Está en la página 1de 242

Infeccin por flavivirus en aves cinegticas en Espaa:

distribucin del virus y alteraciones estructurales


en los tejidos del hospedador

Memoria presentada por


Virginia Gamino Rodrguez
para optar al grado de Doctor
por la Universidad de Castilla-La Mancha

Bajo la direccin de la Doctora


Ursula Hfle Hansen

Ciudad Real, 2013

GRUPO DE SANIDAD Y BIOTECNOLOGA


INSTITUTO DE INVESTIGACIN EN RECURSOS CINEGTICOS
(CSIC-UCLM-JCCM)
DEPARTAMENTO DE CIENCIA Y TECONOLOGA AGROFORESTAL
Y GENTICA
UNIVERSIDAD DE CASTILLA-LA MANCHA

La Dra. Ursula Hfle Hansen hace constar que la tesis titulada Infeccin por
flavivirus en aves cinegticas en Espaa: distribucin del virus y alteraciones
estructurales en los tejidos del hospedador realizada bajo su direccin por Virginia
Gamino Rodrguez en el Instituto de Investigacin en Recursos Cinegticos, rene
los requisitos necesarios para su defensa y aprobacin y, por tanto, para optar al
grado de Doctor.

VB de la Directora

Dra. Ursula Hfle Hansen

Este trabajo de tesis doctoral se realiz gracias a los siguientes proyectos:


Proyecto AG2008-02504GAN, Ministerio de Ciencia e Innovacin
Proyecto PAC08-0296-7771, Junta de Comunidades de Castilla-La Mancha
Proyecto FAU2008-00006, Instituto Nacional de Investigacin y Tecnologa Agraria y
Alimentaria

ndice

NDICE
ESTRUCTURA DE LA TESIS ............................................................................... 1
INTRODUCCIN .................................................................................................... 3
Antecedentes ......................................................................................................... 5
El gnero Flavivirus ............................................................................................... 6
Flavivirus en Europa y Espaa ........................................................................... 10
Patogenia y desarrollo de encefalitis en la infeccin por flavivirus transmitidos
por mosquitos ..................................................................................................... 26
La perdiz roja y su relacin con los flavivirus ...................................................... 37
Uso de aves como modelo experimental de la infeccin por flavivirus ................. 38
Bibliografa .......................................................................................................... 39

OBJETIVOS ......................................................................................................... 61
CAPTULO 1. Revisin sobre la patologa y el tropismo tisular del virus
West Nile en aves infectadas de forma natural ................................................. 65
Resumen ............................................................................................................. 67
Abstract............................................................................................................... 68
Introduction ........................................................................................................ 69
Etiology ............................................................................................................... 69
Eco-epidemiology ................................................................................................ 71
Pathogenesis in birds .......................................................................................... 74
Pathology of natural WNV infection in birds ........................................................ 80
Discussion and conclusion .................................................................................. 91
Acknowledgements .............................................................................................. 94
References ........................................................................................................... 95

CAPTULO 2. Patogenia de la infeccin experimental con dos aislados


mediterrneos del virus West Nile en perdiz roja ...................................... 105
Resumen ........................................................................................................... 107
Abstract............................................................................................................. 108
Introduction ...................................................................................................... 109
Material and methods ....................................................................................... 110
Results .............................................................................................................. 114
Discussion......................................................................................................... 125
Acknowledgements ............................................................................................ 130
References ......................................................................................................... 131

Virginia Gamino Rodrguez

Tesis doctoral

CAPTULO 3. Infeccin experimental de una especie gallincea


euromediterrnea con una cepa norteamericana del virus West Nile ........ 137
Resumen ........................................................................................................... 139
Abstract............................................................................................................. 140
Introduction ...................................................................................................... 141
Material and methods ....................................................................................... 142
Results .............................................................................................................. 146
Discussion......................................................................................................... 156
Acknowledgements ............................................................................................ 161
References ......................................................................................................... 161

CAPTULO 4. Infeccin natural por el virus Bagaza en aves cinegticas


en el sur de Espaa .................................................................................. 167
Resumen ........................................................................................................... 169
Abstract............................................................................................................. 170
Introduction ...................................................................................................... 171
Material and methods ....................................................................................... 171
Results .............................................................................................................. 175
Discussion......................................................................................................... 183
Acknowledgements ............................................................................................ 186
References ......................................................................................................... 187

CAPTULO 5. Hallazgos patolgicos en ojos de aves gallinceas infectadas


por flavivirus ............................................................................................ 191
Resumen ........................................................................................................... 193
Abstract............................................................................................................. 194
Introduction ...................................................................................................... 195
Material and methods ....................................................................................... 195
Results .............................................................................................................. 196
Discussion......................................................................................................... 200
Acknowledgements ............................................................................................ 201
References ......................................................................................................... 202

CAPTULO 6. Deteccin del virus Usutu en zorzales migratorios en


Espaa .................................................................................................... 203
Resumen ........................................................................................................... 205
Abstract............................................................................................................. 205
Introduction ...................................................................................................... 206
The study .......................................................................................................... 206

ii

ndice

Conclusions ...................................................................................................... 208


Acknowledgements ............................................................................................ 209
References ......................................................................................................... 209

SNTESIS ................................................................................................. 211


Bibliografa ........................................................................................................ 222

CONCLUSIONES ...................................................................................... 225

iii

Lista de abreviaturas

LISTA DE ABREVIATURAS
ARN

cido ribonucleico

BLAST

Basic Local Alignment Search Tool

bp

Base pairs

CD3/79

Cluster of differentiation 3/79

Ct

Cycle threshold

DNA

Deoxyribonucleic acid

ELISA

Enzyme-linked immunosorbent assay

GFAP

Glial fibrillary acidic protein

hr

Hour

IgG

Immunoglobulin G

kb

Kilobase

min

Minute

mL

Milliliter

mM

Millimolar

NK cells

Natural killer cells

No./Nos.

Number/Numbers

PCR

Polymerase chain reaction

PFU

Plaque-forming unit

pmol

Picomol

RCA

Ricinus communis agglutinin

RNA

Ribonucleic acid

RT-PCR

Reverse transcription-polymerase chain reaction

sec

Second

sp./spp.

Species

TNF

Tumoral necrosis factor

Units

UFP

Unidades formadoras de placa

Microgram

Microliter

Micrometer

Estructura de la tesis

ESTRUCTURA DE LA TESIS
La presente tesis doctoral se encuentra organizada de acuerdo al orden habitual
con el que se estructura un trabajo cientfico. La primera parte consiste en una
Introduccin en la que se realiza una sntesis sobre la informacin disponible
relacionada con los flavivirus transmitidos por mosquitos, centrndose en aquellos
que se encuentran circulando en Europa y, ms concretamente, en Espaa, y que
producen mortalidad en aves. A continuacin se enumeran los Objetivos generales
y especficos que se han planteado para realizar ste trabajo y que sern abordados
en cada uno de los seis captulos que componen esta tesis.
En el Captulo 1 se hace una revisin detallada sobre la informacin
disponible de la patogenia y los cambios morfolgicos asociados a la infeccin por el
virus West Nile (WNV) en aves. En los Captulos 2 y 3 se describen las lesiones y la
distribucin del virus durante el curso de la infeccin experimental de la perdiz roja
(Alectoris rufa) con dos aislados mediterrneos y un aislado norteamericano,
respectivamente, de WNV. En el Captulo 4 se describen los cambios morfolgicos y
la distribucin del virus en tejidos de perdiz roja, faisn comn (Phasianus
colchicus) y paloma torcaz (Columba palumbus) infectados de forma natural por el
virus Bagaza (BAGV). En el Captulo 5 se realiza una breve descripcin de las
lesiones y la distribucin del antgeno viral en ojos de aves gallinceas cinegticas
infectadas con WNV y BAGV. Finalmente, el Captulo 6 describe el primer caso de
mortalidad asociado a la infeccin por el virus Usutu en aves en Espaa. Todos
estos captulos estn redactados en ingls y corresponden a artculos cientficos en
diferente estado de publicacin.
Por ltimo, en el apartado de Sntesis se realiza una discusin general sobre
los hallazgos ms importantes obtenidos en cada uno de los trabajos que componen
la tesis doctoral, para terminar con las Conclusiones generales obtenidas de los
mismos.
1

Introduccin

Introduccin

ANTECEDENTES
Debido a la situacin estratgica que presenta Espaa en relacin al paso de aves
migratorias entre Europa y frica, su elevada riqueza en humedales (que sirven
como reas de invernada y nidificacin de muchas aves), su proximidad a frica
(considerada el origen de numerosos flavivirus), y las temperaturas relativamente
suaves que se suelen registrar durante gran parte del ao, fundamentalmente en el
rea mediterrnea (que permiten una amplia actividad de vectores capaces de
transmitir flavivirus), existen las condiciones ptimas para la introduccin de un
flavivirus y el establecimiento de una circulacin local y persistencia del mismo. A
pesar de que la circulacin de flavivirus en Espaa se detect hace ms de
cincuenta aos, ha sido en la ltima dcada cuando la actividad de estos virus ha
incrementado, provocando casos de enfermedad y mortalidad en humanos, caballos
y aves. Una situacin similar se est dando no slo en el resto de Europa, sino
tambin en Norteamrica, especialmente en relacin a uno de los flavivirus ms
distribuidos del mundo, considerado patgeno para las aves silvestres, el virus West
Nile (WNV). La emergencia o re-emergencia de la infeccin por un flavivirus est
asociada a factores no slo relacionados con el propio virus, sino tambin con el
ambiente, los vectores que los transmiten y los hospedadores a los que infectan. La
combinacin de estos factores hace que estos virus no resulten igualmente
patgenos en todas las reas del mundo y que no todos los hospedadores se vean
igualmente afectados por la infeccin. En la presente tesis doctoral se han realizado
una serie de estudios destinados a mejorar el conocimiento sobre las causas de la
emergencia o re-emergencia de la infeccin por estos virus en aves, as como de las
diferencias interespecie en la evolucin y las consecuencias de la infeccin.

Virginia Gamino Rodrguez

Tesis doctoral

EL GNERO FLAVIVIRUS
Etiologa
El gnero Flavivirus, de la familia Flaviviridae, comprende aproximadamente 70
virus encuadrados en doce complejos antignicos (Heinz et al., 2000).
Los flavivirus son pequeos (50nm de dimetro) virus de morfologa
redondeada conformados por una nucleocpside de simetra polidrica rodeada por
una envoltura lipdica (Lindenbach y Rice, 2003) (Figura 1a). Su genoma consiste
en una cadena simple de ARN de polaridad positiva de 11kb de longitud que
configura tres protenas estructurales (cpside (C), premembrana/membrana
(prM/M) y envoltura (E)) y siete no estructurales (NS1, NS2A, NS2B, NS3, NS4A,
NS4B y NS5) (Brinton, 2002; Lindenbach y Rice, 2003) (Figura 1b). La protena E es
la ms importante de las protenas de la superficie de la partcula vrica,
participando en la unin a los receptores celulares y la fusin con la membrana
celular. Esta protena constituye el principal determinante antignico del virus,
siendo,

fundamentalmente

el

dominio

III,

el

objetivo

de

los

anticuerpos

neutralizantes del hospedador (Lindenbach y Rice, 2003; Martn-Acebes y Saiz,


2012). Las protenas no estructurales poseen diferentes funciones que, sobre todo,
son importantes en la replicacin vrica, pero tambin ejercen efectos inhibidores
sobre la respuesta inmune del hospedador (Brinton, 2002; Lindenbach y Rice,
2003; Martn-Acebes y Saiz, 2012). La protena NS5 es la de mayor tamao y la ms
conservada (Lindenbach y Rice, 2003) y su secuencia se utiliza para estudios
filogenticos (Kuno et al., 1998; Gaunt et al., 2001).

Introduccin

Figura
1.
a)
Morfologa
de
los
flavivirus
(Ilustracin
extrada
de
http://flavivirus.wordpress.com/general-flavivirus-info/). b) Organizacin del genoma y
funciones de las protenas virales (Ilustracin adaptada de Fernndez-Garca et al., 2009).

Clasificacin
Los flavivirus pueden ser clasificados en base a su secuencia nucleotdica,
antigenicidad, patogenicidad, distribucin geogrfica o forma de transmisin
(Calisher et al., 1989; Kuno et al., 1998; Gould y Solomon, 2008) (Figura 2). En
base

la

forma

de

transmisin,

hasta

el

ao

2009

se

contabilizaron

aproximadamente 54 arbovirus (39 flavivirus transmitidos por mosquitos y 15 por


garrapatas) y 17 flavivirus sin vector conocido (Mackenzie y Williams, 2009). Dentro
de los flavivirus transmitidos por mosquitos a su vez se pueden distinguir dos
grupos, el de los flavivirus transmitidos por Aedes spp. y el de los transmitidos
por Culex spp. (Gaunt et al., 2001; Kuno y Chang, 2005; Gould y Solomon, 2008)
(Figura 2). Sin embargo, todas estas clasificaciones no son absolutas, pudiendo
existir variaciones y discrepancias.

Virginia Gamino Rodrguez

Tesis doctoral

Figura 2. rbol filogentico del gnero Flavivirus (Familia Flaviviridae). Asociacin en grupos
en base a su hospedador invertebrado, hospedador vertebrado y distribucin geogrfica
(Ilustracin extrada de Gould y Solomon, 2008).

Epidemiologa
Existen evidencias de que los flavivirus se originaron en el Viejo Mundo (Gaunt et
al., 2001; Gould et al., 2003), desde donde se distribuyeron a todos los continentes
excepto la Antrtida (Mackenzie y Williams, 2009). De esta forma, ha sido sugerida

Introduccin

la existencia de un ancestro africano para todos los virus que forman parte del
complejo antignico Encefalitis Japonesa (Mackenzie y Williams, 2009).
La circulacin de los flavivirus transmitidos por garrapatas y por mosquitos
Aedes spp., a excepcin del virus del dengue y de la fiebre amarilla, se restringe al
Viejo Continente, mientras que los transmitidos por mosquitos Culex spp. circulan
tanto en el Viejo como en el Nuevo Continente (Gaunt et al., 2001; Gould et al.,
2003) (Figura 2).
La mayora de los flavivirus transmitidos por mosquitos se caracterizan por
ser virus emergentes, es decir, tienden a establecerse en nuevas reas con relativa
facilidad y frecuencia. Esta propiedad se debe sobre todo a su epidemiologa
multifactorial, en la que influyen factores relacionados con el virus, el hospedador,
el vector o el ambiente (Monath, 1993; Kilpatrick et al., 2008; Pfeffer y Dobler,
2010; Jimnez-Clavero, 2012). Algunos ejemplos de flavivirus emergentes son el
virus Usutu (USUV) y su emergencia en Centroeuropa (Weissenbck et al., 2013),
WNV y su emergencia en Norteamrica (Lanciotti et al., 1999) y el virus Bagaza
(BAGV) y su emergencia en el sur de Europa (Agero et al., 2011).

Ecologa
Los flavivirus transmitidos por mosquitos pueden infectar a una gran variedad de
especies de mosquitos y hospedadores vertebrados. Algunos han sido asociados a
importantes mortalidades en humanos (virus del dengue y de la fiebre amarilla) y
otros, aparentemente, no resultan patgenos ni para el hombre ni para otros
vertebrados (virus Cacipacore) (Weissenbck et al., 2010).
La capacidad para producir enfermedad en humanos es considerada muy
importante, pero tambin los efectos econmicos y/o ecolgicos derivados de la
infeccin en animales.

Virginia Gamino Rodrguez

Tesis doctoral

En general, los flavivirus transmitidos por Aedes spp. se mantienen en un


ciclo en el que participan mosquitos y primates (incluido el hombre), asocindose a
casos de enfermedad hemorrgica (Gaunt et al., 2001) (Figura 2). Sin embargo, los
flavivirus transmitidos por Culex spp., donde se encuadran los pertenecientes a los
complejos antignicos Encefalitis Japonesa y Ntaya, suelen mantenerse en ciclos
enzoticos que implican, sobre todo, a mosquitos ornitfagos y aves (Figura 2).
stos ltimos generalmente circulan de forma silenciosa, pero bajo ciertas
circunstancias pueden dar lugar a brotes en sus hospedadores naturales o ser
transmitidos a hospedadores accidentales, como son los mamferos, y entre ellos el
hombre, dando lugar a epizootias o epidemias (Monath, 1993; Gaunt et al., 2001).
Generalmente, los mosquitos que producen este tipo de transmisin no son los
mismos que mantienen el ciclo enzotico y estos hospedadores accidentales, si bien
pueden sufrir casos de encefalitis grave, son considerados como fondos de saco,
ya que no desarrollan una viremia suficiente que permita la transmisin del
patgeno a mosquitos (Gaunt et al., 2001; Mackenzie y Williams, 2009).

FLAVIVIRUS EN EUROPA Y ESPAA


Los flavivirus transmitidos por mosquitos que circulan en Europa y que
recientemente han provocado brotes en sus hospedadores naturales son WNV,
USUV y BAGV. WNV y USUV adems son capaces de producir enfermedad en
humanos y WNV ha dado lugar a episodios de mortalidad en caballos.
Por otro lado, anualmente se producen casos de enfermedad en humanos
asociados a infecciones por flavivirus adquiridos en los trpicos, como son por
ejemplo el virus del dengue, de la fiebre amarilla, de la encefalitis japonesa y el
virus Chikungunya (Hublek, 2008a; Reiter, 2010; Zeller, 2012).
Dada la situacin estratgica de Espaa en relacin al paso de aves
migratorias entre Europa y frica, y la importancia de nuestros humedales como
10

Introduccin

reas de invernada y nidificacin de muchas de estas aves, en nuestro pas existe


un riesgo elevado de aparicin de brotes asociados a la infeccin por virus
introducidos por aves migratorias. La circulacin de flavivirus en nuestro pas ha
sido demostrada por medio de serologa desde los aos 60, sobre todo en la zona
mediterrnea (Filipe y Andrade, 1990; Figuerola et al., 2007a; Bueno-Mar y
Jimnez-Peydr, 2010). La circulacin de WNV haba sido silenciosa hasta hace
relativamente poco tiempo. Sin embargo, en los ltimos seis aos se han producido,
por primera vez, casos de mortalidad en aves asociados a la infeccin por WNV,
BAGV y USUV (Jimnez-Clavero et al., 2008; Agero et al., 2011; Hfle et al., 2013).

Virus West Nile


El virus West Nile es uno de los flavivirus ms ampliamente distribuidos del
mundo, con una gran importancia tanto en sanidad animal como en salud pblica.
WNV fue aislado por primera vez de la sangre de una mujer con un proceso
febril en Uganda en 1937 (Smithburn et al., 1940) y desde ah pas a ser detectado
en numerosas partes del mundo, donde emergieron nuevos genotipos (Petersen,
2009). Los aislados de WNV se han clasificado filogenticamente en 7 linajes, siendo
los linajes 1 y 2 los ms importantes (Mackenzie y Williams, 2009) (Figura 3). El
linaje 1 contiene las cepas consideradas ms patgenas, que estn ampliamente
distribuidas por Europa, Amrica, Oriente Medio, India, frica y Australia (Lanciotti
et al., 2002). El linaje 2 estaba conformado por cepas que slo circulaban en el sur
de frica y Madagascar pero recientemente ha sido detectado en Europa, donde ha
dado lugar a casos de encefalitis grave en humanos, aves y caballos (Bakonyi et al.,
2006; Bagnarelli et al., 2011; Kutasi et al., 2011; Papa et al., 2011). Antes de la
dcada de los 90, a pesar de que WNV era uno de los flavivirus ms distribuidos en
humanos, aves y mosquitos de frica, Oriente Medio y el suroeste de Europa, slo
ocasionalmente daba lugar a infecciones que generalmente eran subclnicas o poco
11

Virginia Gamino Rodrguez

Tesis doctoral

importantes y que afectaban sobre todo a humanos y caballos (Hublek y Halouzka,


1999; Murgue et al., 2002). Sin embargo, a partir de los aos 90 la frecuencia y
gravedad de las infecciones en humanos increment, as como el nmero de casos
en otros vertebrados, incluyendo animales de compaa, de granja y silvestres (van
der Meulen et al., 2005). En el ao 1999 WNV fue introducido en Norteamrica
(Lanciotti et al., 1999; Nash et al., 2001; Ludwig et al., 2002), donde en el ao 2002
emergi un nuevo genotipo (WN02) con capacidad para diseminarse de forma ms
eficiente, que desplaz al anterior (Davis et al., 2005). Desde entonces han ido
emergiendo nuevos genotipos que han dado lugar a miles de casos de encefalitis en
humanos, caballos y aves (CDC; McMullen et al., 2011).

12

Introduccin

Figura 3. rbol filogentico de WNV (Ilustracin extrada de Vzquez et al., 2011).

WNV pertenece al complejo antignico Encefalitis Japonesa y, por tanto, su


reservorio natural son las aves silvestres (McLean et al., 2001). Las aves migratorias

13

Virginia Gamino Rodrguez

Tesis doctoral

juegan un papel importante en la dispersin local y a larga distancia del virus


(Malkinson et al., 2002a,b; Peterson et al., 2003), habindose demostrado que stas
presentan ttulos de anticuerpos frente al virus ms altos que las aves residentes
(Lpez et al., 2008; Dusek et al., 2009; Valiakos et al., 2012). Existen especies de
anfibios, reptiles y mamferos, entre ellos el hombre, que pueden infectarse y sufrir
encefalitis, sin embargo, en la mayor parte de los casos no desarrollan suficiente
viremia como para ser consideradas especies reservorio (Kostiukov et al., 1985,
1986; Rodhain et al., 1985; Turell et al., 2000; Sardelis et al., 2001; Xiao et al.,
2001; Komar et al., 2003a; Klenk et al., 2004; Root, 2012).
WNV ha sido aislado de numerosas especies de mosquitos, pero slo aquellos
en los que se replica y llega hasta las glndulas salivares para poder ser
transmitido, son vectores competentes (Hublek, 2008a). En Europa, los principales
vectores del virus son Culex pipiens, Cx. modestus y Coquillettidia richiardii
(Hublek y Halouzka, 1999).
Otras vas de transmisin de WNV en aves son el consumo de presas
infectadas o agua contaminada y el contacto con individuos infectados (Komar et
al., 2003b; Nemeth et al., 2006a) (Figura 4). Adems, en humanos el virus se puede
transmitir mediante trasplante de rganos, transfusiones de sangre, consumo de
leche materna, a travs de la placenta y en accidentes de laboratorio (CDC
2002a,b,c; Iwamoto et al., 2003; Pealer et al., 2003).

Contacto

Oral

Figura 4. Ciclo de transmisin de WNV (Elaboracin propia).

14

Introduccin

En reas endmicas, la infeccin en aves suele tener lugar en primavera y a


principios de verano, apareciendo los picos de mortalidad desde mitad del verano
hasta principios de otoo. Los casos en humanos y en caballos, si existen, suelen
producirse algunas semanas despus del inicio de la mortalidad en aves (Phalen y
Dahlhausen, 2004).
La susceptibilidad de las aves a la infeccin vara en funcin de la especie,
por lo que no todas llegan a sufrir enfermedad (Komar et al., 2003b). En Europa,
WNV ha sido detectado en numerosas aves residentes y migratorias, acuticas y
terrestres, pero los casos de mortalidad han sido

espordicos, afectando

fundamentalmente a aves rapaces (Bakonyi et al., 2006; Jimnez-Clavero et al.,


2008; Wodak et al., 2011). Por el contrario, en Norteamrica existen en torno a 300
especies de aves susceptibles (CDC) y la tasa de mortalidad en muchas de ellas es
elevada.

Algunas

poblaciones

como

la

de

cuervos

americanos

(Corvus

brachyrhynchos) han sufrido un grave declive debido a la mortalidad provocada por


este virus (LaDeau et al., 2007; Foppa et al., 2011).
En humanos, slo un 20% de las infecciones son de carcter clnico y, de
stas, menos del 1% son neuroinvasivas. La mayor parte de los casos clnicos se
producen en individuos de edad avanzada o inmunocomprometidos y la mortalidad
se mantiene en torno al 10% (Hayes et al., 2006; Sejvar, 2007). En caballos, slo un
10% de individuos desarrolla enfermedad neurolgica, con una mortalidad que
puede llegar al 50% (Castillo-Olivares y Wood, 2004; Garca-Bocanegra et al.,
2011a).
WNV en Espaa
La presencia de anticuerpos frente a WNV fue descrita por primera vez en
humanos y micromamferos entre los aos 60 y 80 (Filipe y Andrade, 1990) (Tabla
1). Posteriormente, se han seguido detectando anticuerpos en humanos, aves y
caballos, fundamentalmente en la zona mediterrnea (Tabla 1).
15

Virginia Gamino Rodrguez

Tesis doctoral

La mayora de los estudios realizados, sobre todo en aves, se centran en el


suroeste de Espaa debido a que es un rea considerada de alto riesgo como
consecuencia de la existencia de zonas de cra de aves migratorias y poblaciones
abundantes de aves silvestres y vectores competentes, as como por su proximidad
a frica, donde WNV es endmico (Garca-Bocanegra et al., 2011b, 2012a,b). La
circulacin de WNV en esta zona es tan importante que en el ao 2006 se detect
en un pool de Cx. pipiens un posible nuevo linaje del virus (Vzquez et al., 2010)
(Tabla 1).
Los estudios serolgicos realizados en aves adems indican que existe
transmisin vertical de anticuerpos (Figuerola et al., 2007a), que hay circulacin
local de WNV en el sur de Espaa (Figuerola et al., 2007b) y que la seroprevalencia
est relacionada con la actividad migratoria (Lpez et al., 2008) y con el tamao
corporal y la ecologa del ave (Figuerola et al., 2008).
El primer caso de enfermedad asociada a la infeccin por WNV se produjo en
humanos en el ao 2004 (Kaptoul et al., 2007) (Tabla 1). En 2007 tuvo lugar el
primer y nico caso de encefalitis y mortalidad en aves y WNV fue aislado de
muestras de dos guilas reales (Aquila chrysaetos) y un guila perdicera (Aquila
fasciata) (Jimnez-Clavero et al., 2008) (Tabla 1). La cepa aislada mostr una gran
homologa gentica con la aislada en Italia en 1998 (Sotelo et al., 2009) y una muy
similar a sta fue detectada y aislada de mosquitos Cx. perexiguus un ao despus
en la cuenca del Guadalquivir (Vzquez et al., 2011) (Tabla 1). En el ao 2010 se
produjo el primer caso de encefalitis causado por WNV en caballos y, coincidiendo
con ste, dos casos de encefalitis en humanos en el suroeste del pas (GarcaBocanegra et al., 2011a; Jess-De La Calle et al., 2012) (Tabla 1). La cepa que
caus este brote mostr mayor homologa gentica con las aisladas en Italia en
2008 y 2009 que con las aisladas en Espaa en 2007 y 2008 (Sotelo et al., 2011a).

16

Introduccin

En el ao 2011 y 2012 tuvieron lugar nuevos casos de enfermedad en caballos en la


misma zona (RASVE, 2013) (Tabla 1).
Tabla 1. Deteccin cronolgica de la circulacin de WNV y otros flavivirus en Espaa.
Elaboracin propia.
Ao de
muestreo

Especie

1960-1970

Humanos

1973

Humanos

Humanos

1978-1979

Micromamferos

Finales de los
70
-

rea de
muestreo

Tipo de deteccin

Referencia bibliogrfica

Galicia, Castilla y
Len, Asturias
Comunidad
Valenciana
Andaluca

Anticuerpos

Gonzlez y Filipe, 1977

Anticuerpos

Sanchs-Bayarri, 1974

Anticuerpos

Lozano, 1980

Anticuerpos

Chastel et al., 1980

Humanos

Aragn,
Extremadura,
Andaluca
Catalua

Anticuerpos

Lozano y Filipe, 1998

Humanos

Andaluca

Anticuerpos

Pujol et al., 2004

2001

Humanos

Catalua

Anticuerpos

Bofill et al., 2006

2001-2005

guila imperial ibrica


(Aquila adalberti)
Aves

Castilla-La
Mancha
Andaluca

Anticuerpos, genoma
y antgeno viral
Anticuerpos

Hfle et al., 2008

Andaluca

Anticuerpos

2003?

Focha comn (Fulica


atra)
Humanos

Andaluca

Anticuerpos

2003-2011

Jabal (Sus scrofa)

Centro y sur de
Espaa

Anticuerpos

2004

Humanos

Catalua

2004-2006

Aves

Andaluca

Anticuerpos, caso
clnico
Anticuerpos

2005-2008

Caballos

Andaluca

Anticuerpos

2006

Halcn de Eleonora
(Falco eleonorae)
Mosquitos (Cx. pipiens)

Islas Canarias

Anticuerpos

Andaluca

Vzquez et al., 2010

2006-2008

Ciervo (Cervus
elaphus)
Zorro (Vulpes vulpes)

Anticuerpos

2006-2009

Aves

Centro y sur de
Espaa
Centro y suroeste
de Espaa
Andaluca

Genoma viral y
aislamiento
Anticuerpos

2007

guila real
guila perdicera
Urraca (Pica pica)

Castilla-La
Mancha

Gutirrez-Guzmn et al.,
2012
Garca-Bocanegra et al.,
2011b
Jimnez-Clavero et al.,
2008

2008

Mosquitos

Andaluca

2009-2010

Cerdo ibrico

2010

Humanos

Centro y suroeste
de Espaa
Andaluca

2003-2005
2003-2006

2006
2006

Anticuerpos
Anticuerpos, genoma
viral y aislamiento,
caso clnico y
mortalidad
Genoma viral y
aislamiento
Anticuerpos
Anticuerpos, caso
clnico

Figuerola et al., 2007a,


2008
Figuerola et al., 2007b
Bernabeu-Wittel et al.,
2007
Boadella et al., 2012;
Gutirrez-Guzmn et al.,
2012
Kaptoul et al., 2007
Lpez et al., 2008, 2011
Jimnez-Clavero et al.,
2007, 2010
Gangoso et al., 2010

Boadella et al., 2012

Vzquez et al., 2011


Gutirrez-Guzmn et al.,
2012
Garca-Bocanegra et al.,
2011a; Jess-De La Calle
et al., 2012

17

Virginia Gamino Rodrguez

Tesis doctoral

Tabla 1. Deteccin cronolgica de la circulacin de WNV y otros flavivirus en Espaa (cont).


Elaboracin propia.
Ao de
muestreo

Especie

rea de
muestreo

Tipo de deteccin

Referencia bibliogrfica

2010

Mulas y burros

Andaluca

Anticuerpos

2010-2012

Caballos

Andaluca

2011-2012

Perdiz roja (Alectoris rufa)


Faisn comn
(Phasianus colchicus)

Andaluca

Anticuerpos y
genoma viral, caso
clnico y mortalidad
Anticuerpos

Garca-Bocanegra et al.,
2012c
Garca-Bocanegra et al.,
2011a, 2012a,d; RASVE,
2013
Llorente et al., 2013

Estudio de la infeccin por WNV en aves


La infeccin por WNV en aves ha sido fundamentalmente

estudiada en

Norteamrica, puesto que es donde ms casos de mortalidad se han producido.


Existen varios trabajos describiendo las lesiones macroscpicas y microscpicas
asociadas a la infeccin natural, fundamentalmente en crvidos y en rapaces, pero
tambin numerosos trabajos experimentales en los que se han descrito la patogenia
de la infeccin as como la distribucin temporal de WNV en los diferentes tejidos.
En

Europa,

existen menos

trabajos

de esta

naturaleza,

los

que

hay

fundamentalemente se refieren a aves rapaces diurnas (Bakonyi et al., 2006; Hfle


et al., 2008; Wodak et al., 2011; Busquets et al., 2012; Ziegler et al., 2012; Dridi et
al., 2013).
En el captulo 1 de esta tesis profundizaremos en los datos existentes sobre
la dinmica de la infeccin por WNV en aves.

Virus Usutu
El virus Usutu fue aislado por primera vez de mosquitos Culex neavei en Sudfrica
en 1959 (Woodall, 1964) y de otros mosquitos y aves en el continente africano en
las dcadas posteriores (Williams et al., 1964; Adam y Digoutte, 2005). En el ao
1996, el virus lleg a Europa produciendo mortalidad en mirlo comn (Turdus
merula) en Italia (Weissenbck et al., 2013). A partir de ese momento, se sospecha
18

Introduccin

que se estableci una circulacin local entre aves y mosquitos del centro de Europa,
dando lugar a brotes posteriores que han afectado exclusivamente a aves
(Weissenbck et al., 2002, 2003; Bakonyi et al., 2007; Manarolla et al., 2010; Savini
et al., 2011; Steinmetz et al., 2011; Becker et al., 2012) (Tabla 2). La homologa
gentica de las cepas que circulan en el centro de Europa es muy elevada (Chvala et
al., 2007; Steinmetz et al., 2011; Peletto et al., 2012), pero en la Cuenca
Mediterrnea circulan adems otras cepas que probablemente hayan sido
introducidas posteriormente desde frica (Busquets et al., 2008; Savini et al., 2011;
Vzquez et al., 2011; Peletto et al., 2012).
Al igual que WNV, USUV pertenece al complejo antignico Encefalitis
Japonesa, por lo que su reservorio natural son las aves. En Europa, los principales
vectores de USUV pertenecen al gnero Culex (Weissenbck et al., 2007; Tamba et
al., 2011).
Las aves del orden Passeriformes, fundamentalmente el mirlo comn, y las
del orden Strigiformes son las ms susceptibles a la infeccin (Weissenbck et al.,
2002; Bakonyi et al., 2007). Sin embargo, la circulacin del virus ha sido detectada
mediante PCR, aislamiento vrico y serologa en varias especies de aves residentes y
migratorias en diferentes pases europeos (Tabla 2).

19

Virginia Gamino Rodrguez

Tesis doctoral

Tabla 2. Deteccin de USUV en muestras de aves en diferentes pases de Europa. Elaboracin


propia.
Pas

Ao

Tipo de deteccin

Referencia bibliogrfica

Alemania

2000-2005
2011
2000-2006

Anticuerpos
Genoma y antgeno viral, aislamiento
Genoma y antgeno viral

2005-2006
2001-2002
2004
1996
2005
2006-2008
2007
2008-2009
2010
2006

Genoma y antgeno viral, aislamiento


Anticuerpos
Anticuerpos
Genoma y antgeno viral
Anticuerpos
Genoma y antgeno viral
Anticuerpos
Anticuerpos, genoma viral,
aislamiento viral
Genoma viral
Anticuerpos

Linke et al., 2007


Becker et al., 2012
Weissenbck et al., 2002, 2003; Chvala
et al., 2004, 2007; Meister et al., 2008
Bakonyi et al., 2007
Buckley et al., 2003
Buckley et al., 2006
Weissenbck et al., 2013
Rizzoli et al., 2007
Manarolla et al., 2010
Lelli et al., 2008
Savini et al., 2011; Tamba et al., 2011

2005
2011-2012
2006-2007
2009

Anticuerpos
Genoma viral, aislamiento viral
Anticuerpos, genoma y antgeno viral
Genoma y antgeno viral

Austria
Hungra
Inglaterra
Italia

Polonia
Repblica Checa
Suiza

Calzolari et al., 2012


Hublek, 2008b
Hublek, 2008c
Hublek, 2012
Steinmetz et al., 2007, 2011
Steinmetz et al., 2011

La susceptibilidad de los humanos a la infeccin parece ser baja en


comparacin a la que existe frente a otros flavivirus como WNV, sin embargo, USUV
fue aislado de un paciente con fiebre y erupciones en frica (Williams et al., 1964;
Adam y Digoutte, 2005). Adems, en Europa ha sido detectado por PCR en un
paciente en Austria y en pacientes inmunodeprimidos con un proceso neurolgico
en Italia (Weissenbck et al., 2007; Cavrini et al., 2009, 2011; Pecorari et al., 2009),
y por PCR y serologa en donantes de sangre asintomticos en Italia y Alemania
(Gaibani et al., 2010, 2012; Allering et al., 2012).
Se desconoce la susceptibilidad de otros vertebrados a la infeccin, pero en
Italia y Serbia se han detectado anticuerpos frente a USUV en caballos (Lupolovic et
al., 2011; Savini et al., 2011).
USUV en Espaa
La informacin relativa a la circulacin de USUV en Espaa es muy escasa. El virus
se detect por primera vez en un pool de mosquitos Cx. pipiens en Catalua en el
ao 2006 (Busquets et al., 2008) y en un pool de Cx. perexiguus en el sur de
20

Introduccin

Espaa en el ao 2009, mostrando ambas cepas una gran homologa gentica


(Vzquez et al., 2011). En un trabajo realizado en 2013 se ha demostrado la
presencia de anticuerpos frente al virus en una perdiz roja en el sur de Espaa
(Llorente et al., 2013).
Estudio de la infeccin por USUV en aves
Los hallazgos clnicos y los cambios morfolgicos asociados a la infeccin natural
por USUV en aves han sido descritos fundamentalmente en mirlo comn y en
algunas especies de rapaces nocturnas. Los signos clnicos que se pueden observar
son depresin, apata, anorexia, erizamiento de las plumas, incapacidad para volar,
incoordinacin y convulsiones, y muerte rpida, aproximadamente a los tres das
del comienzo de la signologa (Manarolla et al., 2010; Steinmetz et al., 2011; Becker
et al., 2012). Las lesiones macroscpicas sobre todo se detectan en hgado y en bazo
y microscpicamente lo ms caracterstico es la presencia de encefalitis y necrosis e
infiltrados inflamatorios multiorgnicos (Tabla 3).
Existen muy pocos estudios experimentales por lo que se tienen pocos datos
sobre la patogenia de la infeccin por USUV en aves. En un experimento realizado
con pollos de gallina domstica (Gallus domesticus) y en otro realizado con ganso
domstico (Anser anser domesticus) los animales no mostraron signos clnicos y las
lesiones macroscpicas y microscpicas fueron moderadas (Chvala et al., 2005,
2006). En ninguno de los casos se consigui demostrar la presencia del antgeno
viral mediante inmunohistoqumica (IHQ). Se detect material gentico del virus en
tejidos desde el da 2-3 post inoculacin (pi) hasta el da 14-16, viremia en muy
pocos individuos entre los das 5 y 7, y desarrollo anticuerpos el da 10 pi. Ninguna
de las dos especies, por tanto, puede ser considerada como un hospedador
competente o altamente susceptible a la infeccin por USUV. Los resultados
obtenidos en pollos de gallina son muy similares a los descritos en la infeccin
experimental con WNV (Senne et al., 2000). No es as en el caso de ganso
21

Virginia Gamino Rodrguez

Tesis doctoral

domstico, en el que la infeccin experimental con WNV dio lugar a una encefalitis
fatal, demostrando que esta especie es ms susceptible a la infeccin por WNV que
por USUV (Swayne et al., 2001).

22

Weissenbck et al., 2003; Chvala et


al., 2004; Manarolla et al., 2010;
Steinmetz et al., 2011

Clulas glomerulares y epiteliales


tubulares del rin

Fibrocitos y clulas musculares de la


cpsula del bazo, macrfagos y
clulas dendrticas
Clulas inflamatorias
intravasculares, clulas del ovario

Hepatitis, necrosis coagulativa


heptica y hemorragias

Nefritis y degeneracin de clulas


epiteliales tubulares

Necrosis coagulativa esplnica,


hiperplasia folicular y de la pulpa
roja

Enteritis seromucosa

Hepatomegalia, congestin heptica y


focos hepticos amarillentos

Nefromegalia

Esplenomegalia

No descritas

Sistema gastrointestinal

Sistema endocrino

Sistema urinario

Sistema inmunitario

Otros

Leucocitolisis

Weissenbck et al., 2002, 2003;


Chvala et al., 2004; Bakonyi et al.,
2007; Manarolla et al., 2010;
Steinmetz et al., 2011; Becker et al.,
2012; Calzolari et al., 2012

Clulas de Kupffer en hgado

Proventriculitis y abscesos en criptas


intestinales

Weissenbck et al., 2002; Chvala et


al., 2004; Bakonyi et al., 2007;
Manarolla et al., 2010; Steinmetz et
al., 2011; Weissenbck et al., 2013

Weissenbck et al., 2002, 2003;


Chvala et al., 2004; Bakonyi et al.,
2007; Manarolla et al., 2010;
Steinmetz et al., 2011; Becker et al.,
2012

Weissenbck et al., 2002; Chvala et


al., 2004; Bakonyi et al., 2007;
Manarolla et al., 2010

Ganglios autnomos, clulas


glandulares y de la tnica muscular
de proventrculo y molleja, y clulas
epiteliales de las criptas y tnica
muscular de intestino

Miocarditis y miocitolisis

No descritas

Sistema cardiovascular

Weissenbck et al., 2003; Chvala et


al., 2004; Bakonyi et al., 2007;
Manarolla et al., 2010; Steinmetz et
al., 2011; Becker et al., 2012

Miofibrillas y clulas intersticiales del


corazn, clulas musculares y
endoteliales de la pared de grandes
vasos

Neumona y edema

Congestin, edema y hemorragia


pulmonar

Sistema respiratorio

Weissenbck et al., 2002, 2003;


Chvala et al., 2004; Bakonyi et al.,
2007; Manarolla et al., 2010;
Steinmetz et al., 2011; Becker et al.,
2012; Weissenbck et al., 2013
Weissenbck et al., 2002; Chvala et
al., 2004; Manarolla et al., 2010;
Steinmetz et al., 2011; Becker et al.,
2012

Neuronas, clulas de la gla y clulas


inflamatorias

Neuronofagia, gliosis, manguitos


perivasculares, satelitosis,
degeneracin y necrosis neuronal y
edema

Hiperemia en meninges y
parnquima cerebral

Sistema nervioso central

REFERENCIA BIBLIOGRFICA

Clulas parenquimatosas del pulmn

ANTGENO VIRAL

LESIONES MICROSCPICAS

LESIONES MACROSCPICAS

SISTEMA ORGNICO

Tabla 3. Infeccin natural por USUV en aves. Lesiones macroscpicas, microscpicas y distribucin del antgeno viral (IHQ) en los diferentes sistemas
orgnicos del hospedador. Elaboracin propia.

Introduccin

23

Virginia Gamino Rodrguez

Tesis doctoral

Virus Bagaza
El virus Bagaza es un flavivirus relativamente desconocido. Fue aislado por primera
vez de mosquitos del gnero Culex en la Repblica Centroafricana en 1966
(Digoutte, 1978) y despus detectado en mosquitos en otros pases de frica y la
India (Traore-Lamizana et al., 1994; Diallo et al., 2005; Bondre et al., 2009).
BAGV pertenece al grupo antignico Ntaya (Calisher et al., 1989), donde se
encuentran otros flavivirus como el de la meningoencefalitis del pavo (ITMV), que es
considerado sinnimo de BAGV (Kuno et al., 1998). ITMV fue descrito por primera
vez en Israel en 1960 (Komarov y Kalmar, 1960) y es un virus de alta patogenicidad
para aves domsticas, fundamentalmente pavos (Meleagris gallopavo), causando un
proceso neurolgico muy similar al encontrado en la infeccin por otros flavivirus
(Komarov y Kalmar, 1960; Barnard et al., 1980). Este flavivirus slo ha sido
detectado en Oriente Medio y el sur de frica (Kuno et al., 1998) y aparentemente
no es patgeno para humanos (Guy y Malkinson, 2008).
Se desconoce cul es el reservorio de BAGV. Algunos estudios han indicado
que son las aves y que se transmite fundamentalmente por mosquitos del gnero
Culex, al igual que WNV y USUV (Gaunt et al, 2001). Hasta hace relativamente poco
se desconoca la suceptibilidad de las aves a la infeccin por BAGV, pero al ser
genticamente muy similar a ITMV se especulaba que podra causar enfermedad en
el hospedador aviar (Kuno y Chang, 2007).
La suceptibilidad de los humanos est poco caracterizada, sin embargo, en el
ao 1996 se detectaron anticuerpos frente al virus en pacientes con encefalitis en la
India (Bondre et al., 2009) y Woolhouse y cols. (2006) identificaron a BAGV como
un patgeno emergente o re-emergente capaz de producir enfermedad en humanos.
BAGV en Espaa
El primer caso de mortalidad asociado a la infeccin por BAGV en aves se produjo
en el suroeste de Espaa en el verano de 2010 (Agero et al., 2011). BAGV afect
24

Introduccin

fundamentalmente a perdiz roja (Alectoris rufa) y faisn comn (Phasianus


colchicus), con una mayor mortalidad en la primera (Agero et al., 2011; GarcaBocanegra et al., 2012b).
El modo en el que BAGV lleg a Espaa se desconoce, sin embargo, su
secuencia gentica mostr una gran homologa con la cepa aislada en frica y en
menor medida con la detectada en la India (Agero et al., 2011), por lo que se
baraja la posibilidad de su introduccin por aves migratorias desde frica (Agero
et al., 2011; Garca-Bocanegra et al., 2012b). La elevada morbilidad de la infeccin
pudo estar relacionada con la abundancia de vectores competentes y la alta
densidad de aves en la zona (Garca-Bocanegra et al., 2011a, 2012b).
Estudio de la infeccin por BAGV en aves
Existen muy pocos trabajos en los que se describan las lesiones asociadas a la
infeccin por BAGV en aves. En general, los signos clnicos y las lesiones
macroscpicas y microscpicas encontradas son similares a los provocados por la
infeccin por ITMV (Komarov y Kalmar, 1960; Barnard et al., 1980). Las aves
infectadas en el brote de Espaa mostraron debilidad, postracin, incoordinacin,
prdida de peso y diarrea blanquecina (Agero et al., 2011; Garca-Bocanegra et al.,
2012b). Las lesiones macroscpicas principales fueron caquexia y congestin
generalizada y dentro de las microscpicas lo ms caracterstico fue la presencia de
meningoencefalitis y neuritis (Garca-Bocanegra et al., 2012b).
En el captulo 4 de esta tesis se darn ms datos sobre la infeccin natural
por este virus en aves.

25

Virginia Gamino Rodrguez

Tesis doctoral

PATOGENIA Y DESARROLLO DE ENCEFALITIS EN LA INFECCIN POR


FLAVIVIRUS TRANSMITIDOS POR MOSQUITOS
La patogenia de la infeccin tras la inoculacin de un flavivirus por un mosquito
infectado en un hospedador vertebrado suele seguir el mismo esquema (Figura 5).
En mamferos, una vez que el virus es inoculado en la piel, y tras una posible
replicacin en los queratinocitos (Lim PY et al., 2011), ste es transportado por
clulas dendrticas o clulas de Langerhans desde la zona de inoculacin hacia los
linfonodos regionales (Johnston et al., 2000) (Figura 5). Aqu, se produce la
replicacin inicial del flavivirus (McMinn et al., 1996) ante la cual el hospedador
desarrolla una respuesta inmune inmediata en la que intervienen, entre otros,
mediadores como el interfern (IFN) tipo I y clulas como los macrfagos (Chambers
y Diamond, 2003; Arjona y Fikrig, 2009; Lim SM et al., 2011) (Figura 5). Diversos
trabajos han indicado que la produccin de IFN por las clulas dendrticas de la piel
es la primera lnea de defensa (Libraty et al., 2001) y que los macrfagos son clulas
diana de algunos flavivirus, jugando un papel importante en la patogenia de la
enfermedad (Steele et al., 1999; Chambers y Diamond, 2003; Garca-Tapia et al.,
2006). Desde los linfonodos regionales el virus pasa a la circulacin sangunea, a
travs de la cual llega a diferentes rganos, establecindose una infeccin sistmica
y producindose una segunda replicacin vrica (Figura 5). Los flavivirus muestran
un amplio tropismo celular y tisular pudiendo infectar clulas epiteliales,
parenquimatosas, endoteliales y musculares, as como fibroblastos (Chambers y
Diamond, 2003; Weingartl et al., 2004). Ante la infeccin sistmica, el hospedador
desarrolla una respuesta inmune en la que intervienen, adems de otros efectores,
linfocitos B y T que reaccionan frente a diferentes determinantes antignicos del
virus, como la protenas E y NS1, y que tratan de frenar la replicacin vrica
(Chambers y Diamond, 2003; Samuel y Diamond, 2006) (Figura 5). La informacin
sobre la fase inicial de la patogenia en aves es muy limitada, pero se sospecha que
26

Introduccin

tras una amplificacin local en la zona de inoculacin se produce una rpida


viremia e infeccin sistmica (Nemeth et al., 2011, para ms detalles ver captulo 1).
Los flavivirus han desarrollado numerosos mecanismos que les permiten evadir la
respuesta inmune del hospedador (Ye et al., 2013), por lo que en funcin del nivel
de replicacin perifrica y la efectividad de la respuesta inmune y, por tanto, del
nivel de viremia, stos pueden llegar a alcanzar el sistema nervioso central (SNC)
dando lugar a encefalitis (Albrecht, 1968; McMinn et al., 1996; Diamond et al.,
2003; Chambers y Diamond, 2003; Samuel y Diamond, 2006; Lim SM et al., 2011)
(Figura 5).

27

Virginia Gamino Rodrguez

Tesis doctoral

Figura 5. Patogenia de la infeccin por un flavivirus transmitido por mosquitos en un


hospedador mamfero (Ilustracin modificada de Suthar et al., 2013).

28

Introduccin

En el caso de WNV, la entrada va oral o inhalatoria han sido sugeridas en


humanos y han sido demostradas en animales infectados de forma natural y
experimental (Nir et al., 1959, 1965; Odelola y Oduye, 1977; Garmendia et al.,
2000; Kuno, 2001; CDC 2002a, 2003; Komar et al., 2003b; Miller et al., 2003;
Austgen et al., 2004; Klenk et al., 2004). Sin embargo, debido al desconocimiento
de la importancia epidemiolgica de estos tipos de transmisin, existen muy pocos
trabajos en los que se haya estudiado la patogenia. En un experimento realizado en
el ao 1965, la exposicin de ratones a aerosoles contaminados con WNV provoc la
entrada del virus al cerebro a travs de los bulbos olfatorios sin necesidad de
viremia previa, as como la replicacin inicial del virus en pulmn y diseminacin
posterior a sangre, bazo y otros tejidos (Nir et al., 1965). Por otro lado, la infeccin
experimental por va oral en ratones permiti detectar el virus en cerebro dos das
antes que en sangre, por lo que, de nuevo, se asumi que el virus accedi al SNC a
travs de los bulbos olfatorios sin necesidad de una replicacin perifrica previa
(Odelola y Oduye, 1977). En general, tanto en aves como en otros hospedadores, la
infeccin por va oral provoca un retraso en la aparicin de viremia, aunque sta
suele alcanzar los mismos niveles que en el caso de la transmisin por la picadura
de un mosquito, las lesiones suelen ser ms moderadas y en muchos casos los
individuos infectados no muestran signos clnicos ni existe mortalidad (Komar et
al., 2003b; Klenk et al., 2004; Sbrana et al., 2005; Nemeth et al., 2006a,b;
Steinman et al., 2006).
La patogenia de la infeccin por un flavivirus est influenciada por factores
relacionados con el virus y el hospedador. Existen numerosos determinantes
genticos del virus que modulan su interaccin con el hospedador y una simple
mutacin puede dar lugar a un cambio en su virulencia, ya sea por una
modificacin en la capacidad de replicacin, en la susceptibilidad a la respuesta
inmune del hospedador, o en la sensibilidad a la temperatura corporal del mismo
29

Virginia Gamino Rodrguez

Tesis doctoral

(Brinton et al., 2002; Kinney et al., 2006; Ye et al., 2013). En animales infectados
experimentalmente se ha demostrado que la existencia de cambios en las protenas
virales como la protena E, la NS3 o la NS4b pueden modificar la virulencia de un
flavivirus (Beasley et al., 2005; Wicker et al., 2006; Brault et al., 2007). As por
ejemplo, la glicosilacin de la protena E de WNV incrementa la replicacin
perifrica en pollos infectados de forma experimental y finalmente su virulencia
(Murata et al., 2010; Totani et al., 2011) y la sustitucin del aminocido treonina
por prolina en la posicin 249 de la protena NS3 incrementa la virulencia de WNV
en la infeccin experimental de cuervos americanos (Brault et al., 2004, 2007). El
estado inmunitario del hospedador es el factor ms importante del que depende la
susceptibilidad a la infeccin (Solomon y Winter, 2004). La capacidad de respuesta
inmune est a su vez condicionada por numerosos factores como la existencia de
inmunidad previa frente al virus o a flavivirus relacionados antignicamente (Tesh
et al., 2002; Fang y Reisen, 2006), la presencia de enfermedades concomitantes
(Murray et al., 2006) y la edad (Eldadah et al., 1967). De la edad del hospedador
depende la expresin de receptores celulares a los que se pueda unir el virus o de
factores intracelulares que participan en la patogenia de la infeccin (Chambers y
Diamond, 2003). Numerosos autores han indicado que la susceptibilidad a la
infeccin tambin tiene componentes hereditarios (Chambers y Diamond, 2003;
Bigham et al., 2011; Clark et al., 2012).
En funcin de la combinacin de estos factores la infeccin por un flavivirus
puede desencadenar tres cuadros clnicos diferentes (Monath, 1986):
a) Infeccin inaparente, con una viremia transitoria, replicacin perifrica
limitada y ausencia de neuroinvasin.
b) Encefalitis subclnica, con una viremia moderada, establecimiento tardo de
infeccin en el SNC y eliminacin del virus con un desarrollo mnimo de
lesiones.
30

Introduccin

c) Encefalitis severa, con una viremia temprana y de gran magnitud, invasin


del SNC y desarrollo de lesiones severas.

Desarrollo de encefalitis asociada a la infeccin por un flavivirus


La mayora de los trabajos publicados describen el desarrollo de encefalitis asociada
a la infeccin por flavivirus en roedores y humanos, y fundamentalmente se centran
en la infeccin por WNV (revisin en McMinn, 1997; Chambers y Diamond, 2003;
Lim SM et al., 2011; Cho y Diamond, 2012; Clark et al., 2012).
La habilidad de un flavivirus para provocar un proceso neurolgico depende
de su neuroinvasividad, es decir, la capacidad para replicarse perifricamente
alcanzando suficiente viremia para invadir el SNC, y de su neurovirulencia, o
capacidad para iniciar una infeccin citoptica en el SNC y provocar encefalitis
(Monath, 1986; McMinn, 1997). La mayora de las infecciones del SNC se producen
por va hematgena (Chambers y Diamond, 2003; Turtle et al., 2012), pero debido a
las caractersticas estructurales de la barrera hematoenceflica (BHE) (Abbott et al.,
2006, 2010) (Figura 6), el paso del virus a travs de la misma requiere mecanismos
especiales como pueden ser:
a) Infeccin y replicacin del virus en las clulas endoteliales de la BHE (Liou et
al., 1998; Verma et al., 2009).
b) Disrupcin de la BHE por medio de factores que alteren su permeabilidad (en
ratones, la produccin perifrica de TNF- en respuesta a la infeccin
experimental con WNV altera las uniones estrechas entre las clulas
endoteliales de la BHE permitiendo el paso de partculas vricas y clulas
inflamatorias (Wang et al., 2004)).

31

Virginia Gamino Rodrguez

Tesis doctoral

Figura 6. Estructura de la BHE. La BHE es una barrera fsica conformada por clulas
endoteliales conectadas por uniones estrechas que hacen que sta acte como una barrera
selectiva, forzando el paso trascelular de la mayora de molculas. Rodeando a las clulas
endoteliales se sitan pericitos y una lmina basal (BL1). El parnquima cerebral a su vez est
rodeado por otra lmina basal (BL2) tras la cual se sita la capa limitante de gla, formada por un
entramado constituido por las porciones finales de los astrocitos y de algunas clulas de la
microgla. En algunos vasos sanguneos existe un espacio perivascular (Virchow-Robin) entre BL1
y BL2 (Ilustracin extrada de Abbot et al., 2010).

Existen otros mecanismos de acceso al SNC como son la difusin pasiva del
virus a travs de las clulas endoteliales del plexo coroideo (Kramer-Hmmerle et
al., 2005), la infeccin de neuronas olfatorias tras la invasin del epitelio olfatorio
(McMinn, 1997), el transporte axonal retrgrado desde neuronas motoras infectadas
perifricamente (Samuel et al., 2007a) o el transporte por medio de clulas
inflamatorias infectadas, proceso que se conoce con el nombre de caballo de Troya
(Garca-Tapia et al., 2006). El mecanismo de neuroinvasin depende de la
virulencia del virus y de la va de entrada del mismo en el hospedador (Beasley et
al., 2002).
Las principales clulas diana de los flavivirus en el SNC son las neuronas
(Desai et al., 1995; Shieh et al., 2000; Steele et al., 2000; Xiao et al., 2001;
Shrestha et al., 2003), pudiendo producir su muerte por necrosis o por apoptosis
(Xiao et al., 2001; Shrestha et al., 2003; Weissenbck et al., 2004; Samuel et al.,
2007b). El predominio de una u otra forma de muerte celular ha sido relacionado
con la virulencia y la dosis infectiva del virus en ratones y cultivos celulares (Xiao et
32

Introduccin

al., 2001; Chu y Ng, 2003). En ocasiones, las neuronas que estn muriendo como
consecuencia de la infeccin liberan citoquinas txicas que daan otras neuronas
que no estn infectadas (Shrestha et al., 2003; Kumar et al., 2010). Los flavivirus
adems pueden infectar clulas de la gla in vivo e in vitro y aunque las
consecuencias de este tropismo estn menos estudiadas, algunos trabajos in vitro
sugieren su papel en el mantenimiento de la infeccin (Jordan et al., 2000; Steele et
al., 2000; Diniz et al., 2006; Thongtan et al., 2010).
En el momento en el que el virus infecta las neuronas se inicia la respuesta
inmune del hospedador (Cho y Diamond, 2012). El SNC dispone de una serie de
componentes celulares que le permiten responder a estmulos de forma inmediata:
a) Clulas de la microgla: consideradas como los macrfagos residentes del
SNC, con capacidad para fagocitar y presentar antgenos a las clulas T y
producir citoquinas (Gehrmann et al., 1995; Kettenmann et al., 2011).
b) Astrocitos: su funcin bsica es el mantenimiento de la homeostasis para
una correcta actividad neuronal (Montgomery, 1994; Magistretti y Ransom,
2002). Su papel en la inflamacin es menos importante que el de la microgla
pero pueden actuar como clulas fagocitarias y presentadoras de antgeno,
as como liberar molculas que intervienen en el proceso inflamatorio
(Montgomery, 1994).
c) Otras clulas: en mamferos se ha visto que existen otras poblaciones
celulares en menor proporcin como son macrfagos y clulas dendrticas
perivasculares, coroideas y menngeas, adems de linfocitos T, B y monocitos
en el lquido cefalorraqudeo (LCR) (Ousman y Kubes, 2012; Ransohoff y
Engelhardt, 2012).
Por tanto, la primera lnea de defensa del SNC frente a la infeccin por un
flavivirus es la activacin de la microgla y los astrocitos, la expresin de molculas
de adhesin, y la produccin de mediadores de la inflamacin que terminar en el
33

Virginia Gamino Rodrguez

Tesis doctoral

reclutamiento de clulas inflamatorias desde la periferia (Chambers y Diamond,


2003; Garca-Tapia et al., 2007; Rezai-Zadeh et al., 2009; Cho y Diamond, 2012;
Ousman

Kubes,

2012).

El

SNC

es

considerado

como

un

lugar

inmunoprivilegiado, entre otras razones debido a que las caractersticas de la BHE


hacen que el acceso de clulas inflamatorias perifricas sea mucho menor que en el
caso de otros tejidos (Muldoon et al., 2013). No obstante, existen tres vas por las
que estas clulas pueden acceder al SNC (Ransohoff et al., 2003):
1. De la sangre al LCR a travs del plexo coroideo, considerndose una va de
entrada en condiciones fisiolgicas (Figura 7).
2. De la sangre al espacio subaracnoideo y de ah al espacio perivascular de
Virchow-Robin (Figura 8).
3. De la sangre directamente al espacio perivascular intraparenquimatoso. En
este caso requieren atravesar la BHE y la lmina basal endotelial, por lo que
slo se va a producir en situaciones patolgicas en las que se altere su
permeabilidad (Wang et al., 2004).

Figura 7. Entrada de leucocitos en el LCR.


Las clulas inflamatorias se unen a citoquinas
expresadas en el endotelio fenestrado y entran
por diapdesis en el estroma del plexo
coroideo. Una vez aqu los leucocitos se
pueden unir a otras citoquinas expresadas por
las clulas epiteliales del plexo coroideo y
migrar hacia el LCR (Ilustracin extrada de
Ransohoff et al., 2003).

34

Introduccin

Figura 8. Entrada de leucocitos en el espacio subaracnoideo. La sangre llega desde las ramas
finales de la arteria cartida interna al espacio subaracnoideo. Cuando entran en el parnquima
del cerebro, los vasos estn rodeados inicialmente por un espacio perivascular (Virchow-Robin)
que a su vez conecta con el espacio subaracnoideo (Ilustracin extrada de Ransohoff y
Engelhardt, 2012).

En la encefalitis asociada a la infeccin por un flavivirus participan sobre


todo linfocitos T CD4+ y CD8+ pero tambin linfocitos B, neutrfilos y monocitos
(Sampson et al., 2001; Kelley et al., 2003; Brhin et al., 2008; Rezai-Zadeh et al.,
2009). Estas clulas suelen ser retenidas en el espacio perivascular, donde se
preparan para reaccionar frente al virus, atravesando posteriormente la capa
limitante de gla para penetrar en el parnquima nervioso (Bechmann et al., 2007).
La magnitud y el tipo de respuesta inflamatoria en el SNC depende de la
virulencia del virus, la va de infeccin y el estado inmunitario del hospedador
(Chambers y Diamond, 2003). En ocasiones la respuesta es tan intensa que puede
tener efectos perjudiciales sobre la integridad y la funcionalidad de los componentes
del SNC (bystander injury) (Shrestha et al., 2003; Brhin et al., 2008; Wee Yong et
al., 2010; Zhang et al., 2010). La importancia del papel que juega en el desarrollo
de enfermedad neurolgica la muerte neuronal por accin directa del virus o
secundaria a la inflamacin no est demasiado clara (Turtle et al., 2012). En
ratones se ha visto que los linfocitos T CD8+ y las clulas productoras de
anticuerpos son importantes para la recuperacin de la encefalitis, pudiendo

35

Virginia Gamino Rodrguez

Tesis doctoral

permanecer en el SNC durante varias semanas (Wang et al., 2003; Stewart et al.,
2011).
El virus puede persistir en el SNC de humanos, aves, roedores y otros
mamferos infectados de forma natural o experimental durante varias semanas o
meses, dando lugar o no a secuelas neurolgicas (Pogodina et al., 1983; Ravi et al.,
1993; Xiao et al., 2001; Penn et al., 2006; Nemeth et al., 2009a,c; Appler et al.,
2010; Sadek et al., 2010; Wheeler et al., 2012).
Existen diversos factores vricos que van a modular la neuroinvasividad y la
neurovirulencia. La protena E es considerada el determinante antignico ms
importante, ya que es la que interacciona con los receptores celulares, pero existen
regiones no estructurales y no codificantes en el genoma viral que tambin ejercen
un efecto modulador (Monath y Heinz, 1996; Clark et al., 2012). La glicosilacin de
la protena E de WNV ha sido asociada a un incremento en la neuroinvasividad en
ratones (Shirato et al., 2004; Beasley et al., 2005), en parte porque los virus con
gran habilidad para replicarse perifricamente en el hospedador presentan una
mayor capacidad neuroinvasiva (Huang y Wong, 1963; Albrecht, 1968; Shirato et
al., 2004). Existen adems diferentes factores genticos en humanos, aves y
roedores que han sido relacionados con la susceptibilidad a sufrir encefalitis en la
infeccin por estos virus (Monath, 1986; Sangster et al., 1994; Chambers y
Diamond, 2003; Clark et al., 2012; Tag-El-Din-Hassan, 2012).
A pesar de los numerosos estudios experimentales realizados en aves, la
informacin sobre el mecanismo de entrada de los flavivirus en el SNC as como los
mediadores y componentes celulares que intervienen en el desarrollo de encefalitis y
muerte neuronal es muy escasa. La BHE en aves es considerada similar a la de
mamferos (Stewart y Wiley, 1981). La presencia de vasculitis, hemorragias y
manguitos perivasculares en algunas especies podra relacionarse con la llegada del
virus va hematgena. De igual forma, la deteccin del antgeno viral en clulas
36

Introduccin

endoteliales del SNC podra indicar una infeccin y replicacin del virus en las
mismas (Wnschmann et al., 2004; Lopes et al., 2007; Nemeth et al., 2011),
aunque la llegada por medio de clulas del sistema inmune infectadas no se
descarta, ya que en numerosas ocasiones se detecta el antgeno viral en stas antes
que en ninguna otra clula del SNC (Steele et al., 2000; Weingartl et al., 2004;
Gibbs et al., 2005).
Las clulas inflamatorias que intervienen en el desarrollo de encefalitis
asociada a la infeccin por un flavivirus en aves tampoco estn caracterizadas, sin
embargo, es probable que sean muy similares a las encontradas en mamferos. En
la encefalitis asociada a la infeccin por otros virus neurotrpicos, adems de
participar astrocitos y clulas de la microgla, se produce una infiltracin de
linfocitos T y B y macrfagos (Ecco et al., 2011; Brjer et al., 2012).

LA PERDIZ ROJA Y SU RELACIN CON LOS FLAVIVIRUS


La perdiz roja es una ave galliforme mediterrnea cuya poblacin se distribuye de
forma natural en el noroeste de Italia, centro y sur de Francia y la Pennsula
Ibrica, y que ha sido introducida en el sur de Inglaterra y las islas Canarias
(Blanco-Aguiar et al., 2003). Ms del 50% de dicha poblacin se encuentra en la
Pennsula Ibrica (Aebischer y Lucio, 1997), dnde presenta un gran valor
socioeconmico y ecolgico, ya que es una de las especies de caza menor ms
importante (Bernabeu, 2000) y un recurso trfico de una gran variedad de especies,
alguna de ellas amenazada como el guila imperial ibrica (Caldern, 1975; BlancoAguiar, 2007). La perdiz roja ha sufrido un declive en el 95% de sus reas de
distribucin (Aebischer y Potts, 1994; Aebischer y Lucio, 1997). La prdida del
hbitat, la introduccin de especies alctonas y la sobreexplotacin de recursos
parecen

estar

relacionadas

con

este

hecho

(Blanco-Aguiar,

2007).

Como

consecuencia de la elevada demanda cinegtica y el declive de sus poblaciones, esta


37

Virginia Gamino Rodrguez

Tesis doctoral

especie es producida en granjas cinegticas de manera intensiva y al aire libre


(Aebischer y Potts, 1994), y las altas densidades que se alcanzan en estas
instalaciones pueden favorecer o incrementar el riesgo de transmisin o aparicin
de enfermedades (Blanco-Aguiar, 2007). Esto, unido al hecho del bajo o nulo
control sanitario de las repoblaciones (Gortzar, 1998), puede dar lugar a graves
problemas sanitarios, no slo para esta especie sino para muchas otras que estn
presentes en su entorno o que tengan relaciones ecolgicas con la misma.
En general las aves galliformes son consideradas poco susceptibles a la
infeccin por WNV (Guy y Malkinson, 2008; van der Meulen et al., 2005). La
mayora no muestra signos clnicos ni muere a causa de la enfermedad, si bien
desarrolla viremia y produce anticuerpos (Senne et al., 2000; Swayne et al., 2000;
Langevin et al., 2001; Komar et al., 2003b). Sin embargo, en Norteamrica se
produjeron sendos brotes de elevada mortalidad en granjas de perdiz chukar
(Alectoris chukar) y faisanes imperiales (Lophophorus impeyanus) (Wnschmann y
Ziegler, 2006) y la perdiz roja demostr ser susceptible a la infeccin experimental
con cepas mediterrneas de WNV, mostrando signos clnicos y mortalidad (Sotelo et
al., 2011b). Por otro lado, y como se ha mencionado anteriormente, tanto la perdiz
roja como el faisn comn son susceptibles a la infeccin natural por BAGV,
sucumbiendo incluso a la enfermedad, pero se desconoce su susceptibilidad a la
infeccin por USUV.

USO DE AVES COMO MODELO EXPERIMENTAL DE LA INFECCIN POR


FLAVIVIRUS
Como ya ha sido mencionado anteriormente, en el estudio experimental de
enfermedades infecciosas que afectan a humanos, entre ellas la infeccin por
flavivirus, se emplean fundamentalmente roedores. Estos modelos experimentales,
aunque con ciertas diferencias, presentan un sistema inmune similar al humano,
38

Introduccin

permitiendo emular de forma ms o menos fiable la patogenia de enfermedades que


afectan al hombre (Buer y Balling, 2003).
En relacin a los flavivirus, los ratones y hmsteres han sido ampliamente
utilizados para estudios de virulencia y tropismo viral, identificacin de factores
vricos y del hospedador que intervienen en la patogenia, as como en tests de
eficacia y seguridad de vacunas y tratamientos teraputicos (Clark et al., 2012). Sin
embargo, cuando se desea realizar estudios relacionados con la patogenia de la
infeccin en aves, reservorio natural de la mayora de los flavivirus transmitidos por
mosquitos, la utilidad de los roedores disminuye. Ciertas aves galliformes,
fundamentalmente los pollos de gallina domstica, han sido utilizadas para
estudios experimentales desde hace ms de 100 aos ya que, al igual que los
roedores, son fciles de manejar y suponen un bajo coste de adquisicin,
mantenimiento y alimentacin (Davis, 2003). La perdiz roja adems rene las
caractersticas de ser una especie autctona, distribuida en los ambientes
mediterrneos donde pueden persistir y amplificarse diferentes flavivirus, ser
fcilmente manejable en cautividad y tener un gran potencial como especie
centinela debido a la amplia distribucin de granjas cinegticas en las zonas de
inters. Por ello, los trabajos experimentales incluidos en esta tesis doctoral se han
desarrollado en esta especie.

BIBLIOGRAFA
Abbott NJ, Ronnback L, Hansson E. Astrocyte-endothelial interactions at the
blood-brain barrier. Nat Rev Neurosci 2006, 7(1):41-53.
Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. Structure and
function of the blood-brain barrier. Neurobiol Dis 2010, 37(1):13-25.
Adam F, Digoutte JP: Virus dAfrique (base de donnes). Viruses of Africa
(database). Dakar: World Health Organization Collaborating Reference and
Research Centre for arboviruses and haemorrhagic fever viruses (CRORA),
Institut
Pasteur
de
Dakar
2005.
[http://www.pasteur.fr/recherche/
banques/CRORA/]
39

Virginia Gamino Rodrguez

Tesis doctoral

Aebischer NJ, Potts GR: Red-legged partridge. In: Birds in Europe, their
conservation status. Edited by Tucker GM, Heat MF. Cambridge: BirdLife
International; 1994:214-215.
Aebischer NJ, Lucio AJ: Red-legged Partridge Alectoris rufa. In: The EBCC atlas
of European breeding birds: Their distribution and abundance. Edited by
Hagemeijer WJM, Blair MJ. London: T & AD Poyser; 1997:208-209.
Agero M, Fernndez-Pinero J, Buitrago D, Snchez A, Elizalde M, San Miguel E,
Villalba R, Llorente F, Jimnez-Clavero MA. Bagaza virus in partridges and
pheasants, Spain, 2010. Emerg Infect Dis 2011, 17(8):1498-1501.
Albrecht P. Pathogenesis of neurotropic arbovirus infections. Curr Top Microbiol
Immuno 1968, 43:44-91.
Allering L, Jost H, Emmerich P, Gunther S, Lattwein E, Schmidt M, Seifried E,
Sambri V, Hourfar K, Schmidt-Chanasit J. Detection of Usutu virus infection
in a healthy blood donor from south-west Germany, 2012. Euro Surveill
2012, 17(50).
Appler KK, Brown AN, Stewart BS, Behr MJ, Demarest VL, Wong SJ, Bernard KA.
Persistence of West Nile virus in the central nervous system and periphery
of mice. PLoS One 2010, 5(5):e10649. doi: 10.1371/journal.pone.0010649.
Arjona A, Fikrig E: Innate Immune Responses to West Nile Virus Infection. In:
West Nile Encephalitis Virus Infection: Viral Pathogenesis and the Host Immune
Response. Edited by Diamond MS. New York: Springer Science+Business
Media; 2009:169-187.
Austgen LE, Bowen RA, Bunning ML, Davis BS, Mitchell CJ, Chang GJ.
Experimental infection of cats and dogs with West Nile virus. Emerg Infect
Dis 2004, 10(1):82-86.
Bagnarelli P, Marinelli K, Trotta D, Monachetti A, Tavio M, Del Gobbo R,
Capobianchi M, Menzo S, Nicoletti L, Magurano F, Varaldo P. Human case of
autochthonous West Nile virus lineage 2 infection in Italy, September
2011. Euro Surveill 2011, 16(43).
Bakonyi T, Ivanics T, Erdelyi K, Ursu K, Ferenczi E, Weissenbock H, Nowotny N.
Lineage 1 and 2 strains of encephalitic West Nile virus, central Europe.
Emerg Infect Dis 2006, 12(4):618-623.
Bakonyi T, Erdelyi K, Ursu K, Ferenczi E, Csorgo T, Lussy H, Chvala S, Bukovsky
C, Meister T, Weissenbock H, Nowotny N. Emergence of Usutu virus in
Hungary. J Clin Microbiol 2007, 45(12):3870-3874.
Barnard BJ, Buys SB, Du Preez JH, Greyling SP, Venter HJ. Turkey meningoencephalitis in South Africa. Onderstepoort J Vet Res 1980, 47(2):89-94.
Beasley DW, Li L, Suderman MT, Barrett AD. Mouse neuroinvasive phenotype of
West Nile virus strains varies depending upon virus genotype. Virology
2002, 296(1):17-23.
Beasley DW, Whiteman MC, Zhang S, Huang CY, Schneider BS, Smith DR,
Gromowski GD, Higgs S, Kinney RM, Barrett AD. Envelope protein
40

Introduccin

glycosylation status influences mouse neuroinvasion phenotype of genetic


lineage 1 West Nile virus strains. J Virol 2005, 79(13):8339-8347.
Bechmann I, Galea I, Perry VH. What is the blood-brain barrier (not)? Trends
Immunol 2007, 28(1):5-11.
Becker N, Jost H, Ziegler U, Eiden M, Hoper D, Emmerich P, Fichet-Calvet E,
Ehichioya DU, Czajka C, Gabriel M, Hoffmann B, Beer M, Tenner-Racz K, Racz
P, Gunther S, Wink M, Bosch S, Konrad A, Pfeffer M, Groschup MH, SchmidtChanasit J. Epizootic emergence of Usutu virus in wild and captive birds in
Germany. PLoS One 2012, 7(2):e32604. doi: 10.1371/journal.pone.0032604.
Bernabeu RL: Evaluacin econmica de la caza en Castilla-La Mancha. PhD
Thesis, Albacete: Universidad de Castilla-La Mancha, 2000.
Bernabeu-Wittel M, Ruiz-Prez M, del Toro MD, Aznar J, Muniain A, de Ory F,
Domingo C, Pachon J. West Nile virus past infections in the general
population of Southern Spain. Enferm Infecc Microbiol Clin 2007, 25(9):561565.
Bigham AW, Buckingham KJ, Husain S, Emond MJ, Bofferding KM, Gildersleeve H,
Rutherford A, Astakhova NM, Perelygin AA, Busch MP, Murray KO, Sejvar JJ,
Green S, Kriesel J, Brinton MA, Bamshad M. Host genetic risk factors for
West Nile virus infection and disease progression. PLoS One 2011,
6(9):e24745. doi: 10.1371/journal.pone.0024745.
Blanco-Aguiar JA, Virgs E, Villafuerte R: Perdiz roja (Alectoris rufa). In: Atlas de
las aves reproductoras de Espaa. Edited by Mart R, del Moral JC. Madrid:
DGCoNa-SEO; 2003:212-213.
Blanco-Aguiar JA: Variacin espacial en la biologa de la perdiz roja (Alectoris
rufa); una aproximacin multidisciplinar. PhD Thesis, Madrid: Universidad
Complutense de Madrid, 2007.
Boadella M, Dez-Delgado I, Gutirrez-Guzmn AV, Hfle U, Gortzar C. Do wild
ungulates allow improved monitoring of flavivirus circulation in Spain?
Vector Borne Zoonotic Dis 2012, 12(6):490-495.
Bofill D, Domingo C, Cardenosa N, Zaragoza J, de Ory F, Minguell S, Snchez-Seco
MP, Domnguez A, Tenorio A. Human West Nile virus infection, Catalonia,
Spain. Emerg Infect Dis 2006, 12(7):1163-1164.
Bondre VP, Sapkal GN, Yergolkar PN, Fulmali PV, Sankararaman V, Ayachit VM,
Mishra AC, Gore MM. Genetic characterization of Bagaza virus (BAGV)
isolated in India and evidence of anti-BAGV antibodies in sera collected
from encephalitis patients. J Gen Virol 2009, 90(Pt 11):2644-2649.
Brault AC, Langevin SA, Bowen RA, Panella NA, Biggerstaff BJ, Miller BR, Komar N.
Differential virulence of West Nile strains for American crows. Emerg Infect
Dis 2004, 10(12):2161-2168.
Brault AC, Huang CY, Langevin SA, Kinney RM, Bowen RA, Ramey WN, Panella NA,
Holmes EC, Powers AM, Miller BR. A single positively selected West Nile

41

Virginia Gamino Rodrguez

Tesis doctoral

viral mutation confers increased virogenesis in American crows. Nat Genet


2007, 39(9):1162-1166.
Brhin AC, Mouries J, Frenkiel MP, Dadaglio G, Despres P, Lafon M, Couderc T.
Dynamics of immune cell recruitment during West Nile encephalitis and
identification of a new CD19+B220-BST-2+ leukocyte population. J
Immunol 2008, 180(10):6760-6767.
Brinton MA. The molecular biology of West Nile Virus: a new invader of the
western hemisphere. Annu Rev Microbiol 2002, 56:371-402.
Brjer C, Agren EO, Uhlhorn H, Bernodt K, Jansson DS, Gavier-Widen D.
Characterization of encephalitis in wild birds naturally infected by highly
pathogenic avian influenza H5N1. Avian Dis 2012, 56(1):144-152.
Buckley A, Dawson A, Moss SR, Hinsley SA, Bellamy PE, Gould EA. Serological
evidence of West Nile virus, Usutu virus and Sindbis virus infection of
birds in the UK. J Gen Virol 2003, 84(Pt 10):2807-2817.
Buckley A, Dawson A, Gould EA. Detection of seroconversion to West Nile virus,
Usutu virus and Sindbis virus in UK sentinel chickens. Virol J 2006, 3:71.
Bueno Mar R, Jimnez Peydr R. Current status and eco-epidemiology of
mosquito-borne arboviruses (Diptera: Culicidae) in Spain. Rev Esp Salud
Publica 2010, 84(3):255-269.
Buer J, Balling R. Mice, microbes and models of infection. Nat Rev Genet 2003,
4(3):195-205.
Busquets N, Alba A, Allepuz A, Aranda C, Ignacio Nuez J. Usutu virus sequences
in Culex pipiens (Diptera: Culicidae), Spain. Emerg Infect Dis 2008,
14(5):861-863.
Busquets N, Bertran K, Costa TP, Rivas R, de la Fuente JG, Villalba R, Solanes D,
Bensaid A, Maj N, Pages N. Experimental West Nile virus infection in GyrSaker hybrid falcons. Vector Borne Zoonotic Dis 2012, 12(6):482-489.
Caldern J: Sobre el papel de la perdiz roja (Alectoris rufa) en la dieta de los
depredadores ibricos. PhD Thesis, Sevilla: Universidad de Sevilla, 1975.
Calisher CH, Karabatsos N, Dalrymple JM, Shope RE, Porterfield JS, Westaway EG,
Brandt WE. Antigenic relationships between flaviviruses as determined by
cross-neutralization tests with polyclonal antisera. J Gen Virol 1989, 70 (Pt
1):37-43.
Calzolari M, Gaibani P, Bellini R, Defilippo F, Pierro A, Albieri A, Maioli G, Luppi A,
Rossini G, Balzani A, Tamba M, Galletti G, Gelati A, Carrieri M, Poglayen G,
Cavrini F, Natalini S, Dottori M, Sambri V, Angelini P, Bonilauri P. Mosquito,
bird and human surveillance of West Nile and Usutu viruses in EmiliaRomagna Region (Italy) in 2010. PLoS One 2012, 7(5):e38058. doi:
10.1371/journal.pone.0038058.
Castillo-Olivares J, Wood J. West Nile virus infection of horses. Vet Res 2004,
35(4):467-483.
42

Introduccin

Cavrini F, Gaibani P, Longo G, Pierro AM, Rossini G, Bonilauri P, Gerundi GE, Di


Benedetto F, Pasetto A, Girardis M, Dottori M, Landini MP, Sambri V. Usutu
virus infection in a patient who underwent orthotropic liver
transplantation, Italy, August-September 2009. Euro Surveill 2009, 14(50).
Cavrini F, Della Pepa ME, Gaibani P, Pierro AM, Rossini G, Landini MP, Sambri V.
A rapid and specific real-time RT-PCR assay to identify Usutu virus in
human plasma, serum, and cerebrospinal fluid. J Clin Virol 2011, 50(3):221223.
Centers
for
Disease
Control
and
Prevention
[http://www.cdc.gov/ncidod/dvbid/westnile/birds&mammals.htm]

(CDC)

Centers for Disease Control and Prevention. Possible West Nile virus
transmission to an infant through breast-feeding--Michigan, 2002. MMWR
Morb Mortal Wkly Rep 2002a, 51(39):877-878.
Centers for Disease Control and Prevention. Laboratory-acquired West Nile virus
infections--United States, 2002. MMWR Morb Mortal Wkly Rep 2002b,
51(50):1133-1135.
Centers for Disease Control and Prevention. Intrauterine West Nile virus
infection--New York, 2002. MMWR Morb Mortal Wkly Rep 2002c, 51(50):11351136.
Centers for Disease Control and Prevention: West Nile virus infection among
turkey breeder farm workers--Wisconsin, 2002. MMWR Morb Mortal Wkly Rep
2003, 52(42):1017-1019.
Chambers TJ, Diamond MS. Pathogenesis of flavivirus encephalitis. Adv Virus
Res 2003, 60:273-342.
Chastel C, Launay H, Rogues G, Beaucournu JC. Arbovirus infections in Spain:
serological survey on small mammals. Bull Soc Pathol Exot Filiales 1980,
73(4):384-390.
Cho H, Diamond MS. Immune responses to West Nile virus infection in the
central nervous system. Viruses 2012, 4(12):3812-3830.
Chu JJ, Ng ML. The mechanism of cell death during West Nile virus infection is
dependent on initial infectious dose. J Gen Virol 2003, 84(Pt 12):3305-3314.
Chvala S, Kolodziejek J, Nowotny N, Weissenbock H. Pathology and viral
distribution in fatal Usutu virus infections of birds from the 2001 and
2002 outbreaks in Austria. J Comp Pathol 2004, 131(2-3):176-185.
Chvala S, Bakonyi T, Hackl R, Hess M, Nowotny N, Weissenbock H. Limited
pathogenicity of Usutu virus for the domestic chicken (Gallus domesticus).
Avian Pathol 2005, 34(5):392-395.
Chvala S, Bakonyi T, Hackl R, Hess M, Nowotny N, Weissenbock H. Limited
pathogenicity of usutu virus for the domestic goose (Anser anser f.
domestica) following experimental inoculation. J Vet Med B Infect Dis Vet
Public Health 2006, 53(4):171-175.
43

Virginia Gamino Rodrguez

Tesis doctoral

Chvala S, Bakonyi T, Bukovsky C, Meister T, Brugger K, Rubel F, Nowotny N,


Weissenbock H. Monitoring of Usutu virus activity and spread by using
dead bird surveillance in Austria, 2003-2005. Vet Microbiol 2007, 122(34):237-245.
Clark DC, Brault AC, Hunsperger E. The contribution of rodent models to the
pathological assessment of flaviviral infections of the central nervous
system. Arch Virol 2012, 157(8):1423-1440.
Davis K: The Experimental Use of Chickens and Other Birds in Biomedical and
Agricultural Research. PhD Thesis, Boston: New England Anti-Vivisection
Society, 2003.
Davis CT, Ebel GD, Lanciotti RS, Brault AC, Guzman H, Siirin M, Lambert A,
Parsons RE, Beasley DW, Novak RJ, Elizondo-Quiroga D, Green EN, Young DS,
Stark LM, Drebot MA, Artsob H, Tesh RB, Kramer LD, Barrett AD.
Phylogenetic analysis of North American West Nile virus isolates, 20012004: evidence for the emergence of a dominant genotype. Virology 2005,
342(2):252-265.
Desai A, Shankar SK, Ravi V, Chandramuki A, Gourie-Devi M. Japanese
encephalitis virus antigen in the human brain and its topographic
distribution. Acta Neuropathol 1995, 89(4):368-373.
Diallo M, Nabeth P, Ba K, Sall AA, Ba Y, Mondo M, Girault L, Abdalahi MO, Mathiot
C. Mosquito vectors of the 1998-1999 outbreak of Rift Valley Fever and
other arboviruses (Bagaza, Sanar, Wesselsbron and West Nile) in Mauritania
and Senegal. Med Vet Entomol 2005, 19(2):119-126.
Diamond MS, Shrestha B, Marri A, Mahan D, Engle M. B cells and antibody play
critical roles in the immediate defense of disseminated infection by West
Nile encephalitis virus. J Virol 2003, 77(4):2578-2586.
Digoutte JP. Bagaza (BAG) strain: Dak Ar B 209. Am J Trop Med Hyg 1978,
27:376-377.
Diniz JA, Da Rosa AP, Guzman H, Xu F, Xiao SY, Popov VL, Vasconcelos PF, Tesh
RB. West Nile virus infection of primary mouse neuronal and neuroglial
cells: the role of astrocytes in chronic infection. Am J Trop Med Hyg 2006,
75(4):691-696.
Dusek RJ, McLean RG, Kramer LD, Ubico SR, Dupuis AP, 2nd, Ebel GD, Guptill
SC. Prevalence of West Nile virus in migratory birds during spring and fall
migration. Am J Trop Med Hyg 2009, 81(6):1151-1158.
Ecco R, Susta L, Afonso CL, Miller PJ, Brown C. Neurological lesions in chickens
experimentally infected with virulent Newcastle disease virus isolates.
Avian Pathol 2011, 40(2):145-152.
Eldadah AH, Nathanson N, Sarsitis R. Pathogenesis of West Nile Virus
encephalitis in mice and rats. 1. Influence of age and species on mortality
and infection. Am J Epidemiol 1967, 86(3):765-775.

44

Introduccin

Fang Y, Reisen WK. Previous infection with West Nile or St. Louis encephalitis
viruses provides cross protection during reinfection in house finches. Am J
Trop Med Hyg 2006, 75(3):480-485.
Fernndez-Garca MD, Mazzon M, Jacobs M, Amara A. Pathogenesis of flavivirus
infections: using and abusing the host cell. Cell Host Microbe 2009, 5(4):318328.
Figuerola J, Jimnez-Clavero MA, Rojo G, Gmez-Tejedor C, Soriguer R. Prevalence
of West Nile virus neutralizing antibodies in colonial aquatic birds in
southern Spain. Avian Pathol 2007, 36(3):209-212.
Figuerola J, Soriguer R, Rojo G, Gmez Tejedor C, Jimnez-Clavero MA.
Seroconversion in wild birds and local circulation of West Nile virus, Spain.
Emerg Infect Dis 2007, 13(12):1915-1917.
Figuerola J, Jimnez-Clavero MA, Lpez G, Rubio C, Soriguer R, Gmez-Tejedor C,
Tenorio A. Size matters: West Nile Virus neutralizing antibodies in resident
and migratory birds in Spain. Vet Microbiol 2008, 132(1-2):39-46.
Filipe AR, Andrade HR. Arboviruses in the Iberian Peninsula. Acta Virol 1990,
34(6):582-591.
Flavivirus.wordpess.com [http://flavivirus.wordpress.com/general-flavivirus-info/]
Foppa IM, Beard RH, Mendenhall IH. The impact of West Nile virus on the
abundance of selected North American birds. BMC Vet Res 2011, 7:43. doi:
10.1186/1746-6148-7-43.
Gaibani P, Pierro AM, Cavrini F, Rossini G, Landini MP, Sambri V. False-positive
transcription-mediated amplification assay detection of West Nile virus in
blood from a patient with viremia caused by an Usutu virus infection. J
Clin Microbiol 2010, 48(9):3338-3339.
Gaibani P, Pierro A, Alicino R, Rossini G, Cavrini F, Landini MP, Sambri V.
Detection of Usutu-virus-specific IgG in blood donors from northern Italy.
Vector Borne Zoonotic Dis 2012, 12(5):431-433.
Gamino V, Gutirrez-Guzmn AV, Fernndez-de-Mera IG, Ortiz JA, Durn-Martn
M, de la Fuente J, Gortzar C, Hfle U. Natural Bagaza virus infection in
game birds in southern Spain. Vet Res 2012, 43(1):65. doi: 10.1186/12979716-43-65.
Gangoso L, Grande JM, Llorente F, Jimnez-Clavero MA, Prez JM, Figuerola J.
Prevalence of neutralizing antibodies to West Nile virus in Eleonora's
Falcons in the Canary Islands. J Wildl Dis 2010, 46(4):1321-1324.
Garca-Bocanegra I, Jan-Tllez JA, Napp S, Arenas-Montes A, Fernndez-Morente
M, Fernndez-Molera V, Arenas A. West Nile fever outbreak in horses and
humans, Spain, 2010. Emerg Infect Dis 2011a, 17(12):2397-2399.
Garca-Bocanegra I, Busquets N, Napp S, Alba A, Zorrilla I, Villalba R, Arenas A.
Serosurvey of West Nile virus and other flaviviruses of the Japanese
encephalitis antigenic complex in birds from Andalusia, southern Spain.
Vector Borne Zoonotic Dis 2011b, 11(8):1107-1113.
45

Virginia Gamino Rodrguez

Tesis doctoral

Garca-Bocanegra I, Jan-Tllez JA, Napp S, Arenas-Montes A, Fernndez-Morente


M, Fernndez-Molera V, Arenas A. Monitoring of the West Nile virus
epidemic in Spain between 2010 and 2011. Transbound Emerg Dis 2012a,
59(5):448-455.
Garca-Bocanegra I, Zorrilla I, Rodrguez E, Rayas E, Camacho L, Redondo I,
Gmez-Guillamn F. Monitoring of the Bagaza Virus Epidemic in Wild Bird
Species in Spain, 2010. Transbound Emerg Dis 2012b, 60(2):120-126.
Garca-Bocanegra I, Arenas-Montes A, Jan-Tllez JA, Napp S, Fernndez-Morente
M, Arenas A. Use of sentinel serosurveillance of mules and donkeys in the
monitoring of West Nile virus infection. Vet J 2012c, 194(2):262-264.
Garca-Bocanegra I, Arenas-Montes A, Napp S, Jan-Tllez JA, Fernndez-Morente
M, Fernndez-Molera V, Arenas A. Seroprevalence and risk factors
associated to West Nile virus in horses from Andalusia, Southern Spain. Vet
Microbiol 2012d, 160(3-4):341-346.
Garca-Tapia D, Loiacono CM, Kleiboeker SB. Replication of West Nile virus in
equine peripheral blood mononuclear cells. Vet Immunol Immunopathol 2006,
110(3-4):229-244.
Garca-Tapia D, Hassett DE, Mitchell WJ, Jr., Johnson GC, Kleiboeker SB. West
Nile virus encephalitis: sequential histopathological and immunological
events in a murine model of infection. J Neurovirol 2007, 13(2):130-138.
Garmendia AE, Van Kruiningen HJ, French RA, Anderson JF, Andreadis TG,
Kumar A, West AB. Recovery and identification of West Nile virus from a
hawk in winter. J Clin Microbiol 2000, 38(8):3110-3111.
Gaunt MW, Sall AA, de Lamballerie X, Falconar AK, Dzhivanian TI, Gould EA.
Phylogenetic
relationships
of
flaviviruses
correlate
with
their
epidemiology, disease association and biogeography. J Gen Virol 2001, 82(Pt
8):1867-1876.
Gehrmann J, Matsumoto Y, Kreutzberg GW. Microglia: intrinsic immuneffector
cell of the brain. Brain Res Brain Res Rev 1995, 20(3):269-287.
Gibbs SE, Ellis AE, Mead DG, Allison AB, Moulton JK, Howerth EW, Stallknecht
DE. West Nile virus detection in the organs of naturally infected blue jays
(Cyanocitta cristata). J Wildl Dis 2005, 41(2):354-362.
Gonzlez MT, Filipe AR. Antibodies to arboviruses in northwestern Spain. Am J
Trop Med Hyg 1977, 26(4):792-797.
Gortzar C: Las repoblaciones con perdiz roja. In: La perdiz roja. Edited by
Fedenca. Madrid: Grupo editorial V; 1998:119-134.
Gould EA, de Lamballerie X, Zanotto PM, Holmes EC. Origins, evolution, and
vector/host coadaptations within the genus Flavivirus. Adv Virus Res 2003,
59:277-314.
Gould EA, Solomon T. Pathogenic flaviviruses. Lancet 2008, 371(9611):500-509.

46

Introduccin

Gutirrez-Guzmn AV, Vicente J, Sobrino R, Perz-Ramrez E, Llorente F, Hfle U.


Antibodies to West Nile virus and related flaviviruses in wild boar, red
foxes and other mesomammals from Spain. Vet Microbiol 2012, 159(3-4):291297.
Guy JS, Malkinson M: Arbovirus infections. In: Diseases of Poultry. Edited by Saif
YM. Ames: Blackwell Publishing; 2008:414-425.
Hayes EB, Gubler DJ. West Nile virus: epidemiology and clinical features of an
emerging epidemic in the United States. Annu Rev Med 2006, 57:181-194.
Heinz FX, Purcell MS, Gould EA, Howard CR, Houghton M: Family Flaviviridae.
In: Virus Taxonomy Seventh Report of the International Committee on Taxonomy
of Viruses. Edited by van Regenmortel MHV, Fauquet CM, Bishop DHL,
Carstens EB, Estes MK, Lemon SM, Maniloff J, Mayo MA, McGeoch DJ, Pringle
CR et al. San Diego: Academic Press; 2000:860-878.
Hfle U, Blanco JM, Crespo E, Naranjo V, Jimnez-Clavero MA, Snchez A, de la
Fuente J, Gortzar C. West Nile virus in the endangered Spanish imperial
eagle. Vet Microbiol 2008, 129(1-2):171-178.
Hfle U, Gamino V, Fernndez de Mera IG, Mangold AJ, Ortz JA, de la Fuente J.
Usutu Virus in Migratory Song Thrushes, Spain. Emerg Infect Dis 2013,
19(7). doi: 10.3201/eid1907.130199.
Huang CH, Wong C. Relation of the peripheral multiplication of Japanese B
encephalitis virus to the pathogenesis of the infection in mice. Acta Virol
1963, 7:322-330.
Hublek Z, Halouzka J. West Nile fever--a reemerging mosquito-borne viral
disease in Europe. Emerg Infect Dis 1999, 5(5):643-650.
Hublek Z. Mosquito-borne viruses in Europe. Parasitol Res 2008a, 103 Suppl
1:S29-43.
Hublek Z, Wegner E, Halouzka J, Tryjanowski P, Jerzak L, Sikutova S, Rudolf I,
Kruszewicz AG, Jaworski Z, Wlodarczyk R. Serologic survey of potential
vertebrate hosts for West Nile virus in Poland. Viral Immunol 2008b,
21(2):247-253.
Hublek Z, Halouzka J, Juricova Z, Sikutova S, Rudolf I, Honza M, Jankova J,
Chytil J, Marec F, Sitko J. Serologic survey of birds for West Nile flavivirus
in southern Moravia (Czech Republic). Vector Borne Zoonotic Dis 2008c,
8(5):659-666.
Hublek Z, Rudolf I, Capek M, Bakonyi T, Betasova L, Nowotny N. Usutu Virus in
Blackbirds (Turdus merula), Czech Republic, 2011-2012. Transbound Emerg
Dis 2012. doi: 10.1111/tbed.12025.
Iwamoto M, Jernigan DB, Guasch A, Trepka MJ, Blackmore CG, Hellinger WC,
Pham SM, Zaki S, Lanciotti RS, Lance-Parker SE, DiazGranados CA, Winquist
AG, Perlino CA, Wiersma S, Hillyer KL, Goodman JL, Marfin AA, Chamberland
ME, Petersen LR. Transmission of West Nile virus from an organ donor to
four transplant recipients. N Engl J Med 2003, 348(22):2196-2203.
47

Virginia Gamino Rodrguez

Tesis doctoral

Jess-De La Calle I, Espinosa-Garca MJ, Prez-Ramos S, Cruz-Rosales E. First


confirmed cases of human meningoencephalitis due to West Nile virus in
Andalusia, Spain. Enferm Infecc Microbiol Clin 2012, 30(7):426-427.
Jimnez-Clavero MA, Tejedor CG, Rojo G, Soriguer R, Figuerola J. Serosurvey of
West Nile virus in equids and bovids in Spain. Vet Rec 2007, 161(6):212.
Jimnez-Clavero MA, Sotelo E, Fernndez-Pinero J, Llorente F, Manuel Blanco J,
Rodrguez-Ramos J, Prez-Ramrez E, Hfle U. West Nile virus in golden
eagles, Spain, 2007. Emerg Infect Dis 2008, 14(9):1489-1491.
Jimnez-Clavero MA, Llorente F, Sotelo E, Soriguer R, Gmez-Tejedor C, Figuerola
J. West Nile virus serosurveillance in horses in Donana, Spain, 2005 to
2008. Vet Rec 2010, 167(10):379-380.
Jimnez-Clavero MA. Animal viral diseases and global change: bluetongue and
West Nile fever as paradigms. Front Genet 2012, 3:105. doi:
10.3389/fgene.2012.00105.
Johnston LJ, Halliday GM, King NJ. Langerhans cells migrate to local lymph
nodes following cutaneous infection with an arbovirus. J Invest Dermatol
2000, 114(3):560-568.
Jordan I, Briese T, Fischer N, Lau JY, Lipkin WI. Ribavirin inhibits West Nile
virus replication and cytopathic effect in neural cells. J Infect Dis 2000,
182(4):1214-1217.
Kaptoul D, Viladrich PF, Domingo C, Niubo J, Martinez-Yelamos S, De Ory F,
Tenorio A. West Nile virus in Spain: report of the first diagnosed case (in
Spain) in a human with aseptic meningitis. Scand J Infect Dis 2007,
39(1):70-71.
Kelley TW, Prayson RA, Ruiz AI, Isada CM, Gordon SM. The neuropathology of
West Nile virus meningoencephalitis. A report of two cases and review of
the literature. Am J Clin Pathol 2003, 119(5):749-753.
Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. Physiology of microglia.
Physiol Rev 2011, 91(2):461-553.
Kilpatrick AM, Meola MA, Moudy RM, Kramer LD. Temperature, viral genetics,
and the transmission of West Nile virus by Culex pipiens mosquitoes. PLoS
Pathog 2008, 4(6):e1000092. doi: 10.1371/journal.ppat.1000092.
Kinney RM, Huang CY, Whiteman MC, Bowen RA, Langevin SA, Miller BR, Brault
AC. Avian virulence and thermostable replication of the North American
strain of West Nile virus. J Gen Virol 2006, 87(Pt 12):3611-3622.
Klenk K, Snow J, Morgan K, Bowen R, Stephens M, Foster F, Gordy P, Beckett S,
Komar N, Gubler D, Bunning M. Alligators as West Nile virus amplifiers.
Emerg Infect Dis 2004, 10(12):2150-2155.
Komar N. West Nile virus: epidemiology and ecology in North America. Adv
Virus Res 2003a, 61:185-234.

48

Introduccin

Komar N, Langevin S, Hinten S, Nemeth N, Edwards E, Hettler D, Davis B, Bowen


R, Bunning M. Experimental infection of North American birds with the
New York 1999 strain of West Nile virus. Emerg Infect Dis 2003b, 9(3):311322.
Komarov A, Kalmar E. A hitherto undescribed disease-turkey meningoencephalitis. Vet Rec 1960, 72:257-261.
Kostiukov MA, Gordeeva ZE, Bulychev VP, Nemova NV, Daniiarov OA. The lake
frog (Rana ridibunda)--one of the food hosts of blood-sucking mosquitoes in
Tadzhikistan--a reservoir of the West Nile fever virus. Med Parazitol (Mosk)
1985(3):49-50.
Kostiukov MA, Alekseev AN, Bulychev VP, Gordeeva ZE. Experimental evidence
for infection of Culex pipiens L. mosquitoes by West Nile fever virus from
Rana ridibunda Pallas and its transmission by bites. Med Parazitol (Mosk)
1986(6):76-78.
Kramer-Hammerle S, Rothenaigner I, Wolff H, Bell JE, Brack-Werner R. Cells of
the central nervous system as targets and reservoirs of the human
immunodeficiency virus. Virus Res 2005, 111(2):194-213.
Kumar M, Verma S, Nerurkar VR. Pro-inflammatory cytokines derived from
West Nile virus (WNV)-infected SK-N-SH cells mediate neuroinflammatory
markers and neuronal death. J Neuroinflammation 2010, 7:73. doi:
10.1186/1742-2094-7-73.
Kuno G, Chang GJ, Tsuchiya KR, Karabatsos N, Cropp CB. Phylogeny of the
genus Flavivirus. J Virol 1998, 72(1):73-83.
Kuno G. Transmission of arboviruses without involvement of arthropod
vectors. Acta Virol 2001, 45(3):139-150.
Kuno G, Chang GJ. Biological transmission of arboviruses: reexamination of
and new insights into components, mechanisms, and unique traits as well
as their evolutionary trends. Clin Microbiol Rev 2005, 18(4):608-637.
Kuno G, Chang GJ. Full-length sequencing and genomic characterization of
Bagaza, Kedougou, and Zika viruses. Arch Virol 2007, 152(4):687-696.
Kutasi O, Bakonyi T, Lecollinet S, Biksi I, Ferenczi E, Bahuon C, Sardi S, Zientara
S, Szenci O. Equine encephalomyelitis outbreak caused by a genetic lineage
2 West Nile virus in Hungary. J Vet Intern Med 2011, 25(3):586-591.
LaDeau SL, Kilpatrick AM, Marra PP. West Nile virus emergence and large-scale
declines of North American bird populations. Nature 2007, 447(7145):710713.
Lanciotti RS, Roehrig JT, Deubel V, Smith J, Parker M, Steele K, Crise B, Volpe KE,
Crabtree MB, Scherret JH, Hall RA, MacKenzie JS, Cropp CB, Panigrahy B,
Ostlund E, Schmitt B, Malkinson M, Banet C, Weissman J, Komar N, Savage
HM, Stone W, McNamara T, Gubler DJ. Origin of the West Nile virus
responsible for an outbreak of encephalitis in the northeastern United
States. Science 1999, 286(5448):2333-2337.
49

Virginia Gamino Rodrguez

Tesis doctoral

Lanciotti RS, Ebel GD, Deubel V, Kerst AJ, Murri S, Meyer R, Bowen M, McKinney
N, Morrill WE, Crabtree MB, Kramer LD, Roehrig JT. Complete genome
sequences and phylogenetic analysis of West Nile virus strains isolated
from the United States, Europe, and the Middle East. Virology 2002,
298(1):96-105.
Langevin SA, Bunning M, Davis B, Komar N. Experimental infection of chickens
as candidate sentinels for West Nile virus. Emerg Infect Dis 2001, 7(4):726729.
Lelli R, Savini G, Teodori L, Filipponi G, Di Gennaro A, Leone A, Di Gialleonardo L,
Venturi L, Caporale V. Serological evidence of USUTU virus occurrence in
north-eastern Italy. Zoonoses Public Health 2008, 55(7):361-367.
Libraty DH, Pichyangkul S, Ajariyakhajorn C, Endy TP, Ennis FA. Human
dendritic cells are activated by dengue virus infection: enhancement by
gamma interferon and implications for disease pathogenesis. J Virol 2001,
75(8):3501-3508.
Lim PY, Behr MJ, Chadwick CM, Shi PY, Bernard KA. Keratinocytes are cell
targets of West Nile virus in vivo. J Virol 2011, 85(10):5197-5201.
Lim SM, Koraka P, Osterhaus AD, Martina BE. West Nile virus: immunity and
pathogenesis. Viruses 2011, 3(6):811-828.
Lindenbach BD, Rice CM. Molecular biology of flaviviruses. Adv Virus Res 2003,
59:23-61.
Linke S, Niedrig M, Kaiser A, Ellerbrok H, Muller K, Muller T, Conraths FJ, Muhle
RU, Schmidt D, Koppen U, Bairlein F, Berthold P, Pauli G. Serologic evidence
of West Nile virus infections in wild birds captured in Germany. Am J Trop
Med Hyg 2007, 77(2):358-364.
Liou ML, Hsu CY. Japanese encephalitis virus is transported across the
cerebral blood vessels by endocytosis in mouse brain. Cell Tissue Res 1998,
293(3):389-394.
Llorente F, Prez-Ramrez E, Fernndez-Pinero J, Soriguer R, Figuerola J, JimnezClavero MA. Flaviviruses in Game Birds, Southern Spain, 20112012. Emerg
Infect Dis 2013, 19(6):1024-1026. doi: 10.3201/eid 1906.130122.
Lopes H, Redig P, Glaser A, Armien A, Wunschmann A. Clinical findings, lesions,
and viral antigen distribution in great gray owls (Strix nebulosa) and
barred owls (Strix varia) with spontaneous West Nile virus infection. Avian
Dis 2007, 51(1):140-145.
Lpez G, Jimnez-Clavero MA, Tejedor CG, Soriguer R, Figuerola J. Prevalence of
West Nile virus neutralizing antibodies in Spain is related to the behavior
of migratory birds. Vector Borne Zoonotic Dis 2008, 8(5):615-621.
Lpez G, Jimnez-Clavero MA, Vzquez A, Soriguer R, Gmez-Tejedor C, Tenorio A,
Figuerola J. Incidence of West Nile virus in birds arriving in wildlife
rehabilitation centers in southern Spain. Vector Borne Zoonotic Dis 2011,
11(3):285-290.
50

Introduccin

Lozano A: Arboviruses in Spain. In: Arboviruses in the Mediterranean countries.


Edited by Vesenjak-Hirjan J. Stuttgart: Gustav Fisher Verlag; 1980.
Lozano A, Filipe AR. Antibodies against the West Nile virus and other arthropodtransmitted viruses in the Ebro Delta region. Rev Esp Salud Publica 1998,
72(3):245-250.
Ludwig GV, Calle PP, Mangiafico JA, Raphael BL, Danner DK, Hile JA, Clippinger
TL, Smith JF, Cook RA, McNamara T. An outbreak of West Nile virus in a
New York City captive wildlife population. Am J Trop Med Hyg 2002,
67(1):67-75.
Lupulovic D, Martn-Acebes MA, Lazic S, Alonso-Padilla J, Blzquez AB, EscribanoRomero E, Petrovic T, Saiz JC. First serological evidence of West Nile virus
activity in horses in Serbia. Vector Borne Zoonotic Dis 2011, 11(9):1303-1305.
Mackenzie JS, Williams DT. The zoonotic flaviviruses of southern, south-eastern
and eastern Asia, and Australasia: the potential for emergent viruses.
Zoonoses Public Health 2009, 56(6-7):338-356.
Magistretti PJ, Ransom BR: Astrocytes. In: Neuropsychopharmacology: The Fifth
Generation of Progress. Edited by Davis KL, Charney D, Coyle JT, Nemeroff C.
Philadelphia: Lippincott Williams & Wilkins; 2002:133-145.
Malkinson M, Banet C, Weisman Y, Pokamunski S, King R, Drouet MT, Deubel V.
Introduction of West Nile virus in the Middle East by migrating white
storks. Emerg Infect Dis 2002a, 8(4):392-397.
Malkinson M, Banet C. The role of birds in the ecology of West Nile virus in
Europe and Africa. Curr Top Microbiol Immuno 2002b, 267:309-322.
Manarolla G, Bakonyi T, Gallazzi D, Crosta L, Weissenbock H, Dorrestein GM,
Nowotny N. Usutu virus in wild birds in northern Italy. Vet Microbiol 2010,
141(1-2):159-163.
Martn-Acebes MA, Saiz JC. West Nile virus: a re-emerging pathogen revisited.
World J Virol 2012, 1(2):51-70.
McLean RG, Ubico SR, Docherty DE, Hansen WR, Sileo L, McNamara TS. West Nile
virus transmission and ecology in birds. Ann N Y Acad Sci 2001, 951:54-57.
McMinn PC, Dalgarno L, Weir RC. A comparison of the spread of Murray Valley
encephalitis viruses of high or low neuroinvasiveness in the tissues of
Swiss mice after peripheral inoculation. Virology 1996, 220(2):414-423.
McMinn PC. The molecular basis of virulence of the encephalitogenic
flaviviruses. J Gen Virol 1997, 78 2711-2722.
McMullen AR, May FJ, Li L, Guzman H, Bueno R, Jr., Dennett JA, Tesh RB, Barrett
AD. Evolution of new genotype of West Nile virus in North America. Emerg
Infect Dis 2011, 17(5):785-793.
Meister T, Lussy H, Bakonyi T, Sikutova S, Rudolf I, Vogl W, Winkler H, Frey H,
Hubalek Z, Nowotny N, Weissenbock H. Serological evidence of continuing

51

Virginia Gamino Rodrguez

Tesis doctoral

high Usutu virus (Flaviviridae) activity and establishment of herd


immunity in wild birds in Austria. Vet Microbiol 2008, 127(3-4):237-248.
Miller DL, Mauel MJ, Baldwin C, Burtle G, Ingram D, Hines ME, 2nd, Frazier KS.
West Nile virus in farmed alligators. Emerg Infect Dis 2003, 9(7):794-799.
Monath TP: Pathobiology of the flaviviruses. In: "The Togaviridae and the
Flaviviridae". Edited by Schlesinger S, Schlesinger MJ. New York: Plenum Press;
1986:375-440.
Monath TP: Arthropod-borne viruses. In: Emerging viruses. Edited by Morse S.
New York: Oxford University Press; 1993:138-148.
Monath TP, Heinz FX: Flaviviruses. In: Fields Virology. 3rd. Edited by Fields BN,
Knipe DM, Howley PM. Philadelphia: LippincottRaven; 1996:961-1034.
Montgomery DL. Astrocytes: form, functions, and roles in disease. Vet Pathol
1994, 31(2):145-167.
Muldoon LL, Alvarez JI, Begley DJ, Boado RJ, Del Zoppo GJ, Doolittle ND,
Engelhardt B, Hallenbeck JM, Lonser RR, Ohlfest JR, Prat A, Scarpa M,
Smeyne RJ, Drewes LR, Neuwelt EA. Immunologic privilege in the central
nervous system and the blood-brain barrier. J Cereb Blood Flow Metab 2013,
33(1):13-21.
Murata R, Eshita Y, Maeda A, Maeda J, Akita S, Tanaka T, Yoshii K, Kariwa H,
Umemura T, Takashima I. Glycosylation of the West Nile Virus envelope
protein increases in vivo and in vitro viral multiplication in birds. Am J
Trop Med Hyg 2010, 82(4):696-704.
Murgue B, Zeller H, Deubel V. The ecology and epidemiology of West Nile virus
in Africa, Europe and Asia. Curr Top Microbiol Immuno 2002, 267:195-221.
Murray K, Baraniuk S, Resnick M, Arafat R, Kilborn C, Cain K, Shallenberger R,
York TL, Martinez D, Hellums JS, Hellums D, Malkoff M, Elgawley N, McNeely
W, Khuwaja SA, Tesh RB. Risk factors for encephalitis and death from West
Nile virus infection. Epidemiol Infect 2006, 134(6):1325-1332.
Nash D, Mostashari F, Fine A, Miller J, O'Leary D, Murray K, Huang A, Rosenberg
A, Greenberg A, Sherman M, Wong S, Layton M. The outbreak of West Nile
virus infection in the New York City area in 1999. N Engl J Med 2001,
344(24):1807-1814.
Nemeth NM, Gould D, Bowen R, Komar N. Natural and experimental West Nile
virus infection in five raptor species. J Wildl Dis 2006a, 42(1):1-13.
Nemeth NM, Hahn DC, Gould DH, Bowen RA. Experimental West Nile virus
infection in Eastern Screech Owls (Megascops asio). Avian Dis 2006b,
50(2):252-258.
Nemeth NM, Kratz GE, Bates R, Scherpelz JA, Bowen RA, Komar N. Clinical
evaluation and outcomes of naturally acquired West Nile virus infection in
raptors. J Zoo Wildl Med 2009a, 40(1):51-63.

52

Introduccin

Nemeth NM, Oesterle PT, Bowen RA. Humoral immunity to West Nile virus is
long-lasting and protective in the house sparrow (Passer domesticus). Am J
Trop Med Hyg 2009b, 80(5):864-869.
Nemeth NM, Young G, Ndaluka C, Bielefeldt-Ohmann H, Komar N, Bowen R.
Persistent West Nile virus infection in the house sparrow (Passer
domesticus). Arch Virol 2009c, 154(5):783-789.
Nemeth NM, Thomsen BV, Spraker TR, Benson JM, Bosco-Lauth AM, Oesterle PT,
Bright JM, Muth JP, Campbell TW, Gidlewski TL, Bowen RA. Clinical and
pathologic responses of American crows (Corvus brachyrhynchos) and fish
crows (C ossifragus) to experimental West Nile virus infection. Vet Pathol
2011, 48(6):1061-1074.
Nir YD. Airborne West Nile virus infection. Am J Trop Med Hyg 1959, 8:537-539.
Nir Y, Beemer A, Goldwasser RA. West Nile Virus infection in mice following
exposure to a viral aerosol. Br J Exp Pathol 1965, 46(4):443-449.
Odelola HA, Oduye OO. West Nile virus infection of adult mice by oral route.
Arch Virol 1977, 54(3):251-253.
Ousman SS, Kubes P. Immune surveillance in the central nervous system. Nat
Neurosci 2012, 15(8):1096-1101.
Papa A, Xanthopoulou K, Gewehr S, Mourelatos S. Detection of West Nile virus
lineage 2 in mosquitoes during a human outbreak in Greece. Clin Microbiol
Infect 2011, 17(8):1176-1180.
Pealer LN, Marfin AA, Petersen LR, Lanciotti RS, Page PL, Stramer SL, Stobierski
MG, Signs K, Newman B, Kapoor H, Goodman JL, Chamberland ME.
Transmission of West Nile virus through blood transfusion in the United
States in 2002. N Engl J Med 2003, 349(13):1236-1245.
Pecorari M, Longo G, Gennari W, Grottola A, Sabbatini A, Tagliazucchi S, Savini G,
Monaco F, Simone M, Lelli R, Rumpianesi F. First human case of Usutu virus
neuroinvasive infection, Italy, August-September 2009. Euro Surveill 2009,
14(50).
Peletto S, Lo Presti A, Modesto P, Cella E, Acutis PL, Farchi F, Ciotti M, Zehender G,
Ciccozzi M. Genetic diversity of Usutu virus. New Microbiol 2012, 35(2):167174.
Penn RG, Guarner J, Sejvar JJ, Hartman H, McComb RD, Nevins DL, Bhatnagar J,
Zaki SR. Persistent neuroinvasive West Nile virus infection in an
immunocompromised patient. Clin Infect Dis 2006, 42(5):680-683.
Petersen L: Global epidemiology of West Nile virus. In: West Nile encephalitis
virus infection. Edited by MS D. New York: Springer; 2009:1-24.
Peterson AT, Vieglais DA, Andreasen JK. Migratory birds modeled as critical
transport agents for West Nile Virus in North America. Vector Borne Zoonotic
Dis 2003, 3(1):27-37.

53

Virginia Gamino Rodrguez

Tesis doctoral

Pfeffer M, Dobler G. Emergence of zoonotic arboviruses by animal trade and


migration. Parasit Vectors 2010, 3(1):35. doi: 10.1186/1756-3305-3-35.
Phalen DN, Dahlhausen B. West Nile virus. Seminars in Avian and Exotic Pet
Medicine 2004, 13(2).
Pogodina VV, Frolova MP, Malenko GV, Fokina GI, Koreshkova GV, Kiseleva LL,
Bochkova NG, Ralph NM. Study on West Nile virus persistence in monkeys.
Arch Virol 1983, 75(1-2):71-86.
Pujol E, Fuertes EF, Saavedra J, Ruiz S, Daz A, Vzquez I, Domingo C, Figuerola J,
Soriguer R, Guilln J. Seroprevalencia frente al virus West Nile en una
poblacin centinela. Avances en Enfermedades Infecciosas 2004, 29.
Ransohoff RM, Kivisakk P, Kidd G. Three or more routes for leukocyte migration
into the central nervous system. Nat Rev Immunol 2003, 3(7):569-581.
Ransohoff RM, Engelhardt B. The anatomical and cellular basis of immune
surveillance in the central nervous system. Nat Rev Immunol 2012,
12(9):623-635.
Ravi V, Desai AS, Shenoy PK, Satishchandra P, Chandramuki A, Gourie-Devi M.
Persistence of Japanese encephalitis virus in the human nervous system. J
Med Virol 1993, 40(4):326-329.
Red

de Alerta Sanitaria Veterinaria (RASVE)


RASVE_2008/Publica/Focos/Consulta.aspx]

[http://rasve.magrama.es/

Reiter P. Yellow fever and dengue: a threat to Europe? Euro Surveill 2010,
15(10):19509.
Rezai-Zadeh K, Gate D, Town T. CNS infiltration of peripheral immune cells: DDay for neurodegenerative disease? J Neuroimmune Pharmacol 2009,
4(4):462-475.
Rizzoli A, Rosa R, Rosso F, Buckley A, Gould E. West Nile virus circulation
detected in northern Italy in sentinel chickens. Vector Borne Zoonotic Dis
2007, 7(3):411-417.
Rodhain F, Petter JJ, Albignac R, Coulanges P, Hannoun C. Arboviruses and
lemurs in Madagascar: experimental infection of Lemur fulvus with yellow
fever and West Nile viruses. Am J Trop Med Hyg 1985, 34(4):816-822.
Root JJ. West Nile virus associations in wild mammals: a synthesis. Arch Virol
2012. doi: 10.1007/s00705-012-1516-3.
Sadek JR, Pergam SA, Harrington JA, Echevarria LA, Davis LE, Goade D, Harnar J,
Nofchissey RA, Sewell CM, Ettestad P, Haaland KY. Persistent
neuropsychological impairment associated with West Nile virus infection.
J Clin Exp Neuropsychol 2010, 32(1):81-87.
Sampson BA, Armbrustmacher V. West Nile encephalitis: the neuropathology of
four fatalities. Ann N Y Acad Sci 2001, 951:172-178.

54

Introduccin

Samuel MA, Diamond MS. Pathogenesis of West Nile Virus infection: a balance
between virulence, innate and adaptive immunity, and viral evasion. J Virol
2006, 80(19):9349-9360.
Samuel MA, Wang H, Siddharthan V, Morrey JD, Diamond MS. Axonal transport
mediates West Nile virus entry into the central nervous system and
induces acute flaccid paralysis. Proc Natl Acad Sci USA 2007a,
104(43):17140-17145.
Samuel MA, Morrey JD, Diamond MS. Caspase 3-dependent cell death of
neurons contributes to the pathogenesis of West Nile virus encephalitis. J
Virol 2007b, 81(6):2614-2623.
Sanchs-Bayarri V. Contribucin al estudio de la serologa de las infecciones
por Arbovirus. Hospital General 1974, 14(5):417-424.
Sangster MY, Urosevic N, Mansfield JP, Mackenzie JS, Shellam GR. Mapping the
Flv locus controlling resistance to flaviviruses on mouse chromosome 5. J
Virol 1994, 68(1):448-452.
Sardelis MR, Turell MJ, Dohm DJ, O'Guinn ML. Vector competence of selected
North American Culex and Coquillettidia mosquitoes for West Nile virus.
Emerg Infect Dis 2001, 7(6):1018-1022.
Savini G, Monaco F, Terregino C, Di Gennaro A, Bano L, Pinoni C, De Nardi R,
Bonilauri P, Pecorari M, Di Gialleonardo L, Bonfanti L, Polci A, Calistri P, Lelli
R. Usutu virus in Italy: an emergence or a silent infection? Vet Microbiol
2011, 151(3-4):264-274.
Sbrana E, Tonry JH, Xiao SY, da Rosa AP, Higgs S, Tesh RB. Oral transmission of
West Nile virus in a hamster model. Am J Trop Med Hyg 2005, 72(3):325-329.
Sejvar JJ. The long-term outcomes of human West Nile virus infection. Clin
Infect Dis 2007, 44(12):1617-1624.
Senne DA, Pedersen JC, Hutto DL, Taylor WD, Schmitt BJ, Panigrahy B.
Pathogenicity of West Nile virus in chickens. Avian Dis 2000, 44(3):642-649.
Shieh WJ, Guarner J, Layton M, Fine A, Miller J, Nash D, Campbell GL, Roehrig JT,
Gubler DJ, Zaki SR. The role of pathology in an investigation of an
outbreak of West Nile encephalitis in New York, 1999. Emerg Infect Dis
2000, 6(4):370-372.
Shirato K, Miyoshi H, Goto A, Ako Y, Ueki T, Kariwa H, Takashima I. Viral
envelope protein glycosylation is a molecular determinant of the
neuroinvasiveness of the New York strain of West Nile virus. J Gen Virol
2004, 85(Pt 12):3637-3645.
Shrestha B, Gottlieb D, Diamond MS. Infection and injury of neurons by West
Nile encephalitis virus. J Virol 2003, 77(24):13203-13213.
Smithburn KC, Hughes TP, Burke AW, Paul JH. A neurotropic virus isolated from
the blood of a native of Uganda. Am J Trop Med Hyg 1940, s1-20(4):471-492.

55

Virginia Gamino Rodrguez

Solomon T, Winter PM. Neurovirulence


encephalitis--evidence from clinical
2004(18):161-170.

Tesis doctoral

and host factors in flavivirus


epidemiology. Arch Virol Suppl

Sotelo E, Fernndez-Pinero J, Llorente F, Agero M, Hoefle U, Blanco JM, JimnezClavero MA. Characterization of West Nile virus isolates from Spain: new
insights into the distinct West Nile virus eco-epidemiology in the Western
Mediterranean. Virology 2009, 395(2):289-297.
Sotelo E, Fernndez-Pinero J, Llorente F, Vzquez A, Moreno A, Agero M, Cordioli
P, Tenorio A, Jimnez-Clavero MA. Phylogenetic relationships of Western
Mediterranean West Nile virus strains (1996-2010) using full-length
genome sequences: single or multiple introductions? J Gen Virol 2011a,
92(Pt 11):2512-2522.
Sotelo E, Gutirrez-Guzmn AV, Del Amo J, Llorente F, El-Harrak M, Prez-Ramrez
E, Blanco JM, Hfle U, Jimnez-Clavero MA. Pathogenicity of two recent
Western Mediterranean West Nile virus isolates in a wild bird species
indigenous to Southern Europe: the red-legged partridge. Vet Res 2011b,
42(1):11. doi: 10.1186/1297-9716-42-11.
Steele KE, Linn MJ, Schoepp RJ, Komar N, Geisbert TW, Manduca RM, Calle PP,
Raphael BL, Clippinger TL, Larsen T, Smith J, Lanciotti RS, Panella NA,
McNamara TS. Pathology of fatal West Nile virus infections in native and
exotic birds during the 1999 outbreak in New York City, New York. Vet
Pathol 2000, 37(3):208-224.
Steinman A, Banet-Noach C, Simanov L, Grinfeld N, Aizenberg Z, Levi O, Lahav D,
Malkinson M, Perk S, Shpigel NY. Experimental infection of common garter
snakes (Thamnophis sirtalis) with West Nile virus. Vector Borne Zoonotic Dis
2006, 6(4):361-368.
Steinmetz H, Bakonyi T, Chvala S, Weissenbck H, Eulenberger U, Hatt J, Robert
N, Hoop R, Nowotny N: Emergence of Usutu virus in Switzerland. In:
Proceedings 43rd international symposium on diseases of zoo and wild animals:
16-20 May 2007; Edinburgh; 2007:129-131.
Steinmetz HW, Bakonyi T, Weissenbock H, Hatt JM, Eulenberger U, Robert N, Hoop
R, Nowotny N. Emergence and establishment of Usutu virus infection in
wild and captive avian species in and around Zurich, Switzerland--genomic
and pathologic comparison to other central European outbreaks. Vet
Microbiol 2011, 148(2-4):207-212.
Stewart BS, Demarest VL, Wong SJ, Green S, Bernard KA. Persistence of virusspecific immune responses in the central nervous system of mice after
West Nile virus infection. BMC Immunol 2011, 12:6. doi: 10.1186/1471-217212-6.
Stewart PA, Wiley MJ. Structural and histochemical features of the avian bloodbrain barrier. J Comp Neurol 1981, 202(2):157-167.
Suthar MS, Diamond MS, Gale M, Jr. West Nile virus infection and immunity.
Nat Rev Microbiol 2013, 11(2):115-128.
56

Introduccin

Swayne DE, Beck JR, Zaki S. Pathogenicity of West Nile virus for turkeys. Avian
Dis 2000, 44(4):932-937.
Swayne DE, Beck JR, Smith CS, Shieh WJ, Zaki SR. Fatal encephalitis and
myocarditis in young domestic geese (Anser anser domesticus) caused by
West Nile virus. Emerg Infect Dis 2001, 7(4):751-753.
Tag-El-Din-Hassan HT, Sasaki N, Moritoh K, Torigoe D, Maeda A, Agui T. The
chicken 2'-5' oligoadenylate synthetase A inhibits the replication of West
Nile virus. Jpn J Vet Res 2012, 60(2-3):95-103.
Tamba M, Bonilauri P, Bellini R, Calzolari M, Albieri A, Sambri V, Dottori M,
Angelini P. Detection of Usutu virus within a West Nile virus surveillance
program in Northern Italy. Vector Borne Zoonotic Dis 2011, 11(5):551-557.
Tesh RB, Travassos da Rosa AP, Guzman H, Araujo TP, Xiao SY. Immunization
with heterologous flaviviruses protective against fatal West Nile
encephalitis. Emerg Infect Dis 2002, 8(3):245-251.
Thongtan T, Cheepsunthorn P, Chaiworakul V, Rattanarungsan C, Wikan N, Smith
DR. Highly permissive infection of microglial cells by Japanese
encephalitis virus: a possible role as a viral reservoir. Microbes Infect 2010,
12(1):37-45.
Totani M, Yoshii K, Kariwa H, Takashima I. Glycosylation of the envelope protein
of West Nile Virus affects its replication in chicks. Avian Dis 2011,
55(4):561-568.
Traore-Lamizana M, Zeller HG, Mondo M, Hervy JP, Adam F, Digoutte JP.
Isolations of West Nile and Bagaza viruses from mosquitoes (Diptera:
Culicidae) in central Senegal (Ferlo). J Med Entomol 1994, 31(6):934-938.
Turell MJ, O'Guinn M, Oliver J. Potential for New York mosquitoes to transmit
West Nile Virus. Am J Trop Med Hyg 2000, 62(3):317-324.
Turtle L, Griffiths MJ, Solomon T. Encephalitis caused by flaviviruses. QJM 2012,
105(3):219-223.
Valiakos G, Touloudi A, Athanasiou LV, Giannakopoulos A, Iacovakis C, Birtsas P,
Spyrou V, Dalabiras Z, Petrovska L, Billinis C. Serological and molecular
investigation into the role of wild birds in the epidemiology of West Nile
virus in Greece. Virol J 2012, 9:266. doi: 10.1186/1743-422X-9-266.
van der Meulen KM, Pensaert MB, Nauwynck HJ. West Nile virus in the
vertebrate world. Arch Virol 2005, 150(4):637-657.
Vzquez A, Snchez-Seco MP, Ruiz S, Molero F, Hernndez L, Moreno J, Magallanes
A, Tejedor CG, Tenorio A. Putative new lineage of west nile virus, Spain.
Emerg Infect Dis 2010, 16(3):549-552.
Vzquez A, Ruiz S, Herrero L, Moreno J, Molero F, Magallanes A, Snchez-Seco MP,
Figuerola J, Tenorio A. West Nile and Usutu viruses in mosquitoes in Spain,
2008-2009. Am J Trop Med Hyg 2011, 85(1):178-181.

57

Virginia Gamino Rodrguez

Tesis doctoral

Verma S, Lo Y, Chapagain M, Lum S, Kumar M, Gurjav U, Luo H, Nakatsuka A,


Nerurkar VR. West Nile virus infection modulates human brain
microvascular endothelial cells tight junction proteins and cell adhesion
molecules: Transmigration across the in vitro blood-brain barrier. Virology
2009, 385(2):425-433.
Wang Y, Lobigs M, Lee E, Mullbacher A. CD8+ T cells mediate recovery and
immunopathology in West Nile virus encephalitis. J Virol 2003,
77(24):13323-13334.
Wang T, Town T, Alexopoulou L, Anderson JF, Fikrig E, Flavell RA. Toll-like
receptor 3 mediates West Nile virus entry into the brain causing lethal
encephalitis. Nat Med 2004, 10(12):1366-1373.
Wee Yong V. Inflammation in neurological disorders: a help or a hindrance?
Neuroscientist 2010, 16(4):408-420.
Weingartl HM, Neufeld JL, Copps J, Marszal P. Experimental West Nile virus
infection in blue jays (Cyanocitta cristata) and crows (Corvus
brachyrhynchos). Vet Pathol 2004, 41(4):362-370.
Weissenbck H, Kolodziejek J, Url A, Lussy H, Rebel-Bauder B, Nowotny N.
Emergence of Usutu virus, an African mosquito-borne flavivirus of the
Japanese encephalitis virus group, central Europe. Emerg Infect Dis 2002,
8(7):652-656.
Weissenbck H, Kolodziejek J, Fragner K, Kuhn R, Pfeffer M, Nowotny N. Usutu
virus activity in Austria, 2001-2002. Microbes Infect 2003, 5(12):1132-1136.
Weissenbck H, Bakonyi T, Chvala S, Nowotny N. Experimental Usutu virus
infection of suckling mice causes neuronal and glial cell apoptosis and
demyelination. Acta Neuropathol 2004, 108(5):453-460.
Weissenbck H, Chvala S, Bakonyi T, Nowotny N: Emergence of Usutu virus in
central Europe: diagnosis, surveillance and epizoology. In: Emerging pests
and vector-borne disease in Europe. Edited by Takken W, Knols B. Wageningen:
Wageningen Academic Publishers; 2007:153-168.
Weissenbck H, Hublek Z, Bakonyi T, Nowotny N. Zoonotic mosquito-borne
flaviviruses: worldwide presence of agents with proven pathogenicity and
potential candidates of future emerging diseases. Vet Microbiol 2010, 140(34):271-280.
Weissenbck H, Bakonyi T, Rossi G, Mani P, Nowotny N. Usutu Virus, Italy, 1996.
Emerg Infect Dis 2013, 19(2):274-277.
Wheeler SS, Vineyard MP, Woods LW, Reisen WK. Dynamics of West Nile virus
persistence in House Sparrows (Passer domesticus). PLoS Negl Trop Dis
2012, 6(10):e1860. doi: 10.1371/journal.pntd.0001860.
Wicker JA, Whiteman MC, Beasley DW, Davis CT, Zhang S, Schneider BS, Higgs S,
Kinney RM, Barrett AD. A single amino acid substitution in the central
portion of the West Nile virus NS4B protein confers a highly attenuated
phenotype in mice. Virology 2006, 349(2):245-253.
58

Introduccin

Williams MC, Simpson DI, Haddow AJ, Knight EM. The isolation of West Nile
Virus from man and of Usutu virus from the birdbiting mosquito Mansonia
aurites (Theobald) in the Entebbe area of Uganda. Ann Trop Med Parasitol
1964, 58:367-374.
Wodak E, Richter S, Bago Z, Revilla-Fernandez S, Weissenbock H, Nowotny N,
Winter P. Detection and molecular analysis of West Nile virus infections in
birds of prey in the eastern part of Austria in 2008 and 2009. Vet Microbiol
2011, 149(3-4):358-366.
Woodall JP. The viruses isolated from arthropods at the East African Virus
Research Institute in the 26 years ending December 1963. Proc E Afr Acad
1964, 2.
Woolhouse MEJ, Gowtage-Sequeria S, Evans B. T16: quantitative analysis of the
characteristics of emerging and reemerging human pathogens. 2006.
[http://www.bis.gov.uk/foresight/MediaList/foresight/media%20library/BISPa
rtners/Foresight/docs/infectiousdiseases/~/media/BISPartners/Foresight/doc
s/infectious-diseases/t16.ashx]
Wnschmann A, Shivers J, Carroll L, Bender J. Pathological and
immunohistochemical
findings
in
American
crows
(Corvus
brachyrhynchos) naturally infected with West Nile virus. J Vet Diagn Invest
2004, 16(4):329-333.
Wnschmann A, Ziegler A. West Nile virus-associated mortality events in
domestic Chukar partridges (Alectoris chukar) and domestic Impeyan
pheasants (Lophophorus impeyanus). Avian Dis 2006, 50(3):456-459.
Xiao SY, Guzman H, Zhang H, Travassos da Rosa AP, Tesh RB. West Nile virus
infection in the golden hamster (Mesocricetus auratus): a model for West
Nile encephalitis. Emerg Infect Dis 2001, 7(4):714-721.
Ye J, Zhu B, Fu ZF, Chen H, Cao S. Immune evasion strategies of flaviviruses.
Vaccine 2013, 31(3):461-471.
Zeller H. Is Japanese encephalitis emerging in Europe? Euro Surveill 2012,
17(32).
Zhang B, Patel J, Croyle M, Diamond MS, Klein RS. TNF-alpha-dependent
regulation of CXCR3 expression modulates neuronal survival during West
Nile virus encephalitis. J Neuroimmunol 2010, 224(1-2):28-38.
Ziegler U, Angenvoort J, Fischer D, Fast C, Eiden M, Rodriguez AV, RevillaFernandez S, Nowotny N, de la Fuente JG, Lierz M, Groschup MH.
Pathogenesis of West Nile virus lineage 1 and 2 in experimentally infected
large falcons. Vet Microbiol 2013, 161(3-4):263-273.

59

Objetivos

Objetivos

Como se ha expuesto anteriormente, en las ltimas dcadas estn incrementado de


forma importante los casos de mortalidad en aves silvestres asociados a la infeccin
por virus del gnero Flavivirus tanto en Norteamrica como en Europa, siendo
Espaa uno de los pases en los que se han registrado mortalidades en los ltimos
aos. Sin embargo, aunque se barajan numerosas posibilidades, las causas que
han llevado a esta situacin continan siendo objeto de estudio. Por otro lado,
aunque WNV es un flavivirus considerado como patgeno importante para las aves,
se ha visto que la infeccin no afecta a todas las especies por igual y que,
aparentemente, las consecuencias de dicha infeccin son menores para las aves
nativas europeas que para las que habitan en Norteamrica. A pesar de esto,
apenas existen trabajos en los que se estudie la patogenia o las lesiones asociadas a
la infeccin por WNV en aves endmicas de Europa. Finalmente, y aunque la
infeccin por WNV se caracteriza por dar lugar a un proceso neurolgico, no existen
trabajos que determinen los componentes celulares inflamatorios que intervienen
en el desarrollo de la encefalitis en aves. Por lo tanto, los objetivos de esta tesis
doctoral son:

Objetivo general
Estudiar los cambios morfolgicos y el tropismo celular y tisular del virus en aves
infectadas por flavivirus.

Objetivos especficos
1) Evaluar la informacin disponible sobre la patogenia y las lesiones asociadas
a la infeccin por WNV en aves para determinar los mecanismos patognicos
de los que deriva la variacin interespecie en la presentacin clnica de la
enfermedad y la mortalidad asociada (Captulo 1).

63

Virginia Gamino Rodrguez

Tesis doctoral

2) Describir la patogenia de la infeccin experimental con tres cepas diferentes


de WNV en perdiz roja. Determinar los componentes celulares inflamatorios
que intervienen en el desarrollo de la encefalitis as como su evolucin
durante el curso de la infeccin (Captulos 2 y 3).

3) Estudiar las diferencias en la patogenia, severidad de lesiones y nivel de


replicacin viral en la infeccin experimental con tres cepas diferentes de
WNV en perdiz roja. Determinar la existencia de diferencias en la
susceptibilidad, patogenia y lesiones asociadas a la infeccin experimental de
un ave endmica europea con WNV respecto a lo descrito en aves de
Norteamrica (Captulos 2 y 3).

4) Describir las lesiones y la distribucin tisular del virus en la infeccin


natural de perdiz roja, faisn comn y paloma torcaz por BAGV. Determinar
las existencia de diferencias interespecie en relacin a estos aspectos
(Captulo 4).

5) Caracterizar las lesiones y la distribucin del antgeno viral en secciones


oculares de aves galliformes cinegticas infectadas con WNV y BAGV.
Determinar la va de entrada de WNV al ojo y los hallazgos microscpicos que
puedan explicar la ceguera observada en perdices rojas infectadas por BAGV
(Captulo 5).

6) Caracterizar el primer caso de infeccin por USUV en aves en Espaa.


Determinar el origen de la cepa infectante y describir brevemente las lesiones
asociadas a la infeccin (Captulo 6).

64

Captulo 1
Revisin sobre la patologa y el tropismo
tisular del virus West Nile en aves
infectadas de forma natural

Gamino V, Hfle U. Pathology and tissue tropism of natural West Nile


virus infection in birds: a review. Veterinary Research 2013, 44(1):39.
doi: 10.1186/1297-9716-44-39.

Captulo 1

RESUMEN
El virus West Nile (WNV) es un flavivirus transmitido por artrpodos distribuido
mundialmente y capaz de infectar a una gran variedad de hospedadores
vertebrados, siendo las aves su reservorio natural. Aunque este virus haba sido
considerado un patgeno de escasa importancia para las aves, desde la dcada de
los 90, y especialmente tras ser introducido en el continente norteamericano en
1999, miles de aves han muerto como consecuencia de la infeccin. En la presente
revisin se realiza una sntesis sobre la patogenia y la patologa asociadas a la
infeccin por WNV en aves, resaltando las diferencias en la distribucin y severidad
de lesiones y la presencia de antgeno viral entre rdenes y familias de aves. A pesar
de la diferencias encontradas entre especies en la susceptibilidad a la infeccin, en
un ave infectada por WNV existen lesiones y antgeno viral en la mayora de los
rganos. Las diferencias encontradas en cuanto a la distribucin y severidad de
lesiones y la presencia de antgeno viral estn relacionadas, en parte, con el curso
de la infeccin, segn sea sta no progresiva, aguda o crnica. Es muy probable que
la patogenia se vea modulada por la combinacin de numerosas variables del
hospedador, factores ambientales y factores intrnsecos del virus como la virulencia
y la patogenicidad de la cepa infectante.

67

Virginia Gamino Rodrguez

Tesis doctoral

ABSTRACT
West Nile virus (WNV) is a globally distributed arthropod-borne flavivirus capable of
infecting a wide variety of vertebrates, with birds as its natural reservoir. Although
it had been considered a pathogen of little importance for birds, from the 1990s,
and especially after its introduction in the North American continent in 1999,
thousands of birds have succumbed to West Nile infection. This review summarizes
the pathogenesis and pathology of WNV infection in birds highlighting differences in
lesion and antigen distribution and severity among bird orders and families. Despite
significant species differences in susceptibility to infection, WNV associated lesions
and viral antigen are present in the majority of organs of infected birds. The nonprogressive, acute or more prolonged course of the disease accounts for part of the
differences in lesion and viral antigen distribution and lesion severity. Most likely a
combination of host variables and environmental factors in addition to the intrinsic
virulence and pathogenicity of the infecting WNV strain influence the pathogenesis
of the infection.

68

Captulo 1

INTRODUCTION
West Nile virus (WNV) is an arthropod-borne virus of the genus Flavivirus capable of
infecting a wide variety of vertebrates, with birds as its natural reservoir (McLean
and Ubico, 2007). It was first isolated in Uganda in 1937 from a woman with a
febrile process (Smithburn et al., 1940). During the 1960s WNV was one of the
most widely distributed flaviviruses in humans, birds and mosquitoes in Africa, the
Middle East and south-western Europe, where it was considered a pathogen of little
importance, causing subclinical infection or sporadic self-limiting outbreaks in
horses and humans (Murgue et al., 2002; Dauphin et al., 2004). From the 1990s
the frequency and severity of human infections as well as the number of cases in
other vertebrates including companion, farm and wild animals has increased (van
der Meulen et al., 2005). In Europe, WNV causes disease in horses and humans,
and recently sporadic mortality events have been reported primarily in birds of prey
(Bakonyi et al., 2006; Hfle et al., 2008; Jimnez-Clavero et al., 2008). In contrast,
after its introduction in North America in 1999, where it caused one of the most
important outbreaks in New York, thousands of birds, horses and humans have
died of the disease (CDCa; LaDeau et al., 2007). Nowadays it is one of the most
widely distributed flaviviruses in the world and an important public and animal
health and conservation concern.

ETIOLOGY
West Nile virus belongs to the genus Flavivirus of the family Flaviviridae. It
has been serologically classified within the Japanese encephalitis antigenic group.
The viral positive sense single-stranded RNA genome encodes a polyprotein which is
translated into different structural (envelope protein E, membrane precursor
protein prM and capsid protein C) and non-structural (NS1, NS2a, NS2b, NS3,
NS4a, NS4b, and NS5) proteins that play an important role in the viral host range,
69

Virginia Gamino Rodrguez

Tesis doctoral

tissue tropism, viral replication and assembly, and host immune system
stimulation (Deubel et al., 2001; Brinton, 2002; Lindenbach and Rice, 2003). Based
on phylogenetic analysis, WNV strains have been grouped into seven genetic
lineages (Mackenzie and Williams, 2009) but there are two major lineages, lineage 1
and lineage 2. Lineage 1 is widespread and contains isolates from Europe, America,
the Middle East, India, Africa and Australia (Lanciotti et al., 2002; Charrel et al.,
2003). Lineage 2 had originally only been isolated in Southern Africa and
Madagascar but has recently also been detected in Europe (Bakonyi et al., 2006;
Papa et al., 2011a; Valiakos et al., 2011; Savini et al., 2012). In general lineage 1
viruses were considered to be more virulent than lineage 2 viruses, however it has
been demonstrated that both lineages contain neuroinvasive phenotypes and some
of the recent outbreaks in Europe, that also affect birds, have been caused by the
latter (Bakonyi et al., 2006; Wodak et al., 2011; Savini et al., 2012) (Table 1).
Increased virulence of WNV strains has been associated to genetic and aminoacidic
changes in structural and non-structural proteins (Charrel et al., 2003; Sotelo et
al., 2009; McMullen et al., 2013). Substitution to a proline in position 249 of the
non-structural protein NS3 has been associated with increased virulence in WNV
lineage 1 strains in American crows (Corvus brachyrhynchos) (Brault et al., 2007)
and the same substitution has been detected in recent lineage 2 strains causing
important outbreaks in Europe (Papa et al., 2011b).

70

Captulo 1

Table 1. Bird species found dead or with symptoms of encephalitis in Europe in which the
presence of WNV has been evidenced by real time RT-PCR or by virus isolation.
Year

Country

Order

Family

Species

20012004

Spain

Falconiformes

Accipitridae

2003

Hungary

Anseriformes

Anatidae

2004

Hungary

Falconiformes

Accipitridae

France

Passeriformes

Corvidae

Spanish imperial
eagle
(Aquila adalberti)
Domestic goose
(Anser anser
domesticus)
Goshawk
(Accipiter gentilis)
Common magpie
(Pica pica)
House sparrow
(Passer
domesticus)
Goshawk

Passeridae

2005

Hungary

Falconiformes

Accipitridae

2007

Spain

Falconiformes

Accipitridae

2008

Austria

Falconiformes

Accipitridae

Sparrow hawk
(Accipiter nisus)
Golden eagle
(Aquila chrysaetos)
Bonellis eagle
(Aquila fasciata)
Sparrow hawk

Lineage
1

(Hfle et al., 2008)

(Glvits et al., 2005;


Bakonyi et al., 2006)

(Bakonyi et al., 2006;


Erdlyi et al., 2007)
(Jourdain et al.,
2007b)

(Bakonyi et al., 2006;


Erdlyi et al., 2007)

(Jimnez-Clavero et
al., 2008)

(DEFRA; Wodak et al.,


2011)

Goshawk
Falconidae

Italy

Psittaciformes

Strigopidae

Charadriiformes

Laridae

Columbiformes

Columbidae

Passeriformes

Corvidae

Suliformes

Phalacrocoracidae

2009

Austria

Falconiformes

Accipitridae

2011

Italy

Columbiformes

Columbidae

2012

Italy

Falconiformes

Accipitridae

Gyrfalcon
(Falco rusticolus)
Kea
(Nestor notabilis)
Herring gull
(Larus argentatus)
Pigeon
(Columba livia)
Common magpie
Carrion crow
(Corvus corone)
Eurasian jay
(Garrulus
glandarius)
Great cormorant
(Phalacrocorax
carbo)
Goshawk
Collared dove
(Streptopelia
decaocto)
Goshawk

Reference

(DEFRA)
1

(Monaco et al., 2010)

(Wodak et al., 2011)

(Savini et al., 2012)

(Savini et al., 2013)

ECO-EPIDEMIOLOGY
The virus is maintained in an enzootic ornitophilic-mosquito-bird cycle (Hublek
and Halouzka, 1999). Migratory birds are considered to play an important role in
the local and long distance viral dispersal (Malkinson and Banet, 2002; Peterson et

71

Virginia Gamino Rodrguez

Tesis doctoral

al., 2003; Rappole et al., 2006; Jourdain et al., 2007a). The most susceptible
species to infection belong to the family Corvidae of the order Passeriformes such as
the American crow, the blue jay (Cyanocitta cristata), the black-billed magpie (Pica
hudsonia) or the fish crow (Corvus ossifragus) (Komar et al., 2003; LaDeau et al.,
2007). Some species of this order such as the American robin (Turdus migratorius)
or the house sparrow (Passer domesticus) are also considered the main reservoir of
WNV in urban areas of North America and Europe (Langevin et al., 2005; Kilpatrick
et al., 2006). Other vertebrates susceptible to infection include amphibians, reptiles
and mammals, among which the virus can be especially pathogenic for equids and
humans. However, most of them are considered accidental or dead-end hosts as
they rarely develop sufficient viremia to infect feeding mosquitoes (van der Meulen
et al., 2005; Kramer et al., 2008).
Although WNV transmission in birds occurs primarily by mosquitoes of the
genus

Culex,

transmission

through

infected

prey

or

contaminated

water

consumption and horizontal contact transmission have also been documented


(Langevin et al., 2001; Swayne et al., 2001; Banet-Noach et al., 2003; Komar et al.,
2003; Nemeth et al., 2006a). The virus is maintained in a silent mosquito-bird cycle
in natural habitats (rural cycle) but can cause outbreaks in humans and horses
when it is introduced in humanized habitats where mosquitoes with a wider host
range are present, establishing an urban cycle (Jimnez-Clavero, 2012). In
endemic areas wild bird infection usually starts in spring and early summer,
mortality peaks from midsummer to early fall, and human and horse cases take
place a few weeks after the onset of bird mortalities (Phalen and Dahlhausen,
2004).
Before the 1990s WNV had been isolated in different parts of Europe, Asia
and Africa but was rarely associated with clinical disease. Although some studies
conducted in Africa in the 1950s underlined the role of birds as amplifying hosts
72

Captulo 1

and mortality was reported in host competence studies, especially in hooded crows
(Corvus corone sardonius) (Work et al., 1955), prior to 1997 WNV was considered
low-pathogenic for birds (Zeller and Schuffenecker, 2004). In 1998 a large outbreak
took place in Israel with high mortality in birds, particularly in geese (Anser anser
domesticus), and WNV was isolated from the brain of dead free-living storks (Ciconia
ciconia) (Malkinson et al., 2002). Currently, WNV is considered endemic in Europe
(Jimnez-Clavero, 2012). Although viral circulation has been reported in many bird
species, cases of encephalitis or mortality due to viral infection in wild birds have
been sporadic (Zeller and Schuffenecker, 2004) and observed mainly in raptors
(Table 1). In North America WNV was detected for the first time in 1999 in New York
(Lanciotti et al., 1999) and since then it has caused thousands of bird deaths being
detected in at least 326 bird species (CDCb) (Table 2). WNV became endemic within
ten years of its introduction in this continent (De Filette et al., 2012). The
epidemiological behavior in North America, with an intense viral circulation, differs
from the epidemiological behavior in other parts of the world including Europe, and
the reasons remain largely unknown (Jimnez-Clavero, 2012). Viral phenotype, host
heterogeneity in the area, vector abundance, feeding activity or host preference, and
host susceptibility have been identified as possible modulating factors (Ostfeld and
Keesing, 2000; Brinton, 2001, 2002; Ezenwa et al., 2006; Hamer et al., 2009;
Jimnez-Clavero, 2012). Host susceptibility to infection has been associated to
geographic range, mating and breeding systems, body size, migratory behavior and
to co-evolution with the virus or antigenically-related flaviviruses (Gancz et al.,
2004; Fang and Reisen, 2006; Reisen and Hahn, 2007; Figuerola et al., 2008; Kwan
et al., 2012).

73

Virginia Gamino Rodrguez

Tesis doctoral

Table 2. Bird orders and families from North America in which mortality due to WNV infection
has been demonstrated.
ORDER

FAMILY

Anseriformes

Anatidae

Apodiformes

Apodidae, Trochilidae

Caprimulgiformes

Caprimulgidae

Casuariiformes

Dromaiidae

Charadriiformes

Charadriidae, Laridae

Ciconiiformes

Ardeidae, Cathartidae, Ciconiidae, Threskiornithidae

Columbiformes

Columbidae

Coraciiformes

Alcedinidae, Bucerotidae

Cuculiformes

Cuculidae

Falconiformes

Accipitridae, Falconidae

Galliformes

Numididae, Odontophoridae, Phasianidae

Gaviiformes

Gaviidae

Gruiformes

Aramidae, Gruidae, Rallidae

Musophagiformes

Musophagidae

Passeriformes

Bombycillidae, Cardinalidae, Cinclidae, Corvidae, Emberizidae, Estrildidae,


Fringillidae, Icteridae, Laniidae, Mimidae, Paridae, Parulidae, Passeridae,
Sittadae, Sturnidae, Thraupidae, Troglodytidae, Turdidae, Vireonidae

Pelecaniformes

Pelecanidae, Phalacrocoracidae

Phoenicopteriformes

Phoenicopteridae

Piciformes

Picidae

Podicipediformes

Podicipedidae

Psittaciformes

Aratingidae, Cacatuidae, Psittacidae

Sphenisciformes

Spheniscidae

Strigiformes

Strigidae, Tytonidae

Struthioniformes

Struthionidae

Tinamiformes

Tinamidae

PATHOGENESIS IN BIRDS
Most information about the pathogenesis of WNV infection is derived from
experimental studies done in mammals, mainly rodents.
In mammals, after mosquito blood-feeding, WNV replicates in the skin and is
transported by Langerhans dendritic cells to draining lymph nodes. There the virus
replicates and primary viremia and peripheral organ dissemination take place
(Samuel and Diamond, 2006). On the contrary, the exact mechanism and sites of

74

Captulo 1

WNV replication in avian hosts are still not well understood. It is supposed that,
like in mammals, it replicates locally at the inoculation site and is rapidly
distributed to all organ systems (Nemeth et al., 2011). However, it has been
demonstrated that the virus can be detected in the blood as early as 30-45 min
after the mosquito feeding period, suggesting that in birds local replication is not
necessary for the primary viremia (Reisen et al., 2007). In general, WNV can be
isolated from the blood of infected birds at one day post-infection (dpi) (one day
later in less susceptible species such as chicken or turkeys or in case of oral
infection) (Senne et al., 2000; Komar et al., 2003; Weingartl et al., 2004). Viremia
can peak as early as 2-3 dpi in geese and some Passeriformes such as crows and
jays, or 4-6 dpi in raptors, owls and chicken (Senne et al., 2000; Swayne et al.,
2001; Weingartl et al., 2004; Nemeth et al., 2006b; Ziegler et al., 2013). Mean peak
viremia is higher in those birds that finally succumb to the disease (Langevin et al.,
2005). WNV can be detected in the blood until day 6-7 pi in geese, passerines and
owls and up to 10 dpi in raptors and turkeys (Swayne et al., 2000; Banet-Noach et
al., 2003; Nemeth et al., 2006b; Lapointe et al., 2009; Ziegler et al., 2013).
Little is known about innate avian host defenses against WNV infection. In
mammals this includes IFN production, complement activation, phagocytosis and
cytotoxicity, with the participation of macrophages, neutrophils, NK cells and T
cells (Samuel and Diamond, 2006; Suthar et al., 2013).
WNV infects all major organ systems and a wide variety of individual cell
types. The targeting of cells of the mononuclear phagocytic system may play an
important role in the pathogenesis in infected birds, as the virus can replicate
within these cells and disseminate to a wide variety of tissues (Steele et al., 2000;
Weingartl et al., 2004). As early as 1 dpi WNV can be detected in the spleen of crows
and one day later it is widely distributed. In this species the highest viral titer in
tissues can be reached on day 4 pi, decreasing from day 5 pi (Weingartl et al.,
75

Virginia Gamino Rodrguez

Tesis doctoral

2004). On the contrary, in falcons and owls maximum viral titer in tissues can be
delayed until day 7-8 pi, decreasing from day 9 pi, and while in falcons the virus
remains detectable on day 14 pi (Busquets et al., 2012), it can be totally cleared
from most tissues of owls (Nemeth et al., 2006a,b). Flaviviruses have evolved
different strategies that modulate the host immune response (Ye et al., 2013) and
depending on the effectiveness of this response and, therefore, viremia level, WNV
can reach the central nervous system (CNS). While WNV can be detected in the
brain of crows as early as 2 dpi, in owls it may not be present before 5 dpi
(Weingartl et al., 2004; Nemeth et al., 2006b). In the former, maximum viral titer
can be reached on day 4 pi while this may not take place in the latter until day 8 pi
or until 14 dpi in falcons (Weingartl et al., 2004; Nemeth et al., 2006b; Busquets et
al., 2012). The mechanisms by which WNV crosses the blood-brain barrier and
reaches the CNS remain unknown. Some authors have suggested that TNF-
mediated change in endothelial cell permeability may facilitate its entry, at least in
mammals (Wang et al., 2004). Other mechanisms proposed are the infection of or
passive transport through the endothelium or epithelial cells of the choroid plexus
(McMinn, 1997), direct axonal retrograde transport from olfactory or motor neurons
(Monath et al., 1983; Samuel et al., 2007) or transport by infected immune cells
(Garca-Tapia et al., 2006). In birds, WNV detection in endothelial cells by
immunohistochemistry (IHC) and the presence of perivascular inflammatory
infiltrates (Wnschmann et al., 2004a; Lopes et al., 2007) may indicate that WNV
reaches the CNS via the blood stream and infects endothelial cells, although viral
transport through infected immune cells may also be probable (Weingartl et al.,
2004).
The development of clinical disease is caused by the invasion of the CNS
and/or other major organs such as the liver, spleen, kidney and heart (Steele et al.,
2000). In most cases, clinical signs appear approximately on 5 dpi in experimentally
76

Captulo 1

WNV infected birds, but may be absent both in experimental and natural infections
(Fitzgerald et al., 2003; Komar et al., 2003; Bertelsen et al., 2004; Nemeth et al.,
2006a; Wnschmann and Ziegler, 2006; Nemeth et al., 2011; Sotelo et al., 2011).
Unspecific clinical signs include depression, anorexia, dehydration and ruffled
feathers. In more than 60% of infections convulsions are present, approximately in
30% appear ataxia, abnormal head posture and head movements, and in up to 20%
tremors, uncoordinated flight, paresis and disorientation are observed (Steele et al.,
2000; Fitzgerald et al., 2003; D'Agostino and Isaza, 2004; Wnschmann et al.,
2004b; Joyner et al., 2006; Saito et al., 2007; Palmieri et al., 2011). The
development of impaired vision and blindness is common in raptors and owls
(Wnschmann et al., 2004b, 2005; Gancz et al., 2006). Long-term sequelaes have
been detected in long-lived birds such as raptors, in which relapses of neurologic
signs, feather pulp abnormalities and abnormal molt can persist up to 4 years
(Nemeth et al., 2009a), and may have a negative impact on the longevity of these
species (Nemeth et al., 2006a, 2009a).
Systemic infection of the host also activates the adaptive immune response
that includes B and T cell activation and antibody production (Samuel and
Diamond, 2006; Suthar et al., 2013). In most bird species seroconversion can first
be detected between 4-6 dpi (Weingartl et al., 2004; Sotelo et al., 2011) and
neutralizing antibodies, at least in some species, persist for longer than a year and
can be transferred from adult females to their progeny (Gibbs et al., 2005a; Hahn et
al., 2006; Wilcox et al., 2007; Nemeth et al., 2009b). In mice it has been indicated
that CD8+ and CD4+ T lymphocytes combined with antibodies and chemokines are
essential for viral clearance from the CNS and peripheral organs (Lobigs et al.,
2009). In birds it has been demonstrated that the virus can persist in different
organs such as the spleen, kidney, eye, brain or the skin (Komar et al., 2003;
Reisen et al., 2006; Nemeth et al., 2009c; Wheeler et al., 2012a,b). Wheeler et al.
77

Virginia Gamino Rodrguez

Tesis doctoral

(2012a) demonstrated that viral persistence in naturally infected Passeriformes can


last for 4 months extending to up to 6 months in experimentally infected birds.
Viral persistence consequences for the avian host are still not clear (Wheeler et al.,
2012a) but it can play an important role in viral overwintering and mosquito
infection in case of host immune defenses impairment and viremia recrudescence
(Semenov et al., 1973; Reisen et al., 2006; Wheeler et al., 2012c).
In case of host defense failure, death often occurs within 24 hours or two to
four days after the onset of clinical signs in experimentally or naturally infected
birds, respectively (Komar et al., 2003; Glvits et al., 2005; Wnschmann and
Ziegler, 2006; Erdlyi et al., 2007). Sometimes death is not directly related to WNV
associated tissue lesions but to concurrent disease such as trauma or bacterial,
fungal or parasitic infections (Steele et al., 2000; Wnschmann et al., 2005; Nemeth
et al., 2006a; Saito et al., 2007; Hfle et al., 2008; Nemeth et al., 2009a).
The outcome of WNV infection and development of lesions and disease in
individual birds depend mainly on viral and host factors. Glycosylation in the E
protein of WNV has been demonstrated to increase peripheral viremia and virulence
for birds and neuroinvasiveness in rodents (Shirato et al., 2004; Murata et al.,
2010). Brault et al. (2007) experimentally demonstrated that a T249P NS3
substitution in the WNV NY99 strain increased virulence for American crows.
However, this substitution appears not to have the same effect for some other North
American bird species and an experimental study has shown that this substitution
alone in European WNV strains does not necessarily lead to an increased virulence
for susceptible European birds (Sotelo et al., 2011). Papa et al. (2011b) related the
H249P NS3 substitution to the increased virulence of the lineage 2 WNV strain that
caused the outbreak in humans in Greece in 2010 (Papa et al., 2010), but the role
of this substitution in the virulence for birds is not clear, as lineage 2 strains that
have caused mortalities in birds do not show this change (Papa et al., 2011b; Savini
78

Captulo 1

et al., 2012, 2013). Therefore, the presence of other genetic markers of virulence
could be implicated (Botha et al., 2008; McMullen et al., 2013). The existence of
natural infections of goshawks (Accipiter gentilis) by either WNV lineage 1 and 2
strains allows comparison of the associated pathology. In both cases viral cellular
and tissue tropism is similar but lesion localization and nature slightly differ. Thus
for example splenic lymphoid depletion, nephritis and hepatitis present in lineage 2
infected goshawks are not described in those infected by lineage 1 which in change
have hepatic and myocardial necrosis (Wnschmann et al., 2005; Erdlyi et al.,
2007; Wodak et al., 2011). Another determinant factor is the ability of the WNV
strain to replicate at the high corporal temperature of the avian host, as has been
demonstrated for the WNV NY99 genotype (Kinney et al., 2006). Moreover, viral
inoculation dose should not be forgotten, although dose-dependent pathological
differences are not always evidenced in experimental infections (Ziegler et al., 2013).
Finally, infection route is other viral factor that can influence WNV pathogenicity.
Some authors have indicated that in orally infected birds, tissue lesions are less
severe and the incidence of clinical signs or mortality is lower than in those
subcutaneously infected (Komar et al., 2003; Nemeth et al., 2006a).
Bird species is an important host factor that influences the pathogenesis of
WNV infection, as has been demonstrated by numerous authors (Steele et al., 2000;
Komar et al., 2003; Nemeth et al., 2006a; Wnschmann and Ziegler, 2006; Lopes et
al., 2007). Also, the immune status of the host plays an essential role in the
pathogenesis, as it determines the capacity of the host to clear the virus (Samuel
and Diamond, 2006). It can be modulated by the presence of previous immunity
against the virus or cross immunity against antigenically-related flaviviruses, the
presence of concurrent diseases, the influence of hormonal factors and stress, as
well as age (Eldadah et al., 1967; Langevin et al., 2001; Fang and Reisen, 2006;
Wnschmann and Ziegler, 2006; Nemeth and Bowen, 2007; Nemeth et al., 2008;
79

Virginia Gamino Rodrguez

Tesis doctoral

Shirafuji et al., 2009). Age not only determines the maturity of the immune system
but also the expression of cellular receptors that play an important role on viral
entry and intracellular factors that participate in the pathogenesis of the disease
(Chambers and Diamond, 2003). Finally, it has been demonstrated in avian and
rodent models that genetic factors influence host susceptibility to flaviviral
infections (Sangster et al., 1994; Tag-El-Din-Hassan et al., 2012).

PATHOLOGY OF NATURAL WNV INFECTION IN BIRDS


The pathology of natural WNV infection in birds has been documented in more than
57 species of at least 13 families and 10 orders.
Because of the wide cellular and tissue tropism of WNV, there are no
pathognomonic macroscopic lesions. Bird species with low susceptibility to WNV
infection such as chicken may have no observable macroscopic lesions at necropsy
(Senne et al., 2000) and those highly susceptible such as crows or jays will die
rapidly after a short incubation period and may have few acute or no observable
lesions (Bertelsen et al., 2004; Wnschmann et al., 2004a). In contrast, birds that
survive longer will have more pronounced macroscopic lesions, that may be chronic
in the case of individuals that survive days to months (Wnschmann et al., 2005;
Lopes et al., 2007). Emaciation, dehydration, multiorgan hemorrhages, petechiae
and congestion are the most characteristic macroscopic changes, but also
splenomegaly, hepatomegaly, myocardial pallor and pale mottling in the liver,
spleen or kidney. Cerebral atrophy and malacia can be observed in raptors (Steele
et al., 2000; Wnschmann et al., 2005; Nemeth et al., 2006a; Wnschmann and
Ziegler, 2006; Lopes et al., 2007; Palmieri et al., 2011).
In the vast majority of bird families, microscopic lesions due to WNV infection
are predominantly found in the CNS, heart, kidney, spleen and liver (Table 3).
Pathological changes can be the result of the direct effect of the virus or secondary
80

Captulo 1

to the host inflammatory response (Chambers and Diamond, 2003; Pauli et al.,
2007; Lim et al., 2011). Lymphoplasmacytic and histiocytic infiltrates, cellular
degeneration and necrosis, and hemorrhages are the main microscopic findings.
However, there are differences in antigen abundance and distribution and in
microscopic lesion severity and distribution depending on the species infected or
the period between infection and death of the analyzed individual (McLean and
Ubico, 2007) (Table 3 and figure 1). Highly susceptible species such as crows and
jays usually have large amounts of virus in the blood and WNV antigen is widely
distributed in major organs. Microscopic changes in these cases are acute with
minimal inflammatory reaction and can be absent in the CNS (Weingartl et al.,
2004; Wnschmann et al., 2004a; Gibbs et al., 2005b). In contrast, birds that
survive longer, such as raptors and owls, may develop chronic lesions that can be
also found in the CNS (Wnschmann et al., 2005; Nemeth et al., 2006a).
120

Percentage of tissues

100

n=15
n=14

n=4 n=9

n=2

n=18
n=18

n=10
n=10

n=13
n=15

n=7

n=7

n=10

n=19 n=19 n=19

n=19

80

60

n=10

n=10

Lesion
Antigen

40

20

Bird order

Figure 1. Lesion extension and antigen distribution among examined tissues in WNV infected
bird orders as reported in the reviewed literature. Each column represents the percentage of
tissues collected (n) in which lesions or antigen is present.

81

Virginia Gamino Rodrguez

Tesis doctoral

Central nervous system. CNS lesions can appear on 8-9 dpi in falcons and owls,
increasing their severity during the course of infection, and can be maintained
chronically (Nemeth et al., 2006a,b; Busquets et al., 2012). In chicken, the
appearance of lesions can be delayed until 21 dpi (Senne et al., 2000).
Lymphoplasmacytic and histiocytic meningoencephalitis characterized by gliosis,
perivascular cuffing and glial nodules is the main microscopic finding. This
inflammation is usually present in the molecular layer of the cerebellum and in
some cases also in the cerebrum, mesencephalon and medulla oblongata. Neuronal
necrosis and degeneration are usually found in raptors and owls (Nemeth et al.,
2006a; Ellis et al., 2007). Vasculitis is detected in red-tailed hawks (Buteo
jamaicensis), yellow-billed magpies (Pica nuttalli) and house sparrows (Nemeth et
al., 2006a; Ernest et al., 2010; O'Brien et al., 2010) and hemorrhages are observed
in guanay cormorants (Phalacrocorax bougainvillea), turkeys and owls (Steele et al.,
2000; Gancz et al., 2006; Zhang et al., 2006) (Table 3). WNV antigen is usually
detected by IHC in the CNS. Cells showing antigen labeling include neurons
(including Purkinje cells) and glial cells as well as other inflammatory cells and
vascular endothelial cells.
Eye. The eye is not routinely microscopically studied due to the difficulty of
obtaining sections suitable for interpretation. However, it has been more closely
examined in raptors and owls, probably because of the importance of vision in these
species. Moderate to marked lymphoplasmacytic and histiocytic inflammatory
infiltrates, disarray of the retinal pigmented epithelial cell layer and retinal cell
necrosis and mineralization have been described in naturally WNV infected hawks
(Pauli et al., 2007). Birds of the order Psittaciformes and Strigiformes only show
inflammatory infiltrates that are mainly detected in the conjunctiva, iris, choroid,
pecten and retina (Gancz et al., 2006; Stockman et al., 2010) (Table 3). WNV
antigen is detected in different cells of the retina and in inflammatory infiltrating
82

Captulo 1

cells. Although Wnschmann et al. (2004a) described the presence of WNV antigen
in endothelial cells of the pecten and choroid in the eye of infected American crows,
they did not describe associated lesions in this species. In contrast, WNV antigen
presence has not been documented in the eye of Psittaciformes (Stockman et al.,
2010).
Peripheral nervous system. WNV related lesions in the peripheral nervous system
have only been described in raptors and owls (Ellis et al., 2007). Mononuclear or
mixed inflammation is usually present in the sciatic nerve and in the myenteric,
proventricular and ventricular ganglia (Table 3). Neuronal positivity can be detected
by IHC.
Heart. Myocardial lesions may not be detected in falcons until 14 dpi, but appear
earlier in owls (8 dpi) and crows (6 dpi) (Weingartl et al., 2004; Nemeth et al.,
2006b; Busquets et al., 2012). Lymphoplasmacytic and histiocytic myocarditis with
myocardial necrosis, degeneration, mineralization or fibrosis and hemorrhages are
the main microscopic findings. Vasculitis can be seen in blue jays and American
kestrels (Falco sparverius) (Gibbs et al., 2005b; Nemeth et al., 2006a) (Table 3).
Cardiomyocytes, inflammatory and endothelial cells, and smooth muscle cells of
arteries usually show immunostaining. Although the heart is usually affected in
WNV infected birds, Anseriformes rarely have lesions in this tissue, even though
viral antigen is demonstrated by IHC (Steele et al., 2000).
Spleen and other lymphoid organs. Splenic lesions can appear as early as 2 dpi in
jays and crows, increasing in severity as infection progresses (Weingartl et al., 2004)
but in falcons they may not be prominent until 7 dpi (Busquets et al., 2012). In
this organ, lymphoid necrosis or apoptosis with fibrin deposition and hemosiderosis
are

consistently

observed.

Hemorrhages

are

described

in

ducks,

guanay

cormorants, and black-crowned night heron (Nycticorax nycticorax) (Steele et al.,


2000) and lymphoid depletion in goshawks and owls (Gancz et al., 2004; Erdlyi et
83

Virginia Gamino Rodrguez

Tesis doctoral

al., 2007) (Table 3). Viral antigen is detected in dendritic cells/macrophages and
smooth muscular cells of arterioles. Necrosis can be found in bursal and thymic
lymphoid cells (Ellis et al., 2007; Himsworth et al., 2009) but these tissues have
been only analyzed in some naturally WNV exposed birds, most probably because
they are only present in juvenile birds (Table 3).
Liver. Similarly to the spleen, hepatic lesions can be found in crows as early as 3
dpi and increase in severity during the progress of the infection (Weingartl et al.,
2004). In owls and falcons on day 5 pi these lesions are mild changing to severe by
day 14 pi (Nemeth et al., 2006b; Busquets et al., 2012). Lymphoplasmacytic and
histiocytic hepatitis as well as coagulative hepatocyte necrosis are the most
frequent lesions. Hemorrhages in goshawks and blue jays (Gibbs et al., 2005b;
Erdlyi et al., 2007), biliary duct hyperplasia in northern bald eagles (Haliaeetus
leucocephalus alascanus) and Chilean flamingos (Phoenicopterus chilensis) (Steele et
al., 2000), and vasculitis in American kestrels can also be found (Nemeth et al.,
2006a) (Table 3). Birds that show hemosiderosis in the spleen usually also have it
in the liver (Zhang et al., 2006; Ellis et al., 2007; Ernest et al., 2010) (Table 3). WNV
antigen is mainly seen in hepatocytes and Kupffer cells. Anseriformes do not show
lesions (Glvits et al., 2005) but WNV antigen is detected by IHC.
Kidney. Renal lesions can be moderate on 6 dpi in crows and 9 dpi in owls but
may not become apparent until 14 dpi in falcons (Nemeth et al., 2006b; 2011;
Busquets et al., 2012). Lymphoplasmacytic and histiocytic interstitial nephritis and
tubular epithelial cell degeneration and/or necrosis are detected in most WNV
infected birds. Glomerular cell degeneration and/or necrosis are described in birds
of the order Psittaciformes and Strigiformes (Gancz et al., 2006; Palmieri et al.,
2011), hemorrhages in ducks (Himsworth et al., 2009) and vasculitis in American
kestrels (Nemeth et al., 2006b) (Table 3). IHC demonstrates WNV antigen in tubular

84

Captulo 1

and collecting duct epithelial cells, glomerular cells, infiltrating inflammatory cells,
interstitial fibroblasts and, less frequently, in endothelial cells.
Lung. WNV antigen labeling and associated lesions are not usually described in the
lung or if present they are mild. Lymphohistiocytic inflammatory infiltrates have
occasionally been described and interstitial edema and necrosis can also be found.
In naturally WNV infected Anseriformes and Psittaciformes pulmonary lesions have
not been documented (Himsworth et al., 2009; Stockman et al., 2010) (Table 3).
Viral antigen is detected mainly in inflammatory cells although it can also be found
in epithelial cells of the air capillaries.
Gastrointestinal tract. Lesions in the gastrointestinal tract are mainly described in
the proventriculus, ventriculus and intestines with enterocyte and intestinal crypt
cell necrosis, lymphoplasmacytic and histiocytic infiltrates, and proventricular
gland and ventricular hemorrhages as main microscopic findings. In owls, but even
less so in goshawks, lesions in the gastrointestinal tract are rarely found or if
present changes are mild (Fitzgerald et al., 2003; Erdlyi et al., 2007) (Table 3).
Viral antigen is usually detected in enterocytes and intestinal crypt cells,
inflammatory cells and smooth muscle cells of the lamina muscularis.
Endocrine system. Lymphoplasmacytic and histiocytic pancreatitis and acinar cell
necrosis are present in most species (Table 3). WNV antigen is detected in exocrine
cells. Adrenalitis has been described in different bird species but adrenal gland cell
necrosis has only been documented in Psittaciformes (Palmieri et al., 2011) (Table
3). Viral antigen is present in both cortical and medullary cells. Thyroid gland has
rarely been analyzed in the available studies but follicular cell necrosis has been
described in ducks (Himsworth et al., 2009) and thyroiditis in goshawks
(Wnschmann et al., 2005) (Table 3). Viral antigen in follicular cells has only been
described in owls and goshawks (Wnschmann et al., 2005; Gancz et al., 2006;
Erdlyi et al., 2007).
85

Virginia Gamino Rodrguez

Tesis doctoral

Gonads. WNV associated lesions in gonads have only been documented in owls in
which authors described oophoritis, epididimitis and necrosis of granulosa cells,
oocytes and seminiferous tubular cells (Gancz et al., 2006) (Table 3). Viral antigen
has been detected in different species in oocytes, theca and granulosa cells, stromal
cells, seminiferous tubule cells and infiltrating inflammatory cells.
Skeletal muscle. In the skeletal muscle WNV infection can produce myositis,
myofibril degeneration, necrosis and fibrosis (Table 3). IHC shows viral antigen in
myofibers but this is not consistently observed (Stockman et al., 2010).
Skin.

Lymphocytic

dermatitis

has

only

been

described

in

WNV

infected

Psittaciformes (Palmieri et al., 2011) (Table 3). Viral antigen is detected in


keratinocytes of the basal layer of the epidermis, fibrocytes and macrophages of the
dermis and subcutis, interstitial cells, and epithelial cells of the feather pulp.
Although in raptors and owls skin lesions are not described, viral antigen is usually
detected (Wnschmann et al., 2005).
Bone marrow. Cellular necrosis in American crows (Wnschmann et al., 2004a)
and hypercellularity in turkeys (Zhang et al., 2006) are the only lesions that have
been described in the bone marrow (Table 3). WNV antigen can be detected in
hematopoietic cells.
Despite all the cited differences among bird orders, families or even species, lesions
and viral antigen are described in the majority of organs in WNV infected birds
(Figure 1).

86

ANAT

FAMILY

+
+
+
+

Neuronal vacuolization

Meningeal inflammatory infiltrates

Gliosis

Perivascular cuffs

Glial nodules

Vasculitis

Hemorrhage

Inflammatory infiltrates

+
+
+

Myofibril necrosis

Myocytolisis-mineralization

Inflammatory infiltrates

Vasculitis

Hemorrhage

HEART

Inflammatory infiltrates

ND

NT

Inflammatory infiltrates

PERIPHERAL NERVOUS SYSTEM

NT

Retinal cell necrosis

EYE

Neuronal necrosis-degeneration

SPINAL CORD

Neuronal necrosis-degeneration

BRAIN

TISSUE/Lesion

ANSE

ORDER*

ND

NT

NT

ND

ND

LARI

CHAR

ND

NT

NT

ND

ND

ND

ND

ND

ND

ND

ND

ND

ND

ARDE

CICO

ACCI

NT

NT

NT

ND

ND

FALC

FALC

ND

NT

NT

ND

ND

PHAS

GALL

Table 3. Distribution of specific lesions in tissues of naturally WNV infected birds.

ND

ND

ND

ND

CORV

NT

NT

NT

NT

NT

LANI

PASS

NT

NT

NT

ND

ND

PASS

ND

NT

NT

ND

ND

PHAL

PELE

ND

NT

NT

ND

ND

PHOE

PHOE

ND

ND

ND

PSIT

PSIT

STRI

STRI

Captulo 1

87

ANAT

FAMILY

+
+

Fibrin deposition

Lymphoid cell depletion

Hemosiderosis

Hemorrhage

Bursal lymphoid cell atrophy-apoptosis

Thymic lymphoid cell necrosis

Kupffer cell necrosis

Inflammatory infiltrates

Vasculitis

Biliary duct hyperplasia

Hemosiderosis

Hemorrhage

Inflammatory infiltrates

Vasculitis
+

Glomerular cell necrosis

Hemorrhage

Tubular epithelial cell necrosis

KIDNEY

Hepatocyte necrosis

LIVER

Bursal epithelial cell atrophy-apoptosis

OTHER LYMPHOID ORGANS

Lymphoid cell necrosis/apoptosis

SPLEEN

TISSUE/Lesion

ANSE

ORDER*

ND

ND

ND

ND

ND

ND

ND

ND

ND

ND

ND

ND

NA

NA

NA

ND

ND

ND

ND

ND

LARI

CHAR

ND

ND

ND

ND

ND

ND

ND

NA

NA

NA

ARDE

CICO

NA

ACCI

NA

NA

NA

FALC

FALC

NA

NA

NA

PHAS

GALL

Table 3. Distribution of specific lesions in tissues of naturally WNV infected birds (cont).

NA

NA

NA

CORV

ND

ND

ND

ND

ND

NA

NA

NA

LANI

PASS

NA

PASS

NA

NA

NA

PHAL

PELE

NA

NA

NA

ND

ND

ND

ND

ND

PHOE

PHOE

ND

ND

PSIT

PSIT

STRI

STRI

Virginia Gamino Rodrguez


Tesis doctoral

88

ANAT

FAMILY

Inflammatory infiltrates

Vasculitis

Edema

Intestinal crypt cell necrosis

Inflammatory infiltrates

Hemorrhage

ND
ND
+

Pancreatic inflammatory infiltrates

Adrenal gland cell necrosis

Adrenal inflammatory infiltrates

Thyroid gland cell necrosis

Thyroid inflammatory infiltrates

ND

Inflammatory infiltrates

ND
ND

Myofibril degeneration-necrosis

Inflammatory infiltrates

SKELETAL MUSCLE

ND

Cellular necrosis

GONADS

Pancreatic acinar cell necrosis

ENDOCRINE SYSTEM

Enterocyte necrosis

GASTROINTESTINAL TRACT

Cellular necrosis

LUNG

TISSUE/Lesion

ANSE

ORDER*

NA

NA

ND

ND

NA

NA

ND

ND

ND

ND

ND

ND

ND

ND

ND

ND

ND

ND

LARI

CHAR

NA

NA

ND

ND

NA

NA

ND

ND

ND

ND

ND

ND

ND

ND

ND

ND

ND

ND

ARDE

CICO

ACCI

NA

NA

NA

NA

ND

ND

FALC

FALC

ND

ND

NA

NA

ND

ND

PHAS

GALL

ND

ND

NA

NA

NA

NA

CORV

Table 3. Distribution of specific lesions in tissues of naturally WNV infected birds (cont).

NA

NA

ND

ND

NA

NA

NA

NA

LANI

PASS

NA

NA

NA

NA

NA

NA

NA

NA

ND

ND

ND

ND

ND

ND

PASS

NA

NA

ND

ND

NA

NA

ND

ND

ND

ND

ND

ND

PHAL

PELE

NA

NA

ND

ND

NA

NA

ND

ND

ND

ND

ND

ND

PHOE

PHOE

ND

ND

PSIT

PSIT

ND

ND

STRI

STRI

Captulo 1

89

NA

Hypercellularity

NA

NA

NA

NA

LARI

CHAR

NA

NA

NA

NA

ARDE

CICO

ND

ND

ND

ACCI

NA

NA

NA

FALC

FALC

NA

PHAS

GALL

NA

ND

CORV

NA

NA

ND

NA

LANI

PASS

NA

NA

NA

NA

PASS

NA

NA

NA

NA

PHAL

PELE

NA

NA

NA

NA

PHOE

PHOE

ND

ND

PSIT

PSIT

*ANSE = Anseriformes; CHAR = Charadriiformes; CICO = Ciconiiformes; FALC = Falconiformes; GALL = Galliformes; PASS = Passeriformes; PELE = Pelecaniformes; PHOE =
Phoenicopteriformes; PSIT = Psittaciformes; STRI = Sitrigiformes.
ANAT = Anatidae; LARI = Laridae; ARDE = Ardeidae; ACCI = Accipitridae; FALC = Falconidae; PHAS = Phasianidae; CORV= Corvidae; LANI = Laniidae; PASS = Passeridae; PHAL =
Phalacrocoracidae; PHOE = Phoenicopteridae; PSIT = Psittacidae; STRI = Strigidae.
ND: No described lesion. Tissues that were taken during necropsy but for which there are no lesions described.
NA: Tissue sample no analyzed.
+ Lesion present. Lesion described by at least one author for the tissue.
Lesion absent. Lesion stated as absent or not specifically described by any author for the tissue.

NA

NA

Cellular necrosis

BONE MARROW

Inflammatory infiltrates

SKIN

Fibrosis

SKELETAL MUSCLE (cont)


ND

ANAT

FAMILY

TISSUE/Lesion

ANSE

ORDER*

Table 3. Distribution of specific lesions in tissues of naturally WNV infected birds (cont).

ND

ND

STRI

STRI

Virginia Gamino Rodrguez


Tesis doctoral

90

Captulo 1

DISCUSSION AND CONCLUSION


West Nile virus, a pathogen considered of little importance for birds before the
1990s (Zeller and Schuffenecker, 2004), is nowadays one of the most widely
distributed arboviruses in the world that causes thousands of bird deaths, with a
locally significant impact on populations of native species in North America (LaDeau
et al., 2007; Foppa et al., 2011).
In this work we have reviewed the pathogenesis of WNV infection in
experimentally infected birds and the main pathologic findings that have been
described in natural infections. The information that has been reviewed indicates
on one hand that in most infected birds the virus first appears in the spleen, after
which it spreads rapidly to other organs such as the kidney, lung, heart and liver,
reaching later the CNS. However, tissue distribution takes place earlier in bird
species highly susceptible to infection such as Passeriformes and later in less
susceptible species such as Falconiformes, Galliformes or Strigiformes. On the
other hand, the reviewed information indicates that most infected birds that show
clinical disease and some of those that die without previous clinical manifestations
have macroscopic and/or microscopic lesions, but that there may be differences in
their distribution and severity between species even within the same family.
Accordingly, considerable differences exist among species in expression of clinical
disease that are not always clearly related to the severity and distribution of lesions.
Thus for example, nervous clinical signs do not always correlate with pathological
findings in the brain such as neuronal necrosis.
Acute lesions such as encephalitis and hemorrhages in different tissues are
consistently found in the vast majority of the described bird families. Hemorrhagic
fever associated to WNV infection has mainly been described in humans (Nash et
al., 2001; Paddock et al., 2006) in which hemorrhages are thought to be related to
direct or indirect microvascular damage (Paddock et al., 2006). Thus similarly, the
91

Virginia Gamino Rodrguez

Tesis doctoral

vasculitis and endothelial WNV antigen found in some bird species could explain
the presence of blood extravasation. A study on human pathogenic flaviviruses has
associated sequence signatures in the envelope protein of the virus with the
primary syndrome that they produce (encephalitis or hemorrhagic disease) (Barker
et al., 2009), but envelope protein sequence is not available for most WNV strains
that infected the birds from which lesion descriptions exist. One lesion that is not
consistently found in every bird family or even in different species within families is
hemosiderosis in the spleen and liver. Avian hepatic hemosiderosis has been
observed in relation to iron overload in captive wild forest birds but is also
frequently associated to hemolytic processes in acute infectious disease (Cork et al.,
1995). Although Passeriformes develop necrosis and mild inflammation in the heart,
spleen, liver or kidney, they show only mild encephalitic lesions and neuronal
necrosis is absent (Steele et al., 2000; Wnschmann et al., 2004a; O'Brien et al.,
2010). This may be related to the higher susceptibility of this order to WNV
infection, leading to rapid viral distribution and host death that does not allow
development of encephalic lesions (Gibbs et al., 2005b). The absence of encephalitis
in

juvenile

chukar

partridges

(Alectoris

chukar)

and

Impeyan

pheasants

(Lophophorus impeyanus) (Wnschmann and Ziegler, 2006) could have the same
explanation. In contrast, neuronal degeneration, necrosis and phagocytosis are well
documented in Strigiformes and Falconiformes, potentially because the course of
the disease in these species is more prolonged. The detection of WNV antigen or
genome by IHC or real time RT-PCR in a particular tissue, even when there are no
observable macroscopic or microscopic lesions, or the apparent absence of viral
antigen immunolabeling although microscopic related lesions are present, may have
the same underlying cause (Gibbs et al., 2005b; Gancz et al., 2006). Thus, lesion
description and viral antigen detection should be considered together in order to
enable us to understand the pathogenesis of WNV infection in a particular host.
92

Captulo 1

Many pathologic differences are related to the time post-infection the animal
died which, in fact, is related to its susceptibility to infection and immune capacity
at the time the virus is inoculated, as well as the infection route (oral vs. mosquito),
the infective dose and the virulence of the infecting virus. Looking for example at
the description of Lopes et al. (2007), species specific factors are evident as in this
case the infecting virus, route and dose were probably very similar. Based upon the
evaluated information, apparently there are species in which the individuals
respond early and strongly to the infection clearing the virus rapidly, suffering thus
only mild microscopic lesions and causing only few individuals to succumb to the
disease (Busquets et al., 2012). On the contrary, other species potentially mount a
weak immune response of late onset that leads to high tissue viral loads and death
due to the severity of tissue lesions during the first general viremia (Lapointe et al.,
2009; Nemeth et al., 2011). Finally, there is a third group of species which
maintains low levels of viral replication that leads to chronic infection which can
finally produce host death or long-term sequelaes (Nemeth et al., 2006a).
However, we should consider that the literature about the pathology of
natural WNV infection in birds is limited to a number of species that may not
necessarily reflect the complete host spectrum of species affected by natural
disease. Thus, the information is unbalanced, meaning that the absence of lesions
in certain tissues in any particular bird species could be related to the limited
number of studies available. The reason why most studies about the pathology of
WNV infection have been carried out in raptors and owls may be related to the
higher visibility of dead or clinically affected animals of these species in the field.
Furthermore, these animals are frequently maintained in facilities such as zoos or
rehabilitation centers where they are observed daily and where necropsies of fresh
carcasses can be performed in case of death. Additionally, studies in corvids in the
US are probably driven by the magnitude of mortalities in these species and their
93

Virginia Gamino Rodrguez

Tesis doctoral

visibility for the public as they are relatively abundant in urban and suburban
areas. The lack of information about WNV infection pathology in other species could
be related to the lack of fresh carcasses that enable a detailed pathological
description.
Certain bird species considered resistant to the disease in the field such as
the red-legged partridge (Alectoris rufa) develop lesions and succumbed to the
disease in experimental studies (Sotelo et al., 2011). Therefore, it is important to
consider that lesion and viral antigen distribution between experimentally and
naturally

infected bird

species

is

probably

different,

and

that numerous

contributing factors in the field are generally not reproduced under experimental
conditions.
In conclusion, differences in pathology of WNV infection in different bird
species are most probably related to a combination of host and environmental
factors in addition to the intrinsic virulence and pathogenicity of the infecting
strain. The non-progressive, acute or more prolonged course of the disease
accounts for part of the differences in the distribution of lesion and viral antigen
and in the severity of lesions. Finally, although experimental infections cannot
completely reproduce field situations, they can increase our understanding of
pathologic pathways and help in the identification of key factors that influence the
outcome of an infection in a given host.

ACKNOWLEDGEMENTS
This review is part of the work supported by grant AG2008-02504GAN funded by
the Spanish Ministry for Science and Innovation. V. Gamino (323/09) is a research
fellow supported by the regional government of Castilla La Mancha (JCCM). We
acknowledge F. Ruiz-Fons for critically reviewing an early draft of the paper.

94

Captulo 1

REFERENCES
Bakonyi T, Ivanics T, Erdlyi K, Ursu K, Ferenczi E, Weissenbck H, Nowotny N:
Lineage 1 and 2 strains of encephalitic West Nile virus, central Europe.
Emerg Infect Dis 2006, 12(4):618-623.
Banet-Noach C, Simanov L, Malkinson M: Direct (non-vector) transmission of
West Nile virus in geese. Avian Pathol 2003, 32(5):489-494.
Barker WC, Mazumder R, Vasudevan S, Sagripanti J-L, Wu CH: Sequence
signatures in envelope protein may determine whether flaviviruses
produce hemorrhagic or encephalitic syndromes. Virus Genes 2009, 39(1):19.
Bertelsen MF, Olberg RA, Crawshaw GJ, Dibernardo A, Lindsay LR, Drebot M,
Barker IK: West Nile virus infection in the eastern loggerhead shrike
(Lanius
ludovicianus
migrans):
Pathology,
epidemiology,
and
immunization. J Wildl Dis 2004, 40(3):538-542.
Botha EM, Markotter W, Wolfaardt M, Paweska JT, Swanepoel R, Palacios G, Nel
LH, Venter M: Genetic determinants of virulence in pathogenic lineage 2
West Nile virus strains. Emerg Infect Dis 2008, 14(2):222-230.
Brault AC, Huang CYH, Langevin SA, Kinney RM, Bowen RA, Ramey WN, Panella
NA, Holmes EC, Powers AM, Miller BR: A single positively selected West Nile
viral mutation confers increased virogenesis in American crows. Nat Genet
2007, 39(9):1162-1166.
Brinton MA: Host factors involved in West Nile virus replication. Ann N Y Acad
Sci 2001, 951:207-219.
Brinton MA: The molecular biology of West Nile Virus: a new invader of the
western hemisphere. Annu Rev Microbiol 2002, 56:371-402.
Busquets N, Bertran K, Costa TP, Rivas R, de la Fuente JG, Villalba R, Solanes D,
Bensaid A, Maj N, Pags N: Experimental West Nile virus infection in GyrSaker hybrid falcons. Vector Borne Zoonotic Dis 2012, 12(6):482-489.
Centers
for
Disease
Control
and
Prevention
[http://www.cdc.gov/ncidod/dvbid/westnile/index.htm]

(CDCa)

Centers
for
Disease
Control
and
Prevention
[http://www.cdc.gov/ncidod/dvbid/westnile/birdspecies.htm]

(CDCb)

Chambers TJ, Diamond MS: Pathogenesis of flavivirus encephalitis. Adv Virus


Res 2003, 60:273-342.
Charrel RN, Brault AC, Gallian P, Lemasson JJ, Murgue B, Murri S, Pastorino B,
Zeller H, de Chesse R, de Micco P, de Lamballerie X: Evolutionary relationship
between Old World West Nile virus strains. Evidence for viral gene flow
between Africa, the Middle East, and Europe. Virology 2003, 315(2):381-388.
Cork SC, Alley MR, Stockdale PHG: A quantitative assesment of hemosiderosis
in wild and captive birds using image-analysis. Avian Pathol 1995, 24(2):239254.
95

Virginia Gamino Rodrguez

Tesis doctoral

D'Agostino JJ, Isaza R: Clinical signs and results of specific diagnostic testing
among captive birds housed at zoological institutions and infected with
West Nile virus. J Am Vet Med Assoc 2004, 224(10):1640-1643.
Dauphin G, Zientara S, Zeller H, Murgue B: West Nile: worldwide current
situation in animals and humans. Comp Immunol Microbiol Infect Dis 2004,
27(5):343-355.
De Filette M, Ulbert S, Diamond M, Sanders NN: Recent progress in West Nile
virus diagnosis and vaccination. Vet Res 2012, 43. doi: 10.1186/1297-971643-16.
Department
for
Environment
Food
and
Rural
Affairs
(Defra)
[http://archive.defra.gov.uk/foodfarm/farmanimal/diseases/monitoring/docu
ments/wnv-austria.pdf]
Deubel V, Fiette L, Gounon P, Drouet MT, Khun H, Huerre M, Banet C, Malkinson
M, Despres P: Variations in biological features of West Nile viruses. Ann N Y
Acad Sci 2001, 951:195-206.
Eldadah AH, Nathanso.N, Sarsitis R: Pathogenesis of West Nile Virus
encephalitis in mice and mats .I. Influence of age and species on mortality
and infection. Am J Epidemiol 1967, 86(3):765-775.
Ellis AE, Mead DG, Allison AB, Stallknecht DE, Howerth EW: Pathology and
epidemiology of natural West Nile viral infection of raptors in Georgia. J
Wildl Dis 2007, 43(2):214-223.
Erdlyi K, Ursu K, Ferenczi E, Szeredi L, Rtz F, Skre J, Bakonyi T: Clinical and
pathologic features of lineage 2 West Nile virus infections in birds of prey
in Hungary. Vector Borne Zoonotic Dis 2007, 7(2):181-188.
Ernest HB, Woods LW, Hoar BR: Pathology associated with West Nile virus
infections in the yellow-billed magpie (Pica nuttalli): a California endemic
bird. J Wildl Dis 2010, 46(2):401-408.
Ezenwa VO, Godsey MS, King RJ, Guptill SC: Avian diversity and West Nile virus:
testing associations between biodiversity and infectious disease risk. Proc
Biol Sci 2006, 273(1582):109-117.
Fang Y, Reisen WK: Previous infection with West Nile or St. Louis encephalitis
viruses provides cross protection during reinfection in house finches. Am J
Trop Med Hyg 2006, 75(3):480-485.
Figuerola J, Jimnez-Clavero MA, Lpez G, Rubio C, Soriguer R, Gmez-Tejedor C,
Tenorio A: Size matters: West Nile Virus neutralizing antibodies in resident
and migratory birds in Spain. Vet Microbiol 2008, 132(1-2):39-46.
Fitzgerald SD, Patterson JS, Kiupel M, Simmons HA, Grimes SD, Sarver CE, Fulton
RM, Steficek BA, Cooley TM, Massey JP, Sikarskie JG: Clinical and pathologic
features of West Nile virus infection in native North American owls (Family
Strigidae). Avian Dis 2003, 47(3):602-610.

96

Captulo 1

Foppa IM, Beard RH, Mendenhall IH: The impact of West Nile virus on the
abundance of selected North American birds. Vet Res 2011, 7:43. doi:
10.1186/1746-6148-7-43.
Gancz AY, Barker IK, Lindsay R, Dibernardo A, McKeever K, Hunter B: West Nile
virus outbreak in North American owls, Ontario, 2002. Emerg Infect Dis
2004, 10(12):2135-2142.
Gancz AY, Smith DA, Barker IK, Lindsay R, Hunter B: Pathology and tissue
distribution of West Nile virus in North American owls (family : Strigidae).
Avian Pathol 2006, 35(1):17-29.
Garca-Tapia D, Loiacono CM, Klelboeker SB: Replication of West Nile virus in
equine peripheral blood mononuclear cells. Vet Immunol Immunopathol 2006,
110(3-4):229-244.
Gibbs SEJ, Hoffman DM, Stark LM, Marlenee NL, Blitvich BJ, Beaty BJ,
Stallknecht DE: Persistence of antibodies to West Nile virus in naturally
infected rock pigeons (Columba livia). Clin Diagn Lab Immunol 2005a,
12(5):665-667.
Gibbs SE, Ellis AE, Mead DG, Allison AB, Moulton JK, Howerth EW, Stallknecht
DE: West Nile virus detection in the organs of naturally infected blue jays
(Cyanocitta cristata). J Wildl Dis 2005b, 41(2):354-362.
Glvits R, Ferenczi E, Ivanics E, Bakonyi T, Mat T, Zarka P, Palya V: Cooccurrence of West Nile Fever and circovirus infection in a goose flock in
Hungary. Avian Pathol 2005, 34(5):408-414.
Hahn DC, Nemeth NM, Edwards E, Bright PR, Komar N: Passive West Nile virus
antibody transfer from maternal Eastern Screech-Owls (Megascops asio) to
progeny. Avian Dis 2006, 50(3):454-455.
Hamer GL, Kitron UD, Goldberg TL, Brawn JD, Loss SR, Ruiz MO, Hayes DB,
Walker ED: Host selection by Culex pipiens mosquitoes and West Nile virus
amplification. Am J Trop Med Hyg 2009, 80(2):268-278.
Himsworth CG, Gurney KEB, Neimanis AS, Wobeser GA, Leighton FA: An outbreak
of West Nile virus infection in captive lesser scaup (Aythya affinis)
ducklings. Avian Dis 2009, 53(1):129-134.
Hfle U, Blanco JM, Crespo E, Naranjo V, Jimnez-Clavero MA, Snchez A, de la
Fuente J, Gortzar C: West Nile virus in the endangered Spanish imperial
eagle. Vet Microbiol 2008, 129(1-2):171-178.
Hublek Z, Halouzka J: West Nile fever - a reemerging mosquito-borne viral
disease in Europe. Emerg Infect Dis 1999, 5(5):643-650.
Jimnez-Clavero MA, Sotelo E, Fernndez-Pinero J, Llorente F, Manuel Blanco J,
Rodrguez-Ramos J, Prez-Ramrez E, Hfle U: West Nile virus in golden
eagles, Spain, 2007. Emerg Infect Dis 2008, 14(9):1489-1491.
Jimnez-Clavero MA: Animal viral diseases and global change: bluetongue and
West
Nile
fever
as
paradigms.
Front
Genet
2012,
3.
doi:
10.3389/fgene.2012.00105.
97

Virginia Gamino Rodrguez

Tesis doctoral

Jourdain E, Gauthier-Clerc M, Bicout DJ, Sabatier P: Bird migration routes and


risk for pathogen dispersion into western Mediterranean wetlands. Emerg
Infect Dis 2007a, 13(3):365-372.
Jourdain E, Schuffenecker I, Korimbocus J, Reynard S, Murri S, Kayser Y,
Gauthier-Clerc M, Sabatier P, Zeller HG: West Nile virus in wild resident
birds, Southern France, 2004. Vector Borne Zoonot Dis 2007b, 7(3):766-771.
Joyner PH, Kelly S, Shreve AA, Snead SE, Sleeman JM, Pettit DA: West Nile virus
in raptors from Virginia during 2003: Clinical, diagnostic, and
epidemiologic findings. J Wildl Dis 2006, 42(2):335-344.
Kilpatrick AM, Daszak P, Jones MJ, Marra PP, Kramer LD: Host heterogeneity
dominates West Nile virus transmission. Proc R Soc B-Biol Sci 2006,
273(1599):2327-2333.
Kinney RM, Huang CY, Whiteman MC, Bowen RA, Langevin SA, Miller BR, Brault
AC: Avian virulence and thermostable replication of the North American
strain of West Nile virus. J Gen Virol 2006, 87(Pt 12):3611-3622.
Komar N, Langevin S, Hinten S, Nemeth N, Edwards E, Hettler D, Davis B, Bowen
R, Bunning M: Experimental infection of North American birds with the
New York 1999 strain of West Nile virus. Emerg Infect Dis 2003, 9(3):311322.
Kramer LD, Styer LM, Ebel GD: A global perspective on the epidemiology of
West Nile virus. Annu Rev Entomol 2008, 53:61-81.
Kwan JL, Kluh S, Reisen WK: Antecedent avian immunity limits tangential
transmission of West Nile virus to humans. PLoS One 2012, 7(3):e34127.
doi: 10.1371/journal.pone.0034127.
LaDeau SL, Kilpatrick AM, Marra PP: West Nile virus emergence and large-scale
declines of North American bird populations. Nature 2007, 447(7145):710713.
Lanciotti RS, Roehrig JT, Deubel V, Smith J, Parker M, Steele K, Crise B, Volpe KE,
Crabtree MB, Scherret JH, Hall RA, MacKenzie JS, Cropp CB, Panigrahy B,
Ostlund E, Schmitt B, Malkinson M, Banet C, Weissman J, Komar N, Savage
HM, Stone W, McNamara T, Gubler DJ: Origin of the West Nile virus
responsible for an outbreak of encephalitis in the northeastern United
States. Science 1999, 286(5448):2333-2337.
Lanciotti RS, Ebel GD, Deubel V, Kerst AJ, Murri S, Meyer R, Bowen M, McKinney
N, Morrill WE, Crabtree MB, Kramer LD, Roehrig JT: Complete genome
sequences and phylogenetic analysis of West Nile virus strains isolated
from the United States, Europe, and the Middle East. Virology 2002,
298(1):96-105.
Langevin SA, Bunning M, Davis B, Komar N: Experimental infection of chickens
as candidate sentinels for West Nile virus. Emerg Infect Dis 2001, 7(4):726729.

98

Captulo 1

Langevin SA, Brault AC, Panella NA, Bowen RA, Komar N: Variation in virulence
of West Nile virus strains for house sparrows (Passer domesticus). Am J
Trop Med Hyg 2005, 72(1):99-102.
Lapointe DA, Hofmeister EK, Atkinson CT, Porter RE, Dusek RJ: Experimental
infection of Hawai'i 'Amakihi (Hemignathus virens) with West Nile virus
and competence of a co-occurring vector, Culex quinquefasciatus:
potential impacts on endemic Hawaiian avifauna. J Wildl Dis 2009,
45(2):257-271.
Lim SM, Koraka P, Osterhaus AD, Martina BE: West Nile virus: immunity and
pathogenesis. Viruses 2011, 3(6):811-828.
Lindenbach BD, Rice CM: Molecular biology of flaviviruses. Adv Virus Res 2003,
59:23-61.
Lobigs M, Mllbacher A, Regner M: CD4+ and CD8+ T-Cell Immune Responses in
West Nile Virus Infection. In: West Nile Encephalitis Virus Infection. Edited by
Diamond MS. New York: Springer Science+Business Media; 2009: 287-307.
Lopes H, Redig P, Glaser A, Armien A, Wunschmann A: Clinical findings, lesions,
and viral antigen distribution in great gray owls (Strix nebulosa) and
barred owls (Strix varia) with spontaneous West Nile virus infection. Avian
Dis 2007, 51(1):140-145.
Mackenzie JS, Williams DT: The zoonotic flaviviruses of southern, south-eastern
and eastern Asia, and Australasia: the potential for emergent viruses.
Zoonoses Public Health 2009, 56(6-7):338-356.
Malkinson M, Banet C: The role of birds in the ecology of West Nile virus in
Europe and Africa. Curr Top Microbiol Immunol 2002, 267:309-322.
Malkinson M, Banet C, Weisman Y, Pokamunski S, King R, Drouet MT, Deubel V:
Introduction of West Nile virus in the Middle East by migrating white
storks. Emerg Infect Dis 2002, 8(4):392-397.
McLean R, Ubico S: Arboviruses in Birds. In: Infectious Diseases of Wild Birds.
Edited by Thomas N, Hunter D, Atkinson C. Iowa: Blacwell Publishing; 2007:
17-62.
McMinn PC: The molecular basis of virulence of the encephalitogenic
flaviviruses. J Gen Virol 1997, 78:2711-2722.
McMullen AR, Albayrak H, May FJ, Davis CT, Beasley DW, Barrett AD: Molecular
evolution of lineage 2 West Nile virus. J Gen Virol 2013, 94(Pt 2):318-325.
Monaco F, Lelli R, Teodori L, Pinoni C, Di Gennaro A, Polci A, Calistri P, Savini G:
Re-emergence of West Nile virus in Italy. Zoonoses Public Health 2010, 57(78):476-486.
Monath TP, Cropp CB, Harrison AK: Mode of entry of a neurotropic arbovirus
into the central nervous system. Reinvestigation of an old controversy.
Lab Investig 1983, 48(4):399-410.

99

Virginia Gamino Rodrguez

Tesis doctoral

Murata R, Eshita Y, Maeda A, Maeda J, Akita S, Tanaka T, Yoshii K, Kariwa H,


Umemura T, Takashima I: Glycosylation of the West Nile Virus envelope
protein increases in vivo and in vitro viral multiplication in birds. Am J
Trop Med Hyg 2010, 82(4):696-704.
Murgue B, Zeller H, Deubel V: The ecology and epidemiology of West Nile virus
in Africa, Europe and Asia. Curr Top Microbiol Immuno 2002, 267:195-221.
Nash D, Mostashari F, Fine A, Miller J, O'Leary D, Murray K, Huang A, Rosenberg
A, Greenberg A, Sherman M, Wong S, Layton M, Campbell GL, Roehrig JT,
Gubler DJ, Shieh WJ, Zaki S, Smith P, Working WNOR: The outbreak of West
Nile virus infection in the New York City area in 1999. N Engl J Med 2001,
344(24):1807-1814.
Nemeth NM, Gould D, Bowen R, Komar N: Natural and experimental West Nile
virus infection in five raptor species. J Wildl Dis 2006a, 42(1):1-13.
Nemeth NM, Hahn DC, Gould DH, Bowen RA: Experimental West Nile virus
infection in Eastern Screech Owls (Megascops asio). Avian Dis 2006b,
50(2):252-258.
Nemeth NM, Bowen RA: Dynamics of passive immunity to West Nile virus in
domestic chickens (Gallus gallus domesticus). Am J Trop Med Hyg 2007,
76(2):310-317.
Nemeth NM, Kratz GE, Bates R, Scherpelz JA, Bowen RA, Komar N: Naturally
induced humoral immunity to West Nile virus infection in raptors.
Ecohealth 2008, 5(3):298-304.
Nemeth NM, Kratz GE, Bates R, Scherpelz JA, Bowen RA, Komar N: Clinical
evaluation and outcomes of naturally acquired West Nile virus infection in
raptors. J Zoo Wildl Med 2009a, 40(1):51-63.
Nemeth NM, Oesterle PT, Bowen RA: Humoral Immunity to West Nile virus Is
long lasting and protective in the house sparrow (Passer domesticus). Am J
Trop Med Hyg 2009b, 80(5):864-869.
Nemeth NM, Young G, Ndaluka C, Bielefeldt-Ohmann H, Komar N, Bowen R:
Persistent West Nile virus infection in the house sparrow (Passer
domesticus). Arch Virol 2009c, 154(5):783-789.
Nemeth NM, Thomsen BV, Spraker TR, Benson JM, Bosco-Lauth AM, Oesterle PT,
Bright JM, Muth JP, Campbell TW, Gidlewski TL, Bowen RA: Clinical and
pathologic responses of American crows (Corvus brachyrhynchos) and fish
crows (C ossifragus) to experimental west nile virus Infection. Vet Pathol
2011, 48(6):1061-1074.
O'Brien VA, Meteyer CU, Reisen WK, Ip HS, Brown CR: Prevalence and pathology
of West Nile virus in naturally infected house sparrows, western Nebraska,
2008. Am J Trop Med Hyg 2010, 82(5):937-944.
Ostfeld RS, Keesing F: The function of biodiversity in the ecology of vectorborne zoonotic diseases. Can J Zool 2000, 78:2061-2078.

100

Captulo 1

Paddock CD, Nicholson WL, Bhatnagar J, Goldsmith CS, Greer PW, Hayes EB,
Risko JA, Henderson C, Blackmore CG, Lanciotti RS, Campbell GL, Zaki SR:
Fatal hemorrhagic fever caused by West Nile virus in the United States.
Clinical Infectious Diseases 2006, 42(11):1527-1535.
Palmieri C, Franca M, Uzal F, Anderson M, Barr B, Woods L, Moore J, Woolcock P,
Shivaprasad HL: Pathology and immunohistochemical findings of West Nile
virus infection in psittaciformes. Vet Pathol 2011, 48(5):975-984.
Papa A, Danis K, Baka A, Bakas A, Dougas G, Lytras T, Theocharopoulos G,
Chrysagis D, Vassiliadou E, Kamaria F, Liona A, Mellou K, Saroglou G,
Panagiotopoulos T: Ongoing outbreak of West Nile virus infections in
humans in Greece, July - August 2010. Euro Surveill 2010, 15(34).
Papa A, Xanthopoulou K, Gewehr S, Mourelatos S: Detection of West Nile virus
lineage 2 in mosquitoes during a human outbreak in Greece. Clin Microbiol
Infect 2011a, 17(8):1176-1180.
Papa A, Bakonyi T, Xanthopoulou K, Vzquez A, Tenorio A, Nowotny N: Genetic
characterization of West Nile virus lineage 2, Greece, 2010. Emerg Infect Dis
2011b, 17(5):920-922.
Pauli AM, Cruz-Martnez LA, Ponder JB, Redig PT, Glaser AL, Klauss G, Schoster
JV, Wnschmann A: Ophthalmologic and oculopathologic findings in redtailed hawks and Cooper's hawks with naturally acquired West Nile virus
infection. J Am Vet Med Assoc 2007, 231(8):1240-1248.
Peterson AT, Vieglais DA, Andreasen JK: Migratory birds modeled as critical
transport agents for West Nile Virus in North America. Vector Borne Zoonot
Dis 2003, 3(1):27-37.
Phalen DN, Dahlhausen B: West Nile virus. Seminars in Avian and Exotic Pet
Medicine 2004, 13(2).
Rappole JH, Compton BW, Leimgruber P, Robertson J, King DI, Renner SC:
Modeling movement of West Nile virus in the western hemisphere. Vector
Borne Zoonot Dis 2006, 6(2):128-139.
Reisen WK, Fang Y, Lothrop HD, Martinez VM, Wilson J, Oconnor P, Carney R,
Cahoon-Young B, Shafii M, Brault AC: Overwintering of West Nile virus in
Southern California. J Med Entomol 2006, 43(2):344-355.
Reisen WK, Fang Y, Martinez V: Is nonviremic transmission of West Nile virus by
Culex mosquitoes (Diptera: Culicidae) nonviremic? J Med Entomol 2007,
44(2):299-302.
Reisen WK, Hahn DC: Comparison of immune responses of brown-headed
cowbird and related blackbirds to west Nile and other mosquito-borne
encephalitis viruses. J Wildl Dis 2007, 43(3):439-449.
Saito EK, Sileo L, Green DE, Meteyer CU, McLaughlin GS, Converse KA, Docherty
DE: Raptor mortality due to West Nile virus in the United States, 2002. J
Wildl Dis 2007, 43(2):206-213.

101

Virginia Gamino Rodrguez

Tesis doctoral

Samuel MA, Diamond MS: Pathogenesis of West Nile virus infection: a balance
between virulence, innate and adaptive immunity, and viral evasion. J Virol
2006, 80(19):9349-9360.
Samuel MA, Wang H, Siddharthan V, Morrey JD, Diamond MS: Axonal transport
mediates West Nile virus entry into the central nervous system and
induces acute flaccid paralysis. Proc Natl Acad Sci U S A 2007, 104(43).
17140-17145.
Sangster MY, Urosevic N, Mansfield JP, Mackenzie JS, Shellam GR: Mapping the
Flv locus controlling resistance to flaviviruses on mouse chromosome 5. J
Virol 1994, 68(1):448-452.
Savini G, Capelli G, Monaco F, Polci A, Russo F, Di Gennaro A, Marini V, Teodori L,
Montarsi F, Pinoni C, Pisciella M, Terregino C, Marangon S, Capua I, Lelli R:
Evidence of West Nile virus lineage 2 circulation in Northern Italy. Vet
Microbiol 2012, 158(3-4):267-273.
Savini G, Puggioni G, A DIG, G DIF, Rocchigiani AM, Polci A, Marini V, Pinoni C,
Rolesu S, Marruchella G, Lorusso A, Monaco F: West Nile virus lineage 2 in
Sardinian wild birds in 2012: a further threat to public health. Epidemiol
Infect 2013:1-4.
Semenov BF, Chunikhin SP, Karmysheva V, Iakovleva NI: Study of chronic forms
of arbovirus infections in birds. 1. Experiments with West Nile, Sindbis,
Bhandja and Sicilian mosquito fever viruses. Vestn Akad Med Nauk SSSR
1973, 28(2):79-83.
Senne DA, Pedersen JC, Hutto DL, Taylor WD, Schmitt BJ, Panigrahy B:
Pathogenicity of West Nile virus in chickens. Avian Dis 2000, 44(3):642-649.
Shirafuji H, Kanehira K, Kubo M, Shibahara T, Kamio T: Experimental West Nile
virus infection in aigamo ducks, a cross between wild ducks (Anas
platyrhynchos) and domestic ducks (Anas platyrhynchos var. domesticus).
Avian Dis 2009, 53(2):239-244.
Shirato K, Miyoshi H, Goto A, Ako Y, Ueki T, Kariwa H, Takashima I: Viral
envelope protein glycosylation is a molecular determinant of the
neuroinvasiveness of the New York strain of West Nile virus. J Gen Virol
2004, 85(Pt 12):3637-3645.
Smithburn KC, Hughes TP, Burke AW, Paul JH: A neurotropic virus isolated from
the blood of a native of Uganda. Am J Trop Med Hyg 1940, s1-20(4):471-492.
Sotelo E, Fernndez-Pinero J, Llorente F, Agero M, Hfle U, Blanco JM, JimnezClavero MA: Characterization of West Nile virus isolates from Spain: new
insights into the distinct West Nile virus eco-epidemiology in the Western
Mediterranean. Virology 2009, 395(2):289-297.
Sotelo E, Gutirrez-Guzmn AV, del Amo J, Llorente F, El-Harrak M, Prez-Ramrez
E, Manuel Blanco J, Hfle U, Jimnez-Clavero MA: Pathogenicity of two
recent Western Mediterranean West Nile virus isolates in a wild bird

102

Captulo 1

species indigenous to Southern Europe: the red-legged partridge. Vet Res


2011, 42. doi: 10.1186/1297-9716-42-11.
Steele KE, Linn MJ, Schoepp RJ, Komar N, Geisbert TW, Manduca RM, Calle PP,
Raphael BL, Clippinger TL, Larsen T, Smith J, Lanciotti RS, Panella NA,
McNamara TS: Pathology of fatal West Nile virus infections in native and
exotic birds during the 1999 outbreak in New York City, New York. Vet
Pathol 2000, 37(3):208-224.
Stockman J, Hawkins MG, Burns RE, Fang Y, Brault AC, Lowenstine LJ: West Nile
virus infection in a green-winged macaw (Ara chloropterus). Avian Dis
2010, 54(1):164-169.
Suthar MS, Diamond MS, Gale M, Jr.: West Nile virus infection and immunity.
Nat Rev Microbiol 2013, 11(2):115-128.
Swayne DE, Beck JR, Zaki S: Pathogenicity of West Nile virus for turkeys. Avian
Dis 2000, 44(4):932-937.
Swayne DE, Beck JR, Smith CS, Shieh WJ, Zaki SR: Fatal encephalitis and
myocarditis in young domestic geese (Anser anser domesticus) caused by
West Nile virus. Emerg Infect Dis 2001, 7(4):751-753.
Tag-El-Din-Hassan HT, Sasaki N, Moritoh K, Torigoe D, Maeda A, Agui T: The
chicken 2'-5' oligoadenylate synthetase A inhibits the replication of West
Nile virus. Jpn J Vet Res 2012, 60(2-3):95-103.
Valiakos G, Touloudi A, Iacovakis C, Athanasiou L, Birtsas P, Spyrou V, Billinis C:
Molecular detection and phylogenetic analysis of West Nile virus lineage 2
in sedentary wild birds (Eurasian magpie), Greece, 2010. Euro Surveill
2011, 16(18).
van der Meulen KM, Pensaert MB, Nauwynck HJ: West Nile virus in the
vertebrate world. Arch Virol 2005, 150(4):637-657.
Wang T, Town T, Alexopoulou L, Anderson JF, Fikrig E, Flavell RA: Toll-like
receptor 3 mediates West Nile virus entry into the brain causing lethal
encephalitis. Nat Med 2004, 10(12):1366-1373.
Weingartl HM, Neufeld JL, Copps J, Marszal P: Experimental west nile virus
infection in blue jays (Cyanocitta cristata) and crows (Corvus
brachyrhynchos). Vet Pathol 2004, 41(4):362-370.
Wheeler SS, Langevin SA, Brault AC, Woods L, Carroll BD, Reisen WK: Detection
of persistent West Nile virus RNA in experimentally and naturally infected
avian hosts. Am J Trop Med Hyg 2012a, 87(3):559-564.
Wheeler SS, Vineyard MP, Woods LW, Reisen WK: Dynamics of West Nile virus
persistence in House Sparrows (Passer domesticus). PLoS Negl Trop Dis
2012b, 6(10):e1860. doi: 10.1371/journal.pntd.0001860.
Wheeler SS, Vineyard MP, Barker CM, Reisen WK: Importance of recrudescent
avian infection in West Nile virus overwintering: incomplete antibody
neutralization of virus allows infrequent vector infection. J Med Entomol
2012c, 49(4):895-902.
103

Virginia Gamino Rodrguez

Tesis doctoral

Wilcox BR, Yabsley MJ, Ellis AE, Stallknecht DE, Gibbs SEJ: West Nile virus
antibody prevalence in American crows (Corvus brachyrhynchos) and fish
crows (Corvus ossifragus) in Georgia, USA. Avian Dis 2007, 51(1):125-128.
Wodak E, Richter S, Bago Z, Revilla-Fernndez S, Weissenbck H, Nowotny N,
Winter P: Detection and molecular analysis of West Nile virus infections in
birds of prey in the eastern part of Austria in 2008 and 2009. Vet Microbiol
2011, 149(3-4):358-366.
Work TH, Hurlbut HS, Taylor RM: Indigenous wild birds of the Nile Delta as
potential West Nile virus circulating reservoirs. Am J Trop Med Hyg 1955,
4(5):872-888.
Wnschmann A, Shivers J, Carroll L, Bender J: Pathological and
immunohistochemical
findings
in
American
crows
(Corvus
brachyrhynchos) naturally infected with West Nile virus. J Vet Diag Invest
2004a, 16(4):329-333.
Wnschmann A, Shivers J, Bender J, Carroll L, Fuller S, Saggese M, van Wettere A,
Redig P: Pathologic findings in red-tailed hawks (Buteo jamaicensis) and
Cooper's hawks (Accipiter cooperi) naturally infected with West Nile virus.
Avian Dis 2004b, 48(3):570-580.
Wnschmann A, Shivers J, Bender J, Carroll L, Fuller S, Saggese M, van Wettere A,
Redig P: Pathologic and immunohistochemical findings in goshawks
(Accipiter gentilis) and great horned owls (Bubo virginianus) naturally
infected with West Nile virus. Avian Dis 2005, 49(2):252-259.
Wnschmann A, Ziegler A: West Nile virus-associated mortality events in
domestic chukar partridges (Alectoris chukar) and domestic Impeyan
pheasants (Lophophorus impeyanus). Avian Dis 2006, 50(3):456-459.
Ye J, Zhu B, Fu ZF, Chen H, Cao S: Immune evasion strategies of flaviviruses.
Vaccine 2013, 31(3):461-471.
Zeller HG, Schuffenecker I: West Nile virus: An overview of its spread in europe
and the Mediterranean Basin in contrast to its spread in the Americas. Eur
J Clin Microbiol Infect Dis 2004, 23(3):147-156.
Zhang Z, Wilson F, Read R, Pace L, Zhang S: Detection and characterization of
naturally acquired West Nile virus infection in a female wild turkey. J Vet
Diag Invest 2006, 18(2):204-208.
Ziegler U, Angenvoort J, Fischer D, Fast C, Eiden M, Rodriguez AV, RevillaFernandez S, Nowotny N, de la Fuente JG, Lierz M, Groschup MH:
Pathogenesis of West Nile virus lineage 1 and 2 in experimentally infected
large falcons. Vet Microbiol 2013, 161(3-4):263-273.

104

Captulo 2
Patogenia de la infeccin experimental con
dos cepas mediterrneas del virus West Nile
en perdiz roja

Gamino V, Prez-Ramrez E, Gutirrez-Guzmn AV, Sotelo E, JimnezClavero MA, Hfle U. Pathogenesis of two Mediterranean West Nile
virus strains in experimentally infected red-legged partridges. En
preparacin.

Captulo 2

RESUMEN
La re-emergencia del virus West Nile (WNV) en Europa y la cuenca mediterrnea en
las ltimas dos dcadas se atribuye a cambios aminoacdicos que se han producido
en las cepas del virus con el consecuente incremento en su virulencia para
humanos, caballos y aves. En el presente trabajo, estudiamos las diferencias en la
patogenia de la infeccin experimental de perdices rojas (Alectoris rufa), de siete
semanas de edad, con dos cepas mediterrneas de WNV, Morocco/2003 (MO03) y
Spain/2007 (SP07). El objetivo fue determinar las causas del diferente curso de la
infeccin y la diferente mortalidad encontrados en un trabajo previo. Adems,
estudiamos la dinmica de activacin y llegada de clulas inflamatorias al cerebro y
cerebelo de las perdices infectadas. En ambas infecciones, el da 6 post inoculacin
(dpi) fue cuando la carga viral fue mayor, el antgeno viral estuvo ms ampliamente
distribuido y fue ms abundante, y las lesiones microscpicas fueron ms severas.
Los rganos ms afectados fueron el corazn, hgado y bazo. En el cerebro y
cerebelo, la microgla fue la poblacin inflamatoria ms prevalente y las clulas T
CD3+ infiltraron este tejido el 6 dpi. Comparando ambas infecciones, se observaron
relativamente pocas diferencias en la carga viral, distribucin del virus y naturaleza
y severidad de las lesiones; sin embargo, en las perdices infectadas con MO03 las
lesiones microscpicas en el cerebro y cerebelo aparecieron de forma ms aguda y
fueron ms severas, lo que dio lugar a una encefalitis ms marcada. Considerando
nuestros resultados, el mayor impacto de la infeccin por MO03 para las perdices
rojas est probablemente relacionado con marcadores especficos que hacen que
esta cepa sea ms neurovirulenta.

107

Virginia Gamino Rodrguez

Tesis doctoral

ABSTRACT
West Nile virus (WNV) re-emergence in Europe and the Mediterranean basin in the
last two decades is thought to be related to amino acid substitutions in the virus
strains with a subsequent increase in their virulence for humans, horses and birds.
In the present work, we studied differences in the pathogenesis of the experimental
infection of 7-week-old red-legged partridges (Alectoris rufa) with two different
Mediterranean WNV strains, Morocco/2003 (MO03) and Spain/2007 (SP07). The
objective was to elucidate the reasons of the different infection course and mortality
observed in a previous work. Additionally, we studied the dynamics of inflammatory
cell activation and recruitment into the brain of the infected partridges. In both
infections, day 6 post-inoculation (dpi) was the day when viral load was higher,
virus antigen was more widespread and abundant, and microscopic lesions were
more severe. The most affected organs were the heart, liver and spleen. In the brain,
the more prevalent inflammatory cell population was microglia and CD3+ T cells
infiltrated this tissue on 6 dpi. Comparing both infections, few differences were
observed in viral load, virus distribution and lesion nature and severity; however, in
MO03 infected partridges microscopic lesions in the brain were more acute and
severe, which led to a more marked encephalitis. Considering our results, the
higher impact of MO03 infection on red-legged partridges is probably related to
specific markers of this WNV strain that make it more neurovirulent.

108

Captulo 2

INTRODUCTION
West Nile virus (WNV) is an arthropod-borne flavivirus whose natural cycle involves
bird hosts and mosquito vectors, with horses and humans as accidental or deadend hosts (Kramer et al., 2008). Nowadays, it is considered one of the most widely
distributed arboviruses in the world, causing thousands of human, equine and bird
encephalitis cases and mortalities, both in the Old and New World (CDC; ECDC). In
the Mediterranean basin, WNV activity has increased in the last two decades, and it
has been associated to several outbreaks affecting mainly humans and horses
(Murgue et al., 2001; Sotelo et al., 2011a). In this area, with the exception of the
outbreak reported in migrating storks (Ciconia ciconia) and geese (Anser anser
domesticus) in Israel between 1998-1999 (Malkinson et al., 2002), there have only
been sporadic cases of avian mortality (Jourdain et al., 2007; Jimnez-Clavero et
al., 2008; Monaco et al., 2010; Savini et al., 2012, 2013). WNV re-emergence is
thought to be associated mainly to genetic factors of the virus and/or
environmental factors (Sotelo et al., 2011a). The variability of amino acid sequence
of Mediterranean WNV isolates between 1998 and 2003 was low. However, recent
isolates show higher frequency of amino acid substitutions that can lead to
phenotypic changes (Sotelo et al., 2009; 2011a), as demonstrated in field and in
several experimental studies (Beasley et al., 2005; Davis et al., 2005; Wicker et al.,
2006). In birds, genetic differences in WNV strains have been shown to modify the
degree of peripheral virus replication and its virulence (Brault et al., 2004; Langevin
et al., 2005; Totani et al., 2011).
In a recent experimental study we demonstrated that the red-legged partridge
(Alectoris rufa), an endemic Mediterranean gallinaceous bird species (Blanco-Aguiar
et al., 2003), is susceptible to the experimental infection with two Western
Mediterranean WNV strains, Morocco/2003 (MO03) and Spain/2007 (SP07) (Sotelo
et al., 2011b). However, virulence of both strains was markedly different; with 70%
109

Virginia Gamino Rodrguez

Tesis doctoral

mortality in MO03 infected partridges as compared to 30% mortality in SP07


infected birds. In that study, it was indicated that one or a combination of the 13
amino acid differences detected between the two WNV strains were probably
determining differences in their virulence. Nevertheless, in the aforementioned
study, detailed pathogenesis of the infection by each strain that led to differences in
the onset of clinical signs and mortality was not addressed.
For this reason, in the present study, we compare the dynamics of virus
appearance and distribution as well as of microscopic lesion distribution and
severity in different tissues during the course of the experimental infection of redlegged partridges with these two Mediterranean WNV strains. Additionally, and
considering the importance of the central nervous system (CNS) infection in the
pathogenesis of WNV disease, we study the dynamics of immune cell activation and
recruitment into the brain of the partridges. To our knowledge, this has not been
previously documented in WNV infected birds.

MATERIAL AND METHODS


Animals
Recently hatched red-legged partridge chicks were obtained from a commercial
breeder and raised in the experimental farm of the Instituto de Investigacin en
Recursos Cinegticos (IREC) until they were six weeks of age. Prior to the
experimental infection, they were tested by ELISA for the presence of WNV
antibodies (ID Screen West Nile Competition, IdVet, Montpellier, France) and by
real time RT-PCR for detection of WNV genome in cloacal and oral swabs, in order
to ensure they were not previously infected by the virus (Sotelo et al., 2011b).

110

Captulo 2

Viruses
In this study we used two different strains of WNV that had been isolated in the
Mediterranean basin; Morocco/2003 (MO03), isolated from a horse and cultivated
in Vero cells, and Spain/2007 (SP07), isolated from a golden eagle (Aquila
chrysaetos) and cultivated in BSR cells (clone of BHK-21 cell line). Details on the
inoculates are given in Sotelo et al. (2011b).

Experimental infection
Partridges were transported to the biosafety level 3 (BSL-3) facilities in the Centro
de Investigacin en Sanidad Animal (CISA). After five days for acclimatization, two
groups of ten 7-week-old red-legged partridges were subcutaneously inoculated in
the cervical region with 104 PFU/individual of either WNV SP07 or MO03 diluted in
up to 0.1 mL Dulbecco's Minimum Essential Medium (DMEM) (supplemented with
2 mM L-glutamine, 100 U/mL penicillin and 100 g/mL streptomycin). A control
group was sham-inoculated with an equivalent volume of DMEM and placed in a
separate cage. Inoculated partridges were observed daily for clinical signs or death
and two birds of each group were euthanized by intravenous injection of
embutramide (T61 , Intervet Schering-Plough, Madrid, Spain) on days 3 (Nos. 14), 6 (Nos. 5-8) and 14 (Nos. 9-11) post-inoculation (dpi).
The experiment was performed following biosafety animal welfare and ethical
rules applicable in the EU. Food and water were provided ad libitum throughout the
experiment.

Sample collection
Detailed necropsies were performed on euthanized individuals. Samples of brain,
heart, lung, liver, spleen, kidney, thymus, bursa of Fabricius and feather pulp were
collected into sterile polypropylene tubes filled with 1 mL of Hanks' balanced
solution (10% glycerol, 200 U/mL penicillin, 200 g/mL strepctomycin, 100 U/mL
111

Virginia Gamino Rodrguez

Tesis doctoral

polymixin B sulphate, 250 g/mL gentamicin and 50 U/mL nystatin) and stored at
-70 C until analysis. In addition, samples of brain, oral mucosa, thymus, heart,
trachea, lung, liver, spleen, kidney, small and large intestine, pancreas, cecal
tonsils, bursa of Fabricius, pectoral muscle and skin with feather follicles were fixed
in 10% neutral buffered formalin.

Virus genome detection


RNA was extracted from tissue samples after homogenization and tested by real
time RT-PCR for the presence of WNV genome as described in Sotelo et al. (2011b).

Histopathology
Formalin-fixed tissue samples were trimmed, embedded in paraffin and processed
to obtain hematoxylin and eosin stained sections. These were independently
examined by two different investigators (UH and VG) to determine the presence of
WNV associated lesions. When lesions were present, these were graded according to
their distribution (focal, multifocal or diffuse) and severity (mild, moderate or
marked).

Immunohistochemistry
Tissue sections were also mounted on Vectabond reagent (Vector Laboratories,
Inc., Burlingame, CA) pretreated slides. Immunohistochemical detection of WNV
antigen was performed using a rabbit polyclonal antibody (BioReliance, Product 81015, Rockville, MD) following the protocol described previously (Gamino et al.,
2012). We also characterized the inflammatory cell population in the cerebrum and
cerebellum. For that purpose, we used primary antibodies, reagents and protocols
detailed in table 1. In all cases endogenous peroxidase activity was inhibited with a
peroxidase blocking reagent (Dako EnVision+System-HRP (AEC), DakoCytomation,
Carpinteria, CA) (CD3, CD79, GFAP) or with 3% H202 diluted in methanol (RCA),

112

Captulo 2

rinses were performed using 0.1% Tris-buffered saline/Tween20 (TBS 0.05 M, pH


7.5), non-specific primary antibody labeling was blocked with 2% albumin from
bovine serum (BSA) (Sigma-Aldrich Chemie, Steinheim, Germany) diluted in 0.1%
TBS/Tween20, and sections were counterstained with Mayer's hematoxylin.
In the immunohistochemistry (IHC) for WNV antigen detection, tissue
sections of experimentally infected red-legged partridges, in which presence of WNV
had been confirmed by real time RT-PCR, served as positive controls. Negative
controls included several sections with substitution of the primary antibody by 2%
BSA-0.1% TBS/Tween20 and negative rabbit antibody (BioReliance, Product 81
015), as well as tissue sections of a red-legged partridge negative for WNV infection.
In the IHC for T and B cells, positive controls included sections of spleen and
bursa of Fabricius of red-legged partridges. Negative controls included substitution
of the primary antibody by 2% BSA-0.1% TBS/Tween20 and a brain section of a
red-legged partridge that tested negative for WNV presence by real time RT-PCR. A
brain section of a non-infected partridge of the same age served as reference for
RCA and GFAP.
We graded virus antigen staining according to its distribution and
abundance. Moreover, we characterized the distribution of inflammatory cells in the
brain and analyzed changes in their relative abundance during the course of
infection.

113

Virginia Gamino Rodrguez

Tesis doctoral

Table 1. Detail of reagents and protocols used in the IHC for inflammatory cell characterization
in the CNS of experimentally WNV infected red-legged partridges.
Cell
population/
marker

Antibody
reference*

Pretreatment**

Primary
antibody
dilution

Primary
antibody
incubation

Secondary
antibody

Detection
system

Microgliamacrophages

Lectin RCA-1
biotinylated

Citrate buffermicrowave heat


(22 min)

1:600

45 min RT

Goat antirabbit IgG

ABC-DAB

Astrocytes

Polyclonal rabbit
anti-GFAP

Proteinase K
(7 min RT)

1:500

4C ON

T cells

Polyclonal rabbit
anti-human CD3

1:500

4C ON

AEC+
substrate
chromogen

B cells

Monoclonal rabbit
anti-human
CD79a

Labelled
polymer-HRP
anti-rabbit

1:10

45 min RT

Citrate buffermicrowave heat


(22 min)

*Primary antibody products: RCA-1 product No. B-1085 (Vector Laboratories); GFAP product No. Z0334
(DakoCytomation, Glostrup, Denmark); CD3 product No. A0452 (DakoCytomation); CD79a product No. RM-9118
(Thermo Fisher Scientific, Runcorn, UK).
**Proteinase K (DakoCytomation); RT: room temperature (22-25 C).
Antibodies were diluted in 2% BSA-0.1% TBS/Tween20.
ON: overnight.
Goat anti-rabbit IgG (Vector Laboratories) was diluted 1:200 in 0.1% TBS/Tween20 and applied for 1 hr at RT;
Labelled polymer-HRP anti-rabbit (Dako EnVision+System-HRP (AEC), DakoCytomation) was applied according to
manufacturer's recommendation.
Avidin-biotinylated enzyme complex (ABC system, Vector Laboratories) was applied for 30 min according to
manufacturer's recommendation and 3,3-diaminobenzidine tetrahydrochloride (DAB, Vector Laboratories) was
applied for 30 sec according to manufacturer's recommendations. AEC+substrate chromogen (Dako
EnVision+System-HRP (AEC), DakoCytomation) was applied for 15 min (CD3, CD79) and 3 min (GFAP) according
to manufacturer's recommendation.

RESULTS
Macroscopic findings
Macroscopic lesions were observed in the necropsy of all euthanized animals from 3
dpi on. The most affected organs were the heart, spleen, liver and kidneys, with no
remarkable difference between SP07 and MO03 inoculated partridges. Main
macroscopic lesions were pale myocardium and pale hepatic, splenic and renal
parenchyma, which, in some cases, also showed diffuse petechiae. On 14 dpi, one
SP07 inoculated bird (No. 10) had congestion in the kidney and spleen that also
was observed in the heart and the lung of one MO03 infected partridge (No. 11).
Partridge No. 11 showed cerebral vessel injection that was also observed in one
SP07 infected bird (No. 9).

114

Captulo 2

Virus genome detection


In the WNV real time RT-PCR, all tissue samples collected in the necropsy of
inoculated animals tested positive from 3 dpi on, with lower Ct values (i.e. higher
viral loads) on 6dpi (Table 2). Ct values were similar between MO03 and SP07
infected partridges or slightly lower in some tissues of the MO03 infected group
(Table 2). On 3 dpi, the spleen followed by the kidney and heart were the organs
with highest viral load (Table 2). On 6 dpi, the highest viral load was detected in the
heart but also in the spleen and kidney (Table 2). On this dpi, the feather pulp was
markedly positive, especially in bird No. 8, and the brain showed the lowest Ct
values (Table 2). On 14 dpi, viral loads were low in most cases, and several tissues
were negative in the SP07 infected partridge (No. 10) (Table 2). Control animals
tested negative for WNV RNA throughout the experiment.
Table 2. Detection of WNV genome by real time RT-PCR (Ct values) in tissues of experimentally
infected red-legged partridges.
3 dpi
SP07

6 dpi
MO03

SP07

14 dpi
MO03

SP07

MO03

TISSUE

No.1

No. 2

No. 3

No. 4

No. 5

No. 6

No. 7

No. 8

No. 9

No. 10 No. 11

Brain

30.3

32.8

31.4

33.9

25.2

26.5

25.2

24.8

NA

No Ct

37.6

Heart

28.7

28.5

25.4

24.8

19.6

20.3

23.7

18.8

NA

38.0

33.8

Lung

NA

30.9

31.0

30.1

26.3

25.7

29.2

24.2

NA

No Ct

38.8

Liver

NA

30.2

29.6

30.9

28.7

25.7

30.8

28.2

NA

No Ct

No Ct

21.8

NA

36.1

34.0

20.9

NA

37.5

36.1

Spleen
Kidney

25.3
26.3

23.7
27.5

26.5
24.5

24.4
26.0

29.7
23.0

22.7
22.4

24.6
23.7

Thymus

27.6

29.3

31.9

23.5

30.2

23.4

33.0

NA

NA

No Ct

29.0

Bursa of Fabricius

NA*

32.0

32.8

34.2

27.9

24.8

31.5

29.8

NA

No Ct

31.3

No Ct

31.4

27.4

31.3

18.2

20.8

22.8

16.9

NA

34.6

33.7

Feather pulp

*NA: tissue sample no analyzed.


No Ct: tissue sample with Ct 40 (considered negative).

Histopathology
Histologic lesions appeared as early as 3 dpi, but were more severe and widespread
on 6 dpi (Table 3). The most affected tissues in both groups were the heart, spleen,
liver and skin with feather follicles (Figures 1-4). The brain, lung and kidney were
115

Virginia Gamino Rodrguez

Tesis doctoral

moderately affected and the oral mucosa, pancreas, cecal tonsils, thymus and
intestines showed, in most cases, mild lesions (Table 3). There were no microscopic
lesions in the trachea and pectoral muscle.
Main

microscopic

findings

were

the

presence

of

lymphoplasmacytic,

histiocytic and/or granulocytic inflammatory infiltrates, cellular necrosis and/or


degeneration, and hemosiderosis in the spleen and liver (Table 3 and figures 1-4). In
the brain, the most consistently observed lesion was endothelial cell swelling
(Figure 5), both in the cerebrum and cerebellum, but also mild neuronal necrosis
(affecting also Purkinje cells) diffuse gliosis or glial nodules, and rare perivascular
cuffing (Table 3 and figures 5 and 6). All these lesions were observed in both groups
but were slightly more severe in some tissues of MO03 infected partridges,
especially in the brain (Table 3).
On 3 dpi, the main microscopic finding was the presence of widespread
moderate to marked inflammatory infiltrates, although mild cardiac myofiber
degeneration was also observed (Table 3). In the brain, endothelial cell swelling was
the only detected lesion, mainly in MO03 infected partridges (Table 3). On 6 dpi,
microscopic lesions increased in severity, with the appearance of cellular necrosis
in many tissues, which was especially severe in the liver in both groups (Table 3
and figures 1 and 3). One MO03 infected partridge (No. 8) showed multifocal
macrophages with foamy and distended cytoplasm in the spleen (Figure 2). On this
dpi, mild to moderate neuronal necrosis and gliosis appeared in the brain of MO03
infected partridges (Table 3 and figures 5 and 6). On 14 dpi, microscopic lesions
were still widespread, with moderate to marked severity in the heart and spleen
that was milder in the liver (Table 3). On this dpi, renal tubular epithelial cell
necrosis appeared and both SP07 and M003 infected birds showed mild to
moderate lesions in the brain (Table 3).

116

Captulo 2

Incidental findings included the presence of granulomas in the liver and


pancreas of birds euthanized on 3 and 6 dpi and coccidian oocysts in the
enterocytes of the large intestine. These parasites were especially numerous in
individuals euthanized on 6 dpi and in partridges infected with MO03. Thus, one
MO03 infected partridge (No. 8) showed an associated severe necrosis of the
intestinal mucosa.
Table 3. Microscopic lesions and IHC staining gradation on different days post-inoculation (dpi)
in tissues of experimentally WNV infected red-legged partridges.
3 dpi

6 dpi

SP07

MO03

14 dpi

SP07

MO03

SP07

MO03

No. 1

No. 2

No. 3

No. 4

No. 5

No. 6

No. 7

No. 8

No. 9

No. 10

No. 11

Neuronal necrosis

Gliosis

++

Perivascular cuffing

Endothelial cell swelling

++

++

++

++

Immunohistochemical staining

Purkinje cell necrosis

++

NA*

Gliosis

++

NA

++

Perivascular cuffing

NA

Endothelial cell swelling

++

++

+++

++

++

NA

++

Immunohistochemical staining

NA

Cerebrum

Cerebellum

Heart
Myofiber necrosis-degeneration

+++

++

+++

+++

++

++

Inflammatory infiltrate

++

++

+++

+++

+++

+++

++

+++

Immunohistochemical staining

++

++

+++

BALT necrosis

Inflammatory infiltrate

+++

++

+++

+++

+++

+++

+++

Immunohistochemical staining

Lung

Liver
Hepatocyte necrosis

+++

+++

Inflammatory infiltrate

++

++

++

++

++

++

+++

++

Hemosiderosis

++

++

++

Immunohistochemical staining

Lymphoid cell necrosis

++

Lymphoid cell depletion

++

+++

+++

++

++

+++

+++

++

+++

Eosinophilic material deposits

+++

+++

++

++

++

++

Hemosiderosis

+++

+++

+++

++

Immunohistochemical staining

Spleen

Granulocytic infiltrate

117

Virginia Gamino Rodrguez

Tesis doctoral

Table 3. Microscopic lesions and IHC staining gradation on different days post-inoculation (dpi)
in tissues of experimentally WNV infected red-legged partridges (cont).
3 dpi

6 dpi

SP07
No. 1

MO03

No. 2

No. 3

14 dpi

SP07

MO03

SP07

MO03

No. 4

No. 5

No. 6

No. 7

No. 8

No. 9

No. 10

No. 11

Kidney
Tubular epithelial cell necrosis

+++

+++

+++

Inflammatory infiltrate

++

++

++

Immunohistochemical staining

++

Inflammatory infiltrate

NA

Immunohistochemical staining

NA

Inflammatory infiltrate

++

NA

++

Immunohistochemical staining

NA

Acinar cell necrosis

NA

Immunohistochemical staining

NA

Lymphoid cell necrosis

NA

NA

NA

NA

++

NA

NA

NA

NA

Immunohistochemical staining

NA

NA

NA

NA

NA

NA

NA

NA

Lymphoid cell necrosis

NA

++

NA

NA

Lymphoid cell depletion

NA

++

++

++

NA

NA

Immunohistochemical staining

NA

NA

NA

Duodenum

Large intestine

Pancreas

Cecal tonsils

Bursa of Fabricius

Skin + feather follicle


Inflammatory infiltrate skin

NA

+++

NA

NA

NA

++

Inflammatory infiltrate feather


pulp

+++

NA

+++

NA

NA

NA

NA

+++

NA

+++

Immunohistochemical staining

NA

NA

NA

Tissue lesions were graded according to their distribution and severity: , no lesion; +, focal and mild or moderate /
multifocal and mild; ++, focal and marked / multifocal and moderate / diffuse and mild; +++, multifocal and marked /
diffuse and moderate or marked.
WNV antigen immunostaining was graded according to its distribution and percentage of stained cells: , negative
staining; , focal single cells; +, focal or multifocal and < 20% cells stained; ++, multifocal or diffuse and 20-80% cells
stained; +++, multifocal or diffuse and > 80% cells stained.
*NA: tissue sample no analyzed.

118

Captulo 2

Figure 1. Heart; partridge inoculated with SP07 No. 6, 6 dpi. Diffuse and marked necrosis and
degeneration of cardiac myofibers, and infiltration of lymphoplasmacytic and histiocytic cells. HE,
x100. Inset: detail of necrotic cardiac myofibers which show degeneration, fragmentation and
accumulation of hyaline material in the cytoplasm. Infiltration of mononuclear inflammatory cells is
also observed. HE, x400.
Figure 2. Spleen; partridge inoculated with MO03 No. 8, 6 dpi. Distension of the cytoplasm of
splenic macrophages by a foamy material (black arrows) or a red-brown pigment (white arrows) that
displace the nuclei. Mild infiltration of granulocytes (arrowheads). HE, x400.
Figure 3. Liver; partridge inoculated with SP07 No. 6, 6 dpi. Multifocal to coalescing marked
necrosis of hepatocytes near the central vein. HE, x40. Inset: detail of the necrotic hepatocytes which

119

Virginia Gamino Rodrguez

Tesis doctoral

show picnosis and lysis of the nuclei and degeneration of the cytoplasm. Mild infiltration of
lymphoplasmacytic and histiocytic cells is also observed. HE, x400.
Figure 4. Feather follicle; partridge inoculated with MO03 No. 11, 14 dpi. Diffuse and moderate
infiltration of lymphoplasmacytic, histiocytic and granulocytic cells in the feather pulp. HE, x200.
Inset: detail of an inflammatory nodule composed by lymphocytes, histiocytes and granulocytes. HE,
x400.
Figure 5. Cerebrum; partridge inoculated with MO03 No. 7, 6 dpi. Focal necrosis of neurons,
degeneration of the neuropil and infiltration of lymphocytic and glial cells. HE, x400. Inset: swelling of
the nuclei of endothelial cells (arrowheads). HE, x400.
Figure 6. Cerebellum; partridge inoculated with MO03 No. 7, 6 dpi. Focal necrosis of Purkinje cells
(arrow) and glial cells (arrowheads), and a focus of gliosis and degeneration in the molecular layer. HE,
x400.

Immunohistochemistry
Virus antigen
Virus antigen was detected by IHC in several tissues from 3 dpi on, mainly in
inflammatory cells but also in different parenchymal and epithelial cells.
Immunopositivity was mild to moderate in most cases and WNV antigen was more
widespread on 6 dpi and in the tissues of MO03 infected partridges (Table 3).
On 3 dpi, virus antigen was detected in macrophages in the spleen (Figure 7)
and in inflammatory cells and myofibers of the heart. In MO03 infected partridges,
there was also mild immunostaining in tubular epithelial cells of the kidney, single
acinar cells of the pancreas and a small group of inflammatory cells in the cecal
tonsils. On 6 dpi, WNV antigen was present in inflammatory cells in the lung, heart,
spleen, kidney and pancreas. It was also detected in cardiac myofibers (Figure 8),
glomerular mesangial cells (Figure 9) and tubular epithelial cells of the kidney,
acinar cells of the pancreas and in enterocytes of the crypts and myofibers of the
lamina muscularis of the large intestine. In MO03 infected birds, virus antigen was
also present in some vascular walls in the spleen, hepatocytes and Kupffer cells of
the liver, and in one vascular wall and myofibers of the lamina muscularis of the
duodenum. On 14 dpi, IHC for WNV antigen detection was negative in every tissue.

120

Captulo 2

Inflammatory cells in the brain


Microglia activation and macrophage infiltration
In both groups there was an early reaction of microglial cells, but differences in
their relative abundance and morphology were detected during the course of the
infection and between SP07 and MO03 infected birds.
In the cerebrum, microglia activation and macrophage infiltration were more
prominent in the peripheral pallium and especially evident near the lateral
ventricle. On 3 dpi, few cells with no more than two thin cellular processes were
observed, but there was also mild presence of amoeboid cells, characterized by a
larger soma and short thick cellular processes, and rounded cells, corresponding
both to activated microglia and macrophages. In one MO03 infected bird (No. 3), few
neurons and vessels were surrounded by amoeboid cells. On 6 dpi, relative
abundance of stained microglia, mainly with an amoeboid morphology, and
macrophages increased both in SP07 and MO03 inoculated birds. Many of these
cells were grouped surrounding neurons and vessels, especially in MO03 infected
birds. On this dpi, SP07 infected partridges also showed numerous ramified cells
composed by more than three thin cellular processes. On 14 dpi, ramified cells were
scarce but amoeboid cells and macrophages still remained. Reactive microglial cells
were slightly more abundant in SP07 infected birds, especially in case No. 10.
In the cerebellum, amoeboid microglial cells and macrophages were
especially abundant in the molecular layer, mainly from 6 dpi on (Figure 10). These
cells were diffusely distributed or forming nodules that in many cases were
surrounding Purkinje cells. On 6 dpi, microglia activation was also moderate in the
granular layer and white matter, especially in MO03 infected partridges. Similarly
to the cerebrum, cellular activation was lower on 14 dpi.

121

Virginia Gamino Rodrguez

Tesis doctoral

Astrocyte activation
GFAP+ cells were detected from 3 dpi on, with only mild changes in their
distribution and relative abundance during the infection course and between
infected groups. In comparison to a non-infected partridge, there were mild
differences in staining distribution but none in astrocyte morphology or staining
intensity.
In the cerebrum, all inoculated animals showed positive staining in
astrocytes with long cellular processes located in the subpial zone, in astrocytes
with medium cellular processes surrounding some vessel walls, and in star-shaped
astrocytes in the pallium near the lateral ventricle (hippocampus) (Figure 11). Mild
astrocytosis (i.e. increased number of astrocytes) was detected in the lamina
medularis dorsalis in MO03 infected birds.
In the cerebellum, there was mild astrocytosis in the granular layer, slightly
more marked on 6 dpi and in MO03 infected birds. In the MO03 infected group,
especially in one bird euthanized on 6 dpi (No. 7), some GFAP+ fibers invade the
molecular layer among Purkinje cells. In this group, there was also a moderate
astrocytosis in the white matter.
T cell infiltration
Although scarce CD3+ T cells were detected in the vascular lumen of some vessels
in MO03 infected partridges on 3 dpi, it was on 6 dpi when these cells were more
numerous, especially in MO03 infected birds.
In the cerebrum, T cell infiltration was more marked in the peripheral
pallium. These cells were mainly diffusely distributed, but also formed part of
perivascular infiltrates and of inflammatory foci/nodules associated, in some cases,
with neuronal necrosis. T cells were also detected within the vascular lumen. On 6
dpi, partridge No. 8 (MO03) showed many T cells forming part of large inflammatory

122

Captulo 2

nodules. On 14 dpi, cellular infiltration decreased, more drastically so in the MO03


infected group.
In the cerebellum, most T cells were distributed in the molecular layer, in
some cases forming part of inflammatory nodules surrounding Purkinje cells
(Figure 12). There was also moderate infiltration in the granular layer and mild
infiltration in the white matter. On 6 dpi, this infiltration was much more
prominent in MO03 infected birds, especially in case No. 7, decreasing markedly in
both groups on 14 dpi.
Few T cells were detected in meningeal vessels, only in MO03 infected
partridges. In some cases, brain zones of T cell infiltration corresponded to brain
zones of microglia activation and/or macrophage infiltration.
B cell infiltration
B cells were not stained in the brain of the red-legged partridges on any dpi.

123

Virginia Gamino Rodrguez

Tesis doctoral

Figure 7. Spleen; partridge inoculated with SP07 No. 1, 3 dpi. WNV antigen in the cytoplasm of
splenic macrophages. IHC for WNV with Mayer's hematoxylin counterstain, x400.
Figure 8. Heart; partridge inoculated with SP07 No. 6, 6 dpi. WNV antigen in the cytoplasm of
cardiac myofibers. IHC for WNV with Mayer's hematoxylin counterstain, x100. Inset: detail of WNV
antigen in the cytoplasm of cardiac myofibers. IHC for WNV with Mayer's hematoxylin counterstain,
x400.
Figure 9. Kidney; partridge inoculated with MO03 No. 8, 6 dpi. WNV antigen in the cytoplasm of
glomerular mesangial cells. IHC for WNV with Mayer's hematoxylin counterstain, x400.
Figure 10. Cerebellum; partridge inoculated with MO03 No. 8, 6 dpi. RCA-1 positive staining in
the cytoplasm of phagocytic foamy macrophages in the molecular layer. IHC for microglia
activation/macrophages with Mayer's hematoxylin counterstain, x400.

124

Captulo 2

Figure 11. Cerebrum; partridge inoculated with MO03 No. 8, 6 dpi. GFAP positive staining in
astrocytes diffusely located in the subpial zone, surrounding blood vessels and diffusely distributed
within the parenchyma near the lateral ventricle (LV). IHC for astrocytes with Mayer's hematoxylin
counterstain, x100. Inset: detail of stained astrocytes with medium cellular processes surrounding a
blood vessel and star-shaped astrocytes within the parenchyma. IHC for astrocyte activation with
Mayer's hematoxylin counterstain, x400.
Figure 12. Cerebellum; partridge inoculated with MO03 No. 7, 6 dpi. CD3 positive staining in T
cells located near the Purkinje cell layer. IHC for T cells with Mayer's hematoxylin counterstain, x400.

DISCUSSION
In this work, we studied differences in the pathogenesis of the experimental
infection of red-legged partridges with two different Mediterranean WNV strains, in
order to elucidate the different infection course and mortality observed in a previous
work (Sotelo et al., 2011b).
In both groups, WNV genome and antigen as well as macroscopic and
microscopic lesions were detected as early as 3 dpi. On this dpi, the most affected
tissues were the heart, liver, spleen and kidney and virus antigen was detected
mainly in inflammatory cells, suggesting that these cells play an important role in
WNV infection pathogenesis in red-legged partridges. However, day 6 pi should be
considered critical in the course of the infection, since at that time viral load in
tissues was highest, virus antigen was more widespread and abundant, and
microscopic lesions were more severe. On this dpi, most birds suffered encephalitis
and virus antigen was detected both in inflammatory cells and parenchymal and
epithelial cells of several tissues. On 14 dpi, tissue lesions were still observed,
however, viral loads were low or absent and detectable virus had been cleared from
many tissues, suggesting that, at that time, partridges had eliminated most
infecting virus. Nevertheless, potential virus maintenance in the tissues where viral
load was relatively low should be considered, as persistent infection has been
demonstrated in experimentally and naturally WNV infected birds (Reisen et al.,
2006; Wheeler et al., 2012).

125

Virginia Gamino Rodrguez

Tesis doctoral

Microscopic lesions found in this work were similar to those described in


other WNV infected gallinaceous birds (Steele et al., 2000; Wnschmann and
Ziegler, 2006; Zhang et al., 2006) but, while we found endothelial cell swelling,
gliosis and neuronal necrosis in the brain, CNS lesions were not described in
chukar partridges (Alectoris chukar) naturally infected with the virus (Wnschmann
and Ziegler, 2006). The presence of marked inflammatory infiltrates in the feather
pulp and skin from 3 dpi onwards was probably associated to the high viral load
found, but we were not able to demonstrate virus antigen by IHC. Nevertheless,
WNV has been detected in the skin and feather follicles in naturally and
experimentally infected birds (Wnschmann et al., 2005; Gancz et al., 2006;
Shirafuji et al., 2008), and feather-picking has been suggested as a potential way of
horizontal WNV transmission (Banet-Noach et al., 2003). The severe increase in the
number of intestinal coccidian oocysts from 3 to 6 dpi, only in inoculated animals,
suggests that immunosuppression associated to WNV infection exacerbated this
secondary parasitation. In fact, secondary pathological processes in WNV infected
birds have been documented on numerous occasions (Saito et al., 2007; Hfle et al.,
2008; Nemeth et al., 2009).
In general, viral loads obtained in the real time RT-PCR correlated well with
virus antigen detected by IHC and microscopic lesions, with some exceptions such
as the brain. Despite the fact that this tissue showed microscopic lesion and tested
positive for WNV genome, virus antigen was not detected by IHC. Different
sensitivity of PCR and IHC could be the reason (Steele et al., 2000). However, it
would be possible that the mild to moderate neuronal necrosis found in our study
was more related to the local inflammatory response than to viral infection and
replication within neurons and other parenchymal cells, especially on 14 dpi.
Once WNV reaches the CNS, it rapidly infects neurons, but also glial and
endothelial cells (Steele et al., 2000; Fitzgerald et al., 2003; Erdlyi et al., 2007;

126

Captulo 2

Lopes et al., 2007). This activates the CNS immune response that, at least in
mammals, includes microglia and astrocyte activation and proliferation, chemokine
and cytokine release, and leukocyte recruitment (Samuel and Diamond, 2006;
Brhin et al., 2008; Suthar et al., 2013). In our study, the most prevalent
inflammatory cell population was microglia, mainly on 6 dpi, when these cells
change their morphology to an activated amoeboid form. Microglial cells are one of
the resident immune effectors of the CNS (Kettenmann et al., 2011) and are
considered essential in the anti-flavivirus response in the CNS (Maximova et al.,
2009). However, in case viral infection cannot be controlled, prolonged activation of
these cells can have detrimental effects, contributing to neuron damage (Brhin et
al., 2008). In fact, in our study, mild to moderate neuronal necrosis was detected on
6 and 14 dpi, days on which the reaction of these immune effectors was more
intense. We also detected RCA-1 lectin+ round shaped cells that, in many cases,
corresponded to macrophages, which were especially numerous on 6 and 14 dpi.
The other resident immune effector of the CNS, astrocytes, played a limited role in
the encephalitis of the red-legged partridges, as we only found mild differences in
their distribution or abundance during the course of the infection and compared to
a non-infected partridge of the same age. The presence of GFAP+ fibers among
Purkinje cells in the molecular layer of the cerebellum, mainly in one MO03 infected
partridge, potentially corresponded to Bergmann glia (Kalman et al., 1993).
Astrocytes constitute 50% of the glial cell population in the brain and are a
structural support of the CNS (Montgomery, 1994). In case of CNS damage, their
role as phagocyte and antigen-presenting cell is less important than that of the
microglia (Montgomery, 1994). Astrocytes also participate in WNV-associated
encephalitis in humans (Kelley et al., 2003) and are able to reduce WNV-induced
neuropathology in mice (Hussmann et al., 2013). Some authors have indicated that
these cells can react later than the microglia and that their maximum activity can

127

Virginia Gamino Rodrguez

Tesis doctoral

occur 14 days after CNS injury (Montgomery, 1994). Therefore, it is possible that
astrocytosis and/or astrogliosis would have been detected later if our experimental
study had carried on. In response to activation of microglia and astrocytes, among
other stimuli, CD3+ T cell infiltrated the brain, especially on 6 dpi and mainly the
cerebral pallium and molecular layer of the cerebellum. T cells act destroying virusinfected cells and producing cytokines that increase immune cell recruitment and
stimulate other immune effectors (Dorries, 2001). In WNV infection, these cells are
essential for virus clearance and for recovery from the disease (Shrestha and
Diamond, 2004; Sitati and Diamond, 2006), both in humans and rodent models
(Sampson and Armbrustmacher, 2001; Brhin et al., 2008). However, it has been
indicated that a robust response of these inflammatory cells can also have
detrimental effects (Wang et al., 2003). The failure to detect B cells in the CNS of
the partridges may be related to primary antibody malfunction rather than to a true
absence of B cell infiltration. Nevertheless, B cell infiltration in WNV-induced
encephalitis is much lower than that of T cells, at least in rodents and humans
(Kelley et al., 2003; Wang et al., 2003). These cells are usually detected in
perivascular inflammatory infiltrates, so the low number of positive cells detected in
this compartment in our study could be related to the absence of B cell staining.
Despite the fact that similar infection dynamics was found in experimentally
WNV SP07 and MO03 infected partridges, some differences were noticeable in viral
loads, virus antigen distribution and microscopic lesions severity. However, the
most striking differences were found in the CNS, where microscopic lesions and
encephalitis were more acute and severe in MO03 infected partridges.
WNV infection pathogenesis depends mainly on viral and host factors, which
determine the level of viral replication and, therefore, the severity of the infection. In
our work, the experimentally infected partridges were maintained under similar
conditions and were challenged at the same age, so immunologic status should be

128

Captulo 2

very similar between groups. For this reason, we think that the aforementioned
differences were most probably related to factors related to the WNV strain. Genetic
changes that lead to amino acidic substitutions can modify virulence of WNV
strains, in fact substitution of threonine by proline in position 249 of the NS3
protein has been identified as a WNV virulence marker in American crows (Corvus
brachyrhynchos) (Brault et al., 2007). Although this amino acid substitution is
present in the SP07 strain, in our experimental study this did not lead to an
increased virulence, as had already been indicated (Sotelo et al., 2011b).
Considering that SP07 and MO03 differ in 13 amino acid positions (Sotelo et al.,
2009a), it is probable that other markers are determining the virulence of these two
Mediterranean WNV strains.
According to our results, in red-legged partridges, the different virulence of
the MO03 and SP07 WNV strains appears to be driven by differences in the
pathogenesis of the CNS infection, as differences in pathological findings and virus
replication in other tissues were not enough to explain the higher mortality induced
by the Moroccan strain. Pathogenesis of WNV infection in the CNS depends mainly
on the capacity of the virus strain to enter the CNS (neuroinvasiveness) and
produce lesions (neurovirulence) (Chambers and Diamond, 2003). Some authors
have indicated that, once a certain viremia level is reached, the ability of a WNV
strain to enter the CNS is much more important than its own neurovirulence
(Brault et al., 2004; Beasley et al., 2005). Although mean peak viremia titer was
higher in the group inoculated with MO03, there were no statistically significant
differences (Sotelo et al., 2011b), and, in both groups, WNV was present in the
brain as early as 3 dpi. Moreover, on 6 dpi, despite viral load being very similar
between infected groups, a more intense inflammatory reaction and more severe
microscopic lesions were observed in MO03 infected partridges. For these reasons,
we suspect that, in this study, differences in neurovirulence between the strains

129

Virginia Gamino Rodrguez

Tesis doctoral

were probably more important than their neuroinvasiveness and it is probable that
a combination of the higher neurovirulence of MO03 with the more exacerbated
inflammatory reaction resulted in a more acute and severe CNS pathology.
Nevertheless, the limited number of birds analyzed in the study forces us to be
cautious with the interpretation of the results obtained.
In conclusion, experimental WNV infection of red-legged partridges with two
different Western Mediterranean strains resulted in a similar pathogenesis with
slightly higher viral loads, higher distribution of virus antigen and a higher severity
of lesions in MO03 infected individuals. The more acute and severe lesions in the
brain of MO03 infected partridges in combination with an apparent similar viral
load imply that the higher impact of MO03 infection on this species may be related
to a higher neurovirulence of this strain. Further studies should enable us to
elucidate the specific markers that are determining this higher neurovirulence.

ACKNOWLEDGEMENTS
We thank the personnel of the experimental farm of the University of Castilla-La
Mancha La Galiana for their effort in this study. We would like to acknowledge the
Junta de Comunidades de Castilla-La Mancha (JCCM) for their support. This study
is a contribution of the epidemiological network of rehabilitation centers in the
community of Castilla-La Mancha, and the REVISA network, and has been
supported by the project PAC08-0296-7771 (JCCM), and from INIA-MARM funds
(INIA CC08-020). V. Gamino (323/09) is a research fellow supported by the regional
government of Castilla La Mancha (JCCM) and Elisa Perez Ramirez is a fellow
from the National Research Council (CSIC).

130

Captulo 2

REFERENCES
Banet-Noach C, Simanov L, Malkinson M. Direct (non-vector) transmission of
West Nile virus in geese. Avian Pathol 2003, 32(5):489-494.
Beasley DW, Whiteman MC, Zhang S, Huang CY, Schneider BS, Smith DR,
Gromowski GD, Higgs S, Kinney RM, Barrett AD. Envelope protein
glycosylation status influences mouse neuroinvasion phenotype of genetic
lineage 1 West Nile virus strains. J Virol 2005, 79(13):8339-8347.
Blanco-Aguiar JA, Virgs E, Villafuerte R: Perdiz roja (Alectoris rufa). In: Atlas de
las aves reproductoras de Espaa. Edited by Mart R, del Moral JC. Madrid:
DGCoNa-SEO; 2003:212-213.
Brault AC, Langevin SA, Bowen RA, Panella NA, Biggerstaff BJ, Miller BR, Komar N.
Differential virulence of West Nile strains for American crows. Emerg Infect
Dis 2004, 10(12):2161-2168.
Brault AC, Huang CYH, Langevin SA, Kinney RM, Bowen RA, Ramey WN, Panella
NA, Holmes EC, Powers AM, Miller BR. A single positively selected West Nile
viral mutation confers increased virogenesis in American crows. Nature
Genet 2007, 39(9):1162-1166.
Brhin AC, Mouries J, Frenkiel MP, Dadaglio G, Despres P, Lafon M, Couderc T.
Dynamics of immune cell recruitment during West Nile encephalitis and
identification of a new CD19+B220-BST-2+ leukocyte population. J
Immunol 2008, 180(10):6760-6767.
Centers
for
Disease
Control
and
Prevention
[http://www.cdc.gov/ncidod/dvbid/westnile/index.htm]

(CDC)

Chambers TJ, Diamond MS. Pathogenesis of flavivirus encephalitis. Adv Virus


Res 2003, 60:273-342.
Davis CT, Ebel GD, Lanciotti RS, Brault AC, Guzman H, Siirin M, Lambert A,
Parsons RE, Beasley DW, Novak RJ, Elizondo-Quiroga D, Green EN, Young DS,
Stark LM, Drebot MA, Artsob H, Tesh RB, Kramer LD, Barrett AD.
Phylogenetic analysis of North American West Nile virus isolates, 20012004: evidence for the emergence of a dominant genotype. Virology 2005,
342(2):252-265.
Dorries R. The role of T-cell-mediated mechanisms in virus infections of the
nervous system. Curr Top Microbiol Immunol 2001, 253:219-245.
European
Centre
for
Disease
Prevention
and
Control
(ECDC)
[http://www.ecdc.europa.eu/en/healthtopics/west_nile_fever/West-Nile-fevermaps/Pages/index.aspx]
Erdlyi K, Ursu K, Ferenczi E, Szeredi L, Rtz F, Skre J, Bakonyi T. Clinical and
pathologic features of lineage 2 West Nile virus infections in birds of prey
in Hungary. Vector Borne Zoonotic Dis 2007, 7(2):181-188.
Fitzgerald SD, Patterson JS, Kiupel M, Simmons HA, Grimes SD, Sarver CE, Fulton
RM, Steficek BA, Cooley TM, Massey JP, Sikarskie JG. Clinical and pathologic
131

Virginia Gamino Rodrguez

Tesis doctoral

features of West Nile virus infection in native North American owls (Family
strigidae). Avian Dis 2003, 47(3):602-610.
Gamino V, Gutirrez-Guzmn AV, Fernndez-de-Mera IG, Ortz JA, Durn-Martn
M, de la Fuente J, Gortzar C, Hfle U. Natural Bagaza virus infection in
game birds in southern Spain. Vet Res 2012, 43(1):65. doi: 10.1186/12979716-43-65.
Gancz AY, Smith DA, Barker IK, Lindsay R, Hunter B. Pathology and tissue
distribution of West Nile virus in North American owls (family: Strigidae).
Avian Pathol 2006, 35(1):17-29.
Hfle U, Blanco JM, Crespo E, Naranjo V, Jimnez-Clavero MA, Snchez A, la
Fuente J, Gortzar C. West Nile virus in the endangered Spanish imperial
eagle. Vet Microbiol 2008, 129(1-2):171-178.
Hussmann KL, Samuel MA, Kim KS, Diamond MS, Fredericksen BL. Differential
replication of pathogenic and nonpathogenic strains of West Nile virus
within astrocytes. J Virol 2013, 87(5):2814-2822.
Jimnez-Clavero MA, Sotelo E, Fernndez-Pinero J, Llorente F, Manuel Blanco J,
Rodriguez-Ramos J, Prez-Ramrez E, Hfle U. West Nile virus in golden
eagles, Spain, 2007. Emerg Infect Dis 2008, 14(9):1489-1491.
Jourdain E, Schuffenecker I, Korimbocus J, Reynard S, Murri S, Kayser Y,
Gauthier-Clerc M, Sabatier P, Zeller HG. West Nile virus in wild resident
birds, Southern France, 2004. Vector Borne Zoonotic Dis 2007, 7(3):766-771.
Kalman M, Szekely AD, Csillag A. Distribution of glial fibrillary acidic proteinimmunopositive structures in the brain of the domestic chicken (Gallus
domesticus). J Comp Neurol 1993, 330(2):221-237.
Kelley TW, Prayson RA, Ruiz AI, Isada CM, Gordon SM. The neuropathology of
West Nile virus meningoencephalitis. A report of two cases and review of
the literature. Am J Clin Pathol 2003, 119(5):749-753.
Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. Physiology of microglia.
Physiol Rev 2011, 91(2):461-553.
Kramer LD, Styer LM, Ebel GD. A global perspective on the epidemiology of
West Nile virus. Annu Rev Entomol 2008, 53:61-81.
Langevin SA, Brault AC, Panella NA, Bowen RA, Komar N. Variation in virulence
of West Nile virus strains for house sparrows (Passer domesticus). Am J
Trop Med Hyg 2005, 72(1):99-102.
Lopes H, Redig P, Glaser A, Armien A, Wnschmann A. Clinical findings, lesions,
and viral antigen distribution in great gray owls (Strix nebulosa) and
barred owls (Strix varia) with spontaneous West Nile virus infection. Avian
Dis 2007, 51(1):140-145.
Malkinson M, Banet C, Weisman Y, Pokamunski S, King R, Drouet MT, Deubel V.
Introduction of West Nile virus in the Middle East by migrating white
storks. Emerg Infect Dis 2002, 8(4):392-397.
132

Captulo 2

Maximova OA, Faucette LJ, Ward JM, Murphy BR, Pletnev AG. Cellular
inflammatory response to flaviviruses in the central nervous system of a
primate host. J Histochem Cytochem 2009, 57(10):973-989.
Monaco F, Lelli R, Teodori L, Pinoni C, Di Gennaro A, Polci A, Calistri P, Savini G.
Re-emergence of West Nile virus in Italy. Zoonoses Public Health 2010, 57(78):476-486.
Montgomery DL. Astrocytes: form, functions, and roles in disease. Vet Pathol
1994, 31(2):145-167.
Murgue B, Murri S, Triki H, Deubel V, Zeller HG. West Nile in the Mediterranean
basin: 1950-2000. Ann N Y Acad Sci 2001, 951:117-26.
Nemeth NM, Kratz GE, Bates R, Scherpelz JA, Bowen RA, Komar N. Clinical
evaluation and outcomes of naturally acquired West Nile virus infection in
raptors. J Zoo Wildl Med 2009, 40(1):51-63.
Reisen WK, Fang Y, Lothrop HD, Martinez VM, Wilson J, Oconnor P, Carney R,
Cahoon-Young B, Shafii M, Brault AC. Overwintering of West Nile virus in
Southern California. J Med Entomol 2006, 43(2):344-355.
Saito EK, Sileo L, Green DE, Meteyer CU, McLaughlin GS, Converse KA, Docherty
DE. Raptor mortality due to West Nile virus in the United States, 2002. J
Wildl Dis 2007, 43(2):206-213.
Sampson BA, Armbrustmacher V. West Nile encephalitis: the neuropathology of
four fatalities. Ann N Y Acad Sci 2001, 951:172-178.
Samuel MA, Diamond MS. Pathogenesis of West Nile virus infection: a balance
between virulence, innate and adaptive immunity, and viral evasion. J Virol
2006, 80(19):9349-9360.
Savini G, Capelli G, Monaco F, Polci A, Russo F, Di Gennaro A, Marini V, Teodori L,
Montarsi F, Pinoni C, Pisciella M, Terregino C, Marangon S, Capua I, Lelli R.
Evidence of West Nile virus lineage 2 circulation in Northern Italy. Vet
Microbiol 2012, 158(3-4):267-273.
Savini G, Puggioni G, A DIG, G DIF, Rocchigiani AM, Polci A, Marini V, Pinoni C,
Rolesu S, Marruchella G, Lorusso A, Monaco F. West Nile virus lineage 2 in
Sardinian wild birds in 2012: a further threat to public health. Epidemiol
Infect 2013:1-4.
Shirafuji H, Kanehira K, Kubo M, Shibahara T, Kamio T. Experimental West Nile
virus infection in jungle crows (Corvus macrorhynchos). Am J Trop Med Hyg
2008, 78(5):838-842.
Shrestha B, Diamond MS. Role of CD8+ T cells in control of West Nile virus
infection. J Virol 2004, 78(15):8312-8321.
Sitati EM, Diamond MS. CD4+ T-cell responses are required for clearance of
West Nile virus from the central nervous system. J Virol 2006, 80(24):1206012069.

133

Virginia Gamino Rodrguez

Tesis doctoral

Sotelo E, Fernndez-Pinero J, Llorente F, Agero M, Hoefle U, Blanco JM, JimnezClavero MA. Characterization of West Nile virus isolates from Spain: new
insights into the distinct West Nile virus eco-epidemiology in the Western
Mediterranean. Virology 2009, 395(2):289-297.
Sotelo E, Fernndez-Pinero J, Llorente F, Vzquez A, Moreno A, Agero M, Cordioli
P, Tenorio A, Jimnez-Clavero MA. Phylogenetic relationships of Western
Mediterranean West Nile virus strains (1996-2010) using full-length
genome sequences: single or multiple introductions? J Gen Virol 2011a,
92(Pt 11):2512-2522.
Sotelo E, Gutirrez-Guzmn AV, del Amo J, Llorente F, El-Harrak M, Prez-Ramrez
E, Blanco JM, Hoefle U, Jimnez-Clavero MA. Pathogenicity of two recent
Western Mediterranean West Nile virus isolates in a wild bird species
indigenous to Southern Europe: the red-legged partridge. Vet Res 2011b,
42. doi: 10.1186/1297-9716-42-11.
Steele KE, Linn MJ, Schoepp RJ, Komar N, Geisbert TW, Manduca RM, Calle PP,
Raphael BL, Clippinger TL, Larsen T, Smith J, Lanciotti RS, Panella NA,
McNamara TS. Pathology of fatal West Nile virus infections in native and
exotic birds during the 1999 outbreak in New York City, New York. Vet
Pathol 2000, 37(3):208-224.
Suthar MS, Diamond MS, Gale M, Jr. West Nile virus infection and immunity.
Nat Rev Microbiol 2013, 11(2):115-128.
Totani M, Yoshii K, Kariwa H, Takashima I. Glycosylation of the envelope protein
of West Nile Virus affects its replication in chicks. Avian Dis 2011,
55(4):561-568.
Wang Y, Lobigs M, Lee E, Mullbacher A. CD8+ T cells mediate recovery and
immunopathology in West Nile virus encephalitis. J Virol 2003,
77(24):13323-13334.
Wheeler SS, Langevin SA, Brault AC, Woods L, Carroll BD, Reisen WK. Detection
of persistent West Nile virus RNA in experimentally and naturally infected
avian hosts. A J Trop Med Hyg 2012, 87(3):559-564.
Wicker JA, Whiteman MC, Beasley DW, Davis CT, Zhang S, Schneider BS, Higgs S,
Kinney RM, Barrett AD. A single amino acid substitution in the central
portion of the West Nile virus NS4B protein confers a highly attenuated
phenotype in mice. Virology 2006, 349(2):245-253.
Wnschmann A, Shivers J, Bender J, Carroll L, Fuller S, Saggese M, van Wettere A,
Redig P. Pathologic and immunohistochemical findings in goshawks
(Accipiter gentilis) and great horned owls (Bubo virginianus) naturally
infected with West Nile virus. Avian Dis 2005, 49(2):252-259.
Wnschmann A, Ziegler A. West Nile virus-associated mortality events in
domestic chukar partridges (Alectoris chukar) and domestic Impeyan
pheasants (Lophophorus impeyanus). Avian Dis 2006, 50(3):456-459.

134

Captulo 2

Zhang Z, Wilson F, Read R, Pace L, Zhang S. Detection and characterization of


naturally acquired West Nile virus infection in a female wild turkey. J Vet
Diagn Invest 2006, 18(2):204-208.

135

Captulo 3
Infeccin experimental de una especie
gallincea euromediterrnea con una cepa
norteamericana del virus West Nile

Gamino V, Escribano-Romero E, Blzquez AB, Gutirrez-Guzmn AV,


Saiz JC, Hfle U. Experimental infection of a Euro-Mediterranean
gallinaceous bird species with a North American West Nile virus
strain. En preparacin.

Captulo 3

RESUMEN
Mientras que la infeccin por el virus West Nile (WNV) da lugar a la muerte de miles
de aves en Norteamrica, la mortalidad de aves europeas es de carcter espordico
y afecta principalmente a las aves rapaces. El diferente comportamiento ecoepidemiolgico y la diferente virulencia de WNV entre estos dos escenarios y para
las diferentes especies de aves han sido relacionados, entre otras cosas, con la cepa
circulante del virus y la susceptibilidad del hospedador a la infeccin. En el
presente estudio inoculamos nueve perdices rojas (Alectoris rufa), de nueve
semanas de edad, con 107 UFP de una cepa norteamericana de WNV (NY99) con la
finalidad de determinar la susceptibilidad de una especie de ave euromediterrnea
endmica a esta cepa. Adems, estudiamos la dinmica de activacin y
reclutamiento de clulas inflamatorias en el sistema nervioso central (SNC) de las
aves eutanasiadas. La perdiz roja demostr ser susceptible a la infeccin con WNV
NY99, desarrollando lesiones y sucumbiendo a la enfermedad. Los tejidos ms
afectados fueron el corazn y el SNC. En este ltimo, las clulas de la microgla, los
astrocitos y las clulas T participaron en la encefalitis causada por WNV. A pesar de
la alta dosis de inoculacin utilizada, la replicacin del virus en tejidos fue mnima
y, en los individuos eutanasiados, no se detectaron lesiones severas, ms
probablemente relacionadas con la respuesta inflamatoria del hospedador, hasta el
da 10 post inoculacin, demostrando que esta cepa del virus es relativamente poco
virulenta para la perdiz roja.

139

Virginia Gamino Rodrguez

Tesis doctoral

ABSTRACT
While West Nile virus (WNV) infection produces thousands of bird deaths in
Northern America, European bird mortalities are rather sporadic and mainly
reported in raptors. The different eco-epidemiological behavior and virulence of
WNV between these two scenarios and among bird species have been related,
among others, to the circulating virus strain and host susceptibility to the infection.
In the present study we inoculated nine 9-week-old red-legged partridges (Alectoris
rufa) with 107 PFU of a North American WNV strain (NY99) in order to determine the
susceptibility of an endemic Euro-Mediterranean bird species to this strain.
Moreover, we studied the dynamics of inflammatory cell activation and recruitment
into the central nervous system (CNS) of euthanized birds. The red-legged partridge
proved to be susceptible to WNV NY99 infection, developing lesions and
succumbing to the disease. The most affected tissues were the heart and the CNS.
In the latter, microglial cells, astrocytes and T cells participated in the encephalitis
caused by WNV. Despite the high viral dose inoculated, viral replication in tissues
was minimal and severe lesions, most probably related to host inflammatory
response, were not detected in euthanized birds until day 10 post-inoculation,
demonstrating that this virus strain is of relatively low virulence for the red-legged
partridge.

140

Captulo 3

INTRODUCTION
West Nile virus (WNV) is a globally distributed mosquito-borne flavivirus whose
main reservoir are birds (Kramer et al., 2008). Before the 90's this arbovirus was
considered a pathogen of limited importance for birds (Zeller and Schuffenecker,
2004), but nowadays this scenario has changed, although with differences in the
eco-epidemiological behavior and virulence of the virus between the New and the
Old World and among bird species. While in Northern America WNV infection leads
to numerous outbreaks with thousands of bird deaths (CDC), European bird
mortalities are rather sporadic and mainly reported in raptors (Erdlyi et al., 2007;
Jimnez-Clavero et al., 2008; Wodak et al., 2011). Many of these differences are
related to the probability of the host to be infected by the virus and to susceptibility
to suffer disease once infected. On one hand, infection probability is influenced by
factors such as host geographic range, mating and breeding system, migratory
behavior and body size (Gancz et al., 2004; Reisen and Hahn, 2007). On the other
hand, infection pathogenesis is modulated by virulence determinants of the virus
strain (Beasley et al., 2004; Brault et al., 2007) and host capacity to fight the
infection (Gancz et al., 2004), which depends on the presence of previous immunity
against the virus or antigenically-related flaviviruses (Fang and Reisen, 2006;
Nemeth et al., 2008; Kwan et al., 2012), genetic factors (Tag-El-Din-Hassan et al.,
2012), age (Eldadah et al., 1967; Shirafuji et al., 2009) and the presence of
concurrent disease (Hfle et al., 2008) among others.
Susceptibility to both New and Old World

WNV strains has been

experimentally studied in bird species in Europe (Sotelo et al., 2011; Busquets et


al., 2012; Dridi et al., 2013; Ziegler et al., 2013). European hybrid falcons have
proven to be susceptible to different New York/1999 strains (Busquets et al., 2012;
Ziegler et al., 2013) and an endemic Mediterranean gallinaceous bird species, the
red-legged partridge (Alectoris rufa), developed disease and mortality up to 70%
141

Virginia Gamino Rodrguez

Tesis doctoral

after the experimental infection with two different Mediterranean WNV strains
(Sotelo et al., 2011). Based upon these results, the aim of the present study was to
describe the dynamics of virus appearance and distribution, as well as of
macroscopic and microscopic lesion distribution and severity in different tissues
during the course of the experimental infection of red-legged partridges with a WNV
New York/1999 strain. We also studied the dynamics of inflammatory cell
activation and recruitment into the brain of the WNV infected partridges, as this
information is available for murine models experimentally infected with similar
WNV strains, but not for birds.

MATERIAL AND METHODS


Animals
Recently hatched red-legged partridge chicks were raised in the experimental farm
of the Instituto de Investigacin en Recursos Cinegticos (IREC) until they were
eight weeks of age. In order to confirm that these birds had not been previously
infected by WNV, prior to the experimental infection they were tested by ELISA (ID
Screen West Nile Competition, IdVet, Montpellier, France) and by real time RTPCR (in cloacal and oropharyngeal swabs) as described previously (Crdoba et al.,
2007).

Virus
In this study we used a cell culture-passaged WNV New York/1999 (NY99) strain
(Crdoba et al., 2007; Martn-Acebes and Saiz, 2011).

Experimental infection
Partridges were transported to our biosafety level 3 (BSL-3) facilities in the Instituto
Nacional de Investigacin y Tecnologa Agraria y Alimentaria (INIA), where they were
142

Captulo 3

given five days for acclimatization. Nine 9-week-old partridges were subcutaneously
inoculated in the cervical region with 107 PFU/individual of the WNV NY99 strain
diluted in up to 0.1 mL Dulbecco's Minimum Essential Medium (DMEM)
(supplemented with 2 mM L-glutamine, 100 U/mL penicillin and 100 g/mL
streptomycin). Animals were observed daily for clinical signs or death and were
euthanized by intravenous injection of embutramide (T61 , Intervet ScheringPlough, Madrid, Spain) on days 3 (No. 2), 7 (No. 4), 10 (Nos. 6 and 7) and 14 (Nos. 8
and 9) post-inoculation (dpi).
All animals were handled in strict accordance with the guidelines of the
European Community 86/609/CEE and the protocols were approved by the
Committee on Ethics of animal experimentation of the INIA (permit number 2010015). Food and water were provided ad libitum throughout the experiment.

Sample collection
Detailed necropsies were performed on dead (Nos. 1, 3 and 5) and euthanized (Nos.
2, 4, 6-9) individuals. Samples of brain, heart, liver, spleen and kidney were
collected into sterile polypropylene tubes and stored at -70 C until analysis.
Additional samples of brain, oral mucosa, thymus, trachea, lung, heart, liver,
spleen, kidney, adrenal gland, testicle, ovary, small and large intestine, pancreas,
cecal tonsils, bursa of Fabricius, spinal cord, pectoral muscle and skin with feather
follicles were fixed in 10% neutral buffered formalin.

Virus genome detection


WNV RNA was extracted using a commercial Kit (Speedtools RNA virus extraction
kit, Biotools B&M Labs S.A, Madrid, Spain) and quantified by real time RT-PCR as
described previously (Crdoba et al., 2007). For RNA quantification a standard
curve was generated with previously titrated WNV (10 610-1 PFU) and samples were

143

Virginia Gamino Rodrguez

Tesis doctoral

considered negative when Ct35, equivalent to 102 PFU/sample (Blzquez and Saiz,
2010).

Histopathology
Formalin-fixed tissue samples were trimmed, embedded in paraffin and processed
to obtain 4 m sections that were stained with hematoxylin and eosin. These were
independently examined by two different investigators (UH and VG) to determine
the presence of WNV associated lesions. When lesions were present, these were
graded according to their distribution (focal, multifocal or diffuse) and severity
(mild, moderate or marked).

Immunohistochemistry
Tissue sections were also mounted on Vectabond reagent (Vector Laboratories,
Inc.,

Burlingame,

CA)

pretreated

slides

for

immunohistochemistry

(IHC).

Immunohistochemical detection of WNV antigen was performed using a rabbit


polyclonal antibody (BioReliance, Product 81-015, Rockville, MD) applying the same
protocol

used

previously

(Gamino

et

al.,

2012).

For

inflammatory

cell

characterization in the cerebrum, cerebellum, optic lobe and pons and medulla
oblongata, we used different antibodies, reagents and protocols detailed in table 1.
Endogenous peroxidase activity was inhibited with a peroxidase blocking reagent
(Dako EnVision+System-HRP (AEC), DakoCytomation, Carpinteria, CA) (CD3,
CD79, GFAP) or with 3% H202 diluted in methanol (RCA), rinses were done using
0.1% Tris-buffered saline/Tween20 (TBS 0.05 M, pH 7.5), non-specific primary
antibody labeling was blocked with 2% albumin from bovine serum (BSA) (SigmaAldrich Chemie, Steinheim, Germany) diluted in 0.1% TBS/Tween20, and sections
were counterstained with Mayer's hematoxylin.
In the IHC for WNV antigen detection, tissue sections of experimentally
infected red-legged partridges in which presence of WNV had been confirmed by real
144

Captulo 3

time RT-PCR served as positive controls. Negative controls included substitution of


the primary antibody by 2% BSA-0.1% TBS/Tween20 and a negative rabbit
antibody (BioReliance, Product 81015), as well as tissue sections of a red-legged
partridge negative in the WNV real time RT-PCR. Spleen and bursa of Fabricius
sections of red-legged partridges served as positive controls in the IHC for T and B
cells. In this case, negative controls included substitution of the primary antibody
by 2% BSA-0.1% TBS/Tween20 and a brain section of a red-legged partridge
negative in the WNV real time RT-PCR. A brain section of a non-infected partridge of
the same age served as reference for RCA and GFAP.
Virus antigen staining was graded according to its distribution and
abundance. We also determined the distribution of inflammatory cells within the
brain parenchyma and analyzed changes in their relative abundance.
Table 1. Detail of reagents and protocols used in the IHC for inflammatory
characterization in the CNS of experimentally WNV NY99 infected red-legged partridges.

cell

Cell
population/
marker

Antibody
reference*

Pretreatment**

Primary
antibody
dilution

Primary
antibody
incubation

Secondary
antibody

Detection
system

Microgliamacrophages

Lectin RCA-1
biotinylated

Citrate buffer
Microwave heat
(22 min)

1:600

45 min RT

Goat antirabbit IgG

ABC-DAB

Astrocytes

Polyclonal rabbit
anti-GFAP

Proteinase K
(7 min RT)

1:500

4C ON

T cells

Polyclonal rabbit
anti-human CD3

1:500

4C ON

Labelled
polymer-HRP
anti-rabbit

AEC+
substrate
chromogen

B cells

Monoclonal rabbit
anti-human CD79a

1:10

45 min RT

Citrate buffer
Microwave heat
(22 min)

*Primary antibody products: RCA-1 product No. B-1085 (Vector Laboratories); GFAP product No. Z0334
(DakoCytomation, Glostrup, Denmark); CD3 product No. A0452 (DakoCytomation); CD79a product No. RM-9118
(Thermo Fisher Scientific, Runcorn, UK).
**Proteinase K (DakoCytomation); RT: room temperature (22-25 C).
Antibodies were diluted in 2% BSA-0.1% TBS/Tween20.
ON: overnight.
Goat anti-rabbit IgG (Vector Laboratories) was diluted 1:200 in 0.1% TBS/Tween20 and applied for 1 hr at RT;
Labelled polymer-HRP anti-rabbit (Dako EnVision+System-HRP (AEC), DakoCytomation) was applied according to
manufacturer's recommendation.
Avidin-biotinylated enzyme complex (ABC system, Vector Laboratories) was applied for 30 min according to
manufacturer's recommendation and 3,3-diaminobenzidine tetrahydrochloride (DAB, Vector Laboratories) was
applied for 30 sec according to manufacturer's recommendations. AEC+substrate chromogen (Dako
EnVision+System-HRP (AEC), DakoCytomation) was applied for 15 min (CD3, CD79) and 3 min (GFAP) according
to manufacturer's recommendations.

145

Virginia Gamino Rodrguez

Tesis doctoral

RESULTS
Morbidity and mortality
Clinical signs were observed in the three partridges that died on days 2, 7 and 8 pi
(mortality of 45.5%). These birds showed depression, ruffled feathers and
recumbence, dying 24 hours after the onset of clinical signs.

Macroscopic findings
Macroscopic lesions compatible with WNV infection were detected in every
necropsied partridge, with minimal differences between dead and euthanized birds,
with the exception of the emaciation found only in fatally infected birds. Lesions
were more widespread between days 7 and 10 pi and the most affected organs were
the heart, liver, spleen and kidney. Pallor of the myocardium (3/9), hepatic (3/9),
splenic (3/9) and renal parenchyma (3/9), splenomegaly (2/9) and hepatomegaly
(2/9), as well as multifocal yellowish or reddish areas in the liver (2/9) were the
main macroscopic findings.
Additional macroscopic lesions, probably non-related to WNV infection, such
as dilation of intestinal loops (7/9) and injection of serosa vessels (4/9) (mainly in
the

duodenum and

cecum), and

distension

of the

crop

with a whitish

pseudomembrane covering the mucosa (2/9) were observed in some animals.

Virus genome detection


All tissue samples were negative for the presence of WNV genome in the real time
RT-PCR, with the exception of the heart of the bird found dead on 7 dpi (No. 3) and
the brain of one of the birds euthanized on 10 dpi (No. 6).

146

Captulo 3

Histopathology
Microscopic changes were observed early in the course of the infection, but while
these were severe in the bird found dead on 7 dpi, in euthanized partridges a
similar severity was not observed until 10 dpi (Table 2).
The most markedly affected tissues were the brain, spinal cord and heart.
While in fatally infected animals the spleen, liver, kidney, large intestine, pancreas
and bursa of Fabricius showed moderate lesions, in euthanized birds these tissues
were mildly affected or lesions were absent (Table 2). No microscopic changes were
detected in the trachea, cecal tonsils, testicles, ovaries, adrenal gland, and skin and
feather follicles.
Main

microscopic

findings

were

the

presence

of

lymphoplasmacytic,

histiocytic and/or granulocytic inflammatory infiltrates, cellular necrosis and


degeneration and hemosiderosis in the spleen and liver (Table 2 and figures 1-4).
While fatally infected animals showed moderate to marked lymphoid cell depletion
and necrosis in lymphoid organs, this was rarely observed in the euthanized ones.
The CNS mainly showed gliosis, perivascular cuffing and endothelial cell swelling,
but also neuronal necrosis and phagocytosis (Table 2 and figures 5 and 6). The bird
euthanized on 3 dpi only showed endothelial cell swelling, and moderate to marked
lesions detected in the CNS of birds found dead on 7 and 8 dpi, were not observed
until 10 dpi in euthanized animals.
As incidental findings, one fatally infected (No. 3) and five euthanized (Nos. 2
and 6-9) partridges showed moderate to marked hyperplasia, necrosis and
desquamation of the mucosa of the crop, associated to inflammatory infiltrates. In
these individuals pseudohyphae and blastospores of Candida sp. were confirmed in
keratin debris by staining with periodic-acid Schiff (PAS). Moreover, partridges
found dead on 7 and 8 dpi (Nos. 3 and 5) had mild to moderate amounts of

147

Virginia Gamino Rodrguez

Tesis doctoral

coccidian oocysts in the cecum, and those euthanized on 7 and 14 dpi (Nos. 4, 8
and 9) showed pancreatic granulomas.
Table 3. Microscopic lesions and antigen staining gradation on different days post-inoculation
(dpi) in tissues of experimentally WNV NY99 infected red-legged partridges.
2dpi

3dpi

7dpi

7dpi

8dpi

10dpi

No. 1

No. 2

No. 3

No. 4

No. 5

No. 6

Neuronal necrosis

Neuronal phagocytosis

++

Gliosis

+++

Perivascular cuffing

++

Endothelial cell swelling


Immunohistochemical
staining**
Cerebellum

++

Purkinje cell necrosis

Purkinje cell phagocytosis

Gliosis

14dpi

No. 7

No. 8

No. 9

++

++

++

++

++

++

++

++

+++

+++

++

++

++

Perivascular cuffing

++

Endothelial cell swelling

+++

++

++

Immunohistochemical staining

Neuronal necrosis

Neuronal phagocytosis

++

Gliosis

++

++

Perivascular cuffing

++

Endothelial cell swelling

++

++

Immunohistochemical staining

Neuronal necrosis

++

Neuronal phagocytosis

Gliosis

++

++

++

Perivascular cuffing

++

++

++

Endothelial cell swelling

++

++

++

++

Immunohistochemical staining

Neuronal necrosis

++

Neuronal phagocytosis

++

++

Gliosis

Perivascular cuffing

Endothelial cell swelling

++

++

++

++

++

+++

++

Immunohistochemical staining

Myofiber necrosis-degeneration

+++

+++

++

++

Inflammatory infiltrate

++

+++

++

+++

++

++

++

Immunohistochemical staining

++

++

Inflammatory infiltrate

+++

++

Immunohistochemical staining

Cerebrum*

Optic lobe

Pons + medulla oblongata

Spinal cord

Heart

Lung

148

Captulo 3

Table 3. Microscopic lesions and antigen staining gradation on different days post-inoculation
(dpi) in tissues of experimentally WNV NY99 infected red-legged partridges (cont).
2dpi

3dpi

7dpi

7dpi

8dpi

No. 1

No. 2

No. 3

No. 4

No. 5

No. 6

10dpi
No. 7

No. 8

14dpi
No. 9

Hepatocyte necrosis

+++

Hemosiderosis

Inflammatory infiltrate

Immunohistochemical staining

++

Follicular cell necrosis

Lymphoid depletion

+++

+++

Hemosiderosis

++

+++

Granulocytic infiltrate

+++

++

++

Eosinophilic material deposits

+++

+++

Immunohistochemical staining

Epithelial cell necrosis

++

++

Epithelial cell degeneration

++

Inflammatory infiltrate

++

Immunohistochemical staining

Glandular cell necrosis

Immunohistochemical staining

Inflammatory infiltrate

+++

++

Immunohistochemical staining

Inflammatory infiltrate

++

+++

Immunohistochemical staining

Granulocytic infiltrate

Immunohistochemical staining

Acinar cell necrosis

+++

++

Inflammatory infiltrate

++

Immunohistochemical staining

Bursa of Fabricius
Epithelia/lymphoid cell
necrosis
Lymphoid depletion

+++

++

Lymphoid cell necrosis

NA

Lymphoid depletion

NA

Granulocytic infiltrate

++

+++

NA

++

Immunohistochemical staining

NA

Liver

Spleen

Kidney

Proventriculus

Gizzard

Duodenum

Large intestine

Pancreas

Thymus

Partridges found dead


Euthanized partridges
*Tissue lesions were graded according to their distribution and severity: , no lesion; +, focal and mild or moderate /
multifocal and mild; ++, focal and marked / multifocal and moderate / diffuse and mild; +++, multifocal and marked /
diffuse and moderate or marked.
**WNV antigen immunostaining was graded according to its distribution and percentage of stained cells: , negative
staining; , focal single cells; +, focal or multifocal and < 20% cells stained; ++, multifocal or diffuse and 20-80% cells
stained; +++, multifocal or diffuse and > 80% cells stained.
NA: tissue sample no analyzed.

149

Virginia Gamino Rodrguez

Tesis doctoral

Figure 1. Heart, partridge found dead No. 5, 8 dpi. Focal and marked necrosis and degeneration of
cardiac myofibers, and infiltration of lymphoplasmacytic and histiocytic cells. Necrotic myofibers show
lysis of the nuclei, and fragmentation and accumulation of hyaline material in the cytoplasm. HE,
x400.
Figure 2. Liver, partridge found dead No. 3, 7 dpi. Diffuse and marked necrosis of hepatocytes, and
mild infiltration of lymphoplasmacytic and histiocytic cells. Necrotic hepatocytes show lysis and
picnosis of the nuclei, and degeneration of the cytoplasm. HE, x400.
Figure 3. Spleen, partridge found dead No. 6, 10 dpi. Distension of the cytoplasm of macrophages
by a red-brow pigment (arrows) and moderate infiltration of granulocytes (arrowheads). HE, x400.
Figure 4. Pancreas, partridge found dead No. 3, 7 dpi. Multifocal and marked necrosis of acinar
cells. Necrotic cells show lysis and picnosis of the nuclei and degeneration of the cytoplasm. HE, x400.

150

Captulo 3

Figure 5. Cerebrum, partridge found dead No. 5, 8 dpi. Diffuse and moderate gliosis with
perivascular infiltration of lymphoplasmacytic and histiocytic cells and satellitosis (arrowheads). HE,
x200.
Figure 6. Cerebellum, euthanized partridge No. 7, 10 dpi. Focal necrosis of Purkinje cells and focal
and moderate gliosis and degeneration in the molecular layer. HE, x400.

Immunohistochemistry
Virus antigen
Although the amount of WNV antigen detected by IHC was scarce, it was
widespread, especially on 8 and 10 dpi (Table 2).
Similar cell types were stained in euthanized and fatally infected birds. WNV
antigen was detected in cardiac myofibers (Figure 7), glomerular mensangial and
tubular epithelial cells of the kidney, acinar cells of the pancreas, and rare vascular
walls in the spleen and inflammatory cells in the lamina muscularis of the gizzard.
In fatally infected birds, WNV antigen was also observed in hepatocytes and Kupffer
cells (Figure 8), and in proventricular gland epithelial cells. By contrast, in
euthanized partridges, it was detected in splenic macrophages and in vascular walls
and muscular fibers of the lamina muscularis of the large intestine. In the CNS, the
partridge euthanized on 7 dpi had only one intravascular inflammatory cell stained
in the cerebrum and the two birds euthanized on 10 dpi showed positive staining
within the cytoplasm of one Purkinje cell, several neuronal axons and in very few
glial cells in the cerebellum (Figure 9). On 14 dpi, virus antigen was only detected in
bird No. 8, in one inflammatory cell in the spinal cord and one cardiac myofiber.
Inflammatory cells in the brain
Inflammatory cells that participated in the encephalitis associated to WNV infection
were only analyzed in euthanized partridges.

151

Virginia Gamino Rodrguez

Tesis doctoral

Microglia activation/macrophage infiltration


Microglial cells reacted as early as 3 dpi, but there were changes in their
morphology and abundance during the course of the infection and among brain
regions.
On 3 dpi, most microglial cells displayed a poorly ramified morphology,
although some amoeboid cells, characterized by a large soma and short thick
cellular processes, were observed. On 7 dpi, most cells changed to an amoeboid
morphology, and rounded cells, which corresponded both to activated microglia and
infiltrating macrophages, were also observed. All these cells were especially
numerous on 10 dpi, diffusely distributed or forming nodules or foci that, in some
cases, surrounded dying neurons and blood vessels. On 14 dpi, microglia remained
very active. Some macrophages were also detected in the vascular lumen of
meningeal vessels.
In the cerebrum, on 3 dpi, ramified cells were diffusely distributed but, from
7 dpi on, these were more numerous and intensely stained in the peripheral
pallium. Nevertheless, from 7 dpi on, amoeboid and rounded cells were
predominant and especially active in the parenchyma near the lateral ventricle,
where some cells were also detected near ependymal cells.
In the cerebellum, activated microglial cells were mainly detected in the
molecular layer. In this layer, on 10 dpi, and especially in bird No. 7, most cells
were grouped forming numerous nodules, many of them surrounding necrotic
Purkinje cells (Figure 10). Moderate activation was also observed in the white
matter that, in change, was mild in the granular layer.
In the optic lobe, activated microglial cells were detected in every layer, but
microglia nodules were mainly located in the stratum griseum et fibrosum
superficiale. These cells were especially numerous in bird No. 6 (10 dpi).

152

Captulo 3

The pons and medulla oblongata were the brain regions that showed more
marked microglia activation. As early as 3 dpi, ramified microglial cells were
diffusely distributed, but amoeboid and rounded cells were also abundant. On 10
dpi, and also on 14 dpi, there were multiple microglia nodules, also located in some
neuronal nuclei.
Astrocyte activation
GFAP+ cells were detected from 3 dpi on. Mild changes in cellular morphology,
abundance and distribution compared to a non-infected partridge were noticed, but
none in staining intensity. On 14 dpi, astrocyte activation was slightly higher.
In the cerebrum, GFAP+ cells with long cellular processes were stained in the
subpial region and many vessels were surrounded by astrocytes with medium
cellular processes, mainly located in the peripheral parenchyma. Mild astrocytosis
was detected in the peripheral pallium, especially near the lateral ventricle,
characterized by star-shaped cells with short or medium cellular processes.
In the cerebellum, the granular layer showed scarce stained astrocytes,
mainly located near the Purkinje cell layer (Bergmann glia) (Figure 11). GFAP+ cells
were also scarce in the white matter and absent in the molecular layer.
In the optic lobe, numerous astrocytes were stained in the stratum opticum,
some of them surrounding blood vessels. On 14 dpi, mild astrocytosis was detected
in the stratum album centrale, and the ependymal cells surrounding the ventricle
were GFAP+.
In the pons and medulla oblongata, astrocytes with long cellular processes
were stained in the dorsal and ventral subpial zone and many vessels were
surrounded by GFAP+ cells. On 3 and 7 dpi, mild astrocytosis was detected in the
medulla oblongata that, in change, was detected on 10 dpi in the pons. On 14 dpi,
moderate diffuse astrocytosis was observed, especially in bird No. 8.

153

Virginia Gamino Rodrguez

Tesis doctoral

T cell infiltration
CD3+ T cells were detected early, but changes in their distribution and abundance
during the course of the infection and differences among brain regions were
observed.
On 3 and 7 dpi, CD3+ T cells were mainly observed in the vascular lumen of
meningeal and parenchymal vessels, and only scarce cells infiltrated the
interstitium of the cerebrum, cerebellum and pons and medulla oblongata. It was
on 10 dpi when T cells were especially numerous in the brain. On this dpi, these
cells were mainly diffusely distributed or forming part of inflammatory nodules or
foci, but were also observed in the perivascular space and vascular lumen. On 14
dpi, while the cellular infiltration decreased markedly in the cerebellum and optic
lobe, numerous T cells were still detected in the cerebrum and pons and medulla
oblongata.
In the cerebrum, T cells were diffusely distributed in the parenchyma.
Numerous T cells were detected in inflammatory nodules, inflammatory foci and in
some perivascular cuffs, especially in bird No. 7 (10 dpi).
In the cerebellum, the molecular layer was markedly infiltrated on 10 dpi,
showing numerous cells diffusely distributed and forming part of inflammatory
nodules, many of them surrounding Purkinje cells, which in most cases showed
necrosis. At this time, moderate infiltration was also detected in the granular layer
and white matter, especially in bird No. 7.
The optic lobe was the brain region in which less T cell infiltration was
observed. Although T cells were detected in every layer, these were slightly more
numerous in the stratum opticum and stratum griseum et fibrosus superficiale
and, to a lesser extent, in the stratum griseum and album centrale.

154

Captulo 3

The pons and medulla oblongata were the brain regions most markedly
infiltrated by T cells, either diffusely distributed or forming part of inflammatory
nodules (Figure 12), both on 10 and 14 dpi.
B cell infiltration
CD79a+ cells were not detected in any brain region on any dpi.

155

Virginia Gamino Rodrguez

Tesis doctoral

Figure 7. Heart; partridge found dead No. 5, 8 dpi. WNV antigen in the cytoplasm of cardiac
myofibers. IHC for WNV with Mayer's hematoxylin counterstain, x400.
Figure 8. Liver; partridge found dead No. 3, 7 dpi. WNV antigen in the cytoplasm of hepatocytes
and Kupffer cells. IHC for WNV with Mayer's hematoxylin counterstain, x400.
Figure 9. Cerebellum; euthanized partridge No. 6, 10 dpi. WNV antigen in the cytoplasm and axons
of Purkinje cells. IHC for WNV with Mayer's hematoxylin counterstain, x400.
Figure 10. Cerebellum; euthanized partridge No. 7, 10 dpi. RCA-1 positive staining in the
cytoplasm of activated microglia/macrophages forming nodules in the molecular layer. IHC for
microglia activation/macrophages with Mayer's hematoxylin counterstain, x400.
Figure 11. Cerebellum; euthanized partridge No. 8, 14 dpi. GFAP positive staining in cell processes
of astrocytes diffusely distributed in the granular layer near the Purkinje cell layer (Bergmann glia).
IHC for astrocyte activation with Mayer's hematoxylin counterstain, x100.
Figure 12. Medulla oblongata; euthanized partridge No. 7, 10 dpi. CD3 positive staining in the
cytoplasm of T cells forming an inflammatory nodule in the neuropil. IHC for T cells with Mayer's
hematoxylin counterstain, x400.

DISCUSSION
In this work we have demonstrated that the juvenile red-legged partridge, an
endemic Euro-Mediterranean gallinaceous bird species, is susceptible to the
experimental infection with a North American WNV strain, developing clinical signs,
macroscopic and microscopic lesions, and succumbing to the disease. To our
knowledge, this is the first experimental infection of a European gallinaceous bird
species with a North American WNV strain and one of the few experimental studies
that have been done with a European endemic bird species.
Gallinaceous birds have been traditionally considered little susceptible to
WNV infection (Komar et al., 2003). However, sporadic cases of West Nile disease
and mortality have been described in Northern America (Steele et al., 2000; Zhang
et al., 2006), where an outbreak among chukar partridges (Alectoris chukar), a bird
species closely-related to the red-legged partridge, occurred (Wnschmann and
Ziegler, 2006). Although experimental infections in gallinaceous birds have
demonstrated their little susceptibility to WNV infection (Swayne et al., 2000), the
red-legged partridge proved to be susceptible to two Mediterranean WNV strains in
a previous experimental infection, suffering moderate (30%) to high (70%) mortality,
developing high viremia levels and producing high titers of neutralizing antibodies
(Sotelo et al., 2011). While no WNV associated mortality event has been recorded in
156

Captulo 3

gallinaceous birds in Europe, the red-legged partridge and the ring-necked


pheasant (Phasianus colchicus) have found to be susceptible to natural infection by
a closely-related flavivirus, Bagaza virus (Gamino et al., 2012).
In the present work, we only detected WNV genome by real time RT-PCR in
the heart of one partridge that succumbed to the infection on 7 dpi and in the brain
of one animal euthanized on 10 dpi. This could be related to poor virus replication
and a rapid virus clearance by the host, which matches with the scarce virus
antigen detected by IHC. However, most infected partridges developed multiorgan
macroscopic and

microscopic

lesions,

probably

more

related

to

the

host

inflammatory response than to virus replication. These lesions, as expected, were


more severe and widespread in fatally infected birds and as infection progressed.
The most severely affected tissues were the CNS and the heart, as it had been
previously described in Gyr-Saker hybrid falcons inoculated with the same WNV
strain (Busquets et al., 2012).
One partridge died as early as 2 dpi; however, this bird only showed mild
microscopic changes and we could not detect WNV by real time RT-PCR or IHC.
Considering that the partridge euthanized on 3 dpi had only mild acute lesions and
WNV antigen was scarcely present in different tissues, and that even highly
susceptible species such as crows or jays do not usually succumb to WNV infection
until 4-5 dpi (Weingartl et al., 2004; Nemeth et al., 2011), we cannot rule out other
underlying causes.
Incidental findings such as the presence of coccidian oocysts and Candida
sp. in some individuals were probably enhanced by the impairment of host defenses
by the viral infection, as it had been previously described in natural WNV infections
(Hfle et al., 2008).
In the CNS of euthanized birds, although resident immune cells (i.e.
microglia and astrocytes) showed mild reaction as early as 3 dpi, it was on 10 dpi
157

Virginia Gamino Rodrguez

Tesis doctoral

when the inflammatory reaction was more marked, virus genome was detected by
real time RT-PCR, virus antigen was detected by IHC, and moderate to marked
microscopic lesions were observed. A similar sequence of virus antigen appearance,
development of lesions, and inflammatory reaction in the CNS has been described
in rodent models experimentally infected with WNV NY99 strains (Brhin et al.,
2008) and in red-legged partridges experimentally infected with Mediterranean WNV
strains (the authors, under revision); however, in both cases, this took place earlier
in the course of the infection.
The more prevalent inflammatory cell population on 10 dpi was microglia.
These resident immune cells act as CNS macrophages, phagocytizing damaged cells
and releasing immune mediators (Gehrmann et al., 1995), and are essential in the
anti-flavivus response (Brhin et al., 2008; Maximova et al., 2009). The other
resident cells, astrocytes, showed mild changes during the course of the infection
and, as expected, were more active on 14 dpi. These cells also participate in the
encephalitis associated to WNV infection in humans and mice (Kelley et al., 2003;
Hussmann et al., 2013). Microglia and astrocyte activation, together with other
stimuli, resulted in CD3+ T cell recruitment to the CNS, which infiltrated the
parenchyma from 7 dpi onwards. These immune cells are able to destroy cells
infected by the virus and are essential for CNS recovering from WNV infection, both
in humans and rodents (Sampson and Armbrustmacher, 2001; Brhin et al., 2008).
All these inflammatory cells were especially active in the pons and medulla
oblongata. This could be related to the fact that, as in humans (Sampson et al.,
2000) and horses (Cantile et al., 2001), the brain stem is the primary target for
WNV. In fact, infected birds usually show microscopic lesions in this region (Steele
et al., 2000; Gancz et al., 2006; Zhang et al., 2006). The absence of CD79a stained
cells was probably related to malfunction of the primary antibody rather than the
real absence of B cell infiltration. Nevertheless, studies done in humans and
158

Captulo 3

rodents have indicated that B cells play a limited role in the encephalitis associated
to WNV infection and are mainly detected in the perivascular space (Kelley et al.,
2003; Wang et al., 2003). Despite the important role of inflammatory cells in the
recovery of the CNS from WNV infection, it has been demonstrated that an
exacerbated and prolonged activation or recruitment of these cells can have
detrimental effects, contributing to neuron damage (Wang et al., 2003; Brhin et al.,
2008). It is probable that, in our study, CNS lesions were more related to host
response than to virus replication within cells, as virus genome was only detected in
one animal euthanized on 10 dpi and virus antigen was scarcely present only in the
cerebellum of the two individuals euthanized on the same dpi.
Lesions observed in red-legged partridges were similar to those described in
birds naturally and experimentally infected with WNV (Steele et al., 2000; Nemeth
et al., 2006; Ziegler et al., 2013). Although similar moderate to marked microscopic
lesions have been described in the CNS of some naturally infected gallinaceous
birds (Steele et al., 2000; Zhang et al., 2006), these are not always present both in
natural and experimental infections (Senne et al., 2000; Swayne et al., 2000;
Wnschmann and Ziegler, 2006). However, differences in infection conditions such
as the specific virus strain and inoculation dose, and the age, species and number
of birds infected, could be determining differences in pathological findings. In fact,
considering the poor virus replication in tissues and the relatively low mortality
found

in our study,

despite

the

high virus inoculation dose

used

(107

PFU/individual), it might be possible that, if we had given a lower dose, viral


replication, development of lesions and clinical disease might not have taken place
or, at least, might have been milder.
If we compare the infection pathogenesis and pathogenicity observed in the
present study with the results of the experimental infection of the same species
with Mediterranean WNV strains, differences are evident, mainly compared to
159

Virginia Gamino Rodrguez

Tesis doctoral

Morocco/2003 (the authors, under revision). In the present study, virus replication
in tissues was much lower and microscopic lesions in euthanized partridges were
milder and appeared later in the course of the infection (the authors, under
revision). Although birds inoculated with the Mediterranean isolates were two weeks
younger, and host susceptibility to infection is strongly related to this factor
(Wnschmann and Ziegler, 2006; Shirafuji et al., 2009), these were inoculated with
104 PFU, which is three logs lower than the inoculation dose of the present study.
From our point of view, this implies that WNV NY99 is less virulent for the redlegged partridge than Morocco/2003 and Spain/2007. However, the low number of
birds used in both studies and the different experimental conditions, prevents us
from drawing clear conclusions.
Summarizing, in the present study we have demonstrated that an endemic
Euro-Mediterranean gallinaceous bird species is susceptible to a North American
WNV strain, developing lesions, such as myocarditis and encephalitis, clinical
signs, and succumbing to the disease. Considering our results, together with the
fact that some European birds have suffered mortalities due to WNV infection
(Bakonyi et al., 2006; Monaco et al., 2010; Wodak et al., 2011; Bakonyi et al.,
2013), the low consequences of WNV infection for birds in Europe, and especially in
the Mediterranean basin, where outbreaks of West Nile fever have mainly affected
humans and equids (Murgue et al., 2001; Monaco et al., 2010; Garca-Bocanegra et
al., 2011), apparently are not related with a low susceptibility of European and
Mediterranean birds to WNV infection and disease. Other factors such as the
presence of previous immunity in the host, host and vector abundance in the area,
the degree of biodiversity or difficulties to identify WNV infection as the cause of
bird mortalities should be considered among others.

160

Captulo 3

ACKNOWLEDGEMENTS
This study was supported by grants AG2008-02504GAN and SAF-2008-04232
funded by the Spanish Ministry for Science and Innovation, grants FAU2008 000600-00 and RTA2011-0036 funded by the Spanish Institute for Agricultural and
Alimentary Investigation and Technology (INIA), and The Network of Animal Disease
Infectiology and Research Facilities, NADIR-UE-228394, funded by the E.U. V.
Gamino (323/09) is a research fellow supported by the regional government of
Castilla La Mancha (JCCM). We are grateful to the personnel of the experimental
farm of the University of Castilla La Mancha, La Galiana, for their help during
the breeding of partridges.

REFERENCES
Bakonyi T, Ivanics E, Erdlyi K, Ursu K, Ferenczi E, Weissenbck H, Nowotny N.
Lineage 1 and 2 strains of encephalitic West Nile virus, central Europe.
Emerg Infect Dis 2006, 12(4):618-623.
Bakonyi T, Ferenczi E, Erdlyi K, Kutasi O, Csorgo T, Seidel B, Weissenbck H,
Brugger K, Ban E, Nowotny N. Explosive spread of a neuroinvasive lineage 2
West Nile virus in Central Europe, 2008/2009. Vet Microbiol 2013. pii:
S0378-1135(13)00164-8. doi: 10.1016/j.vetmic.2013.03.005.
Beasley DW, Davis CT, Whiteman M, Granwehr B, Kinney RM, Barrett AD.
Molecular determinants of virulence of West Nile virus in North America.
Arch Virol Suppl 2004(18):35-41.
Blzquez AB, Siz JC. West Nile virus (WNV) transmission routes in the murine
model: intrauterine, by breastfeeding and after cannibal ingestion. Virus
Res 2010, 151(2):240-243. doi: 10.1016/j.virusres.2010.04.009.
Brault AC, Huang CY, Langevin SA, Kinney RM, Bowen RA, Ramey WN, Panella NA,
Holmes EC, Powers AM, Miller BR. A single positively selected West Nile
viral mutation confers increased virogenesis in American crows. Nat Genet
2007, 39(9):1162-1166.
Brhin AC, Mouries J, Frenkiel MP, Dadaglio G, Despres P, Lafon M, Couderc T.
Dynamics of immune cell recruitment during West Nile encephalitis and
identification of a new CD19+B220-BST-2+ leukocyte population. J
Immunol 2008, 180(10):6760-6767.

161

Virginia Gamino Rodrguez

Tesis doctoral

Busquets N, Bertran K, Costa TP, Rivas R, de la Fuente JG, Villalba R, Solanes D,


Bensaid A, Maj N, Pags N. Experimental West Nile virus infection in GyrSaker hybrid falcons. Vector Borne Zoonotic Dis 2012, 12(6):482-489.
Cantile C, Del Piero F, Di Guardo G, Arispici M. Pathologic and
immunohistochemical findings in naturally occuring West Nile virus
infection in horses. Vet Pathol 2001, 38(4):414-421.
Centers
for
Disease
Control
and
Prevention
[http://www.cdc.gov/ncidod/dvbid/westnile/birdspecies.htm]

(CDC)

Crdoba L, Escribano-Romero E, Garmendia A, Saiz JC. Pregnancy increases the


risk of mortality in West Nile virus-infected mice. J Gen Virol 2007, 88:476480.
Dridi M, Vangeluwe D, Lecollinet S, van den Berg T, Lambrecht B. Experimental
infection of Carrion crows (Corvus corone) with two European West Nile
virus (WNV) strains. Vet Microbiol 2013. pii: S0378-1135(13)00061-8. doi:
10.1016/j.vetmic.2012.12.043.
Eldadah AH, Nathanson N, Sarsitis R. Pathogenesis of West Nile Virus
encephalitis in mice and rats. 1. Influence of age and species on mortality
and infection. Am J Epidemiol 1967, 86(3):765-775.
Erdlyi K, Ursu K, Ferenczi E, Szeredi L, Ratz F, Skare J, Bakonyi T. Clinical and
pathologic features of lineage 2 West Nile virus infections in birds of prey
in Hungary. Vector Borne Zoonotic Dis 2007, 7(2):181-188.
Fang Y, Reisen WK. Previous infection with West Nile or St. Louis encephalitis
viruses provides cross protection during reinfection in house finches. Am J
Trop Med Hyg 2006, 75(3):480-485.
Gamino V, Gutirrez-Guzmn AV, Fernndez-de-Mera IG, Ortz JA, Durn-Martn
M, de la Fuente J, Gortzar C, Hfle U. Natural Bagaza virus infection in
game birds in southern Spain. Vet Res 2012, 43(1):65. doi: 10.1186/12979716-43-65.
Gancz AY, Barker IK, Lindsay R, Dibernardo A, McKeever K, Hunter B. West Nile
virus outbreak in North American owls, Ontario, 2002. Emerg Infect Dis
2004, 10(12):2135-2142.
Gancz AY, Smith DA, Barker IK, Lindsay R, Hunter B. Pathology and tissue
distribution of West Nile virus in North American owls (family : Strigidae).
Avian Pathology 2006, 35(1):17-29.
Garca-Bocanegra I, Jan-Tellez JA, Napp S, Arenas-Montes A, Fernndez-Morente
M, Fernndez-Molera V, Arenas A. West Nile fever outbreak in horses and
humans, Spain, 2010. Emerg Infect Dis 2011, 17(12):2397-2399.
Gehrmann J, Matsumoto Y, Kreutzberg GW. Microglia: intrinsic immuneffector
cell of the brain. Brain Res Brain Res Rev 1995, 20(3):269-287.
Hfle U, Blanco JM, Crespo E, Naranjo V, Jimnez-Clavero MA, Snchez A, de la
Fuente J, Gortzar C. West Nile virus in the endangered Spanish imperial
eagle. Vet Microbiol 2008, 129(1-2):171-178.
162

Captulo 3

Hussmann KL, Samuel MA, Kim KS, Diamond MS, Fredericksen BL. Differential
replication of pathogenic and nonpathogenic strains of West Nile virus
within astrocytes. J Virol 2013, 87(5):2814-2822.
Jimnez-Clavero MA, Sotelo E, Fernndez-Pinero J, Llorente F, Manuel Blanco J,
Rodrguez-Ramos J, Prez-Ramrez E, Hfle U. West Nile virus in golden
eagles, Spain, 2007. Emerg Infect Dis 2008, 14(9):1489-1491.
Kelley TW, Prayson RA, Ruiz AI, Isada CM, Gordon SM. The neuropathology of
West Nile virus meningoencephalitis. A report of two cases and review of
the literature. Am J Clin Pathol 2003, 119(5):749-753.
Komar N, Langevin S, Hinten S, Nemeth N, Edwards E, Hettler D, Davis B, Bowen
R, Bunning M. Experimental infection of North American birds with the
New York 1999 strain of West Nile virus. Emerg Infect Dis 2003, 9(3):311322.
Kramer LD, Styer LM, Ebel GD. A global perspective on the epidemiology of
West Nile virus. Annu Rev Entomol 2008, 53:61-81.
Kwan JL, Kluh S, Reisen WK. Antecedent avian immunity limits tangential
transmission of West Nile virus to humans. PLoS One 2012, 7(3):e34127. doi:
10.1371/journal.pone.0034127.
Martn-Acebes MA, Saiz J-C. A West Nile virus mutant with increased resistance
to acid-induced inactivation. J Gen Virol 2011, 92:831-840.
Maximova OA, Faucette LJ, Ward JM, Murphy BR, Pletnev AG. Cellular
inflammatory response to flaviviruses in the central nervous system of a
primate host. J Histochem Cytochem 2009, 57(10):973-989.
Monaco F, Lelli R, Teodori L, Pinoni C, Di Gennaro A, Polci A, Calistri P, Savini G.
Re-emergence of West Nile virus in Italy. Zoonoses Public Health 2010, 57(78):476-486.
Murgue B, Murri S, Triki H, Deubel V, Zeller HG. West Nile in the Mediterranean
basin: 1950-2000. Ann N Y Acad Sci 2001, 951:117-126.
Nemeth N, Gould D, Bowen R, Komar N. Natural and experimental West Nile
virus infection in five raptor species. J Wildl Dis 2006, 42(1):1-13.
Nemeth NM, Kratz GE, Bates R, Scherpelz JA, Bowen RA, Komar N. Naturally
induced humoral immunity to West Nile virus infection in raptors.
Ecohealth 2008, 5(3):298-304.
Nemeth NM, Thomsen BV, Spraker TR, Benson JM, Bosco-Lauth AM, Oesterle PT,
Bright JM, Muth JP, Campbell TW, Gidlewski TL, Bowen RA. Clinical and
pathologic responses of American crows (Corvus brachyrhynchos) and fish
crows (C ossifragus) to experimental west nile virus Infection. Vet Pathol
2011, 48(6):1061-1074.
Reisen WK, Hahn DC. Comparison of immune responses of brown-headed
cowbird and related blackbirds to west Nile and other mosquito-borne
encephalitis viruses. J Wildl Dis 2007, 43(3):439-449.
163

Virginia Gamino Rodrguez

Tesis doctoral

Sampson BA, Ambrosi C, Charlot A, Reiber K, Veress JF, Armbrustmacher V. The


pathology of human West Nile Virus infection. Hum Pathol 2000, 31(5):527531.
Sampson BA, Armbrustmacher V. West Nile encephalitis: the neuropathology of
four fatalities. Ann N Y Acad Sci 2001, 951:172-178.
Senne DA, Pedersen JC, Hutto DL, Taylor WD, Schmitt BJ, Panigrahy B.
Pathogenicity of West Nile virus in chickens. Avian Dis 2000, 44(3):642-649.
Shirafuji H, Kanehira K, Kubo M, Shibahara T, Kamio T. Experimental West Nile
virus infection in aigamo ducks, a cross between wild ducks (Anas
platyrhynchos) and domestic ducks (Anas platyrhynchos var. domesticus).
Avian Dis 2009, 53(2):239-244.
Sotelo E, Gutirrez-Guzmn AV, Del Amo J, Llorente F, El-Harrak M, Prez-Ramrez
E, Blanco JM, Hfle U, Jimnez-Clavero MA. Pathogenicity of two recent
Western Mediterranean West Nile virus isolates in a wild bird species
indigenous to Southern Europe: the red-legged partridge. Vet Res 2011,
42(1):11. doi: 10.1186/1297-9716-42-11.
Steele KE, Linn MJ, Schoepp RJ, Komar N, Geisbert TW, Manduca RM, Calle PP,
Raphael BL, Clippinger TL, Larsen T, Smith J, Lanciotti RS, Panella NA,
McNamara TS. Pathology of fatal West Nile virus infections in native and
exotic birds during the 1999 outbreak in New York City, New York. Vet
Pathol 2000, 37(3):208-224.
Swayne DE, Beck JR, Zaki S. Pathogenicity of West Nile virus for turkeys. Avian
Dis 2000, 44(4):932-937.
Tag-El-Din-Hassan HT, Sasaki N, Moritoh K, Torigoe D, Maeda A, Agui T. The
chicken 2'-5' oligoadenylate synthetase A inhibits the replication of West
Nile virus. Jpn J Vet Res 2012, 60(2-3):95-103.
Wang Y, Lobigs M, Lee E, Mullbacher A. CD8+ T cells mediate recovery and
immunopathology in West Nile virus encephalitis. J Virol 2003,
77(24):13323-13334.
Weingartl HM, Neufeld JL, Copps J, Marszal P. Experimental West Nile virus
infection in blue jays (Cyanocitta cristata) and crows (Corvus
brachyrhynchos). Vet Pathol 2004, 41(4):362-370.
Wodak E, Richter S, Bago Z, Revilla-Fernndez S, Weissenbock H, Nowotny N,
Winter P. Detection and molecular analysis of West Nile virus infections in
birds of prey in the eastern part of Austria in 2008 and 2009. Vet Microbiol
2011, 149(3-4):358-366.
Wnschmann A, Ziegler A. West Nile virus-associated mortality events in
domestic chukar partridges (Alectoris chukar) and domestic Impeyan
pheasants (Lophophorus impeyanus). Avian Dis 2006, 50(3):456-459.
Zeller HG, Schuffenecker I. West Nile virus: an overview of its spread in Europe
and the Mediterranean basin in contrast to its spread in the Americas. Eur
J Clin Microbiol Infect Dis 2004, 23(3):147-156.
164

Captulo 3

Zhang Z, Wilson F, Read R, Pace L, Zhang S. Detection and characterization of


naturally acquired West Nile virus infection in a female wild turkey. J Vet
Diagn Invest 2006, 18(2):204-208.
Ziegler U, Angenvoort J, Fischer D, Fast C, Eiden M, Rodriguez AV, RevillaFernandez S, Nowotny N, de la Fuente JG, Lierz M, Groschup MH.
Pathogenesis of West Nile virus lineage 1 and 2 in experimentally infected
large falcons. Vet Microbiol 2013, 161(3-4):263-273.

165

Captulo 4
Infeccin natural por el virus Bagaza en
aves cinegticas en el sur de Espaa

Gamino V, Gutirrez-Guzmn AV, Fernndez-de-Mera IG, Ortz JA,


Durn-Martn M, de la Fuente J, Gortzar C, Hfle U. Natural Bagaza
virus infection in game birds in southern Spain. Veterinary Research
2012, 43(1): 64. doi: 10.1186/1297-9716-43-65.

Captulo 4

RESUMEN
A finales del verano de 2010 el virus Bagaza (BAGV), un flavivirus transmitido por
mosquitos que nunca haba sido detectado previamente en Europa, caus un brote
de elevada mortalidad en perdiz roja (Alectoris rufa) y faisn comn (Phasianus
colchicus). En el presente trabajo se estudiaron los hallazgos clnicos y la
distribucin de lesiones y del antgeno viral en aves cinegticas infectadas de forma
natural con BAGV, con el fin de explicar el impacto aparentemente ms severo en la
poblacin de perdiz roja. En ambas especies de galliformes y, en menor medida, en
paloma torcaz (Columba palumbus), la infeccin dio lugar a signos nerviosos.
Adems, en las perdices sta caus una hemosiderosis severa en el hgado y en el
bazo que, por el contrario, no estuvo presente en los faisanes y fue menos evidente
en las palomas. Mientras que el antgeno viral fue detectado en el endotelio vascular
en diversos rganos de las perdices y en el bazo de las palomas, en faisanes slo fue
encontrado en neuronas y clulas de la gla en cerebro. Estos hallazgos indican la
existencia de un tropismo endotelial por parte de BAGV y un proceso hemoltico
severo en perdiz roja que se sum al proceso neurolgico encontrado en las tres
especies.

169

Virginia Gamino Rodrguez

Tesis doctoral

ABSTRACT
In late summer 2010 a mosquito-borne flavivirus not previously reported in Europe
called Bagaza virus (BAGV) caused high mortality in red-legged partridges (Alectoris
rufa) and ring-necked pheasants (Phasianus colchicus). We studied clinical findings,
lesions and viral antigen distribution in naturally BAGV infected game birds in
order to understand the apparently higher impact on red-legged partridges. The
disease induced neurologic signs in the two galliform species and, to a lesser extent,
in common wood pigeons (Columba palumbus). In red-legged partridges infection by
BAGV caused severe haemosiderosis in the liver and spleen that was absent in
pheasants and less evident in common wood pigeons. Also, BAGV antigen was
present in vascular endothelium in multiple organs in red-legged partridges, and in
the spleen in common wood pigeons, while in ring-necked pheasants it was only
detected in neurons and glial cells in the brain. These findings indicate tropism of
BAGV for endothelial cells and a severe haemolytic process in red-legged partridges
in addition to the central nervous lesions that were found in all three species.

170

Captulo 4

INTRODUCTION
In late summer 2010 an extremely high mortality was observed among game birds
in southern Spain, especially in red-legged partridges (Alectoris rufa), but also in
ring-necked pheasants (Phasianus colchicus), that was determined to be due to a
flavivirus not previously reported in Europe, Bagaza virus (BAGV) (Agero et al.,
2011). BAGV is a relatively unknown member of the Ntaya group of the genus
Flavivirus that was first isolated in the Central African Republic in 1966 from a pool
of Culex spp. mosquitoes (Digoutte, 1978).
Although a recent study has examined the epidemiology of the outbreak more
closely (Garca-Bocanegra et al., 2012), information on the pathogenesis of BAGV is
very limited. Thus, in the present study we review the main clinical findings, lesions
and viral antigen distribution in wild birds naturally infected with BAGV. Moreover,
we compare these features among BAGV-infected red-legged partridges, ring-necked
pheasants and common wood pigeons (Columba palumbus), in order to understand
the pathogenesis of the disease and the reason for the extreme impact of the
disease in red-legged partridges in comparison to other affected species.

MATERIAL AND METHODS


Study area
The study area is located on the Mediterranean coast, in south-western Spain
(between 36 20 N and 5 48 O). The climate is Mediterranean, with wet winters
and dry summers. The habitat is a mosaic of intensively managed crops, primarily
rice and vegetables, and open woodland (dehesa) with cork oaks (Quercus suber),
and stone pines (Pinus pinea).

171

Virginia Gamino Rodrguez

Tesis doctoral

Postmortem examination and sampling


Recently dead or moribund red-legged partridges (n = 6), ring-necked pheasants
(n = 5) and common wood pigeons (n = 2) were collected in the area during three
consecutive days. Oral and cloacal swabs were taken and stored in viral transport
medium (Hanks' balanced salt solution containing 10% glycerol, 200 U/mL
penicillin, 200 g/mL streptomycin, 100 U/mL polymixin B sulphate, 250 g/mL
gentamicin and 50 U/mL nystatin (Munster et al., 2007)) and frozen immediately in
liquid nitrogen.
Detailed necropsies were carried out, and a complete set of tissues was taken
(brain, oral mucosa, pectoral muscle, trachea, lung, heart, liver, spleen, pancreas,
duodenum, caecal tonsils, kidney, bursa of Fabricius, thymus and skin with feather
follicles)

and

fixed

in

10%

neutral

buffered

formalin

for

histopathologic

examination. Additionally, samples of heart, kidney, brain and spleen were also
collected into sterile containers and frozen immediately in liquid nitrogen.

Real time RT-PCR and sequence analysis


Swabs and frozen tissue samples were processed upon arrival at the laboratory.
Nucleic acid (RNA) was extracted using High Pure RNA Tissue Kit (Roche
Diagnostics, Barcelona, Spain), and analyzed by real time RT-PCR for the presence
of West Nile Virus (WNV) and flavivirus genome in general. For WNV detection, a
TaqMan MGB multiplex real time RT-PCR using a commercial kit (QuantiTEC
ProbeW RT-PCR, Qiagen, Madrid, Spain) was applied as described before (JimnezClavero et al., 2006). Flavivirus detection was carried out as described previously by
generic SYBR Green (Qiagen, Madrid, Spain) real time RT-PCR (Moureau et al.,
2007).
A generic nested RT-PCR previously described (Snchez-Seco et al., 2005)
was used to confirm flavivirus detection. Reactions were performed using the Access

172

Captulo 4

RT-PCR System (Promega, Madison, WI, USA) in an automated DNA thermal cycler
model TC-512 (Techne, Cambridge, UK). Briefly, 5 L (around 10 g) of nucleic acid
preparation was added to 45 L of a RT-PCR mix containing 2 mM MgSO4, 0.2 mM
of dNTP, 1 AMV/Tfl 5 reaction buffer, 5 U Tfl DNA polymerase, 5 U AMV reverse
transcriptase and 40 pmols of each primer (Flavi1+ and Flavi1-) and amplified using
an initial incubation at 38 C for 45 min followed by 94 C for 2 min, and 40 cycles
of 94 C for 30 sec, 47 C for 1 min, and 68 C for 1 min and 15 sec. The nested
PCR reaction was carried out in a final volume of 50 L, with similar concentrations
as in the first reaction, using primers Flavi2+ and Flavi2-, and 1 L of the product
of the first amplification. The mix was subjected to 94 C for 2 min, followed by 40
PCR cycles with similar conditions to those used in the primary generic RT-PCR.
Subsequently, 8 L of each PCR product was subjected to electrophoresis on
a 2% agarose gel to check the size of amplified fragments by comparison to a DNA
molecular weight marker (1 kb Plus DNA Ladder, Promega, Madison, WI, USA).
The DNA bands from the nested and the first generic amplification were
resin-purified (Wizard, Promega Madison, WI, USA) and cloned into pGEM-T
(Promega, Madison, WI, USA). At least four independent clones were sequenced
from both ends for each positive sample (Secugen SL, Madrid, Spain). Sequence
similarity search was performed using BLAST (BLAST).

Histopathology and immunohistochemistry


Formalin-fixed tissues were trimmed, embedded in paraffin, sectioned at 4 m and
processed to obtain haematoxylin-eosin stained sections. These were examined
independently by two different investigators (VG and UH).
Furthermore, the avidin-biotin-peroxidase complex (ABC) method was used
on paraffin-embedded tissue sections for immunohistochemical demonstration of
flavivirus antigen. For this purpose, a rabbit polyclonal antibody against WNV that
has been shown to cross-react with other flaviviruses (Steele et al., 2000), was used
173

Virginia Gamino Rodrguez

Tesis doctoral

(BioReliance, Product 81015, Rockville, Maryland, USA). After deparaffinization


and hydration of the sections, they were incubated with 3% H2O2 diluted in
methanol for 30 min at room temperature (2025C) in order to block endogenous
peroxidase activity. After rinsing with water, the sections were treated with
proteinase K (Diagnostic BioSystems, Pleasanton, California) for 15 min at room
temperature. They were then rinsed with water, followed by three 5 min rinses in
0.1% Tris-buffered saline/Tween20 (TBS 0.05 M, pH 7.5), and incubated 1 hr at
room temperature with 2% albumin from bovine serum (Sigma-Aldrich Chemie,
Steinheim, Germany) diluted in 0.1% TBS/Tween20. The primary antibody was
applied overnight at 4C at a dilution of 1:1000 in 2% albumin-0.1% TBS/Tween20.
After three 5 min rinses in 0.1% TBS/Tween20, sections were incubated with a
biotinylated goat anti-rabbit IgG (Vector Laboratories, Burlingame, California, USA)
diluted 1:200 in 0.1% TBS/Tween20 for 1 hr at room temperature. After three 5
min rinses in 0.1% TBS/Tween20 an avidin-biotinylated enzyme complex (ABC
system, Vector laboratories) was applied for 30 min at room temperature according
to manufacturer's recommendations. For development, sections were incubated for
1 min with 3,3-diaminobenzidine tetrahydrochloride according to manufacturer's
recommendations (Vector Laboratories). Sections were counterstained with Mayer's
haematoxylin. Negative controls included substitution of the primary antibody by
2% albumin from bovine serum diluted in 0.1% TBS/Tween20 and negative rabbit
antibody (BioReliance, Product 81015). Tissue sections of experimentally infected
red-legged partridges in which presence of WNV had been confirmed by real time
RT-PCR served as positive controls.

174

Captulo 4

RESULTS
Morbidity and mortality
The outbreak affected mostly gallinaceous game birds and caused nervous signs
and high mortality. Apparently the primary species affected by the outbreak were
red-legged partridges and ring-necked pheasants. Carcasses and individuals
displaying clinical signs of apparent blindness, ataxia and lack of coordination
belonged primarily to these two species, although a low number of affected
Columbiformes (common wood pigeon) had also been observed. Mortality or nervous
signs in other avian species such as corvids or birds of prey were not detected.

Real time RT-PCR and sequence analysis


All samples tested negative for genomes of WNV lineage 1 and 2 by real time RTPCR. Flavivirus genome was detected by real time RT-PCR in all tested individuals
in at least one of the samples analyzed (swabs and tissues, Table 1). A positive
signal was obtained most frequently in the brain (67%) and oral swabs (67%) of
partridges, and in the kidney (80%), and brain (100%) of pheasants. In one of the
wood pigeons sampled, all tested tissues were positive for flavivirus, while only the
kidney of the other one was positive and oral and cloacal swabs were negative in
both individuals (Table 1).
Only one partridge brain sample yielded a DNA product sufficient for cloning
and sequencing in the primary conventional generic nested flavivirus RT-PCR. In
the sequence similarity search performed by using BLAST, the analyzed sequence
of 1048 bp showed 92% sequence identity to previously reported BAGV strain
DakAr B209 [GenBank: AY632545.2]. In the case of the nested PCR, the 104 bp
amplicon was sequenced from 8 different samples; 3 brain samples and 2 oral
swabs from partridges, and 3 brain samples from pheasants, showing 97-98%
sequence identity to the previously described BAGV (Table 1).
175

Virginia Gamino Rodrguez

Tesis doctoral

Table 1. Detection of flavivirus genome in BAGV infected game birds. Detection of flavivirus
genome by rt RT-PCR (Ct values) and by sequencing from conventional PCR in different tissues and
swabs.
Partridge

Wood
pigeon

Pheasant

Sample
No.1

No.2

No.3

No.4

No.5

No.6

No.1

No.2

No.3

No.4

No.5

No.1

No.2

19

30

23*

19*

29

31

Clocacal swab

29

31

Brain

22*

22*

16*

30

26*

23*

24*

30

30

30

Heart

32

23

23

27

31

Spleen

30

30

27

Kidney

29

30

28

29

27

29

29

30

Oral swab

*Sequenced 104 bp amplicon from conventional PCR.


Sequenced 1048 bp amplicon from conventional PCR.
Samples that did not show amplification.

Gross and microscopic lesions


The distribution, nature and severity of lesions varied considerably between species.
Two partridges and two pheasants were emaciated. The most striking macroscopic
lesions in partridges included pallor of the pectoral muscle (4/6), pancreas (4/6)
and myocardium (3/6) and injection of encephalic (3/6) and epicardial (4/6)
vessels. Besides thickening of the pericardium (2/5) or pallor of the myocardium
(2/5), no significant gross lesions were observed in pheasants. Equally, no
significant macroscopic lesions other than encephalic vessel injection (2/2) were
found in wood pigeons.
In all cases, the most prominent microscopic lesions were congestion,
necrosis and mononuclear inflammatory infiltrates consisting of lymphoid cells,
plasma cells and histiocytes, although to a different degree in individuals of the
three species (Table 2).

176

Captulo 4

The most affected systems were the central nervous system and the spleen.
Lesions in the brain were characterized by congestion, gliosis, neuronal necrosis
and phagocytosis, perivascular cuffing, capillary endothelial cell swelling and
Purkinje cell necrosis and disappearance. The perivascular infiltrates and glial
nodules were constituted by lymphocytes, plasma cells and histiocytes and were
mainly present in the gray matter of the cerebrum, the brain stem and molecular
layer of the cerebellum (Table 2 and figures 1-4). In the spleen, lymphoid cell
depletion, necrotic foci of lymphoid cells, thickened capsule, and multifocal
granulocytic infiltrates were evident. Other organs affected included the kidney,
heart and liver, where necrosis and lymphoplasmacytic and histiocytic infiltrates
were the most important lesions (Table 2 and figures 5 and 6).
While partridges showed severe haemosiderosis in the spleen and liver, this
was absent in pheasants and less evident in wood pigeons (Table 2 and figures 7
and 8). All examined partridges and, to a lesser extent wood pigeons, had Kupffer
cells and hepatocytes in the liver and macrophages in the spleen heavily laden with
brown pigment. Using Perls' stain, this brown pigment was shown to contain iron,
indicating that in fact, it corresponds to haemosiderin (Figures 9 and 10). In
pheasants no iron/haemosiderin was evidenced in the liver or spleen using this
technique.

177

Virginia Gamino Rodrguez

Tesis doctoral

Table 2. Microscopic lesions in BAGV infected game birds.


Table 2. Microscopic lesions in BAGV infected game birds.
Partridge

Lesion

Wood
pigeon

Pheasant

No.1

No.2

No.3

No.4

No.5

No.6

No.1

No.2

No.3

No.4

No.5

No.1

No.2

Neuronal necrosis

NA

Neuronophagia

NA

Satellitosis

NA

Gliosis

NA

Glial necrosis

NA

Perivascular cuffing

NA

Endothelial cell swelling

NA

Purkinje cell necrosis

NA

Purkinje cell disappearance

NA

Gliosis

NA

Perivascular cuffing

NA

Endothelial cell swelling

NA

Neuronal necrosis

NA

NA

Gliosis

NA

NA

Cerebrum

Cerebellum

Optic lobe

Glial necrosis

NA

NA

Perivascular cuffing

NA

NA

Endothelial cell swelling

NA

NA

Miofibrilar necrosis
degeneration

NA

Inflammatory infiltrate

NA

Septal epithelial cells


necrosis

Inflammatory infiltrate

Capsular thickening

NA

NA

Lymphoid cell necrosis

NA

NA

Lymphoid cell depletion

NA

NA

Haemosiderosis

NA

NA

Granulocytic infiltration

NA

NA

Vascular wall thickening

NA

NA

Endothelial cell swelling

NA

NA

Hepatocellular necrosis

Inflammatory infiltrate

Haemosiderosis

Tubular epith. cell degen.

Inflammatory infiltrate

Heart

Lung

Spleen

Liver

Kidney

178

Captulo 4

Table 2. Microscopic lesions in BAGV infected game birds (cont).


Partridge

Lesion

Wood
pigeon

Pheasant

No.1

No.2

No.3

No.4

No.5

No.6

No.1

No.2

No.3

No.4

No.5

No.1

No.2

NA

NA

NA

NA

NA

NA

NA

NA

NA

Large intestine
Epithelial cell necrosis
Pancreas
Acinar cell necrosis
Skin
Inflammatory infiltrate

+ Presence of lesion.
Absence of lesion.
NA: tissue sample no analyzed.

Figure 1. Cerebrum; ring-necked pheasant. Diffuse necrosis of neurons and multifocal perivascular
infiltration of lymphoplasmacytic cells. HE, 100.
Figure 2. Cerebellum; ring-necked pheasant. Necrosis and disappearance of Purkinje cells, and mild
infiltration of lymphoplasmacytic and histiocytic cells in the molecular layer. HE, 200.
Figure 3. Cerebrum; red-legged partridge. Diffuse necrosis of neurons and diffuse and moderate
infiltration of lymphoplasmacytic and histiocytic cells. Necrotic neurons show picnotic nuclei and
retracted cytoplasm. HE, 200.
Figure 4. Cerebellum; red-legged partridge. Disappearance of Purkinje cells, which are replaced by
glial cells (arrowhead), and endothelial cell swelling in the white matter. HE, 100. Inset: detail of the
endothelial cell swelling. HE, x400.

179

Virginia Gamino Rodrguez

Tesis doctoral

Figure 5. Heart; red-legged partridge. Multifocal and moderate necrosis of cardiac myofibers and
infiltration of lymphoplasmacytic and histiocytic cells. Necrotic myofibers show fragmentation and
accumulation of hyaline material in the cytoplasm. HE, 100.
Figure 6. Kidney; red-legged partridge. Multifocal and moderate interstitial infiltration of
lymphoplasmacytic and histiocytic cells. HE, 100.
Figures 7. Spleen; red-legged partridge. Diffuse brown pigment in the cytoplasm of splenic
macrophages. HE 100.
Figure 8. Liver; red-legged partridge. Multifocal brown pigment in the cytoplasm of hepatocytes and
Kupffer cells. HE, 100.
Figure 9. Spleen; red-legged partridge. Blue staining in the cytoplasm of splenic macrophages. Perls'
stain, 100.
Figure 10. Liver; red-legged partridge. Blue staining in the cytoplasm of hepatocytes and Kupffer
cells. Perls' stain, 100

180

Captulo 4

Using immunohistochemistry, BAGV antigen was detected in numerous


organs of the partridges, especially in capillary endothelial cells, while in the wood
pigeon it was only detected in the spleen and in the pheasants in the central

Percentage of individuals with positive


immunostaining

nervous system (Figure 11).


n=6

100
90

n=6

n=6

80
70
60
50
40

n=5
n=2

n=6

30
20

Red-legged partridge

n=6

n=6
n=6

n=5

Ring-necked pheasant

n=5

Common wood pigeon

10
0

Tissue
Figure 11. Distribution of positive flavivirus (presumptive BAGV) immunostaining in naturally
BAGV infected red-legged partridges, ring-necked pheasants and common wood pigeons. Each
column represents the percentage of individuals that showed positive immunostaining in each organ
and n is the number of individuals in which each tissue was tested.

In pheasants, only the cytoplasm of neurons and glial cells of the thalamus
and optic lobe, and Purkinje cells of the cerebellum were found to contain BAGV
antigen (Figures 12 and 13). In partridges, endothelial cells of capillaries of the
cerebrum, cerebellum, spleen, heart, kidney, pectoral muscle, caeca and adrenal
gland contained BAGV antigen in their cytoplasm. Other cells that tested positive
were neurons and glial cells of the cerebrum, Purkinje cells of the cerebellum,
cardiomyocytes of the heart and endothelial cells of the glomerular mesangium in
the kidney (Figures 14-17). One of the wood pigeons had BAGV antigen in capillary
endothelial cells in the spleen. As swabs and tissues tested by real time RT-PCR
were negative for WNV, and the only flavivirus identified was BAGV, we assumed

181

Virginia Gamino Rodrguez

Tesis doctoral

that positive labeling in immunohistochemistry preparations indicated presence of


BAGV antigen.

Figure 12. Cerebrum; ring-necked pheasant. BAGV antigen in the cytoplasm of neurons. IHC using
a cross-reactive WNV antibody with Mayer's haematoxylin counterstain, x100.
Figure 13. Cerebellum; ring-necked pheasant. BAGV labeling in the cytoplasm of Purkinje cells. IHC
using a cross-reactive WNV antibody with Mayer's haematoxylin counterstain, x200. Inset: detail of
the immunostaining. IHC using a cross-reactive WNV antibody with Mayer's haematoxylin
counterstain, x400.
Figure 14. Cerebrum; red-legged partridge. BAGV labeling in the cytoplasm of neurons and capillary
endothelial cells. IHC using a cross-reactive WNV antibody with Mayer's haematoxylin counterstain,
x200.

182

Captulo 4

Figure 15. Cerebellum; red-legged partridge. BAGV antigen in capillary endothelial cells and in the
cytoplasm of Purkinje cells. IHC using a cross-reactive WNV antibody with Mayer's haematoxylin
counterstain, x100. Inset: detail of the immunostaining. IHC using a cross-reactive WNV antibody with
Mayer's haematoxylin counterstain, x400.
Figure 16. Heart; red-legged partridge. BAGV antigen in capillary endothelial cells. IHC using a
cross-reactive WNV antibody with Mayer's haematoxylin counterstain, x400.
Figure 17. Kidney; red-legged partridge. BAGV antigen in capillary endothelial cells of the
glomerular mesangium. IHC using a cross-reactive WNV antibody with Mayer's haematoxylin
counterstain, x400.

Incidental findings included avian tuberculosis, anthracosis and coccidian


oocysts. Two of the pheasants and one of the wood pigeons had lesions compatible
with avian tuberculosis, in which presence of acid resistant bacilli was confirmed by
Ziehl-Neelsen stain. All of the partridges and one pheasant had brown pigment and
crystalline structures within the cytoplasm of peribronchial macrophages in the
lungs, compatible with anthracosis. Finally, two of the partridges showed less than
five coccidian oocysts/cross sections in the mucosal epithelium of the large
intestine.

DISCUSSION
The BAGV outbreak in 2010 in Spain is the first documented occasion in which this
virus has caused disease and mortality in birds, with a strikingly most severe effect
on a game bird species, the red-legged partridge (Agero et al., 2011). Recently,
authors reporting on the parallel outbreak of WNV in horses in the same region
made the hot summer and high mosquito abundance responsible for both
outbreaks (Garca-Bocanegra et al., 2011). While several cases of WN fever in horses
were notified, no new mortality events among game birds were detected during
2011.
Pathology due to BAGV infection had not been previously described in any
species, as the presence of neutralizing antibodies against BAGV in persons with
acute encephalitis in India, could not link the infection clearly to disease symptoms
(Bondre et al., 2009). However, based on sequence analysis, BAGV has been shown
183

Virginia Gamino Rodrguez

Tesis doctoral

to be synonymous to Israel turkey meningoencephalitis virus (ITMV) (Kuno et al.,


1998), which causes a disease characterized by nonpurulent meningoencephalitis
with lymphocytic perivascular infiltrates and focal myocardial necrosis in turkeys in
Israel and South Africa, and is controlled by vaccination with live attenuated
vaccines (Komarov and Kalmar, 1960; Ianconescu et al., 1975; Barnard et al.,
1980). The close genetic relationship between the two viruses may mean that BAGV
is similarly pathogenic to at least some bird species (Garca-Bocanegra et al., 2012).
A local hunting estate in the area of the outbreak that conducts direct
transect counts on game birds prior to and after the hunting season, reported
reduction of red-legged partridge numbers by 86% and in ring-necked pheasants by
29%. Due to the high mortality in red-legged partridges, only ring-necked pheasants
were hunted in winter 2010/2011. Hunting bag data from the same hunting estate,
showed a reduction in the female/male ratio with respect to previous years (2.4
females/male as opposed to 4 females/male), suggesting that female pheasants
were more affected than males.
The degree to which the impact of BAGV in red-legged partridge populations
is higher than in ring-necked pheasants and other birds is evidenced by differences
in mortality and in the incidence of concomitant disease observed. In all examined
partridges BAGV infection appeared to be the primary cause of death (2/6) or
disease (4/6), while two of five pheasants and one of the two wood pigeons had
severe advanced lesions of concurrent chronic disease (avian tuberculosis). The
reason of the higher reduction in numbers in female pheasants, as opposed to
males is unclear. With view to a potential higher impact of BAGV, disease due to
ITMV has also been reported to be more severe and frequent in female than in male
turkeys (Barnard et al., 1980). However, other effects such as higher susceptibility
of female pheasants to predation, other diseases, or toxic substances employed in
agriculture in the area cannot be ruled out.

184

Captulo 4

Differences in the pathogenicity of BAGV for the three species could partly be
explained by the differences observed in the distribution and severity of lesions,
distribution of viral antigen, and the severe haemosiderosis in partridges, that was
moderate in wood pigeons and absent in pheasants. In partridges, BAGV apparently
has a wide tropism, targeting different cell types, but especially capillary endothelial
cells. In pheasants, neurotropism appears to be somewhat more important while in
pigeons only endothelial cells of splenic capillaries seemed to contain BAGV
antigen.
One of the most well studied flaviviruses that is known to cause disease in
birds is WNV, of which information is available on pathology in naturally infected
humans, horses and birds as well as experimental avian and mouse models
(Sampson et al., 2000; Steele et al., 2000; Cantile et al., 2001; Samuel and
Diamond, 2006). WNV is known to vary greatly in its virulence in avian species
although the mechanism and reasons are still poorly understood (Steele et al.,
2000; Wnschmann et al., 2005; Brault, 2009). As an example, both endothelial
and neural tropism has been described in native North American avian species after
natural WNV infection (Wnschmann et al., 2004a,b; Lopes et al., 2007). Also, in
humans and mouse models WNV has been shown to have a diverse cell tropism,
leading to a variety of lesions and clinical manifestations (Hayes et al., 2005;
Samuel and Diamond, 2006).
The severe haemosiderosis in red-legged partridges that is the most striking
difference to lesions due to BAGV in ring-necked pheasants has been described
previously in birds naturally infected by WNV. Hepatic haemosiderosis is most well
known as associated to iron overload in captive wild forest birds but is also
frequently associated to haemolytic processes in infectious disease (Cork et al.,
1995). Haemosiderosis and haemorrhage in relation to WNV has been described
previously

in

the

spleen and

liver of naturally

infected

North American
185

Virginia Gamino Rodrguez

Tesis doctoral

Passeriformes such as the blue jay (Cyanocitta cristata) and the house sparrow
(Passer domesticus) as well as raptors and owls from the North American continent
(Gibbs et al., 2005; Ellis et al., 2007; O'Brien et al., 2010). It has also been
described in wild turkeys (Meleagris gallopavo) (Zhang et al., 2006), but in an
outbreak in farmed chukar partridges (Alectoris chukar) and Impeyan pheasants
(Lophophorus impeyanus), only erythrocytophagocytosis was reported in the spleen,
while haemosiderosis was not observed (Wnschmann and Ziegler, 2006). With view
to WNV-associated haemolysis in other species, a few cases of WNV-associated
haemorrhagic fever in humans have been described (Paddock et al., 2006). A recent
study has associated sequence signatures in the envelope protein of human
pathogenic flaviviruses with the primary syndrome that they produce (encephalitis
or haemorrhagic disease) (Barker et al., 2009). These authors speculated that the
electrostatic charge differences caused by the presence of either glycosilated
asparagine (Asn, haemorrhagic viruses) or Aspartic acid (Asp, encephalitic viruses)
at position 67 of the domain II of the envelope protein could be responsible for the
phenotype and related disease syndrome. It would be of interest to further
characterize the outbreak-related virus, in order to study these and other features.
In conclusion, BAGV is more pathogenic for red-legged partridges than for
ring-necked pheasants and common wood pigeons, and causes a severe haemolytic
process in this species. Further (experimental) studies will be necessary to
determine the factors that trigger BAGV susceptibility and pathogenesis of the
infection in red-legged partridges.

ACKNOWLEDGEMENTS
This study was supported by grant AG2008-02504GAN funded by the Spanish
Ministry for Science and Innovation. A.V. Gutirrez-Guzmn (PAC08-0296-7771)
and V. Gamino (323/09) are research fellows supported by the regional government
186

Captulo 4

of Castilla La Mancha (JCCM) and I.G. Fernndez-de-Mera was funded by MCINN


Program Juan de la Cierva, JCI-2009-04518, Spain. We acknowledge the help of
game wardens with sample and data collection in the field, and the assistance of M.
Pizarro with the photography of microscopic lesions.

REFERENCES
Agero M, Fernndez-Pinero J, Buitrago D, Snchez A, Elizalde M, San Miguel E,
Villalba R, Llorente F, Jimnez-Clavero MA: Bagaza virus in partridges and
pheasants, Spain, 2010. Emerg Infect Dis 2011, 17(8):1498-1501.
Barker WC, Mazumder R, Vasudevan S, Sagripanti J-L, Wu CH: Sequence
signatures in envelope protein may determine whether flaviviruses
produce hemorrhagic or encephalitic syndromes. Virus Genes 2009, 39(1):19.
Barnard BJ, Buys SB, Du Preez JH, Greyling SP, Venter HJ: Turkey meningoencephalitis in South Africa. Onderstepoort J Vet Res 1980, 47(2):89-94.
Bondre VP, Sapkal GN, Yergolkar PN, Fulmali PV, Sankararaman V, Ayachit VM,
Mishra AC, Gore MM: Genetic characterization of Bagaza virus (BAGV)
isolated in India and evidence of anti-BAGV antibodies in sera collected
from encephalitis patients. J Gen Virol 2009, 90(Pt 11):2644-2649.
Brault AC: Changing patterns of West Nile virus transmission: altered vector
competence and host susceptibility. Vet Res 2009, 40(2):43. doi:
10.1051/vetres/2009026.
Basic Local Alignment Search Tool [http://blast.ncbi.nlm.nih.gov/Blast.cgi].
Cantile C, Del Piero F, Di Guardo G, Arispici M: Pathologic and
immunohistochemical findings in naturally occuring West Nile virus
infection in horses. Vet Pathol 2001, 38(4):414-421.
Cork SC, Alley MR, Stockdale PH: A quantitative assessment of haemosiderosis
in wild and captive birds using image analysis. Avian Pathol 1995, 24(2):239254.
Digoutte JP: Bagaza (BAG) strain: Dak Ar B 209. Am J Trop Med Hyg 1978,
27:376-377.
Ellis AE, Mead DG, Allison AB, Stallknecht DE, Howerth EW: Pathology and
epidemiology of natural West Nile viral infection of raptors in Georgia. J
Wildl Dis 2007, 43(2):214-223.
Garca-Bocanegra I, Jan-Tllez JA, Napp S, Arenas-Montes A, Fernndez-Morente
M, Fernndez-Molera V, Arenas A: West Nile fever outbreak in horses and
humans, Spain, 2010. Emerg Infect Dis 2011, 17(12):2397-2399.

187

Virginia Gamino Rodrguez

Tesis doctoral

Garca-Bocanegra I, Zorrilla I, Rodrguez E, Rayas E, Camacho L, Redondo I,


Gmez-Guillamn F: Monitoring of the Bagaza Virus Epidemic in Wild Bird
Species in Spain, 2010. Transbound Emerg Dis 2012, 60(2):120-126.
Gibbs SEJ, Hoffman DM, Stark LM, Marlenee NL, Blitvich BJ, Beaty BJ,
Stallknecht DE: Persistence of antibodies to West Nile virus in naturally
infected rock pigeons (Columba livia). Clin Diagn Lab Immunol 2005,
12(5):665-667.
Hayes EB, Sejvar JJ, Zaki SR, Lanciotti RS, Bode AV, Campbell GL: Virology,
pathology, and clinical manifestations of West Nile virus disease. Emerg
Infect Dis 2005, 11(8):1174-1179.
Ianconescu M, Hornstein K, Samberg Y, Aharonovici A, Merdinger M: Development
of a new vaccine against turkey meningo-encephalitis using a virus
passaged through the Japanese quail (Coturnix coturnix japonica). Avian
Pathol 1975, 4(2):119-131.
Jimnez-Clavero MA, Aguero M, Rojo G, Gomez-Tejedor C: A new fluorogenic realtime RT-PCR assay for detection of lineage 1 and lineage 2 West Nile
viruses. J Vet Diagn Invest 2006, 18(5):459-462.
Komarov A, Kalmar E: A hitherto undescribed disease-turkey meningoencephalitis. Vet Rec 1960, 72:257-261.
Kuno G, Chang GJ, Tsuchiya KR, Karabatsos N, Cropp CB: Phylogeny of the
genus Flavivirus. J Virol 1998, 72(1):73-83.
Lopes H, Redig P, Glaser A, Armien A, Wunschmann A: Clinical findings, lesions,
and viral antigen distribution in great gray owls (Strix nebulosa) and
barred owls (Strix varia) with spontaneous West Nile virus infection. Avian
Dis 2007, 51(1):140-145.
Moureau G, Temmam S, Gonzalez JP, Charrel RN, Grard G, de Lamballerie X: A
real-time RT-PCR method for the universal detection and identification of
flaviviruses. Vector Borne Zoonotic Dis 2007, 7(4):467-477.
Munster VJ, Baas C, Lexmond P, Waldenstrom J, Wallensten A, Fransson T,
Rimmelzwaan GF, Beyer WE, Schutten M, Olsen B, Osterhaus AD, Fouchier
RA: Spatial, temporal, and species variation in prevalence of influenza A
viruses in wild migratory birds. PLoS Pathog 2007, 3(5):e61.
O'Brien VA, Meteyer CU, Reisen WK, Ip HS, Brown CR: Prevalence and pathology
of West Nile virus in naturally infected house sparrows, western Nebraska,
2008. Am J Trop Med Hyg 2010, 82(5):937-944.
Paddock CD, Nicholson WL, Bhatnagar J, Goldsmith CS, Greer PW, Hayes EB,
Risko JA, Henderson C, Blackmore CG, Lanciotti RS, Campbell GL, Zaki SR:
Fatal hemorrhagic fever caused by West Nile virus in the United States.
Clin Infect Dis 2006, 42(11):1527-1535.
Sampson BA, Ambrosi C, Charlot A, Reiber K, Veress JF, Armbrustmacher V: The
pathology of human West Nile Virus infection. Hum Pathol 2000, 31(5):527531.
188

Captulo 4

Samuel MA, Diamond MS: Pathogenesis of West Nile Virus infection: a balance
between virulence, innate and adaptive immunity, and viral evasion. J Virol
2006, 80(19):9349-9360.
Snchez-Seco MP, Rosario D, Domingo C, Hernandez L, Valdes K, Guzman MG,
Tenorio A: Generic RT-nested-PCR for detection of flaviviruses using
degenerated primers and internal control followed by sequencing for
specific identification. J Virol Methods 2005, 126(1-2):101-109.
Steele KE, Linn MJ, Schoepp RJ, Komar N, Geisbert TW, Manduca RM, Calle PP,
Raphael BL, Clippinger TL, Larsen T, Smith J, Lanciotti RS, Panella NA,
McNamara TS: Pathology of fatal West Nile virus infections in native and
exotic birds during the 1999 outbreak in New York City, New York. Vet
Pathol 2000, 37(3):208-224.
Wnschmann A, Shivers J, Bender J, Carroll L, Fuller S, Saggese M, van Wettere A,
Redig P: Pathologic findings in red-tailed hawks (Buteo jamaicensis) and
Cooper's hawks (Accipiter cooper) naturally infected with West Nile virus.
Avian Dis 2004a, 48(3):570-580.
Wnschmann A, Shivers J, Carroll L, Bender J: Pathological and
immunohistochemical
findings
in
American
crows
(Corvus
brachyrhynchos) naturally infected with West Nile virus. J Vet Diagn Invest
2004b, 16(4):329-333.
Wnschmann A, Shivers J, Bender J, Carroll L, Fuller S, Saggese M, van Wettere A,
Redig P: Pathologic and immunohistochemical findings in goshawks
(Accipiter gentilis) and great horned owls (Bubo virginianus) naturally
infected with West Nile virus. Avian Dis 2005, 49(2):252-259.
Wnschmann A, Ziegler A: West Nile virus-associated mortality events in
domestic chukar partridges (Alectoris chukar) and domestic Impeyan
pheasants (Lophophorus impeyanus). Avian Dis 2006, 50(3):456-459.
Zhang Z, Wilson F, Read R, Pace L, Zhang S: Detection and characterization of
naturally acquired West Nile virus infection in a female wild turkey. J Vet
Diag Invest 2006, 18(2):204-208.

189

Captulo 5
Hallazgos patolgicos en ojos de
gallinceas infectadas por flavivirus

aves

Gamino V, Escribano-Romero E, Gutirrez-Guzmn AV, Blzquez AB,


Saiz JC, Hfle U. Oculopathologic findings in flavivirus infected
gallinaceous birds. Veterinary Pathology. En revisin.

Captulo 5

RESUMEN
En el presente trabajo estudiamos el curso de aparicin de lesiones y antgeno viral
en las estructuras oculares de nueve perdices rojas (Alectoris rufa), de nueve
semanas de edad, infectadas de forma experimental con el virus West Nile (WNV).
De forma adicional, se estudiaron las lesiones y la distribucin del antgeno viral en
los ojos de seis perdices rojas y tres faisanes comunes (Phasianus colchicus)
infectados de forma natural con el virus Bagaza (BAGV), con el fin de explicar la
aparente ceguera observada. La rpida aparicin de lesiones microscpicas y
antgeno viral en el ojo de las perdices infectadas experimentalmente con WNV,
incluso antes que en el sistema nervioso central, fue indicativo de la llegada del
virus al ojo va hematgena. Las perdices infectadas con BAGV presentaron una
mayor reaccin inflamatoria y una distribucin ms amplia del antgeno viral en la
retina en comparacion con los faisanes y las perdices infectadas experimentalmente
con WNV. Esto podra explicar el mayor efecto de la infeccin sobre la visin en las
primeras. Nuestros resultados indican que, en aves gallinceas de caza, la
replicacin de un flavivirus y el desarrollo de lesiones en estructuras oculares
varan en funcin del virus y la especie infectada.

193

Virginia Gamino Rodrguez

Tesis doctoral

ABSTRACT
Using eye samples of nine 9-week-old experimentally West Nile virus (WNV) infected
red-legged partridges (Alectoris rufa), time course of lesions and WNV antigen
appearance in ocular structures was examined. Additionally, eye samples of six redlegged partridges and three ring-necked pheasants (Phasianus colchicus) naturally
infected with Bagaza virus (BAGV) were used to study lesions and flavivirus antigen
distribution in relation to apparent blindness. The rapid onset of microscopic
lesions and early presence of viral antigen in the eye of experimentally WNV infected
partridges,

prior

to

the

central

nervous

system

involvement,

suggested

hematogenous spread of the virus into the eye. BAGV infected partridges had a
more pronunced inflammatory reaction and more widespread flavivirus antigen
distribution in the retina compared to pheasants and experimentally fatally WNV
infected partridges, which could explain the more prominent effect on the vision.
Our results suggest that flavivirus replication and development of lesions in ocular
structures of gallinaceous game birds vary with the specific virus and host species
involved.

194

Captulo 5

INTRODUCTION
Flaviviruses are small single-stranded RNA viruses with a virtually worldwide
distribution. West Nile virus (WNV), a pathogen of importance for birds, has a wide
tissue tropism, and the eye is one of the target organs in infected avian hosts.
Vision impairment and ocular lesions have been described, mainly in raptors
(Wnschmann et al., 2004, 2005; Pauli et al., 2007), but little is known about the
mechanisms by which this virus reaches the eye and produces ocular lesions (Pauli
et al., 2007).
Another flavivirus, Bagaza virus (BAGV), recently caused an outbreak among
game birds in south-western Spain, producing neurologic signs in ring-necked
pheasants (Phasianus colchicus) and, more severely, in red-legged partridges
(Alectoris rufa), which also showed apparent blindness (Gamino et al., 2012).

MATERIAL AND METHODS


For the present study, we collected eye samples of nine 9-week-old red-legged
partridges (Nos. 1 to 9) subcutaneously inoculated with 107 PFU of a cell culture
passaged New York/1999 (NY99) WNV strain, and euthanized on days 3 (No. 2), 7
(No. 4), 10 (Nos. 6 and 7) and 14 (Nos. 8 and 9) post-inoculation (dpi), or found
dead during the experiment presumably due to WNV infection (Nos. 1, 3 and 5).
These samples were examined in order to follow the appearance and distribution of
lesions and antigen during the course of the infection.
Moreover, we collected eye samples of six sexually mature red-legged
partridges (Nos. 10 to 15) and three ring-necked pheasants (Nos. 16 to 18) that died
or were euthanized in a moribund state during the recent BAGV outbreak in Spain
(Gamino et al., 2012). These samples were examined in order to compare
microscopic lesions and flavivirus antigen distribution between the two avian

195

Virginia Gamino Rodrguez

Tesis doctoral

species, with the aim of amplifying our recent description of species specific tissue
tropism of this virus and investigating blindness observed in the affected partridges.
Hematoxylin

and

eosin

stain

(HE)

and

viral

antigen

detection

by

immunohistochemistry (IHC) were performed. For the IHC, a rabbit polyclonal


antibody against WNV that has been shown to cross-react with other flaviviruses
(BioReliance, Product 81015, Rockville, MD), was used applying the protocol
described previously (Gamino et al., 2012), but using 3-Amino-9-ethylcarbazole
(AEC) as visualizing method. These sections were examined under a light
microscope by two different investigators (VG and UH). For comparative purposes
between species (BAGV) or among different dpi (WNV), the distribution and
abundance of the viral antigen were subjectively scored as absent, mild (focal or
multifocal, <10% cells stained), moderate (multifocal, 10-50% cells stained) or
marked (diffuse, >50% cells stained).

RESULTS
WNV and BAGV infection was confirmed in all experimentally and naturally infected
birds by real time RT-PCR and sequencing as previously described (Crdoba et al.,
2007; Gamino et al., 2012)
Unlike BAGV infected partridges, none of the experimentally WNV infected
birds showed visual impairment.
Experimentally

WNV

infected

partridges

that

were

euthanized

had

intravascular lymphoplasmacytic, histiocytic and, less frequently, granulocytic


inflammatory cells in the choroid, pecten and ciliary body on various dpi (Table 1).
Mild inflammation in the connective tissue of the sclera (7 dpi, No. 4) and ciliary
body (7 and 14 dpi, Nos. 4 and 9, respectively) and endothelial cell swelling in the
choroid (10 dpi, No. 7), periocular muscle and pecten (14 dpi, Nos. 8 and 9) were
also detected (Table 1 and figure 1). Compared to euthanized birds, fatally WNV
196

Captulo 5

infected

partridges

showed

mononuclear

inflammatory

cells

and

muscular

degeneration in the iris on days 2 and 7 pi (Nos. 1 and 3, respectively) and mild
optic neuritis on 7 dpi (No. 3) (Table 1). In both euthanized and fatally infected
partridges, WNV antigen was only detected in the rods and cones processes. The
immunostaining was mild and appeared earlier in euthanized birds (Table 1 and
figure 2).
Table 1. Lesion and antigen distribution in the eyes of experimentally WNV infected red-legged
partridges, collected between days 2-14 post-inoculation (dpi).
2 dpi
(No. 1)
Tissue

3 dpi
(No. 2)*

7 dpi
(No. 3)

7 dpi
(No. 4)*

8 dpi
(No. 5)

10 dpi
(Nos.
6 and 7)*

14 dpi
(Nos.
8 and 9)*

HE

IHC

HE

IHC

HE

IHC

HE

IHC

HE

IHC

HE

IHC

HE

IHC

Periocular muscle

+d

+d

+d

Conjunctiva

NA

NA

NA

NA

+a,b

+a,b

Ciliary body

+a,c

+a,c

+c

+a

Choroid

+c

+a,c

+c,d

+d

+c

Pecten

+c

+c

+c,d

+c,d

Optic nerve

+a

NA

NA

Retina

++

Iris

Partridges found dead.


* Euthanized partridges.
Lesion evaluation in HE stained sections: () lesion absence; (+) lesion presence:(a) inflammatory cell infiltration;
(b) myofiber degeneration; (c) intravascular inflammatory cells; (d) endothelial cell swelling.
Immunostaining scores: () negative; (+) mild; (++) moderate; (+++) marked.
NA: no analyzed.

In BAGV infected partridges, the presence of mononuclear or mixed


intravascular and infiltrating inflammatory cells was marked. These were widely
distributed in every ocular structure, including the optic nerve. Additionally,
myofibril degeneration (Nos. 11-14) and endothelial cell swelling (Nos. 13 and 14)
were detected, but only in the periocular muscle. In pheasants, inflammatory

197

Virginia Gamino Rodrguez

Tesis doctoral

infiltrates were mild, with the exception of the pecten and the optic nerve (Figures 3
and 4), and absent in the periocular muscle. Endothelial cell swelling was observed
in the choroid and pecten of case No. 16. In both species, BAGV antigen was
detected in different cell layers of the retina, scarcer in pheasants, with differences
in staining distribution and abundance (Figures 5-7).

198

Captulo 5

Figure 1. Pecten; experimentally WNV infected partridge No. 8, 14 dpi. Nuclear swelling in
capillary endothelial cells. HE, x400.
Figure 2. Retina; experimentally WNV infected partridge No. 6, 10 dpi. WNV antigen in
photoreceptor processes. IHC for WNV with Mayer's hematoxylin counterstain, x400.
Figure 3. Pecten; naturally BAGV infected pheasant No. 16. Diffuse and marked infiltration of
lymphoplasmacytic, histiocytic and granulocytic cells which is expanding the pecten. HE, x40. Inset:
detail of the presence of numerous intravascular and perivascular inflammatory cells. HE, x400.
Figure 4. Optic nerve; naturally BAGV infected pheasant No. 16. Diffuse and moderate gliosis. HE,
x400.
Figure 5. Retina; naturally BAGV infected partridge No. 13. BAGV antigen in photoreceptor
processes and within vertical cell processes in the inner nuclear layer. IHC using cross-reactive WNV
antibody with Mayer's hematoxylin counterstain, x400.
Figure 6. Retina; naturally BAGV infected partridge No. 15. BAGV antigen in photoreceptor
processes, within cell cytoplasm and vertical cell processes in the inner nuclear layer, and in cell

199

Virginia Gamino Rodrguez

Tesis doctoral

cytoplasm in the ganglion cell layer. IHC using cross-reactive WNV antibody with Mayer's hematoxylin
counterstain, x400.

100%

Pa

Ph

Pa

Ph

Pa

Ph

Pa

Ph

90%

Percentage of individuals

80%
70%

Marked

60%

Moderate
Mild

50%

Absent

40%
30%
20%
10%
0%
Rods and
cones

Outer nuclear

Inner nuclear

Ganglion cell

Retinal layer

Figure 7. Immunostaining scores of BAGV antigen in different layers of the retina of red-legged
partridges (Pa) and ring-necked pheasants (Ph) naturally infected with the virus.

DISCUSSION
In the present study, we observed similar lesions in the eyes of experimentally WNV
infected partridges to those described in naturally infected raptors and owls
(Wnschmann et al., 2004, 2005; Gancz et al., 2006). Ocular infection of
flaviviruses is thought to occur either hematogenously during viremia or by
extension from the central nervous system (CNS) to the retina via the optic nerves
(Pauli et al., 2007). Studying eye samples of the experimentally WNV infected
partridges, microscopic lesions appeared very early in the course of the infection
and, in euthanized animals, WNV antigen was detected as early as 3 dpi.
Considering that WNV RNA, viral antigen and microscopic lesions were detected
later in the CNS of the same birds (7-10 dpi, data not shown), we assumed that
WNV distribution into the eye occurred hematogenously.

200

Captulo 5

In this study, we also found differences in the inflammatory response and


antigen abundance in the eyes of BAGV infected partridges and pheasants. The
mild inflammatory reaction and the scarce BAGV antigen presence in the retina of
pheasants could be related to the apparently lower virulence of this virus in this
species (Agero et al., 2011; Gamino et al., 2012). On the contrary, the higher
abundance and wider distribution of BAGV antigen in the retina of infected
partridges accompanied by a more severe inflammatory reaction may be the cause
of the more prominent effect of BAGV on visual capacity in this species, which
matches clinical observation of apparent blindness that was absent in the
experimental WNV infection. Nevertheless, pathological differences could be also
related to the experimental conditions in the WNV infection, the age of the infected
birds or the virus.
In conclusion, virus replication and development of lesions in ocular
structures of flavivirus infected gallinaceous game birds vary with the virus and
host species involved. Especially, retinal involvement can influence the development
of impaired vision, as evidenced here in the case of BAGV infected red-legged
partridges. Vision impairment could have an important impact on the survival of
individuals in the field that adds to primary mortality from the disease.

ACKNOWLEDGEMENTS
This study was supported by grants AG2008-02504GAN and SAF-2008-04232
funded by the Spanish Ministry for Science and Innovation, grants FAU2008 000600-00 and RTA2011-0036 funded by the Spanish Institute for Agricultural and
Alimentary Investigation and Technology (INIA), and The Network of Animal Disease
Infectiology and Research Facilities, NADIR-UE-228394, funded by the E.U. We are
grateful to F. Ruiz-Fons for assistance with figure 7 and to J. Rodriguez-Ramos for
initial critical discussion of the interpretation of ocular histopathology.
201

Virginia Gamino Rodrguez

Tesis doctoral

REFERENCES
Agero M, Fernndez-Pinero J, Buitrago D, Snchez A, Elizalde M, San Miguel E,
Villalba R, Llorente F, Jimnez-Clavero MA. Bagaza virus in partridges and
pheasants, Spain, 2010. Emerg Infect Dis 2011, 17(8):1498-1501.
Crdoba L, Escribano-Romero E, Garmendia A, Saz JC. Pregnancy increases the
risk of mortality in West Nile virus-infected mice. J Gen Virol 2007, 88:476480.
Gamino V, Gutirrez-Guzmn A-V, Fernndez-de-Mera IG, Ortz J-A, Durn-Martn
M, de la Fuente J, Gortzar C, Hfle U. Natural Bagaza virus infection in
game birds in southern Spain. Vet Res 2012, 43. doi: 10.1186/1297-9716-4365.
Gancz AY, Smith DA, Barker IK, Lindsay R, Hunter B. Pathology and tissue
distribution of West Nile virus in North American owls (family : Strigidae).
Avian Pathol 2006, 35(1):17-29.
Pauli AM, Cruz-Martinez LA, Ponder JB, Redig PT, Glaser AL, Klauss G, Schoster
JV, Wunschmann A. Ophthalmologic and oculopathologic findings in redtailed hawks and Cooper's hawks with naturally acquired West Nile virus
infection. J Am Vet Med Assoc 2007, 231(8):1240-1248.
Wnschmann A, Shivers J, Bender J, Carroll L, Fuller S, Saggese M, van Wettere A,
Redig P. Pathologic findings in red-tailed hawks (Buteo jamaicensis) and
Cooper's hawks (Accipiter cooperi) naturally infected with West Nile virus.
Avian Dis 2004, 48(3):570-580.
Wnschmann A, Shivers J, Bender J, Carroll L, Fuller S, Saggese M, van Wettere A,
Redig P. Pathologic and immunohistochemical findings in goshawks
(Accipiter gentilis) and great horned owls (Bubo virginianus) naturally
infected with West Nile virus. Avian Dis 2005, 49(2):252-259.

202

Captulo 6
Deteccin del virus Usutu
migratorios en Espaa

en

zorzales

Hfle U, Gamino V, Fernndez-de-Mera IG, Mangold AJ, Ortz JA, de la


Fuente J. Usutu Virus in Migratory Song Thrushes, Spain. Emerging
Infectious Diseases 2013, 19(7):1173-1175. doi: 10.3201/eid1907.130199.

Captulo 6

RESUMEN
En noviembre del ao 2012 se detect el virus Usutu (USUV) en zorzales comunes
(Turdus philomelos), afectados por un proceso de encefalitis aguda, que se
encontraban invernando en el sur de Espaa. El carcter no residente de esta
especie en el sur de nuestro pas y la secuencia de la cepa viral detectada
proporcionan evidencias de la introduccin de USUV por aves migratorias
procedentes del norte de Europa y una posible recrudescencia de una infeccin
persistente.

ABSTRACT
We detected Usutu virus (USUV) in migratory wintering song thrushes (Turdus
philomelos) with acute encephalitis in southern Spain in November 2012. The nonresident nature of this species in southern Spain and sequence data provides
evidence of USUV introduction by northern European migrants and a possible
recrudescence of a persistent infection.

205

Virginia Gamino Rodrguez

Tesis doctoral

INTRODUCTION
Usutu virus (USUV), a member of the Japanese Encephalitis antigenic group, was
first detected in 1959 in mosquitoes in South Africa (Woodall, 1964), and emerged
in 1996 in blackbirds (Turdus merula) in Italy (Weissenbck et al., 2013). Recent
cases of USUV infection in asymptomatic human blood donors (Allering et al., 2012)
and severe disease in immunocompromised patients (Pecorari et al., 2009) have
evidenced its zoonotic potential.
Epidemiology and molecular phylogeny of USUV isolated in Italy, Austria,
Hungary, Switzerland and Germany suggest establishment of stable endemic
mosquito bird cycles in Europe (Chvala et al., 2007; Becker et al., 2012). Where
active vector surveillance programs exist, USUV is detected in mosquitoes prior to
bird mortality and human cases. USUV similar to African strains was detected in
Spanish mosquitoes in 2006 and 2009 (Busquets et al., 2008; Vzquez et al., 2011).

THE STUDY
In November 2012, two live song thrushes (Turdus philomelos) with neurological
signs were recovered from a mortality of approximately ten birds in a hunting estate
in southern Spain. After death, a full detailed necropsy was conducted and samples
were collected for virus detection and histopathology. Total RNA was extracted from
oral and cloacal swabs, serum from a cardiac blood clot, and heart, kidney, spleen
and brain samples using High Pure RNA Tissue Kit (Roche Diagnostics, Barcelona,
Spain) and analyzed by generic flavivirus SYBR Green real time RT-PCR (Qiagen,
Madrid, Spain) and by a generic conventional nested flavivirus RT-PCR (Gamino et
al., 2012). The product of the first generic PCR (1,048 bp) was resin purified, cloned
into pGEM-T (Promega, WI, USA) and sequenced. The obtained sequence was
compared to European and African USUV sequences available in GenBank using
BLAST (BLAST). In addition to histopathology, a polyclonal primary rabbit antibody
206

Captulo 6

directed against WNV, with proven cross-reactivity to other flaviviruses, was used
for viral antigen detection by immunohistochemistry (Gamino et al., 2012).
The thrushes were an adult male and female in poor body condition that had
greenish urate soiled feathers around the cloaca. Subcutaneous or visceral fat
deposits were absent and the pectoral muscle was partially atrophied, more severely
in the male. Both birds showed severe generalised congestion.
The serum, brain and pool of cloacal and oropharyngeal swabs of both birds
yielded a strongly positive signal in the generic flavivirus real time RT-PCR.
Sequencing of the PCR product obtained in the generic flavivirus RT-PCR (GenBank
accession number KC437386) showed a 96-97% homology to published USUV
sequences. Nucleotide sequence analysis revealed a higher homology to Northern
European strains (97% to BH65/11-02-03 [HE599647] and Meise H, Germany
[JQ219843]; Budapest, Hungary [EF206350]; Italy 2009 [JF266698]; and Vienna
2001, Austria [AY453411]) than to USUV isolated in South Africa (96% to SAAR 1776 [AY453412]), a finding also supported by phylogenetic analysis of USUV
strains with similar results for Maximum Likelihood and Neighbor-Joining methods
(Figure 1A, data not shown).
Histologically,

both

birds

had

severe

encephalitis

characterized

by

congestion, neuronal and Purkinje cell necrosis, gliosis, satellitosis, neuronophagia,


endothelial cell swelling and vasculitis. Other lesions included multiorgan
congestion, necrosis of renal tubular epithelium and moderate hemosiderosis in the
liver and spleen. Intravascular cross sections of microfilariaes were detected in
pulmonary capillaries. Virus antigen was detected in neurons in the cerebral
hemispheres and brain stem, and in glial cells throughout the brain (Figure 1B).
Rare Purkinje cells and neurons in peripheral ganglia (e.g. of the gizzard) as well as
cardiac myofibers and renal tubular epithelial cells were positive.

207

Virginia Gamino Rodrguez

Tesis doctoral

Figure 1. Usutu virus in migratory song thrushes in Spain. (A) Phylogenetic analysis of European
and African USUV. The evolutionary history was inferred using the Neighbor-Joining method. The
optimal tree with the sum of branch length = 1.18014408 is shown. The percentage of replicate trees
in which the associated taxa clustered together in the bootstrap test (1000 replicates) is shown next to
the branches. The tree is drawn to scale, with branch lengths in the same units as those of the
evolutionary distances used to infer the phylogenetic tree. The evolutionary distances were computed
using the Maximum Composite Likelihood method and are in the units of the number of base
substitutions per site. The analysis involved 10 nucleotide sequences. All ambiguous positions were
removed for each sequence pair. There were a total of 929 positions in the final dataset. Evolutionary
analyses were conducted in MEGA5. USUV are identified by their GenBank accession numbers and a
capital letter indicating the country of origin (A = Austria, G = Germany, I = Italy, H = Hungary, S =
Spain, SA = South Africa); the sequences of the last two branches correspond to outgroup viruses used
to root the tree (M = Murray Valley encephalitis virus [GenBank AF161266], J = Japanese encephalitis
virus [GenBank HM596272]). The song thrush strain from Spain 2012 is highlighted with a black
triangle. (B) Immunohistochemical staining with cross-reacting antibody showing USUV antigen
labeling in a Purkinje cell of the cerebellum of a song thrush that died from encephalitis. Original
magnification x400.

CONCLUSIONS
The

molecular

genetic

analysis,

histopathology

and

immunohistochemistry

confirmed encephalitis due to USUV in two migratory wintering song thrushes


collected in a small mortality event in southern Spain. Based upon phylogenetic
analysis, the USUV infecting the diseased birds was more similar to USUV from
Austria, Hungary and Germany, than to the USUV isolated from mosquitoes in
Spain (Busquets et al., 2008; Vzquez et al., 2011) or in the African continent. This
result together with the fact that song thrushes are a non-resident migratory
wintering bird species in southern Spain (MAGRAMA) provides circumstantial
evidence of USUV introduction in Spain during bird migration from northern
Europe. Persistence of USUV in song thrushes and recrudescence of the infection
during southward migration with concomitant filarial infection, together with the
208

Captulo 6

presence of USUV in a local endemic cycle from previous introductions, high vector
abundance and high viral loads in infectious mosquitoes are possible scenarios for
this outbreak.
Our data implies that introduction of USUV (and potentially other
flaviviruses such as WNV lineage 2, not yet detected in Spain) from northern
Europe in addition to local endemicity and introduction from Africa occurs, and
that zoonotic European USUV strains may be co-circulating with strains of African
origin. At this time it is not clear if the Spanish/African lineage differs in virulence
for humans from the European/African lineage. However, virus introduction by
northern migrants in combination with locally favourable conditions for vector
populations implies a risk for virus amplification and transmission and disease
outbreaks in humans and horses outside the currently established mosquitotrapping period (MayOctober) of targeted flavivirus surveillance programs.

ACKNOWLEDGEMENTS
This research was supported by European Union grants 278976 from the following
programs of the EMIDA ERANET (Coordination of European Research on Emerging
and Major Infectious Diseases of Livestock; www.emida-era.net): ANTIGONE
(Anticipating the Global Onset of Novel Epidemics) and APHAEA (Harmonised
Approaches in Monitoring Wildlife Population Health).

REFERENCES
Allering L, Jost H, Emmerich P, Gunther S, Lattwein E, Schmidt M, Seifried E,
Sambri V, Hourfar K, Schmidt-Chanasit J. Detection of Usutu virus infection
in a healthy blood donor from south-west Germany, 2012. Euro Surveill
2012, 17(50).
Basic Local Alignment Search Tool (BLAST) [http://blast.ncbi.nlm.nih.gov/]
Becker N, Jost H, Ziegler U, Eiden M, Hoper D, Emmerich P, Fichet-Calvet E,
Ehichioya DU, Czajka C, Gabriel M, Hoffmann B, Beer M, Tenner-Racz K, Racz
209

Virginia Gamino Rodrguez

Tesis doctoral

P, Gunther S, Wink M, Bosch S, Konrad A, Pfeffer M, Groschup MH, SchmidtChanasit J. Epizootic emergence of Usutu virus in wild and captive birds in
Germany. PLoS One 2012, 7(2):e32604.
Busquets N, Alba A, Allepuz A, Aranda C, Ignacio Nuez J. Usutu virus sequences
in Culex pipiens (Diptera: Culicidae), Spain. Emerg Infect Dis 2008,
14(5):861-863.
Chvala S, Bakonyi T, Bukovsky C, Meister T, Brugger K, Rubel F, Nowotny N,
Weissenbock H. Monitoring of Usutu virus activity and spread by using
dead bird surveillance in Austria, 2003-2005. Vet Microbiol 2007, 122(34):237-245.
Gamino V, Gutirrez-Guzmn AV, Fernndez-de-Mera IG, Ortiz JA, Durn-Martn
M, de la Fuente J, Gortzar C, Hfle U. Natural Bagaza virus infection in
game birds in southern Spain. Vet Res 2012, 43(1):65. doi: 10.1186/12979716-43-65.
Pecorari M, Longo G, Gennari W, Grottola A, Sabbatini A, Tagliazucchi S, Savini G,
Monaco F, Simone M, Lelli R, Rumpianesi F. First human case of Usutu virus
neuroinvasive infection, Italy, August-September 2009. Euro Surveill 2009,
14(50).
Spanish Ministry of Agriculture, Food and Environment (MAGRAMA): Atlas de las
aves
reproductoras
de
Espaa
[http://www.magrama.gob.es/es/
biodiversidad/temas/inventarios-nacionales/inventario-especies-terrestres/
inventario-nacional-de-biodiversidad/index2010-11-11_20.52.58.7490.aspx]
Vzquez A, Ruiz S, Herrero L, Moreno J, Molero F, Magallanes A, Snchez-Seco MP,
Figuerola J, Tenorio A. West Nile and Usutu viruses in mosquitoes in Spain,
2008-2009. Am J Trop Med Hyg 2011, 85(1):178-181.
Weissenbck H, Bakonyi T, Rossi G, Mani P, Nowotny N. Usutu Virus, Italy, 1996.
Emerg Infect Dis 2013, 19(2):274-277.
Woodall JP. The viruses isolated from arthropods at the East African Virus
Research Institute in the 26 years ending December 1963. Proc E Afr Acad
1964, 2.

210

Sntesis

Sntesis

Los tres flavivirus que han sido objeto de estudio de la presente tesis doctoral, y que
circulan en Espaa produciendo mortalidad en aves, son agentes patgenos
emergentes en cuya epidemiologa influyen numerosos factores relacionados con el
virus, el hospedador, el ambiente y el vector. La OIE considera como una
enfermedad emergente a toda aquella enfermedad infecciosa que cumpla uno o
varios de los siguientes requisitos:
1) Una infeccin que aparece como consecuencia de la evolucin o de un
cambio en un agente patgeno existente.
2) Un

patgeno

una

enfermedad

no

descrita

previamente

que

es

diagnosticada por primera vez.


3) Una infeccin conocida que se distribuye hacia una nueva zona geogrfica o
una nueva poblacin hospedadora.
El ejemplo ms claro del primer tipo de enfermedad emergente en nuestro
pas es la infeccin por WNV. A pesar de que su circulacin en Espaa se conoce
desde hace medio siglo, no haba dado lugar a casos de enfermedad y mortalidad
hasta

la

ltima

dcada.

Este

flavivirus

ha

sido

ampliamente

estudiado,

fundamentalmente desde su introduccin en Norteamrica en el ao 1999, dada las


graves consecuencias que esto supuso, y que sigue suponiendo, no slo para aves
sino tambin para quidos y humanos. Despus de su introduccin en el Nuevo
Continente, este virus ha re-emergido como patgeno para humanos, quidos y
aves en el Viejo Continente. No obstante, como ya se ha indicado previamente en
esta tesis, mientras que WNV ha causado miles de muertes de aves en
Norteamrica,

los

brotes

detectados

en

Europa

son

muy

escasos

fundamentalmente parecen afectar a aves rapaces. Adems, mientras que algunas


especies como los crvidos han demostrado ser muy susceptibles a la infeccin,
otras, como las encuadradas en el orden Galliformes, si bien pueden infectarse,
raramente desarrollan enfermedad y sucumben a la misma. Ante esta situacin, se
213

Virginia Gamino Rodrguez

Tesis doctoral

han realizado numerosos trabajos que tratan de explicar el porqu de la reemergencia de este flavivirus, el porqu de las diferencias en su comportamiento
eco-epidemiolgico entre Europa y el continente norteamericano y el porqu de las
diferencias en las consecuencias de la infeccin para las diferentes especies de
aves.

En relacin a

esto,

puesto

que

existen todava

pocos

estudios

experimentales realizados en aves, se plantearon los tres primeros captulos de esta


tesis doctoral.
El Captulo 1 se centr, fundamentalmente, en realizar una revisin de la
informacin existente sobre la patogenia, lesiones y tropismo celular y tisular en la
infeccin por WNV en aves, resaltando las diferencias encontradas entre rdenes y
familias que pudiesen explicar el hecho de que en unas especies la infeccin sea
ms grave que en otras. Con los datos recopilados se comprob que, efectivamente,
existen diferencias entre especies, incluso dentro de la misma familia, en relacin a
la abundancia y distribucin del antgeno viral as como en la severidad y
distribucin de las lesiones macroscpicas y microscpicas. Estas diferencias estn
relacionadas, fundamentalmente, con el momento post infeccin en el que el
hospedador muere, lo que a su vez est determinado por la susceptibilidad del
mismo a la infeccin. De esta forma, existen especies en las que la respuesta a la
infeccin es dbil, y en muchos casos tarda, permitiendo una elevada replicacin
del virus en los tejidos que provoca la muerte rpida del hospedador. En este grupo
se podran encuadrar algunas aves del orden Passeriformes, como los cuervos o los
arrendajos, que presentan lesiones de carcter agudo, sobre todo de tipo
inflamatorio, en algunos casos ausentes en el SNC, ya que estos individuos pueden
morir incluso antes de que el virus llegue a este tejido. Existe un segundo grupo de
hospedadores capaz de mantener una replicacin viral baja, que da lugar a una
infeccin crnica, la cual finalmente puede llevar a la muerte del individuo o al
desarrollo de secuelas a largo plazo. En este grupo podran encuadrarse las aves

214

Sntesis

rapaces, en las que es caracterstica la presencia de lesiones crnicas que tambin


son detectadas en el SNC. Finalmente, existe un tercer grupo de hospedadores que
responde a la infeccin de forma efectiva, en muchos casos de forma temprana,
eliminando el virus rpidamente, y en el que slo algunos individuos mueren como
consecuencia de la enfermedad. En este ltimo grupo se encuadraran algunas aves
del orden Galliformes, en las que se detectan lesiones de carcter moderado, en
muchos casos ms relacionadas con la respuesta inflamatoria del hospedador que
con la replicacin activa del virus.
No obstante, aunque la especie tiene cierto valor predictivo, el desarrollo de
lesiones, enfermedad y muerte asociado a la infeccin por WNV est modulado por
muchos otros factores relacionados, sobre todo, con el virus y el hospedador y que,
en muchos casos, hacen difcil predecir el desenlace de la infeccin. Como ya se ha
indicado anteriormente, se ha visto que una simple modificacin gentica y
aminoacdica en el virus puede generar un cambio en su virulencia, ya sea por una
mayor capacidad para replicarse o por una mayor habilidad para evadir la
respuesta inmune del hospedador (Brault et al., 2004; Ye et al., 2013). De este
modo, la emergencia o re-emergencia de la virulencia de WNV se ha relacionado en
gran parte con este factor (Brault et al., 2007; Sotelo et al., 2011a). Por otro lado,
cualquier factor que modifique la capacidad de respuesta del hospedador frente al
virus, como puede ser la edad, la existencia de enfermedades concurrentes, o la
presencia

de

inmunidad

previa

frente

al

flavivirus

infectante

otros

antignicamente relacionados, influye en la evolucin de la infeccin (Eldadah et


al., 1967; Fang y Reisen, 2006; Hfle et al., 2008).
Para determinar cmo las variaciones genticas y aminoacdicas de WNV
pueden modificar su virulencia para las aves, y si existen diferencias en la
susceptibilidad a la infeccin entre aves de Norteamrica y Europa, en los captulos
2 y 3 estudiamos la patogenia de la infeccin experimental de un ave mediterrnea
215

Virginia Gamino Rodrguez

Tesis doctoral

endmica con tres cepas de WNV pertenecientes al linaje 1. En el Captulo 2, se


infectaron perdices rojas con dos cepas mediterrneas del virus, MO03 y SP07. El
objetivo fundamental fue estudiar las causas que pudiesen explicar el diferente
curso de la infeccin y la distinta mortalidad provocada por ambas cepas descritos
en un trabajo previo (Sotelo et al., 2011b). Se encontraron diferencias entre las dos
cepas en relacin a la distribucin del virus y nivel de replicacin viral en tejidos,
as como en la severidad de las lesiones, pero stas fueron consideradas
insuficientes como para determinar la mayor mortalidad causada por MO03. La
diferente patogenia de la infeccin en el SNC, donde MO03 dio lugar a una
aparicin de lesiones ms severas de forma ms temprana y a una reaccin
inflamatoria ms marcada, podra explicar la distinta mortalidad. Las lesiones que
WNV

provoca

en

el

SNC

estn

relacionadas

con

la

neuroinvasividad

neurovirulencia de la cepa. En nuestro trabajo la neuroinvasividad de ambas cepas


fue muy similar, puesto que el nivel de viremia, la llegada al SNC y el nivel de
replicacin viral en este tejido fueron muy similares. Por ello, determinamos que fue
la diferente neurovirulencia la que dio lugar a la diferente gravedad de la infeccin.
Sin embargo, an se desconocen los determinantes especficos que hacen que
MO03 resulte ms neurovirulenta. En el Captulo 3, se realiz un estudio similar,
pero en este caso las perdices fueron infectadas con una cepa norteamericana de
WNV, NY99. El objetivo de este trabajo fue determinar si esta especie es tambin
susceptible a la infeccin con esta cepa. Aunque las perdices desarrollaron lesiones
y murieron como consecuencia de la enfermedad, la replicacin del virus en los
tejidos fue mnima, a pesar de la alta dosis de inoculacin (107 UFP) que se utiliz.
As mismo, al comparar la patogenia de la infeccin en los individuos eutanasiados
con la observada en el estudio anterior, se demostr que las lesiones fueron menos
severas y que aparecieron de forma ms tarda en el curso de la infeccin. El hecho
de que las perdices infectadas con NY99 tuviesen dos semanas ms de edad podra

216

Sntesis

estar influyendo en estas diferencias; sin embargo, es ms probable que la causa


est asociada a una menor virulencia de la cepa NY99 de WNV.
Dados los resultados de estos dos trabajos, se demuestra, por un lado, que la
perdiz roja es susceptible a la infeccin por WNV, desarrollando lesiones similares a
las descritas en otras aves (Steele et al., 2000), que incluso llegan a producir la
aparicin de signos clnicos y mortalidad. Por otro lado, que las tres cepas de WNV
estudiadas

presentan

una

diferente

virulencia

para

esta

especie,

siendo

especialmente elevada la de MO03. Sin embargo, se desconoce qu determinantes


genticos o aminoacdicos (Sotelo et al., 2009) estn marcando esta diferente
virulencia, al no haber podido relacionarla con la sustitucin de treonina por
prolina en la posicin 249 de la protena NS3, considerada como un marcador de
virulencia para cuervos americanos (Brault et al., 2007). De cualquier forma,
considerando nuestros resultados, las diferencias a nivel gentico o protenico
podran explicar, al menos en parte, la emergencia o re-emergencia de la virulencia
de este flavirus para las aves y para otros hospedadores. No obstante, la
epidemiologa de WNV es muy compleja, y existen otros factores que podran estar
contribuyendo a una mayor circulacin del virus como, por ejemplo, el cambio
climtico (Jimnez-Clavero et al., 2012). Adems, es posible que el incremento de la
sensibilizacin de la poblacin sobre la importancia que tiene la infeccin por este
virus para las aves y, sobre todo, para los humanos, y la mejora de los planes de
vigilancia estn contribuyendo a la deteccin de casos que antes pasaban
desapercibidos (Burt et al., 2002).
Aunque ambos estudios son experimentales y han sido realizados con un
bajo nmero de animales, y se desconoce cmo transcurrira la infeccin en
condiciones naturales, se podra decir que al menos esta ave endmica europea es
moderadamente susceptible a la infeccin por WNV y que en Europa circulan cepas
que han demostrado ser virulentas para sta y otras especies de aves, tanto de
217

Virginia Gamino Rodrguez

Tesis doctoral

forma experimental como natural (Jimnez-Clavero et al., 2008; Bakonyi et al.,


2013; Dridi et al., 2013). Por lo tanto, las menores consecuencias de la infeccin
por WNV para las aves europeas no parecen estar relacionadas ni con la baja
virulencia de las cepas circulantes ni con la baja susceptibilidad de las especies de
aves a la infeccin. Es probable que aquellos factores que modulan la efectividad de
la transmisin del virus o la probabilidad de infeccin del hospedador, como
pueden ser la abundancia de vectores, su actividad y la preferencia de hospedador,
o la abundancia de hospedadores reservorio y la biodiversidad (Ezenwa et al., 2006;
Brault, 2009; Hamer et al., 2011), estn jugando un papel mucho ms importante.
No obstante, el constante contacto de las aves del Viejo Continente con WNV, o con
flavivirus antignicamente relacionados, que hace que stas desarrollen inmunidad,
tambin podra explicar la baja incidencia de la enfermedad (Reiter, 2010).
La enfermedad asociada a la infeccin natural por BAGV encontrada en aves
cinegticas del sur de Espaa, constituye un ejemplo del segundo tipo de
enfermedad emergente previamente citado, ya que este flavivirus nunca haba sido
asociado a ningn caso de enfermedad o mortalidad. En el Captulo 4 se realiz,
por primera vez, una descripcin detallada de los hallazgos microscpicos y de la
distribucin del antgeno viral en aves infectadas de forma natural por este virus,
tratando fundamentalmente de determinar el porqu de su mayor virulencia para la
perdiz roja en comparacin con el faisn comn y la paloma torcaz. Los hallazgos
clnicos y las lesiones encontradas fueron muy similares a lo que se observa en la
infeccin por otros flavivirus, entre ellos WNV, detectndose una encefalitis
marcada en las tres especies. El mayor impacto de esta infeccin en la perdiz roja
se relacion con una distribucin tisular del virus mucho ms amplia y, sobre todo,
con el grave proceso hemoltico que provoc exclusivamente en esta especie. Este
estudio nos sirvi para corroborar que no todas las especies se ven igualmente
afectadas por la infeccin por un flavivirus, y podra constituir un ejemplo de lo que
218

Sntesis

ocurre cuando un nuevo virus es introducido en una poblacin hospedadora que


nunca antes haba contactado con el mismo. Sin embargo, se desconoce desde
cundo llevaba circulando BAGV en nuestro pas. El hecho de que fuese un brote
muy localizado, afectando a pocas especies, y que no se detectasen mortalidades
posteriores, pudo deberse a que fue un caso puntual favorecido por las condiciones
climatolgicas de ese ao y la elevada abundancia de vectores, as como la alta
densidad de aves en la zona. Es posible que la presencia de inmunidad cruzada
frente a otros flavivirus y el desarrollo de inmunidad frente BAGV (Llorente et al.,
2013) hayan evitado la aparicin de nuevas mortalidades.
A pesar de que, como se ha indicado en el apartado anterior, los signos
clnicos y las lesiones fueron muy similares en la infeccin por BAGV y WNV,
consideramos interesante la deteccin de ceguera slo en las perdices infectadas
por BAGV. Esto nos llev a plantear el estudio que constituye el Captulo 5, en el
que se determin que la ceguera de las perdices infectadas por BAGV est
relacionada con una inflamacin ocular ms severa y una mayor replicacin del
virus en la retina. Estos hallazgos son similares a los que se obtienen si se
comparan los encontrados en rapaces diurnas y nocturnas infectadas por WNV. En
las primeras se observa ceguera o visin alterada como consecuencia de la
presencia de infiltrados inflamatorios oculares extensos y una grave afectacin de la
retina asociada la replicacin del virus. Por el contrario, en las rapaces nocturnas, a
pesar de presentar una inflamacin moderada, la retina rara vez se ve afectada,
haciendo que sea poco frecuente la deteccin de signos relacionados con la
infeccin ocular (Wnschmann et al., 2005; Lopes et al., 2007; Pauli et al., 2007).
Por otro lado, nuestro estudio nos permiti determinar que la entrada de WNV
NY99 en el ojo de la perdiz roja se produce por va hematgena durante la viremia
primaria.

219

Virginia Gamino Rodrguez

Tesis doctoral

La infeccin por USUV podra encuadrarse en el tercer tipo de enfermedad


emergente citado al principio de este captulo de sntesis, puesto que ya se conoca
su virulencia para las aves, sobre todo de la familia Turdidae, y, aunque el virus
haba sido detectado en mosquitos (Busquets et al., 2008; Vzquez et al., 2011), el
Captulo 6 constituye el primer trabajo en el que se describe la infeccin por USUV
en aves en nuestro pas. Los dos zorzales infectados mostraron lesiones muy
similares a las descritas en aves de Centroeuropa infectadas por el mismo virus
(Chvala et al., 2004). El hecho de que el zorzal comn sea un ave migratoria que
pasa los inviernos en Espaa (MAGRAMA) y que la cepa detectada mostrara mayor
homologa con las circulantes en Centroeuropa que con las detectadas previamente
en Espaa y frica, parece indicar que, probablemente, estas aves vinieron
infectadas y que el estrs asociado a la migracin recrudeci la infeccin,
provocando la muerte de las mismas. No obstante, es posible que esta cepa ya
estuviese circulando en Espaa y los zorzales se infectasen en nuestro pas. Puesto
que no se ha detectado enfermedad o mortalidad en otras aves de la zona, es difcil
determinar qu consecuencias tiene la presencia de esta cepa.
Los hallazgos microscpicos incidentales de los trabajos anteriores indican
una frecuente asociacin de la infeccin por estos flavivirus con la presencia de
otros patgenos. En unos casos estos patgenos aprovecharn la inmunosupresin
provocada por el flavivirus para multiplicarse, contribuyendo al deterioro del estado
de salud del hospedador, y en otros sern la causa de la inmunosupresin del
hospedador, haciendo que finalmente sucumba a la infeccin por el flavivirus. De
esta forma, los coccidios detectados en el intestino grueso y la candidiasis
observada en el buche de las perdices objeto de estudio de los captulos 2 y 3,
probablemente

vieron

favorecida

su

proliferacin

por

la

inmunosupresin

provocada por la infeccin con WNV. Por el contrario, la presencia de tuberculosis


crnica en algunos de los faisanes y las palomas infectadas por BAGV posiblemente
220

Sntesis

contribuy a la susceptibilidad de estos hospedadores a la infeccin por el


flavivirus.
En conjunto, en esta tesis hemos demostrado, por un lado, que en Espaa
circulan flavivirus capaces de infectar a aves cinegticas, dando lugar a la aparicin
de lesiones que, en algunos casos, puede llegar a producir la muerte del
hospedador. Sin embargo, tambin hemos visto que su eco-epidemiologa es
extremadamente compleja, y que la susceptibilidad del hospedador o la virulencia
del virus slo determinan una parte de la misma. Debido a que los tres flavivirus
comparten hospedadores susceptibles y vectores, que se ha demostrado que
circulan de forma silenciosa en nuestro pas (Llorente et al., 2013) y que al menos
WNV y USUV son zoonticos, existe un riesgo de aparicin de epizootias o
epidemias, sin embargo, su prediccin resulta muy complicada. Dada la
importancia socioeconmica que tienen algunas de estas aves, fundamentalmente
la perdiz roja, la infeccin por flavivirus podra tener consecuencias graves en caso
de

provocar grandes

mortalidades.

Adems,

considerando

que

estas

aves

constituyen el recurso trfico de otras especies (algunas de ellas amenazadas), que


estos virus se replican en tejidos y que la transmisin por ingestin de tejidos
infectados ha sido demostrada, al menos para WNV (Komar et al., 2003), la
infeccin por flavivirus tambin podra tener consecuencias ecolgicas graves.
Por otro lado, la perdiz roja ha demostrado ser un buen modelo experimental
de la infeccin por WNV, permitiendo ampliar los conocimientos sobre la patogenia
de la infeccin por este flavivirus en aves.
Considerando los resultados obtenidos en esta tesis, sera interesante
plantear futuros estudios que nos permitiesen, por un lado, determinar los factores
que hacen que la cepa MO03 de WNV sea ms virulenta para la perdiz roja y, por
otro lado, determinar la patogenia de la infeccin por BAGV en aves, para poder

221

Virginia Gamino Rodrguez

Tesis doctoral

esclarecer, entre otras cosas, las causas de la severa hemlisis detectada en la


perdiz roja y su diferente virulencia para distintas especies de aves. Por ltimo,
consideramos interesante determinar si la cepa de USUV ya estaba circulando en
Espaa o, en caso de haber sido introducida por los zorzales, dnde se infectaron y
si la cepa sigue circulando en nuestro pas.

BIBLIOGRAFA
Bakonyi T, Ferenczi E, Erdlyi K, Kutasi O, Csorgo T, Seidel B, Weissenbck H,
Brugger K, Ban E, Nowotny N. Explosive spread of a neuroinvasive lineage 2
West Nile virus in Central Europe, 2008/2009. Vet Microbiol 2013. pii:
S0378-1135(13)00164-8. doi: 10.1016/j.vetmic.2013.03.005.
Brault AC, Langevin SA, Bowen RA, Panella NA, Biggerstaff BJ, Miller BR, Komar N:
Differential virulence of West Nile strains for American crows. Emerg Infect
Dis 2004, 10(12):2161-2168.
Brault AC, Huang CY, Langevin SA, Kinney RM, Bowen RA, Ramey WN, Panella NA,
Holmes EC, Powers AM, Miller BR: A single positively selected West Nile
viral mutation confers increased virogenesis in American crows. Nat Genet
2007, 39(9):1162-1166.
Brault AC: Changing patterns of West Nile virus transmission: altered vector
competence and host susceptibility. Vet Res 2009, 40(2):43. doi:
10.1051/vetres/2009026.
Burt FJ, Grobbelaar AA, Leman PA, Anthony FS, Gibson GV, Swanepoel R:
Phylogenetic relationships of southern African West Nile virus isolates.
Emerg Infect Dis 2002, 8(8):820-826.
Busquets N, Alba A, Allepuz A, Aranda C, Ignacio Nuez J. Usutu virus sequences
in Culex pipiens (Diptera: Culicidae), Spain. Emerg Infect Dis 2008,
14(5):861-863.
Chvala S, Kolodziejek J, Nowotny N, Weissenbock H. Pathology and viral
distribution in fatal Usutu virus infections of birds from the 2001 and
2002 outbreaks in Austria. J Comp Pathol 2004, 131(2-3):176-185.
Dridi M, Vangeluwe D, Lecollinet S, van den Berg T, Lambrecht B. Experimental
infection of Carrion crows (Corvus corone) with two European West Nile
virus (WNV) strains. Vet Microbiol 2013. pii: S0378-1135(13)00061-8. doi:
10.1016/j.vetmic.2012.12.043.
Eldadah AH, Nathanson N, Sarsitis R: Pathogenesis of West Nile Virus
encephalitis in mice and rats. 1. Influence of age and species on mortality
and infection. Am J Epidemiol 1967, 86(3):765-775.

222

Sntesis

Ezenwa VO, Godsey MS, King RJ, Guptill SC: Avian diversity and West Nile virus:
testing associations between biodiversity and infectious disease risk. Proc
Biol Sci 2006, 273(1582):109-117.
Fang Y, Reisen WK: Previous infection with West Nile or St. Louis encephalitis
viruses provides cross protection during reinfection in house finches. Am J
Trop Med Hyg 2006, 75(3):480-485.
Hamer GL, Chaves LF, Anderson TK, Kitron UD, Brawn JD, Ruiz MO, Loss SR,
Walker ED, Goldberg TL. Fine-scale variation in vector host use and force of
infection drive localized patterns of West Nile virus transmission. PLoS One
2011, 6(8):e23767. doi: 10.1371/journal.pone.0023767.
Hfle U, Blanco JM, Crespo E, Naranjo V, Jimnez-Clavero MA, Snchez A, de la
Fuente J, Gortzar C: West Nile virus in the endangered Spanish imperial
eagle. Vet Microbiol 2008, 129(1-2):171-178.
Jimnez-Clavero MA, Sotelo E, Fernndez-Pinero J, Llorente F, Manuel Blanco J,
Rodrguez-Ramos J, Prez-Ramrez E, Hfle U. West Nile virus in golden
eagles, Spain, 2007. Emerg Infect Dis 2008, 14(9):1489-1491.
Jimnez-Clavero MA: Animal viral diseases and global change: bluetongue and
West Nile fever as paradigms. Front Genet 2012, 3:105. doi:
10.3389/fgene.2012.00105.
Komar N, Langevin S, Hinten S, Nemeth N, Edwards E, Hettler D, Davis B, Bowen
R, Bunning M. Experimental infection of North American birds with the
New York 1999 strain of West Nile virus. Emerg Infect Dis 2003, 9(3):311322.
Llorente F, Prez-Ramrez E, Fernndez-Pinero J, Soriguer R, Figuerola J, JimnezClavero MA. Flaviviruses in Game Birds, Southern Spain, 20112012. Emerg
Infect Dis 2013, 19(6):1024-1026. doi: 10.3201/eid 1906.130122.
Lopes H, Redig P, Glaser A, Armien A, Wnschmann A: Clinical findings, lesions,
and viral antigen distribution in great gray owls (Strix nebulosa) and
barred owls (Strix varia) with spontaneous West Nile virus infection. Avian
Dis 2007, 51(1):140-145.
Ministerio de Agricultura, Alimentacin y Medio Ambiente (MAGRAMA): Atlas de las
aves
reproductoras
de
Espaa
[http://www.magrama.gob.es/es/
biodiversidad/temas/inventarios-nacionales/inventario-especies-terrestres/
inventario-nacional-de-biodiversidad/index2010-11-11_20.52.58.7490.aspx]
Organizacin Mundial de Sanidad Animal (OIE) (2012): Cdigo Sanitario para los
Animales Terrestres (2012) [http://www.oie.int/es/normas-internacionales/
codigo-terrestre/acceso-en-linea/]
Pauli AM, Cruz-Martinez LA, Ponder JB, Redig PT, Glaser AL, Klauss G, Schoster
JV, Wunschmann A: Ophthalmologic and oculopathologic findings in redtailed hawks and Cooper's hawks with naturally acquired West Nile virus
infection. J Am Vet Med Assoc 2007, 231(8):1240-1248.

223

Virginia Gamino Rodrguez

Tesis doctoral

Reiter P: West Nile virus in Europe: understanding the present to gauge the
future. Euro Surveill 2010, 15(10):19508.
Sotelo E, Fernndez-Pinero J, Llorente F, Agero M, Hoefle U, Blanco JM, JimnezClavero MA. Characterization of West Nile virus isolates from Spain: new
insights into the distinct West Nile virus eco-epidemiology in the Western
Mediterranean. Virology 2009, 395(2):289-297.
Sotelo E, Fernndez-Pinero J, Llorente F, Vzquez A, Moreno A, Agero M, Cordioli
P, Tenorio A, Jimnez-Clavero MA: Phylogenetic relationships of Western
Mediterranean West Nile virus strains (1996-2010) using full-length
genome sequences: single or multiple introductions? J Gen Virol 2011a,
92(Pt 11):2512-2522.
Sotelo E, Gutirrez-Guzmn AV, del Amo J, Llorente F, El-Harrak M, Prez-Ramrez
E, Blanco JM, Hoefle U, Jimnez-Clavero MA. Pathogenicity of two recent
Western Mediterranean West Nile virus isolates in a wild bird species
indigenous to Southern Europe: the red-legged partridge. Vet Res 2011b,
42. doi: 10.1186/1297-9716-42-11.
Steele KE, Linn MJ, Schoepp RJ, Komar N, Geisbert TW, Manduca RM, Calle PP,
Raphael BL, Clippinger TL, Larsen T, Smith J, Lanciotti RS, Panella NA,
McNamara TS: Pathology of fatal West Nile virus infections in native and
exotic birds during the 1999 outbreak in New York City, New York. Vet
Pathol 2000, 37(3):208-224.
Vzquez A, Ruiz S, Herrero L, Moreno J, Molero F, Magallanes A, Snchez-Seco MP,
Figuerola J, Tenorio A: West Nile and Usutu viruses in mosquitoes in Spain,
2008-2009. Am J Trop Med Hyg 2011, 85(1):178-181.
Wnschmann A, Shivers J, Bender J, Carroll L, Fuller S, Saggese M, van Wettere A,
Redig P: Pathologic and immunohistochemical findings in goshawks
(Accipiter gentilis) and great horned owls (Bubo virginianus) naturally
infected with West Nile virus. Avian Dis 2005, 49(2):252-259.
Ye J, Zhu B, Fu ZF, Chen H, Cao S: Immune evasion strategies of flaviviruses.
Vaccine 2013, 31(3):461-471.

224

Conclusiones

Conclusiones

1. Aunque la mayora de las aves infectadas por WNV desarrollan lesiones


macroscpicas y/o microscpicas, existen diferencias interespecie en la
naturaleza, severidad o distribucin de las mismas relacionadas con el
momento post infeccin en el que el animal muere, lo que est determinado por
la susceptibilidad del mismo a la infeccin.

2. La

perdiz roja

es

susceptible a

la

infeccin experimental

con WNV,

desarrollando lesiones muy similares a las encontradas en otras aves infectadas


por el virus.

3. La perdiz roja constituye un modelo experimental apropiado para estudiar la


patogenia de la infeccin por WNV en aves.

4. La infeccin experimental de la perdiz roja con WNV provoca una encefalitis en


la que participan componentes celulares inflamatorios similares a los descritos
en roedores y humanos infectados por el mismo virus.

5. Las diferencias genticas y aminoacdicas entre diferentes cepas de WNV dan


lugar a variaciones en la patogenia de la infeccin experimental de la perdiz roja
que se traducen en una diferente patogenicidad.

6. El diferente comportamiento eco-epidemiolgico de WNV entre Norteamrica y


Europa no parece estar asociado a la menor susceptibilidad de las aves
europeas a la infeccin ni a la menor virulencia de las cepas circulantes en
Europa.

227

Virginia Gamino Rodrguez

Tesis doctoral

7. BAGV es un flavivirus emergente capaz de producir enfermedad en la perdiz


roja, el faisn comn y la paloma torcaz, siendo ms virulento para la primera,
al mostrar un tropismo tisular ms amplio y generar un proceso hemoltico
grave.

8. La infeccin por BAGV causa ceguera en la perdiz roja como consecuencia del
desarrollo de lesiones inflamatorias oculares severas y la intensa replicacin del
virus en la retina.

9. La llegada de la cepa NY99 de WNV al ojo de perdices rojas infectadas de forma


experimental ocurre va hematgena.

10. La cepa de USUV que produjo el evento de mortalidad en zorzal comn en


Espaa probablemente fue introducida por los propios zorzales desde el centro
de Europa. Esta cepa dio lugar a una enfermedad similar a la encontrada en
otras aves infectadas por el mismo virus, que posiblemente se vio favorecida por
el estrs causado por la migracin.

228

También podría gustarte