l. The genomes of these five canine isolates showed restriction enzyme digestion patterns that were nearly identical to those of FHV -1, but completely different from CHV. The disease potential of these canine F HV-l-like isolates remained unclear in this study. 1.104 isolated FHV-l from the thymus and spleen of a 17 week old Kramer eta cross breed dog with a history of distemper-like illness, diabetes mellitus, and pancreatic atrophy. Following culture of the virus in Crandell feline kidney cells, three healthy puppies were inoculated intravenously and intranasally with the viral suspension. No clinical signs were produced. 12 In a study investigating spotted hyenas, only one of 240 animals had serum antibodies against FHV-l.105 This suggests that hyenas unlikely play a role as a reservoir for FHV-l in the Serengeti ecosystem. 1.7 Feline herpesvirus 1 disease - Systemic manifestations Feline herpesvirus-1 infection causes upper respiratory tract symptoms, ocular manifestations and reproductive failure.106 The primary, acute disease affects kittens more commonly and more severely. Following an incubation period of 24-48 hours, a sudden onset of sneezing, followed by profuse serous nasal and ocular discharge, hypersalivation, fever, anorexia and lethargy may be observed.”1’27’70’79’106 Due to secondary bacteria] invasion, the discharge may progress from serous to mucopurulent.106 In severe cases, the virus may invade the lungs leading to viral pneumonia and possibly secondary bacteria] pneumonia.70’82’83’107 Bordetella bronchiseptica has been identified as a frequent secondary invader.108 Primary FHV—l infection is self limiting in most cases, and symptoms regress within 10-14 days.70 Chronic sinusitis may follow severe primary infection and may result in necrosis and distortion of the turbinates.“66 In rare cases of FHV-l infection, oral ulceration has been observed. However, oral ulceration is more commonly associated with feline calicivirus (FCV) infection.69’106 As a rare sequela to FHV-l infection, neurological signs have been described.109 Abortion has been associated with FHV-l infection. However, experimental studies have suggested that abortion is most likely due to the severe systemic illness, rather than a direct effect of the virus itself.88 Hickman et a].110 investigated a natural outbreak of F HV-l infection in a specific pathogen free cat colony. No cases of abortion were seen, even in severely affected pregnant queens. 1 '0 l3 Cutaneous ulceration has been described with FHV—l infection in domestic cats and also in Cheetahsm'l '6 Histopathologic examination of the lesions reveals ”5’1'6 Typically a marked eosinophilic necrotizing and ulcerative changes. inflammatory response is noted within these lesions. However, the infiltrate may also be neutrophilic in some cases.1 1 H '6 Holland et al.117 showed that FHV -1 DNA can be detected in the skin of cats with herpetic dermatitis and suggested that the virus may play a causative role in the disease. Furthermore, a PCR assay for FHV -1 was shown to be useful in the diagnostic process with high sensitivity and specificity.”7 1.8 Feline herpesvirus 1 disease - Ocular manifestations 1.8.1 Conjunctivitis Feline herpesvirus-1 infection is the most common cause of conjunctivitis in cats and is often accompanied by respiratory disease.‘18 Bilateral conjunctivitis characterized by blepharospasm, chemosis, serous ocular discharge, and hyperemia is the most common ocular manifestation during acute FHV-l infection.4’2l”0’71"19‘122 Diffuse necrosis of the conjunctiva] epithelium and intranuclear inclusions within conjunctiva] epithelial cells are present by day four post-infection.4 Around day 8 post-infection, cytopathic effects of the virus cause the conjunctiva] epithelium to slough.4 The serous ocular discharge can progress over several days to mucopurulent discharge as polymorphonuclear cells as well as low numbers of macrophages infiltrate the conjunctiva] stroma.4’120 Bacteria] infections may worsen the mucopurulent discharge, and pseudodiphtheritic membrane formation may occur.1 '8 Most cats recover within 10-20 days with no other ocular sequelaef“120 However, more severe infection or immunosuppression may result in chronic or recurrent 14 conjunctivitis.123 F HV-l infection in adult cats typically causes conjunctiva] hyperemia, intermittent blepharospasm, and mild serous ocular discharge. '24 Conjunctiva] cytologic findings depend on the duration of the disease process and the presence or absence of other infectious agents. 1 18 The predominant cell type is the lymphocyte, although macrophages are also a common finding. l 18"” With chronic or secondary infection, more neutrophils and giant epithelial cells may be seen.1 18“” Epithelial intranuclear inclusion bodies may occur, but are rarely observed in clinical C3868.121 1.8.2 Keratitis Epithelial keratitis commonly occurs during primary infection in young cats, which resolves spontaneously in most cases.125 Feline herpesvirus-l induced corneal ulceration in adult cats is likely due to reactivation of the virus.l ”"20 This condition is usually accompanied by varying degree of conjunctivitis.70 Systemic immunosuppression, environmental stress, and systemic administration of corticosteroids have been associated with reactivation of latent virus.12°‘125 In the adult cat, herpetic keratitis can be unilateral or bilateral and is usually not associated with upper respiratory disease.”8’125 Corneal edema may precede the formation of comea] ulcers. 1 18 During the acute stage of ocular infection, the corneal lesions tend to be microdendritic.”9 Early corneal ulcers may also be dendritic. However, they often progress quickly to larger geographic ulcers.4’70 Dendritic shaped corneal ulcers are the classical pattern of corneal ulcerations induced by FHV-1.4’7°’”9’126"27 They result from the direct cytopathic effect of the virus”:5 .127 Sometimes, ulcers may also occur as numerous punctate lesions, that may coalesce to form a larger superficial ulcer.“8 Occasionally herpetic ulcerations progress, likely due to secondary bacterial infection, to involve deeper stromal layers or cause corneal perforation.7O Chronic ulcers may 15 incite a corneal vascular response, although the intensity varies greatly.70 FHV-l induced corneal ulcerations may resolve spontaneously, whereas others may develop . . 70 rnto indolent ulcers. 1.8.3 Stromal keratitis Herpetic stromal keratitis caused by HSV-l is the leading cause of infectious corneal blindness in the industrialized world. Changes associated with stromal keratitis are thought to be due to the inflammatory response to chronic or repeated recrudescent infection. Several observations support the conclusion that the pathogenesis of herpetic stromal keratitis is immune mediated, including the observation that this manifestation of ocular herpetic disease is uncommon in humans who are immunodeficient.128 Furthermore, large numbers of inflammatory cells infiltrate the cornea, including polymorphonuclear neutrophils, macrophages, Langerhans cells, natural killer cells, plasma cells and T cellsm"32 The importance of the immune response in stromal keratitis is also supported by observations that in T cell-deficient mice, ocular infection with HSV-l was not associated with the development of marked stromal disease. '33 The exact immunological mechanism of the development of stromal keratitis is not known. Originally the chronic changes believed to result from immune complex deposition because immunoglobulins and herpesvirus particles were found in the stromal” However, there is evidence, that a chronic low-level viral infection persists within stromal keratocytes providing a continuous stimulus to antigen-specific T lymphocytes that kill the keratocytes by a cytotoxic mechanism.I35 Furthermore, CD4 T cells reactive against HSV-l antigens have been isolated from chromic stromal keratitis lesionsm’137 The pathogenesis of corneal scarring and vascularization in stromal keratitis is still uncertainm’139 HSV-l infection can induce production of 16 different angiogenic factors such as vascular endothelial growth factor (VEGF), matrix metalloproteinases (MMP) 2 and 9, platelet-derived grth factor (PDGF), B- fibrosing growth factor (bFGF), macrophage inflammatory protein 2 (MIP-2) and monocyte chemotactic protein 1 (MCP—l).'4°’142 These factors are mainly produced by polymorphonuclear neutrophils. However, VEGF is also expressed in corneal epithelial and stromal cells. It appears to be produced by cells close to those infected by the virus.140 It is possible that interleukin 1 (IL-1) and interleukin 6 (IL-6), produced initially by infected cells, induced the production of VEGF. Stromal keratitis is a rare manifestation of corneal FHV -1 infection.“’80 However, due to its potential to cause vision threatening corneal scarring, it is considered the most significant manifestation of feline ocular herpetic disease.4 It is usually associated with chronic epithelial ulceration and characterized by stromal edema, deep vascularization and the presence of inflammatory cells.4 In experimentally induced ocular feline herpesvirus-1 infection, the virus was observed to preferentially infect conjunctiva] epithelium. However, FHV-l was able to replicate in corneal epithelium, and only minimal viral antigen was detected in the corneal stroma . 80 When cats were treated with subconjunctival administration of betamethasone followed by topical application of FHV-l to the scarified cornea, a stromal keratitis developed, characterized by geographic epithelial ulceration, interstitial edema, deep vascularization, and inflammatory cell infiltratesfl’80 To elaborate the mechanism through which stromal disease occurs, Nasisse et a].143 studied the histologic, immunologic, and virologic features of FHV-l induced keratitis in 18 experimentally infected cats. Stromal disease was induced by local immunosuppression via application of three consecutive daily subconjunctival injections of 4 mg betamethasone prior to inoculation with FHV-l .143 Prior to the 17 development of stromal keratitis, prolonged absence of the corneal epithelium, decreased numbers of circulating lymphocytes, decreased mitogen responses, and acquisition of viral antigen by the corneal stroma were noted.'43 The first cells to arrive in infected corneas were neutrophils. The subsequent arrival of B and T lymphocytes was supportive of a specific antiviral immune response.143 Fibrosis and scarring developed secondary to the stromal inflammation. The authors concluded, that it is probable that the corticosteroid-mediated suppression of the normal immune function allowed uncontrolled local virus replication, which combined with delayed corneal epithelial healing led to stromal accumulation of virus.143 The development of stromal keratitis was likely a result of the immune response to viral antigen.‘43 1.8.4 Keratoconjunctivitis sicca Decreased tear production in cats has long been suspected to be a potential sequelae to FHV-l infection.”9 Clinical signs can include conjunctiva] hyperemia, a dry appearance of the cornea, cornea] epithelial hyperplasia and corneal ulceration.120 The cause remains speculative, but it has been assumed that excretory duct occlusion or lacrimal adenitis is responsible.4’70’80 In most cats, keratoconjunctivitis sicca is transient, with tear production increasing with resolution of active FHV-l related disease.70 However, a decrease in tear production associated with FHV-l infection can also be permanent.“’70 Experimentally, a temporary keratoconjunctivitis sicca developed in 5 of 10 cats treated pre-inoculation with three consecutive daily subconjunctival injections of betamethasone.80 Aqueous tear production returned to normal in all cats by day 60 post-infection. Using specific fluorescent antibody test and virus isolation, Bistner, et a]. failed to demonstrate the presence of FHV-l within the lacrimal gland.121 However, using a PCR assay, Reubel et a], detected FHV-l DNA in two of five lacrimal glands 18 of latently infected cats, as defined by negative virus isolation from swabs of oral and ocular secretions. 144 Fluorescein labeled antibody has been used to examine human lacrimal glands during both acute and asymptomatic stages of HSV-1 corneal infection. Viral antigen was detected in 2/3 patients with acute disease and in 4/7 asymptomatic patients with a history of previous herpetic infection.145 A case of dacryoadenitis was described in an immunodeficient patient.‘46 A biopsy sample from the right lacrimal gland was positive on immunohistochemisty for HSV-l. However, virus isolation was negative for HSV-l, VZV, cytomegalovirus, and enterovirus, but positive for adenovirus. The patient was treated with systemic antiviral therapy (acyclovir). The patient recovered with the exception of mild bilateral dry eye symptoms. Duration of the dry eye could not be evaluated because the patient was lost to follow-up. Lim and Cullen147 evaluated Schirmer tear test values and tear film break up times in cats with conjunctivitis. Lower mean tear film break up times, but not lower Schirmer tear test values were found in cats that had FHV—l DNA detected by PCR in their blood compared to cats that did not. The authors concluded that tear film instability may play a role in the pathogenesis of feline conjunctivitis, although it is not clear if it is a predisposing factor or a result of it. In a recent study the effect of primary FHV-l infection on tear film break up time and Schirmer tear test values, as well as the relationship between these parameters and conjunctival goblet cell density, histological inflammation, and clinical disease severity was evaluated.148 Tear film break up time and goblet cell density decreased dramatically during the first week following viral inoculation, continued to decline until 15 and 21 days post-inoculation and remained abnormal for the duration of the study (29 days)!“ The total disease score and clinical and 19 histological evidence of conjunctivitis increased in all 6 cats during the first week, but began to normalize thereafter. Schirmer tear test values remained elevated for 29 days following inoculation, likely due to the continued tear-film instability. ”8 1.8.5 Ophthalmia neonatorum and symblepharon Ophthalmia neonatorum as a presentation of early FHV-l infections was Lm Two to four week old kittens with ocular lesions initially described by Bistner et a had generalized disease as well characterized by rhinitis, tracheitis, bronchopneumonia, and hepatic necrosism Infection with FHV-l before the eyelids are open can result in mucopurulent conjunctivitis which causes distension of the eyelids. ‘20 Symblepharon formation, the adhesion of conjunctiva to itself or to the cornea, may be observed as a sequela to FHV-l infection.“’120 The profound epithelial necrosis caused by FHV-l precedes this condition, leading to adhesions and scar - 4.19.120 formation. 1 1.8.6 Corneal sequestrum Feline corneal sequestrum (FCS) is a disease unique to the catm’lso characterized by degeneration of collagen and accumulation of brown pigment.“8"51’152 The pigment appears to be water soluble and not melanin or hemosiderin.”(’)’151’153 However, Featherstone et al.”4 found particles within corneal sequestra which were consistent with the appearance of melanin particles. The clinical appearance is characteristic. Lesions are variable in pigmentation, round to oval shaped and usually located centrally or paracentrally.'49’155 Corneal vascularization, 118,119,156 perilesiona] stromal edema, and inflammation are common in chronic disease. Clinical signs include blepharospasm, epiphora, enophthalrnos, elevation of the 20 nictitans, chemosis and hyperemia of the conjunctiva as well as photophobia and 149,150,157,158 The decreased activity. condition can occur unilaterally or 149,150,158 118,119,150,158,159 118.158 bilaterally. Predispositions for Persian, Himalayan, 160 158 118,150,158 Colorpoint, American Shorthair and Siamese cats are reported. Histopathologically the sequestrum is characterized by stromal degeneration, cell loss and dessication, often surrounded by a zone of inflammatory cells.”8’151'153’156’161 The etiology of FCS is unknown. Feline herpesvirus-1 DNA has been detected in 18-55. 1% of keratectomy specimens from cats with corneal sequestrum using polymerase chain reaction (PCR).'62"63 Using immunofluorescent antibody tests (IF A), FHV-l was detected in 22.9% of corneal sequestra examined.‘58 1.8.7 Eosinophilic keratitis Infection with F HV-l has also been implicated in the development of eosinophilic keratitis. "’4 Clinically, the patient presents with a pink to white, irregular, vascularized lesion in the peripheral limba] area that may invade the superficial cornea, conjunctiva, and nictitating membrane and be present unilaterally or bilaterally.“3’”9’I65 Cytologic examination usually demonstrates the presence of eosinophils, mast cells, lymphocytes, or plasma cellsm’166 Histopathological findings are chronic granulomatous inflammation characterized by infiltration of plasma cells, lymphocytes, numerous eosinophilic granulocytes and occasionally mast cells. 1 19’1“ Using IFA, corneal scraping specimens from cats affected with eosinophilic keratitis were positive for FHV-l in 9 of 27 cases (33.3%).164 Nasisse at al.163 detected F HV-l DNA using PCR assay in 76.3% of corneal scrapings from cats with eosinophilic keratitis, but only in 5.9% of those from clinically normal cats. The higher detection rate in the second study is explained by the higher sensitivity of PCR 21 compared to IF A. The role of FHV-l in the development of eosinophilic keratitis has to be further elucidated. '20 1.8.8 Anterior uveitis Herpes infections that involve the anterior uvea have been described in humans and are generally not associated with simultaneous or past keratitis.167 Utilizing PCR and hybridization techniques, Bustos et al. detected HSV-l DNA in 20/30 (66.6%) human ciliary ganglia from patients with no known active HSV-l ‘68 This suggests that the ciliary ganglion may be an infection at the time of death. additional site of HSV-1 latency in humans.168 Latent HSV-l was also found in ciliary ganglia from experimentally infected mice.31 This suggests that anterior uveitis may result from virus reactivation within the ciliary ganglia rather than within the 1.169 concluded from another study, that trigeminal ganglion. However, Labetoulle eta infection of the iris and the ciliary body results from transfer of virus through the sympathetic system via the superior cervical ganglion. If additional sites of latency within the autonomic system exist in cats, a possible role of FHV-l in feline anterior uveitis could be explained. Maggs et a].170 utilized a PCR assay to detect FHV-l DNA in the aqueous humor of 11 of 73 cats (14%) with idiopathic anterior uveitis or uveitis due to T oxoplasma gondii. Intraocular production of FHV-l antibodies was noted in 22/44 cats with idiopathic uveitis and 11/29 cats with toxoplasma uveitis, whereas intraocular production of FHV-l antibodies was never detected in cats without uveitis.170 In a study by Powell et a].,171 four specific pathogen free kittens were inoculated with FHV-l by use of an atomizer administering V2 of the volume into the oropharynx and ‘A of the volume into each nostril. Two of the cats developed mild anterior uveitis 11 and 25 days post-infection respectively that resolved within one 22 week of appearance. No posterior segment lesions were detected.l7l These findings suggests that F HV-l may play a role in feline anterior uveitis. 1.9 Feline herpesvirus 1 disease - Diagnosis The characteristic clinical signs and the self-limiting nature of most acute phase FHV-l infections often make obtaining a definitive diagnosis less critical.75’120 Application of Rose Bengal stain, a vital dye that stains dead and degenerating cells, to the ocular surface can be helpful to identify early dendritic lesions prior to the disruption of the corneal epitheliumno’lm‘I72 Application of sodium fluorescein to the ocular surface will identify corneal epithelial disruption.120 Conjunctiva] cytology may be performed in acute, primary F HV-l infections particularly for the presence of intranuclear inclusion bodies.”8 May-Gruenwald-Giemsa stain has been recommended for identifying intranuclear inclusions.121 Conjunctiva] cytology is rarely diagnostic because the predominant cell type observed is the neutrophil and intranuclear inclusions are infrequent.126 The diversity of clinical signs in chronic FHV-l disease make a definitive diagnosis with viral identification more desirable/5’120 Tests that are currently available either rely on detection of the organism or demonstration of an immunologic response to the organism. The methodologies available include virus isolation (VI), detection of virus neutralizing serum antibodies (SN), enzyme-linked immunosorbent assay (ELISA), immunofluorescent antibody testing (IFA), or polymerase chain reaction assays (PCR).75 1.9.1 Virus isolation In 1958, Crandell and Maurer21 isolated a feline virus associated with intranuclear inclusion bodies, which was designated as the C-27 virus. In 1959, the 23 cytopathogenic features of feline rhinotracheitis virus in tissue cultures of feline renal cells were described as: development of intranuclear inclusion bodies, formation of giant cells, and a slight reduction in the metabolic rate of infected cells.28 Virus isolation (VI) has been the gold standard for the diagnosis of alphaherpesviruses.75 Due to F HV-l ’5 rapid replication and production of characteristic cytopathic effects in cell culture, V1 is relatively simple to perform provided viable virus was collected and transported to the laboratory.75 Dacron or cotton swabs are recommended to collect the samples as alginate swabs can be inhibitory to some herpesviruses.173 After collection of the sample, it should be refrigerated and shipped promptly.75 1.9.2 Serology Regardless of methodology, serologic titers are not considered useful in the diagnosis of FHV-l infection because of the high rate of vaccination against F HV-l with resultant systemic antibody titers.70’75’123 Therefore titers are unlikely beneficial, because serology does not differentiate between immune response to vaccination or natural infection.75 In one study, no correlation between F HV -1 seropositivity or titer magnitude and the likelihood of detecting virus or presence of clinical signs could be demonstrated.64 The serum neutralization (SN) test is commonly used to detect F HV-l infection in catsm’m"76 However, FHV-l is difficult to detect with this method, because of the poor irnmunogenicity of the virus.177 The presence of antibody in serum suggests prior infection but does not necessarily correlate with clinical disease.178 On the other hand FHV -1 has been documented in ocular tissues of many cats negative for antibody by serum neutralization test. '79 Enzyme-linked immunosorbent assays (ELISA) for detection of feline herpesvirus-1 antibody have been developedm’178 These tests are more sensitive than 24 serum neutralization.178 However in one study, seroprevalence, as assessed by ELISA did not vary significantly between normal cats and cats suspected to be undergoing acute or chronic FHV-l infections, nor did it correlate with the likelihood of viral detection utilizing IFA or VI.64 1.9.3 Fluorescent antibody test Fluorescent antibody tests can be performed as a direct test (polyclonal fluorescein conjugated antibody) or an indirect test (polyclonal unlabelled antibody, which then reacts with a conjugated antibody). The antibody then detects FHV-l epitopes on the cell surface.75 However, the indirect test is more sensitive than the 180 direct test. Samples are usually obtained by conjunctiva] or corneal scraping or by biopsy.75"2° It is necessary to collect the sample prior to topical application of fluorescein dye, as this will interfere with the interpretation of the test result.181 Carlson and Scott found the test specific and sensitive for diagnosing cats showing clinical sings of FHV—l infection.182 In contrast, other studies found different limitations of the fluorescent antibody test, such as lack of significant differences in detection rates between acutely or chronically affected and normal animalsf’4 and the subjective nature of the laboratory interpretation.“126 Particularly in chronic infections, it is possible that viral epitopes may be bound to host antibody, which results in a false negative test.126 Burgesser et a].183 determined the sensitivity and specificity for indirect immunofluorescence to be 29% and 96%, respectively, compared to PCR. These limitations and the availability of other diagnostic methods, make fluorescent antibody tests appear to be of limited clinical application.75 25 1.9.4 PCR assays Us“ and The polymerase chain reaction was introduced in 1987 by Mullis et a dramatically changed the field of molecular biology as well as disease diagnosis in man and animals. To obtain a sample for FHV-l detection, host cells need to be sampled, since the virus is an obligate intracellular agent.75 Biopsy samples are recommended.75 However, in other studies, FHV-l DNA was obtained by the use of Schirmer tear test strips or microbrush swabs.34’185 Laboratories often suggest the samples are placed in a small volume of a physiological fluid such as phosphate buffered saline, frozen, and shipped overnight on ice for analysis.” However, Maggs et a].186 assessed the effect of shipping temperature on results of one PCR assay for detection of FHV-l DNA. PCR results did not differ for identical samples sent overnight on ice or mailed over several days at ambient daytime maximum temperatures between 17 an 47 °C.'86 Multiple PCR assays, some of which are commercially available, have been described for use in the detection of FHV-l DNA.85‘144’183"87"89 PCR analysis for F HV-l is commonly based on DNA-amplification of the thymidine kinase gene.126’l44 PCR and nested PCR are highly sensitive for the detection of active or latent FHV-l - - 2 .144, , . . . infections.I 6 162 183 These PCR techmques have shown to be more sensrtrve than the use of immunofluorescent antibody testing or virus isolation, but detection limits ,l44,l , 7- 9 vary greatly.“ 8318 I8 Nested PCR utilizes a second primer pair internal to the initial primer. Stiles et a].188 demonstrated a nested PCR assay to be more sensitive for identification of F HV- ] in cats with conjunctivitis than virus isolation or fluorescent antibody tests. In another study comparing virus isolation, single PCR, and nested PCR for the detection of FHV-l in ocular and nasal secretions, nested PCR was shown to be 4.8 times more 26 '89 When 22 samples that were negative for virus isolation sensitive than single PCR. were assayed, 19/22 (86.4%) were positive by nested PCR and 2/22 (9%) were positive by single PCR.189 Real time PCR not only allows detection of, but also the quantification of a given DNA sequence.190 Accumulation of PCR product is measured through a labelled fluorogenic probe.190 Advantages of real time PCR over conventional PCR include simultaneous quantification of input target DNA within the PCR process, no necessity for post-amplification steps, and therefore reduced chance for cross contamination.190 The procedure is fast and highly efficient.190 A real time PCR assay was developed for the quantification of FHV-l DNA in ocular fluid samples of clinically diseased cats.84 A good correlation between viral titer and quantitative PCR was observed during early infection.84 In the second stage of disease, the viral titers decreased, while the PCR signal remained high, and in the final stage of the disease, virus isolation was negative and the PCR signal decreased consistently.84 The authors concluded that comparing the results of virus isolation and quantitative PCR in consecutive samples would allow one to track the course of the infection.84 Low et al.‘91 assessed the prevalence of FHV-l DNA in cats with and without conjunctivitis using conventional PCR and quantitative PCR. The concordance between the two assays for detection of FHV-l DNA was 92.5%. '91 No significant difference for FHV- l prevalence was found between cats with conjunctivitis and cats without conjunctivitis. '91 Another study compared six published PCR assays for detection of FHV-l DNA in 15 clinical samples and five vaccines licensed for use in preventing FHV-l associated disease.192 Detection rates for the clinical samples varied between 29% and 86%.192 All six PCR assays detected FHV-l DNA in all five vaccines tested.192 27 Therefore, a positive PCR results cannot differentiate a vaccinal strain from natural strains and does not differentiate sub-clinically infected cats from those with clinical disease as a result of FHV-l infection.“192 1.9.5 In situ Hybridization In order to obtain information regarding latent F HV-l infection, Ohmura et al.193 utilized in situ hybridization to detect FHV-l LATs in the trigeminal ganglia of 1.194 performed in situ hybridization with a experimentally infected cats. Suchy et a digoxigenin-labeled double-stranded DNA probe for FHV-l on different tissues from a severely affected cat. Hybridization signal was found only in nuclei of intact epithelial cells in the basal layers of the epidermis, hair follicles of the eyelids, nose, superior lip and paw, as well as in conj unctival epithelium of palpebrae and nictitating membrane and in a few superficial cells of both corneas.I94 Kim and Yi'95 optimized an in situ hybridization protocol for the detection of F HV -1 DNA by using microwave irradiation as pre-treatment. Although this technique may be of limited value in diagnosis of clinical cases, it offers great potential in studying FHV-l infection of different tissues as in situ hybridization allows for specific localization of the virus and evaluation of tissue morphology simultaneously. Sensitivity of in situ hybridization largely depends on many variables in the utilized protocol including fixation of the tissue, exposure of target nucleic acid, selection of probe, type of probe labelling, denaturation of doublestranded DNA, hybridization conditions, and detection conditions. 196 1.9.6 FHV-l detection in different tissues Despite the high incidence of F HV-l infection within the feline population and many investigations into its pathology, much remains unknown. For example, FHV-l 28 latency is stated to occur only in the trigeminal ganglia.2 During the latent period, clinical signs are absent and the virus cannot be isolated using standard cell culture techniquesfm‘197 However, several studies have detected FHV-l DNA by PCR in 13%188 to 31%183 of conjunctival and 5.9%,163 46%, '62 and 49%198 of comeal samples from clinically normal cats. Despite the PCR positive results of conjunctival samples, 183 1188 FHV-l was detected either infrequently or not at al by virus isolation (VI) or fluorescent antibody (FA) staining. In these samples, F HV -1 was not quantified, nor was the active or latent viral status or specific viral location within each tissue identified. It is known that FHV-l demonstrates a tropism for conjunctival epithelium and also replicates within corneal epithelium.”80 In one study, immunohistochemical staining of corneal and conjunctival epithelial cells revealed the diffuse presence of viral antigen during the acute stage of infection.80 In cats with experimentally induced stromal keratitis, viral antigen persisted within the corneal stroma between collagen lamellae.I43 However these cats were immunosuppressed by three consecutive daily subconjunctival injections of betamethasone prior to inoculation with the virus. In clinically normal cats with PCR detectable virus, whether the virus persists in epithelial cells, becomes localized in the underlying stroma, or has any effect on the surrounding tissues is not known. Viral location is critical, as acquisition of viral antigen by the corneal stroma is the factor currently thought to precede development of stromal keratitis.143 The ability of the virus to infect other ocular tissues has not been widely explored. Studies to identify sites of F HV-l latency have concentrated particularly on the trigeminal ganglion as this is an important site of viral latency for many other herpesviruses.l97’199 Gaskell et al.‘99 isolated FHV-l from the trigeminal ganglia of 29 3/ 17 cats latently infected with field virus using a tissue fragment culture technique. F HV-l could not be detected in four cats previously infected with a vaccine strain of FHV-l or in four cats which were administered the vaccine and 48 hours later were inoculated with field virus. 199 The authors suggest that the vaccine may prevent latent infection. Nasisse et a1.200 isolated FHV-l in trigeminal ganglia from 5/26 latently infected cats using a similar tissue culture method. FHV-l was also isolated from the trigeminal ganglia from 4/4 cats with active infection.200 Reubel et al.144 developed and utilized a PCR assay to detect F HV-l in various tissues in latently infected cats. Latency was defined by concurrent negative virus isolation for those tissues. FHV-l was detected by PCR in optic nerves (6/6), trigeminal ganglia (8/9), olfactory bulbs (7/9), nasal turbinates (7/9), corneas (6/6), lacrimal glands (2/5), salivary glands (1/9), oral fauces (3/7), tonsils (1/9), cerebrum (1/9), conjunctiva (1/9), and mesenteric lymph node (1/9). No virus was detected by PCR in cerebral cortex, thymus, trachea, lung, liver, spleen, kidney, or peripheral blood monocular cells. However, 5/9 cats received intravenous methylprednisolone acetate 10 days prior to necropsy. In all five cats virus could be isolated from oral swabs obtained at the time of necropsy. In these cases, it is likely that a recrudescent infection was present at the time of euthanasia. In 4/4 cats viral DNA was detected utilizing PCR from oral swabs. Vaginal swabs were negative in all 5 cats. In a similar study conducted by Weigler et al.85 various tissues from 10 cats latently infected with FHV-l were evaluated for the presence of FHV-l by PCR. Latency was confirmed by concurrent negative virus isolation. All 20 trigeminal ganglia, 15/20 optic nerves, and 6/20 corneas were positive for F HV -1 DNA on PCR. 30 FHV -1 was detected in 4/10 optic chiasma and 2/9 olfactory bulbs. Virus was not detected within the uvea. Detection of latency associated transcripts (LATs) is probably the most accurate way to truly define the latent stage of infection.”197 With the detection of LATs it is also possible to distinguish true latency from low-level persistent infection.“ Using in situ hybridization and northern blot analysis, Ohmura et a1.193 detected transcripts of opposite strand to the immediate-early mRNA in the trigeminal ganglia of 3/5 experimentally infected cats. In another study,198 FHV-l DNA was detected by PCR in 45/92 (48.9%) corneas and in 38/92 (41.3%) trigeminal ganglia of cats without clinical signs of FHV -1 infection. The investigators also developed a reverse transcriptase PCR (RT— PCR) to detect FHV -1 LATs. None of the 39 tested corneas and four of the 16 tested trigeminal ganglia had positive results for F HV-l LATs.198 These studies to identify FHV -1 sites of latency suggest that FHV-l follows a pattern similar to HSV-l during latency. However, studies to evaluate whether F HV-l infects and establishes latency in autonomic ganglia have not yet been undertaken. 1.10 Feline herpesvirus 1 disease - Treatment and prevention Upper respiratory tract infection during the acute stage of the disease is in most cases self-limiting. Systemic broad-spectrum antibiotics are indicated to prevent or treat secondary bacterial infections.66’70’71 Supportive care is also very important.120 It is necessary to keep oro-nasal pathways clear. Frequent removal of nasal and ocular discharge is recommended.7l"06’202 Intravenous or subcutaneous fluid administration 66,71,202 can become necessary to achieve rehydration. Nutritional support is indicated in cases of severe anorexia.“202 31 Corneal ulceration should be treated with topical broad spectrum antibiotics to prevent secondary bacterial infections, and topical antiviral therapy should be considered.6‘r”70’l '8’203 In kittens with primary FHV-l infection with corneal ulceration or early symblepharon formation, topical antiviral therapy can be beneficial.(’6’70’203 In these cases frequent mechanical break-down of the symblepharon is also recommendedm’203 In older cats with recurring bouts of FHV—l conjunctivitis, therapy depends on the severity, duration, and frequency of the outbreaks.7°’203 Mild transient conjunctivitis does not require specific treatment, as it is usually self- limitingm‘120 If the conjunctivitis does not resolve or a corneal ulcer is present, topical antiviral therapy, as discussed below, is indicatedm’l 18’125 Mechanical debridement of the corneal ulcer can be helpfulm’203 to enhance the healing by removal of any loose epithelium, as well as removal of viral particles. Specific antiviral and adjunctive therapy, such as oral supplementation with the amino acid L-lysine, or application of interferons are described below in detail. Topical application of corticosteroids is the recommended initial treatment in cases of eosinophilic keratitis;1 '8'125 however, in cats where an association with F HV- 1 is suspected, other treatment options should be considered insteadm’204 Oral administration of megestrol acetate, 5 mg/day during the first week and 2.5-5 mg/week for maintenance dosage as treatment of eosinophilic keratitis has been recommended. 1 '9’204 Due to the potential of serious systemic side effects, such as temperament changes, polyphagia, diabetes mellitus, iatrogenic Addison’s syndrome, endometritis, cystic endometrial hyperplasia, mammary hypertrophy and neoplasia, megestrol acetate is not recommended as the initial choice of therapy.”"”205 Topical 204,206 application of cyclosporine can be used alternatively. In a recent study by Spiess et a1.,206 33 cats were treated with topical cyclosporine 1.5% two or three times daily. 32 83% of these cats showed marked improvement and 6% showed mild improvement of the disease. While, three animals showed no response, the likely cause was thought to be poor owner compliance. Two animals developed blepharitis, which resolved after treatment. 1.1 1 Antiviral therapy A multitude of antiviral drugs are licensed for treatment of viral diseases in humans. As a result of the many similarities between HSV-l and FHV-l, several of the drugs are employed in the treatment of herpetic disease in cats. However, differences in the biologic behaviour of these viruses support the need for continued research into and development of antiviral drugs specifically for use against F HV-l in cats. Frequent administration of the antiviral compounds is crucial in order for therapy to be effective.207 This is often a problem for cat owners, as many of the available topical antivirals are topically irritating.120 The presently available antiviral drugs are also virustatic. Therefore the drugs are effective only during the replicative phase of infection and latent FHV-l remains unaffected."207’208 Precise targeting of viral specific replicative processes is difficult as the virus utilizes host cell machinery 207 For many antiviral agents, the mechanism of action does not for replication. adequately differentiate infected from non-infected cells, which can lead to host toxicity.207 1.11.1 Nucleoside analogues The structure of these medications is similar to the nucleosides required for viral and host DNA and RNA synthesis. However, due to slight modifications the nucleoside analogues are able to disrupt the viral replication cycle.207 Following an 33 intracellular phosphorylation, the nucleoside analogues are incorporated into newly synthesized mammalian and viral DNA strands.1 DNA synthesis is then disrupted by mechanisms that are similar for all nucleoside analogues and are described in further detail below. 1.11.2 Acyclovir Acyclovir (9-(2-hydroxyethoxymethyl)guanine (acycloguanosine)) is similar to guanosine, but it contains an acyclic sugar group.l Acyclovir is a specific, non- toxic drug, that is highly efficacious against herpes simplex virus and, to some extent, varicella zoster virus.l Activation of this pro-drug requires three kinases to be present in the cell to convert acyclovir to a triphosphate derivate, the actual antiviral drug.1 The first step, phosphorylation of acyclovir into the monophosphate, is catalyzed by a herpesvirus-specific thymidine kinase.209'2” Cellular kinases perform the two remaining phosphorylation steps to produce acyclovir triphosphate."207 Acyclovir triphosphate has a greater affinity for the viral DNA polymerase than the naturally occurring deoxyguanine triphosphate, and is therefore preferentially incorporated into growing DNA via a competitive mechanism."207 The primary activities of acyclovir are related to its ability to decrease viral replication by chain termination of replication DNA by inactivating viral DNA polymerase.207 As acyclovir lacks the 3'- hydroxyl group present in the cyclic sugar ring of naturally occurring nucleotides, the growing DNA chain terminates, and further DNA replication is blocked."207 When acyclovir triphosphate is bound to the DNA template in the presence of the next nucleotide to be attached, the viral DNA polymerase is inactivated by the formation of an irreversible complexm’213 Further DNA replication is shut down by these mechanisms; therefore, no further production of infectious virions is possible.207 34 Acyclovir is highly specific for cells infected with herpes simplex virus or varicella zoster virus.1 The specificity depends on the virus-encoded kinase.l Differences or mutations in the viral thymidine kinase prevent the initial step of monophosphorylation leading to antiviral resistance.207 Since the development of acyclovir in 1974, many variations of chain-terminating nucleoside analogues have been developed in order to increase efficacy and bioavailability.1 Information regarding efficacy of acyclovir against FHV—l is described below. 1.11.3 Valacyclovir Valacyclovir (L-valine, 2-[(2-amino-l ,6-dihydro-6-oxo-9H-purin-9- yl)methoxy]ethyl ester, monohydrochloride) is the hydrochloride salt of the L-valyl 1,207,214 ester derivate of acyclovir. Following oral administration, valacyclovir is nearly completely converted by a liver hydrolase to acyclovir and L-valine during 25'2” It is therefore considered to be a pro-pro-drug.l intestinal or hepatic metabolism. The mean absolute oral bioavailability of acyclovir after administration of valacyclovir is four-fold higher (54.4 i 9.1%)214’2'8 than the limited oral bioavailability of acyclovir (15 to 21%).219 The bioavailability of valacyclovir in cats is 2.3-fold greater than that of acyclovir;220 however, severe systemic side effects such as bone marrow suppression, hepatic necrosis, and renal tubule epithelial necrosis have been documented.221 Due to its extremely toxic properties, valacyclovir is considered unsuitable for use in cats.221 1.11.4 Ganciclovir Ganciclovir (9-(1,3-dihydroxy-2-propoxymethyl)guanine) is an acyclic guanosine analogue, which is structurally closely related to acyclovir.2°7'222 Following phosphorylation to its triphosphorylated form, ganciclovir inactivates viral DNA 35 polymerase by competing with deoxyguanosine triphosphate for incorporation into the replicating DNA.207 Its incorporation into viral DNA-strands also results in 223 The toxicity of ganciclovir is greater than that of termination of elongation. acyclovir in vivo and in vitro. It can cause severe neutropenia in humans.207 Similar to acyclovir, the oral bioavailaility is limited. Therefore a valine ester of ganciclovir (valganciclovir) has been synthesized.222 Like valacyclovir, valganciclovir will be converted to release the parent compound, ganciclovir.222 1.11.5 Penciclovir Penciclovir (9-(4-hydroxy-3 -hydroxymethyl-but-1-yl)guanine) was first 2.224 The activity of penciclovir against several herpesviruses was synthesized in 197 first evaluated in 1987.225 It is an acyclic guanosine nucleoside analogue with a structure similar to acyclovir.207’222’225 Like acyclovir and ganciclovir, penciclovir must be initially phosphorylated by a viral thymidine kinase, followed by two more phosphorylation steps to reach its active form, penciclovir-triphosphate.207’222 This active form has been shown to inhibit herpesvirus DNA polymerase. However the possibility exists for internal incorporation of penciclovir residues into viral DNA and further chain extension.226 The first phosphorylation step of penciclovir to penciclovir-monophosphate is more efficient than the phosphorylation of acyclovir. However, the penciclovir-triphosphate formed in infected cells is a less effective inhibitor of herpes simplex virus DNA polymerase than acyclovir-triphosphate.227 Eamshaw et al.227 showed that for HSV-l DNA polymerase, the K for penciclovir triphosphate is about lOO-fold greater than that for acyclovir triphosphate. Therefore, a greater drug concentration is needed to inhibit viral DNA polymerase and hence inhibit the virus. Although penciclovir showed a similar spectrum of antiviral activity and selectivity when compared to acyclovir, the oral bioavailability was even lower 36 than that of acyclovir.222 Attempts at improving bioavailability led to the development . . 2 of famcrclovrr.22 1.11.6 Famciclovir F amciclovir (Diacetyl-6-deoxy-9-(4-hydroxy-3 -hydroxymethyl-but—1 - yl)guanine) is the diacetyl derivate of penciclovir.228 Following oral administration of famciclovir, it is efficiently converted to penciclovir.229 The absolute bioavailability of penciclovir from famciclovir following a single oral dose is 77%. The final products are penciclovir and the harmless by-product, acetic acid.229 Administered orally, famciclovir is highly effective against HSV-l in a variety of different animal models.230 In a quantitative study on the prevention of HSV-1 latency in mice, significantly less latent virus was detected in the ganglia of mice treated with 231 famciclovir as compared to those treated with valacyclovir. Oral administration of famciclovir appears to be well tolerated in cats.232 1.11.7 Idoxuridine Idoxuridine (IDU, 5-iodo-2'-deoxyuridine) was the first available antiviral agent, initially synthesized in 1959.233 It is still used in veterinary medicine. In human medicine it has largely been replaced by newer agents, such as acyclovir.207 Idoxuridine is a thymidine analogue, which does not require a virally encoded thymidine kinase for phosphorylation."211 Since it can be phosphorylated by both 1’2” Idoxuridine has a cellular and viral kinases, it has less specificity than acyclovir. direct inhibitory effect on virus-specific DNA polymerase causing suppression of viral DNA synthesis.211 Idoxuridine also causes disruptive replicative events as incorporation of the thymidine analogue into newly synthesized viral DNA produces abnormal viral proteins?” When administered systemically, it combines with cellular 37 DNA and can cause severe systemic side effects, including neoplastic changes, 2” Ophthalmic preparations are often compounded as genetic mutations, or infertility. 0.5% ointment or 0.1% solution,234 which are well tolerated and reasonably effective in the treatment of FHV-l disease in cats.207 1.11.8 Trifluridine Heidelberger et al. first synthesized 5-trifluoromethyl-Z-deocyuridine (TFT) in 1964.235 Like idoxuridine, trifluridine is structurally similar to deoxythymidine, a pyrimidine nucleoside used in DNA synthesis.207 It acts by incorporation into viral DNA, followed by inhibition of transcription of late virus-specific DNA?” As with idoxuridine, trifluridine is also independent of viral thymidine kinase for phosphorylation because it can be phosphorylated by cellular or viral kinases to its active form.233 Trifluridine is commercially available as a 1% ophthalmic solution.234 Clinically in cats, it is less well tolerated than idoxuridine or Vidarabinem'234 1.11.9 Vidarabine Vidarabine (9-Beta-d-rabinofuranosyladenine (Ara-A, adenine arabinoside)) was first synthesized in 1960 as a potential anti-neoplastic agent.2'l In 1964, its antiviral properties were reported.236 Vidarabine is a nucleoside analogue of adenosine, which does not require viral thymidine kinase for phosphorylation and 237 It selectively inhibits the replication of DNA viruses and some RNA activation. viruseszm’z” Following the three successive phosphorylation steps performed by host cellular enzymes, it is a competitive inhibitor of cellular and viral DNA polymerases, but has more potency against viral enzymes.21 "237 Systemic use is limited by its toxic neurologic and gastrointestinal side effects.”1 Vidarabine is available as a 3% 38 ophthalmic ointment with a recommended application frequency of every three 234 hours. Clinically, it is reasonably tolerated by cats.207 1.1 1.10 Cidofovir Cidofovir ((S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine) was first described in 1987 as a broad-spectrum anti-DNA viral agent active against herpesviruses (HSV-1, HSV-2, VZV, CMV), adeno- and poxviruses.238 Cidofovir does not require viral thymidine kinase for phosphorylation as cellular host enzymes perform the two phosphorylation steps required to reach its active form.207 Nephrotoxicity of cidovovir when given orally necessitates limitation of the dosage in humans.222 l-[((S)-2-Hydroxy-2-Oxo-l ,4,2-Dioxaphosphorinan-5-yl)Methyl]cytosine is the chemically stable pro-drug of cidofovir, which has reduced nephrotoxicity as compared to cidofovir in rats239 and in humans.240 Within the cells, the pro-drug is converted to cidofovir by an intracellular cyclic CMP phosphodiesterase.24' To increase the limited oral bioavailability, alkoxyalkyl esters have been prepared.242 Cidofovir was able to reduce clinical disease and viral shedding in cattle with bovine herpesvirus type 1 infection.243 Topical administration of cidofovir was highly effective in rabbit models in the treatment of experimental herpetic keratitis?“247 1.1 1 .1 1 Bromovinyldeoxyuridine Bromovinyldeoxyuridine, BVDU [(E)-5-(2-bromovinyl-2'-deoxyuridine)] is a pyrimidine analogue of the nucleoside thymidine.207 For phosphorylation it requires viral-encoded thymidine kinase like acyclovir.207 F HV -1 thymidine kinase does not effectively phosphorylate this agent, making BVDU relatively ineffective against FHV—l .207 Because of the low potency, BVDU is not used in veterinary medicine.207 39 1.11.12 Ribavirin 1-Beta-d-ribofuranosyl-l,2,4-triazole—3ocarboxamide, (ribavirin or virazole) is a triazole purine analogue, which inhibits a broad spectrum of RNA and DNA viruses.“ Following phosphorylation to its 5'-monophosphate form, it directly inhibits the synthesis of guanine nucleosides. Ribavirin triphosphate also blocks ATP and GTP binding to RNA polymerase.248 Systemic administration may lead to a number of side effects, such as extravascular hemolysis, bone marrow suppression, gastrointestinal toxicity, and central nervous signsm’248 These side effect make it unsuitable for clinical use. 1.12 Efficacy of nucleoside analogues against FHV-l 1.249 evaluated the in vitro susceptibility of feline herpesvirus-1 to Nasisse et a various antiviral drugs and found the relative potency of these compounds was trifluridine >> idoxuridine > Vidarabine > bromovinyldeoxyuridine >> acyclovir. Maggs and Clarke250 conducted a study in order to establish the in vitro efficacy against FHV-l of ganciclovir, cidofovir, penciclovir and foscamet. Furthermore the results were compared to the in vitro efficacy of acyclovir and idoxuridine. The relative potency of these compounds was idoxuridine = ganciclovir >> cidofovir > penciclovir >> acyclovir >> foscamet. The authors concluded that clinical trials with ganciclovir, cidofovir and penciclovir are warranted. Van der Meulen et al.25 I compared the in vitro efficacy of different antiviral drugs against FHV-l. Ganciclovir, 9-(2-phosphonylmethoxyethyl)-2,6-diaminopurine (PMEDAP) and cidofovir were most potent; adefovir and foscamet were interrnediately potent; and acyclovir was least potent. However, all antiviral drugs were able to significantly reduce plaque size in Crandell-Rees feline kidney (CRFK) 4O cells when compared with the untreated control. None of the compounds were toxic for the CRFK cells at antiviral concentrations. A study to assess the effect of cidofovir on viability of feline corneal epithelial cells, replication of FHV—l, and virus-induced cytopathic changes revealed that cidofovir in vitro was highly efficacious against FHV-l infection of a primary culture of feline corneal epithelial cells, but had cytostatic effects on the cultured cells.252 A recent study by Fontenelle et al.253 found that a twice daily application of 0.5% cidofovir solution in both eyes significantly decreased the amount of viral shedding, as determined by quantitative real time PCR, and the severity of clinical disease in cats with experimentally induced ocular FHV -1 infection. Owens et al.220 studied the pharmacokinetics of acyclovir and valacyclovir in cats. Nephrotoxicity and bone marrow suppression were observed with repeated dosing of acyclovir and valacyclovir in cats with experimentally induced FHV-l infection. F uthermore, neither compound appeared efficacious against F HV-l infection. In another study conducted to determine whether orally administered valacyclovir could be used safely and effectively to treat cats with primary F HV-l infection, severe systemic side effects such as coagulative necrosis of the renal tubular epithelium, centrilobular atrophy and hepatic necrosis, and severe bone marrow (1.221 depression were note The experiment had to be terminated after 12 days for humane reasons. Therefore treatment of cats with systemic acyclovir or valacyclovir is not recommended. However, Williams et al.254 evaluated the efficacy of topical ophthalmic acyclovir applied five times daily as a treatment for FHV-l keratitis in cats and concluded that topical acyclovir can have a beneficial effect against FHV-l conjunctivitis and keratitis. 41 A study evaluating the pharmacokinetics and safety of penciclovir following oral administration of famciclovir in cats revealed that famciclovir dosages of 15 mg/kg orally administered every eight hours are unlikely to result in plasma penciclovir concentrations with activity against FHV-l.232 No adverse side effects with the oral administration of famciclovir were observed. However, anecdotal reports suggestive favourable responses after administration of 6-10 mg/kg orally of famciclovir every 12 to 24 hours to clinically affected animals.232 1.13 Non-nucleoside antivirals 1.13.1 Foscarnet Phosphonoformic acid (PF A) is a pyrophosphate analogue inhibiting in a non- competitive way the pyrophosphate binding site of DNA polymerase?” Foscamet does not require phosphorylation. Its efficacy is therefore not dependent on viral thymidine kinase.1 It has been shown to have activity against herpesviruses, human immunodeficiency virus, and other RNA and DNA viruses.255 Foscamet is administered intravenously, and due to its severe renal toxicity it is recommended only for life threatening infections for which other antiviral drugs are no longer 250 effective.1 Maggs and Clarke investigated the antiviral activity of foscamet against FHV-l in vitro and reported it to be relatively ineffective. 1.13.2 L-Lysine Tankersley256 in 1964 studied the amino acid requirements of herpes simplex virus in human cells. Results showed that cells infected with herpes simplex virus deprived of arginine support neither cytopathic effects nor virus replication. However, when arginine was replaced, a prompt and extensive infection followed. Lysine was 42 not required but rather exhibited a partially inhibitory effect upon virus multiplication.256 A later study confirmed that the yield of several DNA viruses in cell culture is 257 Arginine deprivation largely dependent on the presence of arginine in the medium. in the media of HSV cell cultures causes a selective decrease in the synthesis of viral macromolecules. It seems that deprivation of arginine may be more detrimental to virus multiplication than restriction of other amino acids because several specific viral proteins are large in size and rich in arginine.257 A study evaluating the relationship of arginine-lysine antagonism to herpes simplex grth in tissue culture concluded that arginine deficiency suppressed HSV replication while lysine antagonized the viral growth-promoting action of arginine.258 Walsh et al.259 investigated lysine’s therapeutic response when administered to patients with herpes simplex and found lysine to be an effective form of treatment for herpetic infection. L-lysine was shown to reduce the occurrence of, severity of and healing time for recurrent HSV infectionm’m Multiple studies have investigated the mechanism of the arginine-lysine antagonism. Kadirvel and Kratzer262 concluded that the lysine-induced arginine deficiency is a metabolic effect rather than an inhibition of gastrointestinal tract absorption of arginine. Another study supports arginine and lysine competition for a (1.263 Therefore an increase of L—lysine plasma common transport system in the bloo concentration saturates the transport mechanism and thereby reduces the availability of arginine. Arginine is an essential amino acid for cats and cannot be restricted in feline diets due to their extreme sensitivity to an arginine deficiency. Morris and Rogers264 fed cats that were fasted overnight a single meal of a complete amino acid diet 43 without arginine. Signs of severe hyperammonemia developed and one cat died 4.5 l.264 The authors suggest that the rapidity of the onset hours after ingestion of the mea of symptoms arises from a combination of l.) a rapid depletion of free arginine in the liver and therefore of omithine in the postabsorbtive state, and 2.) by inhibition of arginine release from protein catabolism after feeding as a result of the anabolic response of incoming amino acids. Maggs et al.265 examined the effect of various concentrations of L-lysine and L-arginine on in vitro replication of feline herpesvirus-1. Results showed that arginine had a substantial grth promoting effect on F HV -1 and supplementation of the viral culture medium with lysine attenuated this effect. Data indicated that high concentrations of L-lysine reduce in vitro replication of FHV-l in media containing low concentrations of arginine.265 Cats that received oral administration of L-lysine 500 mg every 12 hours beginning 6 hours prior to inoculation of FHV-l developed less severe conjunctivitis after inoculation than cats receiving placebo.266 Plasma L-lysine and arginine levels were measured prior to the study and on days 3, 14, and 22, which was 1 day after cessation of oral administration of L-lysine. Mean plasma L-lysine concentrations were significantly higher than in untreated cats. Arginine plasma concentrations did not differ significantly. The authors did note that if food was withheld prior to oral administration of 500 mg or 1000 mg L-lysine, 50% of the cats vomited within 1 hour.266 Therefore L-lysine should be given with a small amount of food. Another study examined the effect of orally administered L-lysine 400 mg once daily on clinical signs of FHV—l and ocular shedding of virus in latently infected 267 cats The authors found decreased viral shedding but no significant change in clinical signs during reactivation. Plasma L-lysine and arginine concentrations were 44 assessed on day 30 at 3 and 24 hours following L-lysine administration. Three hours following administration, the mean plasma lysine concentration was significantly increased compared to the control group. Mean plasma arginine concentration did not vary significantly between treated and untreated cats. No significant difference in mean plasma lysine or arginine concentrations were detected between the treatment and the control group 24 hours after administration of L-lysine. These data suggest that twice-daily administration of L-lysine would be more beneficial. 1.13.3 Interferons Interferons (IFNs) are glycoproteins produced by cells in response to viral infection.268 They are members of the cytokine family and are important mediators of the initial, innate and adaptive immune system.1 In addition to their antiviral properties, interferons possess anti-proliferative and immune modulating properties.l Although the protein was described in 1954,269 it was named interferon in 1957 due to its viral interference.270 Human interferons are reported to have antiviral efficacy 271-273 against herpes simplex virus as sole agents or administered in combination with ”“76 Topical application early in the course of HSV-1 infection nucleoside analogues. reduces the severity and duration of oral or genital herpes277 and shortens the period of viral shedding.278 Using topical human leucocyte interferon 3x1045 units/ml twice daily in humans with HSV, dendritic ulcers healed more rapidly and viral shedding was significantly inhibited.279 A study comparing the in vitro effect of an undefined feline interferon and two recombinant human alpha-interferons on feline herpesvirus- 1, feline calicivirus, and vesicular stomatitis virus found that treatment with these interferons reduced the viral yield for each of these three viruses as compared with that of the control cultures.280 45 Sandmeyer et al.281 investigated the effect of interferon-alpha on viability of feline corneal epithelial cells, replication of feline herpesvirus-1, and virus-induced cytopathic changes in primary cultures of feline corneal epithelial cells. Interferon- alpha did not have cytotoxic effects and at a concentration of 105 fU/ml was moderately effective against FHV-l infection in primary cell cultures of feline corneal epithelial cells.28' In another study, the effect of recombinant feline interferon omega (rFeIFN-w) administered topically and orally prior to subsequent F HV-l infection in d.282 No beneficial effects of pre-treatment with rFeIFN-w in the cats was investigate course of primary FHV-l infection in cats was observed.282 A cat with a longstanding feline herpesvirus-induced facial dermatitis was successfully treated with rFeIFN- (0.283 The cat received 1.5 million units (MU)/kg on day 0. Half of the dose was injected perilesionally and intradennally, the other half subcutaneously on the lateral thorax. Great improvement was noted after only 2 days. The cat received consecutive injections on days 2, 19, 21, and 23. Four months later at the last recheck examination, the lesion had significantly regressed. Considering the different results of the various studies or case reports, further investigations and controlled clinical trials would be helpful to assess the in viva therapeutic effect of interferons against FHV-l infection. Route of application and time point of application in relation to viral infection as well as type of interferon may play an important role for its efficacy. 1.13.4 Bovine lactoferrin Lactoferrin is an iron binding glycoprotein that has been shown to possess 1 284-287 1 288-290 1 29] 1292-295 9 9 9 antibacteria antifunga antiprotozoa and antivira properties. Mucosal cells of many mammalian species produce lactoferrin. Fujihara et al.295 reported a significant decrease in HSV-l titers following topical administration of 1% 46 lactoferrin using a mouse corneal infection model. Another study also found bovine lactoferrin to have potent antiviral activity against HSV-l infection by competing with the virus for cellular binding sites.296 Bovine lactoferrin was also effective against FHV -1 in cell culture.297 A further study found bovine lactoferrin to have an inhibitory effect on the in vitro replication of F HV-l prior to and during, but not following viral adsorption.298 When CRFK cells were exposed to lactoferrin prior to or during viral adsorption, FHV -1 replication was inhibited by 87-96%. No cytotoxic effect of lactoferrin was observed. The authors concluded that lactoferrin may inhibit F HV-l adsorption to the cell surface and/or the penetration of the virus into the cell. Controlled clinical studies to evaluate the in vivo therapeutic effect against F HV-l infections are warranted. 1.13.5 Vaccination Vaccines against FHV-l have been available for a number of years. There are several types of F HV-l vaccines commercially available, which are invariably given in association with feline calicivirus (FCV) vaccinesf’6 Modified live virus (MLV), and adjuvanted inactivated virus vaccines are available for parental administration. In some countries, intranasal MLV vaccines are available.(’6 Currently available vaccines induce reasonable protection against disease in previously unexposed cats.“"97 However, although severe disease is uncommon in a vaccinated cat, milder forms of FHV-l disease can occur, especially given a high challenge dosem None of the currently available vaccines appear to completely protect against F HV -1 infection or the development of the carrier state. However, both viral shedding and latency load of wild type virus after viral challenge may be reduced in vaccinated cats compared to unvaccinated controls.'97‘299'3°‘ 47 Cocker et al.302 concluded from their study that intranasal vaccination with a cold adapted strain of FHV-l two days prior to challenge gave partial protection, and vaccination four days before challenge gave complete protection against feline viral rhinotracheitis. Weigler et 31.301 evaluated the biology of intranasal vaccination with a temperature sensitive FHV-l mutant. Twelve cats were vaccinated with a temperature sensitive mutant strain of F HV -1 intranasally. Four weeks post vaccination, 6 cats were challenged with a virulent FHV-l field strain. Eight weeks post vaccination, all 12 cats were subjected to a stress protocol (intravenous injection of 30 mg cyclophosphamide followed by 10 mg dexamethasone 24 hours later) in order to reactivate latent infections. Cats were euthanized 13 weeks post vaccination. Utilizing PCR and VI it was shown that all 12 cats shed virus throughout the 4 weeks following vaccination. Although no clinical signs were observed in the 6 cats that were challenged, FHV-l was recovered 4 weeks post challenge. Following euthanasia F HV-l during latency was detected in trigeminal ganglia, optic nerves, corneas, optic chiasma, olfactory bulbs, palatine tonsils and nasal turbinates. The frequency distribution of FHV-l detected by PCR was identical for vaccine-only and vaccine- challenge groups. Therefore, latent infection with vaccine virus can be established following intranasal vaccination. The authors were not able to differentiate vaccine strain from challenge strain of F HV-l by the methods utilized in this study. Sussman et a1.300 performed quantitative PCR to detect latent wild-type F HV-l in the trigeminal ganglia, olfactory bulbs, and brain stems of cats either unvaccinated or subcutaneously vaccinated with FHV-lCV, F HVfi-galglgEA, or FHV-lSA prior to challenge with a virulent FHV-l field strain. Latent FHV-l could be detected at significantly higher concentrations in trigeminal ganglia from cats that were either not vaccinated or vaccinated with FHVB-galglgEA than in ganglia from cats that were 48 vaccinated with either FHV-lSA or FHV-ICV. In the olfactory bulbs or brain stems only extremely low levels of FHV-l DNA could be detected. Another study found that administration of a single dose of a commercially available FHV-l intranasal vaccine within several days prior to exposure lessened clinical signs of disease and F HV-l shedding compared to unvaccinated cats.299 Vaccines against FHV-l are generally safe, and although occasional mild transient clinical signs can follow their use, it is unclear if this is due to the FHV-l or 303 Kruger et al.304 reported that cats vaccinated subcutaneously the F CV component. or intranasally with high doses of a recombinant FHV-l strain responded with only mild clinical signs and developed strong immunity against subsequent virulent virus challenge. Care should be taken to avoid inadvertent oronasal administration of attenuated FHV-l vaccines labelled for intramuscular or subcutaneous administration as disease may be induced.7"304'306 This occurs because vaccines labelled for intramuscular or subcutaneous administration are truly virulent strains. When administered intramuscularly or subcutaneously, virulence is completely masked. When this commercially available vaccine is administered by the oronasal route to cats disease is induced as seen with natural infection.304 A number of genes have been targeted for the deve10pment of recombinant FHV-l vaccines. These targeted genes mainly encode enzymes involved in DNA synthesis, such as thymidine kinase and ribonucleotide reductase genes, and a number of glycoprotein genes.197 Yokoyama et 31.307 developed a recombinant FHV-l attenuated live vaccine, which was deleted in a defined region within the thymidine kinase gene. This attenuated live vaccine caused only mild symptoms and provided siginificant protection against challenge with the parent F HV-l strain. The deletion also served as a genetic marker for PCR assays and allowed for easy detection of and 49 differentiation of the vaccinal strain. The level of protection was later improved through the addition of a late promoter gC in the construct.308 Glycoproteins that are essential can be developed for use as subunit vaccines; whereas genes of non-essential glycoproteins can be targeted for the construction of ‘97 Recombinant poxviruses and baculoviruses that express the F HV- deletion mutants. l glycoprotein D have been constructed to characterize its properties and potential as an immunogen.309’3 '0 Also, multiple FHV-l deletion/insertional mutants have been 300,304,31 l 312 developed including gE-gl deletion mutants, a gI insertional mutant, and an insertional mutant of ORF2, downstream of glycoprotein C313 These mutants are generally less virulent and are reasonably protective against disease, especially when administered via the oro-nasal route.66 However, since the protection offered by these new mutants is not superior to conventionally attenuated vaccines, none has been marketed so far.66 1.14 Conclusions Herpesviruses are of major medical importance across all species. Despite extensive research in the past 150 years, many details of the biological nature of the different herpesviruses still have to be elucidated. FHV -1 remains a widespread pathogen within the feline population worldwide. Expansion of our current knowledge and understanding will potentially offer new perspectives for prevention and therapy of the multiple manifestations of feline herpetic disease. Expanded knowledge of the herpesvirinae also becomes critical because of their potential new applications. A large area of current research focuses on the utilization of HSV-1 as a viral vector for gene therapy and its potential as an oncolytic virus. If cats and humans will be purposely inoculated with the herpesvirinae as therapeutic agents, we must ensure that all ramifications of that infection are known. 50 Through genetic engineering and recombinant DNA techniques, the herpesvirinae are being utilized in the creation of novel vaccines which function as delivery agents. The delivery vaccine consists of an attenuated live vector in which foreign genetic information from one or more other pathogens has been incorporated in order to induce protective immunity against those pathogens and potentially against the field type variants of the vector.314 Recombinant F HV-l has been evaluated as a . . . . . . . . -1 vector for rmmunrzatron agarnst feline rmmunodeficrency vrrus,315 3 8 308,319,320 321 calicivirus, and toxoplasmosis. Viral oncolysis, the destruction of neoplastic cells by replicating viruses is a promising strategy for cancer treatment. While numerous viruses may be used for this type of therapy, HSV-l is an attractive vector due to its high infectivity, ease of genetic engineering, and a large transgene capacity.322 Positron emission tomography (PET) has become a useful tool for tracking sites of viral replication.323 HSV thymidine kinase serves as the PET reporter gene. The PET scan provides a novel, non-invasive, easily repeatable instrument to ascertain the location and magnitude of viral replication and tumor lysis.322 51 2. CHAPTER — ANATOMY AND HISTOLOGY OF TISSUES RELEVANT TO THIS STUDY 2.1 Cornea The cornea is the anterior continuation of the sclera, the fibrous tunic of the globe. The transition zone between the cornea and the sclera is called the limbus. The careful arrangement of the fibrous structure and the absence of blood vessels and pigment allow transmission of light. The cat cornea is slightly conical and has a rather steep curvature with a radius of 8-9 mm.324 The shape of the cornea is almost circular with the horizontal diameter (16 mm) slightly greater than the vertical diameter (15.5 ).324 This shape allows 40-42 diopters of light refraction.325 In the cat the central mm thickness of the cornea varies only slightly from the periphery. The average overall thickness is 0.62 mm — 0.68 mm.324‘325 The most anterior of the corneal layers, the corneal epithelium, is a continuation of the conjunctiva, and is composed of nonkeratinized, stratified squamous epithelium.324‘326 The epithelium has one basal layer of columnar cells, two 324 The most or three layers of polyhedral cells, and 7-10 layers of squamous cells. superficial layer of squamous cells is in direct contact with the precomeal tear film and has a microplicated surface.327 The corneal stroma (substantia propria) comprises 90% of the thickness of the cornea — approximately 0.55 mm in the catm’325 Parallel bundles of collagen fibrils form lamellae, with each lamella running the entire diameter of the cornea.325 The collagen fibrils within a lamella are parallel, but between lamellae, they vary in direction.325 The fixed fibrocytes of the cornea, the keratocytes, lie flattened between 325 the lamellae. These cells and their extensions form and maintain the stromal 52 325 lamellae. The precise organization of the corneal stroma is the most important factor in maintaining corneal clarity.3’25‘327 Descemet’s membrane, a homogeneous, acellular membrane, is the exaggerated basement membrane of the corneal endothelium.325 It is produced throughout life, becoming thicker as the animal ages.325 In cats, the thickness of Descemet’s membrane ranges from 8-12 “111.324 The innermost layer of the cornea, the endothelium, is a single layer of flattened hexagonal cells.325’327 Its thickness in cats is 3-4 pm.”4 The avascular cornea is nourished by the capillary loops at the limbus, the pre- corneal tear film, and the aqueous humor.327 Branches of the ciliary nerves that arise from the ophthalmic nerve, a branch of the trigeminal nerve innervate the cornea.327 2.2 Conjunctiva The anterior corneal epithelium is continuous with the bulbar conjunctiva at the corneoscleral junction.327 The epithelium of the lower nasal and middle fomix and the lower tarsal portion of the palpebral conjunctiva contain the highest density of goblet cells.325 The bulbar conjunctiva essentially lacks goblet cells.325 Two layers are distinguished within the substantia propria of the conjunctiva, a superficial adenoid layer, which contains lymphatic follicles and glands, and a deep, fibrous layer. The 325 fibrous layer contains the nerves and vessels. A superficial and a deep plexus of lymphatics are present in the conjunctiva.325 Drainage extends to the parotid lymph nodes and the submaxillary lymph nodes.325 2.3 Lacrimal gland The lacrimal gland lies dorsolaterally enclosed within the periorbita.324‘325 The gland is 1 mm thick with an equal width and a length of 14 mm.324 The thickness is 53 324 maximal centrally and tapers towards the periphery. Its surface is slightly lobulated, 324 and its anterior edge lies very close to the limbus. The lacrimal gland is 324 tubuloalveolar and of serous quality. It is composed of columnar cells and the secreting acini are arranged in lobules of different sizes, supported by a relatively 324 sparse stroma of connective tissue. The lumen of the interlobular ducts is 324 approximately 30 pm in diameter. The lateral artery, a branch of the internal 24 d.3 maxillary artery serves the lacrimal glan Two lacrimal veins enter into the external rete, which is drained by the ophthalmic vein from which it drains into the external jugular vein.324 The lacrimal nerve, a branch of the ophthalmic division of the trigeminal nerve, receives sympathetic fibers via the internal carotic plexus and parasympathetic fibers via the pterygopalatine ganglion.328 2.4 Nictitans gland The gland of the nictitans surrounds the caudal end of the shaft of the cartilagenous plate of the third eyelid.325 It is thin, and has a flat roughly triangular shape with its apex pointing caudally towards the apex of the orbit.324 Most of the gland lies on the palpebral side of the cartilage, where it is 1 to 1.5 mm thick, whereas 324 on the bulbar side the gland is very thin. It is a serous gland in the cat.325 Although 324 the acini are positive for PAS suggesting a seromucoid type. The nictitans gland is 24 (1.3 much more richly endowed with septa than the lacrimal glan These septa carry 324 many prominent interlobular ducts and blood vessels. The ducts within the gland 324 Many fat cells are found within the septa.324 The have diameters up to 60 um. nictitans gland receives its parasympathetic innervation via the infratrochlear nerve, a branch of the nasociliary nerve which is a branch of the ophthalmic division of the trigeminal nerve.328 54 2.5 Trigeminal ganglion 329 The trigeminal nerve is the 5th cranial nerve. It has both motor and sensory 329 . . . . . 2 , 29 components. The Radzx sensorza rs much larger than the Radzx motorra.3 8 3 The trigeminal nerve enters the trigeminal canal on the rostromedial aspect of the petrosal 9 bone}2 Within this canal is the large trigeminal ganglion. It contains sensory cell bodies of the afferent, sensory fibersm’329 The three branches of the trigeminal nerve emerge from the trigeminal ganglion: the ophthalmic nerve, the maxillary nerve, and the mandibular nervem’329 The Radix motoria, which also contains afferent, proprioceptive fibers, crosses the trigeminal ganglion medially and unites rostrally with one sensory branch forming the mandibular nerve, which is a mixed nervem’329 The ophthalmic nerve consists of sensory axons from the eyelids, the globe and the nose.329 The nerve passes through the orbital fissure of the skull. Within the periorbita, it divides into three branches: the frontal nerve, lacrimal nerve, and nasociliary nerve.329 These branches innervate the skin of the upper eyelid, lacrimal gland, cornea, conjunctiva, nasal cavity, and the skin of the nostril.329 The maxillary nerve is sensory to the lower eyelids, nasal mucosa, upper teeth, upper lip, and nose, and its distal branches contain postganglionic parasympathetic axons (via fibers from the pterygopalatine ganglion) that innervate the lacrimal, nasal, and palatine glandsm’329 The nerve leaves the cranial cavity through the round foramen, alar canal, and rostral alar foramen and courses rostrally to the maxillary 329 foramen. In the pterygopalatine fossa the maxillary nerve gives rise to the zygomatic nerve, pterygopalatine nerve, and infraorbital nerve.329 The mandibular nerve is both motor and sensory. It exits the cranial cavity 328 through the oval foramen. The nerve divides into multiple branches: masseteric nerve, lateral and medial pterygoid nerves, tensor tympani nerve, tensor veli palatine 55 nerve, auriculotemporal nerve, the inferior alveolar nerve, the mylohyoid nerve, and the lingual nervem’329 The mandibular nerve is sensory to the buccal cavity, tongue, teeth of the lower jaw, the lower lip, in part the skin of the head, and mucosa of the intraosseous part of the external ear canal.329 2.6 Ciliary ganglion The oculomotor nerve is the 3rd cranial nerve. It consists of somatic efferent neurons that innervate several of the striated extraocular muscles (dorsal rectus muscle, medial rectus muscle, ventral rectus muscle, ventral oblique muscle, and parts of the retractor bulbi muscle) and visceral efferent neurons (parasympathetic) that innervate ocular smooth muscle.‘7’28’329 The nerve exits the cranial cavity through the . , 9 orbrtal fissue.328 32 Upon entering the orbit, the nerve divides into a smaller dorsal branch and a ventral branchm’329 The ventral branch continues rostrally passing lateral and slightly ventral to the optic nerve, where it terminates in a number of branches to the medial rectus, ventral rectus, and ventral oblique musclesm’329 At its termination is the ciliary ganglion where the preganglionic parasympathetic fibers 329 synapse with postganglionic neurons. The two short ciliary nerves contain the postganglionic axons, innervating the ciliary muscle and the iris sphincter muscle of the eye.328-330 56 3. CHAPTER - HYPOTHESES AND SPECIFIC AIMS This investigation was designed to follow infection with FHV-l through the acute phase and into latency. The viral quantity, location, replicative status, and local tissue effect of the virus were assessed through virus isolation, quantitative PCR, in situ hybridization, and routine histopathologic examination of corneas, conjunctiva, lacrimal glands, nictitans glands, trigeminal ganglia, and ciliary ganglia at various time points during the infectious process. 3.1 Hypotheses 1. F HV-l DNA is present during both the acute and latent stages within the corneas, conjunctiva, lacrimal glands, nictitans glands, trigeminal ganglia, and cilary ganglia of experimentally infected cats. 2. FHV-l infects lacrimal tissue and leads to inflammation within the lacrimal gland and nictitans gland. 3.2 Specific aims l. Localization of F HV-l within the cornea, conjunctiva, lacrimal gland, and nictitans gland by in situ hybridization. 2. Quantification of F HV -1 within cornea, conjunctiva, lacrimal gland, nictitans gland, trigeminal ganglion, and cilary ganglion during the acute and latent stages of infection by use of a real-time PCR assay. 3. Use of virus isolation to determine whether the F HV-l is active or latent when detected in the tissues using the real-time PCR assay. 57 4. Assessment of local tissue effect of the virus within cornea, conjunctiva, lacrimal gland, nictitans gland, trigeminal ganglion, and cilary ganglion during the acute and latent stages of infection. 58 4. CHAPTER — RESEARCH PROTOCOL, RESULTS, AND DISCUSSION 4.1 Introduction Feline herpes virus 1 (FHV-l) infection is widespread within the cat population.4 Herpesviral ocular disease can be quite severe. Typical acute manifestations include ocular discharge, blepharospasm, conjunctivitis, keratitis, and 4,79 corneal ulceration. Chronic ocular manifestations and sequelae may include persistent conjunctivitis, symblepharon, stromal keratitis and possibly corneal sequestration and eosinophilic keratitis.77'80’163 Decreased tear production in cats has long been suspected to be a potential sequela to FHV-l infection.33 I The cause remains speculative, but it has been assumed that either excretory duct occlusion or lacrimal adenitis is responsiblefl’80 To the authors knowledge, histopathologic examination of the lacrimal tissues has not been performed to support either of these theories. Experimentally, a temporary keratoconjunctivitis sicca developed in 5 of 10 cats treated pre-inoculation with three consecutive daily subconjunctival injections of betamethasone.80 However, aqueous tear production returned to normal in all cats by day 60 post-infection.80 Bistner, et al.m failed to demonstrate the presence of F HV-l within the lacrimal gland utilizing specific fluorescent antibody staining or virus isolation. Interestingly, fluorescein- labelled antibody has been used to examine human lacrimal glands and revealed HSV-1 viral antigen in 2/3 patients with acute herpetic keratitis and in 4/7 asymptomatic patients with a history of previous herpetic infection.145 The potential for recrudescent infection is high because the virus remains latent in approximately 80% of infected cats and 50% of FHV-l recovered cats will shed virus spontaneously or in response to stress.65 During the latent period clinical signs are absent and the virus cannot be isolated using standard cell culture 59 techniques.38 FHV-l DNA has been detected utilizing PCR assays in the nasal turbinates, oral fauces, tonsils, salivary glands, mesenteric lymph nodes, conjunctiva, corneas, lacrimal glands, optic nerves, trigeminal ganglia, olfactory bulbs, and cerebra of latently infected cats.85’I44 Latency was confirmed by concurrent negative virus isolation.38 Furthermore, using in situ hybridization and northern blot analysis of trigeminal ganglia, Ohmura et al. detected transcripts of the strand opposite to the immediate-early mRNA in cats latently infected with FHV-l.193 These transcripts likely represent latency associated transcripts (LATs) which are the only transcripts present during latency and may therefore serve as markers of latency.”38 Townsend et al.198 also detected LATs utilizing a reverse transcriptase PCR assay in 4/16 trigeminal ganglia of clinically normal cats. However, to the authors knowledge histologic examination of the latently infected neural tissues to determine the effect of the F HV -1 infection has not been performed. In humans with a history of previous herpes simplex-l (HSV-l) infection, the ciliary ganglion has been identified as a site of latency. Bustos et al.168 identified and detected HSV-l utilizing PCR in 20/30 ciliary ganglia from human cadavers with no history of active herpetic lesions at the time of death. However, the ciliary ganglion has not been evaluated for presence of FHV-l during either active or latent infections in cats. The objectives of this study are to utilize virus isolation, a quantitative real- time polymerase chain reaction assay, histopathologic examination, and in situ hybridization to localize and quantify the amount of F HV-l present and evaluate the associated morphological changes within the corneas, conjunctiva, lacrimal glands, 60 nictitans glands, trigeminal ganglia, and ciliary ganglia of experimentally inoculated specific pathogen free cats during both the acute and latent phases of infection. 4.2 Materials and Methods 4.2.1 Animals Fifteen 6-month-old female SPF cats were obtained from a commercial vendor. Cats were housed in individual cages with treatment and control groups in separate rooms (Biocontainment Level-2) with controlled temperature, humidity, and lighting. All personnel donned sterile disposable Tyvek coveralls, gloves, head covers, and shoes covers upon entering a room. Cats were fed a combination of dry and moist diets. All cats were acclimated for 7 days before viral inoculation. A11 cats were determined to be serologically negative for FHV -1 virus neutralizing antibody prior to inoculation. Blood samples were collected on days 7, 14, and 21 post inoculation from the group 3 cats for evaluation of FHV-l virus neutralizing antibodies. 4.2.2 Inoculation and clinical scoring Cats were anesthetized via mask administration of halothane and oxygen. Twelve cats were infected by instilling 1.05 ml (1x105 TCIDso/ml) of the C-27 strain of FHV-l into the lefl and right conjunctival sacs (350 pl each), external nares (150 pl each), and oropharynx (50 ul) of each cat. The other 3 cats served as controls. The C27 strain of FHV -1 was obtained from a commercial source (ATCC, Manassas, VA) and was certified to be a purified suspension. Clinical signs including fever, sneezing, ocular or nasal discharge, anorexia, dyspnea, and dehydration were scored daily for each cat as described (U .S. 61 Department of Agriculture, Animal and Plant Health Inspection Service, National Animal Veterinary Services Laboratory, 1985) by 3 investigators (Jacobi, Wasserrnan, Townsend) (Appendix A). Previously published work304 had established that all animals should be exhibiting ocular signs on day 6, all animals should be at the peak of ocular and respiratory signs on day 10, and latency should be established with resolution of clinical signs by day 30. Therefore, on days 6 (group 1), 10 (group 2) and 30 (group 3), four inoculated and one control cats were humanely euthanized and samples were immediately collected. All experiments were performed in full compliance with the ARVO Statement for the Use of Animals in Ophthalmic and Vision Research and with approval from the Michigan State University Animal Care and Use Committee. 4.2.3 Samples Aseptic, bilateral sample collection (corneas, conjunctiva, lacrimal glands, nictitans glands, trigeminal ganglia, ciliary ganglia) occurred immediately following euthanasia. Particular care was taken to avoid cross contamination between tissue samples by alcohol dipping and flaming of the instruments for each use. Tissue samples or entire ganglia from the right side were placed in Eppendorf tubes containing one sterile homogenizing bead with 800 u] of Bovamick’s buffer and homogenized for further virus isolation and PCR analysis. Tissue samples from the left cornea, conjunctiva, lacrimal gland, and nictitans gland were divided. One half was placed immediately in 10% formalin; the other V2 was frozen at -80 °C. Due to their limited size, the entire left trigeminal ganglion and ciliary ganglion were placed immediately in 10% formalin. 62 4.2.4 Virus isolation Following homogenization in the Qiagen Mixer mill (QIAGEN Inc., USA) and collection of a 200 pl aliquot for DNA extraction, the remaining suspension was transferred to a 15 ml conical tube. The volume was brought to 3 ml with Eagle minimal essential medium (EMEM) supplemented with 10% fetal bovine serum (FBS). The homogenate was then filtered (0.45 pm). Filtered tissue extracts (500 pl) were inoculated onto monolayers of Crandell feline kidney (CRFK) cells grown in 24- well microtiter plates. All cultures were performed in duplicate. The cultures were observed daily for five days with an inverted microscope (Nikon Eclipse TSlOO) for the cytopathic effects (CPE) characteristic of FHV-l. A sample was considered positive for FHV -1 only if it produced CPE within both culture wells. If only one culture was positive, another culture was performed for the sample, again in duplicate. The result was then considered positive if CPE was produced in one or both of the repeat cultures. 4.2.5 Quantitative real time PCR 4.2.5.1 DNA extraction A mixture of 100 pl tissue homogenate and 100 pl phosphate buffered saline (PBS) was utilized for DNA extraction. Extraction controls, performed for each extraction, consisted of 200 pl PBS. Purification of DNA was performed using a commercially available kit (DNeasy Tissue Kit, QIAGEN, Valencia, CA) in accordance with the manufacturer’s protocol. The samples were eluted with 100 pl Buffer AB (10 mM Tris-Cl, 0.5 mM EDTA; pH 9.0). DNA was quantified using a spectrophotometer (NanoDrop 1000, Therrno scientific, Wilmington, DE). 63 4.2.5.2 PCR amplification of F HV-l A real-time SYBR Green-based PCR specific for FHV -1 has been previously described.332 The primers for this assay target a 478-bp fragment of the FHV-l gE gene. The sequence of the forward primer is 5’-GGT CAT GTG TAA TGT TGA CG- 3’, and the reverse primer is 5’- GTC TTT GGT TCT GAT GAG AG-3’.3“ The sensitivity of this assay is 10 viral genome copies. The diagnostic utility of the assay has been confirmed by its ability to amplify the targeted DNA fragment of 48 known clinical isolates of FHV-l. The 50 ul real time PCR reaction mixture consisted of 25 pl 2x SYBR Green mix (Taq DNA polymerase, dNTPs, SYBR Green I, 5.0 mM MgC12; QIAGEN QuantiTect SYBR Green PCR Kit, Valencia, CA), 1 u] forward primer (25 pmol/ul), 1 ul reverse primer (25 pmol/ul), 100 ng of template DNA and ddeO. PCR amplification was performed on an iCyclerTM iQ® System (Bio-Rad Laboratories, Hercules, CA) with the following cycling conditions: pre-denaturation at 95 °C for 15 min, followed by 40 cycles of 94 °C for 30 sec, 50 °C for 30 sec and 72 °C for l min. Amplification was continuously monitored in real time by quantifying the amount of fluorescence emitted at 530 nm at each annealing step. Following the final elongation step at 72 °C for 60 seconds, a post-PCR melt curve analysis was performed, amplicon melt temperatures were determined by raising the temperature in 0.5 °C increments from 55 °C to 95 °C and monitoring changes in emitted fluorescence (Figures 4.1 and 4.2). All reactions were conducted in duplicate. DNA extraction controls and reagent controls were included in each run. A standard curve of threshold cycle (Ct) in relation to starting viral copy number was constructed (Figures 4.3 and 4.4). The standard curve for F HV-l was generated by a series of 10- fold dilutions (10l - 106) of FHV-l DNA. Samples for the standard curve were assayed in triplicate for each run and served also as the positive control. 64 Melt Curve Chart: Data 2006-05-15 2150.0pd RFU 55 60 65 70 80 85 90 95 Temperature, Celsius Figure 4.1. Standard melt curve chart. Melt Peak Chart: Data 2006-05-15 2150.0pd -d( RFU )ldT 55 6O 65 7O 75 80 85 90 95 Temperature, Celsius Figure 4.2. Standard melt peak chart. 65 Melt Peak Chart: Data 2006-05—1 5 2150.0pd 2200 2000 1 800 1 600 1400 1 200 1 000 800 600 400 200 O PCR Base Line Subtracted Curve Fit RFU 0 5 10 15 20 25 30 35 40 45 Temperature, Celsius Figure 4.3. Standard amplification chart, 10 fold dilutions of 106 genomic copies of FHV-l, triplicate. 0 Standard 40 35 - i- if :7 1111 25 * 20 15......4...... O 2 4 6 8 Log Starting Quantity, copy number IL Threshold Cycle 1 L111 4 q + SYBR IE=85.6% R"2=0.987 slope=-3.725 y-int=39... PCR Standard Curve: Data 2006-05-15 2150.0pd Figure 4.4. PCR standard curve. 66 4.2.5.3 Statistical analysis Data were analysed using the Wilcoxon Rank -Sum test for non Gaussian data distribution. Following Bonferroni’s adjustment for multiple comparisons, the level of significance was p < 0.017. For all tissues mean copy numbers of the 3 time points (day 6, day 10, and day 30 post inoculation) were compared. 4.2.5.4 Histopathology Following fixation in 10% formalin, tissues were paraffin-embedded, sectioned at 5 pm, deparaffinized, and stained with hematoxylin and eosin (HE). All samples were examined with a light microscope. 4.2.5.5 In situ hybridization In situ hybridization was performed to specifically localize FHV-l within the cornea, conjunctiva, lacrimal gland, and nictitans gland. The in situ protocol was first optimized by evaluating various pretreatment methods: 1.) baking for 30 min at 60 °C prior to deparaffinization of the section followed by enzymatic digestion (100 ul/ml proteinase K in TBS buffer (50 mM Tris HCl, pH 7.4, 10 mM EDTA, 10 mM NaCl)) for 15 minutes at 37 °C; 2.) enzymatic digestion following deparaffinization without previous baking; 3.) microwaving: slides covered with 10 mM Tris HCl, pH 8.1 + 1 mM EDTA buffer were microwaved at full power for 5 or 10 minutes. Also antibody dilutions of 1:250 and 1:500 were evaluated (Figure 4.5). All optimization was performed on F HV-l pellets. Superior results were obtained by pre-treating via microwaving for 10 minutes and using an antibody dilution of 1:250 (Figure 4.6). This pretreatment protocol was used for all tissue sections. 67 ‘ ~0- niés ‘ Is an...) .‘ gm ' Figure 4. 5. FHV- 1 pellet pre-treated with different protocols. A. Enzymatic digestion with proteinase K using an antibody dilution of 1:500. B. Enzymatic digestion with proteinase K using an antibody dilution of 1:250. C. Baking for 30 min at 60 °C prior to deparaffinization of the section followed by enzymatic digestion with proteinase K using an antibody dilution of 1:250. D. Micowaving for 10 minutes using an antibody dilution of 1:500. 68 ‘ — ‘ , A - F . n - r r .- _ v - ._ ‘ . . o , 1,. I, .'f _ t ‘ . : . at, ' “F r , , > - ‘r-O . 4 pre-treated via micowaving for 10 minutes using an [.8 1.3.- ‘. Figure 4.6. FHV-l pellet antibody dilution of 1:250. Comea, conjunctiva, lacrimal gland, and nictitans gland were examined in duplicate. The FHV-l DNA probe used targets a portion of the gE gene. The 30-mer synthetic oligonucleotide probe with the sequence 5' CGA TGA CAG AAG GTG CCG CAC TTT TAG TCG ATG GG 3' was synthesized and labeled with digoxigenin-l l-dUTP by a commercial supplier (IDTDNA, Coralville, IA). Positive (FHV-l pellet) and negative controls (canine nictitating membrane, F HV-l pellet with no probe applied) were included with each batch of slides. Paraffin sections were prepared on silane-coated slides. Sections were then dewaxed in xylene (2 x 10 min), taken through a series of graded ethanols (l x 5 min in 100%, 95%, 75%, and 50%) and washed in nanopure water (2 x 5 min).195 Sections were then placed in Tris/41., ' J' ' ‘ ‘ " acid (EDTA) buffer (10 mM Tris HCl, pH 8.1 + 1 mM EDTA) and microwaved at full power continuously for 10 69 minutes. Sections were allowed to cool for 15 minutes and nanopure water was gradually added over 5 minutes to reduce the temperature. ‘95 Tissues were then fixed in 4% paraformaldehyde in phosphate buffered saline (PBS) for 10 minutes.333 Slides were allowed to air dry and the tissue sections were layered with 75 pl of prehybridization solution containing 4X saline sodium citrate (SCC); 50 mM NaHzPO4/Na2HPO4, pH 7.0; 1 mM EDTA; 5X Denhardt’s solution (1% Ficoll, 1% polyvinylpyrrolidone and 1% bovine serum albumin); 10 pl sonicated salmon sperm DNA(500 pg/ml); formamide to 25%; and nanopure water. The solution was held in place by the use of a liquid blocker (PAP PEN, Invitrogen, Carlsbad, CA). Prehybridization was allowed for 60 minutes at room temperature. Then hybridization solution consisting of the prehybridization solution and 200 ng/ml DIG labeled probe was layered over each section. Slides were heated for 6 minutes at 95 °C and immediately quenched on ice for 1 minute. Hybridization followed at 25 °C overnight in a humidified chamber. Sections were then washed twice in 2X SCC for 5 minutes and 0.1X SCC for 10 minutes at room temperature. Slides were rinsed briefly in buffer 1 (100 mM Tris HCL, 150 mM NaCl, pH 7.5). The tissues were then covered with buffer 2 (100 mM Tris HCL, 150 mM NaCl, pH 7.5, and 0.5% blocking reagent) and cover-slipped with parafilm. Slides were allowed to sit for 15 minutes at room temperature. Following removal of the cover slips, slides were rinsed briefly in buffer 1. Then, 30 pl of 1:250 anti-DIG AP, Fab fragment antibody conjugate was layered onto the tissues, which were cover-slipped with parafilm, and placed in a humidified chamber at 25 °C for 2 hours. Slides were then washed twice for 10 minutes in buffer 1 and allowed to equilibrate for 5 minutes in buffer 3 (0.1 M Tris HCL, pH 9.5, 0.1 M NaCl) at room temperature. Then 20 pl of 1:50 NBT/BCIP solution was layered onto each tissue, cover-slipped with glass cover slips, and covered with aluminum foil to prevent illumination. Color was allowed to develop overnight at 25 °C. Following removal of the cover-slips, the reaction was stopped by rinsing the slides briefly in 10 mM Tris HCl, pH 8.1 + 1 mM EDTA. Slides were then washed in nanopure water. Neutral red counter-stain was layered over each tissue for 1 minute and then rinsed with nanopure water. Slides were then cover-slipped (Irnmu-mount, Shandon, Pittsburgh, PA) and evaluated under a light microscope. 4.3 Results 4.3.1 Clinical features All inoculated cats euthanized on day 30 post inoculation developed antibodies against F HV-l with titers ranging from 1:32 to 1:128. The control cat did not develop antibodies against FHV-l. None of the control cats showed any signs of disease at any time. Sneezing developed in 11/12 inoculated cats with a peak between day 3 and 9 post-inoculation. None of the cats developed a cough, rales, open mouth breathing, or corneal or cutaneous ulceration. Blepharospasm of variable severity and duration was noted in 10/ 12 inoculated cats. All 12/ 12 inoculated cats developed serous or in some cases mucopurulent ocular discharge of variable duration and severity between days 3-14 post-inoculation. Conjunctivitis was found in all 12/ 12 inoculated cats of variable duration and severity starting on day 3 post-inoculation. Other signs noted in variable severity and duration were nasal discharge in 10/12 inoculated cats, salivation in 3/ 12 inoculated cats and oral ulceration in 9/ 12 inoculated cats. Two of the 12 inoculated cats developed mild signs of dehydration, 6/ 12 cats developed a transient, mild anorexia, and 5/ 12 cats had a transient pyrexia (>102.5 °F) between days 3 and 7 post-inoculation. None of the cats required or received any treatment 71 during the study. See clinical scores in Table 4.1. Detailed data are shown in Appendix B. Day of euthanasia post inoculation Cat number Clinical score Day 6 1 8 2 7 12 4 4 5 5 0 Day 10 6 24 7 19 8 16 9 31 10 0 Day 30 11 20 3 44 13 27 14 20 15 0 Table 4.1. Clinical scores. 4.3.2 Virus isolation None of the tissues were positive on virus isolation for the control cats. Virus isolation from the four inoculated cats euthanized on day 6 and 10 post-inoculation 72 demonstrated 2/4 positive trigeminal ganglia, 2/4 positive ciliary ganglia, and 4/4 positive lacrimal glands, nictitans glands, corneas, and conjunctiva respectively. Virus isolation from the four inoculated cats euthanized on day 30 post-inoculation demonstrated only 1 positive sample; a corneal specimen. All other tissues from the group 3 cats were negative for virus isolation of FHV-l. Results are summarized in the Table 4.2. Data are shown in detail in Appendix C. Cornea 4/4 4/4 'unctiva 4/4 4/4 Tri ' 2/4 2/4 C' ' ' 2/4 2/4 Table 4.2. Summarized results of virus isolation (positive/total) 4.3.3 Quantitative real time PCR FHV-l DNA could not be detected in any of the tissue samples from the control cats. In all infected animals FHV -1 DNA could be detected in cornea, conjunctiva, trigeminal ganglia and ciliary ganglia on days 6 and 10 post inoculation. All inoculated cats had positive trigeminal ganglia on day 30. In addition, one cat was positive for FHV-l DNA in the corneal, conjunctival, and ciliary ganglion samples on day 30. The three remaining inoculated cats were negative for those tissue samples on day 30. (Table 4.3) Copy numbers per 100 ng DNA were similar for the corneal samples in all cats on day 6 and day 10. In conjunctival samples, copy numbers tended to be lower on day 10 than on day 6 post-inoculation. For the trigeminal ganglion and the ciliary ganglion, copy numbers tended to be increased on day 10 compared to day 6. However, statistical analysis using the Wilcoxon Rank-Sum test with a level of significance of p < 0.017 following Bonferroni’s adjustment for multiple comparisons, 73 showed that none of the comparisons between the 3 different time points were significant. PCR results for the cornea, conjunctiva, lacrimal gland and the nictitans gland tissues for cats # 4, 7, and 12 revealed positive extraction controls. PCR for these samples was not repeated for the glandular tissues due to the fact that upon histologic examination, conjunctival tissue was found to be adherent to the glandular material and could have resulted in false positive results because of possible presence of F HV- 1 in the conjunctiva rather than in the gland itself. PCR was repeated for the corneal and conjunctival tissues of cats # 4, 7, and 12 in duplicate. Results are summarized in the Table 4.3. Data are shown in detail in Appendix D. 10 30 Cornea 4/4 4/4 1/4 tiva 4/4 4/4 2/4 Tri ' 4/4 4/4 4/4 C' ' ° 4/4 4/4 1/4 Table 4.3. Summarized results of quantitative real time PCR (positive/total) 4.3.4 Histopathology None of the control cats showed any histomorphologic abnormalities in any of the tissues examined. Conj unctival changes characterized by epithelial cell attenuation or loss and lymphoplasmacytic inflammation were seen in all 12 inoculated cats (Figure 4.7), although the changes were milder in cats euthanized on day 30 post- inoculation. In these cats areas of conjunctival re-epithelialization were present. All corneal sections were normal except for those from one cat (cat # 9) euthanized on day 10 post-inoculation. Multiple, solitary, degenerate epithelial cells and a mild lymphoplasmacytic infiltrate characterized the changes within the corneal section (Figure 4.8). 74 The glandular tissues within all sections of lacrimal and nictitans glands appeared normal (Figure 4.9). However, histopathologic examination revealed that multiple specimens of lacrimal and nictitans gland contained the adjacent conjunctiva. The conjunctiva in these samples exhibited changes consistent with those noted in all other conjunctival samples. All 4 trigeminal ganglion samples obtained on day 6, 3 samples obtained on day 10, and 1 sample obtained on day 30 post-inoculation revealed histopathologic changes including ganglion cell necrosis, drop out or loss of ganglion cells, increased numbers of glial cells, and lymphoplasmacytic inflammation (Figure 4.10). All ciliary ganglia evaluated appeared normal. Due to the small size of the ganglia from one cat euthanized on day 10, no appropriate section of the trigeminal ganglion could be obtained. From 1 cat (day 10) and 2 cats (day 30 post inoculation) no appropriate section of the ciliary ganglion could be obtained. 75 r v-2- Figure 4.8. Corneal sample from cat # 9. (HE) Figure 4.7. Conjunctiva] sample from cat # l2. (HE) 76 :. .- r- . . W '3‘ ' I! ”HWLMO??'%§L£" 1 ~ 14% .. .7?va ‘ - P' -'.' ,. in _. cat # 2. HE . " pr . . ‘V r , f ' 3 j: " ' .. a; _‘-. {5.9 g ‘ 3.: “14.2%: .. ‘ Figure 4.10. Trigeminal ganglion sample from cat # 1. (HE) 4.3.5 In situ hybridization Results of in situ hybridization are summarized in Table 4.4. A blue to brown reaction product confined to the nuclei of intact epithelial cells characterized a positive hybridization signal. None of the control cats showed positive results in any of the tissue samples. In conjunctival samples the positive cells were found adjacent to areas of inflammation (Figure 4.11). No inflammatory changes were noted adjacent to the positive superficial corneal epithelial cells (Figure 4.12) Hybridization signals were not detected within the lacrimal or nictitans glands. However, positive hybridization signals were detected within the overlying conjunctiva. Corneal samples of cat # 2 were not available for evaluation via in situ hybridization. The tissue quality of all 4 samples from the group 3 (day 30) cats was very poor. One cat appeared to have a positive hybridization signal in the cornea, but this result may be spurious due to the poor tissue morphology. ISH Cat N o. Cornea Con'unctiva Lacrimal Nictitans J Gland Gland 1 + + - - 2 Not avail. Day6 12 - +++ I I 4 + 5 (control) - 6 - 7 - - - - Day 10 8 - - - - 9 - - - - 10 (control) - - - - 11 3 Day 30 13 Poor tissue morphology 14 15 (control) Table 4.4. Summarized results of in situ hybridization 78 .933 A a My it“ " . 50 pm ‘5' ,- Figure 4.11. In situ hybridization of the conj unctival sample from cat # 12. 50 pm Figure 4.12. In situ hybridization of the corneal sample from cat # 4. 79 4.4 Discussion Clinical signs of natural as well as experimentally induced FHV -1 infection can be quite severe.70’79 In the present study infected cats generally exhibited mild signs of upper respiratory and ocular disease. A possible explanation for the mild clinical signs compared to natural infection or experimental infection with other viral strains could be viral attenuation over time leading to a reduced pathogenicity of the C27 strain of FHV-l. Similar to observations by Bistner et al.12 I no clinical signs of keratitis were observed in any of the cats at any point in time during this primary infection. However, fluorescein staining and Rose Bengal staining of the cornea were not performed daily. Therefore very subtle lesions may not have been detected. Oral ulcers are reported to occur rarely in association with feline herpes virus 1 infection and are more commonly associated with feline calicivirus infectionm’106 Although the areas of oral ulceration noted were generally small and transient, 75% of inoculated cats were affected. It is possible those small ulcers are overlooked in clinical cases or that field strains do not induce development of oral ulceration. The oropharyngeal administration of 50 pl of viral suspension may also represent a greater viral inoculum than generally reaches those tissues during natural infection. At 30 days post-inoculation, latency should be established with resolution of clinical signs.304 Latency has been previously defined as the absence of detectable FHV-l as assessed by virus isolation, but detectable viral DNA using PCR assays.85 ’144 In this study, virus isolation was performed immediately following tissue harvest. Infectious virus was isolated from all ocular tissues and 50% of the ganglia on day 6 and day 10 post-inoculation. This is consistent with virus shedding as expected with active disease. On day 30 all tissue samples from 3 of the inoculated 80 cats were negative on V1. This is consistent with establishment of latency, which has previously been shown to occur by this time post inoculation.304 However, F HV-l could be isolated from the corneal sample of one cat. From the same cat FHV-l DNA could also be detected using quantitative real-time PCR in the cornea, conjunctiva, and both the ciliary and trigeminal ganglia. Although the cat did not show any clinical signs at the time of euthanasia, these results suggest the cat was either suffering a recrudescent infection or failed to clear the primary infection. A similar scenario has been noted in studies by Stiles et a1.162 and Townsend et al.198 in which F HV -1 DNA was detected in the corneas of clinically normal cats. In these studies virus isolation was not performed. However, in the study by Townsend et al.198 LATs could not be detected in the cornea, suggesting the infection was not latent. Histopathological examination revealed typical changes for the conjunctival tissues in all inoculated cats. However, glandular tissues from all lacrimal and nictitans glands were normal. These results do not support the suggestion by Nasisse4 and Nasisse et al.80 that lacrimal adenitis is responsible for FHV-l related keratoconjunctivitis sicca (KCS). As inflammatory changes were noted in the overlying conjunctiva, perhaps occlusion of the excretory ductules is the cause of FHV-l related KCS. Clinically none of the cats demonstrated clinical signs consistent with KCS. However, tear production was not evaluated in this study. Inoculation with a more virulent strain of F HV-l might cause KCS to develop and better elucidate the cause. Conjunctival tissue exhibiting changes consistent with conjunctivitis was noted in sections of the lacrimal and nictitans glands. Therefore, the results of the virus isolation for these tissues are suspect, since FHV-l could be isolated from conjunctiva during the acute phase. Unfortunately lacrimal tissue could not be 81 harvested without contamination from the overlying conjunctiva. For this reason, quantitative real-time PCR was not repeated for the glandular tissue samples with positive extraction controls. All trigeminal ganglia examined histopathologically during the acute infectious state showed signs of lymphoplasmacytic inflammation. However, only 1 trigeminal ganglion obtained from a cat on day 30 showed histomorphological changes; a drop out of ganglion cells, but no concurrent inflammation. These results suggest that the trigeminal ganglia developed transient inflammation during the acute infection. The absence of lymphocytes during the latent phase is very interesting. Current studies of HSV-1 in man suggest lymphocytes play a key role in preventing reactivation of the virus within neuronal tissues“0 In contrast to the inflammation seen within the acute phase trigeminal ganglia, no histomorphologic changes could be detected in any of the ciliary ganglia. This occurred despite a 50% isolation rate on V1 and a 100% detection rate on quantitative real time PCR during the acute phase of FHV-l infection. It is possible that localized inflammatory changes do not occur or they were missed on histopathologic examination due to the limited number of sections obtained from the ganglia. The F HV-l DNA copy numbers in corneal samples as determined by quantitative real-time PCR were similar in samples obtained on day 6 to samples collected on day 10 post-inoculation. Except for one cat’s corneal sample in which recrudescent or persistent infection is suspected, all other cats euthanized on day 30 had negative tissue samples. DNA copy numbers in conjunctival tissues tended to be lower on day 10 than on day 6. Therefore it is possible that the infection clears from the conjunctiva before it clears from the cornea. For the trigeminal ganglia a transient increase on day 10 was noted before the viral copy number decreased by day 30. A 82 similar phenomenon of detectable viral DNA increase on day 10 was noted in the ciliary ganglia. Therefore we would speculate that these ganglia reach their peak viral load later than the surface ocular tissues. In contrast to findings in a previous study300 F HV-l DNA copy number within the trigeminal ganglia during latency was only slightly lower than during active infection. In that study quantification was carried out using an automated electrochemiluminescence detector. The higher sensitivity of the quantitative real time PCR assay utilized in the present study could explain the difference in viral copy numbers between the two studies. However, our results are similar to those seen in murine models of HSV-1.3%336 On day 30 a low copy number of FHV-l DNA was detected only in the ciliary ganglion from the cat with a suspected recrudescent or persistent infection. All other ciliary ganglia were negative. From these data we cannot conclude that the ciliary ganglion is a site of latency for F HV-l in cats. Further investigation will be necessary. All cats euthanized on day 6 post-inoculation were positive on ISH within corneal and/or conjunctival tissues. In contrast none of the cats euthanized on day 10 post-inoculation had a clearly positive result in any of the tissues. The positive control samples processed at the same time demonstrated strong positive hybridization signals. Furthermore, these tissues were processed with other tissues in which positive hybridization signals were detected. These results are somewhat surprising as virus isolation for conjunctival and corneal samples was positive for all inoculated cats euthanized on days 6 and 10. Furthermore, viral load as determined by quantitative real time PCR was similar for ISH-positive and -negative cats euthanized on day 6 and 10. The strongest positive hybridization signal was detected within conjunctival epithelial cells from cat #12 euthanized on day 6 post-inoculation. This cat also had 83 the highest viral load within corneal and conjunctival tissue samples based upon the RT-PCR results. However, no positive signal was found within this cat’s cornea. FHV -1 positive cells were generally sparse in positive tissues. Therefore it would be possible to have examined sections that did not contain cells harboring sufficient virus to produce a detectable positive hybridization signal. Also, cats with severe conjunctivitis had necrotized or very attenuated epithelium. This leaves less intact epithelial cells in which to detect the virus. The overall sensitivity of the ISH to detect FHV-l utilizing this protocol was poor when compared to the sensitivity of the RT- PCR assay. A possible explanation could be the localized distribution of FHV-l infected cells within the tissues. No virus could be detected with this method within glandular tissue samples examined. This is consistent with the normal tissue morphology shown in standard histopathologic examination. However, F HV-l in low quantities could be present. Furthermore the clinical signs seen in this study were generally mild. Therefore it is possible that F HV-l infects lacrimal and nictitans glands in more severe infections. All tissues of cats euthanized on day 30 showed poor cellular morphology in the sections examined by ISH. The morphology of the same tissues was excellent for standard histology. This is difficult to explain, since these tissues were processed for the ISH in different runs together with other tissues that demonstrated good morphology. It could be possible that fixation of the tissues post euthanasia was either insufficient or excessive and the intense treatment during ISH led to the morphological changes noted. Repeating the ISH for these tissues with varying conditions could help to explain the changes found. In future studies, additional tissues such as uvea, retina, optic nerve, olfactory bulb, brainstem, cerebellum, cerebrum (visual cortex), optic chiasm, cranial cervical 84 ganglion, otic ganglion, pterygopalatine ganglion, mandibular salivary gland, submandibular lymph node, parotid salivary gland, parotid lymph node, nasal turbinates, tonsil, lung, trachea, oral fauces, and retropharyngeal lymph node could be examined to identify other targets of FHV-l during both the acute and latent stage of infection. Herpesvirinae have been detected within some of these tissues during studies of HSV-1 infection in murine models31 and pseudorabies virus infection in pigletsm‘338 The presence of virus within these tissues could expand our understanding of the pathology induced by FHV-l infection and the patterns of recrudescence noted clinically. Modulation of apoptosis may be a key factor allowing herpesvirinae to establish latency.339'340 However, the ability of FHV-l infection to influence cellular apoptosis has not been investigated. If FHV-l infection can block apoptosis of infected cells, this is a mechanism by which the host defenses are evaded. Apoptosis could be assayed by measuring caspase-3 activity. Caspase activation is thought to be the most specific indicator of apoptosis and caspase 3, the key converging enzyme is ubiquitously distributed making it an excellent choice for a marker of apoptosis.34' More sensitive methods to detect FHV-l could be utilized including in situ PCR or laser capture of individual cells for PCR analysis. Furthermore, a different FHV-l strain exhibiting higher virulence than C27 could be used for inoculation. This could also facilitate the detection rate and help to identify further targets of FHV-l. In conclusion, this study confirmed the ability of F HV-l to acutely infect not only conjunctival epithelium, corneal epithelium, and trigeminal ganglion, but also the ciliary ganglion. The ciliary ganglion may serve as an additional site of latency for FHV-l and could explain the preponderance of ocular lesions noted during recrudescent infections. However additional studies to prove this hypothesis are 85 required. The copy number within latent trigeminal ganglia was much higher than those previously reported due to the increased sensitivity of the assay utilized. The copy numbers reported are similar to those found for HSV-1 in murine models of infection. However, despite an initial lymphoplasmacytic inflammatory response and persistence of the virus, the ganglia appeared normal by day 30 post-inoculation. This lack of inflammation is in direct contrast with the current theory that lymphocytes are a key factor in regulation of the latent state. The theory that lacrimal adenitis contributes to the development of KCS was not supported as there was no evidence of FHV-l infection or secondary inflammation within the glandular tissues at any time. 86 APPENDICES 87 5. APPENDIX A USDA Scoring Method for FHV-la Clinical signs 1 Days I Score Fever 103.0 to 103.9 °F 1 each day 104.0 to 104.9 °F 2 each day 2105 °F 3 each day Conjunctivitis Serous discharge only 1 to 3 l a 2 Mucopurulent discharge 1 to 2 2 3 to 5 4 26 6 Rhinitis Serous discharge only 1 to 3 l a 2 Mucopurulent discharge 1 to 2 2 3 to 5 4 26 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 l to 2 3 2 4 Hypothermia <99 °F 2 each day Oral ulcers (linguinal or oral mucosa) 1 ulcer <4 mm 1 to 5 2 6 to 9 3 210 4 Multiple ulcers <4 mm 1 to 4 3 5 to 8 5 2 7 Ulcer or ulcers >4 mm 1 to 4 5 5 to 8 7 3 9 Salivating 1 each day Extenal ulcers (lip or nares) Nonbleeding ulcer 4 Bleeding ulcer 6 Death 15 a US. Department of Agriculture, Animal and Plant Health Inspection Service, National Animal Veterinary Services Laboratory (1985). Table 5.1. USDA Scoring Method for FHV-l 88 6. APPENDIX B sneezing ca'123456789 15 14 13 3 11 10 (O .s .rrorurburmuoo Figure 6.1. Results of the clinical examination: sneezing. elevated body temperature C31 15 14 13 3 11 10 123456789 (D a ANNAO‘IO’VQ Figure 6.2. Results of the clinical examination: elevated body temperature. 89 dehydration can123456789 15 14 13 3 11 1o (.0 .a ANN-“mafia Figure 6.3. Results of the clinical examination: dehydration. anorexia cat 15 14 13 3 11 10 9 123456789 .3 ANNAU’IO’NQ Figure 6.4. Results of the clinical examination: anorexia. 90 ptyalism cat123456789 15 14 13 3 11 10 ID .5 ANNAUIQVQ Figure 6.5. Results of the clinical examination: ptyalism. oral ulceration cat 15 14 13 3 11 10 123456789 (D A ANNAU‘IU’VQ Figure 6.6. Results of the clinical examination: oral ulceration. 91 nasal discharge cat 15 14 13 3 11 10 9 123456789 —I gumbmmwm Figure 6.7. Results of the clinical examination: nasal discharge. blepharospasm W 1 2 3 4 5 6 7 8 9 15 14 13 3 11 1o (.0 _I ANN‘U‘O’NO’ Figure 6.8. Results of the clinical examination: blepharospasm. 92 ocular discharge cat1234567 9 15 14 13 3 11 1o 9 d ANN‘U’IU’NO Figure 6.9. Results of the clinical examination: ocular discharge. conjunctivitis cat123456789 15 14 13 3 11 1o 9 .n ANNAUIOINQ Figure 6.10. Results of the clinical examination: conjunctivitis. 93 7. APPENDIX C DAY 6 Sample ControlS Cat4 Cat 1 Cat2 Cat 12 — + + Trigeminal Ganglia Ciliary Ganglia — Lacrimal Gland — Cornea — ++++ Conjunctiva — Nictitating Gland — + +++++ ++++ ++++ Table 7.1. Virus isolation day 6 post inoculation DAY 10 Sample Control 10 Cat6 Cat7 Cat8 Cat9 Trigeminal Ganglia — + + _ _ Ciliary Ganglia — Lacrimal Gland — Cornea — ++++ Conjunctiva — Nictitating Gland — + + + + + ++++ ++++ + Table 7.2. Virus isolation day 10 post inoculation DAY 30 Sample Control 15 Cat 3 Cat 11 Cat 13 Cat 14 Trigeminal Ganglia — — _ _ _ Ciliary Ganglia — — _ _ _ Lacrimal Gland — — _ ._ _ Cornea - _ + _ _ Conjunctiva — _ _ _ _ Nictitating Gland — — _ .. _ Table 7.3. Virus isolation day 30 post inoculation 94 8. APPENDIX D Cat # Repeat#1 Repeat#2 AveragL Std Dev ML 1 172,000 224,000 198,000 36,770 2 1,370,000 1,510,000 1,440,000 98,995 4 56,500 86,900 71,700 21,496 12 269,000 384,000 326,500 81,317 5 O O 0 0 DAY 10 P1. 6 226,000 298,000 262,000 50,912 7 73,400 74,500 73,950 778 8 177,000 223,000 200,000 32,527 9 189,000 247,000 218,000 41,012 10 O 0 0 0 DAY 30 Pl. 3 O 0 0 0 1 1 22,600 28,000 25,300 3,818 13 0 O 0 0 14 0 0 0 0 15 O O O 0 Table 8.]. PCR results of cornea (Copy no. / 100 ng DNA) 95 Cat # Repeat#1 Repeat#2 Average Std Dev DAY 6 P1. 1 125,000 174,000 149,500 34,648 2 610,000 648,000 629,000 26,870 4 434,000 317,000 375,500 82,731 12 2,510,000 2,410,000 2,460,000 70,71 1 5 0 O O 0 DAY 10 Pl. 6 4,200 5,510 4,855 926 7 619,000 493,000 556,000 89,095 8 16,500 19,600 18,050 2,192 9 27,500 29,500 28,500 1,414 10 O O O 0 DAY 30 P1. 3 0 O 0 O 11 105 122 114 12 13 O O O 14 5 14 9 6 15 0 O 0 O Table 8.2. PCR results of conjunctiva (Copy no. / 100 ng DNA) 96 Table 8.3. PCR results of lacrimal gland (Copy no. / 100 ng DNA) Marked fields: contamination of sample, assay not repeated. 97 Table 8.4. PCR results of nictitans gland (Copy no. / 100 ng DNA) Marked fields: contamination of sample, assay not repeated. 98 Cat # Repeat#1 Remain Average Std Dev DAY 6 P1. 1 16,500 16,200 16,350 212 2 12,200 13,000 12,600 566 4 14,100 16,700 15,400 1,838 12 10,800 10,600 10,700 141 5 0 0 O 0 DAY 10 P1. 6 17,500 31,800 24,650 10,1 12 7 57,000 37,700 47,350 13,647 8 13,400 13,200 13,300 141 9 50,400 52,400 51,400 1,414 10 0 0 0 0 DAY 30 P1. 3 15,900 34,200 25,050 12,940 11 3,110 3,130 3,120 14 13 5,370 5,790 5,580 297 14 13,500 16,100 14,800 1,838 15 0 0 0 O Table 8.5. PCR results of trigeminal ganglia (Copy no. / 100 ng DNA) 99 Cat # Repeat#l Repeat#2 Average Std Dev DAY 6 P1. 1 239 192 216 33 2.440 1.630 2.035 573 4 5.660 3.230 4.445 1.718 12 149 65 107 59 5 0 0 0 0 DAY 10 P1. 6 5.130 4.560 4.845 403 7 26.000 47.600 36.800 15.274 8 685 512 599 122 9 410 364 387 33 10 O O 0 0 DAY 30 P1. 3 0 0 0 0 11 100 115 108 11 13 O 0 l4 0 O 15 0 0 0 0 Table 8.6. PCR results of ciliary ganglia (Copy no. / 20 ng DNA) 100 10. 11. 12. 13. 9. BIBLIOGRAPHY Flint SJ, Enquist LW, Racaniello VR, et al. Principles of virology. 2nd ed. Washington, DC: ASM Press, 2004. Roizrnan B, Carmichael LE, Deinhardt F, et a1. Herpesviridae. Definition, provisional nomenclature, and taxonomy. The Herpesvirus Study Group, the International Committee on Taxonomy of Viruses. Intervirology 1981;16:201- 217. Roizrnan B. The family Herpesviridae: general description, taxonomy, and classification In: B. R, ed. The herpesviruses. New York: NY: Plenum Press, 1982;pp. 1—23. Nasisse MP. Feline herpesvirus ocular disease. Vet Clin North Am Small Anim Pract 1990;20:667-680. Davison AJ, Seberle R, Hayward GS, et al. Herpesviridae In: Fauquet CM, Mayo MA, Maniloff J, et al., eds. Virus Taxonomy: Eighth report of the International Committee on Taxonomy of Viruses. San Diego: Elsevier, 2005;193-212. Rolle M, Mayr A, Gedek B, et al. Medizinische Mikrobiologie, Infections- and Seuchenlehre. 6 ed, 1993. Murphy F, Gibbs P, Horzinek M, et al. Veterinary Virology. 3 ed: San Diego: Academic Press, 1999. Martina BE, Airikkala MI, Harder TC, et al. A candidate phocid herpesvirus vaccine that provides protection against feline herpesvirus infection. Vaccine 2001;20:943-948. Harder TC, Harder M, Vos H, et al. Characterization of phocid herpesvirus—1 and -2 as putative alpha- and gammaherpesviruses of North American and European pinnipeds. J Gen Virol 1996;77 (Pt l):27-35. Harder TC, Harder M, de Swart RL, et al. Major immunogenic proteins of phocid herpes-viruses and their relationships to proteins of canine and feline herpesviruses. Vet Q 1998;20:50-55. Harder TC, Osterhaus AD. Molecular characterization and baculovirus expression of the glycoprotein B of a seal herpesvirus (phocid herpesvirus-1). Virology 1997;227:343-352. Lebich M, Harder TC, Frey HR, et a1. Comparative immunological characterization of type—specific and conserved B-cell epitopes of pinniped, felid and canid herpesviruses. Arch Virol 1994;136:335-347. Martina BE, Harder TC, Osterhaus AD. Genetic characterization of the unique short segment of phocid herpesvirus type 1 reveals close relationships among 101 14. 15. l6. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. alphaherpesviruses of hosts of the order Camivora. J Gen Virol 2003;84:1427- 1430. Martina BE, van de Bildt MW, Kuiken T, et a1. Immunogenicity and efficacy of recombinant subunit vaccines against phocid herpesvirus type 1. Vaccine 2003;21:2433-2440. Willoughby K, Bennett M, Williams RA, et al. Sequences of the ribonucleotide reductase-encoding genes of felid herpesvirus 1 and molecular phylogenetic analysis. Virus Genes 1997;15:203-218. Willoughby K, Bennett M, McCracken CM, et al. Molecular phylogenetic analysis of felid herpesvirus 1. Vet Microbiol 1999;69:93-97. Maeda K, Horimoto T, Mikami T. Properties and functions of feline herpesvirus type 1 glycoproteins. J Vet Med Sci 1998;60:881-888. Rota PA, Maes RK, Ruyechan WT. Physical characterization of the genome of feline herpesvirus-1. Virology 1986;154:168-179. Grail A, Harbour DA, Chia W. Restriction endonuclease mapping of the genome of feline herpesvirus type 1. Arch Virol 1991;116:209-220. Roizman B, Pellet P. The family of herpesviridae In: Fields BN, Howler P, Griffin DE, et al., eds. Fields’ Virology. 4 ed. Philadelphia: Lippincott Williams & Williams, 2001 ;2381-2385. Crandell RA, Maurer FD. Isolation of a feline virus associated with intranuclear inclusion bodies. Proc Soc Exp Biol Med 1958;97:487-490. Maeda K, Horimoto T, Norimine J, et al. Identification and nucleotide sequence of a gene in feline herpesvirus type 1 homologous to the herpes simplex virus gene encoding the glycoprotein B. Arch Virol 1992;127:387-397. Sodeik B, Ebersold MW, Helenius A. Microtubule-mediated transport of incoming herpes simplex virus 1 capsids to the nucleus. J Cell Biol 1997;136:1007-1021. Fenwick M, Roizman B. Regulation of herpesvirus macromolecular synthesis. VI. Synthesis and modification of viral polypeptides in enucleated cells. J Virol 1977;22:720-725. Fenwick ML, Owen SA. On the control of immediate early (alpha) mRNA survival in cells infected with herpes simplex virus. J Gen Virol 1988;69 (Pt ll):2869-2877. Roizman B, Knipe DM. Herpes simplex viruses and their replication In: Fields BN, Howler P, Griffin DE, et al., eds. Field is Virology. 4 ed. Philadelphia: Lippincott Williams & Williams, 2001;2399-2443. Crandell RA. Feline viral rhinotracheitis (F VR). Adv Vet Sci Comp Med 1973;17:201-224. 102 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. Crandell RA, Despeaux EW. Cytopathology of feline viral rhinotracheitis virus in tissue cultures of feline renal cells. Proc Soc Exp Biol Med 1959; 101 :494-497. Crandell RA, F abricant CG, Nelson-Rees WA. Development, characterization, and viral susceptibility of a feline (Felis catus) renal cell line (CRF K). In Vitro l973;9:176-185. Jones C. Alphaherpesvirus latency: its role in disease and survival of the virus in nature. Adv Virus Res 1998;51:81-133. Labetoulle M, Maillet S, Efstathiou S, et al. HSVl latency sites after inoculation in the lip: assessment of their localization and connections to the eye. Invest Ophthalmol Vis Sci 2003;44:217-225. Baringer JR, Swoveland P. Recovery of herpes-simplex virus from human trigeminal ganglions. N Engl J Med 1973;288:648-650. Bastian FO, Rabson AS, Yee CL, et al. Herpesvirus hominis: isolation from human trigeminal ganglion. Science 1972;178:306-307. Rock DL. The molecular basis of latent infections by alphaherpesviruses. Semin Virol 1993;42157-165. Rock DL. Latent infection with bovine herpesvirus type 1. Sem Virol 1994;5z233-240. Rock DL, Nesbum AB, Ghiasi H, et al. Detection of latency-related viral RNAs in trigeminal ganglia of rabbits latently infected with herpes simplex virus type 1. J Virol 1987;61:3820-3826. Stevens JG, Wagner EK, Devi-Rao GB, et al. RNA complementary to a herpesvirus alpha gene mRNA is prominent in latently infected neurons. Science 1987;235:1056-1059. Borchers K, Wolfinger U, Ludwig H. Latency-associated transcripts of equine herpesvirus type 4 in trigeminal ganglia of naturally infected horses. J Gen Virol 1999;80 (Pt 8):2165-2171. Dobson AT, Sederati F, Devi-Rao G, et al. Identification of the latency- associated transcript promoter by expression of rabbit beta-globin mRN A in mouse sensory nerve ganglia latently infected with a recombinant herpes simplex virus. J Virol 1989;63:3844-3851. Zwaagstra JC, Ghiasi H, Slanina SM, et al. Activity of herpes simplex virus type 1 latency-associated transcript (LAT) promoter in neuron-derived cells: evidence for neuron specificity and for a large LAT transcript. J Virol 1990;64:5019-5028. Steiner I, Spivack JG, O'Boyle DR, 2nd, et al. Latent herpes simplex virus type 1 transcription in human trigeminal ganglia. J Virol 1988;62:3493-3496. 103 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. Sawtell NM, Thompson RL. Herpes simplex virus type 1 latency-associated transcription unit promotes anatomical site-dependent establishment and reactivation from latency. J Virol 1992;66:2157-2169. Thompson RL, Sawtell NM. The herpes simplex virus type 1 latency- associated transcript gene regulates the establishment of latency. J Virol 1997;71:5432-5440. Chen SH, Kramer MF, Schaffer PA, et al. A viral function represses accumulation of transcripts from productive-cycle genes in mouse ganglia latently infected with herpes simplex virus. J Virol 1997;71:5878-5884. Garber DA, Schaffer PA, Knipe DM. A LAT-associated function reduces productive-cycle gene expression during acute infection of murine sensory neurons with herpes simplex virus type 1. J Virol 1997;71:5885-5893. Steiner I, Spivack JG, Lirette RP, et al. Herpes simplex virus type 1 latency- associated transcripts are evidently not essential for latent infection. Embo J 1989;8:505-511. Hill JM, Sedarati F, Javier RT, et al. Herpes simplex virus latent phase transcription facilitates in vivo reactivation. Virology 1990;174:117-125. Pemg GC, Dunkel EC, Geary PA, et al. The latency-associated transcript gene of herpes simplex virus type 1 (HSV-1) is required for efficient in vivo spontaneous reactivation of HSV-1 from latency. J Virol 1994;68:8045-8055. Block TM, Deshmane S, Masonis J, et al. An HSV LAT null mutant reactivates slowly from latent infection and makes small plaques on CV-l monolayers. Virology 1993;192:618-630. Devi-Rao GB, Bloom DC, Stevens JG, et al. Herpes simplex virus type 1 DNA replication and gene expression during explant-induced reactivation of latently infected murine sensory ganglia. J Virol 1994;68: 1271-1282. Leib DA, Bogard CL, Kosz-Vnenchak M, et al. A deletion mutant of the latency-associated transcript of herpes simplex virus type 1 reactivates from the latent state with reduced frequency. J Virol 1989;63:2893-2900. Pemg GC, Slanina SM, Ghiasi H, et al. The effect of latency-associated transcript on the herpes simplex virus type 1 latency-reactivation phenotype is mouse strain-dependent. J Gen Vir012001;82:1117-1122. Pemg GC, Jones C, Ciacci-Zanella J, et al. Virus-induced neuronal apoptosis blocked by the herpes simplex virus latency-associated transcript. Science 2000;287:1500-1503. Branco F J , Fraser NW. Herpes simplex virus type 1 latency-associated transcript expression protects trigeminal ganglion neurons from apoptosis. J Virol 2005;79:9019-9025. 104 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. Gupta A, Gartner JJ, Sethupathy P, et al. Anti-apoptotic function of a microRNA encoded by the HSV-1 latency-associated transcript. Nature 2006;442:82-85. J in L, Pemg GC, Mott KR, et al. A herpes simplex virus type 1 mutant expressing a baculovirus inhibitor of apoptosis gene in place of latency- associated transcript has a wild-type reactivation phenotype in the mouse. J Viral 2005;79:12286-12295. Inman M, Pemg GC, Henderson G, et al. Region of herpes simplex virus type 1 latency-associated transcript sufficient for wild-type spontaneous reactivation promotes cell survival in tissue culture. J Viral 2001;75:3636- 3646. Verjans GM, Hintzen RQ, van Dun JM, et al. Selective retention of herpes simplex virus-specific T cells in latently infected human trigeminal ganglia. Proc Natl Acad Sci U S A 2007;104:3496-3501. Theil D, Derfuss T, Paripovic I, et al. Latent herpesvirus infection in human trigeminal ganglia causes chronic immune response. Am J Pathol 2003;163:2179-2184. Hufner K, Derfuss T, Herberger S, et a1. Latency of alpha-herpes viruses is accompanied by a chronic inflammation in human trigeminal ganglia but not in dorsal root ganglia. J Neuropathol Exp Neural 2006;65:1022-1030. Crandell RA, Rehkemper J A, Niemann WH, et al. Experimental feline viral rhinotracheitis. J Chronic Dis 1961 ;138: 191-196. Ellis TM. Feline respiratory virus carriers in clinically healthy cats. Aust Vet J 1981;57:115-118. Studdert MJ, Martin MC. Virus diseases of the respiratory tact of cats. 1. Isolation of feline rhinotracheitis virus. Aust Vet J 1970;46:99-104. Maggs DJ, Lappin MR, Reif J S, et al. Evaluation of serologic and viral detection methods for diagnosing feline herpesvirus-1 infection in cats with acute respiratory tract or chronic ocular disease. J Am Vet Med Assoc 1999;214:502-507. Gaskell RM, Povey RC. Experimental induction of feline viral rhinotracheitis virus re-excretion in F VR-recovered cats. Vet Rec 1977; 100: 128-133. Gaskell R, Dawson S, Radford A, et al. Feline herpesvirus. Vet Res 2007;38:337-354. Gaskell RM, Povey RC. Re-excretion of feline viral rhinotracheitis virus following corticosteroid treatment. Vet Rec 1973;93:204-205. Povey RC, Johnson RH. A survey of feline viral rhinotracheitis and feline picomavirus infection in Britain. J Small Anim Pract 1971;12:233-247. 105 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. Gaskell RM, Radford AD, Dawson S. Feline infectious respiratory disease In: Chandler EA, Gaskell CJ,Gaskell RM, eds. Feline medicine and therapeutics: Blackwell Publishing, 2004;577-595. Stiles J. Feline herpesvirus. Vet Clin North Am Small Anim Pract 2000;30:1001-1014. Povey RC. Feline respiratory infections--a clinical review. Can Vet J 1976;17:93-100. Povey RC. A review of feline viral rhinotracheitis (feline herpesvirus I infection). Comp Immunol Microbiol Infect Dis l979;2:373-3 87. Hartrnann K. Virale lfenctionskrankheiten In: Kraft W,Hirschberger J, eds. Kleintierkrankheiten. 3 ed. Stuttgart: Eugen Ulmer GmbH & Co, 2000;] 19- 126. Wardley RC, Povey RC. The pathology and sites of persistence associated with three different strains of feline calicivirus. Res Vet Sci 1977;23:15-19. Maggs DJ. Update on pathogenesis, diagnosis, and treatment of feline herpesvirus type 1. Clin Tech Small Anim Pract 2005;20:94-101. Storey ES, Gerding PA, Scherba G, et al. Survival of equine herpesvirus-4, feline herpesvirus-l, and feline calicivirus in multidose ophthalmic solutions. Vet Ophthalmol 2002;5I263-267. Nasisse MP, Guy J S, Stevens JB, et al. Clinical and laboratory findings in chronic conjunctivitis in cats: 91 cases (1983-1991). J Am Vet Med Assoc 1993;203:834-837. Gaskell RM, Povey RC. Feline viral rhinotracheitis: sites of virus replication and persistence in acutely and persistently infected cats. Res Vet Sci 1979;27:167-174. Hoover EA, Rohovsky MW, Griesemer RA. Experimental feline viral rhinotracheitis in the germfree cat. Am J Pathol 1970;58:269-282. Nasisse MP, Guy J S, Davidson MG, et al. Experimental ocular herpesvirus infection in the cat. Sites of virus replication, clinical features and effects of corticosteroid administration. Invest Ophthalmol Vis Sci 1989;30:1758-1768. Tham KM, Studdert MJ. Clinical and immunological responses of cats to feline herpesvirus type 1 infection. Vet Rec 1987;120:321-326. Shields RP, Gaskin JM. Fatal generalized feline viral rhinotracheitis in a young adult cat. J Am Vet Med Assoc 1977;170:439-441. Spradbrow PB, Carlisle C, Watt DA. The association of a herpesvirus with generalised disease in a kitten. Vet Rec 1971;89:542-544. 106 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. 96. 97. Vogtlin A, Fraefel C, Albini S, et a]. Quantification of feline herpesvirus 1 DNA in ocular fluid samples of clinically diseased cats by real-time TaqMan PCR. J Clin Microbiol 2002;40:519-523. Weigler BJ, Babineau CA, Sherry B, et al. High sensitivity polymerase chain reaction assay for active and latent feline herpesvirus-l infections in domestic cats. Vet Rec 1997;140:335-338. Hoover EA, Griesemer RA. Bone lesions produced by feline herpesvirus. Lab Invest 1971;25:457-464. Bittle JL, Peckham JC. Genital infection induced by feline rhinotracheitis virus and effects on newborn kittens. J Am Vet Med Assoc 1971;158:Suppl 2:927- 928. Hoover EA, Griesemer RA. Experimental feline herpesvirus infection in the pregnant cat. Am J Pathol 1971;65:173-188. Maeda K, Yokoyama N, F uj ita K, et al. Heparin-binding activity of feline herpesvirus type 1 glycoproteins. Virus Res 1997;52:169-176. Thompson NL, Sabine M, Hyne RH. Herpesviruses isolated from Cheetahs. Aust VetJ 1971;47:458. Scherba G, Hajjar AM, Pemikoff DS, et al. Comparison of a cheetah herpesvirus isolate to feline herpesvirus type 1. Arch Virol 1988;100:89-97. Herrmann SC, Gaskell RM, Ehlers B, et al. Characterization of the feline herpesvirus genome and molecular epidemiology of isolates from natural outbreaks and latent infections. In: Wittman G, Gaskell RM,Rziha HJ, eds. Latent herpesvirus infections in veterinary medicine. Boston, MA: Nijhoff, 1984;321-336. Boever WJ, McDonald S, Solorzano RF. Feline viral rhinotracheitis in a colony of clouded leopards. Vet Med Small Anim Clin 1977;72:1859-1866. Truyen U, Stockhofe-Zurwieden N, Kaaden OR, et al. A case report: encephalitis in lions. Pathological and virological findings. Dtsch T ierarztl Wachenschr 1990;97:89-91 . Spencer JA. Survey of antibodies to feline viruses in free-ranging lions. South- Afi'ican Journal of Wildlife Research 1991;21:59-61. Spencer JA, Morkel P. Serological survey of sera from lions in Etosha National Park. South-Afr T ydskr Natuurnav 1993;23 :60-61. Hofrnann-Lehmann R, F ehr D, Grob M, et al. Prevalence of antibodies to feline parvovirus, calicivirus, herpesvirus, coronavirus, and immunodeficiency virus and of feline leukemia virus antigen and the interrelationship of these viral infections in free-ranging lions in east Afiica. Clin Diagn Lab Immunol 1996;32554-562. 107 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. ll]. Eberle R, Baldwin CJ, Black D, et al. Feline herpesvirus infections in Bobcats (Lynx Rufus): Disease in experimentally inoculated animals. Journal of Zoo and Wildlife Medicine 1991;22:175-183. Paul-Murphy J, Work T, Hunter D, et al. Serologic survey and serum biochemical reference ranges of the free-ranging mountain lion (Felis concolor) in California. J Wildl Dis 1994;30:205-215. Pratelli A, Tempesta M, De Palma MG, et a]. Restriction endonuclease analysis of feline herpesvirus 1 DNA isolated from wild felids. Vet Rec 1999;144:537-538. - Nakamura K, Ikeda Y, Miyazawa T, et a]. Comparison of prevalence of feline herpesvirus type 1, calicivirus and parvovirus infections in domestic and leopard cats in Vietnam. J Vet Med Sci 1999;61:1313-1315. Evermann JF, McKeiman AJ, Ott RL, et al. Diarrhea] condition in dogs associated with viruses antigenically related to feline herpesvirus. Cornell Vet 1982;72:285-291. Rota PA, Maes RK, Evermann JF. Biochemical and antigenic characterization of feline herpesvirus-l-like isolates from dogs. Arch Virol 1986;89:57-68. Kramer JW, Evermann JF, Leathers CW, et al. Experimental infection of two dogs with a canine isolate of feline herpesvirus type 1. Vet Pathol 1991;28:338-340. Harrison TM, Mazet JK, Holekamp KE, et a1. Antibodies to canine and feline viruses in spotted hyenas (Crocuta crocuta) in the Masai Mara National Reserve. J Wildl Dis 2004;40:1-10. Wolf A. Other feline viral diseases In: Ettinger SJ,Feldmann EC, eds. Textbook of veterinary internal medicine: disease of the dog and cat. 5 ed. Philadelphia: W.B. Saunders Company, 2000;445-448. Love DN. Feline herpesvirus associated with interstitial pneumonia in a kitten. Vet Rec 1971;89:178-181. Welsh RD. Bordetella bronchiseptica infections in cats. J Am Anim Hosp Assoc 1996;32:153-158. Gaskell RM, Wardley RC. Feline viral respiratory disease: a review with particular reference to its epizootiology and control. J Small Anim Pract 1978;19:1-16. Hickman MA, Reubel GH, Hoffman DE, et al. An epizootic of feline herpesvirus, type 1 in a large specific pathogen-free cat colony and attempts to eradicate the infection by identification and culling of carriers. Lab Anim 1994;28:320-329. F lecknell PA, Orr CM, Wright AI, et a]. Skin ulceration associated with herpesvirus infection in cats. Vet Rec 1979;104:313-315. 108 112. 113. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. Johnson RP, Sabine M. The isolation of herpesviruses from skin ulcers in domestic cats. Vet Rec 1971;89:360-362. Junge RE, Miller RE, Boever WJ, et al. Persistent cutaneous ulcers associated with feline herpesvirus type 1 infection in a cheetah. J Am Vet Med Assoc 1991;198:1057-1058. Munson L, Wack R, Duncan M, et al. Chronic eosinophilic dermatitis associated with persistent feline herpes virus infection in cheetahs (Acinonyx jubatus). Vet Pathol 2004;41:170-176. Hargis AM, Ginn PE, Mansell JEKL, et al. Ulcerative facial and nasal dermatitis and stomatitis in cats associated with feline herpesvirus 1. Veterinary Dermatology 1999;10:267-274. Hargis AM, Ginn PE. Feline herpesvirus l-associated facial and nasal dermatitis and stomatitis in domestic cats. Vet Clin North Am Small Anim Pract 1999;29:1281-1290. Holland JL, Outerbridge CA, Affolter VK, et a1. Detection of feline herpesvirus 1 DNA in skin biopsy specimens from cats with or without dermatitis. J Am Vet Med Assoc 2006;229:1442-1446. Whitley RD, Whitley EM, McLaughlin SA. Diagnosing And Treating Disorders Of The Feline Conjunctiva And Cornea. Veterinary Medicine l993;88:1138-&. Glaze M, Gelatt K. Feline ophthalmology In: Gelatt K, ed. Veterinary Ophthalmology. 3rd edn. ed. Philadelphia: Lippincott Williams & Wilkins, 1999;997—1052. Andrew SE. Ocular manifestations of feline herpesvirus. J Feline Med Surg 2001;3z9-l6. Bistner SI, Carlson JH, Shively JN, et a]. Ocular manifestations of feline herpesvirus infection. J Am Vet Med Assoc 1971;159:1223-1237. Horvath Z, Bartha A, Papp L, et al. On feline rhinotracheitis. Acta Vet Acad Sci Hung 1965;15:417-420. Gaskell RM, Dawson S. Feline respiratory disease In: Greene CE, ed. Infectious disease of the dog and cat. 2 ed. Philadelphia: W.B. Saunders, 1998;97-106. Whitley RD, Moore CP. Advances In Feline Ophthalmology. Veterinary Clinics OfNarth America-Small Animal Practice 1984;14: 1271-1288. Nasisse MP. Diagnosis and treatment of conjunctiva] and corneal diseases. Vet Q 1995;17 Suppl l:S42-43. Nasisse MP, Weigler B]. The diagnosis of ocular feline herpesvirus infection. Veterinary and Comparative Ophthalmology 1997;7z44-51. 109 127. 128. 129. 130. 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. 141. Roberts SR, Dawson CR, Coleman V, et al. Dendritic keratitis in a cat. J Am Vet Med Assoc 1972;161:285-289. Liesegang TJ. Classification of herpes simplex virus keratitis and anterior uveitis. Cornea 1999;18:127-143. Pepose J S, Nestor MS, Gardner KM, et a]. Composition of cellular infiltrates in rejected human corneal allografts. Graefes Arch Clin Exp Ophthalmol 1985;222:128-133. Youinou P, Colin J, Mottier D. Immunological analysis of the cornea in herpetic stromal keratitis. J Clin Lab Immunol 1985;17:105-106. Youinou P, Colin J, Ferec C. Monoclonal antibody analysis of blood and cornea T lymphocyte subpopulations in herpes simplex keratitis. Graefes Arch Clin Exp Ophthalmol 1986;224:131-133. Miller JK, Laycock KA, Nash MM, et al. Corneal Langerhans cell dynamics after herpes simplex virus reactivation. Invest Ophthalmol Vis Sci 1993;34:2282-2290. Streilein JW, Dana MR, Ksander BR. Immunity causing blindness: five different paths to herpes stromal keratitis. Immunol Today 1997;18:443-449. Meyers-Elliott RH, Pettit TH, Maxwell WA. Viral antigens in the immune ring of Herpes simplex stromal keratitis. Arch Ophthalmol 1980;98:897-904. Carr DJ, Harle P, Gebhardt BM. The immune response to ocular herpes simplex virus type 1 infection. Exp Biol Med (Maywood) 2001;226:353-366. Verjans GM, Remeijer L, van Binnendijk RS, et al. Identification and characterization of herpes simplex virus-specific CD4+ T cells in corneas of herpetic stromal keratitis patients. J Infect Dis 1998; 177:484-488. Koelle DM, Reymond SN, Chen H, et a]. Tegument-specific, virus-reactive CD4 T cells localize to the cornea in herpes simplex virus interstitial keratitis in humans. J Viral 2000;74:10930-10938. Kaye S, Choudhary A. Herpes simplex keratitis. Prag Retin Eye Res 2006;25:355-380. Deshpande S, Banerjee K, Biswas PS, et al. Herpetic eye disease: immunopathogenesis and therapeutic measures. Expert Rev Mol Med 2004;621-14. Zheng M, Deshpande S, Lee S, et al. Contribution of vascular endothelial growth factor in the neovascularization process during the pathogenesis of herpetic stromal keratitis. J Viral 2001;75:9828-9835. Yang YN, Bauer D, Wasmuth S, et al. Matrix metalloproteinases (MMP-2 and 9) and tissue inhibitors of matrix metalloproteinases (TIMP-1 and 2) during 110 142. 143. 144. 145. 146. 147. 148. 149. 150. 151. 152. 153. the course of experimental necrotizing herpetic keratitis. Exp Eye Res 2003;77:227-237. Lee S, Zheng M, Kim B, et al. Role of matrix metalloproteinase-9 in angiogenesis caused by ocular infection with herpes simplex virus. J Clin Invest 2002;110:1105-1111. Nasisse MP, English RV, Tompkins MB, et al. Immunologic, histologic, and virologic features of herpesvirus-induced stromal keratitis in cats. Am J Vet Res 1995;56:51-55. Reubel GH, Ramos RA, Hickman MA, et al. Detection of active and latent feline herpesvirus 1 infections using the polymerase chain reaction. Arch Virol 1993; 132:409-420. Kaufman HE, Brown DC, Ellison ED. Herpes virus in the lacrimal gland, conjunctiva and cornea of man--a chronic infection. Am J Ophthalmol 1968;65:32-35. Foster W], Jr., Kraus MD, Custer PL. Herpes simplex virus dacryoadenitis in an immunocompromised patient. Arch Ophthalmol 2003;121:911-913. Lim CC, Cullen CL. Schirmer tear test values and tear film break-up times in cats with conjunctivitis. Vet Ophthalmol 2005;82305-310. Lim CC, Reilly CM, Thomasy SM, et al. Effects on Tear film Dynamics of experimental feline herpesvirus infection. American College of Veterinary Ophthalmologists Annual Conference 2007;46. Startup FG. Corneal Necrosis And Sequestration In The Cat - A Review And Record Of 100 Cases. Journal Of Small Animal Practice 1988;29:476-486. Martin CL. Feline ophthalmologic diseases. Mod Vet Pract 1982;63:385-390. Lightowler C, Briihl-day R, HD H. Feline cornea] sequestrum. Feline Practice 1987;17:9—12. Bellhom R. Sequestrum of the feline cornea. Proceedings of the American College of Veterinary Ophthalmologists 1970:14—22. Gelatt KN, Peiffer L, Stevens J. Chronic ulcerative keratitis and sequestrum in the domestic cat. Journal of the American Animal Hospital Association 1973;92204— 154. 155. 213. Featherstone HJ, Franklin VJ, Sansom J. Feline corneal sequestrum: laboratory analysis of ocular samples from 12 cats. Veterinary Ophthalmology 2004;72229-238. Andrew SE, Tou S, Brooks DE. Comeoconjunctival transposition for the treatment of feline corneal sequestra: a retrospective study of 17 cases (1990- 1998). Vet Ophthalmol 2001;4:107—111. 111 156. 157. 158. 159. 160. 161. 162. 163. 164. 165. 166. 167. 168. 169. 170. Knecht CD, Schiller AG, Small E. Focal degeneration of the cornea with sequestration in a cat. J Am Vet Med Assoc 1966;149:1192-1193. Pentlarge V. Corneal sequestration in cats. Compendium of Continuing Education for the Practicing Veterinarian 1989;11:24—32. Morgan RV. Feline Corneal Sequestration - A Retrospective Study Of 42 Cases (1987-1991). Journal Of The American Animal Hospital Association 1994;30:24-28. Featherstone HJ, Sansom J. Feline corneal sequestra: a review of 64 cases (80 eyes) from 1993 to 2000. Veterinary Ophthalmology 2004;72213-227. Vawer GD. Corneal mummification in colourpoint cats. Vet Rec 1981;109:413. Souri E. The feline corneal nigrum. Vet Med Small Anim Clin 1975;70:531- 534. Stiles J, McDermott M, Bigsby D, et al. Use of nested polymerase chain reaction to identify feline herpesvirus in ocular tissue from clinically normal cats and cats with corneal sequestra or conjunctivitis. Am J Vet Res 1997;58:338-342. Nasisse MP, Glover TL, Moore CP, et al. Detection of feline herpesvirus 1 DNA in corneas of cats with eosinophilic keratitis or corneal sequestration. Am J Vet Res 1998;59:856-858. Morgan RV, Abrams KL, Kern TJ. Feline eosinophilic keratitis: a retrospective study of 54 cases: (1989-1994). Veterinary and Comparative Ophthalmology 1996;6zl3 l-134. Allgoewer I, Schaffer EH, Stockhaus C, et al. Feline eosinophilic conjunctivitis. Vet Ophthalmol 2001;4:69-74. Prasse KW, Winston SM. Cytology and histology of feline eosinophilic keratitis. Veterinary and Comparative Ophthalmology 1996;6z74-81. Van der Lelij A, Ooijman FM, Kijlstra A, et al. Anterior uveitis with sectoral iris atrophy in the absence of keratitis: a distinct clinical entity among herpetic eye diseases. Ophthalmology 2000;107:1164-1170. Bustos DE, Atherton SS. Detection of herpes simplex virus type 1 in human ciliary ganglia. Invest Ophthalmol Vis Sci 2002;43:2244-2249. Labetoulle M, Kucera P, Ugolini G, et al. Neuronal propagation of HSV] from the oral mucosa to the eye. Invest Ophthalmol Vis Sci 2000;41:2600-2606. Maggs DJ, Lappin MR, Nasisse MP. Detection of feline herpesvirus-specific antibodies and DNA in aqueous humor from cats with or without uveitis. Am J Vet Res 1999;60:932-936. 112 171. 172. 173. 174. 175. 176. 177. 178. 179. 180. 181. 182. 183. 184. Powell CC, Kordick DL, Lappin MR. Inoculation with Bartonella henselae followed by feline herpesvirus 1 fails to activate ocular toxoplasmosis in chronically infected cats. J Feline Med Surg 2002;4z107-110. Strubbe T, Gelatt K. Ophthalmic examination and diagnostic procedures. Veterinary Ophthalmology. 3rd edn. ed. Baltimore: Lippincott, Williams & Wilkins, 1999;440—441. Hanson BR, Schipper IA. Effects of swab materials on infectious bovine rhinotracheitis virus. Am J Vet Res 1976;37:707-708. Crandell RA. Virologic and immunologic aspects of feline viral rhinotracheitis virus. J Am Vet Med Assoc 1971;1582Suppl 2:922-926. Mochizuki M, Konishi S, Ogata M. Studies on cytopathogenic viruses from cats with respiratory infections. III. Isolation and certain properties of feline herpesviruses. Nippon Juigaku Zasshi 1977;39:27-37. Mochizuki M, Konishi SI, Ogata M. Sero-diagnostic aspects of feline herpesvirus infection. Nippon Juigaku Zasshi 1977;39:191-194. Satoh Y, Iizuka K, F ukuyama M, et al. An enzyme-linked immunosorbent assay using nuclear antigen for detection of feline herpesvirus 1 antibody. J Vet Diagn Invest 1999;11:334-340. Dawson DA, Carman J, Collins J, et a]. Enzyrne-linked immunosorbent assay for detection of feline herpesvirus 1 IgG in serum, aqueous humor, and cerebrospinal fluid. J Vet Diagn Invest 1998;10:315-319. Maggs DJ, Lappin MR, Collins J. Assessment of serum antibody measurement and virus detection methods in the diagnosis of F HV -1 infection in cats. American College of Veterinary Ophthalmologists, 26th annual conference 1995; 1. Nasisse MP. Manifestations, diagnosis, and treatment of ocular herpesvirus infection in the cat. Campend Contin Educ Vet Pract 1982;42962-970. da Silva Curie] JMA, Nasisse MP, Hook JR, et al. Topical fluorescein dye: effects on immunofluorescent antibody test for feline herpesvirus keratoconjunctivitis. Veterinary and Comparative Ophthalmology 1991;] :99- 104. Carlson JH, Scott FW. An immunofluorescence diagnostic test for feline viral rhinotracheitis. Am J Vet Res 1978;39:465-467. Burgesser KM, Hotaling S, Schiebel A, et al. Comparison of PCR, virus isolation, and indirect fluorescent antibody staining in the detection of naturally occurring feline herpesvirus infections. J Vet Diagn Invest 1999;11:122-126. Mullis KB, Faloona FA. Specific synthesis of DNA in vitro via a polymerase- catalyzed chain reaction. Methods Enzymol 1987;155:335-350. 113 185. 186. 187. 188. 189. 190. 191. 192. 193. 194. 195. 196. 197. Wasserrnan NT, M. TW, S. T, et al. The ability to detect FHV -1 using a real time PCR assay performed on samples collected with either microbrush swabs or Schirmer tear test strips compared to virus isolation from conjunctival swabs. ACVO 37th annual conference 2006;61. Maggs DJ, Clarke HE, Smith SJ. Effect of shipping conditions on PCR detection of feline herpesvirus (FHV-l) DNA in clinical ocular samples. 35th Annual Conference of the American College of Veterinary Ophthalmologists 2004;20. Sykes JE, Browning GF, Anderson G, et al. Differential sensitivity of culture and the polymerase chain reaction for detection of feline herpesvirus 1 in vaccinated and unvaccinated cats. Arch Virol 1997;142:65-74. Stiles J, McDermott M, Willis M, et al. Comparison of nested polymerase chain reaction, virus isolation, and fluorescent antibody testing for identifying feline herpesvirus in cats with conjunctivitis. Am J Vet Res 1997;58:804-807. Hara M, Fukuyama M, Suzuki Y, et al. Detection of feline herpesvirus 1 DNA by the nested polymerase chain reaction. Vet Microbiol 1996;48:345-352. Lutz H, Leutenegger C, Hofrnann-Lehmann R. The role of polymerase chain reaction and its newer developments in feline medicine. J Feline Med Surg 1999;1:89-100. Low HC, Powell CC, Veir JK, et al. Prevalence of feline herpesvirus 1, Chlamydophila felis, and Mycoplasma spp DNA in conjunctival cells collected from cats with and without conjunctivitis. Am J Vet Res 2007;68:643-648. Maggs DJ, Clarke HE. Relative sensitivity of polymerase chain reaction assays used for detection of feline herpesvirus type 1 DNA in clinical samples and commercial vaccines. Am J Vet Res 2005;66:1550-1555. Ohmura Y, Ono E, Matsuura T, et al. Detection of feline herpesvirus 1 transcripts in trigeminal ganglia of latently infected cats. Arch Virol 1993;129:341-347. Suchy A, Bauder B, Gelbmann W, et al. Diagnosis of feline herpesvirus infection by immunohistochemistry, polymerase chain reaction, and in situ hybridization. J Vet Diagn Invest 2000;12:186-191. Kim 0, Yi SJ. Optimization of in situ hybridization protocols for detection of feline herpesvirus 1. J Vet Med Sci 2003;65:1031-1032. Brown C. In situ hybridization with riboprobes: an overview for veterinary pathologists. Vet Pathol 1998;35:159-167. Gaskell R, Willoughby K. Herpesviruses of carnivores. Vet Microbiol 1999;69:73-88. 114 198. 199. 200. 201. 202. 203. 204. 205. 206. 207. 208. 209. 210. 211. 212. Townsend WM, Stiles J, Guptill-Yoran L, et al. Development of a reverse transcriptase-polyrnerase chain reaction assay to detect feline herpesvirus-1 latency-associated transcripts in the trigeminal ganglia and corneas of cats that did not have clinical signs of ocular disease. Am J Vet Res 2004;65:314-319. Gaskell RM, Dennis PE, Goddard LE, et al. Isolation of felid herpesvirus I from the trigeminal ganglia of latently infected cats. J Gen Viral 1985;66 (Pt 2):39l-394. Nasisse MP, Davis BJ, Guy JS, et al. Isolation of feline herpesvirus 1 from the trigeminal ganglia of acutely and chronically infected cats. J Vet Intern Med 1992;6:102-103. Laycock KA, Lee SF, Stulting RD, et al. Herpes simplex virus type 1 transcription is not detectable in quiescent human stromal keratitis by in situ hybridization. Invest Ophthalmol Vis Sci 1993;34:285-292. Kraft W. Virale Infektionskrankheiten In: Kraft W,Diirr UM, eds. Katzenkrankheiten, Klinik und Therapie. 4 ed. Hannover: Schaper, 1996;123- 1 59. Stiles J. Feline herpesvirus. Clin Tech Small Anim Pract 2003;18:178-185. Martin CL. Krankheiten des Auges In: Kraft W,Diirr UM, eds. Katzenkrankheiten. Honnover: Schaper GmbH, 1996;783-844. Plumb DC. Plumb's veterinary drug handbook. 5th ed. Ames, Iowa: Blackwell, 2005. Spiess AK, Sapienza J S, Myordomo A. Treatment of feline eosinophilic proliferative keratitis with topical cyclosporine: 30 cases. ACVO 38th Annual Conference 2007;47. Galle LE. Antiviral therapy for ocular viral disease. Vet Clin North Am Small Anim Pract 2004;34:639-653. Greene CE, Watson A. Antiviral drugs In: Greene CE, ed. Infectious Disease of the Dog and Cat. 2 ed. Philadelphia: W.B. Saunders Company, 1998;6-9. Elion GB, Furman PA, F yfe JA, et a]. Selectivity of action of an antiherpetic agent, 9-(2-hydroxyethoxymethyl) guanine. Proc Natl Acad Sci U S A 1977;74:5716-5720. Fyfe JA, Keller PM, F urrnan PA, et al. Thymidine kinase from herpes simplex virus phosphorylates the new antiviral compound, 9-(2- hydroxyethoxymethyl)guanine. J Biol Chem 1978;253:8721-8727. Gustafson DP. Antiviral therapy. Vet Clin North Am Small Anim Pract 1986;16:1181-1189. Derse D, Cheng YC, Furman PA, et al. Inhibition of purified human and herpes simplex virus-induced DNA polymerases by 9-(2- 115 213. 214. 215. 216. 217. 218. 219. 220. 221. 222. 223. 224. 225. 226. hydroxyethoxymethyl)guanine triphosphate. Effects on primer-template function. J Biol Chem 1981;256:11447-11451. F urman PA, St Clair MH, Spector T. Acyclovir triphosphate is a suicide inactivator of the herpes simplex virus DNA polymerase. J Biol Chem 1984;259:9575-9579. Jacobson MA. Valaciclovir (BW256U87): the L-valyl ester of acyclovir. J Med Virol 1993;Suppl 1:150-153. de Miranda P, Bumette TC. Metabolic fate and pharrnacokinetics of the acyclovir prodrug valaciclovir in cynomolgus monkeys. Drug Metab Dispos 1994;22:55-59. Bumette TC, de Miranda P. Metabolic disposition of the acyclovir prodrug valaciclovir in the rat. Drug Metab Dispos 1994;22:60-64. Bumette TC, Harrington J A, Reardon JE, et al. Purification and characterization of a rat liver enzyme that hydrolyzes valaciclovir, the L-valyl ester prodrug of acyclovir. J Biol Chem 1995;270:15827-15831. Weller S, Blum MR, Doucette M, et al. Pharmacokinetics of the acyclovir pro- drug valaciclovir after escalating single- and multiple-dose administration to normal volunteers. Clin Pharmacol Ther 1993;54:595-605. de Miranda P, Blum MR. Pharmacokinetics of acyclovir after intravenous and oral administration. J Antimicrob Chemother 1983;12 Suppl B129-37. Owens JG, Nasisse MP, Tadepalli SM, et al. Pharmacokinetics of acyclovir in the cat. J Vet Pharmacol Ther 1996;19:488-490. Nasisse MP, Dorman DC, Jamison KC, et al. Effects of valacyclovir in cats infected with feline herpesvirus 1. Am J Vet Res 1997;58:1141-1144. De Clercq E, Field HJ. Antiviral prodrugs - the development of successful prodrug strategies for antiviral chemotherapy. Br J Pharmacol 2006;147:1-1 1. Heit MC, Riviere JE. Antifungals and antiviral drugs In: Adams RH, ed. Veterinary pharmacology and therapeutics. 7 ed. Ames: Iowa State University Press, 1995. Pandit UK, Grose WFA, Eggelte TA. A new class of nucleoside analogues. Synthesis of Nl-pyrimidiny-and N9-purinyl-4'-hydroxy-3- (hydroxymethyl)butanes. Synth Commun 1972;2z345-351. Boyd MR, Bacon TH, Sutton D, et al. Antiherpesvirus activity of 9-(4- hydroxy-3-hydroxy-methylbut-1-y1)guanine (BRL 39123) in cell culture. Antimicrob Agents Chemother 1987;3 l : 123 8-1242. Vere Hodge RA, Perkins RM. Mode of action of 9-(4-hydroxy-3- hydroxyrnethyl-but-l-yl)guanine (BRL 39123) against herpes simplex virus in MRC-S cells. Antimicrob Agents Chemother 1989;33:223-229. 116 227. 228. 229. 230. 231. 232. 233. 234. 235. 236. 237. 238. 239. 240. Eamshaw DL, Bacon TH, Darlison SJ, et al. Mode of antiviral action of penciclovir in MRC-5 cells infected with herpes simplex virus (HSV-l), HSV- 2, and varicella-zoster virus. Antimicrob Agents Chemother 1992;36:2747- 2757. Hamden MR, Jarvest RL, Boyd MR, et al. Prodrugs of the selective antiherpesvirus agent 9-[4-hydroxy-3-(hydroxymethyl)but-l-yl] guanine (BRL 39123) with improved gastrointestinal absorption properties. J Med Chem 1989;32:1738-1743. Vere Hodge RA. F amciclovir and penciclovir. The mode of action of famciclovir including its conversion to penciclovir. Antiviral Chem Chemother 1993;4267-84. Field HJ. Famciclovir - origins, progress and prospects. Expert Opin Invest Drugs 1996;5z925-938. Thackray AM, Field HJ. Famciclovir and valaciclovir differ in the prevention of herpes simplex virus type 1 latency in mice: a quantitative study. Antimicrob Agents Chemother 1998;42:1555-1562. Thomasy SM, Maggs DJ, Moulin NK, et al. Pharmacokinetics and safety of penciclovir following oral administration of famciclovir to cats. Am J Vet Res 2007;68: 1252-1258. Prusoff WH. Synthesis and biological activities of iododeoxyuridine, an analog of thymidine. Biochim Biophys Acta 1959;32:295-296. Hom MM. Masby’s Ocular Drug Consult. St. Louis: Mosby Elsevier, 2006. Heidelberger C, Parsons DG, Remy DC. Syntheses Of 5-Trifluoromethyluraci1 And 5-Trifluoromethyl-2'-Deoxyuridine. J Med Chem 1964;7zl-5. Johnson AW, Jervey ED. Adenine Arabinoside And S-Mercaptouracil In Herpes Keratitis. Arch Ophthalmol 1964;72:826-828. Bean B. Antiviral therapy: current concepts and practices. Clin Microbiol Rev 1992;52146-182. de Clercq E, Sakuma T, Baba M, et a]. Antiviral activity of phosphonylmethoxyalkyl derivates of purine and pyrimidines. Antiviral Res 1987;8z261-272. Bischofberger N, Hitchcock MJ, Chen MS, et al. 1-[((S)-2-hydroxy-2-oxo- 1,4,2-dioxaphosphorinan-5-yl)methyl] cytosine, an intracellular prodrug for (S)-1 -(3-hydroxy-2-phosphonylmethoxypropyl)cytosine with improved therapeutic index in vivo. Antimicrob Agents Chemother 1994;38:2387-2391. Cundy KC, Barditch-Crovo P, Petty BG, et al. Clinical pharmacokinetics of 1- [((S)-2-hydroxy-2-oxo-1,4,2—dioxaphosphorinan-5-yl)methyl]cytosine in human immunodeficiency virus-infected patients. Antimicrob Agents Chemother 1999;43:271-277. 117 24]. 242. 243. 244. 245. 246. 247. 248. 249. 250. 25]. 252. 253. Mendel DB, Cihlar T, Moon K, et al. Conversion of l-[((S)-2-hydroxy-2-oxo- 1,4,2-dioxaphosphorinan-5-y1)methyl]cytosine to cidofovir by an intracellular cyclic CMP phosphodiesterase. Antimicrob Agents Chemother 1997;41:641- 646. Painter GR, Hostetler KY. Design and development of oral drugs for the prophylaxis and treatment of smallpox infection. Trends Biotechnal 2004;22:423-427. Gilliam SE, Field HJ. The effect of (S)-l-(3-hydroxy-2-phosphonyl- methoxypropyl)cytosine (HPMPC) on bovine herpesvirus-1 (BHV-l) infection and reactivation in cattle. Antiviral Res 1993;20:21-32. Kaufman HE, Vamell ED, Thompson HW. Trifluridine, cidofovir, and penciclovir in the treatment of experimental herpetic keratitis. Arch Ophthalmol 1998;] 16:777-780. Gordon YJ, Romanowski EG, Araullo-Cruz T. HPMPC, a broad—spectrum topical antiviral agent, inhibits herpes simplex virus type 1 replication and promotes healing of dendritic keratitis in the New Zealand rabbit ocular model. Cornea 1994;13:516-520. Romanowski EG, Bartels SP, Gordon YJ. Comparative antiviral efficacies of cidofovir, trifluridine, and acyclovir in the HSV-l rabbit keratitis model. Invest Ophthalmol Vis Sci 1999;40:378-384. Kaufman HE, Vamell ED, Thompson HW. Cidofovir and experimental herpetic stromal disease. Arch Ophthalmol 1999;117:925-928. Reyes GR. Ribavirin: recent insights into antiviral mechanisms of action. Curr Opin Drug Discav Devel 2001;4:651-656. Nasisse MP, Guy J S, Davidson MG, et al. In vitro susceptibility of feline herpesvirus-1 to Vidarabine, idoxuridine, trifluridine, acyclovir, or bromovinyldeoxyuridine. Am J Vet Res 1989;50:158-160. Maggs DJ, Clarke HE. In vitro efficacy of ganciclovir, cidofovir, penciclovir, foscamet, idoxuridine, and acyclovir against feline herpesvirus type-l. Am J Vet Res 2004;65:399-403. van der Meulen K, Garre B, Croubels S, et al. In vitro comparison of antiviral drugs against feline herpesvirus 1. BMC Vet Res 2006;2zl3. Sandmeyer LS, Keller CB, Bienzle D. Effects of cidofovir on cell death and replication of feline herpesvirus-1 in cultured feline corneal epithelial cells. Am J Vet Res 2005;66:217-222. F ontenelle JP, Powell CC, Veir JK, et al. Effect of topical ophthalmic application of cidofovir on experimentally induced primary ocular feline herpesvirus-l infection in cats. Am J Vet Res 2008;69:289-293. 118 254. 255. 256. 257. 258. 259. 260. 261. 262. 263. 264. 265. 266. 267. 268. Williams DL, Robinson JC, Lay E, et al. Efficacy of topical aciclovir for the treatment of feline herpetic keratitis: results of a prospective clinical trial and data from in vitro investigations. Vet Rec 2005;157:254-257. Crumpacker CS. Mechanism of action of foscamet against viral polymerases. Am J Med 1992;92:3S-7S. Tankersley RW, Jr. Amino Acid Requirements Of Herpes Simplex Virus In Human Cells. J Bacteriol 1964;87:609-613. Spring SB, Roizman B, Spear PG. Selective failure of protein synthesis in herpesvirus-infected cells deprived of arginine. Virology 1969;38:710-712. Griffith RS, DeLong DC, Nelson JD. Relation of arginine-lysine antagonism to herpes simplex grth in tissue culture. Chemotherapy 1981;27:209-213. Walsh DE, Griffith RS, Behforooz A. Subjective response to lysine in the therapy of herpes simplex. J Antimicrob Chemother 1983;12:489-496. Griffith RS, Walsh DE, Myrmel KH, et al. Success of L-lysine therapy in frequently recurrent herpes simplex infection. Treatment and prophylaxis. Dermatologica 1987;175:183-190. Griffith RS, Norins AL, Kagan C. A multicentered study of lysine therapy in Herpes simplex infection. Dermatologica 1978;156:257-267. Kadirvel R, Kratzer FH. Uptake of L-arginine and L-lysine by the small intestine and its influence on arginine-lysine antagonism in chicks. J Nutr 1974;104:339-343. Banos G, Daniel PM, Pratt OE. Saturation of a shared mechanism which transports L-arginine and L-lysine into the brain of the living rat. J Physiol 1974;236:29-41. Morris JG, Rogers QR. Ammonia intoxication in the near-adult cat as a result of a dietary deficiency of arginine. Science 1978;199:431-432. Maggs DJ, Collins BK, Thorne JG, et al. Effects of L-lysine and L-arginine on in vitro replication of feline herpesvirus type-1. Am J Vet Res 2000;61:1474- 1478. Stiles J, Townsend WM, Rogers QR, et al. Effect of oral administration of L- lysine on conjunctivitis caused by feline herpesvirus in cats. Am J Vet Res 2002;63:99-103. Maggs DJ, Nasisse MP, Kass PH. Efficacy of oral supplementation with L- lysine in cats latently infected with feline herpesvirus. Am J Vet Res 2003;64:37-42. Stewart WE, 2nd. Interferon nomenclature recommendations. J Infect Dis 1980;142:643. 119 269. 270. 271. 272. 273. 274. 275. 276. 277. 278. 279. 280. 281. 282. Nagano Y, Kojima Y, Sawai Y. [Immunity and interference in vaccinia; inhibition of skin infection by inactivated virus]. C R Seances Soc Biol F il 1954;148:750-752. Isaacs A, Lindenmann J. Virus interference. I. The interferon. Proc R Soc Lond B Biol Sci 1957;147:258-267. Neumann-Haefelin D, Sundmacher R, Sauter B, et al. Effect of human leukocyte interferon on vaccinia-and herpes virus-infected cell cultures and monkey corneas. Infect Immun 1975;12:148-155. McGill J], Collins P, Cantell K, et al. Optimal schedules for use of interferon in the corneas of rabbits with herpes simplex keratitis. J Infect Dis 1976;133 Supple13-17. Trousdale MD, Robin JB, Stebbing N. Reevaluation of human alpha interferons against herpesvirus infection of the rabbit eye. Curr Eye Res 1987;6z269-272. de Koning EW, van Bij sterveld OP, Cantell K. Combination therapy for dendritic keratitis with acyclovir and alpha-interferon. Arch Ophthalmol 1983;101:1866-1868. Sundmacher R, Cantell K, Mattes A. Combination therapy for dendritic keratitis. High-titer alpha-interferon and trifluridine. Arch Ophthalmol 1984;102:554-555. Taylor JL, Punda-Polic V, O'Brien WJ. Combined anti-herpes virus activity of nucleoside analogs and interferon. Curr Eye Res 1991;10 Suppl:205-211. Ho M. Interferon as an agent against herpes simplex virus. J Invest Dermatol 1990;95:158S-160S. Smolin G, Matli M, Okumoto M, et al. Use of recombinant interferon to prevent recurrent herpesvirus shedding. Curr Eye Res 1984;3: 1069-1074. Sundmacher R, Neumann-Haefelin D, Cantell K. Successful treatment of dendritic keratitis with human leukocyte interferon. A controlled clinical study. Albrecht Von Graefes Arch Klin Exp Ophthalmol 1976;201:39—45. Fulton RW, Burge LJ. Susceptibility of feline herpesvirus 1 and a feline calicivirus to feline interferon and recombinant human leukocyte interferons. Antimicrob Agents Chemother 1985;28:698-699. Sandmeyer LS, Keller CB, Bienzle D. 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 2005;66:210-216. Haid C, Kaps S, Gonczi E, et al. Pretreatment with feline interferon omega and the course of subsequent infection with feline herpesvirus in cats. Vet Ophthalmol 2007; 10:278-284. 120 283. 284. 285. 286. 287. 288. 289. 290. 291. 292. 293. 294. 295. 296. 297. Gutzwiller ME, Brachelente C, Taglinger K, et al. Feline herpes dermatitis treated with interferon omega. Vet Dermatol 2007;18:50-54. Arnold RR, Cole MF, McGhee JR. A bactericidal effect for human lactoferrin. Science 1977;197:263-265. Bellamy W, Takase M, Yamauchi K, et al. Identification of the bactericidal domain of lactoferrin. Biochim Biophys Acta 1992;l 121:130-136. Bellamy W, Takase M, Wakabayashi H, et al. Antibacterial spectrum of lactoferricin B, a potent bactericidal peptide derived from the N-terminal region of bovine lactoferrin. J Appl Bacterial 1992;73:472-479. Hoek KS, Milne JM, Grieve PA, et al. Antibacterial activity in bovine lactoferrin-derived peptides. Antimicrob Agents Chemother 1997;41:54-59. Bellamy W, Wakabayashi H, Takase M, et al. Killing of Candida albicans by lactoferricin B, a potent antimicrobial peptide derived from the N-terminal region of bovine lactoferrin. Med Microbial Immunol 1993;182:97-105. Kirkpatrick CH, Green 1, Rich R, et al. Inhibition of growth of Candida albicans by iron-unsaturated lactoferrin: relation to host-defense mechanisms in chronic mucocutaneous candidiasis. J Infect Dis 1971;124:539-544. Soukka T, Tenovuo J, Lenander-Lumikari M. Fungicidal effect of human lactofenin against Candida albicans. FEMS Microbiol Lett 1992;69:223-228. Turchany JM, Aley SB, Gillin FD. Giardicidal activity of lactoferrin and N- terminal peptides. Infect Immun 1995;63:4550-4552. van der Strate BW, Beljaars L, Molema G, et al. Antiviral activities of lactoferrin. Antiviral Res 2001;52:225-239. Marchetti M, Pisani S, Antonini G, et a]. Metal complexes of bovine lactoferrin inhibit in vitro replication of herpes simplex virus type 1 and 2. Biometals 1998;] 1:89-94. Sato R, Inanami 0, Tanaka Y, et al. Oral administration of bovine lactoferrin for treatment of intractable stomatitis in feline immunodeficiency virus (F IV)- positive and FIV-negative cats. Am J Vet Res 1996;57:1443-1446. Fujihara T, Hayashi K. Lactoferrin inhibits herpes simplex virus type-l (HSV- 1) infection to mouse cornea. Arch Viral 1995;140:1469-1472. Hasegawa K, Motsuchi W, Tanaka S, et al. Inhibition with lactoferrin of in vitro infection with human herpes virus. Jpn J Med Sci Biol 1994;47:73-85. Hirano N, Sato R, K. Y, et al. Antiviral effect of bovine lactoferrin on feline herpesvirus infection. fourth International Conference on Lactoferrin: Structure, Function, and Applications 1999;233-237. 121 298. 299. 300. 301. 302. 303. 304. 305. 306. 307. 308. 309. 310. Beaumont SL, Maggs DJ, Clarke HE. Effects of bovine lactoferrin on in vitro replication of feline herpesvirus. Vet Ophthalmol 2003;6:245-250. Lappin MR, Sebring RW, Porter M, et al. Effects of a single dose of an intranasal feline herpesvirus 1, calicivirus, and panleukopenia vaccine on clinical signs and virus shedding after challenge with virulent feline herpesvirus 1. J Feline Med Surg 2006;8:158-163. Sussman MD, Maes RK, Kruger JM. Vaccination of cats for feline rhinotracheitis results in a quantitative reduction of virulent feline herpesvirus- ] latency load after challenge. Virology 1997;228:379-382. Weigler BJ, Guy J S, Nasisse MP, et al. Effect of a live attenuated intranasal vaccine on latency and shedding of feline herpesvirus 1 in domestic cats. Arch Virol 1997;142:2389-2400. Cocker FM, Newby TJ, Gaskell RM, et al. Responses of cats to nasal vaccination with a live, modified feline herpesvirus type 1. Res Vet Sci 1986;41:323-330. Gaskell RM, Gettinby G, Graham SJ, et a1. Veterinary Products Committee working group report on feline and canine vaccination. Vet Rec 2002;150:126- 134. Kruger JM, Sussman MD, Maes RK. 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 1996;220:299-308. Povey RC. Feline respiratory disease - which vaccine? Feline Practice 1977;7:12-16. Povey RC, Wilson MR. A comparison of inactivated feline viral rhinotracheitis and feline caliciviral disease vacccines with live-modified viral vaccines. Feline Practice 1978;8z35-42. Yokoyama N, Maeda K, Tohya Y, et al. Pathogenicity and vaccine efficacy of a thymidine kinase-deficient mutant of feline herpesvirus type 1 in cats. Arch Virol 1996;141:481-494. Yokoyama N, F uj ita K, Damiani A, et a]. Further development of a recombinant feline herpesvirus type 1 vector expressing feline calicivirus immunogenic antigen. J Vet Med Sci 1998;60:717-723. Maeda K, Ono M, Kawaguchi Y, et al. Expression and properties of feline herpesvirus type 1 gD (hemagglutinin) by a recombinant baculovirus. Virus Res 1996;46:75-80. Spatz SJ, Rota PA, Maes RK. Identification of the feline herpesvirus type 1 (FHV-l) genes encoding glycoproteins G, D, I and E: expression of FHV-l glycoprotein D in vaccinia and raccoon poxviruses. J Gen Virol 1994;75 (Pt 6):]235-1244. 122 3]]. 312. 313. 314. 315. 316. 317. 318. 319. 320. 321. 322. 323. 324. Sussman MD, Maes RK, Kruger JM, et al. 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 1995;214:12-20. Willemse MJ, Chalmers WS, Sondermeijer P]. In vivo properties of a feline herpesvirus type 1 mutant carrying a lacZ insertion at the g1 locus of the unique short segment. Vaccine 1996;14:1-5. Willemse MJ, Chalmers WS, Cronenberg AM, et al. The gene downstream of the gC homologue in feline herpes virus type 1 is involved in the expression of virulence. J Gen Virol 1994;75 (Pt 11):3107-3116. Yokoyama N, Maeda K, Mikami T. Recombinant viral vector vaccines for the veterinary use. J Vet Med Sci 1997;59:311-322. Sato E, Yokoyama N, Miyazawa T, et a1. Efficient expression of the envelope protein of feline immunodeficiency virus in a recombinant feline herpesvirus type 1 (FHV-l) using the gC promoter of FHV-l. Virus Res 2000;70:13-23. Sato E, Yokoyama N, Maeda K, et al. Construction of a recombinant feline herpesvirus type 1 expressing Gag precursor protein of feline immunodeficiency virus. Arch Virol 1998;143:453-466. Sato E, Miyazawa T, Nishimura Y, et al. Further development of a recombinant feline herpesvirus type 1 expressing the Gag protein of feline immunodeficiency virus. Arch Virol 2001;146:379-387. Willemse MJ, van Schooneveld SH, Chalmers WS, et a1. Vaccination against feline leukaemia using a new feline herpesvirus type 1 vector. Vaccine 1996;14:1511-1516. Yokoyama N, Maeda K, Tohya Y, et a]. Recombinant feline herpesvirus type 1 expressing immunogenic proteins inducible virus neutralizing antibody against feline calicivirus in cats. Vaccine 1996;14:1657-1663. Yokoyama N, Maeda K, F ujita K, et al. Vaccine efficacy of recombinant feline herpesvirus type 1 expressing immunogenic proteins of feline calicivirus in cats. Arch Virol 1996;141:2339-2351. Mishima M, Xuan X, Yokoyama N, et al. Recombinant feline herpesvirus type 1 expressing Toxoplasma gondi ROP2 antigen inducible protective immunity in cats. Parasitol Res 2002;88:144-149. Kuruppu D, Dorfrnan JD, Tanabe KK. HSV-l viral oncolysis and molecular imaging with PET. Curr Cancer Drug Targets 2007;7z175-180. Kuruppu D, Brownell AL, Zhu A, et al. Positron emission tomography of herpes simplex virus 1 oncolysis. Cancer Res 2007;67:3295-3300. Prince JH, Diesem CD, Eglitis I, et al. Anatomy and histology of the eye and orbit in domestic animals. Springfield: Charles C Thomas, 1960. 123 325. 326. 327. 328. 329. 330. 331. 332. 333. . 334. 335. 336. 337. 338. 339. Samuelson DA. Ophthalmic Anatomy In: Gelatt K, ed. Veterinary E Ophthalmology. 3rd ed. Philadelphia: Lippincott Williams & Wilkins, 1999;31-150. Fine BS, Yanoff M. Ocular Histology A Text and Atlas. 2nd ed. Hagerstown: Harper & Row, 1979. Murphy CJ, Pollock RVS. The Eye In: Saunders, ed. Miller's Anatomy of the dog. 3rd ed. Philadelphia, 1993;1009-1057. Bdhme G. Nervensystem, Sinnesorgane, endokrine Driisen. 3. Auflage ed. Berlin und Hamburg: Verlag Paul Parey, 1992. Evans HE, Kitchell RL. Cranial Nerves and Cutaneous Innervation of the Head In: Saunders, ed. Miller's Anatomy of the dog. 3rd ed. Philadelphia: Saunders, 1993;953-987. Scagliotti RH. Comparative neuro-ophthalmology. Veterinary Ophthalmology. 3rd ed. Philadelphia: Lippincott Williams & Wilkins, 1999;1307—1400. Glaze MB, Gelatt KN. Feline Ophthalmology In: Gelatt K, ed. Veterinary Ophthalmology. 3 ed: Lippincott Williams &Williams, 1998;997-1052. Tai S WA, Hannala J, et al. Detection of acute and latent feline herpesvirus-1 (F HV-l) infections by SYBR Green-based real-time PCR. Phi Zeta Research Day 2004. Kim O, Chae C. In situ hybridization for the detection and localization of porcine epidemic diarrhea virus in the intestinal tissues from naturally infected piglets. Vet Pathol 2000;37:62-67. Sawtell NM. Comprehensive quantification of herpes simplex virus latency at the single-cell level. J Viral 1997;71:5423-5431. Sawtell NM, Poon DK, Tansky CS, et al. The latent herpes simplex virus type 1 genome copy number in individual neurons is virus strain specific and correlates with reactivation. J Viral 1998;72:5343-5350. Wang K, Lau TY, Morales M, et al. Laser-capture microdissection: refining estimates of the quantity and distribution of latent herpes simplex virus 1 and varicella-zoster virus DNA in human trigeminal Ganglia at the single-cell level. J Viral 2005;79:14079-14087. Wheeler JG, Osorio FA. Investigation of sites of pseudorabies virus latency, using polymerase chain reaction. Am J Vet Res 1991;52: 1799-1803. Brown TT, Jr., Shin KO, Fuller FJ. Detection of pseudorabies viral DNA in tonsillar epithelial cells of latently infected pigs. Am J Vet Res 1995 ;56:587- 594. Jerome KR, Fox R, Chen Z, et a]. Herpes simplex virus inhibits apoptosis through the action of two genes, Us5 and Us3. J Viral 1999;73:8950-8957. 124 340. Miles D, Athmanathan S, Thakur A, et al. A novel apoptotic interaction between HSV-1 and human corneal epithelial cells. Curr Eye Res 2003;26:165-174. 341. Gown AM, Willingham MC. Improved detection of apoptotic cells in archival paraffin sections: immunohistochemistry using antibodies to cleaved caspase 3. J Histochem Cytochem 2002;50:449-454. 125 ’. .‘r’riiir-r' swu~*:n:-‘urrr‘lrer.1 _ . .. -\1‘!!r'.-\'-1.'. .~ Illllllllllll ‘. l 9 111