thesis

304
MOLECULAR CHARACTERISATION, ATTENUATION AND INACTIVATION OF VERY VIRULENT INFECTIOUS BURSAL DISEASE VIRUS FOR THE DEVELOPMENT OF TISSUE CULTURE-BASED VACCINES By MAJED H. MOHAMMED Thesis Submitted to the School of Graduate Studies, Universiti Putra Malaysia, in Fulfilment of the Requirements for the Degree of Doctor of Philosophy July 2010

Upload: majed-mohammed

Post on 16-Jul-2015

287 views

Category:

Documents


4 download

TRANSCRIPT

Page 1: Thesis

MOLECULAR CHARACTERISATION, ATTENUATION AND INACTIVATION OF VERY VIRULENT INFECTIOUS BURSAL DISEASE VIRUS FOR THE

DEVELOPMENT OF TISSUE CULTURE-BASED VACCINES By

MAJED H. MOHAMMED

Thesis Submitted to the School of Graduate Studies, Universiti Putra Malaysia, in Fulfilment of the Requirements for the Degree of Doctor of

Philosophy

July 2010

Page 2: Thesis

2

DEDICATED WITH LOVE AND GRATITUDE TO:

MY DEAREST (THE SPIRIT OF MY FATHER), MOTHER, WIFE (MAYADA), TWO LOVELY SONS (ALI AND MOHAMMED)

Abstract of thesis presented to the Senate of Universiti Putra Malaysia in fulfilment of the requirement for the degree of Doctor of Philosophy

Page 3: Thesis

3

MOLECULAR CHARACTERISATION, ATTENUATION AND INACTIVATION OF VERY VIRULENT INFECTIOUS BURSAL DISEASE VIRUS FOR THE

DEVELOPMENT OF TISSUE-CULTURE BASED VACCINES

By

MAJED H. MOHAMMED

July 2010

Chairman: Profesor Dr. Mohd Hair Bin Bejo, PhD

Faculty: Veterinary Medicine

Infectious bursal disease (IBD), an economically important infectious viral

disease of poultry, is caused by IBD virus (IBDV) belonging to Avibirnavirus

genus of Birnaviridae family. The disease causes considerable mortality and

immunosuppression. Emergence of very virulent IBDV (vvIBDV) strains in

different parts of the world in late 1980‟s including Malaysia in 1991, have

demanded further research efforts in understanding the added complexicity of

the disease process and the means to control and prevent outbreaks of the

disease. Treatment of IBD is of no value and the disease can only be controlled

and prevented by proper vaccination programme and biosecurity. It was the

objectives of the study to determine the molecular characteristics and effects of

attenuation and inactivation of Malaysian field isolates of vvIBDV for tissue

culture based IBD vaccines development. Three IBDV isolates identified as

UPM04190, UPM94273 and UPM0081 with an accession number of AY791998,

AF527039 and EF208038, respectively were propagated in specific-pathogenic-

free (SPF) embryonated chickens egg via chorioallontoic membrane (CAM) for

Page 4: Thesis

4

three times and infected onto two types of continuous cell line namely the DF-1

and Vero cell lines. The UPM0081 vvIBDV isolate successfully infected these

cells while the other vvIBDV isolates failed. The virus was passaged serially 20

and 9 times in Vero cells and DF-1 cell lines, respectively. The cytopathic effects

(CPEs) were observed and virus from each passage was confirmed through

indirect immunoperoxidase staining test. The UPM0081 was adapted to Vero

cells and DF-1 cells line in fourth and third passage, respectively.

The molecular characteristics of the virus at different passages in Vero cells and

two passages in DF-1 cell line were characterized by using reverse transcriptase

polymerase chain reaction (RT-PCR). The nucleotide base sequence of a 643

bp fragment of genome segment A containing the partial coding sequence of

VP2 and the entire hyper-variable region were determined. No apparent

changes by sequence analysis of selected passage in VP2 gene at passage 5

(UPM0081T5) and passage 7 (UPM0081T7) in Vero cells and DF-1 cell line.

One amino acid substitution change occurred in passage 8 (UPM0081T8) and

passage 9 (UPM0081T9): 222 (A to P). Further changes in the VP2 gene were

recorded in passage 10 (UPM0081T10), passage 15 (UPM0081T15), and

passage 20 (UPM0081T20) 222: (A to P), 242 (I to V), 253 (Q to H), 256 (I to V),

279: (D to N), 284: (A to T), 294 (I to L), 326 (S to L), and 330 (S to R). Amino

acid substitution at positions 279 (D to N) and 284 (A to T) were commonly

found in the attenuated IBDV strains.

Page 5: Thesis

5

The pathogenicity and immunogenicity properties of the UPM0081 vvIBDV

passages 10, 15 and 20 isolates on Vero cells were evaluated in this study. The

results revealed that only UPM0081T10 was still pathogenic to SPF chickens. It

caused clinical signs, gross lesions, 25% mortality and histological changes in

bursa of Fabricius. Neither clinical signs nor gross lesions were observed in the

SPF chickens inoculated with either UPM0081T15 or UPM0081T20. Efficacy

test demonstrated that both UPM0081T15 and UPM0081T20 could provide

100% protection in highly susceptible SPF chickens when challenged with

vvIBDV (UPM0081) at virus titer of 107.8 ELD50/0.1 mL per chicken.

The UPM0081T15 and UPM0081T20 IBDV isolates were inactivated using

either Binary ethyleneimine (BEI) or Electrolysed water-Catholyte-Anolyte

(ECA). Complete inactivation of UPM0081T15 with titer of 106.7 TCID50/0.1 mL

and UPM0081T20 with titer of 107.4 TCID50/0.1ml occurred after 24 hours with

either BEI or ECA. The inactivated viruse suspension and an equal volume of

Freund‟s incomplete adjuvant were mixed together (water-in-oil) emulsion and

injected subcutaneously into 42-day-old SPF chickens to determine the safety

and immunogenicity of the inoculum. Neither clinical signs nor gross lesions

were observed in both groups of chickens before and after vvIBDV challenged.

High and protective level IBD antibody titer was recorded more in BEI than ECA

groups at 2 weeks post infection and 2 weeks post challenged. The study

showed that both the inactivated UPM0081T15 and UPM0081T20 either in BEI

or ECA was safe and could provide 100% protection against vvIBDV challenged

Page 6: Thesis

6

with titer of 107.8 EID50/ 0.1 mL, while that of ECA could not protect fully SPF

chicken against bursal lesion.

In conclusion, vvIBDV UPM0081 was successfully adapted and attenuated in

continuous cell line (Vero cells) after fifteen and twenty passages. The

attenuated and inactivatted local vvIBDV named UPM0081T15 and

UPM0081T20 conferred full protection to the immunized SPF chickens against

vvIBDV.

Abstrak tesis yang dikemukakan kepada Senat Universiti Putra Malaysia sebagai memenuhi keperluan untuk ijazah Doktor Falsafah

PENCIRIAN SECARA MOLEKUL, PELEMAHAN DAN INAKTIVASI VIRUS PENYAKIT BERJANGKIT BURSA YANG AMAT VIRULEN UNTUK PEMBANGUNAN VAKSIN YANG BERASASKAN KULTUR TISU

Page 7: Thesis

7

Oleh

MAJED H. MOHAMMED

July 2010

Pengerusi: Profesor Dr. Mohd Hair Bejo, PhD

Fakulti: Perubatan Veterinar

Wabak penyakit infeksi bursa (IBD) adalah sejenis jangkitan virus yang menular

di kalangan ayam dan menjejas kepentingan ekonomi. Penyakit ini adalah

disebabkan oleh virus penyakit bursa berjangkit (IBDV) yang tergolong dalam

genus Avibirnavirus dari keluarga Birnaviridae. IBD menyebabkan kadar

kematian yang tinggi serta boleh melemahkan imun dan daya tahan untuk

melawan penyakit. Kehadiran strain yang amat virulen IBDV (vvIBDV) di serata

dunia pada penghujung tahun 1980an, termasuk di negara Malaysia dalam

tahun 1991 telah meningkatkan keperluan kajian penyelidikan demi memahami

proses jangkitan yang kompleks serta mengenalpasti kaedah untuk mengawal

dan mencegah penyakit ini. Rawatan perubatan tidak akan memberi kesan

kecuali dengan kaedah vaksinasi serta biosekuriti. Objektif penyelidikan ini

adalah untuk membuat pencirian di peringkat molekul serta mengesan kesan

sampingan daripada proses pelemahan vvIBDV di kalangan isolat IBDV dari

Malaysia dalam sel kultur untuk tujuan perkembangan vvIBDV vaksin. Tiga

IBDV asingan tempatan yang dinamakan UPM04190, UPM94237 and

UPM0081 dengan nombor perolehan AY791998, AF527039 and EF208038

telah di biak ke dalam telur ayam spesifik-pathogen-bebas (SPF) melalui

disuntikan ke dalam membran korioalontoik (CAM) sebanyak tiga kali serta telah

Page 8: Thesis

8

suntik ke dalam dua jenis sel kultur jenis berurutan iaitu sel DF-1 dan sel Vero.

Isolat UPM0081 vvIBDV telah berjaya menyebabkan jangkitan di dalam sel

tersebut manakala asingan yang lain gagal disesuaikan ke dalam sel kultur.

Virus tersebut telah di pasage sebanyak dua puluh kali di dalam sel Vero dan

sebanyak sembilan kali di dalam sel DF-1. Kesan sitopatik (CPEs) telah

dikesan dan setiap virus dari setiap pasage telah dikenal pasti melalui ujian

imunoperoxidase tidak terus. UPM0081 telah diadaptasi ke dalam sel Vero pada

pasage yang ke empat dan di dalam sel DF-1 pada pasage yang ke tiga.

Pencirian molekul virus pada waktu yang berbeza di dalam sel Vero dan dua

pasage di dalam sel DF-1 telah dikaji melalui tindak balas transkripsi balik reaksi

rangkaian polimerasi (RT-PCR). Rangkaian nukleotida pada kedudukan 643 bp

dalam genom segmen A mempunyai separa kodon gen protein virus 2 (VP2)

dan juga seluruh bahagian variable tinggi telah dikesan. Analisis jujukan

menunjukkan beberapa pasage di dalam gen VP2 gene pada pasage 5

(UPM0081T5) dan juga pasage 7 (UPM0081T7) di dalam sel Vero dan DF-1

tidak menunjukkan sebarang perubahan. Seterusnya satu perubahan

melibatkan penukaran asid amino telah berlaku di dalam pasage 8

(UPM0081T8) dan juga pasage 9 (UPM0081T9) 222 (A to P). Perubahan

seterusnya di dalam gen VP2 telah dikesan di dalam pasage 10 (UPM0081T10),

15 (UPM0081T15), 20 (UPM0081T20): 222 (A to P), 242 (I to V), 253 (Q to H),

256 (I to V), 279 (D to N), 284 (A to T), 294 (I to L), 326 (S to L) dan juga 330 (S

to R). Perubahan asid amino pada kedudukan 279 (D to N) dan 284 (A to T)

kerap di kesan dalam IBDV strain yang lemah.

Page 9: Thesis

9

Kepatogenan dan keimunan UPM0081 vvIBDV pasage 10, 15 and 20 isolat

dalam sel Vero telah dikaji dalam kajian ini. Keputusan kajian menunjukkan

bahawa hanya UPM0081T10 masih menampilkan cirri-ciri patogenisitinya di

dalam ayam SPF. Jangkitan dengan strain ini mengakibatkan kesan klinikal

termasuk pembentukan lesi, 25% kematian dan juga perubahan patologi di

dalam bursa Fabricius. Walaubagaimanapun, pemerhatian yang sama tidak

berlaku dengan strain UPM0081T15 ataupun UPM0081T20.

Ujian keberkesanaan telah menunjukkan bahawa UPM0081T15 dan juga

UPM0081T20 boleh memberi 100% perlindungan ke atas ayam SPF yang

sangat sesuai menerima jangkitan apabila disuntik dengan vvIBDV (UPM0081)

mengunakan virus titer 107.8 ELD50/0.1 mL untuk setiap ayam.

UPM0081T15 dan UPM0081T20 IBDV isolat telah dibunuh dengan

mengunakan Binary ethyleneimine (BEI) atau Electrolysed water-Catholyte-

Anolyte (ECA). UPM0081T15 dengan virus titer 106.5 TCID50/0.1 mL dan

UPM0081T20 dengan virus titer 107TCID50/0.1mL telah dikesan mati

sepenuhnya seawal 24 jam dengan menggunakan BEI atau ECA. Virus yang

telah dibunuh berserta adjuvan Freund‟s tidak lengkap dalam kuantiti yang

sama telah di campurkan dan disuntik di bawah kulit ayam SPF berumur 42 hari

ke dalam ayam SPF untuk menguji kepatogenan dan keimunan inokulum.

Kedua dua kumpulan ayam tidak menunjukkan sebarang perubahan klinikal

selepas infeksi dengan vvIBDV. Kadar antibodi yang tinggi dan melindung

Page 10: Thesis

10

telah direkod dengan mengunakan BEI berbanding ECA pada minggu ke dua

selepas suntikan dan minggu ke dua selepas infeksi dengan vvIBDV.

Kajian penyelidikan ini menunjukkan bahawa UPM0081T15 dan UPM0081T20

yang telah dibunuh dengan mengunakkan BEI ataupun ECA adalah selamat

dan boleh menyebabkan 100% perlindungan terhadap vvIBDV dengan

menggunakan virus titer 107.1 EID50/ 0.1 mL, manakala ECA tidak dapat

memberi perlindungan yang sepenuhnya di dalam ayam SPF chicken daripada

bursal lesi di bursa Fabricius.

Kesimpulannya, vvIBDV UPM0081 telah berjaya disesuaikan dan dilemahkan

di dalam sel jenis berurutan (sel Vero) selepas lima belas hingga dua puluh

pasage. Virus vvIBDV daripada asingan tempatan ini yang lemah dan telah

dimatikan dan dinamakan sebagai UPM0081T15 dan UPM0081T20 boleh

memberi perlindungan sepenuhnya kepada ayam SPF terhadap jangkitan

vvIBDV.

ACKNOWLEDGEMENT

All praise for Almighty Allah, Lord of all creations Who has granted me

His blessings throughout my life and backed me up to luxuriate in

the researches of this study.

Page 11: Thesis

11

I would like to express my heartiest gratitude and appreciation to my supervisor,

Professor Dr. Mohd Hair Bejo for providing his invaluable advice, constant

guidance, encouragement and incitement that has stimulated me to accomplish

my PhD research. I want to thank him for granting me a generous opportunity to

work in his laboratory as a graduate student. His honest advice, patience,

thorough guidance and calm demeanor has steered my research towards

success. He challenged me to set my bench mark even higher and to look for

solutions to problems rather than focus on the problem. I have learned to have

confidence in myself and in my work as a result. And I would like to thank him

for his never ending support he had for me during my long journey of doctorate

study program. He was the brother, the friend, and even sometimes the father

who I lost before being my research advisor, reconstruct my whole life by

teaching me the true meaning of doing my best for anything encountered, and to

set goals more aggressive and ambitious.

Thank you professor.

I would like to express my sincere thanks and appreciation to Professor Datin

Paduka Dr. Aini Ideris, and Professor Dr. Abdul Rahman Omar, my co-

supervisors for their constructive instructions, proper guidance and motivation

throughout my study period.

One more time I would like to thank gratefully each of Mr. Saipuzaman Ali, Mr.

Mohd Kamaruddin and Mrs. Siti Khadijah the laboratory staffs. And also to all

Page 12: Thesis

12

my friends in the laboratory for always being willing to render assistance

throughout the course of my study.

I would also like to thank Universiti Putra Malaysia for the award of the graduate

research fellowship (GRF) which has supported me during my study.

I have no words to express gratitude to my family in Iraq, (the spirit of my father)

and my mother who always encouraged me to obtain higher education, special

thanks to my brother, sister, my wife and all other family members for their moral

support and countless prayers throughout the course of my life.

May Allah give them a long, prosperous and happy life (Aa‟meen)

I certify that an Examination Committee met on 6th July 2010 to conduct the final examination of Majed H. Mohammed on his Doctor of Philosophy thesis entitled “Molecular Characterisation, Attenuation and Inactivation of Very Virulent Infectious Bursal Disease Virus for the Development of Tissue-Culture Based Vaccines” in accordance with Universiti Pertanian Malaysia (Higher Degree) Act 1980 and Universiti Pertanian Malaysia (Higher Degree) Regulations 1981. The Committee recommends that the candidate be awarded the relevant degree. Members of the Examination Committee are as follows:

RASEDEE @ MAT BIN ABDULLAH, PhD Professor, Faculty of Veterinary Medicine, Universiti Putra Malaysia. (Chairman)

Page 13: Thesis

13

SITI SURI ARSHAD, PhD Associate Professor Faculty of Veterinary Medicine Universiti Putra Malaysia (Member) JASNI BIN SABRI, PhD Associate Professor Faculty of Veterinary Medicine Universiti Putra Malaysia (Member) EMDADUL HAQUE CHOWDHURY, PhD Professor, Department of Pathology Faculty of Veterinary Science Bangladesh Agriculture Science 2202 Mymensingh Bangladesh

___________________________________

HASANAH MOHD GHAZALI, PhD Professor and Dean School of Graduate Studies Universiti Putra Malaysia Date:

This thesis submitted to the Senate of Universiti Putra Malaysia has been accepted as fulfilment of the requirement for the degree of Doctor of Philosophy. The members of the Supervisory Committee were as follows: Mohd Hair Bejo, PhD Professor Faculty of Veterinary Medicine Universiti Putra Malaysia (Chairman) Abdul Rahman Omar, PhD Professor Faculty of Veterinary Medicine Universiti Putra Malaysia (Member) Aini Ideris, PhD Professor

Page 14: Thesis

14

Faculty of Veterinary Medicine Universiti Putra Malaysia (Member)

HASANAH MOHD GHAZALI, PhD

Professor and Dean School of Graduate Studies Universiti Putra Malaysia Date: 12 August 2010

DECLARATION I declare that the thesis is my original work except for quotations and citations which have been duly acknowledged. I also declare that it has not been previously, and is not concurrently, submitted for any other degree at Universiti Putra Malaysia or at any other institution.

MAJED H. MOHAMMED

Date: 6 July 2010

Page 15: Thesis

15

TABLE OF CONTENTS

Page

DEDICATION ii

ABSTRACT iii

ABSTRAK vii

ACKNOWLEDGEMENTS xi

APPROVAL xiii

DECLARATION

LIST OF TABLES

LIST OF FIGURES

LIST OF ABBREVIATIONS

CHAPTER

xv

xxi

xxiv

xxix

1 INTRODUCTION 1

2 LITERATURE REVIEW 2.1 Infectious Bursal Disease 9 2.1.1 Clinical Signs and Gross Lesions 11 2.1.2 Histopathology 14 2.1.3 Pathogenesis 16 2.1.4 Immunosuppression 18 2.1.5 Epidemiology of IBD 20 2.1.6 Transmission 21 2.2 Infectious Bursal Disease Virus 22 2.2.1 IBDV Genome 23 2.2.2 IBDV Proteins 25 2.2.3 Antigenic and Virulence Variation 27 2.3 Isolation Adaptation and Attenuation of IBDV 31 2.3.1 Chicken Embryos 31 2.3.2 Cell Culture 32 2.4 General Information on the Immune System 36 2.4.1 Innate Immunity 37 2.4.2 Adaptive Immunity 38 2.4.3 Humoral (B-cell mediated) Immunity 38 2.4.4 Cell-mediated (T-cell mediated)Immunity 39 2.4.5 Relationship between B-and-T-cells 40 2.4.6 Effect of IBDV on innate immunity 40 2.4.7 Effect of IBDV on humoral immunity 40 2.4.8 Effect of IBDV on cellular immunity 41 2.5 Vaccination 42 2.5.1 Live Virus Vaccines 43

Page 16: Thesis

16

2.5.2 Inactivated of Virus 46 2.5.3 Recombinant and DNA Vaccine 48 2.5.4 Anti-viral Drugs 51 3 ADAPTATION AND ATTENUATION OF vvIBDV ISOLATES

IN TISSUE CULTURE FOR DEVELOPMENT OF VACCINES 53

3.1 Introduction 53 3.2 Materials and Methods 57 3.2.1 IBDV Isolates 57 3.2.2 IBDV Inoculums Preparation 58 3.2.3 Propagation of Viruses in SPF Embryonated Chicken

Eggs via Chorioallantoic Membrane 58

3.2.4 Adaptation, Replication and Attenuation of vvIBDV in Cell Culture

60

Vero Cell Line 60 DF-1 Cell Line 60 3.2.5 Resuscitation of Frozen Cell Line 61 3.2.6 Sub Culturing of Adherent Monolayer 61 3.2.7 Infection of Vero Cell and DF-1 Cells Monolayer 62 3.2.8 Harvesting of Virus 63 3.2.9 Adaptation and Attenuation 63 3.2.10 Tissue Culture Infective Dose 50 (TCID50) 64 3.2.11 IBDV Identification and Confirmation 64 3.2.12 Indirect Immunoperoxidase Staining Test 65 3.3 Rusults 66 3.3.1 Chorio-allantoic Membrane for UPM94372 66 3.3.2 Chorio-allantoic Membrane for UPM04019 66 3.3.3 Chorio-allantoic Membrane for UPM0081 67 3.3.4 IBDV Replication and Adaptation in Vero Cell Line 70 3.3.5 IBDV Replication and Adaptation in DF 1 Cell Line 70 3.3.6 IBDV Titration (TCID50/ml) 75 3.3.7 IBDV Identification though Indirect Immunoperoxidase

Staining (IIPS) Test 75

3.4 Discussion 84 4 MOLECULAR CHARACTERIZATION OF THE ADAPTED

AND ATTENUATTED vvIBDV ISOLATE 89

4.1 Introduction 89 4.2 Materials and Methods 92 4.2.1 Sample Preparation 92 4.2.2 RNA Extraction 92 4.2.3 Determination of RNA Concentration 93 4.2.4 Primer Design 94 4.2.5 Reverse Transcription and PCR Reaction 94 4.2.6 Gel Electrophoresis and Ethidium Bromide Staining 95 4.2.7 Purification of RT-PCR Products 96 4.2.8 Molecular Cloning of Amplified Products and Analysis 97

Page 17: Thesis

17

of Recombinant Plasmid 4.2.9 Plasmid Extraction and Purification 98 4.2.10 Sequence Assembly and analysis Using

Bioinformatics Software 99

4.2.11 Phylogenetic Tree Construction 101 4.3 Results 101 4.3.1 Amplification of the Hypervariable Region of VP2

Gene 101

4.3.2 PCR Analysis of Recombinant Colonies 102 4.3.3 Nucleotide Sequence Analysis 102 4.3.4 Amino Acid Sequence Analysis 104 4.3.5 Phylogenetic Analysis 105 4.4 Discussion 130 5 PATHOGENICITY AND IMMUNOGENCITY OF THE

ATTENUATED vvIBDV IN SPF CHICKENS

134

5.1 Introduction 134 5.2 Materials and Methods 137 5.2.1 Chickens 137 5.2.2 Selection of IBDV Isolates 138 5.2.3 Adaptation of IBDV to Embryonated SPF Eggs 138 5.2.4 Tissue Culture Infective Dose 50 (TCID

50) 138

5.2.5 Experimental Design 138 5.2.6 Experiment 1 139 5.2.7 Experiment 2 140 5.2.8 IBD Challenge 142 5.2.9 Histopathology 142 5.2.10 Histopathological Lesion Scoring 143 5.2.11 Collection of Samples for Serological Test 143 5.2.12 Antibody Assay 144 5.2.13 Reverse Transcriptase Polymerase Chain Reaction

(RT-PCR) 144

5.2.14 Statistical Analysis 145 5.3 Rusults 145 5.3.1 Clinical Signs 145 Experiment 1 145 Experiment 2 146 5.3.2 Body Weight 149 Experiment 1 149 Experiment 2 150 5.3.3 Bursa Weight 151 Experiment 1 151 Experiment 2 152 5.3.4 Bursa to Body Weight Ratio 153 Experiment 1 153 Experiment 2 154

Page 18: Thesis

18

5.3.5 Gross Pathology 155 Experiment 1 155 Experiment 2 156 5.3.6 Histopathological Changes and Lesion Scoring 163 Experiment 1 163 Experiment 2 164 5.3.7 Enzyme Linked Immunosorbent Assay (ELISA) 181 Experiment 1 181 Experiment 2 181 5.3.8 Detection of the Virus or Viral RNA using RT-PCR 182 5.4 Discussion 183 6 SAFETY AND IMMUNOGENICITY OF THE

INACTIVATED ATTENUATED vvIBDV IN SPF CHICKENS 187

6.1 Introduction 187 6.2 Materials and Methods 191 6.2.1 Virus and Cells 191 6.2.2 Harvesting of Virus 191 6.2.3 Tissue Culture Infective Dose 50 (TCID

50) 192

6.2.4 Inactivation of vv IBDV 192 Binary ethylenmine (BEI) Treatment 192 Electrolysed water-Catholyte-Anolyte (ECA)

Treatment 193

6.2.5 Determination of Time Required to Inactivate Virus 193 6.2.6 Perparation of Killed- Virus Oil Emulsion 194 6.2.7 Experimental Design 194 6.2.8 Microscopic Examination and Lesion Score 195 6.2.9 Determination of ELISA Titer Against Inactivated

IBDV Vaccine 196

6.2.10 Reverse Transcriptase Polymerase Chain Reaction (RT-PCR)

196

6.2.11 Statistical analysis 197 6.3 Results 197 6.3.1 Inactivation of the Virus Attenuated vvIBDV 197 6.3.2 Clinical Signs 198 6.3.3 Body Weight 200 6.3.4 Bursa Weight 201 6.3.5 Bursa to Body Weight Ratio (1x10-3) 202 6.3.6 Gross Lesions 203 6.3.7 Histological Lesions Score 207 6.3.8 Antibody Titer (ELISA) 212 6.3.9 Detection of the Virus or Viral RNA using RT-PCR 213 6.4 Discussion

214

7 GENERAL DISCUSSION, CONCLUSION AND 220

Page 19: Thesis

19

RECOMENDATION FOR FUTURE RESEARCH

7.1 General Discussion 220 7.2 Conclusion 226 7.3 Recommendation for Further Research 228

BIBLOGRAPHY 230 APPENDICES 260 BIODATA OF STUDENT 271 LIST OF PUBLICATIONS 273

Page 20: Thesis

20

LIST OF TABLES

Table Page

3.1 Mortality of SPF embryonated eggs following vvIBDV inoculation into CAM route

70

3.2 Percentage of CPE monolayer Vero cells following UPM0081 vvIBDV inoculation

73

3.3 Percentage of CPE monolayer DF-1 cells following vvIBDV inoculation

74

3.4 Virus titer determined by tissue culture Infective Dose 50 (TCID50) 75

4.1 Primers used to amplify the HPVR VP2 gene 94

4.2 IBDV isolates used in the sequence analyses 100

4.3 Number of nucleotide differences in HPVR of VP2 gene between IBDV isolate

109

4.4 Sequence identity matrix of VP2 genes nucleotides of IBDV isolates

110

4.5 Summary of the proposed molecular markers (amino acid residues) of UPM0081T10, UPM0081T15 and UPM0081T20 atIBDV isolates with other published IBDV strains

111

4.6 Number of amino acids differences in HPVR of VP2 gene between IBDV isolates

112

4.7 Sequence identity matrix of VP2 genes amino acids of IBDV isolates

113

5.1 Groups of SPF chickens inoculated with attenuated vvIBDV passage 15 and 20 and challenged with vvIBDV at day 14 post inoculation

141

5.2 Rate of mortality and the percentage of protection based on the number of chickens that survived at day 7 post challenged

149

5.3 Experiment 1: body, bursa, bursa to body weight ratio (1 x 103), lesion scoring and ELISA titer of SPF chicken inoculated

175

Page 21: Thesis

21

attenuated vvIBDV and control group

5.4 Experiment 2: body weight (g) of chickens in the inoculated attenuated vvIBDV and control group

176

5.5 Experiment 2: body, bursa, bursa to body weight ratio (1 x 103) and lesion scoring of SPF chicken inoculated attenuated vvIBDV and uninoculated challenge group

177

5.6 experiment 2: body, bursa, bursa to body weight ratio (1 x 103) and lesion scoring of SPF chicken inoculated attenuated vvIBDV and control group

177

5.7 Experiment 2: bursa weight (g) of chickens in the inoculated attenuated vvIBDV and control group

178

5.8 Experiment 2: bursa to body weight ratio (1 x 103) of chickens in the inoculated attenuated vvIBDV and control group

179

5.9 Experiment 2: lesions scoring of chickens in the inoculated attenuated vvIBDV and control group

180

5.10 Antibody titers (mean titer ± standard deviation) to IBD determined by ELISA in the attenuated vvIBDV inoculated groups

182

6.1 Different time interval to inject SPF embryonated eggs by two kinds of killed vvIBDV (BEI and ECA)

193

6.2 Different groups of chickens inoculated with two types of inactivated vvIBDV (BEI and ECA) and the control group

195

6.3 Mortality of SPF embryonated eggs following inoculation (BEI and ECA) into CAM route

197

6.4 Efficacy of the inactivated attenuated vvIBDV (UPM0081) in SPF chickens

200

6.5 Body weight of chickens in the inactivated attenuated vvIBDV inoculated and control group at 2 weeks post challenged

201

6.6 Bursa weight of chickens in the inactivated attenuated vvIBDV inoculated and control group at 2 weeks post challenge

202

6.7 Bursa to body weight ratio of chickens in the inactivated attenuated vvIBDV inoculated and control group at 2 weeks post challenged

203

6.8 Lesion score of chickens in the inactivated attenuated vvIBDV 212

Page 22: Thesis

22

inoculated and control group at 2 weeks post challenge

6.9 Antibody titers to IBDV determined by ELISA in the inactivated attenuated vvIBDV inoculated and uninoculated groups after two weeks of post inoculated and two weeks post challenged

213

Page 23: Thesis

23

LIST OF FIGURE Figure Page

3.1a

3.1b

(A):Uninfected control embryonated SPF chicken eggs. (B): UPM94273 dead embryo with severe haemorrhage (C): UPM04019 dead embryo with severe haemorrhage. (D): UPM0081 the embryo infected showed severe petechial to ecchymotic haemorrhage (arrows)

68 69

3.2 (A) Uninfected control Vero cells monolayer. (B) Cytopathic effect of UPM0081 isolate in 4th passage days 15 pi. The arrows show cell rounding and aggregation. 10 x . Bar = 200 µm

76

3.3 (A) Vero cell monolayer in 6th passage days 8 pi (B). Vero cell monolayer in passage 12th, days 6 pi. The arrows shows cell rounding and aggregate in clumps and granulated in cytoplasm. 10x. Bar = 200 µm

77

3.4 (A) Vero cell monolayer in passage 13th, day 3 pi (B). Vero cell monolayer in 20th passage days 4 pi. The detachment of cells from the substrate, with the eventual destruction of the entire monolayer. 10 x. Bar = 200 µm

78

3.5 (A) Uninfected control DF-1 cells monolayer. (B) Cytopathic effect of UPM0081 isolate in 3rd passage days 5 pi. The arrow shows cell rounding and clumping. 10 x. Bar = 200 µm

79

3.6 (A) DF-1 monolayer in 4th passage day 5 pi, affected cells were more concentrated with granular cytoplasm (B). DF-1 cells passage 5th day 4 pi, the arrow shows detachment of cells from the substrate (B). 10 x. Bar = 200 µm

80

3.7 (A) DF-1 cell monolayer in passage 6th, day 3 pi (B). DF-1 cell monolayer in 9th passage, days 3 pi the arrow shows degenerated cells and more detachment of cells from the substrate. 10 x. Bar = 200 µm

81

3.8 Identification of IBD antigens in Vero cells culture using infected cell cultures stained with HRP-conjugated antibody. (A) Uninfected control Vero cells. (B) Vero cells infected with UPM0081 at 20th passage days 2 pi. Note specific intracytoplasmic brownish colouration. 10 x. Bar = 200 µm

82

3.9 Identification of IBD antigens in DF-1 cells culture using infected cell cultures stained with HRP-conjugated antibody. (A) Uninfected control DF-1. (B) DF-1 infected with UPM0081 at passage 4 day 2 pi. Note

83

Page 24: Thesis

24

specific intracytoplasmic brownish colouration. 10x. Bar = 200 µm

4.1 Hypervariable region (643pb) of IBDV VP2 genes Lane 1- Negative control; Lane 2 positive UPM0081D5; Lane 3 positive UPM0081D7; Lane 4 positive UPM0081T5 and Lane 5 positive UPM0081T7; M- 100 bp DNA marker (Promega, USA)

107

4.2 Hypervariable region (643pb) of IBDV VP2 genes Lane 1- positive UPM0081T8; Lane 2 positive UPM0081T9; Lane 3 positive UPM0081T10; Lane 4 positive UPM0081T15 and Lane 5 positive UPM0081T20; Lane 6- Negative control; M- 100 bp DNA marker (Promega, USA)

107

4.3 PCR screening on white colonies amplification of IBDV genes Lane 1, 2 and 3 white colonies positive for VP2 gene passages (UPM0081D5, UPM0081T5 and UPM0081T7 respectively; Lane 4 Negative control; M- 100 bp DNA marker (Promega, USA)

108

4.4 PCR screening on white colonies amplification of IBDV genes Lane 1,2,3,4,5,6 and 7 white colonies positive for VP2 gene passages (UPM0081D7, UPM0081T8, UPM0081T9, UPM0081T10, UPM0081T15 and UPM0081T20 respectively; Lane 7 Negative control; M- 100 bp DNA marker (Promega, USA)

108

4.5 Nucleotide sequences of HPVR of VP2 from nucleotide 516-1158 (numbering of Bayliss et al., 1999) of the UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages compared with other published IBDV strains. A dot indicated position where the sequence is identical to others

114

4.6 Amino acid sequence aligment of UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages

121

4.7 Phylogenetic tree based on nucleotide sequence of HPVR of VP2 gene of IBDV isolates, displaying relationship of UPM0081T10, UPM0081T15 and UPM0001T20 passages and other published atIBDV strains

124

4.8 Phylogenetic tree based on amino acids sequence of HPVR of VP2 gene of IBDV isolates, displaying relationship of UPM0081T10, UPM0081T15 and UPM0001T20 passages and other published atIBDV strains

125

4.9 Sequence nucleotide difference of VP2 genes of IBDV isolates

126

4.10 Sequence nucleotide identity matrix of VP2 genes of IBDV isolates

127

Page 25: Thesis

25

4.11 Sequence nucleotide difference of VP2 genes of IBDV isolates

128

4.12 Sequence amino acid identity matrix of VP2 genes of IBDV isolates

129

5.1a

5.1b

Experiment 1 (preliminary study): bursa of Fabricius in SPF chickens day 14 pi. (A) Group C: normal. (B) Group A: Passage 10 bursa of Fabricius with mild to moderate odema with yellowish gelatinous material (arrow) Experiment 1 (preliminary study): bursa of Fabricius in SPF chickens day 14 pc. (C) Group B: passage 15 normal

157

158

5.2a

5.2b

Experimental 2 (challenged groups): bursa of Fabricius in SPF chickens day 7pc. (A) Group1 (b): passage 15 normal. (B) Group 2(b): passage 20 normal. Experimental 2 (challenged groups): bursa of Fabricius in SPF chickens. (C) Group 3(b): control positive severee haemorrhages day 4 pc.

159

160

5.3a

5.3b

Experiment 2 (challenged groups): proventriculus and gizzard in SPF chickens day 7pc. (A) Group 1 (b): passage 15 normal (B) Group 2 (b): passage 20 normal Experiment 2 (challenged groups): proventriculus and gizzard in SPF chickens day 7pc. (C) Group 3 (b): control positive hemorrhage on the mucosa of the proventriculus at the junction with the gizzard (arrow).

161

162

5.4a

5.4b

Experiment 1 (preliminary study): day 14 pi. bursa of Fabricius (A) Control group: No lesions were observed lesion score of 0 (B) Group B: Normal, large active follicles consist of lymphoid cells (arrow) lesion score of 0 (. HE, 10x. Bar = 200µm. Experiment 1 (preliminary study): bursa of Fabricius. (C) Group A: Oedematous bursa with degeneration, necrosis (arrow) and infiltration of inflammatory cells (arrow), follicular cyst (arrow) in the medulla, lesion score of 5 at day 2 pi. (D) Group A: More severe lymphoid necrosis (arrow) in the mudella, lesion score of five at day 5 pi. HE, 20x. Bar = 100 µm

167

168

5.5a

5.5b

Experiment 2 (sacrificed groups): day 7 pi. bursa of Fabricius. (A) Group 1(a): Mild degeneration and necrosis of the follicles (arrow) lesion score of 1 (B) Group 2(a) Mild degeneration and necrosis of the follicles (arrow) lesion score of 1. HE, 10x. Bar = 200µm Experiment 2 (sacrificed groups): day 7 pi. bursa of Fabricius. (C) Group 3(a): very clear cortex and medulla packed with healthy follicles, lesion score of 0. HE, 10x. Bar = 200µm

169

170

Page 26: Thesis

26

5.6a

5.6b

Experiment 2 (challenged groups): day 7 pc. bursa of Fabricius. (A) Group 1(b): Mild degeneration and necrosis of the follicles (arrow), lesion score of 1 (B) Group 2(b) Mild degeneration and necrosis of the follicles (arrow) lesion score of 1 HE, 10x. Bar = 200µ

Experiment 2 (challenged groups): day 7 pc. (C) Group 3(b): Depletion of bursa follicles with cysts contains cell debris with fibrinous exudates at medulla follicle (arrow), the interstitial connective tissues were obvious, edematous and infiltrated with inflammatory cells (arrow), lesion score of 5. HE, 20x. Bar = 100µm

171

172

5.7a

5.7b

Experiment 2 bursa of Fabricius (mortality groups): day 7 pc. (A) Group 1(c): Mild lymphoid deplesion (arrow), lesion score of 1 (B) Group 2(c): Mild lymphoid deplesion (arrow), lesion score of 1. HE, 10x. Bar = 200µm Experiment 2 (mortality groups): day 7 pc. bursa of Fabricius. (C) Group 3(c): Mild lymphoid deplesion (arrow), lesion score of 1. HE, 10x. Bar = 200µm

173

174

5.8 Hypervariable region (643pb) amplification of IBDV VP2 genes. Lane 1 Day 1; Lane 2 Day 3; Lane 3 Day 5 Day ; Lane 4 Day 7; Lane 5 Day

183

10; Lane 6 Day 14; Lane 7 Day 21; and Lane 8 Negative control; M- 100 bp DNA marker (Promega, USA)

6.1a

6.1b

bursa of Fabricius (BF) in SPF chickens. (A) Group C1: normal (B) Group C2: severee haemorrhagic bursa of Fabricius (BF) in SPF chickens. (C) Group BEIP15: normal (D) Group BEIP20: normal.

205

206

6.2 bursa of Fabricius (A) Group C1 (Control negative): Apparently normal lymphoid follicles, lesion score of 0 (B) Group C2 (Control positive): lesion score of 5, day 2 pi, severe follicular necrosis with cyst formation on the follicles (arrow) and infiltration of inflammatory cells and oedema fluid at interstitial space (arrow). HE, 20x. Bar = 100µm

209

6.3 bursa of Fabricius day 14 pc. (A) Group BEIP15: Mild degeneration and necrosis of the follicles (arrow), lesion score of 1 (B) Group BEIP20: Mild degeneration and necrosis of the follicles (arrow), lesion score of 1. HE, 10x. Bar = 200µm

210

6.4 bursa of Fabricius day 14 pc. (A) Group ECAP15: Mild to moderate lymphoid necrosis (arrow), lesion score of 1.5. (B) Group ECAP20: Mild to moderate lymphoid necrosis (arrow), lesion score of 1.5. HE, 10x. Bar = 200µm

211

Page 27: Thesis

27

6.5 Hypervariable region (643pb) amplification of IBDV VP2 genes. (1)

BEIP15 negative (2) BEIP20 negative (3) ECAP15 negative (4) ECAP20 negative (5) C2 posative. (M) 100 bp DNA marker (Promega, USA).

214

Page 28: Thesis

28

LIST OF ABBREVIATIONS

ATV Antibiotic-trypsin versene

AA Amino acid sequences

AGID Agar gel immunodiffusion

AGPT Agar gel precipitation test

atIBDV Attenuated strain of infectious bursal disease virus

BEI Binary ethylenimine

BF Bursa of Fabricius

bp Base pair

CAM Chorioallantoic membrane

caIBDV Classical strain of infectious bursal disease virus

cDNA Complementary deoxyribonucleic acid

CEF Chicken embryo fibroblast

CMI Cell-mediated immunity

DAB Diaminobenzidine tetrahydrochloride

ddH2O Deionized double-distilled water

DMSO Dimethylsulphoxide

DNA Deoxyribonucleic acid

dNTP Deoxynucleoside triphosphate

dsDNA Double-stranded DNA

ECA Electrolysed water-Catholyte-Anolyte

EID50 Embryo effective dose fifty

ELISA Enzyme-linked immunosorbent assay

FBS Fetal bovine serum

HE Haematoxylin-and-eosin

HPVR Hypervariable region

IBD Infectious bursal disease

IBDV Infectious bursal disease virus

IPNV Infectious pancreatic necrosis virus

IPS Immunoperoxidase staining technique

Page 29: Thesis

29

IPTG Isopropyl-ß-D-thiogalactosidase

kb Kilobase pair

kD Kilo Dalton

LB Luria-Bertani

Min Minute

NaCl Sodium chloride

nt Nucleotide

OD Optical density

OIE Office international des epizooties

ORF Open reading frame

PBS Phosphate-buffered saline

pH Hydrogen ion exponent

pi post infection

% Percentage

PCR Polymerase chain reaction

RNA Ribonucleic acid

rpm Revolution per minute

RT-PCR Reverse transcriptase-polymerase chain reaction

RT Room temperature

SPF Specific-pathogen-free

TAE Tris-acetate-EDTA

UPM Universiti Putra Malaysia

vaIBDV Variant strain of infectious bursal disease virus

Vero Green Monkey Kidney

vvIBDV Very virulent strain of infectious bursal disease virus

w/v Weight per volume

X-gal 5-bromo-4-choro-3-indolyl-ß-D-galactopyranoside

µg Microgram

µl Microliter

µm Micrometer

Page 30: Thesis

30

Amino Acid Single/Three Letter Amino Acid Code

Alanine A Ala

Arginine R Arg

Asparagine N Asn

Aspartic Acid D Asp

Glutamine Q Gln

Glutamic Acid E Glu

Glycine G Gly

Isoleucine I IIe

Leucine L Leu

Lycine K Lys

Methionine M Met

Phenylalanine F Phe

Proline P Pro

Serine S Ser

Threonine T Thr

Thyptophan W Trp

Valine V Val

Page 31: Thesis

31

CHAPTER 1

INTRODUCTION

Infectious bursal disease virus (IBDV) also called Gumboro disease after the

geographic location in the state of Delaware where the first outbreak occurred,

causes immunosuppression in young chickens and an acute disease in chickens

between 3 to 6 weeks old (Ramm et al., 1991). The disease is endemic with

95% presence as reported by of the Office of International Epizooties (OIE)

member countries, in spite of intensive vaccination and biosafety practices (van

den Berg., 2000). This indicates that the current control measures of this virus

are not very effective.

The disease causes economic losses due to increase susceptibility to other

pathogens (bacterial, viral and protozoan) and decrease vaccination efficacy.

Impaired growth and death are also common and the mortality rates do vary

from insignificant levels to 100%, depending on the strain involved in the

outbreak (Lasher and Shane, 1994).

The target organ of IBDV is the bursa of Fabricius, which is a specific reservoir

for B lymphocyte cells in avian species. The severity of the disease has been

reported to be directly related to the number of susceptible cells present in the

bursa. Therefore, the age range of chickens susceptible to IBDV infection is

between 3 to 6 weeks, when the bursa of Fabricius is at its maximum

Page 32: Thesis

32

development. Massive growth of the virus in the bursal cells causes cellular

destruction and the subsequent dissemination of the virus causes disease and

death.

Infection with IBDV often results in immunosuppression (Allan et al., 1972). The

immunosuppressive effects with classical IBDV (caIBDV) appears to be more

pronounced if the virus exposure occurs within the first 2-3 weeks of age of the

chickens as the degree of immunosuppression varies, depending on the

virulence of the virus and time of infection. Immunosuppression may be

accompanied by overt clinical or subclinical outbreaks of infectious bursal

disease (IBD). In this case, the humoral immune response is clearly depressed,

but transient depression of the cellular immune response occurs (Confer et al.,

1981).

IBDV is a member of the genus Avibirnavirus in the family Birnaviridae. The

member of this family contain a genome consists of two segments of double-

stranded RNA (dsRNA), designated A and B (Dobos et al., 1979; Muller et al.,

1979b), with icosahedral symmetry and a diameter of about 60nm (Hirai and

Shimakura, 1974). The virus has five proteins recognized as VP1 to VP5. The

smaller RNA segment known as segment B of the genome, with a length of

about 2.8 kb encodes for VP1, which is a 90-kD multifunctional protein with

polymerase and capping enzyme activities (Spies et al., 1987). The larger

segment A with a length of about 3.2 kb encodes for VP2, VP3, VP4 and VP5.

The VP2 and VP3 are the major proteins of the virions constituting 51% and

Page 33: Thesis

33

40%, respectively of the total proteins and contain the major neutralizing

epitopes. The VP2 has the serotype specific epitope and VP3 has a group

specific antigen. VP4 is a minor protein involved in the processing of the

precursor polyprotein (Fahey et al., 1989).

Many IBDV has been characterized molecularly using the hypervariable region

which is located in VP2 of IBDV genome (Brown et al., 1994). This region

encodes the main host protective immunogen polypeptides of the virus (Azad et

al., 1987; Becht et al., 1988; Fehey et al., 1989). It consists of 145 amino acids

from amino acid positions 206 to 350 and within this region there are two

hydrophilic peaks. The first peak (peak A) is from amino acid positions 212 to

224 and the second peak (peak B) is from amino acid positions 314 to 324

(Bayliss et al., 1990; Heine et al., 1991; Brown et al., 1994). Specific amino acid

changes within hypervariable region and serine heptapeptide motif sequence

(SWSASGS), which is adjacent to peak B, are potential sites responsible for

virus attenuation or antigenic determination (Heine et al., 1991; Yamaguchi et

al., 1996b). In addition, several amino acid molecules change at the

hypervariable domain of the VP2 gene which has been used to differentiate the

virus into very virulent, attenuated, variant and classical strains. The amino acid

residue changes at 222 (P to A), 242 (V to I), 253 (H to Q), 256 (V to I), 294 (L to

I), and 299 (N to S), are the markers for vvIBD (Cao et al; 1998; Brown et al.,

1994; Rudd et al., 2002) while the marker for variant strain are at 245 (G to S)

and 249 (Q to K) and that of attenuated strains are at 279 (D to N), and 284 (A

to T) (Yamaguchi et al., 1996b; Cao et al., 1998).

Page 34: Thesis

34

The cloacal bursa and spleen are the tissue of choice for the isolation of IBDV,

but the bursa is the most common tissue chosen to isolate IBDV. Other organs

contain the virus, but at a lower concentration and probably only because of the

viremia (Lukert and Saif, 2003). The chorioallontoic membrane (CAM) of 9-11

days old embryos was also the most sensitive route for isolation of the virus

(Hitchner, 1970). The IBDV do infect and grow in various primary cell culture of

avian origin like chicken embryo kidney (CEK), chicken embryo bursa (CEB) and

chicken embryo fibroblast (CEF) cells (Raymond and Hill, 1979; Yamaguchi et

al., 1996a).

Mammalian continuous cell lines had also been reported to be susceptible to

IBDV and these include RK-13 derived from rabbit kidney (Rinaldi et al., 1972),

Vero cells derived from adult African green monkey kidney (Leonard, 1974;

Jackwood et al., 1987; Kibenge et al., 1988; Peilin et al., 1997; Ahasan et al.,

2002) BGM-70 derived from baby grivet monkey kidney (Jackwood et al., 1987),

MA-104 derived from foetal rhesus monkey (Jackwood et al., 1987), and OK

derived from ovine kidney (Kibenge and Mckenna, 1992).

The use of these continuous cell lines of mammalian origin has been found to

have advantages over the use of primary cell culture of avian origin. Continuous

cell lines are easier to handle and maintain compared to primary cell culture,

and are free from vertically transmitted extraneous viruses (Hassan et al., 1996).

Its usage will be timely for laboratories that have limited or no access to specific

pathogen free (SPF) eggs or chicks. Thus, if higher virus titer could be obtained

Page 35: Thesis

35

from continuous cell lines, it will be valuable and economical to adopt the cell

lines to grow IBDV.

Conventional immunizations with live and killed vaccine are the principle

methods for control of IBD in chickens. Live virus vaccines are generally derived

from the serial passages in embryonated eggs or tissue culture (van den Berg,

2000). The degree of attenuation of the vaccine strains can be classified as mild,

intermediate and hot depending on the its ability to cause the varying degree of

histological lesions. Although serotype 1 vaccine strains cause no mortality, its

use still cause different degrees of bursal lesions that range from mild to

moderate or even severe (van den Berg, 2000). The higher the virulence of the

vaccine virus strain, the more severe damage of the bursal lymphocytes resulted

(Kelemen et al., 2000). Nonetheless, as it should be, the lesion caused by the

vaccine strain is less severe than the field strain (Rosales et al., 1989a).

The major problem with active immunization of maternally immune chickens is

ability to determine the proper time of vaccination that allows for adequate

replication of the vaccine virus and at the same time protects young chicken

from disease. The time of vaccination varies with the level of maternal

antibodies, route of vaccination and virulence of the vaccine virus. For a

successful vaccination program, factors like environmental stresses,

management and flock profiling for the presence of maternal antibodies should

be taken into account (Lukert and Saif, 2003). Inactivated vaccines are usually

used in the breeder hens for them to pass down high, uniform, and persistent

Page 36: Thesis

36

antibody titres to the progeny (Cullen and Wyeth, 1976; Wyeth and Cullen,

1978; Wyeth and Cullen, 1979; Guittet et al., 1992). For the vaccination to be

effective, the hens must be previously vaccinated with a live virus or had been

exposed to the virus in the farm. Inactivated vaccines are administered to the

layers through subcutaneous or intramuscular routes at sixteen to twenty week

old. In this way, the chicks will have the protective maternal antibodies up to

thirty days (Wyeth and Cullen, 1979; Box, 1989; van den Berg and Meulemans,

1991; Wyeth et al., 1992). However, the chicks will not be protected from the

challenge of the highly pathogenic IBDV strains at later age (Wyeth and Cullen,

1979; Van den Berg and Meulemans, 1991).

Inactivated vaccine is usually prepared from the bursal homogenates of infected

chicks, or from viral cultures on embryonated eggs or tissue culture, where the

virus is inactivated by formaldehyde and various alkylating agents like

Binaryethylenimine (BEL), betapropiolactone and prepared as the oil emulsions

(van den Berg, 2000). Killed virus vaccines in oil adjuvant are used to boost and

prolong immunity in breeder flocks, but they are not practical and desirable for

inducing a primary response in young chicken (Lukert and Saif, 2003). Oil-

adjuvant vaccines are most effective in chicken that have been primed with live

virus either in the form of vaccine or field exposure to the virus (Wyeth and

Cullen, 1979).

To date, several types of IBD vaccines were imported for use in Malaysia. They

include live attenuated and killed vaccines. The evaluation on the safety and

Page 37: Thesis

37

efficacy of the imported IBD vaccines for local used available in the market

commercially demonstrated that most of the vaccines studied were consider to

be unsafe and not effective to confer full protection against the vvIBDV

challenged. The failure of those IBD vaccines to induce IBD antibody titer had

been previously reported (Hair-Bejo et al., 1995a; 1995b). Despite the

vaccination program adopted, frequent outbreaks of IBD do occur from time to

time. The worst was the emergence of a new highly pathogenic strain of

(vvIBDV) which complicates the immunization programme of the disease.

Differences in the antigenicity between the vaccine and field viruses have been

recognised as one of the major reason for vaccination failure.

This antigenic variation has been reported to be present among the recent field

strains of the virus (Jackwood, 2005) and this could be attributed to the failure of

protection by the existing vaccines.

In the present study, it is believed that attempt to develop local live attenuated

and killed vaccines in tissue culture, has opened great opportunity to a great and

potential for the control of IBDV infection and its associated immune

suppression. The use of local vaccine has helped to control IBD in regional

regions where outbreaks were not controlled by commercially available vaccines

(Hair-Bejo, et al., 1995b).

Page 38: Thesis

38

The objectives of this study were:

1. to adapt and attenuate vvIBDV isolates in tissue cultures for development of

vaccines.

2. to determine the molecular characteristic of the adapted and attenuated

vvIBDV isolate.

3. to determine the pathogenicity and immunogenicity of the attenuated vvIBDV

in SPF chickens

4. to determine the safety and immunogenicity of the inactivated attenuated

vvIBDV in SPF chickens

Page 39: Thesis

CHAPTER 2

LITERATURE REVIEW

2.1 Infectious Bursal Disease

Infectious bursal disease (IBD) is a highly contagious viral disease of young

chickens characterised by destruction of the lymphoid cells in the bursa of

Fabricius. Other lymphoid organs are also affected but to a lesser degree

(Cheville, 1967; Lukert and Saif, 1997). The disease in a fully susceptible

chicken flock, occurs at 3 to 6 weeks of age and the economic impact of the

disease are manifold which includs losses due to morbidity and mortality.

Immunosuppression experienced by the surviving chickens could exacerbate

infections with other disease agents coupled with reduced chicken‟s ability to

respond to vaccination. The economic impact of the disease is influenced by

pathogenicity of the virus, susceptibility of the flock, presence of other prevalent

pathogens, the environment and poor management practices (Saif, 1998).

The causative agent for IBD is a bisegmented, double stranded RNA virus that

belongs to the family Birnavirideae of the genus Avibirnavirus (Dobos et al.,

1979; Muller et al., 1979b). Two distinct serotypes have been recognized.

Pathogenic strains are grouped in serotype 1 viruses while serotype 2 strains

are non-pathogenic.

Page 40: Thesis

40

Until 1987, the virus strains were of low virulence causing less then 2% mortality

and the disease was satisfactorily controlled by vaccination. But in 1986, an

outbreaks of IBD were reported, despite vaccination with a classical strain of

IBD vaccine (Jackwood and Saif, 1987). In 1987, very virulent IBDV (vvIBDV)

was isolated in Holland and Belgium (Chettel et al., 1989; van den Berg, 2000).

The mortality rate associated with vvIBDV infection in 3 to 14 weeks old

replacement pullet had been reported to reach 70% while that of broiler flocks

was 30% mortality (van den Berg and Meulamans, 1991). The pathogenic

disease attributed to this strain had spread worldwide including in Malaysia

(Hair-Bejo, 1992), China (Gaudry, 1993), Indonesia (Rudd et al., 2002), Russia

(Shcherbakova et al., 1998) and Japan (Nunoya et al., 1992). The vvIBDV

strains are characterised by severe damage of the bursa and higher mortality

rate in susceptible flocks. These vvIBDV strains, are antigenically similar to the

classical but can established infection in chicken with antibody levels that are

protective against classical strains. The emergence of the vvIBDV has

complicated the immunization programmes against the disease. Early

vaccination may result in failure due to the interference by the maternally

derived antibody (MDA), while delay may cause field virus infection. Therefore

vvIBDV have become an economically important pathogen in the poultry

industries worldwide (Yamaguchi et al., 1997; Chen et al., 1998; Eterradosi et

al., 1998).

Page 41: Thesis

41

2.1.1 Clinical Signs and Gross Lesions

The incubation period of IBD range from 2-4 days. The infection of susceptible

broiler or layer pullet flocks is characterized by acute onset of depression.

Chickens are disinclined to move and peck at their vents (Cosgrove, 1962). In

acute outbreaks, the chicks appear sleepy and have a reduce food intake.

Terminally, birds may show sternal or lateral recumbency with coarse tremor

(Lasher and Shane, 1994). White or watery diarrhea, solid vent feathers and

vent pecking are seen. The feathers are ruffled, the birds have an unsteady gait

and may become prostrate and trembling prior to death (Cosgrove, 1962;

Chettle et al., 1989; Hair-Bejo, 1993; Lasher and Shane, 1994; Lukert and Saif,

1997).

The short duration of clinical signs and mortality pattern are considered to be of

diagnostic significance in IBD (Lasher and Shane, 1994). Affected flocks

showed depression for 5-7 days during which mortality rises rapidly for the first

two days then declines sharply as clinical normality returns (Parkhurst, 1964).

There is usually 100% morbidity, but the mortality varies depending on the virus

strains.

Clinical signs alone are not sufficient to make a diagnosis, but when combined

with gross lesions, it is possible to arrive at a preliminary diagnosis (Saif, 1998).

Changes in lymphoid organs are typical of the disease. The bursa of Fabricius,

which is the main target of the virus, undergoes major changes beginning at 3

Page 42: Thesis

42

days post infection post. Infection (pi). It increases in size reaching twice the

normal size by 4 days pi followed by atrophy, and reaching one third of its

original weight by 8 days pi (Saif, 1998).

By day 2 or 3 pi, the bursa usually has a gelatinous yellowish transudate

covering the serosal surface. Longitudinal striations became prominent and the

color changed from white to creamy. The transudate disappeared as the bursa

returned to its normal size and the organs turned gray during the period of

atrophy (Lukert and Saif, 2003).

The tissue distribution and severity of lesions is dependent on the subtype and

pathogenicity of the virus (Rosenberger and Cloud., 1986). Infected birds are

dehydrated and have darkened discoloration of pectoral muscles. Hemorrhages

occur in thigh and pectoral muscles and are also reported from the mucosa at

the proventriculus-gizzard junction and on the serosal surface and the bursa

(Hanson, 1962). Extensive hemorrhages could be seen on the entire bursa.

There is increased mucus in the intestine and renal changes are observed in

diseased birds which had been attributed to dehydration (Lukert, and Saif,

2003). The kidneys, tubules and ureters are so distended and filled with urates

that they appeared white (Cosgrove, 1962).

Pathologic changes in the spleen and thymus were less prominent than those of

the bursa (Cosgrove, 1962; Inoue et al., 1994). The spleen might be slightly

enlarged and usually had small gray foci uniformly dispersed on the surface

Page 43: Thesis

43

(Inoue et al., 1994). Lesions in these organs are noticed at the same time as the

changes occurred in the bursa. These lesions resolved within 1 or 2 days of

appearance (Helmboldt and Garner, 1964).

The vvIBDV infections are characterized by severe clinical signs, high mortality,

and a sharp death curve followed by rapid recovery. The vvIBDV strains have

the same clinical signs and incubation period of 4 days as classical viruses

(caIBDV) but the acute phase is exacerbated (van den Berg, 2000). The vvIBDV

strains cause more severe lesions in the cecal tonsils, thymus, spleen and bone

marrow and a greater decrease in thymic weight index as compared to the

(caIBDV) strains but, bursal lesions are similar. It has been shown that the

pathogenicity of field strains of IBDV correlated with lesion production in non-

bursal lymphoid organs. The results also suggest that pathogenicity of IBDV

may be associated with virus antigen distribution in non-bursal lymphoid organs

(Tanimura et al., 1995).

Chickens affected by the variant IBDV (vaIBDV) are characterized by severe

bursal atrophy and immunosuppression (Lukert and Saif, 1997) without showing

the inflammation induced symptoms associated with the infection of caIBDV

(Sharma et al., 1989). Attenuated strains have been adapted to chick embryo

fibroblast (CEF) cells or other cell lines. These strains do not cause disease in

chickens, and therefore some of them are being used as live vaccines (Lim et

al., 1999).

Page 44: Thesis

44

2.1.2 Histopathology

Histolopathologic changes occur in the bursa, spleen, thymus, Harderian gland

and cecal tonsils. The first obvious lesion of infection occurs in the bursa of

Fabricius and it is the most severely affected organ. Degeneration and necrosis

of individual lymphocytes in the medullary region of the bursa occur as early as

1 day post infection. Lymphocyte degeneration is accompanied by nuclear

pyknosis and formation of lipid droplets in the cytoplasm (Cheville, 1967).

Degenerating lymphocytes are surrounded by macrophages. Lymphocytes are

replaced by heterophils, pyknotic debri, and hyperplastic reticuloendothelial

cells.

By 3 or 4 days post infection, all lymphocytes would have been affected. At this

point of time the bursal weight increases due to edema, hyperemia, and

accumulation of heterophils. As the inflammatory reaction subsides, cystic

cavities appear in the medullary region of the bursal follicles. Necrosis and

phagocytosis of the heterophils take place and fibroplasia occurs in the inter-

follicular connective tissue (Helmboldt and Garner, 1964; Cheville, 1967; Lukert

and Saif, 2003). The proliferation of the bursal epithelial layer occurs producing

glandular structures of columnar epithelial cells containing globules of mucin.

Follicular regeneration and repopulation of follicles with the lymphocytes occur

but healthy follicles are not formed during the observed time span of 18 days

(Helmboldt and Garner, 1964).

Page 45: Thesis

45

The spleen shows hyperplasia of the reticuloendothelial cells around the

adenoid sheath arteries during the early stages of infection. Lymphoid necrosis

occurs in the peri-arteriolar lymphoid sheath by 3 days pi. The spleen recovers

shortly without any sustainable damage to the germinal follicles (Cheville, 1967;

Lukert and Saif, 2003).

Changes in thymus and cecal tonsils appear shortly after infection and include

areas of lymphoid necrosis and hyperplasia of the reticular and epithelial

components in the medullary region of thymic follicles (Cheville, 1967). The

damage is less extensive than in the bursa and is quickly repaired by 12 days pi

(Cheville, 1967).

The Harderian gland is reported to be severely affected by the virus in 1 day old

chickens (Survashe et al., 1979). Normally, the gland is populated with plasma

cells as the chicken ages but the infection prevents this infiltration. Harderian

gland of the chickens infected at 1 day of age has 5-10 folds fewer plasma cells

than those of uninfected chickens from 1-7 weeks of age (Dohms et al., 1981).

However, lymphoid follicles and heterophil populations in the Harderian gland

are not affected by IBDV infection, nor could necrotic or degenerative changes

be found in the acini or excretory ducts.

In contrast, the broilers infected at 3 weeks of age have a 51% reduction in

plasma cell content at 5-14 days pi (Dohms et al., 1981). Plasma cell numbers

reduction was temporary and the levels became normal after 14 days. Histologic

Page 46: Thesis

46

lesions appearing in the kidneys were nonspecific and resulted from dehydration

(Helmboldt and Garner, 1964). The liver had some slight perivascular infiltration

of monocytes (Peters, 1967).

2.1.3 Pathogenesis

Pathogenesis is the process through which the virus cause injury to the host

leading to mortality, disease or immunosuppression. The different pathotypes of

IBDV have different degree of pathogenicity, virulence and antigen distribution in

different organs (Lukert and Hitchner, 1984). The natural infection is usually via

the oral route accompanied by the gut associated lymphoid cells (Becht, 1980).

Following oral inoculation of IBDV in susceptible birds, the virus replicate

primarily in the macrophage and lymphoid cells of the gut-associated lymphoid

tissue during 4 to 6 hours pi (Kaufer and Weiss, 1976) and leads to primary

viremia. Then virus travels to liver via portal vein and localized in the bursa of

Fabricius as the target organ via blood stream where IBDV replication occur at

13 hour post inoculation (Muller et al., 1979a). After massive replication in the

follicle of the bursa of Fabricius, the virus will be released into the blood as

secondary viremia. This will be followed by virus replication and destruction to

another organ such as cecal tonsil, spleen, bone marrow, gut associated

lymphoid tissue and also replication in bursa of Fabricius (Muller et al., 1979a;

Becht, 1980). Consequently, clinical sign and mortality occur within 48 to 72

hours (Kaufer and Weiss, 1976). The cause of death in clinical IBD is mainly due

Page 47: Thesis

47

to circulatory failure as a result of severe hemorrhages (Hair-Bejo, 1993).

Severe dehydration owing to diarrhea and reduce water intake could also lead to

circulatory failure and death (Hair-Bejo, 1993).

Haemorrhage in IBDV infected chicken can be due to impairment of the clotting

mechanism due to destruction of thrombocyte (Skeeles et al., 1980) and

depletion of haemolytic component (Skeelas et al., 1980). In addition

haemorrhages can also be the result of formation of immune complexes

culminating to an Arthus reaction.

Microscopic lesion particularly in the bursa of Fabricius is similar to an Arthus

reaction, which is caused by deposition of antigen antibody complement

complexes which in turn induces production of chemotactic factors,

haemorrhages and leukocytes infiltration (Skeeles et al., 1979). Two week old

chicks showed less circulating complement than 8 weeks old chicks and did not

show the Arthus reaction (Skeelas et al., 1979). In addition, IBDV infected

chickens showed prolonged clotting time, which has consequently induced

hemorrhagic lesions in the birds (Skeeles et al., 1979).

The target organ of IBDV is the bursa of Fabricius at its maximum development.

Orally inoculated IBDV in bursectomized and non-bursectomized birds showed

that the replication of the virus occurred in the gut-associated lymphoid tissues

(Muller et al., 1979a; Kaufer and Weiss, 1980) and the second replication, in the

Page 48: Thesis

48

bursa of Fabricius that is responsible for the high titer of the virus and also for

clinical signs and mortality.

IBDV has a predilection for actively dividing immunoglobulin G and M bearing

cells (Hirai et al., 1981). This makes the B lymphocytes to be the main cells

affected by the virus. Since the maturation B lymphocyte occurs in the bursa of

Fabricius, this organ and the lymphocytes are the most affected during infection.

Therefore infected chicken became deficient in the production of optimum levels

of antibodies against divers antigen (Faragher et al., 1974; Giambrone et al.,

1977).

2.1.4 Immunosuppression

IBDV infected chickens are immunosuppressed and susceptible to other avian

pathogens, such as Mycoplasma gallisepticum (Nunoya et al.,1995),

Staphylococcus aureus (Santivatr et al., 1981; McNamee, 2000), Escherichia

coli (Igbokwe et al., 1996), Eimeria tenella (Giambrone et al., 1977; Anderson et

al.,1977), Newcastle disease virus (NDV) (Almassy and Kakuk, 1976; Westbury,

1978; Rosales et al., 1989b), chicken anaemia virus (CAV) (Yuasa et al.,1980;

Rosenberger and Cloud, 1989), reovirus (Moradian et al., 1990), Marek‟s

disease virus (Cho, 1970), infectious laryngotracheitis virus (Rosenberger and

Gelb, 1978), infectious bronchitis virus (IBV) (Winterfield et al., 1978; Pejkovski

et al., 1979), and adenovirus infection (Fadley et al., 1976). Simultaneous

infections by IBV and IBDV usually lead to secondary infection of the respiratory

Page 49: Thesis

49

tract caused by E.coli (Naqi et al., 2001). IBDV-infected chickens also failed to

response to anti-coccidial drug treatments during the coccidiosis outbreak and

this result in high mortality (McDougald et al., 1979).

IBDV induced immunosuppression may be due to the direct destruction of B

lymphocytes (Ramm et al., 1991; Saif, 1998), and possibly the elimination of

crucial elements within the bursal microenvironment (Ramm et al., 1991).

Infection of day-old SPF chicks with a virulent IBDV strain reduced the antibody

production against IBV in tears as well as in serum (Thompson et al., 1997;

Gelb, et al., 1998). Studies showed that immunosuppression caused by IBDV

infection could last for at least up to six weeks of age (Wyeth, 1975; Giambrone,

1979; Lucio and Hitchner, 1980). The most severe and longest-lasting

immunosuppression occurs when day-old chicks are infected with IBDV (Allan et

al., 1972; Faragher et al., 1974; Sharma et al., 1989). Fortunately this is

uncommon in the field because chicks usually have high maternal antibody (van

den Berg, 2000). However, at two to three weeks when maternal antibody

wanes, then the infection is likely to occur (van den Berg, 2000). In situations

where the bursa of Fabricius of young birds were destructed, this has been

shown to affect the effectiveness of the subsequent vaccination programmes

(Giambrone et al., 1976).

Page 50: Thesis

50

2.1.5 Epidemiology of IBD

The actual distribution of IBD around the world is difficult to ascertain because of

the subclinical nature of the disease. The first outbreak due to the classical IBDV

(caIBDV) occurred in 1957 in the US town of Gumboro and was initially

described as avian nephrosis (Cosgrove, 1962). It was characterized by flock

morbidity of 10-25% and mortality averaging 5% (Lasher and Shane, 1994). The

disease later discovered in 1971 in India (Mohanty et al., 1971), 1973 in Japan

(Hirai et al., 1974), 1974 in Australia, (Firth, 1974), and 1974 in United Kingdom

(Edwards, 1981). The prevalence of clinical IBD was reduced following the

introduction of live vaccines from 1966 onwards (Edgar and Cho, 1965).

In 1983, antigenic variant IBDV (vaIBDV) was reported in the USA (Jackwood

and Sommer, 1999), in China (Cao et al., 1998) and in Australia (Sapats and

Ignjatovic, 2000). Chickens vaccinated with caIBDV vaccines were not protected

against these new „variant strains‟ and they succumbed to immunosuppressive

form of the disease (Ture et al., 1993; Vakhaira et al., 1994).

The vvIBDV strains, a newly evolved strain associated with very high mortality

were first observed in Europe in the late 1980s (Chettle et al., 1989; van den

Berg et al., 1991). To date, vvIBDV infections have been documented in Europe

(Chettle et al., 1989; Pitcovski et al., 1998), Asia (Japan) (Nunoya et al., 1992;

Lin et al., 1993), China (Cao et al, 1998), Malaysia (Hair-Bejo, 1992) and Africa

Page 51: Thesis

51

(Zierenberg et al., 2000). Until now, none of vvIBDV was reported in United

States, Australia, Canada and New Zealand (Sapat and Ignijatovic, 2000).

It was hypothesized that the initial outbreaks of IBD in the USA arose by

mutation of an Aquabirnavirus such as infectious pancreatic necrosis virus

(IPNV) (Lasher and Shane, 1994), there is no published evidence that IBDV

serotype 1 existed in turkey flocks prior to 2003, although an earlier report

suggested that turkeys might be infected with IBDV serotype 1 and 2 (McNulty

et al., 1979). The latest report by Owoade et al (2004) showed that turkeys

should be considered to be susceptible to vvIBDV infection.

2.1.6 Transmission

IBD has been an economically significant, widely distributed condition affecting

flocks of chickens. The causal virus is transmitted laterally by direct and indirect

contact between infected and susceptible flocks (Lasher and Shane, 1994), but

not transmitted vertically by transovarian route (Lukert and Saif, 1997). Indirect

transmission of virus most probably occurs on fomites (feed, clothing and litter)

or through air (Benton et al., 1967). There is no evidence of egg transmission of

the virus and no carrier state has been detected in chickens (Saif, 1998).

Infected chickens shed IBDV at one day after infection and can transmit the

virus for at least 14 days (Vindervogel et al., 1976), but not exceeding 16 days

(Benton et al., 1967; Winterfield et al., 1972; Lasher and Shane, 1994).

Page 52: Thesis

52

Operation of multi-age broiler and pullet replacement farms, defects in

biosecurity or proximity of farms to road used to transport poultry may contribute

to high prevalence of infection (Lasher and Shane, 1994). The virus can remain

viable for up to 60 days in poultry house litter (Vinervogel et al., 1976). In

addition, rodent, wild birds and insects including mites may be playing an

important role in transmission of IBDV (Brady, 1970). Beside, the lesser meal

worm was recognized as a carrier and the virus has been isolated from

mosquitoes and evidence of infection in rats has been reported but there is no

indication that either species is a reservoir for the virus (Saif, 1998). In contrast

Pages-Mante, et al (2004) show that the possibility that dog could eventually be

carrier of IBDV after eating infected chicken either voluntarily or accidentally.

2.2 Infectious Bursal Disease Virus

The etiological agent of the disease is infectious bursal disease virus (IBDV)

belonging to the family Birnaviridae of the genus Avibirnavirus. The genus name

Birnavirus was proposed to describe viruses with 2 segments of double stranded

RNA. Other viruses included in this group are infectious pancreatic necrotic virus

(IPNV) of fish, tellina virus, oyster virus, blotched snakehead virus (BSVN) (Da

Costa et al., 2003) and crab virus of bivalve mollusks belonging to

Aquabirnavirus while Drosophila X virus belongs to genus Entomobirnavirus. All

of these contain two segments of double stranded RNA surrounded by a single

protein capsid of icosahedral symmetry (Dobos et al., 1979)

Page 53: Thesis

53

2.2.1 IBDV Genome

IBDV contains a genome composed of two segments and double stranded RNA

(dsRNA), designated A and B (Dobose et al., 1979; Mundt and Muller, 1995).

The dsRNA genome is enclosed within a non-enveloped icosahedral capsid

approximately 60nm in diameter (Mundt and Muller, 1995). The larger segment

A (3.4kb) contain two open reading frames (ORFs) of 3,039 pb and 438 pb,

which partially overlap at 5‟ end of the genome (Bayliss et al., 1990; Mundt et

al., 1995). The larger ORF encodes a 110KDa precursor polyprotein (NH2-VP2-

VP4-VP3-COOH) which is autocatalytically cleaved by cis-acting viral protein

VP4 into three proteins designated precursor VP2(pVP2)(48kDa), VP3(23KDa).,

and VP4 (28KDa) (Sanchez and Rodriguez 1999; Lejal et al., 2000; Birgham et

al., 2002). The pVP2 is further processed into VP2 (38KDa) during maturation of

the viral particle (Sanchez and Rodriguez., 1999; Lejal et al., 2000; Birgham et

al., 2002). VP2 the major structural protein of the viral capsid, carries highly

conformational epitopes responsible for the induction of neutralising antibodies

that confer protective immunity (Azad et al., 1987; Becht et al., 1988; Jagadish

et al., 1988). VP3 is the second structural protein of the viral capsid, recognized

by non-neutralising antibodies that often cross-react with both serotypes

(Hudson et al., 1986; Bottcher et al., 1997). The smaller ORF of segment A

encodes VP5 (17KDa), a 145 amino acid non structural protein of unknown

function (Mundt et al., 1995). VP5 has been shown for viral replication and

infection, but plays an important role in the release of viral progeny from infected

cells which are important for its pathogenicity (Mundt et al., 1995; Mundt et al.,

Page 54: Thesis

54

1997). The smaller segment B (2.8kb) encodes VP1 (90KDa), RNA-dependent

RNA polymerase (RdRp) with capping enzyme activities (Mundt et al., 1995).

Several attempt to elucidate the residues responsible for the pathogenicity of

IBDV has identified conserved amino acid substitutions throughout both genome

segments (Lejal et al., 2000). Development and application of reverse system

for IBDV has shown that neither the non-coding regions (NCRs), nor residue

within VP1 or the N terminus of VP2 is responsible to increase pathogenicity of

IBDV (Mundt and Vakharia, 1996; Yao et al., 1998). These results suggest that

virulence determinants reside within the VP2, VP4 and/or VP3 proteins.

Comparison of the deduced amino acid sequence of the large segment of IBDV

strains showed that the most amino acid change occurs in the central

hypervariable region between residues 206 and 350 of VP2 protein (Bayliss et

al., 1990). VP2 has been shown to be the variable region which encodes the

neutralisation antigenic epitope (Chen et al., 1998). This region is highly

conformation dependent, and it is constituted by hydrophobic fragment flanked

by hydrophilic peaks (Fahey et al., 1989; Fahey et al., 1991; van den Berg et al.,

1996). VP2 of the virus is shown to be responsible for increased apoptosis in a

variety of different mammalian cell lines (Fernendez Aries et al., 1997).

Page 55: Thesis

55

2.2.2 IBDV Proteins

Five viral proteins have been described in the IBDV virion namely VP1, VP2,

VP3, VP4 and VP5 (Nick et al., 1976). VP1, VP4 and VP5 are non-structural

viral proteins whereas VP2 and VP3 are structural viral proteins (Mundt et al.,

1995; Nagarajan and Kibenge, 1997).

VP1, a RNA dependent RNA polymerase of the IBDV, is present in small

amounts (3%) in the virion. It is 90 kDa in molecular weight (Lasher and Shane,

1994). VP1 is both a free polypeptide and a genome-linked protein (Muller and

Nitschke, 1987; Kibenge and Dharma, 1997). It plays a key role in the

encapsidation of the viral particles (Lombardo et al., 1999).

VP2 is a 454 amino acid long polypeptide that builds up the external virus capsid

(Kibenge et al., 1988). Expression or deletion studies have shown VP2 amino

acid positions 206 to 350 to represent a major conformational, neutralizing

antigenic domain (Azad et al., 1987). Most amino acid changes between IBDV

strains are clustered in this region, thus referred to as VP2 variable domain

(Bayliss et al., 1990). This domain is composed of hydrophobic amino acid

flanked by two hydrophilic peaks A and B, which span amino acid 210 to 225

and amino acid 312 to 324, respectively (Azad et al., 1987). Variations in IBDV

antigenicity have been shown to depend on changes in peaks A and B. Two

smaller hydrophilic areas of VP2 variable domain, amino acid 248 to 252 and

279 to 290 were recently reported to also influence IBDV antigenicity (van den

Page 56: Thesis

56

Berg et al., 1996). Only two mutations of the VP2 (Q253H and A284T) are

enough to attenuate a vvIBDV strains (UK661 isolate) and enabling it to grow in

cell culture (van Loon et al., 2002). VP2 is an important IBDV structure protein

as the antigenic site that is responsible for the induction of neutralizing

antibodies are centrally located on VP2 gene (Fahey et al., 1989; Becht et al.,

1988). Monoclonal antibodies (MAbs) had been successfully raised against VP2

and VP3 but only those reacting to VP2 have the ability to neutralize the virus

(Azad et al., 1987; Becht et al., 1988; Snyder et al., 1988). Thus, it was

suggested that the hyper variable region of the VP2 gene is responsible for the

virus antigenicity and the induction of host neutralizing antibodies (Schnitzler et

al., 1993). VP2 is also an apoptotic inducer where its expression in various

mammalian cell lines leads to apoptosis (Fernandez-Arias et al., 1997). In vivo

studies and molecular characterization suggest that some of the VP2 residues

may play a role in molecular determinants for the virulence, cell tropism and

pathogenic phenotype of vvIBDV (Brandt et al., 2001).

VP3 is a group specific antigen which is recognized by non-neutralising

antibodies. VP3 is 40% of the complete virion protein with 32 kDa molecular

weight (Becht et al., 1988; Oppling et al., 1991). It is responsible for the

structural integrity of the virion and has been identified as a major antigenic

component of the virus (Fahey et al., 1985). VP3 reacts with serotypes1 and 2

and perform as an intermediate, which interacts with both the VP2 and VP1, and

the formation of VP1-VP2 complexes is likely to be an important step in the

morphogenesis of IBDV particles (Lombardo et al., 1999).

Page 57: Thesis

57

VP4 is fourth viral protein with 28 kDa molecular weight. It is a non-structural

polypeptide, representing 6% of the viral protein. VP4 is involved in the

autoprocessing of the virus polyprotein producing VPa, VP3 and VP4 (Lasher

and Shane, 1994; van den Berg, 2000). The amino acids for this proteolytic

activity have been identified to be a serine lysine catalytic dyad (S652 and K

692) (Lejal et al., 2000).

VP5 is also nonstructural IBDV protein that has been identified in IBDV infected

cells. The VP5 is located at the second ORF on the segment A of the IBDV

genome which encodes polyprotein of 21 kDa molecular weight. This

polypeptide more probably has a regulatory function and may play a key role in

virus release and dissemination (Mundt et al., 1995; Lombardo et al., 2000).

2.2.3 Antigenic and Virulence Variation

IBDV is endemic throughout the world but several different antigenic and

pathogenic types exist in specific geographic locations. Two serotypes of IBDV

are recognized by the virus neutralization test. These two serotypes are

antigenically distinct (Mcferran et al., 1980). Serotype 1 viruses are pathogenic

to chickens and differ in their virulence (Winterfield et al., 1978). They cause

lesions in the bursa of Fabricius by lymphocytic depletion (Schroder et al., 2000)

whereas, serotype 2 viruses are avirulent to chickens and are isolated mainly

from turkeys (Ismail et al., 1988; Kibenge et al., 1991).

Page 58: Thesis

58

Serotype 1 viruses can be broadly divided into classic (ca), variant (va) and very

virulent (vv) IBDVs. Until 1987, the strains of virus were of low virulence and

were controlled by vaccination. Emergence of variant viruses was first reported

in USA in 1987. These viruses were reported to undergo an antigenic drift

against which the classical IBD vaccines were not protective (Jackwood and

Saif, 1987; Snyder et al., 1992).

Six antigenic subtypes of IBDV serotype 1 viruses have been identified by the

virus neutralization test (Jackwood and Saif, 1987). Variant viruses that were

found in the USA and Australia are different from the classic viruses in terms of

pathogenicity and immunogenicity. They overcome the immunity induced by

classic serotype 1 viruses and cause rapid bursal atrophy with minimal or no

inflammatory response (Mcferran et al., 1980; Jackwood and Saif, 1987; Hassan

and Saif, 1996).

Vaccination with one serotype 1 subtype did not ensure protection from

challenge with another subtype suggesting that variant viruses are antigenically

different from classical viruses (Mcferran et al., 1980; Jackwood and Saif, 1987;

Ismail and Saif, 1991; Hassan and Saif, 1996). Variant viruses present in the

USA and Australia are not closely related to each other (Sapats and Ignjatovic,

2000). Significant antigenic differences exist among serotype 1 strains as

detected by virus neutralization and this led to the grouping of the serotype 1

viruses into 6 subtypes (Hassan and Saif, 1996) hence virus neutralization test

Page 59: Thesis

59

proved to be serotype specific and could distinguish between the two serotypes

(Jackwood et al., 1982; Jackwood et al., 1985; Hassan and Saif 1996).

Serotype 2 viruses are immunologically distinct from serotype 1 viruses since

vaccination with serotype 2 (OH) viruses did not confer protection against

serotype 1. Cross protection studies indicated that the variant viruses were

different from other subtypes of serotype 1 IBDVs. Both serotype 1 and 2

viruses share common group antigens which could be detected by (AGPT),

flourescent antibody test and ELISA (Jackwood et al., 1982; Jackwood et al.,

1985; Jackwood and Saif 1987; Chettle et al., 1989). Capsid proteins VP2 and

VP3 contain epitopes that are responsible for group antigenicity (Becht et al.,

1988). The VP2 carries the serotype specific antigens responsible for the

induction of neutralizing protective antibodies (Azad et al., 1987; Becht et al.,

1988).

VP2 is the major host-protective immunogen of IBDV and it contains the

determinants responsible for antigenic variation (Fahey et al., 1989; Brown et

al., 1994; Vakharia et al., 1994). The antigenic site which is responsible for the

induction of neutralizing antibodies against IBDV, are centrally located on VP2

gene and known as hypervariable region (Azad et al., 1987; Becht et al., 1988).

The sequences of the major host-protective immunogen VP2 are highly

conserved except the central Accl-Spel (206-350) restriction fragment of

hypervariable region (Heine et al., 1991; Brown and Skinner, 1996).

Representing only 16% of segment A, this region displays the greatest amount

Page 60: Thesis

60

of amino acid sequence variations between the pathogenic serotype 1 strains

(Becht, 1980; Kibenge et al., 1990).

The HPVR encodes for the immunodominant viral epitopes (Becht et al., 1988;

Fahey et al., 1989) or neutralizing antigenic epitopes. There are at least three

distinct, non-overlapping and conformation-dependent epitopes (Azad et al.,

1987; Becht et al., 1988; Oppling et al., 1991). These epitopes are located at the

central variable region of the VP2 gene and is comprised of 145 amino acids

from amino acids 206-350. Within this region, there are two hydrophilic peaks

which is highly conformation dependent (Fahey et al., 1991; van den Berg et al.,

1991).

The first peak is from amino acid 212 to 224 where as the second peak is from

amino acids 314 to 324 (Fahey et al., 1989; Bayliss et al., 1990; Heine et al.,

1991; Schnitzler et al., 1993; Brown et al., 1994). Within the first hydrophilic

peak of the hydrophilic region, position 222 appears to play a crucial role in

epitope formation. In classical virus strain, proline (P) is found at position 222,

while glutamine (Q), therionine (T) or serine (S) are found in variant strains and

alanine (A) is found in vv strains (Vakharia et al., 1994; Dormitorio et al., 1997).

Minor mutations in the hydrophilic peaks can result in antigenic drift (Schnitzler

et al., 1993). The amino acid residue changes at the P222A (proline to alanine),

V256I (valine to isoleucine) and L294I (leucine to isoleucine) can be used as a

marker for vvIBDV, G254S (glycine to serine) and Q249K (glutamine to lysine)

for variant strains whereas, amino acid changes at D279N (aspartic acid to

Page 61: Thesis

61

aspargines) and A248T (alanine to theronine) are common among attenuated

strains (Yamaguchi et al., 1996b; Cao et al., 1998). The hydrophilic regions are

thought to play an important role for the formation and stabilization of the virus

neutralizing epitopes (Heine et al., 1991; Schnitzler et al., 1993; Vakharia et al.,

1994).

In addition, specific amino acid changes do occur within the HPVR, an adjacent

downstream serine-rich heptapeptide sequence (SWSASGS) which are located

after the second hydrophilic region, amino acid residue 326 to 332 have been

proposed as potential sites responsible for virus attenuation (Heine et al., 1991;

Vakharia et al., 1994; Yamaguchi et al., 1996b; Dormitorio et al., 1997) or

antigenic determinants associated with the virulence of IBDV (Brown et al.,

1994).

2.3 Isolation, Adaptation and Attenuation of IBD Virus

2.3.1 Chicken Embryos

Initially, most workers had difficulty in isolating of the virus in chicken embryos.

Landgraf et al. (1967) reported a typical experience using the allantoic sac route

of inoculation. Hitchner (1970) demonstrated that chorioallantoic membrane

(CAM) of 9-11 days old embryos was the most sensitive route of isolation of

IBDV. Hitchner (1970) observed that most mortality occurred between the 3rd

and 5th days post inoculation as affected embryos had edematous distention of

Page 62: Thesis

62

the abdomen, petechiae and congestion of the skin and occasionally echymotic

hemorrhages in the toe joints and cerebrum.

2.3.2 Cell Culture

Many strains of IBDV have been adapted to primary cell culture of chicken

embryo origin and cytopathic effects have been observed. These cells include

chicken embryo kidney (CEK), chicken embryo bursa (CEB) and chicken

embryo fibroblast (CEF) cells (Lukert and Davis, 1974; McNulty et al., 1979).

Cell culture adapted IBDV grows in several mammalian continuous cell lines

such as RK-13 derived from rabbit kidney (Rinaldi et al, 1972), Vero cells

derived from adult African green monkey (Leonard, 1974; Lukert et al., 1975;

Jackwood et al., 1987), BGM-70 cells derived from baby grivet monkey kidney

and MA-104 cells derived from rhesus monkey kidney (Jackwood et al., 1987).

Continuous cell lines has been found to yield higher virus titers compared to

primary cell culture, thus are more suitable to use for vaccine production. Three

strains of serotype1 IBDV (SAL, D78, 2512), one of the serotype 2 (OH) and one

vaIBDV strain (Variant A) were grown in Vero and (CEF) cell culture. The latent

period in Vero cells ranged from 12-18 hours, which has longer than 4-6 hours

period observed in CEF cultures from strains SAL, D78 and OH. There was

more extensive maturation phase and higher yield of virus in Vero cells than in

CEF cultures. Total titers of theses viruses of 5.35 to 6.10 log10 TCID50/ mL in

CEF occurred 24-40 hours post infection (pi) although the CPEs were not seen

Page 63: Thesis

63

until 72 hours pi. By comparison, their total infectious virus titers of 6.85 to 8.35

log10 TCID50/mL in Vero cell occurred from 48 hours pi coinciding with

appearance of CPEs. The growth curve of variant A in Vero cells differed from

other viruses by showing steadily extracellular and cell associated virus titer

throughout the 72 hours observation period. Only very low titers of variant A

were obtained in CEF cultures and no growth curve in CEF was reported

(Kibenge et al., 1988).

Vero cell line was found to be more susceptible than ovine kidney (OK) cell line

for IBDV. Kibenge et al., (1992) used OK cell line, Vero cell line and CEF

culture to attempt IBDV isolation from 26 suspected samples. Virus was isolated

from 2 of 26, 3 of 26 and 3 of 25 samples on OK, Vero and CEF cultures,

respectively. However, in contrast to IBDV replication in Vero and CEF, isolated

virus was unable to induce serially sustained CPEs during successive passages

in OK cell line. The cytopathogenicity of chloroform un-treated virus passages

on OK cells was revived and maintained upon passages in Vero cells (Kibenge

et al., 1992). An initial single passage of suspected field material in OK cells

followed by further passages in Vero cells resulted in virus isolation from 6 of 26

samples which was a better recovery than when either cell line was used alone

or when CEF culture was used. Twenty of twenty six samples were originally

positive when examined by nucleic acid hybridization with radio-labeled IBDV-

cDNA, indicating that some of the samples that were negative upon virus

isolation using OK and Vero cells may have contained inactivated virus. When

two variant strains of IBDV, IN and E were serially passaged in BGM-70 cell line

Page 64: Thesis

64

for 30 times and 40 times respectively, it resulted in loss of pathogenicity.

However, both viruses maintained their antigenicity and immunogenicity as

demonstrated by immunofluorescence and virus neutralization tests. When

inactivated preparation of both passaged viruses was inoculated in SPF chicken,

satisfactory protection was obtained (Tsai and Saif, 1992).

A variant IBDV strain 977 was passaged in cell culture, plaque purified and

attenuated by serial passages at a high multiplicity of infection in CEF. Cell

culture passaged virus caused less bursal atrophy and splenomegaly than did

the original isolate and retained immunogenicity (Bayyari et al., 1996).

Mohamed et al. (1996b) investigated the pathogenicity of bursa derived and

tissue culture attenuated classic (STC) and variant (IN) serotype 1 strains of

IBDV. The IN bursa derived virus caused bursal necrosis and atrophy earlier

than bursa derived STC virus. Both viruses lost their pathogenicity after four

passages in BGM-70. A statistically significant level (p<0.05) of protection was

observed in SPF chicken vaccinated with the attenuated IN virus used as a live

or inactivated vaccine followed by homologous (STC) challenged with bursa

derived virus (Hassan et al., 1996; Mohamed et al.,1996a).

Mohamed et al. (1996a) also investigated the effect of host system on the

pathogenicity, immunogenicity and antigenicity of IBDV. One classic (SAL) and

one variant (IN) strain of IBDV were passaged separately six times in three host

systems BGM-70 continuous cell line, CEF and embryonated chicken eggs.

Passages in BGM-70 cells or CEF resulted in loss of pathogenicity but virus

Page 65: Thesis

65

passaged in embryos maintained its pathogenicity (Mohamed et al., 1996a).

Although the CEF and Vero cells infected with IBDV exhibited the biochemical

features of apoptosis, agarose gel electrophoresis of DNA extracted from IBDV

infected cells revealed the characteristic laddering pattern of DNA fragmentation

which was more intense in infected CEF than Vero cells. The appearance of

apoptotic nucleosomal DNA fragments in IBDV infected CEF was independent

of virus replication and occurred at an early stage following an in vitro infection

(Tham and Moon, 1996).

Highly virulent the vvIBDV strains were adapted through serial passages in

embryonated eggs. The embryonated egg-adapted vvIBDV was successfully

adapted to grow CEF with CPEs. The embryonated egg and cell culture adapted

virus strains had significantly reduced pathogenicity and did not kill any young

chicken in experimental infection. The bursal lesions of the adapted strain-

infected chicken were similar to those observed in classic strain-infected

chicken. Cross virus neutralization analysis showed antigenic diversity between

the cell culture adapted vvIBDV and classical strains. Immunization with adapted

strains in chicken showed good protection against the infection of vvIBDV,

especially, in case of 3 days post-immunization challenged hence adapted virus

strains showed effective immunogenicity hence they appeared to provide a new

and effective live vaccine against vvIBDV (Yamaguchi et al., 1996b).

Yamaguchi et al. (1996b) studied the changes in the virus population during

serial passage in chicken and chicken embryo fibroblast cells. Two attenuated

Page 66: Thesis

66

infectious bursal disease virus used as commercial live vaccine were passage

five successive times in SPF chicken and CEF cell. Both attenuated strains

increased in virulence during the passage in susceptible chicken as evidenced

by the decrease in bursa to body weight ratio. A direct nucleotide sequence

analysis of the VP2 hypervariable domain amplified by RT-PCR revealed that

the nucleotide at position 890 (T) in both strains was (A) after the passage in

chicken. In addition, the nucleotide at position 890 (A) was T or C after the

subsequent passage in CEF cells. Because of the nucleotide differences, the

amino acid residue at position 235 (His) in both vaccines was Gln after the

passage in chicken, and the amino acid residue Gln was changed back to His

during subsequent passage in CEF cells. The digestion of the amplified

fragment with restriction endonuclease Stu 1 and Neo 1 which recognize the

sequence difference at position 890, showed the population of the virus that had

amino acid Gln at position 253 was gradually increase during the passage in

chicken. The population of the virus that had amino acid His at position 253 was

gradually increased during the subsequent passage in CEF cells.

2.4 General Information on the Immune System

The immune system is an important part of any live entity, protecting the host

from infections existing in the environment such as viruses, bacteria and

parasites and from other non-infectious foreign substance such as protein and

polysaccharide (Abbas, et al 2001; Calder and Kew, 2002). Bone marrow, lymph

nodes, spleen, and the thymus are essential elements of the immune response

Page 67: Thesis

67

of chicken to microorganism. The first is the innate (or natural) immunity and the

second is the adaptive (specific, acquired) immunity (Abbas et al., 2001).

2.4.1 Innate Immunity

The innate immune system is the initial level of immune response that combats

infections. Its properties are defined in the germ line. Innate immunity has no

memory property. It consists of anatomic, physiologic and phagocytic / endocytic

barriers and chemical protection such as gastric acid (Medzhitov and Janeway,

1997). These anatomic barriers are the first line of defence against invaders.

They include the skin and mucous membranes. Physiological barriers in innate

response, such as pH, temperature and oxygen tension limit microbial growth.

Phagocytic cells are critical in the defence against pathogens. Some primary cell

of the innate immunity system include phagocytic / endocytic barriers such as

(heterophils), monocytes and phagocytic macrophages. These cells have

specific receptors associated with common bacterial molecules. Monocytes and

lymphocytes can create and secrete cytokines which are non-immunoglobulin

Polypeptides, in response to interaction with a specific antigen (Ag), a non-

specific Ag or a non-specific soluble stimulus. Cytokines affect the magnitude of

inflammatory or immune responses. They regulate other cells of the immune

system. The secretion of cytokines may be triggered by the interaction of a

lymphocyte with its specific Ag but cytokines are not Ag-specific. Thus, they

bridge innate and adaptive immunities. Macrophages are important phagocytic

cells that participate in non specific and specific immunity. They can destroy

Page 68: Thesis

68

infected cells and ingested microbes and support other cells of the immune

system to generate an immune response (Abbas et al., 2001).

2.4.2 Adaptive Immunity

When the innate immune system cannot handle and destroy the encountered

pathogen, adaptive immunity is the next line of defence in its support. Acquired

immunity is very specific and has an immunologic memory. The immunologic

memory allows this specific immunity to remember the molecular features of a

pathogen that has been previously encountered and handled. Adaptive immunity

includes both humoral and cell-mediated immune response (Abbas et al., 2001).

2.4.3 Humoral (B cell-mediated) Immunity

Humoral immunity can combat certain infections through circulating antibodies

such as immunoglobulin (Ig) (Devereux, 2002). The antibodies are generated as

soon as a germ is encountered and remain in the immune system.

Immunoglobulin molecules are the cell surface receptor of B-lymphocytes

derived from the bursa of Fabricius in chicken. Antibodies in birds fall into three

major categories: IgM, IgG (also called IgY) and IgA. It has been observed that

mature B-cells, which have a single antigen specificity, travel towards different

lymphoid organs in order to properly interact with an antigen (Abbas et al.,

2001). The antibodies produced are usually incapable of struggling against

Page 69: Thesis

69

viruses and some types of bacteria intracellularly. However, they are powerful at

destroying extracellular pathogens.

2.4.4 Cell-mediated (T-cell mediated) Immunity

Cell- mediated immune response becomes active when the humoral immune

response is not capable of eliminating the antigen (Erf, 2004). T-lymphocytes

play an important role in the cell-mediated immune system and are capable of

handling and mitigating the risk of intracellular pathogens (Chen et al., 1991;

Devereux, 2002).

T-cell can recognize antigens through the T-cell receptor (TCR) and other

accessory adhesion molecules. All T-cells express the CD3 complex but T-cell

has discrete subpopulation, thus distinguishing them as cytotoxic or regulatory

T-cells. Cytotoxic cells eliminate mostly virus-infected and tumor cells, they are

inclined to express the CD8 complex, a specific molecule on their surface (Chan

et al., 1988; Janeway et al., 2001). Regulatory T-cells, also called T- helper cells

(Th) express the CD4 cell-surface molecules and play a major role in the

immune system (Astile et al., 1994). Such cells produce cytokines that are

needed for T- and B- cells to become active (Chan et al., 1988; Janeway et al.,

2001). These cytokines are capable of activating component of non-specific

immunity and thus enhance better functioning of the immune system. The Th-

cells are subdivided into type-1 T-helper cells (Th1) and type-2 T-helper cells

(Th2). The classification of regulatory T-cells is based on the profile of cytokines

Page 70: Thesis

70

produce and their function (Bottomly, 1988). Th1-cells an important role in cell

mediated immune response while Th2-cells participate in the induction of a

strong humoral immune response (Constant and Bottomly, 1997).

2.4.5 Relationship Between B- and T-cells

B-cells do not need antigen-presenting cells, because B-cells can bind directly

with antigens. However, they do need cytokines created by Th cells in order to

be completely active and become antibody-producing plasma cells (T-

dependent response). Consequently B-cells obtain support from Th-cells.

Nevertheless, it is known that there are certain antigens, such as T- independent

antigens, that activate B-cells irrespective of Th- cells (Abbas et al., 2001).

2.4.6 Effect of IBDV on Innate Immunity

IBDV modulates macrophage functions. There is indirect evidence that the in

vitro phagocytic activity of these cells may be compromised (Lam, 1998).

Macrophages are important cells in the immune system and the altered

functions of these cells may influence normal immune responsiveness in birds.

2.4.7 Effect of IBDV on Humoral Immunity

IBDV has an affinity for the immature B lymphocytes (Sivanandan and

Maheswaran, 1980) and actively dividing B lymphocytes thereby causing a

Page 71: Thesis

71

complete lysis of IgM bearing B cells which in turn result in the decrease in

circulating IgM

cells. Infected chicken produces less level of antibodies against

the antigen (Kim et al., 1999). Only primary antibody responses are affected.

Secondary responses remain unaltered (Rosenberger et al 1994; Sharma et al.,

1989). IBDV induced humoral deficiency is reversible and overlaps with the

restoration of bursal morphology (Sharma et al., 2000).

Chickens infected with IBDV at 1 day of age were found to be completely

deficient in serum IgG and produced only a monomeric immunoglobulin M (IgM)

(Ivanyi, 1975). IgG levels varied depending on the age at the time of infection

(Hirai et al., 1981). The number of B cells in peripheral blood was reduced after

infection with IBDV, but T cells were not appreciably affected (Hirai et al., 1981;

Sivanandan and Maheswaran, 1980). The adverse effect on antibody responses

is due to the damage to the B cells in the bursa and the blood since the virus

has a predilection for actively dividing B cells as compared to the mature B cells

(Sivanandan and Maheswaran, 1980).

2.4.8 Effect of IBDV on Cellular Immunity

T-cells in spleen and peripheral circulation are affected during IBDV infection

(Confer et al., 1981; Sivanandan and Maheswaran, 1980; Kim et al., 1999). The

mitogenic inhibition of T cells occurred early, during the first 3 to 5 days of virus

exposure but later returned to normal levels. During the period of mitogenic

inhibition, T cells of IBDV infected chickens also failed to secrete IL-2 upon in

Page 72: Thesis

72

vitro stimulation with mitogens (Kim et al., 1999; Sharma and Fredericksen,

1987).

2.5 Vaccination

IBDV is highly infectious, very resistant in the environment and can persist in the

poultry houses after cleaning and disinfection. The virus is also resistant to ether

and chloroform. It is inactivated at pH 12.0 but unaffected at pH 2.0.

Consequently the virus can persist in the chicken houses for long periods

(Benton et al., 1967). Therefore, hygienic measures alone are not enough to

control this disease and vaccination is the principle method used for the control

of IBD in chicken (Kibenge et al., 1988).

The most common strategy followed to control IBD is by achieving passive

and/or active immunity in chickens (van den Berg, 2000). Passive immunity is

referred to the transfer of IBDV specific, neutralizing antibodies from

hyperimmunized parent flocks to their progeny (Sharma and Rosenberger,

1978). These maternally derived antibodies protect baby chick from early

immunosuppressive effect caused by IBDV. Passive immunity conferred to

progeny chicks normally last up to 21 days of age approximately. However, the

vaccination of parent breeders with an inactivated IBDV oil-emulsified vaccine

extends the range of maternal antibody protection up to 30-38 days of age

(Lucio and Hitchner, 1979; Baxendale and Lutticken, 1981; Lukert and Saif,

1997). Attempts have been made to confer passive protection by performing

Page 73: Thesis

73

parenteral inoculation of IBDV specific immunoglobulins in chicks of 1 day of

age (Lucio et al., 1996). However, these approaches are not routinely used in

the field.

Active immunity is accomplished when doing vaccination of broiler breeder and

layer flocks with live and/or inactivated oil-emulsified vaccines. Generally, live

vaccines are used to prime the immune system so that an IBDV specific

antibody response is induced. In contrast, killed vaccines are used to boost the

active immunity developed in chicken (Lasher and Shane, 1994; Lukert and Saif,

1997).

2.5.1 Live Virus Vaccines

Live virus vaccines are generally derived from the serial passages in

embryonated eggs or tissue culture (van den Berg, 2000). The degree of

attenuation of the vaccine strains can be classified as mild, intermediate and

hot, depending on the its ability to cause the varying degree of histological

lesions (Office International des Epizooties, 2000). Although serotype 1 vaccine

strains cause no mortality, it is still causing different degrees of bursal lesions

that range from mild to moderate or even severe (van den Berg, 2000). The

higher the virulence of the vaccine virus strain, the more the damage that is

observed in the bursa of the vaccinated chicken (Kelemen et al., 2000).

Nonetheless, as it should be, the lesion caused by the vaccine strain is less

severe than the field strain (Rosales et al., 1989b).

Page 74: Thesis

74

The mild strain is mainly used in the breeder vaccination programme.

Vaccination with the mild strain is usually affected by maternal antibody

interference, therefore, such vaccine is usually used between the fourth and

eight week of age, depending on whether the grandparent birds have or have

not been vaccinated with oil-emulsion inactivated vaccine before lay (van den

Berg, 2000). Intermediate vaccines are used for broiler and pullet vaccination

(Mazariegos et al., 1990), and sometimes given to breeder chicks when the

flocks are at risk of early challenge of highly pathogenic strains. Day-old

vaccination using intermediate vaccine may protect the chicks that have

insufficient maternal antibody (van den Berg, 2000). Besides, early vaccination

will spread the vaccine virus in the farm premises and provides indirect

vaccination to the other susceptible chicks (van den Berg, 2000). In high-risks

farms, two vaccinations are generally practice. The time of vaccination depends

on the flocks‟ maternal antibody titres. Route of vaccination is usually through

drinking water, although nebulisation could also be used (van den Berg, 2000).

To achieve higher maternal antibody in the progeny, vaccination of broiler

breeders with live IBD vaccine is common (Wyeth, 1980). Meanwhile,

vaccination of parent chickens with a commercial live IBD vaccine under field

conditions at varying ages and by different routes may result in the variable

susceptibility to the disease in their chicks (Wyeth et al., 1992).

One of the major problems in the use of live IBDV vaccines is optimizing the

time for immunizing chicken flocks. Timing of these vaccines usually depends

upon the level for maternal antibodies circulating in serum as determined by

Page 75: Thesis

75

serology (ELISA), the route of vaccine administration and pathogenicity of the

vaccine virus to be used. Thus, development of an appropriate vaccination

program is very difficult due to difference in the protecting maternal antibody

levels seen in progeny form different breeder flocks. The myriad of antibody titer

within parent flocks induce a wide variability of antibody levels in progeny. In

consequence, some chicks may be refractory to vaccination for up to 4 weeks of

age, while other may be susceptible to IBDV and ready to be immunized within

the first week of age (Lasher and Shane, 1994; Lukert and Saif, 1997).

In ovo vaccination with antibody-mixed live vaccine provides an alternative

mean of vaccination, in which the interference from the maternal antibodies is

avoided and the chicken are protected against IBD (Haddad et al., 1997). Whitfill

et al., (1995) developed this type of IBD vaccine by mixing the anti-IBDV

antibody with the virus particles and this was referred as “antibody-mixed live

vaccine (Whitfill et al., 1995). The vaccine was administered through In ovo

route to the SPF embryos and was reported to be safer and more potent than

the conventional IBD vaccine because it delayed the appearance of bursal

lesions, produced higher geometric mean antibody titers against IBDV,

generated protective immunity against challenge and produced no early

mortality (Johnston et al., 1997). The working mechanism of antibody-mixed live

vaccine was thought to be related to its specific cellular interaction with the

follicular dendritic cells in spleen and bursa (Jeurissen et al., 1998). The

disadvantages of In ovo vaccination using antibody-mixed live vaccine might be

the transient bursal destruction, observed both in SPF and commercial broilers

Page 76: Thesis

76

(Ivan et al., 2001). Some reported that the vaccine may cause bursal atrophy

(Corley et al., 2001; Corley and Giambrone, 2002) and cell-mediated

immunosuppression (Corley and Giambrone, 2002).

2.5.2 Inactivated Vaccines

Inactivated vaccines are usually used in the breeder hens for them to pass down

high, uniform, and persistent antibody titres to the progeny (Cullen and Wyeth

1976; Wyeth and Cullen 1978; Wyeth and Cullen 1979; Guittet et al., 1992). For

the vaccination to be effective, the hens must be previously vaccinated with a

live virus or had been exposed to the virus in the farm. Inactivated vaccines are

administered to the layers through subcutaneous or the intramuscular route at

sixteen- to twenty-week-old. In this way, the chicks will have the protective

maternal antibodies up to thirty days (Wyeth and Cullen 1976; Box 1989; van

den Berg and Meulemans 1991; Wyeth et al., 1992). However, the chicks will

not be protected from the challenge from the vvIBDV strains at later age (Wyeth

and Cullen 1979; van den Berg and Meulemans 1991).

Inactivated vaccine is usually prepared from the bursal homogenates of infected

chicks or from viral cultures on embryonated eggs or tissue culture, where the

virus is then inactivated by heat which generally is ineffective due to protein

denaturataion that affect the immunogenicity. Chemical inactivation with

formaldehyde and some alkylating compound like Binaryethylenimine (BEI) and

betapropiolactone has had success (Kuby, 1994). Formalin affects numerous

Page 77: Thesis

77

chemical grouping of proteins that cause the phenomenon of "membrane effect"

which tend to "close" the outer protein shell of the virus capsid before the nucleic

acid of the infectious genome is destroyed. In conditions of prolonged

incubation, the infectious nucleic acid can emerge and leads to a replication of

the virulent virus. This phenomenon can cause a subclinical infection or even

lead to disease (Brown, 1997). BEI, a member of group of alkylating substances

"aziridines" reacts very little with proteins and does not change the antigenic

components of virus. BEI has an inactivation reaction that is more specific for

the nucleic acid and it produces antigenically superior vaccine (Bahnemann,

1990). Habib et al., (2006) reported that 0.001 and 0.002 M BEI completely

inactivated the IBDV after 36 hours, whereas 0.1 and 0.2% formalin inactivated

the virus after 24 hours. On the basis of antibody titres, the BEI inactivated

vaccines were found twice the efficient as formalin inactivated vaccines.

Various adjuvants have been used in order to enhance the immune response

against specific antigens since 1925, when Ramon (1925) reported that it was

possible to enhance artificially the diphteric and tetanic antitoxin levels by the

addition of some substances. Most vaccine adjuvants used for poultry include

classical formulations, including water-in-oil (W/O), oil-in-water (O/W), saponins

and alum-based formulations. The exact mechanisms of such vaccine adjuvant

remain undefined (Schijns, 2000). Inactivated vaccines that make use of a W/O

emulsion as adjuvant are usually prepared by emulsifying an aqueous solution

comprising the inactivated antigen, a suitable oil and emulsifying agents until a

W/O emulsion is obtained in which the antigens are homogeneously distributed

Page 78: Thesis

78

over the aqueous phase. One of the most widely used adjuvants is the W/O

emulsion, Freund's complete adjuvant and incomplete adjuvants. In complete

adjuvant antigen suspended in W/O emusions with killed Mycobacterium

tuberculosis bacteria do stimulate strong T cell responses. Freund‟s incomplete

adjuvant (FIA) has the same oil surfactant mixture as FCA but does not contain

Mycobacteria. FIA is frequently used to boost animals that received a primary

antigen injection in FCA. It can also be used as the adjuvent for the primary

injection which favour humoral immunity without CMI (Lascelles, et al., 1989).

Mineral oil can be used to prepare W/O vaccines using arlacel A (oil-soluble

surfactant). The oil to water phase ratio was usually kept at ratio 1:1. Mineral oil

has high viscosity and some time it is difficult to inject the vaccine as it may lead

to local irritation (Hassan et al., 1992).

2.5.3 Recombinant and DNA Vaccines

IBDV proteins expressed in other prokaryotic systems can serve as IBD

recombinant vaccine. The recombinant IBDV protein will be a more effective

vaccine if it precisely mimics the authentic molecular structure of the viral protein

(Martinez-Torrecuadrada et al., 2003). Structural proteins of IBDV had been

expressed in the baculovirus expression system. The baculovirus-expressed

protein induces immunological response (Wang et al., 2000) and protects the

chickens from IBDV challenge (Vakharia et al., 1993; Snyder et al., 1994;

Vakharia et al., 1994; Pitcovski et al., 1996). However, the protection is

incomplete, evidence by the presence of bursal damage after IBDV challenge

Page 79: Thesis

79

(Dybing and Jackwood, 1998). In comparison with virus-like particles (VLP),

VPX tubules, and polyprotein-derived mix structures, the baculovirus-expressed

VP2 capsids elicit stronger immune response (Martinez-Torrecuadrada et al.,

2003). Hens vaccinated with baculovirus-derived recombinant VP2 vaccine

could pass down their maternal antibody that last for at least 20 days after

hatching to their progeny (Yehuda et al., 2000). Efforts to use improved

technology for the production recombinant IBDV protein using baculovirus

expression system is still ongoing (Wang and Doong, 2000).

VP2 had also been expressed in other expression vectors such as the

herpesvirus (Darteil et al., 1995; Tsukamoto et al., 2002), Marek‟s disease virus

(Tsukamoto et al., 1999; Tsukamoto et al., 2000), fowl adenovirus (Sheppard et

al., 1998), fowlpox virus (Bayliss et al., 1991; Boyle and Heine 1993; Tsukamoto

et al., 2000), and Semliki forest virus (Phenix et al., 2001), in which they may

serve as recombinant IBD vaccines. Recombinant fowlpox vaccine protects the

chickens from the IBDV-induced bursal damage but its efficacy depends on the

titre of the challenge virus and the chicken genotype (Shaw and Davison 2000).

In addition, the effective application of recombinant fowlpox (VP2) vaccine may

be restricted to the wing web and parenteral routes of inoculation (Boyle and

Heine 1994). In eukaryotic expression system, VP2 expressed in the yeast

confer passive protection against IBDV (Fahey et al., 1989; Macreadie et al.,

1990); probably because the multimeric forms yeast-derived VP2 were highly

immunogenic (Azad et al., 1991). Expression of VP2 in E. coli was not

immunogenic (Azad et al., 1991). Aside from single type of recombinant vaccine,

Page 80: Thesis

80

the dual-viral vector approach, an approach that uses Marek's disease and

Fowlpox viruses in expressing vvIBDV host-protective antigen may serve as a

quick and safe method in inducing strong and long-lasting protective immunity

against vvIBDV (Tsukamoto et al., 2000). Recently, a study by Cao et al., (2005)

showed that immunized SPF chickens with recombinant T4 bacteriophage

displaying VP2 protein elicited specific antibodies and have protection against

vvIBDV infection.

DNA vaccine could provide efficacious protection for chickens against IBDV

infection (Fodor et al., 1999; Chang et al., 2001). Effective DNA vaccine

included the VP2 gene in the plasmid DNA (Chang et al., 2003).

Transcutaneous plasmid-dimethylsulfoxide (DMSO) delivery technique for avian

nucleic acid immunization had been described (Heckert et al., 2002). DMSO

enhances liposome-mediated transfection of nucleic acid in chicken

macrophage cells and this phenomenon was exploited for the transcutaneous

delivery of naked DNA through the intact skin of the chickens. DNA-based IBD

vaccine had been delivered using this technique and the chickens were

protected against IBD (86% survival) (Heckert et al., 2002). Recently, Hsieh et

al. (2007) indicated that a prime-boost approach by priming with DNA vaccine

encoding the large segment gene of the IBDV and boosting with killed IBD

vaccine can adequately protect SPF chickens against challenge by homologous

or heterologous IBDV.

Page 81: Thesis

81

Recombinant vaccines offer several advantages of vaccines such as the

absence of residual pathogenicity, low sensitivity to maternal antibodies and low

risk of selection of mutants (Bayliss et al., 1991; Heine and Boyle 1993; Darteil

et al., 1995; Tsukamoto et al., 1999). But the primary problems have been to

deliver sufficient quantities of vaccines to elicit an effective immune response

and to mass-vaccinate thousands of birds in a flock (Oshop et al. 2003).

2.5.4 Anti-viral Drugs

Antiviral drugs have had very limited use in veterinary practise. It seems likely

that some of these drugs will be effective against IBDV. For example, by feeding

Azadirachta indica (Neem) dry leaves powder to the IBDV-infected birds,

scientist found that the bird‟s humoral and cell-mediated immune response

improved (Sadekar et al., 1998). Supplementation of ascorbic acid at 1,000 ppm

in the diet has been found to be beneficial to the chickens vaccinated against

IBD (Amakye-Anim et al., 2000). This is probably because ascorbic acid has

shown to ameliorate the immunosuppression caused by IBDV vaccination and

thus improved the humoral and cellular immune responses of the vaccinated

birds (Wu et al., 2000). Moreover, ascorbic acid supplemented birds have higher

body weight gains in comparison with the non-supplemented group (Amakye-

Anim et al., 2000). L-arginine has also been found to play a vital role in

modulation of protective immune response against IBDV (Tayade et al. 2006).

Recently, Oliveira et al. (2009) indicated that the inclusion of

Page 82: Thesis

82

mannanoligosaccharides (MOS) in broiler diets increased the immune response

to vaccinations against IBDV and NDV.

Virus neutralization factor (VNF) is a class of non-specific antiviral agents

produced in vivo in chickens in response to viral infection has been found to

directly inactivate IBDV particles (Whitfill et al., 1991). The recombinant

interferon alpha, which has antiviral effect, has also been shown to suppressed

IBDV plaque formation in a dose-dependent manner and ameliorated IBDV and

NDV infection in both SPF and commercial chickens (Mo et al., 2001). The

effect of the interferon therapy depends on the route of administration, in

commercial chickens than in SPF chickens (Mo et al., 2001).

Page 83: Thesis

CHAPTER 3

ADAPTATION AND ATTENUATION OF vvIBDV ISOLATES IN TISSUE CULTURE FOR DEVELOPMENT OF VACCINES

3.1 Introduction

Since the first description of Infectious bursal disease (IBD) by Cosgrove in 1957

(Cosgrove, 1962), IBD has become an important viral disease threatening the

chicken industry worldwide. Its causative agent is IBD virus (IBDV) which has

two known serotypes 1 and 2. Serotype 1 IBDV strains are pathogenic for

chickens and they do cause serious problems in the poultry industry with

individual strains differing markedly in their virulence. Serotype 2 strains which

have been isolated from fowl, turkey and duck (McFerran et al., 1980) are not

pathogenic for chicken.

Since the mid 1980‟s a new IBDV serotype 1 pathotype has emerged and it is

characterized by an acute course with unusually high mortality. It was first

described by Box (1989) in the Netherlands and ever since such virulent

pathotype have been reported worldwide (Chettle et al., 1989; van den Berg et

al., 1991; Fabio et al., 1999).

IBDV belongs to the genus Avibirnavirus of the family Birnaviridae (Leong et al.,

2000), the genome encodes five viral polypeptides, designated VP1 to 5

(Sharma et al., 2000). The VP2 and VP3 are the major structural proteins of the

Page 84: Thesis

54

viral particles and the VP4 is a proteolytic enzyme-like protein, which is involved

in the processing of the precursor polypeptide. The function of VP5 has not been

defined although it has been shown not to be essential for viral replication and

infection (Mundt et al., 1997; Yao et al., 1998), but plays an important role in the

release of viral progeny from infected cells (Lombardo et al., 2000). The VP1 is

known as an RNA-dependent RNA polymerase (RdRp) (Spies et al., 1987) with

capping enzyme activities (Spies and Muller, 1990).

IBDV has its major effect on lymphoid tissues of chicken. The bursa of Fabricius

and spleen are the tissue of choice for the isolation of IBDV, but the bursa is the

most commonly used. Other organs contain the virus, but at a lower

concentration (Lukert and Saif, 2003).

The chorioallontoic membrane (CAM) route of inoculation in 9-11 days old

embryos is the most sensitive route for the isolation of the virus. Classic IBDVs

(caIBDV) usually kill the embryos in 3-5 days with congestion and subcutaneous

hemorrhages in the embryos (Hitchner, 1970), while variant strains of IBDV

(vaIBDV) do not kill the embryos, but result in embryo stunting, discoloration,

splenomegaly and hepatic necrosis (Lukert et al., 1975).

As result of the time consuming, nature and the over burden cost implication

with the use of specific-pathogen-free (SPF) embryos or for SPF chicks, which

are the traditional method of propagating IBDV, there had been a call for the use

of cell culture. The use of cell culture in growing avian viruses has become an

Page 85: Thesis

55

increasingly economical, less laborious, continuous and efficient tool with an

advantage of measuring virus effects outside the host animal. There are two

major cell culture, the cell culture of avian origin and that of mammalian origin

that had gained continuous used in the various investigations associated with

IBDV ( Kibenge et al.,1988; Peilin et al., 1997; Ahasan et al., 2002).

Primary cell cultures of chicken embryo kidney (CEK) cells and chicken embryo

fibroblast (CEF) had been use for the adaptation and attenuation of many IBDV

isolates (Lukert et al., 1974; McNulty et al., 1979; Martin et al., 1998) with

formation of small plaque (SP) and large plaque (LP) clones (Lange et al.,

1987). A continuous fibroblast cell line of Japanese quail origin was also found

to support the replication of IBDV and several other viral pathogens of poultry

(Cowen and Braune, 1988).

On the other hand, mammalian continuous cell lines had been reported to be

susceptible to IBDV. Of these include RK-13 (Petek et al., 1973), Vero cells

(Leonard 1974; Lukert et al., 1975; Jackwood et al., 1987), MA-140 and BGM-70

cells (Jackwood et al., 1987). Previous studies comparing the replication of

IBDV in Vero and CEK cultures revealed no visible or significant differences in

virus titers (Leonard, 1974), although the cycle of replication was reported to be

longer in Vero cells than in CEK cells (Lukert et al., 1975).

Lukert et al. (1975) reported that initial passage of IBDV in Vero cells did not

produce visible cytopathic effect (CPE) until the 4th passage at 13th days post

Page 86: Thesis

56

inoculation while that of CEF cultures were noticeable 3 days after virus

inoculation with visible aggregates of tiny round refractive cells which later

spread to the entire cell sheet. These altered cells later became detached from

the dish, leaving empty areas in the cell sheet (Cho et al., 1979).

Pathogenic bursa-derived IBDV is difficult to adapt to cell cultures as extensive

serial blind passages in cell culture (Hassan and Saif,1996), in the CAM as well

as in the yolk sac of embryonated chicken eggs (Yamaguchi et al., 1996a) are

needed to achieve the growth of the virus. Tsai and Saif, (1992) reported the

failure of a very virulent IBDV (vvIBDV) isolate, E Del-IBDV that was grown in

BMG-70 cells, to replicate in chicken embryo fibroblast (CEF) cells. The possible

reason for this is yet to be understood.

The use of continuous cell lines has several advantages over the primary cell

culture of avian origin as they are easy to handle, maintain and free from

vertically transmitted extraneous viruses of avian origin (Hassan et al., 1996).

They are also valuable and economical method of growing IBDV for mass

commercial vaccine production (Rasool and Hussain, 2006).

However, there is a dearth of information on the adaptation of very virulent

vvIBDV isolates to continuous cell line. Hence this investigation attempt to

adapt, propagate and attenuate vvIBDV Malaysian isolates to cell culture of

mammalian and avian origin namely Vero cells and DF-1, respectively with the

Page 87: Thesis

57

hope of achieving a positive step towards a cheaper means of IBD vaccine

production.

Therefore, the objectives of this study were to adapt, propagate and attenuate

local vvIBDV to Vero cells and DF-1 cells, and determine the virus titres in these

two cell lines.

3.2 Materials and Methods

3.2.1 IBDV Isolates

Three vvIBDV isolates obtained in Malaysia were used in the study namely

UPM94273, UPM04190 and UPM0081, with an accession number of

AY791998, AF527039 and EF208038, respectively. The isolates were kindly

supplied by Prof. Dr. M. Hair-Bejo, Universiti Putra Malaysia.

Page 88: Thesis

58

3.2.2 IBDV Inoculum Preparation

The CAM of the IBDV infected chicken eggs were grounded separately by using

sterile mortars and sand to make 1:2 (w/v) dilution of each in sterile phosphate

buffer saline (PBS) pH 7.2 (Appendix A). Processed samples were centrifuged

at 3000 rpm for 15 minutes at 4°C (MSE, Mistral 4L, UK). The supernatants

were collected and filtered through a 0.45 µm filter (Sartorius, Germany). The

samples were treated with antibiotics-antimycotics (Appendix A) (GIBCO

Laboratories, N.Y.1072 USA) in 1:10 suspension and incubated at 4°C for 1

hour. The samples were used immediately or stored at -80°C until used (Heto

ultra freeze, Denmark).

3.2.3 Propagation of Viruses in SPF Embryonated Chicken Eggs via Chorioallantoic Membrane

Nine to eleven days old SPF embryonated chicken eggs were candled for

viability. The eggs were marked at the side approximately midway along the

long axis where the vein structure was well developed and at area of about 0.5

cm below and parallel to the base of air cell. The eggs were placed horizontally

and disinfected with 70% alcohol. A hole was then drilled through the eggshell at

the top of the air cell and on the side of the egg that had been marked. It was

punched carefully not to penetrate the CAM. Holding the eggs in the same

position, the air of the air cell was drawn out by rubber bulb and an artificial air

cell was formed directly over the CAM (Yamaguchi et al., 1996a).

Page 89: Thesis

59

The CAM homogenate (0.1mL), which was prepared earlier from the IBDV

infected CAM, was then inoculated into the artificial air cell using 27 gauge

needle. The hole at air sac was sealed without turning the eggs from their

horizontal position. The eggs were rocked gently to distribute the inoculums

evenly over the CAM surface. The inoculated SPF embryonated chicken eggs

were placed horizontally into incubator at 37C and monitored daily. It was

candled at least twice a day for any abnormalities for 7 days. Death embryo

within the first 24 hours was considered to be non-significance and can be due

to non-specific causes such as trauma and injury during handling. Death embryo

after 24 hours was kept in the refrigerator at least 24 hours and then examined

for the pathological changes in sterile condition. The CAM from dead embryos

was collected in sterile condition as soon as possible. The samples (UPM94273,

UPM04190 and UPM0081) were then processed using the same procedure as

described earlier (section 3.2.2). The samples were then further passaged until

the third passage. The third-passaged CAMs homogenate was kept in -80oC

until use.

Page 90: Thesis

60

3.2.4 Adaptation, Replication and Attenuation of vvIBDV in Cell Culture

3.2.4.1 Vero Cell Line

Adult African green monkey kidney cells (Vero cell line, ATCC Lot number

58078553), were obtained from the Virology Laboratory, Faculty of Veterinary

Medicine, Universiti Putra Malaysia.

3.2.4.2 DF-1 Cell Line

Secondary embryo fibroblasts derived from an EV-0 embryo were obtained from

the Virology Laboratory, Faculty of Veterinary Medicine, Universiti Putra

Malaysia.

Page 91: Thesis

61

3.2.5 Resuscitation of Frozen Cell Line

Frozen ampoule was removed from the liquid nitrogen container wearing over

coat, mask and gloves. The lower half of ampoule was submerged in a water

bath at 37oC for 2 minutes and then transferred it to class-ll safety cabinet. The

outside of the ampoule was cleaned with a tissue moistened with 70% alcohol

and the lid was gently opened. The cells were slowly pipetted and dropped into

a 25 cm2 tissue culture flask having 5 mL pre-warmed growth medium to dilute

out the DMSO. The flask was closed and kept in CO2 incubator at 5% CO2 and

37oC. The cells were examined twice daily under inverted microscope

(Olympus®, Japan).

3.2.6 Sub Culturing of Adherent Monolayer

Vero cell lines and DF-1 cells were used for tissue culture repassaging. The old

growth medium covering the confluent monolayer of the Vero cells and DF-1

cells in 25 cm2 sterile disposable polystyrene cell culture flask (Nunc Easyflasks,

Sigma) was removed. The cells were rinsed twice with warm 37oC sterile PBS,

pH7.2. An amount of 5 mL of warm antibiotic-trypsin versene (ATV) (Appendix

A) solution was added to chelate the cells. When the cells started to detach, the

ATV was poured away and the cell culture flask was shaken vigorously to

Page 92: Thesis

62

complete cell detachment. The cells were then resuspended in sufficient amount

of RPMI growth medium containing 10% (v/v) fetal bovine serum (FBS), and 1%

(v/v) antibiotic-antimycotic. This allowed preparation of two or three 25 cm2 cell

culture flasks from the original one of 25 cm2. The new flasks were incubated at

37oC in CO2 incubator (Thermo Forma, USA) and observed daily until the

confluent monolayers of cells were formed, usually within 2-3 days. Upon

formation of monolayers, the old media were replaced with fresh RPMI

maintenance medium with the same content as the growth medium, except that

FBS was reduced to 1% (v/v) (Appendix A).

3.2.7 Infections of Vero Cell and DF-1 Cells Monolayer

Three IBDV isolates (UPM94273, UPM04019 and UPM0081) were used to

infect the healthy, semi-confluent monolayers of Vero cells and DF-1 cells with

concentration of seed cells were 3 x 106 and 1.5 x 105 cells/mL, respectively.

The growth medium of the flasks was removed and the monolayers were

washed twice with prewarmed sterilized PBS. 0.1 mL of vvIBDV inoculums was

dispensed over the each monolayer, the inoculums was spread uniformly over

each monolayer and flasks were incubated at 37oC for I hour with intermittent

rotation to allow the virus to adsorb on the surface of Vero cells and DF-1 cells.

Five mL of sterilized pre warmed maintenance medium (Appendix A) was added

in each flask and incubated them at 37oC in 5% CO2. The monolayers were

Page 93: Thesis

63

examined twice daily under inverted microscope for cytopathogenic effects

(CPEs).

3.2.8 Harvesting of Virus

If the CPEs were found, infected cells were harvested. The virus infected cells

and culture medium were repeated frozen and thawed three times. After clarified

by low-speed centrifugation at 3,000 rpm for 20 minutes at 4oC, the supernatant

fluids were harvested and passed through a 0.45 µm filter, aliquot, and stored at

-80oC or used as IBDV inoculums inoculated in subsequent cell cultures for

further passages.

3.2.9 Adaptation and Attenuation

The passage 1 (P1) virus was inoculated again to fresh, healthy and semi

confluent monolayers of Vero cells and DF-1 cells using the same technique for

CPEs. The virus was harvested by three freeze-thaw cycles, clarified and

labeled as passage 2 (P2). In this way IBDV was adapted to Vero cells or DF-1

cells when the CPEs were clear and consistent. The adapted virus was serially

passage until it became attenuated (non-pathogenic).

Page 94: Thesis

64

3.2.10 Tissue Culture Infective Dose 50 (TCID50)

Titration of the virus was achieved by observation of the CPEs. The two

passages of 6th and 9th in DF-1 cells and the three passages of 10th, 15th and

20th in Vero cells were frozen and thawed for three times before they were

clarified at 4000 rpm for 20 minutes at 4oC. The supernatant was collected and

serial ten fold dilutions from this supernatant were prepared in PBS. Vero cells

and DF-1 cells monolayer were prepared in 96-well tissue culture microtitration

plate. Each dilution of virus (100 µl) was inoculated in each well of first row,

leaving the last two well as negative control. The plate was incubated at 37oC in

5% CO2 for 5 days, the plate was examined twice daily for CPEs. The virus titer

was determined by Reed-Muench method (1938). This method evaluates an

endpoint where 50% of the cell cultures were infected. The cell culture infectious

dose affecting 50% of the cultures (TCID50) was calculated using a formula that

takes into account the accumulated percentage of infected cultures (Appendix

B).

3.2.11 IBDV Identification and Confirmation

The growth of IBDV on Vero cells and DF-1 cells monolayers was identified

through its CPEs. The characteristics changes in the infected monolayers were

carefully noted after each passage. The time for the appearance of CPEs of the

Page 95: Thesis

65

adapted and attenuated virus on Vero cells and DF-1 cells was also recorded.

The virus of each passage was obtained from the infected tissue culture filtrate

and subjected to indirect immunoperoxidase (IIPS).

3.2.12 Indirect Immunoperoxidase Staining Test

The indirect immunoperoxidase test (IIP) was done according to the method of

Guvenc et al., 2004. The infected Vero cell and DF-1 cells were fixed with cold

methanol: acetone (50:50 v/v) for 5 minutes. The glass slides were then

immersed in 1% H2O2 in absolute methanol for 30 minutes. The PBS was then

added to the glass slide for 15 minutes. The glass slides were then air dried.

The hyper immune serum kindly provided by Prof. Dr. M. Hair-Bejo was diluted

1:1000 with PBS and added to the glass slide incubated for 1 hour in room

temperature. The glass slides were then washed 3 times with PBS for 5 minutes

each. The rabbit anti-chicken IgG-HRP conjugated secondary antibody (Bio-

Red, USA) was then added to the glass slides (1: 1000) and incubated for 1

hour at room temperature. DAB substrate solution (DAB reagent set, Invitrogen,

USA) was then added to the glass slides and incubated for 10 minutes in a dark

room. The slides were mounted with buffer glycerol and examined under light

microscope.

Page 96: Thesis

66

3.3 Results

3.3.1 Chorio-allantoic Membrane for UPM94273

There was 60% mortality of the embryo in CAM within 7 day post infection (pi) in

passage one and two. The CAM was mildly congested while the embryos were

haemorrhagic. On the third passage, 80% mortality occurred within day 7 pi. At

this passage, the embryos were severely hemorrhagic and oedematous (Table

3.1, Figure 3.1).

3.3.2 Chorio-allantoic Membrane for UPM04019

There was 100% mortality in the embryo in CAM within 7 day pi following

passage one and two. The CAM was severely congested while the embryos

were haemorrhagic. On the third passage, mortality occurred at day 5 pi. At this

stage, the embryos were severely hemorrhagic and oedematous (Table 3.1,

Figure 3.1).

Page 97: Thesis

67

3.3.3 Chorio-allantoic Membrane for UPM0081

There was 100% mortality in the embryo in CAM within 5 day pi in the passage

one, two and three. The CAM was severely congested. The dead embryos

showed severe petechial to ecchymotic hemorrhages (Table 3.1, Figure 3.1).

Page 98: Thesis

68

Figure 3.1a: (A): Uninfected control embryonated SPF chicken eggs. (B): UPM94273 dead embryo with severe haemorrhage

Page 99: Thesis

69

Figure 3.1b: (C): UPM04019 dead embryo with severe haemorrhage. (D): UPM0081 the embryo infected showed severe petechial to ecchymotic haemorrhage (arrows)

Page 100: Thesis

70

Table 3.1: Mortality of SPF embryonated eggs following vvIBDV inoculation into CAM route

vvIBDV strains

Passage

No. of SPF eggs

Cumulative mortality (Days post inoculation)

Total no. of SPF egg viable

Mortality (%)

1

2

3

4

5

6

7

1

5

0a/5b

0/5

1/5

1/5

2/5

2/5

3/5

2

60 UPM94273

2

5

0/5

1/5

1/5

2/5

2/5

2/5

3/5

2

60

3

5

0/5

2/5

2/5

3/5

3/5

3/5

4/5

1

80

1

5

0/5

2/5

2/5

3/5

4/5

4/5

5/5

0

100 UPM04019

2

5

0/5

2/5

3/5

3/5

3/5

4/5

5/5

0

100

3

5

0/5

2/5

4/5

4/5

5/5

5/5

5/5

0

100

1

5

0/5

1/5

3/5

3/5

5/5

5/5

5/5

0

100 UPM0081

2

5

0/5

2/5

2/5

4/5

5/5

5/5

5/5

0

100

3

5

0/5

2/5

3/5

4/5

5/5

5/5

5/5

0

100

a: Total number of eggs dead b: Total number of eggs inoculated

Page 101: Thesis

71

3.3.4 IBDV Replication and Adaptation in Vero Cell Line

The CPE was not observed in the first six passages for UPM94273 and

UPM04019. However, the UPM0081 was successfully adapted at 4th passage

with the formation of visible CPEs and attenuated in Vero cells at the 20th

passage. With UPM0081, at 4th and 5th passages, there was complete CPEs

up to 15 and 14 days pi, respectively (Figure 3.2), while at 6th, 9th, and 12th

passages, CPE was observed on 8, 7 and 6 days pi, respectively (Figure 3.3),

and at 13th to 20th passages, the CPE was observed at 4 days pi. (Figure 3.4)

(Table 3.2). The CPE in Vero cells was characterized by aggregates of tiny

round refractive cells that later spread to the entire cell sheet. These abnormal

cells later detached from the surface to leave empty areas in the cell culture.

3.3.5 IBDV Replication and Adaptation in DF 1 Cell Line

The CPE was not observed in the first six passages of UPM94273 and

UPM04019 in DF1 cell line. Again, UPM0081 showed CPE at the third to the

ninth passages. The CPE at the third passage started with rounding and

clumping of few cells at 3 days pi which later progressed to enhanced rounding

and clumping of cells at 5 days pi (Figure 3.5). At 4th passage, the CPE was

similar to those of the 3rd passage, but more concentrated with granular

cytoplasm. At 5th passage however, there was cellular degeneration and

detachment from the surface monolayer by 3 days pi, (Figure 3.6). At 4 days pi,

there was rounding and clumping of large number of cells with intense

Page 102: Thesis

72

cytoplasmic granulation and detachment of cells with few cells floating in the

media. At 6th to 9th passages, the CPE were characterized by degenerative and

more detachment of cells at 3 days pi (Figure 3.7, Table 3.3).

Page 103: Thesis

73

Table 3.2: Percentage of CPE monolayer Vero cells following UPM0081vvIBDV inoculation

vvIBDV passage

No. of

monolayers

plates

CPEs (Days post inoculation)

Total no. of CPE/ plate

CPE

(%)

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

1 4 0a/4b 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0 0

2 4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0 0

3 4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0 0

4 4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 4/4 4 100

5 4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 4/4 – 4 100 6 4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 4/4 – – – – – – – 4 100

7 4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 4/4 – – – – – – – 4 100

8 4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 4/4 – – – – – – – 4 100 9 4 0/4 0/4 0/4 0/4 0/4 0/4 4/4 – – – – – – – – 4 100

10 4 0/4 0/4 0/4 0/4 0/4 0/4 4/4 – – – – – – – – 4 100

11 4 0/4 0/4 0/4 0/4 0/4 0/4 4/4 – – – – – – – – 4 100 12 4 0/4 0/4 0/4 0/4 0/4 4/4 – – – – – – – – – 4 100

13 4 0/4 0/4 0/4 4/4 – – – – – – – – – – – 4 100

14 4 0/4 0/4 0/4 4/4 – – – – – – – – – – – 4 100 15 4 0/4 0/4 0/4 4/4 – – – – – – – – – – – 4 100

16 4 0/4 0/4 0/4 4/4 – – – – – – – – – – – 4 100 16 4 0/4 0/4 0/4 4/4 – – – – – – – – – – – 4 100

18 4 0/4 0/4 0/4 4/4 – – – – – – – – – – – 4 100

19 4 0/4 0/4 0/4 4/4 – – – – – – – – – – – 4 100 20 4 0/4 0/4 0/4 4/4 – – – – – – – – – – – 4 100

a: Total number of CPE b: Total number of plates inoculated – The cells were harvested.

Page 104: Thesis

74

Table 3.3: Percentage of CPE monolayer DF-1 cells following vvIBDV inoculation

vvIBDV passage

No. of

monolayers

plates

CPEs (Days post inoculation)

Total no. of CPE/

plate

CPE

(%)

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

1

4

0a/4b

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0

0

2

4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0/4

0

0

3

4

0/4

0/4

0/4

0/4

4/4

4

100

4

4

0/4

0/4

0/4

4/4

4

100

5

4

0/4

0/4

0/4

4/4

4

100

6

4

0/4

0/4

4/4

4

100

7

4

0/4

0/4

4/4

4

100

8

4

0/4

0/4

4/4

4

100

9

4

0/4

0/4

4/4

4

100

a: Total number of CPE b: Total number of plates inoculated – The cells were harvested.

Page 105: Thesis

75

3.3.6 IBDV Titration (TCID50/mL)

As shown in Table 3.4, the TCID50 of UPM0081 strain on Vero cell and DF1 cell

lines ranged from 103.5 to 107.4 and 101.5 to 102.5, respectively. The IBDV titre

continued to increase from P6 to P20 (Appendix B).

Table3.4: Virus titer determined by Tissue Culture Infective Dose50 (TCID50)

vvIBDV Passages

TCID50 on the Vero cells and DF-1 cell line

Vero cells

DF-1 cells

P6 103.5 101.5 P9 104.5 102.5

P10 104.7 ND P15 106.7 ND P20 107.4 ND

ND (Not done) 3.3.7 IBDV Identification through Indirect Immunoperoxidase Staining (IIPS) Test

In IIPS technique, the cytoplasm of the IBDV infected Vero cells and DF-1 cells

with CPE were positively stained light brown (Figures 3.8 and 3.9).

Page 106: Thesis

76

Figure 3.2: (A) Uninfected control Vero cells monolayer. (B) Cytopathic effect by UPM0081 isolate at 4th passage at day 15 pi. The arrow shows cell rounding and aggregation. 10 x. Bar = 200 µm

Page 107: Thesis

77

Figure 3.3: (A) Vero cell monolayer at 6th passage at day 8 pi (B). Vero cell monolayer at passage 12th at day 6 pi. The arrows show cell rounding and aggregate in clumps and granulation in cytoplasm. 10x. Bar = 200 µm

Page 108: Thesis

78

Figure 3.4: (A) A Vero cell monolayer at passage 13th at day 3 pi (B). A Vero cell monolayer at 20th passage at day 4 pi. The arrows show detachment of cells from the substrate with the eventual destruction of the entire monolayer. 10 x. Bar = 200 µm

Page 109: Thesis

79

Figure 3.5: (A) Uninfected control DF-1 cells monolayer. (B) Cytopathic effect of UPM0081 isolate of the 3rd passage at day 5 pi. The arrow shows cell rounding and clumping. 10 x. Bar = 200 µm

Page 110: Thesis

80

Figure 3.6: (A) DF-1 monolayer of the 4th passage at day 5 pi. Affected cells are more concentrated with granular cytoplasm. (B) DF-1 cells at 5th passage at day 4 pi. The arrow shows detachment of cells from the substrate (B). 10 x. Bar = 200 µm

Page 111: Thesis

81

Figure 3.7: (A) DF-1 cell monolayer at 6th passage at day 3 pi. (B) DF-1 cell monolayer at 9th passage at day 3 pi. The arrow shows degenerate cells and more detachment of cells from the substrate. 10 x. Bar = 200 µm

Page 112: Thesis

82

Figure 3.8: Identification of IBD antigens in Vero cells culture using infected cell cultures stained with HRP-conjugated antibody. (A) Uninfected control Vero cells. (B) Vero cells infected with UPM0081 at 20th passage at day 2 pi. The arrow shows the presence of specific intracytoplasmic brownish colouration. 20 x. Bar = 100 µm

Page 113: Thesis

83

Figure 3.9: Identification of IBD antigens in DF-1 cells culture using infected cell cultures stained with HRP-conjugated antibody. (A) Uninfected control DF-1. (B) DF-1 infected with UPM0081 at 4th passage at day 2 pi. The arrow shows the presence of specific intracytoplasmic brownish colouration. 20 x. Bar = 100 µm

Page 114: Thesis

84

3.4 Discussion

This study was initiated to adapt and propagate local vvIBDV isolates in two cell

lines namely Vero cells and DF-1 cells. The three Malaysia isolates of vvIBDV

namely the UPM94273, UPM04019 and UPM0081 were first adapted through

serial passage in SPF embryonated eggs and the embryo showed characteristic

lesions of oedema, distension of the abdomen, generalized congestion and

haemorrhages of the body. The CAM inoculated with UPM04190 and UPM0081

isolates resulted in 100% embryonic death at passage three, while UPM94273

resulted in 50% to 80% embryonic death during three passages. These lesions

were consistent with other studies of vvIBDV (Hair-Bejo et al., 1995a;

Yamaguchi et al., 1996a), while the pathogenicity in the embryo suggests the

virulence of these isolates.

The embryonated egg adapted UPM0081 strain was successfully adapted to

grow in Vero cells after four passages and in DF-1 cells after three passages,

while the other failed to adapt on Vero cell and DF-1 cells after six passages.

The failure of some of the vvIBDV (UPM94273 and UPM04019) to adapt is

consistent with the findings of some investigators that it is usually difficult to

adapt and grow pathogenic bursal derived field strains in vitro as some wild type

strains had been reported to fail to adapt to mamallian cell culture, despite

increased serial blind passages (McFerran et al., 1980; Kibenge and McKenna,

1992; Hassan et al., 1996). The poor adaptation rates of vvIBDV to CEF have

Page 115: Thesis

85

been reported previously (Lee and Lukert, 1986). The possible reason for this

failure is not certain as some workers suggested a receptor theory which is still

very much under investigation (Zhu et al., 2008).

The growth pattern of UPM0081 in Vero cells was clear and consistent. It

showed CPEs of IBDV at the fourth passage onward up to 20th passage

(Ahasan et al., 2002), but slightly varied from findings of Rasool and Hussain

(2006) where the vvIBDV was adapted to Vero cell line after the third

passage.This observation was sharply in contrast to that of Tsai and Saif (1992)

and Hassan and Saif (1996) who showed CPE at second passage in BGM-70

cell line (baby monkey kidney cell line). The difference might be due to the cell

culture passage level of the virus strains used or variation in sensitivity of cell

culture to different strains.

The difference between primary cell culture and secondary cell culture to adapt

vvIBDV had been reported by many researchers. Some scientists were able to

detect CPEs in CEF only on 5th passage onwards (Pahar and Rai, 1997; Mathur

et al., 1999; Gupta et al., 2001), while in this study, CPE was evident in DF1 (a

cell line derived from EV-0 embryo) from the third passage with UPM0081 as

other vvIBDV isolates failed to adapt. This findings is in line with reports of other

workers (Wang et al., 2009) with IBDV Gt strain in CEF and DF-1 cells where

CPE was observed clearly at 48 hours pi. and became more obvious at 60

hours pi.

Page 116: Thesis

86

The classical method of detecting the replication of virus in the cell culture using

the cytopathic effect was augmented in this study with indirect

immunoperoxidase and the viral antigen was observed as brownish

intracytoplasmic granules in both Vero cells and DF-1 cells, These findings is

similar to the report of Guvenc et al., (2004).

Of note is the low titer obtained from DF-1 cells when compared to Vero cell line

as infectious virus titer in Vero cell line is 100 times more than that obtained with

DF-1 at passage 9. This observation is in sharp constrast with that of Lukert et

al., (1974) where approximately equal titers were obtained in Vero and CEK cell

line, but it similar to the report of Kibenge et al, (1988) where Vero cell line gave

180 times more than the total virus titer in CEF culture.The possible reason for

this discrepancy may be related to species difference in the origin of the two cell

lines and the receptors for attachment and replication (Kim et al., 2006). It has

been reported that DF1 uses a molecule that contain chicken heat shock protein

(cHsp90α) (Lin et al., 2007), while Vero cells utilise RAVP2 and RVVP2

receptors (Yip et al., 2007). However, the knowledge of these receptors or

coreceptors in IBDV is very poor. The variation in the mitotic and metabolic

rates of the individual cell lines can also be adduced for the difference. It is

pertinent to note that Vero cell line, BGM-70 cell line were from the kidney of

green monkey and baby monkey, respectively which strongly suggest an

inherent ability of kidney cells to permit the growth of the virus. It will be

Page 117: Thesis

87

interesting therefore to investigate more on the tropism of the virus in live

animals.

It is known that secondary cell lines have several advantages over the primary

cell cultures as the cost of maintenance of secondary cell line is lower than

production of CEF from SPF chicken embryos. And also, primary cell cultures

have short life span as a continuous cell line could afford continuous source of

cells by simple protocol when compared to primary cell cultures, which is time

consuming and laborious, In addition, some primary avian cell culture may

contain exogenous avian virus not found in secondary cell culture (van den

Berg, 2000).

To product large quantities of virus for adaptation, attenuation and vaccination

development, it is important that the virus grows to high titers in the in vitro

tissue culture systems. The UPM0081 strain produced higher titers when

propagated in Vero cells than in DF-1 cell line, the possible reason for this

observation is not clear.

It was concluded that only UPM0081 vvIBDV strain was successfully adapted

and attenuated in the two continuous cell lines, while the other strains

(UPM04190 and UPM94273) failed. The virus was first propagated in SPF

embryonated chicken eggs for three times prior to inoculation into the cell lines.

The UPM0081 vvIBDV strain yielded higher titer in Vero cells than DF-1 cells.

Page 118: Thesis

88

Thus, the propagation and attenuation of UPM0081 strain in Vero cells line has

high potential to be used for future IBD vaccines development.

Page 119: Thesis

CHAPTER 4

MOLECULAR CHARACTERISATION OF THE ADAPTED AND ATTENUATED vvIBDV ISOLATE

4.1 Introduction

Infectious bursal disease virus (IBDV) is a double-stranded RNA (dsRNA) virus

belonging to the genus Avibirnavirus within the family Birnaviridae (Kibenge et

al., 1988). There are two distinct serotypes of IBDV (McFerran et al., 1980). The

Serotype 1 strains cause immunosuppression as well as an acute fatal disease

of young chickens while Serotype 2 viruses are avirulent for chickens. Serotype

1 viruses are further categorized as classical virulent, antigenic variant and very

virulent strains depending on their pathogenicity and/or antigenicity. Non-

enveloped icosahedral IBDV particles contain the bi-segmented dsRNA

genome. The major open reading frame (ORF) in the larger genome segment A

encodes a polyprotein which is co-translationally and autocatalytically cleaved

into the two structural viral proteins VP2 and VP3, and a viral protease VP4

(Hudson et al., 1986). A second ORF in the segment A encodes a nonstructural

protein, VP5 (Muller and Becht, 1982). The smaller segment B encodes the

VP1, which is an RNA-dependent RNA polymerase (RdRp) (Spies et al., 1987).

The PCR has been used to detect and characterise IBDV (Spies et al., 1987;

Wu et al., 1992). The PCR uses two oligonucleotide primers to hybridize the

Page 120: Thesis

90

segments of the template DNA, allowing DNA polymerase to produce more

DNA by copying the template DNA lying between the 3 ends of primers. The

oligonucleotides hybridize specifically to the target sequence and in the

presence of DNA polymerase coupled with excess deoxynucleotide

triphosphates, prime new DNA synthesis (Wu et al., 1992). IBDV being a double

stranded RNA virus, needs a reverse transcription (RT) in the PCR for IBD to

synthesize cDNA.

The hyper variable (hv) region in VP2 is the most investigated area of the IBDV

genome. It is an extreme sequence which encodes the immune dominant viral

epitopes (Becht et al., 1988). This epitope region contains only a 145 amino

acid (aa) fragment spanning from aa 206 to 350, which comprises the central hv

portion of the major viral capsid structure protein VP2. Within this region lies two

hydrophilic peaks, the first peak is from aa 212 to 224 and the second peak is

from aa 314 to 324 (Fahey et al., 1989; Bayliss et al., 1990; Brown et al., 1994).

Specific amino acid changes within the hv region and an adjacent downstream

serine-rich heptapeptide sequence (SWSASGS) and amino acid residues 326 to

332 have been proposed as potential sites responsible for virus attenuation or

antigenic determination (Heine et al., 1991; Vakharia et al., 1994; Yamaguchi et

al., 1996b; Dormitorio et al., 1997). In addition, several amino acid residue

changes at the VP2 gene can be used to differentiate the very virulent (vv),

attenuated (at), variant (va) and classical (ca) strains of IBDV. The amino acid

residue changes at P222A (proline to alanine), V256I (valine to isoleosine) and

Page 121: Thesis

91

L294I (leucine to isoleosine) can be used as a marker for vvIBDV, G254S

(glycine to serine) and Q249K (glutamine to lysine) for vaIBDV, while amino acid

changes at D279N (aspartic to aspargine) and A284T (alanine to threonine) has

been demonstrated for atIBDV (Yamaguchi et al., 1996b; Cao et al., 1998).

Sequence analysis of the structural protein VP2 of the isolate was focused

especially on the region between residue 206 and 360 which is referred to as

very variable region (Dormitorio et al., 1997; Lin et al., 1993).

The passage of the virus in vitro has been associated with attenuation of

virulence as evidenced by a reduction in the ability of the virus to induce bursal

lesions (Yamaguchi et al., 1996a). Thus, attenuated vvIBDV can be produce by

in vitro with extended serial passages. The Vero cells-adapted strains showed

considerable reduction in virulence (Hyuk and Soo, 2004), but the possible

genomic changes for the attenuation of vvIBDV in different cell lines have not

received sufficient attention especially with local isolates of vvIBDV.

Therefore, the objectives of the study were:

1. to amplify, clone and sequence the hypervariable region of VP2 gene

(643-bp) of the UPM0081 IBDV isolate adapted and attenuated in Vero

cells and DF-1

2. to determine the molecular characteristic of the adapted and attenuated

IBDV in the cell lines

Page 122: Thesis

92

4.2 Materials and Methods

4.2.1 Sample Preparation

The 5, 7, 8, 9, 10, 15, and 20 passages of UPM0081 vvIBDV isolate in Vero

cells and the 5 and 7 passages in DF-1 cells were choosen as the sample for

reverse transcriptase-polymerase chain reaction (RT-PCR). The virus infected

cells from the tissue culture flasks were repeatedly frozen and thawed three

times. After being clarified by low-speed centrifugation at 3000 rpm for 20

minutes at 4°C (Beckman, USA), the resultant supernatant fluids were

harvested and further clarified with centrifugation at 10,000 rpm for 30 minutes

at 4°C. The collected supernatant was passed through a 0.45 µm filter before

stored or used for RNA extraction.

4.2.2 RNA Extraction

The viral RNA was extracted by using Trizol reagent according to the

manufacturer‟s instruction (Gibco BRL, Life Technologies, USA). The partial

sample with the volume of 250 µl was mixed with 750 µl Trizol Reagent solution

in a 1.5 mL Eppendorf tube and incubated at room temperature (RT) for 5

minutes. Two hundred µl of chloroform was then added into the sample mixture.

The sample mixture was then covered tightly and shaken vigorously for 15

second, followed by incubation at RT for 15 minutes. The mixture was then

centrifuged at 12, 000g (Centrifuge 5417R, Eppendorf, USA) for 20 minutes at

Page 123: Thesis

93

4°C which separated the sample into three phases: the upper aqueous phase,

whitish interphase and the lower red phenol-chloroform phase. Only the upper

aqueous phase was pippetted slowly and genteelly into a new eppendorf tube

and 800 µl of isopropanol was added and left to stand in RT for 20 minutes. The

sample was then centrifuged at 12, 000g for 15 minutes at 4°C to pellet the

dsRNA. The supernatant was removed and the dsRNA pellet was then

resuspended in 1 mL of 75% ethanol and followed by centrifugation at 12, 000g

for 15 minutes at 4°C. The supernatant was discarded and 1 mL of 100%

ethanol was added to the pellet, re-suspended and centrifuged at 12, 000g for

15 minutes at 4°C. The pellet was then air dry. The pellet was then dissolved in

10 µl diethlpyrocarbonate (DEPC) treated deionized double-distilled water

(ddH2O). The RNA solution if not used immediately was kept in –80°C (Wu and

Lin, 1992).

4.2.3 Determination of RNA Concentration

The concentration and purity of extracted RNA was determined by using

spectrophotometer according to the method described by Sambrook et al.

(1989). The optical density (OD) of RNA was measured at its maximum

absorbance wavelength of 260nm. Once the OD unit was equivalent to 40 µl/mL

of dsRNA, the extracted RNA was read at wavelength of 260nm and 280

whereby the ratio of 260nm to 280nm estimates the purity of RNA. The

acceptable absorbance ratio of 260/280OD was in the range of 1.8-2.0.

Page 124: Thesis

94

4.2.4 Primer Design

One set primers designated as P1 and P2 (Liu et al., 1994) were used for RT-

PCR amplification.The P1 and P2 primers were used for amplification of the

hypervariable region of VP2 gene of the IBDV isolates as shown in (Table 4.1).

Table4.1: Primers used to amplify the HPVR VP2 gene

Primer Sequences Position (5‟ to 3‟)a

Product

(size)

P1

TCA CCG TCC TCA GCT TAC

622-639

HPVR

(643 bp) P2

TCA GGA TTT GGG ATC AGC

1247-1264

4.2.5 Reverse Transcription and PCR Reaction

The following protocol was according to Fiza et al. (2006). The genome of IBDV

was denatured by adding (1 µl) dimethyl sulfoxide (DMSO), reverse and forward

primers (1 µl) and (4 µl) DEPC water in heating block for 5 minutes at 95°C and

immediately placed on ice for 2-3 minutes. Reverse transcriptase was carried

out at 42°C for 1 hour in a total of 20 µl volume containing 9 µl of denatured

RNA, 4 µl MgCl2, 5 unit of AMV, 4 µl of reaction buffer, 2 µl of dNTP, and 0.5 µl

Rnasin. After heating to 99°C for I minute, 5 µl of cDNA was added into PCR

reaction. This mixture contained 1 µl of 10mM dNTP, 4 µl of MgCl2, 6 µl of PCR

Page 125: Thesis

95

buffer, (1µl) Reverse and Forward Primer, 5 µl cDNA, (31.5 µl) water and (0.5

µl) Taq Polymerase. PCR was performed in the thermocycler (MJ, BioRad,

USA) using the following temperature profile for 35 cycle 94°C for 1 minute,

52°C for 1 minute, 72°C for 2 minutes and then the reaction was allowed to go

at 72°C for 10 minutes.

4.2.6 Gel Electrophoresis and Ethidium Bromide Staining

The following protocol was according to Fiza et al. (2006). Agarose gel

electrophoresis was done using a 7 x 10 cm Mini-Sub cell GT gel

electrophoresis apparatus (Bio-Rad, USA). The 1% agarose gel was prepared

by adding 1g of agarose powder in 100 mL TAE (Tris-acetate-EDTA) buffer

(Appendix A) and boiled until the powder dissolved. The agarose solution was

cooled down to about 60°C before pouring onto a mini gel tray that had been

placed with an 8 wells comb and bordered with the gel casting gates within the

gel electrophoresis apparatus. The gel was allowed to solidify at room

temperature and gently the comb and gates were removed. TAE buffer was

pour onto the electrophoresis apparatus and submerged the solidified gel. DNA

ladder (100bp) (Promega, USA) was used as DNA marker. The DNA ladder

marker was prepared by mixing 10 µl of marker and 2 µl of 6x loading dye

solution. Other wells were loaded with 10 µl of RT-PCR products that had been

pre-mixed with 2 µl of loading dye. Gel electrophoresis was run at 80V for 35

min.

Page 126: Thesis

96

After electrophoresis was completed, the gel was transferred from the

electrophoresis apparatus and submerged into ethidium bromide staining

solution for 15-30 minutes. The gel was de-stained with distiled water for 15

minutes and placed onto the UV transluminator to visualize the nucleic acids

bands.

4.2.7 Purification of RT-PCR Products

Gel purification system from GeneAll® (GeneAll®, Seoul, Korea) or “Gel SV”

(GelSV®, Seoul, Korea) was used to extract the DNA from the gel slice. In order

to obtain good quality of DNA, the handling time on the transilluminator was kept

to the minimum. The purification of the PCR products from the gel was

performed according to the manufacturers‟ protocol (GeneAll®, Seoul, Korea).

The gel slice was transferred to a new 1.5 mL tube and weighed. Three volumes

(µl) of GB buffer were added for each volume (mg) of gel and the mixture was

incubated in a 50°C water bath until the gel melted. The tube was vortexed

every 3 minutes to facilitate the dissolution of gel. The mixture was transferred

on to a SV centrifuge column and was centrifuged for 1 minute after which the

pass-through was discarded. Then five hundred µl of GB buffer was added to

the column and centrifuged for 30 seconds and the pass-through discarded.

Seven hundred µl of NW buffer was added and centrifuged twice for 30 seconds

and discarded. The column was then transferred to a new tube and 30 µl of EB

buffer was added to the column; the column was left to stand on the tube for 1

Page 127: Thesis

97

minute and then centrifuged for 1.5 minutes. The tube containing the desired

DNA was stored at -80°C.

4.2.8 Molecular Cloning of Amplified Products and Analysis of Recombinant Plasmid

The PCR products were directly cloned into PCR®2.1-TOPO® vector using the

TOPO TA Cloning ® kit (Invitrogen, USA) by referring to its protocol. TOPO®

cloning reaction solution was set up by mixing 4 µl of fresh PCR product with 1

µl of salt solution and 1 µl of TOPO® vector. The reaction solution was gently

mixed, incubated for 5 minutes at room temperature and placed on ice. An

aliquot of 2 µl of the reaction solution was mixed with a vial of thawed One

Shot® Escherichia coli cells and incubated on ice for 5 minutes. The cells were

heat-shocked at 42ºC for 30 seconds without shaking and then immediately

incubated on ice for 1 minute. The cells were added with 250 µl of LB broth and

shook horizontally (200 rpm) at 37ºC for 1 hour. The transformed cells were

spread on Ampicillin-added selective LB plates in a volume of 10-50 µl per plate.

The plates were then incubated at 37ºC in inverted position for at least 12 hours

to allow the transformed cells to grow.

Based on blue white selection, 10 white colonies were picked and subcultured

onto another selective LB plates. At the same time, these colonies were also

being screened for positive transformation using PCR. Each PCR reaction

mixture consisted of 1 µl of 10x PCR buffer, 1µl of 25 Mm MgCl2, 1µl of 10dNTP,

Page 128: Thesis

98

0.5 µl of each P1 and P2 primers, 0.5 µl of Taq polymerase, 6.5 µl of ddH2O,

and a dot of the picked colony. The PCR program was set to 94ºC for 3 minutes

(for 1 cycle) and followed by 35 cycles of the following: 94ºC for 1 minute, 48ºC

for 1 minute and 72ºC for 10 minutes. Positive clone was identified by the

presence of band of interest after gel electrophoresis. A dot of the positive clone

was added into 4 mL of ampicillin-added LB broth and incubating in 37ºC with

horizontal shaking 200 rpm for about 12 hours. The culture clone, if not used

immediately was mixed with sterial glycerol (0.85 mL culture to 0.15 mL

glycerol) and stored at -80 ºC.

4.2.9 Plasmid Extraction and Purification

Plasmids from the positive clones were extracted and purified using Gene All

Kit® (Fragment, SV, Seoul, Korea) following manufacturer recommendations.

Briefly, the overnight culture of positive clone was first pelleted and all medium

was removed. The pellet was re-suspended in 250 µl of RNase-added cell

suspension buffer (Buffer S1) until homogenous. This was followed by adding

250 µl of cell lysis solution (Buffer S2) and gently mixed by inverting the capped

tube five times and incubated at RT for 5 minutes. Immediately after the

incubation 350 µl of neutralization buffer (Buffer S3) was added and the mixture

was centrifuged at 12, 000 g for 10 minutes. The supernatant was loaded onto a

spin column and centrifuged at 12, 000 g for 1 minute. The flow-through was

discharged. The cartridge was washed by adding 700 µl of wash buffer (Buffer

Page 129: Thesis

99

PW) onto the cartridge and centrifuged at 12, 000 g for 1 min. The cartridge was

re-centrifuged to remove residual wash buffer. The plasmid was eluted from the

cartridge by adding 50 µl of EB buffer, incubated at room temperature for 1

minute and centrifuged at 12, 000 g for 1 min. The purity and concentration of

the plasmid was determined using spectrophotometry and the plasmid was

stored at -20oC if not used immediately.

4.2.10 Sequence Assembly and Analysis Using Bioinformatics Software

The sequencing data were initially aligned and processed by using the basic

BLAST (Basic Local Alignment Search Tool) search programme of National

Center for Biotechnology Information (NCBI)

(http://www.ncbi.nim.nih.gov/BLAST). Sequence editing, multiple alignments

and analysis were done by using bioinformatics software: BioEdit version 7.0.4,

GeneDoc version 2.6.002 (Nicholas et al., 1997), and Clustal-XTM version 1.83

(Thompson et al., 1997). Twenty four sequences were downloaded from

GenBank and compared with UPM0081, UPM0081T10, UPM0081T15 and

UPM0081T20 (Table 4.2).

Page 130: Thesis

100

Table 4. 2: IBDV isolates used in the sequence analyses

Isolate

Strain

A

Accession

Country

B

References

UPM0081

VV

AY520910

Malaysia

Tan et al., 2004

UPM94268 VV AY333088 Malaysia Tan et al., 2004

UPM92/04 VV AF262030 Malaysia Hoque et al., 2001

UPM4/230 VV AY520911 Malaysia Tan et al., 2004

UPM94/274 VV AF527039 Malaysia Phong et al., 2003

B00/73 VV AY520909 Malaysia Tan et al., 2004

UK661 VV X92760 United Kingdom Brown and Skinner, 1996 Tasek94 VV AF322444 Indonesia Rudd et al., 2002

OKYM VV D49706 Japan Yamaguchi et al.,1996

HK-46 VV AF051838 Hong Kong Cao et al., 1998

Ehime 91 VV AB024076 Japan Tsukamoto et al., 1999

SH 95 VV AY 134874 China Sun et al., 2003

SH 92 VV AF533670 Korea Kim et al., 2004

CT CA AJ310158 France Lejal et al., 2000

Kal2001 CA AY311479 Egypt EL-Zeedt et al., 2003

GLS VA M97346 USA Vakharia et al., 1994

Variant A VA M64285 USA Lana et al., 1992

E/DEL VA X54858 USA Vakharia et al., 1992

UPM0081T10 AT* FJ824699 Malaysia Mohammed et al., 2009

UPM0081T15 AT* FJ898322 Malaysia Mohammed et al., 2009

UPM0081T20 AT* FJ898321 Malaysia Mohammed et al., 2009

Soroa AT AF140705 Cuba Femandez-Arias et al.,1988

OKYMT AT AJ427340 Japan Yamaguchi et al., 1996

PBG98 AT D00868 United Kingdom Spies et al., 1989

D78 AT AF499929 USA Hudson et al., 2003

AmerVH9907 AT AJ577092 Vietnam Ghazi et al., 2003

OH Serotype 2 M66722 USA Kibenge et al., 2008

Note: Strain represented IBDV strains: VV = very virulent; CA = classical; VA = variant; AT = attenuated. Country was the place of isolation. AT* indicated the strain identity as determined in this study.

Page 131: Thesis

101

4.2.11 Phylogenetic Tree Construction

Phylogenetic tree was constructed based on the hypervariable region of the

nucleotide sequence from position 516 to 1058 (Bayliss et al., 1990) and

deduced amino acid from 173 to 386 of the VP2 gene. Twenty four published

IBDV isolates were included in this study to investigate the evolutionary

relationships among the isolates. Sequences were aligned using ClustalX soft

were version 1.83. Translation to amino acid sequences were done using

biology work bench (version 3.2). The tree views were edited using Tree View X

version (http://evolgen.biol.metrou.ac.ip/TE/TE_man.html).

4.3 Results 4.3.1 Amplification of the hypervariable Region of VP2 Gene

The dsRNA from the 5, 7, 8, 9, 10, 15 and 20 passages of UPM0081 vvIBDV in

Vero cells namely as UPM0081T5, UPM0081T7, UPM0081T8, UPM0081T9,

UPM0081T10, UPM0081T15, and UPM0081T20 respectively and from the 5

and 7 passages in DF-1 cells namely as UPM0081D5 and UPM0081D7,

respectively were extracted and synthesized to cDNA. The hypervariable region

of VP2 gene was successfully amplified. All the amplified cDNA showed

identical mobility on a 1% agarose gel. The PCR amplification was done by

using primer P1-P2 generate specific DNA band of 643 bp (Figures, 4.1 and

4.2).

Page 132: Thesis

102

4.3.2 PCR Analysis of Recombinant Colonies

The amplified VP2 from the UPM0081T5, UPM0081T7, UPM0081T8,

UPM0081T9, UPM0081T10, UPM00815 and UPM0081T20 passages in Vero

cells and UPM0081D5 and UPM0081D7 passages in DF-1 cells were

successfully cloned in the PCR 2.1- TOPO TA vector. The plasmids were

screened for insertion using the PCR technique from the white colonies.

Expected size of the inserted VP2 gene (643 bp) was identified from the white

colonies (Figures, 4.3 and 4.4).

4.3.3 Nucleotide Sequence Analysis

To determine the genetic changes, the VP2 variable region sequences of IBDV

passaged in Vero cells namely as UPM0081T5, UPM008T7, UPM0081T8,

UPM0081T9, UPM0081T10, UPM0081T15, and UPM0081T20 and two

passaged in DF-1 cells UPM0081D5 and UPM0081D7 from 516 to 1158 as

described by Bayliss et al. (1990) were analysed and aligned with their parental

strain and published various IBDV strains (Table 4.2). There was no insertion or

deletion of nucleotide sequence for the seventh passages strain in Vero cells

and two passages in DF-1 cells when compared with published sequences used

in this study: vvIBDV (UPM94268, UPM92/04, UPM4/230, UPM94/273, B00/73,

UK661, Tasek.94. OKYM, HK46, Ehime 91, SH/95, SH/92), caIBDV (CT,

KAL2001), variant IBDV (GLS, Variant A, E//DEL), attenuated IBDV (Soroa,

Page 133: Thesis

103

OKYMT. PBG98, D78, AmerVH9907), and a virulent serotype 2 IBDV (OH)

(Figure, 4.5).

Sequence analysis of selected passages showed that there were no changes in

the nucleotide sequence in two passages (UPM008T5 and UPM008T7) in Vero

cells and UPM008D5 andUPM008D7 in DF-1 cells. The first changes was

observed in passage 8 (UPM0081T8) at nucleotide position 585, 774 and 1056

from A, T, and C to C, C, and G, respectively. In passage 9 (UPM0081T9) eight

nucleotides changes at position 585, 664, 696, 774, 489, 990, 1056, and 1083

from A, G, T, T, A, T, A, and T to C, C, G, C, G, A, G, and C, respectively. A

total of 29 nucleotides mutation were detected in passages 10, 15 and 20

(UPM0081T10, UPM0081T15, and UPM0081T20) from A, C, A, G, T, T, C, A,

C, A, A, A, A, T, T, A, G, A, G, A, C, C, A, T, C, A, A, T and C to G, T, C, C, G,

C, T, G, T, G, G, C, G, C, C, G, A, G, A, C, T, T, G, A, T, G, G, C, and T at

nucleotide position of 537, 540, 585, 664, 696, 699, 702, 724, 726, 735, 746,

759, 766, 774, 777, 822, 835, 846, 850, 880, 888, 977, 948, 990, 1014, 1050,

1056, 1083, and 1052, respectively.

The differences of nucleotides with IBDV strain are as shown in Table 4.3. The

number of nucleotide difference in the hypervariable region of VP2 gene for

UPM0081T10, UPM0081T15 and UPM0081T20 IBDV isolates were 29 to 36

with vvIBDV strains, 13 to 14 with caIBDV strain, 27 to 32 with vaIBDV strains

and 13-17 with atIBDV strains. The degree of identity nucleotide for

Page 134: Thesis

104

UPM0081T10, UPM0081T15 and UPM0081T20 showed highest nucleotide

homology to atIBDV strains and classical strains (97%). The degree of identity

was down to 94 to 95% when compared with vvIBDV, and vaIBDV strains

(Table 4.4, Figures, 4.9 and 4.10). Meanwhile, the number of nucleotide

difference in the hypervariable region of VP2 gene for very virulent parental

strain UPM0081 was 1 to 11 with vvIBDV, 39 to 40 with caIBDV strains, 39 with

vaIBDV strains, and 29 to 42 with atIBDV (Table 4.3). The degree of identity

nucleotide for UPM0081 was greatest with vvIBDV strains (98 to 99%). Degree

of identity was down to 93 to 95% when compared with classical strains, variant

strains and attenuated strains (Table 4.4).

4.3.4 Amino Acid Sequence Analysis

The 214 amino acids that resides between 173-386 of the segment A of the

hypervariable regions of VP2 genes were aligned with published IBDV strains

(Figure 4.6). Both passages (UPM008T5 and UPM008T7) in Vero cells and DF-

1 cells (UPM008D5 and UPM008D7) had the same amino acid when compared

with parental strain (UPM0081) and vvIBDV strains (UPM94268, UPM92/04,

UPM4/230, UPM94/273, B00/73, UK661, Tasek.94. OKYM, HK46, Ehime 91,

SH/95, SH/92). The following amino acids were found at these positions: Ala

[222], IIe [242], Gln [253], IIe [256], Ala [284], IIe [294] and Ser [299] in the

HPVR of VP2 gene of theses isolates (Table 4.5).

Page 135: Thesis

105

The number of amino acid difference in the hypervariable region of VP2 gene

for UPM0081T10, UPM0081T15 and UPM0081T20 IBDV isolates were 10 to 13

with vvIBDV, 4 to 5 with caIBDV, 9 to 13 with vaIBDV and 4 to 7 with atIBDV

(Table 4.6). The degree of identity amino acid for UPM0081T10, UPM0081T15

and UPM0081T20 showed highest nucleotide homology to atIBDV strains 96 to

98%, while the degree of identity was down to 93 to 95% when compared with

very virulent strains (Table 4.7, Figures, 4.11 and 4.12).

Meanwhile the number of amino acid difference in the hypervariable region of

VP2 gene for very virulent parental strain UPM0081 was 1 to 3 with vvIBDV, 11

to 12 with caIBDV strains, 11 to 13 with vaIBDV strains and 11 to 14 with

atIBDV (Table 4.6). The degree of identity of amino acid for UPM0081 was

greatest with vvIBDV strains (98 to 99%). The degree of identity was down to 94

to 95% when compared with attenuated strains (Table 4.7).

4.3.5 Phylogenetic Analysis

The overall grouping and branching of the phylogenetic tree showed two main

group base amino acid and nucleotide (Figures 4.7 and 4.8). The group one

showed the serotype 2 strain (OH) while the other group was that of very

virulent, attenuated, classical and variant strains of serotype 1. The branches of

serotype 1 contained three subgroups; UPM0081, UPM94268, UPM92/04,

UPM4/230, UPM94/273, B00/73, UK661, Tasek.94. OKYM, HK46, Ehime 91,

Page 136: Thesis

106

SH/95 as well as SH/92 which belong to the vvIBDV subgroup 1 branch;

UPM0081T10, UPM0081T15, and UPM0081T20 which were more closely

related to another group that had Soroa, OKYMT. PBG98, D78 and Amer

VH9907 isolates which are regarded as typical attenuated and ca IBDV strains,

such as CT and KAL2001 subgroup 2 branch, and finally subgroup 3 branch

which consists of vaIBDV strain such as GLS, Variant A and E/DEL.

Page 137: Thesis

107

Figure 4.1: Hypervariable region (643pb) of IBDV VP2 genes Lane 1- Negative control; Lane 2 positive UPM0081D5; Lane 3 positive UPM0081D7; Lane 4 positive UPM0081T5 and Lane 5 positive UPM0081T7; M- 100 bp DNA marker (Promega, USA).

Figure 4.2: Hypervariable region (643pb) of IBDV VP2 genes Lane 1- positive UPM0081T8; Lane 2 positive UPM0081T9; Lane 3 positive UPM0081T10; Lane 4 positive UPM0081T15 and Lane 5 positive UPM0081T20; Lane 6- Negative control; M- 100 bp DNA marker (Promega, USA).

Page 138: Thesis

108

Figure 4.3: PCR screening on white colonies amplification of IBDV genes Lane 1, 2 and 3 white colonies positive for VP2 gene passages (UPM0081D5, UPM0081T5 and UPM0081T7 respectively; Lane 4 Negative control; M- 100 bp DNA marker (Promega, USA).

Figure 4.4: PCR screening on white colonies amplification of IBDV genes. Lane 1,2,3,4,5,6 and 7 white colonies positive for VP2 gene passages (UPM0081D7, UPM0081T8, UPM0081T9, UPM0081T10, UPM0081T15 and UPM0081T20 respectively; Lane 7 Negative control; M- 100 bp DNA marker (Promega, USA).

Page 139: Thesis

109

Table 4.3: Number of nucleotide differences in HPVR of VP2 gene between IBDV isolates Passage Isolate Strain No.

Diff Parent isolate

Isolate Strain N. Diff

UPM0081T20

vs.

UPM0081

VV

29

UPM0081

vs.

UPM0081

VV

ID

UPM0081T20 vs. UPM94268 VV 30 UPM0081 vs. UPM94268 VV 1 UPM0081T20 vs. UPM92/04 VV 30 UPM0081 vs. UPM92/04 VV 1 UPM0081T20 vs. UPM4/230 VV 33 UPM0081 vs. UPM4/230 VV 4 UPM0081T20 vs. UPM94/274 VV 31 UPM0081 vs. UPM94/274 VV 4 UPM0081T20 vs. B00/73 VV 36 UPM0081 vs. B00/73 VV 7 UPM0081T20 vs. UK661 VV 31 UPM0081 vs. UK661 VV 6 UPM0081T20 vs. Task94 VV 35 UPM0081 vs. Task94 VV 9 UPM0081T20 vs. OKYM VV 33 UPM0081 vs. OKYM VV 6 UPM0081T20 vs. HK-46 VV 34 UPM0081 vs. HK-46 VV 7 UPM0081T20 vs. Ehime 91 VV 34 UPM0081 vs. Ehime 91 VV 7 UPM0081T20 vs. SH 95 VV 34 UPM0081 vs. SH 95 VV 11 UPM0081T20 vs. SH 92 VV 30 UPM0081 vs. SH 92 VV 5 UPM0081T20 vs. CT CA 13 UPM0081 vs. CT CA 40 UPM0081T20 vs. Kal2001 CA 14 UPM0081 vs. Kal2001 CA 39 UPM0081T20 vs. GLS VA 27 UPM0081 vs. GLS VA 39 UPM0081T20 vs. Variant A VA 32 UPM0081 vs. Variant A VA 39 UPM0081T20 vs. E/DEL VA 29 UPM0081 vs. E/DEL VA 39 UPM0081T20 vs. UPM0081T10 AT 0 UPM0081 vs. UPM0081T10 AT 29 UPM0081T20 vs. UPM0081T15 AT 0 UPM0081 vs. UPM0081T15 AT 29 UPM0081T20 vs. UPM0081T20 AT 0 UPM0081 vs. UPM0081T20 AT 29 UPM0081T20 vs. Soroa AT 13 UPM0081 vs. Soroa AT 40 UPM0081T20 vs. OKYMT AT 17 UPM0081 vs. OKYMT AT 42 UPM0081T20 vs. PBG98 AT 13 UPM0081 vs. PBG98 AT 42 UPM0081T20 vs. D78 AT 15 UPM0081 vs. D78 AT 39 UPM0081T20 vs. AmerVH9907 AT 15 UPM0081 vs. AmerVH9907 AT 40 UPM0081T20 vs. OH Serotype

2 157 UPM0081 vs. OH Serotype

2 168

Note:“No. Diff” is the nucleotide differences between passage (UPM0081T20) and parent isolate (UPM0081). All the three passages (UPM0081T10, UPM0081T15 and UPM0081T20) were similar and closely related to atIBDV publish gene bank by having 13-17 nucleotides difference, while parent isolate having high nucleotides difference 29-42 nucleotides. Strain represented IBDV strains: VV = very virulent; CA = classical; VA = variant; AT = attenuated

Page 140: Thesis

110

Table 4.4: Sequence identity matrix of VP2 genes nucleotides of IBDV isolates

Passage Isolate Strain Seq. Iden

Parent isolate

Isolate Strain Seq. Iden

UPM0081T20

vs.

UPM0081

VV

0.954

UPM0081

vs.

UPM0081

VV

ID

UPM0081T20 vs. UPM94268 VV 0.953 UPM0081 vs. UPM94268 VV 0.998 UPM0081T20 vs. UPM92/04 VV 0.953 UPM0081 vs. UPM92/04 VV 0.998 UPM0081T20 vs. UPM4/230 VV 0.948 UPM0081 vs. UPM4/230 VV 0.993 UPM0081T20 vs. UPM94/273 VV 0.951 UPM0081 vs. UPM94/273 VV 0.993 UPM0081T20 vs. B00/73 VV 0.949 UPM0081 vs. B00/73 VV 0.989 UPM0081T20 vs. UK661 VV 0.951 UPM0081 vs. UK661 VV 0.990 UPM0081T20 vs. Task94 VV 0.954 UPM0081 vs. Task94 VV 0.986 UPM0081T20 vs. OKYM VV 0.948 UPM0081 vs. OKYM VV 0.990 UPM0081T20 vs. HK-46 VV 0.947 UPM0081 vs. HK-46 VV 0.989 UPM0081T20 vs. Ehime 91 VV 0.947 UPM0081 vs. Ehime 91 VV 0.982 UPM0081T20 vs. SH 95 VV 0.947 UPM0081 vs. SH 95 VV 0.982 UPM0081T20 vs. SH 92 VV 0.953 UPM0081 vs. SH 92 VV 0.992 UPM0081T20 vs. CT CA 0.979 UPM0081 vs. CT CA 0.937 UPM0081T20 vs. Kal2001 CA 0.978 UPM0081 vs. Kal2001 CA 0.939 UPM0081T20 vs. GLS VA 0.958 UPM0081 vs. GLS VA 0.939 UPM0081T20 vs. Variant A VA 0.950 UPM0081 vs. Variant A VA 0.939 UPM0081T20 vs. E/DEL VA 0.954 UPM0081 vs. E/DEL VA 0.939 UPM0081T20 vs. UPM0081T10 AT 1.000 UPM0081 vs. UPM0081T10 AT 0.959 UPM0081T20 vs. UPM0081T15 AT 1.000 UPM0081 vs. UPM0081T15 AT 0.959 UPM0081T20 vs. UPM0081T20 AT ID UPM0081 vs. UPM0081T20 AT 0.959 UPM0081T20 vs. Soroa AT 0.979 UPM0081 vs. Soroa AT 0.939 UPM0081T20 vs. OKYMT AT 0.973 UPM0081 vs. OKYMT AT 0.943 UPM0081T20 vs. PBG98 AT 0.979 UPM0081 vs. PBG98 AT 0.943 UPM0081T20 vs. D78 AT 0.976 UPM0081 vs. D78 AT 0.939 UPM0081T20 vs. AmerVH9907 AT 0.976 UPM0081 vs. AmerVH9907 AT 0.937 UPM0081T20 vs. OH Seroty

pe 2 0.736 UPM0081 vs. OH Serot

ype 2 0.739

Note: “Seq. Iden” is the sequence nucleotide identity matrix between passage (UPM0081T20) and parent isolate (UPM0081). All the three passages (UPM0081T10, UPM0081T15 and UPM0081T20) were similar and closely related to atIBDV publish gene bank by having 97% sequence identity matrix, while parent isolate having low sequence identity matrix 93-95%. Strain represented IBDV strains: VV = very virulent; CA = classical; VA = variant; AT = attenuated

Page 141: Thesis

111

Table 4.5: Summary of the proposed molecular markers (amino acid residues) of UPM0081T10, UPM0081T15 and UPM0081T20 atIBDV isolates with other published IBDV strains. IBD Isolate

Strain

222

242

249

253

256

279

284

294

326

330

UPM0081

VV

A

I

Q

Q

I

D

A

I

S

S

UPM94268 VV A I Q Q I D A I S S UPM92/04 VV A I Q Q I D A I S S UPM4/230 VV A I Q Q I D A I S S UPM94/274 VV A I Q Q I D A I S S B00/73 VV A I Q Q I D A I S S UK661 VV A I Q Q I D A I S S Task94 VV A I Q Q I D A I S S OKYM VV A I Q Q I D A I S S HK-46 VV A I Q Q I D A I S S Ehime 91 VV A I Q Q I D A I S S SH 95 VV A I Q Q I D A I S S SH 92 VV A I Q Q I N A I S S CT CA P V Q H V N T L S R Kal2001 CA P V R H V N T L S R GLS VA T V K H V N T L S S Variant A VA Q V K Q V N A L S S E/DEL VA T V K Q V N A L S S UPM0081T10 AT P V R H V N T L L R UPM0081T15 AT P V R H V N T L L R UPM0081T20 AT P V R H V N T L L R Soroa AT P V R H V N T L S R OKYMT AT P V Q H V N T L S K PBG98 AT P V R H V N T L S R D78 AT P V Q H V N T L L R AmerVH9907 AT P V Q H V N T L S R OH Serotype2 P V S I I T T N I V

Note: The proposed molecular markers for atIBDV which were Pro [222], Val [242], Arg [249], His [253], Val [256], Asn [279], Thr [284], Leu [294], Leu [326] and Arg [330] were found in the HPVR of VP2 gene of these three isolates (Kwon and Kim, 2004; Wang, et al., 2004). Strain represented IBDV strains: VV = very virulent; CA = classical; VA = variant; AT = attenuated

Page 142: Thesis

112

Table 4.6: Number of amino acids differences in HPVR of VP2 gene between IBDV isolates

Passage Isolate Strain No. Diff

Parent isolate

Isolate Strain N. Diff

UPM0081T20

vs.

UPM0081

VV

10

UPM0081

vs.

UPM0081

VV

ID

UPM0081T20 vs. UPM94268 VV 11 UPM0081 vs. UPM94268 VV 1 UPM0081T20 vs. UPM92/04 VV 11 UPM0081 vs. UPM92/04 VV 1 UPM0081T20 vs. UPM4/230 VV 13 UPM0081 vs. UPM4/230 VV 3 UPM0081T20 vs. UPM94/273 VV 13 UPM0081 vs. UPM94/273 VV 3 UPM0081T20 vs. B00/73 VV 12 UPM0081 vs. B00/73 VV 2 UPM0081T20 vs. UK661 VV 11 UPM0081 vs. UK661 VV 1 UPM0081T20 vs. Task94 VV 12 UPM0081 vs. Task94 VV 3 UPM0081T20 vs. OKYM VV 11 UPM0081 vs. OKYM VV 1 UPM0081T20 vs. HK-46 VV 11 UPM0081 vs. HK-46 VV 1 UPM0081T20 vs. Ehime 91 VV 11 UPM0081 vs. Ehime 91 VV 1 UPM0081T20 vs. SH 95 VV 12 UPM0081 vs. SH 95 VV 2 UPM0081T20 vs. SH 92 VV 10 UPM0081 vs. SH 92 VV 2 UPM0081T20 vs. CT CA 4 UPM0081 vs. CT CA 12 UPM0081T20 vs. Kal2001 CA 5 UPM0081 vs. Kal2001 CA 11 UPM0081T20 vs. GLS VA 9 UPM0081 vs. GLS VA 13 UPM0081T20 vs. Variant A VA 11 UPM0081 vs. Variant A VA 11 UPM0081T20 vs. E/DEL VA 13 UPM0081 vs. E/DEL VA 13 UPM0081T20 vs. UPM0081T10 AT 0 UPM0081 vs. UPM0081T10 AT 10 UPM0081T20 vs. UPM0081T15 AT 0 UPM0081 vs. UPM0081T15 AT 10 UPM0081T20 vs. UPM0081T20 AT ID UPM0081 vs. UPM0081T20 AT 10 UPM0081T20 vs. Soroa AT 4 UPM0081 vs. Soroa AT 12 UPM0081T20 vs. OKYMT AT 7 UPM0081 vs. OKYMT AT 12 UPM0081T20 vs. PBG98 AT 4 UPM0081 vs. PBG98 AT 14 UPM0081T20 vs. D78 AT 5 UPM0081 vs. D78 AT 11 UPM0081T20 vs. AmerVH9907 AT 6 UPM0081 vs. AmerVH9907 AT 12 UPM0081T20 vs. OH Serotype

2 145 UPM0081 vs. OH Serotype

2 147

Note:“ No. Diff” is the amino acid differences between passage (UPM0081T20) and parant strain (UPM0081). All the three passages (UPM0081T10, UPM0081T15 and UPM0081T20) were similar and closely related to atIBDV publish gene bank by having 4-7 amino acid difference, while parent isolate had high amino acid difference 10-14 amino acid. Strain represented IBDV strains: VV = very virulent; CA = classical; VA = variant; AT = attenuated

Page 143: Thesis

113

Table 4.7: Sequence identity matrix of VP2 genes amino acids of IBDV isolates

Passage Isolate Strain Seq. Iden

Parent isolate

Isolate Strain Seq. Iden

UPM0081T20

vs.

UPM0081

VV

0.953

UPM0081

vs.

B0081

VV

ID

UPM0081T20 vs. UPM94268 VV 0.948 UPM0081 vs. UPM94268 VV 0.995 UPM0081T20 vs. UPM92/04 VV 0.948 UPM0081 vs. UPM92/04 VV 0.995 UPM0081T20 vs. UPM4/230 VV 0.939 UPM0081 vs. UPM4/230 VV 0.985 UPM0081T20 vs. UPM94/273 VV 0.939 UPM0081 vs. UPM94/273 VV 0.985 UPM0081T20 vs. B00/73 VV 0.943 UPM0081 vs. B00/73 VV 0.990 UPM0081T20 vs. UK661 VV 0.948 UPM0081 vs. UK661 VV 0.995 UPM0081T20 vs. Task94 VV 0.943 UPM0081 vs. Task94 VV 0.985 UPM0081T20 vs. OKYM VV 0.948 UPM0081 vs. OKYM VV 0.995 UPM0081T20 vs. HK-46 VV 0.948 UPM0081 vs. HK-46 VV 0.995 UPM0081T20 vs. Ehime 91 VV 0.948 UPM0081 vs. Ehime 91 VV 0.995 UPM0081T20 vs. SH 95 VV 0.943 UPM0081 vs. SH 95 VV 0.990 UPM0081T20 vs. SH 92 VV 0.953 UPM0081 vs. SH 92 VV 0.990 UPM0081T20 vs. CT CA 0.981 UPM0081 vs. CT CA 0.943 UPM0081T20 vs. Kal2001 CA 0.976 UPM0081 vs. Kal2001 CA 0.948 UPM0081T20 vs. GLS VA 0.957 UPM0081 vs. GLS VA 0.939 UPM0081T20 vs. Variant A VA 0.948 UPM0081 vs. Variant A VA 0.948 UPM0081T20 vs. E/DEL VA 0.939 UPM0081 vs. E/DEL VA 0.939 UPM0081T20 vs. UPM0081T10 AT 1.000 UPM0081 vs. UPM0081T10 AT 0.953 UPM0081T20 vs. UPM0081T15 AT 1.000 UPM0081 vs. UPM0081T15 AT 0.953 UPM0081T20 vs. UPM0081T20 AT ID UPM0081 vs. UPM0081T20 AT 0.953 UPM0081T20 vs. Soroa AT 0.981 UPM0081 vs. Soroa AT 0.943 UPM0081T20 vs. OKYMT AT 0.967 UPM0081 vs. OKYMT AT 0.943 UPM0081T20 vs. PBG98 AT 0.981 UPM0081 vs. PBG98 AT 0.943 UPM0081T20 vs. D78 AT 0.967 UPM0081 vs. D78 AT 0.948 UPM0081T20 vs. AmerVH9907 AT 0.971 UPM0081 vs. AmerVH9907 AT 0.943 UPM0081T20 vs. OH Serotype

2 0.325 UPM0081 vs. OH Serotype

2 0.326

Note: “Seq. Iden” is the sequence amino acid identity matrix between passage (UPM0081T20) and parent isolate (UPM0081). All the three passages (UPM0081T10, UPM0081T15 and UPM0081T20) were similar and closely related to atIBDV publish gene bank by having 96-98% sequence identity matrix, while parent isolate having low sequence identity matrix 94-95%. Strain represented IBDV strains: VV = very virulent; CA = classical; VA = variant; AT = attenuated

Page 144: Thesis

114

Figure 4.5: Nucleotide sequences of HPVR of VP2 from nucleotide 516-1158 (numbering of Bayliss et al., 1999) of the UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages compared with other published IBDV strains. A dot indicated position where the sequence is identical to others.

Page 145: Thesis

115

Figure 4.5: Nucleotide sequences of HPVR of VP2 from nucleotide 516-1158 (numbering of Bayliss et al., 1999) of the UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages compared with other published IBDV strains. A dot indicated position where the sequence is identical to others. .

Page 146: Thesis

116

Figure 4.5: Nucleotide sequences of HPVR of VP2 from nucleotide 516-1158 (numbering of Bayliss et al., 1999) of the UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages compared with other published IBDV strains. A dot indicated position where the sequence is identical to others.

Page 147: Thesis

117

Figure 4.5: Nucleotide sequences of HPVR of VP2 from nucleotide 516-1158 (numbering of Bayliss et al., 1999) of the UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages compared with other published IBDV strains. A dot indicated position where the sequence is identical to others.

Page 148: Thesis

118

Figure 4.5: Nucleotide sequences of HPVR of VP2 from nucleotide 516-1158 (numbering of Bayliss et al., 1999) of the UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages compared with other published IBDV strains. A dot indicated position where the sequence is identical to others.

Page 149: Thesis

119

Figure 4.5: Nucleotide sequences of HPVR of VP2 from nucleotide 516-1158 (numbering of Bayliss et al., 1999) of the UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages compared with other published IBDV strains. A dot indicated position where the sequence is identical to others.

Page 150: Thesis

120

Figure 4.5: Nucleotide sequences of HPVR of VP2 from nucleotide 516-1158 (numbering of Bayliss et al., 1999) of the UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages compared with other published IBDV strains. A dot indicated position where the sequence is identical to others.

Page 151: Thesis

121

Figure 4.6: Amino acid sequence aligment of UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages Note: Deduced amino acid sequence of HPVR of VP2 from residue 173-386 (numbering of the segment A polyprotein of Bayliss et al., 1990) of the UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages and other published IBDV strains. Major hydrophilic domain (Peak A and B) and minor hydrophilic domains were boxed by black line. The serine rich heptapeptide was shown by underline.

Page 152: Thesis

122

Figure 4.6: Amino acid sequence aligment of UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages Note: Deduced amino acid sequence of HPVR of VP2 from residue 173-386 (numbering of the segment A polyprotein of Bayliss et al., 1990) of the UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages and other published IBDV strains. Major hydrophilic domain (Peak A and B) and minor hydrophilic domains were boxed by black line. The serine rich heptapeptide was shown by underline.

Page 153: Thesis

123

Figure 4.6: Amino acid sequence aligment of UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages Note: Deduced amino acid sequence of HPVR of VP2 from residue 173-386 (numbering of the segment A polyprotein of Bayliss et al., 1990) of the UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages and other published IBDV strains. Major hydrophilic domain (Peak A and B) and minor hydrophilic domains were boxed by black line. The serine rich heptapeptide was shown by underline.

Page 154: Thesis

124

.

Figure 4.7: Phylogenetic tree based on nucleotide sequence of HPVR of VP2 gene of IBDV isolates, displaying relationship of UPM0081T10, UPM0081T15 and UPM0001T20 passages and other published atIBDV strains.

Page 155: Thesis

125

Figure 4.8: Phylogenetic tree based on amino acids sequence of HPVR of VP2 gene of IBDV isolates, displaying relationship of UPM0081T10, UPM0081T15 and UPM0001T20 passages and other published atIBDV strains.

Page 156: Thesis

126

Seq 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27

1UPM0081 ID 29 29 29 1 1 4 4 7 6 9 6 7 7 11 5 40 39 39 39 39 40 42 42 39 40 168

2UPM0081T10 ...... ID 0 0 30 30 33 31 36 31 35 33 34 34 34 30 13 14 27 32 29 13 17 13 15 15 157

3UPM0081T15 ...... ...... ID 0 30 30 33 31 36 31 35 33 34 34 34 30 13 14 27 32 29 13 17 13 15 15 157

4UPM0081T20 ...... ...... ...... ID 30 30 33 31 36 31 35 33 34 34 34 30 13 14 27 32 29 13 17 13 15 15 157

5UPM94368 ...... ...... ...... ..... ID 0 3 3 6 5 8 5 6 6 10 4 39 38 38 38 38 39 41 41 38 39 167

6UPM92/04 ...... ...... ...... ..... ..... ID 3 3 6 5 8 5 6 6 10 4 39 38 38 38 38 39 41 41 38 39 167

7UPM4/230 ...... ...... ...... ..... ..... ..... ID 4 9 8 11 8 9 9 12 7 42 41 41 41 41 42 44 44 41 42 164

8UPM94/273 ...... ...... ...... ..... ..... ..... ..... ID 9 8 11 8 9 9 11 7 40 39 37 37 37 40 42 42 39 40 170

9B00/73 ...... ...... ...... ..... ..... ..... ..... ..... ID 11 14 11 10 10 16 10 42 41 39 41 41 42 44 44 41 42 167

10UK661 ...... ...... ...... ..... ..... ..... ..... ..... ..... ID 7 6 7 7 11 5 38 37 37 37 37 38 40 40 37 38 171

11Task94 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ID 9 10 10 14 8 41 40 41 41 41 41 43 42 40 41 170

12 OKYM ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ID 7 7 9 5 40 39 39 39 39 40 42 42 39 40 165

13HK-46 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0 12 4 39 38 36 36 38 39 41 41 38 39 165

14Ehime91 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 12 4 39 38 36 36 38 39 41 41 38 39 168

15SH95 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 10 41 40 39 38 38 41 43 43 40 41 167

16SH92 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 37 36 36 36 36 37 39 39 36 37 153

17CT ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 1 24 31 26 0 4 2 4 4 152

18KAL2001 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 23 30 25 1 3 3 3 3 165

19GLS ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 21 17 24 26 26 24 24 163

20Variant A ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 19 41 28 33 31 31 164

21E/DEL ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 26 2 28 26 26 153

22Soroa ...... ….. ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... …. ..... ID 6 2 4 4 154

23OKYMT ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 6 6 6 151

24PBG98 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 6 6 149

25D78 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 4 153

26Amer07 ….. ….. ….. …. …. …. …. ..... …. ..... ..... ..... ..... ..... ..... ..... ..... ..... …. …. …. ..... ..... ..... ..... ID 152

27OH ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID

Figure 4.9: Sequence nucleotide difference of VP2 genes of IBDV isolates Note: Twenty seven IBDV isolates were compared: vvIBDV(UPM94268, UPM92/04, UPM4/230, UPM94/273, B00/73, UK661, Tasek.94. OKYM, HK46, Ehime 91, SH/95 and SH/92), caIBDV (CT and KAL2001), variant (GLS, Variant A and E//DEL), attenuated (UPM0081T10, UPM0081T15, UPM0081T20, Soroa, KTI/99. PBG98, D78 and AmerVH9907), and a virulent serotype 2 (OH). Note that the three atIBDV isolates in this study (UPM0081T10, UPM0081T15 and UPM0081T20) were had 13 to 17 nucleotide difference when compare with publish atIBDV, while 29 to 36 with vvIBDV.

Page 157: Thesis

127

Seq 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27

1UPM0081 ID 0.954 0.954 0.954 0.998 0.998 0.993 0.993 0.989 0.990 0.986 0.990 0.989 0.982 0.982 0.992 0.937 0.939 0.939 0.939 0.939 0.937 0.943 0.934 0.939 0.937 0.739

2UPM0081T10 ..... ID 1.000 1.000 0.953 0.953 0.948 0.951 0.949 0.951 0.945 0.948 0.947 0.947 0.947 0.953 0.979 0.978 0.958 0.950 0.954 0.979 0.973 0.979 0.976 0.976 0.756

3UPM0081T15 ..... ..... ID 1.000 0.953 0.953 0.948 0.951 0.949 0.951 0.945 0.948 0.947 0.947 0.947 0.953 0.979 0.978 0.958 0.950 0.954 0.979 0.973 0.979 0.976 0.976 0756

4UPM0081T20 ..... ..... ..... ID 0.953 0.953 0.948 0.951 0.949 0.951 0.945 0.948 0.947 0.947 0.947 0.953 0.979 0.978 0.958 0.950 0.954 0.979 0.973 0.979 0.976 0.976 0.756

5UPM94368 ..... ..... ..... ..... ID 1.000 0.995 0.995 0.990 0.992 0.987 0.992 0.990 0.990 0.984 0.993 0.939 0.940 0.940 0.940 0.940 0.939 0.936 0.936 0.939 0.939 0.741

6UPM92/04 ..... ..... ..... ..... ..... ID 0.995 0.995 0.990 0.992 0.987 0.992 0.990 0.990 0.984 0.993 0.939 0.940 0.940 0.940 0.940 0.939 0.936 0.936 0.939 0.939 0.741

7UPM4/230 ..... ..... ..... ..... ..... ..... ID 0.992 0.986 0.987 0.982 0.987 0.986 0.986 0.979 0.989 0.934 0.936 0.936 0.936 0.936 0.934 0.931 0.931 0.934 0.954 0.741

8UPM94/273 ..... ..... ..... ..... ..... ..... ..... ID 0.986 0.987 0.982 0.987 0.986 0.986 0.982 0.989 0.937 0.939 0.942 0.942 0.942 0.937 0.934 0.934 0.937 0.937 0.741

9B00/73 ..... ..... ..... ..... ..... ..... ..... ..... ID 0.982 0.978 0.982 0.984 0.984 0.975 0.984 0.934 0.936 0.939 0.936 0.936 0.934 0.931 0.931 0.934 0.934 0.736

10UK661 ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.989 0.990 0.986 0.989 0.982 0.992 0.940 0.942 0.942 0.942 0.942 0.940 0.937 0.937 0.940 0.940 0.736

11Task94 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.986 0.984 0.984 0.978 0.987 0.936 0.937 0.936 0.936 0.936 0.936 0.933 0.933 0.936 0.936 0.736

12 OKYM ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.989 0.989 0.986 0.992 0.937 0.939 0.939 0.939 0.939 0.937 0.934 0.934 0.937 0.937 0.736

13HK-46 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 1.000 0.981 0.993 0.939 0.940 0.944 0.944 0.940 0.939 0.936 0.936 0.939 0.939 0.744

14Ehime91 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.981 0.993 0.939 0.940 0.944 0.944 0.940 0.939 0.936 0.936 0.939 0.939 0.744

15SH95 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.984 0.936 0.937 0.939 0.940 0.940 0.936 0.933 0.933 0.942 0.936 0.739

16SH92 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.942 0.944 0.944 0.944 0.944 0.942 0.939 0.939 0.993 0.942 0.741

17CT ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.998 0.962 0.951 0.959 1.000 0.993 0.996 0.995 0.993 0.763

18KAL2001 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.964 0.953 0.961 0.989 0.995 0.995 0.962 0.995 0.764

19GLS ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.967 0.973 0.962 0.959 0.959 0.951 0.962 0.744

20Variant A ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.970 0.951 0.948 0.948 0.959 0.951 0.747

21E/DEL ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.959 0.956 0.956 0.993 0.959 0.746

22Soroa ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.993 0.996 0.990 0.993 0.763

23OKYMT ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.990 0.990 0.990 0.761

24PBG98 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.993 0.990 0.761

25D78 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.993 0.766

26Amer07 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.769

27OH ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID

Figure 4.10 : Sequence nucleotide identity matrix of VP2 genes of IBDV isolates Note: Twenty seven IBDV isolates were compared: vvIBDV(UPM94268, UPM92/04, UPM4/230, UPM94/273, B00/73, UK661, Tasek.94. OKYM, HK46, Ehime 91, SH/95 and SH/92), caIBDV (CT and KAL2001), variant (GLS, Variant A and E//DEL), attenuated (UPM0081T10, UPM0081T15, UPM0081T20, Soroa, KTI/99. PBG98, D78 and AmerVH9907), and a virulent serotype 2 (OH). Note that the three atIBDV isolates in this study (UPM0081T10, UPM0081T15 and UPM0081T20) where identical (97%) to other atIBDV while The degree of identity was down to (95%) when compared with very virulent strains.

Page 158: Thesis

128

Seq 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27

1UPM0081 ID 10 10 10 1 1 3 3 2 1 3 1 1 1 2 2 12 11 13 11 13 12 12 14 11 12 147

2UPM0081T10 ...... ID 0 0 11 11 13 13 12 11 12 11 11 11 12 10 4 5 9 11 13 4 7 4 5 6 145

3UPM0081T15 ...... ...... ID 0 11 11 13 13 12 11 12 11 11 11 12 10 4 5 9 11 13 4 7 4 5 6 145

4UPM0081T20 ...... ...... ...... ID 11 11 13 31 12 11 12 11 11 11 12 10 4 5 9 11 13 4 7 4 5 6 145

5UPM94368 ...... ...... ...... ..... ID 0 2 2 1 0 2 0 0 0 1 1 11 10 12 10 12 11 11 13 10 11 147

6UPM92/04 ...... ...... ...... ..... ..... ID 2 2 1 0 2 0 0 0 1 1 11 10 12 10 12 11 11 13 10 11 147

7UPM4/230 ...... ...... ...... ..... ..... ..... ID 2 3 2 4 2 2 2 3 3 12 11 14 12 14 12 12 14 12 13 147

8UPM94/273 ...... ...... ...... ..... ..... ..... ..... ID 3 2 4 2 2 2 3 3 12 11 12 10 12 12 12 14 12 13 147

9B00/73 ...... ...... ...... ..... ..... ..... ..... ..... ID 1 3 1 1 1 2 2 12 11 13 11 13 12 12 14 11 12 147

10UK661 ...... ...... ...... ..... ..... ..... ..... ..... ..... ID 2 0 0 0 1 1 11 10 12 10 12 11 11 13 10 11 147

11Task94 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ID 2 2 2 3 3 12 11 13 11 13 12 12 14 11 12 147

12 OKYM ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ID 0 0 1 1 11 10 12 10 12 11 11 13 10 11 147

13HK-46 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0 1 1 11 10 12 10 12 11 11 13 10 11 147

14Ehime91 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 1 1 11 10 12 10 12 11 11 13 10 11 147

15SH95 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 2 12 11 13 11 13 12 12 14 11 12 147

16SH92 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 10 9 11 9 11 10 10 12 9 10 147

17CT ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 1 7 9 11 0 3 2 3 4 144

18KAL2001 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 7 9 11 1 2 3 2 3 144

19GLS ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 7 8 7 8 9 7 8 146

20Variant A ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 3 9 10 11 9 10 147

21E/DEL ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 11 12 13 11 12 147

22Soroa ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 3 2 3 4 144

23OKYMT ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 5 4 5 144

24PBG98 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 5 6 144

25D78 ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 2 144

26Amer07 …. …. …. …. …. …. …. ..... …. ..... ..... ..... ..... ..... ..... ..... ..... ..... …. …. …. ..... ..... ..... ..... ID 145

27OH ...... ...... ...... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID

Figure 4.11: Sequence nucleotide difference of VP2 genes of IBDV isolates Note: Twenty seven IBDV isolates were compared: vvIBDV(UPM94268, UPM92/04, UPM4/230, UPM94/273, B00/73, UK661, Tasek.94. OKYM, HK46, Ehime 91, SH/95 and SH/92), caIBDV (CT and KAL2001), variant (GLS, Variant A and E//DEL), attenuated (UPM0081T10, UPM0081T15, UPM0081T20, Soroa, KTI/99. PBG98, D78 and AmerVH9907), and a virulent serotype 2 (OH). Note that the three atIBDV isolates in this study (UPM0081T10, UPM0081T15 and UPM0081T20) were had 4 to7 amino acid difference when compare with publish atIBDV, while 10 to 13 with vaIBDV.

Page 159: Thesis

129

Seq 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27

1UPM0081 ID 0.953 0.953 0.953 0.995 0.995 0.985 0.985 0.990 0.995 0.985 0.995 0.995 0.995 0.990 0.990 0.943 0.948 0.939 0.948 0.939 0.943 0.943 0.934 0.948 0.943 0.326

2UPM0081T10 ..... ID 1.000 1.000 0.948 0.948 0.939 0.939 0.943 0.948 0.943 0.948 0.948 0.948 0.943 0.953 0.981 0.976 0.957 0.948 0.939 0.981 0.967 0.981 0.967 0.971 0.325

3UPM0081T15 ..... ..... ID 1.000 0.948 0.948 0.939 0.939 0.943 0.948 0.943 0.948 0.948 0.948 0.943 0.953 0.981 0.976 0.957 0.948 0.939 0.981 0.967 0.981 0.967 0.971 0325

4UPM0081T20 ..... ..... ..... ID 0.948 0.948 0.939 0.939 0.943 0.948 0.943 0.948 0.948 0.948 0.943 0.953 0.981 0.976 0.957 0.948 0.939 0.981 0.967 0.981 0.967 0.971 0.325

5UPM94368 ..... ..... ..... ..... ID 1.000 0.990 0.990 0.995 1.000 0.990 1.000 1.000 1.000 0.995 0.995 0.948 0.953 0.943 0.953 0.943 0.948 0.948 0.939 0.953 0.948 0.316

6UPM92/04 ..... ..... ..... ..... ..... ID 0.990 0.990 0.995 1.000 0.990 1.000 1.000 1.000 0.995 0.995 0.948 0.953 0.943 0.953 0.943 0.948 0.948 0.939 0.953 0.948 0.316

7UPM4/230 ..... ..... ..... ..... ..... ..... ID 0.990 0.985 0.990 0.981 0.990 0.990 0.990 0.985 0.985 0.943 0.948 0.943 0.942 0.934 0.934 0.942 0.934 0.943 0.939 0.316

8UPM94/273 ..... ..... ..... ..... ..... ..... ..... ID 0.985 0.990 0.981 0.990 0.990 0.990 0.985 0.985 0.943 0.948 0.943 0.952 0.943 0.943 0.942 0.934 0.943 0.939 0.316

9B00/73 ..... ..... ..... ..... ..... ..... ..... ..... ID 0.985 0.985 0.995 0.995 0.995 0.990 0.990 0.943 0.948 0.939 0.948 0.939 0.934 0.942 0.934 0.948 0.942 0.316

10UK661 ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.990 1.000 1.000 1.000 0.985 0.995 0.948 0.953 0.943 0.953 0.943 0.948 0.948 0.939 0.953 0.948 0.316

11Task94 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.990 0.990 0.990 0.985 0.985 0.943 0.948 0.939 0.948 0.939 0.943 0.942 0.934 0.948 0.943 0.316

12 OKYM ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 1.000 1.000 0.995 0.995 0.948 0.953 0.943 0.935 0.943 0.948 0.948 0.939 0.953 0.948 0.316

13HK-46 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 1.000 0.995 0.995 0.948 0.953 0.943 0.953 0.943 0.948 0.948 0.939 0.953 0.948 0.316

14Ehime91 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.995 0.995 0.948 0.953 0.943 0.953 0.943 0.948 0.948 0.939 0.953 0.948 0.316

15SH95 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.990 0.943 0.948 0.939 0.948 0.939 0.942 0.943 0.934 0.948 0.943 0.316

16SH92 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.953 0.957 0.948 0.957 0.948 0.953 0.953 0.942 0.957 0.943 0.316

17CT ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.995 0.967 0.957 0.948 1.000 0.985 0.990 0.985 0.981 0.330

18KAL2001 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.967 0.957 0.948 0.995 0.990 0.985 0.990 0.985 0.330

19GLS ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.967 0.962 0.967 0.962 0.957 0.967 0.962 0.320

20Variant A ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.985 0.967 0.953 0.948 0.957 0.953 0.316

21E/DEL ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.948 0.942 0.939 0.948 0.943 0.316

22Soroa ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.985 0.990 0.985 0.981 0.330

23OKYMT ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.967 0.981 0.976 0.330

24PBG98 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.976 0.971 0.330

25D78 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.985 0.330

26Amer07 ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID 0.330

27OH ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ..... ID

Figure 4.12: Sequence amino acid identity matrix of VP2 genes of IBDV isolates Note: Twenty seven IBDV isolates were compared: vvIBDV(UPM94268, UPM92/04, UPM4/230, UPM94/273, B00/73, UK661, Tasek.94. OKYM, HK46, Ehime 91, SH/95 and SH/92), caIBDV (CT and KAL2001), variant (GLS, Variant A and E//DEL), attenuated (UPM0081T10, UPM0081T15, UPM0081T20, Soroa, KTI/99. PBG98, D78 and AmerVH9907), and a virulent serotype 2 (OH). Note that the three atIBDV isolates in this study (UPM0081T10, UPM0081T15 and UPM0081T20) where identical (98%) to other atIBDV while the degree of identity was down to (95%) when compared with very virulent strains.

Page 160: Thesis

130

4.4 Discussion

The hypervariable region (HPVR) of VP2 is among the most studied parts of the

IBDV genome. The nucleotide and amino acid sequence data obtained for

different IBDV isolates showed this region to be the most diverse region of the

viral genome (Bayliss et al., 1990). The HPVR has also been shown to play a

key role in host immunity by forming a conformational epitopes that is the major

target to which host virus neutralizing antibodies are directed (Fahey et al.,

1989; Eterradossi et al., 1998). The domain composing this epitope is composed

of a collation of hydrophobic amino acids which are flanked on either side by two

hydrophilic peaks (aa 212-224 and aa314-324) (Lana et al., 1992). Substitution

of amino acid within the HPVR, particularly within the hydrophilic peaks, has

previously been associated with changes in viral antigenicity (Jackwood and

Saif, 1987; Ismail et al., 1990; Snyder 1990).

Various studies have demonstrated that passages of virulent viruses in

embryonated egg or tissue culture have decreased virulence and effective

immunogenicity (Yamaguchi et al., 1996b). In this study, the differences in

genetic composition between parental IBDV strains, UPM0081 and two

mammalian cell line (Vero cells and DF-1) passages culture derivatives as it

relates to the nucleotide and amino acid sequences of the VP2 variable region

were determined.

Page 161: Thesis

131

Sequence analyses of selected passages showed that there were no apparent

changes in the VP2 in two passages, Vero and DF-1 cells (UPM0081T5,

UPM008T7, UPM0081D5 and UPM0081D7). A few nucleotide changes in the

VP2 gene with no resultant amino acid substitution by the (UPM0081T8)

passage level, did not significantly affect the homology of amino acid sequence

with parental vvIBDV UPM0081 strains as the homolgy remained at 99%. The

result of the sequencing in (UPM0081T9) passage showed the first mutation in

amino acid sequence at position 222 (A to P) in the major hydrophilic peak A

VP2 gene. The change at this position has been reported to be useful in

predicting the antigenic or pathotypic characteristics of IBDV as A at position

222 has been associated with very virulent viruses, while P has been associated

with attenuated viruses which was the case in this study (Cao et al., 1998).

Further changes in the VP2 gene also occurred in UPM0081T10, UPM0081T15,

and UPM0081T20 passages as detected in the minor hydrophilic peak 1 and

minor hydrophilic peak 2 at position 242 (I to V), 249 (Q to R), 253 (Q to H), 256

(I to V), 279 (D to N) , 284 (A to T) and 294 (I to L). The amino acid substitutions

at positions 279 (D to N) and 284 (A to T) are commonly found in the attenuation

strain as observed in this study (Yamaguchi et al., 1996b). This substitution has

been reported when some highly virulent strains were attenuated and adapted to

CEF (Yamaguchi et al., 1996b; Lim et al., 1999).

Page 162: Thesis

132

Some researchers also demonstrated the importance of VP2 in both the cellular

tropism and pathogenicity of IBDV but the alteration was that of two specific

amino acid 253 (Q to H) and 284 (A to T) within the VP2 protein which resulted

in both adaptations to tissue culture and attenuation in SPF chickens (Mundt

1999; Brandt et al., 2001; van Loon et al., 2002).

From the various reports, it is likely that amino acid 284 plays a central role in

infection of tissue culture (Yamaguchi et al., 1996b; Cao et al., 1998). However,

other amino acids are also important for adaptation. The probable mechanism

that resulted in the additional exchange of amino acid 253 (Q to H) which in turn

enhanced the amount of obtained virus after passaging needed to be

understood in order to explain IBDV adaptation, virus attachment and cell

tropism. It has been suggested that the three determinants (253, 279 and 284)

are located in the most exposed loops of projection domain of VP2, therefore

alternations of three residues of VP2 provides the virus an ability to penetrate

cell culture and grow to give high titer (Boot et al., 2000; Liu and Vakharia, 2004;

Coulibaly et al., 2005).

The importance of the change in the heptapeptide region in UPM0081T10,

UPM0081T15 and UPM0081T20, which changed from SWSASGS (residue 326-

332) in parental IBDV strain (UPM0081) to LWSARGS in cell culture adapted

IBDV strain is not certain. However the amino acid at position 330 (S) in parental

Page 163: Thesis

133

IBDV has been reported to be associated with virulence (Mundit, 1999), hence

the sequences of amino acids in the serine-rich heptapeptide may be useful in

the determination of the virulence of IBDV. It could also be useful to study the

effect of the type of tissue culture, and the passage level on the sequences of

amino acids in the serine-rich heptapeptide of IBDV.

The phylogenetic tree analysis was constructed based on nucleotide (residue

516-1158) and amino acid (173-386) by Clustal X method. The analysis showed

that (UPM0081T10, UPM0081T15 and UPM0081T20) and atIBDV strains (D78

and AmerVH9907) are in a branch in one group and they are similar to other

atIBDV.

It was concluded that the UPM0081T10, UPM0081T15 and UPM0081T20 were

successfully adapted and attenuated in Vero cells as atIBDV strain. The

phylogenetic analysis showed that Malaysian atIBDV isolates shared a common

origin with other attenuated isolates. The most favorable mutation among IBDV

passages used in this study was found in the HPVR of VP2 gene with amino

acid substitutions occurring between 253 (Q to H) and 256 (I to V) and also 279

(D to N) and 284 (A to T). This mutations observed could play a key role in the

adaptation and attenuation of Malaysian field vvIBDV strain (UPM0081) at

different passage levels to Vero cells. The attenuated isolates could in turn be

possible candidates for development live and inactivated IBD vaccines.

Page 164: Thesis

CHAPTER 5

PATHOGENICITY AND IMMUNOGENCITY OF THE ATTENUATED vvIBDV IN SPF CHICKENS

5.1 Introduction

Infectious bursal disease virus (IBDV) causes a highly contagious

immunosuppressive disease in young chickens, which is characterized by the

depletion of the lymphoid follicles of the bursa of Fabricius (Cosgrove, 1962;

Hirai et al., 1981; Okoye et al., 1984). Infection caused by other etiologic agents

such as IBDV infection has been reported to diminish the chicken‟s ability to

respond to vaccination (Cho et al., 1970; Allan et al., 1972; Faragher et al.,

1974; Giambrone et al., 1977).

Infectious bursal disease (IBD) manifests in two forms clinical and subclinical

which is age related. The clinical form of the disease occurred in chickens

between 3 to 6 weeks of age and is responsible for losses due to impaired

growth and death and carcass condemnation from skeletal muscle hemorrhages

(Fadley, 1983). Chicken infected with the classical IBDV (caIBDV) when less

than 3 weeks of age do not exhibit clinical infection, but develop subclinical

infection characterized by microscopic lesions in the bursa of Fabricius and

immunosuppression (Edwards, 1981). The bulk of the economic losses are

associated with subclinical IBDV infection due to the consequent severe and

prolonged immunosuppression (Ivanyi and Morris, 1976).

Page 165: Thesis

135

IBDV is a lymphotropic pathogen with a special predilection for differentiating

cells in the bursa of Fabricius. IBDV infection induces B-cell apoptosis, necrosis,

and bursal atrophy with a concomitant suppression of the humoral response

(Sivanandan and Maheswaran, 1980). The damage to the bursa varies with

some caIBDV strains which induced severe inflammatory response (Benton et

al., 1967; Tanimura et al., 1995) and other strains do result in bursal atrophy

with little or no inflammation (Allan, et al., 1972; Faragher et al., 1974; Tanimura

et al., 1995).

No treatment is available against IBDV infection as most poultry industry uses

different preventive programmes in order to minimize the effects of virus

exposure. These programmes include biosecurity, breeder flock vaccination

and/or progeny vaccination (Lucio and Hitchner, 1980). Although biosecurity is

an important step in the prevention of early exposure to infectious agent, IBDV is

very resistant to inactivation which results in its persistence in poultry houses for

long periods. Therefore, the poultry industry rely more on passive or active

immunity for the control and prevention of IBDV infection. The time of

vaccination in breeders and/or progeny is critical for the success since maternal

antibody can interfere with progeny vaccination (Wood et al., 1981).

The vaccination of chicken with IBD vaccine derived from the classical strains of

the virus had been successful until 1988 with the emergence of very virulent

form of IBDV (vvIBDV) in Europe and variant strains (vaIBDV) in the United

Page 166: Thesis

136

States which had been reported to induced massive mortalities in vaccinated

poultry flocks (Chettle el al., 1989; Rautenshlein et al., 2005). Numerous IBD

commercial vaccines are available and these vaccines have been subjectively

classified as mild, intermediate and intermediate plus (hot) IBD vaccines (Lukert

and Saif, 2003) based on the level of the attenuation of the vaccine virus

employed. The vaIBDV isolates had been reported to vary pathologically and

serologically from the caIBDV strains and they have been reported to have

different neutralizing epitopes which may account for the vaccine failure

observed in the field. It is therefore imperative that vaccines made from local

strains which is antigenically close to that of the wild-type viruses that the

present in the bird‟s environment will provide better protection (Jackwood,

2005). Apart from the antigenic closeness, literature abounds on the effect of

cell culture adaptation and passage level on pathogenicity and immunogenicity

of IBD vaccine. The adaptation and attenuation of IBDV in BGM (grivet monkey

kidney cell line) has been well described and the virus loss its pathogenicity at

more than 30 passages in the cell line (Tsai and Saif, 1992; Hassan and Saif,

1996). The replication of the attenuated virus in host had also been a subject of

debate as the titre induced by these attenuated IBD vaccines in some reports

had been low and some were not protective (Tsai and Saif, 1992). This study

therefore seek to evaluate the pathogenicity and immunogenicity of a Malaysian

isolate of vvIBDV (UPM0081) which was attenuated, propagated and adapted

in Vero cell line with the view of understanading the probable effect of the host

Page 167: Thesis

137

system on the immunogenicity, antigenicity and pathogenicity of the virus at

different passages.

The objectives of this study therefore were:

1. to determine the pathogenicity and immunogenicity of the attenuated

Malaysian isolate of vvIBDV (UPM0081) in Vero cells and in specific-

pathogen- free (SPF) chickens.

2. to determine the efficacy of the attenuated vvIBDV in SPF chickens

5.2 Materials and Methods

5.2.1 Chickens

One-day-old SPF chicks were housed in isolation units at the Experimental

House, Faculty of Veterinary Medicine, Universiti Putra Malaysia and reared

under standard conditions. Clean drinking water and commercial feed were

provided to the chicks.

Page 168: Thesis

138

5.2.2 Selection of IBDV Isolates

A local isolate of vvIBDV serotype 1 identified as UPM0081 strain with an

accession number of AY520910 was first passaged 3 times in 9 to 11-day-old

SPF embryonated chicks eggs prior to adaptation and attenuation in Vero cells

on passages 10, 15 and 20 as previously described (Section 3.2.9). The virus

titer on passages 10, 15 and 20 were 104.7, 106.7 and 107.4 TCID50/ 0.1 mL,

respectively (Section 3.2.10).

5.2.3 Adaptation of IBDV to Embryonated SPF Eggs

As previously described in (Section 3.2.3)

5.2.4 Tissue Culture Infective Dose 50 (TCID50

)

As previously described in (Section 3.2.10)

5.2.5 Experimental Design

Two separate experiments were conducted. The first was the preliminary study

to determine the pathogenicity and immunogenicity of the two vvIBDV

attenuated passages 10 and 15 in SPF chicks and the second to determine the

Page 169: Thesis

139

pathogenicity and immunogenicity of the two vvIBDV attenuated passaged 15

and 20 in SPF chicks.

5.2.6 Experiment 1

Eighteen, 42-day-old SPF chickens were allocated randomly into 3 groups

namely groups A, B and C with 8, 8 and 2 chickens in each group, respectively.

The chickens in group A were inoculated orally with the attenuated P10 IBDV

isolate (UPM0081T10) at a dose of 104.7 TCID50/ 0.1 mL. The chickens in group

B were inoculated with the attenuated P15 isolate (UPM0081T15) at a dose of

106.7 TCID50/ 0.1 mL while, the group C acted as the control group and remained

uninoculated. The chicken in each group were kept in different isolation unit and

were monitored daily of any abnormality and clinical signs. At day 14 post

inoculation (pi), the survived chickens were sacrificed. Blood samples and body

weight were collected. The gross lesions were recorded and bursa of Fabricius

was weight for evaluation of bursa to body weight ratio. Samples of bursa of

Fabricius were collected and fixed in 10% buffered formalin for histopathology

examination.

Page 170: Thesis

140

5.2.7 Experiment 2

One hundred and twenty five SPF chickens of 21-day-old were allocated

randomly into three main groups namely as group 1 (passage 15), group 2

(passage 20) and group 3 (control) with 40 chickens per group except for group

3 (45 chickens). The group 1 was further divided into three sub groups namely

subgroup 1 (a): 30 chickens which served as the sacrificed group. They were

inoculated orally (0.1 mL) with IBDV isolate (UPM0081T15) at a dose of 106.7

TCID50/ 0.1 mL. Subgroup 1 (b): five chickens were inoculated with same dose

orally and served as the positive challenge group and subgroup 1 (c): five

chickens were inoculated with the same dose and served as the mortality group.

The group 2 chickens were also divided into three subgroups. Subgroup 2 (a):

30 chickens which served as sacrificed group were inoculated orally (0.1mL)

with IBDV isolate (UPM0081T20) at a dose of 107.4 TCID50/ 0.1 mL. Subgroup 2

(b): five chickens were inoculated with the same dose orally and served as the

positive challenge group and subgroup 2 (c): five chickens were inoculated with

the same dose orally and served as the mortality group. The group 3 was

subdivided into three subgroups. Subgroup 3 (a): 35 chickens served as

sacrificed group. Subgroup 3 (b): five chickens were served as the positive

challenge group (uninoculated challenged) and subgroup 3 (c): five chickens

were left without inoculation and without challenged. The chickens were

inoculated with the IBDV at 21-day-old and challenged with field isolate of

Page 171: Thesis

141

vvIBDV (UPM0081) with a titer of 107.8 EID50/0.1mL at 35-day old. Five chickens

from subgroup 3 (a) were sacrificed at day 0 before IBDV inoculation. The

chickens from all subgroups 1 (a), 2 (a) and 3 (a) were sacrificed at days

1,3,5,7,10 and 14 post inoculation (pi) and 21 days in subgroups 1 (b), 2 (b),3

(b), 1(c), 2(c) and 3(c) or 1 week post vvIBDV challenged (Table 5.1). At

necropsy, blood samples were collected for detection of IBD antibody. The body

weight, bursa weight, bursa to body weight ratio and gross lesions of the

chickens were recorded. The bursa of Fabricius were collected and fixed into

10% buffered formalin for histology and the other part was for detection of IBDV

using (RT–PCR).

Table 5.1: Groups of SPF chickens inoculated with attenuated vvIBDV passage 15 and 20 and challenged with vvIBDV at day 14 post inoculation.

Definition

Time of sampling ( Days pi)

Time of sampling ( Days pc)

No. of chickens

0 1 3 5 7 10 14

7

1 / (P15)

(a) Attenuated vvIBDV inoculated (sacrificed)

– 5 5 5 5 5 5

30

(b) Attenuated vvIBDV inoculated (challenge)

– – – – – – –

5

5

c) Attenuated vvIBDV inoculated (% of mortality)

– – – – – – –

5

5

2 / (P20)

( a) Attenuated vvIBDV inoculated (sacrificed)

– 5 5 5 5 5 5

30

(b) Attenuated vvIBDV inoculated (challenge)

– – – – – – –

5

5

c) Attenuated vvIBDV inoculated (% of mortality)

– – – – – – –

5

5

3 / (control)

(a) Sacrificed without inoculation

5 5 5 5 5 5 5

35

(b) Without inoculation and vvIBDV challenge

– – – – – – –

5

5

(c) Without inoculation and without challenge (% of mortality)

– – – – – – –

5

5

Page 172: Thesis

142

5.2.8 IBD Challenge

The attenuated vvIBDV inoculated and control challenge groups were

challenged at 14 day post inoculation with a titer of 107.8 EID50/0.1mL of vvIBDV

(UPM0081) via the orally route and kept under strict observation for clinical

signs, gross lesions up to 7 days post challenged.

5.2.9 Histopathology

According to the method of Tanimura et al. (1995). Samples collected from the

experimentally infected SPF chickens were fixed in 10% buffered formalin for at

least 24 hours to be further processed for histopathological examination. The

bursa was trimmed to the thickness of 0.5 cm followed by dehydration through

grade series of alcohol and xylene in the automatic tissues processor (Leica

5500, Germany). The tissues were then embedded in paraffin wax, cut into 4

µm sections and mounted on glass slide. They were dewaxed and stained with

haematoxylin and eosin (H&E) (Lillie, 1965) (Appendix C), and examined under

light microscope to observe histological changes.

Page 173: Thesis

143

5.2.10 Histopathological Lesion Scoring

The histological scoring of bursa of Fabricius was performed based on the

previously described methods (Hair-Bejo et al., 2000). Briefly, bursa of Fabricius

lesions were scored in a scale of 0 to 5 based on the presence of lymphoid cell

necrosis, degeneration, oedema, hetrophil infiltration, and follicular cysts

formation. The score of 0 represented no lesion observed, 1 for mild, 2 for mild

to moderate, 3 for moderate, 4 for moderate to severe and 5 for severe lesions

which were divided into chronic and acute forms (Appendix D).

5.2.11 Collection of Samples for Serological Test

The blood samples were collected to assess the immunity prior and after the

attenuated vvIBDV inoculation at days 0, 1, 3, 5, 7, 10, 14 and 21 post

inoculation (pi). The blood was collected from different groups of chickens via

heart or wing vein using 2 and 5 mL sterile syringe and poured into test tubes

kept in slanting position to collect the serum. The samples were stored at -80oC

till used.

Page 174: Thesis

144

5.2.12 Antibody Assay

The technique of the test was followed as described by Howie and Thorsen,

(1981), and was conducted by One Point Health Company using precoated

ELISA kit (BioChek, UK). Briefly, the test samples were diluted to five hundred

fold (1:500) dilution. The diluted samples (100 µl) were dispensed into the

appropriate 96-well plates coated with IBDV viral antigens. The plates were

incubated for 30 min at room temperature (RT) and were washed with 300 µl of

ddH2O 5 times at the end of incubation period followed by addition of 100 µl of

Anti-chicken: alkaline phosphates conjugated into each well. The plate were

allowed to incubate at RT for 30 min and washed 5 times again before adding

100 µl of p-Nitrophenyl phosphate (PNPP) substrate solution into each test well

which was then incubated for 15 min at RT. Finally, 100 µl of stop solution was

added into each well to stop the reaction and the absorbance were read at 405

nm (Dynatech MR7000, USA).

5.2.13 Reverse Transcriptase Polymerase Chain Reaction (RT-PCR)

Bursal tissue were collected from each of the experiemntal groups for DNA

detection using the reverse transcriptase polymerase chain reaction (RT-PCR).

The procedure used to extract the double- stranded RNA genome of IBDV from

bursal tissues was in accordance to that described by Jackwood et al., (1997).

Page 175: Thesis

145

Briefly, a 4.0 µl volume of each RNA sample in dimethyl sulfoxide (DMSO) was

removed and then amplified by RT–PCR as described previously (Jackwood, et

al., 1997).

5.2.14 Statistical Analysis

The average of body weight, bursa weight, bursa to body weight ratio, lesions

score and mean antibody titer of the inoculated challenge were compared with

those of uninoculated control group for statistical analysis of significant using

analysis of variance (two way ANOVA) followed by Duncan‟s multiple range test

was used as the post hoc produced by using SPSS version 15 for windows

(Norusis, 2004).

5.3 Results 5.3.1 Clinical Signs 5.3.1.1 Experiment 1 Group A: Passage 10

All the chickens in this group did not show any clinical signs of IBDV infection in

the first day pi. At day 2 pi, some of the chickens exhibited typical clinical signs

of IBD including anorexia, severe depression and ruffled feathers. Two out of

eight (25%) chickens died at day 2 and 4 pi.

Page 176: Thesis

146

Group B: Passage 15

No clinical signs and mortality were observed in any bird throughout the

experiment.

Group C: Control Group

No clinical signs and mortality were observed in any bird throughout the

experiment.

5.3.1.2 Experiment 2

Passage 15

Group 1(a): Attenuated vvIBDV Inoculation (Sacrificed)

No clinical signs were observed in this group throughout the experiment.

Group 1(b): Attenuated vvIBDV Inoculation (vvIBDV challenge)

All the chickens did not show any abnormal IBD clinical signs and were 100%

protected against vvIBDV challenged (Table 5.2).

Page 177: Thesis

147

Group 1(c): Attenuated vvIBDV Inoculation (% of mortality)

No abnormal clinical signs of IBD were recorded throughout the experiment.

Passage 20

Group 2(a): Attenuated vvIBDV Inoculation (Sacrificed)

No abnormal clinical signs were observed in the chicken throughout the

experiment.

Group 2(b): Attenuated vvIBDV Inoculation (vvIBDV challenged)

No abnormal clinical signs were observed and the chickens were 100%

protected against vvIBDV challenged (Table 5.2).

Group 2(c): Attenuated vvIBDV Inoculation (% of mortality)

No abnormal clinical signs of IBD were recorded throughout the experiment.

Page 178: Thesis

148

Control

Group 3(a): Non Attenuated vvIBDV Inoculated (Sacrificed)

No clinical signs were observed in any of the chicken throughout the experiment.

Group 3(b): Uninoculated and vvIBDV Challenge (vvIBDV challenge)

No abnormal clinical signs were observed during the first day of the experiment.

At day 2 pi almost all of the chickens exhibited typical clinical signs of IBD

including, anorexia, sever depression and ruffled feathers. However, no diarrhea

and urate were observed. Five out of five chickens (100%) died at day 5 post

challenged (Table 5.2).

Group 3(c): Non Attenuated vvIBDV Inoculation (%of mortality)

No abnormal clinical signs were observed in any chickens throughout the

experiment.

Page 179: Thesis

149

Table 5.2: Rate of mortality and the percentage of protection based on the number of chickens that survived at day 7 post challenged

Challenged groups

Total accumulative death

Number of death/total number of chicken

% of % of

Mortality Protection

Days (Post challenged)

1 2 3 4 5 6 7

Group 1(b): Attenuated vvIBDV inoculation and vvIBDV challenge

0/5 0/5 0/5 0/5 0/5 0/5 0/5

0% 100%

Group 2(b): Attenuated vvIBDV inoculation and vvIBDV challenge

0/5 0/5 0/5 0/5 0/5 0/5 0/5

0% 100%

Group 3(b): Without inoculation and vvIBDV challenge

0/5 2/5 2/5 3/5 5/5 5/5 5/5

100% 0%

All chickens that survived from vvIBDV challenged showed no significant clinical abnormality observed at day 7 post-challenged. 5.3.2 Body Weight 5.3.2.1 Experiment 1 Groups A, B and C

There were no difference in the body weight of chickens between groups B

(867.5 ± 46.5g) and C (875.0 ± 35.3g), while mean body weight of chickens

group A (695.0 ± 102.5g) were significant lower (p<0.05) than the group B

(Table 5.3).

Page 180: Thesis

150

5.3.2.2 Experiment 2

Groups 1(a), 2(a) and 3(a)

Body weight of chickens from the group 1(a) increased significantly (p<0.05)

from day old (82.6 ± 3.7g) to day 14 (351.0 ± 6.5g). The body weight of the

group 2(a) also had significant increased (p<0.05) from day 5 (137.0 ± 8.3g) to

day 14 (355.4 ± 7.7g) (Table 5.4). The body weight of the chickens from the

group 3(a) showed significant increased (p<0.05) from day 1 (81.6 ± 2.1g) to

day 14 (357.0 ± 5.7g). There was no significant difference (p>0.05) in the body

weights of the chickens among groups 1(a), 2(a) and 3(a), respectively

throughout the experiment (Table 5.4).

Groups 1(b), 2(b) and 3(b)

No significant differences (p>0.05) in body weight were observed between

groups 1(b) (478.0 ± 5.7g) and 2(b) (480.0 ± 6.1g) at day 7 post challenged. The

mean body weights of chickens group 3(b) (441.0 ± 13.8g) was significantly

(p<0.05) lower than those of chickens attenuated vvIBDV inoculated with

passage 15 and 20 and challenged with vvIBDV in groups 1(b) and 2(b)

respectively (Table 5.5).

Page 181: Thesis

151

Groups 1(c), 2(c) and 3(c)

There were no significant difference (p>0.05) in the body weight of chicken in all

groups 1(c) 468.0 ± 29.2g, 2(c) 478.2 ± 29.4g and 3(c) 478.8 ± 29.0g at day 7

post challenged (Table 5.6).

5.3.3 Bursa Weight 5.3.3.1 Experiment 1

Groups A, B and C

No difference was observed in the bursa weight of chickens in groups B (2.8 ±

0.2g) and C (2.9 ± 0.1g) at day 14 post inoculation. The chickens in group A (1.5

± 0.2g) which were significantly lower (p<0.05) than the group B (Table 5.3).

Page 182: Thesis

152

5.3.3.2 Experiment 2

Groups1 (a), 2(a) and 3(a)

The chickens in group 1(a) had a significant increased (p<0.05) bursal weight

from day 1 (0.14 ± 0.04g) to day 14 (1.87 ± 0.06g). The bursa weight in the

groups 2 (a) and 3 (a) were also increased from day 1 (0.13 ± 0.03g) and (0.14

± 0.02g) to day 14 (1.86 ± 0.06g) and (1.88 ± 0.05g), respectively. There was no

significant difference (p>0.05) in bursal weight among these groups throughout

the experiment (Table 5.7).

Groups 1(b), 2(b) and 3(b)

The mean bursal weight (0.4 ± 0.1g) of the group 3(b), birds was significantly

lower (p<0.05) than those of the attenuated vvIBDV inoculated challenged birds

in both groups 1(b) (2.5 ± 0.1) and 2(b) (2.5 ± 0.1), while there was no significant

difference (p>0.05) between the groups 1(b) and 2(b) (Table 5.5).

Page 183: Thesis

153

Groups 1(c), 2(c) and 3(c)

There were no significant difference (p>0.05) in the bursa weight of chicken in

all groups 1(c) 2.34 ± 0.1g, 2(c) 2.42 ± 0.1g and 3(c) 2.48 ± 0.1g at day 7 post

challenged (Table 5.6).

5.3.4 Bursa to Body Weight Ratio 5.3.4.1 Experiment 1

Groups A, B and C (1x103)

No difference was observed in the bursa to body weight ratio of chickens in

groups B (3.2 ± 0.4) and C (3.2 ± 0.1) at day 14 post inoculation. The chickens

in group A (2.1 ± 0.4) were significantly lower (p<0.05) than the group B (Table

5.3).

Page 184: Thesis

154

5.3.4.2 Experiment 2

Groups1 (a), 2(a) and 3(a) (1x103)

The bursa to body weight ratio gradually increased (p<0.05) from day 1 (1.7 ±

0.4) to day 10 (5.0 ± 0.2) in group 1(a) and remained stable (p>0.05) from day

10 (5.0 ± 0.2) to day 14 (5.1 ± 0.1). The ratio also increased from day 1 (1.6 ±

0.4) to day 10 (5.1 ± 0.1) in group 2(a) and also remain stable (p>0.05) from day

10 (5.1 ± 0.1) to day 14 (5.1 ± 0.1). The ratio in group 3(a) also followed similar

pattern with increased from day 1 (1.6 ± 0.2) to day 10 (5.0 ± 0.1) and similar

stability in values (p>0.05) from day 10 (5.0 ± 0.1) to day 14 (5.2 ± 0.1) (Table

5.8). There was no significant (p>0.05) difference among group 1(a), group 2(a)

and group 3(a) throughout the experiment (Table 5.8).

Groups1 (b), 2(b) and 3(b) (1x103)

There was no significant (p>0.05) differences in the bursa body weight ratio

between two groups 1(b) (5.2 ± 0.2) and 2(b) (5.1 ± 0.1) while the ratio of

chickens non inoculated and challenged with vvIBDV group 3(b) (0.9 ± 0.1) was

significantly (p<0.05) lower than those of chickens inoculated with passage 15

and 20 and challenged with vvIBDV in groups 1(b) and 2(b), respectively

(Table 5.5).

Page 185: Thesis

155

Groups 1(c), 2(c) and 3(c) (1x103)

There were no significant difference (p>0.05) in the bursa weight of chicken in

all groups 1(c) (5.0 ± 0.1), 2(c) (5.1 ± 0.1) and 3(c) (5.2 ± 0.1g) at day 7 post

challenged (Table 5.6).

5.3.5 Gross Pathology 5.3.5.1 Experiment 1

Groups A, B and C

The bursa of Fabricius (BF) remained normal in groups B and C throughout the

experiment. However in group A, there was mild petechial haemorrhage in the

breast muscle at day 3 pi. At day 4 pi, the BF showed mild to moderate oedema

and necrosis with yellowish gelatinous material (Figures 5.1a and b).

Page 186: Thesis

156

5.3.5.2 Experiment 2

Groups 1(a), 2(a) and 3(a)

All sacrificed chickens did not show any gross lesions throughout the

experimental period

Groups 1(b), 2(b) and 3(b)

The BF and other organs remained normal in the attenuated vvIBDV inoculated

and challenged with vvIBDV in the groups 1(b) and 2 (b). However in

uninoculated and challenged group 3(b), at 2-4 days post challenged, the BF

was swollen (inflamed) and appeared odematous with severe hemorrhage

(Figures 5.2a and b), Hemorrhages of the proventricular mucosa and at the

junction with the gizzard was recorded (Figures 5.3a and b).

Groups 1(c), 2(c) and 3(c)

All sacrificed chickens did not show any gross lesions at day 7 pc.

Page 187: Thesis

157

Figure 5.1a: Experiment 1 (preliminary study): bursa of Fabricius in SPF chickens at day 14 pi. (A) Group C: Control normal. (B) Group A: Passage 10 bursa of Fabricius with mild to moderate odema with yellowish gelatinous material (arrow)

Page 188: Thesis

158

Figure 5.1b: Experiment 1 (preliminary study): bursa of Fabricius in SPF chickens at day 14 pi. (C) Group B: passage 15 normal

Page 189: Thesis

159

Figure 5.2a: Experimental 2 (challenged groups): bursa of Fabricius in SPF chickens at day 7pc. (A) Group1 (b): passage 15 normal. (B) Group 2(b): passage 20 normal.

Page 190: Thesis

160

Figure 5.2b: Experimental 2 (challenged groups): bursa of Fabricius in SPF chickens. (C) Group 3(b): control positive with severe haemorrhages at day 4 pc

Page 191: Thesis

161

Figure 5.3a: Experiment 2 (challenged groups): proventriculus and gizzard in SPF chickens at day 7pc. (A) Group 1 (b): passage 15 normal (B) Group 2 (b): passage 20 normal

Page 192: Thesis

162

Figure 5.3b: Experiment 2 (challenged groups): proventriculus and gizzard in SPF (C) Group 3 (b): control positive hemorrhage on the mucosa of the proventriculus at the junction with the gizzard (arrows) at day 4 pc

Page 193: Thesis

163

5.3.6 Histopathological Changes and Lesion Scoring

5.3.6.1 Experiment 1

Group A

The histopathological changes were severe in the bursa of Fabricius. The

changes were hyperemia, haemorrhage and oedema, followed by lymphoid

depletion, bursal atrophy and cyst formation. The lesion score was 4.66 ± 0.4

(Figures 5.4b, Table 5.3).

Group B

There was no visible histological lesion in bursa of Fabricius and the lesion

score was (0.4 ± 0.1) ( Figure 5.4a, Table 5.3).

Group C

The bursa follicles were apparently normal with

lesion score of 0.5 ± 0.07 (Figure 5.4a).

Page 194: Thesis

164

5.3.6.2 Experiment 2

Groups 1(a), 2(a) and 3(a)

Groups 1(a)

The bursa of Fabricius was characterized by normal to mild tissue reaction.

There was no significant difference (p>0.05) in the lesion score from day 1 (0.34

± 0.54) to day 14 pi (0.42 ± 0.08) throughout the experiment (Table 5.9, Figure

5.5a).

Group 2(a)

Histopathological lesions in bursa of Fabricius remained unchanged (p>0.05)

from day 1 (0.34 ± 0.05) to day 14 (0.38 ± 0.14) throughout the experiment

(Table 5.9, Figure 5.5a).

Group 3(a)

No observable lesions were present in the bursa of Fabricius (p>0.05) from day

1 (0.32 ± 0.44) to day 14 (0.36 ± 0.08). There was no significant difference

(p>0.05) in the lesion score between groups 1(a), 2 (a) and 3 (a) throughout the

experiment (Figures 5.5b, Table 5.9).

Page 195: Thesis

165

Groups 1(b), 2(b) and 3(b)

Groups 1(b)

The bursa of Fabricius was characterized by normal to mild tissue reaction with

active follicles with lesion scoring of 0.8 ± 0.2 (Figure 5.6a, Table 5.5).

Group 2(b)

The bursa of Fabricius was characterized by normal to mild tissue reaction with

active follicles (Figure 5.6a). There was no significant difference (p>0.05) in the

lesion score between groups 2(b) (0.9 ± 0.1) and 1(b) (0.8 ± 0.2) (Table 5.5).

Group 3(b)

The histopathological feature was remarkable. There was severe tissue reaction

as many lymphoid follicles appeared heavily depleted of lymphoid cells. There

was also lymphoid cells aggregation in the cortex, of some follicles while cellular

vacuolation and cyst still existed in some follicles. The interstitial space was

infiltrated with inflammatory cells. The lesion score (4.5 ± 0.5) was significantly

(p<0.05) higher than those of chickens in groups 1(b) and 2(b) respectively

(Table 5.5, Figures 5.6b).

Page 196: Thesis

166

Groups 1(c), 2(c) and 3(c)

Groups 1(c)

All chickens in this group showed normal to mild bursitis with lesion score of 0.4

± 0.1 (Table 5.6, Figure 5.7a).

Groups 2(c)

The bursa of Fabricius was characterized by normal to mild tissue reaction with

lesion score of 0.4 ± 0.1 (Table 5.6, Figure 5.7a).

Groups 3(c)

All chickens in this group showed normal to mild bursitis with lesion score of 0.3

± 0.1. There was no significant difference (p>0.05) in the lesion score among

groups 1(c ), 2(c) and 3(c) (Table 5.6, Figure 5.7b).

Page 197: Thesis

167

Figure 5.4a: Experiment 1 (preliminary study) at day 14 pi. bursa of Fabricius (A) Control group: No lesions were observed, lesion score of 0. (B) Group B: Normal, large active follicles consist of lymphoid cells ( ), lesion score of 0. HE, 10x. Bar = 200µm

Page 198: Thesis

168

Figure 5.4b: Experiment 1 (preliminary study) bursa of Fabricius (C) Group A: Oedematous bursa with degeneration, necrosis ( ) and infiltration of inflammatory cells ( ), follicular cyst ( ) in the medulla, lesion score of 5 at day 2 pi. (D) Group A: More sever lymphoid necrosis ( ) in the medulla, lesion score of 5 at day 5 pi. HE, 20x. Bar = 100 µm

Page 199: Thesis

169

Figure 5.5a: Experiment 2 (sacrificed groups) day 7 pi. bursa of Fabricius (A) Group 1(a): Mild degeneration and necrosis of the follicles ( ), lesion score of 1. (B) Group 2(a): Mild degeneration and necrosis of the follicles ( ), lesion score of 1. HE, 10x. Bar = 200µm

Page 200: Thesis

170

Figure 5.5b: Experiment 2 (sacrificed groups) day 7 pi. bursa of Fabricius (C) Group 3(a): Very clear cortex and medulla packed with healthy follicles, lesion score of 0. HE, 10x. Bar = 200µm

Page 201: Thesis

171

Figure 5.6a: Experiment 2 (challenged groups) at day 7 pc. bursa of Fabricius (A) Group 1(b): Mild degeneration and necrosis of the follicles ( ), lesion score of 1. (B) Group 2(b): Mild degeneration and necrosis of the follicles ( ), lesion score of 1. HE, 10x. Bar = 200µm

Page 202: Thesis

172

Figure 5.6b: Experiment 2 (challenged groups) at day 7 pc. bursa of Fabricius (C) Group 3(b): Depletion of bursa follicles with cysts contains cell debris and fibrinous exudates at medulla follicle ( ), the interstitial connective tissues were obvious, edematous and infiltrated with inflammatory cells ( ), lesion score of 5. HE, 20x. Bar = 100µm

Page 203: Thesis

173

Figure 5.7a: Experiment 2: bursa of Fabricius (mortality groups) at day 7 pc. (A) Group 1(c): Mild lymphoid depletion ( ), lesion score of 1. (B) Group 2(c): Mild lymphoid depletion ( ), lesion score of 1. HE, 10x. Bar = 200µm

Page 204: Thesis

174

Figure 5.7b: Experiment 2 (mortality groups) at day 7 pc. bursa of Fabricius (C) Group 3(c): Mild lymphoid depletion ( ), lesion score of 1. HE, 10x. Bar = 200µm

Page 205: Thesis

175

Table 5.3: Experiment 1: body, bursa, bursa to body weight ratio (1 x 103), lesion scoring and ELISA titer of SPF chicken inoculated attenuated vvIBDV and control group

Group / passage

Weights, lesion scoring and ELISA titer at day 14 pi (Mean ± SD)

Body weight

Bursa weight

Bursa body weight

ratio (x103)

Lesion scoring

ELISA titer

A / P10

695.0 ± 102.5

a

(n = 6)

1.5 ± 0.2

a

(n = 6)

2.1 ± 0.4

a

(n = 6)

4.66 ± 0.4

a

(n = 6)

8174.2 ± 264.5

a

(n = 6)

B / p15

867.0 ± 46.5b

(n = 8)

2.8 ± 0.2

b

(n = 8)

3.2 ± 0.4

b

(n = 8)

0.47 ± 0.1

b

(n = 8)

2808.0 ± 413.0

b

(n = 8)

ab Values with different subscripts within column differ significantly at p<0.05 due to different passages (P10 and P15) n number of sacrificed chicken

Page 206: Thesis

176

Table 5.4: Experiment 2: body weight (g) of chickens in the inoculated attenuated vvIBDV and control group

Group / passage

Body weight (Mean ± SD g)

Time (Days pi)

1 3 5 7 10 14

1(a) / p15

82.6 ± 3.7

a

P

(n= 5)

104.0 ± 4.5

ab

P

(n= 5)

135.0 ± 7.9

b

P

(n= 5)

199.6 ±30.0

c

P

(n= 5)

300.0 ± 15.8

d

P

(n= 5)

351.0 ± 6.5

e

P

(n= 5)

2(a) / p20

82.8 ± 3.8

a

P

(n= 5)

105.8 ± 9.1

a

P

(n= 5)

137.0 ± 8.3

b

P

(n= 5)

202.2 ± 7.1

c

P

(n= 5)

305.2 ± 10.9

d

P

(n= 5)

355.4 ± 7.7

e

P

(n= 5)

3(a) / control

81.6 ± 2.1

a

P

(n= 5)

106.2 ± 5.0

b

P

(n= 5)

136.6 ±5.9

c

P

(n= 5)

208.0 ± 5.7

d

P

(n= 5)

306.6 ± 7.5

e

P

(n= 5)

357.0 ± 5.7

f

P

(n= 5)

ab Values with different subscripts within rows differ significantly at p<0.05 due to time effects pq Values with different subscripts within column differ significantly at p<0.05 due to different passages (P15 and P20) and control group n number of sacrificed chicken

Page 207: Thesis

177

Table 5.5: Experiment 2: body, bursa, bursa to body weight ratio (1 x 103) and lesion scoring of SPF chicken inoculated attenuated vvIBDV and uninoculated challenge group

Group / passage

Weights and lesion scoring at day 7 pc. (Mean ± SD g)

Body weight

Bursa weight

Bursa body weight ratio

(x103)

Lesion scoring

1(b) / P15

478.0 ± 5.7

a

(n = 5)

2.5 ± 0.1

a

(n = 5)

5.2 ± 0.2

a

(n = 5)

0.8 ± 0.2

a

(n = 5)

2(b) / p20

480.0 ± 6.1

a

(n = 5)

2.5 ± 0.1

a

(n = 5)

5.1 ± 0.1

a

(n = 5)

0.9 ± 0.1

a

(n = 5)

3(b) uninoculated

challenge

441.0 ± 13.8

b

(n = 5)

0.4 ± 0.1

b

(n = 5)

0.9 ± 0.1

b

(n = 5)

4.5 ± 0.5

b

(n = 5)

ab Values with different subscripts within column differ significantly at p<0.05 due to different passages (P15 and P20) and control group n number of sacrificed chicken Table 5.6: Experiment 2: body, bursa, bursa to body weight ratio (1 x 103) and lesion scoring of SPF chicken inoculated attenuated vvIBDV and control group

Group / passage

Weights and lesion scoring at day 7pc. (Mean ± SD )

Body weight

Bursa weight

Bursa body weight ratio

(x103)

Lesion scoring

1 (c) / P15

468.0 ± 29.2a

(n = 5)

2.34 ± 0.1a

(n = 5)

5.0 ± 0.1a

(n = 5)

0.4 ± 0.1a

(n = 5)

2 (c) / P20

478.2± 29.4a

(n = 5)

2.42 ± 0.1a

(n = 5)

5.1 ± 0.1a

(n = 5)

0.4 ± 0.1a

(n = 5)

3 (c) / control

478.8 ± 29.0 a

(n = 5)

2.48 ± 0.1a

(n = 5)

5.2 ± 0.1a

(n = 5)

0.3 ± 0.1a

(n = 5)

ab Values with different subscripts within column differ significantly at p<0.05 due to different passages (P15 and P20) and control group n number of sacrificed chicken

Page 208: Thesis

178

Table 5.7: Experiment 2: bursa weight (g) of chickens in the inoculated attenuated vvIBDV and control group

Group / passage

Bursa weight (Mean ± SD g)

Time (Days pi)

1 3 5 7 10 14

1(a) / p15

0.14 ± 0.04

a

P

(n= 5)

0.29 ± 0.08

ab

P

(n= 5)

0.48 ± 0.04

b

P

(n= 5)

0.73 ± 0.06

c

P

(n= 5)

1.53 ± 0.05

d

P

(n= 5)

1.87 ± 0.06

e

P

(n= 5)

2(a) / p20

0.13 ± 0.03

a

P

(n= 5)

0.2 8 ± 0.03

ab

P

(n= 5)

0.37 ± 0.01

b

P

(n= 5)

0.72 ± 0.05

c

P

(n= 5)

1.56 ± 0.03

d

P

(n= 5)

1.86 ± 0.06

e

P

(n= 5)

3(a) / control

0.14 ± 0.02

a

P

(n= 5)

0.32 ± 0.04

b

P

(n= 5)

0.40 ± 0.04

b

P

(n= 5)

0.72 ± 0.51

c

P

(n= 5)

1.56 ± 0.06

d

P

(n= 5)

1.88 ± 0.05

e

P

(n= 5)

ab Values with different subscripts within rows differ significantly at p<0.05 due to time effects pq Values with different subscripts within column differ significantly at p<0.05 due to different passages (P15 and P20) and control group n number of sacrificed chicken

Page 209: Thesis

179

Table 5.8: Experiment 2: bursa to body weight ratio (1 x 103) of chickens in the inoculated attenuated vvIBDV and control group

Group / passage

Bursa to body weight ratio 1 x 10

3 (Mean ± SD)

Time (Days pi)

1 3 5 7 10 14

1(a) / p15

1.7 ± 0.4a

P

(n= 5)

2.8 ± 0.1b

P

(n= 5)

2.9 ± 0.3 bc

P

(n= 5)

3.4 ± 0.3c

P

(n= 5)

5.0 ± 0.2d

P

(n= 5)

5.1 ±0.1d

P

(n= 5)

2(a) / p20

1.6 ± 0.4a

P

(n= 5)

2.6 ± 0.1b

P

(n= 5)

2.7 ± 0.2b

P

(n= 5)

3.5 ± 0.3c

P

(n= 5)

5.1 ± 0.1d

P

(n= 5)

5.1 ± 0.1d

P

(n= 5)

3(a) / control

1.6 ± 0.2a

P

(n= 5)

2.8 ± 0.2b

P

(n= 5)

2.8 ± 0.3b

P

(n= 5)

3.4 ± 0.2c

P

(n= 5)

5.0 ± 0.1d

P

(n= 5)

5.2 ± 0.1d

P

(n= 5)

ab Values with different subscripts within rows differ significantly at p<0.05 due to time effects pq Values with different subscripts within column differ significantly at p<0.05 due to different passages (P15 and P20) and control group n number of sacrificed chicken

Page 210: Thesis

180

Table 5.9: Experiment 2: lesions scoring of chickens in the inoculated attenuated vvIBDV and control group

Group / passage

Lesion scoring (Mean ± SD g)

Time (Days pi)

1 3 5 7 10 14

1(a) / p15

0.34± 0.54a

P

(n= 5)

0.36 ± 0.08a

P

(n= 5)

0.38 ± 0.04a

P

(n= 5)

0.36 ± 0.05a

P

(n= 5)

0.36 ± 0.13a

P

(n= 5)

0.42 ± 0.08a

P

(n= 5)

2(a) / p20

0.34 ± 0.05a

P

(n= 5)

0.36 ± 0.08a

P

(n= 5)

0.40 ± 0.10a

P

(n= 5)

0.38 ± 0.08a

P

(n= 5)

0.44 ± 0.08a

P

(n= 5)

0.38 ± 0.14a

P

(n= 5)

3(a) / control

0.32 ± 0.44 a

P

(n= 5)

0.36 ± 0.08a

P

(n= 5)

0.36 ± 0.05a

P

(n= 5)

0.32 ± 0.13a

P

(n= 5)

0.34 ± 0.05a

P

(n= 5)

0.36 ± 0.08a

P

(n= 5)

ab Values with different subscripts within rows differ significantly at p<0.05 due to time effects pq Values with different subscripts within column differ significantly at p<0.05 due to different passages (P15 and P20) and control group n number of sacrificed chicken

Page 211: Thesis

181

5.3.7 Enzyme Linked Immunosorbent Assay (ELISA) 5.3.7.1 Experiment 1

At two weeks post inoculation, there was a significant difference in the mean

antibody titer (p<0.05) between group A (8174 ± 264) and B (2808 ± 413), while

no antibody was detected in the control group (Table 5.3).

5.3.7.2 Experiment 2

The IBDV antibody titers were detected in the both groups 1 (a) (2066 ± 2452)

and 2 (a) (4205 ± 4223) at day 5 post inoculation (pi). The antibody titers

increased at day 7 pi (8844 ± 18989) and (7663 ± 4244) in group 1(a) and 2(a),

respectively. The maximum antibody titers of 10849 ± 3448 and 11329 ± 2943

in group1 (a) and group 2(a), respectively were observed at 10 days pi. At day

14 pi the antibody titer decreased in both groups 8067 ± 5517 and 6169.4 ±

4013 in group 1(a) and 2(a), respectively. The lowest antibody titers were found

in the group 1(b) 3245 ± 347 and 2(b) 4735 ± 3244 at day 21 pi. There was no

significant difference (p>0.05) in antibody titers of the two groups 1 (a) and

group 1(b) from day 1 to day 21 pi. The control group had no detectable IBD

antibody titer throughout the entire experiment (Table 5.10).

Page 212: Thesis

182

Table 5.10: Antibody titers (mean titer ± standard deviation) to IBD determined by ELISA in the attenuated vvIBDV inoculated groups

(Days post inoculation)

ELISA titer (Mean ± SD)

Group / passage

1 (a) / p15

2(a) / p20

1

0 ± 0a p

0± 0a p

3

0 ± 0a p

0.± 0a p

5

2066 ± 2452b p

4205± 4223b p

7

8844 ± 18989c p

7663 ± 4244bc p

10

10849 ± 3448c p

11329 ± 2943c p

14

8067 ±5517c p

6169 ± 4013b p

21

3245 ± 347b p

4735 ±3244b p

ab Values with different subscripts within column differ significantly at p<0.05 pq Values with different subscripts within rows differ significantly at p<0.05

5.3.8 Detection of the Virus or Viral RNA using RT-PCR

The viral RNA was detected in the bursa homogenates from chickens inoculated

with attenuated vvIBDV passage 15 and 20 in group1 (a), 2 (a) and 3 (a) at day

1, 3,5,7,10,14 pi and group 1(c), 2(c) and 3(c) at day 21 pi using RT-PCR

(Figure 5.8).

Page 213: Thesis

183

Figure 5.8: Hypervariable region (643pb) amplification of IBDV VP2 genes. Lane 1 Day 1; Lane 2 Day 3; Lane 3 Day 5 ; Lane 4 Day 7; Lane 5 Day 10; Lane 6 Day 14; Lane 7 Day 21; and Lane 8 Negative control; M- 100 bp DNA marker (Promega, USA).

5.4 Discussion

In the present study, the Vero cells adapted vvIBDV strain (UPM0081) after 10,

15 and 20 passages were evaluated in SPF chicks for its pathogenicity and

immunogenicity. In the first experiment, the results of the pathogenicity test

indicated that P10 was still pathogenic as evident by the higher bursa lesion

score of 4 and this was in agreement with various investigators especially on

vaccine evaluation (Cursiefen et al., 1979; Muskett et al., 1979; Edwards et al.,

1982). Such vaccine candidate that induces bursal lesions were graded as

intermediate hot vaccines since they induce bursal damage and severe

immunosuppression that are indistinguishable from the field strains (Mazariegos

et al., 1990). The vaccine of this nature are useful to induce active immunity in

chicks with very high level of maternal immunity and in areas where IBD is a

Page 214: Thesis

184

serious challenge. The fact this attenuated virus induces bursal lesion also

agreed with the findings of Raue et al. (2004) that with lower level of attenuation

of IBDV, there is a considerably increase risk of reversion to virulence. In the

case of P10, it can be concluded that the 10 serial passage in Vero cell line is

not enough for sufficient loss of pathogenicity needed for vaccine production

(Rodriguez-Chavez et al. (2002). This observation further revealed the influence

of host system and passage level on the pathogenicity of this strain (Hassan

and Saif 1996).

In this study, the vvIBDV at P15 lost its pathogenicity in SPF chicken as evident

by lower bursal lesion score. This was also observed when the caIBDV passage

in BGM-70 or CEF cells resulted in loss of pathogenicity (Hassan and Saif,

1996) at 6 passages and loss of replication at 30 and 40 passages in BMG 70

cells (Tsai and Saif 1992). The loss of pathogenicity may be because there was

no optimal microenviroment and host factors that are necessary for efficient

virus replication in cells like Vero cells (Lange et al., 1987).

The examination of bursal histopathology, bursa body weight ratio and

percentage protection are common measures used to evaluate the virulence of

vvIBDV. In this study, these methods revealed that the P15 and P20 UPM0081

were not pathogenic as there was no significant difference in the bursa weight

and bursal body weight ratio of the chickens in inoculated groups and that of the

control group. This was not the case with the inoculated and challenged group

Page 215: Thesis

185

and non inoculated and challenged group as there was a significant difference

(p<0.05) in the bursa weight and bursal body weight ratio (Hassan and Saif

1996; Yamagoshi et al., 1996b; Rasool and Hussain, 2006). The

histopathological features and the bursa lesion score in inoculated groups with

Vero cell adapted, attenuated vvIBDV UPM0081 at P15 and P20 also concured

with the fact that the attenuated strain is non pathogenic and do confer 100%

protection to SPF chicks against vvIBDV field strain (Abdel-Alim and Saif

(2001).

The immunogenicity of P10 and P15 Malaysian vvIBDV isolate revealed that

significant antibody level were detected in serum samples from P10 inoculated

group than P15 inoculated group in experiment 1 after 14 days post inoculation,

and this is may be related to the level of attenuation and tropism of the

attenuated virus from P10 virus as it induced obvious clinical syndrome and

bursal damage which may account for the high level of antibody recorded. In

experiment 2 however, the antibody titers in both inoculated groups were

detected after 5 days post inoculation (Ashraf et al., 2005). The antibody titer

increased rapidly at day 7 post inoculation, and the maximum antibody titers

were found at day 10 post inoculation in both groups, these results suggest that

the induced antibody level in both inoculated groups is sufficient to protect the

chicks and this was substantiated by the 100% protection against vvIBDV

challenge. Although the IBD titres observed in this study was not compared with

that from embryo or bursal derived cell line adapted vvIBDV, the study clearly

Page 216: Thesis

186

showed that Vero cell attenuated vvIBDV in passages 15 and 20 do induce

sufficient antibody titre to efficiently result in 100% protection against field

vvIBDV. It was reported that chickens innoculated with cell culture derived IBDV

strains showed poorer immunity when compared with birds that received bursa

derived or embryo derived strains (Rodriguez-Chavez et al., 2002). In contrast,

the VN titres obtained in propagated vaccines in BGM70 and BF cells did not

differ significantly (Hassan and Saif, 1996).

The virus was detected in bursal tissue collected from chickens inoculated with

Vero cells adapted vvIBDV (P15 and P20) at 1, 3,5,7,10,14 and 21 days pi by

RT-PCR. These showed that the virus at this level of passage is replicating

actively in the bursal of Fabricius of inoculated chickens. This was also

observed by Abdel-Alim, (2000) where IBDV was detected in the bursal

homogenates of chicks inoculated with the cell culture adapted virus up to 21

days pi. This ability to replicate was reported to be lost in IBDV passaged in

BGM-70 30 to 40 times (Tsai and Saif 1992). This feature has not been

previously described in Vero attenuated vvIBDV.

It was concluded that the Malaysian vvIBDV (UPM0081) adapted and

attenuated in Vero cell line passage 15 and 20 could provide 100% protection to

SPF chickens from vvIBDV challenged. Hence, the attenuated strains

established in this study may provide a useful virus seed for vaccine

development in the future against vvIBDV field challenge.

Page 217: Thesis

CHAPTER 6

SAFETY AND IMMUNOGENICITY OF THE INACTIVATED ATTENUATED vvIBDV IN SPF CHICKENS

6.1 Introduction

Infectious bursal disease virus (IBDV) is the causative agent of a highly

contagious disease of young chickens. The virus infects and multiplies in

immature B lymphocyte found in the bursa of Fabricius, which can result in

immunosuppression (Faragher et al., 1974; Nick et al., 1976). Recent research

had indicated the emergence of vvIBDV with difference in virulence and the

antigenic variation associated with the strain has been the greatest obstacle for

successful control of this disease (van den Berg, 2000).

Presently, there is no treatment against IBDV infection and the effect of virus

exposure. The induction of active immunity by vaccine is still the effective

method of control, but this should include biosecurity measures (Lucio and

Hitchner, 1979; Baxendale and Luttiken, 1981). The preparation of an effective

vaccine is precluded by the isolation and characterisation of an appropriate

IBDV strains with subsequent attenuation before the preparation into live or

killed vaccines. The inactivation of antigen can be carried out by physical or

chemical agents. For physical methods, heat and radiation are involved while for

chemical reagents, beta propiolactone, formaldehyde, aziridines and other

Page 218: Thesis

188

derivatives had used (Bahnemann, 1990). Of these, formalin had been the

most popular before the advent of Binary ethylenimine (BEI), a type of alkylating

group compounds (aziridines) which reacts poorly with proteins and for this

reason, it does not alter the antigenic components of the virus. BEI has an

inactivation reaction that is more specific for the nucleic acid and has been

known to produce antigenically superior vaccine (Bahnemann, 1990). BEI has

been used to inactivate different viruses such as rabies (Larghi and Nebel,

1980), foot-and-mouth disease virus (Dilovski and Tekerlekov, 1983),

bluetongue virus (Stott et al., 1979), porcine parvovirus (Buonavoglia et al.,

1988), African horse sickness virus (Soliman et al., 1996) and Newcastle

disease virus (King, 1991). The use of this agent in preparation of IBD vaccine

are few in literature (Habib et al., 2006).

On the other hand, Electrolysed water-Catholyte-Anolyte (ECA) is an activated

solution with a highly oxidized anolyte solution which functions as a very fast

acting antimicrobial agent that destroys viruses and other microorganisms

(Anonymous, 1997). Studies have suggested that the hypochlorous acid in the

course of its production can penetrate microbial cell membranes and in turn

exert antimicrobial action through the oxidation of key metabolic systems

(Albrich et al., 1986; Barrette et al., 1989; Hurst et al., 1991). The use of this

agent in the inactivation of IBD viruses has not received adequate attention.

Since the purpose of vaccination is the production of a strong immune response

to the administered antigen, this feat often requires the addition of an adjuvant

Page 219: Thesis

189

(Bomford, 1998). Commonly, water in oil (W/O) emulsions is recommended for

small ruminants, bovine, poultry and fishes when long term immunity is required.

In the case of IBD, mineral oil based emulsions had been used as adjuvant in

order to protect birds against IBDV infection (Benjamin and Hitchner, 1978). The

W/O emulsions usually allow for the reduction of the vaccine dose or the antigen

concentration, which is important to make such vaccines to be cost effective

(Aucouturier et al., 2001).

Inactivated vaccines are most commonly used to vaccinate layer and breeder

birds prior to the laying period. Most inactivated vaccines are used to boost

immunity that has been provided by priming with live vaccines. Alternatively, the

inactivated vaccines that are expected to provide life-long immunity without prior

priming are used in breeders to confer immunity to progeny especially in

endemic IBD areas (van den Berg, 2000).

Freund‟s Complete Adjuvant (FCA), a mixture of a non-metabolizable oil, such

as a mineral oil, a surfactant (Arlacel A) and mycobacteria (M. tuberculosis ) has

been used for many years to enhance immunologic response to antigens, and

even today is considered to be one of the most effective adjuvants (Jackson and

Fox, 1995). However, despite FCA being an effective adjuvant, it has attending

problem and hazards associated with its use especially at the injection site

where it may results in a chronic inflammation response that may be severed

and painful, depending on the site as well as the quantity and quality of adjuvant

Page 220: Thesis

190

injected (Broderson, 1989). The inflammatory response may result in the

formation of chronic granulomas, sterile abscesses, and/or ulcerating tissue

necrosis (Stills and Bailey, 1991). FCA is also a potential hazard for laboratory

personnel as accidental self inoculation can result in tuberculin sensitization

followed by chronic local inflammation, which responds poorly to antibiotic

treatment. Accidental splashing of FCA in the eye can result in severe ocular

irritation and even blindness (Kleinman et al., 1993). Freund‟s Incomplete

Adjuvent (FIA) is similar to FCA but does not contain Mycobacterium. FIA is

frequently used to boost immune system after the use of live vaccines.

The objectives of the study were:

1. to determine the safety and immunogenicity of the attenuated and inactivated

Malaysian isolate of vvIBDV (UPM0081) in specific-pathogen-free (SPF)

chickens.

2. to determine the efficacy of the inactivated vvIBDV in SPF chickens.

Page 221: Thesis

191

6.2 Materials and Methods 6.2.1 Virus and Cells

Passage 15 (UPM0081T15) and passage 20 (UPM0081T20) of vvIBDV were

used as previously described (Section 3.2.6). Briefly Vero cell were grow in

polystyrene 150 cm2 (Nunc Easyflasks, Denmark) containing 30 mL RPMI

supplemented with 10% FBS. A confluent cell monolayer was infected with

vvIBDV (UPM0081) and the virus was allowed to adsorb for 60 minutes at 37oC

in 5% CO2 with intermittent rotation to allow the virus to adsorb on the surface of

Vero cells. After viral adsorption, the inoculums was removed and replaced with

30 mL RPMI containing 1% FBS and retained in an incubator at 37oC in 5%

CO2, for 4 days.

6.2.2 Harvesting of Virus

The Vero cells infected with the IBDV were harvested when cytopathic effect

(CPE) reached 90%. The IBDV-infected cells were centrifuged at 3000 rpm for

20 minutes at 4oC the resultant supernatant fluids were harvested, filtered

through a 0.45 µm filter (Sartorius, Germany), aliquot and stored at -80oC.

Page 222: Thesis

192

6.2.3 Tissue Culture Infective Dose 50 (TCID50

)

As previously described (Section 3.2.10)

6.2.4 Inactivation of vv IBDV

The inactivation of the two Vero cell adapted and attenuated vvIBDV at passage

15 (P15) and 20 (P20) were carried out using Binary ethylenimine (BEI) and

Electrolysed water-Catholyte-Anolyte (ECA) treatment.

6.2.4.1 Binary ethylenmine (BEI) Treatment

Bromoethylamine (BEA) (Sigma, USA) was converted to BEI by adding 2.05g

BEA to 100 mL 0.175N NaOH (0.7g/100 mL deionized water) warmed to 37oC

for one hour. The BEI preparation (2%) was added to the virus suspensions at

P15 or P20 with the virus titer of TCID50=106.7 (P15) and TCID50=107.4 (P20). A

control group without addition of BEI was included. The residual BEI was

hydrolysed in samples by the addition of 1 mol/L sterile Na thiosulfate (Merck)

solution at 10% of the volume of the BEI used (Habib et al., 2006).

Page 223: Thesis

193

6.2.4.2 Electrolysed water-Catholyte-Anolyte (ECA) Treatment

ECA solution, an anolyte with pH 2.2 was used to inactivate the virus by adding

0.5 mL of viral suspensions at P15 (TCID50=106.7) or P20 (TCID50=107.4) to 4.5

mL of the anolyte solution to make 1/10 dilution. A control group without addition

of ECA was included.

6.2.5 Determination of Time Required to Inactivate Virus

Samples from ECA (P15 and P20) and BEI (P15 and P20) as the treated

viruses and the control group were incubated for different times namely as 6,

12, 24, 30 and 36 hours at 37.5oC to determine the inactivation time of the virus.

After incubation period, the treated virus was inoculated in to 5, 10-day-old SPF

embryonated chicken eggs through chorioallantoic membrane (CAM) route

(Table 6.1). After inoculation, all eggs were sealed with melted wax and were re-

incubated at 37.5oC. Inoculated eggs were candled daily for lesions associated

with IBDV (Rosenberger and Cloud, 1985).

Table 6.1: Different time interval to inject SPF embryonated eggs by two kinds of killed vvIBDV (BEI and ECA)

Incubation time (hours at 37

oC)

Treatment / passage

Total SPF

eggs

BEI(P15)

BEI(P20)

ECA(P15)

ECA(P20)

Control

6 1 1 1 1 1 5 12 1 1 1 1 1 5 24 1 1 1 1 1 5 30 1 1 1 1 1 5 36 1 1 1 1 1 5

Page 224: Thesis

194

6.2.6 Perparation of Killed- Virus Oil Emulsion

One volum of killed virus suspecsion (P15 and P20 treated with BEI and ECA)

was mixed with an equal volume of Freund’s incomplete adjuvant (Sigma, USA)

by using a Waring blender at the highest speed 20000 rpm for 15 minutes. An

equal volume of 2% Tween-80 was added to the mixture and the emulsion was

mixed again for 15 minutes giving a final 1:3 dilution of virus suspension

(Benjamin and Hitchner 1978).

6.2.7 Experimental Design

A total of 30, 42-day-old SPF chickens, were divided into six groups namely the

B1, B2, E1, E2, C1 and C2 with 5 birds in each group (Table 6.2). B1 group (BEI

P15), B2 group (BEI P20), E1 group (ECA P15), E2 (ECA P20), C1 (control

negative) without inoculation and unchallenged and C2 (control positive) without

inoculation and challenged. The inoculum in the groups B1, B2, E1, and E2

were mixed with Freund‟s incomplete adjuvant, and injected at 42-day-old

chickens subcutaneously (0.1mL/dose) with IBDV isolate P15 and P20. At 56

days post inoculation the chicken challenged with vvIBDV field strain

(UPM0081) with the titer of 107.8 EID50/0.1mL through the oral route. One

chicken from each group was sacrificed at day 0 prior to inoculation and all

chickens from the groups were sacrificed at day 70 or two weeks post

challenged. The clinical signs and mortality were recorded up to 10 days post

Page 225: Thesis

195

challenged. Blood samples were collected for detection of IBD antibody. The

body weight, bursa weight, bursa to body weight ratio were recorded. Samples

from bursa were collected and a part was fixed in 10% formalin for

histopathology, and the other was used for IBDV detection using RT-PCR.

Table. 6.2 Different groups of chickens inoculated with two types of inactivated vvIBDV (BEI and ECA) and the control group

Group / Passage

Definition

Time of sampling

( Day 0 before inoculation

Time of sampling ( Days 14

pc)

No. of chickens

BEI / P15

Inactivated attenuated vvIBDV inoculated & vvIBDV

challenged

1

4

5

BEI / P20

Inactivated attenuated vvIBDV inoculated & vvIBDV

challenged

1

4

5

ECA / P15

Inactivated attenuated vvIBDV inoculated & vvIBDV

challenged

1

4

5

ECA / P20

Inactivated attenuated vvIBDV inoculated & vvIBDV

challenged

1

4

5

C1

Uninoculated & unchallenged

1

4

5

C2

Uninoculated & challenged

1

4

5

6.2.8 Microscopic Examination and Lesion Score

The collected bursa of Fabricius were fixed in 10% buffered formalin, processed

embedded and cut in a thin sections. The slides were stained using a standard

haematoxlin and eosin staining programme. After the staining, each slide was

dipped in xylene, mounting and cover-slipped. Tissue sections were examined

under a light microscope and lesions were recorded (Hair-Bejo et al., 2000),

(Appendex D).

Page 226: Thesis

196

6.2.9 Determination of ELISA Titer Against Inactivated IBDV Vaccine

The technique of the test was followed as described by Howie and Thorsen,

(1981), and was conducted by One Point Health Company using precoated

ELISA kit (BioChek, UK). Briefly diluted test sera (diluted in phosphate buffer at

1:500) were added into the appropriate wells, already coated with IBDV

antigens and the plate was incubated at 37°C for 30 minutes. The contents of

wells were aspirated and plate was washed five times with 300 µl of ddH2O. 100

μl of anti-chicken alkaline phosphates was added to each well and the plate was

incubated at 37°C for 30 minutes. The plate was washed as above. 100 μl of p-

Nitrophenyl phosphate (PNPP) was added to each well and the plate was

blanked in the air and the reading was recorded by reading the optical density

(OD) spectrophotometrically at 450nm. Positive and negative sera were used as

controls as recommended by the manufacturer.

6.2.10 Reverse Transcriptase Polymerase Chain Reaction (RT-PCR)

Total RNA from bursa of Fabricius was extracted by using the Trizol reagent.

Amplification of the hypervariable region of VP2 genes was carried out by using

reverse transcriptase polymerase chain reaction (RT-PCR). Primers used for

amplification were: (P1) TCA CCG TCC TCA GCT TAC and (P2) TCA GGA TTT

GGG ATC AGC (Jackwood, et al., 1997).

Page 227: Thesis

197

6.2.11 Statistical analysis

As previously described (Section 5.2.14)

6.3 Results

6.3.1 Inactivation of the Virus Attenuated vvIBDV

The two Vero cells adapted IBDV P15 and P20 for each of BEI and ECA were completely inactivated at 24 hours treatment (Table 6.3)

Table 6.3: Mortality of SPF embryonated eggs following inoculation (BEI and ECA) into CAM route Incubation time (hours at 37

oC)

Group / passage N0. Of SPF eggs Cumulative mortality

No. of viable SPF eggs

Mortality (%)

6

BEI / P15 1 1a/1

b 0 100

BEI / P20 1 1/1 0 100 ECA / P15 1 1/1 0 100

ECA / P20 1 1/1 0 100 Control 1 0/1 1 0

12

BEI / P15 1 1/1 0 100 BEI / P20 1 1/1 0 100 ECA / P15 1 1/1 0 100

ECA / P20 1 1/1 0 100

Control 1 0/1 1 0

24

BEI / P15 1 0/1 1 0

BEI / P20 1 0/1 1 0

ECA / P15 1 0/1 1 0

ECA / P20 1 0/1 1 0

Control 1 0/1 1 0

30

BEI / P15 1 0/1 1 0 BEI / P20 1 0/1 1 0

ECA / P15 1 0/1 1 0

ECA / P20 1 0/1 1 0

Control 1 0/1 1 0

36

BEI / P15 1 0/1 1 0

BEI / P20 1 0/1 1 0 ECA / P15 1 0/1 1 0

ECA / P20 1 0/1 1 0

Control 1 0/1 1 0

Page 228: Thesis

198

6.3.2 Clinical Signs

Control Groups

C1 (Control Negative)

No clinical signs and mortality were observed in any chickens throughout the

experiment.

C2 (Control Positive)

No abnormal clinical signs occurred at day 1 post challenged (pc). However, at

day 2 pc, all the chicken showed clinical signs of severe depression, ruffled

feather, and anorexia. There was 100% mortality at day 4 pc (Table 6.4).

BEI Groups

Group: BEI P15

All the chickens in this group did show any abnormal clinical signs throughout

the trial. There was 100% protection against the vvIBDV challenged (Table 6.4).

Page 229: Thesis

199

Group BEI p20

No abnormal clinical signs were recorded throughout the trail. There was 100%

protection against vvIBDV challenged (Table 6.4).

ECA Groups

Group: ECA P15

No abnormal clinical signs were observed throughout the trail. The chickens

were 100% protected against vvIBDV challenged (Table 6.4).

Group: E2 (ECA P20)

No clinical signs of IBD were observed in any of the chickens throughout the

trail. The chickens were 100% protected against vvIBDV challenged (Table 6.4).

Page 230: Thesis

200

Table 6.4: Efficacy of the inactivated attenuated vvIBDV (UPM0081) in SPF chickens

Challenged groups

Total accumulative death

Number of death/total number of chicken

% of % of

Mortality Protection

Days (Post challenged)

1 2 3 4 5 6 7 8 9 10

BEI/P15

0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4

0% 100%

BEI/P20

0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4

0% 100%

ECA/P15

0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4

0% 100%

ECA/20

0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4 0/4

0% 100%

C2

0/4 1/4 1/4 4/4 4/4 4/4 4/4 4/4 4/4 4/4

100% 0%

6.3.3 Body Weight The mean body weight for each group were as indicated in Table 6.5. There

was no significant difference (p>0.05) observed in the body weight among the

groups: 980.0 ± 86.4g (BEIP15), 967.5 ± 91.0g (BEIP20) and 995.0 ± 42.0g

(C1), except in groups ECAP15 and ECAP20 (732.5 ± 53.8)g and (737.0 ±

65.0g), which were significantly lower (p<0.05) when compared with the groups

BEIP15, BEIP20 and C1, respectively (Table 6.5).

Page 231: Thesis

201

Table 6.5: Body weight of chickens in the inactivated attenuated vvIBDV inoculated and control group at 2 weeks post challenged

Body weight (Mean ± SDg)

Group / passage

BEI / P15

BEI / P20 ECA / P15 ECA / P20 C1 / CN C2 / CP

980.0 ± 86.4

a

(n=4)

967.5 ± 91.0

a

(n=4)

732.5 ± 53.8

b

(n=4)

737.5 ± 65.0

b

(n=4)

995.0 ± 42.0

a

(n=4)

NA

NA: Not available due to the death of chickens abValues with different superscripts within rows differ significantly at p<0.05 due to treatment effects n-number of chickens sacrificed

6.3.4 Bursa Weight

There was no significant difference (p>0.05) in the bursa weight of chickens

among groups C1 (3.07 ± 0.31g), BEIP15 (3.01 ± 0.22g), BEIP20 (2.90 ± 0.26g)

and ECAP15 (2.40 ± 0.35g), except for chickens in group ECAP20 (2.3 ± 0.42g)

which were significant lower (p<0.05) than the groups C1 and BEIP15 (Table

6.6).

Page 232: Thesis

202

Table 6.6: Bursa weight of chickens in the inactivated attenuated vvIBDV inoculated and control group at 2 weeks post challenge

Bursa weight (Mean ± SDg)

Group / passage

BEI / P15

BEI / P20 ECA / P15 ECA / P20 C1 / CN C2 / CP

3.01 ± 0.22

a

(n=4)

2.90 ± 0.26

ab

(n=4)

2.40 ± 0.35

ab

(n=4)

2.30 ± 0.42

b

(n=4)

3.07 ± 0.31

a

(n=4)

NA

NA: Not available due to the death of chickens abValues with different superscripts within rows differ significantly at p<0.05 due to treatment effects n-number of chickens sacrificed

6.3.5 Bursa to Body Weight Ratio (1x10-3)

There was no significant difference (p>0.05) between the groups BEIP15 (3.06

± 0.04) and BEIP20 (3.08 ± 0.46), when compared to the group C1 (3.09 ±

0.32). However the bursa to body weight ratio in the groups ECAP15 (2.07 ±

0.43) and ECAP20 (2.05 ± 0.17) were significantly (p<0.05) lower than groups

BEIP15, BEIP20 and C1 (Table 6.7).

Page 233: Thesis

203

Table 6.7: Bursa to body weight ratio of chickens in the inactivated attenuated vvIBDV inoculated and control group at 2 weeks post challenged

Bursa to body weight ratio (x 10

3)(Mean ± SDg)

Group / passage

BEI / P15

BEI / P20 ECA / P15 ECA / P20 C1 / CN C2 / CP

3.06 ± 0.04 a

(n=4)

3.08 ± 0.46 a

(n=4)

2.07 ± 0.43 b

(n=4)

2.05 ± 0.17 b

(n=4)

3.09 ± 0.32 a

(n=4)

NA

NA: Not available due to the death of chickens abValues with different superscripts within rows differ significantly at p<0.05 due to treatment effects n-number of chickens sacrificed

6.3.6 Gross Lesions

Group: C1 (Control Negative)

The chickens did not shown any gross lesions of IBD after two weeks post

challenged (Figure 6.1a)

Group: C2 (Control Positive)

Severe haemorrhagic, oedematous and necrosis of bursa of Fabricius were

observed in the dead chickens at days 2 and 4 post challenged (Figure 6.1a).

Page 234: Thesis

204

Group: BEI P15

The chickens did not shown any gross lesions of IBD after two weeks post

challenged (Figure 6.1b).

Group: BEI P20

The chickens did not shown any gross lesions of IBD after two weeks post

challenged (Figure 6.1b)

Group ECA P15

All chickens showed smaller bursal size after two weeks post challenged as

compared to the size of the BF pre vaccination.

Group ECA P20

All chickens showed smaller bursal size after two weeks post challenged as

compared to the size of the BF pre vaccination.

Page 235: Thesis

205

Figure 6.1a: bursa of Fabricius (BF) in SPF chickens. (A) Group C1: normal (B) Group C2: severe haemorrhages and oedema

Page 236: Thesis

206

Figure 6.1b: bursa of Fabricius (BF) in SPF chickens. (C) Group BEIP15: normal (D) Group BEIP20: normal

Page 237: Thesis

207

6.3.7 Histological Lesions Score

Group: C1

All chickens in this group showed normal to mild bursitis with lesion score of

0.45 ± 0.17 (Figure 6.2, Table 6.8).

Group: C2

Histopathological changes at day 2 post-challenged was severe lymphoid

depletion, lymphocytolysis, degeneration and formation of follicular cyst with

fibrin exudates in the medulla and mononuclear inflammatory cells, oedema in

the interstitial connective tissue with lesion score of 4.72 ± 0.32. (Figure 6.2,

Table 6.8).

Group: BEIP15

The bursa of Fabricius was characterized by normal to mild tissue reaction with

lesion score of 0.62 ± 0.09 (Figure 6.3, Table 6.8).

Page 238: Thesis

208

Group: BEIP20

All chickens in this group showed normal to mild bursitis with lesion score of

0.50 ± 0.81 (Figure 6.3, Table 6.8).

Group: ECAP15

All the chickens in this group showed mild to moderate degeneration and

necrosis of lymphoid cells in the some of lymphoid follicles especially in the

medulla with lesion score of 1.45 ± 0.12 (Figure 6.4, Table 6.8).

Group: ECAP20

All chickens in this group showed mild to moderate bursitis with lesion score of

1.35 ± 0.25 (Figures 6.4, Table 6.8)

Page 239: Thesis

209

Figure 6.2: bursa of Fabricius. (A) Group C1 (Control negative): Normal lymphoid follicles, lesion score of 0. (B) Group C2 (Control positive): lesion score of 5, day 2 pi, sever follicular necrosis with cyst formation in the follicles ( ) and infiltration of inflammatory cells and oedema fluid at interstitial space ( ). HE, 20x. Bar = 100µm

Page 240: Thesis

210

Figure 6.3: bursa of Fabricius at day 14 pc. (A) Group BEIP15: Mild degeneration and necrosis of the follicles ( ), lesion score of 1. (B) Group BEIP20: Mild degeneration and necrosis of the follicles ( ), lesion score of 1. HE, 10x. Bar = 200µm

Page 241: Thesis

211

Figure 6.4: bursa of Fabricius at day 14 pc. (A) Group ECAP15: Mild lymphoid cells necrosis ( ), lesion score of 1. (B) Group ECAP20: Mild cells lymphoid necrosis ( ), lesion score of 1. HE, 10x. Bar = 200µm

Page 242: Thesis

212

Table 6.8: Lesion score of chickens in the inactivated attenuated vvIBDV inoculated and control group at 2 weeks post challenge

Bursa lesion scoring (Mean ± SD)

Group / passage

B1 / P15

B2 / P20 E1 / P15 E2 / P20 C1 / CN C2 / CP

0.62 ± 0.09a

(n=4)

0.50 ± 0.81a

(n=4)

1.45 ± 0.12b

(n=4)

1.35 ± 0.25b

(n=4)

0.45 ± 0.17a

(n=4)

4.72 ± 0.32c

(n=4)

abcValues with different superscripts within rows differ significantly at p<0.05due to treatment effects n number of chickens sacrificed

6.3.8 Antibody Titer (ELISA)

There was no significant difference (p>0.05) between the mean antibody titer at

two weeks post inactivated attenuated vvIBDV inoculation in the two types of

inactivate IBDV (BEI and ECA): 12303 ± 4515 (BEIP15), 13557 ± 2479

(BEIP20), 12131 ± 1932 (ECAP15), except for chickens in group ECAP20 (6615

± 3886) which are significant lowered (p<0.05) when compared to BEIP15,

BEIP20 and ECAP15 groups. At 2 weeks post challenged there was no

significant difference (p>0.05) in mean titers in the three groups: 10262 ± 6405

(BEIP15), 8847 ± 4992 (BEIP20) and 6756 ± 1214 (ECAP15), except for

vaccinated group ECAP20 (3397 ± 1698) which had a significantly lowered titers

(p<0.05) when compared to the groups BEIP15 and BEIP20 (Table 6.9).

Page 243: Thesis

213

6.3.9 Detection of the Virus or Viral RNA using RT-PCR

No viral RNA was detected in the bursa homogenates from chickens inoculated

with inactivated vvIBDV in groups BEIP15, BEIP20, ECAP15 and ECAP20 at

day 14 pc (Figure 6.5).

Table 6.9: Antibody titers to IBDV determined by ELISA in the inactivated attenuated vvIBDV inoculated and uninoculated groups after two weeks of post inoculated and two weeks post challenged.

Antibody titer ( Mean ± SD)

Time (Weeks pi & pc)

Group / passage

2 weeks pi

2weeks pc

BEIP15

12303 ± 4515a

p

10262 ± 6405a

p

BEIP20

13557 ± 2479a p

8847 ± 4992a

q

ECAP15

12131 ± 1932a p

6756 ± 1214ab

q

ECAP20

6615 ± 3886b p

3397 ± 1698b

q

CI

ND

ND

C2

ND

NA

ab Values with different subscripts within column differ significantly at p<0.05 pq Values with different supscripts within rows differ significantly at p<0.05

NA: Not available due to the death of chickens ND: Not detected

Page 244: Thesis

214

Figure 6.5: Hypervariable region (643pb) amplification of IBDV VP2 genes at day 14 pc. (1) BEIP15 negative (2) BEIP20 negative (3) ECAP15 negative (4) ECAP20 negative (5) C2 positive. (M) 100 bp DNA marker (Promega, USA).

6.4 Discussion

The study showed that UPM0081T15 and UPM0081T20 attenuated vvIBDV

were successfully inactivated by Binary ethylenimine (BEI) and Electrolysed

water-Catholyte-Anolyte (ECA) 24 hours after the treatment. The inactivated

vvIBDV conjugated with incomplete Freund‟s adjuvant could provide full

protection (100%) against vvIBDV (UPM0081) challenged. Inoculation of

inactivated IBDV could give complete protection with no obvious IBD clinical

signs, as it was reported previously (Maas et al., 2001).

Page 245: Thesis

215

The mean body weight of treatment groups (C1, BEIP15 and BEIP20) was not

significantly (p>0.05) different throughout the experimental study while that of

groups ECAP15 and ECAP20 was significant lower (p<0.05) when compared to

that of groups C1, BEIP15 and BEIP20 at week 2 post vaccination. The bursal

weight of the chicken followed similar pattern like the body weight except that of

group ECAP15 was significantly different from ECAP20 group, the exactly

reason to this is unknown.

As regards the bursa to body weight ratio, there was no significant difference

(p>0.05) in BEI treatment groups (B1, B2) and the control negative group (C1).

These results corroborated those reported by Hassan and Saif (1996) where

they also reported no significant difference in the bursa to body weight ratio of

chicken administered with BGM-70 attenuated and inactivated IN and STC

strains of IBDV and the control group. The chickens in groups (ECAP15 and

ECAP20) on the other hand had a significantly (p<0.05) lower bursa body

weight ratio when compared with (C1). This clearly showed the superiority of

BEI in effectively inactivating the vvIBDV and also in protecting the chicken

against bursal damage.

Histopathological assessment also revealed similar pattern as the bursa lesion

score was normal to mild in treatment groups C1, BEIP15 and BEIP20 groups

while that of ECAP15 and ECAP20 had mild to moderate.

Page 246: Thesis

216

On the basis of humoral immune response, the results showed that the

inactivated passage 15 UPM0081T15 and passage 20 UPM0081T20 of vvIBDV

used in all the treatment groups were immunogenic with increased in antibody

titers in all inoculated groups 2 weeks pi. It is evident that groups BEIP15,

BEIP20 and ECAP15 had no significant titres (p>0.05) while group ECAP20 was

significantly (p<0.05) lower 2 weeks pi (Jackwood, 2005; Habib et al., 2006)

while that with ECAP15 and ECAP20 were observed probably for the first time

and this shows that ECA has a potential as a inactivating and immunogenic

agent especially with IBD viruses. The possible mechanism of inactivation of

ECA as regards IBDV needed to be investigated while that of BEI and its use in

the inactivation of viruses for vaccine development abound in literature

(Buonavoglia et al., 1988; Bahnemann, 1990).

Post challenged titres measured by ELISA followed the same pattern with the

post inoculation titres except that the titres were lowered and this are quite

understandable as the field virus has been reported to mop up antibodies.

Again, it could be deduced from the post challenged titers that the BEI-

inactivated groups gave higher antibody titers than ECA inactivated groups

although ECAP15 group had better response than ECAP20. The probable

reason for the higher titers in ECAP15 group may not be acertain but it could be

related with the level of passage as ECAP15 had 15 passages when compared

to 20 passages before inactivation in ECAP20. This could best be said to be a

function of the available viral antigen in each group.

Page 247: Thesis

217

The result of this study also showed that a single dose of the inactivated

passage 15 UPM0081T15 and passage 20 UPM0081T20 of IBDV in both

groups (BEI and ECA) could give 100% protection against vvIBDV challenged,

which is in contrast with the report of 100 % protection obtained with the use of

two doses of killed IBD vaccines at a week interval in 3 weeks SPF chickens

(Hsieh et al., 2007).

Since the current facts showed that the humoral immune response plays the

principal role in defense against vvIBDV (Lukert and Saif, 1997), there may be a

need to study the cell mediated immune response and the effect of vaccine

inactivated by these substances (BEI and ECA) on the cellular mechanism since

T cells are also important in the protection against virulent IBDV (Rautenschlein

and Sharma, 2002).

In order to have a better understanding of the chemical inactivation of the IBDV,

the efficacy of these chemicals was studied by using two passages (P15 and

P20) of attenuated Vero adapted IBDV, the probable effect of the passages was

evident in ECA groups with P15 giving promising results in terms of immune

response pre- and post challenged. The results obtained with ECA group do not

totally showed that the ECA inactivated passage 15 UPM0081T15 and passage

20 UPM0081T20 of IBDV could not be useful as a vaccine, as it could be given

twice to enhance the immune titers in endemic IBD areas or in chicks with high

maternal antibodies. This sugestion needed to be verified before adoption.

Page 248: Thesis

218

The scope of this study did not cover the possible effect of the adjuvant

employed but it was assumed that the adjuvant also enhanced the humoral

response that was recorded and the use of tween-80 in the combination with

Freund‟s incomplete adjuvant (FIA) made the adjuvant less viscous and this

may have reduced the side effects associated with the adjuvant and the

possible alteration of the antigenicity of IBDV (Stone et al.,1978).

The chemical interactions between the BEI, ECA and incomplete Freund‟s

adjuvant needed to be investigated as this may give a clue to the difference in

the humoral and histopathological scores observed in the two treatment groups.

In this experiment, the possibility of replication of the inactivated virus was also

investigated by using RT-PCR. No viral genetic material was detected in the

bursa of Fabricius of the inoculated birds at two weeks post challenged,

indicating that the IBDV was inactivated and there was no replication of viral

RNA or inactivated virus in bursa of Fabricius. This further showed that the

reversal to virulence as in the case of attenuated live vaccine is not possible in

this case.

It was concluded that chickens inoculated once with inactivated (BEI or ECA)

attenuated vvIBDV UPM0081 in Vero cell at passages 15 and 20 could

adequately protect against vvIBDV challenged. It also revealed that antibody

specific to IBD detected by ELISA in BEI treated virus was higher than ECA

Page 249: Thesis

219

treated virus. Although both inoculated viruses (BEI or ECA) were able to

protect chicken against vvIBDV challanged, ECA treated virus could not provide

protection against bursal damage in challenged chickens.

Page 250: Thesis

CHAPTER 7

GENERAL DISCUSSION, CONCLUSION AND RECOMMENDATIONS FOR FUTURE RESEARCH

7.1 General Discussion

Since the control of IBD is based mainly on vaccination, continuous efforts were

directed towards developing effective and economic vaccines against IBDV. The

work presented in this dissertation was initiated to investigate the adaptation

and attenuation local vvIBDV in tissue culture, and possible vaccine production

with thr improving the efficacy of local attenuated and inactivated vaccines

which are employed in the effective control of IBD in Malaysia, and world wide.

In this experiment, a total of three Malaysia vvIBDV field strains designated as

UPM04190, UPM94273 and UPM0081 with an accession number of AY791998,

AF527039 and EF208038, respectively were used in first set of investigations.

The inoculation of theses isolates into the embryonated chicken eggs was

observed to result in embryonic death and the lesions observed was similar to

those observed in other vvIBDV strains (Hair-Bejo et al.,1995a; Yamaguchi et

al., 1996a).

In this study, the adaptation and attenuation of a local strain, in two continuous

cell lines namely Vero cells and DF-1 cells was demonstrated. UPM0081 strain

was successfully adapted to grow in Vero cells after four passages and DF-1

Page 251: Thesis

221

cells after three passages, while the other isolates failed to adapt on Vero cells

and DF-1 cells after six passages. These findings further reinforced the view

that several wild strains are difficult to replicate and grow in vitro, and some, if

they eventually grow, often require several weeks and blind passages

(McFerran et al., 1980; Hassan et al., 1996). In this investigation, there was no

evidence of growth by CPEs in the first, second and third passages while at the

fourth, clear and constant CPEs of IBDV were found in Vero cells monolayer.

The CPEs involved aggregation of Vero cells, formation of round, refractile cells

and finally granulation and detach from the surface. In DF-1 cells, the first and

second passages of the UPM0081 strain did not produce any CPEs, while the

third gave clear and consistent CPEs of cell rounding, granulation and

detachment from the substrate. The growth of the virus in infected cell cultures

(Vero cells and DF-1 cells) was further confirmed with stained HRP-conjugated

antibody in IIPS. The infected cell cultures appeared as specific intracytoplasmic

brown colour when examine under light microscope.

When the virus titers obtained were compared in Vero and DF-1 cells, the result

showed that Vero cells yielded higher titers than DF-1 cells, and for this reason,

it was concluded that the Vero adapted attenuated virus should be further

studied for possible adoption as a candidate for an attenuated and inactivated

local vaccine development.

Page 252: Thesis

222

During the last few years, many molecular studies have detected genomic

changes that account for attenuation in tissue culture of IBDV. The comparison

of the local attenuated (at) IBDV strains obtained in this study, with other IBDV

strains was done by molecular methods since the molecular characterization of

IBDV isolates from different geographical regions and identification of

differences among them had helped in developing a correct and effective

vaccine (Pitcovski et al., 1998).

In this study, Vero cells and DF-1 cells adapted vvIBDV local isolates were

used. The viral RNA extraction was done using Trizol reagent. RT-PCR was

performed for all the RNA samples using P1 and P2 pair of primers specific to

HPVR of VP2 gene to confirm the presence and genetic mutation of IBDV in cell

line. The expected 643-bp was obtained for each passage and cloned in the

PCR 2.1- TOPO TA Cloning.

In this study, the first changes of amino acid residues, occurred at positions 222

(A to P) in passage 8 (UPM0081T8) and further mutation of amino acid residue

was observed at positions 242 (I to V), 249 (Q to R), 253 (Q to H), 256 (I to V),

279 (D to N) , 284 (A to T) and 294 (I to L), in passages 10, 15 and 20

(UPM0081T10, UPM0081T15, and UPM0081T20). These mutations were

similar to those reported for IBDV attenuated strains (Yamaguchi et al., 1996b).

Page 253: Thesis

223

It is interesting to note that all the three amino acid substitution at position 253

(Q to H), 279 (D to N) and 284 (A to T) are within the areas of the protein

predicted to be associated with antigenic variation, as well as back bone

flexibility. It is possible that these changes especially at residue (284) could

result in significant functional rearrangement of the virus capsid that is

advantageous for virus replication and attenuation in Vero cells.

In phylogenetic tree based on segment A, IBDV strains are usually distinctly

grouped into separate branches according to their serotype and virulence. In

this study, UPM0081T10, UPM0081T15 and UPM0081T20 IBDV passages

were closely related to atIBDV strains and they formed a subbranch with classic

attenuated (D78) and atIBDV AmerVH9907. Generally, many attenuated

vaccines had been successfully used to develop attenuated live vaccine as

evidenced by a reduction in the ability of the virus to induce bursal lesions

(Yamaguchi et al., 1996a).

Based on the pathogenicity and immunogenicity attenuated vvIBDV in SPF

chicken in this study, the first preliminary investigation revealed that P10

(UPM0081T10) was still pathogenic, as it caused mortality (25%) in SPF

chicken and bursa lesion score of 4 (Raue et al., 2004), while the P15

(UPM0081T15) virus was observed to have lost their pathogenicity after

passage in Vero cells as evident by no mortality, bursal lesion score of 0. These

Page 254: Thesis

224

findings are similar to that reported for other attenuated IBDV strains (Guittet et

al., 1992).

It should be noted that passaging in vitro of an IBDV isolate does not only

reduces strain virulence but also changes its antigenicity. Therefore, most of the

live attenuated vaccines available today have a different passage history and

they show a different level of virulence and antigenicity. Thus, it is believed that

an antigenically superior vaccine can best be obtained from vvIBDV strains

provided the attenuation is adequately achieved (Melchior and Melson 1989) as

observed in this study.

It was also noted that the inoculated passage 10 (UPM0081T10) had higher IBD

antibody titer than the passage 15 (UPM0081T15) at 2 weeks post inoculation.

This is may be due to the fact that the virus in passage 10 is still more

pathogenic and subsequently immunogenic than passage 15. The study

showed that passages 15 (UPM0081T15) and 20 (UPM0081T20) were safe and

could confer 100% protection against vvIBDV field challenged with no clinical

signs of IBD.

The two Vero cells adapted attenuated UPM0081T15 and UPM0081T20

vvIBDV also provided full protection from bursal lesion against the vvIBDV

challenged. These findings support the previous report by Ismail and Saif

Page 255: Thesis

225

(1991), in which two virulent strains were adapted and passaged in BGM cell

line (BGM-70).

Both viruses (UPM0081T15 and UPM0081T20) also produced titers detectable

after 5 days post inoculation which increased rapidly at day 7pi, and the

maximum antibody titers were found at day 10 pi in both groups, these results

suggest that the acquired serum antibody in both groups, was sufficient to fullly

protect the chickens (100%) against vvIBDV challenge.

The study went further to showed that viral antigen was detectable in the bursa

homogenates of SPF chickens up to 21 days using RT-PCR after inoculation of

two passages (UPM0081T15 and UPM0081T20) Vero adapted attenuated

isolates. This observation was similar to the findings of Abdel-Alim, (2001) who

also found detectable RNA in bursa haemogenates up to 21 days.

The pathogenecity and immunogenicity of two types of inactivated local vvIBDV

(UPM0081T15 and UPM0081T20) isolates were also evaluated in SPF

chickens. The two vaccine candidates were successfully inactivated by Binary

ethylenimine (BEI) and Electrolysed water-Catholyte-Anolyte (ECA), and they

were found to fully protected (100%) the SPF chickens against challenge

vvIBDV strain (UPM0081) challenged, throughout the period of the experimental

study. The BEI treated inactivated Vero adapted isolates fully protected the

birds against bursal damage when challenged with vvIBDV strain (UPM0081)

Page 256: Thesis

226

while the chicken vaccinated with ECA treated inactivated Vero adapted isolates

had mild to moderate bursal lesions. The findings also indicated that the killed

vaccines used (BEI and ECA) were capable of inducing satisfactory serologic

response as evident by the increased in antibody titers observed in all the

vaccinated birds, but BEI-inactivated vaccines gave higher antibody titers than

ECA inactivated vaccines.

In the present study, the possible influence of Freund‟s incomplete adjuvant

(FIA) water in oil (W/O) emulsions on the inactivated (BEI and ECA) IBDV

antigen was also explored. This study showed that the adjuvant combined with

the inactivating agents do gave satisfactory and protective humoral response in

BEI inactivated IBDV while the possible reason for the low titers in ECA

inactivated IBDV may not be unconnected with the combination.

7.2 Conclusion

This study has shown that the Malaysian vvIBDV strains UPM0081 strain was

successfully propagated and attenuated in Vero cells and DF-1 cell lines with

higher titers recorded in Vero cells, hence Vero cell line could be used as a

model to study the growth kinetic of the IBDV isolate.

Based on molecular techniques, the IBDV passages, UPM0081T10,

UPM0081T15 and UPM0081T20 were characterised as atIBDV. The sequence

Page 257: Thesis

227

analysis showed that most mutation among these isolates focused on the HPVR

of VP2 gene especially between 253 (Q to H), 256 (I to V), 279 (D to N) and 284

(A to T) and these mutations could be said to play an important role in the

adaptation Malaysian vvIBDV isolate (UPM0081) to Vero and DF-1 cell lines.

The phylogenetic analysis also showed that these cell line adapted isolates

were evolutionary closed to atIBDV strains.

It is also clear from the present findings that the inoculation of Malaysian

vvIBDV adapted and attenuated in Vero cell line passages 15 and 20 conferred

full protection to the immunized SPF chickens.

The water in oil emulsion Freund‟s Incomplete Adjuvant inactivated (BEI and

ECA) vaccine candidates also protected the chickens fully from vvIBDV

challenged. However, the antibody response to IBD detected by ELISA in group

with BEI was higher than the ECA group. Although the two groups were able to

protect SPFchickens from mortality and clinical signs of the disease, the group

with BEI fully protected against bursal lesion while the ECA group could not.

From this study, these possible vaccine candidates may be recommended for a

field trial to further evaluate the safety and efficacy of these possible vaccines

on a larger scale.

Page 258: Thesis

228

7.3 Recommendation for Further Research

1- To demonstrate the role of receptors in the adaptation and attenuation of

vvIBDV in Vero cells using monoclonal antibodies.

2- To investigate the use of chicken stem cell line in the adaptation and

attenuation of vvIBDV.

3- To further the investigation into the influence of serum or protein and insulin-

like growth factor I (IGF-I) on the ability of vvIBDV strains to infect and replicate

in cell culture.

4- To improve the quality of attenuated and inactivated vaccine candidates by

using different mechanisms of increasing vaccine virus concentration so as to

provide adequate protection by a single dose.

5- To study the cell mediated immune (CMI) response of chicken using the

attenuated and inactivated vaccines candidates.

6- To adopt the use of real time RT-PCR in the evaluation of vaccination

program in order to differentiate between wild IBDV and the spread of vaccine

Page 259: Thesis

229

virus in poultry flocks. It will also help to obtain the virus titers than the

qualitative result from RT-PCR.

7- To conduct field trail on the safety and efficacy of the live and inactivated

atIBDV seed virus for vaccine development.

Page 260: Thesis

230

BIBLIOGRAPHY

Abbas, A.K., Lichtman, A.H. and Pober, J.S. (2001). Cellular and molecular

immunology. (A.K. Abbas, Lichtman, and J.S. Pober ED.), W. B. Saunders, Philadelphia, PA.

Abdel-Alim, G.A. (2000). Immunogenicity and Pathogenicity of very virulent

strain infectious bursal disease virus in Chickens. Ph.D. thesis, Department of Veterinary Preventive Medicine. University of Ohio state.

Abdel-Alim, G.A. and Saif, Y.M. (2001). Immunogenicity and antigenicity of very

virulent strains of infectious bursal disease viruses. Avian Dis., 45: 92–101.

Ahasan, M.M., Hossain, K.M., and Islam, M.R. (2002). Adaptation of infectious

bursal disease virus on Vero cell line. Online J. Biol. Sci., 2(9): 633-635. Albrich, J., Callahan, J.H.K.B. and Hurst, J.K. (1986). Effects of the putative

neutrophil-generated toxin, hypochlorous Acid, on membrane permeability and transport systems of Escherichia coli. J. Clin. Invest., 78:177- 184.

Allan, W.H., Faragher, J.T. and Cullen, G.A. (1972). Immunosuppression by the

infectious bursal agent in chickens immunised against Newcastle disease. Vet Rec., 90:511-2.

Almassy, K. and Kakuk, T. (1976). Immunosuppressive effect of a naturally

acquired subclinical bursal agent infection on vaccination against Newcastle disease.Vet Rec., 99(22):435-7.

Amakye-Anim, J., Lin, T.L., Hester, P.Y., Thiagarajan, D., Watkins, B.A. and

Wu, C.C. (2000). Ascorbic acid supplementation improved antibody response to infectious bursal disease vaccination in chickens. Poult Sci., 79(5):680-8.

Anderson, W.I., Reid, W.M., Lukert, P.D. and Fletcher, O.J. J. (1977). Influence

of infectious bursal disease on the development of immunity to Eimeria tenella. Avian Dis., 21(4): 637-41.

Anonymous, (1997). Principle of formation of electrolytic water. Hoshizaki

Electric Co. Ltd., Sakae, Toyoake, Aichi, Japan.

Page 261: Thesis

231

Ashraf, S., Abdel-Alim. G., Al-Natour, M.Q. and Saif, Y.M. (2005). Interference between mild and pathogenic strains of infectious bursal disease virus in chickens. Avian Dis., 49: 99-103.

Astile, T.P., Vainio, O. and Lassila, O. (1994). Central role of CD4 T cells in

avian immune response. Poultry Sci., 73: 1019-1026. Aucouturier, J., Dupuis, L. and Ganne, V. (2001). Adjuvants designed for

veterinary and human vaccines. Vaccine, 19: 2666–2672. Azad, A.A., Jagadish, M.N., Brown M.A. and Hudson, P.J. (1987). Deletion

mapping and expression in Escherichia coli of the large genomic segment of a birnavirus. Virol., 161:145-52.

Azad, A.A., McKern, N.M., Macreadie, I.G., Failla, P., Heine, H.G., Chapman,

A., Ward, C.W. and Fahey K.J. (1991). Physicochemical and immunological characterization of recombinant host-protective antigen (VP2) of infectious bursal disease virus. Vaccine, 9(10):715-22.

Bahnemann, H.G. (1990). Inactivation of viral antigens for vaccine preparation

with particular reference to the application of binary ethylenimine. Vaccine, 8(4): 299-303.

Barrette, W.C., Hannum, D. M. and Wheeler, W. D. (1989). General mechanism

for the bacterial toxicity of hypochlorous acid: Abolition of ATP production. J. Am. Chem. Soc., 28: 9172-9178.

Baxendale, W. and Lutticken, D. (1981). The results of field trails with an

inactivated Gumboro vaccine. Dev. Biol. Stand., 51: 211-219. Bayliss, C.D., Peters, R.W., Cook, J.K., Reece, R.L., Howes, K., Binns, M.M.

and Boursnell, M.E. (1991). A recombinant fowlpox virus that expresses the VP2 antigen of infectious bursal disease virus induces protection against mortality caused by the virus. Arch Virol., 120(3-4) :193-205.

Bayliss, C.D., Spies, U., Shaw, K., Peters, R.W., Papageorgiou, A., Muller, H.

and Boursnell, M. E. (1990). A comparison of the sequences of segment A of four infectious bursal disease virus strains and identification of a variable region in VP2. J Gen Virol., 71 (6):1303-12.

Bayyari, G.R., Story, J.D., Beasley, J.N. and Skeeles, J.K. (1996).

Pathogenicity studies of an Arkansas variant infectious bursal disease virus. Avian Dis., 40: 516-336.

Becht, H. (1980). Infectious bursal disease virus. Curr. Topic. Microbiol and

Immunol., 90:107-121.

Page 262: Thesis

232

Becht, H., Muller, H. and Muller, H.K. (1988). Comparative studies on structural and antigenic properties of two serotypes of infectious bursal disease virus. J Gen Virol., 69 (3):631-40.

Benjamin, L. and Hitchner, B. (1978). Infectious Bursal Disease Emulsified

Vaccine: Effect upon Neutralizing-Antibody Levels in the Dam and Subsequent Protection of the Progeny. Avian Dis., 32:466-470.

Benton, W.J., Cover, M.S. and Rosenberger, J.K. (1967). Studies on the

transmission of the infectious bursal agent of (IBA) of chickens. Avian Dis., 11:430-438.

Birgham, C., Mundt, E. and Gorbalenya, A.E. (2002). A non-conical proteinase

lacking the ATPase domain employs the ser-lys catalic dyad to exercise broad control over the life cycle of a double-stranded RNA virus. Embo. J., 114-123.

Bomford, R. (1998). Will adjuvants be needed for vaccines of the future? Dev.

Biol. Stand., (92):13–17. Boot, H.J., Huurne, A.A., Hoekman, A.J.W., Peeters, B.P.H. and Gielkens,

A.L.J. (2000). Rescue of very virulent and mosaic infectious bursal disease virus from cloned cDNA: VP2 is not the sole determinant of the very virulent phenotype. J. Virol., 74:6701-6711.

Bottcher, B., Kiselev N.A., Stel'Mashchuk, V.Y., Perevozchikova N.A., Borisov,

A.V. and Crowther, R.A. (1997). Three-dimensional structure of infectious bursal disease virus determined by electron cryomicroscopy. J. Virol., 71(1):325-30.

Bottomly, K. (1988). Afunctional dichotomy in CD4 lymphocytes. Immunol.

Today, 9:268-279. Box, P.G. (1989). High maternal antibodies help chickens beat virulent virus.

World Poult., 53:17-9. Boyle, D.B. and Heine, H.G. (1993). Recombinant fowlpox virus vaccines for

poultry. Immunol Cell Biol., 71:391-7. Boyle, D.B. and Heine, H.G. (1994). Influence of dose and route of inoculation

on responses of chickens to recombinant fowlpox virus vaccines. Vet Microbiol., 41(1-2):173-81.

Brady, J. (1970). Litter mites and their effects on poultry. World Poult Sci J.,

26(3):658-68.

Page 263: Thesis

233

Brandt, M., Yao, K., Liu, M., Heckert, R.A. and Vakharia, V.N. (2001). Molecular determinants of virulence, cell tropism, and pathogenic phenotype of infectious bursal disease virus. J Virol., 75:11974-82.

Broderson, J.R. (1989). A retrospective review of lesions Associated with the

use of Freund‟s adjuvant. Lab. Anim. Sci., 39:400-405. Brown, F., (1997). Viral vaccines. In: Vaccine Manual. The production and

quality control of veterinary vaccines for use in developing countries. FAO, UN. Rome, Italy :35-40.

Brown, M.D. and Skinner, M.A. (1996). Coding sequences of both genome

segments of a European 'very virulent' infectious bursal disease virus. Virus Res., 40:1-15.

Brown, M.D., Green, P. and Skinner, M. A. (1994). VP2 sequences of recent

European 'very virulent' isolates of infectious bursal disease virus are closely related to each other but are distinct from those of 'classical' strains. J Gen Virol., 75 (3):675-80.

Buonavoglia, C., Iovane, G., Trani, L.D., Falcone, F., Orefice, L. and Di Trani, L.

(1988). Evaluation of the immunogenicity in pigs guineapigs and chickens of an inactivated porcine parvovirus vaccine. Acta Medica Veterinaria., 34:445–452.

Calder, P.C. and Kew, S. (2002). The immune system: a target for functional

food. British J. of Nutr., 88:156-176. Cao, Y.C., Shi, Q.C., Ma, J.Y., Xie, Q.M. and Bi, Y.Z. (2005). Vaccination

against very virulent infectious bursal disease virus using recombinant T4 bacteriophage displaying viral protein VP2. Acta Biochim. Biophys. Sin. (Shangahai)., 37: 657-664.

Cao, Y.C., Yeung, W.S., Law, M., Bi, Y.Z., Leung, F.C. and Lim, B.L. (1998).

Molecular characterization of seven Chinese isolates of infectious bursal disease virus: classical, very virulent, and variant strains. Avian Dis., 42:340-51.

Chan, M.M., Chen, C.H., Ager, L.L. and Cooper, M.D. (1988). Identification of

the avian homologues of mammalian CD4 and cD8 antigens. J. Immunol., 140:2133-2138.

Chang, H.C., Lin, T.L. and Wu, C.C. (2001). DNA-mediated vaccination against

infectious bursal disease in chickens. Vaccine, 20(3-4):328-35.

Page 264: Thesis

234

Chang, H.C., Lin, T.L. and Wu. C.C. (2003). DNA vaccination with plasmids containing various fragments of large segment genome of infectious bursal disease virus. Vaccine, 21(5-6):507-13.

Chen, C.H., Pickel, J.M., Lahti, J.M. and Cooper, M.D. (1991). Surface markers

on avian immune cells. In “Avian cellular immunology” (J.M. Sharma, Ed.), CEC press, Boca Raton, FL, pp. 1-22.

Chen, H.Y., Zhou, Q., Zhang, M.F. and. Giambrone, J.J. (1998). Sequence

analysis of the VP2 hypervariable region of nine infectious bursal disease virus isolates from mainland China. Avian Dis., 42:762-9.

Chettle, N., Stuart J.C., and Wyeth, P.J. (1989). Outbreak of virulent

infectious bursal disease in East Anglia. Vet Rec., 125:271. Cheville, N.F. (1967). Studies on the pathogenesis of Gumboro disease in the

bursa of Fabricius, spleen and thymus of the chicken. Am J Pathol., 51:527-551.1967.

Cho, B.R. (1970). Experimental dual infections of chickens with infectious bursal

and Marek's disease agents. I. Preliminary observation on the effect of infectious bursal agent on Marek's disease. Avian Dis., 14(4):665-75.

Cho, B.R., Raymond, R.G. and Hill, R.W. (1979). Growth of infectious bursal

disease virus with plaque formation in chick embryo fibroblast cell culture. Avian Dis., 23: 200–17.

Confer, A.W., Springer, W.T., Shane, S.M. and Donovan, J.F. (1981).

Sequential mitogen stimulation of peripheral blood lymphocytes from chickens inoculated with infectious bursal disease virus. Am J Vet Res., 42:2109-13.

Constant, S.L. and Bottomly, K. (1997). Induction of Th1 and Th2 CD4 T cell

response: alternative approaches. Annu. Rev. Immunol., 16:297-322. Corley, M.M. and Giambrone, J.J. (2002). Immunosuppression in specific-

pathogen-free broilers administered infectious bursal disease virus vaccines by In ovoroute. Avian Dis., 46(4):810-815.

Corley, M.M., Giambrone, J.J. and Dormitorio, T.V. (2001). Detection of

infectious bursal disease vaccine viruses in lymphoid tissues after In ovovaccination of specific-pathogen-free embryos. Avian Dis., 45(4):897-905.

Cosgrove, A.S. (1962). An apparently new disease of chickens-Avian

Nephrosis. Avian Dis., 6:385-389.

Page 265: Thesis

235

Coulibaly, F., Chevalier, C., Gutsche, I., Pous, J., Navaza, J., Bressanelli, S., Delmas, B. and Rey, F.A. (2005). The birnavirus crystal structure reveals structural relationships among icosahedral viruses. Cell., 120: 761-772.

Cowen, B.S. and Braune, M.O. (1988). The propagation of avian viruses in a

continuous cell line (QT35) of Japanese quail origin. Avian Dis., 32:282-97.

Cullen, G.A. and Wyeth, P.J. (1976). Response of growing chickens to an

inactivated IBD antigen in oil emulsion. Vet Rec., 99(21):418. Cursiefen, D., Kaufer, I. and Becht, H. (1979) Loss of virulence in a small plaque

mutant of the infectious bursal disease virus. Arch of Virol., 59: 39-46. Da Costa, B., Soignier, S., Chevalier, C., Henry, C., Thory, C., Huet J.C. and

Delmas, B. (2003). Blotched snakehead virus is a new aquatic birnavirus that is slightly more related to avibirnavirus than to aquabirnavirus. J Virol., 77:719-25.

Darteil, R., Bublot, M., Laplace, E., Bouquet, J.F., Audonnet, J.C. and Riviere,

M. (1995). Herpesvirus of turkey recombinant viruses expressing infectious bursal disease virus (IBDV) VP2 immunogen induce protection against an IBDV virulent challenge in chickens. Virol., 211(2):481-90.

Devereux, G. (2002). The immune system an Overview. In: Calder, C.J., Field

and Gill, H.S.(Eds). Nut and imm fun., 4:1175-1182. Dilovski, M. and Tekerlekov, P. (1983). Comparative testing of the quality of

foot-and-mouth disease vaccines prepared with different virus inactivators. Vet Med Nauki., 20(5-6):41–45.

Dobos, P., Hill, B.J., Hallett, R., Kells, D.T., Becht, H. and Teninges, J.D.

(1979). Biophysical and biochemical characterization of five animal Viruses with bisegmented double-stranded RNA genomes. J Virol., 32:593-605.

Dohms, J.E., Lee, K.P. and Rosenberger, J.K. (1981). Plasma cell changes in

the gland of Harder following infectious bursal disease virus infection of the chicken. Avian Dis., 25:683-95.

Dormitorio, T.V., Giambrone, J.J. and Duck, L.W. (1997). Sequence

comparisons of the variable VP2 region of eight infectious bursal disease virus isolates. Avian Dis., 41:36-44.

Page 266: Thesis

236

Dybing, J.K. and Jackwood, D.J. (1998). Antigenic and immunogenic properties of baculovirus-expressed infectious bursal disease viral proteins. Avian Dis., 42(1):80-91.

Edgar, S.A. and Cho, Y. (1965). Avian nephrosis (Gumboro disease) and its

control by immunization. Poult Sci., 44:1366 (abstract). Edwards, K.R., Muskett, K.C. and Thornton, D.H. (1982). Duration of

imunosuppression caused by a vaccine strain of infectious bursal disease virus. ResVet Sci., 32: 79-83.

Edwards, W.A. (1981). Subclinical infectious bursal disease in the broiler

industry: Interim report. Vet Rec., 81:88-89. Erf, G.F. (2004). Cell-mediated immunity in poultry. Poultry Sci., 83: 850-590. Eterradossi, N., Arnauld, C., Toquin, D. and Rivallan, G. (1998). Critical amino

acid changes in VP2 variable domain are associated with typical and atypical antigenicity in very virulent infectious bursal disease viruses. Arch Virol., 143:1627-36.

Fabio, D.J., Rossini, L.I., Eterradossi, N., Toquin, M.D. and Gardin, Y. (1999).

European-like pathogenic infectious bursal disease viruses in Brazil. Vet Rec., 145, 203-204.

Fadley, A.M. and Nazerian, K. (1983). Pathogenesis of infectious bursal disease in chickens infected with virus at various ages. Avian Dis., 27: 714-723.

Fadley, A.M., Winterfield, R.W. and Olander, H.J. (1976). Role of the bursa of

Fabricius in the pathogenicity of inclusion body hepatitis and infectious bursal disease viruses. Avian Dis., 20(3):467-77.

Fahey, K.J., Erny, K. and Crooks, J. (1989). A conformational immunogen on

VP-2 of infectious bursal disease virus that induces virus neutralizing antibodies that passively protect chickens. J Gen Virol., 70 (6):1473-81.

Fahey, K.J., McWaters, P., Brown, M. A., Erny, K., Murphy, VJ. and. Hewish,

D.R. (1991). Virus-neutralizing and passively protective monoclonal antibodies to infectious bursal disease virus of chickens. Avian Dis., 35:365-73.

Fahey, KJ., O'Donnell, I.J. and Bagust, T.J. (1985). Antibody to the 32K

structural protein of infectious bursal disease virus neutralizes viral infectivity in vitro and confers protection on young chickens. J Gen Virol., 66:2693-702.

Page 267: Thesis

237

Faragher, J.T., Allan, W.H. and. Wyeth, P.J. (1974). Immunosuppressive effect of infectious bursal agent on vaccination against Newcastle disease. Vet Rec., 95:385-8.

Fernandez-Arias, A., Martinez, S. and Rodriguez, J.F. (1997). The major

antigenic protein of infectious bursal disease virus, VP2, is an apoptotic inducer. J Virol., 71:8014-8.

Firth, G.A. (1974). Letter: Occurrence of an infectious bursal syndrome within an

Australian poultry flock. Aust Vet J., 50(3):128-30. Fiza, M.I., Hair-Bejo, M., Omar, A.R. and Aini, I. (2006). Molecular

characterization of recent infectious bursal disease virus isolates from Malaysia. Acta Virol., 50(1): 45-51.

Fodor, I., Horvath, E., Fodor, N., Nagy, E., Rencendorsh, A., Vakharia, V.N. and

Dube, S.K. (1999). Induction of protective immunity in chickens immunised with plasmid DNA encoding infectious bursal disease virus antigens. Acta Vet Hung., 47(4):481-92.

Gaudry, (1993). Experience with very virulent IBDV. IN: proceeding of the 13th

Annual Meeting of the American Veterinary Association, Minneapolis, Minnesota, pp. 156.

Gelb, J., Nix, WA. and Gellman, S.D. (1998). Infectious bronchitis virus

antibodies in tears and their relationship to immunity. Avian Dis., 42(2):364-74.

Giambrone, J.J. (1979). Effects of early infectious bursal disease virus infection

on immunity to Newcastle disease in adult chickens. Poult Sci., 58(4):794-8.

Giambrone, J.J., Eidson, C.S. and Kleven, S.H. (1977). Effect of infectious

bursal disease on the response of chickens to Mycoplasma synoviae, Newcastle disease virus, and infectious bronchitis virus. Am J Vet Res., 38:251-3.

Giambrone, J.J., Eidson, C.S., Page, R.K., Fletcher, O.J., Barger, B.O. and

Kleven, S.H. (1976). Effect of infectious bursal agent on the response of chickens to Newcastle disease and Marek's disease vaccination. Avian Dis., 20(3):534-44.

Guittet, M., LeCoq, H., Picault, J.P., Eterradossi, N. and Bennejean, G. (1992).

Safety of infectious bursal disease vaccines: assessment of an acceptability threshold. Dev Biol Stand., 79:147-52.

Page 268: Thesis

238

Gupta, A.K., Oberoi, M.S. and Maiti, N.K. (2001). Detection of infectious bursal disease virus strains of varying pathogenicity under field conditions. Indian J. Comp. Microbiol. Immunol. Infect. Dis., 22:26-28.

Guvenc, T., Haziroglu, R., Yarim, M. and Tunca, R. (2004) Diagnosis of

infectious bursal disease by immunoperoxidase technique.

Vet Fak Derg., 51:237-238.

Habib, M., Iftikhar, H., Hamid, I., Zong, Y., Jiang, S. and Ning, N. (2006). Immunogenicity of formaldehyde and binary ethylenimine inactivated infectious bursal disease virus in broiler chicks. J Zhejiang Univ Science B., 7(8):660-664.

Haddad, E.E., Whitfill, C.E., Avakian, A.P., Ricks, C.A., Andrews, P.D., Thoma,

J.A. and Wakenell, P.S. (1997). Efficacy of a novel infectious bursal disease virus immune complex vaccine in broiler chickens. Avian Dis., 41(4):882-9.

Hair-Bejo, M. (1992). An outbreak of infectious bursal disease in broiler. InThe

Forth Veterinary Association Malaysia Congress, Kuala Lumpur, Malaysia, pp. 35-36.

Hair-Bejo, M. (1993). Infectious bursal disease in broilers: pathological changes

and virus detection. J. Vet. Malaysia, 5:49-51. Hair-Bejo, M., Hafiza, H., Moonafizad, M. and Aini, I. (1995a). Safety and

efficacy of an infectious bursal disease vaccine in replacement layer. In proc. of 7th Veterinary Association Malaysia Scientific Congress., October 6-8, 1995, Saremban, pp. 51-53.

Hair-Bejo, M., Salina, S., Hafiza, H. and Julaida, S. (2000). In ovovaccination

against infectious bursal disease in broiler chickens. J. Vet. Malaysia, 12:63-69.

Hair-Bejo, M., ThuZar, T., Moonafizd, M. and Aini, I. (1995b). Safety and

efficacy of infectious bursal disease vaccine in commercial broilers. In proc. of 7th Nathional Biotechnology Seminar., November, 20-22, 1995, Langkawi, pp. 138-141.

Hanson, B.S. (1962). Post- mortem lesions diagnostic of certain poultry lesions.

Vet Rec., 80:109-119. Hassan, M.K. and Saif, Y.M. (1996). Influence of the host system on the

pathogenecity, immunogenecity and antigenecity of infectious bursal disease virus. Avian Dis., 40:553-561.

Page 269: Thesis

239

Hassan, M.K., Natour, M.Q,.Ward L.A. and Saif, Y.M. (1996). Pathogenicity, attenuation and immunogenicity of infectious bursal disease virus. Avian Dis., 40:567-571.

Hassan, N., Khashaba, E., Bording, F.E., Asily, S.E., Ebiary, E.E., Wahab, S.

and Barhoma, N. (1992). Trail for production of aluminum hydroxide gel and oil adjuvant inactivated Newcastle disease vaccines in comparison with commercial oil vaccine. Assiut Vet. Med. J., 27:123-133.

Heckert, A., Elankumaran, S., Oshop, G.L. and Vakharia, V.N. (2002). A novel

transcutaneous plasmid-dimethylsulfoxide delivery technique for avian nucleic acid immunization. Vet Immunol Immunopathol., 89:(1-2):67-81.

Heine, H.G. and Boyle, D.B. (1993). Infectious bursal disease virus structural

protein VP2 expressed by a fowlpox virus recombinant confers protection against disease in chickens. Arch Virol., 131(3-4):277-92.

Heine, H.G., Haritou, M., Failla, P., Fahey, K. and Azad, A. (1991). Sequence

analysis and expression of the host-protective immunogen VP2 of a variant strain of infectious bursal disease virus which can circumvent vaccination with standard type I strains. J Gen Virol., 72 (8):1835-43.

Helmboldt, C.F. and Garner, E. (1964). Experimentally induced Gumboro

disease (IBD). Avian Dis., 8:561-575. Hirai, K. and Calnek, B.W. (1979). In vitro replication of infectious bursal disease

virus in established lymphoid cell lines and chicken B lymphocytes. Infect Immun., 25(3):964-70.

Hirai, K. and Shimakura, S. (1974). Structure of infectious bursal disease Virus.

J. of Virol, 14: 957-964. Hirai, K., Funakoshi, T., Nakai, T. and. Shimakura S. (1981). Sequential

changes in the number of surface immunoglobulin-bearing B lymphocytes in infectious bursal disease virus-infected chickens. Avian Dis., 25:484-96.

Hirai, K., Shimakura, S., Kawamoto, E., Taguchi, F., Kim, S.T., Chang, C.N.

and Iritani Y. (1974). The immunodepressive effect of infectious bursal disease virus in chickens. Avian Dis., 18:50-7.

Hitchner, S.B. (1970). Infectivity of infectious bursal disease virus for

embryonating eggs. Poult Sci., 49:511-16.

Page 270: Thesis

240

Howie, R. and Thorsen, J. (1981). An enzyme-linked immunosorbent assay (ELISA) for infectious bursal disease virus. Can J Comp Med., 45(1):51–55.

Hsieh, M.K., Wu, C.C. and Lin, T.L. (2007). Priming with DNA vaccine and

boosting with killed vaccine conferring protection of chickens against infectious bursal disease. Vaccine, 25:7629-7635.

Hudson, P.J., McKern, N.M., Power, B.E. and Azad, A.A. (1986). Genomic

structure of the large RNA segment of infectious bursal disease virus. Nucleic Acids Res., 14:5001-12.

Hurst, J.K., Barrete, W.C., Michel, B.R. and Rosen, H. (1991). Hypochlorous

acid and myeloperoxidase-catalyzed oxidation of iron-sulfur clusters in bacterial respiratory dehydrogenases. Eur. J. Biochem., 202:1275-1282.

Hyuk, M.K. and Soo, J. K.(2004). Sequence analysis of the variable VP2 Gene

of Infectious Bursal Disease Viruses Passaged in Vero Cells. Virus Genes, 28:(3):285–291.

Igbokwe, I.O., Salako, M.A., Rabo, J.S. and Hassan S.U. (1996). Outbreak of

infectious bursal disease associated with acute septicaemic colibacillosis in adult prelayer hens. Rev Elev Med Vet Pays., Trop 49(2):110-3.

Inoue, M., Fukuda, M. and Miyano, K. (1994). Thymic lesions in chicken

infected with infectious bursal disease virus. Avian Dis., 38:839-46. Ismail, N.M., Saif, Y.M. and Moorhead, P.D. (1988). Lack of pathogenicity of five

serotype 2 infectious bursal disease viruses in chickens. Avian Dis., 32(4):757-9.

Ismail, N.M. and Saif, Y.M. (1990). Differentiation between antibodies to

serotypes 1 and 2 infectious bursal disease viruses in chicken sera. Avian Dis., 34:1002-4.

Ismail, N.M. and Saif, Y.M. (1991). Immunogenicity of infectious bursal disease

viruses in chickens. Avian Dis., 35:460-9. Ismail, N.M., Saif, Y.M., Wigle, W.L., Havenstein, G.B. and Jackson, C. (1990).

Infectious bursal disease virus variant from commercial leghorn pullets. Avian Dis., 34: 141-145.

Ivan, J., Nagy, N., Magyar, A., Kacskovics, I. and Meszaros, J. (2001).

Functional restoration of the bursa of Fabricius following In

Page 271: Thesis

241

ovoinfectious bursal disease vaccination. Vet Immunol Immunopathol., 79(3-4):235-48.

Ivanyi, J. (1975). Immunodeficiency in the chicken. II. Production of monomeric

IgM following testosterone treatment or infection with Gumboro disease. Immunology, 28:1015-21.

Ivanyi, J. and Morris, R. (1976). Immunodeficiency in the chicken. IV. An

immunological study of infectious bursal disease. Clin Exp Immunol., 23(1): 154-65.

Jackson, L.R. and Fox, J.G. (1995). Institutional policies and guidelines on

adjuvants and antibody production. ILAR Journal, 37(3):141-150. Jackwood, D J., Saif, Y.M. and Hughes, J. H. (1987). Replication of Infectious

bursal disease virus in continuous cell lines. Avian Dis., 31: 370-375. Jackwood, D.H. and Saif, Y.M. (1987). Antigenic diversity of infectious bursal

disease viruses. Avian Dis., 31(4):766-70. Jackwood, D.J. (2005). The role of vaccination on the control of infectious bursal

disease virus. 77th Northeastern Conference on avian Disease. Pp 1-3. Jackwood, D.J. and Sommer, S.E. (1999). Restriction fragment length

polymorphisms in the VP2 gene of infectious bursal disease viruses from outside the United States. Avian Dis., 43:310-4.

Jackwood, D.J., Jackwood, R.J. and Sommer, S.E. (1997). Identification and

comparison of point mutations associated in classic and variant infectious bursal disease viruses. Virus Res., 49: 131 – 137.

Jackwood, D.J., Saif, Y.M. and Hughes, J.H. (1982). Characteristics and

serologic studies of two serotypes of infectious bursal disease virus in turkeys. Avian Dis., 26(4):871-82.

Jackwood, D.J., Saif. Y.M. and Moorhead, P.D. (1985). Immunogenicity and

antigenicity of infectious bursal disease virus serotypes I and II in chickens. Avian Dis., 29(4):1184-94.

Jagadish, M.N., Staton, V.J., Hudson, P.J. and Azad, A.A. (1988). Birnavirus

precursor polyprotein is processed in Escherichia coli by its own virus-encoded polypeptide. J Virol., 62:1084-7.

Janeway, C.A., Travers, P., Walport, M. and Shlomchik, M. (2001). Immunology:

The immune system in health and disease 5th ed. Garland Publishing, New York.

Page 272: Thesis

242

Jeurissen, S.H., Janse, E.M., Lehrbach, P.R., Haddad, E.E., Avakian, A. and Whitfill, C.E. (1998). The working mechanism of an immune complex vaccine that protects chickens against infectious bursal disease. Immunology, 95(3):494-500.

Johnston, P.A., Liu, H., O'Connell, T., Phelps, P., Bland, M., Tyczkowski, J.,

Kemper, A., Harding, T., Avakian, A., Haddad, E., Whitfill, C., Gildersleeve, R. and Ricks, C.A. (1997). Applications in In ovo technology. Poult Sci., 76(1):165-78.

Kaufer I. and Weiss, E. (1980). Significance of bursa of Fabricius as target

organ in infectious bursal disease of chickens. Infect Immun., 27(2):364-7.

Kaufer, I. and Weiss, E. (1976). Electron-microscope studies on the

pathogenesis of infectious bursal disease after intrabursal application of the causal virus. Avian Dis., 20:483-496.

Kelemen, M., Forgach, K., Ivan, J., Palya, V., Suveges, T., Toth, B. and

Meszaros, J. (2000). Pathological and immunological study of an In ovocomplex vaccine against infectious bursal disease. Acta Vet Hung., 48(4):443-54.

Kibenge, F.S. and Dhama, V. (1997). Evidence that virion-associated VP1 of

avibirnaviruses contains viral RNA sequences. Arch Virol., 142:1227-36. Kibenge, F.S. and McKenna, P.K. (1992). Isolation and propagation of infectious

bursal disease virus using the ovine kidney continuous cell line. Avian Dis., 36:256-261.

Kibenge, F.S., Dhillon, A.S. and. Russell, R.G. (1988). Growth of serotypes I

and II and variant strains of infectious bursal disease virus in Vero cells. Avian Dis., 32(2):298-303.

Kibenge, F.S., Jackwood, D.J. and Mercado C.C. (1990). Nucleotide sequence

analysis of genome segment A of infectious bursal disease virus. J. Gen. Virol., 71(3):569-77.

Kibenge, F.S., McKenna, P.K. and Dybing, J.K. (1991). Genome cloning and

analysis of the large RNA segment (segment A) of a naturally avirulent serotype 2 infectious bursal disease virus. Virol., 184:437-40.

Kim, I.J., Karaca, K.T.L., Pertile, S.A., Erickson, and Sharma, J.M. (1999).

Enhanced expression of cytokine genes in spleen macrophages during acute infection with infectious bursal disease virus in chickens. Vet Immunol Immunopathol., 61:331-41.

Page 273: Thesis

243

Kim, J.K., Fahad, A.M., Shanmukhappa, K. and Kapil, S. (2006). Defining the cellular target(s) of porcine reproductive and respiratory syndrome virus blocking monoclonal antibody 7G10. J. virol., 80 (2): 689-696.

King, D.J. (1991). Evaluation of different methods of inactivation of Newcastle

disease virus and avian influenza virus in egg fluids and serum. Avian Dis., 35(3):505–514.

Kleinman, N.R., Kier, A.B., Diaconu, E. and Lass, J.H. (1993). Posterior paresis

induced by Freund's adjuvant in guinea pigs. Lab. Anim. Sci., 43:364-366.

Kuby, N.P. (1994). Vaccines. In: Immunology 2nd Edition. W.H. Freeman and

Company. USA. PP: 469-482. Kwon, H.M. and Kim, S.J. (2004). Sequence analysis of the variable VP2 gene

of infectious bursal disease viruses passaged in Vero cells. Virus Genes, 28: 285-291.

Lam, K.M. (1998). Alteration of chicken heterophil and macrophage functions by

the infectious bursal disease virus. Microb Pathog., 25:147-55. Lana, D.P., Beisel, C.E. and Silva, R. F. (1992). Genetic mechanisms of

antigenic variation in infectious bursal disease virus: analysis of a naturally occurring variant virus. Virus Genes, 6:247-25.

Landgraf, H., Vielitz, E. and Kirsch, R. (1967). Occurence of an infectious

disease affecting the bursa of Fabricius (Gumboro disease). Dtsch Tierarztl Wochenschr., 74:6-10.

Lange, H., Mjller, H., Ufer, I. and Becht, H. (1987). Pathogenic and structural

properties of wild type infectious bursal disease virus (IBDV) and virus grown in vitro. Archives of Virology, 92:187-196.

Larghi, O.P. and Nebel, A.E. (1980). Rabies virus inactivation by binary

ethylenimine: new method for inactivated vaccine production. Journal of Clinical Microbiology, 11(2):120–122.

Lascelles, A.K., Eagleson. G., Beh, K.J. and Watson, D.L.(1989). Significance of

Freund‟s adjuvent/antigen injection granuloma in the maintenance of serum antibody response. Vet. Immunol. Immunopathol., 22(1): 15-27

Lasher, H. and Shane, S. (1994). Infectious bursal disease virus. World's Poult.

Sci. J., 50:133-166.

Page 274: Thesis

244

Lee, L.H. and Lukert, P.D. (1986). Adaptation and antigenic variation of infectious bursal disease virus. J. Chin. Soc. Vet. Sci., 12:297-304

Lejal, N., Da Costa, B., Huet, J. C. and Delmas, B. (2000). Role of Ser-652

and Lys-692 in the protease activity of infectious bursal disease virus VP4 and identification of its substrate cleavage sites. J Gen Virol., 81:983-92.

Leonard, J. (1974). Immunogenicity of cell culture adapted infectious bursal

disease virus. M.Sc. Thesis, University of Georgia, Athens. G.A. Leong, J.C., Brown, D., Dobos, P., Kibenge, F. S. B., Ludert, J. E., Müller, H.,

Mundt, E. and Nicholson, B. (2000). Family Birnaviridae. In Virus Taxonomy. Seventh Report of the International Committee on the Taxonomy of Viruses, pp. 481-490. Edited by M.H.V. van Regenmortel, C. M. Fauquet, D.H.L. Bishop, E.B. Carstens, M. K. Estes, S. M. Lemon, J. Maniloff, M.A. Mayo, D.J. McGeoch, C. R. Pringle & R. B. Wickner. San Diego: Academic Press.

Lillie, R.D. (1965). Histopathologic technic and practical histochemistry. (3d ed).

Mac Graw book company, pp 142. Lim, B.Y., Cao, T.Y. and Mo, C. (1999). Adaptation of very virulent infectious

bursal disease virus to chicken embryonic fibriblasts by site-direct mutagenesis of residues 279 and 284 of viral coat protein VP2. J. Virol., 73:2854-2862.

Lin, T.W., Lo, C.W., Lai, S.Y., Fan, R.J., Lo, C.J., Chou, Y.M., Thiruvengadam,

R., Wang, A.H. and Wang, M.Y. (2007). Chicken heat shock protein 90 is a component of the putative cellular receptor complex of infectious bursal disease virus. J. Virol., 81(16): 8730-41.

Lin, Z., Kato, A., Otaki, Y., Nakamura T., Sasmaz E. and Ueda, S. (1993).

Sequence comparisons of a highly virulent infectious bursal disease virus prevalent in Japan. Avian Dis., 37(2):315-23.

Liu, H., Giambrone, J.J. and Dormitorio T. (1994). Detection of genetic

variations in serotype 1 isolates of infectious bursal disease virus using polymerase chain reaction and restriction endonuclease analysis. J. Virol. Meth., 48: 281-291.

Liu, M. and Vakharia, V.N. (2004). VP1 protein of infectious bursal disease virus

modulates the virulence in vivo. Virol., 330: 62–73. Lombardo, E., Maraver, A., Castn, J.R., Rivera, J., Fernandez-Arias, A.,

Serrano, A., Carrascosa, J. L. and Rodriguez, J.F. (1999). VP1, the

Page 275: Thesis

245

putative RNA-dependent RNA polymerase of infectious bursal disease virus, forms complexes with the capsid protein VP3, leading to efficient encapsidation into virus-like particles. J Virol., 73:6973-83.

Lombardo, E., Maravet, A., Espinosa, I., Fernandez- Arias, A. and Rodriguez,

J.F. (2000). VP5, the nonstructural polypeptide of infectious bursal disease virus, accumulates within the host plasma membrane and induces cell lysis. Virol., 277: 345-357.

Lucio, B. and Hitchner, S.B. (1979). Infectious bursal disease emulsified

vaccine:Effect upon neutralizing-antibody levels in the dam and subsequent protection of the progeny. Avian Dis., 23:466-478.

Lucio, B. and Hitchner, S.B. (1980). Immunosuppression and active response

induced by infectious bursal disease virus in chickens with passive antibody. Avian Dis., 24:189-96.

Lucio, B., Morales, G.A., Toscamo, C.A. and Chape, B.J. (1996). Challenge

protection with specific infectious bursal disease virus strains. Proceedings of the Western Poultry Disease Conference, University of California, 45:178-182.

Lukert, P.D. and Hitchner, S.B. (1984). Infectious bursal disease. In: Disease of

poultry (8th ed.). Iowa, state University Press, Ames USA, pp. 566-576. Lukert, P.D. and saif, Y.M. (1997). Infectious bursal disease. In: Disease of

poultry (11th ED) Iowa state University press, Ames Iowa. YSE. pp: 721-738.

Lukert, P.D. and. Davis, R. B. (1974). Infectious bursal disease virus: growth

and characterization in cell cultures. Avian Dis., 18:243-50. Lukert, P.D., and Saif, Y.M. (2003). Infectious Bursal Disease. In: Diseases of

Poultry, 11th ed. Y. M. Saif, H. J. Barnes., A. M. Fadly., J. R. Glisson., L. R. McDougald., and D. E. Swayne., eds. Iowa State Press, Ames, Iowa., pp. 161-179.

Lukert, P.D., Leonaed, J. and Davis, R.B. (1975). Infectious bursal disease

antigen production and immunity. Am. J. Vet. Res., 36:539-540. Luna, G. (1968). Manual of Histological Staining Method of the Armed Forces Institute of Pathology. 3rd ed., McGraw-Hill Book Company, New York. Maas, R.A., venema, S., Oei, H.L., pol, M.A., Claassen, J.T.M. and Huurne,

A.H.M. (2001). Effecacy of inactivated infectious bursal disease (IBD) vaccine: coparison of serology with protection of progeny chickens

Page 276: Thesis

246

against IBD virys strain of varying virulence. Avian pathol., (30): 345-354.

Macreadie, I.G., Vaughan, P.R., Chapman, A.J., McKern, N.M., Jagadish, M.N.,

Heine, H.G., Ward, C.W., Fahey, K.J. and Azad, A.A. (1990). Passive protection against infectious bursal disease virus by viral VP2 expressed in yeast. Vaccine, 8(6):549-52.

Martin, H., Foster, N.D., Bottoli, I., Iacovoni, S.J. and Peter, K.V. (1998). The

DF-1 Chicken Fibroblast Cell Line: Transformation Induced by Diverse Oncogenes andCell Death Resulting from Infection by Avian Leukosis Viruses. Virology, 248, 295–304.

Martinez-Torrecuadrada, J.L., Saub, I.N., Pages-Mante, A., Caston, J.R.,

Espuna, E. and Casal, J.I. (2003). Structure-dependent efficacy of infectious bursal disease virus (IBDV) recombinant vaccines. Vaccine, 21(23):3342-50.

Mathur, S., Kapoor, S. and Kalra, S.K. (1999). Adaptation of K2 strain of IBD

virus in CEF cell cultures. Indian J. Comp. Microbiol. Immunol. Infect. Dis., 20: 68-69.

Mazariegos, L.A., Lukert, P.D. and Brown, J. (1990). Pathogenicity and

immunosuppressive properties of infectious bursal disease "intermediate" strains. Avian Dis., 34(1):203-8.

McDougald, L.R., Karlsson, T. and Reid, W.M. (1979). Interaction of infectious

bursal disease and coccidiosis in layer replacement chickens. Avian Dis., 23(4):999-1005.

McFerran, J.B., McNulty, M.S., McKillop, E.R., Conner, T.J., McCracken, R. M.,

Collins, D.S. and Allan, G. M. (1980). Isolation and serological studies with infectious bursal disease viruses from fowl, turkey and duck: Demonstration of a second serotype. Avian Pathol., 9:395-404.

McNamee P.T. (2000). Bacterial chondronecrosis with osteomyelitis ('femoral

head necrosis') of broiler chickens: a review. Avian Pathol., 29(4):253-81.

McNulty, M.S., Allan, G.M. and McFerran, J.B. (1979). Isolation of infectious

bursal disease virus from turkeys. Avian Pathol., 8:205-212. Medzhitov, R. and Janeway, C.A. (1997). Innate immunity. Cell, 91(3):295-298. Melchior, F.W.J. and Melson, L. (1989). Attenuation of infectious bursal disease

virus strai and vaccine thereform. United States Patent., No 4,824,668.

Page 277: Thesis

247

Mo, C.W., Cao, Y.C. and Lim, B.L. (2001). The in vivo and in vitro effects of chicken interferon alpha on infectious bursal disease virus and Newcastle disease virus infection. Avian Dis., 45(2):389-99.

Mohamed, K.M., Hassan, K. M.A. and Saif, Y.M. (1996a). Influence of the host

system on the pathogenicity and antigencity of infectious bursal disease virus. Avian Dis., 40:553-561.

Mohamed, K.M., Hassan, M.Q., AL-Natour, Ward, L.A., and saif, Y.M. (1996b).

Pathogenicity attenuation and immunogenicity of infectious bursal disease virus. Avian Dis., 40: 567-571.

Mohanty, G.C., Pandey, A.P. and. Rajya, B.S. (1971). Infectious bursal disease

virus in chickens. Cur. Sci., 40:181-184. Moradian, A., Thorsen, J. and Julian, R.J. (1990). Single and combined

infections of specific-pathogen-free chickens with infectious bursal disease virus and an intestinal isolate of reovirus. Avian Dis., 34(1):63-72.

Muller, H. and Becht, H. (1982). Biosynthesis of virus-specific proteins in cells

infected with infectious bursal disease virus and their significance as structural elements for infectious virus and incomplete particles. J Virol., 44:384-92.

Muller, H. and Nitschke, R. (1987). The two segments of the infectious bursal

disease virus genome are circularized by a 90,000-Da protein. Virol., 159:174-7.

Muller, H., Lange, H. and Becht, H. (1979a). Immunofluorescence studies of

early virus propagation after oral infection with infectious bursal disease virus (IBDV). Zentrabl. Veterimaedmed. Med., 26:345-352.

Muller, H., Scholtissek, C., and Becht, H. (1979b). The genome of infectious

bursal disease virus consists of two segments of double-stranded RNA. J Virol., 31:584-9.

Mundt, E. (1999). Tissue culture infectivity of different strains of infectious bursal

disease virus is determined by distinct amino acids in VP2. Journal of General Virology, 80, 2067-2076.

Mundt, E. and Muller H. (1995). Complete nucleotide sequences of 5'- and 3'-

noncoding regions of both genome segments of different strains of infectious bursal disease virus. Virol., 209:10-8.

Page 278: Thesis

248

Mundt, E., and Vakharia, V.N. (1996). Synthetic transcripts of double-stranded birnavirus genome are infectious. In :Proc. Natl. Acad. Sci. USA. 93:11131-11136.

Mundt, E., Beyer, J. and Muller, H. (1995). Identification of a novel viral protein

in infectious bursal disease virus-infected cells. J Gen Virol., 76 (2):437-43.

Mundt, E., Kollner, B. and Kretzschmar, D. (1997). VP5 of infectious bursal

disease virus is not essential for viral replication in cell culture. J Virol., 71:5647-51.

Muskett, J.C., Hopkins, I.G., Edwards, K.R. and Thornton, D.H. (1979).

Comparison of two infectious bursal disease vaccine strains: efficacy and potential hazards in and maternally immune birds. Vet Rec., 4: 332-334

Nagarajan, M.M. and. Kibenge, F.S. (1997). The 5'-terminal 32 basepairs

conserved between genome segments A and B contain a major promoter element of infectious bursal disease virus. Arch Virol., 142:2499-514.

Naqi, S., Thompson, G., Bauman, B., and Mohammed, H. (2001). The

exacerbating effect of infectious bronchitis virus infection on the infectious bursal disease virus-induced suppression of opsonization by Escherichia coil antibody in chickens. Avian Dis., 45(1):52-60.

Nicholas, K.B., Nicholas, H.B. and Deerfield, D.W. (1997). GeneDoc: Analysis

and visualization of genetic variation. Embnet news., 4:14. Nick, H., Cursiefen, D. and. Becht, H. (1976). Structural and growth

characteristics of infectious bursal disease virus. J Virol., 18:227-34.1976.

Norusis, M.J. (2004). SPSS 13.0 advanced statistical procedure companion.

Engelewood Cliffs: Prentice Hall. Nunoya, T., Otaki, Y., Tajima, M., Hiraga, M., and Saito, T. (1992). Occurrence

of acute infectious bursal disease with high mortality in Japan and pathogenecity of field isolated in specific pathogen free Chicken. Avian Dis., 36: 597-609.

Nunoya, T., Yagihashi, T., Tajima, M. and Nagasawa, Y. (1995). Occurrence of

keratoconjunctivitis apparently caused by Mycoplasma gallisepticum in layer chickens. Vet Pathol., 32(1):11-8.

Page 279: Thesis

249

Okoye, J.O.A. and Uzoukwo, M. (1984). 1. Histopathogenesis of infectious bursal disease in the bursa of Fabricius. 2.Persistence of infectious bursal virus and the appearance of precipitins in infected chickens. Tropical Veterinarian, 2: 91-102.

Oliveira, M.C., Figueiredo-Lima D.F., Faria Filho, D.E., Marques, R.H. and

Moraes, V.M.B. (2009). Effect of mannanoligosaccharides and/or enzymes on antibody titers against infectious bursal and Newcastle disease viruses Arq. Bras. Med. Vet. Zootec., 61 (1): 6-11.

Oppling, V., Muller, H. and Becht, H. (1991). The structural polypeptide VP3 of

infectious bursal disease virus carries group- and serotype-specific epitopes. J Gen Virol ., 72 (9):2275-8.

Oshop, G.L., Elankumaran, S., Vakharia, V.N. and Heckert, R.A. (2003). In

ovodelivery of DNA to the avian embryo. Vaccine, 21:1275–1281.

Owoade, A. A., Mulders, M.N., Kohnen, J., Ammerlaan, W. and Muller, C. P. (2004). High sequence diversity in infectious bursal disease virus serotype 1 in poultry and turkey suggests West-African origin of very virulent strains. Arch. Virol., 149:653-672.

Pages-Mante, A., Maldonado, D.T. and Saubi, N. (2004). Dog as carrier of

infectious bursal disease virus. Avian Pathol., 33(2), 205-209. Pahar, B. and Rai, A. (1997). The characterization of infectious bursal disease

virus strains/ isolates from field outbreaks in India. Vet. Res. Comm., 21: 289-301.

Parkhurst, R.T. (1964). On- the farm studies of Gumboro Disease in broilers.

Avian Dis., 8:584-596. Peilin, W., Baoxiang, C. and Jinsong, L. (1997). Adaptation and propagation of

infectious bursal disease in Vro cells. J. Vet. Res. Tech., 27:23-24. Pejkovski,C., Davelaar, F.G. and Kouwenhoven, B. (1979). Immunosuppressive

effect of infectious bursal disease virus on vaccination against infectious bronchitis. Avian Pathol., 8:95-106.

Petek, N., Aprile, N.D. and Cancellotti, F. (1973). Biological and physico-

chemical properties of the infectious bursal disease virus (IBDV). Avian Pathol., 2: 135-152.

Peters, G. (1967). Histology of Gumboro disease. Berl Munch Tierarztl

Wochensch., 80:394-396.

Page 280: Thesis

250

Phenix, K.V., Wark, K., Luke, C.J., Skinner, M.A., Smyth, J.A., Mawhinne, K.A. and Todd, D. (2001). Recombinant Semliki Forest virus vector exhibits potential for avian virus vaccine development. Vaccine, 19(23-24):3116-23.

Pitcovski, J., Di Castro, D., Shaaltiel, Y., Azriel, A., Gutter, B., Yarkon,i E.,

Michael, A., Krispel, S. and Levi, B.Z. (1996). Insect cell-derived VP2 of infectious bursal disease virus confers protection against the disease in chickens. Avian Dis., 40(4):753-61.

Pitcovski, J., Goldberg, D., Levi BZ, Di Castro D., Azriel A, Krispel S., Maray T.

and Shaaltiel Y. (1998). Coding region of segment A sequence of a very virulent isolate of IBDV--comparison with isolates from different countries and virulence. Avian Dis., 42(3):497-506.

Ramm, H.C., Wilson, T.J., Boyd, R.L., Ward, H.A., Mitrangas, K. and Fahey,

K.J. (1991). The effect of infectious bursal disease virus on B lymphocytes and bursal stromal components in specific pathogen-free (SPF) White Leghorn chickens. Dev Comp Immunol., 15(4):369-81.

Ramon, G. (1925). Sur l‟augmentation anormale de l‟antitoxine chez les

chevaux producteurs de se´rum antidiphte´rique. Bull Soc CentrmMed Vet., 101:227.

Rasool, M.H. and Hussain, I. (2006). Preparation and evaluation of Vero-cell

infectious bursal disease vaccine in Pakistan. Vaccine, 24: 2810–2814. Raue, R., Islam, M.N., Islam, K.M., Islam, S.C., Badhy, P.M., and Muller H.

(2004). Reversion of molecularly engineered, partially attenuated, very virulent infectious bursal disease virus during infection of commercial chickens. Avian Pathol., 33(2), 181-189.

Rautenschlein, S., Kraemer, C., Vanmarke J. and Montiel, E. (2005). Protective

efficacy of intermediate and intermediate plus infectious bursal disease virus IBDV vaccines against very virulent IBDV in commercial broilers. Avian. Dis., 49(2):231 -7.

Rautenschlein, S. and H.Y. Sharma, (2002). The role of T cells in protection by

an inactivated infectious bursal disease virus vaccine. Veterinary immunology and immunopathology, (89): 159-167.

Raymond, R.G. and Hill, R.W. (1979). Growth of infectious bursal disease virus

with plaque formation in chick embryo fibroblast cell culture. Avian Dis., 23(1):209-216.

Page 281: Thesis

251

Reed, L.J. and Muench, S. (1938) A simple method of estimating fifty percent end point. Am. J. Hyg., 27,493497.

Rinaldi, A., Cessi, D., Cervio, G. and Iodetti, T. (1972). Attenuation of infectious

bursal disease virus and vaccination trial under laboratory and field conditions. Avian pathol., 3:51-57.

Rodriguez-Chavez, I.R., Rosenberger, J.K. and Cloud, S.S. (2002).

Characterization of the antigenic, immunogenic, and pathogenic variation of infectious bursal disease virus due to propagation in different host systems (bursa,embryo, and cell culture). II. Antigenicity at the epitope level. Avian Pathol., 31: 473-483.

Rosales, A.G., Villegas, P, Lukert, P.D., Fletcher, O.J, Mohamed, M.A, Brown J.

(1989a). Pathogenicity of recent isolates of infectious bursal disease virus in specific-pathogen-free chickens: protection conferred by an intermediate vaccine strain. Avian Dis., 33(4):729-34.

Rosales, A.G., Villegas, P., Lukert, P.D., Fletcher, O.J. and Brown, J. (1989b).

Immunosuppressive potential and pathogenicity of a recent isolate of infectious bursal disease virus in commercial broiler chickens. Avian Dis., 33(4):724-8.

Rosenberg, J.K., Sharma, J. M., Belzer, S. W., Nordgren, R. M. and Naqi, S.

(1994). Flow cytometric analysis of B cell and T cell subpopulations in specific-pathogen-free chickens infected with infectious bursal disease virus. Avian Dis., 38(1), 16-21.

Rosenberger, J.K. and Gelb, J. (1978). Response to several avian respiratory

viruses as affected by infectious bursal disease virus. Avian Dis., 22(1):95-105.

Rosenberger, J.K. and Cloud, S.S. (1985). Isolation and characterization of

variant infectious bursal disease viruses. Abs. J. Am. Vet. Med. Asso., 198:357.

Rosenberger, J.K. and Cloud, S.S. (1986). Isolation and characterization of

variant infectious bursal disease virus. J Am Vet Med Assoc., 189:357. Rosenberger, J.K., and Cloud, S.S. (1989). The effects of age, route of

exposure, and coinfection with infectious bursal disease virus on the pathogenicity and transmissibility of chicken anemia agent (CAA). Avian Dis., 33(4):753-9.

Page 282: Thesis

252

Rudd, M. F., Heine, H. G., Sapats, S. I., Parede, L. and Ignjatovic, J. (2002) Characterization of an Indonesian very virulent strain of infectious Bursal disease virus. Arch. Virol., 147:1303-1322.

Sadekar, R.D., Kolte, A.Y., Barmase, B.S. and Desai, V.F. (1998).

Immunopotentiating effects of Azadirachta indica (Neem) dry leaves powder in broilers, naturally infected with IBD virus. Indian J Exp Biol., 36(11):1151-3.

Saif, Y.M. (1998). Infectious bursal disease and hemorrhagic enteritis. Poul.

Sci., 77:1186-1189. Sambrook, J., Fritsch, E.F. and Maniatis, T. (1989). Molecular Cloning: a

laboratory manual. 2nd ed. N.Y., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, 1989. 1659 p. ISBN 0-87969-309-6.

Sanchez, A.B. and Rodriguez J.F. (1999). Proteolytic processing in infectious

bursal disease virus: identification of the polyprotein cleavage sites by site-directed mutagenesis. Virol., 262:190-9.

Santivatr, D, Maheswaran, S.K., Newman, J.A. and Pomeroy, B.S. (1981).

Effect of infectious bursal disease virus infection on the phagocytosis of Staphylococcus aureus by mononuclear phagocytic cells of susceptible and resistant strains of chickens. Avian Dis., 25(2):303-11.

Sapats, S. I. and Ignjatovic, J. (2000). Antigenic and sequence heterogeneity of

infectious bursal disease virus strains isolated in Australia. Arch Virol., 145:773-85.

Schijns, V.E.J.C. (2000). Immunological concepts of vaccine adjuvant activity.

Curr. Opin. Immunol., 12: 456–463. Schnitzler, D., Bernstein, F., Muller H. and Becht, H. (1993). The genetic basis

for the antigenicity of the VP2 protein of the infectious bursal disease virus. J. Gen. Virol., 74 (8):1563-71.

Schroder, A., van Loon A.A., Goovaerts, D. and Mundt, E. (2000). Chimeras in

noncoding regions between serotypes I and II of segment A of infectious bursal disease virus are viable and show pathogenic phenotype in chickens. J. Gen. Virol., 81: (2):533-40.

Sharma, J. and Rosenberger, J.K. (1978). Infectious buesal disease and

reovirus infection of chickens: Immune response and vaccine control. Avian Immunity Basic Pratice, 21:149-150.

Page 283: Thesis

253

Sharma, J.M. and Fredericksen, T.L. (1987). Mechanism of T cell immunosuppression by infectious bursal disease virus of chickens. Prog. Clin. Biol. Res., 238:283-94.

Sharma, J.M., Dohms, J.E. and Metz, A.L. (1989). Comparative pathogenesis of

serotype 1 and variant serotype 1 isolates of infectious bursal disease virus and their effect on humoral and cellular immune competence of specific-pathogen-free chickens. Avian Dis., 33(1):112-24.

Sharma, J.M., Kim, I.J., Rautenschlein, S. and Yeh, H.Y. (2000). Infectious

bursal disease virus of chickens: pathogenesis and immunosuppression. Dev Comp Immunol., 24(2-3):223-35.

Shaw. I. and Davison, T.F. (2000). Protection from IBDV-induced bursal

damage by a recombinant fowlpox vaccine, fpIBD1, is dependent on the titre of challenge virus and chicken genotype. Vaccine, 18(28):3230-41.

Shcherbakova, L.O., Lomakin, A.I., Borisov, A.V., Drygin V.V. and Gusev, A.A.

(1998). Comparative analysis of the VP2 variable region of the gene from infectious bursal disease virus isolates. Mol Gen Mikrobiol Virusol., 35-40.

Sheppard, M., Werne, W., Tsatas, E., McCoy, R., Prowse, S. and Johnson, M.

(1998). Fowl adenovirus recombinant expressing VP2 of infectious bursal disease virus induces protective immunity against bursal disease. Arch Virol ., 143(5):915-30.

Sivanandan, V. and Maheswaran, S. K.(1980). Immune profile of infectious

bursal disease: I. Effect of infectious bursal disease virus on peripheral blood T and B lymphocytes of chickens. Avian Dis., 24:715-25.

Skeeles, J. K., Lukert, P.D. and DeBuysscher, E. V. (1979). Infectious bursal

disease viral infections. Complement and virus- neutralizing antibody response following infection of susceptible chickens. Avian Dis., 23:95-106.

Skeeles. J.K., and Lukert, P.D. (1980). Studies with an attenuated cell-culture

adapted infectious bursal disease virus: Replication sites and persistence of the virus in specific-pathogen-free chickens. Avian Dis., 24:43-47.

Snyder, D.B. (1990). Current field status of infectious bursal disease virus.

Avian Pathol., 19, 419-423.

Page 284: Thesis

254

Snyder, D.B., Lana, D.P., Cho B.R. and Marquardt, W.W. (1988). Group and strain-specific neutralization sites of infectious bursal disease virus defined with monoclonal antibodies. Avian Dis., 32:527-34.

Snyder, D.B., Vakharia, V.N. and Savage, P.K. (1992). Naturally occurring-

neutralizing monoclonal antibody escape variants define the epidemiology of infectious bursal disease viruses in the United States. Arch Virol., 127:89-101.

Snyder, D.B., Vakharia, V.N., Mengel-Wherea, S.A., Edwards, G.H., Savage,

P.K., Lutticken, D. and Goodwin, M.A. (1994). Active cross-protection induced by a recombinant baculovirus expressing chimeric infectious bursal disease virus structural proteins. Avian Dis., 38(4):701-7.

Soliman, I.M.A., El-Kabany, M.M.A., Kalad, M.A. and Abdel-Baky, M.H. (1996).

Immune response in horses vaccinated with a combined freeze dried vaccine and tetanus toxoid. Assiut Vet Med J., 35(70):209–221.

Spies, U. and Muller, H. (1990). Demonstration of enzyme activities required for

cap structure formation in infectious bursal disease virus, a member of the birnavirus group. J Gen Virol., 71 (4):977-81.

Spies, U., Muller, H. and Becht. H. (1987). Properties of RNA polymerase

activity associated with infectious bursal disease virus and characterization of its reaction products. Virus Res., 8:127-40.

Stills, H.F. and Bailey M.Q. (1991). The use of Freund‟s Complete Adjuvant,

20(4):25-31. Stone. H.D., Burgh, M., Hapkins, S.R., Yoder, H.W. and Beard, C.W. (1978).

Preparation of inactivated oil emulsion vaccines with avian viral mycoplasma antigens. Avian Dis., 22:666-674.

Stott, J.L., Osburn, B.I. and Barber, T.L. (1979). The current status of research

on an experimental inactivated bluetongue virus vaccine. Proc Annu Meet U S Anim Health Assoc., 83:55–62.

Survashe, B.D., Aitken, I.D. and Powell, J.R. (1979). The response of the

harderian gland of the fowl to antigen given by the ocular route. I. histological changes. Avian Pathol., 8:77-93.

Tanimura, N., Tsukamoto, K., Nakamura, K., Narita, M. and Maeda, M. (1995).

Association between pathogenicity of infectious bursal disease virus and viral antigen distribution detected by immunohistochemistry. Avian Dis., 39:9-20.

Page 285: Thesis

255

Tayade, C., Jaiswal, T.N., Mishra, S.C. and Koti, M. (2006). L-Arginine stimulates immune response in chickens immunized with intermediate plus strain of infectious bursal disease vaccine. Vaccine, 24 552–560.

Tham, K. M., and Moon, C.D. (1996). Apoptosis in cell cultures induced by

infectious bursal disease virus following in vitro infection. Avian Dis., 40:109-13.1996.

Thompson, G., Mohammed, H., Bauman, B. and Naqi, S. (1997). Systemic and

local antibody responses to infectious bronchitis virus in chickens inoculated with infectious bursal disease virus and control chickens. Avian Dis., 41(3):519-27.

Thompson, J.D., Gibson, T.J., Plewniak, F., Jeanmougin F. and Higgins, D.G.

(1997). The CLUSTAL_X windows interface: flexible strategies for multiple sequence alignment aided by quality analysis tools. Nucleic Acids Res., 25(24):4876-82.

Tsai, H.I., and Saif, Y.M. (1992). Effect of cell culture passage on the

pathogenicity and immunogenicity of infectious bursal disease virus. Avian Dis., 36:415-442.

Tsukamoto, K., Kojima, C., Komori, Y., Tanimura, N., Mase, M. and Yamaguchi,

S. (1999). Protection of chickens against very virulent infectious bursal disease virus (IBDV) and Marek's disease virus (MDV) with a recombinant MDV expressing IBDV VP2. Virol., 257(2):352-62.

Tsukamoto, K., Saito, S., Saeki, S., Sato, T., Tanimura, N., Isobe, T., Mase, M.,

Imada, T., Yuasa, N. and Yamaguchi, S. (2002). Complete, long-lasting protection against lethal infectious bursal disease virus challenge by a single vaccination with an avian herpesvirus vector expressing VP2 antigens. J Virol., 76(11):5637-45.

Tsukamoto, K., Sato, T., Saito, S., Tanimura, N., Hamazaki, N., Mase, M. and

Yamaguchi, S. (2000). Dual-viral vector approach induced strong and long-lasting protective immunity against very virulent infectious bursal disease virus. Virol., 269(2):257-67.

Ture, O., Tsai, H.J. and Saif, Y. M. (1993). Studies on antigenic relatedness of

classic and variant strains of infectious bursal disease viruses. Avian Dis., 37:647-54.

Vakharia, V.N., Snyder, D.B., He, J., Edwards, G.H., Savage, P.K. and Mengel-

Whereat, S.A. (1993). Infectious bursal disease virus structural proteins expressed in a baculovirus recombinant confer protection in chickens. J. Gen Virol ., 74 (Pt 6):1201-6.

Page 286: Thesis

256

Vakharia, V.N., Snyder, D.B., Lutticken, D., Mengel-Whereat, S.A., Savage, P.K., Edwards, G.H. and Goodwin, M.A. (1994). Active and passive protection against variant and classic infectious bursal disease virus strains induced by baculovirus-expressed structural proteins. Vaccine. 12:452-6.

van den Berg T.P., Gonze, M. and Meulemans, G. (1991). Acute infectious

bursal disease in poultry: isolation and characterization of a highly virulent strain. Avian Pathol., 20(1):133-43.

van den Berg, T.P. (2000). Acute infectious bursal disease in poultry: a review.

Avian Pathol., 29:175-194. van den Berg, T.P. and Meulemans, G. (1991). Acute infectious bursal disease

in poultry: protection afforded by maternal derived antibodies and interference with live vaccination. Avian Pathol., 20:409-421.

van den Berg, T.P., Gonze, M., Morales, D. and Mueulemans, G. (1996).

Acute infectious bursal disease in poultry:immunological and molecular basis of antigenicity of a highly virulent strain. Avian Pathol., l 25:751-76.

van Loon, A. A., de Haas, N., Zeyda, I. and Mundt, E. (2002). Alteration of

amino acids in VP2 of very virulent infectious bursal disease virus results in tissue culture adaptation and attenuation in chickens. J Gen Virol., 83:121-9.

Vindevogel, H., Gouffaux M., Meulemans G., Duchatel J.P. and Halen, P.

(1976). Maladie de Gumboro:distribution et persistence du virus chez la poussin inocule. Etudes sur la transmission de la maladie. Avian Pathol., 5:31-8.

Wang M. and Doong, S. (2000.) A pH-based fed-batch process for the

production of a chimeric recombinant infectious bursal disease virus (IBDV) structural protein (rVP2H) in insect cells. Process Biochem., 35(9):877-84.

Wang, M.Y., Kuo, Y.Y., Lee, M.S., Doong, S.R., Ho, J.Y. and Lee, L.H.(2000).

Self-assembly of the infectious bursal disease virus capsid protein, rVP2, expressed in insect cells and purification of immunogenic chimeric rVP2H particles by immobilized metal-ion affinity chromatography. Biotechnol Bioeng., 67(1):104-11.

Wang, X., Jiang, P., Dee, S., Wu, J., Liu, X. and Xu, J. (2003). Efficacy of DNA

vaccines against infectious bursal disease virus in chickens enhanced

Page 287: Thesis

257

by coadministration with CpG oligodeoxynucleotide. Avian Dis., 47(4):1305-12.

Wang, X.M., Zeng, X.W.H.L., Gao, C.Y. and Wei, P. ( 2004). Changes in VP2

gene during the attenuation of very virulent infectious bursal disease virus strain Gx isolated in China. Avian Dis., 48: 77–83.

Wang, Y., Qi, X., Gao, H., Gao, Y., Lin, H., Song, X., Pei, L. and Wang, X.

(2009). Comparative study of the replication of infectious bursal disease virus in DF-1 cell line and chicken embryo fibroblasts evaluated by a new real-time RT-PCR. J. Virol. Methods., 157: 205–210.

Westbury, H.A. (1978). Interaction between infectious bursal disease virus and

Newcastle disease virus in chickens. Aust Vet J., 54(7):349-51. Whitfill, C.E., Haddad, E.E., Ricks, C.A., Skeeles, J.K., Newberry, L.A., Beasley,

J.N., Andrews, P.D., Thoma, J.A. and Wakenell, P.S. (1995). Determination of optimum formulation of a novel infectious bursal disease virus (IBDV) vaccine constructed by mixing bursal disease antibody with IBDV. Avian Dis., 39(4):687-99.

Whitfill, C.E., Presson, B., Newberry, L., Andrews, P., Cox, E., Skeeles, K.,

Gyles, N.R. and Thoma, J.A. (1991). Action spectrum of antiviral factor from chicken sera. Poult Sci., 70(12):2450-9.

Winterfield, R.W., Fadly, A.M. and Bickford, A. (1972). Infectivity and distribution

of infectious bursal disease virus in the chicken. Persistence of the virus and lesions. Avian Dis., 16(3):622-32.

Winterfield, R.W., Hoerr, F.J. and Fadly, A.M. (1978). Vaccination against

infectious bronchitis and the immunosuppressive effects of infectious bursal disease. Poult Sci., 57(2):386-91.

Wood, G.W., Muskett J.C. and Thornton, D.H. (1981). Theinteraction of live

vaccine and maternal antibody inprotection against infectious bursal disease. Avian Pathol., 10: 365-373.

Wu, C.C. and Lin, T.L. (1992). Detection of infectious bursal disease virus in

digested formalin-fixed paraffin-embedded tissue sections by polymerase chain reaction. J Vet Diagn Invest., 4(4):452-5.

Wu, C.C., Dorairajan, T. and Lin, T.L. (2000). Effect of ascorbic acid

supplementation on the immune response of chickens vaccinated and challenged with infectious bursal disease virus. Vet Immunol Immunopathol., 74(1-2):145-52.

Page 288: Thesis

258

Wyeth P.J. and Cullen GA. (1979). The use of an inactivated infectious bursal disease oil emulsion vaccine in commercial broiler parent chickens. Vet Rec., 104(9):188-93.

Wyeth P.J. and Cullen, G.A. (1978). Transmission of immunity from inactivated

infectious bursal disease oil-emulsion vaccinated parent chickens to their chicks. Vet Rec., 102(16):362-3.

Wyeth P.J., Chettle. N.J and Mohepat, A.R. (1992). Use of an inactivated

infectious bursal disease oil emulsion vaccine in commercial layer chicks. Vet Rec., 130(2):30-2.

Wyeth, P.J. (1975). Effect of infectious bursal disease on the response of

chickens to S typhimurium and E coli infections. Vet Rec., 96(11):238-43.

Wyeth, P.J. (1980). Passively transferred immunity to IBD following live

vaccination of parent chickens by two different routes. Vet Rec., 106(13):289-90.

Wyeth, P.J. and Cullen, G.A. (1976). Maternally derived antibody-effect on

susceptibility of chicks to infectious bursal disease. Avian Pathol., 5:253-60.

Yamaguchi, T., Kondo, T., Inoshima, Y., Ogawa, M., Miyoshi, M., Yanai, T.,

Masagi, T., Fukushi, H. and Hirai, K. (1996a). In- vitro attenuation of highly virulent infectious bursal disease virus: Some characteristics of attenuated strains. Avian Dis., 40(3):501-509.

Yamaguchi, T., Ogawa, M., Inoshima, Y., Miyoshi, M., Fukushi, H. and Hirai, K.

(1996b). Identification of sequence changes responsible for the attenuation of highly virulent infectious bursal disease virus. Virology, 223:219-23

Yamaguchi, T., Ogawa, M., Miyoshi, M., Inoshima, Y., Fukushi, H., and Hirai, K.

(1997). Sequence and phylogenetic analyses of highly virulent infectious bursal disease virus. Arch Virol., 142:1441-58.

Yao, K., Goodwin, M. A., and. Vakharia V.N. (1998). Generation of a mutant

infectious bursal disease virus that does not cause bursal lesions. J Virol., 72:2647-54.

Yehuda, H., Godfried, Y., Goldway, M., Gutter, B., Michael, A., Shaaltiel, Y.,

Levi, B.Z. and Pitcovski, J. (2000). Transfer of antibodies elicited by baculovirus-derived VP2 of a very virulent bursal disease virus strain to progeny of commercial breeder chickens. Avian Pathol., 29(1):13-20.

Page 289: Thesis

259

Yip, C.W., Yeung, Y.S., Ma, C.M., Lam, P.Y., Hon, C.C., Zeng, F. and Leung, F.C. (2007). Demonstration of receptor binding properties of VP2 of very virulent strain infectious bursal disease virus on Vero cells. Virus Res., 123(1): 50-6.

Yuasa, N., Taniguch,i T., Noguchi, T. and Yoshida, I. (1980). Effect of infectious

bursal disease virus infection on incidence of anemia by chicken anemia agent. Avian Dis., 24:202-9.

Zhu, S.L., Wu, G.P., Zhang, and Zhu, G.Q. (2008). The cellular receptors for

infectious bursal disease virus. African J. of Bio., 7(25):4832–4835. Zierenberg, K., Nieper H., van den Berg, T.P., Ezeokoli C.D. Voss M. and

Muller, H. (2000). The VP2 variable region of African and German isolates of infectious bursal disease virus: comparison with very virulent, "classical" virulent, and attenuated tissue culture-adapted strains. Arch Virol., 145(1):113-25.

Page 290: Thesis

260

APPENDIX A

Buffer and Media

1. PBS (pH 7.4)

NACl 8.0 g KCl 0.2 g Na2HPO4 1.15 g KH2PO4 0.2 g The above chemicals were dissolved in 800 mL distilled water. The final volume of the buffer was then brought up to 1000 mL after adjusted the pH to 7.4. The buffer was autoclaved at 121 ºC for 15 minutes at 151 bs pressure. 3. Antibiotic-Antimycotic (100X) Liquid Penicillin G (sodium salt) 10,000 units/mL Streptomycin Sulfate 10,000 ug/mL Amphotericin B 25 ug/mL 0.85% Saline 100 mL 4. Growth Medium RPMI 1640 (Gibco) 10 g FBS 100 mL NaHCO3 0.58 g Distilled Water 1 litre Add 1 mL of antibiotic-antimycotic. Sterile by filtration using 0.22 µm pore size filter and store at 4 ºC. 5. Maintenance Medium RPMI 1640 (GIBCO) 10 g TPB 10 mL FBS 10 mL NaHCO3 3.7 g Distilled Water 1 litre

Page 291: Thesis

261

Add 1 mL of antibiotic-antimycotic. Sterile by filtration using 0.22 µm pore size filter and store at 4 ºC. Note:

For the growth and maintenance medium, 1 mL of each antibiotic-antimycotic must be added. Then warm it to 37ºC before use.

FBS must be inactivated at 56 ºC for 1 hour to inactivate the non-specific inhibitor before it was used in maintenance media in case of cell culture inoculation with particular virus.

6. Antibiotic-Trypsin-Versine (ATV)

Trypsin 0.5 g Versine (EDTA) 0.2 g

NaCl 8 g KCl 0.4 g Dextrose 1 g NaHCO3 0.58 g Penicillin 2 x 100 units Streptomycin 100 mg Phenol Red 0.02 g The above are made up to 1000 mL with distilled water and the resulting solution is sterilized by 0.22 µm pore size filter and store at 4 ºC.

7. TAE Buffer (10x stock)

40 mM Tris-base 48.4 g 20 mM Glacial acetic-acid 11.4 mL 0.5 M EDTA 20 mL In 1000 mL of distilled water.

Page 292: Thesis

262

APPENDIX B

Table B1: Fifty Percent Tissue Culture Infective Dose (TCID50/mL) DF-1p6

Dilution Infected Non-infected Cumulative effect

Ratio Percentage CPE No CPE

10-1 3 1 4 1 4/5 80

10-2 1 3 1 4 1/5 20

10-3 0 4 0 8 0/8 0

10-4 0 4 0 12 0/12 0

10-5 0 4 0 16 0/16 0

10-6 0 4 0 20 0/20 0

10-7 0 4 0 24 0/24 0

10-8 0 4 0 28 0/28 0

%50below%50aboveyInfectivit

%50%50aboveyInfectivitpointEnd

= 0.5

TCID50 = Dilution where CPE > 50% + 0.5 = 1 + 0.5 = 1.5 / 100 µl One unit 101.5 TCID50/0.1 mL

Page 293: Thesis

263

Table B2: Fifty Percent Tissue Culture Infective Dose (TCID50/mL) DF-1p9

Dilution Infected Non-infected Cumulative effect

Ratio Percentage CPE No CPE

10-1 4 0 8 1 8/8 100

10-2 3 1 4 1 4/5 80

10-3 1 3 1 4 1/5 20

10-4 0 4 0 8 0/8 0

10-5 0 4 0 12 0/12 0

10-6 0 4 0 16 0/16 0

10-7 0 4 0 20 0/20 0

10-8 0 4 0 24 0/24 0

%50below%50aboveyInfectivit

%50%50aboveyInfectivitpointEnd

= 0.5

TCID50 = Dilution where CPE > 50% + 0.5 = 2 + 0.5 = 2.5 / 100 µl One unit 102.5 TCID50/0.1 mL

Page 294: Thesis

264

Table B3: Fifty Percent Tissue Culture Infective Dose (TCID50/mL) Vero p6

Dilution Infected Non-infected Cumulative effect

Ratio Percentage CPE No CPE

10-1 4 0 12 0 12/12 100

10-2 4 0 8 0 8/8 100

10-3 3 1 4 1 4/5 80

10-4 1 3 1 4 1/5 20

10-5 0 4 0 8 0/8 0

10-6 0 4 0 12 0/12 0

10-7 0 4 0 16 0/16 0

10-8 0 4 0 20 0/20 0

%50below%50aboveyInfectivit

%50%50aboveyInfectivitpointEnd

= 0.5

TCID50 = Dilution where CPE > 50% + 0.5 = 3 + 0.5 = 3.5 / 100 µl One unit 103.5 TCID50/0.1 mL

Page 295: Thesis

265

Table B4: Fifty Percent Tissue Culture Infective Dose (TCID50/mL) Vero p9

Dilution Infected Non-infected Cumulative effect

Ratio Percentage CPE No CPE

10-1 4 0 16 0 16/16 100

10-2 4 0 12 0 12/12 100

10-3 4 0 8 0 8/8 100

10-4 3 1 4 1 4/5 80

10-5 1 3 1 4 1/5 20

10-6 0 4 0 8 0/8 0

10-7 0 4 0 12 0/12 0

10-8 0 4 0 16 0/16 0

%50below%50aboveyInfectivit

%50%50aboveyInfectivitpointEnd

= 0.5

TCID50 = Dilution where CPE > 50% + 0.5 = 4 + 0.5 = 4.5 / 100 µl One unit 104.5 TCID50/0.1 mL

Page 296: Thesis

266

Table B5: Fifty Percent Tissue Culture Infective Dose (TCID50/mL) Vero p10

Dilution Infected Non-infected Cumulative effect

Ratio Percentage CPE No CPE

10-1 4 0 17 0 17/17 100

10-2 4 0 13 0 13/13 100

10-3 4 0 9 0 9/9 100

10-4 3 1 5 1 5/6 83.3

10-5 2 2 2 3 2/5 40

10-6 0 4 0 7 0/7 0

10-7 0 4 0 11 0/11 0

10-8 0 4 0 15 0/15 0

%50below%50aboveyInfectivit

%50%50aboveyInfectivitpointEnd

= 0.77 TCID50 = Dilution where CPE > 50% + 0.77 = 4 + 0.77 = 4.77 / 100 µl One unit 104.77 TCID50/0.1 mL

Page 297: Thesis

267

Table B6: Fifty Percent Tissue Culture Infective Dose (TCID50/mL) Vero p15

Dilution Infected Non-infected Cumulative effect

Ratio Percentage CPE No CPE

10-1 4 0 25 0 25/25 100

10-2 4 0 21 0 21/21 100

10-3 4 0 17 0 17/17 100

10-4 4 0 13 0 13/13 100

10-5 4 0 9 0 9/9 100

10-6 3 1 5 1 5/6 83

10-7 2 2 2 3 2/5 40

10-8 0 4 0 7 0/7 0

%50below%50aboveyInfectivit

%50%50aboveyInfectivitpointEnd

= 0.76

TCID50 = Dilution where CPE > 50% + 0.76 = 6 + 0.76 = 6.76 / 100 µl One unit 106.7 TCID50/0.1 mL

Page 298: Thesis

268

Table B7: Fifty Percent Tissue Culture Infective Dose (TCID50/mL) Vero p20

Dilution Infected Non-infected Cumulative effect

Ratio Percentage CPE No CPE

10-1 4 0 28 0 28/28 100

10-2 4 0 24 0 24/24 100

10-3 4 0 20 0 20/20 100

10-4 4 0 16 0 16/16 100

10-5 4 0 12 0 12/12 100

10-6 4 0 8 0 8/8 100

10-7 2 2 4 2 4/6 66

10-8 2 2 2 4 2/6 33

%50below%50aboveyInfectivit

%50%50aboveyInfectivitpointEnd

= 0.45

TCID50 = Dilution where CPE > 50% + 0.45 = 7 + 0.45 = 7.45 / 100 µl One unit 107.4 TCID50/0.1 mL

Page 299: Thesis

269

APPENDIX C

Chemicals for Histopathology

1. 10% Buffered Formalin (Luna, 1968) 37-40% formalin 100 mL Distilled water 900 mL Sodium phosphate monobasic 4 mL Sodium phosphate dibasic 6.5 mL (anhydrous) 2. Haematoxylin and Eosin Staining (H & E) i. Harris Hematoxylin

Distilled water 200 mL Hematoxylin crystal 1 g Ammonium or potassium sulfate 20 g Mercuric oxide (red) 0.5 g The hematoxylin is dissolved in the alcohol, whilst the ammonium is dissolved in water with the aid of heat. Both the hematoxylin and the alum solution are combined and boiled. The addition of mercuric oxide make the solution takes a dark purple colour. Filter the solution before using or storing.

Page 300: Thesis

270

APPENDIX D

LESION SCORING Table G: Lesion scoring for bursa of Fabricius

Lesion Scoring Description

0 (Normal) Normal or undetectable lesion

1 (Mild) Mild degeneration and necrosis especially at the medullary region of lymphoid.

2 (Mild to Moderate) Mild to moderate degeneration and necrosis of lymphoid cells in some lymphoid follicles especially in the medulla. Interstitial connective tissue becomes oedematous and filled with inflammatory cells.

3 (Moderate) Moderate necrotized follicles involving both the cortex and medulla. Pyknotic nuclei were scattered in follicles. The interstitial space was obvious and present of heterophils, macrophages and a few erythrocytes and fibroblasts. Epithelial lining was thickened and vacuolated in some areas.

4 (Moderate to severe) Moderate to severe depletion of lymphoid cells. Lymphoid cells aggregation was found in the cortex of some follicles. Necrotic cells and cysts were present in some follicles especially in the medulla. The interstitial space was infiltrated and pack with fibrinous connective tissues. The intra and extra follicular areas might be hyperaemic and haemorrhagic. Epithellium was thickened, corrugated and vacuolated in some areas.

5 (Severe) Severe follicular necrosis and degeneration involving both cortex and medulla. Follicular cysts with fibrinous exudates and cell debris were frequently observed. The interstitial connective tissue was obvious, oedematous and infiltrated with mild to moderate inflammatory cells. The epithelial lining of the bursa was thickened and vacuolated

Or

Moderate to severe atrophy of the bursa follicles with cellular degeneration and necrosis. Cysts might be present in some follicles. The interstitial connective tissue as obvious and infiltrated with fibroblast and inflammatory cells

Page 301: Thesis

271

BIODATA OF AUTHOR

The author, Majed Hameed Mohammed, was born in 21st of April 1965, in Baghdad,

Iraq. He was the eldest son in his family of five. He is married and has got 2 children.

He did his primary education in Baghdad from 1971 to 1977. The author continued his

secondary school education at AL-Mansoor secondary school and he finished his final

public examination successfully on 1983 with an average grade 82%.

On October, 1983, the author furthered his study in Bachelor Science of Veterinary

Medicine in Faculty of Veterinary Medicine, Baghdad University. After finishing the

degree he graduated on 1st of July 1989 with final average of (77) and he ranked (3) in a

class numeration of 185 students.

Immediately after completion the bachelor degree, he worked as a researcher assistant

at Pathology and Poultry Department Laboratory, Faculty of Veterinary Medicine,

Baghdad University. He spent 6 months during his service as a research assistant and

later joined to the ranks of military army to perform his compulsory service in his

country. Then after one year, he enrolled as a full-time candidate pursuing his study in

Master Science program in field of poultry diseases at the same Faculty of Veterinary

Medicine on 1991.

On 1993, the author was awarded a Master‟s degree in poultry diseases with standard

very good for submission of thesis titled “The effect of Gumboro disease on the

incidence of airsacculitis”. After completion all the requirements for M.Sc. degree, the

Page 302: Thesis

272

author has got a job as a lecturer on 1993 in the same Department of Poultry Disease,

Veterinary Medicine.

The author engaged to work as a part time in the field of Veterinary supervision of

poultry farms and as a veterinarian at a center of agricultural research and animal

production.

On 2006, the author got an offer letter to persue his Ph.D. programme at Faculty of

Veterinary Medicine, Universiti Putra Malaysia in the field of Pathology. Upon

completing his Ph.D. the author plans to embark on an academic career and further

play an active role ensha‟allah through post doctorate research programme.

Page 303: Thesis

273

LIST OF PUBLICATIONS

PATENT Novel attenuated and inactivated tissue culture adapted infectious bursal disease vaccine against very verulent infectious bursal disease virus Infection, (submitted to innovation and commercialization center UPM for patent submission) JOURNALS 1. Mohammed, M.H., Hair-Bejo, M., Omar, A.R. and Aini, I. (2010). Adaptation, attenuation and molecular characterisation of very virulent infectious bursal disease virus of Malaysia isolate in Vero cells. Acta Vet. Hungarica (to be submitted). 2. Mohammed, M.H., Hair-Bejo, M., Omar, A.R. and Aini, I. (2010). Pathogenicity and Immunogenicity of very virulent infectious bursal disease virus adapted in Vero cells in Malaysia. Acta Vet. Scandinavica (to be submitted). IN PROCEEDING 1. Mohammed, M.H., Hair-Bejo, M., Omar, A.R. and Aini, I. (2008). Adaptation and propagation of very virulent infectious bursal disease virus of Malaysian isolate in mammalian cell line. In: Proceeding of 29th Malaysian Society of Animal Production (MSAP) Annual Conference, 25-27 May 2008, Bayview Beach Resort, Penang. P.36 2. Mohammed, M.H., Hair-Bejo, M., Omar, A.R. and Aini, I. (2008). Adaptation and propadation of very virulent infectious bursal disease virus of Malaysian isolate in DF-1 cell line. In Proceeding of 20th Veterinary Association Malaysia (VAM) congress, 15th -17thAugust 2008, Equatorial Hotel Bangi-Putrajaya. P. 51 3. Mohammed, M.H., Hair-Bejo, M., Omar, A.R. and Aini, I. (2009). Efficacy of inactivated very virulent infectios bursal disease virus isolate of Malaysia in specific pathogenic free chickens.In Proceeding of 21th Veterinary Association Malaysia (VAM) congress, 7-9th August 2009, the Legend, Water Chalets Port Dickson, Negeri Sembilan. P.20 GENBANK

1. Mohammed, M.H., Hair-Bejo, M., Omar, A.R. and Aini, I (2009). Accession number FJ824699 (IBD UPM0081T10 VP2 gene). In: GenBank – http://www.ncbi.nlm.nih.gov/ 2. Mohammed, M.H., Hair-Bejo, M., Omar, A.R. and Aini, I (2009). Accession number FJ898322 (IBD UPM0081T15 VP2 gene). In: GenBank – http://www.ncbi.nlm.nih.gov/

Page 304: Thesis

274

3. Mohammed, M.H., Hair-Bejo, M., Omar, A.R. and Aini, I (2009). Accession number FJ898321 (IBD UPM0081T20 VP2 gene). In: GenBank – http://www.ncbi.nlm.nih.gov/