Anda di halaman 1dari 70

TUGAS MATA KULIAH

ILMU PENYAKIT VIRAL

“AFRICAN SWINE FEVER (ASF)”


Oleh:

PALAGAN SENOPATI SEWOYO (1709511062)


MARTINA TIODORA SITOHANG (1709511063)
NI MADE ERNAWATI (1709511065)
SAMUEL EVAN (1709511066)
IDA BAGUS KRISNA PRADNYADANA (1709511098)
I PUTU DWI KOMALA PUTRA (17095110103)
AGUSTINHO MOREIRA BELO (17095110127)
ZELIA XIMENES (17095110133)
LABORATORIUM VIROLOGI
FAKULTAS KEDOKTERAN HEWAN
UNIVERSITAS UDAYANA
TAHUN
2019
KATA PENGANTAR

Puji syukur kehadirat Tuhan Yang Maha Esa berkat limpahan rahmat dan
karunia-Nya, paper yang berjudul “AFRICAN SWINE FEVER (ASF)” dapat
diselesaikan dengan baik dan tepat waktu dalam tujuan pemenuhan tugas mata
kuliah ilmu penyakit viral. Tak lupa kami ucapkan terima kasih kepada para dosen
mata kuliah ilmu oenyakit viralyang telah membimbing dan mengajar kami:

1. Prof. drh. I NYOMAN MANTIK ASTAWA, Ph.D.


2. Prof. Dr. drh. GUSTI AYU YUNIATI KENCANA, MP.
3. Prof. Dr. drh. I Gusti Ngurah Kade Mahardika
4. Dr. drh. Ida Bagus Kade Suardana, M.Si.

Penulis sadar masih banyak kekurangan yang terdapat pada paper ini, jadi
sangat diperlukan saran dan kritik membangun dari pembaca. Besar harapan paper
ini dapat bermanfaat bagi mahasiswa Fakultas Kedokteran Hewan pada khususnya,
ataupun bidang-bidang saintek lain yang menekuni tentang parasitologi pada
umumnya. Akhir kata kami ucapkan terima kasih

Penulis

Denpasar, 2019

ii
DAFTAR ISI

KATA PENGANTAR .................................................................................... ii


DAFTAR ISI ................................................................................................... iii
DAFTAR GAMBAR ...................................................................................... iv
DAFTAR TABEL ......................................................................................... v
BAB I PENDAHULUAN ............................................................................... 1
1.1 Latar Belakang ............................................................................... 3
1.2 Rumusan Masalah .......................................................................... 1
1.3 Tujuan ............................................................................................ 1

BAB II PEMBAHASAN ................................................................................ 2


2.1 Etiologi ........................................................................................... 2
2.2 Epidemiologi .................................................................................. 3
2.2.1 Spesies Terdampak ............................................................... 4
2.2.2 Cara penularan dan penyebaran ............................................ 4
2.3 Patogenesis .................................................................................... 5
2.4 Patologi .......................................................................................... 6
2.5 Gejala Klinis................................................................................... 9
2.6 Diagnosis ........................................................................................ 11
2.7 Diagnosis Banding ......................................................................... 11
2.8 Imunitas, Pencegahan, dan Kontrol ............................................... 13
BAB III PENUTUP ........................................................................................ 15
3.1 Kesimpulan ................................................................................... 15
3.2 Saran ............................................................................................... 15
DAFTAR PUSTAKA .................................................................................... 16

iii
DAFTAR GAMBAR

Gambar 2.1 .................................................................................................... 2

Gambar 2.2 ..................................................................................................... 3

Gambar 2.3 ..................................................................................................... 5

Gambar 2.4 ..................................................................................................... 8

Gambar 2.5 ..................................................................................................... 8

Gambar 2.6 ..................................................................................................... 9

iv
DAFTAR TABEL

Tabel 2.1 ......................................................................................................... 6

Tabel 2.2 .......................................................................................................... 12

v
1

BAB I

PENDAHULUAN

1.1. Latar Belakang

African swine fever (ASF) merupakan penyakit viral pada babi yang
mengakibatkan angka mortalitas yang tinggi pada babi Hal ini menyebabkan
kehilangan ekonomi yang luarbiasa dan tidak bisa dicegah karena tidak adanya
vaksin yang efektif dan metode kontrol penyakit. Penyakit ini disebabkan oleh virus
yang pertama kali diidentifikasi di Kenya pada tahun 1920-an. Kemudian,
penyebaran terjadi begitu pesat sampai ke Eropa, dan sampai sekarang di Amerika
Selatan, dan karibia. Penyakit ini diberantas dari Eropa (kecuali Sardinia) pada
tahun 1990-an melalui kontrol drastis dan program pemberantasan. Pada paper ini
akan dibahas lebih lanjut mengenai penyakit viral ini.

1.2. Rumusan Masalah


1. Apa agen penyebab (etiologi) dari penyakit african swine fever ?
2. Bagaimana epidemiologi dari penyakit ini?
3. Bagaimana patogenesis dari penyakit ini?
4. Bagaimana gejala klinis dari penyakit ini?
5. Bagaimana diagnosis dari penyakit ini?
6. Apa diagnosis banding dari penyakit ini?
7. Bagaimana pengobatan dan pencegahan dari penyakit ini?
1.3. Tujuan Penulisan
1. Untuk mengetahui agen penyebab dari penyakit african swine fever (ASF)
2. Untuk mengetahui epidemiologi dari penyakit ini.
3. Untuk mengetahui patogenesis dari penyakit ini
4. Untuk mengetahui gejala klinis dari penyakit ini
5. Untuk mengetahui bagaimana cara mendiagnosis penyakit ini
6. Untuk mengetahui diagnosis banding dari penyakit ini
7. Untuk mengetahui bagaimana pengobatan dan pencegahannya.
2

BAB II

PEMBAHASAN

2.1. Etiologi

Gambar 2.1. Pohon filogenik yang membandingkan protein dUTPase yang


dienkode oleh African swine fever virus (AFSV) dengan virus DNA lain.
African swine fever virus (ASFV) merupakan virus DNA beramplop yang
merupakan anggota dari genus Asfivirus dan famili Asfarviridae (Asfar = African
swine fever dan virus terkait). African swine fever virus merupakan satu-satunya
arbovirus DNA yang diketahui dan ditransmisikan melalui caplak dari genus
Ornithodoros. Strain virus dibedakan berdasarkan virulensi terhadap babi, yang
mana memiliki rentang dari yang penyakit sangat letal atau mematikan hingga
penyakit yang subklinis. Strain juga dapat dibedakan berdasarkan sekuens genetik,
dan geb enkode-virus p72 (yang juga dikenal sebagai p73) yang dapat digunakan
untuk genotiping virus.
3

Gambar 2.2. (A) Pewarnaan negatif virion menunjukkan garis heksagonal dari
kapsid dengan amplop (B dan C) Pewarnaan negatif dari kapsid yang rusak yang
menunjukkan susunan kapsomer. (D) Bagian tipis dari virion menunjukkan
lapisan ganda yang mengelilingi inti. (Skala 100nm) (Sumber: J.L. Carraascosa)
Asfarvirus berdiameter kurang lebih 200 nm, dan memiliki nukleokapsid
yang dikelilingi oleh lapisan lipid internal dan sebuah kapsid ikosahedral kompleks.
Kapsid terdiri atas sebuah susunan heksagonal prisma dengan lubang sentral.
Genom terdiri atas molekul tunggal dari DNA untai ganda linier. Lebih dari 50
protein terdapat di dalam virion, termasuk beberapa enzim dan faktor yang
diperlukan di dalam proses transkripsi mRNA.

2.2. Epidemiologi

African swine fever (ASF) pertama kali ditemukan di Kenya pada tahun
1921, dan sejak itu telah menyebar secara cepat ke negara-negara afrika lain. ASF
pertama kali lolos dari afrika pada tahun 1957 dan mencapai Portugal melalui
kontaminasi produk pangan yang digunakan untuk babi. Setelah wabah ini yang
secara cepat dikontrol, ASFV masuk Portugal pada tahun 1960 dan pada waktu ini
menyebar ke seluruh semenanjung Iberian, dan persisten lebih dari 30 tahun. ASFV
menyebar secara sporadis ke negara lain di Eropa dan Amerika (Brazil, Republik
Dominika, Kuba, dan Haiti.) ASF telah diberantas pada negara-negara ini kecuali
kepulauan Italia Sardinia, yang telah terdampak sejak 1978.
4

Pada tahun 2007, ASFV memasuki Georgia kemungkinan melalui kontaminasi


pangan yang digunakan untuk babi. Dari bagian ini, ASFV menyebar secara cepat
melalui negara dan memberi dampak negara tentangga seperti Armenia,
Azerbaijan, dan Federasi Russia. Pada daerah geografis ini, ASFV mempengaruhi
babi domestik dan liar dan menyebar dari utara ke barat. Pada tahun 2012, wabah
pertama di deklarasi di Ukraina, dan 2013 pada Belarus. Dari negara ini, ASF
berlanjut menyebar sebelum mencapai perbatasan Uni Eropa pada tahun 2014, yang
mana ditemukan bangkai babi liar pada negara Lithuania dan Polandia.

2.2.1 Spesies terdampak

African swine fever menginfeksi babi domestik dan anggota dari famili
Suidae, termasuk babi hutan (Potamochoerus aethiopicus, P. porcus, dan
Sus scrofa ferus). Semua usaha untuk menginfeksi hewan lain tidak
berhasil.

2.2.2 Cara penularan dan penyebaran

Rute transmisi Lokasi dan tanggal


Limbah daging babi mentah pada Lisbon, 1957
bandara Malta. 1978
Sardinia, 1978
Georgia, 2007
Pemindahan produk pangan babi Portugal, 1960
Spanyol, 1960
Italia, 1983
Belgia, 1985
Penularan secara alami oleh babi Federasi Russia, 2007
liar yang terinfeksi Lithuania, 2014
Polandia, 2014
Estonia, 2014
Caplak yang terinfeksi Portugal, 1999
5

Penularan virus ini adalah melalui caplak Ornithodoros moubata (tick-


borne disease) sebagai vektor penyakit. Diansumsikan munculnya pertama
kali penyakit ini adalah akibat siklus alami caplak argasid dan babi hutan,
akan tetapi virus menyebar diluar lingkungan tradisional dan menginvasi
babi pada Eropa. Penyebaran virus ini juga karena akibat transportasi
produk-produk pangan babi. Produk yang terinfeksi ini kemungkinan dapat
diimpor secara ilegal untuk mendapatkan keuntungan.

2.3. Patogenesis

Gambar 2.3. Interaksi virus ASF dengan makrofag host.


Infeksi african swine fever pada babi domestik mengakibatkan leukopenia,
limfopenia, trombositopenia, dan apoptosis baik limfsoit maupun sel-sel fagositik
mononuklear. Kemampuan virus African swine fever untuk secara efisien
menyebabkan sitopatologi pada makrofag merupakan faktor penting dalam
virulensi virus. Pada makrofag yang terinfeksi, virus secara efektif menghambat
ekspresi sitokin pro-inflammatory seperti TNF (Tumor Necrosis Factor), interferon
(IFN)-1, dan interleukin (IL)- 8, akan tetapi menyebabkan ekspresi mengubah
faktor pertumbuhan (growth factor) – beta. Sebaliknya, peningkatan ekspresi TNF
juga dilaporkan pada kasus infeksi ASF secara in vitro dan in vivo. Strain dari virus
ASF dengan virulensi yang berbea memiliki kemampuan ekspresi memicu ataupun
menghambat sitokin pro-inflammatory atau IFN yang berbeda. Penghambatan
6

peradangan dimediasi paling tidak oleh bagian gen viral A238L, yang mana
mengenkode sebuah protein yang mirip dengan faktor penghambat transkripsi
seluler, nuklear faktor kB (NFkB).

Jika infeksi terjadi melalui sistema pernafasan, virus bereplikasi pertama


kali pada tonsil faringeal dan limfe nodus mukosa nasal, sebelum menyebar secara
cepat ke dalam tubuh melalui viremia primer yang mana virion dihubungkan
dengan eritrosit maupun leukosit. Infeksi umum berlanjut, dengan titer virus yang
tinggi (sampai 109 dosis infeksius per ml darah atau per gram jaringan) dan semua
cairan sekresi dan eksresi mengandung jumlah virus infeksius yang banyak.

Babi yang lolos dari infeksi akut mungkin terlihat sehat atau terlihat kronis,
namun kedua grup ini tetap terjadi infeksi yang persisten. Infeksi yang persisten ini
bahkan tidak menunjukkan gejala klinis. Durasi infeksi persisten belum diketahui,
namun kadar virus yang rendah telah di deteksi pada jaringan sampai lebih dari
setahun setelah paparan.

2.4. Patologi

Tabel 2.1. Persebaran lesi yang diamati pada bentuk-bentuk ASF

ASF ASF Akut ASF Subakut ASF Kronik


Perakut
Demam Tinggi Tinggi Moderat Irreguler
ataupun
tidak ada
Trombositopenia - Tidak ada / Sementara Tidak ada
ringan
Kulit Eritema Eritema Eritema Area
nekrotik
Limfonodus - Gastrohepatik Gambaran umum Bengkak
dan renal seperti bekuan
dengan aspek darah
kelereng
7

Limpa - Splenomegali Hiperemi parsial Terjadi


hiperemik splenomegali perbesaran
atau infark fokal dengan
warna
normal
Ginjal - Hemoragi Hemoragi -
Petekie pada petekie pada
korteks korteks, medulla,
dan pelvis,
edema perirenal
Paru-paru - Edema - Pleuritis
alveolar hebat dan
pneumonia
Kantung - Hemoragi Edema dinding -
empedu petekie
Jantung - Hemoragi Hemoragi pada Perikarditis
pada epikardium, fibrinosa
epikardium endokardium,
dan dan
endokardium hidroperikardium
Tonsil - - - Nekrotik
fokal
Alterasi - - Abortus Abortus
reproduksi
Sumber: Danrdiri, 1979, Gomez Villamandos et al., 1995a, Hervas et al., 1996,
Carrasco et al. 1997a, Sances Viszaino dan Aria, 2012.
8

Gambar 2.4. (A) Hemoragi subkutan pada telinga (B) Splenomegali


Sumber: R. Harutyan, Pusat Diagnostik dan Epizootik Universitas Illinois
Pada kasus akut yang fatal pada babi domestik, lesi berat paling menonjol
pada sistem limfoid dan vaskular. Hemoragi terjadi secara luas, limfo nodus
visceral menyerupai gumpalan darah. Terdapat ptekie yang ditandai pada semua
permukaan serosa, limfo nodus, epikardium dan endokardium, kortek ginjal, vesica
urinaria, edema dan kongesti pada kolon dan paru-paru. Limpa seringkali
mengalami perbesaran dan rapuh. Penyakit yang kronik ditandai oleh ulkus kulit,
pneumonia, perikarditis, pleuritis, dan arthritis.

Gambar 2.5. (A) Splenomegali dengan warna keungu-unguan pada seluruh


rongga abdominal (hiperemik splenomegali) (B) Beberapa limfo nodus yang
menunjukkan derajat keparahan hemoragi. (C) Potongan limfo nodus dengan
tampilan kelereng (D) hemoragi petekia pada korteks renalis
9

Gambar 2.6. ASF subakut. Edema yang intens pada dinding kantung empedu (A)
dan di sekeliling ginjal. (B) Limpa dengan hiperemik splenomegali parsial.
Hemoraagik dan edematus gastrohepatic (D) dan ginjal (E) limfonodus (F) ginjal
dengan hemoragik intens pada korteks, medulla, dan pelvis.
2.5. Gejala Klinis

Gambaran klinis ASF dapat berbeda-beda, bergantung pada virulensi isolat


virus, rute dan dosis infeksi dan karakteristik host. Tampilan klinis dari ASF pada
peternakan biasa dengan dosis kecil infeksi, tidak akan menyebabkan angka
motalitas yang tinggi ataupun gejala klinis, kecuali demam dan kematian akibat
perdarahan pada limfo nodus. Biasanya, beberapa hari kemudian, karena
peningkatan sirkulasi virus, ledakan infeksi dapat diamati dengan mortalitas yang
meningkat dan ciri-ciri lesi ataupun gejala klinis.

Perakut

Bentuk ini dipicu oleh strain ASFV yang sangat virulen yang ditandai
demam yang sangat tinggi (41-42 derajat celsius), kehilangan nafsu makan, tidak
10

aktif, hipernoea, dan hiperemi kutaneus. Hewan biasanya mati secara mendadak
setelah 1-4 setelah menunjukkan gejala klinis dan tidak ada lesi terdapat pada organ.

Akut

Gejala akut atau hiperakut dari African swine fever pada babi yang rentan
ditandai dengan hemoragi yang hebat dengan mortalitas tinggi. Setelah masa
inkubasi 5 – 15 hari, babi mengalami demam (40.5-42 derajat C) yang persisten
selama sekitar 4 hari. Dimulai 1-2 hari setelah demam, terjadi tidak nafsu makan,
diare, inkoordinasi, dan lesu. Babi akan mati pada stadium ini tanpa gejala lain.
Pada beberapa babi terjadi dispnea, muntah-muntah, leleran (discharge) nasal dan
konjungtival, sianosis pada telinga dan moncong, dan hemoragi pada hidung dan
anus. Babi bunting biasanya terjadi abortus. Angka mortalitas seringkali 100%,
dengan babi domestik mati setelah 1-3 hari setelah demam.

Subakut

Bentuk penyakit ini disebabkan oleh isolat virus dengan virulensi moderat
/ sedang, dan memberikan dampak pada hewan yang menunjukkan gejala klinis
yang mirip dengan bentuk akut. Babi yang terdampak biasanya mati setelah 7 – 20
hari dan tingkat mortalitas memiliki rentang 30 sampai 70%. Babi yang berhasil
selamat biasanya akan pulih setelah 3 sampai 4 minggu. Babi yang telah pulih masih
dapat mengeksresikan virus sampai 6 minggu post infeksi. Kematian hewan dapat
terjadi dalam dua stadium berbeda: trombositopenia yang intens / fase leukopenia
atau pada fase pemulihan ketika hemoragi muncul akibat eritodiapedesis oleh
vasodilatasi, khususnya pada hewan muda. Babi yang mengalami ASF sub akut
akan mengalami demam moderat sampai tinggi, ascites, hidroperikardium dan
edema pada dinding kantung empedu. Limpa biasanya menunjukkan hiperemi
parsial splenomegali.

Kronis

Bentuk kronis dari ASF diakibatkan oleh infeksi virus dengan virulensi
rendah, yang telah diobservasi pada Spanyol dan Portugal. Bentuk ini ditandai
dengan lesi nekrotik pada kulit dan arthritis. Maka dari itu, bentuk ini belum pernah
11

dideskripsikan pada negara-negara dengan ASFV jangka panjang (seperti Afrika


dan Sardinia). Telah dihipotesiskan bentuk kronis mungkin berasal dari evolusi
alami isolat ASFV pada studi vaksin yang dilakukan di semenjanjung Iberian pada
tahun 1960-an.

2.6. Diagnosis

Gejala klinis dari African swine fever mirip dengan beberapa penyakit,
termasuk bakterial septikemia seperti eriseplas, salmonellosis akut, akan tetapi
masalah utama dalam mendiagnosis adalah membedakan dengan swine fever klasik
(hog cholera). Setiap penyakit demam pada babi yang berhubungan dengan
hemoragi yang menyebar (hemoragik diatesis) dan angka kematian yang tinggi
harus meningkatkan kecurigaan terhadap African swine fever. Diagnosis infeksi
kronis bermasalah karena gejala klinis dan lesi pada babi yang terdampak berbeda-
beda dan sangat bervariasi. Konfirmasi atau peneguhan laboratorium sangat
penting, dari sampel darah, limfa, ginjal, limfo nodus viseral, dan tonsil harus
dikumpulkan untuk isolasi virus, deteksi antigen dengan PCR (Polymerase Chain
Reaction) untuk mendeteksi gen p72. Isolasi virus dilakukan di sumsum tulang babi
atau kultur leukosit darah perifer. Setelah isolasi awal, virus dapat diadaptasi untuk
tumbuh di berbagai lapisan sel, seperti sel Vero. Deteksi antigen didapatkan dengan
pewarnaan immunofluorescence pada jaringan.

2.7 Diagnosis Banding

Ketika terjadi stadium awal dari ASF, terutama ketika jumlah yang sedikit
hewan terdampak, diagnosis belum pasti. Keberadaan lesi non-spesifik dan
presentasi kematian yang kecil dapat dengan mudah dibingungkan oleh penyakit-
penyakit hemoragik babi. Karena itu, diagnosis banding berdasarkan lesi patologi
anatomi penyakit babi berikut: classical swine fever (CSF) / hog cholera, high-
patogenic procine reproductive and respiratory syndrome (HP-PRRS), eriseplas
babi, salmonelosis septikemia, dan procine dermatitis nephropathy syndrome
(PDNS). Karakteristik spesifik telah dirangkum pada tabel berikut.
12

Tabel 2.2. Diagnosis Banding

ASF Akut ASF CSF HP- Erisepla Salmonel DNS


Subakut PRRS s babi osis
Septikem
ia
Kulit Eritema Eritema Eritema Sianosi Lesi Sianosis Makula
s diamond dan
Papula
Limfo Aspek Hemora Multiple Bengka Aspek Bengkak -
nodus “marbled” gi infark k “marble
pada d”
tepi
Limpa Hiperemik Hiperem Henorag Hemora Hiperem Splenom Glomeru
splenomeg i Parsial i petekie gi ik egali onephrit
ali dengan ptekie splenom is
fokal egali
infark
Ginjal Hemoragi Hemora Hemora - Hemora Hemorag -
petekie gi gi gi i ptekie
Petekie petekie petekie
Kantung Hemoragi Edema Hemora Bengka - - -
empedu petekie dinding gi ptekie k
dengan
hemora
gi
Tonsil - - Area - - - -
nekrotik
Intestin Hemoragi Petekie Button - - Enterokol -
petekie ulcers itis
nekrotik
13

Trombo Tidak ada / Sementa Intens Atrofi - - -


sitopeni Ringan ra intersiti
a al
pneumo
nia
Lesi lain - - Gejala Arthirits Gejala -
Saraf dan saraf,
Kongeni endokar Kongesti
tal ditis mukosa
Malform vegetatif gastric,
asi nekrosis
fokal
pada liver
dan
bronchop
neumonia

2.8. Imunitas, Pencegahan dan kontrol

Baik komponen humoral dan seluler (termasuk limfosit spesifik CD8+)


berkontribusi dalam respon imun protektif terhadap virus African swine fever.
Respon antibodi terhadap virus ASF telah terbukti melindungi babi dari tantangan
mematikan. Namun, penawar antibodi untuk protein virion p30, p54, dan p72 tidak
cukup memberikan perlindungan yang diperantarai oleh antibodi.

Pencegahan dan pengendalian African swine fever dapat diperumit oleh


beberapa faktor, termasuk kurangnya vaksin yang efektif, penularan virus dalam
daging segar dan beberapa produk daging babi yang disembuhkan, adanya infeksi
persisten pada beberapa babi, kebingungan diagnostik dengan agen yang
menyebabkan sindrom penyakit serupa seperti hog cholera dan peranan caplak
lunak di berbagai belahan dunia. Keberadaan virus dalam caplak dan babi hutan di
14

banyak negara Afrika sub-sahara menjadikan sulit, bukan tidak mungkin untuk
memutus siklus penyebaran virus.

Di belahan dunia lain, pada negara yang bebas African swine fever, mereka
memanajemen status bebas virus dengan melarang impor babi hidup dan produk
babi dari negara-negara terinfeksi, dan dengan memonitor penghancuran sisa-sisa
makanan dari kapal maupun pesawat yang terlibat dalam rute internasional. Vaksin
belum tersedia untuk mencegah infeksi ASF. Konsekuensinya, pencegahan
berdasarkan penghindaran dari penyakit itu sendiri. Rute-rute penyakit sebisanya
harus ditutup / di blok untuk menurunkan resiko.
15

BAB III

PENUTUP

3.1 Kesimpulan

African swine fever virus (ASFV) merupakan virus DNA beramplop yang
merupakan anggota dari genus Asfivirus dan famili Asfarviridae. Penularannya
dapat melalui caplak dari genus Ornithodoros. Strain virus dibedakan berdasarkan
virulensi terhadap babi. Virus ini pertama kali di Kenya pada tahun 1921, setelah
itu menyebar secara cepat ke negara-negara lain. Gambaran patologis dari ASF
berbeda, bergantung dari bentuk ASF, yakni perakut, akut, subakut, dan kronis.
Diagnosis dari penyakit ini bisa dengan deteksi antigen menggunakan PCR untuk
mendeteksi gen. p72. Diagnosis banding dari penyakit ini adalah CSF, HP-PRRS,
Eriseplas babi, salmonellosis septikemia, dan PDDNS. Vaksin belum tersedia
sehingga diperlukan penghindaran terhadap ASF dengan penutupan / memblok
rute-rute penularan.

3.2 Saran
16

DAFTAR PUSTAKA

MachLachlan, N.J & Edward J.D. (2010). Fenner’s Veterinary Virology. 4th Ed.
USA: Elsevier Inc.
United States Departement of Agriculture (2019). Disease Response Strategy
African Swine Fever. USA: U.S. Departement of Agriculture
Alcrudo D.B. et al. (2017). African Swine Fever: Detection and Diagnosis – A
Manual for Veterinarians. USA: Food and Agriculture Organization of the
United Nations.
Galindo I. & Covadonga A. (2017). African Swine Fever Virus: A Review. Viruses.
Vol. 9: 130
Vizcaino J.M.S et al. (2015). An Update on the Epidemiology and Pathology of
African Swine Fever. J. Comp. Path. Vol. 152:9-21
Blome S. et al. (2013). Pathogenesis of African swine fever in domestic pigs and
European wild boar. Virus Research. Vol. 122:130
Howey E.B. et al. (2013). Pathogenesis of highly virulent African swine fever virus
in domestic pigs exposed via intraoropharyngeal, intranasopharyngeal, and
intramuscular inoculation, and by direct contact with infected pigs. Virus
Research. Vol 328:339
J. Comp. Path. 2015, Vol. 152, 9e21 Available online at www.sciencedirect.com

ScienceDirect

www.elsevier.com/locate/jcpa
INFECTIOUS DISEASE: REVIEW ARTICLE

An Update on the Epidemiology and Pathology


of African Swine Fever
anchez-Vizcaıno*, L. Mur*, J. C. Gomez-Villamandos†
J. M. S
and L. Carrasco†
* Centro VISAVET and Animal Health Department, Universidad Complutense de Madrid, Av. Puerta de Hierro SN 28040,
Madrid and † Department of Comparative Pathology, Universidad de Cordoba, Cordoba, Spain

Summary
African swine fever (ASF) is one of the most important infectious diseases of swine and has major negative con-
sequences for affected countries. ASF is present in many sub-Saharan countries, Sardinia and several countries
of eastern and central Europe, where its continuous spread has the swine industry on heightened alert. ASF is a
complex disease for which no vaccine or treatment is available, so its control is based on early detection and
rapid control of spread. For a robust and reliable early detection programme it is essential to be able to recog-
nize the clinical signs and pathological changes of ASF, keeping in mind that in most cases the first introduc-
tions don’t show high mortality nor characteristic clinical signs or lesions, but fever and some hemorrhagic
lymph nodes. Knowledge of the main characteristics of this infection, including its current distribution and
routes of transmission, is also essential for preventing and controlling ASF. This review addresses each of these
topics and aims to update knowledge of the disease in order to improve early detection of ASF in the field and
allow implementation of public health programmes.
Ó 2014 Elsevier Ltd. All rights reserved.

Keywords: African swine fever; clinical signs; differential diagnosis; lesions

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
African Swine Fever Epidemiology and Main Routes of Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
Clinical Presentation and Lesions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Peracute African Swine Fever . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Acute African Swine Fever . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Subacute African Swine Fever . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
Chronic African Swine Fever . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
Differential Diagnosis between African Swine Fever and Other Haemorrhagic Diseases . . . . . . . . . . . . . . . . . . . . . . . 14
Classical Swine Fever . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
Porcine Reproductive and Respiratory Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
Swine Erysipelas . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
Septicaemic Salmonellosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
Porcine Dermatitis and Nephropathy Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
Laboratory Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
Early Detection, Prevention and Control . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18

Correspondence to: J. M. Sanchez-Vizcaıno (e-mail: jmvizcaino@ucm.es).

0021-9975/$ - see front matter Ó 2014 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.jcpa.2014.09.003
10 anchez-Vizcaıno et al.
J.M. S

Introduction Despite efforts made over past decades, there is no


vaccine available for preventing and controlling
African swine fever (ASF) is one of the most impor-
ASFV infection. Several strategies have been studied;
tant infectious diseases of swine and is present in
however, the lack of neutralizing antibodies, genetic
many African countries, some eastern and central Eu-
variability and presence of some gaps in knowledge
ropean countries and Sardinia. ASF must be notified
about ASF pathogenesis and immune modulation
to the World Organization of Animal Health (OIE)
make discovery-based approaches difficult.
and its presence leads to immediate restrictions on
the pig and pork trade.
ASF is caused by infection with a complex DNA vi- African Swine Fever Epidemiology and Main
rus, the ASF virus (ASFV), which is the only member Routes of Introduction
of the family Asfarviridae (Dixon et al., 2005). ASFV
ASF was first described in Kenya in 1921
is a large enveloped virus of approximately 200 nm in
(Montgomery, 1921) and since then it has spread
diameter that contains double-stranded DNA of
rapidly to other African countries. ASF first escaped
170e193 kilobase pairs (Dixon et al., 2013). ASFV
from Africa in 1957 to reach Portugal via contami-
is composed of more than 50 structural proteins and
nated waste containing infected pig products that
produces more than 150 proteins in infected macro-
were used to feed pigs. After this incursion, which
phages (Salas and Andres, 2013), many of which
was rapidly controlled, ASFV re-entered Portugal
are highly immunogenic. Hence, infection causes a
in 1960 and this time it spread to the whole Iberian
strong humoral immune response that persists for
Peninsula, where it persisted for more than 30 years
long periods of time. Nevertheless, the antibodies pro-
(Arias and Sanchez-Vizcaıno, 2002). During this
duced are not able to neutralize ASF infection effec-
period (1960e1995), ASFV spread sporadically to
tively (Neilan et al., 2004) and serotyping is not
other countries in Europe and America (i.e. Brazil,
possible. Consequently, classification is based on gen-
Dominican Republic, Cuba and Haiti). ASF has
otyping procedures through the analysis of some
been eradicated from all of these countries except
genome regions, such as the C-terminal region of
from the Italian island of Sardinia, which has been
the gene encoding vp72 (Bastos et al., 2003). Based
affected since 1978 (Sanchez-Vizcaıno and Arias,
on the differences observed in this region, circulating
2012; Costard et al., 2013a, b).
isolates of ASFV have been classified into 22 geno-
ASF has also continued to spread on the African
types (Boshoff et al., 2007).
continent and reached West African countries and
ASFV replicates in mononuclear phagocytic cells
some islands that were previously free of the disease.
of both domestic and wild swine. The virus infects
Some authors have stated that this spread started in
monocytes and macrophages (Malmquist and Hay,
1994 due to a combination of factors including: (1)
1960), but infection of T or B lymphocytes has never
increasing pig production on the continent, (2) pres-
been observed (Minguez et al., 1988). The virus also
ence of non-symptomatic pigs that could act as reser-
replicates in endothelial cells (Carrasco et al.,
voirs to spread the disease without being noticed and
1996a), hepatocytes, renal tubular epithelial cells
(3) globalization. These factors, together with the eco-
(G omez-Villamandos et al., 1995a) and neutrophils.
nomic crisis of the present century, could be the origin
After initial replication in these primary sites, ASFV
of the spread of ASFV to Eastern Europe (Penrith and
spreads through the blood or the lymphatic system,
Vosloo, 2009; Sanchez-Vizcaıno et al., 2013).
where it persists for long periods of time in the absence
In 2007, ASFV entered Georgia through the port of
of neutralizing antibodies and moves towards second-
Poti (Beltran-Alcrudo et al., 2008), potentially via
ary sites of replication.
contaminated food used to feed pigs. From this region,
ASFV also replicates in soft ticks of the genus
ASFV spread rapidly through the country and
Ornithodoros, which act as virus reservoirs. These ticks
affected neighbouring countries including Armenia,
are involved in the epidemiological cycle of ASF in
Azerbaijan and the Russian Federation. In this
eastern and southern Africa (Ornithodoros moubata)
geographical region, ASFV affects domestic and
and have also been observed during infection on the
wild boar and has spread to the north and west. In
Iberian Peninsula (Ornithodoros erraticus). Other Orni-
2012, the first outbreaks were declared in Ukraine,
thodoros spp. have been demonstrated to be susceptible
followed by Belarus in 2013 (Sanchez-Vizcaıno
to ASF infection (European Food Safety Authority,
et al., 2013). From these countries, ASF continued
2010). ASFV strain Georgia 2007/1 can also replicate
to spread before reaching the European Union
in O. erraticus ticks (Diaz et al., 2012), but the potential
(EU) borders in 2014, when several dead wild boars
role of these ticks in virus transmission is still unknown
were found in Lithuania and Poland. Since then,
in other European regions.
several cases have been reported in Estonia and
Epidemiology and Pathology of African Swine Fever 11

Latvia and repeatedly in Lithuania and Poland Clinical Presentation and Lesions
(OIE, 2014).
ASF can have different clinical presentations and
At this time, the critical situation of ASF in East
pathological lesions, depending on the virulence of
and Central Europe poses a serious risk to other EU
the virus isolate, the route and dose of infection and
countries, which could suffer the severe consequences
the host characteristics. The clinical presentation of
of trade restrictions, as has already happened with
ASF in a naive farm with a low dose of virus infection,
Russian Federation bans. However, it is important
will not cause high mortality nor characteristic clin-
to remember that the main source of ASFV is pres-
ical signs, except fever and deaths with some hemor-
ently Africa and from there, it could spread to other
rhagic lymph nodes. Usually, few days later, due to
disease-free countries/regions and could potentially
the increased viral circulation, more explosive infec-
spread and cause high-impact consequences, as have
tions could be observed with more mortality and
occurred previously in Europe.
characteristic clinical signs and lesions. Therefore,
The main routes for the spread of ASF involve the
every dead animal that present fever in a high risk
transport of infected animals and, more frequently,
area, should be tested for ASF.
the transport of infected products (Table 1). These in-
ASFV strains are usually classified as highly viru-
fected products could be imported illegally to make
lent, moderately virulent and low virulent (Pan and
profit or could involve catering waste containing in-
Hess, 1984). Highly virulent strains are usually respon-
fected pig products. Indeed, this was the means by
sible for the peracute (pigs dead 1e4 days post infec-
which ASF was introduced into many disease-free
tion [dpi]) and acute forms (animals dead 3e8 dpi)
countries in the 1970s (Sanchez-Vizcaıno and Arias,
of the disease, while moderately virulent strains are
2012) and the cause of the last introduction in 2007
involved in the acute (pigs dead 11e15 dpi) and sub-
(Beltran-Alcrudo et al., 2008). Other sources of infec-
acute (animals die after 20 dpi) forms. In clinical
tion are related to contaminated fomites and trans-
terms, acute ASF develops over a 7-day period,
port, including livestock vehicles or infected
compared with 10e20 days for the subacute form of
material. Natural ranging of wild boar is one of the
the disease. Chronic ASF has been associated with
most serious concerns for the EU due to the proximity
infection by moderate-to-low virulence isolates
of affected territories, as demonstrated by the recent
(Mebus and Dardiri, 1979; McVicar, 1984), which
cases detected at EU borders and risk assessment an-
were only described in Spain, Portugal and the
alyses (De la Torre et al., in press). However, this route
Dominican Republic when ASF infection in these
is not likely to cause distant outbreaks in disease-free
areas was endemic. The main lesions observed in the
territories such as Asia without prior suspicion or
different forms of ASF are summarized in Table 2.
detection. Other factors, such as infected ticks or
airborne transmission, are also important, but only
at a local level and should be ruled out as potential Peracute African Swine Fever
sources for ASF infection in distant disease-free terri- This form is induced by highly virulent ASFV strains
tories. and is characterized by high fever (body temperature
Table 1
41e42 C), loss of appetite, inactivity, hyperpnoea
Main sources of ASF infection into free areas of Europe and cutaneous hyperaemia. Animals usually die sud-
denly 1e4 days after the onset of clinical signs and no
Transmission route Location and date of lesions are evident in organs.
occurrence

Raw pork waste at Lisbon, 1957 Acute African Swine Fever


airport/port Malta, 1978
Sardinia, 1978 This is the most usual form of the disease and is
Georgia, 2007 induced by highly or moderately virulent virus
Movement of pork or pig Portugal, 1960 strains. Animals with acute ASF display fever (body
product Spain, 1960 temperature 40e42 C) and a tendency to crowd
Italy, 1983 together; they also exhibit loss of appetite, inactivity,
Belgium, 1985 apathy and early leucopenia, induced by lymphope-
Natural ranging of Russian Federation, 2007 nia and changes in monocyte numbers (Colgrove
infected wild boar Lithuania, 2014 et al., 1969; Pan and Hess, 1984; Gomez-
Poland, 2014 Villamandos et al., 1995a; Carrasco et al., 1996b;
Estonia, 2014 Sanchez-Vizcaıno and Arias, 2012). Severe
Infected ticks Portugal, 1999 pulmonary oedema, giving rise to respiratory
changes, is a characteristic finding in pigs infected
12 anchez-Vizcaıno et al.
J.M. S

Table 2
Main lesions observed in the different forms of ASF

Peracute ASF Acute ASF Subacute ASF Chronic ASF

Fever High High Moderate Irregular or absent


Thrombocytopenia Absent Absent or slight (late) Transient Absent
Skin Erythema Erythema Erythema Necrotic areas
Lymph nodes e Gastrohepatic and renal with The majority of lymph nodes Swollen
marbled aspect resemble a blood clot
Spleen e Hyperaemic splenomegaly Partial hyperaemic splenomegaly Enlarged with normal
or focal infarct colour
Kidney e Petechial haemorrhages, mainly Petechial haemorrhages in cortex, e
in cortex medulla and pelvis; perirenal oedema
Lung e Severe alveolar oedema e Pleuritis and pneumonia
Gall bladder e Petechial haemorrhages Wall oedema e
Heart e Haemorrhages in epicardium and Haemorrhages in epicardium and Fibrinous pericarditis
endocardium endocardium; hydropericardium
Tonsils e e e Necrotic foci
Reproductive alteration e e Abortion Abortion

with highly virulent strains of ASF. Affected animals Dardiri, 1979; Gomez-Villamandos et al., 1995a;
die in shock, usually 1 week after fever begins and Hervas et al., 1996; Carrasco et al., 1997a, b; S
anchez-
foam is generally observed around the mouth and Vizcaıno and Arias, 2012).
nose (Sierra et al., 1990; Carrasco et al., 1996a, 2002).
Affected pigs show erythema, which particularly
Subacute African Swine Fever
affects the skin of the ears, tail, distal extremities,
chest, abdomen and perianal area. Cyanosis may This form of the disease is caused by moderately
also be observed 1e2 days prior to death on the skin virulent isolates and affected animals show similar clin-
of the ears, abdomen and perianal areas. Small foci ical signs to those animals with the acute form of the dis-
of cutaneous necrosis (more characteristic of infec- ease, although clinical signs tend to be less marked.
tions with strains of moderate virulence) and subcu- However, the vascular changes observed in subacute
taneous haematomas also occur. Other clinical signs forms of ASF, mainly haemorrhage and oedema, are
include mucoid nasal discharge, epistaxis, vomiting, more intense than those reported in the acute form of
abdominal pain, constipation or diarrhoea, which is the disease (Gomez-Villamandos et al., 1995a, 2013).
initially mucoid, but may later become bloody (mel- Haemorrhages related to the development of intense,
aena) (Moulton and Coggins, 1968; Mebus and although transient, thrombocytopenia are observed
Dardiri, 1979; Gomez-Villamandos et al., 2013). in the early and mid stages of the disease (Villeda
Abortion (caused by fever) may occur in pregnant et al., 1993a, b; Gomez-Villamandos et al., 1998).
sows; in some cases, abortion may be the first sign of Abortion is usually the first clinical sign of these
an outbreak. Almost 90e100% of pigs with these forms. Affected pigs usually die within 7e20 days and
signs will die within 7 days. the mortality rate can range from 30 to 70%.
Together with erythema and cyanosis of the skin, the Survivors usually recover within 3e4 weeks.
animals that develop acute ASF present with charac- Recovered pigs can still excrete the virus up to 6
teristic hyperaemic splenomegaly in which the spleen weeks after infection. Death of animals may occur at
can be up to six times larger than normal. An affected two different stages: in the intense thrombocytopenia/
spleen may have rounded edges, be friable in consis- leucopenia phase or in the recovery phase when
tency and purpleeblack in colour and may occupy haemorrhages appear due to erythrodiapedesis by
the entire abdominal cavity from one side to the other vasodilation, especially in young animals (G omez-
(Fig. 1A). Lymph nodes, mainly the gastrohepatic and Villamandos et al., 1998, 2013).
renal nodes, display haemorrhages in the medulla Pigs suffering from the subacute form of ASF display
(Fig. 1B), which is why sections of affected lymph nodes a moderate to high fever, ascites, hydropericardium
sometimes have a marbled appearance (Fig. 1C). Kid- and characteristic oedema of the wall of the gall-
neys usually show petechial haemorrhages in the cortex bladder and bile duct (Fig. 2A), as well as in the sur-
(Fig. 1D) and renal pelvis. Other lesions that pigs usu- rounding area of kidneys (perirenal oedema)
ally show with acute forms of the disease are petechial (Fig. 2B). The spleen usually shows initial partial hy-
haemorrhages in the mucosa of the urinary bladder, peraemic splenomegaly (Fig. 2C) that progressively re-
epicardium, endocardium and pleura (Mebus and verses, leaving some focal damage, which eventually
Epidemiology and Pathology of African Swine Fever 13

Fig. 1. Acute ASF. (A) An enlarged, purpleeblack spleen crosses the entire abdominal cavity (hyperaemic splenomegaly). (B) Several
lymph nodes display degrees of haemorrhage, most intensely in the medulla. (C) Cut sections of lymph nodes with marbled appear-
ance. (D) Petechial haemorrhages in the renal cortex.

disappears, or focal infarction with different stages of through the natural evolution of the ASFV isolates em-
organization. Lymph nodes, mainly the gastrohepatic ployed in the vaccination studies performed on the Ibe-
(Fig. 2D) and renal nodes (Fig. 2E), as well as the sub- rian Peninsula in the 1960s. To confirm this hypothesis,
mandibular, retropharyngeal, mediastinal, mesenteric several molecular studies to elucidate the similarity be-
and inguinal nodes, are haemorrhagic, oedematous tween chronic forms and vaccine isolates are underway
and friable, which is why they often look like dark red (Sanchez-Vizcaıno, unpublished observations).
haematomas (S anchez-Vizcaıno and Arias, 2012; The chronic form of the disease, which has no spe-
Gomez-Villamandos et al., 2013). Renal cific clinical signs, was mainly detected during sero-
haemorrhages (Fig. 2F) are more intense (petechiae logical screening to eradicate the disease in Spain, in
and ecchymoses) and more extensive (cortex, medulla carrier animals, of which around 10% can spread
and pelvis) than in acute forms, but are not linked to the virus for long periods of time, which likely plays
the endothelial lesions reported in acute ASF a key role in the persistence of the disease (Arias
(Gomez-Villamandos et al., 1995a; Hervas et al., 1996). and Sanchez-Vizcaıno, 2002). These animals usually
have necrotic lesions of the skin and arthritis
Chronic African Swine Fever (Sanchez-Botija, 1982), delayed growth, emaciation,
The chronic form of ASF is caused by infection by low lameness, respiratory signs, abortion and low mortal-
virulence strains, observed in Spain and Portugal, as ity (Arias et al., 1986). Unlike other forms of ASF,
well as the Dominican Republic. This form is charac- chronic forms are characterized by the absence of
terized by necrotic lesions of the skin and by arthritis vascular lesions and by the presence of lesions in
(Sanchez-Botija, 1982). However, this form of the dis- which bacteria are involved, such as fibrinous pleuri-
ease has never been described in those countries where tis and/or pericarditis, pleural adhesions, necrotic
ASFV has been present for long periods of time (e.g. pneumonia, fibrinous arthritis/periarthritis and
Africa and Sardinia). Therefore, it has been hypothe- necrotic skin lesions, as well as necrotic areas on the
sized that the chronic form may have originated tonsils and the tongue (Moulton and Coggins, 1968;
14 anchez-Vizcaıno et al.
J.M. S

Fig. 2. Subacute ASF. Intense oedema of the wall of the gall bladder (A) and surrounding the kidney (B). (C) Spleen with partial hyper-
aemic splenomegaly. Haemorrhagic and oedematous gastrohepatic (D) and renal (E) lymph nodes. (F) Kidneys with intense hae-
morrhage in the cortex, medulla and pelvis.

Arias et al., 1986). This form of the infection is not syndrome (PDNS). The specific characteristics of
currently in circulation. these differential diagnoses are summarized in Table
3 and descriptions of each disease, emphasizing the
Differential Diagnosis between African similarities and differences with ASF, are presented
Swine Fever and Other Haemorrhagic below.
Diseases
Classical Swine Fever
As discussed previously, during the early stages of
ASF, particularly when low numbers of animals are CSF, also known as ‘hog cholera’, is a fatal disease
affected, diagnosis is not straightforward. The pres- caused by a small enveloped RNA virus belonging
ence of non-specific lesions and a small percentage to the family Flaviviridae, genus Pestivirus (Van
of deaths can easily be confused with other porcine Regenmortel et al., 2000). CSF takes various clinical
haemorrhagic diseases. Therefore, differential diag- forms (i.e. peracute, acute, chronic and congenital)
noses based on the gross lesions include the following depending on the virus strain involved, the environ-
swine diseases: classical swine fever (CSF), high- ment, the time of infection and the individual host
pathogenic porcine reproductive and respiratory syn- response (Trautwein, 1988).
drome (HP-PRRS), swine erysipelas, septicaemic However in the acute and chronic forms of the dis-
salmonellosis and porcine dermatitis nephropathy ease, affected pigs show similar clinical signs:
Table 3
Differential diagnosis of African swine fever on the basis of gross lesions

Epidemiology and Pathology of African Swine Fever


ASF (acute) ASF (subacute) CSF HP-PRRS Swine erysipelas Septicaemic salmonellosis DNS

Skin Erythema Erythema Erythema Cyanosis Diamond skin lesions Cyanosis Macules and papules
Lymph nodes Marbled aspect Haemorrhagic Marbled aspect Swelling or marbled Marbled aspect Swelling Marbled aspect
aspect
Spleen Hyperaemic Partial hyperaemic Multiple infarcts Scattered infarcts or Hyperaemic Splenomegaly e
splenomegaly splenomegaly or focal at the margin white spots on the surface splenomegaly
infarct
Kidney Petechial Petechial haemorrhages; Petechial Petechial Petechial Petechial Glomerulonephritis
haemorrhages perirenal oedema haemorrhages haemorrhages haemorrhages haemorrhages
Gall bladder Petechial Wall oedema Petechial e e e e
haemorrhages haemorrhages
Tonsils e e Necrotic areas Swelling or with e e e
haemorrhages
Intestine Petechial Petechial Button ulcers e e Necrotic enterocolitis e
haemorrhages haemorrhages
Thrombocytopenia Absence or Transient Intense (early) Absent Absent Absent Absent
slight (late)
Other lesions e e Nervous signs Thymic atrophy Arthritis and Nervous signs; congestion e
Congenital Interstitial pneumonia vegetative endocarditis of gastric mucosa;
malformation (chronic form) necrotic foci in the liver
(congenital form) and bronchopneumonia

African swine fever, ASF; classical swine fever, CSF; high-pathogenic porcine reproductive and respiratory syndrome, HP-PRRS; dermatitis and nephropathy syndrome, DNS; e, no change.

15
16 anchez-Vizcaıno et al.
J.M. S

anorexia, lethargy, conjunctivitis, respiratory 2014). The main lesions are found in the lungs,
signs and constipation followed by diarrhoea. These which display interstitial pneumonia (Han et al.,
clinical signs, together with the haemorrhages (pete- 2014), and in lymphoid organs, where severe thymic
chiae and ecchymoses) of the skin, kidneys, tonsils, atrophy and swelling and haemorrhage in the tonsils
gallbladder and ileocaecal junction observed in the and lymph nodes may be observed (located in the me-
acute form, could be confused easily with ASF. dulla with a cut surface showing a marbled appear-
Some of the most characteristic lesions of CSF ance), together with scattered infarcts or white spots
include the presence of multifocal infarction of the on the surface of the spleen. Petechial haemorrhages
margin of the spleen, which takes a triangular shape, may be observed in the renal cortex (Wang et al.,
and the presence of necrotic foci on the tonsils 2011). Although haemorrhages in kidneys and lymph
(Cheville and Mengeling, 1969; Trautwein, 1988; nodes are present in HP-PRRSV, the absence of hy-
Potier et al., 2006). Although these lesions are not peraemic splenomegaly and the intensity of respira-
always present together, they represent, together with tory distress, due to interstitial pneumonia, are used
the neurological signs (induced by non-purulent to make the differential diagnosis with ASF.
meningoencephalitis), the best differentiation between
CSF and the acute and subacute forms of ASF.
Swine Erysipelas
Another characteristic lesion of the subacute and
chronic forms of CSF, which allows a differential diag- Swine erysipelas (SE) manifests commonly as acute or
nosis to be made with ASF, is the presence of ‘button subacute septicaemia and chronic proliferative lesions
ulcers’ in the small (mainly ileum and ileocaecal caused by Erysipelothrix rhusiopathiae, a small gram-
valves) and large intestines (caecum and colon) positive bacillus (Sneath et al., 1986). Acute SE is
(Cheville and Mengeling, 1969; Trautwein, 1988; characterized by sudden onset, with occasional sud-
Van Oirschot, 1999; Potier et al., 2006). den death, in which animals have a high fever
Additionally, the onset of thrombocytopenia and (body temperature 40e42 C) for 5e7 days and also
haemorrhages also differs between CSF and ASF. display severe depression and leucopenia. The sub-
Haemorrhages in CSF are associated with early acute forms are less severe than the acute form and
intense thrombocytopenia, which coincides with the abortion may occur in sows that come into contact
appearance of fever and the start of viraemia with acute or subacute SE during pregnancy (Wood
(Cheville and Mengeling, 1969; Trautwein, 1988; and Henderson, 2006).
Bautista et al., 2002). However, in ASF, slight The most characteristic lesion of SE is the presence
thrombocytopenia in the late stage can be detected of rhomboid urticarial lesions (or ‘diamond skin’ le-
in acute forms or transient thrombocytopenia may sions) in the acute non-fatal form from days 2e3 of
be present in the early and mid stages of subacute infection, which appear as small square or rhomboid
disease (Villeda et al., 1993a, b). areas with light pink to dark purple colouration that
are usually raised and firm to the touch. In the acute
fatal form of SE lesions are similar to those of septicae-
Porcine Reproductive and Respiratory Syndrome
mia, with extensive dark purple discolouration over
PRRS is caused by two genotypes of the PRRS virus the ears, tail, abdomen, the posterior aspect of the
(PRRSV), a member of the family Arteriviridae thighs, and the jowls. The lungs are congested and oe-
(Nelsen et al., 1999). PRRS is characterized by respi- dematous and haemorrhages (petechial to ecchy-
ratory problems in growing and finishing pigs and by motic) may be seen in the cortex of the kidneys, the
reproductive failure in sows, with late-term abortion, heart (particularly in the epicardium and the muscu-
stillbirth and increased preweaning mortality. lature of the atria) and the serosa of the stomach. The
Recently, however, highly pathogenic strains (HP- spleen may be congested and markedly enlarged (hy-
PRRSV) have been isolated in China and south- peraemic splenomegaly) (Wood and Henderson,
east Asia and in Eastern Europe (Garcıa-Nicolas 2006), which is similar to the acute form of ASF.
et al., 2013; Gomez-Laguna et al., 2013). Infection The involvement of lymph nodes depends on the
with HP-PRRSV strains is associated with severe area that they drain; usually the peripheral nodes
clinical signs, pulmonary lesions and aberrant host show marked congestion to haemorrhage of the
immune responses (Gomez-Laguna et al., 2013). medulla and the cut surface reveals a marbled
Pigs infected with HP-PRRS strains display high appearance (Wood and Henderson, 2006).
mortality accompanied by high fever (body tempera- Despite the similarities between acute SE and ASF
ture 40e42 C) severe lethargy, anorexia, cough, se- (i.e. clinical course, presence of haemorrhages, similar
vere dyspnoea, lameness and cyanosis of the ears, spleen lesions), there are several characteristics that
limbs and perineum (Zhou et al., 2009; Han et al., differ between these diseases. Firstly, the presence of
Epidemiology and Pathology of African Swine Fever 17

diamond skin lesions is typical in acute SE, but not in Affected pigs are anorexic and depressed, with
ASF. Additionally, the peripheral lymph nodes are little or no pyrexia (Drolet et al., 1999). There
usually the most affected in acute SE, while the gas- are redepurple macules and papules on the skin
trohepatic and renal nodes are mainly affected in caused by necrotizing vasculitis, mainly affecting the
acute ASF. hindlimbs and perineal area. These occasionally coa-
lesce to form large, irregular patches and plaques.
With time, these lesions become covered by dark
Septicaemic Salmonellosis
crusts and gradually fade (usually after 2e3 weeks),
Swine salmonellosis is caused by Salmonella cholerae- sometimes leaving scars. In severe cases, the flank
suis and has been related to the development of sep- and lateral thoracic wall may also be involved. On
ticaemia or enterocolitis (Baskerville and Dow, rare occasions, multifocal lesions are observed
1973). Pigs with septicaemic salmonellosis are inap- randomly over the entire body. Pigs that die from
petent, lethargic, febrile (body temperature acute PDNS have bilaterally enlarged kidneys with
40.5e41.6 C), have a moist cough with slight expira- small red dots in the renal cortex that correspond to
tory dyspnoea, uncoordinated gait, tremor, paraly- fibrinonecrotizing glomerulonephritis. Lymph nodes
sis, convulsions and recumbency. These clinical may be enlarged and the medulla may be congested,
signs may result in death. At 3e4 dpi, animals suffer which is why the cut surface sometimes has a marbled
from diarrhoea with watery yellow faeces. In most appearance (Segales et al., 2006).
outbreaks, which are frequently associated with
stressful situations, morbidity is variable but usually
Laboratory Diagnosis
<10%, although the mortality rate is high (Griffith
et al., 2006). Considering the similarities with other swine diseases,
Pigs that die from septicaemic salmonellosis display and the potential appearance of unspecific clinical
cyanosis of the ears, feet and tail and the ventral signs, the laboratory diagnosis of ASF is essential in
abdomen. Cutaneous icterus can be severe, although order to make a correct diagnosis. There are several
is not present consistently. Petechial haemorrhages techniques that can distinguish the 22 different
can be present in the renal cortex and on the epicar- ASFV genotypes. A correct diagnosis of ASF should
dium. There is splenomegaly, with crossing of the include virological and serological detection to obtain
abdominal cavity, but the spleen has normal colour. a complete picture of disease status (Sanchez-
The gastrohepatic and mesenteric lymph nodes are Vizcaıno and Mur, 2013). If ASF is suspected, collec-
swollen. Other lesions include hepatomegaly and tion of samples of serum and blood is required in addi-
miliary foci of necrosis in the liver and congestion of tion to the main target organs, including spleen,
the gastric fundic mucosa. The lungs are diffusely con- lymph nodes, kidney, lung and bone marrow. Oral
gested, often with interlobular oedema and sometimes fluid samples can also be used to detect ASF anti-
haemorrhage. Cranioventral bronchopneumonia is bodies (Mur et al., 2013).
not uncommon. In pigs that survive the first days, a For virological detection, the most commonly used
serous to necrotic enterocolitis develops (Griffith techniques are virus isolation and haemadsorption
et al., 2006). (HAD) tests, polymerase chain reaction (PCR) tech-
The main differences between septicaemic salmo- niques and direct immunofluorescence. Virus isolation
nellosis and the acute and subacute forms of ASF in macrophages and HAD (Malmquist and Hay,
are the absence of vascular lesions in the spleen and 1960) is the gold standard for ASF diagnosis and
lymph nodes, the existence of nervous signs and the required for the first detection of the disease in
presence of necrotic enterocolitis and miliary foci of disease-free territories or regions. However, as it is a
necrosis in the liver. laborious and time-consuming process, it is normally
used only in reference laboratories. PCRs are currently
the mostly widely used tests thanks to the reliability of
Porcine Dermatitis and Nephropathy Syndrome
the technology, even with badly preserved tissues.
PDNS is a haemorrhagic syndrome associated with There are conventional (Ag€ uero et al., 2003) and
infection with porcine circovirus type 2 (Rosell et al., real-time PCR (King et al., 2003) tests, both of which
2000; Segales et al., 2006). This syndrome affects have been validated by the OIE. Another virus detec-
nursing, growing and adult pigs. Acutely affected tion test is direct immunofluorescence (DIF), which is
pigs usually die within a few days of onset of clinical designed to detect ASF antigens in smears or sections of
signs, but surviving pigs tend to recover and gain infected tissue. For this purpose, antibodies against
weight 7e10 days after the syndrome begins (Segales ASFV conjugated with fluorescein are used for visual-
et al., 1998). izing the presence of antigens.
18 anchez-Vizcaıno et al.
J.M. S

Serology has been employed widely in eradication Such information campaigns help counteract one of
and control programmes for ASF (Arias and the most important problems in the control of animal
Sanchez-Vizcaıno, 2002). As no vaccine is available, diseases: delayed detection in the field. Knowledge of
the presence of ASF antibodies implies previous infec- the clinical presentation and pathological lesions
tion. Consequently, serology is appropriate for detect- compatible with ASF is essential for effective control.
ing potential carrier animals in endemic situations However, as it was mentioned before, the presentation
(Arias and Sanchez-Vizcaıno, 2002). The enzyme of the disease in the field does not always present the
linked immunosorbent assay (ELISA) is the most typical clinical characteristics and lesions. Conse-
commonly used serological test as it can be widely em- quently, every animal in a high risk area presenting
ployed for screening purposes. Several ELISA tests high fever and dead should be analysed for ASF to
are available. Presently, the OIE uses a validated make an early detection and avoid the spread of the
in-house soluble ASF antigen (Sanchez-Vizcaıno explosive infection. Otherwise, disease can spread (as
et al., 1982) and a commercial ELISA. Other ELISAs has already occurred with other animal diseases such
based on recombinant antigens (Gallardo et al., 2009) as CSF or foot-and-mouth disease in the EU) for weeks
are also used. The immunoblotting (Pastor et al., or even months before suspected samples arrive at the
1989) and immunoperoxidase (Pan et al., 1978) tests laboratory.
are employed for confirmation purposes or to comple- The control measures for ASF are based on rapid
ment the detection of ASF antibody. field detection and diagnosis confirmation, slaughter-
ing animals in infected holdings and banning move-
ments of pigs and pig-derived products in affected
Early Detection, Prevention and Control
areas. Other factors, such as the involvement of wild
There is no vaccine available for preventing ASF boar and/or ticks in disease appearance, should be an-
infection. Consequently, prevention is based on alysed as potential sources of infection and the mea-
avoiding introduction of the disease. Several routes sures required to control them should be applied
of entry and risk assessments have been described whenever necessary.
for different EU countries (Mur et al., 2012a, b,
2014; Costard et al., 2013a; De la Torre et al., in
press). The main control measures are to monitor Acknowledgements
the entry of pigs and pig products and to forbid the
This study was funded partially by the European
use of food waste for feeding pigs. Food waste from
Research Project ASFORCE (‘Targeted research
infected areas should be incinerated and never used
effort on African swine fever’, grant agreement
to feed pigs (Sanchez-Vizcaıno and Arias, 2012).
311931). L. Mur holds a scholarship (FPU program)
Contact made between wild boar and domestic pigs
from the Spanish Ministry of Education. The authors
is also an important risk for ASF-free countries
would like to dedicate this paper to all researchers
located near infected areas. Separating domestic
who have worked or collaborated with our team for
free-ranging pig production from areas containing
the last 30 years in studying ASF; particularly Prof.
wild boars is also important, as well as increasing bio-
A. Jover and Prof. M. A. Sierra, and the importance
security by fencing pig farms to control against this
contribution of Dr. I. Minguez and Dr. C. Patta who
route of transmission. A publication on assessing the
have now both died, but their work on ASF research
risk of introduction of ASF into the EU by wild
and control will not be forgotten.
boar has described the importance of this transmission
route for ASFV (De la Torre et al., in press).
If risk assessments are performed for estimating the
References
risk of introduction of ASF, they should be used to in-
crease awareness and surveillance in the critical loca- Ag€
uero M, Fernandez J, Romero L, Mascaraque CS,
tions/times/pathways indicated by the analysis Arias M et al. (2003) Highly sensitive PCR assay for
(Mur et al., 2014). Another important control mea- routine diagnosis of African swine fever virus in clinical
sure is access to a good early detection programme samples. Journal of Clinical Microbiology, 41, 4431e4434.
Arias ML, Escribano JM, Rueda A, S anchez-Vizcaıno JM
that includes raising awareness among veterinarians
(1986) La peste porcina Africana. Medicine Veterinaire, 3,
and farmers by providing them with updated infor- 333e350.
mation on risks. Basic ASF concepts, especially Arias M, Sanchez-Vizcaıno JM (2002) African swine fever
regarding transmission routes, how to avoid intro- eradication: the Spanish model. In: Trends in Emerging
duction and clinical signs and lesions, should be ad- Viral Infections of Swine, A Morilla, K Jin,
dressed in leaflets, posters and by other information J Zimmerman, Eds., Iowa State University Press,
campaigns. Ames, pp. 133e139.
Epidemiology and Pathology of African Swine Fever 19

Baskerville A, Dow C (1973) Pathology of experimental Diaz AV, Netherton CL, Dixon LK, Wilson AJ (2012) Afri-
pneumonia in pigs produced by Salmonella choleraesuis. can swine fever virus strain Georgia 2007/1 in Ornithodoros
Journal of Comparative Pathology, 83, 207e215. erraticus ticks. Emerging Infectious Diseases, 18, 1026e1028.
Bastos AD, Penrith ML, Cruciere C, Edrich JL, Dixon LK, Chapman DAG, Netherton CL, Upton C
Hutchings G et al. (2003) Genotyping field strains of Af- (2013) African swine fever virus replication and geno-
rican swine fever virus by partial p72 gene characterisa- mics. Virus Research, 173, 3e14.
tion. Archives of Virology, 148, 693e706. Dixon LK, Escribano JM, Martins C, Rock DL, Salas ML
Bautista MJ, Ruiz-Villamor E, Salguero FJ, S anchez- et al. (2005) Asfarviridae. In: Eighth Report of the Interna-
Cordon PJ, Carrasco L et al. (2002) Early platelet aggre- tional Committee on Taxonomy of Viruses, CM Fauquet,
gation as a cause of thrombocytopenia in classical swine MA Mayo, J Maniloff, U Desselberger, LA Ball, Eds.,
fever. Veterinary Pathology, 39, 84e91. Elsevier Academic Press, London, pp. 135e143.
Beltran-Alcrudo D, Lubroth J, Depner K, De la Roque S Drolet R, Thibault S, D’Allaire S, Thomson JR, Done SH
(2008) African swine fever in the Caucasus. FAO (1999) Porcine dermatitis and nephropathy syndrome
EMPRES Watch,1e8, April 2008. (PDNS): an overview of the disease. Journal of Swine
Boshoff CI, Bastos AD, Gerber LJ, Vosloo W (2007) Ge- Health and Production, 7, 283e285.
netic characterisation of African swine fever viruses European Food Safety Authority. (2010) Scientific opinion
from outbreaks in southern Africa (1973e1999). Veteri- on the role of tick vectors in the epidemiology of
nary Microbiology, 121, 45e55. Crimean-Congo hemorrhagic fever and African swine
Carrasco L, Bautista MJ, G omez-Villamandos JC, Martın fever in Eurasia. EFSA Journal, 8, 1703.
de las Mulas J, Chac on-M et al. (1997a) Development of Gallardo C, Reis AL, Kalema-Zikusoka G, Malta J,
microscopic lesions in splenic cords of pigs infected with Soler A et al. (2009) Recombinant antigen targets for
African swine fever virus. Veterinary Research, 28, 93e99. serodiagnosis of African swine fever. Clinical and Vaccine
Carrasco L, Chacon M, de Lara F, Martın de las Mulas J, Immunology, 16, 1012e1020.
Gomez-Villamandos JC et al. (1997b) Ultrastructural Garcıa-Nicol as O, Baumann A, Vielle NJ, G omez-
changes related to lymph node haemorrhages in acute Laguna J, Quereda JJ et al. (2013) Virulence and
African swine fever. Research in Veterinary Science, 62, genotype-associated infectivity of interferon-treated
199e204. macrophages by porcine reproductive and respiratory
Carrasco L, de Lara FC, G omez-Villamandos JC, syndrome viruses. Virus Research, 179, 204e211.
Bautista MJ, Villeda CJ et al. (1996a) The pathogenic G
omez-Laguna J, Salguero FJ, Pallares FJ, Carrasco L
role of pulmonary intravascular macrophages in acute Af- (2013) Immunopathogenesis of porcine reproductive
rican swine fever. Research in Veterinary Science, 61, 193e198. and respiratory syndrome in the respiratory tract. Veter-
Carrasco L, de Lara FC, Martın de las Mulas J, G omez- inary Journal, 195, 148e153.
Villamandos JC, Perez J et al. (1996b) Apoptosis in G
omez-Villamandos JC, Bautista MJ, Carrasco L,
lymph nodes in acute African swine fever. Journal of Chac on M, De Lara F et al. (1998) Thrombocytopenia
Comparative Pathology, 115, 415e428. associated with apoptotic megakaryocytes in a viral hae-
Carrasco L, N ~ez A, Salguero FJ, Dıaz San Segundo F,
un morrhagic syndrome induced by a moderately virulent
Sanchez-Cordon PJ et al. (2002) African swine fever: strain of African swine fever virus. Journal of Comparative
expression of interleukin-1a and tumour necrosis fac- Pathology, 118, 1e13.
tor-a by pulmonary intravascular macrophages. Journal G
omez-Villamandos JC, Bautista MJ, S anchez-Cord on PJ,
of Comparative Pathology, 126, 194e201. Carrasco L (2013) Pathology of African swine fever: the
Cheville NF, Mengeling WL (1969) The pathogenesis of role of monocyte-macrophage. Virus Research, 173,
chronic hog cholera (swine fever). Histologic, immuno- 140e149.
fluorescent and electron microscopic studies. Laboratory G
omez-Villamandos JC, Herv as J, Mendez A, Carrasco L,
Investigation, 20, 261e274. Martın de las Mulas J et al. (1995a) Experimental Afri-
Colgrove GS, Haelterman ED, Coggins L (1969) Patho- can swine fever: apoptosis of lymphocytes and virus
genesis of African swine fever in young pigs. American replication in other cells. Journal of General Virology, 76,
Journal of Veterinary Research, 30, 1343e1359. 2399e2405.
Costard S, Jones BA, Martinez-Lopez B, Mur L, de la Griffith RW, Schwartz KJ, Meyerholz DK (2006) Salmo-
Torre A et al. (2013a) Introduction of African swine fe- nella. In: Disease of Swine, 9th Edit., BE Straw,
ver into the European Union through illegal importa- JJ Zimerman, S D’Allaire, DJ Taylor, Eds., Blackwell
tion of pork and pork products. PLoS One, 8, e61104. Publishing, Ames, pp. 739e754.
Costard S, Mur L, Lubroth J, S anchez-Vizcaıno JM, Han D, Hu Y, Li L, Tian H, Chen Z et al. (2014) Highly
Pfeiffer DU (2013b) Epidemiology of African swine fe- pathogenic porcine reproductive and respiratory syn-
ver virus. Virus Research, 173, 191e197. drome virus infection results in acute lung injury of the
De la Torre A, Bosch J, Iglesias I, Mu~ noz MJ, Mur L et al. infected pigs. Veterinary Microbiology, 169, 135e146.
(in press) Assessing the risk of African swine fever intro- Herv as J, G
omez-Villamandos JC, Mendez A, Carrasco L,
duction into the European Union by wild boar. Trans- Sierra MA (1996) The lesional changes and pathogen-
boundary and Emerging Disease, http://dx.doi.org/10. esis in the kidney in African swine fever. Veterinary
1111/tbed.12129. Research Communications, 20, 285e299.
20 anchez-Vizcaıno et al.
J.M. S

King DP, Reid SM, Hutchings GH, Grierson SS, Pan IC, Shimizu M, Hess W (1978) African swine fever:
Wilkinson PJ et al. (2003) Development of a TaqManÒ microplaque assay by an immunoperoxidase method.
PCR assay with internal amplification control for the American Journal of Veterinary Research, 39, 491e497.
detection of African swine fever virus. Journal of Virolog- Pastor M, Laviada M, S anchez-Vizcaıno JM, Escribano J
ical Methods, 107, 53e61. (1989) Detection of African swine fever virus antibodies
Malmquist WA, Hay D (1960) Hemadsorption and cyto- by immunoblotting assay. Canadian Journal of Veterinary
pathic effect produced by African swine fever virus in Research, 53, 105e107.
swine bone marrow and buffy coat cultures. American Penrith M-L, Vosloo W (2009) Review of African swine fe-
Journal of Veterinary Research, 21, 104e108. ver: transmission, spread and control. Journal of the South
McVicar JW (1984) Quantitative aspects of the transmis- African Veterinary Association, 80, 58e62.
sion of African swine fever. American Journal of Veterinary Potier MF, Mesplede A, Vannier P (2006) Classical swine
Research, 45, 1535e1541. fever and other pestiviruses. In: Disease of Swine, 9th
Mebus CA, Dardiri AH (1979) Additional characteristics Edit., BE Straw, JJ Zimerman, S D’Allaire,
of disease caused by the African swine fever viruses iso- DJ Taylor, Eds., Blackwell Publishing, Ames,
lated from Brazil and the Dominican Republic. Proceed- pp. 291e298.
ings of the Annual Meeting of the US Animal Health Rosell C, Segales J, Ramos-Vara JA, Folch JM, Rodrıguez-
Association, 83, 227e239. Arrioja GM et al. (2000) Identification of porcine circo-
Minguez I, Rueda A, Dominguez J, S anchez-Vizcaıno JM virus in tissues of pigs with porcine dermatitis and ne-
(1988) Double labeling immunohistological study of Af- phropathy syndrome. Veterinary Record, 146, 40e43.
rican swine fever virus-infected spleen and lymph nodes. Salas ML, Andres G (2013) African swine fever virus
Veterinary Pathology, 25, 193e198. morphogenesis. Virus Research, 173, 29e41.
Montgomery ER (1921) On a form of swine fever occurring S
anchez-Botija C (1982) African swine fever. New develop-
in British East Africa (Kenya Colony). Journal of Compar- ments. Revue Scientifique et Technique, 4, 1065e1094.
ative Pathology and Therapeutics, 34, 159e191. S
anchez-Vizcaıno JM, Arias M (2012) African swine fever.
Moulton J, Coggins L (1968) Comparison of lesions in In: Diseases of Swine, 10th Edit., JJ Zimerman,
acute and chronic African swine fever. Cornell Veteri- L Karriker, A Ramirez, K Schwartz, G Stevenson,
narian, 58, 364e387. Eds., John Wiley & Sons, Ames, pp. 396e404.
Mur L, Gallardo C, Soler A, Zimmermann J, Pelayo V et al. S
anchez-Vizcaıno JM, Mur L (2013) African swine fever
(2013) Potential use of oral fluid samples for serological diagnosis update. In: Vaccines and Diagnostics for Trans-
diagnosis of African swine fever. Veterinary Microbiology, boundary Animal Diseases, JA Roth, JA Richt,
165, 135e139. IA Morozov, Eds., Karger, Basel, pp. 159e165.
Mur L, Martınez-L opez B, Costard S, de la Torre A, S
anchez-Vizcaıno JM, Mur L, Martınez-L opez B (2013)
Jones BA et al. (2014) Modular framework to assess the African swine fever (ASF): five years around Europe.
risk of African swine fever virus entry into the European Veterinary Microbiology, 26, 45e50.
Union. BMC Veterinary Research, 10, 145. S
anchez-Vizcaıno JM, Tabares E, Salvador E, S anchez-
Mur L, Martinez-Lopez B, Martinez M, Costard S, Bojita C (1982) Semipurified structural viral protein
Wieland B et al. (2012a) Quantitative risk assessment for the detection of African swine fever antibodies by
for the introduction of African swine fever virus into the indirect ELISA technique. Current Topics in Veterinary
the European Union by legal import of live pigs. Trans- Medicine and Animal Science, 22, 214e222.
boundary and Emerging Diseases, 59, 134e144. Segales J, Allan GM, Domingo M (2006) Porcine circovi-
Mur L, Martınez-L opez B, Sanchez-Vizcaıno JM (2012b) rus diseases. In: Disease of Swine, 9th Edit., BE Straw,
Risk of African swine fever introduction into the Euro- JJ Zimerman, S D’Allaire, DJ Taylor, Eds., Blackwell
pean Union through transport-associated routes: re- Publishing, Ames, pp. 299e307.
turning trucks and waste from international ships and Segales J, Piella J, Marco E, Mateu-de-Antonio EM,
planes. BMC Veterinary Research, 8, 149. Espuna E et al. (1998) Porcine dermatitis and nephrop-
Neilan JG, Zsak L, Lu Z, Burrage TG, Kutish GF et al. athy syndrome in Spain. Veterinary Record, 142,
(2004) Neutralizing antibodies to African swine fever vi- 483e486.
rus proteins p30, p54, and p72 are not sufficient for Sierra MA, Carrasco L, G omez-Villamandos JC, Martin
antibody-mediated protection. Virology, 319, 337e342. de las Mulas J, Mendez A et al. (1990) Pulmonary intra-
Nelsen CJ, Murtaugh MP, Faaberg KS (1999) Porcine vascular macrophages in lungs of pigs inoculated with
reproductive and respiratory syndrome virus compari- African swine fever virus of differing virulence. Journal
son: divergent evolution on two continents. Journal of of Comparative Pathology, 102, 323e334.
Virology, 73, 270e280. Sneath PHA, Mair NS, Sharpe ME, Holt JG (1986)Bergey’s
OIE (2014) WAHID Database. Disease Information. Available Manual of Systematic Bacteriology. Williams & Wilkins,
at: http://web.oie.int/wahis/public.php?page¼disease_ Baltimore, pp. 1245e1249.
immediate_summary (accessed 25 June 2014 2012). Trautwein G (1988) Classical swine fever and related viral
Pan IC, Hess WR (1984) Virulence in African swine fever: diseases. 3. Pathology and pathogenesis of the diseases.
its measurement and implications. American Journal of In: Developments in Veterinary Virology, B Liess, Ed., Mar-
Veterinary Research, 45, 361e366. tinus Nijhoff Publishing, Boston, pp. 27e54.
Epidemiology and Pathology of African Swine Fever 21

Van Oirschot JT (1999) Hog cholera. In: Diseases of Swine, Wang G, Song T, Yu Y, Liu Y, Shi W et al. (2011) Immune
8th Edit., BE Straw, S D’Allaire, WL Mengeling, responses in piglets infected with highly pathogenic
DJ Taylor, Eds., Blackwell Science, Malden, porcine reproductive and respiratory syndrome virus.
pp. 159e172. Veterinary Immunology and Immunopathology, 142, 170e178.
Van Regenmortel MHV, Fauquet CM, Bishop DHL, Wood RL, Henderson LM (2006) Erysipelas. In: Diseases of
Carstens EB, Estes MK et al. (2000) Virus taxonomy, Swine, 8th Edit., BE Straw, S D’Allaire, WL Mengeling,
classification and nomenclature of viruses. Seventh Report DJ Taylor, Eds., Blackwell Science, Malden, pp. 629e638.
of the International Committee on Taxonomy of Viruses, Aca- Zhou L, Zhang J, Zeng J, Yin S, Li Y et al. (2009) The 30-
demic Press, London, pp. 867e872. amino-acid deletion in the Nsp2 of highly pathogenic
Villeda CJ, Williams SM, Wilkinson PJ, Vi~ nuela E (1993a) porcine reproductive and respiratory syndrome virus
Haemostatic abnormalities in African swine fever. A emerging in China is not related to its virulence. Journal
comparison of two virus strains of different virulence of Virology, 83, 5156e5167.
(Dominican Republic 78 and Malta 78). Archives of
Virology, 130, 71e83.
Villeda CJ, Williams SM, Wilkinson PJ, Vi~ nuela E (1993b)
Consumption coagulopathy associated with shock in
acute African swine fever. Archives of Virology, 133,
467e475. ½ Received,
Accepted, September 23rd, 2014 
September 4th, 2014
Journal of General Virology (1998), 79, 1439–1443. Printed in Great Britain
.......................................................................................................................................................................................................... SHORT COMMUNICATION

The pathogenesis of African swine fever in the resistant


bushpig
C. A. L. Oura, P. P. Powell, E. Anderson† and R. M. E. Parkhouse
Institute for Animal Health, Pirbright Laboratory, Ash Road, Pirbright, Surrey GU24 0NF, UK

eating warthog tissue or whole ticks containing virus


Bushpigs and warthogs are natural reservoir hosts (Thomson et al., 1980). The susceptibility of bushpigs to ASFV
of African swine fever virus (ASFV) in the wild, was first demonstrated by Montgomery (1921). Since then,
showing no clinical signs of disease when infected however, the one further study on bushpigs was limited to the
with the same highly virulent isolates of ASFV that isolation of ASFV from several animals (DeTray, 1963).
induce rapid, haemorrhagic death in domestic pigs. Bushpigs are thought to be natural reservoir hosts of ASFV.
In contrast to domestic pigs, infection of bushpigs They show no clinical signs of disease when infected with
with Malawi isolate results in low levels of virus virulent and haemorrhagic isolates of ASFV that kill domestic
replication and lymphocyte apoptosis within the pigs within 5–7 days of infection. The aim of this study was to
spleen, and a relatively low spread of virus to other investigate the role of the bushpig as a reservoir host of ASFV
lymphoid tissues. However, at 10 days post-in- and to compare the pathogenesis of ASF in bushpigs and
fection, a high degree of apoptosis was seen in B domestic pigs. Specifically, the levels of ASFV replication in
lymphocytes of the B cell follicles in bushpig lymph tissues of bushpigs and pigs were assessed by virus titrations ;
nodes. Virus infected cells were present amongst the pathology, cell tropism and organ distribution of ASFV
the apoptotic B cells of these follicles, suggesting were investigated by immunocytochemistry, and compared to
that indirect factors released from ASFV infected similar studies in the domestic pig which are reported in the
macrophages signal surrounding lymphocytes to accompanying paper (Oura et al., 1998 a). Such comparisons of
enter apoptosis. The susceptibility/resistance of disease immunopathology may lead to an understanding of
domestic pigs/bushpigs to ASFV may serve as a why bushpigs survive infections with ASFV whereas domestic
unique veterinary model for the recently emerging pigs die, and provide important clues for the design of future
haemorrhagic disease of man. control strategies, such as a vaccine.
All bushpig infection studies were carried out on an island
on Lake Kariba, Northern Zimbabwe, with two separate
African swine fever virus (ASFV) is a non-pathogenic DNA groups of bushpigs. The bushpigs were captured from an
virus of warthogs and bushpigs which persists in Africa via a ASFV-free area in Zimbabwe. Of the first group of four
natural cycle of transmission between the warthog (Phaco- bushpigs, around 6 months old, three were infected intra-
choerus aethiopicus), bushpig (Potamchoerus porcus) and the soft muscularly with 10% HAD (50 % haemadsorbing units per g
&!
tick (Ornithodorus moubata). Warthogs are born free from tissue or per ml blood) Malawi isolate (Haresnape et al., 1984)
infection and may be infected early in life following the bite of of ASFV and sacrificed 5, 10 and 15 days post-infection (p.i.),
an infected tick. Virus replicates in the warthog and produces and the fourth was an uninfected control. The second group of
a low degree of viraemia for a few weeks, which is sufficient to two bushpigs, also 6 months old, was infected intramuscularly
infect a proportion of ticks that feed on the viraemic young with 10% HAD Malawi isolate of ASFV and sacrificed at 5
&!
warthogs (Thomson, 1985). Domestic pigs in Africa acquire and 10 days p.i. Blood, liver, spleen, kidney, gastro-hepatic
infection from wildlife reservoirs of the virus primarily by the lymph node, mesenteric lymph node, parotid lymph node,
bite of an infected tick (Plowright, 1977). Also, under submandibular lymph node, thymus, lung and tonsil were
experimental conditions, domestic pigs have been infected by removed and rapidly frozen (liquid N ) in OCT compound
#
(Tissue Tec) or fixed in 4 % paraformaldehyde in PBS for 18 h,
Author for correspondence : P. P. Powell. then paraffin embedded. Sections (5 µm) were placed onto
Fax ­44 1483 232448. e-mail penny.powell!bbsrc.ac.uk Superfrost Plus slides (BDH). As a direct comparison, Large
White¬Landrace pigs weighing 30 kg were inoculated intra-
† Present address : Veterinary Research Laboratory, PO Box 8101, muscularly with the highly virulent, 100 % fatal and haem-
Causeway, Harare, Zimbabwe. adsorbent Malawi isolate of ASFV (10% HAD per pig). One
&!

0001-5319 # 1998 SGM BEDJ


C. A. L. Oura and others

tissue. A high degree of virus replication (87³5 infected


cells}mm#) was seen in the red pulp of the spleen (Fig. 2 B) and
in cells located both outside and within the epithelium of
crypts in the tonsil (13³1±1 infected cells}mm#) (Fig. 2 D).
Interestingly, in the bushpig ASFV replication was seen in cells
of the tonsillar crypt at 5, 10 and 15 days p.i. These infected
cells are shed into the pharynx and, if they are coughed up,
may be a mode of transmission of the virus. Indeed, direct
transmission of ASFV (Malawi isolate) from bushpig to
domestic pig has been achieved under experimental conditions
(Anderson et al., 1998).
The organ distribution of the virus was significantly
different in bushpigs compared to domestic pigs. In bushpig
Fig. 1. Comparison of virus titrations in bushpigs and domestic pigs lymphoid tissues, virus replication was seen in the spleen but
infected with the Malawi isolate of ASFV. Virus titrations (log10 50 % there was very limited escape of the virus to other lymphoid
haemadsorbing units per g tissue or per ml blood, HAD50) in blood, organs. This was in marked contrast to the domestic pig, where
spleen and tonsil removed from bushpigs and domestic pigs infected with
Malawi isolate of ASFV. initial high levels of virus replication in the spleen were rapidly
followed by escape of the virus and high levels of virus
replication in other lymphoid organs (Oura et al., 1998 a). The
pig was used as the uninfected control, one pig was sacrificed higher degree of infection in the lymphoid tissue of domestic
at 2 days p.i., and pigs were sacrificed in pairs at 3, 4, 5, 6 and pigs was associated with severe irreversible pathological
7 days p.i. damage and apoptosis of lymphocytes which were not seen in
Virus levels in bushpig and domestic pig tissues and blood the bushpig lymphoid tissues. The number of ASFV infected
were titrated by the haemadsorption assay described pre- cells correlates with the degree of lymphocyte apoptosis,
viously by Wilkinson et al. (1977). Titres of between 5 and 7 indicating that ASFV infected macrophages, possibly through
log HAD were measured in bushpig tissues (blood, spleen, the release of cytokines or other apoptotic mediators, may be
"! &!
liver, tonsil, lymph nodes, kidney and lung) at 5, 10 and 15 responsible for the extensive lymphocyte apoptosis. Therefore,
days p.i. When compared with tissue virus titres in Malawi the reason why bushpigs survive infection with virulent
infected domestic pig tissues, the bushpig titres were around 2 isolates of ASFV may be because the virus is not so extensively
log HAD lower (Fig. 1). These results indicate that disseminated throughout the mononuclear phagocytic system
"! &!
considerably less virus replication occurred in ASFV infected as clearly occurs in the domestic pig. With fewer infected
bushpig tissues compared to domestic pig tissues. Similar macrophages in the peripheral lymphoid tissue there would be
levels of blood and tissue virus titrations (³1 log HAD ) less release of cytokines or vasoactive substances such as
"! &!
were measured in the two separate groups of infected bushpigs. reactive oxygen intermediates or nitric oxide, and thus less
Cell tropism and organ distribution of ASFV Malawi isolate destructive pathology and lymphocyte apoptosis.
were determined by immunocytochemistry on bushpig tissues There was a striking difference between bushpig lymph
taken from two bushpigs at 5 and 10 days p.i. and one bushpig nodes at 5 and 10 days p.i. The terminal deoxynucleotidyl
at 15 days p.i. Standard immunohistochemical techniques were transferase (TdT)-mediated dUTP end labelling technique
carried out as described previously (Oura et al., 1998 b). The (TUNEL) was used to label DNA strand breaks in apoptotic
levels of virus replication in tissues stained with anti-vp73 cells in tissue sections (Oura et al., 1998 a). At 10 days p.i. many
MAb 4H3 (Cobbold et al., 1996) were quantified by counting B cells in many of the follicles had pyknotic nuclei and were
the number of infected cells per mm#. The mean number of apoptotic (Fig. 3 A, B). In some B cell follicles 70–80 % of B
infected cells and standard error from 30 fields were calculated. lymphocytes were apoptotic. Also, interestingly, virus rep-
As might have been predicted from the virus titrations, results lication was detected in cells in the B cell follicles (Fig. 3 C). The
reveal that there was considerably less virus replication in most likely phenotypes of infected cells in the follicles are
bushpig tissues compared to domestic pig tissues. There was tingible body macrophages or follicular dendritic cells. In order
an initial low level of virus replication of (3±5³0±5 infected to identify the phenotype of the apoptotic cells in the follicles,
cells}mm#) at 5 days p.i. in cells in the red pulp of the spleen lymph node cryosections from bushpigs, 10 days p.i. with
(Fig. 2 A). At this time-point there was no detectable replication Malawi, were immunostained with anti-CD21 B cell specific
in the lymph nodes ; however, in the tonsil there was virus MAb C35 (Saalmuller, 1996). The vast majority of the cells in
replication (3±2³0±5 infected cells}mm#) in cells located within the follicles were B cells (Fig. 3 D). This demonstrated that the
the epithelium of tonsillar crypts (Fig. 2 C). This was in marked apoptotic cells in the follicles were B lymphocytes. The degree
contrast to the domestic pig in which, at 5 days p.i., there was of apoptosis of B lymphocytes in B cell follicles of bushpig
extensive replication of ASFV (Malawi isolate) in all lymphoid lymph nodes, at 10 days p.i., was considerably higher than the

BEEA
Pathogenesis of ASF in the bushpig

Fig. 2. Immunocytochemical localization of vp73 protein in bushpig and domestic pig tissues infected with ASFV (Malawi
isolate). Paraffin embedded sections, stained with anti-vp73 MAb 4H3, visualized with DAB (brown, arrow) and counterstained
with haematoxylin. (A) Bushpig spleen, 5 days p.i. with Malawi (¬80) ; (B) domestic pig spleen, 5 days p.i. with Malawi
(¬80) ; (C) bushpig tonsil, 5 days p.i. with Malawi (¬80) ; (D) domestic pig tonsil, 5 days p.i. with Malawi (¬160). W, White
pulp ; R, red pulp ; C, crypt.

degree of apoptosis seen in follicles of lymph nodes from TUNEL. There was considerably less apoptosis observed in
domestic pigs, 10 days p.i. with the moderately virulent Malta germinal centres of the vaccinated pig compared to the
isolate of ASFV (Oura et al., 1998 a). In order to stimulate an bushpig at 10 days p.i. (data not shown). While not conclusive,
exuberant immune response in lymph node germinal centres, a this indicates that the observed exuberant apoptosis is more
pig was vaccinated twice with foot-and-mouth disease virus likely to result from a pathological component than a normal
vaccine (O1 serotype, Bayer) 3 weeks apart. Seven days after immune activation.
the second vaccine the pig was sacrificed and the draining Apoptosis of lymphocytes and ASFV infection of cells in
lymph node was removed, paraffin embedded and stained with bushpig lymph nodes were mostly confined to the B cell

BEEB
C. A. L. Oura and others

Fig. 3. Apoptosis in bushpig lymph node infected with ASFV (Malawi isolate). Paraffin embedded lymph node sections from a
bushpig, 10 days p.i. with Malawi isolate (¬160). (A) Stained with haematoxylin and eosin ; (B) stained with TUNEL ;
(C) immunostained with anti-vp73 MAb 4H3 ; (D) lymph node cryosection from a bushpig, 10 days p.i. with Malawi isolate,
immunostained with anti-CD21 MAb C35. In immunohistochemical and TUNEL staining, signal was visualized with DAB
substrate (brown) and sections were counterstained with haematoxylin. F, B cell follicle.

follicles, with low levels of apoptosis and ASFV infection apoptotic mediators, inducing surrounding B cells to enter
occurring in T cell zones of the cortex and paracortex. This apoptosis ; (2) the lymphocyte selection process is impaired
observation is in marked contrast to the situation in domestic through damage to follicular dendritic cells so most of the B
pigs infected with Malawi isolate where the majority of ASFV cells in the germinal centres are selected for programmed cell
infected cells and apoptosis of lymphocytes were located in the death, with few lymphocytes being positively selected for
T cell areas (Oura et al., 1998 a ; Carrasco et al., 1996). There are continued survival ; or (3) the process of affinity maturation is
many possible causes of this B lymphocyte apoptosis which compromised so that only low affinity antibody-producing B
include : (1) ASFV infected tingible body macrophages are lymphocytes are produced. Because only high affinity
indirectly, perhaps through the release of cytokines or other antibody-producing lymphocytes are selected for survival in

BEEC
Pathogenesis of ASF in the bushpig

the germinal centres, the majority of B lymphocytes enter References


apoptosis. Anderson, E. C., Hutchings, G., Mukarati, N. & Wilkinson, P. J. (1998).
ASFV has evolved in a natural cycle between the reservoir African swine fever virus infection of the bushpig (Potamchoerus porcus)
hosts : the bushpig, the warthog and the soft tick. Sufficiently and their significance in the epidemiology of the disease. Veterinary
high viraemias occur in the bushpig to allow infection of Microbiology (in press).
feeding ticks, but ASFV replication in lymphoid tissues is kept Carrasco, L., Chacon, M., de Lara, F., Martin de las Mulas, J., Gomez-
in check, and pathological damage and apoptosis in lymphoid Villamandos, J. C., Perez, J., Wilkinson, P. J. & Sierra, M. A. (1996).
Apoptosis in lymph nodes in acute African swine fever. Journal of
tissue are kept to a minimum so that the hosts survive infection Comparative Pathology 115, 415–428.
with no clinical signs of disease. Also, a controlled level of
Cobbold, C., Whittle, J. T. & Wileman, T. (1996). Involvement of the
apoptosis in uninfected lymphocytes may result in a diminished endoplasmic reticulum in the assembly and envelopment of African swine
immune response enabling ASFV infected cells to survive the fever virus. Journal of Virology 70, 8382–8390.
immune system for sufficient time to produce a productive DeTray, D. E. (1963). African swine fever. Advances in Veterinary Science
infection. In addition, bushpigs may also control the apoptosis 8, 299–333.
of B lymphocytes at the time of selection of high affinity Haresnape, J. M., Wilkinson, P. J. & Mellor, P. S. (1984). Isolation of
antibody producing B lymphocytes (10 days p.i.). This would African swine fever virus from ticks of the Ornithodorus moubata complex
halt the production of high affinity anti-ASFV antibody and (Ixodoidae : Argasidae) collected within the African swine fever enzootic
facilitate long term survival of extracellular virus adsorbed to area of Malawi. Epidemiology and Infection 101, 173–185.
the surface of red blood cells, enabling its transmission to Montgomery, R. E. (1921). On a form of swine fever occurring in British
feeding ticks. This may also have important implications for East Africa (Kenya colony). Journal of Comparative Pathology 34, 243–262.
the persistence of the ASFV in the bushpig and suggests that Oura, C. A. L., Powell, P. P. & Parkhouse, R. M. E. (1998 a). African
swine fever : a disease characterized by apoptosis. Journal of General
antibody mediated immunity may not be critically important Virology 79, 1427–1438.
for bushpig survival. Characterization of bushpig and domestic
Oura, C. A. L., Powell, P. P. & Parkhouse, R. M. E. (1998 b). Detection
pig antibody responses are being carried out. of African swine fever virus in infected pig tissues by immuno-
In contrast, the domestic pig is not a natural host of ASFV. cytochemistry and in situ hybridisation. Journal of Virological Methods (in
The factors that keep both the virus and apoptosis in check in press).
bushpigs are ‘ out of control ’ in domestic pigs. High levels of Plowright, W. (1977). Vector transmission of African swine fever virus.
virus replication in lymphoid tissue result in uncontrolled In Hog Cholera}Classical Swine Fever and African Swine Fever, EEC
apoptosis of lymphocytes, a severely impaired immune Publication EUR 5904 EN, pp. 575–587. Edited by B. Liess. Luxembourg :
response, and perhaps aberrant levels of cytokines contributing Office for Official Publications of the European Communities.
to the haemorrhagic pathology. Therefore domestic pigs die Saalmuller, A. (1996). Characterisation of swine leukocyte differ-
entiation antigens. Immunology Today 17, 352–354.
within 5–7 days of infection.
ASF with its differential pathology, haemorrhagic in the Thomson, G. R. (1985). The epidemiology of ASF : the role of free-
living hosts in Africa. Onderstepoort Journal of Veterinary Research 52, 201.
susceptible domestic pig versus non-pathogenic in the resistant
Thomson, G. R., Gainaru, M. D. & Van Dellen, A. F. (1980). Ex-
bushpig, may be considered as a veterinary model for the
perimental infection of warthog (Phacochoerus aethiopicus) with ASFV.
recently emerging haemorrhagic diseases of man. Onderstepoort Journal of Veterinary Research 47, 19–22.
Wilkinson, P. J., Donaldson, A. I., Greig, A. & Bruce, W. (1977).
We thank Dr Stuart Hargreaves, Director of Veterinary Services, Transmission studies with African swine fever virus. Infection of pigs by
Harare, Zimbabwe for providing laboratory space in Zimbabwe. Thanks airborne virus. Journal of Comparative Pathology 87, 487–495.
to Mick Denyer for help with the animal work. The field work was funded
by the Overseas Development Administration (ODA) and the work was
also funded in part by MAFF Contract 1506. Received 11 November 1997 ; Accepted 3 February 1998

BEED
Virus Research 173 (2013) 122–130

Contents lists available at SciVerse ScienceDirect

Virus Research
journal homepage: www.elsevier.com/locate/virusres

Review

Pathogenesis of African swine fever in domestic pigs and European wild boar
Sandra Blome ∗ , Claudia Gabriel, Martin Beer
Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493 Greifswald-Insel Riems, Germany

a r t i c l e i n f o a b s t r a c t

Article history: African swine fever (ASF) is among the most important viral diseases that can affect domestic and feral
Available online 6 November 2012 pigs. Both clinical signs and pathomorphological changes vary considerably depending on strain viru-
lence and host factors. Acute infections with highly virulent virus strains lead to a clinical course that
Keywords: resembles a viral haemorrhagic fever that is characterized by pronounced depletion of lymphoid tissues,
African swine fever apoptosis of lymphocyte subsets, and impairment of haemostasis and immune functions. It is generally
Pathogenesis
accepted that most lesions can be attributed to cytokine-mediated interactions triggered by infected
Domestic pigs
and activated monocytes and macrophages, rather than by virus-induced direct cell damage. Neverthe-
European wild boar
less, most pathogenetic mechanisms are far from being understood. This review summarizes the current
knowledge and discusses implications and research gaps.
© 2012 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
2. Clinical and pathological observations upon ASFV infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
2.1. ASFV infection results in a wide range of clinical pictures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
2.2. Gross pathological findings and virus detection mirror clinical courses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
2.3. Blood count changes and phenotypic characteristics vary in the course of acute and chronic infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
3. Changes in primary target cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 125
3.1. Susceptibility of monocyte/macrophages to ASFV infection increases upon maturation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 125
3.2. Secretory and phagocytic activation of monocyte/macrophages leads to proinflammatory and procoagulant responses . . . . . . . . . . . . . . . . 125
4. Viral distribution, secondary replication sites and pathomorphological correlates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 126
4.1. Viral replication leads to widespread lymphoid depletion through destruction and apoptosis of monocyte/macrophages and
uninfected lymphocytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 126
4.2. Haemorrhagic lesions are associated with release of cytokines by infected monocyte/macrophages rather than direct endothelial
cell damage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 126
4.3. Impairment of platelets and coagulation changes contribute to evolution of a haemorrhagic syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 127
4.4. Additional cytokine and acute phase responses link pathogenesis and immune response . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
5. Résumé and gap analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128

1. Introduction family (Takamatsu et al., 2011). Due to the fact that soft ticks of
the genus Ornithodoros can be involved in the infection cycle, ASFV
Due to its relevance for animal health and pig industry, African is regarded as the only DNA virus that can be classified as an ARBO
swine fever (ASF) is one of the most important viral diseases of (arthropod borne) virus (Kleiboeker and Scoles, 2001).
domestic pigs. It was shown that it impairs pig production consid- In the vertebrate host, the virus replicates in cells of the
erably, especially in Africa (Penrith and Vosloo, 2009). The causative mononuclear phagocyte system (in older literature referred to
agent, African swine fever virus (ASFV), is a large and complex as reticuloendothelial system), predominantly monocytes and
DNA virus of the genus Asfivirus within the Asfarviridae virus macrophages although several other cell types can be infected,
especially in the later stages of the disease (Carrasco et al., 1996b,
1996d; Fernandez et al., 1992a; Mebus, 1987, 1988; Perez et al.,
∗ Corresponding author. Tel.: +49 38351 7 1144; fax: +49 38351 7 1275. 1994; Ramiro-Ibanez et al., 1995). It could be demonstrated in
E-mail addresses: sandra.blome@fli.bund.de (S. Blome), new-born piglets that primary viraemia occurs approximately 8 h
claudia.gabriel@fli.bund.de (C. Gabriel), martin.beer@fli.bund.de (M. Beer). post infection. Secondary viraemia is detected from 15 to 24 h post

0168-1702/$ – see front matter © 2012 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.virusres.2012.10.026
S. Blome et al. / Virus Research 173 (2013) 122–130 123

infection and after 30 h, virus is found in almost all organs (Col- up to somnolence, anorexia, slight catarrhal conjunctivitis, vomi-
grove et al., 1969, cited by the EFSA Scientific Report 2009). During ting, watery diarrhoea, accelerated respiratory and pulse rate, and
viraemia, haemadsorbing ASFV isolates are found associated with ataxia/incoordination prior to death (Gabriel et al., 2011; Kolbasov
erythrocytes (Quintero et al., 1986; Wardley and Wilkinson, 1977), and Blome, unpublished data). In these studies, it could be demon-
but also with lymphocytes and neutrophils (Plowright et al., 1994). strated that the clinical course in domestic pigs was irrespective of
Infection correlates with a wide range of clinical syndromes from the route of infection (oro-nasal, intramuscular or infection through
almost inapparent disease to a haemorrhagic fever like illness with direct contact with infected pen-mates). In all experimental infec-
high mortality (Penrith and Vosloo, 2009; Takamatsu et al., 2011). tions, mortality was 100% in less than 10 days. A similar picture
Nowadays it is generally accepted that the massive destruc- was observed upon oral or intramuscular infection of European
tion of macrophages plays a major role in pathogenesis, especially wild boar although skin colour and perfusion could not be assessed
in the impaired haemostasis due to the release of active sub- (Blome et al., 2012; Gabriel et al., 2011). In wild boar, severe depres-
stances including cytokines, complement factors and arachidonic sion, anorexia, diarrhoea, and respiratory distress predominated.
acid metabolites (Penrith et al., 2004b). The genesis of thrombo- Furthermore, central nervous symptoms including ataxia and con-
cytopenia that occurs particularly during the course of acute and vulsions as well as epistaxis were observed in some cases prior to
subacute ASFV infection is controversially discussed. While some death (Gabriel et al., 2011; Kolbasov and Blome, unpublished data).
authors show evidence that infection and destruction of megakary-
ocytes may play a role (Rodriguez et al., 1996a), other researchers 2.2. Gross pathological findings and virus detection mirror
did not find structural or ultrastructural correlates for an clinical courses
impaired thrombocytopoiesis (Gomez-Villamandos et al., 1997a).
Pigs infected with ASFV generally suffer from severe lymphopenia Like the major clinical signs, pathological findings vary consid-
that could be attributed to apoptosis of lymphocytes. Production of erably depending on the course of disease. Frequently observed
pro-inflammatory cytokines by infected macrophages is strongly lesions in domestic pigs include enlarged and haemorrhagic gas-
implicated in induction of apoptosis in lymphocyte populations trohepatic and renal lymphnodes, splenomegaly with necrotic foci,
(Gomez-Villamandos et al., 1995a, 2003; Oura et al., 1998a). petechiae in kidneys, ecchymoses in serosae, alveolar haemor-
The also described chronic forms of ASF might have an auto- rhages and oedema of the lungs (Kleiboeker, 2002; Maurer and
immune component, and lesions could result from the deposition of Griesemer, 1958; Colgrove et al., 1969, reviewed by Rodriguez et al.,
immune-complexes in tissues such as kidneys, lungs and skin with 1996a). Secondary infections may complicate the clinical and path-
their subsequent binding to complement (EFSA Scientific Report omorphological picture. The same is seen in European wild boar of
2009; Plowright et al., 1994). all age classes upon highly virulent infection under experimental
Despite intensive research efforts, most of these pathogenetical conditions (Blome et al., 2012; Gabriel et al., 2011), and also in other
aspects are still far from being understood. This review compiles the feral pigs (McVicar et al., 1981). Field data showed that all courses
current knowledge of pathogenetical mechanisms and their clinical of the disease including almost inapparent ones are possible (Perez
and pathomorphological correlates, discusses their implications, et al., 1998). Exemplary pathological findings in European wild boar
and identifies research gaps. are depicted in Figs. 1 and 2.
Particularly during the clinical phase of infection, virus and viral
DNA can be detected by virus isolation and real-time PCR in all se-
2. Clinical and pathological observations upon ASFV and excretions of infected pigs, but the viral load varies consid-
infection erably (Gabriel et al., 2011). While high amounts of virus or viral
DNA can be detected in blood samples, all swab samples (rectal,
2.1. ASFV infection results in a wide range of clinical pictures pharyngeal, nasal, and preputial/vaginal) show considerably lower
viral loads (Ekue et al., 1989; Gabriel et al., 2011). In detail, up to
In both domestic pigs and wild boar, clinical signs of ASF vary 3 × 106 copies can be detected per ␮l DNA from 200 ␮l of blood,
considerably from peracute deaths to inapparent courses and cor- whereas only 4–8 × 103 copies per ␮l can be found in rectal swabs,
relate to a wide range of pathomorphological observations. Reports and 2–6 × 102 copies per ␮l in oropharyngeal swabs (Gabriel and
on naturally resistant domestic pigs in Africa remain controver- Blome, unpublished data). Exemplary data depicting the detection
sially discussed and do not seem to have a genotypic correlate as of viral DNA in blood and swab samples from European wild boar
the offspring of less susceptible pigs does not inherit this advan- and domestic in-contact pigs are shown in Table 1. It is not sur-
tage (Penrith et al., 2004a). Upon infection with a virulent strain, prising that contagiosity is mainly linked to blood and that contact
clinical signs develop after an incubation period of 2 to 7 (sel- alone is less efficient (Heuschele, 1967).
dom up to 14) days (Mebus, 1988). They may include high fever,
reddened skin at the acra, severe depression, anorexia, conjunctivi- 2.3. Blood count changes and phenotypic characteristics vary in
tis, vomiting, watery to bloody diarrhoea, accelerated respiratory the course of acute and chronic infections
and pulse rate, abortion in pregnant sows, cyanosis, and incoor-
dination (EFSA Scientific Report, 2009). Acute lethal forms can In general, infection with ASFV is accompanied by leukopenia
be accompanied by haemorrhagic lesions (petechiae, epistaxis) and thrombocytopenia, but extent and nature may vary with strain
(Gomez-Villamandos et al., 2003). Characteristics are thrombo- virulence and host factors.
cytopenia, petechial bleedings, and apparently increased vascular It has been reported by Wardley and Wilkinson (1977) that
permeability with extravasation of blood components. In less acute infection with the highly virulent Kirawira isolate (KWH/12) (Greig
disease courses, respiratory signs (coughing, sneezing, and dysp- and Plowright, 1970) leads to a decrease in leucocyte counts while
noea) and gastrointestinal lesions (mainly watery diarrhoea but the red blood count remains almost unchanged. In this study,
also obstipation) are frequently seen. Depending on strain viru- lymphocyte numbers decreased while an increased number of neu-
lence, mortality ranges from 3 to 100%. trophils, especially juvenile subsets, was observed. Virus could be
The ASFV strain currently affecting the Trans-Caucasian demonstrated in all major blood fractions, but over 90% were asso-
countries, Russia, and Ukraine, is highly virulent and leads to ciated with red blood cells. These findings are in agreement with
the above mentioned clinical picture with high fever, reddened earlier observations by Detray and Scott (1957). In contrast, Gomez-
skin and cyanosis, especially upon agitation, severe depression Villamandos et al. (1997a) did not observe significant changes
124 S. Blome et al. / Virus Research 173 (2013) 122–130

Table 1
Real-time PCR results from EDTA blood, oropharyngeal and faecal swab samples depicted as quantification cycle (cq) values. Wild boar were infected orally with 2 ml of
a spleen suspension containing 106 tissue culture infectious doses 50% of the highly virulent Caucasian ASFV strain per ml. Domestic pigs were co-housed two days after
inoculation of the wild boar. Wb: Wild boar; Dp: Domestic Pig; nd: not done. Grey shaded values indicate fever (±40 ◦ C) at that time point. −Negative result in qPCR; †Animal
was dead at this time point; *Serum instead of EDTA-blood sample.

Days post infection of wild boar

Animal Samples 0 1 2 3 4 5 6 7 9 13 17 20

Wb 1 Blood – nd – nd nd 23
Oropharyngeal swab – – – – nd – †
Faecal swab – – – – nd –
Wb 2 Blood – nd – nd nd 22 20 24
Oropharyngeal swab – – – – nd 37 37 37 †
Faecal swab – – – – nd – 38 -
Wb 3 Blood – nd – nd nd 28 22 23
Oropharyngeal swab – – – – nd – 38 34 †
Faecal swab – – – – nd 37 34 33
Wb 4 Blood – nd – nd nd 25 26 26
Oropharyngeal swab – – – 37 nd – 34 37 †
Faecal swab – – – – nd 30 29 33
Wb 5 Blood – nd 39 nd nd 25 23
Oropharyngeal swab – – – – nd 39 35 †
Faecal swab – – – – nd – 29
Wb 6 Blood – nd – nd nd 23 24
Oropharyngeal swab – – – 37 nd – 34 †
Faecal swab – – – – nd 35 32
Dp 1 Blood – nd nd nd nd nd 39 nd – 21 20 †
Dp 2 Blood – nd nd nd nd nd – nd – 23 nd †
Dp 3 Blood – nd nd nd nd nd – nd – – nd 29*

in total white blood cell counts, percentages of monocytes and different clinical courses. Moreover, interpretation of tendencies
platelets counts upon infection with the highly virulent Malawi by the authors varied to a certain extent. Even after infection with
1983 ASFV isolate (Haresnape et al., 1988), but lymphopenia and the Kirawira isolate of ASFV, leukopenia barely reached patholog-
neutrophilia was seen. This discrepancy may arise from slightly ical values (i.e. values <10 × 103 cells per ␮l) before the animals
succumbed to infection.
Other studies showed that acute infection with highly vir-
ulent viruses leads to a severe decrease of macrophages and
B-lymphocytes (reduction of 60% with respect to pre-infection
values) after a short period with increased B-lymphocyte counts
(probably due to polyclonal activation period of B-lymphocytes)
and an increase in macrophages at the beginning of infection
(Ramiro-Ibanez et al., 1997). In this study, severe leukopenia was
observed in global white blood cell counts 7 days post infection with
the highly virulent E70 isolate of ASFV. These findings coincided
with reduced relative numbers of lymphocytes and monocytes.
Affected lymphocyte subsets were, apart from the already men-
tioned B-lymphocytes, CD4+ T-helper cells. Furthermore, CD8+
cytotoxic T-cells increased by the end of the first week.
Investigation of phenotypic antigens and activation markers
revealed that at the peak of viral titres, an increase is seen in both

Fig. 1. Gross pathological lesions in a European wild boar piglet. The boar (approx-
imately six months of age) was infected oronasally with 3 × 106 tissue culture
infectious doses 50% (see table above) of the highly virulent Caucasian ASFV isolate. Fig. 2. Mandibular lymph nodes of a crossbred weaner domestic pig (approximately
This animal was euthanized in a moribund state at eight days post infection, show- 30 kg). This animal acted as in-contact control to European wild boar piglets which
ing severe depression, high fever (41.8 ◦ C), tachypnoea, uncontrolled movements of were orally inoculated with 2 × 106 tissue culture infectious doses 50% (see table
hind legs and inability to stand. (A) Acute diffuse ecchymosis and petechiae on the above) of the Caucasian ASFV isolate. The animal was brought into the same pen
mucosal surface of the stomach. (B) Kidney with numerous acute petechiae on the three days after inoculation, and died 16 days after initial contact. Note high-grade,
renal cortex. acute, diffuse haemorrhages and enlargement.
S. Blome et al. / Virus Research 173 (2013) 122–130 125

SLA II (MHC-II) and CD8+ expression. These findings are accom- expressing higher levels of macrophage specific markers and SLA
panied by down regulation of IL-2 (CD25) receptor expression II antigens were most susceptible to ASFV infection. Especially
(Ramiro-Ibanez et al., 1997). As IL-2 is crucial for the modulation expression of CD163, an acute phase regulated receptor, and sur-
of regulatory and effector lymphocyte functions, T-cell response face antigen 4E9 (now known as porcine CD107a, or lysosomal
seems to be influenced. Similar changes in IL-2 receptor and SLA II associated membrane protein 1) correlates with permissiveness to
expression were reported by Canals et al. (1995) after investigation ASFV infection (Sanchez-Torres et al., 2003). The fact that infection
of ASFV-infected cell cultures. could be inhibited by anti-CD163 or anti-4E9 monoclonal antibod-
Studies using a moderately virulent ASFV strain from the ies demonstrates that both antigens play a role in the initial stages
Dominican Republic (DR II, isolated in 1979) showed that virus of viral infection (Sanchez-Torres et al., 2003). This is an interesting
could be found in the isolated erythrocyte fraction as well as in finding as it is known that CD163+ monocytes produce more TNF-␣,
plasma and mononuclear leukocytes from peripheral blood after express high levels of adhesion molecules and are better at pre-
oro-nasal infection (Knudsen and Genovesi, 1987). In this case, senting antigens to T-cells when compared to CD163− monocytes
virus infection had only minor effects on the number of circulat- (Chamorro et al., 2005). This may have an impact on the influence
ing leukocytes as just slight increases in neutrophils and decreases on endothelial cells and apoptosis induction capacity as well as
in lymphocyte counts were observed early upon infection. Respon- the timeframe of pathogenesis. The more mature scavenger type
siveness of mononuclear cells to mitogenic stimuli was preserved macrophage seems to fulfil the needs for ASFV replication much
throughout the trial. The latter disagrees with studies by other better than monocytes. One explanation could be the need for rapid
authors where severe impairment of responsiveness to mitogenic vesicle acidifications and lysosomal enzyme activity (Basta et al.,
stimuli was observed (Gonzalez et al., 1990; Wardley, 1982). 1999). Nevertheless, further investigations are needed to confirm
Chronically infected animals were shown to exhibit marked and consolidate these results.
differences in blood count changes. During this disease course, B- In peripheral blood of experimentally infected animals, mainly
lymphocytes and macrophages showed an increase of 100% within monocytes and macrophages are positive for viral p30 antigen as
the first week after infection with the attenuated E75CV1 4 strain shown by flow cytometric analyses with a maximum percentage
with a peak that coincided with high viraemia, onset of fever, and ranging from 6 to 31% at days 7–9 post infection with an attenuated
detectable antibodies (Ramiro-Ibanez et al., 1997). These values strain and at day 5 after virulent ASFV infection (Ramiro-Ibanez
were back to normal after the second week post infection. Both et al., 1995). The above mentioned influence of the maturation
CD8+ and CD4+ T cells showed an elevation during the second week state may explain the relatively low number of infected cells on
of infection. In this case, both SLA I (MHC-I) and SLA II showed the peak of infection and thus viral replication. In addition to the
an elevated expression on peripheral mononuclear cells indica- monocyte/macrophage subset, a small proportion of granulocytes
tive for stimulation of the immune system. These findings were (7–21% of all infected cells) is also infected. Neither time point after
in agreement with in vitro studies and findings in spleen tissue sec- infection nor virus isolate led to significant differences regarding
tions (Gonzalez-Juarrero et al., 1992a, 1992b). At the third week affected cell subsets (Ramiro-Ibanez et al., 1995). Infection of neu-
post infection, the expression was significantly decreased. This may trophils was confirmed by Carrasco et al. (1996d) who could show
have implications for viral clearance through an impaired antigen that both mature and immature neutrophils can harbour virus.
presentation. Here again, a decrease in IL-2 receptor expression Especially the latter are discussed as transport vehicles for virus
was observed, underlining the possible role in the pathogenesis of spreading (Gomez-Villamandos et al., 1997a).
lesions (Canals et al., 1995).
3.2. Secretory and phagocytic activation of
monocyte/macrophages leads to proinflammatory and
3. Changes in primary target cells procoagulant responses

3.1. Susceptibility of monocyte/macrophages to ASFV infection Upon ASFV infection, several macrophage subpopulations
increases upon maturation show signs of secretory and/or phagocytic activation. Moreover,
destruction of monocytes/macrophages leads to release of cel-
Without tick involvement, ASFV enters the body via the tonsils lular components. Activated monocytes/macrophages secrete a
or dorsal pharyngeal mucosa to the mandibular or retropharyngeal wide range of mediators including proinflammatory cytokines
lymph nodes, from where the virus spreads systemically through such as IL-1, IL-6, and TNF-␣ (Murtaugh et al., 1996). These
viraemia (Greig, 1972; Plowright et al., 1994). Thereafter, the virus cytokines can trigger acute phase reactions, inflammation, acti-
is detectable in almost all tissues. Highest titres are observed in tis- vation of endothelial cells, and apoptosis. Among these factors,
sues containing a large component of the mononuclear phagocyte TNF-␣ is particularly important (Gomez del Moral et al., 1999).
system (reticuloendothelial cellular elements) like the spleen and It can induce vascular changes (vasodilatation and increased per-
lymphnodes (Heuschele, 1967). meability) and modulates the activation status of the vascular
The main target cells of ASFV, which are entered by endo- endothelium (procoagulant/anticoagulant). Moreover, TNF-␣ is
cytosis, probably involving the mechanism of macropinocytosis involved in the control of apoptosis.
(Sanchez et al., 2012), belong to the monocyte/macrophage lineage It could be demonstrated that an increase in the production of
(Malmquist and Hay, 1960; Sanchez-Torres et al., 2003; Sierra et al., TNF-␣, IL-1␣ and IL-1␤, and IL-6 coincides with onset of fever, vas-
1990). This cell population is highly diverse and comprises different cular damage, and changes in lymphoid structures (Salguero et al.,
phenotypes, activation and maturation stages. In addition, infection 2002). Detection of cytokines in tissues correlates with detection
of dendritic cells is observed which may interfere with humoral of VP73 antigen in cells of the monocyte/macrophage lineage and
immune responses (Gregg et al., 1995). an increase in serum levels of TNF-␣ and IL-1␤.
It was observed that different subsets of monocytes and Comparison of cytokine responses in macrophages infected
macrophages show a different susceptibility to ASFV, both in vivo in vitro with low and highly virulent ASFV strains indicate that
(Carrasco et al., 1992; Rodriguez et al., 1996a) and in vitro. attenuated strains show an altered response favouring cytokines
Bone marrow derived cells and fresh blood monocytes are less involved in cellular immunity, namely IFN-␣ and IL12p40 (Gil et al.,
susceptible to in vitro infection than alveolar macrophages. In 2008). Thus, survival of animals may at least partly depend on the
general, it could be shown that cells of higher maturation stages nature of cytokine responses. An interesting finding in line with
126 S. Blome et al. / Virus Research 173 (2013) 122–130

both above mentioned considerations is that pulmonary intravas- Besides general lymphoid depletion in the spleen, infected
cular macrophages but not alveolar macrophages are infected macrophages attached to smooth muscle cells were observed in
(Carrasco et al., 2002; Oura et al., 1998b). Infection of these cells the splenic cords of affected animals upon highly virulent infection.
leads to intense activation and consequently expression of IL-1␣ These cells rapidly disappeared and were replaced by numerous
and TNF-␣. A clear correlation was observed between this cytokine erythrocytes associated with fibrin deposits. Fibrin deposition and
response and interstitial oedema and fibrin microthrombi forma- platelet activation was probably due to the exposure of collagen
tion in septal capillaries (Carrasco et al., 2002). These findings were upon destruction of infected macrophages (Carrasco et al., 1997a).
supported by earlier studies of the same group that showed that In the tonsils of acutely infected animals, an increased number
activation of pulmonary intravascular macrophages could play a of monocytes/macrophages was observed along with an increase
major role in the genesis of lung-associated lesions (Carrasco et al., in the expression of proinflammatory cytokines (especially TNF-␣
1996a). and IL-1␣). This finding was accompanied by lymphocyte apoptosis
(Fernandez de Marco et al., 2007).
Comparison of moderately and highly virulent ASFV strains
4. Viral distribution, secondary replication sites and showed no general differences in terms of cell tropism or organ
pathomorphological correlates distribution, but significantly more severe tissue destruction with
increasing virulence (Oura et al., 1998a). The cellular depletion
4.1. Viral replication leads to widespread lymphoid depletion described above was previously attributed to necrosis, but it could
through destruction and apoptosis of monocyte/macrophages and be shown beyond doubt that the changes are attributable to apop-
uninfected lymphocytes totic mechanisms (Carrasco et al., 1996c; Gomez-Villamandos et al.,
1995a; Oura et al., 1998a; Ramiro-Ibanez et al., 1996, 1997). It
Immunohistochemical studies upon infection with moder- concerns both infected cells of the monocyte/macrophage lineage
ately and highly virulent ASFV strains showed that infection as well as a large number of uninfected lymphocytes (Ramiro-
and depletion is initially found in monocytes/macrophages in Ibanez et al., 1996). Apoptosis is also present in lymphoid tissues
lymphoid organs, especially lymphnodes and spleen (Ramiro- associated with tonsils, bronchia, and the gastrointestinal system
Ibanez et al., 1997; Rodriguez et al., 1996a). Later on, additional (Ramiro-Ibanez et al., 1997) as well as in the liver and kidney tissues
cell types in various organs including megakaryocytes (Edwards (Gomez-Villamandos et al., 1995a). Apoptotic cells have been found
et al., 1985), tonsillar epithelial cells (Gomez-Villamandos et al., from 2 to 4 days post infection onwards in spleen and lymphnodes,
1997b; Rodriguez et al., 1996a), hepatocytes, kidney cells and from day 3 post infection in the thymus (Oura et al., 1998a;
and endothelial cells (Gomez-Villamandos et al., 1995a) get Salguero et al., 2002). These cells were mainly lymphocytes, both
infected. In vivo haemadsorption is observed in hepatic sinu- in B- and T-cell areas of lymphoid organs (Salguero et al., 2002).
soids, lymph sinuses, and red splenic pulp (Sierra et al., Order and extent of depletion of these cell subsets varied among
1991). studies using different ASFV strains (Carrasco et al., 1996c; Oura
In the liver, Kupffer cell infection is accompanied by activa- et al., 1998a; Ramiro-Ibanez et al., 1997).
tion, erythrophagocytosis, in vivo haemadsorption, and lymphocyte In line with the findings in the tonsils, the appearance of lym-
attachment (Gomez-Villamandos et al., 1995b). It was observed phocyte apoptosis in splenic and thymic structures as well as
that Kupffer cell and sinusoidal circulating cell counts increase lymphnodes coincided with expression of TNF-␣, IL-1␣ and IL-1␤
initially, and that the former show secretory activation in terms by activated monocytes/macrophages (Salguero et al., 2005). Thus,
of IL-1␣, TNF-␣, and IL-6 expression (Sanchez-Cordon et al., proinflammatory cytokines seem to be responsible for apoptosis
2008). Thus, infectable cells are concentrated. Fibrin networks and following ASFV infection.
microthrombi are found in sinusoidal lumina of peripheral lobular Based on these findings, research groups subsequently identi-
areas. Beside this macrophage population, infection of endothelial fied several ASFV genes involved in programmed cell death both in
cells is observed from seven days post inoculation with highly viru- an inhibitory or an inducing manner (Afonso et al., 1996; Brun et al.,
lent ASFV such as the Malawi ’83 strain (Gomez-Villamandos et al., 1996; Chacon et al., 1995; Galindo et al., 2008; Hernaez et al., 2004a,
1995b). 2004b; Hurtado et al., 2004; Nogal et al., 2001; Portugal et al., 2009;
The kidney is another organ with severe pathomorphologi- Revilla et al., 1997; Rodriguez et al., 2002; Zsak and Neilan, 2002).
cal changes including haemorrhages. In acute ASF, proliferative In contrast to changes seen in the course of acute-lethal
glomerulonephritis including hyperplasia of collecting ducts infection, the chronic course is characterized by a rather severe
(Gomez-Villamandos et al., 1995c, 1995d) is seen while in sub- hyperplasia of lymphoid organs as evidenced by enlarged lymphn-
acute forms immune-mediated, subendothelial and mesangial odes with hypercellularity (Ramiro-Ibanez et al., 1997). In general,
deposits of immunoglobulins and complement prevail (Hervas immune complexes may play a role in subacute ASFV infections
et al., 1996a). Haemorrhages seen in the kidney are attributed by (Fernandez et al., 1992c).
different authors to endothelial dysfunction, aggravated by virus In recovering pigs, Oura et al. (1998a) found that virus persisted
replication in endothelial cells in the finals stages of the disease in lymphnodes and tonsils up to 48 days post infection. Virus was
(Gomez-Villamandos et al., 1995c). In contrast, other groups dis- located in cells surrounded by apoptotic lymphocytes (up to 32 dpi).
cuss endothelial damage as direct cause for the observed petechiae This confirms again the role of apoptosis of uninfected lymphocytes
(Sierra et al., 1989). Upon acute infection with highly virulent induced by cytokines released by infected macrophages.
ASFV strains, the renal interstitium exhibits intense oedema and
macrophage infiltrates. ASFV replication in mesangial cells and 4.2. Haemorrhagic lesions are associated with release of
renal collecting duct epithelial cells could account for virus in cytokines by infected monocyte/macrophages rather than direct
urine (without haematuria) (Gomez-Villamandos et al., 1995d). endothelial cell damage
In addition, virus replication in fibroblasts and smooth muscle
cells of small blood vessels is seen in later stages. In case of Infection and destruction of monocytes/macrophages in lym-
subacute and chronic forms, immune mediated events including phoid organs coincides with the appearance of first haemorrhagic
immune-complex deposits are discussed as a cause of intersti- lesions in these organs starting from day 3 post infection
tial haemorrhages associated with diapedesis (Hervas et al., 1996a, with a virulent ASFV strain (Salguero et al., 2005). Among the
1996b). sites with frequent haemorrhages are gastro-hepatic and renal
S. Blome et al. / Virus Research 173 (2013) 122–130 127

lymphnodes (haemorrhagic enlargement). Following infection was normal during infection as measured by incorporation of the
with the Malawi ’83 isolate, these changes are accompanied by radionuclides into platelets. Gomez-Villamandos et al. (Gomez-
activation of endothelial cells and fibrin deposition in the vessel Villamandos et al., 1997a) also demonstrated that neither vascular
lumina. Moreover, increased fenestration and exposure of basal changes nor viral replication in different bone marrow cell popu-
membrane to the blood stream could be observed (Carrasco et al., lations gave rise to impaired bone marrow functions. They even
1997b). Following infection with highly virulent ASFV isolates, acti- observed an increased haematopoiesis. Thus, it remains debatable
vation of capillary endothelial cells was also accompanied by an whether impaired thrombocytopoiesis plays a major role in the
increased fibrinolytic activity and high levels of fibrin monomers pathogenesis of thrombocytopenia in ASF despite the fact that bone
(Villeda et al., 1995). marrow lesions are observed.
In early pathology and pathogenesis studies, the haemorrhagic Consumption of platelets in the periphery, the second dis-
picture was attributed to direct infection of endothelial cells cussed mechanism leading to thrombocytopenia, could be due to
(Maurer and Griesemer, 1958). This hypothesis was strengthened microthrombi as a correlate for disseminated intravascular coag-
by in vitro infection of endothelial cells (Wilkinson and Wardley, ulation. In acute forms of ASF, fibrin depositions with embedded
1978) and demonstration of virus and viral antigen in endothelial platelets are frequently observed (Rodriguez et al., 1996a). Mech-
cells of kidney and liver (Fernandez et al., 1992a, 1992b). Later stud- anisms leading to these microthrombi could include systemic
ies showed that only a minority of endothelial cells was infected in release of enzymes, cytokines, complement factors, and products
organs with obvious haemorrhagic pathology, and that only in the of the arachidonic acid metabolism. These factors are known as
late stages of the disease, infection of endothelial cells was found at pathogenetic mechanisms in other diseases featuring the same
all. At this time, haemorrhagic lesions had been present already for clinical and pathomorphological correlates (Lange et al., 2011). It
some days. For this reason, infection of endothelial cells does not has to be stated that the acute phase of thrombocytopenia coin-
seem to be the primary cause of haemorrhage (Gomez-Villamandos cides with marked viraemia and high levels of immunoglobulin.
et al., 1997b; Perez et al., 1994). Thus, immune mediated microthrombi and aggregates involving
It is now an acknowledged opinion that neither vascular antigen-antibody complexes cannot be ruled out. In conclusion,
nor lymphoid lesions are directly linked to viral replication in it seems likely that both impairment of thrombocytopoiesis and
endothelial cells or lymphocytes (Carrasco et al., 1997a; Gomez- peripheral consumption play a role in the genesis of thrombocyto-
Villamandos et al., 1995a, 1995c). These findings are in line with the penia.
observations known for several other viral diseases with haemor- Beside the obvious changes in count, both activation and
rhagic lesions (Gomez-Villamandos et al., 2003; Lange et al., 2011). degranulation of platelets could be observed by Gomez-
Upon ASFV infection, activated endothelial cells coincide with Villamandos et al. (1996) after highly and moderately virulent ASFV
ASFV replication in neighbouring monocytes and macrophages. infection. This finding coincided with infection and activation of
Thus, stimulation of endothelial cells is likely to result from monocytes/macrophages and probably the secretion of platelet
cytokine release. Proinflammatory cytokines such as TNF-␣ and IL- activating factors. After highly virulent infection, platelet aggrega-
1 are known to stimulate a procoagulant state of the endothelium tion and viscous metamorphosis was observed that coincided also
and finally activate the coagulation cascade. with endothelial lesions. In contrast, these changes were much less
prevalent in animals infected with the moderately virulent ASFV
4.3. Impairment of platelets and coagulation changes contribute strain. Virus positive platelets that may contribute to the spreading
to evolution of a haemorrhagic syndrome of the virus were observed in the final phase of the disease.
In pigs surviving the first weeks of infection, degranulation of
African swine fever virus infection is accompanied by antibody-coated leukocytes led to secondary platelet aggregation
thrombocytopenia. This could either be due to impairment of and vasoactive amine release facilitating immune-complex depo-
thrombocytopoiesis or peripheral consumption. sition, e.g. in glomeruli (Slauson and Sanchez-Vizcaino, 1981).
In order to investigate the first option, studies were undertaken Another factor of haemostasis is the coagulation system. Infec-
to explore the changes in bone marrow. Here, infection of 25–30% tion with virulent ASFV isolates leads several coagulation changes
of bone marrow megakaryocytes could be shown from six days post that can be measured through clotting times. In detail, ASFV infec-
infection with the moderately virulent E75 strain (Rodriguez et al., tion leads to a prolongation of the activated partial thromboplastin
1996a, 1996b). This finding was consistent with the appearance of time (aPTT), prothrombin time (PT), and thrombin clotting time
a marked thrombocytopenia at that time. Thus, impairment and (TCT) from 4 days post infection onwards. These findings are accom-
destruction of platelet precursors could play an important role in panied by the above mentioned acute thrombocytopenia and an
the genesis of thrombocytopenia. Infection of peripheral platelets, increase in factor VIII-related antigen (Edwards et al., 1984). In
however, was rare. An explanation for this could be that infected addition, Villeda et al. (1993b) could show that apart from pro-
megakaryocytes are destroyed completely in the bone marrow longation of aPTT and PT, reduced levels of coagulation factors are
compartment and do not further maturate into platelets. present in plasma upon highly virulent ASFV infection. This could be
In contrast, Perez et al. (1997) could only demonstrate a much either attributed to excess of consumption (disseminated intravas-
lower percentage of infected megakaryocytes both upon infection cular coagulation, DIC) or decreased production. The latter is less
with a highly virulent (Malawi ’83) and a moderately virulent (DR probable as almost normal liver enzyme activity was observed in
’78) ASFV isolate. After infection with the highly virulent isolate, a the course of the underlying infection. While the aPTT is a marker
peak of 9.5% positive cells was reached at day seven and only little for the intrinsic coagulation pathway that is often affected in the
damage was observed. Upon infection with the moderately virulent course of DIC syndromes, prolonged PT-values are indicative for an
strain, less than 1% of megakaryocytes was infected but severe dam- involvement of the extrinsic coagulation pathway. Taken together,
age was seen together with markedly decreased numbers. These the observed increases are indicative for a more global activation
findings are quite in line with studies conducted by Edwards et al. of the coagulation system.
(1985) who found that 2–10% of the megakaryocytes were infected Moreover, activation of the fibrinolytic system was observed
with an ASFV strain of moderate virulence. Furthermore, Edwards (Villeda et al., 1993b). Activation of the coagulation system can
and Dodds (1985) could show in functional studies, i.e. platelet be achieved by expression of tissue factor (coagulation factor III)
survival times using cohort labelling with [75 Se] selenmethion- by endothelial cells. This phenomenon is observed at least in vitro
ine, that despite obvious thrombocytopenia, thrombocytopoiesis upon ASFV infection (Vallee et al., 2001) and could be explained
128 S. Blome et al. / Virus Research 173 (2013) 122–130

by procoagulant mediators released by infected macrophages. In uninfected lymphocytes, (3) activation of endothelial cells and the
the respective experiments in porcine aortic endothelial cells, coagulation system, and (4) impairment of innate immune func-
these findings were accompanied by inhibition of inflammatory tions. Despite these parallels and the possibility to conclude from
responses. Another possibility is the impairment of degradation of research in these related fields, neither virulence factors nor disease
activated coagulation factors and impaired production of coagula- mechanisms are fully understood. Gaps are especially related to the
tion factors in the liver. While the former seems to be among the role of host factors, the differences of ASFV strains concerning their
options, the impact of disturbed production seems rather moder- virulence, mechanisms and implications of chronic disease courses,
ate as only few hepatocytes are infected and haemorrhages are seen and the role of ASF-specific immunity for pathogenesis. The under-
much earlier than infection of non-macrophage cells. standing of these major points might be the key element for the
Concluding, there is evidence for a DIC as global coagulation development of a first efficacious ASF vaccine.
tests show prolonged clotting times, decreased levels of fibrino-
gen are observed, and consumption of coagulation factors can be
References
seen along with reduced levels of plasminogen (Villeda et al., 1993a,
1993b). Afonso, C.L., Neilan, J.G., Kutish, G.F., Rock, D.L., 1996. An African swine fever virus
Bc1-2 homolog, 5-HL, suppresses apoptotic cell death. Journal of Virology 70 (7),
4858–4863.
4.4. Additional cytokine and acute phase responses link Afonso, C.L., Piccone, M.E., Zaffuto, K.M., Neilan, J., Kutish, G.F., Lu, Z., Balinsky, C.A.,
pathogenesis and immune response Gibb, T.R., Bean, T.J., Zsak, L., Rock, D.L., 2004. African swine fever virus multigene
family 360 and 530 genes affect host interferon response. Journal of Virology 78
(4), 1858–1864.
Different studies on host range and virulence of ASFV demon- Basta, S., Knoetig, S.M., Spagnuolo-Weaver, M., Allan, G., McCullough, K.C., 1999.
strated that IFN response and its control by viral gene products Modulation of monocytic cell activity and virus susceptibility during differenti-
ation into macrophages. Journal of Immunology 162 (7), 3961–3969.
play a major role in determining the virulence of ASFV isolates Blome, S., Gabriel, C., Dietze, K., Breithaupt, A., Beer, M., 2012. High virulence of
(Afonso et al., 2004). Attenuated ASFV strains that lack the host African swine fever virus caucasus isolate in European wild boars of all ages.
range determinant have been shown to induce myxovirus resis- Emerging Infectious Diseases 18 (4), 708.
Brun, A., Rivas, C., Esteban, M., Escribano, J.M., Alonso, C., 1996. African swine fever
tance (Mx) genes that are known to mediate the antiviral response
virus gene A179L, a viral homologue of bcl-2, protects cells from programmed
to several RNA viruses. Recently, it was shown that Mx gene prod- cell death. Virology 225 (1), 227–230.
ucts are also able to inhibit the replication of ASFV in vitro and may Canals, A., Dominquez, J., Tomillo, J., Babin, M., Alonso, F., 1995. Inhibition of IL-2R
and SLA class II expression on stimulated lymphocytes by a suppressor activity
thus also mediate the antiviral state towards ASFV (Netherton et al.,
found in homogenates of African swine fever virus infected cultures. Archives
2009). of Virology 140 (6), 1075–1085.
An acute phase response has been observed upon highly virulent Carpintero, R., Alonso, C., Pineiro, M., Iturralde, M., Andres, M., Le Potier, M.F., Madec,
ASFV infection as evidenced by increased serum concentrations of F., Alava, M.A., Pineiro, A., Lampreave, F., 2007. Pig major acute-phase protein and
apolipoprotein A-I responses correlate with the clinical course of experimentally
C-reactive protein (CRP), serum amyloid A (SAA), and haptoglobin induced African Swine Fever and Aujeszky’s disease. Veterinary Research 38 (5),
starting from day 3 post infection (Sanchez-Cordon et al., 2007). 741–753.
Acute phase proteins are mainly synthesized by hepatocytes upon Carrasco, L., Bautista, M.J., Gomez-Villamandos, J.C., Martin de las Mulas, J., Cha-
con, M.d.L.F., Wilkinson, P.J., Sierra, M.A., 1997a. Development of microscopic
cytokine stimuli, especially proinflammatory mediators such as lesions in splenic cords of pigs infected with African swine fever virus. Veterinary
TNF-␣, IL-1, and IL-6. Activated Kupffer cells can lead to induction Research 28 (1), 93–99.
of an acute phase response. In turn, Kupffer cells can be activated Carrasco, L., Chacon, M.d.L.F., Martin de Las Mulas, J., Gomez-Villamandos, J.C., Sierra,
M.A., Villeda, C.J., Wilkinson, P.J., 1997b. Ultrastructural changes related to the
by proinflammatory cytokines such as TNF-␣ or IL-1 (Gabay and lymph node haemorrhages in acute African swine fever. Research in Veterinary
Kushner, 1999). In the study mentioned above, high serum concen- Science 62 (3), 199–204.
trations of SAA and CRP coincided with high serum concentrations Carrasco, L., de Lara, F.C., Gomez-Villamandos, J.C., Bautista, M.J., Villeda, C.J., Wilkin-
son, P.J., Sierra, M.A., 1996a. The pathogenic role of pulmonary intravascular
of IL-1 and expression of IL-1 and TNF-␣ by Kupffer cells. The high-
macrophages in acute African swine fever. Research in Veterinary Science 61
est haptoglobin concentration coincides with the largest number of (3), 193–198.
IL-6-secreting hepatic macrophages. No impairment of this unspe- Carrasco, L., de Lara, F.C., Martin de las Mulas, J., Gomez-Villamandos, J.C., Hervas, J.,
Wilkinson, P.J., Sierra, M.A., 1996b. Virus association with lymphocytes in acute
cific immune response could be observed.
African swine fever. Veterinary Research 27 (3), 305–312.
In an additional study, Carpintero et al. (2007) demonstrated Carrasco, L., de Lara, F.C., Martin de las Mulas, J., Gomez-Villamandos, J.C., Perez, J.,
high levels of pig major acute phase protein and a decrease Wilkinson, P.J., Sierra, M.A., 1996c. Apoptosis in lymph nodes in acute African
of apolipoprotein A-I is seen upon ASFV infection. In this case, swine fever. Journal of Comparative Pathology 115 (4), 415–428.
Carrasco, L., Fernandez, A., Gomez Villamandos, J.C., Mozos, E., Mendez, A., Jover,
haptoglobin levels were significantly increased, but only minor dif- A., 1992. Kupffer cells and PIMs in acute experimental African swine fever.
ferences were seen for C-reactive protein. Histology and Histopathology 7 (3), 421–425.
In addition, it was recently shown that some serological Carrasco, L., Gomez-Villamandos, J.C., Bautista, M.J., Martin de las Mulas, J., Villeda,
C.J., Wilkinson, P.J., Sierra, M.A., 1996d. In vivo replication of African swine fever
responses have a direct impact on the pathogenesis: some anti- virus (Malawi ’83) in neutrophils. Veterinary Research 27 (1), 55–62.
bodies show higher titres in animals developing lesions, e.g. Carrasco, L., Nunez, A., Salguero, F.J., Diaz San Segundo, F., Sanchez-Cordon, P.,
antibodies against the K196R/thymidine kinase, while an effective Gomez-Villamandos, J.C., Sierra, M.A., 2002. African swine fever: Expression of
interleukin-1 alpha and tumour necrosis factor-alpha by pulmonary intravas-
immune response is accompanied by antibodies raised against the cular macrophages. Journal of Comparative Pathology 126 (2–3), 194–201.
A104R/histone like protein (Reis et al., 2007). Moreover, recovery of Chacon, M.R., Almazan, F., Nogal, M.L., Vinuela, E., Rodriguez, J.F., 1995. The African
swine leucocyte antigen expression during ASFV infection is indica- swine fever virus IAP homolog is a late structural polypeptide. Virology 214 (2),
670–674.
tive for an effective immune response and influences viral clearance
Chamorro, S., Revilla, C., Alvarez, B., Alonso, F., Ezquerra, A., Dominguez, J., 2005.
(replacement/survival of macrophages in the spleen) (Gonzalez- Phenotypic and functional heterogeneity of porcine blood monocytes and its
Juarrero et al., 1992a). relation with maturation. Immunology 114 (1), 63–71.
Detray, D.E., Scott, G.R., 1957. Blood changes in swine with African swine fever.
American Journal of Veterinary Research 18 (68), 484–490.
Edwards, J.F., Dodds, W.J., 1985. Platelet and fibrinogen kinetics in healthy and
5. Résumé and gap analysis
African swine fever-affected swine: [75 Se]selenomethionine-labeling study.
American Journal of Veterinary Research 46 (1), 181–184.
The pathogenesis of ASF shares several features with viral Edwards, J.F., Dodds, W.J., Slauson, D.O., 1984. Coagulation changes in African
haemorrhagic fevers of humans and animals. Among these are (1) swine fever virus infection. American Journal of Veterinary Research 45 (11),
2414–2420.
primary replication in cells of the monocyte/macrophage lineage, Edwards, J.F., Dodds, W.J., Slauson, D.O., 1985. Megakaryocytic infection and throm-
(2) occurrence of cytokine-mediated lesions including apoptosis of bocytopenia in African swine fever. Veterinary Pathology 22 (2), 171–176.
S. Blome et al. / Virus Research 173 (2013) 122–130 129

Ekue, N.F., Wilkinson, P.J., Wardley, R.C., 1989. Infection of pigs with the Cameroon and low virulence: immunohistochemical and ultrastructural studies. Journal
isolate (Cam/82) of African swine fever virus. Journal of Comparative Pathology of Veterinary Diagnostic Investigation 7 (1), 23–30.
100 (2), 145–154. Greig, A., 1972. Pathogenesis of African swine fever in pigs naturally exposed to the
European Food Safety Authority (EFSA), 2009. Scientific report on African Swine disease. Journal of Comparative Pathology 82 (1), 73–79.
Fever submitted to EFSA prepared by Sánchez-Vizcaíno, J.M., Martínez-López, Greig, A., Plowright, W., 1970. The excretion of two virulent strains of African swine
B., Martínez-Avilés, M., Martins, C., Boinas, F., Vial, L., Michaud, V., Jori, F., Etter, fever virus by domestic pigs. Journal of Hygiene 68 (4), 673–682.
E., Albina, E. and Roger, F., 1-141. Haresnape, J.M., Wilkinson, P.J., Mellor, P.S., 1988. Isolation of African swine fever
Fernandez, A., Perez, J., Carrasco, L., Bautista, M.J., Sanchez-Vizcaino, J.M., Sierra, M.A., virus from ticks of the Ornithodoros moubata complex (Ixodoidea: Argasidae)
1992a. Distribution of ASFV antigens in pig tissues experimentally infected with collected within the African swine fever enzootic area of Malawi. Epidemiology
two different Spanish virus isolates. Zentralblatt fur Veterinarmedizinreine B 39 and Infection 101 (1), 173–185.
(6), 393–402. Hernaez, B., Diaz-Gil, G., Garcia-Gallo, M., Ignacio Quetglas, J., Rodriguez-Crespo,
Fernandez, A., Perez, J., Carrasco, L., Sierra, M.A., Sanchez-Vizcaino, M., Jover, A., I., Dixon, L., Escribano, J.M., Alonso, C., 2004a. The African swine fever virus
1992b. Detection of African swine fever viral antigens in paraffin-embedded tis- dynein-binding protein p54 induces infected cell apoptosis. FEBS Letters 569
sues by use of immunohistologic methods and polyclonal antibodies. American (1-3), 224–228.
Journal of Veterinary Research 53 (8), 1462–1467. Hernaez, B., Escribano, J.M., Alonso, C., 2004b. Switching on and off the cell death
Fernandez, A., Perez, J., Martin de las Mulas, J., Carrasco, L., Dominguez, J., Sierra, M.A., cascade: African swine fever virus apoptosis regulation. Progress in Molecular
1992c. Localization of African swine fever viral antigen, swine IgM, IgG and C1q and Subcellular Biology 36, 57–69.
in lung and liver tissues of experimentally infected pigs. Journal of Comparative Hervas, J., Gomez-Villamandos, J.C., Mendez, A., Carrasco, L., Perez, J., Wilkinson, P.J.,
Pathology 107 (1), 81–90. Sierra, M.A., 1996a. Structural and ultrastructural study of glomerular changes
Fernandez de Marco, M., Salguero, F.J., Bautista, M.J., Nunez, A., Sanchez-Cordon, P.J., in African swine fever. Journal of Comparative Pathology 115 (1), 61–75.
Gomez-Villamandos, J.C., 2007. An immunohistochemical study of the tonsils Hervas, J., Gomez-Villamandos, J.C., Mendez, A., Carrasco, L., Sierra, M.A., 1996b.
in pigs with acute African swine fever virus infection. Research in Veterinary The lesional changes and pathogenesis in the kidney in African swine fever.
Science 83 (2), 198–203. Veterinary Research Communications 20 (3), 285–299.
Gabay, C., Kushner, I., 1999. Acute-phase proteins and other systemic responses to Heuschele, W.P., 1967. Studies on the pathogenesis of African swine fever. I. Quan-
inflammation. New England Journal of Medicine 340 (6), 448–454. titative studies on the sequential development of virus in pig tissues. Archiv fur
Gabriel, C., Blome, S., Malogolovkin, A., Parilov, S., Kolbasov, D., Teifke, J.P., Beer, M., die gesamte Virusforschung 21 (3), 349–356.
2011. Characterization of african Swine Fever virus caucasus isolate in European Hurtado, C., Granja, A.G., Bustos, M.J., Nogal, M.L., Gonzalez de Buitrago, G., de
wild boars. Emerging Infectious Diseases 17 (12), 2342–2345. Yebenes, V.G., Salas, M.L., Revilla, Y., Carrascosa, A.L., 2004. The C-type lectin
Galindo, I., Hernaez, B., Diaz-Gil, G., Escribano, J.M., Alonso, C., 2008. A179L, a viral homologue gene (EP153R) of African swine fever virus inhibits apoptosis both
Bcl-2 homologue, targets the core Bcl-2 apoptotic machinery and its upstream in virus infection and in heterologous expression. Virology 326 (1), 160–170.
BH3 activators with selective binding restrictions for Bid and Noxa. Virology 375 Kleiboeker, S.B., 2002. Swine fever: classical swine fever and African swine fever.
(2), 561–572. The Veterinary clinics of North America. Food Animal Practice 18 (3), 431–451.
Gil, S., Sepulveda, N., Albina, E., Leitao, A., Martins, C., 2008. The low-virulent African Kleiboeker, S.B., Scoles, G.A., 2001. Pathogenesis of African swine fever virus in
swine fever virus (ASFV/NH/P68) induces enhanced expression and production Ornithodoros ticks. Animal Health Research Reviews/Conference of Research
of relevant regulatory cytokines (IFNalpha, TNFalpha and IL12p40) on porcine Workers in Animal Diseases 2 (2), 121–128.
macrophages in comparison to the highly virulent ASFV/L60. Archives of Virol- Knudsen, R.C., Genovesi, E.V., 1987. In vivo and in vitro effects of moderately vir-
ogy 153 (10), 1845–1854. ulent African swine fever virus on mitogenesis of pig lymphocytes. Veterinary
Gomez-Villamandos, J.C., Bautista, M.J., Carrasco, L., Caballero, M.J., Hervas, J., Villeda, Immunology and Immunopathology 15 (4), 323–336.
C.J., Wilkinson, P.J., Sierra, M.A., 1997a. African swine fever virus infection of bone Lange, A., Blome, S., Moennig, V., Greiser-Wilke, I., 2011. Pathogenesis of classi-
marrow: lesions and pathogenesis. Veterinary Pathology 34 (2), 97–107. cal swine fever–similarities to viral haemorrhagic fevers: a review. Berl Munch
Gomez-Villamandos, J.C., Bautista, M.J., Hervas, J., Carrasco, L., de Lara, F.C., Perez, Tierarztl Wochenschr 124 (1-2), 36–47.
J., Wilkinson, P.J., Sierra, M.A., 1996. Subcellular changes in platelets in acute Malmquist, W.A., Hay, D., 1960. Hemadsorption and cytopathic effect produced
and subacute African swine fever. Journal of Comparative Pathology 115 (4), by African Swine Fever virus in swine bone marrow and buffy coat cultures.
327–341. American Journal of Veterinary Research 21, 104–108.
Gomez-Villamandos, J.C., Carrasco, L., Bautista, M.J., Sierra, M.A., Quezada, M., Her- Maurer, F.D., Griesemer, R.A., 1958. The pathology of African swine fever; a com-
vas, J., Chacon Mde, L., Ruiz-Villamor, E., Salguero, F.J., Sonchez-Cordon, P.J., parison with hog cholera. American Journal of Veterinary Research 19 (72),
Romanini, S., Nunez, A., Mekonen, T., Mendez, A., Jover, A., 2003. African swine 517–539.
fever and classical swine fever: a review of the pathogenesis. Deutsche Tier- McVicar, J.W., Mebus, C.A., Becker, H.N., Belden, R.C., Gibbs, E.P., 1981. Induced
arztliche Wochenschrift 110 (4), 165–169. African swine fever in feral pigs. Journal of the American Veterinary Medical
Gomez-Villamandos, J.C., Hervas, J., Mendez, A., Carrasco, L., Martin de las Mulas, Association 179 (5), 441–446.
J., Villeda, C.J., Wilkinson, P.J., Sierra, M.A., 1995a. Experimental African swine Mebus, C.A., 1987. Pathobiology and pathogenesis. In: Becker, Y. (Ed.), African Swine
fever: apoptosis of lymphocytes and virus replication in other cells. Journal of Fever. Martinus Nijhoff, Boston, pp. 21–30.
General Virology 76 (Pt 9), 2399–2405. Mebus, C.A., 1988. African swine fever. Advances in Virus Research 35, 251–269.
Gomez-Villamandos, J.C., Hervas, J., Mendez, A., Carrasco, L., Villeda, C.J., Sierra, M.A., Murtaugh, M.P., Baarsch, M.J., Zhou, Y., Scamurra, R.W., Lin, G., 1996. Inflam-
Wilkonson, P.J., 1995b. A pathological study of the perisinusoidal unit of the liver matory cytokines in animal health and disease. Veterinary Immunology and
in acute African swine fever. Research in Veterinary Science 59 (2), 146–151. Immunopathology 54 (1-4), 45–55.
Gomez-Villamandos, J.C., Hervas, J., Mendez, A., Carrasco, L., Villeda, C.J., Wilkinson, Netherton, C.L., Simpson, J., Haller, O., Wileman, T.E., Takamatsu, H.H., Monaghan, P.,
P.J., Sierra, M.A., 1995c. Pathological changes in the renal interstitial capillaries Taylor, G., 2009. Inhibition of a large double-stranded DNA virus by MxA protein.
of pigs inoculated with two different strains of African swine fever virus. Journal Journal of Virology 83 (5), 2310–2320.
of Comparative Pathology 112 (3), 283–298. Nogal, M.L., Gonzalez de Buitrago, G., Rodriguez, C., Cubelos, B., Carrascosa, A.L., Salas,
Gomez-Villamandos, J.C., Hervas, J., Mendez, A., Carrasco, L., Villeda, C.J., Wilkinson, M.L., Revilla, Y., 2001. African swine fever virus IAP homologue inhibits caspase
P.J., Sierra, M.A., 1995d. Ultrastructural study of the renal tubular system in acute activation and promotes cell survival in mammalian cells. Journal of Virology 75
experimental African swine fever: virus replication in glomerular mesangial (6), 2535–2543.
cells and in the collecting ducts. Archives of Virology 140 (3), 581–589. Oura, C.A., Powell, P.P., Parkhouse, R.M., 1998a. African swine fever: a disease char-
Gomez-Villamandos, J.C., Hervas, J., Moreno, C., Carrasco, L., Bautista, M.J., Caballero, acterized by apoptosis. Journal of General Virology 79 (Pt 6), 1427–1438.
J.M., Wilkinson, P.J., Sierra, M.A., 1997b. Subcellular changes in the tonsils of Oura, C.A., Powell, P.P., Parkhouse, R.M., 1998b. Detection of African swine fever
pigs infected with acute African swine fever virus. Veterinary Research 28 (2), virus in infected pig tissues by immunocytochemistry and in sity hybridisation.
179–189. Journal of Virological Methods 72 (2), 205–217.
Gomez del Moral, M., Ortuno, E., Fernandez-Zapatero, P., Alonso, F., Alonso, C., Penrith, M.L., Thomson, G.R., Bastos, A.D., Phiri, O.C., Lubisi, B.A., Du Plessis, E.C.,
Ezquerra, A., Dominguez, J., 1999. African swine fever virus infection induces Macome, F., Pinto, F., Botha, B., Esterhuysen, J., 2004a. An investigation into nat-
tumor necrosis factor alpha production: implications in pathogenesis. Journal ural resistance to African swine fever in domestic pigs from an endemic area in
of Virology 73 (3), 2173–2180. southern Africa. Revue Scientifique et Technique 23 (3), 965–977.
Gonzalez-Juarrero, M., Lunney, J.K., Sanchez-Vizcaino, J.M., Mebus, C., 1992a. Mod- Penrith, M.L., Thomson, G.R., Bastos, A.D.S., 2004b. African swine fever. In: Coet-
ulation of splenic macrophages, and swine leukocyte antigen (SLA) and viral zer, J.A.W., Tustin, R.C. (Eds.), Infectious Diseases of Livestock with Special
antigen expression following African swine fever virus (ASFV) inoculation. Reference to Southern Africa. , 2nd ed. Oxford University Press, Cape Town,
Archives of Virology 123 (1-2), 145–156. pp. 1087–1119.
Gonzalez-Juarrero, M., Mebus, C.A., Pan, R., Revilla, Y., Alonso, J.M., Lunney, Penrith, M.L., Vosloo, W., 2009. Review of African swine fever: transmission, spread
J.K., 1992b. Swine leukocyte antigen and macrophage marker expression on and control. Journal of the South African Veterinary Association 80 (2), 58–62.
both African swine fever virus-infected and non-infected primary porcine Perez, J., Bautista, M.J., Rodriguez, F., Wilkinson, P.J., Sierra, M.A., Martin de las Mulas,
macrophage cultures. Veterinary Immunology and Immunopathology 32 (3-4), J., 1997. Double-labelling immunohistochemical study of megakaryocytes in
243–259. African swine fever. Veterinary Record 141 (15), 386–390.
Gonzalez, S., Mendoza, C., Sanchez-Vizcaino, J.M., Alonso, F., 1990. Inhibitory effect of Perez, J., Fernandez, A.I., Sierra, M.A., Herraez, P., Fernandez, A., Martin de las Mulas,
African swine fever virus on lectin-dependent swine lymphocyte proliferation. J., 1998. Serological and immunohistochemical study of African swine fever in
Veterinary Immunology and Immunopathology 26 (1), 71–80. wild boar in Spain. Veterinary Record 143 (5), 136–139.
Gregg, D.A., Mebus, C.A., Schlafer, D.H., 1995. Early infection of interdigitating den- Perez, J., Rodriguez, F., Fernandez, A., Martin de las Mulas, J., Gomez-Villamandos, J.C.,
dritic cells in the pig lymph node with African swine fever viruses of high Sierra, M.A., 1994. Detection of African swine fever virus protein VP73 in tissues
130 S. Blome et al. / Virus Research 173 (2013) 122–130

of experimentally and naturally infected pigs. Journal of Veterinary Diagnostic Sanchez-Cordon, P.J., Romero-Trevejo, J.L., Pedrera, M., Sanchez-Vizcaino, J.M.,
Investigation 6 (3), 360–365. Bautista, M.J., Gomez-Villamandos, J.C., 2008. Role of hepatic macrophages dur-
Plowright, W., Thomson, G.R., Neser, J.A., 1994. African swine fever. In: Coetzer, ing the viral haemorrhagic fever induced by African Swine Fever Virus. Histology
J.A.W., Thomson, G.R., Tustin, R.C. (Eds.), Infectious Diseases of Livestock with and Histopathology 23 (6), 683–691.
Special Reference to Southern Africa. Cape Town Oxford University Press, pp. Sanchez-Torres, C., Gomez-Puertas, P., Gomez-del-Moral, M., Alonso, F., Escrib-
567–599. ano, J.M., Ezquerra, A., Dominguez, J., 2003. Expression of porcine CD163 on
Portugal, R., Leitao, A., Martins, C., 2009. Characterization of African swine fever monocytes/macrophages correlates with permissiveness to African swine fever
virus IAP homologue expression in porcine macrophages infected with different infection. Archives of Virology 148 (12), 2307–2323.
virulence isolates. Veterinary Microbiology 139 (1-2), 140–146. Sanchez, E.G., Quintas, A., Perez-Nunez, D., Nogal, M., Barroso, S., Carrascosa, A.L.,
Quintero, J.C., Wesley, R.D., Whyard, T.C., Gregg, D., Mebus, C.A., 1986. In vitro and Revilla, Y., 2012. African swine fever virus uses macropinocytosis to enter host
in vivo association of African swine fever virus with swine erythrocytes. Amer- cells. PLoS Pathogens 8 (6), e1002754.
ican Journal of Veterinary Research 47 (5), 1125–1131. Sierra, M.A., Carrasco, L., Gomez-Villamandos, J.C., Martin de las Mulas, J., Mendez, A.,
Ramiro-Ibanez, F., Escribano, J.M., Alonso, C., 1995. Application of a monoclonal anti- Jover, A., 1990. Pulmonary intravascular macrophages in lungs of pigs inoculated
body recognizing protein p30 to detect African swine fever virus-infected cells with African swine fever virus of differing virulence. Journal of Comparative
in peripheral blood. Journal of Virological Methods 55 (3), 339–345. Pathology 102 (3), 323–334.
Ramiro-Ibanez, F., Ortega, A., Brun, A., Escribano, J.M., Alonso, C., 1996. Apoptosis: a Sierra, M.A., Gomez-Villamandos, J.C., Carrasco, L., Fernandez, A., Mozos, E., Jover, A.,
mechanism of cell killing and lymphoid organ impairment during acute African 1991. In vivo study of hemadsorption in African swine fever virus infected cells.
swine fever virus infection. Journal of General Virology 77 (Pt 9), 2209–2219. Veterinary Pathology 28 (2), 178–181.
Ramiro-Ibanez, F., Ortega, A., Ruiz-Gonzalvo, F., Escribano, J.M., Alonso, C., 1997. Sierra, M.A., Quezada, M., Fernandez, A., Carrasco, L., Gomez-Villamandos, J.C., Mar-
Modulation of immune cell populations and activation markers in the patho- tin de las Mulas, J., Sanchez-Vizcaino, J.M., 1989. Experimental African swine
genesis of African swine fever virus infection. Virus Research 47 (1), 31–40. fever: evidence of the virus in interstitial tissues of the kidney. Veterinary Pathol-
Reis, A.L., Parkhouse, R.M., Penedos, A.R., Martins, C., Leitao, A., 2007. Systematic ogy 26 (2), 173–176.
analysis of longitudinal serological responses of pigs infected experimentally Slauson, D.O., Sanchez-Vizcaino, J.M., 1981. Leukocyte-dependent platelet vasoac-
with African swine fever virus. Journal of General Virology 88 (Pt 9), 2426–2434. tive amine release and immune complex deposition in African swine fever.
Revilla, Y., Cebrian, A., Baixeras, E., Martinez, C., Vinuela, E., Salas, M.L., 1997. Inhibi- Veterinary Pathology 18 (6), 813–826.
tion of apoptosis by the African swine fever virus Bcl-2 homologue: role of the Takamatsu, H., Martins, C., Escribano, J.M., Alonso, C., Dixon, L.K., Salas, M.L., Revilla,
BH1 domain. Virology 228 (2), 400–404. Y., 2011. Asfarviridae. In: King, A.M.Q., Adams, M.J., Carstens, E.B., Lefkowitz, E.J.
Rodriguez, C.I., Nogal, M.L., Carrascosa, A.L., Salas, M.L., Fresno, M., Revilla, Y., 2002. (Eds.), Virus Taxonomy. Ninth Report of the ICTV. Elsevier, Oxford, pp. 153–162.
African swine fever virus IAP-like protein induces the activation of nuclear factor Vallee, I., Tait, S.W., Powell, P.P., 2001. African swine fever virus infection of porcine
kappa B. Journal of Virology 76 (8), 3936–3942. aortic endothelial cells leads to inhibition of inflammatory responses, activation
Rodriguez, F., Fernandez, A., Martin de las Mulas, J.P., Sierra, M.A., Jover, A., 1996a. of the thrombotic state, and apoptosis. Journal of Virology 75 (21), 10372–10382.
African swine fever: morphopathology of a viral haemorrhagic disease. Veteri- Villeda, C.J., Gomez-Villamandos, J.C., Williams, S.M., Hervas, J., Wilkinson, P.J., Vin-
nary Record 139 (11), 249–254. uela, E., 1995. The role of fibrinolysis in the pathogenesis of the haemorrhagic
Rodriguez, F., Martin de las Mulas, J., Herraez, P., Sanchez Vizcaino, J.M., Fer- syndrome produced by virulent isolates of African swine fever virus. Thrombosis
nandez, A., 1996b. Immunohistopathological study of African swine fever and Haemostasis 73 (1), 112–117.
(strain E-75)-infected bone marrow. Journal of Comparative Pathology 114 (4), Villeda, C.J., Williams, S.M., Wilkinson, P.J., Vinuela, E., 1993a. Consumption coagu-
399–406. lopathy associated with shock in acute African swine fever. Archives of Virology
Salguero, F.J., Ruiz-Villamor, E., Bautista, M.J., Sanchez-Cordon, P.J., Carrasco, L., 133 (3-4), 467–475.
Gomez-Villamandos, J.C., 2002. Changes in macrophages in spleen and lymph Villeda, C.J., Williams, S.M., Wilkinson, P.J., Vinuela, E., 1993b. Haemostatic abnor-
nodes during acute African swine fever: expression of cytokines. Veterinary malities in African swine fever a comparison of two virus strains of different
Immunology and Immunopathology 90 (1-2), 11–22. virulence (Dominican Republic ‘78 and Malta ‘78). Archives of Virology 130 (1-2),
Salguero, F.J., Sanchez-Cordon, P.J., Nunez, A., Fernandez de Marco, M., Gomez- 71–83.
Villamandos, J.C., 2005. Proinflammatory cytokines induce lymphocyte Wardley, R.C., 1982. Effect of African swine fever on lymphocyte mitogenesis.
apoptosis in acute African swine fever infection. Journal of Comparative Pathol- Immunology 46 (1), 215–220.
ogy 132 (4), 289–302. Wardley, R.C., Wilkinson, P.J., 1977. The association of African swine fever virus with
Sanchez-Cordon, P.J., Ceron, J.J., Nunez, A., Martinez-Subiela, S., Pedrera, M., Romero- blood components of infected pigs. Archives of Virology 55 (4), 327–334.
Trevejo, J.L., Garrido, M.R., Gomez-Villamandos, J.C., 2007. Serum concentrations Wilkinson, P.J., Wardley, R.C., 1978. The replication of African swine fever virus in
of C-reactive protein, serum amyloid A, and haptoglobin in pigs inoculated with pig endothelial cells. British Veterinary Journal 134 (3), 280–282.
African swine fever or classical swine fever viruses. American Journal of Veteri- Zsak, L., Neilan, J.G., 2002. Regulation of apoptosis in African swine fever virus-
nary Research 68 (7), 772–777. infected macrophages. The Scientific World Journal 2, 1186–1195.
Virus Research 178 (2013) 328–339

Contents lists available at ScienceDirect

Virus Research
journal homepage: www.elsevier.com/locate/virusres

Pathogenesis of highly virulent African swine fever virus in domestic


pigs exposed via intraoropharyngeal, intranasopharyngeal, and
intramuscular inoculation, and by direct contact with infected pigs
Erin B. Howey a,b , Vivian O’Donnell a,c , Helena C. de Carvalho Ferreira a,b ,
Manuel V. Borca a , Jonathan Arzt a,∗
a
Plum Island Animal Disease Center, Foreign Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture,
Greenport, NY 11944, USA
b
Oak Ridge Institute for Science and Education, PIADC Research Participation Program, Oak Ridge, TN 37831, USA
c
Department of Pathobiology and Veterinary Science, University of Connecticut at Storrs, Storrs, CT 06269, USA

a r t i c l e i n f o a b s t r a c t

Article history: To investigate the pathogenesis of African swine fever virus (ASFV), domestic pigs (n = 18) were chal-
Received 9 July 2013 lenged with a range (102 –106 50% hemadsorbing doses (HAD50 )) of the highly virulent ASFV-Malawi
Received in revised form strain by inoculation via the intraoropharyngeal (IOP), intranasopharyngeal (INP), or intramuscular (IM)
13 September 2013
routes. A subsequent contact challenge experiment was performed in which six IOP-inoculated donor
Accepted 13 September 2013
pigs were allowed to have direct contact (DC) with six naïve pigs for exposure times that varied from 24 to
Available online 26 September 2013
72 h. All challenge routes resulted in clinical progression and postmortem lesions similar to those previ-
ously described in experimental and natural infection. The onset of clinical signs occurred between 1 and 7
Keywords:
African swine fever
days post inoculation (dpi) and included pyrexia with variable progression to obtundation, hematochezia,
ASFV melena, moribundity and death with a duration of 4–11 days. Viremia was first detected between 4 and
Transmission 5 dpi in all inoculation groups whereas ASFV shedding from the nasal cavity and tonsil was first detected
Challenge at 3–9 dpi. IM and DC were the most consistent modes of infection, with 12/12 (100%) of pigs challenged
Pathogenesis by these routes becoming infected. Several clinical and virological parameters were significantly differ-
Domestic pig ent between IM and DC groups indicating dissimilarity between these modes of infection. Amongst the
simulated natural routes, INP inoculation resulted in the most consistent progression of disease across
the widest range of doses whilst preserving simulation of natural exposure and therefore may provide a
superior system for pathogenesis and vaccine efficacy investigation.
Published by Elsevier B.V.

1. Introduction Georgia which has subsequently led to spread to Russia, Armenia,


Azerbaijan, Ukraine, and most recently to Belarus (WAHID, 2013),
African swine fever (ASF) is a highly contagious, often fatal, with the range of the virus progressively extending west toward
transboundary disease of domestic and wild suids caused by ASF Europe (Costard et al., 2013).
virus (ASFV) (Tulman et al., 2009). It is the sole member of the In domestic swine, it is generally accepted that under natu-
Asfarviridae family and is a large, double stranded DNA virus with ral conditions, ASFV is primarily transmitted via direct contact
a 170–190 kb genome. Due to the lack of a commercial or experi- with excreted viral particles through nuzzling and/or ingestion
mental vaccine or treatment, large economical losses are associated (Sánchez-Vizcaíno et al., 2012; Wardley et al., 1983). However, the
with ASFV outbreaks (Sánchez-Vizcaíno et al., 2012). Such an event detailed mechanism of this mode of transmission has yet to be
occurred in 2007, when ASFV was introduced into the Republic of explicitly defined. Arthropod-borne transmission via Ornithodoros
spp. ticks, which is a relevant means of transmission of ASFV within
the wild suid population and from wild suids to domestic swine, is
∗ Corresponding author at: Foreign Animal Disease Research Unit, USDA/ARS a minor method of transmission between domestic pigs (Arzt et al.,
Plum Island Animal Disease Center, P.O. Box 848, Greenport, NY 11944, USA. Tel.: 2010; Jori et al., 2013; Penrith, 2009).
+1 631 323 3336; fax: +1 631 323 3006. An extensive portion of in vivo ASF research has utilized the
E-mail addresses: Erin.Howey@ars.usda.gov (E.B. Howey), intramuscular (IM) route of inoculation (Ballester et al., 2010;
Vivian.ODonnell@ars.usda.gov (V. O’Donnell), Helena.Ferreira@ars.usda.gov
(H.C. de Carvalho Ferreira), Manuel.Borca@ars.usda.gov (M.V. Borca),
Carrasco et al., 1996, 1997; Childerstone et al., 1998; Fernández
Jonathan.Arzt@ars.usda.gov (J. Arzt). de Marco et al., 2007; Gómez-Villamandos et al., 1995a, 1995b,

0168-1702/$ – see front matter. Published by Elsevier B.V.


http://dx.doi.org/10.1016/j.virusres.2013.09.024
E.B. Howey et al. / Virus Research 178 (2013) 328–339 329

1997a, 1997b, 1998; Oura et al., 1998; Pan and Hess, 1984; Salguero additional dilution to achieve the desired viral dose in 2 ml of
et al., 2004). This method results in highly reproducible clinical media.
disease. However, since IM inoculation does not result in viral
interaction with the oral and upper respiratory mucosal surfaces, 2.2. Animals
it bypasses many of the innate defense mechanisms the virus
would normally encounter through natural infection. Therefore, Conventional castrated male Yorkshire pigs from a herd that
it may be a suboptimal inoculation method to examine the early is demonstrated to be free of porcine reproductive and respira-
pathogenesis and previremic stages of disease. Similarly, these tory syndrome virus were used for all experiments. Animals were
limitations make IM challenge, an unnatural model for vaccine dewormed and vaccinated for common porcine pathogens includ-
assessment. ing porcine circovirus 2 prior to arrival at our facility. Pigs were
The conventional pathogenesis of ASF has been investigated approximately 3–4 months old and 22–27 kg upon arrival. All
in vivo using simulated natural routes of inoculation including animal procedures were performed following Protocol 225-10-R
intranasal (IN) (de Carvalho Ferreira et al., 2012, 2013a; Greig, approved by the Plum Island Animal Disease Center Institutional
1972; Greig and Plowright, 1970; Heuschele, 1967; Plowright et al., Animal Care and Use Committee (IACUC), which ensured ethi-
1968), intraoral (IO) (Boulanger et al., 1967; Colgrove et al., 1969), cal and humane treatment of experimental animals. The animal
combined oronasal (ON) (Boinas et al., 2004; Hamdy and Dardiri, experiments were carried out in BSL-3 Ag isolation rooms at Plum
1984; McVicar, 1984; Mebus et al., 1978; Mebus and Dardiri, 1979, Island Animal Disease Center. Animals were kept within the hous-
1980) and direct contact/aerosol (DC) (de Carvalho Ferreira et al., ing facilities 7 days prior to the start of the experiments to allow
2013b; Wilkinson and Donaldson, 1977; Wilkinson et al., 1977, for acclimation to the new environment. All animals were fed daily
1981). Within all of the studies noted, the main route of virus and had unlimited access to water.
detection was through virus isolation and was usually performed All animals except for donors in the DC study were allowed to
only on the blood, serum or a limited number of tissues. Few survive until terminal disease. Severely moribund animals were
have focused on virus detection within the secretions (de Carvalho humanely euthanized intravenously with 85.8 mg/kg of sodium
Ferreira et al., 2012; Greig and Plowright, 1970; McVicar, 1984; pentobarbital. Animals without clinical disease were humanely
Wilkinson et al., 1983; Ekue et al., 1989). While these reports have euthanized at the end of the study as described above.
contributed to current understanding of the viral dynamics and
pathogenesis of ASF, they fail to capture some specific information 2.3. Study design
which may contribute to develop the next generation of rationally
designed countermeasures. Specifically, confirmation of the pri- 2.3.1. Direct inoculation studies
mary site(s) of infection and elucidation of virus–host interactions Experiments were performed to compare three routes of direct
during early pathogenesis would facilitate the process of develop- inoculation. Eighteen pigs were used in these experiments. Six pigs
ing and validating effective prophylaxis. Furthermore, variability were assigned to each of the three inoculation route groupings:
in methodologies including virus strain, dose, and route of chal- intramuscular (IM), intranasopharyngeal (INP) and intraoropha-
lenge has confounded comparison across studies. The importance ryngeal (IOP). Inoculation route groupings were further subdivided
of standardizing experimental models including the description into three inoculation dosage groups: low dose 102 HAD50 (n = 2),
and interpretation of clinical and pathologic findings in experi- mid dose 104 HAD50 (n = 2) and high dose 106 HAD50 (n = 2). IM-
mental ASF has recently been emphasized (Galindo-Cardiel et al., inoculated pigs received a 2 mL injection of inoculum within the
2013). The goal of such standardization is to facilitate compari- right semimembranosus muscle. Pigs assigned to INP inoculation
son of studies across research groups for the purpose of advancing were sedated with an intramuscular injection of telazol (3 mg/kg),
ASF vaccine development whilst avoiding redundancy of research ketamine (8 mg/kg), xylazine (4 mg/kg), placed in sternal recum-
activities. bency and 2 mL inoculum was instilled through the nares into the
In the current study, we compared four routes of challenge of nasopharynx via a 15 gauge silicon catheter (Abbott Laboratories,
domestic pigs with ASFV in order to characterize route- and dose- IL). Pigs assigned to IOP inoculation received 2 mL of the inoculum
dependent viral dynamics and the transmission of ASFV via direct perorally via syringe with an attached 6 in. sterile metal cannula.
contact. This was done to determine how different means of expo- Animals were sedated, placed in dorsal recumbency and 2 mL of the
sure/inoculation alter the pathogenesis of ASFV in swine and to inoculum was deposited in the ventral portion of the soft palate and
determine which route most closely simulates natural infection palatine tonsil.
with optimum experimental reproducibility.
2.3.2. Direct contact (DC) transmission studies
2. Materials and methods Additional experiments were performed to characterize ASFV
transmission from IOP-inoculated pigs and viral dynamics in pigs
2.1. Virus challenged by direct contact. Donor (n = 6) and naïve pigs (n = 6)
were initially housed in two separate rooms. Donor pigs were inoc-
The highly pathogenic ASFV isolate Malawi Lil-20/1 was orig- ulated via the intraoropharyngeal method as described above with
inally isolated from Ornithodoros sp. ticks during a field outbreak 2 mL of inoculum at a dose of 105 HAD50 of the same ASFV Malawi
in Malawi in 1983 and obtained from L. Dixon (Institute of Ani- Lil-20/1 described above. Upon detection of pyrexia (rectal tem-
mal Health, Pirbright Laboratory, Woking, Surrey, United Kingdom) perature greater than or equal to 40 ◦ C) in at least 4 donor pigs (6
(Borca et al., 1998). The stock virus was passaged once in primary days post inoculation (dpi)), contact pigs were transferred into the
porcine macrophages derived from heparinized swine blood. The donor room and allowed to comingle with inoculated pigs. Feed
tissue culture supernatant was then stored as 108 50% hemadsorb- was withheld for approximately 4 h to encourage contact between
ing dose (HAD50 ) aliquots at −70 ◦ C. pigs. After 24 h (1 day post contact (dpc)), two contact animals were
The inoculum was prepared by initially diluting stock virus 10- removed and isolated into a separate room and two donor animals
fold in Dulbeco’s Modified Eagle’s Medium (DMEM) (Gibco; Invit- were euthanized to maintain the donor/naïve pig ratio. This pro-
rogen, NY) containing 1% antibiotic/antimycotic (10,000 units/mL cess was repeated at 48 and 72 h (2 and 3 dpc). Contact-exposed
of penicillin, 10,000 ␮g/mL of streptomycin, and 25 ␮g/mL of animals were then monitored for onset and progression of clinical
Fungizone® ) (Gibco; Invitrogen, NY) and subsequently performing disease.
330 E.B. Howey et al. / Virus Research 178 (2013) 328–339

Table 1
Clinical signs and scoring value for ASF.

Characteristic Score

Behavior and mentation 0 Normal, alert, responsive


1 Mildly obtunded. Slightly reduced liveliness, stands up unassisted, resists restraint or rectal thermometer
2 Obtunded. Reluctant to stand but will do so when assisted; decreased resistance to restraint or rectal thermometer
3 Intermittent ataxia, disorientation, can still stand/walk or will not stand/walk even when assisted, still conscious
4 Moribund. Nonambulatory, unconscious/nonresponsive

Neurologic signs 0 Normal


2 Unambiguous neurologic signs (e.g. convulsions, seizures)

Defecation 0 Normal to mildly soft stools


1 Profuse watery diarrhea, ±mild hematochezia or melena
2 Severe to marked hematochezia/melena

Body temperaturea 0 38–40 ◦ C


1 Temperature greater than or equal to 40 ◦ C at any point of study
2 Temperature greater than or equal to 40 ◦ C for at least 2 subsequent days
3 Temperature greater than or equal to 41 ◦ C
4 Temperature less than 38 ◦ C
a
Temperature scores are never reduced.

2.4. Clinical evaluation animals will appear in a separate manuscript (Howey et al., in
progress).
Incidence proportion of ASF was calculated for each dose/route
combination as [(number of successfully infected pigs for that dose 2.6. Virus isolation and titration
and route) ÷ (all pigs subjected to that dose and route)]. For all stud-
ies, clinical evaluations were performed on all animals until death Blood, serum, nasal swabs, tonsil scrapings, urine (postmortem
or termination of the experiment. Clinical signs used to charac- only) and tissues from infected pigs were screened for ASFV
terize progression of ASF are listed in Table 1. These clinical signs by virus isolation and titration (VI) on primary porcine blood
were assigned a numerical value based on severity and significance. macrophage cell cultures prepared from defibrinated swine blood
The sum of the scores of all clinical signs present for each pig was as previously described (Zsak et al., 2005). Presence of virus was
recorded daily. determined by identification of infected cells by rosette formation
(hemadsorption) and titers expressed as doses calculated by the
2.5. Sample collection and processing Spearman–Karber method (Mahy and Kangro, 1996).

Sample collection including whole blood with EDTA, clotted 2.7. Immunomicroscopy
blood for the collection of serum, swab specimens from the nasal
cavity and superficial scrapings from the tonsil of the soft palate Microscopic localization of the virus in tissues positive
were performed on the day of inoculation at 0 days post inocu- for ASFV via VI was performed via immunohistochemistry
lation (dpi) followed by sample collection every other day. Pigs (IHC). Immunofluorescence (IFA) was used to detect virus in
were manually restrained for sample collection. Whole blood and combination with other cellular markers in order to further
serum were collected via the jugular vein. Tonsil scrapings were determine/characterize phenotypes of infected cells. All primary
collected with a sterile spoon by gently scraping along the surface antibodies were extensively tested prior to this study in sections of
of the tonsil of the soft palate, exfoliating superficial epithelial cells tissue from ASFV infected or non-infected pigs, using IHC, to ensure
while making sure to not disrupt the mucosa. Nasal swabs were specificity.
collected bilaterally using sterile cotton swabs extended just cau- OCT-embedded tissue samples for IHC and IFA were cryosec-
dal to the alar fold. Following collection, nasal swabs and tonsil tioned onto electrostatically charged glass slides and fixed for
scrapings were immediately immersed in 1 mL of DMEM with 5% 10 min in acetone at −20 ◦ C. Slides were blocked for 2 h at 20 ◦ C
antibiotic/antimycotic; then, all samples were transferred on ice with Phosphate buffered saline with Tween (PBST) containing 6%
to the laboratory for processing. All samples were transferred to mixed serum and 2% powdered non-fat milk. A mouse monoclonal
cryovials and stored at −70 ◦ C until they were analyzed by virus primary antibody 1D9, targeting the ASFV VP30 (Cuesta-Geijo et al.,
isolation (VI). 2012; Galindo et al., 2012) (kindly provided by Javier Dominguez
Necropsies were performed as soon as possible following Juncal, Departamento de Biotecnología, Instituto Nacional de Inves-
euthanasia or natural death. Tissues collected during postmortem tigación y Tecnología Agraria y Alimentaria (INIA)), was diluted
examination included: tonsil of the soft palate, lingual tonsil, nasal 1:250 in blocking buffer and applied to tissue sections that were
tonsil, dorsal soft palate (rostral and caudal), dorsal nasophar- then incubated for 20 h at 4 ◦ C.
ynx, caudal nasal turbinate, epiglottis, trachea, bronchial mucosa, For IHC, specific anti-ASFV immunoreactivity was detected
lung, thymus, liver, spleen, kidney, adrenal gland, thyroid, pan- using a commercial micropolymer alkaline phosphatase detection
creas, small intestine, large intestine, bone marrow, and lymph system (Mach 3 AP kit; Biocare, CA) as per manufacturers’ recom-
nodes including: retropharyngeal, submandibular, hilar, gastro- mendation with an alkaline phosphatase substrate (Vector Red;
hepatic, renal, inguinal and popliteal. Urine was also taken via direct Vector Laboratories, CA). Slides were counterstained with Gill’s
aspiration from the urinary bladder at the time of necropsy. Tis- hematoxylin and cover slipped using routine methods. A duplicate
sue samples collected were placed in cryovials as described above serial section of each tissue screened was treated with a mouse
and cryomolds, embedded in Optimal Cutting Temperature (OCT) monoclonal anti-foot and mouth disease virus (FMDV) antibody
medium (Tissue-Tek O.C.T. compound, Sakura Finetek, CA) and then (10GA4, targeting FMDV O1-Brugge (Arzt et al., 2009)) serving as a
frozen in liquid nitrogen. Detailed description of tissues from these negative control.
E.B. Howey et al. / Virus Research 178 (2013) 328–339 331

Table 2
Clinical characteristics of pigs infected with ASFV by different routes and dosages.

Group No. infected/total no. Total days survival (±SD) Pyrexia Clinical signs

Days to onset (±SD) Days to onset (±SD) Maximum clinical score (±SD)

IM
Overall 6/6 8.5 (±1.8)c 5.2 (±1.2)c 4.5 (±1.2)c 6.3 (±1.4)c
102 (n = 2) 2/2 10.5 (±0.3) 5.5 (±0.7) 5.5 (±0.7) 7.0 (±1.4)
104 (n = 2) 2/2 8.0 (±1.4) 5.5 (±0.7) 5.0 (±0.0) 6.5 (±2.1)
106 (n = 2) 2/2 7.0 (±0.0) 4.5 (±2.1) 3.0 (±0.0) 5.5 (±0.7)

INP
Overall 5/6 10.3 (±1.9)c 5.0 (±2.7)c 2.6 (±2.5)c 6.2 (±2.8)c
102 (n = 2) 1/2 11.0 (±1.4) 7.0 (±0.0)a 7.0 (±0.0)a 9.0 (±0.0)a
104 (n = 2) 2/2 11.0 (±1.4) 7.0 (±0.0) 1.5 (±0.7) 5.0 (±1.4)
106 (n = 2) 2/2 9.0 (±2.8) 2.0 (±0.0) 1.5 (±0.7) 6.0 (±4.2)

IOP
Overall 4/6 10.5 (±2.4)c 5.8 (±1.3)c 4.3 (±2.4)c 4.8 (±4.0)c
102 (n = 2) 0/2 13.0 (±0.0) – – –
104 (n = 2) 2/2 10.0 (±2.8) 6.5 (±0.7) 3.5 (±3.5) 6.5 (±5.0)
106 (n = 2) 2/2 8.5 (±0.7) 5.0 (±1.4) 5.0 (±1.4) 3.0 (±0.0)

DC
Overall 6/6 10.3 (±1.2)c 5. (±1.8)c 5.0 (±1.9)c 6.8 (±2.0)c
1 dpc (n = 2) 2/2 10.5 (±0.7) 5.0 (±0.0) 4.5 (±0.7) 7.5 (±0.7)
2 dpc (n = 2) 2/2 10.5 (±2.1) 3.5 (±2.1) 3.5 (±2.1) 8.0 (±1.4)
3 dpc (n = 2) 2/2 10.0 (±1.4) 7.0 (±0.0) 7.0 (±0.0) 5.0 (±2.8)

IOP donor
Overall 4/6 –b 3.6 (±0.6) 4.0 (±1.7) 4.0 (±2.5)

–, not detected; SD, standard deviation.


a
One animal affected within group.
b
Survival not calculated due to predetermined euthanasia.
c
No statistical difference between group overall values (IM, INP, IOP and DC).

Multichannel IFA was performed similarly as described for IHC, 2.8.2. Clinical signs, viremia and shedding
except ASFV antigen detection was performed in combination with For all clinical and virological parameters examined, the
primary antibodies with specificity for host cellular markers includ- mean ± standard deviation (SD) for each infection group/dose com-
ing pancytokeratin (180059, Zymed, Invitrogen, NY) for epithelium, bination was calculated using data values collected per individual
CD163 and CD172a for macrophages (MCA 2311, and MCA2312GA, animal per time point. The effect of the inoculation/infection route
respectively AbD Serotec, UK). After incubation of primary antibod- used to establish infection was compared across groups (both
ies for 20 h at 4 ◦ C, isotype-specific secondary antibodies labeled within the direct inoculation studies and between direct inoc-
with fluorescent dyes (AF 350, 488, 594, and 647, AlexaFluor; ulation and contact studies) for the outcome variables present
Molecular Probes Inc., OR) were applied and slides were incubated in Tables 2 and 3. These comparisons were made using regres-
for 1 h at 37 ◦ C. Images were obtained using a Nikon Eclipse 90i sion analysis, using a linear model, considering the 4 types of
multichannel epi-fluorescent microscope equipped with 350, 488, inoculation/infection route as categorical explanatory variables:
594, and 647 excitation filter cubes and digital camera. intramuscular (IM), intranasopharyngeal (INP), intraoropharyngeal
(IOP) and direct contact (DC). A linear model was also used to assess
2.8. Statistical analyses the effects of different exposure levels applied in the pigs infected
by direct contact, with exposure as explanatory variable (with 3
All statistical analyses were performed using R statistical pro- exposure levels – 1 day, 2 days and 3 days) and variables present
gram (R core Team, 2013)), employing different R packages in Tables 2 and 3 as outcome. Linear model assumptions were con-
(explained in detail below). Statistical significance was considered firmed through visual inspection of the residual plots. The statistical
as p < 0.05. All data reported herein is not statistically significant power of these experiments was analyzed retrospectively using the
unless explicitly stated. package “pwr” (Champely, 2012). The study did not have enough
power to test for possible interactions of type of inoculation route
2.8.1. Survival analysis and dose used for inoculation. The power of the analysis using only
Duration of survival for each pig was calculated as the total the explanatory variable type of inoculation/infection route (deter-
number of days from inoculation/exposure to natural death or mining differences between inoculation/infection groups) varied
euthanasia due to moribundity. Survival functions were calculated from 0.58 to 0.31, depending on the number of missing values.
for pigs belonging to the IM, INP, IOP and DC groups and com-
pared through a Cox proportional hazard regression model using 3. Results
the package “Survival” for R software (Therneau, 2013). Multiple
comparisons were made: (1) between inoculation/infection routes 3.1. Direct inoculation studies
(IM, INP, IOP and DC), (2) within inoculated groups (IM, INP and
IOP) comparing type of inoculation route and dose, (3) within each 3.1.1. Inoculation and infectivity
dose (comparing different inoculation routes), (4) within each route Successful infection of individual pigs was determined by the
(comparing doses), and (5) within the DC pigs (comparing expo- presence of characteristic clinical signs combined with detection
sure length). Survival was not calculated for donor pigs (IOP donor of ASFV within the blood components (viremia). In the IM group,
group) from the contact experiment since these pigs were eutha- all animals regardless of dose were successfully infected (2/2 high
nized at predetermined times regardless of clinical disease. dose, 2/2 mid dose, and 2/2 low dose (100% incidence)). In the INP
332 E.B. Howey et al. / Virus Research 178 (2013) 328–339

Table 3
Virologic parameters of pigs infected with ASFV by different routes and dosages.

Group No. infected/ Viremia Shedding


total no.
Days to onset Duration Maximum Days to onset of Maximum Days to onset of Maximum
(±SD) titer tonsil shedding titer (±SD)b nasal shedding titer (±SD)b
(±SD) (±SD)
(±SD) (±SD)b,c,g

IM
Overall 6/6 4.3 (±1.6)e 5.2 (±1.3)e 6.5 (±0.6)c 8.0 (±1.2)e 2.4 (±0.4)e 9.0 (±0.0)a,e 2.1 (±0.0)a,e
102 (n = 2) 2/2 6.0 (±1.4) 5.5 (±0.7) 6.1 (±0.0)c 9.0 (±0.0)a 2.8 (±0.0)a 9.0 (±0.0)a 2.1 (±0.0)a
104 (n = 2) 2/2 4.0 (±1.4) 5.0 (±2.8) 6.4 (±0.9)c 8.0 (±1.4) 2.4 (±0.5) – –
106 (n = 2) 2/2 3.0 (±0.0) 5.0 (±0.0) 6.9 (±0.2)c 7.0 (±0.0)a 2.1 (±0.0)a – –

INP
Overall 5/6 5.4 (±1.1)e 5.6 (±1.7)e 7.0 (±1.2) 7.5 (±1.7)e 3.3 (±1.0)e 7.7 (±3.5)e 4.2 (±1.7)e,f
102 (n = 2) 1/2 5.0 (±0.0)a 6.0 (±0.0)a 5.8 (±0.0)a 9.0 (±0.0)a 2.8 (±0.0)a – –
104 (n = 2) 2/2 5.0 (±1.4) 7.0 (±0.0) 8.3 (±0.0)c 6.0 (±0.0) 4.0 (±0.7) 6.0 (±2.8) 5.2 (±0.5)
106 (n = 2) 2/2 6.0 (±1.4) 4.0 (±1.4) 6.3 (±0.4) 9.0 (±0.0)a 2.3 (±0.0)a 11.0 (±0.0)a 2.3 (±0.0)a

IOP
Overall 4/6 5.0 (±1.2)e 5.3 (±1.7)e 8.5 (±0.1) 5.5 (±1.9)e,f 5.3 (±1.7)f 6.5 (±1.0)e 5.2 (±1.1)f
102 (n = 2) 0/2 – – – – – – –
104 (n = 2) 2/2 6.0 (±0.0) 5.0 (±2.8) 8.6 (±0.0) 7.0 (±1.4) 4.4 (±0.5) 7.0 (±1.4) 4.7 (±0.2)
106 (n = 2) 2/2 4.0 (±0.0) 5.5 (±0.7) 8.4 (±0.2) 4.0 (±0.0) 6.2 (±2.3) 6.0 (±0.0) 5.8 (±1.4)

DC
Overall 6/6 5.2 (±1.5)e 6.2 (±1.7)e 8.8 (±0.3) 3.5 (±2.4)f 5.2 (±0.8)f 7.7 (±0.6)e 4.7 (±0.1)f
1 dpc (n = 2) 2/2 4.0 (±1.4) 7.5 (±2.1) 8.7 (±0.2) 1.5 (±0.7) 4.7 (±0.5) 8.0 (±0.0) 4.6 (±0.4)
2 dpc (n = 2) 2/2 5.0 (±1.4) 6.5 (±0.7) 8.8 (±0.4) 4.0 (±2.8) 5.8 (±0.0) 7.5 (±0.7) 4.7 (±0.2)
3 dpc (n = 2) 2/2 6.5 (±0.7) 4.5 (±0.7) 8.9 (±0.5) 5.0 (±2.8) 5.2 (±1.2) 7.5 (±0.7) 4.8 (±0.0)

IOP donor
Overall 4/6 4.8 (±2.2) –d 7.4 (±3.2) 2.8 (±1.5) 4.1 (±1.2) 6.5 (±1.3) 4.6 (±1.3)

–, not detected; SD, standard deviation.


a
One animal affected within group.
b
Log10 HAD50 /mL.
c
All titers derived from whole blood except ‘c’ below (serum derived).
d
Duration of viremia not calculated due to predetermined euthanasia.
e
Group overall values (IM, INP, IOP and DC) with a common superscript are not significantly different (p > 0.05).
f
Group overall values (IM, INP, IOP and DC) with a common superscript are not significantly different (p > 0.05).
g
No statistical differences were calculated for this group.

and IOP groups, incidence was 100% within the mid (2/2) and high 3.1.3. Clinical signs
dose (2/2) groups; however amongst low dose animals, 1/2 pigs For all route-dose combinations, the most consistent and com-
in the INP group and the entire (0/2) IOP group did not become monly earliest detected clinical sign was pyrexia ranging from 40
infected (low dose incidence = 50%, 0% respectively). to 41.9 ◦ C. Diarrhea, hematochezia and melena were common in
all groups during the late stages of infection, except the contact
3.1.2. Survival experiment donors. Less consistent clinical signs included obtunda-
The IM-inoculated pigs had the shortest overall survival (mean tion, oculonasal discharge, coughing, and dermal hyperemia which
8.5 ± 1.8 dpi, n = 6) (Table 2, Figs. 1A and 2), however, no statisti- were often transient and variable throughout all groups. Termi-
cally significant differences were found between the IM, INP and nal stage ASF was characterized by moribundity including a rapid
IOP groups in the Cox proportional hazard analysis. IM-inoculated and substantial decrease in body temperature, lack of response to
pigs also had the shortest survival following onset of pyrexia (mean stimulus, inability to stand or ambulate, and occasionally, seizure-
4.3 ± 1.9 days, n = 6). Mean survival after pyrexia was longest within like behavior. There were no statistically significant differences
the low dose group (6 ± 0.0 days, n = 2) and equal within the mid (p > 0.05) between the IM, INP, IOP and DC routes for total days of
and high dose groups (3.5 ± 2.1 days, n = 2 per group). Amongst survival, days to onset of pyrexia and clinical signs, and maximum
the INP-inoculated pigs, overall mean survival was 10.3 ± 1.9 dpi clinical score (Table 2).
with 5.5 ± 1.8 days survival following the onset of pyrexia. Sur- Amongst the IM-inoculated pigs, the onset of clinical signs
vival was shortest within the high dose group (9.0 ± 2.8 days, was shortest within the high dose group (Table 2, Fig. 2) includ-
n = 2) compared to the mid and low dose groups (11.0 ± 1.4 days, ing pyrexia (4.5 ± 2.1 days) and obtundation (6.0 ± 0.0 days)
n = 2 per group). Overall survival within the intraoropharyngeal which occurred in all pigs (n = 6). Melena/hematochezia was
group was the longest compared to all other routes with a mean observed in one animal from the mid and high dose groups,
of 10.5 ± 2.4 dpi (n = 6). Survival following onset of pyrexia was with the shortest onset (5.0 ± 0.0 days) within the high dose.
4.5 ± 2.1 days; however, the number of days to onset of pyrexia The overall highest mean clinical score was 6.3 ± 1.4 with the
was the longest (5.8 ± 1.3 dpi). highest mean clinical score observed within the low dose group
The dose used for inoculation had a significant effect on the pig’s (n = 2). The IM-inoculated pigs, had significantly earlier onset of
survival, with pigs inoculated with the low dose (102 HAD50 ASFV) obtundation and onset of melena/hematochezia (p < 0.05) in com-
having a significantly longer survival (p = 0.02) than pigs inoculated parison to every group, or in comparison to the DC and INP routes
with a higher dose (106 HAD50 ASFV). However, when groups were individually.
compared within the same dose (i.e. across route), there were no The lowest overall number of days to the onset of clinical
statistical differences between the different inoculation routes. signs was amongst the INP-inoculated pigs (mean 2.6 ± 2.5 dpi, 5/6
E.B. Howey et al. / Virus Research 178 (2013) 328–339 333

Fig. 1. Descriptive survival analyses for ASFV-exposed pigs relating dose/exposure length to survival duration within the direct inoculation studies (A) and direct contact
study (B).

pigs). However, onset within the low dose group was substantially pigs had significantly different results in comparison with pigs
longer (7 days) than the mid and high dose groups (1.5 ± 0.7 days) infected by DC. Specifically these pigs had longer days to onset
(Fig. 2D–F). The lowest overall number of days to onset of pyrexia of tonsil shedding, and lower maximum ASFV titers in tonsil and
was amongst the INP-inoculated pigs (mean 5.0 ± 2.7 dpi, n = 2) nasal swabs (Table 3).
with the high dose group having the shortest onset (2.0 ± 0.0 days, The overall number of days to onset of viremia was the longest
n = 2) compared to the low (1/2 pigs) and mid dose (2/2 pigs) groups in the INP groups at 5.4 ± 1.1 dpi (5/6 pigs). The number of days of
(7.0 ± 0.0 days). Obtundation and melena/hematochezia occurred viremia averaged 5.6 ± 1.7 days with the high dose group having
in 4 out of 6 pigs with respective means to onset of 9.5 ± 1.0 and the shortest length of viremia (Fig. 2C, Table 3). The overall mean
10.0 ± 0.8 days. The mean maximum clinical score amongst the maximum serum/whole blood titer was 7.0 ± 1.2 log10 HAD50 /mL.
INP-inoculated pigs was 6.2 ±;2.8 with the highest clinical score Tonsillar ASFV was detected in 4 out of 6 INP-inoculated ani-
occurring within the low dose group (9.0 ± 0.0). mals with a mean onset of 7.5 ± 1.7 days and a max titer of
The mean maximum clinical score for the IOP groups was 3.3 ± 1.0 log10 HAD50 /mL. Nasal shedding was detected in 3 out
4.8 ± 4.0 (4/6 pigs) with the lowest clinical score found within of 6 animals with a mean onset of 7.7 ± 3.5 dpi and a max
the high dose group. Clinical signs consistent with ASF were titer of 4.2 ± 1.7 log10 HAD50 /mL. The mean difference between
not observed in pigs from the low dose group. Onset of pyrexia the onset of viremia and onset of ASFV detection in secretions
occurred within the mid and high dose groups at 6.5 ± 0.7 and was 1.5 ± 1.9 days. In comparison to DC-exposed pigs, the INP-
5.0 ± 1.4 days respectively (n = 2 per group). Obtundation and inoculated pigs had significantly longer days to onset of tonsil
melena/hematochezia were detected only in one mid dose IOP- shedding (for INP), and significantly lower maximum titers in
inoculated animal starting at 10 and 11 dpi respectively. tonsil.
Within the IOP groups, overall onset of viremia was detected at
3.1.4. Viremia and ASFV shedding 5.0 ± 1.2 dpi with overall duration of viremia for 5.3 ± 1.7 days for 4
The IM-inoculated pigs (n = 6) had the shortest overall period out of 6 pigs inoculated (Table 3). The high dose group had both the
to onset of viremia (mean 4.3 ± 1.6 dpi) and number of days of shortest onset to viremia and shortest duration. The mean maxi-
viremia (5.2 ± 1.3 days) (Table 3). Amongst the IM-inoculated pigs, mum titer from whole blood was 8.5 ± 0.1 log10 HAD50 /mL. Tonsil
the low dose group had both the longest time before onset and and nasal shedding was detected within all infected animals (4/6)
longest duration of viremia (Fig. 2A, Table 3). The highest mean with a mean onset of shedding at 5.5 ± 1.9 and 6.5 ± 1.0 dpi respec-
titer of ASFV in serum was detected within the high dose group tively. The mean maximum titers for tonsil and nasal swabs for
(6.9 ± 0.2 log10 HAD50 /mL). Onset to detection of ASFV from the IOP-inoculated pigs were 5.3 ± 1.7 and 5.2 ± 1.1 log10 HAD50 /mL.
tonsil was shortest within the high dose group. Nasal shedding The period between onset of viremia and viral shedding was the
was only detected in 1 pig within the low dose IM group which shortest of all routes at 0.5 ± 1.2 days. There were no statistical sig-
occurred at 9 dpi. There was an overall mean of 4.0 ± 1.6 days from nificant differences in any of the outcome variables between the
onset of viremia to detection of ASFV in secretions. IM-inoculated IOP and the DC groups.
334 E.B. Howey et al. / Virus Research 178 (2013) 328–339

Fig. 2. Relationship between clinical score and virus shedding in pigs inoculated IM (A–C), INP (D–F), and IOP (G–I) with ASFV Malawi at 102 , 104 and 106 HAD50 . Lines
indicating viral titer in whole blood, serum, tonsil swabs and nasal swabs are expressed on the right Y-axis. The daily clinical score (bars) are expressed on the left Y-axis.

3.2. Direct contact experiment groups. However, there were no statistical differences in the sur-
vival according to the Cox proportional analysis between different
3.2.1. Inoculation and infectivity exposure durations.
For the IOP-inoculated donor pigs from the contact experiment,
the overall incidence of ASFV infection was 66% (4/6 pigs), identical 3.2.3. Clinical signs
to the incidence in IOP-inoculated animals from the direct inocula- Among the IOP-inoculated donors, the mean number of days to
tion studies. Within DC groups, all animals, regardless of exposure onset of pyrexia was 3.6 ± 0.6 dpi. The mean duration to onset of
time, were successfully infected (incidence = 100%) (Table 2). clinical signs was 4.0 ± 1.7 dpi (Table 2). Two animals were pyrexic
for 1–2 days but their temperature returned to within normal limits
3.2.2. Survival before euthanasia. These two pigs did not become viremic nor was
All donor animals were euthanized between 7 and 9 dpi in accor- shedding detected. One pig was not pyrexic by the time of euthana-
dance with the predetermined scheme to maintain 1:1 ratio with sia, but was viremic and shed virus nasally on the day of euthanasia
contact pigs. Naïve pigs were exposed to IOP-inoculated donors for (8 dpi). Four out of six pigs were obtunded with a mean onset of
1, 2, or 3 days. Overall mean survival for all three exposure groups 4.0 ± 2.5 dpi. Donor animals did not have melena or hematochezia.
(n = 6) was 10.3 ± 1.2 dpc with the longest onset of survival follow- Amongst the DC-exposed pigs, onset of clinical signs began with
ing onset of pyrexia (5.9 ± 2.0 days) compared to direct infection pyrexia that was detected first within the 2 dpc-exposure group
routes (Table 2, Fig. 1B). Animals exposed to donors for 3 dpc had the (3.5 ± 2.1 dpc, n = 2) followed by the 1 dpc-exposure (4.5 ± 0.7 dpc,
shortest overall survival compared to the 1 dpc and 2 dpc-exposure n = 2) and 3 dpc-exposure (7.0 ± 0.0 dpc, n = 2) groups. Obtundation
E.B. Howey et al. / Virus Research 178 (2013) 328–339 335

Fig. 3. Relationship between clinical score and virus shedding in donor pigs inoculated IOP (A) with ASFV Malawi at 105 HAD50 and pigs exposed via Direct Contact (DC) for
24 h (B), 48 h (C) and 72 h (D). Lines indicating viral titer in whole blood, serum, tonsil swabs and nasal swabs are expressed on the right Y-axis. The daily clinical score (bars)
are expressed on the left Y-axis.

was detected within 5 out of 6 pigs with a mean onset of lesion amongst pigs was enlarged and often hemorrhagic lymph
10.6 ± 1.1 dpc. Melena/hematochezia was detected in 2 of 6 pigs nodes. The most severe lymphadenomegaly commonly involved
with a mean onset of 11.5 ± 0.7 dpc. The DC-exposed pigs had the the gastrohepatic lymph nodes. Splenomegaly ranged from mild
highest overall maximum clinical score (6.8 ± 2.0); with the highest to severe enlargement. Renal lesions varied from mild cortical
score in the 2 dpc group followed by the 1 dpc and 3 dpc groups. petechia to renomegaly with diffuse hemorrhage and conges-
tion.
Histologically, lymphoid organs of all animals examined had
3.2.4. Viremia and ASFV shedding
perifollicular regions expanded by cellular debris, extensive
The mean detection of onset of viremia occurred within 4 out of
hemorrhage, and distinct fragmented nuclear remnants, most con-
6 of the donor group pigs at 4.8 ± 2.2 dpi with the mean maximum
sistent with lymphocyte apoptosis (Fig. 4A and B). Lymphoid
whole blood titer of 7.4 ± 3.2 log10 HAD50 /mL. The onset of tonsil
follicles were still identifiable but less distinct compared to normal
shedding was detected prior to viremia in 2 pigs. Onset of tonsil and
lymphoid architecture in naïve pigs.
nasal shedding was detected at 2.8 ± 1.5 and 6.5 ± 1.3 dpi respec-
tively. Maximum titers for tonsil and nasal swabs were 4.1 ± 1.2
and 4.6 ± 1.3 log10 HAD50 /mL respectively (Fig. 3A, Table 3). The dif-
3.4. Immunohistochemical localization of ASFV antigens
ference between onset of viremia and shedding averaged 1.3 ± 0.6
days.
Examination of the distribution of virus and its association
Amongst contact-exposed pigs, mean onset of viremia
with specific cellular markers was performed on tissues collected
was 5.2 ± 1.5 dpc and was detected first within the 1 dpc-
from pigs of each inoculation route. Within lymphoid tissues
exposure group (4.0 ± 1.4 dpc, n = 2) followed by the 2 dpc
of pigs with fulminant ASF, there was extensive cell-associated
and 3 dpc-exposure groups (5.0 ± 1.4 and 6.5 ± 0.7 dpc, n = 2
labeling with the anti-ASFV-VP30 antibody. Localization was pre-
per group). The overall mean highest whole blood titer was
dominately within interfollicular regions with few immunopositive
8.8 ± 0.3 log10 HAD50 /mL, which was the highest amongst all routes
cells within follicles (Fig. 4C). Within the tonsil of the soft
(Table 3). Tonsillar detection of ASFV occurred prior to viremia in 4
palate, labeling was most prominent surrounding tonsillar crypts,
pigs. The mean onset of tonsil and nasal shedding was detected at
with few labeled cells within the tonsillar crypt epithelium.
3.5 ± 2.4 and 7.7 ± 0.6 dpc respectively, with mean maximum titers
This VP30-positive labeling did not colocalize with pancytoker-
of 5.2 ± 0.8 and 4.7 ± 0.1 log10 HAD50 /mL. The difference between
atin.
onset of viremia and shedding averaged 1.7 ± 1.7 days.
Within all positive tissues, immunoreactive cells were mor-
phologically consistent with large mononuclear cells (interpreted
3.3. Postmortem lesions as macrophages). Combined multichannel immunofluorescence
demonstrated that cells immunopositive for ASFV were also com-
All four routes produced comparable gross and histologic monly positive for monocyte/macrophage cellular markers such as
lesions in pigs at terminal stages of disease. The most consistent CD163 (Fig. 4D–G) and CD172a (not shown).
336 E.B. Howey et al. / Virus Research 178 (2013) 328–339

Fig. 4. Histopathology of lesions from pigs infected with ASFV-Malawi. Palatine tonsil, animal #38, INP ASFV-Malawi 102 HAD50 /mL; Lymphoid tissues including tonsil of
the soft palate (A and B), lymph nodes, and spleen displayed loss of lymphocytes and karyorrhectic debris. Scale bars: (A) 500 ␮m, (B) 100 ␮m immunohistochemical staining
(C) via Alkaline phosphatase polymer kit with anti-ASFV VP30 (1D9). Positive labeling extensively throughout interfollicular regions. Scale bar 500 ␮m. Inset (c), region of
interest at higher magnification. Multichannel immunofluorescence (D–G). (D) Low magnification three channel image containing anti-ASFV (red), pancytokeratin (green),
and macrophage marker CD163 (aqua). Scale bar 500 ␮m. (E) Three channel image containing Anti-ASFV (red), leukocyte marker macrophage marker CD163 (aqua) and
nuclear staining with DAPI (blue). (F) Three channel image containing anti-ASFV (red), pancytokeratin (green), and nuclear staining with DAPI (blue). (G) Four channel merged
image with colocalization of anti-ASFV (red) and leukocyte marker macrophage marker CD163 (aqua), pancytokeratin (green), and nuclear staining with DAPI (blue). Scale
bar (E–G) 100 ␮m.

4. Discussion clarify and/or challenge previous conclusions derived from a collec-


tion of pathogenesis studies whose work is not directly comparable
The goal of the investigation described herein was to evaluate due to variability of virus strain, dose, inoculation technique, and
dose- and route-dependent effects on the clinical course of ASF and detection methods. In addition, this work contributes to the estab-
viral dynamics of ASFV within domestic pigs. This work serves to lishment of challenge model systems for ASF which closely simulate
E.B. Howey et al. / Virus Research 178 (2013) 328–339 337

natural infection. The goals of the model systems were to achieve differences in the viremia and shedding parameters between the
modes of challenging pigs that were (1) consistent with previ- intramuscular and the direct contact route suggests that the intra-
ous accounts of ASF, (2) consistent across experimental subjects, muscular route is a poor simulator of natural infection. As a result,
(3) compatible with the current understanding of natural routes inoculation via IM should be avoided in experimental studies try-
of direct pig-to-pig transmission, (4) allowed control of dose and ing to mimic a natural course of disease. Contrastingly, both the
timing of challenge, and (5) provided natural engagement of host IOP and INP inoculated pigs had comparable (i.e. not significantly
immune defenses. Recent studies have led toward standardization different) results compared to the DC exposed pigs suggesting that
of clinical scoring systems for ASF (de Carvalho Ferreira et al., 2012; IOP and INP are more likely to closely simulate natural infection.
Galindo-Cardiel et al., 2013), and the current work demonstrates The implied superior infectivity of ASFV via INP inoculation as
adaptation of these systems where appropriate. compared to IOP was reflected in slightly higher incidence pro-
Four routes of challenge (IM, IOP, INP, DC) resulted in a sim- portion and delayed onset of pyrexia and longer survival time
ilar clinical course of ASF that was consistent with previously seen among INP-inoculated pigs in comparison with all success-
described field cases (Ayoade and Adeyemi, 2003; Detray, 1963; fully IOP-inoculated pigs (including donors in DC experiment).
Sánchez Botija, 1982) and experimental models (Boulanger et al., However, IOP led to significantly higher shedding titers in the
1967; Colgrove et al., 1969; Greig, 1972; Greig and Plowright, tonsil compared with INP inoculation. This effect may have been
1970; Heuschele, 1967; McVicar, 1984; Mebus et al., 1978; Mebus due to anatomic and physiological factors currently under inves-
and Dardiri, 1979, 1980; Plowright et al., 1968; Wilkinson and tigation in our laboratory. IOP inoculation directly deposits the
Donaldson, 1977; Wilkinson et al., 1977, 1980). Clinical signs did inoculum along the tonsil of the soft palate. While this tonsil has
not vary substantially according to route of inoculation. The great- been documented as an early site of viral replication (Heuschele,
est consistency was achieved similarly by IM and DC challenge 1967; Plowright et al., 1968), to our knowledge, specific virus–host
which both had 100% incidence of infection. However, these sys- interactions in ASFV primary infection in vivo have never been
tems suffer from unnatural delivery of virus (IM) and inability to explicitly described. Thus, the lower incidence of infection by
precisely control dose and timing of challenge (DC). All pigs chal- the IOP route compared to INP-inoculation could be explained
lenged via DC became infected with ASFV regardless of length of by lack of establishment of systemic disease subsequent to pri-
exposure, thereby demonstrating that only 24 h of exposure to mary infection within the tonsil of the soft palate and other
shedding animals is required for infection with ASFV. This finding oropharyngeal tissues. Since low-dose INP-inoculated pigs had a
is consistent with the previous works that demonstrated trans- higher incidence of successful infection than IOP, the nasophar-
mission after 24 h (Greig and Plowright, 1970) or 6 h (Ekue et al., ynx may provide more permissive conditions for establishing
1989) of exposure to donor pigs. However, other studies have systemic ASFV infection. INP inoculation targets nasopharyngeal
concluded that the quantity of virus shed 24 h following pyrexia tissues, including nasal tonsil, dorsal soft palate and walls/roof
was inadequate to ensure infection in pigs exposed via direct con- of the nasopharynx. The data presented in this study suggest
tact (Heuschele, 1967; Plowright et al., 1968). INP inoculation had that sites targeted by INP inoculation, or other tissues in the
higher incidence whilst maintaining a natural exposure route and respiratory tract, may provide important portals for ASFV infec-
allowing control of dose and timing of challenge. Additionally, the tion.
shortest time to onset of clinical signs was observed within the Increased inoculation dose of ASFV was associated with a sig-
INP groups, however this difference was not statistically signifi- nificant decrease in survival duration of pigs. Animals inoculated
cant. with 106 HAD50 , regardless of route, had a decreased mean number
Precise determination of pig infectious dose 50% ([PID50 ], i.e. the of days to onset of clinical signs (pyrexia, obtundation, hema-
minimum dose necessary to infected 50% of pigs) for the distinct tochezia/melena) and survival compared to the mid and low dosage
inoculation routes was not possible due to logistical constraints groups. The number of days to onset of viremia and shedding was
which dictated some aspects of experimental design. However, the decreased compared to mid and low doses as well, suggesting that
data herein suggests that the required dose of ASFV to establish a higher viral dose resulted in overwhelming primary replication
infection is lowest for IM, intermediate for INP, and highest for IOP and faster systemic dissemination of the virus. Paradoxically, the
delivery of ASFV to domestic swine. Specifically, low dose inocula- high-dose pigs had lower mean maximum clinical scores because
tion (102 HAD50 ) incidence proportions were 100% (IM), 50% (INP), of the rapidity of progression to death before the full severity of dis-
and 0% (IOP). Yet, at 104 HAD50 all routes had 100% incidence of ease had occurred. Thus, maximum clinical score does not (in itself)
infection. This is consistent with previous studies that have demon- describe the severity of disease but must be evaluated in context
strated low ASFV IM dose (lower than 105 TCID50 ) (Pan and Hess, with duration of survival, virological characteristics, and pathologic
1984; McVicar, 1984) and high IOP dose (PID50 higher than 103 findings.
TCID50 ) (de Carvalho Ferreira et al., 2012; Greig, 1972; McVicar, During the clinical phase of infection, ASFV was variably
1984). Additionally, Maurer et al. demonstrated that a minimal detected in whole blood, serum and tonsil and nasal swabs.
infectious dose of 105 HAD50 was necessary to infect pigs orally Amongst all challenge routes, viremia and shedding commonly
with ASFV (Maurer and Griesemer, 1958). The precise manner by either coincided with the onset of pyrexia or it occurred 2 days
which specific routes of inoculation of ASFV correlate with vari- before or after pyrexia, which is in agreement with other studies
able PID50 via distinct pathogenesis mechanisms remains to be (Greig and Plowright, 1970; McVicar, 1984). The DC-exposed pigs
elucidated. had the highest mean ASFV titer within whole blood. Throughout
The route-specific clinical and virological characteristics the course of this study whole blood samples had titers of almost 2
described herein are likely to be the result of distinct route- log10 higher than serum samples. This is consistent with previous
determined, unique pathogenesis events in early ASFV infection. studies that has described greater number of mature virus particles
Pigs inoculated IM had the shortest mean survival duration; how- associated with erythrocytes and within circulating monocytes in
ever the difference was not statistically significant. Shortest onset whole blood, in comparison to free virions within serum (Genovesi
to viremia amongst IM-inoculated pigs was statistically significant. et al., 1988; Wardley and Wilkinson, 1977).
This may be due to more direct access to the vascular system asso- Detection of ASFV within secretions (tonsil scraping and nasal
ciated with deposition within the muscle as compared to contact swabs) varied across infection routes and dosage groups. ASFV
with intact mucosa as occurred with simulated natural challenge titers in tonsil scrapings and nasal swabs were generally quite sim-
routes. Furthermore, the finding of several statistically significant ilar, suggesting that virus was released from both regions and/or
338 E.B. Howey et al. / Virus Research 178 (2013) 328–339

virus quantities equilibrate between nasal and oral secretions. One Conflict of interests
noteworthy exception to this trend was amongst DC-exposed pigs
wherein ASFV was consistently detected in tonsil scrapings (for 1–6 The authors do not have any financial or personal conflict of
days) prior to detection in nasal swabs. Across all three inocula- interests.
tion routes, initial detection of ASFV shedding occurred at the same
time, or subsequent to, virus detection in whole blood and serum.
Acknowledgements
However within the contact challenge study, ASFV was detected
in tonsil scraping samples 1–4 days prior to detection in blood of
EH planned and carried out the live animal experiments, virus
IOP-inoculated donors and DC-exposed pigs. This indicates that,
isolation, immunomicroscopy and drafted the manuscript. VO pro-
under these distinct conditions, it was uniquely possible to detect
vided expertise in conceptual design, as well as optimization and
local viral replication and shedding within the oral cavity prior to
troubleshooting of techniques. HF performed the statistical analysis
systemic dissemination. This is consistent with previous studies in
and helped with manuscript revisions. MB consulted and con-
which ASFV could be detected within oral and pharyngeal swabs
tributed on all aspects of study design and implementation; also
1–2 days prior to viremia and pyrexia (Greig and Plowright, 1970)
provided critical review of manuscript. JA conceived the study,
or as early as 24 h post inoculation (hpi) in lymphoid and respiratory
oversaw the implementation of experiments, helped draft and
tract tissues (Heuschele, 1967; Plowright et al., 1968).
revised the manuscript. All authors read and approved the final
Gross and histologic changes in pigs in the late stages of disease
manuscript.
were similar regardless of route of infection and were consis-
We wish to thank Meghan Tucker, George Smoliga and Ethan
tent with postmortem findings described in field and experimental
Hartwig for superb technical support. Luis Rodriguez and Carolina
cases of ASF. Systemic hemorrhages affecting the kidney and heart
Stenfeldt are acknowledged for critical review of the manuscript.
and pulmonary congestion and edema are similar to previously
Juan Pacheco supported data manipulation and consulted regard-
described lesions (Detray, 1963; Mebus, 1988). Amongst individ-
ing VI technique. In addition, we would like to thank the staff of the
ual pigs, severity of lesions varied from mild to severe based upon
Animal Resource Branch for their support in collecting data during
recently published recommendations for standardization of patho-
animal experiments.
logic findings for ASF (Galindo-Cardiel et al., 2013). The hallmark
This research was funded, in part, by Agricultural Research
depletion and apoptosis of lymphocytes throughout the lymphoid
Service–Current Research Information System project No. 1940-
tissue with subsequent infiltration of macrophages commonly
32000-056-00D. Additional funds came from an interagency
described for ASF (Gómez-Villamandos et al., 2003) was observed
agreement with the Science and Technology Directorate of the U.S.
histologically within this study. Detailed pathological description
Department of Homeland Security under Award Number HSHQPM-
will be forthcoming in a separate manuscript (Howey et al., in
12-X-00005. Erin Howey’s position is funded through the Plum
preparation).
Island Animal Disease Research Participation Program adminis-
Immunohistochemical visualization of ASFV antigen was
tered by the Oak Ridge Institute for Science and Education through
demonstrated in tonsil of the soft palate of pigs in terminal stages
an interagency agreement between the U.S. Department of Energy
of ASF, further confirming the presence and characterizing the dis-
and the U.S. Department of Agriculture.
tribution of virus within tissues. Immunoreactive cells were large
mononuclear cells and colocalization with monocyte/macrophage
cellular antigen suggested that the predominate infected cell type References
is of monocytic origin. Colocalization of ASFV with CD163 supports
Alonso, C., Galindo, I., Cuesta-Geijo, M.A., Cabezas, M., Hernaez, B., Muñoz-Moreno,
previous suggestions that this molecule may serve as an important R., 2013. African swine fever virus–cell interactions: from virus entry to cell
ASFV receptor (Alonso et al., 2013; Sánchez-Torres et al., 2003). survival. Virus Res. 173 (1), 42–57.
Additionally, this is consistent with numerous reports indicat- Arzt, J., Gregg, D.A., Clavijo, A., Rodriguez, L.L., 2009. Optimization of immuno-
histochemical and fluorescent antibody techniques for localization of
ing monocytotropism of ASFV (Detray, 1963; Tulman et al., 2009;
foot-and-mouth disease virus in animal tissues. J. Vet. Diagn. Invest. 21 (6),
Wardley and Wilkinson, 1978). 779–792.
Arzt, J., White, W.R., Thomsen, B.V., Brown, C.C., 2010. Agricultural diseases on the
move early in the third millennium. Vet. Pathol. 47 (1), 15–27.
Ayoade, G., Adeyemi, I., 2003. African swine fever: an overview. Rev. Élev. Méd. Vét.
5. Conclusions Pays Trop. 56 (3–4), 129–134.
Ballester, M., Galindo-Cardiel, I., Gallardo, C., Argilaguet, J.M., Segalés, J., Rodríguez,
The current study has demonstrated that four modes of chal- J.M., Rodríguez, F., 2010. Intranuclear detection of African swine fever virus DNA
in several cell types from formalin-fixed and paraffin-embedded tissues using a
lenging pigs with ASFV result in similar ASF syndromes with slight
new in situ hybridisation protocol. J. Virol. Methods 168 (1–2), 38–43.
variations. The IM route may be advantageous for the study of late Boinas, F.S., Hutchings, G.H., Dixon, L.K., Wilkinson, P.J., 2004. Characterization
stage ASF events; however several significant differences from DC of pathogenic and non-pathogenic African swine fever virus isolates from
Ornithodoros erraticus inhabiting pig premises in Portugal. J. Gen. Virol. 85 (8),
indicated this is not an optimal system to simulate natural infection.
2177–2187.
DC would be preferable when simulating a natural manner of trans- Borca, M.V., Carrillo, C., Zsak, L., Laegreid, W.W., Kutish, G.F., Neilan, J.G., Burrage,
mission was utmost priority; however this system is expensive due T.G., Rock, D.L., 1998. Deletion of a CD2-like gene, 8-DR, from African swine
to the inclusion of additional (donor) animals in each experiment fever virus affects viral infection in domestic swine. J. Virol. 72 (4), 2881–2889.
Boulanger, P., Bannister, G.L., Gray, D.P., Ruckerbauer, G.M., Willis, N.G., 1967. African
and does not allow control of dose quantity and timing. The two swine fever. II. Detection of the virus in swine tissues by means of the modified
simulated natural manners of inoculation, IOP and INP, allowed direct complement-fixation test. Can. J. Comp. Med. Vet. Sci. 31 (1), 7–11.
precise control of timing and dose of ASFV delivery and had similar Carrasco, L., Chacon, M.d.L.F., Martin de Las Mulas, J., Gómez-Villamandos, J.C., Sierra,
M.A., Villeda, C.J., Wilkinson, P.J., 1997. Ultrastructural changes related to the
incidence of successful infection of pigs. Overall, the slightly lower lymph node haemorrhages in acute African swine fever. Res. Vet. Sci. 62 (3),
minimum infectious dose and slightly higher incidence of infec- 199–204.
tion subsequent to INP inoculation in these experiments suggest Carrasco, L., de Lara, F.C., Martín de las Mulas, J., Gómez-Villamandos, J.C., Hervás, J.,
Wilkinson, P.J., Sierra, M.A., 1996. Virus association with lymphocytes in acute
that INP may be the preferred route of inoculation for pathogenesis African swine fever. Vet. Res. 27 (3), 305–312.
and vaccine challenge studies of pigs with ASFV. Further validation Champely, S., 2012. pwr: Basic functions for power analysis. R package version 1.1.1.
of these techniques with larger sample sizes would contribute to http://CRAN.R-project.org/package=pwr
Childerstone, A., Takamatsu, H., Yang, H., Denyer, M., Parkhouse, R.M., 1998. Modu-
stronger conclusions regarding the differences between the infec-
lation of T cell and monocyte function in the spleen following infection of pigs
tion systems described herein. with African swine fever virus. Vet. Immunol. Immunopathol. 62 (4), 281–296.
E.B. Howey et al. / Virus Research 178 (2013) 328–339 339

Colgrove, G.S., Haelterman, E.O., Coggins, L., 1969. Pathogenesis of African swine Michaud, V., Roger, F., 2013. Review of the sylvatic cycle of African swine fever
fever in young pigs. Am. J. Vet. Res. 30 (8), 1343–1359. in sub-Saharan Africa and the Indian Ocean. Virus Res. 173 (1), 212–227.
Costard, S., Mur, L., Lubroth, J., Sánchez-Vizcaíno, J.M., Pfeiffer, D.U., 2013. Epidemi- Mahy, B.W.J., Kangro, H.O. (Eds.), 1996. Virology Methods Manual. Academic Press,
ology of African swine fever virus. Virus Res. 173 (1), 191–197. San Diego.
Cuesta-Geijo, M.A., Galindo, I., Hernáez, B., Quetglas, J.I., Dalmau-Mena, I., Alonso, Maurer, F.D., Griesemer, R.A., 1958. The pathology of African swine fever; a compar-
C., 2012. Endosomal maturation, Rab7 GTPase and phosphoinositides in African ison with hog cholera. Am. J. Vet. Res. 19 (72), 517–539.
swine fever virus entry. PLoS ONE 7 (11), e48853. McVicar, J.W., 1984. Quantitative aspects of the transmission of African swine fever.
de Carvalho Ferreira, H.C., Weesendorp, E., Elbers, A.R., Bouma, A., Quak, S., Stege- Am. J. Vet. Res. 45 (8), 1535–1541.
man, J.A., Loeffen, W.L., 2012. African swine fever virus excretion patterns in Mebus, C.A., Dardiri, A.H., 1979. Additional characteristics of disease caused by the
persistently infected animals: a quantitative approach. Vet. Microbiol. 160 (3–4), African swine fever viruses isolated from Brazil and the Dominican Republic.
327–340. Proc. Annu. Meet. U. S. Anim. Health Assoc. (83), 227–239.
de Carvalho Ferreira, H.C., Weesendorp, E., Quak, S., Stegeman, J.A., Loeffen, W.L., Mebus, C.A., Dardiri, A.H., 1980. Western hemisphere isolates of African swine fever
2013a. Quantification of airborne African swine fever virus after experimental virus: asymptomatic carriers and resistance to challenge inoculation. Am. J. Vet.
infection? Vet. Microbiol. 165 (3–4), 243–251. Res. 41 (11), 1867–1869.
de Carvalho Ferreira, H.C., Backer, J.A., Weesendorp, E., Klinkenberg, D., Stegeman, Mebus, C.A., Dardiri, A.H., Hamdy, F.M., Ferris, D.H., Hess, W.R., Callis, J.J., 1978.
J.A., Loeffen, W.L., 2013b. Transmission rate of African swine fever virus under Some characteristics of African swine fever viruses isolated from Brazil and
experimental conditions. Vet. Microbiol. 165 (3–4), 296–304. the Dominican Republic. Proc. Annu. Meet. U. S. Anim. Health Assoc. (82),
Detray, D.E., 1963. African swine fever. Adv. Vet. Sci. Comp. Med. 19, 299–333. 232–236.
Ekue, N.F., Wilkinson, P.J., Wardley, R.C., 1989. Infection of pigs with the Cameroon Mebus, C.A., 1988. African swine fever. Adv. Virus Res. 35, 251–269.
isolate (Cam/82) of African swine fever virus. J. Comp. Pathol. 100 (2), 145–154. Oura, C.A., Powell, P.P., Parkhouse, R.M., 1998. Detection of African swine fever virus
Fernández de Marco, M., Salguero, F.J., Bautista, M.J., Núñez, A., Sánchez-Cordón, P.J., in infected pig tissues by immunocytochemistry and in situ hybridisation. J.
Gómez-Villamandos, J.C., 2007. An immunohistochemical study of the tonsils in Virol. Methods 72 (2), 205–217.
pigs with acute African swine fever virus infection. Res. Vet. Sci. 83 (2), 198–203. Pan, I.C., Hess, W.R., 1984. Virulence in African swine fever: its measurement and
Galindo-Cardiel, I., Ballester, M., Solanes, D., Nofrarías, M., López-Soria, S., Argilaguet, implications. Am. J. Vet. Res. 45 (2), 361–366.
J.M., Lacasta, A., Accensi, F., Rodríguez, F., Segalés, J., 2013. Standardization of Penrith, M.L., 2009. African swine fever. Onderstepoort J. Vet. Res. 76 (1), 91–95.
pathological investigations in the framework of experimental ASFV infections. Plowright, W., Parker, J., Staple, R.F., 1968. The growth of a virulent strain of African
Virus Res. 173 (1), 180–190. swine fever virus in domestic pigs. J. Hyg. (Lond.) 66 (1), 117–134.
Galindo, I., Hernáez, B., Muñoz-Moreno, R., Cuesta-Geijo, M.A., Dalmau-Mena, I., R Core Team, 2013. R: A Language and Environment for Statistical
Alonso, C., 2012. The ATF6 branch of unfolded protein response and apopto- Computing. R Foundation for Statistical Computing, Vienna, Austria
sis are activated to promote African swine fever virus infection. Cell Death Dis. http://www.R-project.org/
3 (7), e341. Salguero, F.J., Sánchez-Cordón, P.J., Sierra, M.A., Jover, A., Núñez, A., Gómez-
Genovesi, E.V., Knudsen, R.C., Whyard, T.C., Mebus, C.A., 1988. Moderately viru- Villamandos, J.C., 2004. Apoptosis of thymocytes in experimental African Swine
lent African swine fever virus infection: blood cell changes and infective virus Fever virus infection. Histol. Histopathol. 19 (1), 77–84.
distribution among blood components. Am. J. Vet. Res. 49 (3), 338. Sánchez-Torres, C., Gómez-Puertas, P., Gómez-del-Moral, M., Alonso, F., Escrib-
Gómez-Villamandos, J.C., Bautista, M.J., Carrasco, L., Caballero, M.J., Hervás, J., Villeda, ano, J.M., Ezquerra, A., Domínguez, J., 2003. Expression of porcine CD163 on
C.J., Wilkinson, P.J., Sierra, M.A., 1997a. African swine fever virus infection of bone monocytes/macrophages correlates with permissiveness to African swine fever
marrow: lesions and pathogenesis. Vet. Pathol. 34 (2), 97–107. infection. Arch. Virol. 148 (12), 2307–2323.
Gómez-Villamandos, J.C., Bautista, M.J., Carrasco, L., Chacón-Manrique de Lara, F., Sánchez-Vizcaíno, J.M., Mur, L., Martínez-López, B., 2012. African swine fever: an
Hervás, J., Wilkinson, P.J., Sierra, M.A., 1998. Thrombocytopenia associated with epidemiological update. Transbound Emerg. Dis. 59 (s1), 27–35.
apoptotic megakaryocytes in a viral haemorrhagic syndrome induced by a mod- Sánchez Botija, C., 1982. African Swine Fever. New developments. Rev. Sci. Tech. Off.
erately virulent strain of African swine fever virus. J. Comp. Pathol. 118 (1), Int. Epiz. 1 (4), 1065–1094.
1–13. Tulman, E.R., Delhon, G.A., Ku, B.K., Rock, D.L., 2009. African swine fever virus. Curr.
Gómez-Villamandos, J.C., Carrasco, L., Bautista, M.J., Sierra, M.A., Quezada, M., Top. Microbiol. Immunol. 328, 43–87.
Hervás, J., Chacón, M.de L., Ruiz-Villamor, E., Salguero, F.J., Sónchez-Cordón, P.J., Therneau, T., 2013. A Package for Survival Analysis in S. R package version 2.37-4.
Romanini, S., Núñez, A., Mekonen, T., Méndez, A., Jover, A., 2003. African swine http://CRAN.R-project.org/package=survival
fever and classical swine fever: a review of the pathogenesis. Dtsch. Tierarztl. Wardley, R.C., de, M.A.C., Black, D.N., de Castro Portugal, F.L., Enjuanes, L., Hess, W.R.,
Wochenschr. 110 (4), 165–169. Mebus, C., Ordas, A., Rutili, D., Sánchez-Vizcaíno, J., Vigário, J.D., Wilkinson, P.J.,
Gómez-Villamandos, J.C., Hervás, J., Méndez, A., Carrasco, L., Martín de las Mulas, Moura Nunes, J.F., Thomson, G., 1983. African swine fever virus. Brief review.
J., Villeda, C.J., Wilkinson, P.J., Sierra, M.A., 1995a. Experimental African swine Arch. Virol. 76 (2), 73–90.
fever: apoptosis of lymphocytes and virus replication in other cells. J. Gen. Virol. Wardley, R.C., Wilkinson, P.J., 1978. The growth of virulent African swine fever virus
76 (Pt. 9), 2399–2405. in pig monocytes and macrophages. J. Gen. Virol. 38 (1), 183–186.
Gómez-Villamandos, J.C., Hervás, J., Méndez, A., Carrasco, L., Villeda, C.J., Wilkinson, Wardley, R.C., Wilkinson, P.J., 1977. The association of African swine fever virus with
P.J., Sierra, M.A., 1995b. Ultrastructural study of the renal tubular system in acute blood components of infected pigs. Arch. Virol. 55 (4), 327–334.
experimental African swine fever: virus replication in glomerular mesangial Wilkinson, P.J., Donaldson, A.I., 1977. Transmission studies with African swine fever
cells and in the collecting ducts. Arch. Virol. 140 (3), 581–589. virus. The early distribution of virus in pigs infected by airborne virus. J. Comp.
Gómez-Villamandos, J.C., Hervás, J., Moreno, C., Carrasco, L., Bautista, M.J., Caballero, Pathol. 87 (3), 497–501.
J.M., Wilkinson, P.J., Sierra, M.A., 1997b. Subcellular changes in the tonsils of pigs Wilkinson, P.J., Donaldson, A.I., Greig, A., Bruce, W., 1977. Transmission studies with
infected with acute African swine fever virus. Vet. Res. 28 (2), 179–189. African swine fever virus. Infections of pigs by airborne virus. J. Comp. Pathol.
Greig, A., 1972. Pathogenesis of African swine fever in pigs naturally exposed to the 87 (3), 487–495.
disease. J. Comp. Pathol. 82 (1), 73–79. Wilkinson, P.J., Lawman, M.J., Johnston, R.S., 1980. African swine fever in Malta, 1978.
Greig, A., Plowright, W., 1970. The excretion of two virulent strains of African swine Vet. Rec. 106 (5), 94–97.
fever virus by domestic pigs. J. Hyg. (Lond.) 68 (4), 673–682. Wilkinson, P.J., Wardley, R.C., Williams, S.M., 1981. African swine fever virus
Hamdy, F.M., Dardiri, A.H., 1984. Clinical and immunologic responses of pigs to (Malta/78) in pigs. J. Comp. Pathol. 91 (2), 277–284.
African swine fever virus isolated from the Western Hemisphere. Am. J. Vet. Wilkinson, P.J., Wardley, R.C., Williams, S., 1983. Studies in pigs infected with African
Res. 45 (4), 711. swine fever virus (Malta/78). In: Wilkinson, P.J. (Ed.), CEC/FAO Expert Consulta-
Heuschele, W.P., 1967. Studies on the pathogenesis of African swine fever. I. Quan- tion on African Swine Fever Research. September 1981, Sardinia.
titative studies on the sequential development of virus in pig tissues. Arch. Zsak, L., Borca, M.V., Risatti, G.R., Zsak, A., French, R.A., Lu, Z., Kutish, G.F., Neilan,
Gesamte Virusforsch. 21 (3), 349–356. J.G., Callahan, J.D., Nelson, W.M., Rock, D.L., 2005. Preclinical diagnosis of African
http://www.oie.int/wahis 2/public/wahid.php/Wahidhome/Home (accessed swine fever in contact-exposed swine by a real-time PCR assay. J. Clin. Microbiol.
3.09.13). Jori, F., Vial, L., Penrith, M.L., Perez-Sanchez, R., Etter, E., Albina, E., 43 (1), 112–119.
viruses
Review
African Swine Fever Virus: A Review
Inmaculada Galindo and Covadonga Alonso *
Dpto. de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA),
Ctra. de la Coruña km 7.5, 28040 Madrid, Spain; galindo@inia.es
* Correspondence: calonso@inia.es

Academic Editors: Linda Dixon and Simon Graham


Received: 10 April 2017; Accepted: 4 May 2017; Published: 10 May 2017

Abstract: African swine fever (ASF) is a highly contagious viral disease of swine which causes high
mortality, approaching 100%, in domestic pigs. ASF is caused by a large, double stranded DNA virus,
ASF virus (ASFV), which replicates predominantly in the cytoplasm of macrophages and is the only
member of the Asfarviridae family, genus Asfivirus. The natural hosts of this virus include wild suids
and arthropod vectors of the Ornithodoros genus. The infection of ASFV in its reservoir hosts is usually
asymptomatic and develops a persistent infection. In contrast, infection of domestic pigs leads to a
lethal hemorrhagic fever for which there is no effective vaccine. Identification of ASFV genes involved
in virulence and the characterization of mechanisms used by the virus to evade the immune response
of the host are recognized as critical steps in the development of a vaccine. Moreover, the interplay of
the viral products with host pathways, which are relevant for virus replication, provides the basic
information needed for the identification of potential targets for the development of intervention
strategies against this disease.

Keywords: African swine fever virus; ASFV; virus entry; endocytosis; endosomal pathway; host cell
targets; cellular responses; ER stress; apoptosis; autophagy; A179L

1. Introduction
African swine fever (ASF) is a viral disease of swine that leads to a high mortality in domestic
pigs while being asymptomatic in the natural suid reservoir hosts [1–3]. It causes important economic
losses that are unavoidable in the absence of an effective vaccine and the available methods of disease
control are the quarantine of the affected area and the slaughter of the infected animals [4]. ASF is
caused by the ASF virus (ASFV), a double-stranded DNA virus with a complex molecular structure.
It is the only member of the Asfarviridae family [5] and the only DNA virus transmitted by arthropods,
soft ticks of the Ornithodoros genus [3,6]. Soft ticks (Ornithodoros moubata) are involved in the sylvatic
transmission cycle of the virus in Africa and O. erraticus in Europe. The wild boar that suffers an acute
disease similar to the domestic pig appears to be relevant in the transmission cycle in Europe.
The disease caused by this virus was first identified in Kenya in the 1920s [7]. Then, it was
confined to Africa until it spread to Europe in the middle of the last century, and later to South America
and the Caribbean. The disease was eradicated from Europe (except of Sardinia) at the 1990s via
drastic control and eradication programs. However, in 2007, the disease spread again out of Africa
into the Caucasus, especially Georgia, and in 2014 it reached the eastern territory of the European
Union. The latest reports of the disease include an increasing list of EU countries, Poland and the three
Baltic republics [8,9] and very recently Moldova [10]. Due to the absence of vaccines with protective
efficacy, ASF represents a serious threat to all European countries. The epidemiological complexity
of ASF has been clearly demonstrated in eastern and southern Africa, where genetic characterization
of ASFV based on sequence variation in the C-terminal region of the B646L gene encoding the major
capsid protein p72, revealed the presence of 22 genotypes [11,12]. Recently, a new genotype, genotype

Viruses 2017, 9, 103; doi:10.3390/v9050103 www.mdpi.com/journal/viruses


Viruses 2017, 9, 103 2 of 10

XXIII, that shares a common ancestor with genotypes IX and X, which comprise isolates circulating
in Eastern African countries and the Republic of Congo, has been described [13]. This review paper
summarizes the current state of knowledge about ASFV.

2. African Swine Fever Virus


ASFV is a large, enveloped virus with icosahedral morphology and an average diameter of 200 nm.
The viral genome consists of a single molecule of linear, covalently close-ended, double stranded DNA.
The genomes of different isolates vary in length between 170 and 190 Kbp and encode between 151
and 167 open reading frames. ASFV replication cycle is mainly cytoplasmic, but the nucleus is also a
site of viral DNA synthesis at early times [14,15]. The disassembly of the lamina network close to the
sites where the viral genome starts its replication and the redistribution of several nuclear proteins
suggests the existence of sophisticated mechanisms to regulate the nuclear machinery during viral
infection [16].
Transcription of viral genes is strongly regulated. Four classes of mRNAs have been identified
by their distinctive accumulation kinetics—including immediate–early, early, intermediate, and late
transcripts. Immediate–early and early genes are expressed before the onset of DNA replication,
whereas intermediate and late genes are expressed afterwards. The presence of intermediate genes
suggests a cascade model for the regulation of ASFV gene expression [17,18]. Enzymes required
for DNA replication are expressed immediately after virus entry into the cytoplasm from partially
uncoated core particles and using enzymes and other factors packaged in virus particles [17–20].
Virus morphogenesis takes place in the viral factories where the main late phase of DNA replication
also occurs.

3. Virion Structure
The ASFV particle has an icosahedral morphology composed of several concentric domains: the
internal core formed by the central genome contains the nucleoid, which is coated by a thick protein
layer named core shell; an inner lipid envelope surrounding the core; and finally the capsid, which is
the outermost layer of the intracellular virions [21]. The extracellular virions possess an additional
external envelope that is obtained when the virus buds out through the plasma membrane [22].
However, the importance of this envelope is unclear as it is not required for infectivity [23].

4. Viral Entry Mechanisms


The ASFV infectious cycle starts with the viral adsorption and entry into the host cell. Early
studies on ASFV entry characterized this event as a low pH- and temperature-dependent process
consistent with saturable and specific receptor-mediated endocytosis in Vero cells and porcine
macrophages [24–27]. However, the receptor(s) for the virus still remain unknown. The limited
cell tropism of ASFV suggests that a macrophage-specific receptor is required for infection. Successful
infection of porcine macrophages and monocytes by ASFV correlates to the expression of the CD163
scavenger receptor, a hallmark of macrophage maturation. It was previously suggested as a potential
virus receptor, as monoclonal antibodies against this molecule were able to block infection of primary
alveolar macrophages [28]. However, more recent studies have demonstrated that CD163 is not
necessary for infection with the Georgia 2007/1 virus isolates. Gene-edited pigs possessing a complete
knockout of CD163 produced using the CRISPR/Cas9 system showed no differences in clinical
signs, mortality, pathology, or viremia [29]. One conclusion from these studies was that CD163 may
be necessary but insufficient for infection, suggesting that other macrophage surface proteins may
participate in the infection process.
While there is support for receptor-dependent mechanisms of viral entry, such as
clathrin-mediated dynamin-dependent endocytosis [30,31], there is also evidence that ASFV exploits
other mechanisms, such as phagocytosis [32] and macropinocytosis [33,34]. Also, cholesterol is required
Viruses 2017, 9, 103 3 of 10

for a successful
Viruses 2017, 9, 103 entry. These mechanisms occur both in the macrophage target cell and in Vero3cells of 10
using viral isolates adapted to this cell line.
In addition,
addition, some
someASFV
ASFVproteins
proteinsare
areinvolved
involvedinin
thethe entry
entry mechanism
mechanism such
such as p30,
as p30, important
important for
for viral
viral internalization,
internalization, while
while other
other proteins
proteins suchsuch as and
as p12 p12 p54
and have
p54 have
been been identified
identified as potential
as potential viral
viral attachment
attachment proteins
proteins [35–37].
[35–37].

5. ASFV Enters the Endosomal Pathway


ASFV infection by either pathway of entry should finally reach the endocytic pathway pathway [38]. Once
the virus has entered the endocytic pathway, it it must
must passpass through
through different
different endosome
endosome populations
populations
to achieve a successful infection (Figure 1). Endocytic pathway maturation is carefully orchestrated
by proteins and lipids
lipids that
that are
arerecruited
recruitedto tothe
theendosomal
endosomalmembrane.
membrane.Rab Rab GTPase
GTPase protein
protein family
family is
is
thethe major
major regulator
regulator of of
thethe endosomal
endosomal maturation
maturation pathway,
pathway, where
where each
each member
member of of
thethe
RabRab family
family is
is specifically
specifically located
located to atodifferent
a different endosomal
endosomal compartment
compartment [39].[39]. Incoming
Incoming viruses
viruses are found
are found in
in early
early endosomes (EE) labeled with Rab5 and EEA1 markers from very few
endosomes (EE) labeled with Rab5 and EEA1 markers from very few minutes after adsorption. In minutes after adsorption.
In fact,
fact, complete
complete encapsidated
encapsidated virions
virions are only
are only found found
at theatlevel
the level
of EEofbut
EEnot
butinnot
otherin mature
other mature
acidic
acidic compartments
compartments [38].inhibition
[38]. The The inhibition of endosomal
of endosomal acidification
acidification with bafilomycin
with bafilomycin A1 prevents
A1 prevents viral
viral decapsidation
decapsidation and and
onlyonlyunderunder
thisthis condition
condition it is
it is possibletotoobserve
possible observecomplete
complete viruses
viruses inside
multivesicular endosomes positive for CD63 CD63 andand latelate endosomes
endosomes expressing
expressing Rab7.
Rab7. In normal
conditions, late endosomes harbor harbor only
only viral
viral cores
cores lacking
lacking thethe capsid
capsid protein
protein [38].
[38].

Figure 1.1. ASFV


ASFVenters the the
enters host host
cell through a complex
cell through processprocess
a complex involving dynamin-
involving and clathrin-
dynamin- and
mediated endocytosis
clathrin-mediated and macropinocytosis.
endocytosis Only few
and macropinocytosis. Onlyseconds later, later,
few seconds ASFVASFV
progresses through
progresses the
through
endocytic
the pathway
endocytic andand
pathway reaches
reachesmature
mature endosomal
endosomalcompartments
compartmentswherewhereviral
viral decapsidation
decapsidation and
envelope with
fusion of the inner viral envelope with the
the endosomal
endosomal membrane
membrane occurs.
occurs. Newly synthesized virions
exocytosis budding at the plasma
are assembled in the viral factory and will exit the cell either by exocytosis
membrane or through the formation
formation of of apoptotic
apoptotic bodies.
bodies.

Viral decapsidation
Viral decapsidation occurs
occurs at
at the
the acidic
acidic intraluminal
intraluminal pH
pH in
in mature
mature endosomal
endosomal compartments
compartments
between 30
between 30 and
and 45
45 min
min post
post infection
infection (mpi).
(mpi). Mature
Mature endosomal
endosomal compartments
compartments are
are multivesicular
multivesicular
bodies expressing CD63 that are characterized by the presence of intraluminal vesicles and also, late
endosomes expressing Rab7. Dependence on the endosomal maturation for sequential viral
uncoating and penetration has been also described for other viruses [40,41]. Once decapsidated, virus
particles expose the inner envelope which allows their interaction and subsequent fusion of this viral
Viruses 2017, 9, 103 4 of 10

bodies expressing CD63 that are characterized by the presence of intraluminal vesicles and also, late
endosomes expressing Rab7. Dependence on the endosomal maturation for sequential viral uncoating
and penetration has been also described for other viruses [40,41]. Once decapsidated, virus particles
expose the inner envelope which allows their interaction and subsequent fusion of this viral membrane
with the limiting membrane of the endosomes and naked cores can be released into cytosol in order
to start replication. This process is strongly dependent on the cholesterol efflux at the endosomal
membrane. In fact, blocking cholesterol transport at this level causes retention of virions inside
endosomes, inhibiting infection progression [42]. The inner envelope viral protein pE248R is also
involved in viral fusion. This protein shares sequence similarity with some members of the poxviral
entry/fusion complex [34].
Other inhibitors of endosome maturation such as wortmannin, a phosphatidylinositol 3
(PI3)-kinase inhibitor that blocks early endosome fusion, and nocodazole, an inhibitor that disturbs
microtubule-dependent endosomal transport also prevent ASFV infection [38].

6. ASFV Gene Expression and DNA Replication


Incoming ASFV virions should reach their replication site in the perinuclear area close to the
microtubule organizing center (MTOC) [43]. Immediate early and early genes are expressed before the
onset of DNA replication. Both DNA chains are alternatives used as the coding strand. This is possible
due to the action of several enzymes involved in viral transcription that are packed in the viral core.
Following DNA replication, the transcription of intermediate and late genes begins. ASFV commits
approximately 20% of its genome to encode genes involved in the transcription and modification of its
mRNAs. This transcriptional machinery gives to ASFV a relative independence from its host and an
accurate positional and temporal control of its gene expression [44]. The existence of a nuclear stage
in the replication of ASFV DNA has been determined by in situ hybridization and autoradiography
in thin sections of infected cells, although the precise role of the nucleus in viral replication remains
unclear. Sedimentation analysis of replicating viral DNA in alkaline sucrose gradients has shown
that small DNA fragments are pulse-labeled in the nucleus at early times in the replication of the
viral DNA, whereas larger molecules are synthesized in the cytoplasm at later times. The replicative
intermediates that are synthesized both in the nucleus and cytoplasm of ASFV-infected cells consist
of head to head concatemers. It is possible that the nucleus may provide small transcripts or other
factors required for priming virus replication or that an early stage of virus DNA replication should
take place [15].

7. Formation of the Viral Factory


Microtubules are required for the transport of the virus to perinuclear area, where replication takes
place. Integrity of microtubules is required for the formation of viral factories [45] and virus particles
are found associated to stabilized microtubules at entry [46]. Also, nocodazole, which interferes with
the polymerization of microtubule filaments, prevents the correct formation of factories [43,47–49].
Structural protein p54 interacts with the dynein motor protein during virus infection and could
constitute a molecular mechanism for microtubule-mediated virus transport [43].
The viral factory, localized in the cytoplasm close to the nucleus, can be described as a single
and large perinuclear area at the MTOC. On this site, viral proteins and DNA are accumulated and
newly synthesized virions are assembled. A cage made of intermediate filament vimentin surrounds
the viral factory likely to prevent the sensing of viral components into the cytoplasm and concentrate
structural proteins at sites of assembly [50]. There are many features shared between aggresomes
and VFs. However, neither HDAC6 nor Bag3 are required for factory formation, suggesting that
aggresomes and viral factories are not the same structures [51].
Formation of viral replication sites depends on several cellular determinants. For example, Rho
GTPase inhibitors produce an abnormal viral factory size with the accumulation of envelope precursors
and immature virions [46]. This specialized site at the MTOC, contains viral DNA, most of the viral
Viruses 2017, 9, 103 5 of 10

proteins, immature and mature virions, and also abundant virus-induced membranes. Viral factories
contain precursors that develop into icosahedral intermediates by the assembly of the icosahedral
capsid and the core shell domain. The last step of virion morphogenesis will be the encapsidation
of DNA giving rise to mature virions. Finally, the newly formed virus leaves the factory and is
transported to the cell surface by kinesin where it is released by budding [50]. Extracellular virus is
covered by an additional external envelope that is acquired during this budding process [22].

8. ER Stress and Unfolded Protein Response


The virus modifies and interacts with cellular pathways in response to infection. The endoplasmic
reticulum (ER) is an essential organelle for ASFV replication and maturation and a large number of
viral proteins are synthesized in infected cells and accumulated in the ER during the viral life cycle.
This process can trigger ER stress and the unfolded protein response (UPR) of the host cell as the
induction of caspase 12 indicates [52]. Viruses have evolved various mechanisms to counteract these
cellular responses that would limit or inhibit viral replication. This response is a regulatory program
that upregulates a large number of genes, such as ER chaperones and ER-associated degradation
(ERAD) components, which increase the folding capacity of the ER. ASFV induces the upregulation of
the chaperones calnexin and calreticulin, but not ERp57, PDI [52], or BiP/Grp78 [52,53]. Moreover,
ASFV induces selectively the transcription factor 6 (ATF6) signaling pathway of the UPR, but not the
protein kinase-like ER kinase (PERK) or the inositol-requiring enzyme 1 (IRE1) pathways. Thus, the
capacity of ASFV to regulate the UPR signaling cascade may prevent the effects that are detrimental to
the infection, while maintaining those that are beneficial. Importantly, viral protein DP71L is involved
in ATF4 downregulation and in CHOP inhibition [54]. DP71L, homolog of the neurovirulence factor
ICP34.5 of HSV-1 and the cellular gene GADD34, binds to catalytic subunit of protein phosphatase
1 (PP1) and causes the dephosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2 α),
thereby preventing the inhibition of protein synthesis produced by ER stress and the UPR [55].

9. ASFV and Apoptosis


ER stress after ASFV infection is reflected by the activation of caspase 12, which follows similar
temporal dynamics to the activation of mitochondrial caspase 9 and effector caspase 3 [52]. Apoptosis
represents an important innate cellular mechanism to prevent virus infection, and many viruses
have developed strategies for inhibiting or delaying this cellular response in turn [56]. Thus, ASFV
A179L gene encodes an homolog of antiapoptotic Bcl-2 protein to prolong host cell survival until
the replication of the viral genome is completed [57]. This viral Bcl-2 is expressed both at early and
late times after infection and inhibits the action of several pro-apoptotic BH3-only proteins, known
to be rapid inducers of apoptosis, such as activated Bid, BimL, BimS, BimEL, Bad, Bmf, Bik, Puma,
and DP5 [58]. Another ASFV gene, A224L, encodes a member of the family of apoptosis inhibitors
known as IAP proteins and is able to inhibit caspase activation and to promote cell survival [59,60].
Viral IAP not only blocks caspase-3 activation but also activates NF-κB [61]. It is interesting that the
virus encodes an IκB-like molecule (A238L) that interferes with NF-κB activation [62]. A238L and
A224L are expressed at different times during ASFV infection, this suggests that ASFV requires a
low NF-κB activity at early times of infection to avoid immune responses but a higher activity at
late times, probably to prevent apoptosis as the cellular systems are abused [61]. At very late stages
of ASFV infection, infected cells undergo apoptosis [63] and show the characteristic morphological
changes of programmed cell death, including typical membrane blebbing of the infected cell that led
to the formation of numerous vesicles containing virus [45] and this could be an efficient system for
virus spread.

10. ASFV and Autophagy


A179L, the viral Bcl2 homolog of African swine fever virus, not only interacts with pro-apoptotic
Bcl2 family proteins to inhibit apoptosis but also inhibits autophagy by interacting with Beclin1
Viruses 2017, 9, 103 6 of 10

through its BH3 homology domain. ASFV is armed to counteract elimination by autophagy as other
DNA viruses. An example of that is the HSV-1 ICP34.5 protein, which inhibits autophagy by targeting
Beclin1 [64]. ASFV encodes a protein homologous to ICP34.5, which exerts other functions. ASFV
DP71L inhibits the ER stress response activating PP1/protein phosphatase 1 [55]. However, in contrast
to HSV-1 ICP34.5, it does not interact with Beclin1 [65].
In fact, ASFV infection does not induce LC3 activation or autophagosome formation in Vero
cells [65]. Autophagy is a relevant cellular defense mechanism that allows the orderly degradation
and recycling of cellular components. Autophagy eliminates intracellular pathogens and has a crucial
role for innate and adaptive immune responses. Some DNA viruses, such as ASFV and HSV-1, have
developed strategies to keep this cellular response under control to prevent the degradation of newly
assembled virions. In contrast, most RNA viruses have been reported to induce autophagy in infected
cells, and in several cases autophagy may enhance viral replication [66].

11. ASFV Egress


The mature particle is transported from the virus factories to the cell surface through a
microtubule-mediated mechanism [48] depending on the motor protein conventional kinesin [67] and
on the capsid protein pE120R [68]. Once on the cell surface, particles exit the host cell by budding at
the membrane, acquiring an additional envelope [22]. Both intracellular and extracellular viruses are
infectious but structurally and antigenically different [68], and this could have important implications
in the host immune response against ASFV.

12. Potential Vaccines and Antivirals


Previous attempts to develop vaccines against ASFV have failed to induce protective immunity.
Currently, there are several reports of the protection elicited by experimental vaccines based in live
attenuated ASFV (LAV) containing single or double gene deletions in the genome [69] and other
different approaches, including DNA vaccines [70,71]. Some of them might turn into successful
vaccine candidates. Nowadays, a vaccine to be used in the field should meet a number of requirements.
Any potential ASFV vaccine should include markers for differentiation between infected or vaccinated
animals that allows for DIVA diagnostics. Also, vaccine production could not be possible because
of the lack of a cell line supporting the replication of attenuated vaccine viruses without modifying
the virus virulence. At present, wild boars were found to be major players of disease spread in both
the Baltics and Poland. Hence, a live vaccine given as bait for these animals could be crucial to limit
disease spread. In the current scenario, a vaccine would make possible to eradicate the disease together
with infection surveillance by diagnostics. Available diagnostic methods allow both virus detection
and also detection of antibodies for identification of survivors and asymptomatic carriers [72].
Until an effective vaccine is developed, the possibility of using antivirals remains. Antiviral
strategies have been extensively applied in human infections but can be used in animal health.
Antivirals are useful for an early control of virus spread after an outbreak. In addition, the
combination of antivirals with vaccination protocols could be applied to elicit the immune response
required for effective protection against the disease. Here, we have reviewed potential molecular
targets to be considered as targets for antivirals. Antivirals against ASFV described include
resveratrol and oxyresveratrol [73], microalgae [74], cholesterol lowering drugs [46] or inhibitors
of cholesterol transport [42], antitumoral lauryl-gallate [75], anticonvulsivant valproic acid [75],
dynamin inhibitors [38], fluoroquinolones [76], serine protease inhibitors [52], specific peptides [77],
and miscelanea [32]. These could be eventually applied in a quick response to reduce susceptible
animals in order to create ‘safe areas’ around the outbreaks and thus control the spread of the infection.

Acknowledgments: This work was funded by grant AGL2015-69598-R of the Ministerio de Economía, Industria
y Competitividad of Spain.
Conflicts of Interest: The authors declare no conflict of interest.
Viruses 2017, 9, 103 7 of 10

References
1. Parker, J.; Plowright, W.; Pierce, M.A. The epizootiology of african swine fever in Africa. Vet. Rec. 1969, 85,
668–674. [PubMed]
2. Thomson, G.R.; Gainaru, M.D.; van Dellen, A.F. Experimental infection of warthos (Phacochoerus aethiopicus)
with african swine fever virus. Onderstepoort J. Vet. Res. 1980, 47, 19–22. [PubMed]
3. Anderson, E.C.; Hutchings, G.H.; Mukarati, N.; Wilkinson, P.J. African swine fever virus infection of the
bushpig (Potamochoerus porcus) and its significance in the epidemiology of the disease. Vet. Microbiol. 1998,
62, 1–15. [CrossRef]
4. Penrith, M.L.; Vosloo, W. Review of african swine fever: Transmission, spread and control. J. S. Afr. Vet.
Assoc. 2009, 80, 58–62. [CrossRef] [PubMed]
5. Dixon, L.K.; Escribano, J.M.; Martins, C.; Rock, D.L.; Salas, M.L.; Wilkinson, P.J. Asfarviridae. In Virus
Taxonomy. VIIIth Report of the ICTV; Fauquet, C.M., Mayo, M.A., Maniloff, J., Desselberger, U., Ball, L.A., Eds.;
Elsevier/Academic Press: London, UK, 2005; pp. 135–143.
6. Kleiboeker, S.B.; Scoles, G.A.; Burrage, T.G.; Sur, J. African swine fever virus replication in the midgut
epithelium is required for infection of ornithodoros ticks. J. Virol. 1999, 73, 8587–8598. [PubMed]
7. Montgomery, R.E. On a form of swine fever occurring in british east Africa (Kenya Colony). J. Comp. Pathol.
Ther. 1921, 34, 159–191. [CrossRef]
8. Wozniakowski, G.; Kozak, E.; Kowalczyk, A.; Lyjak, M.; Pomorska-Mol, M.; Niemczuk, K.; Pejsak, Z. Current
status of african swine fever virus in a population of wild boar in eastern Poland (2014–2015). Arch. Virol.
2016, 161, 189–195. [CrossRef] [PubMed]
9. Pejsak, Z.; Truszczynski, M.; Niemczuk, K.; Kozak, E.; Markowska-Daniel, I. Epidemiology of african swine
fever in Poland since the detection of the first case. Pol. J. Vet. Sci. 2014, 17, 665–672. [CrossRef] [PubMed]
10. OIE-WAHID. World Animal Health Information Database (Wahid) [Database on the Internet]. World
Organisation for Animal Health (OIE), 2017. [Cited World Animal Health Information System (WAHIS)].
Available online: http://www.Oie.Int/wahis/public.Php?Page=home (accessed on 27 March 2017).
11. Boshoff, C.I.; Bastos, A.D.; Gerber, L.J.; Vosloo, W. Genetic characterisation of african swine fever viruses
from outbreaks in southern Africa (1973–1999). Vet. Microbiol. 2007, 121, 45–55. [CrossRef] [PubMed]
12. Bastos, A.D.; Penrith, M.L.; Cruciere, C.; Edrich, J.L.; Hutchings, G.; Roger, F.; Couacy-Hymann, E.;
Thomson, G.R. Genotyping field strains of african swine fever virus by partial p72 gene characterisation.
Arch. Virol. 2003, 148, 693–706. [CrossRef] [PubMed]
13. Achenbach, J.E.; Gallardo, C.; Nieto-Pelegrin, E.; Rivera-Arroyo, B.; Degefa-Negi, T.; Arias, M.; Jenberie, S.;
Mulisa, D.D.; Gizaw, D.; Gelaye, E.; et al. Identification of a new genotype of african swine fever virus in
domestic pigs from Ethiopia. Transbound. Emerg. Dis. 2016. [CrossRef] [PubMed]
14. Garcia-Beato, R.; Salas, M.L.; Vinuela, E.; Salas, J. Role of the host cell nucleus in the replication of African
swine fever virus DNA. Virology 1992, 188, 637–649. [CrossRef]
15. Rojo, G.; Garcia-Beato, R.; Vinuela, E.; Salas, M.L.; Salas, J. Replication of african swine fever virus DNA in
infected cells. Virology 1999, 257, 524–536. [CrossRef] [PubMed]
16. Ballester, M.; Rodriguez-Carino, C.; Perez, M.; Gallardo, C.; Rodriguez, J.M.; Salas, M.L.; Rodriguez, F.
Disruption of nuclear organization during the initial phase of african swine fever virus infection. J. Virol.
2011, 85, 8263–8269. [CrossRef] [PubMed]
17. Almazan, F.; Rodriguez, J.M.; Andres, G.; Perez, R.; Vinuela, E.; Rodriguez, J.F. Transcriptional analysis of
multigene family 110 of african swine fever virus. J. Virol. 1992, 66, 6655–6667. [PubMed]
18. Rodriguez, J.M.; Salas, M.L.; Vinuela, E. Intermediate class of mRNAs in African swine fever virus. J. Virol.
1996, 70, 8584–8589. [PubMed]
19. Almazan, F.; Rodriguez, J.M.; Angulo, A.; Vinuela, E.; Rodriguez, J.F. Transcriptional mapping of a late gene
coding for the p12 attachment protein of African swine fever virus. J. Virol. 1993, 67, 553–556. [PubMed]
20. Salas, M.L.; Kuznar, J.; Vinuela, E. Polyadenylation, methylation, and capping of the RNA synthesized
in vitro by African swine fever virus. Virology 1981, 113, 484–491. [CrossRef]
21. Salas, M.L.; Andres, G. African swine fever virus morphogenesis. Virus Res. 2013, 173, 29–41. [CrossRef]
[PubMed]
22. Breese, S.S., Jr.; de Boer, C.J. Electron microscope observations of African swine fever virus in tissue culture
cells. Virology 1966, 28, 420–428. [CrossRef]
Viruses 2017, 9, 103 8 of 10

23. Schloer, G.M. Polypeptides and structure of African swine fever virus. Virus Res. 1985, 3, 295–310. [CrossRef]
24. Valdeira, M.L.; Geraldes, A. Morphological study on the entry of African swine fever virus into cells. Biol.
Cell 1985, 55, 35–40. [CrossRef] [PubMed]
25. Alcami, A.; Carrascosa, A.L.; Vinuela, E. Interaction of African swine fever virus with macrophages. Virus
Res. 1990, 17, 93–104. [CrossRef]
26. Alcami, A.; Carrascosa, A.L.; Vinuela, E. The entry of African swine fever virus into vero cells. Virology 1989,
171, 68–75. [CrossRef]
27. Alcami, A.; Carrascosa, A.L.; Vinuela, E. Saturable binding sites mediate the entry of African swine fever
virus into vero cells. Virology 1989, 168, 393–398. [CrossRef]
28. Sanchez-Torres, C.; Gomez-Puertas, P.; Gomez-del-Moral, M.; Alonso, F.; Escribano, J.M.; Ezquerra, A.;
Dominguez, J. Expression of porcine cd163 on monocytes/macrophages correlates with permissiveness to
African swine fever infection. Arch. Virol. 2003, 148, 2307–2323. [CrossRef] [PubMed]
29. Popescu, L.; Gaudreault, N.N.; Whitworth, K.M.; Murgia, M.V.; Nietfeld, J.C.; Mileham, A.; Samuel, M.;
Wells, K.D.; Prather, R.S.; Rowland, R.R. Genetically edited pigs lacking cd163 show no resistance following
infection with the African swine fever virus isolate, georgia 2007/1. Virology 2017, 501, 102–106. [CrossRef]
[PubMed]
30. Galindo, I.; Cuesta-Geijo, M.A.; Hlavova, K.; Munoz-Moreno, R.; Barrado-Gil, L.; Dominguez, J.; Alonso, C.
African swine fever virus infects macrophages, the natural host cells, via clathrin and cholesterol-dependent
endocytosis. Virus Res. 2015, 200, 45–55. [CrossRef] [PubMed]
31. Hernaez, B.; Alonso, C. Dynamin and clathrin-dependent endocytosis in African swine fever virus entry.
J. Virol. 2010, 84, 2100–2109. [CrossRef] [PubMed]
32. Basta, S.; Gerber, H.; Schaub, A.; Summerfield, A.; McCullough, K.C. Cellular processes essential for African
swine fever virus to infect and replicate in primary macrophages. Vet. Microbiol. 2010, 140, 9–17. [CrossRef]
[PubMed]
33. Sanchez, E.G.; Quintas, A.; Perez-Nunez, D.; Nogal, M.; Barroso, S.; Carrascosa, A.L.; Revilla, Y. African
swine fever virus uses macropinocytosis to enter host cells. PLoS Pathog. 2012, 8, e1002754. [CrossRef]
[PubMed]
34. Hernaez, B.; Guerra, M.; Salas, M.L.; Andres, G. African swine fever virus undergoes outer envelope
disruption, capsid disassembly and inner envelope fusion before core release from multivesicular endosomes.
PLoS Pathog. 2016, 12, e1005595. [CrossRef] [PubMed]
35. Gomez-Puertas, P.; Rodriguez, F.; Oviedo, J.M.; Brun, A.; Alonso, C.; Escribano, J.M. The african swine fever
virus proteins p54 and p30 are involved in two distinct steps of virus attachment and both contribute to the
antibody-mediated protective immune response. Virology 1998, 243, 461–471. [CrossRef] [PubMed]
36. Angulo, A.; Vinuela, E.; Alcami, A. Comparison of the sequence of the gene encoding African swine fever
virus attachment protein p12 from field virus isolates and viruses passaged in tissue culture. J. Virol. 1992,
66, 3869–3872. [PubMed]
37. Angulo, A.; Vinuela, E.; Alcami, A. Inhibition of African swine fever virus binding and infectivity by purified
recombinant virus attachment protein p12. J. Virol. 1993, 67, 5463–5471. [PubMed]
38. Cuesta-Geijo, M.A.; Galindo, I.; Hernaez, B.; Quetglas, J.I.; Dalmau-Mena, I.; Alonso, C. Endosomal
maturation, Rab7 GTPase and phosphoinositides in African swine fever virus entry. PLoS ONE 2012,
7, e48853. [CrossRef] [PubMed]
39. Stenmark, H. Rab GTPases as coordinators of vesicle traffic. Nat. Rev. Mol. Cell Biol. 2009, 10, 513–525.
[CrossRef] [PubMed]
40. Lozach, P.Y.; Mancini, R.; Bitto, D.; Meier, R.; Oestereich, L.; Overby, A.K.; Pettersson, R.F.; Helenius, A. Entry
of bunyaviruses into mammalian cells. Cell Host Microbe 2010, 7, 488–499. [CrossRef] [PubMed]
41. Pasqual, G.; Rojek, J.M.; Masin, M.; Chatton, J.Y.; Kunz, S. Old world arenaviruses enter the host cell via the
multivesicular body and depend on the endosomal sorting complex required for transport. PLoS Pathog.
2011, 7, e1002232. [CrossRef]
42. Cuesta-Geijo, M.A.; Chiappi, M.; Galindo, I.; Barrado-Gil, L.; Munoz-Moreno, R.; Carrascosa, J.L.; Alonso, C.
Cholesterol flux is required for endosomal progression of african swine fever virions during the initial
establishment of infection. J. Virol. 2015, 90, 1534–1543. [CrossRef] [PubMed]
Viruses 2017, 9, 103 9 of 10

43. Alonso, C.; Miskin, J.; Hernaez, B.; Fernandez-Zapatero, P.; Soto, L.; Canto, C.; Rodriguez-Crespo, I.; Dixon, L.;
Escribano, J.M. African swine fever virus protein p54 interacts with the microtubular motor complex through
direct binding to light-chain dynein. J. Virol. 2001, 75, 9819–9827. [CrossRef] [PubMed]
44. Rodriguez, J.M.; Salas, M.L. African swine fever virus transcription. Virus Res. 2013, 173, 15–28. [CrossRef]
[PubMed]
45. Hernaez, B.; Escribano, J.M.; Alonso, C. Visualization of the African swine fever virus infection in living cells
by incorporation into the virus particle of green fluorescent protein-p54 membrane protein chimera. Virology
2006, 350, 1–14. [CrossRef] [PubMed]
46. Quetglas, J.I.; Hernaez, B.; Galindo, I.; Munoz-Moreno, R.; Cuesta-Geijo, M.A.; Alonso, C. Small Rho GTPases
and cholesterol biosynthetic pathway intermediates in African swine fever virus infection. J. Virol. 2012, 86,
1758–1767. [CrossRef] [PubMed]
47. Carvalho, Z.G.; de Matos, A.P.; Rodrigues-Pousada, C. Association of African swine fever virus with the
cytoskeleton. Virus Res. 1988, 11, 175–192. [CrossRef]
48. De Matos, A.P.; Carvalho, Z.G. African swine fever virus interaction with microtubules. Biol. Cell 1993, 78,
229–234. [CrossRef]
49. Heath, C.M.; Windsor, M.; Wileman, T. Aggresomes resemble sites specialized for virus assembly. J. Cell Biol.
2001, 153, 449–455. [CrossRef] [PubMed]
50. Stefanovic, S.; Windsor, M.; Nagata, K.I.; Inagaki, M.; Wileman, T. Vimentin rearrangement during African
swine fever virus infection involves retrograde transport along microtubules and phosphorylation of
vimentin by calcium calmodulin kinase II. J. Virol. 2005, 79, 11766–11775. [CrossRef] [PubMed]
51. Munoz-Moreno, R.; Barrado-Gil, L.; Galindo, I.; Alonso, C. Analysis of HDAC6 and BAG3-aggresome
pathways in African swine fever viral factory formation. Viruses 2015, 7, 1823–1831. [CrossRef] [PubMed]
52. Galindo, I.; Hernaez, B.; Munoz-Moreno, R.; Cuesta-Geijo, M.A.; Dalmau-Mena, I.; Alonso, C. The ATF6
branch of unfolded protein response and apoptosis are activated to promote African swine fever virus
infection. Cell Death Dis. 2012, 3, e341. [CrossRef] [PubMed]
53. Netherton, C.L.; Parsley, J.C.; Wileman, T. African swine fever virus inhibits induction of the stress-induced
proapoptotic transcription factor CHOP/GADD153. J. Virol. 2004, 78, 10825–10828. [CrossRef] [PubMed]
54. Zhang, F.; Moon, A.; Childs, K.; Goodbourn, S.; Dixon, L.K. The African swine fever virus DP71L protein
recruits the protein phosphatase 1 catalytic subunit to dephosphorylate eIF2alpha and inhibits chop induction
but is dispensable for these activities during virus infection. J. Virol. 2010, 84, 10681–10689. [CrossRef]
[PubMed]
55. Rivera, J.; Abrams, C.; Hernaez, B.; Alcazar, A.; Escribano, J.M.; Dixon, L.; Alonso, C. The MyD116 African
swine fever virus homologue interacts with the catalytic subunit of protein phosphatase 1 and activates its
phosphatase activity. J. Virol. 2007, 81, 2923–2929. [CrossRef] [PubMed]
56. Benedict, C.A.; Norris, P.S.; Ware, C.F. To kill or be killed: Viral evasion of apoptosis. Nat. Immunol. 2002, 3,
1013–1018. [CrossRef] [PubMed]
57. Brun, A.; Rivas, C.; Esteban, M.; Escribano, J.M.; Alonso, C. African swine fever virus gene a179l, a viral
homologue of BCL-2, protects cells from programmed cell death. Virology 1996, 225, 227–230. [CrossRef]
[PubMed]
58. Galindo, I.; Hernaez, B.; Diaz-Gil, G.; Escribano, J.M.; Alonso, C. A179l, a viral BCL-2 homologue, targets the
core BCL-2 apoptotic machinery and its upstream BH3 activators with selective binding restrictions for bid
and noxa. Virology 2008, 375, 561–572. [CrossRef] [PubMed]
59. Chacon, M.R.; Almazan, F.; Nogal, M.L.; Vinuela, E.; Rodriguez, J.F. The African swine fever virus IAP
homolog is a late structural polypeptide. Virology 1995, 214, 670–674. [CrossRef] [PubMed]
60. Nogal, M.L.; Gonzalez de Buitrago, G.; Rodriguez, C.; Cubelos, B.; Carrascosa, A.L.; Salas, M.L.; Revilla, Y.
African swine fever virus IAP homologue inhibits caspase activation and promotes cell survival in
mammalian cells. J. Virol. 2001, 75, 2535–2543. [CrossRef] [PubMed]
61. Rodriguez, C.I.; Nogal, M.L.; Carrascosa, A.L.; Salas, M.L.; Fresno, M.; Revilla, Y. African swine fever virus
IAP-like protein induces the activation of nuclear factor kappa B. J. Virol. 2002, 76, 3936–3942. [CrossRef]
[PubMed]
62. Revilla, Y.; Callejo, M.; Rodriguez, J.M.; Culebras, E.; Nogal, M.L.; Salas, M.L.; Vinuela, E.; Fresno, M.
Inhibition of nuclear factor kappa b activation by a virus-encoded I kappa b-like protein. J. Biol. Chem. 1998,
273, 5405–5411. [CrossRef] [PubMed]
Viruses 2017, 9, 103 10 of 10

63. Ramiro-Ibanez, F.; Ortega, A.; Brun, A.; Escribano, J.M.; Alonso, C. Apoptosis: A mechanism of cell killing
and lymphoid organ impairment during acute african swine fever virus infection. J. Gen. Virol. 1996, 77,
2209–2219. [CrossRef] [PubMed]
64. Orvedahl, A.; Alexander, D.; Talloczy, Z.; Sun, Q.; Wei, Y.; Zhang, W.; Burns, D.; Leib, D.A.; Levine, B. HSV-1
ICP34.5 confers neurovirulence by targeting the Beclin 1 autophagy protein. Cell Host Microbe 2007, 1, 23–35.
[CrossRef] [PubMed]
65. Hernaez, B.; Cabezas, M.; Munoz-Moreno, R.; Galindo, I.; Cuesta-Geijo, M.A.; Alonso, C. A179l, a new viral
Bcl2 homolog targeting Beclin 1 autophagy related protein. Curr. Mol. Med. 2013, 13, 305–316. [CrossRef]
[PubMed]
66. Klionsky, D.J. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition).
Autophagy 2016, 12, 1–222. [CrossRef] [PubMed]
67. Jouvenet, N.; Monaghan, P.; Way, M.; Wileman, T. Transport of african swine fever virus from assembly
sites to the plasma membrane is dependent on microtubules and conventional kinesin. J. Virol. 2004, 78,
7990–8001. [CrossRef] [PubMed]
68. Andres, G.; Garcia-Escudero, R.; Vinuela, E.; Salas, M.L.; Rodriguez, J.M. African swine fever virus structural
protein pE120R is essential for virus transport from assembly sites to plasma membrane but not for infectivity.
J. Virol. 2001, 75, 6758–6768. [CrossRef] [PubMed]
69. O’Donnell, V.; Risatti, G.R.; Holinka, L.G.; Krug, P.W.; Carlson, J.; Velazquez-Salinas, L.; Azzinaro, P.A.;
Gladue, D.P.; Borca, M.V. Simultaneous deletion of the 9gl and uk genes from the African swine fever virus
georgia 2007 isolate offers increased safety and protection against homologous challenge. J. Virol. 2017, 91,
e01760-16.
70. Lacasta, A.; Ballester, M.; Monteagudo, P.L.; Rodriguez, J.M.; Salas, M.L.; Accensi, F.; Pina-Pedrero, S.;
Bensaid, A.; Argilaguet, J.; Lopez-Soria, S.; et al. Expression library immunization can confer protection
against lethal challenge with african swine fever virus. J. Virol. 2014, 88, 13322–13332. [CrossRef] [PubMed]
71. Reis, A.L.; Abrams, C.C.; Goatley, L.C.; Netherton, C.; Chapman, D.G.; Sanchez-Cordon, P.; Dixon, L.K.
Deletion of African swine fever virus interferon inhibitors from the genome of a virulent isolate reduces
virulence in domestic pigs and induces a protective response. Vaccine 2016, 34, 4698–4705. [CrossRef]
[PubMed]
72. Cubillos, C.; Gomez-Sebastian, S.; Moreno, N.; Nunez, M.C.; Mulumba-Mfumu, L.K.; Quembo, C.J.; Heath, L.;
Etter, E.M.; Jori, F.; Escribano, J.M.; et al. African swine fever virus serodiagnosis: A general review with a
focus on the analyses of african serum samples. Virus Res. 2013, 173, 159–167. [CrossRef] [PubMed]
73. Galindo, I.; Hernaez, B.; Berna, J.; Fenoll, J.; Cenis, J.L.; Escribano, J.M.; Alonso, C. Comparative inhibitory
activity of the stilbenes resveratrol and oxyresveratrol on african swine fever virus replication. Antivir. Res.
2011, 91, 57–63. [CrossRef] [PubMed]
74. Fabregas, J.; Garcia, D.; Fernandez-Alonso, M.; Rocha, A.I.; Gomez-Puertas, P.; Escribano, J.M.; Otero, A.;
Coll, J.M. In vitro inhibition of the replication of haemorrhagic septicaemia virus (VHSV) and African swine
fever virus (ASFV) by extracts from marine microalgae. Antivir. Res. 1999, 44, 67–73. [CrossRef]
75. Hurtado, C.; Bustos, M.J.; Sabina, P.; Nogal, M.L.; Granja, A.G.; Gonzalez, M.E.; Gonzalez-Porque, P.;
Revilla, Y.; Carrascosa, A.L. Antiviral activity of lauryl gallate against animal viruses. Antivir. Ther. 2008, 13,
909–917. [PubMed]
76. Mottola, C.; Freitas, F.B.; Simoes, M.; Martins, C.; Leitao, A.; Ferreira, F. In vitro antiviral activity of
fluoroquinolones against African swine fever virus. Vet. Microbiol. 2013, 165, 86–94. [CrossRef] [PubMed]
77. Hernaez, B.; Tarrago, T.; Giralt, E.; Escribano, J.M.; Alonso, C. Small peptide inhibitors disrupt a high-affinity
interaction between cytoplasmic dynein and a viral cargo protein. J. Virol. 2010, 84, 10792–10801. [CrossRef]
[PubMed]

© 2017 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).

Anda mungkin juga menyukai