+ All Categories
Home > Documents > Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for...

Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for...

Date post: 23-May-2020
Category:
Upload: others
View: 5 times
Download: 0 times
Share this document with a friend
172
1 An Analysis of Bovine immunodeficiency virus and Jembrana disease virus Infections in Bos javanicus Tegan Josephine McNab BSc (Hons) This thesis is presented for the degree of Doctor of Philosophy of Murdoch University. April 2010
Transcript
Page 1: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

1

An Analysis of Bovine immunodeficiency virus and Jembrana

disease virus Infections in Bos javanicus

Tegan Josephine McNab

BSc (Hons)

This thesis is presented for the degree of Doctor of Philosophy

of Murdoch University.

April 2010

Page 2: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

i

Abstract

Two closely related bovine lentiviruses have been described, Jembrana disease virus

(JDV) and Bovine immunodeficiency virus (BIV), that produce very different clinical

manifestations in infected cattle. JDV causes an acute disease with a case fatality rate

of about 21% in Bos javanicus (Bali cattle) and is endemic in the cattle population of

parts of Indonesia. BIV produces a subclinical infection in Bos taurus and buffalo

and serological evidence has shown that this virus has a worldwide distribution,

possibly including Indonesia.

Attempts were made to confirm a previous report that BIV was present in the

B. javanicus population in Indonesia. BIV proviral DNA was not detected in any of

the animals although JDV proviral DNA was detected in 12 of 171 animals, only one

of which was seropositive.

To define the kinetics of BIV infection in B. javanicus and determine the optimal

time for sampling to detect BIV infection, 13 animals were experimentally infected

with the R29 strain of BIV. No clinical effects were detected but proviral DNA was

detected from 4-60 days post-infection (dpi) with peak titres 20 days dpi, and a

transient viraemia from 4 to 14 dpi. An antibody response to TM was detected 12 dpi

but an anti-capsid (CA) antibody response was detected in one animal only and not

until 34 dpi. The results indicated that detection of BIV in infected Bali cattle using

PCR would have a greater chance of success soon after infection and prior to the

onset of a CA antibody response.

To determine the effect of BIV infection on subsequent JDV infection in

B. javanicus, 15 cattle were infected with BIV-R29 and 9 of these were subsequently

infected 42 days later with JDV. The response to BIV was typical of that observed

Page 3: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

ii

previously but BIV infection did not markedly modify the response to subsequent

infection with JDV. In response to JDV infection, all cattle previously infected with

BIV still developed an acute disease process typical of Jembrana disease. The results

suggested that despite the close genetic and antigenic relationship between BIV and

JDV, BIV infection does not confer protection against subsequent JDV infection.

The close antigenic relationship between BIV and JDV is a problem in the

development of specific serological tests and immunosurveillance of JDV infection.

To develop reagents capable of differentiating between antibody to BIV and JDV

infections, peptide mapping was used to define linear B cell epitopes on the matrix

(MA), CA and surface unit (SU) proteins of JDV. Short overlapping peptides that

spanned these regions were synthesised and used in an ELISA format to screen their

reactivity with a panel of bovine sera from animals experimentally infected with

JDVTab87, JDVPul01 or BIV-R29. Peptides representing potential immunoreactive

epitopes were identified that appeared to offer promise in the development of JDV-

specific serological tests and need to be tested further with a panel of sera taken from

naturally infected cattle.

Page 4: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

iii

Declaration

I declare that this thesis is my own account of my research and contains as its main

content work which has not previously been submitted for a degree at any tertiary

education institution.

....................................

Tegan Josephine McNab.

Page 5: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

iv

Contents

ABSTRACT........................................................................................................................................... I

DECLARATION................................................................................................................................III

ACKNOWLEDGEMENTS............................................................................................................... VI

ABBREVIATIONS ...........................................................................................................................VII

PUBLICATION AND INTERNATIONAL CONFERENCE PRESENTATIONS...................... IX

Publications arising from this thesis ............................................................................................ix Manuscripts submitted for publication .........................................................................................ix Oral presentations ........................................................................................................................ix Poster presentations .....................................................................................................................ix

CHAPTER 1: INTRODUCTION ....................................................................................................... 1

CHAPTER 2: LITERATURE REVIEW ........................................................................................... 4

Physiochemical characteristics of retroviruses ............................................................................ 4 Retrovirus taxonomy..................................................................................................................... 4 General structure of lentiviruses .................................................................................................. 5 The lentivirus genome................................................................................................................... 6 Lentivirus replication ................................................................................................................. 10 Common characteristics of lentiviruses...................................................................................... 14

PRIMATE LENTIVIRUSES................................................................................................................... 15 Human immunodeficiency virus.................................................................................................. 15 Simian immunodeficiency viruses............................................................................................... 17

NON-PRIMATE LENTIVIRUSES........................................................................................................... 19 Feline immunodeficiency virus ................................................................................................... 19 FIV in non-domestic feline species ............................................................................................. 21 Equine infectious anaemia virus................................................................................................. 22

SMALL RUMINANT LENTIVIRUSES .................................................................................................... 23 Visna maedi virus ....................................................................................................................... 24 Caprine arthritis encephalitis virus............................................................................................ 25

BOVINE LENTIVIRUSES..................................................................................................................... 25 Bovine immunodeficiency virus .................................................................................................. 27 Jembrana disease virus............................................................................................................... 29

IMMUNE RESPONSE TO INFECTION WITH A LENTIVIRUS.................................................................... 35 Humoral immune response to infection with bovine lentiviruses ............................................... 38

ASSAYS FOR THE DETECTION OF LENTIVIRUS INFECTIONS IN RUMINANTS ........................................ 40 Small ruminant lentiviruses ........................................................................................................ 40 Large ruminant lentiviruses........................................................................................................ 41 Difficulties with serological testing ............................................................................................ 42 Techniques to identify epitopes and distinguish between viral infections .................................. 43

LENTIVIRUS SUPERINFECTION.......................................................................................................... 45 Benefits of superinfection - superinfection resistance ................................................................ 45 Adverse consequences of superinfection..................................................................................... 47

CHAPTER 3: ATTEMPTS TO DETECT BOVINE IMMUNODEFICIENCY VIRUS INFECTION IN BALI CATTLE IN INDONESIA WITH A PCR-BASED ASSAY................... 49

SUMMARY ........................................................................................................................................ 49 INTRODUCTION................................................................................................................................. 50 MATERIALS AND METHODS.............................................................................................................. 51 RESULTS.......................................................................................................................................... 57 DISCUSSION..................................................................................................................................... 62

Page 6: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

v

CHAPTER 4: BOVINE IMMUNODEFICIENCY VIRUS PRODUCES A TRANSIENT VIRAEMIC PHASE SOON AFTER INFECTION IN BOS JAVANICUS.................................. 66

SUMMARY ........................................................................................................................................ 66 INTRODUCTION................................................................................................................................. 67 MATERIALS AND METHODS.............................................................................................................. 68 RESULTS.......................................................................................................................................... 73 DISCUSSION..................................................................................................................................... 80

CHAPTER 5: BOVINE IMMUNODEFICIENCY VIRUS INFECTION ALTERS THE DYNAMICS OF SUBSEQUENT JEMBRANA DISEASE VIRUS INFECTION......................... 83

SUMMARY ........................................................................................................................................ 83 INTRODUCTION................................................................................................................................. 84 MATERIALS AND METHODS.............................................................................................................. 85 RESULTS.......................................................................................................................................... 89 DISCUSSION................................................................................................................................... 100

CHAPTER 6: HUMORAL IMMUNE RESPONSES TO JEMBRANA DISEASE VIRUS DETECTED USING OVERLAPPING SYNTHETIC PEPTIDES SPANNING THE MA, CA AND SU REGIONS OF JDV .......................................................................................................... 105

SUMMARY ...................................................................................................................................... 105 INTRODUCTION............................................................................................................................... 106 MATERIALS AND METHODS............................................................................................................ 107 RESULTS........................................................................................................................................ 111 DISCUSSION................................................................................................................................... 119

CHAPTER 7: GENERAL DISCUSSION...................................................................................... 127

REFERENCES................................................................................................................................. 135

Page 7: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

vi

Acknowledgements

First and foremost, I would like to thank my two supervisors, Dr. Moira Desport and

Professor Graham Wilcox, for their help throughout my PhD. I really appreciate all

the help they’ve given me. In particular, Moira for your input in designing my PhD,

for helping me in the lab and for all the wisdom you’ve passed on to me. To Graham,

for your help in editing my thesis and getting papers ready for submission. I hope

you catch many fish in your retirement!

Thankyou to Robert Dobson for your help with all things statistical, I am sure I still

owe you a few more cakes. Thanks must also be extended to other staff in the

Veterinary and Biomedical Sciences building for your help over the past few years.

Thankyou to the Australian Centre for International Agricultural Research for

providing the funding necessary to undertake this work and to the Australian

Government for providing my scholarship.

Thankyou to the members of the laboratory in Denpasar, particularly Nining and

Putu, for their help in running our animal trials and for the collection of samples.

Without this, it would not have been possible to complete my thesis.

A big thankyou to past and present members of our office: Will, Mark, Andrew,

Linda, Josh, Masa, Yudhi and honouree member Jill A. Thankyou so much for all

your help and good times shared around cake.

To all my friends, Laura G, Jacqui, Gael, Genevieve, Sarah, Nikki, Bry, Celia,

Laura H, Kendle, Michelle, and my entire family, thanks for all the good times. To

everyone at kickboxing, thankyou for helping to keep the stress levels low by letting

me punch and kick you on a regular basis.

Finally, a big thankyou to my parents, Marg and Ross, for being fantastic and so

supportive all the time.

To Bong for just being you.

Page 8: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

vii

Abbreviations

AIDS Acquired immunodeficiency syndrome AGID Agar gel immunodiffusion APOBEC Apolipoprotein B mRNA-editing enzyme-catalytic polypeptide-like AUC Area under the curve BFL Bovine foetal lung BIV Bovine immunodeficiency virus BVDV Bovine viral diarrhoea virus CA Capsid CAEV Caprine arthritis encephalitis virus CE Cell equivalents CNS Central nervous system CCR5 C-C (beta) chemokine receptor 5 CD4 Cluster of differentiation 4 CD8 Cluster of differentiation 8 CTL Cytotoxic T-lymphocyte CXCR4 C-X-C (alpha) chemokine receptor 4 DNA Deoxyribonucleic acid dNTP Deoxynucleotide Triphosphate DMSO Dimethyl sulfoxide dpi Days post-infection ELISA Enzyme linked immunosorbent assay EIAV Equine infectious anaemia virus EDTA Ethylenediamine tetra-acetic acid FIV Feline immunodeficiency virus GAPDH Glyceraldehyde 3-phosphate dehydrogenase HRP Horse radish peroxidase HIV Human immunodeficiency virus HTLV-1 Human T-cell lymphotropic virus type 1 ID Immunodominant IgG Immunoglobulin G IR Immunoreactive IN Integrase JDV Jembrana disease virus LTR Long terminal repeat M-tropic Macrophage tropic VMV Visna maedi virus MHR Major homology region MA Matrix NC Nucleocapsid Nef Negative factor ORF Open reading frame PBMC Peripheral blood mononuclear cells PBS Phosphate-buffered saline PBS-T Phosphate-buffered saline-Tween 20 PCR Polymerase chain reaction pi Post-infection PR Protease

Page 9: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

viii

qPCR Quantitative polymerase chain reaction qRT-PCR Quantitative reverse transcriptase polymerase chain reaction Rev Regulator of expression of virion proteins RT Reverse transcriptase RNA Ribonucleic acid RPMI Roswell Park Memorial Institute SIV Simian immunodeficiency virus SRLV Small ruminant lentivirus SU Surface unit T-tropic T-lymphocyte-tropic TCID50 Median tissue culture infective dose Tat Trans-activator of transcription protein TM Transmembrane glycoprotein U3 3’ Untranslated region U5 5’ Untranslated region Vif Viral infectivity protein VL Viral load Vpr Viral protein R Vpu Viral protein U Vpx Viral protein X WIB Western immunoblotting YT Yeast tryptone broth

Page 10: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

ix

Publication and International Conference Presentations

Publications arising from this thesis

McNab, T., Desport, M., Tenaya, W. M., Hartaningsih, N., and Wilcox, G. E. (2010). Bovine immunodeficiency virus produces a transient viraemic phase soon after infection in Bos javanicus. Vet. Microbiol. 141, 216-223.

Desport, M., Ditcham, W. G., Lewis, J. R., McNab, T. J., Stewart, M. E.,

Hartaningsih, N., and Wilcox, G. E. (2009). Analysis of Jembrana disease virus replication dynamics in vivo reveals strain variation and atypical responses to infection. Virology. 386(2), 310-6.

Lewis, J., McNab, T., Tenaya, M., Hartaningsih, N., Wilcox, G., and Desport, M.

(2009). Comparison of immunoassay and real-time PCR methods for the detection of Jembrana disease virus infection in Bali cattle. J Virol Methods. 159(1), 81-6.

Manuscripts submitted for publication

McNab, T., Desprt, M., Dobson, R., Tenaya, I.W.M., Hartaningsih, N., Wilcox, G.E. (2010) Prior Bovine immunodeficiency virus infection does not inhibit subsequent superinfection by the acutely pathogenic Jembrana disease virus. Virology. Oral presentations

“Bovine immunodeficiency virus infection fails to provide protection against subsequent Jembrana disease virus infection” Presented at the European Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations

“Towards a Jembrana disease virus specific diagnostic immunoassay- peptide mapping of gag and env proteins of bovine lentiviruses” Presented at the European Society for Veterinary Virology Conference in Budapest, Hungary 2009.

Page 11: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

1

Chapter 1: Introduction

Two bovine lentiviruses have been described, Jembrana disease virus (JDV) and

Bovine immunodeficiency virus (BIV). The 2 viruses, although genetically and

antigenically related, have been reported to have very different pathogenic effects in

cattle. JDV causes a severe, acute disease in Bali cattle (Bos javanicus) and a mild

disease or subclinical infection in other breeds of cattle, including B. taurus

(Soeharsono et al., 1990). The disease in Bali cattle is acute and associated with a

marked febrile response, very high titres of infectious virus in the blood and a case

fatality rate of about 21% (Soesanto et al., 1990). BIV infection is generally not

associated with significant clinical changes in B. taurus breeds of cattle, although

they have been reported: one study found an association between BIV and decreased

milk yield in dairy cattle (McNab et al., 1994) and another associated BIV infection

with marked weight loss and concurrent infections, suggesting immunosuppression

(Snider et al., 2003b). JDV appears to have a limited geographic distribution and is

restricted to Indonesia where Bali cattle are found (Hartaningsih et al., 1993), while

serological surveys have provided evidence for a worldwide distribution of BIV in

both cattle and buffalo (Bubalus bubalis) (Gonzalez et al., 2008; McNab et al., 1994;

Meas et al., 1998; Meas et al., 2000a; Meas et al., 2000b; Suarez et al., 1993). There

is also serological evidence of infection with a related non-pathogenic BIV-like virus

in Bali cattle in Indonesia on the island of Sulawesi and also in Bali (Barboni et al.,

2001; Desport et al., 2005). Although infection with a BIV-like virus has been

suspected in Bali cattle in Indonesia, this has not been confirmed. There is no

evidence of the nature of BIV infection in this cattle species and what effect BIV

infection might have on subsequent JDV infection.

Page 12: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

2

This thesis describes the development of a serological diagnostic assay to

differentiate JDV and BIV infection, the effects of BIV in Bali cattle and the

interaction between BIV and JDV infections in Bali cattle. As a background to these

investigations, a review of the literature relating to JDV and the other lentiviruses has

been undertaken and is reported in Chapter 2. The review includes the key features of

the various lentiviruses, comparing their genome arrangement, replication and host

immune responses to infection. It also includes the various techniques that have been

described for diagnosis of lentivirus infections, methods of differentiating between

closely-related lentivirus infections and the effects of infection with multiple strains

of closely related lentiviruses.

Chapter 3 describes an attempt to detect JDV and BIV in Bali cattle on the island of

Bali. Two quantitative PCR (qPCR) assays were developed to detect JDV and BIV

proviral DNA within peripheral blood mononuclear cells (PBMC) of naturally

infected animals.

Despite serological evidence for the presence of BIV in the Bali cattle population on

the island of Bali, the investigations reported in Chapter 3 failed to detect evidence of

BIV in the cattle that were sampled. Investigations were therefore undertaken to

determine the susceptibility of Bali cattle to BIV infection in an effort to better

understand the nature of the infection in this species, and these investigations are

reported in Chapter 4. Nineteen cattle were experimentally infected with the R29

strain of BIV and monitored for up to 65 days after infection. The presence of virus

in plasma and other tissues, the presence of proviral DNA in the PBMC and other

tissues, and the immune response to virus antigens was examined and is reported.

Page 13: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

3

The possibility that BIV infection of Bali cattle might modify the effect of

subsequent JDV infection was also investigated and these results are presented in

Chapter 5. These studies were undertaken to determine what might happen on the

island of Sulawesi, where BIV infection is suspected to occur in Bali cattle, if JDV

were to spread through the Bali cattle population of that island. If prior BIV infection

were to inhibit subsequent JDV infection, it was hypothesised that this might form

the basis of a method of vaccination for the control of Jembrana disease.

Due to the presence of cross-reactive epitopes between JDV and BIV proteins,

current serological assays are not capable of discriminating between antibody to the

2 bovine lentiviruses. During the studies undertaken and reported in Chapters 3, 4

and 5, the difficulty of distinguishing antibody to BIV and JDV made it difficult to

monitor the serological response to the individual virus infections. An attempt was

therefore made to develop a peptide antigen capable of differentiating between

antibody to the 2 viruses. Overlapping virus peptides were synthesized and used in

an enzyme linked immunosorbent assay (ELISA) format with serum samples taken

from JDV and BIV infected cattle to determine their reactivity to the peptides. The

results are reported in Chapter 6.

A general discussion of the results obtained and reported in the thesis is presented in

Chapter 7.

Page 14: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

4

Chapter 2: Literature Review

This review covers the general biological features of the retroviruses and

lentiviruses, in particular the lentiviruses that cause disease in animals. Also

reviewed are the various immune responses to animal lentivirus infection (including

cross-reactive epitopes) and the phenomenon of lentivirus superinfection resistance.

The nomenclature used in this thesis for viral genes and proteins is that suggested by

(Fauquet, 2005) where gene names are in lower case and italicised, eg. env, and

where the abbreviations for the encoded proteins have the initial letter in uppercase

and are not italicised, eg. Env and TM.

Retroviruses

Physiochemical characteristics of retroviruses

The key feature of the family Retroviridae is their mode of replication, involving

reverse transcription of the virion RNA into linear double-stranded DNA (Baltimore,

1970; Temin et al., 1970), and the integration of this double-stranded proviral DNA

into the genome of the cell. This reverses the normal flow of genetic information

from DNA to RNA, hence the name retrovirus (Coffin, 1997).

Retrovirus taxonomy

Many viruses have been classified within the family Retroviridae with a significant

proportion of them associated with disease (Coffin, 1997). On the basis of

evolutionary relatedness, retroviruses are separated into 7 genera: Lentivirus,

Spumavirus, Alpharetrovirus, Betaretrovirus, Gammaretrovirus, Deltaretrovirus and

Epsilonretrovirus (Buchschacher, 2001). The lentiviruses and spumaviruses are

distinct from the other 5 genera in that they do not have oncogenic potential. Some of

the well known oncogenic retroviruses include Rous sarcoma virus, Human T-

Page 15: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

5

lymphotropic virus (HTLV-1) and Mouse mammary tumour virus. Less well

understood and researched are the spumaviruses which cause no known disease, of

which Human spumavirus is the type species (Coffin, 1997; Goff, 2001). The

lentiviruses have been studied extensively since the discovery of Human

immunodeficiency virus (HIV).

Lentiviruses

Most lentivirus infections are characterised by a long asymptomatic period before the

onset of chronic clinical disease with a slow but inevitable progression to death

(Campbell et al., 1998). Examples of lentiviruses inducing this type of chronic

infection include Visna maedi virus (VMV), HIV and Caprine arthritis encephalitis

virus (CAEV). Some, however, induce a rapidly progressive acute disease, including

JDV, Equine infectious anaemia virus (EIAV) and the Simian immunodeficiency

virus (SIV) SIVsmm-PBj. This section will review features of lentiviruses that are

common to the majority of members of the genus, including genome structure and

organisation, their mode of replication and common clinical features of infection.

General structure of lentiviruses

Lentiviruses are roughly spherical in shape with an average virion diameter of

approximately 100 nm including the surrounding bilayered lipoprotein envelope

(Fauquet, 2005). They are sensitive to heat, detergent and formaldehyde (Goff,

2001). The envelope contains 2 types of surface projections, the surface unit (SU)

and transmembrane (TM) glycoproteins (Fauquet, 2005; Wagner, 1999). The internal

virion is composed of the distinctively cone-shaped capsid (CA) which surrounds the

nucleocapsid (NC) and contains protease (PR), integrase (IN) and reverse

transcriptase (RT) enzymes (Figure 2.1). The RNA genome is located within the

nucleocapsid (Goff, 2001).

Page 16: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

6

The lentivirus genome

Lentiviruses have 2 identical linear, positive-sense, single-stranded RNA genomes

which range in size from 7-10 kb (Goff, 2001; Peterlin, 1995). Reverse transcription

takes place from one strand at a time. There are 3 major open reading frames (ORF)

in each strand that transcribe and translate 3 polyproteins which are then cleaved by

proteases into approximately 8 proteins. The 3 polyproteins are Gag, Pol and Env

(Figure 2.2). The gag ORF produces the structural proteins MA, CA and NC. The pol

ORF encodes the intravirion enzymes: RT responsible for copying the single-

stranded RNA genome into the double-stranded DNA, IN which is required to

incorporate the double stranded DNA into the host genome forming the provirus, and

PR, required to cleave the encoded polyproteins into smaller proteins (Miller et al.,

2000; Tobin et al., 1994). The env gene produces the 2 envelope proteins, SU and

TM, which play a vital role in receptor recognition and entry of the virus into the cell

(Fauquet, 2005; Wagner, 1999).

Figure 2.1. (A) Schematic representation of a mature HIV-1 virion, showing the location of the

major viral proteins, the lipoprotein envelope and genomic RNA. (B) Tomogram of a mature HIV-1

particle derived by electron cryotomography. Images from Ganser-Pornillos et al. (2008).

Page 17: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

7

In addition to the obligatory gag, pol and env ORF common to every retrovirus,

lentiviruses also have a number of accessory genes (Table 2.1), including rev, tat, vif,

vpr, vpu and nef (Fauquet, 2005), which modulate the replication of the virus and

probably contribute to clinical latency and pathogenic mechanisms (Clements et al.,

1996). Rev plays an essential role in the replication cycle of all lentiviruses as it

facilitates the export from the nucleus of unspliced RNAs whose translated products

are later utilised in virus replication (Malim et al., 1989). Like Rev, Tat is produced

early in the replication cycle and plays a role in the expression of viral transcripts

from a promoter within the long terminal repeat (LTR) (Miller et al., 2000). HIV Tat

has also been shown to modulate the expression of cellular genes and has numerous

other roles in viral replication and pathogenesis (Chen et al., 2000). FIV is the only

lentivirus without the tat ORF but ORF2 encoded by orf2 is predicted to have Tat-

like activity and to act via cellular transcription factors during the expression of viral

transcripts from the promoter within the viral LTR (Chatterji et al., 2002; Miller et

al., 2000; Miyazawa et al., 1994) but requires additional elements within the LTR,

unlike other lentivirus Tat proteins (Chatterji et al., 2002). It has been proposed that

VMV and CAEV lack a tat ORF and that the ORF that is designated tat in these

viruses (named tat because of its similar position in the genome to the tat ORF in the

primate lentiviruses) instead encodes a Vpr-like accessory protein (Villet et al.,

2003).

Page 18: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

8

Figure 2.2. Genome organisation of lentiviruses. The location of the structural and accessory genes

are indicated, orientated 5’ to 3’. Each virus has 3 major open reading frames, gag, pol and env, which

transcribe and translate 3 polyproteins. Image from Craigo et al. (2010).

Page 19: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

9

The vif ORF is transcribed and translated to Vif (viral infectivity factor), thought to

aid in the infectivity and spread of virus, although its mechanism of action is still

unclear (Clements et al., 1996; Miller et al., 2000). Vif has recently been implicated

in protecting virions against the actions of Apolipoprotein B mRNA-editing enzyme-

catalytic polypeptide-like (APOBEC) proteins. APOBEC3G and APOBEC3F are

cytidine deaminases which are packaged into HIV-1 virions and result in the

production of non-infectious virions due to the hypermutation of HIV proviral DNA.

Vif protects the virus against lethal incorporation of the APOBEC proteins by

marking them for ubiquitin-dependent degradation (Goila-Gaur et al., 2008, Romani

et al., 2009).

Vpr encodes Vpr (viral protein R) which, in HIV-1, mediates the nuclear import of

viral RT complexes in non-dividing cells and alters the cell cycle and proliferation

status of the infected host cell. In HIV-2 and SIVsm, Vpr inhibits cell cycle

progression while Vpx (encoded by vpx) is responsible for the nuclear import of the

viral RT complex (Fletcher et al., 1996; Stivahtis et al., 1997). The vpu ORF encodes

Vpu (viral protein U) that enhances the efficiency of virion production and induces

rapid degradation of CD4 (Maldarelli et al., 1993). The nef ORF encodes Nef

(negative factor) whose function is to decrease the expression of CD4 on T-cells; it is

thought to have this effect by increasing the rate of endocytosis of CD4 on the cell

surface which would ultimately prevent re-infection of cells that already harbour the

virus (Benson et al., 1993; Goff, 2001; Peterlin, 1995). The accessory ORF s2 is

unique to EIAV and its role in virus replication is unclear but it is thought to play a

part in replication and virulence of EIAV (Li et al., 1992; Nilsen et al., 1996).

Page 20: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

10

Table 2.1. Accessory genes of each lentivirus, presence indicated by √ (Chakrabarti

et al., 1987; Chatterji et al., 2002; Clements et al., 1996; Dewhurst et al., 1990;

Freed, 2001; Li et al., 1992; Miller et al., 2000; Nilsen et al., 1996; Stivahtis et al.,

1997; Tobin et al., 1994). SIVsm has a gene encoding the Vpx protein, other SIVs

lack this gene (Stivahtis et al., 1997). The presence of tat within the VMV and

CAEV genomes has been debated (Villet et al., 2003).

HIV-1 HIV-2 SIV FIV VMV CAEV EIAV BIV JDV

vif √ √ √ √ √ √ √ √

vpx √ √

vpr √ √ √

tat √ √ √ √ √ √ √ √

rev √ √ √ √ √ √ √ √ √

vpu √ √

nef √ √ √

tmx √ √ √

Other

ORF 2/A S2

vpw,

vpy

Lentivirus replication

Infection of a cell by a lentivirus commences at the surface of the host cell (Figure

2.3). The SU interacts with specific receptors on the target cell. For example, HIV-1

SU interacts with the CD4 receptor located on the surface of T-lymphocytes and

monocytes/macrophages. The interaction brings about a conformational change in

the TM glycoprotein and ultimately leads to the fusion of the viral envelope with the

Page 21: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

11

membrane of the target cell (Clapham et al., 2002; Clements et al., 1996). The virus

then moves into the cytoplasm where it is partially uncoated and virion enzymes

encoded by pol are released. The virion enzymes RT and RNase H copy the viral

RNA within the partially uncoated virion, generating a double-stranded DNA copy

of the viral RNA genome, referred to as the provirus. The provirus forms a complex

with a number of viral proteins including IN, MA, NC, RT, and possibly others, to

form the pre-integration complex, which translocates to the nucleus. Integrase then

catalyses the insertion of the linear, double-stranded viral DNA into the host cell

chromosome (Freed, 2001). At this point, the proviral DNA may remain integrated

and the cell can remain latently infected, or a productive infection may result. It

remains unclear as to what causes a cell to become productively infected, although it

is thought that the presence of specific transcription factors present in mature cells

may stimulate transcription of viral genes. For example, it has been proposed that the

binding of the transcription factor NF-κB, which is found in activated T-

lymphocytes, to the HIV-1 LTR, is important for transcriptional activation in vitro of

HIV-1 in T-cell lines (Clements et al., 1996; Wagner, 1999). Others have proposed

that defective HIV-1 particles preferentially activate CD4+ T-cells which render

them permissive for HIV replication and help to drive HIV pathogenesis (Finzi et al.,

2006).

Once transcription has been activated, expression of viral mRNA begins when RNA

polymerase II binds to the U3 region of the 5' LTR and transcription then proceeds

towards the 3' end of the provirus and into the host DNA. The RNA is cleaved and

polyadenylated at the R-U5 border of the 3' LTR (Figure 2.4), which yields a

complete unspliced viral genomic RNA suitable for incorporation into the virion

particle. A portion of the RNA produced at this level is then spliced by the cellular

Page 22: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

12

splicing machinery to give rise to one or more subgenomic RNAs. Both the

unspliced and spliced RNAs are exported from the nucleus for translation (Goff,

2001).

Once in the cytoplasm, the RNAs transcribed from gag, pol and env are translated by

ribosomes into polyproteins (large precursor protein molecules). In HIV, the gag

ORF encodes a polyprotein precursor of 55 kDa, designated Pr55Gag, that is cleaved

by viral PR to produce MA (p17), CA (p24), NC (p7) and p6. The pol-encoded

enzymes are initially synthesised as part of a large polyprotein precursor, Pr160GagPol

that is cleaved by the virus encoded protease into PR, RT and IN. The Env precursor,

gp160, is cleaved by cellular protease into SU (gp120) and TM (gp41) (Egberink et

al., 1992; Freed, 2001).

Gag and Gag-Pol precursors assemble beneath the plasma membrane and incorporate

viral genomic RNA during the process of budding, while SU and TM glycoproteins

are also inserted into the viral envelope at this stage. After the virion has been

released, it matures when PR cleaves Gag precursors into their functional subunits.

The mature virion is then able to infect other cells (Tobin et al., 1994).

Page 23: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

13

Figure 2.4. Schematic illustration of the changes to lentivirus long terminal repeats during the change

from viral RNA to proviral DNA to viral mRNA (transcript). Illustrations are orientated 5’ to 3’. The

positions of the R (repeat), U3 and U5 regions are shown. Viral mRNA is expressed when RNA

polymerase II binds to the U3 region of the 5’ LTR. Image from Coffin et al. (1997).

Figure 2.3. Schematic illustration of the lentivirus replication cycle depicting the major events in

the replication cycle. Image from Ganser-Pornillos et al. (2008).

Page 24: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

14

Common characteristics of lentiviruses

Members of the genus Lentivirus share a number of features. They all replicate in

non-dividing terminally differentiated cells, have the ability to integrate their

genomes into the chromosomal DNA of non-dividing infected cells and they are

highly species-specific in terms of their ability to cause disease. Common clinical

features of infection shared by the majority of lentiviruses include long

asymptomatic incubation periods before the onset of a usually chronic disease,

persistence of virus infection in the face of vigorous immune responses including

neutralising antibody and cytotoxic T-lymphocytes (CTL), multi-organ disease and

replication in cells of the immune system and brain (Clements et al., 1996; Mealey et

al., 2004; Trautwein, 1992). It is often the immune response of the host against the

infected cells that results in the wide range of disease symptoms observed (Gonda,

1992).

The ability of the viruses to persist in the infected host is, at least in part, associated

with the capacity of lentiviruses to exhibit a wide array of genetic and antigenic

variations, particularly in env. Variations are produced in response to biological and

immunological selective pressures as the virus (usually) successfully avoids

clearance by defence mechanisms (Leroux et al., 2001; Mealey et al., 2004).

Page 25: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

15

Primate lentiviruses

HIV and SIV cause disease in primates and it is the study of these lentiviruses that

has resulted in most of our knowledge of the molecular biology and mechanisms of

disease associated with lentivirus infections. This section will review HIV and SIV

with a particular focus on their genome organisation, cell tropism, clinical features of

diseases they induce, and species specificity.

Human immunodeficiency virus

Arguably the most studied virus in the world today, HIV was first identified in 1983

as the agent responsible for an acquired immunodeficiency syndrome (AIDS) which

led to opportunistic infections and eventually death (Barre-Sinoussi et al., 1983;

Gallo et al., 1984). It has assumed very significant pandemic proportions and it is

estimated that about 33 million people are infected with the virus (UNAIDS, 2008).

Based on epidemiological and genetic studies, HIV isolates form two clusters, HIV-1

and HIV-2, which are distinguished by variations primarily in env. They also show

differences in transmission rate and pathogenicity (Levy, 2009). HIV-1 is the type

species of the genus Lentivirus and although the published literature describing

aspects of the molecular biology of HIV-1 and the associated infection is very large,

a brief overview only will be given in this review.

Many lentiviruses evade the host immune system via their genetic variability (Lopez

et al., 2006). Phylogenetic analyses of HIV-1 isolates from around the world indicate

that more than 10 major groups of distinct genetic subtypes or clades of HIV-1 can

be distinguished. Clades are differentiated by genomic differences in env of 15% or

greater (Barker, 1995). The distribution of the clades tends to have a geographic

basis: Subtype C (clade C) infections are most commonly found in south Africa,

India, Ethiopia and east Africa. Subtype A clades are found in eastern Europe,

Page 26: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

16

central Asia, west, east and central Africa while subtype B is present in the

Americas, western Europe and east Asia. Subtype D is present in north Africa, the

middle east, east and central Africa (Hemelaar et al., 2006).

In HIV the major determinants of cell tropism are the cell surface receptors used to

gain entry into the cell. HIV-1 uses the CD4 receptor and a co-receptor to enter the

host cell (Sattentau et al., 1988). The main cells that have the 2 receptors needed for

HIV-1 to gain entry are the CD4+ T-helper subset of lymphocytes, the CD4+ cells of

macrophage lineage and some dendritic cells (Clapham et al., 2002; Freed, 2001).

Major co-receptors for HIV-1 include the 2 chemokine receptors CCR5 (expressed

on macrophages) and CXCR4 (expressed on T-cells) but other co-receptors are used

(Deng et al., 1996; Doranz et al., 1996; Feng et al., 1996). CCR5-utilising HIV-1

variants dominate the early phases of HIV-1 infection while CXCR4-utilising HIV-1

variants dominate the latter phases of HIV-1 infection, and the switch from CCR5-

utilising to CXCR4-utilising is associated with accelerated disease progression (Ito et

al., 2003).

HIV-1 will establish infection in both humans and chimpanzees but will only cause

disease in humans, with a few rare exceptions (Freed, 2001, Keele et al., 2009).

There are 3 phases to the course of HIV infections. In the initial phase, there is a

period of rapid virus replication associated with influenza-like symptoms,

commencing about 2 weeks post infection (pi) and lasting for 2-3 weeks.

Subsequently, there is a variable asymptomatic period of weeks to years during

which lymphadenopathy can develop. In the third phase, the destruction of the T-cell

population causes the onset of AIDS, seen in about 30% of infected people within 5-

7 years and later in others (Campbell et al., 1998). The loss of T-cells results in the

body being unable to overcome opportunistic infections, and the immune system

Page 27: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

17

finally fails, which without anti-viral therapy will ultimately lead to death (Wagner,

1999). The loss of CD4+ T-cells and the changes in HIV RNA levels are shown in

Figure 2.5. Key clinical manifestations of HIV-1 infection are immunodeficiency,

lymphadenopathy, opportunistic infections, encephalopathy, emaciation, Kaposi's

sarcoma and other cancers (Gonda, 1992).

Simian immunodeficiency viruses

Simian lentiviruses have been identified in several species of non-human primates by

epidemiological studies using serological assays and virus isolation. Each SIV is

named with a subscript that denotes the species from which the virus was first

isolated, for example those from sooty mangabeys as SIVsmm, from macaques as

SIVmac and from chimpanzees as SIVcpz (VandeWoude et al., 2006). In their natural

host to which they have adapted they do not normally cause disease but when they

infect species in which they do not normally occur, disease frequently results. These

Figure 2.5. The progression from infection with HIV to death in a victim. The patient was infected

at or near week 0. The diagram illustrates the levels of HIV RNA, the gradual decline in CD4+ T-

cells and the onset of clinical signs. Image from Pantaleo et al. (1995).

Page 28: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

18

viruses share many features in common with HIV, particularly nucleotide homology,

genome organisation, size (approximately 9 kb in length), receptor usage as well as

the clinical manifestations of disease they induce in non-natural hosts (Clements et

al., 1996; Sattentau et al., 1988).

All SIV have a tropism for CD4+ macrophages or T-cells. Macrophage-tropic (M-

tropic) SIV can efficiently replicate in macrophages, primary CD4+ T-cells and a

variety of T-cell lines whereas T-cell-tropic (T-tropic) SIV replicate in primary

CD4+ T-cells and in T-cell lines but not macrophages (Edinger et al., 1999). Both M-

and T-tropic SIV strains use CCR5 to gain entry into CD4+ cells (Edinger et al.,

1997). The locations of viral replication are thought to be responsible for the varying

clinical manifestations of SIV infection: replication in cells of the

monocyte/macrophage lineage results in disease manifestations in the central nervous

system and lungs; replication in lymphocytes results in a loss of CD4+ lymphocytes

which in turn results in immunodeficiency and increased susceptibility to

opportunistic infections (Sharma et al., 1992).

While the majority of SIV cause disease after a relatively long period of infection,

there are strains which cause disease after only a short incubation period, such as

SIVsmmPBj14. This strain causes an cute disease and death within 6 to 10 days after

intravenous inoculation into pig-tailed macaques and rapidly replicates to high titres

(Fultz et al., 1994, Tao et al., 1995). The lethal nature of this phenotype is possibly

associated with insertions within the V1 region of env and within the NF-κB

enhancer element in the LTR, which enhances transcription and replication kinetics

(Tao et al., 1995).

Page 29: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

19

SIV that originate in African primate hosts are thought to be relatively ancient

viruses that result in non-pathogenic infections in their native hosts. A similar

situation is seen with lentivirus infections of non-domestic cats (Hahn et al., 2000;

Terwee et al., 2005).

Non-primate lentiviruses

Feline immunodeficiency virus

FIV is associated with acquired immunodeficiency in cats and was first isolated in

California (Pedersen et al., 1987). Since its initial isolation, similarities in the disease

syndrome induced in cats to that of HIV in humans have created considerable interest

in the virus as a potential animal model for HIV infection (Dandekar et al., 1992;

Dua et al., 1994; Gardner et al., 1989; Olmsted et al., 1989). The virus infects several

species of Felidae, including the domestic cat Felis cattus, throughout the world but

particularly in Europe, East Asia, Australasia and North America (Brown et al.,

1994; Duarte et al., 2006; Little et al., 2009; Olmsted et al., 1992). Circumstantial

evidence suggests that the most efficient mode of virus transmission is horizontally

by biting (Yamamoto et al., 1989). Other studies have shown that it can be

transmitted through semen and vertically during pregnancy (Jordan et al., 1998;

Wasmoen et al., 1992).

FIV has a broader target cell range than HIV and replicates in both CD4+ and CD8+

lymphocytes, macrophages and immunoglobulin G-positive lymphocytes (Beebe et

al., 1994; Brown et al., 1991; English et al., 1993). The primary receptor of FIV is

CD134, a T-cell-activation antigen and co-stimulatory molecule (Shimojima et al.,

2004). Known co-receptors include CXCR4 (Willett et al., 1997). The progression

from infection to immunosuppression to death in domestic cats is well characterised

and mirrors HIV-1 infection in humans. There is a transient acute stage of the illness

Page 30: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

20

a few weeks pi that is characterised by fever and lymphadenopathy. The next phase

is asymptomatic, lasting up to 5 years. The third and final phase of infection is

characterised by a gradual decline in CD4+ T-lymphocytes leading to

immunodeficiency. This stage, like AIDS in HIV-infected humans, is characterised

by the occurrence of opportunistic infections, neurological disorders and tumours of

various aetiologies, resulting in death usually within a few months (Dean et al., 1996;

Egberink et al., 1992; Ryan et al., 2003; Sauter et al., 2001).

Analogous to SIVsmmPBj14 inoculation into pig-tailed macaques, FIV-CPGammar also

causes an acute disease with a high case fatality rate. Mortalities range from 50 to

100% in kittens ≤12 weeks of age after a short incubation period following

intravenous inoculation However, in contrast to SIVsmmPBj14, acute phase virulence

was induced by acute-phase virus passage (Diehl et al., 1995) and the severe and

rapidly progressive disease could not be induced in young adult cats (Pedersen et al.,

2001).

Page 31: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

21

FIV in non-domestic feline species

Serological surveys of a number of non-domestic feline species have revealed at least

17 species with FIV-like virus infection (Brown et al., 1994; Olmsted et al., 1989;

VandeWoude et al., 2006; VandeWoude et al., 2002). Isolates from pumas (also

referred to as cougar, mountain lion and panther; Puma concolor), lions (Panthera

leo), Pallas cat (Felis manul) and bobcats (Lynx rufus) have been genetically

characterised (Poss et al., 2006; Poss et al., 2008), and these isolates are distinct from

each other and are related to, but distinct from FIV of the domestic cat (Olmsted et

al., 1992). The standard nomenclature for designation of strains originating from

different species is to append genus and species identifiers for the feline species as a

subscript to FIV. For example FIV isolated from a lion is referred to as FIVple and

domestic cat as FIVfca (Brown et al., 1994; VandeWoude et al., 2006).

The available evidence indicates these viruses have been endemic within cat families

for a long period (Biek et al., 2006). Although the clinical effects of FIV from non-

domestic cat populations have not been well studied, it appears that infections due to

these viruses do not cause widespread disease (Biek, 2006; Brown et al., 1994;

VandeWoude et al., 2002; VandeWoude et al., 2003). The asymptomatic nature of

infection is most probably because of a long evolutionary association between virus

and host (Biek, 2006). Signs of neurologic disease have been reported in captive

lions (Brennan et al., 2006) but this may be attributed to the animals outliving their

“normal” life span. FIVpco from pumas is able to establish a productive infection in

domestic cats but does not cause T-cell dysregulation (unlike FIVfca) or clinical signs

and is able to be cleared from PBMC rapidly (Terwee et al., 2005; VandeWoude et

al., 2003). These cats also generate humoral and cell-mediated immune responses

Page 32: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

22

reactive against both FIV and non-domestic cat isolates of FIV (VandeWoude et al.,

2003).

Equine infectious anaemia virus

EIAV, the cause of a chronic relapsing or intermittent anaemia in horses, was the

first lentivirus to be identified and the first non-plant virus to be discovered (Leroux

et al., 2004; Ligné, 1843; Vallée, 1904). The virus causes a persistent infection in

horses and closely related equids, such as donkeys and mules (O'Rourke et al., 1988;

Spyrou et al., 2003). It is transmitted mechanically mainly by blood-feeding biting

arthropods such as tabanids, or iatrogenically on contaminated needles, but contact

infection can also occur (Hawkins et al., 1976; Kemen et al., 1978; Li et al., 2003;

Williams et al., 1981). Because of its transmission by arthropods, the infection is

most common in geographic areas with long vector seasons (Issel et al., 1988).

EIAV is an exclusively tissue macrophage-tropic lentivirus which utilises the equine

lentivirus receptor-1 to gain entry into macrophages (Sellon et al., 1992; Zhang et al.,

2005). The virus causes a relapsing disease characterised by periods of depression,

fever, diarrhoea, anaemia, thrombocytopenia and haemorrhaging due to severe

depletion of platelets, which are associated with a high level viraemia (Hammond et

al., 1997; Harrold et al., 2000; O'Rourke et al., 1988; Oaks et al., 1998). The disease

cycles begin approximately 1-3 weeks pi, last 3-5 days and occur at irregular

intervals of weeks to months. The periodic disease cycles occur for 8-12 months pi,

after which infected horses normally progress to a subclinical infection lasting for the

life of the infected animal; these persistently infected animals serve as a source of

infection for other animals (Hammond et al., 1997; Issel et al., 1982; Montelaro et

al., 1984; Oaks et al., 1998; Payne et al., 1998; Salinovich et al., 1986). Between the

clinical episodes of disease, viral loads are greatly reduced (of the order of 4- to 733-

Page 33: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

23

fold) and viral transcription within macrophages is restricted (Oaks et al., 1998). In

response to immune pressure, rapid genomic variations occur during replication

which results in altered glycoprotein structures and antigenic changes. Variants have

been detected within 28 days and this variation is responsible for the cyclical nature

of the disease, new antigenic variants having a replication advantage as they are not

susceptible to pre-existing immunity (Ball et al., 1992; Montelaro et al., 1984; Payne

et al., 1984; Salinovich et al., 1986).

Small ruminant lentiviruses

It was initially thought that VMV and CAEV were specific for sheep and goats,

respectively, but recent evidence has shown that VMV and CAEV are capable of

infecting both sheep and goats as well as some small ruminant species living in the

wild (Guiguen et al., 2000; Leroux et al., 1997; Narayan et al., 1980; Pisoni et al.,

2005; Ravazzolo et al., 2006; Shah et al., 2004). As a result, VMV and CAEV are

now collectively referred to as small ruminant lentiviruses (SRLV). The 2 viruses

share many similarities at both the genomic and antigenic level, particularly in gag

and pol (Brinkhof et al., 2007; Jolly et al., 1989; Pasick, 1998; Pyper et al., 1986;

Zanoni, 1998), and there is antigenic cross-reactivity between their CA proteins

(Clements et al., 1980). Some SRLV, however, show significant differences in their

LTR, env and regulatory genes (Jolly et al., 1989; Pyper et al., 1986; Zanoni, 1998).

They also differ phenotypically, with the prototypic Icelandic VMV strains inducing

syncytia and lysis of infected cell cultures whereas the prototypic CAEV-Cork strain

induces syncytia with a persistent but non-lytic infection of cell cultures (Narayan et

al., 1980; Pisoni et al., 2007; Querat et al., 1984).

Page 34: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

24

Visna maedi virus

VMV is a natural pathogen of sheep (Zhang et al., 2000; Zink et al., 1987) and has

been isolated from the majority of sheep-rearing areas of the world, excluding

Australia and New Zealand (Dawson, 1988). VMV was initially reported as a cause

of progressive pneumonia of sheep in South Africa in 1915, then in Montana in the

1920's, then in sheep in Iceland in the 1950’s (Dawson, 1988; Jolly et al., 1989;

Sigurdsson et al., 1952; Sigurdsson et al., 1957). The virus name is derived from the

Icelandic language where “maedi” can be translated as “dyspnoea” (difficult

breathing due to pneumonia) and “visna” as “fading away” (due to a demyelinating

leukoencephalomyelitis), representing the 2 forms of the disease (Pepin et al., 1998).

The main route in which the virus is transmitted is thought to be via ingestion of

infected colostrum and/or milk or via the respiratory tract, involving direct inhalation

of infected respiratory secretions, including the inhalation of infected alveolar

macrophages (McNeilly et al., 2008; Pepin et al., 1998; Preziuso et al., 2004).

Monocytes, lung tissue macrophages and spleen tissue macrophages (but not T-

lymphocytes) are the major cell targets in vivo for VMV replication and viral

expression is greatly increased when the monocytes mature into macrophages

(Gendelman et al., 1986). In PBMC populations, dendritic cells are the most

permissive for viral replication (Gorrell et al., 1992; Zhang et al., 2000).

Clinical disease can take months to years to develop (Nilsen et al., 1996) and consists

of a multi-system disease characterised by a chronic infiltration and proliferation of

mononuclear cells in the lungs (chronic interstitial pneumonia, maedi) and the central

nervous system (affected by a chronic and progressive, paralytic disease

characterised by inflammatory and demyelinating lesions in the CNS leading to

wasting and paralysis, visna) (Narayan et al., 1980). Organs less commonly affected

Page 35: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

25

include the joints (arthritis) and mammary glands (Bird et al., 1993; Deng et al.,

1986; Gorrell et al., 1992; Zink et al., 1987).

Caprine arthritis encephalitis virus

CAEV causes an economically significant disease in infected goats, particularly in

Europe, the Americas, Asia and Australia (Campbell et al., 1998; Herrmann et al.,

2003b; Mselli-Lakhal et al., 2007; Zanoni, 1998). The main route of virus

transmission is via colostrum but horizontal spread is also achieved via infected

secretions if there is direct contact between infected and susceptible animals

(Ravazzolo et al., 2006) or from doe to foetus either prior to or during the birth

process (East, 1993).

The virus, like VMV, infects cells of the monocyte/macrophage lineage and dendritic

cells and virus expression is activated during maturation of monocytes to

macrophages (Narayan et al., 1983). It does not infect lymphocytes and so does not

directly result in immunosuppression, unlike SIV and HIV. Infection results in a

chronic inflammatory disease affecting the joints, central nervous system, lungs and

mammary glands, and depending on the organs affected, can result in emaciation,

respiratory distress, mastitis, paralysis or arthritis (Dawson, 1988; Herrmann et al.,

2003b; Narayan et al., 1983) (Ravazzolo et al., 2006). Up to 40% of infected goats

develop chronic arthritis (Cheevers et al., 1997).

Bovine lentiviruses

Currently there are 2 lentiviruses known to infect cattle, BIV and JDV. The first

bovine lentivirus discovered was BIV (Tobin et al., 1994; Van der Maaten et al.,

1972) while JDV has more recently been identified as a lentivirus (Chadwick et al.,

1995b).

Page 36: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

26

While the clinical and pathological syndromes associated with the 2 bovine

lentiviruses are markedy different, the viruses share a very close genetic and

antigenic relationship (Chadwick et al., 1995b). These are shown in Table 2.2.

Table 2.2. Comparison of putative protein products of JDVTab87 and BIV127.

JDV BIV

Protein Amino acids

Mol. Mass (kDa)

Amino acids

Mol. Mass (kDa)

Amino acid identity

gag precursor 436 48.8 476 53.4 62%

MA 125 14.3 126 14.6 60%

p2L - - 22 2.5

CA 226 25.3 219 24.6 75%

p3 - - 25 2.7

NC 85 9.2 66 7.3 63%

p2 - - 18 1.9

gag/pol precursor 1432 163 1475 168 66%

pol precursor 1027 118 1035 118 68%

env precursor 781 88.8 904 102 31%

SU 422 47.8 555 62.1 24%

TM 359 41.1 349 40.2 39%

vif 197 22.9 198 22.8 55%

tat 97 10.7 103 11.7 54%

(alt. tat)a (114) (12.5) - -

rev 213 23.8 186 20.7 35%

(alt. rev)a (201) (22.4) - -

tmx 164 18.5 159 18.0 29%

vpw - - 54 6.6

vpy - - 80 9.5 aAlternate forms of tat and rev generated by utilisation of alternative splice donor

site. Data sourced from Chadwick et al. (1995b).

Page 37: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

27

Bovine immunodeficiency virus

BIV is a naturally-occurring lentivirus found in cattle and based on serological

surveys, infections occur worldwide (Campbell et al., 1998; McNab et al., 1994;

Meas et al., 2000a; Suarez et al., 1995). The virus may be transmitted horizontally by

iatrogenic routes, blood-sucking arthropods, via natural or artificial insemination or

via milk from infected cows (Egberink et al., 1992; Meas et al., 1998; Meas et al.,

2000a). BIV is best known as a virus infection of B. taurus but New Zealand white

rabbits (Oryctolagus cuniculus) and sheep have been infected experimentally,

resulting in a persistent infection (Gonda, 1992; Jacobs et al., 1994; Pifat et al.,

1992).

Genome structure

The first isolation of BIV was from a cow in Louisiana and this isolate, designated

R29 (Van der Maaten et al., 1972), has since been cloned (Braun et al., 1988) and

sequenced (Garvey et al., 1990). BIV-R29 is antigenically and genetically stable

during long-term, persistent infection (Carpenter et al., 2000).

The BIV proviral genome is 8.96 kb in size and consists of the 3 main ORF gag, pol

and env and 6 accessory genes rev, tat, vif, tmx, vpw and vpy (Figure 2.2). The

functions of vif, tmx, vpw and vpy in BIV are yet to be described (Egberink et al.,

1992; Gonda et al., 1987; Miller et al., 2000; Tobin et al., 1994). The BIV Gag

polyprotein that is translated is processed into a number of proteins, the 3 major

proteins found in all lentiviruses (MA, CA and NC) and 3 smaller proteins (p2L, p3

and p2) (Tobin et al., 1994).

Page 38: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

28

Cell tropism and clinical signs associated with disease

Two studies have indicated that BIV may be pantropic. An initial in vivo study with

BIV isolate FL112 found proviral DNA in CD3+, CD4+ and CD8+ cells, B cells,

monocytes and WC1 cells in both the acute and chronic stages of infection

(Whetstone et al., 1997). A subsequent study involving the R29 isolate detected

proviral DNA and infectious virus within CD2+ (located on CD4+ and CD8+ T-

cells), WC1+ (located on γδ T-cells), mature B cells and monocytes during the early

stages of infection (Heaton et al., 1998). However, it is not clear from either study

whether these cell types were productively infected, as only proviral DNA was

detected. BIV-R29 is able to productively infect primary cultures of embryonic

bovine spleen and lung, Madin-Darby bovine kidney cells and embryonic rabbit

epidermal cells producing syncytia, cell lysis and infectious virus (Ferens et al.,

2007; Gonda, 1992; Heaton et al., 1998; Pifat et al., 1992; Suarez et al., 1993;

Whetstone et al., 1990; Zhang et al., 1997a; Zhang et al., 1997b).

BIV is most efficiently transmitted via infected blood and cell-free and cell-

associated tissue culture-derived virus (Pifat et al., 1992). The re-use of contaminated

needles during vaccination and blood collection, communal sharing of colostrum fed

to calves and the failure to cleanse contaminated instruments, have also been

suggested to play a role in the spread of BIV (Gonda, 1992). Inoculation usually

results in a subclinical infection without significant clinical effects (Flaming et al.,

1993) but clinical signs have been reported and include a transient leucocytosis,

lymphoid hyperplasia, lymph node enlargement, wasting and in some cases immune

suppression (Carpenter et al., 1992; Egberink et al., 1992; Zhang et al., 1997b). The

impact of BIV infection to the overall health of herds has not been established (Tobin

et al., 1994), although there is one Canadian study which suggested BIV infection

Page 39: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

29

had a significant effect on milk yield (McNab et al., 1994). Other studies have

suggested that persistent BIV infection plays a role in reducing functional immune

competence as shown by the frequent development of concurrent infections in BIV-

infected animals (Meas et al., 2000a; Snider et al., 2003a; Zhang et al., 1997b). It

would seem, however, that a direct role for BIV in chronic progressive disease or as

a cofactor in a specific disease is unlikely (Jacobs et al., 1994; Suarez et al., 1995).

Jembrana disease virus

In contrast to BIV, JDV causes a significant acute disease process in Bali cattle (Bos

javanicus) in Indonesia (Dharma, 1997; Kertayadnya, 1997). The first reported

outbreak of Jembrana disease occurred in Sangkargung, a village in the Jembrana

district of the island of Bali in Indonesia (Figure 2.6) in December 1964. The disease

only affected Bali cattle and while reference is sometimes made to an effect on

buffalo (Bubalus bubalis) this was anecdotal and has not been confirmed. The

disease spread through all districts of Bali by August 1965 with an estimated 20,000

to 70,000 deaths. The disease then disappeared but subsequent smaller outbreaks

were detected in 1972 in the Tabanan district and in 1981 in the Karangasem district

of Bali (Ramachandran, 1996; Soeharsono, 1997b).

Page 40: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

30

Figure 2.6. (A) The location of Bali in relation to other Indonesian islands (D.F.A.T., 2008). Districts of

Bali province, adapted from Streetdirectory.com (2009). Jembrana disease was first reported in the district

of Jembrana. Antibodies to JDV have since been detected in Sumatra, Java and Kalimantan. Clinical

disease and virus have also been detected in Sumatra and Kalimantan.

A B

Page 41: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

31

Early investigators proposed Rinderpest virus and then a rickettsia as probable causes

of Jembrana disease (Soeharsono, 1997b). A viral aetiology was subsequently

demonstrated, a conclusion based on filtration studies that suggested the infectious

agent in blood was too small for a rickettsia (Ramachandran, 1996). Based on its

estimated size of 50-100 nm, the probable presence of a lipid-containing envelope,

electron microscopic observations and reverse transcriptase activity, the agent was

then considered as a probable member of the family Retroviridae (Kertayadnya,

1997; Wilcox et al., 1992) and was subsequently identified, based on genome

structure and genomic nucleotide sequence analysis, as a lentivirus (Chadwick et al.,

1995b).

Significance of Jembrana disease

The case fatality rate resulting from experimentally induced Jembrana disease is

approximately 21% (Desport et al., 2009a). During the original outbreak

commencing in 1964, it was retrospectively estimated that 60% of the Bali cattle and

buffalo population on Bali island were affected in the following 12 months, of which

98.9% died (Ramachandran, 1996); this high prevalence and high case fatality rate is

unlikely and the evidence is obscure as at the time there were no veterinary services

on the island. The effect of JDV is widespread throughout Bali, as a consequence of

the important role that Bali cattle play in society. They are a source of employment

and help to sustain and generate profit in the agricultural system via the provision of

draught power and manure which in turn helps to improve soil fertility

(Wiryosuhanto, 1997).

Transmission and distribution of Jembrana disease

The probable modes of transmission of JDV include mechanical and contact

transmission. Virus can be mechanically transferred from infected cattle to

Page 42: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

32

uninfected cattle by iatrogenic means or by arthropods. Examples of iatrogenic

routes include multi-use needles during vaccination programs, as has been shown to

occur in EIAV (Li et al., 2003). Arthropod transmission is thought to be limited to

the acute viraemic episode when there is a high titre of virus in blood of affected

cattle, but it requires reasonably close contact between animals and the restriction of

movement of infected animals by quarantine has helped to reduce the spread of the

virus. The likelihood of contact transmission is supported by the detection of the

virus in secretions such as saliva, urine and milk and the ability to reproduce the

disease by conjunctival and oral inoculation of the virus (Hartaningsih et al., 1993;

Soeharsono et al., 1995b).

The disease has now been detected in 4 Indonesian islands: Bali, Kalimantan

(Indonesian Borneo), West Sumatra and East Java (Figure 2.6). There are no reports

of JDV infection of Bali cattle in any other area of Indonesia and clinical Jembrana

disease has not been reported in other cattle types which has led to the belief that the

disease is specific to Bali cattle (Hartaningsih et al., 1993; Wilcox et al., 1995).

However, other cattle types and buffalo can be infected experimentally and become

infected under field conditions (Soeharsono et al., 1995a) although they do not seem

to develop clinical disease. Bos taurus, B. indicus, crossbred Bali cattle (B. javanicus

x B. indicus) and buffalo can be infected and develop viraemia (Soeharsono et al.,

1990; Soeharsono et al., 1995a). The clinical changes and lesions that occur in these

cattle types are consistent with those observed in Bali cattle, but they are much

milder and would be difficult to detect under field conditions.

Page 43: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

33

The JDV genome

The genome of JDV is 7732 bp in length and is the smallest of all lentivirus

genomes. It contains the 3 major ORF typical of all retroviruses, gag, pol and env, as

well as accessory genes, vif, tat, rev and tmx (Figure 2.2) (Chadwick et al., 1995b;

Chen et al., 1999; Nilsen et al., 1996).

JDV is a unique lentivirus in that it is genetically stable, and there is a high level of

nucleotide conservation in gag, pol and env sequences taken from isolates throughout

Indonesia. Gag sequences in isolates from Bali and Sumatra had 97 to 100%,

nucleotide identity. Env sequences were also unexpectedly conserved with 96-99%

nucleotide identity, and 95 to 99% amino acid identity. The largest divergence was

seen in an isolate from South Kalimantan with only 88% identity to that of the

original JDVTab87 isolate (Desport et al., 2007).

This high level of nucleotide conservation is similar to the LTR regions of EIAV that

have been reported to be highly conserved during persistent infection of horses

(Maury et al., 2005; Reis et al., 2003). It is in contrast to other lentiviruses such as

HIV-1 (Balfe et al., 1990; Lamers et al., 1993; Wolfs et al., 1990), FIV (Brown et al.,

1994; Duarte et al., 2006) and CAEV (Valas et al., 2000) which exhibit greater levels

of genetic variation, particularly in envelope glycoprotein regions, in sequences

obtained from within one individual (Lamers et al., 1993; Simmonds et al., 1990),

between individuals and between different geographic locations (Balfe et al., 1990;

Maki et al., 1992).

Page 44: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

34

Cell tropism and clinical signs associated with Jembrana disease

It has been suggested that during the early stages of JDV infection, initial rounds of

virus replication occur in lymphoid tissue before a rapid and widespread

dissemination of virus to other tissues during the second phase of replication

(Chadwick et al., 1995a). JDV has a tropism for mature IgG-containing cells

(Desport et al., 2009a) and JDV-infected cells are found within spleen, lymphoid

tissues, bone marrow, lung, liver and kidney, as shown by in situ hybridisation

techniques for the detection of JDV genomic RNA (Chadwick et al., 1998).

JDV is not a typical lentivirus in that it causes an acute disease syndrome after a

short incubation period (Chadwick et al., 1995a). The incubation period before the

onset of clinical signs is 4-12 days and the clinical signs continue for 5-12 days

(Soeharsono, 1997a). Major clinical signs include a febrile response, lethargy,

anorexia and enlargement of superficial lymph nodes. Less frequently observed

clinical signs include erosions of oral mucous membranes, hypersalivation, nasal

discharge, diarrhoea and blood in the faeces. JDV has also been implicated in the

suppression of the humoral immune response in infected cattle, perhaps not

surprising as the histopathological changes reflect a disease primarily affecting the

lymphoid system (Dharma et al., 1994; Wareing et al., 1999). The major

haematological changes associated with JDV infection include leucopenia,

lymphopenia, eosinopenia and a slight neutropenia. In addition to these changes, a

mild thrombocytopenia, a normocytic normochromic anaemia, elevated blood urea

concentrations and reduced total plasma protein are also observed during the acute

disease (Soesanto et al., 1990).

Page 45: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

35

Immune response to infection with a lentivirus

The immune response to lentiviruses frequently takes longer to develop than in many

other virus types. For example, ponies infected with EIAV do not seroconvert to

envelope glycoproteins and CA until 3 weeks pi, with Env antibodies being

predominant. However, it requires 6-8 months for the EIAV humoral immune

response to fully mature into a high avidity, conformational epitope-specific response

(Hammond et al., 1997). The EIAV-specific CTL activity develops 3-4 weeks pi and

this correlates with the resolution of the primary viraemia (Hammond et al., 1997).

Studies involving the depletion of CD8+ lymphocytes in rhesus monkeys

subsequently infected with SIVmac support the role of these cells in controlling

viraemia (Schmitz et al., 1999) as do other studies of natural HIV-1 infections in

humans (Koup et al., 1994). Neutralising antibody is detected 2-3 months pi

(Hammond et al., 1997). In sheep infected with VMV, the seroconversion to CA also

occurs about 3 weeks pi (Singh et al., 2006) although serum neutralising antibodies

are not detectable until 1-3 months pi (Petursson et al., 1976), slightly earlier than in

EIAV. The time course of the immune response to HIV infection is summarised in

Figure 2.7 and its relationship to the development of other features of the virus

infection are shown in Figure 2.8.

Page 46: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

36

Also of importance in limiting infection with lentiviruses are non-adaptive immune

responses. Studies of HIV and FIV have shown that natural killer cells have an

important role in controlling acute infection (Howard et al., 2010) while cytidine

deaminases such as the previously mentioned APOBEC family of proteins restrict

viruses shortly after they have entered the cell by interfering with viral DNA

formation (Goila-Gaur et al., 2008, Romani et al., 2009). Other important innate

immune responses include TRIM5α which is thought to inhibit HIV-1 transduction

in rhesus macaque cells, non-coding RNAs such as micro RNAs and silence inducing

RNAs (Strebel et al., 2009), anti-viral cytokines such as interferon and dendritic cells

(Williams et al., 2009).

Page 47: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

37

Figure 2.8. Estimated time course for maturation of the host immune response in EIAV-infected

animals. Image from Leroux et al. (2004). Env-specific cytotoxic T-cells appear within the first month

after infection as do Env- and p26 (CA)-specific IgG. Neutralising antibodies appear after 2-3 months

after infection.

Figure 2.7. Estimated time course for host immune response to acute HIV infection. Image from

Levy (2007).

Page 48: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

38

Humoral immune response to infection with bovine lentiviruses

A number of assays have been used to detect and quantify the immune response to

the bovine lentiviruses, most commonly Western immunoblotting (WIB) and ELISA.

Antigen can be prepared using cell culture systems (Burki et al., 1992; Hammond et

al., 1997; O'Rourke et al., 1988; Whetstone et al., 1991; Whetter et al., 1990),

sucrose-gradient purified plasma from infected animals (Hartaningsih et al., 1994) or

recombinant protein production (Bird et al., 1993; Burkala et al., 1999; Burkala et al.,

1998). Other techniques have been used to monitor the immune response to bovine

lentivirus infections, including immunofluorescent assays (O'Rourke et al., 1988),

agar gel immunodiffusion (AGID) (Burki et al., 1992; Coggins et al., 1972;

Hartaningsih et al., 1994; Whetter et al., 1990) and radioimmunoprecipitation (Beyer

et al., 2001; Morin et al., 2003; O'Rourke et al., 1988). Western immunoblot assays

were more sensitive than AGID tests for detection of antibody to BIV (Whetstone et

al., 1991) and for JDV an ELISA was also more sensitive than AGID (Hartaningsih

et al., 1994).

Experimental infection of Bali cattle with JDV results in the production of antibodies

detectable by ELISA and WIB to the CA protein 6-11 weeks pi, although

occasionally animals can seroconvert as early as 2 weeks pi (Desport et al., 2009a;

Hartaningsih, 1997). The antibody response to JDV peaks 23-33 weeks pi and

persists beyond 59 weeks (Hartaningsih et al., 1994). As JDV has not been cultured

in vitro, there have been limited investigations of the neutralising antibody response

to JDV. There is only a single report available of a neutralising antibody response to

JDV, which relied on the infection of cattle to determine virus neutralisation. The

evidence from this study suggested that neutralising antibody to JDV developed only

after a prolonged period following recovery, and therefore that neutralising antibody

Page 49: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

39

did not seem essential in the recovery from acute Jembrana disease. Antibodies

capable of neutralising JDV were detected in cattle 4 and 28 months after cattle had

recovered from clinical disease and titres were low, in the range of 1:2 to 1:20

(Hartaningsih et al., 2001).

In experimental BIV infections, antibody develops earlier than in JDV infections.

Antibody to the CA protein can be detected as early as 14 days post infection (dpi)

with titres peaking 6-8 weeks pi. Multiple studies have confirmed the early CA

antibody response and determined that it persists for up to 2.5 years after initial

infection (Heaton et al., 1998; Suarez et al., 1995; Whetstone et al., 1990; Whetstone

et al., 1991) although there is a single report that found the CA response to BIV

infection decreased dramatically by 40 weeks pi (Isaacson et al., 1995). Antibodies

capable of neutralising BIV have been detected as early as 17 weeks pi and persist

for 44 months pi (Carpenter et al., 2000). Antibodies against BIV TM could be

detected 4 weeks pi, peaking 10-30 weeks pi and persisted for at least 50 weeks pi

(Scobie et al., 1999); in some cattle they were detected 4 years pi (Isaacson et al.,

1995). The antibody response to the SU protein of BIV was reported to take many

months to develop (Suarez et al., 1995).

The CA and TM proteins of BIV and JDV contain cross-reactive epitopes which has

made serological differentiation between infections with JDV and BIV impossible

(Desport et al., 2005; Kertayadnya et al., 1993; Lu et al., 2002). Attempts have been

made to develop reagents capable of differentiating between these infections but

these have been unsuccessful (Desport et al., 2005).

Page 50: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

40

Assays for the detection of lentivirus infections in ruminants

Small ruminant lentiviruses

Serological methods such as ELISA (using whole virus, recombinant proteins or

synthetic peptides as antigens), competitive ELISA (using monoclonal antibodies),

AGID, radio-immunoprecipitation and WIB are most commonly performed to detect

SRLV infection and there are a number of kits available commercially (Brinkhof et

al., 2007; Brodie et al., 1993; de Andres et al., 2005; Herrmann et al., 2003a; Pasick,

1998). It is important to note, however, that no “gold standard” method of diagnosis

exists (Brinkhof et al., 2007; de Andres et al., 2005).

Of a number of assays recently evaluated by Brinkhof and van Maanen (2007) for

the serodiagnosis of SRLV infections in sheep and goats, the best performing assay

was an ELISA. This assay involved the sensitisation of the solid phase with a

combination of recombinant VMV CA protein produced in E. coli and a TM derived

peptide. The performance of this assay may be due to the simultaneous detection of

antibodies against CA and TM. Antibodies to CA are produced early after infection

but decline once clinical signs appear and TM antibodies are produced later but

persist into the clinical phase. Therefore any assay which combines the detection of

antibodies to these 2 proteins could be expected to cover a greater proportion of the

infection period (Boshoff et al., 1997; Brinkhof et al., 2007).

A number of supplementary tests are often performed to confirm or resolve

indeterminate ELISA results, including AGID, WIB and radioimmunoprecipitation,

and these can be supplemented by PCR assays for the detection of proviral DNA or

viral RNA in PBMC or plasma (Barlough et al., 1994; Brinkhof et al., 2007; Brodie

et al., 1993; de Andres et al., 2005; Wagter et al., 1998; Zanoni et al., 1992).

Page 51: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

41

Large ruminant lentiviruses

Diagnosis of BIV infection plays a crucial role in controlling the spread of infection

and the pathogen-free preparation of vaccines prepared using cattle, for example tick

fever vaccines (Lew et al., 2004). Assays used for detecting BIV infection include

qPCR (Lew et al., 2004), nested PCR, cell culture syncytium assays, WIB assays

(Meas et al., 1998; Meas et al., 2000a; Snider et al., 2003b; Suarez et al., 1995;

Zhang et al., 1997a), and ELISA using recombinant CA and TM protein antigens

(Barboni et al., 2001; Burkala et al., 1999). Techniques to quantify proviral and viral

BIV loads in tissues have not been described.

All attempts to culture JDV in vitro have failed and it has not been possible to detect

JDV infections using virus isolation techniques (Kempster et al., 2002; Wilcox et al.,

1992). Two techniques have been described that enable the detection and

quantification of JDV virus load, the JDV gag quantitative Reverse Transcriptase-

PCR (qRT-PCR) assay and a JDV p26 antigen capture ELISA (Stewart et al., 2005).

The qRT-PCR technique was optimised with plasma samples taken from animals

experimentally infected with JDV, and it was found to be robust and sensitive, with

an apparent sensitivity 100-fold greater than that of a standard RT-PCR. The capture

ELISA was relatively insensitive when compared to the qRT-PCR, although it

provided an economical method for monitoring of virus in the absence of more

sensitive techniques (Stewart et al., 2005).

Page 52: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

42

Difficulties with serological testing

A number of factors have been reported to be critical in the interpretation of

serological assays. In lentivirus infections, one problem encountered is the delayed

nature of the antibody response. For example, in donkeys experimentally infected

with EIAV, the animals respond weakly and produce antibodies 42 dpi while horses

produce antibodies which can be detected as early as 16 dpi (Spyrou et al., 2003).

Hence, testing donkeys less than 42 dpi will yield a false-negative result. Problems

with delayed antibody responses have also occurred in CAEV (Brinkhof et al., 2007;

Rimstad et al., 1993) and JDV infections (Desport et al., 2009a; Hartaningsih et al.,

1994). Another problem in lentivirus infections is that antibody titres to viral proteins

may fluctuate between positive and negative after infection, though reasons for this

are unclear (de Andres et al., 2005).

Problems can occur in the detection of neutralising antibody in non-primate

lentivirus infections (Pozzetto et al., 1986; Sahu et al., 1994). Some North American

strains of VMV do not appear to induce neutralising antibodies (Zink et al., 1987). In

other VMV infections, the time required to develop detectable levels of neutralising

antibody after infection may vary considerably, from 12-14 dpi (Bird et al., 1993)

through to 1-3 months (Petursson et al., 1976). The titre of neutralising antibody

reported in response to lentivirus infections has also varied considerably in different

systems: in response to CAEV in goats it was reported to be of low titre and low

affinity (Kennedy-Stoskopf et al., 1986; Pisoni et al., 2007) as was the response of

horses to EIAV infection (O'Rourke et al., 1988) but the titre of neutralising antibody

in sheep in response to VMV infection was reported to be as high as 1:640 (Narayan

et al., 1978).

Page 53: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

43

Another important factor which should be considered in lentivirus serological tests is

cross-reactive epitopes on viral proteins, which can make serological differentiation

of antigenically related viruses difficult. Antigenic cross-reactivity between structural

proteins of lentiviruses, particularly the CA protein, is well known (Cheevers et al.,

1988; Gnann et al., 1987a). Cross-reactivity has been reported between the CA

protein of JDV and BIV (Burkala et al., 1998; Desport et al., 2005; Kertayadnya et

al., 1993), between the TM glycoprotein of BIV and JDV (Burkala et al., 1998) and

the SU glycoprotein of CAEV and VMV (Gogolewski et al., 1985; Valas et al.,

2000), among others.

Techniques to identify epitopes and distinguish between viral infections

Peptide mapping is a useful technique to identify specific epitopes that differ

between 2 viruses that cross-react antigenically (Valas et al., 2000; Van

Regenmortel, 1999b). The reactivity to peptides produced in this manner provides

information on the antigenic characteristics of similar viruses infecting an animal,

which permits differentiation between the virus infections (Gnann et al., 1987a;

Mordasini et al., 2006; Pisoni et al., 2007). Peptide mapping involves the generation

of a panel of overlapping peptides which cover the entire amino acid sequence of the

protein of interest. These peptides are chemically synthesised and used in immune

binding assays, usually in an ELISA format for high throughput, against a panel of

reference sera taken from natural and experimental infections (Ball et al., 1992;

Bertoni et al., 1994). Synthetic peptides have been used, for example, to identify

epitopes that differentiate HIV-1 and HIV-2 infections (Gnann et al., 1987a; Gnann

et al., 1987b).

Another technique to identify unique epitopes is phage display (Xiao et al., 2008).

This technique is made possible by the expression of peptide libraries on the surface

Page 54: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

44

of filamentous phage particles. The first step in the process involves the insertion of

one random oligonucleotide into one phage. The peptide encoded by the

oligonucleotide is then expressed within the pIII or pVIII coat protein of the

filamentous phage fd or M13 (D'Mello et al., 1999). These random phage display

libraries are subsequently screened for reactive epitopes with an antibody of interest.

The peptide sequence can be determined after amplifying the DNA from the selected

phage by PCR and sequencing techniques (D'Mello et al., 1999; Van Regenmortel,

1999a; Westwood, 2000; Williams, 2000).

Another technique suitable for defining epitopes is the use of protein expression

libraries. To construct expression libraries, DNA from a coding sequence of interest

is digested with DNase I to generate random DNA fragments with an approximate

average of 200 bp in length. These fragments, which can encode peptides, are ligated

into vector DNA (for example λgt11) whilst preserving the reading frame of a fusion

protein such as β-galactosidase. Ligations are packaged into Escherichia coli and the

cloned fragments are expressed as fusion proteins from the recombinant phages. The

random libraries are subsequently screened for reactive epitopes with an antibody of

interest. The peptide sequence can be determined after amplifying the DNA from the

selected phage by PCR and then sequencing this product (Bertoni et al., 2000;

Bertoni et al., 1994; Pancino et al., 1993; Van Regenmortel, 1999a).

One traditional method of defining epitopes is the limited fragmentation of proteins

by chemical cleavage, using cyanogen bromide or enzymatic digestion.

Fragmentation is then followed by immunoblotting of protein fragments (Westwood,

2000). Other methods that have been used include crystallographic analysis of

antigen-antibody complexes and the binding of anti-peptide antibodies to either

natural or chemically modified proteins (Van Regenmortel, 1999a).

Page 55: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

45

Lentivirus superinfection

Dual infection of an animal with 2 viruses can be the result of coinfection or

superinfection. Coinfection occurs when 2 viruses infect at or near the same time

prior to seroconversion. Superinfection is the infection of an animal (or a cell) by 2

genetically distinct viruses where the infection with one virus precedes infection with

the second virus, sometime after seroconversion (Blackard et al., 2002; Gottlieb et

al., 2004; Jurriaans et al., 2008).

A natural case of superinfection of goats with CAEV and VMV has recently been

documented (Pisoni et al., 2007). Several studies have reported superinfection of

HIV-1 infected individuals (Gottlieb et al., 2004; Jurriaans et al., 2008; Piantadosi et

al., 2007; Smith et al., 2006). Between 2002 and 2005, 16 cases of HIV-1

superinfection in humans were reported (Smith et al., 2005). Although this number

seems small, natural cases of superinfection have generated considerable interest as

they challenge the assumption that HIV-1 specific immune responses generated

against primary infection are protective against subsequent infection by different

strains of the same virus (Allen et al., 2003).

Benefits of superinfection - superinfection resistance

Experimental studies have shown that pre-infecting an animal with a relatively less

pathogenic virus protects against challenge with a second, more pathogenic

lentivirus, a situation referred to as superinfection resistance (Table 2.3). This has

been demonstrated in FIV (Terwee et al., 2008; VandeWoude et al., 2002), SIV

(Cranage et al., 1998; Nilsson et al., 1998; Stebbings et al., 2004) and heterologous

SHIV infections (Sealy et al., 2009).

Domestic species of cats that have been asymptomatically infected with non-

pathogenic lion lentivirus (FIVple) or puma lentivirus (FIVpco) have shown resistance

Page 56: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

46

to subsequent challenge with pathogenic FIV (Brown et al., 1994). All cats became

infected with the pathogenic FIV but prior exposure to FIVple or FIVpco ameliorated

the normal clinical effects of FIV infection: CD4+ cell depletion was reduced and, in

some cases, plasma and PBMC FIV loads were reduced (Terwee et al., 2008;

VandeWoude et al., 2002).

Infection with live attenuated SIVmacC8 of cynomolgus macaques ameliorated the

effects of subsequent infection with pathogenic wild-type SIVmacJ5 (Stebbings et al.,

2004), its derivative SIVmac220 (Cranage et al., 1998) and SIVsm (Nilsson et al., 1998).

This was shown by a significant decrease of cell-associated virus and plasma viral

RNA loads (Cranage et al., 1998; Stebbings et al., 2004) and negative virus isolation

in a proportion of animals (Nilsson et al., 1998). Indian rhesus macaques infected

with SIVmacC8 also resisted superinfection with the virulence-reverted form of

SIVmacC8 (Sharpe et al., 1997). Likewise, Indian rhesus macaques infected with

SIVmacGX2 were completely protected against challenge with SIVmac220 (Sharpe et al.,

2004) and macaques infected with SIVmac239∆nef were protected against challenge

with SIVmac251 (Connor et al., 1998; Daniel et al., 1992).

Superinfection resistance holds promise as a way to ameliorate the effects of

lentivirus infection and disease, in effect to act as a potential vaccine. It has been

proposed that if the mechanism of protection conferred could be better understood,

then a safe and effective HIV vaccine, for example, could be developed (Cranage et

al., 1998; Stebbings et al., 2004). The effect of challenge with a non-pathogenic virus

on the course of infection with a pathogenic virus also offers an opportunity to

examine host-virus and virus-virus interactions and their effect on pathogenicity and

resistance to virulent lentivirus infections (VandeWoude et al., 2003).

Page 57: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

47

Mechanisms of superinfection resistance

The mechanisms responsible for protection against superinfection are not well

defined. Protection has not been directly correlated with humoral or cellular immune

responses including virus neutralising activity (Connor et al., 1998; Cranage et al.,

1998; Daniel et al., 1992; Nilsson et al., 1998; Sharpe et al., 1997; Stebbings et al.,

2004; Stebbings et al., 2002; VandeWoude et al., 2002) and in some cases protection

is not dependent on challenge-driven expansion of immunodominant epitope-specific

CD8+ T-cells (Sharpe et al., 2004). While CD8+ T-cells are important for control of

primary viraemia, they do not seem to play a central role in protection against

superinfection (Stebbings et al., 2005). Non-immunological phenomena such as virus

interference or antiviral factors such as CD8 suppression factors induced by defective

particles have been suggested as playing a role in superinfection resistance (Cranage

et al., 1998; Stebbings et al., 2004; Stebbings et al., 2002). Virus interference has

been detected in vitro when cell cultures were infected with a retrovirus and were

relatively resistant to infection by a related retrovirus; the phenomenon occurs only

when both viruses share the same receptor and results from a restricted penetration

into the cell (Corbin et al., 1993).

Adverse consequences of superinfection

While the positive effects of superinfection, namely resistance, have been well

described, superinfection or mixed infection of animals with 2 viruses has resulted in

potentially adverse effects. Goats infected naturally with both CAEV and VMV were

shown to contain chimeric viruses with CAEV-VMV envelope glycoproteins; this

was expected to have dramatic effects on the species specificity of the viruses and

their capacity to cross species barriers (Pisoni et al., 2007).

Page 58: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

48

Table 2.3. Experimental superinfection studies conducted in a range of lentivirus animal model systems.

Model system Primary virus infection

Secondary pathogenic virus strain

Time between primary and secondary infections (weeks)

Effect on pathogenic virus Reference

FIVpco FIV-C 4 Reduced CD4+ T-cell depletion. (Terwee et al., 2008)

FIVpco or FIVple FIV-B 27 Reduced CD4+ T-cell depletion, reduced plasma and PBMC FIV load.

(VandeWoude et al., 2002) FIV in domestic cat

FIVPetaluma FIV-M2 28 Reduced total viral load, reduced CD4+ T-cell depletion.

(Pistello et al., 1999)

SIVmac251 or SIVsmE660

Reciprocal infection

Reduction in peak viraemia, amelioration of infection.

(Yeh et al., 2009)

SIV in rhesus macaques

SIVmacGX2 (nef-disrupted)

SIVmac220 89 or 122 Complete resistance (determined by negative virus isolation).

(Sharpe et al., 2004)

SIVmacC8 SIVmac32H/L28 3 or 20 Reduction of viral RNA and DNA load.

(Berry et al., 2008) SIV in cynomolgus macaques Attenuated

SIVmacC8 SIVmacJ5 3

Reduced cell-associated virus loads, reduced plasma virus load.

(Stebbings et al., 2004)

Page 59: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

49

Chapter 3: Attempts to detect Bovine immunodeficiency virus infection in

Bali cattle in Indonesia with a PCR-based assay

Summary

Attempts were made to provide evidence for the occurrence of BIV in cattle in Indonesia.

One hundred and seventy one genomic DNA and serum samples were taken from Bali

cattle in the Bangli and Tabanan regions on the island of Bali. Genomic DNA samples

extracted from PBMC were screened for the presence of BIV or JDV proviral DNA using

both real time and conventional PCR methods and direct sequencing of any amplified

products to confirm their identity. Serum samples were screened for antibodies against

JDV using a range of antigens in a WIB or ELISA format and 21 of the 171 animals were

identified as being seropositive by a positive WIB reaction with the p26 CA protein of

JDV and at least one other positive serological test. BIV proviral DNA was not detected in

any of the cattle but JDV proviral DNA was detected in 12 animals, only one of which

was seropositive.

Page 60: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

50

Introduction

Two bovine lentiviruses are suspected to circulate in the Bali cattle population of

Indonesia. The presence of Jembrana disease virus (JDV) is well documented and both

the disease and antibodies to the virus have been detected in cattle on the islands of Bali,

Sumatra and Java (Hartaningsih et al., 1993). The disease also now occurs in all

Kalimantan provinces in Indonesian Borneo (Hartaningsih, personal communication).

Despite the widespread distribution of Bali cattle in the eastern islands of Indonesia,

clinical Jembrana disease has not been reported in these areas. However, there are reports

of JDV antibody-positive cattle in some of the regions of Indonesia that are free of clinical

Jembrana disease, including on the island of Sulawesi (Desport et al., 2005), suggesting

the presence of a second non-pathogenic virus that is antigenically related to JDV,

possibly BIV. Serological evidence was also presented for the presence of a BIV-like

virus in Bali cattle in Bali where JDV is endemic (Barboni et al., 2001) although this has

not been confirmed by virus isolation.

The objectives of the investigations reported in this thesis were to attempt confirmation of

the presence of non-pathogenic BIV-like viruses in Bali cattle in Bali, to develop

methodology for the detection and differentiation of infection by these viruses and JDV,

perhaps enabling the determination of the distribution of each virus in Indonesia, and to

investigate the interaction between the 2 viruses in infected Bali cattle. Ideally, these

investigations required the isolation of the non-pathogenic BIV-like virus that is reputedly

present in Indonesia, that would then enable its experimental inoculation into animals not

only to determine the effects of these viruses in Bali cattle but also for the production of

reagents. Use of the local non-pathogenic virus would eliminate the need to import an

Page 61: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

51

exotic strain of BIV for these investigations. Unsuccessful attempts were made previously

to isolate a non-pathogenic bovine lentivirus from cattle in Sulawesi: blood samples were

obtained from antibody-positive Bali cattle in Sulawesi, the samples were transported by

air to Bali and inoculated into Bali cattle on arrival. The inoculated cattle did not

seroconvert to BIV or JDV, suggesting the virus was not present in the inoculum. Further

attempts to detect the Sulawesi virus were abandoned due to political unrest in the area

from where cattle had been sampled and the expense involved (Hartaningsih, personal

communication).

This Chapter describes the results of attempts to confirm the report of Barboni et al.

(2001) that Bali cattle on the island of Bali were infected with BIV, and from which

attempts to isolate the virus could be made.

Materials and methods

Field samples

Peripheral blood samples were taken from 171 Bali cattle (Bos javanicus) in 2 districts

(Bangli and Tabanan) on the island of Bali, Indonesia. They were collected from the

jugular vein, into sterile 10 ml vacutainer tubes containing 15% EDTA (BD) for

extraction of genomic DNA, and into plain tubes for preparation of serum samples for

serological tests.

Preparation of PBMC genomic DNA

After collection into vacutainer tubes containing EDTA, the Bangli samples were

centrifuged (900 g for 10 min at 4°C) and the buffy coat was subsequently transferred into

10 ml tubes containing 6 ml distilled H2O, then mixed by inverting 3 times. Three ml of

2X PBS was then added and mixed by inverting 3 times after which tubes were

Page 62: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

52

centrifuged again (250 g for 10 min at 4°C). The resulting supernatant was discarded and

the pellet washed twice with 10 ml PBS before the pellet was gently resuspended in 1 ml

Hank’s medium supplemented with 20% v/v heat inactivated foetal calf serum and 6% v/v

DMSO (Sigma) and stored in 200 µl aliquots at -20°C until required. Genomic DNA was

extracted from PBMC using the FlexiGene DNA Kit (Qiagen) according to the

manufacturer’s instructions and stored at -20°C until used. The concentration of PBMC

genomic DNA in each sample was determined using a spectrophotometer (Nanodrop ND-

1000).

After collection into vacutainer tubes, the Tabanan blood samples were centrifuged (900 g

for 10 min at 4°C) and the buffy coat was collected and mixed with 2 ml of PBS, then

overlayed onto 6 ml Ficoll (Amersham) in a sterile 10 ml tube and centrifuged at 400 g for

20 min at 4°C. The cells at the interphase were collected and washed 3 times in PBS by

mixing with PBS followed by centrifugation (400 g for 20 min at 4°C). The washed cells

were then resuspended with 1 ml PBS and stored at -20°C until required. Genomic DNA

was extracted from the purified PBMC using the QIAamp DNA Mini Kit (Qiagen)

according to the manufacturer’s instructions and the DNA was stored at -20°C until used.

The concentration of PBMC genomic DNA in the samples was determined using a

spectrophotometer.

Verification of DNA quality

Extracted DNA samples were initially tested by PCR using gene-specific glyceraldehyde

3-phosphate dehydrogenase (GAPDH) primers to test for integrity of the DNA. If no PCR

product was obtained with these primers then the sample was discarded. The GAPDH

primers used were as described previously (Mohan et al., 2001) and the reaction consisted

Page 63: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

53

of 200 ng of PBMC genomic DNA, 1X PCR buffer, 1.25 mM MgCl2, 0.2 mM of each

dNTP, 0.8 mM of each primer (GAPDH F,

CCTTCATTGACCTTCACTACATGGTCTA, GAPDH R,

GCTGTAGCCAAATTCATTGTCGTTACCA; Invitrogen), 0.687 U Taq polymerase, and

ultrapure water to a final volume of 25 µl. All reagents were sourced from Fisher Biotec

unless otherwise stated. Thermal cycling conditions were 1 cycle of 94°C for 3 min, 35

cycles at 94 °C for 30 s, 60°C for 30 s and 72°C for 45 s, a final extension step of 72°C

for 7 min and they were then held at 14°C in a Bio-Rad thermal cycler.

qPCR for detection of JDV proviral DNA

The JDV proviral DNA genome copy number was quantified using a DNA plasmid-based

standard curve derived using JDV plasmid clone #139 as a template as previously

described (Stewart et al., 2005). The JDV sequence within the pT7T3 vector spanned JDV

nucleotides 19 to 2881 (U21603). The qPCR assay specifically amplified 118 bp in JDV

gag. All reactions consisted of 1X iQ Supermix (100 mM KCl, 40 mM Tris-HCl (pH 8.4),

1.6 mM dNTPs, 50 U/ml of iTaq DNA polymerase, 6 mM MgCl2, undefined stabilisers,

Bio-Rad), 0.6 mM of each primer (gag1f-GGGAGACCCGTCAGATGTGGA, gag1r-

TGGGAAGCATGGACAATCA; Invitrogen) prepared as described previously (Stewart et

al., 2005), 0.1 µM fluorogenic probe (Geneworks), 200 ng of extracted PBMC genomic

DNA and made up to a final volume of 10 µl using ultrapure water (Bio-Rad). Thermal

cycling conditions were the same as previously described (Stewart et al., 2005), except

that the reverse transcriptase step was omitted.

Page 64: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

54

Conventional PCR for detection of JDV proviral DNA

A number of DNA samples which were tested by qPCR but provided results below the

cut-off for a positive result but where the results were clearly greater than that for the

JDV-negative control DNA, were re-analysed using conventional PCR. The primer pair

for detecting JDV proviral DNA by conventional PCR (JDV 1:

GCAGCGGAGGTGGCAATTTTGATAGGA, JDV 3:

CGGCGTGGTGGTCCACCCCATG) were located within the gag open reading frame

and specifically amplify a 360 bp fragment (Desport et al., 2007). Reactions conditions

consisted of 1X buffer, 1 mM MgCl2, 0.2 mM of each dNTP, 0.88 mM of each primer

(Invitrogen), 1.374 U Taq polymerase, 400 ng PBMC genomic DNA and made up to a

final volume of 50 µl with ultrapure water. Unless otherwise stated, all reagents were from

Fisher Biotec. Thermal cycling conditions were the same as those described (above) for

the GAPDH primer pair. Reaction conditions in the second round of amplification, where

necessary, were the same as the first except 1 µl of first round PCR product was added

into 25 µl reaction volumes.

Sequence analysis of PCR products

Direct DNA sequencing was performed to confirm and compare the proviral DNA

detected in the 2 cattle populations. If one band was produced by conventional PCR

amplification, the PCR product was purified using a PCR Purification Kit (Qiagen)

according to the manufacturer’s instructions. If multiple bands were produced, the PCR

product of the correct size was excised from the agarose gel and purified using a Gel

Extraction Kit (Qiagen) according to the manufacturer’s instructions. Ten ng of purified

product was sequenced using 1 µl of Big Dye Terminator (Applied Biosystems), 1.5 µl of

Page 65: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

55

Big Dye Terminator sequencing buffer (Applied Biosystems), 3.2 pmoles of JDV 1 or

JDV 3 primer and made up to a volume of 10 µl using ultrapure water (Fisher Biotec).

The sequencing reaction consisted of a 2 min hold at 96°C and 25 cycles of 96°C for 10 s,

60°C for 30 s and 60°C for 4 mins. The sequencing reaction was purified by ethanol

precipitation according to the protocol supplied by Applied Biosystems. Samples were

then sequenced using an ABI 3730 48 capillary machine at the State Agricultural

Biotechnology Centre, Murdoch University.

Sequences were edited using Chromas Lite and aligned using the ClustalW program

(http://www.ebi.ac.uk/Tools/clustalw2/index.html). Phylogenetic analysis was performed

using Phylogeny.fr (http://www.phylogeny.fr/version2_cgi/index.cgi) and phylogenetic

trees were edited using MEGA (http://www.megasoftware.net/index.html).

qPCR for BIV proviral DNA

All animals were tested for the presence of BIV proviral DNA using a qPCR assay as

described previously (Lew et al., 2004), with the following modifications: 1X iQ

Supermix (Bio-Rad), 100 ng of each primer (BIVF1-

ACAAAAACTACGGGAATACCCTACA, BIVR1-

TCTTTTAGATCTCTGTGGGCTCTTTC; Invitrogen), 0.1 mM of probe (6FAM

CCACAATCCCAGGGAGT; Applied Biosystems) and 200 ng of PBMC genomic DNA.

The reaction volume was made up to 10 µl using ultrapure water (Fisher Biotec). The

qPCR assay specifically amplified 73 bp in BIV pol. The limit of quantification of the

assay was 20 copies per reaction (determined by amplification of specific known

quantities of cloned viral DNA).

Page 66: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

56

Serological tests

Serum samples were tested for the presence of antibodies to JDV using ELISA or WIB

with a range of antigens. The assays were conducted by Dr J. Lewis (this laboratory)

using antigens that included a recombinant JDV p26-his, recombinant fused JDV

p26/TM-his, JDV TM peptide and plasma-derived JDV native antigen (kindly provided

by N. Hartaningsih). The WIB assay that utilised native JDV antigen derived from the

plasma of infected cattle was conducted as described previously by Hartaningsih et al.

(1994); results were recorded as positive if there was reactivity with the 26 kDa CA

protein. All other ELISA and WIB assays were conducted and interpreted as described

previously (Lewis, 2009).

The recombinant JDV p26-his construct expressed full-length JDVTab87 CA (Barboni et

al., 2001). The construct was kindly supplied by Dr. Margaret Collins, transformed into

BL21 (DE3) E. coli for protein expression and was purified using Ni-NTA agarose resin

in chromatography columns. The fused p26/TM construct was generated in a previously

described manner (de Andres et al., 2005; Rosati et al., 2004) whereby JDV capsid was

fused directly to the putative TM principle immunodominant domain (PID) epitope,

transformed into E. coli BL21 and purified as described for the recombinant JDV p26-his.

The JDV TM peptide ELISA was prepared as previously described (Ditcham et al., 2009)

and encompassed the PID of JDV TM (Barboni et al., 2001).

As there was no “gold standard” test for JDV antibody detection, samples were considered

“positive” when a positive result was obtained in WIB using the plasma-derived JDV

antigen and at least one other assay was also positive. Antibody to JDV and BIV cannot

be differentiated due to the presence of numerous cross-reactive epitopes on the CA

Page 67: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

57

(Desport et al., 2005; Kertayadnya et al., 1993) and TM proteins (Burkala et al., 1998).

Therefore, the use ofthese antigens in WIB assays will equally detect antibody to both

JDV and BIV.

Results

Serology

Twenty one of the 171 cattle from which DNA was also analysed were seropositive for

antibody to the p26 CA of JDV by WIB utilising native JDV antigen and at least one other

assay (Table 3.1).

PCR

The GAPDH gene was detected by PCR in PBMC genomic DNA samples from 171 cattle

and these were deemed to have DNA of sufficient quality to test using qPCR or

conventional PCR. These 171 samples were then screened for the presence of JDV

proviral DNA by PCR and JDV-specific PCR products were detected in 12 of the 171

PBMC genomic DNA samples tested (Table 3.1). The sequences had between 99 and

100% homology with the reference JDVTab87 strain. The sequences generated are shown in

Figure 3.1.

With the exception of one animal (Tabanan Y26), all cattle in which JDV proviral DNA

was detected were seronegative (Table 3.1). In the 11 seronegative and PCR positive

cattle, 10 (83%) were negative in every serological assay and one (Bangli 35) was positive

in one of the 5 serological assays.

BIV proviral DNA was not detected in any of the 171 PBMC genomic DNA samples

tested.

Page 68: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

58

Table 3.1. Results of PCR diagnostic assays and serological assays (ELISA and WIB) in

171 samples taken from Bali cattle (B. javanicus) in the Bangli and Tabanan region of

Bali, Indonesia. The results shown are those from 52 cattle where any assay provided a

positive result, and where samples provided negative results in all assays the results are

not shown. Italics indicate the sample was positive by both JDV PCR and serology.

Animal

identification

Age

(years)a

BIV

PCR

JDV

PCRb Antigen and serological assay

Final

serological

resultc

CA

ELISA

CA

WIB

Fused

CA/TM

ELISA

Fused

CA/TM

WIB

Native

antigen

WIB

Bangli 5 - - - - + - - -

Bangli 9 - + - - - - - -

Bangli 15 - - - - + - - -

Bangli 17 - - - - + - - -

Bangli 18 - - - - + - - -

Bangli 20 - - - - + - - -

Bangli 27 - + - - - - - -

Bangli 35 - + - - + - - -

Bangli 50 - + - - - - - -

Tabanan R3 5 - - + + - + + +

Tabanan R4 4 - - + + - + + +

Tabanan R9 6 - - + + - + + +

Tabanan R10 6 - - + + + + + +

Tabanan R12 6 - - - + - + - -

Tabanan R16 5 - - + + - + + +

Page 69: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

59

Animal identification

Age (years)a

BIV PCR

JDV PCRb Antigen and serological assay

Final serological

resultc

CA ELISA

CA WIB

Fused CA/TM ELISA

Fused CA/TM

WIB

Native antigen WIB

Tabanan R17 4 - - - + - + - -

Tabanan R18 4 - - + + - + + +

Tabanan R21 4 - - + - - - - -

Tabanan R26 5 - - + - - + + +

Tabanan R28 8 - - + + - + + +

Tabanan R33 10 - - + - + + + +

Tabanan R39 7 - - - - - + - -

Tabanan R43 6 - - + + - + + +

Tabanan R46 3 - - - - - + - -

Tabanan R48 5 - - - - - + - -

Tabanan Y5 4 - - + - - - + +

Tabanan Y15 1.6 - - - - - + - -

Tabanan Y21 6 - - + + - - + +

Tabanan Y22 5 - - + + - + + +

Tabanan Y26 5 - + + + + - + +

Tabanan Y31 7 - - + + - - + +

Tabanan Y43 8 - - + + - - + +

Tabanan Y45 7 - - + + + - - -

Tabanan B5 5 - + - - - - - -

Tabanan B6 4 - - + - - + + +

Tabanan B11 8 - - - + - - - -

Page 70: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

60

Animal identification

Age (years)a

BIV PCR

JDV PCRb Antigen and serological assay

Final serological

resultc

CA ELISA

CA WIB

Fused CA/TM ELISA

Fused CA/TM

WIB

Native antigen WIB

Tabanan B16 9 - - - - - + - -

Tabanan B26 8 - - + - + + + +

Tabanan B35 2 - - + - + - - -

Tabanan B37 7 - - + - + + + +

Tabanan G1 12 - - + - + - + +

Tabanan G4 7 - + - - - - - -

Tabanan G6 7 months - + - - - - - -

Tabanan G7 7 months - - - - + - - -

Tabanan G8 7 - + - - - - - -

Tabanan G11 6 months - + - - - - - -

Tabanan G15 10 - - + - - - - -

Tabanan G16 14 - - + - + - + +

Tabanan G26 4 - - - - + - - -

Tabanan G29 3 - + - - - - - -

Tabanan G31 5 - + - - - - - -

Tabanan G49 8 - - + - + - - +

Total 0 12 26 17 18 21 21 21

a Age data not available for Bangli cattle. b Positive status given where a positive result was obtained in the qPCR or conventional PCR assay and confirmed by direct sequencing. c Positive status given where a positive result was obtained with the JDV plasma-derived antigen WIB and at least one other serological assay.

Page 71: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

61

Bangli 27 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCAGAAAAGTATTGGGAAGCATGG 60 Bangli 35 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCAGAAAAGTATTGGGAAGCATGG 60 Bangli 50 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCAGAAAAGTATTGGGAAGCATGG 60 G31 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCAGAAAAGTATTGGGAAGCATGG 60 Y26 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCAGAAAAGTATTGGGAAGCATGG 59 G29 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCAGAAAAGTATTGGGAAGCATGG 60 G11 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCAGAAAAGTATTGGGAAGCATGG 60 G8 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCAGAAAAGTATTGGGAAGCATGG 60 G6 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCAGAAAAGTATTGGGAAGCATGG 60 B5 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCAGAAAAGTATTGGGAAGCATGG 60 G4 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCAGAAAAGTATTGGGAAGCATGG 60 Bangli 9 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCAGAAAAGTATTGGGAAGCATGG 60 JDV/Tab 87 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCAGAAAAGTATTGGGAAGCATGG 60 JDV/Pul 01 CTGCTGGCGGGCCACCACCCAGAAAATTCAGATATGGCCGAAAAGTATTGGGAAGCATGG 60 BIV127 CTGCTGGCGGGGTACAAACCAGAGAGTACAGAAACGGCCCTAGGATATTGGGAGGCCTTT 60 ********** ** * ***** * * **** * *** * ******** ** * Bangli 27 ATAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 120 Bangli 35 ATAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 120 Bangli 50 ATAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 120 G31 ACAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 120 Y26 ACAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 119 G29 ACAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 120 G11 ACAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 120 G8 ACAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 120 G6 ACAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 120 B5 ACAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 120 G4 ACAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 120 Bangli 9 ACAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 120 JDV/Tab 87 ACAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 120 JDV/Pul 01 ACAATCAGAGAAAGGGAGTCCCAAAAGGAGGAGGAAGGAGAAATTACCAGCATCTACCCA 120 BIV127 ACATATAGAGAAAGGGAGGCCAGAGCTGATAAGGAAGGCGAAATTAAGAGTATTTACCCT 120 * * ************ ** * ** ******* ******* ** ** ***** Bangli 27 CAACTTAGAAAGAACT------------------------------------ 136 Bangli 35 CAACTTAGAAAGAACT------------------------------------ 136 Bangli 50 CAACTTAGAAAGAACT------------------------------------ 136 G31 CAACTTAGAAAGAACT------------------------------------ 136 Y26 CAACTTAGAAAGAACT------------------------------------ 135 G29 CAACTTAGAAAGAACT------------------------------------ 136 G11 CAACTTAGAAAGAACT------------------------------------ 136 G8 CAACTTAGAAAGAACT------------------------------------ 136 G6 CAACTTAGAAAGAACT------------------------------------ 136 B5 CAACTTAGAAAGAACT------------------------------------ 136 G4 CAACTTAGAAAGAACT------------------------------------ 136 Bangli 9 CAACTTAGAAAGAACT------------------------------------ 136 JDV/Tab 87 CAACTTAGAAAGAACT------------------------------------ 136 JDV/Pul 01 CAACTTAGAAAGAACT------------------------------------ 136 BIV127 TCCCTAACACAGAACACACAGAATAAGAAGCAGACATCGAATCAGACAAACA 172 ** * * *****

Figure 3.1. Sequence alignment of gag nucleotide sequences from 12 animals with JDV

taken from Bangli and Tabanan. Reference sequences JDVTab87 (accession number

U21603), JDVPul01 (accession number DQ229295) and BIV127 (accession number

NC_001413.1) are included for comparison. Sequences were aligned using ClustalW2.

“*” indicates all nucleotides are identical in that column.

Page 72: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

62

Discussion

Although JDV proviral DNA was detected in the PBMC of 12 of 171 cattle examined,

which included cattle from 2 adjacent districts of Bali, BIV proviral DNA was not

detected in any of the animals tested. The result does not provide proof of the absence of

BIV in the Bali cattle population of Bali but it also does not provide any support for the

serological evidence reported by Barboni et al. (2001) that cattle within Bali are infected

not only by JDV but with a second antigenically related but presumably non-pathogenic

bovine lentivirus. The methodology used by Barboni et al. (2001) involved the production

of recombinant BIV CA and JDV CA and their use in a WIB format or a combination of

the recombinant CA with a BIV or JDV TM peptide in an ELISA format. Sera were

screened initially using the JDV antigens, then JDV antibody negative sera were screened

using the BIV antigens; if the serum reacted in the second set of assays the sample was

declared BIV seropositive and JDV seronegative. Given the high level of cross-reactivity

between JDV and BIV CA and TM antigens (Desport et al., 2005; Kertayadnya et al.,

1993), including the cross-reactivity of the antigens used in the study, these findings

should be interpreted with considerable caution. Results from this laboratory (Desport et

al., 2005) have shown that the reagents used by Barboni et al. (2001) would not

differentiate between JDV and BIV antibody.

The attempt to identify cattle infected with BIV by the detection of proviral DNA was

unsuccessful but not surprising. It is difficult to detect BIV in naturally infected cattle and

with the exception of the reports describing 3 successful attempts in the USA and Japan

(Suarez et al., 1993; Van der Maaten et al., 1972) and one from Japan (Meas et al., 1998),

this has not been reported, although there have been limited reports of the detection of

Page 73: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

63

proviral BIV DNA in naturally infected cattle (Lew et al., 2004; Meas et al., 1998; Meas

et al., 2000b; Snider et al., 2003b). In the current study, both antibody-positive and

antibody-negative cattle were examined, and 2 specific PCR assays were used. Either both

assays lacked the sensitivity to detect the level of provirus present in cattle, or maybe

BIV-like viruses are present in Bali but were not present in the populations sampled, or

possibly BIV-like viruses do not occur in the cattle population of Bali. Modifications to

the PCR assays, such as degenerate primers and less stringent PCR reaction conditions,

may have assisted in the detection of proviral DNA. However, based on previous

experience in this laboratory, alterations such as these have a tendency to produce false

positive results. Further investigation of the pathogenesis of BIV infection in Bali cattle is

needed to examine the kinetics of virus replication and persistence after infection and the

optimal time for sampling.

JDV proviral DNA was detected in 12 animals sampled, suggesting recent infection, even

though there was no evidence of clinical Jembrana disease immediately preceding and

following the sampling. This is the first reported detection of JDV in clinically normal

cattle. The lack of variation between the PCR amplicons suggests a common virus strain

was circulating in the cattle, although the lack of variation is also consistent with previous

studies reporting a high level of nucleotide conservation in gag sequences of JDV detected

in Bali (Desport et al., 2007) and they may not have arisen from a single source. All the 21

JDV-seropositive cattle were 4 years or older indicating minimal transmission of JDV

between the cattle in the preceding 4 years, and this is consistent with the lack of reports

of Jembrana disease outbreaks in Bali during this period (Hartaningsih, personal

communication).

Page 74: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

64

Proviral DNA positive-seronegative animals are rare but not uncommon after natural

lentivirus infections, and have been reported in cattle infected with BIV (Meas et al.,

2000a), cats infected with FIV (Dandekar et al., 1992) and sheep infected with VMV

(Leginagoikoa et al., 2009). These observations, and the detection of JDV provirus in 12

cattle, only one of which was seropositive, highlights the difficulty associated with

conclusively detecting natural lentivirus infections. There are a number of possible

reasons for the lack of concordance between serological and PCR assays. Firstly, the

antibody response to JDV is delayed and has been reported to be detectable only from 11

weeks post infection (Hartaningsih et al., 1994), hence serological assays would only

detect antibodies in the period after this time and this is a period of active virus replication

(Stewart et al., 2005). The detection of JDV proviral DNA in seronegative animals

suggests recent infection with JDV, prior to seroconversion. Secondly, there are a

proportion of animals infected with JDV that do not mount an antibody response to the

CA antigen (Desport et al., 2009a; Ditcham et al., 2009). These animals are referred to as

atypical responders and account for 15% of all animals experimentally infected but it

seems unlikely that this would account for the lack of antibody in 11 of the 12

PCR-positive animals detected. Thirdly, JDV proviral DNA levels in the PBMC are

hypothesised to persist at very low levels in naturally infected cattle, similar to SRLV

infections (de Andres et al., 2005). JDV proviral DNA has been readily detectable in

experimentally infected cattle, even many months after infection but is normally detected

only with difficulty in naturally infected cattle (Desport, personal communication).

Further studies are required to characterise the response of Bali cattle to inoculation with

BIV, including whether the animals can become actively infected and whether the virus

Page 75: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

65

will persist in the animals over time. It will also be of interest to determine if and at what

level BIV proviral DNA and viral RNA loads occur. The development of reagents that are

able to distinguish between BIV and JDV infections in an ELISA will help in the

identification of BIV infections and in Indonesia may clarify discrepancies between

genomic and antibody based assay results.

Page 76: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

66

Chapter 4: Bovine immunodeficiency virus produces a transient

viraemic phase soon after infection in Bos javanicus

Summary

Infection of Bali cattle in Indonesia with a non-pathogenic bovine lentivirus similar

to BIV is suspected but efforts to detect the virus have been unsuccessful. To define

the kinetics of BIV infection and seroconversion in Bali cattle and determine the

optimal time for sampling for detection of virus in infected cattle, 13 cattle were

infected with the R29 strain of BIV and monitored for up to 65 days. No clinical

signs were observed in the infected cattle following infection. Proviral DNA was

detected in PBMC from 4-60 days with peak titres 20 dpi. There was a transient

viraemia from 4 to 14 dpi with a maximum titre of 1 x 104 genome copies/ml plasma.

An antibody response to the TM glycoprotein commenced 12 dpi but an antibody

response to the CA protein was detected in one animal only and not until 34 dpi. The

results indicated that detection of BIV in infected Bali cattle is similar to B. taurus

with levels of proviral DNA detectable during the early stage of infection. Based on

these results, a CA based serological assay would not identify the majority of

infected cattle.

Page 77: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

67

Introduction

Bali cattle are particularly susceptible to JDV and develop an acute disease process

soon after infection. The acute disease is characterised by a transient febrile

response, enlargement of superficial lymph nodes, high virus titres in the plasma and

a number of haematological changes including leucopenia and thrombocytopenia.

The case fatality rate is about 21% and recovered animals are resistant to re-

challenge with the virus (Desport et al., 2009a; Soeharsono et al., 1990; Soesanto et

al., 1990). In contrast, breeds such as B. taurus (Fresian cattle) and B. indicus

(Ongole cattle) develop a mild febrile response but no other clinical signs of disease

(Soeharsono et al., 1990).

The effects of BIV in Bali cattle are unknown but the pathogenesis of BIV in

B. taurus has been investigated by several groups. Many of the studies that were

undertaken assumed that the virus would produce effects long after infection, akin to

many other lentiviruses, and they therefore followed the infections for long time

intervals. In experimentally infected B. taurus, the original R29 isolate caused no

major clinical signs in the period up to 27 months after infection (Flaming et al.,

1993; Zhang et al., 1997b) although subclinical changes in experimentally infected

cattle were reported by others. Subclinical changes reported have included

lymphocytosis and follicular hyperplasia (Carpenter et al., 1992) and immune

suppression at 3-7 weeks post-infection (Zhang et al., 1997b). Other isolates of BIV

(FL491 and FL112) were reported to cause a transient increase in PBMC (Suarez et

al., 1993). The FL112 isolate caused a transient B-cell lymphocytosis that peaked 14

dpi (Whetstone et al., 1997) and is also reported to cause lymphadenopathy and non-

suppurative meningoencephalitis 12 months post-infection (Munro et al., 1998).

Serological studies have also reported no major associations with significant clinical

Page 78: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

68

changes but one study based in North America found associations between BIV and

decreased milk yield in dairy cattle (McNab et al., 1994) while another reported

marked weight loss with frequent and severe concurrent infections (Snider et al.,

2003b).

This Chapter reports the experimental infection of Bali cattle with BIV that was

conducted with 2 objectives. First, to determine if Bali cattle were more susceptible

to infection with BIV than B. taurus, similar to their greater susceptibility to JDV

than other cattle species (Soeharsono et al., 1995a). Second, to determine the kinetics

of virus replication and persistence of BIV and the development of the antibody

response after infection, so as to provide insights into the optimal periods in which to

sample naturally infected cattle to detect the virus. The experiments were conducted

with the R29 strain of BIV as attempts to detect BIV in Indonesian cattle were not

successful (Chapter 3).

Materials and methods

Animals

Nineteen cattle were obtained from Nusa Penida, an island adjacent to Bali where

Jembrana disease has never been detected and the cattle have been consistently

negative for antibody to JDV and BIV. The cattle were housed indoors as previously

described (Soeharsono et al., 1990). Six weeks prior to infection with BIV the cattle

were vaccinated against Bovine viral diarrhoea virus using Pestigard (Pfizer) twice

at 0 and 4 weeks apart as the FBL cell culture was contaminated with BVDV. BVDV

is a common contaminate of BIV cell cultures and foetal bovine serum (Makoschey

et al., 2003).

Page 79: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

69

Virus

BIV-R29 was obtained from J Brownlie and M Collins, Royal Veterinary College,

UK, passaged in primary bovine foetal lung (BFL) cell cultures grown in RPMI

medium (Invitrogen) supplemented with 10% foetal bovine serum (Thermo

Scientific) and antibiotics in 75 cm2 flasks (Nunc). For inoculation into cattle,

infected cells exhibiting marked syncytium formation were scraped from the surface

of flasks 24 h after infection and the cells suspended in RPMI medium. The titre of

the virus was retrospectively estimated by titration in BFL cell cultures and the total

inoculum administered to each animal determined to be 1.38 x 106 50% tissue culture

infectious doses (TCID50).

Experimental infection and sampling of cattle

The infectious inoculum was divided into 2 equal amounts, one administered

intravenously and the other subcutaneously. Seven virus infected cattle (CB169-

CB175) were monitored for 42 days after infection and 6 (CB177-CB182) for 65

days after infection. An additional 6 cattle (CB183-CB189) were inoculated with an

equivalent volume of uninfected BFL cells in RPMI medium and monitored for 65

days as controls.

Animals were observed daily for the development of clinical signs of disease. Rectal

temperatures were measured daily for the duration of the study. Blood samples were

obtained as required by venipuncture of the jugular vein and used for determination

of total leukocyte counts, extraction of DNA from PBMC, extraction of RNA from

plasma and serum for serological tests.

The 6 cattle monitored for 65 days were killed and a complete post-mortem

examination conducted. Tissue samples (retropharyngeal and prescapular lymph

Page 80: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

70

nodes, spleen, bone marrow, kidney, lung and thymus) were collected into RNAlater

(Ambion) for DNA and RNA extraction.

Extraction of DNA and RNA from tissues

Total DNA was extracted and purified from tissue samples using a DNeasy Tissue

Kit (Qiagen) according to the manufacturer’s instructions.

Total RNA was extracted and purified from tissues using the RNeasy Mini Kit

(Qiagen) according to the manufacturer’s instructions. Disruption and

homogenisation was performed using the TissueLyser (Qiagen) and an optional on-

column DNase digestion step using an RNase-Free DNase Set (Qiagen) was included

to remove any contaminating genomic DNA.

A Ficoll-Paque Plus (GE Healthcare) gradient was used to purify PBMC from

heparinised blood according to the manufacturer’s directions. Genomic DNA was

subsequently extracted from the cells using a QIAamp DNA Mini Kit (Qiagen).

Viral RNA was extracted from plasma using the QIAamp Viral RNA Extraction Kit

(Qiagen) as recommended by the manufacturer.

Quantitation of BIV proviral DNA

Extracted DNA samples were initially tested by PCR using gene-specific (GAPDH)

primers to test for sample integrity. If no product was obtained then the sample was

not used in the analysis. The GAPDH primers used were as described in Chapter 3.

Proviral DNA loads were determined with GAPDH-positive samples by a qPCR

assay as described in Chapter 3.

BIV Proviral DNA loads were normalised to GAPDH copy number according to

previously published methods (Terwee et al., 2008). Briefly, the number of cell

equivalents (CE) in 200 ng of PBMC genomic DNA or 200 ng of tissue DNA was

Page 81: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

71

determined by qPCR using GAPDH-specific primers. All reactions consisted of 1X

iQ SYBR® Green Supermix (Bio-Rad), 400 µM of each primer (GAPDH156F:

GGTGATGCTGGTGCTGAGTA, GAPDH342R: GGCACTGCTGACAATCTTGA;

Invitrogen), 200 ng of extracted DNA and were made up to a final volume of 10 µl

using ultrapure water (Fisher Biotec). Thermal cycling conditions were 1 cycle of

95°C for 3 min, 40 cycles of 95°C for 15 s, 60°C for 30 s, 72°C for 30 s and a final

extension step of 72°C for 10 min. The DNA-based plasmid standard was generated

using the Mohan et al. (2001) GAPDH primers. The BIV proviral DNA copy number

generated using the protocol from Lew et al. (2004) was then expressed as the

number of BIV proviral DNA copies/100 000 cell equivalents (CE). The number of

CE was determined using the GAPDH qPCR protocol.

When DNA yields from PBMC were less than 50 ng/ul and insufficient for qPCR,

samples were tested by conventional PCR using primers (Heaton BIV F, 5’

CCCCAGGTCCCATCAACATTCATC and Heaton BIV R, 5’

GTCTTCCCACATCCGTAACATCTCC) as previously described (Heaton et al.,

1998).

Quantification of BIV RNA

Viral RNA loads were determined using qRT-PCR with primers and probes as

described for the qPCR for provirus except that 2 µl of viral RNA, 0.2 µl iScript

Reverse Transcriptase for One-Step RT-PCR (Bio-Rad) and 1X iScript RT-PCR

reaction mix for probes was included per reaction and a 10 min reverse transcription

incubation step at 50°C was included at the start of the reaction. To determine viral

loads within the tissues, the above protocol was followed except 200 ng of tissue

RNA was included in each reaction instead of 2 µl of viral RNA. Results were

expressed in BIV viral RNA genome copies/µg of total RNA.

Page 82: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

72

Enzyme linked immunosorbent assays

The BIV TM antibody response was determined by ELISA with 1:25 dilutions of

sera and a peptide antigen as previously described (Barboni et al., 2001; Ditcham et

al., 2009), except that the 36 amino acid TM peptide

RVSYLEYVEEIRQKQVFFGCKPHGRYCHFDFGPEEV (Proteomics

International) of BIV-R29 was converted from linear to cyclic form (TMc) as

described previously (Scobie et al., 1999). BIV hyperimmune serum was included as

a positive control and serum from uninfected cattle was used as a negative control in

every plate.

The CA antibody response was determined by ELISA using a recombinant protein

antigen. To produce the CA protein, full length BIV CA protein was cloned into the

pTrcHisB vector and transformed into JM109 chemically competent E. coli. Bacteria

were grown overnight at 37°C at 225 rpm in a 50 ml culture of standard 2X YT

medium with ampicillin (50 µg/ml, Sigma). Forty ml of overnight culture was added

to 1 L of 2X YT medium plus ampicillin and grown at 37°C for 4 h at 225 rpm.

Protein production was induced by the addition of 1 mM isopropyl-beta-D-

thiogalactopyranoside (Sigma) for 4 h. The protein was purified as recommended in

the QIAExpressionist Handbook (Qiagen) and an ELISA protocol was optimised.

One hundred ng of protein was coated onto each well of a Maxisorp 96-well plate

(Nunc) and incubated overnight at 4°C. Plates were then washed 3 times with PBS-T

(137 mM NaCl, 2.7 mM KCl, 10 mM Na2HPO4, 2 mM KH2PO4, 0.05% Tween 20).

One hundred µl of serum diluted 1:100 in PBS-T with 5% w/v skim milk (Fonterra)

was added to each well for 1 h at 37°C. Plates were then washed 3 times with PBS-T

and 100 µl of a 1:2 000 dilution of anti-bovine IgG-HRP (ICN) in PBS-T-skim milk

was added to each well for 1 h at 37°C after which they were washed 3 times with

Page 83: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

73

PBS-T and once with PBS. Colour development was induced by the addition of

peroxidase substrate (Bio-Rad) and the reaction stopped by the addition of 2% w/v

oxalic acid and absorbance read at 405 nm. BIV hyperimmune serum was included

in every plate as a positive control and serum from uninfected cattle was used as a

negative control.

Results

Clinical observations

The 13 Bali cattle infected with BIV-R29 and the 6 BFL-only controls did not

develop any clinical signs of disease during the observation period. Rectal

temperatures and total leukocyte counts remained normal throughout the experiment

and no gross lesions were observed during post-mortem examination of the 7 cattle

killed 65 days after infection.

Quantification of BIV proviral DNA load

Proviral DNA was detected in PBMC of all cattle inoculated with virus during the 65

days after infection. Proviral DNA was first detected 8 dpi in 4 of the 13 animals

(Table 1), it was detected in all 13 cattle 20 dpi but in only 2/13 at 40 dpi and 3/6 at

60 dpi. Maximum proviral DNA titres were 6.2 x102 proviral genome

copies/100 000 CE.

Quantification of BIV RNA load

Plasma viral RNA was detected in 8 of the 13 cattle from 4 to 14 dpi (Table 4.2).

Viral titres ranged from 4.2 x 101 to 1.0 x 104 genome copies/ml plasma.

Page 84: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

74

Table 4.1. BIV proviral DNA detection and quantification by conventional and

qPCR in cattle inoculated with BIV-R29.

Days after infection

Animal

Identification 0 2 4 6 8 12 14 20 40 56 60

CB169 - - - NTa +b + 1.6x101 4.5 x101 - NAd NA

CB170 - - - - - - + 5.1 x101 - NA NA

CB171 - - - NT - + + + - NA NA

CB172 - - NT NT + - + + - NA NA

CB173 - - - - - 4.9x101 c - 4.0 x102 + NA NA

CB174 - - - NT - - 6.6x101 1.6 x102 - NA NA

CB175 - - NT - - - + 1.9 x102 + NA NA

CB177 - - NT NT + + + 3.3 x101 - NT -

CB178 - - - - - - 1.2x102 3.2 x101 - + -

CB179 - - - - NT - + + - + -

CB180 - - - - - - - + - - +

CB181 - - - - - + 7.7x101 6.2 x102 - - +

CB182 - - NT NT + + 6.8x101 2.2 x101 - + +

Percent

positive 0 0 0 0 33.3 46.1 84.6 100 15.3 60.0 50.0

aNT, not tested.

b+, positive result by conventional PCR.

cNumber of BIV proviral genome copies/100 000 peripheral blood mononuclear cell-

equivalents.

dNA, not available

Page 85: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

75

Table 4.2. BIV viral RNA genome copies/ml of plasma determined by qPCR in cattle inoculated with BIV-R29.

Days after infection

Cattle 0-2a 4 6 7 8 9 10 11 12 14 15-34b 15-62c

CB169 - - - - - - - - - - -

CB170 - - - - - - - - - - -

CB171 - - - - - - - - - - -

CB172 - - - - - - - - - - -

CB173 - - - - 4.7 x103 - 5.9 x103 - - -

CB174 - - - - - 5.7 x103 - 5.4 x103 - - -

CB175 - - - - 4.2 x101 2.1 x101 - 3.2 x101 - - -

CB177 - - - - - - - - - - -

CB178 - - - - 2.0 x102 - - 3.2 x102 - - -

CB179 - - - 6.1 x101 - - 2.6 x102 6.3 x102 1.3 x102 - -

CB180 - - - - - 4.8 x102 - - - - -

CB181 - 1.0 x104 - 4.4 x101 1.0 x103 3.1 x103 6.5 x102 8 x101 - - -

CB182 - - - - - - - 3.4 x103 5.6 x103 6.0 x102 -

arepresents results from days 0,1 and 2 p.i. brepresents results from days 15, 16, 18, 20, 27 and 34 p.i. crepresents results from days 15, 16, 18, 20, 27, 34, 40, 41, 43, 45, 47 to 53, 55 to 58, 60 and 62 p.i.

Page 86: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

76

Virus RNA and proviral DNA in tissues 65 days after infection

Proviral DNA and/or viral RNA were detected in all 6 cattle killed 65 dpi

(Table 4.3). Proviral DNA was detected in at least 1 tissue type of each animal and

most commonly in lymphoid tissues. Viral RNA was detected less frequently than

proviral DNA and only in the spleen and prescapular lymph nodes. The levels of

viral RNA detected by qRT-PCR were very low, bordering on undetectable.

Serological response to BIV-R29 infection

Antibody to the BIV TMc and CA antigens were detected by ELISA (Figures 4.1 and

4.2). Increased ELISA absorbance readings with the BIV TM c peptide were detected

12 dpi and were detected in most cattle by 20 dpi. The antibody response to the CA

antigen was markedly less than to the TMc peptide and a strong antibody response to

this protein was detected in one animal only between 34 and 56 dpi (Figure. 4.2).

None of the control group developed detectable TM or CA antibody.

Page 87: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

77

Table 4.3. Viral RNA and proviral DNA quantification in tissues after necropsy of

experimentally infected Bali cattle.

Cattle Tissue

CB177 CB178 CB179 CB180 CB181 CB182

Provirusa + (<20c) + (<20) + (<20) - 21.8 21.1 Spleen

Viral RNAb + (<20) + (<20) 204.0e - - -

Provirus - + (<20) + (<20) + (<20) - + (<20) Lung

Viral RNA - - - - - -

Provirus - - - 36.6 - - Thymus

Viral RNA - - - - - -

Provirus - - - 3849.4 - - Bone marrow

Viral RNA - - - - - -

Provirus - + (<20) 3086.4 - + (<20) + (<20) Retropharyngeal

lymph node Viral RNA - - - - - -

Provirus - 6800.6d + (<20) + (<20) + (<20) - Prescapular lymph

node Viral RNA + (<20) 63.5 - - - -

Provirus - 29.28 - - - - Kidney

Viral RNA - - - - - -

aProviral genome in tissues quantified using qPCR. bViral RNA genome copies in tissues quantified using qRT-PCR. cSamples with values less than 20 but consistently above the negative control. dNumbers refer to BIV proviral genome copies/100 000 cell equivalents. eNumbers refer to BIV viral RNA genome copies/µg total RNA.

Page 88: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

78

0 8 12 14 16 18 20 27 34 56 60-1

0

1

2

3

4

Days post infection

Abs

orba

nce

(405

nm

)

10 20 30 40 50 60-1

0

1

2

3

4

CB181

CB169CB170CB171CB172CB173CB174CB175CB177CB178

CB180

CB182

CB179

CB183

CB186CB187

CB184

Days post infection

Abs

orba

nce

(405

nm

)

Figure. 4.1. TM IgG in serum of cattle after infection with BIV-R29. Antibody was

detected by ELISA with a BIV TMc peptide antigen. Absorbances were normalised

to day 0 readings. ELISA absorbances from mock infected cattle (CB183 – CB187)

are also shown. Top: individual results in 17 cattle. Bottom: box and whisker plot of

the values in the 13 cattle infected with BIV.

Page 89: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

79

10 20 30 40 50 60-1

0

1

2

3

4 CB169CB170CB171CB172CB173CB174CB175

CB177CB178

CB179

CB181CB182CB183

CB186CB187

CB180

CB184

Days post infection

Abs

orba

nce

(405

nm

)

0 8 12 14 16 18 20 27 34 56 60

-1

0

1

2

3

4

Days post infection

Abs

orba

nce

(405

nm

)

Figure. 4.2. CA IgG in serum of cattle after infection with BIV-R29. Antibody was

detected by ELISA with a BIV CA antigen. Absorbances were normalised to day 0

readings. ELISA absorbances from mock infected cattle (CB183 – CB187) are also

shown. Top: individual results in 17 cattle. Bottom: box and whisker plot of the

values in the 13 cattle infected with BIV.

Page 90: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

80

Discussion

The detection of BIV provirus in PBMC in all 13 cattle over the course of the

experiment and within tissues at the end of the experiment 65 dpi, the transient

detection of viral RNA in plasma from 8 of 13 infected cattle and an antibody

response to BIV in inoculated cattle, confirm that the Bali cattle were productively

infected with BIV.

No clinical signs of infection were observed in any of the infected Bali cattle. The

absence of significant clinical effects is similar to other experiments where BIV had

been inoculated into B. taurus (Carpenter et al., 1992; Heaton et al., 1998; Isaacson

et al., 1995; Zhang et al., 1997a). The greater susceptibility to JDV of Bali cattle than

B. taurus is not reflected in their susceptibility to BIV. This absence of clinical signs

in Bali cattle in response to BIV would be consistent with the presence of a BIV-like

virus in Bali cattle on the island of Sulawesi in Indonesia where antibody to JDV has

been detected but there is no evidence of Jembrana disease (Hartaningsih, personal

communication).

BIV proviral DNA was detected in PBMC from 8 dpi until the conclusion of the

experiment at 65 dpi but was not detected in all animals at all sampling occasions.

The highest proportion of BIV proviral DNA positive animals was at 14 and 20 dpi

when 84.4 and 100% of animals were positive, suggesting this was a peak period of

virus replication and indicating an acute phase for BIV infection. After this period

the virus appeared to persist in PBMC, at a level that was often undetectable.

Modifications to the PCR assays may have assisted in the detection of very low

proviral DNA copy numbers.

Provirus was detected in a wide variety of tissues at 65 dpi when the experiment was

concluded, similar to a previous report of infection in B. taurus (Zhang et al., 1997a).

Page 91: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

81

The levels of proviral DNA were low but highest in the lymphoid tissues examined,

spleen and lymph nodes. The low titres were similar to those reported in other

lentivirus infections, including African green monkeys persistently infected with

SIVagm (Gueye et al., 2004) and in horses asymptomatically infected with a cell-

adapted pathogenic EIAV (Harrold et al., 2000).

Although previous studies involving experimental infection of B. taurus with BIV

have detected viral RNA within PBMC subpopulations, either by RT-PCR (Baron et

al., 1995; Wu et al., 2003) or by in situ hybridisation (Carpenter et al., 1992), this

study appears to be the first report documenting the transient nature of the plasma

viraemia in the period soon after infection, similar to that reported in many other

lentivirus infections (Langemeier et al., 1996; Miyake et al., 2006; Ryan et al., 2003;

Stewart et al., 2005). While provirus was detected in all 13 infected cattle, viral RNA

was detected in 8 of 13 cattle and only during the period from 4 to 14 dpi. The levels

of virus RNA detected never exceeded 1 x 104 genome copies/ml plasma, much less

than those detected during infection of Bali cattle with the genetically related JDV

where titres of up to 1.6 x 1012 genome copies/ml plasma have been detected during

the acute disease (Stewart et al., 2005). The transient nature of the plasma viraemia

could be viewed as escape of the virus from host control. While provirus was

widespread in the tissues that were tested, viral RNA was detected in the spleen and

prescapular lymph node only of some animals and was at low levels. Perhaps the

level of replication of BIV relative to JDV may be associated with the relative lack of

pathogenicity of BIV compared to JDV.

There was a rapid and strong response against TM but a poor antibody response to

the CA protein in most infected cattle. The TM response to BIV infection was

similar to that reported by Scobie et al. (1999) in cattle infected with the BIV FL112

Page 92: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

82

isolate and an antibody response was detected as early as 2 weeks pi. The weak CA

response detected in all but one of the 13 infected Bali cattle was surprising as in

B. taurus there is normally a strong response to CA between 2 and 4 weeks pi

(Isaacson et al., 1995; Whetstone et al., 1991). Seroconversion to a gag precursor

also occurred between 2 and 4 weeks pi in rabbits infected with BIV (Pifat et al.,

1992). The reason for the poor antibody response to CA in Bali cattle is unknown. In

response to infection with the genetically and antigenically related JDV, Bali cattle

normally mount a strong albeit delayed immune response against CA (Hartaningsih

et al., 1994) but as in the Bali cattle infected with BIV, a subset of cattle infected

with JDV mount a poor antibody response to the JDV CA and a strong antibody

response to JDV TM (Ditcham et al., 2009). A 10 to 100 times greater antibody

response to envelope proteins compared to CA proteins was also observed in horses

in response to EIAV (O'Rourke et al., 1988).

Results from this study indicate not only that BIV is non-pathogenic in Bali cattle but

that maximum virus replication occurred soon after infection and prior to the onset of

a significant antibody response, certainly prior to the onset of a significant antibody

response to the CA protein. It is likely that future attempts to detect BIV infection in

Bali cattle and possibly other cattle species using PCR based assays, would have

greatest chance of success soon after infection and before the onset of a significant

antibody response.

Page 93: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

83

Chapter 5: Bovine immunodeficiency virus infection alters the

dynamics of subsequent Jembrana disease virus infection

Summary

To determine whether BIV infection is capable of protecting against superinfection

with JDV, 15 animals were infected with BIV-R29 and 42 days after BIV infection,

9 of the BIV infected and 4 mock BIV infected animals were superinfected with

JDVTab87. All cattle were successfully infected with BIV, shown by the presence of

proviral DNA and, in a subset of cattle, a transient viraemia. Strong antibody

responses against the TM glycoprotein and poor antibody responses against the CA

protein were also detected. Despite the development of immune responses against

TM, a region known to contain cross-reactive epitopes, all cattle became infected

with JDV, as indicated by the development of typical clinical signs of Jembrana

disease and an acute phase viraemia.

Page 94: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

84

Introduction

The 2 bovine lentiviruses, BIV (Gonda et al., 1987) and JDV (Chadwick et al.,

1995a; Kertayadnya et al., 1993), are genetically and antigenically related but differ

markedly in pathogenicity. The incidence of clinical Jembrana disease and

serological surveys indicate JDV is common in the Bali cattle population in parts of

Indonesia (Hartaningsih et al., 1993). A BIV-like non-pathogenic bovine lentivirus is

also suspected to occur in the cattle population on Bali island (Barboni et al., 2001)

and antibody to JDV has been detected in Bali cattle on the island of Sulawesi where

there is no clinical evidence of Jembrana disease in the Bali cattle population

(Desport et al., 2005). Antibody to JDV and BIV cannot be differentiated due to the

presence of numerous cross-reactive epitopes on the CA (Desport et al., 2005;

Kertayadnya et al., 1993) and TM proteins (Burkala et al., 1998).

A protective immunity against JDV infection has been induced by vaccination with

inactivated whole virus antigens (Ditcham et al., 2009). It was considered possible

that infection of Bali cattle with a non-pathogenic BIV-like lentivirus might also

induce a protective immune response against Jembrana disease. Non-pathogenic

strains of other lentiviruses have been shown to induce protective immunity against

pathogenic strains of the same virus. Infection of domestic cats with non-pathogenic

lion lentivirus or puma lentivirus ameliorated the effects of subsequent wild-type FIV

infection (Terwee et al., 2008; VandeWoude et al., 2002). Similar results were

obtained in domestic cats pre-infected with chimeric FIV (generated by substituting

part of env of clade A FIVPET with a corresponding region of clade B FIVM2, or vice-

versa) and subsequently inoculated with FIVPET or FIVM2 (Giannecchini et al., 2007).

Infection of macaques infected with attenuated SIVmac also ameliorated the effects of

challenge with pathogenic SIVmac (Cranage et al., 1998; Sharpe et al., 2004) and

Page 95: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

85

SIVsm (Nilsson et al., 1998) although attenuated SIVmac did not provide protection

against the more divergent HIV-2 (Nilsson et al., 1998).

This Chapter describes an experiment to determine whether prior infection of Bali

cattle with BIV would provide protection against superinfection with pathogenic

JDV infection 42 days after the initial BIV infection. This experiment was expected

to provide information that would increase our understanding of the effect JDV

would have on the Bali cattle population if it were introduced onto the island of

Sulawesi where BIV is suspected to occur in the cattle population of that island.

Materials and methods

Animals

Nineteen cattle approximately 18 months of age were obtained from Nusa Penida, an

island adjacent to Bali where Jembrana disease has never been detected and the cattle

have been consistently negative to antibody to JDV and BIV (Hartaningsih et al.,

1993). The cattle were housed indoors as previously described (Soeharsono et al.,

1990). Cattle were screened by PCR and ELISA to ensure they were not infected

with bovine lentiviruses prior to challenge (Chapter 3). Six weeks prior to infection

with BIV the cattle were vaccinated against BVDV with Pestigard® (Pfizer), as

previously described (Chapter 4).

Viruses

The BIV-R29 for infection of cattle was obtained from J Brownlie and M Collins,

Royal Veterinary College, UK. The virus was cultured in primary BFL cells and

titrated in BFL cells as previously described (Chapter 4). An estimated 1.82 x 104

TCID50 was inoculated into each animal, half subcutaneously and half intravenously.

Page 96: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

86

The JDVTab87 (Chadwick et al., 1995b) used for infection of cattle was prepared by

infection of antibody-negative cattle with a suspension of frozen spleen harvested

from infected cattle as described previously (Soeharsono et al., 1990). Plasma from

the infected cattle was obtained 2 days after the development of a febrile response

typical of Jembrana disease, and the approximate ID50 in the plasma determined

using an antigen-capture ELISA as described previously (Stewart et al., 2005). An

estimated 1 000 ID50 of the virus was inoculated intravenously into each animal.

Experimental infection and sampling of cattle

Nine cattle were inoculated with BIV at day 0 and with JDV 42 days later (CB190–

CB197 and CB205). Six cattle were inoculated with BIV at day 0 and were not

subsequently inoculated with JDV (CB198–CB202 and CB204). Four cattle were

inoculated with uninfected BFL cells in RPMI medium at day 0 and subsequently

with JDV 42 days later (CB203, CB206, CB208 and CB210).

Animals were observed daily for clinical signs of disease. Rectal temperatures were

measured daily for the duration of the study. Heparinised blood samples were

obtained as required by venipuncture of the jugular vein and used for extraction of

DNA from PBMC, extraction of RNA from plasma and serum for serological tests.

Extraction of DNA and RNA from peripheral blood

PBMC were purified from 10 ml of heparinised blood using a Ficoll-Paque PlusTM

(GE Healthcare) gradient as recommended by the manufacturer. DNA was extracted

from the PBMC using a QIAamp DNA Mini Kit (Qiagen) as recommended by the

manufacturer. Viral RNA was extracted from plasma using the QIAamp Viral RNA

Extraction Kit (Qiagen) as recommended by the manufacturer.

Page 97: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

87

Detection of BIV proviral DNA

The integrity of genomic DNA was confirmed using gene-specific GAPDH primers

as previously described (Chapter 4). BIV proviral DNA loads were determined using

a conventional PCR assay as described previously (Chapter 4).

Quantification of BIV and JDV RNA in plasma

BIV RNA was quantified as described previously (Chapter 4) and JDV RNA as

described by Stewart et al. (2005), with the following exceptions: 1X RT-PCR

Reaction Mix for Probes (Bio-Rad), 0.2 µl iScript Reverse Transcriptase for One-

Step RT-PCR (Bio-Rad) and 2 µl RNA extracted from plasma was added to each

reaction. The reaction was made up to 10 µl using nuclease free water (Bio-Rad).

ELISA

The BIV TM antibody response was determined by ELISA using a cyclic BIV TM

(BIV TM c) peptide as described previously (Chapter 4). The JDV TM antibody

response was determined by ELISA using a cyclic JDV TM (JDV TMc) peptide as

previously described (Ditcham et al., 2009) as while there was extensive cross-

reactivity between JDV and BIV TM antigens, our experience was that the JDV TMc

peptide provided greater sensitivity in the detection of JDV TM antibody than did the

BIV TM c antigen. The BIV CA antibody response was determined using an ELISA

with a recombinant BIV CA antigen as previously described (Chapter 4) except that

100 ng of protein was coated onto each well and serum was tested at a dilution of

1:100.

Page 98: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

88

Analysis of data

To determine whether prior BIV infection would induce protection against

subsequent JDV infection, differences between the viral load (VL) on the first and

second day of the febrile period when the rectal temperature exceeded 39.3°C, the

peak VL, the duration and magnitude of the VL where the area under the curve

(AUC) was >106 genomes/ml, and the duration of the febrile period, were compared

in JDV-infected cattle that had been previously infected with BIV and in cattle not

previously infected with BIV, as described previously (Desport et al., 2009a;

Ditcham et al., 2009). The AUC where the VL was > 106 for each animal was

estimated and linear interpolation between consecutive observations, in combination

with the previously described piecewise-linear model was used to supply missing

data caused by variations in sampling intervals (Ditcham et al., 2009). The duration

of the infectious period was defined as the period where VL was > 106 genomes/ml.

A baseline of 106 was chosen (Ditcham et al., 2009) as bloodmeal residues of

between 4–10 nl have been reported on the mouthparts of tabanid flies and EIAV at

106 ID/ml in blood can be transmitted by a single fly (Foil et al., 1987). For analysis

of the febrile response, rectal temperatures were divided into ranges adapted from a

previously described method (Muraguri et al., 1999) as low fever (>39.3 °C – 40.2

°C), moderate fever (40.3 °C – 41.2 °C) and high fever (> 41.2 °C). Student’s t-test

was used for statistical comparison of the magnitude and duration of plasma VL and

febrile responses and P-values < 0.05 were considered statistically significant.

Page 99: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

89

Results

Clinical and virological observations in cattle after BIV inoculation

Fifteen BIV inoculated Bali cattle and 4 mock-infected control cattle were monitored

for 42 days and then superinfected with JDV. During the initial 42 day period after

BIV infection, the BIV-infected cattle did not exhibit any change in rectal

temperature or other clinical signs of disease.

BIV proviral DNA was detected in PBMC initially 7 dpi in one animal and

subsequently in PBMC of all 15 cattle inoculated with BIV, confirming that all

animals were infected (Table 5.1). BIV proviral DNA was detected in most cattle

after 17 dpi but at no single time point was BIV proviral DNA detected concurrently

in all 15 cattle. BIV proviral DNA was not detected in the mock-infected cattle.

Plasma viral RNA was detected in 5 of the 15 cattle in the period from 8 to 13 dpi

(Table 5.2) and plasma viral titres ranged from 1.04 to 4.25 log10 genome copies/ml

plasma.

An antibody response to the BIV TMc and CA antigens was detected by ELISA

(Figure 5.1). Antibody to the BIV TMc peptide was detected from 10 dpi and in the

majority of cattle by 41 dpi (Figure 5.1A). Antibody to the BIV CA antigen was

detected from 19 dpi (Figure 5.1C) but the CA response was markedly less than the

response to the BIV TMc peptide (Figure 5.1A). Only some of the BIV-infected

cattle developed CA antibody and only one seroconverted strongly in the 42 day

period after infection. None of the mock-infected cattle developed BIV TMc or CA

antibody (Figure 5.1B and 5.1D).

Page 100: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

90

0 7 10 13 17 21 28 35 41

0.0

0.5

1.0

1.5

2.0

2.5

Days after BIV inoculation

Abs

orba

nce

(405

nm

)

0 7 10 13 17 21 28 35 41

0.0

0.5

1.0

1.5

2.0

2.5

Days after mock BIV inoculation

Abs

orba

nce

(405

nm

)0 1 5 11 19 41

0.0

0.5

1.0

1.5

Days after BIV inoculation

Abs

orba

nce

(405

nm

)

0 1 5 11 19 41

0.0

0.5

1.0

1.5

Days after mock BIV inoculation

Abs

orba

nce

(405

nm

)

A B

C D

Figure 5.1. TMc and CA IgG response in serum of cattle after inoculation with BIV,

shown as box and whisker plots of the ELISA absorbance values. (A) Response

detected by BIV TMc peptide in all 15 cattle inoculated with BIV at day 0.

(B) Response detected by His-BIV CA antigen in all 15 cattle inoculated with BIV at

day 0. (C) Response detected by BIV TMc peptide antigen in all 8 cattle mock

inoculated at day 0. (D) Response detected by His-BIV CA antigen in all 8 cattle

mock inoculated at day 0.

Page 101: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

91

Table 5.1. BIV proviral DNA detected by PCR in cattle inoculated with BIV and

prior to infection with JDV.

Days after BIV infection Animal

0 7 10 17 19 21 42

Cattle inoculated with BIV at day 0 and then JDV at day 42

CB190 -a - - +a - + - CB191 - - + + - + - CB192 - - NT + - + - CB193 - - NT + + + + CB194 - - - + + - + CB195 - - - + + + + CB196 - - - + + - - CB197 - - - + - + - CB205 - - - + + + + Cattle inoculated with BIV only at day 0 CB198 - - - - - - + CB199 - + + - + - + CB200 - - - NTb + - + CB201 - - + + + - - CB202 - - - + - - + CB204 - - + + - - + Cattle inoculated with JDV only at day 42 CB203 - - NT - - - - CB206 - - NT - NT - - CB208 - - - - - - - CB210 - - - - - - - Percent BIV-inoculated cattle positive 0 7 15 85 53 47 60 Cumulative % positive 0 7 15 93 93 93 100 Percent mock-inoculated cattle positive 0 0 0 0 0 0 0

a - and + denote negative and positive PCR results, respectively, for BIV proviral

DNA.

b not tested.

Page 102: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

92

Table 5.2. BIV viral RNA genome copies/ml (log10) of plasma determined by qPCR in cattle inoculated with BIV at day 0 (all cattle)

and JDV at day 42 (CB190 – CB205 only).

Days after BIV inoculation Days after JDV inoculation

Animal 0 - 1a 7 8 9 10 11 13

14-

42c

44-

50d 51 52 53 54 55

56-

63e

Cattle inoculated with BIV at day 0 and JDV at day 42 CB190 -b - - - 1.20 - 4.25 - - - - - - - - CB191 - - - - - - 1.89 - - 1.40 - - - - - CB192 - - - - - - - - - - - - - - - CB193 - - - - 1.79 - - - - - - - - - - CB194 - - - - - - - - - - - 1.86 - - - CB195 - - - - - - - - - 2.18 - 2.57 2.51 1.48 - CB196 - - - - - - - - - - - - - - - CB197 - - - - - - - - - 1.79 - - - - - CB205 - - 1.04 2.45 - - - - - - - - - - - Cattle inoculated with BIV at day 0 only CB198 - - - - - - - - - - - - - - - CB199 - - - - - - - - - - - - - - - CB200 - - - - - - - - - - - - - - - CB201 - - - - - - - - - - - - - - - CB202 - - - - - - - - - - - - - - - CB204 - - - 1.04 - - - - - - - - - - - a represent results from 0 and 1 dpi. b negative. c represents results from 14 - 17, 19, 21, 28, 35 and 41 - 42 dpi. d represents results from 44, 46 and 48 - 50 dpi. e represents results from 56 - 59, 61 and 63 dpi

Page 103: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

93

Clinical and virological observations after JDV inoculation

Forty two days after BIV infection, 9 of the 15 BIV-infected cattle and 4 of the

mock-infected cattle were inoculated with JDV and monitored for a further 15 to 19

days. All the JDV-inoculated cattle developed a febrile response but there were

differences in the responses in the previously BIV-infected and non-BIV-infected

groups (Figure 5.2). Excluding 2 animals CB190 and CB205 that responded

uncharacteristically to JDV infection and were considered atypical responders (see

below), the febrile response in the BIV-infected group started 2 days earlier (a mean

of 7 dpi compared to 9 dpi, P = 0.058), the peak of the febrile response occurred

earlier (a mean of 10 dpi versus 12 dpi, P = 0.067), the peak VL also occurred

significantly sooner (a mean of 10 dpi versus 12 dpi, P = 0.008) and there was a

significantly earlier conclusion of the febrile response (a mean of 13 dpi compared to

15 dpi, P = 0.033, Table 5.3). There were no significant differences between groups

in regards to the duration and severity of the febrile response (Table 5.3). The 6 BIV-

only controls did not develop a febrile response or other clinical signs during the

observation period (Figure 5.2).

JDV plasma RNA was detected in all cattle inoculated with JDV from 3 to 19 dpi

(Figure 5.3). The maximum VL was 2.18 x 1011 genome copies/ml plasma.

Excluding the 2 atypical responders, the cattle infected with JDV, regardless of

whether they had been challenged previously with BIV, developed a viraemia typical

of that reported previously (Desport et al., 2009a; Stewart et al., 2005). As shown in

Figure 5.3, the pattern and magnitude of the viraemia was also similar between

groups, but the viraemia started and finished earlier in the cattle infected previously

with BIV, although these differences were not statistically significant; accordingly,

in cattle previously infected with BIV there was a reduction in the mean duration of

Page 104: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

94

the JDV viraemia that was >106 genomes/ml plasma (P = 0.068). There were no

significant differences between previously BIV-infected and non-BIV-infected

groups in the VL on the first or second day of the febrile period, the peak VL or the

total AUC when the viraemia was ≥106 genomes/ml plasma (Table 5.4).

Two animals, CB190 and CB205, responded atypically to JDV infection. The cattle

had a late onset fever (Figures 5.2 and Table 5.3) and the dynamics of their viraemia

were different to those in the other cattle (Figure 5.3), similar to atypical responders

previously reported (Desport et al., 2009a). These animals had an erratic viraemia

increasing to a maximum titre of approximately 1010 genome copies/ml which

continued through to the conclusion of the experiment. As the viraemia in these

animals did not decrease before the experiment concluded, the data provided for the

duration of the viraemia in Table 5.4 is an underestimation of the actual values.

After inoculation with JDV, plasma BIV RNA was again detected in 4 of the 9

superinfected cattle (Table 5.2). The titre of BIV RNA during this period ranged

from 1.40 to 2.57 log10 genome copies/ml plasma.

Antibody to the JDV TMc antigen was detected (Figure 5.4) in the 15 day period

after JDV infection in a majority of cattle previously infected with BIV (Figure

5.4A), but was not detected in any of the JDV-infected cattle that had not been

infected previously with BIV (Figure 5.4B). ELISA absorbances to the CA protein

increased at 4 days after JDV infection in the cattle that had been previously infected

with BIV (Figure. 5.4D). BIV CA ELISA absorbances remained low in the cattle

infected with JDV only (Figure 5.4F).

Page 105: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

95

0 2 4 6 8 10 12 14 16 18 20 22

37.5

38.0

38.5

39.0

39.5

40.0

40.5

41.0

41.5

42.0

Days after JDV inoculation

Rec

tal t

empe

ratu

re (

°° °°C)

Figure 5.2. Mean rectal temperatures in cattle inoculated with JDV. Y-error bars

represent the standard deviations of rectal temperatures. Shown are the rectal

temperatures in cattle which had been previously inoculated with BIV and responded

to JDV in a typical fashion (○; CB191 – CB197), in cattle which had been

previously infected with BIV and responded to JDV in an atypical fashion (▼;

CB190 and CB205), JDV-only control cattle (●; CB203, CB206, CB208 and CB210)

and BIV-only control cattle JDV (■).

Page 106: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

96

Table 5.3. Effect of previous BIV infection of cattle on the febrile response

following JDV infection.

Days after infection Duration of febrile response

(days)

Animal

Peak

VL

Onset of

febrile

response

Peak

febrile

response

End of

febrile

response

Low Moderate High Total

Cattle infected with BIV 42 days previously

CB190 18 17 18 21 1 3 0 4

CB191 8 5 8 13 4 2 2 8

CB192 9 8 10 12 3 1 0 4

CB193 12 11 13 16 4 1 0 5

CB194 9 4 10 12 8 0 0 8

CB195 11 8 10 15 3 3 1 7

CB196 9 7 10 12 1 3 1 5

CB197 9 7 9 13 3 3 0 6

CB205 18 17 20 21 0 3 1 4

Mean 11

(10)a

9

(7)a

12

(10)a

15

(13)a

3 2 1 6

Variance 15.28 22.75 17.75 13.50 5.50 1.36 0.53 2.75

Cattle not previously infected with BIV

CB203 12 8 10 15 2 5 0 7

CB206 12 10 13 15 4 1 0 5

CB208 11 10 11 14 1 2 1 4

CB210 12 9 12 17 4 4 0 8

Mean 12 9 12 15 3 3 0 6

Variance 0.25 0.92 1.67 1.58 2.25 3.33 0.25 3.33

P-valueb 0.441

(0.008)c

0.487

(0.058)c

0.412

(0.067)c

0.449

(0.033)c

0.425 0.153 0.233 0.376

a mean calculated excluding atypical responders CB190 and CB205. b P-values represent statistical differences between animals infected with BIV and then JDV and those infected with JDV only. c P-value calculated excluding atypical responders CB190 and CB205.

Page 107: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

97

0 2 4 6 8 10 12 14 16 18 20

0

2

4

6

8

10

12

Days after JDV inoculation

log1

0 JD

V v

iral

RN

Age

nom

e co

pies

/ml p

lasm

a

Figure 5.3. Mean plasma viral loads (JDV RNA genome copies/ml plasma) in

animals inoculated with JDV at day 0. Y-error bars represent the standard deviations

of viral loads. Shown are viral loads in cattle which had been previously infected

with BIV and responded to JDV in a typical fashion (o; CB191 – CB197), JDV-only

control cattle (●; CB203, CB206, CB208 and CB210), and cattle which had been

previously infected with BIV and responded to JDV in an atypical fashion

(▼; CB190 and CB205).

Page 108: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

98

Table 5.4. Effect of previous BIV infection on the dynamics of the JDV viral load

(VL) in plasma after JDV inoculation.

Animal VL 1st day of

febrile period

(log10)

VL 2nd day of

febrile period

(log10)

Peak VL

(log10)

AUC ≥106

genome

copies/ml (log10)

Total

days

VL≥106

Cattle infected with BIV 42 days previously

CB190 6.61 9.77 11.34 10.15 5.60

CB191 7.35 10.63 10.12 11.76 11.20

CB192 8.49 9.59 9.59 9.71 7.40

CB193 8.89 9.26 9.26 9.68 8.60

CB194 1.91 4.64 9.98 10.25 7.60

CB195 9.23 10.12 10.57 10.98 10.10

CB196 9.22 9.25 10.31 10.44 9.60

CB197 9.36 9.93 10.72 11.00 8.50

CB205 11.25 10.68 10.68 10.70 9.20

Mean 8.04 9.06 10.08 10.55 9.00

Variance 7.00 4.04 0.27 0.57 1.91

Cattle not previously infected with BIV

CB203 9.98 10.28 10.73 11.18 16.40

CB206 9.40 10.01 10.18 10.55 10.30

CB208 7.54 8.95 10.02 10.22 10.70

CB210 7.46 9.16 10.13 10.60 13.40

Mean 8.59 9.60 10.26 10.64 12.53

Variance 1.65 0.41 0.10 0.15 7.77

P-valuea 0.624 0.692 0.931 0.404 0.068

a P-values represent statistical differences between groups of animals infected with

BIV 42 days prior to infection with JDV and those infected with JDV only.

Page 109: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

99

A D

B E

C F

-1 2 6 10 15

0.0

0.5

1.0

1.5

2.0

2.5

Days after JDV inoculation

Abs

orba

nce

(405

nm

)

-1 4 6 10 15

0.0

0.5

1.0

1.5

2.0

2.5

Days after JDV inoculation

Abs

orba

nce

(405

nm

)

-1 2 6 10 15

0.0

0.5

1.0

1.5

2.0

2.5

Days after mock JDV inoculation

Abs

orba

nce

(405

nm

)

-1 6 15

0.0

0.5

1.0

1.5

2.0

2.5

Days after mock JDV inoculation

Abs

orba

nce

(405

nm

)-1 2 6 10 15

0.0

0.5

1.0

1.5

2.0

2.5

Days after JDV inoculation

Abs

orba

nce

(405

nm

)

-1 6 15

0.0

0.5

1.0

1.5

2.0

2.5

Days after mock JDV inoculation

Abs

orba

nce

(405

nm

)

Figure 5.4. IgG response detected by JDV TMc peptide and His-BIV CA after

inoculation with JDV, shown as box and whisker plots of the ELISA absorbance

values. (A) Response detected by JDV TMc peptide in cattle infected with JDV at

day 0 and infected with BIV 42 days earlier. (B) Response detected by JDV TMc

peptide in cattle infected with JDV at day 0 but not infected previously with BIV.

(C) Response detected by JDV TMc peptide in cattle not infected with JDV at day 0

but infected with BIV 42 days earlier. (D) Response detected by His-BIV CA in

cattle infected with JDV at day 0 and infected with BIV 42 days earlier.

(E) Response detected by His-BIV CA in cattle infected with JDV at day 0 only.

(F) Response detected by His-BIV CA in cattle not infected with JDV at day 0 but

infected with BIV 42 days earlier.

Page 110: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

100

Discussion

Under the conditions with which the experiment was conducted, prior BIV infection

did not prevent subsequent JDV infection or result in significant amelioration of the

normal response of Bali cattle to JDV infection. Although it did not alter the normal

clinical response to JDV infection, prior infection of cattle with BIV altered the

dynamics of their response to JDV and resulted in an earlier onset of Jembrana

disease. Although many of the effects were not significantly different, cattle

previously infected with BIV developed an earlier onset of fever, an earlier peak

febrile response, a significantly earlier peak VL and a significantly earlier resolution

of fever following superinfection with JDV. Although prior BIV infection did not

cause a difference in the total AUC, there was (in cattle previously infected with

BIV) a reduction in the duration of the viraemia that exceeded 106 genome copies/ml

of plasma. The differences were only evident when 2 atypical responders, identified

in a small percentage of all cattle infected with the JDVTab87 strain of JDV (Desport et

al., 2009a), were removed from the analysis. The lack of statistical significance in

those animals that responded typically was probably at least partly due to the wide

variance in the response to JDV of the cattle previously infected with BIV. A case

fatality rate of about 21% is normally evident in experimentally JDV-infected Bali

cattle (Desport et al., 2009a) but no fatalities occurred in any of the JDV-infected

cattle during the 14 day observation period after infection so no effect on the JDV-

associated case fatality rate of prior BIV infection was determined.

The enhanced early replication of a superinfecting virus seen here has not been

reported previously in superinfections with other related lentiviruses. An earlier

onset of disease has, however, been reported with CAEV, JDV, EIAV and FIV

following vaccination. Goats vaccinated with inactivated CAEV developed arthritis

Page 111: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

101

more rapidly after CAEV infection than control animals (McGuire et al., 1986).

Cattle vaccinated with a tissue-derived JDV vaccine had earlier peak VL than control

animals (Ditcham et al., 2009). Horses vaccinated with a recombinant EIAV subunit

vaccine and then challenged with EIAV displayed severe enhancement of viral

infection and exacerbation of disease (Montelaro et al., 1996). Goats vaccinated with

a T-cell priming Gag peptide from CAEV had transiently enhanced virus replication

after CAEV infection compared with control animals, potentially via T-cell

enhancement of virus replication (Nenci et al., 2007) and similar findings have been

reported in FIV vaccine studies (Richardson et al., 1997). The presence of an active,

cross-reactive T-helper cell immune response may explain the earlier start of

viraemia and accelerated febrile response. JDV has recently been identified as

replicating in IgG-containing cells (Desport et al., 2009b) and alternatively, the

earlier replication of JDV in BIV-infected cattle may be related to the B-cell

stimulatory activity of BIV (Whetstone et al., 1997).

The lack of amelioration of the febrile response and the replication of JDV in the

cattle previously infected with BIV occurred despite a strong antibody response to

the BIV TM and, in a proportion of the cattle, an antibody response to the CA at the

time of JDV inoculation. Due to the close antigenic relationship between JDV and

BIV (Desport et al., 2005; Kertayadnya et al., 1993) the result was not expected as

other lentiviruses have been shown to offer protection against infection with closely

related heterologous viruses. Domestic cats infected with non-pathogenic puma or

lion lentiviruses developed humoral and cell-mediated immune responses against

both homologous and heterologous FIV isolated from domestic cats (VandeWoude et

al., 2003), suppressed FIV-induced CD4+ T-cell depletion (Terwee et al., 2008;

VandeWoude et al., 2002) and FIV-induced plasma and PBMC viral loads

Page 112: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

102

(VandeWoude et al., 2002). Domestic cats pre-infected with a chimeric FIV and

subsequently infected with fully virulent FIV ameliorated the clinical effects of the

virulent challenge virus in some challenged cats, and reduced viral RNA and proviral

DNA loads in others (Giannecchini et al., 2007). A lack of neutralising antibody

response against the TM glycoprotein could explain why JDV infection occurred

despite a strong anti-TM antibody response.

It is possible that the R29 strain of BIV may have become attenuated since its

isolation in 1969 (Whetstone et al., 1997) and that this may have affected the result

obtained. It is also possible that the 42 day period between BIV infection and

subsequent JDV infection was too short for an effective protective immunity to

develop. Although there was a strong antibody response to the TM protein of BIV at

the time of JDV infection, and a rapid antibody response to TM and CA after JDV

infection, which is normally delayed until at least 6 weeks after infection

(Hartaningsih et al., 1994), this response may be unrelated to the development of a

protective immune response which may require a longer period to develop with the

bovine lentiviruses. However, in a similar experiment, protection against SIV in

macaques could be achieved within 21 days of infection with a live attenuated SIV,

with partial protection against wild-type SIV provided within 10 days of inoculation

(Stebbings et al., 2004). In contrast, after wild-type FIV infection there was no

protection against the effects of heterologous virus challenge until 2 to 3 years after

infection when animals exhibited a reduced virus load and sometimes a reduced

decline of CD4+ T-cells (Pistello et al., 1999). The possibility that longer term

infections with BIV may induce a protective response to JDV infection needs further

investigation as it would perhaps offer a low cost means of immunizing cattle against

Jembrana disease.

Page 113: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

103

The dose of BIV used for infection and the challenge dose of JDV may also be

factors responsible for lack of resistance to JDV infection following BIV infection.

Studies of SIVmac in macaques have shown that the dose of primary virus affects the

level of resistance to infection with a pathogenic virus (Cranage et al., 1998).

Macaques given higher doses, 2 000 - 20 000 TCID50, of SIVmacC8 were shown to

completely resist infection with SIVmac, shown by a lack of virus detection by PCR

and a lack of virus isolation, whereas animals given lower doses of primary virus, 2 –

200 TCID50, were protected against a loss of CD4 cells only.

While there was no evidence that prior BIV infection provided protection against the

pathological effects of subsequent JDV infection, there was also no evidence that the

prior BIV infection exacerbated subsequent JDV infection, as reported in cases of

HIV superinfection. Epidemiological observations suggest that infection with a

second heterologous strain of HIV-1 has in the majority of cases, accelerated disease

progression after infection, reviewed previously (Smith et al., 2005). It was observed

in the current experiments that JDV infection in cattle previously infected with BIV

reactivated replication of the BIV, and this appears to be an observation seen during

heterologous lentivirus infections. Reactivation of primary infection virus has also

been seen in HIV-2 infected macaques superinfected with SIVmac (Petry et al., 1995)

and in HIV-2 infected baboons superinfected with heterologous HIV-2 (Locher et al.,

1997). Reactivation might be associated with the potent transactivation function of

the JDV Tat protein, which was shown to be a potent transactivator not only of its

own LTR but also the BIV and HIV LTR in vitro (Chen et al., 2000; Chen et al.,

1999). JDV Tat may function similarly in vivo in Bali cattle.

There is no evidence of the outcome of mixed infections with JDV and non-

pathogenic BIV-like viruses under field situations in the Bali cattle population of

Page 114: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

104

Indonesia. Although mixed infection with JDV and a second BIV-like virus has been

reported to occur in Bali cattle on Bali island (Barboni et al., 2001), the difficulty of

detecting BIV-like virus in cattle and the close antigenic relationship of the 2 viruses

has so far precluded the epidemiological study of mixed infection by JDV and

possible non-pathogenic BIV-like viruses. BIV proviral DNA is only transiently

present within PBMC of experimentally infected Bali cattle (Chapter 4), making the

virus difficult to detect even when the animals are known to be infected.

Page 115: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

105

Chapter 6: Humoral immune responses to Jembrana disease virus

detected using overlapping synthetic peptides spanning the MA, CA

and SU regions of JDV

Summary

The mapping of linear B-cell epitopes on the MA, CA and SU regions of Jembrana

disease virus is described. One hundred and fifty five overlapping peptides that

spanned these regions were synthesised and used in an ELISA format to screen a

panel of bovine sera from animals experimentally infected with JDVTab87, JDVPul01 or

BIV-R29. Six immunoreactive (IR) peptides, representing 6 potential epitopes, were

identified when the set of peptides was screened with sera taken following JDVTab87

infections; 1 in MA, 1 in CA and 5 in SU. Numerous IR peptides were identified

when the set of peptides was screened with JDVPul01 sera. BIV-R29 sera also reacted

with many peptides, including the IR peptides identified with the JDVTab87 sera.

However, BIV-R29 sera did not react with some peptides and a combination of these

peptides would enable detection of JDV-only seropositive cattle. These peptides

include MA18, MA19, SU93, SU95, SU103, SU119 and SU135.

Page 116: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

106

Introduction

The occurrence of clinical Jembrana disease and serological surveys for antibody

reactive to JDV indicate that JDV infection is widespread in some islands of

Indonesia including Bali, Java, Sumatra and Kalimantan (Indonesian Borneo) but the

absence of clinical Jembrana disease and the occurrence of antibody to JDV suggests

the occurrence of a non-pathogenic bovine lentivirus, possibly related to BIV, on the

island of Sulawesi (Hartaningsih, personal communication). A BIV-like, non-

pathogenic bovine lentivirus was also reported to occur in the cattle population on

Bali island (Barboni et al., 2001) but this has not been confirmed and efforts to detect

BIV proviral DNA in PBMC of cattle on Bali island were unsuccessful (Chapter 3).

While JDV and BIV are sufficiently different genetically that they can be

differentiated utilising a number of PCR assays (Lew et al., 2004; Lewis et al., 2009)

low proviral DNA loads in PBMC after infection precludes their use as a screening

tool. They are very similar antigenically and attempts to differentiate antibody to the

2 viruses using ELISA and WIB procedures have been unsuccessful due to the

presence of numerous cross-reactive epitopes on the CA, MA and TM proteins

(Desport et al., 2005). Using recombinant, overlapping JDV Gag proteins, previous

attempts have been made to define specific epitopes that differentiate between the 2

bovine lentiviruses antigenically, however these were unsuccessful (Desport et al.,

2005). A number of recombinant proteins were produced and reacted with JDV and

BIV hyperimmune sera as well as monoclonal antibodies in a WIB. These attempts

identified at least three epitopic domains within MA and CA, including the Major

Homology Region (MHR) but could not identify an epitope(s) that differentiated

between the two viruses (Desport et al., 2005). A monoclonal antibody directed

against an epitope which spans the cleavage site between BIV MA and the p2L

Page 117: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

107

protein was reported to differentiate between BIV and JDV antibody using a WIB

assay (Lu et al., 2002; Zheng et al., 2001) but the epitope involved does not appear to

be immunogenic in cattle and linear forms of this epitope can not be used to

differentiate between bovine lentivirus infections (Desport et al., 2005). Reliable

serological surveillance for JDV infection in cattle in Indonesia requires the use of

serological techniques that can differentiate the 2 infections.

Various epitope mapping studies have employed the use of expression libraries and

recombinant proteins to map epitopes and then fine mapping using synthetic peptide

strategies (Bertoni et al., 1994; Chong et al., 1991a; Chong et al., 1991b; Rosati et

al., 1999). These approaches were used to map the antigenicity of the SU

glycoproteins of EIAV (Ball et al., 1992) and CAEV (Valas et al., 2000) and the

antigenicity of CAEV TM (Bertoni et al., 1994). In this Chapter, synthetic peptides

were used to map the epitopes of JDV MA, CA and SU proteins to find epitopes

which can be used for differentiating the 2 viruses in serological assays.

Materials and methods

Source of animal sera

A panel of bovine sera from experimentally infected Bali cattle was used in this

study. The cattle were infected with either JDVTab87, JDVPul01 or BIV-R29 during

various studies of the response of cattle to these viruses (Desport et al., 2009;

Ditcham et al., 2009; Chapters 4 and 5) and serum samples were acquired at various

dpi. Pre-infection serum samples from the same cattle were used to test the

background level of reactivity to each peptide.

Hyperimmune JDV and BIV sera, a gift from N. Hartaningsih (Indonesia) were also

tested. JDV hyperimmune serum was created by inoculating a bovine lentivirus-free

Page 118: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

108

animal with plasma taken from an animal during the febrile phase after infection with

JDVTab87. The animal then received 7 booster inoculations intramuscularly at 2 week

internals with tissue-derived JDV vaccine. Serum was taken 2 weeks after the final

inoculation. BIV hyperimmune serum was produced in a bovine lentivirus-free

animal with cell-culture preparations of BIV-R29 prepared as described in Chapter 4.

This cell culture material was emulsified in Freund’s incomplete adjuvant and

inoculated 3 times at 2-week intervals and serum was collected 2 weeks after the

final inoculation. The first inoculation was from freshly harvested cell culture

material while the final 2 inoculations were from frozen cell culture material. The

reactivity of the JDV hyperimmune serum has previously been reported (Desport et

al., 2005). The BIV hyperimmune serum reacted with JDV CA and BIV CA and SU

proteins on a WIB.

Page 119: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

109

Overlapping synthetic peptides

One hundred and fifty five overlapping synthetic peptides were constructed to cover

the entire amino acid sequence of MA, CA and SU of JDVTab87 (accession number

U21603). The peptides were designed to be 16 amino acids long and to overlap each

other by 11 amino acids. This length allows for coverage of the three regions while

the minimal overlap enables mapping to a fine specificity and takes into account the

significant expense of peptide production. The peptides were synthesised by

automated 9-fluorenylmethyloxycarbonyl chemistry (Mimotopes, Australia) as

previously described (Valas et al., 2000). An amino-terminal biotinylated

tetrapeptide (Ser-Gly-Ser-Gly) was added to all peptides to facilitate epitope

accessibility and absorption to streptavidin-coated wells. All peptides were dissolved

in 200 µl of cell culture grade, 100% DMSO (Sigma-Aldrich). For use in the ELISA,

peptides were further diluted 1:200 in PBS-T (PBS containing 0.1% Tween-20

[Sigma] and 0.1% sodium azide [Sigma]).

ELISA

Serum samples were tested at a dilution of 1:50 against each peptide in a standard

ELISA according to the peptide manufacturer’s instructions. Plates were coated with

100 µl of 5 µg/ml stock streptavidin (Sigma-Aldrich) and left to evaporate overnight

at 37°C. Plates were blocked for 2 h with 200 µl of blocking solution (0.01 M PBS

containing 2.5% gelatin [Bio-Rad], 5% rabbit serum [Invitrogen], 1% sodium

caseinate [MP Biomedicals], 0.1% Tween 20 [Sigma]). All incubation steps were

conducted at room temperature (~25°C) on a rocker platform. Plates were then

washed 4 times with PBS-T. Peptides were further diluted 1:4 in PBS-T and 100 µl

of peptide solution were added to each ELISA well, in duplicate, and incubated for

1 h. Plates were again washed 4 times. Serum was diluted 1:50 using PBS-T and

Page 120: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

110

100 µl added to each well. Plates were incubated for 1 h then washed a further 4

times. Goat anti-bovine IgG HRP-conjugated (MP Biomedicals) was diluted 1:2 000

in 0.01 M PBS with additional 1% sheep serum (Invitrogen), 0.1% Tween 20 and

0.1% sodium caseinate, 100 µl was added to each well. Plates were incubated for 1 h

then washed 4 times with PBS-T and 2 times with PBS only. Substrate (SigmaFast

OPD; Sigma) was prepared as per the manufacturer’s instructions and 100 µl was

added to each well. After colour development, the reactions were stopped with 3 M

H2SO4 and read at 450 nm.

Statistical analysis

A model previously developed and described to map linear epitopes in HIV-1 was

used to analyse the data produced in the ELISA (Loomis-Price et al., 1997). The cut-

off for reactivity of individual peptides was determined as follows: the median and

first quartile values (of the optical densities) were determined separately for each

serum sample and for each block of peptides. The data were normalised by

subtracting the median reactivity of the set from each value in the set and the

standard deviation was calculated:

Standard deviation (σ) = (median – first quartile)/0.675

The data were then divided by the calculated standard deviation and expressed as

normalised reactivity (σ) compared to the median. Measurements above a cut-off of

5 σ were considered positive.

The overlapping peptide set was screened with pre-infection sera to determine the

level of background binding present for each serum sample. Unless the level of

reactivity increased over time, the peptide was scored as non-reactive.

Immunoreactive (IR) peptides were defined as those which reacted with 75% or

Page 121: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

111

greater of the serum samples tested and immunodominant (ID) peptides defined as

those which reacted with 100% of the serum samples tested, as previously reported

(Ball et al., 1992).

Results

When the overlapping peptides were tested against sera from cattle after infection

with JDVTab87, a number of peptides were not reactive against any of the sera tested,

including peptide MA18 in MA (Figure 6.1A), peptides CA34, CA35, CA42 and

CA62 in CA (Figure 6.2A) and peptides SU78, SU85, SU92, SU121, SU138,

SU145, SU148 and SU149 in SU (Figure 6.3A). One peptide (MA24) was identified

as IR in MA (Figure 6.1A), one peptide (CA70) was identified as IR in CA

(Figure 6.2A) and 4 peptides (SU112, SU152, SU154 and SU155) were as IR in SU

(Figure 6.3A). The locations of these peptides on the linear amino acid sequence of

JDVTab87 are shown in Figures 6.5 and 6.6. No peptides were identified as ID in MA,

CA or SU using the Ball et al. (1992) definitions. Using the less stringent definitions

reported in a separate study (Valas et al., 2000), whereby IR peptides were defined

by greater than 20% reactivity with bovine sera and ID peptides defined by greater

than 58% reactivity, 22 MA peptides were IR and 1 was ID, 27 CA peptides were IR

and 4 were ID, and 44 SU peptides were IR and 8 were ID.

When the overlapping peptides were screened with serum taken from cattle

experimentally infected with JDVPul01, a number of peptides were not reactive

against the JDVPul01 sera, including peptides MA6, MA10, MA13, MA14 and MA22

in MA (Figure 6.1B), peptides CA26, CA27, CA38, CA39, CA43, CA44, CA55,

CA62, CA63, CA67, CA68 and CA70 in CA (Figure 6.2B) and peptides SU78-80,

SU83, SU84, SU86- 88, SU90, SU94, SU95, SU111, SU112, SU114, SU116,

SU131, SU135, SU136, SU147 and SU154 in SU (Figure 6.3B). Numerous peptides

Page 122: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

112

were IR against the sera from JDVPul01 infected animals including peptides MA4,

MA15, MA21 and MA25 in MA (Figure 6.1B), peptides CA41, CA49, CA52 and

CA69 in CA (Figure 6.2B) and peptides SU100, SU106, SU108, SU110, SU115,

SU118, SU120, SU121, SU126, SU145 and SU150 in SU (Figure 6.3B). Peptides

CA53, SU96 and SU102 were ID.

When the overlapping peptides were screened with serum taken from cattle

experimentally infected with BIV, peptides not reactive with the BIV sera tested

included peptide MA10, MA12 and MA18 in MA (Figure 6.1C), CA33, CA42 and

CA63 in CA (Figure 6.2C) and peptides SU74, SU83, SU85, SU86, SU88, SU92,

SU93, SU95, SU103, SU105, SU111, SU114, SU119, SU135 and SU137 in SU

(Figure 6.3C). Peptide 25 in MA (Figure 6.1C), peptides CA29, CA53 and CA55 in

CA (Figure 6.2C) and peptides SU126, SU129, SU100, SU118 and SU123 in SU

(Figure 6.3C) were all IR and no peptides were ID against these sera.

The 2 hyperimmune serum samples tested reacted against different peptides.

JDVTab87 hyperimmune serum reacted particularly strongly against peptide SU83 as

well as peptide MA5 (Figure 6.4). In contrast, BIV hyperimmune serum reacted

strongly to peptides in 3 regions of CA: CA57 and 58, peptides CA44 and CA45, and

CA65 (Figure 6.4).

Page 123: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

113

1 5 9 13 17 21 25

0

25

50

75

100

Peptide number

Rea

ctiv

ity

( σσ σσ)

1 5 9 13 17 21 25

0

25

50

75

100

Peptide number

Rea

ctiv

ity

( σσ σσ)

1 5 9 13 17 21 25

0

25

50

75

100

Peptide number

Rea

ctiv

ity

( σσ σσ)

A

B

C

Figure 6.1. Reactivity to peptides derived from the MA sequence of JDVTab87 to

serum samples taken (A) 56-159 days after infection of cattle with JDVTab87, (B)

71-102 days after infection of cattle with JDVPul01 and (C) 35-101 days after infection

of cattle with BIV.

Page 124: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

114

26 31 36 41 46 51 56 61 66 71

0

25

50

75

100

Peptide number

Rea

ctiv

ity

( σσ σσ)

25 30 35 40 45 50 55 60 65 70

0

25

50

75

100

Peptide number

Rea

ctiv

ity

( σσ σσ)

25 30 35 40 45 50 55 60 65 70

0

25

50

75

100

Peptide number

Rea

ctiv

ity

( σσ σσ)

A

B

C

Figure 6.2. Reactivity to peptides derived from the CA sequence of JDVTab87 to

serum samples taken (A) 56-159 days after infection of cattle with JDVTab87, (B) 56-

159 days after infection of cattle with JDVTab87 and (C) 35-101 days after infection of

cattle with BIV.

Page 125: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

115

72 82 92 102 112 122 132 142

0

25

50

75

100

Peptide number

Rea

ctiv

ity

( σσ σσ)

72 82 92 102 112 122 132 142

0

25

50

75

100

Peptide number

Rea

ctiv

ity

( σσ σσ)

72 82 92 102 112 122 132 142

0

25

50

75

100

Peptide number

Rea

ctiv

ity

( σσ σσ)

A

B

C

Figure 6.3. Reactivity to peptides derived from the SU sequence of JDVTab87 to

serum samples taken (A) 56-159 days after infection of cattle with JDVTab87, (B) 71-

102 days after infection of cattle with JDVPul01 and (C) 35-101 days after infection of

cattle with BIV.

Page 126: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

116

10 20 30 40 50 60 70 80 90 100 110 120 130 140 150

-5

0

5

10

15

20

25

Peptide number

Rea

ctiv

ity

( σσ σσ)

10 20 30 40 50 60 70 80 90 100 110 120 130 140 150

-5

0

5

10

15

Peptide number

Rea

ctiv

ity

( σσ σσ)

A

B

Figure 6.4. Reactivity of hyperimmune serum to overlapping peptides spanning the

MA (peptides MA1-25), CA (peptides CA26-71) and SU (peptides SU72-155) of

JDV. A: reactivity with JDVTab87 hyperimmune sera. B: reactivity with BIV-R29

hyperimmune sera. Connecting lines are shown for clarity and are not meant to imply

continuous data. Horizontal line (-----) indicates the cut-off of 5 σ.

Page 127: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

117

JDV MKLSKLEKALKKVRVTPQRDDTYTIGNVLWAIRMCRLMGLDCCIDEAT-AAEVAILIGRF 59

Start gag ►

BIV MKRRELEKKLRKVRVTPQQDKYYTIGNLQWAIRMINLMGIKCVCDEECSAAEVALIITQF 60

** :*** *:*******:*. *****: ***** .***:.* ** *****::* :*

JDV QSLDLQDSPLKGKDEKAILTTLKVLWSLLAGHHPENSDMAEKYWEAWTIRERESQKEEEG 119

Peptide 24

BIV SALDLENSPIRGKEEVAIKNTLKVFWSLLAGYKPESTETALGYWEAFTYREREARADKEG 120

.:***::**::**:* ** .****:******::**.:: * ****:* ****:: ::**

JDV EITSIYPQLRKN---------------FPAVSTSDGSPRYDPDLTKQLKIWADATEKHGV 164

MA♦♦♦♦CA

Peptide 24

BIV EIKSIYPSLTQNTQNKKQTSNQTNTQSLPAITTQDGTPRFDPDLMKQLKIWSDATERNGV 180

**.****.* :* :**::*.**:**:**** ******:****::**

JDV DHHAVNILGVITANLTQSEIRLLLQSTPQWRLDIQLIESKLNAREHAHRVWKESHPEAPK 224

BIV DLHAVNILGVITANLVQEEIKLLLNSTPKWRLDVQLIESKVREKENAHRTWKQHHPEAPK 240

* *************.*.**:***:***:****:******:. :*:***.**: ******

JDV TDEIIGKGLTAAEQATLTTQECRDTYRQWVLEAALEVAQGKHDRPGPINIHQGPKEPYPE 284

BIV TDEIIGKGLSSAEQATLISVECRETFRQWVLQAAMEVAQAKHATPGPINIHQGPKEPYTD 300

*********::****** : ***:*:*****:**:****.** **************.:

JDV FVNKLVTALEGMAAPETTKQYLLDHLSVDHANEDCRAVLLPLGPSAPMERKLEACRAVGS 344

BIV FINRLVAALEGMAAPETTKEYLLQHLSIDHANEDCQSILRPLGPNTPMEKKLEACRVVGS 360

*:*:**:************:***:***:*******:::* ****.:***:******.***

JDV SKQKMQFLAEAFAAINVK-------------------GDGEVQRCYGCGKPGHIRRDCKN 385

CA♦♦♦♦NC

Peptide 70

BIV QKSKMQFLVAAMKEMGIQSPIPAVLPHTPEAYASQTSGPEDGRRCYGCGKTGHLKRNCKQ 420

.*.*****. *: :.:: * : :*******.**::*:**:

JDV QKCFKCGKPGHLQRNCKSKNGRRSSAPSGQRS----GYHQEKTS-VTPSAPPLVLD 436

◄ End gag

BIV QKCYHCGKPGHQARNCRSKNGKCSSAPYGQRSQPQNNFHQSNMSSVTPSAPPLILD 476

***::****** ***:****: **** **** .:**.: * ********:**

Figure 6.5. Linear representation of JDV MA and CA epitopes. A panel of sera taken from animals experimentally infected with JDVTab87 were reacted in ELISA against the panel of JDV MA and CA synthetic peptides. Bovine humoral epitopes are portrayed along the linear amino acid sequence of the JDVTab87 gag precursor protein. Grey highlighted sequences delineate immunoreactive peptides which are recognized by 75% or more of the bovine sera. The BIV gag precursor protein sequence is also shown below the JDV sequence for comparison. The sequences were aligned using ClustalW2 (Larkin, 2007). “*” indicates identical residues, “:” indicates conserved substitutions and “.” indicates semi-conserved substitutions.

Page 128: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

118

JDV MMEEGRKEEPEERGEKSTMRDLLQRAVDKGHLTAREALDRWTLEDHGEIHPWIILFCFAG 60

Start env ►

BIV MDQDLDGAERGERGGGS--EELLQEEINEGRLTAREALQTWINN--GEIHPWVLAGMLSM 56

* :: * *** * .:***. :::*:*******: * : ******:: ::

JDV AIGVIGGWGLRGELNVCMLIVLVVLVPIYWGIGEAARNIDSLDWKWIRKVFIVIIFVLVG 120

BIV GVGML--LGVYCQLPDTLIWILMFQLCLYWGLGETSRELDKDSWQWVRSVFIIAILGTLT 114

.:*:: *: :* :: :*:. : :***:**::*::*. .*:*:*.***: *: :

JDV LLG--------------------------------------------------------- 123

BIV MAGTALADDDQSTLIPNITKIPTKDTEPGCTYPWILILLILAFILGILGIILVLRRSNSE 174

: *

JDV ------------------------------------------------------------

BIV DILAARDTIDWWLSANQEIPPKFAFPIILISSPLAGIIGYYVMERHLEIFKKGCQICGSL 234

JDV ----------------------------------------GCSAQRQHVAMLLSPPGIRL 143

BIV SSMWGMLLEEIGRWLARREWNVSRVMVILLISFSWGMYVNRVNASGSHVAMVTSPPGYRI 294

.*. .****: **** *:

JDV P-STVDIPWFCISNAPIPDCVHWTVQK---PDQKHQQIENVMELQEVLDNATFFEVPDLF 199

BIV VNDTSQAPWYCFSSAPIPTCSSSQWGDKYFEEKINETLVKQVYEQAAKHSRATWIEPDLL 354

.* : **:*:*.**** * . :: :: : : : * . .. : : ***:

JDV DRVYLELARLDANSTGVPVNIPPTGISQVKGDCSTGDIQGMNETLSTRGTLGERTFLSIR 259

Peptide 112

BIV EEAVYELALLSANDS-----------RQVVVENGTDVCSSQNSSTNKGHPMTLLKLRGQV 403

:.. *** *.**.: ** : .*. .. *.: .. .: .: .

JDV PGGWFTNTTVWFCVHWPFGFIQRKEN-----LSEGSAQVRNCLDPINVTEPRVANYSYCP 314

Peptide 134

BIV SETWIGNSSLQFCVQWPYVLVGLNNSDSNISFNSGDWIATNCMHPITLNKS--------- 454

. *: *::: ***:**: :: ::. :..*. . **:.**.:.:.

JDV LEYKGKNYINKGLKCVGGRVDLSSNPEQHTDLLACGTFCQNFRNCDMVSRDILIG-YHPS 373

Peptide 134

BIV AQDLGKNFP--RLTFLDGQLSQLKN-----TLCGHNTNCLKFGNKSFSTNSLILCQDNPI 507

: ***: *. :.*::. .* * . .* * :* * .: :..::: :*

JDV QQKQHIYINHTFWEQANTQWILVQVPNYGFVPVPDTERPWKGGKPRGKRAVGMVIFLLVL 433

SU♦TM

Peptide 152 Peptide 154 Peptide 155

BIV GNDTFYSLSHSFSKQASARWILVKVPSYGFVVVNDTDTPP-SLRIRKPRAVGLAIFLLVL 566

:. . :.*:* :**.::****:**.**** * **: * . : * ****:.******

Figure 6.6. Linear representation of JDV SU epitopes. A panel of sera taken from animals experimentally infected with JDVTab87 were reacted in ELISA against the panel of JDV SU synthetic peptides. Bovine humoral epitopes are portrayed along the linear amino acid sequence of the JDV Env precursor protein. Grey highlighted sequences delineate immunoreactive peptides which are recognized by 75% or more of the bovine sera. Three immunoreactive peptides are shown at the carboxyl end of the protein; these are distinguished by the grey highlighted text (1), the underlined text (2) and the bolded text (3). The BIV Env precursor protein sequence is also shown below the JDV sequence for comparison. The sequences were aligned using

Page 129: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

119

ClustalW2 (Larkin, 2007). “*” indicates identical residues, “:” indicates conserved substitutions and “.” indicates semi-conserved substitutions.

Discussion

The investigations conducted were designed to use synthetic peptides to identify

antigenic sites on the MA, CA and SU that would react differentially to antibody to

JDV and BIV. Synthetic peptides have been used previously to map antigenic sites in

a number of lentiviruses, including EIAV (Ball et al., 1992; Grund et al., 1996;

Soutullo et al., 2007), the SRLV CAEV and VMV (Mordasini et al., 2006; Rosati et

al., 1999; Valas et al., 2000) and HIV-1 (Loomis-Price et al., 1997; Neurath et al.,

1990). They have also been used with other virus systems including human

cytomegalovirus (Greijer et al., 1999), Foot and mouth disease virus (Geysen et al.,

1987) and Epstein-Barr virus (Middeldorp et al., 1988). The peptides were able to

identify specific linear sites which were immunogenic in the native protein antigens.

It is recognised, however, that a limitation of these studies as well as the current

study is that the use of synthetic peptides cannot identify discontinuous

conformation-dependent epitopes which may represent important antigenic

determinants of viral proteins.

Another limitation of this current study was that it was necessary to use serum from

BIV-R29 infected animals to screen the peptides and examine for differences in the

reactions comparative to those in JDV-infected cattle. This BIV strain may not

necessarily have a close antigenic relationship to the putative non-pathogenic bovine

lentivirus in Indonesia. However, this was the only BIV serum available and sera

from antibody-positive cattle in Sulawesi, which would have been a suitable

alternative, were unavailable for this study.

Page 130: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

120

Sera from cattle experimentally infected with JDVTab87 or JDVPul01 were used to

screen the peptides for JDV-reactive epitopes as these are 2 reasonably well defined

strains that have been detected in Indonesia and the samples had a well documented

history. Of a number of JDV strains sequenced, JDVPul01 is the most divergent from

JDVTab87 in env and gag regions (Desport et al., 2007) and we sought to determine

whether the differences extended to the humoral response. The amino acid sequence

homology between the two strains in the entire env region and part of the gag regions

is 97% (Desport et al., 2007). Differences also existed between the reaction of cattle

infected with these 2 JDV strains in regards to peak viral loads and duration of

viraemia (Desport et al., 2009a). Differences were detected in the reactivity of the

peptides between the JDVTab87 and JDVPul01 sera. The JDVPul01 sera had a larger

number of IR peptides, with 3 ID peptides CA53, SU96 and SU102. These

differences could be attributed to the fewer number of cattle analysed and it would be

of interest to screen the peptides with more JDVPul01 sera to confirm these results.

The synthetic peptides used in this study encompassed the complete JDVTab87 MA,

CA and SU regions. The MA, CA and SU proteins were chosen for investigation for

a number of reasons. Firstly, the strongest and earliest immune responses against

JDV are directed at CA (Hartaningsih et al., 1994; Kertayadnya et al., 1993) although

a subset of cattle do not mount an immune response against CA (Desport et al.,

2009a; Ditcham et al., 2009). Matrix was chosen as BIV was reported to contain at

least one unique epitope in the CA that is absent in JDV, at the 6.4-kDa N terminus

of the 29-kDa CA adjacent to MA (Zheng et al., 2001) and strong antibody responses

against MA have been previously identified in experimentally infected animals

(Desport et al., 2005; Kertayadnya et al., 1993). SU was included as anti-Env

responses have been reported to persist beyond 190 weeks after BIV infection whilst

Page 131: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

121

the Gag response wanes 40 weeks after infection (Isaacson et al., 1995), env

sequences between JDV strains are reasonable well conserved (Desport et al., 2007)

and there are a significant number of differences between the sequence of the SU

regions of JDV and BIV (Chadwick et al., 1995b, Figure 6.6, Table 2.2). Little

information is currently available detailing the antigenicity of JDV SU due to

problems expressing recombinant SU.

In the ELISA used to examine differential reactivity of the peptides, a cut-off of 75%

and greater positive reactivity to represent a significant B-cell epitope was used, as

reported by others (Ball et al., 1992). Other studies have utilised lower cut-offs of

33% (Kusk et al., 1992) and 50% (Valas et al., 2000) but a higher cut-off was used in

the current study to identify highly reactive B-cell epitopes in the JDV proteins.

There is potential for the use of IR peptides, those reacting with a high percentage of

serum samples tested, and ID peptides reacting with all serum samples, to facilitate

the development of a peptide-based ELISA able to identify most animals infected

with JDV or both JDV and BIV depending on the peptides used. Several potentially

useful IR peptides were detected.

One IR MA peptide was identified which spanned amino acids 116 to 131 of the CA

and MA proteins. This peptide spanned the MA-CA border and indicates that the

antibodies which recognise this peptide are produced in response to the uncleaved

Gag precursor protein.

Page 132: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

122

Sera from long-term JDV infections (>12 months after infection) were reported to

recognise a protein of the same size as MA (Kertayadnya et al., 1993) and these

findings identify an epitope within MA. Some JDV-infected cattle (CB83-86) have

been reported not to respond to MA within 175 dpi when tested in an ELISA using

recombinant MA (Ditcham et al., 2009) but MA24 reacted with sera from all of these

cattle (Table 6.1). The difference between the responses reported by Ditcham et al.

(2009) and those in the current study may be due to differences associated with the

sequences covered by the antigens. The responses reported by Ditcham et al. (2009)

were those generated when using a full length recombinant MA while MA24 spans

the MA-CA border, encompassing additional sequence. Peptide ELISA has been

shown to be more sensitive than an ELISA using recombinant proteins (Rosati et al.,

1999). Coincidentally, most variation in JDV Gag occurs just before the predicted

cleavage point between the MA and CA proteins (Desport et al., 2007) and this,

combined with the identification of an IR peptide in this region, suggests this may be

a region under pressure from the immune system.

The IR CA peptide reported here is within amino acids 346 – 360 of the Gag

precursor protein and spans the CA-NC border. Like the MA response, it also

suggests that the antibodies are produced in response to the uncleaved Gag precursor

polyprotein. Hyperimmune sera against whole JDV and BIV were reported to not

recognise recombinant proteins encompassing this region, presumable because of the

differences in the way hyperimmune sera is raised compared to sera from natural or

experimental infections (Desport et al., 2005). As shown in Figure 6.1, JDV

hyperimmune serum did react to this peptide while BIV hyperimmune sera did not.

A highly immunogenic domain has been reported in a similar location for EIAV

(Chong et al., 1991b).

Page 133: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

123

There were no IR peptides found within the MHR in CA when the peptides were

screened with JDVTab87 sera. The lack of an JDVTab87 IR peptide in this region is

surprising given that previous studies have shown that it is likely to be an epitopic

domain within JDV (Desport et al., 2005). The MHR is conserved among

retroviruses and is essential for virus assembly, maturation and infectivity

(Mammano et al., 1994) and is assumed to account for some of the cross-reactivity

observed between the Gag proteins of HIV, EIAV and BIV. Previous studies have

shown cross-reactivity between BIV hyperimmune sera and the JDV MHR (Desport

et al., 2005), although confirmation is required. Peptides CA52-59 spanned the MHR

and although they were not identified as IR in the current study, a number of peptides

had high reactivities against JDVTab87 sera, including CA53–55, 57 and CA58.

Peptide CA57 had the second highest reactivity of all the CA peptides with a

reactivity of 62.5%. The CA53 peptide was a JDVPul01 ID peptide and when screened

with BIV sera, peptides CA53 and CA55 were found to be IR, confirming that this is

one region of cross-reactivity. The BIV hyperimmune serum used in this study also

strongly reacted to this region (Figure 6.4). JDV hyperimmune serum also reacted to

the MHR, albeit more weakly than the BIV hyperimmune serum.

Surprisingly, the major homology region identified as an immunodominant domain

in the CA proteins of many of the lentiviruses was not identified as IR or ID using

this method of analysis.This may indicate that the level of stringency was too high or

may be due to the characteristics of the sera chosen or the conformation of the

peptides. Previous studies have shown that conformational changes to peptides, such

as converting the peptides to a cyclical form, can help to increase reactivity (Scobie

et al., 1999). Further studies are required to clarify this discrepancy.

Page 134: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

124

Five IR peptides were found within SU. Peptide SU112 spanned amino acids 201–

216 in the central portion of the protein. The most IR peptide, reactive with 87.5% of

the sera tested, was SU134 and this spanned amino acids 311–326. The remaining 3

peptides, SU152, SU154 and SU155, clustered around the carboxyl end of SU and

spanned amino acids 401–430. The reduction in reactivity for peptide SU153

compared with the reactivity for peptides SU152 and SU154 indicated that there may

be 2 different epitopes at this location, as reported in CAEV SU (Valas et al., 2000).

The clustering of IR peptides around the carboxyl end of SU is similar to CAEV

(Bertoni et al., 2000; Valas et al., 2000), HIV-1 (Palker et al., 1987) and EIAV (Ball

et al., 1992; Grund et al., 1996), although these viruses also have IR regions at the

amino end of SU which were not identified in this current study of JDV. FIV also has

an epitope at the carboxyl end of SU, progressing into the start of TM (Pancino et al.,

1993), similar to the region encompassed by peptides SU154 and SU155 in the

current study. It was suggested that the antigenicity of terminal segments of proteins,

including lentiviral SU glycoproteins, is because these regions are frequently surface

orientated and thus exposed and immunogenic in their native state (Ball et al., 1992;

Valas et al., 2000; Van Regenmortel, 1999b).

No ID peptides were identified with the JDVTab87 serum. This might be associated

with the outbred nature of the Bali cattle population, as has been suggested in the T-

cell epitope mapping in FIV (Dean et al., 2004). A number of ID peptides were

identified with the JDVPul01 serum including 1 in CA and 2 in SU (Figures 6.2 and

6.3). Reactivities with the JDVTab°/87 sera to these peptides ranged from 25 to 50%

(Figures 6.2 and 6.3). Further investigations are required to determine whether the

differences are attributed to strain variation or to the number of samples analysed.

Page 135: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

125

The differential reaction of some peptides to JDV sera compared to BIV sera

suggests some of these peptides could be used to form antigens for potential JDV-

specific and broadly reactive bovine lentivirus serological assays. Peptides which

were reactive against 50% of sera taken from JDVTab87, JDVPul01 and BIV-R29 would

be useful to include in a bovine lentivirus serological assay as these should detect a

majority of cattle infected with these bovine lentiviruses. The areas encompassed by

these peptides are significant regions of cross-reactivity since a majority of both JDV

and BIV sera reacted to these peptides.

Peptides that were not recognised by BIV sera are potential candidates for inclusion

in a JDV-specific ELISA antigen, an approach previously suggested in CAEV (Valas

et al., 2000). Unfortunately, none of these peptides were IR or ID against the JDV

sera only. However, a combination of 5 of these peptides, SU93, SU95, SU103,

SU119 and SU135 from SU, would have reacted with 87.5% of the JDV-only sera,

although this specific combination would not have detected animals infected with

JDVPul01. The addition of 2 extra peptides, MA18 and MA19 would provide

reactivity with the JDVPul01 sera. It is important to note, however, that this approach

would potentially provide a JDV-specific serological assay and not a BIV-specific

assay. Previous diagnostic assays have combined multiple antigens in an ELISA

format (Khan et al., 2006) and it would be practical to combine these peptides in this

format.

This study needs to be extended by testing sera from naturally infected cattle as well

as by testing the longitudinal responses to the IR peptides in experimentally infected

cattle, and applying the tests in a clinical setting in Indonesia. Consideration could

also be given to using selected peptides in the formulation of vaccines capable of

protecting against JDV, as has been reported in a number of lentivirus systems

Page 136: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

126

including SIV (Belyakov et al., 2001; Nehete et al., 2008) and HIV-1 (Hovanessian

et al., 2004). Due to the persistent nature of the anti-TM antibody response in BIV

infections (Isaacson et al., 1995, Scobie et al., 1999), the TM glycoprotein may also

be a promising linear antigenic target and therefore extending this study to the TM

glycoprotein may yield a potential antigen for inclusion in a differential serological

assay. It would also be of interest to test the response of cattle superinfected with

both BIV and JDV and their pattern of reactivity over time.

Page 137: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

127

Chapter 7: General discussion

The close genetic and antigenic relationship between BIV and JDV raised 2 issues

that were investigated and are reported in this thesis. First, in animals that are

infected with both viruses, it was hypothesised that there might be cross-protective

immunity or other interaction between the 2 viruses that could modify their

pathogenesis. Second, there is a need for serological tests that will differentiate

antibody to the pathogenic JDV and other non-pathogenic bovine lentiviruses. The

presence of a BIV-like virus in cattle on the island of Sulawesi where Jembrana

disease does not occur could have a marked effect on the events that might occur if

JDV spreads to that island. In an endemic area, previous infection with a non-

pathogenic bovine lentivirus like BIV might ameliorate the effect of subsequent

infection with JDV, resulting in subclinical JDV infections. Immunosurveillance for

JDV infection would be affected by the presence of a second antigenically cross-

reactive but non-pathogenic bovine lentivirus in the cattle population of Indonesia.

The investigations reported in this thesis have provided information that clarifies

these issues.

Evidence was provided previously for the occurrence of a non-pathogenic bovine

lentivirus in Bali cattle in Indonesia, although this was based on serological evidence

only and the virus has not been detected. This serological evidence, particularly that

presented by Barboni et al. (2001), is difficult to evaluate because other

investigations have demonstrated a very close antigenic relationship between JDV

and BIV and there are no reagents or tests available that allow differentiation of

antibody to the 2 types of virus in cattle sera (Desport et al, 2005). Antibody to JDV

was detected in B. javanicus in Sulawesi where there is no evidence of Jembrana

disease (Hartaningsih, personal communication) suggesting the presence of a non-

Page 138: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

128

pathogenic bovine lentivirus antigenically related to JDV. Blood from a seropositive

animal in Sulawesi was inoculated into Bali cattle in an attempt to transmit the non-

pathogenic bovine lentivirus but these attempts were unsuccessful (Hartaningsih,

personal communication). For further investigation of the effects of the Indonesian

strain of this non-pathogenic bovine lentivirus in Bali cattle, an attempt was made to

detect the virus in cattle on the island of Bali (Chapter 3). A large number of cattle

were screened using PCR and serological assays which detect both BIV and JDV.

While a number of cattle were identified that contained proviral JDV DNA, BIV

proviral DNA was not detected and the investigations reported in Chapter 3 therefore

provide no evidence for the occurrence of a second non-pathogenic bovine lentivirus

in these cattle. Isolation of virus in cell culture was not attempted in this study due to

a lack of suitable facilities.

The lack of evidence of a BIV-like virus reported in Chapter 3, however, was

insufficient to eliminate the possibility that there is a second non-pathogenic bovine

lentivirus in the cattle population of Bali as only a limited number of samples from

one area were screened and the seroprevalence was lower than expected. Previous

investigators have concluded that BIV is difficult to detect in cattle. While numerous

studies have reported serological evidence for BIV infection (Barboni et al., 2001;

Bhatia et al., 2008; Horzinek et al., 1991; Meas et al., 1998; Meas et al., 2000a;

Whetstone et al., 1990) there have been only 3 reports of isolation of the virus from

cattle (Meas et al., 1998; Suarez et al., 1993; Van der Maaten et al., 1972). Suarez et

al. (1993) were only successful in the isolation of virus on 2 occasions from many

samples and only after 4 blind passages of samples in cell culture. A very exceptional

result was reported from Japan, where an unusually high isolation rate of BIV in cell

culture was reported from PBMC and milk-derived leukocytes from BIV-antibody-

Page 139: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

129

positive dairy cattle. These isolates had 99.0 to 99.7% nucleotide sequence identity

with the R29 strain of BIV within a 258 nucleotide amplicon from the pol region.

These authors were also able to identify BIV proviral DNA by nested PCR and

Southern blot in cattle and buffalo samples from Japan, Cambodia and Pakistan

(Meas et al., 1998; Meas et al., 2000a; Meas et al., 2000b). The reports by Meas and

colleagues are interesting as they suggest that infection with BIV is widespread and

reasonably easy to detect in buffalo and cattle in Asian countries, and that virus

isolation was possible from antibody-positive cattle.

The study of BIV infection in Bali cattle reported in Chapter 4 is the first report of

the pathogenesis of BIV infection in this species. Because of the unusual

susceptibility of Bali cattle to JDV and Malignant catarrhal fever virus (Soesanto et

al., 1990) it was hypothesised that these cattle might also show a greater

susceptibility to BIV than do B. taurus but their susceptibility appeared generally

similar to the effects reported in B. taurus (Scobie et al., 1999; Suarez et al., 1993;

Whetstone et al., 1990). We also considered that BIV in Bali cattle might have very

different effects to those observed in B. taurus, as some lentiviruses do not cause

disease in their natural host (VandeWoude et al., 2006) but do so in heterologous

hosts, a phenomenon best described with SIV strains (Apetrei et al., 2004) but also

recognised with other lentiviruses such as puma and lion lentivirus infection in

domestic cats (VandeWoude et al., 1997). It has been reported that infection of B.

taurus with JDV resulted in a mild disease only, of much less severity that observed

in B. javanicus (Soeharsono et al., 1995a), and the reciprocal event, greater

susceptibility of Bali cattle to BIV was a potential outcome.

Bali cattle inoculated with BIV did not develop clinical signs within 62 days of

infection and this suggested that if the putative bovine lentivirus present in Sulawesi

Page 140: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

130

is indeed related to BIV-R29, then it should have no significant effect on animal

health in the Bali cattle population. A result that is significant in terms of the optimal

method for detection of natural BIV infection in Bali cattle, and possibly B. taurus,

was that there was a transient period of viraemia detected after infection and it is

during this period that the virus would be most easily detected. However, in natural

infections, the timing of infection is unknown and therefore these results show that

the choice of assay is an important one. If a recent infection is suspected, then a

genome based assay would be most likely to detect infection but if a longer term

infection is suspected then a ELISA using a TM peptide would be more appropriate.

This transient period of viraemia was analogous to that which occurs in other animal

lentivirus infections such as JDV infection of Bali cattle (Stewart et al., 2005), EIAV

infection of horses (Harrold et al., 2000) and SIVsmm-PBj14 infection of pig-tailed

macaques (Dewhurst et al., 1990; Fultz et al., 1989), although BIV was found to have

significantly lower titres than these viruses. It is suggested that BIV infection of B.

taurus should be re-examined using sensitive techniques such as qRT-PCR to see

whether a similar acute phase viraemia is produced. Sensitive techniques need to be

used in such an investigation as the level of viraemia in Bali cattle infected with BIV

was low and virus was not detected consistently in any animal, reflecting the

difficulty of detecting BIV infection even in experimentally infected cattle. The

majority of infected cattle had a significant TM antibody response but a poor CA

response but this occurred after the transient viraemic period, suggesting that

detection of BIV proviral DNA would be most successful in antibody-negative cattle,

similar to the result obtained for JDV in field cattle (Chapter 3). The ability to detect

BIV prior to the onset of an antibody response but not afterwards is in contrast to the

Page 141: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

131

report of the high frequency of isolation of BIV from antibody-positive cattle in Asia

(Meas et al., 1998).

A limitation of this current study of BIV-R29 in Bali cattle is that an Indonesian BIV

isolate was not discovered and could not be used for experimental studies, and the

BIV-R29 isolate used might not reflect what would occur with the putative BIV-like

virus present in Indonesia. However, other Asian isolates have been shown to be

closely related to BIV-R29 (Meas et al., 1998). The R29 strain of BIV may have a

different pathogenesis to the BIV-like virus present in Indonesian cattle. The results

obtained may also not be typical of BIV infection under field conditions as it is

considered by some that the BIV-R29 strain may have, since its isolation in 1969

(Van der Maaten et al., 1972), become attenuated during its prolonged storage and

passage in cell culture (Whetstone et al., 1997). However, an unsuccessful attempt

was made to increase the virulence of BIV-R29 by its serial passage through cattle

(Whetstone et al., 1990), a technique known to increase the virulence of EIAV

(Orrego et al., 1982), and perhaps this inability to increase its virulence reflects a

natural low pathogenicity. Other BIV strains isolated from Florida were thought to be

more pathogenic than BIV-R29, causing a B-cell lymphocytosis in experimentally

infected cattle, but the effects of the Florida isolates in cattle were still very mild

(Suarez et al., 1993; Whetstone et al., 1997). It is possible, however, that the

inoculation of the Florida isolates into Bali cattle may produce results different to

that observed with the R29 strain.

The lower antibody response to the CA versus the TM antigen was an unexpected

finding in BIV-infected Bali cattle. Animals infected with lentiviruses normally

develop a strong antibody response to CA and this response is the first to be detected

(Rosati et al., 2004; Soutullo et al., 2007). A strong response to CA has also been

Page 142: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

132

documented in Bali cattle infected with JDV (Hartaningsih et al., 1994) although

there is a subset of JDV infected Bali cattle that do not develop a detectable CA

antibody response (Desport et al., 2009a; Ditcham et al., 2009). A loss of Gag-

reactivity in the presence of a sustained Env response has previously been reported in

B. taurus infected with BIV (Isaacson et al., 1995).

In several lentivirus systems, infection with a non-pathogenic lentivirus is able to

protect against subsequent superinfection with a closely related pathogenic lentivirus

(Cranage et al., 1998; VandeWoude et al., 2002). An investigation of whether the

same was true for BIV and JDV in Bali cattle showed that BIV-R29 infection did not

induce a protective immune response against subsequent infection 42 days later with

JDV. All BIV-infected animals inoculated with JDV became infected and had a

viraemic phase and a febrile response typical of JDV infection. However, while

previous BIV infection did not protect against subsequent JDV infection, there was

no enhancement of JDV infection even though it did cause an earlier onset and

resolution of fever that was associated with JDV infection. The result suggests that if

BIV-R29 is related to the BIV-like lentivirus circulating within the cattle population

of Sulawesi, if JDV were introduced into the cattle population of Sulawesi it would

not lead to disease enhancement.

The lack of any evidence of a protective immunity against JDV as a consequence of

previous BIV infection was unexpected, considering the extensive antigenic cross-

reactivity of JDV and BIV proteins and evidence of an antibody response to BIV at

the time of superinfection with JDV. It will be necessary to determine if protection

against JDV can be achieved by altering the parameters of the superinfection.

Perhaps if the dose of BIV used to infect the cattle was increased this would affect

the result: higher doses of SIVmacC8 provided greater protection against challenge

Page 143: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

133

with SIVmac (Cranage et al., 1998). Alternatively, the time interval between BIV and

JDV infection could be important: greater protection has been provided by a

decreased time of 21 days between superinfection with closely related simian

lentiviruses (Stebbings et al., 2004) but it is also possible that a longer interval may

be required for the development of a cell-mediated immune response. Although

desirable, analysis of cell-mediated immune responses was not possible and so the

type of immune response required to control infection could not be adequately

assessed.

An important technical requirement for immunosurveillance of JDV infection in

Indonesia is the development of reliable and specific serological tests. JDV and BIV

contain an extensive array of cross-reactive epitopes on several proteins (Desport et

al., 2005; Kertayadnya et al., 1993) and using serological assays, differentiation of

infection by the 2 viruses using serological assays is currently not possible. The

investigation reported in Chapter 6 where a series of overlapping peptides spanning

the MA, CA and SU regions of JDV was used to identify epitopes that would react

specifically to JDV and not BIV has identified potential peptides that could be used

for this purpose. Seven peptides were identified within the MA-CA and the CA-NC

junctions, while 5 were identified in SU, that reacted with >75% of sera from JDV-

infected cattle. Unfortunately, these peptides also reacted with some sera from BIV-

infected cattle and could not therefore be used for the development of a JDV-specific

serological assay. A combination of these peptides could, however, be used as an

alternative antigen for the development of a broadly-reactive bovine lentivirus

serological test. A number of peptides were identified which reacted with sera from

JDV-infected cattle only and hence have potential for development of a JDV-specific

serological assay. These peptide combinations must now be tested using an extensive

Page 144: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

134

array of sera from naturally infected cattle from areas of Indonesia where infection

with both BIV and JDV have been suspected, such as Bali, and in regions where

infection with a non-pathogenic BIV is suspected and where Jembrana disease has

not been detected, such as Sulawesi. A longitudinal series of serum samples from

individual cattle also needs to be tested to determine how the antibody response to

the peptides changes over time.

In conclusion, the results reported in this thesis have made a significant original

contribution to our understanding of BIV infection in Bali cattle that indicate BIV is

unlikely to be a significant pathogen in these cattle. The results obtained have also

provided insights into the interaction of BIV and JDV in Bali cattle which suggest

that previous BIV infection will not provide significant cross-protective immunity

against subsequent JDV infection, and that BIV infection is unlikely to provide a

simple method of vaccinating cattle against JDV infection. The results have also

provided information about epitopes of the Gag and SU proteins of JDV that indicate

there are potential epitopes on these proteins that could be used for the development

of JDV-specific serological tests needed in Indonesia.

Page 145: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

135

References

Allen, T. M., and Altfeld, M. (2003). HIV-1 superinfection. J Allergy Clin Immunol. 112(5), 829-35; quiz 836.

Apetrei, C., Gormus, B., Pandrea, I., Metzger, M., ten Haaft, P., Martin, L. N., Bohm, R., Alvarez, X., Koopman, G., Murphey-Corb, M., Veazey, R. S., Lackner, A. A., Baskin, G., Heeney, J., and Marx, P. A. (2004). Direct inoculation of simian immunodeficiency virus from sooty mangabeys in black mangabeys (Lophocebus aterrimus): first evidence of AIDS in a heterologous African species and different pathologic outcomes of experimental infection. J. Virol. 78(21), 11506-18.

Balfe, P., Simmonds, P., Ludlam, C. A., Bishop, J. O., and Brown, A. J. (1990). Concurrent evolution of human immunodeficiency virus type 1 in patients infected from the same source: rate of sequence change and low frequency of inactivating mutations. J. Virol. 64(12), 6221-33.

Ball, J. M., Rushlow, K. E., Issel, C. J., and Montelaro, R. C. (1992). Detailed mapping of the antigenicity of the surface unit glycoprotein of equine infectious anemia virus by using synthetic peptide strategies. J. Virol. 66(2), 732-42.

Baltimore, D. (1970). RNA-dependent DNA polymerase in virions of RNA tumour viruses. Nature. 226(5252), 1209-11.

Barboni, P., Thompson, I., Brownlie, J., Hartaningsih, N., and Collins, M. E. (2001). Evidence for the presence of two bovine lentiviruses in the cattle population of Bali. Vet. Microbiol. 80(4), 313-27.

Barker, E., Barnett, SW, Stamatatos, L, Levy, JA (1995). The Human Immunodeficiency Viruses. In "The Retroviridae" (J. Levy, Ed.), Vol. 4, pp. 1-96. 4 vols. Plenum Press, New York.

Barlough, J., East, N., Rowe, J. D., Van Hoosear, K., DeRock, E., Bigornia, L., and Rimstad, E. (1994). Double-nested polymerase chain reaction for detection of caprine arthritis-encephalitis virus proviral DNA in blood, milk, and tissues of infected goats. J Virol Methods. 50(1-3), 101-13.

Baron, T., Mallet, F., Polack, B., Betemps, D., and Belli, P. (1995). The bovine immunodeficiency-like virus (BIV) is transcriptionally active in experimentally infected calves. Arch. Virol. 140(8), 1461-7.

Barre-Sinoussi, F., Chermann, J. C., Rey, F., Nugeyre, M. T., Chamaret, S., Gruest, J., Dauguet, C., Axler-Blin, C., Vezinet-Brun, F., Rouzioux, C., Rozenbaum, W., and Montagnier, L. (1983). Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS). Science. 220(4599), 868-71.

Page 146: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

136

Beebe, A. M., Dua, N., Faith, T. G., Moore, P. F., Pedersen, N. C., and Dandekar, S. (1994). Primary stage of feline immunodeficiency virus infection: viral dissemination and cellular targets. J. Virol. 68(5), 3080-91.

Belyakov, I. M., Hel, Z., Kelsall, B., Kuznetsov, V. A., Ahlers, J. D., Nacsa, J., Watkins, D. I., Allen, T. M., Sette, A., Altman, J., Woodward, R., Markham, P. D., Clements, J. D., Franchini, G., Strober, W., and Berzofsky, J. A. (2001). Mucosal AIDS vaccine reduces disease and viral load in gut reservoir and blood after mucosal infection of macaques. Nat. Med. 7(12), 1320-6.

Benson, R. E., Sanfridson, A., Ottinger, J. S., Doyle, C., and Cullen, B. R. (1993). Downregulation of cell-surface CD4 expression by simian immunodeficiency virus Nef prevents viral super infection. J Exp Med. 177(6), 1561-6.

Berry, N., Stebbings, R., Ferguson, D., Ham, C., Alden, J., Brown, S., Jenkins, A., Lines, J., Duffy, L., Davis, L., Elsley, W., Page, M., Hull, R., Stott, J., and Almond, N. (2008). Resistance to superinfection by a vigorously replicating, uncloned stock of simian immunodeficiency virus (SIVmac251) stimulates replication of a live attenuated virus vaccine (SIVmacC8). J Gen Virol. 89(Pt 9), 2240-51.

Bertoni, G., Hertig, C., Zahno, M. L., Vogt, H. R., Dufour, S., Cordano, P., Peterhans, E., Cheevers, W. P., Sonigo, P., and Pancino, G. (2000). B-cell epitopes of the envelope glycoprotein of caprine arthritis-encephalitis virus and antibody response in infected goats. J Gen Virol. 81(Pt 12), 2929-40.

Bertoni, G., Zahno, M. L., Zanoni, R., Vogt, H. R., Peterhans, E., Ruff, G., Cheevers, W. P., Sonigo, P., and Pancino, G. (1994). Antibody reactivity to the immunodominant epitopes of the caprine arthritis-encephalitis virus gp38 transmembrane protein associates with the development of arthritis. J. Virol. 68(11), 7139-47.

Beyer, J. C., Chebloune, Y., Mselli-Lakhal, L., Hotzel, I., Kumpula-McWhirter, N., and Cheevers, W. P. (2001). Immunization with plasmid DNA expressing the caprine arthritis-encephalitis virus envelope gene: quantitative and qualitative aspects of antibody response to viral surface glycoprotein. Vaccine. 19(13-14), 1643-51.

Bhatia, S., Sood, R., Bhatia, A. K., Pattnaik, B., and Pradhan, H. K. (2008). Development of a capsid based competitive inhibition enzyme-linked immunosorbent assay for detection of bovine immunodeficiency virus antibodies in cattle and buffalo serum. J Virol Methods. 148(1-2), 218-25.

Biek, R., Ruth, T. K., Murphy, K. M., Anderson, C. R., Jr., Johnson, M., DeSimone, R., Gray, R., Hornocker, M. G., Gillin, C. M., and Poss, M. (2006). Factors associated with pathogen seroprevalence and infection in Rocky Mountain cougars. J Wildl Dis. 42(3), 606-15.

Biek, R., Ruth, T.K., Murphy, K.M., Anderson, C.R., Poss, M (2006). Examining effects of persistent retroviral infection on fitness and pathogen susceptibility in a natural feline host. Can J Zool. 84, 365-373.

Page 147: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

137

Bird, P., Blacklaws, B., Reyburn, H. T., Allen, D., Hopkins, J., Sargan, D., and McConnell, I. (1993). Early events in immune evasion by the lentivirus maedi-visna occurring within infected lymphoid tissue. J. Virol. 67(9), 5187-97.

Blackard, J. T., Cohen, D. E., and Mayer, K. H. (2002). Human immunodeficiency virus superinfection and recombination: current state of knowledge and potential clinical consequences. Clin Infect Dis. 34(8), 1108-14.

Boshoff, C. H., Dungu, B., Williams, R., Vorster, J., Conradie, J. D., Verwoerd, D. W., and York, D. F. (1997). Detection of Maedi-Visna virus antibodies using a single fusion transmembrane-core p25 recombinant protein ELISA and a modified receiver-operating characteristic analysis to determine cut-off values. J Virol Methods. 63(1-2), 47-56.

Braun, M. J., Lahn, S., Boyd, A. L., Kost, T. A., Nagashima, K., and Gonda, M. A. (1988). Molecular cloning of biologically active proviruses of bovine immunodeficiency-like virus. Virology. 167(2), 515-23.

Brennan, G., Podell, M. D., Wack, R., Kraft, S., Troyer, J. L., Bielefeldt-Ohmann, H., and VandeWoude, S. (2006). Neurologic disease in captive lions (Panthera leo) with low-titer lion lentivirus infection. J Clin Microbiol. 44(12), 4345-52.

Brinkhof, J., and van Maanen, C. (2007). Evaluation of five enzyme-linked immunosorbent assays and an agar gel immunodiffusion test for detection of antibodies to small ruminant lentiviruses. Clin Vaccine Immunol. 14(9), 1210-4.

Brodie, S. J., Pearson, L. D., Snowder, G. D., and DeMartini, J. C. (1993). Host-virus interaction as defined by amplification of viral DNA and serology in lentivirus-infected sheep. Arch. Virol. 130(3-4), 413-28.

Brown, E. W., Yuhki, N., Packer, C., and O'Brien, S. J. (1994). A lion lentivirus related to feline immunodeficiency virus: epidemiologic and phylogenetic aspects. J. Virol. 68(9), 5953-68.

Brown, W. C., Bissey, L., Logan, K. S., Pedersen, N. C., Elder, J. H., and Collisson, E. W. (1991). Feline immunodeficiency virus infects both CD4+ and CD8+ T lymphocytes. J. Virol. 65(6), 3359-64.

Buchschacher, G. L., Jr. (2001). Introduction to retroviruses and retroviral vectors. Somat Cell Mol Genet. 26(1-6), 1-11.

Burkala, E. J., Ellis, T. M., Voigt, V., and Wilcox, G. E. (1999). Serological evidence of an Australian bovine lentivirus. Vet. Microbiol. 68(1-2), 171-7.

Burkala, E. J., Narayani, I., Hartaningsih, N., Kertayadnya, G., Berryman, D. I., and Wilcox, G. E. (1998). Recombinant Jembrana disease virus proteins as antigens for the detection of antibody to bovine lentiviruses. J Virol Methods. 74(1), 39-46.

Page 148: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

138

Burki, F., Rossmanith, W., and Rossmanith, E. (1992). Equine lentivirus, comparative studies on four serological tests for the diagnosis of equine infectious anaemia. Vet. Microbiol. 33(1-4), 353-60.

Campbell, R. S., and Robinson, W. F. (1998). The comparative pathology of the lentiviruses. J Comp Pathol. 119(4), 333-95.

Carpenter, S., Miller, L. D., Alexandersen, S., Whetstone, C. A., VanDerMaaten, M. J., Viuff, B., Wannemuehler, Y., Miller, J. M., and Roth, J. A. (1992). Characterization of early pathogenic effects after experimental infection of calves with bovine immunodeficiency-like virus. J. Virol. 66(2), 1074-83.

Carpenter, S., Vaughn, E. M., Yang, J., Baccam, P., Roth, J. A., and Wannemuehler, Y. (2000). Antigenic and genetic stability of bovine immunodeficiency virus during long-term persistence in cattle experimentally infected with the BIV(R29) isolate. J Gen Virol. 81(Pt 6), 1463-72.

Chadwick, B. J., Coelen, R. J., Sammels, L. M., Kertayadnya, G., and Wilcox, G. E. (1995a). Genomic sequence analysis identifies Jembrana disease virus as a new bovine lentivirus. J Gen Virol. 76 ( Pt 1), 189-92.

Chadwick, B. J., Coelen, R. J., Wilcox, G. E., Sammels, L. M., and Kertayadnya, G. (1995b). Nucleotide sequence analysis of Jembrana disease virus: a bovine lentivirus associated with an acute disease syndrome. J Gen Virol. 76 ( Pt 7), 1637-50.

Chadwick, B. J., Desport, M., Brownlie, J., Wilcox, G. E., and Dharma, D. M. (1998). Detection of Jembrana disease virus in spleen, lymph nodes, bone marrow and other tissues by in situ hybridization of paraffin-embedded sections. J Gen Virol. 79 ( Pt 1), 101-6.

Chakrabarti, L., Guyader, M., Alizon, M., Daniel, M. D., Desrosiers, R. C., Tiollais, P., and Sonigo, P. (1987). Sequence of simian immunodeficiency virus from macaque and its relationship to other human and simian retroviruses. Nature. 328(6130), 543-7.

Chatterji, U., de Parseval, A., and Elder, J. H. (2002). Feline immunodeficiency virus OrfA is distinct from other lentivirus transactivators. J. Virol. 76(19), 9624-34.

Cheevers, W. P., Beyer, J. C., and Knowles, D. P. (1997). Type 1 and type 2 cytokine gene expression by viral gp135 surface protein-activated T lymphocytes in caprine arthritis-encephalitis lentivirus infection. J. Virol. 71(8), 6259-63.

Cheevers, W. P., and McGuire, T. C. (1988). The lentiviruses: maedi/visna, caprine arthritis-encephalitis, and equine infectious anemia. Adv Virus Res. 34, 189-215.

Chen, H., He, J., Fong, S., Wilcox, G., and Wood, C. (2000). Jembrana disease virus Tat can regulate human immunodeficiency virus (HIV) long terminal repeat-

Page 149: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

139

directed gene expression and can substitute for HIV Tat in viral replication. J. Virol. 74(6), 2703-13.

Chen, H., Wilcox, G., Kertayadnya, G., and Wood, C. (1999). Characterization of the Jembrana disease virus tat gene and the cis- and trans-regulatory elements in its long terminal repeats. J. Virol. 73(1), 658-66.

Chong, Y. H., Ball, J. M., Issel, C. J., Montelaro, R. C., and Rushlow, K. E. (1991a). Analysis of equine humoral immune responses to the transmembrane envelope glycoprotein (gp45) of equine infectious anemia virus. J. Virol. 65(2), 1013-8.

Chong, Y. H., Payne, S. L., Issel, C. J., Montelaro, R. C., and Rushlow, K. E. (1991b). Characterization of the antigenic domains of the major core protein (p26) of equine infectious anemia virus. J. Virol. 65(2), 1007-12.

Clapham, P. R., and McKnight, A. (2002). Cell surface receptors, virus entry and tropism of primate lentiviruses. J Gen Virol. 83(Pt 8), 1809-29.

Clements, J. E., Narayan, O., and Cork, L. C. (1980). Biochemical characterization of the virus causing leukoencephalitis and arthritis in goats. J Gen Virol. 50(2), 423-7.

Clements, J. E., and Zink, M. C. (1996). Molecular biology and pathogenesis of animal lentivirus infections. Clin Microbiol Rev. 9(1), 100-17.

Coffin, J., Hughes, SH, Varmus, HE, Ed. (1997). Retroviruses: Cold Spring Harbour Laboratory Press.

Coggins, L., Norcross, N. L., and Nusbaum, S. R. (1972). Diagnosis of equine infectious anemia by immunodiffusion test. Am J Vet Res. 33(1), 11-8.

Connor, R. I., Montefiori, D. C., Binley, J. M., Moore, J. P., Bonhoeffer, S., Gettie, A., Fenamore, E. A., Sheridan, K. E., Ho, D. D., Dailey, P. J., and Marx, P. A. (1998). Temporal analyses of virus replication, immune responses, and efficacy in rhesus macaques immunized with a live, attenuated simian immunodeficiency virus vaccine. J. Virol. 72(9), 7501-9.

Corbin, A., and Sitbon, M. (1993). Protection against retroviral diseases after vaccination is conferred by interference to superinfection with attenuated murine leukemia viruses. J. Virol. 67(9), 5146-52.

Craigo, J. K., Montelaro, R.C. (2010). Lentivirus Tropism and Disease. In "Lentiviruses and Macrophages: Molecular and Cellular Interactions" (M. Desport, Ed.), pp. 410. Caister Academic Press.

Cranage, M. P., Sharpe, S. A., Whatmore, A. M., Polyanskaya, N., Norley, S., Cook, N., Leech, S., Dennis, M. J., and Hall, G. A. (1998). In vivo resistance to simian immunodeficiency virus superinfection depends on attenuated virus dose. J Gen Virol. 79 ( Pt 8), 1935-44.

Page 150: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

140

D'Mello, F., Kairo, S. K., Howard, C. R., and Partidos, C. D. (1999). Mapping the specificity of an anti-feline immunodeficiency virus monoclonal antibody using a filamentous phage displayed peptide library. Vaccine. 18(3-4), 371-5.

D.F.A.T. (2008). Map of Indonesia. Australian Department of Foreign Affairs and Trade, Canberra.

Dandekar, S., Beebe, A. M., Barlough, J., Phillips, T., Elder, J., Torten, M., and Pedersen, N. (1992). Detection of feline immunodeficiency virus (FIV) nucleic acids in FIV-seronegative cats. J. Virol. 66(7), 4040-9.

Daniel, M. D., Kirchhoff, F., Czajak, S. C., Sehgal, P. K., and Desrosiers, R. C. (1992). Protective effects of a live attenuated SIV vaccine with a deletion in the nef gene. Science. 258(5090), 1938-41.

Dawson, M. (1988). Lentivirus Diseases of Domesticated Animals. Journal of Comparative Pathology. 99, 401-419.

de Andres, D., Klein, D., Watt, N. J., Berriatua, E., Torsteinsdottir, S., Blacklaws, B. A., and Harkiss, G. D. (2005). Diagnostic tests for small ruminant lentiviruses. Vet. Microbiol. 107(1-2), 49-62.

Dean, G. A., LaVoy, A., and Burkhard, M. J. (2004). Peptide mapping of feline immunodeficiency virus by IFN-gamma ELISPOT. Vet. Immunol. Immunopathol. 100(1-2), 49-59.

Dean, G. A., Reubel, G. H., Moore, P. F., and Pedersen, N. C. (1996). Proviral burden and infection kinetics of feline immunodeficiency virus in lymphocyte subsets of blood and lymph node. J. Virol. 70(8), 5165-9.

Deng, H., Liu, R., Ellmeier, W., Choe, S., Unutmaz, D., Burkhart, M., Di Marzio, P., Marmon, S., Sutton, R. E., Hill, C. M., Davis, C. B., Peiper, S. C., Schall, T. J., Littman, D. R., and Landau, N. R. (1996). Identification of a major co-receptor for primary isolates of HIV-1. Nature. 381(6584), 661-6.

Deng, P., Cutlip, R. C., Lehmkuhl, H. D., and Brogden, K. A. (1986). Ultrastructure and frequency of mastitis caused by ovine progressive pneumonia virus infection in sheep. Vet. Pathol. 23(2), 184-9.

Desport, M., Ditcham, W. G., Lewis, J. R., McNab, T. J., Stewart, M. E., Hartaningsih, N., and Wilcox, G. E. (2009a). Analysis of Jembrana disease virus replication dynamics in vivo reveals strain variation and atypical responses to infection. Virology. 386(2), 310-6.

Desport, M., Stewart, M. E., Mikosza, A. S., Sheridan, C. A., Peterson, S. E., Chavand, O., Hartaningsih, N., and Wilcox, G. E. (2007). Sequence analysis of Jembrana disease virus strains reveals a genetically stable lentivirus. Virus Res. 126(1-2), 233-44.

Desport, M., Stewart, M. E., Sheridan, C. A., Ditcham, W. G., Setiyaningsih, S., Tenaya, W. M., Hartaningsih, N., and Wilcox, G. E. (2005). Recombinant Jembrana disease virus gag proteins identify several different antigenic

Page 151: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

141

domains but do not facilitate serological differentiation of JDV and nonpathogenic bovine lentiviruses. J Virol Methods. 124(1-2), 135-42.

Desport, M., Tenaya, I. W., McLachlan, A., McNab, T. J., Rachmat, J., Hartaningsih, N., and Wilcox, G. E. (2009b). In vivo infection of IgG-containing cells by Jembrana disease virus during acute infection. Virology. 393(2), 221-7.

Dewhurst, S., Embretson, J. E., Anderson, D. C., Mullins, J. I., and Fultz, P. N. (1990). Sequence analysis and acute pathogenicity of molecularly cloned SIVSMM-PBj14. Nature. 345(6276), 636-40.

Dharma, D. M. (1997). The Pathology of Jembrana Disease. In "Jembrana Disease and the Bovine Lentiviruses. Proceedings of a workshop 10-13 June 1996 Bali, Indonesia." (G. Wilcox, Soeharsono, S, Dharma, DMN, Copland, JW, Ed.), Vol. 75, pp. 26-33. Australian Centre for International Agricultural Research, Canberra.

Dharma, D. M., Ladds, P. W., Wilcox, G. E., and Campbell, R. S. (1994). Immunopathology of experimental Jembrana disease in Bali cattle. Vet. Immunol. Immunopathol. 44(1), 31-44.

Diehl, L.J., Mathiason-Dubard, C.K., O’Neil, L.L., Obert, L.A., and Hoover, E.A. (1995). Induction of accelerated feline immunodeficiency disease by acute-phase virus passage. J. Virol. 69, 6149-57.

Ditcham, W. G., Lewis, J. R., Dobson, R. J., Hartaningsih, N., Wilcox, G. E., and Desport, M. (2009). Vaccination reduces the viral load and the risk of transmission of Jembrana disease virus in Bali cattle. Virology. 386(2), 317-24.

Doranz, B. J., Rucker, J., Yi, Y., Smyth, R. J., Samson, M., Peiper, S. C., Parmentier, M., Collman, R. G., and Doms, R. W. (1996). A dual-tropic primary HIV-1 isolate that uses fusin and the beta-chemokine receptors CKR-5, CKR-3, and CKR-2b as fusion cofactors. Cell. 85(7), 1149-58.

Dua, N., Reubel, G., Moore, P. F., Higgins, J., and Pedersen, N. C. (1994). An experimental study of primary feline immunodeficiency virus infection in cats and a historical comparison to acute simian and human immunodeficiency virus diseases. Vet. Immunol. Immunopathol. 43(4), 337-55.

Duarte, A., and Tavares, L. (2006). Phylogenetic analysis of Portuguese Feline Immunodeficiency Virus sequences reveals high genetic diversity. Vet. Microbiol. 114(1-2), 25-33.

East, N. E., Rowe, J.D., Dahlberg, J.E., Theilen, G.H., Pedersen, N.C. (1993). Modes of transmission of caprine arthritis encephalitis virus infection. Small Ruminant Research. 10, 251-262.

Edinger, A. L., Amedee, A., Miller, K., Doranz, B. J., Endres, M., Sharron, M., Samson, M., Lu, Z. H., Clements, J. E., Murphey-Corb, M., Peiper, S. C., Parmentier, M., Broder, C. C., and Doms, R. W. (1997). Differential

Page 152: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

142

utilization of CCR5 by macrophage and T cell tropic simian immunodeficiency virus strains. Proc Natl Acad Sci U S A. 94(8), 4005-10.

Edinger, A. L., Clements, J. E., and Doms, R. W. (1999). Chemokine and orphan receptors in HIV-2 and SIV tropism and pathogenesis. Virology. 260(2), 211-21.

Egberink, H., and Horzinek, M. C. (1992). Animal immunodeficiency viruses. Vet Microbiol. 33(1-4), 311-31.

English, R. V., Johnson, C. M., Gebhard, D. H., and Tompkins, M. B. (1993). In vivo lymphocyte tropism of feline immunodeficiency virus. J. Virol. 67(9), 5175-86.

Fauquet, C. M., Mayo, M.A., Manilof, J., Desselberger, U., Ball, L.A., Ed. (2005). Virus Taxonomy: Eighth Report of the International Committee on Taxonomy of Viruses: Elsevier.

Feng, Y., Broder, C. C., Kennedy, P. E., and Berger, E. A. (1996). HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science. 272(5263), 872-7.

Ferens, W. A., and Hovde, C. J. (2007). The non-toxic A subunit of Shiga toxin type 1 prevents replication of bovine immunodeficiency virus in infected cells. Virus Res. 125(1), 29-41.

Finzi, D., Plaeger, S. F., and Dieffenbach, C. W. (2006). Defective Virus Drives Human Immunodeficiency Virus Infection, Persistence, and Pathogenesis. Clin Vaccine Immunol. 13(7), 715-721.

Flaming, K., van der Maaten, M., Whetstone, C., Carpenter, S., Frank, D., and Roth, J. (1993). Effect of bovine immunodeficiency-like virus infection on immune function in experimentally infected cattle. Vet. Immunol. Immunopathol. 36(2), 91-105.

Fletcher, T. M., 3rd, Brichacek, B., Sharova, N., Newman, M. A., Stivahtis, G., Sharp, P. M., Emerman, M., Hahn, B. H., and Stevenson, M. (1996). Nuclear import and cell cycle arrest functions of the HIV-1 Vpr protein are encoded by two separate genes in HIV-2/SIV(SM). Embo J. 15(22), 6155-65.

Foil, L. D., Adams, W. V., McManus, J. M., and Issel, C. J. (1987). Bloodmeal residues on mouthparts of Tabanus fuscicostatus (Diptera: Tabanidae) and the potential for mechanical transmission of pathogens. J. Med. Entomol. 24(6), 613-6.

Freed, E. O. (2001). HIV-1 replication. Somat Cell Mol Genet. 26(1-6), 13-33.

Fultz, P. N., McClure, H. M., Anderson, D. C., and Switzer, W. M. (1989). Identification and biologic characterization of an acutely lethal variant of simian immunodeficiency virus from sooty mangabeys (SIV/SMM). AIDS Res Hum Retroviruses. 5(4), 397-409.

Page 153: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

143

Fultz, P.N., and Zack, P.M. (1994) Unique lentivirus-host interactions: SIVsmmPBj14 infection of macaques. Virus Res. 32(2), 205-25.

Gallo, R. C., Salahuddin, S. Z., Popovic, M., Shearer, G. M., Kaplan, M., Haynes, B. F., Palker, T. J., Redfield, R., Oleske, J., Safai, B., and et al. (1984). Frequent detection and isolation of cytopathic retroviruses (HTLV-III) from patients with AIDS and at risk for AIDS. Science. 224(4648), 500-3.

Ganser-Pornillos, B. K., Yeager, M., and Sundquist, W. I. (2008). The structural biology of HIV assembly. Curr. Opin. Struct. Biol. 18(2), 203-17.

Gardner, M. B., and Luciw, P. A. (1989). Animal models of AIDS. FASEB J. 3(14), 2593-606.

Garvey, K. J., Oberste, M. S., Elser, J. E., Braun, M. J., and Gonda, M. A. (1990). Nucleotide sequence and genome organization of biologically active proviruses of the bovine immunodeficiency-like virus. Virology. 175(2), 391-409.

Gendelman, H. E., Narayan, O., Kennedy-Stoskopf, S., Kennedy, P. G., Ghotbi, Z., Clements, J. E., Stanley, J., and Pezeshkpour, G. (1986). Tropism of sheep lentiviruses for monocytes: susceptibility to infection and virus gene expression increase during maturation of monocytes to macrophages. J. Virol. 58(1), 67-74.

Geysen, H. M., Rodda, S. J., Mason, T. J., Tribbick, G., and Schoofs, P. G. (1987). Strategies for epitope analysis using peptide synthesis. J. Immunol. Methods. 102(2), 259-74.

Giannecchini, S., Pistello, M., Isola, P., Matteucci, D., Mazzetti, P., Freer, G., and Bendinelli, M. (2007). Role of Env in resistance of feline immunodeficiency virus (FIV)-infected cats to superinfection by a second FIV strain as determined by using a chimeric virus. J. Virol. 81(19), 10474-85.

Gnann, J. W., Jr., McCormick, J. B., Mitchell, S., Nelson, J. A., and Oldstone, M. B. (1987a). Synthetic peptide immunoassay distinguishes HIV type 1 and HIV type 2 infections. Science. 237(4820), 1346-9.

Gnann, J. W., Jr., Nelson, J. A., and Oldstone, M. B. (1987b). Fine mapping of an immunodominant domain in the transmembrane glycoprotein of human immunodeficiency virus. J. Virol. 61(8), 2639-41.

Goff, S. (2001). Retroviridae: The Retroviruses and Their Replication. 4 ed. In "Fundamental Virology" (D. Knipe, Howley, PM, Ed.), pp. 843-911. Lippincott, Williams & Wilkins, Philadelphia.

Gogolewski, R. P., Adams, D. S., McGuire, T. C., Banks, K. L., and Cheevers, W. P. (1985). Antigenic cross-reactivity between caprine arthritis-encephalitis, visna and progressive pneumonia viruses involves all virion-associated proteins and glycoproteins. J Gen Virol. 66 ( Pt 6), 1233-40.

Page 154: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

144

Goila-Gaur, R., and Strebel, K. (2008). HIV-1 Vif, APOBEC, and Intrinsic Immunity. Retrovirology. 5(51).

Gonda, M. A. (1992). Bovine immunodeficiency virus. Aids. 6(8), 759-76.

Gonda, M. A., Braun, M. J., Carter, S. G., Kost, T. A., Bess, J. W., Jr., Arthur, L. O., and Van der Maaten, M. J. (1987). Characterization and molecular cloning of a bovine lentivirus related to human immunodeficiency virus. Nature. 330(6146), 388-91.

Gonzalez, E. T., Licursi, M., Vila Roza, V., Bonzo, E., Mortola, E., Frossard, J. P., and Venables, C. (2008). Evidence of bovine immunodeficiency virus (BIV) infection: serological survey in Argentina. Res Vet Sci. 85(2), 353-8.

Gorrell, M. D., Brandon, M. R., Sheffer, D., Adams, R. J., and Narayan, O. (1992). Ovine lentivirus is macrophagetropic and does not replicate productively in T lymphocytes. J. Virol. 66(5), 2679-88.

Gottlieb, G. S., Nickle, D. C., Jensen, M. A., Wong, K. G., Grobler, J., Li, F., Liu, S. L., Rademeyer, C., Learn, G. H., Karim, S. S., Williamson, C., Corey, L., Margolick, J. B., and Mullins, J. I. (2004). Dual HIV-1 infection associated with rapid disease progression. Lancet. 363(9409), 619-22.

Greijer, A. E., van de Crommert, J. M., Stevens, S. J., and Middeldorp, J. M. (1999). Molecular fine-specificity analysis of antibody responses to human cytomegalovirus and design of novel synthetic-peptide-based serodiagnostic assays. J Clin Microbiol. 37(1), 179-88.

Grund, C. H., Lechman, E. R., Pezzuolo, N. A., Issel, C. J., and Montelaro, R. C. (1996). Fine specificity of equine infectious anaemia virus gp90-specific antibodies associated with protective and enhancing immune responses in experimentally infected and immunized ponies. J Gen Virol. 77 ( Pt 3), 435-42.

Gueye, A., Diop, O. M., Ploquin, M. J., Kornfeld, C., Faye, A., Cumont, M. C., Hurtrel, B., Barre-Sinoussi, F., and Muller-Trutwin, M. C. (2004). Viral load in tissues during the early and chronic phase of non-pathogenic SIVagm infection. J Med Primatol. 33(2), 83-97.

Guiguen, F., Mselli-Lakhal, L., Durand, J., Du, J., Favier, C., Fornazero, C., Grezel, D., Balleydier, S., Hausmann, E., and Chebloune, Y. (2000). Experimental infection of Mouflon-domestic sheep hybrids with caprine arthritis-encephalitis virus. Am J Vet Res. 61(4), 456-61.

Hahn, B. H., Shaw, G. M., De Cock, K. M., and Sharp, P. M. (2000). AIDS as a zoonosis: scientific and public health implications. Science. 287(5453), 607-14.

Hammond, S. A., Cook, S. J., Lichtenstein, D. L., Issel, C. J., and Montelaro, R. C. (1997). Maturation of the cellular and humoral immune responses to persistent infection in horses by equine infectious anemia virus is a complex and lengthy process. J. Virol. 71(5), 3840-52.

Page 155: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

145

Harrold, S. M., Cook, S. J., Cook, R. F., Rushlow, K. E., Issel, C. J., and Montelaro, R. C. (2000). Tissue sites of persistent infection and active replication of equine infectious anemia virus during acute disease and asymptomatic infection in experimentally infected equids. J. Virol. 74(7), 3112-21.

Hartaningsih, N., Dharma, D. M., Soeharsono, S., and Wilcox, G. E. (2001). The induction of a protective immunity against Jembrana disease in cattle by vaccination with inactivated tissue-derived virus antigens. Vet. Immunol. Immunopathol. 78(2), 163-76.

Hartaningsih, N., Sulistyana, K, Wilcox, GE (1997). Serological Test for JDV Antibodies and Antibody Response of Infected Cattle. In "Jembrana Disease and the Bovine Lentiviruses" (G. Wilcox, Soeharsono, S, Dharma, DMN, Copland, JW, Ed.). Australian Centre for International Agricultural Reseach.

Hartaningsih, N., Wilcox, G. E., Dharma, D. M., and Soetrisno, M. (1993). Distribution of Jembrana disease in cattle in Indonesia. Vet. Microbiol. 38(1-2), 23-9.

Hartaningsih, N., Wilcox, G. E., Kertayadnya, G., and Astawa, M. (1994). Antibody response to Jembrana disease virus in Bali cattle. Vet. Microbiol. 39(1-2), 15-23.

Hawkins, J. A., Adams, W. V., Jr., Wilson, B. H., Issel, C. J., and Roth, E. E. (1976). Transmission of equine infectious anemia virus by Tabanus fuscicostatus. J. Am. Vet. Med. Assoc. 168(1), 63-4.

Heaton, P. R., Johnstone, P., and Brownlie, J. (1998). Investigation of the cellular tropism of bovine immunodeficiency-like virus. Res Vet Sci. 65(1), 33-40.

Hemelaar, J., Gouws, E., Ghys, P. D., and Osmanov, S. (2006). Global and regional distribution of HIV-1 genetic subtypes and recombinants in 2004. Aids. 20(16), W13-23.

Herrmann, L. M., Cheevers, W. P., Marshall, K. L., McGuire, T. C., Hutton, M. M., Lewis, G. S., and Knowles, D. P. (2003a). Detection of serum antibodies to ovine progressive pneumonia virus in sheep by using a caprine arthritis-encephalitis virus competitive-inhibition enzyme-linked immunosorbent assay. Clin Diagn Lab Immunol. 10(5), 862-5.

Herrmann, L. M., Cheevers, W. P., McGuire, T. C., Adams, D. S., Hutton, M. M., Gavin, W. G., and Knowles, D. P. (2003b). Competitive-inhibition enzyme-linked immunosorbent assay for detection of serum antibodies to caprine arthritis-encephalitis virus: diagnostic tool for successful eradication. Clin Diagn Lab Immunol. 10(2), 267-71.

Horzinek, M., Keldermans, L., Stuurman, T., Black, J., Herrewegh, A., Sillekens, P., and Koolen, M. (1991). Bovine immunodeficiency virus: immunochemical characterization and serological survey. J Gen Virol. 72 ( Pt 12), 2923-8.

Hovanessian, A. G., Briand, J. P., Said, E. A., Svab, J., Ferris, S., Dali, H., Muller, S., Desgranges, C., and Krust, B. (2004). The caveolin-1 binding domain of

Page 156: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

146

HIV-1 glycoprotein gp41 is an efficient B cell epitope vaccine candidate against virus infection. Immunity. 21(5), 617-27.

Howard, K.E., Reckling, S.K., Egan, E.A., and Dean, G.A. (2010). Acute mucosal pathogenesis of feline immunodeficiency virus is independent of viral dose in vaginally infected cats. Retrovirology. 7(2).

Isaacson, J. A., Roth, J. A., Wood, C., and Carpenter, S. (1995). Loss of Gag-specific antibody reactivity in cattle experimentally infected with bovine immunodeficiency-like virus. Viral Immunol. 8(1), 27-36.

Issel, C. J., Adams, W. V., Jr., Meek, L., and Ochoa, R. (1982). Transmission of equine infectious anemia virus from horses without clinical signs of disease. J. Am. Vet. Med. Assoc. 180(3), 272-5.

Issel, C. J., Rushlow, K., Foil, L. D., and Montelaro, R. C. (1988). A perspective on equine infectious anemia with an emphasis on vector transmission and genetic analysis. Vet. Microbiol. 17(3), 251-86.

Ito, Y., Grivel, J. C., and Margolis, L. (2003). Real-time PCR assay of individual human immunodeficiency virus type 1 variants in coinfected human lymphoid tissues. J Clin Microbiol. 41(5), 2126-31.

Jacobs, R. M., Smith, H. E., Whetstone, C. A., Suarez, D. L., Jefferson, B., and Valli, V. E. (1994). Haematological and lymphocyte subset analyses in sheep inoculated with bovine immunodeficiency-like virus. Vet Res Commun. 18(6), 471-82.

Jolly, P. E., and Narayan, O. (1989). Evidence for interference, coinfections, and intertypic virus enhancement of infection by ovine-caprine lentiviruses. J Virol. 63(11), 4682-8.

Jordan, H. L., Howard, J. G., Bucci, J. G., Butterworth, J. L., English, R., Kennedy-Stoskopf, S., Tompkins, M. B., and Tompkins, W. A. (1998). Horizontal transmission of feline immunodeficiency virus with semen from seropositive cats. J. Reprod. Immunol. 41(1-2), 341-57.

Jurriaans, S., Kozaczynska, K., Zorgdrager, F., Steingrover, R., Prins, J. M., van der Kuyl, A. C., and Cornelissen, M. (2008). A sudden rise in viral load is infrequently associated with HIV-1 superinfection. J Acquir Immune Defic Syndr. 47(1), 69-73.

Kashiwase, H., Ishimura, M., Ishikawa, Y., and Nishigaki, T. (1997). Characterization of one monoclonal antibody against feline immunodeficiency virus p24 and its application to antigen capture ELISA. J Virol Methods. 68(2), 183-92.

Keele, B.F., Holland-Jones, J., Terio, K.A., Estes, J.D., Rudicell, R.S., Wilson, M.L., Li, Y., Learn, G.H., Beasley, M.T., Schumacher-Stankey, J., Wroblewski, E., Mosser, A., Raphael, J., Kamenya, S., Lonsdorf, E.V., Travis, D.A., Mlengeya, T., Kinsel, M.J., Else, J.G., Silvestri, G., Goodall, J., Sharp, P.M., Shaw, G.M., Pusey, A.E., and Hahn, B.H. (2009). Increased mortality and

Page 157: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

147

AIDS-like immunopathlogy in wild chimpanzees infected with SIVcpz. Nature. 460(7254), 515-519.

Kemen, M. J., McClain, D. S., and Matthysse, J. G. (1978). Role of horse flies in transmission of equine infectious anemia from carrier ponies. J. Am. Vet. Med. Assoc. 172(3), 360-2.

Kempster, S., Collins, M. E., and Brownlie, J. (2002). Tat protein expression in MDBK cells does not confer susceptibility to bovine immunodeficiency virus. Arch. Virol. 147(3), 643-9.

Kennedy-Stoskopf, S., and Narayan, O. (1986). Neutralizing antibodies to visna lentivirus: mechanism of action and possible role in virus persistence. J. Virol. 59(1), 37-44.

Kertayadnya, G., Soeharsono, S, Hartaninsih, N, Wilcox, GE (1997). The Physiochemical Characteristics of a Virus Associated with Jembrana Disease. In "Jembrana Disease and the Bovine Lentiviruses. Proceedings of a workshop 10-13 June 1996 Bali, Indonesia" (G. Wilcox, Soeharsono, S, Dharma, DMN, Copland, JW, Ed.), Vol. 75, pp. 43-48. Australian Centre for International Agricultural Research, Canberra.

Kertayadnya, G., Wilcox, G. E., Soeharsono, S., Hartaningsih, N., Coelen, R. J., Cook, R. D., Collins, M. E., and Brownlie, J. (1993). Characteristics of a retrovirus associated with Jembrana disease in Bali cattle. J Gen Virol. 74 ( Pt 9), 1765-78.

Khan, I.H., Mendoza, S.,Yee, J., Deane, M., Venkateswaran, K., Zhou, S.S., Barry, P.A., Lerche, N.W., and Luciw, P.A. (2006). Clin Vacc Immunol. 13(1), 45-52.

Koup, R. A., Safrit, J. T., Cao, Y., Andrews, C. A., McLeod, G., Borkowsky, W., Farthing, C., and Ho, D. D. (1994). Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J. Virol. 68(7), 4650-5.

Kusk, P., Bugge, T. H., Lindhardt, B. O., Hulgaard, E. F., and Holmback, K. (1992). Mapping of linear B-cell epitopes on the major core protein p24 of human immunodeficiency virus type 1 (HIV-1). AIDS Res Hum Retroviruses. 8(10), 1789-94.

Lamers, S. L., Sleasman, J. W., She, J. X., Barrie, K. A., Pomeroy, S. M., Barrett, D. J., and Goodenow, M. M. (1993). Independent variation and positive selection in env V1 and V2 domains within maternal-infant strains of human immunodeficiency virus type 1 in vivo. J. Virol. 67(7), 3951-60.

Langemeier, J. L., Cook, S. J., Cook, R. F., Rushlow, K. E., Montelaro, R. C., and Issel, C. J. (1996). Detection of equine infectious anemia viral RNA in plasma samples from recently infected and long-term inapparent carrier animals by PCR. J Clin Microbiol. 34(6), 1481-7.

Page 158: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

148

Larkin MA, B. G., Brown NP, Chenna R, McGettigan PA, McWilliam H, Valentin F, Wallace IM, Wilm A, Lopez R, Thompson JD, Gibson TJ and Higgins DG (2007). ClustalW and ClustalX version 2. Bioinformatics. 23(21), 2947-2948.

Leginagoikoa, I., Minguijon, E., Berriatua, E., and Juste, R. A. (2009). Improvements in the detection of small ruminant lentivirus infection in the blood of sheep by PCR. J Virol Methods. 156(1-2), 145-9.

Leroux, C., Cadore, J. L., and Montelaro, R. C. (2004). Equine Infectious Anemia Virus (EIAV): what has HIV's country cousin got to tell us? Vet. Res. 35(4), 485-512.

Leroux, C., Chastang, J., Greenland, T., and Mornex, J. F. (1997). Genomic heterogeneity of small ruminant lentiviruses: existence of heterogeneous populations in sheep and of the same lentiviral genotypes in sheep and goats. Arch. Virol. 142(6), 1125-37.

Leroux, C., Craigo, J. K., Issel, C. J., and Montelaro, R. C. (2001). Equine infectious anemia virus genomic evolution in progressor and nonprogressor ponies. J. Virol. 75(10), 4570-83.

Levy, J. (2007). Acute HIV Infection and Cells Susceptible to HIV Infection. 3rd ed. In "HIV and the Pathogenesis of AIDS". ASM Press, Washington, DC.

Levy, J. A. (2009). HIV pathogenesis: 25 years of progress and persistent challenges. Aids. 23(2), 147-60.

Lew, A. E., Bock, R. E., Miles, J., Cuttell, L. B., Steer, P., and Nadin-Davis, S. A. (2004). Sensitive and specific detection of bovine immunodeficiency virus and bovine syncytial virus by 5' Taq nuclease assays with fluorescent 3' minor groove binder-DNA probes. J Virol Methods. 116(1), 1-9.

Lewis, J. (2009). Murdoch University, Perth.

Lewis, J., McNab, T., Tenaya, M., Hartaningsih, N., Wilcox, G., and Desport, M. (2009). Comparison of immunoassay and real-time PCR methods for the detection of Jembrana disease virus infection in Bali cattle. J Virol Methods. 159(1), 81-6.

Li, F., Craigo, J. K., Howe, L., Steckbeck, J. D., Cook, S., Issel, C., and Montelaro, R. C. (2003). A live attenuated equine infectious anemia virus proviral vaccine with a modified S2 gene provides protection from detectable infection by intravenous virulent virus challenge of experimentally inoculated horses. J Virol. 77(13), 7244-53.

Li, Y., Hui, H., Burgess, C. J., Price, R. W., Sharp, P. M., Hahn, B. H., and Shaw, G. M. (1992). Complete nucleotide sequence, genome organization, and biological properties of human immunodeficiency virus type 1 in vivo: evidence for limited defectiveness and complementation. J. Virol. 66(11), 6587-600.

Page 159: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

149

Ligné, M. (1843). Mémoire et observations sur une maladie de sang, connue sous le nom d'anhémie hydrohémie, cachexie acquise du cheval. Rec. Med. Vet. Ec. Alfort 30-44.

Little, S., Sears, W., Lachtara, J., and Bienzle, D. (2009). Seroprevalence of feline leukemia virus and feline immunodeficiency virus infection among cats in Canada. Can. Vet. J. 50(6), 644-8.

Locher, C. P., Blackbourn, D. J., Barnett, S. W., Murthy, K. K., Cobb, E. K., Rouse, S., Greco, G., Reyes-Teran, G., Brasky, K. M., Carey, K. D., and Levy, J. A. (1997). Superinfection with human immunodeficiency virus type 2 can reactivate virus production in baboons but is contained by a CD8 T cell antiviral response. J Infect Dis. 176(4), 948-59.

Loomis-Price, L. D., Levi, M., Burnett, P. R., van Hamont, J. E., Shafer, R. A., Wahren, B., and Birx, D. L. (1997). Linear epitope mapping of humoral responses induced by vaccination with recombinant HIV-1 envelope protein gp160. J. Ind. Microbiol. Biotechnol. 19(1), 58-65.

Lopez, M., Soriano, V., Lozano, S., Martinez, P., Sempere, J., Gonzalez-Lahoz, J., and Benito, J. (2006). Impact of Gag sequence variability on level, phenotype, and function of anti-HIV Gag-specific CD8(+) cytotoxic T lymphocytes in untreated chronically HIV-infected patients. AIDS Res Hum Retroviruses. 22(9), 884-92.

Lu, M., Zheng, L., Mitchell, K., Kapil, S., Wood, C., and Minocha, H. (2002). Unique epitope of bovine immunodeficiency virus gag protein spans the cleavage site between p16(MA) and p2L. Clin Diagn Lab Immunol. 9(6), 1277-81.

Maki, N., Miyazawa, T., Fukasawa, M., Hasegawa, A., Hayami, M., Miki, K., and Mikami, T. (1992). Molecular characterization and heterogeneity of feline immunodeficiency virus isolates. Arch. Virol. 123(1-2), 29-45.

Makoschey, B., van Gelder, P. T., Keijsers, V., and Goovaerts, D. (2003). Bovine viral diarrhoea virus antigen in foetal calf serum batches and consequences of such contamination for vaccine production. Biologicals. 31(3), 203-8.

Maldarelli, F., Chen, M. Y., Willey, R. L., and Strebel, K. (1993). Human immunodeficiency virus type 1 Vpu protein is an oligomeric type I integral membrane protein. J. Virol. 67(8), 5056-61.

Malim, M. H., Hauber, J., Le, S. Y., Maizel, J. V., and Cullen, B. R. (1989). The HIV-1 rev trans-activator acts through a structured target sequence to activate nuclear export of unspliced viral mRNA. Nature. 338(6212), 254-7.

Mammano, F., Ohagen, A., Hoglund, S., and Gottlinger, H. G. (1994). Role of the major homology region of human immunodeficiency virus type 1 in virion morphogenesis. J. Virol. 68(8), 4927-36.

Maury, W., Thompson, R. J., Jones, Q., Bradley, S., Denke, T., Baccam, P., Smazik, M., and Oaks, J. L. (2005). Evolution of the equine infectious anemia virus

Page 160: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

150

long terminal repeat during the alteration of cell tropism. J. Virol. 79(9), 5653-64.

McGuire, T. C., Adams, D. S., Johnson, G. C., Klevjer-Anderson, P., Barbee, D. D., and Gorham, J. R. (1986). Acute arthritis in caprine arthritis-encephalitis virus challenge exposure of vaccinated or persistently infected goats. Am J Vet Res. 47(3), 537-40.

McNab, W. B., Jacobs, R. M., and Smith, H. E. (1994). A serological survey for bovine immunodeficiency-like virus in Ontario dairy cattle and associations between test results, production records and management practices. Can J Vet Res. 58(1), 36-41.

McNeilly, T. N., Baker, A., Brown, J. K., Collie, D., Maclachlan, G., Rhind, S. M., and Harkiss, G. D. (2008). Role of alveolar macrophages in respiratory transmission of visna/maedi virus. J. Virol. 82(3), 1526-36.

Mealey, R. H., Leib, S. R., Pownder, S. L., and McGuire, T. C. (2004). Adaptive immunity is the primary force driving selection of equine infectious anemia virus envelope SU variants during acute infection. J. Virol. 78(17), 9295-305.

Meas, S., Kabeya, H., Yoshihara, S., Ohashi, K., Matsuki, S., Mikami, Y., Sugimoto, C., and Onuma, M. (1998). Seroprevalence and field isolation of bovine immunodeficiency virus. J Vet Med Sci. 60(11), 1195-202.

Meas, S., Ohashi, K., Tum, S., Chhin, M., Te, K., Miura, K., Sugimoto, C., and Onuma, M. (2000a). Seroprevalence of bovine immunodeficiency virus and bovine leukemia virus in draught animals in Cambodia. J Vet Med Sci. 62(7), 779-81.

Meas, S., Seto, J., Sugimoto, C., Bakhsh, M., Riaz, M., Sato, T., Naeem, K., Ohashi, K., and Onuma, M. (2000b). Infection of bovine immunodeficiency virus and bovine leukemia virus in water buffalo and cattle populations in Pakistan. J Vet Med Sci. 62(3), 329-31.

Middeldorp, J. M., and Meloen, R. H. (1988). Epitope-mapping on the Epstein-Barr virus major capsid protein using systematic synthesis of overlapping oligopeptides. J Virol Methods. 21(1-4), 147-59.

Miller, R. J., Cairns, J. S., Bridges, S., and Sarver, N. (2000). Human immunodeficiency virus and AIDS: insights from animal lentiviruses. J. Virol. 74(16), 7187-95.

Miyake, A., Ibuki, K., Enose, Y., Suzuki, H., Horiuchi, R., Motohara, M., Saito, N., Nakasone, T., Honda, M., Watanabe, T., Miura, T., and Hayami, M. (2006). Rapid dissemination of a pathogenic simian/human immunodeficiency virus to systemic organs and active replication in lymphoid tissues following intrarectal infection. J Gen Virol. 87(Pt 5), 1311-20.

Miyazawa, T., Kawaguchi, Y., Kohmoto, M., Tomonaga, K., and Mikami, T. (1994). Comparative functional analysis of the various lentivirus long terminal

Page 161: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

151

repeats in human colon carcinoma cell line (SW480 cells) and feline renal cell line (CRFK cells). J Vet Med Sci. 56(5), 895-9.

Mohan, M., Malayer, J. R., Geisert, R. D., and Morgan, G. L. (2001). Expression of retinol-binding protein messenger RNA and retinoic acid receptors in preattachment bovine embryos. Mol Reprod Dev. 60(3), 289-96.

Montelaro, R. C., Grund, C., Raabe, M., Woodson, B., Cook, R. F., Cook, S., and Issel, C. J. (1996). Characterization of protective and enhancing immune responses to equine infectious anemia virus resulting from experimental vaccines. AIDS Res Hum Retroviruses. 12(5), 413-5.

Montelaro, R. C., Parekh, B., Orrego, A., and Issel, C. J. (1984). Antigenic variation during persistent infection by equine infectious anemia virus, a retrovirus. J Biol Chem. 259(16), 10539-44.

Mordasini, F., Vogt, H. R., Zahno, M. L., Maeschli, A., Nenci, C., Zanoni, R., Peterhans, E., and Bertoni, G. (2006). Analysis of the antibody response to an immunodominant epitope of the envelope glycoprotein of a lentivirus and its diagnostic potential. J Clin Microbiol. 44(3), 981-91.

Morin, T., Guiguen, F., Bouzar, B. A., Villet, S., Greenland, T., Grezel, D., Gounel, F., Gallay, K., Garnier, C., Durand, J., Alogninouwa, T., Mselli-Lakhal, L., Mornex, J. F., and Chebloune, Y. (2003). Clearance of a productive lentivirus infection in calves experimentally inoculated with caprine arthritis-encephalitis virus. J. Virol. 77(11), 6430-7.

Mselli-Lakhal, L., Guiguen, F., Greenland, T., Mornex, J. F., and Chebloune, Y. (2007). In vitro cross-species infections using a caprine arthritis encephalitis lentivirus carrying the GFP marker gene. J Virol Methods. 143(1), 11-5.

Munro, R., Lysons, R., Venables, C., Horigan, M., Jeffrey, M., and Dawson, M. (1998). Lymphadenopathy and non-suppurative meningo-encephalitis in calves experimentally infected with bovine immunodeficiency-like virus (FL112). J Comp Pathol. 119(2), 121-34.

Muraguri, G. R., Kiara, H. K., and McHardy, N. (1999). Treatment of East Coast fever: a comparison of parvaquone and buparvaquone. Vet. Parasitol. 87(1), 25-37.

Narayan, O., Clements, J. E., Strandberg, J. D., Cork, L. C., and Griffin, D. E. (1980). Biological characterization of the virus causing leukoencephalitis and arthritis in goats. J Gen Virol. 50(1), 69-79.

Narayan, O., Griffin, D. E., and Clements, J. E. (1978). Virus mutation during 'slow infection': temporal development and characterization of mutants of visna virus recovered from sheep. J Gen Virol. 41(2), 343-52.

Narayan, O., Kennedy-Stoskopf, S., Sheffer, D., Griffin, D. E., and Clements, J. E. (1983). Activation of caprine arthritis-encephalitis virus expression during maturation of monocytes to macrophages. Infect Immun. 41(1), 67-73.

Page 162: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

152

Nehete, P. N., Nehete, B. P., Hill, L., Manuri, P. R., Baladandayuthapani, V., Feng, L., Simmons, J., and Sastry, K. J. (2008). Selective induction of cell-mediated immunity and protection of rhesus macaques from chronic SHIV(KU2) infection by prophylactic vaccination with a conserved HIV-1 envelope peptide-cocktail. Virology. 370(1), 130-41.

Nenci, C., Zhano, M.L., Vogt, H.R., Obexer-Ruff, G., Doherr, M.G., Zanoni, R., Peterhans, E., and Bertoni, G. (2007). Vaccination with a T-cell-priming Gag peptide of caprine arthritis encephalitis virus enhances virus replication transiently in vivo. J. Gen. Virol. 88, 1589-93.

Neurath, A. R., Strick, N., and Lee, E. S. (1990). B cell epitope mapping of human immunodeficiency virus envelope glycoproteins with long (19- to 36-residue) synthetic peptides. J Gen Virol. 71 ( Pt 1), 85-95.

Nilsen, B. M., Haugan, I. R., Berg, K., Olsen, L., Brown, P. O., and Helland, D. E. (1996). Monoclonal antibodies against human immunodeficiency virus type 1 integrase: epitope mapping and differential effects on integrase activities in vitro. J. Virol. 70(3), 1580-7.

Nilsson, C., Makitalo, B., Thorstensson, R., Norley, S., Binninger-Schinzel, D., Cranage, M., Rud, E., Biberfeld, G., and Putkonen, P. (1998). Live attenuated simian immunodeficiency virus (SIV)mac in macaques can induce protection against mucosal infection with SIVsm. Aids. 12(17), 2261-70.

O'Rourke, K., Perryman, L. E., and McGuire, T. C. (1988). Antiviral, anti-glycoprotein and neutralizing antibodies in foals with equine infectious anaemia virus. J Gen Virol. 69 ( Pt 3), 667-74.

Oaks, J. L., McGuire, T. C., Ulibarri, C., and Crawford, T. B. (1998). Equine infectious anemia virus is found in tissue macrophages during subclinical infection. J. Virol. 72(9), 7263-9.

Olmsted, R. A., Hirsch, V. M., Purcell, R. H., and Johnson, P. R. (1989). Nucleotide sequence analysis of feline immunodeficiency virus: genome organization and relationship to other lentiviruses. Proc Natl Acad Sci U S A. 86(20), 8088-92.

Olmsted, R. A., Langley, R., Roelke, M. E., Goeken, R. M., Adger-Johnson, D., Goff, J. P., Albert, J. P., Packer, C., Laurenson, M. K., Caro, T. M., and et al. (1992). Worldwide prevalence of lentivirus infection in wild feline species: epidemiologic and phylogenetic aspects. J. Virol. 66(10), 6008-18.

Orrego, A., Issel, C. J., Montelaro, R. C., and Adams, W. V., Jr. (1982). Virulence and in vitro growth of a cell-adapted strain of equine infectious anemia virus after serial passage in ponies. Am J Vet Res. 43(9), 1556-60.

Palker, T. J., Matthews, T. J., Clark, M. E., Cianciolo, G. J., Randall, R. R., Langlois, A. J., White, G. C., Safai, B., Snyderman, R., Bolognesi, D. P., and et al. (1987). A conserved region at the COOH terminus of human immunodeficiency virus gp120 envelope protein contains an immunodominant epitope. Proc Natl Acad Sci U S A. 84(8), 2479-83.

Page 163: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

153

Pancino, G., Chappey, C., Saurin, W., and Sonigo, P. (1993). B epitopes and selection pressures in feline immunodeficiency virus envelope glycoproteins. J. Virol. 67(2), 664-72.

Pantaleo, G., Menzo, S., Vaccarezza, M., Graziosi, C., Cohen, O. J., Demarest, J. F., Montefiori, D., Orenstein, J. M., Fox, C., Schrager, L. K., and et al. (1995). Studies in subjects with long-term nonprogressive human immunodeficiency virus infection. N. Engl. J. Med. 332(4), 209-16.

Pasick, J. (1998). Maedi-visna virus and caprine arthritis-encephalitis virus: distinct species or quasispecies and its implications for laboratory diagnosis. Can J Vet Res. 62(4), 241-4.

Payne, S., Parekh, B., Montelaro, R. C., and Issel, C. J. (1984). Genomic alterations associated with persistent infections by equine infectious anaemia virus, a retrovirus. J Gen Virol. 65 ( Pt 8), 1395-9.

Payne, S. L., Qi, X. M., Shao, H., Dwyer, A., and Fuller, F. J. (1998). Disease induction by virus derived from molecular clones of equine infectious anemia virus. J. Virol. 72(1), 483-7.

Pedersen, N. C., Ho, E. W., Brown, M. L., and Yamamoto, J. K. (1987). Isolation of a T-lymphotropic virus from domestic cats with an immunodeficiency-like syndrome. Science. 235(4790), 790-3.

Pederson, N.C., Leutenegger, C.M., Woo, J., and Higgins, J. (2001). Virulence differences between two field isolates of feline immunodeficiency virus (FIV-APetaluma and FIV-CPGammar) in young adult specific pathogen free cats. Vet. Immunol. Immunol. 79, 53-67.

Pepin, M., Vitu, C., Russo, P., Mornex, J. F., and Peterhans, E. (1998). Maedi-visna virus infection in sheep: a review. Vet. Res. 29(3-4), 341-67.

Peterlin, M. (1995). Molecular Biology of HIV. In "The Retroviridae " (J. Levy, Ed.), Vol. 4, pp. 185-238. 4 vols. Plenum Press, New York.

Petry, H., Dittmer, U., Stahl-Hennig, C., Coulibaly, C., Makoschey, B., Fuchs, D., Wachter, H., Tolle, T., Morys-Wortmann, C., Kaup, F. J., and et al. (1995). Reactivation of human immunodeficiency virus type 2 in macaques after simian immunodeficiency virus SIVmac superinfection. J. Virol. 69(3), 1564-74.

Petursson, G., Nathanson, N., Georgsson, G., Panitch, H., and Palsson, P. A. (1976). Pathogenesis of visna. I. Sequential virologic, serologic, and pathologic studies. Lab Invest. 35(4), 402-12.

Piantadosi, A., Chohan, B., Chohan, V., McClelland, R. S., and Overbaugh, J. (2007). Chronic HIV-1 infection frequently fails to protect against superinfection. PLoS Pathog. 3(11), e177.

Page 164: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

154

Pifat, D. Y., Ennis, W. H., Ward, J. M., Oberste, M. S., and Gonda, M. A. (1992). Persistent infection of rabbits with bovine immunodeficiency-like virus. J. Virol. 66(7), 4518-24.

Pisoni, G., Bertoni, G., Puricelli, M., Maccalli, M., and Moroni, P. (2007). Demonstration of Co-Infection with and Recombination of Caprine Arthritis-Encephalitis Virus and Maedi-Visna Virus in Naturally Infected Goats. J. Virol.

Pisoni, G., Quasso, A., and Moroni, P. (2005). Phylogenetic analysis of small-ruminant lentivirus subtype B1 in mixed flocks: evidence for natural transmission from goats to sheep. Virology. 339(2), 147-52.

Pistello, M., Matteucci, D., Cammarota, G., Mazzetti, P., Giannecchini, S., Del Mauro, D., Macchi, S., Zaccaro, L., and Bendinelli, M. (1999). Kinetics of replication of a partially attenuated virus and of the challenge virus during a three-year intersubtype feline immunodeficiency virus superinfection experiment in cats. J. Virol. 73(2), 1518-27.

Poss, M., Idoine, A., Ross, H. A., Terwee, J. A., Vandewoude, S., and Rodrigo, A. (2006). Recombination in feline lentiviral genomes during experimental cross-species infection. Virology.

Poss, M., and Ross, H. (2008). Evolution of the long terminal repeat and accessory genes of feline immunodeficiency virus genomes from naturally infected cougars. Virology. 370(1), 55-62.

Pozzetto, B., Le Bihan, J. C., and Gaudin, O. G. (1986). Rapid diagnosis of echovirus 33 infection by neutralizing specific IgM antibody. J Med Virol. 18(4), 361-7.

Preziuso, S., Renzoni, G., Allen, T. E., Taccini, E., Rossi, G., DeMartini, J. C., and Braca, G. (2004). Colostral transmission of maedi visna virus: sites of viral entry in lambs born from experimentally infected ewes. Vet. Microbiol. 104(3-4), 157-64.

Pyper, J. M., Clements, J. E., Gonda, M. A., and Narayan, O. (1986). Sequence homology between cloned caprine arthritis encephalitis virus and visna virus, two neurotropic lentiviruses. J. Virol. 58(2), 665-70.

Querat, G., Barban, V., Sauze, N., Filippi, P., Vigne, R., Russo, P., and Vitu, C. (1984). Highly lytic and persistent lentiviruses naturally present in sheep with progressive pneumonia are genetically distinct. J. Virol. 52(2), 672-9.

Ramachandran, S. (1996). Early Observations and Research on Jembrana Disease in Bali and Other Indonesian Islands. In "Jembrana Disease and the Bovine Lentiviruses" (G. Wilcox, Soeharsono, S, Dharma, DMN, Copland, JW, Ed.), pp. 5-9. Australian Centre for International Agricultural Research, Canberra.

Ravazzolo, A. P., Nenci, C., Vogt, H. R., Waldvogel, A., Obexer-Ruff, G., Peterhans, E., and Bertoni, G. (2006). Viral load, organ distribution, histopathological lesions, and cytokine mRNA expression in goats infected

Page 165: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

155

with a molecular clone of the caprine arthritis encephalitis virus. Virology. 350(1), 116-27.

Reis, J. K., Craigo, J. K., Cook, S. J., Issel, C. J., and Montelaro, R. C. (2003). Characterization of EIAV LTR variability and compartmentalization in various reservoir tissues of long-term inapparent carrier ponies. Virology. 311(1), 169-80.

Richardson, J., Moraillon, A., Baud, S., Cuisinier, A.M., Sonigo, P., and Pancino, G. (1997). Enhancement of Feline Immunodeficiency Virus (FIV) infection after DNA Vaccination with the FIV envelope. J. Virol. 71(12), 9640-49.

Rimstad, E., East, N. E., Torten, M., Higgins, J., DeRock, E., and Pedersen, N. C. (1993). Delayed seroconversion following naturally acquired caprine arthritis-encephalitis virus infection in goats. Am J Vet Res. 54(11), 1858-62.

Romani, B., Engelbrecht, S., and Glashoff, R.H. (2009). Antiviral roles of APOBEC proteins against HIV-1 and suppression by Vif. Arch. Virol. 154, 1579-88.

Rosati, S., Mannelli, A., Merlo, T., and Ponti, N. (1999). Characterization of the immunodominant cross-reacting epitope of visna maedi virus and caprine arthritis-encephalitis virus capsid antigen. Virus Res. 61(2), 177-83.

Rosati, S., Profiti, M., Lorenzetti, R., Bandecchi, P., Mannelli, A., Ortoffi, M., Tolari, F., and Ciabatti, I. M. (2004). Development of recombinant capsid antigen/transmembrane epitope fusion proteins for serological diagnosis of animal lentivirus infections. J Virol Methods. 121(1), 73-8.

Ryan, G., Klein, D., Knapp, E., Hosie, M. J., Grimes, T., Mabruk, M. J., Jarrett, O., and Callanan, J. J. (2003). Dynamics of viral and proviral loads of feline immunodeficiency virus within the feline central nervous system during the acute phase following intravenous infection. J. Virol. 77(13), 7477-85.

Sahu, S. P., Alstad, A. D., Pedersen, D. D., and Pearson, J. E. (1994). Diagnosis of eastern equine encephalomyelitis virus infection in horses by immunoglobulin M and G capture enzyme-linked immunosorbent assay. J Vet Diagn Invest. 6(1), 34-8.

Salinovich, O., Payne, S. L., Montelaro, R. C., Hussain, K. A., Issel, C. J., and Schnorr, K. L. (1986). Rapid emergence of novel antigenic and genetic variants of equine infectious anemia virus during persistent infection. J. Virol. 57(1), 71-80.

Sattentau, Q. J., Clapham, P. R., Weiss, R. A., Beverley, P. C., Montagnier, L., Alhalabi, M. F., Gluckmann, J. C., and Klatzmann, D. (1988). The human and simian immunodeficiency viruses HIV-1, HIV-2 and SIV interact with similar epitopes on their cellular receptor, the CD4 molecule. Aids. 2(2), 101-5.

Sauter, S. L., and Gasmi, M. (2001). FIV vector systems. Somat Cell Mol Genet. 26(1-6), 99-129.

Page 166: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

156

Schmitz, J. E., Kuroda, M. J., Santra, S., Sasseville, V. G., Simon, M. A., Lifton, M. A., Racz, P., Tenner-Racz, K., Dalesandro, M., Scallon, B. J., Ghrayeb, J., Forman, M. A., Montefiori, D. C., Rieber, E. P., Letvin, N. L., and Reimann, K. A. (1999). Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science. 283(5403), 857-60.

Scobie, L., Venables, C., Hughes, K., Dawson, M., and Jarrett, O. (1999). The antibody response of cattle infected with bovine immunodeficiency virus to peptides of the viral transmembrane protein. J Gen Virol. 80 ( Pt 1), 237-43.

Sealy, R., Zhan, X., Lockey, T.D., Martin, L., Blanchard, J., Traina-Dorge, V., and Hurwitz, J.L. (2009). SHIV infection protects against heterologous pathogenic SHIV challenge in macaques: A gold-standard for HIV-1 vaccine development? Curr. HIV Res. 7(5), 497-03.

Sellon, D. C., Perry, S. T., Coggins, L., and Fuller, F. J. (1992). Wild-type equine infectious anemia virus replicates in vivo predominantly in tissue macrophages, not in peripheral blood monocytes. J. Virol. 66(10), 5906-13.

Shah, C., Huder, J. B., Boni, J., Schonmann, M., Muhlherr, J., Lutz, H., and Schupbach, J. (2004). Direct evidence for natural transmission of small-ruminant lentiviruses of subtype A4 from goats to sheep and vice versa. J. Virol. 78(14), 7518-22.

Sharma, D. P., Zink, M. C., Anderson, M., Adams, R., Clements, J. E., Joag, S. V., and Narayan, O. (1992). Derivation of neurotropic simian immunodeficiency virus from exclusively lymphocytetropic parental virus: pathogenesis of infection in macaques. J. Virol. 66(6), 3550-6.

Sharpe, S. A., Cope, A., Dowall, S., Berry, N., Ham, C., Heeney, J. L., Hopkins, D., Easterbrook, L., Dennis, M., Almond, N., and Cranage, M. (2004). Macaques infected long-term with attenuated simian immunodeficiency virus (SIVmac) remain resistant to wild-type challenge, despite declining cytotoxic T lymphocyte responses to an immunodominant epitope. J Gen Virol. 85(Pt 9), 2591-602.

Sharpe, S. A., Whatmore, A. M., Hall, G. A., and Cranage, M. P. (1997). Macaques infected with attenuated simian immunodeficiency virus resist superinfection with virulence-revertant virus. J Gen Virol. 78 ( Pt 8), 1923-7.

Shimojima, M., Miyazawa, T., Ikeda, Y., McMonagle, E. L., Haining, H., Akashi, H., Takeuchi, Y., Hosie, M. J., and Willett, B. J. (2004). Use of CD134 as a primary receptor by the feline immunodeficiency virus. Science. 303(5661), 1192-5.

Sigurdsson, B., Grimsson, H., and Palsson, P. A. (1952). Maedi, a chronic, progressive infection of sheep's lungs. J Infect Dis. 90(3), 233-41.

Sigurdsson, B., Palsson, P., and Grimsson, H. (1957). Visna, a demyelinating transmissible disease of sheep. J. Neuropathol. Exp. Neurol. 16(3), 389-403.

Page 167: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

157

Simmonds, P., Balfe, P., Ludlam, C. A., Bishop, J. O., and Brown, A. J. (1990). Analysis of sequence diversity in hypervariable regions of the external glycoprotein of human immunodeficiency virus type 1. J. Virol. 64(12), 5840-50.

Singh, I., McConnell, I., and Blacklaws, B. (2006). Immune response to individual maedi-visna virus gag antigens. J. Virol. 80(2), 912-9.

Smith, D. M., Richman, D. D., and Little, S. J. (2005). HIV superinfection. J Infect Dis. 192(3), 438-44.

Smith, D. M., Strain, M. C., Frost, S. D., Pillai, S. K., Wong, J. K., Wrin, T., Liu, Y., Petropolous, C. J., Daar, E. S., Little, S. J., and Richman, D. D. (2006). Lack of neutralizing antibody response to HIV-1 predisposes to superinfection. Virology. 355(1), 1-5.

Snider, T. G., 3rd, Coats, K. S., Storts, R. W., Graves, K. F., Cooper, C. R., Hoyt, P. G., Luther, D. G., and Jenny, B. F. (2003a). Natural bovine lentivirus type 1 infection in Holstein dairy cattle. II. Lymphoid tissue lesions. Comp Immunol Microbiol Infect Dis. 26(1), 1-15.

Snider, T. G., 3rd, Hoyt, P. G., Coats, K. S., Graves, K. F., Cooper, C. R., Storts, R. W., Luther, D. G., and Jenny, B. F. (2003b). Natural bovine lentiviral type 1 infection in Holstein dairy cattle. I. Clinical, serological, and pathological observations. Comp Immunol Microbiol Infect Dis. 26(2), 89-101.

Soeharsono, S., Budiantono, A, Sulistyana, K, Tenaya, M, Hartaningsih, N, Dharma, DMN, Soesanto, S, Wilcox, GE (1997a). Clinical Changes in Bali Cattle and Other Ruminants Following Infection with Jembrana Disease Virus. In "Jembrana Disease and the Bovine Lentiviruses. Proceedings of a workshop 10-13 June 1996 Bali, Indonesia." (G. Wilcox, Soeharsono, S, Dharma, DMN, Copland, JW, Ed.), Vol. 75, pp. 10-25. Australian Centre for International Agricultural Research, Canberra.

Soeharsono, S., Hartaningsih, N., Soetrisno, M., Kertayadnya, G., and Wilcox, G. E. (1990). Studies of experimental Jembrana disease in Bali cattle. I. Transmission and persistence of the infectious agent in ruminants and pigs, and resistance of recovered cattle to re-infection. J Comp Pathol. 103(1), 49-59.

Soeharsono, S., Teken Temadja, IGN (1997b). The Occurrence and History of Jembrana Disease in Indonesia. 75 ed. In "Jembrana Disease and the Bovine Lentiviruses. Proceedings of a workshop 10-13 June 1996 Bali, Indonesia" (G. Wilcox, Soeharsono, S, Dharma, DMN, Copland, JW, Ed.), pp. 2-4. Australian Centre for International Agricultural Research, Canberra.

Soeharsono, S., Wilcox, G. E., Dharma, D. M., Hartaningsih, N., Kertayadnya, G., and Budiantono, A. (1995a). Species differences in the reaction of cattle to Jembrana disease virus infection. J Comp Pathol. 112(4), 391-402.

Page 168: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

158

Soeharsono, S., Wilcox, G. E., Putra, A. A., Hartaningsih, N., Sulistyana, K., and Tenaya, M. (1995b). The transmission of Jembrana disease, a lentivirus disease of Bos javanicus cattle. Epidemiol Infect. 115(2), 367-74.

Soesanto, M., Soeharsono, S., Budiantono, A., Sulistyana, K., Tenaya, M., and Wilcox, G. E. (1990). Studies on experimental Jembrana disease in Bali cattle. II. Clinical signs and haematological changes. J Comp Pathol. 103(1), 61-71.

Soutullo, A., Santi, M. N., Perin, J. C., Beltramini, L. M., Borel, I. M., Frank, R., and Tonarelli, G. G. (2007). Systematic epitope analysis of the p26 EIAV core protein. J. Mol. Recognit. 20(4), 227-37.

Spyrou, V., Papanastassopoulou, M., Psychas, V., Billinis, C., Koumbati, M., Vlemmas, J., and Koptopoulos, G. (2003). Equine infectious anemia in mules: virus isolation and pathogenicity studies. Vet. Microbiol. 95(1-2), 49-59.

Stebbings, R., Berry, N., Stott, J., Hull, R., Walker, B., Lines, J., Elsley, W., Brown, S., Wade-Evans, A., Davis, G., Cowie, J., Sethi, M., and Almond, N. (2004). Vaccination with live attenuated simian immunodeficiency virus for 21 days protects against superinfection. Virology. 330(1), 249-60.

Stebbings, R., Berry, N., Waldmann, H., Bird, P., Hale, G., Stott, J., North, D., Hull, R., Hall, J., Lines, J., Brown, S., D'Arcy, N., Davis, L., Elsley, W., Edwards, C., Ferguson, D., Allen, J., and Almond, N. (2005). CD8+ lymphocytes do not mediate protection against acute superinfection 20 days after vaccination with a live attenuated simian immunodeficiency virus. J. Virol. 79(19), 12264-72.

Stebbings, R. J., Almond, N. M., Stott, E. J., Berry, N., Wade-Evans, A. M., Hull, R., Lines, J., Silvera, P., Sangster, R., Corcoran, T., Rose, J., and Walker, K. B. (2002). Mechanisms of protection induced by attenuated simian immunodeficiency virus. Virology. 296(2), 338-53.

Stewart, M., Desport, M., Hartaningsih, N., and Wilcox, G. (2005). TaqMan real-time reverse transcription-PCR and JDVp26 antigen capture enzyme-linked immunosorbent assay to quantify Jembrana disease virus load during the acute phase of in vivo infection. J Clin Microbiol. 43(11), 5574-80.

Stivahtis, G. L., Soares, M. A., Vodicka, M. A., Hahn, B. H., and Emerman, M. (1997). Conservation and host specificity of Vpr-mediated cell cycle arrest suggest a fundamental role in primate lentivirus evolution and biology. J. Virol. 71(6), 4331-8.

Streetdirectory.com (2009). Bali overview. Streetdirectory.co.id, Singapore.

Strebel, K., Luban, J., and Jeang, K.T. (2009). Human cellular restriction factors that target HIV-1 replication. BMC Medicine. 7(48).

Page 169: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

159

Suarez, D. L., VanDerMaaten, M. J., Wood, C., and Whetstone, C. A. (1993). Isolation and characterization of new wild-type isolates of bovine lentivirus. J. Virol. 67(8), 5051-5.

Suarez, D. L., and Whetstone, C. A. (1995). Identification of hypervariable and conserved regions in the surface envelope gene in the bovine lentivirus. Virology. 212(2), 728-33.

Tao, B., and Fultz, P.N. (1995). Molecular and Biological Analyses of Quasispecies during Evolution of a Virulent Simian Immunodeficiency Virus, SIVsmmPBj14. J. Virol.69(4), 2031-7.

Temin, H. M., and Mizutani, S. (1970). RNA-dependent DNA polymerase in virions of Rous sarcoma virus. Nature. 226(5252), 1211-3.

Terwee, J. A., Carlson, J. K., Sprague, W. S., Sondgeroth, K. S., Shropshire, S. B., Troyer, J. L., and Vandewoude, S. (2008). Prevention of immunodeficiency virus induced CD4+ T-cell depletion by prior infection with a non-pathogenic virus. Virology. 377(1), 63-70.

Terwee, J. A., Yactor, J. K., Sondgeroth, K. S., and Vandewoude, S. (2005). Puma lentivirus is controlled in domestic cats after mucosal exposure in the absence of conventional indicators of immunity. J. Virol. 79(5), 2797-806.

Tobin, G. J., Sowder, R. C., 2nd, Fabris, D., Hu, M. Y., Battles, J. K., Fenselau, C., Henderson, L. E., and Gonda, M. A. (1994). Amino acid sequence analysis of the proteolytic cleavage products of the bovine immunodeficiency virus Gag precursor polypeptide. J. Virol. 68(11), 7620-7.

Trautwein, G. (1992). Immune mechanisms in the pathogenesis of viral diseases: a review. Vet Microbiol. 33(1-4), 19-34.

UNAIDS (2008). Report on the global AIDS epidemic. UNAIDS. August 2008.

Valas, S., Benoit, C., Baudry, C., Perrin, G., and Mamoun, R. Z. (2000). Variability and immunogenicity of caprine arthritis-encephalitis virus surface glycoprotein. J. Virol. 74(13), 6178-85.

Vallée, H., Carré, H (1904). Sur la nature infectieuse de l'anémie du cheval. C. R. Acad. Sci. 139, 331-333.

Van der Maaten, M. J., Boothe, A. D., and Seger, C. L. (1972). Isolation of a virus from cattle with persistent lymphocytosis. J Natl Cancer Inst. 49(6), 1649-57.

Van Regenmortel, M. H. V. (1999a). Molecular Dissection of Protein Antigens and the Prediction of Epitopes First ed. In "Synthetic Peptides as Antigens". Elsevier Science, Amsterdam.

Van Regenmortel, M. H. V. (1999b). The use of peptides for diagnosing viral infections. In "Synthetic Peptides as Antigens" (M. H. V. Van Regenmortel, Muller, S, Ed.). Elsevier Science, Amsterdam.

Page 170: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

160

VandeWoude, S., and Apetrei, C. (2006). Going wild: lessons from naturally occurring T-lymphotropic lentiviruses. Clin Microbiol Rev. 19(4), 728-62.

VandeWoude, S., Hageman, C. A., O'Brien, S. J., and Hoover, E. A. (2002). Nonpathogenic lion and puma lentiviruses impart resistance to superinfection by virulent feline immunodeficiency virus. J Acquir Immune Defic Syndr. 29(1), 1-10.

VandeWoude, S., Hageman, C. L., and Hoover, E. A. (2003). Domestic cats infected with lion or puma lentivirus develop anti-feline immunodeficiency virus immune responses. J Acquir Immune Defic Syndr. 34(1), 20-31.

VandeWoude, S., O'Brien, S. J., and Hoover, E. A. (1997). Infectivity of lion and puma lentiviruses for domestic cats. J Gen Virol. 78 ( Pt 4), 795-800.

Villet, S., Bouzar, B. A., Morin, T., Verdier, G., Legras, C., and Chebloune, Y. (2003). Maedi-visna virus and caprine arthritis encephalitis virus genomes encode a Vpr-like but no Tat protein. J Virol. 77(17), 9632-8.

Wagner, E., Hewlett MJ (1999). "Basic Virology." Blackwell Science Inc.

Wagter, L. H., Jansen, A., Bleumink-Pluym, N. M., Lenstra, J. A., and Houwers, D. J. (1998). PCR detection of lentiviral GAG segment DNA in the white blood cells of sheep and goats. Vet Res Commun. 22(5), 355-62.

Wareing, S., Hartaningsih, N., Wilcox, G. E., and Penhale, W. J. (1999). Evidence for immunosuppression associated with Jembrana disease virus infection of cattle. Vet. Microbiol. 68(1-2), 179-85.

Wasmoen, T., Armiger-Luhman, S., Egan, C., Hall, V., Chu, H. J., Chavez, L., and Acree, W. (1992). Transmission of feline immunodeficiency virus from infected queens to kittens. Vet. Immunol. Immunopathol. 35(1-2), 83-93.

Westwood, O., M.R., Hay, Frank C., Ed. (2000). Epitope Mapping A Practical Approach. Oxford: Oxford University Press.

Whetstone, C. A., Suarez, D. L., Miller, J. M., Pesch, B. A., and Harp, J. A. (1997). Bovine lentivirus induces early transient B-cell proliferation in experimentally inoculated cattle and appears to be pantropic. J. Virol. 71(1), 640-4.

Whetstone, C. A., VanDerMaaten, M. J., and Black, J. W. (1990). Humoral immune response to the bovine immunodeficiency-like virus in experimentally and naturally infected cattle. J. Virol. 64(7), 3557-61.

Whetstone, C. A., VanDerMaaten, M. J., and Miller, J. M. (1991). A western blot assay for the detection of antibodies to bovine immunodeficiency-like virus in experimentally inoculated cattle, sheep, and goats. Arch. Virol. 116(1-4), 119-31.

Whetter, L., Archambault, D., Perry, S., Gazit, A., Coggins, L., Yaniv, A., Clabough, D., Dahlberg, J., Fuller, F., and Tronick, S. (1990). Equine infectious anemia

Page 171: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

161

virus derived from a molecular clone persistently infects horses. J. Virol. 64(12), 5750-6.

Wilcox, G. E., Chadwick, B. J., and Kertayadnya, G. (1995). Recent advances in the understanding of Jembrana disease. Vet Microbiol. 46(1-3), 249-55.

Wilcox, G. E., Kertayadnya, G., Hartaningsih, N., Dharma, D. M., Soeharsono, S., and Robertson, T. (1992). Evidence for a viral aetiology of Jembrana disease in Bali cattle. Vet. Microbiol. 33(1-4), 367-74.

Willett, B. J., Picard, L., Hosie, M. J., Turner, J. D., Adema, K., and Clapham, P. R. (1997). Shared usage of the chemokine receptor CXCR4 by the feline and human immunodeficiency viruses. J. Virol. 71(9), 6407-15.

Williams, D. L., Issel, C. J., Steelman, C. D., Adams, W. V., Jr., and Benton, C. V. (1981). Studies with equine infectious anemia virus: transmission attempts by mosquitoes and survival of virus on vector mouthparts and hypodermic needles, and in mosquito tissue culture. Am J Vet Res. 42(9), 1469-73.

Williams, K.C. and Burdo, T.H. (2009). HIV and SIV infection- the role of cellular restriction factors and immune responses in viral replication and pathogenesis. APMIS. 117(5-6), 400-412.

Williams, S., van der Logt, P., Germaschewski, V. (2000). Phage display libraries. In "Epitope Mapping" (O. Westwood, M.R., Hay, Frank C., Ed.). Oxford University Press, Amsterdam.

Wiryosuhanto, S. (1997). Bali Cattle- Their Economic Importance in Indonesia. In "Jembrana Disease and the Bovine Lentiviruses. Proceedings of a workshop 10-13 June 1996 Bali, Indonesia." (G. Wilcox, Soeharsono, S, Dharma, DMN, Copland, JW, Ed.), Vol. 75, pp. 34-42. Australian Centre for International Agricultural Research, Canberra.

Wolfs, T. F., de Jong, J. J., Van den Berg, H., Tijnagel, J. M., Krone, W. J., and Goudsmit, J. (1990). Evolution of sequences encoding the principal neutralization epitope of human immunodeficiency virus 1 is host dependent, rapid, and continuous. Proc Natl Acad Sci U S A. 87(24), 9938-42.

Wu, D., Murakami, K., Morooka, A., Jin, H., Inoshima, Y., and Sentsui, H. (2003). In vivo transcription of bovine leukemia virus and bovine immunodeficiency-like virus. Virus Res. 97(2), 81-7.

Xiao, C., Liu, Y., Jiang, Y., Magoffin, D. E., Guo, H., Xuan, H., Wang, G., Wang, L. F., and Tu, C. (2008). Monoclonal antibodies against the nucleocapsid proteins of henipaviruses: production, epitope mapping and application in immunohistochemistry. Arch. Virol. 153(2), 273-81.

Yamamoto, J. K., Hansen, H., Ho, E. W., Morishita, T. Y., Okuda, T., Sawa, T. R., Nakamura, R. M., and Pedersen, N. C. (1989). Epidemiologic and clinical aspects of feline immunodeficiency virus infection in cats from the continental United States and Canada and possible mode of transmission. J. Am. Vet. Med. Assoc. 194(2), 213-20.

Page 172: Bovine immunodeficiency virus and Jembrana disease virus … · 2013-07-09 · Society for Veterinary Virology Conference in Budapest, Hungary 2009. Poster presentations “Towards

162

Yeh, W. W., Jaru-Ampornpan, P., Nevidomskyte, D., Asmal, M., Rao, S. S., Buzby, A. P., Montefiori, D. C., Korber, B. T., and Letvin, N. L. (2009). Partial protection of Simian immunodeficiency virus (SIV)-infected rhesus monkeys against superinfection with a heterologous SIV isolate. J. Virol. 83(6), 2686-96.

Zanoni, R. G. (1998). Phylogenetic analysis of small ruminant lentiviruses. J Gen Virol. 79 ( Pt 8), 1951-61.

Zanoni, R. G., Nauta, I. M., Kuhnert, P., Pauli, U., Pohl, B., and Peterhans, E. (1992). Genomic heterogeneity of small ruminant lentiviruses detected by PCR. Vet. Microbiol. 33(1-4), 341-51.

Zhang, B., Jin, S., Jin, J., Li, F., and Montelaro, R. C. (2005). A tumor necrosis factor receptor family protein serves as a cellular receptor for the macrophage-tropic equine lentivirus. Proc Natl Acad Sci U S A. 102(28), 9918-23.

Zhang, S., Troyer, D. L., Kapil, S., Zheng, L., Kennedy, G., Weiss, M., Xue, W., Wood, C., and Minocha, H. C. (1997a). Detection of proviral DNA of bovine immunodeficiency virus in bovine tissues by polymerase chain reaction (PCR) and PCR in situ hybridization. Virology. 236(2), 249-57.

Zhang, S., Wood, C., Xue, W., Krukenberg, S. M., Chen, Q., and Minocha, H. C. (1997b). Immune suppression in calves with bovine immunodeficiency virus. Clin Diagn Lab Immunol. 4(2), 232-5.

Zhang, Z., Watt, N. J., Hopkins, J., Harkiss, G., and Woodall, C. J. (2000). Quantitative analysis of maedi-visna virus DNA load in peripheral blood monocytes and alveolar macrophages. J Virol Methods. 86(1), 13-20.

Zheng, L., Zhang, S., Wood, C., Kapil, S., Wilcox, G. E., Loughin, T. A., and Minocha, H. C. (2001). Differentiation of two bovine lentiviruses by a monoclonal antibody on the basis of epitope specificity. Clin Diagn Lab Immunol. 8(2), 283-7.

Zink, M. C., Narayan, O., Kennedy, P. G., and Clements, J. E. (1987). Pathogenesis of visna/maedi and caprine arthritis-encephalitis: new leads on the mechanism of restricted virus replication and persistent inflammation. Vet. Immunol. Immunopathol. 15(1-2), 167-80.


Recommended