Anda di halaman 1dari 20

Internasional Ilmiah Jaringan Penelitian ISRN Kedokteran Hewan Volume 2012, ID Artikel 495.

830, 14 halaman doi: 10,5402 /


2012 / 495.830

Review Pasal felid Herpes tipe 1 Infeksi di Kucing: A Model


host Alam untuk Alphaherpesvirus Patogenesis
Roger Maes
Departemen Pathobiology dan Investigasi Diagnostik dan Mikrobiologi dan Genetika Molekuler, College of Veterinary
Medicine, Michigan State University, East Lansing, MI 48824, USA
Correspondence harus ditujukan kepada Roger Maes, maes@dcpah.msu.edu~~V~~aux
Diterima 11 September 2012; Diterima 20 Oktober 2012
Editor Akademik: MH Kogut, V. Nair, dan S. Takai
Copyright 2012 Roger Maes. Ini adalah sebuah artikel akses terbuka didistribusikan di bawah lisensi Creative Commons
Atribusi, yang memungkinkan penggunaan tak terbatas, distribusi, dan reproduksi dalam media apapun, asalkan karya asli benar
dikutip.
Feline herpesvirus 1 (FeHV-1) adalah alphaherpesvirus yang menyebabkan kucing viral rhinotracheitis, penyakit virus penting
dari kucing di seluruh dunia. Akut FeHV-1 infeksi dikaitkan dengan kedua pernapasan atas dan tanda-tanda mata. Setelah fase
akut penyakit latency seumur hidup didirikan, terutama di sel neuron sensorik. Seperti halnya virus herpes simpleks manusia
reaktivasi latency dapat mengakibatkan luapan baru, yang dapat memanifestasikan dirinya dalam bentuk lesi mata yang serius.
FeHV-1 infeksi pada kucing merupakan model tuan rumah alami yang berguna untuk identifikasi gen virulensi virus yang
berperan dalam replikasi di portal mukosa masuk atau mediator dari pembentukan, pemeliharaan, atau reaktivasi latency. Ini juga
merupakan sistem model untuk menentukan mekanisme imunitas bawaan dan adaptif dan strategi imunisasi yang dapat
menyebabkan perlindungan yang lebih baik terhadap ini dan infeksi alphaherpesvirus lainnya.

1. Pendahuluan
felid herpesvirus 1 (FeHV-1) diklasifikasikan dalam Ordo: Herpesvirales, Keluarga: Herpesviridae, Subfamili:
Alphaher- pesvirinae, dan genus: Varicellovirus [1]. Karakteristik anggota Alphaherpesvirinae pendek siklus mereka
replicated kation, induksi latency seumur hidup, utama dalam Rons neutrofil, dan, dalam banyak kasus, kisaran
inang yang sempit. Kedua virus herpes manusia dan hewan adalah anggota subfamili Alphaher- pesvirinae. Jenis
manusia herpes virus simpleks 1 (HSV 1) dan 2 (HSV-2), masing-masing, menyebabkan luka dingin dan lesi genital.
Varicella zoster virus (VZV) merupakan agen penyebab cacar, serta reaktivasi laten VZV DNA menyebabkan herpes
zoster. Beberapa herpesviruses mamalia, selain FeHV-1, yang diklasifikasikan dalam keluarga ini memiliki sapi
herpesvirus-1 (BoHV-1), yang menyebabkan penyakit pernafasan dan aborsi pada sapi, kuda herpesvirus-1 (EHV-1),
yang menyebabkan penyakit pernapasan, aborsi, dan dalam beberapa kasus neutrofil penyakit rological pada kuda,
Suid herpesvirus 1, juga dikenal sebagai pseudorabies (PRV) dan virus penyakit Aujeszky, yang mengarah ke
penyakit pernapasan, aborsi, penyakit saraf pada babi, dan canid herpesvirus-1 (CaHV-1 ), yang bertanggung jawab
untuk
kematian neonatal pada anak anjing dan juga penyakit pernapasan dan mata di remaja dan anjing dewasa. Contoh
virus herpes alfa burung yang menular Laringotrakheitis virus (ILTV), menyebabkan penyakit pernafasan parah
pada unggas, dan Marek Penyakit virus (MDV), yang menginduksi imunosupresi dan T-sel limfoma.
FeHV-1 infeksi menyebabkan rhinotracheitis virus kucing (FVR), yang tidak hanya menyumbang sekitar
setengah dari semua infeksi virus pernapasan bagian atas didiagnosis kucing, tetapi juga merupakan penyebab
penting dari lesi mata pada kucing. Seperti halnya untuk infeksi alphaherpesvirus lainnya, fase akut FVR diikuti
oleh latency seumur hidup. Selama tahap laten, virus FeHV-1 DNA tetap dalam bentuk episom, terutama dalam inti
neuron ganglion sensorik. Transkripsi RNA virus sangat terbatas, dan virus menular tidak diproduksi. Pembalikan
negara laten, yang disebabkan oleh stres alami atau administrasi kortikosteroid, dapat menginduksi reaktivasi virus
dalam sel yang terinfeksi secara laten, yang mengarah ke baru duction pro virus menular. Diaktifkan kembali virus

menular kemudian perjalanan ke pinggiran oleh transportasi aksonal anterograde, berpotensi menyebabkan tandatanda klinis (luapan baru), dan dapat menyebabkan penularan virus [2-5].
Sejak FeHV-1 merupakan patogen utama kucing, dengan piratory res- dan komponen mata penyakit yang mirip
dengan virus herpes manusia, dan latency yang mudah diaktifkan kembali dalam kondisi alamiah, FeHV-1 infeksi
pada kucing dianggap model tuan rumah alami yang baik untuk mempelajari patogenesis molekuler komparatif
infeksi alphaherpesvirus akut dan laten dan untuk menguji strategi imunisasi baru.

2. Karakteristik virus
Ukuran FeHV-1 virion berkisar 120-180 nm. Mereka terdiri dari inti yang mengandung genom untai ganda virus
DNA, sebuah kapsid ikosahedral mengelilingi inti, lapisan tegument sekitarnya kapsid, dan sebuah amplop lipid
bilayer yang paku glikoprotein yang menonjol [6, 7].
FeHV-1 terutama menginfeksi kucing domestik, tapi singa dan cheetah juga rentan [3, 8] .Dalam vitro, FeHV-1
ulangan hanya di sel asal kucing. Alphaherpesviruses yang secara genetik terkait dengan FeHV-1 adalah canid
herpesvirus 1 (CaHV- 1) dan virus herpes phocid (PhHV) 1 dan 2 [3, 9-11].

3.Genomic Organisasi
Laboratoriumkami [12] melaporkan peta Sal Saya pertama dari genom C-27 strain FeHV-1 dan menetapkan bahwa
ukurannya adalah sekitar 134 kb. Grail et al. [13] kemudian memetakan genom dari FeHV-1 B927 saring dan
disepakati di bahwa genom hanya 126 kb dalam ukuran. Organisasi genom dari kedua FeHV-1 strain ini ditemukan
mirip dengan varicelloviruses lainnya. Pada dasarnya, FeHV- 1 genom terdiri dari dua segmen DNA yang unik,
disebut sebagai panjang Unik (UL) dan Unik pendek (US) daerah. Wilayah AS genom diapit oleh sepasang identik,
tetapi urutan terbalik ditunjuk Internal Ulangi Pendek (IRS) dan Terminal Ulangi Pendek (TRS).
Kami baru melaporkan urutan genom lengkap pertama dari FeHV-1, serta konstruksi dan terization-sifat dari
klon BAC berisi seluruh genom virus. Urutan genom lengkap berasal dari kedua FeHV-1 BAC dan DNA virion
dimurnikan. Data ini menunjukkan bahwa FeHV-1 genom adalah 135.797 bp dalam ukuran dan memiliki kandungan
GC dari 45%. Sebanyak 78 frame baca terbuka yang diperkirakan, pengkodean 74 protein yang berbeda. Gen
pemerintah, rencana kerja ditemukan colinear dengan yang paling varicelloviruses lain yang genom telah diurutkan
[14].
Semua alphaherpesviruses dianggap memiliki pola replikasi yang mirip dengan salah satu dari HSV-1 [6, 7 ].
FeHV- 1 sebelumnya telah terbukti mengandung 23 virion terkait protein [15]. Delapan glikoprotein awalnya telah
tified iDEN-, ditunjuk sebagai gB, gC, gD, GE, GG, gH, GI, dan gL. Pemeriksaan urutan lengkap baru-baru berasal
menunjukkan bahwa FeHV-1 genom sebenarnya berisi total 13 glikoprotein amplop [14].
Kebanyakan penelitian tentang fungsi FeHV-1 gen telah difokuskan pada peran glikoprotein amplop [16] ,
karena peran diperkirakan mereka dalam merangsangimun host pelindung
respondan, karena itu, potensi mereka untuk vaksin pembangunan pemerintah.

4. Infeksi akut
FeHV-1 biasanya mempengaruhi anak kucing dan kucing remaja. Kebanyakan anak kucing dilindungi oleh
kekebalan pasif sampai mereka sekitar 2 bulan.
Patogenesis FHV-1 didasarkan pada dua mekanisme yang berbeda. Yang pertama adalah bahwa FeHV-1 adalah
virus sitolitik. Contoh efek cytolytic nya adalah ulserasi di mukosa dan kornea. Mekanisme kedua yang kebalobatan iated, secara klinis mewujudkan dirinya sebagai stroma keratitis. Pertanyaan penting yang berkaitan dengan
mekanisme patogenetik kedua ini adalah sumber stimulasi antigenik mengemudi reaksi ini [17].
Sumber utama FeHV-1 transmisi oronasal dan okular sekresi dari kucing infeksi akut. Penularan virus juga dapat
dikaitkan dengan reaktivasi latency. Anak kucing dengan kekebalan pasif residual mungkin tidak menunjukkan
tanda-tanda klinis bila terkena tetapi menjadi laten terinfeksi [18].

Setelah masuk melalui rute oronasal, FeHV-1 ulangan secara luas di mukosa dari saluran pernapasan atas dan
biasanya menyebabkan penyakit pernapasan bagian atas parah pada hewan rentan . Masa inkubasi bervariasi dari 2
sampai 6 hari. Situs replikasi utama FeHV-1 meliputi mukosa septum hidung, konka, nasofaring, junctivae con-, dan
trakea atas. Replikasi juga terjadi di amandel dan kelenjar getah bening rahang bawah.
Pernapasan FeHV-1 infeksi akut ditandai inisiasi tially dengan demam, inappetence, dan bersin, diikuti dengan
keluarnya cairan hidung serosa, yang dapat menjadi mukopurulen setelah 5- 7 hari. Selain itu, replikasi lisan virus
dapat mengakibatkan air liur berlebihan dan air liur air liur. Kadang batuk dan dyspnea mungkin terjadi. Sariawan,
ciri khas infeksi kucing calicivirus, dapat terjadi sebagai akibat dari FeHV-1 infeksi rongga mulut tapi jarang [3].
Manifestasi okular terkait dengan FeHV-1 tion infeksi telah ditinjau oleh Gould [5 ]. Di anak kucing neonatal
ophthalmia neonatorum telah dijelaskan dan dapat menyebabkan kerusakan kornea serius. Konjungtivitis hyperemic
akut, menyebabkan debit mata dan chemosis, fitur infeksi akut, terjadi dalam hubungan dengan tanda-tanda
pernapasan bagian atas. Pembentukan ulkus epitel bercabang, disebut ulserasi sebagai dendritik, adalah fitur
patognomonik dari mata FeHV-1 infeksi akut. Dalam sebuah tinjauan terbaru dari ulkus kornea etiologi pada kucing,
Hartley [19] menyatakan "menganggap FHV-1 kecuali jika terbukti sebaliknya." Kadang-kadang, borok yang lebih
besar, disebut sebagai ulkus kornea geografis, mengembangkan. Kedua dendritik dan geografis ulserasi kornea juga
dapat terjadi akibat reaktivasi latency. Komponen lain dari lesi yang terkait dengan luapan baru adalah konjungtiva
dan / atau peradangan kornea, yang lebih ringan daripada terlihat selama penyakit akut.
FeHV-1 adalah terutama pernafasan atas dan patogen okular, dengan keterlibatan hanya sporadis paru-paru.
Tingkat viremia rendah, diduga terkait dengan sensitivitas suhu alami virus ini, yang akan mendukung replikasi pada
saluran pernapasan bagian atas. Paparan dari ratu hamil dapat menyebabkan aborsi, tetapi infeksi FeHV-1 infeksi
bukanlah penyebab umum aborsi di kucing. Pada anak kucing neonatal, infeksi dapat menggeneralisasi dan ciated
asso- dengan tanda-tanda neurologis dan tingkat kematian yang tinggi.

5. Alphaherpesvirus Latency Konsep


Sebuah tanda biologi alphaherpesvirus adalah bahwa tion infeksi akut diikuti oleh ketekunan seumur hidup dari
genom virus dalam bentuk laten pada jaringan saraf dan limfoid. . Latency dan reaktivasi periodik latency
merupakan bagian integral dari siklus hidup alphaherpesviruses dan elemen penting dalam kelangsungan hidup dan
transmisi mereka
Siklus latency-reaktivasi operasional terdiri dari tiga langkah utama: pembentukan, pemeliharaan, dan tion
reactiva-definisi.
Pembentukan latency dengan mensyaratkan bahwa virus mencapai jaringan di mana latency akan yang mapan.
Proses ini dimulai selama fase akut replikasi virus di situs mukosa periferal. Ujung saraf saraf sensorik innervating
situs replikasi virus mengambil partikel virus dan subpartikel selama fase ini. Partikel-partikel ini diangkut dalam
axoplasm dari akson saraf ini dengan proses disebut transportasi aksonal sebagai retrograde. Ketika virus mencapai
ganglia sensorik, menginfeksi neuron dan sel lainnya. Infeksi ini akut jenis sel ganglion berlangsung selama sekitar
satu minggu. Neuron adalah jenis sel di mana latency didirikan. Dalam rangka untuk mencapai hal ini, ekspresi gen
litik direpresi, sedangkan terkait transkrip latency- (LAT) dinyatakan, yang menghasilkan beberapa spesies RNA
oleh splicing. Ini beberapa spesies secara kolektif disebut sebagai LATs. Tingkat rendah atau transkripsi sporadis gen
segera-awal dan dini dapat terjadi tetapi tidak cukup untuk memulai infeksi produktif. Tidak ada virion menular
dapat dideteksi dalam ganglia selama infeksi laten. LAT RNA disambung, dan intron stabil dalam bentuk tali
penjerat ternak, yang disebut 2-kb LAT, diproduksi dalam nukleus. Disambung LAT mRNA yang diangkut ke
sitoplasma, di mana beberapa ORFs kecil dapat diterjemahkan menjadi protein.
Selama fase pemeliharaan latency, DNA virus hadir dalam neuron dalam bentuk episom. DNA virus tidak benarbenar statis selama fase pemeliharaan latency, tetapi aktivitas transkripsi dari genom terbatas untuk wilayah yang
disebut sebagai transkrip latency-terkait atau LAT.
Fase pemeliharaan latency adalah reversibel. Dengan kata lain, di bawah pengaruh rangsangan logis tertentu

alam atau pharmaco-, reaktivasi DNA virus laten dapat terjadi. Replikasi virus dimulai lagi, dan virion menular
kemudian perjalanan kembali ke pinggiran, menggunakan sama sensorik saraf "jalan raya" yang digunakan untuk
mencapai ganglia. Virus menular dapat dideteksi lagi oleh isolasi virus atau PCR dari hidung, mulut, atau swab
mata. Biasanya tanda-tanda klinis yang terkait dengan proses pengaktifan secara signifikan lebih ringan daripada
yang terlihat selama infeksi primer, dan reaktivasi pasti bisa tanpa gejala. Virus penumpahan dihasilkan dari
reaktivasi juga biasanya pada tingkat yang lebih rendah dan durasi lebih pendek dari
yang terlihat selama infeksi primer. Namun, mengaktifkan virus masih dapat menjadi sumber signifikan dari paparan
dan penyakit utama dalam sepenuhnya rentan host yang berada di kontak dekat dengan hewan yang reaktivasi
berlangsung. Reaktivasi terjadi di hanya sebagian kecil dari neuron yang terinfeksi secara laten, biasanya kurang
dari 0,05%. Terinfeksi secara laten neuron yang reaktivasi berlangsung tidak bertahan hidup. Hal ini menjelaskan
mengapa defisit sory sen- tidak dihubungkan dengan pengaktifan di ganglia saraf sensorik. Sejak reservoir neuron
yang terinfeksi secara laten tetap besar di bawah kondisi ini, reaktivasi berulang dapat berlangsung sepanjang hidup
dari tuan rumah.
Pemahaman kami saat ini regulasi latency berasal terutama dari studi tentang HSV-1 dan BoHV-1 [20- 22 ].
Berikut ringkasan terutama berasal dari sangat baik review yang sangat baru-baru HSV-1 latency oleh dan Perng
Jones [20].
5.1. Peran Latency-Associated Transkrip (LATs). Infeksi akut neuron ganglia trigeminal menghasilkan produk
ekspresi gen beracun yang membuat mereka rentan terhadap kerusakan dan kematian. Selain itu, kerusakan DNA sel
yang disebabkan oleh replikasi virus merangsang jalur mitokondria apoptosis. Virus herpes mencoba untuk melawan
apoptosis dan dengan demikian meningkatkan kemampuan replikatif mereka, dengan pengkodean beberapa gen
antiapoptotic, salah satunya adalah gen LAT. Karena ada redundansi dalam kemampuan antiapoptotic virus selama
fase akut, apoptosis neuron selama tahap ini dicegah cukup efisien.
Hal ini sangat penting bahwa apoptosis dicegah juga selama berlangsung pembentukan dan pemeliharaan tahap
latency. Hal ini terutama penting dalam neuron permisif, dimana replikasi virus ekstensif telah terjadi selama fase
akut. LAT diberikannya sifat antiapoptotic melalui mikro-RNA (miRNAs). Sebuah mekanisme yang LAT-dikodekan
Mirna mengatur apoptosis menargetkan mengubah faktor pertumbuhan beta, inducer kuat apoptosis [23, 24].
Hal ini penting untuk memahami interaksi antara genom virus laten dan neuron yang menyebabkan reactiva- tion
, karena ini merupakan prasyarat untuk akhirnya mengendalikan proses ini. LAT memainkan peran penting dalam
reaktivasi in vivo dari latency. Dalam penelitian eksperimental telah menunjukkan bahwa reaktivasi spontan sangat
terganggu jika gen LAT dihapus.
5.2. Peran tegument Protein VP16. Thompson et al. [25] baru-baru ini menggambarkan peran sentral yang
dimainkan oleh VP16 protein tegument dalam semua tahap HSV latency. Sebelum pembentukan replikasi virus
latency terjadi di neuron permisif. Dalam sel rentan di permukaan mukosa VP16, komponen virion memasuki sel,
dikombinasikan dengan faktor selular, mengaktifkan gen awal langsung. Transport aksonal dari VP16 menjadi
neuron tidak efisien, yang akan mempromosikan latency. Agar VP16 untuk memulai infeksi litik, perlu disintesis de
novo, sebuah proses yang mengharuskan penghambatan neuronal diatasi.
Sangat menarik, lokus LAT dianggap mengekspresikan riboregulators bahwa memediasi sintesis VP16. Telah
menunjukkan bahwa, dengan tidak adanya transkripsi LAT, setengah dari neuron ditakdirkan untuk terinfeksi secara
laten bukan memasuki siklus litik dan mati. Sebaliknya ketika represi diatasi, neuron menjadi lytically terinfeksi,
dan virus menular yang dihasilkan menyebar baik di dalam ganglia dan kembali ke permukaan mukosa di mana
infeksi dimulai. Tujuan dari infeksi litik adalah untuk meningkatkan jumlah sel-sel yang terinfeksi secara laten.
Stres, menyebabkan reaktivasi, dihipotesiskan untuk meningkatkan produksi novo dari VP16 dengan mekanisme
yang masih dalam penyelidikan. The VP16 dihasilkan kemudian memulai umpan balik dengan gen IE dan hasil di
tion reactiva- virus dalam jumlah yang sangat terbatas neuron yang terinfeksi secara laten.
5.3. Peran Sel-Mediated Responses Immune lokal. Sel T, terutama limfosit CD8 + T, telah ditemukan untuk menjadi

penting bagi akut mengendalikan infeksi HSV di sensorik gan- glia. Produksi antigen virus pada ganglia meningkat
trigeminal sampai 3 hari setelah infeksi, tetapi tidak lagi terdeteksi pada 7 hari setelah infeksi. Seperti penurunan
produksi antigen, ada peningkatan dari berbagai jenis dari berbagai jenis sel limfoid, seperti makrofag, sel
pembunuh alami (NK), dan sel CD8 + T tertentu sekitarnya neuron yang terinfeksi.
Diperkirakan bahwa sel-sel T, terutama limfosit CD8 + T , menghambat pengaktifan dari latency. Kegigihan sel
efektor imun dalam ganglia trigeminal (TG) menyiratkan bahwa rendahnya tingkat protein virus disajikan dan
bahwa respon imun terjadi. Dalam mouse HSV-1 model, telah menunjukkan bahwa replikasi DNA virus, transkripsi,
dan produksi protein virus berlangsung di 1 neuron per 10 TG. Neuron individu dianggap akan mengalami
"reaktivasi molekul spontan" dan secara konsisten dikelilingi oleh manset dari infiltrasi sel darah putih. Dua
mekanisme yang sel-sel infiltrasi mencegah elevasi reacti- adalah produksi interferon gamma dan limfosit monositdimediasi sitotoksisitas.

6. Reaktivasi Latency
The trigeminal ganglion dianggap sebagai situs utama dari latensi untuk FeHV-1 meskipun studi terbaru tersirat
jaringan lain sebagai lokasi potensial [26, 27].
Reaktivasi spontan adalah mungkin tetapi tidak sering terjadi. Lebih umum yang mengarah ke reaktivasi laten
FeHV-1 adalah hasil dari tekanan lingkungan atau fisiologis, seperti perubahan di perumahan atau menyusui.
Tingkat frekuensi reaktivasi telah dilaporkan 18% sebagai akibat dari bergerak kucing dengan lingkungan baru dan
40% sebagai hasil dari menyusui [28-30]. Fase lag antara stressor yang mengarah ke reaktivasi dan penumpahan
sebenarnya virus menular adalah sekitar 4-11 hari, dan virus ekskresi berlangsung selama kurang lebih 6 hari ratarata. Virus ekskresi oleh kucing di mana acara pengaktifan berlangsung berkisar 1-13 hari [29 31]. Selama ini virus
menular dapat didemonstrasikan pada sekresi mata dan oronasal. Reaktivasi dapat berupa asimtomatik atau
berhubungan dengan tanda-tanda klinis. Reaktivasi gejala disebut sebagai luapan baru. Reaktivasi DNA virus laten
pada kucing dewasa dapat menyebabkan ulserasi kornea, disertai dengan berbagai tingkat
konjungtivitis [32]. Sejak herpes keratitis stroma disebabkan oleh HSV-1 merupakan penyebab utama kebutaan
menular di negara-negara industri, infeksi mata dari FeHV-1 pada kucing dianggap sebagai model tuan alam yang
sangat baik.
Pemberian kortikosteroid telah dilaporkan menyebabkan reaktivasi di 70 % dari kucing yang terinfeksi secara
laten [3]. Virus menular dilakukan oleh transportasi aksonal anterograde ke jaringan perifer, biasanya untuk sel-sel
di atau dekat tempat infeksi awal, dan merupakan sumber potensi penularan virus [6, 7].
Peran reaktivasi dalam epidemiologi virus herpes alfa adalah langsung berhubungan dengan frekuensi dengan
yang terjadi. Beberapa virus herpes, termasuk FeHV-1, reaksi tivate jauh lebih mudah daripada yang lain dari negara
laten, baik di bawah kondisi alam dan eksperimental. Kemudahan oleh yang laten FeHV-1 DNA diaktifkan kembali
merupakan elemen penting dalam pembenaran FeHV-1 infeksi kucing sebagai model tuan rumah alami untuk
mempelajari patogenesis molekuler dari virus herpes latency dan pendekatan untuk mencegahnya.

7. Diagnosis
klinis, ada tumpang tindih antara simptomatologi akut FeHV-1 dan kucing calicivirus (FCV), penyakit pernapasan
utama lain dari kucing. Fitur yang membedakan dari FeHV-1 infeksi adalah demam tinggi dan ulserasi kornea.
Sebaliknya, borok lidah, langit-langit mulut, dan faring yang lebih khas atau ditemui lebih sering pada infeksi
calicivirus.
Metode diagnostik laboratorium yang paling umum untuk menunjukkan keberadaan FeHV-1 atau komponen
virus dalam homogenat jaringan atau usapan termasuk antibodi fluoresen langsung (FA) tes, isolasi virus (VI) dan
PCR [3, 5, 18].
tes antibodi Fluorescent dilakukan pada Tival conjunc- atau jaringan kornea. Tes ini jauh kurang umum
digunakan sekarang daripada dulu. Fluorescein topikal, digunakan untuk memvisualisasikan bisul, harus dihindari

sebelum sampel mengumpulkan.


Diagnosis Laboratorium FeHV-1 akut kini paling umum dilakukan oleh virus isolasi (VI) atau PCR,
menggunakan oronasal dan ekstrak swab konjungtiva sebagai sampel. VI mendeteksi virus menular dan telah
menjadi laboratorium diag- standar emas nostic [4, 28].
Beberapa tes PCR telah dijelaskan untuk digunakan dalam deteksi FeHV-1 DNA. Sangat baik real-time PCR
assay berbasis TaqMan, dijelaskan oleh V ogtlin et al. [33], menargetkan porsi lestari dari gB gen FeHV-1. Uji ini
bertekad untuk menjadi sangat spesifik untuk FeHV-1, dan batas deteksi adalah antara 0,6 dan 6TCID
50.

Titer virus menular dan DNA virus berkorelasi pada rentang pengenceran lebar.
The real-time PCR (qPCR) dievaluasi pada berurutan col lected ekstrak cairan mata. Awal selama infeksi, disebut
sebagai fase 1, korelasi antara titer virus dan sinyal qPCR sangat tinggi. Selanjutnya, selama apa yang disebut fase 2,
penurunan cepat dalam titer virus menular terlihat, sedangkan sinyal qPCR tetap tinggi. Selama tahap akhir, disebut
sebagai fase 3, virus menular tidak lagi terdeteksi, dan sinyal PCR kuantitatif juga menurun. Analisis deteksi virus
digabungkan dan qPCR hasilnya pada 20 sampel klinis memungkinkan penulis andal menentukan fase infeksi
selama sampel telah dikumpulkan. Menyadari biaya pengujian gabungan, ia menyarankan untuk menguji sampel
berturut-turut oleh qPCR untuk mencapai tujuan ini.
Maggs [4] menunjukkan 3 aspek diagnosis laboratorium FeHV-1 yang bisa jadi sangat frustasi bagi dokter.
Sedangkan konfirmasi akut FeHV-1 tidak selalu diperlukan, penting untuk mengkonfirmasi bahwa lesi kronis
disebabkan oleh FeHV-1. Sayangnya, deteksi FeHV-1 atau komponen virus pada lesi ini bisa sulit. Aspek kedua dari
diagnosis laboratorium yang mengarah ke salah tafsir tions adalah kenyataan bahwa FEHV-1 atau l DNA virus dapat
dideteksi dari sampel kucing klinis normal. Hal itu menunjukkan bahwa deteksi FeHV-1 atau komponennya dapat
cidental bertepatan, konsekuensial, atau kausal. Membedakan antara kemungkinan ini jelas penting.
Virus menetralisir titer antibodi ditentukan oleh tes VN, yang biasanya digunakan untuk mendeteksi infeksi
sebelumnya atau kemanjuran vaksinasi. Virus antibodi bisa rendah dan lambat untuk berkembang. Seperti yang
ditunjukkan oleh Dawson et al. [34], tingkat rendah antibodi tidak berarti tidak adanya perlindungan terhadap
penyakit klinis.

8. Pengobatan dan Pengendalian


8.1. Pengobatan suportif. Pedoman pengelolaan penyakit FeHV-1-diinduksi telah diterbitkan oleh The European
Dewan Penasehat Penyakit Cat (ABCD) [18]. Seperti halnya bagi banyak infeksi virus, terapi suportif sedang
disarankan. Antibiotik spektrum luas yang mencapai penetrasi yang baik ke dalam saluran pernapasan harus istered
admin- dalam semua kasus akut untuk mencegah infeksi bakteri sekunder. Asupan makanan yang enak dan
beraroma juga penting, karena kucing yang terinfeksi mengembangkan anoreksia dari hilangnya indra penciuman
mereka atau, lebih jarang, keberadaan ulkus di rongga mulut. Pada kucing dengan tanda-tanda klinis yang parah,
restorasi cairan, elektrolit, dan keseimbangan asam-basa diperlukan, sebaiknya intravena. Dekongestan nasal, obat
mukolitik, dan pengabutan dengan garam bisa semua tanda-tanda klinis tingkat amelio-. Tetes mata atau salep, bila
digunakan, harus diberikan beberapa kali sehari.
8.2. Antiviral Therapy. Terapi antivirus terdiri dari top antivirus ically atau sistemik diberikan atau penggunaan
terapi ajuvan. Perbandingan 8 obat antivirus diberikan secara topikal menunjukkan bahwa keberhasilan tertinggi
diperoleh dengan trifluridine, berdasarkan potensi dan penetrasi kornea. Kedua dalam efektivitas adalah idoxuridine,
yang memiliki biaya yang lebih rendah dan tampaknya kurang menjengkelkan [4].
Antiviral analog nukleosida biasanya digunakan untuk mengobati infeksi HSV dan VZV. Mereka diubah
menjadi trifosfat oleh virus kinase timidin dan enzim host lain dalam sel yang terinfeksi dan kompetitif menghambat
virus DNA polimerase. Hal ini untuk mencegah rantai DNA perpanjangan [35] dan, sebagai hasilnya, mengganggu
replikasi virus.

Penggunaan agen ini terhadap FeHV-1 infeksi sebagian besar telah terbatas pada pemberian topikal. Generasi
pertama NRTI, termasuk acyclovir dan prodrugyang,
valasiklovir memiliki sedikit efikasi terhadap FeHV-1 in vitro dan efek moderat in vivo. Lebih penting lagi, ketika
administratif tered sistemik mereka menghasilkan efek samping yang serius pada kucing, termasuk mielosupresi,
hepatotoksisitas dan I-City nephrotox- di tingkat terapeutik [36, 37].
Menurut pedoman dari Dewan Penasehat Eropa untuk Penyakit Cat (ABCD), trifluridine adalah pengobatan
topikal pilihan pada kucing dengan mata FHV-1 tions manifestasi. Asiklovir, gansiklovir, dan idoxuridine juga
nyarankan- gested untuk penggunaan topikal. Telah dicatat bahwa, kecuali untuk asiklovir, ada kurangnya
dikendalikan in vivo studi khasiat untuk agen ini dalam literatur [18]. Khasiat kation appli topikal sidofovir pada
mata FeHV-1 infeksi primer telah ditunjukkan [38].
Meskipun penelitian ini tidak terkontrol, trasi administratif oral famsiklovir telah dilaporkan aman dan manjur
dalam mengobati tanda-tanda mata, penyakit kulit, dan rhinosinusitis diinduksi oleh FeHV-1 infeksi [39].
terapi ajuvan yang digunakan untuk mengobati FeHV-1 infeksi adalah L-lisin, laktoferin, dan interferon. Llysine adalah antagonis dari arginin; yang terakhir telah terbukti menjadi penting untuk HSV-1 dan FeHV-1 sintesis
protein [40]. Pengobatan dengan L-lisin, oleh karena itu, menurun replikasi virus dan telah terbukti memiliki
beberapa efek penghambatan terhadap kedua virus herpes manusia dan FeHV-1 infeksi. Masalah dengan konsentrasi
arginin diet rendah adalah kerentanan diucapkan kucing kekurangan arginin [40, 42].
Oral suplementasi dengan L-lisin mengurangi keparahan eksperimen diinduksi FeHV-1 konjungtivitis [42] dan virus
okular penumpahan terkait dengan pengaktifan infeksi laten [40]. Disarankan untuk digunakan pada awal penyakit
akut atau sebagai sarana mengurangi keparahan penyakit dan virus shedding pada waktu stres [3]. Telah
didemonstrasikan bahwa L-Lysine aman pada tingkat dosis oral yang relatif tinggi. Laktoferin adalah glikoprotein
pengikat zat besi mamalia. Telah terbukti [43] untuk menghambat FeHV-1 replikasi in vitro, berpotensi sebagai
akibat dari campur dengan pengikatan FeHV-1 mengikat reseptor seluler dan / atau tion penetra- virus ke dalam sel
rentan.
Interferon adalah sitokin yang dirilis oleh sel darah putih dan mengganggu virus sel-sel menyebar. Interferonalfa (IFN-) administrasi telah terbukti menurunkan tanda-tanda klinis yang terkait dengan infeksi akut [3].

9. Imunitas dan Vaksinasi


Primer infeksi FeHV-1 menginduksi baik respon imun humoral dan seluler. Imunitas aktif disebabkan oleh alam
infeksi FeHV-1 atau imunisasi melindungi kucing dari penyakit, tetapi tidak dari infeksi. Tanda-tanda klinis yang
ringan telah diamati pada reexposure secepat 150 hari setelah infeksi primer [18, 44, 45]. Virus menetralisir titer
antibodi umumnya rendah dan dalam beberapa kasus tidak terdeteksi setelah infeksi primer; meskipun setelah
paparan lebih lanjut untuk virus, mereka cenderung naik ke tingkat yang lebih moderat dan setelahnya tetap cukup
stabil [3, 46]. Sejak FeHV-1 target mata dan saluran pernapasan bagian atas, mukosa respon imun juga memainkan
peran penting [47].

Imunitas pasif berlangsung selama 2 sampai 10 minggu, tergantung pada konsentrasi kolostrum dan asupan.
Beberapa anak kucing dengan rendahnya tingkat antibodi dari ibu yang berasal yang terkena virus lapangan dapat
mengembangkan infeksi subklinis latency dan [48]. Atau, anak kucing tersebut juga akan menanggapi vaksinasi
awal. Sebaliknya, di beberapa anak kucing dari ibu antibodi yang diturunkan cukup tinggi untuk tetap berada di
tingkat campur di 12-14 minggu usia [3, 49].
Rekomendasi Vaksinasi telah disediakan oleh The European Dewan Penasehat untuk Penyakit Cat (ABCD) dan
The American Association . Praktisi Feline Feline Vaksin Advisory panel
panel ABCD merekomendasikan satu atau dua dosis vaksinasi rejimen bangsa awal: dosis pertama diberikan
pada 9 minggu usia dan yang kedua pada 12 minggu usia. Ini diikuti dengan penguat tahunan [18].
The American Association of Praktisi Feline Feline Vaksin Advisory Panel menyarankan bahwa dosis imunisasi
primer harus diberikan sedini usia 6 minggu, dengan dosis tambahan setiap 3 sampai 4 minggu sampai 16 minggu
umur. Dosis penguat adalah untuk diberikan 1 tahun setelah dosis terakhir dari seri utama. Dosis penguat berikutnya
kemudian diberikan setiap 1-3 tahun [50].
Semua vaksin komersial saat melawan FVR juga mengandung kucing calicivirus (FCV) dan virus
panleukopenia kucing (FPV) komponen dan secara kolektif disebut vaksin FVRCP. Perlindungan disebabkan oleh
ini vaksin trivalen umumnya terendah terhadap komponen FeHV-1 [51, 52].
Kedua diubah-hidup dan dilemahkan vaksin FVRCP untuk penggunaan sistemik tersedia di Amerika Serikat [50,
53]. Modified-live vaccines (MLVs) are routinely used, but they have residual virulence and may induce clinical
signs if administered incorrectly [54]. Because of safety concerns, inactivated vaccines are mostly preferred for use
in pregnant queens, and in cats that are infected with feline leukemia virus (FeLV) or feline immunodeficiency virus
(FIV) [50].
In addition to vaccines labeled for systemic immuniza- tion, an intranasal multivalent vaccine containing a
FeHV-1 component is commercially available. Testing under exper- imental conditions showed that this vaccine was
safe and induced protection against the clinical signs of field virus exposure within a week after vaccination [53],
versus 2-3 weeks with a systemically administered vaccine [55].

10. New Approaches to Immunization


10.1. Virulence Genes and Deletion Mutant Vaccines. As discussed earlier, currently available vaccines cannot
totally protect cats from field virus infection and, as a consequence, from field virus latency [5660].
A better understanding of herpesvirus virulence factors is a prerequisite for the generation of safe and efficacious
deletion mutant vaccines. Candidate genes for deletion are those encoding the nonessential glycoproteins gC, gE,
and gG, the US3 gene encoding a protein kinase, the UL 23, gene encoding thymidine kinase. The combination of
BAC cloning of herpesvirus genomes and the introduction of
recombineering to rapidly generate mutants within alpha- herpesviruses cloned as BACs have been very useful tools
to generate mutants with vaccine potential.
Glycoprotein E (gE) is a virulence factor of FeHV-1. Glycoprotein E (gE) and glycoprotein I (gI) form a heterodimer that functions in virus cell-to-cell spread of the virus and transsynaptic spread of infection throughout the
host nervous system, an important component of neurovir- ulence. gE/gI are nonessential glycoproteins, except for
MDV [61]. As an in vitro indicator of reduced virulence, gE/gI mutants have a smaller plaque size and reduced
capacity for cell-to-cell spread [6266]. A functional gE/gI heterodimer appears to play an even greater role in the
spread of VZV [6769].
We previously constructed a gE/gI deletion mutant by conventional in vivo recombination and reported that cats
vaccinated subcutaneously with high doses of the recombi- nant FeHV-1 strain responded with only mild clinical

signs and developed strong immunity against subsequent virulent virus challenge [70]. We also compared the
intranasal and subcutaneous routes of administration of this strain and assessed its ability to induce protective
immunity and prevent virus shedding after challenge. The only concern we had is that this mutant had some residual
virulence when adminis- tered intranasally at high dosage levels [54].
Kaashoek et al. [71] constructed gE-, TK-, and gE-TK- deletion mutants of BoHV-1 and examined their
virulence and immunogenicity in calves. After intranasal inoculation, the TK mutant showed some residual
virulence, whereas the gE and gE-TK mutants were completely avirulent. The calves inoculated with these deletion
mutants were protected against clinical disease after challenge exposure and shed significantly less challenge virus
than control calves.
Recently, an EHV-1 gE mutant was evaluated as a modified live virus (MLV) vaccine. Colostrum-deprived foals
inoculated intranasally (IN) or intramuscularly (IM) with the gE mutant did not exhibit any clinical signs of
respiratory disease except for mild nasal discharge in one of the IN inoculated foals on Days 1 and 3 after infection.
In contrast, foals inoculated IN with the revertant had biphasic fever, mucopurulent nasal discharge, and
submandibular lymph node swelling. The efficacy of the gE mutant against wild type EHV-1 challenge infection
was assessed using foals pre- viously vaccinated twice IM with 105 or 106 plaque-forming units (pfu) of the gEmutant at an interval of 3 weeks. These foals exhibited no respiratory disease signs after IM immu- nization and
developed a good virus neutralizing antibody response to EHV-1 after the second dose. Following a wild- type EHV1 challenge infection, vaccinated foals showed milder clinical symptoms than foals vaccinated with a pla- cebo, and
challenge virus shedding was significantly reduced [72].
The thymidine kinase (TK) gene of alphaherpesviruses is a virulence factor. Comparisons of the amino acid
sequences of herpesvirus TK proteins showed that these proteins are highly divergent, sharing only short regions of
imperfect amino acid identity. Nunberg et al. [73] first identified the TK gene of FeHV-1 using PCR with highly
degenerate oligo- nucleotide primers. Yokoyama et al. [74] inserted the gene encoding the feline calicivirus capsid
protein into the TK locus of FHV-1 and designated the recombinant C730ldfTK- Cap. In a pilot study, 2 cats were
inoculated intranasally and orally with C730ldfTK-Cap, and one cat was inoculated via the same routes with
C730ldfTK. Virus-neutralizing (VN) antibody against both FeHV-1 and FCV was induced with C730ldfTK-Cap,
and against FeHV-1 with C730ldfTK.
The US3 gene of FeHV-1 encodes a serine/threonine pro- tein kinase (PK), and its amino acid sequence is
conserved in the subfamilyAlphaherpesvirinae [7577]. Possible functions of PK include blocking of apoptosis
induced by both viral and cellular proteins [7881], regulation of the nuclear egress of progeny nucleocapsids [82,
83], and control of the morphology of infected cells [84, 85]. Kimman et al. [86] demonstrated that a PK-mutant of
pseudorabies virus (PRV) has strongly reduced virulence, and animals inoculated with PK-gE-PRV mutant and
subsequently challenged with wild- type virus has reduced virus shedding.
Glycoprotein C (gC) homologues have been extensively studied in several alphaherpesviruses. gC homologues
are nonessential for herpesvirus replication in vitro, but they mediate several important biological functions. First of
all, gC is involved in the initial step of viral attachment by interacting with heparan sulfate on cell surface, as
demon- strated in HSV-1, PRV, BHV-1, and EHV-1 [8790]. gC deficient mutants attach to cells with reduced
efficiency [90]. Secondly, gCs of HSV-1 and -2 can bind the complement component C3b [91, 92]. Binding of this
complement factor may protect herpesvirus-infected cells from complement- mediated lysis [93]. Viruses lacking
complement-binding domains are less virulent than wild-type virus [87, 91, 92]. The gC of FeHV-1 has been shown
to be the dominant heparin-binding glycoprotein that mediates the initial stage of viral adsorption, as observed in
other herpesviruses [94]. However, it remains to be determined whether FeHV-1 gC protects virus-infected cells
from complement-mediated lysis.
Willemse et al. [95] first determined a partial sequence of gC. They also found that the adjacent UL45 gene can
be cotranscribed with gC. The complete sequence of FeHV-1 gC was later determined by Maeda et al. [96]. Based
on the amino acid sequence deduced from the nucleotide sequence, they predicted that gC is a membrane

glycoprotein contain- ing a characteristic N-terminal hydrophobic signal sequence, nine potential N-linked
glycosylation sites, and C-terminal transmembrane and cytoplasmic domains. Maeda et al. [97] further demonstrated
that gC is the major heparin-binding glycoprotein involved in the initial step in virus adsorption to cells as observed
in gCs of other herpesviruses. In addition, they found that gC can agglutinate murine red blood cells, and that
infection of FeHV-1 is inhibited by the addition of soluble heparin in cells cultures.
The gG glycoprotein of herpesviruses interacts with chemokines, which are involved in the regulation of leukocyte trafficking and function and the regulation of inflamma- tion and immunosurveillance. The gG glycoprotein of
alpha- herpesviruses can exist in three different forms: membrane- bound full length, membrane bound truncated or
secreted. The full length form, present in FeHV-1 and EHV-1, can
also exist as a truncated secreted form. The secreted form functions as a viral chemokine-binding protein (vCKBP)
and is now classified under the vCKBP-4 subfamily [98].
Van de Walle et al. [99] used EHV-1 as a model to provide the first molecular determination of the residues in
gG of EHV-1 involved in chemokine binding and interaction with target cells. In a very recent study, Thormann et
al. [100] constructed recombinant viruses to show that the ability of the gG of EHV-1 to interfere with chemokine is
not entirely mediated by its chemokine- binding region.
The gG of FeHV-1 exists in booth membrane-bound and secreted forms. The secreted form shows in vitro
binding to bind to a number of chemokines. The membrane bound displays true viroreceptor characteristics [101].
Virulence characteristics of the gG of several alphaher- pesviruses have been investigated. It has been shown
previ- ously that the deletion of gG in PRV does not have a signif- icant effect on viral virulence [102]. In contrast,
the admin- istration of a gG-deleted ILTV to birds, the natural host of this virus, showed that the gG deletion
resulted in significant reduction in virulence. Importantly, virulence could be restored with a revertant, and the
transcription of genes adja- cent to the gG deletion was not affected by the gG deletion. Immunization with the gG
deletion mutant was shown to be protective against virulent virus challenge in experimental birds [103105].
Herpesviruses have multiple immune evasion genes with various evasion mechanisms.
UL49.5 is a gene present in the genome of several mem- bers of the varicellovirus genus, such as EHV-1, BoHV1, and PRV. UL49.5 inhibits transporter associated with antigen processing (TAP) and downregulates cell-surface
expres- sion of major histocompatibility complex (MHC) class I molecules [106].
A BoHV-1 UL49.5 null mutant was shown to no longer have the TAP inhibition and MHC-I downregulation
properties of the parent virus [107]. In a follow-up study, the pathogenicity and immune responses in calves infected
with BoHV-1 UL49.5 null mutant and the parent wild type strain were compared. Both strains replicated similarly in
the nasal epithelium, and both groups had similar clinical scores. BoHV-1 antigen-specific CD8+ T-cell proliferation
as well as CD8+ T-cell cytotoxicity in calves infected with the BoHV-1 UL49.5 null mutant peaked by 7 days after
infection, 1 week earlier than in calves infected with the wild type strain. In addition, virus neutralizing antibody
(VN) titers and IFN- -producing peripheral blood mononuclear cells (PBMCs) in the UL49.5 mutant virus-infected
calves also peaked 7 days and 14 days earlier, respectively. This study indicated that while immune responses peak
earlier, deleting UL49.5 by itself did not sufficiently attenuate this alphaherpesvirus to make it a vaccine candidate
[108].
10.2. Generating Mutants by BAC Clone Recombineering. BAC cloning and recombineering are two state-of-the-art
techniques to facilitate the process of mutagenesis. BACs are single copy F-factor-based plasmid vectors, which can
stably hold 300 kb or more of foreign DNA [109]. The BACs' larger capacity and greater stability over the other
vectors have enabled the cloning of an entire herpesvirus genome into a single plasmid. These properties have also
made BAC the vector of choice for the cloning of herpesvirus genomes.
Recombineering is a powerful method for fast and efficient manipulation of the BAC. It allows DNA cloned in
E. coli to be modified via lambda () red-mediated homologous recombination, obviating the need for restriction
enzymes and DNA ligases. Specific bacterial strains, for example, E. coli SW105, have been constructed for this

purpose [110 112]. A defective prophage (mini-) is inserted into the E. coli genome and encodes heat-shock
inducible genes that make recombineering possible. Linear DNA (PCR product, oligonucleotide, etc.,) with
sufficient homology in the 5 and 3 ends to a target DNA molecule already present in the bacteria (plasmid, BAC, or
the bacterial genome itself) can be electroporated into heat-shocked and electrocompetent bac- teria cells and
undergoes homologous recombination with the target molecule. Utilizing recombineering techniques, site-specific
mutations can be introduced anywhere in the viral genome. All mutagenesis steps can be strictly controlled and
analyzed in E. coli, and the manipulated viral genome can be stably maintained in the E. coli.
The entire FeHV-1 genome was previously cloned as a BAC in our lab, from which the complete FeHV-1
genomic sequence was derived [14]. The BAC-cloned virus was characterized in vitro and in vivo. Prior to defining
the in vitrogrowth characteristics of the BAC-cloned virus, the BAC cassette was excised from the cloned virus
genome. We then performed plaque size analysis and constructed multiple- step growth curves for the FeHV1BAC and its C-27 parent strain. Plaques produced by the C-27 strain and FeHV- 1BAC virus were
morphologically undistinguishable from each other. The mean plaque diameter of the FeHV-1BAC virus was
101.05% of that of the C-27 parent strain and not significantly different. Multistep growth curve analysis showed
that they can grow to a similar titer.
To investigate possible attenuation resulting from BAC cloning itself, a preliminary challenge experiment was
car- ried out, using four specific-pathogen-free (SPF) cats. Two cats were inoculated intranasally with the FeHV1BAC virus, and the other two cats were inoculated intranasally with either the C-27 strain or cell culture medium.
The main conclusion from the in vivo experiment was that the BAC clone-derived virus behaved very similarly to
its C-27 parent strain both in vitro and in vivo, making it an excellent start- ing platform for introducing mutations
aimed at deleting virulence-inducing genes from the FeHV-1 genome [14].

11. Mucosal Vaccination and


Epitope-Based Vaccines
A major goal of strategies to immunize against alphaher- pesvirus infections is to prevent primary infection, which
would in turn prevent primary disease and the establishment of latency and subsequent latency reactivation. Latency
reactivation has been shown to occur frequently, leading to virus shedding, which is asymptomatic in most cases.
Natural
infection provides protection against reinfection of primary mucosal replication sites for a certain period of time.
This provides a rationale for the development of immunization strategies at the mucosal level.
Innate immune responses are the first to develop after natural infection. The recognition of alphaherpesvirus
com- ponents by toll-like receptors is an important mechanism for induction of these responses. HSV and its
components bind to TLR 2, 3, 7, and 9. Synthetic agonists have been designed to transiently activate the innate
immune response [113].
In human medicine, the majority of the efforts to develop immunization strategies against herpesvirus infections
have been focused on the prevention of genital herpes. However, it is also well recognized that ocular HSV-1
infection is a leading worldwide cause of herpetic keratitis, which can lead to corneal blindness. Like is the case for
FeHV-1, the most severe ocular HSV-1 infections are the result of repeated reactivation events. It is clear that
mucosal delivery is the best approach to generate secretory immunity and cytotoxic T- cell responses at mucosal
sites.
Long-term efforts to immunize against human alphaher- pesvirus infections have included subunit vaccines,
modi- fied-live vaccines, replication-defective vaccines, viral vector vaccines, and naked DNA vaccines. Despite
these efforts, there are no licensed vaccines available.
One of the current approaches to mucosal immunization focuses on the development of a multiepitope selfadjuvant lipopeptide vaccine. A recent overview of this approach by the group that has pioneered it highlights its
promise, but also the hurdles that still have to be overcome [114]. They point out that, based upon recent trials, the

induction of neu- tralizing antibodies is not sufficient for protection. Implied from these results is that the induction
of appropriate and adequate protective T-cell responses is a crucial part of the development of protective immunity.
The essential compo- nents of a protective immune response can be the prevention of primary infection or the
prevention or reduction of reac- tivation events. It is clear from their work, and that of others, that individuals that
are latently infected with HSV, have frequent reactivation events associated with virus shedding. This reactivation is
not associated with clinical signs in most individuals, which are therefore termed asymptomatic individuals. In
contrast, the term symptomatic individuals is used for those in which frequent reactivation is associated with clinical
signs. An important element of the strategy is to characterize the unique T-cell repertoire in HSV-positive individuals
that do not suffer from frequent symptomatic reactivation. It has been determined that a set of human T- cell
epitopes from HSV-1 gB and gD are strongly recognized by T-cells from asymptomatic individuals, but not by T
cells from symptomatic individuals. In contrast, another nonoverlapping set of gB and gD epitopes is recognized by
symptomatic individuals. The results of recent immu- nization of asymptomatic HLA transgenic rabbits showed that
immunization with asymptomatic CD8+ epitopes from HSV-1 gD induced strong CD8+ immune responses and
reduced HSV-1 shedding and tears and corneal lesions fol- lowing ocular challenge virus administration.
The authors emphasize repeatedly that the following five existing hurdles need to be overcome: (1) reasons for
subop- timal immunity resulting from natural infection, (2) optimal effector mechanisms for protective immunity
against the acute and latent phases of the disease, (3) knowledge about immunoevasive strategies, (4) distinction
between protective versus pathogenic antigens, and (5) design of a an appropri- ate vaccine delivery system. They
recognize that candidate vaccines need to be tested in relevant animal models if they cannot be directly evaluated in
the natural host. An already existing human HLA transgenic rabbit model and the development of a similar guinea
pig model are crucial tools in this respect.

12. In Vitro Approaches to


Molecular Pathogenesis
Ocular infection with FeHV-1 results from viral exposure of conjunctival and corneal tissue. Since corneal lesions
are an important disease manifestation, both during both the acute phase and as a result of reactivation, finding an
effective therapy against development of ocular disease has high priority. Sandmeyer et al. [115] have reported the
develop- ment of primary corneal cell culture system, which is useful for in vitro pathogenesis of ocular disease and
also for the testing of potential antivirals [116]. Using this system they showed that IFN- was not toxic to ocular
cells and had a limited effect of virus production in FeHV-1-infected corneal cells. They speculated that a
combination of IFN- and other antivirals may act synergistically [117].
Pathogenesis studies of FeHV-1 have almost exclusively been done on live animals. Since the tracheal mucosa is
an important replication site of FeHV-1, tracheal organ cultures are a good in vitro model to study viral invasiveness
and local immune responses. Leeming et al. [118] established feline tracheal organ cultures and showed that these
could be maintained for at least 5 days. Infection of these cultures at different multiplicities of infection (MOI),
ranging from 0.1 to 100, showed that the virus replicated extensively in these cultures and produced coalescing
necrosis of tracheal epithe- lium and disruption of ciliary activity.
Since mucosal surfaces are the primary replication sites of FeHV-1, it is important to understand viral replication
strategies and the local immune responses generated at these sites to better combat this mucosal pathogen. As
indicated earlier, it is well known that systemically administered vac- cines can prevent clinical signs but cannot
prevent reinfec- tion and the associated development of latency.
Quintana et al. [119] recently developed an equine respi- ratory epithelium cell culture system consisting of
culturing dissociated primary epithelial cells at a liquid air interface. This is a meaningful in vitro system since
epithelial cells not only provide a physical barrier against viral invasion, but also play a significant role in
development of immunity by expressing toll-like receptors, by secreting cytokines, chemo- kines, and host defense
peptides and by playing some role in antigen presentation. It was shown that epithelial cell cul- tures grown under
these conditions were morphologically

similar to intact airway epithelium. These cultures were also shown to be immunologically competent, but some
proper- ties were altered by in vitro culture under sterile conditions. The authors concluded that the addition of
antigenic stimuli and/or immune cells could reverse this situation.
Mucosal explants have recently been shown for several herpesviruses to be an excellent system to study kinetics
of viral invasion, as determined by the ability of a particular herpesvirus to get across the epithelial basement
membrane. This system has been used to compare the invasiveness of different herpesviruses. It can, however, also
be used for strain comparison and to study the role of individual or combinations of viral genes as determinants of
viral viru- lence [120123].
FeHV-1 infection of cats is an excellent natural host model to study mechanisms involved in establishment,
maintenance, and reactivation of latency. As discussed above, latency is established in all cats following natural
infection and is readily reactivated by a variety of natural stimuli or administration of corticosteroids.
De Regge et al. [124] reported the development of a homologous in vitro model to study the interaction of alphaherpesviruses and trigeminal ganglion neurons. The system consists of two concentric culture chambers. The inner
and outer chambers are separated from one another by a silicon barrier, which is impermeable to both virus and cell
culture medium. After 2-3 weeks in culture, axons from neuronal cell bodies present in the inner chamber grow
through the silicon barrier into the outer chamber. Infection of these axons, either with HSV-1 or PRV, exclusively
led to infection of neurons in the inner chamber and the subsequent spread of infection from these neurons to other
neurons and nonneu- ronal cells. This system thus allows an in vivo-like infection of neuronal cells via retrograde
axonal transport. It is, there- fore, very useful to study mechanisms involved in latency establishment, maintenance,
and reactivation. In a follow- up study, De Regge et al. [125] used this system to examine the role of IFN-, an
important component of the innate immune system. The data showed that IFN- was indeed able to establish latency
in these cultures and that latency was maintained after its removal. LAT transcripts, a prominent feature of latency,
were detected in the cultures by RT-PCR and the latent viral DNA could be reactivated by treatment with forskolin.

13. A New Approach to Antiviral Therapy


Ribonucleic acid interference (RNAi), initiated by chemi- cally synthesized 21-mer or 27-mer small interfering
RNAs (siRNA), is an alternative method to the use of standard antiviral therapy. Wilkes and Kania [126] have
explored the potential of this method in vitro. The initial target was gD- specific mRNA, based upon the fact that the
gD glycoprotein plays an important role in viral attachment to susceptible cells and also in the induction of
protective neutralizing anti- body responses. Two of the six siRNAs they tested induced a significant reduction of
virus replication in CRFK cells infected with FeHV-1. In a follow-up study Wilkes and
Kania [127] selected siRNAs specific for the FeHV-1 DNA polymerase mRNA, the gD mRNA, or a combination of
both. The hypothesis behind the targeting of the DNA polymerase was that more complete inhibition of viral
replication would occur when an early rather than a late transcript was targeted. This proved to be the case, since the
highest level of inhibition was obtained with a combination of 2 siRNAs targeting the FeHV-1 DNA polymerase
transcript. Potential in vivo use of this approach is based upon the fact that siRNAs can be taken up effectively when
applied to mucosal surfaces.

14. Conclusions
Recent expansion in our molecular knowledge of FeHV-1 will ultimately not only be of benefit to the health of
domestic cats but will also contribute to our understanding of shared aspects of herpesvirus biology. FeHV-1
infection in cats also has potential, as a natural host system, to develop more effective immunization and treatment
procedures against alphaherpesvirus infections in animals and humans.

References
[1] AJ Davison, R. Eberle, B. Ehlers et al., The order Herpesvi- rales, Archives of Virology, vol. 154, no. 1, pp. 171177, 2009.
[2] N. James Maclachlan and J. Edward Dubovi, Eds., Fenner's Veterinary Virology, Academic Press Elsevier, 4th edition, 2001.
[3] R. Gaskell, S. Dawson, A. Radford, and E. Thiry, Feline herpesvirus, Veterinary Research, vol. 38, tidak ada. 2, pp. 337

354, 2007. [4] DJ Maggs, Update on pathogenesis, diagnosis, and treat- ment of feline herpesvirus type 1, Clinical Techniques
in Small Animal Practice, vol. 20, tidak ada. 2, pp. 94101, 2005. [5] D. Gould, Feline Herpesvirus-1. Ocular manifestations,
diagnosis and treatment options, Journal of Feline Medicine and Surgery, vol. 13, tidak ada. 5, pp. 333346, 2011. [6] PE Pellett
and B. Roizman, The family Herpesviridae: a brief introduction, in Fields Virology, DM Knipe and PM Howley, Eds., pp.
24792499, Lippincott Williams & Wilkins, Philadelphia, Pa, USA, 2007. [7] B. Roizman, DM Knipe, and RJ Whitley, Herpes
simplex viruses, in Fields Virology, DM Knipe and PM Howley, Eds., pp. 25022601, Lippincott Williams & Wilkins,
Philadelphia, Pa, USA, 2007. [8] RC Povey, A review of feline viral rhinotracheitis (feline herpesvirus I infection),
Comparative Immunology, Micro- biology and Infectious Diseases, vol. 2, tidak ada. 2-3, pp. 373387, 1979. [9] TC Harder, M.
Harder, H. Vos et al., Characterization of phocid herpesvirus-1 and -2 as putative alpha- and gamma- herpesviruses of North
American and European pinnipeds, Journal of General Virology, vol. 77, tidak ada. 1, pp. 2735, 1996. [10] TC Harder, M.
Harder, RL De Swart, ADME Osterhaus, and B. Liess, Major immunogenic proteins of phocid herpes-viruses and their
relationships to proteins of canine and feline herpesviruses, Veterinary Quarterly, vol. 20, tidak ada. 2, pp. 5055, 1998. [11] TC
Harder and ADME Osterhaus, Molecular charac- terization and baculovirus expression of the glycoprotein B of a seal
herpesvirus (phocid herpesvirus-1), Virology, vol. 227, no. 2, pp. 343352, 1997.
[12] PA Rota, RK Maes, and WT Ruyechan, Physical char- acterization of the genome of feline herpesvirus-1, Virology, vol.
154, no. 1, pp. 168179, 1986. [13] A. Grail, DA Harbour, and W. Chia, Restriction endonu- clease mapping of the genome of
feline herpesvirus type 1, Archives of Virology, vol. 116, no. 14, pp. 209220, 1991. [14] SHS Tai, M. Niikura, HH Cheng, JM
Kruger, AG Wise, and RK Maes, Complete genomic sequence and an infectious BAC clone of feline herpesvirus-1 (FHV-1),
Virology, vol. 401, no. 2, pp. 215227, 2010. [15] D. Fargeaud, C. Benoit Jeannin, F. Kato, and G. Chappuis, Biochemical study
of the feline herpesvirus 1. Identification of glycoproteins by affinity, Archives of Virology, vol. 80, no. 2-3, pp. 6982, 1984.
[16] K. Maeda, T. Horimoto, and T. Mikami, Properties and functions of feline herpesvirus type 1 glycoproteins, Journal of
Veterinary Medical Science, vol. 60, tidak ada. 8, pp. 881888, 1998. [17] MP Nasisse, JS Guy, MG Davidson, WA Sussman, and
NM Fairley, Experimental ocular herpes virus infection in the cat. Sites of virus replication, clinical features and effects of
corticosteroid administration, Investigative Ophthalmol- ogy and Visual Science, vol. 30, tidak ada. 8, pp. 17581768, 1989.
[18] E. Thiry, D. Addie, S. Bel ak et al., Feline herpesvirus infection ABCD guidelines on prevention and management, Journal
of Feline Medicine and Surgery, vol. 11, tidak ada. 7, pp. 547 555, 2009. [19] C. Hartley, Aetiology of Corneal ulcers. Assume
FHV-1 unless proven otherwise, Journal of Feline Medicine and Sur- gery, vol. 12, tidak ada. 1, pp. 2435, 2010. [20] GC Perng
and C. Jones, Towards an understanding of the herpes simplex virus type 1 latency-reactivation cycle, Interdisciplinary
Perspectives on Infectious Diseases, vol. 2010, Article ID 262415, 18 pages, 2010. [21] C. Jones, Herpes simplex virus type 1
and bovine her- pesvirus 1 latency, Clinical Microbiology Reviews, vol. 16, no. 1, pp. 7995, 2003. [22] C. Jones and S.
Chowdhury, A review of the biology of bovine herpesvirus type 1 (BHV-1), its role as a cofactor in the bovine respiratory
disease complex and development of improved vaccines, Animal Health Research Reviews, vol. 8, tidak ada. 2, pp. 187205,
2007. [23] JR Kent, W. Kang, CG Miller, and NW Fraser, Herpes simplex virus latency-associated transcript gene function,
Journal of NeuroVirology, vol. 9, tidak ada. 3, pp. 285290, 2003. [24] A. Gupta, JJ Gartner, P. Sethupathy, AG Hatzigeorgiou,
and NW Fraser, Anti-apoptotic function of a microRNA encoded by the HSV-1 latency-associated transcript, Nature, vol. 442,
no. 7098, pp. 8285, 2008. [25] RL Thompson, CM Preston, and NM Sawtell, De novo synthesis of VP16 coordinates the exit
from HSV latency in vivo, PLoS Pathogens, vol. 5, tidak ada. 3, 2009. [26] S. Jacobi, Molecular pathology of FHV-1 infection
within feline cornea, conjunctiva, lacrimal gland, nictitans gland, trigeminal ganglion and ciliary ganglion [Ph.D. thesis],
Michigan State University, 2008. [27] WM Townsend, J. Stiles, L. Guptill-Yoran, and SG Krohne, Development of a reverse
transcriptase-polymerase chain reaction assay to detect feline herpesvirus-1 latency-asso- ciated transcripts in the trigeminal
ganglia and corneas of cats that did not have clinical signs of ocular disease, Ameri- can Journal of Veterinary Research, vol. 65,
no. 3, pp. 314319, 2004.
[28] TM Ellis, Feline respiratory virus carriers in clinically healthy cats, Australian Veterinary Journal, vol. 57, no. 3, pp. 115
118, 1981. [29] RM Gaskell and RC Povey, Experimental induction of feline viral rhinotracheitis virus re-excretion in FVRrecovered cats, Veterinary Record, vol. 100, tidak ada. 7, pp. 128 133, 1977. [30] RM Gaskell and RC Povey, Transmission of
feline viral rhinotracheitis, Veterinary Record, vol. 111, no. 16, pp. 359 362, 1982. [31] RM Gaskell and RC Povey,
Reexcretion of feline viral rhinotracheitis virus following corticosteroid treatment, Veterinary Record, vol. 93, tidak ada. 7, pp.
204205, 1973. [32] J. Stiles, Feline herpesvirus, Veterinary Clinics of North
America, vol. 30, tidak ada. 5, pp. 10011014, 2000. [33] A. V ogtlin, C. Fraefel, S. Albini et al., Quantification of feline
herpesvirus 1 DNA in ocular fluid samples of clinically diseased cats by real-time TaqMan PCR, Journal of Clinical
Microbiology, vol. 40, tidak ada. 2, pp. 519523, 2002. [34] DA Dawson, J. Carman, J. Collins, S. Hill, and MR Lappin,
Enzyme-linked immunosorbent assay for detection of feline herpesvirus 1 IgG in serum, aqueous humor, and cere- brospinal
fluid, Journal of Veterinary Diagnostic Investiga- tion, vol. 10, tidak ada. 4, pp. 315319, 1998. [35] R. Snoeck, Antiviral

therapy of herpes simplex, Interna- tional Journal of Antimicrobial Agents, vol. 16, no. 2, pp. 157 159, 2000. [36] MP Nasisse,
DC Dorman, KC Jamison, BJ Weigler, EC Hawkins, and JB Stevens, Effects of valacyclovir in cats infected with feline
herpesvirus 1, American Journal of Veterinary Research, vol. 58, no. 10, pp. 11411144, 1997. [37] JG Owens, MP Nasisse, SM
Tadepalli, and DC Dorman, Pharmacokinetics of acyclovir in the cat, Journal of Veterinary Pharmacology and Therapeutics,
vol. 19, tidak ada. 6, pp. 488490, 1996. [38] JP Fontenelle, CC Powell, JK Veir, SV Radecki, and MR Lappin, Effect of topical
ophthalmic application of cidofovir on experimentally induced primary ocular feline herpesvirus-1 infection in cats, American
Journal of Veteri- nary Research, vol. 69, no. 2, pp. 289293, 2008. [39] R. Malik, NS Lessels, S. Webb et al., Treatment of
feline herpesvirus-1 associated disease in cats with famciclovir and related drugs, Journal of Feline Medicine and Surgery, vol.
11, tidak ada. 1, pp. 4048, 2009. [40] DJ Maggs, MP Nasisse, and PH Kass, Efficacy of oral supplementation with L-lysine in
cats latently infected with feline herpesvirus, American Journal of Veterinary Research, vol. 64, tidak ada. 1, pp. 3742, 2003.
[41] DJ Maggs, BK Collins, JG Thorne, and MP Nasisse, Effects of L-lysine and L-arginine on in vitro replication of feline
herpesvirus type-1, American Journal of Veterinary Research, vol. 61, tidak ada. 12, pp. 14741478, 2000. [42] J. Stiles, WM
Townsend, QR Rogers, and SG Krohne, Effect of oral administration of L-lysine on conjunctivitis caused by feline herpesvirus
in cats, American Journal of Veterinary Research, vol. 63, tidak ada. 1, pp. 99103, 2002. [43] SL Beaumont, DJ Maggs, and HE
Clarke, Effects of bovine lactoferrin on in vitro replication of feline her- pesvirus, Veterinary Ophthalmology, vol. 6, tidak ada.
3, pp. 245 250, 2003. [44] RM Gaskell and RC Povey, Experimental induction of feline viral rhinotracheitis virus re-excretion
in FVR- recovered cats, Veterinary Record, vol. 100, tidak ada. 7, pp. 128 133, 1977.
[45] TE Walton and JH Gillespie, Feline viruses. VII. Immu- nity to the feline herpesvirus in kittens inoculated experi- mentally
by the aerosol method, The Cornell Veterinarian, vol. 60, tidak ada. 2, pp. 232239, 1970. [46] RM Gaskell and RC Povey, The
dose response of cats to experimental infection with feline viral rhinotracheitis virus, Journal of Comparative Pathology, vol. 89,
no. 2, pp. 179191, 1979. [47] MR Lappin, RW Sebring, M. Porter, SJ Radecki, and J. Veir, Effects of a single dose of an
intranasal feline her- pesvirus 1, calicivirus, and panleukopenia vaccine on clinical signs and virus shedding after challenge with
virulent feline herpesvirus 1, Journal of Feline Medicine and Surgery, vol. 8, tidak ada. 3, pp. 158163, 2006. [48] RM Gaskell
and RC Povey, Transmission of feline viral rhinotracheitis, Veterinary Record, vol. 111, no. 16, pp. 359 362, 1982. [49] S.
Dawson, K. Willoughby, RM Gaskell, G. Wood, and WS Chalmers, A field trial to assess the effect of vaccina- tion against
feline herpesvirus, feline calicivirus and feline panleucopenia virus in 6-week-old kittens, Journal of Feline Medicine and
Surgery, vol. 3, tidak ada. 1, pp. 1722, 2001. [50] JR Richards, TH Elston, RB Ford et al., The 2006 American association of
feline practitioners Feline Vaccine Advisory Panel report, Journal of the American Veterinary Medical Association, vol. 229, no.
9, pp. 14051441, 2006. [51] MR Lappin, J. Andrews, D. Simpson, and WA Jensen, Use of serologic tests to predict resistance
to feline herpesvirus 1, feline calicivirus, and feline parvovirus infection in cats, Journal of the American Veterinary Medical
Association, vol. 220, no. 1, pp. 3842, 2002. [52] FW Scott and CM Geissinger, Long-term immunity in cats vaccinated with
an inactivated trivalent vaccine, American Journal of Veterinary Research, vol. 60, tidak ada. 5, pp. 652658, 1999. [53] MR
Lappin, RW Sebring, M. Porter, SJ Radecki, and J. Veir, Effects of a single dose of an intranasal feline her- pesvirus 1,
calicivirus, and panleukopenia vaccine on clinical signs and virus shedding after challenge with virulent feline herpesvirus 1,
Journal of Feline Medicine and Surgery, vol. 8, tidak ada. 3, pp. 158163, 2006. [54] JM Kruger, MD Sussman, and RK Maes,
Glycoproteins gI and gE of feline herpesvirus-1 are virulence genes: Safety and efficacy of a gI-gE- deletion mutant in the
natural host, Virology, vol. 220, no. 2, pp. 299308, 1996. [55] MR Lappin, J. Veir, and J. Hawley, Feline panleukopenia virus,
feline herpesvirus-1, and feline calicivirus antibody responses in seronegative specific pathogen-free cats after a single
administration of two different modified live FVRCP vaccines, Journal of Feline Medicine and Surgery, vol. 11, tidak ada. 2, pp.
159162, 2009. [56] RM Gaskell, Upper respiratory disease in the cat (includ- ing chlamydia): control and prevention, Feline
Practice, vol. 21, pp. 2934, 1993. [57] DA Harbour, PE Howard, and RM Gaskell, Isolation of feline calicivirus and feline
herpesvirus from domestic cats 1980 to 1989, Veterinary Record, vol. 128, no. 4, pp. 7780, 1991. [58] KM Tham and MJ
Studdert, Clinical and immunological responses of cats to feline herpesvirus type 1 infection, Veterinary Record, vol. 120, no.
14, pp. 321326, 1987. [59] BJ Weigler, JS Guy, MP Nasisse, SI Hancock, and B. Sherry, Effect of a live attenuated intranasal
vaccine on latency and shedding of feline herpesvirus 1 in domestic cats, Archives of Virology, vol. 142, no. 12, pp. 23892400,
1997. [60] N. Yokoyama, K. Maeda, Y. Tohya et al., Pathogenicity and vaccine efficacy of a thymidine kinase-deficient mutant
of feline herpesvirus type 1 in cats, Archives of Virology, vol. 141, no. 3-4, pp. 481494, 1996. [61] D. Schumacher, B. Karsten
Tischer, SM Reddy, and N. Osterrieder, Glycoproteins e and I of marek's disease virus serotype 1 are essential for virus growth
in cultured cells, Journal of Virology, vol. 75, no. 23, pp. 1130711318, 2001. [62] P. Balan, N. Davis-Poynter, S. Bell, H.
Atkinson, H. Browne, and T. Minson, An analysis of the in vitro and in vivo phenotypes of mutants of herpes simplex virus type
1 lacking glycoproteins gG, gE, gI or the putative gJ, Journal of General Virology, vol. 75, no. 6, pp. 12451258, 1994. [63] KS
Dingwell, CR Brunetti, RL Hendricks et al., Herpes simplex virus glycoproteins E and I facilitate cell-to-cell spread in vivo and
across junctions of cultured cells, Journal of Virology, vol. 68, no. 2, pp. 834845, 1994. [64] KS Dingwell and DC Johnson,

The herpes simplex virus gE-gI complex facilitates cell-to-cell spread and binds to components of cell junctions, Journal of
Virology, vol. 72, no. 11, pp. 89338942, 1998. [65] H. Otsuka and X. Xuan, Construction of bovine herpes- virus-1 (BHV-1)
recombinants which express pseudorabies virus (PRV) glycoproteins gB, gC, gD, and gE, Archives of Virology, vol. 141, no. 1,
pp. 5771, 1996. [66] FA Zuckermann, TC Mettenleiter, C. Schreurs, N. Sugg, and T. Ben-Porat, Complex between
glycoproteins gI and gp63 of pseudorabies virus: its effect on virus replication, Journal of Virology, vol. 62, tidak ada. 12, pp.
46224626, 1988. [67] RJ Frink, R. Eisenberg, G. Cohen, and EK Wagner, Detailed analysis of the portion of the herpes simplex
virus type 1 genome encoding glycoprotein C, Journal of Virology, vol. 45, no. 2, pp. 634647, 1983. [68] S. Mallory, M.
Sommer, and AM Arvin, Mutational analysis of the role of glycoprotein I in varicella-zoster virus replication and its effects on
glycoprotein E conformation and trafficking, Journal of Virology, vol. 71, no. 11, pp. 8279 8288, 1997. [69] S. Mallory, M.
Sommer, and AM Arvin, Analysis of the glycoproteins I and E of varicella-zoster virus (VZV) using deletional mutations of
VZV cosmids, Journal of Infectious Diseases, vol. 178, no. 5, supplement 1, pp. S22S26, 1998. [70] MD Sussman, RK Maes,
JM Kruger, SJ Spatz, and PJ Venta, A feline herpesvirus-1 recombinant with a deletion in the genes for glycoproteins gI and gE
is effective as a vaccine for feline rhinotracheitis, Virology, vol. 214, no. 1, pp. 1220, 1995. [71] MJ Kaashoek, FAC Van
Engelenburg, A. Moerman, ALJ Gielkens, FAM Rijsewijk, and JT Van Oirschot, Vir- ulence and immunogenicity in calves of
thymidine kinase- and glycoprotein E-negative bovine herpesvirus 1 mutants, Veterinary Microbiology, vol. 48, tidak ada. 1-2,
pp. 143153, 1996. [72] K. Tsujimurai, T. Shiosei, T. Yamanakai, M. Nemotoi, T. Kondoi, and T. Matsumurai, Equine
herpesvirus type 1 mutant defective in glycoprotein E gene as candidate vaccine strain, The Journal of Veterinary Medical
Science, vol. 71, pp. 14391448, 2009. [73] JH Nunberg, DK Wright, GE Cole et al., Identification of the thymidine kinase gene
of feline herpesvirus: use of degenerate oligonucleotides in the polymerase chain reaction to isolate herpesvirus gene homologs,
Journal of Virology, vol. 63, tidak ada. 8, pp. 32403249, 1989.
[74] N. Yokoyama, K. Maeda, Y. Tohya, Y. Kawaguchi, K. Fujita, and T. Mikami, Recombinant feline herpesvirus type 1
expressing immunogenic proteins inducible virus neutraliz- ing antibody against feline calicivirus in cats, Vaccine, vol. 14, tidak
ada. 17-18, pp. 16571663, 1996. [75] MC Frame, FC Purves, DJ McGeoch, HS Marsden, and DP Leader, Identification of the
herpes simplex virus protein kinase as the product of viral gene US3, Journal of General Virology, vol. 68, part 10, pp. 2699
2704, 1987. [76] DJ Mcgeoch and AJ Davison, Alphaherpesviruses possess a gene homologous to the protein kinase gene
family of eukaryotes and retroviruses, Nucleic Acids Research, vol. 14, tidak ada. 4, pp. 17651777, 1986. [77] FC Purves, RM
Longnecker, DP Leader, and B. Roizman, Herpes simplex virus 1 protein kinase is encoded by open reading frame US3 which is
not essential for virus growth in cell culture, Journal of Virology, vol. 61, tidak ada. 9, pp. 28962901, 1987. [78] R. Leopardi,
C. Van Sant, and B. Roizman, The herpes simplex virus 1 protein kinase Us3 is required for protection from apoptosis induced
by the virus, Proceedings of the National Academy of Sciences of the United States of America, vol. 94, no. 15, pp. 78917896,
1997. [79] J. Munger, AV Chee, and B. Roizman, The Us3 protein kinase blocks apoptosis induced by the d120 mutant of herpes
simplex virus 1 at a premitochondrial stage, Journal of Virology, vol. 75, no. 12, pp. 54915497, 2001. [80] J. Munger and B.
Roizman, The Us3 protein kinase of herpes simplex virus 1 mediates the posttranslational modification of BAD and prevents
BAD-induced programmed cell death in the absence of other viral proteins, Proceedings of the National Academy of Sciences of
the United States of America, vol. 98, no. 18, pp. 1041010415, 2001. [81] PD Ogg, PJ McDonell, BJ Ryckman, CM Knudson,
and RJ Roller, The HSV-1 Us3 protein kinase is sufficient to block apoptosis induced by overexpression of a variety of Bcl-2
family members, Virology, vol. 319, no. 2, pp. 212224, 2004. [82] AE Reynolds, BJ Ryckman, JD Baines, Y. Zhou, L. Liang,
and RJ Roller, UL31 and UL34 proteins of herpes simplex virus type 1 form a complex that accumulates at the nuclear rim and
is required for envelopment of nucleocapsids, Journal of Virology, vol. 75, no. 18, pp. 88038817, 2001. [83] AE Reynolds, EG
Wills, RJ Roller, BJ Ryckman, and JD Baines, Ultrastructural localization of the herpes simplex virus type 1 UL31, UL34, and
US3 proteins suggests specific roles in primary envelopment and egress of nucleocapsids, Journal of Virology, vol. 76, no. 17,
pp. 89398952, 2002. [84] A. Kato, M. Tanaka, M. Yamamoto et al., Identification of a physiological phosphorylation site of the
herpes simplex virus 1-encoded protein kinase Us3 which regulates its optimal catalytic activity in vitro and influences its
function in infected cells, Journal of Virology, vol. 82, no. 13, pp. 6172 6189, 2008. [85] T. Murata, F. Goshima, Y. Nishizawa
et al., Phosphorylation of cytokeratin 17 by herpes simplex virus type 2 US3 protein kinase, Microbiology and Immunology,
vol. 46, tidak ada. 10, pp. 707719, 2002. [86] TG Kimman, N. de Wind, T. de Bruin, Y. de Visser, and J. Voermans, Inactivation
of glycoprotein gE and thymidine kinase or the US3-encoded protein kinase synergistically decreases in vivo replication of
pseudorabies virus and the induction of protective immunity, Virology, vol. 205, no. 2, pp. 511518, 1994.
[87] BC Herold, D. WuDunn, N. Soltys, and PG Spear, Glyco- protein C of herpes simplex virus type 1 plays a principal role in
the adsorption of virus to cells and in infectivity, Journal of Virology, vol. 65, no. 3, pp. 10901098, 1991. [88] TC Mettenleiter,
L. Zsak, F. Zuckermann, N. Sugg, H. Kern, and T. Ben-Porat, Interaction of glycoprotein gIII with a cellular heparinlike
substance mediates adsorption of pseudorabies virus, Journal of Virology, vol. 64, tidak ada. 1, pp. 278286, 1990. [89] K.
Okazaki, T. Matsuzaki, Y. Sugahara et al., BHV-1 adsorp- tion is mediated by the interaction of glycoprotein gIII with

heparinlike moiety on the cell surface, Virology, vol. 181, no. 2, pp. 666670, 1991. [90] N. Osterrieder, Construction and
characterization of an equine herpesvirus 1 glycoprotein C negative mutant, Virus Research, vol. 59, no. 2, pp. 165177, 1999.
[91] RJ Frink, R. Eisenberg, G. Cohen, and EK Wagner, Detailed analysis of the portion of the herpes simplex virus type 1
genome encoding glycoprotein C, Journal of Virology, vol. 45, no. 2, pp. 634647, 1983. [92] J. Lubinski, L. Wang, D.
Mastellos, A. Sahu, JD Lambris, and HM Friedman, In vivo role of complement-interacting domains of herpes simplex virus
type 1 gC, Journal of Experimental Medicine, vol. 190, no. 11, pp. 16371646, 1999. [93] LF Fries, HM Friedman, and GH
Cohen, Glycoprotein C of herpes simplex virus 1 is an inhibitor of the complement cascade, Journal of Immunology, vol. 137,
no. 5, pp. 1636 1641, 1986. [94] K. Maeda, N. Yokoyama, K. Fujita, M. Maejima, and T. Mikami, Heparin-binding activity of
feline herpesvirus type 1 glycoproteins, Virus Research, vol. 52, no. 2, pp. 169176, 1997. [95] MJ Willemse, WSK Chalmers,
AM Cronenberg, R. Pfundt, IGL Strijdveen, and PJA Sondermeijer, The gene downstream of the gC homologue in feline herpes
virus type 1 is involved in the expression of virulence, Journal of General Virology, vol. 75, no. 11, pp. 31073116, 1994. [96]
K. Maeda, N. Yokoyama, K. Fujita, and T. Mikami, Identi- fication and characterization of the feline herpesvirus type 1
glycoprotein C gene, Virus Genes, vol. 14, tidak ada. 2, pp. 105109, 1997. [97] K. Maeda, N. Yokoyama, K. Fujita, M.
Maejima, and T. Mikami, Heparin-binding activity of feline herpesvirus type 1 glycoproteins, Virus Research, vol. 52, no. 2,
pp. 169176, 1997. [98] GR van de Walle, KW Jarosinski, and N. Osterrieder, Alphaherpesviruses and chemokines: pas de deux
not yet brought to perfection, Journal of Virology, vol. 82, no. 13, pp. 60906097, 2008. [99] GR van de Walle, ML May, W.
Sukhumavasi, J. Von Einem, and N. Osterrieder, Herpesvirus chemokine-binding glycoprotein G (gG) efficiently inhibits
neutrophil chemo- taxis in vitro and in vivo, Journal of Immunology, vol. 179, no. 6, pp. 41614169, 2007. [100] N. Thormann,
GR van de Walle, W. Azab, and N. Oster- rieder, The role of secreted glycoprotein G of equine herpes- virus type 1 and type 4
(EHV-1 and EHV-4) in immune modulation and virulence, Virus Research on line, vol. 169, no. 1, pp. 203211, 2012. [101] B.
Costes, M. Thirion, B. Dewals et al., Felid herpesvirus 1 glycoprotein G is a structural protein that mediates the binding of
chemokines on the viral envelope, Microbes and Infection, vol. 8, tidak ada. 11, pp. 26572667, 2006.
[102] TG Kimman, N. De Wind, N. Oei-Lie, JMA Pol, AJM Berns, and ALJ Gielkens, Contribution of single genes within the
unique short region of Aujeszky's disease virus (suid herpesvirus type 1) to virulence, pathogenesis and immunogenicity, Journal
of General Virology, vol. 73, no. 2, pp. 243251, 1992. [103] JM Devlin, GF Browning, CA Hartley et al., Glyco- protein G is a
virulence factor in infectious laryngotracheitis virus, Journal of General Virology, vol. 87, no. 10, pp. 2839 2847, 2006. [104]
JM Devlin, GF Browning, CA Hartley, and JR Gilk- erson, Glycoprotein G deficient infectious laryngotracheitis virus is a
candidate attenuated vaccine, Vaccine, vol. 25, tidak ada. 18, pp. 35613566, 2007. [105] JM Devlin, A. Viejo-Borbolla, GF
Browning et al., Evaluation of immunological responses to a glycoprotein G deficient candidate vaccine strain of infectious
laryngotra- cheitis virus, Vaccine, vol. 28, tidak ada. 5, pp. 13251332, 2010. [106] MC Verweij, AD Lipi nska, D. KoppersLalic et al., Structural and functional analysis of the TAP-inhibiting UL49.5 proteins of varicelloviruses, Molecular
Immunology, vol. 48, pp. 20382051, 2011. [107] H. Wei, Y. Wang, and SI Chowdhury, Bovine herpesvirus type 1 (BHV-1)
U(L) 49.5 luminal domain residues 30 to 32 are critical for MHC-I down-regulation in virus-infected cells, PLoS ONE, vol. 6,
tidak ada. 10, Article ID e25742, 2011. [108] H. Wei, J. He, DB Paulsen, and SI Chowdhury, Bovine herpesvirus type 1 (BHV1)mutant lacking UL 49.5 luminal domain residues 3032 and cytoplasmic tail residues 80- 96 induces more rapid onset of virus
neutralizing antibody response and cellular immune responses in calves than the wild-type strain Cooper, Veterinary
Immunology and Immunopathology, vol. 147, pp. 223229, 2012. [109] H. Shizuya, B. Birren, UJ Kim et al., Cloning and
stable maintenance of 300-kilobase-pair fragments of human DNA in Escherichia coli using an F-factor-based vector, Proceedings of the National Academy of Sciences of the United States of America, vol. 89, no. 18, pp. 87948797, 1992. [110] EC Lee,
D. Yu, J. Martinez de Velasco et al., A highly efficient Escherichia coli-based chromosome engineering sys- tem adapted for
recombinogenic targeting and subcloning of BAC DNA, Genomics, vol. 73, no. 1, pp. 5665, 2001. [111] S. Warming, N.
Costantino, DL Court, NA Jenkins, and NG Copeland, Simple and highly efficient BAC recombineering using galK selection,
Nucleic Acids Research, vol. 33, tidak ada. 4, p. e36, 2005. [112] D. Yu, HM Ellis, EC Lee, NA Jenkins, NG Copeland, and DL
Court, An efficient recombination system for chromosome engineering in Escherichia coli, Proceedings of the National
Academy of Sciences of the United States of America, vol. 97, no. 11, pp. 59785983, 2000. [113] CL McGowin and RB Pyles,
Mucosal treatments for her- pes simplex virus: insights on targeted immunoprophylaxis and therapy, Future Microbiology, vol.
5, tidak ada. 1, pp. 1522, 2010. [114] G. Dasgupta, AB Nesburn, SL Wechsler, and L. Ben- Mohamed, Editorial: developing an
asymptomatic mucosal herpes vaccine: the present and the future, Future Microbiol- ogy, vol. 5, tidak ada. 1, pp. 14, 2010.
[115] LS Sandmeyer, CB Keller, and D. Bienzle, Culture of feline corneal epithelial cells and infection with feline herpesvirus-1
as an investigative tool, American Journal of Veterinary Research, vol. 66, no. 2, pp. 205209, 2005.
[116] LS Sandmeyer, CB Keller, and D. Bienzle, Effects of cidofovir on cell death and replication of feline herpesvirus-1 in
cultured feline corneal epithelial cells, American Journal of Veterinary Research, vol. 66, no. 2, pp. 217222, 2005. [117] LS
Sandmeyer, CB Keller, and D. Bienzle, Effects of interferon- on cytopathic changes and titers for feline herpesvirus-1 in

primary cultures of feline corneal epithelial cells, American Journal of Veterinary Research, vol. 66, no. 2, pp. 210216, 2005.
[118] G. Leeming, ML Meli, P. Cripps et al., Tracheal organ cultures as a useful tool to study Felid herpesvirus 1 infection in
respiratory epithelium, Journal of Virological Methods, vol. 138, no. 1-2, pp. 191195, 2006. [119] AM Quintana, GA Landolt,
KM Annis, and GS Hussey, Immunological characterization of the equine air- way epithelium and of a primary equine airway
epithelial cell culture model, Veterinary Immunology and Immunopathol- ogy, vol. 140, no. 3-4, pp. 226236, 2011. [120] A.
Vandekerckhove, S. Glorieux, WVD Broeck, A. Gryspeerdt, KM van der Meulen, and HJ Nauwynck, In vitro culture of equine
respiratory mucosa explants, Veteri- nary Journal, vol. 181, no. 3, pp. 280287, 2009. [121] S. Glorieux, W. Van den Broeck,
KM van der Meulen, K. Van Reeth, HW Favoreel, and HJ Nauwynck, In vitro culture of porcine respiratory nasal mucosa
explants for studying the interaction of porcine viruses with the respira- tory tract, Journal of Virological Methods, vol. 142, no.
1-2, pp. 105112, 2007. [122] S. Glorieux, W. Van den Broeck, KM van der Meulen, K. Van Reeth, HW Favoreel, and HJ
Nauwynck, In vitro culture of porcine respiratory nasal mucosa explants for studying the interaction of porcine viruses with the
respiratory tract, Journal of Virological Methods, vol. 142, no. 1-2, pp. 105112, 2007. [123] L. Steukers, S. Glorieux, AP
Vandekerckhove, HW Favor- eel, and HJ Nauwynck, Diverse microbial interactions with the basement membrane barrier,
Trends in Microbiology, vol. 20, pp. 147155, 2012. [124] N. De Regge, HW Favoreel, K. Geenen, and HJ Nauwynck, A
homologous in vitro model to study interactions between alphaherpesviruses and trigeminal ganglion neurons, Veteri- nary
Microbiology, vol. 113, tidak ada. 3-4, pp. 251255, 2006. [125] N. De Regge, N. Van Opdenbosch, HJ Nauwynck, S. Efstathiou,
and HW Favoreel, Interferon alpha induces establishment of alphaherpesvirus latency in sensory neurons in vitro, PLoS ONE,
vol. 5, tidak ada. 9, Article ID e13076, 2010. [126] RP Wilkes and SA Kania, Use of interfering RNAs targeted against feline
herpesvirus 1 glycoprotein D for inhi- bition of feline herpesvirus 1 infection of feline kidney cells, American Journal of
Veterinary Research, vol. 70, no. 8, pp. 10181025, 2009. [127] RP Wilkes and SA Kania, Evaluation of the effects of small
interfering RNAs on in vitro replication of feline herpesvirus-1, American Journal of Veterinary Research, vol. 71, no. 6, pp.
655663, 2010.

Journal Insects
of
Hindawi Publishing Corporation http://www.hindawi.com Volume 2014
International Journal of Microbiology
Hindawi Publishing Corporation http://www.hindawi.com Volume 2014

Biotechnology Research International


Hindawi Publishing Corporation http://www.hindawi.com Volume 2014
International Genomics
Journal of
Hindawi Publishing Corporation http://www.hindawi.com
Volume 2014
International Agronomy
Journal of
Hindawi Publishing Corporation http://www.hindawi.com Volume 2014

Journal Veterinary of
Medicine
Hindawi Publishing Corporation http://www.hindawi.com Volume 2014

Scientifica
Hindawi Publishing Corporation http://www.hindawi.com Volume 2014
International Cell Biology
Journal of
Hindawi Publishing Corporation http://www.hindawi.com Volume 2014

Submit your manuscripts at http://www.hindawi.com


Veterinary Medicine International
Hindawi Publishing Corporation http://www.hindawi.com Volume 2014

Journal of Parasitology Research


Hindawi Publishing Corporation http://www.hindawi.com Volume 2014

Case Reports in Veterinary Medicine


Hindawi Publishing Corporation http://www.hindawi.com Volume 2014

Journal Animals
of
Hindawi Publishing Corporation http://www.hindawi.com Volume 2014

The Scientific World Journal


Hindawi Publishing Corporation http://www.hindawi.com Volume 2014

Zoology

International Journal of
Hindawi Publishing Corporation http://www.hindawi.com
Volume 2014
International Ecology Journal of
Hindawi Publishing Corporation http://www.hindawi.com Volume 2014

Psyche
Hindawi Publishing Corporation http://www.hindawi.com Volume 2014
International Evolutionary Journal Biology
of
Hindawi Publishing Corporation http://www.hindawi.com Volume 2014

Applied Environmental Soil Science


&
Hindawi http://www.hindawi.com
Publishing Corporation
Volume 2014
Journal Viruses
of
Hindawi Publishing Corporation http://www.hindawi.com Volume 2014

Anda mungkin juga menyukai