THE PATHOGENESIS OF EHV1 IN HORSES: NOVEL ... of contents 5 TABLE OF CONTENTS LIST OF ABBREVIATIONS...

170
THE PATHOGENESIS OF EHV1 IN HORSES: NOVEL INSIGHTS FROM EXPERIMENTAL INOCULATIONS AND FIELD SITUATIONS Gryspeerdt Annick Thesis submitted in fulfilment of the requirements for the degree of doctor (Ph.D.) in Veterinary Sciences, Faculty of Veterinary Medicine, Ghent University, 2011 Promotors: Prof. Dr. H. Nauwynck Laboratory of Virology Department of Virology, Parasitology and Immunology Prof. Dr. G. Van de Walle Department of Comparative Physiology and Biometrics Faculty of Veterinary Medicine

Transcript of THE PATHOGENESIS OF EHV1 IN HORSES: NOVEL ... of contents 5 TABLE OF CONTENTS LIST OF ABBREVIATIONS...

THE PATHOGENESIS OF EHV1 IN HORSES: NOVEL

INSIGHTS FROM EXPERIMENTAL INOCULATIONS AND

FIELD SITUATIONS

Gryspeerdt Annick

Thesis submitted in fulfilment of the requirements for the degree of doctor (Ph.D.) in

Veterinary Sciences, Faculty of Veterinary Medicine, Ghent University, 2011

Promotors:

Prof. Dr. H. Nauwynck

Laboratory of Virology

Department of Virology, Parasitology and Immunology

Prof. Dr. G. Van de Walle

Department of Comparative Physiology and Biometrics

Faculty of Veterinary Medicine

3

ISBN: 9789058642554 EAN: 9789058642554

© 2011 by Laboratory of Virology, Ghent University, Salisburylaan 133, 9820

Merelbeke, Belgium

The author and promoter give the authorization to consult and copy parts of this work for

personal use only. Every other use is subject to the copyright laws. Permission to

reproduce any material contained in this work should be obtained from the author.

Annick Gryspeerdt was supported by a doctoral grant from the Institute for the promotion

of Innovation through Science an Technology Flanders (IWT-Vlaanderen).

Table of contents

5

TABLE OF CONTENTS

LIST OF ABBREVIATIONS

I INTRODUCTION

1. EQUINE HERPESVIRUS 1 (EHV1): general characteristics

1.1 History

1.2 General characteristics

2. EQUINE HERPESVIRUS 1 and the cell

2.1 Virus structure

2.2 Replication cycle

3. EQUINE HERPESVIRUS 1 and the horse

3.1 Epidemiology and prevalence

3.2 Pathogenesis and disease

3.3 Diagnosis

3.4 Disease control

II AIMS OF THE THESIS

III DIFFERENCES IN REPLICATION KINETICS AND CELL TROPISM

BETWEEN NEUROVIRULENT AND NON-NEUROVIRULENT EHV1

STRAINS DURING THE ACUTE PHASE OF INFECTION IN HORSES

IV EXPRESSION OF LATE VIRAL PROTEINS IS HAMPERED IN

INFECTED NASAL MUCOSAL LEUKOCYTES BUT NOT IN

EPITHELIAL CELLS DURING EARLY PATHOGENESIS OF EQUINE

HERPESVIRUS TYPE 1 (EHV1) INFECTION

V DESCRIPTION OF AN UNUSUALLY LARGE OUTBREAK OF

NERVOUS SYSTEM DISORDERS CAUSED BY EQUINE HERPESVIRUS

1 (EHV1) IN 2009 IN BELGIUM

VI GENERAL DISCUSSION

SUMMARY

SAMENVATTING

CURRICULUM VITAE

BIBLIOGRAPHY

DANKWOORD

7

LIST OF ABBREVIATIONS

Ab anitbody

ACV acyclovir

AHV asinine herpesvirus

BM basement membrane

C complement

CD cluster of differentiation

CML cell of the monocytic lineage

CNS central nervous system

CO2 carbon dioxide

ConA concanavalin A

CSF cerebrospinal fluid

CTL cytotoxic T-lymphocyte

D aspartic acid

DC dendritic cell

DMEM Dulbecco’s modified eagle medium

DMSO dimethyl sulfoxide

DNA deoxyribonucleic acid

dpi days post inoculation

E early

EEL equine embryonic lung

EHM equine herpes myelitis

EHV equine herpesvirus

ELISA enzyme-linked immunosorbent assay

ER endoplasmatic reticulum

FCS fetal calf serum

FITC fluoresceine isothiocyanate

g glycoprotein

HCMV human cytomegalovirus

hpi hours post inoculation

IE1 immediate early polypeptide

8

IFN interferon

Ig immunoglobulin

IL interleukin

IONO ionomycin

IPMA immunoperoxidase monolayer assay

IR1 immediate early gene

IV intravenous

L late

mAb monoclonal antibody

MEM minimum essential medium

MHC major histocompatibility complex

MLV modified-live vaccine

mRNA messenger ribonucleic acid

ORF open reading frame

PBMC peripheral blood mononuclear cells

PBS phosphate buffered saline

PCR polymerase chain reaction

PDB phorbol dibutyrate

PHA phytohaemagglutinin

PMW pokeweed mitogen

PRV pseudo rabies virus

RK13 rabbit kidney 13

RLFP restriction length polymorphism

ROCK Rho-associated coiled-coil kinase

RPMI Roswell Park Memorial Institute

RSD arginine-serine-aspartic acid

SD standard deviation

SN seroneutralization

SNP single nucleotide polymorphism

siRNA small interfering ribonucleic acid

TAP transporter associated with antigen processing

TGN trans-Golgi network

TCID tissue culture infectious dose

9

TK thymidine kinase

TR Texas Red

URT upper respiratory tract

USDA United States Department of Agriculture

VACV valacyclovir

VN virus neutralizing

WAHV wild ass herpesvirus

ZHV zebra herpesvirus

Review of the literature

11

CHAPTER I

REVIEW OF THE LITERATURE

1. EQUINE HERPESVIRUS 1 (EHV1): general characteristics

1.1 History

1.2 General characteristics

2. EQUINE HERPESVIRUS 1 and the cell

2.1 Virus structure

2.2 Replication cycle

3. EQUINE HERPESVIRUS 1 and the horse

3.1 Epidemiology and prevalence

3.2 Pathogenesis and disease

3.2.1 Pathogenesis

3.2.2 Symptoms

3.2.3 Treatment

3.3 Diagnosis

3.4 Disease control

Review of the literature

13

1. EQUINE HERPESVIRUS 1 (EHV1): general characteristics

1.1 History

Equine herpesvirus 1 (EHV1) was first described in the early 1930s after a

necropsy examination of an aborted fetus (Dimock & Edwards, 1933), but virus

isolation from such cases succeeded only decades later (Jackson & Kendrick, 1971;

Saxegaard, 1966). The first recorded observation to indicate that major antigenic

differences may exist among clinical isolates of EHV1, was made in 1959 (Shimizu et

al., 1959). Thirteen years elapsed before a high correlation between antigenic subtype

and anatomic origin of the virus isolates was described (Burrows & Goodridge, 1972).

Until 1981, EHV1 and EHV4 were considered as two subtypes of the same virus,

namely ‘EHV1’, causing febrile rhinopneumonitis, ataxia, abortions and neonatal foal

disease in horses (Allen & Bryans, 1986). Unambiguous differentiation into EHV1

and EHV4 came from viral genomic fingerprints or so-called restriction length

polymorphisms (RLFP), but official recognition of the distinction between those two

viruses came almost one decade later (Roizman et al., 1992; Sabine et al., 1981;

Studdert et al., 1981; Turtinen et al., 1981).

1.2 General characteristics

1.2.1 Classification

The revised family Herpesviridae retains the viruses of mammals, birds and

reptiles. Members are highly disseminated in nature, with isolations from most animal

species of at least one and frequently several diverse herpesviruses (Davison et al.,

2009; Roizman & Pellett, 2001). Assignment to the Herpesviridae family is made by

the criterion of the characteristic herpes virion architecture. The known herpesviruses

appear to share four significant biological properties: (1) although the exact array of

enzymes may vary from one herpesvirus to another, all herpesviruses specify a large

array of enzymes involved in nucleic acid metabolism, DNA synthesis and processing

of proteins, (2) synthesis of viral DNA and capsid assembly occurs in the nucleus, (3)

production of infectious progeny virus is always accompanied by the destruction of

Review of the literature

14

the infected cell and (4) herpesviruses are able to remain latent in their natural hosts

(Roizman & Pellett, 2001).

Herpesviruses differ with respect to their biologic properties and are classified into

three different subfamilies based on host range, length of the reproduction cycle,

spread in cell culture, destruction of infected cells and capacity to establish latent

infections primarily but not exclusively in sensory ganglia (Roizman & Pellett, 2001):

(i) members of the Alphaherpesvirinae show a variable host range, short reproduction

cycle, rapid spread in cell culture, efficient destruction of infected cells and capacity

to establish latent infections primarily, but not exclusively, in sensory ganglia; (ii) a

restrictive host range is rather characteristic for the members of the subfamily

Betaherpesvirinae. They have a long reproduction cycle, with a slow infection

progress in cell culture. Cytomegalia is frequently seen in infected cells and latency

can be found in secretory glands, lymphoreticular cells, kidneys and other tissues. (iii)

Finally, host range of the members of the Gammaherpesvirinae is limited to the

family or order to which the natural host belongs. Replication is followed by a lytic

infection and latency is frequently demonstrated in lymphoid tissue.

Equid herpesvirus 1 (EHV1) or equine abortion virus is a member of the order

Herpesvirales, Family Herpesviridae, subfamily Alphaherpesvirinae, genus

Varicellovirus (Davison et al., 2009).

1.2.2 Herpesviruses in equids

In equids, fourteen herpesviruses have been identified so far: six belong to the

subfamily Alphaherpesvirinae and eight to the Gammaherpesvirinae (Table 1). The

horse is the natural host to alphaherpesvirus types 1 (EHV1), 3 (EHV3, coital

exanthema virus), 4 (EHV4) and gammaherpesvirus types 2 (EHV2) and 5 (EHV5).

The donkey is the host of EHV1-homologue asinine herpesvirus type 3 (AHV3),

EHV3-homologue asinine herpesvirus type 1 (AHV1) and asinine gammaherpesvirus

type 2 (EHV2) (Browning et al., 1988; Crabb & Studdert, 1990; 1995; Ficorilli et al.,

1995). In 2002, two new herpesviruses (AHV4 and AHV5) were isolated from the

lungs of donkeys suffering from respiratory disease (Kleiboeker et al., 2002). The

isolation of a new EHV1-related neurotropic virus from a gazelle (Gazelle

herpesvirus- 1, EHV9) and an unknown herpesvirus from a donkey with neurological

Review of the literature

15

disease, indicates that the list of presently known equid herpesviruses is likely to grow

(Fukushi et al., 1997; Hartley et al., 1999; Vengust et al., 2008).

Table 1. Known herpesviruses of equids

Virus

Abbreviation Subfamily Host Clinical signs

Equine herpesvirus

1

EHV1 α horse respiratory disease, abortion, neonatal

foal death, neurological disorders

Equine herpesvirus

2

EHV2 γ horse rhinitis, conjunctivitis,

immunodepression

Equine herpesvirus

3

EHV3 α horse coital exanthema

Equine herpesvirus

4

EHV4 α horse respiratory disorders, (abortion)

Equine herpesvirus

5

EHV5 γ horse respiratory tract disease, multinodular

pulmonary fibrosis

Asinine herpesvirus

1

AHV1

(EHV6)

α donkey lesions on external genitalia and udder

Asinine herpesvirus

2

AHV2

(EHV7)

γ donkey ?

Asinine herpesvirus

3

AHV3

(EHV8)

α donkey rhinitis?

Asinine herpesvirus

4

AHV4 γ donkey pneumonia

Asinine herpesvirus

5

AHV5 γ donkey pneumonia

?

? γ donkey neurological disease

Zebra herpesvirus 1

ZHV1 γ Zebra ?

Wild ass

herpesvirus

WAHV γ wild

ass

?

Gazelle herpesvirus

9

EHV9 α gazelle encephalitis

Review of the literature

16

Of all equid herpesviruses, EHV1 and EHV4 are clinically, economically and

epidemiologically the most relevant pathogens. EHV1 infections can result in

respiratory disease, abortion, fatal neonatal illness and neurologic syndromes (Allen

& Bryans, 1986). In contrast to EHV1 infections, EHV4 infection is mostly limited to

signs of upper respiratory tract disease, while abortion and nervous system disorders

are extremely rare (Ostlund, 1993). EHV2 is suggested to be ubiquitous in the equine

population. EHV2 has been implicated in upper respiratory disease, pyrexia,

inappetance, lymphoadenopathy, immuno-suppression, general malaise and poor

performance, and a high prevalence of the virus could be found in horses with upper

respiratory tract disease, abortion and severe ataxia. However, a correlation between

clinical signs and EHV2 as a causative agent could not be determined (Borchers et al.,

1997; Borchers et al., 1998; Kershaw et al., 2001; Schlocker et al., 1995). EHV3

causes an acute and self-limiting venereal disease characterized by the formation of

papules, vesicles, ulcers, and pustules on genital mucosa and skin (Blanchard et al.,

1992; Pascoe et al., 1968). Little is known about the distribution and natural genesis

of EHV5. A relationship with respiratory tract disease is suspected and two recent

reports suggest an association between the development of equine multinodular

pulmonary fibrosis and EHV5 infection (Verryken et al., 2010; Williams et al., 2007;

Wong et al., 2008).

Review of the literature

17

2. EQUINE HERPESVIRUS 1 and the cell

2.1 Virus structure

Morphologically, herpesviruses are distinct from all other viruses. They contain a

linear double-stranded DNA genome of 125-290 kbp in the form of a torus (Furlong

et al., 1972). The DNA is contained within a icosadeltahedral capsid, which is

surrounded by a proteinaceous matrix, designated the tegument. The latter is

surrounded by a lipid envelope, containing membrane-associated proteins (Davison et

al., 2009; Roizman & Knipe, 2001; Roizman & Pellett, 2001) (Figure 1).

Figure 1. Electronmicroscopic photomicrograph (left) and schematic drawing (right) of an EHV1

virion (picture from University of Wisconsin)

Up till now, 12 different glycoproteins have been described for EHV1: gB, gC,

gD, gE, gG, gH, gI, gK, gL, gM, gN and gp2 (Turtinen & Allen, 1982). These

envelope proteins are involved in virus attachment, penetration, egress and cell-to-cell

spread.

Glycoprotein B is an essential glycoprotein of EHV1. Functional analysis of gB

revealed that this glycoprotein is clearly involved in the penetration process. In

common with gB homologues of herpes simplex virus and other herpesviruses, gB

also plays a role in the cell-cell fusion process (Wellington et al., 1996b). However,

gB-negative EHV1 retained some infectivity and the absolute requirement of gB for

Envelope glycoproteins

gB, gC, gD, gE, gG, gH,

gI, gK, gL, gM,

gN and gp2

Envelope

Tegument

Capsid

Double

stranded DNA

Review of the literature

18

fusion process in EHV1 is not entirely clear (Neubauer et al., 1997). Glycoprotein D

is the second essential protein of EHV1. Monoclonal antibodies against gD strongly

neutralize virus infectivity and inhibit the penetration of the virus into the cell,

suggesting an important and essential function of gD in viral cell penetration (Csellner

et al., 2000; Van de Walle et al., 2008a; Whittaker et al., 1992). Glycoprotein D is

also associated with spontaneous cell-cell fusion, which is consistent with a role in

virus spread and is one of the functions attributed to gD homologues in

alphaherpesviruses (Csellner et al., 2000; Wellington et al., 1996a).

Glycoprotein C was shown to play an important role in the early steps of infection

and in release of virions (Osterrieder, 1999). It also acts as a complement receptor on

EHV1-infected cells, capable of binding the third component of complement (C3)

(Huemer et al., 1995). Glycoprotein G is a viral chemokine-binding protein, capable

of binding a broad range of chemokines, including CCL3 and IL8, thus modulating

pulmonary inflammation and virus replication during an EHV1 infection (Bryant et

al., 2003; Van de Walle et al., 2007; Van de Walle et al., 2008b).

EHV1 glycoprotein M is involved in both virus entry and direct cell-to-cell spread

of virus (Osterrieder et al., 1996). Processing of gM inside the infected cell depends

on the expression of gN (Rudolph et al., 2002). Studies with an EHV1 recombinant

virus containing deletions of glycoprotein E and I showed that these glycoproteins

facilitate cell-to-cell spread and may be important factors in EHV1 virulence

(Matsumura et al., 1998). Glycoprotein K is also critically involved in direct cell-to-

cell spread, as well as in secondary envelopment and/or egress. It facilitates virus

penetration and is at least partially responsible for syncytium formation at later time

points post infection (Neubauer & Osterrieder, 2004).

Although the presence of gH and gL has been demonstrated in infected cells, their

functions in the replication cycle of EHV1 remain unclear (Robertson et al., 1991;

Stokes et al., 1996).

Last, gp2 is a unique glycoprotein that has no positional or structural counterpart

in herpesviruses of other species. Although its function is not fully known, it was

shown to function in virus attachment (Sun et al., 1996) and in vivo, gp2 is proposed

to function as an immuno-modulatory protein (Smith et al., 2005). The simultaneous

absence of gM and gp2 had an additive negative effect on egress but not on secondary

envelopment or cell-to-cell spread (Rudolph & Osterrieder, 2002).

Review of the literature

19

2.2 Replication cycle

The replication cycle of EHV1 is divided into several steps. A schematic

representation of the various steps is shown in Figure 2.

EHV1 attachment to cells is mediated via an interaction between glycoproteins

gB, gC and cellular heparan sulphate (Osterrieder, 1999). After attachment, an

interaction between gD and a putative entry receptor is required to complete the

fusion between the viral envelope and the cellular membrane (Csellner et al., 2000;

Frampton et al., 2005). Recently, it has been shown that Major Histocompatibility

Complex Class I acts as a functional entry receptor for this glycoprotein (Kurtz et al.,

2010; Sasaki et al., 2011). EHV1 can enter cells via fusion at the plasma membrane in

equine endothelial cells or by endocytosis/phagocytosis in peripheral blood

mononuclear cells (Van de Walle et al., 2008a). Successful infection through either

mechanism requires the activation of Rho-associated coiled-coil kinase 1 (ROCK1)

(Frampton et al., 2007). Recently, it has been shown that EHV1 entry via endocytosis

is triggered by the interaction between cellular integrins and the RSD motif present in

gD (Van de Walle et al., 2008a). Subsequent to fusion between the two lipid

membranes, the nucleocapsid is released into the cytoplasm of the infected cell, where

it is transported along microtubules to the nuclear pore of the cell. Transport is both

dependent on the integrity of the microtubule network and the minus-end microtubule

motor protein, dynein. Integrity is in part promoted by EHV1, as it actively induces

the acetylation of tubulin, a marker of microtubule stabilization. Also the cellular

kinase ROCK1 has an important role in virus trafficking to the nucleus (Frampton et

al., 2010). At the pore, the nucleocapsid releases its DNA into the nucleus, leaving an

empty capsid at the cytoplasmic side of the complex (Roizman & Pellett, 2001).

Transcription of the viral genome, replication of viral DNA and assembly of new

capsids take place in the nucleus, but all viral proteins are synthesized in the

cytoplasm. Viral DNA is transcribed throughout productive infection by host RNA

polymerase II, but with the participation of viral factors at all stages of infection.

EHV1 transcription is co-ordinately regulated into immediate-early (IE), early (E) and

late (L) phases in a cascade fashion (Caughman et al., 1985). Several of the gene

products are enzymes and DNA-binding proteins involved in viral DNA replication.

First, the immediate early gene, IR1, is expressed (Gray et al., 1987a; Gray et al.,

Review of the literature

20

1987b). This IE gene region is transcribed into a single mRNA, but multiple species

of the IE protein exist (Robertson et al., 1988). The major IE polypeptide (IE1)

functions as a regulatory protein that is capable of modulating gene expression from

both early and late promotors and also of trans-repressing its own promoter

(Caughman et al., 1985; Gray et al., 1987a). Early genes are expressed before DNA

replication and encode proteins required for DNA replication, e.g. DNA polymerase

and other enzymes such as thymidine kinase. These viral proteins promote viral DNA

replication, and in a late phase, expression of late genes is stimulated. The late genes

are expressed after viral DNA replication has started and their expression is enhanced

by other viral proteins. Late genes mostly encode viral structural components like

capsid proteins and envelope glycoproteins.

Like transcription, viral DNA replication takes place in the nucleus, resulting in

multiple copies of the EHV1 genome. These newly synthesized DNA copies have to

assemble with capsid proteins to form nucleocapsids. Upon translation and expression

in the cytoplasm, capsid proteins enter the nucleus, after which assembly and

incorporation of viral DNA molecules occur, a process called encapsidation.

Assembly occurs in several stages. DNA is packaged into preassembled capsids, after

which the virus matures and nucleocapsids bud at the inner leaflet of the nuclear

membrane, resulting in enveloped particles in the perinuclear space. Egress from the

perinuclear cisterna primarily occurs by fusion of the primary envelope with the outer

leaflet of the nuclear membrane, resulting in intracytoplasmic naked nucleocapsids

(Granzow et al., 2001; Mettenleiter, 2004; Mettenleiter et al., 2006). Inside the

cytoplasm, the majority of the tegument is added to the nucleocapsids (Mettenleiter,

2002; Mettenleiter et al., 2009). Unlike capsid proteins, viral envelope proteins are

co-translationally transported into the endoplasmatic reticulum (ER) and subsequently

incorporated into various cellular membranes while passing through the secretory

pathway (Granzow et al., 2001). The intracytoplasmic naked nucleocapsids are

enveloped at membranes of the trans-Golgi network (TGN) (Granzow et al., 2001).

The enveloped particles are then released through secretory vesicles. Little is known

about viral proteins involved in egress, i.e. transport of vesicles containing enveloped

virions to the plasma membrane and subsequent virus release by fusion of vesicle and

plasma membrane (Mettenleiter et al., 2009).

Review of the literature

21

Figure 2. The EHV1 replication cycle (adapted from Garré, 2008). The first step in the replication of EHV1

is the attachment of free virions to the surface of the target cell (1). After endocytosis (2a) and/or fusion of

the viral envelope with the plasma membrane (2b), the nucleocapsid is released into the cytoplasm (3) and

transported into the nucleus (4). DNA replication (5) and transcription (6) occur and RNA molecules are

transported to the cytoplasm and translated to proteins (7, 9). Capsid proteins are redirected into the nucleus

where encapsidation occurs (8). Viral envelope proteins are co-translationally transported into the

endoplasmatic reticulum (10) and subsequently incorporated into various cellular membranes, while passing

through the secretory pathway. The nucleocapsid leaves the nucleus via budding through the inner leaflet of

the nuclear membrane (11) and subsequent fusion with the outer leaflet of the nuclear membrane, resulting

in entry of naked nucleocapsids in the cytoplasm (12). The nucleocapsid acquires its secondary envelope at

the Golgi apparatus (13) and then leaves the cell via vesicle-mediated exocytosis (14, 15, 16)

2a

2b

3

4

2a

2b

3

4

Review of the literature

22

3. EQUINE HERPESVIRUS 1 and the horse

3.1 Epidemiology and prevalence

A schematic overview of the epidemiology of EHV1 is depicted in Figure 3

Equine herpesvirus 1 infections are endemic in horse populations worldwide, and

most animals are probably infected during the first year of life (Allen & Bryans, 1986;

Allen et al., 1999; Borchers et al., 2006). However, due to antigenic similarity

between EHV1 and EHV4, the interpretation of data from serological surveys was

complicated by the lack of a type-specific antibody test until early 1990 (Crabb &

Studdert, 1993). It should also be indicated that sero-epidemiological studies are

complicated in horse populations in which EHV1 and/or EHV4 vaccination is

practised. Retrospective testing of sera from Thoroughbred horses collected between

1967-1974 and 1993 gave values of 9-28% for EHV1-specific antibodies (Crabb &

Studdert, 1995). Another study found an incidence of EHV1 antibodies in mares and

foals of 26,2% and 11,4% respectively (Gilkerson et al., 1999).

Latency by alphaherpesviruses is an important epidemiological strategy ensuring

survival and spread within the natural host population. When horses are first infected,

latency is established in the lymph nodes of the respiratory tract, the trigeminal

ganglion and CD5+/CD8

+ leukocytes (Baxi et al., 1995; Chesters et al., 1997;

Edington et al., 1994; Slater et al., 1994). Months or years after primary infection, the

latent virus may again become manifest with renewed replication and with the

potential for initiating new outbreaks of disease in its host as well as in susceptible

cohorts. As EHV1 infection already occurs in the first weeks or months of life, it is

assumed that viral reactivation in latently infected mares leads to this early foal

infection (Gilkerson et al., 1999; Marenzoni et al., 2008). Therefore, lactating mares

may be the primary source of EHV1 infection of foals and the latter may further

transmit the virus to other mares and foals (Gilkerson et al., 1997). However, a recent

study could not demonstrate infection of sentinel horses with EHV1 after direct

contact with horses undergoing experimental corticosteroid-induced recrudescence of

latent infection (Pusterla et al., 2010). Failure to effectively transmit EHV1 to

Review of the literature

23

susceptible horses may have resulted from the low level and short period of viral

shedding in these latently infected horses.

Estimates of the prevalence of EHV1 infection based on viral detection

technologies vary strongly. Latent EHV1 could be found by polymerase chain

reaction (PCR) in 87.5% of examined bronchial lymph nodes and a similar level in the

trigeminal ganglia of 40 horses examined at slaughter (Edington et al., 1994). In pools

of tissue with peripheral blood leukocytes, nasal swabs and abattoir specimens from

116 animals, EHV1 DNA was found in 88% of the analysed samples in Brazil

(Carvalho et al., 2000). In a recent study, the prevalence of latent EHV1 in

submandibular lymph nodes was 54% (Allen et al., 2008). Current estimates of the

prevalence of latent EHV1 suggest a rate in excess of 60%, which is probably more

limited by detection technology than by the actual infection rate. We should presume

that the majority of horses is latently infected with EHV1 (Lunn et al., 2009). The

establishment of latency provides a permanent reservoir for the virus and makes total

disease eradication highly unlikely. It also diminishes the effectiveness of antiviral

therapy or immunotherapy and complicates vaccination protocols for control of EHV1

infections (Allen & Bryans, 1986).

It is thought that subclinical EHV1 infections are common in horses and play an

important role in EHV1 epidemiology, as they result in frequent spread and a high

risk of exposure particularly in open horse populations subjected to stress and

introduction of new animals. However, recent reports in both horses exposed to stress

and healthy horses demonstrated that subclinical shedding of EHV1 is infrequent and

at a very low level (Brown et al., 2007; Wang et al., 2007). These observations

suggest that spread of EHV1 among adult horses is typically accompanied by clinical

disease, either respiratory disorders, abortion or equine herpes myelitis (EHM) (Lunn

et al., 2009).

Either after recrudescence from latency or after exogenous introduction, EHV1

infections can spread rapidly within groups of horses. Evidence of widespread

horizontal infection by direct or indirect contact has been reported repeatedly for

outbreaks of EHV1 induced abortion and neurological disease (Drummer et al., 1995;

Friday et al., 2000; McCartan et al., 1995). Aerosol transmission cannot be excluded

and putatively depends on the amount of infectious virus released by the source,

Review of the literature

24

climatic conditions, dilution of the aerosolized virus, and distance between horses

(van Maanen, 2002).

Figure 3. Epidemiology of EHV1 infection in the horse population

Reactivation

of latent virus

Subclinical disease

Respiratory disease

Abortion/neonatal disease

Nervous system disorders

Virus shedding

Mare to foal Horse to horse

on same premise

Other horses outside premise

Other premise

Exogenous

introduction

of virus

Stress factor

Reactivation

of latent virus

Subclinical disease

Respiratory disease

Abortion/neonatal disease

Nervous system disorders

Virus shedding

Mare to foal Horse to horse

on same premise

Other horses outside premise

Other premise

Subclinical disease

Respiratory disease

Abortion/neonatal disease

Nervous system disorders

Virus shedding

Mare to foal Horse to horse

on same premise

Mare to foal Horse to horse

on same premise

Other horses outside premise

Other premise

Exogenous

introduction

of virus

Stress factorExogenous

introduction

of virus

Stress factor

Review of the literature

25

3.2 Pathogenesis and disease

3.2.1 Pathogenesis

A schematic overview of the complete pathogenesis is depicted in Figure 4.

Primary replication. Horses acquire an EHV1-infection through direct contact or

inhalation of infectious secretions (Allen & Bryans, 1986). After inhalation, EHV1

binds to and penetrates the epithelial cells of the upper and lower respiratory tract

(Kydd et al., 1994b; Patel et al., 1982). Following a rapid replication, EHV1

disseminates into the connective tissue, infecting local blood vessels and regional

lymph nodes (Kydd et al., 1994b; van Maanen, 2002). How the virus penetrates the

basement membrane and reaches the underlying connective tissue and lymphoid

tissue remains unsolved to date.

Viremia. Via a still unknown pathway, EHV1 infects mononuclear cells which

can enter the blood circulation resulting in a cell-associated viremia lasting for 8 to 18

days post infection (Edington et al., 1986; Gibson et al., 1992; Gleeson & Coggins,

1980; Patel et al., 1982). The exact identity of these carrier cells still remains

uncertain. A study from 1983 showed that T-lymphocytes, B-lymphocytes and

monocytes are all able to harbour EHV1, but the predominant mononuclear cells

harbouring virus were found to be T-lymphocytes (Scott et al., 1983). Remarkably,

after disruption of these cells, no infectious EHV1 could be found during virus

isolation, suggesting that EHV1 exists in mononuclear cells in a non-infective form,

or is blocked at a certain replication level until activation occurs (Gleeson & Coggins,

1980; Scott et al., 1983; van der Meulen et al., 2006). A later study confirmed the

latter hypothesis. The majority of infected PBMC do not show viral envelope proteins

on their surface during viremia (van der Meulen et al., 2003b). During a recent study

using PBMC isolated from experimentally infected horses, viral genomic DNA was

found by means of PCR techniques in all PBMC subpopulations, with CD8+

lymphocytes and B-lymphocytes as most frequently infected cells (Wilsterman et al.,

2010). These results indicate that different PBMC subpopulations may play different

roles in EHV1 viremia.

Review of the literature

26

Secondary replication. Transfer of EHV1 from viremic cells to endothelial cells

of target organs causes infection, followed by replication of EHV1 and subsequent

damage to these organs. Smith et al. showed that activation of adhesion molecules in

endothelial cells of the nasal mucosa and the reproductive tract is a key step in

transferring virus from infected leukocytes to endothelial cells. Indeed, an in vitro

flow system using equine veins and arteries showed that EHV1 was only transferred

to endothelial cells if both leukocytes and endothelial cells expressed these surface

molecules (Smith et al., 2002; Smith et al., 2001). An infection model with EHV1-

infected leukocytes on the one hand and carotid artery and brain endothelial cells on

the other hand has recently been established (Goehring et al., 2010a). In this model,

neutralizing antibodies could only prevent infection of endothelial cells when an

indirect contact model was used. In the case of a direct contact model between the two

cell types, neutralizing antibodies could not prevent transfer of virus to endothelial

cells. This further confirms the importance of the direct transfer mechanism of EHV1

from infected leukocytes to endothelial cells.

Transfer of EHV1 from infected leukocytes to placental maternal endothelial cells

plays a major role in the pathogenesis of abortion. Infection of endothelial cells in the

umbilical cord seems to be the primary route of dissemination of virus in the fetus.

These infected cells may then act as centres for the development of focal lesions of

parenchymal necrosis. Further spread of the virus with widespread vasculitis,

thrombosis, cotyledonary infections and ischemic damage to the endometrium with

replication of EHV1 in foetal tissues leads to subsequent abortion (Edington et al.,

1991).

Review of the literature

27

Figure 4. Schematic overview of the pathogenesis of EHV1. 1) Attachment/penetration and replication

of virus in epithelial surfaces of the upper respiratory tract; 2) spread of virus through the basement

membrane; 3) spread to connective tissue (3a) and draining lymph nodes (3b); 4) infection of

leukocytes; 5) leukocyte associated viremia; 6) transfer of virus from infected leukocytes to endothelial

cells of target organs; 7) replication and spread of virus in target organs. Full arrow: known

mechanism; striped arrow: unknown mechanism.

Secondary replication of EHV1 in the nervous system of horses causes EHM.

Different independent researchers agreed that lesions accompanying central nervous

system disorders were characterized by a focal vasculitis with associated oedema,

haemorrhage and thrombosis. Axonal and myelin degeneration occurred focally and

secondary to these vascular changes (Jackson et al., 1977; Mumford & Edington,

1980; Platt et al., 1980). As actual isolation of EHV1 from the nervous system is very

rare, it was postulated that lesions could be induced by circulation immune

complexes. However, in 1986, Edington et al. showed that although recovery of virus

1

3a

45

2

3b

67

Review of the literature

28

from the central nervous system (CNS) was low, immunofluorescence indicated that

endothelial tropism of the virus was the initiating and persisting feature. The action of

immune complexes in EHV1 infections seems to be secondary and localized

(Edington et al., 1986). In contrast to several other alphaherpesviruses, which can

cause encephalitis through primary neurotropism, EHV1 seems to be non-neurotropic

and its propensity to induce myeloencephalopathy reflects a marked

endotheliotropism (Edington et al., 1986; Jackson et al., 1977; Whitwell & Blunden,

1992; Wilson, 1997). Vascular lesions result in secondary hypoxic degeneration of

nervous tissue, causing nervous system disorders due to the impairment of blood flow

and metabolic exchange.

Abortion versus nervous system disorders. For many years, it has been

suspected that distinct isolates of EHV1 might be responsible for outbreaks of either

abortion and neonatal foal death or neurological disease. The difference in pathogenic

potential of EHV1 strains to induce abortion or neurological damage appears to be

correlated with differences in the ability to establish infection at vascular endothelial

sites, in particular within the endometrium and central nervous system (Edington et

al., 1986; Mumford et al., 1994; Patel et al., 1982; Platt et al., 1980). Infection with

neurovirulent strains may result in severe clinical disease, including a high frequency

of abortion and a less frequent occurrence of paralysis, while infection with non-

neurovirulent strains is restricted to mild respiratory signs, with infrequent induction

of abortion and no neurological disease (Mumford et al., 1994; Smith et al., 1992;

Smith et al., 2000). It was noticed that horses inoculated with neurovirulent strains of

EHV1 developed both a greater magnitude and duration of PBMC-associated viremia

than animals inoculated with non-neurovirulent strains of the virus, which contributes

to the risk for development of neurological signs (Allen & Breathnach, 2006).

Recently, it was demonstrated that a variation of a single amino acid is strongly

associated with neurological versus non-neurological disease outbreaks (Nugent et al.,

2006). This study compared different isolates recovered from neurological and non-

neurological outbreaks over the course of 30 years. It was found that a single

nucleotide polymorphism (SNP) in the catalytic subunit of the viral DNA polymerase

(pol), causing a substitution of asparagine (N) by aspartic acid (D) at amino acid

position 752 at the ORF30 position of the DNA polymerase, was a major contributory

Review of the literature

29

factor in determining whether a disease outbreak will include animals presenting

neurological signs. To strengthen the finding that the single amino acid exchange by

itself influences neurovirulence, several studies have been performed, using

recombinant viruses of non-neurological EHV1 strains with an inserted point

mutation for the neurological variant and vice versa, further confirming the previous

finding (Goodman et al., 2007; Van de Walle et al., 2009). However, horses with

neurological disease upon infection with a non-neurovirulent EHV1 isolate, as well as

a high prevalence of neurovirulent strains in equine abortions have been reported,

suggesting that other factors such as host and/or environmental factors will also

contribute to the onset of neurological disease (Fritsche & Borchers, 2010; Goehring

et al., 2006; Perkins et al., 2009; Pronost et al., 2010; Smith et al., 2010).

In recent years, outbreaks of neurological disease due to EHV1 infections have

been reported with increasing frequency and severity (Friday et al., 2000; Lunn et al.,

2009; McCartan et al., 1995; USDA, 2007; van Maanen et al., 2001). Unfortunately,

despite 40 years of worldwide research, EHV1-induced neurological disease cannot

be adequately prevented nor fully explained.

3.2.2 Symptoms

An overview of different symptoms following EHV1 infection, is shown in Figure 5.

Respiratory disorders. Primary replication of EHV1 in tissues of the upper

respiratory tract is mostly accompanied with signs of a mild, self-limiting upper

respiratory tract infection (Gibson et al., 1992). Infected horses spread large amounts

of virus via nasal secretions, allowing the virus to spread to other susceptible animals.

Infectious virus can be detected from day 1 till 7 to 14 days post infection (Gibson et

al., 1992; Heldens et al., 2001; van der Meulen et al., 2006) and in some cases even

till 21 days post infection (Garré et al., 2009; Patel et al., 1982).

Young horses usually develop fever, serous to mucopurulent nasal discharge and

swelling of draining lymph nodes, while older horses mainly show mild or subclinical

disease (Coggins, 1979). Conjunctivitis, depression and anorexia can be observed in

some cases (Edington et al., 1986; Kydd et al., 1994a; Patel et al., 1982).

Review of the literature

30

In sporadically reported cases, EHV1 targets the pulmonary endothelium in young

adults, which causes a severe pulmonary oedema (Del Piero & Wilkins, 2001; Del

Piero et al., 2000). Such severely affected horses die in acute respiratory distress.

Abortion and neonatal foal death. Secondary replication of EHV1 in endothelial

cells of the pregnant uterus plays a major role in the occurrence of EHV1-induced

abortion or neonatal death (Edington et al., 1991). The incubation period for abortion

after respiratory infection with EHV1 varies from 6 days (Gleeson & Coggins, 1980)

to 4 months (Mumford et al., 1987). Virologically positive foetuses, including

placenta, are often fresh and abortion occurs suddenly and spontaneously. However,

abortions with virological negative foetuses are also described (Gleeson & Coggins,

1980; Smith et al., 1992).

EHV1-induced abortion can be preceded by mild upper respiratory tract disease in

the mare but usually no signs of disease or warning signs of impending abortion are

noticed. Following abortion, virus is cleared rapidly from the genital tract, and future

breeding capacity is not impaired unless some uterine damage has occurred due to

dystocia (Smith, 1997). Although mares are not immune to subsequent infection,

EHV1-induced abortions in successive pregnancies are rare (Doll et al., 1955;

Ostlund, 1993). EHV1-induced abortions usually involve only 1 or 2 mares in a herd,

although sometimes abortion storms can occur in which more than 50 % of the foals

are lost (Mumford et al., 1987; Van Maanen et al., 2000).

If infection occurs close to term, the mare may deliver an apparently normal or

weak living foal. These foals usually develop jaundice and respiratory distress and

mostly die within 7 days post partum despite of intensive treatment, due to extensive

viral damage and secondary bacterial infection (Murray et al., 1998; Perkins et al.,

1999).

Field studies have shown that 95% of EHV1-induced abortions occur during the

last 4 months of gestation (Allen & Bryans, 1986; Doll, 1952; Doll & Bryans, 1963).

EHV1-abortion has been recorded on rare occasions as early as the fourth month of

gestation (Prickett, 1970). This apparent resistance of the early pregnant mare to

abortion is not completely understood.

Review of the literature

31

Nervous system disorders.

Secondary replication of EHV1 in the endothelial cells of blood vessels of the

nervous system is the first step in the development of nervous system disorders

(Edington et al., 1986). Neurological disease may affect either single horses or groups

of animals within a herd. An incubation period of 6 to 10 days between initial

infection and the occurrence of nervous system disorders has been described (Jackson

et al., 1977; van Maanen et al., 2001).

Although the disorders may be preceded by signs of respiratory disease such as

fever or nasal discharge one or two weeks earlier, they can also occur without any

previous signs (Carroll & Westbury, 1985; Greenwood & Simson, 1980; Stierstorfer

et al., 2002; van Maanen et al., 2001). Neurological disease is usually acute in onset

and can display a variable set of clinical signs. Most commonly, signs vary from mild

ataxia to complete paralysis, accompanied by bladder and/or rectal paralysis (Jackson

et al., 1977; Stierstorfer et al., 2002). In less common cases, oedema of the hind legs,

swollen testis, urticaria and central nervous disorders such as blindness and torticollis

can be observed (Friday et al., 2000; Greenwood & Simson, 1980; van der Meulen et

al., 2003a). The prognosis for horses with ataxia due to EHV1 myeloencephalitis is

favourable, with the possibility to regain their normal performance level. However, if

the horse becomes recumbent for more than 24 hours, the prognosis is grave and

euthanasia is indicated (McCartan et al., 1995; van Maanen et al., 2001).

Although animals of various age, gender and breed can be affected, a retrospective

study of Goehring et al. (2006) indicated that certain pony breeds and females were

more commonly affected. Although neurological problems have been reported in

weanlings, the clinical picture in young horses seems to be less severe than in adults,

suggesting age to be an important risk factor as well (Goehring et al., 2006;

Greenwood & Simson, 1980).

Review of the literature

32

Figure 5. Different EHV1-related symptoms. A: respiratory disorders; B: abortion; C: neonatal foal

disease; D: nervous system disorders

3.2.3 Treatment

Antiviral therapy. Treatment of EHV1 infections is limited to symptomatic care

since no specific antiviral drugs are available for treatment of EHV1 infections in

horses to date. Experimental antiviral drugs have yet to prove clinical efficacy and

several of them have been tested in field as well as under experimental conditions.

During several natural outbreaks of EHV1, treatment with the antiviral drug

acyclovir has been reported with conflicting results (Friday et al., 2000; Henninger et

al., 2007; van der Meulen et al., 2003a). However, since therapeutic evaluations in

case-control studies under field conditions are difficult due to a lack of good control

animals, great care should be taken in interpreting these results. Acyclovir was also

shown to have serious limitations due to poor oral bioavailability. Therefore, high

therapeutic concentrations are needed in order to reach the desired effect, making this

drug a costly medication with questionable efficacy (Garré et al., 2009; Garré et al.,

A

C

B

D

Review of the literature

33

2007; Maxwell et al., 2008a). When using the pro-drug valacyclovir, sufficiently high

acyclovir levels could be reached in plasma and nasal mucus after oral treatment, but

no effect of this treatment was seen on clinical signs, viral shedding and viremia of

EHV1-infected ponies (Garré et al., 2009). The latter was in contrast to the results of

another study, showing that oral treatment with valacyclovir significantly decreased

signs of EHV1 disease (Maxwell et al., 2008b). The protective effect was greatest

when therapy was initiated before viral infection and continued for two weeks.

However, initiation of therapy at the onset of fever also decreased sings of disease as

compared to control horses. These encouraging results should be viewed cautiously in

light of the small number of horses studied.

Recently, synthetic siRNAs against envelope glycoprotein B and the origin-

binding protein helicase were designed, and their efficacy to limit and prevent EHV1

infection was tested in vitro and in a murine model. The results obtained during this

study, indicated that siRNA treatment could be of great importance during an

outbreak of EHV1 in a horse population, as not only the severity of clinical disease

but also the number of affected animals and the magnitude of nasal shedding could be

reduced (Fulton et al., 2009). Although there was no significant difference in viral

shedding and viremia between treated and control groups during an in vivo

experiment with EHV1-infected horses, euthanasia necessitated in case of

neurological disease was significantly reduced after application of EHV1 specific

siRNA (Brosnahan et al., 2009).

Supportive therapy. Despite many experimental approaches, no systemic

antiviral therapeutic agents are approved for the use in horses or have a proven

efficacy. Therefore, supportive therapy is of paramount importance in the field.

Treatment strategies are aimed at easing clinical signs and preventing secondary

complications. The level of care necessary is dependent on the severity of clinical

signs.

Respiratory infections of immunocompetent animals are generally self-limiting

and require only good nursing care. Broad-spectrum antibiotics can be administered to

assist in combating secondary bacterial infections and high fever can be treated with

antipyretics. Stress avoidance is paramount and horses in training should receive

adequate rest before resuming work (Ostlund, 1993; van Maanen, 2002).

Review of the literature

34

Abortion occurs suddenly with complete expulsion of fetus and placenta and

complications are seldom. Therefore, after an uncomplicated partus, usually no

therapy for the aborting mare is required (Ostlund, 1993; van Maanen, 2002).

However, it is advisable to wash tail and hind quarters with disinfectants to avoid

further viral contamination of the environment and/or other animals (Goulden et al.,

1989).

Weak, but live born affected foals or, less frequently, foals that initially appear

healthy but rapidly deteriorate due to intractable bacterial infections should be treated

with intensive therapy like provision of warmth, oxygen, cardiovascular and

nutritional support and appropriate drugs (Murray et al., 1998; van Maanen, 2002).

However, these measures are almost always unsuccessful, as these foals are almost

always bound to die within a few days due to extensive damage in parenchymal and

lymphoreticular organs and subsequent bacterial infections (Allen & Bryans, 1986;

Goulden et al., 1989).

Animals showing nervous system disorders should receive appropriate intensive

care. Non-recumbent animals should be housed in a deeply bedded box, until

coordination improves. Severely ataxic horses may be temporarily assisted by a sling

to prevent secondary complications associated with recumbency. Regular urinary

catheterization and rectum evacuation may be required in case of bladder or rectum

paralysis (Friday et al., 2000; McCartan et al., 1995). Horses which are able to stand,

should be turned out or hand walked as soon as possible (Greenwood & Simson,

1980; McCartan et al., 1995; van Maanen et al., 2001). Recumbent animals not able

to be supported by a sling should be maintained in a sternal position in a well-bedded

stable if possible. They should have easy access to food and water. Intravenous

rehydratation may be necessary for horses unable to remain in sternal position. Horses

in lateral recumbency should be manually rotated every few hours. Antibiotics are

indicated because of the higher risk of developing aspiration pneumonia and/or

decubital ulcers. Horses can also be treated with corticosteroids in order to reduce the

inflammatory response in the nervous system. However, considerable care should be

taken when treating horses with such drugs during an outbreak, as they induce

immunosuppression and may stimulate a longer and higher spread of virus which can

make them even contra-indicated (Edington et al., 1985; Goehring & Sloet van

Oldruitenborgh-Oosterbaan, 2001; Slater et al., 1994). Therefore, only short-acting

Review of the literature

35

corticosteroids, if at all, should be used, for a limited period (McCartan et al., 1995;

Reed & Toribio, 2004). Other anti-inflammatory drugs such as dimethyl sulfoxide

(DMSO) and non-steroidal anti-inflammatory drugs can be used, although their

capacity to inhibit the development of the lesions of EHM is unknown. Recovery of

non-recumbent animals is usually complete, although improvement is gradual and

several months may elapse before maximal improvement (Ostlund, 1993). However,

when a horse is recumbent for more then 24 hours, the prognosis is grave and

euthanasia may be indicated (van Maanen et al., 2001).

3.3 Diagnosis

Virus culture and isolation are considered the gold standard test for the laboratory

diagnosis of EHV1 (Lunn et al., 2009). Virus culture, isolation and identification of

EHV1 is strongly supportive of a diagnosis of EHV1-induced disease in a horse with

compatible clinical signs. The choice of sample depends on the disease syndrome

encountered (Ostlund, 1993). Both nasal and nasopharyngeal swabs can be collected

during the first phase of infection or in case of respiratory symptoms. A recent study

showed that nasal swabbing represents a viable alternative to the less well tolerated

nasopharyngeal swabbing technique for the molecular detection of EHV1 in horses

(Pusterla et al., 2008a). Heparinized whole blood can also be collected at this stage, as

viremia, associated with the buffy coat fraction, often coincides with clinical signs of

respiratory disease, abortion and neurologic disease. In case of abortion, virus can be

isolated from different foetal tissues. Viral culture and positive identification can be

accomplished in as little as 2-3 days in a laboratory when the sample contains a high

viral load. However, virus isolation is often unsuccessful during outbreaks, because

the peak of virus shedding may already have passed and local antibodies may interfere

(Mumford, 1984; van Maanen et al., 2001). The likelihood of detecting EHV1 during

outbreaks of disease can be increased by testing in-contact horses, especially during

episodes of fever.

Recently, PCR techniques have become more and more common as a diagnostic

test because of their high analytical sensitivity and specificity in case of low viral load

(Sharma et al., 1992; van Maanen et al., 2001; Varrasso et al., 2001). Conventional

PCR, however, is limited in its sensitivity, because no distinction between the

Review of the literature

36

presence of infectious virus and latent infection can be made. The random testing or

screening of healthy horses for EHV1 by conventional PCR should therefore be

avoided (Lunn et al., 2009). The use of novel EHV1 PCR platforms, such as real time

PCR, allows for a more sensitive detection, a greater specificity and calculation of

viral loads (Elia et al., 2006; Hussey et al., 2006). Determination of viral load can

offer important advantages as it can allow for better characterization of disease stage,

assessment of risk of exposure to other horses and monitoring of response to treatment

(Pusterla et al., 2009b; Pusterla et al., 2008b; Pusterla et al., 2009c). Concerning the

circulation of neurovirulent and non-neurovirulent strains of EHV1 in the field, real-

time PCR tests have been developed that distinguish between these two biovars

(Allen, 2007; Pusterla et al., 2009c).

Serologic testing demonstrating a 4-fold or greater increase in serum antibody titre

by serum-neutralizing (SN) or complement-fixation tests on acute and convalescent

samples collected 3 weeks apart, provides presumptive evidence of EHV1 infection

(Kydd et al., 2006). Although serologic testing has limitations in confirming a

diagnosis of EHV1 infection in an individual horse, testing of paired serum samples

from in-contact horses is recommended. However, serologic testing is limited due to

cross-reactivity between EHV1 and EHV4, and most laboratories cannot distinguish

these two viruses (Hartley et al., 2005). Therefore, a specific ELISA test based on the

C-terminal portion of glycoprotein G of both viruses has been developed (Drummer et

al., 1995).

Cerebrospinal fluid analysis with the presence of xanthochromia, a yellowish

appearance of cerebrospinal fluid, is often supportive of an EHM diagnosis. In

addition, increased protein concentrations with or without a monocytic pleocytosis are

typical findings. Histopathology on brain and spinal cord, with typical vasculitis and

thrombosis of small blood vessels, is an essential method for confirming EHV1

infection in a horse with suspected EHM post mortem (Schultheiss et al., 1997).

3.4 Disease control

Vaccination. EHV1 remains a leading cause of upper respiratory tract infection,

viral abortion and nervous system disorders in horses worldwide, despite the

customary use of commercial vaccines (Allen et al., 1999; Kydd et al., 2006).

Review of the literature

37

Following natural infection, EHV1 efficiently induces a local mucosal humoral

response, which offers the potential for immune exclusion of virus at the respiratory

tract epithelium portal of entry. However, neither inactivated, nor attenuated vaccines

are able to induce detectable mucosal antibodies (Breathnach et al., 2001). The

currently available vaccines are not able to induce a significant level of protection

against EHV1-induced viremia. However, several vaccines are able to induce a

significant level of protection against either abortion or nervous system disorders (van

der Meulen et al., 2007).

The purpose of vaccination is twofold. First, vaccination is meant to minimize

virus replication in the respiratory tract upon infection, thus limiting nasal shedding

and the occurrence of respiratory disorders. Second, vaccination should prevent the

occurrence of abortion and/or nervous system disorders. The first purpose of

vaccination seems to be fulfilled by the available vaccines, as demonstrated by

reduced nasal virus titres and the reduced severity of respiratory disease upon

challenge infection of vaccinated horses (Burrows et al., 1984; Goodman et al., 2006;

Heldens et al., 2001). However, the second purpose to protect horses against EHV1-

induced viremia and subsequent abortion or neurological disorders is highly variable

and none of them can guarantee 100% protection. In view of this, we will briefly

review several vaccination/challenge studies performed with commercially available

vaccines.

Many studies have addressed the potential of inactivated vaccines in the battle

against EHV1. Inactivated virus vaccines can stimulate high titres of serum virus

neutralizing (VN) antibody, which cannot only reduce the amount and duration of

virus shedding but also prime the mucosal compartment (Breathnach et al., 2001).

However, they fail to induce cytotoxic T-leukocyte (CTL) responses which have

proven to be of paramount importance to limit EHV1 infection (Kydd et al., 2003;

Minke et al., 2004). Several case-control studies have been performed with the

commercially available vaccine Duvaxyn EHV1,4® (Foote et al., 2002; Heldens et al.,

2001). This inactivated vaccine clearly reduced clinical symptoms, the duration of

virus shedding and the quantity of virus shed. Additionally, a significant reduction in

the occurrence of abortion was noticed. However, vaccination under field conditions

could not prevent continuous circulation of both EHV1 and EHV4 in vaccinated

populations (Foote et al., 2006). Two other studies were performed with another

Review of the literature

38

inactivated and oil-adjuvanted vaccine Pneumabort K®. Neither of these studies could

demonstrate a difference in febrile responses, clinical signs and subsequent abortion

rates between vaccinated and control mares (Bürki et al., 1990; Burrows et al., 1984).

However, a recent study with the latter vaccine did not only show a significant

reduction of clinical signs and nasal shedding after three vaccinations, but also

demonstrated a significant reduction in the number of days of viremia (Goehring et

al., 2010c). A possible influence on abortion or nervous system disorders was not

evaluated during this study.

Modified-live vaccines (MLV) probably stimulate CTL responses and prime the

animals for mucosal antibodies, making them valuable candidates to limit EHV1

infections. They induce a rapid onset of immunity which is both broad and long

lasting. However, care should be taken when using these vaccines as reversion to

virulence remains a possible risk (Minke et al., 2004). A study of Bürki et al. (1990)

showed that all horses became infected and developed viremia upon challenge after

vaccination with the MLV Prevaccinol® (Bürki et al., 1990). Two out of 4 mares

aborted, but the study lacked a control group of non-vaccinated mares. Another study

compared a commercially available inactivated vaccine (Flu-vac Innovator 6®) and a

commercially available modified-live vaccine (Rhinomune®) (Goodman et al., 2006).

The modified-live vaccine induced significantly lower VN antibodies, suggesting a

bias towards a cytotoxic immune response and virus shedding was significantly lower.

Although the lymphocyte viral gene copies were similar, viremia lasted for a shorter

period after vaccination with Rhinomune®. Upon challenge, none of the horses

showed nervous system disorders, in contrast to the horses vaccinated with Flu-vac

Innovator 6® (3/5) and the control group (3/5). Also a recent study showed a great

decrease in clinical symptoms and nasal shedding after vaccination with Rhinomune®.

However a decrease in duration of viremia could not be noticed (Goehring et al.,

2010c).

Other alternative approaches for potential vaccination against EHV1-induced

disease such as DNA vaccines, live-vectored vaccines and gene-deleted mutants are

not yet commercially available, but have undergone pilot studies. A study of Soboll et

al. (2006) could demonstrate a limited immune response and protection following

DNA vaccination with plasmids encoding gB, gC, gD, IE and early proteins of EHV1

(Soboll et al., 2006). A recent study showed that a vaccine with the IE gene of EHV1

Review of the literature

39

in a recombinant modified-live vaccinia vector provides not only a reduction in

clinical disease, but also a reduction of cell-associated viremia (Soboll et al., 2010).

Unfortunately, until now, the currently available vaccines do not provide complete

protection as they especially stimulate high titres of circulating antibodies and are

unlikely to stimulate cytotoxic effector lymphocytes. As EHV1-induced protective

immunity is only short-lived, vaccination has to be repeated every 6 months (Kydd et

al., 2006). Future vaccination strategies should be aimed at stimulating both CD8+ and

CD4+ elements of cell-mediated immunity, in particular CTLs, but at the same time

maximise stimulation of mucosal and plasma VN antibody (Kydd et al., 2006).

Routine management. The overall goals in management of EHV1 infections are

to minimize exposure of horses to exogenous virus, to maximize immune

preparedness to forestall disease in the event of exposure, and to decrease the

likelyhood of latent virus recrudescence under influence of stress (Ostlund, 1993; van

der Meulen et al., 2007). Management has to be concentrated mainly on prevention of

the more serious sequelae of an EHV1 infection like abortion storms and outbreaks of

neurological disease (van Maanen, 2002).

Retrospective analysis of equine herpesviral disease outbreaks often can be linked

to a source of exogenous virus. Age segregation of horses on a farm is one of the best

tactics to limit spread of herpesviruses. In particular, pregnant mares should be

separated from young stock and transient horses. Ideally, the pregnant mare

population should be routinely subdivided into small groups according to expected

delivery dates (Ostlund, 1993). Personnel should handle pregnant mares before

contacting other horses on the premises. New horses should be isolated for a period of

3 weeks, with daily monitoring of rectal temperature and general health, before

mixing with the resident population (Allen & Bryans, 1986; Ostlund, 1993).

Vaccination programs should be instituted on a farm-wide basis and should

include adult horses and non-breeding animals as well as young stock and pregnant

mares (Ostlund, 1993) (see chapter 3.4 Disease control, vaccination).

Although some stress is unavoidable, efforts can be made to avoid unnecessary

stress and to be diligent in routine disease surveillance among animals known to be

recently subjected to stress (Ostlund, 1993). Stress factors such as poor feeding

Review of the literature

40

conditions, overcrowding, travelling in late pregnancy and separation from mates

should be minimized (van Maanen, 2002).

Management in case of an outbreak. The three priorities for management in case

of an outbreak of EHV1 are an early diagnosis, prevention of further spread and

thorough management of clinical cases (Allen, 2002; Pusterla et al., 2009a).

The importance of an early diagnosis of EHV1 is crucial, as several specific

interventions may be implemented in case of an EHV1-induced outbreak. Measures

designed to contain potential EHV1 spread are necessary until EHV1 is either

confirmed or excluded (Lunn et al., 2009). A good understanding of the appropriate

diagnostical methods and understanding what samples to take is vital for outbreak

management (see chapter 3.3 Diagnosis).

To prevent further spread of virus to other horses or facilities, quarantine

measurements of the whole facility are necessary for at least 4 weeks after the last

clinical case. A recent paper even describes the necessity of isolating actively EHV1-

shedding horses in a separate airspace under strict biosecurity and isolation

procedures (Goehring et al., 2010b). All affected animals should be kept isolated and

no horses should be allowed to enter or leave the property until quarantine is lifted

(Allen & Bryans, 1986; Pusterla et al., 2009a; van Maanen, 2002). Areas

contaminated by virus should be carefully cleaned with detergents. In the face of an

EHV1 outbreak, vaccination can be used for horses at increased risk of exposure and

to reduce the spread of infectious virus. There is some controversy associated with

this practice, because of the concern that EHM might be associated with a history of

frequent vaccination (Henninger et al., 2007). However, the latter statement should be

interpreted with care, as the majority of horses that was vaccinated 3-4 times a year

were older than 5 years. The latter has been described as a risk factor for the

development of neurologic disease (Goehring et al., 2006).

Clinical cases should be handled as described in chapter 3.2.3 Treatment.

Review of the literature

41

References

Allen, G. P. (2002). Epidemic disease caused by equine herpesvirus-1: Recommendations

for prevention and control. Equine Vet Educ 4, 177-183.

Allen, G. P. (2007). Development of a real-time polymerase chain reaction assay for

rapid diagnosis of neuropathogenic strains of equine herpesvirus-1. J Vet Diagn

Invest 19, 69-72.

Allen, G. P., Bolin, D. C., Bryant, U., Carter, C. N., Giles, R. C., Harrison, L. R.,

Hong, C. B., Jackson, C. B., Poonacha, K., Wharton, R. & Williams, N. M. (2008). Prevalence of latent, neuropathogenic equine herpesvirus-1 in the

Thoroughbred broodmare population of central Kentucky. Equine Vet J 40, 105-

110.

Allen, G. P. & Breathnach, C. C. (2006). Quantification by real-time PCR of the

magnitude and duration of leucocyte-associated viraemia in horses infected with

neuropathogenic vs. non-neuropathogenic strains of EHV-1. Equine Vet J 38, 252-

257.

Allen, G. P. & Bryans, J. T. (1986). Molecular epizootiology, pathogenesis, and

prophylaxis of equine herpesvirus-1 infections. Prog Vet Microbiol Immunol 2,

78-144.

Allen, G. P., Kydd, J. H., Slater, J. D. & Smith, K. C. (1999). Recent advances in

understanding the pathogenesis, epidemiology and immunological control of

equid herpesvirus-1 (EHV-1) abortion. Proceedings of the Eight International

Conference on Equine Infectious Diseases, 129-146.

Baxi, M. K., Efstathiou, S., Lawrence, G., Whalley, J. M., Slater, J. D. & Field, H. J. (1995). The detection of latency-associated transcripts of equine herpesvirus 1 in

ganglionic neurons. J Gen Virol 76, 3113-3118.

Blanchard, T. L., Kenney, R. M. & Timoney, P. J. (1992). Venereal disease. Vet Clin

North Am Equine Pract 8, 191-203.

Borchers, K., Thein, P. & Sterner-Kock, A. (2006). Pathogenesis of equine herpes-

associated neurological disease: a revised explanation. Equine Vet J 38, 283-287.

Borchers, K., Wolfinger, U., Goltz, M., Broll, H. & Ludwig, H. (1997). Distribution

and relevance of equine herpesvirus type 2 (EHV-2) infections. Arch Virol 142,

917-928.

Borchers, K., Wolfinger, U., Ludwig, H., Thein, P., Baxi, S., Field, H. J. & Slater, J. D. (1998). Virological and molecular biological investigations into equine herpes

virus type 2 (EHV-2) experimental infections. Virus Res 55, 101-106.

Breathnach, C. C., Yeargan, M. R., Sheoran, A. S. & Allen, G. P. (2001). The

mucosal humoral immune response of the horse to infective challenge and

vaccination with Equine herpesvirus-1 antigens. Equine Vet J 33, 651-657.

Brosnahan, M. M., Damiani, A., van de Walle, G., Erb, H., Perkins, G. A. & Osterrieder, N. (2009). The effect of siRNA treatment on experimental equine

herpesvirus type 1 (EHV-1) infection in horses. Virus research,

doi:10.1016/j.virusres.2099.1010.1017.

Brown, J. A., Mapes, S., Ball, B. A., Hodder, A. D., Liu, I. K. & Pusterla, N. (2007). Prevalence of equine herpesvirus-1 infection among Thoroughbreds residing on a

farm on which the virus was endemic. J Am Vet Med Assoc 231, 577-580.

Browning, G. F., Ficorilli, N. & Studdert, M. J. (1988). Asinine herpesvirus genomes:

comparison with those of the equine herpesviruses. Arch Virol 101, 183-190.

Review of the literature

42

Bryant, N. A., Davis-Poynter, N., Vanderplasschen, A. & Alcami, A. (2003). Glycoprotein G isoforms from some alphaherpesviruses function as broad-

spectrum chemokine binding proteins. EMBO J 22, 833-846.

Bürki, F., Rossmanith, W., Nowotny, N., Pallan, C., Mostl, K. & Lussy, H. (1990). Viremia and abortions are not prevented by two commercial equine herpesvirus-1

vaccines after experimental challenge of horses. Veterinary Quaterly 12, 80-86.

Burrows, R. & Goodridge, D. (1972). In vivo and in vitro studies of equine

rhinopneumonitis strains. Proc 3rd Int Conf of Equine Infect Dis, Paris, 306-321.

Burrows, R., Goodridge, D. & Denyer, M. S. (1984). Trials of an inactivated equid

herpesvirus 1 vaccine: challenge with a subtype 1 virus. Vet Rec 114, 369-374.

Carroll, C. L. & Westbury, H. A. (1985). Isolation of equine herpesvirus 1 from the

brain of a horse affected with paresis. Australian Veterinary Journal 62, 345-346.

Carvalho, R., Oliveira, A. M., Souza, A. M., Passos, L. M. & Martins, A. S. (2000). Prevalence of equine herpesvirus type 1 latency detected by polymerase chain

reaction. Arch Virol 145, 1773-1787.

Caughman, G. B., Staczek, J. & O'Callaghan, D. J. (1985). Equine herpesvirus type 1

infected cell polypeptides: evidence for immediate early/early/late regulation of

viral gene expression. Virology 145, 49-61.

Chesters, P. M., Allsop, R., Purewal, A. & Edington, N. (1997). Detection of latency-

associated transcripts of equid herpesvirus 1 in equine leukocytes but not in

trigeminal ganglia. J Virol 71, 3437-3443.

Coggins, L. (1979). Viral respiratory disease of horses. Vet Clin North Am, large Anim

Pract I, 59-72.

Crabb, B. S. & Studdert, M. J. (1990). Comparative studies of the proteins of equine

herpesviruses 4 and 1 and asinine herpesvirus 3: antibody response of the natural

hosts. J Gen Virol 71 ( Pt 9), 2033-2041.

Crabb, B. S. & Studdert, M. J. (1993). Epitopes of glycoprotein G of equine

herpesviruses 4 and 1 located near the C termini elicit type-specific antibody

responses in the natural host. J Virol 67, 6332-6338.

Crabb, B. S. & Studdert, M. J. (1995). Equine herpesvirus 4 (equine rhinopneumonitis

virus) and 1 (equine abortion virus). Advances in Virus Research 45, 153-190.

Csellner, H., Walker, C., Wellington, J. E., McLure, L. E., Love, D. N. & Whalley, J. M. (2000). EHV-1 glycoprotein D (EHV-1 gD) is required for virus entry and

cell-cell fusion, and an EHV-1 gD deletion mutant induces a protective immune

response in mice. Arch Virol 145, 2371-2385.

Davison, A. J., Eberle, R., Ehlers, B., Hayward, G. S., McGeoch, D. J., Minson, A. C., Pellett, P. E., Roizman, B., Studdert, M. J. & Thiry, E. (2009). The order

Herpesvirales. Arch Virol 154, 171-177.

Del Piero, F. & Wilkins, P. A. (2001). Pulmonary vasculotropic EHV-1 infection in

equids. Veterinary Pathology 38, 474.

Del Piero, F., Wilkins, P. A., Timoney, P. J., Kadushin, J., Vogelbacker, H., Lee, J. W., Berkowitz, S. J. & La Perle, K. M. D. (2000). Fatal nonneurological EHV-1

infection in a yearling filly. Veterinary Pathology 37, 672-676.

Dimock, W. W. & Edwards, P. R. (1933). Is there a filterable virus of abortion in

mares? Kentucky Agricultural Experiment Station Bulletin 333, 297-301.

Doll, E. R. (1952). Seasonal incidence and fetal age in equine viral abortion. Cornell

Veterinarian 42, 505-509.

Doll, E. R. & Bryans, J. T. (1963). Epizootiology of equine viral rhinopneumonitis. J

Am Vet Med Assoc 142, 31-37.

Review of the literature

43

Doll, E. R., Crowe, M. E. W., Bryans, J. T. & McCollum, W. H. (1955). Infection

immunity in equine virus abortion. Cornell Veterinarian 45, 387-410.

Drummer, H. E., Reynolds, A., Studdert, M. J., MacPherson, C. M. & Crabb, B. S. (1995). Application of an equine herpesvirus 1 (EHV1) type-specific ELISA to

the management of an outbreak of EHV1 abortion. Vet Rec 136, 579-581.

Edington, N., Bridges, C. G. & Huckle, A. (1985). Experimental reactivation of equid

herpesvirus 1 (EHV1) following the administration of corticosteroids. Equine Vet

J 17, 369-372.

Edington, N., Bridges, C. G. & Patel, J. R. (1986). Endothelial cell infection and

thrombosis in paralysis caused by equid herpesvirus-1: equine stroke. Arch Virol

90, 111-124.

Edington, N., Smith, B. & Griffiths, L. (1991). The role of endothelial cell infection in

the endometrium, placenta and foetus of equid herpesvirus 1 (EHV-1) abortions.

Journal of Comparative Pathology 104, 379-387.

Edington, N., Welch, H. M. & Griffiths, L. (1994). The prevalence of latent Equid

herpesviruses in the tissues of 40 abattoir horses. Equine Vet J 26, 140-142.

Elia, G., Decaro, N., Martella, V., Campolo, M., Desario, C., Lorusso, E., Cirone, F. & Buonavoglia, C. (2006). Detection of equine herpesvirus type 1 by real time

PCR. J Virol Methods 133, 70-75.

Ficorilli, N., Studdert, M. J. & Crabb, B. S. (1995). The nucleotide sequence of asinine

herpesvirus 3 glycoprotein G indicates that the donkey virus is closely related to

equine herpesvirus 1. Arch Virol 140, 1653-1662.

Foote, C. E., Love, D. N., Gilkerson, J. R., Wellington, J. E. & Whalley, J. M. (2006). EHV-1 and EHV-4 infection in vaccinated mares and their foals. Vet Immunol

Immunopathol 111, 41-46.

Foote, C. E., Love, D. N., Gilkerson, J. R. & Whalley, J. M. (2002). Serological

responses of mares and weanlings following vaccination with an inactivated

whole virus equine herpesvirus 1 and equine herpesvirus 4 vaccine. Vet Microbiol

88, 13-25.

Frampton, A. R., Jr., Goins, W. F., Cohen, J. B., von Einem, J., Osterrieder, N., O'Callaghan, D. J. & Glorioso, J. C. (2005). Equine herpesvirus 1 utilizes a

novel herpesvirus entry receptor. J Virol 79, 3169-3173.

Frampton, A. R., Jr., Stolz, D. B., Uchida, H., Goins, W. F., Cohen, J. B. & Glorioso, J. C. (2007). Equine herpesvirus 1 enters cells by two different pathways, and

infection requires the activation of the cellular kinase ROCK1. J Virol 81, 10879-

10889.

Frampton, A. R., Jr., Uchida, H., von Einem, J., Goins, W. F., Grandi, P., Cohen, J. B., Osterrieder, N. & Glorioso, J. C. (2010). Equine herpesvirus type 1 (EHV-1)

utilizes microtubules, dynein, and ROCK1 to productively infect cells. Vet

Microbiol 141, 12-21.

Friday, P. A., Scarratt, W. K., Elvinger, F., Timoney, P. J. & Bonda, A. (2000). Ataxia and paresis with equine herpesvirus type 1 infection in a herd of riding

school horses. J Vet Intern Med 14, 197-201.

Fritsche, A. K. & Borchers, K. (2010). Detection of neuropathogenic strains of Equid

Herpesvirus 1 (EHV-1) associated with abortions in Germany. Vet Microbiol 147,

176-180.

Fukushi, H., Tomita, T., Taniguchi, A., Ochiai, Y., Kirisawa, R., Matsumura, T., Yanai, T., Masegi, T., Yamaguchi, T. & Hirai, K. (1997). Gazelle herpesvirus

Review of the literature

44

1: a new neurotropic herpesvirus immunologically related to equine herpesvirus 1.

Virology 227, 34-44.

Fulton, A., Peters, S. T., Perkins, G. A., Jarosinski, K. W., Damiani, A., Brosnahan, M., Buckles, E. L., Osterrieder, N. & Van de Walle, G. R. (2009). Effective

treatment of respiratory alphaherpesvirus infection using RNA interference. PLoS

One 4, e4118.

Furlong, D., Swift, H. & Roizman, B. (1972). Arrangement of herpesvirus

deoxyribonucleic acid in the core. J Virol 10, 1071-1074.

Garré, B., Gryspeerdt, A., Croubels, S., De Backer, P. & Nauwynck, H. (2009). Evaluation of orally administered valacyclovir in experimentally EHV1-infected

ponies. Vet Microbiol 135, 214-221.

Garré, B., Shebany, K., Gryspeerdt, A., Baert, K., van der Meulen, K., Nauwynck, H., Deprez, P., De Backer, P. & Croubels, S. (2007). Pharmacokinetics of

acyclovir after intravenous infusion of acyclovir and after oral administration of

acyclovir and its prodrug valacyclovir in healthy adult horses. Antimicrob Agents

Chemother 51, 4308-4314.

Gibson, J. S., Slater, J. D., Awan, A. R. & Field, H. J. (1992). Pathogenesis of equine

herpesvirus-1 in specific pathogen-free foals: primary and secondary infections

and reactivation. Arch Virol 123, 351-366.

Gilkerson, J., Love, D. & Whalley, J. M. (1997). Serological evidence of equine

herpesvirus (EHV-1) infection in Thoroughbred foals 30-120 days of age.

Australian Equine Veterinarian 15, 128-134.

Gilkerson, J. R., Whalley, J. M., Drummer, H. E., Studdert, M. J. & Love, D. N. (1999). Epidemiology of EHV-1 and EHV-4 in the mare and foal populations on a

Hunter Valley stud farm: are mares the source of EHV-1 for unweaned foals. Vet

Microbiol 68, 27-34.

Gleeson, L. J. & Coggins, L. (1980). Response of pregnant mares to equine herpesvirus

1 (EHV1). Cornell Veterinarian 70, 391-400.

Goehring, L. S., Hussey, G. S., Ashton, L. V., Schenkel, A. R. & Lunn, D. P. (2010a). Infection of central nervous system endothelial cells by cell-associated EHV-1.

Vet Microbiol doi:10.1016/j.vetmic.2010.08.030.

Goehring, L. S., Landolt, G. A. & Morley, P. S. (2010b). Detection and management of

an outbreak of equine herpesvirus type 1 infection and associated neurological

disease in a veterinary teaching hospital. J Vet Intern Med 24, 1176-1183.

Goehring, L. S. & Sloet van Oldruitenborgh-Oosterbaan, M. M. (2001). The mystery

of equine herpes myeloencephalopathy. Equine Veterinary Education 13, 36-42.

Goehring, L. S., van Winden, S. C., van Maanen, C. & Sloet van Oldruitenborgh-Oosterbaan, M. M. (2006). Equine herpesvirus type 1-associated

myeloencephalopathy in The Netherlands: a four-year retrospective study (1999-

2003). J Vet Intern Med 20, 601-607.

Goehring, L. S., Wagner, B., Bigbie, R., Hussey, S. B., Rao, S., Morley, P. S. & Lunn, D. P. (2010c). Control of EHV-1 viremia and nasal shedding by commercial

vaccines. Vaccine 28, 5203-5211.

Goodman, L. B., Loregian, A., Perkins, G. A., Nugent, J., Buckles, E. L., Mercorelli,

B., Kydd, J. H., Palu, G., Smith, K. C., Osterrieder, N. & Davis-Poynter, N. (2007). A point mutation in a herpesvirus polymerase determines

neuropathogenicity. PLoS Pathog 3, 1583-1592.

Goodman, L. B., Wagner, B., Flaminio, M. J., Sussman, K. H., Metzger, S. M., Holland, R. & Osterrieder, N. (2006). Comparison of the efficacy of inactivated

Review of the literature

45

combination and modified-live virus vaccines against challenge infection with

neuropathogenic equine herpesvirus type 1 (EHV-1). Vaccine 24, 3636-3645.

Goulden, B. C., Wilks, C. & Jones, D. (1989). Equine herpesvirus (EHV1) disease.

Publication of the New-Zealand Equine Research Foundation, 1-32.

Granzow, H., Klupp, B. G., Fuchs, W., Veits, J., Osterrieder, N. & Mettenleiter, T. C. (2001). Egress of alphaherpesviruses: comparative ultrastructural study. J Virol

75, 3675-3684.

Gray, W. L., Baumann, R. P., Robertson, A. T., Caughman, G. B., O'Callaghan, D. J. & Staczek, J. (1987a). Regulation of equine herpesvirus type 1 gene

expression: characterization of immediate early, early, and late transcription.

Virology 158, 79-87.

Gray, W. L., Baumann, R. P., Robertson, A. T., O'Callaghan, D. J. & Staczek, J. (1987b). Characterization and mapping of equine herpesvirus type 1 immediate

early, early, and late transcripts. Virus Res 8, 233-244.

Greenwood, R. E. & Simson, A. R. (1980). Clinical report of a paralytic syndrome

affecting stallions, mares and foals on a thoroughbred studfarm. Equine Vet J 12,

113-117.

Hartley, C. A., Drummer, H. E. & Studdert, M. J. (1999). Gazelle herpesvirus 1

(GHV-1): Closely related to EHV-1 and AHV-3. The nucleotide sequence of the

glycoprotein G homologue of equine herpesvirus 3 (EHV-3) indicates EHV-3 is a

distinct equid hperesvirus. Brief report. Arch Virol 144, 2023-2033.

Hartley, C. A., Wilks, C. R., Studdert, M. J. & Gilkerson, J. R. (2005). Comparison of

antibody detection assays for the diagnosis of equine herpesvirus 1 and 4

infections in horses. Am J Vet Res 66, 921-928.

Heldens, J. G., Hannant, D., Cullinane, A. A., Prendergast, M. J., Mumford, J. A.,

Nelly, M., Kydd, J. H., Weststrate, M. W. & van den Hoven, R. (2001). Clinical and virological evaluation of the efficacy of an inactivated EHV1 and

EHV4 whole virus vaccine (Duvaxyn EHV1,4). Vaccination/challenge

experiments in foals and pregnant mares. Vaccine 19, 4307-4317.

Henninger, R. W., Reed, S. M., Saville, W. J., Allen, G. P., Hass, G. F., Kohn, C. W. & Sofaly, C. (2007). Outbreak of Neurologic disease Caused by Equine

Herpesvirus-1 at a University Equestrian Center. J Vet Intern Med 21, 157-165.

Huemer, H. P., Nowotny, N., Crabb, B. S., Meyer, H. & Hubert, P. H. (1995). gp13

(EHV-gC): a complement receptor induced by equine herpesviruses. Virus

Research 37, 113-126.

Hussey, S. B., Clark, R., Lunn, K. F., Breathnach, C., Soboll, G., Whalley, J. M. & Lunn, D. P. (2006). Detection and quantification of equine herpesvirus-1 viremia

and nasal shedding by real-time polymerase chain reaction. J Vet Diagn Invest 18,

335-342.

Jackson, T. A. & Kendrick, J. W. (1971). Paralysis of horses associated with equine

herpesvirus-1 infection. J Am Vet Med Assoc 159, 1351-1357.

Jackson, T. A., Osburn, B. I., Cordy, D. R. & Kendrick, J. W. (1977). Equine

herpesvirus 1 infection of horses: studies on the experimentally induced

neurologic disease. Am J Vet Res 38, 709-719.

Kershaw, O., von Oppen, T., Glitz, F., Deegen, E., Ludwig, H. & Borchers, K. (2001). Detection of equine herpesvirus type 2 (EHV-2) in horses with

keratoconjunctivitis. Virus Res 80, 93-99.

Kleiboeker, S. B., Schommer, S. K., Johnson, P. J., Ehlers, B., Turnquist, S. E., Boucher, M. & Kreeger, J. M. (2002). Association of two newly recognized

Review of the literature

46

herpesviruses with interstitial pneumonia in donkeys (Equus asinus). J Vet Diagn

Invest 14, 273-280.

Kurtz, B. M., Singletary, L. B., Kelly, S. D. & Frampton, A. R., Jr. (2010). Equus

caballus major histocompatibility complex class I is an entry receptor for equine

herpesvirus type 1. J Virol 84, 9027-9034.

Kydd, J. H., Smith, K. C., Hannant, D., Livesay, G. J. & Mumford, J. A. (1994a). Distribution of equid herpesvirus-1 (EHV-1) in respiratory tract associated

lymphoid tissue: implications for cellular immunity. Equine Vet J 26, 470-473.

Kydd, J. H., Smith, K. C., Hannant, D., Livesay, G. J. & Mumford, J. A. (1994b). Distribution of equid herpesvirus-1 (EHV-1) in the respiratory tract of ponies:

implications for vaccination strategies. Equine Vet J 26, 466-469.

Kydd, J. H., Townsend, H. G. & Hannant, D. (2006). The equine immune response to

equine herpesvirus-1: the virus and its vaccines. Vet Immunol Immunopathol 111,

15-30.

Kydd, J. H., Wattrang, E. & Hannant, D. (2003). Pre-infection frequencies of equine

herpesvirus-1 specific, cytotoxic T lymphocytes correlate with protection against

abortion following experimental infection of pregnant mares. Vet Immunol

Immunopathol 96, 207-217.

Lunn, D. P., Davis-Pointer, N., Flaminio, M. J., Horohov, D. W., Osterrieder, K., Pusterla, N. & Townsend, H. G. (2009). Equine Herpesvirus-1 Consensus

Statement. J Vet Intern Med 23, 450-461.

Marenzoni, M. L., Passamonti, F., Cappelli, K., Veronesi, F., Capomaccio, S., Supplizi, A. V., Valente, C., Autorino, G. & Coletti, M. (2008). Clinical,

serological and molecular investigations of EHV-1 and EHV-4 in 15 unweaned

thoroughbred foals. Vet Rec 162, 337-341.

Matsumura, T., Kondo, T., Sugita, S., Damiani, A. M., O'Callaghan, D. J. & Imagawa, H. (1998). An equine herpesvirus type 1 recombinant with a deletion in

the gE and gI genes is avirulent in young horses. Virology 242, 68-79.

Maxwell, L. K., Bentz, B. G., Bourne, D. W. & Erkert, R. S. (2008a). Pharmacokinetics of valacyclovir in the adult horse. J Vet Pharmacol Ther 31,

312-320.

Maxwell, L. K., Bentz, B. G., Gilliam, L. L., Ritchey, J. W., Holbrook, T. C.,

McFarlane, D., Rezabek, G. B., MacAllister, C. G. & Allen, G. P. (2008b). Efficacy of Valacyclovir Against Clinical Disease After EHV-1 Challenge in

Aged Mares. Proceedings of the Annual Convention of the AAEP 54, 198-199.

McCartan, C. G., Russell, M. M., Wood, J. L. & Mumford, J. A. (1995). Clinical,

serological and virological characteristics of an outbreak of paresis and neonatal

foal disease due to equine herpesvirus-1 on a stud farm. Vet Rec 136, 7-12.

Mettenleiter, T. C. (2002). Herpesvirus assembly and egress. J Virol 76, 1537-1547.

Mettenleiter, T. C. (2004). Budding events in herpesvirus morphogenesis. Virus Res 106,

167-180.

Mettenleiter, T. C., Klupp, B. G. & Granzow, H. (2006). Herpesvirus assembly: a tale

of two membranes. Curr Opin Microbiol 9, 423-429.

Mettenleiter, T. C., Klupp, B. G. & Granzow, H. (2009). Herpesvirus assembly: an

update. Virus Res 143, 222-234.

Minke, J. M., Audonnet, J. C. & Fischer, L. (2004). Equine viral vaccines: the past,

present and future. Vet Res 35, 425-443.

Mumford, J. A. (1984). The development of diagnostic techniques for equine viral

diseases. Veterinary Annals 24, 182-189.

Review of the literature

47

Mumford, J. A. & Edington, N. (1980). EHV1 and equine paresis. Vet Rec 106, 277.

Mumford, J. A., Hannant, D. A., Jesset, D. M., O'Neill, T., Smith, K. C. & Ostlund, E. N. (1994). Abortigenic and neurological disease caused by experimental

infection with equine herpesvirus 1. Proceedings of the 7th International

Conference of Equine Infectious diseases, Newmarket, UK, 261-275.

Mumford, J. A., Rossdale, P. D., Jessett, D. M., Gann, S. J., Ousey, J. & Cook, R. F. (1987). Serological and virological investigations of an equid herpesvirus 1 (EHV-

1) abortion storm on a stud farm in 1985. Journal of Reproduction and Fertility,

supplement 35, 509-518.

Murray, M. J., del Piero, F., Jeffrey, S. C., Davis, M. S., Furr, M. O., Dubovi, E. J. & Mayo, J. A. (1998). Neonatal Equine Herpesvirus Type 1 Infection on a

Thoroughbred Breeding Farm. J Vet Intern Med 12, 36-41.

Neubauer, A., Braun, B., Brandmuller, C., Kaaden, O. R. & Osterrieder, N. (1997). Analysis of the contributions of the equine herpesvirus 1 glycoprotein gB

homolog to virus entry and direct cell-to-cell spread. Virology 227, 281-294.

Neubauer, A. & Osterrieder, N. (2004). Equine herpesvirus type 1 (EHV-1)

glycoprotein K is required for efficient cell-to-cell spread and virus egress.

Virology 329, 18-32.

Nugent, J., Birch-Machin, I., Smith, K. C., Mumford, J. A., Swann, Z., Newton, J. R., Bowden, R. J., Allen, G. P. & Davis-Poynter, N. (2006). Analysis of equid

herpesvirus 1 strain variation reveals a point mutation of the DNA polymerase

strongly associated with neuropathogenic versus non-neuropathogenic disease

outbreaks. J Virol 80, 4047-4060.

Osterrieder, N. (1999). Construction and characterization of an equine herpesvirus 1

glycoprotein C negative mutant. Virus Research 59, 165-177.

Osterrieder, N., Neubauer, A., Brandmuller, C., Braun, B., Kaaden, O. R. & Baines, J. D. (1996). The equine herpesvirus 1 glycoprotein gp21/22a, the herpes simplex

virus type 1 gM homolog, is involved in virus penetration and cell-to-cell spread

of virions. J Virol 70, 4110-4115.

Ostlund, E. N. (1993). The equine herpesviruses. Vet Clin North Am Equine Pract 9,

283-294.

Pascoe, R. R., Spradbrow, P. B. & Bagust, T. J. (1968). Equine coital exanthema. Aust

Vet J 44, 485.

Patel, J. R., Edington, N. & Mumford, J. A. (1982). Variation in cellular tropism

between isolates of equine herpesvirus-1 in foals. Arch Virol 74, 41-51.

Perkins, G. A., Ainsworth, D. M., Erb, H. N., del Piero, F., Miller, M., Wilkins, P. A., Palmer, J. & Frazer, M. (1999). Clinical, haematological and biochemical

findings in foals with neonatal Equine herpesvirus-1 infection compared with

septic and premature foals. Equine Vet J 35, 422-426.

Perkins, G. A., Goodman, L. B., Tsujimura, K., Van de Walle, G. R., Kim, S. G., Dubovi, E. J. & Osterrieder, K. (2009). Investigation of the prevalence of

neurologic equine herpes virus type 1 (EHV-1) in a 23-year retrospective analysis

(1984-2007). Vet Microbiol 139, 375-378.

Platt, H., Singh, H. & Whitwell, K. E. (1980). Pathological observations on an outbreak

of paralysis in brood mares. Equine Vet J 12, 118-126.

Prickett, M. E. (1970). The pathology of disease caused by equine herpesvirus-1.

Proceedings of the 2nd International Conference on Equine Infectious diseases,

Paris, 24-33.

Review of the literature

48

Pronost, S., Leon, A., Legrand, L., Fortier, C., Miszczak, F., Freymuth, F. & Fortier, G. (2010). Neuropathogenic and non-neuropathogenic variants of equine

herpesvirus 1 in France. Vet Microbiol 145, 329-333.

Pusterla, N., David Wilson, W., Madigan, J. E. & Ferraro, G. L. (2009a). Equine

herpesvirus-1 myeloencephalopathy: a review of recent developments. Vet J 180,

279-289.

Pusterla, N., Hussey, S. B., Mapes, S., Johnson, C., Collier, J. R., Hill, J., Lunn, D. P. & Wilson, W. D. (2010). Molecular investigation of the viral kinetics of equine

herpesvirus-1 in blood and nasal secretions of horses after corticosteroid-induced

recrudescence of latent infection. J Vet Intern Med 24, 1153-1157.

Pusterla, N., Hussey, S. B., Mapes, S., Leutenegger, C. M., Madigan, J. E., Ferraro, G. L., Wilson, W. D. & Lunn, D. P. (2009b). Comparison of four methods to

quantify Equid herpesvirus 1 load by real-time polymerase chain reaction in nasal

secretions of experimentally and naturally infected horses. J Vet Diagn Invest 21,

836-840.

Pusterla, N., Mapes, S. & Wilson, W. D. (2008a). Diagnostic sensitivity of

nasopharyngeal and nasal swabs for the molecular detection of EHV-1. Vet Rec

162, 520-521.

Pusterla, N., Mapes, S. & Wilson, W. D. (2008b). Use of viral loads in blood and

nasopharyngeal secretions fot the diagnosis of EHV-1 infection in field cases. Vet

Rec 162, 728-729.

Pusterla, N., Wilson, W. D., Mapes, S., Finno, C., Isbell, D., Arthur, R. M. & Ferraro, G. L. (2009c). Characterization of viral loads, strain and state of equine

herpesvirus-1 using real-time PCR in horses following natural exposure at a

racetrack in California. Vet J 179, 230-239.

Reed, S. M. & Toribio, R. E. (2004). Equine herpesvirus 1 and 4. Vet Clin North Am

Equine Pract 20, 631-642.

Robertson, A. T., Caughman, G. B., Gray, W. L., Baumann, R. P., Staczek, J. & O'Callaghan, D. J. (1988). Analysis of the in vitro translation products of the

equine herpesvirus type 1 immediate early mRNA. Virology 166, 451-462.

Robertson, G. R., Scott, N. A., Miller, J. M., Sabine, M., Zheng, M., Bell, C. W. & Whalley, J. M. (1991). Sequence characteristics of a gene in equine herpesvirus 1

homologous to glycoprotein H of herpes simplex virus. DNA Seq 1, 241-249.

Roizman, B., Desrosiers, R. C., Fleckenstein, B., Lopez, C., Minson, A. C. & Studdert, M. J. (1992). The family Herpesviridae: an update. The Herpesvirus

Study Group of the International Committee on Taxonomy of Viruses. Arch Virol

123, 425-449.

Roizman, B. & Knipe, D. M., (eds) (2001). Herpes Simplex Viruses and Their

Replication. Philadelphia: Lipincott Williams and Wilkins.

Roizman, B. & Pellett, P. E., (eds) (2001). The Family Herpesviridae: A Brief

Introduction. Philadelphia: Lippincott Williams & Wilkins.

Rudolph, J. & Osterrieder, N. (2002). Equine Herpesvirus Type 1 Devoid of gM and

gp2 Is Severely Impaired in Virus Egress but Not Direct Cell-to-Cell Spread.

Virology 293, 356-367.

Rudolph, J., Seyboldt, C., Granzow, H. & Osterrieder, N. (2002). The gene 10

(UL49.5) product of equine herpesvirus 1 is necessary and sufficient for

functional processing of glycoprotein M. J Virol 76, 2952-2963.

Sabine, M., Robertson, G. R. & Whalley, J. M. (1981). Differentiation of sub-types of

equine herpesvirus I by restriction endonuclease analysis. Aust Vet J 57, 148-149.

Review of the literature

49

Sasaki, M., Hasebe, R., Makino, Y., Suzuki, T., Fukushi, H., Okamoto, M., Matsuda, K., Taniyama, H., Sawa, H. & Kimura, T. (2011). Equine major

histocompatibility complex class I molecules act as entry receptors that bind to

equine herpesvirus-1 glycoprotein D. Genes Cells 16, 343-357.

Saxegaard, F. (1966). Isolation and identification of equine rhinopneumonitis virus

(equine abortion virus) from cases of abortion and paralysis. Nordic Veterinary

Medicine 18, 504-510.

Schlocker, N., Gerber-Bretscher, R. & von Fellenberg, R. (1995). Equine herpesvirus

2 in pulmonary macrophages of horses. Am J Vet Res 56, 749-754.

Schultheiss, P. C., Collins, J. K. & Hotaling, S. F. (1997). Immunohistochemical

demonstration of equine herpesvirus-1 antigen in neurons and astrocytes of horses

with acute paralysis. Vet Pathol 34, 52-54.

Scott, J. C., Dutta, S. K. & Myrup, A. C. (1983). In vivo harboring of equine

herpesvirus-1 in leukocyte populations and subpopulations and their quantitation

from experimentally infected ponies. American Journal of Veterinary Research

44, 1344-1348.

Sharma, P. C., Cullinane, A. A., Onions, D. E. & Nicolson, L. (1992). Diagnosis of

equid herpesviruses -1 and -4 by polymerase chain reaction. Equine Vet J 24, 20-

25.

Shimizu, T., Ishizaki, R., Ishii, S., Kawakami, Y., Kaji, R., Sugimura, K. & Matumoto, M. (1959). Isolation of equine abortion virus from natural cases of

equine abortion in horse kidney cell culture. Jap J exp Med 29, 643-649.

Slater, J. D., Borchers, K., Thackray, A. M. & Field, H. J. (1994). The trigeminal

ganglion is a location for equine herpesvirus 1 latency and reactivation in the

horse. J Gen Virol 75, 2007-2016.

Smith, D. J., Hamblin, A. & Edington, N. (2002). Equid herpesvirus 1 infection of

endothelial cells requires activation of putative adhesion molecules: an in vitro

model. Clin Exp Immunol 129, 281-287.

Smith, D. J., Hamblin, A. S. & Edington, N. (2001). Infection of endothelial cells with

equine herpesvirus-1 (EHV-1) occurs where there is activation of putative

adhesion molecules: a mechanism for transfer of virus. Equine Vet J 33, 138-142.

Smith, K. C. (1997). Herpesviral Abortion in Domestic Animals. The Veterinary Journal

153, 253-268.

Smith, K. C., Whitwell, K. E., Binns, M. M., Dolby, C. A., Hannant, D. & Mumford, J. A. (1992). Abortion of virologically negative fetuses following experimental

challenge of pregnant mares with equid herpevirus 1. Equine Vet J 24, 256-259.

Smith, K. C., Whitwell, K. E., Mumford, J. A., Hannant, D., Blunden, A. S. & Tearle, J. P. (2000). Virulence of the V592 isolate of equid herpesvirus-1 in

ponies. Journal of Comparative Pathology 122, 288-297.

Smith, K. L., Allen, G. P., Branscum, A. J., Cook, R. F., Vickers, M. L., Timoney, P. J. & Balasuriya, U. B. R. (2010). The increased prevalence of neuropathogenic

strains of EHV-1 in equine abortions. Vet Microbiol 141, 5-11.

Smith, P. M., Kahan, S. M., Rorex, C. B., von Einem, J., Osterrieder, N. & O'Callaghan, D. J. (2005). Expression of the full-length form of gp2 of equine

herpesvirus 1 (EHV-1) completely restores respiratory virulence to the attenuated

EHV-1 strain KyA in CBA mice. J Virol 79, 5105-5115.

Soboll, G., Breathnach, C. C., Kydd, J. H., Hussey, S. B., Mealey, R. M. & Lunn, D. P. (2010). Vaccination of ponies with the IE gene of EHV-1 in a recombinant

Review of the literature

50

modified live vaccinia vector protects against clinical and virological disease. Vet

Immunol Immunopathol 135, 108-117.

Soboll, G., Hussey, S. B., Whalley, J. M., Allen, G. P., Koen, M. T., Santucci, N., Fraser, D. G., Macklin, M. D., Swain, W. F. & Lunn, D. P. (2006). Antibody

and cellular immune responses following DNA vaccination and EHV-1 infection

of ponies. Vet Immunol Immunopathol 111, 81-95.

Stierstorfer, B., Eichhorn, W., Schmahl, W., Brandmuller, C., Kaaden, O. R. & Neubauer, A. (2002). Equine herpesvirus type 1 (EHV-1) myeloencephalopathy:

a case report. J Vet Med B Infect Dis Vet Public Health 49, 37-41.

Stokes, A., Alber, D. G., Greensill, J., Amellal, B., Carvalho, R., Taylor, L. A., Doel, T. R., Killington, R. A., Halliburton, I. W. & Meredith, D. M. (1996). The

expression of the proteins of equine herpesvirus 1 which share homology with

herpes simplex virus 1 glycoproteins H and L. Virus Res 40, 91-107.

Studdert, M. J., Simpson, T. & Roizman, B. (1981). Differentiation of respiratory and

abortigenic isolates of equine herpesvirus 1 by restriction endonucleases. Science

214, 562-564.

Sun, Y., MacLean, A. R., Aitken, J. D. & Brown, S. M. (1996). The role of the gene 71

product in the life cycle of equine herpesvirus 1. J Gen Virol 77 ( Pt 3), 493-500.

Turtinen, L. W. & Allen, G. P. (1982). Identification of the envelope surface

glycoproteins of equine herpesvirus type 1. J Gen Virol 63, 481-485.

Turtinen, L. W., Allen, G. P., Darlington, R. W. & Bryans, J. T. (1981). Serological

and molecular comparisons of several equine herpesvirus type 1 strains. Am J vet

Res 42, 2099-2104.

USDA (2007). Equine herpesvirus myeloencephalopathy: A potentially emerging disease.

Available at: http://wwwaphisusdagov/vs/nahss/equine/ehv.

Van de Walle, G. R., Goupil, R., Wishon, C., Damiani, A., Perkins, G. A. & Osterrieder, N. (2009). A Single-Nucleotide Polymorphism in a Herpesvirus

DNA Polymerase Is Sufficient to Cause Lethal Neurological disease. J Infect Dis

200, 20-25.

Van de Walle, G. R., May, M. L., Sukhumavasi, W., von Einem, J. & Osterrieder, N. (2007). Herpesvirus chemokine-binding glycoprotein G (gG) efficiently inhibits

neutrophil chemotaxis in vitro and in vivo. J Immunol 179, 4161-4169.

Van de Walle, G. R., Peters, S. T., VanderVen, B. C., O'Callaghan, D. J. & Osterrieder, N. (2008a). Equine herpesvirus 1 entry via endocytosis is facilitated

by alphaV integrins and an RSD motif in glycoprotein D. J Virol 82, 11859-

11868.

Van de Walle, G. R., Sakamoto, K. & Osterrieder, N. (2008b). CCL3 and Viral

Chemokine-Binding Protein gG Modulate Pulmonary Inflammation and Virus

Replication during Equine Herpsvirus 1 Infection. J Virol 82, 1714-1722.

van der Meulen, K., Caij, B., Pensaert, M. & Nauwynck, H. (2006). Absence of viral

envelope proteins in equine herpesvirus 1-infected blood mononuclear cells during

cell-associated viremia. Vet Microbiol 113, 265-273.

van der Meulen, K., Vercauteren, G., Nauwynck, H. & Pensaert, M. (2003a). A local

epidemic of equine herpesvirus 1-induced neurological disorders in Belgium.

Vlaams Diergenskund Tijds 72, 366-372.

van der Meulen, K. M., Gryspeerdt, A. C., Vandekerckhove, A. A., Garre, B. A. & Nauwynck, H. J. (2007). The protective properties of vaccination against equine

herpesvirus 1-induced viremia, abortion and nervous system disorders. Vlaams

Diergenskund Tijds 76, 186-194.

Review of the literature

51

van der Meulen, K. M., Nauwynck, H. J. & Pensaert, M. B. (2003b). Absence of viral

antigens on the surface of equine herpesvirus-1-infected peripheral blood

mononuclear cells: a strategy to avoid complement-mediated lysis. J Gen Virol 84,

93-97.

van Maanen, C. (2002). Equine herpesvirus 1 and 4 infections: an update. Vet Q 24, 58-

78.

van Maanen, C., Sloet van Oldruitenborgh-Oosterbaan, M. M., Damen, E. A. & Derksen, A. G. (2001). Neurological disease associated with EHV-1-infection in

a riding school: clinical and virological characteristics. Equine Vet J 33, 191-196.

Van Maanen, C., Willink, D. L., Smeenk, L. A. J., Brinkhof, J. & Terpstra, C. (2000). An equine herpesvirus 1 (EHV1) abortion storm at a riding school. Vet Q

22, 83-87.

Varrasso, A., Dynon, K., Ficorilli, N., Hartley, C. A., Studdert, M. J. & Drummer, H. E. (2001). Identification of equine herpesviruses 1 and 4 by polymerase chain

reaction. Aust Vet J 79, 563-569.

Vengust, M., Wen, X. & Bienzle, D. (2008). Herpesvirus-associated neurological

disease in a donkey. J Vet Diagn Invest 20, 820-823.

Verryken, K., Saey, V., Maes, S., Borchers, K., Van de Walle, G., Ducatelle, R. & Deprez, P. (2010). First report of multinodular pulmonary fibrosis associated with

equine herpesvirus 5 in Belgium. Vlaams Diergen Tijds 79, 297-301.

Wang, L., Raidal, S. L., Pizzirani, A. & Wilcox, G. E. (2007). Detection of respiratory

herpesviruses in foals and adult horses determined by nested multiplex PCR. Vet

Microbiol 121, 18-28.

Wellington, J. E., Lawrence, G. L., Love, D. N. & Whalley, J. M. (1996a). Expression

and characterization of equine herpesvirus 1 glycoprotein D in mammalian cell

lines. Arch Virol 141, 1785-1793.

Wellington, J. E., Love, D. N. & Whalley, J. M. (1996b). Evidence for involvement of

equine herpesvirus 1 glycoprotein B in cell-cell fusion. Arch Virol 141, 167-175.

Whittaker, G. R., Taylor, L. A., Elton, D. M., Giles, L. E., Bonass, W. A., Halliburton, I. W., Killington, R. A. & Meredith, D. M. (1992). Glycoprotein

60 of equine herpesvirus type 1 is a homologue of herpes simplex virus

glycoprotein D and plays a major role in penetration of cells. J Gen Virol 73 ( Pt

4), 801-809.

Whitwell, K. E. & Blunden, A. S. (1992). Pathological findings in horses dying during

an outbreak of the paralytic form of Equid herpesvirus type 1 (EHV-1) infection.

Equine Vet J 24, 13-19.

Williams, K. J., Maes, R., Del Piero, F., Lim, A., Wise, A., Bolin, D. C., Caswell, J.,

Jackson, C., Robinson, N. E., Derksen, F., Scott, M. A., Uhal, B. D., Li, X., Youssef, S. A. & Bolin, S. R. (2007). Equine multinodular pulmonary fibrosis: a

newly recognized herpesvirus-associated fibrotic lung disease. Vet Pathol 44, 849-

862.

Wilson, W. D. (1997). Equine herpesvirus 1 myeloencephalopathy. Veterinary Clinics of

North America-Equine Practice 13, 53-&.

Wilsterman, S., Soboll-Hussey, G., Lunn, D. P., Ashton, L. V., Callan, R. J., Hussey, S. B., Rao, S. & Goehring, L. S. (2010). Equine herpesvirus-1 infected peripheral

blood mononuclear cell subpopulations during viremia. Vet Microbiol

doi:10.1016/j.vetmic.2010.10.004.

Wong, D. M., Belgrave, R. L., Williams, K. J., Del Piero, F., Alcott, C. J., Bolin, S.

R., Marr, C. M., Nolen-Walston, R., Myers, R. K. & Wilkins, P. A. (2008).

Review of the literature

52

Multinodular pulmonary fibrosis in five horses. J Am Vet Med Assoc 232, 898-

905.

Aims of the thesis

53

CHAPTER II

AIMS OF THE THESIS

Aims of the thesis

55

AIMS OF THE THESIS

Equine herpesvirus 1 (EHV1) is the most important cause of infectious abortion in

horses worldwide and can also lead to severe nervous system disorders with frequent fatal

outcome. Infection occurs via respiratory route and after local replication in tissues of the

upper respiratory tract, the virus spreads via a cell-associated viremia to target internal

organs such as the uterus or the nervous system. Secondary replication of EHV1 in

endothelial cells of these organs can result in late term abortion or nervous system

disorders.

Vaccination with the currently available vaccines cannot prevent clinical symptoms

such as abortion or nervous system disorders. Moreover, treatment of these symptoms is

only supportive and specific medication is not available. Therefore, an improvement of

existing vaccines and/or the development of therapies are necessary. Unfortunately, at the

start of this thesis, several important steps in the pathogenesis of this disease were still not

fully clarified, making it difficult to find new directions in the strategy to develop better

vaccines and to create drugs that inhibit steps of the pathogenesis. In order to achieve this

goal, studies in the natural host of EHV1, the horse, under both experimental and field

conditions, are of paramount importance to obtain relevant and correct information about

the pathogenesis of this disease.

Firstly, a thorough knowledge of the complete pathogenesis from primary replication

and invasion via cell-associated viremia to the target organs is urgent. Therefore, the first

aim of this thesis was to gain a more detailed insight in the invasion of neurologic and

non-neurologic strains of EHV1 from the upper respiratory tract to the carrier cells in the

blood stream by means of experimental infection studies in horses (Chapter III). The

second aim was to examine how individual EHV1-infected cells escape from immune

effector mechanisms at the primary site of replication (Chapter IV).

Secondly, to develop suitable vaccination and treatment strategies during natural

occurring outbreaks of disease, field information about the epidemiology of the virus is

needed. The third and last aim was therefore to gain more information about naturally

occurring outbreaks in Belgium in terms of risk factors, protective properties of

vaccination, the effect of experimental therapies and diagnosis (Chapter V).

Replication kinetics of EHV1

57

CHAPTER III

DIFFERENCES IN REPLICATION KINETICS

AND CELL TROPISM BETWEEN

NEUROVIRULENT AND NON-NEUROVIRULENT

EHV1 STRAINS DURING THE ACUTE PHASE OF

INFECTION IN HORSES

Veterinary microbiology 142 (2010), 242-253

Annick C. Gryspeerdt, A.P. Vandekerckhove, B. Garré, F. Barbé, G.R. Van de Walle, H.J.

Nauwynck

Replication kinetics of EHV1

59

SUMMARY

Equine herpesvirus 1 (EHV1) replicates in the respiratory tract of horses, after which

infected leukocytes transport virus throughout the body, resulting in abortion or nervous

system disorders. Two EHV1 strains circulate in the field: neurovirulent and non-

neurovirulent. To investigate differences in replication in the upper respiratory tract

(URT), an experimental inoculation study in ponies was performed with both strains. Two

groups of six ponies, were inoculated intranasally with 106.5

TCID50 of either strain.

Clinical signs, nasal shedding and viremia were evaluated. At early time points post

inoculation (pi), one pony of each group was euthanized. Tissues were collected for

titration and immunostainings. Number and size of EHV1-induced plaques were

calculated, and individual EHV1-infected cells were quantified and characterized.

Inoculation with either strain resulted in nasal shedding and replication in several tissues

of the URT. Both strains replicated in a plaquewise manner in epithelium of the nasal

mucosa, but replication in epithelium of the nasopharynx was largely limited to non-

neurovirulent EHV1. Plaques were never able to cross the basement membrane, but

individual infected cells were noticed in the connective tissue of all examined tissues for

both strains. The total number of these cells however, was 3-7 times lower with non-

neurovirulent EHV1 compared to neurovirulent EHV1. CD172a+ cells and CD5

+

lymphocytes were important target cells for both strains. Interestingly, in lymph nodes, B-

lymphocytes were also important target cells for EHV1, irrespective of the strain. Viremia

was detected very early pi and infected cells were mainly CD172a+ for both strains. In

summary, these results are valuable for understanding EHV1 pathogenesis at the port of

entry, the URT.

Replication kinetics of EHV1

60

1 INTRODUCTION

Infectious respiratory tract disease has been recognized as the cause of major health

problems in horses. The most important respiratory pathogen is equine herpesvirus 1

(EHV1) and infections with this virus cause serious economic losses in the horse industry

worldwide (Allen & Bryans, 1986; Ostlund, 1993; van Maanen, 2002). The upper

respiratory tract is the first line of defence against respiratory pathogens and is also the

primary replication site of EHV1, where it causes upper respiratory disorders (Kydd et

al., 1994a; b). In addition, EHV1 can spread via infected blood leukocytes to internal

organs, for example the pregnant uterus, causing symptoms such as abortion and neonatal

foal death (Allen & Bryans, 1986). EHV1 can also reach the central nervous system,

where replication in endothelial cells results in severe nervous system disorders with

frequent fatal outcome. Neurological disease in horses caused by infection with certain

isolates of EHV1 is a severe condition which is poorly understood (McCartan et al., 1995;

Stierstorfer et al., 2002; van der Meulen et al., 2003a). The pathogenesis for developing

this devastating condition is largely unknown and full protection against these secondary

and severe symptoms can not be obtained by vaccination with the currently available

vaccines (Goodman et al., 2006; Heldens et al., 2001; van der Meulen et al., 2007).

Therefore, an improvement of the existing vaccines and/or the development of alternative

strategies for the prevention and treatment of EHV1-induced infections are necessary.

Importantly, this implies a good understanding of the pathogenesis of EHV1 during the

acute phase of infection.

It has been suggested that distinct isolates of EHV1, differing in pathogenic capacity,

circulate in the field. Nugent et al. (2006) indicated that a single nucleotide polymorphism

(SNP) in the DNA polymerase is strongly associated with neurological versus non-

neurological disease outbreaks. Studies in naturally infected horses with nervous system

disorders have revealed a more robust cell-associated viremia in horses infected with

neurovirulent isolates of EHV1, in contrast to horses infected with non-neurovirulent

isolates (Allen & Breathnach, 2006). This was further confirmed by reverse genetic

experiments of EHV1 strains with a sole mutation of the DNA polymerase SNP, where

experimental inoculation with such strains altered neurologic disease in horses (Goodman

et al., 2007; Van de Walle et al., 2009). Moreover, in vitro experiments with these strains

revealed differences in leukocyte tropism which could explain the difference in clinical

Replication kinetics of EHV1

61

outcome (Goodman et al., 2007). Still, the exact identity of carrier cells susceptible to

EHV1 infection and hereby responsible for dissemination of infectious virus to sites of

secondary replication, is a matter of debate. Blood samples from experimentally infected

ponies were collected and examined for the presence of neurovirulent EHV1 (Scott et al.,

1983). T-lymphocytes were found to be the most important cell fraction to harbour virus

in vivo. However, no full characterization of these cells was performed. Different studies

with in vitro EHV1-infected leukocytes have also been performed to determine the

identity of these cells. Hereby, mainly monocytes were susceptible to infection with non-

neurovirulent EHV1 (van der Meulen et al., 2000). After stimulation with mitogens, T-

lymphocytes became more susceptible, followed by B-lymphocytes (van der Meulen et

al., 2001). Despite these studies, information on possible differences between

neurovirulent and non-neurovirulent EHV1 strains at the port of entry, and identity of

carrier cells during viremia remains limited.

To investigate this, six Shetland ponies were experimentally inoculated with a

neurovirulent EHV1 strain and six with a non-neurovirulent EHV1 strain. These animals

were subsequently euthanized to collect different tissues of the upper respiratory tract.

Tissues that were positive for EHV1 on titration were stained with markers for different

cell types to determine and quantify the carrier cells of EHV1 in the upper respiratory

tract. In addition, peripheral blood mononuclear cells (PBMC) were also collected to

quantify and identify the infected cell type during cell-associated viremia.

2 MATERIALS AND METHODS

2.1 Horses

Twelve male Shetland ponies, between the age of 6 months and 2 years, were used in

this study. They were housed inside isolated stables. They were fed daily with a

commercial, complete feed. Drinking water and hay were supplied ad libitum.

Prior to the experiment, ponies were monitored for 4 weeks. Rectal temperatures were

measured daily and complement-dependent seroneutralization (SN)-tests and

immunoperoxidase monolayer assays (IPMA) were performed weekly to determine EHV-

specific antibody titres.

Replication kinetics of EHV1

62

2.2 Virus and inoculation

Ponies were divided into two groups of 6. For the first group the Belgian EHV1 strain

03P37, isolated from the blood of a paralytic horse in 2003, was used for inoculation

(Garré et al., 2009; van der Meulen et al., 2003a). The second group was inoculated with

the Belgian strain 97P70, which was isolated from an aborted fetus (van der Meulen et

al., 2006). The strains were typed in the DNA polymerase as D752 and N752 respectively in

cooperation with the Animal Health Trust in the United Kingdom (Nugent et al., 2006).

Based on their origin and genotyping, these strains will be referred as neurovirulent and

non-neurovirulent in this paper. Virus stocks used for inoculation were at the 6th

passage;

2 passages in rabbit kidney cells (RK13) and 4 subsequent passages in equine embryonic

lung cells (EEL). Both viruses had virtually identical single-step growth properties in

RK13 cells for intra- and extracellular virus yields at all tested time points post infection

(data not shown).

Ponies were inoculated oronasally with a total amount of 20ml virus suspension

containing 106.5

tissue culture infectious dose 50 (TCID50). Ten ml of the virus suspension

was administered intranasally (5 ml per nostril) using a thin probe and 10 ml was

inoculated orally with a syringe. The virus titre was confirmed by titration of the

inoculum on RK13 cells.

2.3 Serological examination

For the seroneutralization (SN)-test, two-fold dilution series of the sera were prepared

in minimum essential medium (MEM). Fifty micro litre of these serial dilutions were

incubated for 23 hours (h) at 37°C with a fixed number of infectious virus (300 TCID50 of

EHV1 strain Arabica in 50 µl). Hereafter, 25 µl of guinea pig complement was added.

After 1 h of incubation, the mixture of serum, virus and complement was added to RK13

cells. Inoculated cultures were further incubated at 37°C in an atmosphere containing 5%

CO2. After 7 days of incubation, the cultures were analysed for the presence of cytopathic

effect. The neutralization titre was calculated as the reciprocal of the highest dilution of

the serum that was able to completely block EHV1 infection in RK13 cells.

For the immunoperoxidase monolayer assay (IPMA), RK13 cells were inoculated with

103 TCID50 of the EHV1 strain 97P70. After 28 h, cells were washed, dried at 37°C for 1

Replication kinetics of EHV1

63

h and stored at -20°C until use. Plates were thawed and subsequently fixed with 4%

paraformaldehyde and a solution containing 1% hydrogen peroxide in methanol.

Following extensive washing, serial 2-fold dilutions of the sera were added and cells were

incubated for 1 h at 37°C. Cells were incubated with peroxidase-labeled goat anti-horse Ig

(Jackson ImmunoResearch Laboratories Inc., PA, USA) and after 1 h, a substrate solution

of 3-amino-9-ethylcarbazole in 0.05 M acetate buffer with 0.05% hydrogen was added to

each well. Following 20 minutes of incubation at 37°C, substrate solution was replaced

with acetate buffer to block the enzymatic staining reaction. The IPMA titre was

calculated as the reciprocal value of the highest serum dilution that induced visual

staining of infected RK13 cells as determined by light microscopy.

2.4 Clinical examination

Horses were monitored daily for clinical signs by physical examination and

measurement of rectal temperatures. Fever was defined as a rectal temperature ≥ 38.5°C.

During clinical examination, nasal discharge (serous, mucous or purulent), lymph node

swelling, tachypnea and coughing were evaluated. In addition, neurological exams were

performed: walking the horse in a straight line, walking on and of a step, walking the

horse in tight circles, pulling the horse’s tail and backing the horse.

2.5 Virological examination

Nasopharyngeal mucus samples were taken daily until euthanasia. Immediately after

collection, swabs were immersed in transport medium containing phosphate-buffered

saline (PBS) supplemented with 10% fetal calf serum (FCS), 1000 U/ml penicillin, 1

mg/ml streptomycin and 0.5 mg/ml kanamycin. EHV1 in nasopharyngeal secretions was

titrated on monolayers of RK13 cells, exactly as described previously (van der Meulen et

al., 2003b).

On 1, 2, 3, 4, 5 and 7 days post inoculation, one pony of each group was euthanized

with an overdose of Natriumpentobarbital® (Kela, Hoogstraten, Belgium). After

euthanasia, different tissue samples were collected from the upper respiratory tract.

Tissues from slaughterhouse horses were collected and served as uninfected control

samples. Per tissue, two equal squares of 1 cm2 were collected from both left and right

Replication kinetics of EHV1

64

side of the head. From each side, one square was frozen immediately with

(immunofluorescence) or without (virus titration) methylcellulose medium

(Methocel®MC, Sigma-Aldrich, St. Louis, MO, USA) at -70°C. To determine viral

replication, 20% suspensions of collected tissues were made and titrated on RK13 cells.

Titres of infected tissues were determined after 7 days by means of Reed and Muench’s

formula (Reed & Muench, 1938).

The experimental design was approved by the local ethical committee of the Faculty

of Veterinary Medicine.

2.6 Quantification and characterization of individual infected cells in tissues of the

upper respiratory tract

Immunofluorescent double stainings were used to quantify and characterize individual

infected cells in tissues that were EHV1-positive after titration on RK13 cells. First,

cryosections (16 µm) were incubated with either monoclonal antibody (mAb) HT23A,

DH59B or 1.9/3.2 (VMRD Inc, Pullman, WA, USA) against CD5 (T-lymphocytes),

CD172a (cells from the monocyte lineage, CML) or IgM (B-lymphocytes) respectively,

and subsequently incubated with Texas Red®-labelled goat anti-mouse Abs (Molecular

Probes, Eugene, OR, USA). Epithelial cells were identified based on the pancytokeratin

marker MNF116 (Abcam, Cambridge, UK) and/or morphological features. Second, viral

EHV1 proteins were stained by incubation with biotinylated polyclonal horse anti-EHV1

IgG (van der Meulen et al., 2000), followed by streptavidin-FITC® (Molecular Probes,

Eugene, OR, USA). As controls, (i) stainings were performed on uninfected tissue and (ii)

isotype control antibodies were included. All cryosections were analyzed using confocal

microscopy (Leica TCS SP2 Laser scanning spectral confocal system, Leica

Microsystems GmbH, Wetzlar, Germany). Number and latitude of EHV1-induced

plaques were calculated using the software program ImageJ. Latitude was used to

evaluate plaque size and therefore the degree of cell-to-cell spread. In order to quantify

infected cells in nasal septum, two main regions were taken into account, on the one hand

regions with and on the other hand regions without plaques in the epithelium. These

regions were then subdivided into three equal regions underneath the epithelium: A, B

and C for regions with epithelial plaques and D, E and F for regions without epithelial

plaques (Fig 4). For nasopharynx, tubal/nasopharyngeal tonsils and lymph nodes,

Replication kinetics of EHV1

65

subdivisions into different zones of functional importance were made (Fig 5, 6 and 7

respectively). All regions were evaluated for the presence of plaques and/or individual

infected cells. In addition, the total number of infected cells (positive for staining with

anti-EHV1 Abs), equine immune cells (positive for staining with the mAbs against

immune cell markers) and infected equine immune cells (double positive) were

determined.

2.7 Quantification and characterization of viremia

Heparinized blood samples were taken daily until euthanasia and peripheral blood

mononuclear cells (PBMC) were isolated by density centrifugation on Ficoll-Paque,

according to manufacturer’s instructions (Pharmacia Biotech AB, Uppsala, Sweden). To

determine the magnitude of viremia, co-cultivation of PBMC on RK13 cells was

performed as previously described (van der Meulen et al., 2003b). Remaining PBMC

were resuspended in RPMI supplemented with 30% FCS and 20% Dimethyl Sulfoxide

(DMSO) and frozen in liquid nitrogen for later examination to identify the nature of

infected cells. Hereby, a double immunofluorescence staining was performed on acetone-

fixed cell smears of PBMC. EHV1 expression was detected using a biotinylated

polyclonal horse anti-EHV1 IgG (van der Meulen et al., 2003b), followed by streptavidin-

FITC®. The identity of infected cells was determined using the mAbs HT23A, DH59B or

1.9/3.2, as described above, followed by the incubation with Texas Red®-labelled goat

anti-mouse Abs. Samples were analyzed using confocal laser scanning microscopy.

3 RESULTS

3.1 EHV-status of the ponies before inoculation

The inclusion criterium for ponies used in this study was a complete absence of EHV-

specific antibodies by either SN-test (<2) or IPMA (<10). In addition, during the 4-week

observation period prior to the experiment, (i) no raise in temperature was noted, (ii) no

EHV-specific antibodies were detected, and (iii) no virus was isolated from

nasopharyngeal swabs. The use of naïve horses is an important tool to minimize variation

Replication kinetics of EHV1

66

in clinical and virological outcome due to differences in immunological status (Cornick et

al., 1990; Heldens et al., 2001; Matsumura et al., 1996).

3.2 Experimental inoculation with either strain induces fever and upper respiratory

tract disease in ponies

All ponies developed fever upon EHV1 inoculation with exception of the two ponies

that were euthanized 24 hours post inoculation (hpi). Fever started at 48 hpi for the

neurovirulent strain (5/5 animals) and at 36 (3/5 animals) or 48 hpi (2/5 animals) for the

non-neurovirulent strain. High temperatures lasted for the complete observation period,

with peak temperatures of 40.7°C for the neurovirulent strain and 40.1°C for the non-

neurovirulent strain. Serous nasal discharge was noted after inoculation with both strains,

starting at 1 day post inoculation (dpi). A transition to purulent nasal discharge was seen

in both groups from 3 dpi. Breathing problems, consisting of tachypnea or coughing, were

observed in two ponies of each group. Swelling of mandibular lymph nodes was observed

in the majority of the ponies and occurred from 1 dpi in 2/6 animals inoculated with a

neurovirulent strain and in 4/6 animals inoculated with a non-neurovirulent strain. Painful

retropharyngeal lymph nodes were noted in 5/6 ponies inoculated with a non-

neurovirulent strain, in contrast to only 2 ponies inoculated with a neurovirulent strain.

None of the ponies showed neurological signs, irrespective of the strain used for

inoculation. The latter is not too unexpected, as the incubation period of EHV1-induced

neurological disorders is normally 6 to 10 days (Edington et al., 1986; Jackson et al.,

1977; Mumford & Edington, 1980). Moreover, nervous system disorders have been

described to occur more frequently in older horses and certain breeds (Garré et al., 2009;

Goehring et al., 2006; Greenwood & Simson, 1980; Jackson et al., 1977; McCartan et al.,

1995).

Replication kinetics of EHV1

67

3.3 Experimental inoculation with either strain results in nasal shedding and viral

replication in the upper respiratory tract, but differences were observed in the

preferential epithelial replication site between strains

When evaluating nasal shedding, by titrations of nasal swab samples, five out of six

ponies inoculated with the neurovirulent strain shed virus from 1 dpi, with a peak

excretion at 2 dpi (Fig 1). Nasal shedding then slowly decreased until 7 dpi. For the non-

neurovirulent strain, 6 out of 6 ponies shed virus at 1 dpi, reaching a peak at 2 dpi with

remaining high titres until 7 dpi (Fig 1).

Figure 1. Nasal viral shedding. Ponies were inoculated with a neurovirulent or a non-neurovirulent strain

of EHV1. At 0, 1, 2, 3, 4, 5 and 7 days post inoculation (dpi), nasopharyngeal swabs were collected and

titrated as described in materials and methods. - : mean values

Next, different tissue samples were collected from the upper respiratory tract of one

pony per group per day for viral titrations. Virus was never isolated in negative control

tissues and could not be detected in any tissue at 1 dpi for the neurovirulent strain, but

was found in the nasopharynx of ponies inoculated with the non-neurovirulent strain (Fig

2). From 2 dpi, virus was mainly recovered from different parts of the nasal septum and

the nasopharynx for both strains (Fig 2). Septum and nasopharynx remained positive for

EHV1 until 7 dpi, with the exception of day 5 for the neurovirulent strain where no virus

0

2

4

6

8

0 1 2 3 4 5 7

Days post inoculation

Vir

us

tite

r (l

og

10

TC

ID5

0/g

ram

)

n=6

neurovirulent

non-neurovirulent

0

2

4

6

8

0 1 2 3 4 5 7

Days post inoculation

Vir

us

tite

r (l

og

10

TC

ID5

0/g

ram

)

n=6

neurovirulent

non-neurovirulent

neurovirulent

non-neurovirulent

Replication kinetics of EHV1

68

was detected in the nasopharynx (Fig 2) and day 4 for the non-neurovirulent strain where

the distal nasal septum was negative (Fig 2). No difference in virus titres could be noticed

in the nasal septum, but titres found in the nasopharynx seemed to be higher after

inoculation with the non-neurovirulent strain of EHV1 compared to the neurovirulent

strain (Fig 2). However, because of the limited number of animals used, we should be

careful to draw any conclusions based on these results. Virus was also found in lymphoid

tissues such as the tubal and nasopharyngeal tonsils and mandibular lymph nodes (Fig 2).

In general, titres found in tubal and nasopharyngeal tonsils (Fig 2) were higher for the

non-neurovirulent strain. Titres detected in mandibular lymph nodes started rising from 3

dpi (neurovirulent strain) or 4 dpi (non-neurovirulent strain), and at 7 dpi, mandibular

lymph nodes were positive for the non-neurovirulent strain only (Fig 2). EHV1 could also

be isolated for both strains at different time points pi from the ethmoid, the cranial part of

the trachea and 3 different tonsils (data not shown).

Replication kinetics of EHV1

69

Figure 2. Viral replication in different tissues of the upper respiratory tract. Ponies were inoculated

with either neurovirulent or non-neurovirulent EHV1 and euthanized at 0, 1, 2, 3, 4, 5 and 7 days post

inoculation (dpi). Different tissues were collected and titrated as described in materials and methods.

Finally, replication of EHV1 was evaluated in the epithelium of different tissues of

the upper respiratory tract by immunofluorescence. Hereby, EHV1-induced plaques were

analyzed and plaque latitudes measured. In the epithelium of the entire nasal septum,

EHV1-induced plaques were seen at different time points pi for both strains (Fig 3A).

These plaques were defined as groups of infected epithelial cells. Since results for

proximal, intermedial and distal nasal septum were virtually identical, only results for the

intermedial nasal septum are shown. Plaques were not present in the epithelium at 0 and 1

dpi (Fig 3A). However, individual infected cells were observed at 1 dpi, which were

identified as CD172a+ cells, as determined by the marker DH59B, or epithelial cells, as

determined by the pancytokeratin marker (data not shown). For the neurovirulent strain,

0

2

4

6

0 1 2 3 4 5 7

0

2

4

6

0 1 2 3 4 5 7

Vir

us

tite

r (l

og

10

TC

ID5

0/g

ram

tis

sue)

Days post inoculation

0

2

4

6

0 1 2 3 4 5 7

Intermedial nasal septum

0

2

4

6

0 1 2 3 4 5 7

Intermedial nasal septum

Tubal and nasopharyngeal tonsils

0

2

4

6

0 1 2 3 4 5 7

Distal nasal septum

0

2

4

6

0 1 2 3 4 5 7

Distal nasal septum

0

2

4

6

0 1 2 3 4 5 7

Proximal nasal septum

0

2

4

6

0 1 2 3 4 5 7

Proximal nasal septum

Mandibular lymph nodes

0

2

4

6

0 1 2 3 4 5 7

Nasopharynx

0

2

4

6

0 1 2 3 4 5 7

Nasopharynx

neurovirulent

non-neurovirulent

neurovirulent

non-neurovirulent

Replication kinetics of EHV1

70

EHV1-induced plaques were observed in the epithelium of the nasal mucosa starting from

2 till 7 dpi (Fig 3A). At 2, 3 and 4 dpi, these plaques consisted of epithelial cells and

rarely one or two CD172a+ cells. From 5 dpi, EHV1-positive cells became separated from

the basement membrane (BM) and these cells were exclusively positive for the DH59B

marker which identifies cells of the monocytic lineage (CML) (Fig 3A). This observation

was not seen in ponies inoculated with a non-neurovirulent strain. There, plaques were

present in the epithelium of the nasal mucosa and nasopharynx from 2 till 7 dpi (Fig 3A &

B). These plaques consisted of epithelial cells and rarely some CD172a+ cells, but no

separation of EHV1-positive cells was observed (Fig 3A). No differences were observed

in both number (data not shown) and size (Fig 3A) of EHV1-induced plaques in the

intermedial nasal mucosa at different time points pi for both strains. In the epithelium of

the nasopharynx, plaques could be found for the non-neurovirulent strain from 2 till 7 dpi,

with the exception of 4 dpi (Fig 3B). In contrast, no plaques were found for the

neurovirulent strain in the nasopharyngeal epithelium, with the exception of one small

plaque at 3 dpi (Fig 3B). Interestingly, plaques were not able to cross the BM at any time

point pi, but single EHV1-infected cells could be noticed in the connective tissue from 1

dpi. Plaques or individual cells could never be visualised in uninfected control tissues or

in tissues stained with isotype control antibodies.

Replication kinetics of EHV1

71

Figure 3. Plaque formation in epithelium of intermedial nasal septum and nasopharynx. Tissues of the

intermedial part of the nasal septum (A) and nasopharynx (B) were collected and latitude of plaques were

determined. For each tissue, ten consecutive sections were made and the total number of individual plaques

was determined. This number is presented above the relevant histogram. The largest measurement of

latitude of each individual plaque was used to determine plaque latitude. The sum was used to calculate the

mean plaque latitude ± SEM. Representative immunofluorescence pictures of EHV1-induced plaques in the

intermedial nasal septum are shown for neurovirulent or non-neurovirulent EHV1 for each dpi. BM:

basement membrane, objective 20x, NI: non-infected epithelial cells.

A

neuro-

virulent

B

Days post inoculation

Days post inoculation

Pla

que

lati

tud

e(µ

m)/

0.8

mm

2

NI NI

0

20

40

60

80

100

120

140

160

0 1 2 3 4 5 7

neurovirulent

non-neurovirulentP

laq

ue

lati

tud

e(µ

m)/

0.8

mm

2

neurovirulent

non-neurovirulent

0

20

40

60

80

100

120

140

160

0 1 2 3 4 5 7

non-

neuro-

virulent

BM

BM

7

3

2

9

10

31

12

12

2

48

1

A

neuro-

virulent

B

Days post inoculation

Days post inoculation

Pla

que

lati

tud

e(µ

m)/

0.8

mm

2

NI NI

0

20

40

60

80

100

120

140

160

0 1 2 3 4 5 7

neurovirulent

non-neurovirulentP

laq

ue

lati

tud

e(µ

m)/

0.8

mm

2

neurovirulent

non-neurovirulent

0

20

40

60

80

100

120

140

160

0

20

40

60

80

100

120

140

160

0 1 2 3 4 5 7

non-

neuro-

virulent

BM

BM

7

3

2

9

10

31

12

12

2

48

1

Replication kinetics of EHV1

72

Taken together, these results show that EHV1 infection results in nasal shedding and

replication in several tissues of the upper respiratory tract, irrespective of its neurovirulent

potential. However, whereas both types of EHV1 replicate equally in the epithelium of

the nasal mucosa, replication in the nasopharynx was limited to non-neurovirulent EHV1.

3.4 Neurovirulent EHV1 uses mainly CD172a+ cells as carrier cells in tissues of the

upper respiratory tract, whereas non-neurovirulent EHV1 shows a tropism for

both CD172a+ cells and CD5

+ cells

As mentioned before, EHV1-induced plaques never crossed the BM, irrespective of

the strain used. However, individual infected cells were observed in the underlying

connective tissue and the total number was 3-7 times lower with non-neurovirulent EHV1

compared to neurovirulent EHV1. Individual infected cells were never observed in

uninfected control tissues or in tissues stained with isotype control antibodies.

To identify the nature of these cells, cryosections of tissues were made and double

immunofluorescence stainings were performed.

Replication kinetics of EHV1

73

Nasal septum. In the connective tissue of the nasal septum, no differences were

observed in the total amount of infected cells present beneath epithelium with plaques or

without plaques, with the exception of 5 and 7 dpi for the neurovirulent strain, where a

marked increase in infected cells was observed in region A (Fig 4). Such increase was not

seen in the regions located underneath epithelium without plaques (Fig 4).

Figure 4. Quantification and identification of single EHV1-infected cells in different zones of the

intermedial nasal septum for neurovirulent and non-neurovirulent strains. Every tenth section of a

serially sectioned block of tissue was collected. Ten sections were analysed for each cell marker separately,

giving a total number of 30 sections. The total number of infected cells present in the intermedial nasal

septum per zone and per day, is shown in the upper right table. The results for the identification of single

infected cells are shown in the lower graphs and are represented as the mean of ten sections ± SEM. ROI:

region of interest.

B

A

C F

E

D

a

b

plaque

d

c

d

Strain Roi Total number of single infected cells

present at … dpi/30 sections

0 1 2 3 4 5 7

A - - 34 17 34 137 82

B - - 52 13 62 8 60

Neurovirulent

C - - 24 8 40 4 18

D 0 3 24 34 62 11 9

E 0 0 24 10 58 11 5

F 0 0 28 9 22 8 2

A - - 3 0 4 1 0

B

C

D

E

F

Non-

neurovirulent - - 4 3 15 0 0

- - 0 1 0 0 0

0 0 6 0 19 8 0

0 0 0 0 17 6 0

0 0 0 0 25 3 0

CD5+

CD172a+

IgM+

CD5+CD5+

CD172a+CD172a+

IgM+IgM+

of

infe

cted

cell

s/1

0 s

ecti

on

s

Days post inoculation

Schematic overview of the structure of the nasal

septum and different counting regions

a: epithelium; b: connective tissue; c: glands;

d: blood vessel

CD5+

CD172a+

IgM+

Days post inoculation

0

10

20

30

40

50

60

70

80

90

0 1 2 3 4 5 7

CD5+

CD172a+

IgM+

CD5+CD5+

CD172a+CD172a+

IgM+IgM+

Days post inoculation

0

10

20

30

40

50

60

70

80

90

0 1 2 3 4 5 70

10

20

30

40

50

60

70

80

90

0 1 2 3 4 5 7

Neurovirulent strain Non-neurovirulent strain

Replication kinetics of EHV1

74

Nasopharynx. EHV1-infected cells were present in the connective tissue of the

nasopharynx from 1 till 7 dpi, irrespective of the strain used, however, the localization of

these cells appeared to be strain-dependent (Fig 5). Whereas for the neurovirulent strain,

single infected cells were mainly found in epithelium (region a) and connective tissue

(region b), single infected cells were located in connective tissue (region b) and

lymphocyte aggregates (region c) for the non-neurovirulent strain (Fig 5).

Figure 5. Quantification and identification of single EHV1-infected cells in different zones of the

nasopharynx for neurovirulent and non-neurovirulent strains. Every tenth section of a serially

sectioned block of tissue was collected. Ten sections were analysed for each cell marker separately, giving a

total number of 30 sections. The total number of infected cells present in the nasopharynx per zone and per

day, is shown in the upper right table. The results for the identification of single infected cells are shown in

the lower graphs and are represented as the mean of ten sections ± SEM. ROI: region of interest.

a

b

d

c

e

Strain Roi Total number of single infected cells

present at … dpi/30 sections

0 1 2 3 4 5 7

a 0 3 3 7 2 0 1

b 0 22 24 118 22 18 23

Neurovirulent

c 0 0 0 0 0 0 0

d 0 0 0 0 0 0 0

e 0 0 0 0 0 0 0

a 0 0 0 0 0 0 0

b 0 1 320 15 28 18 1

Non-

neurovirulent

c 0 0 0 0 99 16 1

d 0 0 0 0 0 0 0

e 0 0 0 0 0 1 0

CD5+

CD172a+

IgM+

CD5+CD5+

CD172a+CD172a+

IgM+IgM+

Days post inoculation

CD5+

CD172a+

IgM+

CD5+CD5+

CD172a+CD172a+

IgM+IgM+

Days post inoculation

c

Schematic overview of the structure of the

nasopharynx and different counting regions

a: epithelium; b: connective tissue; c: lymphocyte

aggregate; d: deeper connective tissue; e: blood vessels

Neurovirulent strain Non-neurovirulent strain

0

5

10

15

20

25

30

35

40

0 1 2 3 4 5 70

5

10

15

20

25

30

35

40

0 1 2 3 4 5 7

of

infe

cted

cell

s/10

sec

tion

s

Replication kinetics of EHV1

75

Tubal and nasopharyngeal tonsils. For both EHV1 strains, single infected cells were

observed starting from 2 dpi (Fig 6). They were mostly present in the connective tissue

underneath the epithelium (region b) and lymphocyte aggregates (region c, Fig 6), but

single infected cells could also be observed in epithelium/fundus (region a), deeper

connective tissue (region d), blood vessels (region e) and glands (region f) (Fig 6).

Figure 6. Quantification and identification of single EHV1-infected cells in different zones of the tubal

and nasopharyngeal tonsils for neurovirulent and non-neurovirulent strains. Every tenth section of a

serially sectioned block of tissue was collected. Ten sections were analysed for each cell marker separately,

giving a total number of 30 sections. The total number of infected cells present in the tubal and

nasopharyngeal tonsils per zone and per day, is shown in the upper right table. The results for the

identification of single infected cells are shown in the lower graphs and are represented as the mean of ten

sections ± SEM. ROI: region of interest.

Strain Roi Total number of single infected cells

present at … dpi/30 sections

0 1 2 3 4 5 7

a 0 0 5 77 95 0 14

b 0 0 6 273 538 1 51

Neurovirulent

c 0 0 3 260 241 2 0

d 0 0 0 86 150 2 7

e 0 0 0 133 85 0 2

f 0 0 0 38 117 1 13

a

b

d

c

e

f

a 0 0 7 23 7 18 5

b 0 0 39 173 51 50 2

Non-

neurovirulent

c 0 0 10 34 31 487 8

d 0 0 44 78 14 8 2

e 0 0 0 5 3 0 2

f 0 0 2 0 2 4 13

CD5+

CD172a+

IgM+

CD5+CD5+

CD172a+CD172a+

IgM+IgM+

Days post inoculation

CD5+

CD172a+

IgM+

CD5+CD5+

CD172a+CD172a+

IgM+IgM+

Days post inoculation

Schematic overview of the structure of tubal

and nasopharyngeal tonsils and different

counting regions

a: epithelium; b: connective tissue; c: lymphoid aggregate;

d: deeper connective tissue; e: blood vessel; f: glands

Neurovirulent strain Non-neurovirulent strain

0

50

100

150

200

250

300

350

400

0 1 2 3 4 5 70

50

100

150

200

250

300

350

400

0 1 2 3 4 5 7

of

infe

cted

cell

s/10 s

ect

ions

Replication kinetics of EHV1

76

Mandibular lymph nodes. For the neurovirulent strain, most of the infected cells

were found in the marginal sinus of the mandibular lymph nodes (region c, Fig 7). Their

number increased until 4 dpi and rapidly decreased afterwards, with rarely any infected

cells left at 7 dpi. Furthermore, infected cells were also observed in the capsule and

trabecula (region a), the medulla (region f) and in fewer amounts in vessels of the capsule

and the cortical nodules (regions b and d, Fig 7). In these regions a peak number of

infected cells was observed at 4 and 5 dpi (Fig 7). For the non-neurovirulent strain, single

cells were only found from 4 till 7 dpi and were mostly found in the marginal sinus

(region c) and medulla (region f) (Fig 7).

Figure 7. Quantification and identification of single EHV1-infected cells in different zones of the

mandibular lymph node for neurovirulent and non-neurovirulent strains. Every tenth section of a

serially sectioned block of tissue was collected. Ten sections were analysed for each cell marker separately,

giving a total number of 30 sections. The total number of infected cells present in the mandibular lymph

node per zone and per day, is shown in the upper right table. The results for the identification of single

infected cells are shown in the lower graphs and are represented as the mean of ten sections ± SEM. ROI:

region of interest.

Strain Roi Total number of single infected cells

present at … dpi/30 sections

0 1 2 3 4 5 7

a 0 0 0 40 10 162 1

b 0 12 2 16 40 12 0

Neurovirulent

c 0 17 6 106 846 170 34

d 0 2 2 31 36 4 19

e 0 0 0 3 1 0 0

f 0 0 9 49 70 14 39

a

b

c d

e f

a

b

c d

e f

CD5+

CD172a+

IgM+

CD5+CD5+

CD172a+CD172a+

IgM+IgM+

Days post inoculation

CD5+

CD172a+

IgM+

CD5+CD5+

CD172a+CD172a+

IgM+IgM+

Days post inoculation

a 0 0 0 0 4 5 1

b 0 0 0 0 0 0 0

Non-

neurovirulent

c 0 1 2 4 40 39 1

d 0 0 0 0 5 17 8

e 0 0 0 0 0 0 0

f 0 0 0 0 41 82 1

Schematic overview of the structure of the

mandibular lymph node and different counting

regions

a: capsule and trabecula; b: vessels of the capsule; c:

marginal sinus; d: cortical nodule; e: vessels in medulla;

f: medullaNeurovirulent strain Non-neurovirulent strain

0

50

100

150

200

250

300

350

400

0 1 2 3 4 5 70

50

100

150

200

250

300

350

400

0 1 2 3 4 5 7

of

infe

cted

cell

s/10

sec

tio

ns

Replication kinetics of EHV1

77

Immunofluorescence stainings, using mAbs against immune cell surface markers

showed that in all tissues of the upper respiratory tract, infected cells were largely

CD172a+ CML, followed by a slightly lower amount of infected CD5

+ lymphocytes when

the neurovirulent EHV1 strain was used for inoculation (Table I). These results were very

similar to what was observed with the non-neurovirulent strain, although a more

substantial amount of infected CD5+ lymphocytes was observed in these different tissues

of the upper respiratory tract (Table I). IgM+ lymphocytes were always present, but rarely

infected with the exception of the mandibular (Table I) and retropharyngeal (data not

shown) lymph nodes, were these cells were an important target cell type for both strains.

Table I. Percentage of marker positive EHV1-infected individual cells in different tissues of the upper

respiratory tract

Percentage of marker positive EHV1-infected individual cells

Neurovirulent strain Non-neurovirulent strain

Region of interest

CD172a+ CD5

+ IgM

+ CD172a

+ CD5

+ IgM

+

Nasal septum 53.4 ± 22,8 16.2 ± 11.7 0.3 ± 0.8 43.9 ± 12.8 23.6 ± 8.6 0

Nasopharynx 69.5 ± 20.7 0 8.3 ± 20.4 37.9 ± 35.5 13.6 ± 5.7 1.1 ± 2.7

Tubal and

nasopharyngeal tonsils

66.4 ± 20.7 11.2 ± 12.8 4.8 ± 5.6 28.7 ± 3.6 21.2 ± 5.9 0.5 ± 1.0

Mandibular lymph

nodes

71.3 ± 17.9 7.3 ± 9.5 8.3 ± 8.8 42.6 ± 21 26.7 ± 5.7 7.7 ± 10.8

3.5 Ponies became viremic upon experimental inoculation with either strain, and

CD172a+ cells were the most important carrier cells in the blood

Transmission of EHV1 from PBMC to susceptible RK13 cells, as determined by co-

cultivation, was observed in 5 out of 6 ponies upon experimental inoculation with

neurovirulent EHV1 (Fig 8). No differences in the amount of infected PBMC was noted,

except for one pony, which showed a higher level of viremia at 4 and 5 dpi compared to

the other infected ponies (Fig 8). For the non-neurovirulent strain, viremia was observed

in all six infected ponies, with no differences in magnitude for the first 4 dpi (Fig A).

Replication kinetics of EHV1

78

However, at day 5 and 7 pi, the non-neurovirulent strain seemed to retain a higher viremia

level then the neurovirulent strain, but due to the low amount of animals left at those

days, no conclusions could be drawn (Fig 8).

Figure 8. Quantification of viremia for neurovirulent and non-neurovirulent strains. Ponies were

inoculated with a neurovirulent or a non-neurovirulent strain of EHV1. At 0, 1, 2, 3, 4, 5 and 7 days post

inoculation (dpi), blood samples were collected and cocultivation of PBMC was performed as mentioned in

materials and methods.

In addition, around 2 x 107 mononuclear cells for each strain were analyzed by

confocal microscopy to detect infected cells and further characterize their nature. These

cells were a mixture of PBMC from three ponies collected at 2, 3 and 4 dpi. For the

neurovirulent strain, fourteen EHV1-infected cells were found and eleven were

n=6

neurovirulent

non-neurovirulent

0

10

20

30

40

0 1 2 3 4 5 7

Days post inoculation

Nu

mb

ero

f p

laques

/ 1

07

PB

MC

n=6

neurovirulent

non-neurovirulent

neurovirulent

non-neurovirulent

0

10

20

30

40

0

10

20

30

40

0 1 2 3 4 5 7

Days post inoculation

Nu

mb

ero

f p

laques

/ 1

07

PB

MC

Replication kinetics of EHV1

79

characterized as CD172a+ cells (78.5%), whereas three were CD5

+ (21.5%). No infected

B-lymphocytes were found. For the non-neurovirulent strain, nine EHV1-infected cells

were identified, consisting of seven CD172a+ cells (77.8%), one CD5

+ T-lymphocyte

(11.0%) and one cell remained unidentified. Also here, no infected B-lymphocytes were

found.

These data indicate that CD172a+ cells appear to be the main target cell during EHV1-

induced viremia, irrespective of the neurovirulency of EHV1.

4 DISCUSSION

Despite several studies conducted on the pathogenesis of equine herpesvirus 1

(EHV1), it still remains unclear how this virus invades the tissues of the upper respiratory

tract and invades the deeper tissues to initiate a cell-associated viremia. Furthermore, two

strains of EHV1 circulate in the field, namely non-neurovirulent and neurovirulent strains,

the latter causing neurological disease, a severe condition which is poorly understood. In

the present study, we used the two strains of EHV1 to gain more insight into the

pathogenesis of these strains during the acute phase of infection in the upper respiratory

tract in vivo. To this end, Shetland ponies were experimentally inoculated with either a

non-neurovirulent or a neurovirulent EHV1 strain and upon euthanasia, several tissues

from the upper respiratory tract were collected and analyzed.

It was found that EHV1 has different approaches to successfully establish an infection

in the upper respiratory tract. Firstly, we observed that EHV1, irrespective of the strain,

can replicate in the epithelium of the nasal septum and/or nasopharynx by means of virus-

induced plaques. This replication appears to occur in a restricted way, as no increase in

number or size of plaques was observed over time. In addition, a repulsion of EHV1-

induced plaques was observed from 5 dpi onwards for the neurovirulent strain, and a

decrease in plaque latitude was seen for the non-neurovirulent strain starting at day 3 pi.

This reduction in replication might be attributed to the production of interferon (IFN), as

(i) the presence of interferon in nasal secretions after experimental inoculation of horses

with EHV1 has previously been described and (ii) peak concentrations of IFN-α were

noted at day 4 and day 7 pi (Bridges & Edington, 1986; Edington et al., 1989). In the

present study, IFN-α was also found in nasal swabs of experimentally infected ponies

(data not shown), which further points towards the importance of IFN to limit EHV1

Replication kinetics of EHV1

80

replication in the epithelium of the upper respiratory tract. Interestingly, even though the

non-neurovirulent strain was able to replicate in a plaquewise manner in the epithelium of

the nasopharynx, no plaques could be observed at any time point pi in the nasopharynx

after inoculation with the neurovirulent strain. As the epithelial structure of both tissues is

identical, with the exception of the presence of lymphoid aggregates in the nasopharynx,

the reason for this observation remains elusive.

Another interesting observation for both EHV1 strains was that we were unable to

observe EHV1-infected plaques under the basement membrane (BM) at any time point pi,

indicating that EHV1 does not reach the underlying tissue by breaking down the BM.

This is in striking contrast to previous findings with another and closely related

alphaherpesvirus, pseudorabies virus (PRV), which has been shown to cross the BM in a

plaque wise manner between 12 and 24 hpi in a porcine respiratory explant model

(Glorieux et al., 2009). This indicates that different alphaherpesviruses may use different

mechanisms to invade the underlying connective tissue. Indeed, whereas PRV was shown

to break down the BM and replicates further in the underlying tissue in a plaquewise

manner, our study with EHV1 demonstrated only single infected cells below the BM.

This is in agreement with previous studies on EHV1, where the presence of single

infected cells in tissues of the upper respiratory tract has been reported (Edington et al.,

1986; Kydd et al., 1994b). We could further demonstrate that these infected cells in the

underlying tissue were immune cells (as discussed in more detail below), which makes it

tempting to speculate that immune cells in the epithelium become infected with EHV1

and are responsible for transporting the virus to deeper connective tissue. Indeed, we did

found EHV1-infected cells in the epithelium of nasal septum and/or nasopharynx, which

were further characterized to belong to the monocytic lineage (CML) by using the cell

surface maker CD172a.

Not only differences concerning the identity of the EHV1-infected immune cells

present in the underlying tissues were noticed between both strains, moreover, differences

concerning their localization were also observed. When analyzing the identity of the

single EHV1-infected cells in the connective tissue of nasal septum and nasopharynx,

several differences could be observed between the two EHV1 strains. Firstly, the total

number of infected cells in the nasal septum was 10 to 20 times higher for the

neurovirulent strain compared to the non-neurovirulent strain. Secondly, while individual

infected cells in the nasopharynx were mainly present in the connective tissue upon

Replication kinetics of EHV1

81

neurovirulent EHV1 inoculation, individual cells infected with the non-neurovirulent

EHV1 seemed to migrate deeper into the lymphoid follicles of the nasopharynx in a time

wise manner. Thirdly, CD172a+ CML were the most important carrier cells for both

neurovirulent and non-neurovirulent EHV1, although these cells seemed to be more

important for the neurovirulent strain. In addition, both strains were able to infect CD5+

T-lymphocytes, with a higher amount of infected cells for the non-neurovirulent strain

(Table I). When analyzing the data on the identity of infected cells in lymphoid tissues of

the upper respiratory tract, we again observed that the most prominent target cells for

EHV1 were CD172a+ CML, irrespective of the strain used. This is in line with previous

studies, where EHV1-infected macrophages and lymphoblasts were detected in lymph

nodes after inoculation with EHV1 isolates from both an aborted fetus and a paralyzed

mare (Patel et al., 1982). Moreover, another EHV1 experimental inoculation study

showed that lymphocytes, monocytes, macrophages and plasma cells in interstitium and

lymph nodes all expressed EHV1 antigens, as determined by immunohistochemical

staining using anti-EHV1 polyclonal Abs (Kydd et al., 1994b). In the present study, the

majority of EHV1-positive cells was found in the marginal sinus of lymph nodes. At

those sites, transport is possible to extra follicular interdigitating cells in the lymph node

where both mature and virgin lymphocytes have access to antigen, hereby initiating

humoral immune responses (Tew et al., 1990). Surprisingly, and in contrast to what was

observed in nasal septum and nasopharynx, IgM+ B-lymphocytes were also an important

target cell for EHV1. In vitro it has been previously shown that a 6- to 14-fold increase of

EHV1-infected peripheral blood mononuclear cells (PBMC) could be obtained after in

vitro stimulation with proliferating drugs such as pokeweed mitogen (PWM),

concanavalin A (ConA), phytohaemagglutinin (PHA), or ionomycin and phorbol

dibutyrate (IONO/PDB) (van der Meulen et al., 2001). Together with the fact that B-

lymphocytes are known to undergo extensive proliferation in lymphoid follicles (Fu &

Chaplin, 1999), we would like to reason that the elevated percentage of EHV1-infected B-

lymphocytes observed in lymphoid tissues, is due to the proliferation stage of these

immune cells.

The onset of a cell-associated viremia could be detected as early as one day after

experimental inoculation with EHV1, indicating that EHV1-infected cells can enter the

blood stream very rapidly. Moreover, we identified these EHV1-positive cells as mainly

CD172a+ CML. Despite the fact that only 14 infected cells were found, we strongly feel

Replication kinetics of EHV1

82

that this low number is of relevance, as it is known that EHV1-induced viremia is

generally very low with numbers ranging from 1 to 10 positive cells/0.8 x 107 PBMC

(van der Meulen et al., 2006). Combining the results on the identity of EHV1-infected

cells in the connective tissues of the primary port of entry, the upper respiratory tract,

with the results on the identity of the EHV1-infected cells in the blood, we would like to

hypothesize that the CD172a+ cells we observed could be dendritic cells (DCs). The

reasoning behind such hypothesis is as follows. The marker DH59B that was used in the

present study to identify CD172a+ cells is a pan granulocyte/monocyte marker which has

been used in several studies as a DC marker (Ahn et al., 2002; Tumas et al., 1994). In

addition, there is evidence indicating that local tissue DCs are able to re-enter the

bloodstream, which might facilitate the spread of pathogens from tissue to tissue, carried

by DCs serving as Trojan horses (Randolph et al., 2008). Finally, DCs have been shown

to play an important role during the pathogenesis of alphaherpesviruses in general

(Bosnjak et al., 2005; Novak & Peng, 2005; Pollara et al., 2005), and for EHV1 in

specific, both murine and equine blood-derived DCs are susceptible to EHV1 in vitro

(Siedek et al., 1999; Steinbach et al., 1998). Unfortunately, little is known about equine

DCs in general and mucosal DCs in specific in this species, and more research is needed

to elucidate the role of DCs in EHV1 infection and/or other viral infections in equines.

Regardless, the results presented here are in striking contrast to another in vivo study,

where mainly blood T-lymphocytes were found to harbour EHV1 (Scott et al., 1983).

However, in the latter study, leukocyte populations were solely separated by glass-bead

columns and no direct characterization was performed. A possible explanation for the

discrepancy between cell types infected during cell-associated viremia could be the strain

used. In a previous study an American strain, isolated from an aborted fetus, was used

(Scott et al., 1983) whereas in the present study, we used two Belgian EHV1 strains.

Surprisingly, we observed CD172a+ cells as the main carrier cell type for both the

neurovirulent and the non-neurovirulent strain, indicating that the EHV1 strain does not

seem to correlate with a difference in cell tropism during viremia. This is again in contrast

to another study, where a difference in leukocyte tropism between neuro- and non-

neurovirulent EHV1 strains has been demonstrated (Goodman et al., 2007). Hereby it was

observed that the EHV1 strain Ab4, which has the neurovirulent genotype, mainly

infected CD4+ lymphocytes, whereas mutating this strain into the non-neurovirulent

genotype resulted in a preferential infection of monocytes and B-cells. However, these

Replication kinetics of EHV1

83

experiments were performed with in vitro infected PBMC, which most likely explains the

discrepancy with our in vivo data.

Acknowledgements

This research was supported by the Institute for the promotion of Innovation through

Science and Technology in Flanders (IWT-Vlaanderen). The authors like to thank C.

Boone and N. Monteiro for titrations, and C. Bracke for isolation of PBMC and

performing SN-tests.

Replication kinetics of EHV1

84

References

Ahn, J. S., Konno, A., Gebe, J. A., Aruffo, A., Hamilton, M. J., Park, Y. H. & Davis, W. C. (2002). Scavenger receptor cysteine-rich domains 9 and 11 of WC1 are

receptors for the WC1 counter receptor. J Leukoc Biol 72, 382-390.

Allen, G. P. & Breathnach, C. C. (2006). Quantification by real-time PCR of the

magnitude and duration of leucocyte-associated viraemia in horses infected with

neuropathogenic vs. non-neuropathogenic strains of EHV-1. Equine Vet J 38, 252-

257.

Allen, G. P. & Bryans, J. T. (1986). Molecular epizootiology, pathogenesis, and

prophylaxis of equine herpesvirus-1 infections. Prog Vet Microbiol Immunol 2,

78-144.

Bosnjak, L., Jones, C. A., Abendroth, A. & Cunningham, A. L. (2005). Dendritic cell

biology in herpesvirus infections. Viral Immunol 18, 419-433.

Bridges, C. G. & Edington, N. (1986). Innate immunity during Equid herpesvirus 1

(EHV-1) infection. Clin Exp Immunol 65, 172-181.

Cornick, J., Martens, J., Martens, R., Crandell, R., McConnell, S. & Kit, S. (1990). Safety and efficacy of a thymidine kinase negative equine herpesvirus-1 vaccine

in young horses. Can J Vet Res 54, 260-266.

Edington, N., Bridges, C. G. & Griffiths, L. (1989). Equine interferons following

exposure to equid herpesvirus-1 or -4. J Interferon Res 9, 389-392.

Edington, N., Bridges, C. G. & Patel, J. R. (1986). Endothelial cell infection and

thrombosis in paralysis caused by equid herpesvirus-1: equine stroke. Arch Virol

90, 111-124.

Fu, Y. X. & Chaplin, D. D. (1999). Development and maturation of secondary lymphoid

tissues. Annu Rev Immunol 17, 399-433.

Garré, B., Gryspeerdt, A., Croubels, S., De Backer, P. & Nauwynck, H. (2009). Evaluation of orally administered valacyclovir in experimentally EHV1-infected

ponies. Vet Microbiol 135, 214-221.

Glorieux, S., Favoreel, H. W., Meesen, G., de Vos, W., Van den Broeck, W. & Nauwynck, H. J. (2009). Different replication characteristics of historical

pseudorabies virus strains in porcine respiratory nasal mucosa explants. Vet

Microbiol 136, 341-346.

Goehring, L. S., van Winden, S. C., van Maanen, C. & Sloet van Oldruitenborgh-Oosterbaan, M. M. (2006). Equine herpesvirus type 1-associated

myeloencephalopathy in The Netherlands: a four-year retrospective study (1999-

2003). J Vet Intern Med 20, 601-607.

Goodman, L. B., Loregian, A., Perkins, G. A., Nugent, J., Buckles, E. L., Mercorelli,

B., Kydd, J. H., Palu, G., Smith, K. C., Osterrieder, N. & Davis-Poynter, N. (2007). A point mutation in a herpesvirus polymerase determines

neuropathogenicity. PLoS Pathog 3, 1583-1592.

Goodman, L. B., Wagner, B., Flaminio, M. J., Sussman, K. H., Metzger, S. M., Holland, R. & Osterrieder, N. (2006). Comparison of the efficacy of inactivated

combination and modified-live virus vaccines against challenge infection with

neuropathogenic equine herpesvirus type 1 (EHV-1). Vaccine 24, 3636-3645.

Greenwood, R. E. & Simson, A. R. (1980). Clinical report of a paralytic syndrome

affecting stallions, mares and foals on a thoroughbred studfarm. Equine Vet J 12,

113-117.

Replication kinetics of EHV1

85

Heldens, J. G., Hannant, D., Cullinane, A. A., Prendergast, M. J., Mumford, J. A.,

Nelly, M., Kydd, J. H., Weststrate, M. W. & van den Hoven, R. (2001). Clinical and virological evaluation of the efficacy of an inactivated EHV1 and

EHV4 whole virus vaccine (Duvaxyn EHV1,4). Vaccination/challenge

experiments in foals and pregnant mares. Vaccine 19, 4307-4317.

Jackson, T. A., Osburn, B. I., Cordy, D. R. & Kendrick, J. W. (1977). Equine

herpesvirus 1 infection of horses: studies on the experimentally induced

neurologic disease. Am J Vet Res 38, 709-719.

Kydd, J. H., Smith, K. C., Hannant, D., Livesay, G. J. & Mumford, J. A. (1994a). Distribution of equid herpesvirus-1 (EHV-1) in respiratory tract associated

lymphoid tissue: implications for cellular immunity. Equine Vet J 26, 470-473.

Kydd, J. H., Smith, K. C., Hannant, D., Livesay, G. J. & Mumford, J. A. (1994b). Distribution of equid herpesvirus-1 (EHV-1) in the respiratory tract of ponies:

implications for vaccination strategies. Equine Vet J 26, 466-469.

Matsumura, T., O'Callaghan, D. J., Kondo, T. & Kamada, M. (1996). Lack of

virulence of the murine fibroblast adapted strain, Kentucky A (KyA), of equine

herpesvirus type 1 (EHV-1) in young horses. Vet Microbiol 48, 353-365.

McCartan, C. G., Russell, M. M., Wood, J. L. & Mumford, J. A. (1995). Clinical,

serological and virological characteristics of an outbreak of paresis and neonatal

foal disease due to equine herpesvirus-1 on a stud farm. Vet Rec 136, 7-12.

Mumford, J. A. & Edington, N. (1980). EHV1 and equine paresis. Vet Rec 106, 277.

Novak, N. & Peng, W. M. (2005). Dancing with the enemy: the interplay of herpes

simplex virus with dendritic cells. Clin Exp Immunol 142, 405-410.

Nugent, J., Birch-Machin, I., Smith, K. C., Mumford, J. A., Swann, Z., Newton, J. R., Bowden, R. J., Allen, G. P. & Davis-Poynter, N. (2006). Analysis of equid

herpesvirus 1 strain variation reveals a point mutation of the DNA polymerase

strongly associated with neuropathogenic versus non-neuropathogenic disease

outbreaks. J Virol 80, 4047-4060.

Ostlund, E. N. (1993). The equine herpesviruses. Vet Clin North Am Equine Pract 9,

283-294.

Patel, J. R., Edington, N. & Mumford, J. A. (1982). Variation in cellular tropism

between isolates of equine herpesvirus-1 in foals. Arch Virol 74, 41-51.

Pollara, G., Kwan, A., Newton, P. J., Handley, M. E., Chain, B. M. & Katz, D. R. (2005). Dendritic cells in viral pathogenesis: protective or defective? Int J Exp

Pathol 86, 187-204.

Randolph, G. J., Ochando, J. & Partida-Sanchez, S. (2008). Migration of dendritic cell

subsets and their precursors. Annu Rev Immunol 26, 293-316.

Reed, L. J. & Muench, H. (1938). A simple method of estimating fifty per cent

endpoints. Am J Hyg 27, 493-497.

Scott, J. C., Dutta, S. K. & Myrup, A. C. (1983). In vivo harboring of equine

herpesvirus-1 in leukocyte populations and subpopulations and their quantitation

from experimentally infected ponies. Am J Vet Res 44, 1344-1348.

Siedek, E. M., Whelan, M., Edington, N. & Hamblin, A. (1999). Equine herpesvirus

type 1 infects dendritic cells in vitro: stimulation of T lymphocyte proliferation

and cytotoxicity by infected dendritic cells. Vet Immunol Immunopathol 67, 17-32.

Steinbach, F., Borchers, K., Ricciardi-Castagnoli, P., Ludwig, H., Stingl, G. & Elbe-Burger, A. (1998). Dendritic cells presenting equine herpesvirus-1 antigens

induce protective anti-viral immunity. J Gen Virol 79, 3005-3014.

Replication kinetics of EHV1

86

Stierstorfer, B., Eichhorn, W., Schmahl, W., Brandmuller, C., Kaaden, O. R. & Neubauer, A. (2002). Equine herpesvirus type 1 (EHV-1) myeloencephalopathy:

a case report. J Vet Med B Infect Dis Vet Public Health 49, 37-41.

Tew, J. G., Kosco, M. H., Burton, G. F. & Szakal, A. K. (1990). Follicular dendritic

cells as accessory cells. Immunol Rev 117, 185-211.

Tumas, D. B., Brassfield, A. L., Travenor, A. S., Hines, M. T., Davis, W. C. & McGuire, T. C. (1994). Monoclonal antibodies to the equine CD2 T-lymphocyte

marker, to a pan-granulocyte monocyte marker and to a unique pan-B lymphocyte

marker. Immunobiology 192, 48-64.

Van de Walle, G. R., Goupil, R., Wishon, C., Damiani, A., Perkins, G. A. & Osterrieder, N. (2009). A Single-Nucleotide Polymorphism in a Herpesvirus

DNA Polymerase Is Sufficient to Cause Lethal Neurological disease. J Infect Dis

200, 20-25.

van der Meulen, K., Caij, B., Pensaert, M. & Nauwynck, H. (2006). Absence of viral

envelope proteins in equine herpesvirus 1-infected blood mononuclear cells during

cell-associated viremia. Vet Microbiol 113, 265-273.

van der Meulen, K., Vercauteren, G., Nauwynck, H. & Pensaert, M. (2003a). A local

epidemic of equine herpesvirus 1-induced neurological disorders in Belgium.

Vlaams Diergen Tijds 72, 366-372.

van der Meulen, K. M., Gryspeerdt, A. C., Vandekerckhove, A. A., Garre, B. A. & Nauwynck, H. J. (2007). The protective properties of vaccination against equine

herpesvirus 1-induced viremia, abortion and nervous system disorders. Vlaams

Diergen Tijds 76, 186-194.

van der Meulen, K. M., Nauwynck, H. J., Buddaert, W. & Pensaert, M. B. (2000). Replication of equine herpesvirus type 1 in freshly isolated equine peripheral

blood mononuclear cells and changes in susceptibility following mitogen

stimulation. J Gen Virol 81, 21-25.

van der Meulen, K. M., Nauwynck, H. J. & Pensaert, M. B. (2001). Mitogen

stimulation favours replication of equine herpesvirus-1 in equine blood

mononuclear cells by inducing cell proliferation and formation of close

intercellular contacts. J Gen Virol 82, 1951-1957.

van der Meulen, K. M., Nauwynck, H. J. & Pensaert, M. B. (2003b). Absence of viral

antigens on the surface of equine herpesvirus-1-infected peripheral blood

mononuclear cells: a strategy to avoid complement-mediated lysis. J Gen Virol 84,

93-97.

van Maanen, C. (2002). Equine herpesvirus 1 and 4 infections: an update. Vet Q 24, 58-

78.

Expression of late proteins is hampered

87

CHAPTER IV

EXPRESSION OF LATE VIRAL PROTEINS IS

HAMPERED IN INFECTED NASAL MUCOSAL

LEUKOCYTES BUT NOT IN EPITHELIAL CELLS

DURING EARLY PATHOGENESIS OF EQUINE

HERPESVIRUS TYPE 1 (EHV1) INFECTION

Adapted from:

The Veterinary Journal (conditionally accepted, 2011)

Annick C. Gryspeerdt, A.P. Vandekerckhove, H. Bannazadeh Baghi, G.R. Van de Walle,

H.J. Nauwynck

Expression of late proteins is hampered

89

SUMMARY

Equine herpesvirus 1 (EHV1) starts its replication by infection in the epithelial cells of

the upper respiratory tract (URT), where after the virus invades the lamina propria of the

URT using single mononuclear cells, hereby reaching the bloodstream which allows

further spread of this virus throughout the body. In addition, EHV1 has developed various

strategies to escape from immune effector mechanisms. One of these strategies is to

interfere with antibody-dependent cell lysis by hampering proper viral protein expression

on the cell surface.

Because epithelial and mononuclear cells are crucial cell populations in the early

pathogenesis of EHV1, the present study aimed to evaluate the expression of the late viral

proteins gB, gC, gD and gM in these type of cells, using several experimental systems.

Firstly, epithelial-like rabbit kidney (RK13) cells and peripheral blood mononuclear cells

(PBMC) were infected with EHV1 in vitro. Secondly, the recently established ex vivo

system of equine nasal mucosal explants was used and thirdly, an in vivo evaluation was

performed by means of nasal mucosa tissues of experimentally EHV1-infected ponies.

Our salient findings were that the late proteins gB, gC, gD and gM were clearly expressed

in almost all late-infected epithelial cells, whereas the expression pattern was totally

different in infected mononuclear cells: gB and gM were expressed in 60 to 90% of these

cells, gC and gD in only 20%.

These results show that the expression of late viral proteins during the early

pathogenesis of EHV1 depends upon the cell type infected. Furthermore, this study

underlines the use of nasal mucosal explants as a valuable alternative to experimental in

vivo studies in the horse.

Expression of late proteins is hampered

90

1 INTRODUCTION

Equine herpesvirus 1 (EHV1), a member of the Alphaherpesvirinae, is an ubiquitous

equine respiratory pathogen, affecting horses worldwide (Allen & Bryans, 1986;

Brosnahan & Osterrieder, 2009). After an initial replication in the epithelial cells of the

upper respiratory tract (URT), EHV1 can spread through the basement membrane (BM)

carried by single infected mononuclear immune cells, hereby reaching the underlying

connective tissue (Vandekerckhove et al., 2010). Via a cell-associated viremia in

peripheral blood mononuclear cells (PBMC), EHV1 is then transported to its internal

target organs, the pregnant uterus and the central nervous system, where viral replication

in endothelial cells can cause abortion, neonatal foal death and nervous system disorders

through vascular damage (Smith & Borchers, 2001; Smith et al., 1996). As a result of the

short-lived immunity following an EHV1 infection, viral replication in the respiratory

tract can re-occur as soon as two months and viremia can be detected as early as six

months after primary infection (Edington & Bridges, 1990; Gibson et al., 1992). High

levels of circulating EHV1-specific antibodies are still present when re-infection with

EHV1 occurs, suggesting that EHV1 recognition by the immune system is seriously

hampered (Burrows et al., 1984; Gleeson & Coggins, 1980; Heldens et al., 2001;

Mumford et al., 1987).

The genome of EHV1 encodes at least 12 known glycoproteins, designated

glycoprotein B (gB), gC, gD, gE, gG, gH, gI, gK, gL, gM, gN and gp2. Herpesviral

glycoproteins are present in the viral envelope as distinct, projected spikes and because

they are involved in various steps of the virus infection cycle, such as attachment,

penetration, egress and cell to cell spread, they represent major determinants of

pathogenicity, virulence and host cell tropism (Spear & Longnecker, 2003). For example,

glycoprotein C and gM are non-essential glycoproteins, with gC being involved in the

first unstable binding of the virus to glycosaminoglycan moieties of cell surface heparin

sulphate and in the process of egress (Osterrieder, 1999; Sugahara et al., 1997), and gM

contributing to virus penetration and cell-to-cell spread of the virus (Osterrieder et al.,

1996). The essential protein gD interacts with a putative entry receptor to attain a stable

binding (Csellner et al., 2000; Frampton et al., 2005) and is furthermore also involved in

cell-to-cell spread of the virus (Csellner et al., 2000; Wellington et al., 1996a). The

essential protein gB is involved in the penetration of the virus into the host cell and cell-

Expression of late proteins is hampered

91

associated spread of the virus to uninfected neighbouring cells (Neubauer et al., 1997;

Wellington et al., 1996b).

In general, upon infection of a cell with an alphaherpesvirus, these viral envelope

proteins are synthesised and incorporated into various cellular membranes (Mettenleiter,

2002). The presence of viral proteins in the plasma membrane of infected cells renders

them susceptible to antibody (Ab)-dependent cell lysis, through binding of virus-specific

Abs on viral proteins. Subsequently, infected cells are destructed by phagocytic cells and

natural killer cells or via activation of the complement cascade (Hansch et al., 1997;

Hirsch, 1982; Janeway et al., 2001). Several herpesviruses have developed strategies to

interfere with immune effector mechanisms and EHV1 is no exception in this regard (van

der Meulen et al., 2006b). One strategy of EHV1 is to interfere with Ab-dependent cell

lysis by hampering envelope protein expression on the cell surface. An in vitro study by

van der Meulen et al. (2003b) demonstrated that approximately 70% of all EHV1-infected

PBMC did not express viral envelope proteins on their surface, despite a clear

intracellular expression (van der Meulen et al., 2003b). This absence of viral proteins on

the surface of these infected PBMC was shown to render them insensitive to Ab-

dependent complement-mediated lysis and Ab-dependent cell-mediated cytotoxicity (van

der Meulen et al., 2003b). Importantly, in an in vivo study of experimentally inoculated

ponies, 98% of the infected PBMC collected during cell-associated viremia did not show

viral envelope proteins on their plasma membrane, nor were they found intracellularly,

and only immediate early and at least some early proteins were present (van der Meulen

et al., 2006a). Based on these results, it was hypothesized that EHV1-infected PBMC are

at an early stage of infection during viremia and that further replication steps, including

proper expression of late proteins, only occurs upon adhesion to endothelial cells at the

target organs (van der Meulen et al., 2006b).

The main purpose of the present study was to evaluate the expression of several late

viral envelope proteins, including gB, gC, gD and gM in the different cell types which

become infected in the URT, a first step during the early pathogenesis. Two important

cell types during early pathogenesis were analysed in three different model systems: in

vitro with the standard epithelial RK13 cell line and PBMC, ex vivo by means of the

recently established equine nasal mucosal explant system and finally in vivo, with tissues

collected from experimentally infected ponies.

Expression of late proteins is hampered

92

2 MATERIALS AND METHODS

2.1 Virus

Two different strains were used in this study to take into account a possible influence

of the neurovirulent/non-neurovirulent nature of EHV1. On the one hand the Belgian

EHV1 strain 03P37 was used, isolated from the blood of a paralytic horse (Garré et al.,

2009; van der Meulen et al., 2003a), and on the other hand the Belgian strain 97P70,

isolated from an aborted fetus (van der Meulen et al., 2006a). These strains were typed in

their DNA polymerase in cooperation with the Animal Health Trust in the United

Kingdom (Nugent et al., 2006) and based on their origin and genotyping, these strains are

referred to in this paper as neurovirulent (D752) and non-neurovirulent (N752) respectively.

Virus stocks used for inoculation were at the 6th

passage; 2 passages in rabbit kidney cells

(RK13) and 4 subsequent passages in equine embryonic lung cells (EEL). Both viruses

have previously been shown to display virtually identical in vitro single-step growth

properties (Gryspeerdt et al., 2010).

2.2 Antibodies

For the detection of the late proteins gB, gC, gD and gM, specific antibodies were

used: mouse monoclonal antibodies (mAbs) 4B6 against gB (Osterrieder et al., 1995),

1B6 against gC (Huemer et al., 1995), 107D1D against gD (a kind gift from Merial,

France), and the monospecific anti-gM mouse antiserum F6 against gM (Osterrieder et

al., 1996).

To identify EHV1-infected cells, two antibodies were used: either rabbit polyclonal

antibodies against the immediate early (IE) protein (Caughman et al., 1988), or horse

polyclonal anti-EHV1 antibodies. The latter antibodies were obtained by

hyperimmunisation of a horse (van der Meulen et al., 2003b). The polyclonal antibodies

were purified on a protein G column and subsequently biotinylated (Amersham

International, Buckinghamshire, UK). To ensure that these antibodies could recognise the

late proteins of EHV1, transfection studies were performed. To this end, RK13 cells were

transfected with 1 µg of plasmid DNA expressing gB, gC, gD or gM (a kind gift from

Prof. Osterrieder, Freie Universitaet Berlin), using lipofectamin (Invitrogen). After 48h,

Expression of late proteins is hampered

93

RK13 cells were fixed and stained by double immunofluorescence stainings as described

below. All transfected cells, as identified by a positive staining for each late protein

respectively, were also positive for the anti-EHV1 polyclonal antibodies, indicating that

these EHV1-specific antibodies are able to recognise all late proteins evaluated in the

present study. In addition, kinetic infection studies in RK13 cells showed that these

polyclonal antibodies stain all infected cells from 24 hpi onwards when the late

glycoproteins are fully expressed, indicating that these antibodies are a useful tool to

detect cells in a late stage of infection. These horse polyclonal anti-EHV1 antibodies are

especially advantageous to evaluate infected cells in the nasal mucosa because these cells

are at different stages of infection and they always identify cells in the late stage of

infection. Rabbit polyclonal antibodies against the IE protein cannot discriminate between

cells early in infection from cells late in infection with an abortive infection.

2.3 Expression of late proteins

(i) In RK13 cells and PBMC.

Cells of the epithelial-like rabbit kidney (RK13) cell line were seeded on glass inserts

in 24-well culture plates (Nunc A/S) at a density of 300 000 cells/well and were further

cultured in growth medium (MEM supplemented with 100 U/ml penicillin, 0.1 mg/ml

streptomycin, 0.1 mg/ml kanamycin, 0.3 mg/ml glutamine and 5% foetal calf serum) at

37°C for 24 hours . Then, cultures were infected with either neurovirulent strain 03P37 or

non-neurovirulent strain 97P70 at a multiplicity of infection (MOI) of 10. After 1 hour,

virus was removed by rinsing the cells twice with culture medium and after 0, 4, 8, 13 and

24 subsequent hours of incubation, the cells were fixed with methanol for 20 minutes at -

20°C and stored at -70°C. Infected cells were stained as described below and were

analyzed by confocal laser scanning microscopy (Leica TCS SP2 laser scanning spectral

confocal system, Leica Microsystems GmbH, Wetzlar) and the Leica confocal software.

Hereby, individual cells were counted from their appearance (3 hpi) until the appearance

of plaques (8hpi). Newly infected individual cells present after 13 hpi were not taken into

account as they represent a second replication cycle.

Blood samples were collected from three different, healthy adult horses by jugular

venipuncture and collected on heparin (Leo). PBMC were isolated by density

centrifugation on Ficoll-Paque (Pharmacia Biotech AB), washed 3 times with PBS and

Expression of late proteins is hampered

94

resuspended in complete medium containing 2/1 MEM/RPMI 1640, supplemented with

10% fetal calf serum (Grainer), 0.05 mM 2-mercapto-ethanol (Gibco), 100 U/ml

penicillin (Continental Pharma), 0.1 mg/ml streptomycin (Certa), 0.1 mg/ml kanamycin

(Kela), 0.3 mg/ml glutamin (BDH Biochemical), 1% non-essential amino-acids 100x

(Gibco) and 1 mM sodium pyruvate (Gibco). After 24 hours of cultivation, PBMC were

inoculated at a MOI of 10 with either neurovirulent strain 03P37 or non-neurovirulent

strain 97P70. After 1 hour of incubation at 37 °C, cells were washed thoroughly with

RPMI and cultured in medium supplemented with 10 U/ml heparin (Leo). Cell smear

samples were made 48 hours post inoculation (pi) and fixed in acetone for 20 minutes.

(ii) In ex vivo nasal mucosal explants.

The cultivation and infection of nasal mucosal explants was performed exactly as

previously described (Vandekerckhove et al., 2010). In brief, immediately after slaughter,

the head was removed from the carcass and longitudinally sawn into 2 equal sections.

Tissue from the deep intranasal part of the septum of 3 different horses was collected. The

tissues were transported on ice in phosphate buffered saline (PBS), supplemented with 1

µg/ml gentamycin, 1 mg/ml streptomycin, 1000 U/ml penicillin, 1 mg/ml kanamycin and

5 µg/ml amphotericin, to the laboratory. Mucosal explants were stripped from the surface

of the different tissues by use of surgical blades (Swann-Morton). The stripped mucosa

was divided into equal explants of 25 mm2 and placed epithelium upwards on fine-

meshed gauze for culture at an air-liquid interface. Only a thin film of serum-free medium

DMEM (50%)/RPMI (50%) supplemented with 0.3 mg/ml glutamine (BHD

Biochemical), 1 µg/ml gentamycin (Gibco), 0.1 mg/ml streptomycin (Certa) and 100

U/ml penicillin (Continental Pharma)) covered the explants, thereby mimicking the

air/liquid interface found in the respiratory tract of the living animal. Explants were

maintained at 37°C in an atmosphere containing 5% CO2. Explants were inoculated after

24 h of culture by immersion of the explant in 1 ml of inoculum, containing 106.5

TCID50

of either EHV1 strain for 1 h at 37°C and 5% CO2. After incubation, explants were

washed twice with warm medium and transferred back to their gauze. At 48 hours post

inoculation (pi), explants were collected, embedded in methylcellulose medium

(Methocel® MC, Sigma-Aldrich, St.Louis) and samples were frozen at -70 °C.

Expression of late proteins is hampered

95

(iii) In nasal mucosa of experimentally in vivo infected ponies. Six EHV-negative

ponies (SN titer <2 and IPMA titer <10) were divided in two groups of three and

inoculated either with strain 97P70 or 03P37 as described previously (Gryspeerdt et al.,

2010). In brief, ponies were inoculated oronasally with 20 ml of a virus suspension

containing 106.5

tissue culture infectious dose 50 (TCID50). Ten ml of the virus suspension

was administered intranasally (5 ml per nostril) using a thin probe and 10 ml was

inoculated orally with a syringe. On two dpi, all ponies were euthanized with an overdose

of Natriumpentobarbital® (Kela, Hoogstraten, Belgium), and the intermedial part of the

nasal septum was collected. Tissue samples were immediately frozen with

methylcellulose medium (Methocel®MC, Sigma-Aldrich, St. Louis, MO, USA) at -70°C.

This experimental design was approved by the local ethical committee of the Faculty of

Veterinary Medicine, Ghent University, Belgium.

2.4 Double immunofluorescence stainings

To visualise the presence of gB, gC, gD and gM in infected cells, the specific

antibodies (mAbs) against these glycoproteins were used in a first step, followed by

incubation with secondary Texas Red®-labelled goat anti-mouse antibodies (Molecular

Probes, Eugene, OR, USA). For the identification of EHV1-infected cells, either the

rabbit polyclonal antibodies against the IE protein (RK13 cells), or the biotinylated horse

polyclonal anti-EHV1 antibodies (PBMC, nasal mucosa) were used. Subsequently,

samples were incubated with secondary FITC®-labelled goat anti-rabbit antibodies

(Molecular Probes, Eugene, OR, USA) for the polyclonal rabbit anti-IE antibody or

streptavidin-FITC® (Molecular Probes, Eugene, OR, USA) for the biotinylated polyclonal

anti-EHV1 antibody. Antibodies were incubated for 1 h at 37 °C and 5 % CO2 and

washed thoroughly with phosphate buffered saline (PBS) in between staining steps. As

negative controls, (i) stainings were performed on uninfected RK13, PBMC and nasal

mucosa and (ii) isotype specific control antibodies on infected cells and tissues were

included. All cryosections were analyzed using confocal microscopy (Leica TCS SP2

Laser scanning spectral confocal system, Leica Microsystems GmbH, Wetzlar, Germany).

Expression of late proteins is hampered

96

3 RESULTS

3.1 Late proteins gB, gC, gD and gM are properly expressed in in vitro infected

epithelial-like RK13 cells

In a first experiment, RK13 cells were used to analyse the expression kinetics of the

viral EHV1 glycoproteins gB, gC, gD and gM during an EHV1 infection. To this end, the

immediate early protein (IE) was used to identify EHV1-infected cells and double

immunofluorescent stainings were performed to detect the viral glycoproteins gB, gD, gC

and gM. EHV1-infected cells were observed as early as 1 hpi and their number increased

over time (data not shown). From 8 hpi, all late proteins tested started to be expressed in

individual EHV1-infected cells irrespective of the strains used, with gB and gM being

expressed in a larger percentage of the infected cells than gC and gD (Figure 1).

Expression of these late proteins in infected cells further increased over time and finally

at 24 hpi, the expression of gB, gC, gD and gM was found in all infected cells. This time

point co-incided with the time point when the polyclonal anti-EHV1 antibodies recognise

infected cells and therefore, this Ab was used in all further experimentation to identify

late infected cells.

Expression of late proteins is hampered

97

Figure 1. Kinetic study of the expression of late proteins gB, gC, gD and gM in EHV1-infected RK13

cells inoculated with neurovirulent strain 03P37 (A) and non-neurovirulent strain 97P70 (B). All data

represent means + SD of triplicate independent experiments. EHV1-infected cells were firstly incubated

with mouse monoclonal antibodies against gB, gC, or gD and a monospecific mouse antiserum against gM,

followed by Texas Red®-labelled goat anti-mouse antibodies. Secondly, they were incubated with a

polyclonal rabbit anti-IE antibody, followed by FITC®-labelled goat anti-rabbit antibodies.

3.2 Expression of the late proteins gC and gD is hampered in in vitro EHV1-infected

PBMC

Next, we wanted to evaluate the expression of the late proteins gB, gC, gD and gM in

a cell type which is also relevant during the pathogenesis of EHV1 infection, namely

peripheral blood mononuclear cells (PBMC). PBMC were isolated from the blood and

infected in vitro with the neurovirulent or non-neurovirulent EHV1 strain. At 48 hpi,

A

B

0

20

40

60

80

100

0 5 10 15 20 25

Hours post inoculation

Exp

ress

ion

of

late

pro

tein

sin

IE-p

osi

tiv

eR

K1

3 c

ells

(%)

gB

gC

gD

gM

0

20

40

60

80

100

0 5 10 15 20 25

Hours post inoculation

gB

gC

gD

gM

Exp

ress

ion

of

late

pro

tein

sin

IE-p

osi

tiv

eR

K1

3 c

ells

(%)

A

B

0

20

40

60

80

100

0 5 10 15 20 25

Hours post inoculation

Exp

ress

ion

of

late

pro

tein

sin

IE-p

osi

tiv

eR

K1

3 c

ells

(%)

gB

gC

gD

gM

0

20

40

60

80

100

0 5 10 15 20 25

Hours post inoculation

gB

gC

gD

gM

Exp

ress

ion

of

late

pro

tein

sin

IE-p

osi

tiv

eR

K1

3 c

ells

(%)

Expression of late proteins is hampered

98

infected cells were identified using the polyclonal anti-EHV1 Ab and expression of the

late proteins gB, gC, gD and gM was evaluated in these infected cells. For both strains,

expression of the late proteins gB and gM was almost 100% in infected cells (Figure 2A).

In contrast, expression of gC and gD was only seen in 15 to 25% of EHV1-infected

PBMC (Figure 2A). These results indicate that expression of certain late proteins which

are normally expressed in infected rabbit epithelial cells at 24 hpi, are impaired in their

expression in leukocytes at 48 hpi.

3.3 Expression of late proteins gC and gD is also hampered in EHV1-infected

leukocytes but not epithelial cells from nasal mucosa in explants and experimentally

infected ponies

To assess the relevance of the observations which were made with the epithelial-like

RK13 cells and PBMC, we repeated the double immunofluorescence stainings in

cryosections of (i) infected nasal mucosal explants and (ii) nasal tissue of experimentally

infected ponies. Previous work in our laboratory has shown that in both models, the nasal

epithelial cells become infected with EHV1. In addition, single EHV1-infected cells are

also observed, which are mainly present under the BM in connective tissue and which

were identified as leukocytes (Gryspeerdt et al., 2010; Vandekerckhove et al., 2010).

In nasal explants and in nasal tissues of in vivo inoculated horses, infected epithelial

cells showed a full expression of late proteins tested (Figure 3), which is in line with what

was observed in RK13 cells at 24 hpi. Individual infected leukocytes, located in the

connective tissue below the BM showed the same tendency as in vitro infected PBMC.

Hereby, around 80% to 90% of late infected cells showed expression of the late proteins

gB and gM in nasal explants and in vivo harvested nasal tissues, whereas the expression

of the late proteins gC and gD in these EHV1-infected leukocytes fluctuated around 10 to

15% in in vivo harvested nasal tissues and nasal mucosal explants respectively (Figure 2B

and 2C). No clear differences in expression levels were found in cells infected with either

the neurovirulent 03P37 or the non-neurovirulent 97P70 (Figure 2B and 2C).

Expression of late proteins is hampered

99

Figure 2. Expression of late proteins gB, gC, gD and gM in late infected leukocytes at 48 hours post

inoculation with neurovirulent strain 03P37 and non-neurovirulent strain 97P70 in three different

systems: (A) in vitro infected PBMC, (B) nasal mucosal explants and (C) nasal mucosa of experimentally

infected ponies. All data represent means + SD of triplicate independent experiments. Firstly, samples were

incubated with mouse monoclonal antibodies against gB, gC, gD or gM, followed by Texas Red®-labelled

goat anti-mouse antibodies. Secondly, they were incubated with the polyclonal horse anti-EHV1 antibody,

followed by streptavidin-FITC®.

A

B

C

0

10

20

30

40

50

60

70

80

90

100

gB gC gD gM

Pre

sence

of

late

pro

tein

sin

in

vit

ro

infe

cted

PB

MC

(%

)

neurovirulent

non-neurovirulent

neurovirulent

non-neurovirulent

0

10

20

30

40

50

60

70

80

90

100

gB gC gD gM

Pre

sence

of

late

pro

tein

sin

sin

gle

EH

V1

-infe

cte

d

cel

lsin

nas

alex

pla

nts

(%)

0

10

20

30

40

50

60

70

80

90

100

gB gC gD gM

Pre

sence

of

late

pro

tein

sin

sin

gle

EH

V1

-infe

cted

cell

sin

nas

alti

ssue

(%)

Expression of late proteins is hampered

100

Figure 3. Expression of late proteins gB, gC, gD and gM in late infected epithelial cells. Since results were

virtually identical for all three systems and both strains tested, only immunofluorescence pictures of plaques in

in vitro infected nasal mucosal tissue is shown. First, a staining with the biotinylated equine polyclonal anti-

EHV1 IgG, followed by streptavidin-FITC® was performed (A). Second, a staining with specific mouse

antibodies against gB, gC, gD or gM followed by secondary Texas Red®

-labelled goat anti-mouse antibodies

was performed (B).

A

B

A

B

Expression of late proteins is hampered

101

4 DISCUSSION

Equine herpesvirus 1 (EHV1) starts its pathogenesis in the upper respiratory tract

where primary replication occurs and results in upper respiratory tract disease. After

invasion through the basement membrane (BM) barrier, the virus enters blood vessels and

rapidly disseminates in the body via a cell-associated viremia. Recent research has shown

that EHV1 replicates in epithelial cells by means of plaque formation and in migrating

individual cells, allowing EHV1 to cross the BM. The latter cells were identified as cells

of the monocytic lineage, T-lymphocytes and a small amount of B-lymphocytes

(Gryspeerdt et al., 2010; Kydd et al., 1994; Vandekerckhove et al., 2010). Hence, two

main cell populations appear to play a crucial role in the early pathogenesis of EHV1,

namely epithelial cells on the one hand and leukocytes on the other hand.

In the present study, we studied the expression of the late envelope glycoproteins

(g)B, gC, gD and gM in rabbit and horse epithelial cells as well as equine leukocytes, in

three frequently used EHV1 infection models. Hereby, our salient findings were that in

epithelial cells, a complete replication cycle of EHV1 was seen, with expression of all

four tested glycoproteins, gB, gC, gD and gM in 100% of the infected cells. This,

however, was in striking contrast to EHV1-infected leukocytes. In this cell type,

expression of the late proteins gB and gM was observed in 60 to 90% of infected cells,

whereas expression of the late proteins gC and gD never exceeded 20% of EHV1-infected

cells. As these results were consistent in vitro as well as in vivo infected cells, we

concluded that this phenomenon is truly cell-type dependent and not just merely a result

of the experimental set-up used.

A decreased expression of late proteins in certain cell types has already been

described for another herpesvirus, the human cytomegalovirus (HCMV). For this human

herpesvirus, viral gene expression in placental trophoblasts was shown to be restricted to

immediate-early or early gene products during HCMV infection (Sinzger et al., 1993;

Toth et al., 1995). In another study, where a human trophoblast continuous cell line was

used, expression of immediate-early, early and late genes was only delayed when

compared to expression in a fibroblastic continuous cell line (Halwachs-Baumann et al.,

1998). Hence, these studies indicate the importance of the cell type used when evaluating

protein expression after viral infection. For EHV1, quantitative dot blot hybridization

experiments revealed that expression of the late genes gD and gK was significantly lower

Expression of late proteins is hampered

102

in placental trophoblast cells than in foetal epithelial lung tissue (Kimura et al., 2004).

This low level of late gene transcription was linked with a limited production of viral

progeny and the lack of immunoreactivity for viral antigen in trophoblasts infected with

EHV1. However, all these studies have studied EHV1 protein expression in the later

stages of pathogenesis at the secondary replication sites, and therefore, our study is the

first to describe these events of decreased late protein expression in certain cell types

during the early stages of pathogenesis at the upper respiratory tract. The underlying

mechanism responsible for this decreased expression of EHV1 late proteins in certain cell

types, however, remains unclear. These findings could be an explanation why no plaque

formation in the connective tissue of the upper respiratory tract could be found in

previous studies (Gryspeerdt et al., 2010; Vandekerckhove et al., 2010), as gD is an

essential glycoprotein for virus spread and was found to be present in only very low

amounts of individual infected PBMC in the present study.

When combining our present results with data from previously performed studies in

the authors’ laboratory (van der Meulen et al., 2006a; van der Meulen et al., 2003b), it

could be hypothesized that EHV1-infected leukocytes which show a hampered expression

of late proteins, are the cells reaching the blood stream and subsequently causing a cell-

associated viremia. This is supported by the results of van der Meulen et al. (2006),

stating that 98% of EHV1-infected PBMC present in the blood during cell-associated

viremia in experimentally inoculated ponies lacked expression of certain late viral

envelope proteins on their cell surface. They did, however, show expression of the

immediate early protein and at least one early protein, ICP22, indicating that these cells

were truly infected (van der Meulen et al., 2006a). So one could hypothesize that these

cells are in an early stage of infection and that some activation signals need to trigger the

expression of envelope proteins on the cell surface to free the way for transmission of

infectious virus at the secondary replication sites. Such hypothesis has already been put

forward a long time ago by Scott et al. (1983). They described that although virus could

be recovered after cocultivation of intact leukocytes with a permissive cell line, no

infectious virus could be found after disruption of these cells (Scott et al., 1983). All this

indicates that EHV1-infected leukocytes exist in a non-infective or subvirion form.

Indeed, an in vitro study showed that activation of adhesion molecules in endothelial cells

of the nasal mucosa, and the reproductive tract, is a key step in transferring virus from

infected leukocytes to endothelial cells (Smith et al., 2002; Smith et al., 2001).

Expression of late proteins is hampered

103

In conclusion, although this is only a basic study where cells in a later stage of

infection were studied, these results indicate that the expression of certain viral proteins

during the early pathogenesis of EHV1 depends upon the cell-type infected. Furthermore,

this study underlines again the use of nasal mucosal explants as a valuable alternative to

experimental in vivo studies in the horse.

ACKNOWLEDGEMENTS

This research was supported by the Institute for the promotion of Innovation through

Science and Technology in Flanders (IWT-Vlaanderen). We would like to thank Prof. Dr.

N. Osterrieder for kindly providing the plasmids used in transfection studies.

Expression of late proteins is hampered

104

References

Allen, G. P. & Bryans, J. T. (1986). Molecular epizootiology, pathogenesis, and

prophylaxis of equine herpesvirus-1 infections. Prog Vet Microbiol Immunol 2,

78-144.

Brosnahan, A. J. & Osterrieder, K. (2009). Equine herpesvirus-1: a review and update.

Equine Vet J in press.

Burrows, R., Goodridge, D. & Denyer, M. S. (1984). Trials of an inactivated equid

herpesvirus 1 vaccine: challenge with a subtype 1 virus. Vet Rec 114, 369-374.

Caughman, G. B., Robertson, A. T., Gray, W. L., Sullivan, D. C. & O'Callaghan, D. J. (1988). Characterization of equine herpesvirus type 1 immediate early proteins.

Virology 163, 563-571.

Csellner, H., Walker, C., Wellington, J. E., McLure, L. E., Love, D. N. & Whalley, J. M. (2000). EHV-1 glycoprotein D (EHV-1 gD) is required for virus entry and

cell-cell fusion, and an EHV-1 gD deletion mutant induces a protective immune

response in mice. Arch Virol 145, 2371-2385.

Edington, N. & Bridges, C. G. (1990). One way protection between equid herpesvirus 1

and 4 in vivo. Research in veterinary science 48, 235-239.

Frampton, A. R., Jr., Goins, W. F., Cohen, J. B., von Einem, J., Osterrieder, N., O'Callaghan, D. J. & Glorioso, J. C. (2005). Equine herpesvirus 1 utilizes a

novel herpesvirus entry receptor. J Virol 79, 3169-3173.

Garré, B., Gryspeerdt, A., Croubels, S., De Backer, P. & Nauwynck, H. (2009). Evaluation of orally administered valacyclovir in experimentally EHV1-infected

ponies. Vet Microbiol 135, 214-221.

Gibson, J. S., Slater, J. D., Awan, A. R. & Field, H. J. (1992). Pathogenesis of equine

herpesvirus-1 in specific pathogen-free foals: primary and secondary infections

and reactivation. Arch Virol 123, 351-366.

Gleeson, L. J. & Coggins, L. (1980). Response of pregnant mares to equine herpesvirus

1 (EHV1). Cornell Veterinarian 70, 391-400.

Gryspeerdt, A. C., Vandekerckhove, A. P., Garre, B., Barbe, F., Van de Walle, G. R. & Nauwynck, H. J. (2010). Differences in replication kinetics and cell tropism

between neurovirulent and non-neurovirulent EHV1 strains during the acute phase

of infection in horses. Vet Microbiol 142, 242-253.

Halwachs-Baumann, G., Wilders-Truschnig, M., Desoye, G., Hahn, T., Kiesel, L., Klingel, K., Rieger, P., Jahn, G. & Sinzger, C. (1998). Human trophoblast cells

are permissive to the complete replicative cycle of human cytomegalovirus. J

Virol 72, 7598-7602.

Hansch, G. M., Rother, K. O. & Till, G. O., (eds) (1997). The Complement System.

Berlin, Heidelberg: Springer-Verlag.

Heldens, J. G., Hannant, D., Cullinane, A. A., Prendergast, M. J., Mumford, J. A.,

Nelly, M., Kydd, J. H., Weststrate, M. W. & van den Hoven, R. (2001). Clinical and virological evaluation of the efficacy of an inactivated EHV1 and

EHV4 whole virus vaccine (Duvaxyn EHV1,4). Vaccination/challenge

experiments in foals and pregnant mares. Vaccine 19, 4307-4317.

Hirsch, R. L. (1982). The complement system: its importance in the host response to

viral infection. Microbiol Rev 46, 71-85.

Expression of late proteins is hampered

105

Huemer, H. P., Nowotny, N., Crabb, B. S., Meyer, H. & Hubert, P. H. (1995). gp13

(EHV-gC): a complement receptor induced by equine herpesviruses. Virus

Research 37, 113-126.

Janeway, C. A., Travers, P., Walport, M. & Shlomchik, M., (eds) (2001). The

humoral immune response. New York, USA: Garland Publishing.

Kimura, T., Hasebe, R., Mukaiya, R., Ochiai, K., Wada, R. & Umemura, T. (2004). Decreased expression of equine herpesvirus-1 early and late genes in the placenta

of naturally aborted equine fetuses. Journal of Comparative Pathology 130, 41-47.

Kydd, J. H., Smith, K. C., Hannant, D., Livesay, G. J. & Mumford, J. A. (1994). Distribution of equid herpesvirus-1 (EHV-1) in respiratory tract associated

lymphoid tissue: implications for cellular immunity. Equine Vet J 26, 470-473.

Mettenleiter, T. C. (2002). Herpesvirus assembly and egress. J Virol 76, 1537-1547.

Mumford, J. A., Rossdale, P. D., Jessett, D. M., Gann, S. J., Ousey, J. & Cook, R. F. (1987). Serological and virological investigations of an equid herpesvirus 1 (EHV-

1) abortion storm on a stud farm in 1985. Journal of Reproduction and Fertility,

supplement 35, 509-518.

Neubauer, A., Braun, B., Brandmuller, C., Kaaden, O. R. & Osterrieder, N. (1997). Analysis of the contributions of the equine herpesvirus 1 glycoprotein gB

homolog to virus entry and direct cell-to-cell spread. Virology 227, 281-294.

Nugent, J., Birch-Machin, I., Smith, K. C., Mumford, J. A., Swann, Z., Newton, J. R., Bowden, R. J., Allen, G. P. & Davis-Poynter, N. (2006). Analysis of equid

herpesvirus 1 strain variation reveals a point mutation of the DNA polymerase

strongly associated with neuropathogenic versus non-neuropathogenic disease

outbreaks. J Virol 80, 4047-4060.

Osterrieder, N. (1999). Construction and characterization of an equine herpesvirus 1

glycoprotein C negative mutant. Virus Research 59, 165-177.

Osterrieder, N., Neubauer, A., Brandmuller, C., Braun, B., Kaaden, O. R. & Baines, J. D. (1996). The equine herpesvirus 1 glycoprotein gp21/22a, the herpes simplex

virus type 1 gM homolog, is involved in virus penetration and cell-to-cell spread

of virions. J Virol 70, 4110-4115.

Osterrieder, N., Wagner, R., Brandmuller, C., Schmidt, P., Wolf, H. & Kaaden, O. R. (1995). Protection against EHV-1 challenge infection in the murine model after

vaccination with various formulations of recombinant glycoprotein gp14 (gB).

Virology 208, 500-510.

Scott, J. C., Dutta, S. K. & Myrup, A. C. (1983). In vivo harboring of equine

herpesvirus-1 in leukocyte populations and subpopulations and their quantitation

from experimentally infected ponies. American Journal of Veterinary Research

44, 1344-1348.

Sinzger, C., Muntefering, H., Loning, T., Stoss, H., Plachter, B. & Jahn, G. (1993). Cell types infected in human cytomegalovirus placentitis identified by

immunohistochemical double staining. Virchows Archiv A, Pathological Anatomy

and Histopathology 423, 249-256.

Smith, D. J., Hamblin, A. & Edington, N. (2002). Equid herpesvirus 1 infection of

endothelial cells requires activation of putative adhesion molecules: an in vitro

model. Clin Exp Immunol 129, 281-287.

Smith, D. J., Hamblin, A. S. & Edington, N. (2001). Infection of endothelial cells with

equine herpesvirus-1 (EHV-1) occurs where there is activation of putative

adhesion molecules: a mechanism for transfer of virus. Equine Vet J 33, 138-142.

Expression of late proteins is hampered

106

Smith, K. C. & Borchers, K. (2001). A study of the pathogenesis of equid herpesvirus-1

(EHV-1) abortion by DNA in-situ hybridization. Journal of Comparative

Pathology 125, 304-310.

Smith, K. C., Mumford, J. A. & Lakhani, K. (1996). A comparison of equid

herpesvirus-1 (EHV-1) vascular lesions in the early versus late pregnant equine

uterus. Journal of Comparative Pathology 114, 231-247.

Spear, P. G. & Longnecker, R. (2003). Herpesvirus entry: an update. J Virol 77, 10179-

10185.

Sugahara, Y., Matsumura, T., Kono, Y., Honda, E., Kida, H. & Okazaki, K. (1997). Adaptation of equine herpesvirus 1 to unnatural host led to mutation of the gC

resulting in increased susceptibility of the virus to heparin. Arch Virol 142.

Toth, F. D., Mosborg-Petersen, P., Kiss, J., Aboagye-Mathiesen, Hager, G., Juhl, C.

B., Gergely, L., Zdravkovic, M., Aranyosi, J., Lampé, L. & Ebbesen, P. (1995). Interactions between human immunodeficiency virus type 1 and human

cytomegalovirus in human term syncytiotrophoblastcells coinfected with both

viruses. J Virol 69, 2223-2232.

van der Meulen, K., Caij, B., Pensaert, M. & Nauwynck, H. (2006a). Absence of viral

envelope proteins in equine herpesvirus 1-infected blood mononuclear cells during

cell-associated viremia. Vet Microbiol 113, 265-273.

van der Meulen, K., Vercauteren, G., Nauwynck, H. & Pensaert, M. (2003a). A local

epidemic of equine herpesvirus 1-induced neurological disorders in Belgium.

Vlaams Diergenskund Tijds 72, 366-372.

van der Meulen, K. M., Favoreel, H. W., Pensaert, M. B. & Nauwynck, H. J. (2006b). Immune escape of equine herpesvirus 1 and other herpesviruses of

veterinary importance. Vet Immunology and Immunopathology 111, 31-40.

van der Meulen, K. M., Nauwynck, H. J. & Pensaert, M. B. (2003b). Absence of viral

antigens on the surface of equine herpesvirus-1-infected peripheral blood

mononuclear cells: a strategy to avoid complement-mediated lysis. J Gen Virol 84,

93-97.

Vandekerckhove, A. P., Glorieux, S., Gryspeerdt, A. C., Steukers, L., Duchateau, L., Osterrieder, N., Van de Walle, G. R. & Nauwynck, H. J. (2010). Replication

kinetics of neurovirulent versus non-neurovirulent equine herpesvirus type 1

strains in equine nasal mucosal explants. J Gen Virol 91, 2019-2028.

Wellington, J. E., Lawrence, G. L., Love, D. N. & Whalley, J. M. (1996a). Expression

and characterization of equine herpesvirus 1 glycoprotein D in mammalian cell

lines. Arch Virol 141, 1785-1793.

Wellington, J. E., Love, D. N. & Whalley, J. M. (1996b). Evidence for involvement of

equine herpesvirus 1 glycoprotein B in cell-cell fusion. Arch Virol 141, 167-175.

Characteristics of a natural occurring outbreak of EHV1

107

CHAPTER V

DESCRIPTION OF AN UNUSUALLY LARGE

OUTBREAK OF NERVOUS SYSTEM DISORDERS

CAUSED BY EQUINE HERPESVIRUS 1 (EHV1) IN

2009 IN BELGIUM

Adapted from:

Vlaams Diergeneeskundig Tijdschrift 80 (2011), 139-145

Annick C. Gryspeerdt, A.P. Vandekerckhove, J. Van Doorsselaere, G.R. Van de

Walle, H.J. Nauwynck

Characteristics of a natural occurring outbreak of EHV1

109

SUMMARY

Neurological disease in horses caused by neurovirulent strains of EHV1 has been reported

with increasing frequency during the last decade. Therefore, concern has been voiced

within the US horse industry that the neurologic form of EHV1 may be intensifying in

prevalence and/or morbidity and mortality. Full protection against these severe signs

cannot be obtained by vaccination with the currently available vaccines and effective

antiviral therapies are not available. In Belgium, outbreaks of EHV1-induced abortions

are a yearly recurrent phenomenon, but outbreaks of equine herpes myelitis (EHM) are

extremely rare. This report describes an unusually large outbreak of EHV1-induced

neurological disease in Antwerp, Belgium, which involved at least 13 different premises.

Seven of them were characterized in more detail and were included in this research. A

total of 212 horses were exposed to EHV1 and a morbidity of 26% was seen. The

incidence of neurological disorders in the affected horses was 43%. Furthermore, the

outbreak was characterized by the rapid occurrence of ataxia and paralysis immediately

after disappearance of fever. EHV1 was diagnosed as the etiological agent by means of

virus isolation and/or the detection of seroconversion. Isolated virus was classified as

neurovirulent EHV1, with all isolates belonging to group 4 upon sequencing in the

ORF30 and ORF68 region. The latter confirms the suspicion of the outbreak spreading

from the first premise. The extent of this current outbreak and the high percentage of

horses with neurological signs, along with the fact that EHM is only sporadically seen in

Flanders, might indicate that the neurological form of EHV1 is possibly also emerging in

Belgium.

Characteristics of a natural occurring outbreak of EHV1

110

1 INTRODUCTION

Equine herpesvirus 1 (EHV1), a member of the Alphaherpesvirinae, is endemic in the

horse population worldwide. After exposure, virus replicates in tissues of the upper

respiratory tract. Primary replication is followed by a leukocyte-associated viremia which

enables EHV1 to reach internal organs (Gryspeerdt et al., 2010; Kydd et al., 1994a; b). A

second replication in endothelial cells at the site of the pregnant uterus or the nervous

system may result in abortion and neonatal foal death or nervous system disorders

respectively (Allen & Bryans, 1986; Gilkerson & Barrett, 2008). The latter, also

designated equine herpes myelitis (EHM), is a severe condition which is still poorly

understood despite the correlation between a single amino acid variation in the ORF30

region of the viral DNA polymerase and neurological disease (Lunn et al., 2009; Nugent

et al., 2006; Van de Walle et al., 2009).

EHV1 is a ubiquitous pathogen with a worldwide distribution and yearly problems

with respiratory disorders and abortion are common. In contrast, outbreaks of nervous

system disorders caused by infection with EHV1 seem to be more rare. However, during

the last decade, large outbreaks of EHM have been reported in the USA with enhancing

frequency and the fatal outcomes after substantial suffering appeared to increase (Lunn et

al., 2009). In Belgium, EHV1 is also endemic in the horse population and EHV1-induced

abortion is a yearly recurrent event. However, EHM is rare and mostly involves a limited

number of horses on a single premise (van der Meulen et al., 2000). To date, only one

description of EHV1-induced neurological disorders has been reported in Belgium. No

extensive spread of EHM was seen during this episode and severe disease was limited to

the affected premise. The signs in neurologically affected horses consisted mainly of

ataxia and paralysis, but cerebral dysfunctions were also noticed (van der Meulen et al.,

2003).

The current report describes a large outbreak of EHV1-induced neurological disease

in Flanders in 2009, that started on a riding school and spread to at least 12 premises that

had been in direct contact with the riding school. The extent of this outbreak, along with

the higher percentage of horses with neurological signs, without the occurrence of

cerebral dysfunctions, and the rapid occurrence of severe signs raises the intriguing

question if EHM is also emerging in Belgium.

Characteristics of a natural occurring outbreak of EHV1

111

2 MATERIALS AND METHODS

2.1 Description of the premises

During an outbreak of EHV1-induced nervous system disorders in Antwerp, Flanders

(June 2009), fever and neurological disease were observed on at least 13 premises. All

these premises had attended a jumping contest on May 30th

, 2009, at a riding school

where the first cases of neurological signs occurred (premise n° 1). Although 13 premises

reported neurological disease, only the data of 7 premises were included in this report. On

these premises, EHV1 infection was confirmed by virus isolation and/or seroconversion

(Table II). The other 6 affected premises had several horses with neurological problems,

but refused to give any further information and were therefore not included in this

research (Figure 1). Information about vaccination state, management and housing can be

found in Figure 1 and Table I. Each premise was visited for evaluation of clinical signs

and local veterinarians were asked to collect blood samples from horses suspected of

infection with EHV1 for both virus isolation from leukocytes and detection of

seroconversion in paired serum samples.

2.2 Serological examination

The presence of EHV-specific antibodies was determined with a complement-

dependent seroneutralization (SN)-test in acute serum samples and convalescent serum

samples three weeks later. Two-fold dilution series of the sera were prepared in MEM.

Fifty µl of these serial dilutions were incubated for 23 hours (h) at 37°C with a fixed

number of infectious virus (300 TCID50 of EHV1 strain Arabica in 50 µl). Hereafter, 25

µl of guinea pig complement was added. After 1 h of incubation, the mixture of serum,

virus and complement was added to RK13 cells. Inoculated cultures were further

incubated at 37°C in an atmosphere containing 5% CO2. After 7 days of incubation, the

cultures were analyzed for the presence of cytopathic effect. The neutralization titre was

calculated as the reciprocal of the highest dilution of serum that was able to completely

block EHV1 infection in RK13 cells. Seroconversion was present when the antibody titre

of the convalescent sample was at least four times higher than in the acute sample.

Characteristics of a natural occurring outbreak of EHV1

112

2.3 Virological examination

Virus isolation from peripheral blood mononuclear cells (PBMC) was performed on

RK13 cells. Unclotted blood samples (EDTA) were taken from horses with clinical signs

(fever and/or neurological disorders). PBMC were isolated by means of density

centrifugation on Ficoll-Paque (Pharmacia Biotech AB, Uppsala, Sweden) and

subsequently inoculated on monolayers of RK13 cells. Monolayers were incubated at

37°C in an atmosphere containing 5% CO2 and examined daily for 7 consecutive days for

the presence of cytopathic effects.

2.4 PCR amplification and sequencing of ORF30 and ORF68

DNA was extracted from EHV1 isolates using a QiaAmp DNA extraction kit (Qiagen

Benelux, Venlo, The Netherlands). Amplification of ORF30 was performed using the

primers ORF30fw (gctacttctgaaaacggaggc) and ORF30rev (ctatcctcagacacggcaaca) and

amplification of ORF68 with the primers ORF68atg1 (atgggtgtggtcttaattac) or ORF68fw

(aagcattgccaaacagttcc) in combination with ORF68end1 (aacgtgtggatgtccggcc). PCR

products were treated with Exonuclease I and Antarctic Phosphatase (New England

Biolabs, Ipswich, USA) and used directly for cycle sequencing with a Big Dye

Terminator Cycle sequencing kit V1.1

(Applied Biosystems, Foster City, USA).

Additional internal oligonucleotides were used for sequencing of ORF68. Cycle

sequencing reaction products were purified using ethanol precipitation and separated on

an ABI Genetic Analyzer 310 (Applied Biosystems, Foster City, USA). The sequences

were analyzed and compiled using Align, ClustalW and Sixframe in the workbench

(workbench.sdsc.edu) and BlastN and BlastP at www.ncbi.nlm.nih.gov. To validate the

sequencing, previously characterized strains 97P70 (Genbank accession number:

GU271940), 03P37 (# GU271941) and Ab4 (# AY665713) were included (Garré et al.,

2007; Nugent et al., 2006).

Characteristics of a natural occurring outbreak of EHV1

113

2.5 Treatment

All affected animals received supportive therapy consisting of anti-inflammatory

drugs and antibiotics. Horses with severe ataxia and/or bladder paralysis were hung in

slings and the bladder was emptied manually twice a day. Some animals received an

antiviral therapy, consisting of 40 mg of valacyclovir/kg body weight, administered 3

times daily for different consecutive days (Garré et al., 2009). The doses were prepared

by mixing the calculated amount of valacyclovir with water and administered orally.

Because of cost restraints and the limited amount of valacyclovir that was available, only

four affected horses were treated for a different number of consecutive days.

3 RESULTS

3.1 Description of the number, age and sex of affected animals during the outbreak

On the first premise, a morbidity (affected horses/total n° of horses present) of 21%

was seen during the outbreak and this premise reported to vaccinate twice a year with two

different inactivated vaccines (Figure 1). The animals on premise n° 4 had a variable

vaccination status. The morbidity was only 9%. However, clear clinical disease was only

seen in non-vaccinated animals and not all animals were checked for signs of EHV1. All

other premises did not vaccinate against EHV1 and morbidity varied from 25% (premise

n° 5) up to 100% (premise n° 2, 3, 6 and 7). Overall, a morbidity of around 23% was

observed during this outbreak.

The age of the affected animals varied from 3 to 21 years. No correlation between the

severity of clinical signs and age could be noticed during the course of this outbreak.

In total, 43% of the affected horses developed nervous system disorders. Of all

infected females 55% (18/33) developed nervous system disorders. Only 24% (5/21) of

all infected males developed nervous system disorders (Table I). Of the affected horses

with neurological signs 30% (7/23) were eventually euthanized and these were mainly

female (6/7 or 86%) (Table I).

Characteristics of a natural occurring outbreak of EHV1

114

Figure 1. Schematic overview of the course of the outbreak of EHV1-induced neurological disorders

at the different premises. NSD: nervous system disorders; open management: public riding school with

incoming and outgoing horses; half-open management: private premise, but where horses attend

competitions at other premises; housing with direct contact: stables separated from each other by bars;

housing with indirect contact: stables separated from each other by full walls.

Table I. Description of the animals on the premises with EHV1 associated neurological disorders

Total n° of horses present

Total n° of affected horses

N° of horses with neurological

disorders

Premise

Male Female Total Male Female Total (%) Male Female Total (%)

1 53 43 96 8 12 20 (21%) 2 6 8 (40%)

2 1 2 3 1 2 3 (100%) 0 1 1 (33%)

3 2 10 12 0 3 3 (25%) 0 3 3 (100%)

4 42 38 80 2 5 7 (9%) 0 1 1 (14%)

5 3 4 7 3 4 7 (100%) 1 4 5 (71%)

6 2 1 3 2 1 3 (100%) 1 1 2 (67%)

7 5 6 11 5 6 11 (100%) 1 2 3 (27%)

108 104 212 21 33 54 (26%) 5 18 23 (43%)

Premise 1

First case: May 14th

Last case: June 16th

N°of horses present: 96

% morbidity: 21

% of horses with NSD: 40

Management: open

Housing: direct contact

Vaccination state: 2x/year

Premise 2

First case: June 5th

Last case: June 20th

N°of horses present: 3

% morbidity: 100

% of horses with NSD: 33

Management: half open

Housing: indirect contact

Vaccination state: none

Premise 3

First case: June 9th

Last case: June 10th

N°of horses present: 12

% morbidity: 25

% of horses with NSD: 100

Management: half open

Housing: direct, indirect contact

Vaccination state: none

Premise 4

First case: June 9th

Last case: June 19th

N°of horses present: 80

% morbidity: 9

% of horses with NSD: 14

Management: open

Housing: indirect contact

Vaccination state: variable

Premise 5

First case: June 9th

Last case: June 14th

N°of horses present: 7

% morbidity: 100

% of horses with NSD: 71

Management: half open

Housing: indirect contact

Vaccination state: none

Premise 6

First case: June 15th

Last case: June 28th

N°of horses present: 3

% morbidity: 100

% of horses with NSD: 67

Management: half open

Housing: direct, indirect contact

Vaccination state: none

Premise 7

First case: June 17th

Last case: June 17th

N°of horses present: 11

% morbidity: 100

% of horses with NSD: 27

Management: half open

Housing: direct, indirect contact

Vaccination state: none

6 additional premises

with reports of

neurological disorders,

but without any

addtional information,

nor confirmation of

EHV1

Characteristics of a natural occurring outbreak of EHV1

115

3.2 Methodical description of clinical symptoms during the outbreak

The most prominent clinical sign present, was fever (>39.5°C) that lasted for 2-5 days

and swelling of lymph nodes. During pyrexia, most of the horses showed a temporary

weakness of the tail. In the majority of the affected horses, a slight serous nasal discharge

was noted. On premise n° 1 icterus and oedema of the hind legs were also reported. These

symptoms were not regularly observed in the six other affected premises, as only three of

them reported oedema of the hind legs in a few horses. In two horses, urticaria were seen

during the pyretic period and two other horses suffered from muscle trembling after

disappearance of fever. During the outbreak, 43% of the affected horses showed nervous

system disorders (Table I). This occurred in most (65%) cases immediately after

disappearance of pyrexia, but some horses (35%) developed ataxia and paralysis without

preceding elevated body temperatures. Since body temperatures were not checked

regularly in the beginning of the outbreak, a period of fever could have passed unnoticed.

During this outbreak, moderate as well as severe ataxia were noticed. Severe ataxia was

accompanied by bladder paralysis in approximately 50% of the cases. Several horses

became quadriplegic shortly after developing ataxia, but quadriplegia also occurred

without previous neurological symptoms. In total, seven horses were euthanized during

this outbreak. Six of these horses (from premise n° 1) were presented with paralysis

before an etiological diagnosis was made and were euthanized within 24 hours after the

onset of neurological symptoms. The last horse (premise n° 4) was euthanized two weeks

after the occurrence of paralysis since no improvement of the situation was seen. All other

horses with neurological symptoms that survived were clinically normal 4 months later

and regained normal performance levels, with the exception of one mare that is again

suffering from urinary incontinence after a clinically normal period of four months.

3.3 Evaluation of antiviral treatment during outbreak

During the outbreak, four horses were treated orally with the antiviral drug

valacyclovir (Table II). Horse A was a gelding of 4 years of age (premise n° 1). He was

treated when the fever ceased after 5 days and the horse showed muscle tremors. As the

muscle tremors stopped after one day and no other symptoms of EHV1 infection were

observed, the therapy was stopped after 2 days. Horse B was a mare of 7 years (premise

Characteristics of a natural occurring outbreak of EHV1

116

n° 2), and the valacyclovir treatment started at the moment she became quadriplegic. This

horse was unable to stand and was hung up in a sling. Two days later, she was not yet

able to stand up on her own, but she could stand without the help of the sling and her

condition improved rapidly. Treatment was stopped after 5 days. One week later, she was

still slightly ataxic but was able to stand up and regained bladder control. Treatment of

horse C (female, 5 years, premise n° 2) was started when she had difficulties standing up

and was found severely ataxic. She was treated for 3 consecutive days. One week later,

she was still obviously ataxic but this slowly improved over the next couple of weeks.

Bladder paralysis lasted for almost three weeks. Finally, horse D (female, 7 years,

premise n° 3) was treated after she became quadriplegic for more than 36 hours. She was

lifted up in a sling and could barely stand on her own. The first two days after the

treatment was initiated, the horse showed a significant improvement. She was able to

stand on her own in the sling and even tried to walk. After three days, however, treatment

was stopped due to a lack of available valacyclovir. The horse remained stable for several

days, but no further improvement of clinical symptoms was observed. Two weeks after

the onset of symptoms, the horse was able to stand in the sling and started to regain

bladder control, but was not able to lay in sternal position or to stand up on her own.

Because this caused severe anxiety for the horse, she was euthanized.

Table II. Treatment of different animals with valacyclovir

Horse Gender Age Symptoms Duration of treatment Improvement

A ♂ 4 tremor 2 days +++

B ♀ 7 quadriplegia 5 days ++

C ♀ 5 severe ataxia 3 days +

D ♀ 7 quadriplegia 3 days ± †

†: euthanasia

3.4 Diagnosis

Blood samples were collected from seven horses with high fever, six horses with

ataxia and two horses with quadriplegia at the moment of sampling (Table III). EHV1

was isolated by means of PBMC co-cultivation from six of the seven horses with high

fever (86%), from four of the six horses with ataxia (67%) and from the two horses with

Characteristics of a natural occurring outbreak of EHV1

117

quadriplegia (100%) (Table III). Seroconversion of EHV-specific antibodies was

evaluated for thirteen of the fifteen horses, both at the time of clinical signs (acute

sample) as well as three weeks later (convalescent sample). Twelve horses seroconverted

for EHV1 (Table III).

All EHV1 isolates were typed as neurovirulent by sequencing of the D752 allele in the

ORF30 region (e.g. 09P240, # GU271939). In addition, all viruses were sequenced in

their ORF68 region, a genetic marker for grouping isolates into six major geographically

restricted groups (Nugent et al., 2006). All isolates in this outbreak of neurological

disease belonged to group 4. These genetic analyses, along with a direct link between the

confirmed premises and premise n° 1, further confirmed the assumption that all EHV1

infections on the different premises were caused by the same virus spread from premise

n° 1.

Table III. Results from different diagnostic tools to confirm EHV1 infection in discussed horses

Antibody titre in serum Premise Horse Signs

at the

moment of

sampling

Isolation of

EHV1 from

PBMC Acute sample Convalescent

sample

1 1 Fever + 128 1536

2 quadriplegia + 64 NA

2 1 Fever + 48 512

2 Fever + 16 256

3 1 Ataxia + 192 512

2 Ataxia + 96 1024

3 Ataxia - 16 256

4 1 quadriplegia + 96 NA

5 1 Fever + 4 1536

2 Fever + 12 768

3 Fever - 8 1024

4 Fever + 12 2048

6 1 Ataxia + 32 1536

7 1 Ataxia + 4 1536

2 Ataxia - 8 1536

NA: not available due to euthanasia

Characteristics of a natural occurring outbreak of EHV1

118

4 DISCUSSION

The present paper reports a large outbreak of EHV1-induced neurological disease in

Antwerp, Flanders in June 2009. How the present epidemic started on this premise is not

known, however two weeks before the first horse became recumbent and had to be

euthanized, a 5-year old mare was presented with fever, lasting for 4 days. It might be

hypothesized that this mare either reactivated latent EHV1 or got infected with EHV1 due

to many contacts of visiting horses on the premise. Secondary outbreaks on other

premises then occurred as a consequence of direct contact with infected horses from the

first premise. Both the first premise as well as secondary infected premises were put

under quarantine for three weeks and all horses without clinical symptoms in the

neighbourhood were vaccinated. The outbreak was successfully controlled and one month

after the onset of the outbreak, no more cases of EHV1 infection were reported.

An interesting feature of the present outbreak was the rapid occurrence of ataxia and

paralysis immediately after disappearance of fever. Previous reports mention an

incubation period of 7 to 10 days for the development of nervous system disorders after

experimental inoculation (Edington et al., 1986; Jackson et al., 1977). Interestingly,

although sex and age did not seem to influence the incidence of becoming infected, mares

appeared to be predisposed to develop severe ataxia and paralysis. Indeed, 55% of the

infected mares developed nervous system disorders (18/33), whereas this was only 24%

(5/21) for the infected male horses. The higher sensitivity of mares to develop severe

symptoms after EHV1 infection has already been described, however, no explanation for

this exists to date (Goehring et al., 2006; McCartan et al., 1995).

Although the efficacy of current commercial vaccines to protect horses against EHV1-

related symptoms is highly variable and none of them can guarantee 100% protection,

several vaccines have been shown to induce significant protection against either abortion

or nervous system disorders under experimental and field conditions (Goodman et al.,

2006; van der Meulen et al., 2007). The premises which were mainly affected during this

outbreak had an inadequate or no vaccination protocol at all. This observation has also

been reported during other outbreaks of EHV1-induced neurological disease, where most

outbreaks occurred on premises with unvaccinated horses (Goehring et al., 2006; van der

Meulen et al., 2003). During this outbreak, morbidity rates were lower (7 to 21%) in

vaccinated premises (n° 1 and 4) compared to the non-vaccinated premises (25 to 100%)

Characteristics of a natural occurring outbreak of EHV1

119

(Figure 1). On premise n° 4, where both vaccinated and unvaccinated horses were

present, fever and neurological disease was only reported in their unvaccinated horses

whereas the vaccinated horses showed no clinical signs of EHV1 infection. This could

indicate that vaccinated horses have a clinical protection against severe symptoms of

EHV1. Whether this protection is the result of a reduced spread of the virus or due to a

virological effect is not clear. Interestingly, although lower morbidity rates were observed

at the vaccinated premises n° 1 and 4, no difference could be seen in the number of horses

that eventually developed nervous system disorders compared to the non-vaccinated

premises (Table I). In Belgium, only inactivated vaccines are registered for use in horses

which mainly stimulate humoral immunity, whereas it is suggested that cell-mediated

immunity is of paramount importance to limit EHV1 infection (Kydd et al., 2006; Kydd

et al., 2003).

During this outbreak, four horses on three different premises were treated orally with

the antiviral drug valacyclovir. Experimental in vivo studies to evaluate the efficacy of

valacyclovir to reduce viral shedding and provide clinical recovery have recently been

performed, but with contradictory results (Garré et al., 2009; Maxwell et al., 2008).

During this outbreak, 8 horses showed paralysis. Two out of three treated horses (66%)

regained normal prestations, while five out of five (100%) untreated horses had to be

euthanized. Although the treated horses seemed to recover quite fast during treatment

compared to untreated horses with the similar clinical features, it is difficult to evaluate

the benefit of valacyclovir treatment during an outbreak, due to the limited amount of

treated horses versus non-treated horses and the variable treatment periods. Still,

extensive supportive care remains indispensable during an outbreak of EHV1-induced

neurological disorders to increase the likelyhood of recovery.

Both virus isolation from peripheral blood mononuclear cells (PBMC) as well as

seroconversion were used to diagnose EHV1 during this outbreak. EHV1 was

successfully isolated from PBMC during the present outbreak. This is in contrast to

previous published studies, mentioning that virus isolation from PBMC in outbreak

situations can be problematic (Lunn et al., 2009; van Maanen et al., 2001). In addition,

seroconversion has also been reported to have limitations in confirming a diagnosis of

EHV1, especially in horses with neurological signs (Lunn et al., 2009). Interestingly,

during the current outbreak all horses with neurological disorders seroconverted, with the

exception of one horse. An explanation for the successful diagnosis during this outbreak

Characteristics of a natural occurring outbreak of EHV1

120

could be the rapid occurrence of severe signs during the current outbreak which increased

the chance of detecting infectious virus and seroconversion. Low initial EHV1 antibodies

in a largely unvaccinated population could also have contributed to the easy detection of

seroconversion.

Although some interesting findings concerning risk factors, vaccination, treatment and

diagnosis were observed during this outbreak, conclusions drawn from natural occurring

outbreaks should be taken with care, owing to the small numbers involved and variability

in both vaccination and anti-viral treatment.

During the only other reported outbreak of EHV1-induced neurological disease in

Belgium six years ago, a morbidity of 42% was seen with 15% of the horses showing

neurological disorders. Ataxia and paralysis were observed, but also cerebral disorders

such as blindness, torticollis and severe apathy were seen (van der Meulen et al., 2003).

Cerebral disorders were not observed during this recent outbreak, but interestingly, this

outbreak was characterized by the rapid onset of ataxia and paralysis immediately after

the disappearance of fever. In addition, the incidence of neurological signs in the EHV1-

affected horses was much higher in the current outbreak (43%) compared to the outbreak

in 2003 (15%) (van der Meulen et al., 2003). These observations might possibly point

towards a change in neurovirulent potential of naturally circulating EHV1 strains.

In summary, concern has been voiced within the U.S. horse industry that the

neurologic form of EHV1 might be emerging, based on an increased number of EHM

outbreaks with a larger number of seriously affected horses throughout the United States

over the last decade (USDA, 2007). Until now, no such evolution was seen in Belgium

and no reports of EHM outbreaks have been reported with the exception of one limited

outbreak in 2003. The present report describes a recent large outbreak of EHV1 with a

rapid spread and a remarkably high number of animals suffering of EHM. Together with

the fact that also in 2010 two small outbreaks were reported, the question raises whether

the neurological form of EHV1 is also emerging in Belgium and hence Europe. However,

it is not clear if these observations are related to an evolving disease with a changed

virulence and behaviour or if they are the consequence of a greater awareness and the

development of better diagnostic methods (USDA, 2007). Many data gaps exist, and more

investigations need to be conducted to better understand this evolving situation and to

identify other possible factors playing a role. Therefore, a good surveillance system in

Belgium and Europe would be of great importance to monitor outbreaks of EHM, to gain

Characteristics of a natural occurring outbreak of EHV1

121

better insights into the virulence of naturally circulating EHV1 strains and to determine if

EHM is in fact an emerging disease.

Acknowledgements

This research was supported by the Institute for the promotion of Innovation

through Science and Technology in Flanders (IWT-Vlaanderen). The authors like to

thank C. Boone for the help with the processing of all samples and Ine Vanherpe for

performing PCR and sequencing. We would also like to thank the veterinarians Marc

Schelkens, Jacques Corstiens, Wim Vermeiren, Dries Vermeiren, Kamiel Warnants, Jef

Nuytten and all horse owners for providing samples and detailed information.

Characteristics of a natural occurring outbreak of EHV1

122

REFERENCES

Allen, G. P. & Bryans, J. T. (1986). Molecular epizootiology, pathogenesis, and

prophylaxis of equine herpesvirus-1 infections. Prog Vet Microbiol Immunol 2,

78-144.

Edington, N., Bridges, C. G. & Patel, J. R. (1986). Endothelial cell infection and

thrombosis in paralysis caused by equid herpesvirus-1: equine stroke. Arch Virol

90, 111-124.

Garré, B., Gryspeerdt, A., Croubels, S., De Backer, P. & Nauwynck, H. (2009). Evaluation of orally administered valacyclovir in experimentally EHV1-infected

ponies. Vet Microbiol 135, 214-221.

Garré, B., van der Meulen, K., Nugent, J., Neyts, J., Croubels, S., De Backer, P. & Nauwynck, H. (2007). In vitro susceptibility of six isolates of equine herpesvirus

1 to acyclovir, ganciclovir, cidofovir, adefovir, PMEDAP and foscarnet. Vet

Microbiol 122, 43-51.

Gilkerson, J. R. & Barrett, E. J. (2008). Equine herpesvirus neurological disease.

Equine Vet J 40, 102-103.

Goehring, L. S., van Winden, S. C., van Maanen, C. & Sloet van Oldruitenborgh-Oosterbaan, M. M. (2006). Equine herpesvirus type 1-associated

myeloencephalopathy in The Netherlands: a four-year retrospective study (1999-

2003). J Vet Intern Med 20, 601-607.

Goodman, L. B., Wagner, B., Flaminio, M. J., Sussman, K. H., Metzger, S. M., Holland, R. & Osterrieder, N. (2006). Comparison of the efficacy of inactivated

combination and modified-live virus vaccines against challenge infection with

neuropathogenic equine herpesvirus type 1 (EHV-1). Vaccine 24, 3636-3645.

Gryspeerdt, A. C., Vandekerckhove, A. P., Garre, B., Barbe, F., Van de Walle, G. R. & Nauwynck, H. J. (2010). Differences in replication kinetics and cell tropism

between neurovirulent and non-neurovirulent EHV1 strains during the acute phase

of infection in horses. Vet Microbiol 142, 242-253.

Jackson, T. A., Osburn, B. I., Cordy, D. R. & Kendrick, J. W. (1977). Equine

herpesvirus 1 infection of horses: studies on the experimentally induced

neurologic disease. Am J Vet Res 38, 709-719.

Kydd, J. H., Smith, K. C., Hannant, D., Livesay, G. J. & Mumford, J. A. (1994a). Distribution of equid herpesvirus-1 (EHV-1) in respiratory tract associated

lymphoid tissue: implications for cellular immunity. Equine Vet J 26, 470-473.

Kydd, J. H., Smith, K. C., Hannant, D., Livesay, G. J. & Mumford, J. A. (1994b). Distribution of equid herpesvirus-1 (EHV-1) in the respiratory tract of ponies:

implications for vaccination strategies. Equine Vet J 26, 466-469.

Kydd, J. H., Townsend, H. G. & Hannant, D. (2006). The equine immune response to

equine herpesvirus-1: the virus and its vaccines. Vet Immunol Immunopathol 111,

15-30.

Kydd, J. H., Wattrang, E. & Hannant, D. (2003). Pre-infection frequencies of equine

herpesvirus-1 specific, cytotoxic T lymphocytes correlate with protection against

abortion following experimental infection of pregnant mares. Vet Immunol

Immunopathol 96, 207-217.

Lunn, D. P., Davis-Pointer, N., Flaminio, M. J., Horohov, D. W., Osterrieder, K., Pusterla, N. & Townsend, H. G. (2009). Equine Herpesvirus-1 Consensus

Statement. J Vet Intern Med 23, 450-461.

Characteristics of a natural occurring outbreak of EHV1

123

Maxwell, L. K., Bentz, B. G., Gilliam, L. L., Ritchey, J. W., Holbrook, T. C.,

McFarlane, D., Rezabek, G. B., MacAllister, C. G. & Allen, G. P. (2008). Efficacy of Valacyclovir Against Clinical Disease After EHV-1 Challenge in

Aged Mares. Proceedings of the Annual Convention of the AAEP 54, 198-199.

McCartan, C. G., Russell, M. M., Wood, J. L. & Mumford, J. A. (1995). Clinical,

serological and virological characteristics of an outbreak of paresis and neonatal

foal disease due to equine herpesvirus-1 on a stud farm. Vet Rec 136, 7-12.

Nugent, J., Birch-Machin, I., Smith, K. C., Mumford, J. A., Swann, Z., Newton, J. R., Bowden, R. J., Allen, G. P. & Davis-Poynter, N. (2006). Analysis of equid

herpesvirus 1 strain variation reveals a point mutation of the DNA polymerase

strongly associated with neuropathogenic versus non-neuropathogenic disease

outbreaks. J Virol 80, 4047-4060.

USDA (2007). Equine herpesvirus myeloencephalopathy: A potentially emerging disease.

Available at: http://wwwaphisusdagov/vs/nahss/equine/ehv.

Van de Walle, G. R., Goupil, R., Wishon, C., Damiani, A., Perkins, G. A. & Osterrieder, N. (2009). A Single-Nucleotide Polymorphism in a Herpesvirus

DNA Polymerase Is Sufficient to Cause Lethal Neurological disease. J Infect Dis

200, 20-25.

van der Meulen, K., Nauwynck, H. & Pensaert, M. (2000). Equine herpesvirus type 1

abortion, neonatal foal death and nervous system disorders diagnosed in Belgium

in 1999. Vlaams Diergenskund Tijds 69, 38-41.

van der Meulen, K., Vercauteren, G., Nauwynck, H. & Pensaert, M. (2003). A local

epidemic of equine herpesvirus 1-induced neurological disorders in Belgium.

Vlaams Diergenskund Tijds 72, 366-372.

van der Meulen, K. M., Gryspeerdt, A. C., Vandekerckhove, A. A., Garre, B. A. & Nauwynck, H. J. (2007). The protective properties of vaccination against equine

herpesvirus 1-induced viremia, abortion and nervous system disorders. Vlaams

Diergenskund Tijds 76, 186-194.

van Maanen, C., Sloet van Oldruitenborgh-Oosterbaan, M. M., Damen, E. A. & Derksen, A. G. (2001). Neurological disease associated with EHV-1-infection in

a riding school: clinical and virological characteristics. Equine Vet J 33, 191-196.

General discussion

125

CHAPTER VI

GENERAL DISCUSSION

General discussion

127

GENERAL DISCUSSION

Equine herpesvirus 1 (EHV1) is a ubiquitous equine alphaherpesvirus. After

replication in tissues of the upper respiratory tract (URT), EHV1 spreads via a leukocyte-

associated viremia to internal organs such as the pregnant uterus and the nervous system

where a second replication cycle is initiated in the endothelial cells of these organs. The

viral replication can lead to upper respiratory tract disease, abortion or neonatal foal death

and equine herpes myelitis (EHM) (Allen & Bryans, 1986; van Maanen, 2002). In

veterinary medicine, vaccination is the most designated weapon against viral infections.

Several vaccines attempting to prevent severe consequences of infection with EHV1 are

currently on the market. Unfortunately, to date, none of these is able to fully prevent the

EHV1-induced severe secondary symptoms (van der Meulen et al., 2007). Therefore,

effective antiviral medicines could be of great importance to limit virus replication and

clinical signs during acute and recurrent viral infections. To date, no antiviral drugs are

registrated for the use in horses, neither are they efficient enough to battle EHV1

infections. One of the major problems in developing efficient vaccines or antiviral drugs

is that the pathogenesis and immune evasion strategies of EHV1 are not fully known,

which necessitates further research on this matter.

In vivo experiments are a necessary tool to study the pathogenesis of specific

pathogens in animals. However, in the case of horses, such experiments are expensive,

labour-intensive and are often a sensitive matter from an ethical point of view. Therefore,

attempts have been made to find appropriate models to replace those horse experiments.

Laboratory animals may be convenient and valuable tools for use in experimental systems

of disease in large animals. However, they are often criticized because of their failure to

provide relevance to the disease in the target species. The most frequently used in vivo

model for the assessment of virological and histological characteristics of EHV1-induced

disease in horses has been the murine model. The extent to which valid comparisons and

extrapolations about pathogenesis can be made from mouse to horse remains highly

questionable (Walker et al., 1999). For example, the EHV1 strain RacL11 causes only

mild disease in horses (Van de Walle et al., personal communication), while in mice,

inoculation with RacL11 results in severe symptoms and even death (von Einem et al.,

2007). Even though the mouse model is suggested to provide a valid method for

General discussion

128

investigation into several aspects of EHV1-induced disease (Walker et al., 1999),

investigating the pathogenesis of EHV1 in its natural host, the horse, remains inevitable.

Therefore, EHV1 pathogenesis studies in the horse were performed in this thesis, which

provides relevant and unambiguous new insights. It was found that EHV1 replicates in a

plaquewise manner in the epithelium of the upper respiratory tract. Plaques were not able

to penetrate the basement membrane, but individual infected cells could be found in the

connective tissue from 1 dpi. These cells were mainly CD172a+ cells and CD5

+

lymphocytes. Viremia was detected from 1 dpi, with again CD172a+ cells as main carrier

cells. Infected epithelial cells and mucosal leukocytes were examined more thoroughly

and expression of viral late glycoproteins gB, gC, gD and gM was evaluated. It was found

that although a complete replication cycle with formation of plaques was seen in

epithelial cells, individual infected cells showed a low expression of late glycoproteins gC

and gD. The latter indicates that the protein expression in EHV1-infected immune cells is

hampered. Interestingly, the results obtained in this thesis were virtually identical as the

results obtained in the recently established in vitro model of equine nasal mucosa

(Vandekerckhove et al., 2010). Therefore, the latter model could be a valuable tool for the

study of EHV1 pathogenesis in vitro.

Based on the observations above, several new insights in the pathogenesis of EHV1

were obtained and some missing links were elucidated. Therefore, an improved model on

the pathogenesis of EHV1 in the upper respiratory tract is put forward in this thesis

(Figure 1). Upon infection, EHV1 replicates in epithelial cells of the nasal septum and

nasopharynx, which results in a productive infection with plaque formation and nasal

virus shedding (2). This nasal shedding allows transmission of virus to susceptible contact

animals and ensures that EHV1 can persist in the equine population. This productive

infection in epithelial cells, however, is self-limiting, most likely due to IFN-α

production. In addition, EHV1 seems to cross the basement membrane through the help of

CD172a+ carrier cells, probably mucosal dendritic cells (3). We postulate that these

infected carrier cells can transport the virus across the basement membranes and through

connective tissues in the direction of vascular endothelial cells (4a) on the one hand and

lymphatic epithelium (4b) on the other hand. As a result, a cell-associated viremia is

originated (5), together with migration of infected immune cells to local lymph nodes,

hereby initiating the start of a specific immune response. During viremia, also CD172a+

cells were the main carrier cells, suggesting that these cells indeed originate during the

General discussion

129

early stages of infection in the upper respiratory tract. Although epithelial cells undergo a

complete replication cycle with the progeny of new infectious virus particles, infected

leukocytes seem to have a hampered expression of some late proteins. The same

observation has been previously demonstrated, as it was seen that during viremia, 98% of

EHV1-infected PBMC present in the blood stream lacked expression of certain late

proteins (van der Meulen et al., 2003b). When we combine the latter results with our

findings, we can conclude that infected leukocytes in the upper respiratory tract could be

the source of the cell-associated viremia. These cells undergo an incomplete replication

cycle even in the early steps of pathogenesis, which might be a mechanism to protect

these cells from destruction by the immune system. Interestingly, the latter could explain

why despite high antibody levels, viremia is still observed in immune animals. Transfer of

EHV1 from viremic cells to endothelial cells of target organs (6) causes infection,

followed by replication of EHV1 and subsequent damage to these organs (7).

Figure 1. Adapted schematic overview of the pathogenesis of EHV1. 1) Attachment/penetration of virus in

epithelial cells of the upper respiratory tract; 2) replication of virus in epithelial cells by means of plaque

formation; 3) spread of virus through the BM by means of individual infected leukocytes; 4) spread of

infected leukocytes to vascular endothelial cells (4a) and draining lymph nodes (4b); 5) leukocyte

associated viremia in CD172a+ carrier cells; 6) transfer of virus from infected leukocytes to endothelial cells

of target organs; 7) replication and spread of virus in target organs. Full arrow: known mechanism; striped

arrow: unknown mechanism.

1

4a

5

67

3

4b

2

General discussion

130

New insights in the pathogenesis and the natural occurrence of EHV1, may provide

new directions in the amelioration of vaccines and the development of new drugs for

treatment. Indeed, at present, the two most important tools in the battle against EHV1 are

vaccination and antiviral therapy.

An efficient vaccine against EHV1 is of great importance to control outbreaks of

EHV1 and prevent the development of serious problems such as abortion and nervous

system disorders. Firstly, during this research, we have observed that the first steps in the

pathogenesis of EHV1 occur very rapidly. During primary replication in tissues of the

upper respiratory tract, single infected cells are already seen at 1 day pi (dpi) and plaque

formation was seen from 2 dpi. Already after 1 or 2 dpi, single infected cells were present

beneath the basement membrane and viremia could be detected. The latter implies that

vaccines can only prevent viremia and subsequent spread to internal organs if an effective

local immunity can be established at the level of the upper respiratory tract, preventing

the origin of individual infected cells beneath the basement membrane and the subsequent

viremia. Because horses have been shown to be protected against viremia up to 6 months

after a natural infection with EHV1 (Breathnach et al., 2001; Edington & Bridges, 1990),

local immunity can prevent viremia at least for some time. This makes a modified life

vaccine (MLV) a good candidate in vaccine development and a suitable recombinant

EHV1 MLV could be a virus which is able to replicate in the epithelium of the upper

respiratory tract but unable to penetrate the basement membrane by the use of individual

infected cells. As it has been shown that the mucosal nasal explant model is an accurate

alternative for in vivo testing, this model can be used to rapidly screen several potential

MLV candidates before starting expensive and labour-intensive in vivo experiments

(Vandekerckhove et al., 2009; Vandekerckhove et al., 2010). Hereby, suitable

recombinant viruses could be selected for the use in in vivo experiments based on their

inability to penetrate the basement membrane by using individual infected immune cells.

Secondly, during viremia, carrier cells of EHV1 are present in a blocked stage as certain

late glycoproteins are not expressed (van der Meulen et al., 2006). In fact, we could

demonstrate that this immune evasion strategy already took place in the early stages of

infection, as soon as individual infected cells appeared in the connective tissues following

penetration of the basement membrane (Gryspeerdt et al., 2011). Such immune evasion

strategy could supposedly protect them against antibody-dependent destruction,

suggesting that humoral antibodies are of no use during this phase of infection (van der

General discussion

131

Meulen et al., 2003b). However, a humoral protection could be of importance in the last

step of the pathogenesis, when virus is transferred from its carrier cells to the endothelium

of the target organs. Here, antibodies can play a role in either (i) preventing virus to be

transmitted from carrier cells to endothelial cells, or (ii) limiting the extent of plaque

formation in target organs. Unfortunately, a recent study demonstrated that virus could be

transferred in vitro to endothelial cells despite the presence of neutralizing antibodies

(Goehring et al., 2010), meaning that humoral immunity will probably be of limited use

in this step of the pathogenesis. However, antibodies may still play an important role in

limiting replication in target organs and thus limiting the damage and the occurrence of

severe symptoms. The latter could be of interest for future research, where the effect of

neutralizing antibodies on plaque formation and virus spread in endothelial cells of target

organs should be evaluated.

A thorough knowledge on the pathogenesis of the virus is of paramount importance

for the development of antivirals. Based on the results of this thesis, we are now able to

predict where an antiviral could exert its action. When we look at the rapid invasion of

EHV1 into the connective tissue, combined with the fact that carrier cells are already in a

blocked stage during early pathogenesis, there are only three places where antivirals could

have an effect on the replication of EHV1 in the horse: (i) the entry and primary

replication of the virus in the mucosa of the URT, (ii) the transmission of virus from

infected PBMC to endothelial cells, and (iii) the secondary replication of the virus in

endothelial cells at the target organs. To have a rapid effect on the primary replication in

the epithelium of the URT, an antiviral with a metaphylactic use, applied locally in the

nose to protect horses from getting a productive infection, would be the most suitable. An

antiviral with an effect on the amount and duration of viremia will be difficult since

during viremia the infected leukocyte seems to be blocked, which limits the possible

targets of antivirals (van der Meulen et al., 2003b). Due to the rapid course of an EHV1

infection, most animals during an outbreak will already be in a late stage of infection,

namely the secondary replication in endothelial cells of the target organs, before a therapy

can be initiated. An antiviral which either inhibits and/or limits the extent of this

secondary replication seems to have great potential in limiting damage at secondary target

organs and therefore severe symptoms. However, until now, the latter is difficult to

evaluate since there is no appropriate in vitro or in vivo model to study the possible effect

of an antiviral at secondary replication sites. Given the fact that after oral valacyclovir

General discussion

132

treatment no influence on nasal shedding or viremia was observed (Garré et al., 2009),

but that a clinical effect and a more rapid recovery is suspected (Maxwell et al., 2008), it

seems that an effect of valacyclovir on the secondary replication of EHV1 cannot be

excluded. Interestingly, during the outbreak of neurological disease studied in this thesis,

a rapid spread of the virus was seen followed by a rapid appearance of neurological

disorders. Some horses did not show previous signs such as nasal discharge, fever or

ataxia and were merely found paralyzed, necessitating euthanasia. As treatment should

preferentially be initiated before severe symptoms such as paralysis become apparent

(Maxwell et al., 2008), any antiviral treatment would have been initiated too late in the

course of disease. As there is no way to predict in which stage of infection the horse is

during a natural outbreak of EHV1, an efficient antiviral drug should have both a

metaphylactic and a curative action. The best way to achieve this goal would be a local

treatment via aerosol, to prevent primary replication in tissues of the upper respiratory

tract. Horses with (severe) symptoms however, should be treated in a way to prevent or

inhibit further extension of endothelial damage and should be treated orally or

intravenously with a curative antiviral drug. Future research should be focussed on how

EHV1 penetrates through the basement membrane and gets into the blood stream. Also

the mechanism of transmission of the virus from infected PBMC to endothelial cells

should be studied. These are two phenomena where future antivirals could possibly

interfere with the pathogenesis of the virus, hereby limiting severe consequences after

infection with EHV1.

In 2009, several outbreaks of abortion and neonatal death were seen during the first

period of the year. Exceptionally, a large outbreak of EHV1-induced neurological disease

was seen in Antwerp. Concern has been voiced within the U.S. horse industry that the

neurologic form of EHV1 is emerging. The latter is based on the detection of an increased

number of EHM outbreaks throughout the United States over the last decade (USDA,

2007). Until now, no such evolution was seen in Belgium and no reports of large EHM

outbreaks have been observed with the exception of one limited outbreak in 2003 (van der

Meulen et al., 2003a). The recent large outbreak of EHM showed a rapid spread and a

remarkably high number of animals with severe neurological symptoms. The latter,

together with the report of two novel outbreaks in 2010, raises the question whether the

neurological form of EHV1 is also emerging in Belgium, and hence, Europe. In 2009, one

General discussion

133

outbreak of EHM was reported in the UK, one in France and three in Ireland. In 2010,

five outbreaks were reported in France, two in the UK and one in Italy. Until now, three

outbreaks of EHM have been reported in France in 2011. Unfortunately, most European

countries do not report outbreaks of EHM, providing insufficient data to decide if EHM is

possibly emerging in Europe. Additionally, it is not clear if the reported observations are

related to an evolving disease with a changed virulence and behaviour or if they are the

consequence of a greater awareness and the development of better diagnostic methods

(USDA, 2007). Many data gaps exist, and more investigations need to be conducted to

better understand this evolving situation and to identify other possible factors playing a

role. Therefore, a good surveillance system in Belgium and Europe would be of great

importance to monitor outbreaks of EHM and to get better insights into the virulence of

naturally circulating EHV1 strains. An efficient communication system and a centralized

diagnostic centre would be of paramount importance to achieve this goal. Diagnostical

methods should be quick and efficient to have a rapid diagnosis during suspected

outbreaks of EHV1 (Lunn et al., 2009). Although virus isolation proved to be a reliable

method to confirm infection within two days, PCR techniques could be of importance as

results can be achieved within a few hours and information could be given about the

neurovirulence of the isolated EHV1 strain and the quantity of virus present (Allen, 2007;

Pusterla et al., 2008; Pusterla et al., 2009b).

Worldwide, several guidelines are available of what needs to be done during an

outbreak of EHV1. These guidelines vary widely both within and between countries. A

protocol on how to manage an EHV1 outbreak, from being prepared to collect samples

appropriately and how to contain the situation would be of great importance (USDA,

2007). Some guidelines found in literature, formed the basis of our approach during the

outbreak of EHM and were checked for their efficiency (Allen, 2002; Lunn et al., 2009;

Pusterla et al., 2009a). Based on our observations, we would like to discuss some of these

guidelines.

In the case of a suspected outbreak of EHV1, immediate measures should be taken to

prevent any possible spread of the virus within the affected premise or to other premises

(Lunn et al., 2009). Affected horses should be put in isolation and contact with other

horses or care takers should be completely avoided. Other horses should be monitored for

the appearance of fever or other symptoms suggesting an early infection (van Maanen,

2002). If possible, all horse movement in affected regions should be avoided until three

General discussion

134

weeks after the last reported case. During the outbreak of EHM in Antwerp, isolation and

quarantine measures proved to be of great importance to prevent further spread of the

virus. Suspected premises were closed until EHV1 infection was either confirmed or

excluded. Quarantine measures in confirmed premises were only lifted three weeks after

the last observed clinical case. Major horse events were cancelled and all horse

movements in affected regions were discouraged.

An early and accurate diagnosis of the cause of the observed symptoms, with virus

culture and isolation as the gold standard test, is of paramount importance to further

determine the strategies to be followed in managing the outbreak (Lunn et al., 2009).

Several samples were collected during the outbreak: unclotted blood samples for isolation

of PBMC, nasal swabs and paired serum samples. Isolation of EHV1 by means of

cocultivation of PBMC proved to be an important and efficient diagnostic tool. Within

two days, virus could be isolated from affected horses with fever, ataxia or paralysis.

Although nasal swabbing could be a good alternative in an even earlier stage of infection,

during this outbreak, no virus could be isolated from collected swabs. An explanation for

this observation could be inappropriate conservation techniques or the long transport

period under non-ideal circumstances. A seroconversion could be noticed in almost all

infected horses, but such methodology takes about 4 weeks before the final results are

known. Therefore, the best samples to collect in an acute stage of infection, at least in our

hands, seemed to be unclotted blood samples. All samples should be immediately put at

4°C and sent as soon as possible to the laboratory.

EHV1 infections in horses are very difficult to treat. The virus has a peculiar

pathogenesis which makes it hard to interfere. Until now, no antivirals have been licensed

in Belgium for the use in horses, limiting treatment to intensive supportive therapy only

(van Maanen, 2002), along with supportive antibiotics and anti-inflammatory drugs.

During several naturally occurring outbreaks of EHV1-induced neurological disease,

attempts were made to prevent infection or treat horses with severe symptoms using

antiviral drugs (Friday et al., 2000; Henninger et al., 2007; van der Meulen et al., 2003a).

Acyclovir and its prodrug valacyclovir are the most popular drugs in this matter. During

the outbreak in Antwerp, a limited number of animals were treated orally with

valacyclovir. Treated horses received an oral dose of valacyclovir of 40 mg/kg body

weight, three times a day. This treatment was initiated as soon as possible. Preferentially a

treatment should be started before paralysis occurs in order to stop this process. However,

General discussion

135

evaluation of a drug in clinical cases will always remain difficult as animals are mostly

not treated under identical conditions and EHV1-negative animals need to be present as

controls to exclude any influence of the immune system. Despite these limitations, it was

observed in this outbreak that valacyclovir-treated animals seemed to recover more

rapidly from clinical symptoms during treatment than non-treated animals. Treatments

with anti-inflammatory drugs and antibiotics to prevent secondary bacterial infections

seemed to contribute to a better recovery. Finally, intensive care greatly improved the

chance of recovery in the severely affected horses, emphasizing the benefit of such

treatment.

The development of a fully protective vaccine against EHV1 would be the most

efficient way to eradicate EHV1-induced problems from the horse population. Until an

efficient vaccine has been found, management procedures are still the most important

measures to prevent or control an outbreak of EHV1 (Lunn et al., 2009; van Maanen,

2002). During the present outbreak, vaccination of horses against EHV1 was highly

recommended. Within an affected premise, horses without clinical symptoms were

vaccinated. Further, owners of large premises and organisers of competitions were

advised to prohibit contact between their horses and visiting horses, and to allow only

horses on their premises which were vaccinated. To date, only inactivated vaccines are

commercially available in Belgium. Although they can not completely prevent abortion or

nervous system disorders, they seemed to protect horses partially from abortion or

nervous system disorders during the outbreak of EHM. It was observed that morbidity

rates during the outbreak of 2009 were the lowest on the first affected premise where a

correct vaccination protocol was used. Moreover, and as far as we know, virus only

spread to premises where horses were not vaccinated against EHV1, suggesting some

clinical protection of vaccinated horses against severe symptoms (Henninger et al., 2007;

Studdert et al., 2003).

In conclusion, given the peculiar and rapid pathogenesis of EHV1, the development of

a curative antiviral will be a very difficult task, as the points of action of such antiviral are

rather limited. Therefore, the best way to protect horses from severe complications due to

an EHV1 infection, is to prevent them from getting infected. This goal can be achieved in

two ways: i) the development of a fully protective vaccine and/or ii) the development of

an antiviral with a metaphylactic action to protect in-contact horses during the course of

General discussion

136

an outbreak. Until further research is able to fulfil the above mentioned goals,

management procedures are still very important measures.

General discussion

137

References

Allen, G. P. (2002). Epidemic disease caused by equine herpesvirus-1: Recommendations

for prevention and control. Equine Vet Educ 4, 177-183.

Allen, G. P. (2007). Development of a real-time polymerase chain reaction assay for

rapid diagnosis of neuropathogenic strains of equine herpesvirus-1. J Vet Diagn

Invest 19, 69-72.

Allen, G. P. & Bryans, J. T. (1986). Molecular epizootiology, pathogenesis, and

prophylaxis of equine herpesvirus-1 infections. Prog Vet Microbiol Immunol 2,

78-144.

Breathnach, C. C., Yeargan, M. R., Sheoran, A. S. & Allen, G. P. (2001). The

mucosal humoral immune response of the horse to infective challenge and

vaccination with Equine herpesvirus-1 antigens. Equine Vet J 33, 651-657.

Edington, N. & Bridges, C. G. (1990). One way protection between equid herpesvirus 1

and 4 in vivo. Research in veterinary science 48, 235-239.

Friday, P. A., Scarratt, W. K., Elvinger, F., Timoney, P. J. & Bonda, A. (2000). Ataxia and paresis with equine herpesvirus type 1 infection in a herd of riding

school horses. J Vet Intern Med 14, 197-201.

Garré, B., Gryspeerdt, A., Croubels, S., De Backer, P. & Nauwynck, H. (2009). Evaluation of orally administered valacyclovir in experimentally EHV1-infected

ponies. Vet Microbiol 135, 214-221.

Goehring, L. S., Hussey, G. S., Ashton, L. V., Schenkel, A. R. & Lunn, D. P. (2010). Infection of central nervous system endothelial cells by cell-associated EHV-1.

Vet Microbiol doi:10.1016/j.vetmic.2010.08.030.

Gryspeerdt, A. C., Vandekerckhove, A. P., Bannazadeh Baghi, H., Van de Walle, G. R. & Nauwynck, H. J. (2011). Expression of late viral proteins is hampered in

infected nasal mucosal leukocytes but not in epithelial cells during early

pathogenesis of equine herpesvirus type 1 (EHV1) infection. The Veterinary

Journal in press.

Henninger, R. W., Reed, S. M., Saville, W. J., Allen, G. P., Hass, G. F., Kohn, C. W. & Sofaly, C. (2007). Outbreak of Neurologic disease Caused by Equine

Herpesvirus-1 at a University Equestrian Center. J Vet Intern Med 21, 157-165.

Lunn, D. P., Davis-Pointer, N., Flaminio, M. J., Horohov, D. W., Osterrieder, K., Pusterla, N. & Townsend, H. G. (2009). Equine Herpesvirus-1 Consensus

Statement. J Vet Intern Med 23, 450-461.

Maxwell, L. K., Bentz, B. G., Gilliam, L. L., Ritchey, J. W., Holbrook, T. C.,

McFarlane, D., Rezabek, G. B., MacAllister, C. G. & Allen, G. P. (2008). Efficacy of Valacyclovir Against Clinical Disease After EHV-1 Challenge in

Aged Mares. Proceedings of the Annual Convention of the AAEP 54, 198-199.

Pusterla, N., David Wilson, W., Madigan, J. E. & Ferraro, G. L. (2009a). Equine

herpesvirus-1 myeloencephalopathy: a review of recent developments. Vet J 180,

279-289.

Pusterla, N., Mapes, S. & Wilson, W. D. (2008). Use of viral loads in blood and

nasopharyngeal secretions fot the diagnosis of EHV-1 infection in field cases. Vet

Rec 162, 728-729.

Pusterla, N., Wilson, W. D., Mapes, S., Finno, C., Isbell, D., Arthur, R. M. & Ferraro, G. L. (2009b). Characterization of viral loads, strain and state of equine

General discussion

138

herpesvirus-1 using real-time PCR in horses following natural exposure at a

racetrack in California. Vet J 179, 230-239.

Studdert, M. J., Hartley, C. A., Dynon, K., Sandy, J. R., Slocombe, R. F., Charles, J. A., Milne, M. E., Clarke, A. F. & El-Hage, C. (2003). Outbreak of equine

herpesvirus type 1 myeloencephalitis: new insights from virus identification by

PCR and the application of an EHV-1-specific antibody detection ELISA.

Veterinary Record 153, 417-423.

USDA (2007). Equine herpesvirus myeloencephalopathy: A potentially emerging disease.

Available at: http://wwwaphisusdagov/vs/nahss/equine/ehv.

van der Meulen, K., Caij, B., Pensaert, M. & Nauwynck, H. (2006). Absence of viral

envelope proteins in equine herpesvirus 1-infected blood mononuclear cells during

cell-associated viremia. Vet Microbiol 113, 265-273.

van der Meulen, K., Vercauteren, G., Nauwynck, H. & Pensaert, M. (2003a). A local

epidemic of equine herpesvirus 1-induced neurological disorders in Belgium.

Vlaams Diergenskund Tijds 72, 366-372.

van der Meulen, K. M., Gryspeerdt, A. C., Vandekerckhove, A. A., Garre, B. A. & Nauwynck, H. J. (2007). The protective properties of vaccination against equine

herpesvirus 1-induced viremia, abortion and nervous system disorders. Vlaams

Diergenskund Tijds 76, 186-194.

van der Meulen, K. M., Nauwynck, H. J. & Pensaert, M. B. (2003b). Absence of viral

antigens on the surface of equine herpesvirus-1-infected peripheral blood

mononuclear cells: a strategy to avoid complement-mediated lysis. J Gen Virol 84,

93-97.

van Maanen, C. (2002). Equine herpesvirus 1 and 4 infections: an update. Vet Q 24, 58-

78.

Vandekerckhove, A., Glorieux, S., Broeck, W. V., Gryspeerdt, A., van der Meulen, K. M. & Nauwynck, H. J. (2009). In vitro culture of equine respiratory mucosa

explants. Vet J 181, 280-287.

Vandekerckhove, A. P., Glorieux, S., Gryspeerdt, A. C., Steukers, L., Duchateau, L., Osterrieder, N., Van de Walle, G. R. & Nauwynck, H. J. (2010). Replication

kinetics of neurovirulent versus non-neurovirulent equine herpesvirus type 1

strains in equine nasal mucosal explants. J Gen Virol 91, 2019-2028.

von Einem, J., Smith, P. M., Van de Walle, G. R., O'Callaghan, D. J. & Osterrieder, N. (2007). In vitro and in vivo characterization of equine herpesvirus type 1

(EHV-1) mutants devoid of the viral chemokine-binding glycoprotein G (gG).

Virology 362, 151-162.

Walker, C., Love, D. N. & Whalley, J. M. (1999). Comparison of the pathogenesis of

acute equine herpesvirus 1 (EHV-1) infection in the horse and the mouse model: a

review. Vet Microbiol 68, 3-13.

Summary

139

SUMMARY

Summary

141

SUMMARY

Equine herpesvirus 1 (EHV1) is the most important infectious cause of abortion

worldwide. Infection can also result in severe nervous system disorders with frequent

fatal outcome. Horses are infected via the respiratory system. After local replication in the

upper respiratory tract, virus spreads via a cell-associated viremia to the uterus and/or the

nervous system. Replication in endothelial cells of these organs can result in abortion or

nervous system disorders. Vaccination with current available vaccines against EHV1 can

not offer full protection against these severe symptoms. Therefore there’s an urgent need

for improvement of existing vaccines or the development of alternative strategies for

prevention or treatment of EHV1 infections. However, the lack of knowledge about the

invasion strategies of EHV1 is an important obstacle. A thorough knowledge about the

pathogenesis of EHV1 is crucial for vaccine development or efficient therapy.

In Chapter 1, an overview of the current knowledge of EHV1 was given. First a short

overview of the common characteristics of EHV1 was given. Further, interactions of the

virus with the cell and with the horse as host were reviewed.

In Chapter 2, the aims of the studies described in the thesis are put forward. The first

aim was to gain a more detailed insight in the invasion strategies of EHV1 strains from

the upper respiratory tract until the origin of a cell-associated viremia. The second aim

was to examine how individual EHV1-infected cells escape from immune effector

mechanisms at the primary site of replication. The third and last aim was to gain more

information about naturally occurring outbreaks in Belgium in terms of risk factors,

protective properties of vaccination, the effect of experimental therapies and diagnosis.

The first aim was addressed in Chapter 3. An experimental inoculation study in

ponies was performed with both a neurovirulent and a non-neurovirulent strain. Two

groups of six ponies were inoculated intranasally with either strain. Clinical signs, nasal

shedding and viremia were evaluated. At early time points post inoculation, one pony of

each group was euthanized. Tissues were collected for titration and immunostainings.

Number and size of EHV1-induced plaques were calculated, and individual EHV1-

infected cells were quantified and characterized. Inoculation with either strain resulted in

Summary

142

nasal shedding and replication in several tissues of the upper respiratory tract. Both

strains replicated in a plaquewise manner in epithelium of the nasal mucosa, but

replication in epithelium of the nasopharynx was largely limited to non-neurovirulent

EHV1. Plaques were never able to cross the basement membrane, but individual infected

cells were noticed in the connective tissue of all examined tissues for both strains. The

total number of these cells however, was 3-7 times lower with non-neurovirulent EHV1

compared to neurovirulent EHV1. CD172a+ cells and CD5

+ lymphocytes were important

target cells for both strains. Interestingly, in lymph nodes, B-lymphocytes were also

important target cells for EHV1, irrespective of the strain. Viremia was detected very

early post inoculation and carrier cells were mainly CD172a+ for both strains.

The second aim was addressed in Chapter 4. Since both epithelial and mononucleair

cells appeared to be crucial cell types during the pathogenesis of EHV1, we studied if

EHV1 uses immuno-evasive techniques in these cell types during the early stages of

pathogenesis. Therefore, the expression of viral late proteins gB, gC, gD and gM in these

target cells was studied in different experimental models. Firstly, RK13 cells and

peripheral blood mononuclear cells were infected with EHV1 in vitro. Secondly, the

recently established equine nasal mucosal explant system was used and thirdly, nasal

mucosal tissues of experimentally EHV1-infected ponies were analyzed. Our salient

findings were that for all three systems tested, the late proteins gB, gC, gD and gM were

expressed on EHV1-infected epithelial cells. On the contrary, gB and gM expression was

observed in 60 to 90% of infected mononuclear cells, whereas expression of gC and gD

on these cells was hampered, because only 20% of EHV1-infeced mononuclear cells

showed gC and gD expression. These results indicate that the expression of certain viral

proteins depends upon the cell type infected and that a hampered expression could be a

mean of EHV1 to stay invisible for the immune system during the early pathogenesis of

EHV1.

The third aim was addressed in Chapter 5. To gain more information about EHV1

problematics in Belgium, a recent outbreak of EHV1-induced neurologic disease in

Flanders which involved at least 13 premises, was both clinically as genetic analyzed. A

total of 212 horses was exposed to EHV1 and a morbidity of 26% was seen. The

incidence of neurological disorders in the affected horses was 43%. Furthermore, the

Summary

143

outbreak was characterized by the rapid occurrence of ataxia and paralysis immediately

after disappearance of fever. Morbidity and virus spread was remarkably higher at

premises were no or an incorrect vaccination schedule was used. Mares seemed to be

more sensible for the development of nervous system disorders with fatal outcome. A

positive influence of valacyclovir treatment during the course of disease was seen if

treatment was initiated immediately after appearance of nervous system disorders. EHV1

was diagnosed in each case by means of virus isolation and/or the detection of

seroconversion. Isolated virus was classified as neurovirulent EHV1, with al isolates

belonging to group 4 upon sequencing in the ORF30 and ORF68 region. The latter

confirms the suspicion of the outbreak spreading from the first premise.

Following general conclusions can be made from the studies performed in the

present thesis:

1. EHV1 has a rapid course after infection and replicates in the epithelium of the

upper respiratory tract in a plaquewise manner. These plaques do not penetrate the

basement membrane, but individual infected cells can be observed in the

connective tissue. These cells are mainly CD172a+ cells and CD5

+ lymphocytes.

At the time point of appearance of these individual infected cells, a cell-associated

viremia could be noticed, harbouring the virus mainly in CD172a+ cells.

2. In the beginning of infection individual infected cells are already in a blocked

stage, allowing them to elope the immune system. Contrary to infected epithelial

cells who expressed al tested glycoproteins, individual infected cells only express

gC and gD in only 20% of the cases.

3. As observed in America, equine herpes myelitis caused by infection with EHV1

seems to be emerging in Belgium. After the large outbreak in 2009, also in 2010

two small outbreaks of EHM were observed, showing a larger number of horses

with nervous system disorders and a fast spread of the virus. Again, this points out

the necessity of a thorough research into the pathogenesis of EHV1, in order to

develop efficient vaccines and antiviral therapy. Vaccinated horses seem to have a

certain clinical protection against the appearance of severe symptoms. Diagnosis

is possible by means of virus isolation and seroconversion. If treatment with

Summary

144

valacyclovir is started at an early time point in infection, horses seem to have a

quicker and more complete recovery.

Samenvatting

145

SAMENVATTING

Samenvatting

147

SAMENVATTING

Infectie met het equiene herpesvirus 1 (EHV1) is wereldwijd de belangrijkste

infectieuze oorzaak van abortus bij paarden. Infectie kan ook resulteren in ernstige

zenuwstoornissen met veelal fatale afloop. Paarden worden geïnfecteerd via het

ademhalingsstelsel. Na lokale vermeerdering in de bovenste luchtwegen verspreidt het

virus zich via een celgeassocieerde viremie naar baarmoeder en/of zenuwstelsel. Daar kan

vermeerdering van EHV1 ter hoogte van endotheelcellen resulteren in abortus of

zenuwstoornissen. Vaccinatie met de huidig beschikbare vaccins tegen EHV1 kan geen

volledige bescherming bieden tegen deze ernstige symptomen. Er is dus nood aan

verbetering van vaccins of en het ontwikkelen van alternatieve strategieën voor preventie

of behandeling van EHV1 infecties. Een belangrijke hinderpaal hierbij is het gebrek aan

kennis over de invasie van EHV1. Een grondige kennis van de pathogenese van EHV1 is

onontbeerlijk voor de ontwikkeling van efficiënte vaccins of een doelgerichte therapie.

In Hoofdstuk 1 werd een overzicht gegeven van de huidige kennis over EHV1. Eerst

werd een beknopte uitleg gegeven over de algemene eigenschappen van het virus.

Vervolgens werden de interacties van het virus met de cel en met het paard als gastheer

besproken.

In Hoofdstuk 2 werden de doelstellingen van de verschillende studies uiteengezet. De

eerste doelstelling bestond erin om een meer gedetailleerd inzicht te verkrijgen in de

invasie van EHV1 stammen van de bovenste ademhalingswegen tot aan de

celgeassocieerde viremie. De tweede doelstelling was na te gaan hoe individuele EHV1-

geïnfecteerde cellen er in slagen te ontsnappen aan de immuun effector mechanismen op

de plaats van primaire vermeerdering. De derde en laatste doelstelling was om het verloop

van natuurlijke infecties met EHV1 beter in kaart te brengen wat betreft risicofactoren,

mogelijke bescherming door middel van vaccinatie, het effect van experimentele

therapieën en diagnostische middelen.

In Hoofdstuk 3 kwam de eerste doelstelling aan bod. Een experimentele inoculatie

met zowel een neurovirulente als een niet-neurovirulente stam van EHV1 werd

uitgevoerd bij pony’s. Twee groepen van 6 pony’s werden intranasaal geïnoculeerd met

Samenvatting

148

één van beide stammen. Klinische tekenen, uitscheiding via de neus en viremie werden

geëvalueerd. Tijdens de eerste dagen na infectie werd telkens 1 pony van elke groep

geëuthanaseerd. Er werden weefsels verzameld voor titratie en immunofluorescentie

kleuringen. Het aantal en de grootte van EHV1-geïnduceerde plaques werd bepaald en

EHV1-geïnfecteerde individuele cellen werden gekwantificeerd en gekarakteriseerd.

Inoculatie met elke stam resulteerde in virusuitscheiding via de neus en een replicatie in

verschillende weefsels van de bovenste ademhalingswegen. Beide stammen

vermeerderden door middel van plaque formatie in de mucosa van de neus, maar

replicatie in het epitheel van de nasopharynx was grotendeels gelimiteerd tot niet-

neurovirulent virus. Plaques doorbraken op geen enkel moment de basaalmembraan, maar

voor beide stammen werden individueel geïnfecteerde cellen gevonden in het

bindweefsel. Het aantal individuele cellen was echter 3-7 maal lager voor niet-

neurovirulent EHV1 vergeleken met neurovirulent EHV1. CD172a+ cellen en CD5

+

lymfocyten waren de belangrijkste doelwitcellen voor beide stammen. In lymfeknopen

bleken B-lymfocyten ook belangrijke dragercellen te zijn voor EHV1, onafhankelijk van

welke stam gebruikt werd. Viremie werd reeds vroeg na infectie vastgesteld en de

geïnfecteerde cellen bleken voor beide stammen vooral CD172a+ te zijn.

In Hoofdstuk 4 kwam de tweede doelstelling aan bod. Gezien het feit dat zowel

epitheliale als mononucleaire cellen cruciale celtypes bleken te zijn gedurende de

pathogenese van EHV1, werd in dit hoofdstuk nagegaan of EHV1 in deze cellen reeds

vroeg in de pathogenese gebruik maakt van immuno-evasieve technieken. Daarom werd

de expressie van de late virale proteïnen gB, gC, gD en gM in deze doelwitcellen

nagegaan in verschillende experimentele modellen. Ten eerste werden RK13 cellen en

perifere bloed mononucleaire cellen in vitro geïnfecteerd met EHV1. Ten tweede werd

het onlangs gerealiseerde equiene nasale explant systeem gebruikt. Ten derde werd de

expressie nagegaan in nasale mucosa van experimenteel geïnfecteerde pony’s. In elk van

de drie geteste systemen bleken de late eiwitten gB, gC, gD en gM volledig tot expressie

te komen in EHV1-geïnfecteerde epitheliale cellen. Dit was in tegenstelling tot

mononucleaire cellen, waar gB en gM expressie werd gezien bij 60 tot 90% van de

geïnfecteerde cellen, terwijl gC en gD expressie enkel werd gezien bij 20% van de

geïnfecteerde cellen. De expressie van bepaalde virale proteïnen lijkt dus afhankelijk te

Samenvatting

149

zijn van het celtype dat geïnfecteerd wordt en het lijkt er op dat EHV1 op deze manier

kan verborgen blijven voor het immuun systeem in vroege stadia van infectie.

In Hoofdstuk 5 kwam de derde doelstelling aan bod. Om meer informatie te

verkrijgen over de EHV1 problematiek in België, werd een recente uitbraak van EHV1-

geïnduceerde neurologische stoornissen in Vlaanderen, waarbij minstens 13 bedrijven

werden getroffen, zowel klinisch als genetisch grondig onderzocht. In totaal werden 212

paarden blootgesteld aan EHV1 en werd een morbiditeit van 26% gezien. De incidentie

van neurologische stoornissen bij aangetaste paarden bedroeg 43%. Verder werd deze

uitbraak gekarakteriseerd door het snelle optreden van ataxie en paralyse na een korte

periode van koorts. De morbiditeit en spreiding van het virus was merkelijk hoger op

bedrijven waar niet of niet correct werd gevaccineerd. Merries leken gevoeliger voor het

ontwikkelen van zenuwstoornissen met fatale afloop. Indien onmiddellijk na het

verschijnen van zenuwstoornissen gestart werd met een valacyclovir behandeling, werd

een positief effect gezien op het ziekteverloop. Diagnose was in alle gevallen mogelijk

door ofwel virusisolatie uit perifere bloed mononucleaire cellen ofwel bepaling van

seroconversie bij gepaarde serum stalen. Het geïsoleerde virus werd geclassificeerd als

neuropathogeen, behorende tot groep 4 na sequenering in de ORF30 en ORF68 regio. Dit

laatste bevestigt het vermoeden dat de uitbraak verspreid werd vanuit het eerste

aangetaste bedrijf.

Op basis van de studies uitgevoerd in deze thesis kunnen de volgende algemene

conclusies naar voor gebracht worden:

1. EHV1 kent een snel verloop na infectie en vermeerdert in het epitheel van de

bovenste ademhalingswegen door middel van plaque formatie. Deze plaques zijn

niet in staat om de basaalmembraan te doorbreken, maar individueel geïnfecteerde

cellen kunnen worden teruggevonden in het bindweefsel. Deze cellen zijn vooral

CD172a+ cellen en CD5

+ lymfocyten. Op het moment van het verschijnen van

deze individueel geïnfecteerde cellen kon een celgeassocieerde viremie

waargenomen worden, waarbij het virus zich vooral in CD172a+ cellen bevindt.

2. Reeds in het begin van de infectie bevinden individueel geïnfecteerde cellen zich

in een geblokkeerde staat, waarbij ze kunnen ontsnappen aan het immuun

mechanisme. In tegenstelling tot geïnfecteerde epitheelcellen die alle geteste

Samenvatting

150

glycoproteines tot expressie brachten, werden in de individueel geïnfecteerde

leukocyten enkel in 20% van de gevallen gC en gD tot expressie gebracht.

3. Naar analogie met gebeurtenissen in Amerika, lijkt ook in België equiene herpes

myelitis door EHV1 in opmars. Na een grote uitbraak in 2009, waren er ook twee

kleine uitbraken in 2010, waarbij het voorkomen van zenuwstoornissen in een

groter percentage van de gevallen optrad en de spreiding sneller verliep. Dit

benadrukt nogmaals het belang van grondig onderzoek naar de pathogenese van

EHV1, voor gerichte vaccin- en therapie ontwikkeling. Gevaccineerde paarden

lijken een bepaalde graad van klinische bescherming te bezitten tegenover het

optreden van ernstige symptomen. Diagnose is mogelijk door middel van

virusisolatie en seroconversie. Bij het tijdig starten van een behandeling met

valacyclovir lijken paarden sneller en vollediger te herstellen.

Curriculum Vitae

151

CURRICULUM VITAE

Curriculum Vitae

153

CURRICULUM VITAE

Annick Gryspeerdt werd geboren op 3 april 1981 te Roeselare. Na het beëindigen

van het secundair onderwijs, richting Latijn-Wetenschappen aan het Bisschoppelijk

Lyceum der Grauwe Zusters te Roeselare, begon ze in 1999 met de studies

Diergeneeskunde aan de Universiteit Gent. In 2005 behaalde ze het diploma van

dierenarts met onderscheiding.

Onmiddellijk na het beëindigen van haar studies trad zij in dienst bij de vakgroep

Virologie, Parasitologie en Immunologie bij het laboratorium voor Virologie. Ze

verrichtte er een onderzoeksstudie getiteld “De pathogenese van EHV1 bij paarden bij

experimentele inoculaties en veldstudies”. Naast het verrichten van onderzoek, was

Annick ook verantwoordelijk voor het verder uitbreiden van de diagnostiek van virale

ziekten bij het paard en het routinematig uitvoeren van deze diagnostiek. Tevens was ze

betrokken bij het praktische onderricht van laatstejaars studenten in de optierichting

paard.

Annick Gryspeerdt is auteur en mede-auteur van verschillende wetenschappelijke

publicaties in internationale tijdschriften. Ze nam eveneens deel aan meerdere nationale

en internationale congressen en was verantwoordelijk voor het geven van verschillende

lezingen, waaronder een opleidingsnamiddag voor dierenartsen en sensibilisatie van

paardeneigenaren.

Bibliography

155

BIBLIOGRAPHY

Bibliography

157

BIBLIOGRAPHY

Publications in international journals

Gryspeerdt, A. C., Vandekerckhove, A. P., Garré, B., Barbé, F., Van de Walle, G. R. &

Nauwynck, H. J. (2009). Differences in replication kinetics and cell tropism

between neurovirulent and non-neurovirulent EHV1 strains during the acute phase

of infection in horses. Vet Microbiol 142, 242-253.

Gryspeerdt, A. C., Chiers, K., Govaere, J., Vercauteren, G., Ducatelle, R., Van de Walle,

G. R. & Nauwynck, H. J. (2009). Neonatal foal death due to infection with equine

arteritis virus in Belgium. Vlaams Diergen Tijds 78, 189-193.

Gryspeerdt, A. C., Vandekerckhove, A. P., Bannazadeh Baghi, H., Osterrieder, N., Van

de Walle, G. R. & Nauwynck H. J. (2011). Expression of late proteins is hampered

in infected nasal mucosal leukocytes but not in epithelial cells during early

pathogenesis of equine herpesvirus type 1 (EHV1) infection. Vet J (accepted)

Gryspeerdt, A. C., Vandekerckhove A. P., Van Doorsselaere, J., Van de Walle, G. R. &

Nauwynck, H. J. (2011). Description of an unusually large outbreak of nervous

system disorders caused by equine herpesvirus 1 (EHV1) in 2009 in Belgium.

Vlaams Diergen Tijds 80, 139-145.

Van der Meulen, K. M., Gryspeerdt, A. C., Vandekerckhove, A. P., Garré, B. A. &

Nauwynck, H. J. (2008). The protective properties of vaccination against equine

herpesvirus 1-induced viremia, abortion and nervous system disorders. Vlaams

Diergen Tijds 76, 186-194.

Garré, B. A., Gryspeerdt, A. C., Croubels, S., De Backer, P. & Nauwynck, H. J. (2009).

Evaluation of orally administered valacyclovir in experimentally EHV1-infected

ponies. Vet Microbiol 135, 214-221.

Bibliography

158

Garré, B., Shebany, K., Gryspeerdt, A. C., Baert, K., van der Meulen, K., Nauwynck, H.

J., Deprez, P., De Backer, P. & Croubels, S. (2007). Pharmacokinetics of

acyclovir after intravenous infusion of acyclovir and after oral administration of

acyclovir and its prodrug valacyclovir in healthy adult horses. Antimicrob Agents

Chemother 51, 4308-4314.

Vandekerckhove, A. P., Glorieux, S., Gryspeerdt, A. C., Steukers, L., Duchateau, L.,

Osterrieder, N., Van de Walle, G. R. & Nauwynck, H. J. (2010). Replication

kinetics of neurovirulent versus non-neurovirulent equine herpesvirus type 1

strains in equine nasal mucosal explants. J Gen Virol 91, 2019-2028.

Vandekerckhove, A. P., Glorieux, S., Gryspeerdt, A. C., Steukers, L., Van Doorsselaere

J., Osterrieder N., Van de Walle, G. R. & Nauwynck, H. J (2011). Equine

alphaherpesviruses (EHV) differ in their efficiency to infect mononuclear cells

during early steps of infection in nasal mucosa explants. Vet Mic (in press).

Vandekerckhove, A. P., Glorieux, S., Van Den Broeck, W. V., Gryspeerdt, A. C., van

der Meulen, K. M. & Nauwynck, H. J. (2009). In vitro culture of equine

respiratory mucosa explants. Vet J. 181, 280-287.

Oral presentations

Vandekerckhove, A. P., Van den Broeck, W., van der Meulen K. M., Glorieux, S.,

Gryspeerdt, A. C. & Nauwynck, H. J. (2007). Replication of equine herpesvirus

1 in equine respiratory mucosa explants. 32nd International Herpesvirus

Workshop, Asheville, North Carolina, VS.

Gryspeerdt, A. C., Vandekerckhove, A. P. & Nauwynck, H. J. (2008). Replication of

equine herpesvirus 1 in tissues of the upper respiratory tract during the acute phase

of infection in horses. 33rd International Herpesvirus Workshop, Estoril, Portugal.

Gryspeerdt, A. C., Vandekerckhove, A. P., Van de Walle, G. R. & Nauwynck H. J.

(2009). Differences between the replication of neurovirulent en non-neurovirulent

Bibliography

159

EHV1 strains in the upper respiratory tract in vivo. 34th

International Herpesvirus

Workshop, Ithaca, New York, VS.

Vandekerckhove, A. P., Gryspeerdt, A. C., Glorieux, S., Van de Walle, G. R. &

Nauwynck, H. J. (2009). Replication of non-neurovirulent versus neurovirulent

equine herpesvirus type 1 strains: a study in equine respiratory mucosa explants.

34th

International Herpesvirus Workshop, Ithaca, New York, VS.

Gryspeerdt, A. C., Vandekerckhove, A. P., Van de Walle, G. R. & Nauwynck H. J.

(2009). Differences between the replication of neurovirulent en non-neurovirulent

EHV1 strains in the upper respiratory tract in vivo. Summer Course on Infection

and Immunity, 29th

– 30th

September, Ghent, Belgium.

Vandekerckhove, A. P., Gryspeerdt, A. C., Glorieux, S., Van de Walle, G. R. &

Nauwynck, H. J. (2009). Replication of non-neurovirulent versus neurovirulent

equine herpesvirus type 1 strains: a study in equine respiratory mucosa explants.

Symposium of the Belgian Society for Microbiology, 11th

December, Brussels,

Belgium.

Gryspeerdt, A. C. & Nauwynck, H. J. (2009). Prevalence and occurrence of clinical

EHV1 in Flanders, Belgium. Equine Influenza and Herpesvirus – Current Disease

Situation, 16th

October, Berlin, Germany.

Gryspeerdt, A. C. (2010). Voorkomen, behandeling en preventie van rhinopneumonie in

Vlaanderen. Studienamiddag rond virale ziekten bij het paard, Vlaams

Paardenloket, 27th

March, Nazareth, Belgium.

Gryspeerdt, A. C. & Nuytten, J. (2010). Welke lessen kunnen getrokken worden uit de

recente uitbraken van virale ziekten bij het paard? Belgian Equine Practitioners

Society (BEPS), 27ste

studienamiddag, 13th

November, Leuven, Belgium.

Bibliography

160

Gryspeerdt, A. C. (2010). Voorkomen, behandeling en preventie van rhinopneumonie in

Vlaanderen. Studienamiddag rond virale ziekten bij het paard, Vlaams

Paardenloket, 4th

December, Lummen, Belgium.

Gryspeerdt, A. C. (2011). Rhinopneumonie: waarheden en onwaarheden over recente

uitbraken. Baanpraktijk de Morette, 3th

March, Essene, Belgium.

Abstracts

Vandekerckhove, A. P., Van den Broeck, W., van der Meulen K. M., Glorieux, S.,

Gryspeerdt, A. C. & Nauwynck, H. J. (2007). Replication of equine herpesvirus

1 in equine respiratory mucosa explants. Symposium of the Belgian Society for

Microbiology, 23rd

November, Brussels, Belgium.

Gryspeerdt, A. C., Vandekerckhove, A. P. & Nauwynck, H. J. (2008). Replication of

equine herpesvirus 1 in tissues of the upper respiratory tract during the acute phase

of infection in horses. Symposium of the Belgian Society for Microbiology, 12th

December, Brussels, Belgium. BSM award for excellent poster

Gryspeerdt, A. C.*, Vandekerckhove, A. P.*, Van de Walle, G. R. & Nauwynck, H. J.

(2009). In vitro respiratory mucosa explants are useful tools to study respiratory

tract diseases in horses. 3d ESVV Veterinary Herpesvirus Symposium, Insel

Riems, Germany. * equally contributed

Dankwoord

161

DANKWOORD

Dankwoord

163

DANKWOORD

Na ettelijke jaren van hard werken, bloed, zweet, tranen, maar ook veel memorabele

momenten, ligt het hier dan eindelijk: mijn doctoraat. Het is moeilijk te vatten wat een

grote waarde dit kleine boekje heeft. Na al die jaren verdienen heel wat mensen een

woord van dank en dat in het ongetwijfeld meest gelezen hoofdstuk van elk doctoraat…

Eerst en vooral wil ik hier graag mijn promotoren danken. Hans, jou wil ik graag

bedanken om me de kans te geven in jouw laboratorium te werken. Ik denk dat er weinig

werkplaatsen zijn waar de sfeer tussen de werknemers zo goed zit. Ik denk ook dat er

maar weinig manieren zijn om jou te beschrijven. Zelden heb ik zo een gedreven en

hyper-enthousiaste wetenschapper meegemaakt. Je bent echt een uniek persoon! Ik hoop

dat het labo virologie nog lang zo mag groeien en bloeien. Karen, jij was de persoon die

me geïntroduceerd heeft in de wondere wereld van de rhinopneumonie. Met jouw hulp is

de IWT-beurs binnengehaald en kon mijn onderzoek echt starten. Je hebt het werk niet

kunnen afmaken, maar je hebt het startschot gegeven zodat ik op eigen benen kon staan.

Mijn schrijfsels zijn nog steeds niet top, maar ik zal me altijd de verbeterde versies met

kleurcodes herinneren die ik in het begin van jou terugkreeg. Gerlinde, jou ben ik heel

veel dankbaarheid verschuldigd. We werden gewaarschuwd dat er een felle madam in

aantocht was, die de tucht in onze paardengroep moest herstellen. Die felle madam bleek

echter een heel aangename en gedreven onderzoeker te zijn, die er als eerste in slaagde

om een artikel van mij gepubliceerd te krijgen. Honderden schrijfsels heb je verbeterd en

hoewel ik nog steeds geen bestseller auteur kan genoemd worden, is er toch enige

verbetering merkbaar. Verder stond je altijd voor me klaar toen het weer eens moeilijk

ging met mijn doctoraat. Dankzij jouw motivering ligt dit boekje hier vandaag! En ik

denk dat ik zeker mag zeggen dat je goed bezig bent, Professor Van de Walle! Ik ben er

zeker van dat je een prachtig labo zal uitbouwen! Herman, ik wil je graag bedanken voor

je geloof in mijn onderzoek en praktijkgerichte aanpak. Bedankt voor je luisterend oor

tijdens moeilijke momenten en voor de aangename congressen wanneer je van de partij

was!

Ook de overige leden van de begeleidings- en examencommissie zou ik hartelijk

willen bedanken voor de tijd die ze vrijgemaakt hebben en voor hun waardevolle en

Dankwoord

164

opbouwende kritieken: Prof. dr. P. Deprez, Prof. dr. J. Kydd, dr. K. van Maanen, mevr. B.

Caij, dr. K. van der Meulen, dr. J. Nuytten en dr. T. Picavet.

Het agentschap voor Innovatie door Wetenschap en Technologie wil ik bedanken voor

de financiële steun die het mogelijk heeft gemaakt om dit doctoraat tot een goed einde te

brengen.

Eerst wil ik even de moeite nemen om de fantastische laboranten en technische

krachten van het labo te bedanken. Zij zijn het hart en ziel van de dienst en zorgen er

dagelijks voor dat alles op wieltjes loopt, wat met al dat volk geen evidente taak kan

genoemd worden. Chris, ondertussen ben je op pensioen, maar ik zal al het werk dat je

voor mij gedaan hebt nooit vergeten. Je was de onbetwiste kampioen wat betreft het

isoleren van paardenleukocyten! Bovendien was je een zeer aangename persoon en

hebben we samen ook veel plezier gehad. Ik hoop dat je geniet van je pensioen!

Vervolgens hebben we Lieve. Ondanks het feit dat we nooit echt rechtstreeks hebben

samengewerkt, kan ik me nog goed herinneren dat je me in het begin ogenblikkelijk hebt

duidelijk gemaakt dat het een slecht idee was om mijn voorraden slechts op de dag van

mijn experiment te checken. Geregeld ben ik met rode kaken tot bij jou gekomen om te

zeggen dat mijn medium weer eens op was of beschimmeld was. Ik heb dankzij je strenge

aanpak echter zeer snel geleerd om me wat beter te organiseren, waarvoor dank! Chantal,

jou moet ik bedanken voor het opgroeien van ontelbare virusstocks, want zonder virus

geen experimenten! Vervolgens hebben we Carine. Wij hebben heel veel samengewerkt

en het is altijd met het grootste plezier geweest. Je bent een zeer aangename en

hardwerkende vrouw en niets was ooit te veel voor jou. Ik kan zelfs al niet meer tellen

hoeveel weefsels je vakkundig getitreerd hebt. Het enthousiasme waarmee we de

zoveelste diagnose verwerkten was legendarisch! En het plezier dat we hadden als we

weer iets geïsoleerd hadden na veel problemen maakte het de moeite waard ☺. Ik hoop

dat de diagnostiek even goed blijft draaien en ik hoop dat we daar in de toekomst nog veel

contact over hebben. Nele en Melanie hebben me ook geregeld uit de brand geholpen.

Nele door het biotinyleren van ontelbare antistoffen en Melanie met het ingeven van

diagnose uitslagen en het inspringen als het praktische werk mij weer eens boven het

hoofd groeide. Dries heeft de ondankbare taak gekregen om me af en toe moleculair te

begeleiden. Helaas voor hem ben ik er zeker van dat ik op dat vlak vervloekt ben.

Bedankt voor de moeite die je er toch telkens weer instak ondanks de hardnekkige

Dankwoord

165

tegenvallende resultaten. Jan Van Doorsselaere wil ik bedanken voor alle sequeneringen

die hij voor mij heeft uitgevoerd. Verder hebben we de technische krachten nog, waarvan

vooral Geert een eervolle vermelding verdient. Je hebt me op hele vroege uren geholpen

met de paardjes en zulke goede hulp heb ik nooit meer gehad. Ik hoop dat je gelukkig

bent op je nieuwe werk! Fernand is ondertussen ook al aan zijn pensioen begonnen, maar

heeft ook dikwijls zijn steentje bijgedragen bij diverse experimenten, waarvoor mijn

dank! Bart en Zeger, jullie hebben de moeilijke taak gekregen dit duo op te volgen. Ik

hoop dat jullie je draai vinden en genieten van jullie job! Marijke mag ook zeker niet

vergeten worden, de persoon die als moeilijke taak heeft om het labo netjes te houden.

Bedankt voor het onderhouden van mijn bureautje en de vele leuke gesprekken! Mieke,

Ann, Gert en Louise, bedankt voor alle praktische dingen die jullie dag na dag moeten

zien te regelen. Jullie zijn echt de duizendpoten van het labo! Dirk, bedankt voor het

oplossen van vele computergerelateerde problemen, maar toch ben ik nog steeds heel erg

blij met mijn PC’tje ☺. Mocht ik mij in de verre toekomst ooit eens een Mac aanschaffen,

dan zal ik eens aan jou terug denken!

En dan hebben we de collega’s. Er zijn er in die 6 jaar zoveel gekomen en gegaan dat

ik echt hoop dat ik niemand vergeten ben!

Ik ben mijn tijd op het labo gestart in de paardenbureau. Annelies, jij was mijn collega

van het eerste uur, eerst als laatstejaars, vervolgens als Phd student. Ik herinner me vooral

onze vreugdedans in het bureau van Hans toen we vernamen dat we beide onze IWT-

beurs gehaald hadden. Verder heb je me ontelbare keren geholpen met

paardenexperimenten. Ik wil je bedanken voor alle leuke jaren. Ik hoop dat je als “the last

one standing” de eer van de paardengroep hoog kunt houden en dat je het EHV onderzoek

nieuw leven kan inblazen. Ik wens je een prachtig leven toe met je allerliefste Lieven en

hoop dat jullie heel gelukkig worden samen! Filip had de ondankbare taak om als enige

man ons bureautje te bevolken. Ik moet zeggen, ik ken niemand waarbij de spreuk “stille

waters, diepe gronden” zo goed van toepassing was! Je kon je mannetje heel goed staan

tussen al dat vrouwengeweld! Zelfs de Flair werd op de duur opgenomen in je

literatuurlijst ☺. Hartelijk dank voor alle gezellige etentjes, gesprekjes en je hulp met de

paardjes. Ik hoop dat we elkaar in de toekomst nog geregeld eens zien en ik wens je veel

geluk toe met de bouw van je droomhuis!

Dankwoord

166

Na vier jaar werd het tijd voor een nieuw onderkomen, wat werd gevonden in de

beroemde “eerste bureau”. Sarah G., als ancien van deze bureau heb je de nieuwelingen

hartelijk ontvangen! Ik wil je bedanken voor de talloze gezellige etentjes en het

onvergetelijke congres dat we samen in Portugal hebben mogen meemaken. Ik hoop dat

alles vlot loopt met je onderzoek en dat je heel gelukkig mag worden met je toekomstige

man! Amy, you arrived a bit later, but you are really the sweetest girl in the office! I am

really glad that your husband finally arrived in Belgium and I hope you will enjoy your

time here in Belgium. I want to thank you for all the nice Chinese gifts I received from

you and I wish you all the best with your research! Hossein, you got the very difficult task

to find our well-hidden equine dendritic cells. I hope you will find them eventually! Keep

practising with your bike and then you will avoid becoming as fat as I once was ☺.

Sabrina! You are the most energetic person I ever met! It is really a delight to have such

person in the office! With you, nothing will ever be boring! I hope you will succeed in

your Phd, you really deserve it after all the work you’ve put in to it. Enjoy your beautiful

daughter and always remain your cheerful self! Mieke, jij bent stiekem binnengeglipt toen

ik op zwangerschapsverlof was, maar ik heb de indruk dat je je plaatsje al goed veroverd

hebt! Ik wens je veel geluk toe in je onderzoek! Lennert, tja, er zijn zoveel woorden

waarmee jij beschreven kunt worden, maar saai is er in elk geval niet één van! Je brengt

altijd plezier in het bureau en bent altijd in voor een grapje. Ik ben blij dat er nog een

echte veearts in het labo aanwezig is! Veel succes in Amerika en ik ben zeker dat jij er op

professioneel vlak wel zal geraken!

Verder heb ik nog een heel tijdelijk onderkomen gevonden in mijn oude bureau

tijdens het “pimpen” van ons nieuw bureau (kunnen jullie nog volgen?), waar ik hartelijk

werd ontvangen door Sjouke en Annebel. Bedankt voor alle leuke en serieuze gesprekken

en Sjouke, bedankt voor de hulp met het streepje aan de bovenkant van mijn pagina’s! Je

bent een ferme madam en je zal er zeker komen in het leven!

Marc, jij verdient ook zeker een eervolle vermelding! Met jou is er altijd tijd voor

plezier en dat al sinds ons eerste jaar in Gent. Ik vond het super dat je onze ploeg op het

labo kwam vergezellen. De sfeer op het labo is er enkel maar op vooruit gegaan! Bedankt

voor alle amusante gesprekken, maar ook voor de steun die ik altijd van jou heb gekregen.

Werp je maar op het schrijven en haal dat doctoraat binnen! Merijn en Wander, jullie wil

ik bedanken voor de talloze vragen die jullie beantwoord hebben naar aanleiding van mijn

moleculaire problemen! Niettegenstaande het feit dat mijn moleculaire vervloeking overal

Dankwoord

167

stokken in de wielen stak, heb ik wel veel nuttige tips gekregen! Wander, het was fijn om

een compagnon te hebben bij de finishing touch van onze doctoraten. Ik hoop dat je in de

toekomst iets meer relax je deadlines zal halen!

Ik denk dat ons labo ondertussen wel gekend is als één van de grootste van de

faculteit diergeneeskunde. Volgens mij is het ook één van die uitzonderlijke plekken waar

de sfeer tussen de werknemers echt super is. Daarom wil ik graag iedereen bedanken die

nog niet aan bod is gekomen, zowel onze inheemse (Matthias, Tim, Veerle, Ytse, Hannah,

Annelike, Dominique, Ben, Lowiese, Sebastiaan, Hanne, Inge, Karen, Miet, Céline,

Thary en Nina) als uitheemse mensen (Sabine, Ilias, Eva, Dipu, Joao, Tù, Uladzimir,

Xiaoyun, Jun, Lang, Liping, Wenfeng, Yu, Zhongfang and Maria) en natuurlijk iedereen

die zijn/haar carrière op het labo nog maar net gestart is! Elk van jullie draagt zijn steentje

bij tot de tweede thuis die het labo voor veel mensen geworden is.

Nog een woordje voor de collega’s die het labo reeds verlaten hebben, maar die zeker

ook een plaatsje in dit dankwoord verdienen. Barbara, ik weet niet zo goed wat ik hier

moet neerschrijven. Ik herinner me nog de eerste keer dat je me moest wegwijs maken in

het labo en je me heel vreemd aankeek omdat je me maar “raar en luid” vond. Maar heel

snel hebben we een gezamenlijke passie ontdekt, die ons tot heel goede vriendinnen heeft

gemaakt. Je hebt me door de moeilijke periode in het labo geholpen en hebt me zowel

privé als werkgerelateerd heel veel plezier bezorgd. We hebben van in de vroege tot de

late uurtjes samen gewerkt en om het met je eigen woorden te zeggen “bedankt voor heel

veel en nog zoveel meer”… Hopelijk vinden jij en Eric jullie droomhuis en kunnen jullie

een prachtig leven uitbouwen! Sarah C., jij was ook één van de meest opmerkelijke

personen in het labo. Ik vind het super dat we nog af en toe contact hebben en hoop dat

het je op alle vlakken voor de wind blijft gaan! Leslie, ik heb grote bewondering voor het

feit dat je zo goed weet wat je wil in het leven en daar ook helemaal voor gaat. Veel

succes met je nieuwe job en bedankt voor alle leuke gesprekken!

Hoewel het minder gebruikelijk is, wil ik ook alle paarden en pony’s bedanken die

een bijdrage geleverd hebben aan dit doctoraat. Ze hebben me veel plezier bezorgd en een

aantal van hen hebben ondertussen een super thuis gevonden! Een speciale vermelding

wilde ik graag geven aan Cointreau, het veulen dat ons bloed, zweet en tranen gekost

heeft omwille van zijn speelse karakter. Ik denk dat de jacht op Cointreau het meest

memorabele moment was in die 6 jaar. Ik vergeet nooit de hulp van Prof. dr. Gasthuys,

die in volledige jacht outfit, verrekijker en laarzen inclusief, er in slaagde onze ontsnapte

Dankwoord

168

dwarsligger na twee dolle dagen van zijn touw te verlossen… Helaas bleek onze

ontsnapkampioen twee weken later nog steeds niet bereid om in zijn lekker warme

stalletje te komen staan, waardoor er weer een paar dolle jachturen moesten gespendeerd

worden, deze keer met dank aan de sterke mannen van verloskunde. Ondertussen is

Cointreau een flinke vijfjarige geworden en doet hij zijn uiterste best voor zijn nieuwe

baasje! Ik hoop dat hij een lang en gelukkig leven tegemoet gaat!

In de loop van mijn doctoraatsjaren heb ik ook enorm veel steun mogen ontvangen

van mijn collega dierenartsen in de praktijk. Ik wil hen graag bedanken voor alle hulp en

suggesties die ze mij geboden hebben. Ze hielden me met mijn voeten op de grond en

zorgden ervoor dat ik niet vergat waarom virologisch onderzoek voor paarden zo

belangrijk is. Hierbij moet zeker een speciale plaats gegeven worden aan mijn ”mentor”

Jef. Het is door jou dat ik 6 jaar geleden de stap genomen heb om in het onderzoek te

beginnen. Jij was het spreekwoordelijke duwtje in de rug dat ik toen nodig had om er

helemaal in te vliegen. Gedurende al die jaren heb je echter mijn interesse in de praktijk

levendig gehouden en heb je er op gehamerd dat de virologie voor paarden dringend

verder moest onderzocht worden. Ook ons eigen onderzoekje met de honderden swabs en

bloedjes mag daarbij zeker niet vergeten worden. Evenals het ongeduld waarmee je belde

om de uitslag van de zoveelste diagnose. Hierbij wil ik je nog eens uitgebreid danken

voor alles wat je me bijgebracht hebt aan ervaring en “West-Vlaamse levenswijsheden”

en voor de vele uren ontspanning en plezier als het onderzoek mij weer eens te zwaar

werd. Ik hoop dat we in de toekomst nog evenveel praktijkplezier mogen meemaken en

dat er af en toe nog eens een glas champagne geleegd mag worden ☺.

Ook buiten het labo zijn er zoveel vrienden die me geholpen en gesteund hebben.

Lieve Leen, je woont misschien wel ver weg, maar het contact is er na meer dan tien jaar

nog steeds en onze schaarse ontspannende uitstapjes en de deugddoende gesprekken zal

ik nooit vergeten! Julie, in al die jaren zijn er honderden en honderden mails over en weer

gevlogen! Super dat we nog altijd zo een leuk contact hebben en ik hoop dat dat nog jaren

mag blijven duren. Bedankt voor al je steun en begrip! Je bent een super meid en

supermama! Sophie, jij bent er maar later bijgekomen, maar op die korte tijd ben je echt

een goede vriendin geworden. Bedankt voor je altijd luisterende oor en voor alle leuke

etentjes die er al geweest zijn. Veel geluk met je prachtige gezinnetje! En last, but not

least: de familie Demeyer en hun bonte beestenboel. Ik kan mij al lang niet meer

Dankwoord

169

herinneren wanneer ik bij jullie ben binnen gewaaid. Ik weet enkel nog dat het begon met

een brommertje zonder benzine en 100 frank. Zoals jullie het zelf zeggen ben ik bij jullie

ontvangen als vijfde dochter en ik denk dat jullie maar al te goed beseffen wat jullie

daarmee in huis gehaald hebben ☺. Jullie staan stuk voor stuk altijd voor me klaar en

hebben gezorgd voor jaren ontspanning, plezier en serieuze momenten. Dank vanuit de

grond van mijn hart! Miss Domino moet ik nog danken voor de hersenschudding die ze

me bezorgde vlak voor ik mijn project moest indienen… Gelukkig heeft haar zoon dat

ruimschoots goed gemaakt met de ontspannen momenten gedurende de laatste loodjes

van mijn doctoraat!

Wie hier zeker niet mogen ontbreken zijn mijn allerliefste ouders. Zonder jullie steun,

of het nu op financieel vlak was, of door jullie rotsvaste geloof dat ik er wel zou komen in

mijn leven, zou ik nooit staan waar ik nu sta. Jullie hebben me altijd gemotiveerd om

datgene te doen wat ik wilde en hebben ervoor gezorgd dat ik mijn grote droom als

veearts kon waarmaken. Ik ben jullie oneindig dankbaar hiervoor en besef dat er velen

zijn die die steun moeten missen. Mijn zus Charlotte en broertjes Filip en Frederic moet

ik natuurlijk ook danken, want ze hebben heel wat geklaag mogen aanhoren! Ook de

aanhangsels zijn hier vandaag aanwezig en wil ik danken omdat ze onze familie nog

groter en gezelliger gemaakt hebben!

Liefste Jan-Bart, jij hebt het gedurende al die jaren het zwaarst te verduren gekregen.

Je moest het regelmatig ontgelden als de stress weer eens te hoog opflakkerde. Gelukkig

kan je wel tegen een stootje. Ons leven is op korte tijd grondig omgegooid met ons eigen

huisje op een ver afgelegen plaats en onze prachtige, lieve dochter. Nu komt er weer een

verandering met een nieuwe werkuitdaging bij. Ik wil je danken voor al de steun die ik al

gekregen heb en die ik nog steeds krijg, want jij begrijpt wat een carrière vrouw is en

vooral wat een gestresseerde carrièrevrouw betekent! Lieve Aline, sinds jij er bent, weet

ik wat belangrijk is in het leven, weet ik waarom ik het doe en kan ik alles wat beter

relativeren. Met je stralende glimlach kon je me altijd weer doen smelten. Ik hoop dat je

mag opgroeien tot een gelukkige vrouw en dat je evenveel passies mag kennen als je

mama en papa.

Dit dankwoord is zeer uitgebreid geworden en dat maakt me blij, want het betekent

dat er veel mensen zijn die het de moeite waard vonden om me te steunen. Ik hoop vanuit

Dankwoord

170

de grond van mijn hart dat ik niemand ben vergeten, daarom nogmaals: dank aan

iedereen, dit is jullie doctoraat!