You are on page 1of 267

A LABORATORY MANUAL FOR THE

ISOLATION,
IDENTIFICATION, AND
CHARACTERIZATION -

OF AVIAN
UH* ** w/ «

Fifth Edition

THE AMERICAN ASSOCIATION OF AVIAN PATHOLOGISTS


A LABORATORY MANUAL FOR THE
ISOLATION, IDENTIFICATION
AND CHARACTERIZATION
OF AVIAN PATHOGENS

Fifth Edition

American Association of Avian Pathologists

Editorial Committee

Louise Dufour-Zavala, Editor-in-Chief


David E. Swayne
John R. Glisson
Janies E. Pearson
Willie M. Reed
Mark W. Jackwood
Peter R. Woolcock

Copies Available from:


American Association of Avian Pathologists
953 College Station Road
Athens, GA 30602-4875
Frederic J. Hoerr

Cover:
1. Direct fluorescent antibody test on primary chicken embryo kidney cells showing
syncytia formed by ILT virus in primary chick embryo kidney cells. (Rey Resurreccion,
GPLN, Oakwood, GA) 2. Ninety well serum plate prepared for ELISA testing for the
detection of AE antibodies. (Len Chappell, GPLN, Oakwood, GA). 3. Bacterial culture on
a TSI slant. (Doug Waltman, GPLN, Oakwood, GA). 4. Reverse transcriptase-polymerase
chain reaction (RT-PCR) and restriction fragment length polymorphism (RFLP) analysis if
the spike (SI) glycoprotein gene of the Arkansas strain of infectious bronchitis virus
(IBV). Lanes 1 and 2 are molecular weight markers. Lane 3 is the IBV amplicon digested

9 with BstYI, lane 4 is the IBV amplicon digested with Haein, and lane 5 is the IBV
amplicon digested with XcmI. (Mark Jackwood, PDRC, Athens, GA). 5. Ten-fold
dilution series of avian influenza virus RNA run with the USDA type A influenza M gene
real-time RT-PCR test on the Applied Biosystems 7500 FAST system. (Erica Spackman,
1 SEPRL, Athens, GA). 6. Microphotograph of Mycoplasma gallisepticum colonies on agar
(35x) (Stan Kleven, PDRC, Athens, GA). 7. Bio Merieux API 20 system for bacteria
identification using a series of biochemical tests. (Doug Waltman, GPLN, Oakwood, GA).
3 4
8. Embryonated egg candling to locate the chorio-allantoic sac for inoculation of a virus
isolation sample. (Rey Resurreccion, GPLN, Oakwood, GA). 9. Wet mount of Aspergillus

2 sp. conidiofores after culture in Sabouraud's dextrose agar (100X). The isolate was
obtained from a case of systemic aspergillosis in 9-wk-old broiler breeder pullets.
(Guillermo Zavala, PDRC, Athens, GA).
Pictures 2,3,7,8 and Page design by HMM Photography (Heidi Migalla).

5 6
7 1-------- 1 8

Copyright © 1975, 1980, 1989,1998,2008 by American Association of Avian Pathologists, Inc. Athens, Georgia

All rights reserved

Copyright is not claimed for Chapters 22, 36

Chapters 16,29, 35,40,44, 49: US Government copyrighted, user rights reserved, printed by permission

Chapters 30, 31: (British) Crown copyrighted and (British) Crown user rights reserved, printed by permission

Chapter 6: (Australian) Crown copyrighted and (Australian) Crown user rights reserved, printed by permission

Mention of a trademark or proprietary product does not constitute a guarantee or warranty of the product by the author, the supporting
institutions or the American Association of Avian Pathologists and does not imply its approval to the exclusion of other products that also
may be suitable

Fifth edition 2008

Previously entitled Isolation and Identification ofAvian Pathogens

Library of Congress Catalog Card Number: 2008924452

ISBN 978-0-9789163-2-9

All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted, in any form or by any means,
electronic, mechanical, photocopying, recording, or otherwise, without the prior written permission of the copyright owner.

Printed in the United States of America

10 987654321

Printed by
OmniPress, Inc., Madison, Wisconsin 53704

ii
A Laboratory Manual for the Isolation, Identification and Characterization
of Avian Pathogens

TABLE OF CONTENTS

1. Diagnostic Principles. Frederic J. Hoerr...................................................................................................................................1


2. Salmonellosis. W. Douglas Waltman and Richard K. Gast.................................................................................................... 3
3. Colibacillosis. Margie D. Lee and Lisa K. Nolan..................................................................................................................10
4. Pasteurellosis, Avibacteriosis, Gallibacteriosis, Riemerellosis, and Pseudotuberculosis.
John R. Glisson, Tirath S. Sandhu, and Charles L. Hofacre....................................................................................................12
5. Bordetellosis. Mark W. Jackwood........................................................................................................................................ 20
6. Infectious Coryza. Pat J. Blackall........................................................................................................................................ 22
7. Campylobacter in Poultry. Jaap A. Wagenaar and Wilma F. Jacobs- Reitsma..................................................................... 27
8. Spirochetosis. Stephen R. Collett........................................................................................................................................... 31
9. Erysipelas. George L. Cooper and Arthur A. Bickford........................................................................................................ 36
10. Listerosis. George L. Cooper and Arthur A. Bickford........................................................................................................ 39
11. Staphylococcosis. Stephan G. Thayer and W. Douglas Waltman.........................................................................................42
12. Streptococcosis and Enterococcosis. Stephan G. Thayer and W. Douglas Waltman............................................................ 44
13. Clostridial Diseases. Stephan G. Thayer and David A. Miller............................................................................................. 47
14. Tuberculosis. Susan Sanchez and Richard M. Fulton.......................................................................................................... 53
15. Mycoplasmosis. Stanley H. Kleven......................................................................................................................................59
16. Chlamydiosis. Arthur A. Andersen and Daisy Vanrompay................................................................................................. 65
17. Omithobacteriosis. Richard P. Chin and Bruce R. Charlton................................................................................................ 75
18. Mycoses and Mycotoxicoses. R.D. Wyatt............................................................................................................................ 77
19. Adenovirus. Brian M. Adair and J. Brian McFerran.............................................................................................................84
20. Hemorrhagic Enteritis of Turkeys Marble Spleen Disease of Pheasants. F. William Pierson and Sctott D. Fitzgerald....... 90
21. Infectious Laryngotracheitis. Deoki N. Tripathy and Maricarmen Garcia........................................................................... 94
22. Marek’s Disease. Patricia S. Wakenell and Jagdev M. Sharma........................................................................................... 99
23. Duck Virus Enteritis. Peter R. Woolcock............................................................................................................................ 106
24. Herpesviruses of Free-Living and Pet Birds. Erhard F. Kaleta........................................................................................... 110
25. Pox. Deoki N. Tripathy and Willie M. Reed....................................................................................................................... 116
26. Budgerigar Fledgling Disease and Other Avian Polyomavirus Infections. Branson W. Ritchie and Phil D. Lukert........ 120
27. Psittacine Beak and Feather Disease. Branson W. Ritchie and Phil D. Lukert................................................................... 122
28. Chicken Anemia Virus. M. Stewart McNulty and Daniel Todd......................................................................................... 124
29. Avian Influenza. David E. Swayne, Dennis A. Senne and David L. Suarez....................................................................... 128
30. Newcastle Disease Virus and Other Avian Paramyxoviruses. Dennis J. Alexander and Dennis A. Senne.........................135
31. Avian Metapneumovirus. Richard E. Gough and Janice C. Pedersen................................................................................. 142
32. Infectious Bronchitis. Jack Gelb, Jr. and Mark W. Jackwood............................................................................................. 146
33. Turkey Coronavirus. Mark W. Jackwood and James S. Guy.............................................................................................. 150
34. Enteric Viruses. Don Reynolds and Ching Ching Wu.................................................................................................... 153
35. Oncornaviruses, Leukosis/Sarcomas and Reticuloendotheliosis. Aly M. Fadly, Richard L. Witter, and Henry D. Hunt.. 164
36. Avian Encephalomyelitis. Louis van der Heide................................................................................................................... 173
37. Duck Hepatitis. Peter R. Woolcock............................................................................................................................ 175
38. Turkey Viral Hepatitis. Willie M. Reed.............................................................................................................................. 179
39. Viral Arthritis/Tenosynovitis and Other Reovirus Infections. John K. Rosenberger and Erica Spackman.........................181
40. Arbovirus Infection. Eileen N. Ostlund and James E. Pearson........................................................................................... 184
41. Infectious Bursal Disease. John K. Rosenberger, Y.M. Saif, and Daral J. Jackwood......................................................... 188
42. Parvovirus of Waterfowl. Richard E. Gough...................................................................................................................... 191
43. Cell-Culture Methods. Karel A. Schat and Holly S. Sellers................................................................................................ 195
44. Virus Propagation in Embryonating Eggs. Dennis A. Senne..............................................................................................204
45. Virus Identification and Classification. Pedro Villegas and Ivan Alvarado........................................................................209
46. Titration of Biological Suspensions. Pedro Villegas........................................................................................................... 217
47. Serologic Procedures. Stephan G. Thayer and Charles W. Beard.......................................................................................222
48. Molecular Identification Procedures. Daral J. Jackwood and Mark W. Jackwood..............................................................230
49. Antigen Detection Systems. Mary J. Pantin-Jackwood and Sandra S. Rosenberger.......................................................... 233
Appendix of Abbreviations and Acronyms used in the Text...................................................................................................... 241
Quick Reference Diagnostic Chart.............................................................................................................................................. 244
Index............................................................................................................................................................................................ 247

iii
CONTRIBUTING AUTHORS

Brian Adair Richard P. Chin


Veterinary Sciences Division California Veterinary Diagnostic Laboratory System
Agri-Food and Biosciences Institute Fresno Branch
Stoney Road School of Veterinary Medicine
Stormont University of California - Davis
Belfast BT4 3SD 2789 S. Orange Ave.
Northern Ireland Fresno, California 93725
E-mail: Brian.Adair@afbini.gov.uk E-mail: rpchin@ucdavis.edu
FAX: +44(0)2890525773 FAX: (559) 485-8097

Dennis J. Alexander Stephen R. Collett


Central Veterinaiy Laboratory (Weybridge) Department of Population Health
New Haw, Addlestone College of Veterinary Medicine
Surrey KT15 3NB The University of Georgia
United Kingdom 953 College Stations Road
E-mail: d.j.alexander@vla.defra.gsi.gov.uk Athens, Georgia 30602-4875
FAX: +44-1932-357856 E-mail: colletts@uga.edu
FAX: (706) 542-5630
Ivan R. Alvarado
120 Spring Lake Pointe George L. Cooper
Athens, GA 30605 School of Veterinary Medicine
E-mail: ialvarado@lahintemational.com University of California-Davis
FAX: (404) 506-9013 Calif. Vet. Diagnostic Lab. System
Turlock Branch
Dr. Arthur A. Andersen P.O. Box P
USDA-ARS Turlock, California 95381
Former Address: E-mail: gcooper@cvdls.ucdavis.edu .
National Animal Disease Center FAX: (209) 667-4261
P.O.Box 70, Ames, IA 50010
Email: aanderse@nadc.usda.gov Aly M. Fadly
USDA- Agricultural Research Service
Charles W. Beard Avian Disease and Oncology Laboratory (ADOL)
130 Red Fox Run 3606 East Mount Hope Road
Athens, GA 30605 East Lansing, Michigan 48823
E-mail: charlesb9@charter.net E-mail: fadly@msu.edu
FAX: (706) 548-6410 FAX: (517) 337-6776

Arthur A. Bickford Scott D. Fitzgerald


University of California-Davis Diagnostic Center for Population and Animal Health
California Veterinaiy Diagnostic Laboratory System College of Veterinary Medicine
Turlock Branch Michigan State University
1650 Simon Drive PO Box 30076
Turlock, California 95382 Lansing, Michigan 48909-7576
E-mail: aabickford@ucdavis.edu E-mail: fitzgerald@dcpah.msu.edu
FAX: (209) 632-4258 FAX: (517)355-2152

Pat J. Blackall Richard M. Fulton


Animal Research Institute Diagnostic Center for Population and Animal Health
Locked Mail Bag No. 4 Michigan State University
Moorooka, Queensland 4105 PO Box 30076
Australia Lansing, Michigan 48909
E-mail: blackap@dpi-qld.gov.au E-mail: fulton@dcpah.msu.edu
FAX: +61-7-33629-429 FAX: (517)355-2152

Bruce R. Charlton Maricarmen Garcia


California Veterinary Diagnostic Laboratory System Department of Population Health
Turlock Branch College of Veterinary Medicine
School of Veterinary Medicine The University of Georgia
University of California - Davis 953 College Station Rd.
3327 Chicharra Way Athens, Georgia 30602-4875
Coulterville, California 95311 E-mail: mcgarcia@uga.edu
E-mail: brcharlton@ucdavis.edu FAX: (706) 542-5630
FAX: (209) 667-4267

iv
Richard K. Gast Daral J. Jackwood
USDA, ARS Food Animal Health Research Program
Russell Research Center Ohio Agricultural Research and Development Center
950 College Station Road The Ohio State University
Athens, GA 30605 1680 Madison Ave
E-mail: rgast@seprl.usda.gov Wooster, Ohio 44691
FAX: (706) 546-3035 E-mail: jackwood.2@osu.edu
FAX: (330) 263-3760
Jack Gelb, Jr.
Department of Animal and Food Sciences Mark W. Jackwood
College of Agricultural Sciences Department of Population Health
531 South College Avenue College of Veterinary Medicine
University of Delaware The University of Georgia
Newark, Delaware 19716-2150 953 College Station Road
E-mail: jgelb@udel.edu Athens, Georgia 30602-4875
FAX: (302)831-2822 E-mail: mjackwoo@uga.edu
FAX: (706) 542-5630
John R. Glisson
Department of Population Health Wilma F. Jacobs-Reitsma
College of Veterinary Medicine RIKILT Institute of Food Safety
The University of Georgia Bomsesteeg 45
953 College Station Rd. 6708 PD Wageningen
Athens, Georgia 30602-4875 The Netherlands
E-mail: iglisson@uga.edu E-mail: Wilma.jacobs@wur.nl
FAX: (706) 542-5630 FAX: +31-317-417717

Richard E. Gough Erhard F. Kaleta


Central Veterinary Laboratory (Weybridge) Institute for Avian and Reptile Medicine
New Haw, Addlestone Justus-Liebig-University
Surrey KT15 3NB Frankfurter Strabe 91, D-35392 Giefien
United Kingdom Germany
E-mail: r.e.gough@vla.defra.gsi.gov.uk E-mail: erhard.f.kaleta@vetmed.uni-giessen.de
FAX: +44-1932-357856 FAX: +49-641-201548

Janies S. Guy Stanley H. Kleven


North Carolina State University Department of Population Health
College of Veterinary Medicine College of Veterinary Medicine
Department of Population Health & Pathobiology University of Georgia
4700 Hillsborough Street 953 College Station Rd
Raleigh, North Carolina 27606 Athens, Georgia 30602-4875
E-mail: Jim Guy@NCSU.edu E-mail: skleven@uga.edu
FAX: (919)513-6464 FAX: (706) 542-5630

Frederic J. Hoerr Margie D. Lee


Thomas Bishop Sparks Department of Population Health
State Diagnostic Laboratory Poultry Diagnostic and Research Center
P. O. Box 2209 The University of Georgia
Auburn, Alabama 36831-2209 953 College Station Road
E-mail: hoerrfj@vetmed.auburn.edu Athens, Georgia 30602-4875
FAX: (334) 8244-7206 E-mail: mdlee@uga.edu
FAX: (706) 542-5771
Charles Hofacre
Department of Population Health Phil D. Lukert
College of Veterinary Medicine Department of Medical Microbiology
The University of Georgia College of Veterinary Medicine
953 College Station Road University of Georgia
Athens, Georgia 30602-4875 Box 207
E-mail: chofacre@uga.edu Colbert, Georgia 30628
FAX: (706) 542-5630 E-mail: plukert@uga.edu
FAX: (706) 542-5771
Henry Hunt
USDA- Agricultural Research Service J. Brian McFerran
Avian Disease and Oncology Laboratory (ADOL) 19 Knocktem Gardens
3606 East Mount Hope Road Belfast BT4 3LZ
East Lansing, Michigan 48823 Northern Ireland
E-mail: hunt@msu.edu FAX: +44-1232-658040
FAX: (517) 337-6776

v
M. Stewart McNulty Willie M. Reed
Department of Agriculture Purdue University
Veterinary Sciences Division Dean, School of Veterinary Medicine
Stormont, Belfast BT4 3SD Lynn Hall Room 1176
Northern Ireland West Lafayette, Indiana 47907-2026
FAX: +44-1232-525773 E-mail: wreed@purdue.edu
E-mail: m-mcnulty @utvintemet. com FAX: (765) 496-1261

David A. Miller Donald L. Reynolds


National Veterinary Services Laboratories 2520 Veterinary Administration
USDA-APHIS-VS Iowa State University
1800 Dayton Road 1802 Elwood Drive
Ames, Iowa 50010 Ames, Iowa 50011
E-mail: damiller@aphis.usda.gov E-mail: DLR@iastate.edu
FAX: (515) 239-8569 FAX: (515) 294-8956

Lisa K, Nolan Branson W. Ritchie


Dept, of Vet Microbiology and Preventive Medicine Department of Medical Microbiology
College of Veterinary Medicine College of Veterinary Medicine
1802 Elwood Dr VMRI #2 University of Georgia
Iowa State University Athens, Georgia 30602
Ames, LA 50011 FAX: (706) 542-6460
E-mail: lknolan@iastate.edu Email: britchie@uga.edu
FAX: (515) 233-2136
John K. Rosenberger
Eileen N. Ostlund Aviserve, LLC
Head Equine Ovine Viruses Section Delaware Technology Park
Diagnostic Virology 1 Innovation Way, Suite 100
NVSL Newark, DE 19711
Ρ,Ο,Βοχ 844 E-mail: aviserve@aol.com
Ames, IA 50010 FAX: (302) 368-2975
E-mail: Eileen.Ostlund@aphis.usda.gov
FAX: (515) 663-7348 Sandra S. Rosenberger
Aviserve, LLC
Mary J. Pantin-Jackwood Delaware Technology Park
Southeast Poultry Research Laboratory USDA-ARS 1 Innovation Way, Suite 100
934 College Station Road Newark, DE 19711
Athens, Georgia 30605 E-mail: aviserve@aol.com
E-mail: mary.pantin-jackwood@ars.usda.gov FAX: (302) 368-2975
FAX: (706) 546-3161
Y. M. Saif
Janies E. Pearson Food Animal Health Research Program
4016 Phoenix Ohio Agricultural Research and Development Center
Ames, Iowa 50014 The Ohio State University
(515) 292-9435 1680 Madison Avenue
E-mail: ipearson34@aol.com Wooster, Ohio 44691
E-mail: saif.l@osu.edu
Janice C. Pedersen FAX: (330) 263-3677
Avian Section, Diagnostic Veterinary Services Laboratory
NVSL Susan Sanchez
1437 270th Street Athens Diagnostic Laboratory
Madrid, Iowa 50156 0103 Athens V.M. Diag Lab
E-mail: Janice.c.pedersen@aphis.usda.gov 501 D.E. Brooks Dr
FAX: (515) 663-7348 Athens, GA 30602
E-mail: ssanchez@uga.edu
Frank W. Pierson FAX: (706)542-5568
Center for Molecular Medicine and Infectious Diseases
Virginia-Maryland Reg. College of Vet. Medicine Tirath S. Sandhu
Virginia Polytechnic Institute and State University Cornell University Duck Research Laboratory
Blacksburg, Virginia 24061 37 Howell Place
E-mail: pierson@vt.edu P.O. Box 427
FAX: (540) 231-3426 Speonk, New York 11972
E-mail: Sandhu37@optonline.net

vi
Karel A. Schat Stephen G. Thayer
Unit of Avian Health Department of Population Health
College of Veterinary Medicine
College of Veterinary Medicine The University of Georgia
Cornell University 953 College Station Road
Ithaca, New York 14853 Athens, Georgia 30602-4875
E-mail: kas24@comell.edu E-mail: sthayer@uga.edu
FAX: (607) 253-3384 FAX: (706) 542-0252

Holly Sellers Daniel Todd


Department of Population Health Dept of Agriculture and Rural Development from N. Ireland
Poultry Diagnostic and Research Center Veterinary Sciences Division
The University of Georgia Stormont, Belfast BT4 3SD
953 College Station Road UK
Athens, GA 30602-4875 E-mail: Daniel.Todd@dardni.gov.uk
E-mail: hsellers@uga.edu Tel: +44 2890 525773
FAX: (706)542-5630
Deoki N. Tripathy
Dennis A. Senne Department of Veterinary Pathobiology
National Veterinary Services Laboratories College of Veterinary Medicine
Veterinary Services University of Illinois
Animal and Plant Health Inspection Service 104 West McHenry
United States Department of Agriculture Urbana, Illinois 61801
1800 Dayton Road E-mail: tripathy@uiuc.edu
Ames, Iowa 50010 FAX: (217) 244-7421
E-mail: dennis.a.senne@aphis.usda.gov
FAX: (515) 663-7348 Daisy Vanrompay
Ghent University
Jagdev M. Sharma Faculty of Bioscience engineering
Department of Veterinary PathoBiology Department of molecular biotechnology
258 Veterinary Science Building Coupure Links 653
College of Veterinary Medicine 9000 Ghent, Belgium
University of Minnesota E-mail: daisy.vanrompay@UGent.be
1971 Commonwealth Avenue FAX: +32 09 2646219
St. Paul, Minnesota 55108
E-mail: sharmOO 1 @umn.edu Louis Van Der Heide
FAX: (612) 625-5203 Dept. Of Pathobiology U-89
PO Box 37
Erica Spackman 12 Yeomans Road
USDA-Agriculture Research Service Columbia, Connecticut 06237
Southeast Poultry Research Laboratory E-mail: louisandingrid@yahoo.com
934 College Station Road FAX: (860) 486-2794
Athens, GA 30605
E-mail: erica.spackman@ars.usda.gov Pedro Villegas
FAX: (706)546-3161 Department of Population Health
College of Veterinary Medicine
David L. Suarez University of Georgia
USDA-Agriculture Research Service 953 College Station Road
Southeast Poultry Research Laboratory Athens, Georgia 30602-4875
934 College Station Road E-mail: pedrov@uga.edu
Athens, GA 30605 FAX: (706) 542-5630
E-mail: david.suarez@ars.usda.gov
FAX: (706)546-3161 Jaap A. Wagenaar
Department of Infectious Diseases and Immunology
David E. Swayne OIE Reference Laboratory for Campylobacteriosis
USDA-Agricultural Research Service and WHO Collaboration Centre for Campylobacter
Southeast Poultry Research Laboratory Faculty of Veterinary Medicine
934 College Station Road Utrecht University
Athens, Georgia 30605 P.O. Box 80.165
E-mail: david.swayne@ars.usda.gov 3508 TD Utrecht
FAX: (706) 546-3161 The Netherlands
E-mail: i .wagenaar@uu.nl
FAX: +31 (0)30-2533199

vii
Patricia S. Wakenell Peter R. Woolcock
Department of Veterinary Medicine California Veterinary Diagnostic Laboratory System
Population Health & Reproduction Fresno Branch
University of California School of Veterinary Medicine
Davis, California 95616 University of California - Davis
E-mail: pswakenell@ucdavis. edu 2789 South Orange Ave
FAX: (530) 752-5845 Fresno, California 93725
E-mail: prwoolcock@ucdavis.edu
W. Douglas Waltman FAX: (559) 485-8097
Georgia Poultry Laboratory
P.O. Box 20 Ching-Ching Wu
4457 Oakwood Road Purdue University
Oakwood, Georgia 30566 Animal Disease Diagnostic Laboratory
E-mail: dwaltman@gapoultrylab.org 406 South University Street
FAX: (770) 535-1948 West Lafayette, Indiana 47907-2065
E-mail: Wuc@purdue.edu
Richard L. Witter FAX: (765) 497-1405
USDA- Agricultural Research Service
Avian Disease and Oncology Laboratory Roger Wyatt
3606 East Mount Hope Road 195 Edgewood Dr. SW
East Lansing, Michigan 48823 Athens, GA 30606
E-mail: witterr@msu.edu (706) 548-2297
FAX: (517) 349-0817 E-mail: mycotec@charter.net

viii
PREFACE

This manual has its origins in the need for a book to codify standardized method for testing and evaluating poultry vaccines. The
National Academy of Sciences sponsored the original publication entitled Methods for the Examination of Poultry Biologies, and completed
two successive revisions. In 1975, the American Association of Avian Pathologists (AAAP) accepted responsibility for the publication, but
because the need for standardizing testing of vaccines had been met, the scope and purpose of the manual was broadened to be a resource for
laboratory procedures for the isolation and identification of disease-causing agents. The title was changed to Isolation and Identification of
Avian Pathogens for the 1st edition and to A Laboratory Manual for the Isolation and Identification of Avian Pathogens for the 3rd edition.
The manual was intended as a bench resource for daily use in the diagnostic laboratory.

With the 4th edition, the focus had shifted from a manual of procedures for isolation and identification of pathogens to a more
encompassing manual for diagnosis of the disease and isolation and/or demonstration of the pathogen. This change was needed because
many clinical specimens are presented as unknowns for determination of etiology and some pathogens are difficult to isolate, but can be
demonstrated with newer molecular or immunologic techniques.

With the 5th edition, the AAAP appointed Dr. Louise Dufour-Zavala as Editor-in-Chief. David Swayne served as advisor to the Editor-
in-Chief and section editor. John Glisson, Willie M. Reed, Mark W. Jackwood and James E. Pearson continued as section editors for their
expertise in bacteriology, veterinary diagnostics, molecular biology, and virology. Peter Woolcock was newly appointed to support the
virology section.

The 5th edition has a new title to reflect the molecular advances and modem testing procedures allowing us to characterize, type,
speciate and serotype several avian pathogens. It also has a new chapter on Turkey Coronavirus. A color plate with tissue culture images is
included. Improvements to individual chapters include updated terminology and information on molecular techniques.

In the appendix section, we removed the appendix of sources and the appendix of reference antisera as they are now readily available on
the Internet.

The editorial committee thanks all of the 67 contributors who prepared new chapters or revised existing chapters for the 5th edition. We
also thank Crissie Boyd, Georgia Poultry Laboratory Network, for her tremendous clerical support in formatting the Manual. We thank Heidi
Migalla, HMM photography, for the design of the cover and Aaron Nord at Omni Press for the printing services.

Editorial Committee:

Louise Dufour-Zavala, Editor-in-Chief


David E. Swayne
John R. Glisson
Mark W. Jackwood
James £. Pearson
Willie M. Reed
Peter Woolcock

ix
X
1
Diagnostic Principles
Frederic J. Hoerr

In avian diagnostic medicine the usual immediate challenge is pneumovirus, infectious bronchitis, infectious laryngotracheitis,
making a definitive diagnosis for the presenting problem of infectious bursal disease, chicken infectious anemia, reovirus,
morbidity or mortality. When the case is probed deeper, however, hemorrhagic enteritis virus, mycoplasmosis, infectious coryza,
evidence may emerge of a concurrent or preceding infectious pasteurellosis, and others. For example, isolation of infectious
disease, management or nutritional problem, or other condition that bronchitis virus is an important first step in understanding a
contributes to the presenting problem. In food supply medicine respiratory disease. If the virus is determined to be a new serotype,
involving a population of animals, the challenge is to evaluate one a significant change in vaccine virus serotype will be required to
or more diagnoses for priority of response. In practice, the control the disease. If the isolated virus is indistinguishable from a
diagnostic results are considered along with animal welfare, vaccine strain administered at an earlier age, it may indicate the
production economics, and public health in the development of need for improvements in vaccine administration, or point to
strategies for treatment, prevention, and control of the disease. immunosuppression from infectious bursal disease, chicken
Three factors influence the expression of an infectious disease: the infectious anemia, or other diseases.
virulence of the causative organism, the level of exposure or dose of Some vaccine viruses are readily isolated for several days post­
the inoculum, and the susceptibility of the host. Within a poultry administration. Extended periods of virus shedding in a flock may
flock, each individual reacts according the net influence of these reflect variable immunity existing at the time of vaccine
factors. The uniformity of exposure and resistance among the administration. For respiratory viruses, isolation of a vaccine virus
individuals in the flock will eventually influence flock performance for a prolonged period after administration can be indicative of so-
and possibly food safety. called rolling reactions. In this situation, poor uniformity in flock
immunity will result in some chicks being resistant to virus
HISTORICAL PERSPECTIVE infection at the time of vaccine administration, then becoming
susceptible during the end of the shedding period of a pen mate.
The very existence of this book makes a statement about the The attenuated virus infection spreads from bird-to-bird resulting in
importance of infectious diseases that affect avian species. vaccination reactions of prolonged duration. Attenuated vaccine
Detection and characterization of infectious pathogens have viruses may re-acquire virulence characteristics during this process.
advanced substantially in recent decades, building on classical Rolling reactions and lengthened periods of vaccine virus isolation
methods for cultivation, isolation, characterization, and also occur with uneven or ineffective vaccine administration
immunological assay. Procedures based on the specific molecular technique. Vaccine virus shed from infected pen mates eventually
genetic characteristics of disease agents are now extensively applied infects susceptible chicks causing uneven and sometimes harsh
for the detection and characterization of infectious pathogens. reactions within the flock. Differentiating vaccine strains from wild­
Diagnostic technology today uses a variety of sensitive and specific type or field-challenge viruses or bacteria in the laboratory may
methods that may not require the actual cultivation and isolation of require molecular genetic sequencing and analysis.
an organism for a diagnosis. Isolation of an agent still has an The presence of co-pathogens can increase the severity of a viral
important place in the investigative process. Isolates are important disease such as infectious bronchitis or the respiratory form of
in assessing virulence and pathogenesis, and in the development of Newcastle disease. Mycoplasma infections and elevated
vaccines. Detection technologies offer significant gains and concentrations of ammonia and airborne dust increase the severity
substantial advantages in the number of samples that can be tested of respiratory viral disease. In these situations, the actual cause of
in a short time, and in the overall efficiency of testing. Enzyme- death or economic loss in the form of condemnations at slaughter
linked immunological detection (antigen capture ELISA) is used for from respiratory compromise or septicemia caused by Escherichia
viral and bacterial diseases. The polymerase chain reaction and coli. While the laboratory effort may focus on the viral infection,
nucleotide probes offer many approaches for identification of the cumulative effects of the co-pathogens must also take into
fragments of genetic code specific to a species or biotype of consideration for effective treatment and control. This situation
organism. A panel or multiplex of serological assays can reveal the requires a comprehensive approach to diagnostics, including
spectrum of infectious agents that have infected an individual bird. virology, bacteriology, serology, and pathology.
Simultaneous statistical analysis of ELISA titers for various Specific-pathogen-free sentinel birds are useful in identifying
infectious pathogens helps to assess the frequency and distribution specific primary infectious pathogens. These are typically used
of the infection or vaccine-induced humoral immunity within the when interference from overwhelming co-pathogens or vaccine
flock. Advances in diagnostic expertise that improve sensitivity and strains obscures the isolation effort of primary pathogen. Sentinel
specificity, and reduce the time required for the test generally lead birds can be placed in a flock for a specific time and then brought
to improvements in disease prevention and control. This is essential into the laboratory for pathogen isolation and identification
to safeguarding poultry health in the ever larger size of poultry procedures. Selective immunization of the sentinels focuses the
flocks under the care of food supply medicine. susceptibility and thereby increases the efficiency of identifying the
challenge strain.
SIGNIFICANCE OF IDENTIFIED PATHOGENS The relative significance of an isolate also depends on whether it
has food safety or public health significance. For example,
An isolated or detected disease agent should be evaluated for Salmonella, Escherichia, and Campylobacter can be pathogenic for
diagnostic significance relative to the case history, clinical disease, poultry but even in the apparent absence of disease they can be
and lesions. Poultry in commercial flocks may experience significant contaminants on processed poultry or poultry products.
sequential or simultaneous disease challenges, as well as exposure Avian influenza virus has strain-variable virulence among different
to agents in attenuated live vaccines. In a diagnostic investigation, poultry species, and some strains have considerable public health
both field challenge agents and attenuated live agents may be significance.
detected and require further characterization and differentiation.
Examples of live vaccines include those for Newcastle disease
1
Frederic J. Hoerr

DISEASE REPORTING microorganisms. Maintenance and calibration of equipment should


include but not be limited to scales, precision pipettes, incubators,
The isolation and identification of certain pathogens carries the refrigerators, freezers, autoclaves, spectrophotometers and other
responsibility of reporting to state/provincial or federal regulatory visualization apparatuses, and electrophoresis equipment.
officials, who in turn have responsibilities to report internationally Recording, documentation and review of laboratory procedures
to the World Organization for Animal Health (OIE) (1). The OIE should be routine. Commercial test kits should be used according to
website maintains a current list of internationally reportable manufacturer’s recommendations. Inventories of reagents and test
diseases which includes Newcastle disease, turkey rhinotracheitis, kits should be managed with respect to expiration dates.
any H5 or H7 avian influenza, Pullorum disease, and others.
Reporting regulations vary among countries, states, and provinces, LABORATORY SAFETY
as do the definitions of reportable strains of an agent. Laboratory
diagnosticians generally report to the next level of authority in their The avian diagnostic laboratory today should operate under
organization. Question about the responsibility to report a disease standard biosafety criteria (4). Although some avian procedures can
should be directed to the state or regional veterinary regulatory be conducted at biosafety level 1, biosafety level 2 is desirable
health official. Trade, regulatory and other legal issues may because some avian pathogens are agents of moderate potential
surround the isolation and accurate identification of certain hazard to personnel. Biosafety level 3 may be required for viruses
pathogens, or serological evidence thereof. Erroneous reporting, and bacteria that may cause serious or potentially lethal disease as a
misidentification or failure to identify an agent may carry serious result of exposure by the inhalation route. Salmonella,
consequences. In the United States, the USDA National Veterinary Campylobacter, Chlamydia, Erysipelas, Escherichia and Newcastle
Services Laboratory is the agency that should be consulted about disease can infect persons causing clinical disease from mild
agent identification, and in other countries, the appropriate central conjunctivitis to systemic illness. Certain strains of highly
reference laboratory. pathogenic influenza virus may cause human fatalities. Fungal
cultures carelessly handled can result in massive release of spores
QUALITY MANAGEMENT into the laboratory environment. Laboratory workers should be
informed about these risks and trained in proper procedures for
A quality management program to include standard operating routine and specialized aspects of laboratory duties. Laboratories
procedures for the isolation and identification of infectious should be kept clean and orderly, and surfaces frequently cleaned
pathogens improves the overall consistency and quality of the and disinfected. Warning notices should be displayed by
laboratory effort. A quality management program evaluates laboratories which contain hazardous substances or conditions.
laboratory procedures for the yield of relevant and timely data. It Biosafety cabinets of adequate rating should be maintained and
involves quality assessment by specifying performance parameters fully functional. Eating, drinking, and smoking should be prohibited
and setting limits for acceptable performance, and quality in laboratory work areas. Personal protective equipment including
improvement by correcting problems and preventing their appropriate laboratory clothing is always in order, and respiratory
reoccurrence. Quality management helps to ensure that the masks and protective eye glasses should be worn when zoonotic
laboratory information is accurate, reliable and reproducible, with pathogens are suspected.
the goal of eliminating or reducing test variation within and
between laboratories. The elements of a quality program comprise STORAGE AND ARCHIVING OF ISOLATES
written procedure manuals, record keeping, documentation, and
retention, training, education and evaluation of personnel, A diagnostic laboratory has a wealth of information and valuable
proficiency testing, laboratoiy safety, and maintenance and material pass through it. The agents isolated from commercial
monitoring of equipment. poultry represent a selection process involving large homogeneous
The International Standards Organization (ISO) is a developer and animal populations under performance rearing conditions. These
publisher of international standards for laboratoiy quality isolates have value for investigations of virulence and pathogenesis;
management (2). ISO is a non-governmental organizational network epidemiology of emerging agents; evaluation of new drugs and
composed of the national standards institutes that currently vaccines, and potentially as new vaccines. Isolates from
represent 157 countries. ISO document 17025 is the main standard noncommercial poultry have value in comparative studies and in
for diagnostic laboratories worldwide, including the USDA epidemiology. All viruses and most bacteria require ultracold (-70
National Veterinary Services Laboratory. These standards are F or lower) storage that can be achieved with specialty freezers or
reflected in the essential requirements for accreditation by the liquid nitrogen storage. Some bacteria and fungi can be stored in
American Association of Veterinary Laboratoiy Diagnosticians special media at room temperature. Specific information about
(AAVLD), which accredits publically-supported diagnostic storage and archiving of samples is provided in the text.
laboratories in the United States and Canada (3). The AAVLD
defines laboratoiy quality management to involve validation of test References
methods, among other criteria. Validation of a test requires ongoing
documentation of internal or inter-laboratory performance using 1. OIE: World Organization for Animal Health,
known reference standard(s) for the species and/or diagnostic http://www.oie.int/eng/maladies/en classification2007.htm?eld7.
specimen(s) of interest, and one or more of the following: the test is 2. International Organization for Standardization,
endorsed or published by reputable technical organization http://www.iso.org/iso/home.htm.
(including the American Association of Avian Pathologists 3. American Association of Veterinary Laboratory Diagnosticians.
Isolation and Identification of Avian Pathogens); published in a AAVLD Essential Requirements for An Accredited Veterinary
peer-reviewed journal with sufficient documentation to establish Medical Diagnostic Laboratory.
diagnostic performance and interpretation of results; or http://web.memberclicks.com/mc/page.do?sitePageId=27915&orgl
documentation of an internal or inter-laboratory comparison to an d=aavld.
accepted methodology or protocol. 4. Centers for Disease Control and Prevention. BMBL Section ΙΠ:
In the avian diagnostic laboratory, quality management may Laboratoiy Biosafety Criteria.
include the utilization of check tests, reference sera, and reference http: //www.cdc. gov/od/ohs/biosfty/bmbl4/bmbl4 s3 .htm.
strains of infectious agents, and maintenance of stock cultures of
2
2
SALMONELLOSIS
W. Douglas Waltman and Richard K. Gast

SUMMARY. Avian Salmonella infections are important as both a cause of clinical disease in poultry and as a source of food-bome
transmission of disease to humans. Host-adapted salmonellae (5. pullorum and S. gallinarum) are responsible for severe systemic diseases,
whereas numerous serotypes of non-host-adapted paratyphoid salmonellae are often carried subclinically by poultry and thereby may
contaminate poultry products. Crowding, mal-nutrition, and other stressful conditions as well as unsanitary surroundings can exacerbate
mortality and performance losses due to salmonellosis, especially in young birds.
Agent Identification. Salmonella infections in poultry flocks are diagnosed principally by the isolation of the bacteria from clinical tissue
samples, such as pooled internal organs and sections of the intestinal tract. Egg contents may also be cultured to detect S. enteritidis.
Preliminary identification of infected or colonized flocks is often obtained by culturing poultry house or hatchery environmental samples,
such as drag swabs, litter, dust, feed, and hatch residue. Clinical and environmental samples are generally subjected to selective enrichment,
with nonselective preenrichment sometimes employed when salmonellae are present in very small numbers or are potentially injured.
Delayed secondary enrichment often improves Salmonella recovery from many sample types. Samples are plated on selective agar and
bacterial colonies are identified as Salmonella by further biochemical and serological testing.
Serologic Detection in the Host. Serologic identification of infected poultry plays an important role in programs for controlling the spread
of Salmonella in commercial flocks, especially in regard to S. pullorum and 5. gallinarum. Agglutination tests, particularly the rapid whole­
blood plate test, are used for verification of the Salmonella status of flocks participating in the National Poultry Improvement Plan.

INTRODUCTION measures be followed any time individuals go onto a farm or into a


hatchery.
Avian Salmonella infections are important as both a cause of
clinical disease in poultry and as a source of food-bome Clinical Cases
transmission of disease to humans. Host-adapted salmonellae are Samples taken from the internal organs of the birds with systemic
responsible for pullorum disease (S. pullorum) and fowl typhoid salmonellosis are typically free of competing bacteria. In such
(S. gallinarum), which are severe systemic diseases that have cases, isolation is usually relatively simple, and may only require
become relatively rare in countries with testing and eradication direct plating on nonselective media. Liver, spleen, heart, heart
programs (13). Infections with non-host-adapted (paratyphoid) blood, ovary or yolk sac, synovial fluid, eye, and brain (if torticollis
salmonellae are common in all types of birds (9,10), although some is observed) are excellent sources for recovery. Sterile cotton-tipped
serotypes are found predominantly in a fairly limited range of hosts swabs are preferred to wire inoculating loops, except for small
(e.g., S. arizonae is isolated mostly from turkeys). Crowding, mal­ organs in young birds, because swabs transfer more tissue. Several
nutrition, and other stressful conditions as well as unsanitary tissues should be cultured from each bird, as tissues with lesions do
surroundings can exacerbate mortality and performance losses due not always yield salmonellae. Portions of several tissues may be
to salmonellosis. Paratyphoid salmonellae can also cause human pooled and added to selective enrichment broth to increase the
illness, and food-bome Salmonella outbreaks can lead to severe possibility of isolation (Fig. 2.1). Sections of the intestinal tract,
economic losses to poultry producers as a result of regulatory especially the ceca and cecal tonsils, may be cultured by inoculation
actions, market restrictions, or reduced consumption of poultry into selective enrichment broth.
products.
Serologic Reactors
CLINICAL DISEASE Because of the potential consequences involved in culturing
serologic reactors, especially pullorum-typhoid, these cases should
Most salmonellae are transmitted horizontally, but some serotypes be cultured more intensively than routine clinical cases. Also, in
(such as S. pullorum and S. enteritidis) can be transmitted vertically some situations reactor birds may be asymptomatic, the salmonellae
and often produce highly persistent flock infections. Pullorum may be present in smaller numbers than in clinical cases, or they
disease primarily affects chickens, turkeys, and other fowls during may be more localized. Recent use of antimicrobial agents may
the first few weeks of life. Fowl typhoid is also observed in mature prevent the recovery salmonellae, although they may not
poultry. These diseases are uncommon in pet birds species and completely clear the organism from the bird.
pigeons. Paratyphoid salmonellae, particularly some strains of Reactors should be evaluated by both direct and selective culture
S. enteritidis, can occasionally cause morbidity or mortality in procedures (Fig. 2.1) (15,17). Tissues showing any pathologic
young birds of diverse species. lesions should be sampled using a sterile cotton-tipped swab and
Histopathologic lesions associated with avian salmonellosis include inoculated onto a nonselective plating medium. If desired the swab
fibrinopurulent perihepatitis and pericarditis; purulent synovitis; may be broken off into a tube of nonselective broth, incubated at
focal fibmoid necrosis, lymphocytic infiltrations, and small 35-37 C for 24 hr, and plated onto nonselective agar.
granulomae in various visceral organs; and serositis of the Portions of the liver, spleen, heart, gall bladder, ovaiy and oviduct
pericardium, the pleuroperitoneum, and the serosa of the intestinal should be pooled (10-15 g total) and then minced, blended, or
tract and mesentery (9,10,13). Infections with S. arizonae can stomached. Selective enrichment broth is added to the tissue (one
produce encephalitis and hypopyon in young turkeys and chickens. part tissue homogenate to 10 parts broth) and incubated at 35-37 C
for 24 hr, and if negative they are incubated for an additional 24 hr.
SAMPLE COLLECTION After the internal organ samples have been removed, the intestinal
samples may be taken. Portions of the ceca and the cecal tonsils
Both clinical tissue and environmental samples should be collected (and other sections of the intestinal tract including the crop may
as aseptically as possible to prevent cross-contamination. This added) should be pooled and minced, blended, or stomached.
includes the use of sterile sampling materials (e.g., swabs, scoops, Selective enrichment broth is added to the sample (one part sample
bags) and disposable gloves. It is critical that strict biosecurity to 10 parts broth), incubated typically at 41 C for 24 hr, and plated
on selective plating media.
3
W. Douglas Waltman and Richard K. Gast

One-Day-old Hatchlings levels above 0.85 appear to promote the environmental survival and
It is often important to determine the presence of Salmonella multiplication of salmonellae.
contamination or infection in 1-day-old chicks or poults. Four The frequency and extent of sampling often varies according to the
methods have been used. The first method, approved by the purpose of the monitoring. For example, the NPIP has established
National Poultry Plan(NPIP), takes 25 1-day-old chicks (in groups guidelines for testing breeding flocks for salmonellae (15).
of 5) and pools the internal organs, yolk sac, and intestinal tracts Environmental samples pose a challenge to the detection of the
from each group in three separate sterile plastic bags (16). Selective salmonellae, because salmonellae are often present in low numbers,
enrichment broth is added to all five groups of pooled samples. salmonellae may be present but injured, and large populations of
A second method cultures chick meconium collected during chick other bacteria are often present. Therefore, highly selective
handling for sexing. Pools of about 5 g of meconium are collected enrichment and plating media must be used.
from the chicks into sterile plastic bags and inoculated with Drag Swabs. Flock infection or contamination can be conveniently
selective enrichment broth. detected by use of drag swabs. Properly assembled and sterilized 3-
A third method cultures papers that line the boxes the chicks are or 4-square inch gauze pads moistened with DSSM are drawn by 3-
transported in from the hatcheiy to the farm. The surface of these 4-ft lengths of cord across the surface of the floor litter or dropping
chick papers may be swabbed with double-strength skim milk pit for 15-20 min. The full length of the occupied building is
(DSSM)-moistened gauze pads or by directly placing pieces of the traversed one or more times. The swabs are placed in individual
paper into a sterile plastic bag and adding selective enrichment sterile plastic bags at the farm and transported as soon as possible to
broth. the laboratory on ice packs. Selective enrichment broth is added
A fourth method, with good sensitivity, uses 50-100 chicks that directly to the bags containing the swabs (15). An alternative to the
have been held for 48-72 hr with only clean water available for gauze pad is a commercially available sponge. Caution is advised to
consumption. This promotes widespread dissemination of infection make sure these sponges are specially made for culturing purposes,
among boxmates and increases the likelihood of detection by because many household-type sponges contain bacteriocidal or
culturing the paper liner or even by cloacal swabbing of the birds in bacteriostatic substances.
some cases. However, an important difficulty in using this method In studies with broiler chickens, results from several unpooled drag
is that the chicks must be maintained without cross contamination swabs closely reflected intestinal excretion rates of salmonellae in
from external sources of Salmonella. the flock (7). Such sampling circumvents more cumbersome
Cloacal Swabs methods of environmental monitoring (5).
Cloacal swabs have been shown to be an unreliable means of Floor Litter. Samples should be collected from representative dry
detecting salmonellae from birds (4). Generally, salmonellae are areas of floor litter; wet and caked ateas, including those around
excreted intermittently, and often in low numbers. Furthermore waterers and feeders, should be avoided because salmonellae
cloacal swabbing of birds requires laborious culture of 300-500 survival may be poor in such areas. A sterile wooden tongue
birds to provide dependable results. depressors or similar device is used to collect 5 g of dry litter from
Egg Culture the upper 2.5-5.0 cm (1-2 inches) of floor litter into a sterile plastic
The surface of intact eggs can be sampled by immersion in 30-50 bag from five sites in the pen or house. Additional sample pools
ml of selective enrichment broth in a plastic bag, with soaking or should be collected from other areas of the house or pen to obtain
manual rubbing for at least 30 sec before removal of the egg. the following ratio of samples to birds per pen: fewer than 500
Eggshells can also be manually crushed in selective enrichment birds, 5 pooled samples; 500-2500 birds, 10 pooled samples; 2500
broth to allow more complete access of the media to internal birds or more, 15 pooled samples (15). The sample numbers
surfaces. After disinfection of eggshells (generally in 70% ethanol suggested above are necessary for reasonable dependability in
or 2% tincture of iodine), egg contents can be cultured at a standard monitoring semimature or mature flocks in which excretion rates
1:10 ratio in selective or nonselective media. may be very low.
The contents of 10-30 eggs are often pooled for culturing because Nest Boxes or Egg Belts. Loose litter in nests is a preferred sample
of the usual low incidence and level of Salmonella contamination. site after breeder hens have been in lay 2-4 wk. Following the same
To allow salmonellae to multiply to easily detectable levels (and to procedure for the nest litter as given for the floor litter, collect fine
minimize media consumption), pools of egg contents should be material from the bottoms of at least five nests for each pooled
incubated for at least 1 day at 37 C or 3 days at 25 C before sample. Samples need not be weighed or measured after some
subculturing into broth media. Incubated egg pools can be experience is acquired, but weight should not exceed 10-15 g per
transferred directly onto selective agar, but significantly greater pooled sample.
detection sensitivity can be attained by using one or more Many companies no longer use wood shavings in their nest boxes
enrichment steps. because of automated egg collection. In lieu of sampling nest
Environmental Monitoring shavings, swabbing the inside of an equal number of nest boxes
Environmental monitoring or surveillance has become a useful using DSSM moistened gauze pads has been shown to be effective.
method for predicting potential infection or colonization of flocks Each pad may be used to sample 5-10 nest boxes and then placed
with the paratyphoid salmonellae. Environmental sampling has been into a sterile bag for culture. It is a good practice to combine
shown to be effective and less invasive than other sampling sampling the floor, by using either litter or drag swabs, with nest
methods. Although environmental monitoring may show good sampling.
predictive evidence of salmonellae in a flock, it is an indirect In lieu of swabbing the nest boxes in houses with mechanical nests,
indicator and flocks should not be diagnosed as infected based the egg belts may be swabbed with DSSM-moistened swabs,
solely on the environmental sampling. usually at the front of the house.
The probability of detecting salmonellae in environmental samples Dust. In some regions, dust has been a more dependable source for
depends on the number of salmonellae excreted into the isolation of salmonellae than litter. Local experience should be a
environment by the flock, the survival of salmonellae in the sample guide for determining the most dependable source. Samples are
locations, the intensity of sampling, and the culture methods used. collected by DSSM moistened gauze pads or by wooden tongue
Fecal shedding of salmonellae is often greatest during the first few depressors from 15 or more sites of dust accumulation per pen.
weeks of life, when the susceptibility of chicks to infection is Depending on the volume collected, five samples may be pooled
greatest. Litter surface water activity (equilibrium relative humidity) into one. Use cotton-tipped swabs only for hard-to-reach areas.

4
Chapter 2 Salmonellosis

Cage Housing. The best methods of sampling for cage houses have PREFERRED CULTURE MEDIA AND SUBSTRATES
not been fully determined. Methods in current use include sterile
gauze pads moistened with DSSM to aseptically collect material Recommended Steps, Media, and Incubation Times and
from egg transport belts and elevators, rollers, diverters, and manure Temperatures.
scrapers or cables. All segments of the house should be sampled. The isolation and identification flow chart in Fig. 2.1 details the
The drag swab method is also used with layer cages that allow selection and use of preferred media in a variety of testing
droppings to accumulate under cages. situations. Selective enrichment broths are typically inoculated at a
Hatcheries. The area most likely to yield salmonellae is the 1:10 ratio of sample to broth, except for RV enrichment, which is
hatcher during or following hatching. Selective Agar plates, opened inoculated at a 1:100 ratio from a pre-enrichment broth.
and exposed for 5-10 min to the air in various parts of the hatcheiy Typically, internal organ samples and samples having lower
building, have been used to monitor hatcheries. However, failure to background levels of bacteria may be incubated at 35-37 C.
find salmonellae on such plates is not a reliable indication of Intestinal and environmental samples having higher levels of
freedom from hatchery contamination. background bacteria are commonly incubated at 40-43 C. Because
A sterile tongue depressor may be used to collect at least one of potential incubator problems and the sensitivity of some strains
heaping tablespoonful of fluff and dust from each hatcher. This to higher temperatures, the preferred incubation temperature is
sample is placed into a sterile plastic bag with selective enrichment 41+0.5 C. Routine monitoring of the temperature of incubators is
broth. critical when using the higher temperatures.
The most reliable sample has been the hatch residue (2). About 10- Selective enrichment broths should be incubated 20-24 hr and
15 g of eggshells and other hatch residue remaining after the chicks plated. Use of a delayed secondary enrichment (DSE) procedure is
are removed from the hatch trays is placed into a sterile plastic bag highly recommended (see below), but if not, the enrichment broths
to which selective enrichment broth is added. Alternatively, several should at least be incubated an additional 24 hr and replated.
hatch trays may be swabbed with gauze pads and pooled.
Feed and Feed Ingredients. About 100 g of feed or feed Precautions for Environmental and Intestinal Samples
ingredients should be collected into a sterile plastic bag representing Isolation of salmonellae from environmental and intestinal
multiple sites in a lot of feed. Care should be taken to prevent cross monitoring samples is much more demanding than isolation from
contamination of the samples. samples collected from bacteremic (clinically affected) or carrier
The Association of Official Analytical Chemists (AOAC) (serologic reactor) birds. In bacteremic or carrier birds, salmonellae
International method is the most widely accepted procedure for populations are often high, particularly with respect to bacterial
culturing feed and feed ingredients (1). Twenty-five grams of feed competitors. In contrast, salmonellae levels are ordinarily low in
is mixed with 225 ml of lactose broth. The mixture is allowed to environmental or intestinal samples where competing bacteria are
stand for 1 hr and the pH is adjusted to 6.8 + 0.2. The culture is found in high levels. These competitors, if not controlled, seriously
incubated at 35 C for 24 hr. One milliliter of the culture is impair media detection efficiency, resulting in false negatives.
transferred into 10 ml of tetrathionate (TT) broth and 0.1 ml is The dependable isolation of salmonellae from environmental and
transferred into Rappaport-Vassiliadis (RV) broth. The two intestinal samples is, therefore, significantly improved by
enrichment broths are incubated at 43 C and 42 C, respectively. incubation of enrichment broths at 41 + 0.5 C for a full 24-36-hr
After 24 hr, the cultures are inoculated onto bismuth sulfite, period, use of novobiocin- or tergitol (niaproof) 4-supplemented
Hektoen enteric (HE), and xylose lysine desoxycholate (XLD) media, DSE of primary selective enrichment broths, and use of a
agars. plate-streaking technique that produces well-separated colonies.
Some investigators have suggested that this procedure be modified
by using universal preenrichment or buffered peptone water (BPW) Non-Selective Media
as the preenrichment broth; by reducing the selective enrichment Salmonellae grow well on such nonselective broth media as veal
incubation temperatures to 41 C; and by replacing the plating media infusion, brain-heart infusion, or nutrient broth. Nonselective agars
with more selective agars, such as brilliant green supplemented with that may be used include blood agar and nutrient agar. MacConkey
novobiocin (BGN) and xylose lysine tergitol 4 (XLT4) agars. agar, although mildly selective, is frequently used as a nonselective
medium.
Shipping and Storing Samples These media are preferred for tissues from clinical cases and
Holding Media. Studies have shown that the best moistening agent serologic reactors in which the likelihood of competing bacteria is
and holding media for environmental swabs is DSSM (12). This low. Use of nonselective media is important when S. pullorum or S.
medium is prepared by dissolving 200 g Bacto Skim Milk (BD gallinarum are suspected, as some strains may be inhibited by
Diagnostics Systems, Sparks, MD) in 1 liter distilled or deionized selective media.
water in a large flask and autoclaving. The DSSM may be stored in
the refrigerator. Pre-enrichment
Sample Storage Times and Temperatures. Specimens from Preenrichment is the process of inoculating nonselective broth
clinical diagnostic consignments and serologic reactors should be media with the sample and incubating it for 24 hr at 35-37 C.
cultured immediately upon collection and no more that 24 hr after Typically, 1.0 ml of the preenriched culture is transferred to 10 ml
collection even if they are refrigerated. Meconium, freshly voided of a TT enrichment broth or 0.1 ml is transferred to 10 ml of RV
feces, and water should be refrigerated at Ί-b C as soon as possible enrichment broth. The purpose of preenrichment is to revive injured
and inoculated into appropriate culture media within 24 hr. Drag salmonellae that may be present in some samples. Typical
swabs and other swabs containing DSSM may be refrigerated for 3 preenrichment media include lactose broth and BPW.
days or frozen for 7 days before inoculation of selective enrichment
broth (12). Because the survival rate of salmonellae in floor litter Selective Enrichment Broths
samples varies considerably, even under refrigeration, it is best to Tetrathionate Enrichments. Tetrathionate broths are the preferred
culture floor litter within 2 days of collection. This is advisable selective enrichments for salmonellae isolation. Older formulations,
despite the fact that some dry environmental samples have yielded such as Mueller-Kauffrnann TT brilliant green broth, require the
salmonellae when held for several months at room temperature and addition of both iodine and brilliant green solutions to the broth
low humidity. immediately before use. However, newer formulations, such as TT
broth, Hajna, or TT, Hajna and Damon (BD Diagnostic Systems,
5
W. Douglas Waltman and Richard K. Gast

Sparks, MD) are supplied already containing brilliant green and Xylose Lysine Desoxycholate, XLD Supplemented with
require only the addition of an iodine solution to the broth base. The Novobiocin, and XLT4 Agars. The differential characteristics of
newer TT formulations contain more nutrients and buffers. The these media include the lysine decarboxylation and H2S-producing
iodine solution differs for the individual TT formulations, so care abilities of most salmonellae. Hydrogen sulfide-positive
must be taken to ensure the appropriate one is used and that it is salmonellae colonies are nearly or fully jet black. Proteus (inhibited
prepared and stored correctly. on XLDN and XLT4) colonies may appear blackish, but the color is
Selenite Enrichments. The use of selenite enrichments are no less brilliant, tending to have a gray or green cast as the colonies
longer recommended because they have a short shelf life, are not as age. Colonies of Citrobacter freundii ordinarily have black centers
effective at higher incubation temperatures (3), potentially and prominent creamy-colored borders on these media.
mutagenic (11), and in many geographic areas are considered to be Other Plating Media. Bismuth sulfite and Hektoen enteric agars,
a hazardous waste. although widely used in human clinical situations and advocated by
Rappaport-Vassiliadis Enrichments. A selective enrichment that the Food and Drug Administration (FDA) and AOAC International
is gaining acceptance and appears to be replacing selenite for food and feed, lack the sensitivity and specificity of BGN and
enrichments is RV broth. The RV enrichment procedure begins with XLT4. Modified lysine iron agar (MLIA) incorporates novobiocin
preenrichment of the sample in BPW and then inoculation into RV and is similar in recovery to BGN or XLDN. Rambach agar, a new
broth at a ratio of 1:100. The RV broth should be incubated at 41- chromogenic agar, appears to be effective, but it is more expensive
42 C. than other options. Recently, several other chromogenic agars have
been developed for the isolation of Salmonella. The most
Delayed Secondary Enrichment (DSE) thoroughly studied of these, Miller-Mallinson (MM) media, has
After the 24-hr incubation of the TT and plating onto BGN and been found to be very effective.
XLT4 agars, the TT culture is left at room temperature for 5-7 days.
If the original plating was negative for salmonellae, 0.5-1.0 ml of Rapid Salmonella Detection Techniques
the original TT broth is transferred into 10 ml of fresh TT broth and A number of rapid tests have been approved for detecting
incubated at 37 C for 24 hr. The new TT broth is plated and salmonellae in various samples. Typically these have been approved
processed as before (16). Studies have shown that DSE increases based on comparison to AOAC International culture procedures (1).
the recovery rates of salmonellae from most sample types (18). The rapid tests have several advantages over conventional culture,
such as obtaining results in less than 48 hr, being less labor
Selective Plating Media intensive, and having the possibility of automation. For many
Rationale for Using. Because of the high levels of nonsalmonellae sample types, especially clinical, processing plant, and food
bacteria in most intestinal and environmental samples, selective samples, these rapid tests appear to be veiy effective. However,
plating media must be used to assist the selective enrichment broth some sensitivity and specificity problems are apparent with feed
in inhibiting other bacteria. Conventional salmonellae plating media and environmental samples.
lacks the selectivity necessary for these samples. Many media can A variety of rapid detection systems are commercially available,
inhibit most coliforms, however, the primary problems are with including antigen-capture enzyme-linked immunosorbent assay
species of Proteus, Providencia, Morganella, and Pseudomonas. (AC-ELISA) systems, DNA probes, polymerase chain reaction
These bacteria resemble salmonellae on some plating media and systems, immunodiffusion, immunofluorescence, magnetic bead
must be screened, resulting in high percentages of false-positive systems, bioluminescence, and other novel applications. A listing of
cultures. The addition of 20 pg of novobiocin (N1628 Sigma these tests may be found at the FDA-BAM website. Currently,
Chemical Co., St. Louis, Mo.) per ml of plating media, especially NPIP has approved a rapid ruthenium-labeled sandwich
brilliant green (BG) (14) and XLD agars, results in the inhibition of immunoassay and two PCR systems for detecting Salmonella in
Proteus. Another plating medium that has proven to be effective is environmental samples (15). As technology advances the sensitivity
XLT4 (BD Diagnostic Systems, Sparks, MD). This medium inhibits and specificity of rapid systems will surely increase and allow more
Proteus and Pseudomonas aeruginosa. A combination of two widespread use.
plating media that are predicated on different selective and
differential characteristics is advocated. BGN and XLT4 are two AGENT IDENTIFICATION
good choices (8). The use of BGN agar, which demonstrates the
positive hydrogen sulfide (H2S) production characteristic, increases Basic Identification Screening Media
the likelihood of detecting atypical strains. Hydrogen sulfide­ The combined use of TSI and LI agar slants is generally sufficient
negative strains undetected on XLT4 should be obvious on BGN, for presumptive identification of most salmonellae-suspect colonies.
because they usually remain lactose-negative. Conversely, lactose­ At least three well-separated colonies are selected for transfer to
positive strains (eg., S. arizonae and others) undetected on BGN each set of TSI and LI agar slants. Each colony is stabbed into the
should be obvious on XLT4 because they usually remain MS­ butt of the TSI and LI agars and streaked across the slants. The
positive. tubes are read after incubation for 24 hr at 37 C. The use of more
Brilliant Green (BG) Supplemented with Sulfapyridine or selective plating media (BGN and XLT4) to screen these
Sulfadiazine (BGS), and BGN Agars. Brilliant green agar has salmonellae suspect colonies results in fewer false positives
been supplemented with antimicrobial compounds to make it more (nonsalmonellae bacteria).
selective. The purpose of adding sulfapyridine or sulfadiazine and The presence of multiple serotypes may be more likely in some
novobiocin was primarily to inhibit Proteus species. Salmonellae samples than in others. Also, in situations where S. enteritidis, S.
colonies on these media are usually transparent pink to deep pullorum, qt S. typhimurium are being monitored, and other
fuchsia, surrounded by a reddish medium. These colonies may lose salmonellae are present, screening many more colonies may be
this characteristic appearance if there is a heavy growth of lactose- necessary to ensure the absence of S. enteritidis, S. pullorum, or S.
fermenting colonies or if the colonies are not well separated. Some typhimurium serotypes. The Colony Lift Immunoassay (Synbiotics
nonsalmonellae bacteria produce colonies similar in appearance to Corp., San Diego, CA) may be useful in detecting group D
salmonellae and must be screened further. Host-adapted S. pullorum salmonellae in these situations (6).
and 5. gallinarum colonies are smaller and grow slower than non- Triple Sugar Iron (TSI) Agar. Most salmonellae produce an
host-adapted salmonellae. Consequently, all plates should be alkaline (red) slant and acid (yellow) butt, with gas bubbles in the
incubated for 48 hr. agar and a blackening due to H2S production that often obscures the
6
Chapter 2 Salmonellosis

acid reaction in the butt of the tube. Salmonella gallinarum does not SEROLOGIC DETECTION IN THE HOST
form gas in TSI, whereas S. pullorum may show weak gas
production. Both of these Salmonella may or may not show H2S Serologic Testing of Poultry Flocks
production. Some paratyphoid strains may be H2S-negative in this Several serologic tests have been developed for detecting
medium. antibodies to salmonellae. The most commonly used tests for
Lysine Iron (LI) Agar. Salmonellae will show lysine pullorum-typhoid include the whole blood plate (WBP) (not
decarboxylation, with a deeper purple (alkaline) slant and alkaline approved for use in turkeys), rapid serum plate, standard
or neutral butt with slight blackening due to H2S production, with macroscopic tube agglutination, microagglutination, and
the exceptions noted in the previous paragraph. LI agar is useful in microantiglobulin tests (9,15). Possibly the most widely used of
differentiating common intestinal flora such as Proteus and these tests is the WBP test, which can be performed in the field
Citrobacter from further consideration. Proteus (also Providencia using commercially available antigens. A drop of fresh blood is
and Morganella) produces a reddish or port-wine colored slant, mixed with a drop of antigen on a plate, mixed, and observed for
indicating lysine deamination, and a yellow (acid) butt on LI agar agglutination within 2 min.
medium. Citrobacter gives a purple (alkaline) slant and a yellow The tube agglutination test is also commonly used. It requires sera
(acid) butt, with some H2S production. Rarely, both slant and butt for testing with an antigen that may be obtained from the NVSL. In
are yellow. the macroscopic tube agglutination test the sera are diluted 1:25 to
1:50 for chickens or 1:25 for turkeys. The sera are mixed with the
Biochemical and Serological Confirmation antigen, incubated at 37 C for 20-24 hr, and observed for
Biochemical Identification. Further biochemical tests may be agglutination. Because the WBP test is not approved for turkeys, the
necessary to confirm an isolate as Salmonella. Table 2.1 lists tube agglutination or rapid serum agglutination tests are typically
several typical biochemical reactions of salmonellae. Key tests for used.
differentiating salmonellae from other bacteria include those for Agglutination tests have also been used for detecting antibodies to
urea {Proteus and most Providencia and Morganella are urease various paratyphoid salmonellae, especially S. typhimurium, S.
positive), beta-galactocidase (salmonellae with the exception of arizonae, and S. enteritidis. These tests have met with varying
S. arizonae, are negative whereas C. freundii and other coliforms degrees of success due to sensitivity and specificity problems and
are positive), and indole (Escherichia coli and Escherichia tarda thus have not found widespread commercial application. Because S.
produce indole, salmonellae do not). enteritidis and S. pullorum share somatic antigens, pullorum-
Commercially available identification systems may also be very typhoid antigen preparations have sometimes been applied for
useful in confirming isolates. They use a number of biochemical detecting antibodies to S. enteritidis.
tests in a simple to use format. Enzyme-linked immunosorbent assays (ELISAs) have been
Serological Typing. The presumptive salmonellae colonies developed for detecting antibodies to various salmonellae. The most
identified by the TSI and LI agar reactions should be serologically widely used ELISAs have been developed for detecting antibodies
typed. The first phase of serologic typing is to serogroup the isolates to S. enteritidis. These ELISAs have been used with some success,
based on their somatic O-group antigens using commercially particularly in Europe, but concerns about their specificity still
available polyvalent somatic antisera. Individual polyvalent antisera persist.
are tested against each isolate in a simple plate agglutination assay. The various serologic tests for detecting antibodies to salmonellae,
Bacterial growth from an agar plate is emulsified in a small amount especially the agglutination tests, are subject to false positive
of physiologic saline to form a milky suspension, and one drop of reactions. Confirmation of all positive serologic tests by culturing
polyvalent O antisera is mixed with it on a slide or plate. The the bird for salmonellae is important.
agglutination reaction is read within 60 sec. After determining
which polyvalent antiserum agglutinates with the isolate, additional DIFFERENTIATION FROM CLOSELY RELATED AGENTS
testing is performed using individual single factor O-group antisera
that comprised the polyvalent antiserum. In this maimer, each Young birds with generalized Salmonella infections may show
isolate is typed to a specific serogroup. signs and lesions identical to any bacteremia. In severe outbreaks of
The next step, which is often performed at a reference laboratory, salmonellosis, fibrinous liver and heart lesions may be very similar
is to determine the serotype. The serotype is based on the flagellar to those seen with air sac disease (colibacillosis). The heavy
antigen present on almost all salmonellae, except 5. pullorum and yellowish white cheesy exudate covering the retina of turkey poults
S. gallinarum. Commercially produced antisera are available for with S. arizonae infection and occasionally with other paratyphoid
serotyping most isolates (BD Diagnostic Systems, Sparks, MD). infections can be confused with signs of aspergillosis. Nervous
The serotyping procedure involves an antigen extraction step with signs associated with infection of the brain of fowl may resemble
formalin, followed by a microtube agglutination test. those of Newcastle disease or other diseases affecting the central
nervous system. Joint involvement may be mistaken for synovitis or
bursitis due to other infectious agents.

7
W. Douglas Waltman and Richard K. Gast

Figure 2.1 General Salmonella /solation and identification

A Hajna TT or Mueller-Kauffmann tetrathionate enrichment broths.


For RV broth, follow special inoculation and preenrichment instructions of manufacturer
B Beef extract, veal infusion, or comparable non-selective media.
A broth detect can help detect low Salmonella levels in live birds.
C BGN in combination with XLT4 is preferred (refer to text)
D Colony lift immunoassays can significantly increase the reliability detecting Group D Salmonella
(S. enteritidis, S. pullorum, etc. on plating agars (refer to text)
E If combined results with TSI and LI agars, additional identification media, and O-group screening
procedures are inconclusive, restreak original colony onto selective plating agar to check for purity.
F Reevaluate if epidemiologic, necropsy or other information strongly suggests the presence of an unusual
strain of Salmonella.

8
Chapter 2 Salmonellosis

Table 2.1 Typical biochemical reactions of Salmonella K 4. Fanelli, M J., W. W. Sadler, C. E. Franti, and J. R. Brownell.
Paratyphoid Localization of salmonellae within the intestinal tract of chickens. Avian
Media S. pullorum S. gallinarum S. arizonae
salmonellae Dis. 15:366-375. 1971.
Dextrose A(G)b A AG AG 5. Kingston, D. J. A comparison of culturing drag swabs and litter for
Lactose - - - (-)C identification of infections with Salmonella spp. in commercial chicken
flocks. Avian Dis. 25:513-516. 1981.
Sucrose - - - - 6. Lamichhane, C. M, S. W. Joseph, W. D. Waltman, T. Secott, E. M Odor,
Mannitol A(G) A AG AG J. deGraft-Hanson, E. T. Mallinson, V. Vo, and M Blankford. Rapid
Maltose (-)D A AGe AG detection of Salmonella in poultry using the colony lift assay. In:
Dulcitol - A AG - Proceedings of the Southern Poultry Science Society. Atlanta, Ga. Poult.
Sci. (Suppl. 1)74:198. 1995.
Malonate - - - +F
7. Mallinson, E. T., C. R. Tate, R. G. Miller, B. Bennett, and E. Russek-
Urea - - - - Cohen. Monitoring poultry farms for Salmonella by drag swabs sampling
Motility - - + + and antigen capture immunoassay. Avian Dis. 33:684—690. 1989.
A = acid produced; G = gas produced; 0 = variable reaction 8. Miller, R. G., C. R. Tate, E. T. Mallinson, and J. A. Scherrer. Xylose
B Blood agar is generally used as the non-selective agar plate. If desired, the lysine tergitol 4: an improved selective agar medium for the isolation of
Salmonella. Poult. Sci. 70:2429-2432. 1991.
swabs or pieces of tissue may be inoculated into a non-selective broth, such
9. Gast, R. K. Paratyphoid infections. In: Diseases of poultry, 11th ed. Y. M
cas brain-heart infusion or nutrient broth. Saif, H. J. Barnes, J. R. Glisson, A M Fadly, L. R. McDougald, and D. E.
Brilliant green agar supplemented with novobiocin (BGN) in combination
Swayne, eds. Iowa State University Press, Ames, Iowa. pp. 583-613. 2003.
with xylose-lysine-tergitol (niaproof) 4 (XLT4) agar are preferred (refer to 10. Nagaraja, K. V., B. S. Pomeroy, and J. E. Williams. Arizonosis. In:
text). If S. pullorum is suspected, MacConkey (MAC) agar may be Diseases of poultry, 9th ed. B. W. Calnek, H. J. Bames, C. W. Beard, W. M
substituted for XLT4 agar).
Reid, and H. W. Yoder, Jr., eds. Iowa State University Press, Ames, Iowa,
D Colony lift immunoassays may increase the reliability of detecting Group
pp. 130-137. 1991.
D Salmonella (e.g. S. enteritidis, S. pullorum, etc) on plating media (refer to 11. Noda, Μ, T. Takano, and H. Sakurai. Mutagenic activity of selenium
text). compounds. Mutat. Res. 66:175-179. 1979.
E Salmonella typhimurium var. Copenhagen, as isolated from pigeons, 12. Opara, Ο. O., L. E. Carr, C. R. Tate, R. G. Miller, E. T. Mallinson, L. E.
frequently forms no acid in maltose broth and can be confused with S. Stewart, and S. W. Joseph. Evaluation of possible alternatives to double­
pullorum. However, it is motile. strength skim milk used to saturate drag swabs for Salmonella detection.
F Turns deep Prussian blue within 24 hr. Avian Dis. 38:293-296. 1994.
13. Shivaprasad, Η. N. Pullorum Disease and Fowl Typhoid. In: Diseases of
poultry, 11th ed. Y.M Saif, H. J. Bames, J; R. Glisson, A. M Fadly, L. R.
McDougald, and D. E. Swayne, eds. Iowa State University Press, Ames,
ACKNOWLEDGMENT Iowa. pp. 568-582. 2003.
14. Tate, C. R., R. G. Miller, E. T. Mallinson, and L. W. Douglass. The
The guidance supplied by Edward T. Mallinson is very gratefully isolation of salmonellae from poultry environmental samples by several
acknowledged enrichment procedures using plating media with and without novobiocin.
Poultry Sci. 69:721-726. 1990.
REFERENCES 15. United States Department of Agriculture (USDA). National Poultry
Improvement Plan and Auxiliary Provisions. Animal and Plant Health
1. Association of Official Analytical Chemists (AOAC) International. Inspection Service, 91-55-063. USDA, Washington, D.C. Feb 2004.
Official methods of analysis, 16th ed. P. Cunniff, ed. AOAC International, 16. Waltman, W. D., A. M Home, C. Pirkle, and T. G. Dickson. Use of
Gaithersburg, Md. pp. 55-94. 1996. delayed secondary enrichment for the isolation of Salmonella in poultry and
2. Bailey, J. S., N. A. Cox, and Μ E. Berrang. Hatchery-acquired poultry environments. Avian Dis. 35:88-92. 1991.
salmonellae in broiler chicks. Poult. Sci. 73:1153-1157. 1994. 17. Waltman, W. D., and A. M Home. Isolation of Salmonella from
3. Carlson, V. L., and G. H. Snoeyenbos. Comparative efficacies of selenite chickens reacting in the pullorum-typhoid agglutination test. Avian Dis.
and tetrathionate enrichment broths for the isolation of Salmonella 37:805-810. 1993.
serotypes. Am. J. Vet. Res. 35:711-718. 1974. 18. Waltman, W. D., A. M Home, and C. Pirkle. Influence of enrichment
incubation time on the isolation of Salmonella. Avian Dis. 37:884—887.
1993.

9
3
COLIBACILLOSIS
Margie D. Lee and and Lisa K. Nolan

SUMMARY. Escherichia coli is the causative agent of colibacillosis in poultry. The disease results from a systemic infection involving the
blood, joints, and/or air sacs of birds. Young birds (4-8 wk old) may die of acute septicemia that is preceded by only a brief period of
anorexia and depression. At necropsy, lesions are sparse but may include swollen, dark-colored liver and spleen and increased fluid in the
body cavities. Birds surviving the acute phase may develop fibrinopurulent airsacculitis, pericarditis or arthritis. Whether lesion-associated
isolates are primary pathogens or whether environmental factors are responsible is unknown at this time.
Agent Identification. Isolation of E. coli is significant if made from the internal organs or blood from fresh carcasses. MacConkey agar is
selective and differential for E. coli and is preferred for primary isolation. Presumptive positive colonies (pink on MacConkey that produce
A/A reaction on triple sugar iron agar and that are oxidase-negative, gram-negative rods should be confirmed as E. coli by a positive indole
test and the inability to produce H2S. But some isolates do not form pink colonies on MacConkey, therefore multiple colonies should be
separately inoculated on triple sugar iron agar to detect these pathogens.
Serologic Detection in the Host. Host serology is not useful for diagnosis because many birds have antibody to normal intestinal flora
isolates of E. coli.

INTRODUCTION fibrinopurulent lesions suggest subacute colibacillosis, swab


samples of exudate should be collected from the pericardial sac, air
Colibacillosis of poultry is a common systemic infection caused by sacs, and joints. Lesions present more than 1 wk are often sterile.
avian pathogenic Escherichia coli (APEC). The disease is When postmortem changes are obvious, bone marrow samples may
economically important to poultry production worldwide. be useful because they are less likely than other tissues to contain
Colibacillosis occurs in many forms. It may occur as intestinal E. coli. Escherichia coli isolates survive well on sealed
colisepticemia, which typically leads to death. However, some agar slants for storage and shipping. For long-term storage, mix E.
birds fully recover from colisepticemia, and others may recover coli broth culture with sterile glycerol 1:1 and store at -20 to -60 C.
with sequelae (2). Colibacillosis may also be localized, manifesting
as omphalitis, yolk sac infection, cellulitis, swollen head syndrome, PREFERRED CULTURE MEDIA AND SUBSTRATES
enteritis, acute vaginitis, salpingitis, or peritonitis (2,12). For E. coli
infections to become clinically apparent, adverse environmental Escherichia coli grows well in most commonly used culture
factors or other infectious agents are usually required (2, 3). A media, but differential media are useful for primary isolation.
foodbome link between human disease and APEC has not been MacConkey's agar is a selective and differential medium for the
established. However, recent reports of similarities between the isolation of enteric organisms. Tryptose blood agar with 5% bovine
virulence attributes of APEC and human extraintestinal pathogenic blood can be used as a primary culture medium to support growth of
E. coli, suggest that some APEC may be capable of causing disease E. coli and other bacterial pathogens. Several differential
in human beings (4,10). biochemical characteristics can be obtained through use of Kligler’s
iron agar or triple sugar iron agar slants. All the above media are
CLINICAL DISEASE readily available from BD Diagnostics (Sparks, Md.).
Microbiology identification kits such as the API 20E (bioMerieux
Clinical signs of colibacillosis are nonspecific and vaiy with the Vitek, Hazelwood, Mo.) and Enterotube (Becton Dickinson
age of bird, duration of infection, organs involved, and concurrent Microbiology Systems) are useful for performing the biochemical
disease conditions. In young (4 to 8-wk-old) broilers and poults tests and are available commercially.
dying of acute septicemia, death is preceded by a brief period of
anorexia, inactivity, and somnolence. At necropsy, lesions are AGENT IDENTIFICATION
sparse except for swollen, dark-colored liver and spleen and
increased fluid in all body cavities. Birds surviving the septicemia Colony Morphology and Biochemical Features
phase of the disease are unthrifty and develop subacute Blood samples should be diluted 1:10 in brain-heart infusion broth
fibrinopurulent airsacculitis, pericarditis, perihepatitis, and then inoculated directly onto MacConkey’s plates. Swabs from
lymphocyte depletion in the bursa and thymus. Airsacculitis,a lesions can be used to streak MacConkey’s plates. MacConkey’s
classic lesion of colibacillosis, occurs following respiratory agar should be incubated aerobically for 18-24 hr at 37 C for the
exposure to large numbers of E. coli, but also occurs as a sequel to primary isolation of E. coli. Escherichia coli, Enterobacter, and
bacteremia. Other less common lesions are arthritis, osteomyelitis, Klebsiella ferment lactose and can be distinguished from the other
salpingitis, and pneumonia (1,3,6). Cellulitis, responsible for enteric organisms. On MacConkey's agar, most E. coli and some
substantial economic losses for the poultry industry, has also been Enterobacter isolates produce 1-2-mm-diameter hot-pink colonies
associated with E. coli infection (12). (lactose positive) whereas Klebsiella and Enterobacter aerogenes
form large mucoid pink colonies (8). However slow lactose-
SAMPLE COLLECTION fermenting E. coli are frequently isolated from cases of
colibacillosis and these may not form pink colonies. If primary
Only internal organs or blood, not feces or intestine, are useful cultures reveal large numbers of a predominant colony type
samples. Because normal intestinal flora E. coli readily invade suggestive of E. coli, pick several of these colonies and use them to
other tissues after death, specimens from fresh carcasses are separately inoculate triple sugar iron agar (or Kligler’s iron agar),
necessary. When acute colisepticemia is suspected, heart blood and sulfide-indole-motility (SIM) medium, and blood agar plates. If the
liver should be sampled aseptically. One ml of blood collected by clinical signs suggest colibacillosis infection, and no hot-pink
needle and syringe can be used to inoculate broth media (1:10), colonies are seen on the MacConkey’s plates, pick a couple of non­
which is used to streak agar plates. Sterile culture swabs or pink colonies to inoculate blood agar, triple sugar iron agar and
inoculation loops can be stabbed into the liver parenchyma after SIM. A Gram stain and the oxidase reaction can be performed on
searing the capsule with a flamed scalpel or spatula. When colonies from blood agar plates. Escherichia coli, Enterobacter,
10
Chapter 3 Colibacillosis

and Klebsiella are oxidase-negative, gram-negative rods. On triple Antimicrobial Susceptibility


sugar iron agar (or Kligler’s iron agar), these organisms produce APEC isolates are frequently resistant to more than one antibiotic.
acid (even the slow lactose-fermenting E. coli) and gas but not H2S. Seventy to 90% of isolates are resistant to sulfa drugs, tetracyclines,
On SIM medium, E. coli is positive for the indole reaction, positive streptomycin, and gentamicin. It is not uncommon to find isolates
or negative for motility, and negative for H2S, whereas that are multi-resistant to greater than 3 antibiotics. Resistance to
Enterobacter and Klebsiella are usually indole-negative (Fig. 3.1). fluoroquinolones is less frequently detected; a recent study (13)
The site of sample collection, condition of the carcass, and nature reported that 84% of isolates were susceptible to enrofloxacin.
of the lesion are important when deciding whether isolation of E.
coli is relevant. The isolation of a pure culture of E. coli from the SEROLOGIC DETECTION IN THE HOST
organs or blood of moribund birds or freshly dead carcasses is
indicative of colibacillosis. Pathogenicity of E. coli isolates has Serology is not commonly used to detect E. coli infection.
traditionally been established by inoculating young (less than 3-wk-
old) chicks or poults parenterally with 0.1 ml of overnight broth DIFFERENTIATION FROM CLOSELY RELATED AGENTS
culture. Pathogenic isolates should produce death or characteristic
lesions of colibacillosis within 3 days. Alternately, inoculation of Colibacillosis should be distinguished from other bacterial
embryonated eggs may be used to establish the pathogenicity of E. infections causing fatal septicemia or fibrinopurulent inflammation
coli isolates (7). Also, many APEC share a complex of plasmid- of air sacs, pericardium, joints, and other viscera. Diseases to be
associated virulence genes, whose presence may be helpful in considered in the differential diagnosis include mycoplasmosis,
distinguishing APEC from non-pathogenic strains (9). Hence, salmonellosis, pasteurellosis, pseudotuberculosis, erysipelas,
multiplex PCR, targeting these common genes, may have value in chlamydiosis, and staphylococcosis. Colibacillosis is a common
confirming the pathogenic nature of E. coli isolates (11). complication of concurrent viral respiratory or enteric infections.

REFERENCES

1. Arp, L.H. Pathology of spleen and liver in turkeys inoculated with


Escherichia coli. Avian Pathol. 11:263-279. 1982.
2. Barnes, H.J., J.-P. Vaillancourt, and W.B. Gross. Colibacillosis. In:
Diseases of poultry, 11th ed Y.M Saif, H. J. Bames, J.R. Glisson, A.M
Fadly, L.R. McDougald, and D.E. Swayne, eds. Iowa State University
Press, Ames, Iowa. pp. 631-652. 2003.
3. Gross, W.B. Colibacillosis. In: Diseases of poultry, 9th ed. B.W.
Calnek, H.J. Bames, C.W. Beard, W.M Reid, and H.W. Yoder, eds. Iowa
State University Press, Ames, Iowa. pp. 138-144. 1991.
4. Johnson, J.R., A.C. Murray, A. Gajewski, M Sullivan, P. Snippes, MA.
Kuskowski, and K.E. Smith. Isolation and Molecular Characterization of
nalidixic acid-resistant extraintestinal pathogenic Escherichia coli from
retail chicken products. Antimicrob Agents Chemother. 47: 2161-2168.
2003.
5. Kaper, J. B., J.P. Nataro, and H.L.T. Mobley. Pathogenic Escherichia
coli. Nature Rev. Microbiol. 2: 123-140. 2004.
6. Nakamura, K., M Maecla, Y. Imada, T. Imada, and K. Sato. Pathology of
spontaneous colibacillosis in a broiler flock. Vet. Pathol. 22:592-597. 1985.
7. Nolan, L.K., R.E. Wooley, J. Brown, K.R. Spears, H.W. Dickerson, and
M Dekich. Comparison of a complement resistance test, a chicken embryo
lethality test, and the chicken lethality assay for determining virulence of
avian Escherichia coli. Avian Dis. 36:395-397. 1992.
8. Quinn, P.J., ME. Carter, B.K. Markey, and G.R Carter. Clinical
veterinary microbiology. Mosby-Year Book Limited, London, England,
pp. 209-236. 1994.
Figure 3.1. Escherichia coli identification scheme .Diagnosis of 9. Rodriguez-Siek, K.E., C.W. Giddings, M Fakhr, C. Doetkott, T.J.
colibacillosis is dependent on distinguishing pathogenic isolates from Johnson, and L.K. Nolan. Characterizing an APEC pathotype. Vet Res. 36:
nonpathogenic, normal intestinal flora isolates of E. coli. 1-16. 2005.
10. Rodriguez-Siek, K.E., C.W. Giddings, T.J. Johnson, M Fakhr, C.
Serogrouping of Isolates Doetkott, and L.K. Nolan. Comparison of Escherichia coli implicated in
Escherichia coli serogroups are based on O antigens; but serotypes human urinary tract infection and avian colibacillosis. Microbiol. 151:
of E. coli are based on O antigens, as well as flagellar and/or 2097-2110. 2005.
11. Skyberg, J.A, S.M Home, C.W. Giddings, R.E. Wooley, P.S. Gibbs, and
capsular antigens (5). There is great diversity of serogroups/types L.K. Nolan. Characterizing avian Escherichia coli isolates with multiplex
among APEC, although some occur more commonly than others PCR Avian Dis. 47:1441-1447,2003.
(e.g., 078 and 02), and many isolates are not typable (9). Routine 12. Vaillancourt, J.-P., and H.J. Bames. Coliform Cellulitis. In: Diseases
serotyping of APEC is usually impractical, but for epidemiologic of poultry, 11th ed. Y.M Saif, H. J. Bames, J.R Glisson, A.M Fadly, L.R.
studies, isolates can be serotyped for a fee by the Gastroenteric McDougald, and D.E. Swayne, eds. Iowa State University Press, Ames,
Iowa. pp. 652-656. 2003.
Disease Center (Pennsylvania State University, University Park,
13. Zhao, S., J.J. Maurer, S. Hubert, J.F. De Villena, P.F. McDermott, J.
Penn.). Meng, S. Ayers, L. English, and D.G. White. Antimicrobial susceptibility
and molecular characterization of avian pathogenic Escherichia coli isolates.
Vet. Microbiol. 107:215-24,2005.

11
4
PASTEURELLOSIS, AVIBACTERIOSIS, GALLIBACTERIOSIS, RIEMERELLOSIS, AND PSEUDOTUBERCULOSIS
John R. Glisson, Tirath S. Sandhu, and Charles L. Hofacre

SUMMARY. In avian hosts, certain members of the genera Pasteurella, Avibacterium, Gallibacterium and the species Riemerella
anatipestifer and Yersenia pseudotuberculosis cause septicemic and respiratory disease. Acute diseases caused by these Gram-negative
bacteria are often characterized by high flock mortality and morbidity. Clinical signs of acute disease include ruffled feathers, depression,
increased respiratory rate, cyanosis, emaciation, and diarrhea. Lesions may consist of hemorrhages, swollen liver and spleen, focal necrotic
areas in the liver, airsacculitis, and increased pericardial and peritoneal fluids. With R. anatipestifer infection, common gross lesions in ducks
are fibrinous pericarditis, hepatitis and airsacculitis. Chronic disease signs produced by Pasteurella sp., R. anatipestifer and Y.
pseudotuberculosis can include ocular and nasal discharge, swelling of tissues such as joints and wattles, and stunted growth.

Pasteurella
This genus includes P. multocida, the cause of fowl cholera. Two similar organisms, Avibacterium gallinarum (formerly Pasteurella
gallinarum) and Gallibacterium anatis biovar haemolytica (formerly Pasteurella haemolytica) are include in this chapter because they can be
isolated from the respiratory tract and have been associated with respiratory disease in poultry (1,4). Five serogroups of P. multocida (A, B,
D, E and F) have been isolated from avian species, but serogroup A strains are the major cause of fowl cholera. Sixteen somatic serotypes
may occur among strains of P. multocida. However, somatic serotypes 1, 3 and 4 are most commonly isolated.
Agent Identification. Pasteurellas are identified by cell and colony morphology, Gram stain, and reactions in biochemical and other tests. A
mucopolysaccharidase test can be used for presumptive identification of P. multocida serogroups A, D and F. Gel diffusion precipitin tests
are used to determine somatic serotype.
Serologic Detection in the Host. Serologic tests are not commonly used to detect infections by Pasteurella sp. in poultry.

Riemerella anatipestifer
Riemerella anatipestifer infection occurs in ducklings usually at 1-8 wk of age. Primary isolations of R. anatipestifer are made on blood or
trypticase soy agar incubated at 37C in a candle jar or CO2 incubator. At least twenty one serotypes have been identified using agglutination
tests.
Agent Identification. Riemerella anatipestifer is identified by cell and colony morphology, Gram stain, and biochemical characteristics.
Fluorescent-antibody technique can be used to detect and identify R. anatipestifer in tissues or exudates from infected birds.
Serologic Detection in the Host. Serum antibodies against R. anatipestifer can be detected in poultry by ELISA.

Yersenia pseudotuberculosis
Pseudotuberculosis, caused by infection with Y pseudo tuberculosis, is infrequently reported in poultry. Selective media are used for
isolation of the bacterium from feces. There are six serotypes (I-VI) based upon heat-stable antigens using agglutination and agglutination­
adsorption tests. Among strains isolated from birds, serotype I is the most common. Serotypes V and VI have not been reported in birds.
Agent Identification. Yersenia pseudotuberculosis is identified by cell and colony morphology, Gram stain, and reactions in biochemical
and other tests. The bacterium is motile at 25 C.
Serologic Detection in the Host. Serologic tests are not used to detect Y. pseudotuberculosis infections in poultry.

Pasteurella multocida
INTRODUCTION
stage, particularly when the infection is caused by organisms of low
The disease caused by infection with Pasteurella multocida is virulence. Finding dead birds may be the first sign of fowl cholera.
usually called fowl cholera; however, the term avian cholera is Other typical signs are depression, diarrhea, ruffled feathers,
frequently used when the disease occurs in wild birds. The name increased respiratory rate, and cyanosis. Commonly observed
Pasteurella septica infection was sometimes used in older European lesions in birds dying of acute fowl cholera include passive
literature. Fowl cholera is a common, widely distributed disease of hyperemia, hemorrhages, swollen liver, focal necrotic areas in the
major economic importance in the United States. The disease liver and spleen, and increased pericardial and peritoneal fluids. In
affects all species of birds. Among commercially raised birds, general, the signs of chronic fowl cholera include swelling of
turkeys and Japanese quail are particularly susceptible. Outbreaks affected tissues, such as joints and sternal bursae, and exudate from
in wild waterfowl are common and frequently cause high mortality. conjunctivae and turbinates. The focal lesions are generally
Fowl cholera occurs as a primary disease that does not require characterized by fibrinosuppurative exudate and various degrees of
predisposing factors, although predisposing factors may increase necrosis and fibroplasia.
severity of outbreaks. Subclinical infections apparently do not exist
in normal flocks, but normal-appearing birds that have survived SAMPLE COLLECTION
outbreaks of the disease frequently remain infected and may serve
as carriers. Pasteurella multocida can be isolated readily from the liver, bone
marrow, and heart blood of birds that die of acute fowl cholera and
CLINICAL DISEASE usually from localized lesions of chronic cholera (5). Bone marrow
and brain are recommended when specimens are not fresh or when
Fowl cholera may affect birds of any age, but it rarely occurs in contamination of tissues seems likely (25). To obtain specimens for
commercially raised poultry of less than 8 wk of age. The infection microbiologic examination, the surface of the tissue is seared with a
often occurs as an acute septicemic disease with high morbidity and heated spatula, and a sterile cotton swab or wire loop is inserted
mortality (5). Chronic fowl cholera may follow the septicemic through the seared surface. The specimen is transferred to an agar
12
Chapter 4 Pasteurellosis, Avibacteriosis, Gallibacteriosis, Riemerelloisis, And Pseudotuberculosis

medium and incubated at 37 C. Pasteurella multocida grows strains of P. multocida. A gel-diffusion precipitin test (GDPT) is
aerobically and anaerobically. used for serotyping based on differences in somatic antigens
Cultures of P. multocida are moderately stable, generally surviving (somatic serotyping) (9). Sixteen serotypes (1-16) have been
storage or transportation if maintained in a humid environment, reported (2); strains representing each of these 16 serotypes have
such as on agar slants in screw-capped tubes. Stab cultures in agar been isolated from avian hosts. Frequently, antigens from a single
medium in screw-capped tubes generally survive for weeks. Long­ strain react with more than one type of serum, resulting in serotypes
term storage is best accomplished using lyophilization. such as 3,4 and 3, 4, 12. An indirect (passive) hemagglutination
test is used for serogrouping based on differences in capsular
PREFERRED CULTURE MEDIA AND SUBSTRATES antigens (capsular serogrouping) (3). Five capsular serogroups (A,
B, D, E, and F) have been reported (20). Serogroup A, D, and F
Dextrose starch agar (DSA), blood agar, or trypticase soy agar strains produce capsules containing mucopolysaccharides, and
(Becton Dickinson Microbiology Systems, Sparks, Md.) are presumptive identification of these serogroups can be made using
recommended for primary isolation of P. multocida. The likelihood specific mucopolysaccharidases in a disk-diffusion test (16). Except
of isolation may be improved by supplementing these media with for serogroup E, strains representing all serogroups have been
5% heat-inactivated serum. The organisms grow readily in tryptose isolated from avian hosts.
or trypticase soy broth. Antisera used in determining somatic serotypes are prepared in
chickens (9, 17). Such typing antisera are available from the
AGENT IDENTIFICATION National Veterinary Services Laboratories, Animal and Plant Health
Inspection Service (APHIS), United States Department of
Colony Morphology Agriculture (USDA), Ames, Iowa. Laboratories wishing to obtain
On DSA, 24-hr colonies are circular, 1-3 mm in diameter, smooth, such sera should contact the APHIS-USDA Veterinarian-in-Charge
transparent, glistening, and butyrous. Colonies on blood agar are in their state.
similar to those on DSA but are grayish and less translucent. Antigens for the GDPT are prepared from 18-to-24-hr growth of
Observation of 24-hr colonies on DSA or other translucent agar heavily seeded DSA in petri dishes. The cells from one dish are
using a dissection microscope and obliquely transmitted lighting (5) suspended in 1.0 ml of 0.85% NaCl, 0.02M phosphate, and 0.3%
provides information on whether or not cells are capsulated. formalin solution, pH 7.0. The suspension is heated in a water bath
Colonies that are iridescent contain capsulated cells, whereas at 100 C for 1 hr and centrifuged at 4000 x g for 30 min. The
colonies that are noniridescent (blue or blue-gray) contain supernatant fluid is used for antigen.
uncapsulated cells. Pasteurella multocida produces a distinctive The agar gel consists of 0.9% Noble hgar (Difco, Detroit, Mich.)
odor when grown on agar media. in 8.5% NaCl solution. Five milliliters of warm (46 C) melted agar
is flooded onto a 25 x 75-mm microscope slide; wells, 4 mm in
Cell Morphology diameter and 6 mm from center to center, are cut. Antigen is placed
Pasteurella multocida cells are typically rods of 0.2-0.4 χ 0.6-2.5 in a well and antisera are placed in opposing wells. The slide is
pm occurring singly or occasionally in pairs or short chains. When placed in a petri dish to prevent drying, and the results are recorded
grown under unfavorable conditions or after repeated subculture, after 24 hr at 37 C.
cells tend to become pleomorphic. Cells in tissues or exudate Antisera used in the indirect hemagglutination test for capsule
usually show bipolar staining with Giemsa or Wright’s stain. serogrouping are prepared in Pasteurellα-free rabbits (18, 20).
Capsules can be demonstrated using an indirect india-ink method of Preparation of high-titered sera requires repeated intravenous
staining. A loopful of dilute bacterial specimen is mixed with a inoculations with formalin-killed capsulated organisms. Currently,
loopful of india ink on a microscope slide, a coverslip is applied, antisera for capsule serogrouping are not available from commercial
and the preparation is examined microscopically at a high or government laboratories.
magnification. Capsules appear as clear halos around the bacterial
cells when examined in this manner. SEROLOGIC DETECTION IN THE HOST

Biochemical and Other Tests Commercially available enzyme-linked immunosorbent assay


Evaluation of reactions in differential media is generally made (ELISA) kits may be used to detect a serological response to P.
after 2 days of incubation at 37 C and again after 5 days of multocida infection in poultry (IDEXX, Westbrook, Maine and
incubation at room temperature (21 C), in case of delayed reactions. Synbiotics, Gaithersbury, MD). ELISAs are used primarily to
The carbohydrate broth media used for identification (Table 4.1) is measure the serologic response following the use of inactivated P.
phenol red broth base containing 1% of the carbohydrate substrate. multocida vaccines in poultry. Serologic tests are rarely used for
For detection of hydrogen sulfide (H2S), a filter paper strip diagnosis of fowl cholera.
impregnated with lead acetate is suspended above modified H2S
broth (10) during incubation. The presence of indole is indicated by DIFFERENTIATION FROM CLOSELY RELATED AGENTS
development of a dark red color when a small amount of modified
Kovac’s indole test reagent is added to a 24-hr culture consisting of Differentiation of fowl cholera from other diseases caused by
2% tryptose in 0.85% NaCl solution (7). Oxidase production is Pasteurella, Avibacterium, Gallibacterium, Riemerella
determined using Kovac’s oxidase-test reagent and indirect filter anatipestifer, and Yersinia pseudotuberculosis depends primarily
paper procedure (12). upon isolation and differentiation of causative organisms.
Fructose, galactose, glucose, and sucrose are fermented without Differential physiologic reactions of these organisms are indicated
gas production. Inositol, inulin, maltose, salicin, and rhamnose are in Table 4.1. Several other diseases that are not caused by closely
not fermented. Indole and oxidase are almost always produced. related agents, such as infectious coryza, fowl typhoid, fowl plague,
Neither hemolysis of blood nor growth on MacConkey’s agar occur. duck plague, and infectious synovitis may have histories, signs, or
Differential characteristics are listed in Table 4.1. lesions similar to those of fowl cholera. Differentiation of fowl
cholera from these diseases also depends upon isolation and
Serologic Identification identification of causative organisms.
Serologic tests are rarely used for identification of P. multocida.
However, these are commonly used for antigenic characterization of
13
John R. Glisson, Tirath S. Sandhu, and Charles L. Hofacre

Table 4.1. Differential characteristics of Pasteurella, Avibacterium, Gallibacterium, Riemerella, and Yersinia.

Test P. multocida A. gallinarum G.anatis biovar R. anatipestifer Y. pseudotuberculosis


haemolytica
Arabinose -U - - - +
Dextrin -u + -U +
Dulcitol -u - - -
Fructose + + + +
Galactose + + + +
Glucose + + + +
Glycerol +u + + +
Inositol - V +u -
Inulin - - - -
Lactose u - +u -
Maltose + -u +
Mannitol + - + +
Mannose + + + +
Melibiose +
Raffinose -u + V -
Rhamnose -u - - +
Salicin - - - +U
Sorbitol +u V + -
Sucrose + + + V
Trehalose -u +u +u . +
Xylose +u - +u +
Gelatin - - - +U +
Hemolysis - - + - -
H2S +u +u +u - V
Indole + - - -
Litmus milk - si. acid U si. alk si. Alk
MacConkey - V - +U
Motility - - - +(25 C)
Methyl red - - - +
Nitrate reduced + + + - +
Oxidase + + + + +
Urease - - - V +
A+ = reaction, - = no reaction, U = usual reaction, V = variable reaction, si. Acid = slightly increased acidity, si. alk = slightly increased
alkalinity, (25 C) =- incubated at 25 C.

Avibacterium gallinarum
INTRODUCTION
Swollen and inflamed wattles resulting from infection with only A.
Infection with A. gallinarum often occurs in the respiratoiy tracts gallinarum have been reported in chickens (13).
of chickens and turkeys (6, 13), but disease is generally manifested
only when this infection occurs with other respiratoiy tract SAMPLE COLLECTION
infections. Attempts to isolate A.gallinarum from normal flocks
have not been successful. The infection is widely distributed. It has Avibacterium gallinarum often can be isolated from the sinuses,
been reported in the United States, Australia, Japan, Nigeria, Iran, nasal cleft, trachea, air sacs, and lungs of birds exhibiting
and Israel. Generally it is not considered to be of economic respiratory disease. The organism is less frequently isolated from
significance. livers, heart blood, and joints. Avibacterium gallinarum grows
aerobically and anaerobically. Specimens are collected using the
CLINICAL DISEASE same methods as described for P. multocida.

Disease manifestations in which A. gallinarum infection occurs PREFERRED CULTURE MEDIA AND SUBSTRATES
usually affect the respiratory tract and have a chronic course.
The same media are used as described for P. multocida.
14
Chapter 4 Pasteurella, Avibacteriosis, Gallibacteriosis, Riemerellosis, and Pseudotuberculosis

AGENT IDENTIFICATION gallinarum using serologic tests, but specific serotypes have not
been defined. Serologic testing has indicated that minor antigens of
Colony and Cell Morphology and Biochemistry some strains of A. gallinarum are shared with P. multocida.
On DSA or blood agar at 37 C, 24-hr colonies are 1-2 mm in
diameter, smooth, entire, low convex, and transparent. Colonies on SEROLOGIC DETECTION IN THE HOST
DSA are iridescent when observed with obliquely transmitted light,
often with concentric rings. Serologic tests are not used to detect A. gallinarum infections in
Avibacterium gallinarum cells are similar to P. multocida; that is, poultry.
Gram-negative short rods that occur as single organisms and short
chains, stain bipolarly, and are capsulated. They become DIFFERENTIATION FROM CLOSELY RELATED AGENTS
pleomorphic with repeated subculturing and growth under less than
optimum conditions. Differentiation of A. gallinarum infections from those caused by
Glucose, sucrose, and maltose are fermented without gas Pasteurella and Y. pseudotuberculosis is accomplished by
production. Lactose is not fermented, and indole is not produced. determination and comparison of selected physiologic
Neither hemolysis of blood nor growth on MacConkey’s agar occur. characteristics (Table 4.1). Other infections that may cause similar
Differential characteristics are listed in Table 4.1. signs or lesions include infectious coryza, mycoplasmosis, and
bordetellosis. Differentiation depends upon isolation and
Serologic Identification identification or differentiation of the agents.
Serologic tests are rarely used for identification of A. gallinarum.
Antigenic differences have been demonstrated among strains of A.

Gallibacterium anatis biovar haemolytica


INTRODUCTION AGENT IDENTIFICATION

Gallibacterium anatis biovar haemolytica is generally considered Colony Morphology


to be a secondary pathogen that requires some predisposing factor On blood agar at 37 C, 24-hr colonies are generally 0.5 mm in
before producing disease. However, too little information is diameter, smooth, entire, low convex, and translucent. A wide zone
available to clearly establish the role of this organism in avian of β-hemolysis is produced on blood agar. On DSA, the colonies
disease production. In normal-appearing flocks, the reported are iridescent when observed with obliquely transmitted light and
incidence of infection is quite high in chickens but low in turkeys often exhibit concentric rings.
and geese. Infections in avian hosts are widely distributed, as
indicated by isolations in the United States, England, Germany, Cell Morphology
France, Israel, Syria, Nigeria, Taiwan, and Australia. This disease G. anatis biovar haemolytica cells are Gram-negative, nonmotile,
is not considered economically important. non-spore-forming, pleomorphic rods (0.5 x 1.6 pm) that often
exhibit bipolar staining. They generally occur singly but
CLINICAL DISEASE occasionally form short chains.

Disease manifestations related to G. anatis biovar haemolytica Biochemical and Other Tests
infections most frequently involve the respiratory tract. Septicemic Glucose, sucrose, mannose, glycerol, and fructose are fermented
manifestations with petechial hemorrhages in the viscera and areas without gas production within 24 hr of incubation at 37 C. Lactose
of liver necrosis have been reported. Salpingitis and peritonitis may is usually fermented after 3 days. Arabinose, dulcitol, rhamnose,
occur in laying hens. inulin, and salicin are not fermented. Indole is not produced.
Differential characteristics are indicated in Table 4.1.
SAMPLE COLLECTION
Serologic Identification
G. anatis biovar haemolytica usually can be isolated from the Serologic tests are not used for identification of G. anatis biovar
trachea, lungs, liver, or oviduct of an infected bird. haemolytica. Little effort has been made to serotype strains isolated
from avian hosts, and no avian serotypes have been established.

SEROLOGIC DETECTION IN THE HOST

Serologic tests are not used to detect G. anatis biovar haemolytica


infection in poultry.

Riemerella anatipestifer
INTRODUCTION the disease can cause mortality as high as 75% in ducks, especially
at farms where infection persists because hatches are frequently
Riemerella anatipestifer infection, also known as infectious moved from one pen to another to create space for the next hatch.
serositis, duck septicemia, new duck disease, or anatipestifer Adverse environmental conditions and concomitant disease often
syndrome is a septicemic disease of ducks, geese, turkeys, and predispose flocks to epomitics of R. anatipestifer infection. The
various other birds caused by R. anatipestifer. The disease is disease is not of public health importance. In the United States,
prevalent worldwide and causes significant economic loss due to federal or state notification is not required.
high mortality, weight loss, and condemnations. The acute form of

15
John R. Glisson, Tirath S. Sandhu, and Charles L. Hofacre

CLINICAL DISEASE long and 0.1-0.4 pm wide and may show bipolar staining. A
capsule has been shown with the india ink method of staining.
The acute form of the disease usually occurs in ducklings 1-8 wk Riemerella anatipestifer does not ferment sugars in routine media
of age. Chronic infections may occur in older birds. High mortality (Table 4.1), but has been reported to produce acid in glucose,
has been reported in turkeys 6-15 wk of age (23). Riemerella mannose, maltose and dextrin when grown in buffered single
anatipestifer infections have also been reported in swans, pheasants, substrate medium (11). The organism liquefies gelatin and
guinea fowl, partridges, quail, and chickens. Clinical signs of the produces a slight alkaline reaction in litmus milk. Some strains
disease include ocular and nasal discharge, sneezing, greenish produce urease and arginine dihydrolase. No growth occurs on
diarrhea, tremors of the head, neck and legs, ataxia, and coma. The MacConkey’s agar and no hemolysis takes place on blood agar.
common gross lesions are fibrinous pericarditis, perihepatitis, The organism produces oxidase, catalase, and phosphatase, but is
airsacculitis, and meningitis. In females, the oviduct is filled with indole negative.
caseous yellowish white exudate. Chronic and localized infections Riemerella anatipestifer is identified by enzymatic activity
result in synovitis/arthritis and dermatitis. Infections originate from (Apizyme - API laboratory Products Ltd., St-Laurent, Quebec,
exposure via the respiratory tract or through abrasions or cuts in the Canada). It is positive for leucine-, valine-, and cystine-arylmidases,
skin. phosphoamidase, α-glucosidase, and negative for a- and β-
galactosidases, β-glucuronidase, β-glucosidase, α-mannosidase, β-
SAMPLE COLLECTION glucosaminidase, ornithine and lysine decorboxylases (22).

Riemerella anatipestifer can readily be isolated from heart blood, Serologic Identification
brain, pericardial exudate, air sacs, lungs, oviduct, and liver. Using agglutination tests, 21 serotypes have been reported (15,
Isolations should be attempted from infraorbital sinuses and trachea 21). Antisera are made in rabbits. Antigen for rabbit inoculation is
to detect carriers or inapparent infections. Specimens should be made by harvesting 24 to 48-hr growth from an agar plate in 0.85%
obtained by a wire loop or sterile swab through a seared surface. In NaCl solution containing 0.3% formalin. The inactivated cells are
chronic or localized infections, R. anatipestifer can be isolated from washed twice by centrifugation in the above solution and adjusted
the exudate. The specimen is streaked directly onto the surface of to an optical density of 0.2 at 525 nm in a spectrophotometer.
agar media, which are then incubated at 37 C in a candle jar for 24- Young rabbits are immunized by inoculating through the marginal
72 hr. Incubation under increased CO2 and humidity in a candle jar ear vein successive doses of 0.05, 0.1, 0.2, 0.5, 1.0, 1.0, 1.5, and 2.0
or CO2 incubator enhances the growth for primary isolation. If the ml of standardized cell suspension, at 3- to 4-day intervals. Rabbits
cultures cannot be made within a reasonable time, the specimens or are bled for serum collection when maximum titers are obtained.
tissues should be kept at 4 C or frozen for shipping or later Agglutination titers are low, usually on the order of 1:50 to 1:400.
processing. The plate agglutination test is done by mixing a drop of undiluted
antiserum with one or two colonies from a 24-hr growth.
PREFERRED CULTURE MEDIA AND SUBSTRATES Agglutination within a few seconds indicates a homologous
serotype reaction. The tube agglutination test is performed by
Although R. anatipestifer grows readily on routine media, growth is mixing an equal amount of formalinized cell suspension (adjusted
enhanced by using enriched media such as blood agar or trypticase to 0.2 optical density at 525 nm) with each dilution of serum and
soy agar containing 0.05% yeast extract. Tryptose broth or incubating at 37 C. Agglutination is recorded after 24 and 48
trypticase soy broth is used for broth cultures. Addition of hr.The fluorescent antibody technique may be used to detect and
gentamicin (5 mg/liter) to enriched solid medium has proven identify R. anatipestifer cells directly in tissues or exudate from
helpful for isolation of R. anatipestifer from sinuses, skin, and infected birds.
contaminated specimens from the processing plant, even though
growth of other organisms is not completely inhibited. SEROLOGIC DETECTION IN THE HOST

AGENT IDENTIFICATION Enzyme-linked immunosorbent assay (ELISA) is commonly used


for early detection of R. anatipestifer infections (8). Cell lysate is
Colony and Cell Morphology and Biochemistry used as an antigen in ELISA. It is not serotype specific and will
Riemerella anatipestifer is identified on the basis of growth, show positive reactions with antisera against heterologous
morphologic and biochemical characteristics. After 24 hr of growth serotypes, but ELISA is more sensitive than agglutination test. The
on blood agar, colonies are 1-2 mm in diameter, convex, entire, GDPT is unreliable because of an apparent discrepancy in duck
transparent, glistening, and butyrous. Some strains may produce immunoglobulins, which are almost nonfunctional in precipitin
mucoid growth on solid media. On trypticase soy agar, colonies reactions (24).
appear bluish and are iridescent when observed with obliquely
transmitted light. Some strains have been observed to produce off- DIFFERENTIATION FROM CLOSELY RELATED AGENTS
white growth, which may change to grayish brown after 3-5 days.
Growth in broth produces slight turbidity. Diagnosis should be based on isolation and identification of the
Most R. anatipestifer strains become nonviable after 3^4 days on a causative organism, because similar gross lesions are produced by
solid medium at room temperature or 37 C. In broth the organisms P. multocida, Escherichia coli, and fecal streptococcal infections.
may survive for 2-3 wk under refrigeration. Cultures can be stored Ducklings infected with Salmonella may sometime show nervous
as freeze-dried for longer periods. Riemerella anatipestifer cells are signs similar to those of R. anatipestifer infection. Chlamydiosis
Gram-negative, nonmotile, non-spore-forming rods that occur should also be considered in differential diagnosis.
singly, in pairs, or occasionally in chains. The cells are 1-5 pm

16
Chapter 4 Pasteurella, Avibacteriosis, Gallibacteriosis, Riemerellosis, and Pseudotuberculosis

Yersinia pseudotuberculosis
INTRODUCTION Cell Morphology
Yersinia pseudotuberculosis cells are Gram-negative rods of 0.5 x
The disease caused by infection with Y.pseudotuberculosis is 0.8-5.0 pm. Coccoid forms usually stain bipolar. Neither spores
pseudotuberculosis (19). It can affect domesticated, feral, or caged nor capsules are formed. Peritrichous flagella usually develop
birds and a variety of mammals, including humans. Although this between 20 C and 30 C. Motility is best shown in semisolid media
disease is infrequently reported in the United States and is not at 25 C.
considered economically important, it has caused death losses as
high as 80% in turkey flocks. Biochemical and Other Tests
Maltose, trehalose, and usually sucrose are fermented, and acid but
CLINICAL DISEASE not gas is produced. Production of H2S is variable. Urease and
catalase are produced, but not indole and oxidase. No hemolysis
Pseudotuberculosis usually occurs as a septicemia or bacteremia of occurs on blood agar. Differential characteristics are listed in Table
short duration, followed by chronic focal infections. In very acute 4.1.
cases, birds may die before other signs of disease are observed, Serologic Identification
although signs usually are present for 2 wk or more. Affected birds Serologic testing is not used for identifying strains of
exhibit weakness, ruffled feathers, diarrhea, and breathing Y. pseudotuberculosis but is used to characterize strains (19). There
difficulties. Emaciation and paralysis occur occasionally. Swollen are six serotypes (I-VI), four of which have two subtypes each (A
livers and spleens and enteritis are common lesions in acute cases. and B). These serotypes and subtypes are based on 15 heat-stable
Necrotic foci in visceral organs and muscles and enteritis are somatic antigens (1-15), which are demonstrated by agglutination
common lesions in chronic cases. Osteomyelitis occurs in some and agglutination-adsorption tests. Additional serotypes VH and
affected turkey flocks. Vin and a subtype C of serotype Π have been proposed. Among
strains from birds, serotype I is most common, followed by
SAMPLE COLLECTION serotypes Π and IV. Serotypes V and VI have not been reported in
birds, and type ΠΙ is rare. Five heat-labile flagellar antigens (a, b, c,
Yersinia pseudotuberculosis can usually be isolated from blood, d, e) are recognized but are rarely used for serologic
liver, spleen, or lung using blood or trypticase soy agar and characterization.
incubation at 37 C (19). The method of Paterson and Cook (14) is
recommended for isolation of Y pseudotuberculosis from feces. To SEROLOGIC DETECTION IN THE HOST
use this method, inoculate the surface of Paterson and Cook agar
medium with a 10% suspension of feces in phosphate buffer (pH Serologic techniques are not used to detect Y. pseudotuberculosis
7.6). Recovery of Y. pseudotuberculosis is enhanced by storing the infection in poultry.
fecal suspension at 3-4 C for 7 days or longer before inoculating
agar. The organism grows both aerobically and anaerobically. DIFFERENTIATION FROM CLOSELY RELATED AGENTS

PREFERRED CULTURE MEDIA AND SUBSTRATES Differentiation of Y. pseudotuberculosis from avian Pasteurella is
based on differences in biochemical characteristics, as indicated in
Trypticase soy agar and blood agar are effective for primary Table 4.1. Diseases with similar lesions or signs, such as
isolation. Differential media are as described for P. multocida. For tuberculosis, E. coli infections, and salmonellosis, are differentiated
isolation from feces, Paterson and Cook medium is recommended from pseudotuberculosis through isolation and identification or
(14) (Table 4.2). biochemical differentiation of causative agents.

AGENT IDENTIFICATION REFERENCES

Colony Morphology 1. Blackall, P.J. H. Christensen, T. Beckenham, L.L. Blackall, and M


After incubation at 37 C for 24 hr on trypticase soy agar, colonies Bisgaard Reclassification of Pasteurella gallinarum, [Haemophilus]
are smooth, round, entire, grayish yellow, butyrous, and 0.5-1 mm paragallinarum, Pasteurella avium and Pasteurella volantium as
in diameter. Older colonies are 2-3 mm in diameter, raised, flat, Avibacterium gallinarum gen. nov., comb, nov., Avibacterium
paragallinarum comb, nov., Avibacterium avium comb, nov. and
dry, and irregular with rough edges.
Avibacterium volantium comb. nov. Int. J. Evol. Microbiol. 55: 353-362,
2005.
Table 4.2. Medium of Paterson and Cook. 2. Brogden, K. A., K. R. Rhoades, and K. L. Heddleston. A new serotype
Constituents Amount (ml) of Pasteurella multocida associated with fowl cholera. Avian Dis. 22:185-
Trypsinized meat agar 190. 1978.
200.0 3. Carter, G. R. Studies on Pasteurella multocida. I. A hemagglutination
Peptic digest of sheep blood test for the identification of serological types. Am. J. Vet. Res. 16:481^484.
10.0
Novobiocin (0.2%) 1955.
0.5 4. Christensen, Η., M. Bisgaard, A.M Bojesen, R. Mutters, and J.E. Olsen.
Erythromycin (0.2%) Genetic relationships among avian isolates classified as Pasteurella
0.5 haemolytica, Actinobacillus salpingitidis or Pasteurella anatis with proposal
Mycostatin (50,000 units/ml) of Gallibacterium anatis gen. nov., comb. nov. and description of additional
0.8
Crystal violet (0.1%) genomospecies with Gallibacterium gen. nov. Int. J. Syst. Evol. Microbiol.
0.5 53: 275-287,2003.
5. Glisson, J. R., C. L. Hofacre and J. P. Christensen. Fowl cholera. In:
Diseases of poultry, 11th ed Y. M. Saif, H. J. Barnes, J. R. Glisson, A M
Fadly, L. R. McDougald, and D. E. Swayne, eds. Iowa State University
Press, A Blackwell Publishing Company, Ames, Iowa. pp. 658-676. 2003.

17
John R Glisson, Tirath S. Sandhu, and Charles L. Hofacre

6. Hall, W. J., K. L. Heddleston, D. H. Legenhausen, and R. W. Hughes. 19. Rimler, R B., and J. R. Glisson. Pseudotuberculosis. In: Diseases of
Studies on pasteurellosis. I. A new species of Pasteurella encountered in poultry, 10th ed. B. W. Calnek, H. J. Bames, C. W. Beard, L. R.
chronic fowl cholera. Am. J. Vet. Res. 16:598-604. 1955. McDougald, and Y. M Saif, eds. Iowa State University Press, Ames, Iowa,
7. Hans, G. H., and S. Gabriel. Modified stable Kovac’s reagent for pp. 314-317. 1997.
detection of indole. Am. J. Clin. Pathol. 26:1373-1375. 1956. 20. Rimler, R. B., and K. R. Rhoades. Serogroup F, a new capsule
8. Hatfield, R. Μ., B. A Morris, and R. R. Henry. Development of an serogroup of Pasteurella multocida. J. Clin. Microbiol. 25:615-618. 1987.
enzyme-linked immunosorbent assay for the detection of humoral antibody 21. Sandhu, T. S. Riemerella anatipestifer infection. In: Diseases of
to Pasteurella anatipestifer. Avian Pathol. 16:123-140. 1987. poultry, 11th ed. Y. M Saif, H. J. Bames, J. R. Glisson, A. M Fadly, L. R.
9. Heddleston, K. L., J. E. Gallagher, and P. A. Rebers. Fowl cholera: gel McDougald, and D. E. Swayne, eds. Iowa State University Press, A
difiusion precipitin test for serotyping Pasteurella multocida from avian Blackwell Publishing Company Ames, Iowa. pp. 676-682. 2003.
species. Avian Dis. 16:925-936. 1972. 22. Segers, P., W. Mannheim, M Vancanneyt, K. DeBrandt, K. H. Hinz,
10. Heddleston, K. L., T. Goodson, L. Liebovitz, and C. I. Angstrom. K. Kersters, and P. Vandamme. Riemerella anatipestifer gen. nov., comb,
Serological and biochemical characteristics of Pasteurella multocida from nov., the causative agent of septicemia anserum exudativa, and its
free-flying birds and poultry. Avian Dis. 16:925-936. 1972. phylogenetic affiliation within the Flavobacterium-Cytophaga rRNA
11. Hinz, K. Η., M Ryll, and B. Kohler. Detection of acid production from homology group. Int. J. Syst. Bacteriol. 43:768-776. 1993.
carbohydrates by Rimerella anatipestifer and related organisms using the 23. Smith, J. M, D. D. Frame, G. Cooper, A. A. Bickford, G. Y.
buffer^ single substrate test. Vet. Microbiol. 60:277-284. 1998. Ghazikhanian, and B. J. Kelly. Pasteurella anatipestifer infection in
12. MacFaddin, J. F. Oxidase test. In: Biochemical tests for identification commercial meat-type turkeys in California. Avian Dis. 31:913-917. 1987.
of medical bacteria, 2nd ed. Williams and Wilkins, Baltimore, Md. pp. 24. Toth, T. E., and N. L. Norcross. Precipitating and agglutinating
249-253. 1980. activity in duck anti-soluble protein immune sera. Avian Dis. 25:338-352.
13. Mushin, R., R. Bock, and M Abrams. Studies on Pasteurella 1981.
gallinarum. Avian Pathol. 6:415-423. 1977. 25. Waltman, W.D. and A.M Home. Characteristics of fowl cholera
14. Paterson, J. S. and R Cook. A method for the recovery of Pasteurella diagnosed in Georgia 1989-1991. Avian Dis. 37:616-621, 1993.
pseudotuberculosis from feces. J. Pathol. Bacteriol. 85:241-242. 1963.
15. Pathanasophon, P., P. Phuektes, T. Tanticharoenyos, W. Narongsak, and
T. Sawada. A potential new serotype of Riemerella anatipestifer isolated
from ducks in Thailand. Avian Pathol. 31:267-270. 2002.
16. Rimler, R. B. Presumptive identification of Pasteurella multocida
serogroups A, D, and F by capsule depolymerisation with
mucopolysaccharidases. Vet. Rec. 134:191-192. 1994.
17. Rimler, R B., R. D. Angus, and M Phillips. Evaluation of the
specificity of Pasteurella multocida somatic antigen-typing antisera
prepared in chickens using ribosome-lipopolysaccharide complexes as
inocula. Am. J. Vet. Res. 50:29-31. 1989.
18. Rimler, R. B., and K. A. Brogden. Pasteurella multocida isolated from
rabbits and swine: serologic types and toxin production. Am J. Vet. Res.
47:730-737. 1986.

18
5
BORDETELLOSIS
Mark W. Jackwood

SUMMARY. Bordetellosis is an acute highly contagious disease of the upper respiratory tract of young turkeys (4-8 wk of age). The
disease is caused by a gram-negative nonfermentative bacteria, Bordetella avium. Members of the genus Bordetella are well known for their
ability to colonize and damage ciliated epithelial surfaces in the respiratory tract and B. avium is no exception. Bordetellosis is most severe
when it occurs in conjunction with other respiratory infections, especially Newcastle disease, or when turkeys are immunosuppressed.
Mortality can be high when poor management (inadequate ventilation, dust, chilling, or filthy conditions) is a complicating factor and there is
an associated colibacillosis. Bordetella avium is an opportunistic pathogen in chickens.
Agent Identification. Bordetellosis can be diagnosed using a combination of clinical signs (snick or cough with a catarrhal nasal discharge)
and bacterial culture.
Serologic Detection in the Host. Enzyme-linked immunosorbent assays and microagglutination tests are accurate and sensitive for detecting
antibodies in turkeys. Test results can be an aid in diagnosis as well as monitoring vaccination.

INTRODUCTION sp. Cultures should be taken early during the infection while cilia
are heavily colonized with B. avium. If cultures are taken late in the
In the 1970s an acute upper respiratory disease emerged as a major infection the most predominant bacteria isolated will likely be
disease problem in young turkeys. The disease was more severe the Escherichia coli.
earlier posthatch turkeys become infected. Initially some confusion
occurred as to the identification of the causative agent but a PREFERRED CULTURE MEDIA AND SUBSTRATES
definitive study established the agent as a new species in the genus
Bordetella and it was given the name Bordetella avium (7). In the Bordetella avium grows on a variety of enriched agar-based media
early years following its recognition, the disease and associated but the bacteria is usually isolated from clinical material using
colibacillosis produced high mortality but in recent years the MacConkey agar because this agar quickly demonstrates the
disease or the susceptibility of turkeys to the disease has changed bacteria’s nonfermenting characteristic and differentiates it from E.
and the number and severity of reported outbreaks has lessened. coli, another common bacteria found in the trachea of turkeys with
Some of the possible explanations for the change in the character of respiratory disease. Initial isolation of B. avium is best done on
the disease include the presence of maternal antibody, as it is well MacConkey agar increased to 2.5% agar content to slow the spread
established that passive immunity is highly protective (9), or that of faster growing organisms (6). Growth of B. avium in liquid
strains of B. avium have emerged that are not as virulent. The media requires that the cultures be aerated by agitation or other
significance of bordetellosis is not as great now as when it was first means because it is a strict aerobe.
recognized but it still is considered to be a major cause of
respiratory disease in young turkeys. AGENT IDENTIFICATION
Bordetella avium has also been isolated from chickens. Bordetella
avium is an opportunistic pathogen in chickens because clinical After 24 hr of incubation on MacConkey agar, colonies of
disease can only be reproduced following initial infection with an B. avium are clear and pinpoint in size. If cultures have been
upper respiratory disease virus vaccine such as infectious bronchitis secured early they are often pure but when cultures are taken late
virus or Newcastle disease virus (4). Recent studies have reported there is often contamination with other bacteria especially E. coli.
B. avium in mallards, wild turkeys, and a Canada goose, (10) and When such contamination occurs it is important to look in the more
antibodies against B. avium have been detected in peafowl (2). diluted part of the streak for typical colonies.
Three colony types of B. avium have been identified. The most
CLINICAL DISEASE typical type is that described above with colonies being small,
compact, translucent, and pearl-like with entire edges and glistening
Bordetellosis is characterized by an abrupt onset of a snick surfaces. These colonies will be 0.2-1 mm in diameter after 24 hr
(sneezing) in young turkeys. Other signs include watery eyes, and 1-2 mm after 48 hr. Many isolates develop a raised brown-
submaxillary edema, and a clear nasal discharge that can usually be tinged center when grown to >48 hr on MacConkey’s agar. A
expressed by placing gentle pressure on the nares. Mouth breathing, smaller percentage of isolates dissociate into a rough colony type
altered vocalization, dyspnea, huddling, and decreased consumption having a dry appearance and a serrated irregular edge. Rough
of feed and water are also common. Morbidity is high 80-100% in colonies represent a phase-shift to a nonpathogenic form of the
young turkeys, and mortality is low (1% to 10%) except when other bacterium (3).
infections occur simultaneously (colibacillosis) or when Biochemical tests that are used in distinguishing B. avium from
management is poor, then mortality can be high (>50%). The other nonfermenting bacteria include the oxidase test (+), catalase
infection also adversely affects growth rate and toxins associated test (+), urease test (-), nitrate reduction test (-), and the ability to
with the bacteria damage tracheal cartilage beneath colonized alkalinize certain amides and organic salts. A microcupule test kit
ciliated respiratory epithelium. Mortality has been reported from called the Rapid API 20 NE (Non-Fermenter Test, bioMerieux-
suffocation due to excessive mucus production in the trachea and Vitek, Hazelwood, Mo.) can be used to identify B. avium.
when damaged tracheas collapse (12). A very useful correlation has been made between the ability of
strains of B. avium to hemagglutinate guinea pig red blood cells
SAMPLE COLLECTION and pathogenicity (6). This association with pathogenicity is
thought to be related to the ability of pathogenic strains to adhere to
The bacterium is best cultured by swabbing the anterior trachea cilia. The hemagglutination assay is performed by heavily
through a midcervical aseptic opening. Bacterial culture of sinus inoculating solid media (blood agar, brain-heart infusion agar, or
material is usually not productive because of overgrowth by Proteus MacConkey’s agar) with suspected B. avium isolates and incubating
19
Mark W. Jackwood

36-48 hr at 37 C. Bacterial growths are washed from the plate with 2. Drop 0.1 ml of stock antigen into row A, columns 1-8 of each
phosphate-buffered saline (PBS) and diluted to a concentration of plate.
approximately 5 χ 109 cells/ml (0.5 optical density at 600 nm). 3 Drop 0.05 ml of PBS with 0.01% merthiolate into rows B
Equal volumes of bacterial suspension and erythrocyte suspension through H, columns 1-8.
(2% packed-cell volume in PBS) are mixed on a glass slide or plate 4 Make twofold dilutions of B. avium antigen down from row A
and observed for agglutination after gentle rocking. Alternatively, through row H.
50:1 mixture of bacterial suspension and erythrocyte suspension 5. Add 0.05 ml of the appropriate serum dilution (1:10 through
(0.5% packed-cell volume in PBS) are mixed in U-bottom shaped 1:640, in columns 1 through 7, respectively) to rows A through H.
wells of microtitration plates and allow to stand 1 hr at 25 C. A 6. Add 0.05 ml PBS with 0.01% merthiolate to column 8, rows A
reference strain of B. avium and a negative control should be through H, for the antigen control.
included with each test run. 7. Incubate plates at room temperature for 24 hr and read. A
Comparable results can be obtained using erythrocytes fixed in positive test has complete absence of a button, whereas a negative
formaldehyde (F. W. Pierson, Virginia Polytechnic Institute and test has a distinct button in the bottom of the well. For the optimum
State University, pers. comm.). Fixed guinea pig erythrocytes are B. avium antigen dilution, pick the antigen dilution well that has a
prepared as follows: well-defined, visible button in the negative control serum and
1. Collect 5 ml of guinea pig blood with 1 ml of 3% sodium citrate detects antibody (no button) in the positive control serum. It is
and mix well. necessary to do a block titration only once for each lot of B. avium
2. Centrifuge at 500 x g (2000 rpm) for 10 min. Resuspend antigen produced. Mark the appropriate dilution on the stock
erythrocytes in fixative containing 40 ml of PBS and 10 ml of 40% antigen for later reference.
formaldehyde. The final pH of the solution should be The actual microagglutination test is run as follows:
approximately 7.4. 1. Make a 1:5 dilution of each serum sample, including both
3. Mix gently on a rotary shaker at room temperature overnight. positive and negative controls.
4. Centrifuge at 500 x g (2000 rpm) for 10 min and wash pellet 10 2 Drop 0.05 ml PBS with 0.01% merthiolate into each well of a
times with PBS. microtitration plate.
5. Resuspend fixed erythrocytes in PBS at a final concentration of 3. Drop 0.05 ml of diluted serum to the first row of wells. Be sure
2% and store at 4 C. lite fixed cells are stable for at least 6 mos. to include positive and negative control sera with each plate.
4. Make twofold dilutions of the sera from 1:10 to 1:1280 down the
SEROLOGIC DETECTION IN THE HOST plate.
5. Dilute B. avium stock antigen in PBS with 0.01% merthiolate
An in-house enzyme-linked immunosorbent assay (ELISA) can be using the dilution factor from the block titration. The antigen
produced to detect B. avium antibodies in turkeys (8,9). A should be passed through a syringe with a 22 gauge needle to break
commercially available ELISA has been developed and marketed up all antigen clumps.
for B. avium in turkeys (Synbiotics Corporation, San Diego, CA.). 6. Drop 0.05 ml of diluted B. avium antigen into all wells.
The commercially available ELISA kit is excellent and the results 7. Incubate the test plate at room temperature for 24 hr and read.
are very compatible with the in-house ELISA test (9). Any titer 1:20 or greater is considered to be positive.
Prior to the development of the ELISA a microagglutination The microagglutination test is not as user friendly as the ELISA
procedure was performed (5) as outlined below. but the test is sensitive and accurate. It detects immunoglobulin M
Microagglutination antigen is produced as follows: (IgM), which is the first antibody produced following infection. The
1. Inoculate 10 ml of veal infusion broth or brain-heart infusion microagglutination test will not detect maternal antibody, which is
broth with an isolated colony of B. avium and incubate aerobically immunoglobulin G (IgG). High levels of maternal antibody protect
at 37 C for 48 hr in a shaking-water bath or shaking incubator. poults during the first 2 wk of life when the birds are most
2. Inoculate 500 ml of sterile broth in a 1000-ml flask with 10 ml of susceptible to the disease (12). Antibody titers (IgG) to B. avium
broth culture from step 1. Incubate in a shaking-water bath or can be detected by ELISA 2 wk postinfection, with peak titers
incubator at 37 C for 48 hr. When given ideal growth conditions, B. occurring between 3 and 4 wk postinfection (5,8,9).
avium will sometimes produce a filamentous form in broth culture,
which interferes with the microagglutination test (1). This problem DIFFERENTIATION FROM CLOSELY RELATED AGENTS
can be circumvented by growing the bacterium on agar and
harvesting the cells in PBS. Then, continue with step 3. Bordetellosis in poultry should be differentiated from
3. At 48, 49, and 50 hr, add 2.5 ml of a 1% solution of mycoplasmosis, chlamydiosis, cryptosporidiosis, Omithobacterium
neotetrazolium chloride stain. Incubate 4 hr at 37 C following the rhinotracheale, and viral respiratory diseases, particularly
last addition of neotetrazolium chloride. Newcastle disease, influenza, and turkey rhinotracheitis
4. Add 0.2 ml of a 0.1% solution of merthiolate and incubate (pneumovirus infection). The most closely related agents to be
overnight at 37 C. differentiated are Bordetella bronchiseptica and Bordetella hinzii
5. Centrifuge for 30 min at 10,960 x g. Wash and centrifuge (also referred to as B. avium-tike bacteria in the literature). Both can
antigen three times in PBS with 0.01% merthiolate. be isolated from the upper respiratory tract, but neither have shown
6. Centrifuge antigen in a graduated tube at 10,960 x g for 20 min, to cause disease in turkeys. B. avium causes agglutination of guinea
discard the supernatant, and resuspend to make a 1:20 dilution of pig erythrocytes, is urease-negative, and does not grow on minimal
packed cells in PBS with 0.01% merthiolate. This is the stock essential medium agar or in 6.5% NaCl broth. B. bronchiseptica
antigen. Pass the stock antigen through a 22-gauge needle with a may cause hemagglutination, but it produces a positive reaction for
syringe, run a block titration, aliquot the remaining antigen, and urease. B. hinzii does not hemagglutinate but will grow on minimal
freeze the aliquots at -30 C or below. essential medium agar and in 6.5% NaCl broth (6). Molecular tests
Microagglutination antigen standardization (block titration) is as have been used to distinguish between B. avium and B. hinzii.
follows: Ribotyping using PvuH or restriction endonuclease analysis using
1. Use separate microtitration plates for both B. avzwm-positive and Hinfl and DdeV enzymes clearly distinguish between the two
negative sera. First make serial twofold dilutions of sera in test organisms but the REA test was found to be best as a routine
tubes beginning with 1:10 and ending with 1:640. About 0.5 ml of epidemiologic tool (11).
each dilution is needed for one microtitration plate.
20
Chapter 5 Bordetellosis

REFERENCES
8. Lindsey, D. G., P. D. Andrews, G. S. Yarborough, J. K. Skeeles, B.
1. Domingo, D. D., M. W. Jackwood, and T. P. Brown. Filamentous forms Glidewell-Erickson, G. Campbell, and Μ B. Blankford. Evaluation of a
of Bordetella avium', culture conditions and pathogenicity. Avian Dis. commercial ELISA kit for detection and quantification of antibody against
36:706-713. 1992. Bordetella avium. In: Proc. 75th Annual Meeting of the Conference of
2. Hollamby, S., J. G. Sikarskie, and J. Stuht. Survey of peafowl (Pavo Research Workers in Animal Diseases, Chicago, Ill. Abstract #31. Nov.
cristatus) for potential pathogens at three Michigan zoos. J. Zoo Wild. Med. 14—15,1994.
34:375-379. 2003. 9. Neighbor, N. K., J. K. Skeeles, J. N. Beasley, and D. L. Kreider. Use of
3. Jackwood, M W., D. A. Hilt, S. M Mendes, and P. D. Cox. Bordetella an enzyme-linked immunosorbent assay to measure antibody levels in
avium phase-shift markers: characterization of whole cell, cell envelope, and turkey breeder hens, eggs, and progeny following natural infection or
outer membrane proteins. J. Vet. Diag. Invest. 7:402-404. 1995. immunization with a commercial Bordetella avium bacterin. Avian Dis.
4. Jackwood, M. W., S. M McCarter, and T. P. Brown. Bordetella avium'. 35:315-320. 1991.
an opportunistic pathogen in leghorn chickens. Avian Dis. 39:360-367. 10. Raffel, T. R., K. B. Register, S. A. Marks, and L. Tempel. Prevalence
1995. of Bordetella avium infection in selected wild and domesticated birds in the
5. Jackwood, D. J., and Y. M Saif. Development and use of a eastern USA. J. Wildl. Dis. 38:40-46. 2002.
microagglutination test to detect antibodies to Alcaligenes faecalis in 11. Register, K. B., R. E. Sacco, and G. E. Nordholm. Comparison of
turkeys. Avian Dis. 24:685-701. 1980. ribotyping and restriction enzyme analysis for inter- and intraspecies
6. Jackwood, M W.,Y. M. Saif, P. D. Moorhead, and R. N. Dearth. discrimination of Bordetella avium and Bordetella hinzii. J. Clin. Microbiol.
Further characterization of the agent causing coryza in turkeys. Avian Dis. 41:1512-1519. 2003.
29:690-705. 1985. 12. Skeeles, J. K., and L. H. Arp. Bordetellosis (turkey coryza). In:
7. Kersters, K., K.-H. Hinz, A. Hertle, P. Segers, A. Lievens, O. Diseases of Poultry, 10th ed. B. W. Calnek, H. J. Barnes, C. W. Beard, L. R.
Siegmann, and J. De Ley. Bordetella avium sp. nov. isolated from the McDougald, and Y. M Saif. Iowa State University Press, Ames, Iowa. pp.
respiratory tracts of turkeys and other birds. Int. J. Syst. Bacteriol. 34:56- 275-287. 1997.
70. 1984.

21
6
INFECTIOUS CORYZA
Pat J. Blackall

SUMMARY. Infectious coryza, an acute upper respiratory tract disease of chickens, is caused by the bacterium Avibacterium
paragallinarum. The main impact of the disease is a drop in egg production. Other manifestations such as airsacculitis in broilers, swollen
head-like syndrome, arthritis, and septicemia are either unusual or probably due to complications associated with other infectious agents.
Agent Identification. Diagnosis of infectious coryza is preferably made by the isolation and identification, by biochemical properties, of the
bacterium. A polymerase chain reaction (PCR) test, which can be applied either to suspect colonies or directly to samples from chickens, is
now available. The PCR test is particularly suited for those laboratories that lack suitable expertise and experience in the growth and
phenotypic identification of A. paragallinarum or where samples are transported for long periods to the laboratory. Although most isolates
of A. paragallinarum are dependent upon V factor for growth in artificial media (meaning they show the traditional satellitic growth), some
isolates are V factor-independent. This variation in growth factor requirements, along with the existence of nonpathogenic V factor­
dependent organisms, increases the need for biochemical identification or the use of the PCR test. Serotyping of isolates is important to
guide the use of vaccines.
Serologic Detection in the Host. A range of serologic tests to detect antibodies has been described with hemagglutination-inhibition tests
now being the most widely used.

INTRODUCTION When the disease occurs in chicken flocks in developing countries,


the added presence of other pathogens and stress factors can result
Infectious coryza is an acute respiratory disease of chickens. The in disease outbreaks that are associated with greater economic
clinical syndrome has been recognized since the 1930s and has been losses than those reported in high health flocks in developed
described in the early literature as roup, contagious or infectious countries. In China, outbreaks of infectious coryza have been
catarrh, and uncomplicated coryza (6). Early workers identified the associated with morbidities of 20 to 50% and mortalities of 5 to
causative agent as Haemophilus gallinarum, an organism that 20% (13). In India, outbreaks of infectious coryza are often
required both X (hemin) and V (nicotinamide adenine dinucleotide; complicated with fowl cholera and can result in high mortalities e.g.
NAD) factors for growth in vitro. However, from the 1960s to the one farm experienced 50% mortality in a 14,000 bird layer flock
1980s, all isolates of the disease-producing agent have been shown (33). Arthritis and septicemia, possibly complicated by the presence
to require only V factor and have been termed Haemophilus of other pathogens, have been reported in broiler and layer flocks in
paragallinarum (6). V factor-independent H. paragallinarum South America (27). Infectious coryza can also be a problem in the
isolates have been encountered in the Republic of South Africa (21) village production system e.g. there are reports of outbreaks in such
and Mexico (15). Most recently, a polyphasic taxonomic study has chickens in Thailand (32) and Indonesia (24). Overall, there is
shown that the Haemophilus paragallinarum is not a member of the considerable evidence that infectious coryza outbreaks can have a
genus Haemophilus (2). Thus, H. paragallinarum was allocated to much greater impact in developing countries than in developed
a new genus - Avibacterium - along with several other chicken countries.
associated members of the family Pasteurellaceae (2). Hence, the
causal agent of infectious coryza is now called Avibacterium SAMPLE COLLECTION
paragallinarum, an organism that can be either V factor-dependent
or -independent. This text will use the new terminology of A. Two to three acutely diseased chickens should be killed and the
paragallinarum - even if the primary publications being cited used skin over their sinuses seared with a heated spatula. The skin is
the older terminologies. then incised with a sterile scalpel blade, and a sterile cotton swab is
The disease occurs worldwide and causes economic losses due to inserted into the sinus cavity. Typically, in the early phase of the
an increased number of culls and a marked reduction from 10% to disease, A. paragallinarum is found in pure culture in the sinus.
more than 40% in egg production, particularly on multiage farms. Swabs of the trachea and air sacs may be taken, although the
The disease is essentially limited to chickens and does not threaten organism is less frequently isolated from these areas. Avibacterium
public health. paragallinarum is a fragile organism that does not survive for more
than 5 hr outside of birds. Sinus swabs held in Ames Transport
CLINICAL DISEASE medium (without charcoal) can yield positive cultures for up to 8
days at transport temperatures of either 25 C or room temperature
Infectious coryza may occur in growing chickens and layers. The (9).
most common clinical signs are nasal discharge, facial swelling, Live bird sampling is also possible. In this technique, gentle
lacrimation, anorexia, and diarrhea. Decreased feed and water milking pressure is exerted on the sinus area and mucus forced from
consumption retards growth in young stock and reduces egg the nostril. The expressed mucus should be sampled by a
production in laying flocks (6). In layers, the disease can have a bacteriologic loop, with care being taken to avoid touching the
modi greater impact than the relatively simple scenario described surface of beak or nostril. A swab of the expressed mucus is also
above. As an example, an outbreak of the disease in older layer the optimal sample for the A. paragallinarum PCR. Swabs
birds in California, which was not associated with any other collected by this method will still yield a PCR positive reaction
pathogen, caused a total mortality of 48% and a drop in egg after storage in glycerol-enriched phosphate buffered saline for 180
production from 75 to 15.7% over a 3 wk period (8). The disease days at either 4 C or -20 C (12).
can have significant impact in meat chickens. In California, two
cases of infectious coryza, one complicated by the presence of PREFERRED CULTURE MEDIA AND SUBSTRATES
Mycoplasma synoviae, caused a swollen head like syndrome and
increased condemnations, mainly due to airsacculitis, that varied Artificial Media
from 8.0 to 15% (14). Blood agar is commonly used for the isolation of A.
22
Chapter 6 Infectious Coryza

paragallinarum. The medium is prepared from a dehydrated base whereas A. paragallinarum will colonize and produce typical
such as Bacto-tryptose-blood-agar base (Difco, Detroit, Mich.) and disease.
is enriched with 5% erythrocytes, which may be from any animal.
The inoculum is streaked onto the blood agar plate in the AGENT IDENTIFICATION
conventional maimer, after which the plate is cross-streaked with a
nurse culture. Blood agar is deficient in V factor, and the role of the Background
nurse culture is to excrete excess V factor to support the growth of Avibacterium paragallinarum is not the only growth factor­
A. paragallinarum . Although several bacterial species are possible dependent organism that can be isolated from chickens.
nurse cultures, it is recommended that Staphylococcus hyicus, a Nonpathogenic avian hemophili have been recognized since the
normal inhabitant of the skin of chickens, be used. Avibacterium 1930s. The organisms have been the subject of several taxonomic
paragallinarum is typically grown in the presence of 5% CO2 at 37 changes - being initially named as Haemophilus avium (16) and
C. A convenient procedure is to use candle jars if CO2 incubators then as Pasteurella volantium, Pasteurella avium, and Pasteurella
are not available. species A (22). In a recent polyphasic study, these non-pathogenic
An alternative isolation (and maintenance) medium that is organisms have been transferred to the genus Avibacterium as A.
particularly suited to laboratories in developing countries has been avium, A. volantium and Avibacterium species A (2). The final
described by Terzolo et al. (31). This medium consists of Columbia member of the new genus is A. (Pasteurella) gallinarum.
blood agar base (Becton Dickinson Microbiology Systems, Sparks, A range of other hemophili, none of which are yet assigned to
Md.) with 7% lysed equine blood. The lysed equine blood is named species, have been isolated from birds other than chickens.
prepared by holding fresh equine blood at 56 C for 40 min with The identification of these other hemophili will not be discussed
occasional stirring. The lysed blood can be stored at -20 C. A further here.
selective version of the medium, which should only be used in
parallel with the nonselective medium, contains bacitracin (5 U/ml), Morphology
cloxacillin (5 pg/ml), and vancomycin (25 pg/ml). The plates are Examination of a direct smear of the exudate by Gram stain can be
incubated under a microaerophilic atmosphere. useful for initial assessment of the microbial flora. The finding of
Several complex media have been described that support growth gram-negative rods in direct smears, however, is not sufficient
of avian hemophili (a term used generically to refer to bacteria grounds for definitive diagnosis and must be followed by culture.
within this group). Such media, although not suitable for isolation After incubation of blood agar plates for 24—^48 hr, V factor­
due to problems with overgrowth by contaminants, are particularly dependent isolates of A. paragallinarum produce tiny dewdrop
useful for characterization tests following initial isolation. Two colonies up to 0.3 mm in diameter adjacent to the nurse culture.
media that have proven very useful are Haemophilus maintenance The colonies become smaller with increasing distance from the
medium (HMM) and supplemented test medium agar (TM/SN), nurse culture. For the satellitic growth to be obvious, cultures must
originally described by Rimler et al. (25, 26). HMM base consists be examined within 24-48 hr. The V factor-independent A.
of 1% polypeptone (BBL), 1% biosate peptone (BBL), 0.24% beef paragallinarum isolates produce small colonies (1-2 mm) that do
extract, 0.005% para-aminobenzoic acid, 0.005% nicotinamide, not show satellitic growth. Colonies of A. avium, A. volantium,
0.1% starch, 0.05% glucose, 0.9% NaCl, 0.23% leptospira base Avibacterium species A show satellitic growth and are typically
Ellinghausen-McCullaugh-Johnson-Harris (EMJH) (Difco), and much bigger than V factor-dependent A. paragallinarum. Some
2% Noble agar (Difco). Immediately before being poured, this isolates of A. volantium may produce a yellowish pigment.
medium is supplemented with 0.0025% reduced NAD and 1%
chicken serum. TM/SN base consists of 1% biosate peptone (BBL), Growth Requirements
1% NaCl, 0.1% starch, 0.05% glucose, and 1.5% Noble agar Most avian hemophili have a requirement for V factor but not for
(Difco) and is supplemented with 5% oleic albumin complex, 1% X factor (6). The determination of the growth factor requirements
chicken serum, 0.0005% thiamine, and 0.0025% reduced NAD. A of these organisms is not an easy process. The use of some brands
modified version of TM/SN which consists of brain heart infusion of commercial growth factor discs on media such as brain-heart
agar that is supplemented with 5% oleic albumin complex, 1% infusion agar or susceptibility test agar can result in a high
chicken serum, 0.0005% thiamine, and 0.0025% reduced NAD has percentage of cultures that falsely appear to be both X and V factor­
proven to be as suitable as the original formula given above. Broth dependent. The combination of Oxoid growth factor discs
versions of HMM and TM/SN are prepared by omission of the agar. (Unipath, Ltd., distr. Unipath, Ogdensburg, N.Y.) and TM/S
(TM/SN without added NAD) has been shown to be suitable for
Chicken Embryos growth factor testing (5). Some isolates of avian hemophili may
Avian hemophili can be propagated in 5 to 7-day-old chicken have such lowered V factor requirements that the serum must be
embryos, commonly by inoculation via the yolk sac route. After omitted from TM/S.
overnight incubation, large numbers of hemophili are present in the Alternative tests such as the porphyrin test (18) for X factor testing
yolk, which then can be harvested and preserved by freezing at -70 or the use of purified hemin (X factor) and NAD (V factor) as
C (or lower) or by lyophilization. supplements to otherwise complete media are possible but are
generally too complex for diagnostic laboratories.
Chicken Inoculation V factor-independent isolates of A. paragallinarum have been
Another efficient diagnostic procedure is to inoculate suspect recognized to date in the Republic of South Africa (21) and Mexico
exudate into the infraorbital sinus of two or three susceptible (15). Hence, diagnostic bacteriologists need to be aware of the
chickens (preferably 4 wk old or more). The appearance of the possibility of such variants emerging in other areas.
typical clinical signs of infectious coryza in 24-48 hr is diagnostic. Typically, A. paragallinarum isolates fail to grow in air, although
On occasion, if the number of viable organisms is low, particularly some strains will develop this ability on subculturing. A. avium,
in chronic cases, the incubation period may be delayed for up to 1 A. volantium and Avibacterium species A all grow vigorously in air.
wk. In such cases, a second passage may be required to produce the
typical rapid onset of clinical signs. If the exudate is heavily Physicochemical Properties
contaminated with extraneous bacteria, the chicken inoculation test The ability to reduce nitrate to nitrite and ferment glucose without
can be more reliable than culture. In such instances, most of the the formation of gas is common to all of the avian hemophili.
extraneous bacteria will be cleared by the host defense mechanisms, Oxidase activity, the presence of the enzyme alkaline phosphatase,
23
Pat J. Blackall

and a failure to produce indole or hydrolyse urea or gelatin are also Kume serogroups, the use of absorbed antisera allows the
uniform characteristics. A. paragallinarum is catalase-negative, recognition of serovars, with the nine currently recognized Kume
whereas all other members of the genus Avibacterium including the serovars being A-l, A-2, A-3, A-4, B-l, C-l, C-2, C-3, and C-4 (4).
hemophilic species of A. avium, A. volantium and Avibacterium The antisera, with the exception of the antisera for B-l and C-3,
species A are catalase-positive (2). need to be absorbed to allow recognition of the Kume serogroups.
Carbohydrate fermentation patterns of the avian hemophili are The antisera to be absorbed are diluted 1 in 40 in PBS-B-G. The
generally determined using a phenol red broth (Difco) containing absorption is performed using antigens adjusted to 64 HA units and
1% NaCl, 0.0025% NADH, 1% chicken serum, and 1% at five times die volume of the diluted serum. The adjusted antigen
carbohydrate. For routine identification, the use of this broth and a is centrifuged and the supernatant discarded. The pelleted antigen
dense inoculation (i.e., the modified reference technique of Blackall is resuspended in the diluted antiserum. The suspension is left at
(1)), is a most suitable approach for determining fermentation room temperature for 2 hr and then overnight at 4 C. The
patterns. For larger studies, a replica plating technique is more suspension is then centrifuged and the supernatant retained as the
suitable (1). absorbed antiserum (still at 1 in 40 dilution). Depending on the
Table 6.1 presents those properties that allow full identification of antiserum, a succession of absorptions may need to be performed.
the avian hemophili. The properties of all members of the genus Once the antigen absorption has been completed, the sera need to be
Avibacterium are presented. The failure of A. paragallinarum to absorbed with fixed erythrocytes, using 5 times the volume of fixed
ferment either galactose or trehalose clearly separates it from all erythrocytes compared with the serum. The relevant volume of
other members of the genus, both hemophilic and non-hemophilic. erythrocytes is centrifuged and the supernatant discarded. The
pelleted erythrocytes are resuspended in the diluted antiserum. The
Serotyping Tests suspension is left at room temperature for 2 hr and then overnight at
Two different, but interrelated, serotyping schemes for A. 4C. The suspension is then centrifuged and the supernatant retained
paragallinarum have been used mainly the Page (23) and the Kume as the absorbed antiserum (still at 1 in 40 dilution). The Kume
(19) schemes. The most widely recognized and applied scheme is scheme has not been widely applied as it is technically demanding
the Page scheme, which recognizes three serovars (A, B, and C) of to perform.
A. paragallinarum . Although the original scheme was based on an
agglutination test, the Page serovars are best recognized by a Molecular Identification
hemagglutination-inhibition (HI) test. The antigen for this HI test A polymerase chain reaction (PCR) test that is specific for A.
can be produced by either of two methods. In the first method, paragallinarum has been described (11). The test has been
whole bacterial cells are harvested from an overnight broth and evaluated with a wide range of A. paragallinarum isolates and is
stored at 4 C in 1/100 of the initial broth volume for at least 3 days specific and sensitive. The test, termed the HP-2 PCR, has been
(3). Alternatively, the antigen can be produced by the technique shown to be suitable for use on purified DNA extracts as well as on
originally used in the Kume scheme (19). In this method, cells of A. crude colony preparations obtained from isolation plates. The HP-2
paragallinarum are harvested, washed in phosphate-buffered saline PCR can be used directly on swabs of nasal mucus obtained from
(PBS) (pH 7.0), resuspended in 0.5 M KSCN/0.425 M NaCl to a the squeezing of the nostril. The test can be inhibited in the
density equivalent to a MacFarland nephelometer tube number 5 presence of blood and swabs obtained via by slicing open the infra­
(Difco), held at 4 C for 2 hr with agitation, and then sonicated. The orbital sinus during necropsy are not optimal for use in the HP-2
antigen is washed three times in PBS and resuspended in PBS with PCR. Direct PCR examination of swabs has been shown to
0.01% merthiolate to a density equivalent to a MacFarland outperform culture in China (10, 12). The HP-2 PCR is particularly
nephelometer tube number 5. With either antigen type, the HI test useful in regions where both NAD-independent A. paragallinarum
should be performed with glutaraldehyde-fixed chicken and Omithobacterium rhinotracheale are present. Molecular typing
erythrocytes. The erythrocytes are prepared by collecting fresh methods such as restriction endonuclease analysis (7) and
chicken blood into an equal volume of Alsever’s solution. The ribotyping (20) have proved useful in epidemiologic studies. A
suspension is centrifuged and the erythrocytes are washed three PCR-based typing method - ERIC-PCR - has also shown a capacity
times in 0.15 M NaCl. The erythrocytes are then suspended to 1% to sub-type isolates of A. paragallinarum (30).
(v/v) in a glutaraldehyde-salts solution and held at 4 C for 30 min.
The glutaraldehyde-salts solution is prepared by diluting 25% Maintenance
glutaraldehyde to 1% in a solution containing one volume of 0.15 M Although the initial isolation of A. paragallinarum from acute
Na2HPO4 (pH 8.2), nine volumes of 0.15 M NaCl, and five volumes infectious coryza is not difficult, it is a fragile organism requiring
of distilled water. The fixed erythrocytes are collected by special care for propagation and maintenance in the laboratory.
centrifugation, washed five times in 0.15 M NaCl and five times in Cultures can be maintained on blood agar plates by weekly
distilled water and finally resuspended to 30% (v/v) in distilled passages. Cultures incubated for 24-48 hr at 37 C and then stored
water containing 0.01% merthiolate. This stock of fixed at 4 C in a candle jar will remain viable for up to 2 wk. Cultures
erythrocytes is held at 4 C, and a working dilution of 1% is prepared can be preserved by the lyophilization or freezing (at -70 C or
in PBS (pH 7.0) containing bovine serum albumin (0.1%) and lower) of infected yolk (see the section on chicken embryos).
gelatin (0.001%) (PBS-B-G). Reports that Page serovar B is not a Storage at -70 C of a heavy suspension in the commercial bead
true serovar (28) have been discounted, and Page serovar B has systems now commonly available is also possible.
been conclusively shown to be serologically distinct (34).
The Kume scheme is a hemagglutination-inhibition serotyping SEROLOGIC DETECTION IN THE HOST
scheme using bacterial cells that have been treated with potassium
thiocyanate and then sonicated, and glutaraldehyde-fixed chicken Although a range of serologic tests for the detection of antibodies
erythrocytes. The preparation of the antigen and the chicken to A. paragallinarum have been described (6), only
erythrocytes for the Kume scheme has already been described in the hemagglutination-inhibition (HI) tests are in widespread use. The
section on the Page serotyping scheme. The nomenclature of the various HI tests differ in the methods used to prepare the antigen
Kume scheme has been changed since the original publication. and the type of red blood cells used. For the purposes of this
Under die altered terminology the three Kume serogroups are overview, the three main HI tests have been termed the simple,
termed A, B, and C (4). This terminology emphasizes that the extracted, and treated HI tests. Although most of these HI tests
Kume serogroups correspond to the Page serovars. Within the were originally described as tube or macroplate tests, all can also be
24
Chapter 6 Infectious Coryza

performed in microtiter trays using appropriate volumes. The (one volume of antigen with eight volumes of erythrocytes for 2 hr
simple HI test is based on whole bacterial cells of a Page serovar A at 37 C with serum recovered by centrifugation and regarded as
organism (17). Cells are harvested and suspended in PBS being a one in five dilution).
containing 0.01% merthiolate (pH 7.0). Pretreatment of the sera The extracted HI test has been used to detect antibodies to Page
may be necessary to eliminate nonspecific agglutinins. A 5% serovars A, B, and C in vaccinated chickens (35). Vaccinated
suspension of chicken erythrocytes is added to the sera (1:5 chickens with HI titers of 1:5 or greater in the HI tests based on the
proportion) and the mixture is incubated for 2 hr at room simple and extracted antigens have been found to be protected
temperature and 12 hr at 4 C (29). Two-tenths milliliter of antigen against subsequent challenge (29).
(containing 20 HA units/ml) is added to 0.2 ml of serially diluted
serum (initial dilution of 1:5). After incubation at room temperature DIFFERENTIATION FROM CLOSELY RELATED AGENTS
for 10 min, 0.4 ml of 0.5% chicken erythrocyte suspension
containing 0.02% (v/v) gelatin is added. The HI titers are Infection with A. paragallinarum must be differentiated from other
determined after incubation for 30-40 min at room temperature. diseases, such as chronic respiratory disease, chronic fowl cholera,
This test has been performed mainly using antigen prepared from A. fowl pox, omithobacteriosis (due to O. rhinotracheale), swollen
paragallinarum strain 221. head syndrome (associated with avian pneumovirus) and
The extracted HI test is based on potassium thiocyanate-extracted hypovitaminosis A, which can produce similar clinical signs.
and sonicated cells of A. paragallinarum and glutaraldehyde-fixed Because A. paragallinarum often occurs in mixed infections, one
chicken erythrocytes (29). The preparation of the antigen and the should consider the possibility of other bacteria or viruses as
chicken erythrocytes has already been described in the section on complicating agents, particularly if mortality is high and the disease
the Page serotyping scheme. Although the methodology of the takes a prolonged course.
extracted HI test is as described for the simple HI test, the sera are The simple isolation of a satellitic, gram-negative organism is not
pretreated with 10% glutaraldehyde-fixed erythrocytes to eliminate sufficient to identify an isolate as A. paragallinarum. For
nonspecific hemagglutinins, and PBS containing 0.1% bovine laboratories with limited facilities, sufficient evidence for a
serum albumin and 0.001% gelatin is used as the diluent. diagnosis of infectious coryza would be the isolation of a catalase­
The extracted HI test has been used to detect antibodies in negative, gram-negative organism that exhibits satellitic growth and
chickens receiving inactivated vaccines based on Page serovar C fails to grow in air, together with a history of a rapidly spreading
organisms (29). In chickens infected with a serovar C organism, the acute coryza in a flock.
majority of the birds remain negative in this test (36). Note that, in The use of the PCR is recommended as the definitive test.
this report, the erythrocytes were fixed in formalin instead of Laboratories without access to PCR technology should determine
glutaraldehyde (36). growth factor requirements and the ability to ferment glucose,
The treated HI test is based on hyaluronidase-treated whole galactose, and trehalose. At this level, A. paragallinarum can be
bacterial cells of A. paragallinarum and formaldehyde-fixed differentiated from the other members of the genus Avibacterium.
chicken erythrocytes (37). Whole bacterial cells that have been For laboratories with extensive resources, a complete phenotypic
adjusted to 10 times the optical density of 0.4 at 540 nm are treated identification based on the properties shown in Table 6.1 is
with hyaluronidase (50 units/ml) in phosphate buffer (pH 6.0) in a possible. As V factor-independent A. paragallinarum have been
waterbath at 37 C for 2 hr. The treated antigen is then washed twice isolated, diagnostic microbiologists must be aware of the fact that
in PBS and then resuspended in the original volume of PBS. non-satellitic bacteria should be considered as suspect
The test uses the same methodology as that described for the A. paragallinarum if they show the typical biochemical properties
extracted HI test except that the erythrocytes are 1.0% of the species and have been obtained from chickens showing upper
formaldehyde-fixed chicken erythrocytes. The diluent used in this respiratory disease.
HI test is the same as that used in the extracted HI test. All sera are
pretreated with 50% (v/v) formaldehyde-fixed chicken erythrocytes

Table 6.1. Distinguishing properties of the species within the genera A vibacteriumK

Property Avibacterium Avibacterium Avibacterium Avibacterium avium Avibacterium sp. A.


gallinarum paragallinarum volantium
Catalase +B - + + +
Symbiotic growth - V + + +
Growth in Air + - + + +
Acid from
L-arabinose - - - - +
D-galactose + - + + +
Lactose V - V - -
D-mannitol - + + - V
Maltose + V + - V
D-sorbitol - + V - -
Trehalose + - + + +
ONPG V - + - V
A All species are Gram-negative and non-motile. All species reduce nitrate, are oxidase positive and ferment glucose. Most isolates of Avibacterium
paragallinarum require an enriched carbon dioxide (5-10%) atmosphere and most will show an improved growth in the presence of 5-10% chicken serum.
Most isolates of Avibacterium gallinarum show an improved growth in an enriched carbon dioxide (5-10%) atmosphere.® + = positive (>90%), - = negative
(>90%), V = variable reaction

25
Pat J. Blackall

ACKNOWLEDGEMENT 17. Iritani, Y., G. Sugimori, and K. Katagiri. Serologic response to


Haemophilus gallinarum in artifically infected and vaccinated chickens.
The authors would like to acknowledge the contribution of Dick Yamamoto Avian Dis 21:1-8. 1977.
who has been the senior author or co-author for previous editions of this 18. Kilian, M. A rapid method for the differentiation of Haemophilus strains.
text Acta. Pathol. Microbiol. Immunol. Scand. Sect. B 82:835-842. 1974.
19. Kume, K., A. Sawata, T. Nakai, and M. Matsumoto. Serological
REFERENCES classification of Haemophilus paragallinarum with a hemagglutinin system
J. Clin. Microbiol. 17:958-964. 1983.
1. Blackall, P. J. An evaluation of methods for the detection of carbohydrate 20. Miflin, J. K., R. F. Homer, P. J. Blackall, X. Chen, G. C. Bishop, C. J.
fermentation patterns in avian Haemophilus species. J. Microbiol. Methods Morrow, T. Yamaguchi, and Y. Iritani. Phenotypic and molecular
1.275-281. 1983. characterization of V-factor (NAD)-independent Haemophilus
2. Blackall, P. J., H. Christensen, T. Beckenham, L. L. Blackall, and M. paragallinarum. Avian Dis 39:304-308. 1995.
Bisgaard. Reclassification of Pasteurella gallinarum, [Haemophilus] 21. Mouahid, Μ., M. Bisgaard, A. J. Morley, R. Mutters, and W. Mannheim.
paragallinarum, Pasteurella avium and Pasteurella volantium as Occurrence of V-factor (NAD) independent strains of Haemophilus
Avibacterium gallinarum gen. nov., comb, nov., Avibacterium paragallinarum. Vet. Microbiol. 31:363-368. 1992.
paragallinarum comb, nov., Avibacterium avium comb. nov. and 22. Mutters, R., K. Piechulla, K.-H. Hinz, and W. Mannheim Pasteurella
Avibacterium volantium comb. nov. International Journal of Systematic and avium (Hinz and Kunjara 1977) comb. nov. and Pasteurella volantium sp.
Evolutionary Microbiology 55:353-362. 2005. nov. Int. J. Syst. Bacteriol. 35:5-9. 1985.
3. Blackall, P. J., L. E. Eaves, and G. Aus. Serotyping of Haemophilus 23. Page, L. A. Haemophilus infections in chickens. 1. Characteristics of 12
paragallinarum by the Page scheme: comparison of the use of agglutination Haemophilus isolates recovered from diseased chickens. Am. J. Vet. Res.
and hemagglutination-inhibition tests. Avian Dis 34:643-645. 1990. 23:85-95. 1962.
4. Blackall, P. J., L. E. Eaves, and D. G. Rogers. Proposal of a new serovar 24. Poemomo, S., Sutarma, M. Rafiee, and P. J. Blackall. Characterization
and altered nomenclature for Haemophilus paragallinarum in the Kume of isolates of Haemophilus paragallinarum from Indonesia. Aust. Vet. J.
hemagglutinin scheme. J. Clin. Microbiol. 28:1185-1187. 1990. 78:759-762. 2000.
5. Blackall, P. J., and J. G. Farrah. An evaluation of commercial discs for the 25. Rimler, R. B. Studies of the pathogenic avian haemophili. Avian Dis
determination of the growth factor requirements of the avian haemophili. 23:1006-1018. 1979.
Vet Microbiol. 10:125-131. 1985. 26. Rimler, R B., E. B. Shotts Jr, J. Brown, and R. B. Davis. The effect of
6. Blackall, P. J., and M. Matsumoto. Infectious coryza. In: Diseases of sodium chloride and NADH on the growth of six strains of Haemophilus
Poultry, 11th ed. Y. M. Saif, H. J. Bames, J. R. Glisson, A. M. Fadly, L. R. species pathogenic to chickens. J. Gen. Microbiol. 98:349-354. 1977.
McDougald, and D. A. Swayne, eds. Iowa State University Press, pp. 691- 27. Sandoval, V. E., H. R. Terzolo, and P. J. Blackall. Complicated
703 infectious coryza cases in Argentina. Avian Dis 38:672-678. 1994.
7. Blackall, P. J., C. J. Morrow, A. Mclnnes, L. E. Eaves, and D. G. Rogers. 28. Sawata, A., K. Kume, and Y. Nakase. Biologic and serologic
Epidemiologic studies on infectious coryza outbreaks in northern New South relationships between Page's and Sawata's serotypes of {THaemophilus
Wales, Australia, using serotyping, biotyping, and chromosomal DNA paragallinarum}. Am. J. Vet. Res. 41:1901-1904. 1980.
restriction endonuclease analysis. Avian Dis 34:267-276. 1990. 29. Sawata, A., K. Kume, and Y. Nakase. Hemagglutinin of Haemophilus
8. Bland, Μ. P., A. A. Bickford, B. R. Charlton, G. C. Cooper, F. Sommer, paragallinarum serotype 2 organisms: occurrence and immunologic
and G. Cutler. Case Report: A severe infectious coryza infection in a multi­ properties of hemagglutinin. Am J. Vet. Res. 43:1311-1314. 1982.
age layer complex in central California. In 51st Western Poultry Disease 30. Soriano, V. E., G. Tellez, B. M. Hargis, L. Newberry, C. Salgado-
Conference/XXVH convention anual ANECA. Peurto Vallajarta, Mexico, p. Miranda, and J. C. Vazquez. Typing of Haemophilus paragallinarum strains
56-57. 2002 by using enterobacterial repetitive intergenic consensus-based polymerase
9. Bragg, R. R., P. Jansen Van Rensburg, E. Van Heerden, and J. Albertyn. chain reaction. Avian Dis 48:890-895. 2004.
The testing and modification of a commercially available transport medium 31. Terzolo, H. R, F. A. Paolicchi, V. E. Sandoval, P. J. Blackall, T.
for the transportation of pure cultures of Haemophilus paragallinarum for Yamaguchi, and Y. Iritani. Characterization of isolates of Haemophilus
serotyping. Onderstepoort J Vet Res 71:93-98. 2004. paragallinarum from Argentina. Avian Dis 37:310-314. 1993.
10. Chen, X., Q. Chen, P. Zhang, W. Feng, and P. J. Blackall. Evaluation of 32. Thitisak, W., O. Janviriyasopak, R S. Morris, S. Srihakim, and R. V.
a PCR test for the detection of Haemophilus paragallinarum in China. Kruedener. Causes of death found in an epidemiological study of native
Avian Pathol. 27:296-300. 1998. chickens in Thai villages. Proc. 5th. Inter. Sym Vet. Epidemiol. Economics
11. Chen, X., J. K. Miflin, P. Zhang, and P. J. Blackall. Development and pp. 200-202. 1988.
application of DNA probes and PCR tests for Haemophilus paragallinarum. 33. Tongaonkar, S., S. Deshmukh, and P. Blackall. Characterisation of
Avian Dis 40:398-407. 1996. Indian isolates of Haemophilus paragallinarum. In 51st Western Poultry
12. Chen, X., C. Song, Y. Gong, and P. J. Blackall. Further studies on the Disease Conference/XXVII convention anual ANECA. Peurto Vallajarta,
use of a polymerase chain reaction test for the diagnosis of infectious Mexico, p. 58. 2002
coryza Avian Pathol. 27:618-624. 1998. 34. Yamaguchi, T., P. J. Blackall, S. Takigami, Y. Iritani, and Y. Hayashi.
13. Chen, X., P. Zhang, P. J. Blackall, and W. Feng. Characterization of Pathogenicity and serovar-specific hemagglutinating antigens of
Haemophilus paragallinarum isolates from China. Avian Dis 37:574-576. Haemophilus paragallinarum serovar B strains. Avian Dis 34:964-968.
1993. 1990.
14. Droual, R., A. A. Bickford, B. R. Charlton, G. L. Cooper, and S. E. 35. Yamaguchi, T., P. J. Blackall, S. Takigami, Y. Iritani, and Y. Hayashi.
Channing. Infectious coryza in meat chickens in the San Joaquin Valley of Immunogenicity of Haemophilus paragallinarum serovar B strains. Avian
California. Avian Dis 34:1009-10016. 1990. Dis 35:965-968. 1991.
15. Garcia, A. J., E. Angulo, P. J. Blackall, and A. M. Ortiz. The presence of 36. Yamaguchi, T., Y. Iritani, and Y. Hayashi. Serological response of
nicotinamide adenine dinucleotide-independent Haemophilus chickens either vaccinated or artificially infected with Haemophilus
paragallinarum in Mexico. Avian Dis 48:425-429. 2004. paragallinarum. Avian Dis 32:308-312. 1988.
16. Hinz, K.-H., and C. Kunjara. Haemophilus avium, a new species from 37. Yamaguchi, T., Y. Iritani, and Y. Hayashi. Hemagglutinating activity
chickens. Int. J. Syst. Bacteriol. 27:324-329. 1977. and immunological properties of Haemophilus paragallinarum field isolates
in Japan. Avian Dis 33:511-515. 1989.

26
7
Campylobacter INFECTIONS IN POULTRY
Jaap A. Wagenaar and Wilma F. Jacobs-Reitsma

SUMMARY. Poultry may easily become colonized with Campylobacter jejuni and/or Campylobacter coli. These bacterial species do not
cause clinical disease in poultry but contamination of the meat during laughter and processing is a well recognized source of foodbome
illness in humans.
Agent Identification. Campylobacter is isolated from poultry feces and cecal contents using selective agar media under microaerobic
conditions. Subsequently, suspect isolates are confirmed to be Campylobacter and identified to the species level by using a limited number of
biochemical tests or appropriate PCR tests.
Serologic Detection in the Host. Campylobacter infections are not routinely diagnosed by serologic assays.

INTRODUCTION CLINICAL DISEASE

The gut of poultry easily becomes colonized with the thermophilic Sporadic cases of vibrionic hepatitis in poultry have been
Campylobacter species C. jejuni and C. coli. Other Campylobacter described, supposedly caused by Campylobacter. However, there is
species are very rarely (< 1%) isolated from poultry. Also wild birds only the suggestion of a causative role of Campylobacter without
frequently are found to carry Campylobacter spp., with a strong any proof (7). C. jejuni may cause illness in ostriches but the
association of C. lari present in seagulls. Campylobacters are economic loss due to Campylobacter infections are assumed to be
commensal organisms without causing any clinical disease in very limited (28).
poultry, though there is an immune response and the presence of the Both C. jejuni and C. coli have a high prevalence in poultry, but
organism is detected by the host. The significance of both species generally are considered as normal gut inhabitants. Co­
Campylobacter in poultry is for reasons of human public health and infections with multiple strains frequently occur. There is a strong
not for reasons of clinical poultry diseases. Vertical transmission of seasonality in the prevalence in broiler flocks with higher isolation
the infections does not occur, and eggs are not contaminated. rates in summer (18).
Contamination of meat products is the main issue and consequently Control programs in poultry to reduce the number of infected
C. jejuni and C. coli are of importance only in broilers and other flocks are implemented to prevent human illness from contaminated
meat producing avian species. poultry meat. Epidemiology of Campylobacter infections in broiler
Many broiler flocks are colonized at the age of slaughter because flocks is still not completely elucidated. Recently a systematic
poultry can be infected with low numbers of Campylobacter and review based on UK data and comprising 159 research papers was
Campylobacter is generally present in the environment with published on risk factors for introduction of Campylobacter into
reservoirs in both domesticated and wild warm-blooded animals broiler flocks (1). Partial depopulation (thinning) and multiple
(18, 32). The fact that the organism colonizes in high concentrations poultry houses on a farm were identified as contributing factors
results in shedding of high levels of Campylobacter and intense associated with increased risk, and hygiene barrier, parent company
contamination of the environment. Cleaning and disinfection of and certain seasons of rearing were associated with decreased risk.
poultry houses and the surroundings of the houses between
production cycles is difficult. When the houses are stocked with a Diagnostics
new flock of day-old chickens, the infection cycle can easily start Testing poultry for presence of Campylobacter is performed for
again. When Campylobacter-^QsitiNQ birds are processed in the particular research objectives, for more general monitoring reasons
slaughterhouse, the meat may easily become contaminated with (observing trends), or to identify the Campylobacter status of the
bacteria from the gut contents. Campylobacter will only multiply individual flock. The latter may be used to decide on scheduled
under specific atmospheric conditions with reduced oxygen tension slaughter of negative flocks followed by positive flocks.
and within a temperature range of 30 - 45 C. These conditions are
not present during processing, transport and storage of the meat. In Monitoring programs
contrast with e.g. Salmonella, the Campylobacter concentration will Monitoring programs are implemented to identify trends in
not increase along the food chain in case of troubles with Campylobacter infections and to evaluate the feasibility of control
maintaining chilled conditions. At the consumer level, programs. It offers the possibility to link the poultry data to the
Campylobacter will easily die off during proper heating of the meat human Campylobacter data in order to assess the contribution of
but the risk for humans is mainly in cross contamination from the poultry to the human burden of illness. A good example is the
raw meat to ready-to-eat products like salads. Campylobacter is a obligatory monitoring of Campylobacter in broilers in the EU as
highly infectious organism so even ingesting low doses is prescribed by Zoonoses Directive 2003/99/EC. According to this
associated with a relatively high probability of human illness. directive the monitoring of broiler flocks started on January 1st,
In addition to foodbome infections, humans may become ill from 2005 with the European Food Safety Authority (EFSA) as the
direct contact with infected animals like poultry, but also pets like responsible agency for compilation and reporting of data collected
(young) cats and dogs. Petting zoos are identified as a risk factor by the EU member states. At this moment (February 2006) the EU
especially when less attention is paid to personal hygiene. is unique in the world for this monitoring program.
Campylobacter is one of the most important causes of bacterial (http ://www.efsa. eu.int/science/monitoring zoonoses/reports/catind
gastro-enteritis in humans (30). Campylobacteriosis in humans is ex en.html).
characterized by diarrhea and abdominal cramps. Complications
that may occur are reactive arthritis and the Guillain-Barre Individual flocks
syndrome (10, 25). In some European countries (Denmark, Iceland, Norway) poultry
flocks are tested for Campylobacter prior to slaughter, and negative
and positive flocks are slaughtered separately. The meat from
positive flocks is treated (freezing or heat treatment) to reduce the
Campylobacter concentration, whereas the meat from negative

27
Jnp A Wagenaar and Wilma F. Jacobs-Reitsma

flocks is sold as fresh meat. As consumers are exposed to reduced fecal samples is not performed routinely, enrichment media will not
levels of Campylobacter compared to the situation without be discussed in this chapter.
separation, this approach aims to reduce the burden of illness. The Several commercial enzyme immunoassays are available for the
effect of this approach is strongly dependent on the reliability (in detection of Campylobacter in human stool samples but these tests
particular sensitivity) of the diagnostic procedure, including are not validated for routine use in poultry fecal samples.
collection and transport of samples, and performance of the
detection assay. Selective media for isolation
Many media currently are in use for the bacteriological culture of
SAMPLE COLLECTION Campylobacter spp. and the majority also are commercially
available. A recommendation for a specific medium cannot be
Isolation of Campylobacter from feces and contents of the ceca is given. Experience in laboratories is an important factor in the
described here as these samples are commonly used for the choice of the medium. There are differences in medium preferences
diagnosis of a Campylobacter infection in poultry. Carcass samples in different places in the world. Many European countries use
can be analyzed but this needs a specific approach including mCCDA whereas in the US Campy-cefex and CVA are more
selective enrichment. Isolation of Campylobacter from carcass and commonly used (see list below). A detailed description of the
meat samples is described in detail elsewhere (12,13). developments in Campylobacter detection and the variety of
existing media is given by Corry et al. (8, 9). Campylobacter-
Collection of samples selective media can be divided into two main groups: blood­
A Campylobacter infection rapidly spreads within a flock (31), containing media and charcoal-containing media. Both blood
and close to 100% of the animals become colonized, shedding >106 components and charcoal remove toxic oxygen derivatives. The
cfu Campylobacter per gram feces for a prolonged period of time selectivity of the media is determined by the antimicrobials used.
(at least till slaughter age of broilers). At a within-flock prevalence Cefalosporins (generally cefoperazone) are used, sometimes in
of >40%, 10 samples will be sufficient to detect the Campylobacter combination with other antibiotics (e.g. vancomycin, trimethoprim).
infection at a 95% confidence level. In case of scheduled slaughter Cycloheximide (actidione) or amphotericin B are used to inhibit
it is most important to know the actual Campylobacter status of the yeasts and molds (17). All media allow the growth of C. jejuni and
flock just prior to slaughter. Consequently, the samples should be C. coli but the main difference between the media is the degree of
taken as close to slaughter as possible: at the farm or even at the inhibition of contaminating flora depending on the combination of
slaughterhouse. In the latter case, reliable results can be obtained by antimicrobials used. There is no medium available that allows
taking intact ceca for examination. Living birds can be sampled by growth of C. jejuni and inhibits C. coli or vice versa. To some
taking fecal/cecal droppings or cloacal swabs. For reliable detection extent, other Campylobacter species (e.g. C. lari, C. upsaliensis,
of Campylobacter by culture, freshly voided feces should be C. helveticus, C. fetus and C. hyointestinalis) will also be able to
collected. All samples must be prevented from drying out before grow on some of these media, especially at the less selective
culture. At the slaughterhouse, ceca can be cut with sterile scissors temperature of 37 C. Examples of selective blood-containing solid
from the remaining part of the intestines and submitted intact to the media are: Preston agar (4), Skirrow agar (24), Campy-cefex (29),
laboratory in a plastic bag or Petri-dish. CVA (22).
Examples of selective charcoal-based solid media are:mCCDA
Transport of samples (modified Charcoal Cefoperazone Deoxycholate Agar)(ll), slightly
Campylobacter easily dies off during transport of samples and modified version of the originally described CCDA (5), Karmali
precautions have to be taken to prevent the samples from agar or CSM (Charcoal-Selective Medium) (14), CAT agar
dehydration, atmospheric oxygen, sunlight and elevated (Cefoperazone, Amphotericin, Teicoplanin agar), facilitating
temperature. When swabs are used, a transport medium (like Amies, growth of C. upsaliensis (2).
Cary Blair or Stuart) must be used. These transport swabs are
available commercially. Inoculation of selective media
When only small amounts of fecal/cecal samples can be collected Samples are sown on to the solid selective agar medium by using a
and transport swabs are not available, shipment of the specimen in loop or a swab. Plating for single colonies needs special attention.
transport media is recommended. Several transport media have been
described: Cary-Blair, modified Cary-Blair, modified Stuart Passive filtration
medium, Campythioglycolate medium, alkaline peptone water and Passive filtration avoids the use of selective media and is useful
semisolid motility test medium. Good recovery results have been for the isolation of the more antimicrobial-sensitive Campylobacter
reported using Cary-Blair (15, 23). species (26). In resource-poor countries this method can be used as
No specific recommendation on the temperature for transportation an alternative for the use of expensive selective media. For passive
can be made, but it is clear that freezing and high temperatures filtration, a suspension of the feces is made in PBS (approximately
reduce viability. In general high temperatures (>20 C), low 1/10 dilution). Approximately 100 μΐ of this suspension are then
temperatures (<0 C) and especially fluctuations in temperature must carefully layered on to a 0.45 or 0.65 pm filter, which has been
be avoided. During laboratory processing, samples can be kept at previously placed on top of a nonselective blood agar plate. Care
room temperature for short periods to avoid unnecessary must be taken not to allow the inoculum to spill over the edge of the
temperature shocks. When the time between sampling and filter. The bacteria are allowed to migrate through the filter for 30-
processing is longer, storage at 4 (±2) C is advised. Transport to the 45 min at 37 C or room temperature. The filter is then removed, the
laboratory and subsequent processing should therefore be as rapid fluid that has passed through the filter is spread with a loop or
as possible (preferably the same day, but within at least 3 days). spreader to facilitate the isolation of single colonies. The plate is
incubated microaerobically at 41.5 C (or at 37 C for isolation of
PREFERRED CULTURE MEDIA AND SUBSTRATES non-thermophilic Campylobacter species).

Isolation of Campylobacter from fecal/cecal or intestinal samples Incubation Atmosphere


is usually performed by direct plating onto selective medium or by A microaerobic atmosphere of 5-10% oxygen, 5-10% carbon
using the filtration method on nonselective agar. As enrichment of dioxide (and preferably 5-9% hydrogen) is required for optimal
growth (9). Appropriate microaerobic atmospheric conditions may
28
Chapter 7 Campylobacter Infections in Poultry

be produced by commercial gas generator kits or by (repeated) gas cultures of C. jejuni/C. coli (often also including C. lari) are
jar evacuations followed by atmosphere replacement with bottled commercially available.
gasses. Variable atmosphere incubators can be used but they usually Identification of Campylobacter to the species level. When
need large gas volumes in particular when they are frequently incubation takes place at 42 C, the most frequently encountered
opened. species from poultry samples are C. jejuni and C. coli. However,
low frequencies of other species have been described. Few
Temperature of incubation biochemical tests are available to discriminate between C. jejuni, C.
Media may be incubated at 37 C or 41.5 C, but it is common coli and C. lari. A positive hippurate test surely indicates C. jejuni
practice to incubate at 41.5 C to reduce growth of contaminants and because this is the sole hippurate-positive species within the
to select for optimal growth of C. jejuni/C. coli. (6). Incubation at Campylobacter genus, though some hippurate-negative C. jejuni
41.5 C instead of at 42 C was chosen to harmonize with Salmonella strains have been reported (27).
and Escherichia coli 0157 isolation protocols (12). Table 7.2 gives some basic classical phenotypic characteristics of
the most important thermophilic Campylobacter species in poultry
Time of incubation (12). More extensive speciation schemes have been described in the
C. jejuni and C. coli usually show growth on solid media within literature (20).
24M8 hr at 41.5 C. As the additional number of positive samples
obtained by prolonged incubation is very low, 48 hr of incubation is Table 7.2. Basic phenotypic characteristics of selected thermophilic
recommended for routine diagnosis (12, 6). Campylobacter species.
Characteristics C. jejuni C. coli C. lari

AGENT IDENTIFICATION Hydrolysis of hippurate + - -


Indoxyl acetate + + -
Growth characteristics
On Skirrow or other blood-containing agars, characteristic Key: + = positive; - = negative
Campylobacter colonies are slightly pink, round, convex, smooth
and shiny, with a regular edge. On charcoal-based media such as Susceptibility testing to identify Campylobacter to the species
mCCDA, the characteristic colonies are greyish, flat and moistened, level formerly could be used to discriminate between nalidixic acid­
with a tendency to spread, and may have a metal sheen. sensitive C. jejuni/C. coli and nalidixic acid- resistant C. lari. Due
to the frequently acquired resistance In C. jejuni/C. coli and the
Confirmation identification of nalidixic acid-susceptible C. lari strains, this
A pure culture is required for confirmatory tests, but a preliminary identification method nowadays is obsolete. Biochemical speciation
confirmation can be obtained by direct microscopic examination of may be supplemented or even replaced with molecular methods.
suspect colony material. Speciation results always should be confirmed using defined
According to ISO Campylobacter confirmation is based upon the positive and negative controls.
characteristics listed in Table 7.1 (12). Detection of hippurate hydrolysis. Suspend a loopful of growth
from a suspect colony in 400 μΐ of a 1% sodium hippurate solution
Table 7.1. Confirmatory tests for thermophilic Campylobacter. (care should be taken not to incorporate agar). Incubate at 37 C for
Confirmatory test Result for thermophilic Campylobacter 2 hr, then slowly add 200 μΐ 3.5% ninhydrin solution to the side of
Morphology Small curved bacilli the tube to form an overlay. Reincubate at 37 C for 10 min, and read
Motility Characteristic (highly motile and cork­
the reaction. Positive reaction: dark purple/blue. Negative reaction:
screw like) clear or grey. If commercially available hippurate hydrolysis test
Oxidase + disks are used, follow the manufacturer’s instructions.
Micro-aerobic growth at 25 C Detection of indoxyl acetate hydrolysis. Place a suspect colony
Aerobic growth at 41.5 C
on an indoxyl acetate disk and add a drop of sterile distilled water.
If indoxyl acetate is hydrolysed a color change to dark blue occurs
Key: + = positive; - = negative
within 5-10 min. No color change indicates hydrolysis has not
taken place. If commercially available indoxyl acetate hydrolysis
Microscopic examination of morphology and motility.
test disks are used, follow the manufacturer’s instructions.
Freshly grown material from a suspect colony is suspended in saline
and evaluated, preferably by a phase-contrast microscope, for
Molecular Detection and Identification of Campylobacter
characteristic, spiral or curved slender rods with a corkscrew-like
PCR-based methods for the detection of Campylobacter in animal
motility. Older cultures show less motile coccoid forms.
fecal samples and enriched meat samples have been previously
Detection of oxidase. Take material from a suspect colony and
described in the literature (16, 19). One of these assays is in use in
place it on to a filter paper moistened with oxidase reagent. The
Denmark for routine screening of cloacal swabs from broilers at the
appearance of a violet or deep blue colour within 10 sec is a
slaughterhouse (3, 16).
positive reaction. If a commercially available oxidase test kit is
A variety of DNA probes and polymerase chain reaction (PCR)-
used, follow the manufacturer’s instructions.
based identification assays have been described for Campylobacter
Microaerobic growth at 25 C. Inoculate the pure culture onto a
species. On et al. evaluated the specificity of 11 PCR-based
non-selective blood agar plate and incubate at 25 C in a
identification assays for C. jejuni and C. coli (21).
microaerobic atmosphere for 48 hr. Examine the plate for visible
growth of Campylobacter.
Typing
Aerobic growth at 41.5 C. Inoculate the pure culture onto a non- For several foodbome pathogens like Salmonella serotyping and
selective blood agar plate and incubate at 41.5 C in an aerobic
phage typing of isolates are important tools to trace infections and
atmosphere for 48 hr. Examine the plate for visible growth of
to perform epidemiological studies. Many typing methods for
Campylobacter.
Campylobacter have been described, both phenotypic (serotyping
Latex agglutination tests. In addition to the described
and phage typing) and molecular based (33). For outbreak
confirmatory tests latex agglutination tests for confirmation of pure
investigations any typing method will be suitable. It has to be
noticed that for Campylobacter the current typing methods can only
29
Jaap A Wagenaar and Wilma F. Jacobs-Reitsma

be used for tracing infections restricted in time and geographical isolation of Campylobacter organisms from feces. J. Clin. Microbiol.
area. Although further typing of strains is often requested for 23:456-459. 1986.
epidemiological reasons, the biology of Campylobacter (natural 15. Luechtefeld, N.W., W.L. Wang, MJ. Blaser, and L.B. Reller.
Evaluation of transport and storage techniques for isolation of
competent species with DNA-uptake) hampers the use of large-
Campylobacter fetus subsp. jejuni from turkey cecal specimens. J. Clin.
scale typing in Campylobacter epidemiology. Microbiol. 13:438-443. 1981.
16. Lund, M, A. Wedderkopp, M Waino, S. Nordentoft, D.D. Bang, K.
SEROLOGIC DETECTION IN THE HOST Pedersen, and M Madsen. Evaluation of PCR for detection of
Campylobacter in a national broiler surveillance programme in Denmark. J.
Although intestinal Campylobacter colonisation of poultry is Appl. Microbiol. 94:929-935. 2003.
associated with circulating and mucosal antibody responses, no 17. Martin, K.W., K.L. Mattick, M Harrison, and T.J. Humphrey.
validated serological tests are commonly used for detection of Evaluation of selective media for Campylobacter isolation when
cycloheximide is replaced with amphotericin B. Lett. Appl. Microbiol.
infected birds.
34:124-129. 2002.
18. Newell, D.G., and J. A. Wagenaar. Poultry infections and their control at
REFERENCES
the farm level. In: Campylobacter, Second Edition. I. Nachamkin, and MJ.
Blaser, eds. ASM Press, Washington DC, USA. pp. 497-509. 2000.
1. Adkin, A, E. Hartnett, L. Jordan, D. Newell, and H. Davison. Use of a
19. Olsen, J.E., S. Abo, W. Hill, S. Notermans, K. Wemars, P.E. Granum,
systematic review to assist the development of Campylobacter control
T. Popvic, H.N. Rasmussen, and O. Olsvik. Probes and polymerase chain
strategies in broilers. J. Appl. Microbiol. 100:306-315. 2006.
reaction for the detection of food-borne bacterial pathogens. Int. J. Food
2. Aspinall, S.T., D.R.A. Wareing, P.G. Hayward, and D.N. Hutchinson.
Microbiol. 28:1-78. 1995.
Selective medium for thermophilic Campylobacters including
20. On, S.L.W. Identification methods for Campylobacters, Helicobacters,
Campylobacter upsaliensis. J. Clin. Pathol. 46:82^-831. 1993.
and Related organisms. Clin. Microbiol. Rev. 9:405-422. 1996.
3. Bang, D.D., K. Pedersen, and M Madsen. Development of a PCR assay
21. On, S.L.W., and P.J. Jordan. Evaluation of 11 PCR assays for species­
suitable for Campylobacter spp. mass screening programs in broiler
level identification of Campylobacter jejuni and Campylobacter coli. J. Clin.
production. J. Rapid Meth. Automat. Microbiol. 9:97-113. 2001.
Microbiol. 41:330-336. 2003.
4. Bolton, F.J., and L. Robertson. A selective medium for isolating
22. Reller, L.B., S. Merritt, and L. G. Reimer. Controlled evaluation of an
Campylobacter jejuni/coli. J. Clin. Pathol. 35:462-467. 1982.
improved selective medium for isolation of Campylobacter jejuni from
5. Bolton, F.J., D.N. Hutchinson, and D. Coates. Blood-free selective
human faeces. Abstr. C274 ASM Annual Meeting, p 357. 1983.
medium for isolation of Campylobacter jejuni from faeces. J. Clin.
23. Sjogren, E., G.B. Lindblom, and B. Kaijser. Comparison of different
Microbiol. 19:169-171. 1984.
procedures, transport media, and enrichment media for isolation of
6. Bolton, F.J., D.N. Hutchinson, and G. Parker. Reassessment of selective
Campylobacter species from healthy laying hens and humans with diarrhea.
agars and filtration techniques for isolation of Campylobacter species from
J. Clin. Microbiol. 25:1966-1968. 1987.
faeces. Eur. J. Clin. Microbiol. Infect. Dis. 7:155-160. 1988.
24. Skirrow, MB. Campylobacter enteritis: a new disease. Brit. Med. J.
7. Burch, D. Avian vibrionic hepatitis in laying hens. Vet. Rec. 157:528.
2:9-11. 1977.
2005.
25. Skirrow, MB., and MJ. Blaser. Clinical aspects of Campylobacter
8. Corry, J.E.L., H.I. Atabay, S.J. Forsythe, and L.P. Mansfield Culture
infection. In: Campylobacter, Second Edition. I. Nachamkin, and MJ.
media for the isolation of Campylobacters, helicobacters and arcobacters. In:
Blaser, eds. ASM Press, Washington DC, USA. pp. 69-88. 2000.
Handbook of Culture Media for Food Microbiology, Second Edition. J.E.L.
26. Steele, T.W., and S.N. McDermott. The use of membrane filters applied
Corry, G.D.W. Curtis, and R.M Baird, eds. Elsevier, Amsterdam, The
directly to the surface of agar plates for the isolation of Campylobacter
Netherlands, pp. 271-315. 2003.
jejuni from feces. Pathology 16:263-265. 1984.
9. Corry, J.E.L., D.E. Post, P. Colin, and MJ. Laisney. Culture media for
27. Steinhauserova, I., J. Ceskova, K. Fojtikova, and I. Obrovska.
the isolation of Campylobacters. Int. J. Food Microbiol., 26:43-76. 1995.
Identification of thermophilic Campylobacter spp. by phenotypic and
10. Friedman, C.R., J. Neimann, H.C. Wegener, and R.V. Tauxe R.V.
molecular methods. J. Appl. Microbiol. 90:470-475. 2001.
Epidemiology of Campylobacter jejuni infections in the United States and
28. Stephens, C.P., S.L. On, and J.A. Gibson. An outbreak of infectious
other industrialized nations. In: Campylobacter, Second Edition. I.
hepatitis in commercially reared ostriches associated with Campylobacter
Nachamkin, and MJ. Blaser, eds. ASM Press, Washington DC, USA pp.
coli and Campylobacter jejuni. Vet. Microbiol. 61:183-190. 1998.
121-138. 2000.
29. Stem, N. J., B. Wojton, and K. Kwiatek. A differential-selective
11. Hutchinson, D.N., and F.J. Bolton. Improved blood-free selective
medium and dry ice-generated atmosphere for recovery of Campylobacter
medium for the isolation of Campylobacter jejuni from faecal specimen. J.
jejuni. J. FoodProt. 55:515-517. 1992.
dim Pathol. 37:956-957. 1984.
30. Tauxe, R.V. Emerging food home pathogens. Int. J. Food Microbiol.
12. ISO 10272-1:2006(E) and ISO/TS 10272-2:2006(E). Microbiology of
78:3141. 2002.
food and animal feeding stuffs - Horizontal method for the detection and
31. Van Gerwe, T.J.W.M, A. Bouma, W.F. Jacobs-Reitsma, J. van den
enumeration of Campylobacter spp. Part 1: Detection method, Part 2:
Broek, D. Klinkenberg, J.A. Stegeman, and J.A.P. Heesterbeek. Quantifying
Colony-count technique. International Organization for Standardization
transmission of Campylobacter spp. among broilers. Appl. Environm.
(ISO), ISO Central Secretariat, 1 rue de Varembe, Case Postale 56, CH -
Microbiol. 71:5765-5770. 2005.
1211, Geneva 20, Switzerland, www.iso.org.
32. Waldenstrom, J., T. Broman, I. Carlsson, D. Hasselquist, R.P.
13. Jacobs-Reitsma, W.F. Campylobacter in the food supply. In:
Achterberg, J.AWagenaar, and B. Olsen. Prevalence of Campylobacter
Campylobacter, Second Edition. I. Nachamkin, and MJ. Blaser, eds. ASM
jejuni, C. lari, and C. coli in different ecological guilds and taxa of
Press, Washington DC, USA. pp. 467-481. 2000.
migrating birds. Appl. Environ. Microbiol. 68:5911-5917. 2002.
14. Karmali, MA, A.E. Simor, M Roscoe, P.C. Fleming, S.S. Smith, and
33. Wassenaar, T.M, and D.G. Newell. Genotyping of Campylobacter spp.
J. Lane. Evaluation of a blood-free, charcoal-based, selective medium for the
Appl. Environ. Microbiol. 66:1-9. 2000.

30
8
SPIROCHETOSIS
Stephen R. Collett

SUMMARY. Spirochete infections of birds are caused by helical-shaped bacteria of the order Spirochaetales, family Spirochaetaceae. Two
distinct syndromes exist because of differences in spirochete colonization, pathogenesis, and lesion production- nonrelapsing, tickbome,
acute septicemic borreliosis caused by Borrelia anserina, and subacute-to-chronic intestinal disorders of varying severity caused by a diverse
group of spirochetes.

SEPTICEMIC BORRELIOSIS

Soft ticks in the genus Argas are the reservoir and transmit B. anserina to poultry and free-living avian species. Transmission is through
ingestion of ticks or penetration through mucous membranes or skin and results in septicemia. Strains of B. anserina vary in virulence.
Infection may result in a mild illness or one characterized by mortality approaching 100%. Clinical signs are not specific, but finding larval
ticks on the bird or nymph and adult ticks in the bird’s environment, or presence of pinpoint, cutaneous hemorrhages, especially on the
shanks, where ticks have been feeding, associated with an ill bird increase the likelihood of the disease. Diagnosis is based on identifying the
organism or its antigen in affected birds or finding antibodies in unvaccinated, recovered birds.
Agent Identification. When clinical signs are present, slender, spiral organisms usually can be identified in stained or wet smears of blood
or bufiy coat cells. Darkfield microscopy of bufiy coat smears is especially useful. Spirochetal antigens can be demonstrated by serologic
methods in spleen and liver for several days after organisms disappear from the circulation. Borrelia anserina cannot be grown in
conventional media, but can be isolated by subinoculation of blood into young birds or in chick embryos via yolk-sac inoculation.
Serologic Detection in the Host. Several serologic tests have been developed in areas where the disease is prevalent, but none has been
adopted for widespread use. Serology has proved useful in distinguishing antibodies of B. burgdorferi, which can also infect birds, from
those of B. anserina, and for differentiation of serotypes.

INTESTINAL SPIROCHETOSIS

Spirochetes can be part of the normal large-intestinal flora in many avian species. In other birds, spirochete infection of the large intestine
results in avian intestinal spirochetosis (AIS). Three clinical entities have been described in field cases or experimental studies associated
with intestinal spirochete infection-subclinical infections, mild-to-moderate enteric disease, and severe enteric disease.
Agent Identification. A presumptive diagnosis of AIS is made if clinical signs and lesions of large intestinal disease are present and
spirochetes are visualized in cecal droppings, cecal tissue, or fecal specimens by dark-field, light, or immunofluorescent microscopy.
Isolation of spirochetes on antibiotic-containing agar plates under anaerobic conditions is needed for confirmation. Taxonomic classification
requires biochemical and/or molecular characterization.
Serologic Detection in the Host. Serologic detection is of limited value because of poor sensitivity and specificity of currently available-
methods. Serology will not differentiate infections of non-pathogenic from pathogenic intestinal spirochetes.

SEPTICEMIC BORRELIOSIS

INTRODUCTION it causes significant economic losses. It has occasionally been


identified in the southwestern United States (7). Free-ranging
Avian septicemic borreliosis (spirochetosis) is a tickbome, poultry are more commonly affected, although introduction into
nonrelapsing septicemia of chickens, turkeys, geese, ducks, and a intensively reared commercial flocks can result in high losses.
number of nondomesticated avian species caused by the spirochete Indigenous breeds of chickens are generally more resistant than
Borrelia anserina. The disease was first identified in affected geese imported breeds (19). Where the disease is endemic, exposure erf
in Russia in 1891 (3). young chicks to infective ticks while the chicks still have high
Certain species of soft ticks in the genus Argas, including Argas maternal antibodies results in mild or nonclinical infections and
persicus, A. sanchezi, and A. arboreus, serve as reservoirs and subsequent resistance, which is maintained by continued,
vectors of the organism. Infection of avian species is not essential intermittent exposure. Clinical disease occurs when susceptible
for survival of the organism; transovarial and transstadial populations are placed in contact with infective ticks through
transmission within the tick ensure the continuing survival of introduction of birds into flocks or environments where ticks are
B. anserina (40). Horizontal infection can occur by cannibalism, present or birds parasitized by larval ticks are placed into
accidental transfer of contaminated blood or infective excreta to susceptible flocks (21,29).
susceptible birds, mechanically via other biting arthropods, or by
ingestion of organisms in carcasses or secretions from infected birds CLINICAL DISEASE
or infected ticks (21). Virulent strains can cause infection by direct
penetration of intact mucous membranes, including conjunctiva, Infection ranges from inapparent to very severe depending on
and skin (18). Recovered birds are not carriers, but may have a degree and type of exposure and virulence of the strain <rf
prolonged period of poor growth and production and persistent spirochete infecting the bird. Strains causing outbreaks in the
neurologic signs. Relapses do not occur (10). United States are usually of intermediate to low virulence (7). Onset
Occurrence of spirochetosis coincides with distribution of tick of clinical signs generally occurs 3-12 days after exposure to
vectors and a seasonal pattern often occurs that is related to tick infective ticks, although it may be shorter following experimental
activity. The disease is endemic in most tropical, subtropical, and inoculation (19,21).
warm temperate areas, particularly arid and semiarid regions, where Clinical signs are not specific for spirochetosis. Affected birds «
inactive, cyanotic, anorexic, dehydrated, experience rapid weight
31
Stephen R. Collett

loss, have fluid to mucoid green fecal droppings containing excess expressing the tube's contents subcutaneously. Weekly transfer of
bile and white to pale yellow urates, and are usually febrile. Birds virulent strains is easily accomplished by clipping a toenail of an
may show nervous signs, become comatose, and have subnormal infected chick short enough to obtain a drop of blood and then
body temperatures prior to death. Recovered birds often have a instilling the drop into the eye of a 1-day-old chick to be exposed-
prolonged convalescent period and residual weakness or paralysis Use of 1-day-old chicks will ensure that spirochetemia is still
of one or both wings and/or legs (21,26,29). present at 7 days postinoculation.
Nonhemolytic anemia in affected birds results from
erythrophagocytosis causing decreased total erythrocytes, AGENT IDENTIFICATION
hemoglobin, and packed cell volumes. Anemia is most severe after
organisms disappear from the blood. Other hematologic changes Finding B. anserina in blood or tissues from ill birds is diagnostic
include leucocytosis, primarily resulting from increased of septicemic borreliosis. The organism is usually readily identified
mononuclear cells, granulocytopenia, and prolonged blood clotting in dried or wet blood smears. On stained smears it is a long, slender,
(31,32). helical bacterium, usually with 5-8 spirals, that measures 6-30 x
Enlargement of the spleen with a prominent mottled appearance is 0.3 pm. Borrelia anserina stains blue to purple with dyes used
characteristic of spirochetosis. Mottling is caused by hyperplasia routinely for hematology (5). It can also be stained with most other
and/or fibrinoid necrosis of mononuclear phagocytes surrounding dyes used for demonstrating bacteria although the Gram-stain
sheathed capillaries. Liver enlargement, occasionally with pale or procedure is generally not used because of considerable background
red foci, and renal swelling and pallor are also typical lesions of staining of the blood. Stained blood films can be held for later
spirochetosis. Pale and red foci in the liver result from necrosis and examination, provide a permanent record, and do not require special
hemorrhage, respectively (2,26). Extensive muscle necrosis and microscopic equipment.
hemorrhage occurred in infected pheasants (23). Serositis, which is Spirochetes are also readily identified in wet smears by dark-field,
typical of most other bacterial septicemias, is not a finding in phase-contrast, or interference-contrast microscopy. Wet smears are
spirochetosis, although mild pericarditis can infrequently be seen. preferred for identifying low numbers of organisms as may occur in
Lesions are less evident or may be absent when infection results the early or late stages of the disease or when infections are caused
from strains of low virulence (7,9). by low-virulent strains, and for demonstrating spirochetes in
decomposed tissues or ticks. Care needs to be taken to differentiate
SAMPLE COLLECTION pseudospirochetes from spirochetes when examining wet smears.
Spirochetes have a definite, uniform, spiral (wavy) appearance, are
Spirochetes are usually present in the blood during the febrile highly motile and show directed, purposeful movement when
period but may be too few to detect very early in the disease and viable, and may or may not adhere to blood cells.
rapidly disappear following recovery (11,15,26). If death occurs Pseudospirochetes, which are most likely extrusions from
prior to their disappearance, spirochetes can be easily identified in degenerating erythrocytes, are slender but do not have a spiral
wet smears of clotted blood for several days, especially if carcasses shape, are of variable lengths and morphology, tend to alter in shape
have been refrigerated (26). Stained smears of decomposed tissues as they float in the medium, frequently have a small terminal hook
are generally unsatisfactory due to background staining. or mass, have no purposeful movement, and are frequently attached
Subinoculation of blood or tissues from dead birds may or may not to blood cells (14).
result in infection as organisms identified in wet smears may be Spirochetes in blood can be concentrated in the buffy coat by
nonviable. Spirochetal antigen can be demonstrated in tissues, centrifugation (13). A convenient method of demonstrating
especially spleen and liver, by agar-gel immunodiffusion (AGID) spirochetes is to centrifuge blood in a microhematocrit tube, score
for several days after spirochetes are no longer present in the the tube with a diamond-tipped pen at the interface of the packed
circulation (1). Mucoid droppings passed by affected birds contain erythrocytes and buffy coat, break the tube, and express the buffy
spirochetes and are infective when fed to susceptible chicks or coat and plasma onto a microscopic slide. Place a cover slip on the
poults (23,26). sample and gently press it to spread the buffy coat into an even
circle. Allow the wet smear to sit for approximately 5 min and then
PREFERRED CULTURE MEDIA AND SUBSTRATES examine the margin of the buffy coat for spirochetes that have
emerged into the plasma. Samples can be quickly and efficiently
Borrelia anserina cannot be cultured using routine bacteriologic evaluated with this technique.
media or techniques. The organism has been grown in Barbour- In histopathologic sections, spirochetes can be demonstrated by a
Stoenner-Kelly medium, but lost virulence after 12 passages (22). number of silver impregnation procedures (7,21).
Subinoculation of blood or tissues into susceptible young chicks or On electron microscopy, borreliae have 15-22 periplasmic flagella
poults, with daily examination of blood for 10-14 days subterminally that overlap centrally so that 30-44 flagella are
postinoculation is the best method for isolating the spirochete. present in the middle of the spirochete (5).
Inoculation of susceptible 1-day-old chicks or embryos after 18
days of incubation will result in birds with prolonged, high SEROLOGIC DETECTION IN THE HOST
spirochetemia persisting for as long as 21 days with spirochetes
comprising as much as 4% of total blood volume. The organism is Several serologic tests including agglutination, immobilization,
also readily isolated by yolk-sac inoculation of chicken or turkey precipitin, and fluorescent antibody procedures (27) been developed
embryos (24). Embryo mortality generally occurs 4-7 days and used in areas where spirochetosis is prevalent to detect
postinoculation; if not, viable embryos should be checked on the antibodies in serum and egg yolk (3). Spirochetal antigens can be
sixth day following inoculation. Spirochetes are present in high detected in organs, especially liver and spleen, after organisms have
numbers in tissues, blood, and blood-contaminated embryonic disappeared from the blood by immunodiffusion or
fluids. Mammals are resistant to infection. immunofluorescence (2). Serology has also been used to determine
Strains can be maintained in infective blood stored at -70 C (an that there are different antigenic strains of B. anserina (8,20,38).
equal volume of dimethylsulfoxide can be added to assist in
cryoprotection), in infected ticks, or by weekly transfer of blood
into 1-day-old chicks. Frozen blood can be stored in capillary tubes,
which can be used to puncture the skin and expose the bird by
32
Chapter 8 Spirochetosis

DIFFERENTIATION FROM CLOSELY RELATED AGENTS resulting from Marek’s disease, leukosis, and type Π adenovirus
infection. Lesions characteristic of Marek's disease and leukosis
Spirochetosis must be differentiated from other types of should be present in other tissues and typical intranuclear inclusions
septicemia. Negative bacteriologic cultures will rule out and/or antigen can usually be demonstrated when splenomegaly
colibacillosis, pasteurellosis, and other less common systemic results from type Π adenovirus infection.
bacterial diseases. Absence of serositis (except for occasional mild Borrelia burgdorferi will infect birds but does not cause clinical
pericarditis) will usually distinguish spirochetosis from illness. Spirochetemia may or may not be present but would
chlamydiosis. Splenomegaly occurring in spirochetosis, although probably be low making it unlikely that organisms would be found
not a constant finding when birds are infected with low-virulent in blood smears. Specific antigens and antibodies can be used to
strains of B. anserina, must be distinguished from splenomegaly differentiate B. anserina from B. burgdorferi infections (12,39).

INTESTINAL SPIROCHETES

INTRODUCTION isolation, but success of isolation is lower than for cultures of cecal
mucosa.
Intestinal spirochetes are a heterogenous group of spiral bacteria
that colonize the large intestine of a variety of mammalian and PREFERRED CULTURE MEDIA AND SUBSTRATES
avian hosts including swine, humans, and domestic poultry (36,37).
Such colonization may be part of the normal intestinal flora, or can Avian intestinal spirochetes are anaerobic (36,37). Primary
be associated with or be the cause of clinical disease. Avian isolation has been accomplished on various solid media systems
intestinal spirochetes are related to but distinct from B. anserina, the used originally to isolate swine intestinal spirochetes. Typically,
spirochetal etiology of nonrelapsing tickbome acute septicemic these systems contain a blood agar base, such as trypticase-soy agar
borreliosis. with 5-10% sheep blood, and one to five selective antibiotics (i.e.,
Spirochetes isolated from intestinal tracts of birds include 400 pg/ml spectinomycin, 6.25 pg/ml rifampin, 25 pg/ml
Brachyspira (Serpulina) hyodysenteriae, B .innocens, B. pilosicoli spiramycin, 12.5 pg/ml vancomycin, or 12.5 pg/ml colistin) to
B. intermedia, B. alvinipulli and B. murdochii. However, most inhibit indigenous anaerobic intestinal microflora (4). An anaerobic
avian intestinal spirochetes are unclassified. atmosphere such as 94% N2/6% CO2 provides a good environment
for primary isolation, but the addition of 1% O2 enhances growth of
CLINICAL DISEASE some isolates. Incubation is at 37-42 C for a minimum of 10 days;
however growth usually occurs within 2-5 days.
Avian intestinal spirochetosis (AIS) is a subacute to chronic,
nonsepticemic, intestinal disorder characterized by spirochetes in AGENT IDENTIFICATION
the cecum and/or rectum, and variable clinical illness, morbidity,
and mortality. Three clinical entities exist - subclinical infections, Visual demonstration of helical-shaped bacteria in feces or cecal
mild to moderate disease, and severe disease. In the first of these droppings by dark-field or light microscopy is sufficient for
clinical entities, spirochetes identified in ceca of most wild and tentative identification of spirochetes. Confirmation of helical­
many domestic birds are not associated with clinical disease and are shaped bacteria as spirochetes requires visualization of distinctive
considered to be nonpathogenic (35). In the second clinical form, ultrastructural features, demonstration of spirochete antigens
AIS in domestic poultry presents with diarrhea, pasty vents, wet (immunofluorescent microscopy), or isolation in culture. However,
feces containing increased crude fat, retarded growth rates, delayed ultrastructural morphology or antigen demonstration will not
onset of laying, production of fecal-stained eggshells, and frothy, distinguish between spirochete groups or species. Culturing with
yellow to brown fluid in dilated ceca (36,37). Daily mortality rates further biochemical or molecular characterization is necessary for
are normal or minimally increased. In the third clinical entity, AIS categorization or taxonomic classification. In addition, because
in juvenile common rheas presents with necrotizing typhlitis and spirochetes can be normal flora or produce subclinical infections,
mortality rates up to 80% (4,23). Some birds show depression, have characteristic clinical signs and lesions must be present to
reduced body weights, and pass watery feces with caseous cores 1- distinguish spirochetes of normal flora from those of AIS.
2 days before death (37). However, production of AIS in rheas may
require synergism between spirochetes and indigenous, anaerobic Cell Morphology and Staining
bacilli of the ceca. Spirochetes are gram-negative, helical-shaped bacteria (6). Avian
intestinal spirochetes are readily identified by the characteristic
SAMPLE COLLECTION corkscrew type of motility in wet mounts of cecal contents using
dark-field microscopy. Spirochete cells are easily visualized in
Colonization is most frequent in the cecum and rectum, impression smears or tissue sections of cecal mucosa. Spirochetes
occasionally extending into the ileum (37). Spirochetes primarily are dark brown to black using silver-stain techniques and blue using
colonize the ciypt lumina, and to a lesser extent the intestinal Wright-Giemsa stains.
contents adjacent to villous epithelium. On electron microscopy, each spirochete cell contains a central
Fresh cecal droppings or cecal mucosa are optimal specimens for protoplasmic cylinder, two sets of endocellular periplasmic flagella
isolation or direct demonstration of intestinal spirochetes. Samples (axial filaments), and an outer envelope (outer sheath) (6).
chilled at 4 C for up to 1 wk are acceptable, but frozen samples, Visualization of periplasmic flagella is diagnostic of spirochetes.
especially those stored at -20 C, will have low success rates for Individual cells have lengths of 7-19 pm, diameters of 0.25-0.6
primary isolation. For live birds, use of noninvasive sampling such pm, amplitudes of 0.45-0.79 pm, and wavelengths of 2.7-3.7 pm
as cloacal swabs or feces are practical alternatives for spirochete (37).

33
Stephen R Collett

Table 8.1. Multilocus enzyme electrophoresis (MEE), phenotypic, and biochemical characteristics of avian intestinal spirochetes
(16,25,33,34,36).

Taxonomic classification

Brachyspiraa Brachispira Brachyspira. Unnamed Brachispira Brachyspira Brachyspira


hyodysenteriae intermedia innocens alvinipulli murdochii pilosicoli

Host species Common rhea Chicken Chicken Chicken Chicken Chicken Chicken,
(Rhea waterfowl
americana)

Pathogenicity Severe Moderate None Unknown Mild Unknown Mild

MEE profile A b c d e F g

Periplasmic 8:16:8 8:16:8 8:16:8 8:16:8 8:16:8 8:16:8 5:10:5


flagellar ratios

β-Hemolysis Strong Variable Weak Weak Weak Weak Weak

Indole + + + - - - -

API ZYM profile 14.0.13.11.1


10.0.4.10.1
14.0.15.11.1 14.0.12.3.0
14.0.4.2.1 14.0.12.7.1
14.0.4.10.1 14.0.12.3.0 14.0.12.3.1 14.0.4.3.1 14.0.4.3.0
14.0.12.10.1 6.0.12.6.1
14.0.13.3.1 14.0.13.11.8
10.0.13.10.1
14.0.13.15.1

Colony Morphology threshold of ~ 103 cells/g feces making it more sensitive and rapid
Because of the vigorous motility, spirochetes spread rapidly over (1 day vs. 7-10 days) than the traditional culture/PCR techniques.
agar plates and do not form discrete colonies. With some isolates,
growth is detectible by hemolysis of the blood agar. However, with SEROLOGIC DETECTION IN THE HOST
nonhemolytic or slow-growing weakly β-hemolytic spirochetes,
areas of growth are evident as a dull sheen on the agar surface. Serologic tests have been used to survey avian species and swine
Bacterial growth on plates should be confirmed as spirochetes by to determine exposure to intestinal spirochetes (35,37). The tests are
characteristic motility on wet mounts using dark-field microscopy not based on species-specific antigens, have low specificity and
or spiral morphology as visualized in Wright-Giemsa or Gram- sensitivity, and do not distinguish between infections of
stained smears. nonpathogenic and pathogenic spirochetes. AGID provides a single
Isolated bacteria can be verified as spirochetes by transmission test method to screen birds from diversely different orders and
electron microscopy. However, demonstration of spirochete families for spirochete infections (35).
antigens by direct or indirect fluorescent (27) or Agar gel immunodiffusion plates (0.6% agar, 0.6% agarose, 7%
immunohistochemical methods is easier and more rapid than NaCl, 3% polyethylene glycol, and 0.05 mg/ml sodium azide) have
electron microscopy. Categorization or taxonomic classification spirochete antigen placed into the center well and two-fold dilutions
requires additional testing such as serotyping, growth-inhibition of unknown, nonhemolyzed sera in surrounding wells (35). Plates
testings, biochemical differentiation profiling, rRNA restriction are incubated at 20 C and read on day 3. Positive and negative
pattern analysis, or multilocus enzyme electrophoresis (MEE) control sera should be included in each test run. The spirochete test
(17,36,37). antigen is derived by washing known spirochete cells from culture
plates (brain-heart infusion agar plates with 10% fetal calf serum)
Biochemical Properties with 0.9% phosphate- buffered saline (PBS), centrifugation twice to
Avian intestinal spirochetes contain alkaline and acid pellet, final resuspension in 2% sodium dodecyl sulfate in PBS,
phosphatases, esterase, esterase lipase, β-galactosidase, and sonication for 30 sec at 4 C, and heating at 60 C for 30 min.
phosphorylase (34). Differences in hemolysis patterns, indole Other tests for antispirochete antibody detection include enzyme-
production, and the presence or absence of α-galactosidase and a- linked immunosorbent assay, plate and microagglutination tests,
glucosidase activities have been used to categorize avian and passive hemolysis assay, and indirect fluorescent antibody test (36).
mammalian intestinal spirochete isolates and to predict the potential
for causing disease (Table 8.1). Use of an API ZYM® (bioMerieux- DIFFERENTIATION FROM CLOSELY RELATED AGENTS
Vitek, Hazelwood, Mo.) biochemical coding system and MEE have
been useful in characterizing avian and mammalian isolates In poultry, spirochetes identified in fecal specimens should be
(17,25,34). distinguished from other spiral bacteria including Campylobacter,
Arcobacter, Helicobacter, and Spirillum (36). Many spiral bacteria
PCR Assay can be nonpathogenic indigenous flora of gastrointestinal tracts of
Phillips et al. (28) recently reported a two-step nested duplex PCR birds. In cases of chronic diarrhea or pasty vents nutritional
(2S-N-D-PCR) assay for detection of the two most commonly problems such as excess dietary salt or fats should be investigated.
reported species in chicken (B. pilosicoli and B. intermedia). When Other causes of chronic diarrhea include enteric salmonellosis,
performed on fecal samples this 2S-N-D-PCR assay has a detection colibacillosis, and coccidiosis.
34
Chapter 8 Spirochetosis

In rheas, intestinal spirochetes as the cause of necrotizing typhlitis 19. Khogali, A. R., and A.M Shomein. Studies on spirochaetosis in fowls
must be distinguished from other potential causes including group B in the Sudan. I. Epizootiology and experimental transmission. Bull. Epizoot
Salmonella, Clostridium perfringens, C. sordelli, and Histomonas Dis. Afr. 22:251-254. 1973.
20. Kligler, I. J., D. Hermoni, and M Perek. Studies on fowl spirochetosis.
meleagridis (36). Furthermore, the gross cecal lesions of eastern
Π. Presence of serologically differentiated types of spirochetes. J. Comp.
equine encephalitis virus (EEEV) infection can be confused with Pathol. Ther. 51:206-212. 1938.
AIS, but EEEV also produces hemorrhage in the small intestine and 21. Knowles, R., B. M Das Gupta, and B. C. Basu. Studies in avian
widespread petechiation and necrosis in visceral organs. spirochaetosis. Indian Med. Res. Mem 22:1-113. 1932.
22. Levine, J. F., M J. Dykstra, W. L. Nicholson, R. L. Walker, and G.
REFERENCES Massey. Attenuation of Borrelia anserina by serial passage in liquid
medium. Res. Vet. Sci. 48:64-69. 1990.
1. Al-Attar, M A., and F. M. Jahanly. Demonstration by immunodiffusion 23. Mathey, W. J., Jr., and P. J. Siddle. Spirochetosis in pheasants. J. Am.
agar-gel test of Borrelia anserina antigens in the organs of infected chickens. Vet. Med Assoc. 126:123-126. 1955.
Avian Dis. 18:463^166. 1974. 24. McKercher, D. G. The propagation of Borrelia anserina in embryonated
2. Bandopadhyay, A. C., and J. L. Vegad. Observations on the pathology of eggs employing the yolk sac technique. J. Bacteriol. 59:446-447. 1950.
experimental avian spirochaetosis. Res. Vet. Sci. 35:138-144. 1983. 25. McLaren, A. J., D. J. Trott, D. E. Swayne, S. L. Oxberry, and D. J.
3. Bames, H. J. Spirochetosis (borreliosis). In: Diseases of Poultry, 10th ed. Hampson. Genetic and phenotypic characterization of intestinal spirochetes
B. W. Calnek, H. J. Bames, C. W. Beard, L. R. McDougald, and Y. M Saif, colonizing chickens, and allocation of known pathogenic isolates to three
eds. Iowa State University Press, Ames, Iowa. pp. 318-324. 1997. distinct genetic groups. J. Clin. Microbiol. 35:412-417. 1997.
4. Buckles, E. L., K. A. Eaton, and D. E. Swayne. Cases of 26. McNeil, E., W. R. Hinshaw, and R. E. Kissling. A study of Borrelia
spirochete_associated necrotizing typhlitis in captive common rheas (Rhea anserina infection (spirochetosis) in turkeys. J. Bacteriol. 57:191-206. 1949.
americana). Avian Dis. 41:144-148. 1997. 27. Prudovsky, S., A. Hadani, M Rubina, and A. Skiair. The use of the
5. Burgdorfer, W., and T. G. Schwan. Borrelia. In: Manual of clinical indirect fluorescent antibody technique in avian spirochaetosis. Avian
microbiology, 5th ed. A. Balows, W. J. Hausler, Jr., K. L. Herrmann, H. D. Pathol. 7:421-425. 1978.
Isenberg, and H. J. Shadomy, eds. American Society of Microbiologists, 28. Phillips, N. D., T. La, and D. J. Hampson. Development of a two-step
Washington, D.C. pp. 560-566. 1991. nested duplex PCR assay for the rapid detection of Brachyspira pilosicoli
6. Canale Parola, E. Order I. Spirochaetales Buchanan 1917, 163. In: and Brachyspira intermedia in chicken faeces. Veterinary Microbiology
Bergey's manual of systematic bacteriology, vol. 1. N. R. Krieg, ed. 116. 239-245. 2006.
Williams and Wilkins, Baltimore, Md. pp. 38 70. 1984. 29. Rao, S. B. V. Spirochaetosis in poultry. Research Series No. 18. ndian
7. Cooper, G. L., and A A Bickford. Spirochetosis in California game Council of Agricultural Research, New Delhi, India. 1958.
chickens. Avian Dis. 37:1167-1171.1993. 30. Sagartz, J. E., D. E. Swayne, K. A. Eaton, J. R. Hayes, K. D. Amass, R.
8. DaMassa, A. J., and Η. E. Adler. Avian spirochetosis: natural Wack, and L. Kramer. Necrotizing typhlocolitis associated with a spirochete
transmission by Argas (Persicargas) sanchezi (Ixodoidea:Argasidae) and in rheas (Rhea americana). Avian Dis. 36:282 289. 1992.
existence of different serologic and immunologic types of Borrelia anserina 31. Soliman, Μ K., A. A. S. Ahmed, S. El Amrousi, and I. H. Moustafa.
in the United States. Am. J. Vet. Res. 40:154-157. 1978. Cytological and biochemical studies on the blood constituents of normal and
9. DaMassa, A. J., and Η. E. Adler. Avian spirochaetosis: enhanced spirochete-infected chickens. Avian Dis. 10:394-400. 1966.
recognition of mild strains of Borrelia anserina with bursectomized and 32. Soni, J. L., and A. G. Joshi. A note on strain variation in Akola and
dexamethasone-treated chickens. J. Comp. Pathol. 89:413-420. 1979. Jabalpur strains of Borrelia anserina. Zentralbl. Veterinaermed. Reihe B
10. Dickie, C. W., and J. Barrera. A study of the carrier state of avian 27:70-72. 1980.
spirochetosis in the chicken. Avian Dis. 8:191-195. 1964. 33. Stanton, T. B., D. Postic, and N. S. Jensen. Serulina alvinipulli sp.
11. Djankov, I., I. Soumrov, T. Lozeva, and P. Penev. Persistence and Nov., a new Serpulina species that is enteropathogenic for chickens. Int J.
excretion of Treponema anserinum (Sakharoff, 1891) in hens. Zentralbl. of Systematic Bacteriology. 48, 669-676. 1998.
Veterinaermed. ReiheB 17:544-548. 1970. 34. Stoutenburg, J. W. Studies of intestinal spirochetes in avian species.
12. Fabbi, Μ, V. Sambri, A. Marangoni, S. Magnino, F. S. Basano, R. MS. Thesis. Ohio State University, Columbus, Ohio. 1993.
Cevenini, and C. Genchi. Borrelia in pigeons: no serological evidence of 35. Stoutenburg, J. W., D. E. Swayne, T. M Hoepf, R. Wack, and L.
Borrelia burgdorferi infection. J. Vet. Med. Ser. B 42:503-507. 1995. Kramer. Frequency of intestinal spirochetes in avian species from a zoologic
13. Goldschmid, J. M, and K. Mahomed. The use of the microhematocrit collection and private rhea farms in Ohio. J. Zoo Wildl. Med. 26:272_278.
technic for the recovery of Borrelia duttoni from the blood. Am. J. Clin. 1995.
Pathol. 58:165-169. 1972. 36. Swayne, D. E. Avian intestinal spirochetosis. In: Diseases of poultry,
14. Greene, R. T., R. L. Walker, and C. E. Greene. Pseudospirochetes in 10th ed. B. W. Calnek, H. J. Bames, C. W. Beard, L. R. McDougald, and Y.
animal blood being cultured for Borrelia burgdorferi. J. Vet. Diagn. Invest. M Saif, eds. Iowa State University Press, Ames, Iowa. pp. 325-332. 1997.
3:350-352. 1991. 37. Swayne, D. E., and A J. McLaren. Avian intestinal spirochaetes and
15. Gross, W. M., and M R. Ball. Use of fluorescein-labeled antibody to avian intestinal spirochaetosis. In: Intestinal spirochaetes in domestic
study Borrelia anserina infection (avian spirochetosis) in the chicken. Am. J. animals and humans. D. J. Hampson and T. B. Stanton, eds. CAB
Vet. Res. 25:1734-1739. 1964. International, Wallingford, United Kingdom, pp. 267_300. 1997.
16. Hampson, D. J., and T La. Reclassification of Serpulina intermedia and 38. Verma, R. K., A. R. Muley, and K. N. P. Rao. Some observations on the
Serpulina murdochii in the genus Brachyspira as Brachyspira intermedia strain variation of Borrelia anserina. Indian Vet. J. 68:818-821. 1991.
comb. nov. and Brachyspira murdochii comb. nov. Int. J. of Systematic 39. Walker, R. L., R. T. Greene, W. L. Nicholson, and J. F. Levine. Shared
Evolutionary Microbiology 56,1009-1012. 2006 flagellar epitopes of Borrelia burgdorferi and Borrelia anserina. Vet
17. Hunter, D., and T. Wood An evaluation of the API ZYM system as a Microbiol. 19:361-371. 1989.
means of classifying spirochaetes associated with swine dysentery. Vet. Rec. 40. Zaher, M A., Z. R. Soliman, and F. M Diab. An experimental study of
104:383 384. 1979. Borrelia anserina in four species of Argas ticks. 2. Transstadial survival and
18. Kapur, H. R. Transmission of spirochaetosis through agents other than transovarial transmission. Z. Parasitenkd 53:213-223. 1977.
Argas persicus. Indian J. Vet. Sci. Anim. Hush. 10:354-360. 1940.
9
ERYSIPELAS
George L. Cooper and Arthur A. Bickford

SUMMARY. Erysipelothrix rhusiopathaiae, the causal agent of erysipelas, is one of two species of bacteria included in the genus
Erysipelothrix (Erysipelothrix tonsillarum is a recent addition).
Agent Identification. The organism can be cultured directly onto blood agar plates, on selective media such as Packer’s, or in
Erysipelothrix selective broth. Cultures should be incubated at 37 C in 5%-10% CO2 Presumptive identification of the bacterium is based on
morphologic and Gram-staining characteristics, and on the ability of the organism to produce H2S in triple sugar iron agar. Definitive
identification requires further biochemical characterization or animal inoculation. Twenty-two serovars of E. rhusiopathiae have been
identified on the basis of heat-stable somatic antigens. Most isolates are serovar 1 or 2, however, pathogenicity and immunogenicity are
apparently not associated with serotype. Multilocus enzyme electrophoresis (MLEE) and ribotyping methods have been used to characterize
different strains of E. rhusiopathiae. A PCR amplification system has been developed for the rapid identification of E. rhusiopathiae DNA
in tissues.
Serologic Detection in the Host. Host serology is not useful for diagnosis of erysipelas.

INTRODUCTION brachial vein or the carefully exposed heart. For direct culture of
various internal organs (liver, spleen, kidney, lungs and bone
Erysipelas is an acute septicemic disease that occurs worldwide in marrow), wire culture loops or cotton-tipped swabs can be inserted
turkeys, chickens, waterfowl, numerous additional avian species, into tissues after flaming or searing die exposed surface. Solid
and in mammals, including humans. The disease is caused by the media should be streaked immediately, and the swabs may be
bacteria Erysipelothrix rhusiopathiae. Carrier animals and bacteria broken off into tubes of broth medium. Cultures of blood and
persisting in decomposing animal carcasses, meat or fish and their parenchymatous organs should be prepared from several birds to
processed products, ectoparasites, sewage, water, and soil present assure recovery during optimal stages of infection. Samples from
obvious sources of infection. Maturing male turkeys are decomposed cadavers or from organs likely to be contaminated with
predisposed to infection through skin lacerations inflicted by other bacteria should be directly cultured or collected in media
fighting, whereas the disease in mature turkey hens has been containing inhibitors such as erysipelothrix selective broth (ESB)
attributed to insemination with E. rhusiopathiae-conianahiated (7). This is also true of cultures of feces, soil, sewage, feeds, and
semen from carrier males. Erysipelothrix rhusiopathiae can cause a other materials which may be done as part of flock-or plant­
painful localized would infection (erysipeloid) in humans and is an monitoring programs.
occupational hazard for veterinarians and for people processing If specimens are to be shipped without being cultured on site, it is
fresh meat and fish. recommended that they be taken as soon as possible after death or,
preferably, from moribund birds. Livers, spleens, and bone marrow
CLINICAL DISEASE (unopened femur or tibia) are the best sources, but hearts and
affected joints might also be included from birds with subacute or
Among birds, male turkeys from 18-20 wk of age are most chronic infections. Specimens for shipping should be removed from
frequently affected, although the disease has been documented in cadavers as aseptically as possible, placed in sterile leak-proof
chickens, ducks, geese, pheasants, emus, and other birds. Wound containers, packed on ice or cold packs, and transported to the
infections associated with de-toeing procedures in day-old turkeys bacteriology laboratory as expeditiously as possible. Organ
have been reported as well. The onset of acute septicemic erysipelas specimens or joint exudates may be frozen if immediate shipment is
is sudden, with only a few hours of rapidly progressing signs not possible. Storage at 70 C is recommended.
(depression, somnolence, ruffled feathers, and prostration) before Erysipelothrix rhusiopathiae is remarkably persistent in
death. The lesions, apart from irregular areas of erythema and environmental specimens. The bacterium remains viable for up to 5
edema of infected skin, are those expected with bacterial septicemia days in water and up to 15 days in sewage. The bacterium persists
(i.e., generalized congestion with scattered serosal hemorrhages and for long periods in smoked or pickled meats and for up to 9 mo in
swelling of parenchymous organs). Purulent arthritis and vegetative buried carcasses. Erysipelothrix rhusiopathiae is killed by heating
valvular endocarditis may be seen in chronic erysipelas. The at 60 C for 15 min, and growth is inhibited by 10% (w:v) NaCl.
dominant microscopic lesions in acute erysipelas involve vascular Low temperatures and alkaline conditions in organic matter favor
damage, as evidenced by generalized congestion, edema, focal survival of the bacterium, whereas heat and direct sunlight diminish
hemorrhage, disseminated septic fibrin thrombi, and numerous its viability. Erysipelothrix rhusiopathiae can be preserved for
bacterial aggregates engulfed by cells of the macrophage- months by stab inoculation of screw-cap tubes of nutrient agar (pH
phagocytic system (1). Focal to diffuse necrosis may be seen in 7.4). After overnight growth, the cap is tightly closed and tubes are
liver and spleen. maintained in the dark at room or refrigerator temperatures.

SAMPLE COLLECTION PREFERRED CULTURE MEDIA AND SUBSTRATES

Handling of potentially infectious material can be hazardous in Samples from acutely affected birds should be inoculated onto
that even minor scratches and abrasions can become infected. The blood agar, McConkey’s agar, and triple sugar iron (TSI) agar for
use of rubber gloves, disinfectants, and sterile instruments is rapid preliminary isolation of E. rhusiopathiae as well as other
recommended. septicemic bacteria. Because specimens from carrier birds may
In birds with fulminant septicemia, E. rhusiopathiae is distributed contain few bacteria, isolation of E. rhusiopathiae will be enhanced
widely in blood and all parenchymatous organs, which are the by use of heavier inocula in enriched broth media (brain-heart
preferred sources of samples for isolation of the bacteria. Aseptic infusion broth with 5% serum). Isolation of E. rhusiopathiae from
collection of blood for culture can be achieved from either the potentially contaminated specimens (feed, intestine, cecal tonsils,
36
Chapter 9 Erysipelas

decomposed organs, and other materials) requires selective media medium (Andrade’s base with 10% rabbit serum). Running known
with inhibitory chemicals (5). For such isolation, ESB consisting of strains of E. rhusiopathiae for comparison with the test isolate is
tryptose broth with added antibiotics and Packer’s medium recommended. Lactose, glucose, levulose, and dextrin are usually
consisting of tryptose agar with sodium azide, crystal violet, and fermented; some acid production may be noted in sucrose,
serum are recommended (7). ESB medium should not be stored arabinose, dulcitol, mannitol, xylose, galactose, and fructose.
longer than 2 wk. Broth cultures should be inoculated 18-24 hr at 37 Hyaluronidase and neuraminidase are elaborated by many strains
C, and cultures on solid media should be incubated 24-48 hr. (4,10), and a recent report notes the consistent production of
Pure cultures of E. rhusiopathiae can be propagated successfully coagulase by numerous isolates tested (9). Vitex automated
in embryonated chicken eggs (6). Colonies can be picked directly systems, as well as API Coryne System (bioMerieux Vitek, Inc.,
from solid media with a bacteriologic needle that is then inserted Hazelwood, Mo.), a miniaturized biochemical test kit, can be used
into the yolk sac of embryonated eggs at 6-8 days of incubation. to identify E. rhusiopathiae (1).
Alternatively, 0.01 ml of pure broth culture can be delivered into
the yolk sac using a hypodermic needle. Embryos are killed by the Antigen Detection
bacterium within 24-48 hr. Harvested yolk is an excellent source of The definition of antigenic types of E. rhusiopathiae has been
high-titered bacterial inoculum and can be preserved for long primarily a research endeavor, largely because there is no
periods of storage at 70 C or by lyophilization. established relationship between serotype and immunogenicity or
Mouse inoculation can be used as a means of purifying mixed pathogenicity. From an epidemiological standpoint, however,
cultures and of assessing pathogenicity of E. rhusiopathiae isolates. maintaining a database on existing serotypes could be of value. The
To eliminate other bacteria, mice are exposed by swabbing scarified procedures for serotyping, including preparation of antigens and
ear skin with a cotton swab from broth cultures at 24, 48, and 96 hr antisera for the 26 known serotypes, are well established (3,4,8,10).
of incubation. Mice usually die 3-6 days after exposure, and E. One serologic procedure potentially critical to identification of
rhusiopathiae can be isolated in pure culture from heart blood or some E rhusiopathiae strains is the mouse protection test (6). Each
livers. Apparently, bacteria other than E. rhusiopathiae do not have of two or more mice are injected in the loose skin of the flank with
the ability to invade scarified skin of mice. For pathogenicity 0.1 ml of a 24 hr broth culture. The opposite flank is injected with
assessment, mice are inoculated subcutaneously or intraperitoneally 0.3 ml of equine hyperimmune anti-erysipelas serum (Anchor
with 0.1 ml of 24 hr broth culture; pathogenic isolates should kill Laboratories, St. Joseph, Mo.). Another group of mice receives the
mice within 6 days. Not all turkey isolates are pathogenic to mice, culture only. It is recommended that a third group of mice receive a
so pathogenicity may also need to be assessed directly in known mouse-virulent E. rhusiopathiae strain and antiserum. If the
susceptible turkeys by skin scarification or subcutaneous routes. test mice are killed by the unknown bacterium within a 6-day
observation period and are protected by specific erysipelas
AGENT IDENTIFICATION antiserum, one can conclude that the bacterium is E. rhusiopathiae.
A fluorescent antibody technique has been found to be useful as an
In stained smears from the serosal or cut surface of liver or spleen, aid in identifying E. rhusiopathiae isolates and in detecting
E. rhusiopathiae appears as a straight or curved rod (0.2-0.4 x 0.5- intestinal infection (6), but like other serologic procedures it is not
2.5 um) often occurring in clumps. The bacterium is Gram-positive widely applied in diagnostic laboratories. Fluorescein-labeled
but decolorizes readily. It has no capsule, does not form spores, and antibody to E. rhusiopathiae is not commercially available (4).
is non motile.
Molecular Identification
Cultural Characteristics A genus specific PCR amplification system has been used with pig
Although E. rhusiopathiae will grow aerobically, growth is samples for the rapid detection of E. rhusiopathiae. (2). Testing
enhanced if cultures are incubated in an atmosphere of 5%-10% methodologies utilized for the classification of different strains of E.
CO2 . After incubation of agar plates for 24-72 hr at 37 C, tiny rhusiopathiae include restriction fragment length polymorphism
pinpoint colonies progress to small, round, translucent 0.7-1-mm- (ribotyping) (2), and multilocus enzyme electrophoresis (MLEE)
diameter colonies. In mixed cultures these colonies can be easily (2). Erysipelothrix tonsillarum, the only other species in the genus
overshadowed by larger, more rapidly growing contaminant Erysipelothrix, differs from E. rhusiopathiae only by DNA-DNA
bacteria. On blood agar, a very narrow zone of greenish hemolysis homology. All strains belong to serovar 7.
frequently occurs around smooth colonies. In older cultures,
colonies may dissociate to flat, opaque, rough forms that are slightly SEROLOGICAL DETECTION IN THE HOST
larger than the smooth colonies. Bacterial morphology in rough
colonies is dramatically different from that noted above, in that the The absence of a widely applied serologic test for erysipelas in
cells become long, filamentous, and beaded, 4-15 pm in length, and birds suggests either a lack of demand or the unavailability of
often form tangled chains. In stab cultures in nutrient gelatin, E practicable testing technology. A tube agglutination test applied
rhusiopathiae forms a characteristic “bottlebrush” growth pattern primarily to research and epidemiology problems has been
along the stab line after 3-5 days of incubation, but gelatin is not described (6).
liquefied.
DIFFERENTIATION FROM CLOSELY RELATED AGENTS
Biochemical Properties
Erysipelothrix rhusiopathiae is catalase and oxidase negative, does The acute septicemic form of avian erysipelas might be confused
not produce indole, does not reduce nitrate, does not hydrolyze with a number of other acute infectious diseases, including fowl
esculin, and produces no change or weak acid in litmus milk, and cholera (pasteurellosis), staphylococcosis, colibacillosis,
most strains produce H2S in TSI agar. The latter change is one of streptococcosis, salmonellosis, and highly virulent avian influenza
the most reliable presumptive tests for the identification of E. or Newcastle disease. However, Gram staining of smears prepared
rhusiopathiae. TSI agar tubes should be fresh (not more than 2 wk directly from affected organs or from isolated bacterial colonies will
in storage); the butt is stabbed and the slant is streaked. The black clearly distinguish E. rhusiopathiae from most other septicemic
discoloration along the stab line is usually well developed by 24-48 bacteria. The only significant exception might be septicemic
hr, and there is an acid (yellow) butt and an acid slant. Carbohydrate listeriosis. Distinction of E. rhusiopathiae from Listeria
fermentation patterns are variable and require a standardized monocytogenes has been addressed in Chapter 10 on listeriosis.
37
George L. Cooper and Arthur A. Bickford

REFERENCES 6. Corstvet, R. Erysipelas, In: Isolation and identification of avian


pathogens, 2nd ed. S B. Hitchner, C.H. Domermuth, H.G. Purchase, and J.E.
1. Bickford, A.A, RE. Corstvet, and A S. Rosenwald. Pathology of Williams, eds. American Association of Avian Pathologists, College Station,
experimental erysipelas in turkeys. Avian Dis. 22:503-518. 1978. Tex., pp.23-26. 1980.
Σ Bille, J., J.R Rocourt, and B. Swaminathan. Listeria and Erysipelothrix, 7. Cottral, G.E. Manual of standardized methods for veterinary
In: Manual of Clinical Microbiology, 8th ed P.R. Murray, E.J. Baron, J.H. microbiology. Cornell University Press, Ithaca, N.Y. 1978.
Jorgensen, MA. Pfaller, and R.H. Yolken, eds. American Society for 8. Eamans, G.J., MJ. Turner, and R.E. Catt. Serotypes of Erysipelothrix
Microbiology, Washington, D.C. pp. 461-471. 2003. rhusiopathiae in Australian pigs, small ruminants, poultry and captive wild
3. Bisgaard, Μ, V. Norrung, and N. Tomoe. Erysipelas in poultry, birds and animals. Aust. Vet. J. 65:249-252. 1988.
prevalence of serotypes and epidemiological investigations. Avian Pathol. 9. Tesh, MJ., and R.L. Wood Detection of coagulase activity in
9:355-362. 1980. Erysipelothrix rhusiopathiae. J. Clin. Microbiol. 26:1058-1060. 1988.
4. Bricker, J.M, and Y.M Saif. Erysipelas, In: Diseases of Poultry, 11th ed. 10. Wood, R.L. Erysipelas, In: Diseases of swine, 6th ed A.D. Leman, B.
Y.M Saif, H.J. Barnes, J.R. Glisson, A.M. Fadly, L.R. McDougald, and Straw, R.D. Glock, W.L. Mengeling, RH.C. Penny, and E. School, eds.
DE. Swayne, eds. Iowa State University Press, Ames, Iowa. Pp. 845-862. Iowa State University Press, Ames, Iowa. Pp. 571-583. 1986.
2003.
5. Carter, G.R Diagnostic procedures in veterinary bacteriology and
mycology, 4th ed Charles C. Thomas, Springfield, Π1. 1984.

38
10
LISTERIOSIS
George L. Cooper and Arthur A. Bickford

SUMMARY. Listeria monocytogenes, the causal agent of listeriosis, grows well on most laboratory media, and can be cultured directly on
blood agar or blood agar with 0.05% potassium tellurite. Isolation of the organism directly from tissues is usually not difficult; however
isolation from the brain can be troublesome and may require culturing from the floor of the medulla. Listeria monocytogenes has been
subdivided into 13 serovars, based on the presence of different flagellar (H) and somatic (O) antigens, however, most cases of listeriosis are
caused by only three serotypes, l/2a, l/2b, and 4B.
Agent Identification. A definitive diagnosis of listeriosis usually requires the isolation and identification of the etiologic agent and the
demonstration of characteristic histologic lesions. Immunoassays that use monoclonal antibodies, DNA probe assays, and a PCR test are
available commercially for the rapid detection of L. monocytogenes in food products. L. monocytogenes can be detected in tissues (fresh or
paraffin block) by PCR-based tests, or direct immunofluorescence using specific monoclonal antibodies conjugated to fluorescein
isothiocyanate. Methodologies used to characterize the various strains of L. monocytogenes include phage typing, multilocus enzyme
electrophoresis, rRNA typing, DNA restriction enzyme analysis, pulsed-field gel electrophoresis, and random amplification of polymorphic
DNA.
Serologic Detection in the Host. Diagnostic serologic tests have not been reliable due to cross-reactions and the tendency of the organism
to autoagglutinate.

INTRODUCTION a glass tissue grinder at a ratio of 1:10 in a general nutrient broth


such as trypticase soy broth or brain-heart infusion broth (3).
Listeria monocytogenes has been isolated from a broad range of Inoculated broth media should be incubated at 35 C for 5-7 days
mammals, birds, and fish, and is widely distributed in nature. The and examined daily for growth.
bacterium is especially common in the temperate zones of both
hemispheres, where it can be found in soil, in animal feces, and on PREFERRED CULTURE MEDIA AND SUBSTRATES
vegetable matter such as silage. Although ordinarily thought of as a
disease of ruminants and humans, listeriosis has been well Samples can be cultured directly onto blood agar plates or blood
documented in domestic fowl, where it is generally considered as agar containing 0.05% potassium tellurite (which inhibits many
opportunist and is often associated with other disease conditions. gram-negative bacteria). Specimens can be held at temperatures of
Outbreaks are usually sporadic and mortality rates low, although 4 C for cold enrichment, and samples may be removed for culturing
losses of up to 40% have been reported in some chicken flocks. L. at weekly intervals for periods of up to 4 mo. The cold enrichment
monocytogenes has been recognized as a serious food bom technique is reportedly useful for culturing contaminated specimens
pathogen in humans and is of major public health concern and to the (3). Contaminated clinical specimens, feces, and food products can
food industry. Outbreaks have been traced to raw or undercooked be cultured with a two-stage enrichment procedure. The U.S.
poultry meat products which are considered a significant source of Department of Agriculture (USDA) method and the Netherlands
human infection (6). Government Food Inspection Service (NGFIS) methods are used
together at the Centers for Disease Control and Prevention to isolate
CLINICAL DISEASE L. monocytogenes from non-sterile clinical specimens and foods (2).
The USDA method involves enrichment of the specimen in
The most common avian hosts of L. monocytogenes are chickens, University of Vermont (UVM) primary selective enrichment broth
ducks, turkeys, geese, and canaries (1). Infection in other avian (1 part sample plus 9 parts of broth) for 24 hr at 30 C. 0.1ml of
species has been reported less frequently. All age groups are broth is then plated onto lithium chloride-phenylethanol-
susceptible, but the disease is primarily one of young birds, where it moxalactam (LPM) agar and Oxford or modified Oxford (MOX)
may cause a septicemic infection, with focal necrosis in the liver agar plates, and 0.1 ml of the primary enrichment broth is added to
and myocardium, pericarditis, and, occasionally, encephalitis. 10 ml of UVM secondary selective enrichment broth. The
Outbreaks of the encephalitic form have been seen in commercial secondary enrichment broth is incubated for 24 hr at 30 C before
broiler flocks, where the predominant clinical sign is torticollis (4). being subcultured onto selective agar plates. The NGFIS method
Lesions seen microscopically include perivascular cuffing, focal involves primary enrichment of the specimen in liquid polymyxin-
microabscesses in the midbrain and medulla, and gliosis and acriflavin-lithium-chloride-ceftazidime-esculin-mannitol
satellitosis in the cerebellum. In the septicemic form, multiple focal (PALCAM) egg yolk broth for 24-48 hr at 30 C. The secondary
hepatic abscesses and myocardial degeneration are common. enrichment broth is plated onto PALCAM agar plates and incubated
Salpingitis occurred in hens following the acute systemic phase of for an additional 24-48 hr under microaerobic conditions (5%
the infection. Bacteria may be found in Kupffer’s cells of liver or in oxygen, 7.5% hydrogen, and 80% nitrogen). LPM agar plates are
the periphery of brain lesions. examined with oblique lighting (Henry illumination), where Listeria
Infiltration of plasma cells, lymphoid cells, and macrophages are colonies appear blue and colonies of other bacteria appear yellowish
characteristic of the lesions (1). or orange. On Oxford and MOX agars, Listeria colonies appear
black due to hydrolysis of esculin, and the formation of black iron­
SAMPLE COLLECTION phenol compounds in the medium. On PALCAM agar, Listeria
colonies have a gray-green appearance. A chromogenic medium has
Listeria monocytogenes grows well on most laboratory media and been recently developed to help distinguish L. monocytogenes and
can be isolated readily from blood, but isolation directly from the L. ivanovii from other Listeria spp. on culture plates (5). Tests
brain, may be difficult. Culturing directly from the floor of the utilizing this medium include the BCM chromogenic agar test
medulla may increase the chances of isolation. Alternatively, tissue (Biosynth International, Naperville), and the CHROMagar Listeria
specimens may be macerated with a mortar and pestle or blended in test (Mast Diagnostics, Reinfeld, Germany). On chomogenic agar
39
George L. Cooper and Arthur A. Bickford

plates, colonies L. monocytogenes and L. ivanovii appear blue with Molecular Identification
a white halo, while other species of Listeria are either inhibited, or Based on DNA-DNA hybridization analysis and 16S rRNA
appear colorless, or blue without the halo (5). Suspect colonies can studies, Listeria have been divided into seven species (2).
be subcultured on nutrient agar slants. Pure cultures may be Listeria monocytogenes has been subdivided into 13 serotypes, or
lyophilized or stored indefinitely in defribrinated rabbit blood at 70 serovars, based on the possession of different flagellar (H) and
C. somatic (O) antigens. More than 90% of all cases of listeriosis
reported worldwide in humans and animals are produced by
AGENT IDENTIFICATION serotypes 4B, l/2a, and l/2b. A commercial kit is available for
serotyping Listeria isolates (Denka Seiken, Tokyo, Japan), and
Morphology antisera for serotyping can be obtained from Difco (Difco
Listeria monocytogenes is a slender, non-spore forming, gram­ Laboratories/ Becton Dickinson and Co., Sparks, MD) (2). Other
positive, rod-shaped bacterium. In older or rough cultures, long methods used for typing L. monocytogenes include phage typing,
filaments 6-20 pm in length may develop. In young cultures the multilocus enzyme electrophoresis (MLEE), ribotyping, random
organism may appear as a coccobacillus. In stained preparations fragment length polymorphism (RFLP), and pulsed-field gel
the organisms may have a diphtheroid appearance. electrophoresis (PFGE) (5). An automated RiboPrinter System
(Qualicon, Wilmington, Del.) facilitates rapid and easy subtyping of
Biological properties L. monocytogenes, and the CDC has established a network of public
Listeria monocytogenes grows at a temperature range of 4-42 C, health laboratories that routinely subtype food-borne pathogens
with an optimum of 35 C. The organism is somewhat resistant to using PFGE pattern analysis (PFGE@cdc.gov) (2).
heat but is killed in 60 min at 55 C. It can grow in 10% salt at pH The rapid detection of Listeria spp. in food samples from selective
9.6 and can survive in 20% salt. It can survive for years at 4 C in enrichment broths can be facilitated by immunoassays using
broth and survives for long periods of time in the environment on monoclonal antibodies, such as Listeria Tek (Organon-Teknika,
silage, hay, or in the soil. The organism will grow aerobically, but Tumhoout, Belgium) and VIDAS Listeria Assay (bioMerieux,
the addition of 5%-10% CO2 to the atmosphere enhances growth. Marcy-Etoile, France). These tests are genus specific, whereas
Positive motility can be demonstrated by an umbrellalike growth DNA probes such as L. monocytogenes assay (Gene-Trak,
pattern produced along the stab line in motility test medium. The Naperville, Iowa), and Accuprobe (Gen-Probe Inc., San Diego,
end-over-end tumbling motion that is characteristic of Listeria Calif.) are specific for L. monocytogenes (2). Commercially
monocytogenes can best be demonstrated in a hanging drop available PCR tests used for testing food products include the BAX
preparation of a 2-hr broth culture incubated at room temperature PCR system (Qualicon, Wilmington, Del.), and the Probelia assay
(25 C). After 24-48 hr of growth, small, smooth, gray translucent (Sanofi Diagnostic Pasteur, Marne la Coquette, France). Real-time
colonies, 0.2-0.4 mm in diameter, are formed, and on blood agar a PCR tests are being developed to identify L. monocytogenes in food
narrow zone of beta hemolysis is usually produced. On clear media, and clinical samples, with the added advantage of being able to
such as LPM, the colonies appear blue (small young colonies) to quantify the bacteria as well (5). Listeria monocytogenes can be
white (older colonies) when viewed with an oblique light source (45 detected in tissues (either fresh or in paraffin blocks) by PCR-based
degree angle). Turbidity is produced in broth cultures, which tends tests, and by a direct immunoflorescence test using specific L.
to settle on standing. monocytogenes monoclonal antibodies conjugated to Fluorscein
isothiocyanate. (2). A 30-min chemiluminescence DNA probe
Physicochemical properties (Gen-Probe, San Diego, Calif.) is available for rapid confirmation
Acid without gas is produced from glucose, maltose, lactose, of L. monocytogenes from colonies on primary isolation plates (2).
sucrose, trehalose, rhamnose, and salicin. No fermentation of
arabinose, dulcitol, inulin, raffinose, or xylose occurs. The organism Table 10.1 Characteristics to differentiate Listeria species.
is usually methyl red-positive, Voges-Proskauer-positive, catalase­
positive, esculin-positive, and produces a positive reaction on the Type of
Rhamnose
Listeria species Mannitol Xylose Nitrate
Christie, Atkins and Muench-Peterson (CAMP) test with Hemolysis
Staphylococcus aureus. B-lysin discs (Remel, Lenexa, Kans.) can +
L. monocytogenes β - - -
be used for enhancement of L. monocytogenes hemolysis. The
bacterium is urease and gelatinase-negative, and no H2S produced L. ivanovii β - - + -
in die butt of triple sugar iron agar. Several miniaturized
L. innocua β - +/- - -
biochemical test kits are available commercially for identifying
Listeria spp., and include M *\-Listeria test (bioMerieux-Vitex, Inc., L. seeligeri β - - + -
Hazelwood, Mo.) and Micro ID Listeria (Organon-Teknika,
L. welsh β - +/- + -
Tumhoout, Belgium).
Mice, rabbits, and guinea pigs are susceptible to experimental L. grayi β + - - -
infection, and have been used to isolate the organism and to
L. murrayi β + +/- - +
evaluate pathogenicity. The Anton test has been used as a
presumptive test, wherein a purulent conjunctivitis occurs 24-36 hr
after the instillation of a drop of broth culture in the conjunctival sac
SEROLOGIC DETECTION IN THE HOST
of a rabbit or guinea pig. An immunocompromised-mouse model
has been developed in which relatively small numbers of virulent
Diagnostic immunologic tests have been used with various degrees
Listeria can cause death within 3 days, and a cell culture
of success. These include hemagglutination-inhibition, complement
cytotoxicity assay using the human intestinal epithelial cell line
fixation, gel diffusion and precipitation. The tendency of the
Caco-2 has been developed to determine the virulence of Listeria
organism to autoagglutinate, and problems with cross reaction have
isolates in vitro (2). The organism can also be isolated in 10-day-old
limited the usefulness of many serologic tests. Cross reactions are
chicken embryos inoculated via the allantoic sac.
common with Staphylococcus aureus, some group D Streptococcus,
and also some gram-negative organisms such as coliforms.

40
Chapter 10 Listeriosis

DIFFERENTIATION FROM CLOSELY RELATED AGENTS which is often isolated from mixed flora specimens on selective
media, from some strains of L. monocytogenes, which may produce
The systemic form of listeriosis may resemble many other only faint or narrow zones of hemolysis. Erysipelas is also caused
septicemic diseases, such as staphylococcosis, streptococcosis, by a slender gram-positive rod-shaped bacterium, but unlike L.
erysipelas, salmonellosis, pasterellosis, colisepticemia, and other monocytogenes, Erysipelothrix rhusiopathiae produces hydrogen
bacterial or viral infections in which nonspecific findings such as sulfide, does not produce catalase, and gives a negative reaction on
depression and acute mortality are often seen. A differential the Anton test.
diagnosis of the neuologic form of the disease should include avian
encephalomyelitis; Newcastle disease; encephalomalacia; Marek’s REFERENCES
disease; chronic localized Pasteurella multocida infection of the
1. Barnes, H.J. Miscellaneous and Sporadic Bacterial Infections, In:
middle ear, meninges, and cranial bones; and the neurologic form of
Diseases of Poultry, 11th ed., Y.M Saif, H.J. Barnes, J.R. Glisson, A M
aspergillosis. The histologic lesions seen in the brain are suggestive Fadly, L.R. McDougald, and D.E. Swayne, eds. Iowa State University Press,
of listeriosis, and isolation and identification of the etiologic agent Ames, Iowa. Pp. 845-862. 2003
should confirm the diagnosis. Listeria monocytogenes is sometimes 2. Bille, J., J.R. Rocourt, and B. Swaminathan. Listeria and Erysipelothrix,
overlooked on culture plates when it is confused with In: Manual of Clinical Microbiology, 8th ed. P.R. Murray, E.J. Baron, J.H.
nonpathogenic “diphtheroid” organisms, Lactobacillus, Brochothrix Jorgensen, MA. Pfaller, and R.H. Yolken, eds. American Society for
spp., Kurthia spp., or with a beta hemolytic Streptococcus. Listeria Microbiology, Washington, D.C. Pp. 461-471. 2003.
monocytogenes is easily discolorized and can be confused with a 3. Carter, G.R. Diagnostic procedures in veterinary bacteriology and
mycology, 4th ed. Charles C. Thomas, Springfield Ill. Pp 191-195. 1994.
gram-negative bacillus. A positive catalase reaction will separate L.
Cooper, G.L. An encephalitic form of listeriosis in broiler chickens. Avian
monocytogenes from the streptococci, and the positive motility and Dis. 33:182-185. 1989.
the ability to hydrolyze esculin should separate it from the 4. Gasanov, U., D. Hughes, and P.M Hansbro. Methods for the Isolation
corynebacteria. Lactobacillus spp. generally are nonmotile and and Identification of Listeria Spp. and Listeria monocytogenes: A Review.
catalase-negative. Brochothrix spp. is unable to grow at 37 C, and FEMS Microbiology Review 29(5), 851-75. 2005.
Kurthia spp. are strictly aerobic and esculin negative. The CAMP 5. Gray, ML., and A.H. Killinger. Listeria monocytogenes and Listeria
test is useful for differentiating nonhemolytic Listeria innocua, Infections. Bacteriol. Rev. 30:339-382. 1966.

41
11
STAPHYLOCOCCOSIS
Stephan G. Thayer and W. Douglas Waltman

SUMMARY. Staphylococcosis is a common disease condition in both chickens and turkeys. It is almost always associated with a break in
the skin or mucus membrane or with immunosuppression.
Agent Identification. The causative agent, Staphylococcus aureus, is easy to isolate using almost any bacterial culture media but 5% sheep
or bovine blood agar is preferred. Staphylococcus aureus produces circular smooth colonies that are 1-3 mm in diameter within 18-24 hr
with most strains being β-hemolytic and pigmented white to yellow. Gram-staining yields gram positive cocci in grapelike clusters.
Staphylococcus aureus is catalase positive. S. aureus can be identified with the latex agglutination test.
Serologic Detection in the Host. There is no serologic test available to diagnose Staphylococcus infection.

INTRODUCTION is some evidence that certain major histocompatibility B complex


genotypes may play a role in susceptibility to staphylococcal
In recent years, staphylococcosis has become one of the most skeletal disease (6).
important bacterial diseases of poultry. This has resulted from the
common practice of growing chickens and turkeys to a greater SAMPLE COLLECTION
weight because of product demands for further processing.
Chickens and turkeys also have been highly selected for their ability Specimens from which staphylococci are cultured are blood,
to achieve rapid growth. Such selection has added to stress by exudate from swollen joints or foot pads, yolk, and swab stabs from
creating various leg problems that predispose poultry to the internal organs such as liver, spleen, and kidneys. In the field,
development of staphylococcosis. exudate from swollen joints can be conveniently collected by
Historically, staphylococcosis has been a significant problem arthrocentesis using a 1-ml syringe and a 20 to 26-gauge needle;
because of the ubiquitous nature of the bacterium in the poultry care must be taken to first disinfect the skin where the needle is
farm environment. The portals of entry start with the open navel of introduced. No special precautions are needed beyond standard
newly hatched chicks from contaminated hatchers and infection is procedures in handling and transporting specimens, as
associated with contaminated hatching eggs. The sources of staphylococci are very hardy.
infection progress to wounds associated with beak trimming, toe
trimming, toe scratches, wood splinters, and sharp metal edges, with PREFERRED CULTURE MEDIA AND SUBSTRATES
increasing opportunities for injury as birds rapidly gain weight.
Numerous Staphylococcus spp. are found in the poultry Staphylococci grow readily on ordinary culture media. Specimens
environment, but S. aureus is the most frequent disease causing from internal organs or fluids, where background contamination
agent. Staphylococcus spp. are normal inhabitants of skin and would normally be low, can be inoculated directly onto sheep or
mucus membranes and readily reside in all the environments where bovine blood agar plates. The blood used in the agar must be free
poultry are hatched, reared, and even processed. Staphylococcus of antibiotics. Culture materials taken from external tissues or the
aureus definitely can cause disease, but it, along with other environment, where heavy background contamination exists, should
Staphylococcus spp. especially S. epidermidis, is thought to be inoculated onto selective media. Phenyethanol (PEA), Mannitol-
suppress other possible pathogens through interference or salt agar or Staphylococcus medium 110 (BD Diagnostic Systems,
competitive exclusion. Other Staphylococcus spp. that have been Sparks, MD) are commonly used, and Mannitol salt agar and Staph
isolated from lesions or possible disease situations include S. hyicus medium 110 function as both a selective and differential medium
(fibrinoheterophilic blepharitis) (4) and S. cohnii (liver spots) (13). for S. aureus. Other selective media that might be used are
Columbia CNA agar or tellurite glycine agar (BD Diagnostic
CLINICAL DISEASE Systems, Sparks, MD).

Staphylococcosis is a term used to describe any of several clinical AGENT IDENTIFICATION


conditions in poultry where S. aureus is the causative agent.
Infections occur when there is a disruption of the skin or a mucus A rapid tentative identification can be made by spreading a thin
membrane allowing entrance or penetration of S. aureus into deeper film of material from synovial fluid or tissues such as the liver
tissues. Staphylococcus aureus infections have been observed most (impression smear) directly onto a microscope slide and observing
frequently in the bones, tendon sheaths, and joints, but infections for gram-positive cocci. Diagnosis is confirmed by isolating
are also observed, although less frequently, in the skin, sternal staphylococci (S. aureus) from tissues with lesions. In most cases
bursa, yolk sac, heart, vertebrae, eyelids, and as granulomas in the where classical lesions of staphylococcosis are present, S. aureus is
liver and lungs. In older birds, usually in lay, S. aureus can cause a readily isolated in large numbers from the involved tissues.
septicemia with acute deaths (1). On agar cultures, staphylococci produce 1 to 3 mm diameter,
The most common important clinical conditions associated with opaque, smooth, circular, entire, raised, colonies in 18-24 hr.
staphylococcosis are osteomyelitis, arthritis, periarthritis, and Staphylococcus aureus isolates are mostly hemolytic but
synovitis. The bones most frequently involved are the proximal occasionally non-hemolytic at 24 hr on blood agar. Non-hemolytic
tibiotarsus and proximal femur. Less commonly involved sites are can be a misinterpretation of colonies with weak hemolysis hidden
the proximal tarsometatarsus, distal femur, distal tibiotarsus, behind the colony. Avian isolates are frequently white developing
proximal humerus, ribs, and vertebrae. In affected birds, the cream to yellow pigmentation after 48 hr or more. On mannitol-salt
femoral head often separates from the shaft by a fracture through agar, S. aureus colonies are surrounded by a yellow halo indicating
the neck of the femur when the coxofemoral joint is disarticulated mannitol fermentation. Typical colonies are examined
(femoral head necrosis). Grossly affected bones have focal yellow microscopically to confirm that they are composed of gram-positive
crumbly areas that contain caseous exudate. Affected joints are staphylococci. The latex agglutination test (Remel, Inc. Lenexa,
swollen and filled with white to yellow exudate (1, 7, 8, 11). There KS) is sufficient, in most cases, to differentiate S. aureus from other
42
Chapter 11 Staphylococcosis

Staphylococcus species. Coagulase testing can be accomplished others), viruses (viral arthritis), and nutritional deficiencies. Other
using commercially available desiccated rabbit plasma containing bacteria and molds are also responsible for hatchery related
either citrate or ethylenediaminetetracetic acid. A rapid slide infections. Staphylococcal septicemia can resemble any bacterial
coagulase test can be performed by making a heavy, smooth septicemic condition of poultry, including fowl cholera,
suspension of a colony in one drop of reconstituted plasma on a colibacillosis (E. coli), and fowl typhoid (Salmonella gallinarum).
glass slide and observing for clumping in 10 sec; this detects the To differentiate Staphylococcosis from these diseases the laboratory
clumping factor (cell-bound coagulase). A tube coagulase test is testing described in this chapter is essential.
more accurate than the rapid slide test and detects extracellular
(free) coagulase. The tube test uses 0.5 ml of reconstituted plasma REFERENCES
that is thoroughly mixed with either 0.1 ml of an overnight broth
culture or a portion of a well isolated colony. The mixture is 1. Andreason, Claire B. Staphylococcosis. In: Diseases of poultry, 11th ed
Y. M Saif, H. J. Bames, J. R. Glisson, A.M Fadly, L. R. McDougald, and
incubated for 4 hr at 37 C, and any degree of clotting represents a
D.E. Swayne, eds. Iowa State Press, Ames, Iowa. pp. 798-804. 2003.
positive test. Since S. aureus, S. hyicus, and S. intermedius are all 2. Boynukara, B., K. Gurturk, T. Golhan, I. H. Ekin, and E. Ogun.
positive for coagulase there is still a need for a test specific for S. Comparison of latex agglutination test with protein A, clumping factor, and
aureus. Commercial latex agglutination tests for S. aureus have coagulase tests for identification of staphylococci isolated from avian. Eur.
been found specific and reliable when used for the identification of J. of Med 4:58-60. 1999.
avian isolates (2, 5, 14). 3. Butterworth, A. Infectious components of broiler lameness: A review.
When latex agglutination positive (coagulase-positive) World’s Poultry Science Journal. 56:327-352.1999
staphylococci are isolated in large numbers from birds presenting 4. Cheville, N. F., J. Tappe, M Ackermann, and A. Jensen. Acute
fibrinopurulent blepharitis and conjunctivitis associated with
typical signs of staphylococcosis, a diagnosis is confirmed.
Staphylococcus hyicus, Escherichia coli, and Streptococcus sp. in chickens
Isolation of low numbers of S. aureus is not always indicative of and turkeys. Vet. Pathol. 25:369-375. 1988.
staphylococcosis since they are part of the normal flora of healthy 5. Doem, G.V. Evaluation of commercial latex agglutination test for
birds. identification of Staphylococcus aureus. J. Clin. Microbiol. 15:416-418.
Various species of coagulase-negative staphylococci can be 1982.
isolated from poultry. Occasionally these may appear as 6. Finegold, S. M, and E. J. Baron. Micrococcaceae: Staphylococci,
opportunists when they can be isolated repeatedly from the same Micrococci, and similar organisms. In: Bailey and Scott's diagnostic
flock however none of these species have been incriminated as a microbiology, 10th ed B. A. Forbes, D. F. Sahm, and A. Weissfeld, eds. C.
V. Mosby Co., St. Louis, Mo. pp. 606-618. 1999.
cause of major disease in poultry. Should it be necessary to identify
7. Glisson, J. R. , and J. A. Smith. Staphylococcal synovitis in broiler
these staphylococci, the following scheme can be used: breeders. In: Proceedings of the avian skeletal disease symposium. 127th
1) Isolate onto blood agar. Employ selective medium such as PEA Annual Meeting of the American Veterinary Medical Association/American
or mannitol salt agar where necessary to isolate from contaminated Association of Avian Pathologists. San Antonio, TX. 1990.
specimens. Colonies from selective media need to be subcultured 8. Joiner, K.S., F.J. Hoerr, E. Van Santen, and S.J. Ewald The Avian Major
onto TSA/blood agar before testing. Histocompatibility Complex Influences Bacterial Skeletal Disease in Broiler
2) After confirmation of Gram positive cocci, test for the Breeder Chickens. Vet. Pathol. 42:275-281. 2005.
production of catalase by picking 24-hr colonies from TSA and 9. MacFaddin, Jean F. Biochemical tests for the identification of medical
bacteria. 3rd edition. Lippincott, Williams and Wilkins, Baltimore, MD.
immersing the bacteria in a drop of 3% H2O2 on a glass slide.
2000.
Observe for vigorous bubbling indicating breakdown of H2O2 and 10. Mannerman, Tammy L. Staphylococcus, Micrococcus, and other
release of oxygen gas. Catalase-positive cultures are of the family catalase positive cocci that grow aerobically. In: Manual of clinical
Micrococcaceae. microbiology, 8th ed Patrick R. Murray, Ellen Jo Barron, James H.
3) Test for sensitivity to lysostaphin (Sigma Chemical, St. Louis, Jorgensen, Michael A. Pfaller, and Robert H. Yolken. eds. American
MO) by placing 5 drops of lysostaphin solution into 5 ml of a 24 Society for Microbiology, Washington, D.C. pp. 384-404. 2003.
hour heart infusion broth culture of the suspect catalase positive 11. Mutalib, A., C. Riddell, and A. D. Osborne. Studies on the
cocci. Add 5 drops of PBS (control) to a second suspension. pathogenesis of staphylococcal osteomyelitis in chickens. I. Effect of stress
on experimentally induced osteomyelitis. Avian Dis. 27:141-156. 1983.
Incubate at 35C for up to 2 hr. Observe at 30, 60 and 120 min.
12. Naim, Μ E. Bacterial osteomyelitis and synovitis of the turkey. Avian
Clearing of the suspension is a positive test indicating sensitivity to Dis. 17:504—517. 1973.
lysostaphin. Turbidity indicates insensitivity. Species of the genus 13. Norton, R. A., G. R. Bayyari, J. K. Skeeles, W. E. Huff, and J. N.
Staphylococcus are sensitive to lysostaphin, and species of the Beasley. A survey of two commercial turkey farms experiencing high levels
genus Micrococcus are resistant to it. of liver foci. Avian Dis. 38:887-894. 1994.
4) Confirm S. aureus identification with one of the latex 14. Roberson, J. R., L.K. Fox, D.D. Hancock, T.E. Besser. Evaluation of
agglutination tests since coagulase-positive isolates can be S. Methods for differentiation of coagulase-positive staphylococci. J. Clin.
aureus, S. hyicus, or S. intermedius. Microbiol. 30:3217-3219. 1992.
5) Test latex-negative and coagulase-negative cultures with a
battery of biochemical tests to identify the species. Several
commercial microassay kits are available that include the API
Staph, ID 32 Staph and RAPIDEC Staph (bioMerieux-Vitek,
Hazelwood, Mo.).
More extensive coverage of the above diagnostic procedures is
published elsewhere (2, 5, 9, 10, 14).

SEROLOGIC DETECTION IN THE HOST

Serologic tests are not used for the diagnosis of staphylococcosis.

DIFFERENTIATION FROM CLOSELY RELATED AGENTS

Lameness and leg disorders have several causes in poultry. These


include other bacteria (Escherichia coli, Mycoplasma synoviae, and
43
12
STREPTOCOCCOSIS AND ENTEROCOCCOSIS
Stephan G. Thayer and W. Douglas Waltman

SUMMARY. Streptococcosis is caused by Streptococcus zooepidemicus from the Lancefield type C antigenic serogroup and Enterococcus
spp. reclassified from the Lancefield type D streptococcal antigenic serogroup. Streptococcus zooepidemicus most commonly affects mature
chickens. The Enterococcus spp. are commonly referred to as fecal streps and include E. faecalis, E. /aecium, E. durans, E. avium and E.
hirae. Enterococcus spp. can affect almost all avian species. Disease caused by both bacterial groups can be either acute, subacute, or
chronic. Septicemia, omphalitis, arthritis, salpingitis, valvular endocarditis, myocarditis, and osteomyelitis are common lesions associated
with infection.
Agent Identification. Blood and MacConkey’s agar and other selective media can be used to isolate these organisms. Differential
characteristics can be identified using selective media, hemolysis on blood agar, and fermentation of mannitol, L-arabinose, and sorbitol.
Differential diagnosis of streptococcal and enterococcal infections should include staphylococcus, colibacillosis, pasteurellosis, and
erysipelas.
Serologic Detection in the Host. Serologic techniques are not commonly employed to detect infection by streptococci in poultry.

INTRODUCTION less commonly, infarcts in the lung, kidney, and brain. Lesions in
the heart valves consist of fibrin, bacteria, heterophils,
Streptococcosis of avian species is worldwide in distribution macrophages, and fibroblasts. Other microscopic lesions related to
occurring as acute septicemic or chronic infections with mortality endocarditis include cerebral vasculitis and infarcts;
ranging from 0.5% to 50% (19). Streptococci are ubiquitous in leptomeningitis; glomerulonephritis; and thrombosis of pulmonary
nature and are commonly found in poultry environments. These vessels (13). Focal granulomas may be observed in any tissue due
organisms also are part of the normal intestinal flora of poultry and to septic thrombi. Liver infarcts are characterized by portal venous
wild birds. Streptococcoses are considered to be secondary thrombosis and necrosis. Molecular tools such as 16S rRNA
infections (21). sequencing offer discriminatory power not seen in conventional
The application of new bacteriologic techniques, especially DNA- biochemical approaches. The result is* description of new species
DNA and DNA-rRNA hybridization, has led to the reclassification such as S. gallinaceus. It is considered an opportunistic pathogen
of many bacterial species, including Streptococcus spp. (10, 17). which is associated with septicemia (5, 6). Another new species S.
The most common Streptococcus spp. associated with avian disease gallolyticus is associated with septicemic disease in pigeons.
is Streptococcus zooepidemicus, from the Lancefield antigenic Enterococcus spp. affect avian species of all ages. The most
serogroup type C. The Lancefield serogroup D has been serious infections cause early embryonic death and mortality of
reclassified as Enterococcus spp. Members of this group are found very young birds. Transmission can be by oral, aerosol, and egg
as opportunists in the intestinal tract. Potential pathogenic transmission routes (3, 9 ). Entry may also occur through skin
enterococci in avian species are Enterococcus faecalis), E. faecium, injuries. Enterococcus species such as E. faecalis, E. faecium appear
E. durans, and E. avium (5). Enterococcus hirae has been identified to be the most common with other species showing occasional
as the cause of growth depression in young chickens but has not involvement. Enterococcus durans has been reported with
been associated with embryo mortality or disease in young chicks bacteremia and encephalomalacia in young chickens 4 to 10 days of
(1,6). age from two separate flocks (4, 22).

CLINICAL DISEASE SAMPLE COLLECTION

Streptococcus zooepidemicus infection occurs almost exclusively Preferred tissues for collection include liver, spleen, blood, yolk,
in mature chickens, but may cause mortality in wild birds (11). embryonic fluids, subcutaneous fluid, and material from suspected
With acute S. zooepidemicus infections, clinical signs include lesions. Such samples usually yield large numbers of organisms,
lassitude, blood-stained tissue and feathers around the head, yellow often in pure culture. Cultures should be taken from clinically
droppings, emaciation, and pale comb and wattles. In some cases, affected birds immediately after euthanasia. Embryo fluids,
the presence of dead birds may be the only sign. especially yolk, are preferred specimens for culturing unpipped
Gross lesions of acute streptococcosis include splenomegaly, eggs. Culture of pipped eggs is to be confined to yolk and body
hepatomegaly (with or without red, tan or white foci; the latter tissue and/or fluids. Embryonic pipped egg samples should be
being miliary to 1 cm in diameter), enlarged kidneys, subcutaneous aseptically taken as soon after hatch as possible. Sterile swabs
congestion, serosanguinous subcutaneous and/or pericardial fluid, should be used to obtain adequate quantities of material for culture.
and peritonitis. Omphalitis is observed in chicks and poults. Best results are obtained by immediately streaking swabs on blood
Microscopically the liver has dilated sinusoids with increased agar after sample collection. If swabs remain uncultured for more
intrasinusoidal heterophils. Thrombosis, infarction, and multiple than 2 hr, they should be incubated in a general purpose broth (heart
areas of necrosis with heterophil accumulation are observed if gross infusion, trypticase soy (BD Diagnostic Systems, Sparks, Md.),
liver foci are present. Splenomegaly is characterized by congestion tryptose, etc.) for 2-^ hr before being streaked onto blood agar. If
and hyperplasia of the macrophage-phagocytic system (13). With increased bacterial numbers are desired, overnight incubation in
subacute or chronic streptococcosis, signs include loss of body nutrient broth is recommended. Swabs to be processed the
weight, lameness, pyrexia, and occasional head tremors. Gross following day should be transported in a silica gel or a dry filter
lesions may include fibrinous arthritis, tenosynovitis, salpingitis, transport system (10) (Carter, Rice, Storrs & Bennett, East Hartford,
pericarditis, and peritonitis; necrotic myocarditis; and valvular Conn.). Streptococcal cultures may be stored on tightly capped agar
endocarditis. slants at 4 C (several months), stored in blood at -70 C (1-2 yr), or
Osteomyelitis has been observed in turkeys. Associated with lyophilized (20 yr).
valvular endocarditis are infarcts of the liver, spleen, and heart, and,

44
Chapter 12 Streptococcosis and Enterococcosis

Table 12.1 Differential Characteristics of Streptococci and Enterococci from poultry (9,11,16,18,19, 21).

Growth on FermentsM Ferments


Identification Hemolysis Ferments Sorbitol
*MacConkey annitol L-arabinose
E. avium α/γ + /- + + +
E. durans
** α /γ + /- - - -
E. faecalis α/γ + /- + - +
E. faecium a /γ + /- + + -
***
E. hirae α/γ + /- - - -
S. equi ss. zooepidemicus β - - + +
* Literature - without / with crystal violet
** Sucrose (-), Raffinose (-)
*** Sucrose (+), Raffinose (+)

PREFERRED CULTURE MEDIA AND SUBSTRATES Serologic Identification


For positive identification of streptococci, one must conduct
Blood agar, MacConkey’s agar, or a selective medium with 6.5% group-specific precipitin, agglutinin, and/or fluorescent antibody
NaCl combination of equal ionic strength can be used for primary tests. Several descriptions of these tests have been published (10).
or secondary culture (7, 10, 11). The selective medium will also A latex agglutination test for rapid detection of Lancefield Group C
favor growth of staphylococci that can be differentiated by the streptococci has been developed that can be used for identification
catalase test. Another selective medium for enterococci is the use of 5. zooepidemicus (14).
of bile esculin medium (BD Diagnostic Systems, Sparks, Md.) on
which enterococci form a dark brown to black pigment after 24 h of SEROLOGIC DETECTION IN THE HOST
incubation at 35 C. Listeria monocytogenes and some of the
Enterobacteriaceae spp. can display the same reaction on esculin Serologic techniques are not commonly employed to detect
medium (9, 17, 18, 19). Group D streptococci can be further infection by streptococci in poultry.
differentiated by their ability to grow at 45 C and by being
thermostable at 60 C for 30 min. DIFFERENTIATION FROM CLOSELY RELATED AGENTS

AGENT IDENTIFICATION Differential diagnosis should exclude other bacterial septicemic


diseases, such as staphylococcosis, colibacillosis, pasteurellosis, and
Streptococci and Enterococci are nonmotile facultative anaerobes. erysipelas.
In gram-stained histopathologic sections and/or gram-stained tissue
and blood smears, the streptococci are non-spore-forming, gram­ REFERENCES
positive, spherical bacteria occurring singly, in pairs, or as short
1. Abdul-Aziz, T. A. Pathogenicity of Enterococcus hirae for chicken
chains. For routine diagnostic purposes, S. zooepidemicus and the
embryos and betamethazone-treated chicks. Res. Vet. Sci. 56:397-398.
enterococci can be identified on the basis of colony and microscopic 1994.
morphology, gram-stain reaction, a negative catalase reaction, and 2. Baele, M, L. A. Devriese, P. Butaye, andF. Haesebrouck. Composition
difference in the type of hemolysis on blood agar. Streptococcus of enterococcal and streptococcal flora from pigeon intestines.
zooepidemicus is β-hemolytic and enterococci are a- or γ-hemolytic. J. of Appl. Microbiol. 92:348-351.2002
Enterococci can be differentiated into species on the basis of growth 3. Brittingham, M C., S. A. Temple, and R. M Duncan. A survey of the
or no growth on MacConkey’s agar and fermentation of mannitol, prevalence of selected bacteria in wild birds. J. Wildl. Dis. 24:299-307.
sorbitol, and L-arabinose (Table 12.1) (17, 19, 20, 22). Colonies 1988.
4. Cardona, C. J., A. A. Bickford, B. R. Charlton, and G. I. Cooper.
are small and usually grayish in 24 hr and can appear from mucoid Enterococcus durans infection in young chickens associated with bacteremia
to rough. Enterococci may or may not grow on MacConkey, and encephalomalacia. Avian Dis. 37:234-239. 1993.
depending on the formulation. Several formulations exist and those 5. Chadfield, M S., J. P. Christensen, H. Christensen, and M Bisgaard
with crystal violet will prevent the growth of enterococci. Characterization of streptococci and enterococci associated with septicaemia
Another approach is the application of the Strep ID triplate in broiler parents with high prevalence of endocarditis. Avian Pathol.
(Remel, Inc, Lenexa, KS.) which includes blood agar, bile esculin 33:610-617. 2004
agar, and PYR agar. The combination of Gram stain, catalase 6. Chadfield, M S., J. P. Christensen, J. Juhl-Hansen, H. Christensen, and
reaction, hemolytic character, bile esculin reaction and PYR M Bisgaard Characterization of Enterococcus hirae outbreaks in broiler
flocks demonstrating increased mortality because of septicemia and
reaction will identify most avian streptococci to the Lancefield endocarditis and/or altered production parameters. Avian Dis. 49:16-23.
grouping. It must be emphasized that the characterization and 2005
differentiation described are presumptive diagnostic procedures. 7. Chapin, Kimberle C., and Patrick R Murray. Media. In: Manual of
More comprehensive identification can be accomplished through clinical microbiology, 7th ed P. R Murray, E. J. Baron, M A. Pfaller, F. C.
the use of additional biochemical tests or commercial systems. Tenover, and RH. Yolken, eds. American Society for Microbiology Press,
Washington, D.C. pp. 297-305. 1999.
8. Devriese, Luc A., P. Vandamme, B. Pot, M Vanrobaeys, K. Kersters,
and F. Haesebrouck. Differentiation between Streptococcus gallolyticus
strains of human clinical and veterinary origins and Streptococcus bovis
strains from the intestinal tracts of ruminants. J. Clin. Microbiol. 36:3520-
3523. 1998.
9. Domermuth, C. H., and W. B. Gross. A medium for isolation and
tentative identification of fecal streptococci, and their role as pathogens.
Avian Dis. 13:394-399. 1969.

45
Stephan G. Thayer and W. Douglas Waltman

10. Farrow, J. A. and M D. Collins. Enterococcus hirae, a new species that 17. Quinn, P. J., Μ E. Carter, B. K. Markey, and G. R. Carter Streptococci
includes amino acid assay strain NCDO 1258 and strains causing growth and related cocci. In: Clinical Veterinary Microbiology. Wolfe Publishing-
depression in young chickens. Int. J. Syst. Bacteriol. 35:73-75. 1985 Mosby Yearbook Limited. London. 1994.
11. Farrow, J. A., E. D. Jones, B. A. Phillips, and M D. Collins. 18. Rouff, K. L., RA. Whiley, and D. Beighton. Streptococcus. In:
Taxonomic studies on some group streptococci. J. Gen. Microbiol. Manual of clinical microbiology, 8th ed. P. R. Murray, E. J. Baron, J. H
129:1423-1432. 1983. Jorgensen , M A. Pfaller, and R. H. Yolken., eds. American Society for
12. Hernandez, D. J., E. D. Roberts, L. G. Adams, and T. Vera. Microbiology, Press, Washington, D.C. pp. 405-421. 2003.
Pathogenesis of hepatic granulomas in turkeys infected with Streptococcus 19. Teixeira, L. M and R. R. Facklam. Enterococcus. In: Manual of
faecalis var. liquefaciens. Avian Dis. 15:201-216. 1972. Clinical Microbiology, 8th ed. P. R. Murray, E. J. Baron, J. H. Jorgensen,
13. Inzana, T. J., and B. Iritani. Rapid detection of group C streptococci M A. Pfaller, and R. H. Yolken, eds. American Society for Microbiology
from animals by latex agglutination. J. Clin. Microbiol. 27:309-312. 1989. Press, Washington, D.C. pp. 422-433, 2003.
14. Jensen, W. I. An outbreak of streptococcosis in eared grebes (Podiceps 20. Vanrobaeys, M, F. Haesebrouck, R. Ducatelle, and P. De HerdL
nigricollis). Avian Dis. 23:543-546. 1979. Identification of virulence associated markers in the cell wall of pigeon
15. Jortner, B. S., and C. F. Helmboldt. Streptococcal bacterial endocarditis Streptococcus gallolyticus strains. Vet. Microbiol. 73:319-325. 2000.
in chickens. Vet. Pathol. 8:54—62. 1971. 21. Wages, D. P. Enterococcosis. In: Diseases of poultry, 11th ed. Y.M
16. MacFaddin, Jean F. Biochemical tests for the identification of medical Saif, H. J. Bames, J. R. Glisson, A. M Fadly, L. R. McDougald, and D. E.
bacteria. 3rd edition. Lippincott, Williams and Wilkins, Baltimore, MD. Swayne, eds. Iowa State Press, Ames, Iowa. pp. 809-812. 2003.
2000. 22. Wages, D. P. Streptococcosis. In: Diseases of poultry, 11th ed. Y.M
Saif, H. J. Bames, J. R. Glisson, A. M Fadly, L. R. McDougald, and D. E
Swayne, eds. Iowa State Press, Ames, Iowa. pp. 805-808. 2003.

46
13
CLOSTRIDIAL DISEASES
Stephan G. Thayer and David A. Miller

SUMMARY. The genus Clostridium includes the Gram-positive, spore-forming, catalase-negative, anaerobic bacilli. More than 100
species of Clostridium have been described, and many have been recovered from environmental and clinical specimens of wild and domestic
avian species. Pathogenicity of Clostridia is mediated primarily through potent exotoxins. Clostridial diseases may occur as soft tissue
infections, intoxications, and toxico-infections. Four economically significant avian clostridial diseases are discussed in this chapter
botulism, ulcerative enteritis, necrotic enteritis, and gangrenous dermatitis.

BOTULISM

Botulism is caused by intoxication with one of eight neurotoxins produced by the anaerobic bacillus Clostridium botulinum. Most avian
outbreaks have been caused by toxin type C. Outbreaks of high mortality have occurred in free-ranging and confined poultry and wild avian
species worldwide. Mortality may reach 40% in chickens and turkeys and has reached 100% on pheasant farms. Sources of preformed toxin
include feed, carcasses, maggots, or decaying plant material. In some outbreaks, no source of toxin could be identified, leading to the
conclusion that toxin production can occur in vivo.
Agent Identification. A presumptive diagnosis is made based on clinical signs including a characteristic flaccid paralysis that begins in the
legs and progresses cranially to the wings, neck, and eyelids.
Serologic Detection in the Host. Serologic testing is of little value because the intoxicating dose of neurotoxin is less than the dose required
to produce detectable levels of antitoxin. Confirmation of a diagnosis of botulism is usually based on demonstration of the toxin in serum of
affected birds by mouse bioassay or PCR. Isolation of C. botulinum from tissues of affected birds is not definitive evidence for diagnosis of
botulism, because the organism has been isolated from healthy birds.

ULCERATIVE ENTERITIS

Ulcerative enteritis, known as quail disease, is an acute infection of young chickens, turkeys, and game birds caused by Clostridium
colinum. Quail are more susceptible than other avian species. The disease typically occurs along with or after periods of stress or other
diseases, such as coccidiosis, aplastic anemia, or bursal disease. Typical outbreaks are characterized by sudderi deaths without prior clinical
signs, watery white diarrhea in quail, and mortality up to 10% in chickens and 100% in quail.
Agent Identification. Presumptive diagnosis is based on gross lesions, including multiple necrotic areas and ulceration in small intestine and
cecum, liver necrosis, and enlarged hemorrhagic spleen. Confirmation is by isolation and biochemical characterization or demonstration of
C. colinum antigens by fluorescent antibody tests. An enterotoxemic syndrome in young broilers characterized by duodenitis, nephrosis, and
high mortality has also been associated with C. colinum infection.
Serologic Detection in the Host. A complement fixation test has been used to detect carrier birds.

NECROTIC ENTERITIS

Necrotic enteritis, caused by Clostridium perfringens types A and C, is characterized by intestinal mucosal necrosis in chickens, turkeys,
and other avian species. Predisposing factors include high-energy-high-protein rations and mucosal damage mediated by high-fiber litter and
coccidia. Occurrence of the disease also appears to be influenced by age and genetic factors. Outbreaks have been reported most frequently in
2 to 5-wk-old broilers on litter. Clinical signs are nonspecific and birds may die without prior signs. Gross lesions include necrosis of the
mucosa in the jejunum and ileum and the formation of pseudomembranes that adhere to mucosal surfaces.
Agent Identification. A presumptive diagnosis is based on gross and microscopic lesions and is confirmed by isolation of the etiologic
agent.
Serologic Detection in the Host. Serologic testing is not used in diagnosis of necrotic enteritis.

GANGRENOUS DERMATITIS

Gangrenous dermatitis, a sporadic disease of complex etiology, is characterized by necrosis of skin, subcutaneous tissue, and underlying
musculature. Signs include depression, inappetence, incoordination, leg weakness, and ataxia. C. perfringens type A, Clostridium septicum,
and Staphylococcus aureus have been most frequently associated with the disease. Predisposing factors include infection with agents that
cause immunosuppression, including infectious bursal disease virus, chick anemia virus, and avian adenoviruses. Outbreaks have been
reported in chickens and turkeys 17 days to 20 wk of age, but most commonly 4 to 8-wk old broilers have been affected. Mortality rates have
ranged from 1% to 60%.
Agent Identification. The disease is usually diagnosed on the basis of gross and microscopic lesions and isolation of Clostridium znAJtx
Staphylococcus species.
Serologic Detection in the Host. Serologic tests are not used for diagnosis of gangrenous dermatitis.

47
Stephan G. Thayer and David A. Miller

BOTULISM

INTRODUCTION botulinum from tissues of affected birds has generally not been
regarded as definitive evidence to support a diagnosis of botulism,
Botulism, sometimes referred to as limbemeck or western duck because the organisms have been isolated from apparently normal
sickness, results from intoxication or toxico- infection with birds (8,25). However, other investigators reported recovering the
Clostridium botulinum. The disease occurs worldwide in free- organism from tissues of birds only if the flock had a history of
ranging and confined poultry and wild avian species (8). Although botulism (8).
the disease is usually sporadic and highly fatal, it may appear in a
variety of different ways, including a recurrent paralytic condition PREFERRED CULTURE MEDIA AND SUBSTRATES
in successively reared flocks on some farms (10). Eight toxigenic
types (A, B, C alpha, C beta, D, E, F, and G) of C. botulinum have Cold gelatin diluent (pH 6.2) containing 0.4 M phosphate buffer
been identified. Most avian outbreaks have been caused by C. and 0.2% gelatin may be used to extract specimens for toxin assays
botulinum type C. Although rare, botulism of types A and E has and to dilute specimens prior to inoculation of media for isolation of
occurred in small chicken flocks fed human food waste. A C. botulinum (15). Chopped meat-glucose-starch (CMGS) medium
contaminated feed source was incriminated in one type A outbreak may be used as an enrichment medium for isolation of the
in chickens (7, 25). Epizootics of type E botulism have occurred in organisms (25). McClung-Toabe egg yolk agar plates are used to
wild birds in the Great Lakes area and research has suggested that streak growth from the enrichment medium for isolated colonies
live fish can be a vector for type E toxin to waterfowl (2, 44). The and to detect lipase-positive colonies (15).
disease most often results from ingestion of preformed neurotoxin,
but no source of toxin could be identified in many broiler outbreaks. AGENT IDENTIFICATION
This has lead to the conclusion that ingested bacteria elaborate the
toxin in vivo as has been shown to occur in humans with toxin type The mouse bioassay for detection and typing of toxin in avian
B (8,15). Litter, decaying plant matter, feed, maggots and other serum samples has been commonly used to demonstrate the
insect larvae, small crustaceans, animal carcasses, and feces have presence of neurotoxin (15,25,44). However the mouse bioassay is
been incriminated as sources of preformed toxin (7,13,24,25,35). rapidly being replaced by in-vitro assays such as the amplified
Maggots have been shown to contain up to 40,000 mouse lethal enzyme-linked immunosorbent assay * (ELISA), antigen-capture
doses of toxin per gram (13). ELISA, and immuno-polymerase chain reaction assays. These
assays approach or are as sensitive as the mouse bioassay
CLINICAL DISEASE (5,33,43,44).
Clinical and environmental specimens intended for isolation of C.
The neurotoxin acts at peripheral cholinergic nerve termini by botulinum may be inoculated into CMGS medium for enrichment in
blocking release of acetylcholine (8,24). Flaccid paralysis of legs, a ratio of 1 g of specimen to 10 ml of medium. Prior to inoculation,
wings, neck, and eyelids is a characteristic sign. In mild tissues should be macerated in a small amount of gelatin diluent,
intoxications, only leg paralysis may be observed and affected birds and fecal specimens should be diluted 1:4 in gelatin diluent and the
may appear dull and reluctant to move. If forced to move, birds supernatant used as inoculum. Two tubes of CMGS medium should
appear lame. Paralysis progresses cranially from the legs to the be held in a 100 C water bath for 10 min. One tube is then placed in
wings, neck, and eyelids. Birds may sit with their necks and wings cold water and the other in an 80 C water bath. After equilibration,
outstretched and legs tucked underneath, unable to rise. The eyelids 1 ml or approximately 1 g of specimen is inoculated into each tube.
may droop or be closed completely and birds may appear comatose After inoculation, the 80 C tube is held at that temperature an
(8.25.35) . The characteristic flaccid neck paralysis seen in chickens additional 10 min, followed by cooling. Both tubes are incubated
and turkeys may not be seen in pheasants (8,24,35). If the birds are anaerobically at 30 C for 4 days. Aliquots of the cultures are
lifted, a curled-toe paralysis may be evident in turkeys. Broilers centrifuged at 12,000 x g and the supernatants are recovered for
may have diarrhea with increased urates in feces and turkey feces toxin assays. Positive enrichment cultures should be streaked onto
may be darker than normal (8,35). In some outbreaks, the disease egg yolk agar plates, which are incubated anaerobically at 35 C for
has been limited to certain pens or sections of a chicken or turkey 48 hr. If growth is observed only on plates from unheated
house and only male turkeys have been affected (8,35). Attack rates enrichment cultures, ethanol treatment of enrichment cultures may
of 30-50% are common in chickens and turkeys. Mortality rates be necessary to recover less heat-stable spore forming organisms.
may reach 40% in chickens and turkeys and have reached 100% on To confirm the identity of isolates, lipase-positive colonies may be
some pheasant farms (8,23,35). Recovering birds usually regain incubated anaerobically in CMGS medium for 4 days at 30 C
muscle control in the eyelids first, then in neck and wings, and followed by biochemical testing and toxin typing by mouse
finally in legs over a 3 to 5-day period if given ready access to feed bioassay ELISA, PCR or other methods (15).
and water (9,35).
SEROLOGIC DETECTION IN THE HOST
SAMPLE COLLECTION
Serologic testing of affected birds is of little value because the
Serum is the specimen of choice for demonstrating the presence of intoxicating dose of neurotoxin is less than the dose required to
neurotoxin in birds. The toxin may also be demonstrated in the produce detectable levels of antitoxin. For this reason, recovering
liver, spleen, crop, and gastrointestinal contents. Specimens should birds do not develop protective immunity (8).
be collected from live birds showing clinical signs, if possible,
because postmortem decomposition may result in toxin production DIFFERENTIATION FROM CLOSELY RELATED AGENTS
from C. botulinum in the intestines leading to a misdiagnosis. Feed,
larvae, carcasses, and environmental specimens may be collected Clinically, botulism may be similar to Marek’s disease, Newcastle
for toxin analysis to provide evidence of the source of intoxication disease, avian encephalomyelitis, chemical intoxications, and
(8.13.35) . Specimens should be frozen and transported to a musculoskeletal problems in poultry. In waterfowl, fowl cholera,
laboratory as soon as possible after collection. Isolation of C. chemical intoxication, and lead poisoning should be considered.
48
Chapter 13 Clostridial Diseases

Differential diagnosis is based on clinical signs and a lack of gross nature can be demonstrated by holding culture supernatant at 100 C
and microscopic lesions. Definitive diagnosis is usually based on for 10 min, followed by mouse inoculation. Isolates of C. botulinum
demonstration of toxin in serum of affected birds, followed by can be differentiated from other Clostridia by biochemical testing
identification of the toxin type (8,25). The neurotoxin’s heat labile and toxin typing (15).

ULCERATIVE ENTERITIS

INTRODUCTION
AGENT IDENTIFICATION
Ulcerative enteritis, originally referred to as quail disease, is
caused by Clostridium colinum. The disease has worldwide Characteristic gross lesions include multiple necrotic areas and
distribution and has been reported in chickens, wild and domestic ulceration in the small intestine and ceca, areas of liver necrosis,
turkeys, pigeons, pheasants, grouse, partridges, and robins, but quail and an enlarged hemorrhagic spleen. In quail, marked hemorrhagic
are the most susceptible species. Although the disease has been enteritis is apparent in the duodenum. Ulcers in ceca may have
reported in adult birds, it usually affects younger birds. Outbreaks in central depressions containing dark debris (25,39). Chickens may
chickens frequently occur after or during periods of stress or other have clearly circumscribed yellow ulcers 1-5 mm in diameter in the
disease conditions, such as coccidiosis, aplastic anemia, and bursal mucosa that are detectable from the serosal side (21). Large Gram­
disease. Birds become infected by ingestion of the organism in feed, positive bacilli with subterminal spores can be demonstrated in
water, or litter. Premises remain contaminated with spores for smears of necrotic liver tissue by Gram stain. Clostridium colinum
prolonged periods after outbreaks. It has been suggested that is fastidious and some laboratories have experienced difficulty in
surviving birds become chronic carriers (2,25). isolating the organism from birds with characteristic gross lesions
(21,39). Specimens intended for isolation of C. colinum should be
CLINICAL DISEASE inoculated into pre-reduced tryptose phosphate medium and
incubated anaerobically at 37 C for 2 days. Colonies are 1-2 mm in
Birds may die in good physical condition without prior clinical diameter, round, semi translucent, convex, and have filamentous
signs. Quail may have watery white feces. Birds may be listless margins. Growth in broth may occur within 12-16 hr after
with eyes partially closed and have a humped appearance. inoculation and is characterized by gas production that subsides 6-8
Emaciation with breast muscle atrophy may be seen in birds in hr after it begins (1,25). Biochemical characterization should be
which the clinical course is 1 wk or more. Mortality may reach 10% used to confirm the identity of an isolate. The typical gas
in chickens and 100% in quail. Because some surviving birds have chromatogram will indicate acetic and formic acid production
resisted subsequent experimental challenges, active immunity (1,21). A fluorescent antibody test for diagnosis of ulcerative
appears to develop after outbreaks. However, survivors treated with enteritis has correlated well with presumptive diagnoses based on
antibiotics may remain susceptible to infection (39). An unusual gross lesions. An agar gel diffusion test has been used to detect
enterotoxemic syndrome in 12 to 25-day-old broilers caused by C. soluble C. colinum antigens in intestinal contents, but cross
colinum has also been reported in Israel. Clostridium colinum was reactions with Clostridium perfringens types A and C are a problem
the only organism consistently isolated from acutely infected birds (39).
exhibiting severe duodenitis, nephrosis, and mortality up to 50%
(27). In another report a combination of Clostridim perfringens, SEROLOGIC DETECTION IN THE HOST
Capillaria sp., Eimeria sp., Histomonas sp., and E. coli were
reported in an epizootic that involved quail (29). A complement fixation test has been used to detect carrier birds
(39).
SAMPLE COLLECTION
DIFFERENTIATION FROM CLOSELY RELATED AGENTS
Clostridium colinum may be isolated in pure culture from liver
specimens (24). The organism may also be recovered from spleen Clinically, ulcerative enteritis may be similar to coccidiosis,
and intestinal lesions (39,21). Swabs or tissues collected at histomoniasis, and necrotic enteritis. Because coccidiosis may occur
necropsy should be cultured as soon as possible, and should be before or during ulcerative enteritis outbreaks, the etiologic agents
placed in anaerobic transport tubes (Starswab Anerobic Transport of both diseases may be present in specimens submitted to
System, VWR International) if transportation to a laboratory is diagnostic laboratories. Coccidia may be demonstrated in fecal or
necessary. impression smears. Histomoniasis may present cecal and liver
lesions similar to those of ulcerative enteritis (39). Histomonas may
PREFERRED CULTURE MEDIA AND SUBSTRATES be demonstrated in lesions by histopathologic examination.
Clostridium perfringens, the cause of necrotic enteritis, may be
The medium of choice for isolation of C. colinum is tryptose distinguished from C. colinum by cultural and biochemical
phosphate agar with 0.2% glucose and 0.5% yeast extract. Ihe pH methods. Clostridium difficile resembles C. colinum biochemically,
should be adjusted to 7.2, and the medium autoclaved, followed by but differs in raffinose fermentation and gelatin hydrolysis (25,39).
cooling to 56 C and addition of 8% horse plasma. The same
medium without agar (broth) may also be used (39).

49
Stephan G. Thayer and David A. Miller

NECROTIC ENTERITIS

INTRODUCTION minimize overgrowth by other organisms. If transportation of


specimens to a laboratory is necessary, use of anaerobic transport
Necrotic enteritis is a sporadic disease of avian species swabs (Starswab Anerobic Transport System, VWR International)
characterized by necrosis of the intestinal mucosa primarily in the is recommended (25,40).
jejunum and ileum with the formation of pseudomembranes (25).
This disease has been reported from most areas of the world in PREFERRED MEDIA AND SUBSTRATES
which poultry are produced. Recent restrictions on the use of
antibiotics and some anticoccidials has played a role in the increase Anaerobic blood agar and PEA agar (Phenyethanol alcohol agar)
in the incidence of necrotic enteritis in some parts of the world (19). can be used to isolate C. perfringens from specimens collected at
The disease affects both broilers and layers and has also been necropsy. PEA agar is useful where contamination with facultative
reported in wild and domestic ducks and geese, capercaillies, and enterics and swarming enteric anaerobes interfere. Classical double
red lories (4,26,37,40). The etiologic agents are C. perfringens types zone hemolysis is useful in detection of C. perfringens colonies
A and C. In earlier work the alpha toxin produced by type A and though it may be observed as only a beta-hemolysis without the
type C organisms and the beta toxin produced by type C organisms double-zone characteristic. McClung-Toabe egg yolk agar can be
were thought to be responsible for the necrosis of intestinal mucosa used to detect lecithinase and lipase production by isolates. For
(40). However, more recent work has discounted the role of the determination of toxin types, isolates should be propagated in
Alpha toxin requirement in the production of necrotic lesions in the cooked meat medium containing 1% glucose (16,40).
intestine (20). Plasmid encoded toxin genes appear to be stable
under normal transportation and culture conditions (17). AGENT IDENTIFICATION
Toxinotyping of C. perfringens can be done by PCR and multiplex
PCR (16). Predisposing factors include high-energy-high-protein Diagnosis of necrotic enteritis is usually made on the basis of
diets, rations containing high levels of fish meal, wheat, rye, barley, gross and microscopic lesions and isolation of toxigenic Cl.
and oat groats, as well as damage to the intestinal mucosa induced perfringens. Gram stains of impression smears from intestinal
by high-fiber litter and coccidial infections (4,8,14,18,21,25,31). mucosa may reveal large numbers of Gram-positive bacilli (14).
Coccidiosis has been the most frequently diagnosed concurrent Clostridium perfringens can be isolated by inoculation of pre­
intestinal disease (11). Genetic susceptibility may also be a factor reduced blood agar and PEA plates incubated anaerobically at 37 C
influencing occurrence in chickens, as evidenced by significantly overnight. Isolates are characterized by smooth, circular colonies 2-
different mortality rates between different major histocompatibility 4 mm in diameter surrounded by an inner β-hemolytic zone and an
complex genotypes (34). Outbreaks of necrotic enteritis have outer zone of incomplete β-hemolysis. Though this double zone is
occurred most frequently in broilers 2-5 wk of age raised on litter, the classical appearance this characteristic is not always seen. Gram
but have also occurred in 3 to 6-mo-old layers raised in floor pens stains typically reveal short, thick rods with blunt ends, usually
and 12 to 16-wk-old caged layer replacements. In turkeys the without spores. Classical biochemical methods can be used to
disease has occurred primarily in birds 6-11 wk of age. The confirm identity of isolates. Typical gas chromatograms indicate
frequency of disease occurrence has been greater in male than in production of large amounts of acetic and butyric acids with smaller
female turkey flocks (11,40). Mortality rates up to 15% have been amounts of propionic and formic acids. The presence of lecithinase
reported in chickens (34). Not much is known about the role of and absence of lipase can be observed on egg yolk agar. As an
immunity in necrotic enteritis but it has been shown that protection alternative to classical biochemical methods which may take 48
is associated with immunization or infection with virulent more hours to complete, the use of a pre-formed enzyme
C. perfringens but not with avirulent strains (22). identification system (RapID ANA Π System, Remel, Inc., Lenexa,
KS) permits 4-6 hr identification. Toxin types can be identified by
CLINICAL DISEASE inoculation of isolates into pre-reduced cooked meat-glucose
medium, which is incubated anaerobically at least 6 hr at 37 C.
Clinical signs may be nonspecific. Birds exhibit depression, Cultures should be centrifuged and supernatants tested for toxins.
inappetence, reluctance to move, diarrhea, and ruffled feathers. The To 1.2-ml aliquots of supernatants, 0.3 ml of C.perfringens types A
clinical course is frequently short and birds may be found dead and C antitoxins are added, followed by neutralization at room
without premonitory signs (25,40). Turkeys may be well fleshed temperature for 30 min, and injection of 0.5 ml intravenously into
and feathered but severely dehydrated (14). Gross lesions are pairs of mice. A third group of mice receives supernatant mixed
frequently confined to jejunum and ileum and include yellow or with normal horse serum as a control. Mice should be observed for
green pseudomembranes adherent to the mucosa (25). Cecal lesions 24 hr. If the isolate is type A, mice inoculated with supernatants
have also been reported and may include small areas of mucosa neutralized by type A and type C antitoxins will survive. If the
covered by a thick whitish brown pseudomembrane in turkeys isolate is type C, only mice receiving supernatants neutralized by
(14,40). Intestines may be friable with a thickened wall distended type C antitoxin will survive (25,40).
by gas and contain bile-stained fluid or a granular core of Toxin typing can also be accomplished using multiplex PCR to
fibrinonecrotic debris and litter (18,40). Tylosin phosphate at 100 determine the presence of genes coding for the different toxin types
ppm can be used to reduce C. perfringens colonization and thus the in C. perfringens isolates or in C. perfringens in the test sample
severity of necrotic lesions (6). (16).

SAMPLE COLLECTION SEROLOGIC DETECTION IN THE HOST

Clostridium perfringens can be isolated from intestinal contents, Serologic testing is not used in the diagnosis of necrotic enteritis.
mucosal scrapings, hemorrhagic lymphoid nodules, and swabs of
pseudomembranous lesions. Specimens should be fresh and
processed as soon as possible to increase likelihood of recovery and
50
Chapter 13 Clostridial Diseases

DIFFERENTIATION FROM CLOSELY RELATED AGENTS


by ascending infection if the bile ducts (cholangiohepatitis). Also a
Necrotic enteritis may resemble ulcerative enteritis and multifocal hepatitis with accompanying fibrinoid necroses may be
coccidiosis. Because necrotic enteritis and coccidiosis may occur seen. Cecal involvement is usually minimal. Cultural and
simultaneously, the etiologic agents of both diseases may be biochemical methods can be used to distinguish C. perfringens
present in specimens submitted to diagnostic laboratories. Lesions from C. colinum. Coccidia can be demonstrated by microscopic
of necrotic enteritis can be found in the jejunum and ileum. These examination of fecal smears, mucosal impression smears, or
appear as ulcers or light yellow spots on the mucosal surface. intestinal sections (19,25,40).
Occasionally lesions may be seen in the liver thought to be affected

GANGRENOUS DERMATITIS

INTRODUCTION SAMPLE COLLECTION

Gangrenous dermatitis is a sporadic disease of chickens and Specimens or swabs of exudates and affected tissues, including
turkeys characterized by necrosis of the skin, subcutaneous tissue, skin and underlying muscle, should be processed for bacterial
and possibly underlying musculature. The disease has been referred isolation as soon as possible after collection. Isolation of
to by a variety of other names, including necrotic dermatitis, Clostridium species can be enhanced by use of anaerobic transport
gangrenous cellulitis, gangrenous dermatomyositis, avian malignant swabs (Star-Swab Anerobic Transport System, VWR Scientific) if
edema, gas edema disease, and wing rot. The etiology appears to be transportation to a laboratory is necessary.
complex and may include various species of aerobic and anaerobic
bacteria. Clostridium sordellii, C. novyi, C. sporogenes, C. PREFERRED MEDIA AND SUBSTRATES
subterminale, Escherichia coli, Staphylococcus epidermidis, other
Staphylococcus and Streptococcus species, and Gallibacterium Media for isolation and identification of Staphylococcus species
anatis biovar haemolytica (formerly Pasteurella haemolytica) have and other facultative bacteria that may be associated with
been isolated from comparable gross lesions (3,25,30). Clostridium gangrenous dermatitis have been described in previous chapters.
chauvoei has also been isolated from chickens exhibiting Isolation of C. perfringens has been described earlier in this chapter
inflammatory lesions on the skin of the head with necrosis of the and specimens for isolation of C. septicum should be inoculated
comb (28). However, C. septicum, C. perfringens type A, and onto blood agar and PEA (Phenylethanol) agar plates to control
Staphylococcus aureus, either singly or in combination, have been swarming. McClung-Toabe egg yolk agar can be used to detect
most frequently associated with gangrenous dermatitis. lecithinase and lipase production of isolates (25,41).
Predisposing factors are other infectious agents that suppress
immunologic functions, such as infectious bursal disease virus, AGENT IDENTIFICATION
chick anemia virus, and avian adenoviruses. Some evidence exists
of a genetic effect on susceptibility, because progeny from certain Gangrenous dermatitis is usually diagnosed on the basis of gross
breeder flocks have consistently developed die disease after and microscopic lesions and isolation of Clostridium and/or
placement. Outbreaks have been reported in chickens and turkeys Staphylococcus species. Clostridium septicum and other clostridial
17 days to 20 wk of age, but most commonly 4 to 8-wk-old broilers species can be isolated on pre-reduced blood agar and PEA agar
have been affected. One report documented the annual recurrence of plates incubated anaerobically at 37 C overnight. On blood agar, C.
gangrenous dermatitis caused by C. septicum (42). Outbreaks have septicum will swarm the entire plate overnight. C. sordellii can
also been reported in 6 to 20-wk-old layers, 20-wk-old broiler partially cover a BA plate in the same time frame. Other species
breeders, chickens following caponization, and turkey breeder hens. such as C. perfringens will form defined colonies. PEA will control
Mortality rates have ranged from 1 to 60% (25,41). swarming and permit selection of defined colonies for further
isolation as well as help control contamination and unwanted
CLINICAL DISEASE growth. Isolated colonies can be easily seen and motile Clostridia
will have irregular margins. Once isolation is achieved use of a pre­
Signs include depression, inappetence, incoordination, leg formed enzyme identification system (RapID ANA Π System,
weakness, and ataxia. The clinical course may be short, usually less Remel, Inc., Lenexa, KS) permits 4-6 hr identification. Fluorescent
than 24 hr, and birds are often found dead without prior signs. Gross antibody conjugates are available for presumptive identification of
lesions include dark, wet areas of skin devoid of feathers overlying several clostridial species in specimens and in culture. A more
wings, breast, abdomen, or legs. Serosanguinous fluid, with or classical approach would include growth on egg yolk agar
without gas, may accumulate under affected skin and diffuse into demonstrating a lack of lecithinase and lipase production. The
surrounding subcutaneous tissues under apparently normal skin. identity of isolates could then be confirmed biochemically. The
The underlying musculature may appear gray or tan with fluid and typical gas chromatogram will indicate production of large amounts
gas between muscle bundles (41). A cellulitis in turkeys of acetic and butyric acids, usually with large amounts of formic
characterized by swelling and vesicular lesions on the tailheads with acid (25,41).
gelatinous fluid accumulation over the breast has been reported (3).
Usually no involvement of internal organs is observed, but SEROLOGIC DETECTION IN THE HOST
occasionally birds may have discrete white foci of liver necrosis.
Significant atrophy of the bursa of Fabricius has been reported in Serologic testing is not used to diagnose gangrenous dermatitis.
some outbreaks. On histopathologic examination, large basophilic
bacilli and small cocci may be seen in subcutaneous tissues (30,41). DIFFERENTIATION FROM CLOSELY RELATED AGENTS

Because viral infections may predispose birds to gangrenous


dermatitis, viral isolation and identification should be done (25). A
variety of other skin conditions may appear similar to this disease,
51
Stephan G. Thayer and David A. Miller

including mycotic dermatitis, ulcerative dermatitis of broilers, 21. Kondo, F. Ulcerative enteritis in broiler chickens caused by Clostridium
plantar pododermatitis of turkeys, scabby hip lesions of broilers, colinum and in vitro activity of 19 antimicrobial agents in tests on isolates.
ulcerative squamous cell carcinoma, nutritional deficiencies, and Poult. Sci. 67:1424-1430. 1988.
22. Kulkami, R. R., V. R. Parreira, S. Sharif, and J. F. Prescott. Clostridium
genetically related slow feathering of male chickens. Mycotic perfringens antigens recognized by broiler chickens immune to necrotic
dermatitis can be diagnosed by demonstration of fungal elements in enteritis. Clin. Vacc. Immunol. 13:1358-1362. 2006.
impression smears or isolation of the agents. Ulcerative dermatitis 23. Kurazono, Η., K. Shimozawa, and G. Sakaguchi. Botulism among
and plantar pododermatitis are usually associated with wet or poor penned pheasants and protection by vaccination with Cl toxoid. Res. Vet.
litter conditions. Staphylococcus species and C. perfringens have Sci. 38:104-108. 1985.
been recovered in pure culture from some birds with scabby hip 24. Linares, J. A., R. L. Walker, A. A. Bickford, G. L. Cooper, and B. R.
lesions and may be primary or secondary etiologic agents of that Charlton. An outbreak of type C botulism in pheasants and subsequent
condition (32). Squamous cell carcinoma can be diagnosed by occurrence in chickens. J. Vet. Diagn. Invest. 6:272-273. 1994.
25. Miller, David A. Clostridial Diseases. In: Isolation and Identification of
histopathologic examination. Avian Pathogens, 4th ed. D. E. Swayne, J. R. Glisson, M. W. Jackwood, J.
E. Pearson, W. M. Reed (eds.). American Association of Avian Pathologists:
REFERENCES Kennett Square, PA, 61—68. 1998.
26. O’Toole, D., K. Mills, R. Ellis, R. Farr, and M Davis. Clostridial
1. Berkhoff, H. A. Clostridium colinum sp. nov., nom. rev., the causative enteritis in red lories (Eos bomea). J. Vet. Diagn. Invest. 5:111-113. 1993.
agent of ulcerative enteritis (quail disease) in quail, chickens, and pheasants. 27. Perelman, B., S. Mints, M. Zjut, E. Kuttin, and S. Machny. An unusual
Int. J. Syst. Bacteriol. 35:155-159. 1985. Clostridium colinum infection in broiler chickens. Avian Pathol. 20:475-
2. Brand, C. J., S. M. Schmitt, R. M . Duncan, and T. M. Cooley. An 480. 1991.
outbreak of type E botulism among common loons (Gavia immer) in 28. Prukner-Radovcic, E., L. Milakovic-Novak, S. Ivesa-Petricevic, andN.
Michigan’s upper peninsula. J. Wildl. Dis. 24:471-476. 1988. Grgic. Clostridium chauvoei in hens. Avian Pathol. 24:201-206. 1995.
3. Carr, D., D. Shaw, D. A. Halvorson, B. Rings, and D. Roepke. Excessive 29. Radi, Zaher Ahmed An Epizootic of Combined Clostridium
mortality in market-age turkeys associated with cellulitis. Avian Dis. perfringens, Eimeria spp., and Capillaria spp. Enteritis and Histomonas spp.
40:736-741. 1996. Hepatitis with Escherichia coli septicemia in Bobwhite Quails (Colinus
4. Chakrabarty, A K., B. R. Boro, A. Mukit, B. M Dutta, and Μ M. virginianus). Int’l J. Poult. Sci. 3:438-441. 2004.
Baruah. An epomitic of enteritis in brahminy ducks (Todoma ferruginea) 30. Reece, R. L., V. D. Beddome, and D. A. Ban. Diseases diagnosed in
due to enterotoxigenic strains of Clostridium perfringens type C. J. Zoo. broiler chicken flocks in Victoria, Australia, 1977 to 1984. Vet. Rec.
Anim. Med. 14:24-27. 1983. 116:315-320. 1985.
5. Chao, Η. Y., Y. C. Wang, S. S. Tang, and H. W. Liu. A highly sensitive 31. Riddell, C., and X. M Kong. The influence of diet on necrotic enteritis
lmmuno-polymerase chain reaction assay for Clostridium botulinum in broiler chickens. Avian Dis. 36:499-503. 1992.
neurotoxin type A. Toxicon. 43:27-34. 2004. 32. Scanlan, C. M., and B. M Hargis. A bacteriologic study of scabby-hip
6. Collier, C. T., J. D. van der Klis, B. Deplancke, and D. B. Anderson. lesions from broiler chickens in Texas. J. Vet. Diagn. Invest. 1:170-173.
Effects of tylosin on bacterial mucolysis, Clostridium perfringens 1989.
colonization, and intestinal barrier function in a chick model of necrotic 33. Sharma, S. K., J. L. Ferreira, B. S. Eblen, and R. C. Whiting. Detection
enteritis. Antimicrob. Agents Chemother. 47:3311-3317. 2003. of type A, B, E, and F Clostridium botulinum toxins in foods by using and
7. Craven, S. E., Colonization of the Intestinal Tract by Clostridium amplified enzyme-linked immunosorbent assay with digoxigenin-labeled
perfringens and Fecal Shedding in Diet-Stressed and Unstressed Broiler antibodies. Appl. Environ. Microbiol. 72:1231-1238. 2006.
Chickens. Poult. Sci. 79:843-849. 2000. 34. Siegel, P. B., A S. Larsen, C. T. Larsen, and E. A Dunnington.
8. Dohms, J. E. Botulism In: Diseases of Poultry, 11th ed. Y.M. Saif, H. J. Resistance of chickens to an outbreak of necrotic enteritis as influenced by
Barnes, J. R. Glisson, A. M Fadly, L. R. McDougald, and D. E. Swayne, major histocompatibility genotype and background genome. Poult. Sci.
eds. Iowa State Press, Ames, Iowa. pp. 785-791. 2003. 72:1189-1191. 1993.
9. Dohms, J. E., P. H. Allen, and J. K. Rosenberger. Cases of type C 35. Smart, J. L. Type C botulism in intensively farmed turkeys. Vet. Rec.
botulism in broiler chickens. Avian Dis. 26:204-210. 1982. 113:198-200. 1983.
10. Dohms, J. E., P. H. Allen, and S. S. Cloud. The immunization of broiler 36. Smith, G. R., and J. C. Oliphant. Diagnosis of botulism in water birds.
chickens against type C botulism Avian Dis. 26:340-345. 1982. Vet. Rec. 112:457^158. 1983.
11. Droual, R., T. B. Farver, and A. A. Bickford. Relationship of sex, age, 37. Stuve, G., M Hofshagen, and G. Holt. Necrotizing lesions in the
and concurrent intestinal disease to necrotic enteritis in turkeys. Avian Dis. intestine, gizzard, and liver in captive capercaillies (Tetrao urogallus)
39:599-605. 1995. associated with Clostridium perfringens. J. Wildl. Dis. 28:598-602. 1992.
12. Fach, P., M Gibert, R. Griffais, and M. R. Popoff. Investigation of 38. Thomas, R. J. Detection of Clostridium botulinum types C and D by
animal botulism outbreaks by PCR and standard methods. FEMS Immunol. ELISA. Aust. Vet. J. 68:111-113. 1991.
Med. Microbiol. 13:279-285. 1996. 39. Wages, D. P., Ulcerative Enteritis (Quail Disease). In: Diseases of
13. Foreyt, W. J., and F. R. Abinati. Maggot-associated type C botulism in Poultry, 11th ed. Y.M Saif, H. J. Barnes, J. R. Glisson, A. M Fadly, L. R.
game farm pheasants. J. Am. Vet. Med. Assoc. 177:827-828. 1980. McDougald, and
14. Gadzinski, P., and R. J. Julian. Necrotic enteritis in turkeys. Avian Dis. D. E. Swayne, eds. Iowa State University Press, Ames, Iowa. pp. 776-781.
36:792-798. 1992. 2003.
15. Hatheway, C. L. Laboratory procedures for cases of suspected infant 40. Wages, D. P., and K. Opengart. Necrotic Enteritis. In: Diseases of
botulism Rev. Infect. Dis. 1:647-651. 1979. Poultry, 11th ed. Y.M. Saif, H. J. Barnes, J. R. Glisson, A. M Fadly, L. R.
16 Heikinheimo, A., and H. Korkeala. Multiplex PCR assay for McDougald, and D. E. Swayne, eds. Iowa State University Press, Ames,
toxmotyping Clostridium perfringens isolates obtained from Finnish broiler Iowa. pp. 781-785. 2003.
chickens. Lett. Appl. Microbiol. 40:407-411. 2005. 41. Wages, D. P., and K. Opengart. Gangrenous Dermatitis. In: Diseases of
17. Johansson, Anders. Clostridium perfringens the causal agent of necrotic Poultry, 11th ed. Y.M. Saif, H. J. Barnes, J. R. Glisson, A. M Fadly, L. R.
enteritis in poultry. Doctoral thesis. Swedish University of Agricultural McDougald, and D. E. Swayne, eds. Iowa State Press, Ames, Iowa. pp. 791-
Sciences. Uppsala, Sweden. 2006. 795. 2003.
18. Kaldhusdal, M., and M Hofshagen. Barley inclusion and avoparcin 42. Willoughby, D. H, A. A Bickford, G. L. Cooper, and B. R. Charlton.
supplementation in broiler diets: 2. Clinical, pathological, and Periodic Recurrence of Gangrenous Dermatitis associated with Clostridium
bacteriological findings of necrotic enteritis. Poult. Sci. 71:1145-1153. septicum in a Broiler Chicken Operation. J. Vet. Diag. Invest. 8:259-261.
1992. 1996.
19. Kaldhusdal, Magne, and Atle Lovland. Clostridial necrotic enteritis and 43. Wu H.C., Y. L. Huang, S. C. Lai, Y. Y. Huang, and M. F. Shaio.
choiangiohepatitis. The Elanco Global Enteritis Symposium Abtract G-l- Detection of Clostridium botulinum neurotoxin A using immuno-PCR. Lett.
14. Cambridge, July 9-11. 2002. Appl. Microbiol. 32:321-325. 2001.
20. Keybum, Anthony L., Scott A. Sheedy, Mark E. Ford, Mark M 44. Yule, Adam M., Ian K. Barker, John W. Austin and Richard D. Moccia.
Williamson, Milena M. Awad, Julian I. Rood, and Robert J. Moore. Alpha­ Toxicity of Clostridiium botulinum type E neurotoxin to Great Lakes fish:
Toran of Clostridium perfringens is not an essential virulence factor in Implications for avian botulism. J. Wildlife Dis. 42: 479-493. 2006.
necrotic enteritis in chickens. Infect. Immun. 74:6496-6500. 2006.
52
14
TUBERCULOSIS
Susan Sanchez and Richard M. Fulton

SUMMARY. Tuberculosis in commercial poultry occurs at a very low frequency. Tuberculosis is most common in small hobby poultry
flocks and pet birds; it is occasionally reported in captive bird collections and may occur in wild birds. The disease in birds is caused by
Mycobacterium avium subsp. avium. Mycobacteria are hardy organisms that can survive in the environment for several years. Avian
tuberculosis is a chronic disease leading to severe emaciation. The most common route of infection is fecal-oral.
Agent Identification. Multiple white nodular lesions on and/or in internal organs warrant the inclusion of avian tuberculosis in the
differential diagnosis. Finding acid fast bacilli in smears of the lesions and/or with histopathology allows confirmation of a diagnosis of
avian tuberculosis. Culture with identification of the organism and/or positive PCR further confirms the diagnosis.
Serologic Detection in the Host. Serologic tests are not readily available and do not appear to be as reliable as serologic tests of other
disease agents. Tuberculin testing may be a useful tool to detect infected chickens within a flock.

INTRODUCTION Tubercles may also be found on the serosal surfaces of intestines as


well as within the parenchyma of other internal organs, such as
Tuberculosis in commercial poultry has a very low rate of lung, and are postulated to reflect route of exposure (27). On cut
occurrence. It is most commonly found is small hobby poultry surface tubercles may be solid and white or have central areas that
flocks and pet birds. Occasionally, it is reported to occur in wild are yellow and easily crumbled, which suggests caseation. (10, 14,
birds and captive bird collections (14, 26). The disease in birds is 16, 40).
mostly caused by Mycobacterium avium subsp. avium serotypes 1, A presumptive diagnosis of tuberculosis may be made at necropsy
2, or 3. Mycobacterium genavense has been isolated with based on emaciation and nodular lesions. Bone marrow nodules are
increasing frequency from pet and zoo collection birds (7, 18, 19, also supportive of that diagnosis, as they are not often seen in other
20, 25, 26, 28, 33). Mycobacteriosis in poultry is a chronic disease diseases. For definitive diagnosis, further testing such as acid-fast
leading to emaciation and death (43). M avium belongs to the slow stain of smears or lesions, culture with identification, and/or
growing non-tuberculous bacteria group and together with M. polymerase chain reaction (PCR) is necessary.
intracellulare (serotypes 7, 12 to 20, and 22 to 28) form the
Mycobacterium avium Complex (MAC). There is evidence for a Microscopic Lesions
third species in this group, generally referred to as MAC-X (45). Microscopic lesions of avian tuberculosis consist of granulomas.
MAC organisms’ reservoir is in the environment; they have been They may be composed of aggregated epithelioid macrophages or
isolated from water, soil and plants. All members of the MAC have have centrally-located, granular debris, derived from local and/or
been isolated from animals. M. avium may be further divided into inflammatory cells, which is surrounded by epithelioid
three subspecies namely M. avium sbsps. paratuberculosis isolated macrophages with admixed multinucleated giant cells and a
from ruminants and free-ranging birds (2, 8), M. avium sbsps. peripheral zone of low numbers of lymphocytes and plasma cells.
silvaticum isolated from a penguin and wood pigeons and M. avium Calcification and fibrosis are not features of avian tuberculosis as in
subsp. avium isolated from birds. In 2002 Mijs and his mammalian tuberculosis. In addition, multi-nucleated giant cells
collaborators suggested that M. avium sbsps. avium should be are few in number in avian tuberculosis compared to that of
further subdivided based on phenotypic and genotypic grounds in to mammals. Acid-fast bacilli are usually found in large numbers in
M. avium sbsps avium for isolates originating from birds and epithelioid macrophages. (10,43)
Mycobacterium avium sbsps. hominissuis (serotypes 4, 6, 8 to 11,
and 21) for isolates recovered from humans and animals (1, 23). SAMPLE COLLECTION
Although disease in poultry is mostly attributed to one subspecies
in pet and wild birds, speciation of mycobacterium isolates is Smears and Sections
warranted as they have been reported as being infected with several Acid-fast smears and histopathologic sections can be prepared
subspecies of M. avium and species of Mycobacterium including M. from tubercles. Smears may be made by placing small tubercles
tuberculosis, M. bovis, M. fortuitum, and M. genavense (6, 7, 19). between two glass slides, compressing the tubercle between the
slides followed by sliding one slide off of the other. Some tubercles
CLINICAL DISEASE can have the surface scraped with a scalpel blade followed by
impression smears of the lesion. Slides are then air dried followed
Birds of all ages, and most likely all species, appear to be by staining with an acid-fast stain or auramine-rhodamine or both at
susceptible to M. avium sbsps avium. Signs and lesions are most the same time.
common in older birds due to the slow progression of the illness. For histopathologic sections, select parenchymal or serosal
Not all birds in an infected production flock will show signs of tubercles for fixation and processing. Experimentally inoculated
illness even though they are infected. Some birds will go out of egg chickens routinely exhibit miliary lesions in spleen, and liver while
production while others, although infected, will continue to lay lesions are less frequently observed in bone marrow, lung, and
eggs. (16) Infected birds lose weight gradually, become unthrifty in thymus (10, 32). Thus, in infection of short duration, tubercles may
appearance, and eventually die. Birds may not show signs of illness be easily overlooked. In early stages of the disease, a tuberculin
or gross lesions in the early stages of the disease. positive chicken may have few or no visible lesions.
Histopathologic examination of liver, spleen, and bone marrow
Gross Lesions from these birds may reveal granulomas not grossly visible.
Tubercles are the most prominent gross lesions of avian
tuberculosis; they appear as white to yellow to tan nodules of
various sizes (from pinpoint up to more than a centimeter).
Tubercules are found most commonly, in order of frequency, in the
parenchyma of spleen, liver, bone marrow, and thymus (10).
53
Susan Sanchez and Richard M. Fulton

Isolation and Identification Fluorochrome Stain. An alternate method for a quick


For bacteriologic culture select spleen, liver, bone marrow, or identification is to use fluorescent microscopy; in this case, the
tubercles on the peritoneal surface, in that order of preference. acid-fast stain is a mixture of auramine and rhodamine. Auramine-
Femur marrow is useful from birds with postmortem autolysis. rhodamine are non-specific fluorocromes that bind selectively to
Specimens must be removed with sterile instruments and ground to mycolic acids, staining target organisms fluoresce orange-yellow
a paste in sterile conditions with sufficient sterile water to form a under UV light. Slides must be examined under ultraviolet light
suspension thin enough to pipette. If the bird is freshly killed, the with a microscope equipped with special filters (37). This stain is
tissue suspension can be transferred directly to slants of solid more sensitive due to its minimal background allowing for the study
media. Sample decontamination should only be attempted if the of the smear with lower magnification. It also has the advantage that
sample is thought to be contaminated or culture is attempted from it can be used in conjunction with other stains.
feces. The decontamination process also destroys mycobacterial
cells and concentration of mycobacteria is recommended after this Isolation and Identification
process to enhance isolation. The most common decontaminant is Any doubt about the diagnosis can be resolved only by isolating
sodium hydroxide (see Appendix I for detail method). the microorganism as described under sample collection above. For
In some instances isolation of the causative agent is not necessary epidemiologic studies, avian mycobacteria must be isolated in pure
or not possible yet confirmation of mycobacteriosis is important. In culture for serotyping. Isolation and identification of M. avium may
such situations detection of mycobacterial DNA using PCR is a fast require a minimum of 4 wk and negative samples can only be
and very reliable option. Results can be obtained within the same discarded after 8 wk.
day of sample collection (34, 35). All initial growth must be stained for acid-fast bacilli to ensure
purity of culture. For optimal recovery, samples may be plated in
PREFERRED CULTURE MEDIA AND SUBSTRATES both liquid and solid media. Culture in liquid media is faster than on
solid media. Media can be selective (includes antibiotics) if
For primary cultures, only solid media are recommended. contamination is suspected or is elected to forgo the
M. avium sbsps. avium grows well on commercially available media decontamination process. The following media enhance the growth
used for isolating Mycobacterium tuberculosis, such as of MAC organisms and may be used preferentially in a poultry
Lowenstein-Jensen (LJ) inspissated egg medium or Middlebrook diagnostic laboratory (see appendix 1 for a more extensive list of
7H10 agar (Difco, Detroit, Mich.). The addition of sodium media): Lowenstein-Jensen medium or other suitable slants, tubes
pyruvate, 4.1 g per liter of 7H10 medium, is recommended for of Proskauer-Beck liquid medium containing 5% serum or
isolating M, avium sbsps. avium from tissues (41). Suitable growth Middlebrook which contains 2% glycerol. Isolated mycobacteria
will develop on blood agar, especially if the base agar has 2.0% can be used in subsequent tests for pathogenicity determination and
glycerol. If subcultures in liquid media are desired, Proskauer- in in vitro differential tests (12). PCR has been used for rapid
Beck liquid medium containing 5.0% serum or Middlebrook 7H9 identification of MAC organisms (34). Restriction fragment length
liquid medium may be used (37). Pet birds can be infected with M. polymorphism (RFLP) typing, using the insertion sequence IS 1245
genavense, which just like M. avium silvaticum, requires as probe, allows for the classification as a “bird type” isolate (23,
mycobactin supplementation for growth. LJ slants supplemented 36). Isolates from the same serotype can belong to different
with mycobactin should be set up side by side with non­ IS1245RFLP types (36); however, available probes do not provide
supplemented LJ slants. for differentiation of serovars (9).

AGENT IDENTIFICATION Incubation


An atmosphere of 5-10% carbon dioxide (CO2) and a temperature
Smears and Section of 41 C are optimum for M. avium sbsps. avium, but 37 C is
Acid-Fast Stains. In domestic poultry, M. avium infection may be satisfactory. Cultures can be transferred to an air incubator after 7
diagnosed when acid-fast bacilli are demonstrable in smears of days of CO2 incubation. Commercial sachet CO2 generators are
typical lesions. The Kinyoun modification of the Ziehl-Neelsen acceptable alternatives in the absence of CO2 incubators. Cultures
stain is recommended because the procedure is performed without should be examined twice a week for the first 4 wk (removing any
heat (37) (Table 14.2). Using new slides, make thin smears from with contamination) and then once a week for 8 wk or longer if
scrapings of cut surfaces of lesions; air-dry and fix by flaming for 1 there are positive smears or histopathology. This regime will also
sec. Cover the slide with a strip of filter paper and flood with allow for the isolation of other Mycobacterium spp. present in pet
carbolfuchsin solution for 5 min. Wash with water and carefully birds.
remove paper. Apply decolorizer until thick areas are clear and no
more stain flows off. Wash with water. Flood with counterstain for Growth and Morphologic Characteristics
1 min. Wash with water, drain, and blot with new paper or air-dry. In lesions, M, avium sbsps. avium is a short (0.5-1.5-pm) acid-fast
The acid-fast bacilli in lesions will be red coccoid or bacillary cells, rod. In cultures, M. avium sbsps avium may develop as acid-fast
0.5-1.0 pm long, occurring singly or in large clumps. branching filaments up to 10 cm in length. Growth can be seen as
Examining sections from acid-fast bacilli may be less fruitful than soon as 14-21 days on solid medium incubated at 37 C
examining smears because of the small amount of tissue seen per (temperature range 35 to 41 C). M. avium sbsps. hominissuis will
field. For histopathology, a technique should be chosen that permits grow at 45 C while isolates of M. avium sbsps. avium will not grow
study of the lesion and staining for acid-fast bacilli as described by at this temperature; is the only way to presumptively distinguish
Feldman (13). between the two subspecies without the aid of molecular
diagnostics (23). Colonies are flat, smooth, and glistening; rough,
with irregular edge and non-pigmented variants are common with in
the same culture. The color varies from colorless to light buff to
slightly yellowish, which becomes more prominent with age (3,12).
In liquid medium, the growth is turbid, with sediment, but no
pellicle.

54
Chapter 14 Tuberculosis

Biochemical Characteristics (Gene-Probe, San Diego, CA, USA) System and the INNO-LiPA
Mycobacterium avium does not produce niacin, does not hydrolyze Mycobacteria v2 (Innogenetics, Ghent, Belgium) are among the
Tween 80, does not produce urease and does not reduce nitrate, but most widely used. They allow for species identification after
does reduce tellurite (5). Acid-fast bacteria isolated from typical bacterial culture (14). PCR has the advantage of being a very fast
tuberculosis lesions in poultry and having these features can be and specific test allowing results to be obtained within hr of the
assumed to be M. avium sbsps. avium. sample arriving at the laboratory without the need for culture. There
Niacin Production. With rare exceptions of the slowly growing are many published validated assays that can be used if the
mycobacteria, only M. tuberculosis yields a positive reaction for diagnostic laboratory has molecular diagnostic capability (4, 17, 22,
niacin. A luxuriant 3-4-wk growth on Lowenstein-Jensen medium 28, 29, 34, 35, 44). In the clinical veterinary laboratory is becoming
or a 2-wk growth in Proskauer-Beck liquid medium is necessary to widely accepted that the future of mycobacteria identification lies in
test for niacin production. Add 1 ml of sterile water to the growth the use of PCR due to its speed and accuracy.
on solid medium and allow to stand for 20 min on a horizontal
slant; add 1 ml of aniline solution (Table 14.3) and, after 5 min, add Typing of Isolates
1 ml of the cyanogen bromide solution (Table 14.3). A yellow Serological typing of isolates using agglutination allows for the
color will appear if niacin is present. This method is rarely recognition of a least 28 serovars of M. avium, and
performed any more as impregnated paper strips are commercially Mycobacterium intracellulare and is based on their surface
available. For the paper strip method, add water as described and glycopetidolipids. However, not all isolates can be reliably typed
stand for 20 min for extraction, collected 0.5ml of the water and using this method as some are non-typable. Isolates belonging to
placed in a sterile container with the paper strip, cap fast and let it serovars 1, 2 and 3 are ascribed to M. avium sbsps. avium, serovars
stand for 15min. A positive reaction will show as yellow color of 4 to 6, 8 to 11 and 21 to M avium sbsps. hominissui, and servars 7,
the liquid (not of the strip). 12 to 20 and 22 to 28 to M. intracellualre isolates (1). Serovar
Urease Hydrolysis. M. avium and M. intracellulare do not prevalence in poultry varies among countries and host species (21).
hydrolyze urea. This test is useful in distinguishing some pigmented Newer typing methods using the ELISA format detecting cell-wall-
strain M. avium from M. scrofulaceum, and other slow growing specific targets and using monoclonal antibodies are currently
mycobacteria. The test media is prepared by mixing 1 part of urea available for the major serovars as well as thin-layer
agar base concentrate with 9 parts of distilled water and dispensed chromatography of lipids. Procedures for preparing the antisera and
in 4ml volumes into tubes. A loopful of bacteria is emulsified in a for conducting the tests have been described in detail elsewhere
test tube containing this media and incubated at 37 C for 3 days. A (31). Molecular typing of isolates, is also useful; pulse field gel
positive reaction is indicated by a pink or red color change. electrophoresis and large DNA restriction fragment length
Tween 80 Hydrolysis. Mycobacterium avium does not hydrolyze polymorphism (RFLP) are very discriminatory. RFLP typing, using
Tween 80 in 10 days, whereas the similar-appearing the insertion sequence IS 1245 as probe, has recently been shown to
Mycobacterium terrae, Mycobacterium gas tri, and Mycobacterium allow for the classification as a “bird type” isolate (23, 36). Isolates
triviale will do so in less then 5 days (36). Inoculate substrate from the same serotype can belong to different IS 1245 RFLP types
(Table 14.4) with a 5-mm loopful of bacteria. Hydrolysis of the (36); however, available probes do not provide for differentiation of
Tween 80 is indicated when the solution becomes pink. Observe at serovars (8). Serotyping in combination with RFLP may be useful
24hrs, 5 days, and 10 days and record color change. M. avium will in epidemiological studies.
produce no change in color.
Nitrate Reduction. Mycobacterium avium does not reduce nitrate, Pathogenicity
but some slow-growing mycobacteria, particularly M. triviale, may Intracellular survival of M. avium is due to this bacterium’s ability
reduce nitrate. Suspend a 5-mm loopful of 10-to-14-day-old to inhibit macrophage phagosomal maturation. This has been
growing culture in 0.5 ml of sterile water and add 2 ml of buffered mainly attributed to the ability of the organism to inhibit
sodium nitrate solution (Table 14.5); place in a water bath at 37 C phagolysosome fusion in macrophages and the presence of an
for 2 hr, and then add 1 drop of the HC1 solution, 2 drops of the electron-transparent extracellular capsule following the
sulfanilamide solution, and 2 drops of the naphthylaminediamine-2 accumulation of glycopeptidolipids in the cell envelope (30). These
HC1 solution. Cultures of M. avium sbsps. avium reveal no color glycopeptidolipids have been found to be relatively resistant to
change, indicating a negative test. If nitrate is reduced, a deep pink macrophage degradation; therefore, they persist during infection
color will occur. and may play a role in altering host response.
Tellurite Reduction. Mycobacterium avium gives a positive test Intravenous injection of 0.01 mg of growing culture into chickens
for tellurite reduction, whereas the superficially similar M. terrae, or rabbits results in disseminated disease with enlargement of the
M. gas tri, and M. triviale do not. The medium used is spleen and liver and death in 30-60 days. Subcutaneous or
commercially available Middlebrook 7H9 liquid medium or intramuscular injections of this amount of inoculum cause disease
Proskauer-Beck medium without glycerol but with 5% serum. The that progresses more slowly, but is still fatal. In guinea pigs, 0.01
medium is dispensed in screw-capped tubes in 5.0-ml amounts. mg given subcutaneously causes only a local abscess that eventually
Inoculate with a 10 to 14-day-old growing culture using a 5-mm may heal (13). M. avium can cause disease and tuberculin
loop. Incubate for 7-10 days, then add 0.2 ml sterile 0.2% hypersensitivity in many species of mammals, including humans
potassium tellurite solution and reincubate for 3 days. A definite (42). Strains of M. avium serovar 1 have been isolated in patients
black deposit on the bottom of the tube indicates reduction of the with acquired immune deficiency syndrome (15).
potassium tellurite. Laboratories with only occasional need to
identify M. avium or other mycobacteria may send cultures to a SEROLOGIC DETECTION IN THE HOST
reference laboratory for supplemental tests.
Several serologic tests for diagnosis have been described, but their
Molecular identification practicality must await further studies (38). An enzyme-linked
Mycobacterial culture is the gold standard for definitive diagnosis immunosorbent assay has been developed (40).
of mycobacteriosis in birds after the microscopic examination of The tuberculin test is a satisfactory means of detecting Μ avnat
tissues and/or exudates. However, these methods are slow and infection in a flock of chickens. The M. avium purified protein
cannot reliably determine the exact species or subspecies (34, 35). derivative tuberculin prepared by the U.S. Department of
Molecular probes, such those of the commercial assays AccuProbe Agriculture is used without dilution. Lyophilized purified protein
55
Susan Sanchez and Richard M. Fulton

derivative tuberculin must be reconstituted according to directions and therefore would not have acid-fast bacilli in smears or in
of the manufacturer. With a 25-gauge needle, about 0.05 ml of examined sections of lesions. Thus, smears or histopathology are
tuberculin is injected into the skin of one wattle to form a bleb or diagnostic in these instances. In cases of persistent unexplained
blanched area. Each bird is examined in 48 hr. A positive test is death loss within a flock where birds die in the early stages of
indicated by edema of the wattle, with swelling up to five times the infection without granuloma formation, many birds may have to be
normal thickness, as compared with the other wattle (38). Chickens examined over time to confirm the presence of Mycobacterium
with extensive tuberculous lesions may fail to react. In early stages avium subsp. avium.
of the infection, the tuberculin test may be positive before any
grossly visible lesions are present. The tuberculin test may be APPENDIX I
applied at 1 or 2-mo intervals for monitoring the occurrence of the
disease; but because each bird must be handled twice, tuberculin Sodium Hydroxide decontamination method
testing may not be practical. Decontamination is necessary because any extraneous
microorganisms will contaminate and overgrow the culture of
DIFFERENTIATION FROM CLOSELY RELATED AGENTS slowly growing mycobacteria. Place 10-20 ml of tissue suspension
in the bottom of a 50-ml screw-capped centrifuge tube and add an
In chickens and in other birds, any disease that causes granulomas equal amount of decontaminant (Table 14.1).
or produces nodules may mimic avian tuberculosis grossly. Thus, Agitate for 5-10 sec on a vortex mixer. Allow to stand at room
differentiation from other causes require further analysis. Bacteria, temperature for 15 min; tissue with evidence of contamination can
such as Escherichia coli, Staphylococcus sp., Enterococcus sp. and stand for no more than 25 min. Neutralize with HC1 and then
Salmonella sp., and fungal agents such as Aspergillus sp. produce centrifuge for 10 min at 1,000 x g. Do not use the centrifuge brake.
granulomas that may be confused with avian tuberculosis. (If smears of the original material reveal one or more acid-fast
Neoplasms whether caused by viruses, such as Marek’s disease bacilli per field, concentration by centrifugation is not necessary.)
virus or avian leukosis virus, or spontaneously occurring may Remove the supernatant fluid aseptically and place in 5% phenol.
mimic avian tuberculosis by forming solid masses within the Transfer the sediment to slants of solid media with a swab or a 5-
parenchyma or on the serosal surface of organs. None of these mm loop.
diseases mimicking avian tuberculosis will contain acid-fast bacilli
Table 14.1. Reagents of Kinyoun stain for acid-fast bacilli.

Constituent Amount

Carbolfuchsin solutionA
Basic fuchsin 4.0 g
Phenol (melted) 8.0 ml
Ethyl alcohol (95%) 20.0 ml
Distilled water 68.0 ml

Decolorizer
HC1 (concentrate) 12.0 ml
Ethyl alcohol (95%) 388.0 ml

Counterstain
Methylene blue 4.0 g

Tap water 400.0 ml

A Allow mixture to stand for 24 hr, then filter through paper and store in a stoppered bottle.

Table 14.2. Aniline and cyanogen bromide solutions for the niacin test.

Constituent Amount

Aniline solution
Aniline 4.0 g
Alcohol (absolute) 96.0 ml

Cyanogen bromide solution


Cyanogen bromide 10.0 g
Distilled water 90.0 ml

Table 14.3. Substrate for Tween 80 hydrolysis test*1.

Constituent Amount

Phosphate buffer, 0.067 M, pH 7.0


100.0 ml
Tween 80
0.5 ml
Neutral red, 0.1% aqueous solution 2.0 ml

ADispense in 4.0-ml amounts in screw-capped tubes and autoclave for 10 min at 121 C. The solution should have an amber color.

56
Chapter 14 Tuberculosis

Table 14.4. Reagents for the nitrate reduction test.

Constituent Amount

Buffered nitrate solution


NaNO3 0.085 g
KH2PO4 0.117 g
Na2HPO4.-12H2O 0.485 g
Distilled water 100.0 ml

Autoclave for 10 min at 121 C


HC1 (concentrate) 50.0 ml
Distilled water 50.0 ml
Sulfanilamide 0.2%
Naphthylaminediamine-2 HC1 0.1%

Table 14.5. Reagents used for decontamination of samples.

Constituent Amount

Decontaminant
NaOH (reagent-grade) 20.0 g
Sterile distilled water 1000.0 ml
Neutralizer
HC1 (concentrate) 82.5 ml
Sterile distilled water 917.5 ml
Bromcresol purple solution 4.5 ml

Bromcresol purple solution


Bromcresol purple 1-2 g
Alcohol (95%) 50.0 ml
Distilled water 50.0 ml

ACKNOWLEDGEMENT lymphoid systems in avian tuberculosis. Am. J. Path. 64:-97-122,1971.


11. Cromie, R. L., M J. Brown, N. A. Forbes, J. Morgan and J. L. Stanford.
The authors wish to thank Charles O. Thoen for previous contributions to Av. Pathology. 22:617-630,1993.
this chapter. 12. Engbaek, H. C., E. H. Runyon, and A. G. Karlson. Mycobacterium
avium Chester: designation of the neotype strain. Int. J. Syst. Bacteriol.
REFERENCES 21:193-196. 1971.
13. Feldman, W. H. Avian tuberculosis infections. Williams & Wilkins
1. Bartos, Μ, P. Hlozek, P. Svastova, L. Dvorska, T. Bull, L. Matlova, I. Co., Baltimore, Md. pp. 116-138. 1938.
Parmova, I. Kuhn, J. Subbs, M Moravkova, J. Kintr, V. Beran, I. 14. Gerhold, R. W., and J. R. Fisher. Avian tuberculosis in a wild turkey.
Melicharek, M Ocepek and I. Pavlik Identification of members of Av. Diseases 49:164-166,2005
Mycobacterium avium species by ACCU-Probes, serotyping, and single 15. Good, R. C. Opportunistic pathogens in the genus Mycobacterium.
IS900, IS907, IS7245 and IS907-flanking region PCR with internal Annu. Rev. Microbiol. 39:347-369. 1985.
standards. J. Microbiol. Meth. 2005 Jul 29; [Epub ahead of print] 16. Gonzalez, M. A. Rodriguez-Bertos, I. Gimeno, J. M Flores and M
2. Beard, P. Μ, M J. Daniels, D. Henderson, A. Pirie, K. Rudge. D. Pizarro. Outbreak of avian tuberculosis in 48-wk-old commercial layer hen
Buxton, S. Rhind, A. Creig, M R. Hutchings, I. McKendrik, K. Stevenson, flock. Av. Diseases 46:1055-1061. 2002.
and J. M Sharp. Paratuberculosis infection of non-ruminant wildlife in 17. Gyimesi, Z. S., I. H. Stalis, J. M Miller, and C. O. Thoen. Detection of
Scotland. J. Clin. Microbiol. 39:1517-1521. 2001. Mycobacterium avium in formalin-fixed tissues of captive birds using
3. Belisle, J. T., and P. J. Brennan. Molecular basis of colony morphology polymerase chain reaction. J. Zoo. Wildl. Med. 30:348-353. 1999.
in Mycobacterium avium. Res. Microbiol. 145(3):237-242. 1994. 18. Hoop, R. K., E. C. Bottger, P. Ossent, and M salfinger.
4. Boian, Μ, T., E. Avaniss-Aghajani, R Walker, T. Aronson, T. Tran, N. Mycobacteriosis due to Mycobacterium genavense in six pet birds. J. Clin.
Glover, O. G. W. Berlin, L. Woods, C. Brunk, J. L. Li, S. Froman, and A. Microbiol. 31:990-993. 1993.
Holtzman. Identification of Mycobacterium genavense in intestinal tissue 19. Hoop, R. K., E. C. Bottger, and G. E. Pfyffer. Etiological agents of
from a parakeet using two polymerase chain reaction methods: are pets are mycobacteriosis in pet birds between 1986 and 1995. J. Clin. Microbiol.
reservoir of infection in AIDS patients? AIDS 111:255-256. 1997. 34:991-992. 1996.
5. Boughton, E. Tuberculosis caused by Mycobacterium avium. Vet. Bull. 20. Kiehn, T. E., H. Hoefer, E. C. Bottger, R. Ross, M. Wong, F. Edwards,
39:457^165. 1969. N. Antinoff, and D. Armstrong. Mycobacterium genavense infections in pet
6. Britt, J. Ο., E. B. Howard and W. J. Rosskopf. Psittacine tuberculosis. animals. J. Clin. Microbiol. 34:1840-1842. 1996.
Cornell Vet. 70:218-225,1980. 21. Meissner, G., J. Viallier, and D. Coullioud. Identification serologique
7. Buogo, C. H., L. Bacciarinini, N. Robert, T. Bodmer, and J. Nocolet. de 1590 souches de Mycobacterium avium isolees en France et en
Presence of Mycobacterium genavense in birds. Schweiz. Arch. Tierheilkd. Allamagne Federate. Ann. Microbiol. (Inst. Pasteur) 129A: 131-137. 1978.
139:397-402. 1997. 22. Mendenhall, Μ K., S. L. Ford, C. L. Emerson, R. A. Wells, L. G.
8. Com J. L., E. J. B. Manning, S. Sreevatsan, and J. R. Fisher. Isolation of Gines, and I. S. Eriks. Detection and differentiation of Mycobacterium
Mycobacterium avium subsps. paratuberculosis from free-ranging birds and avium and Mycobacterium genavense by polymerase chain reaction and
mammals on livestock premises. Appl. Environ. Microbiol. 71: 6963-6967. restriction enzyme digestion analysis. J. Vet. Diagn. Invest. 12 :57-60. 2000.
2005. 23. Mijs, W., P. de Haas, R. Rossau, T. Van Der Laan, L. Rigouts, F.
9. Crawford, J. T. Development of rapid techniques for identification of Portaels and D. van Soolingen. Molecular evidence to support a proposal to
Mycobacterium avium infection. Res. Microbiol. 145(3): 177-180. 1994. reserve the designation Mycobacterium avium subsp. avium for bird-type
10. Cheville, N. F. and W. D. Richards. The influence of thymic and bursal isolates and “M. avium subsps. hominissuis ” for the human/porcine type of
M. avium. Int. J. Syst. Evol. Microbiol. 52:1505-1518. 2002.

57
Susan Sanchez and Richard M Fulton

24. Montali, R. J., M Bush, C. O. Thoen, and E. Smith. Tuberculosis in 36. Thegerstrom J., B. Marklund, S. Hoffner, D. Axelsson-Olsson, J.
captive exotic birds. J. Am. Vet. Med. Assoc. 169:920-927. 1976. Kauppinen, and B. Olsen. Mycobacterium avium with the bird type IS 1245
25. Panigraphy B., F. D. Clark, and C. F. Hall. Mycobacteriosis in RFLP profile is commonly found in wild and domestic animals, but rarely in
psittacine birds. Avina Dis. 27:1166-1168. 1983. humans. Scjnd. J. Infect. Dis. 37:15-20. 2005.
26. Painter, K. S., Avian tuberculosis caused by Mycobacterium avium 37. Thoen, C. O. Mycobacterium. In: Diagnostic procedures in veterinary
serotype 3 in captive wildfowl. Vet. Record. 4557-458,1997. bacteriology and mycology, 5th ed. G. C. Carter and J. L. Cole, eds.
27. Peavy, G. M., S. Silverman, E. B. Howard, R. S. Cooper, L. J. Rich and Academic Press, San Diego, Calif, pp. 287-298. 1990.
G. N. Thomas. Pulmonary tuberculosis in a sulfur-crested cockatoo. J. Am. 38. Thoen, C. O. Tuberculosis. In: Diseases of poultry. 10th ed. B. W.
Vet. Med. Assoc. 69:915-919,1976. Calnek, H. J. Bames, C. W. Beard, L. R. McDougald, and Y. M Saif, eds.
28. Portaels F., L. Realini, L. Bauwens, B. Hirschel, W. M Meyers, and W. Iowa State University Press, Ames, Iowa. pp. 167-178. 1997.
De Meurichy. Mycobacteriosis caused by Mycobacterium genavense in 39. Thoen, C. O., and R. Chiodini. Mycobacterium. In: Pathogenisis of
birds kept in a zoo: 11-year survey. J. Clin. Microbiol. 34:319-323. 1996. bacterial infections in animals C. L. Gyles and C. O. Thoen, eds. Iowa
29. Ramis, A., L. Ferrer, A. Aranaz, E. Liebana, A. Mateos, L. Dominguez, State University Press, Ames, Iowa. pp. 44-56. 1993.
C. Pascual, J. Fdez-Garayazabal, and M D. Collins. Mycobacterium 40. Thoen, C. O., W. G. Eacret, and E. M. Himes. An enzyme-labeled
genavense infection in canaries. Av. Diseases. 40:246-251. 1996. antibody in chickens infected with Mycobacterium avium serotype 2. Avian
30. Rastogi, N., and W. W. Barrow. Cell envelope constituents and the Dis. 22:162-165. 1978.
multifaceted nature of Mycobacterium avium pathogenicity and drug 41. Thoen, C. Ο., E. M. Himes, and A. G. Karlson. Mycobacterium avium
resistance. Res. Microbiol. 145:243-252. 1994. complex. In: The mycobacteria: a sourcebook. G. P. Kubica and L. G.
31. Schaefer, W. B. Serologic identification and classification of the Wayne, eds. Marcel Dekker Inc., New York. pp. 1251-1275. 1984.
atypical mycobacteria by their agglutination. Am. Rev. Respir. Dis. 42. Thoen, C. O., and D. E. Williams. Tuberculosis, tuberculoidoses and
92(2): 85-93. 1965. other mycobacterial infections. In: Handbook of zoonoses, 2nd ed.. G. W.
32. Smith, I. M., S. T. Licence, and R. Hill. Effect of repeated injections of Beran, ed. CRC Press, Boca Raton, Fla. pp. 41-60. 1994.
iron dextran on the haematological, serological and pathological changes in 43. Thorel, M F., H. Huchzermeyer, R. Weiss, and J. J. Fontaine.
experimental avian tuberculosis. Res. Vet. Science. 25:284-289,1978. Mycobacterium avium infections in animals. Literature review. Vet. Res.
33. Tell L. A., L. Woods, And R.I. Cromie. Mycobacteriosis in birds. Rev. 28: 439-447.
Sci. Tech. Off. Int. Epizoot. 20:180-203. 2001. 44. Thornton C. G., M R. Cranfield, K. M MacLellan, T. L. Brink, Jr., J. D.
34. Tell L. A., C. M Leutenegger, R. S. Larsen, D. W. Agnew, L. Keener, Strandberg, E. A. Carlin, J. B. Torrelles, J. N. Maslow, J. L. B. Hasson, D.
M L. Needham, and B. A. Rideout. Real-time polymerase chain reaction M Heyl, S. J. Sarro, D. Chatteijee, and S. Passen. Processing postmortem
testing for the detection of Mycobacterium genavense and Mycobacterium specimens with Cig-caboxypropylbetaine and analysis by PCR to develop
avium Complex species in avian samples. Av. Diseases. 47:1406-1415. and antemortem test for Mycobacterium avium infections in ducks. J. Zoo
2003a. Wildl. Med. 30:11-24. 1999.
35. Tell L. A., A. L. Woods, and J. Foley. M L. Needham and R. L. 45. Wayne, L., R. Good, M Krichevsky, Z. Blacklock, H. David, D.
Walker. Diagnosis of mycobacteriosis: comparison of culture, acid-fast Dawson, W. Gross, J. Hawkins, V. Levy-Frebault, C. McManus, F. Portaels,
stains, and polymerase chain reaction for the identification of S. Rusch-Gerdes, K. Schroder, V. Silcox, M Tsukamura, L. Van den Breen
Mycobacterium avium in experimentally inoculated Japanese quail and M Yarkrus. Fourth report of the cooperative open-ended study of slowy
(Coturnix japonica}. Av. Diseases. 47:444-452. 2003b. growing mycobacteria of die international Working Group on mycobacterial
Taxonomy. Int. J. Syst. Bacteriol. 41: 463-472. 1991.

58
15
MYCOPLASMOSIS
Stanley H. Kleven

SUMMARY. The pathogenic avian mycoplasmas, Mycoplasma gallisepticum, Mycoplasma synoviae, Mycoplasma meleagridis, and
Mycoplasma iowae, are associated with respiratory disease, synovitis, poor performance, skeletal deformities, or embryo mortality in
chickens and turkeys. These mycoplasmas are egg transmitted, and (except for M. iowae) are covered by control programs administered
under the National Poultry Improvement Plan. The majority of broilers and turkeys in the United States are reared free of infection.
Mycoplasma gallisepticum and M. synoviae are frequently found in commercial layers; egg production losses due to M. gallisepticum are
generally controlled by vaccination.
Agent Identification. Serologic diagnosis is frequently verified by isolation and identification of the etiologic agent or by polymerase chain
reaction. All of the pathogenic avian mycoplasmas exhibit significant intraspecies variability in virulence, tissue tropism, and antigenic
makeup. DNA fingerprinting using random amplified polymorphic DNA or targeted gene sequencing may be utilized for epidemiologic
studies or for identifying specific strains, such as vaccine strains of M. gallisepticum.
Serologic Detection in the Host. Serum plate agglutination tests or enzyme-linked immunosorbent assays are used for screening and
preliminary diagnosis, and reactors are generally confirmed by hemagglutination inhibition. No serologic tests are available for M. iowae.

INTRODUCTION disappear from lesions after a few weeks, but they persist in the
upper respiratory tract. For culture of embryonating eggs for
Mycoplasmas are small prokaryotic organisms lacking a cell wall. M. gallisepticum, M. synoviae, or M. meleagridis, samples of yolk
Numerous species of avian Mycoplasma have been described and that contain a portion of the yolk membrane should be included. For
characterized. Table 15.1, adapted from Jordan (26) with M. iowae, culture of the esophagus of pipped embryos or 1-day-old
information from other sources (6, 7, 8, 17, 18, 34, 44, 45), lists poults, or from cloaca, phallus or semen of breeding adult turkeys
these species along with their biochemical reactions and usual host. should be obtained.
At least four species of Mycoplasma apparently are isolated from In the case of M. meleagridis, culture of the phallus or vagina of
ratites. Future additions to this list are likely as additional isolates breeding-age turkeys may be useful. In some cases, culture of other
are characterized. tissues such as oviduct, cloaca, or brain may be desirable.
When specimens must be stored or shipped to a laboratoiy,
CLINICAL DISEASE inoculate broth medium with the specimen on the farm and ship by
overnight carrier to the laboratory. Pre-wetting of the swabs in broth
Four avian mycoplasmas are commonly recognized as poultry medium or PBS may improve recovery rates. Inoculated broth
pathogens: Mycoplasma gallisepticum, Mycoplasma synoviae, cultures are viable for several days at room temperature, although
Mycoplasma meleagridis, and Mycoplasma iowae. they should be incubated as soon as possible. Swabs may be kept
M. gallisepticum is a cause of respiratory disease and egg moist at refrigerator temperature up to 24 hr if necessary.
production drops in chickens, turkeys, and other avian species, and Alternatively, tracheas, fluids, or tissues can be collected and frozen
M. gallisepticum is the most economically important of the avian on dry ice. Isolation of mycoplasmas is generally successful when
mycoplasmas (39). Severe airsacculitis, coughing, rales, and poor specimens are preserved on dry ice and arrive at the laboratory still
growth may be observed in broilers or turkeys infected with frozen. Shipment on wet ice may be necessary in some cases, but
M. gallisepticum, with heavy condemnations from airsacculitis at specimens should be cultured within 24 hr, if possible.
processing. Swollen sinuses are commonly seen in turkeys. No special procedures for serum collection are needed for
Mycoplasma gallisepticum infection is widespread in house finches serologic testing. However, nonspecific agglutination may occur
(Carpodacus mexicanus) in the eastern United States (40, 41), and with frozen sera, so fresh serum should be used for serum plate
has been shown to be a cause of conjunctivitis. Mycoplasma agglutination tests. Either fresh or frozen serum is suitable for other
synoviae causes lesions of synovitis in chickens, turkeys, and other serologic tests.
species and is also involved in respiratory disease (31).
Mycoplasma meleagridis, found only in turkeys, causes respiratory PREFERRED CULTURE MEDIA AND SUBSTRATE
disease in young turkeys and is involved in stunting, poor
feathering, and leg problems (11). Mycoplasma iowae is a cause of No single medium formulation has been universally accepted as
late embryo mortality and reduced hatchability in turkeys (9). optimum for the growth of all avian Mycoplasma species.
Mycoplasma gallisepticum, M. synoviae, M. meleagridis, and Mycoplasmas are fastidious organisms that require a protein-based
M. iowae are egg-transmitted, and may be spread laterally by direct medium enriched with 10%—15% serum or serum factors. Glucose
or indirect contact. The mode of egg transmission of M. meleagridis is fermented by M. gallisepticum, M. synoviae, M. iowae, and
is venereal; infection of the phallus of the male results in several other species and is a common supplement. A yeast source
contaminated semen, which contaminates the oviduct of the female. factor may be beneficial and is usually supplied by commercial
yeast autolysate or by fresh yeast extract. Mycoplasma synoviae
SAMPLE COLLECTION requires nicotinamide adenine dinucleotide (NAD), and cysteine
hydrochloride is added as a reducing agent for the NAD.
During the acute stages of infection (up to 60-90 days Mycoplasmas are resistant to penicillin (because they lack a cell
postinfection), the population of organisms in the upper respiratoiy wall) and are relatively resistant to thallium; most media contain
tract and the incidence of infection in the flock are high. In such these components to avoid bacterial and mycotic contamination. In
cases 5-10 cultures from the trachea or choanal cleft are often general, horse serum should be used in media for M. meleagridis,
sufficient. However, in chronic cases or when infection with swine serum for M. synoviae, and either horse or swine serum for
Mycoplasma strains of low pathogenicity is suspected, as many as M. gallisepticum. Serum should be heat inactivated at 56 C for 30
30-100 individuals should be cultured. When airsacculitis or min. Because of the variability in the ability of batches of serum to
synovitis is present, lesions should be cultured. Organisms tend to support growth, each batch should be tested before use.
59
Stanley H. Kleven

Table 15.2 shows examples of commonly used formulations for A commonly used method for Mycoplasma identification is
media that have been shown to support the growth of all avian growth inhibition (12). The surface of agar plates is evenly coated
mycoplasmas and acholeplasmas. These are modifications of media with a moderate inoculum (usually a 1:10 to 1:1000 dilution of a
described by Frey et al. (19) and Bradbury (5). broth culture; too large an inoculum may give false negative
Cotton swabs from trachea, air sac, or other tissue, or results), and filter paper discs impregnated with specific
approximately 0.1 ml of fluid from swollen sinuses or joints, are hyperimmune serum are placed on the surface. After incubation at
inoculated into 3-5 ml of broth medium, and the swab is discarded. 37 C for several days, a zone of inhibition of colony formation
Pieces of air sac, trachea, or other tissue may be inoculated directly surrounding the disc indicates a positive test. The growth inhibition
into broth medium; however, the inoculum should be small, because procedure requires the use of pure cultures. Cultures may be cloned
tissue enzymes tend to break down glucose and cause a drop in pH, by aspirating single colonies with a Pasteur pipette and inoculating
or tissue inhibitors may be present. Therefore, too large an broth medium. After sufficient growth occurs in broth, the medium
inoculum may inhibit growth. Some laboratories make serial should be filtered through a 0.45 pm filter to eliminate clumped
dilutions of the inoculum in broth to avoid the inhibitory effects of a cells, and re-plated on agar. Cultures should be cloned up to three
large inoculum. Sometimes agar plates are inoculated times to ensure purity.
simultaneously with the broth, but broth cultures are generally more In many laboratories, direct immunofluorescence is the preferred
sensitive. Incubation is at 37 C. Ordinarily, aerobic incubation is method of identifying isolates of avian mycoplasmas (47), although
sufficient, although some laboratories may prefer to incubate under many laboratories prefer an indirect procedure. Fluorescein is used
CO2. For M. synoviae, M. gallisepticum, and other glucose to conjugate high titered specific rabbit antiserum. Two to three
fermenters, cultures are incubated until the phenol red indicator drops of diluted conjugate are placed on suspect colonies inside
changes in color from red to orange or yellow, and then plates and a plastic cylinder on an agar plate, which is incubated at 37 C for 30
fresh tubes of broth are inoculated. Do not allow broth cultures to min. Washing is accomplished by filling the cylinder with
continue to incubate after the phenol red indicator changes to phosphate-buffered saline (PBS) and removing it by gentle suction.
yellow, because mycoplasmas (especially M synoviae) may be The stained agar blocks that contain the colonies can then be placed
sensitive to low pH. For nonfermenters, cultures are plated at about on a glass slide and examined with a fluorescent microscope at 35-
5 days of incubation and again at 10-14 days. Agar plates can be *.
100 Microscopes equipped with an ultraviolet (UV) light
divided into sections so that six to eight cultures can be inoculated source transmitted through a dry darkfield condenser are useful for
on a single agar plate. Cotton swabs, microbiological loops, or this purpose; for this procedure, agar blocks should be mounted face
Pasteur pipettes can be used to inoculate agar plates. Plates are down (toward the UV light source). This setup has the advantage of
incubated in a closed jar to prevent dehydration. For M. meleagridis making location of colonies using incandescent light easy; the light
and M. iowae, primary isolation directly on agar plates may be more source can then be switched to UV to examine the colonies for
successful than inoculating broth medium. fluorescence. Epifluorescence also works well; colonies should be
Agar plates are examined for Mycoplasma colonies under low mounted face up. Immunofluorescence has the advantage of
magnification (about 35 x) using an ordinary light microscope with accuracy and speed (an isolate can be examined within 1-2 hr), and
the light intensity reduced, or with a dissecting microscope. With it can be effectively used with mixed cultures.
several of the nonpathogenic species, such as Acholeplasma
laidlawii, Mycoplasma gallinarum, or Mycoplasma gallinaceum, Molecular identification
colonies may be observed as early as 24 hr postinoculation. Polymerase chain reaction (PCR) can be used to identify the
However, with the pathogenic species (M. gallisepticum, species of avian Mycoplasma isolates. The PCR product is cut with
M. synoviae, and M. meleagridis), colonies are usually not observed one or more restriction enzymes, and the patterns can be used to
until 4—5 days of incubation at 37 C, but it some instances they may identify the species (15, 37). In addition, species-specific DNA
be present after 3 days. Mixed cultures are common, especially in probes (29) or species specific PCR primers (3, 4, 20, 24, 27, 28,
multiage commercial layers, which may make recovery of the 35, 36, 38,42,43) can be used.
pathogenic species more difficult. The most commonly encountered Mycoplasma gallisepticum strains differ in virulence and
nonpathogenic species are M. gallinarum and M. gallinaceum. antigenicity. Additional strain variation has been detected by
polyacrylamide gel electrophoresis of mycoplasmal proteins (30),
AGENT IDENTIFICATION by restriction endonuclease analysis of DNA (32), and by Southern
blot hybridization using a DNA probe prepared from the ribosomal
Tiny, smooth colonies 0.1-1 mm in diameter with dense, elevated RNA gene of Mycoplasma capricolum (48).
centers are suggestive of Mycoplasma species. Although A rapid and accurate method for DNA fingerprinting utilizes
biochemical reactions (Table 15.1) may be useful at times, arbitrary primed PCR or random amplified polymorphic DNA. This
identification of isolates is ordinarily by serologic methods, using technique utilizes short, arbitrary PCR primers, which generate
hyperimmune serum prepared in rabbits (46). For most tests, it is reproducible patterns in agarose gels (10, 13, 14, 21). This method
important to ensure that the antiserum does not contain antibodies is rapid and simple, and has proven to be very useful for rapid
against the serum present in the medium being used to propagate identification of strains of M. gallisepticum, M. synoviae, M.
the Mycoplasma isolate to be identified. This can be accomplished meleagridis, and M. iowae for epidemiologic studies. However,
by growing the antigen to be used for hyperimmunization in there may be problems with reproducibility, so strains to be
medium containing serum from the animal to be immunized. compared must be run on the same gel. Also, interpretation of
However, in most cases it is sufficient to grow antigen for preparing banding patterns which appear to be similar can be difficult.
hyperimmune serum in medium containing serum from a species Gene targeted sequencing (GTS), using PCR primers for the mgc2,
other than the serum being used in ordinary Mycoplasma medium. gapA, pvpA, and MGA 0309 genes of M. gallisepticum can be used
For example, cultures to be used for immunizing rabbits are grown to provide an accurate and reproducible method of typing of strains
in medium containing calf serum, and medium containing swine which will allow rapid global comparisons between laboratories
serum is used for all other purposes. (16). Preliminary strain identification with diagnostic PCR primers
for the mgc2 gene for M. gallisepticum or the vlhA gene for
M. synoviae by sequencing of die PCR product allows for
preliminary strain identification without the need for prior isolation
of the organism (24, 25). However, unrelated strains sometimes
60
Chapter 15 Mycoplasmosis

have identical sequences using these primers, and further M. synoviae, and M. meleagridis HI tests varies from other HI
characterization may be necessary. procedures as follows (1): Mycoplasma HI tests are usually
Amplified Fragment Length Polymorphism (AFLP) is a powerful conducted in microtiter plates, using 4 hemagglutinating units of
and reproducible tool for identification of avian Mycoplasma antigen per test using the β procedure (constant antigen-diluting
species (25) or for differentiating among strains of M. gallisepticum serum), although in practice the antigen concentration can be
(23), but the method is somewhat complex and requires DNA reduced somewhat to improve sensitivity as long as negative control
sequencing equipment and expensive software. sera still are solidly negative. The first serum dilution (1:10) is
made in buffer and is used as a serum control. All higher serum
SEROLOGIC DETECTION IN THE HOST dilutions are made directly in antigen. An alternative method is to
prepare serum dilutions in buffer and then add antigen. Some
Serologic testing is the basis of voluntary control programs within batches of antigen tend to give occasional low-titered background
each state and as part of the National Poultry Improvement Plan (1). reactions up to 1:20 to 1:40 with negative sera. In such cases, the
Screening of poultry flocks for infection with the pathogenic antigen strength should be increased enough to suppress that effect,
mycoplasmas is generally accomplished with the serum plate even at the expense of sacrificing sensitivity. Also, reading positive
agglutination (SPA) test (1). Generally, 10% of the flock (or a results only when hemagglutination is 100% inhibited helps to
minimum of 300 birds) is tested before the onset of egg production, eliminate nonspecific reactions. Generally, HI titers of 1:40 to 1:80
and approximately 30 birds per flock are tested every 60-90 days or greater are considered to be positive, but the interpretation of
thereafter. Serologic testing is also used as a laboratory diagnostic results must be done on a flock basis. The HI test is considered to
aid. be highly specific but less sensitive than the SPA test. Infected birds
The SPA test is quick, inexpensive, and highly sensitive. Infected may not test positive until 2-3 wk or longer after infection. In
birds may test positive as early as 7-10 days after infection. The test addition, antigenic variation exists among M. gallisepticum strains
involves the use of specific stained antigens for M. gallisepticum, as measured by HI. Antigen prepared from one M. gallisepticum
M. synoviae, and M. meleagridis. Antigens are available strain may not adequately detect HI antibodies in chickens infected
commercially (Intervet America, Millsboro, Del.; and Charles River with a different strain (33).
Laboratories, Wilmington, Mass.). Approximately 0.02 ml of serum Enzyme-linked immunosorbent assays have also been developed
and 0.03 ml of antigen are mixed on a glass plate. The plate is in several laboratories (16,28), and commercial test kits are
rotated for 2-3 min, and the tests are examined for visible available (IDEXX, Westbrook, Maine, and Synbiotics, San Diego,
clumping. Antigens obtained from different sources may differ in Cal.). Such systems are reliable for detecting antibodies against M.
sensitivity and specificity (2), but variations also exist between gallisepticum or M. synoviae. The kits detect antibodies at about the
batches. Occasionally, agglutination tests are insensitive for the same time after infection as the HI test. Although the quality of the
detection of M. synoviae antibodies in turkeys. commercial test kits is good, false positive results may sometimes
The greatest disadvantage of the SPA test is low specificity (false occur.
positive reactions). These have been related to medium components, In many situations, serologic testing may not give a definitive
primarily serum, adhering to the surface of the Mycoplasma diagnosis, especially when positive agglutination reactions occur
organisms used to prepare the antigen, although false positive that cannot be confirmed by HI testing. Retesting may be required
reactions may often be unexplained (2). False positive reactions are in such cases. When retesting is needed, isolation and identification
commonly seen after chickens or turkeys have been vaccinated with of the organism or PCR may result in a faster and more definitive
inactivated oil emulsion vaccines against other infectious agents, diagnosis than repeated serologic testing. No serologic tests exist
especially if there are remnants of serum in the vaccine (22). Such for avian mycoplasmas other than M. gallisepticum, M. synoviae,
false positive reactions may persist up to 4-8 wk or longer after and M. meleagridis.
vaccination. In addition to false positive reactions related to
medium components, cross-reacting antigens shared among DIFFERENTIATION FROM CLOSELY RELATED AGENTS
Mycoplasma species or between mycoplasmas and bacteria may
occur. Several cross-reacting antigens between M. gallisepticum Mycoplasma infections of poultry may be readily confused with
and M. synoviae are known to occur. other respiratory diseases and may be obvious only when present in
Flocks with SPA reactors should be confirmed as positive or conjunction with complicating respiratory infections such as
negative with the hemagglutination-inhibition (HI) test or other Newcastle disease or infectious bronchitis. Secondary bacterial
acceptable serologic test, or the agent can be detected by culture or invasion associated with airsacculitis is often significant;
by PCR. Some laboratories prefer to use serum dilutions with the Escherichia coli is the bacterial species most frequently isolated.
SPA for confirming reactors. Serial twofold dilutions of the serum Mycoplasma gallisepticum infection must be ruled out whenever
are made in a buffer such as PBS, and the SPA test is conducted on sinusitis or respiratory disease is present in turkeys, although M.
the diluted sera. Sera that react at 1:8 or 1:10 or greater are gallisepticum infection may be confused with avian influenza,
considered positive. Overall, the SPA dilution system works well, chlamydiosis, Newcastle disease, fowl cholera, or respiratory
but weak but specific SPA reactors may be negative with the serum cryptosporidiosis.
dilution test, and strong false positive reactors may react at 1:8 or Mycoplasma synoviae infection should be considered whenever
greater. synovitis lesions are present. Differentiation from infectious
Agglutination reactors are generally confirmed with the HI test. tenosynovitis, infection with Staphylococcus aureus, or other
Antigen is prepared by harvesting an actively growing culture by bacterial infection is essential. Mycoplasma meleagridis infection in
centrifugation, suspending the packed cells in a small amount of turkeys should be considered when air sac lesions are found in
PBS, and mixing with an equal volume of glycerol. Aliquots are young poults or when stunting and/or skeletal problems are present.
preserved by freezing at -70 C. Hemagglutination antigen for M. Mycoplasma iowae infection should be ruled out in cases of reduced
gallisepticum and M. synoviae are available commercially (Charles hatchability with late embryo mortality in turkeys.
River SPAFAS, Storrs, Conn.) The procedure for M. gallisepticum,

61
Stanley H. Kleven

Table 15.1. Characteristics of avian mycoplasmas from various hosts

Mycoplasma species Usual host Glucose fermentation Arginine hydrolysis

Acholeplasma laidlawiiK Various +

M. anatis Duck +

M. anseris Goose +

M. buteonis Buteo hawk +

M. cloacale Turkey +

M. columbinasale Pigeon +

M. columbinum Pigeon +

M. columborale Pigeon +

M. corogypsi Black vulture +

M. falconis Saker falcon . +

M. gallinaceum Chicken +

M. gallinarum Chicken +

M. gallisepticum Chicken and Turkey +

M. gallopavonis Turkey +

M. glycophilum Chicken +

M. gypis Griffon vulture +

M. imitans Duck, goose, partridge +

M. iners Chicken +

M. iowae Turkey + +

M. lipofaciens Chicken + +

M. meleagridis Turkey +

M. pullorum Chicken +

M. sturni European starling +

M. synoviae Chicken and turkey +

Ureaplasma galloraleQ Chicken and turkey

62
Chapter 15 Mycoplasmosis

Table 15.2. Formulations for two commonly used media for the isolation and propagation of avian mycoplasmas.
Medium Ingredient Amount
Frey’s medium BBL Mycoplasma broth base (Becton Dickinson, Sparks, Md) 22.5 g
Glucose 3g
Swine serum 120 ml
Yeast extract 35 ml
Cysteine hydrochloride® 0.1 g
Nicotinamide adenine dinucleotide (NAD)B 0.1 g
Phenol red (1%) 2.5 ml
Thallium acetate (10%)c 3ml
Penicillin G potassium9 106 units
Distilled water q.s. 1000 ml

Adjust pH to 7.8 with 20% NaOH and filter sterilize®4"

PPLO broth Difco PPLO broth without crystal violet (Becton Dickinson, Sparks, Md.) 21 g
Glucose 10g
Swine serum 150 ml
Yeast extractA 100 ml
Cysteine hydrochloride® 0.1 g
Nicotinamide adenine dinucleotide (NAD)® 0.1 g
Phenol red (1%) 2.5 ml
Thallium acetate (10%)c 3 ml
Penicillin G potassium9 106 units
Distilled water q.s. 1000 ml

Adjust pH to 7.8 with 20% NaOH and filter sterilize®4"


A Fresh yeast extract (also available commercially) is made by placing 250 g dry baker’s or brewer’s yeast in 1 liter distilled H2O and allowing to soak 1 hr. Heat to
boiling, allow to cool, and centrifuge at 3000 x g for 20 min. Decant the supemate and adjust the pH of the fluid to 8.0 with 0.1 M NaOH. Clarify by filtration
through coarse filter paper and sterilize by filtration. Dispense in aliquots and store at 20 C.
B Reduced NAD is required for M synoviae only. A 1% solution each of NAD and cysteine is mixed in equal parts, and 20 ml is added per liter of medium
c For potentially contaminated specimens, add an extra 20 ml of 1% thallium acetate per liter of medium to bring the total concentration to 1:2000. Add thallium
acetate to the distilled H2O before the other ingredients to prevent precipitation of protein.
D For potentially contaminated material, an extra 2 χ 106 units of penicillin may be added per liter of medium; 200 mg to 1 g of ampicillin per liter of medium will
substitute.
E Alternatively, all ingredients except cysteine/NAD, serum, penicillin, and yeast extract may be autoclaved at 121 C for 15 min, and the remaining ingredients are
added aseptically after sterilization by filtration.
F For agar medium, use 1% of a purified agar such as Difco purified agar. All components except cysteine/NAD, serum, and penicillin are sterilized by autoclaving
at 121 C for 15 min. Cool to 50 C and aseptically add the above components that have been sterilized by filtration and warmed to 50 C. Mix and pour plates to a
depth of approximately 5 mm

REFERENCES 6. Bradbury, J.M. and M Forrest. Mycoplasma cloacale, a new species


isolated from a turkey. Int. J. Syst. Bacteriol. 34:389-392. 1984.
1. Anonymous, National Poultry Improvement Plan and Auxiliary 7. Bradbury, J.M, M. Forrest, and A. Williams. Mycoplasma lipofaciens, a
Provision. Vol. APHIS-91-55-063. Washington, DC. 2004. new species of avian origin. Int. J. Syst. Bacteriol. 33:329-335. 1983.
2. Avakian, A.P., S.H. Kleven, and J.R. Glisson. Evaluation of the 8. Bradbury, J.M, F. Jordan, T. Shimizu, L. Stipkovits, and Z. Varga.
specificity and sensitivity of two commercial enzyme-linked immunosorbent Mycoplasma anseris sp. nov found in geese. Int. J. Syst. Bacteriol. 38:74-76.
assay kits, the serum plate agglutination test, and the hemagglutination­ 1988.
inhibition test for antibodies formed in response to Mycoplasma 9. Bradbury, J.M and S.H. Kleven, Mycoplasma iowae infection. In:
gallisepticum. Avian Dis. 32:262-272. 1988. Diseases of Poultry, 11th, ed Y.M Saif, et al., Editors. Iowa State
3. Bencina, D., M Drobnic-Valic, S. Horvat, M Narat, S.H. Kleven, and P. University Press: Ames, Iowa. p. 766-771. 2003.
Dove. Molecular basis of the length variation in the N-terminal part of 10. Charlton, B.R., A.A. Bickford, R.L. Walker, and R. Yamamoto.
Mycoplasma synoviae hemagglutinin. FEMS Microbiol Lett. 203:115-23. Complementary randomly amplified polymorphic DNA (RAPD) analysis
2001. patterns and primer sets to differentiate Mycoplasma gallisepticum strains. J.
4. Boyle, J.S., R.T. Good, and C.J. Morrow. Detection of the turkey Vet. Diag. Invest. 11:158-61. 1999.
pathogens Mycoplasma meleagridis and M iowae by amplification of genes 11. Chin, R.P., G. Y. Ghazikhanian, and I. Kempf. Mycoplasma meleagridis
coding for ribosomal-RNA. J. Clin. Microbiol. 33:1335-1338. 1995. infection. In: Diseases of Poultry, 11th. ed Y.M. Saif, et al., Editors. Iowa
5. Bradbury, J.M Rapid biochemical tests for characterization of the State University Press: Ames, Iowa. p. 744-756. 2003.
Mycoplasmcfates. J. Clin. Microbiol. 5:531-534. 1977.

63
Stanley H. Kleven

12. Clyde, W.A. Mycoplasma species identification based upon growth 31. Kleven, S.H., Mycoplasma synoviae infection. In: Diseases of Poultry,
inhibition by specific antisera. J. Immunol. 92:958-965. 1964. 11th. ed. Y.M Saif, et al., Editors. Iowa State University Press: Ames, Iowa,
13. Fan, H.H., S.H. Kleven, and MW. Jackwood. Application of p. 756-766. 2003.
polymerase chain reaction with arbitrary primers to strain identification of 32. Kleven, S.H., G.F. Browning, D.M Bulach, E. Ghiocas, C.J. Morrow,
Mycoplasma gallisepticum. Avian Dis. 39:729-735. 1995. and K.G. Whithear. Examination of Mycoplasma gallisepticum strains using
14. Fan, H.H., S.H. Kleven, and MW. Jackwood. Studies of intraspecies restriction endonuclease DNA analysis and DNA-DNA hybridisation. Avian
heterogeneity of Mycoplasma synoviae, M meleagridis, and M iowae with Pathol. 17:559-570. 1988.
arbitrarily primed polymerase chain reaction. Avian Dis. 39:766-777. 1995. 33. Kleven, S.H., C.J. Morrow, and K.G. Whithear. Comparison of
15. Farmer, K.L., G.E. Hill, and S.R. Roberts. Susceptibility of a naive Mycoplasma gallisepticum strains by hemagglutination-inhibition and
population of house finches to Mycoplasma gallisepticum. J. Wildl. Dis. restriction endonuclease analysis. Avian Dis. 32:731-741. 1988.
38:282-286. 2002. 34. Koshimizu, K., R. Harasawa, I.J. Pan, H. Kotani, M Ogata, E.B.
16. Ferguson, N.M, D. Hepp, S. Sun, N. Ikuta, S. Levisohn, S.H. Kleven, Stephens, and MF. Barile. Ureaplasma gallorale sp. nov. from the
and M Garcia. Use of molecular diversity of Mycoplasma gallisepticum by oropharynx of chickens. Int. J. Syst. Bacteriol. 37:333-338. 1987.
gene-targeted sequencing (GTS) and random amplified polymorphic DNA 35. Laigret, F., J. Deaville, J.M Bove, and J. Bradbury. Specific detection
(RAPD) analysis for epidemiological studies. Microbiol. 151:1883-1893. of Mycoplasma iowae using polymerase chain reaction. Molec. Cell. Probes.
2005. 10:23-29. 1996.
17. Forrest, M. and J.M Bradbury. Mycoplasma glycophilum, a new 36. Lauerman, L.H., Mycoplasma PCR Assays, in Nucleic Amplification
species of avian origin. J. Gen. Microbiol.?597-603. 1984. Assays for Diagnosis of Animal Diseases, ed. L.H. Lauerman,
18. Forsyth, MH., J.G. Tully, T.S. Gorton, L. Hinckley, S. Frasca Jr., H.J. Editor.American Association of Veterinary Laboratory Diagnosticians:
Van Kruiningen, and S.J. Geary. Mycoplasma stumi sp. nov., from the Auburn, AL. p. 41-52. 1998.
conjunctiva of a European starling (Stumus vulgaris). Int. J. Syst. Bacteriol. 37. Lauerman, L.H., A.R. Chilina, J.A. Closser, and D. Johansen. Avian
46:716-719. 1996. Mycoplasma identification using polymerase chain reaction amplicon and
19. Frey, ML., R.P. Hanson, and D.P. Anderson. A medium for the restriction fragment length polymorphism analysis. Avian Dis. 39:804-811.
isolation of avian Mycoplasmas. Am. J. Vet. Res. 29:2163-2171. 1968. 1995.
20. Garcia, Μ, N. Ikuta, S. Levisohn, and S.H. Kleven. Evaluation and 38. Lauerman, L.H., F.J. Hoerr, A.R. Sharpton, S.M Shah, and V.L. van
Comparison of Various PCR Methods for Detection of Mycoplasma Santen. Development and application of a polymerase chain reaction assay
gallisepticum Infection in Chickens. Avian Dis. 48:125-132. 2005. for Mycoplasma synoviae. Avian Dis. 37:829-834. 1993.
21. Geary, S.J., MH. Forsyth, S.A. Saoud, G. Wang, D.E. Berg, and C.M 39. Ley, D.H., Mycoplasma gallisepticum infection. In: Diseases of Poultry,
Berg. Mycoplasma gallisepticum strain differentiation by arbitrary primer 11th. ei Y.M Saif, et al., Editors. Iowa State University Press: Ames, Iowa,
PCR (RAPD) fingerprinting. Molec. Cell. Probes. 8:311-316. 1994. p. 722-744. 2003.
22. Glisson, J.R., J.F. Dawe, and S.H. Kleven. The effect of oil emulsion 40. Ley, D.H., J.E. Berkhoff, and J.M McLaren. Mycoplasma gallisepticum
vaccines on the occurrence of nonspecific plate agglutination reactions for isolated from house finches (Carpodacus mexicanus) with conjunctivitis.
Mycoplasma gallisepticum and Mycoplasma synoviae. Avian Dis. 28:397- Avian Dis. 40:480-483. 1996.
405. 1984. 41. Luttrell, M.P., J.R. Fischer, D.E. Stallknecht, and S.H. Kleven. Field
23. Hong, Y, M. Garcia, S. Levisohn, P. Savelkoul, V. Leiting, I. investigation of Mycoplasma gallisepticum infections in house finches
Lysnyansky, D.H. Ley, and S.H. Kleven. Differentiation of Mycoplasma (Carpodacus mexicanus) from Maryland and Georgia. Avian Dis. 40:335-
gallisepticum strains using amplified fragment length polymorphism and 341. 1996.
other DNA-Based Typing Methods. Avian Dis. 49:43-49. 2005. 42. Moalic, P.M, F. Gesbert, F. Laigret, and I. Kempf. Evaluation of
24. Hong, Y., M Garcia, L. Leiting, D. Bencina, L. Dufour-Zavala, G. polymerase chain reaction for detection of Mycoplasma meleagridis in
Zavala, and S.H. Kleven. Specific Detection and Typing of Mycoplasma turkeys. Proc. World Vet. Poultry Assoc. 11:73. 1997.
synoviae Strains in Poultry with PCR and DNA Sequence Analysis 43. Morina, R., D. Bencina, and P. Dove. Polymorphisms in the 5' - vlhA
Targeting the Hemagglutinin Encoding Gene vlhA. Avian Dis. 48:606-616. gene enable differentiation of Mycoplasma synoviae strains, in International
2004. Organization for Mycoplasmology. 2002. Vienna.
25. Hong, Y., M Garcia, S. Levisohn, I. Lysnyansky, V. Leiting, P.H.M. 44. Panangala, V.S., J.S. Stringfellow, K. Dybvig, A. Woodard, F.-. Sun,
Savelkoul, and S.H. Kleven. Evaluation of Amplified Fragment Length D.L. Rose, and MM Gresham. Mycoplasma corogypsi sp. nov., a new
Polymorphism for Differentiation of Avian Mycoplasma Species. J. Clin. species from the footpad abscess of a black vulture, Coragyps atratus. Int. J.
Microbiol. 43:909-912. 2005. Syst. Bacteriol. 43:585-590. 1993.
26. Jordan, F.T.W., Recovery and identification of avian mycoplasmas, in 45. Poveda, J.B., J. Giebel, J. Flossdorf, J. Meier, and H. Kirchhoff.
Methods in Mycoplasmology. Volume Π, Diagnostic Mycoplasmology. J.G. Mycoplasma buteonis sp. nov., Mycoplasma falconis sp. nov., and
Tully and S. Razin, Editors. Academic Press: New York. p. 69-79. 1983. Mycoplasma gypis sp. nov., 3 species from birds of prey. Int. J. Syst.
27. Kempf, I. DNA amplification methods for diagnosis and Bacteriol. 44:94-98. 1994.
epidemiological investigations of avian Mycoplasmosis. Acta Vet. Hung. 46. Senterfit, L.B., Preparation of antigens and antisera. Vol. 1,
45:373-386. 1997. Mycoplasma Characterization. In: Methods in Mycoplasmology. S. Razin
28. Kempf, I., A. Blanchard, F. Gesbert, M Guittet, and G. Bennejean. and J.G. Tully, Editors. Academic Press: New York, N.Y. p. 401-404. 1983.
Comparison of antigenic and pathogenic properties of Mycoplasma iowae 47. Talkington, F.D. and S.H. Kleven. A classification of laboratory strains
strains and development of a PCR-based detection assay. Res. Vet. Sci. of avian Mycoplasma serotypes by direct immunofluorescence. Avian Dis.
56:179-185. 1994. 27:422-429. 1983.
29. Khan, MI., B.C. Kirkpatrick, and R. Yamamoto. A Mycoplasma 48. Yogev, D., D. Halachmi, G.E. Kenny, and S. Razin. Distinction of
gallisepticum strain-specific DNA probe. Avian Dis. 31:907-909. 1987. species and strains of Mycoplasmas (Mollicutes) by genomic DNA
30. Khan, MI., K.M Lam, and R. Yamamoto. Mycoplasma gallisepticum fingerprints with an rRNA gene probe. J. Clin. Microbiol. 26:1198-1201.
strain variations detected by sodium dodecyl sulfate-polyacrylamide gel 1988.
electrophoresis. Avian Dis. 31:315-20. 1987.

64
16
CHLAMYDIOSIS
Arthur A. Andersen and Daisy Vanrompay

SUMMARY. Avian chlamydiosis is caused by avian strains of Chlamydophila psittaci. Currently six serovars (serovars A through F) of C.
psittaci are known from birds. The serovars appear to be related to the host species, with serovar A predominately found in psittacine birds
and serovar B in pigeons. Some uncertainty still exists about the natural hosts of the other serovars; however, serovar D is the serovar seen in
turkeys during outbreaks with high mortality and high risks to humans. Natural infections of C. psittaci have been reported to occur in most
species of birds. Chlamydiosis is an Office International des Epizooties (OIE) list B disease, having public health importance and being
significant in international trade. Most states require the treatment of known infected birds prior to slaughter.
Agent Identification. Diagnosis of avian chlamydiosis preferably is made by isolation and identification of the agent. PCR and other
molecular tests are replacing isolation in a number of laboratories because of improved sensitivity of recently developed tests and to increase
laboratory safety. Other diagnostic methods such as enzyme-linked immunosorbent assay for detection of the antigen, and direct visualization
of the organism by Giemsa staining or by indirect fluorescent antibody tests, are satisfactory when signs of the disease are present. In
necropsied birds the use of immunohistochemistry is continuing to increase in importance for confirmation of chlamydiosis.
Serological Detection in the Host. A number of serological tests are available. The complement fixation (CF) test is considered the standard
in most countries. A high CF titer in a majority of individuals in a flock with clinical signs is presumptive evidence of an active infection. A
fourfold increase in titer in an individual bird is considered to be diagnostic for a current infection.

INTRODUCTION infections result in fever, anorexia, lethargy, diarrhea, and


occasionally shock and death.
Avian chlamydiosis (psittacosis, ornithosis, parrot fever) is caused Turkeys most commonly are infected by serovar D, with mortality
by Chlamydophila psittaci. The family Chlamydiaceae was rates of 5^W% unless early antibiotic treatment is instituted (6,45).
recently reclassified into two genera and nine species (15). The Vasculitis, pericarditis, splenitis, and lateral nasal adenitis are
new genera Chlamydia and Chlamydophila correlate with the typical postmortem findings. Experimentally, virulent turkey
former species Chlamydia trachomatis and Chlamydia psittaci. The strains cause little, if any, disease in chickens, pigeons, and
genus Chlamydia includes C. trachomatis (human), C. suis (swine), sparrows; however, cockatiels and parakeets succumb rapidly to
and C. muridarum (mouse, hamster). The genus Chlamydophila infection by these agents.
includes C. psittaci (avian), C. felis (cats), C. abortus (sheep, goats, Turkey outbreaks with low mortality and little or no human
cattle), C. caviae (guinea pigs), and the former species C. involvement usually are caused by the pigeon serovar. Mortality
pneumoniae (human) and C. pecorum (ruminants). All known avian usually is less than 5%. Necropsy findings include pneumonitis, air
strains belong to the species Chlamydophila psittaci, which includes sacculitis, and hepatosplenomegaly.
six avian strains (serovars A through F) (2,4), and two mammalian Chlamydiosis is a very common chronic infection of psittacine
isolates (WC and M56). WC and M56 are isolates from epizootics birds (42). The disease is of public health significance because of
in cattle and muskrats, respectively. The avian serovars are the popularity of psittacine birds as pets and the increased
relatively host-specific. Serovars A and B are usually associated placement of these birds in child-care facilities and in homes for the
with psittacine birds and pigeons, respectively. The natural hosts of aged. Infections cause conjunctivitis, enteritis, air sacculitis,
the other serovars are more uncertain. Serovar C has primarily been pneumonitis, and hepatosplenomegaly. Droppings are often green to
isolated from ducks and geese, and serovar D has been isolated yellow-green. Many of the birds become chronically infected but
from turkeys. Serovar F is a single isolate from a psittacine bird. show no clinical signs until stressed. These birds often shed
The host range of serovar E is the most diverse of the strains: it is chlamydiae intermittently and serve as a source of infection for
isolated from about 20% of pigeons, from many cases of fatal humans and other birds.
chlamydiosis in ratites, from outbreaks in ducks and turkeys, and Chlamydiosis is a common chronic infection of pigeons. Clinical
occasionally from humans. signs include conjunctivitis, blepharitis, and rhinitis (6). Survivors
The term "virulent” when used with serovar D is a misnomer, as become asymptomatic carriers. In recent years, chlamydiosis due to
virulence is highly dependent on the strain of chlamydia and the serovar E has been reported in a number of ratites.
species of bird infected. However, in the literature, ’’virulent" Chlamydiosis in ducks is rare in the United States, but is a serious
primarily refers to infections in turkeys, with serovar D being the economic and occupational health problem in Europe (6).
most virulent serovar in this species. Serovar D is the serovar most Trembling, conjunctivitis, rhinitis, and diarrhea are seen in infected
likely to be transmitted to humans following exposure in turkey ducks. Mortality can range up to 30%.
processing plants.
United States Department of Agriculture (USDA) regulations SAMPLE COLLECTION
prohibit the movement of poultry, carcasses, or offal from any
premises where avian chlamydiosis has been proven by isolation of The laboratory diagnosis of avian chlamydiosis usually includes
the chlamydial agent. State regulatory agencies may also impose the isolation and identification of the organism from the host.
quarantines on intrastate movement of diseased flocks and may Because C. psittaci requires living cells to multiply, isolation
require antibiotic treatment of infected birds. Interstate movement requires inoculation of either laboratory animals or cell cultures.
of birds, but not eggs, from infected flocks is prohibited by the Specimens should be collected aseptically, as contaminant bacteria
Animal and Plant Health Inspection Service of the USDA and by may interfere with isolation of the chlamydiae.
the U.S. Department of Health and Human Services. Proper handling of clinical samples is necessary to prevent loss of
infectivity. If specimens are used to inoculate animals or cell
CLINICAL DISEASE cultures immediately, most diluents will be adequate; however, if
the specimen is to be shipped or stored, a special diluent should be
Depending on the chlamydial serovar and the avian host, used. A diluent consisting of sucrose-phosphate-glutamate (SPG)
chlamydiae cause pericarditis, air sacculitis, pneumonia, lateral was developed for rickettsiae and has proven satisfactory for
nasal adenitis, peritonitis, hepatitis, and splenitis. Generalized transport of chlamydial field samples (10,43). The recommended
65
Arthur A. Andersen and Daisy Vanrompay

medium for chlamydiae consists of SPG buffer (sucrose, 74.6 birds. Mice and guinea pigs can also be used when chlamydiae-free
g/liter; KH2PO4, 0.512 g/liter; K2HPO4, 1.237 g/liter; and L- animal colonies are available.
glutamic acid, 0.721 g/liter), which can be sterilized by autoclaving.
To this is added fetal calf serum (10%), vancomycin and AGENT IDENTIFICATION
streptomycin (100 pg/ml), and nystatin and gentamicin (50 pg/ml).
The antibiotics reduce the effect of contamination even when Microscopic Examination of Specimens
samples are shipped at ambient temperatures. This medium also Histochemical Staining. Chlamydiae can be detected in smears of
serves as a laboratory diluent and a medium for freezing of exudates and feces and in impression smears of liver and spleen by
chlamydiae. a variety of techniques. Gimesa and Gimenez stains are commonly
For isolation and identification of C. psittaci from acute and used. The modified Gimenez technique (Table 16.1; B.D. Van Do-
subclinical cases of chlamydiosis, the following samples should be Kamp, pers. comm.) is routinely used by several laboratories for
collected: pharyngeal/choanal slit swabs (3), lung tissue, thickened detection of chlamydial inclusions in smears and paraffin-embedded
exudate-coated air sacs and free exudate, thickened pericardial tissue sections.
tissues and exudate, sections of enlarged spleen and liver, intestinal Procedure for smears:
mucosa at sites of hyperemia, colon contents, and conjunctival and 1) Fix in methanol for 5 min.
nasal discharges. In the live bird, pharyngeal/choanal slit swabs, 2) Stain in solution 3 for 10 min.
fecal samples, and cloacal swabs are the preferred specimens (3). 3) Rinse in tap water.
All three specimens should be collected, as no one sample will 4) Counterstain in solution 6 for 2 min.
provide an isolation in all cases. Other specimens that can be 5) Rinse in tap water.
collected, depending on the disease signs present, include whole 6) Air dry.
blood, conjunctival scrapings or exudate, and peritoneal exudate. Procedure for paraffin sections:
When companion birds are being tested for subclinical infection 1) Deparaffmize and hydrate in distilled H2O.
before placement in facilities, repeated sampling is necessary. 2) Stain in solution 3 for 10 min.
The same tissues can be collected for PCR and other molecular 3) Rinse in tap water.
tests. If the DNA extraction will not be done immediately, it may be 4) Dip in solution 4 until no more red runs out of the section.
beneficial to collect the specimen in a DNA stabilization buffer 5) Rinse in tap water.
(13). 6) Counterstain in solution 6 for 20 dips.
7) Dip in two changes of 95% alcohol, for five dips each.
Hazards to Humans in Handling Specimens 8) Dehydrate, clear, and mount.
Potentially infected birds and specimens must be handled with 9) Chlamydiae will appear red against a green background.
caution, as the avian strains can cause serious or fatal disease in
humans. Humans can also become infected through aerosol Immunohistochemical Staining.
exposure or contact with dried excrement. Precautions must be Immunohistochemistry is another method for detection of
taken to prevent creation of infectious aerosols during necropsies or chlamydiae in cytologic and histologic preparations (35,45). The
by mechanical manipulation of suspensions of organisms in use of immunohistochemical staining on formalin-fixed sections is
syringes, centrifuges, tissue grinders, pipettes, or other laboratory gaining in popularity because a number of laboratories are using
equipment, and to prevent the contamination of skin with infectious automatic staining equipment. The technique also allows
exudate or material. The use of biological safety hoods and gloves laboratories to take routine samples for histopathology and then
is recommended during the handling of any potentially infectious retrospectively examine them for chlamydiae if necessary. The
material. technique is more sensitive than histochemical staining, but
experience is required to interpret findings. Cross-reactions with
Storage of Specimens some bacteria, fungi, and epithelial intercellular material dictate that
The stability of chlamydiae during storage depends upon the morphology must be considered in making a positive diagnosis.
material in which it is contained. Chlamydiae in tissue specimens or Chlamydial antigen is most often found in macrophages in areas of
yolk-sac suspension can be preserved indefinitely by storage at -70 inflammation. Antigen can be found in the lateral nasal gland up to
C (5,6). Note that chlamydiae harvested from cell culture require 50 days post infection. Inflamed air sacs and pericardium also
special media during freezing. A satisfactory method for freezing consistently contain antigen. However, care must be taken to avoid
them is to replace the cell culture medium with SPG buffer before misinterpreting hemosiderin as antigen. Antigen-positive tissue and
freezing, as the organism is highly susceptible to the presence of hematoxylin-and-eosin-stained sections are used as controls.
sodium ions in the medium (43). The transport medium described For immunohistochemical staining of paraffin sections, a genus­
earlier will provide adequate stability during freezing. At 4 C, the specific monoclonal or polyclonal antibody is used as the primary
organism can survive with gradual loss of infectivity for 30 days or antibody. The antibodies should be produced to formalin-
longer when in heavily infected tissues or in SPG transport medium inactivated chlamydiae and selected for reaction with formalin-
(43). inactivated chlamydiae. The specific procedure used for staining the
paraffin sections will depend on the equipment available, as most
PREFERRED CULTURE MEDIA AND SUBSTRATES techniques will give satisfactory results.

Chlamydiae are obligate intracellular parasites that multiply in the Isolation of the Organism
cytoplasm of animal cells. The preferred method of isolation of Preparation of Inoculum. Tissue specimens, fecal samples, and
avian strains in most laboratories is inoculation of cell cultures. swabs are routinely used as samples for the isolation of chlamydiae.
Modifications of standard cell culture techniques are used to The processing of the samples is similar for inoculation of cell
enhance the growth of the organism and are discussed under agent cultures, embryonated eggs, or laboratory animals. Diluents such as
identification below. beef heart infusion broth, phosphate-buffered saline (PBS; pH 7.2),
Until recently, the standard method for the isolation of chlamydiae and cell culture media often are used to prepare a 20%-40%
has been inoculation of chicken embryos. Chicken embryos will suspension of the homogenized sample. These diluents, with
support the growth of all known strains of chlamydiae infecting antibiotics, are satisfactory when the samples are to be inoculated
within 24 h and will not be frozen. For routine use, a number of
66
Chapter 16 Chlamydiosis

laboratories are using SPG buffers, such as the transport medium is most commonly used; it can be added to the medium at the rate of
described earlier or Bovamick's buffer (10,43). These buffers have 0.5-2.0 pg/ml at the time of inoculation of the monolayer. The
the added advantage of stabilizing the agent during refrigeration or concentration needed will depend on the cell line used. Emetine
freezing of the samples. must be removed after treatment and replaced with medium (836);
Before the inoculation of animals or cell cultures, contaminated this is considered by some to be an argument for its use. The
samples must be treated by one of three basic methods: treatment monolayer is treated for 5 min with a concentration of 0.5 μg/ml of
with antibiotics (8), treatment with antibiotics plus low-speed emetine. After removal of the emetine and its replacement with
centrifugation (6), or treatment with antibiotics plus filtration (8). A medium, the monolayer is ready for use. The effect of these drugs
number of antibiotics that do not inhibit chlamydiae are available. A on the replication of the chlamydiae is strain dependent: growth of
standard procedure is to homogenize the sample in diluent most, but not all, strains is enhanced by treatment of the monolayer
containing 500-1000 pg/ml each of streptomycin, vancomycin, and with one of these drugs.
kanamycin. Amphotericin B will control yeast and fungal growth. Chlamydiae are known to have a low infection rate in cell cultures,
Other antibiotic solutions are often used, but penicillin, tetracycline, and methods to enhance the infection rate are often used. A
and chloroamphenicol should be avoided, as they will inhibit the common method to increase attachment of the chlamydiae to the
growth of chlamydiae. cells is to centrifuge the inoculum onto the monolayer at 1000-3000
If the sample is lightly contaminated, adequate treatment for x g for 30-90 min. Adsorption is enhanced by temperatures near 37
inoculation into chicken embryos, guinea pigs, mice, or cell culture C. The inoculum is often removed and replaced with cell culture
consists of homogenization of the sample in an antibiotic solution. medium containing an antimetabolite. Incubation is usually at 37-
The sample is often left in the antibiotic solution for 24 hr before 39 C. Cultures must be examined at regular intervals by an
the inoculation of cell cultures or animals. If the sample is heavily appropriate staining method for the identification of chlamydiae.
contaminated, such as a fecal sample, it should be homogenized in Staining is usually done at days 2-3 and days 5-6. Cultures that are
antibiotics and centrifuged at 1000-2000 x g for 30 min. The negative at day 6 are harvested and repassed. Disruption of the
surface layer and the bottom layer should be discarded; the monolayer by freeze-thawing should not be done, as it will destroy
supernatant fluid is collected and recentrifuged, and the final the chlamydiae.
supernatant fluid is inoculated into the experimental host or cell Staining of coverslips. The preferred method for fixing of the
culture. If contamination persists, the sample may also be passed monolayer is to remove the medium, wash once with PBS, and fix
through a filter of 0.45-0.8 pm pore size. with acetone for 2-10 min. If the cell culture vessel is plastic, the
Cell Cultures. Cell cultures are the most common and convenient monolayer can be fixed with a mixture of 50% acetone and 50%
method for the isolation of C. psittaci. The most commonly used methyl alcohol. The preferred method for staining is the direct FA
cell lines are BGM, McCoy, HeLa, Vero, and L-929, although a method (5,6,8), but a number of other methods can be used. With
number of other cell cultures can be used. A recent study showed the FA method, the fluorescein-conjugated antichlamydia serum is
BGM to be the most sensitive, with Vero and L-929 listed as applied to the coverslip and incubated 30 min at 37 C. The
satisfactory (49). Standard cell culture medium is used, containing coverslips are then washed three times with PBS, air-dried, and
5%-10% fetal calf serum and antibiotics that do not inhibit the mounted in an inverted position on a glass slide. The chlamydial
growth of chlamydiae. In our laboratory, 10 pg/ml of gentamicin inclusions will fluoresce a light green color under ultraviolet
and 2 pg/ml of amphotericin B are standard for use. microscopy. Commercially prepared conjugates using monoclonal
The laboratory equipment and supplies available will determine antibodies are available and are highly specific. Conjugates also can
the type of vessel used to grow the cell culture monolayer for be prepared from polyclonal sera from rabbits, guinea pigs, sheep,
incubation with the homogenated samples. The equipment, or goats. The main problem is obtaining a specific, high-titer
however, must be suitable for the following procedures: antiserum. The fluorescein conjugate is prepared using standard
identification by direct fluorescent antibody (FA) or another techniques (5,6,8).
appropriate technique of staining of the infected monolayer; Other techniques to demonstrate chlamydial inclusions are the
centrifugation of the inoculum into the monolayer to enhance indirect immunofluorescent technique, the immunoperoxidase
infectivity; possible blind passage at 3-6 days to increase sensitivity technique (35), and direct staining techniques, such as Pierce-Van
of isolation; examination of the sample two to three times during a Der Kamp (PVK), Gimenez, Giemsa, or Macchiavello's stains. The
passage; and protection of humans against possible infection. Small immunoperoxidase, Macchiavello, PVK, and Gimenez staining
flat-bottomed vials (I-dram shell vials) or bottles, with 12-mm techniques allow the use of standard light microscopes.
diameter glass coverslips, will meet these requirements and are Chicken Embryos. A number of laboratories still use chicken
often used (8,9) because the cell culture monolayer can be grown embryos for primary isolation of chlamydiae. The standard
directly on the coverslip. Several vials (often four to six) are procedure is to inject up to 0.5 ml of inoculum into the yolk sacs of
inoculated with each sample to permit fixing and staining at various 6 to 7 day-old embryos (6). Incubation is at 39 C, as multiplication
times and to permit repassaging of negative samples after 4-6 days of chlamydiae is greatly increased in comparison with incubation at
of inoculation. Multiwell disposable cell culture dishes in sets of 37 C. Replication of the chlamydiae usually will cause the death of
four are used in our laboratory. These work well when processing the embryo within 5-12 days after inoculation. If no deaths occur,
large numbers of samples; however, cross contamination between two additional blind passages are usually made before calling the
wells can be a problem. The infected monolayers are fixed and sample negative. Chlamydial infection typically causes vascular
stained directly in 96 multiwell dishes. Inclusions are visualized by congestion of the yolk-sac membranes, which are harvested and
inverting the plate under a FA microscope. homogenized as a 20% yolk-sac suspension. This suspension can be
Chlamydiae can be isolated from cells that are replicating frozen to preserve the strain or inoculated into other eggs or into
normally. Most diagnosticians, however, prefer to use cell culture monolayers.
nonreplicating cells for two reasons: to provide increased nutrients Identification of the organism requires preparation of an antigen
for the replication of chlamydiae, and because nonreplicating cells from the infected yolk sac and testing for the chlamydial group
can be maintained for longer periods for observation. Host-cell antigen by the complement fixation (CF) test or other appropriate
replication can be suppressed either by irradiation or by cytotoxic serologic test. Laboratories now often inoculate cell culture
chemicals; the use of chemicals is becoming more common. monolayers with the yolk-sac suspension and stain for chlamydial
Cytotoxic chemicals include 5—iodo—2-deoxyiodine, cytocholasin inclusions at 48-72 hr by the direct FA test described above. The
B, cycloheximide, and emetine hydrochloride (36). Cycloheximide typical chlamydial inclusion is a round or hat-shaped body in the
67
Arthur A. Andersen and Daisy Vanromp ay

cytoplasm. In some virulent strains the inclusions break up rapidly 6. Phosphate-buffered saline (PBS): 10 mM Na2HPO4, 10 mM
and the chlamydial antigen is found dispersed throughout the NaH2PO4, 145 mM NaCl, pH 7.0. Adjust pH by adding NaH2PO4.
cytoplasm. 7. Tris-EDTA (TE) buffer: 10 mM Tris-HCl, pH 8.0, 1 mM EDTA,
Enzyme-Linked Immunosorbent Assays. A number of pH 8.0 (ethylene diamine tetra-acetic acid).
manufacturers are producing enzyme-linked immunosorbent assay 8. Sodium dodecyl sulfate (SDS) solution: 10 mg/ml in TE.
(ELISA) kits for detection of Chlamydia trachomatis in humans. 9. Phenol: saturated solution in TE buffer. If two separate phases
These test kits will detect C. psittaci as they react to the are visible, use the lower phase only.
lipopolysaccharide (LPS) or group antigen. The advantages of 10. Chloroform-isoamyl alcohol: 24:1 (v/v).
ELISA are that it is rapid, does not require a high level of expertise, 11. Isopropanol, analytical or molecular biology grade
and is safe for the technician because the sample can be inactivated.
A number of the test kits have been evaluated for use in avian Procedure for DNA extraction from nasal, pharyngeal, conjunctival
samples (50), although none of the kits are licensed for use in swabs or faecal swabs:
veterinary medicine. One of the problems of some of these tests is 1. Pipet 500 μΐ of lysis buffer I into a 2-ml microfuge tube
that the chlamydiae LPSs share some epitopes with the LPSs in containing the swab.
other gram-negative bacteria, which can lead to a high number of 2. Vortex mix thoroughly for 1 min.
false positives. This problem has been reduced or eliminated in the 3. Centrifuge at 12,000 x g for 30 s.
more recently developed kits by the use of monoclonal antibodies. 4. Put the swab into a 1-ml pipet tip whose lower half was cut off
However, these kits still lack sensitivity: a few hundred organisms and place it all into a fresh microfuge tube.
are needed for a positive reaction. The general rule with these tests 5. Centrifuge at 12,000 x g for 1 min to press the remaining liquid
is that a diagnosis of chlamydiosis can be made when a strong out of the swab.
positive reaction is seen along with clinical signs of chlamydiosis. 6. Add the liquid to that in the first microfuge tube. If you sampled
Because of the number of false positives, a positive test without your swabs into GIT buffer, please
signs cannot be considered significant. 7. Centrifuge the liquid at 12,000 x g for 15 min.
8. Discard the supernatant and resuspend the pellet in 50 μΐ lysis
Polymerase chain reaction buffer I.
The possibilities of diagnostic detection of Chlamydophila psittaci 9. Add 20 μΐ proteinase K solution I and incubate at 60 C for 2 h.
are considerably improved with the introduction of molecular 10. Inactivate the proteinase K by heating at 95 C for 15 min.
methods, particularly the polymerase chain reaction (PCR), which 11. Centrifuge at 12,000 x g for 5 min to remove debris.
permits direct identification from clinical specimens and accelerates 12. Use 5 μΐ of the supernatant for PCR.
diagnosis from several days to a single working day. Tests 13. For faecal swabs an additional purification of DNA by phenol
published in the literature utilize two different genomic target extraction and ethanol or isopropanol precipitation is recommended.
regions for amplification, namely the ribosomal RNA gene region 14. When the swabs are taken in guanidinium isothiocyanate
(33,16,32) and the gene encoding the MOMP antigen designated buffer, step 1 should be vortex mix thoroughly for 1 min followed
ompl or ompA (25,27,56). by centrifugation at 12,000 x g for 30 s and afterwards removing all
The sensitivity of the PCR largely depends on the amount and the buffer except 50 μΐ. Resuspend the pellet in the buffer and add
quality of the extracted DNA. Optimal extraction of nucleic acids the proteinase K solution as in step 9.
from a wide range of clinical samples is one of the most under
appreciated, but nevertheless challenging and important steps. Procedure for DNA extraction from organ tissues:
Proper extraction must efficiently release nucleic acids from 1. Boil 100 mg of homogenized tissues in 200 μΐ of water in a
samples, remove PCR inhibitors, avert the degradation of nucleic plastic tube for 10 min. Subsequently, allow the tube to cool to
acids, and ensure adequate concentration of the nucleic acids after room temperature.
extraction. This is relatively easily achievable for viral and 2. Optionally, proteinase digestion can be carried out to increase
mammalian amplification targets, but much more difficult and the final yield of DNA: add 200 μΐ SDS solution and 20 μΐ of
inconsistent for nucleic acid targets of bacteria, as these pathogens proteinase K solution I to the tube and incubate at 55 C for 1 h.
frequently possess highly resistant outer membranes, and specific 3. Add 200 μΐ of phenol.
methods for disruption of these membranes are necessary for 4. Vortex mix vigorously for 1 min.
quantitative recovery of nucleic acids from these organisms. In 5. Centrifuge at 12,000 x g for 5 min.
some cases, concentration of target organisms by physical means 6. Transfer the upper aqueous phase into a fresh microfuge tube.
increases assays’ sensitivity, but it is important to ascertain the 7. Add 200 μΐ of chloroform-isoamyl alcohol and vortex mix at the
enrichment effect (13). Such methods include immunomagnetic, highest intensity for 1 min.
centrifugal, or filter concentration (31,1,37). Several in-house-made 8. Transfer the upper aqueous phase into a fresh microfuge tube.
and commercial DNA extraction methods have been described 9. Precipitate DNA by adding 120 μΐ of isopropanol. Thoroughly
(54,38; Harkinezhad et al., 2005, unpublished results). The mix the reagents and incubate at room temperature for 10 min.
following are used in a number of laboratories and are designed for 10. Collect DNA by centrifugation at 12,000 x g for 10 min.
specific sample types. Discard supernatant.
11. Allow DNA pellet to air-dry for 30 min.
Materials for the simplest methods for chlamydial DNA extraction 12. Dissolve pellet in 20 μΐ of water. Use 1 μΐ for the PCR.
from various specimens:
1. Water. Deionized water must be used for all buffers and Procedure for DNA extraction from faeces:
dilutions. 1. Add 200 μΐ of water to 100 mg of faeces and vortex mix
2. Lysis buffer I: 100 mM Tris-HCl, pH 8.5, 0.05% (v/v) Tween® vigorously for 1 min.
20. 2. Boil the suspension for 10 min.
3. Lysis buffer Π: 0.01 M Tris-HCl, 0.05 M KC1, 0.0025 M 3. Add 300 μΐ pf phenol to each tube for DNA extraction and
MgC12.6H2O, 0.5% (v/v) Tween® 20, pH 8.3 vigorously vortex mix for 1 min.
4. Proteinase K solution I: 10 mg/ml in water. 4. Centrifuge at 14,000 x g for 5 min.
5. Proteinase K solution Π: 20 mg/ml in water 5. Transfer the (upper) aqueous phase into fresh microfuge tubes.
6. Add 300 μΐ of chloroform-isoamyl alcohol to each tube.
68
Chapter 16 Chlamydiosis

7. Vortex mix at the highest intensity. although detection by use of a fluorimeter is also possible by using
8. Centrifuge at 14,000 x g for 5 min. this method. The sensitivity of the nested PCR-EIA was established
9. Transfer the (upper) aqueous phase into fresh microfuge tubes. at 0.1 infection-forming units (IFU). Specificity was 100%. An
10. Add 200 μΐ of isopropanol for DNA precipitation. Thoroughly internal inhibition control was included to rule out the presence of
mix the reagents and incubate at room temperature for 10 min. inhibitors of DNA amplification.
11. Centrifuge at 14,000 x g for 10 min. Discard the supernatant.
12. Allow DNA pellet to air-dry for 30 min. Nested PCR-EIA procedure
13. Dissolve pellet in 20 μΐ of water. Use 1 μΐ for the PCR. The nested ompA PCR-EIA was developed using external and
internal primers generating a biotin-fluorescein dual labelled
Alternatively, commercial DNA extraction kits can be used for internal PCR product of 472 bp (Table 16.2).
the extraction of chlamydial DNA. In our hands the QIAamp® Both inner and outer sense primers are located within the first
DNA Mini Kit performed the best for pharyngeal swabs while the conserved domain (CD1) of ompA, whereas the inner anti-sense
High Pure PCR Template Preparation Kit performed best for primer is located in CD3 and the outer anti-sense primers overlap
cloacal swabs and faeces. The use of commercial kits can be CD4 and variable domain 4 (Fig 16.2) First round PCR occurred in
recommended especially when dealing with PCR inhibitors. They 50 mM KC1, 20 mM Tris-HCl (pH 8.3), 2 mM MgCl2, 0.1%
contain a special buffer reagent for lysis of the bacterial and Tween20, 200 μΜ each dNTP, 1.25 μΜ each external primer
eukaryotic host cells mostly based on a guanidine-detergent lysing (Table 16.2) and 0.1 U SuperTaq polymerase (5 U/μΙ). After an
solution first described by Chomczynski and Sacchi (12). initial denaturation at 95 C for 5 min, 20 cycles of one min at 95 C,
Simultaneous disruption of cells and inactivation of nucleases is two min at 59 C and three min at 72 C, with a final extension at 72
achieved by lysis in strong denaturing reagents, for instance 4M C for 5 min were performed. Second round amplification was
guanidinium isothiocyanate (GIT) or 4M guanidinium performed under similar conditions, using labelled internal primers
hydrochloride. Guanidinium isothiocyanate (GIT) is 2.5 times more (each 10 μΜ; Table 16.2), adapted annealing temperature (47 C)
effective on a molar basis than guanidine HC1. Both anion and and adapted number of cycles (25). Subsequently, nested PCR
cation act as denaturants in the GIT buffer but only the guanidine generated a fluorescein/biotin dual-labelled product of 472 bp.
cation in GIT-HCL buffer. An optimal RNase digestion is intended To allow colorimetric detection of the ompA PCR products, 50 μΐ
to remove cellular RNA. The lysate is then centrifuged through a of the PCR product diluted 1:10 in PBS supplemented with 3%
mini-column, where the released DNA is effectively bound to a BSA was transferred in duplicate to streptavidin-coated microtiter
solid phase: modified silica, hydroxyl apatite, or a special filter plates (2 pg/well for 3 hr at 37 C) and incubated at 37 C for one
membrane. After washing, the DNA can be eluted with an elution hour. Non-specific binding places .were blocked overnight (4 C)
buffer or water. DNA prepared in this manner is usually of high with 5% BSA in PBS. Subsequently, the plates were washed twice
purity and free of PCR inhibitors. with PBS and incubated (1 hour, 37 C) with a horseradish
Tissue sample homogenization in cell culture medium, PBS, or peroxidase labelled anti-fluorescein antibody (Invitrogen), diluted
TBS and/or freeze-thaw cycles of samples lead to rapid loss of 1/1000 in PBS supplemented with 3% BSA. Following incubating
chlamydial target DNA. Specimens that contain low target numbers and washing with PBS, the ABTS substrate solution (2,2' azino-di-
before freezing often become negative after cryostorage. The 3-ethylbenzothiazoline sulphonate, KPL) was added to the wells.
preservation of DNA from degradation is critically important. De Absorbencies were read at 450 nm after incubating for 30 min at 37
Graves et al., (13) advise to collect all specimens for PCR analysis C (TiterTek MultiskanR Plus, MKH, TechGen International). Three
in a DNA stabilization reagent. Such reagents are commercially positive controls consisting of serial 10-fold dilutions of PCR
available; for instance the RNA/DNA Stabilization Reagent for products generated from 5 IFU of C. psittaci and five negative
Blood/Bone Marrow® from Roche Applied Science. This controls (water) were included in each assay. Results were positive
stabilization reagent is based on the denaturation of proteins in a if the absorbance exceeded the cut off value of the mean of negative
concentrated solution of guanidinium isothiocyanate and a reducing controls plus three times the standard deviation.
agent, as introduced by Chirgwin et al., (11) for the inactivation of Note: rules to remember
ribonuclease during RNA isolations. However, it also works 1. Work in separate rooms (three levels) for DNA extraction (level
perfectly well to stabilize DNA as demonstrated by De Graves et 1), preparation of the PCR
al., (13) extracting chlamydial DNA from 10% (w/v) tissue reaction mix and PCR (level 2) and visualization of the result (level
suspensions of lungs from mice and by Harkinezhad et al., (2005, 3). Use a biosafety cabinet for DNA extraction and preparation of
unpublished results) extracting chlamydial DNA from avian the PCR reaction mix.
pharyngeal and cloacal swabs stored in this reagent as well as 2. Use separate sets of supplies and pipets in these levels.
faeces. 3. Avoid bringing amplified DNA to level 1 and 2.
The choice of a PCR test depends on the skill of the personnel in 4. Take supplies from storage cabinets and not from the room
the laboratory and the equipment available. Nested procedures are where the PCR analyses are being performed.
more sensitive than traditional PCR and in most cases are also more 5. Store all reagents (including water) as aliquots and record the
sensitive than real-time PCR. Recently, two highly sensitive nested lots.
PCR assays have been developed to detect Chlamydophila psittaci 6. The use of positive displacement pipets or filter tips in
in avian samples (38,48). The nested PCR of Sachse and Hotzel recommended.
(38) is based on a first amplification generating a genus-specific 7. Change gloves frequently.
ompA product (576-597 bp) followed by a second amplification 8. Uncap the tubes carefully to prevent aerosols.
using one species-specific and one genus-specific primer generating 9. Minimize sample handling.
a Chlamydophila psittaci-specific amplicon (389-404 bp). PCR 10. Add non-sample components to the reaction mixture before
results are visualized by agarose gel electrophoresis using ethidium addition of DNA and cap each tube after adding the sample DNA
bromide containing gels. The sensitivity of the nested PCR-EIA was before proceeding to the next sample.
established at 1 to 0.1 infection forming unit (IFU). Van Loock et 11. Use a positive control that amplifies weakly but consistently, as
al. (48) developed a nested PCR-enzyme immuno assay (PCR-EIA). one of the major mistakes is using a strongly positive control and
The fluorescein-biotin labelled PCR products were immobilized on thus augmenting the risk of post-PCR carry-over from the amplified
streptavidin-coated microtiter plates and detected with anti­ positive control.
fluorescein peroxidase conjugate and a colorimetric substrate,
69
Arthur A. Andersen and Daisy Vanromp ay

12. Use pre- and post-amplification methods for the inactivation of used next to the TaqMan probes. Genotype E/B could only be
possible contaminating nucleic acids. detected by use of a minor groove binder (MGB) probe. Genotype­
Real-time PCRA number of real-time PCR tests have been specific probes (Applied Biosystems) listed in Table 16.3 were 5’
developed. At least two of these tests have been used successfully and 3’ labelled with the reporter dye 6-carboxyfluorescein (FAM)
with avian samples. One uses the TaqMan system and the other uses and the quencher dye carboxytetramethylrhodamine (TAMRA),
the LightCycler system. The technologies are similar and either test respectively. Genotype-specific reactions were performed with the
with minor modifications can readily be adapted to other systems. LightCycler 2.0 instrument in LightCycler Capillaries, using the
The real-time PCR systems have the advantage that their sensitivity LightCycler FastStart DNA MasterPLUS Hybridization Probes kit,
approaches that of the nested PCR systems, and they require no in a total reaction volume of 20 μΐ (9 μΐ PCR grade water, 2 μΐ of
additional pipetting or handling of the PCR product following the primer (300 nM) / probe (300 nM) I competitor (50 nM or 150 nM)
initial set-up of the PCR mix. This reduces both the time needed for mixture, 4 μΐ 5 x Master Mix, 5 μΐ of DNA template). Cycling
the test and the incidence of contamination. conditions were as follows: 95 C for 10 min and subsequently 50
The TaqMan test amplifies a 132bp segment of the 23S rDNA cycles of 95 C for 10 s followed by 63 C (genotype B) or 60 C (all
(15). In our laboratory we have found its sensitivity to be other genotypes), for 20 s. Genotypes A, B and E could only be
comparable to isolation using samples from experimentally infected distinguished by using competitor oligonucleotides enhancing the
animals (Andersen, unpublished data). TaqMan probe specificity. The detection of genotype E/B was made
Recently a SYBRGreen-based real-time PCR was developed possible by use of a minor groove binder (MGB) probe (Applied
targeting the rDNA ribosomal spacer of Chlamydophila psittaci Biosystems) (29). Standard Ct-value graphs obtained from testing
(19). The test could detect 10 rDNA copies/μΐ DNA extract and serial dilutions of purified control plasmids (108 to 101) were used
could detect all known Chlamydophila psittaci genotypes (20). for quantification. Clinical samples from case studies were tested in
Real-time PCR was performed with the LightCycler 2.0 Instrument the presence of genotype-specific control plasmids (50 ompA
(Roche, Applied Science, Penzberg, Germany) using the copies/μΐ) to check for PCR inhibitors. Ct-values for clinical
LightCycler FastStart DNA MasterPLUS SYBR Green I kit and samples were plotted against standard graphs and C. psittaci
LightCycler Capillaries. The reaction mixture (20 μΐ) was prepared genotypes present in clinical samples (No) were quantified using the
according to the manufacturer’s protocol: 11 μΐ PCR grade water, LightCycler 4.0 software. The sensitivity of the assay was found to
2 μΐ of primer mixture (300 nM forward primer and reverse primer), be 10 ompA copies/μΐ DNA extract.
2 μΐ lOx Master Mix, 5 μΐ of DNA template. Cycling conditions
were as follows: 50 cycles of 95 C for 10 s, 63 C for 10 s and 72 C DNA microarray-based detection andddentification
for 8 s. All default program settings were used. Standard graphs of Microarrays can be coupled with PCR where they serve as a set of
the Cycle threshold (Ct) values, obtained by testing tenfold serial parallel dot-blots to enhance product detection and identification of
dilutions (108 to 101) of the purified species-specific inhibition bacterial isolates. Recently, Sachse et al. (39) developed a DNA
control plasmid, were used for quantification. Ct-values were microarray-based detection and identification method for
automatically converted into initial template quantities (No) using Chlamydia and Chlamydophila spp. The test was developed using
the LightCycler Software 4.0. DNA from clinical samples was the ArrayTube platform (CLONDIAG®chip technologies). Unique
always tested in the presence of control plasmid (50 copies/μΐ) to species-specific hybridization patterns were obtained for all nine
check for PCR inhibitors. species of the family Chlamydiaceae on both microarray types. The
present assay proved suitable for unambiguous species
AGENT IDENTIFICATION identification of chlamydial cell cultures and showed a potential for
direct detection of these bacteria from clinical tissue.
Previously the standard procedure for the identification of
chlamydiae was to confirm the presence of chlamydiae by preparing SEROLOGICAL DETECTION IN THE HOST
an antigen from the egg harvest and testing it in the CF test with
known antisera. Currently the determination is made at the time of Serology
isolation by using fluorescein-labeled antichlamydial antibodies Despite the introduction of very sensitive and specific tests like
following inoculation of cell culture monolayers as described PCR, the idea of serological diagnosis still lingers in the minds of
above. Distinguishing between C. psittaci and the other species of several veterinary clinicians. However, serology alone is not
chlamydia is not of major importance, as all avian isolates have particularly useful in diagnosing a current chlamydial infection in
been in the species C. psittaci. Knowing the particular strain can be birds because of the high prevalence of this infection in birds and
important in identifying the source of infection and in the long-term, up to several months, persistence of anti-chlamydial
epidemiologic studies. Monoclonal antibodies have been developed antibodies. In most bird species, there is a high background rate of
that can readily serotype the six known serovars seen in birds and a anti-chlamydial antibodies, and until we have more information on
number of serotypes from other animals (2,4). PCR restriction the disease pattern in certain bird species in relation to direct
fragment length polymorphism procedures are available that can identification of the bacteria and serology, we are unable to
distinguish the different chlamydial species and avian strains comment on the real significance of antibody titers obtained. Thus,
(14,41). Isolates can also be readily identified by sequence analysis to determine if a single bird is infected, serology should always be
of the major outer membrane protein gene (ompA) (20). Sequence used in conjunction with antigen or gene detection or paired sera
analysis has the advantages that it can characterize new strains and should be examined. However, obligatory examination of paired
that in epidemiological studies it can track strains with minor DNA sera removes serology from immediate clinical relevance. A
changes. positive test is evidence that the bird was infected by the bacterium
but does not necessarily indicate an active infection. False negative
Genotype-specific real-time PCR results can occur in birds with acute infections that are sampled
Recently a genotype-specific real-time PCR was developed before sero-conversion. Treatment with antibiotics may also delay
detecting all known ompA genotypes (19). The genotype-specific and/or diminish the antibody response. The main serological
real-time PCR could easily distinguish genotypes C, D and F by use methods that are been used for detecting chlamydial antibodies are:
of TaqMan probes. Genotypes A, B and E could not be 1) various methods of elementary body agglutination (EBA), 2) the
distinguished from each other by simply using TaqMan probes. For complement fixation test (CF), 3) an indirect (micro)
this purpose non-fluorescent competitor oligonucleotides had to be immunofluorescence (ΜΠ7) test and 4) several commercial ELISAs.
70
Chapter 16 Chlamydiosis

The EBA detect primarily IgM antibodies and thus can detect early the serodiagnosis of Chlamydophila psittaci infections. At present,
infections (22,23,24). Titers > 1:10 in budgerigars, cockatiels and an ELISA using recombinant MOMP of Chlamydophila psittaci has
lovebirds; and titers > 1:20 in larger birds are frequently seen in already been described (52; Verminnen et al., 2005, unpublished
cases of recent infection. A negative result does not guarantee that a results) for testing avian sera. The test is not species-specific as the
bird is free of infection as the sensitivity of the test is rather low. recombinant MOMP, produced by transiently tranfected COS7 cells
The direct complement fixation (DCF) test detects avian IgG but (51), comprises all four variable domains as well as conserved
not IgM. An advantage is that there is a readily available domains and thus detects antibodies against all members of the
microprocedure. Disadvantages are: 1) test antigen commercially genus Chlamydophila and Chlamydia (53). However, this is not a
unavailable, 2) the test cannot be used for testing sera from avian problem when testing avian sera as birds can only become infected
species whose immunoglobulins do not fix complement, such as by Chlamydophila psittaci. Unfortunately, Chlamydophila psittaci-
small psittacine birds, 3) it is only relatively sensitive, 4) it cannot specific ELISAs are not yet available.
be used to differentiate between IgG and IgM antibodies, making it
necessary to test paired sera, and 5) the technique is fairly laborious DIFFERENTIATION FROM CLOSELY RELATED AGENTS
when there is a large number of samples to be tested. The modified
DCF test is more sensitive but has the same disadvantages as the The gross lesions caused by chlamydiae may resemble those
DCF test (5,21,23,47). The indirect IF test detects all caused by Pasteurella, mycoplasmas, and coliforms when a
immunoglobulin isotypes and is, as is the MIF test, widely used to systemic infection occurs. Pasteurella- and coliform-caused
detect Chlamydia trachomatis, Chlamydophila pneumoniae and diseases can readily be eliminated from consideration, as the
Chlamydophila psittaci antibodies in human sera using selected fibrinous exudate from uncomplicated chlamydial infection will be
strains of these bacteria (40,55,17). The MIF test appears to be more free of bacteria that can be cultured on standard bacteriologic
sensitive than the complement fixation tests. Some years ago a large media. Confirmation of chlamydial infections can be obtained by
number of commercially available ELISAs were evaluated for isolation and identification of viable chlamydiae from infected
demonstrating Chlamydophila psittaci antibodies in birds. All of tissues or by demonstration of chlamydiae by specific
these ELISAs were highly sensitive but showed low specificity, as immunohistochemical techniques.
they were mainly based on the use of whole chlamydial organisms,
LPS, or chlamydial outer membrane fractions of LPS and
lipoglycoprotein nature (44,7,18,30,47). When using these sources
and antigen, false-positives due to the presence of antibodies cross-
reactive to the chlamydial LPS or hsp60 cannot be ruled out. More
recently, peptide-based ELISA systems, or ELISAs using
recombinant LPS, have become commercially available for the
specific detection of Chlamydia trachomatis, Chlamydophila
pneumoniae and Chlamydophila abortus antibodies (Medac,
Savyon, Labsystems), (28,34,26,46. These tests performed as well
as the MIF assay, but are less time-consuming, less expensive, and
easier to perform. In the future this principle might also be useful in

Table 16.1. Modified Giminez technique or Pierce-Van Der Kamp (PVK) stain.

Constituent Amount

Distilled H2O 450.0 ml


Solution 1:
Phenol 5.0 ml

Basic fuchsin 2-5 g


Add to:
95% Ethanol 50.0 ml

Incubate (tightly capped) at 37 C for 48 hr. Filter and store at room temperature.
Na2HPO4 11.65 g
Solution 2: NaH.POAO 2.47 g
Distilled H2O (pH 7.5) Sufficient quantity 1 liter
Solution 1 20.0 ml
Solution 3:
Solution 2 25.0 ml
Let stand 10 min, filter, and use.
Solution 4: 0.5% Citric acid (aqueous)
Fast green 0.2 g
Solution 5: Distilled H2O 100.0 ml
Glacial acetic acid 0.2 ml
Solution 5 20.0 ml
Solution 6:
Distilled H2O 50.0 ml

71
Arthur A. Andersen and Daisy Vanrompay

Table 16.2 Primer sequences for the nested PCR/EIA

Oligonucleotide bp Sequence (5’- 3’)

Forward outer 21 CCT GTA GGG AAC CCA GCT GAA

Reverse outer 22 GGT TGA GCA ATG CGG ATA GTA T

Fluorescein—forward inner 17 GCA GGA TAC TAC GGA GA

Biotin—reverse inner 18 GGA ACT CAG CTC CTA AAG

Table 16.3. Genotype specific primers and probes and competitors


Oligonucleotide Sequence (5’-3’) Positiona Specificity
CpPsSSfor TTATTAAGAGCTATTGGTGGATGCC 1 822 Cp. psittaci

CpPsSSrev AACGTATAATGGTAGATGATTAATCTACCG 1 972


CpPsGASfor GGTTTTCAGCTGCAAGCTCAA 488 Genotype A
CpPsGASprobe CTACCGATCTTCCAACGCAACTTCCTAACG 512
CpPsGAScompetitorB CTACCGATCTTCCAATGCAACTTCCTAACGb 512
CpPsGASrev CCACAACACCTTGGGTAATGC 565 .
CpPsGBSfor AATAGGGTTTTCAGCTACCAACTCAA 483 Genotype B
CpPsGBSprobe TCTACCGATCTTCCAATGCAACTTCCTAACGTA 511
CpPsGBScompetitorA TCTACCGATCTTCCAACGCAACTTCCTAACGTA 511
CpPsGBScompetitotE+E/B TCTACCGAGCTTCCAATGCAACTTCCTAACGTA 511
CpPsGBSrev CCACAACACCTTGGGTAATGC 565
CpPsGCSfor GCATCGCTCAACCTAAATTGG 929 Genotype C
CpPsGCSprobe TCTGCTGTTATGAACTTGACCACATGGAACC 952
CpPsGCSrev ATTGTGGCTTCCCCTAAAAGG 1 009
CpPsGDSfor AACCACTTGGAACCCAACACTTT 969 Genotype D
CpPsGDSprobe AGGAAAGGCCACAACTGTCGACGG 993
CpPsGDSrev CGAAGCAAGTTGTAAGAAGTCAGAGTAA 1 062
CpPsGESfor CCAAGCCTTCTAGGATCAAGGA 982 Genotype E
CpPsGESprobe TACTTTGCCCAATAATGGTGGTAAGGATGTTCTATC 1 005
CpPsGEScompetitorA+B TGCTTTGCCCAATAATAGTGGTAAGGATGTTCTATC 1005
CpPsGEScompetitorE/B TGCTTTGCCCAATAATGCTGGTAAGGATGTTCTATC 1 005
CpPsGESrev CGAAGCAATTTGCAAGACATCA 1 062
CpPsGFSfor GCAACTTTTGATGCTGACTCTATCC 904 Genotype F
CpPsGFSprobe CATCGCTCAACCTAAATTAGCCGCTGC 930
CpPsGFSrev GTTCCATGTGGTCAAGTTCAAAAC 981
CpPsGE/BSfor CCAAGCCTTCTAGGATCAACCA 982 Genotype E/B
CpPsGE/BSprobe TGCTTTGCCCAATAATGCTGc 1 005
CpPsGE/BScompetitorA+B TGCTTTGCCCAATAATAGTG 1 005
CpPsGE/BScompetitorE TACTTTGCCCAATAATGGTG 1 005
CpPsGE/BSrev TGCAAGACATCAGATAGAACATCCTT 1 052

72
Chapter 16 Chlamydiosis

ML-INF02-F ML-INR03-B

472 bp
<=?
>

872 bp
< >

70 90 184 200 ompA 638 655 920 941

CD1 VD1 CD2 VD2 CD3 VD3 CD4 VD4 CD5

◄-- 1068 bp —►

Figure 16.1: Location of the outer and inner primers in the ompA gene. Numbering according to ompA sequences in Genbank

REFERENCES 15. Everett, K.D.E., RM. Bush, and A.A. Andersen. Emended description
of the order Chlamydiales, proposal of Parachlamydiaceae fam nov. and
1. Al-Soud, W.A, P.--G. Lantz, A. Backman, P. Olcen, and P. Rydstrom. A Simkaniaceae fam. nov., each containing one monotypic genus, revised
sample preparation method which facilitates detection of bacteria in blood taxonomy of the family Chlamydiaceae, including a new genus and five new
cultures by the polymerase chain reaction. J. Microbiol. Meth. 32:217-224. species, and standards for the identification of organisms. Inti. J. Systematic
1998. Bacteriol. 49:415-440. 1999.
2. Andersen, A. A. Serotyping of Chlamydia psittaci isolates using serovar— 16. Everett, K.D.E., L.J. Hornung, and A.A. Andersen. Rapid detection of
specific monoclonal antibodies with the microimmunofluorescence test. J. the Chlamydiaceae and other families in the Order Chlamydiales'. Three
Clin. Microbiol. 29:707-711. 1991. PCR Tests. J. Clin. Microbiol. 37(3):575-580. 1999.
3. Andersen, A.A. Comparison of pharyngeal, fecal, and cloacal samples 17. Fernandez, F., J. Gutierrez, J. Mendoza, J. Linares, and M.J. Soto. A
for the isolation of Chlamydia psittaci from experimentally infected new microimmunofluorescence test for the detection of Chlamydia
cockatiels and turkeys. J. Vet. Diagn. Invest. 8:448-450. 1996. pneumoniae specific antibodies. J. Basic Microbiol. 44(4):275-9. 2004.
4. Andersen, A. A. Two new serovars of Chlamydia psittaci from North 18. Fudge, A.M. Blocking antibody ELISA testing of pet birds:
American birds. J. Vet. Diagn. Invest. 8:159-164. 1997. Implications for chronic infections. Semen. Avian Exotic Pet Med. 2:167—
5. Andersen, A.A. Avian chlamydiosis. In: OIE Manual of standards for 170. 1993.
diagnostic tests and vaccines for terrestrial animals (mammals, birds and 19. Geens, T., A. Dewitte, N. Boon, and D. Vanrompay. Development of a
bees), 5th ed. Office International des Epizooties (OIE), Paris, France, pp. Chlamydophila psittaci species—specific and genotype—specific real-time
856-867. 2004. PCR. Vet. Res. 2005, in press.
6. Andersen, A.A., and D. Vanrompay. Avian chlamydiosis (psittacosis, 20. Geens, T., A. Desplanques, M. Van Loock, B.M. Bonner, E.F. Kaleta,
ornithosis). In: Diseases of poultry, 11th ed. Y.M. Saif, ed. Iowa State S. Magnino, A.A Andersen, K.D. Everett, and D. Vanrompay. Sequencing
University Press, Ames, Iowa. pp. 863-879. 2003. of the Chlamydophila psittaci ompA gene reveals a new genotype, E/B, and
7. Bendheim U., I. Wodowski, M. Ordonez, and A. Naveh. Development of the need for a rapid discriminatory genotyping method. J. Clin. Microbiol.
an ELISA—Kit for antibody detection in psittacine birds. IV DVG Tagung 43(5):2456—61. 2005.
uber Vogelkrankheiten. Munchen. pp. 193-201. 1993. 21. Grimes, J.E., D.N. Phalen, and F. Arizmendi. Chlamydia latex
8. Bevan, B.J., and C.D. Bracewell. Chlamydiosis in birds in Great Britain. agglutination antigen and protocol improvement and psittacine bird anti-
2. Isolations of Chlamydia psittaci from birds sampled between 1976 and chlamydial immunoglobulin reactivity. Avian Dis. 37(3):817-24. 1993.
1984. J. Hyg. 96:453-458. 1986. 22. Grimes, J.E., and F. Arizmendi. Usefulness and limitations of three
9. Bevan, B.J., G.A. Cullen, and W.MF. Read. Isolation of Chlamydia serologic methods for diagnosing or excluding chlamydiosis in birds. J.
psittaci from avian sources using growth in cell culture. Avian Pathol. Am. Vet. Med. Assoc. 209:747-750. 1996.
7:203-211. 1978. 23. Grimes, J.E., T.N. Tully, Jr., F. Arizmendi, and D.N. Phalen.
10. Bovamick, M.R., J.C. Miller, and J.C. Snyder. The influence of certain Elementary body agglutination for rapidly demonstrating chlamydial
salts, amino acids, sugars, and proteins on the stability of rickettsiae. J. agglutinins in avian serum with emphasis on testing cockatiels. Avian Dis.
Bacteriol. 59:509-522. 1950. 38:822-831. 1994.
11. Chirgwin, J.M., A.E. Przybyla, RJ. MacDonald, and W.J. Rutter. 24. Grimes, J.E. Evaluation and interpretation of serological responses in
Isolation of biologically active ribonucleic acid from sources enriched in psittacine bird chlamydiosis and suggested complementary diagnostic
ribonuclease. Biochemistry, 18:5294-5299. 1979. procedures. J. Avian Med. Surg. 10:75—83. 1996.
12. Chomczynski, P, and N. Sacchi. Single-step method of RNA isolation 25 Hewinson, R.G., P.C. Griffiths, B.J. Bevan, S.E. Kirwan, M.E. Field,
by acid guanidinium thiocyanate—phenol—chloroform extraction. Anal. M.J. Woodward, and M. Dawson. Detection of Chlamydia psittaci DNA in
Biochem 162(1): 156—9. 1987. avian clinical samples by polymerase chain reaction. Vet. Microbiol.
13. DeGraves, F.J., D. Gao, and B. Kaltenboeck. High-sensitivity 54(2): 155-66. 1997.
quantitative PCR platform. Biotechniques 34(1): 106-10, 112-5. 2003. 26. Hoymans, V.Y., J.M. Bosmans, L. Van Renterghem, R. Mak, D. Ursi,
14. Everett, K.D.E., and A. A. Andersen. 1999. Identification of nine species F. Wuyts, C.J. Vrints, and M. Ieven. Importance of methodology in
of the Chlamydiaceae using PCR-RFLP. Inti. J. Systematic Bacteriol. determination of Chlamydia pneumoniae seropositivity in healthy subjects
49:803-813. and in patients with coronary atherosclerosis. J. Clin. Microbiol. 41(9)4049-
-53. 2003.
27. Kaltenbock, Β., N. Schmeer, and R. Schneider. Evidence for numerous
ompl alleles of porcine Chlamydia trachomatis and novel chlamydial
species obtained by PCR. J. Clin. Microbiol. 35(7): 1835-41. 1997.

73
Arthur A. Andersen and Daisy Vanrompay

28. Kaltenboeck, B., D. Heard, F.J. DeGraves, and N. Schmeer. Use of 43. Spencer, W.N., and F.W.A. Johnson. Simple transport medium for the
synthetic antigens improves detection by enzyme—linked immunosorbent isolation of Chlamydia psittaci from clinical material. Vet. Rec. 113:535—
assay of antibodies against abortigenic Chlamydia psittaci in ruminants. J. 536. 1983.
Clin. Microbiol. 35(9):2293-8. 1997b. 44. Sting, R., and H.M. Hafez. Purification of Chlamydia psittaci antigen by
29. Kutyavin, I.V., I.A. Afonina, A. Mills, V.V. Gom, E.A. Lukhtanov, E.S. chromatografie on polymyxin B agarose for use in die enzyme-linked
Belousov, MJ. Singer, D.K. Walburger, S.G. Lokhov, A.A. Gall, R. immunosorbent assay (ELISA). Zentralbl. Bakteriol. 277: 436—445. 1992.
Dempcy, MW. Reed, R.B. Meyer, and J. Hedgpeth. 3'-minor groove 45. Tappe, J.P., A.A. Andersen, and N.F. Cheville. Respiratory and
binder-DNA probes increase sequence specificity at PCR extension pericardial lesions in turkeys infected with avian or mammalian strains of
temperatures. Nucleic Acids Res. 28:655-661. 2000. Chlamydia psittaci. Vet. Pathol. 26:386-395. 1989.
30. Ley, D.H., K. Flammer, and P. Cowen. Performance characteristics of 46. Tiran, A., K. Tiesenhausen, E. Karpf, J. Orfila, G. Koch, H.J. Gruber,
diagnostic tests for avian chlamydiosis. J. Assoc. Avian Vet. 7:203-207. O. Tsybrovskyy, and B. Tiran. Association of antibodies to chlamydial
1993. lipopolysaccharide with the endovascular presence of Chlamydophila
31. Lindqvist, R., B. Norling, and S.T. Lambertz. A rapid sample pneumoniae in carotid artery disease. Atherosclerosis. 173(1):47—54. 2004.
preparation method for PCR detection of food pathogens based on buoyant 47. Tully, T.N.,Jr., S.M. Shane, J.E. Grimes, R.P. Poston, and MT.
density centrifugation. Lett. Appl. Microbiol. 24:306—310. 1997. Kearney. Comparison of procedures to detect Chlamydia psittaci antibodies
32. Madico, G., T.C. Quinn, J. Boman, and C. A. Gaydos. Touchdown in cockatiels (Nymphicus hollandicus). Avian Dis. 40(2): 266—71. 1996.
enzyme time release—PCR for detection and identification of Chlamydia 48. Van Loock, Μ, K. Verminnen, T.O. Messmer, G. Volckaert, B.M
trachomatis, C. pneumoniae, and C. psittaci using the 16S and 16S—23 S Goddeeris and D. Vanrompay. Use of a nested PCR-enzyme immunoassay
spacer rRNA genes. J. Clin. Microbiol. 38(3): 1085-93. 2000. with an internal control to detect Chlamydophila psittaci in turkeys. 2005,
33. Messmer, T. O., S. K. Skelton, J. F. Moroney, H. Daugharty, and B. S. BMC Infect. Dis. 5:76, 2005.
Fields. Application of a nested, multiplex PCR to psittacosis outbreaks. J. 49. Vanrompay, D., R. Ducatelle, and F. Haesebrouck. Diagnosis of avian
Clin. Microbiol. 1997; 35:2043-2046. chlamydiosis: Specificity of the modified Gimenez staining on smears and
34. Morre, S.A., C. Munk, K. Persson, S. Kruger-Kjaer, R. van Dijk, C.J. comparison of the sensitivity of isolation in eggs and three different cell
Meijer, and A. J. van Den Brule. Comparison of three commercially cultures. J. Vet. Med. Ser. B 39:105-112. 1992.
available peptide-based immunoglobulin G (IgG) and IgA assays to 50. Vanrompay, D., A. Van Nerom, R. Ducatelle, and F. Haesebrouck.
microimmunofluorescence assay for detection of Chlamydia trachomatis Evaluation of five immunoassays for detection of Chlamydia psittaci in
antibodies. J. Clin. Microbiol. 40(2):584-7. 2002. cloacal and conjunctival specimens from turkeys. J. Clin. Microbiol.
35. Moore, F.M, and ML. Petrak. Chlamydia immunoreactivity in birds 32:1470-1474. 1994.
with psittacosis: Localization of chlamydiae by the peroxidase­ 51. Vanrompay, D., E. Cox, J. Mast, B. Goddeeris, and G. Volckaert. High-
antiperoxidase method. Avian Dis. 29:1036-1042. 1985. level expression of Chlamydia psittaci major outer membrane protein in
36. Paul, I.D. The growth of Chlamydia in McCoy cells treated with COS cells and in skeletal muscles of turkeys. Infect. Immun. 66(11): 5494-
emetine. Med. Lab. Sci. 39:15-32. 1982. 500. 1998.
37. Ridstrom, P., R. Knutsson, P. Wolffs, M Dahlenborg, and C. Lofstrom. 52. Vanrompay, D., E. Cox, P. Kaiser, S. Lawson, M Van Loock, G.
Pre—PCR processing of samples. In: PCR detection of microbial pathogens. Volckaert, and B. Goddeeris. Protection of turkeys against Chlamydophila
K. Sachse, and J. Frey, eds. Humana Press, Totowa, NJ. pp. 31-50. 2003. psittaci challenge by parenteral and mucosal inoculations and the effect of
38. Sachse, K. and H. Hotzel. Detection and differentiation of Chlamydiae turkey interferon-gamma on genetic immunization. Immunology.
by nested PCR. In: PCR detection of microbial pathogens. K. Sachse and J. 103(1): 106-12. 2001.
Frey, eds. Humana Press, Totowa, NJ. pp. 123-136. 2003. 53. Vanrompay, D., T. Geens, A. Desplanques, T.Q. Hoang, L. De Vos, M
39. Sachse, K, H. Hotzel, P. Slickers, T. Ellinger, and R. Ehricht. DNA Van Loock, E. Huyck, C. Mirry, and E. Cox. Immunoblotting, ELISA and
microarray-based detection and identification of Chlamydia and culture evidence for Chlamydiaceae in sows on 258 Belgian farms. Vet.
Chlamydophila spp. Mol. Cell Probes 19(1):41—50. 2005. (Epub 2004 Nov Microbiol. 99(1):59—66. 2004.
Π) 54. Wilson, P.A., J. Phipps, D. Samuel, andN.A. Saunders. Development of
40. Salinas, J., MR. Caro, and F. Cuello. Comparison of different a simplified polymerase chain reaction—enzyme immunoassay for the
serological methods for the determination of antibodies to Chlamydia detection of Chlamydia pneumoniae. J. Applied Bacteriol. 80:431—438.
psittaci in pigeon sera. Zentralbl. Veterinarmed. B. 40(4):239—44. 1993. 1996.
41. Sayada, C., A.A. Andersen, C. Storey, A. Milon, F. Eb, N. Hashimoto, 55. Wong, K.H, Skelton S.K., and H. Daugharty. Utility of complement
K. Hirai, J. Elion, and E. Denamur. Usefulness of ompl restriction mapping fixation and microimmunofluorescence assays for detecting serologic
for avian Chlamydia psittaci isolate differentiation. Res. Microbiol. responses in patients with clinically diagnosed psittacosis. J. Clin.
146:155-165. 1995. Microbiol. 32(10):2417-21. 1994.
42. Smith, K.A., K.K. Bradley, MG. Stobierski, and L.A. Tengelsen. 56. Yoshida, Η., Y. Kishi, S. Shiga, and T. Hagiwara. Differentiation of
Compendium of measures to control Chlamydophila psittaci (formerly Chlamydia species by combined use of polymerase chain reaction and
Chlamydia psittaci) infection among humans (psittacosis) and pet birds, restriction endonuclease analysis. Microbiol. Immunol. 42(5):411—4. 1998.
2005. J. Am. Vet. Med. Assoc. 226(4):532-39. 2005.
17
ORNITHOBACTERIOSIS
Richard P. Chin and Bruce R. Charlton

SUMMARY. Omithobacteriosis is a respiratory disease of avian species caused by the bacterium Ornithobacterium rhinotracheale. Gross
lesions in infected birds include a fibrinoheterophilc pneumonia, airsacculitis, and pericarditis. Ornithobacterium rhinotracheale is a
pleomorphic gram-negative rod-shaped bacterium that grows on blood agar. Seventeen serotypes (A through Q) are currently recognized,
though serotype A is the major one isolated. The bacterium has been isolated from numerous avian species, primarily infecting chickens and
turkeys.
Agent Identification. Isolation and identification of the bacterium are required to make a diagnosis.
Serologic Detection in the Host. Enzyme-linked immunosorbent assays and serum plate agglutination tests have been developed to detect
antibodies.

INTRODUCTION airsacculitis, pericarditis, peritonitis, and a mild tracheitis could


occur.
Ornithobacterium rhinotracheale is associated with respiratory
disease in avian species, primarily turkeys and chickens, but the SAMPLE COLLECTION
bacterium also has been isolated from chukars, rooks, and a
partridge, pheasant, and pigeon (2,4,10). The first reported isolation The primary site for isolating O. rhinotracheale is the respiratory
of O. rhinotracheale was made from turkeys in Germany in 1981 tract. Taking tracheal cultures from live birds should be done using
(6). The bacterium has since been isolated from birds throughout a sterile swab. Care must be taken to prevent contamination as
the world. large numbers of other bacteria can easily overgrow the pinpoint
Prior to being named in 1994, O. rhinotracheale had been colonies of O. rhinotracheale. Samples can be taken at necropsy
identified as Pasteurella-\&Q organism, Kingella-tiks bacterium, using sterile swabs (cotton or dacron) from the trachea, infraorbital
Taxon 28, and pleomorphic gram-negative rod bacterium. sinuses, lungs, and air sacs. Reports have been made of isolation of
Identification of O. rhinotracheale by biochemical test kits can be O. rhinotracheale from joints, heart, brain, and fiver, and in chronic
challenging. Current commercially available identification systems disease, from the thoracic vertebrae and tendon sheaths. Isolates
do not have O. rhinotracheale listed in their database and may can be stored at -70 C.
misidentify it as Gallibacterium anatis biovar haemolytica
(formerly Pasteurella haemolytica), Weeksella spp., or PREFERRED CULTURE MEDIA AND SUBSTRATES
Flavobacterium meningosepticum, depending on the test system and
variable reactions of this bacterium. For primary isolation, O. rhinotracheale gjtows readily on 5%
sheep blood agar. It grows aerobically, microaerobically, and
CLINICAL DISEASE anaerobically. The best growth occurs in air enriched with 7.5%-
10% CO2 at 37 C, although growth occurs from 30 to 42 C.
Whether or not O. rhinotracheale is a primary pathogen is still Ornithobacterium rhinotracheale does not grow on MacConkey
uncertain. In many cases, in both chickens and turkeys, infection agar.
with O. rhinotracheale is secondary to other respiratory disease About 90% of the isolates appear to be resistant to gentamicin and
agents (9). polymyxin B (11). Hence, 5pg/ml each of gentamicin and
In chickens, infections have been reported in 3-to-6-wk-old birds, polymyxin B can be added to blood agar media for selective
and in layers 20-50 wk of age (5,6,8,9,12). Mild respiratory signs isolation of O. rhinotracheale. However, blood agar plates without
occur from about 3 to 4 wk of age, mortality mildly increases, and antibiotics should always be used in tandem to prevent missing the
airsacculitis condemnation rates at the slaughter plant are higher. In 10% antibiotic-susceptible isolates.
broiler breeders, the disease primarily occurs between 20 and 50 wk
of age, during peak egg production (5). Mortality is slightly AGENT IDENTIFICATION
increased, feed intake is decreased, and mild respiratory signs
occur. There may be egg production decrease, eggshell may be of Colony and Cell Morphology
poor quality, and egg size may decrease. On blood agar, pinpoint colonies, less than 1 mm, can be detected
In turkeys, infections have been detected as early as 2 wk of age, at 24 hr of incubation. At 48 hr, colonies will be approximately 1-2
but severe lesions are seen in older birds (>14wk of age) and mm in diameter, gray, circular, and convex with an entire edge.
breeders (3,5,11). Infection with O. rhinotracheale at 2 wk of age Some isolates from chickens have a reddish glow. Plates should be
causes respiratory signs and nasal discharge, followed by facial held for 48 h to look for the small colonies typical of
edema and swelling of the infraorbital sinuses (9). Birds will O. rhinotracheale as they can easily be overgrown by other bacteria
appear depressed with ruffled feathers, and display a decrease in such as Escherchia coli. Ornithobacterium rhinotracheale cultures
feed and water intake. Subsequently, mortality increases. In older can have a distinct smell similar to butyric acid.
birds, a sudden increase in mortality may be the only sign. Slight Ornithobacterium rhinotracheale is a gram-negative, highly
depression and gasping, marked dyspnea, and expectoration of pleomorphic, nonmotile, nonsporulating bacterium. It appears
blood-stained mucus can be seen just prior to death. In breeder primarily as short, plump rods 0.2-0.9 pm x 1-3 pm, and less
flocks, egg production can decrease slightly (2-5%) (3). Mortality frequently as long filamentous rods or club-shaped rods (10).
rates usually range between 2% and 11% in chickens and turkeys.
Clinical cases reveal unilateral and bilateral lung consolidation Biochemical Characteristics
with fibrinous exudate on the pleura in both chickens and turkeys Biochemical tests for O. rhinotracheale are inconsistent. Listed in
(3,11). These lesions are similar to those seen in fowl cholera in Table 17.1 are some of the more consistent reactions.
turkeys, but are less severe. In addition, a fibrinoheterophilic
75
Richard P. Chin and Bruce R. Charlton

Table 17.1. Some phenotypic characteristics of Ornithobacterium DIFFERENTIATION FROM CLOSELY RELATED AGENTS
rhinotracheale.
Test Reaction Ornithobacterium rhinotracheale infection should be
Blood agar Growth differentiated from other diseases with pneumonia and airsacculitis,
MacConkey agar No growth
such as fowl cholera, colibacillosis, mycoplasmosis, and
Gram stain Pleomorphic gram-neg. rod
Catalase Negative
chlamydophylosis. This is dependent upon isolation and
Oxidase Positive identification of O. rhinotracheale from infected tissues.
Triple sugar iron agar No change
β-Galactosidase (ONPG) Positive REFERENCES
Indole Negative
1. Back, A, D. Halvorson, G. Rajashekara, K. V. Nagaraja. Development of
a serum plate agglutination test to detect antibodies to Ornithobacterium
The API-20NE identification strip (bioMerieux-Vitek, Hazelwood,
rhinotracheale. J. Vet. Diagn. Invest. 10:84-86. 1998
Mo.) is routinely used, although O. rhinotracheale is not included 2. Charlton, B. R., S. E. Channing-Santiago, A. A. Bickford, C. J. Cardona,
in their database, and there are various biocodes. The most common R. P. Chin, G. L. Cooper, R Droual, J. S. Jeffrey, C. U. Meteyer, H. L.
biocodes are 0220004 (61%) and 0020004 (38.5%) (11). If the Shivaprasad, and R. L. Walker. Preliminary characterization of a
isolate is positive for arginine dihydrolase (0.5%) biocodes of pleomorphic gram-negative rod associated with avian respiratory disease. J.
0320004 and 0120004 are produced. The API ZYM system Vet. Diagn. Invest. 5:47-51. 1993.
(bioMerieux-Vitek, Hazelwood, Mo.) is useful for determining 3. Chin, R. P., P. C. M van Empel, and Η. M Hafez. Ornithobacterium
enzymatic activities. In this system O. rhinotracheale consistently rhinotracheale infection. In: Diseases of poultry, 11th ed. Y. M Saif, H. J.
produces five negative reactions (lipase, β-glucuronidase, β- Bames, J. R. Glisson, A. M Fadly, L. R. McDougald, and D. E. Swayne,
eds. Iowa State University Press, Ames, Iowa. pp. 683-690. 2003.
glucosidase, α-mannosidase, and α-fucosidase). Another rapid test
4. De Rosa, M., R. Droual, R. P. Chin, H. L. Shivaprasad, and R. L.
system, the RapID NF Plus, (Remel/Atlanta, Norcross, Ga.) also Walker. Ornithobacterium rhinotracheale infection in turkey breeders.
appears to be suitable for identification of O. rhinotracheale (7). Avian Dis. 40:865-874. 1996.
Field isolates generated 5 unique biocodes: 472264 (41.8%), 5. Hafez, Η. M Current status on the role of Ornithobacterium
476264 (31.8%), 676264 (18.2%), 672264 (7.3%) and 472044 rhinotracheale (ORT) in respiratory disease complexes in poultry. Arch.
(0.9%). Carbohydrate fermentation is extremely variable. Geflugelk. 60:208-211. 1996.
Analysis of cellular fatty acids can aid in identifying 6. Hinz, K.-H., and Η. M Hafez. The early history of Ornithobacterium
O. rhinotracheale. Predominant fatty acids detected using the rhinotracheale (ORT). Arch. Geflugelk 61:95-96. 1997.
7. Post, K. W., S. C. Murphy, J. B. Boyette, and P. M Resseguie.
Microbial Identification System (Microbial ID, Newark, Del.) are
Evaluation of a commercial system for the identification of
15:0 iso, 16:0, 15:0 iso 3OH, 17:0 iso, 16:0 3OH, 17:0 iso 3OH, and Ornithobacterium rhinotracheale. J. Vet. Diagn. Invest. 11:97-99. 1999.
unknown peaks with equivalent chain lengths of 13.566 and 16.580 8. Travers, A. F. Concomitant Ornithobacterium rhinotracheale and
(2). Newcastle disease infection in broilers in South Africa. Avian Dis. 40:488-
Seventeen serotypes of O. rhinotracheale are reported, serotype A 490. 1996.
through Q. These have been differentiated using the agar gel 9. van Beek, P.N.G.M, P. C. M van Empel, G. Van Den Bosch, P. K.
immunodiffusion test (8), (P. van Empel, pers. comm.). Some Storm, J. H. Bongers, and J. H. du Preez. Ademhalingsproblemen,
geographical differences exist in serotypes found throughout the groeivertraging en gewrichtsontsteking bij kalkoenen en vleeskuikens door
een Pasteurella-achtige bacterie: Ornithobacterium rhinotracheale or “Taxon
world. Most isolates found in Europe and the United States are
28.” Tijdschr. Diergeneeskd 110:99-101. 1994.
serotype A. After serotype A, types B, D, and E are the predominate 10. Vandamme P., P. Segers, M. Vancanneyt, K. van Hove, R. Mutters, J.
serotypes found in Europe. Ninety-five percent of the strains Hommez, F. Dewhirst, B. Paster, K. Kersters, E. Falsen, L. A Devriese, M
isolated from chickens are serotype A. Bisgaard, K.-H. Hinz, and W. Mannheim. Ornithobacterium rhinotracheale
gen. nov., sp. nov., isolated from the avian respiratory tract. Int. J. Syst.
SEROLOGIC DETECTION IN THE HOST Bacteriol. 44:24-37. 1994.
11. van Empel, P. C. M, and Η. M Hafez. Ornithobacterium
Enzyme-linked immunosorbent assays (ELISAs) have been rhinotracheale: a review. Avian Pathol. 28: 217-227. 1999.
developed and appear adequate for detecting antibodies to most 12. van Empel, P., H. van den Bosch, D. Goovaarts, and P. Storm.
Experimental infection in turkeys and chickens with Ornithobacterium
serotypes of O. rhinotracheale (5,8,11). Commercial ELISA kits are rhinotracheale. Avian Dis. 40:858-864.1996.
available in Europe and appear to detect antibodies for serotypes A
through M. A serum plate agglutination test has also been
developed to detect antibodies to O. rhinotracheale (1).

76
18
MYCOSES AND MYCOTOXICOSES
R. D. Wyatt

SUMMARY. Mold-related disease in avian species can be divided into two broad categories, namely mycoses and mycotoxicoses. Mycoses
are typically defined as infection of tissue by a particular mold species. In general terms, Aspergillus, Dactylaria, and Microsporum are those
mold species most apt to be responsible for mycoses in avian species. Additionally, Candida, a genus of polymorphic yeasts (exhibiting yeast
cells, hyphae and pseudohyphae) can also infect the upper gastrointestinal tract. All of these agents should be considered economically
important to domestic poultry, however, Aspergillus is by far the most common.
Mycotoxins are a broad and diverse group of toxic metabolites produced by toxigenic molds when these molds are permitted to grow on
certain feedstuffs. Consumption of such moldy feedstuffs by avian species may result in clinical signs of disease, dependent upon the specific
mycotoxin that is contaminating the ingested feedstuff. The three most important mold genera that are known to have species that posses the
capability to produce mycotoxins are Fusarium, Aspergillus, and Penicillium. Trichothecenes, a rather large group of mycotoxins with
similar chemical structures, are produced primarily by species of Fusarium. Aflatoxin, perhaps the most toxic of all mycotoxins, is the most
important mycotoxin produced by Aspergillus. Ochratoxin is also an economically important mycotoxin and is produced by species of
Penicillium and Aspergillus.
Isolation and identification of molds responsible for mycoses can be accomplished in infected tissues by employing basic microbiological
techniques. Additionally, histological examination of infected tissue and examination of the etiological agent and tissue response can also
confirm the identity of die pathological agent.
Isolation and identification of specific mycotoxins requires extraction of the contaminated feedstuff followed by chemical analysis of the
extract. Additionally, flock history, management practices and a working knowledge of specific clinical signs of various mycotoxicoses is
essential to identify the specific mycotoxin involved in a disease outbreak.

ASPERGILLOSIS

INTRODUCTION

The term “fungus” is a general term used to describe any member dramatically. Consequently, the signs of mycotoxicoses will vary
of a very broad taxonomic classification of eukaryotic depending on the specific mycotoxin(s) involved in the disease.
microorganisms (protists) including lichens, mushrooms, yeasts, Signs of disease are also influenced by various interacting factors
rusts, smuts, molds, and numerous other organisms. These such as the presence of other diseases, nutritional status of affected
organisms have no chlorophyll, are aerobic, and have a very rigid birds, environmental conditions, and husbandry practices (11, 13,
cell wall containing chitin. Molds are a subset of fungi that degrade 17, 34, 35, 37, 39). Hence, a rapid and accurate diagnosis of specific
organic material in the environment. They are ubiquitous, mycotoxicoses is often very difficult.
saprophytic, parasitic, and their spores are resistant to many
environmental conditions, especially desiccation. When growing in CLINICAL DISEASE
culture, most molds will first produce single filaments known as
hyphae and as the culture matures it will appear filamentous and Aspergillosis can be caused by any member of the genus
assume a "wooly” appearance. Mature cultures are frequently Aspergillus, however, Aspergillus fumigatus is the most common
pigmented due to the production of pigments by the mycelium or etiologic agent for this disease in poultry (7, 18, 22). The lungs
pigments associated with spores. Pathogenic molds typically and air sacs are the typical tissues involved, however, infection of
reproduce by formation of spores or conidia; whereas, Candida the eye, sinuses, and meninges can occur. When responsible for
albicans reproduces by budding. A relatively small percentage of mycoses, Aspergilli are generally considered opportunistic
molds in die environment are causative of disease of poultry. pathogens. Immunosuppression, respiratory tract irritation,
Nevertheless, based upon routine exposure of poultry to molds, the antibiotic therapy and die presence of other infective agents can
probability for disease outbreaks to occur in commercial poultry is predispose birds to aspergillosis (2).
significant. Infections of the upper respiratory tract by Aspergillus In young broilers, poults, and quail, and a variety of wild birds (7,
fumigatus and other Aspergilli and infections of the upper 12, 22) high morbidity and mortality are commonly observed.
gastrointestinal tract of poultry by Candida albicans are well- Improperly fumigated hatching eggs and improperly disinfected
documented (7,40). incubators and hatchers can result in high exposure rates to the
Other mold species are merely environmental inhabitants with soil spores of Aspergilli resulting in infection of chicks with the disease
being their natural ecological niche. These molds are typically progressing rapidly. Consequently, the term “brooder pneumonia”
associated with degradation of living or dead organic material. is often used to describe acute aspergillosis in young birds.
Those degrading living organic material are often plant pathogens; Impairment of respiratory function followed by lethargy and
whereas, those degrading dead organic material are best known for anorexia can lead to death in young birds within 24-72 hr. Adult
their capability to produce secondary metabolites such as antibiotics turkeys are also susceptible to aspergillosis. Although the signs of
(e.g. Penicillium chrysogenum). Some molds possess the genetic the disease are similar to those of young turkeys, chickens and
capability to produce highly toxic secondary metabolites during quail, the disease is generally of a longer duration. Coughing,
their growth on organic material. These toxic metabolites are sneezing, and general labored breathing are commonly observed.
referred to as mycotoxins. Ingestion of mycotoxins by poultry in Upon necropsy, the internal lesions can be described as focal
grain or finished feed will result in disease referred to as caseous nodules on the air sacs, imbedded within the lungs and on
mycotoxicosis. Mycotoxicoses in poultry are quite diverse with the mucosal surface of the trachea. Occasionally, greenish mold
each being caused by a specific mycotoxin and each toxicosis growth can be visualized on the air sacs. This is typically associated
characterized by a primary target organ system, target organ, or with the chronic form of the disease. When this occurs, the lesions
target tissue. The toxicity of various mycotoxins to poultry differ associated with other internal organs may appear yellowish in color.

77
R. D. Wyatt

SAMPLE COLLECTION AGENT IDENTIFICATION

Upon necropsy, areas of suspected infected tissue should be Presumptive identification of Aspergillus is based upon
carefully identified, aseptically excised from surrounding tissue and microscopic morphology and growth on various microbiological
immediately transferred to a sterile vessel. Retrieval of suspect media. Aspergilli typically exhibit hyaline (glassy appearance),
tissue from both the periphery and center of a lesion is septate, and branched hyphae. The color of the conidiophore can be
recommended. Retrieved tissue should be subjected to both direct colorless, brown or green depending upon the species (7). The
microscopic examination and microbiological culturing. Care conidiophore with be rather long (> 300 microns) in the case of
should be taken in maintaining aseptic conditions since Aspergilli A. niger, and short (<300 microns) in length in the case of A.
are common in the laboratoiy microflora. When samples are being fumigatus (9). The round or spherical conidia supported by the
prepared for microscopic examination, immediate fixing of the coniophore are usually arranged in either a radiate or columnar
specimen is imperative. Also, culturing procedures should be fashion. Aspergillus fumigatus is typically in a columnar
initiated without delay following removal of the infected tissue. configuration; whereas, A. niger is typically in a radiate
configuration.
PREFERRED CULTURE MEDIA AND SUBSTRATES
Direct Microscopic Examination
Place a small subsample of the suspected infected tissue in a sterile Specimens of infected tissue can be mixed with 10% KOH on a
container with a sufficient volume of sterile saline to facilitate sterile microscope side and then stabilized by covering with a cover
disruption of tissue. Disruption of tissue can be accomplished with a slip. With large pieces of tissue, the KOH concentration can be
sterile glass rod or sterile spatula. Alternatively, infected tissue can increased to 20% and allowed to incubate to permit sufficient
be transferred directly to culture medium in a Petri dish or clarification of the tissue to permit visualization of the mold.
thoroughly homogenized in a tissue grinder or microblendor prior to Infecting mold can be visualized and detected by the presence of
transfer. In any case, following aseptic transfer of the specimen to branching septate hyphae or mycelium. If mold is detected in the
the surface of the Petri dish, streaking or spreading of the specimen tissue it is advisable to prepare another specimen for histological
over the surface of the Petri dish should be accomplished with a examination.
sterile inoculating loop.
Several types of microbiological media can be used for culturing Histological Examination of Infected Tissue
Aspergilli. Sabouraud’s dextrose agar, malt agar, and potato Suspect tissue should first be fixed in 10% neutral buffered
dextrose agar have proven ideal for the growth of numerous species formalin, embedded in paraffin and sections prepared for staining.
of Aspergillus (2). Regardless of the choice of medium, fortification Several stains are available for detection and identification of mold
of the medium with 40-50 micrograms of chloramphenicol/ml of in tissue. Examples are Girdley’s fungus, periodic acid-Schiff
medium should be employed to inhibit any bacteria that may be in (PAS), hematoxylin and eosin (H&E) and Gomori methenamine
the specimen. This is essential since bacteria, if present, will grow silver (GMS) (5). Of these, GMS is usually preferred (5). However,
more rapidly than molds present in the specimen. The rapid growth routinely stained specimens with H&E will stain most species of
of bacteria will impede or even prevent the development of the Aspergilli, but is not acceptable for most other mold genera. Stained
slower growing molds. Incubation of the Petri dish cultures should sections infected with mold will have structures similar to those
be done at 30 C or up to 10 days. described above. Hyaline, septate, and branched hyphae with a
During incubation of the inoculated cultures, Aspergillus growth consistent diameter can be detected. In some cases of aspergillosis,
will typically be evident within 2-3 days of incubation and will first the conidial head and even conidia may be present.
appear white in color and filamentous in texture. If the mold
sporulates, it may turn light green to gray in color by day 5-10. The
reverse of the Petri dish culture will typically be white to tan in
color. Not all Aspergilli display a greenish coloration during spore
formation, but A. fumigatus and many other species do.

CANDIDIASIS

CLINICAL DISEASE poor feathering, lethargy, and C. albicans septicemia in rare cases
(40).
Avian candidiasis, also known as crop mycosis, is an infection of
the upper gastrointestinal tract. Turkeys are more susceptible to SAMPLE COLLECTION
candidiasis than chickens (40). The primary etiologic agent for this
disease is Candida albicans, however, C. tropicalis, C. labrata, C. In intact birds, the upper gastrointestinal tract (mouth, esophagus,
parapsilosis, C. krusei, and C. lusitaniae have also been isolated and crop) can be swabbed with a dry sterile cotton swab by gently
from outbreaks of candidiasis (2,40). C. albicans is common in rubbing the swab on these internal surfaces. At this point, the swab
small numbers in the gastrointestinal tract of birds, however this can be used to inoculate the surface of a Petri dish containing an
opportunistic pathogen tends to colonize the bird during early age, appropriate medium and subsequently incubated. On the other hand,
following long-term use of antibiotics, during stages of after swabbing, the end of the swab can be separated from the swab
immunodeficiency, and during times of generalized stressful stick with sterile scissors while the used portion of the swab is
situations, such as pre-existing infection, poor sanitation, allowed to drop into a sterile container containing a small volume of
overcrowding, poor nutrition, and environmental extremes. neutral buffered saline. Following gentle agitation to release and
During infection, the crop wall will become thickened and opaque suspend any yeast cells into the saline, a measured volume of the
and during advanced cases, focal areas of white yeast growth can be saline can be transferred directly to the surface of a Petri dish
visualized. During advanced cases, the mucosal surface will present containing an appropriate medium, streaked and subsequently
a “Turkish towel” appearance. The esophagus may be characterized incubated. Alternatively, the suspension can be serially diluted and
by having a thick exudate covering the mucosal lining. Affected pour-plate technique used to enumerate the recovered organisms
birds may appear clinically normal or may exhibit poor growth, (2).
78
Chapter 18 Mycoses and Mycotodcoaes

During necropsy, the mouth can be swabbed as described above. somewhat more intense in color and with an overall dull luster to
The crop can be opened with sterile scissors, rinsed with water and the colony surface.
then swabbed with a sterile cotton swab and plated as described.
AGENT IDENTIFICATION
PREFERRED CULTURE MEDIA AND SUBSTRATES
Confirmation of Candida albicans can be accomplished based
Collected specimens should be streaked onto Petri dish cultures upon chlamydospore formation. Cornmeal agar is typically used for
containing Pagano Levin Base (Difco Laboratories, Detroit, MI) chlamydospore formation. The cornmeal agar is inoculated with the
fortified with 100 micrograms of 2, 3, 5-triphenylterazolium suspect yeast, incubated at 25 C for 2-3 days and examined
chloride (TTC) and 500 micrograms of neomycin per ml of medium microscopically for the presence of large chlamydospores typically
and prepared in strict adherence to manufacturer’s along the border of pseudomycelium or at the pseudomycelium
recommendations. Under some situations, the presence of terminus (2).
neomycin-resistant bacteria in the sample will tend to impede the Another confirmation of Candida albicans is based upon germ
growth, enumeration, or visualization of Candida species. If this tube formation. A 0.5 ml volume of sterile serum is placed in a
occurs, gentamicin (50 micrograms per ml of medium) can be sterile test tube and inoculated with actively growing yeast cells
added to the medium in addition to the neomycin. Petri dish cultures (confirmed by microscopic examination) and incubated at 37 C for
of Candida species should be incubated at 30 C for 72 hr. Candida 3 hr. Following incubation, remove of a small volume of serum and
species will appear as rather large round colonies with a regular examine microscopically. The presence of germ tubes originating
border, dull or glistening luster. Depending upon the Candida from parent yeast cells is consistent with of C. albicans (2).
species growing on the surface of the medium, the color of Fermentation of specific carbohydrates, carbohydrate assimilation
individual colonies will be cream to pink to light red in color. tests, and nitrogen utilization tests are extremely useful in
Furthermore, the colonial characteristic is described as having a identification of yeasts, particularly when identifying less frequently
raised center (similar to a fried egg). Candida albicans will encountered yeasts, variants, and atypical isolates. These tests are
typically be cream to light pink in color with the raised center being essential in taxonomic studies, but offer little additional information
in identifying most avian yeast pathogens (16).

CLINICAL DISEASE technique for tissue collection is the same as previously described
for aspergillosis.
Dactylariosis is term used to refer to the fungal encephalitis caused
by the thermophilic mold Ochroconis (Dactylaria) gallopavum PREFERRED CULTURE MEDIA AND SUBSTRATES
(gallopava) (27). For taxonomic purposes, D. gallopava is the now
obsolete synonym for Ochroconis gallopavum. However, based Homogenized portions of affected brain tissue characterized by
upon general usage of D. gallopava by most in the poultry industry, granuloma or necrosis are plated and streaked on Sabouraud’s
this synonym will be used herein. Daciy/arza-caused encephalitis dextrose agar containing 50 micrograms of chloramphenicol/ml of
is known to affect turkey poults, young chicks, quail, and wildbirds medium. Incubation should be done at 37 C for 5 days.
(3, 21, 25, 26). The target tissue of affected birds is the brain,
specifically the cerebrum. Infected tissue will be characterized by AGENT IDENTIFICATION
multiple granuloma and localized necrosis. Affected birds may
exhibit ataxia, loss of motor control, and torticollis. Although this Colonies of Dactylaria will appear as typical mold colonies,
disease is relatively rare, a high percentage of birds within an grayish brown in color, with the underside of the colony being deep
affected flock may exhibit mortality rates from 5-20%. purple-red or intensely wine-colored. This pigment, typically
Thermophilic fungi, including Dactylaria, have been found in produced by Dactylaria will diffuse into the medium and will create
several materials (24, 29, 36), some of which can be used in the a pigmented border surrounding each mold colony or may pigment
poultry industry and may be the source of introduction of the the entire culture plate if numerous colonies are present (10). When
pathogen to susceptible birds. In numerous cases, the utilization of mycelial growth is teased from a colony and placed in a small
aged pine and hardwood shavings as litter has been linked to the volume of mounting fluid, Dactylaria has distinct morphology.
introduction of Dactylaria in a flock. Individual hyphae are septate, the conidiophore is erect and
unbranched, conidia are two-celled, oval to tear-drop in
SAMPLE COLLECTION morphology. Very young conidia may be more round than oval and
may be single celled.
During necropsy, a sample of the granuloma should be collected for
microbiological culture and histopathology assessment. The

RINGWORM (FAVUS)

CLINICAL DISEASE

Favus is a dermatophytosis that affects the comb, wattles, and prevalent resulting in marked feather loss. Affected birds will
feather follicles of chickens and turkeys. The most common appear lethargic and may show anorexia and generalized loss of
etiological agent is Microsporum gallinae (previously Trichophyton condition. This disease is uncommon in commercial poultry flocks
gallinae). This disease is highly transmissible by bird-to-bird are occurs in low frequency in backyard flocks and exotic species.
contact or contact with a contaminated environment. The disease Strict environmental sanitation, isolation, and culling of affected
presents as white, wrinkled or crusty lesions primarily on the comb birds are recommended.
and wattles. Infection of feather shafts and follicles are also
79
R. D. Wyatt

SAMPLE COLLECTION however the color will generally be more intense and diffusion into
the medium can be observed. Direct microscopic and histological
Suspect areas of infection can be removed from the bird by gently examination of recovered mycelial growth will reveal both micro-
scraping affected areas of epidermal tissue. Scrapings can be and macroconidia. Microconidia are usually the more abundant and
cleared with 10% KOH for direct microscopic examination or will have a pear-shaped morphology. Macroconidia are
transferred to microbiological medium for growth of the mold, multicellular (4-6 cells), have a thick wall and are pear to club
isolation, and identification. shaped. Frequently, the branched hyphae will break into chains of
arthroconidia. The same structures can be visualized in scrapings
PREFERRED CULTURE MEDIA AND SUBSTRATES with direct microscopic examination after clearing with 10% KOH
or in histological sections stained with GMS or Periodic Acid Schiff
Microsporum gallinae grow abundantly on Sabouraud’s dextrose (2,4, 5).
agar supplemented with 50 micrograms of chloramphenicol/ml of
medium. Incubation of Petri dish cultures should be done at 27-30
C for 7-14 days.

AGENT IDENTIFICATION

Colonies of M. gallinae will appear as white to cream colored and


will have a suede-like texture. Pigment formation (pink or peach in
color) can be observed along the periphery of developing and aging
cultures. The underside of Petri dish colors will have a similar color
to that observed on the periphery of the top side of the culture,

MYCOTOXICOSES

INTRODUCTION Aspergillus ochraceus are the primary producers of ochratoxin. All


of these mycotoxins are known to adversely affect poultry health
Mycotoxins are defined as highly toxic secondary metabolites and productivity. On the contrary, Fusarium roseum and other
produced by certain molds during the growth of these molds on a Fusarium spp. are known to produce zearalenone. This mycotoxin
suitable substrate. Mycotoxins can either be associated with the is highly toxic to swine and cattle, but has literally no impact on
mold mycelium, extruded into the microenvironment surrounding poultry health. Vomitoxin is another mycotoxin produced by
the mold or both. Several key points are important in understanding Fusarium and has relatively low toxicity to poultry compared to
the relationship of mycotoxins to disease in poultry. First, a many other mycotoxins (38). On the other hand, the prevalence of
relatively small percentage of molds in the environment have the vomitoxin as a common contaminant of com, the primary cereal
genetic capacity for mycotoxin production. Secondly, mold growth grain for poultry and other animals, leads to the conclusion that this
must occur before mycotoxins can be produced in an environment. mycotoxin may be an insidious toxicant in animal agriculture (33).
The most important factor promoting mold growth in nature is the Mycotoxicoses are known to interact with various conditions and
availability of moisture. Molds do not grow and mycotoxins are not circumstances associated with poultry production. For example,
produced in very dry environments, however, the mold spores can aflatoxin tends to exhibit a more pronounced effect on poultry
remain viable for years in environments with almost no water. health when birds are exposed to a low environmental temperature,
Resistance to desiccation is common in molds. Lastly, molds must compared to the response of birds exposed to a high environmental
have a suitable substrate (with sufficient moisture) in order to grow. temperature (17). Young chicks are much more susceptible to
Dead or living organic material is an ideal substrate for mold aflatoxin than are older birds. A low nutritional plane tends to
growth. Most feed grains, grain products and finished poultry feed accentuate the response of poultry to most mycotoxins (34).
are ideal substrates for mold growth. Immunosuppression is caused by several mycotoxins (23,30).
Mycotoxins as a broad class of chemicals are produced by a very During these mycotoxicoses, secondary viral, bacterial and mold
diverse population of molds. Additionally, the chemical structures infections can occur in birds that are experiencing mycotoxicoses.
of mycotoxins differ greatly. Hence, as would be expected, the Consequently, a rapid and accurate diagnosis of mycotoxicosis or
biological activity and pathological manifestations of mycotoxin the specific mycotoxicosis is often difficult. Diagnosis is typically
intoxication will also differ greatly. In fact, some mycotoxins are achieved by evaluation of clinical signs, and under certain
extremely toxic to poultry; whereas, others are literally non-toxic to circumstances even hematological evaluation (38). Furthermore,
all classes of poultry. collection of suspect feed or grain followed by chemical analysis of
Ingestion of a mycotoxin at a concentration sufficient to cause an the sample for specific mycotoxin contamination is most helpful in
alteration of structure or disruption of function of poultry will result achieving a diagnosis.
in disease. This disease is classified as a toxicity, specifically a The following is a brief summary of the major clinical signs of
mycotoxicosis. Mycotoxicoses are usually of an acute nature. This specific mycotoxicoses in poultry. The specific mycotoxicoses
is due to the sporadic occurrence of mycotoxins in feedstuffs and discussed herein are generally considered to be the most prevalent
the management techniques used in the feeding of poultry flocks. in poultry. However, due to the number of mycotoxins known to
Three genera of molds, Fusarium, Aspergillus, and Penicillium, are contaminate poultry feedstuffs, not all potential mycotoxicoses are
typically associated with most mycotoxin formation in poultry mentioned.
feedstuffs. For example, various species of Fusarium produce
numerous mycotoxins that share similar chemical structures. This Aflatoxicosis
class of mycotoxins, known as trichothecenes, are responsible for The major four naturally occurring forms of aflatoxin apt to be
numerous disease outbreaks in poultry. Aspergillus parasiticus and found in feedstuffs are designated as aflatoxin Bb B2, Gb and G2.
A. flavus are the two producers of aflatoxin, perhaps the most toxic Of these, aflatoxin Bj is the most toxic to poultry and is usually
of all mycotoxins to poultry. Penicillium viridicatum and produced in the highest concentration. Aspergillus flavus and
80
Chapter 18 Mycoses and Mycotoxi coses

A. parasiticus are recognized as the molds responsible for the genus Fusarium. Although their chemical structures are similar,
production of aflatoxin in nature. In most feedstuffs, Aspergillus their biological activities are slightly different. In general terms,
flavus tends to produce primarily aflatoxin Bj; whereas, A. their mechanisms of action involve an impact on dermal tissue and
parasiticus tends to produce a mixture of both aflatoxin B] and Gb systemic effects that target lymphatic tissue (33).
A mycotoxicosis will occur in poultry when birds are fed a ration Much of the research with trichothecenes has been conducted with
containing a toxic concentration of the mycotoxin for a period of T-2 toxin, one of the 12, 13, epoxytrichothecenes. This was done
time sufficient for the mycotoxin to impact the function or structure based upon the ease of bioproduction of T-2 toxin in the laboratory
of susceptible tissues. When poultry are fed a ration containing for use in experimentation. It is now recognized that T-2 toxin may
aflatoxins affected birds will appear listless, have rough feathers, not be the primary trichothecene produced in nature or the most
display marked depigmentation of the skin and shanks and may die common trichothecene contaminant of poultry feeds.
from acute toxicosis or from the toxicosis complicated with an Diacetoxyscirpenol (DAS) and deoxynivalenol (DON) are most
infection of viral, bacterial or fungal etiology due to likely more probable as poultry feed contaminants. The primary
immunosuppression associated with aflatoxicosis (6). The basic clinical manifestation of trichothecene toxicosis is oral necrosis
mechanism of action of aflatoxin at the cellular level is to bind presumably resulting by direct contact of contaminated feed with
covalently with DNA with a subsequent inhibition of protein the mucous membranes of the oral cavity and esophagus (42). This
synthesis or disruption of normal regulatory processes. Failure of oral necrosis will clinically manifest itself as small white to cream­
affected birds to produce digestive enzymes is noted during colored necrotic areas on the inside of the upper and lower
aflatoxicosis (19). This results in incomplete digestion of feed, mandible and the borders of the tongue. With some trichothecenes,
passage of undigested feed in the feces, and poor growth and these necrotic areas may involve the esophagus as well. Associated
performance. Birds being processed for meat may show reddening with oral necrosis is a marked decrease in feed intake. Decreased
of the wing tips, reddening of the skin covering the thigh, legs, and feed intake leads to poor growth in broilers and extremely poor
breast, and petechial hemorrhages of the leg, thigh and breast feathering. In commercial layers, decreased feed intake and thin egg
muscle. This response is attributable to capillary fragility resulting shells is commonly observed (38). Necrosis of dermal tissue has
from induction of lysosomal enzymes by aflatoxin (32). The gall been observed in broiler grown on litter contaminated with
bladder will typically be enlarged and elongated and filled with bile trichothecenes and manifests as dermatitis of skin and shanks.
with a pale green color (28). Inhibition of protein synthesis at the ribosomal level has been
The liver is the organ that will respond first and to the greatest associated with trichothecene toxicosis resulting in
degree. Upon necropsy, the liver can be described as enlarged, immunosuppression (33). Among the.systemic effects of T-2 toxin
edematous, fatty, and will tend to have a yellowish discoloration in broiler chickens is an impact on neurological tissue. Hysteroid
due to the accumulation of lipoidal components (28). Occasionally, seizures, impaired righting reflex, and an abnormal wing posture
the liver will show focal hemorrhages with necrotic foci. The spleen following handling of affected birds is observed in birds
will typically be enlarged and may exhibit a mottled appearance. experiencing moderate to severe T-2 toxicosis (38).
Mild kidney enlargement and thymus gland atrophy are also Upon necropsy, oral lesions ranging from a few small areas of
typically observed during aflatoxicosis. Histological examination of necrosis to numerous areas of larger areas of necrosis will be
liver tissue will reveal severe fatty infiltration, edema, bile ductule observed. The higher number of lesions of increased severity in the
hyperplasia, necrosis, hemorrhage, necrosis, and fibrosis (20). oral cavity is usually associated with higher dietary concentrations
Susceptibility of poultry to aflatoxicosis is dependent upon the of the trichothecene or exposure to contaminated feed for an
presence of interacting factors, age of the birds, and species. For extended period of time.
example, breed, strain, and gender of poultry will influence their
susceptibility of aflatoxin poisoning (41). Additionally, the Ochratoxicosis
environmental temperature and the nutritional status of birds are Ochratoxin is a term typically used to refer to either ochratoxin A
also known to influence the susceptibility of poultry to or B, or a mixture of the two. Ochratoxin A is the more toxic to
aflatoxicosis. Young broilers are more susceptible to aflatoxin than poultry. Aspergillus ochraceus and Penicillium viridicatum are
older broilers. In broiler breeder hens exposed to aflatoxin, the hen considered as the primary producers of ochratoxin in nature. The
does not respond to aflatoxin in a significant manner, however the target organs for ochratoxin are primarily the kidney and to a lesser
transfer of minute concentrations of aflatoxin residues to hatching extent the Ever. The primary clinical manifestations of ochratoxin
eggs will result in high embryonic mortality (31). Ducklings, are extreme morbidity, increased mortality, poor growth rate,
turkeys, and chickens respond in similar fashion to aflatoxicosis, impaired feed efficiency, pasty vents, and diarrhea (8, 15).
however, ducklings are the most susceptible, followed by turkey At necropsy, severely enlarged and pale kidneys are the most
poults, and finally chickens (1). notable signs with the liver being of normal size but tan-colored.
Caution should be exercised when attempting to relate dietary The kidney tubules will be filled with urates but visceral or articular
concentrations of aflatoxin and other mycotoxins to various gout is usually not present. Histological examination of renal tissue
physiological and pathological responses. Laboratory typically shows enlarged and necrotic proximal tubular epithelium
experimentation is designed to establish a cause-and-effect (8). Histological examination of the liver will reveal liver
relationship between dietary aflatoxin and specific responses in congestion, focal necrosis, and an unusually high glycogen content
poultry. In general terms, the dietary concentration required to elicit (14). High glycogen concentrations in liver is a specific response
a specific response in laboratory studies is much higher than the and is of diagnostic value for ochratoxicosis.
dietary concentration required elicit the same response under
conditions of commercial poultry production. The underlying Analysis for Mycotoxins
rationale for this is the presence of various interacting factors and Tissue analysis for mycotoxins is most always futile. The
circumstances in the “field situation” that are usually absent in the underlying reason for this is due to the fact that mycotoxin
“laboratory environment” (11). concentrations at the time the tissue sample is obtained will be
extremely low or non-existent. Once most mycotoxins are
Trichothecene Toxicosis consumed, the mycotoxin or its metabolites and adducts are rapidly
The trichothecene mycotoxins are a group similar chemical excreted from the bird and no residues will persist.
compounds, also referred to as the 12, 13, epoxytrichothecenes. The The most promising approach to identify a mycotoxin responsible
trichothecenes are produced in nature by primarily members of the for the disease outbreak is a chemical analysis of the feed being
81
R D. Wyatt

consumed by affected birds. This, however, is not without 9. Dixon, D. M and A. Polak-Wyss. The medically important dematiaceous
difficulties. In many situations, the feed responsible for the disease fungi and their identification. Mycoses. 34:1-18. 1991.
outbreak may have been consumed and a new feed delivered made 10. Georg, L. K., B. W. Biere, and W. B. Cooke. Encephalitis in turkey
poults due to a new fungus species. Sabouraudia 3:239-244. 1964.
to the affected flock. Analysis of the feed in this situation will most
11. Hamilton, P. B. Determining safe levels of mycotoxins. J. Food Protect.
likely reveal no detectable mycotoxin contamination. Since 47:570-575. 1984.
mycotoxin formation does not occur uniformly within a container of 12. Hubbard, G. B., R. E. Schmidt, D. L. Eisenbrandt, W. M. Witt, and K.
feed or grain, sampling is a major problem in attempting to obtain a C. Fletcher. Fungal infection of ventriculi in captive birds. J. Wildlife Dis.
sample indicative of the true field situation. On the other hand, if a 21:25-28. 1985.
feed sample can be obtained (from the farm or as a “retained” 13. Huff, W.E. and J.A. Doerr. Synergism between aflatoxin and ochratoxin
sample) that was fed to the birds, it is important to stabilize the feed in broiler chickens. Poultry Sci. 60:550-555. 1981.
to prevent additional mycotoxin production and to halt all microbial 14. Huff, W. E., J. A. Doerr, and P. B. Hamilton. Decreased glycogen
mobilization during ochratoxicosis in broiler chickens. Appl. Microbiol.
activity so the contaminating mycotoxin will not be degraded. The
37:122-124. 1979.
goal of any sample is that it accurately reflects the whole from 15. Huff, W. E., R. D. Wyatt, T. L. Tucker, and P. B. Hamilton.
whence it was obtained. Feed samples should be stored in a paper Ochratoxicosis in the broiler chicken. Poult. Sci. 53:1585-1590. 1974.
container, frozen, and then analyzed as soon as possible. 16. Lodder, J. The Yeasts, 2nd ed. North-Holland Publishing Co.
The decision of which mycotoxin to analyze for in a feed sample Amsterdam, The Netherlands. 1970.
should be based upon the production traits, clinical signs, and 17. Manning, R. O. and R. D. Wyatt. Effect of cold acclimation on the
histological examination of birds from the affected flock. Since broiler chick’s resistance to acute aflatoxicosis. Poultry Sci. 69:388-396.
several mycotoxins could be involved in an outbreak of a toxicosis, 1990.
18. Okoye, J. O. A., C. N. Okeke, and F. K. O. Ezeobele. Effect of
and since the analysis for each mycotoxin will have inherent
infectious bursal disease virus infection on the severity of Aspergillus flavus
specifics, it is imperative that all information surrounding the aspergillosis of chickens. Avian Pathol. 20: 167-171. 1991.
affected flock be furnished to the laboratory in order to narrow the 19. Osborne, D. J. and P. B. Hamilton. Decreased pancreatic digestive
list of mycotoxin possibilities. enzymes during aflatoxicosis. Poultry Sci. 60:1818-1820. 1981.
The specific method of analysis could involve chemical 20. Quist, C. F., T. Cornish, and R. D. Wyatt. Mycotoxicosis. In: Infectious
absorbency, thin layer chromatography (TLC), high pressure liquid Diseases of Wildbirds. N. J. Thomas, R. Hunter, and C. T. Atkinson, eds.
chromatography (HPLC), or gas chromatography-mass spectral Blackwell Publishing, Oxford, UK. pp. 417-430. 2007.
(GC-MS) methodology. Specific techniques and methods are will 21. Ranck. F.M, L.K. Georg, and D. H. Wallace. Dactylariosis, a newly
recognized fungus disease of chickens. Avian Qis. 18:4-20. 1974.
recognized that will permit an analysis for numerous mycotoxins.
22. Redig, P. T. Aspergillosis in avians, an update. In: Proceedings of the
However, these methodologies are time-consuming, costly, and 19th annual conference on avian medicine and surgery. Mid-Atlantic States
often require specialized equipment and training of laboratory Association of Avian Veterinarians. Lancaster, PA. pp 172-177. 1998.
personnel. These techniques are most appropriate for research 23. Richard, J. L., S. J. Cysewski, A. C. Pier, and G. D. Booth. Comparison
laboratories rather than diagnostic facilities. of effects of dietary T-2 toxin on growth, immunogenic organs, antibody
Exploitation of enzyme-linked immunosorbent assay (ELISA) formation and pathologic changes in turkeys and chickens. Am. J. Vet. Res.
technology has resulted in the development of testing procedures 39:1674-1679. 1978.
for mycotoxins in a variety of commodities. These tests can yield 24. Rippon, J. W., R. Gerhold, and M Heath. Thermophilic and
thermotolerant fungi isolated from the thermal effluent of nuclear power
mycotoxin concentrations with ppb accuracy or can be modified to
generating reactors: dispersal of human opportunistic and veterinary
give a less expensive qualitative result. These tests have proven to pathogenic fungi. Mycopathologia 70:169-179. 1980.
be not only highly accurate and specific but also free from most 25. Salkin, I. F., D. M Dixon, Μ E. Kemna, P. J. Danneman, and J. W.
interfering influences. Among the sources for various testing Grifith. Fatal encephalitis caused by Dactylaria constricta var. gallopava in
supplies are Charm Sciences, Inc. Lawrence, MA; EnviroLogix, a snowy owl chick (Nyctea scandiaca). 28:2845-2847. 1990.
Inc. Portland, ME; Neogen, Corp., Lansing, MI; Romer Labs, Inc. 26. Shane, S. M, J. Markovits, T. G. Snider ΠΙ, and K. S. Harrington.
Union, MO; Strategic Diagnostics Inc. Newark, DE; VICAM, Encephalitis attributed to dactylariosis in Japanese quail chicks (Coturnix
Watertown, MA; and R-Biopharm Rhone, Ltd. Glasgow, Scotland. coturnix japonica). Avian Dis. 29:822-828. 1985.
27. Singh, K., J. Hood, R D. Welsh, J. H. Wyckoff ΠΙ, T. A Snider and D.
REFERENCES A. Sutton. Fatal systemic Phaeohyphomycosis caused by Ochroconis
gallopavum in a dog (Canis familaris}. Vet. Pathol. 43:988-992. 2006.
1. Asplin, F. D. and R B. A. Camaghan. The toxicity of certain groundnut 28. Smith, J. W. and P. B. Hamilton Aflatoxicosis in the broiler chicken.
meals for poultry with special reference to their effect on ducklings and Poultry Sci. 49:207-215. 1970.
chickens. Vet. Rec. 73:1215-1219. 1961. 29. Tansey, M R. and T. D. Brock. Dactylaria gallopava, a cause of avian
2. Benke, E. S. and A. L. Rogers. Medical mycology manual with human encephalitis, in hot spring effluents, thermal soils and self-heated coal waste
mycoses monograph, 4th ed Burgess Publishing Company, Minneapolis, piles. Nature 242:202-203. 1979.
MN. 1980. 30. Thaxton, J. P., Η. T. Tung, and P. B. Hamilton. Immunosuppression in
3. Blalock, H. G., L. K. Georg, and W. T. Derieux. Encephalitis in turkey chickens by aflatoxin. Poultry Sci. 53:721-723. 1974.
poults due to Dactylaria (Diplohinotrichum) gallopava-a. case report and its 31. Trucksess, N. W., L. Stoloff, K. Young, R D. Wyatt, and B.L. Miller.
experimental reproduction. Avian Dis. 17:197-204. 1973. Aflatoxicol and aflatoxins B] and Mi in eggs and tissues of laying hens
4. Chandler, F. W., W. Kaplan, and L. Ajello. Color Atlas and Text of the consuming aflatoxin-contaminated feed. Poult. Sci. 62:2176-2180. 1983.
Histopathology of Mycotic Diseases. Year Book Medical Publishers, 32. Tung, Η. T., J. W. Smith, and P. B. Hamilton Aflatoxin and bruising in
Chicago, 1980. the chicken. Poult. Sci. 50:795-798. 1971.
5. Chandler, F. W. and J. C. Watts. Histologic and immunohistologic 33. Ueno, Y. Trichothecene mycotoxins: mycology, chemistry and
diagnosis of mycotic diseases. International Academy of Pathology, Short toxicology. Advances in Nutrition Research 3:301-305. 1980.
Course No. 12, Atlanta, GA. 1983. 34. Veltmann, J. R, R. D. Wyatt and Μ N. Voigt. The effect of varying the
6. Chang, C. F. and P. B. Hamilton. Impaired phagocytosis by heterophils total sulfur amino acid content in the diets of chicks with aflatoxicosis.
from chickens during aflatoxicosis. Toxicol. Appl. Pharmacol. 48:459-462. Poultry Sci. 60:1748.
1979. 35. Voigt, Μ N., R. D. Wyatt, J. C. Ayres, and P. E. Koehler. Abnormal
7. Converse, K. A. Aspergillosis. In: Infectious Diseases of Wildbirds. N. J. concentrations of B vitamins and amino acids in plasma, bile and liver of
Thomas, R. Hunter, and C. T. Atkinson, eds. Blackwell Publishing, Oxford, chicks with aflatoxicosis. Appl. Environ. Microbiol. 40:870-874. 1980.
UK. pp. 360-374. 2007. 36. Waldrip, D. W., A A. Padhye, L. Ajello and M Ajello. Isolation of
8. Dwiedi, P and R. B. Bums. The natural occurrence of ochratoxin A and Dactylaria gallopava from broiler-house litter. Avian Dis. 18:445-451.
its effect in poultry. A review. Part 1. Epidemiology and toxicity. World’s 1974.
Poultry Science Journal. 42:32-47. 1986.
82
Chapter 18 Mycoses and Mycotoxicoses

37. Wyatt, R.D. Mycotoxin Interactions. In: The Mycotoxin Blue Book. D. 40. Wyatt, R. D., and P. B. Hamilton. Candida species and crop mycosis in
Diaz, ed. Nottingham University Press. Nottingham, UK. pp 269-278. 2005. broiler chickens. Poult. Sci. 54:1663-1666. 1975.
38. Wyatt, R.D. Poultry. In: Mycotoxins and Animal Foods. J. E. Smith and 41. Wyatt, R. D., H. L. Marks, and R. O. Manning. Selection for resistance
R. S. Henderson, eds. CRC Press, Boca Raton, FL. pp. 553-605. to aflatoxin in chickens. Poultry Sci. 66:1901-1904. 1987.
39. Wyatt, R. D., J. A. Doerr, P. B Hamilton, and H. R. Burmeister. Egg 42. Wyatt, R. D., B. A. Weeks, P. B. Hamilton, and H. R. Burmeister.
production, shell thickness and other physiological parameters of laying Severe oral lesions in chickens caused by ingestion of dietary fusariotoxin
hens affected by T-2 toxin. Appl. Microbiol. 29:641-645. 1975. T-2. Appl. Microbiol. 24:251-254. 1972.

83
19
ADENOVIRUSES
Brian M. Adair and J. Brian McFerran

SUMMARY. Avian adenoviruses are divided into three groups, which correspond to general to the Adenovirus family. Group 1, or
conventional adenoviruses (Genus Aviadenovirus), all share a common group antigen, distinct from the mammalian group antigen. The
group 1 viruses are subdivided into 5 species (A to E) on the basis of molecular criteria and at least 12 serotypes. The group 2 adenoviruses
(Genus Siadenovirus) are dealt with in Chapter 20. The third group (Genus Atadenovirus) is represented by a duck virus that partially shares
an antigen with the group 1 viruses and causes egg drop syndrome (EDS) in chickens.
Group 1 adenoviruses are spread both vertically and horizontally, mainly by the fecal-oral route, and are ubiquitous in avian species. The
group 2 (EDS) virus replicates in the uterus of hens and is transmitted either in or on the egg and also in droppings contaminated by
secretions from the oviduct. Chickens can also be infected by direct or indirect contact with contaminated feces from waterfowl.
Because so many healthy birds are infected with group 1 adenoviruses, it is difficult to determine their adenoviruses as pathogens, however
strong evidence exists that some strains can cause inclusion body hepatitis (IBH) and pancreatitis in birds. Others are associated with
hydropericardium syndrome (HPS) or Angarra disease. Infection of laying hens with the group 3 (EDS) virus results in a severe decrease in
egg production, mainly through the production of soft-shelled and shell-less eggs.
Agent identification. Virus isolation is the first stage in adenovirus characterization and subtyping and is best carried out in epithelial cell
cultures such as embryo liver cells. Detection of virus in cell cultures or tissues from diseased birds is normally carried out using
immunostaining (immunofluorescence or immunoperoxidase) techniques, or by molecular methods. Because of the well defined and easily
recognized structure of the adenovirus particle, negative stain and thin section electron microscopy is useful and widely used to confirm
presence of the virus. Subtyping is achieved by molecular methods or by cross neutralization tests with prototype strains.
Serologic Detection in the Host. Serologic tests are of little value except to monitor specific-pathogen-free flocks, where a sensitive test
such as the enzyme-linked immunosorbent assay should be used. The hemagglutination inhibition test is recommended for EDS virus
serology. Latently infected chicks only develop antibody to EDS virus after they produce affected eggs.

INTRODUCTION chickens have generally resulted in mild respiratory signs or none at


all.
Avian adenoviruses can be divided into three subgroups, which are Several serotypes of group 1 adenoviruses have been directly
now separated into 3 distinct Genera of the Adenovirus Family (6). associated with inclusion body hepatitis (IBH) in chickens, turkeys,
The first group (Genus Aviadenovirus) includes the conventional parrots, kestrels, merlins, and pigeons. In chickens, the disease is
avian adenoviruses, which are often referred to in the literature as usually seen in meat producing birds, between 3 and 7 wk of age
group 1 adenoviruses. The second group (Genus Siadenovirus), but has also been recorded in birds as young as 7 days and as old as
consists of the viruses of hemorrhagic enteritis of turkeys and 20 wk. The disease has a sudden onset of mortality, peaking after 3-
marble spleen disease of pheasants, along with the virus causing 4 days but may continue for 2-3 wk. Mortality normally ranges
avian adenovirus splenomegaly (AAS) of chickens. This group is from 5-10%, but can reach 30%; pale swollen livers with
often referred to in the literature as group 2 avian adenoviruses, hemorrhages are observed and there are also numerous eosinophilic
and they are dealt with in Chapter 20. The third group (Genus inclusion bodies present in hepatocytes. Electron microscopy shows
Atadenovirus), contains the viruses isolated from ducks and the presence of large numbers of adenovirus particles in the
chickens, which cause Egg Drop Syndrome (EDS), and are referred inclusion bodies. Many different serotypes of adenovirus have been
to as group 3 avian adenoviruses. The EDS adenovirus has not been associated with IBH. Several outbreaks of disease in Australia, with
identified in chickens in the United States, and if evidence of mortality up to 30%, were associated with 3 serotypes all belonging
infection is detected, animal health officials must be notified. to FAdV species E (Table 19-1), although FAdV8 was the most
common isolate (9).
CLINICAL DISEASE FAdV4 viruses have been implicated in outbreaks of
hydropericardium syndrome (HPS), also known as Angarra disease,
Group 1 Adenoviruses which has caused severe economic losses to the poultry industry in
The group 1 adenoviruses are widely distributed in avian species Pakistan. HPS has also been recorded in India, Kuwait, Iraq, Japan,
as indicated by serological surveys and virological studies, and have and parts of Eastern Europe, as well as South and Central America.
been isolated from both sick and healthy birds. In addition to The disease is characterized by accumulation of fluid in the
chickens and turkeys, they have been isolated from geese, ducks, pericardium with lesions in heart, kidneys and lungs, and presence
pigeons, and budgerigars, and there is reported evidence of of basophilic intra-nuclear inclusion bodies in hepatocytes, along
infection in other species, including gulls, owls, and hawks (13). with multiple areas of focal necrosis and mononuclear cell
They have been isolated from birds with signs of respiratory infiltration (14,16).
disease, depressed egg production, arthritis/ tenosynovitis, uneven
growth, and enteritis, but, in most cases, causal relationships have Group 3 (EDS) viruses
been difficult to demonstrate. Infection of healthy birds is common, The viruses associated with EDS are widespread in waterfowl and
and most adults have been infected by many serotypes. They have in domestic ducks and geese. Three forms of EDS have been
also been isolated from commercial poultry flocks even when recognized. The first, or classical, form infects breeding stock and is
exceptionally high levels of production and fertility are in evidence. vertically transmitted. The virus remains latent until birds come into
Apart from quail bronchitis virus (QBV), caused by an FAdV-1 lay, when it becomes activated, causing classical EDS. The second,
strain (Table 19-1), the association with respiratory disease has not or endemic, form arises from the classic form. Virus spreads into
been proven, although adenoviruses have frequently been isolated commercial egg layers and is often spread by eggs through common
from the respiratory tract, often in association with severe packing stations. The third form is the sporadic form, which infects
respiratory disease. Despite this association, attempts to reproduce flocks via drinking water contaminated by wild or domestic
respiratory disease by experimental inoculation of isolates into waterfowl. This form can give rise to the classical form (if great
grandparents or grandparents were infected) or to the endemic form.
84
Chapter 19 Adenovirus

SAMPLE COLLECTION sac inoculation (13). However, the FAdV-1 and FAdV-5 viruses,
and in particular the FadV-4 strains, associated with HPS (Angara
Group 1 Adenoviruses Disease), appear to grow well in embryonated eggs. FadV-4 strains
The main sites of replication of group 1 adenoviruses are the from HPS cases have been isolated in eggs and cause high levels of
digestive tract and the upper respiratory system. Samples for virus embryo mortality with severe lesions (14).
isolation should therefore include feces or large intestine with feces, The group 3 (EDS) viruses produce high titers of hemagglutinin
and also an aliquot from the affected organ(s), e.g. liver in cases of when inoculated into the allantoic cavity of embryonated duck or
IBH. Virus is also often present in the pharynx and kidney. Ten per geese eggs, but do not produce hemagglutinin or cause embryo
cent suspensions of tissue or feces in cell culture medium, death in embryonated chicken eggs.
homogenized or shaken with glass beads, then frozen and thawed,
provides a convenient method for preparing inocula. Details are AGENT IDENTIFICATION
given in Chapter 43 (Cell culture Methods). Following
centrifugation, the supemates may be stored at -80 C until required On recognition of cytopathic effect, several alternatives are
for inoculation of cell cultures. available to confirm presence of adenovirus.

Group 3 (EDS) viruses Antigen detection


The main site of EDS virus replication is the pouch shell gland Confirmation of adenovirus presence in cell culture can be made by
area of the uterus, although selection of an infected bird can be fluorescent antibody staining of infected cells using adenovirus­
difficult because of the lack of clinical signs and the short duration specific antisera (1,3). This is best done on cell cultures grown for
that virus is present. Consequently, the usual method of detecting an this purpose on sterile glass coverslips (e.g. 13 mm diameter) in
infected flock is by serology. Isolation of the virus can be facilitated Petri dishes, or in 24-well cell culture plates or rectangular
by collecting thin shelled and soft shelled eggs from affected birds, coverslips in tubes. Chick embryo liver cells or chick kidney cells
homogenizing and using this as oral inoculum by feeding to normal are allowed to monolayer on the coverslips, and the supemate is
laying hens, which have no EDS virus antibodies. At the first signs removed and replaced with the specimen containing virus.
of abnormal eggs being produced by the hens (i.e. loss of shell Coverslips are removed with sterile forceps at 24 and 48 hr post
color, thin shells, or no shells), the hens are sacrificed and the pouch
Table 19.1 Classification of Group 1 avian adenoviruses
*
shell gland removed for either immunohistochemical staining or
virus isolation. A 10% suspension of the shell gland is prepared as Group 1 avian adenoviruses (Genus Aviadenovirus)
above. If the birds are kept in cages, spread can be slow. Birds or
eggs for virus isolation should be selected from cages where Group 1 Recognized serotypes within each species and examples
aviadenovirus species of prototype strains
affected eggs are being laid and from adjoining cages where normal
eggs are present. Blood samples for serology should be taken from
the first birds to show signs of EDS i.e laying abnormal eggs. Fowl adenovirus A Fowl adenovirus 1 (FAdV-1) (CELO, 112, QBV, Ote,
Hl)
PREFERRED CULTURE MEDIA AND SUBSTRATES
Fowl adenovirus B Fowl adenovirus 5 (FAdV-5) (340,TR-22, Tipton, M2)
Isolation of adenovirus in cell culture is an essential prerequisite
for full characterization and typing, and for further studies on
disease association. In general, cell systems from the homologous
species should be used. Therefore, although fowl adenoviruses have Fowl adenovirus C Fowl adenovirus 4 (FAdV-4) (506, J2, KR5, H2, K31,
been isolated from turkeys using chicken cells, isolation of turkey 61)
adenovirus strains seem to require turkey cell cultures. Embryonic
Fowl adenovirus 10 (FAdV-10) (C-2B, Mil, CFA20,
fiver or kidney cells, and kidney cells from 1-4 wk old birds are the SA2)
cultures of choice. If chick embryo cells are used, they should have
a predominance of epithelial cells for optimal sensitivity. Chick
embryo fibroblasts or tracheal organ cultures are less sensitive for Fowl adenovirus D Fowl adenovirus 2 (FAdV-2) (GAL-1, 685, SR-48, H3,
aviadenovirus primary isolation. P7)
Because of the problem of latency, the cell cultures must be
prepared from embryos or chicks derived from a flock known to be Fowl adenovirus 3 (FAdV-3) (SR-49, 75, H5)
free of adenovirus. Uninoculated control cultures should be
included, and these should be incubated for approximately 7 days, Fowl adenovirus 9 (FAdV-9) (A2, 90, CFA19)
disrupted by freezing and thawing, or by sonication, and the
Fowl adenovirus 11 (FAdV-11) (380, UF71)
supernatants passed onto fresh cell cultures. Two passes in this
manner should be sufficient to exclude the possibility of latent Fowl adenovirus E Fowl adenovirus 6 (FAdV-6) (CR119,168)
adenovirus being present in the uninoculated cells.
The group 3 (EDS) viruses are best isolated in duck cell cultures, Fowl adenovirus 7 (FAdV-7) (YR36, X-l 1,122)
although chick embryonic liver cells or, to a lesser degree, chicken
kidney cells, can be used instead. Isolation in chick cultures may Fowl adenovirus 8a (FAdV-8a) (58, TR-59, T-8,
require up to five passages before CPE becomes apparent. CFA40)
Supernatants of all cultures should be checked for
Fowl adenovirus 8b (FAdV-8b) (764, B3, VRI-33)
hemagglutination, using chick erythrocytes. Although the virus can
be difficult to isolate, cross-contamination with EDS viruses occurs
readily. Therefore, controls must be passaged in parallel with the
specimens. *Based on Benko et al, (6)
Embryonated eggs are not normally a sensitive medium for
primary isolation of group 1 adenoviruses, although all the inoculation, fixed by immersion in acetone (5 min), air dried, then
prototype fowl adenoviruses have been propagated in eggs by yolk stained by addition of adenovirus antiserum using either direct or
85
Brian M. Adair and J. Brian McFerran

indirect immunofluorescence techniques. In direct Nucleic acid detection


immunofluorescence, infected coverslip cultures are covered by the Presence of adenovirus in tissues or lesions from diseased birds is
conjugated antiserum (conjugated normally with fluorescein often first suspected by demonstration of intranuclear inclusion
isothiocyanate, FITC) known to contain adenovirus group-specific bodies in histopathology studies, although adenoviruses are not the
antibodies at a pre-determined dilution. After incubation at 37 C for only agents which give rise to intranuclear inclusions following
30 min, the coverslips are washed in several changes of PBS, infection. Other viruses e.g. herpesviruses, circoviruses or polyoma
blotted dry, mounted in buffered glycerol, and examined for viruses may give rise to similar inclusions.
presence of fluorescent intranuclear inclusions using a fluorescent In situ hybridization (ISH) has been used for detection of
microscope (1,3). In indirect immunofluorescence, the first serum adenovirus DNA in tissues in these circumstances and can be useful
used for staining is unconjugated and this first phase is followed by in species other than turkeys or chickens where cell cultures are not
secondary staining using an anti-species FITC conjugate (e.g. anti­ available for virus isolation, or where formalin-fixed tissues are the
chicken FITC). Following incubation and washing, the coverslips only specimen submitted to the laboratory for diagnosis.
are mounted and viewed as above (3,4). Adenovirus ISH probes (36-40 bases) based on a highly conserved
An antigen detecting ELISA has also been used for demonstration region of the penton of FAdVIO have been described and used in
of adenovirus antigen in tissues from diseased birds (18). In this ISH (12). This region was chosen since it is highly conserved and
test, a rabbit antiserum against an FAdV8 strain of group I therefore provides maximum cross reactivity between adenovirus
adenovirus was used to capture virus present in tissues or in cell species. The procedure requires the sections to be de-paraffinized
cultures. Ninety six-well ELISA plates are coated with the capture by treatment with xylene, rehydrated by passage through graded
antibody, washed, and the test samples (10% tissue suspension, see alcohols, digested with pepsin, before hybridization to the
above) are added. After incubation for 90 min at 37 C and washing, digoxigen labeled probe. An anti-digoxygenin antibody conjugated
a chicken antiserum to group 1 adenovirus is added and following to alkaline phosphatase was then applied, and the chromogen,
incubation and washing, an anti-chicken serum conjugated with nitroblue tetrazolium (NBT) added. Foci of dye reduction to blue­
horseradish peroxidase is added. The test was shown to be capable black formazan pigment indicated presence of adenovirus nucleic
of detecting less than 100 TCID50 of virus per gram of tissue and acid, which duplicated the pattern of intranuclear inclusions seen in
was capable of detecting any of the 12 recognized adenovirus parallel H&E stained sections. ISH was successfully used to
serotypes (i.e. it was group specific). demonstrate hepatic, renal and intestinal adenovirus infection in
Gel diffusion is relatively insensitive but may be useful to confirm parrots and to exclude herpesvirus and circovirus from the
that an isolate made in cell culture is an adenovirus, if no other diagnosis. The same probes were used ‘in an investigation of
methods are available. Once an isolate has been made in cell inclusion body hepatitis and pancreatitis in broiler chickens, and
culture, a large batch of antigen must be prepared e.g. 12 x 75 cm2 positive adenovirus ISH results were confirmed by thin section
flasks of chick embryo liver or kidney. Following development of electron microscopy and virus isolation (10).
>80% CPE, the cells are disrupted by freezing and thawing, the cell
debris removed by centrifugation, and the supemates, containing Morphology
virus antigens, concentrated by precipitation with ammonium On development of CPE, negative stain electron microscopy is
sulphate overnight or by extraction with arcton. A control used to confirm presence of adenovirus. Simple techniques for
adenovirus antiserum and antigen is required and common lines of diagnostic use of the electron microscope are described in Chapter
precipitation in agar, between the control and test antigen confirms 34 on enteric viruses. Examination of negatively stained material by
presence of adenovirus. electron microscopy is fast and also allows both positive
Growth of group 3 (EDS) virus in cell culture is accompanied by identification of the virus and recognition of other unsuspected
production of a hemagglutinin which agglutinates avian viruses or bacteria. Adenoviruses are normally easily recognized by
erythrocytes but not mammalian erythrocytes. Therefore, while their morphology. They are seen as isosahedral particles (i.e.
fluorescence or other antigen detecting techniques can be applied to particles with 12 vertices and 20 triangular faces) measuring 65-75
detection of EDS virus, hemagglutination presents an easier nm in diameter. Degenerating particles, however, can develop a
alternative. This is carried out by removing some supernatant from circular appearance. Adeno-associated parvoviruses are frequently
the EDS virus-infected culture, following development of CPE, and seen, along with the adenoviruses. These are spherical particles
addition of an equal volume of chicken or duck erythrocytes (0.8% measuring approximately 22 + 3 nm in diameter. If desired,
suspension in PBS). Presence of EDS hemagglutinin results in sensitivity can be improved by use of immunoelectron microscopy,
hemagglutination of the erythrocytes. Hemagglutination-inhibition in which adenovirus antiserum or convalescent serum is mixed with
(HI) (see below) using EDS virus-specific antiserum confirms the the specimen at an appropriate dilution to facilitate clumping of
isolate as EDS virus. If immunofluorescence is to be used for particles, making viewing easier.
detection of EDS virus antigen in infected cultures or tissues, Thin-section electron microscopy has also proved useful for
antisera with specificity for group I adenoviruses cannot be used, demonstration of adenoviruses in cell cultures or to confirm
because of lack of cross reactivity. Specific EDS antisera are presence of adenovirus in tissues or organs from birds in which
required for this purpose. EDS specific antisera have been used for intranuclear inclusions are seen with high frequency in
direct immunostaining of tissue sections to detect EDS antigens. histopathology studies. Cell cultures showing CPE are scraped from
Using the avidin-biotin-peroxidase technique, EDS antigen was the plastic surface, fixed for 2 hr at 4 C in glutaraldehyde, post fixed
detected in tissue sections of infundibulum for 3-5 days post in osmium tetroxide for 2 hr at room temperature, then dehydrated
infection and in large amounts in the pouch shell gland from about 8 in alcohol and embedded in Araldite. Ultrathin sections of infected
days post inoculation, until as long as affected eggs were produced cells stained with uranyl acetate and lead citrate show large
(19). numbers of characteristic particles, 65-75 nm in diameter, often in
crystalline arrays, along with associated inclusion material in the
nucleus (5). Small pieces of tissue shown by histopathology to
contain large numbers of inclusions can also be treated in this way,
and often show similar large concentrations of virus particles. This
can be useful in investigations in species other than chicken and
turkey where virus isolation in cell culture is more difficult, or not

86
Chapter 19 Adenovirus

an option, but is not indicative of a causal relationship to the virus stock is mixed with buffer at pH 3 and pH 7. After incubating
condition. for 1 hour at 37 C, the virus suspensions are immediately titrated.
Adenoviruses should be stable to pH 3 treatment, and show no
Subtyping of Isolates substantial reduction in infectivity. Adenoviruses in cell-culture
The hexon protein of the group 1 avian adenoviruses contains a medium, without serum, are generally inactivated by heating for 30
highly conserved region which gives rise to antibodies which are min at 60 C. At 50 C for 1 hr, 1 M MgCl2 enhances the thermal
‘group specific’ i.e. the antibodies produced following infection inactivation, and this is the basis of the test for stability in the
with one group 1 adenovirus also react with other group 1 viruses in presence of divalent cations.
tests such as gel diffusion and immunofluorescence. Other
antibodies are produced following infection, which react only with SEROLOGIC DETECTION IN THE HOST
the infecting virus and closely related viruses. These type specific
antibodies neutralize virus infectivity and are used to separate group Group specific antibodies
1 viruses into serotypes, using the serum neutralization test (see Because antibodies to avian adenoviruses are widespread in
below). poultry flocks, serological methods are of little significance in
In most cases, establishment an isolate as an adenovirus may be diagnosis, except for the screening of SPF flocks. For this purpose,
sufficient for diagnosis. Further characterization, to establish ELISA is probably the test of choice. Indirect ELISAs have been
relatedness to standard strains, requires more detailed laboratory described and have been shown to be broadly group specific,
testing. The group 1 adenoviruses are subdivided into 5 species (A detecting antibodies to all group 1 avian adenoviruses (7). Antigen
to E; Table 19.1), based largely on molecular criteria, in particular for ELISA was grown by infecting monolayers of chicken kidney
restriction endonuclease analysis (REA) and sequencing data. cells with each avian adenovirus serotype. After 4 days, the cells
Viruses within each species are further subdivided into serotypes were scraped from the plastic, disrupted by sonication in distilled
largely on the result of cross-neutralization tests, although this can water and extracted with fluorocarbon. The antigen preparation was
also be done by PCR. REA of fowl adenovirus serotypes into 5 added to ELISA cuvettes at a predetermined dilution and allowed to
distinct groups was first described by Zsak and Kisary, (21) and has absorb overnight. After washing, test antisera were added, the
proved effective in clarifying relationships between serotypes and in cuvettes were again washed and an anti-chicken serum conjugated
some cases between strains showing differences in virulence e.g. with horseradish peroxidase was added. Following incubation and
strains associated with IBH (9). Polymerase chain reaction (PCR) washing positive reactions were identified by addition of the
primers, based on conserved regions of the hexon gene, are enzyme substrate, at a previously standardized concentration (7).
available which will amplify group 1 adenoviruses belonging to all A similar indirect ELISA for detection of group 3 (EDS) virus
12 reference serotypes (15). Treatment of the PCR product with 4 antibodies has also been described (5). In this test the EDS antigen
endonucleases produces restriction patterns which are specific for was grown in chick embryo liver cells, and an anti-chicken serum
each reference strain. Therefore, the combined use of PCR with conjugated with peroxidase was used with ortho phenylene diamine
REA can be used for detection and typing of all group 1 as the substrate. The test was shown to be equal in sensitivity to an
adenoviruses (15,11). Details of the primers and methodology are EDS serum neutralization test and an indirect fluorescent antibody
given by Meulemans et al., (15), and Hess (11). PCR’s are also test which used EDS virus infected chick embryo liver cells, fixed
available for specific detection of EDS virus (17) and for on 12-well multispot slides.
amplification of DNA from all 3 groups of avian adenoviruses (21). A group specific, microtitre based immunofluorescence test
Subdivision into serotypes can also be done by serum capable of screening large numbers of sera for group 1 adenovirus
neutralization test, which requires substantial cell culture expertise antibodies, has also been described (4). In this test, chick embryo
and access to reference antisera prepared against each prototype liver cells, grown as monolayers in the wells of 96-well, flat
strain. Prototype antisera (usually prepared in rabbits) are titrated bottomed microtitre plates for 48 hr, were infected with FAdVl
against 100 mean tissue culture infective doses (TdD50) of virus virus, for 24 hr, and the plates were fixed by addition of a diluted
and then diluted to contain 20 neutralizing units (e.g., a serum with hypochlorite solution. After washing in PBS, test sera were applied
a neutralizing antibody titer of 1:2048 is diluted to 1:100). The and the plates were incubated at 37 C for 30 min then washed and
strains are described in Table 19-1. For typing of isolates by serum an anti-chicken serum conjugated with FITC added. After
neutralization, the virus isolate is diluted to 1/10 and 1/1000 in cell incubation and washing, the wells were examined for fluorescent
culture medium, mixed with 1 volume of each prototype antiserum nuclei using an inverted fluorescence microscope.
containing 20 neutralizing units, and incubated for 2 hr at 37 C. Double-immunodiffusion or gel diffusion has been commonly
Duplicate monolayers of chick embryo liver or kidney cells, used for detecting avian adenovirus group specific antibodies. It is
growing in 96-well, flat bottomed microtitre plates or in 24 well cell fast, cheap, and simple to use but lacks sensitivity, so birds
culture plates, are inoculated with each virus-serum mixture and the undergoing a primary infection by natural routes may not respond
plates are incubated at 37 C and observed for CPE over a period of with detectable precipitating antibodies. However, if three antigens
6 days. Controls, consisting of virus alone, and antiserum alone are are pooled (e.g. FAdVl, FAdV5 and FAdV9) sensitivity can be
also inoculated. Neutralization of virus CPE indicates the serotype markedly improved (8). Antigen for gel diffusion test may be
of the isolate. Chick embryo liver cells are preferable to chick prepared by inoculating embryonating chicken eggs by the allantoic
kidney cells because the endpoints are more distinct. If desired, route with 0.1 ml of an egg-adapted strain of group 1 adenovirus,
pools of antisera can be used instead of individual antiserum. such as FAdVl. After 72 hr incubation at 37 C, the eggs are
chilled, and the chorioallantoic membranes harvested. One ml PBS
Physicochemical properties is added for each membrane and the membranes are homogenized
The more important physical and chemical properties may be then centrifuged at 1500 x g for 20 min to remove the debris. The
useful if an isolate is recovered, which cannot be neutralized using pooled supemate is stored in small volumes at -80 C as gel diffusion
the prototype antisera. Tests for determining the presence of antigen.
essential lipid, nucleic acid type, and thermal stability may be useful
and are described in Chapter 45 on virus identification and Type specific (neutralizing) antibodies
classification. Other characterization criteria may include Neutralizing antibodies to group 1 adenoviruses may occur in
morphology, site of replication, resistance to pH 3, and stability in chickens, turkeys, quail, pigeons, and other avian species. The
the presence of divalent cations. To test for resistance to pH 3, the neutralization test is similar to that described above for typing of
87
Brian M. Adair and J. Brian McFerran

isolates. Two-fold dilutions of the test sera (e.g. 1/16 to 1/2048) are titration in cell culture should be performed with new isolates. Ten­
added (50ul volumes) to duplicate wells of flat-bottomed microtitre fold dilutions of the virus isolate are prepared and inoculated into
plates. Two hundred TCID50 ,of the appropriate adenovirus serotype duplicate chick embryo liver or kidney cell cultures. The highest
in a 50 μΐ volume, are then added to each well. Following dilution showing CPE is harvested by freezing at -80 C and
incubation for 1 hour at 37 C, to allow neutralization to take place, thawing; the process is repeated a total of 3 times. Electron
50μ1 of chick embryo liver or kidney cell suspension is added to microscope examination should be used to confirm presence of only
each well. Virus controls containing 100, 10, 1.0, and 0.1 TCID50 of one agent. While the procedure is not foolproof, it gives some
virus are also included, and the plates are incubated and examined confidence that most other agents have been removed. Adeno-
for development of viral CPE. Neutralizing endpoints are read when associated parvoviruses have been found in adenovirus stock pools
the virus controls indicate that 100 TCID5o of virus is present in the and have also been isolated from healthy birds. They require
test. presence of adenovirus for their replication, and there is no
The same test can be used for EDS virus to confirm results of HI evidence to suggest an involvement in disease. Molecular tests
tests. In this case the endpoints can be read on the basis of CPE or (PCR) are available for their diagnosis.
by removing the supemate from each well to a parallel microtitre
plate, adding 50μ1 of chicken erythrocytes and observing for REFERENCES
presence of hemagglutination, as described above.
1. Adair, B.M Studies on the development of avian adenoviruses in cell
cultures. Avian Pathol. 7:541-550. 1978
HI test for group 3 (EDS) virus antibodies 2. Adair, B.M, Curran, W.L. and McFerran, J.B. Ultrastructural studies of
EDS virus growth in cell cultures or eggs is accompanied by the replication of fowl adenoviruses in primary cell cultures. Avian Pathol.
production of a hemagglutinating antigen, which agglutinates 8:133-144. 1979.
chicken and duck erythrocytes. Antibody to the virus inhibits the 3. Adair, B.M, McFerran, J.B., Connor, T.J., McNulty, MS. and McKillop,
hemagglutination reaction and this is used as the basis of the HI test E.R. Biological and physical properties of a virus (strain 127) associated
for EDS. The test can be used for detecting infection with EDS with the egg drop syndrome 1976. Avian Pathol. 8: 249-264. 1979.
virus and is not applicable to any other avian adenovirus. Birds do 4. Adair, B.M, McFerran, J.B. and Calvert, V.M Development of a
not usually develop antibody until after they have had EDS. Flocks microtitre fluorescent antibody test for serological detection of adenovirus
infection in birds. Avian Pathol. 9: 291-300. 1980.
that develop EDS through reactivation of latent virus usually do so 5. Adair, B.M., Todd, D., McFerran, J.B. and McKillop, E.R. Comparative
before they are 35 wk old. Birds of all ages are susceptible to the serological studies with egg drop syndrome virps. Avian Pathol. 15: 677-
virus, and if the EDS virus is introduced (e.g., through drinking 685. 1986.
contaminated water), infection can occur at any age. For screening 6. Benko, M, B.Harrach, and W.C. Russell. 2000. Family Adenoviridae. In:
purposes, 1/10 dilutions of test sera in PBS are added to duplicate Virus Taxonomy. Seventh Report of the International Committee on
wells of V-well microtitre plates. An equal volume of EDS antigen, Taxonomy of Viruses. MH.V. van Regenmortel, C.M Fauquet, D.H.L.
diluted to contain 4 hemagglutinating (HA) units, is added to one Bishop, E.B. Carstens, MK. Estes, S.M Lemon, J. Maniloff, MA. Mayo,
well, and 1 volume PBS to the duplicate (control) well. A titration D.J. McGeoch, C.R. Pringle, R.B. Wickner (eds), Academic Press, New
York, San Diego. Pp. 227-238. 2000.
of a positive serum (2-fold dilutions in PBS) should also be
7. Calnek, B.W., Shek, W.R., Menendez, N.A. and Stiube, P. Serological
included. The plates are shaken and left at room temperature for 15 cross-reactivity of avian adenovirus serotypes in an enzyme-linked
min, 1 volume of an 0.8% suspension of chicken erythrocytes is immunosorbent assay. Avian Dis. 26: 897-906 1982.
added to each well. The plates are again shaken and left at room 8. Cowen, B. S. A triple antigen for the detection of type 1 avian adenovirus
temperature until the erythrocytes have settled (about 45 min), and precipitin. Avian Dis. 31:351-354. 1987.
then viewed by holding at an angle of 45 degrees and allowing the 9. Emy, K. M, D. A Barr, and K. J. Fahey. Molecular characterization of
settled erythrocytes to stream. No hemagglutination should occur in highly virulent fowl adenoviruses associated with outbreaks of inclusion
the serum control well, no inhibition should occur with negative body hepatitis. Avian Pathol. 20:597-606. 1991.
10. Goodwin, MA, Latimer, K.S., Resurreccion, R.S., Miller, P.G. and
serum, and the positive serum should be at its correct titer. Finally,
Campagnoli, R.P. DNA in situ hybridization for the rapid diagnosis of
a back-titration carried out on the diluted antigen used in the test massive necrotizing avian adenovirus hepatitis and pancreatitis in chicks.
should indicate that 4 HA units are present. Antigen for HI tests Avian Dis. 40:828-831. 1996.
may be prepared by inoculating 10-day-old embryonated duck eggs 11. Hess, M Detection and differentiation of avian adenoviruses: a review.
allantoically with 0.2 ml of a 1/100 dilution of EDS virus. The Avian Pathol. 29: 195-206. 2000.
allantoic fluid is harvested 4-5 days after inoculation, clarified by 12. Latimer, K.S., Niagro, F.D., Williams, O.C., Ramis, A., Goodwin,
low-speed centrifugation, and stored at -80 C in small volumes. MA, Ritchie, B.W. and Campagnoli, R.P. Avian Dis. 41: 773-782. 1997
Alternatively chick embryo liver or chick embryo kidney 13. McFerran, J.B. and Smyth, J.A Avian Adenoviruses. Rev. Sci. Off. Int.
Epiz., 19: 589-601. 2000.
monolayers may be inoculated with 1000 TCID50 of EDS virus, and 14. Mazaheri, A, Prusas, C., Voss, M and Hess, M Some strains of
incubated until EDS virus CPE is seen in 10% of the cells. The serotype 4 fowl adenoviruses cause inclusion body hepatitis and
flasks of cells are then frozen at -80 C and thawed 3 times and the hydropericardium syndrome in chickens. Avian Pathol. 27: 269-276. 1998.
cell debris removed by centrifugation. The supernatant containing 15. Meulemans, G., Boschmans, M., van den Berg, T.P. and Decaesstecker,
the hemagglutinin is distributed in small volumes and stored frozen M Polymerase chain reaction combined with restriction enzyme analysis for
at -80 C. If inactivation of the antigen is required, this is done by detection and differentiation of fowl adenoviruses. Avian Pathol. 30: 655-
heating for 1 hr at 65 C in a water bath. This is preferred to formalin 660. 2001.
treatment. 16. Nakamura, K., Mase, M, Yamaguchi, S., Shibahara, T. and Yuasa, N.
Pathologic study of specific pathogen free chicks and hens inoculated with
adenovirus isolated from hydropericardium syndrome. Avian Dis. 43: 414-
DIFFERENTIATION FROM CLOSELY RELATED AGENTS 423. 1999.
17. Raue, R. and Hess, M Hexon based PCR’s combined with restriction
Differentiation of Isolates. Isolation attempts in cell cultures will enzyme analysis for rapid detection and differentiation of fowl adenoviruses
often result in the isolation of other agents, particularly reoviruses. and egg drop syndrome virus. J. Virol. Meth. 73: 211-217.
In addition to the different CPE in cells, these viruses produce intra- 18. Saifuddin, M and Wilks, C.R. Development of an enzyme linked
cytoplasmic inclusions. Mycoplasma may also be recovered from immunosorbent assay to detect and quantify adenovirus in chicken tissues.
specimens in cell culture and may be suspected due to their Avian Dis. 34: 239-245. 1990.
reovirus-like CPE and sensitivity to chloroform. To eliminate the
possibility of contamination with other agents, limit dilution
88
Chapter 19 Adenovirus

19. Smyth, J. A., M A. Platten, and J. B. McFerran. A study of the 20. Xie, Z., Fadi, A.A., Girshick, T. and Khan, MI. Detection of avun
pathogenesis of egg drop syndrome in laying hens. Avian Pathol. 17:653— adenovirus by polymerase chain reaction. Avian Dis. 43: 98-105. 1999.
666. 1988. 21. Zsak, L. and Kisary, J. Grouping of fowl adenoviruses based upon the
restriction patterns of DNA generated by Bam Hl and Hind HI. InterviroL
22: 110-114.

89
20
HEMORRHAGIC ENTERITIS OF TURKEYS
MARBLE SPLEEN DISEASE OF PHEASANTS
F. William Pierson and Scott D. Fitzgerald

SUMMARY. Hemorrhagic enteritis of turkeys and marble spleen disease of pheasants are caused by viruses of the family Adenoviridae,
genus Siadenovirus. They are serologically distinct from other avian adenoviruses but are indistinguishable from one another as determined
by agar gel immunodiffusion (AGID) test. Slight differences appear at the DNA level and clinical manifestations of disease vary with the
strain of virus and the host infected. Most gallinaceous birds are susceptible to infection, but disease is of primary importance in turkeys and
pheasants.
Agent Identification. Diagnosis can usually be made on the basis of clinical signs, lesions, histopathology, and the demonstration of viral
antigen or DNA in splenic tissue by AGID and PCR respectively.
Serologic Detection in the Host. Analysis of acute and convalescent sera by AGED or enzyme linked immunosorbent assay may also be of
diagnostic value.

INTRODUCTION Gross Lesions


Turkeys that have died from HE are pale due to loss of blood but
Hemorrhagic enteritis (HE) is an acute viral disease of turkeys, 4 are otherwise in good flesh and often have feed in the crop. The
wk of age and older caused by a virus of the family Adenoviridae, anterior small intestine is congested and the lumen is often filled
genus Siadenovirus, i.e., Turkey Hemorrhagic Enteritis Virus with blood. The intestinal mucosa especially at the level of the
(THEV) (8) In its most severe form the disease is characterized by duodenum has hemorrhage with progression from ecchymoses to
depression, bloody droppings, and death. Survivors as well as those petechiae, proximal to distal. In such cases the spleen may appear
with milder forms of the disease, often develop secondary coliform small and pale but in moribund birds killed prior to extensive blood
infections as a result of transient immunosuppression. This is loss, splenic enlargement with mottling is typically observed
believed to be the result of viral replication within IgM bearing B- (22,36,39).
lymphocytes and macrophages as well as cytokine mediated In pheasants, splenic enlargement with mottling, lung congestion
apoptosis of bystander cells (40). Gastrointestinal lesion formation and edema are the hallmark lesions. Gastrointestinal lesions are
is likewise considered to be immune mediated (40). Prevention of absent (5).
the gastrointestinal form of the disease may be accomplished
through the use of avirulent live-virus vaccines (14,18). Histopathology
Marble spleen disease (MSD) is a condition affecting captive- In cases of HE or MSD splenic tissue that has been fixed in 10%
reared pheasants between 3-8 mo of age and is caused by a virus neutral buffered formalin and stained with hematoxylin and eosin
serologically indistinguishable from THEV (11,28). However, reveals numerous distinct microscopic lesions. These include
MSD virus (MSDV) primarily causes pulmonary edema and death lymphoid follicular depletion due to virus-induced apoptosis and
by asphyxiation in affected pheasants. Mottled enlarged spleens are necrosis as well as prominent hyperplasia of large lymphoreticular
also characteristic (20). Like HE, fulminate MSD is thought to be cells within the white pulp, many of which contain eosinophilic
an immune mediated disease and may be prevented by vaccination intranuclear inclusions (23,29,41,40).
(15). Intestinal samples may be of confirmative value if preserved in
Both THEV and MSD have been reviewed extensively in the Zenker's fixative within a few minutes of death and stained with
literature (20,37). hematoxylin and eosin. Characteristic histopathologic changes
include severe congestion of the mucosa, degeneration and
CLINICAL DISEASE detachment of the villus epithelium, and hemorrhage within the
lamina propria (41).
Signs Occasional intranuclear inclusions may be seen in lymphocytes
The clinical signs of the gastrointestinal form of HE usually and macrophages of the lamina propria, in mononuclear cells
appear 3-4 days after exposure to the virus and include depression, located within the fiver, bone marrow, peripheral blood, lung,
pallor, bloody diarrhea, prostration, and death. Dark red to black pancreas, brain, and renal tubular epithelium (26,29,41,45).
gastrointestinal contents may be expressed from the cloaca of dead Microscopically the splenic lesions seen with MSD are identical
or moribund birds following application of moderate pressure to to those of HE. Lung lesions in pheasants that succumb to MSD are
the abdomen. Clinically affected birds usually die within 24 hr or consistent with the congestion and edema observed grossly (21).
recover completely. The disease spreads quickly through Intranuclear inclusions are occasionally found in mononuclear cells
susceptible flocks and usually subsides within 6-10 days of onset. in the lung, liver, proventriculus, bursa of Fabricius, and bone
The morbidity typically approaches 100% but mortality may range marrow (21).
from less than 1 % to as high as 60% depending on the viral
pathotype (13,24). Due to the immunosuppressive nature of the SAMPLE COLLECTION
virus a second rise in mortality commonly occurs approximately 2
wk after the first due to secondary infections with E. coli, THEV can be recovered from the intestinal contents or spleens of
Staphylococcus sp. or other agents (36, 42, 49). dead or moribund turkeys. However for both THEV and MSDV
The progression of clinical signs associated with MSD in mottled enlarged spleens are by far the best source (12). To obtain
pheasants is also rapid and includes depression, weakness, nasal useful quantities of virus the capsule of the spleen should be
discharge, dyspnea, and death. Gastrointestinal involvement is not removed and the parenchyma pressed through a syringe to disrupt
apparent. Mortality usually ranges between 2 and 3% but may the reticular architecture. This material should be diluted with an
reach as high as 20% over the course of approximately 10 days equal volume of 10 mM phosphate buffered saline (pH 7.4), mixed,
(5,15,33). frozen and thawed 2-3 times, and centrifuged. The resulting
supernatant may be used as a test antigen in immunoprecipitation
reactions or for further propagation in susceptible birds and cell
90
Chapter 20 Hemorrhagic Enteritis of Turkeys Marble Spleen Disease of Pheasants

culture. The virus is extremely stable and can be maintained for be divided into smaller aliquots, refrigerated, and re-liquefied by
extended periods by freezing or lyophilization. Infectivity is heating in a microwave or boiling water prior to the pouring of test
destroyed by drying or application of disinfectants containing plates. Disposable immunodiffusion plates that hold approximately
phenol or sodium hypochlorite (9,10). 15 ml of gel or comparable products are commonly used. Wells
should be 5 mm in diameter and hold approximately 50 ml.
PREFERRED CULTURE MEDIA AND SUBSTRATES. Unknown test material and known positive control antigen should
be placed in alternating wells around a central well containing
THEV and MSDV can be propagated in susceptible turkeys that positive control antiserum. Plates should be incubated in a
are 4 wk-of-age or older. However 5-6 wk of age is generally the humidified chamber at room temperature and read at 24 and 48 hr.
best time for infection due to the natural decline of maternal Bands of precipitation showing lines of identity with known
antibody (17). Birds may be inoculated intravenously or orally with positive antigen are indicative of THEV or MSDV infection.
about 0.25 ml of a IO’2 dilution of supernatant prepared as Quantification of THEV/MSDV antigen. An antigen capture
previously described. Removal of spleens should be performed 3-4 ELISA for HEV has been described in the literature (47). This
days after intravenous inoculation or 5-6 days after oral inoculation technique enables more precise quantitation of viral antigen in
(12). At these times the spleens will be grossly mottled and tissue extracts than does AGID. Competitive PCR may also be
enlarged. Successful in vitro propagation of THEV and MSDV can used for quantification in tissue based on the DNA copy number
be accomplished using MDTC RP19 cells (American Type Culture (2).
Collection, Rockville, MD), a turkey lymphoblastoid cell line Immunohistochemistry. As an adjunct to routine histopathology,
derived from a Marek’s disease virus-induced tumor (34) or in immunofluorescent and immunoperoxidase staining techniques
leukocytes isolated from the peripheral blood of THEV negative have been described that allow accurate identification of THEV or
turkeys (46). MSDV intranuclear inclusions in fixed tissues (19,21,41).

AGENT IDENTIFICATION Molecular Identification


The THEV/MSDV genome is small in comparison to other
Morphology and Physicochemical Properties members of the family Adenoviridae i.e., 26.3 kb in length (38).
THEV and MSDV are DNA viruses (27,31). They produce thin This in addition to the presence of unique open reading frames and
bands in sucrose or cesium chloride gradients at a density of 1.32- relatively high A+T content, formally establishes THEV and
1.34 g/ml (4,27). They are non-enveloped icosahedrons having 252 MSDV as being members of the genus Siadenovirus (7,8,38).
capsomeres of which 240 are non-vertex capsomeres (hexons). The Techniques for determining the presence of viral DNA in tissues
12 vertex capsomeres (pentons) possess a single fiber (43). using both in situ DNA hybridization and polymerase chain
Complete virions range in diameter from 70 to 90 nm (27,44). reactions (PCR) have been described (2,3,25,43). Due to its
enhanced sensitivity PCR may be useful in cases in which the
Biological Properties suspicion of HEV infection is high but presence of viral antigen
The viruses causing HE and MSD are believed to be pathotypic cannot be confirmed by AGID or antigen capture ELISA. A
variants of the same virus. THEV will cause splenic enlargement suitable technique involves the amplification of a 273 bp region of
and mottling in pheasants without producing significant pulmonary the hexon gene. Primers (NHEVF-5’-gtggttcagcagaaagttctt-3’;
lesions (15). Similarly MSDV infection in turkeys is characterized NHEVR-5’-cagtagactcataagcaactat-3’) are added to the mastermix
by splenic enlargement and mottling without gastrointestinal which contains thermostable Taq DNA polymerase, MgCl2 and
involvement (14). The observed variations in clinical disease are dNTPs (Platinum Taq Readymix®, Invitrogen, Grand Island, NY)
probably as much the result of differences in the host immune to a final concentration of 0.2 mM of each primer. Sample DNA
response i.e., target shock organ (37,40) as they are differences extracted from splenic tissue (InstaGene™ Matrix, Bio-Rad
between viral strains. Other variants exist that produce graded Laboratories, Hercules, CA) is then added and the PCR reaction is
intermediate clinical responses in infected birds (14). run (thermocycler program: initial denaturation at 95 C for 2 min;
Mottling and enlargement of the spleen have also been observed then 35 cycles of 95 C for 15 sec, 58 C for 15 sec, 72 C for 30 sec;
in chickens infected with a virus serologically indistinguishable and final extension at 72 C for 10 min). PCR products are
from THEV and MSDV (16). The disease has been referred to as visualized after agar gel electrophoresis (0.8% TBE-agarose gel,
avian adenovirus group Π splenomegaly (AAS) of chickens but 100V for 1 hour) by staining with ethidium bromide and UV trans­
probably only represents the expression of THEV/MSDV in a illumination. Intense bands corresponding with the predicted 273
different gallinaceous host. Except for occasional condemnation bp product are considered positive.
due to suspicion of Marek’s disease or lymphoid leukosis, AAS
appears to be of little economic significance (16). SEROLOGICAL DETECTION IN THE HOST

Antigen Detection Agar Gel Immunodiffusion Test for Antibody


Agar Gel Immunodiffusion (AGID) Test for Antigen. Isolates Anti-HEV or anti-MSDV antibodies can be detected in sera as
of THEV and MSDV are serologically indistinguishable from one early as 2-3 wk post-infection by AGID (12,13,30). The assay is
another. However an AGID may be used to differentiate HEV and identical to that described in the section on agent identification
MSDV from other pathogens (12,13). with the exception that unknown test sera should be alternated with
Viral antigen may be obtained from the spleens of suspect cases known positive control sera and placed in opposition to positive
as described in the section on sample collection. Positive control control (splenic) antigen. Testing of both acute and convalescent
antigen may be prepared as described in the section on preferred sera should be performed.
culture media and substrates. Positive control antiserum may be
obtained 4 wk post-inoculation. Enzyme-Linked Immunosorbent Assay (ELISA)
Immunodiffusion gels may be prepared in the following manner. Enzyme-linked immunosorbent assays have been developed for
Thirty-two (32.0) grams sodium chloride, 0.8 g sodium azide, and the detection of THEV/MSDV antibody (6,35,47) and are available
3.2 g agarose are added to 400 ml distilled water in a 1000-ml commercially (Synbiotics Corporation, San Diego, CA). These
Erlenmeyer flask. The mixture is then stirred on a hot plate until tests are more sensitive than AGID and may be useful in the
boiling, at which time it should clarify. The liquefied gel can then quantitation of low levels of antibody such as that found in young
91
F. William Pierson and Scott D. Fitzgerald

birds prior to and shortly after vaccination. In older birds with high 16. Domermuth, C. H., J. R. Harris, W. B. Gross, and R. T. DuBose. A
levels of antibody e.g., breeders or market age turkeys, additional naturally occurring infection of chickens with a hemorrhagic
dilution of sera may be required to bring optical densities within enteritis/marble spleen disease type of virus. Avian Dis. 23:479-484.1979.
17. Fadly, A. M, and K. Nazerian. Hemorrhagic enteritis of turkeys:
readable limits. In most cases multiplication of the sample to
influence of maternal antibody and age at exposure. Avian Dis. 33:778-86.
positive ratio by the appropriate dilution factor prior to the 1989.
calculation of the log titer will enable comparison with younger 18. Fadly, A Μ, K. Nazerian, K. Nagaraja, and G. Below. Field
birds. vaccination against hemorrhagic enteritis of turkeys by a cell-culture live-
virus vaccine. Avian Dis. 29:768-777. 1985.
DIFFERENTIATION FROM CLOSELY RELATED AGENTS 19. Fasina, S. O., and J. Fabricant. In vitro studies of hemorrhagic enteritis
virus with immunofluorescent antibody technique. Avian Dis. 26:150-
In turkeys the presence of mottled enlarged spleens in the absence 157.1982.
of detectable HEV antigen or intestinal bleeding should arouse 20. Fitzgerald, S. D., and W. M. Reed. A review of marble spleen disease
of ring-necked pheasants. J. Wildl. Dis. 25:455-461. 1989.
suspicion of other diseases such as reticuloendotheliosis (see
21. Fitzgerald, S. D., W. M Reed, and T. Bumstein. Detection of type Π
Chapter 35) or lymphoproliferative disease (1). Splenic avian adenoviral antigen in tissue sections using immunohistochemical
enlargement may also be indicative of a generalized septicemia or staining. Avian Dis. 36:341-347. 1992.
other acute conditions (37, see Chapters 3, 4, and 9). Congestion 22. Gross, W. B. Lesions of hemorrhagic enteritis. Avian Dis. 11:684-693.
and hemorrhage in the proximal small intestine should also prompt 1967.
consideration of coccidiosis (Eimeria meleagrimidis) (32). In 23. Gross, W. B., and C. H. Domermuth. Spleen lesions of hemorrhagic
pheasants mortality associated with asphyxia, dyspnea, pulmonary enteritis of turkeys. Avian Dis. 20:455-466. 1976.
edema, and mottled enlarged spleens may be considered 24. Gross, W. B., and W. E. C. Moore. Hemorrhagic enteritis of turkeys.
Avian Dis. 11:296-307. 1967.
pathognomonic for MSD.
25. Hess, M, R. Raue, and Η. M Hafez. PCR for specific detection of
haemorrhagic enteritis virus of turkeys, an avian adenovirus. J. Virol.
ACKNOWLEGEMENT Methods 81:199-203. 1999.
26. Hussain, I., C. U. Choi, B. S. Rings, D. P. Shaw, and K. V. Nagaraja.
The authors are greatly indebted to Drs. C. H. Domermuth and W. B. Gross Pathogenesis of hemorrhagic enteritis virus infection in turkeys. Zentralbl.
for their contributions to earlier editions of this chapter. Veterinarmed. 40:715-726.1993.
27. litis, J. P., and S. B. Daniels. Adenovirus of ringnecked pheasants:
REFERENCES purification and partial characterization of marble spleen disease virus.
Infect. Immun. 16:701-705. 1977.
1. Biggs, P. M Lymphoproliferative disease of turkeys. In: Diseases of 28. litis, J. P., R. M Jakowski, and D. S. Wyand. Transmission of marble
poultry, 10th ed B. W Calnek. H. J. Barnes, C. W. Beard, L R. McDougald, spleen disease in turkeys and pheasants. Am J. Vet. Res. 36:97-101. 1975.
Y. M Saif, eds. Iowa State University Press, Ames, Iowa. pp. 485-488. 29. Itakura, C., and H. C. Carlson. Pathology of spontaneous hemorrhagic
1997. enteritis of turkeys. Can. J. Comp. Med. 39:310-315.1975.
2. Beach, N., C. Cardona, R.B. Duncan, Jr., D. Wise, and F. W. Pierson. 30. Jakowski, R. M, and D. S. Wyand. Marble spleen disease in ring­
Competitive PCR for the quantification of hemorrhagic enteritis virus. In: necked pheasants: demonstration of agar gel precipitin antibody in
Proceedings of the 73rd Northeastern Conference on Avian Diseases, pheasants from an infected flock. J. Wildl. Dis. 8:261-263. 1972.
College Park, Maryland 2001. 31. Jucker, M T„ J. R. McQuiston, J. V. van den Hurk, S. M, Boyle, and
3. Beach, N., C. Cardona, D. Wise, R. Duncan, and F. W. Pierson. Latency F. W. Pierson. Characterization of the haemorrhagic enteritis virus genome
of hemorrhagic enteritis virus as determined by nested PCR. In: and the sequence of the putative penton base and core protein genes. J. Gen.
Proceedings of the 74th Northeastern Conference on Avian Diseases, Virol. 77:469-479. 1996.
Mystic, Connecticut. 2002. 32. MacDougald, L. R. Coccidiosis. In: Diseases of poultry, 11th ed. Y.M
4. Carlson, H. C., F. AI-Sheikhly, J R. Pettit, and G. L. Seawright. Virus Saif., H.J. Barnes, J.R. Glisson, A.M Fadly. L.R. McDougald, and D.E.
particles in spleens and intestines of turkeys with hemorrhagic enteritis. Swayne, eds. Iowa State University Press, Ames, Iowa. pp. 988. 2003.
Avian Dis. 18:67-73. 1974. 33. Mayeda, B., G. B. West, A A. Bickford, and B. R. Cho. Marble spleen
5. Carlson, H. C., J. R. Pettit, R. V. Hemsley, and W. R. Mitchell. Marble disease in pen-raised pheasants in California. Proc. Amer. Assoc. Vet. Lab.
spleen disease of pheasants in Ontario. Can. J. Comp. Med. 37:281-286. Diagn. 25:261-270. 1982.
1973. 34. Nazerian, K., and A. Fadly. Propagation of virulent and avirulent
6. Davidson, I., A. Aronovici, Y. Weisman, and M Malkinson. Enzyme turkey hemorrhagic enteritis virus in cell culture. Avian Dis. 26:816-
immunoassay studies on the serological response of turkeys to hemorrhagic 827.1982.
enteritis virus. Avian Dis. 29:43-52. 1985. 35. Nazerian, K., and A M Fadly. Further studies on in vitro and in vivo
7. Davison, A J., and B. Harrach. Siadenovirus. In: The springer index of assays of hemorrhagic enteritis virus (HEV). Avian Dis. 31:234-240. 1987.
viruses. C.A. Tidona and G. Darai, eds.: Springer-Verlag, New York, New 36. Pierson, F. W., V. D. Barta, D. Boyd, and W. S. Thompson. Exposure
York, pp.29-33. 2002. to multiple infectious agents and the development of colibacillosis in
8. Davison, A.J., M Benko, and B. Harrach. Genetic Content and Evolution turkeys. J. Appl. Poult. Res. 5:347-357.1996.
of Adenoviruses. J. Gen. Virol. 84:2895-2908. 2003. 37. Pierson, F. W., and S. D. Fitzgerald. Hemorrhagic enteritis and related
9. Domermuth, C. H., and W. B. Gross. Effect of disinfectants and drying infections. In: Diseases of poultry, 11th ed. Y.M Saif, H.J. Barnes, J.R.
on the virus of hemorrhagic enteritis of turkeys. Avian Dis. 15:94-97. 1971. Glisson, A.M. Fadly, L.R. McDougald, and D.E. Swayne, eds. Iowa State
10. Domermuth, C. H., and W. B. Gross. Effect of chlorine on the virus of University Press, Ames, Iowa. pp. 237-247. 2003.
hemorrhagic enteritis of turkeys. Avian Dis. 16:952-953. 1972. 38. Pitcovski, J., M Mualem, Z. Rei-Koren, S. Krispel, E. Shmueli, Y.
11. Domermuth, C. H., and W. B. Gross. Hemorrhagic enteritis of turkeys: Peretz, B. Gutter, G.E. Gallili, A. Michael, and D. Goldberg. The complete
antiserum efficacy, preparation and use. Avian Dis. 19:657-665. 1975. DNA sequence on genome organization of the avian adenovirus,
12. Domermuth, C. H., W. B. Gross, R. T. DuBose, C. S. Douglass, and C. hemorrhagic enteritis virus. Virol 249:307-315. 1998.
B. Reubush, Jr. Agar gel diffusion precipitin test for hemorrhagic enteritis 39 Pomeroy, B. S., and R. Fenstermacher. Hemorrhagic enteritis in
of turkeys. Avian Dis. 16:852-857. 1972. turkeys. Poult. Sci. 16:378-382. 1937.
13. Domermuth, C. H., W. B. Gross, and R. T. DuBose. 40. Rautenschlien, S., and J. M Sharma. Immunopathogenesis of
Microimmunodiffusion test for hemorrhagic enteritis of turkeys. Avian Dis. haemorrhagic enteritis virus (HEV) in turkeys. Devel. Comp. Immuno.
17:439-444. 1973.
24:237-246. 2000.
14. Domermuth, C. H., W. B. Gross, C. S. Douglass, R. T. DuBose, J. R. 41. Saunders, G. K., F. W. Pierson, and J. V. van den Hurk. Haemorrhagic
Harris, and R. B. Davis. Vaccination for hemorrhagic enteritis of turkeys. enteritis virus infection in turkeys: a comparison of virulent and avirulent
Avian Dis. 21:557-565. 1977. virus infections and a proposed pathogenesis. Avian Pathol. 22:47-58.
15. Domermuth, C. H., W. B. Gross, L. D. Schwartz, E. T. Mallinson, and 1993.
R. Britt. Vaccination of ring-necked pheasant for marble spleen disease.
Avian Dis. 23:30-38. 1979.
92
Chapter 20 Hemorrhagic Enteritis of Turkeys Marble Spleen Disease of Pheasants

42. Sponenberg, D. P., C. H. Domermuth, and C. T. Larsen. Field 46. van den Hurk, J. V. Propagation of hemorrhagic enteritis virus in
outbreaks of colibacillosis of turkeys associated with hemorrhagic enteritis normal (non-tumor derived) cell culture. J. Am. Vet. Med. Assoc. 187:307.
virus. Avian Dis. 29:838-842. 1985. 1985.
43. Suresh, M., and J. M Sharma. Pathogenesis of type II avian adenovirus 47. van den Hurk, J. V. Quantitation of hemorrhagic enteritis virus antigen
infection in turkeys: in vivo immune cell tropism and tissue distribution of and antibody using enzyme-linked immunosorbent assays. Avian Dis.
the virus. J. Virol. 70:30-36. 1996. 30:662-671. 1986.
44. Tolin, S. A., and C. H. Domermuth. Hemorrhagic enteritis of turkeys. 48. van den Hurk, J. V. Characterization of the structural proteins of
Electron microscopy of the causal virus. Avian Dis. 19:118-125. 1975. hemorrhagic enteritis virus. Arch. Virol. 126:195-213. 1992.
45. Trampel, D. W., C. U. Meteyer, and A. A Bickford. Hemorrhagic 49. van den Hurk, J. V., B. J. Allan, C. Riddell, T. Watts, and A. A Potter.
enteritis virus inclusions in turkey renal tubular epithelium. Avian Dis. Effect of infection with hemorrhagic enteritis virus on susceptibility of
36:1086-1091. 1992. turkeys to Escherichia coli. Avian Dis. 38:708-716. 1994.

93
21
INFECTIOUS LARYNGOTRACHEITIS
Deoki N. Tripathy and Maricarmen Garcia

SUMMARY. Infectious laryngotracheitis (ILT) is either an acute, highly contagious herpesvirus infection of chickens, characterized by
severe dyspnea, coughing, and rales; or a subacute disease with lacrimation, tracheitis, conjunctivitis, and mild rales. ILT has been reported
from most of the intensive poultry-rearing areas of the United States and many other countries. Mortality varies and may reach 50% in adult
birds, usually due to occlusion of the trachea by hemorrhage or exudate. Following recovery, some birds remain carriers for variable periods
and become a source of infection for susceptible birds. Diagnosis during the acute stage of infection is usually made by clinical signs and
demonstration of intranuclear inclusion bodies in the infected tracheal epithelium.
Agent Identification. Infectious laryngotracheitis is routinely diagnosed by histopathologic examination of tracheal lesions characterized by
the presence of intranuclear inclusion bodies. In addition, viral particles exhibiting typical herpesvirus morphology can be detected by
negative staining of lesion suspensions or by direct examination of ultrathin sections of affected tissues using transmission electron
microscopy. The virus is isolated by inoculation of chorioallantoic membrane (CAM) of 9 to 12-day-old developing chicken embryos.
Lesions develop on the CAM 5 to 7 days post-infection. Etiology is confirmed by histopathologic examination of the CAM for intranuclear
inclusion bodies or electron microscopy for herpesvirus morphology. Isolated viruses can be evaluated for pathogenicity in susceptible hosts,
for example, chickens (development of clinical signs and tracheal lesions), or for growth and pathogenicity of avian cell culture (cytopathic
effect and plaque formation).
Genomic characterization of virus isolates can be based on restriction fragment length polymorphism (RFLP), in which the DNA profiles
are compared after restriction enzyme digestion and subsequent agarose gel electrophoresis. Although cloned genomic fragments have been
used as diagnostic probes, the use of polymerase chain reaction (PCR) for the amplification of a specific portion of the genome have been
used more frequently as a rapid diagnostic method for detection of viral nucleic acid in experimentally and naturally infected birds. Although
PCR has not been used as a routine diagnostic tool, PCR-RFLP procedures has been widely utilized as an epidemiological tool to
differentiate among viral isolates. Other rapid diagnostic methods utilized are the immunoperoxidase staining, indirect fluorescent antibody,
and the agar-gel immunodiffusion (AGID) test, all utilized to detect the viral antigen in affected tissues.
Serologic Detection in the Host. Serologic detection of infection may be important in experimental studies and in measuring immune
response following vaccination. Antibody response can be measured by AGID tests, the immunoperoxidase (IP) test, and the enzyme-linked
immunosorbent assays (ELISA). The immunoperoxidase/indirect fluorescent antibody and AGID tests can also be used to detect the antigen
in affected tissues. Antigenic differences among isolates can be determined by host pathogenicity and virus neutralization.
Vaccination with modified live strains of low virulence is practiced in endemic areas and on farms where a specific diagnosis has been
made. Vaccination in the face of an outbreak shortens the course of the disease.

may be detectable in some cases. In the milder forms of the disease,


INTRODUCTION only a few birds in a flock show classical respiratory signs, and
mortality is low. In such cases, the clinical signs are characterized
Infectious laryngotracheitis (ILT) is an acute respiratory disease of by mild tracheatis, swollen sinus and mild conjunctivitis (32, 37).
chickens caused by an alphaherpesvirus. ILT has been recognized in Turkeys and pheasants have a low degree of susceptibility to ILTV
many countries and is an economically important disease of (18).
chickens (18). In areas with large concentrations of poultry, ILT is
responsible for losses due to lowered egg production and mortality. SAMPLE COLLECTION
As a member of the alpha-herpesvirus family the ILT virus (ILTV)
has the ability to establish latency and to persist in recovered Tracheal exudates, tracheal scrapings and lung should be collected
chickens for variable periods after recovery from the disease. Thus,
for virus isolation in the early acute phase of the disease. A 10%
the carrier state is of considerable epizootiological significance, as suspension is made of the tissue in sterile normal saline, Hanks
carrier birds may shed the reactivated virus and may be responsible
balanced salt solution, or broth by grinding the tissue either with
for new outbreaks of the disease.
sterile fine sand or 60-mesh aluminum oxide with a sterile mortar
and pestle or in a glass grinder. Alternatively tracheal scrapings can
CLINICAL DISEASE
be collected with a sterile scalpel and resuspended in any suitable
sterile transport media. The suspension is centrifuged at 700 x g for
Infection with ILTV is characterized by anorexia, depression, and
10 min, and antibiotics (1000 IU/ml penicillin and 1 g/ml of
severe respiratory distress, with coughing, sneezing, gurgling,
dihydrostreptomycin) are added. The supernatant fluid is held at
gasping, and rales. The neck is often extended during inspiratory
room temperature for 30-50 min before inoculation into the culture
efforts, because the trachea is often partially occluded, and bloody,
substrate.
mucous exudate may be expelled during violent expectorating Tracheal tissue from early acute cases may be collected in Bouin’s
efforts. Conjunctivitis has been characteristic of more recent
or Zenker’s fixatives for demonstration of intranuclear inclusion
outbreaks Although ILTV causes a highly localized infection
bodies by histopathologic examination. Serum samples may be
involving mainly the trachea and the conjunctiva, extratracheal
collected to measure antibody responses by virus-neutralization
spread of the virus to the lung, trigeminal ganglion and brain has (VN), agar-gel immunodiffusion (AGID), enzyme-linked
also been reported.
immunosorbent assay (ELISA), or fluorescent antibody (FA) tests
In severe forms of the disease, mortality is as high as 50% or
to detect specific antibodies.
more, with significant lesions consisting of hemorrhages in the
trachea and larynx. The tracheal lumen is often filled with blood
clots, mucus, caseous yellowish exudate, or a tracheal plug, which
may cause death from asphyxia. Intranuclear inclusion bodies are
detectable in the early stage of the disease on histopathologic
examination of the infected trachea. Air sacculitis and pneumonitis
94
Chapter 21 Infectious Laryngotracheitis

PREFERRED CULTURE MEDIA AND SUBSTRATES Camoys solution and stained either by the Feulgen method or with
acridine orange) inclusion bodies in a few multinucleate giant cell
Routinely ILTV can be isolated from tracheal exudates, tracheal nuclei are observed 12 hr after infection. Degenerate and detached
scrapings or lung tissue suspensions by inoculation of developing cells are seen from 36 to 72 hr, and degeneration may be complete
embryonated eggs, the trachea and infraorbital sinuses of by 72 hr PI. Adult chicken kidney culture, CEK cells, and the LMH
susceptible chickens, or inoculation of chick embryo liver (CEL) cell line have been used as a plaque system for quantitative assay of
and chicken embryo kidney (CEK) monolayer cultures and virus ILTV. Plaques of 1 to 2 mm in diameter develop by 4 to 5 days PI
isolation from tracheas of latently infected birds has been possible (35, 39).
using the tracheal organ culture system (18).
AGENT IDENTIFICATION
Embryo Inoculation
Chicken embryos, 9 to 12 day-old, from specific-pathogen-free Infectious laryngotracheitis (ILT) is routinely diagnosed by
flocks are inoculated on the chorioallantoic membrane (CAM) with histopathologic examination of tracheal lesions for the presence of
a 0.1 to 0.2-ml suspension of tracheal exudate or tracheal and lung intranuclear inclusion bodies. In addition, viral particles exhibiting
tissues. Inoculated embiyos are incubated at 37 C. As early as 3 typical herpesvirus morphology can be detected by negative
days post-inoculation (PI), plaques can be observed on the CAM staining of suspensions from affected tissues or by direct
that have opaque edges and depressed central areas of necrosis. The examination of ultrathin sections with electron microscopy. On
plaques result from proliferation and necrosis of the affected cells inoculated CAM the etiology is confirmed by histopathologic or
and may vary from a few scattered foci to large numbers. Some ultrastructural examination of the CAM lesions for intranuclear
embryos die between 2 to 8 days PI, and the size of surviving inclusion bodies or viral particles with herpesvirus morphology.
embryos is often reduced. Isolated viruses can be evaluated for pathogenicity in susceptible
hosts, for example, in chickens (development of clinical signs and
Chicken Inoculation tracheal lesions), or for susceptibility of avian cell culture
Susceptible chickens can be inoculated by tracheal, infraorbital (cytopathic effect and plaque formation).
sinus, or aerosol methods with a suspension of the specimen. Under A slide-smear technique for diagnosis of ILT has been described
experimental conditions the severity of respiratory signs and (7). Smears from scrapings of laryngeal mucosa (or conjunctiva in
mortality depends on virulence of the viral strain, susceptibility and the case of conjunctivitis) are fixed for 3-5 min in absolute methyl
age of the birds, and the route of inoculation. The virus multiplies alcohol and stained with Giemsa (1 drop of stock per ml of distilled
primarily in the trachea and can be isolated 2 to 7 days after water) for 2 hr at 37 C or overnight at room temperature. Stained
intratracheal inoculation. Virus titer is at a maximum 3 to 4 days PI. smears are washed in tap water and differentiated in absolute
Chickens infected with virulent strains usually develop signs of methyl alcohol, using three to five quick dipping motions, until the
respiratory tract infection by day 3 PI. Most chickens show typical smear has a magenta cast. The smear is washed in tap water, air­
signs of the disease by days 4 and 5. Mild respiratory disease with dried, and examined. Intranuclear inclusion bodies of ILTV are
mild, focal, non-suppurative pneumonitis and air sacculitis may be detectable in the early acute stages of disease. Tracheal impression
observed in some birds. smears stained by Giemsa in early stages of the disease also reveal
Gross changes in birds infected experimentally with virulent ILTV intranuclear inclusion bodies. The inclusions appear purple with a
by the aerosol method consist of severe hemorrhagic laryngitis and pinkish cast and are surrounded by a halo. Inclusions can be
tracheitis by day 3 PI, with extension to the syrinx and bronchi by observed in conjunctival smears in cases with conjunctivitis.
day 4. A yellowish, diphtheritic membrane lining the trachea and Sevoian (38) described a quick method for diagnosis of ILT. This
larynx is observed at about day 6 PI and may be found as a plug in method uses simultaneous fixation and dehydration and
the laiynx and upper trachea by day 8. The tracheal exudate is demonstrates intranuclear inclusion bodies in less than 3 hr. In
expelled by day 10. Gross lesions of the lungs and air sacs are another histologic diagnostic method tracheas and affected tissues
observed in some experimental chickens between days 3 and 7 PI are embedded in wax medium for sectioning and satisfactory slides
and occasionally for longer periods. are prepared in 3 hr (33).
During the incubation stage in experimentally infected, susceptible Direct electron microscopy can be used to identify virions of
birds, isolated areas of epithelial hyperplasia appearing as giant cell typical herpesvirus morphology in lysed cells from tracheal
syncytia occur in the trachea and laiynx by 48 hr PI. Intranuclear scrapings of infected birds. Various serologic tests (e.g., FA,
inclusion bodies are detectable in the syncytia. Necrotizing immunoperoxidase [IP], and AGID), described later in the chapter,
tracheitis with sloughing of the epithelium in cell groups with can be used to detect the ILTV antigen in the specimen.
intranuclear inclusion bodies is seen by day 3 PI. Inclusions are
usually observed from 4 to 6 days PI. At 5 to 7 days PI, the lesion Fluorescent Antibody (FA)
progresses to extensive necrosis of the epithelium, and inclusions The viral antigen can be detected by direct and indirect FA in
are visible less often. In recovered birds, the epithelium regenerates tracheal smears or frozen tracheal sections during the early acute
to normal appearance by day 12 (34). Pneumonitis and airsacculitis phase of the disease (2 to 8 days after exposure) 22). The FA test is
are often present during the acute stage, and giant cell syncytia a useful rapid assay. Direct FA on smears of tracheal scrapings has
containing intranuclear inclusion bodies are observed in the air sac been evaluated on experimentally and naturally infected birds. On
walls and lungs. naturally infected birds, viral antigens were detected from 2 to 14
days PI, while characteristic ILTV lesions were detected by
Cell Culture histopathological examination were detected from day 3 to 10 PI
Chick embryo liver (CEL) and chicken embryo kidney (CEK) (43). In naturally infected birds, FA and histopathological
cells are suitable for isolation of ILTV (23). A chicken liver tumor examination were equally satisfactory in the detection of infected
cell line, LMH, has also been used for propagation of ILTV (39). birds (15).
Early cytopathic changes in the CEK cells after inoculation with
ILTV consist of the development of numerous multinucleate giant Immunoperoxidase (IP)
cells that grow, coalesce, and finally undergo degeneration with An indirect IP method with monoclonal antibody has been used
continued incubation. Large, basophilic (methanol-fixed and stained for detection of ILTV antigen in frozen tracheal sections (17). In a
by the May-Griinwald-Giemsa method), DNA-positive (fixed in recent experimental study (2), IP was more sensitive than other

95
Deoki N. Tripathy and Maricarmen Garcia

diagnostic tests when histopathology, FA, IP, PCR, and sequences (10, 14). In these reports outbreak related isolates
hybridization were compared. collected before the use of vaccination were easily differentiated
from vaccines strains. However, outbreak related isolates obtained
Molecular Identification after the implementation of vaccination were identical to the
Procedures for detection of ILTV DNA using dot-blot currently utilized vaccines. A PCR-RFLP analysis of the viral
hybridization assays with cloned ILTV DNA fragments labeled glycoprotein E (gE) gene demonstrated that different viral
with p32 or digoxigenin have been described (12, 26, 29). These subpopulations are present in the chicken embryo origin (CEO)
procedures demonstrated to be highly sensitive for detection of ILTV vaccine preparations. This assay permitted the identification
ILTV in acutely affected and convalescent chickens when detection of vaccine related isolates in as the cause of outbreaks in the US
was no longer possible by virus isolation. With the advent of (13). Korean field isolates were differentiated from vaccine strains
polymerase chain reaction (PCR) several viral nucleic acid by PCR-RFLP analysis of the glycoprotein G and thymidine kinase
amplification procedures have been described (1, 4, 24, 40, 41, 44, (tk) genes (20). Although vaccine strains and field isolates from
45). Some PCR have included the use of non-isotopically labeled different countries were differentiated successfully using different
probes on membrane hybridization (1, 4), or an ELISA format, to PCR-RFLP none of the reported ILTV DNA differences between
enzymatically detect the amplification products (40). Otherwise vaccine and field isolates has been related to differences in strains
amplification products are visualized by gel electrophoresis. Nested pathogenicity at this moment.
PCR amplifications, with two sets of primers, have been utilized to
increase the sensitivity of detection of ILTV DNA (24). SEROLOGIC DETECTION IN THE HOST
Different samples have been evaluated for the detection of ILTV
DNA by PCR. For example, viral DNA has been successfully Agar-Gel Immunodiffusion
amplified from ILTV infected cell culture supernatants (40), Infectious laryngotracheitis virus antigen is prepared from infected
tracheal scrapings (41, 45), tracheal swabs (44), conjunctival swabs CAMs of embryonated eggs by homogenizing membranes that have
(4), turbinates, trigeminal ganglia (44), and from formalin-fixed, confluent lesions. Large tissue particles are removed by
paraffin embedded tracheal tissues (24). centrifugation at 1600 x g, and the supernatant is used as an antigen.
The ability of PCR to detect ILTV infected birds has been Tracheal antigen is prepared from a bird that either died of ILT or
compared to other diagnostic assays in samples from experimentally was euthanized within 1 wk of the onset of clinical signs. The
(2, 4) and naturally infected birds (45). In tracheas from antigen consists of exudate collected from the lumen and walls of
experimentally infected birds PCR was more effective than virus the larynx, trachea, and extrapulmonary bronchi. If the exudate is
isolation in the detection of ILTV, particularly during late stages of too dry, it is diluted with normal saline. Antigen prepared from
infection when birds have recovered from clinical signs. In naturally either the ILTV-infected CAM or the tracheal exudate is reacted
infected birds PCR detected viral DNA from tracheal samples that with specific antibody in plates prepared with 1.5% Noble agar and
contained bacteria and other viruses that prevented the isolation of 8% sodium chloride in veronal buffer (pH 7.2). Sodium azide in 1%
ILTV in cell culture (45). Molecular methods for differentiating concentration of thimerosal in 0.01% concentration is added as a
ILTV strains have been described. These include restriction preservative. A line or lines of precipitation will develop between
fragment length polymorphism (RFLP) analysis of the viral genome the antigen and antibody wells after 24 to 48 hr of incubation at
(6, 16, 19, 27, 28), reciprocal DNA : DNA hybridization using room temperature if the concentration of antigen is adequate in the
cloned DNA fragments (31, 42), and RFLP of PCR amplified tracheal exudate. Because the amount of precipitating antigen in the
products (9, 10, 13, 14, 20). RFLP analysis of the viral genome tracheal exudate and the amount of live virus will vary with the
requires large amounts of purified viral DNA, reciprocal period of infection and from bird to bird, AGID should be
hybridization protocols call for in-vitro radiolabelling of viral DNA. performed in combination with egg or cell culture inoculation (and
Although these procedures are useful in epidemiological studies tests such as IP, FA, or PCR). A small amount of five virus present
they are cumbersome not practical for larger epidemiological in tracheal exudate will infect the CAM of embryonated eggs or cell
studies, and not feasible for the daily diagnostic routine. The use of culture, whereas an inadequate concentration of precipitating
PCR based methods has greatly facilitated the molecular antigen will not form a line of precipitation with the antibody.
differentiation of ILTV because pure viral DNA is not required, and AGID also can be used to detect the antibody responses of
a large number of isolates from a single outbreak, or from different recovered birds by reacting the sera with known antigen prepared
geographical regions can be analyzed in a timely fashion. from ILTV-infected cell cultures or CAM (25). A positive response
may occur in 25%-50% of the birds, and occasionally in over 80%.
Strain Variability The test is less sensitive than IP, ELISA, FA, and VN. However, it
still may be a useful test for detecting antibodies on a flock basis
Naturally occurring strains of ILTV of variable pathogenicity have
been isolated from field outbreaks. However, not enough (3).
information is available on their characterization. The DNA of
Virus Neutralization
virulent and avirulent strains shows high homology by reciprocal
Infectious laryngotracheitis virus or ILTV antibody can be
DNA-DNA hybridization (30, 42). However, some minor identified by the VN test. Undiluted serum mixed with 10-fold virus
differences can be detected by restriction endonuclease analysis dilutions or serially diluted serum mixed with a single concentration
(27) and by PCR RFLP analysis (9, 11, 14, 42). A PCR-RFLP assay
of virus is held at room temperature for 1^4 hr before inoculation of
developed by Clavijo and Nagy (9) allowed the differentiation of a 9-to-12-day-old embryonated chicken eggs or CEK cell cultures.
virulent LT isolate from a low less virulent Canadian isolate.
Neutralizing antibody is detectable by 1 wk PI (21). A microassay
Australian vaccine and a pathogenic field strain were differentiated and neutralization test with CEK (35) and CEL cells (5) in
by the identification of a 467 base pair insertion/deletion in the
microcell culture trays can be used satisfactorily.
ICP4 gene non-coding region (42). The 467 bp pair deletion present
in the Australian pathogenic field isolate was not detected in other Enzyme-Linked Immunosorbent Assay
pathogenic field isolates from Taiwan (11). However, Taiwanese An ELISA tests has been developed to detect serum antibodies
isolates as well as isolates from England, Scotland, and the against ILTV and is available commercially in United States and
Republic of Ireland were differentiated from vaccine strains by Europe. Current ELISA tests utilize antigen prepared from either
PCR-RFLP analysis of the coding and non-coding ICP4 gene infected CAM or infected cell cultures. Using antigen prepared
96
Chapter 21 Infectious Laryngotracheitis

from either infected CAM or infected cell cultures can detect 16. Guy, J. S., H. J. Bames, L. L. Munger, and L. Rose. Restriction
antibodies in the sera of infected birds as early as 7 days PI (3). The endonuclease analysis of infectious laryngotracheitis viruses: Comparison of
ELISA was shown to be more sensitive than VN in the detection of modified-live vaccine viruses and North Carolina field isolates. Avian Dis.
33:316-323.1989.
ILTV serum antibodies (8). ELISA is an ideal test for survey
17. Guy, J. S., H. J. Bames, and L. G. Smith. Rapid diagnosis of infectious
purposes particularly when it has been utilized to evaluate the laryngotracheitis using a monoclonal antibody-based immunoperoxidase
antibody response of vaccinated flocks (31, 36). Antigen capture procedure. Avian Path. 21:77-86. 1992.
ELISA using ILTV polyclonal or monoclonal antibodies and then 18. Guy, J. S. and T. J. Bagust. Laryngotracheitis. In: Diseases of Poultry,
detecting the capture antigen by enzyme-labeled antibodies has also 11th ed. Y. M Saif, ed. Iowa State University Press, Ames, Iowa. pp. 121-
been utilized as a rapid diagnostic test to detect infected flocks (46). 134.2003.
19. Han, M G. and S. J. Kim. Comparison of virulence and restriction
DIFFERENTIATION FROM CLOSELY RELATED AGENTS endonuclease cleavage patterns of infectious laryngotracheitis virus isolated
in Korea. Avian Pathol. 30: 337 -344. 2001a.
Although acute signs of ILT, characterized by coughing, expulsion 20. Han, M G., and S. J. Kim. Analysis of Korean strains of infectious
larymgotracheitis virus by nucleotide sequences and restriction fragment
of blood, and high mortality, can be suggestive of ILT, many signs
length polymorphism. Veterinary Microbiol. 83:321 - 331. 2001b.
are similar to those of other respiratory diseases. The diphtheritic 21. Hitchner, S. B., C. A. Shea, and P. G. White. Studies on a serum
form of fowlpox with lesions in the trachea may simulate signs of neutralization test for the diagnosis of laryngotracheitis in chickens. Avian
ILT. Respiratory signs caused by mildly virulent strains of ILTV Dis. 2:258-269. 1958.
may be indistinguishable from other respiratory infections, such as 22. Hitchner, S. B., J. Fabricant, and T. J. Bagust. A fluorescent-antibody
mycoplasmosis and infectious bronchitis. Therefore tests such as study of the pathogenesis of infectious laryngotracheitis. Avian Dis. 21:185—
PCR, FA, IP become relevant confirmatory tests in the diagnosis of 194. 1977.
ILTV. 23. Hughes, C. S., and R. C. Jones. Comparison of cultural methods for
primary isolation of infectious laryngotracheitis virus from field material.
Avian Pathol. 17:295-303. 1988.
REFERENCES 24. Humberd, J., Riblet, S., Resurreccion, R.S., Brown, T.P., and Garcia, M.
Detection of infectious laryngotracheitis in formalin-fixed, paraffin
1. Abbas, F., J. R. Andreasen, Jr., and M W. Jackwood. Development of a embedded tissues by nested-PCR Avian Dis. 46: 64-74. 2002.
polymerase chain reaction and a nonradioactive DNA probe for infectious 25. Jordan, F. T. W., and R. C. Chubb. The agar gel diffusion technique in
laryngotracheitis virus. Avian Dis. 40:56-62. 1996. the diagnosis of infectious laryngotracheitis (ILT) and its differentiation
2. Abbas, F., and J. R. Andreasen, Jr. Comparison of diagnostic tests for from fowlpox. Res. Vet. Sci. 3:245-255. 1962.
infectious laryngotracheitis. Avian Dis. 40:290-295. 1996. 26. Keam L, York JJ, Sheppard M, Fahey KJ.Detection of infectious
3. Adair, B. M, D. Todd, E. R. McKillop, and K. Bums. Comparison of laryngotracheitis virus in chickens using a non-radioactive DNA probe.
serological tests for detection of antibodies to infectious laryngotracheitis Avian Dis.35:257-262. 1991.
virus. Avian Pathol. 14:461-469. 1985. 27. Keeler, C. L., Jr., J. W. Hazel, J. E. Hastings, and J. K. Rosenberger.
4. Alexander, H. S., and E. Nagy. Polymerase chain reaction to detect Restriction endonuclease analysis of Delmarva field isolates of infectious
infectious laryngotracheitis virus in conjunctival swabs from experimentally laryngotracheitis virus. Avian Dis. 37:418-426. 1993.
infected chickens. Avian Dis. 41:646-653. 1997. 28. Keller, L. H., C. E. Benson, S. Davison, andR. J. Eckroade. Differences
5. Andreasen J. R, J. Brown, J. R. Glisson and P. Villegas. Reprodicibility among restriction endonuclease DNA fingerprints of Pennsylvania field
of a virus-neutralization test for infectious laryngotracheitis virus. Avian isolates, vaccine strains, and challenge strains of Infectious laryngotracheitis
Dis. 34:185-192. 1989. virus. Avian Dis. 36:575-581. 1992.
6. Andreasen, J. R., J. Glisson, Jr., and P. Villegas. Differentiation of 29. Key, D. W., C. B. Gough, J. B. Derbyshire, and E. Nagy. Development
vaccine strains and Georgia field isolates of Laryngotracheitis virus by theis and evaluation of a non-isotopically labeled DNA probe for the diagnosis of
restriction endonuclease. Avian Dis. 34:646-656. 1990. infectious laryngotracheitis. Avian Dis. 38:467-474. 1994.
7. Armstrong, W. H. A slide smear technique for the diagnosis of 30. Kotiw M, Sheppard M, May JT, Wilks CR. Differentiation between
laryngotracheitis. Avian Dis. 3:80-84. 1959. virulent and avirulent strains of infectious laryngotracheitis virus by
8. Bauer, B.,J. E.Lohr and E. F. Kaleta. Comparison of commercial ELISA DNA: DNA hybridization using a cloned DNA marker. Vet Microbiol.
test kits from Australia and the USA with the serum neutralization test in 11(4):319-30. 1986
cell cultures for the detection of antibodies to the infectious laryngotracheitis 31. Leong, V.Y., J. R. Glisson, R. S. Resurreccion, and I-H. N. Cheng.
virus of chickens. Avian Pathology. 28:65-72. 1999. Infectious laryngotracheitis virus in commercial hens: A serological study
9. Clavijo, A., and E. Nagy. Differentiation of laryngotracheitis virus strains based on enzyme - linked immunosorbent assay. Avian Dis.38:304 - 307.
by polymerase chain reaction. Avian Dis. 41:241-246. 1997. 1994.
10. Chang, P. C., Y. L. Lee, J. H. Shien, and Η. K. Shien. Rapid 32. Linares, J. A., A. A. Bickford, G. L. Cooper, B. R. Charlton, and P. R.
differentiation of vaccine strains and field isolates of infectious Woolcock. An of infectious laryngotracheitis virus in California broilers.
laryngotracheitis virus by restriction fragment length polymorphism of PCR Avian Dis. 38:188-192. 1994.
products. J. Virol. Methods. 66:179-186. 1997. 33. Pirozok, R. P., C. F. Helmboldt, and E. L. Juhgherr. A rapid histological
11. Chang, P. C.,H. K.Shieh, J. H. Shien, and S. W. Kang. A homopolymer technique for the diagnosis of infectious avian laryngotracheitis. J. Am. Vet.
stretch composed of variable numbers of cytidine residues in the terminal Med. Assoc. 130:406-407. 1957.
repeats of infectious laryngotracheitis virus. Avian Dis. 44:125-131.2000. 34. Purcell, D. A. Histopathology of infectious laryngotracheitis in fowl
12. Fatunmbi, Ο. O., W. M Reed, D. L. Schwartz, and D. N. Tripathy. Dual infected by an aerosol. J. Comp. Pathol. 81:421^131. 1971.
infection of chickens with pox and infectious laryngotracheitis (ILT) 35. Robertson, G. M, and J. R. Egerton. Micro-assay systems for infectious
confirmed with specific pox and ILT DNA dot-blot hybridization assays. laryngotracheitis virus. Avian Dis. 21:133-135. 1977.
Avian Dis. 39:925-930. 1995. 36. Sander, J. E. and S. G. Thayer. Evaluation of ELISA titers to infectious
13. Garcia, M, and S. M Riblet. Characterization of Infectious laryngotracheitis. Avian Dis. 41:429-432.1997.
laryngotracheitis virus (ILTV) vaccine strains and field isolates: 37. Sellers, H. S., M Garcia, J. R. Glisson, T. P. Brown, J. S. Sander, and J.
demonstration of viral sub-populations within vaccine preparations. Avian S. Guy. Mild infectious laryngotracheitis in broilers in the Southeast. Avian
Dis. 45:558-566. 2001. Dis. 48:430-436.2004.
14. Graham, D. A., I. E. Mclaren, V. Calvert, D. Torrens, and B. M 38. Sevoian, M A. A quick method for the diagnosis of avian pox and
Meeham. RFLP analysis of recent Northern Ireland isolates of infectious infectious laryngotracheitis. Avian Dis. 4:474-476. 1960.
laryngotracheitis: comparison with vaccine virus and field isolates from 39. Schnitzlein, W. M, J. Radzevicius, and D. N. Tripathy. Propagation of
England, Scotland and Republic of Ireland. Avian Pathol. 29:57-62. 2000. infectious laryngotracheitis virus in avian liver cell line. Avian Dis. 38:211—
15. Goodwin, M. A, M A. Smeltzer, J. Brown, R. S. Resurreccion, and T. 217. 1994.
G. Dickson. Comparison of histopathology to the direct immunofluorescent 40. Shirley MW, Kemp DJ, Sheppard M, Fahey KJ. Detection of DNA
antibody test for the diagnosis of infectious laryngotracheitis in chickens. from infectious laryngotracheitis virus by colourimetric analyses of
Avian Dis. 35:389-391. 1991. polymerase chain reactions. J Virol Methods. 30(3):251 -9. 1990.

97
Deoki N. Tnpathy and Maricarmen Garcia

41. Sholz E., Porter RE. & Guo P. 1994. Differential diagnosis of 44. Williams, R A, M Bennett, J. M. Bradbury, R. M Gaskell, R C.
infectious laryngotracheitis from other avian respiratory disease by a Jones, and F. T.W. Jordan. Demonstration of sites of latency of infectious
simplified PCR procedure. J. Virol. Meth. 50:313-322. 1994 laryngotracheitis virus using the polymerase chain reaction. J. Gen Virol.
42. Trist, Η. M, S. G. Tyack, M A. Johnson, C. T. Prideaux, and M 73:2415-2420.1992.
Sheppard. Comparison of the genomic short regions of a vaccine strain (SA- 45. Williams, R. A., C. E. Savage, and R. C. Jones. A comparison of direct
2) and a virulent strain (CSW-1) of infectious laryngotracheitis virus (Gallid electron microscopy, virus isolataion and a DNA amplification method for the
Herpesvirus 1). Avian Dis. 40:130-139.1996. detection of avian infectious laryngotracheitis virus in fiels material. Avian
43. Wilks, C. R. and Kogan, V. G. An immunofluorescence diagnostic test Pathol. 23:709-720. 1994.
for avian infectious laryngotracheitis. Aus. Vet. Journal. 55:385 - 388. 1979. 46. York, J. J., and K. J. Fahey. Diagnosis of infectious laryngotracheitis
using a monocloal antibody ELISA. Avian Pathol. 17:173-182. 1988.

98
22
MAREK’S DISEASE
Patricia S. Wakenell and Jagdev M. Sharma

SUMMARY. Marek’s Disease (MD) is a herpesvirus-induced neoplastic disease of chickens. Because MD virus (MDV) is ubiquitous and
chickens acquire environmental infection early in life, they must be protected by vaccination in the hatchery. Diagnosis of infection versus
diagnosis of disease is critical as most birds are infected without developing clinical disease. The molecular structure of MDV has been
extensively examined. The virus has three serotypes, serotypes 1, 2, and 3. Among these, only viruses of serotype 1 have pathogenic
potential. MDV isolates can be categorized by serotype-specific monoclonal antibodies. Exposure to MDV results in persistent infection that
lasts for the life of the bird. Infected birds develop viremia as well as antibodies against the virus. Although initially the virus causes a lytic
infection in lymphoid cells, the virus persists in the birds by establishing a long-term latent infection of lymphocytes. Latently infected cells
minimally express viral antigens. Some latently infected cells may undergo transformation and induce tumors. Thus, MD outbreaks are
characterized by development of lymphoid tumors in viscera, skin, and nervous system. MD outbreaks may be associated with mortality,
immunosuppression, or excessive condemnation of carcasses during processing. Commercial chicken flocks can experience MD outbreaks at
any age. A number of vaccines are available to control MD in the field. These vaccines consist of serotype 1 (attenuated), 2, or 3 viruses
administered singly or in combination. Herpes virus of turkeys (HVT), a serotype 3 virus once used as a monovalent vaccine against MD, is
now usually mixed with serotype 1 and 2 viruses. Most commercial broiler chickens in the US receive MD vaccines by in ovo technology, in
which the vaccine is mechanically injected into eggs at embryonation day 18.
Agent Identification. Because most commercial chickens with or without MD have circulating MDV, mere virus isolation or detection of
antibody is not helpful in establishing a firm diagnosis. History of the flock, vaccination protocols, the nature of lesions, viral load,
expression of viral antigens, and the identity of the cells constituting tumors must be carefully considered.
A number of virus isolation strategies are available. Virus isolation in cell cultures is preferred. Because MDV is highly cell-associated,
viable cells removed from the tissues or tumors of infected chickens are generally co-cultivated with susceptible monolayer cells to isolate
the virus. Upon co-cultivation, the latently infected cells transmit the infection to permissive cells and induce herpesvirus-type cytopathology.
The specificity of the cytopathic effect can be confirmed by staining the cell culture monolayers by the immunofluorescence test using anti-
MDV antibodies. A number of molecular techniques have been used to identify viral genome in tissues. These include polymerase chain
reaction, dot-blot hybridization, and in situ hybridization. Assessment of MD viral load in tumors by quantitative PCR and expression of meq
on tumor cell surfaces are promising techniques for confirmation of MDV as the causative agent.
Serologic Detection in the Host. The serologic tests that can identify anti-MDV antibodies include immunofluorescence,
immunohistochemistry, enzyme-linked immunosorbent assay, the agar gel preciptin test, and virus neutralization test.

INTRODUCTION paralysis and/or dilation of the crop due to vagus nerve paralysis.
Affected nerves appear edematous, grayish and devoid of cross
Marek’s disease (MD) is important primarily in chickens, to a striations. Blindness is associated with lymphoid infiltration of the
much lesser degree in quail, and has been rarely observed in iris. Microscopically, MD lesions are composed of an infiltration by
turkeys, pheasants and jungle fowl. Turkeys and other species have a heterogenous population of mononuclear cells.
limited susceptibility. Attempts to infect mammals with MD virus
(MDV) have uniformly failed. The disease is caused by a SAMPLE COLLECTION
widespread, highly contagious, cell-associated, oncogenic
herpesvirus. In susceptible chickens, exposure to pathogenic MDV Since MDV is ubiquitous among chickens, mere demonstration of
may result in a progressive, debilitating disease that may result in the virus or antibody cannot be considered pathognomonic for the
high mortality, reduced egg production, and immunosuppression. cause of death or of an eportnic. The disease itself is not ubiquitous
The disease most commonly occurs in young, sexually immature and differentiation from other agents causing lymphoid tumors can
chickens 2-7 mo old, but can occur at virtually any age beyond 3 be problematic (19, 46, 51). For a positive diagnosis of MD in a
wk. Most commercial flocks are routinely vaccinated against MD. flock, one must consider the history of the flock and the presence of
Monovalent or multivalent vaccines are used. Although extensive characteristic gross and microscopic lesions in clinically sick and
use of vaccines has greatly reduced the incidence of disease dead chickens (52).
outbreaks compared to the pre-vaccination era, MD occasionally If virus isolation is attempted from commercial chickens raised
occurs in the presence of vaccine use. Many outbreaks in recent under field conditions, several MDV types may be isolated.
years have been caused by highly virulent strains of the virus that Chickens acquire infection with these viruses through live vaccines
emerged from less virulent predecessors. MDV vaccines protect or through environmental exposure. The isolated viruses may
against the disease but not against infection with the virus. Upon include herpesvirus of turkeys (HVT, designated serotype 3 MDV)
exposure to MDV, vaccinated or unvaccinated chickens become or serotype 1 or 2 MDV. Optimum sources are the same for
carriers of the virus and persistently shed MDV into the isolation of HVT as for isolation of MDV. Most suitable for virus
environment, thus making eradication difficult. MDV is not isolation are blood and cellular suspensions of spleen or tumor
transmitted vertically. The disease occurs throughout the world and tissue.
virtually all flocks are exposed to the causative virus.
Isolation from Blood
CLINICAL DISEASE Blood is preferred for virus isolations when euthanasia of the bird
is not permitted. In susceptible chickens inoculated experimentally
Clinical signs occur in chickens affected with MD but are of little at 1 day of age, viremia peaks at about 4 wk, and most chickens
help in establishing a diagnosis. The disease is characterized by remain persistently viremic for life (52). Infectivity in the blood is
proliferation of lymphoid cells in various tissues and organs, associated with the leukocyte (lymphocyte) fraction. The test
including peripheral nerves. Birds with visceral tumors are sample consists of 0.2 ml of whole blood or 2 x 106 buffy-coat cells
depressed and often cachectic prior to death. Birds with lymphoid suspended in cell-culture medium. The buffy-coat cells are the
infiltration of peripheral nerves may demonstrate asymmetric partial preferred sample.
99
Patricia S. Wakenell and Jagdev M Sharma

Isolation from Tumors Samples for Antibody


Solid tumor, spleen, kidney, or other tissues are removed Either serum or plasma can be used. Samples are stored and
aseptically, minced with scissors, washed several times with shipped at -20 C. Sera can be shipped at room temperature if a
phosphate-buffered saline (PBS) until reasonably clear of preservative (e.g. merthiolate, benzalkonium chloride or
erythrocytes, and then trypsinized once or twice to obtain single-cell chloroform) is added to prevent bacterial growth.
suspensions. The cells are pelleted by centrifugation, resuspended in
either PBS or cell-culture medium, and adjusted to give 2 x 106 PREFERRED CULTURE MEDIA AND SUBSTRATES
cells per 0.2 ml.
For best results, the cellular preparations should be used The best and most widely used substrate for virus isolation is
immediately after they are processed. If storage is required, the permissive cell cultures. In cell cultures, MDV produces cytopathic
following method is recommended: leukocytes or tissue cells effects (CPEs) typical of herpesviruses. Although serotypes 1, 2,
suspended at 2 x 107 cells/ml in cell-culture medium containing ΙΟ­ and 3 produce herpesvirus-type CPEs, it is possible to differentiate
Ι 5% dimethyl sulfoxide (DMSO) and 15-25% bovine fetal serum between serotypes by staining CPE-bearing cell cultures with
are dispensed in 1 ml amounts in glass or plastic cryovials. The serotype-specific monoclonal antibodies using an indirect
ampules are heavily insulated with paper towels and/or placed in a immunofluorescence test (26). An experienced eye may also be able
Styrofoam container and stored overnight at -70 C. Ampules are to differentiate the morphogenic characteristics of the plaques
then rapidly, to avoid any thawing, transferred to storage in liquid induced by each serotype. Serial passages of individual isolated
nitrogen. Cellular suspensions prepared as above can be held at plaques picked from an agar-overlaid culture containing a mixture
-196 C indefinitely without loss of viability. At the time of use, the of viruses may be used to prepare stocks of single serotypes. Virus
ampules are removed from the liquid nitrogen and quickly (< 2 min) can be isolated by inoculating test materials into embryonating
thawed in tepid water. The ampules are placed on wet ice and used chicken eggs. Isolation of virus by inoculation of susceptible
immediately. For shipping, ampules must be transferred to either chickens is cumbersome and not commonly done, although the
liquid nitrogen dewers (preferred) or dry ice without any thawing at identity of virus isolated in cell cultures or embryos can be
any stage of shipment or packaging. confirmed by reproducing typical disease in chickens.

Isolation of Cell-Free Virus Cell Culture


This procedure is not routine. However, for special purposes (e.g., Cell-Associated Virus. Although cell-associated MDV propagates
the serum-neutralization test or cloning the virus by plaque in most avian cell cultures tested, chicken kidney (CK) cells,
purification in cell cultures), cell-free MDV can be extracted from chicken embryo fibroblasts (CEFs) and duck embryo fibroblasts
feather tips or skin of infected chickens. For skin preparations, (DEFs) are the most commonly used. Most classical isolates of
feathers are clipped off at the surface of major feather tracts, skin serotype 1 MDV replicate poorly or become inconsistent in biologic
strips are removed, minced with scissors, and suspended in sucrose- characteristics when cultivated in CEFs (35). Thus CEFs are
phosphate-glutamine albumin (SGPA) buffer containing sodium generally considered unsuitable for primary virus isolation or
ethylenediamintetraacetate (EDTA). propagation for challenge of these viruses. Serotype 2 and 3 viruses
A 1:5 or 1:10 (w/v) suspension of skin strips in SGPA-EDTA grow readily in CEF. Serotype 1 viruses, passed first in susceptible
buffer is homogenized for 3-5 min and then sonicated for 2 min cells, also adapt to grow in CEF. A cellular suspension (0.2 ml
(four bursts of 30 sec each) with an ultrasonic oscillator with the containing 2 x 106 cells) is inoculated onto each monolayer culture
needle probe set at an intensity of 70 on the meter. The resulting of 24-hr primary CK cells or secondary CEF or DEF, grown in 15 x
suspension is centrifuged at 650 x g, and the supernatant is saved as 60 mm plastic tissue culture plates. CEF and DEF cultures can be
a source of cell-free virus (9). This preparation can be filtered maintained in flasks or roller bottles as well. However, CK
through a 0.45 mm filter pretreated with bovine fetal serum. maintenance for 5-6 days in flasks or roller bottles is difficult. The
Cell-free virus can also be isolated from feather tips. Feathers are cultures are incubated at 37-38 C in a humidified atmosphere
pulled from all major feather tracts, and 3-5 mm parts of the tips are containing 3-5% CO2. The culture medium is first renewed 24 hr
cut with scissors, diluted 1:5 (w/v) with SPGA-EDTA buffer, and post-inoculation (PI) and then on alternate days. For serotype 1, foci
sonicated for 2-3 min. The suspension is cleared by centrifugation of CPEs, usually termed plaques, are counted under a light
and tested for cell-free virus either with or without filtration through microscope 4-8 days PI in CK cells and 10-14 days PI in DEFs.
a 0.45 mm filter. Direct culture of kidney cells from infected chickens can be done
Cell-free virus preparations should be stored at -70 C in glass or (53). However, kidney cells from older birds may not culture well
plastic cryovials. For shipment, ampules should be packed in dry due to cellular age. Kidney cells from older birds can be used to
ice. infect CK monolayers as described.
Cell-Free Virus. Cell-free serotype 1 MDV propagates best in
Table 22.1: Sucrose-phosphate-glutamine-albumin buffer containing primary CK cell cultures. DEF and CEF are less susceptible than
sodium ethylene diaminetetraacetic acid (EDTA) for extraction and titration are CK cells.
of cell-free Marek’s disease virus. The buffer is sterilized by filtration and Virus is diluted in SPGA-EDTA buffer (Table 1). The SPGA-
has a pH of about 6.5
EDTA buffer enhances the virus titer several fold over that with a
Reagents Concentration Weight
standard diluent such as PBS.
Sucrose 0.2180 M 7.462 g
Primary CK cultures, 24-48 hr old, are drained and inoculated with
Monopotassium phosphate 0.0038 M 0.052 g the virus dilutions, two cultures per dilution using 0.2 ml of
Dipotassium phosphate 0.0072M 0.125 g inoculum per culture. After 30 min of adsorption at 37 C, the
L-Monosodium glutamate 0.0049 M 0.083 g medium is added to the cultures. Thereafter, the medium is changed
Bovine albumin powder 1.0% 1.000 g at 48 hr intervals. Because the virus suspension contains EDTA, a
EDTA 0.2% 0.200 g chelating agent, the inoculated cells tend to detach from the dish
Distilled H20 qs adA 100 ml during adsorption. Upon addition of fresh medium, however, the
A qs ad = sufficient quantity to make cells resettle and estabfish monolayers. Plaques are counted under
the light microscope 6-8 days PI, and the number of plaque-forming
units (PFU) per milliliter of virus is calculated.
100
Chapter 22 Marek’s Disease

Chicken Embryo Inoculation The molecular structure of MDV has been examined extensively.
Cell-Associated Virus. Primary virus isolation can be done also by The viral DNA for molecular analysis can be obtained by extraction
inoculating test material into the yolk sac of 4-day-old total cell DNA from cell cultures exhibiting extensive viral CPEi,
embiyonating chicken eggs (45) or intravenously in 10 to 11-day- feather tips, feather follicle epithelium (FFE), lymphoid tissues aid
old embryos. This procedure is rarely used. Virus isolation in cell brain. Separating virus-specific from cellular DNA can be difficult
cultures is preferred. Inoculated and uninoculated control eggs are because the densities of viral and cellular DNA are similar. Many
incubated at 37 C and candled daily. Embryos dying within 24 hr of methods are used but pulse field electrophoresis is recommended
inoculation are discarded, and those dying thereafter are examined (48). Cloning of MDV in bacterial artificial chromosomes (BAC)
for pocks on the chorioallantoic membrane (CAM). When the can generate larger amounts of MDV DNA (36). It has been
embryos are 18 days old, the test is ended by chilling the eggs suggested that viral DNA is integrated in the host DNA at multiple
overnight at 4 C. Positive isolations are characterized by the sites in chromosomes and the integration pattern varies in different
appearance of pocks on the CAM. One disadvantage of primary cell lines derived from in vivo MDV-induced tumors (16,24). The
virus isolation in embryos is that lymphoid cells present in virus- complete sequence has been obtained for the 3 serotypes
free inocula may also occasionally produce pocks on the CAM (5). (1,23,41,44). The genome consists of long and short unique regions
Futhermore, adaptation of MDV to cell cultures by serial passages flanked by terminal repeats (52). Endonuclease digestion patterns
reduces the pock response by embryos (38). differ substantially between the three serotypes (41). Cross
Cell-Free Virus. Inoculum is deposited in the CAM of 9 to 12- hybridization of DNA among the three serotypes under less
day-old embryonating eggs, and the CAM is examined for pocks 7 stringent conditions has shown that these viruses have a collinear
days PI. Cell-free virus can be inoculated intravenously into 10 to relationship. The three types of viruses share significant homology
11-day-old embryos (0.1 ml per embryo), and the CAM is examined at the DNA level and the structure of some of die dominant genes
for pocks 5 days later. such as gB, gC, gD, and gH is quite similar within all serotypes
(1,23,41,44). There are few structural differences between serotype
Chicken Inoculation 1 pathotypes (41).
A 0.2 ml test inoculum containing cell-associated or cell-free virus All three serotypes of MDV can cause latent infection in cells but
is injected intra-abdominally in 1 to 5-day-old chicks of a only unattenuated serotype 1 MDV has oncogenic potential.
susceptible genetic line lacking maternal antibodies to MDV or Latently infected cells have limited or no production of virus and
HVT. The chickens are raised in an isolation environment until they have fewer than 5 copies of the viral genome per cell. This number
are 6-8 wk old (50, 53). Because MDV is ubiquitous and highly may increase to 15 copies when the cell undergoes neoplastic
contagious, strict isolation must be maintained to prevent exposure transformation (20). Viral DNA of transformed cells is highly
to extraneous virus. Chickens that die before the end of the study methylated. Although the molecular basis of oncogenic
should be necropsied, and if gross lesions are absent, sections of transformation by MDV is not clear, the gene meq, containing a
nerves (vagus nerves and brachial and sciatic plexuses and nerves), basic leucine zipper resembling the jun/fos oncogene family, is a
gonads, spleen, kidney, liver and heart should be fixed in 10% strong candidate (25). Some of the transformed cells express meq
neutral buffered formalin and hematoxylin and eosin stained which can assist in the diagnosis of MDV-induced lymphomas
sections examined for histologic changes (30). At the end of the (19,24). The region containing 132 base pair tandem repeats flanks
study, survivors are bled for serum and examined for gross and the unique long portion of the virus genome. This region is
microscopic pathology. Polymerase chain reaction (PCR) can also important because the number of copies of 132 base pair repeats
be performed on tumor and spleen (2,3,4,21,40,55). Antibody can increases following cell culture attenuation of serotype 1 viruses,
be tested by the agar-gel precipitin (AGP) test, immunofluorescent and thus can be used as a marker for reduced oncogenicity (40).
(IF) test, or ELISA. Groups of chickens infected with the virus Identification by Inoculation in Cell Cultures, Chickens, or
develop lesions or antibody or both. Certain highly virulent strains Embryonating Eggs. MDV may be identified by inoculating the
may cause a high incidence of mortality within the first week of test sample in avian cultures (preferably CK cells or DEFs), 1-day-
infection (49,50). This early mortality is associated with extreme old susceptible chicks, or 9 to 12 day-old embryonating chicken
lymphoid cell depletion. eggs. Positive samples will induce herpesvirus-type CPE in cell
cultures, MD in chickens, and pocks on the CAM of eggs.
AGENT IDENTIFICATION Nonpathogenic isolates of MDV, such as serotypes 2 or attenuated
serotype 1, will not induce clinical MD in chickens, although the
Physiochemical and Molecular Characteristics of MDV. MDV chickens will develop specific antibodies.
is ether-sensitive, contains DNA, and belongs to the herpesvirus In cell cultures, MDV produces CPE characteristic of
group (52). The nuclear capsid is 90-100 nm in diameter and herpesviruses. The CPEs are sensitive to inhibitors of DNA
contains 162 capsomeres arranged in an icosahedral symmetry. The synthesis such as 5’-iodo-2’-deoxyuridine (IDU). Within 3-5 days
DNA of MDV is similar for all 3 serotypes and is a buoyant density of inoculation of monolayer cell cultures with MDV, plaques of
of 1.705 g/ml. The guanine plus cytosine content is different CPE appear. These consist of collections of rounded refractile cells.
amongst the 3 serotypes ranging from 43.9-53.6% for serotypes 1 With continued incubation, each plaque grows by including
and 2 and 47.6% for serotype 3.MDV remains highly cell- additional refractile cells at its periphery. In Giemsa stained
associated in vitro, and transfer of infection from one culture to preparations, the refractile cells appear as small or large
another depends on the use of viable infected cells. Destruction of polykaryocytes. Intranuclear inclusions (Cowdry type A) are readily
cell viability generally results in the loss of infectivity. However, seen in the areas of CPEs. MDV adapted to grow in cell cultures by
small amounts of cell-free virus can be extracted from cell cultures serial passage produces large plaques that often develop clear areas
by SPGA buffer (9). (holes) in the center.
Cell-free MDV obtained from extracts of skin or feather pulp The morphology of CPE may also vary with the type of cell
retains its titer for several months at -60 C but not at -20 C. Virus is culture. For instance, in DEF cultures, a focus of CPEs consist of
inactivated in 2 wk at 4 C, in 4 days at 22-25 C, in 18 hrs at 37 C, in round as well as fusiform cells. In CK cell cultures, fusiform cells
30 min at 56 C, and in 10 min at 60 C. The virus can withstand at are rare. The serotype of the CPE inducing virus can be confirmed
least four freeze-thaw cycles and 10 min of ultrasonic vibration. A by staining infected cultures by the IF test using serotype specific
pH of lower than 4 or higher than 10 readily inactivates the virus. monoclonal antibodies.

101
Patricia S. Wakenell and Jagdev M. Sharma

Samples containing oncogenic serotype 1 MDV and HVT may not Virus Detection by Molecular Techniques. These procedures are
induce MD in chickens because of the protective effect of HVT. To not routinely used for diagnosis of MD but have recently become
identify MDV in such samples, inoculated chickens should be popular research tools. Polymerase chain reaction (PCR) technology
maintained in direct contact with uninoculated hatchmates. If can be used to identify viral nucleic acid in productively and
oncogenic serotype 1 MDV is present, the contact exposed chickens latently infected cells as well as in cells from lymphomas. The most
will not be protected against MD by this virus. If the test samples commonly used primers amplify the 132 base pair repeat sequences
contain serotype 1 and serotype 2 MDV, neither the inoculated (4,21,40,55), although primers to amplify other regions of the
chickens nor the contact exposed chickens will develop MD, genome may also be used (4,19,21). The 132 base pair repeat
although both groups of chickens will develop anti-MDV antibody primers can detect virulent or attenuated serotype 1 MDV.
and persistent viremia with both serotypes. Quantitative PCR has been used to detect MDV in latently infected
Several pathotypes of serotype 1 MDV have been isolated from birds (6,33). Real-time PCR has been used to quantify MDV in
chicken flocks. Periodically, highly virulent isolates of serotype 1 tumors (19). Briefly, DNA was extracted from tumors using a
virus have emerged that have increasingly resisted protection by Puragene DNA Isolation kit (Gentra System, Minneapolis, MN).
conventional vaccines. Thus, serotype 1 MDV isolates recovered Samples were amplified using primers for glycoprotein B (gB) and
from field flocks may vary greatly in virulence. The isolates may be glyceraldehydes-3-phosphate dehydrogenase (GAPDH). The
mildly virulent (mMDV), virulent (vMDV), very virulent GeneAmp 5700 (Applied Biosystems, Foster City, CA) was used to
(wMDV), or very very virulent (w+MDV) (49). These pathotypes amplify the samples in a 25 μΐ PCR reaction containing 50 ng DNA,
can be differentiated by chick inoculation (50,51) although the 0.2pm of each primer and SYBR green master mix (Applied
process is very tedious and should be attempted only by well Biosystem, Warrington, UK). The reaction was cycled 40 times.
equipped laboratories proficient in tissue culture techniques and The threshold cycle was determined for each PCR reaction by
having good animal isolation facilities. Briefly, the virus is first establishing a fixed threshold. The relative number of copies of gB
isolated from the test sample in cell cultures. The isolated virus is was compared to that of GAPDH, the reference control (19). High
cloned into a population that contains pure serotype 1 virus without loads of MDV DNA in tumors were considered diagnostic of MDV
detectable contamination with other serotypes of MDV or other as the causative agent.
common viruses that may have been present in the test sample. It is
imperative that the test sample is free of extraneous viruses that Strain Variability
might influence the behavior of the test sample (50,51). The Numerous isolates of MDV have been described. Serologically,
pathotype of the isolated virus is determined by inoculation in MDV isolates can be differentiated into sefbtypes 1 and 2. Serotype
chickens that are either unvaccinated or have been immunized with 1 contains pathogenic isolates and their attenuated forms, and
monovalent HVT, bivalent HVT + SB-1 (serotype 2 MDV) or serotype 2 contains apathogenic isolates. Serotype 1 isolates have
Rispens (attenuated serotype 1) vaccines. All pathotypes cause been differentiated primarily on the basis of pathogenicity for
clinical MD in unvaccinated chickens with the exception of certain chickens. Thus, they fall into one of four general categories:
highly inbred resistant lines. mMDV is protected by HVT, w mMDV, vMDV, wMDV, or w+MDV. Although isolates of
MDV is protected by HVT/SB-1 and w+MDV is fairly well serotypes 1 and 2 cross-react, isolates within each serotype have
protected by Rispens. stronger serologic cross-reactions than the isolates between the two
Direct Demonstration of Viral Antigens in Tissues. Viral serotypes. Highly virulent isolates of MDV (wMDV or w+MDV)
antigens can be detected only in tissues that are productively are of interest because HVT is not an effective vaccine against these
infected with MDV. The FA test using monoclonal or polyclonal isolates, and bivalent vaccines containing HVT and serotype 2
ant-MDV antibodies may be used. Fresh tissues are frozen at -70 C MDV, certain attenuated serotype 1 vaccines or trivalent vaccines
or -196 C and cut at 6-8gm thickness in a freezing microtome. At containing HVT along with serotypes 1 and 2 MDV must be used to
the time of staining, tissue sections are thawed, mounted on glass protect flocks. Diagnosis of infection with wMDV in a flock is a
microscope slides, and stained by the direct or indirect FA test. complicated process that has been detailed previously (50). Using
Viral antigen in the feather follicles may also be detected by real-time reverse transcriptase PCR (54) and/or quantitative PCR to
reacting feather pulp with anti-MDV polyclonal antibody in an determine viral load (19) show promise for differentiation of
AGP test. pathotypes. Although expansions of the 132 base pair repeat have
The immunohistochemistry (IHC) test using an avidin-biotin- indicted attenuation, these are not related to virulence (42).
peroxidase complex (Vecta-stain ABC kit, Vector Laboratories, Mutations in genes such as meq (10) and the terminal and internal
Burlingame, CA) has been done to detect meq, CD30, MATSA, repeat long regions of MDV may also contribute to differences in
p53, pp38 and methyl-3-cytosine in tumors (19). Expression of meq virulence (43).
was considered specific for MD lymphomas. Briefly, the IHC
staining of meq and CD30 were amplified with the tyramide signal Characteristics of Vaccine Strains
reaction using the TSA Biotin System Kit (PerkinElmer Life Although HVT was once the most commonly used vaccine against
Science, Boston, MA). The monoclonal antibody (mAb) for meq MD, many flocks now receive bivalent or trivalent vaccines.
(27) was used at a dilution of 1:1000, mAb for pp38 (Hl9.47, 14) Bivalent vaccines contain HVT and a serotype 2 MDV (e.g., SB-1),
was used at a 1:3200 dilution, mAb for AV37, specific for chicken whereas trivalent vaccines may contain HVT, attenuated serotype 1
CD30 (7) was used at a dilution of 1:25, the mAb for MATSA MDV (e.g., CV1988), and serotype 2 MDV (e.g., SB-1). In some
(14B367 Lee, unpublished data) was used at 1:2000, the mAb for parts of the world, serotype 1 MDV is used alone as an effective
methyl-3-cytosine (WWR International Oncogene Research vaccine against MD.
Product, San Diego, CA) was used at 1:25 and the mAb for p53 is Recombinant MD vaccines have been developed that are quite
specific for mouse p53 but cross reacts with chicken p53 (Pab 240, similar to HVT/SB-1 in protecting chickens against virulent MDV.
18) was used at a dilution of 1:25. The recombinants have been constructed by inserting immunogenic
Molecular procedures can be used to detect the presence of viral genes of MDV, particularly gB, in fowlpox virus or HVT (28,34).
DNA in tissues or cells in which viral antigens are not expressed. Recombinant vaccines containing immunogenic genes from
Dot-blot and in situ hybridization have been used to detect viral multiple disease agents such as MDV and Newcastle disease virus
antigen in feather tip extracts and to localize viral antigens in have also been developed (32).
tissues, respectively (15,17,22). Upon vaccination, chickens remain asymptomatic, although they
develop persistent viremia with all viruses present in the vaccine.
102
Chapter 22 Marek’s

Vaccinated chickens become resistant to tumor formation by the medium, whereas the other lines are minor, produced by
virulent MDV but not to infection with MDV. Thus, the vaccinated antigens intimately associated with infected cells.
chickens become persistently viremic with the vaccine virus(es) as
well as field MDV and can shed virulent MDV, although at a Fluorescent Antibody Test
reduced rate. The best way to ascertain that the flock has been The FA test can be used to detect antibodies if known antigen is
properly vaccinated against MD is to isolate the vaccine virus(es) available (indirect FA) or - the reverse - to detect antigen if know®
from vaccinated chickens, HVT replicates readily in avian cell antibody is available (direct FA). Techniques for both direct and
cultures (8), although CPEs are slightly different from that of MDV. indirect FA tests for the avian system have been reviewed (31). Far
In contrast to MDV plaques, which develop slowly and consist of the direct test, globulin extracted from serum of chickens exposed
rounded clusters rarely extending more than 0.5-1 mm in diameter, to MDV is labeled with fluorescein isothiocyanate. For the indirect
the plaques produced by HVT in CK cells appear early (in 2-3 days) test, labeled anti-chicken gamma globulin is available from several
and consist of large syncytia surrounding a clear central area. HVT commercial sources. Mouse monoclonal anti-MDV antibodies have
plaques grow rapidly, reaching a diameter of 1.5-3 mm in about 10 been developed that may be used in the indirect FA test (26).
days. HVT and MDV plaques can be distinguished in several avian The antigen is prepared in monolayer cultures of CK cells grown
cells cultures, but most clearly in CK cell cultures. The identity of on glass coverslips. Confluent monolayers are inoculated with
the plaques induced by the two viruses may be confirmed by enough MDV to give numerous separated plaques. When the
staining CPE-monoclonal antibodies in an FA test. Serotypes 1 and earliest distinct plaques appear (before CPE become confluent), the
2 antibody will react only with MDV plaques and serotype 3 coverslips are washed once with PBS and then fixed for 5 min in
antibody reacts with HVT plaques. acetone at 4 C. Fixed coverslips can be stored at -20 C and used
Most MD vaccines consist of a suspension of viable, virus-infected within several weeks.
CEF (wet vaccine), although lyophilized, monovalent cell-free HVT
vaccine (diy vaccine) is also available. Serotypes 1 or 2 MDV are Enzyme-Linked Immunosorbent Assay
not available in the lyophilized form. Wet vaccine is supplied in In this test, wells of a 96-well microtiter plate are coated with
frozen, sealed glass ampules. The accompanying handling MDV-infected CEFs or DEFs (11). Monolayer cells cultured in
instructions should be followed carefully. The vaccine should be 100-mm plates are infected with a massive dose of cell-associated
thawed just before used and immediately diluted to the proper dose MDV. When more than 75% cells show CPEs, the infected cells are
level. Concentrations of DMSO used to protect cells during freezing trypsinized and suspended in PBS and used to coat the wells of the
are highly toxic to cells at room temperature (25-30 C). microtiter plate. Alternatively, the cells may be suspended in tissue­
culture medium containing 10% calf serum and 10% DMSO and
SEROLOGIC DETECTION IN THE HOST stored in liquid nitrogen for later use. At the time of use, the frozen
cells are thawed and washed twice with PBS to remove the DMSO
Chickens infected with MDV develop antibody that persists for before coating the wells. Each microtiter well receives 5 x 104
life. Several serologic tests may be used to quantify serum antibody, infected cells (containing about 1.7 x 103 PFU) in 0.1 ml PBS. After
however, usefulness in commercial flocks is limited since all flocks the plates are low-speed centrifuged (1000 rpm) for 10 min, the
have been vaccinated or exposed. Serologic assays are useful for supernatant is discarded and the cells are allowed to dry at room
evaluating chickens with no known prior exposure to MDV vaccine temperature. Antigen-coated plates may be stored at 4 C or -20 C
or field strains (backyard flocks, international flocks). for at least 3 mo without loss of reactivity. The stored plates should
be washed three times with PBS containing 0.1% Tween 80 before
Agar-Gel Precipitin Test use.
In the AGP test, serum is reacted with MD antigens in an agar Duplicate samples of 0.1 ml of test sera and known MD antibody­
medium. Commercial reagents are available. The antigen for this positive and -negative sera diluted in 3% bovine serum albumin are
test is prepared by propagating MDV (of low cell-culture-passage placed in the wells of the microtiter plates. After 1 hr at 37 C, the
level) in CK cell or CEF cultures (12). When CPEs are confluent, wells are washed with PBS and 0.1 ml of a 1:3200 dilution of
the cells are detached from the culture vessel either by scraping the affinity-purified goat anti-chicken immunoglobulin G (IgG)-
vessel with a rubber policeman or trypsinizing and the cells are peroxidase conjugate (Synbiotics, San Diego, CA) is added to each
suspended in PBS or culture medium without tryptose phosphate well. After 1 hr at 37 C, the wells are washed four times and to each
broth (tryptose phosphate broth in the antigen may produce well is added 0.1 ml of substrate containing one volume of 0.05%
nonspecific precipitin bands) at a concentration of about 1 x 107 H2O2 and nine volumes of purified 5-aminosalicylic acid (1 mg per
cells/ml. This suspension is then freeze-thawed three times and used ml in 0.02 M phosphate buffer [pH6.0]). After 30 min at room
as antigen. Feather pulp extracted by squeezing the tips of feathers temperature, absorbance is read at 490 nm wavelength using any
plucked from major feather tracts of infected chickens also has been commercially available ELISA reader. Sera showing an absorbance
used as antigen. For optimum precipitation, a 1% suspension of reading of at least three times that of known antibody-negative sera
Noble agar is made in NaK phosphate buffer (pH 7.4) containing are considered positive. Cheng et al. (11) considered a serum
8% NaCl. About 3 ml of suspension is poured onto a glass positive if the absorbance at a 1:400 dilution was 0.20 units or
microscope slide pretreated with a film of 1:5 dilution of agar higher. The titer of antibody was the reciprocal of the serum
suspension. When the agar solidifies, one central and six peripheral dilution with an absorbance of 0.20 units.
equidistant wells, each 3 mm in diameter and about 3.5 mm apart
are cut with a template or other suitable borer. The central well is Virus Neutralization Test
filled with antigen, and the peripheral wells are filled with serum. This test is used to detect neutralizing antibody in the serum or
To detect nonspecific precipitin bands and enhance weak reactions plasma of infected chickens. Because the cell-free preparations of
each well containing test serum should be adjacent to one MDV prepared from skin or feather-follicle extracts of MDV-
containing known MD-specific serum. Reagents should be diffused infected chickens are generally low in titer, the kinetics of
for 72 hr in a moist chamber at either 37 C or at room temperature. neutralization have not been adequately studied. The test should be
Positive test serum should form a line of identity with the line conducted with cell-free virus suspension in SPGA-EDTA buffer
produced by MD-specific serum. Certain sera may produce up to with a titer of about 103 PFU/ml. One part of twofold or 10-fold
six precipitin lines. The most prominent line, referred to as the A (or serum or plasma dilution and four parts of virus suspension are
gC) line, is produced by the antigen released from infected cells into mixed and incubated for 30 min at 37 C or at room temperature.
103
Patricia S. Wakenell and Jagdev M Sharma

Known positive and negative serum controls should be included in 7. Burgess, S. C., P. Kaiser, and T. F. Davison. A monoclonal antibody that
the test. Each of duplicate cultures of 24-hr-old primary CK cells is recognizes the chicken homologue of CD30, a tumor antigen in Marek’s
then inoculated with 0.2 ml of the serum-virus mixture and disease. In: K. A. Schat (Ed). Current Progress in Avian Immunology. Am.
Assoc. Avian Path. Kennett Square, PA. pp. 232-232. 2001.
absorbed for 30 min at 37 C and fresh medium is then added to the
8. Calnek, B. W., C. Garrido, W. Okazaki, and I. V. Patrascu. In vitro
cultures. Thereafter, medium is changed on alternate days. Plaques methods for assay of turkey herpesvirus. Avian Dis. 16:52-56. 1972.
are counted 6-8 days after inoculation. The titer of the serum is the 9. Calnek, B. W., S. B. Hitchner, and Η. K. Adldinger. Lyophilization of
reciprocal of the serum dilution that causes at least 50% reduction cell-free Marek’s disease herpesvirus and a herpesvirus of turkeys. Appl.
in virus titer (obtained with negative serum control). Microbiol. 20:723-726. 1970.
10. Chang, K. S., K. Ohashi, and M Onuma. Diversity (polymorphism) of
Vaccine Delivery. Within the last decade, a dramatic change has the meq gene in the attenuated Marek’s disease virus (MDV) serotype 1 and
occurred in the method of MD vaccine delivery in commercial MDV-transformed cell lines. J. Vet. Med. Sci. 64:1097-1101. 2002.
11. Cheng, Y. Q., L. F. Lee, E. Smith, and R. L. Witter. An enzyme-linked
chicken flocks. Previously, MD vaccines were administered
immunosorbent assay for the detection of antibodies to Marek’s disease
subcutaneously in newly hatched chickens in the hatchery. Most of virus. Avian Dis. 28:900-911. 1984.
the major hatcheries in the U.S.A. now use in ovo technology to 12. Chubb, R. C., and A E. Churchill. Precipitating antibodies associated
vaccinate chickens against MD (37,39). In this technology, the with Marek’s disease. Vet. Rec. 83:4-7. 1968.
vaccines are injected in eggs at 17-18 days of embryonation using 13. Crespo, R., P. R. Woolcock, A. M Fadly, C. Hall, and H. L.
multiple-head injection machines. Site of vaccine delivery is critical Shivaprasad. Characterization of T-cell lymphomas associated with an
for MDV vaccine efficacy (47). This method has substantially outbreak of reticuloendotheliosis in turkeys. Avian Pathology 31:355-361.
reduced the labor cost associated with posthatch vaccination. 2002.
14. Cui, Z. Z., D. Yan, and L. F. Lee. Marek’s disease virus gene clones
encoding virus-specific phosphorylated polypeptides and serological
DIFFERENTIATION FROM CLOSELY RELATED AGENTS characterization of fusion proteins. Virus Genes 3:309-322. 1990.
15. Davidson, I., M Malkinson, C. Strenger, and Y. Becker. An improved
Clinical MD is often confused with lymphoid leukosis. The main ELISA method using a streptavidin-biotin complex, for detecting Marek’s
source of confusion is the similarity of the gross visceral tumors disease virus in feather tips of infected chickens. J. Virol. Methods 14:237-
produced by the two diseases. Detection of cellular antigens with 241. 1992.
monoclonal antibodies may be used to distinguish tumor cells of 16. Delecluse, H.-J., and W. Hammerschmidt. Status of Marek’s disease
MD and lymphoid leukosis (13,19,29,51). Because virus in established lymphoma cell lines: herpesvirus integration is
common. J. Virol. 67:82-92. 1993.
reticuloendotheliosis virus may cause nerve enlargement or 17. Endoh, D., Y. Kon, M Hayashi, T. Morimura, K. O. Cho, T. Iwasaki,
lymphomas under experimental conditions, it also may be confused and F. Sato. Detection of transcripts of Marek's disease virus serotype 1
with MD. Firm diagnosis depends on flock history, distribution and ICP4 homologue (MDV ICP4) by in situ hybridization. J. of Vet. Mei Sci.
histologic appearance of lesions, identity of cells constituting 58:969-975. 1996.
tumors, identification of viral antigens in tumor cells, and isolation 18. Gannon, J. V., R. Greaves, R. Iggo, and D. P. Lane. Activating
of the etiologic agent (see Chapter 35 on oncornaviruses). Viral mutations in p53 produce a common conformational effect. A monoclonal
antigens may be identified by microscopy techniques using antibody specific for the mutant form. Euro. Mol. Biol. Org. J. 9:1595-1602.
immunofluorescence or immunohistochemhical staining or by 1990.
19. Gimeno, I. M, R. L. Witter, A. M Fadly, and R. F. Silva. Novel criteria
molecular assays such as PCR to detect viral nucleic acid. for the diagnosis of Marek’s disease virus-induced lymphomas. Avian
Another common avian herpesvirus that should differentiate from Pathology 34(4):332-340. 2005.
MDV is infectious laryngotracheitis virus (ILTV). Clinically, ILTV 20. Gimeno, I. M, R. L. Witter, and A. Miles. Marek’s disease (CDRom)
causes a respiratory disease in chickens, and the lesions are more or Slide set. Am. Assoc. Avian Path. Athens, GA. 2004.
less restricted to the upper respiratory tract, without involvement of 21. Handenberg, K. J., O. L. Nielsen, and P. H. Jorgensen. The use of
the nervous system. ILTV produces pocks that are indistinguishable serotype 1- and serotype 3-specific polymerase chain reaction for the
from those produced by MDV on the CAM of embryonated eggs. In detection of Marek’s disease virus in chickens. Avian Pathology 30:243-
avian cell cultures, ILTV becomes cell-free and grows much faster 249. 2001.
22. Holland, M S., C. D. Mackenzie, R. W. Bull, and R. F. Silva. A
than MDV. The CPE of ILTV becomes detectable 8-10 hr after comparative study of histological conditions suitable for both
inoculation and consists of rapidly progressing syncytia formation. immunofluorescence and in situ hybridization in the detection of herpesvirus
Plaques induced by ILTV and MDV may be differentiated by and its antigens in chicken tissues. J. Histochem. Cytochem. 44:259-265.
staining with virus-specific antibodies. 1996.
23. Izumiya, Y, Η. K. Jang, M Ono, and T. Mikami. A complete genomic
REFERENCES DNA sequence of Marek’s disease virus type 2, strain HPRS24. In: K. Hirai
(Ed.). Current Topics in Microbiology and Immunology. Springer-Verlag,
1. Afonso, C. L., E. R. Tulman, Z. Liu, L. Zsak, D. L. Rock, and G. F. Berlin. 255:191-222. 2001.
Kutish. The genome of turkey herpesvirus. J. Virol. 75:971-978. 2001. 24. Jarosinski, K. W., B. K. Tischer, S. Trapp, andN. Osterrieder. Marek’s
2. Baigent, S., V. Nair, and R. Currie. Real-time quantitative PCR for disease virus: Lytic replication, oncogenesis and control. Expert. Rev.
Marek’s disease vaccine virus in feather samples: applications and Vaccines 5(6):761-772. 2006.
opportunities. Dev. Biol. (Basel). 126:271-81. 2006. 25. Kung, H. J., L. Xia, P. Brunovskis, D. Li, J. L. Liu, and L. F. Lee. Meq:
3. Baigent, S. J., L. J. Pelherbridge, K. Howes, L. P. Smith, R. J. Currie, and An MDV-specific bZIP transactivator with transforming properties. In: K.
V. K. Nair. Absolute quantification of Marek’s disease virus genome copy Hirai (Ed.). Current Topics in Microbiology and Immunology. Springer-
number in chicken feather and lymphocyte samples using real-time PCR. J. Verlag, Berlin. 255:245-260. 2001.
Virol. Methods 123(l):53-64. 2005. 26. Lee, L. F., X. Liu, and R. L. Witter. Monoclonal antibodies with
4. Becker, Y., Y. Asher, E. Tabor, I. Davidson, M Malkinson, and Y. specificity for three different serotypes of Marek’s disease viruses in
Weisman. Polymerase chain reaction for differentiation between pathogenic chickens. J. Immunol. 130:1003-1006. 1983.
and nonpathogenic serotype 1 Marek’s disease virus (MDV) and vaccine 27. Liu, J. L., L. F. Lee, and H. J. Kung. Biological properties of the
viruses of MDV serotypes 2 and 3. J. Virol. Methods 40:307-322. 1992. Marek’s disease latent protein MEQ: Subcellular localization and
5. Biggs, P. M, and B. S. Milne. Use of the embryonating egg in studies on transforming potential. In: R. F. Silva, Η. H. Cheng, P. M Coussens, L. F.
Marek’s disease. Am. J. Vet. Res. 32:1795-1809. 1971. Lee, and L. F. Velicer (Eds.). Current Research on Marek’s Disease. Am.
6. Bumstead, N. J., Silliboume, M. Rennie, N. Ross, and F. Davison. Assoc. Avian Path. Kennett Square, PA pp. 271-277. 1996.
Quantification of Marek’s disease virus in chicken lymphocytes using the 28. Nazerian, K., L. F. Lee, N. Yanagida, and R. Ogawa. Protection against
polymerase chain reaction with fluorescence detection. J. Virol. Methods Marek’s disease by fowlpox virus recombinant expressing the glycoprotein
65:75-81. 1997. B of Marek’s disease virus. J. Virol. 66:1409-1413. 1992.

104
Chapter 22 Marek’s Disease

29. Neumann, U., and R. L. Witter. Differential diagnosis of lymphoid 42. Silva, R. F., S. M Reddy, and B. Lupiani. Expansion of a unique region
leukosis and Marek’s disease by tumor associated criteria. I. Studies on in the Marek’s disease virus genome occurs concomitantly with attenuation
experimentally infected chickens. Avian Dis. 23:417-425. 1979. but is not sufficient to cause attenuation. J. Virol. 78:733-740. 2004.
30. Payne, L. N., and P. M Biggs. Studies on Marek’s disease. Π. 43. Spatz, S. J., and R. F. Silva. Polymorphisms in the repeat long regions
Pathogenesis. J. Natl. Cancer Inst. 39:281-302. 1967. of oncogenic and attenuated pathotypes of Marek’s disease virus 1. Virus
31. Purchase, H. G. Fluorescent-antibody techniques in avian research. Genes. Sep. 9. 2006.
Avian Dis. 17:213-226. 1973. 44. Tulman, E. R., C. L. Afonso, Z. Lu, L. Zsak, D. L. Rock, and G. F.
32. Reddy, S. K., J. M Sharma, J. Ahmad, D. N. Reddy, J. K. McMillen, S. Kutish. The genome of a very virulent Marek’s disease virus. J. Virol.
M Cook, M A. Wild, and R. D. Schwartz. Protective efficacy of 74:7980-7988. 2000.
recombinant herpesvirus of turkeys as an in ovo vaccine against Newcastle 45. Von Bulow, V. Diagnosis and certain biological properties of the virus
and Marek’s disease in specific-pathogen-free chickens. Vaccine 14:469- of Marek’s disease. Am. J. Vet. Res. 32:1275-1288. 1971.
477. 1996. 46. Wakenell, P. S. An overview of problems in diagnosis of neoplastic
33. Reddy, S. M, R. L. Witter, and I. M Gimeno. Development of a diseases of poultry. In: Proceedings of the American Association of Avian
quantitative-competitive polymerase chain reaction assay for serotype 1 Pathologists Tumor Virus Symposium. Pp 1-7. 1997.
Marek’s disease virus. Avian Dis. 44:770-775. 2000. 47. Wakenell, P. S., T. Bryan, J. Schaeffer, A. Avakian, C. Williams, and C.
34. Ross, L. J. N., Μ M Binns, P. Tyers, J. Pastorek, V. Zelnik, and S. Whitfill. Effect of in ovo vaccine delivery route on herpesvirus of
Scott. Construction and properties of turkey herpesvirus recombinant turkeys/SB-1 efficacy and viremia. Avian Dis. 46:274-280. 2001.
expressing the Marek’s disease virus homolog of glycoprotein B of herpes 48. Wilson, M R., and P. M Coussens. Purification and characterization of
simplex virus. J. Gen. Virol. 74:371-377. 1993. infectious Marek’s disease virus genomes using pulsed field electrophoresis.
35. Schat, K. A. Isolation of Marek’s disease virus: Revisited. Avian Virology 185:673-680. 1991.
Pathology 34(2):91-95. 2005. 49. Witter, R. L. Increased virulence of Marek’s disease virus field isolates.
36. Schumacher, D. Β., Β. K. Tischer, W. Fuchs, and N. Osterrieder. Avian Dis. 41:149-163. 1997.
Reconstruction of Marek’s disease virus serotype I (MDV-1) from DNA 50. Witter, R. L., B. W. Calnek, C. Buscaglia, I. M Gimeno, and K. A
cloned as a bacterial artificial chromosome and characterization of a Schat. Classification of Marek’s disease viruses according to pathotype:
glycoprotein B negative MDV-1 mutant. J. Virol. 74:11088-11098. 2000. Philosophy and methodology. Avian Pathology 34(2): 75-90. 2005.
37. Sharma, J. M., and B. R. Burmester. Resistance to Marek’s disease virus 51. Witter, R. L., I. M Gimeno and A. M Fadly. Differential diagnosis of
at hatching in chickens vaccinated as embryos with the turkey herpesvirus. lymphoid and myeloid tumors in chickens (CD-ROM), set 27. Am. Assoc.
Avian Dis. 26:134-'49. 1982. Avian Pathologists, Athens, GA. 2005.
38. Sharma, J. Μ, B. D. Coulson, and E. Young. Effect of in vitro 52. Witter, R. L., and K. A. Schat. Marek’s disease. In: Diseases of Poultry,
adaptation of Marek’s disease virus on pock induction on the chorioallantoic 11th ed. Y. M Saif, H. J. Bames, J. R. Glisson, A. M Fadly, L. R.
membrane of embryonated chicken eggs. Infect. Immun 13:292-295. 1976. McDougald, and D. E. Swayne (Eds.). Iowa State University Press, Ames,
39. Sharma, J. M, and R. L. Witter. Embryo vaccination against Marek’s LA. Pp. 407-465. 2003.
disease with serotypes 1, 2, and 3 vaccines administered singly or in 53. Witter, R. L., J. J. Solomon, and G. H. Burgoyne. Cell culture
combination. Avian Dis. 27:453-463. 1983. techniques for primary isolation of Marek’s disease associated herpes
40. Silva, R. F. Differentiation of pathogenic and nonpathogenic serotype 1 viruses. Avian Dis. 13:101-118. 1969.
Marek’s disease viruses (MDVs) by the polymerase chain reaction 54. Yunis, R., K. W. Jarosinski, and K. A. Schat. Association between rate
amplification of the tandem direct repeats within the MDV genome. Avian of viral genome replication and virulence of Marek’s disease herpesvirus
Dis. 36:521-528. 1992. strains. Virology 328:142-150. 2004.
41. Silva, R. F., L. F. Lee, and G. F. Kutish. The genomic structure of 55. Zhu, G. S., T. Ojima, T. Hironaka, T. Ihara, N. Mizukoshi, A. Kato, S.
Marek’s disease virus. In: K. Hirai (Ed.). Current Topics in Microbiology Ueda, and K. Hirai. Differentiation of oncogenic and nononcogenic strains
and Immunology. Springer-Verlag, Berlin. 255:143-158. 2001. of Marek’s disease virus type 1 by using polymerase chain reaction DNA
amplification. Avian Dis. 36:637-645. 1992.

105
23
DUCK VIRUS ENTERITIS
Peter R. Woolcock

SUMMARY. Duck virus enteritis (DVE) is caused by a herpesvirus of the subfamily Alphaherpesvirinae of the family Herpesviridae. No
evidence exists of antigenic variation. Infections have only been reported in ducks, geese, and swans. Birds of all ages are susceptible to
infection and mortality may be high, but the severity of clinical signs varies with the species, sex, and age of the infected birds. Lesions seen
at necropsy are typical of vascular damage.
Agent Identification. Diagnosis of DVE is confirmed by isolation and identification of the virus. Polymerase chain reactions (PCR) have
been developed to detect viral DNA in infected tissues.
Serologic Detection in the Host. Serologic detection of infection is only of importance in non-commercial waterfowl, and may be used to
determine prior exposure of migratory and non-migratory birds to DVE virus.

INTRODUCTION SAMPLE COLLECTION

Duck virus enteritis (DVE) is an acute, sometimes chronic, Carcasses or tissues collected for virus isolation from dead ducks
contagious viral infection occurring naturally only in ducks, geese, should be chilled at 4 C immediately; if they cannot be delivered to
and swans, all of which are members of the family Anatidae of the a diagnostic laboratoiy within 24 hr, the tissues should be frozen,
order Anseriformes. DVE may also be referred to as duck plague, preferably at -70 C or lower, and stored until they can be shipped on
anatid herpes, eendenpest, entenpest, and peste du canard. The dry ice (freezing at temperatures warmer than -70 C may result in
disease has been reported in Asia, Europe, and North America. The loss of viability of the virus). Primary isolation of the virus is best
etiologic agent, a herpesvirus, is a member of the subfamily achieved from samples of liver, spleen, or kidney tissue that have
Alphaherpesvirinae of the family Herpesviridae (16, 17). The been homogenized in buffered saline and clarified by low speed
infection has not been reported in other avian species, mammals, or centrifugation.
humans.
PREFERRED CULTURE MEDIA AND’SUBSTRATES
CLINICAL DISEASE
Virus isolation may be attempted by inoculating clarified tissue
In domestic ducks and ducklings, DVE has been reported in birds homogenates onto cell cultures, or into ducklings or duck embryos.
ranging from 7 days of age to mature breeders. In susceptible
flocks, wild or domestic, the first signs are often sudden, high, and Cell Cultures
persistent mortality, and a significant drop in egg production. In Isolation of DVE virus may be made in primary duck embryo
chronically infected, partially immune flocks, only occasional fibroblasts (DEFs) (8, 13, 19), or preferably, primary Muscovy
deaths occur. Latent infections are established in recovered birds DEFs (MDEFs) (8, 13). Cell monolayers grown in medium
and these carriers may shed the virus in feces or on the surface of consisting of Eagle minimal essential medium (EMEM) containing
eggs over a period of years (16,18). 10% fetal calf serum (FCS), 2 mM glutamine, and 0.17% sodium
Clinical signs and gross lesions associated with a DVE outbreak bicarbonate and gentamicin are washed with serum-free EMEM and
vary not only with the virulence of the strain of virus, but also with then inoculated with the clarified sample homogenate suspected to
the species, age, and sex of the affected waterfowl (1, 17). In contain DVE virus. The virus is added to the cell monolayer and
breeder ducks, the signs may include photophobia, polydypsia, loss incubated for 1 hour at 37 C and then EMEM medium containing
of appetite, ataxia, watery diarrhea, and nasal discharge. Affected 2% FCS, 2 mM glutamine, and 0.17% sodium bicarbonate and
birds often have ruffled feathers and soiled vents. Sick birds may gentamicin is added and the cultures are incubated at 37 C in an
maintain an upright stance by using their wings for support, but atmosphere containing 5% CO2 This method of isolation may be
their overall appearance is one of weakness and depression. In modified to a plaque assay by overlaying the monolayer with
ducklings 2-7 wk of age, mortality may be lower than in older birds maintenance medium containing 1% agarose. Primary Muscovy
and the signs associated with DVE infection include dehydration, duck embryo liver cells may be more sensitive for virus isolation,
loss of weight, a blue coloration of the beaks, and blood-stained (R.E. Gough, pers. comm.). It has been reported (2) that the
vents. isolation of DVE virus in MDEF cells is favored by incubation at
At necropsy, little evidence exists of emaciation in adult ducks. temperatures between 39.5 and 41.5 C. However, an elevated
Prolapse of the phallus may occur in mature males. The gross temperature does not appear to be essential for isolation, which is
lesions are characterized by vascular damage, with tissue often carried out at 37 C. More than one passage may be necessary
hemorrhages and free blood in the body cavities, eruptions or for an isolation. The cytopathic effect is characterized by the
annular hemorrhages (seen as dark red bands around the intestine), appearance of pyknotic, rounded clumped cells that enlarge and
and ulcers with diphtheritic plaques on the mucosal surfaces of the become necrotic after 2-4 days. To confirm the presence of DVE
digestive tract. Lesions occur in all the lymphoid organs and virus, cultures should be stained with a fluorescent antibody
degenerative changes are apparent in the parenchymatous organs. conjugate using a direct or indirect method specific for DVE virus.
Collectively, these lesions are pathognomonic for DVE. The Cell monolayers may also be fixed and then stained with
pathology and histopathology of DVE in white Pekin ducks (Anas hematoxylin and eosin to show syncytial formation, intranuclear
platyrhynchos) has been reviewed (1, 17). inclusions, and marked cytoplasmic granulation.

Ducklings
When inoculated intramuscularly, 1-day-old susceptible ducklings
die within 3-12 days. Muscovy ducklings (Cairina moschata)
(Grimaud Farms, Stockton, Calif.) are more susceptible to DVE
than are white Pekin ducklings. Both gross and microscopic lesions
106
Chapter 23 Duck Virus Enteritis

typical of infection with DVE should be seen on postmortem Biological Properties


examination. Uninoculated ducklings, housed separately, should be Infected cells surrounding necrotic foci contain intranuclear
maintained as controls. The diagnosis may be confirmed either by inclusion bodies. Based on plaque-reduction tests, all strains of
vaccinating ducklings against DVE (Cornell University, Duck DVE virus currently belong to one serotype.
Research Laboratory, Eastport, Long Island, N.Y.) and challenging Chicken embryo-adapted DVE virus is used in the preparation of a
them subsequently with the virus isolate, or by staining of tissues live attenuated vaccine, which is recommended for use in birds 2
with a fluorescent antibody conjugate specific for DVE virus. weeks of age or older. Fattening or breeding ducks may be
However, virulent strains of the virus do exist, against which vaccinated subcutaneously or intramuscularly to produce active
vaccine may be ineffective (12). immunity. The vaccine virus is thought not to spread by contact
The susceptibility of Pekin ducklings to field isolates of DVE virus from vaccinated to unvaccinated ducks because unvaccinated
can be highly variable and therefore unreliable as the primary contact birds remain susceptible to infection.
method of isolation. Virus isolation in birds rather than in egg or
cell cultures is not always practical, but it may be necessary for a Antigen Detection
successful demonstration of the virus. In the author's recent Virus-neutralization (VN) and immunofluorescence tests are most
experience, even though a herpesvirus could be visualized by commonly used to confirm the identity of DVE virus using either
negative stain electron microscopy, and the necropsy and embryonating eggs or cell cultures. A plaque assay for DVE virus
histopathologic changes observed in Muscovy ducks were using duck embryo cell cultures has been described (4).
pathognomonic for DVE, isolation of virus was eventually achieved Hyperimmune antiserum prepared in ewes was used in a direct
only in Muscovy ducklings and not in MDEF cell cultures. fluorescent antibody test for DVE virus in DEF cells, and was
shown to be the next most sensitive assay after isolation in 1 to 9-
Duck Embryos day old ducklings (7). Direct or indirect immunofluorescent
Primary DVE virus isolations can also be made by inoculation tests using antisera to DVE virus prepared in ducks can be used to
onto the chorioallantoic membrane (CAM) of 9 to 14-day-old detect virus in cell cultures and in frozen tissue sections. A reverse
embryonating Muscovy duck eggs. The embryos may die, showing passive hemagglutination test for DVE has been described (5) but
characteristic extensive hemorrhages 4-10 days after inoculation. is reported to be less sensitive than immunofluorescence and plaque
Two to four serial passages of the homogenized CAMs may be assays. An avidin-biotin-peroxidase method of immunoperoxidase
necessary before an isolation is made. This method of isolation is staining to detect DVE antigen in formalin-fixed, paraffin-
not as sensitive as cell cultures or susceptible 1-day-old ducklings. embedded sections of liver and spleen from experimentally infected
Pekin duck embiyos vary in their susceptibility to strains of DVE birds has been described (11); this method could have diagnostic
virus. Embryonating chicken eggs are not very susceptible to potential. Herpesvirus may also be detected in clarified tissue
infection with field strains of DVE virus. The virus can, however, homogenates by negative stain electron microscopy; this is not,
be adapted to chicken embryos by serial passaging. however, positive confirmation that the herpesvirus is DVE virus.

AGENT IDENTIFICATION Molecular identification


Detection of DVE virus by polymerase chain reaction (PCR) has
Morphology been reported (9, 10, 14, 15). Primers have been identified that are
Mature virions are enveloped, have typical herpesvirus able to amplify DNA from DVE virus present in various tissues,
morphology, and range in size from 160 to 380 nm. Naked or including oesophagus, liver and spleen, from an original outbreak
unenveloped nucleocapsids may be seen by negative stain electron and after passage in Muscovy duck embryos
microscopy and are between 100 to 110 nm in diameter. The following detailed protocol for the detection of DVE virus
was provided by Dr. W. R. Hansen, US Geological Survey,
Physicochemical Properties Biological Resources Division, National Wildlife Health Center,
The virus genome consists of DNA and virus particles are 6006 Schroeder Road, Madison, WI 53711, USA. This procedure
sensitive to lipid solvents and are rapidly inactivated at a pH <3 or uses the following commercial items: GeneAmp PCR Reagent Kits
>10. containing dNTPs, lOx amplification buffer for hot start PCR, Taq
DNA polymerase, Lambda PCR control reagents, and Ampliwax
beads (Applied Biosystems), and a 100 base pair molecular size
ladder (Invitrogen).

Extraction of viral DNA This DNA extraction procedure can be used on disrupted cell suspensions from duck plague infected cell culture,
10% ground tissue suspensions, or cloacal swab material in transport medium. This method is used to prepare duck plague DNA for the
known positive PCR controls.
Note: All product transfers in steps i through v are performed inside a biological safety cabinet.

For 10% ground tissue suspension, add 400 μΐ to a 1.5 ml microfuge tube and microfuge at 16,000xg for 5 min. Transfer the
supernatant to a new tube and go to step ii.
i. For cell culture suspensions and cloacal swab material, add 400 μΐ of the sample, or supernatant from step i above, to a 1.5 ml tube
and microfuge at 16,000-20,OOOxg for 45 min, to pellet virus.
ii. Discard the supernatant and resuspend the pellet with 200 μΐ of Tris - EDTA (10 mM Tris-HCl, pH 8.0, 1 mM EDTA) buffer.
iii. Add 10 μΐ of a 5 pg/μΙ proteinase K solution giving a final concentration of 0.2 pg/μΙ, mix thoroughly, and incubate at 56 C for 1
hour.
iv. Add 25 μΐ of 10% sodium dodecyl sulfate solution giving a final SDS concentration of 1%, mix thoroughly, and incubate at 37° C
for one hour.
v. Add 15 μΐ of 5M NaCl giving a final concentration of 0.3 M and mix thoroughly.
vi. Add 300 μΐ of fresh phenol buffered with Tris-HCl, pH 8.0 to the tube, and mix by inverting 50 times.
vii. Microfuge tube at 16,OOOxg for 5 min and transfer the top aqueous phase (sample) to a new tube.
viii. Repeat the phenol extraction steps vii and viii one more time.
107
Peter R Woolcock

ix. Add 500 μΐ of ether to the tube, mix thoroughly, and microfuge at 16,000 x g for one min.
x. Discard the top aqueous phase (ether) and repeat the ether extraction step (x) one more time.
xi. Heat tube with lid open at 56 C for about 15 min or until the smell of ether is gone.
xii. Split tube contents in two and add 2.25 times the sample volume of 100% ethanol to each tube, mix tube contents by inverting tube
several times, and leave at room temp (22 C) for 30 min.
xiii. Microfuge tube at 16,000 x g for 45 min, and discard supernatant.
xiv. Add 200 μΐ of 70% ethanol to gently wash pellet then microfuge at 16,000 x g for 15 min.
xv. Discard the supernatant and diy pellet at 56 C for 30-45 min, with the tube lid open.
xvi. Resuspend the DNA in 30 μΐ of distilled water that is RNAase and DNAase free.
xvii. Store sample tube at 4 C until tested (few days) or at -20 C for long term storage.

Polymerase chain reaction


i. Lower reaction mixtures for the duck plague PCR and the lambda control are prepared in advance in a biosafety cabinet using the
kit manufacturers recommended methods for a hot start PCR. The lower reaction mixture is dispensed into PCR reaction tubes,
sealed with Ampliwax at 80 C as recommended by the manufacturer, and stored at 4 C for 1-2 mo.
ii. The following are the PCR primers for duck plague DNA-directed DNA polymerase gene:
iii. Primer 1 sequence 5’- GAAGGCGGGTATGTAATGTA - 3’ (forward)
iv. Primer 2 sequence 5’ - CAAGGCTCTATTCGGTAATG - 3’ (reverse)
v. The upper reaction mixture is prepared as a master mix according to the kit manufacturer’s recommendations the day of the test,
and distributed to each sample tube including the duck plague and lambda control tubes.
vi. Add 10 μΐ of DNA suspension from the stored sample tubes to PCR lower reaction tubes with corresponding labels.
vii. Place known duck plague DNA diluted to 1 pg/10 μΐ into one control tube and 10 μΐ of distilled water into the no DNA control
tube. Add 10 μΐ of lambda DNA supplied in the kit and 10 μΐ of water to corresponding lambda control tubes.
viii. Place all the tubes in a thermal cycler that is programmed as follows:
One cycle: Hold 94 C for 2 min.
Hold 37 C for 1 min.
Hold 72 C for 3 min.
35 cycles: Hold 94 C for 1 min.
Hold 55 C for 1 min.
Hold 72 C for 2 min.
One cycle: Hold 72 C for 7 min.
Hold 4 C until stored
PCR tubes are stored at 4 C until samples are examined for amplification products.

Electrophoretic analysis of PCR products


i. A fresh lx TAE buffer (40 mM Tris acetate, 1 mM EDTA, pH 8.3) is prepared from a lOx stock for agarose preparation and for use
in the electrophoresis chamber.
ii. A 1% agarose solution is prepared in TAE buffer, heated to dissolve the agarose and, when cool, poured into a gel former with a
comb.
iii. The solidified gel is placed into the electrophoresis chamber and TAE running buffer is added.
iv. PCR test samples, including the duck plague and lambda controls, are mixed 1/10 with Ιμΐ of loading buffer (0.25% (w/v)
bromophenol blue, 0.25% (w/v) xylene cyanol, 0.01 M Tris-HCl, pH 8.0, and 50% (v/v) glycerol) and 10 μΐ of each is added to
individual wells of the gel. The 100 base pair (bp) molecular size markers are added to each side of the gel at 0.4 pg per well.
v. Run the gel for one hour at 120 volts then stain in a 1% ethidium bromide solution for 20 min. Destain the gel for 45 min in
deionized water and view the gel on a UV illuminated light box. Photograph gel to record results.

Duck plague PCR interpretation: A 500 bp amplification band in the lambda control sample indicates the PCR ran successfully. A 446 bp
band in the duck plague known DNA control indicates the duck plague primers are working. A 446 bp band in the unknown test sample
indicates duck plague viral DNA was present. No amplification products will be present in the duck plague or lambda DNA controls. If bands
appear in these negative control products, cross contamination occurred during the and the test must be repeated.

Alternative methods to detect DVE virus DNA have been published by Plummer et al. (14), and Pritchard et al. (15)

SEROLOGIC DETECTION IN THE HOST between 0 and 1.5 were detected in domestic and wild waterfowl
that had not been exposed to DVE; an NI of 1.75 or greater was
Serologic detection of infection is only of importance in considered as evidence of prior exposure to DVE virus (3).
noncommercial waterfowl and may be used to determine prior Alternatively, sera may be screened using a constant-virus varying-
exposure of birds to DVE virus. serum method. In the author’s laboratory, a microtiter neutralization
The humoral response to natural infection with DVE virus is often assay using primary MDEF or DEF cells is used. Serial two-fold
low and antibodies may be short-lived (6); cell mediated immunity dilutions of each serum sample (heat inactivated at 56 C) are
is assumed to also play a role in the infection (16). However, prepared in 50 μΐ of serum-free EMEM in microtiter plates.
detection of neutralizing antibodies to DVE virus in serum is Approximately 102 0 mean tissue culture infective dose (TCID50) of
possible. VN assays, using a constant serum varying virus method, DVE virus in 50 μΐ of EMEM are added to each well and the
may be performed in chick or duck embryos by using embryo- mixtures are allowed to react at 37 C for 1 hr. A suspension of
adapted virus, or in cell cultures. Neutralization indices (NIs) primary MDEF or DEF cells in EMEM supplemented with 2 mM
108
Chapter 23 Duck Vine

L-glutamine, 0.17% sodium bicarbonate, and 10% FCS is adjusted 5. Deng, Μ Y., E. C. Burgess, and T. M Yuill. Detection of dock ρΜρκ
to contain 3 x 105 cells/ml. Cells are next added to the plates at 100 virus by reverse passive hemagglutination test Avian Dis. 28 616-62X.
μΐ per well and the plates are then incubated for up to 96 hr at 37 C 1984.
6. Docherty, D. E., and C. J. Franson. Duck Virus Enteritis. In: Vi ii i··^
in a humidified 5% CO2 atmosphere. Following incubation, cultures Diagnostic Virology, A. E. Castro and W. P. Heuschele, eds. Mosby Yor
are observed daily by light microscopy and finally fixed with 10% Book, St Louis, Missouri, USA. pp. 25-28. 1992.
formol-buffered saline and stained with 1% crystal violet. The 7. Erickson, G. A., S. J. Proctor, J. E. Pearson, and G. A Giwtrfw
plates are read macroscopically. The titer for virus-neutralizing Diagnosis of duck virus enteritis (duck plague). 17th Annual Proceedings of
activity is expressed as the reciprocal of the highest dilution of the American Association of Veterinary Laboratory Diagnostic^·!.
serum at which there is no evidence of cytopathic effects and AAVLD, Madison, Wisconsin, USA Roanoake, Virginia, USA pp. 85-90L
therefore complete virus neutralization has occurred. A titer of less 1974.
than 3 log2 is usually considered negative. A VN titer of 8 or 8. Gough, R. E., and D. J. Alexander. Duck Virus Enteritis in Greai-Brii··,
1980 to 1989. Vet. Rec. 126:595-597. 1990.
greater is considered significant and is evidence of exposure to 9. Hansen, W. R, S. E. Brown, S. W. Nashold, and D. L. Knudso·.
DVE virus (6). VN antibody may also be detected using cell Identification of duck plague virus by polymerase chain reaction. Avian Dti
cultures by mixing sera at a single dilution (e.g., 1:10) with 100- 43:106-115. 1999.
200 TCID50 virus and then testing inoculated cell cultures for non­ 10. Hansen, W. R., S. W. Nashold, D. Docherty, E., S. E. Brown, and D. L
neutralized virus by immunofluorescence. Although this method is Knudson. Diagnosis of Duck Plague in Waterfowl by Polymerase Cham
not quantitative, it can be useful for screening large numbers of Reaction. Avian Dis. 44:266-274. 2000.
sera. These latter methods, using constant virus and varying serum, 11. Islam, M R., J. Nessa, and K. M Halder. Detection of duck plague
require less sera than the NI methods. virus antigen in tissues by immunoperoxidase staining. Avian Path. 22:389-
393. 1993.
12. Kisary, S., and L. Zsak. Comparative studies on duck viral enteritis
DIFFERENTIATION FROM CLOSELY RELATED AGENTS (DVE) virus strains in geese. Avian Path. 12:395-408. 1983.
13. Kocan, R. M Duck plague virus replication in Muscovy duck fibroblast
DVE is usually pathologically distinct from other diseases of cells. Avian Dis. 20:574-580. 1976.
waterfowl; however, hemorrhages and necrosis do occur in other 14. Plummer, P. J., T. Alefantis, S. Kaplan, P. O'Connell, S. Shawky, and
diseases of waterfowl such as duck viral hepatitis, pasteurellosis K. A. Schat. Detection of duck enteritis virus by polymerase chain reaction.
{Pasteurella multocida) and some toxic conditions. Occasionally, Avian Dis. 42:554-564. 1998.
lesions caused by DVE virus could be confused with those 15. Pritchard, L. I., C. Morrissy, K. Van Phuc, P. W. Daniels, and H. A
Westbury. Development of a polymerase chain reaction to detect
produced by highly pathogenic Newcastle disease virus, avian Vietnamese isolates of duck virus enteritis. Vet. Microbiol. 68:149-156.
influenza virus, and poxvirus (16, 17). 1999.
16. Richter, J. Η. M, and M. C. Horzinek. Duck Plague. In: Virus
REFERENCES Infections of Birds, J. B. McFerran and M S. McNulty, eds. Elsevier
Science Publishers B.V., Amsterdam, the Netherlands, pp. 77-90. 1993.
1. Brand, C. J. Duck Plague. In: Field Guide to wildlife diseases, Vol. 1: 17. Sandhu, T. S., and S. A Shawky. Duck Virus Enteritis (Duck Plague).
General field procedures and diseases of migratory birds, M Friend, ed. In: Diseases of Poultry, 11th ed. S. Y.M, H. J. Barnes, J. R. Glisson, A M
Resources publication 167. U.S. Department of the Interior, Fish and Fadly, L. R. McDougald and D. E. Swayne, eds. Iowa State Press, pp. 354-
Wildlife Service, Washington, DC. pp. 117-127. 1987. 363. 2003.
2. Burgess, E. C., and T. M Yuill. Increased cell culture incubation 18. Shawky, S., and K. A. Schat. Latency sites and reactivation of duck
temperatures for duck plague virus isolation. Avian Dis. 25:222-224. 1981. enteritis virus. Avian Dis. 46:308-313. 2002.
3. Dardiri, A H., and W. R. Hess. The incidence of neutralizing antibodies 19. Wolf, K., C. N. Burke, and M C. Quimby. Duck viral enteritis: a
to duck plague virus in serums from domestic ducks and wild waterfowl in comparison of replication by CCL-141 and primary cultures of duck embryo
the United States of America. Proceedings, Annual Meeting of the United fibroblasts. Avian Dis. 20:447-454. 1976.
States Animal Health Association, pp. 225-237. 1967.
4. Dardiri, A H., and W. R. Hess. A plaque assay for duck plague virus.
Can. J. Comp. Med. 32:505-510. 1968.

109
24
HERPESVIRUSES OF FREE-LIVING AND PET BIRDS
Erhard F. Kaleta

SUMMARY. Free-living and pet birds are affected by a large number of different herpesviruses (HVs). The taxonomic status within the
three subfamilies α-, β-, and y-Herpesvirinae is still undetermined for most of the many isolates. HV isolates can be distinguished by
restriction enzyme analysis, cross neutralization tests, and in part by plaque morphology in chick-embryo fibroblast (CEF) and chick embryo
kidney or chick embryo liver cell cultures. Replicating and latently present HVs can be detected in swabs and various tissues by polymerase
chain reactions (PCRs).
Most commonly affected by HVs are various breeds of domestic pigeons, waterfowl, many species of psittacines, owls, black and white
storks, and various finches; eagles, falcons, cormorants, toucans, and quail are less frequently infected. Subclinical infections occur in all
species. Various forms of apparent disease are frequently linked to the presence of debilitating factors such as overcrowding, food primarily
deficiencies, prolonged antibacterial treatment, and poor hygiene. In the acute phase of the disease lesions are primarily hemorrhagic and
inflammatory in nature and in the chronic phases lesions are predominantly necrotic mostly in the pharynx, esophagus, and intestines, but
also in large parenchymal organs and brains.
Agent Identification. Diagnosis of HV infections in live birds is preferably made by virus isolation from buffy coat cells and feather pulp
material. Homogenates of parenchymal organs, bone marrow, or brain tissues from dead birds may be used as inocula for cell cultures of
avian origin. Viral DNA can be detected by PCR. Virus identification is achieved by chloroform treatment of supernatant fluids from heavily
infected cell cultures, cultivation of the isolate in question in the presence of iododeoxyuridine, and electron microscopy. Identification of
serotypes is done by cross neutralization tests.
Serologic Detection in the Host. Subclinical infection is commonly associated with seroconversion. The assay of sera in neutralization tests
identifies infected, possibly also virus positive birds. In many cases several serologically unrelated viruses must be used to detect all
serotypes of HVs known to occur in a particular species of bird. Repeated serologic monitoring is advisable for all bird collections, breeding
stations, and quarantine stations.

and 4 (31,64). Transmission experiments that prove the etiology and


INTRODUCTION genesis of these papillomatous tumors have.not been reported;
2. the hemorrhagic type in infectious laryngotracheitis (ILT)
Domestic, free-living, and pet birds are frequent hosts of a large infections in chickens and in pheasants (11,13,41), peafowl
variety of herpesviruses (HVs). Infections may go unnoticed or may (3,5,13,42), and in guinea fowl (75); and
result in fatal disease. HVs often interfere with captive breeding 3. the necrotic type, which is found in many birds dying of HV
programs or with attempts to re-establish free-living bird infections. These birds include owls (6), falcons (51,74), eagles
populations. In addition, enzootics may occur in wild birds. (16), rural and fancy pigeons (63,71) many psittacines
(30,45,50,54), black storks (Ciconia nigra; 38) and white (Ciconia
CLINICAL DISEASE ciconia) storks (39), quail (40), and cranes (7), as well as
passeriforms of the families Estrildidae (exotic finches), Ploceidae
More than any other viruses, HVs have a tendency for latency and (weavers), and Carduelidae (finches, including the common
persistent infections. Young age, concurrent infections, and canary). Respiratory distress, hepatitis, and disseminated focal
noninfectious noxae may precipitate disease, whereas antibacterial, necrosis located in liver, spleen, and bone marrow are most
antifungal, and antiparasitic treatments may ameliorate the course commonly seen (61,68,72,76).
of the disease. In any case, clinical signs appear primarily as
nonspecific, general depression of common behavioral traits (46). SAMPLE COLLECTION
Psittacines often have yellowish droppings. However, in a large
number of cases, sudden increases in mortality rates may be the Latent, non-productive infections are difficult to detect, and
only signalment evident in many species of birds. Therefore, any cyclophosphamide treatments before sampling may enhance
increased death rate should prompt a detailed examination (18). recovery of the agent. During pairing time in seed-eating birds that
In psittacines, HV-induced mortality is often preceded by feed their offspring with crop-milk and/or macerated seeds (e.g.,
introductions of latently infected birds, such as South American pigeons and some psittacines, but also many passerine species), HV
conures of the genera Pyrrhura and Cyanoliseus (22). These birds infections change from the latent, non-productive state to the
tend to be more often latently infected and are prolonged shedders persistent, productive state.
than are other psittacines (21,23). Falcons, and in particular owls, Swabs of the oropharynx should be placed in complete tissue
often acquire fatal infections from preying on persistently infected culture medium without serum. This medium is suitable for storage
domestic pigeons (2). Housing of large numbers of birds of various of samples at 4 C for about 1 wk. If necessary, infectivity in
species in quarantine stations and pet bird shops provide ideal swabbed epithelial cells can be preserved in freezing medium; (80
circumstances for the horizontal spread of different viruses, ml basal medium, Eagle’s [BME] or M199, 10 ml fetal calf serum
including HV, and also avian paramyxoviruses (10,25,34). [FCS], and 10 ml dimethylsulfoxide [DMSO]; or in sucrose-
Gross lesions in birds dying from HV-induced diseases (17,24,32) phosphate-glutamate albumin [SPGA] buffer) (8).
allow categorization of the disease into three main types: In addition to swabs, tissue specimens should be taken from all
1. the neoplastic type in chickens due to serotype 1 Marek’s disease organs with macroscopically visible lesions, (e.g. liver and spleen)
viruses and in pigeons due to an HV antigenically unrelated to in birds that die from suspected HV disease. Bone marrow
Marek's disease or classical HV infection of pigeons. specimens are valuable if carcasses have advanced autolysis.
Circumstantial evidence for a causal relationship between mucosal Specimens should be minced immediately and immersed in cell­
tumors in neotropical parrots and HVs has been reported (47,55). culture medium with twice the normal strength of antibiotic and
These mucosal tumors in pharynx and cloaca of neotropical parrots antimycotic drugs. Such samples can be kept refrigerated for up to 1
of the genera Ara and Amazona are histomorphologically similar to wk without appreciable loss of infectivity. Freezing of samples
papillomas and are linked to psittacid HVs of genotypes 1, 2, 3, should be avoided. If long-term storage is necessary, tissue
homogenates should be suspended in freezing medium (see above).
110
Chapter 24 Herpesviruses of Free-living and Pei Bmk

Stork HV induce a long-lasting viremia, with the bulk of HV Characterization


infectivity associated with peripheral leukocytes (39). Heparinized Culture filtrates that are free of bacteria, mycoplasmas,
whole blood is aseptically drawn from the wing vein of storks and chlamydiae, and fungi and that induce any of the three types of
further processed for virus isolation, as in the case of Marek's cellular focal alterations described above should be examined
disease herpesvirus (see Chapter 22 on Marek’s disease). further to confirm the presence of HVs. The type of lesions in
affected birds, the tissues that yield isolates, and the types of
PREFERRED CULTURE MEDIA AND SUBSTRATES cellular alterations are an indication, but not proof, of the presence
of HVs. Virus identification involves physicochemical and
Cell cultures are the preferred substrate and any common cell biological properties, and, finally, immunologic identification.
culture medium (e.g., BME, minimum essential medium [MEM] Molecular characterization, e.g. in situ hybridization (ISH),
and modifications, or Ml99) that supports the growth and restriction fragment length polymorphism (RFLP) and several
maintenance of cultured avian embryonic cells or young chick cells techniques of polymerase chain reaction (PCR) have gained
is suitable for the isolation of avian HVs (see Chapter 43 on cell increasing importance.
culture methods). DNA of HVs that are in the non-productive stage
(latency) can be detected by various PCRs. Physicochemical Properties
All known avian HVs multiply well in primary monolayer cultures Essential Lipid. All HVs are sensitive to lipid solvents. HVs
of chicken kidney cells (CKCs) and also chick embryo liver cells enclosed in syncytia may escape the direct action of chloroform,
(CELC). Marek's disease, ILT, and Lake Victoria Cormorant HV leaving some intact particles unaffected. Centrifugation or filtration
grow only in chicken kidney epithelium or CELC. It is laborious to through 220-nm filters before chloroform treatment eliminates this
adapt these three viruses to chick embiyo fibroblasts (CEFs). Other problem.
cells (e.g., duck embiyo fibroblasts or cell lines of mammalian Filtration. An estimate of the average diameter of virions can be
origin) either are not definitely free from adventitious agents or obtained by sequential filtration sets.
yield erratic results. Therefore, such cell cultures cannot be Iododeoxyuridine. Virus replication is inhibited, but not entirely
recommended. Cells may be inoculated immediately after plating or prevented, by halogenated desoxyribonucleosides.
after the formation of subconfluent monolayers. Incubation for Morphology. Cell lysates of heavily inoculated cultures showing
more than 1 wk is not advisable. Instead, inoculated trypsinized cytopathic effect (CPEs) are prepared either by one or two freeze­
cells should be subcultured at intervals of 4-6 days. thaw cycles or by sonication. After clarification by low-speed
Depending on the particular strain of HV, three types of cellular centrifugation, viral particles in the supernatant are pelleted by
alterations can be observed in unstained cultures: ultracentrifugation. The pellet is resuspended in saline, negatively
1) Small plaques consisting of roundish refractile cells on top of stained, and examined in a transmission electron microscope. All
otherwise normal-appearing confluent CKCs or CEFs. complete enveloped HVs have the appearance of a fried egg, that is,
2) Large plaques with a lytic center and rounded cells adjacent to with an irregular envelope and a nucleocapsid roughly in the center.
the periphery of the plaque. With extended incubation, more Naked, nonenveloped particles or particles with disrupted envelopes
and more rounded cells detach and float in the medium. In the may also be seen (9,10,24,45,59,62,69). Naked particles tend to
final stages a netlike structure of unchanged elongated cells form clusters consisting of 3-10 nucleocapsids. Because of
remains. relatively low infectivity titers (usually in the range of ΙΟ4—106
3) Syncytial-type plaques composed initially of large irregular median tissue culture infective doses [TdD50]/ml), negative results
structures. are likely. In such situations, several steps of multiplication,
Upon further incubation, these syncytia detach from the surface of purification, and concentration are needed.
the culture vessel and assume roughly spherical shapes and float in Because of variations in steps used for the processing of samples
the medium and later disintegrate into particulate matter with for electron microscopy and because of inherent differences in the
irregular boundaries and different diameters. Some avian HV examined isolates, diameters of the nucleocapsid vary between 100
isolates may contain small and large plaque variants (35). nm and 150 nm and of the enveloped particles from 150 nm to more
An inoculum can be considered to be negative if the primary culture than 200 nm. Some HVs, such as several stork isolates, have an
and two subsequent subcultures do not yield any of the described envelope that tightly surrounds the nucleocapsid. Others, such as
cytopathologic changes. owl, falcon, eagle, psittacine, crane, and quail HVs, carry a large,
Most HV isolates can be kept frozen without detrimental effects on sometimes folded envelope (40). Spikes are not always clearly
viral titer. However, most of the stork isolates (39) and at least some discernible on the outer surface of the envelope.
of the strains recovered from psittacines and pigeons (33) are highly
cell-associated. Preservation of such strains requires the addition of Biological Properties
SPGA buffer (8,19) before freezing. Embryo Inoculation. Herpesviruses do multiply following
inoculation of yolk sacs or chorioallantoic membranes (CAMs) in
AGENT IDENTIFICATION embryonating eggs from chickens or other birds. Inoculation by
either route results in variable embryo mortality and, if the embryo
Taxonomy survives the inoculation for more than 4 days, distinct pox-like
Herpesvirus species of free-living and pet birds are currently not lesions form on the CAM. The embryo itself may be stunted and
assigned to any of the three subfamilies of the α-, β-, and γ- have disseminated focal necrosis of the liver and spleen;
Herpesvirinae (52,60). Moreover, names for the diseases in various occasionally focal lesions are present on the mucosa of the palatine
bird species or higher taxons have not yet been coined. In this bone. Allantoic fluid does not agglutinate chicken erythrocytes.
paper, names for HV isolates are selected according to suggestions Intranuclear Inclusions. Predominantly eosinophilic Cowdry
made by Roizman (60). Table 24.1 presents an updated list of 27 Type A intranuclear inclusion bodies surrounded by a distinct halo
known and sufficiently characterized HV grouped in chronologic can be detected in hematoxylin-and-eosin-stained sections of liver,
order according to the first description of a defined disease or of the spleen, and bone marrow cells of naturally infected birds
causative virus. Additional data refer to the main host species (or (9,10,24,45,59,62,69); in cultured cells inoculated with avian HVs
group), the provisional species designation (60), the serologic (9,12,15,34,37,42,51,62); and liver and spleen cells of infected
relatedness of the isolates to each other (37,40), and the embryos (6,18,33,48).
predominant type of lesion in naturally infected birds (32).
Ill
Erhard F. Kai eta

Animal Inoculation. Numerous in vivo studies with HVs derived SEROLOGIC DETECTION IN THE HOST
from domestic birds have yielded valuable information on the
development of signs, lateral spread and lesions and contributed to Neutralizing antibody in sera of birds can be detected by the
the knowledge of pathogenesis, prevention and immunoprophylaxis. plaque-reduction method in primary CEF or CKC cultures. Thirty to
Pathogenicity studies with avian HVs and naturally free-living and 100 plaque-forming units of virus are reacted with twofold dilutions
pet birds are generally of limited value. Maintenance of such birds of antisera and incubated for at least 1 hr at room temperature. Two
in biocontainment isolation cabinets is difficult because of the plates are inoculated as negative controls. All cultures are stained
birds’ sometimes extreme demands on the environment and food. with neutral red (2 ml of a 1:40,000 dilution in phosphate-buffered
The general health of such birds and their immune status to HVs are saline per 60-mm petri dish culture) after 5-6 days of incubation
often unknown; their immunocompetence might be impaired. Legal and examined macroscopically for plaques. Plaque numbers are
rulings on animal protection and ethical reasons prevent the expressed as a percentage of negative controls. The serum dilution
handling of endangered species. is then plotted against the probit values, and the titer of the antisera
Data on the host range of HVs can be obtained more effectively by is estimated graphically and enumerated as log2 values.
swabbing the pharynx and cloaca or by assaying purified peripheral A less laborious neutralization test can be performed under less
leukocytes for viremia. Indirect evidence for the presence of natural stringent conditions in 96-well microtiter plates. Twofold serum
infections by various HVs can be gathered by testing serum samples dilutions ranging from 1:2 to 1:256 are incubated with 100 TCID50
for neutralizing antibodies (15,28). The domestic chicken is not of specific HV for at least 1 hr. Freshly prepared CEF at a
susceptible to any avian HVs other than Marek's disease HVs and concentration of 106 cells/ml are added. After 5-6 days of
ILT virus. incubation, plates are stained with crystal violet solution, and serum
Genomic properties. Parts of the genome of some psittacine and titers are macroscopically enumerated. The titer of a serum is the
passerine HVs were studied (27,66,67) using consensus primer highest dilution that produces no CPEs.
PCRs that amplify either a region in the DNA-directed DNA Plaques also can be enumerated by direct or indirect
polymerase gene (70,76) or parts of the UL26 open reading frame immunofluorescence (57,58).
(66,67). The UL6 and UL7 of duck enteritis HV were amplified by Other serologic tests, such as agar gel precipitation, complement
Plummer et al. (56). The results of these studies confirmed in most fixation, or enzyme-linked immunosorbent assay are either
instances the genetic heterogeneity of psittacine HVs, the results of insufficiently sensitive or not yet developed and validated (3).
previous cross neutralization tests and earlier data on the restriction
fragment length polymorphisms (26,27,34,37,40). Although DIFFERENTIATION FROM CLOSELY RELATED AGENTS
assignment of the examined HVs to any of the three established
subfamilies of the family Herpesviridae is not yet achieved, the Using the neutralization test, some avian herpesviruses are shown
genomic data contribute to the phylogenetic evolution of avian, to be serologically closely related (Table 24.1). These are Marek's
mammalian and reptilian HVs (12,14). disease and turkey herpesviruses; HVs of pigeons, owls, falcons,
and eagles; and HVs from cranes and bobwhite quail. No serologic
Immunologic Identification relatedness is detectable among any of the other viruses listed in
Antisera against specific HVs can be produced in rabbits and used Table 24.1. It should be noted that at least five serologically
with the plaque-reduction method in primary CEF or CKC cultures different herpesviruses exist within birds of the order Psittaciformes
(see serologic detection in the host below) to categorize unknown (26). The psittacine HV of the serotype 1 is most frequently
HV isolates into serogroups. Avian HVs are poorly immunogenic in isolated. Also, two serologically distinguishable HVs can be
laboratory rodents. The following procedure has been adopted to recovered from pigeons. The serologic status of HVs recently
raise antisera in rabbits (26,27). Viruses are harvested, and cells are isolated from passeriforms in this laboratory (exotic finches, weaver
disrupted by ultrasonic vibration. Cellular debris is removed by bird, and common canaiy) indicates a relationship to psittacine
centrifugation at 4000 x g for 30 min. The supernatant fluid is viruses (26,27,68).
mixed with 10% (w/v) polyethylene glycol MW 6000, stirred at 4 Any enhanced morbidity and above-normal mortality in free-living
C, and kept overnight (48). After centrifugation at 4000 x g for 10 and pet birds requires a comprehensive postmortem and laboratory
min, the pellet is resuspended in Dulbecco’s phosphate buffer and examination. Certain bacteria (e.g., Salmonella spp., Klebsiella
layered on top of a 12-52% linear sucrose gradient as described by spp., Escherichia coli, Mycobacterium spp., Campylobacter spp.,
Lee et al. (48) and centrifuged at 25,000 x g for 1 hr in a Beckman Pseudomonas spp., Bacillus spp., Erysipelothrix rhusiopathiae, and
SW 27 rotor (Beckman Instruments, Inc, Fullerton, Calif.). The Pasteurella spp.) or bacterial exotoxins (e.g., botulism) may cause
gradient fluid is collected in 2-ml fractions. The same fractions are focal necrosis in livers.
titrated in cell cultures for infectious virus. Fractions with the Hemorrhagic lesions in the respiratory tract might be caused by
highest viral content are mixed in equal parts of complete Freund's certain bacterial infections (e.g., Pasteurella spp.) or by the red
adjuvant and are inoculated intradermally on multiple sites on the worm Syngamus trachea. Highly pathogenic avian influenza and
back of a rabbit. The first antiserum is collected 4 wk later. Booster Newcastle disease viruses grow readily in cell cultures and
injections are given at 2-wk intervals. Rabbits are bled 2 wk after embryonating eggs. Members of both groups of viruses easily can
the third booster injection is given. Serum can be collected and be detected by hemagglutination in cell culture and allantoic fluids
stored until used for serotyping unknown HVs. of inoculated eggs.
Using the plaque-reduction method in primary CEF or CKC Diphtheritic lesions in the intestines might be caused by
cultures, 11 different serotypes (33,37) can be distinguished. In Trichomonas spp. if the organisms are found in the pharynx or
Table 24.1, available data on the serologic relatedness of esophagus. Jejunal diphtheritic lesions and hemorrhages can be
herpesviruses is indicated in the fourth column. induced by Clostridium spp.
Available, yet preliminary, data on DNA restriction endonuclease Chlamydophila (formerly Chlamydia) psittaci, as a possible cause
analysis (2,27) seem to confirm the results of conventional of respiratory distress, is widespread in various birds (42,46). Its
neutralization tests. detection is possible with cell culturing and other techniques.
The use of monoclonal antibodies for identification and Laboratory workers must take special precautions to avoid
differentiation of avian HVs is hampered by the large number of becoming infected.
different viruses from various bird species. So far only one report Several avian viruses that also induce CPEs and/or intranuclear
exists on studies of a psittacine HV (1). inclusions need to be excluded (49,70). These are polyomaviruses
112
Chapter 24 Herpesviruses of Free-living and Pet Birds

(especially psittacines), reoviruses, rotaviruses, and adenoviruses (65). Biological substances such as various preparations of propolis
(in many species, particularity in pigeons); paramyxoviruses (in and several caffeoylics inhibit the multiplication in vitro and in vivo
almost all avian species); orthomyxoviruses (frequent in waterfowl of HVs from pigeons and psittacines (35,44). All these drugs need
and gallinaceous birds); togaviruses and coronaviruses (in turkeys); to be given very early and repeatedly after infection. Local reactions
and also poxiruses (4). Viruses that might be present but do not at the site of injection may occur.
necessarily cause CPEs include picomaviruses (in waterfowl and Several attempts have been made to protect psittacines by
gallinaceous birds), parvoviruses (in geese and Muscovy ducks), adjuvanted vaccines containing partially purified, formol-
and some retroviruses (in ducks and turkeys), in psittacines); inactivated viruses (20,21,29,36,77). The widespread prophylactic
Control and vaccination. All HVs are horizontally transmitted via use of such vaccines is limited by several factors. These include the
saliva or droppings. The detection of shedders by virologic broad antigenic diversity of psittacine HVs (26,66,67), which
examination of swabs taken from the oropharynx and cloaca (see precludes the use of one seed virus for vaccines in all cases and
above) and separation of shedders from noninfected susceptible undesirable side effects such as local inflammatory or
birds greatly helps to reduce fatal infections (23). granulomatous to necrotic reactions at the site of inoculation. In
Cell-free viruses in the environment are sensitive to inactivation addition, strict but variable governmental regulations for such
by ultraviolet light (sunshine) and also heat and chemical ’’niche" products in various countries makes it cumbersome for
disinfectants (73). commercial enterprises to develop and produce these vaccines.
The application of inhibitors of viral DNA synthesis such as Available information on vaccination-challenge experiments or
acyclovir and gancyclovir resulted in reduced rates of morbidity and seroconversion are promising and favor additional studies on
mortality following experimental exposure of Quaker parakeets innocuity, immunogenicity, and duration of protection.
(53). Some strains of pigeon HVs also are sensitive to acyclovir

Table 24.1. Updated list of avian herpesviruses (Hvs).


Susceptible bird Provisional HV
Name of (species)/ species Serologically related
No. disease/infection HV isolated from designationA to no. Type of lesion References
1 Marek s disease Chicken GallidHV2 2 Tumorous
Turkey HV
2 infection Turkey, chicken Meleagrid HV 1 1 None
3 Duck viral enteritis Ducks, geese, swans AnatidHVl None Hemorrhagic
Infectious Chicken, pheasants,
4 laryngotracheitis others GallidHVl None Hemorrhagic
5 Pacheco's disease Psittaciformes Psittacid HV 1 20-23, 25 Necrotic 26,61
6 Pacheco's disease Psittaciformes Psittacid HV 2 None Hemorrhagic 26,44
7 Pacheco's disease Psittaciformes Psittacid HV 3 None Necrotic 26, 33
8 Pacheco's disease Psittaciformes Psittacid HV 4 None Necrotic 26
9 Pacheco's disease Psittaciformes Psittacid HV 5 19 Necrotic 26
10 Smadel 's disease Pigeons ColumbidHVl 12,13,14 Necrotic 11,52
Pharyngo
11 esophagitis Pigeons Columbid HV 2 None Necrotic 33
Hepatosplenitis
12 infectiosa strigum Various owls StrigidHVl 10,13,14 Necrotic 6
Inclusion body
13 hepatitis Falcon FalconidHVl 10,12, 14 Necrotic 50, 73
Inclusion body
14 hepatitis Bald eagle nestling Acciprid HV 1 10,12, 13 Necrotic 16
Lake Victoria Phalacrocoracoid
15 None cormorant HV 1 None None® 19
Inclusion body
16 hepatitis Cranes GruidHVl 17 Necrotic 7, 18
17 None Bobwhite quail PercididHVl 16 Necrotic 40
Black and white
18 None storks Ciconiid HV 1 None Necrotic 34,39
Black-footed
19 None penguin Sphenicid HV 1 4? Hemorrhagic 43
20 None Exotic finch EstrildidHV 1 9 Hepatitis 60,71,76
21 None Weaver bird PloceidHVl 5 Hepatitis 60,71
22 None Canary Serinid HV 1 5 None® 27
23 None Exotic finch EstrildidHV 2 5 None® 27
24 None Exotic finch EstrildidHV 3 5 None® 27
25 None Tragopan Tragopanid HV 1 None None® 27
26 None Superb starling Lamprotomid HV 1 5 Hepatosplenitis 27
27 None Toucan Andigenid HV 1 9 Necrotic 9

A Modified from Roizman (49).


B No lesions observed so far

113
Erhard F. Kaleta

REFERENCES herpesvirus isolates by restriction endonuclease analysis. Avian Pathol.


26:305-316. 1997.
1. Abdala, N. Development of an experimental ELISA for the detection of 28. Heffels, U., K. Fritzsche, E. F. Kaleta, and U. Neumann. Serologische
antigens and antibodies to the virus of Pacheco disease using monoclonal Untersuchungen zum Nachweis virusbedingter Infektionen bei der Taube in
antibodies. J. Vet. Med. Ser. B 41:369-380. 1994. der Bundesrepublik Deutschland. Dtsch. Tieraerztl. Wochenschr. 88:97-
2. Aini, I., L. M Shih, A. E. Castro, and Y. C. Zee. Comparison of 102. 1981.
herpesvirus isolates from falcons, pigeons and psittacines by restriction 29. Hitchner, S. B., and B. W. Calnek. Inactivated vaccine for parrot
endonuclease analysis. J. Wildl. Dis. 29:196-202. 1993. herpesvirus infection (Pacheco’s disease). Am. J. Vet. Res. 41:1280-1282.
3. Bauer, B., J. E. Lohr, and E. F. Kaleta. Comparison of commercial 1980.
ELISA test kits from Australia and the USA with serum neutralization tests 30. Homer, R. F., Μ E. Parker, E. F. Kaleta, and L. Prozesky. Isolation and
in cell cultures for the detection of antibodies to the infectious identification of psittacid herpesvirus 1 from imported psittacines in South
laryngotracheitis virus of chickens. Avian Pathol. 28:65-72. 1999. Africa. J. S. Afr. Vet. Assoc. 63:59-62. 1992.
4. Bolte, A. L., J. Meurer, and E. F. Kaleta. Avian host spectrum of 31. Johne, R., A. Konrath, M-E. Krautwald-Junghanns, E. F. Kaleta, H.
avipoxviruses. Avian Pathol. 28:415-432. 1999. Gerlach, and H. Muller. Herpesviral, but no papovaviral sequences, are
5. Borst, G. H. A., and G. M Lambers. Infectious laryngotracheitis in detected in cloacal papillomas of parrots. Arch. Virol. 147:1869-1880. 2002.
peafowl in the Netherlands. Tijdschr. Diergeneeskd. 108:199-200. 1983. 32. Kaleta, E. F. Herpesvirus-induzierte Infektionen und Krankheiten des
6. Burtscher, H. Die virusbedingte Hepatosplenitis infectiosa strigum 1. Vogels. Tieraerztl. Prax. 11:67-75. 1983.
Mitteilung: Morphologische Untersuchungen. Pathol. Vet. 2:227-255. 1965. 33. Kaleta, E. F. Infections and diseases of birds caused by alpha- and beta-
7. Burtscher, H., and W. Gruenberg. Herpesvirus-Hepatitis bei Kranichen herpesvirinae. In: Proc. 3rd International Symposium on Marek’s disease, S.
(Aves: Gruidae) I. Pathologische Befunde. Zentralbl. Veterinaermed. B Kato, T. Horiuchi, T. Mikomi, K. Kirai, eds. Osaka, Japan, Sept. 12-16. pp.
26:561-569. 1979. 372-388. 1988.
8. Calnek, B. W., S. B. Hitchner, and Η. K. Adi dinger. Lyophilization of 34. Kaleta, E. F. Herpesviruses of birds.—A review. Avian Pathol. 19:193—
cell-free Marek’s disease herpesvirus and herpesvirus of turkeys. Appl. 211. 1990.
Microbiol. 20:723-726. 1970. 35. Kaleta, E. F. EinfluB von Propolis der Honigbiene (Apis mellifera L.)
9. Charlton, B. R., B. C. Barr, A. E. Castro, P. L. Davis, and B. J. Reynolds. auf die Vermehrung einer Klein- und GroB-Plaque-Variante des Tauben-
Herpes viral hepatitis in a toucan. Avian Dis. 34:787-790. 1990. Herpesvirus in vitro. Tieraerztl. Umsch. 46:553-556. 1991.
10. Cheeseman, Μ T., and C. Riddell. Esophagitis due to a herpesvirus 36. Kaleta, E. F., and M Bueno Brinkmann. An outbreak of Pacheco’s
associated with mortality in a psittacine aviary. Avian Dis. 39:658-660. parrot disease in a psittacine bird collection and an attempt to control it by
1995. vaccination. Avian Pathol. 22:785-789. 1993.
11. Cornwell, H. J. C., and A. R. Weir. Herpesvirus infection of pigeons. 37. Kaleta, E. F., U. Heffels, U. Neumann, and T. Mikami. Serological
IV. Growth of the virus in tissue-culture and comparison of its differentiation of 14 avian herpesviruses by plaque reduction tests in cell
cytopathogenicity with that of the viruses of laryngotracheitis and pigeon cultures. In: Proceedings of the 2nd International Symposium of Veterinary
pox. J. Comp. Pathol. 80:517-523. 1970. Laboratory Diagnosticians, Lucerne, Switzerland. June 24—26, 1:38-41.
12. Cracraft, J. Avian evolution, Gondwana biogeography and the 1980.
Cretaceous-Tertiary mass extinct event. Proc. R. Soc. Lond. B 268:459-469. 38. Kaleta, E. F., T. Mikami, H.-J. Marschall, U. Heffels, M Heidenreich,
2000. and B. Stiburek. A new herpesvirus isolated from black storks (Ciconia
13. Crawshaw, G. J., and B. R. Boycott. Infectious laryngotracheitis in nigra). Avian Pathol. 9:301-310. 1980.
peafowl and pheasants. Avian Dis. 26:397-401. 1982. 39. Kaleta, E. F., and N. Kummerfeld. Persistent viraemia of a cell-
14. Davison, A J. Evolution of herpesviruses. Vet Microbiol. 86:69-88. associated herpesvirus in white storks (Ciconia ciconia). Avian Pathol.
2002. 15:447-453. 1986.
15. Docherty, D. E., and R. I. Romaine. Inclusion body disease in cranes: a 40. Kaleta, E. F., H.-J. Marschall, G. Gluender, and B. Stiburek. Isolation
serological follow-up to the 1978 die-off. Avian Dis. 27:830-835. 1983. and serological differentiation of a herpesvirus from bobwhite quail (Colinus
16. Docherty, D. E., R. I. Romaine, and R. Night. Isolation of a herpesvirus virginianus,L. 1758). Arch. Virol. 66:359-364. 1980.
from a bald eagle nestling. Avian Dis. 27:1162-1165. 1983. 41. Kaleta, E. F. and T. Redmann. Die infektiose Laryngotracheitis bei
17. Eskens, U., E. F. Kaleta, and G. Unger. Eine Herpesvirus-bedingte Huhn, Pfau und Fasan sowie Moglichkeiten und Grenzen ihrer Bekampfung
Enzootie - Pacheco’sche Papageienkrankheit in einem Psittazidenbestand. durch attenuierte Lebendimpfstoffe. Tieraerztl. Prax. 25:605-610. 1997.
Tieraerztl. Prax. 22:542-553. 1994. 42. Kaleta, E. F. and E. M A. Taday. Avian host range of Chlamydophila
18. Foerster, S., C. Chastel, and E. F. Kaleta. Crane hepatitis herpesviruses. spp. based on isolation, antigen detection and serology. Avian Pathol.
J. Vet. Med. Ser. B 36:433-441. 1989. 32:435462.
19. French, E. L., H. G. Purchase, and K. Nazerian. A new herpesvirus 43. Kincaid, A. L., and M Cranfield. Herpesvirus-like infection in black­
isolated from a nestling cormorant (Phalacrocorax melanoleucos). Avian footed penguins (Speniscus demersus). J. Wildl. Dis. 24:173-175. 1988.
Pathol. 2:3-15. 1973. 44. Koenig, B., and J. H. Dustmann. The caffeoylics as a new family of
20. Fudge, A. M Psittacine vaccines. Vet. Clin. North Am. Small Anim. natural antiviral compounds. Naturwissenschaften 72:659-661. 1985.
Pract. 21:1273-1279. 1991. 45. Krautwald, M-E., S. Foerster, W. Herbst, B. Schildger, and E. F.
21. Gaskin, J. M, D. A. Arnold, and C. M Robbins. An inactivated vaccine Kaleta. Nachweis eines neuen Herpesvirus bei einem ungewohnlichen Fall
for psittacine herpesvirus infection (Pacheco’s disease). Am. Assoc. Zoo von Pachecoscher Krankheit bei Amazonen und Graupapageien. J. Vet.
Vet. Annu. Proc., Arlington, Va. pp. 102-105. 1980. Med. Ser. B 35:415^120. 1988.
22. Gaskin, J. Μ, B. Raphael, A. Major, and G. Hall. Pacheco’s disease: the 46. Krautwald-Junghanns, M-E., F. Schumacher, and B. Tellhelm
search for the elusive carrier bird. Am. Assoc. Zoo Vet. Annu. Proc., Seattle, Evaluation of the lower respiratory tract in psittacines using radiology and
Wasm. pp. 24-28. 1981. computed tomography. Vet. Radiol. Ultrasound 34:382-390. 1993.
23. Gerlach, H. Viruses. In: Avian medicine: principles and application. B. 47. Krautwald-Junghanns, Μ E., E. F. Kaleta, R. E. Marschang, and K.
W. Ritchie, G. J. Harrison, and L. R. Harrison, eds. Wingers, Lake Worth, Pieper. Untersuchungen zur Diagnostik und Therapie der Papillomatose des
Fl. pp. 862-948. 1994. aviaren Gastrointestinaltrakts. Tieraerztl. Prax. 28:272-278. 2000.
24. Gomez-Villamandos, J. C., E. Mozos, M A. Sierra, and A. Fernandez. 48. Lancz, G. J. Rapid method for the concentration and partial purification
Mortality in psittacine birds resembling Pacheco’s disease in Spain. Avian of herpes simplex viruses types 1 and 2. Arch. Gesamte Virusforsch.
Pathol. 20:541-547. 1991. 42:303-306. 1973.
25. Gough, R. E., and D. J. Alexander. Pacheco’s disease in psittacine birds 49. Lee, L. F., R. L. Armstrong, and K. Nazerain. Comparative studies of
m Great Britain 1987 to 1991. Vet. Rec. 132:113-115. 1993. six avian herpesviruses. Avian Dis. 16:799-805. 1972.
26. Gravendyck, M, S. Tritt, H. Spenkoch-Piper, and E. F. Kaleta. 50. Louzis, C., and J. P. Gollet. Mortalite d’etiologie complexe dans une
Antigenic diversity of psittacine herpesviruses: cluster analysis of antigenic population de psittacides: epidemiologie, clinique et recherche etiologique.
differences obtained from cross-neutralization tests. Avian Pathol. 25:345- Rev. Med. Vet. (Toulouse) 136:639-643. 1985.
357. 1996. 51. Mare, C. J., and D. L. Graham. Falcon herpesvirus, the etiologic agent
27. Gunther, B. M F., B. G. Klupp, M Gravendyck, J. E. Lohr, T. C. of inclusion body disease of falcons. Infect. Immun. 8:118-126. 1973.
Mettenleiter, and E. F. Kaleta. Comparison of the genomes of 15 avian 52. Murphy, F. A., C. M. Fauquet, D. H. L. Bishop, S. A. Ghabrial, A. W.
Jarvis, G. P. Martelli, M A. Mayo, and M D. Summers, eds. Virus

114
Chapter 24 Herpesviruses of Free-living and Pet Bink

taxonomy. In: Classification and nomenclature of viruses. Sixth Report of 65. Thiry, E., H. Vindevogel, P. Leroy, P.-P. Pastoret, A. Schwers, BL
the International Committee on Taxonomy of Viruses. Springer-Verlag, Brochier, Y. Anciaux, and P. Hoyois. In vivo and in vitro effect of acydovir
Vienna, Austria, pp. 114-127. 1995. on pseudorabies virus, infectious bovine rhinotracheitis virus and pigeon
53. Norton, T. M, J. Gaskin, G. V. Kollias, B. Homer, C. H. Clark, and R. herpesvirus. Ann. Rech. Vet. 14:239-245. 1983.
Wilson. Efficacy of acyclovir against herpesvirus infection in Quaker 66. Tomaszewski, E. K., G. van Wilson, W. L. Wigle, and D. N. Phaten
parakeets. Am. J. Vet. Res. 52:2007-2009. 1991. Detection of hetero-geneity of herpesviruses causing Pacheco’s disease b
54. Pacheco, G., and O. Bier. Epizootie chez perroques du Bresil. Relations parrots. J. Clin. Microbiol. 39:533-538. 2001.
avec la psittacose. C. R. Seances Soc. Biol. 105:109-111. 1930. 67. Tomaszewski, E. K., E. F. Kaleta, and D. N. Phalen. Molecular
55. Phalen, D. N., E. Tomaszewski, and G. Wilson. Internal papillomatosis: phylogeny of the psittacid herpesviruses causing Pacheco’s disease
A herpesvirus connection? Proc. Ann. Conf. Assoc. Avian Vet., pp. 45-46. correlation of genotype with phenotypic expression. J. Virol. 77:11260-
1998. 11267. 2003.
56. Plummer, P. J., T. Alefantis, S. Kaplan, P. O’Connell, S. Shawky, and 68. Tomaszewski, E. K., M Gravendyck, E. F. Kaleta, and D. N. Phalen.
K. A. Schat. Detection of duck enteritis virus by polymerase chain reaction. Genetic characte-rization of a herpesvirus isolate from a superb starling
Avian Dis. 42:554-564. 1998. (Lamprotornis superbus) as a psittacid herpesvirus genotype 1. Avian Dis.
57. Potgieter, L. N. D., A. A. Kocan, and K. M Kocan. Isolation of a 48:212-214. 2004.
herpesvirus from American kestrel with inclusion body disease. J. Wildl. 69. Tsai, S. S., J. H. Park, K. Hirai, and C. Itakura. Herpesvirus infections in
Dis. 15:143-149. 1979. psittacine birds in Japan. Avian Pathol. 22:141-156. 1993.
58. Purchase, H. G., C. J. Mare, and B. R. Burmester. Antigenic comparison 70. Van Deventer, D. R., P. Warrener, L. Bennett, E. R. Schultz, S. Coulter,
of avian and mammalian herpesviruses and protection tests against Marek’s R. L. Garber, and T. M Rose. Detection and analysis of diverse herpesviral
disease. In: Proceedings of the 76th Annual Meeting U. S. Animal Health species by consensus primer PCR. J. Clin. Microbiol. 34:1666-1671. 1996.
Association, Miami Beach, Fla., Nov. 5-10. pp. 484-^492. U.S. Animal 71. Vindevogel, H., L. Dagenais, B. Lansival, and P. P. Pastoret. Incidence
Health Association, Richmond, Va. 1972. of rotavirus, adenovirus and herpesvirus infection in pigeons. Vet. Rec.
59. Ramis, A., D. Fondevila, J. Tarres, and L. Ferrer. Immunocytochemical 109:285-286. 1981.
diagnosis of Pacheco’s disease. Avian Pathol. 21:523-527. 1992. 72. von Rotz, A., A. Ruebel, F. Mettler, and R. Hoop. Letal verlaufende
60. Roizman, B. The family herpesviridae: general description, taxonomy, Herpesvirusinfektion bei Goldsamadinen (Chloebia Gouldinae [Gould]).
and classification. In: The herpesviruses, vol. 1. B. Roizman, ed. Plenum Schweiz. Arch. Tierheilkd. 126:651-658. 1984.
Press, New York, N. Y. pp. 1-23. 1982. 73. Wagner, U. Vergleichende Untersuchungen zur Empfindlichkeit
61. Schoenbauer, M, and H. Koehler, fiber eine Virusinfektion bei verschiedener aviarer Herpesvirusisolate gegeniiber chemischen
Prachtfinken (Estrildidae). Kleintierpraxis 27:149-152. 1982. Desinfektionsmitteln. Veterinarxy Medical Thesis. Giessen, Germany. 1993.
62. Simpson, C. F., J. E. Hanley, and J. M Gaskin. Psittacine herpesvirus 74. Ward, F. P., D. G. Fairchild, and J. V. Vuicich. Inclusion body hepatitis
infection resembling Pacheco’s parrot disease. J. Infect. Dis. 131:390-396. in prairie falcon. J. Wildl. Dis. 7:120-124. 1971.
1975. 75. Watanabe, T., and H. Ohmi Susceptibility of guinea fowl to the virus of
63. Smadel, J. E., E. B. Jackson, and J. W. Harman. A new virus disease of infectious laryngotracheitis and egg-drop syndrome—1976. J. Agric. Sci.
pigeons. I. Recovery of the virus. J. Exp. Med. 81:385-398. 1945. Tokyo Nogyo Daigaku 28:193-200. 1983.
64. Styles, D. K., E. K. Tomaszewaski, L. A. Jaeger, and D. N. Phalen. 76. Wellehan, J. F. X., M Gagea, D. A. Smith, W. M Taylor, Y. Berhane,
Psittacid herpesviruses associated with mucosal papillomas in neotropical and D. Bienzle. Characterization of a herpesvirus associated with tracheitis
parrots. Virology 325:24-35. 2004. in Gouldian finches (Erythrura [Chloebia} gouldiae). J. Clin. Microbiol.
41:4054-4057. 2003.
Π. York, S. M, and C. J. York Pacheco virus vaccine studies—A
preliminary report. In: Proceedings of the 32rd Western Poultry Disease
Conference, Davis, Calif. February 8-10. pp. 101-102. 1983.

115
25
POX
Deoki N. Tripathy and Willie M. Reed

SUMMARY. Avian pox is a common viral disease of domestic birds (chickens, turkeys, pigeons, canaries) and wild birds. Approximately
232 species in 23 orders of birds have been reported to acquire natural poxvirus infection. Avian pox is a slow-spreading disease
characterized by the development of proliferative skin lesions (cutaneous form) and/or upper digestive and respiratory tract lesions
(diphtheritic form). The causative agent is a double-stranded DNA virus of the genus Avipoxvirus of the family Poxviridae.
Agent Identification. Pox is routinely diagnosed by histologic examination of proliferative skin, oral, or tracheal lesions for cytoplasmic
inclusion bodies in tissue sections, or viral particles exhibiting typical poxvirus morphology using electron microscopy. The virus is isolated
by inoculation of chorioallantoic membranes (CAMs) of 9 to 12-day embryonating chicken eggs. Pocks develop on the CAM in 5-7 days.
The etiology is confirmed by histopathologic or ultrastructural examination of the CAM lesions for cytoplasmic inclusions or viral particles
with typical poxvirus morphology, respectively.
Immunoperoxidase or indirect fluorescent antibody and agar-gel immunodiffusion (AGID) tests can also be used to detect pox viral antigen
in tissue samples. Antigenic differences among isolates can be determined by immunoblotting, host pathogenicity, and virus neutralization.
Isolated viruses can be evaluated for pathogenicity for susceptible hosts or for susceptibility in avian cell culture. Molecular characterization
is done by restriction fragment length polymorphism (RFLP) analysis of viral genome, polymerase chain reaction (PCR) amplification of
specific genomic fragments and nucleotide sequence determination of the genome or selected fragments.
Serologic Detection in the Host. Serologic detection of infection may be important in experimental studies and for measuring the immune
responses following vaccination. Antibody responses can be measured by AGED, passive hemagglutination, immunoperoxidase, and enzyme-
linked immunosorbent assay (ELISA). Monoclonal antibodies against fowlpox have been used in experimental studies to differentiate strains
of fowlpox virus.
Modified live virus vaccines (fowl pox, pigeon pox, canary pox, turkey pox, and quail pox viruses) of chick-embryo or cell-culture origin
are available commercially.

INTRODUCTION becomes yellowish brown to dark brown. Removal of such lesions


before they are not completely dry leaves a hemorrhagic, moist
Approximately 232 species in 23 orders of birds have been surface. When the scab is dry it drops off, leaving a scar.
reported to acquire a natural poxvirus infection (2). Although the In the diphtheritic form of pox, lesions occur on the mucous
number of distinct causative agents is unknown, all of them belong membrane of the mouth, nares, pharynx, larynx, esophagus, or
to the genus Avipoxvirus of the family Poxviridae. This genus trachea. Mouth lesions can interfere with feeding. Tracheal lesions
includes fowl, turkey, pigeon, canary, and quail viruses, as well as can cause difficulty in breathing and may simulate signs of
other avianpox viruses. Fowlpox virus is a common pathogen of infectious laryngotracheitis in chickens. In layers, the disease causes
chickens and turkeys. The infection is generally manifested as either a drop in egg production, and in young chicks it reduces growth.
cutaneous or diphtheritic forms, although both types of the disease Death occurs in cases with the generalized infection or diphtheritic
may occur in the same bird. In addition, an acute systemic form of form of the disease. The acute systemic disease caused by canary
pox is seen in canaries and results in high mortality. All forms of poxvirus results in high mortality in susceptible canaries. Although
the disease (acute systemic, cutaneous, and diphtheritic) occurring all avian species appear susceptible to pox (29), only fowl poxvirus
either singly or in combination have been observed in canaries (4). has been studied extensively.
In the cutaneous form, proliferative lesions are primarily confined
to unfeathered areas of skin (e.g., legs, head, and eyelids), whereas SAMPLE COLLECTION
in the much more lethal diphtheritic form, the lesions are found in
the mouth, esophagus, and trachea. The acute systemic form of pox Poxviruses are readily isolated from the nodular lesions of infected
in canaries is characterized by fibrinous inflammation of serous birds. Tissues with lesions, preferably recently developed lesions,
membranes, pulmonary edema, and fibrinous pneumonitis. Because can be removed with sterile scissors and forceps by cutting deep
of the economic importance of this disease in commercial poultry, into the epithelial tissue. The material is ground with either sterile
fowl pox virus and pigeon pox virus vaccines of chorioallantoic fine sand or 60-mesh aluminum oxide (Norton Alundum; Fisher
membrane (CAM) or cell-culture origin have been used for more Scientific Co., Pittsburgh, PA) with a sterile mortar and pestle or in
than 60 yr. In recent years, turkey, canary and quail virus vaccines a glass grinder. Hanks’ balanced salt solution, saline solution, or
have also become available commercially. In spite of regular broth is added to make a 10% suspension. The suspension is
vaccination, outbreaks of fowlpox have occurred in all regions of centrifuged for 10 min at about 700 x g to remove large tissue
the US in previously vaccinated chicken flocks. particles. Antibiotics (penicillin and streptomycin) are added to the
supernatant to give respective final concentrations of 1000 IU/ml
CLINICAL DISEASE and 1 mg/ml, and the suspension is held at room temperature for 30
min to 1 hr before inoculation. A piece of tissue should also be
The cutaneous form of pox is characterized by development of collected (in 10% neutral buffered formalin) for histopathologic
nodular lesions on various parts of unfeathered skin. A mild form of examination to reveal cytoplasmic inclusion bodies or for
the disease may remain unnoticed, with only small focal lesions, transmission electron microscopy (TEM) to demonstrate virus
usually on the comb and wattles. In the severe form of the disease, particles by negative staining or in ultrathin sections. Serum
generalized lesions may occur on any part of the body, such as the samples may be collected to determine previous exposure by
comb, wattle, comer of the mouth, around the eyelids, the angle of serologic tests.
the beak, the ventral surface of the wings, and the vent. Skin lesions
may be small and discrete or may involve large areas through the PREFERRED CULTURE MEDIA AND SUBSTRATES
coalescence of adjoining lesions. The surface of lesions is moist for
a short time but dries soon, with a rough, irregular surface that Poxviruses can be isolated by inoculating the suspect material in
116
Chapter 25 Pox

embryonating eggs, susceptible birds, or cell cultures (e.g., chick fluorescent antibody and immunoperoxidase methods (27). Viral
embryo kidney cells, chick embryo dermal cells, or quail cells). antigen can be detected in lesions by indirect fluorescent antibody
or immunoperoxidase tests using specific antiserum.
Embryo inoculation A quick method for diagnosis of avian pox uses simultaneous
The preferred and most convenient hosts are 9 to 12-day-old fixation and dehydration and demonstrates cytoplasmic inclusion in
embryonating chicken embryos from a specific-pathogen-free flock. less than 3 hr (17). Elementary bodies can be demonstrated in
About 0.1 ml of the virus suspension is inoculated on the CAM. impression or squash-preparation smears stained by the Gimenez
Inoculated embryos are incubated at 37 C for 5-7 days and then method (25). Negative-contrast TEM can be used for direct
examined for pocks on the CAM. Pox lesions appear as either focal demonstration of viral particles in clinical material, pocks in CAM,
white opaque pocks or a generalized thickening of the CAM. or cultured cells (11).
Specific diagnosis requires either histologic demonstration of
cytoplasmic inclusion bodies, or viral particles by TEM, or Antigen Detection in Tissues
detection of specific poxvirus antigen in the CAM lesions. Poxviral antigens can be detected in cytoplasmic inclusions of
Occasionally, some strains from wild and pet birds fail to grow on infected cells (cutaneous or cultured) by direct or indirect
the CAM and may require adaptation. immunofluorescent or immunoperoxidase microscopy using
specific primary antiserum. Appropriate samples would include
Chicken inoculation fowlpox-infected CAM, skin, or cell culture (27). The
The suspension made from a lesion is either applied to the comb, immunoperoxidase technique has the advantage of using a light
which is then scarified, or applied with a brush to denuded feather microscope and sections can be stored for more than 2 mo without
follicles (pluck 5 or 6 feathers). Similarly, wing web is another loss of staining.
suitable site that can be used for inoculation. Susceptible chickens
develop lesions at the site of inoculation in 5-10 days. Later, Molecular Identification
generalized lesions may appear during the disease, especially with Restriction fragment length polymorphism has been used for
virulent field strains. The disease is more severe in susceptible comparing closely related DNA genomes (7, 16, 24). In this regard,
young birds than in older birds. genomic profiles of quail, canary, mynah and poxviruses from
Hawaiian endangered forest birds are different from those of fowl
Cell culture poxvirus (9, 24,28).
Primary chick embryo, chick embryo kidney, chick embryo Cloned genomic fragments of fowl poxvirus can be used
dermis, duck embryo, and secondary quail (QT-35) or LMH cells effectively as nucleic acid probes for diagnosis (5). In this
(16) support the growth of fowl poxvirus, producing a cytopathic procedure, viral DNA isolated from legions is hybridized either with
effect (CPE), usually in 4-6 days, although CPE may occur earlier 32P-labeled or nonradioactive-labeled genomic probes. This method
with high-titer inocula. The cells often become round and refractile, is especially useful for differentiation of the diphtheritic form of
followed by degeneration. The virus produces plaques in agar- fowlpox from infectious laryngotracheitis when tracheal lesions are
overlaid monolayers of chick embryo cells or quail (QT-35) cells. present (5).genomic DNA sequences of various sizes can be
Plaques are produced in 3-5 days in QT-35 cells. Adaptation of amplified by polymerase chain reaction (PCR) using specific
strains to cell culture is necessary for plaque formation, because not primers (8, 10, 18, 24). This technique is very sensitive, especially
all strains form plaques. The method has been considered unsuitable when an extremely small amount of virus is present in the sample.
for titration of fowl poxvirus, because it is less sensitive than pock Complete nucleotide sequence of a vaccine like fowlpox virus (1)
formation on the CAM (12). and canarypox virus (30) has been determined. Molecular analysis
reveals that while most strains of fowlpox virus from field isolates
AGENT IDENTIFICATION have integrated reticuloendotheliosis virus (REV) in their genome,
the vaccine strains of fowlpox virus contain remnants of long
Physicochemical, Morphologic, and Biological Properties terminal repeats (LTR) of REV (6, 18, 19, 23).
Fowl poxvirus, the type species of the genus Avipoxvirus, is a
brick-shaped to rectangular virus. Other species of the genus are SEROLOGICAL DETECTION IN THE HOST
morphologically similar. Complete nucleotide sequence of the
genomes of fowlpox virus and canary pox virus has been Agar Gel Immunodiffusion Test
determined. The genome of fowl poxvirus is composed of a single Precipitating antibodies can be detected by reacting test sera against
double-stranded DNA molecule of approximately 288 kilobases (1). viral antigens (25). The antigen can be derived by sonication and
The genome of canary poxvirus is 365 kbp (30). homogenization of infected skin or CAM lesions, as well as by
Fowl poxvirus is resistant to ether. It is inactivated by 1% caustic treatment of infected cell cultures as described for ELISA. The
potash when separated from its matrix. It withstands 1% phenol and lysed cell suspension is centrifuged and the supernatant is used as
0.1% formalin for 9 days. Inactivation occurs by heating at 50 C for antigen. Gel-diffusion medium is prepared with 1% agar, 8%
30 min or 60 C for 8 min. Trypsin has no effect on the DNA or sodium chloride, and 0.01% thimersol. The viral antigen is placed
whole virus. Fowl poxvirus can survive in dried scabs for months or in the central well and the test sera are placed in the peripheral
even years. In this regard, the presence of photolyase gene in the wells. It is important to include a positive and negative control
genome of fowlpox virus appears to play some role in virus serum. The plates are incubated at room temperature. Precipitation
persistence (21, 22). The virus multiplies in the cytoplasm of lines develop in 24-48 hr after incubation of the antigen with
infected cells and forms inclusion bodies (Bollinger bodies) that antibody to homologous or closely related strains. The test is less
contain the elementary bodies (Borrel bodies). It causes sensitive than the ELISA (3) or die passive hemagglutination test
proliferation of the epithelium with ballooning degeneration of the (26, 29).
cells and ultimately epithelial cell death. The cytoplasmic type-A
inclusions vary in size and shape and can be demonstrated in Passive Hemagglutination
histologic sections of skin, or in diphtheritic or CAM lesions. The Antibodies against fowl poxvirus can be measured by a passive
inclusions can be stained by hematoxylin and eosin, acridine hemagglutination test using tanned horse or sheep erythrocytes (26)
orange, and Giemsa stains and the Feulgen reactions. The coated with fowl poxvirus antigen. Passive hemagglutinating
specificity of the viral inclusions can be determined also by antibodies are detectable in some sera of infected birds as early as
117
Deoki N. Tripathy and Willie M. Reed

1 wk after inoculation and may persist for 15 wk; that is, longer REFERENCES
than precipitating antibodies. Cross-reactions occur among avian 1. Afonso, C. L., E. R Tulman, Z. Lu, L. Zsak, G. F. Kutish, and D. L.
poxviruses. Rock. The genome of fowlpox virus. J. Virol. 74:3815-3831, 2000.
2. Bolte, A. L., J. Meurer, and E. F. Kaleta. Avian host spectrum of
avipoxviruses. Avian Pathol. 28:415-432, 1999.
Virus Neutralization 3. Buscaglia, C., R. A. Bankowski, and L. Miers. Cell-culture virus­
Virus-neutralizing antibodies usually appear 1-2 wk after natural neutralization test and enzyme-linked immunosorbent assay for evaluation
infection or experimental inoculation. The antibody response can be of immunity in chickens against fowlpox. Avian Dis. 29:672-680, 1985.
measured either by pock reduction in CAMs or plaque reduction in 4. Donnelly, T. M., and L. A Crane. An epomitic of avian pox in a research
cell cultures (12). aviary. Avian Dis. 28:517-525,1984.
5. Fatunmbi, Ο. O., W. M Reed, D. L. Schwartz, andD. N. Tripathy. Dual
ELISA infection of chickens with pox and infectious laryngotracheitis (ILT)
confirmed with specific pox and ELT DNA DOT-BLOT hybridization
ELISA is a sensitive method to measure humoral antibody
assays. Avian Dis. 39:925-930,1995.
responses (3), and antibodies can be detected by 7 days after 6. Garcia, Μ, N. Narang, W. M Reed, and A. M Fadly. Molecular
infection. Fowlpox virus antigens are prepared from either infected characterization of reticuloendotheliosis virus insertions in the genome of
QT-35 cell monolayers or CAM lesions. Infected cells are pelleted field and vaccine strains of fowlpox virus. Avian Dis. 47:343-354,2003.
(700 x g for 10 min at 4 C), washed with isotonic buffer (10 mM 7. Ghildyal, N., W. M Schnitzlein, and D. N. Tripathy. Genetic and
Tris, pH 8.0, 150 mM NaCl, 5 mM ethylenediamine tetraacetic acid antigenic differences between fowlpox and quailpox viruses. Arch. Virol.
[EDTA]) followed by lysis in hypotonic buffer (10 mM Tris, pH 106:85-92,1989.
8.0, 10 mM KC1, 5 mM EDTA) containing 0.1% Triton X-100 and 8. Kim, T. J., and D. N. Tripathy. Reticuloendotheliosis virus integration in
the fowlpox virus genome: not a recent event. Avian Dis. 45:663-669,2001.
0.025% beta-mercaptoethanol. Nuclei and cellular debris are
9. Kim, Tae-Joong, W. M Schnitzlein, D. McAloose, A. P. Pessier, and D.
removed by low-speed centrifugation (500 x g for 5 min at 4 C) and N. Tripathy. Characterization of an avian pox virus isolated from an Andean
the resulting supernatant is used as a source of fowlpox virus Condor Vultur gryphus. Veterinary Microbiology 96:237-246, 2003.
antigens for ELISA or immunoblotting. To isolate viral antigen 10. Lushow, D., T. Hoffman, and Η. M Hafez. Differentiation of avianpox
from CAM lesions, initial grinding of the lesions with subsequent virus strains on the basis of nucleotide sequences of the 4b gene fragment.
detergent treatment as described earlier would be required. Virus Avian Dis. 48:453-463, 2004.
propagated in chicken embryo fibroblasts and chicken embryo 11. McFerran, J. B., J. K. Clarke, and W. L. Curran. The application of
dermis cells has also been used for antigen. negative contrast electron microscopy to routine veterinary virus diagnosis.
Res. Vet. Sci. 12:253-257,1971.
12. Morita, C. Studies on fowlpox viruses. Π» Plaque-neutralization test.
Strain Variability Avian Dis. 7:93-98,1973.
Fowlpox virus is the type species of the Avipoxvirus genus and 13. Moyer, R. W., Β. M Arif, D. N. Black, D. B. Boyle, R. M Buller, K. R.
has been studied more extensively than other members of this genus Dumbell, J. J. Esposito, G. McFadden, B. Moss, A. A. Mercer, S. Ropp, D.
(i.e., pigeon, canary, turkey, sparrow, junco, quail, mynah and other N. Tripathy, and C. Upton. Family Poxviridae. In: Virus Taxonomy:
avian poxviruses). Differentiation of fowl, turkey, canary, and Classification and Nomenclature of Viruses. Seventh Report of the
pigeon poxviruses on the basis of their pathogenicity for the host International Committeee on Taxonomy of Viruses. Academic Press, San
spectrum has been attempted. Although some strains show Diego, pp. 137-157,2000.
14. Reed, W. M, and Ο. O. Fatunmbi. Pathogenicity and immunological
pathogenicity for several host species, others either are host-specific
relationship of quail and mynah poxviruses to fowl and pigeon poxviruses.
or infect a limited number of species (14, 15). Because natural pox Avian Pathol. 22:395-400,1993.
infections have been reported in several species of wild birds of 15. Reed, W. M, and D. L. Schrader. Immunogenicity and pathogenicity of
different families, as well as in domestic birds, it appears that all mynah pox virus in chickens and bobwhite quail. Poult. Sci. 68:631-638,
avian species are susceptible to avian pox. 1989.
Minor antigenic differences in strains of fowl poxvirus can be 16. Schnitzlein, W. Μ, N. Ghildyal, and D. N. Tripathy. Genomic and
detected by sodium dodecyl sulfate-polyacrylamide gel antigenic characterization of avipoxviruses. Virus Res. 10:65-76,1988.
electrophoresis and immunoblotting (7, 9, 16, 24), although the 17. Sevoian, M A quick method for the diagnosis of avian pox and
infectious laryngotracheitis. Avian Dis. 4:474-477,1960.
majority of the antigens are cross-reactive. Antigenic profiles of
18. Singh, P., T. J. Kim, and D. N. Tripathy. Re-emerging fowlpox:
quail poxvirus and isolates from Hawaiian forest birds are evaluation of isolates from vaccinated flocks. Avian Pathol. 29:449-455,
remarkably different from fowl poxvirus. 2000.
Comparison of the genomic profile of fowl, pigeon, and junco 19. Singh, P., W. M Schnitzlein, and D. N. Tripathy. Reticuloendotheliosis
poxviruses after digestion of their DNA with restriction enzymes virus sequences within the genomes of field strains of fowlpox virus display
and agarose gel electrophoresis revealed that the majority of DNA variability. J. Virol. 77:585-586, 2003.
fragments co-migrate. However, most isolates could be 20. Singh, P., T.-J. Kim, and D. N. Tripathy. Identification and
distinguished by the presence or absence of one or more DNA characterization of fowlpox virus strains using monoclonal antibodies. J.
Vet. Diag. Invest. 15:50-54, 2003.
fragments (9, 16, 24). On the other hand, genomic profiles of quail,
21. Srinivasan, V., W. M Schnitzlein, and D. N. Tripathy. Fowlpox virus
canary, mynah and Hawaiian bird poxviruses are different from encodes for a novel DNA repair enzyme that restores infectivity of UV-light
profiles of fowl poxvirus (7, 9, 28). damaged virus. J. Virol. 75:1681-1688,2001.
22. Srinivasan, V., and D. N. Tripathy. The DNA repair enzyme, CPD-
DIFFERENTIATION FROM CLOSELY RELATED AGENTS photolyase restores the infectivity of UV-damaged fowlpox virus isolated
from infected scabs of chickens. Veterinary Microbiology 108:215-223,
The diphtheritic form of pox in birds with associated respiratory 2005.
signs must be differentiated from other respiratory infections, 23. Tadese, T., and W. M Reed. Detection of specific reticuloendotheliosis
virus sequence and protein from REV-integrated fowlpox virus strains. J.
especially infectious laryngotracheitis, an infection caused by a
Virol. Meth. 110(l):99-104, June 9, 2003.
herpesvirus that produces intranuclear inclusions (5). Oral lesions 24. Tadese, T., and W. M Reed. Use of restriction fragment length
caused by vitamin A, pantothenic acid and biotin deficiency in polymorphism, immunoblotting and polymerase chain reaction in the
young chicks or by T-2 toxin may be mistaken for pox lesions. differentiation of avianpox viruses. J. Vet. Diag. Invest. 15:141-150,2003.
Diphtheritic pox lesions in doves and pigeons may be mistaken for 25. Tadese, T., E. A. Potter, and W. M Reed. Development of a mixed agar
lesions of Trichomonas gallinae, which are diagnosed by gel enzyme assay (AGEA) for the detection of antibodies to poxvirus in
microscopic examination of smears or by culture (29). chicken and turkey sera. J. Vet. Med. Sci. 65:255-258,2003.

118
Chapter 25 Pox

26. Tripathy, D. N., and L. E. Hanson. A smear technique for staining 30. Tripathy, D. N., and W. M Reed. Pox. In: Diseases of Poultry, 11th Ed.
elementary bodies of fowlpox. Avian Dis., 20:609-610,1976. Y. M. Saif, H. J. Bames, B. W. Calneck, J. R. Glisson, A. M Fadly, L. R.
27. Tripathy, D. N., L. E. Hanson, and W. L. Myers. Passive McDougald, and D. E. Swayne, Eds. Iowa State University Press, Ames, pp.
hemagglutination test with fowlpox virus. Avian Dis. 14:29-38,1970. 253-269, 2003.
28. Tripathy, D. N., L. E. Hanson, and A. H. Killinger. Immunoperoxidase 31. Tulman, E. R., C. L. Afonso, Z. Lu, G. F. Kutish, and D. L. Rock. The
technique for detection of fowlpox antigen. Avian Dis. 17:274-278,1973. genome of canarypox virus. J. Virol. 78:353-366,2004.
29. Tripathy, D. N., W. M Schnitzlein, P. J. Morris, D. L. Janssen, J. K.
Zuba, G. Massey, and C. T. Atkinson. Characterization of poxviruses from
forest birds in Hawaii. J. Wildl. Dis. 36:225-230,2000.

119
26
BUDGERIGAR FLEDGLING DISEASE AND OTHER
AVIAN POLYOMAVIRUS INFECTIONS
Branson W. Ritchie and Phil D. Lukert

SUMMARY. Avian polyomavirus, which belongs to the family Papovaviridae, genus Polyomavirus, causes acute, highly fatal disease in
many young psittacines.
Agent Identification. Detection of target segments of viral nucleic acid is most easily made by submitting tissues or swabs for polymerase
chain reaction (PCR) based testing. Culture of the virus may be done in budgerigar embryo cell cultures initially, followed by culture in
chicken embryo fibroblast cell cultures.
Serologic Detection in the Host. The virus-neutralization test is the serologic test of choice. Confirmation of disease in birds with
characteristic clinical or gross lesions requires demonstration of viral nucleic acid by in situ hybridization, immunohistochemical staining for
viral proteins or documentation of viral particles by electron microscopy.

INTRODUCTION Birds recovering from the chronic form appear normal, although
some may die months later from kidney failure (4).
Avian polyomavirus (family Papovaviridae, genus Polyomavirus)
causes an acute, highly fatal disease in budgerigar fledglings as well SAMPLE COLLECTION
as peracute, acute, or chronic infections of many other species of
psittacine birds. Disease is most common in young birds but has also For virus isolation a pool of liver, kidney, spleen, and heart tissues
been documented in adults. In budgerigars the mortality rate can vary should be collected and submitted frozen in a plastic bag. Submit
from 25 to 100%. Substantial evidence exists that the virus is egg- tissue from dying or recently dead birds. Serum samples for serology
transmitted, because intranuclear inclusion bodies are observed in 1- may be collected in capillary tubes, centrifuged to separate the
day-old fledglings. All psittacines should be considered susceptible erythrocytes, and stored or shipped under refrigeration (4).
to natural infection, but some species develop clinical disease more For the detection of viral DNA by PCR-based testing, cloacal swabs
readily than others. Passeriformes and some species of from live birds or swabs of tissue sampled at necropsy are preferred.
Falconiformes are also affected by avian polyomavirus. Gallinaceous Swabs used to culture bacteria are adequate and should be encased in
birds are susceptible to natural infection by avian polyomavirus, but a protective holder. Swabs used for anaerobic bacterial culture with
clinical disease has only been described experimentally in severely gel-type transport media may interfere with the assay and should not
immunocompromised animals. Antibodies generated against be used (4).
psittacine polyomavirus cross-react with the polyomavirus of
finches; however, nucleic acid sequences of these viruses differ. PREFERRED CULTURE MEDIA AND SUBSTRATES
Avian polyomavirus differs from mammalian polyomaviruses in
pathogenicity. Polyomaviruses of mammals typically produce The method of choice for isolation of virus is described by
disease only in immunocompromised hosts. Bozeman et al (2). A 10% homogenized tissue suspension in cell
culture medium containing antibiotics is prepared, and a monolayer
CLINICAL DISEASE of budgerigar embryo fibroblast (BEF) cells is inoculated. After
several passages in BEF cells at 4- or 5-day intervals, most isolates
Budgerigars will replicate in chick embryo fibroblast (CEF) cell cultures. The
Acute fatal infections usually occur in fledglings from 1 to 15 days BEF cell cultures are prepared from 10 to-16-day-old budgerigar
of age. Affected fledglings may die suddenly with no premonitory embryos. The medium used is 60% Ham’s F-10 with glutamine and
signs or they may exhibit abdominal distention, subcutaneous 40% M-199 with Hanks’ salts with glutamine. This medium is
hemorrhages, and reduced feather formation. Some birds develop enriched with 4% tryptose phosphate broth and 5% fetal bovine
neurologic signs with ataxia and tremors of the head and neck serum for growth. Maintenance medium has the fetal bovine serum
several days before death. The death rate is highest in birds affected reduced to 2%.
at an age of less than 15 days. After 15 days, birds that develop
clinical signs may survive and develop feather abnormalities. Avian AGENT IDENTIFICATION
polyomavirus is considered to be one cause of French molt in
budgerigars (1,3). Cell Culture Properties
Cells with enlarged nuclei are first seen 48 hr postinfection with
Nonbudgerigar Psittacine Birds avian polyomavirus. By 72-96 hr, the cells begin to round up and are
Both young and adult nonbudgerigar psittacine birds can experience swollen, with greatly enlarged nuclei. Cells fixed and stained with
clinical infections with avian polyomavirus. Although most Giemsa stain exhibit large, clear intranuclear inclusion bodies with
infections are subclinical, the disease can be severe and often fatal in margination of the chromatin. Infectivity titrations of the virus using
young birds. The peracute form with no premonitory signs is most cytopathic effect (CPE) as the endpoint are held for 7-9 days.
common in young birds and as the age increases the acute form is Titration times may be reduced to 72 hr if immunofluorescence is
usually observed. The signs include depression, anorexia, weight used to detect viral infection.
loss, diarrhea, subcutaneous hemorrhages, and dehydration. Birds
normally die within 12^18 hr after developing clinical signs. Polymerase Chain Reaction
Posterior paresis and paralysis within 18 hr of the onset of the A DNA probe based test has been developed and is commercially
disease is suggestive of polyomavirus infections. Eclectus parrots available (Infectious Diseases Laboratory, Athens, GA.). The DNA
and caiques appear to be highly susceptible to disease (4). probe based test has been shown to be sensitive and specific (1). The
In addition to the peracute and acute forms a chronic/progressive sample of choice for detecting birds that are shedding viral nucleic
disease has been described and is typified by weight loss, polyuria, acid is a cloacal swab. The same test can be used to detect viral
poor feather formation, and recurring bacterial or fungal infections. nucleic acid in whole unclotted blood. A swab of the cut surface of
the spleen, liver, and kidney can be submitted for PCR-based testing
120
Chapter 26 Budgerigar Fledgling Disease and Other Avian Polyomavirus Infections

to document the presence of viral nucleic acid in birds that have died DIFFERENTIATION FROM CLOSELY RELATED AGENTS
with suspicious clinical and gross lesions.
Because of the feathering abnormalities in chronic infections,
Physiochemical Properties polyomavirus infection may be confused with psittacine beak and
Virions have a buoyant density of 1.34 g/cm3 in CsCl and a mean feather disease. French molt can be caused by psittacine beak and
diameter of 42 mm (2). The virions are nonenveloped and contain a feather disease virus, polyomavirus, or both. However, no beak
4981-bp double-stranded DNA genome. Growth in cell culture is lesions have been associated with polyomavirus infections. In
inhibited by 5-iododeoxyuridine. Freezing and thawing of the virus addition, feather loss is not as extensive in polyomavirus infections
for five cycles has no effect on infectivity. The virus is relatively as it is in psittacine beak and feather disease. Inclusion bodies
heat resistant. Incubation of the virus at 56 C for 30 min reduces the associated with both viruses are different; those of polyomavirus are
titer by 0.6 log10, and 56 C for 90 min reduces the titer by 1.0 logi0. large, clear intranuclear inclusions and those of beak and feather
disease virus are more basophilic and occur in the nucleus and
SEROLOGIC DETECTION IN THE HOST cytoplasm. In situ hybridization can also be used to differentiate
these viruses.
A virus-neutralization (VN) test is used for the detection of
antibodies to avian polyomavirus. The test is performed in 96-well REFERENCES
microtiter plates. Virus adapted to CEF cells is used in the test. A
100 mean tissue-culture infective dose (TCID50) dose of virus is used 1. Bernier, G., M Morin, and G. Marsolais. Papovavirus induced feather
abnormalities and skin lesions in the budgerigar: clinical and pathological
and twofold dilutions of serum added. The virus-serum mixture is
findings. Can. Vet. J. 25:307-310. 1984.
incubated 30-60 min at 37 C and then a suspension of CEF cells is 2. Bozeman, L. H., R. B. Davis, D. Gaudry, P. D. Lukert, O. J. Fletcher, and
added in growth medium. After 72 hr the endpoint may be read by M J. Dykstra. Characterization of a papovavirus isolated from fledgling
staining with a fluorescein-isothiocyanate-labeled anti-polyoma virus budgerigars. Avian Dis. 25:972-980. 1981.
globulin or the cells can be held for 7-9 days to read the endpoint by 3. Davis, R. B., L. H. Bozeman, D. Gaudry, O. J. Fletcher, P. D. Lukert, and
CPE. Serum endpoints are determined as the last dilution of serum M J. Dykstra. A viral disease of fledgling budgerigars. Avian Dis. 25:179-
that prevents detectable infection of the cells. Serum titers of 1:20 or 183. 1981.
greater are considered to be positive for VN antibodies. No evidence 4. Ritchie, B. W. Papovaviaridae. In. Avian Viruses: Function and Control,
1st ed. Wingers Publishing, Inc., Lake Worth, Fla. pp. 127-170. 1995.
of varying serotypes has been shown with avian polyomaviruses of
5. Wainwright, P. Ο., P. D. Lukert, R. B. Davis, and P. Villegas. Serological
psittacine birds (5). evaluation of some Psittaformes for budgerigar fledgling disease virus. Avian
Dis. 31:673-676. 1987.
27
PSITTACINE BEAK AND FEATHER DISEASE
Branson W. Ritchie and Phil D. Lukert

SUMMARY. Psittacine beak and feather disease can be an acute or chronic disease of psittacine birds and is characterized by necrotic and
abnormally formed feathers, beak necrosis and fractures, and usually death. The disease is caused by a small virus in the family Circoviridae.
Agent Identification. The virus has not been cultured in vitro, but a DNA probe based test can be used to detect the target segments of viral
nucleic acid in infected birds. Virus purified from infected birds will agglutinate cockatoo erythrocytes. Confirmation of disease in birds with
characteristic clinical changes requires demonstration of viral nucleic acid by in situ hybridization, immunohistochemical staining for viral
proteins or documentation of viral particles by electron microscopy.
Serologic Detection in the Host. A hemagglutination-inhibition test can be used to detect antibodies to the virus, but it is of minimal value in
managing the disease in individual birds or in aviaries.

INTRODUCTION SAMPLE COLLECTION

Psittacine beak and feather disease (PBFD) is usually a chronic Samples that are useful for the detection of psittacine circovirus
disease of psittacine birds that eventually leads to death. It was first nucleic acid are white blood cells and swabs from feather pulp,
described in various species of Australian cockatoos in the early necropsy tissue, or environmental areas. Blood samples should be
1970s (4). The virus that causes this disease belongs to the family of collected in heparin (0.01 ml heparin to 0.49 ml of blood).
viruses called Circoviridae. The family includes viruses that infect Ethylenediaminetetraacetic acid or excessive heparin may result in
Psittaciformes, Columbiformes, Passeriformes, and Anseriformes false negative polymerase chain reaction (PCR) based tests. Clotted
and are the smallest known virus of animals with a 14-17 nm blood is unsuitable for nucleic acid detection. Swabs should be
diameter and circular single stranded DNA containing about 1700- encased in a swab holder and anaerobic swabs with gel-type
2300 bases. In comparison, parvoviruses are 25 nm in diameter and protective media are unacceptable. Serum samples for antibody
have a single- stranded linear DNA with 5000 bases. Psittacine detection by hemagglutination-inhibition (HI) tests may be submitted
circovirus has not been grown in vitro, but it has been characterized in microhematocrit capillary tubes after centrifugation to separate
by purifying the virus from the skin (feather follicles) of affected cells from the serum (1).
birds (2).
All psittacine birds are probably susceptible to the virus and the PREFERRED CULTURE MEDIA AND SUBSTRATES
outcome of infection is largely dependent upon the age of the bird
when infected. Young birds are more susceptible to disease after The virus has not been propagated in embryos or cell cultures, but
infection. Generally, the older the bird the longer the incubation can be identified and characterized by purifying the virus from
period between infection and appearance of disease. The disease homogenized preparations of skin from infected birds. The nucleic
usually occurs in birds less than 3 yr of age but birds up to 20 yr of acid can be extracted and identified by gel electrophoresis or the
age have been diagnosed with PBFD (1). virus visualized by electron microscopy.

CLINICAL DISEASE AGENT IDENTIFICATION

The incubation period for the disease is highly variable and may be Polymerase Chain Reaction (PCR)
influenced by maternal antibodies, virus dose, or the host response to A DNA probe based test has been developed and is available
the infection. The minimum incubation period is 3-4 wk. Virus is commercially (Infectious Diseases Laboratory, Athens, GA). The test
thought to spread by direct contact or through contaminated food and is sensitive and specific (1). Most infected birds clear an infection
water. Evidence exists that the virus can be transmitted vertically with no signs of disease. Birds with no feather abnormalities that or
through the egg (1). blood positive for viral nucleic acid should be retested in 90 days. A
The first clinical sign of disease is the appearance of necrotic or negative test in the absence of clinical changes suggests that the bird
abnormally formed feathers. In young birds (less than 2 mo old), all has cleared the infection. A non-lory psittacine bird with a positive
of the feather tracts may be involved, whereas in older birds the test and developmental feather lesions should be considered infected
disease is more prolonged with progressive feather changes but disease can only be confirmed by histopathology. Lories have
occurring with ensuing molts. Peracute infections occur in neonates been shown to be susceptible to a variant of psittacine circovirus and
with signs of septicemia, pneumonia, enteritis, rapid weight loss, and birds with advanced feather abnormalities can recovery. Specific
death. The acute form is common in young birds during their first PCR-based testing is necessary to differentiate the two types of
feather formation. Usually several days of depression occur, psittacine circovirus that may infect lories and is also required to
followed by sudden changes in developing feathers, which include provide accurate prognostic information. Once a psittacine aviary is
necrotic, broken, bent, and hemorrhagic feathers. Crop stasis and free of PBFD virus a negative flock can be maintained through
diarrhea followed by death in 1-2 wk is a usual occurrence in the quarantine and testing procedures. Until a commercial vaccine is
acute form of the disease. available, PCR-based testing will remain the most valuable tool for
The chronic form of PBFD is characterized by symmetrical, the control of this disease.
progressive appearance of abnormal feathers during successive In situ hybridization can be used to confirm a diagnosis of PBFD
molts. The course of the disease may be protracted and the birds may and differentiate it from budgerigar fledgling disease, which is
experience secondary infections during this period. Birds with caused by avian polyomavirus infections (1).
varying degrees of feathering may live months to years before
eventual death, typically from a secondary infection. The beak Physicochemical Properties
lesions usually follow the feather malformations and changes include Virions are 14-17 nm in diameter and are nonenveloped. The viral
longitudinal fractures and necrosis of the palate area (1). DNA is single-stranded and circular, containing approximately 1800
bases. The virus has a density of 1.37 g/cm3 in CsCl (2), and is
highly resistant to heat and disinfectants. Chicken anemia virus,
122
Chapter 27 Psittacine Beak and Feather Disease

which is in the same family (Circoviridae), resists 80 C for 1 hr and PBFD virus. The PBFD inclusions tend to be basophilic and are
resists 5% phenol (5). found in the nucleus and cytoplasm, whereas polyomavirus produces
large clear intranuclear inclusions. In situ hybridization with viral
SEROLOGIC DETECTION IN THE HOST specific probes can be used to confirm a diagnosis (1).

The virus will hemagglutinate erythrocytes of cockatoos but not REFERENCES


those of sheep or chickens. An HI test can detect antibodies in
1. Ritchie, B. W. Circoviridae. In: Avian Viruses: Function and Control, 1st
infected birds and is typically used for epizootiological surveys. The
ed Wingers Publishing, Inc., Lake Worth, Fla. pp. 223-252. 1995.
HI test is, however, of minimal value in managing the disease in 2. Ritchie, B. W., F. Niagro, P. Lukert, W. Steffans and K. Latimer.
individual birds or in aviaries. Virus for the HI test is prepared by Characterization of a new virus from cockatoos with psittacine beak and
homogenizing feather follicle tracts from affected birds. The virus is feather disease. Virology 171:83-88. 1989.
concentrated on sucrose gradients followed by purification on CsCl 3. Ritchie, B. W., F. Niagro, K. Latimer, P.Lukert, and D. Pesti.
gradients (3). Hemagglutination by psittacine beak and feather disease virus and use of
hemagglutination-inhibition for detection of PBFD virus antibodies. Am J.
DIFFERENTIATION FROM CLOSELY RELATED AGENTS Vet. Res. 52:1810-1815. 1991.
4. Perry, R. A. A psittacine combined beak and feather disease syndrome. In:
Proceedings # 55 of Courses for Veterinarians, Sydney, Australia, pp. 81-
Initially the chronic forms of PBFD may be confused with avian 108. 1981.
polyomavirus (budgerigar fledgling disease). The differentiation of 5. Yuasa, N. Effect of chemicals on the infectivity of chicken anemia virus.
the two diseases may be readily made by PCR-based testing of white Avian Pathol. 21:315-319. 1992.
blood cells from the bird. In some cases the intranuclear inclusions of
avian polyomavirus can be differentiated from those induced by

123
28
CHICKEN ANEMIA VIRUS
M. Stewart McNulty and Daniel Todd

SUMMARY. Chicken anemia virus (CAV) causes a disease in 2 to 4 wk-old chickens characterized by increased mortality, anemia, thymic
atrophy, and subcutaneous hemorrhages. Disease occurs following vertical transmission of CAV from a breeder flock.
Agent Identification. Chicken anemia virus can be detected in affected chickens by virus isolation, immunocytochemical and
immunofluorescence techniques, and a variety of molecular methods that detect CAV DNA.
Serologic Detection in the Host. Enzyme-linked immunosorbent assay is the preferred method for detecting antibodies to CAV.

disease outbreaks have been made from liver, but the virus has also
INTRODUCTION been isolated from skin, spleen, thymus, lung, heart, bursa, muscle,
feces, and bone marrow. For virus isolation in susceptible chicks or
Chicken anemia virus (CAV), formerly known as chicken anemia in MDCC-MSB1 cells (a Marek’s disease virus transformed
agent, is a small DNA virus that causes a disease in chickens called chicken lymphoblastoid cell line), 20% tissue suspensions are made
blue wing, infectious anemia, hemorrhagic syndrome, or anemia using a stomacher or pestle and mortar in RPMI 1640 medium
dermatitis syndrome. This disease is characterized by aplastic (GIBCO-BRL Laboratories, Gaithersburg, Md.) containing 1000IU
anemia, lymphoid depletion in the thymus, atrophy of penicillin per milliliter and 1000pg streptomycin per milliliter.
hematopoietic tissues, subcutaneous and intramuscular Specimens are clarified by centrifugation at 2500 x g for 30 min
hemorrhages, and increased mortality (11). before inoculation. Heat (70 C for 5 min) and chloroform treatment
of inocula, using standard methods, may be performed before
CLINICAL DISEASE inoculation to inactivate other viruses that may be present.
For detection of CAV antigens by immunofluorenscence, acetone-
Serological surveys have shown that most commercial chicken fixed impression smears of thymus and bone marrow, and cryostat
flocks (13) and many specific-pathogen-free (SPF) flocks (14) sections of thymus are prepared using standard methods (10).
possess antibody to CAV. Outbreaks of clinical disease are rare. For immunocytochemistry, tissues should be fixed in 10% neutral
Onset of disease is acute and is usually seen from 2 to 4 wk of age. buffered formalin for no longer than 6 hr; after this time destruction
Disease occurs following infection of in-lay breeding flocks that of CAV antigens occurs (1,24). Fixation for up to 7 days in neutral
have no antibody to CAV. No clinical signs are seen in the buffered formalin has no apparent effect on detection of CAV DNA
breeders, but CAV is vertically transmitted to the progeny. in thymus by in situ hybridization (1).
Affected chicks are produced from eggs laid over a period of 3-6
wk following infection. Diseased birds are depressed and weak, PREFERRED CULTURE MEDIA AND SUBSTRATES
and they may be stunted. Mortality rate is variable, usually about
10%, but it may be as high at 60%. At necropsy, carcasses may be Chicken anemia virus can be isolated either in cultured cells
pale with thin, watery blood and yellowish bone marrow. including MDCC-MSB1 and MDCC-CU147 (5) or by inoculation
Thymuses, and, to a lesser extent, bursas, are atrophic. Livers may of susceptible chicks. Chick inoculation is more straightforward but
be enlarged and yellowish. Skin lesions, in the form of ecchymotic requires the availability of chicks derived from SPF flocks that are
hemorrhages, commonly occur on the wings. These lesions are free of CAV infection. Many SPF chicken flocks have antibody to
prone to secondary bacterial infection leading to gangrenous CAV. Chicks used for isolation of CAV must be free of such
dermatitis. Histologically, lymphocytes are depleted in the thymus, antibodies, as the presence of maternal antibody in chicks confers
particularly in the thymic cortex. The bone marrow is aplastic, with resistance to experimentally induced disease caused by CAV.
hematopoietic tissue replaced by adipose tissue. The disease is Although MDCC-CU147 cells appear to be susceptible to a wider
usually seen in broilers, but it has been seen in replacement laying range of CAV isolates and produce higher virus yields, MDCC-
pullets. A similar condition in older chicks, sometimes MSB1 cells are the most commonly used cells for isolating and
accompanied by inclusion-body hepatitis, may be due to mixed growing CAV. MDCC-MSB1 cells are grown in suspension in
infections of CAV and adenovirus. Vertically infected chicks RPMI 1640 medium (GIBCO-BRL) with 5%-10% fetal bovine
excrete virus, resulting in horizontal spread to their hatchmates. serum at 39 C in a 5% CO2 atmosphere. Following seeding at 2 x
Horizontally acquired CAV infection has been associated with 105 to 5 x 105 cells per milliliter, cell cultures must be subcultured
impaired economic performance in broilers (16). The severity of every 2-3 days. Following infection of MDCC-MSB1 cells with
the disease in horizontally infected chicks is variable, depending on CAV, a cytopathic effect develops that is characterized by an
factors that include age, virus dose and concurrent infections (12). increase in cell size, followed by lysis. CAV-infected MDCC-
To our knowledge, there are no peer-reviewed reports documenting MSB1 cell cultures are not capable of successful subculture. Cell
the presence of CAV in avian species other than domestic fowl. death is also indicated by alkalinity of the medium.
Two tubes or 2 wells in 24 well cell culture plates (Costar,
SAMPLE COLLECTION Coming), each containing 1 ml MDCC-MSB1 cells seeded at 3 x
105 cells per milliliter, are each inoculated with 0.1 ml tissue
The easiest way to detect infected flocks is to examine sera for suspension. Cultures are incubated at 39 C in a 5% CO2
antibodies to CAV. Dead or sick chicks can be submitted for atmosphere for 48 hr. If the inoculum appears to be toxic,
attempted virus isolation, detection of viral antigen, or detection of subculture is carried out for the first two subcultures by transferring
viral nucleic acid. Tissue samples for virus isolation can be sent to 0.3 ml medium and resuspended cells from inoculated cultures to
a laboratory in any virus transport medium. Although CAV is an 1 ml fresh cultures containing 3 χ 105 cells. Where toxicity is not a
extremely heat-resistant virus, samples should be kept cool during problem, subcultures are carried out by transferring 0.1-0.2 ml
transport to discourage the growth of bacteria whose presence may medium and cells from inoculated cultures to 1 ml warm growth
complicate isolation of the virus. Most of the isolates of CAV from medium every 48-72 hr. Isolation of CAV in MDCC-MSB1 cells
may require up to 10 subcultures or passages before a cytopathic
effect is evident.
124
Chapter 28 Chicken Anemia Vine

To isolate CAV in chicks, 1-day-old SPF chicks devoid of 0.1 ml of the virus-serum mixtures are inoculated into each of four
maternal antibody to CAV are inoculated intramuscularly with tubes, each containing 0.9 ml MDCC-MSB1 cell suspension
0.1 ml tissue suspension. After 14 days, heparinized blood samples containing 3 * 105 cells per ml. Appropriate virus, serum, and
are taken, and the hematocrit levels are determined. Levels below positive and negative controls also should be included. MDCC-
27% indicate anemia and suggest the presence of CAV. Following MSB1 cells are then subcultured at 2-3 day intervals into fresh
euthanasia, inoculated birds are examined grossly and histologically medium as described above. If the isolate is CAV, it will be
for CAV lesions. Because low doses of CAV do not produce neutralized by the CAV antiserum and no cytopathic effect will be
anemia experimentally, some birds should be retained and bled evident.
again from 21 to 28 days postinoculation and examined for the Antisera are prepared by intramuscular inoculation of 4-wk-old
presence of antibodies to CAV. More than one passage in SPF SPF chicks with 1 ml of cell-free CAV. A booster intravenous
chicks may be necessary to produce anemia. Once experimental inoculation (1 ml) is given 4 wk later, and the birds are completely
anemia and associated pathology have been reproduced, isolation of bled out after a further 3 wk (13). Hyperimmune rabbit antisera
the virus from livers of inoculated chicks in MDCC-MSB1 cells is (10) or mouse monoclonal antibodies (15) can also be used.
relatively straightforward. Alternatively, CAV antigens and/or More recently developed diagnostic methods are quicker, less
nucleic acids can be demonstrated in the tissues of submitted and/or laborious, and/or cheaper than virus isolation. CAV antigens in
inoculated chicks. diseased birds can be easily and rapidly detected by
immunofluorescent staining of impression smears of thymus and
AGENT IDENTIFICATION bone marrow, and of cryostat sections of thymus.
Immunofluorescence is concentrated in the thymic cortex. Antigens
Chicken anemia virus is a non-enveloped, icosahedral (T = 1) can also be detected by immunocytochemical staining of formalin-
virus, approximately 25 nm in diameter, comprising 60 structural fixed, paraffin-embedded tissues; however, for optimal results
units arranged as 12 pentamers (6). The virus possesses a single­ treatment of the sections with protease XIV (Sigma Chemical
stranded, circular DNA genome, approximately 2.3 kb in size. Company, St. Louis, Mo.) and fixation with formalin for no more
CAV has a density of 1.33-1.34 g/ml in CsCl (12). CAV is than 6 hr are necessary (1,24). Immunocytochemical staining is
classified as the type species of the genus Gyrovirus of the family more technically demanding, slower, and more laborious than
Circoviridae (28). immunofluorescence, and nonspecific staining can occur, but
Chicken anemia virus is a remarkably stable virus. It resists preservation of tissue morphology is much superior.
treatment with lipid solvents such as chloroform and ether. It Chicken anemia virus nucleic acids in tissues from diseased birds
survives heating at 70 C for 1 hr and is only partially inactivated by and infected MDCC-MSB1 cells dan be detected by a number of
a range of commercial disinfectants, including invert soap, techniques, including in situ hybridization (1), dot-blot
amphoteric soap, orthodichlorobenzene, iodine disinfectant, and hybridization (19,30) and polymerase chain reaction (PCR)
sodium hypochlorite, and by formaldehyde fumigation (35). amplification. Dot-blot hybridization methods, which can be
Available evidence suggests that only one serotype of CAV exists. completed in 2-4 days, have a limit of sensitivity of only 105-106
The existence of a second CAV serotype was suggested when an genome copies (1—10 pg CAV DNA) and are likely to be less
antigenically distinct agent was shown to cause similar pathogenic sensitive than virus isolation. Greater speed and sensitivity can be
effects following experimental infection of SPF chickens (26). achieved using PCR amplification methods, many variations of
However, the causal agent needs to be isolated and molecularly which have been developed. PCR methods that involve the visual
characterized to confirm whether it represents a second CAV detection of PCR product following agarose gel electrophoresis,
serotype. Minor antigenic differences between CAV isolates have ethidium bromide staining and exposure to UV light generally have
been revealed by monoclonal antibody binding studies (15). sensitivity limits of 10!-103 genome copies (0.1 - 10 fg) (D. Todd,
Naturally occurring isolates of CAV obtained from chickens appear unpubl. obs.). Hybridization techniques to detect PCR product have
to possess similar pathogenicity for young chicks. Naturally been employed to enhance sensitivity and to demonstrate specificity
occurring apathogenic isolates have been obtained from turkeys (19,20,25,27,33) but their inclusion makes these PCR methods more
(23). CAV can be attenuated by passage in MDCC-MSB1 cells labor-intensive and time-consuming. Nested PCR tests offer the
(29) or chicken embryos (22). CAV does not agglutinate the greatest sensitivity and have been used to detect CAV DNA in
commonly available avian and mammalian erythrocytes (D. Todd, embryonal tissues and egg shell membranes (18) and to demonstrate
unpubl. obs.). the presence of CAV DNA in blood cells and reproductive tissues
The earliest method to identify CAV was isolation of the virus in from chickens with neutralizing antibodies (2,7). The most
SPF chicks or MDCC-MSB1 cells, as described above. sensitive PCR methods are likely to have greater sensitivity than
Confirmation of the identity of putative isolates is achieved by virus isolation. While a highly sensitive PCR test may be the
immunofluorescent staining of infected MDCC-MSB1 cells. method of choice in research investigations and when examining
Infected MDCC-MSB1 cells are pelleted by centrifugation at biologicals for evidence of contamination with CAV, caution must
1500 x g for 10 min, resuspended in a minimal volume of be exercised in unreservedly recommending such tests for routine
phosphate-buffered saline, and smeared onto multispot Teflon- diagnosis of CAV. Given that low levels of CAV DNA can persist
coated slides. These are air-dried, fixed in acetone for 10 min at in chickens for months after infection, detection of virus DNA by a
room temperature, and stained by direct or indirect highly sensitive PCR technique may be of questionable value to the
immunofluorescence using chicken antisera to CAV or monoclonal diagnostician faced with clinical disease problems. The ability to
antibodies that are known to react with all CAV isolates (15). CAV quantify CAV DNA, made possible by applying competitive (34) or
antigens are found in the nuclei of infected cells. Fine granular real time (9) PCR methods, may assist in determining the clinico-
fluorescence usually is present throughout the nuclei, with some pathological significance of CAV in the clinical problem, but these
nuclei containing larger spherical inclusions. methods will not be available in many diagnostic laboratories.
Isolates growing in MDCC-MSB1 cells also can be confirmed as
CAV using a neutralization test. Chicken antiserum to CAV and SEROLOGIC DETECTION IN THE HOST
control chicken serum without antibody to CAV are diluted 1:80 in
RPM1 1640 containing antibiotics, inactivated by treatment at 56 C Antibodies to CAV were originally detected by indirect
for 30 min, cooled, and then added to an equal volume of the immunofluorescence and virus neutralization (4,12). However,
undiluted isolate and incubated for 30 min at 37 C. Samples of both of these tests have now been superseded by enzyme-linked
125
M Stewart McNulty and Daniel Todd

immunosorbent assay (ELISA). A number of ELISAs have been 10. McNeilly, F., G. M Allan, D. A. Moffett, and M S. McNulty.
described (3,8,21,31,32). Most use CAV grown in MDCC-MSB1 Detection of chicken anaemia agent in chickens by immunofluorescence and
cells as antigen. With the indirect ELISA format, antigen-coated immunoperoxidase staining. Avian Pathol. 20:125-132. 1991.
11. McNulty, M S. Chicken anemia agent. In: A laboratory manual for
plates are reacted with dilutions of chicken sera and the presence of
the isolation and identification of avian pathogens, 3rd ed. H. G. Purchase,
chicken antibodies specific to CAV are detected using an anti­ L. H. Arp, C. H. Domermuth, and J. E. Pearson, eds. Kendall/Hunt
chicken immunoglobulin enzyme conjugate. In the competitive or Publishing Company, Dubuque, Iowa. pp. 108-109. 1989.
blocking format, the presence of virus-specific chicken antibodies 12. McNulty, M S. Chicken anaemia agent: a review. Avian Pathol.
in test sera is detected when the reaction between the coated antigen 20:187-203. 1991.
and an enzyme-conjugated virus-specific mouse monoclonal 13. McNulty, M S., T. J. Connor, F. McNeilly, K. S. Kirkpatrick, and J. B.
antibody is prevented or reduced. Competitive or blocking tests McFerran. A serological survey of domestic poultry in the United Kingdom
generally exhibit fewer problems in relation to the non-specific for antibody to chicken anaemia agent. Avian Pathol. 17:315-324. 1988.
14. McNulty, M S., T. J. Connor, and F. McNeilly. A survey of specific­
binding of chicken antibodies to cellular antigens.
pathogen-free chicken flocks for antibodies to chicken anaemia agent, avian
ELISAs are now available commercially (Flockscreen Chicken nephritis virus, and group A rotavirus. Avian Pathol. 18:215-220. 1989.
Infectious Anaemia Antibody ELISA kit, Guildhay Ltd. Guildford, 15. McNulty, M S., D. P. Mackie, D. A. Pollock, J. McNair, D. Todd,
Surrey, United Kingdom; Chicken Anemia Virus (CAV) ELISA K. Mawhinney, T. J. Connor, and F. McNeilly. Production and preliminary
test kit, IDEXX Laboratories, Inc., Westbrook, Maine). Lack of characterization of monoclonal antibodies to chicken anemia agent. Avian
specificity with some ELISAs is not a major problem in serologic Dis. 34:352-358. 1990.
profiling of commercial flocks, but can cause problems when 16. McNulty, M S., S. G. McIlroy, D. W. Bruce, and D. Todd. Economic
testing SPF flocks (17). effects of subclinical chicken anemia agent infection in broiler chickens.
Avian Dis. 35:263-268. 1991.
Following an outbreak of anemia dermatitis syndrome,
17. Michalski, W. P., D. O’Rourke, and T. J. Bagust. Chicken anaemia
retrospective testing of sera from the breeder flocks, if sera are virus antibody ELISA: problems with non-specific reactions. Avian Pathol.
available, will identify one or more breeder flocks that were 25:245-254. 1996.
seronegative at point-of-lay, but that have developed antibodies by 18. Miller, Μ Μ, K. A. Ealey, W. B. Oswald,, and K. A Schat. Detection
the time disease is recognized in their progeny. These breeder of chcken anemia virus DNA in embryonal tissues and eggshell membranes.
flocks are probably the source of vertically transmitted virus. Avian Dis. 47: 662-671. 2003.
19. Notebom, Μ Η. M, C. A. J. Verscheuren, D. J. van Roozelaar, S.
DIFFERENTIATION FROM CLOSELY RELATED AGENTS Veldkemp, A. J. van der Eb, and G. F. de Boer. Detection of chicken
anaemia virus by DNA hybridization and polymerase chain reaction. Avian
Pathol. 21:107-118. 1992.
The differential diagnosis of anemia dermatitis syndrome includes 20. Novak, R., and W. L. Ragland. Competitive DNA hybridization in
those conditions causing anemia, immunosuppression, and microtitre plates for chicken anaemia virus. Mol. Cell. Probe. 15:1-11.
increased mortality. 2001.
21. Pallister, J., K. J. Fahey, and M Sheppard. Cloning and sequencing of
REFERENCES the chicken anaemia virus (CAV) ORF-3 gene, and the development of an
ELISA for the detection of serum antibody to CAV. Vet. Microbiol.
1. Allan, G. M, J. A. Smyth, D. Todd, and M S. McNulty. In situ 39:167-178. 1994.
hybridization for the detection of chicken anemia virus in formalin-fixed, 22. Schrier, C. C. Chicken anaemia agent vaccine. European Patent
paraffin-embedded sections. Avian Dis. 37:177-182. 1993. Application 92202864.2. 1992.
2. Brentano, L., S. Lazzarin, S. S. Bassi, T. A. P. Klein, and K. A Schat. 23. Schrier, C. C. and H. J. M Jagt. Chicken anaemia viruses of low
Detection of chicken anemia virus in the gonads and in the progeny of pathogenicity. US Patent Application 2001/0023664 Al
broiler breeder hens with high neutralizing antibody titres. Vet. Micro. 105: 24. Smyth, J. A, D. A. Moffett, M S. McNulty, D. Todd, and D. P.
65-72. 2005. Mackie. A sequential histopathologic and immunocytochemical study of
3. Brewer, J., J. M Saunders, and N. J. Chettle. The development of an chicken anemia virus infection at one day of age. Avian Dis. 37:324-328.
enzyme linked immunosorbent assay to detect antibodies to chicken anaemia 1993.
virus, and its comparison with the indirect immimofluorescent antibody test. 25. Soine, C., S. K. Watson, E. Rybicki, B. Lucio, R. M Nordgren, C. R.
In: Proceedings of the International Symposium on Infectious Bursal Parrish, and K. A. Schat. Determination of the detection limit of the
Disease and Chicken Infectious Anaemia, Rauischholzhausen, Germany, polymerase chain reaction for chicken infectious anemia virus. Avian Dis.
pp. 408-412. 1994. 37:467^176. 1993.
4. Bulow, V. V., B. Fuchs, and M Bertram. Untersuchungen uber den 26. Spackman, E., S. S. Cloud, C. R. Pope, and J.K. Rosenberger.
Erreger der infectiiosen Anamie bei Huhnerkuken (CAA) in vitro: Comparison of a putative second serotyoe of chicken infectious anemia virus
Vermehrung, Titration, Serumneutralisationtest and indirekter with a prototypical isolate. I. Pathogenesis. Avian Dis. 46: 945-955. 2002.
Immunofluoresenztest. Zentralbl. Veterinaermed. Reihe B 32:679-693. 27. Tham, K. M, and W. L. Stanislawek. Detection of chicken anaemia
1985. agent DNA sequences by the polymerase chain reaction. Arch. Virol.
5. Calnek, B. W., B. Lucio-Martinez, C. Cardona, R. W. Harris, K. A Schat, 127:245-255. 1992.
and C. Buscaglia. Comparative susceptibility of Marek’s disease cell lines to 28. Todd, D., M Bendinelli, P. Biagini, S. Hino, A. Mankertz, S. Mishiro,
chicken infectious anemia virus. Avian Dis. 44:114-124. 2000. C. Niel, H. Okamoto, S. Raidal, B. W. Ritchie, and G. C. Teo.
6. Crowther, R. A, J. A. Berriman, W. L. Curran, G. M Alan, and D. Circoviridae. In: Virus Taxonomy, VUIth Report of the International
Todd. Comparison of the structures of three circoviruses: Chicken anemia Committee for the Taxonomy of Viruses. C.M Fauquet, MA. Mayo, J.
virus, Porcine circovirus type 2 and Beak and feather disease virus. J. Virol. Maniloff, U. Desselberger, and L.A. Ball, eds. , Elsevier/Academic Press,
77: 13036-13041. 2003. London, pp 327-334. 2004.
7. Imai, K., M Mase, K. Tsukamoto, H. Hihara, and N. Yuasa. Persistent 29. Todd, D., T. J. Connor, V. M Calvert, J. L. Creelan, B. M Meehan, and
infection with chicken anaemia virus and some effects of highly virulent M S. McNulty. Molecular cloning of an attenuated chicken anaemia virus
infectious bursal disease virus infection on its persistency. Res Vet. Sci. 67: isolate following repeated cell culture passage. Avian Pathol. 24:171-187.
233-238. 1999. 1995.
8. Lamichlane, C. M, D. B. Snyder, T. Girschick, M A. Goodwin, and S. 30. Todd, D., J. L. Creelan, and M S. McNulty. Dot blot hybridization
L. Miller. Development and comparison of serologic methods for assay for chicken anemia agent using a cloned DNA probe. J. Clin.
diagnosing chicken anemia virus infection. Avian Dis. 36:725-729. 1992. Microbiol. 29:933-939. 1991.
9. Markowski-Grimsrud, C. J., Μ M Miller, and K. A. Schat. Development 31. Todd, D., D. P. Mackie, K. A. Mawhinney, T. J. Connor, F. McNeilly,
of a strain-specific real-time PCR and RT-PCR assays for quantitation of and M S. McNulty. Development of an enzyme-linked immunosorbent
chicken anemia virus. J. Virol. Meth. 101: 135-147. 2002. assay to detect serum antibody to chicken anemia agent. Avian Dis.
34:359-363. 1990.

126
Chapter 28 Chicken Anemia Virus

32. Todd, D., K. A. Mawhinney, D. A. Graham, and A. N. J. Scott. 34. Yamaguchi, S., N. Kaji, Η. M Munang’andu, C. Kojima, M Mase, and
Development of a blocking enzyme-linked immunosorbent assay for the K. Tsukamoto. Quantification of chicken anaemia virus by competitive
serological diagnosis of chicken anaemia virus. J. Virol. Meth. 82: 177-184. polymerase chain reaction. Avian Path. 29: 305-310. 2000.
1999. 35. Yuasa, N. Effect of chemicals on the infectivity of chicken anaemia
33. Todd, D., K. A. Mawhinney, and M S. McNulty. Detection and virus. Avian Pathol. 21:315-319. 1992.
differentiation of chicken anemia virus isolates by using the polymerase
chain reaction. J. Clin. Microbiol. 30:1661-1666. 1992.

127
29
AVIAN INFLUENZA
David E. Swayne, Dennis A. Senne and David L. Suarez

SUMMARY. Avian influenza (Al) is caused by type A influenza virus, in the family Orthomyxoviridae. Type A influenza viruses are
serologically categorized into 16 hemagglutinin (H1-H16) and 9 neuraminidase (N1-N9) subtypes. All subtypes have been identified in
birds. Infections by influenza virus have been reported in a variety of domestic and wild birds, but most infections are subclinical. In poultry,
influenza virus infections can be clinical, but the features are variable depending on virus strain, host species and breed, host age, concurrent
infections, physiologic stresses, and environmental factors. Some H5 and H7 strains are highly pathogenic for poultry, producing a systemic
disease with high mortality, and all H5 and H7 strains should be considered to have the potential to mutate from low (LP) to high
pathogenicity (HP). Both LP and HP H5 and H7 strains isolated from poultry are notifiable to the World Organization for Animal Health
(OIE).
Agent Detection or Identification. Although several diagnostic tests can be used for detection of avian influenza, the initial diagnosis is
preferably made by virus isolation via inoculation of embryonating chicken eggs, demonstration of hemagglutinating activity, and
verification as avian influenza virus by agar-gel immunodiffusion (AGID) or other antigen-detection tests. Once a virus has been isolated,
additional biologic and molecular studies can be performed to characterize the virus that isn’t possible by other means. This includes virus
subtyping, testing of pathogenicity in chickens or other species, and for H5 and H7 subtypes the determination of amino acid sequence at the
hemagglutinin proteolytic cleavage site to develop the appropriate control strategies. We do have an increased ability to rapidly diagnose and
characterize Al by detection of nucleic acids specific for Al viruses and potentially by rapid and sensitive immunoassays, but these tests will
not replace virus isolation in our ability to characterize Al viruses.
Serologic Detection in the Host. Serologic detection of infection in the host is important for moderate-to-large scale surveillance by
industry or government and for early diagnostic detection programs. Detection of type A group-specific antibodies via AGID or enzyme-
linked immunosorbent assay is preferred as the initial step for detection of primary infection, but confirmation and subtype determination via
hemagglutination-inhibition and neuraminidase-inhibition tests are crucial adjuncts.

INTRODUCTION of the head, comb, and wattles; and hemorthages on the serosal and
mucosal surfaces of viscera including the proventriculus, on the
Avian influenza virus (AIV) infections occur in a variety of shanks of the legs, and on the comb and wattles. Ruptured mature
domestic and wild bird species. The endemic nature of AIV in wild ovarian follicles (yolks) and accompanying inflammation
birds including ducks, gulls and shorebirds provides for a constant ("peritonitis”) are frequently observed in active layers with either
but low risk of introduction into poultry populations. In North LP or HPAl viral infections. Birds that die peracutely may exhibit
America, infections in commercial poultry occur primarily in few, if any, obvious clinical signs or gross lesions. The period of
domestic turkeys, with avian and swine influenza viruses. However, time during which virus may be recovered after infection depends
outbreaks can be seen in domestic and imported exotic birds, ratites, greatly upon the strain of virus, the type of sample, and the host
and occasionally in chickens, quail, and upland game birds. species and varies from 1-28 days in domestic poultiy (29) and
Infections in the latter three groups have been detected most often probably longer for wild species. However, AIV is infrequently
in live bird marketing systems traditionally found in large recovered from experimentally infected poultry after 14 days.
metropolitan settings. The greatest incidence of the disease in Detailed reviews on avian influenza are available (2,23,28).
domestic poultry occurs where there is opportunity for co-mingling
or indirect contact with waterfowl, seabirds, and/or pigs. SAMPLE COLLECTION
Historically, the best-known avian influenza virus (AIV) is fowl
plague virus, an H7 subtype, which has caused poultry losses since For Virus Isolation
the late 1800s in many parts of the world. The term fowl plague has Samples from respiratory (trachea, lung, air sac, and sinus
been replaced by highly pathogenic or high pathogenicity avian exudate) and digestive systems are suitable for virus isolation. They
influenza (HPAl) and is caused by some H5 and H7 strains (5). can be swabs or tissue. Cloacal swabs are typically best for AIV
Epizootics of HPAl since 1995 have included: 1) H7N3 in Pakistan isolation from wild birds or domestic ducks. However, both tracheal
during 1995, 2001 and 2004; 2) H5N1 in Asia during 1996-2005; or oropharyngeal, and cloacal swabs should be collected from
3) H7N4 in Australia in 1997; 4) H5N2 in Italy during 1997; 5) domestic poultry. Some HPAl viruses can be isolated from liver,
H7N1 in Italy during 1999-2000; 6) H7N3 in Chile during 2002; 7) spleen, blood, heart, or brain of clinically ill birds.
H7N7 in The Netherlands during 2003; 8) H7N3 in Canada during Dry sterile swabs preferably of a synthetic material, such as
2004; 9) H5N2 in USA during 2004; 10) H5N2 in South Africa Dacron and not calcium alginate, should be used to obtain samples
during 2004, and 11) H7N7 in North Korea (9,10,12,14,16,28). of tracheal, oropharyngeal or sinus exudate and of cloacal contents.
Where there is suspicion or confirmation of Al, state and/or federal Swabs with wooden shafts should be avoided unless the swabs are
authorities should be notified. immediately removed from the specimen tube and discarded after
expelling the swab contents into the transport medium (the wooden
CLINICAL DISEASE shaft may be treated with formalin that can interfere with
virus/RNA detection). The swabs are placed in 2-3.5 ml of sterile
The outcome of infection produced by AIV isolates varies from no transport medium prepared from brain-heart infusion, tryptose,
obvious clinical signs to 100% mortality. Birds of all ages and most, nutrient, or peptone broth, or cell-culture medium containing 1%
if not all, avian species are susceptible to infection. Common signs bovine serum albumin. A soft copper or nichrome loop may be used
or lesions in the major poultry species (chickens and turkeys) for small birds (19). Specimens should be kept at 4 C after
infected with LPAI viruses include slight-to-severe declines in egg collection and during transport. Low levels of antibiotics, e.g. 200
production; increased mortality; diarrhea; respiratory difficulties; pg/ml gentamicin sulfate and 5 pg/ml amphotericin B, can be added
and occasionally, deposits of urates in the kidneys. Infections with to the transport medium to reduce growth of bacterial and fungal
HPAl viruses results in severe disease which can present as edema contaminants during transportation but antibiotics should used as an
128
Chapter 29 Avian Influenza

adjunct to chilling and not as an alternative to chilling. When (20 gg/ml), gentamicin sulfate (1000 pg/ml), and kanamycin sulfate
isolation cannot be attempted immediately, samples can be stored at (650 pg/ml). Supernatant should be kept at 22-25 C for 30 to 60
4 C for up to 48 hr, or frozen (preferably at -70 C) for longer min just before inoculation, to allow the antibiotics to reduce the
periods. Samples can be shipped with frozen gel packs if they will level of bacterial contamination.
arrive at the laboratory within 48 hr. If further reduction in bacterial contamination is required to reduce
embryo deaths or non-viral hemagglutinating activity (HA) of egg
For Reverse Transcriptase Polymerase Chain Reaction (RT- fluids, the supernatant can be filtered through pre-wet 0.22-0.45-μιη
PCR) reusable or disposable membrane filters. However, filtration can
Specimens suitable for virus isolation are generally suitable for remove low levels of virus from samples and reduce isolation rates.
RT-PCR (see collection of samples for virus isolation above). For tissue specimens, a 10% ground suspension is made in
However, tracheal or oropharyngeal swabs are the specimens of antibiotic-containing brain-heart infusion, tryptose, nutrient, or
choice for gallinaceous birds because they are sensitive, easy to peptone broth, or cell-culture medium containing 1% bovine serum
process, and generally contain less extraneous organic material that albumin. Grinding is accomplished in a mechanical homogenizer,
can interfere with RNA recovery and amplification by PCR. stomacher, or mincing with sterile scissors and ground either in a
Caution should be used when testing cloacal swabs by real-time sterile glass tissue grinder or with a sterile mortar and pestle using
RT-PCR (RRT-PCR) because of the concerns with false negative sand or similar abrasive materials. Alternatively, small tissue
results due to poor RNA recovery or presence of PCR interfering fragments (1 cm3) are placed in 2-3.5 ml medium, frozen and
substances. The use of RT-PCR for screening of environmental thawed, vigorously vortexed, centrifuged, and the supernatant
samples needs to be done with caution because there may be a poor diluted in equal volumes of medium containing antibiotics. Tissue
correlation between a positive PCR result and presence of viable suspensions are clarified by low-speed centrifugation (1000-1500 x
virus; e.g. inactivated virus cannot be isolated but may yield a g) and placed at room temperature for 30 to 60 min before
positive RRT-PCR test result. Therefore, RT-PCR is not inoculating embryos.
recommended to assure an environment free of avian influenza Chicken embryos, 9-11 days old, are injected via the allantoic sac
virus, because of the potential for the test to identify inactivated (see Chapter 43 on virus propagation in embryonating eggs) with
virus, providing a false positive. Calcium alginate swabs should be 0.2-0.3 ml of clinical specimen. However, inoculation of embryos
avoided for collection of samples for RT-PCR because they are via the yolk sac route or the use of cell cultures (MDCK or VERO)
reported to produce inhibition for PCR. with added trypsin has been more sensitive for the isolation of
When using RRT-PCR, particular attention should be given to the swine-origin influenza viruses from poultry. Three to five embryos
selection of an appropriate method for RNA extraction to maximize per sample are usually adequate. Eggs are incubated at 35-37 C for
recovery. The RNA extraction step is considered one of the key 3-5 days. If virus is present, some embryos may die following
steps for a successful test. Several commercial kits are suitable for inoculation, but some may not, depending on the virulence of the
the recovery of RNA from clinical specimens. The isolation of good virus. Amnioallantoic fluid (AAF) from eggs with dead embryos as
quality RNA that is void of PCR inhibiting substances is essential well as from eggs with surviving embryos should be collected and
for accurate test results. Organic extraction methods such as Trizol analyzed for HA. A second passage may be required for some
(Invitrogen, Carlsbad, CA) are generally better than silica gel viruses. Additional passages can be made by diluting the AAF 1:10
membrane kits for most specimens and are especially good for in antibiotic medium and inoculating a new set of eggs. The eggs
cloacal swabs and tissues that have a high organic content. from additional passages are incubated and tested using the same
However, the toxic chemicals (phenol, guanidine isothiocyanate, procedure as for the first passage.
and chloroform), and additional labor and processing time required
for this method makes it less desirable than other methods. Silica RT-PCR
gel membrane kits such as RNeasy (Qiagen, Valencia, CA) have RT-PCR procedures have been developed to aid in the detection of
been successfully used for processing tracheal and oropharyngeal ATV in clinical samples and to facilitate assessment of
swabs, but this system has lower sensitivity for tissue suspensions pathogenicity of H5 and H7 ATV (18,33). Both traditional RT-PCR
and cloacal swabs. This is most likely because of the heavy organic assays with analysis of the PCR product on an agarose gel and real­
load in these specimens produces extraneous RNA or other time RT-PCR (RRT-PCR) assays have been described in the
substances that compete for binding sites on the silica membrane literature. Most of the methods have levels of sensitivity
resulting in lower recovery rates of target RNA. The MagMAX kit comparable to virus isolation, and are being commonly used as
(Ambion, Austin, TX) uses specially engineered magnetic beads to primary screening tests for avian influenza throughout the world. Of
absorb RNA and is more resistant to RNA saturation problems the two commonly used methods, each has its strengths and
associated with silica gel membrane kits. The MagMAX kit can be weaknesses. The traditional RT-PCR requires less expensive
performed in 96-well plates and therefore is more suitable than equipment that is available in many diagnostic laboratories, but
other extraction methods for high throughput testing of does require additional labor and cross contamination is an ongoing
oropharyngeal or tracheal swabs. However, the suitability of this concern. The RRT-PCR tests are less labor intensive, can be
method for extraction of tissue suspensions and cloacal swabs has performed more rapidly, and have a reduced probability of cross
not yet been fully evaluated. contamination in the laboratory, but it does require expensive
equipment to perform. Although many different RT-PCR tests for
PREFERRED CULTURE MEDIA AND SUBSTRATES avian influenza have been described, few have been extensively
validated for fitness for purpose with field samples in comparison to
Virus Isolation the performance standard of virus isolation.
The preferred method of diagnosis requires isolation of AIV from In general, both RT-PCR assays have four steps: 1) isolation of
clinical swabs or tissue in embryonating chicken eggs. For cloacal, the RNA; 2) conversion of the viral RNA to cDNA by the use of
tracheal, oropharyngeal, or sinus swabs received in appropriate reverse transcriptase; 3) amplification of the cDNA by PCR; and 4)
transport media, the fluid is centrifuged at low speed (1000-1500 x evaluation of the PCR product (amplicon) for the presence or
g) to sediment debris and the supernatant is added to antibiotic absence of the target DNA sequence. For traditional RT-PCR the
medium. The following are suggested antibiotics and the maximum specificity of the amplicon is confirmed by electrophoresis of the
dose per ml that can be used singularly or in combination: penicillin amplicon in an agarose gel containing ethidium bromide, then
(10,000 IU/ml), streptomycin (10,000 pg/ml), amphotericin B comparing the size of the amplicon to markers of known molecular
129
David E. Swayne, Dennis A. Senne and David L. Suarez

weight. The detection of the PCR product of the proper size likely to come by improvements in the critical control points of
however provides only a presumption of the specific product, and RNA extraction, RT-PCR amplification, and primer and probe
additional confirmation can be provided by several methods design. Opportunities to increase sensitivity, specificity, throughput
including enzyme restriction enzyme endonuclease assays, Southern and ease of use are likely to occur, and will become adopted as part
Blot hybridization, or by direct sequencing. The need for post­ of the official protocol. Additionally, consideration for ongoing
amplification processing of the amplicon is one of the major analysis of test performance needs to be considered for all
drawbacks of using traditional RT-PCR as a diagnostic assay molecular diagnostic tests, since specificity is directly related to
because of the increased labor and the high potential for primer and probe design, and genetic variation or drift may affect
contamination of the laboratory and cross contamination of test performance. Future research goals will include the use of an
samples. Extreme precautions, such as the use of dedicated internal positive control (unrelated to the target cDNA) is also
equipment and separation of sample processing activities from post highly recommended to monitor for presence of PCR inhibiting
amplification processing activities, must be taken to prevent cross substances that could cause a false negative test results. Several
contamination. The RRT-PCR assays differ primarily in the different strategies for an internal control have been described.
analysis of the amplicon, which is done with fluorescently labeled
probes or dyes and special thermocyclers that accurately measure AGENT IDENTIFICATION
light levels at different wavelengths. Several different strategies are
available for the fluorescently labeled probes, including Taqman Physicochemical, Morphologic and Biological Properties
probes, Scorpion probes, Fret probes and others. The Taqman The influenza viruses are a diverse group of viruses that belong to
probes, also known as hydrolysis probes, are the most commonly the family Orthomyxoviridae, are enveloped (ether-sensitive), and
used, and includes both an excitation dye and a quencher dye. contain single-stranded ribonucleic acid (RNA) that is segmented
Normally, the light produced from the excitation dye is dampened and has negative polarity. Two major internal components, the
by the close proximity of the quencher dye. However, when the matrix protein (M) and ribonucleoprotein (RNP), are group-specific
Taqman probe binds to the PCR product, the excitation dye is proteins that designate type specificity (i.e., A, B, or C). There are
cleaved away from the probe and quencher dye by Taq polymerase, two surface glycoproteins, the hemagglutinin (H) and
and the light produced from the excitation dye is no longer neuraminidase (N) glycoproteins, that project from the lipid
dampened by the quencher dye. This light produced is quantitative membrane and participate in the infection process and define
and is proportional to the amount of PCR product present in the subtype specificity (i.e., H1-H16 and N1-N9). Influenza virions
reaction. The light level is taken after every PCR cycle, and the can be either spherical forms or short rods (80-120 nm) or
results can be visualized in real-time with no additional post-PCR filamentous forms (400-800 nm long).· Influenza viruses are
processing. Because the probe must specifically bind to the PCR relatively stable at pH 7-8 but are labile at the low pH range. Al
product, an additional level of assurance is provided that the PCR is viruses are sensitive to heat inactivation with inactivation at
specific for the intended agent. pasteurization temperatures (55.6-63.3 C) in 3 to 6.2 min or
Although many different RT-PCR tests and even alternative RNA minimal cooking temperature (70 C) in a few seconds (26,27).
amplification tests have been described, most of these have never
been validated for use with field samples. Validation is the Initial Identification
determination of fitness for a particular use, and includes testing of Hemagglutinating Activity (HA). The surface H glycoprotein of
a prescribed number of positive and negative field samples against a ATV isolates will bind to receptors on a variety of mammalian and
performance standard. In the U.S. a RRT-PCR assay described by avian erythrocytes, and this phenomenon is the basis for screening
Spackman et al. for detection of ATV RNA in clinical specimens of AAF for presence of hemagglutinating agents. Small amounts of
from poultry was developed and validated with clinical specimens AAF can be harvested aseptically from eggs 24—48 hr after
(n = 1,550) from live-bird market surveillance (22). The AIV assay inoculation with a syringe and needle without killing the embryo
was further validated (n = 3,500) during an outbreak of LPAIH7N2 (see Chapter 44 on virus propagating in embryonating eggs). This
in Virginia in 2002. The assay has three sets of primers and probes early sampling can speed diagnosis because HA or specific ATV
that are used in separate reactions, one to identify any type A antigen can be detected long before the embryo dies. HA is
influenza viruses (Matrix assay), one to identify H5 strains and one determined by making twofold dilutions in a microtiter plate
to identify H7 strains of ATV. The matrix test is targeted to a highly followed by the addition of an equal volume of 0.5% washed
conserved region of the influenza genome, and it can detect any chicken erythrocytes. In some cases alternative RBCs need to be
type A influenza virus, including swine, equine, and human considered. In particular the Hl and H3 swine viruses that
influenza viruses. Because the hemagglutinin gene is known for its commonly infect turkeys no longer hemagglutinate chicken RBCs.
high variability, it has been difficult to develop a single test that The use of turkey RBCs should be considered as an alternative,
would identify all H5 or all H7 viruses. The test validated for use in although guinea pig and horse RBCs may be used (25,30). Diluting
the U.S. has been shown to identify most H5 viruses from around the AAF avoids the occasional prozone effect that can occur with
the world, but the H7 test is more restricted in its geographic reach. undiluted egg fluids.
The test used in the U.S. will identify North American origin H7 If HA is observed, the remaining AAF should be harvested after
viruses, but it will not detect Eurasian H7s. Alternative the egg has been chilled for 4—12 hr at 4 C. Chilling kills the
primer/probe sets are available for H7s from other geographic embiyo and lessens the chance of contaminating the fluids with
regions, and the diagnostician must be aware of the strains that are erythrocytes, which can absorb influenza virions.
likely to occur in their region. Since the matrix (M) assay has the Embryos that survive the incubation period and AAF that lacks
highest level of sensitivity, samples are first screened with the M HA can be discarded after a sample of fluid is removed for an
primers/probe then positive specimens are tested with the H5 and additional egg passage.
H7 assays. In the U.S. this assay is considered an official test for Fluids from AJV-infected eggs occasionally may fail to exhibit
avian influenza, and it being used by more than 39 state HA, particularly when virus levels are less than that needed to cause
laboratories, that have demonstrated proficiency in testing, as a hemagglutination, usually ΙΟ5—106 mean embryo infectious dose
front line surveillance tool for Al. (EID5o)/ml. If the fluids are HA-negative, an additional passage is
The use of rapid molecular diagnostic tools can still be considered optional to be certain that isolations are not missed because of low
to be in its early stages, and additional technical advances should be levels of virus in the sample.
expected. These incremental improvements in performance are
130
Chapter 29 Avian influenza

After demonstrating HA in AAF, the agent responsible for such monoclonal antibody technology to demonstrate type A influenza
HA must be determined. Common hemagglutinating agents for antigens in a solid-phase, flow-through ELISA. This test will detect
birds include AIV, avian paramyxoviruses including Newcastle ATV antigens in AAF or directly in clinical specimens (cloacal and
disease virus (NDV) (see Chapter 30), a hemagglutinating tracheal/orophaiyngeal swabs) from birds using a 15 min procedure
adenovirus (see Chapter 19), and hemagglutinating bacteria. For the (31). These test kits have potential use for rapid screening of poultry
initial determination, some diagnostic laboratories run microtiter flocks for avian influenza (Al), but vary in sensitivity, false positive
hemagglutination-inhibition (HI) tests to ascertain the presence or rate and the chickens must be in the acute stage of the disease, that
absence of NDV. If NDV antiserum inhibits the HA, NDV is is, showing clinical respiratory or systemic signs or be dead, to
present, but this inhibition does not exclude the possibility that the provide sufficient viral antigen (minimum of 1055 EIDso/ml of
fluid also contains one or more other viruses. Confirmation of AIV AAF) for detection. An experimental antigen-capture ELISA (AC-
in HA-positive samples is done by demonstrating the presence of ELISA) using microtiter plates will also detect type A influenza
type- and subtype-specific antigens (see below). viral RNP in cloacal and tracheal/oropharyngeal swabs (11).
However, in waterfowl surveys, presumably because of low virus
Type Classification titers, AC-ELISA had low sensitivity and specificity as compared to
Influenza viruses are grouped into three types (A, B, or C) based virus isolation in chicken embryos (3).
on highly conserved internal components of the virion, namely the Visual Demonstration of AIV Antigen. Demonstration of ATV
M and RNP proteins or genes. All avian, swine, and equine antigen in tissue by immunocytochemistry, immunohistochemistry,
influenza viruses share common type A M and RNP proteins/genes or immunofluorescent microscopy has value for rapid diagnosis of
that can be identified by any of four methods: 1) the agar-gel Al, especially HPAI, but should be utilized only as an adjunct to
immunodiffusion (AGID) test with processed AAF or virus isolation. DFA staining of acetone-fixed tissue impression
chorioallantoic membranes (CAMs) which is the most commonly smears can give a presumptive diagnosis of Al in less than 1.5 hr
used to identify AIV; 2) an antigen-capture solid-phase or flow- (4). Impression smears of liver and kidney are easiest to prepare and
through enzyme-linked immunosorbent assay (ELISA) with AAF or give the most consistent results, especially for diagnosis of HPAI.
suitable clinical specimens; 3) visualization by the direct fluorescent However, impression smears of tracheal mucosa and cloacal bursa
antibody (DFA) and indirect fluorescent antibody (IFA) or also provide satisfactory results and are necessary samples to
immunoperoxidase (IP) tests with tissues or CAM smears and diagnose LPAI. ATV can also be demonstrated in fresh-frozen
frozen sections; and 4) detection of the M gene of ATV in clinical sections by DFA, IFA, and IP tests (20) and in formalin-fixed tissue
specimens, AAF or vaccines by RT-PCR or assays. sections by immunohistochemistry (7,20). However, formalin
The AGID Test to Detect AIV Antigen. The procedure utilizes fixation reduced the ability %of immunofluorescent and
influenza A-positive and negative control sera, influenza A antigen, immunochemistry procedures to detect AIV. Detection of antigen in
and an unknown sample (antigen). Either AAF or CAM from kidney tissue sections by the IP test was consistent only when virus
embryonating eggs can be the unknown antigen, but CAM is titers were 105 ELD5()/g of tissue or higher. The IFA and IP tests are
preferred because of higher virus content. Briefly, CAMs should be considered to be more sensitive than the DFA test.
rinsed in phosphate-buffered saline (PBS) (pH 7.2), pooled, and Molecular Detection of Type-specific RNA. For those
carefully drained, opening pockets to remove excess fluid, before laboratories routinely testing for Al by RRT-PCR, this assay can
they are ground in an electric blender or hand-operated tissue also be used to detect presence of ATV in AAF, vaccines or clinical
grinder and used (undiluted) as antigen in the AGID test (6). specimens utilizing primers/probes specific for RNA that codes for
Alternatively, undiluted AAF can be used as the unknown antigen, the highly conserved M protein, a type-specific protein, or H5, H7
but the infectivity titer probably needs to be at least ΙΟ5-106 mean or other HA subtypes if available.
embryo lethal dose (ELD50) to give satisfactory results (15). If the
undiluted AAF is negative for influenza A antigen by the AGID Subtype Classification
test, it may be necessary to concentrate the AAF by acid Once an isolate or viral proteins have been identified as type A
precipitation of the antigen to eliminate the possibility of a false influenza by either the AGID test, solid-phase ELISA, AC-ELISA,
negative result because of a low virus titer. Briefly, eight to 10 ml or RT-PCR, the next step is H and N subtyping. There are currently
of AAF is acid precipitated (IN HC1, pH 4.0, 4 C) for 60 min in an 16 Η (H1-H16) and 9 N (N1-N9) subtypes recognized as
ice bath. The precipitate is centrifuged at 1000 x g for 10 min at 4 determined by the HI (15) and neuraminidase-inhibition (NI) (32)
C, the supernatant is discarded, and the pellet is resuspended in 80- tests, respectively. The HI and NI tests are performed in microtiter
100 μΐ glycine-Sarkosyl buffer. The resulting antigen is tested by plates with monospecific reference sera directed against whole virus
the AGID test. preparations or purified H and N antigens. Alternatively, molecular
The AGID test can be performed in Petri dishes with 0.9% agarose subtyping assays, utilizing, have been developed for North
(ME grade) in PBS (0.01 M, pH 7.2) with an additional 8% NaCl. A American lineages of H5 and H7 ATV (22) but are not yet available
seven-well template with a center well surrounded by six evenly for the other subtypes. All isolates identified as subtypes H5 and H7
spaced wells is used. The wells are 2.4 mm apart and 5.3 mm in are Notifiable Avian Influenza (NAT) and should be reported to
diameter. state and federal authorities. The H5 and H7 NAI can be LP or
The precipitin line will be present within 24 hr at room HPAI viruses. Subtype determination is beyond the scope of most
temperature and may increase in density for up to 48 hr. If a diagnostic laboratories not specializing in influenza viruses.
precipitin line develops between the suspect antigen and the Assistance is available from the National Veterinary Services
positive serum, and if that line is continuous with the line between Laboratories (NVSL), Ames, Iowa. The NVSL is a reference
the adjacent positive antigen and the antiserum, the agent can be laboratory of the World Organization for Animal Health (OIE).
identified as type A influenza virus. The HI Test to Subtype AIV Isolates. Four HA units of the newly
Antigen Capture to Detect AIV Antigen. Detection of ATV in isolated virus can be tested against sera of known H subtypes with a
samples can be done by demonstrating the presence of specific suspension of 0.5% chicken erythrocytes (see Chapter 47).
influenza viral antigens with colorimetric assays. Commercially However, because of antigenic differences between ATV subtypes
licensed test kits for human and veterinary medical use such as and slight differences even within subtypes, the AIV should be
Directigen Flu A® (Becton Dickinson Microbiology Systems, identified only by experienced personnel using monospecific
Sparks, MD), NOW Flu A® (Binax, Portland, ME) Flu Detect® antisera in reference laboratories.
(Synbiotics, San Diego, CA) and others are available that use
131
David E. Swayne, Dennis A. Senne and David L. Suarez

With HI tests for ATV, care must be taken in selecting reagents steric hindrance. Steric hindrance can occur if the antiserum used
used to identify the H subtype of the virus to avoid problems with for H subtyping also contains N antibodies that are homologous

Table 29.1. Amino acid sequences at the HAO cleavage site of H5 and H7 AIV strains (modified from Senne et al., 1996 (18).
Virus Strain Subtype Pathotype HAO Cleavage site sequence
A/Chicken/Hidalgo/26654-1368/94 H5N2 LPAI PQ------------ ....................RETR
*
GLF
A/Emu/NY/12716-67/94 H5N9 LPAI PQ------------ --------------- RETR*
GLF
A/Turkey/CA/6878/79 H5N3 LPAI PQ------------ ................... RETR
*
GLF
A/Chicken/Queretaro/14588-19/95 H5N2 HPAI PQ------------ -------- RKRKTR
*
GLF
A/Γurkey/England/91 H5N1 HPAI PQ------------ --------- RKRKTR
*
GLF
A/Γ urkey/Ireland/83 H5N8 HPAI PQ------------ --------- RKRKKR
*
GLF
A/Chicken/Hong Kong/220/97 H5N1 HPAI PQ------------ —RERRRKKR
GLF
*
A/Chicken/NY/13142-5/94 H7N2 LPAI p E------------ ------------ NPKTR
*
GLF
A/Turkey/Italy/977/99 H7N1 LPAI P £------------ ------------- 1 PKG R
G
* LF
A/Quail/AR/16309-7/94 H7N3 LPAI PE------------ ----------- -N PKT R
*
G LF
A/Chicken/Chile/176822/02 H7N3 LPAI PE G
------------ K PKT R
* LF
A/Chicken/Canada/AVF VI/04 H7N3 LPAI *
GLF
PE.................------------ NPKTR
A/Chicken/Victoria/85 H7N7 HPAI p e------------ ---- IPKKREKR
GLF
*
A/Chicken/The Netherlands/04 H7N7 HPAI PE------------ ------- 1 PKRRRR
*
GLF
A/Turkey/Italy/4580/99 H7N1 HPAI P E................. —IPKGSRVRR
G
* LF
A/Chicken/Chile/4325/02 H7N3 HPAI PEKPKTCSPLSRCRKT R
G
* LF
A/Chicken/Canada/AVFV2/04 H7N3 HPAI PE----- NPKQA YQKRMTR*GLF
* indicates cleavage site, dashes are used for alignment. Basic amino acids arginine (R) and lysine (K) are shown in bold.

with the unknown isolate. The specific reaction of the N antibodies site of H protein. The virulence marker for HPAI is characterized by
can interfere nonspecifically with the H, leading to nonspecific the presence of multiple basic amino acids at the H cleavage site
inhibition and possible misidentification of an isolate (17). (Table 29.1); however, caution should be exercised because some
The NI Test to Subtype AIV Isolates. The NI test is used to recent HPAI viruses have been shown not to have this virulence
identify the other important surface antigen of the influenza viruses marker (10,24).
(8,32). This test, being more complex than the HI test, is generally The method by which the presence of multiple basic amino acids
performed only by those engaged actively in influenza research or at the cleavage site can be determined has largely been limited to
influenza virus characterization. All nine of the known influenza N RT-PCR amplification of the coding sequences surrounding the
subtypes have been identified in avian species. cleavage site of the H protein, followed by sequencing of the
amplicon by automated sequencing methods (18,33). This approach
Pathogenicity Determination has been used to obtain sequences from a large number of H5 and
Chicken Pathogenicity Test. Assessment of pathogenicity of a H7 subtype viruses representing different temporal, geographical
newly isolated ATV is critical for appropriate control strategies and and host origins. However, as with any molecular based assay,
international trade. Precautions should be taken to assure adequate success in the amplification of the target sequence will depend on
containment of the virus before attempting bird inoculations. the sensitivity of the primers used.
Pathogenicity testing of new isolates should be conducted according Phylogenetic Analysis
to guidelines published by the OIE and the U.S. Animal Health As part of the routine characterization of AIV from new outbreaks,
Association (13). Procedures currently recommended include the viruses will be at least partially sequenced for the hemagglutinin
intravenous inoculation of eight 4 to 8-wk-old chickens, and, if the and other viral genes. The sequence information, as mentioned
isolate is of the H5 or H7 subtype, determination of the amino acid previously can help determine the pathogenic potential for H5 and
sequence at the cleavage site of the H. Isolates that are lethal for six, H7 viruses, and additionally can be used for molecular
seven or eight (>75%) of eight experimentally inoculated chickens, epidemiology purposes. The sequence data can be compared to the
or have an intravenous pathogenicity index (IVPI) >1.2 in 6-wk-old influenza sequence database either through a blast search of the
susceptible chickens are considered HPAI. In addition, H5 or H7 National Center of Biological Information or by phylogenetic
subtype viruses that do not meet the >75% mortality criteria but analysis using a variety of available programs. The phylogenetic
have an amino acid sequence at the H cleavage site that is analysis principally provides a graphical representation of the
compatible with HPAI viruses are also classified as HPAI (see relationships or closeness of viruses. The NCBI blast search is a
Molecular Assessment of Pathogenicity below). Isolates that do not quick and inexpensive way to determine the sequence similarity to
meet the criteria to be classified as HP should be designated simply all the sequence information available in GenBank. Further
as ATV or LPAIV with its corresponding H and N subtype information on the origins of a virus isolate may be gained by
designation. phylogenetic analysis. The determination of the origin of a virus is
Avian influenza virus isolates from any H subtype can be useful to understand how the outbreak may have occurred. For
pathogenic, but to date, only H5 and H7 subtypes have been HP. example, the Virginia LP H7N2 in 2002 affected 197 flocks and
However, the vast majority of H5 and H7 subtypes are LPAI. cost the government in excess of 60 million dollars. Phylogenetic
Molecular Assessment of Pathogenicity. An understanding of the analysis clearly showed that this virus was not a recent introduction
molecular basis for pathogenicity of AIV has made it possible to from wild birds, but was related to the viruses that were being
predict virulence of some strains of H5 and H7 subtype viruses isolated from the live bird marketing system in the Northeast USA
based on presence or absence of a virulence marker at the cleavage (21). This information helped provide the basis for the decision to
132
Chapter 29 Avian Influenza

eradicate the virus and helps us understand that the virus was well are species specific, although the available commercial kits will
adapted to poultry and could spread easily from flock to flock. The detect serologic response in both chickens and turkeys.
minimum sequence information necessary for a reasonable tree is at Development of a competitive ELISA would result in a single test
least 300 base pairs, but the more sequence information available usable for all avian species.
the more discriminatory the phylogenetic trees As sequencing has The HI Test for Antibody Detection in the Host. The HI test
become cheaper and more available, the expectation is more to allows avian influenza H subtypes to be differentiated on the basis
sequence all eight gene segments to provide the most of the antigenic character of the H and is an essential follow-up test
comprehensive view of the isolate possible. for AGID-positive sera samples. This subtype specificity makes the
HI test of limited value in initial screening of suspect birds or
SEROLOGIC DETECTION IN THE HOST flocks, unless secondary spread of a previously identified ATV is
being monitored. A large battery of control sera and test antigens
Routine surveillance in individual birds or flocks for serologic representing all 16 of the known H subtypes would be necessaiy for
evidence of ATV infections is important for early detection and detecting primary infection of an unknown H subtype. However,
regulatory surveillance. Four major tests are used: AGID, ELISA, that still would not rule out the possibility that a serum might
HI and NI. Historically, the virus-neutralization (VN) test has been contain antibodies to a new H subtype not yet described, or because
performed as is done with other viruses, such as NDV. Specificity is of antigenic differences caused by antigenic drift. HI testing for
similar to an HI test, but the VN test is infrequently used for subtyping purposes is best suited for references laboratories,
diagnostic purposes in poultry medicine. especially because sera may need to be treated with a receptor­
The AGID Test for Antibody Detection in the Host. The AGID destroying enzyme to eliminate nonspecific HI reactions (15).
is the preferred serologic surveillance test in the U.S.A, because a The NI Test for Antibody Detection in the Host. The NI test
single test detects serologic response in all bird species and against allows ATV to be differentiated on the basis of the antigenic
infection by all type A influenza viruses. Type A AGID test character of the N and is an essential follow-up test for AGID-
reagents (antigen and positive serum) are readily available from the positive samples. NI testing is best suited for reference laboratories.
NVSL and a commercial company (Charles River-SPAFAS,
Norwich, CT). Alternatively, CAMs can be processed as described DIFFERENTIATION FROM CLOSELY RELATED AGENTS
previously in the section on the AGID test to detect ATV antigen
and used as the test antigen (6). Briefly, the CAM is ground, frozen The influenza viruses produce a wide range of clinical signs and
and thawed three times, and centrifuged at a low speed (500-1500 x lesions, and diagnosis depends upon viral and serologic
g). The supernatant is removed and inactivated with formalin (0.1% identification, especially in the initial flocks. Isolates must be
final concentration). The antigen can be used immediately or after distinguished from all other hemagglutinating agents, including
incubating at 37 C for 36 hr to assure virus inactivation. The CAM some avian adenoviruses, Newcastle disease virus (NDV), and other
from three eggs should yield about 1 ml of antigen. The antigen can avian paramyxoviruses by use of the AGID test with known
be stored at 4 C for several weeks or frozen indefinitely at -10 C. positive monospecific influenza A antiserum, by the use of ATV
An antigen consisting principally of M protein can also be prepared specific antigen capture tests, or by RT-PCR assays specific for
from a virus suspension in AAF (15). Care should be taken to ATV or NDV. Diagnosis can be complicated by the presence of
balance the concentration of antigen to match the antiserum so that other microorganisms, such as mycoplasmas. Dual isolations of
the precipitin line forms midway between the antigen and antiserum influenza virus and Newcastle disease virus are not unusual.
well. Only antigen and antiserum that produces a single, narrow, Standard laboratory techniques using specific antisera to neutralize
bold line that extends completely into adjacent test wells other viruses or antibiotics to control bacterial growth are helpful in
(containing negative serum) should be used. Also, a set of known such situations.
strong and weak positive serums and negative serums should be
used to assure adequate sensitivity and specificity of the reagents. ACKNOWLEDGMENT
A protocol for preparation of antigen and antiserum is available
from the NVSL. The authors thank CW. Beard for contributions to prior editions.
The AGID influenza type A test for antibody detection in the host
REFERENCES
utilizes unknown test sera, influenza A test antigen, and influenza A
positive control antiserum. The precipitin line between the ATV test 1. Adair, B.M, D. Todd, E.R. McKillop, and MS. McNulty. Detection of
antigen and positive antiserum will be present within 24 hr at room influenza A type-specific antibodies in chicken and turkey sera by enzyme-
temperature and will increase in density for up to 48 hr; however, linked immunosorbent assay. Avian Pathol. 18:455-463. 1989.
interpretation of most test results can be made at 24 hr. 2. Alexander, D.J. A review of avian influenza in different bird species.
Sera from infected chickens or turkeys will yield positive AGID Vet.Microbiol. 74:3-13. 2000.
results within 5-6 days and up to 3 mo after the appearance of 3. Alfonso, C.P., B.S. Cowen, and H. Vancampen. Influenza A viruses
clinical signs. Because not all sera will be of the same potency, 20 isolated from waterfowl in two wildlife management areas of Pennsylvania.
J.Wildl.Dis. 31:179-185. 1995.
or more individual sera should be tested from a flock. All AGID- 4. Allan, G.M and MS. McNulty. A direct immunofluorescence test for the
positive sera samples should be subtyped by HI and NI tests. rapid detection of avian influenza virus antigen in tissue impression smears.
An ELISA for Antibody Detection in the Host. Several indirect Avian Pathol. 14:449-460. 1985.
ELISA test kits to detect type A group-specific RNP and M 5. Bankowski, R.A. Introduction and objectives of the symposium. In:
antibodies are commercially available (Synbiotics, San Diego, CA; Proceedings of the First International Symposium on Avian Influenza, RA
IDEXX, Westbrook, ME). ELISA procedures have also been Bankowski, ed. U.S. Animal Health Association, Richmond, Virginia, pp.
developed for internal use by diagnostic laboratories (1). The vii-xivl981.
indirect ELISA is a reliable test amenable to semi-automation and 6. Beard, C.W. Demonstration of type-specific influenza antibody in
mammalian and avian sera by immunodiffusion Bull.World Health Organ.
the rapid survey of large numbers of samples, but test results should
42:779-785. 1970.
be interpreted on a flock and not an individual bird basis. Strong 7. Brown, C.C., H.J. Olander, and D A. Senne. A pathogenesis study of
positive reactions on ELISA correlate well with AGID. However, highly pathogenic avian influenza virus H5N2 in chickens, using
reactions in the suspect range should be retested by AGID or HI immunohistochemistry. J.Comp.Pathol. 107:341-348. 1992.
tests to confirm ATV infections. The threshold for ELISA-positive
reactions should be set to avoid false positives. Indirect ELISA tests
133
David E. Swayne, Dennis A. Senne and David L. Suarez

8. Deusen, R.A., V.S. Hinshaw, D.A. Senne, D. Pellacani, and R.A. Van- 20. Slemons, R.D. and D.E. Swayne. Replication of a waterfowl-origin
Deusen. Micro neuraminidase-inhibition assay for classification of influenza influenza virus in the kidney and intestine of chickens. Avian Dis. 34:277-
A virus neuraminidases. Avian Dis. 27:745-750. 1983. 284. 1990.
9. Elbers, AR.W, T.H.F. Fabri, T.S. de Vries, J.J. de Wit, A Pijpers, and 21. Spackman, E., D.A. Senne, S. Davison, and D.L. Suarez. Sequence
G. Koch. The highly pathogenic avian influenza A (H7N7) virus epidemic in analysis of recent H7 avian influenza viruses associated with three different
The Netherlands in 2003--lessons learned from the first five outbreaks. outbreaks in commercial poultry in the United States. J Virol. 77:13399-
Avian Dis. 48:691-705. 2004. 13402. 2003.
10. Hirst, M, C.R. Astell, M Griffith, S.M Coughlin, M Moksa, T. Zeng, 22. Spackman, E., D.A. Senne, T.J. Myers, L.L. Bulaga, L.P. Garber, ML.
D.E. Smailus, R.A. Holt, S. Jones, MA. Mana, M Petrie, M Krajden, D. Perdue, K. Lohman, L.T. Daum, and D.L. Suarez. Development of a real­
Lawrence, A Mak, R. Chow, D.M Skowronski, Tweed S Aleina, S. Goh, time reverse transcriptase PCR assay for type A influenza virus and the
R.C. Branham, J. Robinson, V. Bowes, K. Sojonky, S.K. Byme, Y. Li, D. avian H5 and H7 hemagglutinin subtypes. J.Clin.Microbiol. 40:3256-3260.
Kobasa, T. Booth, and M Paetzel. Novel avian influenza H7N3 strain 2002.
outbreak, British Columbia. Emerging.infectious. diseases. 10:2192-2195. 23. Suarez, D.L. and C.S. Schultz. Immunology of avian influenza virus: a
2004. review. Dev.Comp.Immunol. 24:269-283. 2000.
11. Kodihalli, S., V. Sivanandan, D.A. Halvorson, K.V. Nagaraja, and MC. 24. Suarez, D.L., D.A Senne, J. Banks, I.H. Brown, S.C. Essen, C.W. Lee,
Kumar. Antigen-capture ELISA for rapid diagnosis of avian influenza virus RJ. Manvell, C. Mathieu-Benson, V. Moreno, J.C. Pedersen, B. Panigrahy,
in commercial turkey flocks. Journal of Veterinary Laboratory H. Rojas, E. Spackman, and D.J. Alexander. Recombination resulting in
Diagnosticians 5:438-440. 1993. virulence shift in avian influenza outbreak, Chile. Emerging Infectious
12. Lee, C.W., D.E. Swayne, J.A. Linares, D.A. Senne, and D.L. Suarez. Diseases 10:693-699. 2004.
H5N2 avian influenza outbreak in Texas in 2004: the first highly pathogenic 25. Suarez, D.L., P.R. Woolcock, A.J. Bermudez, and D.A Senne. Isolation
strain in the United States in 20 years? J. Virol, in press2005. from turkey breeder hens of a reassortant H1N2 influenza virus with swine,
13. OIE. International Animal Health Code. human, and avian lineage genes. Avian Dis. 46:111-121. 2002.
http://www.oie.int/eng/normes/MCodeZA 00003.htm 2002. 26. Swayne, D.E. Microassay for measuring thermal inactivation of H5N1
14. OIE. Highly pathogenic avian influenza in South Africa. OIE Disease high pathogenicity avian influenza virus in naturally-infected chicken meat.
Information 17(33):http://www.oie.int/eng/info/hebdo/AIS 32.HTM2005. International Journal of Food Microbiology In press:2006.
15. Palmer,D.F., M.T.Coleman, W.D.Dowdle, and G.O.Schild. Advanced 27. Swayne, D.E. and J.R. Beck. Heat inactivation of avian influenza and
laboratory techniques for influenza diagnosis. Immunology series no. 6. U.S. Newcastle disease viruses in egg products. Avian Pathol. 33:512-518. 2004.
Department of Health, Education and Welfare, Public Health Service, 28. Swayne, D.E. and D.A. Halvorson. Influenza. In: Diseases of Poultry,
Centers of Disease Control, Atlanta, Georgia. 1975 11th ed. Y.M Saif, H.J. Barnes, AM Fadly, J.R. Glisson, L.R. McDougald,
16. Rojas, H., R. Moreira, P. Avalos, I. Capua, and S. Marangon. Avian and D.E. Swayne, eds. Iowa State University Press, Ames, LA. pp. 135-160.
influenza in poultry in Chile. Vet.Rec. 151:1882002. 2003.
17. Schulman, J.L. and E.D. Kilbourne. Independent variation in nature of 29. Swayne, D.E. and RD. Slemons. Evaluation of the kidney as a potential
hemagglutinin and neuraminidase antigens of influenza virus: site of avian influenza virus persistence in chickens. Avian Dis. 36:937-944.
distinctiveness of hemagglutinin antigen of Hong Kong-68 virus. 1992.
Proc.Natl.Acad.Sci.U.S.A. 63:326-333. 1969. 30. Tang, Y., C.W. Lee, Y. Zhang, D.A Senne, R. Dearth, B. Byrum, D.R.
18. Senne, D.A., B. Panigrahy, Y. Kawaoka, J.E. Pearson, J. Suss, M Perez, D.L. Suarez, and Y.M Saif. Isolation and characterization of H3N2
Lipkind, H. Kida, and R.G. Webster. Survey of the hemagglutinin (HA) influenza A virus from turkeys. Avian Dis. 49:207-213. 2005.
cleavage site sequence of H5 and H7 avian influenza viruses: amino acid 31. Vakharia, V.N., E.T. Mallinson, and P.K. Savage. A 15-min test for Al.
sequence at the HA cleavage site as a marker of pathogenicity potential. Broiler Industry July:42-46. 1996.
Avian.Dis. 40:425-437. 1996. 32. Webster, R.G. and C.H. Campbell. An inhibition test for identifying the
19. Siemens, RD., R.S. Cooper, and J.S. Orsbom. Isolation of type-A neuraminidase antigen on influenza viruses. Avian Dis. 16:1057-1066. 1972.
influenza viruses from imported exotic birds. Avian Dis. 17:746-751. 1973. 33. Wood, G.W., J.W. McCauley, J.B. Bashiraddin, and D.J. Alexander.
Deduced amino acid sequences at the haemagglutinin cleavage site of avian
influenza A viruses of H5 and H7 subtypes. Arch.Virol. 130:209-217. 1993.

134
30
NEWCASTLE DISEASE VIRUS AND OTHER AVIAN PARAMYXOVIRUSES
Dennis J. Alexander and Dennis A. Senne

SUMMARY. Avian paramyxoviruses are members of the family Paramyxoviridae, genus Avulavirus. Nine serotypes are recognized and are
termed APMV-1 (which is synonymous with Newcastle disease virus [NDV]) to APMV-9. APMV-1 viruses naturally infect a large range of
avian species, but other avian paramyxoviruses appear to be restricted in their host distribution. APMV-1, APMV-2, APMV-3, APMV-6 and
APMV-7 viruses have produced significant disease in naturally infected poultry. In poultry, NDVs can cause a range of clinical signs from
subclinical to sudden very high mortality, depending on virus strain and host. Virulent NDV is a World Organization for Animal Health
(OLE) listed notifiable disease and subject to international reporting.
Agent Identification. Diagnosis of all avian paramyxoviruses is by isolation and identification of the virus. Virus isolation is usually done
by the inoculation of embryonating chickens’ eggs. For NDVs, it is necessary to test for pathogenicity to determine if statutory control
measures should be enforced. Vaccination of poultry and other birds for Newcastle disease is practiced throughout the world and vaccination
for APMV-3 infections in turkeys is done in some countries. For NDVs, tests based on RT-PCR have been developed that facilitate detection
of the virus in clinical specimens from infected animals; in addition a molecular-based assessment of virulence is also available.
Serologic Detection in the Host. Serologic tests have limited diagnostic value in vaccinated poultry, but in unvaccinated birds, may provide
a rapid early indication of infection. Caution should be exercised in relating positive serology to disease signs in view of the subclinical
nature of infections of birds with most avian paramyxoviruses and because of cross-reactions that may occur between serotypes.

INTRODUCTION in chickens and turkeys may cause few signs of disease in ducks.
The clinical signs that may be associated with ND are respiratory
Three virus families, Rhabdoviridae, Filoviridae and distress; diarrhea; cessation of egg production; depression; edema of
Paramyxoviridae, form the order Mononegavirales. These are head, face, and wattles; nervous signs; and death. Some, all, or none
viruses with negative-sense, single-stranded, nonsegmented, RNA of these signs may be present.
genomes. The family Paramyxoviridae has two subfamilies. The Avian paramyxovirus type 2 and APMV-3 infections of turkeys
subfamily Pneumovirinae consists of two genera, Pneumovirus, are common in some countries (including the United States) and are
which includes the mammalian respiratory syncytial viruses and usually associated with respiratoiy disease and egg production
Metapneumovirus which includes the avian pneumovirus problems. APMV-2 viruses have afso been reported in chickens.
responsible for turkey rhinotracheitis and swollen head syndrome. APMV-3 viruses have been associated with nervous disease with
The subfamily Paramyxovirinae consists of five genera. The genus high mortality in some captive psittacine species. APMV-6 and
Morbillivirus includes measles, rinderpest, and the distemper AMPV-7 virus infections in turkeys have been associated with
viruses. The genus Paramyxovirus includes Sendai virus and other respiratory signs and elevated mortality. APMV-5 viruses have
mammalian parainfluenza viruses. The genus Henipavirus which is been isolated from budgerigars and lorikeets in association with
formed from the recently discovered Nipah and Hendra viruses. The invariably lethal disease. All other isolations of other serotypes of
genus Rubulavirus includes mumps virus and human parainfluenza avian paramyxoviruses have been made from commercial or feral
viruses 2 and 4. Newcastle disease (ND) virus (APMV-1), and the birds with no associated disease.
other avian paramyxoviruses (APMV-2 to APMV-9) are placed in
the genus Avulavirus (16). The name avian paramyxoviruses has SAMPLE COLLECTION
been retained rather than adopting the name avulaviruses. It should
be noted that avian paramyxovirus type 1 (APMV-1) and ND virus For virus isolation
(NDV) are synonymous, although the term pigeon paramyxovirus For all avian paramyxoviruses, the samples associated most
type 1 (PPMV-1) has been used to distinguish the antigenic variant consistently with successful virus isolation have been feces or
APMV-1 virus responsible for the continuing panzootic in racing cloacal swabs. Viruses have also been isolated frequently from the
and other types of pigeons. Nine distinct serotypes of avian respiratory tract using tracheal/orophaiyngeal swabs. Thus, it is
paramyxovirus have been defined using standard serologic tests (6). important when taking samples for virus isolation that
The prototype strains are listed in Table 30.1. tracheal/oropharyngeal swabs and cloacal swabs or fecal material
Avian paramyxovirus type 1 (or ND) viruses are important are included regardless of the clinical signs or the postmortem
pathogens for birds of all types, and in most countries infection with lesions. Additional samples collected from dead birds should reflect
virulent forms is a notifiable disease, suspicion of which requires the clinical signs (e.g., brain if neurologic signs were evident) and
immediate notification of animal health authorities. It is an OIE the obviously affected organs. Virulent NDV is commonly isolated
listed notifiable disease. NDV has a worldwide distribution, from lung, spleen, liver, heart, and brain. Ideally, all samples should
although in some countries, only viruses of low virulence for be dealt with separately. In practice, it is usual to make pools of the
chickens have been reported. NDV has a large host range and has organs and tissues, but to treat tracheal/oropharyngeal swabs and
been reported to infect more than 240 species of birds, probably all fecal samples separately, both from each other and from organ and
species of bird are susceptible to infection. tissue samples. However, tissues/organs from different birds should
not be pooled, to avoid the possibility of neutralization of ND virus
CLINICAL DISEASE from one bird by specific antibodies from another bird. For birds
showing neurologic signs before death, especially when
The clinical signs seen in birds infected with NDV vary widely investigating APMV-1 infections in pigeons, it is advisable to
and are dependent on factors such as the virus strain, host species, process brain samples separately from other organs and tissues. In
age of host, presence of other organisms, environmental stress, and NDV vaccinated flocks suspected of being infected with virulent
the immune status of the host. In chickens the disease caused by NDV, sampling of daily mortalities has been shown to be more
different strains of NDV may vary from sudden death with 100% productive in detecting the virus than the random sampling of birds
mortality to subclinical infection. The influence of host species may in the flock.
be equally marked and, for example, viruses causing severe disease Swabs should be placed in sufficient antibiotic medium to ensure
full immersion. Fecal samples and finely minced tissues or organs
135
Dennis J. Alexander and Dennis A. Senne

should be placed in antibiotic medium as 10-20% w/v suspensions. PREFERRED CULTURE MEDIA AND SUBSTRATES
The antibiotic medium should be based on phosphate-buffered
saline at pH 7.0-7.4 (checked after the addition of antibiotics). Virus isolation
Protein based media e.g. brain heart infusion (BHI) or tris-buffered Newcastle disease virus and most other avian paramyxoviruses
tryptose broth (TBTB) have also been used and may give added will grow well in a variety of cell culture systems. Avirulent NDVs
stability to the virus, especially during shipping. The antibiotics and other avian paramyxoviruses usually require the addition of
used and their concentrations may be varied to suit local conditions trypsin (0.005-0.01 mg/ml) to facilitate growth in cell cultures.
and availability. Very high levels of antibiotics may be necessary However, the most sensitive method of isolation for all these
for fecal samples; suggested levels are: 10,000 IU/ml penicillin, 10 viruses is the inoculation of embryonating chicken eggs.
mg/ml streptomycin, 0.25 mg/ml gentamycin, and 5,000 IU/ml Samples in antibiotic media should be left at least 30 min at room
nystatin. These levels can be reduced up to fivefold for tissues and temperature before inoculation (this step can be omitted if samples
tracheal swabs. If control of Chlamydophila is desired 0.05-0.1 have been stored overnight or longer at -4 C). Suspensions of
mg/ml oxytetracycline should be included. tissues and organs or swab washings should be centrifuged at 1000
Successful isolation is enhanced by rapid processing of samples. x g for 10-20 min, and 0.1-0.3-ml volumes of the supernatant are
Virus infectivity in tissues or organs is destroyed by putrefaction inoculated into the allantoic cavity of four or five 9 to 11-day-old
and this should be avoided by refrigerating samples at 4 C or on ice embryonating chicken eggs. Eggs should be obtained from a
during transport or storage. Placing samples in antibiotic medium specific-pathogen-free (SPF) flock; if this is not possible, eggs
for transportation should be done in addition to chilling and not as obtained from a flock free of NDV antibodies may be considered.
an alternative to chilling. Most avian paramyxoviruses appear to be Use of eggs from antibody-positive flocks will reduce the ability of
able to survive in fecal samples for long periods even at relatively the virus to grow and the success of virus isolation.
high ambient temperatures. Most NDVs will survive a single Inoculated eggs should be placed at 37 C and candled regularly.
freeze-thaw cycle with little loss in infectivity, but this may not be Eggs with dead or dying embryos and all eggs 5-7 days after
the case with other avian paramyxoviruses. inoculation should be chilled to 4 C, and the allantoic/amniotic
fluids collected and tested for hemagglutinating activity. Ideally,
For RT-PCR negative fluids diluted 1:10 in antibiotic medium should be
The use of real time RT-PCR (rRT-PCR) assays in diagnostic passaged at least once more. Some laboratories use undiluted
laboratories is becoming increasingly popular. A one step rRT-PCR allantoic fluid for the second and subsequent passages; no work has
assay has been shown to be highly sensitive in detecting NDV RNA been done to show which method is the moje sensitive. To expedite
in clinical samples (21) and has been used in the U.S. as a front line diagnosis, some laboratories have used two 3 day passages and
test in place of virus isolation in outbreaks of virulent ND and for reported comparable results to two 6 day passages, but this has not
surveillance. yet been fully evaluated. Positive fluids need to be tested for
Samples suitable for the isolation of NDV, for the most part, also freedom from bacteria. If bacteria are present the fluids may be
can be used for rRT-PCR (see collection of samples for virus passed through a 450-nm membrane filter or centrifuged to remove
isolation above). However, tracheal or oropharyngeal swabs are the bacteria and repassaged in eggs after the addition of more
specimens of choice because they contain the nucleic acid, are easy antibiotics.
to process, and generally contain less extraneous organic material Avian paramyxovirus type 5 viruses do not grow in the allantoic
that can interfere with RNA recovery and amplification by PCR. cavity of chick embryos, and several reports exist of greater success
Particular attention should be given to the selection of an at isolating some other avian paramyxoviruses by inoculation into
appropriate method for RNA extraction to maximize recovery. the yolk sac rather into than the allantoic cavity. For avian
Good quality RNA not containing PCR inhibiting substances is paramyxovirus isolations, consideration should be given to
essential for successful amplification in the rRT-PCR assay. Also, inoculation of material in to the yolk sac of 6 to 8-day-old chick
use of an internal positive control (unrelated to the target cDNA) is embryos in addition to the allantoic cavity of 9 to 11-day-old eggs;
highly recommended to monitor for presence of PCR inhibiting this is essential for APMV-5 viruses.
substances that could cause a false negative test result. Several
commercial kits are suitable for the recovery of RNA from clinical RT-PCR
specimens. The three commercial RNA extraction kits that have For NDV, most RT-PCR assays have been developed to aid in the
been most thoroughly evaluated for rRT-PCR assays are Trizol identification or characterization of isolates by using primers that
reagent (Invitrogen, Carlsbad, CA), RNeasy (Qiagen, Valencia, CA) amplify portions of the genome related to a specific function, for
and MagMAX (Ambion, Austin, TX). Trizol is an organic example the fusion gene cleavage site and virulence (3, 10, 14, 19).
extraction method that works well for most specimens and has been Both conventional RT-PCR and nested RT-PCR assays have been
shown to be best for extraction of RNA from suspensions of tissue. used. More recently, a one step rRT-PCR assay has been developed
Although the Trizol kit will also work well for swab specimens, the to aid in the detection of NDV specific RNA in clinical samples as
toxic chemicals (phenol, guanidine isothiocyanate, and chloroform) well as to provide a rapid method to distinguish between viruses of
required for this method makes it less desirable than other methods. low and high virulence (21). This is especially important in
The RNeasy extraction kit has been used extensively for processing outbreaks of virulent NDV where birds may be co-infected with
tracheal and oropharyngeal swabs but has lower sensitivity for both vaccine and virulent virus.
tissue suspensions and cloacal swabs, most likely because of the In general, RT-PCR assays have four steps: isolation of the RNA;
heavy organic load in these specimens; the extraneous RNA conversion of the viral RNA to cDNA by the use of reverse
competes for binding sites in the extraction column, resulting in transcriptase; amplification of the cDNA by PCR; and evaluation of
lower recovery rates of target RNA. The MagMAX kit can be the PCR product (amplicon) for presence or absence of the target
performed in 96-well plates and therefore is more suitable than DNA sequence. Specificity of the amplicon is usually confirmed by
other extraction methods for high throughput testing. However, the electrophoresis of the amplicon in an agarose gel containing
suitability of this method for extraction of tissue suspensions and ethidium bromide, then comparing the size of the amplicon to
cloacal swabs has not yet been fully evaluated. markers of known molecular weight. However, other methods such,
as enzyme restriction endonuclease assays and direct sequencing
have been used to confirm specificity of the amplicon. The need for
post-amplification processing of the amplican is one of the major
136
Chapter 30 Newcastle Disease Virus and other Avian Paramyxoviruses

drawbacks of using RT-PCR as a diagnostic assay because of the enzyme-linked immunosorbent assay (ELISA), for identifying and
high potential for contamination of the laboratory and cross serotyping isolated viruses. However, the conventional method
contamination of samples. Extreme precautions, such as the use of usually employed is the HI test, which is described in Chapter 47 on
dedicated equipment and separation of sample processing activities serological procedures.
from post amplification processing activities, must be taken to Serotype identification for avian paramyxoviruses is usually
prevent cross contamination. straightforward, although some levels of cross- reaction may be
Real time RT-PCR assays differ from conventional RT-PCR seen between the various serogroups. The most important of these is
assays in that they require special thermocyclers and use the cross-reaction seen in HI tests between APMV-1 and APMV-3
fluorogenic hydrolysis (Taqman) probes or fluorescent dyes that, viruses, particularly when the latter are isolated from psittacines or
respectively, bind to specific targets on the PCR product or non- other caged or exotic birds. Usually, confusion due to this
specifically to any double stranded DNA molecule, to monitor for relationship can be avoided if adequate control sera and antigens are
presence of target DNA after each PCR cycle, thus providing results used.
in real time. The major advantages of the one step rRT-PCR assay Several laboratories have produced monoclonal antibodies (mAbs)
is that results can usually be obtained in less than three hr and the to representatives of some of the avian paramyxovirus serotypes
elimination of the post amplification processing step, thus reducing and these may be employed in HI tests or other tests to give highly
concerns about cross contamination of samples. Unfortunately, specific serotyping of avian paramyxoviruses and even divisions
many smaller laboratories have been hampered from using this within serotypes (see below). mAbs can be added to the initial
technology because of the high start-up costs associated with battery of antisera used for virus identification to give maximum
purchasing the real time thermocyclers. information about an isolate at an early stage.
The rRT-PCR assay described by Wise et al. (21) for detection of
NDV RNA in clinical specimens was developed and validated Morphology and Physicochemical Properties
during an outbreak of virulent ND in the U.S. in 2002-03 and Avian paramyxoviruses are RNA viruses with helical capsid
eventually replaced virus isolation as the front line test during the symmetry and have an nonsegmented, single-stranded genome of
outbreak. The rRT-PCR assay showed a sensitivity of 95% when negative sense. The RNA has a molecular weight of about 5 x 106,
results were compared to virus isolation on more than 1,400 which makes up about 5% by weight of the virus particle.
specimens. The assay has three sets of primers and probes that are Nucleotide sequencing of the complete NDV genome has shown it
used in separate reactions: a matrix primer/probe set that is to consist of 15,156 nucleotides, although there may slight
designed to detect most strains of NDV, a fusion primer/probe set variations with different strains. The capsid of avian
that can identify virulent strains of NDV (including many PPMV-1 paramyxoviruses is assembled in the cytoplasm and becomes
viruses) and a primer/probe set designed to detect low virulent enveloped by modified cell lipoprotein membrane as the virus is
strains of the virus. Samples are first screened with the matrix budded from the cell surface. Two functional virus glycoproteins
primers/probe then positive specimens are tested with the low are inserted in the envelope, one (HN) possesses hemagglutination
virulent and fusion and primers/probe sets to confirm presence of and neuraminidase activities, the other (F) is the fusion protein.
low or highly virulent virus, respectively. At the peak of the During the infection process the HN protein is responsible for
outbreak, between 1,000 and 1,500 samples were tested daily by attaching the virus to the cell and the F protein brings about fusion
rRT-PCR. Testing capacity is mostly controlled by the extraction between the cell and virus membranes to allow the genetic material
method used and availability of sufficient number of real time to enter the cell.
thermocyclers. In the U.S. the rRT-PCR assay has been authorized As seen by negative contrast electron microscopy, avian
for use in more than 35 laboratories as a front line surveillance tool paramyxoviruses consist of pleomorphic particles that are usually
for NDV. rounded and 100-500 nm in diameter or are present as filamentous
forms about 100 nm across. Surface projections on the envelope,
AGENT IDENTIFICATION approximately 8 nm long, represent the HN molecule, with the F
molecules forming smaller projections. Virus envelopes are
Virus Identification frequently disrupted so that a characteristic herring-bone
Hemagglutination activity in bacteria-free fluids will be due to one nucleocapsid about 18 nm across may be seen emerging or free in
of the nine avian paramyxovirus serotypes or one of the 16 virtually all paramyxovirus electron microscope preparations. The
influenza A virus subtypes. Most diagnostic laboratories will be presence of nucleocapsid may serve as a useful method for
primarily concerned with the presence of APMV-1 (NDV), which distinguishing paramyxoviruses from influenza viruses, as the
can be confirmed or ruled out by hemagglutination-inhibition (HI) nucleocapsid of the latter is rarely ever seen.
tests with specific antiserum. Alternatively, procedures based on
RT-PCR [see below] can be used to confirm the presence of Pathogenicity of NDV
APMV-1 (NDV) at this stage. In laboratories where differentiation Isolates of NDV require further diagnostic characterization in
of the avian paramyxovirus serotypes is done routinely, subjecting addition to identification as APMV-1 serotype. Not only may
the hemagglutinating agent to HI tests against a range of specific different NDV strains show extremes of pathogenicity, but also
antisera representing all the serotypes is usually considered most viruses of low virulence are enzootic in feral birds in most
practicale. countries. Use of live vaccines is almost universal, and exacerbation
Antisera used for virus identification is generally prepared in of infections with viruses of low virulence may mimic disease
chickens. Most laboratories have their own methods for the produced by highly virulent virus. Therefore, characterization to
production of such antisera. For APMV-1 viruses of low virulence assess the virulence of the isolated virus is extremely important to
and other avian paramyxoviruses, suitable serum can usually be ensure that the virus conforms to the definitions laid down in
obtained by the infection of 6- to 9-wk-old SPF chickens with 0.1 control and/or eradication policies.
ml of infectious allantoic fluid by injection into the leg muscle and Pathotypes. Newcastle disease virus strains and isolates have
nasal instillation of a further 0.1 ml of infectious allantoic fluid at been grouped into five clinico-pathological groups that relate to the
the same time. This is repeated after 2 wk and the serum collected disease signs and lesions produced in infected fully susceptible
after another 3 wk. chickens (9): 1) viscerotropic velogenic NDV, which produces
Specific polyclonal chicken sera can be used in most serologic acute lethal infections in which hemorrhagic lesions are prominent
tests, such as virus neutralization, agar gel immunodiffusion, and in the gut; 2) neurotrophic velogenic NDV, which produces high
137
Dennis J. Alexander and Dennis A. Senne

mortality preceded by respiratory and neurologic signs (gut lesions Thus there appears to be the requirement of a basic amino acid at
are absent); 3) mesogenic NDV, which produces low mortality, residue 113, a pair of basic amino acids at 115 and 116 plus a
acute respiratory disease, and nervous signs in some birds; 4) phenylalanine at residue 117 if the virus is to be virulent for
lentogenic NDV, which produces mild or inapparent respiratory chickens (Table 30.3). The presence of these basic amino acids at
infections; and 5) asymptomatic enteric NDV, which are avirulent these positions means that cleavage can be affected by a protease or
viruses that appear to replicate primarily in the gut. These groups proteases present in a wide range of host tissues and organs, but for
are not completely clear-cut and some overlapping between the lentogenic viruses, cleavage can occur only with proteases
signs associated with the different groups has been reported. recognizing a single arginine, i.e. trypsin-like enzymes. Lentogenic
If an NDV isolate is suspected to be virulent for chickens or other viruses are therefore restricted in the sites where they are able to
poultry species, it should be submitted to a reference laboratoiy for replicate to areas with trypsin-like enzymes, such as the respiratory
assessment. In the United States such viruses may be propagated and intestinal tracts, whereas virulent viruses can replicate and
only in a biosafety level 3 (BSL-3) laboratoiy, and similar or stricter cause damage in a range of tissues and organs resulting in a fatal
restrictions apply in many other countries. systemic infection.
This molecular basis of virulence has been incorporated into the
Pathogenicity Tests for NDV. internationally recognized definition of viruses that cause ND for
Several pathogenicity tests have been developed to differentiate which control measures should be imposed and trade restrictions
between NDV isolates of high and low virulence with some level of may be applied, as an alternative to the ICPI test. Defining such
standardization. However, international agencies such as the World viruses as those where “Multiple basic amino acids have been
Organization for Animal Health [OIE] have adopted the demonstrated in the virus (either directly or by deduction) at the C-
intracerebral pathogenicity index [ICPI] test as the in vivo test used terminus of the F2 protein and phenylalanine at residue 117, which
for defining the virulence of NDV isolates. is the N-terminus of the Fl protein. The term ‘multiple basic amino
Intracerebral Pathogenicity Index (ICPI). The ICPI is determined acids’ refers to at least three arginine or lysine residues between
by inoculating 0.05 ml of a 1:10 dilution of infectious, bacteria-ffee residues 113 and 116. Failure to demonstrate the characteristic
allantoic fluid in sterile isotonic saline (antibiotics must not be pattern of amino acid residues as described above would require
present) into the brains of each of 10 1-day-old (24 to 40 hr-old) characterization of the isolated virus by an ICPI test. [In this
chicks from SPF parents. The birds are observed daily for 8 days definition, amino acid residues are numbered from the N-terminus
and, at each observation, scored 0 if normal, 1 if sick, and 2 if dead. of the amino acid sequence deduced from .the nucleotide sequence
Birds that are sufficiently sick to be unable to eat or drink must be of the F0 gene, 113-116 corresponds to residues -4 to -1 from the
killed humanely and scored as dead at the next observation. The cleavage site.]”
ICPI value is the mean score per bird per observation. Viruses very It is worth noting that all the so-called mesogenic viruses fall
virulent for chickens give values approaching 2, lentogenic viruses within this definition, possessing basic amino acids at the F0
give values close to 0 (Table 30.2). cleavage site and having ICPI values >0.7.
Some evidence exists that NDV isolates from unusual species may The method by which the presence of multiple basic amino acids
not show their true virulence for chickens in pathogenicity index at the cleavage site can be determined has largely been limited to
tests until passaged several times in chickens. RT-PCR amplification of the coding sequences surrounding the
By internationally accepted definition, viruses that cause ND for fusion protein cleavage site, followed by sequencing of the
which control measures should be imposed and trade restrictions amplicon by automated sequencing methods (19). This approach
may be applied are those with an ICPI of 0.7 or greater. has been used to obtain sequences from a large number of isolates
When tested, other avian paramyxoviruses have usually produced of different temporal, geographical and host origins. However, as
indices similar to those of lentogenic NDV isolates. However, with any molecular based assay, success in the amplification of the
several reports have been made of psittacine APMV-3 viruses target sequence will depend on the sensitivity of the primers used.
producing ICPI values of 1.0 or more, which emphasizes the need Although the primers published by Seal et al. (19) work well for
to differentiate APMV-1 viruses from other avian paramyxoviruses most NDV strains, they have shown limited sensitivity in
correctly. amplifying viruses of genotype 6 isolated from ducks. Other
methods for differentiating low from highly virulent NDV by RT-
Molecular Assessment of Pathogenicity. PCR have been described (3, 10); however, they do not provide
An understanding of the molecular basis that controls the virulence direct confirmation of the presence or absence of multiple basic
of NDV strains (18) has meant that it is now possible, using amino acids at the fusion protein cleavage site
nucleotide sequencing techniques, to assess whether or not an
isolate has the genetic make up to be highly pathogenic for poultry. Newcastle Disease Virus Strain Differentiation Using
The viral F protein brings about fusion between the virus membrane Monoclonal Antibodies
and the cell membrane so that the virus genome enters the cell and Using conventional serologic techniques, NDV strains and isolates
replication can begin. The F protein is therefore essential for had been considered to form an antigenically homogeneous group.
replication, but during replication, NDV particles are produced with This meant that diagnosis usually resulted in little information
a precursor glycoprotein, F0, that has to be cleaved to Fl and F2 relating to the source or the spread of the virus involved. MAbs may
polypeptides, which remain bound by disulphide bonds, for the detect slight variations in antigenicity, such as single amino acid
virus particles to be infectious. This post translation cleavage is changes at the epitope to which the antibody is directed. Several
mediated by host cell proteases. groups of workers have developed mAbs against NDV strains,
The cleavability of the F0 molecule has been shown to be related primarily for diagnostic or epidemilogical purposes. Some workers
directly to the virulence of viruses in vivo. A large number of have used mAbs to distinguish between specific viruses, for
studies have confirmed the presence of multiple basic amino acids example, two groups have described mAbs which distinguish
at the F0 cleavage site in virulent viruses. Usually the sequence has between the common vaccine strains Hitchner Bl and La Sota (11,
been 113RQK/RR
*
F 117 in virulent viruses, but most have had a basic 17) whereas other mAbs have been used in distinguish vaccine
amino acid at position 112 as well. In contrast, viruses of low
virulence usually have the sequence 113K/RQG/ER * L 117.

138
Chapter 30 Newcastle Disease Virus and other Avian Paramyxoviruses

Table 30.1. Avian Paramyxoviruses.


Prototype virus strain Usual natural hosts Disease produced in naturally infected poultry
Very common, worldwide, varies from very severe to inapparent disease,
APMV-1/Newcastle disease virus Numerous depending on strain and host infected
Common, probably worldwide, mild respiratory disease or egg production
APMV-2/chicken/Califomia/Yucaipa/56 Turkeys, passerines problems; severe if exacerbation occurs
Turkeys only, in North America and Europe, mild respiratory disease but
severe egg production problems worsened by exacerbating organisms or
APMV-3a/ turkey/Wisconsin/68 Turkeys environment
APMV-3A/parakeet/Netherlands/449/75 Psittacines, passerines No infection of poultry reported
APMV-4/duck/Hong Kong/D3/75 Ducks None known
APMV-5Zbudgerigar/Japan/Kunitachi/74 Budgerigars, lorikeets No infection of poultry reported
Mild respiratory disease and slightly elevated mortality in turkeys; none in
APMV-6/duck/Hong Kong/199/77 Ducks ducks or geese
Natural infections of turkeys with respiratory disease and ostriches have been
APMV-7/dove/Tennessee/4/75 Pigeons, doves reported
APMV-8/goose/Delaware/l 053/76 Ducks and geese No infection of poultry reported
APMV-9/duck/New York/22/78 Ducks None known

viruses from an epizootic virus in a given area (20). MAb typing


was also used to establish the uniqueness of the variant NDV SEROLOGIC DETECTION IN THE HOST
responsible for the pigeon panzootic and specific mAbs have
proven particularly useful in identify the spread of this virus around Detection of an immune response to NDV or other avian
the world. paramyxoviruses may serve a useful diagnostic function, allowing
However, although use of mAb panels showed that viruses detection of infections in unvaccinated birds and the monitoring of
grouped on their ability to react with the same mAbs shared response to vaccination. Numerous serologic tests may be used to
biological and epizootiological properties and that viruses tend to detect antibodies, but the most commonly used for this group of
remain fairly well conserved antigenically during outbreaks or viruses is the HI test. Details of the procedures for this test are given
epizootics (7) this approach to epizootiological assessments of ND in Chapter 46 on titration of biological suspensions.
outbreaks has been largely supplanted by phylogenetic analysis.
Hemagglutination-Inhibition
Phylogenetic analysis Suitably diluted infectious allantoic fluid may be used as antigen
Nucleotide sequencing, after RT-PCR and phylogenetic analysis, in HI tests for all avian paramyxoviruses except APMV-5. Use of
has been used by a number of authors to assess genetic differences noninfectious antigens usually is desirable; and for NDV formalin-
and genotypes of ND viruses. It has been established that sequences inactivated virus (infectious allantoic fluid treated with 0.1%
of as little as 250 base pairs give meaningful phylogenetic analyses, formalin) is often used as antigen. However, the hemagglutinin­
comparable with those obtained with much longer sequences (15, neuraminidase (HN) protein of some avian paramyxoviruses is not
19), which means that sequencing and phylogenetic analyses can be stable when treated with formalin and for those viruses beta-
done rapidly. In the preliminary characterization studies of NDV, propriolactone (0.05% final concentration) may be used as an
six lineages (I to VI) were determined using restriction enzyme alternative to formalin for inactivation. Hemagglutinating activity
analysis (8). These groupings have subsequently been confirmed, was not detected in the original APMV-5 isolates, but later viruses
and two further lineages (VII and VHT) and several sublineages placed in this serotype have been shown to agglutinate red blood
within these have been identified through nucleotide sequencing cells if concentrated following growth in chick embryo cell cultures.
studies (13). Aldous et al., (4) studied the nucleotide sequences of a Some evidence exists that minor antigenic differences between
375-nucleotide fragment at the 3’ end of the fusion protein gene, strains of NDV can result in different HI titers with the same
which includes the region encoding the nuclear localization signal antiserum. This is particularly true using the variant PPMV-1 virus
sequence and the precursor fusion protein cleavage activation site, as antigen.
of 338 isolates of NDV representing a range of viruses of different
temporal, geographical and host origins. They concluded that the Interpretation of HI Response
isolates divided into six broadly distinct groups (lineages 1 to 6). Serum titers will vary with the amount of antigen used in the test;
Lineages 3 and 4 were further subdivided into four sublineages (a to figures quoted in this section assume 4 HA units of virus. For all
d) and lineage 5 into five lineages (a to e). Essentially lineages 1, 2, avian paramyxovirus serotypes, birds that have not been immunized
4 and 5 corresponded to the earlier defined lineages I, Π, VI and or infected usually have HI titers of less than 1:8, and nonspecific
VH, with comparable sub-lineages but that the geno-groupings ΙΠ, titers above this level are rare for most avian species. However, sera
IV, V, VUI corresponded to their sublineages (3a to 3d). In addition from other species may cause agglutination of chicken red blood
lineage 6 represent a new geno-group. Aldous et al., (4) proposed cells to titers high enough to mask low positive inhibition. This
that genotyping of NDV isolates should become part of diagnostic agglutination, caused by the presence of natural serum agglutinins,
virus characterization for reference laboratories by producing the can be removed by treatment of the sera with concentrated chicken
375-nucleotide sequence of the F gene routinely for all viruses and red blood cells before testing. Alternatively, use of red blood cells
comparing the sequences obtained with other recent isolates and 18 from the homologous species can be considered.
viruses representative of the recognized lineages and sub-lineages. In the absence of vaccination, a positive specific HI response is
Such preliminary analysis should allow rapid epidemiological confirmation of infection of the birds with that serotype. However,
assessment of the origins and spread of the viruses responsible for the possibility of cross-reaction between some avian
ND outbreaks. paramyxoviruses should be considered, especially between viruses
of APMV-1 (NDV) and APMV-3 serotypes.

139
Dennis J. Alexander and Dennis A. Senne

Inactivated vaccines for APMV-3 viruses are used in the United available commercially. The main advantage of ELISAs over more
States and Europe and use of APMV-2 vaccines has been conventional tests, such as HI tests, is that they can be semi­
undertaken in some countries. Little is known of the interpretation automated, enabling results to be obtained rapidly and
of the immune response to vaccines for these serotypes. Although inexpensively, especially when sera are to be screened for
considerable work has been done in attempts to assess the likely antibodies to several viruses (see Chapter 47 on serologic
outcome of infection with virulent NDV at different levels of procedures). As a result, ELISAs have often become the method of
immunity, care must be taken in making such assessments due to choice for flock screening programs, particularly those aimed at
the effects other factors may have. In broad terms titers at the low assessing vaccine response. Studies aimed at standardizing ELISA
end of those measurably positive may protect against death in tests and comparing them with conventional tests, for example
Adair et al., (1), have concluded that ELISAs for NDV antibodies
Table 30.2. Pathotypes and intracerebral pathogenicity indices__________ usually show good reproducibiEty, high comparative sensitivity and
Pathotype Range of indicesA Examples of viruses® specificity, and correlate well with the HI test.
ICPI The ELISAs should be modified and validated for species on
Herts 33, N.Y. Parrot which it is being used. ELISA tests are usually extremely sensitive
Viscerotropic
1.5-2.0 70181, CA2089/72 and this may restrict their value for diagnostic testing when
velogenic
antigenic relationships exist between different viruses, which is the
Neurotropic Texas GB
velogenic
1.5-2.0 case with avian paramyxoviruses. Both these problems could be
*,Roakin Komarov
*, addressed by use of competitive or blocking ELISAs employing one
Mesogenic 1.0-1.5 *,
Mukteswar *
H or more mAb to NDV, although mAb(s) should be evaluated to
Hitchner Bl *
, La determine that they react with all strains of NDV and not other
Lentogenic 0.2-0.5 *,
Sota Clone 30 * avian paramyxoviruses.
Ulster 2C
*, *,
V4
Asymptomatic enteric 0.0-0.2 MC110 DIFFERENTIATION FROM CLOSELY RELATED AGENTS
A ICPI = intracerebral pathogenicity index in 1-day-old chicks, See text
for details. Because of their abiEty to cause agglutination of red blood cells,
B Strains marked with an asterisk are used routinely as vaccines in some usually without concentration or treatment, problems in
countries. differentiating avian paramyxoviruses are primarily from each other
uncomplicated infections. However, even high titers will not and from influenza viruses. Preliminary distinction from influenza
prevent some replication of challenge virus and significant egg viruses can be done by examination in the electron microscope
production losses may occur in flocks with prechallenge mean titers where negative contrast staining will reveal morphologic
as high as 1:256. With most lentogenic live NDV vaccines, HI titers differences, most notably the presence of the characteristic
of 1:16 to 1:64 are usually obtained following a single dose. nucleocapsid emerging from disrupted paramyxovirus particles. The
Repeated vaccination with similar or more virulent Eve virus abiEty of paramyxoviruses, but not influenza viruses, to cause
vaccines may increase the immune response considerably, and if oil hemolysis of chicken red blood ceUs at pH 7.0 has also been used in
emulsion inactivated vaccines are included, titers up to 1:1024 to the past. Influenza A viruses can be confirmed in allantoic fluid by
1:4096 are common. Multiple vaccinations with either Eve-, killed- the agar gel immunodiffusion test with type A positive antiserum,
or combinations of both virus vaccines wiU also increase the level or by the use of antigen capture immunoassays (membrane bound or
of crossreactivity between serotypes of APMVs in the HI test. flow-through) designed to detect influenza A viruses. The antigen
capture immunoassay tests are not Ecensed for veterinary use but
Enzyme-linked Immunosorbent Assays for NDV Antibodies have been shown to detect all 16 subtypes of avian influenza virus
Numerous ELISA tests have been developed for the detection of (ATV) and are highly specific. Differentiation between APMV-1
antibodies to NDV and several ELISA kits for this purpose are and ATV can also be done by the use of RT-PCR (conventional and
real time), with primers directed to the highly conserved matrix
Table 30.3. Amino acid sequences at the F0 cleavage site of APMV-1 protein genes of both APMV-1 and ATV. RT-PCR assays have also
viruses modified from Aldous & Alexander (2) been developed that can discriminate between virulent and vaccine
F0 Cleavage site amino acids 111 to virus. However, it is recommended for definitive identification that
Virus strain Pathotype
119 isolates be submitted to a reference laboratory that can that can use
Herts 33 velogenic FIG-
-GRRQRR
* a battery of polyclonal sera against each of the paramyxoviruses, or
Essex ‘70 velogenic FVG-
*
-GRRQKR initially against the most likely serotypes, in HI tests.
Differentiation of avian paramyxoviruses into serotypes can be
Texas GB velogenic -GRRQKR
FIG-
*
more of a problem due to antigenic relationships between viruses of
617/83 PPMV-1 FIG-
*
-GGRQKR different serotypes, which are apparent in most conventional tests.
34/90 velogenic -GKRQKR
FVG-
* This cross reaction in serologic tests is most marked between
Beaudette C mesogenic FIG-
*
-GRRQKR APMV-1 and APMV-3 viruses (particularly APMV-3 viruses
isolated from psittacines). Usually the use of adequate control
Roakin mesogenic FIG-
*
-GRRQKR
antigens and antisera leaves tittle doubt in serotyping isolates from
La Sota lentogenic *
-GGRQGR
LIG- these groups, but mAbs specific for APMV-1 and APMV-3 have
Hitchner Bl lentogenic LIG-
*
-GGRQGR been employed to simplify differentiation (5).
asymptomatic The development and application of RT-PCR or rRT-PCR assays
D26 LIG-
-GGKQGR
* has made it possible to differentiate quickly NDV from other avian
enteric
asymptomatic paramyxoviruses based on genetic differences and to detect
MC110 LIG-
-GERQER
*
enteric mixtures of low and highly virulent ND virus in the same sample.
asymptomatic However, such assays are not yet available to confirm the presence
1154/98 LIG-
*
-GRRQGR
enteric or distinguish between the other avian paramyxoviruses.
note that all virulent viruses have phenyalalanie (F) at position 117, the Fl
N-terminus
140
Chapter 30 Newcastle Disease Virus and other Avian Paramyxovinee

REFERENCES 11. Erdei, J., K. Bachir, E. F. Kaleta, K. F. Shortridge, and B. LommoL


Newcastle disease vaccine (La Sota) strain specific monoclonal antibody.
1. Adair, B. Μ, M S. McNulty, D. Todd. T .J. Connor, and K. Bums. Arch. Virol. 96:265-269. 1987.
Quantitative estimation of Newcastle disease virus antibody levels in 12. Hanson, R. P., and C. A. Brandly. Identification of vaccine strains of
chickens and turkeys by ELISA. Avian Pathol. 18:175-192. 1989. Newcastle disease virus. Science 122:156-157. 1955.
2. Aldous, E. W. and D. J. Alexander, Technical Review: Detection and 13. Herczeg, J., S. Pascucci, P. Massi, M Luini, L. Selli, I. Capua, I. and Bl
differentiation of Newcastle disease virus (avian paramyxovirus type 1) Lomniczi. A longitudinal study of velogenic Newcastle disease vnws
Avian Pathol. 30(2): 117-128. 2001. genotypes isolated in Italy between 1960 and 2000. Avian Pathol. 30: 163-
3. Aldous, E. W., M. S. Collins, A. McGoldrick, and D. J. Alexander. Rapid 168. 2001.
pathotyping of Newcastle disease virus (NDV) using fluorogenic probes in a 14. Jestin, V., and A. Jestin. Detection of Newcastle disease virus RNA in
PCR assay. Vet. Microbiology. 80:201-212. 2001. infected allantoic fluids by in vitro enzymatic amplification (PCR). Arch.
4. Aldous, E. W., Mynn, J. K. Banks, J. and D. J. Alexander, (A molecular Virol. 118:151-161. 1991.
epidemiological study of avian paramyxovirus type 1 (Newcastle disease 15. Lomniczi, B., E. Wehmann, J Herczeg, A. Ballagi Pordany, E F.
virus) isolates by phylogenetic analysis of a partial nucleotide sequence of Kaleta, O. Werner, G. Meulemans, P. H. Jorgensen, A. P. Mante, A. L
the fiision protein gene. Avian Pathol. 32: 239-357. 2003. Gielkens, I. Capua, and J. Damoser. Newcastle disease outbreaks in recent
5. Alexander, D. Avian Paramyxoviridae— recent developments. In: years in western Europe were caused by an old (VI) and a novel genotype
Proceedings of the 1st European Society for Veterinary Virology Congress, (VII). Archiv. Virol. 143: 49-64. 1998.
Liege. Vet. Microbiol. 23:103-114. 1990. 16. Mayo, M A. A summary of the changes recently approved by ICTV.
6. Alexander, D.J. Newcastle disease, Other Avian Paramyxoviruses and Archiv Virol 147,1655 - 1656. 2002.
Pneumovirus infections: Newcastle disease. In: Diseases of Poultry. Y.M 17. Meulemans, G., M Gonze, M C. Carlier, P. Petit, A. Bumy, and L. E
Saif [ed in chief] Iowa State University Press USA pp 64-87. 2003. Long. Evaluation of the use of monoclonal antibodies to hemagglutination
7. Alexander, D. J., R. J. Manveil, J. P. Lowings, K. M Frost, M S. and fusion glycoproteins of Newcastle disease virus for virus identification
Collins, P. H. Russell, and J. E. Smith. Antigenic diversity and similarities and strain differentiation purposes. Arch. Virol. 92:55-62. 1987.
detected in avian paramyxovirus type 1 (Newcastle disease virus) isolates 18. Rott, R., and H.-D. Klenk. Molecular basis of infectivity and
using monoclonal antibodies. Avian Pathol. 26:399-418. 1997. pathogenicity of Newcastle disease virus. In: Newcastle disease. D. J.
8. Ballagi-Pordany, A., E. Wehmann, J. Herczeg, S. Belak, and B. Alexander, ed. Kluwer Academic Publishers, Boston, Mass. pp. 98-112.
Lomniczi. Identification and grouping of Newcastle disease virus strains by 1988.
restriction site analysis of a region from the F gene. Archiv. Virol. 141: 243- 19. Seal, B. S., D. J. King, and J. D. Bennett. Characterization of Newcastle
261. 1996. disease virus isolates by reverse transcription PCR coupled to direct
9. Beard, C. W., and R. P. Hanson. Newcastle disease. In: Diseases of nucleotide sequencing and development of sequence database for pathotype
poultry, 8th edition. M S. Hofstad, H. J. Barnes, B. W. Calnek, W. M Reid, prediction and molecular epidemiological analysis. J. Clin. Microb.
and H. W. Yoder, eds. Iowa State University Press, Ames, Iowa, pp. 452- 33(10):2624-2630. 1995.
470. 1984. 20. Srinivasappa, G. B., D. B. Snyder, W. W. Marquardt, and D. J. King,
10. Creelan, J. L., D. A. Graham, and S. J. McCullough. Detection and Isolation of a monoclonal antibody with specificity for commonly employed
differentiation of pathogenicity of avian paramyxovirus serotype 1 from vaccine strains of Newcastle disease virus. Avian Dis. 30:562-567. 1986.
field cases using one-step reverse transcriptase-polymerase chain reaction. 21. Wise, M G., J. C. Pedersen, D. A. Senne, D. Kapczynski, D. J. King, D.
Avian Pathology 31:493-499. 2002. L. Surarz, B. S. Seal, and E. Spackman. Development of a real time reverse
transcription-polymerase chain reaction for detection of Newcastle disease
virus RNA in clinical samples. J. Clin. Microbiol. 42(1): 329-338. 2004.

141
31
AVIAN METAPNEUMOVIRUS
Richard E. Gough and Janice C. Pedersen

SUMMARY. Avian metapneumovirus (aMPV)) is an upper respiratory tract infection primarily of turkeys and chickens caused by a virus
belonging to the genus metapneumovirus . The disease can cause significant economic losses in turkey flocks, particularly when exacerbated
by secondary pathogens. Mortality is variable but may exceed 80% in susceptible turkey poults. In turkey breeding flocks infection can
result in serious egg production problems. In chickens infection may lead to mild respiratory signs, egg production problems and swollen
head syndrome (SHS).
Agent Identification. Virus detection and identification can be difficult unless samples are taken early in the course of the disease. Virus
isolation in cell cultures, embryonated hen’s eggs, tracheal organ cultures and molecular methods have all been used successfully to detect
aMPV but the degree of success will depend on the strain of virus. Electron microscopy, virus neutralization and molecular techniques are
widely used to identify the virus. The use of monoclonal antibodies and molecular studies have revealed antigenic diversity between the
isolates studied.
Serologic Detection in the Host. Confirmation of infection is best obtained by serologic tests, particularly enzyme-linked immunosorbent
assay (ELISA) methods. In many countries where the disease is endemic vaccination is practiced which may complicate interpretation of
results.

INTRODUCTION infraorbital sinuses, the so-called swollen head syndrome (SHS),


torticollis, incoordination and depression. Unlike subtype A & B,
Avian metapneumovirus (aMPV) previously referred to as avian the U.S. strain (subtype C) has not been shown to naturally induce
pneumovirus (APV) and avian rhinotracheitis (ART) virus is an disease in chickens. Previous work has demonstrated that different
acute, highly contagious upper respiratory tract infection of turkeys strains of a MPV have a specific tropism for chickens or turkeys
and chickens. (7). Although there is evidence that aMPV can infect other species
In turkeys the virus causes a disease known as turkey rhinotracheitis of birds, clinical signs associated with the virus have been rarely
(TRT). The agent responsible for the disease has been confirmed as reported (12). Subtype D metapneumoviruses have been reported to
a member of the genus Metapneumovirus in the family occur in ducks and is associated with respiratory signs and egg
Paramyxoviridae (24). Until recently, APV was thought to be the production problems (28).
sole member of the Metapneumovirus genus but recent reports from
many countries have indicated that similar viruses have been SAMPLE COLLECTION
detected in humans associated with respiratoiy tract infection in
children (21). Avian metapneumovirus (aMPV) has been further It is very important to take samples for attempted virus isolation in
classified into four subtypes: A, B, C and D, based on nucleotide the early stages of infection. Ideally live birds in the acute phase of
and deduced amino acid sequence data (10). It is probable that other the disease should be sampled using sterile swabs from the upper
subtypes exist but have not yet been detected and identified. respiratory tract. The samples for virus isolation have been nasal
Serologic evidence suggests that the disease is now widespread exudates, choanal cleft swabs and scrapings of sinus and turbinate
throughout the world and of considerable economic importance, tissue. The virus has also been isolated from trachea and lungs, and
particularly in turkeys. Australasia appearing to be the only region occasionally viscera of affected turkey poults. Isolation of virus is
in which aMPV has not been reported. There is serological and rarely successful from birds showing severe chronic signs as the
molecular evidence that aMPV occurs in a variety of other avian extreme clinical signs are usually due to secondary adventitious
species, including pheasants, guinea fowl, ostriches, passerines and agents. This certainly applies to SHS of chickens in which the
various waterfowl (21), but there is no evidence that serious disease characteristic signs appear to be due to secondary Escherichia coli
occurs in these species. infection. Furthermore, for reasons that are unclear, virus isolation
from chickens appears to be more difficult than from turkeys.
CLINICAL DISEASE For samples requiring dispatch to a diagnostic laboratory, it
essential that the samples are sent immediately on ice. Where delays
The various clinical signs associated with aMPV infection in of more than three days are expected, the samples should be frozen
turkeys have been detailed (13). Signs in young turkey poults when collected and sent frozen. Swabs for attempted virus isolation
include snicking, rales, sneezing, nasal discharge, foaming should be sent on ice fully immersed in virus transport medium but
conjunctivitis, swelling of the infraorbital sinuses and those for PCR analysis can be sent dry.
submandibular edema. Secondary adventitious agents can For virus isolation, a 20% (v/v) suspension of the nasal exudate or
dramatically exacerbate the clinical signs. In laying turkeys, homogenised tissue is made in phosphate-buffered saline (PBS) at
infection normally results in only mild respiratory signs but with an pH 7.0-7.4 containing antibiotics. This is then clarified by
associated marked drop in egg production, as much as 70%. In centrifugation at 1000 * g for 10 min and the supernatant passed
some flocks of adult turkeys, subclinical infections have been through a 450 nm membrane filter.
detected by seroconversion. When disease is seen, the morbidity
can be as high as 100%, with mortality ranging from 0.5% in adult PREFERRED CULTURE MEDIA AND SUBSTRATES
birds to 85% in young poults. The clinical signs of infection in
chickens are less characteristic than the disease in turkeys. In To maximize the chances of successfully isolating the virus, a
laying flocks, particularly broiler breeders, there is a marked drop in multiple approach to diagnosis is recommended. This is particularly
egg production, often preceded by respiratory signs. Severe relevant when dealing with different subtypes or genotypes which
respiratoiy distress may occur in broiler chickens particularly when may require varied in vitro methods to isolate the virus. This was
exacerbated by secondary pathogens such as infectious bronchitis illustrated particularly well in North America where it was shown
virus, mycoplasmas, and Escherichia coli. The clinical signs that primary isolation of subtype C aMPV was not associated with
associated with this may be swelling of the periorbital and ciliostasis in tracheal organ cultures. This was in contrast to the
142
Chapter 31 Avian Metapneumovirus

European experience and elsewhere in which tracheal organ is rapidly destroyed by lipid solvents, heat (56 C for 30 min), and
cultures were shown to be the most reliable method for the primary extremes of pH. Studies with a subtype C strain of virus, isolated
isolation of subtype A and B aMPV (10). from turkeys in the USA, reported that the virus was resistant to pH
5-9 for one hour. The study also reported that the viability of the
Tracheal Organ Culture virus was significantly reduced after 6 hr at 50 C, 2 days at 37 C, 4
Tracheal organ cultures are prepared from turkey embryos or very wk at 20 C and less than 12 wk at 4 C. In addition, the study
young turkeys obtained from flocks free of specific antibodies to reported that several disinfectants were effective in reducing the
aMPV. Tracheas from chicken embryo or l-to-2-day-old chicks viability of the virus (29)
may also be used. Transverse sections of trachea are rinsed in PBS
(pH 7.2), placed one to a tube in Eagles medium with antibiotics, Biological Properties
and held at 37 C. For inoculation with infective material, the tubes Unlike other members of the family Paramyxoviridae, aMPV do
are drained, and 0.1 ml of bacteria-free inoculum is added. After not possess hemagglutination (HA) or neuraminidase activity.
incubation for 1 hr at 37 C, growth medium is added and the
cultures are incubated at 37 C on a roller apparatus rotating at 30 Molecular Identification
revolutions per hour. Cultures are examined daily after agitation on Due to the fastidious nature of aMPV, RT-PCR is significantly
a laboratory mixer to remove debris from the lumen. Ciliostasis more sensitive and rapid method for the detection of aMPV than
may occur within 7 days of inoculation on primary passage but standard virus isolation methods (1,23). RT-PCR procedures
usually is produced rapidly and consistently only after several blind targeted to the F, M, and G genes are used for the detection of
passages. aMPV, but are limited in specificity and have not been shown to
detect all subtypes (3,4,22,23). These subtype specific assays are
Culture in Embryonating Eggs successfully used for the detection and diagnosis of endemic strains
Six to 8-day-old embryonating chicken or turkey eggs, from flocks (17,23,26). However, limitations of subtype specific assays need to
known to be free of aMPV antibodies, are inoculated by the yolk- be recognized when conducting diagnostic testing for respiratory
sac route with 0.1-0.2 ml of bacteria-free material from infected disease. Primers directed to conserved regions of the N gene have
birds and incubated at 37 C. Within 7-10 days, there is usually been shown to have broader specificity, detecting representative
evidence of stunting of the embryos with few deaths. Consistent isolates from A, B, C, and D subtypes (3). RT-PCR assays directed
embryo mortality is normally seen only after four to five passages to the G gene have been successfully used for genotype or subtype
so this method of isolation is both time consuming and expensive. identification (14,15,17).
Nasal exudates, choanal cleft swabs, and turbinate specimens
Cell Culture collected 2-7 days post exposure are the preferred specimen. It has
Various cell cultures have been used for the primary isolation of been shown, that aMPV can be detected from specimens collected
aMPV, including chicken embryo cells, VERO cells and more 7-10 days post exposure, however, the viral concentration is
recently the QT-35 cells, with varying degrees of success. considerably less thus reducing the success of detection (1,23). Five
However, once the virus has been adapted to growth in swabs from a single flock can be pooled to increase recovery rate.
embryonating eggs or tracheal organ cultures, in which it grows Isolation of aMPV from chickens is difficult and has succeeded
only to low titers, the virus will readily replicate to high titers only in a limited number of cases, for this reason, molecular tests
following multiple passages in a variety of primary chicken or are the method of choice for the detection of aMPV in chickens
turkey embryo cells and in Vero, BSC-1 and QT-35. The virus (16). In addition, RT-PCR has been used to genotype and confirm
produces a characteristic cytopathic effect (CPE) with syncytial the identity of an aMPV isolated from chickens (17,27). It is
formation within 7 days. important to remember that PCR detects viral RNA, not live virus;
therefore, a positive PCR does not necessarily confirm an active
AGENT IDENTIFICATION infection.

Morphology Antigen Detection


By negative-contrast electron microscopy, the virus can be A number of different assays have been developed for the
identified as having a paramyxovirus-like morphology. detection of aMPV antigens using immunostaining methods. The
Pleomorphic fringed particles, roughly spherical and 80-200 nm in most popular techniques have involved the use of
diameter, are commonly seen. Occasionally much larger immunoperoxidase (IP), immunofluorescence (IF) and immunogold
filamentous forms are present, which may be up to 1000 nm in staining and are described in detail elsewhere (10). These
length. The surface projections are 13-14 nm in length and the techniques, in particular IP and IF, have been used to detect virus
helical nucleocapsid, that can sometimes be seen emerging from specific antigen in both fixed and unfixed tissues and smears from
disrupted particles, is 14 nm in diameter with an estimated pitch of turkeys and chickens.
7 nm per turn.
Immunological Detection Techniques
Physiochemical Properties Monoclonal antibodies have been used in virus neutralization tests
The genome of aMPV is unsegmented and composed of single­ to differentiate subtypes of aMPV (6,8). Neutralization tests using
stranded negative sense RNA of approximately 15 kilobases with a monospecific antiserum can also be used to confirm the identity of
helical symmetry. In sucrose gradients, the buoyant density of an viruses isolated in cell or organ cultures. Because of low
isolate from turkeys was found to be 1.21 g/ml with an approximate concentrations of virus, the method most commonly used is the
molecular weight of 500 x 106. The same virus was also shown to alpha test (constant serum-diluted virus). The serum-virus mixtures
have 8 structural polypeptides of which 2 were glycosylated and 3 are incubated at 37 C for 45 min and then assayed in cell or organ
were non-structural virus-specified proteins. These have now been cultures for viable virus. A reduction in infectivity titer of 102.0 or
identified as follows; nucleoprotein (N), phosphoprotein (P), matrix more is considered to be significant.
protein (M), second matrix protein (M2), surface glycoprotein (G), The immunodiffusion test has also been used to confirm the
fusion protein (F), a small hydrophobic protein (SH) and a viral identity aMPV isolates (13). Briefly, isolates are cultivated in cell
RNA-dependant RNA polymerase (L). In common with other cultures to give an infectivity titer of approximately 106.0 median
members of the family Paramyxoviridae, the infectivity of the virus tissue culture infective doses (TCID50) per ml. After extensive CPE
143
Richard E. Gough and Janice C. Pedersen

has occurred, the cell debris is removed and the supernatant The VN test can be carried out in confluent monolayers of chicken
concentrated by ultracentrifugation. The resulting concentrates are embryo cells in 96-well flat-bottomed microtitre plates. Briefly, 30-
treated with 0.2% N-lauroylsarcosine (Sigma, St. Louis, Mo.) to 300 median TCID of virus are reacted with two-fold serial dilutions
produce antigens for the immunodiffusion test. Using monospecific of test serum at 37 C in 5% CO2. After 1 hr, 25ul is transferred from
aMPV antiserum, the antigens are tested by double each dilution in the neutralization plate to the corresponding well on
immunodiffusion in 1% agarose. After approximately 24 hr at 37 the cell culture plate. The plates are sealed and incubated at 37 C in
C, the tests are examined and any precipitin lines that stain with 5% CO2, together with appropriate serum and virus controls. After 1
02% Coomassie brilliant blue R are interpreted as positive. hr, 200 μΐ of maintenance medium is added to each well, the plates
Included in the test are appropriate positive and negative control are sealed and incubated at 37 C in CO2 for 7 days. The cultures are
antigens and sera. A precipitin line between the reference examined daily for CPE and after 7 days the serum titres are
monospecific antiserum and test antigen confirms the identity of the determined and expressed as the reciprocal (log2) of the highest
virus. dilution of serum that completely inhibits viral CPE. A titre of >23
is considered positive.
SEROLOGIC DETECTION IN THE HOST
DIFFERENTIATION FROM CLOSELY RELATED AGENTS
Due to difficulties in isolating and identifying aMPV,
confirmation of infection is usually achieved by serological Strain Variability
methods, particularly in unvaccinated chicken flocks. The most When aMPV was first detected in Europe, it was believed there
commonly employed method is the enzyme-linked immunosorbent was only one serotype of the virus, represented by subtypes A and
assay (ELISA). Other methods that have been used to detect aMPV B. These were differentiated on the basis of nucleotide sequence
antibodies are virus neutralization, microimmunofluorescence and analysis of the attachment (G) protein gene (14) and by mAb
immunodiffusion tests. analyses (6,8). However, this situation changed with the emergence
Ideally, both acute and convalescent serum samples should be of a different aMPV in North America, designated subtype C (9,25).
obtained for testing. The sera should be heated at 56 C for 30 min More recently, reports from France have indicated the presence of a
before testing; if the testing of the sera is delayed, it should be fourth subtype of aMPV, designated subtype D (2,28). It is probable
stored at -20 C. In chickens, the serological response to aMPV that other strains of aMPV exist, which are genetically distinct from
infection is weak when compared to the response in turkeys (7). subtypes A, B, C and D. These other subtypes of the aMPV may
remain undetected using conventional f*CR based techniques;
Enzyme-Linked Immunosorbent Assay therefore a multi diagnostic approach is recommended when
Numerous commercial ELISA kits, together with in-house assays, investigating outbreaks of respiratory disease in poultry.
have been developed for the detection of aMPV antibodies (5,10)
Although the indirect ELISA has been very useful for screening Newcastle Disease
large numbers of sera for aMPV antibodies, differences in Some strains of Newcastle disease virus and other members of the
sensitivity and specificity between tests have been reported to occur genus Paramyxovirus, such as PMV-3 (see Chapter 30 on
between commercial kits (18,19). This is principally due to Newcastle disease virus and other avian paramyxoviruses), may
variations in the antigenicity and purity of the viral antigen used in cause respiratory disease and egg production problems in chickens
the preparation of the ELISA kit. It appears that that sensitivity of and turkeys that closely resemble aMPV. Paramyxoviruses are
the ELISA is less when a heterologous strain of aMPV is used as similar in morphology but can usually be easily distinguished from
antigen, even though the strain appears closely related by virus aMPV because they possess HA and neuraminidase activity.
neutralization test (10) Competitive or blocking ELISA kits have
also been developed, incorporating an aMPV specific monoclonal Infectious Bronchitis
antibody (mAb).These kits claim to have a broad spectrum of Infections of chickens with infectious bronchitis (IB) virus can
sensitivity and specificity for all subtypes of aMPV and can be used result in respiratory disease and egg production problems that are
for testing sera from a variety of avian species. ELISA antigens similar to aMPV. Swollen head syndrome (SHS) in chickens has
have been prepared in a variety of substrates including various cell also been described as being associated with infectious bronchitis
cultures and tracheal organ cultures (10). virus and E. coli (11,20) Serologic and molecular identification of
the infecting virus is probably the simplest method of making a
Fluorescent Antibody Test differential diagnosis.
Several techniques have been described for the detection of aMPV
antibodies using indirect immunofluorescence (HF) tests on infected Avian Influenza
tissues or cell cultures. Studies have shown that antibodies to Infection of chickens or turkeys with the milder strains of avian
aMPV in infected turkeys can be detected by HF 5 days after the influenza viruses can result in disease similar to aMPV. Virus
appearance of clinical signs (10). The technique has limited isolation and the demonstration of HA activity by influenza viruses
application for the large scale testing of poultry sera and has been will distinguish between the viruses.
mainly used for research purposes.
Bacteria and Mycoplasma
Virus Neutralization Test A wide range of bacteria and Mycoplasma species have been
Antibodies to the virus have been detected by standard virus reported to cause disease signs very similar to aMPV. Frequently,
neutralization (VN) techniques in tracheal organ cultures and such organisms may be present as secondary or adventitious
various sensitive cell cultures, such as CEF, chicken embryo liver, invaders and may cause considerable diagnostic problems.
and VERO cultures (13). Using a neutralization assay in CEF Distinction must rely on negative isolation or demonstration of
cells, it was shown that neutralizing antibody could be detected antibodies to aMPV .
within 5 days of the appearance of clinical signs and was declining
by day 13. There is a good correlation between VN results and
ELISA and immunofluorescence results (10). There are cross
reactions between subtype A and B viruses so the VN test is not
suitable for distinguishing subtype antibodies.
144
Chapter 31 Avian Metapneumovina

REFERENCES
15. Lwamba, H.C.M, R. Alvarez, M.G. Wise, Qingzhong, Yu, D.
1. Alkhalaf, A.N., L.A.Ward, RN.Dearth and Y. M Saif. Pathogenicity, Halvorson, MK. Njenga and B.S. Seal. Comparison of full-length genome
transmissibility and tissue distribution of avian pneumovirus in turkey sequence of Avian metapneumovirus subtype C with other paramyxoviruses^
poults. Avian Dis. 46:650-659. 2002. Virus Research. 107:83-92. 2005.
2. Bayon-Auboyer, Μ H., C. Amauld, D.Toquin and N. Eterradossi. 16. Mase, M,S. Asahi, K.Imai, K.Nakamura and S. Yamaguchi. Detection
Nucleotide sequences of the F, L and G protein genes of two non-A/non-B of turkey rhinotracheitis virus from chickens with swollen head syndrome
avian pneumoviruses (APV) reveal a novel APV subgroup. J. Gen. Virol. by reverse transcriptase-polymerase chain reaction (RT-PCR). J. Vet Med.
81:2723-2733. 2000. Sci. 58:359-361.1996.
3. Bayon-Auboyer, MH., V. Jestin, D. Toquin, M Cherbonnel and N. 17. Mase, M, S. Yamaguchi, K. Tsukamoto, T. Imada, K. Imai and K
Eterradossi. Comparison of F-, G- and N-based RT-PCR protocols with Nakamura. Presence of avian pneumovirus subtypes A and B in Japan.
conventional virological procedures for the detection and typing of turkey Avian Dis. 47:481-484.2003.
rhinotracheitis virus. Arch Virol. 144:1091-1109. 1999. 18. McFarlane-Toms, I.P and R.J.H. Jackson. A comparison of three
4. Bennett, R.S., J. Nezworski, B.T. Velayudhan, K.V. Nagaraja, commercially available ELISA tests for detecting antibodies to turkey
D. H.Zeman, N.Dyer, T.Graham, D.C. Lauer, MK. Njenga and D.A. rhinotracheitis virus (TRTV). In: E. Kaleta and U. Heffels-Redman(Eds).
Halvorson. Evidence of avian pneumovirus spread beyond Minnesota Proceedings of the International Symposium on Infectious Bronchitis and
among wild and domestic birds in central North America. Avian Dis. Pneumovirus infections of Poultry. Rauischolzhausen, Germany, pp26-37.
48:902-908. 2004. 1998.
5. Chiang, S.J., A.M Dar, S.M Goyal, K.V. Nagaraja, D.A. Halvorson and 19. Mekkes, D. R. and J. J. de Wit. Comparison of three commercial ELISA
V. Kapur. A modified enzyme-linked immunosorbent assay for the detection kits for the detection of turkey rhinotracheitis virus antibodies. Avian Pathol.
of avian pneumovirus antibodies. J. Vet. Diagn. Invest. 12:381-384. 2000. 27: 301-305. 1999.
6. Collins, MS., R .E .Gough and D .J .Alexander. Antigenic differentiation 20. Morley, A.J. and D.K. Thomson. Swollen-head syndrome in broiler
of avian pneumovirus isolates using polyclonal antibody and mouse chickens. Avian Dis. 28:238-243. 1984.
monoclonal antibodies. Avian Pathol. 22:469-479. 1993. 21. Njenga, MK., H.M Lwamba and B.S. Seal. Metapneumoviruses in
7. Cooke, J.K.A., S. Kinloch and MM. Ellis. In vitro and in vivo studies in birds and humans. Virus Research. 91:163-169. 2003.
chickens and turkeys on strains of turkey rhinotracheitis virus isolated from 22.. Pedersen, J.C., L. Roland, D.L. Reynolds and A. Ali. The sensitivity
the two species. Avian Pathol. 22:157-170. 1993a. and specificity of a reverse transcriptase-polymerase chain reaction assay for
8. Cook, J. K. A., Β. V. Jones, Μ. M Ellis, Jing Li and D. Cavanagh. the βλάβη pneumovirus (Colorado strain). Avian Dis. 44:681-685. 2000.
Antigenic differentiation of strains of turkey rhinotracheitis virus using 23. Pedersen, J.C., D.A. Senne, B. Panigrahy and D.L. Reynolds. Detection
monoclonal antibodies. Avian Pathol. 22:257-273. 1993b. of avian pneumovirus in tissue and swab specimens from infected turkeys.
9. Cook, J.K.A., MB. Huggins, S.J. Orbell and D.A. Senne. Preliminary Avian Dis. 45:581-592. 2001
antigenic characterization of an avian pneumovirus isolated from 24. Pringle, C.R. Virus Taxonomy-San Diego. Arch.of Virology. 143:1449-
commercial turkeys in Colorado, USA. Avian Pathol. 28:607-617. 1999. 1459. 1998.
10. Cook, J.K.A. and D. Cavanagh. Detection and differentiation of avian 25. Senne, D.A., RK. Edson., J.C. Pedersen and B. Panigrahy. Avian
pneumoviruses (metapneumoviruses). Avian Pathol. 31:117-132. 2002. pneumoxdrus update. Proceedings of American Veterinary Medical
11. Droual, R. and P R. Woolcock. Swollen head syndrome associated with Association 134th Annual Congress. Reno, NV, USA. ppi90.1997.
E. Coli and infectious bronchitis virus in the Central Valley of California. 26. Shin, H.J., F.F. Jirjis, M.C. Kumar, MK. Njenga, D.P. Shaw, S.L. Noll,
Avian Pathol. 23:733-742. 1994. K.V.Nagaraja and D.A. Halvorsen. Neonatal avian pneumovirus infection in
12. Gough, R.E., MS. Collins, W.J. Cox and N.J. Chettle. Experimental commercial turkeys. Avian Dis. 46:239-244. 2002.
infection of turkeys, chickens, ducks, guinea fowl, pheasants and pigeons 27. Tanaka, M,H. Tanuma, N.Kokumai, E.Oishi, T. Obi, K. Hiramatsu and
with turkey rhinotracheitis virus. Vet. Record. 123:58-59. 1988. Y. Shimizu. Turkey rhinotracheitis virus isolated from broiler chicken with
13. Gough, R.E. Avian pneumoviruses. In: Diseases of Poultry, 11th ed. Y. swollen head syndrome in Japan. J. Vet. Med. Sci. 57:939-945.1995.
M Saif, H. J. Bames, J.R. Glisson, A M Fadly, L.R. McDougald and D.E. 28. Toquin, D., MH. Bayon-Auboyer, N. Etteradossi, H. Morin and V.
Swayne. Iowa State University Press, Ames, Iowa, pp 92-99. 2003. Jestin. Isolation of a pneumovirus from a Muscovy duck. Vet. Record.
14. Juhasz, K., and A. J. Easton. Extensive sequence variation in the 145:680
attachment (G) protein gene of avian pneumovirus: evidence for two distinct 29. Townsend, E., D.A. Halvorson, K.E.Nagaraja and D.P.Shaw.
subgroups. J. Gen. Virol. 75:2873-2880. 1994. Susceptibility of an avian pneumovirus isolated from Minnesota turkeys to
physical and chemical agents. Avian Dis. 44:336-342. 2000.
32
INFECTIOUS BRONCHITIS
Jack Gelb, Jr. and Mark W. Jackwood

SUMMARY. Infectious bronchitis (IB) is caused by infectious bronchitis virus (IBV), a member of the family Coronaviridae. IBV is highly
host specific, causing natural infections mainly in chickens. Primary infections in young chickens typically produce respiratory disease but
some strains may cause kidney lesions. Layers and breeders commonly suffer egg production losses with or without evidence of respiratory
disease signs. Numerous serotypes of the virus have been responsible for outbreaks in commercial chickens in spite of the use of attenuated
live and inactivated vaccines. IB is a growing problem for poultry producers because only a limited number of serotypes are available for
vaccination, and the cross-protection elicited against unrelated field strains may be minimal.
Agent Identification. Presumptive diagnosis is made in a suspect flock based on clinical signs consistent with the disease and evidence of
IBV serum antibody production as demonstrated by enzyme-linked immunosorbent assay (ELISA). Isolation and identification of the
causative serotype of IBV is required for definitive diagnosis. Serotype identification may be achieved by virus-neutralization,
hemagglutination-inhibition, type-specific monoclonal antibodies, or reverse transcriptase-polymerase chain reaction followed by restriction
fragment length polymorphism or sequencing. Cross-challenge tests in chickens may be used to determine the potential protection afforded
by immunization with various IBV vaccines or field strains to control the disease.
Serologic Detection in the Host. ELISA is preferred method for detecting serum antibody response, but it does not identify serotype­
specific antibodies. Acute and convalescent sera response is useful in sera diagnosis and for monitoring flock vaccination titers.

INTRODUCTION SAMPLE COLLECTION

Infectious bronchitis (IB) is a highly contagious clinically acute Samples for IBV isolation must be obtained as soon as clinical
disease of the respiratory and urogenital tract of chickens, caused by disease signs are evident. Tracheal swabs are preferred and are
infectious bronchitis virus (IBV), a member of the family placed directly into cold media with antibiotics to suppress bacterial
Coronaviridae. The disease is common throughout the world where and fungal growth and preserve the viability of the virus. Sterile
chickens are produced commercially. Mixed infections involving swabs are used to swab 5-10 clinically affected birds per flock.
IBV, Newcastle disease virus (NDV), avian adenovirus group 1, Cotton-tipped swabs of varying sizes * are available (Fisher
Mycoplasma, and coliform bacteria are common and may confuse Scientific, Pittsburgh, Penn.) depending on the age and breed of the
diagnostic efforts. chicken. The swabs are placed in 2-3 ml of cold sterile tryptose
Many serotypes of IBV are recognized and have practical phosphate broth (TPB) (pH 7.0-7.2) containing 10,000 IU/ml
significance in the control of IB, because immunity following penicillin, 10,000 IU/ml streptomycin, and 250 IU/ml amphotericin
infection or vaccination with one serotype often is not protective B. Swab tubes are immediately placed on ice and are frozen at -20
against infections with unrelated serotypes. In the United States, the C at the earliest convenience.
most often isolated IBV serotypes in commercial poultry are Other tissues, such as lung, kidney, oviduct, cecal tonsil and
Arkansas (Ark), Connecticut (Conn), Delaware (DE/072/92), and proventriculus may be collected using aseptic techniques. Tissues
Massachusetts (Mass). Previously unrecognized antigenic variants are placed in clean, labeled, tightly sealed plastic specimen bags or
have been recovered from multiage commercial layer complexes sterile tubes and are frozen at -20 C. Cloacal swabs may be obtained
with respiratory disease or egg production problems (10). Many and are handled as described above. All swabs and tissue samples
antigenic variant serotypes also have been reported in European should be frozen and transported in an insulated container to a
countries and Australia (7). Undoubtedly, many more strains will be diagnostic virology laboratory.
isolated from these and other countries as emphasis on IBV Because IBV persists in the intestinal tract, isolation of some
surveillance increases. For an in-depth review of IB, refer to strains from cecal tonsil and cloacal swabs is possible for several
Cavanagh and Naqi (4). A detailed discussion of IBV antigen, weeks after the disappearance of clinical signs. Accordingly,
genome and antibody detection assays prepared by De Wit (6) is isolating IBV from these sites does not confirm its role as the
also available. causative agent in a recent disease outbreak.
The frozen TPB containing tracheal and cloacal swabs is thawed,
CLINICAL DISEASE mixed, and incubated at room temperature (about 22 C) for 30-60
min prior to inoculation to reduce the possibility of bacterial and
Chickens are the primary natural host, although pheasants may be fungal contamination. Contamination sometimes associated with
susceptible to IBV or highly similar coronaviruses (3). In chickens a virus isolation from cloacal swabs may be avoided by centrifuging
short incubation period (24—72 hr) is a unique characteristic of the the swab tube broth at 1000 x g for 15 min and then passing the
disease. Young chicks display acute respiratory disease signs and supernatant fluid through a 0.22 or 0.45-μιη sterile syringe filter.
lesions in the trachea. Morbidity can approach 100%, but mortality Tissue homogenates (10% w/v) are prepared in TPB with
is generally below 5% in outbreaks not complicated by secondary antibiotics by disrupting lung, kidney, oviduct, or cecal tonsil using
pathogens or concurrent infections. In layer chickens, IBV causes a glass tissue grinder (Tenbroeck, VWR Scientific Products, West
decreased egg production and quality. Lymphoid cell infiltration Chester, Penn.) or mortar and pestle. The homogenates are
and epithelial cell degeneration of the oviduct wall have been incubated at room temperature for 30-60 min.
observed. Nephropathogenic strains such as Holte, Gray, and Sentinel chickens have been used to help facilitate IBV isolation in
Australian T produce enlarged kidneys with distended tubules and commercial flocks (9). Specific-pathogen-free (SPF) chickens
ureters containing uric acid crystals. Diarrhea, dehydration, immunized against IBV vaccine serotype(s) are placed with IBV-
depression, and death may occur in affected birds. susceptible sentinels in pens or cages in commercial chicken
houses. After a 1-wk exposure period, the field-exposed sentinels
are removed, and tracheal swabs are collected for virus isolation
attempts. Several successive weekly placements increase the
possibility of isolating IBV. The use of IBV-immune sentinels
146
Chapter 32 Infectious Brondnta

provides a better opportunity for isolating IBV antigenic variants DNA. Most strains of IBV are inactivated after 15 min at 56 C. IBV
free of contaminating vaccine virus serotypes that may be cycling in is inactivated by lipid solvents, such as chloroform and ether.
commercial chickens. Sensitivity to lipid solvents can also be used to screen LBV field
isolates for contaminating naked viruses, such as avian adenovirus
PREFERRED CULTURE MEDIA AND SUBSTRATES group 1 and reovirus.

Infectious bronchitis virus is most commonly grown in Serological Identification by Virus Neutralization (VN) and
embryonated eggs, tracheal organ culture (TOC), and chicken Hemagglutination-Inhibition (HI)
kidney cell culture. The embryonated egg is preferred for primary Virus Neutralization. Neutralization of an IBV field isolate by
isolation attempts. known serotype-specific IBV antiserum establishes its identity. The
VN test may be performed in embryonated eggs, chicken kidney
Embryonated Chicken Eggs cell culture, or TOC. The test may be conducted using the constant­
Embryonated chicken eggs from an SPF source are recommended serum diluted-virus (alpha) or diluted-serum constant-virus (beta)
for virus isolation. Ten 9 to 11-day-old embryos are inoculated by method. In addition, a constant-virus constant-serum VN procedure
the chorioallantoic sac route with 0.2 ml of swab tube broth or (5) has been useful for serotyping IBV field isolates. VN tests are
tissue homogenate. The eggs are candled daily, with mortality performed in embryonated eggs by reacting 32-320 mean embryo
between days 2 and 7 postinoculation (PI) considered to be virus­ infectious doses (ΕΠ)50) of an IBV field isolate with antisera to
specific. On day 3 PI, five eggs are removed from the incubator and known strains containing 1-20 units of antibody. The antibody unit
are placed at 4 C for 18-24 hr. Chorioallantoic fluid harvested concentration is determined by titration against the homologous
aseptically from inoculated eggs should be free of bacteria and IBV serotype. The highest dilution of antiserum protecting 50% of
fungi and give a negative hemagglutination (HA) reaction with the inoculated embryos is equal to one antibody unit. As concurrent
chicken red blood cells (CRBCs). The remaining embryos are infections with two or more IBV serotypes are common, reciprocal
incubated up to 7 days and are then observed for typical IBV VN tests are sometimes required to identify all IBV in field isolates.
lesions, such as stunting, curling, clubbing of the down, or urate Antiserum to the field isolate is reacted against known IBV
deposits in the mesonephros of the kidney. Cutaneous hemorrhage serotypes to establish the identities of the causative serotypes.
is often noted with embryo-lethal strains of IBV. Chorioallantoic Hemagglutination Inhibition. Hemagglutination antigen for the HI
membranes are collected, homogenized, and tested for avian test is prepared from chorioallantoic fluid harvested from IBV-
adenovirus group 1 by the immunodiffusion method. Group 1 inoculated embryonated eggs. Neuraminidase type V in PBS (pH
adenovirus infections of commercial chickens are common, and the 7.2) at 1.0 units/ml final concentration is used to treat IBV for 30
virus often produces stunted embryos indistinguishable from IBV- min at 37 C (24). HA antigen titration is performed in standard U-
infected embryos. bottom 96-well microtiter plates using CRBCs. Treatment of IBV
Some IBV field isolates are not embryo-adapted and do not cause with bacterial phospholipase C was initially thought to enable the
death or produce lesions on the first passage. Therefore, a minimum virus to agglutinate CRBCs (1,16). However, HA antigens produced
of three passages should be made before a virus isolation attempt is using highly purified phospholipase C preparations often had
considered to be negative. Particular attention should be given to considerably lower titers than those produced using unpurified
urate deposits in the mesonephros of the kidney in evaluating phospholipase C preparations. Subsequent studies (24,25)
embryos inoculated with field isolates. determined that treatment of IBV with purified neuraminidase
preparations consistently produced high-titer HA antigens. These
Organ Cultures findings suggested that the unpurified phospholipase C preparations
Chicken TOC may be used to isolate IBV. The primary advantage were contaminated with neuraminidase.
in using TOC over embryonated eggs is that non-embryo-adapted The microtiter HI test may be used to serotype IBV (1,16).
field strains of IBV produce a rapid ciliostasis in TOC on the first Neuraminidase-treated HA antigen is first prepared from the field
passage, eliminating the need for multiple passages. A disadvantage isolate. A concentration of 4-8 HA units of antigen is added to
is that a cell culture facility is needed for preparing and maintaining twofold dilutions (1:2 to 1:1024) of serotype-specific known IBV
TOC. Other potential limitations of TOC involve differentiating or antiserum. After incubation at room temperature for 30 min,
detecting viruses other than IBV in field samples. NDV and IBV CRBCs (0.5%-1.0%) are added, and the test is read 30-60 min
produce extensive ciliostasis by 3 days PI. Avian adenovirus later. The inhibition of HA of the field isolate by a known antiserum
replicates in TOC, but because it does not produce rapid and identifies the serotype of IBV but caution must be used when
complete ciliostasis, its presence may go undetected. interpreting this test because cross-reactivity is not uncommon.
Production of Serotyping Antiserum for VN and HL Antiserum
Cell Cultures suitable for serotyping IBV isolates using the VN or HI procedures
Primary isolation of IBV in chicken kidney cell culture is not is produced preferably in SPF chickens. Typically, 3-to-8-wk-old
recommended, because the virus requires adaptation in chickens are inoculated intratracheally with about 105 ELD50 of IBV
embryonated eggs before it will grow in cell culture. per bird. The chickens are placed in isolation rooms or Horsfall
isolation units. Two weeks PI, the chickens are reinoculated by
AGENT IDENTIFICATION intravenous injection with at least ΙΟ5 ΕΠ)50 of IBV. Blood samples
are collected from chickens 2 wk after intravenous injection of IBV.
Physicochemical Properties Serum is harvested, pooled, and inactivated at 56 C for 30 min
Coronaviruses are enveloped, single-stranded RNA viruses with before being used in VN or HI serotyping procedures.
helical symmetry (see Chapter 45 on virus identification and It is important for antisera to be highly specific for the
classification for characterization techniques). The virion is homologous IBV serotype and to not cross-react with heterologous
pleomorphic and has a diameter of 80-120 nm. The envelope virus serotypes. Repeated inoculations (three or more) or the use of
consists of large club-shaped spike proteins that give the surface of adjuvants in preparing serotyping antisera is not advisable because
the virion its characteristic corona appearance. Coronavirus antibodies may cross-react with heterologous IBV serotypes.
replication occurs in the cytoplasm. Inhibitors of DNA, such as the
halogenated nucleotide 5-iodo-2-deoxyuridine, do not inhibit IBV
multiplication, indicating that the virus contains RNA and not
147
Jack Gelb, Jr. and Mark W. Jackwood

Monoclonal Antibody Identification of IBV Antigens serotypes may produce a greater degree of protection than would be
Type-specific monoclonal antibodies specific to the SI subunit of predicted on the basis of serotyping results.
the spike glycoprotein of serotypes Mass, Conn, and Ark have been Cross-challenge tests are conducted by immunizing ten 3-to-6-wk-
produced (13) and used to identify the respective serotypes by old SPF chickens by eye drop with known IBV serotypes and
antigen-capture enzyme-linked immunosorbent assay (AC-ELISA) challenging them 4 wk later by eye drop inoculation with an
(23). Other serotypes may be identified as IBV but not identified to unknown field isolate. Appropriate controls are included in cross­
the specific serotype using a group-specific monoclonal to the M challenge tests. The immunized chickens challenged with the
glycoprotein of IBV (13, 23). Allantoic fluid from IBV-inoculated homologous strain must be protected. Nonimmunized control
embryonated chicken eggs is best suited for identification of IBV by chickens inoculated with the challenge virus must be susceptible.
AC-ELISA although tissue homogenates can be used. Isolation facilities are required to prevent inadvertent cross­
Immunoperoxidase staining (22) and immunofluorescent antibody infection between groups of chickens immunized with different
assays have also been used to identify serotypes using type-specific serotypes. Protection is evaluated most commonly by collecting
IBV monoclonal antibodies. tracheal swabs at 4 or 5 days after challenge and assessing them for
challenge virus by performing virus isolation attempts in
Molecular Identification embryonated eggs. Chickens from which virus is not isolated are
Several reverse transcriptase-polymerase chain reaction (RT-PCR) considered to be protected. Other methods for determining
assays have been developed for identifying IBV serotypes. The protection following challenge include evaluating microscopic
assays use serotype-associated sequence variations in the SI subunit lesions in the trachea or measuring ciliary activity in TOC explants
of the spike glycoprotein gene to identify and differentiate strains (20).
and variants. Antigenically novel field isolates should be used as challenge
Generally, allantoic fluids harvested following the inoculation of viruses following immunization of SPF chickens with vaccine(s)
eggs with clinical samples are used for RNA extraction and RT- containing the serotype(s) currently approved for use in the region
PCR amplification (2, 11, 12). A RT-PCR restriction fragment or country where the isolate was recovered. The results of these
length polymorphism (RFLP) assay has been developed that studies will establish the cross-protective potential of a vaccine(s)
differentiates IBV types based on unique electrophoresis banding against field isolate challenge.
patterns of restriction enzyme-digested fragments of the RT-PCR
amplified SI gene (12,18). The RT-PCR RFLP test can identify all SEROLOGIC IDENTIFICATION IN THE HOST
known serotypes of IBV as well as variant viruses. The RT-PCR
RFLP procedure may be used in conjunction with a RT-PCR assay Serodiagnosis of IBV infection in commercial chickens is best
that uses universal primers to the conserved membrane and performed by demonstrating an ascending serum antibody response
nucleocapsid genes (2). Although that assay does not identify virus in recovered chickens using the enzyme-linked immunosorbent
type, it does amplify all types of IBV and can be used in assay (ELISA). Commercially available ELISA kits detect
conjunction with a biotin-labeled DNA probe (11), or nucleic acid antibodies common to all IBV serotypes (IDEXX Laboratories,
sequencing to verify the presence of IBV in the sample Inc., Westbrook, Maine; Synbiotics Corp., San Diego, California)
SI genotype-specific RT-PCR can also be used to identify specific and are not capable of identifying serotype-specific antibodies
IBV serotypes (14). SI gene primers specific for serotypes Mass, induced by the IBV strain(s) responsible for the outbreak.
Conn, Ark, and JMK are used in conjunction with a universal Nonetheless, the ELISA is a very valuable, inexpensive, and easy to
primer set that amplifies all IBV serotypes. Specific primers for the use tool for serodiagnosis of IBV to rule out other common causes
DE/072/92 and California serotypes have also been developed. of respiratory disease such as NDV, infectious laryngotracheitis
Other IBV types can be detected using the general primers, but the virus, or Mycoplasma. Commercial IBV ELISA kits with computer-
specific serotype cannot be identified. Both RT-PCR RFLP and facilitated data analysis capabilities are available. ELISA systems
genotype specific RT-PCR are capable of detecting more than one using 96-well microtiter plates and a single-serum dilution approach
IBV type in a clinical sample. In addition, clinical samples are used to evaluate immune status in commercial flocks. Acute and
inactivated with an equal volume of buffered (pH 4.5) phenol or convalescent serum samples, preferably from the same chickens in a
spotted on Finders Technology Associates (FTA) cards (Whatman suspect IBV-infected flock, are tested by ELISA. It is recommended
Inc. Florham Park, NJ) can be used as a source of template for RT- that serum from acute samples be stored at -20 C until convalescent
PCR amplification (21). These inactivated samples can be safely samples are obtained. Both groups of sera can then be run at the
transported to the laboratory and with the proper permits they can same time to minimize variability. An increase in antibody titer
be imported from abroad. between acute and convalescent serum samples is indicative of an
Cycle sequencing of the RT-PCR amplified hypervariable amino IBV infection. Commercial ELISA systems for measuring IBV
terminus region of SI may be used to identify previously serum antibody have been compared with VN and HI serology and
recognized field isolates and variants (17,19). Comparison and found to give favorable results (26,27).
analysis of sequences of unknown field isolates and variants with The use of VN or HI tests for serodiagnosis is generally not
reference strains available in GenBank (National Center for recommended. These tests are expensive and tedious to perform on
Biotechnology Information http://www.ncbimlm.nih.gov/) can a routine basis. In addition, interpretation of the results may be
establish potential relatedness and is a significant advantage of difficult because sera of commercial chickens often contain cross­
sequencing. reacting antibodies resulting from multiple infections with
The major uses of RT-PCR tests are virus identification and its heterologous vaccinal and field strain serotypes (8). Antibodies
application in the understanding of epidemiological investigations produced by young chickens tend to be more serotype-specific,
during IBV outbreaks. The RT-PCR tests, as they now exist whereas cross-reactions with heterologous VN and HI antigens are
however, do not provide information on viral pathogenicity. common in breeders and layers (15).

Cross-Challenge Studies
Cross-challenge tests in chickens are an important adjunct to SI
genotyping or serotyping for antigenically characterizing an IBV
field isolate. Immunization of chickens with antigenically distinct

148
Chapter 32 Infectious Bronchitis

DIFFERENTIATION FROM CLOSELY RELATED AGENTS 14. Keeler, C. L., K. L. Reed, W. A. Nix, and J. Gelb. Serotype
identification of avian infectious bronchitis virus (IBV) by RT-PCR of the
Respiratory disease associated with IBV infections may be peplomer (S-l) gene. Avian Dis. 42:275-284. 1998.
15. King, D. J., and S. R. Hopkins. Evaluation of the hemagglutination­
clinically indistinguishable from mild respiratory forms of
inhibition test for measuring the response of chickens to avian infectious
Newcastle disease, infectious laryngotracheitis, and low pathogenic bronchitis virus vaccination. Avian Dis. 27:100-112. 1983.
avian influenza. In these cases, diagnosis depends on isolation and 16. King, D. J., and S. R. Hopkins. Rapid serotyping of infectious bronchitis
identification of the virus or demonstration of an increase in specific virus isolates with the hemagglutination-inhibition test. Avian Dis. 28:727—
antibody production associated with recovery from infection. 733. 1984.
Virulent strains of NDV and infectious laryngotracheitis virus 17. Kingham, B. F., C. L. Keeler, Jr., W. A. Nix, B. S. Ladman, and J. Gelb,
cause a more severe disease than occurs in IB outbreaks. Jr. Identification of avian infectious bronchitis virus by direct automated
Neurological signs or visceral lesions with high mortality are cycle sequencing of the S-lgene. Avian Dis. 44:325-335. 2000.
18. Kwon, Η. Μ, M W. Jackwood, and J. Gelb, Jr. Differentiation of
observed in flocks infected with virulent NDV. Infectious
infectious bronchitis virus serotypes using polymerase chain reaction and
laryngotracheitis virus can produce a hemorrhagic tracheitis with restriction fragment length polymorphism analysis. Avian Dis. 37:194-202.
high mortality in serious outbreaks. In addition, infectious 1993.
laryngotracheitis spreads more slowly in affected flocks. 19. Lee, C-W., D. A. Hilt, and M. W. Jackwood. Typing of field isolates of
Infectious coryza and mycoplasmosis may also resemble IB infectious bronchitis virus based on the sequence of the hypervariable region
complicated by pathogenic coliform bacteria. Swollen head in the SI gene. Veterinary Diagnostic Investigation. 15:344-348. 2003
syndrome is associated with IBV, Escherichia coli, and high 20. Marquardt, W. W., S. K. Kadavil, and D. B. Snyder. Comparison of
ammonia levels. Facial swelling is also observed in infectious ciliary activity and virus recovery from tracheas of chickens and humoral
immunity after inoculation with serotypes of avian infectious bronchitis
coryza and Mycoplasma infections.
virus. Avian Dis. 26:828-834. 1982.
Nephritis observed in chickens infected with nephropathogenic 21. Moscoso, Η., E. O. Raybon, S. G. Thayer, and C. L. Hofacre. Molecular
strains of IBV resembles kidney changes seen in several disease detection and serotyping of infectious bronchitis virus from FTA filter
conditions, such as infectious bursal disease, mycotoxicosis, or paper. Avian Dis. 49:24-29. 2005.
other toxicities. 22. Naqi, S. A monoclonal antibody-based immunoperoxidase procedure for
rapid detection of infectious bronchitis virus in infected tissues. Avian Dis.
REFERENCES 34:893-898. 1990.
23. Naqi, S. A., K. Karaca, and B. Bauman. A monoclonal antibody-based
1. Alexander, D. J., and N. J. Chettle. Procedures for the haemagglutination antigen capture enzyme-linked immunosorbent assay for identification of
and the haemagglutination inhibition tests for avian infectious bronchitis infectious bronchitis virus serotypes. Avian Pathol. 22:555-564. 1993.
virus. Avian Pathol. 6:9-17. 1977. 24. Ruano, M, J. El-Attrache and P. Villegas. A rapid-plate
2. Andreasen, J. R. Jr., M W. Jackwood, and D. A. Hilt. Polymerase chain hemagglutination assay for the detection of infectious bronchitis virus.
reaction amplification of the genome of infectious bronchitis virus. Avian Avian Dis. 44:99-104. 2000.
Dis. 35:216-220. 1991. 25. Shultze, B., D. Cavanagh, and G. Herrler. Neuraminidase treatment of
3. Cavanagh, D., P. Britton, R. E. Gough, K. Mawditt, D. de B. Welchman. avian infectious bronchitis coronavirus reveals a hemagglutinating activity
Coronaviruses from pheasants (Phasianus colchicus) are genetically closely that is dependent on sialic acid-containing receptors on erythrocytes.
related to coronaviruses of domestic fowl (infectious bronchitis virus) and Virology 189:792-794. 1992.
turkeys. Avian Pathol. 31:81-93. 2002. 26. Thayer, S. G., B. N. Nersessian, B. Rivetz, and O. J. Fletcher.
4. Cavanagh, D. and S. A. Naqi. Infectious bronchitis. In: Diseases of Comparison of serological tests for antibodies against Newcastle disease
poultry. 11“' ed. Saif, Y. Μ., H. J. Bames, J. R. Glisson, A. M. Fadly, L. R. virus and infectious bronchitis virus using ImmunoComb® solid-phase
McDougald, and D. E. Swayne. Iowa State University Press Ames, LA. 1 Ol­ immunoassay, a commercial enzyme-linked immunosorbent assay, and the
li 9. 2003. hemagglutination-inhibition assay. Arian Dis. 31:459-463. 1987.
5. Cowen, B. S., and S. B. Hitchner. Serotyping of avian infectious 27. Thayer, S. G., P. Villegas, and O. J. Fletcher. Comparison of two
bronchitis viruses by the virus-neutralization test. Avian Dis. 19:583-595. commercial enzyme-linked immunosorbent assays and conventional
1975. methods for avian serology. Arian Dis. 31:120-124. 1987.
6. De Wit, J.J. Technical review. Detection of infectious bronchitis virus.
Avian Pathol. 29:71-93. 2000.
7. Gelb, J., Jr., C. L. Keeler, Jr., W. A Nix, J. K. Rosenberger, and S. S.
Cloud. Antigenic and S-l genomic characterization of the Delaware variant
serotype of infectious bronchitis virus. Avian Dis. 41:661-669. 1997.
8. Gelb, J., Jr., and S. L. Killian. Serum antibody responses of chickens
following sequential inoculations with different infectious bronchitis virus
serotypes. Avian Dis. 31:513-522. 1987.
9. Gelb, J., Jr., J. K. Rosenberger, P. A. Fries, S. S. Cloud, E. M Odor, J. E.
Dohms, and J. S. Jaeger. Protection afforded infectious bronchitis virus-
vaccinated sentinel chickens raised in a commercial environment. Avian
Dis. 33:764-769. 1989.
10. Gelb, J., Jr., J. B. Wolff, and C. A. Moran. Variant serotypes of
infectious bronchitis virus isolated from commercial layer and broiler
chickens. Avian Dis. 35:82-87. 1991.
11. Jackwood, M W., Η. M Kwon, and D. A. Hilt. Infectious bronchitis
virus detection in allantoic fluid using the polymerase chain reaction and a
DNA probe. Avian Dis. 36:403-409. 1992.
12. Jackwood, M W., N. Μ H. Yousef, and D. A. Hilt. Further
development and use of a molecular serotype identification test for
infectious bronchitis virus. Avian Dis. 41:105-110. 1997.
13. Karaca, K., S. Naqi, and J. Gelb, Jr. Production and characterization of
monoclonal antibodies to three infectious bronchitis virus serotypes. Avian
Dis. 36:903-915. 1992.

149
33
TURKEY CORONAVIRUS
Mark W. Jackwood and James S. Guy

SUMMARY. Turkey coronavirus (TCoV) is the cause of coronaviral enteritis in turkeys. TCoV produces an economically significant
enteric disease in commercial turkeys, and has been associated with poult enteritis and mortality syndrome (PEMS). Young turkeys are
affected and diarrhea is the most common clinical sign.
Agent Identification. Virus can be isolated from fecal droppings, cloacal swabs, intestinal contents, intestines and the bursa of Fabricius.
The virus is propagated in embryonating eggs and identified by electron microscopy, fluorescent antibody tests, immunohistochemistry,
antigen capture enzyme linked immunosorbent assay (ELISA), or reverse transcriptase-polymerase chain reaction (RT-PCR).
Serologic Detection in the Host Antibodies against TCoV can be detected by the indirect fluorescent antibody test, commercial ELISA for
infectious bronchitis virus (IBV) or competitive ELISA. TCoV is closely related to, and should be differentiated from IBV as well as a
number of other viruses that cause enteric disease in turkeys.

INTRODUCTION affected. Characteristic microscopic lesions of the intestinal tract


are an increase in crypt depth, a decrease in villous length, a
Turkey poult enteritis is a severe and extremely important disease decrease in intestinal diameter, cuboidal epithelium with a loss of
in commercial turkeys because it can lead to enormous economic microvili, and an increase in heterophils and lymphocytes in the
losses. An enveloped positive-stranded RNA virus designated lamina propria (13). In the bursa of Fabricius, characteristic
turkey coronavirus (TCoV) is the etiologic agent of turkey microscopic lesions consist of epithelial necrosis with replacement
coronaviral enteritis. The original TCoV described in Minnesota in of the normal pseudostratified columnar epithelium with a stratified
1951 (TCoV [Minnesota]), which causes bluecomb disease, initially squamous epithelium (13).
was believed to be similar to bovine coronavirus, a group Π
coronavirus (9, 22, 24). However, recent studies have shown that SAMPLE COLLECTION
TCoV (Minnesota) and other TCoV isolates associated with
enteritis and PEMS in turkeys are antigenically and genomically Samples for TCoV isolation include fecal droppings, cloacal
similar to each other and to a group ΙΠ coronavirus, infectious swabs, intestinal contents, intestines, or bursa of Fabricius. Because
bronchitis virus (IBV) (3,10, 11). coronaviruses are extremely labile, only fresh droppings should be
In 1997, Guy et al. (10) reported that a TCoV isolate from North collected and the samples should be kept cold (on ice at 4 C or
Carolina turkeys (NC95) was similar to other TCoV isolates as well frozen) at all times. Cold phosphate buffered saline (PBS) pH 7.4 or
as TCoV (Minnesota), and these viruses likewise were similar to minimal essential medium should be added to the clinical samples
IBV. Their conclusions were based on cross reactivity of polyclonal and mixed well or homogenized if the samples are tissues. Next the
antibodies in the fluorescent antibody test and immunoperoxidase samples are clarified by centrifugation, and filtered through a
procedures. In addition, they found that a monoclonal antibody 0.45 pm filter and stored frozen, preferably at -80 C.
directed against the membrane protein of IBV reacted with the
TCoV. Since that report, several other studies conducted on TCoV PREFERRED CULTURE MEDIA AND SUBSTRATES
isolates, showed that the order of the genes at the 3’ end of the
genome was similar to IBV (1, 3, 15, 16, 21) reinforcing the fact The preferred laboratoiy host system is specific-pathogen-free
that IBV and TCoV are closely related. (SPF) embryonating turkey eggs between 15 and 21 days of
The most important structural protein in coronaviruses is the spike incubation. Commercial eggs can be used as long as they do not
glycoprotein. The S glycoprotein forms club shaped projections on contain TCoV specific antibodies. Chicken embryonating eggs
the surface of the virus particles. It is anchored in the envelope of between 16 and 18 days of age can also be used to propagate the
the virus and consists of two subunits designated SI and S2. Spike virus. Eggs are inoculated via the allantoic cavity and virus is
mediates virus attachment to the host cell and is for the most part collected from the gastrointestinal tract at 48 to 72 hr post­
responsible for host cell specificity. In addition neutralizing inoculation.
antibodies are directed against the SI subunit of spike (2). Attempts to grow TCoV in cell culture have been unsuccessful.
Published sequence data for the TCoV spike gene (GenBank Acc. The Bluecomb virus, TCoV (Minnesota), was reported to grow in
Nos. AY342356 and AY342357) shows that TCoV spike and IBV HRT-18 cells, a human rectal adenocarcinoma cell line (7);
spike are clearly different (less than 23% amino acid similarity). however, recent studies have failed to verify this (10).
The TCoV spike sequence was also important in establishing that
different isolates of TCoV appear to be genetically similar (greater AGENT IDENTIFICATION
than 90% similarity).
Electron microscopy. The virus can be identified by negative
CLINICAL DISEASE contrast electron microscopy or immune-electron microscopy. The
characteristic 60 to 180nm pleomorphic, enveloped particles with
Young turkeys less than 4 wk of age are most susceptible to the club-shaped projections (spikes) can be observed in intestinal
disease and clinical signs are usually observed from 2 to 3 days contents, intestines, and bursa of Fabricius. However, virus particles
post-infection (14). Clinical signs can occur suddenly and consist of can be difficult to identify in “dirty samples” and it is recommend
watery, frothy droppings that may contain mucus and urates (13). that the source of virus be from the embryonic gastrointestinal tract
Birds often show depression, anorexia, dehydration, and weight following propagation of the virus in embryonating eggs.
loss. Morbidity is near 100% and mortality can be variable Fluorescent antibody test and immunohistochemistry. Direct
depending on the age of the bird, secondary pathogens, and indirect fluorescent antibody (FA) tests have been described for
management, and environmental conditions (13). Lesions associated detection of TCoV in intestines, bursa of Fabricius, and embryo-
with the disease are pale, thin-walled and flaccid intestines, with infected gastrointestinal tissues (4, 18). Frozen tissue sections are
frothy watery contents (14). Most all of the intestinal tract can be fixed in ice cold acetone for 10 min and stored at 4 C or
affected but usually the lower gut, including the ceca are commonly immediately stained with anti-TCoV antibodies directly conjugated
150
Chapter 33 Turkey Coronavirus

with fluorescein isothiocyanate (FITC) for direct FA or for indirect SEROLOGIC DETECTION IN THE HOST
FA, anti-TCoV antibodies are detected with the appropriate
secondary anti-gammaglobulin conjugated with FITC. Direct and The indirect fluorescent antibody test (IFAT) has been established
indirect FA tests have been shown to be useful for detection of viral to detect antibodies to TCoV (5, 18). Antigen for this test consists
antigens in tissues and antibodies in serum, respectively (18). The either of frozen sections of TCoV-infected embryo intestines or
direct FA test, using FITC-conjugated turkey anti-TCoV antibodies, epithelium exfoliated from bursa of Fabricius of infected turkeys
was found to be suitable for detection of viral antigens during the (13). Frozen tissue sections are prepared from intestinal tissues of
acute stages of the disease, whereas the indirect FA test was useful TCoV-infected turkey embiyos, 24-48 hr after inoculation with
for detection of anti-TCV antibodies, and for monitoring flocks for embryo-adapted TCoV strains. Bursa of Fabricius epithelial cells
exposure to TCoV (18). It was reported that TCoV can be detected from experimentally infected turkeys are spotted onto microscope
from 1 to 35 days post exposure when monoclonal antibodies are slides, air-dried, and fixed in ice-cold acetone. Dilutions of sera are
used in the FA test (4). The sensitivity of that test was 69% and the prepared in PBS and applied to the slides. Following incubation, the
specificity was 96% when compared to virus isolation. slides are washed in PBS and incubated with FITC conjugated anti­
Immunohistochemical procedures (indirect FA and indirect turkey IgG (H&L, Kirkegaard & Perry Laboratories, Gaithersburg,
immunoperoxidase procedures) also were used to detect the virus in MD). The slides are washed and examined for fluorescence with a
frozen tissue sections prepared from intestinal tissues and bursa of microscope containing an ultraviolet light source.
Fabricius (4). Frozen tissue sections were fixed in ice-cold acetone Serodiagnosis of TCoV can be demonstrated by ascending serum
and stored at 4 C prior to immunological staining. Indirect FA antibody titers using the ELISA test. A commercially available
utilized TCoV-specific monoclonal antibodies and FITC-conjugated ELISA test for IBV (IDEXX, Westbrook, Maine) cross-reacts and
anti-mouse IgG. The indirect immunoperoxidase procedure utilized can be used to detect TCoV serum antibodies in turkeys when a
TCoV-specific monoclonal antibodies and a commercially available conjugated secondary antibody against turkey immunoglobulin
avidin-streptavidin immunoperoxidase kit. The virus was detected (goat anti-turkey IgG H&L, Kirkegaard & Perry Laboratories,
from 1 to 35 days post exposure, and the sensitivity of the test was Gaithersburg, MD) is used (17). A competitive ELISA test was also
61%, whereas specificity was 96% when compared to virus reported. That test utilizes a recombinant baculovirus expressed
isolation. TCoV nucleocapsid protein and a biotin-labeled monoclonal
Antigen capture enzyme linked immunosorbent assay. Using antibody against the TCoV nucleocapsid protein (12). The ELISA
TCoV-specific antisera prepared in rabbits and guinea pigs, a tests are reported to detect TCoV antibodies beginning on day 10
double-antibody ELISA was developed to detect TCoV in intestinal through day 28 post exposure in experimentally infected turkeys
contents (8). The test was found to be more sensitive than electron (12), and both tests cross-react with IBV antibodies.
microscopy for detection of TCoV. Additionally, the test was not
specific for TCoV as IBV was also detected. Briefly, TCoV-specific DIFFERENTIATION FROM CLOSELY RELATED AGENTS
antibody is coated onto a 96 well ELISA plate and intestinal
contents from turkey poults with diarrhea are added to the well. Differentiation of TCoV from IBV infection is important because
After incubation, the plate is washed with PBS and secondary of the close relationship between those two viruses. TCoV has been
antibody specific for TCoV conjugated to peroxidase is applied. detected for up to 14 days post exposure in the gastrointestinal tract
Following incubation, the plate is again washed and the appropriate of experimentally infected chickens by RT-PCR (14). IBV,
substrate applied to detect the virus. however, has a strict host range infecting only chickens and
Reverse transcriptase-polymerase chain reaction. Several RT- pheasants (6). In addition, TCoV has a tropism for intestinal and
PCR tests have been developed to detect TCoV in intestinal bursa of Fabricius epithelium of turkeys, whereas IBV typically
contents, fecal droppings, and tissues. All of the tests reported to infects the upper respiratory tract, kidneys and reproductive tract of
date cross-react with IBV. Breslin et al. (4) reported an RT-PCR chickens (11).
test for TCoV that amplifies a 1100 bp region spanning the matrix Other causes of turkey enteric disease include a number of viruses
and nucleocapsid genes. The sensitivity of the test was 160 ΕΠ)50 including turkey astrovirus, reovirus, rotavirus, and enterovirus, as
and the identity of the amplified product was confirmed by nucleic well as bacteria, protozoa and fungi. Differentiation of TCoV from
acid sequencing. Velayudhan et al. (23) used 3 sets of primers those infectious agents can be accomplished using the tests
designed to amplify the polymerase gene and the nucleocapsid gene described above.
of TCoV. The sensitivity was between 1 and 10 ng/ml of RNA and
cross-reactions with IBV were identified with another set of primers REFERENCES
specific for the spike gene of IBV. Finally Sellers et al. (19)
developed a multiplex RT-PCR test that amplifies the nucleocapsid 1. Akin, A., T. L. Lin, C. C. Wu, T. A. Bryan, T. Hooper and D. Schrader.
Nucleocapsid protein gene sequence analysis reveals close genomic
gene of TCoV as well as IBV, and Spackman et al. (20) developed
relationship between turkey coronavirus and avian infectious bronchitis
a real-time RT-PCR test that amplifies the matrix protein gene of virus. Acta Virol. 45:31-38. 2001.
both TCoV and IBV. Sensitivity of the multiplex and real-time tests 2. Boursnell, Μ, Μ M Binns, T. Brown, D. Cavanagh and F. M Tomley.
was from 1-5 ng of RNA and 1100 gene copy numbers respectively. Molecular biology of avian infectious bronchitis virus. Karger,New York
Generally, the RT-PCR test can detect TCoV between 1 and 14 0:65-82. 1989.
days post exposure in cloacal swabs and intestinal tissues with 3. Breslin, J. J., L. G. Smith, F. J. Fuller and J. S. Guy. Sequence analysis of
cloacal swabs providing more consistent results. the turkey coronavirus nucleocapsid protein gene and 3' untranslated region
Procedures for extraction of viral RNA vary but the most widely identifies the virus as a close relative of infectious bronchitis virus. Virus
Res. 65:187-193. 1999.
used is Trizol LS reagent (Invitrogen, Inc., Carlsbad, CA); this
4. Breslin, J. J., L. G. Smith, H. J. Bames and J. S. Guy. Comparison of
reagent is used according to the manufacturer’s instructions. The virus isolation, immunohistochemistry, and reverse transcriptase-polymerase
RT-PCR reaction conditions vary with each set of primers but in chain reaction procedures for detection of turkey coronavirus. Avian Dis.
general follow the recommendations of the RT-PCR kit 44:624-631. 2000.
manufacturers and consist of a 30 to 60 min reverse transcription 5. Breslin, J. J., L. G. Smith and J. S. Guy. Baculovirus expression of turkey
step followed by heat denaturation for 5 min and 35 to 40 PCR coronavirus nucleocapsid protein. Avian Dis. 45:136-143. 2001.
cycles. The PCR products are observed on an agarose gel following 6. Cavanagh, D. and S. A. Naqi. Infectious bronchitis. In: Diseases of
electrophoresis or are sequenced. The real-time RT-PCR products poultry. 11th Saif, Y. Μ, H J. Bames, J. R. Glisson, A. M Fadly, L. R.
McDougald, and D. E. Swayne eds. Iowa State University Press Ames, IA.
were detected with a specific probe labeled with a fluorescent dye.
101-119. 2003.
151
Mark W. Jackwood and James S. Guy

7. Dea, S., S. Garzon and P. Tijssen. Isolation and trypsin-enhanced 17. Loa, C. C., T. L. Lin, C. C. Wu, T. A. Bryan, H. L. Thacker, T. Hooper
propagation of turkey enteric (bluecomb) coronaviruses in a continuous and D. Schrader. Detection of antibody to turkey coronavirus by antibody­
human rectal adenocarcinoma cell line. Am J. Vet. Res. 50:1310-1318. capture enzyme-linked immunosorbent assay utilizing infectious bronchitis
1989. virus antigen. Avian Dis. 44:498-506. 2000.
8. Dea, S. and P. Tijssen. Detection of turkey enteric coronavirus by 18. Patel, B. L., E. Gonder and B. S. Pomeroy. Detection of turkey
enzyme-linked immunosorbent assay and differentiation from other coronaviral enteritis (bluecomb) in field epiomithics, using the direct and
coronaviruses. Am. J. Vet. Res. 50:226-231. 1989. indirect fluorescent antibody tests. Am. J. Vet. Res. 38:1407-1411. 1977.
9. Dea, S., A. J. Verbeek and P. Tijssen. Antigenic and genomic 19. Sellers, H. S., M D. Koci, E. Linnemann, L. A. Kelley and S. Schultz-
relationships among turkey and bovine enteric coronaviruses. J. Virol. Cherry. Development of a multiplex reverse transcription-polymerase chain
64:3112-3118. 1990. reaction diagnostic test specific for turkey astrovirus and coronavirus. Avian
10. Guy, J. S., H. J. Bames, L. G. Smith and J. Breslin. Antigenic Dis. 48:531-539. 2004.
characterization of a turkey coronavirus identified in poult enteritis- and 20. Spackman, E., D. Kapczynski and H. Sellers. Multiplex real-time
mortality syndrome-affected turkeys. Avian Dis. 41:583-590. 1997. reverse transcription-polymerase chain reaction for the detection of three
11. Guy, J. S. Turkey coronavirus is more closely related to avian infectious viruses associated with poult enteritis complex: Turkey astrovirus, turkey
bronchitis virus than to mammalian coronaviruses: A review. Avian coronavirus, and turkey reovirus. Avian Dis. 49:86-91. 2005.
Pathology 29:207-212. 2000. 21. Stephensen, C. B., D. B. Casebolt and N. N. Gangopadhyay.
12. Guy, J. S., L. G. Smith, J. J. Breslin and S. Pakpinyo. Development of a Phylogenetic analysis of a highly conserved region of the polymerase gene
competitive enzyme-linked immunosorbent assay for detection of turkey from 11 coronaviruses and development of a consensus polymerase chain
coronavirus antibodies. Avian Dis. 46:334-341. 2002. reaction assay. Virus Res. 60:181-189. 1999.
13. Guy, J. S. Turkey coronavirus enteritis. In: Diseases of poultry. 11th ed. 22. Tijssen, P., A. J. Verbeek and S. Dea. Evidence of close relatedness
Saif, Y. M, Bames, H. J., Glisson, J. R, Fadly, A. M, McDougald, Swayne, between turkey and bovine coronaviruses. Adv. Exp. Med. Biol. 276:457-
D. E. eds. Iowa State Press Ames, Iowa. 300-307. 2003. 460. 1990.
14. Ismail, Μ M., A. Y. Tang and Y. M. Saif. Pathogenicity of turkey 23. Velayudhan, B. T., H. J. Shin, V. C. Lopes, T. Hooper, D. A. Halvorson
coronavirus in turkeys and chickens. Avian Dis. 47:515-522. 2003. and K. V. Nagaraja. A reverse transcriptase-polymerase chain reaction assay
15. Lin, T. L., C. C. Loa and C. C. Wu. Existence of gene 5 indicates close for the diagnosis of turkey coronavirus infection. J. Vet. Diagn. Invest.
genomic relationship of turkey coronavirus to infectious bronchitis virus. 15:592-596. 2003.
Acta Virol. 46:107-116. 2002. 24. Verbeek, A., S. Dea and P. Tijssen. Genomic relationship between
16. Lin, T. L., C. C. Loa and C. C. Wu. Complete sequences of 3' end turkey and bovine enteric coronaviruses identified by hybridization with
coding region for structural protein genes of turkey coronavirus. Virus Res. BCV or TCV specific cDNA probes. Arch Virol. 121:199-211. 1991.
106:61-70. 2004.

152
34
ENTERIC VIRUSES
Don Reynolds and Ching Ching Wu

SUMMARY. With the exception of hemorrhagic enteritis virus (covered elsewhere) enteric viruses of poultry occur primarily in young
poultry. Characteristic clinical signs include diarrhea, anorexia, litter eating, ruffled feathers and poor growth. Intestines may have lesions;
intestines are typically dilated and are filled with fluid and gaseous contents. The sequela to clinical disease is often stunting and / or ranting
of the birds resulting in uneven flocks. Various viral agents (or combinations of these agents) can cause enteric disease and / or have been
identified from the intestinal contents of birds inflicted with enteric disease. Those viruses that cause and / or are associated with enteric
diseases of young poultry include astrovirases, coronaviruses, enteroviruses, rotaviruses and torovirus.
Agent Identification. A number of techniques have been used to detect enteric viruses. Many of the enteric viruses were first identified by
the direct visualization of the virion using electron microscopy (EM). Techniques that employ EM still remain the method of choice (or may
be the only method available) for detecting many of the enteric viruses (31). Immune electron microscopy (IEM) has been advantageous for
identifying many enteric viruses, especially those viruses for which physicochemical characteristics are little known. The expense,
availability, and experience of EM operator have limited the use of EM for the routine diagnosis of enteric viruses. Other techniques such as
fluorescent antibody (FA), genome electropherotyping, enzyme-linked immunosorbent assay (ELISA), and hemagglutination, have also been
used for the detection of some of the enteric viruses. Advances in molecular biology and genomic sequencing have allowed for the reverse
transcriptase polymerase chain reaction (RT-PCR) to be developed and used effectively for detecting some of the enteric viruses.
Serologic Detection in the Host. Detection of antibodies is used for the diagnosis of certain enteric virus infection (VN, ELISA).

INTRODUCTION SAMPLE COLLECTION

Enteric diseases that occur in young birds (less than 6 wk of age) Intestinal samples collected from birds experiencing clinical
are problematic for poultry producers. A number of viruses have disease (diarrhea, etc.) are of greatest benefit for isolating and
been associated with enteric disease of young birds (35). Some of identifying most enteric viruses. The intestinal tract should be
these viruses are enteropathogens. However, the role that other viruses removed from the bird in its entirety starting from the point where the
play in enteric disease has yet to be determined. For example, in duodenum joins the ventriculus and continuing distally to the
turkeys a disease once known as ’’bluecomb disease” is caused by a cloaca (vent); that is, the entire intestinal tract distal to the
coronavirus. However, a similar condition in turkeys known as "poult ventriculus. The number of tracts needed depends upon the age
enteritis" or "turkey viral enteritis" has been associated with other and size of the birds involved. Typically, intestinal tracts
viruses including rotavirus and astrovirus infections. A similar from five to 10 birds are adequate. Ihe intestinal tracts can be placed
condition in chickens that has been commonly referred to as in any container free from viral contaminants. A sterile plastic bag
"malabsorption syndrome," "stunting syndrome," etc., has been such as a Whirl Pac® (Nasco, Fort Atkinson, WI) is convenient.
associated with numerous viral agents, including reoviruses and The intestinal tract should be cut into small sections of 2-5 cm in length
enteroviruses. Within the past decade, poult enteritis and mortality when placing it into the sterile container (Whirl Pac®). This allows
syndrome (PEMS) has been determined to be an infectious disease the samples to be processed easier and aids obtaining intestinal
perhaps involving enteric viruses. Those enteric viruses that have been contents. The intestinal samples can then be stored and/or shipped by
associated with PEMS include coronaviruses and astrovirases (4). freezing at temperatures of -20 C or lower. Intestinal contents are an
important source of virus-infected material. Although it is not
CLINICAL DISEASE necessary to ligate the intestinal tract, no attempt should be made to
empty the intestinal tract of its contents. Intestinal tracts that have
The clinical disease varies with respect to the severity of clinical signs been fixed in formalin or other fixatives prove to be of limited value,
and symptoms depending on the agent(s) and the avian species because fixation usually results in inactivation of viruses.
involved. However, there are a number of characteristics that are Once intestinal tracts are frozen, they can be stored for indefinite
common among all the enteric virus infections. The clinical disease periods of time, as freezing appears to have minimal effects on most
most commonly occurs within the second or third week of life and lasts enteric viruses. Samples can be removed from the freezer at any
10 to 14 days. Therefore, most birds that contract enteric viral disease convenient time for further processing. Processing the intestinal
recover from the clinical disease by 6 wk of age. Characteristic samples for further virus isolation and identification involves the
clinical signs and symptoms most often include diarrhea (watery following steps:
droppings), anorexia, litter eating, listlessness, and various types 1) Thaw intestinal samples slowly, preferably at 4 C. In some
of enteritis. The intestinal tracts are often filled with watery contents, instances (nonenveloped viruses) freeze-thawing multiple times aids
gas, and, in some instances, partially digested feed. The ceca are in rupturing intestinal cells that harbor virus and breaks down tissue
usually dilated and filled with gaseous, frothy contents. Mortality is for easier handling of the sample.
variable and dependent upon the agent(s) and avian species involved. 2) Dilute the samples with sterile phosphate-buffered saline (PBS;
Generally, mortality is only slightly to moderately increased. pH 7.4) to a workable solution. Other diluents can be used;
However, with coronavirus infections of turkeys, losses of 50% or however, PBS is economical and convenient. The extent of dilution
more have been reported especially in PEMS cases (4, 11). necessary depends on the sample. If the sample consists of watery
Morbidity, as a result of decreased growth (stunting), is of diarrhea, it will take less dilution than a sample consisting of solid
primary concern. Typically, 5% to 20% of a flock that has fecal material. Typically, a dilution of 1:5 to 1:10 (sample:
experienced enteric viral disease becomes stunted and remains so diluent) is adequate.
throughout the growout period. 3) Homogenize the sample. This may be accomplished in a
number of ways. It is most convenient to place the plastic bags
containing the sample and diluent in a stomacher for 3 min and
allow the stomacher to homogenize the sample. By this method, the
sample does not leave its original container (Whirl Pac®), thus
minimizing the risk of contaminating the sample, laboratory, and
personnel. Numerous samples may be done quickly without the
153
Don Reynolds and Ching Ching Wu

necessity of cleaning equipment, etc. Other methods to homogenize particulate matter. The supernatant is saved and used for further
tissue such as using tissue grinders, blenders, or digitally processing. The pellet should not be discarded, because it may
expressing the tracts by hand can be as effective. Another convenient retain appreciable amounts of virus.
method is to “roll” the samples contained within a plastic bag with a 6) The supernatant is filtered through a 0.45 pm filter. Usually, it is
small wall paper roller that can be obtained at a local home advisable to filter the supernatant through larger filters (such as
improvement store. 1.2 pm, 0.8 pm or 0.65 pm) before attempting to filter through
4) Sonicate samples in an ice bath for at least 1.5 min. Sonication a 0.45 pm filter. Positive pressure syringe filters are useful for
should be accomplished by 30-sec intervals of sonication followed this procedure. If the samples are difficult to filter, the
by 30-sec intervals of rest (no sonication) to allow heat dispersion. clarification step (see step 5 above) can be repeated increasing the
Sonication aids in liberating viruses from tissue and organic centrifugation force up to 5,000 x g. The sonicated filtrate can be
material. It also aids in dispersing aggregates of viruses. One frozen and stored. Repeated freezing and thawing may have some
must be careful when using this step, because sonication may unwanted effect on some viruses. Therefore, it is advisable to
affect membraned viruses. Additionally, sonication detaches pili aliquot this material in several containers before freezing. The
from bacteria that are present in the sample. Depending on the collection of additional tissues or the use of other procedures may
intended use of the sample, the pili may pose potential be of greater benefit depending upon the specific agent being
problems. Therefore, one may elect to omit sonication or sought.
sonicate after bacteria have been removed.
5) Clarify the sample by subjecting it to centrifugation
between 400 and 500 x g. This clarifies the sample of most of the

Astroviruses

PREFERRED CULTURE MEDIA AND SUBSTRATES Reverse transcription-polymerase chain reaction (RT-PCR)
assay
Chicken astroviruses have been reported to have been A RT-PCR assay was described in 2000 by Koci et al (19, 20).
propagated in chick embryo liver cells (CEL, see Chapter 43 on PCR primers were designed to amplify two fragments, an 802 bp
primary cell culture for procedures). Briefly, CEL cells were grown fragment of viral polymerase gene amplified by primers MKCap8
to near confluency in 6mm petri dishes (5). The inocula (0.2 ml (TCATCATCCTCTCACACTGG) and MKCapl9
sonicated filtrate, as indicated above) can be adsorbed onto the (AGCAGCAGTAGGTGGCAGTG) and 849 bp fragment of viral
monolayer with gentle rocking motions followed by the addition of capsid gene amplified by primers MKPollO
media. Cytopathic effect (CPE) was observed after 6 days of (TGGCGGCGAACTCCTCAACA) and MKPolll
incubation at 5% CO2 and 38.5 C. (AATAAGGTCTGCACAGGTCG). Total RNA isolated from the
intestines and/or feces (experimentally infected or commercial
AGENT IDENTIFICATION turkeys with an acute enteric disease) using the TRIzol total RNA
isolation reagent (Life Technologies, Rockville, MD) were
Astroviruses are small round, non-enveloped viruses that typically subjected to reverse transcription to cDNA using SuperScript
measure 28 to 30 nm in diameter (5) (with a positive-stranded RNA Reverse Transcriptase (Life Technologies). The cDNA was used as
genome). They have characteristic five-pointed or six-pointed star- target in the PCR amplification using the above-mentioned primers.
like surface projections detected by negatively stained electron The PCR products were electrophoresised in a 1.1% agarose gel in
microscopy (EM). The family Astrovirdidae is divided into 2 TAE buffer, stained with ethidium bromide, and visualized by UV
genera: mammalian and avian astroviruses. light. This RT-PCR assay has specificity for TAstV.
Avian astroviruses have been associated with enteric diseases in
turkeys causing diarrhea and high mortality. Astroviruses were first Multiplex RT-PCR
reported in turkeys of age 6-11 days. Two types of turkey A multiplex RT-PCR was described for the detection of turkey
astroviruses (TAstV) have been identified TAstV-1 and TAstV-2. astrovirus and turkey coronavirus TCoV (37). Intestine and feces
These two types differ immunologically and genetically from the from commercial turkey flocks and from turkey embryo intestines
other avian astroviruses, avian nephritis virus, duck virus hepatitis inoculated with TCoV orTAstV-2 were used to develop the assay.
type 2 and chicken astrovirus (34). PCR primers (TCVnucleo forward, GGTAGCGGTGTTCCTGA,
The complete genomic sequence of TAstV was reported by Koci and TCVnucleo reverse, CCCTCCTTACCTTTAGT) for the
et al (18, 20). The genome is 7,325 nucleotides and contains three detection of TCoV were designed to amplify a 598 bp fragment
ORFs; ORF la, ORF lb and ORF2, and there is a frame shift within the nucleocapsid gene of TCoV. PCR primers for TAstV-2
between ORF la and ORF lb. The protein products for ORF la, and were designed to amplify the 802 bp fragment of viral polymerase
ORF lb are unknown as yet, but are predicted to be non-structural gene as mentioned above. The amplified TCoV nucleocapsid gene
proteins. It is possible that the product of ORFa is a serine protease sequence is conserved with nucleocapsid gene sequence of
and the product of ORF lb is a RNA-dependent RNA polymerase infectious bronchitis virus (IBV). This assay was specific for
(5, 20). ORF2 encodes the viral capsid protein of 73-80 kDa. TAstV-2, TCoV, and IBV. The sensitivity or the detection limit of
Classical tests to detect astroviruses include electron microscopy, the multiplex RT-PCR was between 5 to 10 ng.
fluorescent antibody detection and agar gel diffusion assay. These
tests, while more user friendly, lack the required sensitivity for In situ hybridization
accurate diagnosis. In situ hybridization was described in 2002 by Behling-Kelly et al
Recent advances in molecular biology have opened the door for (6), in which tissue samples were taken from experimentally
the development of many diagnostic assays for the detection of inoculated 3-day-old turkey poults with TAstV-2. Tissues assayed
avian astroviruses and are described here. were intestines, spleen, bursa and thymus. Tissues were de­
paraffinized, digested with proteinase K, and hybridized overnight
with a digoxigenin-labeled riboprobe. The riboprobe was generated
by ZtawHI digested plasmid p25.5, which contains a 1.5 kb segment
of the extreme 3' end of the TAstV-2 genome. In vitro transcription
154
Chapter 34 Enteric Viruses

with T7 RNA polymerase and digoxigenin-labeled UTP was used as day 1 to day 14 PI and the highest proportion of poults positive for
an antisense riboprobe (1.6 kb in length). virus detection was between days 4 and 9 PI.
The in situ hybridization assay is used to detect viral replication in
tissues. It was able to detect TAstV-2 in duodenum, cecal tonsils, SEROLOGICAL DETECTION IN THE HOST
jejunum, distal small intestine and large intestine, but not in bursa,
thymus or spleen. TAstV-2 was detected after 1 day post Neutralization test: A virus neutralization assay was described
inoculation (PI) in cecal tonsils and distal small intestine, then in all recently for Astrovirus (5). In this microtitre neutralization test,
other tissues on day 3 PI. The signals were extensive in distal small two-fold dilutions of serum were mixed with an equal volume of
intestine and cecal tonsils. The signals were relatively infrequent virus diluted in Eagle’s modified essential medium to give 200
by day 9 PI. median tissue culture infective doses per 0.1 ml, the sera were
incubated at room temperature for 1 h and 0.1 ml from each dilution
Multiplex real time RT-PCR was placed into four microtitre wells in a 96-well plate and 0.1 ml
A multiplex real time RT-PCR test was recently developed (38) growth medium and 3xl04 LMH cells. A virus titration series and
for the detection of TAstV-2 and TCoV. Virus detection was negative serum were included in each test. The plates were
evaluated with samples collected from poults inoculated at 1 day of incubated for 5 days at 38.58 C in a CO2 atmosphere and then read
age with each virus. Cloacal swabs and intestinal samples were microscopically. The serum titer was the dilution where only 50%
obtained at 1,2, 3, 4, 6, 9, 14, 17, and 21 days after inoculation and of wells showed CPE. Also, a virus neutralization test in
also from field samples. Primers for the real time RT-PCR for embryonated turkey eggs was described by Tang et al (39), in
astro viruses were designed to amplify a 112 bp fragment of the which 100 EID5o of TAstV 1987 isolate and TAstV 2001 isolate
polymerase gene, the sequence of the forward primer is ( TAV were mixed with six 10-fold serial dilutions of guinea pig antisera,
4248F) 5’-TCC TCC ATG ATT CTC ATA AG- ‘3 and for the then inoculated into 22-day-old turkey embryos by the amniotic
reverse primer (TAV 4360 R) 5’-CTT GAC CTG GCA AAC T-‘3 route. After 96 h of incubation at 37 C, embryos were examined for
and the probe sequence (TAV 4274 PB) 5’- [4J-AAG ATG CGG intestinal lesions.
CGC TTG TA-{TAMRA}-‘3. Primers for TCoV were designed to Enzyme-linked immunosorbent assay. An ELISA system was
amplify a 110 bp fragment of the matrix gene and their sequences developed by Tang et al (39). This system is based on the purified
are: forward primer (TCoV 2F) 5’- AGT GGC TTG CTA AGT-‘3, turkey astrovirus isolates (TAstV1987 and TAstV2001). The
for the reverse primer (TCoV 112R) 5’-GCT TTG GTC ACC AGT- purified antigens were used at 2g/ml in 0.05 M carbonate­
‘3 and for the probe (TCoV 51 PB) 5’-{TXRed}-TAT GCA CAC bicarbonate coating buffer and, after blocking step with 3% bovine
CGG ATA GAC G-{BHQ-2}-‘3. serum albumin in phosphate-buffered saline with 0.1 Tween-20
Assay sensitivity was determined using in vitro transcribed RNA (PBS-T), anti- TAstV 1987 and TAstV 2001 were added in 1:400
and varied by target between 150 gene copies for TAstV-2 alone dilution, then goat anti-guinea pig IgG (H+L) horseradish
and 2200 gene copies for TCoV when multiplexed. The TAstV- peroxidase labeled antibody diluted in 1:20,000 was added, then
2/TCoV test was able to detect the astrovirus isolate (NC/96) and substrate 3,3’, 5,5’-tetramethylbenzidine (TMB) mixed with equal
five additional field isolates as well as TAstV-2. volumes of peroxidase solution B was added for color development
In the poults experimentally inoculated with TAstV-2, the virus at 405 nm.
could be detected in both cloacal swabs and intestinal tissue from

Small Enteric Viruses

PREFERRED CULTURE MEDIA AND SUBSTRATES are mounted on N-aminoethylaminopropyltrimethoxysilane treated


glass slides, processed through xylene-ethanol, and digested with
Enteroviruslike particles from chickens can be serially 0.1% trypsin. The tissue sections are treated with methanol and 3%
propagated by inoculating the chorioallantoic membrane peroxide to quench endogenous peroxidase. Normal goat serum is
(CAM) of embryonated chicken eggs. Enteroviruslike used as blocking agent at a 1:10 dilution. An avidin-biotin block
particles from turkeys can be propagated by inoculating was used to block endogenous biotin in the tissue sections.
embryonated chicken eggs by the yolk sac route. Inocula can Unlabelled goat anti-turkey IgG was added to prevent binding of the
be prepared from intestinal content samples as described above (see biotinylated goat anti-turkey IgG to IgG -producing cells. Hyper
Sample Collection). Procedures for CAM and yolk sac immune turkey anti-enterovirus serum adsorbed with intestinal
inoculation are described in Chapter 44 on virus propagation in powder was added in 1:160 dilution and followed by biotinylated
embryonating eggs. goat anti-turkey IgG. The substrate ExtrAvidin peroxidase
conjugate is added followed by another substrate 3-amino-9-
AGENT IDENTIFICATION ethylcarbazole. Sections were counterstained with Mayer’s
hematoxylin then mounted in glycerol gelatin and examined by
Small enteric viruses (18-24 nm in diameter) are a group of small light microscopy. Positive tissue samples were from ileum, jejunum
viruses that are similar in size and detected by EM but generally and duodenum, with the most severely affected tissues being
cannot be differentiated on the basis of morphology (14), Included jejunum and ileum (16).
in this category are enterovirus, parvovirus, calicivirus and
astroviruses. Enteroviruslike virus was detected in young turkeys Immunofluorescent antibody assay
with enteritis. The virus can be propagated in embryonated turkey The immunofluorescent antibody assay (IFA) is used to detect
eggs, with buoyant density of 1.33 g/ml in CsCl, and has a single­ virus in the infected tissues. Formalin-fixed, paraffin-embedded
stranded RNA genome of approximately 7.5 kb. The standard tissue sections were mounted on glass slides, deparaffinized, and
diagnostic assay for the detection of small enteric viruses is EM in digested with 0.1% trypsin. Slides were incubated with normal goat
which the intestinal contents from affected turkeys are used to serum diluted 1:10 to reduce non-specific background staining.
evaluate the presence of virus (15, 36). Hyperimmune turkey anti-enterovirus serum diluted 1:60 was
added, followed by fluorescein-labeled goat anti-turkey IgG
Immunohistochemistry. Paraffin embedded tissue is used to globulin (Kirkegaard & Perry Laboratories, Gaithersburg, MD).
evaluate the presence of enterovirus-like particles. Embedded tissue Stained sections were examined for infected cells using a
155
Don Reynolds and Ching Ching Wu

fluorescence microscope (Nikon Optiphot; Nikon Inc., Garden City, Reverse transcription-polymerase chain reaction
N.Y.) (16). A turkey enteroviruslike virus was identified to be through cross
immunofluorescence assay and RT-PCR (14). Turkey
Enzyme-Linked Immunosorbent Assay enteroviruslike particle RNA was amplified by RT-PCR with
In 1993, an antigen-capture ELISA was developed for the oligonucleotide primers specific for the polymerase gene (ORF lb)
diagnosis of enterovirus infection in turkey (15, 16). The samples and the capsid protein gene (ORF2) of TAstV-2. RNA samples for
were intestinal contents from naturally and experimentally infected the RT-PCR were extracted from turkey enteroviruslike virus
turkeys. Hyper immune turkey anti-enterovirus serum in 1:1600 infected turkey intestines. The RT-PCR products were of 802 bp
dilution in carbonate buffer was used. Five percent non fat dry milk and 849 bp , respectively. Sequence analysis of these RT-PCR
was used as a blocking reagent. Fecal samples were sonicated and products showed a high degree of similarity to those of TAstV-2
concentrated by centrifugation and diluted in minimum essential (ORFlb; 98.8% and ORF2; 96.9%).
medium at 1:5 dilution. Guinea pig anti-enterovirus antibody was
diluted at 1:800 in tris buffered saline and used as primary antibody SEROLOGICAL DETECTION IN THE HOST
for ELISA. Sheep anti guinea pig IgG peroxidase -labeled antibody
was used at 1:1000 in tris-buffered saline as secondary antibody. 2, Antibodies to enterovirus have been detected by serum
2’-Azino-bis (3-ethyl-benz-thiazoline-6-sulfonic acid) in 0.05 M neutralization and indirect immunofluorescence tests. Serology is
citrate buffer with 3% hydrogen peroxide was used as a substrate. useful to determine the status of enterovirus infection in specific
Color development was read at 410 nm. The sensitivity and pathogen free birds. However, due to the lack of virus isolates and
specificity for this ELISA system compared to EM were 0.963 and reference antisera, routine serological diagnosis is not
879, respectively.
0. recommended (30).

Coronaviruses

PREFERRED CULTURE MEDIA AND SUBSTRATES AGENT IDENTIFICATION

There is currently no known cell culture system that is able to Coronaviruses have been reported to cause clinical diseases in
support turkey coronavirus (TCoV) infection and this is a chickens, turkeys, ducks, geese, pheasants, and pigeons and have
significant limitation both for the ease of diagnosis and the been isolated from these animal hosts. Unlike coronavirus induced
investigation of the virus biology. Turkey coronaviruses can be infectious bronchitis in chickens, the primary target tissue of
propagated/isolated in embryonating chicken or turkey eggs more coronavirus in the other avian species is intestinal tract. Based on
than 15 days of age by amniotic route of inoculation. Twenty two or antigenic relationship between turkey coronavirus (TCoV) and
23-day turkey embryos are commonly used for isolation and other coronaviruses, TCoV was classified as a group 3 coronavirus
propagation of TCoV (21). The inoculum is prepared from the with avian infectious bronchitis virus (IBV). The viral particles of
intestines, intestinal contents, or bursal tissues of suspected turkeys TCoV are surrounded by a fringe of regularly spaced petal-shaped
or turkey poults. The intestines, intestinal contents, or bursal tissues projections attached to the particles by a short stalk. Turkey
are homogenized with 5 volumes of PBS or TBS by motar and coronaviruses are hemagglutinated with rabbit erythrocytes. Turkey
pestle or a tissue homogenizer. The homogenates are centrifuged at coronavirus has the buoyant densities of 1.14 to 1.15 and 1.18 to
500 x g for 30 min. The supernatant is filtered through a 0.45 pm 1.20 g/ml, respectively, using sucrose density gradient
membrane filter membrane prior to inoculation into eggs. If ultracentrifugation (21).
necessary, antibiotics (penicillin or streptomycin) can be added to Coronavirus genome contains a single, positive-strand RNA
the filtrate (inoculum) before inoculation. In addition, the inoculum, molecule, which is about 27 to 33 kilobases (KB) and has a
if needed, can be activated by trypsin by adding 10 pg/ml of type methylated cap at the 5’ end and poly (A) tail at the 3’ end. Genome
ΧΙΠ trypsin to the inoculum, followed by incubation at 37 C for at organization of coronavirus is 5’-polymerase gene-spike gene (S)-
least 1 hr. The air cell end of the egg is disinfected by 70% alcohol membrane protein gene (M)-nucleocapsid gene (N)-3’, in which S,
and iodine. A 26-gauge needle with the syringe containing the M, and N are the structure genes. The whole TCoV genome consists
inoculum is inserted through a hole in the egg shell (created in the of 27,749 nucleotides, excluding poly (A) tail. Turkey coronavirus
air cell end of the egg) and subsequently through the chorioallantoic polyprotein gene encodes two open reading frames. There are 9
membrane and reaches the amniotic cavity. One-hundred to 500 pl open reading frames in the structure protein genes, representing the
of inoculum are injected into the amniotic cavity. The needle is entire S protein gene, tricistronic gene 3, M protein gene, bicistronic
removed and the hole in the egg shell is sealed by parafilm, gene 5, and N protein gene in the order of 5’ to 3’(23).
cellophane tape, or silicone gel. Viruses can be recovered from Turkey coronavirus can be identified and/or confirmed by EM,
turkey embiyo intestines or bursae 2 to 5 days after amniotic IEM, IFA, immunohistochemistry, hemagglutination, RT-PCR, and
inoculation. Turkey coronavirus is detected in turkey embryo multiplex PCR.
intestines or bursae by immunofluorescent antibody assay (IFA)
using a TCoV-specific antiserum or monoclonal antibody. Electron Electron microscopy
microscopy (EM) can also be used for intestinal contents or Intestinal contents or homogenates (3 or 5 grams) are placed into a
homogenates to detect enveloped viruses of 50-200 nm with typical stomacher bag with addition of 50 ml of water and the stomacher
coronavirus peplomers on the membrane. Definitive diagnosis by bag is processed in the stomach machine for 30 sec. Four ml of the
EM can be achieved by immune electron EM (IEM) busing a mixture is added with an equal volume of water and centrifuged at
TCoV-specific antiserum. 8,000 rpm for 5 min. Four ml of the supernatant is added with an
equal volume of water and centrifuged at 20,000 rpm for 60 min.
The supernatant is poured off and the pellet is added with a mixture
consisting of 4 drops of phosphotungstic acid and 3 drops of 0.1 %

156
Chapter 34 Enteric Viruses

bovine serum albumin and vortexed for 30 sec to 1 min. The stained cold ethanol and pellets were resuspended in RNAse-free water (7).
pellet is transferred to a nebulizer, sprayed onto the grids, and In another report (45), the intestines were homogenized in minimom
observed on an electron microscope. Turkey coronavirus particles essential medium (MEM) with penicillin (0.5 U/ml) and
are spherical, enveloped, and surrounded by regularly spaced petal- streptomycin (0.5 mg/ml). The homogenate (20% w/v) was
or pear-shaped projections. They are 50 to 200 nm in diameter (21). centrifuged at 8,000 x g for 10 min. Viral RNA was extracted from
the supernatant of intestinal homogenate using QIAamp viral RNA
Immune electron microscopy mini kit (Qiagen, Valencia, CA) (45). RNA was subjected to
Intestines or intestinal contents are mixed or homogenized with 5 reverse transcription followed by PCR amplification. Several sets of
volumes of TBS and centrifuged at 250 x g for 10 min. The PCR oligonucleotide primers designed from the nucleocapsid (N)
supernatant is filtered by 0.8-pm membrane filter, followed by protein gene sequence of TCoV were used in PCR amplification:
0.45-pm membrane filter. The filtrate is incubated with diluted Nl-upper (CAGCGCCAGTCATCAAAC) and N2-lower
turkey anti-TCoV serum overnight at 4 C. The samples are (TGGTCAAACTTGTCAGGGTCC) amplifying 380 bp (45), N3-
centrifuged at 20,000 rpm for 60 min. The pellet is resuspended in upper (CAAGTAAAGGCGGAAGAAAAC) and N4-lower
distilled water and added with a 3.0% phosphotungstic acid solution (GCCTTAGTAATGCGAGAGCCC) amplifying 417 bp (45), and
to yield a final concentration of 1.5% phosphotungstic acid. The NF (TCTTTTGCCATGGCAAGC) and NR
samples are applied to the grids and observed on an electron (TTGGGTACCTAAAAGTTCATTCTC) amplifying 1230 bp (26).
microscope (16). However, all these primer sets also amplified N gene of infectious
bronchitis virus (IBV).
Immunofluorescent antibody assay
The IFA assay for TCoV antigen detection in the intestine or bursa Multiplex PCR
of Fabricius is a sensitive assay due to the use of a fluorochrome A rapid, sensitive, and specific multiplex PCR method has been
conjugated antibody (14, 22, 25, 32). TCoV antigen can be detected established for specifically differential detection of TCoV, IBV, and
in the intestine of experimentally infected turkeys from 1 to 28 days bovine coronavirus (BCoV) (27). Intestines of turkey embryos
post infection. The small intestine (jejunum and ileum) is frozen infected with TCoV were homogenized with 5-fold volume of
immediately after collection, embedded in embedding medium, and phosphate buffered saline (PBS) solution. These homogenates were
sectioned in 6-pm thickness. Tissue sections are fixed in acetone for centrifuged at 1,500 x g for 10 min. Two hundred microliters of
30 min at room temperature and incubated with turkey antiserum virus-containing supernatants were mixed with 1 ml of RNApure
specific for TCoV at a dilution of 1:40 in dilution buffer, containing reagent (GenHunter, Nashville, TN) and incubated on ice for 10
150 mM phosphate buffer, 0.85 % NaCl, 1 % BSA, and 0.02 % min. After addition of 180 μΐ of chloroform, the mixture was mixed
Tween-20, in a humidifying chamber at room temperature for 30 vigorously for 10 sec and centrifuged at 13,000 x g for 10 min. The
min. After washing with PBS buffer for 3 times, intestinal sections upper aqueous phase was mixed with equal volume of cold
are incubated with fluorescein isothiocyanate (FITC) conjugated isopropanol and incubated on ice for 10 min. The RNA precipitate
goat anti-turkey IgG (H+L) antibody (Kirkegaard & Perry was pelleted by centrifugation at 13,000 x g for 10 min and washed
Laboratories) at a dilution of 1:40 in dilution buffer at room with 70 % ethanol. The RNA was dissolved in 50 μΐ of diethyl­
temperature for 30 min. Sections are read on a fluorescent pyrocarbonate (DEPC) treated sterile double-distilled water. The
microscope (Nikon Optiphot; Nikon Inc.). RNA was combined with random hexamers (50 ng) in 11 μΐ of
DEPC-treated water, heat denatured at 70 C for 3 min, and
Immunohistochemistry immediately placed on ice for 5 min. Reverse transcription buffer
Frozen intestinal or bursal sections can be acetone-fixed, incubated containing 200 units of reverse transcriptase (Superscript Π system,
with TCoV-specific monoclonal antibody (Mab 4.24), and followed Life Technologies, Gaithersburg, MD) and 0.2 mM of each of the
by an immunoperoxidae procedure to reveal TCoV antigens in the four deoxynucleotide triphosphates (Promega Corp., Madison, WI)
enterocytes in tissue sections on a light microscope (7). This assay was added. A total volume of 20 μΐ reverse transcription was
had high specificity (96%) and low sensitivity (61%) when carried out at 42 C for 60 min. The primers used in multiplex PCR
compared to virus isolation (7). were based on the alignments of spike (S) or N gene sequences
among TCoV, IBV, and BCoV to identify the variable and
Hemagglutination conserved regions (23). The upstream primer N103F and
Turkey coronaviruses can agglutinate guinea pig and rabbit downstream primer N102R common to both TCoV and IBV were
erythrocytes. Turkey coronavirus antigens are propagated, purified, designed according to conserved regions of N gene sequences. The
and concentrated as described in the section of virus neutralization sequence of upstream primer N103F (cctgatggtaatttccgttggg) and
for TCoV in this Chapter (9, 21). Erythrocyte suspension of 0.5% that of downstream primer N102R (acgcccatccttaataccttcctc)
are used to mix with equal volume of TCoV antigen. The assay is amplify a 357-bp sequence of TCoV or IBV N gene in the
performed at room temperature for 1 hr. The procedures of conserved regions corresponding to nucleotide position 445 to 801
hemagglutination and hemagglutination inhibition can be referred to of TCoV N gene. The upstream primer S306F
the Chapter on serological procedures. (tgtatctaatttgggtgggtttga) and downstream primer S306R
(ataagctgctaattgaagggatgc) are specific to TCoV and based on the
Reverse transcription-polymerase chain reaction alignments of S gene sequences in the variable regions among these
Several approaches have been used to extract viral RNA from the viruses. This set of primers specifies a 727-bp sequence
intestinal contents/dropping samples. In one method (7), the sample corresponding to nucleotide position 2,019 to 2,745 of TCoV S
was prepared as 20% (w/v) suspensions in TNE buffer (0.01 M gene (23). The upstream primer S3 (atgtgtgtaggtaatggtcctgg) and
Tris-hydrochloride, pH7.4, 0.1 M NaCl, 1 mM downstream primer S6 (agcaactacgaatcataaaa) are specific to BCoV
ethylenediaminetetraacetic acid) and sonicated for 30 sec. These and designed according to variable regions of S gene sequences
suspensions were clarified by centrifugation at 1000 x g for 10 min among these viruses. This set of primers amplifies a 568-bp
at 4 C and then at 8000 x g for 30 min at 4 C. The supernatant was sequence corresponding to nucleotide position 1,488 to 2,055 of
layered onto a 20% (w/v) sucrose cushion and centrifuged at 80,000 BCoV-Quebec S gene. PCR was performed in a 96-well thermal
x g for 2 hr at 4 C. The resultant pellets were incubated in 0.5% cycler (GeneAmp, Perkin-Elmer Cetus Corp., Norwalk, CT). The
sodium dodecyl sulfate for 5 min at room temperature followed by reaction mixtures contained 2μ1 of cDNA, 0.2 μΜ of each of the
two phenol-chloroform extractions. Nucleic acid was precipitated in primers N103F, N102R, S306F, S306R, S3, and S6 in 50 μΐ of PCR
157
Don Reynolds and Ching Ching Wu

buffer comprised of 0.2 mM of each of the four deoxynucleotide plasmid vector (Invitrogen) and transformed into E. coli strain
triphosphates, 5 units of Taq DNA polymerase (Promega), 10 mM TOPI OF' (Invitrogen). Correct clones were selected and subjected
Tris-HCl (pH 9.0), 50 mM KC1, 1.5 mM MgCl2, 0.1 % Triton X- to sequencing reaction using dideoxy-cycle sequencing method with
100, and 0.01 % gelatin. The cyclic parameters of the PCR were 94 the corresponding sequencing primers for both strands. The
C for 30 sec for denaturation, 50 C for 1 min for annealing, 72 C for nucleotide and deduced amino acid sequence similarities among the
1 min for extension for 25 cycles followed by 72 C for 10 min final TCoV isolates were analyzed by the Clustal W method in MegAlign
extension. Two PCR bands of 727 and 357 bp, respectively, were module of the DNAstar program (Lasergene Corp, Madison, WI).
seen in the TCoV isolates. One PCR band of 357 bp was shown in Percent similarities were calculated to find nucleic acid and amino
the IBV strains. One PCR band of 568 bp was obtained for the acid pair distances. Based on the obtained sequences, phylogenetic
BCoV strains and isolates. trees of TCoV isolates were constructed. Pair-wise comparison of
nucleotide sequence distance for the entire 3’-end structural protein
Strain Variability gene sequences among TCoV isolates revealed the similarity scores
Turkey coronavirus was identified in the early 1970s as the major ranging from 92.7 % to 99.4 %. Pair-wise comparison of nucleotide
cause of the most costly disease of turkey encountered in Minnesota and deduced amino acid sequence distance of S protein gene
between 1950s and 1970s. In addition, outbreaks of turkey sequences among TCoV isolates had the similarity scores ranging
coronaviral enteritis occurred in Quebec, Canada in late 1980s, from 93.0 % to 99.7 % at the nucleotide level or from 92.5 % to
Indiana, U.S. in early and mid 1990s, and North Carolina, U.S. and 99.3 % at the amino acid level. Phylogenetic analysis according to
Virginia, U.S. in mid and late 1990s and has continued to remain as the entire 3’-end structural protein region or the deduced amino acid
the disease of threat to the turkey industry in North Carolina, sequence of S protein gene showed that TCoV isolates were
Virginia, Arkansas, Missouri, and other states in the U.S. in the clustered within the same genomic lineage. This indicates that
2000s. Variability of TCoV strains or isolates from different TCoV isolates from various geographic locations in the U.S. are
geographical locations in different time period can be determined closely related genetically (28).
by immunological methods and molecular approaches.
Immunological methods. Immunofluorescent antibody assay, SEROLOGICAL DETECTION IN THE HOST
ELISA, and virus neutralization are immunological methods that
can be used to study variability of TCoV strains or isolates. The Immunofluorescent antibody assay for antibody to TCoV
procedures of these methods have been described in this Chapter. Frozen turkey intestines infected with TCoV were sectioned at 6-
The cross antigenic reactivity of 18 TCoV isolates from various pm thickness and incubated with 2-fold serially diluted turkey
geographical locations in the U.S. was studied by IFA using serum at room temperature for 30 min. The intestinal sections were
antibodies to different TCoV isolates (22). Intestinal sections were subsequently incubated with FITC-conjugated goat anti-turkey IgG
prepared from turkey embryos infected with different TCoV isolates (H+L) antibody (Kirkegaard & Perry Laboratories) at a dilution of
and reacted with polyclonal or monoclonal antibodies specific to 1:40 in dilution buffer at room temperature for 30 min in a
TCoV in immunofluorescent antibody staining. All 18 TCoV humidifying chamber. Sections were read on a fluorescent
isolates had the same antigenic reactivity pattern with the same microscope (Nikon Optiphot; Nikon Inc.). The titer of turkey sera
panel of antibodies. This indicates that TCoV isolates from various was defined as the reciprocal of the highest dilution of test sample
geographic locations in the U.S. are antigenically identical or still having positive staining (25, 32). Alternatively, TCoV-infected
closely related (22). epithelial cells exfoliated from the bursae of Fabricius can be served
Molecular methods. Turkey coronavirus genome is a positive as the source of TCoV antigen. Exfoliated TCoV-infected epithelial
single-stranded capped RNA with a polyadenylated 3’ end. The 5’ cells were spotted onto glass slides, air-dried, and fixed in cold
two-thirds of the coronavirus genome consist of two overlapping absolute acetone for 10 min. FITC-conjugated rabbit anti-chicken
open reading frames (ORFs) that encode non-structural proteins immunoglobulin G (ICN Biomedicals, Inc., Costa Mesa, CA) with a
including the viral RNA-dependent RNA polymerase. Another one- 1:40 dilution was used as the secondary antibody (13).
third nucleotide sequences, including the structural protein genes
and 3’ UTR, in the 3’ end have a total of 6,963 nucleotides in the Antibody-capture enzyme-linked immunosorbent assay
structural protein genes that consist of the entire S protein gene, Based on the positive antigenic cross-reactivity between TCoV
tricistronic gene 3, membrane (M) protein gene, bicistronic gene 5, and IBV, commercially available IBV-coated ELISA plates
and N protein gene in the order of 5’ to 3’ along the genome (23). (IDEXX, Westbrook, ME) were successfully used for the detection
PCR, cloning, sequencing, and sequence analysis (including of anti-TCoV antibody (24). Anti-TCoV hyperimmune turkey
sequence alignment, degree of sequence homology, and serum and normal turkey serum were used as positive or negative
phylogenetic analysis) are the major molecular methods to control sera for optimization of the ELISA. Goat anti-turkey IgG
determine variability of TCoV strains or isolates. The 3’-end (H+L) conjugated with horseradish peroxidase (Kirkegaard & Perry
structural protein gene sequences of TCoV isolates associated with Laboratories) was used as detector antibody (conjugate). The
outbreaks of acute enteritis in Indiana, North Carolina, and differentiation between anti-TCV hyperimmune serum and normal
Minnesota have been studied by carrying out PCR, cloning, and turkey serum were achieved when using 1:40 serum dilution, and
sequencing (28). TCoV isolates were propagated in 22-day-old 1:1,600 conjugate dilution. Ninety six-well microtiter plates coated
turkey embryos. Total RNA was extracted from the infected with IBV antigens were incubated with turkey serum samples (1:40
intestines and intestinal content by using guanidinium thiocyanate dilution) to be tested in quadruplicate and incubated at 37 C for 1
and acid-phenol. The RNA was dissolved in 150 μΐ of diethyl­ hr. The plates were subsequently incubated with goat anti-turkey
pyrocarbonate (DEPC) treated sterile double-distilled water. Two IgG (H+L) conjugated with horseradish peroxidase (1:1,600
micrograms of the total RNA was heat denatured at 100 C for 3 min dilution) at 37 C for 1 hr., followed by the addition tetramethyl
and slowly cooled to 22 C in 15 min in reverse transcription (RT) benzidine (TMB) solution. Two reference wells that contained all
buffer (Life Technologies) containing 40 ng of random hexamers. reagents except serum samples were included in each plate. Positive
The reverse transcription was carried out at 42 C for 60 min. Three and negative control sera as well as test sera were tested in
microliters of cDNA were used in PCR amplifications with the duplicate. The absorbance value of each well was measured at 450
primers spanning for 4 overlapping fragments covering the entire nm using a spectrophotometer (Vmax™ Kinetic Microplate Reader,
3’end structural protein genes as described in a previous report (23). Molecular Devices Corporation, Menlo Park, CA). The ELISA
One microliter of the amplicon was used to ligate with pCR-Π value or S/P ratio of each test serum was calculated as (absorbance
158
Chapter 34 Enteric Viruses

value of sample serum minus absorbance value of negative control The sensitivity and specificity of ELISA relative to IFA test were
serum) divided by (absorbance value of positive control serum 98.7% and 98.8%, respectively. The recombinant TCoV S protein­
minus absorbance value of negative control serum). The optimum based antibody-capture ELISA is useful in detection,
cutoff point to separate positivity and negativity was determined to differentiation, and evaluation of antibody response to TCoV.
be 0.18 by logistic regression analysis. The sensitivity and
specificity of this ELISA relative to IFA test were 93.1 % and 96.7 Competitive enzyme-linked immunosorbent assay
%, respectively. A competitive enzyme-linked immunosorbent assay (cELISA) was
Another ELISA to detect TCoV antibody (26) was developed developed for detection of antibodies to TCoV by using a
using the recombinant N protein of TCoV expressed in E. coli recombinant baculovirus-expressed TCoV N protein (as mentioned
transformed with pTriEx-1 (Novagen, Madison, WI) plasmid above) and biotin-labeled TCoV N protein-specific monoclonal
construct carrying N gene of TCoV. The optimum conditions for antibody (Mab 4.23) (13). The rBTCV/N protein was diluted
differentiation between anti-TCoV serum and normal turkey serum 1:1280 in 0.2 M carbonate/0.2 M bicarbonate buffer and 75 μΐ was
were: coating concentration of recombinant E. coli-expressed TCoV added to each well in 96-well ELISA plates and incubated overnight
N protein at 20 pg/ml, serum dilution of testing turkey serum at 4 C. rBTCV/N protein-coated plates were washed three times
sample at 1:200 to 1:800, and conjugate dilution at 1:10,000 or with 0.01 M PBS, pH 7.2, containing 0.05% Tween 20 (PBST), 200
1:20,000. The optimum cutoff point to separate positivity and μΐ of block buffer (PBST containing 1% nonfat dry milk) was added
negativity was determined to be 0.18 by logistic regression analysis. to each well and incubated for 1 hr at 25 C. After washing three
In addition, Recombinant N protein of TCoV produced by times with PBST, positive and negative control sera and test sera
recombinat baculovirus encoding TCoV N gene (rBTCV/N) has were diluted 1:10 in block buffer and 50 μΐ of each were placed in
been shown to be useful in detecting antibody to TCoV in an duplicate wells and plates were incubated for 60 min at 25 C. A
antibody-capture ELISA (8). Recombinant baculovirus was diluent control consisting of block buffer (50 μΐ) also was placed in
generated by co-transfection of linearized AcMNPV DNA (Bac-N- duplicate wells. Washing three times with PBST, 50 μΐ of biotin-
Bluea DNA, Invitrogen, San Diego, CA) and pBacTCV DNA labeled MAb 4.23 diluted 1:160 in block buffer were added to each
carrying TCoV N gene cloned into pMelBac C vector (Invitrogen). well and incubated for 60 min at 25 C. Washing three times with
The rBTCV/N protein was diluted 1/80 in 0.2 M carbonate/0.2 M PBST, 75 μΐ of streptavidin-horseradish peroxidase (Kirkegaard and
bicarbonate buffer, pH 9.6, added to 96-well ELISA plates, and Perry Laboratories) diluted 1:200 in block buffer was added to each
incubated overnight at 4 C. rBTCV/N protein coated plates were well and incubated for 30 min at 37 C. Washing three times with
washed three times with PBS plus 0.05% Tween 20 (PBST), PBST, 100 μΐ of 2,2’-azino-bis (3- ethylbenzthiazoline-6-sulfonic
followed by 200 μΐ of block buffer (PBST containing 1% nonfat acid) substrate (Kirkegaard and Perry Laboratories) were added to
dried milk) being added to each well and incubated for 1 hr at room each well; color development was stopped after 20 min with 1%
temperature. Following washing three times with PBST, 75 μΐ of (w/v) SDS in water. Plates were read on an ELISA reader (BT 2000
testing serum, diluted 1:40 in block buffer, was added to each well MicroKinetics Reader, Fisher Scientific) at 405 nm. Optical
and incubated for 30 min at 37 C. Seventy-five μΐ of peroxidase- densities of duplicate wells, including positive and negative control
labeled goat anti-chicken immunoglobulin G in 1:50 dilution was sera wells, and diluent wells were averaged. Percentage of
added to each well and incubated for 30 min at 37 C. One-hundred inhibition of optical densities of test serum wells relative to negative
μΐ of 2, 2'-azino-bis (3-ethylbenzthiazoline-6-sulfonic acid) control serum was calculated after subtracting the diluent control,
substrate (Kirkegaard and Perry Laboratories) was added to each which was subtracted from all test and control well averages, to
well; color development was stopped after 15 min with 1% (w/v) yield a corrected value. Percentage of inhibition was calculated as
SDS in water. Plates were read with a ELISA reader at 405 nm. The 100 - (100 x [test serum - diluent/negative control - diluent]). Sera
TCoV N ELISA detected antibodies in all antisera prepared against were considered to be positive if inhibition >45% was observed and
TCV and IBV strains. Samples were considered positive if their negative if inhibition <45%. The cELISA was shown to have a
absorbance was > 0.1. Absorbance values for TCoV and IBV sensitivity of 92.9% and a specificity of 96.2% when compared with
antisera were > 0.3. IFA. The cELISA also detected antibody to IBV.
Since TCoV and IBV N gene sequences are conserved and share
cross-reactivity, antibody-capture ELISA based on IBV antigen or Virus neutralization
recombinant TCoV N protein can detect serum antibody to TCoV as Turkey coronavirus is propagated in embryonating turkey eggs as
well as that to IBV. However, S gene sequences between TCoV and described above and infected intestines are harvested 3 days after
IBV are markedly different, with only 33.8% to 33.9% of sequence inoculation. Collected intestines are homogenized with 5 volumes
homology (23). Therefore, antibody-capture ELISA based on TCoV of TBS by mortar and pestle or a tissue homogenizer. After freeze
S protein should be able to detect TCoV-specific antibody, and thaw three times and sonication, the homogenate is centrifuged
differentiate between antibodies to TCoV and IBV, and evaluate at 8,000 x g for 20 min. The supernatant was ultracentrifuged at
TCoV-specific antibody response in the turkey flocks. A TCoV- 100,000 x g for 3 hr through a cushion of 15 ml of 30% sucrose
specifc antibody-capture ELISA has been achieved for the detection solution. After resuspension of the pellet in TBS, the sample was
of antibody to TCoV in turkey sera using recombinant partial S layered on the top of 40 to 65% sucrose gradient and
protein of TCoV expressed in E. coli transformed with pTriEx-1 ultracentrifuged at 100,000 x g for overnight. Two TCoV-
(Novagen) plasmid construct containing the insert of partial S gene containing fractions with sucrose densities of 1.18 to 1.20 and 1.14
of TCoV as the coating antigen in the ELISA plates (46). A partial to 1.15 g/ml, respectively, are collected, titrated, and used in virus
SI gene fragment of TCoV was ampilified by PCR, cloned into an neutralization (10, 21). The serum samples to be tested are 10-fold
expression vector (pTriExl, Novagen), and expressed in E. coli serially diluted and incubated with 0.01, 0.1, or 1 egg infective dose
(Rosetta cells, Novagen). The recombinant partial SI protein was (EID50) of TCoV at 37 C for 1 h. Two-hundred microliters of
purified by His-Bind column chromatography (Novagen) and used mixture are inoculated into the embryonating turkey eggs as
to coat the ELISA plates. The optimal parameters for differentiation described above. The intestines from the turkey embryos are
between anti-TCoV serum and normal turkey serum by ELISA collected 3 days after inoculation and examined by IFA as described
were: purified recombinant partial S protein at 20 pg/ml, serum above. The virus neutralization titer is expressed as the reciprocal of
dilution 1:200 to 1:800, and conjugate dilution 1:10,000, 1:20,000, the highest dilution of the serum sample capable of neutralizing
or 1:40,000. The optimum cutoff point to separate positivity and virus without causing infection to turkey embryos.
negativity was determined to be 0.32 by logistic regression method.
159
Don Reynolds and Ching Ching Wu

Hemagglutination inhibition After thorough mixing and incubation at room temperature for 30
Turkey coronavirus antigens are propagated, purified, and min the serum-kaolin preparation is centrifuged at 500 x g for 30
concentrated as described above (10, 21) and are diluted to yield min and the supernatant (serum) is used. Erythrocytes are added to
four hemagglutination units. The serum samples to be tested are the kaolin-adsorbed serum in a similar manner. The procedures of
adsorbed with kaolin and mixed with erythrocytes to minimize non­ hemagglutination and hemagglutination inhibition can be referred to
specific agglutination. The serum is diluted to 1:5 with sterile PBS the Chapter 47 on serological procedures.
and added to an equal volume of a 25% kaolin suspension in PBS.

Rotaviruses

PREFERRED CULTURE MEDIA AND SUBSTRATES may contain substances that are toxic to the cells. If this is the
case, dilute the sample 1:25 or 1:50 with serum-free media and
Avian group A rotaviruses have been propagated in primary then add type IX trypsin.
cultures of chicken kidney, turkey kidney, and chicken embryonic 4) The trypsin-activated inoculum is placed onto the cell monolayer.
liver cells. However, the continuous cell line derived from For screw-capped tubes, 0.2 ml of inoculum per tube is typically
fetal rhesus monkey kidney, which is referred to as the MA 104 used. Either tubes can be rotated by hand once every 10 to 15
cell line, is the most widely used and convenient method for the min, or, they can be placed directly into a roller-drum
isolation and propagation of these viruses. apparatus and rotated. The inoculation period is 1 h.
Roller tubes are convenient for initial propagation of rotavirus 5) Following inoculation, the monolayers are washed with serum-
isolates, although when propagation of large amounts of virus free media to get rid of any residual inoculum. Maintenance
is needed, roller bottles can easily be used. Stationary cultures medium, consisting of antibiotic containing serum-free EMEM
of MA 104 cells may also support avian rotavirus growth, and the or DMEM with 1-2 pm/ml of type IX trypsin, is added to each
continuous rolling of cultures (as with roller tubes or bottles) tube.
aids in rotavirus isolation and propagation. 6) Virus-inoculated cells are incubated for 48 to 72 h before
Trypsin activation of rotaviruses is essential for cell culture harvesting. CPE is typically seen within 24 h of inoculation.
propagation, as is the incorporation of trypsin into the maintenance However, initial isolates may need to be blind-passaged several
media. Type IX trypsin (Sigma Chemical Co., St. Louis, MO) is times before CPE is observed. For a complete description of
widely used for activation. Not all type IX trypsin has the same rotavirus CPE in MA104 cells, see Theil et al. (41).
activity. The amount of trypsin activity may be influenced by 7) Virus is harvested between 48 and 72 h after inoculation.
factors such as age and different manufacturers' lots. Therefore, it Tubes containing cells are frozen at -20 C or lower, thawed,
is advisable to titrate the amount of trypsin needed in the sonicated, and clarified by centrifugation at 500 x g for 10
maintenance media before virus inoculation. This is done by min.Virus is contained in the supernatant, which can be used for
incorporating different concentrations of trypsin (usually 0.5, further passage, purification, and so on.
1.0, 1.5 ... 4.0 pg/ml) into the maintenance media and using these
media in roller tubes containing confluent monolayers of MA 104 AGENT IDENTIFICATION
which have not been infected with virus. Usually within hours, the
higher concentrations of trypsin will cause the monolayers to Avian rotaviruses are double-stranded (ds) RNA viruses ranging
detach from the glass roller tube. The optimum concentration in size from 60 to 75 nm. Rotaviruses may be observed as
of trypsin to be used in the maintenance media is that concentration double-shelled particles ranging in approximate size from 70
that will leave at least 50% of the monolayer attached to the glass to 75 nm or as single-shelled particles ranging in approximate
after 24 hours. size from 60 to 65 nm. Rotaviruses possess a distinct
Avian rotaviruses other than Group A rotaviruses have not been morphology that aids in their identification. For more
routinely isolated or propagated in cell culture. Isolation and information see McNulty (29) or Theil (41). The buoyant
propagation of these viruses have been limited to orally density of rotaviruses in cesium chloride is approximately 1.36 g/
infecting birds that are known to be free of other enteropathogens cm3 for double- shelled particles and 1.38 g/cm3 for single­
(specific-pathogen-free [SPF] birds are desirable) with shelled particles. Avian rotaviruses are classified according to
preparations of these agents. Virus can be recovered from the groups based on serologic assays and genome electropherotyping.
intestinal tracts of these birds several days following inoculation. To date, there have been three groups of avian rotaviruses
The following procedure can be used to isolate and identified. The group of rotaviruses isolated most commonly
propagate group A rotaviruses in cell culture: from turkeys is the group D rotavirus, also referred to as
1) Screw-capped tubes are seeded with MA 104 cells and grown rotavirus-like virus and para-rotavirus. Another group
to confluency using either Eagle's minimum essential (EMEM) or frequently isolated from avian species is Group A. A third group
Dulbecco's modified Eagle's medium (DMEM) supplemented (which has not been placed into a designated group) referred to as
with 10% fetal bovine serum (FBS) and antibiotics (penicillin the atypical rotavirus occurs infrequently in turkeys.
50 IU/ml, streptomycin 50 pg/ml, and amphotericin B 0.125 A number of assays can be used to identify rotaviruses. Two such
pg/ml). If the screw-capped tubes form a tight seal, they need assays, IEM and electropherotyping, which are capable of
not be incubated under conditions supplemented with CO2. All detecting and distinguishing the various groups of avian
cultures are incubated at 37 C. rotaviruses, are described under serologic detection and
2) Cell monolayers must be washed with serum-free media (or strain variability. There are commercially available assays that
other balanced salt solutions) three times in order of get rid of are capable of detecting rotaviruses. Examples of such assays
any residual FBS that may inhibit the virus or inactivate the would include latex agglutination assays and ELISAs. As of this
trypsin to be added. writing, kits that are commercially available are directed towards
3) The inoculum containing the virus is prepared from the the detection of mammalian (usually human) group A
sonicated filtrate as described above. The inoculum must first be rotaviruses and are of limited value because they are unable to detect
activated with trypsin before inoculation onto the cell monolayer. group D or atypical avian rotaviruses.
This is accomplished by the addition of type IX trypsin to a
final concentration of 10-20 pg/ml of inoculum. Some samples
160
Chapter 34 Enteric Viruses

Serologic Identification 6) Staining the sample preparation with negative stain. To the
Assays employing immune serum are beneficial for detecting drop containing the resuspended pellet, add 1 drop (from a Pasteur
and differentiating avian rotaviruses. The most commonly used pipet) of 3% phosphotungstic acid (PTA) stain. In this way,
serologic techniques for antigen detection are the FA and LEM the final PTA concentration of the sample preparation is
techniques. For further information on how to perform direct and approximately 1.5%. The 3% PTA stain is prepared by adding
indirect fluorescent antibody techniques, see Gardner (12). The 3 g of PTA in high quality water and adjusting the pH to 7.2-
IEM technique is described below. 7.4 with IN potassium hydroxide. The final staining solution can
Immune Electron Microscopy (IEM). The following be filtered through a 0.22-pm filter and stored in a light-proof
procedures are used: container. Proteinaceous material (e.g. antibody, debris) aids in
1) Preparation of antisera. Convalescent antisera from spreading the sample over the grid. If the sample is relatively
infected birds (SPF birds are preferred) or hyperimmune antisera clean of any debris (such as a sample derived from cell culture),
from inoculated animals can be used. Before being used, sera are then difficulty in achieving the proper spread of the sample over
heat-inactivated at 56 C for 30 min, subjected to the grid may occur. Adding a low percentage of protein (e.g.
ultracentrifugation at approximately 45,000 x g for 45 min, filter- 0.1% bovine serum albumin) to the diluent may help alleviate this
sterilized, and aliquoted. Aliquoted sterile sera can be stored at problem. The sample can be mixed with the stain by pipeting
4 C. A determination of the optimum working dilution can be the preparation up and down with a Pasteur pipet. Place a drop
made by serially diluting the sera in sterile PBS and of the stained preparation onto a 300-or 400-mesh carbon-coated
performing the rest of the IEM procedure. Typically, Formvarg grid. After 1 to 3 min, blot the drop from the grid
working dilutions of convalescent sera will range from 1:50 to using bibulous paper held at one edge of the grid.
1:200 sera:PBS). Working dilutions of hyperimmune sera are 7) Viewing the specimen. View the specimen at 80 KV at
usually much higher. magnifications from 30,000 to 60,000. Clumps of virus usually
2) Preparation of the sample. When examining intestinal samples, have from five to hundreds (sometimes thousands) of virus
the intestinal tracts can he processed as described above (see Sample particles. For descriptions of rotaviruses observed by IEM, see
Collection). The resulting sonicated filtrate is used for the rest Theil et al (41) and Saif et al (36).
of the procedure. When examining cell-culture material, the
supernatant obtained from harvesting infected cells (see Strain variability
Preferred Culture Media and Substrates) is used. Rotaviruses are one of the major etiological agents of
3) Incubation of the sample and antisera. The volume of sample gastroenteritis in various species of mammals and birds. In recent
that can be combined with antisera (’’antisera’’ refers to the years, avian rotaviruses have been isolated from turkeys, chickens,
working dilution of antisera, see above) is dependent upon a guinea fowl and pheasants causing diarrhea and high mortality.
number of variables such as the strength and/or dilution of the antisera, Avian rotaviruses are morphologically identical and antigenically
the amount of sample used, and the equipment available. Typical related to mammalian rotaviruses. Avian rotaviruses contain 11
total volumes of sample plus antisera preparation are 0.5 to 1.0 ml. segments of double stranded (ds) RNA that code for six structural
Ratios of sample to antisera are generally between 2:1 and 4:1, proteins (VP 1-4, VP6 and VP7) and six non structural proteins
(e.g., 0.5 ml sample plus 0.2 ml antisera). The incubation can also (NSP1-6). Rotavirus particles consist of 3 layers: the central core,
vary depending upon the sample and antisera involved. For the inner capsid protein and the outer capsid protein. The inner
maximum effect, the sample/antisera preparation can be capsid protein VP6 is the most abundant protein and possesses the
incubated at 4 C overnight, although incubation for 1 h at room group and subgroup antigens. The complete genomic sequence of
temperature is usually sufficient. avian rotavirus was reported in 2001 by Ito, et al (17).
4) Pelleting the virus/antibody complexes. Following incubation, Rotaviruses isolated from different species can be distinguished by
virus/antibody complexes contained in the sample/antisera the electrophoretic migration patterns (electropherotypes) of their
preparation are pelleted by subjecting the preparation to dsRNA upon agarose gel electrophoresis (44). The characteristic
ultracentrifugation at approximately 55,000 x g for 45 min. Mark electropherotypes of rotaviral dsRNA of different specimens can be
ultracentrifuge tubes before ultracentrifugation in order to used as specific markers of different virus populations. Like
locate pellet (see below). If the sample was prepared from mammalian group A rotaviruses, segments 7, 8 and 9 of avian
intestinal samples (or similar material), it may contain much group A rotaviruses migrate as a triplet. Unlike their mammalian
unwanted debris that may interfere with the final preparation when counterparts, segment 5 of avian rotavirus migrates very close to
viewed on the EM. To remove such debris, the sample/antisera segment 4. Avian electropherogroup A rotaviruses display a 5-1-3-
preparation can be pelleted through a layer of 40% to 60% sucrose. 2 migration pattern, whereas mammalian electropherogroup A
The layer of sucrose will act as a barrier to hold back low-density rotaviruses show a 4-2-3-2 pattern, thus permitting a
debris. The sucrose layer should not be used if the sample is differentiation of avian and mammalian strains (42, 43). There have
relatively pure (such as cell-culture material) because some been a lot of improvements in the diagnosis of mammalian
debris aids in the spread of the stained sample on the grid (see rotaviruses, especially human rotavirus, in recent years but this is
below). Additionally, the sucrose may act to hold back viruses not the case for avian rotaviruses.
with low buoyant densities along with the debris. Therefore, caution Avian rotaviruses can be placed into groups based on serologic
must be taken depending on the intended use of the preparation. If the assays such as FA and IEM procedures (see above), and
sample/antisera preparation is pelleted through the layer of electropherotyping. Rotaviruses have a genome consisting of
sucrose, the sucrose must be washed away by discarding the double-stranded RNA with 11 segments. When the genome is
supernatant (containing the sucrose), resuspending the pellet in extracted from the virus and subjected to electrophoresis on
PBS, and pelleting as before. polyacrylamide gel, the resulting pattern of bands is referred to as
5) Resuspending the sample preparation. Following the virus' electropherotype (see below for procedure). The various
ultracentrifugation, die supernatant is discarded, and the pellet can be groups of avian rotaviruses have distinctive electropherotypes
resuspended in sterile high-quality water or tris buffer (pH 7.2). (banding patterns). Slight variations of avian rotaviruses within a
Note that the ultracentrifuge tubes need to be marked before group have been observed. This is generally not detected by
pelleting so as to locate the pellet, since a visible pellet is serologic assays but is detected by polyacrylamide gel electrophoresis.
rarely identifiable. The pellet should be resuspended in 1 drop Polyacrylamide Gel Electrophoresis of Avian Rotavirus RNA.
(from a Pasteur pipet) of the chosen diluent. The following procedures are used:
161
Don Reynolds and Ching Ching Wu

1) Preparation of virus. The ability to distinguish visible bands in the centrifuged as before. The top layer contains the extracted RNA and
final gel may depend on the amount of background material present in can be used for electrophoresis. Additional techniques to purify
the virus preparation. Although there are procedures that will assist the RNA may include cold ethanol precipitation, and CF 11 cellulose
in concentrating and purifying the viral RNA (see below), depending chromatography; see Theil (40,41).
on the situation, it may be advantageous to purify the virus before RNA 3) Electrophoresis of RNA. The amount of sample needed and the
extraction in order to decrease unwanted background. Intestinal samples conditions that can be used during electrophoresis are largely
can be processed as described above (see sample collection). The dependent on the method and type of equipment being used.
resulting sonicated filtrate can be used or further purified by Standard-sized gels, mini gels, and Phast® gels (Pharmacia Co,
fluorocarbon extraction. Fluorocarbon extraction can be Piscataway, NJ) have been used successfully. Conventional
accomplished by mixing the sample with cold (4 C) 1,1,2- standard-size gels, using a vertical gel apparatus, generally require 20
trichloro-l,2,2-trifluoroethane (Freon) and homogenizing it μΐ of sample, whereas the Phast® system requires 2 μΐ.
with a high-speed mixer for 2 to 3 min. The resulting mixture can be Conventional vertical gels can be run for 4 to 5 hr at 40
centrifuged at 300 to 500 x g to separate the organic from the aqueous mA using 7.5% polyacrylamide gels. Phast® gels can be run
phase. The aqueous phase (top layer) contains virus. The virus at 12 mA for 175 volt-hours (about 1 h) using 10-15%
preparation can be further purified by ultracentrifugation techniques polyacrylamide gradient gels. These two methods are a contrast
such as pelleting the virus through a layer of sucrose (see above) or between the largest/slowest and smallest/fastest techniques that
using isopycnic separation. will allow for reasonable results. The method and parameters used
2) RNA extraction. RNA can be extracted from the virus must be determined by the user.
preparation by first suspending the virus preparation in a reducing 4) Staining the gel. The staining method most commonly used for
buffer (2% sodium dodecyl sulfate, 20% glycerol, 4% 2- dsRNA viruses is the silver staining method. For further information,
mercaptoethanol, 30% urea in tris-glycine buffer). The preparation see Theil (40, 42) and Phastsystem development technique file No.
can be centrifuged and the supernatant retained. Phenol-chloroform 210 available from Pharmacia.
(3:2) is added to the supernatant and mixed. This mixture is

Toroviruses

PREFERRED CULTURE MEDIA AND SUBSTRATES SEROLOGIC DETECTION IN THE HOST

There is currently no cell culture system to support Torovirus Dot-immunobinding avidin-enhanced ELISA
growth, and the virus cannot be propagated in chicken embryos, but Toroviruses react specifically in this assay with anti-sera from
can be propagated by inoculating 24-25 day-old turkey embryos by turkeys infected with torovirus but not with antisera against a range
the amniotic route (1). of common veterinary pathogens such as bovine coronavirus, avian
IBV , Newcastle disease virus, and transmissible gastroenteritis
AGENT IDENTIFICATION virus of swine (2,3). Likewise the following viral agents have been
found not to react with anti-serum from turkeys infected with
Stunting syndrome is an enteric disease of turkeys causing torovirus: turkey coronavirus, bovine coronavirus, bovine Breda-1
diarrhea, reduced weight gain, poor feed efficiency, and virus, bovine Breda-2 virus, avian IBV, avian influenza virus,
malnutrition. Infection usually occurs around 1-3 wk of age. Newcastle disease virus, and transmissible gastroenteritis virus of
Chickens are refractory to infections by turkey torovirus, this swine (2,3).
characteristic aids in differentiating them from other enteric
pathogens such as TCoV. Torovirus has been proposed to be the Virus neutralization
stunting syndrome agent (SSA) based on infection of turkey A virus neutralization assay has been developed in 24-25 day old
embryos following amniotic inoculation with filtrate from intestinal turkey embryos. Virus infection was assayed according to the
cultures passed through a 0.1 micron filter. Electron microscopy of presence of intestinal lesions and by maltase release assay (1).
intestinal fluid and epithelial cell lysate has identified pleomorphic Briefly, turkey embryos were inoculated with turkey torovirus plus
enveloped virus particles of 60-95 nm diameter (2, 3). Loss of antisera via the amniotic cavity at 24-25 days of incubation. The
infectivity with ether treatment confirms that the virus is enveloped. jejunal maltase activity of the torovirus-inoculated turkey embryos
The viral genome is thought to be positive-sense, single-stranded and the intestinal lesions (intestines from the infected turkey
RNA due to the sensitivity of genomic nucleic acid to RNAse embryos were pale, thin walled, and distended with fluid) were used
treatment, and amplification of genome by reverse-transcriptase- as indicators of viral infection (or lack thereof = viral neutralization)
polymerase chain reaction but not by polymerase chain reaction. when compared to positive control embryos (virus only, no antisera,
Torovirus haemagglutinates rat erythrocytes at 4 C and at room contained in the inoculum) and negative control embryos (no virus,
temperature. PBS only in the inoculum).
Fluorescent antibody assay. A FA assay has been developed for
detecting Torovirus in intestinal tissues (33). The FA assay uses DIFFERENTIATION FROM CLOSELY RELATED AGENTS
convalescent antiserum from turkeys inoculated with SSA. There
has also been an indirect fluorescent antibody (IFA) assay The enteric viruses can be differentiated from each other using the
developed for detecting serum antibodies to SSA. SSA can be characteristics and techniques described above. Additionally, other
detected in experimental infections between 2-3 days PI. Peak FA nonviral agents must also be considered when attempting to diagnose
scores were seen 3 days PI and began to wane by 4 days PI. enteric diseases in young birds. Both infectious and noninfectious
Specificity was 95% and sensitivity was 100%. Seroconversion to agents need to be considered. Examples of infectious agents include
SSA starts at around 7 days PI and all birds have seroconverted by protozoal agents (e.g., Eimeria, Cryptosporidia, Hexamita) and
12 days PI. bacterial agents (e.g., Salmonella spp.). Examples of noninfectious
agents may include toxins, and nutrients (deficiencies and excesses).
Enteric disease is a complex disease entity that may have a complex
etiology. Therefore, numerous agents (both viral and nonviral) may be
involved.
162
Chapter 34 Enteric Viruses

REFERENCES 26. Loa, C. C., T. L. Lin, C. C. Wu, T. A. Bryan, T. Hooper, and D.


Schrader. Expression and purification of turkey coronavirus nudeocapsid
1. Ali, A., and D. L. Reynolds. The in vitro propagation of stunting protein in Escherichia coli. J. Virol. Methods 116:161-167. 2004.
syndrome agent. Avian Dis. 42:657-666. 1998. 27. Loa, C. C., T. L. Lin, C. C. Wu, T. A. Bryan, T. Hooper, and D.
2. Ali, A., and D. L. Reynolds. Characterization of the stunting syndrome Schrader. Differential detection of turkey coronavirus, infectious bronchitis
agent: physicochemical properties. Avian Dis. 44:426-433. 2000. virus, and bovine coronavirus by a multiplex polymerase chain reaction. J.
3. Ali, A., and D. L. Reynolds. Characterization of the stunting syndrome Virol. Methods 131:86-91. 2006.
agent: relatedness to known viruses. Avian Dis. 44:45-50, 2000. 28. Loa, C. C., T. L. Lin, C. C. Wu, T. A. Bryan, T. Hooper, and D.
4. Barnes, H. J., and J. S. Guy. Poult enteritis-mortality syndrome. In: Schrader. Comparison of 3’ End Encoding Regions of Turkey Coronavirus
Diseases of Poultry, 11th ed. Y. M Saif, H. J. Barnes, J. R. Glisson, A. M Isolates from Indiana, North Carolina, and Minnesota. Intervirol. 49:230-
Fadly, L. R. McDougald, and D. E. Swayne, eds. Iowa State Press, Ames. Iowa, 238. 2006.
pp. 1171-1180. 2003. 29. McNulty, M. S. Viral enteric infections. Rotavirus infections.
5. Baxendale. W, and T. Mebatsion. The isolation and characterisation of In: Diseases of Poultry,
astroviruses from chickens. Avian Pathol. 33:364-370. 2004. 11th ed. Y. M Saif, H. J. Barnes, J. R. Glisson, A. M Fadly, L. R. McDougald,
6. Behling-Kelly, E., S. Schultz-Cherry, M Koci, L. Kelley, D. Larsen, and and D. E. Swayne, eds. Iowa State Press, Ames. Iowa. pp. 308-320. 2003.
C. Brown. Localization of astrovirus in experimentally infected turkeys as 30. McNulty, M. S., and J. S. Guy. Viral enteric infections.
determined by in situ hybridization. Vet. Pathol. 39:595-598, 2002. Avian enteroviruslike viruses. In: Diseases of Poultry, 11th ed. Y. M
7. Breslin, J. J., L. G. Smith, H. J. Barnes, and J. S. Guy. Comparison of Saif, H. J. Barnes, J. R. Glisson, A. M Fadly, L. R. McDougald, and D. E.
virus isolation, immunohistochemistry, and reverse transcriptase-polymerase Swayne, eds. Iowa State Press, Ames. Iowa pp. 308-320.2003.
chain reaction procedures for detection of turkey coronavirus. Avian Dis. 31. McNulty, M S., W. L. Curran, D. Todd, and J. B. McFerran. Detection
44:624-631. 2000. of viruses in avian faeces by direct electron microscopy. Avian Pathol.
8. Breslin, J. J., L. G. Smith, and J. S. Guy. Baculovirus expression of 8:239-247. 1979.
turkey coronavirus nucleocapsid protein. Avian Dis. 45:136-143. 2001. 32. Patel, B.L., D. R. Deshmukh, and B. S. Pomeroy. Fluorescent antibody
9. Dea, S., G. Marsolais, J. Beaubien, and R. Ruppanner. Coronaviruses test for rapid diagnosis of coronaviral enteritis of turkeys (bluecomb). Am. J.
associated with outbreaks of transmissible enteritis of turkey in Quebec: Vet. Res. 36:1265-1267. 1975.
hemagglutination properties and cell cultivation. Avian Dis. 30:319-326. 33. Reynolds, D. L., J. Oesper, A. Ali. The fluorescent antibody and
1986. indirect fluorescent antibody assays for diagnosing stunting syndrome of
10. Dea, S., and P. Tijssen. Identification of the structural proteins of turkey turkeys. Avian Dis 44:313-317. 2000.
enteric coronavirus. Arch. Virol. 99:173-186,1988. 34. Reynolds, D. L., and S. L. Schultz-Cherry. Astrovirus infections. In:
11. Dziuk, Η. E., O. A. Evanson, and C. T. Larson. Physiologic effects of Diseases of Poultry, 11th ed. Y. M Saif, H. J. Barnes, J. R. Glisson, A. M
fasting and bluecomb in turkeys. Am. J. Vet. Res. 30:1045-1056. 1969. Fadly, L. R. McDougald, and D. E. Swayne, eds. Iowa State Press, Ames. Iowa,
12. Gardner, P. S. Immunofluorescence. In: Clinical pp. 320-326. 2003.
Virology Manual. S. Specter and G. J. Lancz eds. Elsevier 35. Saif, Y. M. Viral enteric infections. Introduction. In: Diseases of
Science Publishing Co., New York. pp. 95-109. 1986. Poultry, 11th ed. Y. M Saif, H J. Barnes, J. R. Glisson, A. M Fadly, L. R.
13. Guy, J. S., L. G. Smith, J. J. Breslin, and S. Pakpinyo. Development of a McDougald, and D. E. Swayne, eds. Iowa State Press, Ames. Iowa. pp. 299-
competitive enzyme-linked immunosorbent assay for detection of turkey 300. 2003.
coronavirus antibodies. Avian Dis. 46: 334-441. 2002. 36. Saif, L. J., Y. M Saif, and K. W. Theil. Enteric viruses in diarrheic
14. Guy, J. S., A. M Miles, L. Smith, F. J. Fuller, and S. Schultz-Cherry. turkey poults. Avian Dis. 29:798-811. 1985.
Antigenic and genomic characterization of turkey enterovirus-like virus 37. Sellers, H. S., M D. Koci, E. Linnemann E, L. A. Kelley, S. Schultz-
(North Carolina, 1988 isolate): identification of the virus as turkey astrovirus Cherry. Development of a multiplex reverse transcription-polymerase chain
2. Avian Dis. 48:206-11. 2004. reaction diagnostic test specific for turkey astrovirus and coronavirus. Avian
15. Hayhow, C. S., and Y. M. Saif. Development of an antigen-capture Dis. 48:531-539, 2004.
enzyme-linked immunosorbent assay for detection of enterovirus in 38. Spackman, E., D. Kapczynski, and H. Sellers. Multiplex real-time
commercial turkeys. Avian Dis. 37: 375-379. 1993. reverse transcription-polymerase chain reaction for the detection of three
16. Hayhow, C. S., Y. M Saif, K. MKerr, and R. E. Whitmoyer. Further viruses associated with poult enteritis complex: turkey astrovirus, turkey
observations on enterovirus infection in specific-pathogen-free turkey coronavirus, and turkey reovirus. Avian Dis. 49:86-91. 2005.
poults. Avian Dis. 37. 124-134. 1993. 39. Tang, Y., and Y. M Saif. Antigenicity of two turkey astrovirus isolates.
17. Ito, Η., M Sugiyama, K. Masubuchi, Y. Mori, and N. Minamoto. Avian Dis. 48:896-901. 2004.
Complete nucleotide sequence of a group A avian rotavirus genome and a 40. Theil, K. W. A modified genome electropherotyping procedure
comparison with its counterparts of mammalian rotaviruses. Virus Res. for detecting turkey rotaviruses in small volumes of intestinal contents.
75:123-38. 2001. Avian Dis. 31:899-903. 1987.
18. Koci, M D., B. S. Seal, and S. Schultz-Cherry. Molecular 41. Theil, K. W., D. L. Reynolds, and Y. M. Saif. Isolation and
characterization of an avian astrovirus. J. Virol. 74:6173-6177. 2000. serial propagation of turkey rotaviruses in a fetal rhesus monkey kidney
19. Koci, M D., B. S. Seal, and S. Schultz-Cherry. Development of an RT- (MA104) cell line. Avian Dis. 30:93-104. 1986.
PCR diagnostic test for an avian astrovirus. J. Virol. Methods 90:79-83. 42. Theil, K. W., D. L. Reynolds, and Y. M. Saif. Genomic variation
2000. among avian rotavirus-like viruses detected by polyacrylamide gel
20. Koci, M D., S. Schultz-Cherry. Avian astroviruses. Avian Pathol. electrophoresis. Avian Dis. 30:829-834. 1986.
31:213-227. 2002. 43. Wani, S. A., M A. Bhat, S. M Ishaq, M A. Ashrafi, A. S. Buchh, and
21. Lin, T. L., C. C. Loa, S. C. Tsai, C. C. Wu, T. A. Bryan, H. L. Thacker, M Haq. Detection of a mammalian-like group A rotavirus in diarrhoeic
T. Hooper, and D. Schrader. Characterization of turkey coronavirus from chicken. Vet. Microbiol. 94:13-18. 2003.
turkey poults with acute enteritis. Vet. Microbiol. 84:179-186. 2002. 44. Yason, C. V., and K. A. Schat. Isolation and characterization of avian
22. Lin, T. L., C. C. Loa, C. C. Wu, T. A. Bryan, T. Hooper, and D. rotaviruses. Avian Dis. 29: 499-508. 1985.
Schrader. Antigenic relationship of turkey coronavirus isolates from 45. Velayudhan, B. T., H. J. Shin, V. C. Lopes, T. Hooper, D. A.
different geographic locations in the United States. Avian Dis. 46:466-472. Halvorson, and K. V. Nagaraja. A reverse transcriptase-polymerase chain
2002. reaction assay for the diagnosis of turkey coronavirus infection. J. Vet.
23. Lin, T. L., C. C. Loa, and C. C. Wu. Complete sequences of 3’ end Diagn. Invest. 15:592-596. 2003.
coding region for structural protein genes of turkey coronavirus. Virus Res. 46. Wu, C.C., M Abdelwahab, C. C. Loa, T. A. Bryan, T. Hooper,and T. L.
106:61-70. 2004. Lin. Recombinant partial turkey coronavirus spike protein in antibody­
24. Loa, C. C., T. L. Lin, C. C. Wu, T. A. Bryan, H. L. Thacker, T. Hooper, capture enzyme-linked immunosorbent assay for detection and
and D. Schrader. Detection of antibody to turkey coronavirus by antibody­ differentiation of antibody to turkey coronavirus. In: Proceedings of the 56th
capture enzyme-linked immunosorbent assay utilizing infectious bronchitis North Central Avian Disease Conference. St. Paul, Minnesota, March, 2005.
virus antigen. Avian Dis. 44:498-506. 2000. p.80.
25. Loa, C. C., T. L. Lin, C. C. Wu, T. A. Bryan, H. L. Thacker, T. Hooper,
and D. Schrader. 2001. Humoral and cellular immune responses in turkey
poults infected with turkey coronavirus. Poultry Sci. 80:1416-1424. 2001.

163
35
ONCORNAVIRUSES
LEUKOSIS/SARCOMAS AND RETICULOENDOTHELIOSIS
Aly M. Fadly, Richard L. Witter and Henry D. Hunt

SUMMARY. Avian leukosis virus (ALV), and reticuloendotheliosis virus (REV) are the most common naturally occurring avian
oncornaviruses associated with neoplastic disease conditions in poultry. ALV infects primarily chickens, whereas REV infects chickens,
turkeys, ducks, geese, pheasants, quail, and probably many other avian species. In addition to causing tumors, both ALV and REV can reduce
productivity and induce immunosuppression and other production problems in affected flocks. No vaccines that protect chickens and turkeys
from infection with ALV or REV are available commercially.
Agent Identification A number of biological assays can be used for the detection of ALV and REV infection. Virologic and serologic
criteria, such as assays for virus, antigens or antibodies are very useful in identification and classification of new isolates, surveillance of
pathogen-free and other breeder flocks for freedom of virus infection, establishing infection with one virus and excluding others, and in
safety testing of vaccines. However, such virologic and serologic assays are not particularly helpful in the differential diagnosis of virus-
induced neoplastic diseases of poultry because avian oncogenic viruses are ubiquitous and infection in the absence of tumor formation is
common. Current technology based on molecular hybridization or immunocytochemistry with monoclonal antibodies to cellular and tumor
specific antigens is being used to develop more sensitive and specific procedures for the differential diagnosis of virus-induced avian
neoplasms.
Serological Detection in the Host. Antibodies to ALV can be detected using virus neutralization, Rous Sarcoma Virus-focus reduction
assay or micro-neutralization ELISA. For REV, IFA, flow cytometry, ELISA, AGP, and VN can be used.

INTRODUCTION development of LL in certain lines of chickens following exposure


to ALV after hatch (3,21).
Lymphoid leukosis (LL), formerly called "big liver disease" or Natural infection with REV seldom results in clinical disease in
"visceral lymphomatosis," is caused by ALV. It is the most chickens. In turkeys, mortality first occurs between 12 and 20 wk of
common naturally occurring neoplastic disease of chickens induced age. Lymphomas are found in the liver, intestine, heart, and other
by the leukosis/sarcoma (L/S) group of avian oncornaviruses (19). visceral organs. Nerves are occasionally enlarged (62). The
In early 1990’s, a novel subgroup (J) ALV was found to be neoplastic cells are large and uniform, but their lineage has not been
associated with a relatively high incidence of myelocytomatosis in characterized. However, in a recent outbreak of RE in commercial
meat-type chickens (36). ALV is transmitted congenitally or by turkeys, the tumor cells were identified as T-cell lymphomas (8). In
contact and occurs worldwide in almost all chicken flocks. Stress or chickens inoculated at hatching with biologicals contaminated with
immunodepression can increase the susceptibility of chickens to REV, a runting disease may develop within 2-3 wk: it is
ALV infection and shedding. characterized by bursal and thymic atrophy, retarded growth,
Reticuloendotheliosis (RE) designates a group of disease abnormal feather development, and severe immunodepression. An
syndromes caused by the REV group of avian oncornaviruses (62). outbreak of lymphomas was reported in broiler breeder flocks that
The most common clinical diseases are chronic lymphomas and a had been vaccinated at 6 days of age with a fowlpox vaccine
runting disease. However, an acute reticulum cell neoplasia can be contaminated with REV (22).
induced by a laboratory strain (strain T) that is a mixture of an Experimental infection of chickens with REV induces two types of
oncogene-containing replication-defective virus and a nondefective lymphomas: a bursal lymphoma of B-cell origin that is
helper virus (59). Infection of chicken flocks with REV is pathologically identical to lymphoid leukosis, and a nonbursal
moderately prevalent, but clinical disease is rarely recognized. The lymphoma of T-cell origin that superficially resembles that of MD
virus is transmitted both vertically and horizontally, and by (62). The response varies greatly with the strain of virus, dose, route
inoculation with contaminated biologicals. of exposure, and species of bird. Because the lesions are usually not
Comprehensive reviews on L/S and REV groups of avian pathognomonic, a diagnosis of clinical disease must be based on
oncornaviruses are available (19, 62). pathologic criteria plus virologic or serologic evidence of infection.
Serotype 2 MD vaccines were also found to enhance the
CLINICAL DISEASE development of REV-induced B-, but not T-cell lymphoma (2).

In addition to LL and sarcomas, the L/S viruses induce a wide SAMPLE COLLECTION
range of neoplastic conditions, such as osteopetrosis,
hemangiomas, erythroblastosis, nephroblastomas, myeloblastosis, Avian Leukosis
and myelocytomatosis. The type of tumor is influenced by the Because ALV is ubiquitous in the chicken population, virus
strain of virus; exposure dose; host genotype; and route, sex, and isolation and the demonstration of antigen or antibody have limited
age at exposure (19). LL, a B-cell lymphoma of chickens, originates or no value in diagnosing field cases of LL. However, testing for
in the follicles of the bursa of Fabricius. Transformed bursa cells virus, antigen, or antibody is and has been instrumental in programs
metastasize to the liver, spleen, and other visceral organs (15). for reducing or eradicating ALV infection. Various materials can
Because the incubation period is relatively long (rarely less than 14 be used to detect virus, antigen, or antibody. Because L/S viruses
wk), LL is usually a neoplastic disease of breeders and commercial are thermolabile it is imperative that materials used for biological
egg-layers, but not of broilers, roasters, or fryers. The clinical signs assays for infectious virus be placed on melting ice immediately
of LL are not specific (19). In advanced cases, enlargement of the after collection and then stored at -70 C until assayed. Samples for
bursa and liver may be detected by palpation. Lesions in liver and detection of ALV group-specific (gs) antigens are usually collected
other visceral organs may be diffuse or nodular. Microscopically, in the buffer that is used in a particular test and stored at -20 C.
LL is characterized by intrafollicular infiltration of bursal follicles Samples to be tested by the enzyme-linked immunosorbent assay
with tumor cells (19). Marek’s disease (MD) vaccines containing (ELISA) are collected in phosphate-buffered saline (PBS)
serotype 2 MD herpesvirus (65) were found to enhance the containing 0.1% Tween 80, whereas samples to be tested by
164
Chapter 35 Oncornaviruses Leukosis/Sarcomas and Reticuloendotheliosis

complement fixation (CF) test are collected in barbital or veronal Tumors removed aseptically should be tested fresh or frozen at or
buffer. below -70 C. A 5-10% tumor homogenate in PBS or TC medium
Whole Blood, Plasma, or Serum. Whole blood or plasma are can be used for virus isolation or for detection of gs antigen.
most commonly used for virus isolation. Blood can be drawn in However, isolation of ALV from a tumor does not necessarily
sterile syringes containing 50 units of preservative-free heparin/ml. suggest a causal relationship between the virus and the tumor, as all
Heparin may interfere with the replication of ALVs other than commercial flocks and most chickens in a flock are exposed to
subgroup A. Other anticoagulants (such as 10% [vohvol] of a 3.5% ALV. Histopathology is the most commonly used procedure to
sodium citrate solution) may be used, or blood for serum can be diagnose and differentiate tumors.
collected in tubes containing 0.1-0.2 ml of 25% heat-inactivated Other tissues, such as comb from newly hatched chicks and feather
chicken embryo extract (from embryos known to be free from tips, can be used for detection of gs antigen, although testing of
ALV) to enhance clotting. Blood should be placed on melting ice as these materials has been compared with other test procedures only
soon as possible after clotting. Whole blood should be shipped on on a limited scale (10). Feather tips are collected in tubes containing
ice-cold packs immediately after collection and should not be the buffer to be used in the assay and then disrupted with a tight-
frozen. Samples of plasma or serum for virus isolation should be fitting Teflon pestle. The homogenate should be sonicated or frozen
shipped on dry ice. To avoid interference by neutralizing antibodies, and thawed three times before testing. Liver and magnum of
ALV should preferably be isolated from fresh samples of oviduct collected from freshly killed chickens are considered to be
leukocytes or whole blood. If delay is necessary, tissue culture (TC) rich sources of virus (19). Also, recently, feather tips have been
medium containing 10% dimethylsulfoxide and 15% bovine serum shown to contain a high titer of ALV (56).
should be added to the samples, and then the samples should be
slowly frozen and stored at -196 C. Reticuloendotheliosis
Plasma or serum samples are unsuitable for detection of The preferred samples for REV isolation are preparations of whole
exogenous ALV by testing for gs antigen, as endogenous gs antigen blood, fresh spleen, and tumor tissue containing intact live cells
may lead to false-positive results (37). from birds of any age with suspect lesions. Cellular inocula can be
Meconium and Cloacal or Vaginal Swabs. Meconium from 1- stored for several hours at 4 C or for longer periods at -70 C or -196
day-old chicks and cloacal or vaginal swabs from older chickens C after slow freezing in TC medium containing 15% bovine serum
can be used to isolate virus or to detect gs antigen (19). Cloacal and 10% dimethylsulfoxide. Although virus can be recovered from
swabs are obtained by rotating a sterile cotton swab about five times plasma and tissue extracts, such cell-free samples are inferior to
in the cloaca. Care should be taken to make sure that the mucosal cellular preparations because the virus is relatively labile and the
surface of the cloaca is swabbed and not to allow excess fecal titers of such preparations are often tow. Whole blood is collected
material to adhere to the swab. Vaginal swabs are taken by everting with heparin (50 units/ml) and used undiluted. Because of the ease
the cloaca as for artificial insemination.Vaginal swabs are not used with which it is collected and handled, whole blood or blood
to test non-laying hens because the cloaca of such hens is not lymphocytes are usually the tissue of choice for virus isolation.
readily everted. For virus isolation, the swab is placed in a tube For the isolation of REV from tissues, about 1-2 g of spleen or
containing TC medium supplemented with 1000 units of penicillin- tumor tissue is collected aseptically, placed in 10-20 ml of TC
G and streptomycin sulfate, 100 pg gentamicin sulfate, and 5 pg medium, gently minced with scalpels or sharp scissors, passed
amphotericin B per ml. Meconium for virus isolation can be repeatedly through the barrel of a 10-ml syringe, and filtered
collected by placing a tube containing 1 ml of such TC medium at through two layers of sterile gauze. The filtrate is then centrifuged,
the cloacal opening and gently squeezing the abdomen. Before and one volume of packed cells (pellet) is resuspended in about nine
testing, solid materials should be removed from samples of volumes of TC medium.
meconium by centrifugation at 2000 rpm for 10 min.
Albumen. Albumen can be used for detection of virus or gs PREFERRED CULTURE MEDIA AND SUBSTRATES
antigen (55). Testing albumen for gs antigen is the most sensitive
and practical means of identifying hens that are likely to transmit Avian Leukosis
ALV congenitally. Samples of thin albumen can be drawn with a Cell Culture. Unlike sarcoma viruses, most ALVs produce no
syringe or Pasteur pipette through a hole punched into the small end visible morphologic changes in culture. Thus, indirect biologic
of an unincubated egg. For virus assays, albumen should be assays such as complement fixation (CF) for avian leukosis
collected from eggs within 48 h of laying. In most cases, hens are (COFAL) (46), enzyme-linked immunoabsorbent assay (ELISA) for
screened for ALV shedding by testing albumen for gs antigen from ALV (ELISA-ALV) (9, 51), phenotypic mixing (PM) (35),
two to three consecutively laid eggs per hen. However, identifying resistance-inducing factor (RIF) (42), and non-producer (NP) cell
intermittent shedder hens may depend on the frequency of testing activation (41) tests are used for the detection of ALVs. ELISA-
and the number of eggs tested per hen. If eggs are to be hatched, 0.3 ALV is the more commonly used test for detection of ALV, because
ml of albumen can be collected. Techniques should be aseptic, and it is more sensitive and less cumbersome than other tests such as the
the hole should be sealed with paraffin wax, adhesive tape, or COFAL and PM tests. These assays require the use of chicken
model cement. To facilitate work with albumen, it should be diluted embryo fibroblasts (CEFs) with specific host range (Table 1). The
two-to-four-fold in the buffer to be used in the test. ALV gs antigen COFAL and ELISA-ALV assays are performed in two phases: 1)
is stable in egg albumen for at least 63 days at 8 C. propagation of ALV in CEFs; and 2) detection of ALV gs antigen
Chicken Embryos. Virus recovery from extracts of chicken by COFAL or by ELISA-ALV.
embryos is closely associated with congenital transmission of ALV. In addition to the standard ELISA and CF test reagents obtainable
However, samples of albumen, blood, and cloacal or vaginal swabs from several commercial sources, both tests require the following
are easier to handle than embryos. Embryo extracts are prepared by specific reagents: 1) ALV-susceptible CEFs that are free from
expressing 9-to-l 1-day-old embryos through a 5-ml syringe and endogenous ALV and gs antigen; 2) reference stock of gs antigen,
into 1 ml of PBS. The suspension is frozen and thawed three times which may be obtained from CEFs infected with ALV or Rous
and then clarified by centrifugation at 2000 rpm for 20 min. The sarcoma virus (RSV); 3) highly specific antisera to L/S viral gs
supernatant is used for virus isolation or for detection of gs antigen. antigen prepared in hamsters or rabbits (19), (monoclonal
Tumors and Other Tissues. Some tumors may contain large antibodies against p27 have been shown to enhance specificity and
amounts of virus, but others may contain no detectable virus. sensitivity of the test used to detect gs antigen [14]); and 4) known

165
Aly M. Fadly, Richard L. Witter and Henry D. Hunt

Table 35.1 Phenotype of CEFs used for detection of exogenous and endogenous AL Vs.
Susceptibility to L/S subgroups
CEFs/ line PhenotvneA Susceptible Resistant to
0 C/E A,B,C,D,J E
15B1 C/O A B, C, D, J, E
alv6B C/AC/E B, C, D, J AE
31V6X15B, C/A B, C, D,J ,E A
DF-l/J C/E,C/J A B, C, D E,J
RFS
** C/O A B, C, D, E, J
aC/E = cells resistant to subgroup E ALVs; C/O = cells susceptible to all subgroups of ALVs.
C/A = cells resistant to subgroup A ALVs; DF-l/J = cells resistant to subgroup E and J ALVs (28).
b= Because alphas line 0 background, cells are also resistant to subgroup E (endogenous) ALV.

*♦= Rapid-Feather Susceptible line (RFS), it is susceptible to all subgroups, but lacks all endogenous virus genes.

ALV or RSV seed stock. Some of these reagents can be obtained or ELISA (see section on Preferred Culture Media and Substrates).
from commercial sources (see appendix). Based on properties of viral envelope glycoproteins, members of
L/S group including ALVs from chickens are classified into six
Reticuloendotheliosis subgroups, A, B, C, D, E, and J (19). Unlike the acute leukemia
Cell Culture. Most methods for diagnosis of REV infection by viruses of L/S group, exogenous ALVs (belonging to subgroups A,
virus isolation are designed to recover nondefective viruses. This B, C, D and J) and endogenous ALVs (subgroup E) are not
can be accomplished by propagation of cellular inocula for 9-14 defective and lack host oncogenes (19). Among the structural
days in avian fibroblast cultures and subsequent demonstration of polypeptides (p27, pl9, pl5, pl2, and plO) shared by all members
viral antigens or the enzyme reverse transcriptase. of die L/S group of avian oncornaviruses including endogenous and
Most, and perhaps all, field strains of REV can be easily exogenous ALVs, p27 is the most abundant. Techniques for
propagated in avian cell cultures. Secondary cultures of CEFs are detecting ALV infection have largely been based on assays for virus
normally used, although cultures of chicken kidney, duck embryo or viral antigen rather than on assays for antibody. In addition to
fibroblasts, turkey embryo fibroblasts, or quail embryo fibroblasts ELISA and CF tests, immunocytochemical staining procedures such
are also suitable. Because REV can be transmitted vertically, as immunofluorescence, immunoperoxidase anti-peroxidase, or
embryos should be from breeders free of infection. The QT-35 cell protein A-gold can be used to detect gs antigen associated with
line, composed of chemically transformed quail fibroblasts, is also ALV particles in sections of various tissues (19, 26). These
susceptible to infection (7). procedures also require the use of specific anti-p27 IgG.
For primary isolation, undrained cultures in 35 or 60-mm dishes Direct ELISA or the CF test can be used for detection of gs
with confluent monolayers are inoculated, usually within 6-24 h antigen in samples of albumen, cloacal swabs, meconium, whole
after plating, with 0.1 ml of a cellular test sample; cell-free samples blood, or feather pulp. ELISA is preferred over the CF test for
should be inoculated onto drained cultures to which medium is testing virus-rich samples such as cloacal swabs, meconium, or
added after adsorption for 30 min. The cultures are maintained for blood that are unsuitable for testing by CF because of
7-9 days at 38 C with the first medium change at 24 h and anticomplement (AC) activity (18).
subsequent changes at 2-to-3-day intervals. A second passage is Reticuloendotheliosis. The properties of REV are typical of
accomplished by placement of supernatant fluid into a new culture oncornaviruses (62). The virions are about 100 nm in diameter and
maintained in a 35-mm dish (0.5 ml per plate) or a 96-well plate bud from the plasma membrane. The particles band at 1.16-1.18
(0.1 ml per well), which is maintained for 5-7 more days. Controls g/cm3 in sucrose gradients. The RNA genome of replication
include cultures inoculated with a prototype nondefective virus such competent strains is about 9.0 kilobases. Several polypeptides have
as strains T or CS, and uninoculated cultures. It is prudent to use been isolated, including two glycoproteins (gp90 and gp20) and five
several uninoculated cultures spaced randomly among test cultures others (plO, pl2, ppl8, pp20, and p30), of which p30 is the major
to monitor adventitious spread of REV, which can be a problem if gs antigen. The particles also contain a reverse transcriptase.
cultures are not handled with care.
Biological Identification
AGENT IDENTIFICATION Avian Leukosis Enzyme-Linked Immunosorbent Assay
for Avian Leukosis Virus The ELISA-ALV consists of two phases;
Physiochemical Properties virus propagation in CEFs and assay for ALV presence by testing
Avian Leukosis Avian L/S viruses contain a core of two single­ supernatant fluid for gs antigen by ELISA.
stranded RNA copies (19). They are about 60% protein by weight For propagation of exogenous ALVs (subgroups A, B, C, D and J),
and are made up of internal protein, reverse transcriptase, and CEFs resistant to subgroup E ALV (C/E) are used. For assays of
envelope proteins. About 35% is lipid derived from the cell endogenous viruses, cells that are susceptible to infection with
membrane and 4% carbohydrate associated with envelope proteins. subgroup E ALV are used.
The specific gravity of the virus is 1.16. These viruses are sensitive 1) Suspend cells in TC medium at a concentration of 2.5 X 10^/ml.
to lipid solvent and low pH and are highly resistant to ultraviolet 2) To the cell suspension, add DEAE-Dextran, or polybrene (see
radiation (19). Because the L/S viruses are thermolabile, long-term appendix) to a final concentration of 2 pg/ml.
storage without loss of infectivity is possible only below -60 C. 3) Transfer 2 ml of cell suspension to 35-mm plates; other suitable
Morphologically, the avian L/S viruses are classified as C-type culture dishes, such as 24 or 48-well microtiter TC plates, can be
particles with a diameter 80 to 100 nm. Members of the avian L/S used.
virus group cannot be differentiated on the basis of ultrastructure.
Avian L/S viruses contain a serologically unique reverse
transcriptase and a group specific (gs) antigen demonstrable by CF
166
Chapter 35 Oncornaviruses Leukosis/Sarcomas and Reticuloendothebosis

4) Test one 35-mm plate per sample, plus one plate per dilution of that does not detect RSV(RAV-0), i.e., in cells genetically resistant
1 6 to subgroup E L/S virus (35).
control ALV being titrated (usually six plates; 10’ to 10’ ) of the
prototype ALV, such as RAV-1 in the case of testing subgroup A Complement Fixation for Avian Leukosis. The COFAL
viruses. In the case of testing unknown samples, two plates for each test is performed in two stages: virus propagation and assay of ALV
of other subgroups (B, C, D and J) of ALV are also included. gs antigen by the CF test.
5) On the next day, add 100 μΐ of sample being tested to TC For propagation of exogenous ALV, primary or secondaiy C/E
medium in the corresponding plate; samples can also be added to CEFs lacking endogenous viruses are used. For assaying ALV gs
the plates on the same day the cells are cultured. Stir inoculum into antigen by CF test, hemolysin, guinea pig complement, and
medium by gently rotating each tray holding plates. antiserum to ALV gs antigen are required. The procedures for
6) Change medium 24 h after samples are added. titration of hemolysin, complement, and antiserum and for running
7) Maintain cultures for 7-9 days after inoculation. A second change the COFAL test have been described (46, 60).
of medium is not necessary if cultures are maintained for 7 days Resistance-Inducing-Factor Test. When susceptible
only. CEFs are all infected with ALV, the cells become resistant to
8) If the objective of the assay is to isolate virus, collect 1 ml of transformation induced by RSV of the same subgroup as the ALV
supernatant fluid from all test plates into sterile tubes and save at - (42). This property of interference has been used extensively for
70 C until results of ELISA are available. assay of ALV by the RIF test and also in determining the virus
9) Add 80 μΐ of 5% Tween-80 to each 35-mm plate; freeze and subgroup (19). The presence of ALV is indicated when the number
thaw cell suspension two times before testing for ALV gs antigen of foci induced by a standard stock of RSV on test cells is a tenth or
by ELISA. less of the number of foci on susceptible control cells. Negative and
For the assay for ALV gs antigen by ELISA, precoated ELISA positive controls should be included in each test.
plates or kits for detection of ALV gs antigen are available from Non-producer Cell Activation Test. The NP test (41)
various commercial sources (see appendix). ELISA plates for requires NP cells that are transformed by a defective strain of RSV
detection of ALV gs antigen can also be prepared in the laboratory (Bryan high-titer strain) and that do not produce virus detectable in
using commercially available rabbit anti-p27 immunoglobulin G the usual tests on C/E cells. A Japanese quail cell line transformed
(IgG). Add 100 μΐ of a 1:1000 dilution (or as recommended by the by the envelope-defective Bryan high-titer strain of RSV [R(-)Q]
manufacturer) of rabbit anti-p27 in carbonate buffer (pH 9.4) to has been used as a source of RSV genome. Test samples are
each well of the ELISA plates. Incubate the plates at room inoculated onto genetically susceptible cells, virus-free CEF
temperature (25 C) overnight and then store them at 4 C. Plates cultures that are then co-cultivated with NP cells. Cultures infected
coated in the laboratory should not be stored for more than 10 days. with ALV can be identified by testing the supernatant in cultures or
If samples are to be tested by commercial ELISA kits, follow the in chicken embryos for presence of RSV. For exogenous ALV, C/E
instructions provided by die manufacturer. Otherwise, the cells are co-cultivated with R(-)Q cells, and supernatant is assayed
following general procedure is recommended for ELISA for ALV: on C/E cells. The R(-)Q cells can also be used in assays for
10) Decant coating solution and drain plates; then wash them three endogenous ALV, as well as in assays for chick helper factor (19).
times with 0.1% Tween-80 in PBS. Transformation (Focus) Assay for Sarcoma Viruses.
11) To minimize nonspecific binding and consequently high Avian sarcoma viruses transform spindle-shaped flat CEFs into
background, 100 μΐ of 5% instant nonfat dry milk in PBS (31) or spherical and refractile foci that can be enumerated by microscopic
1% bovine serum albumin in PBS can be added to each well to examination of cultures (19). Focus formation by RSV was best
block reacting surface before adding test samples to plates; allow seen in cultures grown in base medium 199 and medium F-10,
these blocking agents to incubate for 30 min at 25 C. tryptose phosphate broth, calf serum, and sodium bicarbonate.
12) Add 100 μΐ of test material to each well in the ELISA plate. Genetically susceptible monolayer CEF cultures are inoculated with
Include in each plate wells for known positive and negative test material. The next day, medium is decanted and is replaced
samples, as well as for a reading blank. with an agar overlay. Cultures should be examined daily for RSV-
13) Incubate for 1 h at 38-40 C. induced foci, which usually develop within 4-7 days PI.
14) Carefully decant material from plates and drain; wash three Chicken Embryos. Genetically susceptible virus-free chicken
times with 0.1% Tween-80 in PBS. embryos can be used to assay for ALVs, as well as for avian
15) Add 100μ1 of conjugate (peroxidase-conjugated rabbit anti-p27 sarcoma viruses. Embryos infected with ALV by intravenous or
IgG) to each well. The dilution of conjugate should be determined yolk-sac inoculation hatch as normal chicks and later develop
by titration or as recommended by the manufacturer. neoplasms, such as erythroblastosis and LL (19).
16) Incubate for 1 h at 38-40 C. Avian sarcoma viruses induce characteristic pocks on the
17) Decant conjugate and drain plates; wash three times with 0.1% chorioallantoic membrane 8 days after inoculation of 9 to 11-day-
Tween-80 in PBS. old embryos. This procedure of inoculating the chorioallantoic
18) Add 100μ1 of a freshly prepared enzyme substrate, such as 5 membrane with sarcoma virus is commonly used in determining the
amino salicylic acid in 0.2 M phosphate buffer (pH 6.0). The color genetic susceptibility of chickens to the various groups of the L/S
reaction should develop within 30 min. virus. Embryos inoculated intravenously with sarcoma viruses
19) Read absorbance at 490 nm in an ELISA reader or other usually die from sarcoma and hemorrhage within 4 to 5 days (19).
suitable spectrophotometer. Chickens. Chickens used for in vivo assays of ALVs must be
20) Samples with absorbance values of 0.2 units above values of free from virus infection and genetically susceptible to the tumor
known negative samples are considered positive. and virus concerned. Intra-abdominal or intravenous inoculation of
Phenotypic Mixing (PM) Test. The PM test requires CEFs susceptible 1-day-old chicks with ALV usually leads to
susceptible to infection with viruses of subgroup A, B, C, D, J, E development of LL as well as other tumors (19). The time required
(C/O cells), cells resistant to subgroup E ALV (C/E) and stocks of for the assay depends on the response to be measured and on the
RSV (Rous-associated virus-0) (RSV[RAV-0]). All cells must be method used to measure such a response. For example, a LL
from embryos free from exogenous and endogenous ALV. response can be obtained within 36 wk based on gross lesions and
For virus propagation and phenotypic mixing, the ALV is grown mortality, whereas such a response can be measured at 16-18 wk on
together with RSV under conditions that optimize phenotypic the basis of gross and microscopic examinations of the bursa of
mixing. In assaying for the presence of phenotypically mixed RSV, Fabricius. Generally, the time required for assay of ALV-induced
the RSV with the envelope of the ALV must be detected in a system
167
Aly M. Fadly, Richard L. Witter and Henry D. Hunt

tumors such as erythroblastosis can be shortened if chickens are 7) The cells are then incubated at 4 C for 20 min, washed twice by 1
infected during embryonic life (19). min centrifugations using 200 μΐ FCM, and incubated with an
Unlike ALVs, sarcoma viruses do not exist in chicken populations appropriate dilution of either FITC labeled anti chicken
under natural conditions. The most common route used for in vivo (convalescent chicken serum) or mouse (monoclonal) antibody for
assay of these viruses is subcutaneous in the wing web. The test an additional 15 min at 4 C.
sample (100-200μ1) is inoculated between the two layers of the skin 8) After the final incubation, the cells are washed twice in FCM and
of tiie wing web of 4 to 6-wk-old chickens. Palpable tumors are analyzed by flow cytometry.
usually formed within 7 to 14 days at the site of inoculation. These 9) The MCF of the control and virus specific antisera signal is then
tumors either grow progressively and metastasize, eventually killing computed and the results are analyzed using a modified sample to
the host, or regress completely, leaving a normal appearing wing positive (S/P) ratio.
web. Regression is known to be influenced by host age, genotype, 10) The S/P ratio is calculated by subtracting the MCF obtained
and dose of virus. Chickens that survive RSV-induced tumors may using the negative control serum from the MCF obtained using the
develop LL because ALVs coexist in most sarcoma virus stocks. virus specific reference sera for all cells analyzed (positive control
Sensitivity is maximum after 14 days by most procedures, reference virus and field virus infected cultures). The value obtained
although high concentrations of virus may be detected after shorter from the field virus is then divided by the value obtained from the
periods and endpoint assays may be conducted by enzyme positive control culture infected with the reference virus. An S/P
immunoassay within 8-9 days. ratio of 0.1 or greater indicates a positive field sample.
Flow Cytometry. Microscopic immunofluorescence is a common Caution must be used in the interpretation of negative or slightly
technique used to analyze infected cells for expression of virus positive S/P values (0.1 to 0.12). A negative result does not
proteins and to screen live animals for serum antibodies induced by necessarily confirm the absence of virus in the field sample, it could
retrovirus infection. Flow cytometry is an extension of the simply means the reference antisera used does not recognize the
immunofluorescence technique and has several advantages over infecting virus. A classic example is the situation observed for
microscopic immunofluorescence evaluation. The main advantage different strains of ALV-J. Chicken antibody induced by the HPRS-
of flow cytometry is its ability to rapidly quantitate the intensity of 103 strain of ALV-J recognizes cells infected with HPRS-103 but
the immunofluorescent staining procedure (28, 33). The flow not cells infected with the ADOL-Hcl strain. In contrast, chicken
cytometer can accurately evaluate 2000 cells per second and thus, in antibody induced by strain ADOL-Hcl of ALV-J recognize cells
2 to 5 sec, 5 to 10 thousand cells (events) can be evaluated. The infected with both the HPRS-103 and Hcl strains of ALV-J (29).
fluorescent intensity of each event is accurately measured by a Another potential pitfall is the possibility of a very low titer of virus
photomultiplier which then assigns each event a fluorescent channel in the field sample. The re-cultured cells from all low and negative
of intensity based on a log scale ranging from 0 to 10,000 channels. samples should be re-analyzed to confirm the result.
The fluorescent intensity of 5 to 10,000 events is then presented in Flow cytometry can also be used to screen flocks for serum
graphic form as a normal distribution. The median channel antibody to retroviruses. The techniques are essentially the same.
fluorescent (MCF) intensity of the normal distribution is then Sera from the flock in question are applied to tissue culture cells
calculated. infected with the reference strains of retroviruses. Uninfected cells
Evaluation of CEFs (Line 0 or DFls) infected with field samples are used as a negative control. As indicated above, the MCF of the
of ALV by flow cytometry requires essentially the same reagents field sera is compared to reference sera to compute a S/P ratio. The
used in the virus-neutralization test or the microscopic same precautions and pitfalls apply to the interpretation of negative
immunofluorescence technique. Monoclonal antibodies (45) or serum samples as mentioned above. Also, it has been shown that
reference convalescent chicken serum containing virus specific some serum samples may test positive by flow cytometry, but
antibodies are used to classify and type ALVs expressed in the negative by virus neutralization (29).
infected tissue culture cells. The subgroup of ALV (A, B, C, D or J) Reticuloendotheliosis. A variety of methods exist for the
can be determined using antiserum specific for each subgroup. demonstration of REV infection in cell cultures. However, the
Controls used in this process consist of uninfected cells and cells demonstration of antigens by immunofluorescence or enzyme
infected with purified and well characterized laboratory reference immunoassay are probably the methods of choice. Only methods for
strains of ALVs. Recently, this technique has been shown to be very detection of nondefective strains of REV will be given in detail.
valuable in determining the subgroup of an extraneous ALV Immunofluorescence, Anti-REV positive test serum is easily
isolated from commercial Marek’s disease vaccines (20) obtained from convalescent chickens (30), and satisfactory
1) Samples are inoculated on 35-mm plates containing susceptible fluorescein-conjugated anti-chicken globulin serum is available
CEFs as described below for ALV. commercially. Alternatively, mouse monoclonal antibodies (11)
2) Control cultures consist of uninfected cells and cells infected and the corresponding anti-mouse globulin serum can be used.
with laboratory reference strains. The control and field sample Sample-inoculated monolayer CEFs grown on 96-well, 35 mm
cultures are harvested by trypsinization, pelleted by centrifugation plates, or on glass-coverslips are drained, rinsed with PBS, and
(RCF -300), and resuspended in media. then fixed for 5 min in a mixture of six parts acetone and four parts
3) One fourth of the cells are then re-cultured in 35 cm tissue 95% alcohol and air dried. The fixed monolayers can be stained
culture plates for an additional 7 to 9 days to amplify weak samples immediately or can be stored at -20 C until tested. For staining, a
(see below). standard immunofluorescence technique is used (40).
4) The remainder of the cells are pelleted and resuspended in 1ml REV-infected cells have finely granular or diffuse fluorescence
flow cytometry media (FCM), a phosphate buffered saline with 1% throughout their cytoplasm, but not in the nucleus. It is helpful to
bovine serum albumin and 0.1% sodium azide. critically compare cultures inoculated with test samples with
5) 100μ1 of each sample (control and field samples) are then added positive and negative controls. The use of monoclonal antibodies
to three wells of a 96 well U bottom micro titer plate. facilitates interpretation of the test, because of intense specific
6) The first well of each sample is a media control, the second and staining and the virtual absence of background.
third wells receive control chicken serum or monoclonal antibody Flow cytometry. Flow cytometry can also be used to detect
and the virus specific chicken serum or monoclonal antibody, and identify REV isolates. Using REV convalescent and
respectively (final concentration of antibody is 1:50 for chicken monoclonal antibodies, the test is conducted in a way similar to that
serum and 1:1000 for monoclonal). described above for flow cytometry for ALV.

168
Chapter 35 Oncornaviruses Leukosis/Sarcomas and Reticuloendoihebosxs

Enzyme Immunoassay. An immunoassay based on Proviral DNA from infected cell cultures or tumors will hybridize
monoclonal antibodies has been described (12). ELISA plates are with specific probes, such as pSNV, and can be detected by dot blot
coated with a mixture of monoclonal antibodies 11A25 and 11C237 or Southern blot assays (62). Such tests have powerful specificity
(11), each at a dilution of 1:1000 in coating buffer; plates are then and should be helpful in confirming the identity of putative isolates.
incubated overnight at room temperature (see ELISA-ALV test).
For the test, wells are treated with 0.1 ml of test samples following SEROLOGIC DETECTION IN THE HOST
two freeze-thaw cycles to release antigen. The test is conducted in a
way similar to that described above for ELISA-ALV using an Avian Leukosis
appropriate dilution of a rabbit anti-REV serum and peroxidase- Virus-Neutralization Test. The virus-neutralization test is the
labeled anti-rabbit IgG conjugate as the primary and secondary most sensitive test used to determine subgroup-specific antibody to
antibodies, respectively. L/S viruses in chickens. The test requires reference ALV or RSV
At present, reagents for this test are not commercially available. stocks, reference antibody to the subgroup of L/S virus concerned,
The rabbit serum may be produced by inoculation of a rabbit and genetically susceptible CEF cultures. Virus and antibody stocks
subcutaneously at multiple sites with 2 mg of protein from sucrose- can be prepared in the laboratory or obtained from commercial
gradient-purified strain T virus in Freund's complete adjuvant, sources (see appendix). Stocks of ALV or RSV can be prepared by
followed at 21, 35, and 49 days by booster immunizations, each inoculating susceptible CEF cultures. Subculturing of infected
containing 2 mg of protein in Freund's incomplete adjuvant. Serum cultures or transferring supernatant fluids onto new cultures is used
obtained at 63 days is adsorbed against normal CEFs and acetone- to enhance the level of infection. Supernatant fluid harvested from
dried chicken liver powder until it fails to react with normal chicken CEF cultures prepared from embryos congenitally infected with
cells. Availability of the monoclonal antibodies may be arranged ALV can be used as a source of ALV. Tumors induced by RSV in
with the laboratory of origin (11, 12). wing web or breast muscle can be used as a source of RSV (19).
Complement Fixation. Cells scraped from cultures 14 Reference subgroup-specific antiserum to RSV can be collected
days after inoculation can be tested for REV-specific antigens by from chickens that have regressed sarcomas induced by respective
the complement-fixation (COFAR) test (52). A specific anti-REV virus. Antibody to ALV can be prepared by inoculating susceptible
antiserum obtained from rabbits immunized with purified viral gs 4 to 6 wk old chickens with respective ALV. Sera can be harvested
antigens is used, and the complement-fixation test is conducted in at 6 to 16 wk PI, depending on chicken genotype and strain of virus
the normal manner. used.
Reverse-Transcriptase Assay. Cells obtained from cultures 14 Antibody to ALV is measured by its reaction with RSV, and the
days after inoculation can also be tested for the enzyme reverse neutralization of virus is determined by any of the RSV assay
transcriptase (43). Preceding this test, however, appropriate tests procedures; e.g., in chickens, chicken embryos, or cell culture (19).
must be used to rule out other viruses containing reverse A microneutralization test that uses ALV as indicator virus can also
transcriptase, such as ALV in chickens, C-type oncornavirus in be used (17). The test can be conducted in 96-well microtiter plates,
pheasants, or lymphoproliferative disease (LPD) virus in turkeys and the neutralization of the virus is determined by an ELISA on
(4)· culture fluids.
Cytopathic Effect. Within 5-7 days on initial passage of REV, Only the cell-culture (RSV-focus-reduction and ALV
a low-grade cytopathic effect may be often seen (though not microneutralization-ELISA) will be given in detail.
always). This effect only rarely culminates in total destruction of Rous Sarcoma Virus-Focus Reduction Assay. The focus
the culture (58). Plaque assays have been described but are not reduction assay for L/S virus antibody is conducted as follows:
generally applicable to primary isolation. 1) Dilute serum 1:5 and inactivate it for 30 min at 56 C in a water
Recently, an immunoperoxidase plaque assay has been shown to bath.
be a sensitive and reliable method for detection of REV and 2) Determine the dilution of RSV that will give about 100 or 1500
antibody (5). focus-forming units of virus, respectively, when foci are counted
Although defective REVs have not yet been recognized in field grossly
strains, their presence has not been excluded. The isolation of new or microscopically. The RSV working preparation should be twice
defective strains or the propagation of the defective strain T may the concentration determined in step 2. For example, if a 1:200
require special procedures, such as the in vitro transformation of dilution will give 100 foci per plate, the working preparation should
bone-marrow cells or inoculation of birds or embryos. The defective be a 1:100 dilution.
strain T can also induce foci of transformed cells in quail embryo 3) Mix equal volumes of RSV working preparation and the
fibroblast cultures (27), which may constitute a method of assay. inactivated diluted serum, and incubate the mixture at 37 C for 40
min.
Molecular Identification 4) Assay the residual virus in susceptible CEF cultures.
Avian Leukosis. A polymerase chain reaction (PCR) specific for 5) Include known positive and negative sera in the test as controls.
ALV subgroup A can be used to detect proviral DNA and viral 6) Count RSV foci at 7 days PI. A serum sample that reduces the
RNA in various tissues from ALV infected chickens (61). Recently, number of foci by 90% or more, compared with that of control
several PCR primer sets were developed and have been shown to be negative serum, is considered positive for antibody. Negative serum
useful in typing of various field strains of ALV (25, 32, 33, 48, 49, should fail to inhibit the virus, and the number of foci should
53). compare with the number of controls that have virus only.
Proviral DNA from infected CEFs or tumors will hybridize with Microneutralization-ELISA. The microneutralization-ELISA for
specific probes, such as pRAV-2 and can be detected by dot blot or ALV antibody is performed in two phases: virus neutralization and
Southern blot assays (19). assay for residual ALV in culture fluids by ELISA.
Reticuloendotheliosis. A PCR that amplifies the 291 base pairs The virus-neutralization portion is conducted as follows:
product of REV LTR has recently been shown to be a sensitive and 1) Inactivate sera for 30 min at 56 C; dilute sera 1:5 in TC medium
specific method for detection of various strains REV in infected without serum.
CEF as well as in blood and tumors of infected birds (1, 13). PCR 2) Determine the dilution of ALV that will give 500-1000
and reverse transcriptase PCR can also be used in detection of REV infectious units of virus/ml. Use RAV-1, RAV-2, RAV-49, RAV-
in contaminated vaccines (22, 23, 39, 57). 50, HPRS-103 or RAV-0 to assay for antibody to ALV of
subgroups A, B, C, D, J or E, respectively.
169
Aly M. Fadly, Richard L. Witter and Henry D. Hunt

3) Add 50 μΐ of virus preparation to each well in a microtiter plate, precipitin tests must be confirmed. A convenient method employing
except for three to five wells to be used as cell controls. 96-well plates can be used (6). Twofold dilutions of each test and
4) Add 50 μΐ of inactivated diluted test sera to wells; mix serum control serum in culture medium without calf serum are placed in
with virus by drawing the content of the well into the tip of a pipette wells of 96-well plates at 0.05 ml per well. To each well is added
two to three times. an equal volume of prototype REV (strain T or CS) containing
5) Include known positive and negative sera. about 16 infectious units, as determined by simultaneous titration.
6) Incubate microtiter plates at 37 C for 40 min. After incubation for 30 min at 37 C, CEFs are added to each well,
7) Add 5 X 10^ CEFs; cells must be free from endogenous virus and the plate is incubated for 3 days at 38 C in a humidified cell­
and antigen. culture incubator with 5% carbon dioxide. The monolayer cultures
8) Incubate plates for 7 days without changing medium. in each well are then fixed and stained by indirect
9) Add 20 μΐ of 5% Tween 80 to each well; freeze and thaw at least immunofluorescence as described earlier. The wells are scored as
twice before testing supernatant fluid by ELISA. positive or negative for virus following microscopic examination.
For assay of residual ALV, see section on assay for ALV gs The neutralization titer is the highest dilution of serum (after
antigen by ELISA. A negative ELISA reading indicates that serum mixture with the virus) that completely neutralizes the test virus.
is positive for ALV antibody, and a positive ELISA reading Alternatively, ELISA can be used to assay for residual REV (see
indicates that serum is negative for antibody. enzyme immuno assay for detection of REV).
A direct ELISA for ALV antibody has been developed (19, 50),
and kits for detecting antibodies to subgroups A, B and J ALV are DIFFERENTIATION FROM CLOSELY RELATED AGENTS
commercially available (see appendix). False-positive results due to
cross reactions may occur if antibody to the endogenous virus is Avian Leukosis
present, particularly when using kits for detection of antibody to Viruses of the avian L/S group share gs antigens and some unique
subgroups A and B ALV. characteristics, such as the ability to interact with other members of
the group in RIF, PM, and complementation tests (19). This group
Reticuloendotheliosis of viruses can be differentiated from other avian oncornaviruses
Confirmation of REV infection by serologic procedures involves with C-type particle morphology, such as REV on the basis of a
evaluation of sera from naturally exposed birds of the suspect flock. unique gs antigen, unique reverse transcriptase, and a buoyant
Antibodies are induced at various frequencies and persist for varied density of 1.16. The L/S viruses are classed according to: 1) the
periods. Birds infected congenitally or inoculated with high doses as spectrum of neoplasms they induce (pathotype), and 2) the viral
embryos may be permanently viremic and fail to develop envelope subgroup.
antibodies. At least 10-15 sera per group or flock should be tested. The neoplastic condition of turkeys known as LPD is induced by
Pooling of sera is not recommended. yet another oncornavirus that is distinct from both L/S and RE
Indirect Immunofluorescence. The indirect fluorescent antibody viruses (4). However, the incidence of LPD of turkeys has always
test is useful for detecting REV antibodies in sera and egg-yolk been sporadic and the disease has not been reported during at least
extracts (38). Cultures of CEF in 96-well plates are infected with the last decade (16).
about 50-100 fluorescent focus-forming units of REV, grown for 3- Pathotypes. Viruses of avian L/S group that induce neoplasms
5 days under liquid medium and fixed. In a satisfactory antigen, over a long period of time (slowly transforming viruses) are known
many but not all cells are infected. The unknown sera are used at a as ALVs, whereas viruses that cause acute neoplasms (apparent
1:20 dilution in the staining test as described earlier for virus within 2 to 6 wk) are known as avian sarcoma and defective
isolation. Controls with normal serum and heterologous antiserum leukemia viruses.
on both infected and uninfected cultures are necessary. Tests on Subgroup and Strain Variability. Viruses of the avian L/S group
sera from other avian species require the appropriate conjugated that occur in chickens are divided into six subgroups (A, B, C, D, E
anti-species globulin serum. and J) on the basis of their host range in genetically different
Flow cytometry. Flow cytometry can also be used to serologically chicken cells, their interference with transformation of CEF cultures
identify REV isolates. The test is conducted in a way similar to that by RSV members of the same subgroup, and the viral envelope
described above for flow cytometry for ALV. antigens detected by virus- neutralization tests (19). Subgroup E
Enzyme Immunoassay. An indirect ELISA in 96-well plates can ALV are endogenous ALVs of chickens known to be of low
also be used for detection of antibodies to REV in chicken sera (54). pathogenicity. Other subgroups of avian L/S viruses are endogenous
This test appears to be more sensitive than immunofluorescence or viruses rescued from pheasants (F, G), Hungarian partridge (H) and
serum neutralization. However, sera should be diluted 1:200 to quail (I) (19).
1:400 to avoid false-positive reactions. A commercial kit for Laboratory as well as field isolates that belong to one subgroup of
detection of REV antibodies is available (see appendix). L/S virus are usually termed strains of that subgroup. Tolerance
Agar-Gel Precipitin Test. Antibody-containing sera from REV- induction and tumor incidence and type are influenced by the strain
infected chickens form specific bands within 24-72 h in the standard of virus (19).
agar-gel precipitin test when reacted against suitable antigens, e.g., Characteristics of Vaccine Strains. No commercial vaccine is
extracts of infected cell cultures, lOOx concentrated supernatant available for protection of chickens from infection with ALV.
fluids from infected cell cultures, viremic chicken plasma, or virus Results of experimental vaccination with live ALV on shedding and
particles concentrated by ultracentrifugation and sucrose density­ congenital transmission of the virus were equivocal (19). The use of
gradient banding (30). experimental recombinant L/S viruses as vaccines (19, 44, 66) may
Either antigen or antibody can be detected in test samples by a test prove to be a valuable adjunct to current programs for reduction or
system in which positive antigen is placed in the center well, and eradication of ALV infection.
test sera and positive control sera are placed adjacent to each other
in peripheral wells (30). Specificity of precipitin bands must be Reticuloendotheliosis
confirmed by reactions of identity with positive controls in adjacent Strain Variability. All REV regardless of species of origin, are
wells. antigenically related to each other and serologically distinct from all
Virus Neutralization. The virus-neutralization test is used for other oncornaviruses. However, antigenic characterization of
antibody detection when sera cannot be tested by indirect various REV isolates using a panel of 11 monoclonal antibodies
immunofluorescence or when results of immunofluorescent or
170
Chapter 35 Oncornaviruses Leukosis/Sarcomas and Reticuloendothehoes

(11) revealed the presence of three different subtypes, A, B and C 3. Bacon, L. D., R. L. Witter, and A. M Fadly. Augmentation of
(6). oncornavirus-induced lymphoid leukosis by Marek’s disease herpesviruses
REV-induced immunosuppressive syndromes can usually be in white leghorn chickens. J. Virol 63:504-512. 1989.
4. Biggs, P.M Lymphoproliferative disease of turkeys, in: B. W. Calnek, H_
distinguished from lesions induced by infectious bursal disease or
J. Bames, C. W. Beard, L. R. McDougald, and Y. M Saif (Eds) Diseases of
chicken anemia viruses by histopathology. Atrophy of lymphoid Poultry, 10th Ed, pp. 485-489. Iowa State University Press, Ames, Iowa.
organs with immunodepression, nerve lesions and abnormal feather 1997.
development are characteristics of REV-induced immunodepressive 5. Calvert, J. G., and K. Nazerian. An immunoperoxidase plaque assay for
disease. reticuloendotheliosis virus and its application to a sensitive serum
Characteristics of Vaccine Strains. No vaccines are available, neutralization assay. Avian Dis. 38:165-171. 1994.
and control, when warranted, involves removal of virus-shedding 6. Chen, P. Y., Z. Cui, L. F. Lee, and R. L. Witter. Serologic differences
hens from breeding flocks and protection of progeny against among nondefective reticuloendotheliosis viruses. Arch. Virology 93, 233-
245. 1987.
horizontal infection through good management and strict isolation
7. Cho, B. R. Cytopathic effects and focus formation by
(62, 64). reticuloendotheliosis viruses in a quail fibroblast cell line. Avian Dis
27:261-270, 1983.
Differential Diagnosis of Avian Oncornavirus-Induced Tumors 8. Crespo, R, P. R. Woolcock, , A. M Fadly, C. Hall, and H L.
The pathology of tumor conditions induced by viruses of the L/S Shivaprasad. Characterization of T-cell lymphomas associated with an
group, particularly those of lymphoproliferative nature, can be outbreak of reticuloendotheliosis in turkeys. Avian Pathology.31:355-361.
confused with that of tumors seen in MD and RE. Because ALV, 2002.
REV and MDV are all ubiquitous in the chicken population and 9. Crittenden, L. B., S. McMahon, M S. Halpern, and A. M Fadly.
Embryonic infection with the endogenous avian leukosis virus Rous-
infection in the absence of tumor formation is common, virologic
associated virus-o alters response to exogenous avian leukosis virus
and serologic criteria rarely provide a definitive diagnosis. infection. J. Virol. 61:722-725. 1987.
Techniques based on molecular hybridization, or 10. Crittenden, L. B., and E. J. Smith. A comparison of test materials for
immunocytochemistry with monoclonal antibodies to cellular differentiating avian leukosis virus group-specific antigens of endogenous
antigens can be used in the differential diagnosis of avian viral and exogenous origin. Avian Dis. 28:1057-1070. 1984.
lymphomas (8,24, 63). 11. Cui, Z., L. F. Lee, R. F. Silva, and R L. Witter. Monoclonal antibodies
Avian oncomaviral lymphomas originate from either B-cells against avian reticuloendotheliosis virus: identification of strain specific and
(ALV, REV) or T-cells (REV), whereas MD lymphomas are of T- strain common epitopes. J Immunol. 136:4237-4242. 1986.
12. Cui, Z., L. F. Lee, E. J. Smith, R L. Witter, and T. S. Chang
cell origin (19, 34, 47, 62, 65). These characteristics provide the
Monoclonal-antibody-mediated enzyme-linked immunosorbent assay for
basis for tests that distinguish among B- and T-cell lymphomas detection of reticuloendotheliosis viruses. Avian Dis. 32:32-40. 1988.
using monoclonal antibodies specific for cell surface antigens of B- 13. Davidson, I., A. Borovskay, S. Perl and M Malkinson. Use of the
and T-lymphocytes. polymerase chain reaction for the diagnosis of natural infection of chickens
Because nondefective ALV and REV do not posses viral (v)-onc and turkeys with Marek’s disease virus and reticuloendotheliosis virus.
gene, they transform target cells by integration within a cellular Avian Pathology 24:69-94. 1995.
(c)onc gene such as c-myc resulting in the enhanced expression of 14. DeBoer, G. F., and A D. Μ E. Osterhaus. Application of monoclonal
such gene. The enhanced expression of c-myc is believed to initiate antibodies in the avian leukosis virus gs-antigen ELISA. Avian Pathology.
14:39-55. 1985.
the lymphomagenic process. These molecular changes are the bases
15. Ewert, D. L., and G. F. DeBoer . Avian lymphoid leukosis: Mechanism
for testing tumor DNA for the definitive diagnosis of ALV and of lymphomagenesis. In: K. Perk (ed), Immunodeficiency Disorders, pp. 37-
REV lymphomas. Using appropriate probes such as pRAV-2, 53, Academic Press, Inc., Boston. 1988.
pSNV, or c-myc, Southern blots and hybridization analysis of tumor 16. Fadly, A. M Neoplastic Diseases of Poultry, in: Y.M Saif, H. J.
DNA is used to detect clonal insertion of ALV or REV provirus and Bames, A.M Fadly, J.R Glisson, L.R. McDougald, and D.E. Swayne (Eds)
alteration in c-myc (19, 62). Diseases of Poultry, 11th Ed. pp. 405-407. Iowa State University Press,
PCR has been used for the detection of two copies of the 132-base- Ames, Iowa. 2003.
pair repeat in the DNA extracted from MD lymphomas, but not 17. Fadly, A Μ, T. F. Davison, L. N. Payne, and K. Howe. Avian leukosis
virus infection and shedding in brown leghorn chickens treated with
from DNA extracted from lymphomas induced by ALV or REV
corticosterone or exposed to various stressors. Avian Pathology. 18:283-298.
(65). MD lymphomas probably contain a higher frequency of 1989.
infected cells and more copies of viral DNA per infected cell than 18. Fadly, A. M, W. Okazaki, E. J. Smith, and L. B. Crittenden. Relative
are typically found in tissues of nontumor-bearing chickens (65). efficiency of test procedures to detect lymphoid leukosis virus infection.
Recently, it has been shown that MD-induced tumors have many Poultry .Sci. 60:2037-2044. 1981.
more copies of MDV genome than MDV latently infected tissues. A 19. Fadly, A. M and L. N. Payne. Leukosis/sarcoma group, in: Y.M Sai£
real time PCR technique was developed to differentiate MD- H. J. Bames, A. M Fadly, J. R. Glisson, L. R McDougald, and D. E.
induced tumors and tumors induced by retroviruses in chickens Swayne (Eds) Diseases of Poultry, 11th Ed. pp. 465-516. Iowa State
University Press, Ames, Iowa. 2003.
coinfected with MDV (24, 63). Also, a PCR has been shown to
20. Fadly, A.M, Silva, RF., Hunt, H.D. Detection of exogenous and
detect REV-LTR sequences from lymphomas and brains of REV- endogenous avian leukosis viruses in commercial Marek's disease vaccines.
infected chickens, but not from DNA from LL or MD lymphomas In: Proceedings of the 170th Annual Meeting of the United States Animal
(1 13,22). Health Association, pp. 524-525. 2003.
PCR assays demonstrate the presence or absence of the respective 21. Fadly, A. M, and R L. Witter. Effects of age at infection with serotype
virus and thus the same limitations for tumor diagnosis as virus 2 Marek’s disease virus on enhancement of avian leukosis virus-induced
isolation. lymphomas. Avian Pathology 22:565-576. 1995.
22. Fadly, A. M, R. L. Witter, E. J. Smith, R F. Silva , W. M Reed, F. J.
REFERENCES Hoerr, and M R. Putnam. An outbreak of lymphomas in commercial broiler
1. Aly, Μ Μ, E. J. Smith, and A. M Fadly. Detection of breeder chickens vaccinated with a fowl pox vaccine contaminated with
reticuloendotheliosis virus infection using the polymerase chain reaction. reticuloendotheliosis virus. Avian Pathology, 25:35-47. 1996.
Avian Pathol. 22:543-554. 1993. 23. Garcia, Μ, N. Narrang, W. M Reed, and A. M. Fadly. Molecular
2. Aly, Μ. M, R. L. Witter, and A. M Fadly. Enhancement of Charcterization of reticuloendotheliosis virus (REV) insertion in the genome
reticuloendotheliosis virus-induced bursal lymphomas by serotype 2 of field and vaccine strains of fowlpox virus(FPV). Avian Dis. 47:343-354.
Marek’s disease virus. Avian Pathology, 25:81-94. 1996. 2003.

171
Aly M. Fadly, Richard L. Witter and Henry D. Hunt

24. Gimeno, I. M., R. L. Witter, A. M. Fadly and R. F. Silva. Novel Criteria 48. Silva, R.F., A. M Fadly, and H. D. Hunt. Hypervariability in the
for the diagnosis of Marek’s disease virus-induced lymphomas. Avian envelope genes of subgroup J avian leukosis viruses obtained from different
Pathology. 34:332-340- 2005. farms in the United States. Virology 272:106-111. 2000.
25. Gingerich, E., R. E. Porter, B. Lupiani, and A. M Fadly Diagnosis of 49. Smith, L.M, S.R. Brown, K. Howes, S. McLeod , S.S. Arshad, G.S.
myeloid leukosis induced by a recombinant avian leukosis virus in Barron, K. Venugopal, J.C. McKay and L.N. Payne. Development and
commercial white leghorn egg laying flocks. Avian Dis. 46:745-748. 2002. application of polymerase chain reaction (PCR) tests for the detection of
26. Glika, F., and J. L. Spencer. Immunohistochemical identification of subgroup J avian leukosis virus. Virus Research. 54:87-98. 1998.
group specific antigen in avian leukosis virus infected chickens. Can. J. 50. Smith, E. J., A. M Fadly, and L. B. Crittenden. Observations on an
Comp. Med. 48:322-326. 1984. enzyme-linked immunosorbent assay for the detection of antibodies against
27. Hoelzer, J. D., R. B. Franklin, and H. R. Bose. Transformation by avian leukosis-sarcoma viruses. Avian Dis. 30:488-493. 1986.
reticuloendotheliosis virus: development of a focus assay and isolation of a 51. Smith, E. J., A. Fadly, and W. Okazaki. An enzyme-linked
non-transforming virus. Virology 93:20-30. 1979. immunosorbent assay for detecting avian leukosis-sarcoma viruses. Avian
28. Hunt, H. D., L. F. Lee, D. Foster, R. F. Silva, and A. M Fadly. A Dis 23:698-707. 1979.
genetically engineered cell line resistant to .subgroup J avian leukosis virus 52. Smith, E. J., J. J. Solomon, and R. L. Witter. Complement-fixation test
infection (C/J). Virology. 264:205-210. 1999. for reticuloendotheliosis viruses: limits of sensitivity in infected avian cells.
29. Hunt, H. D., V. Leathers, C. Lamichhane, G. Rosales, E. Jensen, J. Avian Dis. 21:612-622. 1977.
McKay, and A. M Fadly. Comparison of the different techniques (ELISA, 53. Smith, E.J., S. M. Williams, and A. M Fadly. Detection of avian
Fluorescent Antibody, and Virus Neutralization) for detecting serum leukosis virus subgroup J using the polymerase chain reaction. Avian
antibody to the subgroup J Avian Leukosis Virus (ALV-J). Presented at the Dis.42:375-380. 1998.
International Symposium on ALV-J and Other Avian Retroviruses, Editors 54. Smith, E. J., and R. L. Witter. Detection of antibodies against
Kaleta, E.F., Payne, L.N. and Heffels-Redmann, U. Rauischholzhausen, reticuloendotheliosis viruses by an enzyme-linked immunosorbent
Germany. 2000. assay.Avian Dis. 27:225-234. 1983.
30. Ianconescu, M Reticuloendotheliosis antigen for the agar gel 55. Spencer, J. L., L. B. Crittenden, B. R. Burmester, C. Romero, and R. L.
precipitation test. Avian Pathology. 6:259-267. 1977. Witter. Lymphoid leukosis viruses and gs antigen in unincubated chicken
31. Johnson, D. A., J. W. Guatsch, J. R. Sportsman, and J. H. Eder. eggs. Avian Pathol. 5:221-226. 1976.
Improved technique utilizing nonfat dry milk for analysis of proteins and 56. Sung, H. W., S. M Reddy, and A. M Fadly. High virus titer in feather
nucleic acids transferred to nitrocellulose. Gene Anal. Technol. 1:3-8. 1984. pulp of chickens infected with subgroup J avian leukosis virus. Avian Dis.
32. Lupiani, B., S. M. Williams, R. F. Silva, H. D. Hunt, and A. M Fadly, 46:281-286. 2002.
Pathogenicity of two recombinant avian leukosis viruses. Avian Dis. 47:425- 57. Takagi, Μ, I. Kiyoyasu, N. Hideki, T. §asaki, G. Kisako , and H.
432. 2003. Koyama. Detection of contamination of vaccines with the
33. Lupiani, B., H. Hunt, R. Silva, and A. Fadly. Identification and reticuloendotheliosis virus by reverse transcriptase polymerase chain
characterization of recombinant subgroup J avian leukosis viruses (ALV) reaction (RT-PCR). Virus Research, 40:113-121. 1996.
expressing subgroup A ALV envelope. Virology 276:37-43. 2000. 58. Temin, Η. M, and V. K. Kassner. Replication of reticuloendotheliosis
34. Neumann, U., and R. L. Witter. Differential diagnosis of lymphoid viruses in cell cultures: acute infection. J. Virol. 13:291-297. 1974.
leukosis and Marek's disease by tumor-associated criteria. I. Studies on 59. Theilen, G. H., R. F. Zeigel, and M J. Twiehaus. Biological studies
experimentally infected chickens. Avian Dis. 23:417-425. 979. with RE virus (strain T) that induces reticuloendotheliosis in turkeys,
35. Okazaki, W., H. G. Purchase, and B. R. Burmester. Phenotypic mixing chickens, and Japanese quail. J. Natl. Cancer Inst. 37:731-743. 1966.
test to detect and assay avian leukosis viruses. Avian Dis. 19:311-317. 1975. 60. U.S. Department of Agriculture, SAM-405. Supplemental assay method
36. Payne, L. N., S. R. Brown, N. Bumstead, K. Howes, J. A. Frazier and for detecting lymphoid leukosis biocontamination by the COFAL test.
Μ E. Thouless. A novel subgroup of exogenous avian leukosis virus in Biologies Laboratories, Veterinary Services, Animal and Plant Health
chickens. J. Gen. Virology 72:801-807. 1991 Inspection Service, Ames, Iowa. 1973.
37. Payne, L. N., A. M. Gillespie, and K. Howes. Unsuitability of chicken 61. Van Woensel, P. A. M, A. van Blaaderen, R. J. M Mooman, and G. F.
sera for detection of exogenous ALV by the group-specific antigen ELISA. de Boer. Detection of proviral DNA and viral RNA in various tissues early
Vet Rec 132:-555-557. 1993. after avian leukosis virus infection. Leukemia 6 (S3): 1355-1375. 1992.
38. Purchase, H. G., C. G. Ludford, K. Nazerian, and H. W. Cox. A new 62. Witter, R.. L. and A. M Fadly. Reticuloendotheliosis, in: Y.M Saif, H.
group of oncogenic viruses: reticuloendotheliosis, chick syncytial, duck J. Bames, A.M Fadly, J. R. Glisson, L. R. McDougald, and D. E. Swayne
infectious anemia, and spleen necrosis viruses. J. Natl. Cancer Inst. 51:489- (Eds) Diseases of Poultry, 11th Ed. pp. 517-536. Iowa State University
499. 1973. Press, Ames, Iowa. 2003.
39. Reimann, I, and O. Werner . Use of polymerase chain reaction for the 63. Witter, R. L., I. M. Gimeno and A. M Fadly. Differential diagnosis of
detection of reticuloendotheliosis virus in Marek’s disease vaccines and lymphoid and myeloid tumors in chickens[CD-ROM] , set 27. Am. Assoc.
chicken tissues. J. Vet. Med, 43:75-84. 1996. Avian Pathologists, Athens, Georgia. 2005
40. Rodriguez, F. R., and F. Deinhardt. Preparation of a semi-permanent 64. Witter, R. L., and D. W. Salter. Vertical transmission of
mounting medium for fluorescent antibody studies. Virology 12:316- reticuloendotheliosis virus in breeder turkeys. Avian Dis. 33:226-235. 1989.
317.1960. 65. Witter, R, L. and K. A. Schat. Marek’s disease. In: Y.M. Saif, H. J.
41. Rispens, B. Η., P. A. Long, W. Okazaki, and B. R. Burmester. The NP Bames, A.M Fadly, J.R. Glisson, L.R. McDougald, and D.E. Swayne (Eds)
activation test for assay of avian leukosis/sarcoma viruses. Avian Dis. Diseases of Poultry, 11th Ed. pp. 407-465. Iowa State University Press,
14:738-751. 1970. Ames, Iowa. 2003.
42. Rubin, H. A virus in chick embryos which induces resistance in vitro to 66. Wright, S. E., and D. D. Bennett. Avian oncornavirus recombinant
infection with Rous sarcoma virus. Proc. Natl. Acad. Sci. U.S.A. 46:1105- expressing foreign envelope delays tumour formation of ASV-A-induced
1119. 1960. sarcoma. Vaccine 10:375-378. 1992.
43. Sarma, P. S., D. K. Jain, Η. K. Mishra, J. L. Vernon, P. S. Paul, and B.
S. Pomeroy. Isolation and characterization of viruses from natural outbreaks
of reticuloendotheliosis in turkeys. J. Natl. Cancer Inst. 54:1355-1359. 1975.
44. Qin, A., Z. Cui, L.F. Lee, and A. M Fadly. Cloning and expression of
envelope gene of subgroup J avian leukosis virus. Chinese Journal of
Virology. 17:54-59. 2001.
45. Qin, A., L.F. Lee, A. M Fadly, H. Hunt, and Z. Cui. Development and
characterization of monoclonal antibodies to subgroup J avian leukosis
virus. Avian Dis. 45:938-945. 2001.
46. Sarma, P. S., H. C. Turner, and R. J. Huebner. An avian leukosis group­
specific complement-fixation reaction. Application for the detection and
assay of non-cytopathogenic leukosis viruses. Virology 23:313-321. 1964.
47. Siccardi, F. J., and B. R. Burmester . The differential diagnosis of
lymphoid leukosis and Marek's disease. USDA Technical Bulletin 1412.
1970.

172
36
AVIAN ENCEPHALOMYELITIS
Louis van der Heide

SUMMARY. Avian encephalomyelitis (AE) or epidemic tremor is an infectious viral disease of poultry caused by a picomavirus. Chickens,
turkeys, pheasants, and quail are naturally susceptible. AE is mostly an egg-transmitted disease. If susceptible breeder chickens become
infected with AE virus during production, the offspring may have clinical AE in the first 4 wk of life. Horizontal transmission may also occur
between infected and susceptible chicks, or between mature birds as well. Chick losses and decreased egg production are the main economic
factors of AE.
Agent Identification. The primary preferred diagnostic tests are isolation of AE virus in 6-day embryonating chicken eggs by yolk sac
inoculation and/or demonstration of AE viral antigen in tissues from suspect poultry.
Serologic Detection in the Host. Secondary diagnostic tests identify anti-AE viral antibodies in suspect poultry sera by either the virus­
neutralization test using the embryo-adapted Van Roekel strain of AE virus, the chicken embryo susceptibility test, or an enzyme-linked
immunosorbent assay.

INTRODUCTION Virus Isolation by Yolk Sac Inoculation of Embryonating


Chicken Eggs
Avian encephalomyelitis (AE) is an infectious viral disease of For the propagation of AE virus, the embryos used must be from
poultry, which in young chickens, turkeys, pheasants, or quail is an AE-susceptible flock. Five-to-6-day-old embryonating eggs are
characterized by clinical signs of a central nervous system disorder, used for AE virus isolation or propagation. The eggs should be
with high morbidity and variable mortality. Older poultry can specific-pathogen-free eggs that are free of chick embiyonal lethal
become infected but rarely develop clinical signs. orphan (CELO; avian adenovirus) virus, to avoid the confusing
The name epidemic tremor was given to the disease by Dr. similarity of lesions between CELO and AE infection in chicken
Elizabeth Jones in 1934 (11). In 1939 the name avian embryos and hatched chicks. CELO virus can be distinguished from
encephalomyelitis was officially adopted. AE is an egg-transmitted AE virus by neutralization tests with specific antisera.
disease; therefore breeder vaccination is an important preventive For virus isolation, 0.2-0.5 ml of brain suspension, prepared as
measure. above, is inoculated into the yolk sac of 24 6-day-old embryonating
chicken eggs (use 1.5-inch, 22-gauge needles). Twelve embryos are
CLINICAL DISEASE examined for lesions at 12 days PI. Gross lesions consist of embryo
immobility (paralysis), leg muscle atrophy, and sometimes death of
In young chicks, 1—4 wk of age, AE is characterized by neurologic the embryo. If no lesions are found, the other 12 inoculated eggs are
signs such as ataxia, incoordination, paralysis, or rapid tremors of allowed to hatch, and the chicks are observed for clinical signs for
the head and neck. Gross lesions are limited to occasional blindness the first 10 days posthatch. In making primary virus isolations from
from cataracts as sequelae of infection. Microscopic lesions include brains of chickens with clinical AE, Butterfield et al. (3) reported
neuron degeneration (“ghost cells”) particularly in the medulla and that an average of 12 passages were necessary to adapt 19 field AE
anterior horns of the spinal cord. Glial cell satellitosis of isolates to produce lesions in chicken embryos.
degenerating neurons is also common. Numerous circumscribed Embryonating eggs are also used for serial passage of established
lymphoid follicles can be found in the pancreas and lymphocytic virus isolates, for virus titrations, and for neutralization tests of field
aggregates in the muscular tissue of the proventriculus (10). sera with the embryo-adapted Van Roekel strain of AE virus (5,18).
In adult laying birds, AE infection causes no neurologic signs, but
it depresses egg production by 10-15% on a flock basis. Egg Cell Culture
production returns to normal levels in about 2 wk. Virus isolation with chicken embryo cell or organ culture is not
practical, because no cytopathic effects can be observed.
SAMPLE COLLECTION
AGENT IDENTIFICATION
Virus isolation is best from brain material of birds that show early
clinical signs of AE and have not been affected for more than 2 or 3 Avian encephalomyelitis virus is an RNA virus, in the family
days. Brains are harvested aseptically from individual birds or are Picomaviridae, genus Enterovirus (3). The titer of AE virus is not
pooled from several birds and stored at -20 C (regular freezer) or affected by treatment with ether or chloroform (3). AE virus
lower until prepared for inoculation. Brain material is ground in a particles are spherical with diameters ranging from 16.5 to 25 nm
tissue grinder with nutrient broth added to make a 10%-25% (3).
suspension. The suspension is centrifuged at 1000 x g for 10 min,
and the supernatant fluid is collected. Penicillin (10,000 IU per ml) Fluorescent Antibody (FA) Test
and streptomycin (100 :g per ml) are added to inhibit growth of any Direct FA testing has been successful in demonstrating AE viral
contaminating bacteria. antigen in tissues of AE-infected chickens (1,14,17). Cryostat
sections, 6-7pm thick, are cut from brain, pancreas, proventriculus,
PREFERRED CULTURE MEDIA AND SUBSTRATES and, if needed, heart, ventriculus, and spinal cord, at a microtome
temperature of -15 to -20 C, using a suitable mounting compound.
Virus Isolation by Intracerebral Inoculation of 1-Day-Old Chicks Sections are mounted on glass slides, air-dried, fixed in cold
Avian encephalomyelitis-susceptible 1-day-old chicks are acetone for 10 min, and air-dried again. The sections are then
inoculated intracerebrally via a 22 or 23-gauge needle with 0.025- reacted with specific anti-AE virus conjugate for 30 min at room
0.03 ml of brain suspension. Brains of chicks that show clinical temperature in a humidified atmosphere, after which the slides are
signs of AE within 1—4 wk postinoculation (PI) can be harvested washed for 20 min in phosphate-buffered saline (PBS) at pH 7.4,
and used for serial passage in susceptible chicks. and then coverslips are mounted with equal parts PBS and buffered
glycerol at pH 7.4. Slides can then be examined under the
fluorescent microscope.

173
Louis van der Heide

Strain Variability Enzyme-Linked Immunosorbent Assay


All reported AE virus isolates and vaccine strains appear to be The enzyme-linked immunosorbent assay (ELISA) is used to
serologically similar to the embryo-adapted Van Roekel strain and determine antibody levels in blood serum. AE antigen microtiter
are considered to be the same serotype. plates for ELISA are commercially available (Synbiotics
Corporation, San Diego, CA and IDEXX, Westbrook, ME).
Characteristics of Vaccine Strains
Live Virus Vaccine. The most widely used AE vaccine is the live- DIFFERENTIATION FROM CLOSELY RELATED AGENTS
virus vaccine developed from strain 1143 by Calnek et al. (4,6,9).
This strain is a mild field strain that should be kept at a low embryo Similar clinical signs, such as paralysis, ataxia, and incoordination
passage for vaccine use (not more than two passages); otherwise, it in chicks, can also be caused by other etiologies. Marek’s disease,
becomes embryo-adapted and loses its efficacy for oral including transient paralysis, can be distinguished by virus isolation
administration. Preferred age for vaccination is between 14 and 18 and histopathology (10). Newcastle disease can be distinguished by
wk, at least 4 wk before the beginning of the laying period. histopathology (12) and virus isolation with confirmation of a
Application is through the drinking water. Minimum virus titer of paramyxovirus type 1 and serologic tests. Encephalomalacia
strain 1143 should be 103 0 EID5o per bird dose. (vitamin E deficiency) can be distinguished by histopathology (10).
Because the vaccine strain is not embryo-adapted, titration is
accomplished by making serial 10-fold dilutions and inoculating 10 REFERENCES
embryos per dilution. The embryos must be from AE-susceptible
stock and are inoculated via the yolk sac with 0.2 ml per embryo. 1. Braune, Μ O., and R. F. Gentry. Avian encephalomyelitis virus. II.
Pathogenesis in chickens. Avian Dis. 15:648-653. 1971.
As controls, 10 embryos are inoculated similarly with the broth
2. Butterfield, W. K., R. E. Luginbuhl, C. F. Helmboldt, and F. W. Sumner.
diluent. The eggs for each inoculation group are placed in pedigree Studies on avian encephalomyelitis. ΠΙ. Immunization with an inactivated
baskets and allowed to hatch. The hatched chicks are identified by virus. Avian Dis. 5:445-450. 1961.
wing bands and placed in a brooder, together with the controls, for 3. Butterfield, W. K., R. E. Luginbuhl, and C. F. Helmboldt.
observation for 3 days. A record is then made of the number Characterization of avian encephalomyelitis virus (an avian enterovirus).
showing clinical signs of AE out of the original number hatched in Avian Dis. 13:363-378. 1969.
each group. The EfD50 is calculated by the method of Reed and 4. Calnek, B. W. Oral vaccine for avian encephalomyelitis. J. Am. Vet.
Muench (13). Med Assoc. 139:1323. 1961.
5. Calnek, B. W., and J. H. Jehnich. Avian encephalomyelitis studies. Annu.
Killed-Virus Vaccine. A beta-propiolactone-killed Van Roekel Rept. Univ. Mass. Bull. No. 509-62. Mass. Agric. Exp. Sta., University of
strain of AE vaccine is available commercially (Lohmann Animal Massachusetts, Amherst, Mass. 1957.
Health, Waterville, ME). Suitable immunity is attained with a 6. Calnek, B. W. , and J. H. Jehnich. Studies on avian encephalomyelitis. Π.
vaccine whose virus titer before inactivation is 106 ELD50 per Immune responses to vaccination procedures. Avian Dis. 3:225-239. 1959.
ml(2,8). 7. Calnek, B. W., R E. Luginbuhl, P. D. McKercher, and H. Van Roekel.
Committee report on a tentative program for the control of avian
SEROLOGIC DETECTION IN THE HOST encephalomyelitis. Avian Dis. 4:456. 1961.
8. Calnek, B. W., and P. J. Taylor. Studies on avian encephalomyelitis. ΠΙ.
Immune response to betapropiolactone inactivated virus. Avian Dis. 4:116—
Various serologic tests are used to confirm AE virus infections 122. 1960.
for the purpose of retrospective diagnosis. 9. Calnek, B. W., P. J. Taylor, and M Sevoian. Studies on avian
Virus-Neutralization Test Serum from birds suspected of having encephalomyelitis. V. Development and application of an oral vaccine.
been infected with AE can be tested for virus-neutralizing Avian Dis. 5:297-312. 1961.
antibodies by using the Van Roekel strain and the procedure 10. Helmboldt, C. F. Histopathologic differentiation of diseases of the
outlined in chapter 46 on serologic procedures. The test uses 6-day- nervous system of the domestic fowl (Gallus gallus). Avian Dis. 16:229-
old AE-susceptible embryonating eggs. Five eggs per dilution are 240. 1972.
inoculated via the yolk sac with a 22-gauge 1.5-inch needle. The 11. Jones, E. E. Epidemic tremor, an encephalomyelitis affecting young
chickens. J. Expt. Med. 59:781-798. 1934.
embryos are examined 12 days PI for typical AE lesions to 12. Jungherr, E. L., and E. L. Minard. The pathology of experimental avian
determine the titration endpoints. Sera from chickens exposed to AE pneumoencephalitis. Am. J. Vet. Res. 5:125-134. 1944.
usually have a logio neutralization index of 1.5-3.0. 13. Reed, L. T., and H. Muench. A simple method for estimating fifty
percent endpoints. Am. J. Hyg. 27:493-497. 1938.
Embryo Susceptibility Test 14. Sato, G., M Kamada, T. Miyamae, and S. Miura. Propagation of non­
The embryo susceptibility test has been developed to determine egg-adapted avian encephalomyelitis virus in chick embryo brain cell
the AE susceptibility or immunity status of a breeder flock (15,16). culture. Avian Dis. 15:326-333. 1971.
To perform the test, 36 hatching eggs are submitted to the 15. Sumner, F. W., R. E. Luginbuhl, and E. L. Jungherr. Studies on avian
encephalomyelitis. II. Flock survey for embryo susceptibility to the virus.
laboratory. After 6 days of incubation, the eggs are candled, and all
Am. J. Vet. Res. 18:720-723. 1957.
fertile eggs are inoculated via the yolk sac with 0.1 ml of a IO'3 16. Taylor, J. R E., and E. P. Schelling. The distribution of avian
dilution of a suspension of the Van Roekel strain of AE virus (about encephalomyelitis in North America as indicated by an immunity test. Avian
1000 mean embryo infective doses [EID50]). Calnek et al. (7) used a Dis. 4:122-132. 1960.
lower dose of 100 EID50 virus per egg. At 12 days PI, the embryos 17. van der Heide, L. The fluorescent antibody technique in the diagnosis of
are examined for specific AE lesions: muscular dystrophy and avian encephalomyelitis. Tech. Bull. No. 44. Life Sciences and Agr. Expt.
embryo immobility. If all embryos have AE lesions, the breeder Sta., University of Maine, Orono, Maine. 1970.
flock is considered to be completely AE-susceptible. If 70%-100% 18. Van Roekel, H„ K.l. Bullis, O. S. Hint, and Μ K. Clarke. Avain
Encephalomyelitis. Annu. Rept. Bull. No. 388. Mass. Agric. Exp. Sta.,
of the embryos are without lesions, the breeder flock is considered
University of Massachusetts, Amherst, Mass. 1942.
to be adequately immune. The percentage of embryos without
lesions should be reported in the results.

174
37
DUCK HEPATITIS
Peter R. Woolcock

SUMMARY. Duck hepatitis can be caused by at least three different viruses. The more common and internationally widespread is duck
hepatitis virus (DHV) type I (a picomavirus, most closely related to members of the genus Parechovirus (9)), which causes a highly lethal,
acute, contagious infection in ducklings less than 6 wk of age and frequently less than 3 wk old. DHV type I does not cause disease in older
ducks. DHV type Π has only been reported in the United Kingdom. It occurs in ducklings from 10 days to 6 wk of age, and causes pathologic
changes similar to those of DHV type I. DHV type Π is classified as an astrovirus. DHV type HI has been reported only in the United States
and causes liver lesions in young ducklings, but is less virulent than DHV type I. DHV type HI is believed to be a picomavirus, serologically
unrelated to DHV type I virus.
Agent Identification. Diagnosis of hepatitis in ducklings is based upon the characteristic disease pattern in the flock, gross pathological
changes, recovery of virus from dead ducklings, and reproduction of the disease in susceptible ducklings. It is not possible to distinguish
between DHV types I, Π, and ΙΠ on the basis of clinical findings and pathology; however, distinctions can be made from the responses of
ducklings, embryonating eggs, and cell cultures to the virus isolates. A one-step reverse transcriptase polymerase chain reaction for DHV-1
has been reported (10).
Serologic Detection in the Host. Serologic tests have little value in the diagnosis of acute infections caused by DHV types I, Π, and HL
Serum neutralization tests in ovo have been used with all three viruses and in vitro tests have been developed for DHV type 1. These tests
have been used for the assay of immune responses to vaccination and epidemiologic surveys, as well as for virus identification.

INTRODUCTION Duck Hepatitis Virus Type HI


Duck hepatitis virus type ΙΠ causes clinical signs and pathologic
Duck hepatitis virus (DHV) types I, Π, and HI are the cause of changes in young ducklings similar to those seen in DHV type I
acute, rapidly spreading, fatal viral infections in young ducklings, infections (1). DVH type HI has been reported only in the United
characterized primarily by hepatitis. DHV types Π and HI were States, where losses of up to 20% have occurred in ducklings
recognized as separate entities because they induced hepatitis in immune to DHV type I (6).
ducklings immune to DHV type I. The duck is the only known
natural host for all three viruses; these diseases are of no known SAMPLE COLLECTION
public health significance. The diseases are economically important
to all duck growers because of their potentially high mortality if not In infected ducklings, viremia occurs before clinical signs become
controlled. apparent. Virus can be recovered from any vital organ, blood, and
Duck hepatitis virus types I, Π, and ΠΙ should not be confused with feces. Because Ever is the target organ, virus can usually be
duck hepatitis B virus, a hepadnavirus classified in the same group recovered by homogenization of liver tissue as a 20% (w/v)
as mammalian hepatitis B virus. This virus seems to have minimal suspension in buffered saline. The suspension is clarified by low-
significance for commercial duck production (4, 12). speed centrifugation.

CLINICAL DISEASE PREFERRED CULTURE MEDIA AND SUBSTRATES

Duck Hepatitis Virus Type I Duck Hepatitis Virus Type I


Duck hepatitis virus type I affects ducklings less than 6 wk of age The presence of DHV type I may be confirmed by one or more of
in virtually all duck-rearing countries and causes the most common the following procedures:
form of duck hepatitis encountered. The clinical disease is 1) Subcutaneous (SC) or intramuscular (IM) inoculation of the
characterized by sudden onset and rapid spread. Ducklings show isolate into l-to-7-day-old DHV type I-susceptible ducklings.
signs of lethargy and ataxia, lose their balance, fall on their sides, This is probably the most sensitive and reHable method. The
and kick spasmodically before death, which may occur in as little as characteristic clinical disease should follow, with deaths often
1-2 hr following the onset of signs. At death, the head is usually occurring within 24 hr of inoculation. Ducklings should show the
drawn back in the opisthotonos position. Practically all mortality in gross lesions associated with DVH type I. Virus should be isolated
a flock will occur within 3-4 days, with the majority of deaths on from Evers to confirm the diagnosis.
the second day. Gross pathologic changes appear primarily in the 2) Inoculation of serial dilutions of Ever homogenate into the
liver, which becomes enlarged with distinct punctate and aUantoic sacs of embryonating duck eggs (aged 10-14 days) from a
ecchymotic hemorrhages. Splenomegaly and swollen kidneys with DHV type I-free flock, or chicken eggs (aged 8-10 days). DHV
some congestion of renal blood vessels may also be seen (14). type I-infected duck embryos should die within 24—72 hr; chicken
Duck Hepatitis Virus Type II embryos are more variable and erratic in their response and usually
Duck hepatitis virus type Π produced clinical signs and pathologic take 5-8 days to die. The allantoic fluid is opalescent or a pale
changes similar to DHV type I in fattening ducks up to 6 wk of age. greenish yeEow. Gross pathologic changes in the embryos include
It has only been reported in the United Kingdom (5). Affected birds stunting, subcutaneous hemorrhages over the whole body, and
may show signs of polydypsia, have loose droppings, and show edema, particularly of the abdominal and hind limb regions. The
excessive urate excretion. They usually die within 1-2 hr following embryo Evers may be swollen, red and yeEowish in color, and show
the onset of signs. Gross pathologic changes are similar to those some necrotic foci. The Ever lesions and stunting become more
produced by DHV type I, but the enlarged spleens may have apparent in embryos that take longer to die.
scattered pale foci. The alimentary tract is often empty, although the
small intestine may contain mucus; hemorrhagic areas on the
intestinal wall are sometimes seen. Petechial hemorrhages may
occasionally be seen on the heart (5, 6).
175
Peter R. Woolcock

3) Inoculation of primary cultures of duck embryo liver cells (15) Wescodyne (an organic iodine solution; Penetone/West, Tenafly,
with serial dilutions of liver homogenate containing DHV type I N.J.) and undiluted Clorox (sodium hypochlorite, Clorox Company,
cause a cytopathic effect (CPE) that is characterized by cell Oakland, Calif.) (14).
rounding and necrosis. The cell monolayer at the time of infection Duck Hepatitis Virus Type Π. Duck hepatitis virus type Π is
must be washed free of mammalian sera, which inhibit virus stable at 50 C for 60 min. Formaldehyde fiimigation and standard
adsorption (3). When the cultures are overlaid with a maintenance disinfection procedures have eliminated the infection from
medium containing 1% agarose (w/v), the CPE appears as plaques; contaminated premises (5).
the size of the plaques may be controlled by the addition of 0.1%- Duck Hepatitis Virus Type HL Duck hepatitis virus type HI is
0.2 % fetal calf serum (FCS) to the medium. Plaques appear after inactivated at 50 C irrespective of the presence of MgCl2.
72-96 hr incubation.
Biological Properties
Duck Hepatitis Virus Type Π All three DHV types lack the ability to agglutinate avian or
The presence of DHV type Π may be confirmed by one or more of mammalian erythrocytes.
the following procedures:
1) Inoculation (SC or IM) of 1 to 7-day-old susceptible ducklings. A Strain Variability
mortality rate of up to 20% may occur within 2-4 days but the Duck Hepatitis Virus Type I. Duck hepatitis virus type I, often
response may be variable. The gross pathology is similar to that referred to as classical DHV, is genetically highly stable. Viruses
observed in field cases (5). serologically distinct from type I have been reported from India and
2) Inoculation into embryonating chicken or duck eggs, via either Egypt, but their relationship to other DHV types is unknown. An
the amniotic cavity or yolk sac. Usually no embryo deaths occur apparent serologic variant, Bamhardt, has been isolated from one
during early passages, but some mortality and pathologic changes farm in the United States. It shows partial cross-neutralization with
may occur after four passages. Embryos take 6-10 days to show DHV type 1(1, 13).
evidence of infection; when this occurs, the embryos are stunted Duck Hepatitis Virus Type Π. Duck hepatitis virus type Π,
and have green, necrotic livers (6). originally thought to be a variant of type I, is serologically distinct
and is now classified as a separate virus type, namely an astrovirus.
Duck Hepatitis Virus Type HI It has only been isolated from ducklings in the United Kingdom. No
The presence of DHV type HI may be confirmed by one or more serologic variants of type Π virus have been isolated.
of the following procedures: Duck Hepatitis Virus Type HI. Duck.hepatitis virus type HI has
1) Intramuscular or preferably intravenous inoculation of liver been reported only in the United States. There are no serologic
suspension into susceptible 1-day-old ducklings. The mortality rate cross-reactions with DHV types I or Π. Although type HI virus has
may reach 20%, with 60% morbidity. No deaths occur in the first 24 been isolated from more than one geographic region of the United
hr, and all virus-specific losses occur between the second and fourth States, no variants of type HI have been reported.
days following inoculation.
2) Inoculation of the suspension onto the chorioallantoic membrane Characteristics of Vaccine Strains
(CAM) of 10—day—old embryonating duck eggs. The response is Duck Hepatitis Virus Type I. A live-virus vaccine containing
erratic, but some embryo mortality usually occurs within 7-10 days. DHV type I, for use in breeder ducks and in susceptible ducklings,
Infected CAMs have a dry, crusty appearance, beneath which they has been produced from attenuated DHV type I passaged more than
are edematous. The embryos may be stunted and edematous with 80 times in embryonating chicken eggs. The vaccine strain is lethal
hemorrhages in the skin. The liver, kidneys, and spleen become for 8 to 10-day-old chicken embryos, causing mortality and lesions
enlarged. Attempts to cultivate the virus in embryonating chicken within 48-72 hr of inoculation. Lesions seen in the dead embryos
eggs have not been successful. include stunting, subcutaneous hemorrhaging, edema, hemorrhages
in the liver and kidney, and occasional greenish discoloration of the
AGENT IDENTIFICATION liver. Vaccine virus cannot be distinguished from the duckling-
virulent virus on the basis of the types of lesions produced in
Morphology chicken embryos, but the time frame for the development of lesions
All three DHV types are small nonenveloped RNA viruses. DHV in embryos is usually shorter with the vaccine virus than with the
type I has been classified in the family Picornaviridae, most closely duckling-virulent virus. An inactivated DHV type I vaccine has also
related to members of the genus Parechovirus (9). Virus particle been described (16). Both live and inactivated DHV type I vaccines
size has been reported as ranging between 20-40 nm in diameter are licensed for use in the USA.
(14). DHV type Π has astrovirus-like morphology and virions are Duck Hepatitis Virus Type Π. A live attenuated virus vaccine has
28-30 nm in diameter. It is classified in the family Astroviridae as been used experimentally under field conditions to protect
duck astrovirus I (DAstV-I) (11). DHV type ΙΠ is thought to be a ducklings (5). The vaccine virus originated from a field isolate and
picomavirus unrelated to DHV type I and has a particle size of was attenuated by 25 serial passages in embryonating chicken eggs
approximately 30 nm diameter (8). (6). This experimental vaccine, developed in the United Kingdom,
is not licensed for use in the United States.
Physicochemical Properties Duck Hepatitis Virus Type HI. A live attenuated DHV type HI
All three DHV types are resistant to pH 3, trypsin, and lipid vaccine has been used in breeder ducks to provide maternal
solvents (e.g., chloroform). The resistance to chloroform is immunity in newly hatched ducklings. The vaccine, prepared with
advantageous, allowing the purification of infectious viruses while virus that was attenuated by 30 serial passages in embryonating
removing high lipid content of duckling liver samples. Chloroform duck eggs, is still experimental and is not currently licensed for use
acts not only as a lipid solvent but also a protein precipitant. in the United States.
Duck Hepatitis Virus Type L Duck hepatitis virus type I is stable
at 50 C for 60 min, with heat stability unaffected by 1 M divalent Differentiation of DHV Isolates. Duck hepatitis virus type I
cations (Mg2+). The virus, which is highly stable under adverse usually runs a peracute course, and mortality up to 100% within 2-3
environmental conditions, can be inactivated with 1% formaldehyde days of onset is not uncommon. The sudden onset, rapid spread, and
and 2% caustic soda in 2 hr, 2% calcium hypochlorite in 3 hr, acute course of this disease are characteristic. Hemorrhagic lesions
chloramin in 5 hr, 0.2% formalin in 2 hr, 5% phenol, undiluted in the livers of ducklings less than 3 wk of age are practically
176
Chapter 37 Duck Hepatitis

pathognomonic. DHV type ΙΠ rarely causes mortality above 20%. and RT -PCR buffer (50 mM Tris-HCl and 75 mM KC1). In
Mortality caused by DHV type Π ranges between 10% and 50%. addition, the following components were included in the reaction: 4
Duck hepatitis virus type I is the only type to produce a CPE in μΐ (50 ng) RNA template, 1 μΐ (10 pmol μΓ1) of each specific
duck embryo liver cells. DHV type Π may be distinguished by its primer, and DEPC-treated dH2O to a total reaction volume of 20 μΐ.
distinctive morphology by negative stain electron microscopy. DHV A T gradient thermocycler was used for one-step RT-PCR. Reverse
type ΙΠ is the only type to cause drying and thickening of the CAM transcription was performed at 45 C for 30 min, after which the
in embryonating duck eggs. When a mixed infection occurs with enzyme was inactivated at 94 C for 5 min. PCR amplification was
type I and type Π or type ΙΠ virus, methods other than cultural conducted using an initial denaturation for 20 s at 94 C, followed by
techniques must be used, because DHV type I is more virulent and 40 cycles of annealing for 30 s at 52 C, extension for 30 s at 72 C
overwhelms the other virus types in its expression. and denaturation for 20 s at 94 C, and a final extension for 5 min at
72 C. The PCR products (10 μΐ) were separated by electrophoresis
Antigen Detection (100 V) in horizontal 1.5% agarose gels and Tris-acetate buffer (40
Duck Hepatitis Virus Type I. Various serologic tests have been mM tris-acetate, 1 mM EDTA). Gels were stained with ethidium
used for virus identification: bromide (0.5 pg ml'1), visualized under UV light and photographed.
1) One-to-7-day-old ducklings susceptible to DHV type I are The DHV-1 sequences used in this report have been assigned the
passively immunized (SC) with 1-2 ml specific hyperimmune following accession numbers in GenBank: DQ219396, DQ226451,
serum or specific egg-yolk antibody and then challenged IM or SC DQ256132, DQ256133, DQ256134.
24 hr later with 0.2 ml of the virus isolate (titer not determined). A
control group of uninoculated ducklings is similarly challenged. SEROLOGIC DETECTION IN THE HOST
Virus identification is based on 80%-100% survival in the passively
immune ducklings and 80%-100% mortality in the controls. Serologic tests have little value in the diagnosis of acute infections
2) One-to-7-day-old DHV type I-susceptible and DHV type I- caused by DHV types I, Π, and HL
matemally immune ducklings are challenged IM or SC with 0.2 ml All three DHV types have been used in virus-neutralization tests in
of the virus isolate (titer undetermined). Identification is based on ovo, but their success depends on the expression of the virus in the
80%-100% losses in the susceptible ducklings and 80%-100% assay system used; with type Π and ΙΠ viruses this can be a
survival in the maternally immune ducklings. problem. In vitro tests have been developed for DHV type I; these
3) Serial 10-fold dilutions of the virus isolate are mixed with equal include a plaque reduction assay and a microtiter assay (14, 15).
volumes of DHV type I-specific hyperimmune serum diluted 1:5 or The plaque reduction assay may be performed using either primary
1:10. The mixtures are allowed to react at 37 C or room temperature DEK or DEL cells. Primary cell culture monolayers are prepared in
for 1 hr and then are inoculated into susceptible ducklings (0.2 ml Eagles minimum essential medium* (EMEM) containing 5%—10%
SC), into the allantoic cavity of embryonating duck eggs (0.2 ml), FCS, 2 mM glutamine, 0.17% sodium bicarbonate, and gentamicin.
and onto primary duck embryo liver (DEL) cell monolayers (2, 15). Trypsinized cells are seeded into 5-cm-diameter petri dishes, and
Controls in each case consist of the virus isolate mixed with normal then incubated at 37 C in a 5% CO2 atmosphere. Monolayers should
control serum. be nearly confluent at 24-48 hr post-seeding. The monolayers are
Duck Hepatitis Virus Type Π. Serologic techniques have not washed twice with serum-free EMEM or Hanks’ balanced salt
been employed routinely because the immunologic response to solution to remove all traces of FCS before infecting with DHV
infection of both ducklings and duck embryos is poor. However, a type I. Equal volumes of DHV type I suspended in serum-free
neutralization assay has been used for virus identification. Chicken EMEM, adjusted to 200 plaque-forming units per 0.1 ml, are mixed
embryos are inoculated via the amniotic cavity with constant-serum with equal volumes of serially diluted duck sera (twofold dilutions
varying-virus mixtures (5). Cross-protection tests have been in EMEM). The serum samples should be heat inactivated at 56 C
performed in 2 to 4-day-old ducklings; these are inoculated with for 30 min before testing. The virus-serum mixtures are incubated at
antisera to types I and Π, and then challenged 3 days later with the 37 C for 1 hr, and then 0.1-ml aliquots are added to the confluent
virus isolate (5). DHV type Π can be detected by negative stain cell monolayers, three dishes per dilution. The plates are left for 30
electron microscopy of liver and fecal preparations, and has min at room temperature (20-22 C), and then overlaid with agarose
astrovirus-like morphology (5, 6). maintenance medium (EMEM containing 2% chicken serum and
Duck Hepatitis Virus Type HI. The lower incidence of mortality 0.1%-0.2% FCS to which agarose had been added to a final
(maximum of 20%) in ducklings that are experimentally infected concentration of 1% [w/w]). The plates are then placed at 37 C in a
with DHV type ΙΠ makes virus-neutralization tests in vivo difficult. 5% CO2 atmosphere. The number of plaques produced is recorded
A virus-neutralization test using the constant-serum varying-virus after 48 hr of incubation. Plaques may be observed using an oblique
method is possible in embryonating duck eggs inoculated by the light source, or alternatively, monolayers may be fixed with 10%
dropped-CAM route. Attempts to induce a CPE with the virus in formol-buffered saline and stained with 1% crystal violet. Serum
cell cultures have not been successful, although the virus has been antibody titers are expressed as the reciprocal of the highest serum
detected by direct immunofluorescence in experimentally infected dilution that reduces the plaque count by 50%.
cell cultures of duck embryo liver and duck embryo kidney (DEK) A microtiter neutralization assay may be performed using primary
(8). DEK cells. Serial twofold dilutions of each serum sample (heat
inactivated) are prepared in 50 μΐ of serum-free basal medium,
Molecular identification Eagle (BME) in microtiter plates. Approximately IO20 median
A one-step reverse transcriptase polymerase chain reaction for tissue culture infective doses of DHV type 1 in 50 μΐ of BME are
DHV-1 has been reported (10). The assay is based on primers added to each well and the mixtures are allowed to react at 37 C for
specific to the sequence coding for the 3D protein. Primers 1 hr. Primary DEK cells are suspended in BME supplemented with
designated DHV-1 ComF (5’-AAG AAG GAG AAA ATY(C or T) 10% tryptose phosphate broth, 2 mM L-glutamine, 0.17% sodium
AAG GAA GG-3’) and DHV-1 ComR (5’- TTG ATG TCA TAG bicarbonate and 2%-4% chicken serum, and are adjusted to contain
CCC AAS(C or G) ACA GC-3’) flank a 467 base pair DNA 3 x 105 cells per milliliter. Cells are next added to the plates at 100
sequence in the 3D gene. One-step RT-PCR was conducted using μΐ per well and the plates are then incubated for up to 96 hr at 37 C
the Maxime RT-PCR PreMix kit (iNikron Biotechnology, Korea). in a humidified 5% CO2 atmosphere. Following incubation, cells
The 20 μΐ reaction mixtures contained 1U of OptiScript Reverse are fixed with 10% formol-buffered saline and stained with 1%
Transcriptase, 2.5 mM dNTPs, 2.5 U i-StarTaq DNA polymerase crystal violet. The plates are read macroscopically. The titer for
177
Peter R. Woolcock

virus neutralizing activity is expressed as the reciprocal of the 5. Gough, R. E., E. D. Borland, I. F. Keymer, and J. C. Stuart An outbreak
highest dilution of serum at which a monolayer grew, that is, there of duck hepatitis type II in commercial ducks. Avian Path 14:227-236. 1985.
is no evidence of CPE and therefore complete virus neutralization 6. Gough, R. E., and J. C. Stuart Astroviruses in ducks (duck virus hepatitis
type Π). In: Virus infections of birds. J. B. McFerran and M S. McNulty,
has occurred. A titer of less than 4 log2 is considered negative. eds. Elsevier Science Publishers, Β. V., Amsterdam, pp 505-508. 1993.
These neutralization tests have been used to assay humoral immune 7. Gough, R. E., and A. S. Wallis Duck hepatitis type I and influenza in
responses to vaccination and for epidemiologic surveys, as well as mallard ducks (Anas platyrhynchos). Vet Rec 119:602. 1986.
for virus identification. 8. Haider, S. A., and B. W. Calnek In vitro isolation, propagation, and
characterization of duck hepatitis virus type ΙΠ. Avian Dis 23:715-729.
DIFFERENTIATION FROM CLOSELY RELATED AGENTS 1979.
9. Kim, Μ-C., Y.-K. Kwon, S.-J. Joh, A. M Lindberg, J.-H. Kwon, J.-H.
Most viral pathogens other than DHV do not cause disease in Kim, and S.-J. Kim Molecular analysis of duck hepatitis virus type 1 reveals
a novel lineage close to the genus Parechovirus in the family
ducks less than 6 wk old; all three types of DHV are resistant to Picornaviridae. Journal of General Virology 87:3307-3316. 2006.
chloroform, which will inactivate most other viral pathogens, for 10. Kim, Μ-C., Y.-K. Kwon, S.-J. Joh, J.-H. Kwon, J.-H. Kim, and. S-J.
example, duck virus enteritis, Newcastle disease virus, and avian Kim. Development of one-step reverse transcriptase-polymerasae chain
influenza virus. Disease outbreaks caused by serologic variants of reaction to detect duck hepatitis virus type 1. Avian Dis 51. 2007.
DHV type I, type Π, or type HI can present a problem in 11. Koci, M D., and S. Schultz-Cherry. Avian astroviruses. Avian Pathol
determining an etiology, particularly when attenuated classic DHV 31:213-227. 2002.
type I is present as a result of vaccination. Dual infections of DHV 12. Mason, W. S., G. Seal, and J. Summers Virus of Pekin ducks with
type I and Chlamydophyla psittaci (2) and influenza virus (7) have structural and biological relatedness to human hepatitis B virus. J Virol
36:829-836. 1980.
also been reported. Other causes of acute mortality in ducklings 13. Sandhu, T. S., B. W. Calnek, and L. Zeman Pathologic and serologic
include salmonellosis and aflatoxicosis. The latter disease may characterization of a variant of duck hepatitis type I virus. Avian Dis
cause ataxia, convulsions, and opisthotonos, as well as microscopic 36:932-936. 1992.
lesions of bile-duct hyperplasia that are suggestive of duck hepatitis, 14. Woolcock, P. R. Duck Hepatitis. In: Diseases of Poultry. S. Y.M, H. J.
but aflatoxicosis does not cause die same characteristic Bames, J. R. Glisson, A. M Fadly, L. R. McDougald and D. E. Swayne,
hemorrhages in the liver. eds. Iowa State Press, pp 343-354. 2003.
15. Woolcock, P. R. An assay for duck hepatitis virus type I in duck embryo
REFERENCES liver cells and a comparison with other assays. Avian Path 15:75-82. 1986.
16. Woolcock, P. R. Duck hepatitis virus type I: Studies with inactivated
1. Calnek, B. W. Duck virus hepatitis. In: Virus infections of birds. J. B. vaccines in breeder ducks. Avian Path 20:509-522. 1991.
McFerran and M S. McNulty, eds. Elsevier Science Publishers Β. V.,
Amsterdam, pp 485-495. 1993.
2. Chalmers, W. S., H. Farmer, and P. R. Woolcock Duck hepatitis virus
and Chlamydia psittaci outbreak [letter]. Vet Rec 116:223. 1985.
3. Chalmers, W. S. K., and P. R. Woolcock The effect of animal sera on
duck hepatitis virus. Avian Path 13:727-732. 1984.
4. Femholz, D., H. Wetz, and H. Will Hepatitis B Viruses in birds. In: Virus
infections of birds. J. B. McFerran and M S. McNulty, eds. Elsevier Science
Publishers, Β. V, Amsterdam, pp 111-119. 1993.

178
38
TURKEY VIRAL HEPATITIS
Willie M. Reed

SUMMARY. Turkey viral hepatitis (TVH) is a highly contagious, often subclinical disease of turkey poults fewer than 5 wk of age. Because
of the clinical nature of the disease and lack of a readily available serologic assay, the true incidence of infection is unknown. Turkey viral
hepatitis is characterized by multifocal hepatic and pancreatic necrosis, frequently accompanied by inflammatory cells composed
predominantly of lymphocytes and macrophages and lesser numbers of heterophils. The etiologic agent has not been fully characterized but
has morphologic features of a picomavirus.
Agent Identification The virus can be propagated by yolk sac inoculation in 5 to 7 day-old embryonated chicken embryos. Diagnosis is
based on the presence of typical gross and microscopic lesions in the liver and pancreas of affected poults, lack of isolation of other bacterial
and viral pathogens, and rarely by isolation of the virus.
Serologic Detection in the Host. There are no readily available serologic assay.

INTRODUCTION necessary. The supernatant from centrifugation for 20 min at 1500 x


g is injected in 0.2 ml amounts into the yolk sac of 5 to 7 day-old
Turkey viral hepatitis results from infection by picomavirus-like chicken embryos. Embryos are observed for 10 days. Dead embryos
virus that produces hepatic and pancreatic lesions only in turkeys (have cutaneous congestion and hemorrhage.) Mortality usually
(1,2,3,4). The infection is frequently subclinical and occurs only in occurs between 5 and 10 days postinoculation, but low virus titers
turkey poults where poorly defined stressors are necessaiy for in the inoculum may require a second passage of yolk harvest
clinical expression. Chickens, quail, pheasants, ducks, mice, and before a typical mortality pattern develops. Embryonated turkey
rabbits are refractory to infection. It is not of major economic embryos up to 10 days of age are suitable for virus propagation but
importance, and a high standard of sanitation and good husbandry are not the preferred culture system, due to the possible presence of
minimize the effects of the disease. Turkey viral hepatitis was first maternal antibodies. Turkey viral hepatitis virus has not been
described in North America in 1959 and occurs in most, if not all, propagated in cell culture.
areas of intensive commercial turkey production (5,7). It has also
been reported in Italy and the United Kingdom (2). AGENT IDENTIFICATION

CLINICAL DISEASE AND PATHOLOGY The turkey hepatitis virus has not been fully characterized but is
reported to have morphologic features of a picomavirus. It is
Clinical disease is generally seen only in turkeys fewer than 5 wk resistant to chloroform, ether, phenol, and creoline but not to
of age and is marked by sudden mortality, ranging to 25% (2). formalin. The virus will survive in yolk for 6 hr at 60 C, 14 hr at 56
However, morbidity rates of up to 100% have been described in C, and 4 wk at 37 C. It will also survive for 1 hour at pH 2 but not
some flocks (2). Mortality is largely confined to a 4 to 8-day period. at pH 10 (8,9). It passes through a 0.1 nm membrane. Lesions in
Gross lesions of TVH are confined to the liver and pancreas. The infected embryos, while highly suggestive of TVH, are not
liver is generally enlarged and contains disseminated, focal to definitive, and while the finding of characteristic lesions in
coalescing, gray and sometimes depressed foci that may be diagnostic specimens allows for a strong presumptive diagnosis,
obscured by the presence of congestion and foci of hemorrhage. added assurance is secured by reproducing the disease. At least 6,
Pancreatic lesions occur less frequently than hepatic lesions. one to 7-day-old poults are injected intramuscularly,
Lesions in the pancreas occur most commonly on the dorsal surface subcutaneously, or intraperitoneally with 0.2 to 0.5 ml of liver
and consist of round to oval, gray to white foci. suspension (preferably) or with yolk fluid from infected embryos
Microscopic lesions in the Ever are characterized initially by (6). As clinical signs seldom develop, one or more poults should be
vacuolation of hepatocytes followed by dense infiltration with euthanized and examined daily, starting 5 to 7 days after
macrophages, lymphocytes, and mononuclear cells and by inoculation. A comparable isolated control group should likewise be
proliferation of bile ductules. As the lesions progresses, there is examined to preclude preexisting infection. Lesions are usually
prominent necrosis with pooling of blood around necrotic foci, much less severe than in natural infections. Neither lesions nor viral
where separated hepatocytes frequently fuse to form syncytial cells. replication occur in chicks.
Inclusion bodies have not been detected. Pancreatic lesions are
similar and are characterized by acinar cell degeneration and SEROLOGIC DETECTION IN THE HOST
necrosis along with infiltration of macrophages and lymphocytes.
Although vertical transmission of TVH virus is suspected, primarily Although substantial resistance to reinfection has been reported,
based on field observations, this is not proven. no means of serologic identification has been developed. The
characteristic low titer of the virus in yolk following embryo
SAMPLE COLLECTION propagation, usually less than a mean embryo infectious dose of
3.5, has severely hampered further study.
Livers with characteristic lesions are preferred, although isolation
of the virus can be made from a variety of tissues, including DIFFERENTIATION FROM CLOSELY RELATED AGENTS
pancreas, spleen, and kidney, and from the feces. Refrigerated,
intact, dead poults are preferred, but aseptically collected, Bacterial infections, particularly from Salmonella sp., Pasteurella
refrigerated or frozen livers with lesions are satisfactory. multocida, or Escherichia coli, and infections caused by Group 1
and Group 2 avian adenovirus, reovirus, and Histomonas
PREFERRED CULTURE MEDIA AND SUBSTRATES meleagridis must be ruled out.

Tissue homogenates should be prepared for embryo inoculation by


grinding in 5-10 parts of nutrient broth or phosphate-buffered
saline. Antibiotics and antifungals, to control possible
contaminating bacteria and fungi, may be used but are seldom
179
Willie M Reed

ACKNOWLEDGEMENT 4. MacDonald, J. W., C. J. Randall, and M D. Dagless. Picoma-like virus


The contributions of the late Dr. G. H. Snoyenbos, a previous author of this causing hepatitis and pancreatitis in turkeys. Vet. Rec. Ill :322. 1982.
chapter, are acknowledged and appreciated. Mongeau, J. D., R. B. Truscott, A. E. Ferguson, and M C. Connell. Virus
hepatitis in turkeys. Avian Dis. 3:388-396. 1959.
REFERENCES 5. Snoeyenbos, G. H., and Η. I. Basch. Further studies of virus hepatitis in
1. Andral, Β., M Lagadic, G. Bennezeon, D. Toquin, and J. M Florent. turkeys. Avian Dis. 4:477-485. 1960.
Picoma-like viruses of young turkeys: Pathogenesis of a disease of poults 6. Snoeyenbos, G. Η., Η. I. Basch, and M Sevoian. An infectious agent
caused by a picoma-like virus. Avian Pathol. 19:245-254. 1990. producing hepatitis in turkeys. Avian Dis. 3:377-388. 1959.
2. Guy, S. J. Turkey viral hepatitis. In: Diseases of Poultry, 10th ed. B. W. 7. Tzionabos, T., and G. H . Snoeyenbos. Some physicochemical properties
Calnek, H. J. Bames, C. W. Beard, L. R. McDougald, and Y. M Saif, eds. of turkey hepatitis virus. Avian Dis. 9:152-156. 1965.
Iowa State University Press, Ames, Iowa, pp. 773-777,1997. 8. Tzionabos, T., and G. H. Snoyenbos. Clinical, immunological, and
3. Klein, P. N., A. E. Castro, C. U. Meteyer, B. Reynolds, J. A serological observations on turkey virus hepatitis. Avian Dis. 9:578-591.
Swartzmann-Andert, G. Cooper, R. P. Chin, and H. L. Shivaprasad. 1965.
Experimental transmission of turkey viral hepatitis to day-old poults and
identification of associated viral particles resembling picomaviruses. Avian
Dis. 35:115-125. 1991.

180
39
VIRAL ARTHRITIS/TENOSYNOVITIS AND OTHER REOVIRUS INFECTIONS
John K. Rosenberger and Erica Spackman

SUMMARY. Viruses of the family Reoviridae, genus Orthoreovirus, infect a variety of bird species including commercial poultry. Viral
arthritis/tenosynovitis in poultry is the most important pathologic manifestation of avian reovirus infection. A malabsorption syndrome in
chickens that includes stunting, diarrhea, osteoporosis, and fragmentation of the proximal end of the femur has also been associated with
reovirus infection. However, several different etiologic factors have been implicated in malabsorption syndrome. Other disease conditions
associated with reovirus infections include hepatitis, myocarditis, hydropericardium, and respiratory and intestinal tract involvement. Many
reovirus infections are subclinical.

VIRAL ARTHRITIS/TENOSYNOVITIS
Agent Identification. Presumptive diagnosis is based on gross lesions including bilateral enlargement of the shank and tendon bundles
above the hock in chickens, and lesions in chicken embryos inoculated at 5 to 7 days of embryonation via the yolk sac, or cytopathic effects
and syncytia formation in chicken kidney cell cultures. Confirmation of reovirus is through demonstration of specific antigens in embryos,
synovium or cell cultures, by agar gel immunodiffusion, or fluorescent antibody tests.
Serologic Detection in the Host. ELISA, virus neutralization and agar gel immunodiffusion tests can be used to document past infections.
However, because reovirus infections can be subclinical and are ubiquitous, flock history and other information must be evaluated before
interpreting serologic results.

OTHER SYNDROMES
Agent Identification. Diagnosis is best made by virus isolation in embryos or cell cultures with confirmation by detection of reovirus
specific antigens. Experimental reproduction of the field syndrome may be necessary to determine if reovirus is the etiology.
Serologic Detection in the Host. ELISA, virus neutralization and agar gel immunodiffusion tests can be used to document past infections.
However, because reovirus infections can be subclinical and are ubiquitous, flock history and other information must be evaluated before
interpreting serologic results.

INTRODUCTION
Malabsorption Syndrome
Avian reoviruses are prevalent in chickens, turkeys, and other Reovirus infections have been associated with malabsorption
avian species (13,15,16,17,21,26). Avian reoviruses have been syndrome, but their exact etiologic relationship remains unclear.
isolated from chickens affected by an assortment of disease Clinical signs in 1 to 3 wk-old chickens attributed to malabsorption
conditions, including viral arthritis/tenosynovitis (5,9,10,13,15,22), syndrome include poor pigmentation, abnormal feathering,
stunting syndrome (14), respiratory disease (3,6,12,16), enteric osteoporosis, uneven growth, undigested feed in the feces, and
disease (2,6), malabsorption syndrome (11,14), and osteoporosis increased mortality (11,14,25). Lesions may include an enlarged
(23)
. In addition, avian reoviruses have been found frequently in proventriculus, catarrhal enteritis, and lameness associated with
clinically unaffected chickens. The severity and nature of the tenosynovitis.
disease, such as viral arthritis/tenosynovitis that occurs following Lesions observed histopathologically include proventriculitis,
reovirus infection, depend upon host age, virus virulence, and route myocarditis, atrophy of the cloacal bursa (bursa of Fabricius),
of exposure. Turkeys infected with reoviruses may present with pancreatitis, enteritis, and atrophy of intestinal villi (11,14). Clefts
similar disease conditions to those described in chickens and in the femoral growth plate with necrosis of the cartilage and
disease presentation is strain dependent. However, strains that fragmentation of the femoral heads have also been reported (11).
induce a particular disease condition in chickens will not
necessarily induce similar disease in turkeys and vice versa (1, 19). SAMPLE COLLECTION
Therefore, the pathogenic potential of avian reovirus isolates must
be evaluated in their species of origin. The synovial fluid from the tibiotarsal or tibiofemoral joints
should be collected with a sterile swab, or a 10% extract of
CLINICAL DISEASE edematous synovium (tendon sheaths) in nutrient broth or cell­
culture medium prepared with a Tenbroeck grinder (VWR
Viral Arthritis/Tenosynovitis Scientific Products, West Chester, Penn.). Swabs or tissue
The most commonly recognized disease that is associated with homogenates can also be taken from the spleen, cloaca, or trachea.
avian reovirus is viral arthritis/tenosynovitis. The principal signs in The digestive and respiratory phases of reovirus infections are of
4 to 7-wk-old broilers are bilateral enlargement of the shank and the short duration making virus isolation from those tissues difficult.
tendon bundle above the hock, causing limited tendon movement When sampling turkeys the bursa of Fabricius should be collected.
and lameness with occasional rupture of the gastrocnemius tendon. Specimens should be stored at -70 C until used, but can be stored at
Hepatitis, myocarditis, and splenitis may be seen in 1 to 7-day-old -20 C for short periods. Repeated cycles of freezing and thawing
chicks. should be avoided.
Microscopically, hypertrophy and hyperplasia of the synovial cells
occurs (24). The synovial lining is infiltrated with heterophils, PREFERRED CULTURE MEDIA AND SUBSTRATES
lymphocytes, plasma cells, and macrophages. In chronic cases,
synoviae develop villus processes with fibrosis of tendon sheaths. The virus can be cultured in the yolk sac or on the chorioallantoic
Granulomatous inflammation may encompass and replace tendons. membrane (CAM) of embryonating chicken eggs, chick embryo
Lesions in the heart consist of heterophil infiltration between the liver cells, or in chicken kidney cells (CKC) (17). No direct
myocardial fibers, which may be accompanied by focal areas of comparisons have been made between the CKC and embryo
proliferating reticular cells. Perivascular accumulations of methods, but for original isolation the yolk-sac route in chicken
lymphocytes are frequent (7). embryos is preferred.
181
John K. Rosenberger and Erica Spackman

Embryo Inoculation Virus Strain Variability


The eggs should come from a reovirus-free, antibody-negative In Japan, five serotypes of avian reoviruses have been described.
flock. The preferred route of inoculation is the yolk sac in 5 to-7 These were isolated from feces, cloacal swabs, and trachea. Their
day-old embryonating chicken eggs; 0.1-0.2 ml of suspect material pathogenicity for the synovial and tendon tissues of chickens was
is inoculated per egg. The allantoic-sac route of inoculation is not determined (6). In the United States, four serotypes have been
usually not satisfactory. The CAM route can be used to demonstrate identified (16). The isolates from the synovial tissues appeared to be
pock-like lesions and cytoplasmic inclusions. subtypes of a single serotype (17) and are probably similar to other
Yolk-sac inoculation kills the embryo in 3-5 days. The embryo is isolates (5,9). Although the pathogenicity of the avian reoviruses
markedly hemorrhagic or may show a purplish-red discoloration. should be studied further, all isolates were capable of producing an
The internal organs are congested and hemorrhagic. Embryos that arthritis or tenosynovitis on foot-pad inoculation of 2-wk-old
survive until 17-21 days are slightly dwarfed, and the liver, spleen, chickens. The avian reoviruses can be differentiated from human
and heart are enlarged and contain necrotic foci. types 1, 2, or 3 by the inability of the avian reoviruses to
Inoculation on the dropped CAM of 10-day-old embryonated hemagglutinate human O erythrocytes and the inability of human
chicken eggs kills the embryo in 3-5 days, and pock-like lesions types to grow in the yolk sac of chicken embryos. In addition, the
may develop on the CAM. Inclusion bodies can be seen in the agar-gel precipitin and virus-neutralization tests can be used to
mesodermal cells of the CAM by using hematoxylin and eosin stain identify avian reoviruses. Strain differences based on relative
or fluorescein-labeled antibody. pathogenicity can be demonstrated with antigenically similar or
identical reovirus isolates (14).
Cell Culture
Primary CKC from 2 to 6-wk-old chicks are preferred over SEROLOGIC DETECTION IN THE HOST
chicken embryo fibroblasts for the culture of avian reoviruses. The
medium for CKC appears not to be critical. Satisfactory media are The agar-gel immunodiffusion (AGID) test can be used for
minimum essential medium (MEM) or Eagle medium (modified), documenting past infections. The antigen is prepared from ground
with Earl’s salt and glutamine; 10% fetal calf serum and sodium undiluted CAMs from dead embryos that were inoculated when 5-7
bicarbonate at 1.5 g/liter; and for maintenance, 3% calf serum and days old by the yolk-sac route. Noble agar (1.2%) is used, dissolved
sodium bicarbonate at 1.5 g/liter. Antibiotics used per liter of in 0.01 M phosphate buffer at pH 7.2 containing 8% salt and 7.5%
medium are 100,000 units of penicillin, 0.1 g dihydrostreptomycin, glycine. Glycine is not essential but may give a more distinct
and 2.5 mg amphotericin B. A humidified incubator with about 5% reaction. Wells are prepared about 3 mm in diameter and 3 mm
CO2 in air is desirable. apart, one centered in a circle of six. Knowh positive serum samples
Following inoculation of CKC, syncytia formation occurs often as should be placed in wells 1 and 4 so that a line of identity with
early as 24-48 hr. The syncytia float free, leaving holes in the unknown sera can be demonstrated (15). All known avian reovirus
monolayer. The floating syncytia are bound by a membrane serotypes share a common group-specific precipitin antigen.
showing a mass of dead cells with a clear area resembling a halo. Precipitating antibody tends to persist in birds with joint
As the virus becomes more adapted to cell culture, the entire cell involvement but may disappear in 4 wk in many birds, so monthly
sheet may degenerate in 24 hr. testing is desirable in screening for infection.
The neutralization test is best accomplished by using a plaque­
AGENT IDENTIFICATION reduction test in CKC. For chicken serum, twofold dilutions may be
used. Equal amounts of virus containing 100 plaque-forming units
Morphology and Physicochemical Properties are mixed with the serum dilutions and incubated at 37 C for 45
The reoviruses are double-stranded RNA viruses in the family min. A 0.2-ml amount is inoculated onto a CKC monolayer in a 60
Reoviridae, genus Orthoreovirus (4). The nucleic acid type can be x 15-mm petri dish and allowed to adsorb for 1 hr with frequent
determined directly by using metabolic inhibitors (4). Multiplication rotation. Excess fluid is aspirated, and the cells are overlaid with 5
of the virus is not inhibited in the presence of DNA metabolic ml of medium consisting of MEM as listed with 5% calf serum,
inhibitors, actinomycin-D at 0.5 pg/ml, cytosine arabinoside at 100 0.5% lactalbumin hydrolysate, 1% peptone broth, 1.2% purified
pg/ml, or 5-fluoro-2-deoxyuridine at 300 pg/ml in primary CKC agar, 2.2 g/liter sodium bicarbonate, and antibiotics. This is
.
(24) The virus is thermostable and resistant to ether, chloroform, incubated for 5-7 days and overlaid with 2 ml of 0.001% neutral
and a pH of 3.0 (13). Structurally, reovirus particles consist of a 45- red. The plaques are counted in 4—8 hr. The titer of the serum is the
nm core and a 75-nm capsid and often form crystalline arrays in the reciprocal of the highest serum dilution that inhibits 90% of the
cytoplasm of infected cells (24). The capsid is an icosahedron plaques. Serum titers of 40 or more are considered significant.
consisting of 92 capsomeres. Plaque inhibition on the CAM has been used, but the results are
difficult to interpret.
Virus Identification
A direct or indirect fluorescent antibody test with antibody against DIFFERENTIATION FROM CLOSELY RELATED AGENTS
group-specific antigen can be used for agent identification. The
agar-gel precipitin and virus- neutralization tests can also be used to Arthritis/T enosynovitis
identify the virus. Clinical viral arthritis/tenosynovitis must be differentiated from
The direct fluorescent-antibody test is reliable for detecting Mycoplasma synoviae synovitis, bacterial arthritis, and lameness
antigen in infected tissue. The antigen can be demonstrated in the resulting from anatomical deformities. Viral arthritis can be
cytoplasm of infected cells, the synovium, or CKC (6,15). diagnosed in clinical outbreaks when there is a marked bilateral
Molecular methods such as standard and real-time RT-PCR can be enlargement of the digital flexor and metatarsal extensor tendon
used to detect avian reoviruses (18). RNA for virus detection should sheaths. The diagnosis should be confirmed by isolation and
be extracted from the same tissues that are used for virus isolation. identification of the virus. The involved tendon bundle may need
Electropherotyping can be performed by directly running RNA microscopic examination to demonstrate lesions (7). Bacterial
extracted from a specimen on a 2% agarose gel, however high titers arthritis frequently affects a single joint, but dual infection is not
of virus are necessary to visualize the electropherotype (19). uncommon, making identification of the primary etiological agent
difficult.

182
Chapter 39 Viral Arthritis/Tenosynovitis and Other Reovirus Infections

Pathogenic staphylococcus infections are common in affected 10. Olson, N. O., and R. Weiss. Similarity between arthritis virus and
joints; less frequent are Salmonella, Pasteurella, and erysipelas Fahey-Crawley virus. Avian Dis. 16:535-540. 1972.
infections. The pathogenic mycoplasmas {Mycoplasma 11. Page, R. K., O. J. Fletcher, G. N. Rowland, D. Gaudry, and P. Villegas.
Malabsorption syndrome in broiler chickens. Avian Dis. 26:618-624. 1982.
gallisepticum, Mycoplasma synoviae, and Mycoplasma meleagridis)
12. Petek, Μ, B. Felluga, G. Borghi, and A. Baroni. The Crawley agent: a
are frequently responsible for arthritis. Culture and identification of reovirus. Arch. Gesamte Virusforsch. 21:414—424. 1967.
the organisms should be attempted. 13. Robertson, M D., and G. E. Wilcox. Avian Reovirus. Vet. Bull.
Marek’s disease and noninfectious conditions such as rickets, 56:155-174. 1986.
spondylolisthesis, dyschondroplasia, and toxicoses should also be 14. Rosenberger, J. K. Characterization of reoviruses associated with
eliminated as a cause of lameness. runting syndrome in chickens. In: Proceeding No. 66, International Union
of Immunological Societies, Sydney, New South Wales, Australia, pp. 141-
Malabsorption Syndrome 152. 1983.
15. Rosenberger, J. K., and N. O. Olson. Viral arthritis. In: Diseases of
Proventriculitis, growth retardation, and feathering abnormalities
poultry, 10th ed. B. W. Calnek, H. J. Bames, C. W. Beard, L. R_
may be caused by other factors, such as reticuloendotheliosis virus McDougald, and Y. M. Saif, eds. Iowa State University Press, Ames, Iowa,
(25) and mycotoxins (20). Parvovirus (8) and calicivirus (27) have pp. 711-720. 1997.
been implicated as possible etiologies for a malabsorptionlike 16. Sahu, S. P., and N. O. Olson. Comparison of the characteristics of avian
syndrome. reoviruses isolated from digestive and respiratory tract with viruses isolated
from the synovia. Am. J. Vet. Res. 36:847-850. 1975.
Embryo Lesions and Cytopathic Effects in Cell Culture 17. Sahu, S. P., N. O. Olson, and R. W. Townsend. Characterization of
Many viruses cause mortality in chicken embtyos that are avian reoviruses isolated from the synovia and breast blister. Avian Dis.
23:896-903. 1979.
differentiated from reovirus-induced mortality only with difficulty.
18. Spackman, E., D. Kapczynski and H. Sellers. Multiplex Real-time RT-
Poxviruses and infectious laryngotracheitis virus induce pocks on PCR for the Detection of Three Viruses Associated with Poult Enteritis
the CAM that may resemble those associated with reoviruses. Also, Complex: Turkey Astrovirus Turkey Coronavirus, and Turkey Reovirus.
poxviruses, adenoviruses, and herpesviruses may cause unique Avian Dis. 49: 86-91.2005.
cytopathic effects and inclusion bodies like reoviruses in cell 19. Spackman, E., M Pantin-Jackwood, J. M Day and H. Sellers. The
culture. Physicochemical, molecular and/or serologic tests may be Pathogenesis of turkey origin reoviruses in turkeys and chickens. Avian
required for confirming the identity of a reovirus isolate. Path. 34 (4) In Press. 2005.
20. Stuart, B. P., R. J. Cole, E. R. Waller, and R. E. Vesonder.
REFERENCES Proventricular hyperplasia malabsorption syndrome in broiler chickens. J.
Environ. Pathol. Toxicol. 6:369-386. 1986.
1. al Afaleq, A. I. and R. C. Jones. Pathogenicity of three turkey and three 21. van der Heide, L. Viral arthritis/tenosynovitis: a review. Avian Pathol.
chickens reoviruses for poults and chicks with particular reference to 6:271-284. 1977.
arthitis/tenosynovitis. Avian Path. 18, 433-440. 1989. 22. van der Heide, L., M Kalbac, and M Brustolon. Development of an
2. Deshmukh, D. R., and B. S. Pomeroy. Avian reoviruses. ID. Infectivity attenuated apathogenic reovirus vaccine against viral arthritis/tenosynovitis.
and egg transmission. Avian Dis. 13:427—439. 1969. Avian Dis. 27:698-706. 1983.
3. Fahey, J. E., and J. F. Crawley. Studies on chronic respiratory diseases of 23. van der Heide, L., D. Lutticken, and M Horzinek Isolation of avian
chickens. 2. Isolation of a virus. Can. J. Comp. Med. 18:13-21. 1954. reovirus as a possible etiologic agent of osteoporosis "brittle bone disease";
4. Jackson, G. G., and R. L. Muldoon. Viruses causing respiratory infection "femoral head necrosis" in broiler chickens. Avian Dis. 25:847-856. 1981.
in man. IV. Reoviruses and adenoviruses. J. Infect. Dis. 128:812-833. 1973. 24. Walker, E. R., Μ H. Friedman, andN. O. Olson. Electron microscopic
5. Johnson, D. C., and L. van der Heide. Incidence of tenosynovitis in study of an avian reovirus that causes arthritis. J. Ultrastruct. Res. 41:67-79.
Maine broilers. Avian Dis. 15:829-834. 1971. 1972.
6. Kawamura, H., F. Shimizu, M Maeda, and H. Tsubahara. Avian 25. Witter, R. L. Reticuloendotheliosis. In: Diseases of poultry, 10th ed. B.
reovirus: its properties and serological classification. Natl. Inst. Anim. W. Calnek, H. J. Bames, C. W. Beard, L. R. McDougald, and Y. M Saif,
Health Q. (Yatabe) 5:115-124. 1965. eds. Iowa State University Press, Ames, Iowa. pp. 467-484. 1997.
7. Kerr, K. M, and N. O. Olson. Pathology of chickens experimentally 26. Wooley, R. E., T. A. Dees, A. L. Cromack, and J. B. Gratzek. Infectious
inoculated or contact-infected with an arthritis-producing virus. Avian Dis. enteritis of turkeys: characterization of two reoviruses isolated by sucrose
13:729-745. 1969. density gradient centrifugation from turkeys with enteritis. Am. J. Vet. Res.
8. Kisary, J., B. Nagy, and Z. Bitay. Presence of parvovirus in the intestine 33:165-170. 1972.
of chickens showing stunting syndrome. Avian Pathol. 13:339-343. 1984. 27. Wyeth, P. J., N. T. Chettle, and J. Labrano. Avian calicivirus. Vet. Rec.
9. Olson, N. O., and D. P. Solomon. A natural outbreak of synovitis caused 109:477. 1981.
by the viral arthritis agent. Avian Dis. 12:311-316. 1968.

183
40
ARBOVIRUS INFECTION
Eileen N. Ostlund and Janies E. Pearson

SUMMARY. Eastern equine encephalitis (EEE), western equine encephalitis (WEE), Highlands J (HJ), West Nile (WN), and turkey
meningoencephalitis (TME) viruses are arboviruses that cause clinical disease in avian species. Eastern equine encephalitis, WEE, and HJ are
members of the genus Alphavirus in the virus family Togaviridae\ TME and WN belong to the genus Flavivirus in the family Flaviviridae.
Infections with EEE and HJ viruses in avian species have been reported primarily in the eastern and southern United States. Infections with
WEE virus have been reported primarily in the western United States. West Nile virus was first recognized in the United States in New York
in 1999. Its current range in the Western Hemisphere extends across the continental United States as well as into Canada, Mexico and the
Caribbean. Turkey meningoencephalitis or Israel meningoencephalitis has been reported only in Israel and South Africa. Clinical
manifestations of these arboviruses differ among poultry and other affected avian species but evidence of nervous system involvement
including incoordination and paralysis are common. EEE and WEE viruses have been reported to cause disease in poultry, game birds and
ratites. The primary disease that has been associated with HJ virus is a drop in egg production in turkeys. Historically, clinical manifestations
of WN infection in avian species were not described. Since the late 1990’s, WN morbidity and mortality in domestic geese and wild birds,
especially corvids, has been documented. TME virus has only been reported to cause disease in turkeys under field conditions.
Agent Identification. Eastern equine encephalitis, WEE, HJ, TME and WN viruses can be isolated from field specimens by inoculating cell
cultures, newborn mice, or embryonating chicken eggs. Identification of EEE, WEE, and HJ isolates can be achieved by complement­
fixation, immunofluorescence, or plaque-reduction-neutralization (PRN) tests. WN isolates are identified by immunofluorescence. TME virus
is identified by virus neutralization in cell culture. Polymerase chain reaction (PCR) can be used to detect EEE, WEE, HJ and WN in tissues.
Antigen capture enzyme linked immunosorbent assay (ELISA) methods have been described for EEE and WN.
Serologic Detection in the Host. Eastern equine encephalitis, WEE, WN, and HJ virus-specific antibodies can be identified in host species
by PRN or hemagglutination-inhibition (HI) tests. Several ELISAs have been described for detection of WNV, EEE or WEE antibody in
avian serum. TME viral antibody can be identified by the HI test.

INTRODUCTION viral disease. Clinical manifestations of natural WN virus infection


in domestic poultry have been recognized only in geese (1,18).
The arboviruses comprise a group of taxonomically diverse viruses TME has only been reported in Israel and South Africa. Turkeys are
clustered together by their ability to replicate in and to be the only avian species to be clinically affected by TME (11,17).
transmitted by arthropod vectors. Over 530 arboviruses have been Several arboviruses including EEE, WEE, HJ, and WN are
identified. For many arboviruses, birds serve as maintenance hosts; zoonotic with potential to cause disease in humans. Severe
more than 100 arboviruses have been isolated from avian species infections and death caused by EEE and WEE viruses have been
and birds are frequently used as sentinels for the detection of reported in laboratory workers (24). Laboratory acquired infections
arbovirus activity. The bidirectional virus transmission between with WN virus have also occurred (3). Infectious arboviruses may
vectors and maintenance hosts seldom causes ill effects in either. be present in blood, serum and tissues of birds. Therefore any
Only five arboviruses (eastern equine encephalitis [EEE], western laboratory work with avian specimens and arboviruses should be
equine encephalitis [WEE], Highlands J [HJ], West Nile [WN], and done at the appropriate biosafety level. Biosafety Level 2 practices
turkey meningoencephalitis [TME]) have been reported to cause are recommended for processing diagnostic specimens. All bird
disease in avian species. Three of the five: EEE, WEE, and HJ are necropsies should be done in a Class 2 biological safety cabinet. For
members of the genus Alphavirus in the virus family Togaviridae. manipulations of cultures, EEE, WEE and HJ are assigned to
West Nile virus and TME belong to the genus Flavivirus in the Biosafety Level 2. EEE and WEE vaccines are recommended for all
family Flaviviridae. Mosquitoes serve as the primary arthropod personnel who are working with these agents. WN and TME viruses
vector for EEE, WEE, WN and HJ; mosquitoes and/or culicoides are assigned to Biosafety Level 3 (24). Unless suitable containment
are suspected vectors for TME. For each virus, the abundance of facilities and laboratory safeguards are available and used, no
insect species competent for virus transmission is a critical factor in attempts should be made to isolate any of these arboviruses. An
defining geographic areas with virus activity. Vector-borne alternative is to send all tissues from suspicious cases or possible
transmission is seasonal in temperate climates, peaking in late isolates to a reference laboratory. Due to public health concerns,
summer and early autumn. Direct bird to bird transmission through confirmed cases of EEE, WEE, HJ, and WN viral infections should
feather picking and cannibalism can occur. Direct transmission of be reported to public and animal health officials. TME is an exotic
EEE, WEE, TME, and WN viruses has been reported. disease in the United States and suspect or cases must be reported to
Eastern equine encephalitis and WEE viruses have been isolated animal health officials.
from mosquitoes and vertebrates throughout much of the Western
Hemisphere. However, all reports of disease in poultry, game birds, CLINICAL DISEASE
or domestic avian species have been from the United States with
most disease caused by EEE virus. Natural infection with EEE virus Clinical disease caused by EEE viral infection has been reported
has been implicated as a cause of clinical disease in emus, primarily in young chukars, quail, emus, and pheasants in the
pheasants, pigeons, turkeys, chukar partridges, quail and ducks eastern and southern United States. Isolated natural cases also have
(5,9,10,20,26). Clinical avian infections definitively attributable to been reported in young Pekin ducks, turkeys, quail, and whooping
WEE are rare but have been reported in turkeys (4). Older reports of cranes. Newly hatched chickens are very sensitive to experimental
WEE-caused avian disease in the eastern United States were likely infection. Clinical signs of EEE infection typically reflect
due to the antigenically related HJ virus. HJ virus has been reported perturbation of the nervous system. Visceral involvement is less
to cause disease in chukar partridges and turkeys in the eastern common. Signs may include depression, drowsiness,
United States (6,8,10). West Nile virus has a broad distribution incoordination, torticollis, paralysis, tremors and death. Mortality is
including portions of Asia, Africa, Europe and North America. Over quite variable but may approach 100%. A significant drop in egg
150 bird species are susceptible to infection with WN virus (25). production, without mortality, can occur in EEE-infected turkey
Infections in wild birds range from inapparent to fatal with corvid hens (28).
species such as crows and blue jays particularly susceptible to WN
184
Chapter 40 Arbovirus Infection

Clinical disease caused by WEE viral infection of avian species is highly susceptible to arbovirus infection. However, the potential of
rare and historical reports of WEE prior to the ability to distinguish aerosol transmission of virus shed by infected chicks precludes then-
HJ from WEE are tenuous. Historically WEE has been implicated use in routine virus isolation attempts. The laboratory’s choice of
as a cause of neurologic disease and mortality of turkeys. More isolation system depends on several factors including the
recently, WEE was reported in turkeys in California. The disease arboviruses of interest, laboratory facilities, availability of cell
was characterized by a sudden drop in egg quantity and quality (4). cultures, newborn mice and/or embryonating eggs, and personnel
EEE and WEE viral infections have been reported in ratites, expertise. In addition, the vaccination status of laboratory personnel
primarily emus (2,21,26). Emus of all ages have developed illness and potential exposure to zoonotic pathogens during inoculation and
following infections and clinical signs may include hemorrhagic harvesting must be considered.
enteritis. Many arboviruses were first isolated in suckling mice due to then-
Avian viruses in the WEE serogroup isolated in the eastern United broad sensitivity and worldwide availability. For isolation, a 10%
States have been determined to be HJ and some prior reports of suspension of tissue is prepared in phosphate-buffered saline (pH
WEE are now presumed to be HJ based on geographic location of 7.8) containing 0.75% bovine serum albumin (fraction V), 500
the affected birds. Chukar partridges infected with HJ virus can units/ml of penicillin, and 300 units/ml of streptomycin. The
exhibit somnolence, ruffled feathers and recumbency (20). suspension is clarified by centrifugation at 1400 x g for 20 min. One
Mortality of 35% has been reported (6). During an outbreak of HJ to five day old mice (one or two litters) are inoculated intracranially
in turkeys, decreased egg production was the predominant sign (28). with 0.02 ml inoculum using a 26-gauge, 3/8-inch (9.3 mm) needle
HJ was serologically linked to mortality in young turkeys (8). attached to a 1 ml tuberculin syringe. The inoculation site is just
Epomitics of TME generally occur in turkeys older than 8 wk of lateral to the midline into the midportion of one lateral hemisphere.
age and are characterized by drowsiness, incoordination, torticollis, Mice are observed for 10 days. Dead mice are collected daily and
tremors, progressive paresis and paralysis (11,17). In some cases, frozen at -70 C. Mouse brains are harvested for virus identification
the disease may be very acute, with only prostration and death being by aspiration using a 20-gauge, 1-inch (2.5 cm) needle attached to a
observed. Mortality is usually 15-30% but may be up to 80%. In 1 ml or 3 ml syringe. A second passage is made only if virus cannot
laying flocks, egg production declines precipitously but there is no be identified from mice that die following inoculation.
effect on egg quality, fertility or hatchability. Arboviruses can also be isolated in several cell culture systems
Geese naturally infected with West Nile virus display neurologic including cells of vertebrate and insect origin. Tissue suspensions
sings of incoordination, paralysis and recumbency. Juvenile birds for cell culture inoculation are prepared in the same diluent used for
experience a higher mortality than adults. Mortality rates of 25-40% mouse inoculation or cell culture media with antibiotics. The most
have been reported in goslings (1,18). Clinical West Nile infections commonly used cells are primary chicken and duck embryo
have not been identified in other domestic poultry breeds under fibroblasts and continuous lines of Vero, baby hamster kidney-231
natural conditions. Natural and experimental infections of avian (BHK-21), and rabbit kidney-13 (RK-13). Using multiple cell
species with West Nile virus have resulted in significant mortality culture systems is advantageous. Isolation is usually attempted in
in some species. Examples are American crows, fish crows, blue 25-cm2 cell-culture flasks. Confluent cell monolayers are inoculated
jays, ring-billed gulls, Muscovy ducks, and several members of the with 1 ml of tissue suspension. After a 1 hr adsorption period, cells
owl family, Strigidae (7,15). are washed and maintenance medium is added. Cultures are
No pathognomonic gross or histopathologic lesions are associated incubated for 7 days and 1 blind passage is made. EEE, WEE, HJ,
with infections by these arboviruses. A complete description of the WNV, and TME viruses produce a cytopathic effect (CPE) in
pathogenesis, epizootiology, and clinical diseases associated with vertebrate cell culture. Cultures that show CPE are frozen. Infected
arboviruses is reported by Guy (10). arthropod cells may not exhibit CPE. The fluid from the thawed
cultures is used for virus identification.
SAMPLE COLLECTION The chicken embryo is also appropriate for primaiy arbovirus
isolation attempts. Arbovirus infection should always be suspected
The method of choice for diagnosis and identification of arbovirus when an embryolethal virus is isolated from birds with neurotropic
infection is identification of the causal agent, either by isolation of disease. Tissue suspensions can be inoculated via the yolk sac route
the virus or by PCR. Although obtaining a virus isolate permits the into 6 to 8-day-old embryonating chicken eggs. Arboviruses are
most extensive characterization, molecular methods have the typically embryolethal but the infected embryos usually lack
advantage of improved sensitivity for some agents. Virus isolation diagnostic lesions. However, embryos infected with TME virus are
should only be attempted in laboratories with sufficient often a cherry red color before death (11). Embryos should be
biocontainment and biosecurity. Necropsy of birds suspected of incubated for 7 days but deaths usually occur 2-4 days after
being infected with an arbovirus should be performed in biological inoculation. Generally, only one passage is necessary, unless there
safety cabinets. Whole birds should be sent to the laboratory in are dead embryos from which virus cannot be identified.
sealed bags at 4 C. Because viremia may be of short duration, birds Following primary virus isolation in any system, arboviruses are
selected for diagnostic purposes should be those that are just frequently propagated by passage in one or more of the vertebrate
starting to exhibit clinical signs. The virus titer in these birds can be cell cultures listed above.
high, which enhances the possibility of virus isolation. Tissues to
collect include brain, Ever and spleen. Serum should be collected AGENT IDENTIFICATION
from birds that have a subacute infection because antibodies can
often be detected in birds with clinical disease. If tissues cannot be Eastern equine encephalitis, WEE, and HJ viruses belong to the
processed immediately, they should be stored at -70 C. genus Alphavirus of the family Togaviridae. WNV and TME
viruses belongs to the genus Flavivirus in the family Flaviviridae.
PREFERRED CULTURE MEDIA AND SUBSTRATES
Chemical and Physical Characteristics
Arboviruses are typically isolated in the laboratory by inoculation The alphaviruses and flaviviruses have a spherical appearance
of samples into newborn mice, vertebrate cell cultures, insect cell when viewed by electron microscopy. Alphaviruses are 50-70 nm in
cultures, or embryonating eggs. Although primary isolation of some diameter while the flaviviruses are somewhat smaller, measuring
arboviruses can be obtained in multiple systems, no single system is 40-50 nm in diameter. Virions in both groups have a ribonucleic
maximally sensitive for all arboviruses. Newborn chicks are also acid (RNA) core surrounded by a lipid-containing envelope;
185
Eileen N. Ostlund and James E. Pearson

therefore, they are ether and chloroform-sensitive. The capsid serum. Enzyme-linked immunosorbent assays to detect
encloses a single-stranded, positive sense RNA genome. immunoglobulin M (IgM) or IgG antibodies to specific arboviruses
are used in some situations, such as sentinel chicken testing (29).
Virus Identification The species specificity of immunoglobulin “capture” ELISA
Eastern equine encephalitis or WEE viruses can be identified in methods limits their application in diverse avian species. Blocking
infected mouse brains, cell culture fluid, or amniotic-allantoic fluid or competitive ELISAs have greater utility in detecting virus­
by the complement-fixation (CF) test. A 10% suspension of mouse specific antibodies from wide range of species (14).
brain is prepared in veronal buffer; egg or cell culture fluid is used
undiluted or diluted 1:10 in veronal buffer. The fluid or suspension PRN test
is centrifuged at 9000 x g for 30 min, and the supernatant is tested The PRN test is performed in duck embryo fibroblast or Vero cell
against hyperimmune serum or mouse ascites fluid prepared against cultures. The sera can be tested at the 1:10 and 1:100 final dilutions.
the suspected arboviruses using a standard CF procedure (23). The Endpoints can be established on the PRN or HI test. Serum used in
CF test requires overnight incubation of serum-antigen mixture with the PRN assay is tested against 100 plaque-forming units of virus.
seven units of complement. EEE, WEE, HJ, WN, and TME viruses The virus-serum mixture is incubated at 37 C for 1 hr and 15 min
can be identified in cell culture by indirect immunofluorescent before inoculation onto confluent cell monlayers in 25-cm2 flasks.
antibody staining. TME virus can also be identified by virus The inoculum is adsorbed for 1 hr, followed by the addition of 6 ml
neutralization or hemagglutination inhibition (HI) tests. The brains of overlay medium. The overlay medium consists of two solutions
from mice infected with TME virus will agglutinate goose red blood that are prepared separately. Solution I contains 2x Earle’s basic salt
cells (11). A less commonly used method for EEE and WEE virus solution with 2x antimicrobials, 4% fetal bovine serum, 6% of a
identification is the plaque-reduction neutralization (PRN) test as 7.5% solution of sodium bicarbonate and 3.3% of a 1:1500 dilution
outlined in a subsequent section. of neutral red (1:80000). Solution Π consists of 2% Noble agar that
is sterilized and maintained at 47 C. Equal volumes of solutions I
Molecular Methods and Π are mixed and the temperature is adjusted to 47 C just before
Molecular methods to detect the presence of arbovirus nucleic acid use. The test is incubated for 48-72 hr and endpoints are based on
in avian tissues are now commonly used by diagnostic laboratories 90% reduction in the number of plaques, as compared with the
where appropriate equipment and expertise are available. virus-control flasks which should have approximately 100 plaques.
Advantages of molecular detection methods include speed of these
diagnostic assays when compared to virus isolation procedures and Hemagglutination-Inhibition Test
lowered risk of laboratory personnel exposure to zoonotic agents A sucrose-acetone infected mouse brain extract is commonly used
since viruses are not propagated. Disadvantages include lack of as virus (positive) antigen and an uninfected mouse brain extract is
widespread experience and validation of these assays in diagnostic prepared in parallel as a control (negative) antigen. The virus
settings. For the single stranded, positive sense RNA viruses, antigen is inactivated by treatment with β-propiolactone at a final
reverse transcriptase polymerase chain reaction (RT-PCR) methods concentration of 0.1-0.3%. In the absence of an international
are used most frequently (16,19,27,30). Initial PCR tests developed reference serum, the antigen should be titrated against a locally
for arboviruses generally were designed to detect RNA of a single prepared positive control serum. The antigen is diluted so that four
viral pathogen (e.g. EEE). Whole RNA is extracted from diagnostic to eight hemagglutinating units are used in the HI test. The
specimens followed by RT and PCR steps. In some assays, a nested hemagglutination titer and optimum pH for each antigen is
PCR approach is used to enhance sensitivity of detection of small determined with goose red blood cells (RBCs) diluted in pH
quantities of viral RNA. Reaction products or their restriction solution ranging from pH 5.8 to pH 7.2 at 0.2 intervals. Sera are
fragments are analyzed on 2.0-2.6% agarose gels that have been diluted 1/10 in borate saline, pH 9.0, and then heat inactivated at 56
stained with 1 pg/ml of ethidium bromide. An alternate C for 30 min. Kaolin treatment is used to remove nonspecific serum
identification procedure is by hybridization with an oligonucleotide inhibitors. Sera should be adsorbed with a 0.05 ml volume of
probe. Recent advances in PCR procedures include a plethora of washed packed goose RBCs for 20 min at 4 C to remove natural
variations on the “standard” RT-PCR formats. These include real serum agglutinins. Serum is added to a 96-well, round-bottom
time methods that avert the need for PCR product testing and microtiter plate in two-fold dilutions in borate saline, ph 9.0
multiplex assays to detect and differentiate more than one pathogen containing 0.4% bovine serum albumin. Equal volumes of antigen
(12,13,16). An antigen capture assay developed for detection of WN are added to the diluted serum and the plates are incubated at 4 C
RNA in mosquito pools has been successfully applied to avian overnight. RBCs are derived from normal white male geese and
samples (22). washed three times in dextrose-gelatin-veronal buffer and a 7.0%
suspension is then diluted 1/24 in the appropriate pH solution
SEROLOGIC DETECTION IN THE HOST immediately before adding to the plates. Plates with RBCs are
covered and incubated for 30 min at 37C before recording results.
A presumptive diagnosis of arbovirus infection can be made using Positive and negative control sera are incorporated into each test. A
a single sample of serum from recovered birds or birds that have test is considered valid only if the control sera give the expected
clinical signs. Confirmation can be made by testing paired sera results. Titers of 1/10 and 1/20 are suspects; titers of 1/40 and above
collected 1-2 wk apart. The PRN test or a combination of PRN and are antibody positive.
HI tests are the procedures most commonly used to detect antibody
against EEE, WEE, WNV and HJ viruses. The use of the PRN test DIFFERENTIATION FROM CLOSELY RELATED AGENTS
allows differentiation of antibodies produced by antigenically
closely related EEE and WEE viruses. Similarly, PRN tests allow Although clinical signs, species affected, geographic location and
differentiation of WN antibodies from antibodies to the related St. other factors contribute to the differential diagnosis among EEE,
Louis encephalitis virus. Differentiation between HJ and WEE viral WEE, HJ, WNV, and TME, laboratory tests are necessary to
antibodies is very difficult because of the close antigenic confirm the causal agent. In addition, these arboviral infections
relationship. A presumptive differentiation can be made based on produce clinical signs in susceptible species that closely resemble
the part of the country from which the birds originated. The HI test central nervous system disturbances caused by other neurotropic
alone can be used to identify antibody against TME virus (11,17). agents, such as Newcastle disease virus, avian encephalomyelitis
The CF test is rarely used to detect arbovirus antibody in avian virus and Clostridium botulinum. All the arboviruses can produce a
186
Chapter 40 Arbovirus Infection

decrease in egg production similar to that caused by turkey Diagnosticians 42nd Annual Meeting. Minneapolis-St Paul, MN. June , 2003.
rhinotracheitis virus. These agents can be distinguished by isolation p. 49.
and identification of the agent or by serologic tests. Vaccination of 14. Jozan, M, R. Evans, R. McLean, R. Hall, B. Tangredi, L. Reed, and J.
Scott. Detection of West Nile virus infection in birds in the United States by
poultry against arboviruses is not practiced routinely in North
blocking ELISA and immunohistochemistry. Vector Bome Zoonotic Dis.
America. No arbovirus vaccines for use in avian species are 3:99-110. 2003.
licensed in the United States, however, EEE, WEE and WNV 15. Komar, N., S. Langevin, S. Hinten, N. Nemeth, E. Edwards, D. Hettier,
equine vaccines have been used experimentally in birds. A live B. Davis, R. Bowen, and M Bunning. Experimental infection of North
attenuated vaccine for TME virus exists and can be differentiated American birds with the New York 1999 strain of West Nile virus. Emerg.
from virulent strains by the intramuscular pathogenicity index test Infect. Dis. 9:311-322. 2003.
performed in 1-day-old-poults (11). If applicable, vaccination 16. Lanciotti, R. S., A. J. Kerst, R. S. Nasci, M S. Godsey, C. J. Mitchell,
history must be taken into account when interpreting laboratory Η. M Savage, N. Komar, N. A. Panella, B. C. Allen, K. E. Volpe, B. S.
Davis, and J. T. Roehrig. Rapid detection of West Nile virus from human
results.
clinical specimens, field-collected mosquitoes, and avian samples by a
TaqMan reverse transcriptase-PCR assay. J. Clin. Microbiol. 38:4066-4071.
REFERENCES 2000.
17. Malkinson, M Turkey meiningo-encephalitis. In: Virus infections of
1. Austin, R. J., T. L. Whiting, R. A. Anderson, and M A. Drebot. An birds. J. G. McFerran and M S. McNulty, eds. Elsevier Science Publishers
outbreak of West Nile virus-associated disease in domestic geese (Anser Β. V., Amsterdam, The Netherlands, pp. 243-245. 1993.
anser domesticus) upon initial introduction to a geographic region, with 18. Malkinson, M, C. Banet, Y. Khinich, I. Samina, S. Pokamunski, and Y.
evidence of bird to bird transmission. Can. Vet. J. 45:117-123. 2004. Weisman. Use of live and inactivated vaccines in the control of West Nile
2. Ayers, J. R., T. L. Lester, and A. B. Angulo. An epizootic attributable to fever in domestic geese. Ann. N. Y. Acad. Sci. 951:255-261. 2001.
western equine encephalitis virus infection in emus in Texas. J. Am. Vet. 19. Office International des Epizooties. West Nile encephalitis. In: Manual
Med. Assoc. 205:600-601. 1994. of diagnostic tests and vaccines for terrestrial animals, 5th ed. Office
3. Centers for Disease Control and Prevention. Laboratory-acquired West International des Epizooties, Paris, pp. 1064-1071. 2004.
Nile virus infections—United States, 2002. Morb. Mortal. Wkly. Rep. 20. Ranck, Jr, F. M, J. H. Gainer, J. E. Hanley, and S. L. Nelson. Natural
51:1133-1135. 2002. outbreak of eastern and western encephalitis in pen-raised chukars in
4. Cooper, G. L. and H. A. Medina. Egg production drops in breeder Florida. Avian Dis. 9:8-20. 1965.
turkeys associated with western equine encephalitis virus infection. Avian 21. Randolph, K. D., S. L. Vanhooser, and M Hoffman. Western equine
Dis. 43:136-141. 1999. encephalitis virus in emus in Oklahoma. J. Vet. Diagn. Invest. 6:492-493.
5. Eleazer, T. Η., H. G. Blalock, J. H. Warner, Jr., and J. E. Pearson. Eastern 1994.
equine encephalomyelitis outbreak in coturnix quail. Avian Dis. 22:522-525. 22. Stone, W. B., J. C. Okoniewski, J. E Therrien, L. D. Kramer, E. B.
1978 Kauffman, and M Eidson. VecTest as diagnostic and surveillance tool for
6. Eleazer, T. H. and J. E. Hill. Highlands J virus-associated mortality in West Nile virus in dead birds. Emerg. Infect. Dis. 10:2175-2181. 2004.
chukar partridges. J. Vet. Diagn. Invest. 6:98-99. 1994. 23. United States Department of Health, Education, and Welfare. A guide to
7. Fitzgerald, S. D., J. S. Patterson, M Kiupel, H. A Simmons, S. D. the performance of the standardized complement fixation method and
Grimes, C. F. Sarver, R. M Fulton, B. A. Steficek, T. M Cooley, J. P. adaptation to micro test. Centers for Disease Control, Atlanta, Ga. 1974.
Massey, and. J. G. Sikarskie. Clinical and pathologic features of West Nile 24. United States Department of Health and Human Services. Biosafety in
virus infection in native North American owls (Family strigidae). Avian Dis. microbiological and biomedical laboratories, 4th ed. U.S. Government
47:602-610. 2003. Printing Office, Washington, D.C., 1999.
8. Flicken, M D., D. P. Wages, J. S. Guy, J. A. Quinn, and W. H. Emory. 25. van der Meulen, K. Μ, Μ B. Pensaert, and H. J. Nauwynck. West Nile
High mortality of domestic turkeys associated with Highlands J virus and virus in the vertebrate world. Arch. Virol. 150:637-657. 2005.
eastern equine encephalitis virus infections. Avian Dis. 37:585-590. 1993. 26. Veazey, R. S., C. C. Vice, D.-Y. Cho, T. N. Tully, Jr., and S. M Shane.
9. Guy, J. S., H. J. Bames, M D. Flicken, L. G. Smith, W. H. Emory, and Pathology of eastern equine encephalitis in emus (Dromaius
D. P. Wages. Decreased egg production in turkeys experimentally infected novaehollandiae). Vet. Pathol. 31:109-111. 1994.
with eastern equine encephalitis virus or Highlands J virus. Avian Dis. 27. Vodkin, Μ H., G. L. McLaughlin, J. F. Day, R. E. Shope, and R. J.
38:563-571. 1994. Novak. A rapid diagnostic assay for eastern equine encephalomyelitis viral
10. Guy, J. S. and M Malkinson. Arbovirus infections. In: Diseases of RNA. Am. J. Trop. Med. Hyg. 49(6):772-776. 1993.
Poultry, 11th ed. Y. M. Saif, ed. Iowa State Press, Ames, Iowa. pp. 388-399. 28. Wages, D. P., M D. Flicken, J. S. Guy, T. S. Cummings, and S. R.
2003. Jennings. Egg-production drop in turkeys associated with alphaviruses:
11. Ianconescu, M Turkey meningoencephalitis. In: A laboratory manual eastern equine encephalitis virus and Highlands J virus. Avian Dis. 37:1163-
for the isolation and identification of avian pathogens, 3rd ed., H. G. 1166. 1993.
Purchase, L. H. Arp, C. H. Dommermuth, and J. E. Pearson, eds. American 29. Weingartl, Η. Μ, M A. Drebot, Z. Hubalek, J. Halouzka, M
Association of Avian Pathologists, Kennett Square, Pa. pp. 163-164. 1989. Andonova, A. Dibemardo, C. Cottam-Birt, J. Larence, and P. Marszal.
12. Johnson, D. J., E. N. Ostlund, and B. J. Schmitt. Nested multiplex RT- Comparison of assays for the detection of West Nile virus antibodies in
PCR for detection and differentiation of West Nile virus and eastern equine chicken serum. Can. J. Vet. Res. 67:128-132. 2003.
encephalomyelitis virus in brain tissues. J. Vet. Diagn. Invest. 15:488-493. 30. Whitehouse, C. A., A. Guibeau, D. McGuire, T. Takeda, and T. N.
2003. Mather. A reverse transcriptase-polymerase chain reaction assay for
13. Johnson, D. J., E. N. Ostlund, and B. J. Schmitt. Nested multiplex RT- detecting Highlands J virus. Avian Dis. 45:605-611. 2001.
PCR for detection and differentiation of western equine encephalomyelitis
virus, eastern equine encephalomyelitis virus, and West Nile virus,
(abstract). North Central Conference of Veterinary Laboratory

187
41
INFECTIOUS BURSAL DISEASE
John K. Rosenberger, Y. M. Saif, and Daral J. Jackwood

SUMMARY. Infectious bursal disease is an acute lymphocidal disease of young chickens that is caused by a double-stranded RNA virus
that is unusually resistant to inactivation by both heat and many common disinfectants. The virus can be found worldwide in all major
poultry-producing areas. Clinical and subclinical forms of the disease occur in young chickens and are followed by marked
immunosuppression. Very virulent infectious bursal disease viruses cause high morbidity and mortality.
Agent Identification. Reverse transcriptase polymerase chain reaction is the method of choice for virus identification. Virus isolation could
be done but it is not practical unless further studies are planned.
Serologic Detection in the Host. Serologic results from enzyme-linked immunosorbent assay can provide a presumptive diagnosis if
accompanied by clinical disease and gross lesions. Agar-gel precipitin or virus-neutralization tests could be used for serologic testing.

INTRODUCTION Because the disease is acute and the virus infection is transient,
bursae should be harvested from a minimum of five clinically
Infectious bursal disease (IBD) is an acute, highly infectious affected birds and stored frozen, at -20 C or colder, pending
lymphocidal disease of young chickens caused by a double-stranded isolation attempts. A 20% (w/v) suspension of bursal homogenate is
RNA virus. The disease is frequently referred to as Gumboro, prepared in tryptose phosphate broth with antibiotics (10,000 IU/ml
named for the town in Delaware, USA where the disease was first of penicillin and 10,000 mg/ml of dihydrostreptomycin). The
recognized (10). The condition has been recognized in virtually all suspension is centrifuged at 1500 x g for 20 min, and the supemate
major poultry-producing areas worldwide and is considered is collected and stored frozen at -20 C or colder.
economically significant because of its ability to induce a profound
immunosuppression in chickens with increased susceptibility to Molecular Identification
bacterial and viral infections and diminished vaccine responses. A Samples from the bursa are preferred because they contain the
variety of infectious bursal disease viruses (IBDV) varying from the highest concentrations of the virus. Bursa should be harvested from
very mild to the very virulent have been identified. Although IBDV a minimum of five birds/house, pooled and submitted to a
has been isolated from other avian species, most notably turkeys molecular testing laboratory on ice or frozen. In cases where there
and ratites, the disease is currently recognized in the chicken only. are no domestic testing laboratories, bursa samples can be treated to
inactivate the virus but preserve the viral genome prior to
CLINICAL DISEASE international shipment (2). Bursa samples collected for international
shipment should be placed in a solution of phenol, chloroform and
The clinical form of the disease generally occurs in young birds isoamyl alcohol (25:24:1, pH 6.6) for a minimum of 7 days at room
and is often concurrent with waning IBDV maternal antibodies. temperature. The phenol:chloroform:isoamyl alcohol should be
Affected birds may be depressed and anorexic, have ruffled removed before shipment with the appropriate import and
feathers, and excrete a watery, green-tinged, urate-containing fecal declaration documentation.
material. Mortality is variable but can be as high as 60% dependent
on the strain of the virus and concomitant infections and mortality is PREFERRED CULTURE MEDIA AND SUBSTRATES
usually higher in leghorn chickens compared to meat-type birds.
Two to 6 days postinfection, the cloacal bursa (bursa of Fabricius) Virulent and vaccine IBDV can be propagated in 9-to-l 1-day-old
is swollen and occasionally hemorrhagic, and is frequently covered embryonated eggs derived from hens free of IBDV maternal
with a yellowish gelatinous transudate. This is followed by bursal antibodies. The chorioallantoic membrane (CAM) route of
atrophy, which occurs 7-10 days after infection. inoculation is the most sensitive, although embryos inoculated via
Microscopic lesions include lymphoid necrosis in the bursa, the yolk sac are also infected by most isolates. Embryo lesions and
spleen, thymus, Harderian gland, and cecal tonsils. The bursa is mortality rates may vary, depending on the type of IBDV assayed.
most severely affected and is characterized by a medullary Classical IBDV isolates, when inoculated by the CAM route, will
lymphoid cell necrosis followed by replacement with heterophils kill embryos 3-5 days postinoculation. Hie dead embryos are
and macrophages. Some IBDV variants produce only atrophy of the congested, with petechial and ecchymotic hemorrhages evident
bursa (15). along the feather tracts, toe joints, and cerebral area. Livers may be
In the subclinical form of the disease lymphoid tissues are affected necrotic, but usually have a pale appearance, whereas spleens are
but the only visible indication of infection may be a severely most frequently pink or lacking in color, small to normal in size,
atrophied bursa. However, subclinical IBD is considered important with occasional small necrotic foci. CAMs are usually unaffected,
because it results in immunosuppression similar to that induced by although infrequent, isolated surface hemorrhages have been
more pathogenic strains. observed.
Variant strains of IBDV can be isolated and grown in chicken
SAMPLE COLLECTION embryos with relative ease when introduced by the CAM route but
generally will not kill embryos. Live embryos must be examined 6-
Virus Isolation. Infectious bursal disease virus can be readily 7 days postinoculation for typical lesions, which externally consist
isolated from most lymphoid tissues during the early stages of the of cerebral and abdominal edema, stunting, and an off-white or
disease (2-6 days postinfection). However, the bursa is the primary creamy color. Embryos are rarely, if ever, congested or
target organ and generally contains greater concentrations of virus hemorrhagic. Livers are frequently bile-stained and necrotic,
for longer periods than other tissues and accordingly is considered whereas spleens are generally enlarged two to three times but are
the tissue of choice for isolation attempts (10). If the bursa is normal in color. The highest concentrations of virus are found in the
chosen, it should be recognized that it may frequently be CAM and embryo tissues, including viscera, 6-7 days after
contaminated with adventitious agents, such as reovirus, inoculation with either the variant or classical form of the virus. Egg
adenovirus, or chicken anemia virus, which could complicate the fluids usually contain less virus than other parts of the embryo.
identification process.
188
Chapter 41 Infectious Bursal Disease

Although the initial isolation of IBDV is best accomplished by viruses. This technique has also been used to identify molecular
embryo inoculation, the virus may be adapted to cell cultures (5, 10, markers present on many wIBDV strains (8).
12). IBDV can be propagated in chicken embryo bursal cells, Although widely accepted as the method of choice, RFLP assays
kidney cells, and fibroblasts, producing a cytopathic effect. The have some limitations. As a result, real-time RT-PCR is being used
virus has also been isolated on a variety of established mammalian by some laboratories to diagnose and differentiate IBDV strains (4,
cell lines including lymphoid and nonlymphoid cell lines (5, 9). The 11, 13, 14). Nucleotide sequencing of viruses is also gaining
very virulent wIBDVs are also propagated in embryonated chicken popularity but this method is still relatively slow and expensive
eggs. They cause severe lesions in embryo and are highly lethal at compared to real-time RT-PCR.
low virus doses. These viruses are difficult to adapt to tissue
culture. SEROLOGIC DETECTION IN THE HOST

AGENT IDENTIFICATION The AGP test can be used to detect and quantitate antibodies in
convalescent birds. To quantitate the antibodies, usually twofold
Morphology and Physicochemical Properties serum dilutions are made to obtain an endpoint. The antigen is
Infectious bursal disease virus is a nonenveloped icosahedral virus prepared from bursal homogenates of experimentally infected birds
in the family Bimaviridae, genus Bimavirus, that is approximately collected 3-6 days postinfection. The test is performed as described
60 nm in diameter. The genome is a double-stranded RNA that earlier.
consists of two segments with molecular weights of approximately The VN test is also used for antibody quantitation. The test is
2.2 x 106 and 2.5 x 106. Five viral proteins (VP1-VP5) have been routinely performed in microtiter systems using cell-culture-adapted
identified. IBDV is heat-stable, remaining infectious after treatment virus and chicken embryo fibroblasts. Twofold dilutions of serum
at 56 C for at least 5 hr. The virus is unaffected by ether, are prepared in medium 199 and added (0.025 ml/well) to
chloroform, or pH 2 but is inactivated at pH 12. Virus infectivity microplates containing 24-hr-old confluent monolayers of chicken
was markedly reduced following treatment with 0.5% formalin for 6 embryo fibroblasts. The cells are inoculated with 100 plaque­
hr, whereas phenolic derivatives and quaternary ammonium forming units of virus per well and incubated for 5 days at 37 C.
disinfectants were relatively ineffective in reducing virus titer (1). Following incubation, the growth medium (medium 199 and 5%
The stability of the virus is the reason it remains on contaminated fetal calf serum) is removed and cells are rinsed with 10% buffered
farms for long periods in spite of rigorous cleaning and disinfection. formalin for 5 min. The formalin is decanted, and the cells are
stained for 3 min with 1% crystal violet. Neutralization titers are
Virus Identification expressed as the reciprocal of the highest dilution of serum that
The agar gel precipitin (AGP) test can be used to detect IBDV prevents cytopathic effect. The VN test is more sensitive than the
group-specific antigen. Antigen is prepared from bursal AGP test and accordingly should be utilized when antibody titers
homogenates from birds in the acute stage of the disease. The bursal are low or when quantitation of antibody is important.
homogenate is mixed 1:1 (w/v) with sterile saline, frozen and The ELISA is widely used in quantitating LBDV antibodies. Test
thawed three times, and centrifuged at low speed (300 x g), and the kits can be purchased from commercial sources and have the
supernatant is retained as antigen and tested against known IBDV advantage of requiring small quantities of serum with improved
antiserum. The virus-neutralization (VN) test could be used to consistency of results. ELISA results do not always correlate with
identify the virus in embryos or in tissue culture after adaptation to VN test findings.
these host systems.
An antigen-capture enzyme-linked immunosorbent assay (AC- DIFFERENTIATION FROM CLOSELY RELATED AGENTS
ELISA) has been described for detecting and characterizing IBDV
isolates (16, 17). Polyclonal antibodies can also be utilized in the Clinical IBD is readily diagnosed because of the pathognomonic
AC-ELISA and may be more effective for general screening of gross and microscopic bursal lesions observed during the acute
tissue samples for IBDV (10). stage of the disease. However, bursal atrophy, which occurs
Direct and indirect immunofluorescence assays have been shown following clinical or subclinical IBD, may also be present with
to be highly reliable for detection of viral antigens in infected other conditions, such as Marek’s disease, mycotoxicoses, and
tissues. Likewise, immunocytochemistry is used for the same infection with chicken anemia agent or selected reoviruses. The
purpose. histologic response with these conditions is generally different from
Strains of IBDV can be differentiated on the basis of pathotype that found with IBD.
and antigenic configuration. There are at least two distinct serotypes Hemorrhagic lesions seen occasionally with IBD may also be
(5, 6, 10) that can be identified by cross-neutralization and cross observed with chicken anemia virus infections and chemical or drug
challenge tests. Serotype 1 viruses are classified into classic and intoxications. Severe lymphoid cell destruction, massive
variant strains that differ antigenically. The variant isolates may subcutaneous hemorrhages, and altered hematopoiesis are common
damage the bursa with no associated inflammation or edema (15). in birds coinfected with chicken anemia virus and IBDV at a young
Serotype 1 viruses vary in pathogenicity from being nonpathogenic age (<2 wk). Nephrosis and nephritis found occasionally in birds
to very virulent. The wIBDV were identified in the 1980s and that die from IBD are usually the result of water deprivation.
spread to several parts of the world except North America, Nephrotoxic strains of infectious bronchitis virus may induce
Australia, and New Zealand. These viruses are capable of inducing similar lesions but can be differentiated because there are no
high mortality and causing extensive damage to lymphoid tissue. changes in the bursa and the condition is normally associated with
acute respiratory disease.
Molecular Identification Because IBDV is capable of inducing immunosuppression, it may
Reverse transcriptase (RT) followed by the polymerase chain serve as a predisposing factor to other conditions, such as
reaction (PCR) is used to detect IBDV infection in chickens (3, 7, gangrenous dermatitis, hemorrhagic aplastic anemia syndrome,
18). When RT-PCR is followed by restriction enzyme fragment inclusion body hepatitis, and respiratory disease. Involvement of
length polymorphism (RFLP) assays the data can be used to place LBDV in disease complexes of these types is usually diagnosed
IBDV strains into molecular groups (3, 8, 19). The RT-PCR-RFLP retrospectively by demonstrating persistent bursal lesions or
procedures used to generate molecular groups of IBDV are serologic evidence of previous IBDV infection.
designed to assess the nucleotide similarity or diversity among
189
John K Rosenberger, Y. M Saif, and Daral J. Jackwood

REFERENCES

1. Benton, W. J., M. S. Cover, J. K. Rosenberger, and R. S. Lake. 11. Moody A., S. Sellers, and N. Bumstead. Measuring infectious bursal
Physicochemical properties of the infectious bursal agent (IBA). Avian Dis. disease virus RNA in blood by multiplex real-time quantitative RT-PCR. J.
11:438-445. 1967. Virol. Methods. 85: 55-64. 2000.
2. Jackwood, D. J., G. Hanes, and S. Heins-Miller. Infectious bursal disease 12. Okoye, J. O. A Infectious bursal disease of chickens. Vet. Bull.
viral RNA can be amplified using RT/PCR from bursa tissue following 54:425-436. 1984.
inactivation of the virus with phenol:chloroform Avian Dis. 40:457-460. 13. Raue R., and A. Mazaheri. Eine real-time RT-PCR zum nachweis des
1996. virus der infektiosen bursitis auf der grundlage des genomsegments B. Arch
3. Jackwood D. J., and S. E. Sommer. Genetic heterogeneity in the VP2 Geflugelk. 67: 22-27. 2003.
gene of infectious bursal disease viruses detected in commercially reared 14. Raue R., and H. Muller. Detection and quantification of IBDV by real­
chickens. Avian Dis. 42: 321-339. 1998. time RT-PCR. Proceedings, Π. International Symposium on Infectious
4. Jackwood D. J., B. D. Spalding, and S. E. Sommer. Real-time reverse Bursal Disease and Chicken Infectious Anaemia, Rauischholzhausen,
transcriptase-polymerase chain reaction detection and analysis of nucleotide Germany, pp. 271-277. 2001.
sequences coding for a neutralizing epitope on infectious bursal disease 15. Rosenberger, J. K, S. S. Cloud, J. Gelb, E. Odor, and J. E. Dohms.
viruses. Avian Dis. 47: 170-176. 2003. Sentinel bird survey of Delmarva broiler flocks. In: Proceedings of the 20th
5. Jackwood, D. H., and Y. M Saif. Antigenic diversity of infectious bursal National Meeting on Poultry Health and Condemnations, Ocean City, Md.
disease viruses. Avian Dis. 31:766-770. 1987. pp. 94-101. 1985.
6. Kibenge, F. S. B., A. S. Dhillon, and R. G. Russell. Biochemistry and 16. Snyder, D. B., D. P. Lana, B. R. Cho, and W. W. Marquardt. Group and
immunology of infectious bursal disease virus. J. Gen. Virol. 69:1757-1775. strain-specific neutralization sites of infectious bursal disease virus defined
1988. with monoclonal antibodies. Avian Dis. 32:527-534. 1988.
7. Lee, L. H., S. L. Yu, and Η. K. Shien. Detection of infectious bursal 17. Snyder, D. B., D. P. Lana, P. D. Savage, F. S. Yancey, S. A. Mengel,
disease virus infection using the polymerase chain reaction. J. Virol. and W. W. Marquardt. Differentiation of infectious bursal disease viruses
Methods 40:243-254. 1992. directly from infected tissues with neutralizing monoclonal antibodies:
8. Lin Z., A. Kato, Y. Otaki, T. Nakamura, E. Sasmaz, and S. Ueda. evidence of a major antigenic shift in recent field isolates. Avian Dis.
Sequence comparison of a highly virulent infectious bursal disease virus 32:535-539. 1988.
prevalent in Japan. Avian Dis. 37: 315-323. 1993. 18. Wu, C. C., T. L. Lin, H. G. Zhang, V. S. Davis, and J. A Boyle.
9. Liu, H.-J., J. J. Giambrone, and T. Dormitorio. Detection of genetic Molecular detection of infectious bursal disease virus by polymerase chain
variations in serotype I isolates of infectious bursal disease virus using reaction. Avian Dis. 36:221-226. 1992.
polymerase chain reaction and restriction endonuclease analysis. J. Virol. 19. Zierenberg K., R. Raue and H. Muller. Rapid identification of very
Methods 48:281-291. 1994. virulent strains of infectious bursal disease virus by reverse transcriptase-
10. Lukert, P. D., and Y. M. Saif. Infectious bursal disease. In: Diseases of polymerase chain reaction combined with restriction enzyme analysis. Avian
poultry, 10th ed. B. W. Calnek, H. J. Bames, C. W. Beard, L. R. Pathol. 30: 55-62. 2001.
McDougald, and Y. M. Saif., eds. Iowa State University Press, Ames, Iowa,
pp. 721-738. 1997.

190
42
PARVOVIRUS OF WATERFOWL
Richard E. Gough

SUMMARY. Goose parvovirus or Derzsy’s disease is caused by an autonomous replicating parvovirus. The disease can result in 100%
mortality in goslings and Muscovy ducklings less than 10 days of age. A similar parvovirus is responsible for a serious disease of Muscovy
ducklings although goslings appear less susceptible to this virus. Significant differences between the genomes of goose and Muscovy duck
isolates have been reported.
Agent Identification. Diagnosis of goose parvovirus is made by isolating the virus in embryonating goose or Muscovy duck eggs, or in cell
cultures derived from these species. The identification of the isolated virus can be confirmed by electron microscopy or a virus-neutralization
test. Detection of viral antigen in host tissues or culture systems has also been used to confirm infection. Molecular methods such as PCR,
DNA probes and RFLP analysis have been applied to the detection and characterization of waterfowl parvoviruses.
Serologic Detection in the Host. Several serologic assays have been developed to confirm infection, including virus-neutralization, agar-gel
precipitin tests, ELISA and plaque reduction tests. In countries where vaccination is practiced, serological results should be interpreted with
caution. Ideally, both acute and convalescent serum samples should be tested in order to confirm infection.

INTRODUCTION Goslings and ducklings over 4 wk of age rarely show clinical signs.
In acute cases, lesions are commonly found in the heart, which has a
Goose parvovirus (GPV) infection, also known as Derzsy’s pale myocardium characteristically rounded at its apex. The liver,
disease, goose hepatitis, or gosling plague, is a highly contagious spleen, and pancreas may be swollen and congested. A variety of
disease affecting goslings and Muscovy ducklings. Apart from other gross lesions may also be present in cases with a more
these species, the disease has not been reported in other avian or prolonged clinical course. Typically, a serofibrinous perihepatitis
mammalian species. The first description of the disease was from and pericarditis is present with large volumes of straw-colored fluid
China in 1956, but goose parvovirus has now been reported from in the abdominal cavity. Pulmonary edema, liver dystrophy, and
most of the major goose and Muscovy duck farming countries in the catarrhal enteritis may also be present. Less frequently,
world (8). Outbreaks of the disease have frequently been attributed hemorrhages in the thigh and pectoral muscles may be seen.
to egg transmission of the virus from latently infected breeder geese Diphtheritic and ulcerative lesions may be observed in the mouth,
or Muscovy ducks, this results in infection passing to susceptible pharynx, and esophagus, depending on the presence of secondary
birds at the time of hatching or soon thereafter. Birds that survive invaders. Histologically, the main lesions seen are pronounced
infection may become life-long carriers of the virus. More recently degenerative changes in the myocardial cells and hepatocytes with
a parvovirus of Muscovy or Barbary ducks has been detected in vacuolation, fatty infiltration, and the presence of scattered Cowdry
various countries throughout the world, the so-called Muscovy duck type A intranuclear inclusions. Similar changes occur in the
parvovirus (MDPV). This virus was shown to be antigenically pancreas, kidney, spleen, bursa, and thymus. MDPV in young
distinct from GPV, and while causing up to 80% mortality in ducklings causes similar clinical signs and pathology to GPV and
Muscovy ducklings, was not particularly pathogenic for goslings these have been described in detail (4,13,27). It has been suggested
(6,12,14). Molecular studies using restriction enzyme fragment that GPV infection in susceptible breeding geese may result in
length polymorphism and sequencing was able to show differences infertility and poor hatchability (11).
between a selected strain of GPV and a MDPV (29). Both viruses
belong to the Parvoviridae family, although phylogenetic analysis SAMPLE COLLECTION
has suggested that, together with adeno-associated viruses, they
should be separated from other autonomous parvoviruses and Virus isolation and visualization by direct transmission electron
placed in the Dependovirus genus (20). microscopy (TEM) can be attempted on fecal samples collected
during the acute phase of the disease. At postmortem, a selection of
CLINICAL DISEASE tissues, including heart, liver, spleen, and kidneys should be
sampled. For virus isolation, the specimens should be shipped at 4
With GPV the clinical signs, morbidity, and mortality in C in leak-proof containers with ice or other suitable coolant. Where
susceptible goslings and ducklings vary according to the age of the delays in shipping of more than 2 days are expected, the samples
birds when infected. Mortality may reach 100% in goslings should be frozen prior to shipping and sent frozen. As soon after
infected in the hatchery, with the appearance of clinical signs and death as possible appropriate samples, particularly organs or tissues
death 5-10 days later. In birds 2 to 3-wk old, the incubation period showing specific lesions should be placed in 10% neutral buffered
and mortality may vary considerably (18). Initially, affected birds formalin solution for histologic examination. As GPV can be
exhibit anorexia, polydypsia, and weakness with a reluctance to vertically transmitted, embryos that die during incubation or shortly
move. A nasal and ocular discharge occurs in many birds with after hatching should be examined for the presence of virus.
associated head shaking. The uropygial glands and eyelids are Antibodies can be detected in blood samples from healthy ducks
often red and swollen, and a profuse white diarrhea is evident in and geese and their progeny. Samples of yolk from infertile eggs
many of the birds. Examination of the birds, at this stage may, can also be collected for antibody assay.
reveal a fibrinous pseudomembrane covering the tongue and oral
cavity. Goslings that survive the acute phase may develop a more PREFERRED CULTURE MEDIA
prolonged disease characterized by profound growth retardation,
loss of down around the back and neck, and marked reddening of For attempted virus isolation, 20% tissue suspensions are prepared
the exposed skin. Ascitic fluid may accumulate in the abdomen, in a solution of antibiotic phosphate-buffered saline (PBS) and
which causes the goslings to stand in a “penguin-like” posture. inoculated into either embryonating goose or Muscovy duck eggs or

191
Richard E. Gough

cell cultures derived from them. It is essential that the embryonated MDPV in vitro and in tissues (8,26). A digoxigenin-labelled DNA
eggs are derived from unvaccinated flocks known to be free of GPV probe technique has been described for the detection and
and MDPV antibodies. characterization of MDPV isolates (19).

Culture in Embryonating Eggs Antigen Detection


The virus can be isolated following inoculation in the allantoic Viral antigen can be detected by immunofluorescence and
cavity of 10 to 15-day-old embryonating goose or Muscovy duck immunoperoxidase techniques in the viscera, particularly the liver
eggs known to be free of goose parvovirus antibodies. Mortality, and spleen, of infected goslings and ducklings, and in infected cell
with hemorrhages in the embryonic Ever and other tissues, usually cultures and embryonated eggs (1,8,23). For primary antibody,
occurs 5-12 days after incubation at 37 C. After further passages, goose anti-goose parvovirus globulin is obtained by triple
consistent embryo mortality normally occurs between 4 and 6 days precipitation of hyperimmune goose serum using 33% ammonium
post inoculation. A minimum of two passages should be carried out sulfate. The concentration of protein is determined and the dialyzed
before the attempted virus isolation is considered negative as virus gammaglobulin fraction labeled with fluorescein isothiocyanate
may not be detected on primary inoculation. (FITC). When applied to impression smears of liver or spleen from
infected goslings and infected cell cultures the nuclei of the cells
Cell Cultures fluoresce brightly.
Primary cell cultures of 12 to 15-day-old goose or Muscovy duck Antigen-capture enzyme-linked immunosorbent assays (ELISAs)
embryo fibroblasts are suitable for isolating GPV. In common with have also been developed for the detection of goose parvovirus
mammalian parvoviruses, isolation is facilitated by inoculating cell antigen in cell cultures and tissues from goslings (16). However,
cultures before they form a confluent monolayer, preferably at the these techniques have not yet been validated for routine laboratory
time of seeding the cultures. Primary isolation may not always testing.
result in a detectable cytopathic effect (CPE) and several passages Agar-gel precipitin techniques have also been developed to detect
may be required before a CPE is observed. However, in the absence parvovirus antigen in the embryonic tissues and allantoic fluid of
of a CPE, examination of cell lysates by TEM often reveals the infected embryos (7). Following inoculation of 10-to- 15-day-old
presence of parvovirus virions (9). Following adaption the virus goose or Muscovy duck embryos with diagnostic samples, embryos
produces a well-defined CPE 3-5 days after inoculation, consisting that subsequently die can be examined for goose parvovirus antigen.
of rounded refractile cells. The cytopathic effect progresses to The dead embryos and amniotic-allantoic fluids (AAFs) are
complete destruction of the cell monolayer after 6-7 days. Cell harvested, the embryos’ head and limbs removed, and a 20% (w/v)
cultures derived from Muscovy duck embryos are required in order suspension prepared in the AAF. After homogenization, the
to isolate MDPV. As with GPV, virus replication may occur in the suspension is mixed with an equal volume qf trichlorotrifluorethane
absence of a detectable CPE and EM or molecular methods may be (Arcton 113, ICI, London, United Kingdom) for 30 min, followed
required to confirm the presence of the isolate (27). by centrifugation at 3000 x g for 15 min. The supernatant is
removed and further centrifuged at 46,000 x g for 2 hr at 4 C. The
AGENT IDENTIFICATION resulting pellet is resuspended in a minimal volume of deionized
water and treated with 0.1% N-lauroylsarcosine (Sigma Chemical
Morphology Co., St. Louis, Mo.). Agar plates are prepared using 1% Ionager
Direct electron microscopy examination of feces from infected No. 2 (Oxoid, distr. Unipath, Ogdensburg, N.Y.) in 8% sodium
birds or culture-derived material often reveals parvovirus particles. chloride solution, buffered to pH 7.8. The gel pattern consists of a
The intact virions are 20-22 nm in diameter, non-enveloped, and central well 5 mm in diameter surrounded by six outer wells of the
hexagonal in shape with an estimated 32 capsomeres (8). same diameter spaced 3 mm apart. Aliquots of positive goose
Aggregates of virions have also been detected following EM parvovirus precipitating antisera are placed in the wells either side
examination of ultrathin sections of heart and bursae from infected of the test antigen with a known positive control antigen in the
goslings (3). An immune EM technique utilizing specific GPV central well. Diffusion of antiserum and antigen takes place at 20-
monoclonal antibodies has been developed to detect virions in the 22 C and the plates are examined for precipitin lines after
organs of goslings and cells of infected goose and duck embryos approximately 24 hr. A positive result is recorded when the
(1). precipitin line between the known positive control wells is
continuous with the line between the test antigen.
Physicochemical Properties
Goose parvovirus is very resistant to physical and chemical Immunologic Techniques
inactivation. Following heating at 65 C for 30 min, no loss of Neutralization tests in goose and Muscovy duck cell cultures or
infectivity was reported (8). The virus is resistant to ether and embryonated eggs using monospecific GPV and MDPV antisera
chloroform and was stable at pH 3.0 for 1 hr at 37 C. Under in vitro can be used to identify the isolated virus. Cross neutralization tests
conditions, infectivity is completely destroyed following treatment can be used to differentiate GPV from MDPV (2). Other methods
with 0.5% formaldehyde (24). have been described, including a plaque reduction test and a sperm
agglutination-inhibition assay but these techniques have not been
Biological Properties fully validated elsewhere (8).
Unlike several mammalian parvoviruses haemagglutination
activity has not been reported for GPV or MDPV using a variety of SEROLOGIC DETECTION IN THE HOST
red blood cells under a variety of conditions (24)
Serologic tests are useful in evaluating the immune status of
Molecular Identification breeding flocks of geese and Muscovy ducks and their progeny,
Goose parvovirus has a single-stranded DNA genome of about particularly in birds that have been vaccinated. The presence or
5600 bases in length. Three major proteins of 91, 78, and 58 kD and absence of parvovirus antibody in breeder geese will determine the
a fourth lighter protein of 51 kD have been identified (8). susceptibility of the progeny. Serology is also a useful diagnostic
Infectious viral particles have a density of approximately 1.38 g/ml tool in confirming recent outbreaks of the disease in goslings and
in cesium chloride (17). Polymerase chain reaction (PCR) and Muscovy ducklings. Ideally both acute and convalescent serum
sequencing has been used to detect and identify both GPV and samples should be tested. Demonstration of yolk-derived antibody
192
Chapter 42 Parvovirus of Waterfowl

in eggs will also provide information on the levels of maternal- ducklings characterized by hepatitis, splenitis, arthritis and
derived antibody in the offspring. tenosynovitis in 2 to 10 -wk-old birds (5,21).
There are very few other pathogens of goslings and Muscovy
Virus-Neutralization (VN) Test ducklings that produce the strict age-specific disease pattern of
The VN test can be performed in embryonating goose or Muscovy GPV and MDPV. The herpesvirus of duck viral enteritis (duck
duck eggs or primary cell cultures derived from them. A GPV plague) produces disease with high mortality in geese and ducks of
isolate adapted to growth in Pekin and Khaki Campbell duck eggs all ages. Isolation and identification of the causal virus will clearly
has been developed for testing goose and Muscovy duck sera for differentiate it from GPV and MDPV. Duck hepatitis viruses also
GPV antibodies by VN test (7). Preferably the test should be cause fatal diseases in ducks less than 6 wk of age. However, these
conducted using constant virus and diluted serum (beta method). viruses are not pathogenic for geese or Muscovy ducks.
Dilutions of heated (56 C for 30 min) sera are reacted with 60-600 Hemorrhagic nephritis and enteritis of geese (HNEG) is associated
median embryo or tissue culture infective doses of virus. After a with a polyomavirus and affects geese from 4 to 20 wk of age
reaction time of 1 hr at 37 C, the culture systems are inoculated with (10,22). The disease was first reported from particular regions of
aliquots of the serum dilution-virus mixture. The tests are France in the 1970s and was referred to as the late form of Derzsy’s
completed after 7 days and the antibody titers are expressed as log2 disease. HNEG has not been reported to occur in Muscovy ducks.
of the reciprocal of the highest dilution of serum causing complete Pasteurella anatipestifer and Pasteurella multocida organisms
neutralization of virus. Titers of 4 (1/16) or greater are considered may also cause high mortality in goslings and Muscovy ducklings.
positive for GPV and MDPV antibody. Treatment of birds with appropriate antibiotics and culture of the
etiologic agent in suitable media will enable differentiation from
Agar-Gel Precipitin Test GPV and MDPV.
Although less sensitive than the VN test, the agar-gel
immunodiffusion (AGID) test is a useful method for rapidly testing REFERENCES
large numbers of sera for the presence of GPV antibodies (7).
Antigen is extracted and concentrated from infected duck embryos 1. Alexandrov, M R. Alexandrov, I. Alexandrov, S. Zachareiva, S
Lasarova, L. Doumanova, R. Pesher and T. Donev. Flourescent and electron
inoculated with a duck-embryo-adapted parvovirus. The tests are microscopy immunoassays employing polyclonal and monoclonal
performed in agar-gel plates containing 1% Ionagar No. 2 or antibodies for detection of goose parvovirus infection. J Virol Methods, 79
agarose in 8% sodium chloride solution buffered to pH 7.8 as 21-32. 1999.
described previously. Diffusion of antigen and antisera takes place 2. Bames, H. J. Muscovy Duck Parvovirus. In: Disease of Poultry, 10th ed
at room temperature and the plates are examined for precipitin lines B. W. Calnek, H. J. Bames, C. W. Beard, L. R. McDougald and Y. M Saif.
after 24 hr. Positive sera can be diluted by twofold series to Eds. Iowa State University Press, Ames, Iowa, USA. pp. 1032-1033. 1997.
ascertain the AGID antibody titer. The AGID test will not 3. Bergmann, V. Pathology and microscopical detection of virus in the
differentiate between GPV and MDPV antibodies. tissues of goslings with Derzsy’s disease (parvovirus infection). Arch Exp
VetMed, 41: 212-221. 1987.
4. Dalibard, V., G. Plassiart, Y.Cherel, M Hurtrel and M Wyers.
Other Serologic Tests Caracterisation des lesions histologiques de la parvovirus spontanee du
Enzyme-linked immunosorbent assays have been developed to canard de Barberie (Cairina moschata). Diagnostic histopathologique
detect GPV and MDPV antibody in both geese and Muscovy ducks differential avec la reovirose. Recuil de Medecine Veterinaire, 169:763-772.
(8). A blocking ELISA, incorporating anti-goose parvovirus-goose 1993.
IgG has been developed and compared to other serologic tests for 5. Gaudrey, D., J. Tetkoff and J-M Charles. A propos d’un nouveau virus
the detection of GPV antibodies (16). The results showed that the isole chez le canard de Barberie. Bull. Soc. Sci. Vet. Med Lyon. 74: 137-
method was rapid, reliable, easily standardized, and correlated well 143. 1972.
6. Gaudrey, D. and D. Fournier. Latest discoveries on waterfowl pathology:
with a virus-neutralization test. This type of ELISA has also been
a new parvovirus of Muscovy ducks. Proceedings of the 9th International
used to detect MDPV antibodies and there was a good correlation Symposium on Waterfowl. Pisa, Italy, September 16-18. pp33-35.1992.
between antibody values and protection against challenge for at 7. Gough, R. E. Application of the agar gel precipitin and virus
least 7 days (15). Problems may arise with the ELISA test if the neutralization tests to the serological study of goose parvovirus. Avian
anti-goose serum and IgG used are not obtained from GPV and Pathol. 13: 501-509. 1984.
MDPV antibody negative birds. Indirect immunofluorescent 8. Gough, R.E. Goose Parvovirus. In: Diseases of Poultry, 11th ed. Y.M
antibody assays have also been used to detect MDPV and GPV Saif, H.J. Bames, J.R. Glisson, AMFadly, L.R. Mcdougald and
antibodies (25,27). D.E.Swayne, eds. Iowa State University Press, Ames, Iowa, USA. Pp 367-
374. 2003.
9. Gough, R. E., V. Ceeraz, W. J. Cox, V. Palya and T Mato. Isolation and
DIFFERENTIATION FROM CLOSELY RELATED AGENTS identification of goose parvovirus in the UK. Vet Record, 151:424. 2005.
10. Guerin, J-L. Hemorragic nephritis of geese (HNEG). In: Disease of
Various techniques have been developed to differentiate GPV Poultry, 11th ed. Y. M. Saif, H. J. Bames, J. R. Glisson, A. M Fadly, L. R.
from MDPV, including; molecular sequencing of the virus genome, McDougald and D. E. Swayne, eds. Iowa State University Press, Ames,
restriction endonuclease analysis studies and VN tests (14,28). A Iowa, USA. pp. 363-367. 2003.
digoxigenin-labeled DNA probe has been developed for the 11. Holmes, J. P., J. R. Jones, R. E. Gough, D. Welchman, Μ E. Wessels
detection of MDPV. The assay proved sensitive for detecting strains and E. L. Jones. Goose parvovirus in England and Wales. Vet Record, 155:
127. 2004.
of parvovirus and differentiating isolates from vaccine-derived 12. Jestin, V. Le point sur l’epizootie actuelle de maladie de Derzsy cher le
strains (19). canard de Barberie. L’Aviculteur, 516:77-78. 1990.
Avian adenovirus-associated viruses are defective parvoviruses 13. Jestin, V., M Le Bras, and M Cherbonnel. Diagnostic de la maladie de
that may be associated with adenovirus infections. In the absence Derzsy. L’Aviculteur, 521. 49-51.199la.
of a helper adenovirus, these parvoviruses are unable to replicate 14. Jestin, V., M-O. Le Bras, M Cherbonnel, G. Le-Gall Recule and G.
under in vitro conditions. Early investigations into outbreaks of Bennejean. Demonstration of very pathogenic parvoviruses (Derzsy’s
Derzsy’s disease (GPV) frequently resulted in the isolation of disease virus) in Muscovy duck farms. Reel. Med. Vet. 167:849-857. 1991b.
adenoviruses, which, at the time, were thought to be the etiological 15. Jestin, V., Μ O. Le-Bras and M Cherbonnel. Control of Muscovy duck
parvoviruses by vaccination. In: M S. McNulty and J. B. McFerran, eds.
agent of the disease (8). Some early reports also attributed the New and Evolving Diseases of Poultry. Commission of the European
disease to reoviruses, which were subsequently shown to be the Communities, Brussels, pp. 167-181. 1994.
etiological agent of a different disease of goslings and Muscovy
193
Richard E. Gough

16. Kardi, V and E. Szegletes. Use of ELISA procedures for the detection of 24. Schettler, C. H. Virus hepatitis of geese. 111. Properties of the causal
Derzsy’s disease virus of geese and of antibodies produced against it. Avian agent. Avian Pathol, 2: 179-193. 1973.
Pathol. 25: 25-34. 1996. 25. Takehara, K., T. Nakata., K. Takizawa, C. K. Limn, K. Mutoh and M
17. Kisary, J. Buoyant density of goose parvovirus strain B. Acta Vet Acad Nakamura. Expression of goose parvovirus VP1 capsid protein by a
Sci. Hung, 23:205-207. 1976. baculovirus expression system and establishment of a fluorescent antibody
18. Kisary, J. Diagnosis and control of parvovirus infection of geese test to diagnose goose parvovirus infection. Arch Virol. 144: 1639-1645.
(Derzsy’s disease). In: Acute virus infections of Poultry. J. B. McFerran and 1999.
M S. McNulty, eds. For the Commission of the European Communities 26. Tatar-Kis, T., T. Mato, B. Markos and V. Palya. Phylogenetic analysis
Marius Nijhoff Publishers, Dordrecht, The Netherlands. pp239-242. 1986. of Hungarian goose parvovirus isolates and vaccine strains. Avian Pathol,
19. Le Gall- Recule, G and V. Jestin. Biochemical and genomic 33: 438-444. 2004.
characterisation of Muscovy duck parvovirus. Arch Virology, 139: 121-131. 27. Wool cock, P. R, V. Jestin, H. L. Shivaprasad, F. Zwingelstein, C.
1994. Amauld, M D. McFarland, J. C. Pedersen and D. A. Senne. Evidence of
20. Lukashov, V. V. and J. Goudsmit. Evolutionary relationships among Muscovy duck parvovirus in Muscovy ducklings in California. The Vet
parvoviruses: virus-host coevolution among autonomous primate Record, 146: 68-72. 2000.
parvoviruses and links between adeno-associated and avian parvoviruses. J 28. Zadori, Z., J. Erdei, J. Nagy and J. Kisary. Characteristics of the genome
of Virol, 75:2729-2740. 2001. of goose parvovirus. Avian Pathol. 23: 359-364. 1994.
21. Palya, V., R. Glavits, M Dobos-Kovacs, E. Ivanics, E. Nagy, K. 29. Zadori, Z., R. Sefancsik, T. Rauch and J. Kisary. Analysis of the
Banyai, G. Reuter, G. Szucs, A. Dan and M Benko. Reovirus identified as complete nucleotide genome sequences of goose and Muscovy duck
cause of disease in young geese. Avian Pathol. 32: 129-138. 2003. parvoviruses indicate common ancestral origin with Adeno-associated virus
22. Palya, V., E. Ivanics, R. Glavits, A. Dan, T. Mato and P. Zarka. 2. Virology. 212: 562-573. 1995.
Epizootic occurrence of haemorragic nephritis enteritis virus infection of
geese. Avian Pathol. 33: 244-250. 2004.
23. Roszkowski, J., P. Gazdzinski, W. Kozaczynski and M Bartoszcze.
Application of the immunoperoxidase technique for the detection of
Derzsy’s disease virus antigen in cell cultures and goslings. Avian Pathol,
11: 571-578. 1982.

194
43
CELL-CULTURE METHODS
Karel A. Schat and Holly S. Sellers

SUMMARY. Cell cultures are routinely used for the isolation, identification, and propagation of viruses. Successful use of cell cultures
requires proper techniques for the preparation and sterilization of glassware. Several types of media formulations are used for the in vitro
propagation of cells. The compositions of some of the commonly used media are presented. Techniques for the preparation and storage of
primary and secondary cultures using chick embryo fibroblasts, chicken kidney cells, chick embryo kidney cells, and lymphocytes are
described in detail. In addition, brief descriptions are given for the preparation of chick embryo tendon cells and intestinal epithelial cells.
The application of cell cultures for virology is discussed.

INTRODUCTION dependent cells (3, 5, 8, 11). Plastic dishes for bacteriology use
cannot be used for cell cultures.
Cell cultures are routinely used in diagnostic laboratories for the Depending on the application, cell monolayer cultures can be
isolation, identification, and propagation of viruses, coccidia, and propagated in a variety of plastic culture vessels including multi­
for the detection of virus-neutralizing antibodies. Cell cultures have well plates, petri dishes, flasks and roller bottles. For monolayer
many advantages over animals or embryonated eggs for such uses. cultures, the cell yield is directly proportional to the available
Cell cultures are economical, and they are a relatively homogentOUS Surface area of the flask. Virus neutralizations and titrations can
population of cells, free of immunologic and hormonal influences easily be scaled down to a 96-well microplate format requiring
that might affect replication of the virus. Many types of cells can be smaller reagent volumes and easily enabling multiple sample
stored in liquid nitrogen and thus be readily available. In addition, replicates. Petri dishes and flasks are available in a variety of sizes
recent advances in the cloning, sequencing, and expression of avian accommodating a couple of milliliters to several hundred milliliters
cytokines and growth factors will allow the development of cell of media and cells.
cultures using specialized cell types, which cannot be grown using Plastic vessels are routinely sterilized after use by autoclaving and
conventional approaches. The use of specific-pathogen-free (SPF) discarded. It is possible, however, to recycle used plastics by the
material is essential for the successful propagation of cells and following method:
subsequent use for virus isolation. Occasionally, non-SPF eggs may 1) Disinfect the used plastics for 16 hr in 2% sodium hypochlorite
be used for cell culture as a means of directly isolating virus. or in a household microwave oven (2.45 gHZ) for 10 min.
The large number of sequential operations involved in cell culture 2) Wash at least six times in tap water.
means that aseptic techniques are of the utmost importance even in 3) Drain and rinse six times in distilled wafer.
the presence of antibiotics. Contamination risks can be greatly 4) Drain and dry in an incubator at 48 C.
reduced using laminar-flow hoods (horizontal flow for medium 5) Sterilize the plastic materials in a household microwave oven
preparation and vertical flow for pathogenic and nonpathogenic (2.45 gHz) for 3-6 min with the power selection on high. Place a
viruses). Even so, cell culture can be performed satisfactorily in a beaker with 250-500 ml of water in the oven as a heat sink. It is
clean, well-lighted room where dust and traffic are kept to a important to cool down the oven between runs and to keep the
minimum. Aseptic techniques and the speed and dexterity of the plastics dry. Petri dishes can be stacked three high, and flasks
operator are very important factors under such conditions. Freshney should be placed upright with the caps loose. Caps may have to be
(6) and Versteeg (12) provide additional information on cell sterilized separately depending on the manufacturer of the flasks.
cultures. This procedure will allow the use of plastic tissue-culture material
for 10-30 cycles before it starts to deteriorate.
LABORATORY EQUIPMENT Although glassware has been largely replaced by plastics,
glassware can be used for growing cells, but careful washing is
If the culture media and solutions are to be prepared in the essential. The following general procedure is recommended for the
laboratory in reusable glassware, a large supply of pure water must preparation of all glassware used in tissue culture:
be available. The water must be deionized, double-distilled, or both, 1) Disinfect all contaminated material by autoclaving at 20 pounds
to remove all traces of cytotoxic organic and inorganic materials. per square inch (psi) for 20 min.
Reverse osmosis (RO) (Bamstead International, Dubuque, Iowa, 2) Rinse in tap water to wash away all solid matter; if necessary,
and other companies) followed by glass distillation is often used. brush. New glassware (optional) or badly soiled glassware must be
Pure water must be stored and transported in glass or glass-lined immersed for 12 hr in a sulfuric acid-dichromate cleaning solution.
containers. (Dissolve 120 g Na2Cr207.2H20 in 1000 ml tap water and add 1600
Cell attachment and proliferation on the surface of a culture vessel ml concentrated sulfuric acid, technical grade. Place the container in
is critical for the growth of anchorage-dependent cells, which ice-cold water during the preparation, and use safety glasses and
require a nontoxic, biologically inert, and optically transparent heavy gloves.) Rubber stoppers can be treated in 0.5 N sodium
surface that will allow cells to attach and allow movement for hydroxide.
growth. Cells attach and grow well on glass and since it is clear, it is 3) Soak small items in hot water with 75 ml Micro-90® cleaning
ideal for direct examination of attached cells by microscope. Glass solution (Cole Parmer Instrument Company, Vernon Hills, IL) per 4
was the primary material of choice until the 1960s when plastic liters of water for at least 1 hr. Place bottles, flasks, and other larger
culture vessels became available. In most laboratories glass has items in warm water (52 C) with Micro-90® in the ultrasonic
been replaced by plastic, which has greater consistency, superior glassware washer. Put pipettes in holders after removal of the cotton
optical properties, is supplied sterile and is disposable. Although plugs and place them in the glassware washer as is done with other
several plastic substrates are available for this purpose, polystyrene glassware. Micro-90® can be replaced by other alkaline
is the most common, and most economical. Through chemical, disinfectants that do not adsorb to glass.
electric ion discharge or gamma irradiation, the surface of the 4) Rinse at least twice with tap water and twice in RO or double­
polystyrene is modified such that it becomes hydrophilic and distilled water. Wash pipettes in cold water rinsers for at least eight
negatively charged thus allowing attachment of most anchorage rinses with tap water, and then immerse them in RO water.
5) Drain all glassware on draining racks or in a drying oven.
195
Karel A. Schat and Holly S. Sellers

Table 43.1. Compositions of commonly used cell culture media.A


No. Name medium Components Amount
M199,10x 90.0 ml
Tryptose phosphate broth 100.0 ml
1 M20b NaHCO3,10% 6.3 ml
Antibiotic mixture 10.0 ml
Deionized water, qs 1000.0 ml
Ml 99 powder 4.5 g
Ham’s F10 powder 5-5 g
2 F10-199c Tryptose phosphate broth 7.5 ml
NaHCOa 0.85 g
Antibiotic mixture 10.0 ml

Leibovitz’s L-15 powder 8.76 g


McCoy’s 5A (modified) powder 5.0 g
3 Leib-McCoyc NaHCOa 0.94 g
Antibiotic mixture 10.0 ml
Deionized water, qs 1000.0 ml

Lactalbumin hydrolysate, 10
* 100.0 ml
Hanks’ BSS, Ιθχ 90.0 ml
4 LH° Antibiotic mixture 10.0 ml
Deionized water 800.0 ml
NaHCO3 solution, qs to adjust pH to 7.3-7.5

BME powder 9.18g


Tryptose phosphate broth 3.54 ml
5 BMEe NaHCOa 1.04 g
Antibiotic mixture 10.0 ml
Deionized water, qs 1000.0 ml

Leibovitz’sL-15, lx 385.0 ml
McCoy’s 5a, modified, 1 χ 385.0 ml
NaHCOa, 10% 2.0 ml
Tryptose phosphate broth 50.0 ml
2-Mercaptoethanol, 1 mM 10.0 ml
6 LMHf
Sodium pyruvate, 100χ 10.0 ml
Glutamine solution, 200 mM 10.0 ml
Antibiotic mixture 10.0 ml
Chicken serum 100.0 ml
FBS 80.0 ml
Aqs = sufficient quantity; BSS = balanced salt solution; BME = basal medium, Eagle; FBS = fetal bovine serum; CK = chicken kidney; CEl· = chick embryo fibroblast.
bM20 supplemented with 5% FBS (M25) is used as growth medium for CK cells. M20 supplemented with 2% FBS (M22) is used as growth medium for CEFs. Maintenance medium contains
0.25% FBS (M20.25).
c F10-199 and Leib-McCoy supplemented with 4% calf serum are used as growth medium for duck embryo fibroblasts and CEFs. For maintenance medium add 1% calf serum We currently
use Fl0-199 with 0.25% FBS as maintenance medium for CK cells..
dLH supplemented with 10% calf serum is used as growth medium for CEFs. Maintenance medium contains 2% calf serum
eBME supplemented with 5% FBS is used as growth medium for CK cells. Maintenance medium contains 1% FBS.
fLMH is used for cultivation of lymphocytes and lymphoblastoid cell lines. RPMI-1640 can be used to substitute McCoy and Leibovitz. Certain cell fines require considerably less serum.

6) Plug pipettes, place in canisters, and sterilize with dry heat at 160 CELL-CULTURE MEDIA AND STOCK SOLUTIONS
C for 2 hr after the oven gets up to temperature. Wrap small items
and package these items. Materials that will melt or bum can be Culture Media
sterilized by autoclaving (20 psi for 20 min). The use of indicator Cell-culture media must provide the cells with necessary nutrients
tape or ampoules is recommended to ensure that proper and adequate conditions for a normal metabolism. Some media,
temperatures are reached. such as Ml99 and RPMI-1640, are complex, containing 50-100
In addition to the usual glassware and equipment found in a ingredients, and are probably better purchased (either aslOx
bacteriologic laboratory, necessary equipment includes an inverted concentrate or in powdered form). Simpler media, such as Earle’s
microscope, and if cells are cultured in dishes or tubes open to the BSS with lactalbumin hydrolysate, are easily prepared in the
atmosphere, an incubator in which the atmosphere can be laboratory.
maintained with 85% relative humidity and 3-5% carbon dioxide. The choice of medium and the amount and source of serum added
Incubator cleanliness is important to prevent gross contamination of to the medium will depend on a number of factors, such as cell
the working area, thereby inadvertently contaminating the cultures. requirements, intended use of the cell cultures, and price and
Because the high level of humidity in the incubator favors growth availability of the media components and sera. Additional vitamins,
of molds, routine disinfection of the incubator is recommended. amino acids (particularly L-glutamine), and chicken embryo extract
Mouth pipetting is not acceptable. Use either a rubber pipette filler are frequently added to improve growth. The addition of 10% TPB
or an electric apparatus (e.g., Pipet-aid, Drummond Scientific Co., facilitates transformation of cells by Rous sarcoma viruses. Table
Broomall, PA). 43.1 gives examples of media formulations used in several research
laboratories.

196
Chapter 43 Cell-Culture Methods

Table 43 2. Ingredients for preparation of 1 χ concentration of balanced salt solutions (BSSs) and phosphate-buffered saline (PBS).
SolutionA Component Amount®
Hanks’ BSS Earle’s BSS Dulbecco’s BSS PBS
Water 800 800 800 1000
A NaCl 8.0 6.8 8.0 8.0
KC1 0.4 0.4 0.2 0.2
MgSO4.7H2O 0.2 0.2 — —
KH2PO4 0.06 — 0.2 0.26
Na2HPO4.2H2O 0.06 — 0.14 0.57
NaH2PO4.H2O — 0.14 — —
Glucose 1.0 1.0 — —
Phenol red, Na 0.017 0.017 0.017 0.017
B Water 100 100 100 —
CaCl2 0.14 0.2 0.1 —
C Water 100 100 100 —
MgCl2.6H2O — — 1.0 1.8C
NaHCO3 0.35 2.2 — —
A Add solution A to solution B. Sterilize solutions A-B and C by autoclaving at 10 psi for 10 min. Solution C can be sterilized by pressure filtration. After
cooling, solutions A-B and C can be mixed. Omit solutions B and C if these components are not needed. In that case, add 200 ml of water to solution A.
B All quantities are given in g, except water, which is in ml. Use less water than indicated; after all components are in solution, add water to the required
amount. For 10* solutions, use 10* the amounts except for water; solution C is omitted from Hanks' BSS 10χ and Earle's BBS 10*.
c Added directly to solution A.

All media have the following basic composition: Trypsin and Versene (TV) Solution
1) A balanced salt solution. Stock solutions of up to 2.5% trypsin can be prepared or obtained
2) A protein supplement such as serum or amniotic fluid, or protein commercially. The following 10x stock solution of trypsin and
derivatives such as lactalbumin hydrolysate or tryptose-phosphate versene (TV) is often used for avian cell cultures and is prepared as
broth, or amino acids.3) An antibiotic mixture to control accidental follows. Dissolve in 1000 ml of glass-distilled water in order as
microbial contamination. However, the use of antibiotics is listed: 85 g NaCl, 4.0 g KC1,10 g glucose, 3.5 g NaHC03, 106 units
controversial in permanent cell lines because it may mask low-level of penicillin G sodium, 1 g streptomycin sulfate (see below), 5 g
bacterial contaminations. trypsin 1:250 (Invitrogen, Carlsbad, CA and other companies), 2.5 g
Although many of the media components can be bought EDTA, and 20 ml of 0.5% phenol red solution (Invitrogen). Stir for
commercially, it is sometimes easier or necessary to prepare these 2-3 hr at room temperature or overnight at 4 C. Not all of
components in the laboratory. It is imperative to use pure water and the trypsin will go into the solution. The insoluble parts can be
reagents of a tissue culture grade. removed by centrifugation at 180 x g for 20 min or by prefiltration
through 1-2.5 cm of Celite filter aid on a Buchner funnel. Sterilize
Balanced Salt Solutions (BSSs) and Phosphate-Buffered Saline TV solution by filtration. Dispense in 50-100 ml quantities and
(PBS) freeze at -20 C. Use as lx TV solution (final concentrations: 0.05%
Hanks’ BSS (HBSS) and Earle's BSS are frequently used as bases trypsin and 0.025% versene) by mixing 100 ml 10x TV solution
for growth media (Table 43.2). Their function is to maintain a with 900 ml of sterile glass-distilled water. Warm the TV solution
physiologic pH (7.2-7.6) and osmotic pressure and to provide to 37 C before use.
water, glucose, and inorganic ions needed for a normal cell
metabolism. BSSs usually contain a pH indicator (e.g., phenol red). Sodium Bicarbonate Solution
BSSs and PBS are also frequently used to wash inocula and dead To simplify control of pH, sodium bicarbonate is often omitted
cells from cultures, to remove serum-containing media before from salt solutions and media. Sodium bicarbonate is added to the
trypsinization, and to dilute trypsin solutions. However, when medium just before use. Various concentrations from 1.4 to 10%
versene (ethylenediaminetetraacetic acid [EDTA]) is used as the have been used. The authors use a 10% solution in double distilled
cell dispersant, Dulbecco's BBS without calcium and magnesium or RO water sterilized by filtration under positive pressure.
salts (solution A [Table 43.2] made up to 1000 ml of water) should Autoclaving (see Table 43.2) or negative-pressure filtration will
be used for washing cells and diluting the versene. convert the sodium bicarbonate to sodium carbonate. An alternative
When the solutions are being prepared, each salt must be approach is the use of equimolar concentrations of sodium
completely dissolved before the next one is added. For convenience, carbonate and bicarbonate. Store at room temperature in small
Hanks' and Earle's stock solutions A and B (Table 43.2) can be aliquots in tightly capped tubes.
prepared at 10* concentration. In this case, it is advisable to add
NaHC03 separately to the lx solution. The stock solutions can be Neutral Red Solution
autoclaved separately and, after cooling, mixed slowly with A 1% solution of neutral red (Invitrogen) is prepared in water,
constant stirring. Alternatively, they can be mixed as above, sterilized by filtration, and stored at room temperature. A 0.33%
sterilized by filtration, and stored frozen or at 4 C. For use, one part solution is available commercially (Invitrogen).
of the 10x solution is added to nine parts of water, and the solution
is finally sterilized either by filtration or by autoclaving. It is Antibiotic Solution
advisable to omit phenol red when the BSSs or PBS solutions are Antibiotic-antimycotic solutions are commercially available
used in the preparation of materials for immunohistochemical (e.g., from Invitrogen). These solutions contain penicillin G
staining or for molecular biology. sodium (10,000 IU/ml), streptomycin sulfate (10,000 pg/ml),
and amphotericin B (Fungizone®, Invitrogen) (25 pg/ml) in
sterile saline. Add 1 ml of this solution to 100 ml of medium. If it is

197
Karel A. Schat and Holly S. Sellers

Table 43.3. Composition of agar overlay media.


No. Name Composition"41 Amount (ml)
1 2- M20® Ml 99,10x 90.0
NaHCO3,10% 6.3
Tryptose phosphate broth 50.0
Antibiotic mixture 10.0
Deionized water 400.0
Agar 2% 400.0

2 2* LHb Earle’s balanced salt solution 2* containing 1.3% lactalbumin hydrolysate


1% neutral red solutionc 500.0
Antibiotic mixture 10.0
Agar, 1.6%—3% 10.0
480.0
3 2x F10-199 M199, 2χ 225.0 225.0
Ham’s medium F10,2χ 50.0
Tryptose phosphate broth 50.0
Calf serum 10.0
Antibiotic mixture —
NaHCO3, 7.5%, sufficient quantity to adjust pH to7.3-7.5
Agar 1.6% 450.0

A The agar is melted and cooled to 42-45 C; the 2* medium is warmed to 42 C, and the two parts are mixed.
B Can be used with or without serum.
c Neutral red is used for staining cultures infected with cytopathic agents. Live cells stain red and dead cells (plaques) are not stained.

difficult to obtain the commercially available solutions, it is autoclaving at 20 psi for 20 min. After cooling, the agar can be
possible to prepare these solutions in the laboratory. In that case stored at room temperature in tightly capped bottles. Just before use,
vials of penicillin G sodium and dihydrostreptomycin sulfate are the agar is melted in a microwave oven or in a boiling water bath
opened aseptically and dissolved in sterile glass-distilled water (or and then brought to 45 C in a water bath. The agar solution should
PBS) to give 10,000 IU and 10,000 pg, respectively, per ml of stock not be repeatedly melted and solidified. Bactoagar will be
solution. If not treated aseptically, the solution can be sterilized by satisfactory for most uses, but the replication of some viruses is
filtration. Amphotericin B (Fungizone®) can be added at 25 pg/ml. inhibited by the presence of anionic, sulfated polysaccharides.
Amphotericin B is insoluble and must be added aseptically. The Addition of 100 pg diethylaminoethyl-dextran per milliliter of agar
mixture should be stored frozen in convenient volumes. For use, it medium or the use of Noble agar or agarose will reduce this
is thawed and added to all media and solutions in cell culture at a problem.
rate of 1 ml of the antibiotic stock to 100 ml of medium. This will Formulations for agar overlays can be found in Table 43.3. A final
give a final concentration of 100 IU of penicillin, 100 pg of concentration of 0.8-1.0% agar produces a firm gel and can be used
dihydrostreptomycin sulfate, and 0.25 pg of amphothericin B per for virus assays and long-term cultures. For cloning or visualization
milliliter of medium. Higher concentrations are occasionally used. of virus plaques with neutral red, a 1.5% agar overlay is
Stock solutions should not be repeatedly frozen and thawed. An recommended. When coverslips are to be removed from beneath the
alternative to penicillin-streptomycin is gentamicin sulfate at 10 agar, a soft agar containing 0.4-0.6% agar can be used. Note that
mg/ml; use 0.1 ml/100 ml of medium. The use of antibiotics is not the ingredients cannot be mixed and then melted, because the serum
recommended for the control of Mycoplasma. Proper testing of will coagulate. Melt the agar in a microwave oven, and cool to 42-
serum (see below), use of SPF material to establish cell cultures, 45 C in a water bath. Prepare the 2* medium and warm to 37-42 C,
and the use of a Pipet-aid will prevent contamination with mix both parts as recommended, and add to the cultures. The final
mammalian, avian, and human Mycoplasma. temperature of the agar medium will fall fairly rapidly and will not
cause damage to the cells.
Lactalbumin Hydrolysate
This powder is available commercially (Sigma-Aldrich Chemical Sera
Co., St. Louis, MO). The hydrolysate solution is commonly Fetal bovine serum (FBS), calf serum, and chicken serum are most
* solution (2.5%, w/v) in HBSS or Earle’s BSS
prepared as a 10 commonly used in media preparations for avian cell cultures.
without sodium bicarbonate. The solution is sterilized by However, sera from other species can be used successfully. FBS
autoclaving and stored at room temperature. The final concentration and calf serum are best bought commercially, whereas chicken
in the medium is 10% of the stock solution, or 0.25% (w/v). serum can be bought commercially, but testing for the absence of
avian viruses is not always satisfactory. Due to the increased risk of
Tryptose Phosphate Broth (TPB) spreading bovine spongiform encephalitis (BSE) among cattle, the
Tryptose phosphate broth (TPB) (BD, Franklin Lakes, NJ) is country of origin and batch numbers are tracked on all bovine
prepared by dissolving 29.5 g in 1000 ml of water. TPB is serum shipped to the U.S. and Australia. The following procedure is
dispensed, sterilized by autoclaving, and stored at 4 C or room suggested if serum must be prepared from blood: 1) collect blood
temperature. It is used as a constituent of media, particularly for from different individuals from the same species individually and
focus formation by Rous sarcoma virus. TPB is also a good allow it to clot spontaneously; 2) incubate the clotted blood at 37 C
substrate for bacteriologic sterility control: add 1-2 ml of test media for 2 hr (afterwards it can be stored overnight at 4 C); 3) remove the
to 5 ml of TPB and incubate at 37 C for up to 1 wk. serum and centrifuge at 2000 * g for 30 min to remove the
erythrocytes; 4) individual sera can be pooled, but keep pools
Agar separate, and 5) serum can be collected aseptically or sterilized by
Depending on the use, 1.6-3% solutions of bactoagar (BD) are filtration, although it clogs most filters rapidly (see section on
prepared in water. The agar is dissolved and sterilized by sterilization).
198
Chapter 43 Cell-Culture Methods

All batches of sera including those purchased from commercial PREPARATION OF CELL CULTURES
sources should be tested for support of cell growth and for the
presence of Mycoplasma. Most suppliers of sera will supply one Preparation of cell cultures involves removing organs aseptically
100-ml bottle of serum of a given lot to allow testing before from freshly killed animals or from embryonating eggs. The organs
purchasing. The following procedure is recommended for testing to are mechanically cut up into small pieces, washed to remove
support cell growth: 1) set up cultures using lx and 0.5x the regular erythrocytes, and then dispersed into a suspension of single cells by
number of cells, and 2) make a passage (subculture) when the enzymatic digestion with trypsin. The cells are washed free of the
cultures are confluent, preferably followed by a second subculture. trypsin, suspended in a growth medium, counted, and placed in
Subtle differences between batches of serum may not be evident appropriate tissue-culture vessels. These are placed in an incubator,
until after the second subculture. It is important to test the batches and the cells are allowed to grow. Of the innumerable procedures
of serum on all cell types in use. used, the following are recommended.
The medium of the first test cycle can be used for Mycoplasma
testing (cell culture is an excellent enrichment method for Chick Embryo Fibroblast (CEF) Cultures
Mycoplasma). The following method will allow detection of most This procedure can be used for embryo cell cultures of many
common contaminating Mycoplasma species: 1) prepare liquid different species. Embryos are used at about the midpoint in the
medium (Fabricant’s medium Π) using heart-infusion broth (BD) incubation period (9-11 days for chicken embryos).
supplemented with 10% swine serum, 10% fresh yeast extract (see 1) Candle a group of 9 to 11 day-old embryonated chicken eggs for
below), 1000 IU/ml penicillin G sodium, and 0.1% thallium acetate; viability.
2) plates are made as above but with heart-infusion agar (BD) 2) Clean the shell surface by immersing in alcohol for 5 min or by
instead of broth; 3) inoculate 0.2 ml of the sample in a tube with 2 swabbing with tincture of iodine. Do not use soiled eggs.
ml Equid medium; 4) incubate for 3 days at 37 C, plate a loopful on 3) Holding the egg in a gloved hand, allow any excess alcohol to
a quadrant of a plate, and incubate in a candle jar with moist drip off, cut around the shell just below the air cell, and flip away
atmosphere; and 5) examine the plate after 6 days for Mycoplasma the piece of the shell. Remove the embryo by placing the forceps
colonies. Proper preparation of yeast extract is essential and is done under its neck and lifting the embryo out with care so that it breaks
as follows: 1) warm 1000 ml of water to 45 C, add 250 g live loose from its yolk sac. Do not use embryos soiled with yolk; the
bakers' yeast, simmer for 15 min, and cool the suspension; 2) yolk is toxic for cells.
centrifuge at 16,500 x g for 30 min; 3) filter the supernatant through 4) Hold the embryo over a sterile trypsinization flask (Bellco
filter paper and autoclave in bottles or tubes; and 4) store the extract Biotechnology, Vineland, NJ) in such a way that when the head is
at -20 C if desired. Instead of using this method it is also possible cut off, the head falls outside the flask and the body falls inside. The
to test the supernatant fluid by polymerase chain reaction (PCR) embryos can also be placed in a beaker and minced with scissors.
using appropriate primers, but this approach may miss some The eyes, limbs, and viscera can be removed before mincing. This
Mycoplasma species. will yield more homogeneous primary cultures, but it is more
laborious.
Chicken Embryo Extract 5) Transfer the minced tissues to a trypsinization or Erlenmeyer
Eight-to-10-day-old SPF chicken embryos are recommended. flask, or a prescription bottle. Wash the pieces by adding 3 ml
After discarding the eyes, macerate the embryos by forcing them sterile PBS per embryo, swirl, allow the pieces to settle, and decant
through a syringe and an 18-20 gauge needle into a flask containing the supernatant fluids. Repeat the wash until the PBS is clear and
an equal volume of PBS. Store this cell suspension overnight at -20 free of blood, and then pour out as much PBS as possible.
C, then thaw it, centrifuge it for 15 min at 500 x g, and save the 6) Add 40-50 ml prewarmed (37 C) TV solution, shake vigorously,
supernatant. This process can be repeated twice more. After the and incubate at 37 C with intermittent shaking, or use a magnetic
final centrifugation, store the supernatant at -20 C. stirrer. After 5 min, allow the pieces to settle and carefully pour the
supernatant suspension through two layers of sterile cheesecloth
Sterilization into a beaker. Transfer the suspension into large centrifuge flasks
Many of the constituents and sometimes the complete media can containing 5% (v/v) calf serum or FBS and place on ice for at least
be purchased sterile. Some solutions can be sterilized by 5 min. If the embryos were not minced with scissors, break the
autoclaving, for example, PBS, agar solution, versene solution, embryos up in PBS by allowing the stirring bar to rotate moderately
neutral red solution, TPB, lactalbumin hydrolysate, and distilled fast and wash once with TV solution before trypsinization.
water. However, media containing thermolabile compounds, such as 7) Repeat the procedure three or four times until only the white
amino acids, antibiotics, serum, or trypsin, cannot be autoclaved fibrous tissues are left and no more cells disperse, that is, the trypsin
and must be sterilized by filtration. Pressure filtration through is nearly clear. Mix the cellular suspensions collected from all the
membrane filters following procedures recommended by the trypsinizations. Alternatively to trypsinizing three to four times,
manufacturers (Millipore Corp., Billerica MA. or Pall Corporation, trypsinization is possible once for 1 hr; use 20 ml of TV solution
Ann Arbor, MI) is highly recommended. Usually, a single sterile per embryo and incubate for 1 hr at 25 C or 37 C with constant
filter is satisfactory for filtering solutions other than serum or stirring. Harvest the cells as described above.
trypsin. Special filters and filter holders recommended for the 8) Centrifuge for 10 min at about 350 x g. Refrigeration is optional.
filtration of serum products are available (Millipore, Pall). Remove and discard the trypsin.
9) Add 100 ml of growth medium (Table 44.1) for each milliliter of
packed cells, and break up the cell pellets by gentle repeated
pipetting or by gently using a vortex mixer.

Chick Embryo Kidney (CEK) or Chicken Kidney (CK) Cell


Cultures
These cultures can be prepared from kidneys of 19-to-20-day-old
SPF chick embryos or 1 to 3 wk-old SPF chicks. The general
procedure is as follows:
1) For embryo kidneys, remove embryos from the eggs aseptically.
Remove kidneys and collect them in a petri dish containing PBS.
199
Karel A. Schat and Holly S. Sellers

For CK, euthanatize chicks by cervical dislocation or CO2 4) Wash the cells in PBS and resuspend in a lymphocyte medium
inhalation (consult your Institutional Animal Care and Use (e.g., LMH in Table 43.1). Count the cells and adjust to 1 χ 106 to 5
Committee policies for the approved method) and dip in χ 106 cells/ml. Incubate the cells at 41 C in a CO2 incubator.
disinfectant. Peel back the skin and open the abdominal cavity. Viability decreases rapidly within 24—72 hr. The addition of
Remove kidneys aseptically and place them into a prescription conditioned medium (CM) (see below) or stimulation with mitogens
bottle with PBS. for 72 hr followed by the addition of CM can prolong the viability
2) Wash the CK two or three times with PBS by shaking the bottle of the lymphocytes. Cloned avian cytokines are becoming
very vigorously. Allow the kidney pieces to settle and discard the increasingly available as commercial products or can be prepared in
supernatant fluid. Washing is not necessary with CEKs. research laboratories using molecular techniques. The use of these
3) Wash one time as above with 50 ml of prewarmed TV solution interleukines is expected to facilitate the culture of lymphocytes,
Allow the suspension to settle for 2 min before discarding the especially after stimulation with mitogens.
supernatant fluid. Conditioned medium containing interleukins can be prepared as
4) Trypsinize as follows: Add 40-50 ml of fresh, warm TV follows: Prepare spleen lymphocytes as above and resuspend at 2 χ
solution. Shake vigorously and place the bottle at 37 C for 5 min. 107 cells/ml in LMH without serum (LM-base) or with 2% chicken
Carefully decant off the supernatant suspension, passing it through serum (LM-2). Stimulate the cells with concanavalin A bound to
two layers of cheesecloth or monofilament screening fabric sepharose beads (GE Healthcare). Wash the beads several times
(SEFAR America, Depew, NY; cat. no. 03-94/46) into a beaker or with a wash buffer (0.5 M NaCl, 20 mM Tris, pH 7.4) and
flask containing 5% (v/v) calf serum. Keep these cells at 4 C. Care equilibrate the beads overnight in three volumes of LM-base. Mix
must be taken to retain the kidney pieces in the bottle for further equal volumes of lymphocytes and beads at a ratio of 107
trypsinizations. lymphocytes/ml and 0.3% (v/v) beads. Incubate the mixture for 48
5) Repeat step 4, each time adding the harvest to the same vessel hr and harvest the supernatant fluid. This method is far superior to
until enough cells are collected or until the kidney tissue is the use of concanavalin A bound to red blood cells (B. W. Calnek
exhausted. and K. A. Schat, unpubl. data). The supernatant fluid can be used as
6) Pour the cells into centrifuge tubes, centrifuge (10 min at 350 x a source of CM or can be further purified, if desired. Freeze at -70
g) and resuspend the pellet in an appropriate growth medium (Table C for long-term storage.
43.1). Count the cells as outlined for CEFs; count small cell clumps Chick Embryo Liver Cells. Use 14 to 16 day-old embryos.
as one cell. Seed 0.5 χ 106 to 1 χ 106 cells/ml for stationaiy Harvest the livers and mince finely. Liver cells are fragile, so
cultures; double the cell numbers for roller cultures. A complete trypsinize with care. Do not shake or stir vigorously.
monolayer will form in 48 hr. CK cells can easily be Chick Embryo Lung, Heart, and Gonadal Cells. Use 14 to 16
overtrypsinized; the best results will be obtained when numerous day-old embryos for lung cells and 20-day-old embryos for heart
small clumps of three to five cells are present in the suspension. and gonadal cells. Prepare the cells in the same way as CEFs.
7) Calculate the cell yield. A 1% suspension of packed cells as Chick embryo tendon (CET) cells. Harvest the flexor tendons
described above contains about 2 χ 106 cells per milliliter. A more from the feet of 17 to 18-day-old embryos. Incubate the tendons
accurate determination of the number of viable cells requires use of overnight in Ham’s F10 medium supplemented with 50 pg/ml Na
a hemocytometer and a microscope. The latter method includes ascorbate and antibiotics, followed by digestion with 0.25% trypsin
transferring 1 ml of the cell suspension to a tube, adding 1 drop of and 0.2% collagenase for 2-3 hr in Dulbecco’s modified Eagle
1% trypan blue dye, and mixing well. For best results, incubate the medium supplemented with Na ascorbate and antibiotics. Culture
cells with trypan blue for 5 min at room temperature. Cells stained the CET cells in Ham’s F10 medium supplemented with 50 pg/ml
blue are dead and unstained cells are alive. Count all unstained Na ascorbate, antibiotics, and 5% FBS. CET cells have been used
cells; doublets, triplets, and clumps are counted as one cell. for studies of avian reoviruses (7).
Suspensions should have more than 95% Eve cells. If fewer than Avian Intestinal Cells. Recently, avian intestinal cell cultures have
80% are live by this test, the cells are not suitable for culture. Seed been developed and may provide useful tools for the isolation of
cells at 1 χ 106 to 2 χ 106 cells/ml for stationaiy cultures and at 4 χ avian intestinal pathogens and the study of coccidia replication.
106 ml for roller bottles. These cell concentrations will give a Procedures have been reported using turkey neonatal cecal cells and
monolayer in 24 hr; lower cell numbers can be used if confluent turkey neonatal intestinal cells (2, 4). Ceca are excised, flushed
monolayers are not required within 24 hr. several times with HBSS containing antibiotics, slit longitudinally,
and minced into small fragments. Wash the fragments vigorously
Preparation of Other Cell Types several times with HBSS, mince the pieces further with scalpels,
Lymphocyte Cultures. Many viruses will replicate in resuspend the minced pieces in HBSS containing 300 U/ml
lymphocytes. Short-term cultures can be prepared from splenic collagenase and 0.1 g/ml dispase, and incubate for 30 min with
lymphocytes as follows: stirring. Vigorously pipet the pieces and harvest the cells.
1) Remove spleens from chickens as described for kidneys and Resuspend the cells at 1 χ 106 to 2 χ 106 cells/ml growth medium
place the spleens in PBS. consisting of M199 supplemented with 4.5 mg/ml glucose, 5% FBS,
2) Force the spleens gently through a 60-nm nylon bolting screen 5 pg/ml heparin, 2.5 μg/ml insulin, 10 ng/ml epidermal growth
(Sefar America, Depew, NY; cat. no. 3-95/39) using a sterile factor, standard antibiotics, and 0.2% gentamycin. These cells can
syringe plunger and PBS. The nylon screen can be placed over a be passed several times. An avian cell line has been established (4).
small glass beaker, covered with aluminum foil, and autoclaved. Turkey neonatal intestinal cells can also be obtained from
Alternatively, individually wrapped, sterile cell strainers can be longitudinally opened fragments of the total intestinal tract (2).
obtained from BD (Falcon catalog # 352350). Incubation of the fragments in 0.15% TV-acetyl cysteine for 20 min
3) Pour the cell suspension into tubes and centrifuge for 10 min at at room temperature with gentle swirling will remove the mucus.
350 χ g, resuspend the cells in PBS, and layer the suspension Cells are obtained by gently shaking 2-3 cm fragments in Joklik’s
carefully on top of 2-4 ml of Ficoll-Paque (GE Healthcare, modified minimum essential medium (Invitrogen) supplemented
Piscataway, NJ). Centrifuge at 400 χ g for 15 min and harvest the with 25 mM HEPES, 2.5 mg/ml glucose, 1% bovine serum
cells in the interface. albumin, 50 pg/ml gentamicin, and 1 mM EDTA. Discard the cells
harvested after the first 5 min. Harvest cells after 25 min incubation
and wash several times to remove EDTA. Resuspend the cell pellet
200
Chapter 43 Cell-Culture Methods

in 30% Percoll® (GE Healthcare), layer the cell suspension on top used extensively in mammalian cell cultures but not in avian cell
of 60% Percoll®, and centrifuge for 20 min at 500 x g. Harvest the cultures, with a few exceptions. The QT-35 cell line was established
epithelial cells from the top layer, wash several times, and from a methylcholanthrene-induced fibrosarcoma in a Japanese
resuspend in growth medium. The growth medium consists of either quail (10), but this cell lines is latently infected with Marek’s
CMRL-1066 or minimal essential medium supplemented with 25 disease virus (MDV) (13). Several other quail cell lines free of
mM HEPES, 5% FBS, 5% chicken serum, antibiotics, 2 mg contaminating viruses are available (9). The virus-free chick
glucose/ml, 38 pg/ml ascorbic acid, 1.5 pg/ml transferrin, 4 mM L- fibroblast cell line CHCC-OU2 has recently been used for the
glutamine, 10‘8M Na selenite, 20 ng/ml epidermal growth factor, 5 cultivation of MDV (1). Apparently, MDV can remain latent in
pg/ml pentagastrin, IO'8 M deoxycholic acid, 0.2 pM progesterone, these cells if cells are not allowed to reach confluency. Several
25 ng/ml triiodothyronine, 10 pg/ml insulin, and 5 pM putrescine other fibroblast cell lines have been reported, but these are probably
(pH 6.9). Optimal results are obtained by plating the epidermal cells positive for avian retroviruses. Many lymphoblastoid cell lines have
on feeder layers of primary fibroblasts when they reach 40-50% been developed from tumors induced by MDV, avian leukosis
confluency. virus, and reticuloendotheliosis virus.
Primary CEFs and some other primary cell cultures can be
GROWTH AND MAINTENANCE OF CELL CULTURES expanded by making a limited number of passages without
establishing a cell line. Large batches of primary CEFs can be
Dispersed cells prepared by the above procedures can be grown in harvested at 48 hr (see below) and stored in liquid nitrogen (see
many different kinds of containers, depending on the main below) for future use. This technique has several advantages over
objectives. Cultures are usually grown in glass or plastic bottles, frequent preparation of primary cells. SPF eggs and the preparation
petri dishes or cluster plates containing 4-96 wells. The desired of primary cells are expensive. Batches of frozen cells can be
number of cells depends on several factors, including: titrated for optimum growth, which facilitates the use of CEF at a
1) The type of cell and passage level of the cells. In general, moment’s notice.
primary cultures are seeded with more cells per milliliter than In general, CEF cannot be subcultured satisfactorily for more than
secondary cultures. Established cell lines can often be set up at 5 x four passages. However, the addition of 1% chicken serum to the
104 to 1 x 105 cells/ml. growth medium and incubation at 41 C will facilitate additional
2) The use of the cell cultures. Certain viruses will replicate better passages.
in actively dividing cells, whereas other viruses can replicate in
stationary cultures. The optimum level may vary among different Passage of Cell Cultures
laboratories and is often determined by trial and error. Containers Use the following procedure to prepare secondary cultures or to
that are open to the atmosphere (e.g., petri dishes, cluster plates, and passage a cell line:
flasks or tubes with loose caps) should be incubated at 37.5-39 C in 1) Discard the supernatant medium.
an atmosphere of 3-5% CO2 and 80-85% humidity to avoid loss of 2) Wash the cells briefly with PBS (which must be free of Ca and
CO2, rendering the cultures alkaline, and to prevent evaporation of Mg if versene is to be used as dispersant) to remove residual serum
water from the media. Cultures in sealed tubes do not require CO2 (which would inactivate trypsin) and residual bivalent ions (which
or a humidified incubator. If the number of cells and culture would inactivate versene). Sometimes, this is followed by a brief
conditions are correct, a full monolayer should form in 18-24 hr for wash with TV solution.
fibroblastic cells and 2-3 days for epithelial cells. 3) Add just enough TV solution to cover the culture, and incubate
Monolayers on Coverslips at room temperature or 37 C until the cells round up and start
Cells can be grown on glass coverslips placed in petri dishes, or in detaching from the surface. Initially, the time taken for the cells to
Leighton tubes. Coverslips must be prepared as described for other round up is determined by frequent microscopic examination,
glassware. Place sterile coverslips in the dishes or tubes, and add although with experience the microscopic examination is not
the cell suspensions on top of the coverslips. Make sure that the necessary.
coverslips are on the bottom of the dish; air bubbles can be removed 4) Shake or pipette the cells loose, and place the cells and TV
gently with the tip of a pipette. Other procedures are as described solution into a centrifuge tube with 5% serum. Keep the tube on ice
above. Coverslips can be harvested aseptically after a suitable until centrifugation. Rinse the vessels with PBS to collect additional
period. Use a soft (0.5%) agar medium if monolayers are to be cells. The optional step of washing with PBS is important if all cells
overlaid with an agar-containing medium, so that the monolayer is must be harvested.
not pulled off the coverslip during harvesting. Alternatively, 5) Centrifuge at 350 x g for 15 min, discard the supernatant, and
Chamber-slide™ (Lab-Tek®, Nalge Nunc International, Naperville, resuspend cells in growth medium.
IL) can be used for this purpose. 6) If desired, count the cells as described for primary cultures.
7) Dilute the cells, if necessary, and seed them in new culture
Maintenance of Cell Cultures vessels. In general, a primary culture will yield two secondary
Replace the growth medium with a medium with a lower cultures of the same size, whereas established cell lines can be
percentage of serum (maintenance medium) when the cultures have divided into five or more subcultures.
become confluent. Replace the maintenance medium every 2-3 In general, lymphoblastoid cell lines do not attach to the culture
days, depending on pH changes. An agar-overlay medium is often vessels and can be simply subcultured by counting and readjustment
used to maintain CK cells (Table 43.3), although CK cell cultures to 0.5 x 106 cells/ml. Centrifugation over Ficoll-Paque (15 min, at
are routinely maintained for 6-8 days in Fl0-199 medium (Table 400 x g) is occasionally needed to remove dead cells.
44.1) supplemented with 0.2% FBS. If an agar-overlay medium is Freezing of Cells Many cell lines, as well as primary and
used, it is important to add a small amount of fresh agar medium secondary cells, can be frozen for later use. The only equipment
every 2-3 days to prevent the agar from drying out. needed, in addition to that found in most laboratories, is a suitable
low-temperature freezer. Cells can be stored indefinitely in liquid
Cell Lines and Secondary Cultures nitrogen (-196 C) or for weeks to months at -70 C, although
A cell line is a population of cells derived from animal tissue and viability does decline. Actively growing cells between 2-5 days
grown in vitro for at least 6 mo and more than 50 successive after plating will give the best results, although others can be used,
passages. A cloned cell line is a cell line derived from one single including cells dispersed directly from organs and blood cells. The
cell containing only one nucleus. Cell lines and cloned cell lines are procedure is as follows:
201
Karel A Schat and Holly S. Sellers

1) Prepare the cells as outlined in the section on passage of cell 7) Serologic procedures that detect viral antigens in or on cells,
cultures. such as immunofluorescence, enzyme-linked immunosorbent assay,
2) Centrifuge the cells at 350 x g for 10 min, and discard the or complement fixation.
supernatant fluid. 8) Assay of cell-culture materials in another host system.
3) Resuspend the cells in cold freeze medium at a concentration of 9) Detection of viral DNA or RNA by Southern or northern
up to 5 x 107 cells/ml. Freeze medium consists of growth medium hybridization or by PCR or reverse transcriptase-PCR.
(e.g. M20 [Table 43.1] supplemented with 7.5% dimethylsulfoxide See Figure 43.1 for pictures of uninfected cell monolayers and
and 10-15% serum. various CPE.
4) Fill the storage vials and seal without delay.
5) Place the vials in an insulated container in the vapor phase of a Virus Isolation
liquid nitrogen container for a minimum of 45 min or in a freezer at Procedures will depend greatly on the virus suspected to be present
-70 C for 3-24 hr. Optimum freezing requires a gradual lowering of and the type of sample used; however, the following procedures are
the temperature at about 1 C/min. Programmable freezers are generally suitable:
available for this purpose, but they are expensive and cumbersome 1) Collect organs or tissues such as lung, liver, and brain
for freezing small batches of cells. aseptically, cut them into small fragments, and homogenize by
6) Transfer the vials into the liquid phase of the liquid nitrogen mechanical means. Prepare a 10% (w/v) suspension using PBS or
container. BSS with 5x antibiotics in cases of heavy contamination. Swabs
Insulated containers can be made of cardboard, plastic foam, should be placed in tubes containing BSS or growth medium with
wood, metal, or pipette canisters. They must contain enough antibiotics. For shipment, samples should be properly packaged to
insulation to allow the gradual decrease in temperature (e.g., when prevent leakage (consult shippers for current regulations) and
vials with distilled water at room temperature are placed in the shipped frozen. If cell-associated viruses are suspected, heparinized
container, the water will take at least 30 min to freeze after the blood or single cell suspensions need to be obtained. Cell
container is placed at -70 C). suspensions should be prepared from organs without disrupting the
Just before use, cells should be removed from the liquid nitrogen cells using a bolted nylon screen, or in a Tenbroeck homogenizer
storage and placed into water at room temperature. It is advisable to (Fisher Scientific, Pittsburgh, PA) on ice, or by passing minced
add 10% (v/v) of a disinfectant such as 5.25% sodium hypochlorite pieces through a syringe with a 18-gauge needle. If necessary,
(e.g., Clorox, Clorox Company, Oakland, CA) to the water, because freeze cell suspensions as outlined in the section on freezing of cells
viruses can be present in the liquid nitrogen as a result of broken and ship on dry ice.
ampoules. It is important to use a facemask and heavy gloves while 2) Use cell-free suspensions directly as inocula, or freeze and thaw
removing the vial from the liquid nitrogen for protection from glass several times, or treat with ultrasonic vibration to liberate the
fragments should the ampoule explode. The ampoule should be highest amount of virus. The suspensions can be clarified by
agitated constantly until the last piece of ice thaws, and then the centrifugation for 15 min at 1000 x g. Blood or cell suspensions
contents should be immediately removed aseptically and diluted should not be frozen and thawed or centrifuged.
slowly in growth medium. Typically, 1 ml of cell suspension from 3) For cell-free inocula, remove the growth medium from the
the ampoule can be diluted in 10 ml of medium and then further cultures and add a part of the supernatant fluid from step 2 above.
diluted to the final concentration based on the number of viable Sufficient fluid should be present to just cover the monolayer. It is
cells or prior titration of the batch of frozen cells. advisable to inoculate undiluted material as well as 10- and 100-fold
dilutions of the material, because toxic materials in undiluted
APPLICATION OF CELL-CULTURE TECHNIQUES IN samples often cause nonspecific damage to the cells. Change the
VIROLOGY culture medium to maintenance medium when cellular inocula are
used. Add the cells directly to the cell cultures. The virus-containing
The most important factors to be considered in studies involving inoculum cells will settle on the monolayer.
viruses are a cell-culture method susceptible to virus infection, 4) Incubate cultures for 1 hr at 38-39 C.
proper growth medium free of antibodies and nonspecific virus or 5) Wash the monolayer twice with abundant BSS to eliminate
cell inhibitors, and environmental conditions best suited to maintain excess contamination and toxic materials from the sample
the cells during the entire period of the experiment. Virus (optional). If this step proves necessary, wash the monolayer
multiplication can be detected in the following ways: carefully so as not to dislodge cells that are loosely attached. Do not
1) Morphologic alteration of the cells, called cytopathic effect wash the monolayer when a cellular inoculum is used.
(CPE), caused by degenerative changes, or, as with some tumor 6) Add fresh maintenance medium and incubate at 38-39 C.
viruses, transformation. Under suitable culture conditions in which 7) Inspect the infected monolayer and controls daily for at least 7
agar overlays and low doses of virus are used, CPE can be seen as days. If cell degeneration is not seen or is unclear, collect the cells
plaques and transformation as foci. Plaques can be made visible by and the fluids from the containers and make a blind passage onto a
adding 1.0 ml of neutral red (1%) to 100 ml of agar-overlay fresh monolayer. Sometimes two or three blind passages must be
medium. Alternately, the neutral red may be added with the feed made to obtain evidence of CPEs. Some viruses do not produce
agar at a later time. Use 2.0 ml of 1% neutral red per 100 ml of feed CPE in culture and must be examined for other changes. For stocks
agar. Keep the cultures in the dark after the addition of neutral red. of virus, supernatant fluid and cells should usually be harvested
Live cells will stain with neutral red, whereas dead cells will be when CPEs are at a maximum but before all cells die. The
visible as clear plaques. suspension can be frozen and thawed or treated with ultrasonic
2) The formation of giant cells and syncytia. vibration, clarified by low-speed centrifugation, and stored frozen.
3) The appearance of cytoplasmic or nuclear inclusion bodies Cell-associated viruses are frozen as outlined in the section on
shown by using polychrome dyes. freezing of cells and stored in liquid nitrogen. It is advisable to
4) The induction of pH changes in the medium due to changes in harvest the cell-associated virus before maximum CPE, when the
cell metabolism. cells have a better viability.
5) Changes in the hemadsorption properties of the cell membrane.
6) The ability of some noncytopathogenic viruses to interfere with
or enhance the CPEs of other viruses.
202
Chapter 43 Cell-Culture Methods

ACKNOWLEDGEMENT 7. Huang, D. D. Restriction of avian reovirus in primary chicken embryo


tendon cells. Virology 207:117-126. 1995.
The authors are greatly indebted to H. Graham Purchase for his 8. Hudis, M Plasma treatment of solid materials. In: J. R. Hollahan, and A.
contributions to earlier editions of this chapter. T. Bell, Eds. Techniques and applications of plasma chemistry. John Wiley
and Sons, New York. Pp. 113 - 147. 1974.
REFERENCES 9. Li, X., and K. A. Schat. Quail cell lines supporting replication of Marek’s
disease virus serotype 1 and 2 and herpesvirus of turkeys. Avian Dis.
1. Abajoub, A., and P. M Coussens. Development of a sustainable chick 48:803-812. 2004.
cell line infected with Marek’s disease virus. Virology 214:541-549. 1995. 10 Moscovici, C., M G. Moscovici, H. Jiminez, Μ M C. Lai, M J.
2. Ali, A., and D. L. Reynolds. Primary cell culture of turkey intestinal Hayman, and P. K. Vogt. Continuous tissue culture cell lines derived from
epthelial cells. Avian Dis. 40:103-108. 1996. chemically induced tumours of Japanese quail. Cell 11:95-103. 1977.
3. Amstein, C. F., and P. A. Hartman. Adaption of plastic surfaces for tissue 11. Ramsey, W. S., W. Hertl, E. D. Nowlan, and N. J. Binkowski. Surface
culture by glow discharge. J. Clin. Microbiol. 2:46 -54. 1975. treatments and cell attachment. In Vitro 20:802-808. 1984.
4. Augustine, P. C. Establishment of a turkey cecal cell line and 12. Versteeg, J. Color atlas of virology. Year Book Medical Publications
development of turkey coccidia within the cells. Proc. Soc. Exp. Biol. Med. Ltd., London, UK 1985.
206:152-156. 1994. 13. Yamaguchi, T., S. L. Kaplan, P. Wakenell, and K. A. Schat.
5. Curtis, A. S. G., J. V. Forrester, C. Mclnnes, and F. Lawrie. Adhesion of Transactivation of latent Marek’s disease herpesvirus genes in QT35, a quail
cells to polystyrene surfaces. J. Cell. Biol. 97:1500-1506. 1983. fibroblast cell line, by herpesvirus of turkeys. J. Virol. 74:10176-10186.
6. Freshney, R. A. Culture of animal cells. A manual of basic technique, 4th 2000.
ed. John Wiley and Sons, Inc. New York, 2000.

203
I)

E F
20\ *


*><
f i

• ♦ • ***
* 4 ' ·
« ·
*
' · r
Λ
gure 43.1
A. Uninfected primary chicken embryo liver cells at 48 hours post seeding.
B. Reovirus cytopathic effect in primary chicken embryo liver cells at 48 hours post-inoculation.
C. Adenovirus cytopathic effect in primary chicken embryo liver cells at 48 hours post-inoculation.
D. Infectious laryngotracheitis virus cytopathic effect in primary chicken embryo liver cells at 72 hours post-inc
E. Uninfected Vero cells
F. Avian metapneumovirus (aMPV) subtype C-infected Vero cells stained with an aMPV specific monoclonal z
courtesy of D.R. Kapczynski, USDA/ARS/SEPRL).
44
VIRUS PROPAGATION IN EMBRYONATING EGGS
Dennis A. Senne

SUMMARY. The embryonating egg is one of the most versatile and widely used host systems for the isolation and propagation of
several avian viruses. Many variables can affect the success, or failure, of the embryonating egg to support virus growth. Some variables
that are covered in this chapter include the source and species of fertile eggs, age of the embryo, route of inoculation, and conditions for
incubation. This chapter also provides detailed accounts on sample preparation: inoculation of eggs via the allantoic sac, yolk sac,
chorioallantoic membrane, and amniotic sac routes: collection of specimens from inoculated embryos; and methods for serial egg passage.

INTRODUCTION SOURCE OF EGGS AND PRELIMINARY INCUBATION


PROCEDURES
The embryonating chicken egg has long been one of the most
widely used host systems for the isolation, propagation, and Fertile eggs less than 1 wk old should be obtained from vigorous,
characterization of avian viruses and for the production of viral healthy, specific-pathogen-free (SPF) breeder flocks that are
vaccines. The embryo and its supporting membranes provide the regularly monitored for most common avian viral and bacterial
diversity of cell types necessary to culture many different types of pathogens (Sources: Hy-Vac, Adel, Iowa; Charles River-SPAFAS,
viruses. However, the success or failure of this system for Norwich, CT; Sunrise Farms, Catskills, NY ). Most suppliers will
propagating and isolating viruses depends upon several conditions provide testing summaries upon request. The validity of any
(1): route of inoculation, age of embryo, incubation temperature, isolation may be compromised by the presence of egg-transmitted
length of incubation time following inoculation, volume and agents such as avian adenovirus, avian encephalomyelitis virus,
dilution of the inoculum used, and the immune status of the flock Newcastle disease virus, avian leukosis virus, infectious bronchitis
from which the eggs were obtained. To optimize the conditions for virus, infectious bursal disease virus, avian reovirus, chicken
virus isolation and propagation in embryonating eggs, each of these anemia virus. Mycoplasma, and Salmonella (1). Embryonating eggs
parameters should be considered for the particular virus in question. from other species, (e. g., quail, duck, and turkey) may also be used
As with any system used for virus isolation, detection of virus (1). However, some adjustments in the age at which the embryos
infection must be possible. The growth of viruses in the are inoculated may be necessary to allow for differences in
embryonating chicken egg can be detected, in most cases, by one or embryonic development. Duck eggs, for example, require 28 days
more of the following (3): embryo death: lesions in the of incubation to hatch, compared to 21 days for chicken eggs.
chorioallantoic membrane (CAM) such as edema or the formation Antibodies may also be found in the yolk of eggs laid by
of plaques; lesions in the embryo such as stunting, cutaneous specifically immune hens. Eggs from antibody-positive hens may
hemorrhage, and abnormal development of muscles and feathers; be used for virus isolation or propagation if routes of inoculation
and abnormalities in the Visceral organs, including enlargement of other than the yolk sac route are employed and the isolation
the liver and spleen, greenish discoloration of the liver, necrotic foci procedure will be completed before the embryo reaches the 15th
on the liver or heart, and formation of urate deposits in the day of incubation, the time when the embryo begins to absorb
mesonephros. Special care should be taken to differentiate lesions antibodies from the yolk. Antibody-positive eggs should be used
that may be caused by the presence of bacterial agents. only when a satisfactory alternate source is not available.
More direct and definitive method for detection of virus infection When working with some viruses, such as avian leukosis, the
of chicken embryos include: ability of the allantoic amniotic fluid genetic constitution of the breeder flock may play an important role
(AAF) to cause hemagglutination (HA) of washed chicken in determining the suitability of some eggs used for virus isolation
erythrocytes, use of serologic and molecular techniques, such as or propagation; some breeds of chickens are naturally resistant to
immunoassays cDNA probes, polymerase chain reaction (PCR), certain strains of leukosis virus.
and electron microscopy (1,3). Initially, eggs should be incubated in a suitable incubator at a
temperature of 38-39 C and relative humidity of 60-70%. If
STRUCTURE OF THE EMBRYONATING EGG possible, the eggs should be turned several times daily to promote
proper development and prevent adhesions of embryonic
Directly beneath the shell of the egg lies a tough fibrinous eggshell membranes (2). Eggs are usually incubated 6-11 days before being
membrane. This membrane lines the entire inner surface of the egg, inoculated; the length of incubation is determined by the route of
and forms the air cell at the blunt end of the egg (Fig. 44.1). The inoculation to be used.
eggshell membrane, along with the eggshell, helps maintain the Eggs should be candled after 5-6 days of incubation to determine
microbiological integrity of the egg, while allowing diffusion of which are fertile and which are unfertile or not viable. Eggs with
gases in and out of the egg. The distribution of gases within the egg weak embryos or abnormal air cell size or position should be
is facilitated by the formation of the highly vascular CAM. which discarded. Candling is performed in a darkened room using a
serves as the respiratory organ of the embryo. The formation of this universal microscope illuminator or other suitable candling device.
membrane occurs adjacent to the shell membrane throughout the The portable microscope illuminator permits the eggs to be candled
egg. During development, this membrane forms a relatively large while positioned on the egg flat, thus eliminating the need for
cavity known as the allantoic sac, which contains between 5 and 10 handling each egg.
ml of allantoic fluid at 10-14 days of incubation (Fig. 44.1). The
embryo is directly surrounded by the amniotic membrane forming SAMPLE PREPARATION
the amniotic sac, which contains 1-2 ml of amniotic fluid. The
embryo is attached to the yolk sac, which is located approximately The type of specimens submitted for virus isolation will differ
in the center of the egg and supplies the nutrients necessary for depending on the disease suspected but will usually consist of
embryo development (2). tissues, feces, or swabs. Suspensions of these specimens should be

204
Chapter 44 Virus Propagation in Embryonating Eggs

Fig. 44. 1^14.6. Inoculation routes for embryonating eggs.

Fig. 1. Structure of the embryonating


egg (After Hawkes [2]).

Fig. 4. Yolk sac route of


inoculation (After Hawkes [2]).

Fig. 2. Allantoic route of inoculation, Fig. 5. Chorioallantoic membrane


Method A (After Hawkes [2]). (CAM) route of inoculation
(After Hawkes [2]).

Fig. 3. Allantoic route of inoculation, Fig. 6. Amniotic route of inoculation


Method δ (After Hawkes [2]). (After Hawkes [2]).

205
Dennis A. Senne

prepared in Tris-buffered fryptose broth (1. 21 g Tris per liter of Allantoic Sac Inoculations
tryptose broth), nutrient broth, brain-heart infusion broth, or other Method A. The following procedure is the most commonly used.
suitable medium containing antibiotics. The concentration of 1) Candle 9 to 11-day-old embryonating eggs and mark the position
antibiotics used may depend on the type of sample being processed of the air cell.
but could contain, as an upper limit, penicillin (10,000 IU/ml), 2) Place eggs on an egg flat, air cell up, and disinfect the area
streptomycin sulfate (2. 0 mg/ml), gentamicin sulfate (1.0 mg/ml), marked in step 1 above. Drill a small hole through the eggshell just
kanamycin sulfate (0. 65 mg/ml), and amphotericin B (0. 02 above the marked location of the air cell (Fig. 43. 3).
mg/ml). The antibiotics may vary with local conditions and 3) Using a syringe fitted with a 25-gauge 5/8-inch (16-mm) needle,
applications. It is important that the pH of the antibiotic diluent be inoculate 0. 1-0. 3 ml of inoculum per egg by inserting the needle,
adjusted to a range of 7. 0-7. 4. For convenience, a stock solution of to its full length, vertically through the hole or at a slight angle
concentrated antibiotics, adjusted to the appropriate pH for the away from the center of the egg and injecting the desired amount.
diluent being used, can be prepared in advance and stored at 20 C. 4) Seal hole and return the eggs to the incubator. Eggs inoculated by
Tissues can be prepared as 10%—15% suspensions by grinding the allantoic route are normally incubated 3-7 days postinoculation.
with a mortar and pestle, Tenbrock tissue homogenizer, electric Method B. The following procedure may also be used.
homogenizer, or stomacher. Tissue suspensions should be prepared 1) Candle 9 to 11 day-old embryonating eggs, making sure that all
in a biosafety cabinet. Swabs should be diluted in as small a volume air cells are in the normal position.
as required for testing (usually 3-4 ml), then vortexed to expel as 2) Place eggs on an egg flat, air cell up, and disinfect the area
much sample material as possible from the swab fibers. Swabs and directly at the top of the egg (the area over the air cell). Drill a small
tissue suspensions should be centrifuged at 1000-1500 χ g for 20 hole through the eggshell along the center axis at the top of the egg.
min in a refrigerated centrifuge (4-10 C) to sediment tissue debris 3) Using a syringe fitted with a 25-gauge 5/8-inch (16-mm) needle,
and most bacteria. The supernatant should then be aseptically inoculate 0. 1-0. 3 ml of inoculum per egg by inserting the needle
removed and placed in an appropriately sized vial for egg vertically through the hole the entire length of the needle and
inoculation and storage. Specimens should be kept at room injecting the desired amount (Fig. 44.2). Avoid moving the syringe
temperature with antibiotics for 1-2 hr before being inoculated into sideways once the needle is inserted to prevent tearing of the CAM,
eggs to reduce problems with bacteria. which could cause bleeding and death of the embryo.
Specimens heavily contaminated with bacteria that cannot be 4) Seal hole and return the eggs to the incubator. Eggs inoculated
removed by centrifugation or controlled with antibiotics can be by the allantoic route are normally incubated 3-7 days
filtered through a sterile 450-nm membrane filter. Filtering should postinoculation.
be used only as a last resort because aggregates of virus could be
retained by the filter and reduce the chance for successful isolation. Yolk Sac Inoculation
Following inoculation, specimens should be stored at -70 C for 1) Candle 6-to-8-day-old embryonating eggs and mark the center of
future use. the air cell.
2) Place eggs on an egg flat, air cell up, and disinfect the top of the
ROUTES OF INOCULATION egg (the area over the air cell).
3) Drill a small hole through the eggshell along the center axis at
The four most common routes for the inoculation of embryonating the top of the egg (Fig. 44.4).
eggs are via the allantoic sac, yolk sac, CAM, and amniotic sac. In 4) Using a syringe fitted with a 22-gauge 1-inch needle inoculate 0.
diagnostic situations where no particular agent is suspected, it is 1-0. 5 ml of inoculum per egg by inserting the needle vertically to
advisable to use as many routes as possible. The CAM route is its full length and injecting the desired amount. To verify the
preferred because of its sensitivity to a large number of viruses and position of the needle as being in the yolk sac, gently withdraw the
because it is less likely to be affected by bacterial contamination plunger of the syringe; yolk will be aspirated if the needle has
(3). Yolk and allantoic sac routes commonly are used and have entered the yolk sac.
similar sensitivities for isolation of many agents. For initial passage, 5) Seal hole and return the eggs to the incubator. Eggs inoculated by
a minimum of four embryos should be used for each specimen and this route are generally incubated from 3 to 10 days, or in some
each route of inoculation. instances, until the eggs hatch.
Before inoculation, all embryos should be candled for viability,
and the site of inoculation marked and disinfected with a solution of Chorioallantoic Membrane Inoculation
70% ethyl alcohol (ETOH) containing 3.5% iodine and 1.5% 1) Candle 10 to 11-day-old embryonating eggs and mark the side of
sodium iodide. The disinfectant can be applied with a cotton swab the egg approximately midway along the long axis where the vein
or with a sponge that has been cut to fit a plastic screw cap structure is well developed.
(approximately 40 mm in diameter). A hole is made in the shell 2) Place eggs horizontally on an egg flat and disinfect both the air
using a vibrating engraving tool or rotating drill equipped with a cell end and the side of the egg that has been marked in step 1
pointed tip. The drill tip should be disinfected with ETOH-iodine above (Fig. 44.5).
disinfectant before each use to avoid contaminating the inoculation 3) Drill a small hole through the shell and eggshell membrane at the
site. Following inoculation, the hole is sealed with type Π (for center of the air cell. Drill a second hole on the side of the egg,
plastics) glue or melted paraffin. The eggs are then returned to the being careful not to perforate the eggshell membrane.
incubator. If the inoculated eggs are to be incubated to allow the 4) At this point eggs should be taken into a darkened room to
chicks to hatch, the eggs should be turned several times daily to perform and observe the dropping of the CAM. Supplies needed
promote proper embryo development. However, continued turning include a 1-ml tuberculin syringe containing sterile phosphate-
of the eggs is generally not required for most routine virus isolation buffered saline (PBS) and fitted with a 25-gauge needle 5/8-inch
procedures. The procedures described below are similar to (16-mm), and a piece of rubber tubing attached to a vacuum source
previously described procedures (1,2,3). or suction bulb. While holding the egg against the egg candler apply
gentle vacuum to the hole at the air cell. At the hole on the side of
the egg insert the point of a 25-gauge needle just under the shell at a
very shallow angle, perforating the eggshell membrane but not the
CAM. Withdrawing the air from the air cell will cause the CAM to
drop, thus forming a new false air cell directly over the CAM.
206
Chapter 44 Virus Propagation in Embryonating Eggs

Deposition of a very small drop of sterile saline on the hole created for discarding eggs, a container for discarded pipettes, and a pipette­
on the side of the shell is sometimes helpful in facilitating the filling device if pipettes are to be used.
dropping process. The PBS acts like a lubricant to help separate the All eggs from which specimens are to be collected should be
CAM from the eggshell membrane. Once the CAM is dropped seal surface-disinfected with 70% ETOH or other suitable disinfectant.
the hole at the air cell and keep eggs positioned horizontally. The alcohol can be easily and rapidly applied using a gun-type
5) Using a syringe fitted with a 25-gauge 5/8-inch (16-mm) needle mist/spray bottle. The disinfectant should be allowed to dry before
inoculate 0. 1-0. 3 ml of inoculum per egg by inserting the needle harvesting specimens.
vertically just inside the eggshell and injecting the desired amount.
6) Seal the hole, rock the egg gently to distribute the inoculum Collection of AAF
evenly over the CAM surface, and return eggs to the incubator. From Eggs with Dead Embryos.
Eggs should be incubated horizontally throughout the incubation 1) Crack the shell over the air cell by tapping with the blunt end of a
period to prevent the false air cell from shifting, as this may cause sterile forceps. Remove the portion of the shell that forms the air
excessive nonspecific mortality. Eggs inoculated by this route are cell and discard pieces in disinfectant. Discard the forceps in a
usually incubated for 5-7 days. beaker of disinfectant.
2) Use a different forceps to tear the eggshell membrane, the CAM,
Amniotic Sac Inoculation and the amniotic membrane to release the AAF. Depress the
1) Candle 10 to 11-day-old embryonating eggs and mark the general membranes over the yolk sac with the forceps and allow the AAF to
location of the embryo at the base of the air cell. collect and pool above the forceps. Using a 5-ml pipette or syringe
2) Position eggs air cell up and disinfect the area directly at the top and needle, aspirate the fluid and place into a sterile 12 * 75-mm
of the egg. Drill a small hole through the eggshell along the snap-cap tube or other suitable vial. Note: amniotic fluid and
longitudinal axis at the top of the egg. (Fig. 44.6). allantoic fluid can be harvested separately using a syringe and
3) Take eggs to a darkened room because this procedure requires needle and aspirating the fluid from each cavity before the
illumination of the egg with an egg candler while being inoculated. membranes are tom. Carefully peeling back the eggshell membrane
Use a syringe with a 22-gauge P/z-inch (38-mm) needle and aim the from the CAM may be necessary to permit a better view of the
needle toward the shadow of the embryo. When the end of the membranes.
needle approaches the amniotic sac, give a gentle but quick stab 3) Culture a loopful of the AAF for bacteria using blood agar or
toward the embryo to permit the needle to penetrate the amniotic nutrient agar. Incubate plates at 37 C overnight and record results.
membrane, then inject 0. 1-0. 2 ml of inoculum. To verify that the 4) Clarify the AAF by centrifugation at 1500 x g for 10 min and test
needle is in the amniotic sac, carefully move the needle sideways; if the AAF for HA activity using a standard microtiter procedure for
the needles have entered the amniotic sac, the embryo should reflect Newcastle disease virus or avian influenza virus.
the same movement as the tip of the needle. 5) Store AAF at 70 C for passage or other use.
4) Seal the hole and return the eggs to the incubator. Eggs 6) This method can also be used to collect AAF from eggs with
inoculated by this route are generally incubated from 2 to 4 days at embryos surviving the incubation period; eggs should be
a temperature appropriate for the virus being propagated. refrigerated to kill the embryo before collecting the AAF.

COLLECTION OF SPECIMENS FROM EMBRYONATING From Eggs with Live Embryos.


EGGS Allantoic-amniotic fluid can be sampled from eggs with five
embryos 24-72 hr post inoculation without taking the eggs off test.
Inoculated eggs should be candled at least once a day to identify This would be important in situations where a diagnosis is urgently
eggs with dead embryos. Such eggs are removed from the needed. If done carefully, eggs can be returned to the incubator for
incubator. Continued incubation of eggs with dead embryos may continued incubation. Use the following procedure.
result in decreased virus titer due to thermal inactivation and could 1) Drill a hole through the shell at the top of the air cell. Using a
cause changes in the embryonic tissues making it difficult to syringe fitted with a 22-gauge V/2-inch (38-mm) needle, insert the
interpret lesions in the embryo or on the supporting membranes (1). needle toward the shell at an approximate 45- to 60-degree angle
Embryos that die within the first 24 hr should be discarded as from the vertical axis.
nonspecific deaths caused by injury or bacterial contamination. All 2) Aspirate 0. 1-0. 5 ml of AAF and evaluate for evidence of virus
embryo deaths beyond 24 hr should be considered suspect. If the infection using HA, electron microscopy, or other appropriate
inoculated eggs have live embryos following the specified methods.
incubation period, eggs should be refrigerated for a minimum of 4 3) Seal hole and return to incubator.
hr (preferably overnight) to kill the embryo and allow the blood to 4) This technique can also be use to collect AAF from eggs with
clot before harvesting egg contents. The presence of erythrocytes in embryos surviving the incubation period; eggs should be
the allantoic or amniotic fluids can significantly reduce the titer of refrigerated to kill to embryo before collecting the AAF.
some viruses, such as paramyxoviruses and orthomyxoviruses,
which agglutinate erythrocytes. Harvesting the CAM
When working with virus-infected embryonating eggs major Method A. The following procedure is most commonly used.
consideration should be given to procedures used to collect 1) Crack the eggshell over the false air cell by tapping the eggshell
specimens to prevent cross-contamination of samples or with the blunt end of sterile forceps. Remove the eggshell as close
contamination of the laboratory environment. It is recommended to the edge of the false air cell as possible and discard pieces of
that all procedures for harvesting or working with infectious eggshell in disinfectant. Discard the forceps in a beaker of
embryonic tissues and fluids be performed in a class Π (or higher) disinfectant.
microbiological safety cabinet. 2) Observe the CAM for signs of thickening (edema) and plaque
Equipment and supplies needed for harvesting include two 250-ml formation.
beakers filled with disinfectant (one for eggshells and one for used 3) Harvest the CAM by grasping it with sterile forceps, stripping
forceps), sterile curve tipped forceps, sterile 5-ml pipettes or 5 10- away excess fluids with a second set of forceps. Place harvested
ml syringes with l/2-inch (38-mm) 18 or 20-gauge needles for CAM in a sterile petri plate for further examination or in a 12 x 75-
harvesting egg fluids, plastic 12 * 75-mm sterile snap-cap tubes or mm snap-cap tube or other suitable vial for storage.
other suitable vials, a large container double-lined with plastic bags
207
Dennis A. Senne

4) For storage, place CAM in a sterile 12 x 75-mm snap-cap tube or addition, waiting until the inoculated embryo has hatched and the·
other suitable storage vial and freeze at 70 C. collecting specimens from the newly hatched bird is sometimes
Method B. The following procedure may also be used. advantageous. Tissues and organs from embryos and birds should
1) Open egg the same way as for harvesting AAF, as described be collected aseptically using standard necropsy procedures and
above. assayed for evidence of virus infection using appropriate methods.
2) Using sterile forceps, carefully remove the embryo and yolk sac
being careful not to dislodge the CAM from the eggshell. Passage of Egg Material
3) Pour out remaining fluids. In most instances where a virus is not detected on the first passage,
4) Observe the CAM for lesions before removing it from the additional blind passages may be required. If AAF is used as the
eggshell or, for closer examination, remove the CAM from the inoculum for passage, it should be centrifuged for 10-20 min at
eggshell and place in a sterile petri plate and look for thickening of 1500 x g and diluted 1:10 or greater in antibiotic diluent prior to
the membrane and/or plaque formation. inoculation. The CAM, yolk sac, and embryo/bird tissues and
5) For storage, place the CAM in a sterile 12 x 75-mm snap-cap organs should be prepared as a 10% suspension in antibiotic diluent
tube or other suitable storage vial and freeze at 70 C. and centrifuged at 1500 x g for 20 min prior to inoculation.

Harvesting the Yolk Sac Membrane REFERENCES


1) Open the egg in the same way as described above for harvesting
AAF. 1. Cottral, G. E. , ed. Manual of standardized methods for veterinary
microbiology. Cornell University Press, Ithaca, N. Y. pp. 47-52. 1978.
2) Rupture the CAM to allow access to the yolk-sac membrane.
2. Hawkes, R. A. General principles underlying laboratory diagnosis of
3) Grasp the yolk sac membrane with forceps and carefully lift it to viral infections. In: Diagnostic procedures for viral, rikettsial and chlamydial
separate it from the embryo and other membranes. Using a second infections, 5th ed E. H. Lennette, and N. J. Schmidt, eds. American
set of forceps, strip off the excess yolk and place the yolk sac in a Public Health Association, Washington, D. C. pp. 1-48. 1979.
sterile 12 x 75-mm snap-cap tube or other suitable vial for storage. 3. Hitchner, S. B. Virus propagation in embryonating eggs. In: Isolation
4) Store the yolk sac at 70 C. and identification of avian pathogens. S. B. Hitchner, C. H. Domermuth,
H. G. Purchase, J. E. Williams, eds. Creative Printing Company, Endwell,
Harvesting Embryo/Bird Tissues or Organs N. Y.pp. 120-121.1980.
Tissues and/or organs from inoculated embryos may be a valuable
source of virus-infected material and should not be overlooked. In
45
VIRUS IDENTIFICATION AND CLASSIFICATION
Pedro Villegas and Ivan Alvarado

Summary. Since the first observation of the presence of a pathogenic agent smaller than any known bacteria causative of tobacco mosaic
disease by Ivanowsky in 1892 and Beijerink in 1898, virus identification and classification has continuously evolved. The continuous
evolution has been achieved by the constant implementation of new technologies that have contributed to new knowledge, allowing the
classification of previously unclassified viruses or the further reclassification of previously classified viruses. In poultry virology, a clear
example of such evolution is the recent inclusion of two new genera (Atadenovirus and Siadenovirus) in the Adenoviridae family, in addition
to the previously present Aviadenovirus genus (17). Also, the avian encephalomyelitis virus, formerly unassigned to a genus in the
Picomaviridae family, has been included in the new Hepatovirus genus within the same family (17).
Avian viruses have been isolated and identified by techniques now referred to as “classical”. Classical viral detection techniques include the
identification of cytophatic effect (CPE) on cell cultures, complement fixation, hemagglutination-inhibition, enzyme immunoassays and the
use of fluorescent antibodies directly on specimen material or after virus propagation. Specific information about viral morphology,
symmetry, size, shape and the presence or absence of envelope has been obtained by transmission electron microscopy. With the availability
of newer molecular techniques, such as polymerase chain reaction (PCR) and reverse transcriptase (RT) - PCR, many viruses can be detected
without previous isolation. Molecular techniques allow a rapid, novel and more sensitive detection and identification of viruses. Other
techniques, such as crystal X-ray technology have increased our understanding about the structural conformation of particular viral proteins,
such as the structural conformation of the VP2 protein of infectious bursal disease virus (24).

INTRODUCTION • Syncitial formation in primary epithelial cell monolayers, and


hemorrhages and congestion in chicken embiyos produced by
Virus isolation and detection avian reovirus. However, syncytial formation might also be
The presence of a virus in a host is initially suspected by the produced by other viruses and should not be used as a single
occurrence of specific clinical signs and characteristic macroscopic criterium to identify reoviruses.
and/or microscopic lesions present in affected organs. Once a • Embryo stunting is normally observed after inoculation of
particular virus is suspected, the virus can be isolated by inoculating infectious bronchitis virus (IBV) and adenoviruses. A more severe
infectious material in susceptible birds or in susceptible systems, stunting, lack of feather development and strong embryo
such as specific pathogen free (SPF) chicken embryos or cell attachment to the chorioallantoic membrane is usually observed
culture (18). Chicken embryos remain as the most convenient when embryo-adapted strains of IBV are isolated.
method for growing high titer stocks of some viruses for research • Formation of plaques in monolayers of primary cells by Marek’s
and diagnostic laboratories, and vaccine production (18). Some disease and other herpesviruses. Once adapted to grow in primary
particular viruses, such as coronavirus, orthomyxovirus, cells, plaques can be observed in chicken fibroblast monolayers.
paramyxovirus, herpesvirus and poxvirus, have been replicated in Once the virus has been isolated, further quantification can be
one or several tissues in the chicken embryo (18). Three main cell achieved by classic biologic or physical assays, as described (18).
culture systems, primary cell culture, cell strains and cell lines, are Biologic assays, such as plaque assay, endpoint method and pock
used for virus isolation. Recognition of viral growth in cell cultures formation, allows the detection of only infective particles in
is achieved by the presence of cytophatic effect, formation of propagation systems. Physical methods, such as optical density
inclusion bodies or hemoadsorption (18). Cytophatic effect can be measurements, immunologic methods or electron microscopy
observed under the light microscope, with the presence of particle counts, quantify the presence of all viral particles without
morphologic changes induced by the virus in individual cells or a differentiating their infectivity capacity.
group of cells. Inclusion bodies are subtle alterations of the With the availability of molecular techniques, such as nucleic acid
intracellular architecture of individual cells. Hemadsorption hybridization, RNA fingerprinting and viral genome amplification
indirectly measures the synthesis of viral protein in infected cells, by PCR or RT-PCR, many viruses can be detected without previous
detected by the adsorption of erythrocytes to the surface of infected isolation (5). Current restrictions and regulations on the importation
cells. Once the virus is isolated, the size, shape, surface structure and use of infectious materials have further increased the use of
and symmetry of the virions can be determined by transmission molecular techniques. Infectious clinical samples, previously
electron microscope after negative staining of concentrated viral inactivated with formalin, phenol or stamped in Flinders
particles with uranyl acetate or phosphotungstic acid (18). Technology Associates (FTA®) cards, can be imported with minor
The following “classical” observations have been used to identify restrictions, allowing the molecular detection and characterization
the presence of particular viruses in biological samples: of avian viruses (19, 26, 27, 29). Although important information
• The ability to spontaneously hemagglutinate chicken red blood about a particular isolate can be obtained by molecular techniques,
cells is used to identify Newcastle disease virus, influenza virus it is important to emphasize that detection of viral nucleic acid is
and egg drop syndrome. Differential characterization of these not equivalent to virus isolation. Only after isolating the virus, can
viruses can be achieved by viral neutralization using specific valuable information such as pathogenicity be obtained, cross­
polyclonal or monoclonal antibodies. neutralization and protection studies, can be performed with an
• The presence of round and refractive cells detaching from liver isolate and it can also be determined whether there are more than
cell monolayers with the presence of intranuclear inclusion bodies one type of virus in clinical samples. Virus isolation in association
by adenovirus strains. with molecular techniques will give a better understanding of both
• Pock formation on the chorioallantoic membrane of embryonating the phenotype and genotype of a particular avian virus.
eggs with the presence of intracytoplasmic inclusion bodies,
characteristic of fowl pox viruses. Virus classification
The classical system of virus classification, developed by Lwoff,
Home and Tournier in 1962, groups the viruses according to viral

209
Pedro Villegas and Ivan Alvarado

shared properties rather than the properties of the cells or organisms • Orientation: Positive, negative, ambisense.
they infect (18). The classical Linnaean hierarchical system has • Number of segments, size of the genome, nucleotide
been partially adopted by the International Committee on sequence.
Taxonomy Viruses (ICTV) (17). The ICTV, a committee of the • Gene organization.
Virology Division of the International Union of Microbiological • Virion morphology: Size, shape, capsid symmetry, and
Societies, classifies the viruses according to their order, family, the presence or absence of envelope.
subfamily, genus and species. Levels higher than the order, such as • Physical properties such as stability against different pH,
class and phylum, and lower than species, such as strains and temperature, detergents, cations and solvents
variants, are not considered in the ICTV classification. In the ICTV • Characteristics of viral proteins, lipids and carbohydrates.
classification, the family is the highest consistently used taxonomic
• Antigenic properties.
grouping, bearing the most generalized description of a particular
• Biologic properties such as replication strategy,
characteristic shared by its members (18).
transmission, pathogenicity and host range.
The main characteristics that delineate the ICTV classification of
The major characteristics used to classify a virus in a particular
viruses are:
family include the nature of the nucleic acid in the virion, the
• Nature of the nucleic acid in the virion.
presence or absence of envelope, the symmetry of the capsid and
• Type of nucleic acid: DNA or RNA, single stranded (ss) the size of the virion and capsid (18). Table 45.1 summarizes the
or double stranded (ds), linear or circular. specific characteristics of each family with important avian viruses.

Table 45.1 Major characteristics for the classification of avian viruses, according to the guidelines established by the International Committee on Taxonomy
Viruses (ICTV),
Nucleic Acid Symmetry Enveloped/Naked Family Diameter (nm)
Retroviridae 80-130
Enveloped Circoviridae 14-24
Togaviridae 70
Icosahedral
Reoviridae 60-80
RNA Naked Birnaviridae 60
Caliciviridae 35-40
Picornaviridae 28-30
Coronaviridae 80-160
Helical Enveloped Orthomyxoviridae 90-120
Paramyxoviridae 150-300
Polyhedral Enveloped Flaviviridae 25-30
Parvoviridae 18-26
Naked
Adenoviridae 70-90
Icosahedral
DNA Polyomaviridae 40-55
Circoviridae 17-24
Enveloped Hepadnaviridae 42
Herpesviridae 150-200
Complex Enveloped Poxviridae 170 to 200-300 to 456

Viruses are divided in two major groups: DNA and RNA containing viruses. DNA containing viruses are divided in 8 families, six of which contain viruses
causing disease in avian species. RNA containing viruses are divided in approximately 38 families, with 11 families containing recognized avian pathogens.
Table 44.2 lists the ICTV classification of DNA and RNA avian viruses (17).

210
Chapter 45 Virus Identification and Classification

Table 45.2. Families of DNA and RNA viruses with important avian viruses

Type Family Genus Avian Viruses

Poxviridae Avipoxvirus Fowlpox virus, canarypox virus, jucopox virus, pigeonpox virus, psitaccinepox virus,
quailpox virus, sparrowpox virus, starlingpox virus, turkeypox virus.
Fowl adenovirus A (FAV-1)
Fowl adenovirus B (FAV-5)
Fowl adenovirus C (FAV-4, FAV-10)
Fowl adenovirus D (FAV-2, FAV-3, FAV-9, FAV-11)
Aviadenovirus Fowl adenovirus E (FAV-6, FAV-7, FAV-8a, FAV-8b)
Goose adenovirus (GAV-1, GAV-2, GAV-3)
Adenoviridae Pigeon adenovirus
Tentative: Duck and turkey adenovirus
Siadenovirus Turkey hemorrhagic enteritis virus, marble spleen disease virus (pheasant adenovirus
dsDNA 1)
Atadenovirus Egg drop syndrome virus (duck adenovirus A)
Polyomaviridae Polyomavirus Budgerigar fledgling disease virus, French molt in psittacines
Mardivirus Marek’s disease virus (gallid herpesvirus 2 and 3), turkey herpesvirus (meleagrid
herpesvirus 1)
Herpesviridae Iltovirus Infectious laringotracheitis virus (gallid herpesvirus 1)
(Subfamily Unassigned Pacheco’s disease virus (anatid herpesvirus 1)
alphaherpesvirinae) Duck enteritis virus (duck plague)
Pigeon herpesvirus (columbid herpesvirus 1)
Gyro virus Chicken anemia virus
Circoviridae Psittacine beak and feather disease virus
Circovirus Canary, duck, finch, goose and pigeon circovirus
ssDNA Parvovirus Chicken parvovirus
Parvoviridae Dependovirus Avian adeno-associated virus, goose parvovirus, Muscovy duck parvovirus

Orthoreovirus Avian reovirus (viral arthritis)


Reoviridae Tentative: duck reovirus
dsRNA
Rotavirus Chicken rotavirus D, F and G (Rotavirus enteritis)
Birnaviridae Avibirnavirus Infectious bursal disease virus
Avulavirus Newcastle disease virus (APMV-1), APMV 2 - APMV 9
Paramyxoviridae
ssRNA (-)
Metapneumovirus Turkey rhinotracheitis virus
Orthomyxoviridae Influenzavirus A Avian influenza virus
Coronaviridae Coronavirus Infectious bronchitis virus, turkey enteric coronavirus, pheasant coronavirus

Caliciviridae Unassigned Fowl calicivirus


Astroviridae Avastrovirus Turkey, duck and chicken (avian nephritis virus 1 and 2) astrovirus
Hepatovirus Avian encephalomyelitis-like virus
Avian entero-like virus, avian nephritis virus 3, cockatoo entero-like virus, duck
Picornaviridae hepatitis virus I and ΠΊ, guineafowl transmissible enteritis virus, turkey entero-like
ssRNA (+) Unassigned virus, turkey hepatitis virus, turkey pseudoenterovirus 1 and 2
Togaviridae Alphavirus Highlands J virus
Flaviviridae Flavivirus Israel turkey meningoencephalomyelitis virus, West Nile virus
Avian leukosis virus, Rous sarcoma virus, avian myeloblastosis virus, avian sarcoma
Alpharetrovirus virus, avian carcinoma Mill Hill virus 2
Retroviridae Gammaretrovirus Reticuloendotheliosis virus, chick syncytial virus, tragger duck spleen necrosis virus
Circular Hepadnaviridae A vihepadnavirus Duck hepatitis B virus
dsDNA Unassigned Ross’ goose hepatitis B virus
ds = double-stranded; ss = single-stranded; - = negative sense; + = positive sense; RT = reverse transcriptase.

Determining the type of nucleic acid in sucrose gradients can be achieved by depositing the Tris­
resuspended virus onto a 20% to 60% (w/v) sucrose gradient.
Direct Methods. Direct methods involve the concentration and Purified viruses are collected by side puncture of the plastic
purification of the virus, followed by extraction and characterization centrifuge tube with a 21-gauge hypodermic needle.
of the nucleic acid with nucleases or differential chemical Once purified, virus particles are disrupted with a lysis buffer
hydrolysis. After a large quantity of virus is cultivated, cells or containing 0.01M Tris-Cl (pH 7.8), 0.05M EDTA, 0.5% SDS and
tissues are disrupted by homogenization and/or freezing-thawing, proteinase K (50pg/ml). Since proteinase K digests native proteins,
releasing the viral particles. Initially, cellular debris is removed by DNAses or RNAses present in the lysates are rapidly inactivated,
centrifugation (1,000 x g for 10-12 min). The supernatant preserving the viral genetic material. Additional buffers used to
containing the viral particles is deposited on top of a 45% (w/v) disrupt cell or viral membranes are described elsewhere (31). After
sucrose cushion. After centrifugation (140,000 x g for 2 hr at 4 C), membrane disruption, the viral nucleic acid is extracted by adding a
pelleted virus particles are resuspended in Tris buffer (pH 7.6). volume of phenol:chloroform:isoamyl alcohol (24:24:1 v/v)
Viral purification may be performed by adjusting the density of the equivalent to the volume of the aqueous sample. Nucleic acid in the
Tris-resuspended virus to 1.40g/ml with cesium chloride followed aqueous phase is precipitated with ethanol and sodium acetate
by centrifugation (270,000 x g for 16 hr at 20 C). Viral purification (0.3M final concentration). Other commonly used cations to

211
Pedro Villegas and Ivan Alvarado

precipitate nucleic acids in association with ethanol are described SYBR Green I and Π stains, generating bright gold fluorescent
elsewhere (31). Finally, the viral genome is washed with 70% signals. It can detect 25 pg of dsDNA or 1 ng of RNA per band.
ethanol, pelleted and resuspended in RNase/DNase free water. • GelStar®: very stable stain at room temperature. It can detect as
Procedures for the extraction of DNA or RNA from tissues and cell little as 20 pg dsDNA, 25 pg ssDNA and 10 ng of RNA. Like
cultures are fully described elsewhere (31). ethidium bromide, GelStar® stain is a potential mutagen.
The following nucleases allow the differentiation of DNA and • Radiant Red®: highly sensitive stain that specifically detects
RNA viruses: RNA, emitting a red-orange signal with approximately 300 nm
• DNase I (RNase free -Pancreatic Deoxyribonuclease I): DNase I transillumination.
nonspecifically cleaves DNA to release 5’-phosphorylated DNA and RNA viruses can also be differentiated by treating the
dinucleotides, trinucleotides and oligonucleotides. DNase I also nucleic acid with sodium hydroxide (0.3M NaOH), which destroys
degrades ssDNA and the DNA present in RNA-DNA hybrids, RNA but not DNA molecules.
however, the activity for these substrates is reduced . One unit of Indirect methods. Metabolic inhibitors of nucleic acid replication
DNase I corresponds to the amount of enzyme able to degrade 1 can be used to determine if an isolated virus contains a DNA or
pg of DNA in 10 min at 37 C. RNA genome. These methods would be especially valuable in those
• Nuclease SI: under conditions of high ionic strength (0.1 - 0.4 M cases in which concentration, purification and direct
NaCl), low pH (pH 4.2), and the presence of ImM Zn2+ degrades characterization of nucleic acids by direct methods are not possible
ssDNA or RNA molecules to yield 5’-phosphate mono or due to low viral titers during replication.
oligonucleotides. One unit is able to digest 0.5 g of single Thymidine analogs are the simplest antiviral agents used to
stranded DNA in 30 min at 37 C. evaluate if a virus contains DNA (30, 31). Thymidine analogs, such
• RNase A: at low NaCl (ImM) concentrations, specifically attacks as 5-bromo-2N-deoxyuridme (BDU) and 5-iodo-2N-deoxyuridine
single or double stranded RNA 3’ to pyrimidine residues and (IDU), inhibit DNA replication and transcription. A cell-culture
cleaves the phosphate linkage to the adjacent nucleotide. At high system for virus propagation is preferred to evaluate the presence of
NaCl (150mM) concentrations, only attacks ssRNA molecules. DNA viruses using thymidine analogs. Briefly, the presence of a
One unit is the amount of enzyme required to yield an increase in DNA virus in cell cultures is evaluated by comparing the viral titer
absorbance at 286 nm of 0.0146 absorbance unit per min in a 1 ml of cell cultures treated with 50 pg/ml of BDU or IDU with the viral
volume at specific conditions. titer of non-treated cell cultures. DNA containing viruses will
• RNase Tl: cleaves ssRNA molecules by specifically attacking the exhibit a lower viral titer (1 log]0) in cell culture plates treated with
3’ phosphate groups of guanine nucleotides and cleaves the 5’ the thymidine analogs when compared with non treated cell culture
phosphate linkage to the adjacent nucleotide. plates. Controls should be run with known RNA and DNA
After digestion with a particular nuclease, the nucleic acid can be containing viruses. When treated like the unknown virus, the
electrophoresed on agarose or polyacrylamide gels. Different stains control DNA virus should be inhibited by the thymidine analog,
that can be used to selectively visualize nucleic acids are (31,35): while the RNA virus is not inhibited.
• Ethidium bromide: intercalates between base pairs of dsDNA (1 The sensitivity of a virus to actinomycin D is of value in
molecule every 2.5 bases approx). When exposed to ultraviolet differentiating some RNA viruses. Actinomycin D is a polypeptide
light (302 nm), it will fluoresce within the red-orange range of the antibiotic that interferes with the replication of viruses with dsRNA
visible spectrum. ssRNA when folded back into itself, or or dsDNA genomes. By binding to double stranded molecules,
• dsRNA can also be detected. Ethidium bromide is incorporated in Actinomycin D inhibits the synthesis of mRNA, blocking the
the gel or the electrophoresis buffer at a concentration of 0.5 translation of viral and host proteins (28, 31). Influenza virus
pg/ml. requires the transcription of some host messenger RNA as an early
• Methylene blue: used to stain bands of DNA in agarose gels in event in the replicative cycle and is inhibited by actinomycin D.
replacement of ethidium bromide. Its use minimizes the exposure Members of the Reoviridae and Bimaviridae families are also
of DNA molecules to UV irradiation, avoiding the generation of sensitive to actinomycin D because of their double-stranded
pyrimidine dimmers. After electrophoresis, the gel is immersed in genomes. The lack of cell culture toxicity by the concentration of
a solution containing 0.001 to 0.0025% methylene blue in ImM actinomycin D used in the cell culture medium must be previously
Tris-acetate (pH 7.4), 0.1 mM EDTA (pH 8.0). evaluated. Briefly, cell culture medium containing lpg/ml of
• Acridine orange: intercalates with both DNA and RNA actinomycin D is added to the cell culture for 2 hr before virus
molecules. Acridine orange produces fluorescense green at a inoculation. After 24 or 48 hr of virus inoculation, the virus is
wavelength of 525 nm and orange at wavelengths higher than 630 harvested and assayed for infectivity. A significant reduction in titer
when incorporated in DNA and RNA molecules, respectively. in the cultures treated with actinomycin D when compared with
• Silver stain: characterized by its high sensitivity, detecting titers in the untreated cultures indicates a sensitive virus (28).
Table 45.3 summarizes the identification of the type of nucleic
dsDNA molecules in polyacrylamide gels. Bands appear brown
against a yellow background. acid by direct and indirect methods.
• SYBR® dyes: display enhanced fluorescence upon binding Table 45.3 Comparison of direct and indirect methods for the
nucleic acids, showing higher affinity for nucleic acids and higher characterization of the nucleic acid genome of viruses.
sensitivity than ethidium bromide. SYBR® dyes are stimulated at DNA RNA
a wavelength (254 nm), causing the maximum DNA damage. Treatment SS DS SS DS
Three main SYBR® dyes are available: DNase I + + - -
• SYBR® Green I: stains DNA, detecting as little as 60 pg of SI nuclease + - - -
dsDNA per band. It has been commonly used in real time PCR RNase A (ImM NaCl) - - + +
reactions for viral quantitation. RNase A (150 mM NaCl) - - + -
• SYBR® Green Π: used to detect RNA with a greater sensitivity 0.3 M NaOH - - + +
than ethidium bromide. It is able to detect 100 pg of ribosomal Thymidine analogs + + - -
RNA on 1% agarose gels and 1 ng of ribosomal RNA on 5% Actinomycin D - + - +
acrylamide gels. Acridine Orange Red Green Red Green
• SYBR® Gold: detects DNA and RNA molecules after light
SS = single stranded, DS = double stranded; + = digestion or inhibition, - =
stimulation at a 300 nm wavelength. It penetrates gels faster than no digestion or no inhibition.
212
Chapter 45 Virus Identification and Classification

Sensitivity to lipid solvents by passing a small quantity (5 ml) of 10% calf serum in saline
Many viruses are surrounded by a lipid bilayer or envelope, through the filter will neutralize electrostatic charges on the
acquired from host cells during the nucleocapsid budding. Viral membrane. Viral adsorption to filter membranes can be also be
envelopes contain virus encoded proteins essential for infectivity. prevented by resuspending the virus in a Dulbecco’s phosphate
By using lipid solvents such as ether and chloroform, the presence buffered saline solution with 0.1% Tween 80 without Ca2+ or Mg2+
or absence of viral envelope can be evaluated (30). After treatment, (25). The technique should be aseptic throughout. The steps for
enveloped viruses lose their capacity to infect and replicate in sizing a virus through serum-treated filters are as follows:
propagation systems. A. Clarify the virus suspension by centrifugation (9200 x g for 10
min) for 10 min and/or by passing the virus suspension through a
Sensitivity to Ether prefilter membrane. Save a sample for assay.
Add ethyl ether, 20% by volume, to a measured amount of virus in B. Filter the clarified virus suspension through a 200 nm filter.
a screw-cap tube. Seal the tube with tape. Prepare a control with Sterilized filters can be inserted into sterile filter holders or attached
virus and 20% by volume of 0.85% NaCl. Place the tubes at 4 C for to the syringes. Filters that are purchased unsterilized can be
18-24 hr, shaking the tubes manually several times during this sterilized by autoclaving. Save a sample for assay.
period. Pour the contents of each tube into a sterile Petri dish, C. Pass the 200-nm filtrate through a 100-nm filter, save a sample
preferably in a chemical cabinet, and allow the ether to evaporate. for assay, and then pass the remainder through a 50-nm filter. The
Assay each virus. A loss of infectivity in the ether-treated virus of 1 filtrates of all filters are then assayed, and the titers are compared
log10 or greater is indicative of an enveloped virus. with those of the original clarified suspension.
D. The size of the virus may be up to 1.2 times the average pore
Sensitivity to Chloroform diameter of the filter through which no virus passed.
Dilute the sample to be tested 1:10 and deposit 2 ml aliquots in E. The integrity of each filter membrane must be evaluated by
two 15 ml glass centrifuge tubes. Add 0.2 ml of chloroform passing a bacterial culture through the filter immediately after
(CHC13) to one of the aliquot and 0.2 ml of 0.85% NaCl to the other passing the virus through the different filters, testing the filtrate for
aliquot. Vortex the centrifuge tubes for 10 min, keeping the tubes in sterility.
an ice bath between mixes. Allow CHC13 to sediment overnight in a
refrigerator or by centrifuging (1500 rpm for 30 min). Collect the Capsid symmetry
clear layer on the top and allow the remaining CHC13 to evaporate Protective protein structures, known as capsids or nucleocapsids,
in the hood for 10 - 15 min. Use the clear top layer for assay of encase and protect the viral genomes. Depending on the number of
virus infectivity. A loss of infectivity of 1 logio or greater indicates protein subunits and their structure, capsids can have a helical or
the presence of an enveloped virus (30). icosahedral symmetry (18). Helical capsids are organized around a
single axis and are described by the number of units per turn and the
Diameter of the virion axial rise per unit. Icosahedral capsids are formed by nonsymmetric
Virus size and shape is highly dependent on the amount and protein molecules arranged in identical equilateral triangular
arrangement of the proteins and the size and conformation of the structures joined to form an icosahedron. Most of the avian viruses
nucleic acid. Transmission electron microscopy is the most useful present helical or icosahedral capsids. However, some viruses such
way to determine the general morphology and size of a virus as poxviruses form complex capsids with structures still not well
particle. Detailed images of isolated and purified viral particles are understood. The symmetry of the nucleocapsid, obtained by
obtained by the formation of a cast after negative staining of the transmission electron microscope, is not essential to determine the
virus with uranyl acetate or potassium phosphotungstate (23). initial classification of a virus. All the same, many newly
Techniques such as cryoelectron microscopy, in which virus discovered viruses have been detected by the electron microscopy
isolates are preserved by rapid freezing in liquid nitrogen or liquid before they have been cultivated. An example is the rotavirus or the
helium, have permitted a more detailed visualization of electron­ family Reoviridae. The transmission electron microscope is a
scattering contrast from the particle itself and not just from the cast. valuable tool in virology and can give nearly all the information
With the use of cryoelectron microscopy, the study of unstable or required to identify and classify a virus: lipoprotein envelope, size,
relative impure preparations can be achieved (23). Further atomic and morphology. An electron microscopy guide of viruses has been
resolution of viral structures or particular viral proteins can be published by Doane and Anderson (15). The basic procedure entails
achieved by X-ray crystallography after virus crystalization. Large concentration of the virus by ultracentrifugation and resuspension of
and more complex viral particles that cannot be crystallized must be the virus in low-salt or salt-free distilled water. A drop of virus is
dissected into well defined subunits or substructures (23). The size then placed on a plastic-coated grid and mixed with a drop of 2%
of a virus may also be estimated with fair accuracy by the use of phosphotungstic acid in distilled water (adjust to pH 6.5 with 1 N
nitrocellulose, polycarbonate or polyester membranes with KOH). Viral concentrations of 106/ml or greater must be present to
relatively uniform pores. Virus suspensions are mainly filtrated by detect virions with the electron microscope.
stereohindrance; i.e., large particles are not able to pass through The main characteristics used to distinguish between the families
smaller holes (25). Membrane filters with pore diameters of 50 nm, of viruses of greatest importance to avian species are summarized in
100 nm, 200 nm, 300 nm and 450 nm are available. The most table 45.4. Serological tests, such as virus-neutralization, gel­
commonly filters used to determine the diameter of the viruses are precipitation, hemagglutination-inhibition, complement-fixation,
those of 50 nm, 100 nm and 200 nm. Several factors may affect the and enzyme-linked immunosorbent are of great value for the
results of filtration of a virus, including clumping, pleomorphism, identification of viruses. Once the virus is assigned to a particular
occlusion of the pores with cellular debris, and electrostatic charges family, further classification at the strain level can be achieved by
on the filter membrane. Initial filtration of viral suspensions through nucleic acid based techniques. The techniques are discussed
a series of membranes for clarification and/or a 450 nm filter will elsewhere in this manual.
preclude the clogging of membranes by clumping and the presence
of cellular debris. Pretreatment of filters by soaking in calf serum or

213
Pedro Villegas and Ivan Alvarado

Table 45.4 Properties of DNA and RNA used to differentiate families of avian viruses.

Differential Properties
I. Sensitive to thymidine analogs DNA virus
A. Ether sensitive
• 1. Passes 200 nm filter Herpesviridae
• 2. Retained by 200 nm filter PoxviridaeA
B. Ether resistant
• 1. Retained by 50 nm filter Adenoviridae
• 2. Passes 50 nm filter Parvoviridae
Papovaviridae
• 3. Retained by 200 nm filter PoxviridaeA

Π. Resistant to thymidine analogs RNA virus


A. Ether sensitive
• 1. Hemagglutination positive
o Sensitive to actinomycin D Orthomyxoviridae
o Resists actinomycin D Paramyxoviridae
• 2. Hemmaglutination negative
o Sensitive to actinomycin D Retroviridae
o Resists actinomycin D Coronaviridae
Flaviviridae®
B. Ether resistant •
• 1. Sensitive to actinomycin D Reoviridaec
Bimaviridaec
• 3. Resists actinomycin D Picomaviridae®
A The poxviruses may or may not be destroyed by ether and/or chloroform.
B Passes 50 nm filter.
c Titer greatly reduced by 50 nm filter.

Nucleic acid based viral detection and characterization locations lead to different patterns of fragments after separation by
Several molecular methods are currently used not only for the electrophoresis in agarose or polyacrylamide gels. A limitation of
detection and characterization of avian viruses but also for this technique is the lack of detection of mutations present outside
evolutionary and epidemiological studies (12). PCR-based of the recognition sites for the specific restriction enzymes chosen
approaches have been crucial in the ability to characterize and for analysis. Also, identical restriction patterns between two
compare the genomes of viruses and to detect viruses that have been different isolates may require the use of additional restriction
difficult to isolate using classical culture methods. Additional enzymes to show differences (31).
advantages of PCR-based approaches include the high sensitivity • Southern blot analysis. Viral DNA, PCR or RT-PCR amplified
and specificity and the capacity to distinguish individual isolates product, is digested with a restriction enzyme, and the
within a serotype by amplification of a small segment of the viral fragments are electrophoresed in agarose or polyacrylamide
genome containing hypervariable regions. The presence of changes gels, transferred onto a nitrocellulose paper and then hybridized
at the nucleotide level, which may or may not result in amino acid with labeled probes from the entire or specific regions of the
changes, is the basis of subtype classification of viruses (5). genome (31). This technique can be used to determine
The following are the molecular techniques routinely used to similarities and differences between two closely related viruses.
characterize avian viruses: • Heteroduplex mobility assay. Following PCR or RT-PCR
• Nucleotide sequencing. Obtained after genomic amplification reactions, an amplified segment of an isolated virus is denatured
by PCR (DNA viruses) or RT-PCR (RNA viruses), nucleotide and reannealed in the presence of DNA segments from a know
sequencing has the potential to detect even one single mutation virus strain. After re-annealing, three types of bands will be
(5). Commonly used for epidemiological and classification observed, one band consisting of homoduplexes and two other
studies at low levels such as strains within a particular serotype bands consisting of heteroduplexes. Nucleotide mistmatches
or variants. Usually genome segments codifying major present in the heteroduplexes will form bulges, retarding the
antigenic epitopes located at the surface of the virion are movement of the band during electrophoresis in polyacrylamide
amplified and sequenced. However, for evolution studies, gels. The relative retardation of the heteroduplexes is
conserved regions, such as untranslated regions may be proportional to the DNA distance (percent of nucleotide
sequenced. difference) between the strains being compared (6, 8).
• Restriction Fragment Length Polymorphism analysis (RFLP). The application of the previously described techniques in the
Double stranded DNA is cleaved by specific restriction detection and characterization of avian viruses is summarized in
enzymes in a sequence dependent fashion to produce different table 45.5.
length fragments. Presence of nucleotide mutations in specific

214
Chapter 45 Virus Identification and Classification

Table 45.5 Nucleic acid-based methods currently used to detect and characterize avian viruses

Virus Gene(s) of Interest Molecular Technique(s) Objective


RT-PCR (27)
Nucleotide sequencing
Southern blot analysis Pathotyping based on the sequence of cleavage site.
Fusion (F)
Immunosorbent ELISA Epidemiological studies
TaqMan fluorogenic probes
Heteroduplex mobility assay
Newcastle Disease Virus (1,2, 36) Hemagglutinin/
Neuraminidase Epidemiological and evolution studies
Nucleotide sequencing
Epidemiological studies
(H/N)
Viral detection and characterization
Matrix
Epidemiological studies
Nucleocapsid RT-PCR Group specific detection
Membrane RT-PCR Group specific detection
Infectious Bronchitis Virus (4, 10, 14,
RT-PCR
22) Viral detection
SI Nucleotide sequencing
Genetic characterization
Slot blot hybridization
Virus characterization (HA subtyping)
RT-PCR
Hemagglutinin Epidemiological studies
Nucleotide secuencing
Avian Influenza (9)
RT-PCR Virus characterization (NA subtyping)
Neuraminidase
Nucleotide sequencing
Matrix RT-PCR Viral detection
RT-PCR
Nucleotide sequencing Viral detection
RFLP Differentiation of classical (standard and very
Infectious bursal disease (7, 33) VP2
In situ RT-PCR virulent) from variants strains
In situ hybridization
TaqMan fluorogenic probes
Genetic characterization
Chicken infectious anemia/other
VP1 PCR / sequencing Epidemiological studies
circovirus (32, 34)
RT-PCR / sequencing Detection of exogenous and endogenous RNA or
P27, Envelope and PCR / sequencing proviral DNA
Avian Leukosis Virus (16)
LTR In situ hybridization with RNA and Subgroup detection
DNA probes Tropism studies
Avian Adenoviruses (3,21) Hexon PCR/RFLP Virus classification in 5 molecular groups
ICP4 andUS3 PCR Serotype 1 and 3 detection
gD TaqManPCR Viral detection
Marek’s disease virus (13,20)
Meq, gB and Tandem
PCR Serotype 1 identification
repeat
F Southern blot RT-PCR Molecular characterization
Avian Metapneumovirus (11) Matrix RT-PCR Viral detection

8. Berinstein, A., H. Sellers, D. J. King, and B. S. Seal. Use of a


REFERENCES heteroduplex mobility assay to detect differences in the fusion protein
cleavage site coding sequence among Newcastle disease virus isolates. J.
1. Aldous, E. W., and D. J. Alexander. Detection and differentiation of Clin. Microb. 39:3171-3178. 2001.
Newcastle disease virus (avian paramyxovirus type 1). Avian Path. 30:117- 9. Brown, I. H. Advances in molecular diagnosis for avian influenza. Dev.
128. 2001. Biol. 124:93-97. 2006.
2. Aldous, E. W., J. K. Mynn, J. Banks, and D. J. Alexander. A molecular 10. Callison, S. A., M. W. Jackwood, and D. A. Hilt. Molecular
epidemiological study of avian paramyxovirus type 1 (Newcastle disease characterization of infectious bronchitis virus isolates foreign to the United
virus) isolates by phylogenetic analysis of a partial nucleotide sequence of States and comparison with United States isolates. Avian Dis. 45:492-499.
the fiision protein gene. Avian Path. 32:239-257. 2003. 2001.
3. Alvarado, I., P. Villegas, J. El-Attrache, E. Jensen, G. Rosales, F. Perozo, 11. Casey, J. L. Specific detection of avian pneumovirus (APV) US isolates
and L. Purvis. Genetic characterization, pathogenicity and protection studies by RT-PCR. Arch. Virol. 145:1239-1246. 2000.
with an avian adenovirus isolate associated with inclusion body hepatitis. 12. Cavanagh, D. Innovation and discovery: the application of nucleic acid­
Avian Dis. In Press. 2007. based technology to avian virus detection and characterization. Avian Path.
4. Alvarado, I., P. Villegas, N. Mossos, and M W. Jackwood. Molecular 30:581-598. 2001.
characterization of avian infectious bronchitis virus strains isolated in 13. Davidson, I., R. Borenshtain, and Y. Weisman. Molecular identification
Colombia during 2003. Avian Dis. 49:494-499. 2005. of the Marek's disease virus vaccine strain CVI988 in vaccinated chickens. J.
5. Arens, M Methods for subtyping and molecular comparison of human Vet. Medicine. 49:83-87. 2002.
viral genomes. Clin. Microb. Rev. 12:612-626. 1999. 14. De Wit, J. J. Detection of infectious bronchitis virus. Avian Path. 29:71-
6. Banda, A., P. Villegas, and J. El-Attrache. Heteroduplex mobility assay 93. 2000.
for genotyping of infectious bursal disease virus. Avian Dis. 48:851-862. 15. Doane, F. W., and N. Anderson. Electron microscopy in diagnostic
2004. virology. Cambridge University Press, New York. 1987.
7. Banda, A., P. Villegas, and J. El-Attrache. Molecular characterization of 16. Fadly, A. M Isolation and identification of avian leukosis viruses: A
infectious bursal disease virus from commercial poultry in the United States review. Avian Path. 29:529-535. 2000.
and Latin America. Avian Dis. 47:87-95. 2003. 17. Fauquet, C. Μ, M A. Mayo, J. Maniloff, U. Desselberger, and L. A.
Ball. Virus taxonomy, 8th ed. Springer-Verlag Wien, New York. 2005.

215
Pedro Villegas and Ivan Alvarado

18. Flint, S. J., L. W. Enquist, R. M Krug, V. R. Racaniello, and A. M 27. Perozo, F., P. Villegas, C. Estevez, I. Alvarado, and L. Purvis. Use of
Skalka. Principles of Virology: molecular biology, pathogenesis and FTA filter paper for the molecular detection of Newcastle disease virus.
control., 1 ed. American Society of Microbiology Press., Washington, DC. Avian Dis. 35:93-98. 2006.
2000. 28. Puissant, C., and L. M. Houdebine. An improvement of the single-step
19. Hamoud, Μ Μ, P. Villegas, and S. M. Williams. Detection of method of RNA isolation by acid guanidinium thiocyanate-phenol-
infectious bursal disease virus in formalin-fixed paraffin-embedded tissue by chloroform extraction. Bio Techniques 8:148-149. 1990.
immunohistochemistry and real-time reverse transcription-polymerase chain 29. Purvis, L., P. Villegas, and F. Perozo. Evaluation of FTA paper and
reaction. J. Vet. Diag. Invest. 19:35-42. 2007. phenol for storage, extraction and molecular characterization of infectious
20. Handberg, K. J., O. L. Nielsen, and P. H. Jorgensen. The use of serotype bursal disease virus. J. Virol. Methods. 138:66-69. 2006.
1- and serotype 3-specific polymerase chain reaction for the detection of 30. Rovozzo, G. C., and C. N. Burke. A manual of basic virological
Marek's disease virus in chickens. Avian Path. 30:243-249. 2001. techniques. Prentice-Hall, Englewood Cliffs, N. J. 1973.
21. Hess, M Detection and differentiation of avian adenoviruses: A review. 31. Sambrook, J., and D. W. Russell. Molecular cloning: A laboratory
Avian Path. 29:195-206. 2000. manual., 3rd ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor,
22. Jackwood, M W., D. A. Hilt, C. W. Lee, Η. M Kwon, S. A. Callison, NY. 2001.
K. M Moore, H. Moscoso, H. Sellers, and S. Thayer. Data from 11 years of 32. Todd, D. Circoviruses: Immunosuppressive threats to avian species: A
molecular typing infectious bronchitis virus field isolates. Avian Dis. review. Avian Path. 29:373-394. 2000.
49:614-618. 2005. 33. Van den Berg, T. P. Acute infectious bursal disease in poultry: A
23. Knipe, D. Μ, P. M Howley, D. E. Griffin, R. A. Lamb, M A. Martin, review. Avian Path. 29:175-194. 2000.
B. Roizman, and S. E. Straus. Fields Virology. Lippincot Williams & 34. Van Santen, V. L., F. J. Hoerr, and L. H. Lauerman. Genetic
Williams, Philadelphia, PA. 2001. characterization of chicken anemia virus from commercial broiler chickens
24. Lee, C. C., T. P. Ko, C. C. Chou, M Yoshimura, S. R. Doong, Μ Y. in Alabama. Avian Dis. 45:373-388. 2001.
Wang, and A. H. Wang. Crystal structure of infectious bursal disease virus 35. Williams, L. R. Staining nucleic acids and proteins in electrophoresis
VP2 subviral particle at 2.6A resolution: Implications in virion assembly and gels. Biotechnic & Histochemistry 76:127-132. 2001.
immunogenicity. J. Struct. Biol. 155:74-86. 2006. 36. Wise, M G., D. L. Suarez, B. S. Seal, J. C. Pedersen, D. A. Senne, D. J.
25. Lytle, C. D., L. B. Routson, N. B. Jain, M R. Myers, and B. L. Green. King, D. R Kapczynski, and E. Spackman. Development of a real-time
Virus passage through track-etch membranes modified by salinity and a reverse-transcription PCR for detection of Newcastle disease virus RNA in
nonionic surfactant. Appl. Environ. Microbiol. 65:2773-2775. 1999. clinical samples. J. Clin. Microb. 42:329-338. 2004.
26. Moscoso, Η., I. Alvarado, and C. Hofacre. Molecular analysis of
infectious bursal disease virus from bursal tissues collected on FTA filter
paper. Avian Dis. 50:391-396. 2006.

216
46
TITRATION OF BIOLOGICAL SUSPENSIONS
Pedro Villegas

SUMMARY. Methods for titration of biological suspensions (viruses) include the procedures devised by Reed and Muench and by
Spearman-Karber. Appropriate selection of the host to be inoculated as well as dilution range are important considerations to guarantee
reliable results. The methodology to calculate geometric mean titers is described.

INTRODUCTION TYPES OF INFECTIVITY ASSAYS

The activity of a biological suspension, such as a virus, is Two types of infectivity assays are used to quantify viruses:
measured quantitatively by procedures that consist of preparing quantal and enumerative (also known as quantitative or focal). In
dilutions of the suspension and determining the dilution at which a the quantal assay, the response is either positive or negative; the
particular biological activity is still detectable. In the preparation of measurement is based on an “all or none” approach: the host
dilutions to test the infectivity of a biological suspension or virus, survived or died, cytopathic effect (CPE) is present or absent, signs
the following principles should be considered. or lesions of the infection are observed or not present, and so on. In
the enumerative response, the number of infectious virus particles
Diluents present in the suspension can be quantified.
When the virus is being diluted, the diluent must preserve
infectivity and be nontoxic for the host. For most viruses, a Quantal Assays
balanced salt solution or a cell-culture medium can be used for cell­ To perform quantal assays, serial dilutions of the virus are
culture systems and a nutrient or tryptose broth can be used for inoculated in the appropriate host. After variable incubation times,
embryos. With extremely labile viruses, it is often advantageous to at each dilution, the hosts are evaluated and determined to be either
include 2-5% serum, although the serum must be pretested and positive or negative to the effect of the virus. The proportion of the
found to be free of antiviral activity. positive hosts is recorded and used to calculate concentration or titer
of the viral suspension.
Preparation of Dilutions A unit of infectivity used to express the results for the quantal
Separate sterile pipettes must be used for each dilution of the response is the mean infective dose (Π)50), which represents the

Table 46.1. Example of estimating the 50% endpoint by the method of Reed and Muench

Embryos Accumulated Numbers

Virus Dilution Proportion


Number Dead Number Alive Dead Alive Percent Dead
Inoculated Dead/Total

IO’3 5 0 12 i 0 12/12 100


io-4 4 1 7 1 7/8 88
IO'5 2 3 3 4 3/7 43
W6 1 4 1 8 r 1/9 11
suspension, although the same pipette can be used for mixing and amount of agent capable of infecting 50% of the hosts. When the
transferring a single dilution. After the required volume of mixture effect is based on lethality, the term mean lethal dose (LD50) is
is transferred to the next tube, the pipette is changed. While used. Both the Π)50 and the LD50 represent the titer of a biological
dilutions are being prepared, all reagents and the sterile dilution suspension, which is usually expressed as the number of infectious
tubes should be kept on ice. units per unit volume (usually 1 ml). The Π)50 is the recommended
procedure used to express agent infectivity. Other units of
Performing the Test infectivity are also used (mean embryo infective dose [EID50], mean
Begin with the least concentrated preparation (highest dilution) tissue culture infective dose [TCID5o]).
and inoculate the volume desired into each susceptible host by the Calculation of Titers. After performing the titration and
route specified. The same instrument (syringe and needle or pipette) determining the number of individual hosts that respond to each
can be used to inoculate several dilutions in a series when the least dilution, the endpoint dilution must be determined. The endpoint is
concentrated suspension is inoculated first and the most defined as the highest dilution of virus that will produce a
concentrated is inoculated last. detectable effect in 50% of the inoculated hosts. An endpoint
Nonspecific deaths resulting from manipulation of birds or usually cannot be determined by visual examination of the data, so
embryonated chicken eggs during inoculation occur most frequently endpoints are determined mathematically. Several mathematical
within a few hours of inoculation. Deaths up to 18-24 hr methods have been described (1,3,4,5,6,7).
postinoculation are generally considered to be nonspecific and are Method of Reed and Muench. A formula devised by Reed and
eliminated from the calculations. Muench (7) permits interpretation of the 50% endpoint from the
Each group of subjects inoculated should be examined regularly data derived from a quantal response and groups of test subjects.
(daily unless stated otherwise), and relevant observations should be Table 46.1 gives an example of the application of this formula on
recorded. At the end of the specified time, which will vary with the information obtained from inoculation of a lethal virus into
agent, each individual is examined and recorded as unaffected, embryonated eggs. The formula can be applied similarly to rates of
affected, or dead by the appropriate criteria. infection in any host system.
The first column in the table lists the dilutions inoculated. The
second column lists the number of embryos that died, and the third
217
Pedro Villegas

column lists the number of live embryos. The next two columns are Method of Spearman-Karber. Many researchers prefer the
the accumulated numbers obtained from columns two and three. Spearman-Karber (1) method of calculating the ID50. In most
The accumulated numbers for the dead embryos are calculated on instances, this method does not involve a great number of
the assumption that if the host or system was affected at a particular calculations and the results are as accurate as those obtained with
dilution, it should also be affected at the next lowest dilution, which the method of Reed and Muench. The Spearman-Karber method
has a greater virus concentration. Similarly, the accumulated can be used only with data including a 100% response or when one
numbers for the unaffected embryos are calculated on the can reasonably expect a 100% response at the next higher dose
assumption that if the host was not affected at a particular dilution, level. To calculate the ID50 by this method, we use the following
it should not be affected at the next highest dilution, which has a condensed formula:
lower virus concentration. Because of this assumption, the numbers dZri
of affected embryos are added beginning at the highest dilution, and ED50 = x + /2d - n
the numbers of unaffected embryos are added beginning at the
lowest dilution. where
Once the accumulated numbers are calculated, the ratio of the x = the highest dilution level tested,
number of dead to the total number of inoculated hosts is calculated d = the interval between successive logarithmic doses
for each dilution. A percentage is then obtained, and the two (dilution factor),
dilutions that bracket the 50% endpoint are found. In the above Ση = the total number of uninfected hosts, and
example, the 50% endpoint is between the IO'4 (88%) and 10‘5 n = the number of hosts used at each dilution level
(43%) dilutions. We use the following formula to calculate the (constant).
proportionate distance between 10‘4 and 10‘5: When the data given for the calculation of the fD50 by the
method of Reed and Muench are used in the above formula, the
Percentage infected at formula becomes:
dilution next above 50% - 50% 1.0(1+ 3+ 4)
PD = ------------------------------------------------------------------ ID50= 6.0 + ^(1.0) - 5
Percentage infected at Percentage infected 8X)
dilution next - at dilution next = 6.0+ 0.5- 5
above 50% below 50% = 6.0+ 0.5- 1.6
= 4.9
88 - 50 38 = 104 9 per 0.1 ml or 105 9 per ml.
PD = ____ =___ = 0.84 -0.8. When 10-fold dilution series are used to inoculate the appropriate
88 - 43 45 hosts, the formula can be reduced to facilitate the calculations:
Στι
The 50% endpoint can now be calculated using the following ID50 = x + 0.5 - n
formula:
The reason that this is done is that the interval between dilutions (d)
log of the = (log dilution - (PD x log is log 10 or 1.0.
50% endpoint above 50%) dilution factor)
ENUMERATIVE RESPONSE
= -4-(0.8 x 1.0)
= -4.8. Depending on the host system used (e.g., chorioallantoic
membranes of embryonated eggs, or cell culture) and the type of
Therefore, the 50% endpoint dilution is IO’4 8 and has no units. response obtained, titers are expressed as pock-forming units,
plaque-forming units (both of these are abbreviated PFU), or focus­
The titer is defined as the number of infectious units per unit forming units (FFU) per milliliter. For this method, the average
volume. Therefore, the titer of the preparation is the negative number of plaques from duplicate plates or tests (two per dilution)
exponential of the endpoint dilution and is expressed as LD50/dose. has to be correlated with the amount of virus added or injected
If the dose were 0.1 ml, the titer would be 105 8/ml. initially, as follows:
It is usual to include in the expression of log titers only one figure
to the right of the decimal point (1058 LD5o/ml, not 105 8433). virus dilution = IO'5,
Extension of additional figures in the mantissa implies a degree of volume inoculated = 0.1 ml,
accuracy that is not real. plaque counts = 145,138,
If dilutions are other than 10-fold, the proportionate distance must average plaque count = 141, and
be multiplied by the logi0 of the factor; for example, for fivefold titer = 1.4x 108PFU/ml.
dilutions such as 101, IO"1'7, and IO’24, multiply the proportionate
distance by 0.6990. For twofold dilutions, multiply the PD by 0.3; Cell cultures can be used in a quantal or an enumerative response.
for fourfold dilutions multiply by 0.6. For 10-fold dilutions, the log Under liquid medium, viruses that spread extracellularly eventually
of the dilution factor (which is 10) is 1.0. will infect the whole culture and produce CPE or viral antigens. In a
Protective doses (PD) can also be calculated using this procedure, quantal response, replicate cultures can be scored for the presence
as for the PDqq, frequently used to calculate the potency of or absence of virus effect. The enumerative response takes
inactivated vaccines in chickens. To calculate a PD90, the advantage of the fact that an infectious virus particle can infect a
proportional distance will be calculated using the following single cell, multiply, and give rise to a circumscribed change in the
formula: cell monolayer under semisolid medium, producing an enumerative
response or plaque assay. Spread of virus through the supernatant
PD = Percentage of birds protected at the dilution next above 90% - medium must be curtailed by solidifying the medium with agar or
90%, divided by methyl cellulose. Agar and other constituents of the medium are
Percentage of birds protected at the dilution next above 90% - sometimes helpful in maintaining the integrity or health of the
Percentage of birds protected at the dilution next below 90% monolayer of normal cells while the virus-infected cells are
218
Chapter 46 Titration of Biological

changing. With some viruses in cell culture, it may be helpful to add be used to find the correct dilution of the preparation. In the
a vital stain such as neutral red to the solid medium or to add the following example, the method using logarithms and the stepwise
stain on top of the medium after the virus has had a chance to method will be used to find the correct dilution. A host system in
replicate. With other viruses or cultures, the solid medium can be the laboratory must be inoculated with 1000 IDso/ml from a virus
removed, and the cells can be fixed and then stained with preparation that has a titer of 105 4 ID50 per ml.
hematoxylin and eosin or other stains. For some assays, direct gross Method Using Logarithms. The number of ID50S needed is to be
or microscopic observation without staining may be satisfactory. divided into the titer of the preparation:
Many viruses inoculated on the chorioallantoic membrane of 105 4 titer of the virus preparation per milliliter
embryonated eggs produce circumscribed lesions (pocks) that can -ί- 103 0 amount of virus needed in 1.0 ml
be counted. = 102 4 or 2.5 (antilog of 0.4) x 102.
This figure with a negative exponent would represent the dilution at
DILUTIONS which the original preparation must be diluted to obtain the 1000
ID50. The 10‘2 dilution can be prepared and 1 ml of it taken up to a
For biological suspensions, the dilutions used are those in which final volume of 2.5 ml to obtain the correct ID50. If the decimal
two liquids are mixed in different proportions, that is, fraction in the titer is to be eliminated, the antilog of 0.4 should be
volume:volume (or volume/volume, v/v). In serologic and virologic found. This antilog corresponds to 2.5; therefore,
dilutions, a 1:2 (or a 1/2 dilution) is considered to be that dilution 105 4 = 2.5 x 105 ID5()/ml.
composed of one volume of the substance in a total of 2 volumes of
the mixture, that is, one volume of each substance. For the rest of Stepwise Method. The following step-by-step procedure can be
this chapter, we use the proportional (e.g., 1:2) rather than the used to find the dilution containing the 1000 ID50 in 1.0 ml of
traditional (e.g., 1/2) form. A 1:10 dilution is that composed of one inoculum:
volume of the biological reagent and nine volumes of diluent.
Serial dilutions are used to accurately determine the titer of virus 1 ml of 10° dilution contains 105 4 or 2.5 x ΙΟ5 Π)50,
preparations. These serial dilutions are commonly done using 1 ml of 101 dilution contains 104 4 or 2.5 x 104 Π)50,
factors of 2, 5, or 10. The most commonly used serial dilution in 1 ml of IO'2 dilution contains 103 4 or 2.5 x 103 tD50 or 2500 ID50.
avian virology is the 10-fold dilution, although it is well recognized
that the smaller the factor, the more precise the titer will be. At this point, the formula C x V = CN x VN can be used as follows:
Depending on the final volume needed, the 10-fold dilution series when
can be prepared using constant volumes of 9.0, 4.5, or 0.9 ml of C = 2500ID5Q,
diluent. V = 1 ml (assuming that only 1 ml is to be taken from the final
suspension),
CONVERSION OF TITER TO DILUTION C’ = 1000ID50, and
V’ = unknown,
For conversion of a more concentrated solution of known titer or using the above formula:
concentration to a more diluted solution, the following formula can 2500 x 1 = 1000 x V, and
be used: V’ = 2.5
C x V = C’ x V’ Therefore, 1 ml of the IO’2 dilution of the virus preparation has to
where be taken up at a final volume of 2.5 ml, that is, 1.5 ml of diluent and
C = original titer or concentration, 1.0 ml of the IO'2 dilution of virus.
V = original volume,
C’ = required or final titer, concentration, and CALCULATION OF GEOMETRIC MEAN TITERS
V’ = required or final volume.
Individual serologic results are usually expressed as the reciprocal
For example: a virus preparation with a titer of ΙΟ50 EID50/ml is of the endpoint serum dilution. This system is used for twofold
needed to inoculate 600 chickens with ΙΟ30 EID50 in 0.1 dilution series that begin with either a 10-fold or a twofold dilution.
ml/chicken. Examples of these results are 10, 20,40,..., 320; or 2, 4, 8,..., 256.
The first step is to have the virus titers expressed in the same unit These results are usually expressed in arithmetic terms, which can
volume. Therefore, the titer of the virus preparation should be be useful when the results of a few serum samples are to be
calculated per 0.1 ml, which will be 104 0 ΕΠ)50 per 0.1 ml. Now the interpreted. When a large number of results from individual serum
formula can be applied as follows: samples are to be analyzed, however, the arithmetic system of
individual titer expression becomes very time consuming. Also, a
IO40 x V = 1030 x (600 x 0.1) few samples with very high or low titers will give an unreasonable
103 0 x 60 arithmetic mean, which can be misleading when the results are
V= IO40 interpreted. Most of the serologic results found in the literature are
V = 6. presented as geometric mean titers (GMTs), although they are
usually expressed in arithmetic terms. Therefore, knowing the
A total of 6 ml of the original virus preparation should be taken to a procedure for calculating the GMT is important. The general
final volume of 60 ml. It is always advisable to prepare additional formula for calculating a geometric mean (GM) is:
amounts of virus. The above formula can also be used when the
concentrations are expressed as proportions (1:8; 1:15), although it GM="y^IX2X3...Xn
is easier to convert the fraction to a percentage and then apply the where:
formula. X = value of the observation, and
n = number of observations.
Determining Number of Infectious Units
When the titer of a virus preparation is known and a determined The GMT can be determined by a number of methods, including
number of infectious units must be obtained, several methods can those that follow.
219
Pedro Villegas

Method Using Logio Titers 2) Add all of the partial results and divide by the total number of
samples:
The base-10 logarithm (logio) of the reciprocal of the titer of each 4.193 + 9.107 + 8.010 + 3.806 = 25.116
sample is added, and the sum is divided by the total number of 25.115 - 20 = 1.2558, which is the log of the GM.
samples. The resulting number represents the logarithm of the GM. 3) Find the antilog of this amount, and that will be the GMT:
The GMT will be the antilogarithm (10x) of this resulting number; antilog of 1.2558 = 18.02
for example, results of a hemagglutination-inhibition (HI) test GMT =18.02.
obtained from 20 serum samples were as follows (titers expressed
as the reciprocal of the serum dilution): six samples with a titer of 5, Method Using Tube Number (Modified Log2) and Tables
seven samples with a titer of 20, five samples with a titer of 40, and When the end dilution points of serologic tests are recorded by
two samples with a titer of 80. To find the GMT log10: tube number and any twofold dilution series is used, the GMT can
1) Calculate the log10 of each of the titers and multiply by the be easily calculated by referring to a table developed by Brugh (2),
number of samples with that titer: reproduced here by permission (Table 46.2). The GMT can be
log10 5 = 0.698 x 6 = 4.193, calculated when the original dilution is either 1:2, 1:5, 1:10, or 1:20.
log1020= 1.301 x 7 = 9.107,
log10 40 = 1.602 χ 5 = 8.010, and
log10 80= 1.903 x 2 = 3.806.

Table 46.2. Conversion of base-two logarithmic mean titers to geometric mean titers (GMTs) (source, M. Brugh [2]).

Mean titei^Reciprocal of GMT at proportionate distance between dilutions.


1:5 1:10 1:20 0.0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9

1 - - 5 5 6 6 7 7 8 8 9 9
2 1 - 10 11 12 12 13 14 15 16 17 19
3 2 1 20 21 23 25 26 28 30 32 35 37
4 3 2 40 43 46 49 53 57 61 65 70 75
5 4 3 80 86 92 98 106 113 121 130 139 149
6 5 4 160 171 184 197 211 226 243 260 279 299
7 6 5 320 343 368 394 422 453 485 520 557 597
8 7 6 640 686 733 788 844 905 970 1040 1114 1194
9 8 7 1280 1372 1470 1576 1689 1810 1940 2079 2229 2389
10 9 8 2560 2744 2941 3152 3378 3620 3880 4159 4457 4777
11 10 9 5120 5487 5881 6303 6756 7241 7760 8317 8914 9554
12 11 10 10,240 10,975 11,763 12,607 13,512 14,482 15,521 16,635 17,289 19,109
13 12 11 20,480 21,950 23,525 25,214 27,024 28,963 31,042 33,270 35,658 38,217
14 13 12 40,960 43,900 47,051 50,428 54,047 57,926 62,084 66,540 71,316 76,434
15 14 13 81,920 87,800 94,101 100,856 108,094 115,852 124,168 133,079 142,631 152,868
16 15 14 163,840 175,599 188,203 201,711 216,188 231,705 248,335 266,159 285,262 305,736
A Mean of titration endpoints expressed by dilution or tube number. Dilution of test material (for example, serum) in first tube of twofold series.
For assays with an initial dilution of 1:2, use the 1:20 column and divide results by 10.

Depending upon which original dilution was used, the whole Arithmetic Method for Any Dilution Factor
number (integer) obtained from the average of the tube numbers is The titer can be calculated mathematically for a twofold dilution or
found in one of the three columns on the left side of Table 46.2. The for any other dilutions with the following formula:
decimal fraction is then found in the column heads of the rest of the GMT = antilog (average endpoint tube number - 1) x (log of the
table. For example, if the original dilution used in an HI test for 10 factor) + (log of reciprocal of first dilution).
serum samples was 1:5, the results were recorded by tube number as In the example from above:
follows: two samples with endpoint in tube 3, three samples with GMT = antilog (4.4 - 1) x (0.30102) + (log 5)
endpoint in tube 4, four samples with endpoint in tube 5, and one = antilog 1.0234 + 0.6990
sample with endpoint in tube 6. = antilog 1.7224
The average of the tube number is calculated: = 52.77 = 53.
(2 χ 3) + (3 x 4) + (4 χ 5) + (1 x 6) = 44
44 - 10 = 4.40
The whole number 4 is found in the column under 1:5 dilution and
the decimal fraction 0.4 is found on the right side of the table. The
GMT from the table is 53. This method is very useful, especially
when automatic equipment is used and large numbers of samples
are tested.

220
Chapter 46 Titration of Biological Suspensions

REFERENCES 5. Cottral, G. E., ed. Manual of standardized methods for veterinary


microbiology. National Academy of Sciences, National Research Council,
1. Brian W. J., and Η. O. Kangro. Virology methods manual. Academic Washington, D.C. p. 731. 1978.
Press, Inc., San Diego, California, 1996. 6. Hsiung, G. D. Diagnostic virology. Yale University Press, New Haven,
2. Brugh, M A., Jr. A simple method for recording and analyzing Conn. 1982.
serological data. Avian Dis. 22:362-365. 1978. 7. Reed, L. J., and H. Muench. A simple method for estimating fifty percent
3. Burleson, F. G., T. M Chambers, and D. L. Wiedbrauk. Virology—a endpoints. Am. J. Hyg. 27:493-497. 1938.
laboratory manual. Academic Press, Inc., San Diego, California 1992. 8. Villegas, P., and H. G. Purchase. Titration of biological suspensions. In:
4. Campbell, J. M, and J. B. Campbell. Laboratory mathematics: medical A laboratory manual for the isolation and identification of avian pathogens,
and biological applications, 3rd ed. The C. V. Mosby Co., St. Louis, Mo. 3rd ed. H. G. Purchase, L. H. Arp., C. H. Domermuth, and J. E. Pearson,
1984. eds. American Association of Avian Pathologists, Kennett Square, Penn. pp.
186-191. 1989.

221
47
SEROLOGIC PROCEDURES
Stephan G. Thayer and Charles W. Beard

SUMMARY. Avian serology consists of a combination of classical test methods, such as the agar-gel precipitin (AGP) test, the plate
agglutination test, the virus-neutralization (VN) test, and the hemagglutination-inhibition (HI), and the enzyme-linked immunosorbent assay
(ELISA). Most of the tests are relatively easy to perform, but quality control is essential.
The AGP test, also known as the agar-gel immunodiffusion test (AGID) or double immunodiffusion test (DID), is the simplest to set up,
requiring only positive and negative control sera, concentrated antigen, and appropriate agar. Antibodies to disease agents such as avian
influenza virus, hemorrhagic enteritis virus, and adenoviruses can be detected by this method.
Plate agglutination tests are used as screening tests for Salmonella pullorum, Salmonella gallinarum, other Salmonella spp., and
Mycoplasma spp. These tests use stained or unstained antigen mixed with either whole blood or serum. It is important to run confirmatory
tests because plate agglutination tests can give false positive and false negative reactions.
The VN test is more labor intensive and requires expertise in handling Eve viruses, cell culture techniques or embryo inoculation methods.
Expertise evaluating cytopathology or pathologic effects in developing embryos is required. Antibodies to viral diseases such as infectious
bursal disease, viral arthritis, some adenoviruses, infectious bronchitis, and avian encephalomyelitis virus can be assessed by these methods.
VN tests are often preferred as they tend to more closely reflect the in vivo neutralization of virus.
The HI test is used to titrate antibodies to avian diseases agents such as Newcastle disease virus, avian influenza virus (subtype-specific),
infectious bronchitis virus, adenovirus 127, and Mycoplasma. Hemagglutination is a naturally occurring activity with avian influenza viruses,
Newcastle disease viruses, and adenoviruses 127. Hemagglutination can be induced with infectious bronchitis viruses by pretreatment with
neuraminidase.
The ELISA can be used to perform titration of antibodies to a large number of avian viral and bacterial disease agents. Kits are available
commercially and offer the advantage of a single protocol to perform multiple tests on the same sample. The format is the 96-well
microplate, and the test can be semi automated using plate washers and computer-controlled microplate readers.

INTRODUCTION antibodies is the basis of the HI test. It is a convenient and


economical serologic tool that has been applied extensively to the
This chapter discusses the more commonly used serologic control of several avian diseases
* such as ND, IB, and
procedures applied to avian medicine. The discussion does not mycoplasmosis by measuring antibody response to vaccines and as
provide a precise outline to follow for all diseases, because the tests evidence of past infection.
vary considerably with the disease and with the laboratory system The basic components of the HI test are the hemagglutinating
(macro or micro) utilized. This chapter will provide information that (HA) antigen, the serum serially diluted in decreasing
can be used along with other specific serologic recommendations in concentrations, and the erythrocyte suspension. The tests may be
the disease chapters and in other publications to perform the desired done in tubes or in micro test plates. The constant-antigen diluted-
procedures in an accurate and reproducible manner. serum (β technique) is used much more than is the constant-serum
The following suggestions may be helpful in obtaining clean serum diluted-antigen (a technique).
in adequate amounts: The preparation of the HA antigen varies depending upon the
1) Use 1.5 ml snap-cap microfuge tubes collecting about 1.0 ml of laboratory and the disease agent. It can be as simple as
blood. The clot does not stick to the walls and yields about 0.5 ml of concentration of virus from NDV-laden egg fluids 48-72 hr after
clear serum after holding overnight at room temperature. The tubes inoculation or a concentrated suspension of Mycoplasma
can be microcentrifuged for about 30 seconds to help squeeze the microorganisms. A very satisfactory ND-antigen-preparation
clot and the serum be poured off into a separate clean microfuge technique has been described (6). When egg fluid is to be harvested,
tubes for storage at 4 C. The yield of 0.5 ml will be sufficient for the eggs should be chilled at 4 C for several hr to reduce the chance
most testing. of erythrocyte contamination of the fluid. The amount of antigen
2) Use silicone-treated glass tubes. They can be purchased or used in each well of the HI test varies from 8-10 HA units for ND,
treated with silicone in the laboratory (33). to 4 HA units for influenza, to 2-4 HA units for Mycoplasma. The
3) Never fill a tube to more than one fourth to one third of its dilution of the antigen suspension is needed to adjust the HA
capacity. activity to the desired number of HA units per volume used in the
4) After the tube is stoppered, lay it down at about a 5 degree angle test. If an antigen suspension is diluted by twofold transfers (0.5 ml
and allow the blood to clot. into 0.5 ml in tubes or 50 microliters into 50 microliters in a
5) Place samples horizontally in a 37 C incubator for several hours. microplate), resulting in dilutions of 1:2, 1:4, 1:8, and so on, and
6) Leave samples upright at room temperature or in the refrigerator complete agglutination occurs at the 1:512 dilution but not at the
(4 C) overnight. 1:1024 dilution, the suspension has an HA titer of 512 (a 1:512
Serum can then be carefully poured off into vials or pipetted into dilution of the antigen theoretically contains 1 HA unit). That
microliter serum storage plates (31). A modification of a filter-paper means that a 1:51 dilution of the suspension would result in 10 HA
blood-sampling method (10) makes that technique particularly units, a 1:256 dilution in 2 HA units, and so on. If more precision is
suited to micro serology for poultry. desired in determining the HA activity of an antigen suspension,
two sets of twofold dilutions can be made simultaneously: one
HEMAGGLUTINATION-INHIBITION (HI) TEST beginning with 1:10 and one with 1:15. The HA titer will be the
reciprocal of the highest dilution that exhibits HA activity in either
Avian viruses that agglutinate erythrocytes include Newcastle row of dilutions. It is always advisable to back-titrate the antigen
disease (NDV) virus, avian influenza (Al) virus, infectious suspension with the erythrocyte suspension used in the HI test to be
bronchitis (IBV) virus (after concentration and enzyme treatment), certain that the HA activity was actually as calculated. If 8 HA units
and adenovirus 127. Several Mycoplasma species are also capable were calculated to be in the final-use antigen suspension, a twofold
of hemagglutination. The inhibition of hemagglutination by specific (1 into 1) dilution (1:2) should contain 4 HA units; the next twofold
222
Chapter 47 Serologic Procedures

dilution (1:4) should contain 2 HA units; the next (1:8) should 1) Dilute the HA antigen in PBS (pH 7-7.2) based on the HA
contain 1 HA unit; and the next (1:16) should be HA negative. If 10 activity of the antigen to yield 10 HA units in 50 microliters; for
HA units were used, the 1:8 dilution should be HA-positive and the example. If the antigen titer is 1:2560, dilute 1:256. The inactivated
1:16 dilution HA negative. antigen described by Beard et al. (6) is stable and non-infectious.
Two basic variations exist in HI test procedures related to HA 2) Dispense the antigen-saline into round-bottomed microplates
antigen. In NDV and Mycoplasma Hi tests, the serum dilutions are using an 8 channel multichannel micropipette with disposable tips.
usually made in an antigen-saline mixture. In the influenza HI tests, Add 100 microliters to the first column and 50 microliters to the
the HA antigen is usually added to the diluted serum in an other columns except column 12. Column 12 receives 50 microliters
additional step. If the antigen-added method is used, the antigen of PBS and serves as an erythrocyte control. Automated pipetters
should be diluted so that the desired number of HA units occurs in are available to dispense reagents and to make dilutions in
the final volume of the diluted serum together with the added microplates.
antigen before erythrocytes are added. 3) If the sera are stored in microplates (28), 8 or 12 sera can be
The concentration of HA antigen used in the HI test undoubtedly transferred at one time using 8 or 12-channel micropipettes. Add 25
has an influence on the final test results, but slight differences in the microliters of sera to the first row of wells and mix resulting in a 1:5
number of HA units may induce minimal effects on the HI titer of dilution of serum in the first column of each plate. The sera are
the serum tested. Generally, increasing the antigen concentration diluted across each plate using the micropipette set at 50 microliter
results in decreased sensitivity, and decreasing the antigen volumes for mixing in the well and making transfers in the serial
concentration results in increased sensitivity. dilutions. This results in dilutions of 1:5, 1:10, 1:20, etc. ending
Many variables other than the concentration of HA antigen used with a final dilution of 1:2560 in well 10. Well 11 serves as an
can influence HI test results. These variables include the antigen control. It is always a good practice to place positive control
concentration of the erythrocyte suspension, the time between sera of known HI titer in specific locations on the test plates to
mixing the serum and antigen and the addition of erythrocytes, the indicate the amount of variation between tests. This positive control
temperature at which those mixtures are held, and the criteria used serum should be from a pool that will provide a long term supply to
in reading the test. Brugh et al. (11) examined several of the provide a consistent long term control that can be monitored versus
variables for the NDV HI test and recommended a standard holding time. Highly automated micropipettes and liquid-handling robotics
period before adding erythrocytes. When the holding temperature is are available for diluting of sera and addition of reagents.
37 C, the HA activity of the antigen must be stable at that An alternative dilution scheme can be used to provide 1:2, 1:4, 1:8
temperature because antigen degradation could falsely increase HI etc dilution series. Simply add 50 microliters of pre-titered antigen­
titers. saline to each well except row 12. Fifty microliters of test serum is
added to each well in the first column resulting in a 1:2 dilution.
Erythrocyte Suspension After the test serum is mixed, 50 microliters is passed to the next
Erythrocytes can be taken from a single chicken if experience has row, and so on, resulting in dilutions of 1:4, 1:8, and so on. It is not
shown that the donor is satisfactory; otherwise, a pool from a unusual to get some nonspecific HI results at the first one or two
minimum of three chickens is recommended (1). Chickens dilutions when this alternative method is used. The highest dilution
vaccinated against NDV can be used as donors, if necessary, achievable testing 8 sera per plate is 1:1024.
provided that careful attention is given to the erythrocyte-washing 4) Incubate all plates at room temperature for 20-30 min.
procedure. Turkeys are often used as donors when turkey serum is Microplates may be covered with individual lids or with plate
tested for HI activity, so that nonspecific reactions are reduced. sealing tape to avoid evaporation.
When turkey sera are tested, both hemagglutination and HI should 5) After the incubation, add 50 microliters of a 0.5% chicken
be performed using turkey erythrocytes. erythrocyte suspension to each well using a manual or automated
An anticoagulant such as 4% sodium citrate (one part to four parts multi-channel pipet. Gentle swirling of the cell suspension should
blood) or Alsever’s solution (equal volumes) should be in the be done during this step to aid in maintaining a uniform suspension
syringe into which the blood is drawn. After it is mixed gently, the of erythrocytes.
blood is transferred slowly to a large, conical centrifuge tube for 6) The plates are left at room temperature for 45 min. Negative
washing. An equal amount of phosphate-buffered saline (PBS) at serum and antigen controls should exhibit a matt of agglutinated
pH 7.0-7.2 is added and the suspension is centrifuged at 500 x g for erythrocytes. Erythrocyte controls should exhibit a distinct button of
5 min. The supernatant is poured off, and 20-30 volumes of PBS non-agglutinated erythrocytes. A reading mirror aids in reading the
are added to the packed cells. The cells are resuspended gently and results. Results should be recorded as soon as possible. Some
the centrifugation step repeated once more, again pouring off the workers suggest tilting the plates and observing for “tear dropping”
supernatant. The cells can then be used to prepare the 0.5% or "running buttons" to show that the erythrocytes are not
suspension based on volume by adding 0.5 ml of the packed cells to agglutinated.
100 ml of PBS at pH 7.0-7.2. Alternatively, once the erythrocytes 7) Results are numerically reported as the reciprocal of the highest
have been washed 3-4 times they can be adjusted to a 25% dilution of serum at which there was complete inhibition of
suspension in PBS and held for up to 4 days at 4 C. hemagglutination. This value (160 if 1:160 was the highest
Erythrocytes can also be stored in Alsever’s solution at a ratio of 1 inhibitory dilution) is not multiplied by the number of HA units
ml of packed cells to 15 ml of Alsever’s solution by resuspending used in the test, as was once suggested. A convenient method for
the cells in the solution and then holding them at 4 C. The cells can reporting HI results and calculating geometric mean titers has been
be used for up to 6 days if no hemolysis is observed. The cells are reported by Brugh (9).
centrifuged out of the Alsever’s solution, washed 3-4 times in PBS In laboratories where microtest equipment is not available, the HI
and diluted as described as needed for a test. tests can be satisfactorily performed in tubes by using the same
dilutions but passing 0.5 ml instead of 50 microliters. The basic
Test Procedures principles and the results between the macro- and micromethods are
Microtechniques are widely used and recommended for HI tests. comparable.
The techniques are convenient, economical, and reliable with a 10- The elution of the antigen from the agglutinated erythrocytes may
fold reduction in the quantity of reagents used. be erroneously interpreted as HI. The elution problem is often
β Procedure (Diluted-Serum Constant-Virus) encountered when viable or rapidly eluting ND viruses are used as
A typical β procedure NDV HI test can be performed as follows: HA antigen in laboratories that have high ambient temperatures.
223
Stephan G. Thayer and Charles W. Beard

Rapidly eluting viruses should not be utilized as the HA antigen. hemagglutination in the fluids containing the known-positive NDV
Unnecessary hazards of inadvertent cross-contamination in a antiserum. This is a rapid and reliable method of screening injected
multipurpose laboratory can be avoided by inactivating all HA embryos while they are still on the laboratory bench. If it is positive,
antigens. Adding formalin to achieve a final concentration of 0.1% all of the remaining fluid can be harvested for additional testing,
and incubating for 24 hr at 37 C is a satisfactory procedure for should that be desirable. This procedure can be used for influenza
NDV. Beta-propriolactone (BPL) at 0.1% can also be used. Careful viruses with polyvalent avian influenza positive antisera and known
initial handling of BPL is necessary as it is carcinogenic but negative sera. HA typing must be done using type-specific antisera.
hydrolyzes rapidly during the inactivation and becomes inactive.
Other instructions for performing the HI test with minor and major IMMUNODIFFUSION
differences are available from other sources (1, 2, 3, 7, 19). For Immunodiffusion techniques are frequently used in avian medicine
Infectious bronchitis virus HTs the principle differences are to demonstrate and analyze antigen-antibody reactions. The
Incubations are done at 4 C and serum dilutions are performed in techniques permit visualization of the antigen-antibody complexes
plain HI buffer instead of diluted antigen. Regardless of the as precipitates form when these two reactants co-mingle while
procedure used, the use of control sera of known titer is absolutely diffusing in a semisolid medium such as agar. Immunodiffusion is a
necessary to facilitate quality control and give assurance of precise immunological technique, yet costs are minimal and the
consistent performance. If those positive sera are provided by a procedures are simple.
reference laboratory, the sera will make possible comparisons The technique used most commonly in poultry diagnostic
between the different laboratories performing the tests. A laboratories is the two-dimensional double-diffusion procedure, in
comparison of wide-ranging HI results obtained by different which both antigen and antibodies diffuse toward each other from
laboratories varied markedly even when all used the same antigen separate wells in a layer of solidified agar in a petri dish or on a
suspension, which emphasized the need to include control sera of glass slide. This double-diffusion test is known by several names,
known titers (8). such as the agar-gel precipitin (AGP) test, the agar-gel
immunodiffusion (AGID) test, the double Immunodiffusion (DID)
a Procedure (Constant-Serum Diluted-Virus) test, and the Ouchterlony test.
The a procedure employs dilutions of the antigen and a constant The DID test has been described for various poultry diseases,
amount of serum. The serum to be tested is diluted 1:5 or to some including Marek’s disease (29), infectious bursa disease (TBD) (16),
other suitable dilution and substituted for the saline in the infectious bronchitis (36, 37), viral arthritis (23), avian
hemagglutination test. This requires a large volume of serum encephalomyelitis (AE) (20), avian influenza (Al) (5), ND (14),
compared to the β procedure and therefore is not practical in most fowl pox (17), mycoplasmosis (22), and others (21). Each particular
diagnostic situations. The virus dilutions and serum are mixed and application requires appropriate techniques of antigen preparation,
incubated at room temperature for 10 min before the erythrocytes buffer selection, and gel formulation. Therefore, one should first
are added. Incubation and reading of the tests are the same as in the become familiar with the principles of the reaction and specific
hemagglutination test. A hemagglutination test must be performed methodology for each (13, 18, 32).
concurrently for the virus-only titer and serum-virus titer to be The advantage of such a test system is simplicity and speed. With
compared. known reference reagents, a laboratory worker can identify either
Because the β procedure gives a better evaluation of the HI titer of infectious agents or antibodies. The visualization of precipitin lines
sera, the a procedure has been discontinued by most laboratories. that fuse with lines of identity from known reactants is a reliable
For a more detailed description of the a procedure, see Methods for diagnostic procedure that requires little effort and expense.
Examining Poultry Biologies and for Identifying and Quantifying Several methods are used for containing the gel medium. Plastic or
Avian Pathogens (4). glass petri dishes are frequently used. Wells are cut with
commercial or homemade cutters and the agar plugs aspirated (24).
HEMAGGLUTINATION (HA) TEST The usual pattern is six wells around a center well. Wells are 5.3
mm in diameter and spaced 2.4 mm apart. Approximately 15-17 ml
A useful application of the hemagglutination (HA) tests can be of agar in a 100-mm petri dish or 6 ml in a 60-mm petri dish or 2 ml
made during the isolation procedure for NDV or other in a 35-mm petri dish will result in the correct agar thickness (2.8
hemagglutinating viruses. The suspected material is injected into mm). The plug is aspirated with a small pipette attached to a
the allantoic cavity of embryonated eggs and incubated. A small vacuum flask. Sometimes the bottom of the wells must be sealed
amount of the allantoic fluid is removed after the desired period of with a small drop of melted medium to prevent the reagents from
incubation. Prechilling of the egg reduces chances of erythrocyte leaking under the gel instead of diffusing through it. Some workers
contamination during the fluid-sampling procedure. A rapid check use a commercially available triangular plastic plate for DID tests
for HA activity can be done by placing a drop of the allantoic fluid with built-in molds that result in either 5-mm or 7-mm well
on a slide along with a drop of 5% washed chicken erythrocytes. distances (Alpha Gamma Laboratories, Inc., Sierra Madre, Calif.).
Mix by light stirring or by gently rocking the slide. AA positive test The plates should be placed in a moisture chamber, such as a plastic
is indicated by hemagglutination and will have a granular bag or storage container with a snap-on lid that contains a wet paper
appearance. Quantitation of HA activity can be done by pipetting towel to avoid drying of the agar.
0.1 microliters of the allantoic fluid into a microtiter plate or in a A simple DID system can be prepared on ordinary frosted end
glass tube. After making twofold serial dilutions in saline, 0.1 ml of glass microscope slides. The wells can be labeled on the frosted end
a 0.5% suspension of washed chicken erythrocytes is added. of the slide. The slides should be placed in a humidity chamber for
Allantoic fluid from an uninoculated egg serves as a negative incubation. If the temperature in the laboratory is below 21 C, the
control. No hemagglutination should occur in this sample. If HA plates or slides should be held in a 26 C incubator.
activity is observed with the fluid from the injected egg, HI tests The pattern of wells must place each test serum or antigen adjacent
can be done in microtest plates using known ND-positive and ND- to a known-positive reagent. This will make the observation of a
negative chicken sera (preferably from specific-pathogen-free [SPF] continuous line of identity possible and will reveal weak positive
chickens) to determine if NDV is the cause of the hemagglutination. reactions by the slight bending of the end of the precipitin line near
After the dilutions of allantoic fluid are mixed with normal and the suspect well.
immune sera, an equal quantity of 0.5% erythrocytes is added. If the The most commonly used semisolid medium for immunodiffusion
suspect HA agent is NDV, the NDV antiserum will prevent is prepared with 0.7-1.0% pure or partially purified agar and 8.0%
224
Chapter 47 Serologic Procedures

NaCl. Such purity is an important consideration, because it can preferably be produced in SPF chickens from a plaque-purified
influence free diffusion of the reactants. Some bacteriologic-grade virus culture. Stocks of normal serum from SPF birds should be
agars have strongly charged chemical groupings that will bind kept available as control serum during test procedures.
certain antigens, which interferes with the formation of a visible Virus. Virus strains used for neutralization tests should have a high
precipitin band. Special agar noble has proven to perform well for a titer, should not have clumps of virus, and should be well adapted to
several test antigens. Agar may be pre-prepared and stored in 30-40 the host system used. The virus strains should be pure cultures,
ml aliquots in glass screw-capped centrifuge tubes at 4C. To preferably clone-purified, and free from bacteria, fungi, or
prepare the agar the tubes are placed in a boiling water bath until Mycoplasma. Stock viruses are best maintained in small aliquots in
molten. Plates must be prepared fresh on the day of the testing. glass-sealed ampules, screw-cap vials, or plastic cryovials stored at
Punching and aspiration of the wells must always be done just prior -60 C or below.
to loading. Any drying of the agar or wells will compromise the Diluents. The diluent used can be cell-culture medium or other
test. diluents known to be compatible with both the virus and host
The last important factor is reactant concentration. The location of system to be used.
the precipitin band between the antigen and antibody wells is
determined by the relative concentrations and diffusion rates of the β - Neutralization Procedure (Constant-Virus Diluted-Serum)
reactants. For practical purposes, the diffusion rates cannot be In this method, serial dilutions of the serum are tested against a
manipulated, although the reactant concentration must sometimes standard concentration of virus. This technique has certain
be adjusted before the precipitin bands will become visible. The advantages in that it uses small amounts of serum, is the preferred
reason for this is that if there is considerably more of one reactant test for low-titered viruses, and is more useful for demonstrating
than the other, the precipitin reaction may occur within or significant differences in neutralizing antibodies between acute and
immediately adjacent to the well that has the lower concentration, convalescent serum specimens collected from suspect flocks.
which gives apparently negative results. A quantal response in cell culture grown in 96 well microplates is
In addition to detecting antibodies in serum, the immunodiffusion selected as the indicator system in the following example. The 96-
test can be used to detect antibodies in yolk. Yolk is diluted with well cell-culture system can be utilized with the same automated
equal parts of PBS, shaken or mixed on a Vortex mixer for 30-40 dispenser system used for HI tests. Although the principle is the
sec, and centrifuged at 1000 x g for 30 min. The yellow, translucent same for all systems, reagent volumes differ among systems and
supernatant is used in the test. procedures.
Immunodiffusion test results can be read after several hours or 1) For the neutralization part of the test, make serial twofold or
several days, depending primarily on the concentrations of antigen fourfold serum dilutions in cell-culture medium in a microtest plate.
and antibody. Precipitin bands are best seen against a dark 2) Dilute the virus in cell-culture medium to contain the desired
background with the gel illuminated obliquely from the bottom. tissue-culture infective dose (TdD50) in 0.1 ml (100 TCID5o is
Illumination devices are usually homemade (15) but are also frequently used). The working dilution of virus is determined from
available commercially. A Model 653 microscope illuminator a previous titration where serial dilution of the stock virus is
(American Optical, Leica, Inc. Deerfield, Π1.) is very satisfactory performed in a microplate. The last well showing destruction of the
for this purpose. cell monolayer is the endpoint . Back-titration the virus with each
A specific, positive result is recorded when the precipitin line batch of neutralization tests is used to confirm that 100 TCID50 of
between the known-positive control wells is continuous with the virus was used (see Chapter 46 on titration of biological
line between the antigen and the test well. It is important that each suspensions).
test well be positioned adjacent to a positive control well. Slight 3) In a series of tubes or in a microtest plate, thoroughly mix 0.1 ml
reactions are observed when the precipitin line is bent at the test of each of the serum dilutions with the pre-diluted virus containing
well end and partial identity is seen as spurs. Crossed lines are 100 TCID50 in 0.1 ml. For a virus control, mix the virus dilution
interpreted as non-identity or negative. with known normal serum or cell-culture medium.
The immunodiffusion procedure offers many advantages, but it 4) Incubate the test set of tubes and the virus control tube for 1 hr at
lacks the level of sensitivity offered by many other tests. Semi­ 25 C, unless another time or temperature is specified.
quantitation may be achieved by testing serial dilutions of a known 5) For assay of residual virus, inoculate 0.2 ml of each serum-virus
positive sample. mixture into each of five monolayer cultures containing fresh
medium and incubate them at 37 C. Use separate pipettes for each
VIRUS-NEUTRALIZATION (VN) TEST dilution or inoculate the lowest serum dilution first. In the β
procedure, this is necessary because active virus is in highest
Serologic tests using the neutralization of virus for quantitative concentration at the highest serum dilution and may be carried
measurement of antibody are used frequently in diagnostic and forward to the next lower serum dilution in the pipette.
research laboratories. Used in conjunction with known antisera, 6) Examine the cultures at an appropriate reading time for the test
they are useful in identifying unknown viruses and differentiating virus and score the cultures for cytopathic effect as positive or
between viruses. The test has two parts. In the neutralization part, negative.
the virus (at the correct dilution) is mixed with the serum (also at When the indicator system is based on an enumerative response in
the correct dilution) in a test tube. The virus and serum are mixed cell culture or a quantal response in chicken embryos, the
and incubated together at a standard temperature for a given length neutralization parts (steps 1^4) are the same. The assay of residual
of time. In the second part, the residual unneutralized virus is virus will vary with the system and the response, but it will be the
assayed in a suitable indicator system (see Chapter 46 on Titration same as that used to determine the titer of the virus (see Chapter 46
of Biological Suspensions). The general procedure of the test is as on Titration of Biological Suspensions).
follows (modifications are given where necessary for different host
systems). Calculation of Neutralizing Titer for β Method
Sera. Sera for neutralization tests should be sterile (they may be When the residual virus is assayed with a quantal response, the
membrane-filtered), free of any chemical preservatives (phenol, 50% endpoint of neutralization is calculated by the method of Reed
formalin, and so on), and heated at 56 C for 30 min to destroy heat- and Muench (25) (Table 46.1) or Spearman-Karber (12) as
labile nonspecific virus-inhibitory substances. Antiserum to be used explained in Chapter 46. The neutralizing titer or mean protective
as a standard or for the identification of unknown viruses should dose (PD50) is calculated from this endpoint.
225
Stephan G. Thayer and Charles W. Beard

For an enumerative response, the endpoint is the dilution of serum 86-50


that neutralizes 50% (80% or 90% in some systems) of the virus.
The endpoint can be interpolated in the same way that the 86-33
proportionate distance is obtained. Interpolation can also be done
graphically, or by other statistical methods. 36

a Neutralization Procedure (Constant-Serum Diluted-Virus) 53


In this method, serial dilutions of virus are mixed with a standard
dilution of serum (ideally, undiluted serum is used). The serum­ = 0.67.
virus mixtures are incubated and then assayed for residual virus as
above, in any host, usually by quantal response. Calculate the log 10 exponent that is the 50% neutralization
A quantal response in chicken embryos is selected as the indicator endpoint or the PD50: reciprocal of 50% neutralization endpoint =
system in the following example. proportionate distance x log of the serum dilution factor + log of the
1) For the neutralization part, prepare decimal dilutions of the virus lower dilution used to calculate the proportionate distance
and arrange them in a rack as shown in Table 47.2, row 1. = (0.67x0.6) = (1.2)
2) Set up rows of small sterile test tubes for negative (row 2) and = 0.4+1.2
positive (row 3) control sera, and a row for each unknown serum = 1.6
(rows 4 and 5). = 40 (antilog of 101.6)
3) Into each of the tubes in row 2, pipette 0.4 ml of sterile serum 50% neutralization endpoint or PD50 = 1:40.
known to be free of antibody against the agent being studied. In the
absence of such serum, sterile diluent can be used. This row will Calculation of Neutralizing Index for a Method
serve as the negative serum or virus control. Into each of the tubes When the residual virus is assayed by a quantal response, the
in row 3, pipette 0.4 ml of sterile serum known to have antibody endpoint for each serum can be calculated by the method of Reed
against the agent being studied. This row will serve as the positive and Muench or of Spearman-Karber. A hypothetical test result is
control. To each of the tubes in the other rows, add 0.4 ml of the given in Table 47.3. The neutralization index (NI) of the serum is
serum being tested. the difference between the log titer of the virus control (negative
4) Transfer 0.4 ml of the virus dilution from row 1, tube 8, to the serum) and the log titer of the serum-virus mixture. It has no units.
eighth tube in rows 2, 3, 4, and 5, and discard any virus remaining Thus, in the example in Table 47.3:
in the pipette. With the same pipette, transfer 0.4 ml of the virus
dilution from row 1, tube 7, to the seventh tube in the other rows. titer of virus control (or negative serum-virus mixture) = 6.5,
Continue until all tubes have virus. When the titer of the virus is titer of positive serum-virus mixture = 2.5, and
known, three to five dilutions in the control group usually suffice to difference (NI) = 4.0.
bracket the endpoint in subsequent tests. Frequently, the unknown
serum must be tested over a broad range of virus dilutions to ensure The NI is sometimes expressed arithmetically (i.e., in the above
detection of the endpoint. example, 10,000). In this instance, the NI represents the ratio of
5) Mix virus and serum thoroughly by swirling each tube arithmetic endpoint titers of the virus control and positive serum­
individually or by shaking the tube rack vigorously. Allow the virus control. It is very important to distinguish between the log and
mixture to stand at 25 C for 1 hr unless another time or temperature the arithmetic figures, particularly when the NI is low.
is specified.
6) For the assay of residual virus, start inoculating with the least­ It is common practice for laboratories to receive a pooled serum
concentrated dilution of virus in the unknown serum group and sample for antibody assay that is composed of equal parts of sera
inoculate each of at least five embryonating eggs with 0.1 ml by the from several chickens. Such pooled samples can lead to false
appropriate route. Proceed to the next dilution, using the same conclusions on the antibody status of the chickens represented by
needle and syringe. Repeat this until all virus-serum mixtures have the pooled sample. In the above example in which the NI was 4.0,
been inoculated. If more than one serum is being tested, use a supposing that the pooled sample came from 10 chickens, it is
separate needle and syringe for each serum, and inoculate each of possible that 9 of the 10 chickens had no antibody levels whereas 1
these mixtures as above. The virus control mixture should be of the 10 had an NI of 5.0.
inoculated last, using a separate needle and syringe. For very critical The result of the pool would be an NI of approximately 4.0. The
work, use a separate needle and syringe for each serum-virus limited information from pooled samples should be weighed against
mixture. the practicality of testing individual sera.
7) Seal the eggs by appropriate means and return them to the
incubator for the time specified for the virus in use. Candle the eggs AGGLUTINATION
18-24 hr later and discard all eggs containing dead embryos. The clumping or agglutination of bacteria by specific antibodies in
Candle eggs daily and examine dead embryos for lesions, if desired. whole blood and in serum was one of the first serologic procedures
At the end of the experimental period, the remaining live eggs can to be used widely in the control of poultry diseases.
be examined for lesions, if desired. Record the results. Agglutination tests are relied on for diseases such as infectious
coryza, salmonellosis (including pullorum and fowl typhoid), and
The PD50 can be calculated from the example given in Table 47.1 mycoplasmosis. The tests may be done on glass or porcelain plates
by the following formulas: (serum plate test), in test tubes (tube test), or in microplates. Refer
to the chapters on the bacterial diseases in this manual for details of
Percent infected at dilution next above procedures and antigens used for the specific tests for a particular
Proportionate = disease.
distance 50%________ -__________ 50%_______ The serum plate test is frequently used as an initial screening test.
percent infected percent infected If a serum agglutinates the specific stained antigen in the plate test,
at dilution next - at next dilution the serum may then be serially diluted and tested at decreasing
above 50% below 50% concentrations for its ability to agglutinate the antigen in tubes or in
microplates. The agglutination titer is reported as the reciprocal of
226
Chapter 47 Serologic Procedures

the highest dilution of serum at which complete agglutination Once titers for individual serum samples are calculated the titers
occurs. For a small number of serum samples, the samples are are placed into titer groups according to a range of titers assigned to
sometimes diluted and tested by only the serum plate method. The that group, that is, titer group 0 could be all titers between 0 and
application of microtest methodology to the agglutination test for 1500, titer group 1 would be all titers between 1501 and 2000, and
salmonellae (35) and for Mycoplasma has drastically reduced the so on. Histograms are created based on the number of sera in each
expense and time associated with the agglutination test. titer group. The histogram is then used to visualize the distribution
of titers within the group of sera tested.
ENZYME-LINKED IMMUNOSORBENT ASSAY (ELISA)
Sample Size
The ELISA has become the principle serologic assay used by most The sample size required for adequate representation of the
laboratories for serologic testing. The ELISA is based upon the serologic status of a flock increases with the size of the flock.
single dilution antibody titer determination method of Snyder and However, a point of diminishing benefit exists for this approach
Marquardt (26, 27, 28). This method permits the development of when testing broiler flocks. A minimum of 20 samples per flock
flock profiles both on a single bleed approach or on one based on should be tested. Thirty sera per flock is suggested for breeder and
sampling over time. The use of a common protocol for most of the ELISA PROFILE
tests streamlines laboratory throughput. The application of the 10 sera 30 sera
microcomputer to drive the plate reader and process the data
simplifies information management. Additional benefit can be
gained through interfacing the microcomputer for direct input into a
laboratory information management system or company data
warehouse.

Ready to use kits for antibody tests to most of the common avian I lb
bacterial and viral pathogens are commercially available. These
include but are not limited to NDV, IBV, IBD virus, avian reovirus,
jIjuuuijj 1
β K A A k b * A * *.$> «V A t »A « ♦ >6*

Fig 47.1 liter group Titer group


Fig. 47.1
Mycoplasma gallisepticum, Mycoplasma synoviae, Pasteurella
multocida, AE virus, lymphoid leukosis viral antigen and antibody, layer flocks. Sample sizes less than 10 are inadequate, as the
infectious laryngotracheitis virus, avian influenza virus, chicken histogram representing these groups lacks the data points necessary
anemia virus, hemorrhagic enteritis virus, Bordetella avium, and for even marginal representation and interpretation. This is
Salmonella enteritidis. demonstrated in Fig. 47.1. Profile 1 represents a select group of 10
sera from the group of 30 sera seen in profile 2. The character of
Titer Calculation profile 1 is very different from that of profile 2 even though the sera
were from the same flock.
Titer calculations can be based simply on corrected absorbance
value where the mean negative control is subtracted from the test T roubleshooting
sample absorbance. A second method is to take the calculations one Pipetting. The ELISA is a very sensitive test and is susceptible to
step further in the calculation of the sample-positive ratio (S/P). pipetting errors. The most common pipetting errors are due to
The sample absorbance and the mean positive control absorbance improper filling because of loose-fitting pipette tips. This is
are both corrected for background by subtracting the mean negative especially true when using multichannel pipettes. The practice of
control absorbance from each, thus: loading those tips directly from the tip rack without checking fit
will cause gross volumetric delivery errors. Additionally, pipettes
Sample absorbance - NCx should be periodically calibrated for proper delivery.
S/P^_____________________ Plate Washing. Improper washing of the plates can cause
PCx - NCx variability that can be difficult to assess. Contamination of the
upper portions of wells, which may not be washed efficiently, can
where also introduce variability into the test. Careful attention to proper
NCx = Mean absorbance of negative control serum, and delivery and aspiration of wash material is essential for consistency.
PCx = Mean absorbance of positive control serum. Check pipettes and automatic plate washers frequently for proper
volume, rate of aspiration, and thorough aspiration of reagents and
Commercial ELISA systems for poultry serology typically take the wash solutions. Tapping plates on absorbent material after each
titer calculations to the next step by using the double regression wash step is practiced by many labs as a means of eliminating any
approach. The second and final step in this method is the residual wash. Four to five washes after aspiration of each reagent is
application of a linear regression formula to calculate the ELISA recommended. Some manufacturers recommend soaking steps as
titer using the single sample method rather than calculating the titer part of the wash procedure. These help to elute reagents that might
based upon a serial dilution. The linear regression formula defines not be thoroughly washed out of the system using rapid sequential
the relationship between log 10 of the S/P ratio of a single serum washes. The manufacturer’s recommendations should always be
dilution and the log 10 of the observed antibody titers. This typically followed.
takes the form: Bubbles and Plate Surface Contamination. Processing can
sometimes induce small bubbles, which can be easily overlooked.
log 10 titer = slope x [loglO(S/P)] + y intercept Be sure to check wells for bubbles and eliminate them prior to
titer = antilog (slope x [loglO(S/P)] + y intercept). reading the plates. Make it a practice to carefully wipe the bottom
of the plate with a clean tissue before reading. Any moisture or
The S/P ratio is central to this calculation. The actual values used contamination on the bottom of the wells will affect absorbance
for the slope and the y intercept are unique to the specific system. values.

227
Stephan G. Thayer and Charles W. Beard

Commercial Kit Reagents and Quality Control. Kits and Correlation of ELISA serology with conventional HI and VN
reagents should always be stored at 4 C or according to the serology can be done with adequate sample sizes comparing ELISA
manufacturer’s recommendations. Always allow reagents to warm geometric mean titers with geometric mean titers of the HI or VN
to room temperature prior to use. Reagents from different lots of the test. There is veiy poor if any correlation based on single sample
same test should not be used interchangeably. For the purpose of comparisons.
quality control it is always helpful to maintain records of kit lot
numbers and positive and negative control values for each test type. General Comments.
In addition, in-house positive controls can be developed and used to Interpretation of ELISA titers should only be done on a flock basis
give another measure of quality control. It is also a good quality with sample sizes of at least 20 sera per broiler flock. Thirty
control practice to periodically submit groups of previously titered samples per flock are preferred on breeder and layer flocks (30).
sera through the lab to monitor general performance. This can be Mycoplasma ELISA tests should only be used for screening, and the
especially helpful when training new technicians. results must be confirmed or ruled out by HI serology, culture, or
polymerase chain reaction (PCR).
Correlation with HI and VN Tests

Table 47.1. Data for the calculation of the 50% endpoint of a neutralization test by the method of Reed and Muench (25).

Dilution of serum Response Accumulated values


Infectivity Ratio Percent
Numerical Log Infected Not infected Infected Not infected
ratioA infected/Total infected
1:4 10-0.6 5/5 5 0 11 4 0 11/11 100
1:16 10-1.2 4/5 4 1 6 1 6/7 86
1:64 10-1/8 2/5 2 3 2 4 2/6 33
1:256 10-2.4 0/5 0 5 0 9 1 r 0/9 0
A Number infected/number inoculated (experimental number).

Table 47.2. Test procedure using the constant-serum diluted-virus (a) method.

Tuhenn
Row no. Contents 1 2 3 4 5 6 7 8
1 Virus dilution 10-1 10-2 10-3 10-4 10-5 10-6 10-7 10-8
Amount (ml) 2.0 2.0 2.0 2.0 2.0 2.0 2.0 2.0
2 Negative Serum (ml) 0.4A 0.4A 0.4A 0.4A 0.4 0.4 0.4 0.4
3 Positive Serum (ml) 0.4 0.4 0.4 0.4 0.4B 0.4B 0.4B 0.4B
4 Unknown serumC (ml) 0.4 0.4 0.4 0.4 0.4B 0.4B 0.4B 0.4B
5 Unknown serumC
A Assuming the virus has an endpoint of 10-6 or 10-7, these can be omitted to conserve serum and embryos.
B Assuming the serum neutralizes 4 logs of virus and the virus has an endpoint of 10-6 or 10-7, these can
be omitted to conserve serum and embryos.
C An example of unknown serum might be acute and convalescent serum.

Table 47.3. Example of virus-neutralization results. A

Virus dilution (loglO) B

-1 -2 -3 -4 -5 -6 -7 -8 ID50C NI
Negative serum (virus control) 5/5 2 4/5 1/5 0/5 6.5 C

Positive serum 5/5 5/5 0/5 2.5 4.0


Unknown serum 5/5 3/5 0/5 0/5 4.2 2.3
Unknown serum 2/5 0/5 0/5 0/5 <2.8 >3.7
Unknown serum 5/5 5/5 5/5 4/5 >6.0 <0.5

A Abbreviations: ID50 = mean infectious dose, NI = neutralization index.


B Values in body of table are number dead/number inoculated.
C Titer calculated by method of Reed and Muench

228
Chapter 47 Serologic Procedures

REFERENCES 19. King, D. J. A comparison of infectious bronchitis virus


hemagglutination-inhibition test procedures. Avian Dis. 32:335-341. 1988.
1. Alexander, D. J., W. H. Allan, P. M Biggs, C. D. Bracewell, J. H. 20. Lukert, P. D., and R. B. Davis. An antigen used in the agar-gel
Darbyshire, P. S. Dawson, A. H. Harris, F. T. W. Jordan, I. MacPherson, J. precipitin reaction to detect avian encephalomyelitis virus antibodies. Avian
B. McFerran, C. J. Randall, J. C. Stuart, O. Swarbrick, and G. P. Wilding. A Dis. 15:935-938. 1971.
standard technique for hemagglutination inhibition tests for antibodies to 21. McFerran, J. Β., B. Adair, and T. J. Connor. Adenoviral antigens
avian infectious bronchitis virus. Vet. Rec. 113:64. 1983. (CELO, QBV, GAL). Am. J. Vet. Res. 36(2):527-529. 1975.
2. Allan, W. H., and R. E. Gough. A standard hemagglutination inhibition 22. Nonomura, I., and H. W. Yoder, Jr. Identification of avian Mycoplasma
test for Newcastle disease. 1. A comparison of macro and micro methods. isolates by the agar-gel precipitin test. Avian Dis. 21:370-381. 1977.
Vet. Rec. 95:120-123. 1974. 23. Olson, N. O., and R. Weiss. Similarity between arthritis virus and
3. Allen, W. H., J. E. Lancaster, and B. Toth. Newcastle disease vaccines. Fahey-Crawley virus. Avian Dis. 16:535-540. 1972.
Their production and use. FAO Animal Production and Health Series, No. 24. Ranck, F. M., Jr. An easy method for making cutters to use in the agar­
10. Food and Agriculture Organization of the United Nations, Rome. 1978. gel diffusion technique. Avian Dis. 17:870-873. 1973.
4. Anonymous. Methods for examining poultry biologies and for 25. Reed, L. J., and H. Muench. A simple method for estimating fifty
identifying and quantifying avian pathogens. National Academy of Sciences, percent endpoints. Am. J. Hyg. 27:493-497. 1938.
Washington, D C. pp. 83-87. 1971. 26. Snyder, D. B., W. W. Marquardt, and E. Russek. Rapid serological
5. Beard, C. W. Demonstration of type-specific influenza antibody in profiling by enzymelinked immunosorbent assay. Π. Comparison of
mammalian and avian sera by immuno-diffusion. Bull. W.H.O. 42:779-785. computational methods for measuring antibody titer in a single serum
1970. dilution. Avian Dis. 27:474-484. 1983.
6. Beard, C. W., S. R. Hopkins, and J. Hammond. Preparation of Newcastle 27. Snyder, D. B., W. W. Marquardt, E. T. Mallinson, and E. Russek. Rapid
disease virus hemagglutination-inhibition test antigen. Avian Dis. 19:692- serological profiling by enzymelinked immunosorbent assay. I.
699. 1975. Measurement of antibody activity titer against Newcastle disease virus in a
7. Beard, C. W., and W. J. Wilkes. A simple and rapid Micro-test procedure single serum dilution. Avian Dis. 27:161-170. 1983.
for determining Newcastle hemagglutination-inhibition (HI) antibody titers. 28. Snyder, D. B., W. W. Marquardt, E. T. Mallinson, P. K. Savage, and D.
In: Proceedings of the 77th Annual Meeting U.S. Animal Health C. Allen. Rapid serological profiling using enzyme linked immunosorbent
Association, St. Louis, Mo. pp. 596-600. 1973. assay. ΙΠ. Simultaneous measurements of antibody titers to infectious
8. Beard, C. W., and W. J. Wilkes. A comparison of Newcastle disease bronchitis, infectious bursal disease, and Newcastle disease viruses in a
hemagglutination-inhibition test results from diagnostic laboratories in the single serum dilution. Avian Dis. 28:12-24. 1984.
southeastern United States. Avian Dis. 29:1048-1056. 1985. 29. Stone, H. A., and E. A. Holly. Reliability of the agar-gel precipitin test
9. Brugh, M A., Jr. A simple method for recording and analyzing in Marek’s disease studies. Avian Dis. 15:939-945. 1971.
serological data. Avian Dis. 22:362-365. 1978. 30. Thayer, S. G., P. Villegas, and O. J. Fletcher. Comparison of two
10. Brugh, M A., Jr., and C. W. Beard. Collection and processing of blood commercial enzyme-linked immunosorbent assays and conventional
samples dried on paper for microassay of Newcastle disease virus and avian methods for avian serology. Avian Dis. 31:120-124. 1987.
influenza virus antibodies. Am. J. Vet. Res. 41:1495-1498. 1980. 31. Whittemore, A. D., and J. E. Williams. Microsystem for collecting and
11. Brugh, M A., Jr., C. W. Beard, and W. J. Wilkes. The influence of test shipping diagnostic sera. Appl. Microbiol. 24:671-672. 1972.
conditions on Newcastle disease hemagglutination-inhibition titers. Avian 32. Williams, C. A., and M W. Chase. Methods in immunology and
Dis. 22:320-328. 1978. immunochemistry. Academic Press, New York. 3:103-374. 1971.
12. Cottral, G. E., ed. Serology. In: Manual of standardized methods for 33. Williams, J. E. Collection of avian blood samples in microtest plates.
veterinary microbiology. Cornell University Press, Ithaca, N.Y. pp. 60-93. Avian Dis. 17:445-449. 1973.
1978. 34. Williams, J. E. Microtest methodology. In: Isolation and identification
13. Crowle, A. J. Immunodiffusion. Academic Press, New York. 1973. of avian pathogens, 2nd ed. S. B. Hitchner, C. H. Domermuth, H. G.
14. Gelb, J., Jr., and C. G. Cianci. Detergent-treated Newcastle disease virus Purchase, and J. E. Williams, eds. American Association of Avian
as an agar gel precipitin test antigen. Poult. Sci. 66:845-853. 1987. Pathologists, College Station, Tex. pp. 136-140. 1980.
15. Glazier, R. M A simple apparatus for dark ground illumination of 35. Williams, J. E., and A. D. Whittemore. Serological diagnosis of
precipitin lines. J. Biol. Photogr. Assoc. 34:51-52. 1966. pullorum disease with the microagglutination system. Appl. Microbiol.
16. Hirai, K., S. Shimakura, and M Hirose. Immunodiffusion reaction to 21:394-399. 1971.
avian infectious bursal virus. Avian Dis. 16:961-964. 1972. 36. Witter, R. L. The diagnosis of infectious bronchitis of chickens by the
17. Jordan, F. T. W., and R. Chubb. The agar gel diffusion technique in the agar gel precipitin test. Avian Dis. 6:478-492. 1962.
diagnosis of infectious laryngotracheitis (ILT) and its differentiation from 37. Woemle, H. The use of the agar gel diffusion technique in the
fowl pox. Res. Vet. Sci. 3:245-255. 1962. identification of certain avian virus diseases. Veterinarian 4:17-28. 1966.
18. Kabat, E. A., and Μ M Mayer. Experimental immunochemistry.
Charles C. Thomas, Springfield, Π1. 1964.

229
48
MOLECULAR IDENTIFICATION PROCEDURES
Daral J. Jackwood and Mark W. Jackwood

SUMMARY. Molecular identification of avian pathogens relies on the detection of nucleic acid (either RNA or DNA) unique to that
pathogen. In addition, analysis of nucleic acid sequences is being used to identify the species, subspecies, type, subtype, serotype, pathotype,
and in some cases individual strains of the disease-causing agent. Molecular diagnostic tests are based on techniques such as restriction
fragment length polymorphism, hybridization with nucleic acid probes, the polymerase chain reaction, random amplified polymorphic DNA,
and nucleic acid sequencing. Knowledge of these techniques is necessary to understand and interpret the results of molecular diagnostic tests
that are based on that technology. The information presented here is general in nature and the reader is referred to individual chapters for
agent specific molecular identification and typing tests.

INTRODUCTION also available. Most kits use a phenol-based extraction followed by


precipitation of the nucleic acid or a lysis procedure followed by
DNA consists of adenine (A), guanine (G), cytosine (C) and binding of nucleic acids to a silica-based support, usually in a
thymine (T) bases, the order of which makes up the genetic code. column or membrane format. After washing, the nucleic acid is
The bases are linked together by a sugar (deoxyribose) and eluted from the silica-based support in the appropriate buffer.
phosphate backbone, which makes up the structure of a single Some of the DNA extraction methods described for identification
strand of DNA. Hydrogen bonds between bases (A pairs with T and of avian pathogens include QIAamp DNA extraction kit (Qiagen
G pairs with C) hold two strands of DNA together (5). Inc., Valencia, CA) and DNAzol reagent (Invitrogen, Carlsbad,
Like DNA, RNA contains bases, except that instead of T RNA CA). The most common reagent used to extract RNA from cells,
contains uracil (U). RNA is usually single stranded and is not as tissues and infectious agents is TRIzol (Invitrogen, Carlsbad, CA).
stable as DNA. RNA has a ribose and phosphate backbone whereas TRIzol is a phenol and guanidine isothiocyanate solution that
DNA contains a deoxyribose sugar. Messenger RNA acts as a preserves RNA during lysis of cells and infectious agents. The RNA
carrier of genetic information from DNA for the synthesis of is extracted from the TRIzol mixture with chloroform and purified
proteins (5). by precipitation with isopropyl alcohol according to the
The genetic code, or the actual sequence of bases in an organism’s manufacturer’s recommendation. Finally, some kits offer
genome, is unique for all living things. Thus, once the genetic code purification of both DNA and RNA (QIAamp MinElute kits,
is ascertained, that information can be used to specifically identify a Qiagen, Valencia, CA).
poultry pathogen by the presence of its genome.
RESTRICTION FRAGMENT LENGTH POLYMORPHISM
SAMPLE COLLECTION AND STORAGE (RFLP)

DNA and especially RNA are extremely labile and care must be This technique uses restriction enzymes (REs) to determine if two
taken to preserve the nucleic acid when collecting clinical samples pieces of DNA are similar. REs cut double-stranded DNA at
for molecular diagnostic procedures. Most clinical samples specific sequences called recognition sites. The DNA is digested
containing infectious agents, including tissues, blood, swabs from (cut) with a particular enzyme and the resulting strands are
the upper-respiratory tract and intestinal tract, and even bone, can separated by electrophoresis according to their size. If the sizes of
be used for molecular diagnostic procedures. Fresh samples should the DNA strands are not identical, the pattern of bands on the gel
be kept cold or frozen to prevent lysis of the sample and subsequent will not match and it can be concluded that the sequences of the two
destruction of the nucleic acids by RNases and DNases. DNA strands are different at the recognition sites. If the patterns of
Alternatively, clinical samples can be mixed with buffered phenol the bands do match, the sequences are considered to be similar but
(pH 7.4 for DNA, and pH 4.5 for RNA), which inactivates RNases additional testing is needed to verify that the two pieces of DNA are
and DNases and preserves the nucleic acids. An easy method of identical because REs only recognize relatively short nucleotide
preserving and transporting clinical samples for molecular sequences.
diagnostic procedures is the use of Finders Technology Associates Genomic DNA isolated from an unknown disease-causing agent
(FTA) cards (Whatman, Middlesex, UK). The FTA cards are filter can be used to identify a specific bacterium, Mycoplasma or virus,
paper cards that inactivate infectious agents and stabilize nucleic by comparing the RFLP pattern with reference standards. If the
acids. Clinical samples are applied directly to the card and allowed disease-causing agent to be detected contains an RNA genome,
to dry. The cards are easily transported without refrigeration using which is the case with many viruses, the RNA can be converted to
the appropriate permits for transportation of biological samples. DNA using the technique of reverse transcription (RT). When RT
Nucleic acid is purified from a punch taken from the FTA card. is followed by amplification of the DNA by the polymerase chain
reaction (PCR), sufficient DNA can be obtained to generate RFLP
NUCLEIC ACID EXTRACTION AND PURIFICATION data (see the section on PCR). This technique is commonly referred
to as RTZPCR-RFLP.
Nucleic acids (DNA and RNA) can be extracted from almost any In some cases, identification of a disease-causing agent is possible
clinical or laboratory sample. DNA is extracted by a variety of by simply looking for the presence or absence of an RE recognition
methods. One of the easiest methods of extracting DNA from whole site or a combination of RE recognition sites. The assay is
cells and bacteria (including Mycoplasma) is the boiling method. conducted exactly like an RFLP except that the sizes of the
Briefly, cells are pelleted by centrifugation, resuspended in a Tris- resulting DNA fragments are insignificant. The result is positive if
EDTA buffer (pH 7.4) and heated to 100 C for 10 min to lyse the the DNA is cut at any location. Thus, any reduction in size of the
cells. The material can be used directly in the PCR test or DNA can original DNA molecule is indicative of the presence of an RE
be further purified by precipitation using sodium acetate (0.3M, pH recognition site. The technique is usually conducted on short pieces
5.2 final concentration) and an equal volume of cold 100% ethanol of DNA that were generated using RT/PCR and has been commonly
(E1OH). The DNA is then resuspended in the appropriate buffer. called RT/PCR-RE.
Numerous kits for extraction of DNA from a variety of sources are
230
Chapter 48 Molecular Identification Procedures

HYBRIDIZATION AND NUCLEIC ACID PROBES POLYMERASE CHAIN REACTION (PCR)

The basic mechanism behind nucleic acid probe-based diagnostic One of the most significant developments in molecular biology,
tests is hybridization. It involves base pairing (hydrogen bonding) and the basis of numerous new diagnostic tests for poultry disease­
of complementary sequences (G with C and A with T, or U) of causing agents is PCR. Stated simply, PCR amplifies very small
DNA or RNA (3). quantities of DNA to detectable levels (1). The reaction uses Taq
In a hybridization assay, the nucleic acid must be single stranded. polymerase, a DNA polymerase that is stable at high temperatures,
Denaturation of the nucleic acid into single strands is usually to amplify the genome of the disease-causing agent. DNA
accomplished by heating or chemical treatment of the sample. Then polymerase is the enzyme that synthesizes new strands of DNA. It
the denatured nucleic acid is bound to a solid support (nitrocellulose is important that it survive high temperatures because the first step
or nylon filter). Next, a single-stranded nucleic acid probe is mixed in the PCR is heating the reaction to 95 C. Typically, there are
with the nucleic acid bound to the filter, utilizing conditions that three steps in the PCR, which are repeated 30-40 times. These steps
favor hybridization (3). are denaturation at 95 C, primer hybridization at 37-56 C, and
Conditions that can influence whether two strands of nucleic acid polymerization at 72 C. A computer-controlled temperature block
will hybridize, are known as stringency (3). High stringency can be programmed to cycle repeatedly between these different
conditions require that the base pairing between two complementary temperatures, making the PCR automated.
sequences be an exact match. Low stringency conditions will allow Generally, the DNA to be amplified is flanked by a pair of
for some base pair mismatches between the two nucleic acid synthetic primers (short pieces of DNA) that are specific
sequences. The reaction conditions that affect hybridization are the (complementary) for the template DNA to be amplified. The target
type of nucleic acid involved in the hybridization (base pairing DNA is denatured by heating, and the primers are allowed to
between two strands of DNA is not as strong as DNA-RNA base hybridize to the DNA by lowering the temperature of the sample.
pairing), the length of the nucleic acid sequence (a greater number When the polymerization step is completed, the sequence between
of complementary base pairs will hybridize more strongly than a the primers is copied, producing twice as much DNA as was
lesser number), and the base composition of the nucleic acids (G-C originally present in the sample. By repeating the denaturation,
base pairing is stronger than A-T or A-U base pairing). The hybridization, and polymerization steps many times, large amounts
temperature of the reaction mixture and the ionic strength of the of the target DNA are obtained. When reverse transcriptase, which
hybridization buffer (both affect hydrogen bonding between bases) synthesizes DNA from an RNA template, is used in the first step of
also affect hybridization. the reaction, messenger RNA, ribosomal RNA, and the genome of
Nucleic acid probes exist for almost all pathogens important in RNA viruses can also be amplified by PCR.
veterinary medicine. They are available from research laboratories, Diagnostic tests utilize PCR to amplify the nucleic acid of disease­
where they are used as experimental tools, or from commercial causing agents that are present in small numbers, thereby increasing
sources. In some cases, these commercial sources have the sensitivity of the test. Then, the amplified DNA can be further
incorporated probes into diagnostic tests. A nucleic acid probe is a analyzed with a specific nucleic acid probe, or by RE analysis,
single-stranded fragment of DNA or RNA that has been labeled so RFLP analysis, or DNA sequencing.
that it can be detected following a hybridization reaction. Because
unique regions exist in the sequence of bases in all organisms, a REAL TIME PCR
complementary nucleic acid probe can be prepared such that it
hybridizes only to that unique region and specifically detects the Real time PCR utilizes a thermocycler that contains a
disease-causing agent (6). spectrophotometer to measure amplification of the PCR product
There are many ways to obtain and label nucleic acid probes (3). during the PCR run. Fluorescent dyes that bind to the amplified
Typically a gene from the organism that is to be detected must be DNA are included in the reaction mixture and fluorescent
isolated then labeled with either a radioisotope or with monitoring occurs at each of the PCR cycles allowing detection of
nonradioactive substances. Nonradioactive labels are more the amplified product in real time. Several methods of monitoring
attractive than radioisotopes for use in diagnostic tests because they fluorescence have been reported but the most commonly used are
are not as hazardous to handle and can be stored for long time SYBR green, TaqMan® probes, and fluorescence resonance energy
periods. Two of the most frequently used labels are biotin and transfer (FRET). SYBR green is a chemical that fluoresces when it
digoxigenin. Probes labeled with biotin are detected by a interacts with double stranded DNA. During each PCR cycle, new
streptavidin-alkaline phosphatase (or other enzyme) conjugate and DNA is synthesized and is available to bind the SYBR green
the appropriate substrate to produce a color reaction similar to an molecule. Thus, as the amount of DNA increases, so does the
enzyme-linked immunosorbent assay (ELISA) reaction. For amount of SYBR green fluorescence. Fluorescence readings are
digoxigenin-labeled probes, an anti-digoxigenin antibody taken at the end of each PCR cycle.
conjugated with various enzymes can be used with the appropriate TaqMan® probes and FRET utilize fluorescently labeled DNA
substrate to produce a color reaction. When anti-digoxigenin probes specific for the target-amplified sequence. TaqMan® probes
antibodies are conjugated to enzymes such as alkaline phosphatase, have a fluorophore and a quencher attached to either end of a probe.
a chemiluminescent substrate can be used, which emits light that When the probe is intact, the quencher molecule prevents
when exposed to X-ray film provides a very sensitive detection fluorescence from the fluorophore molecule. During PCR the probe
method. hybridizes to the template. Then the 5’ exonuclease activity of the
Oligonucleotides, which are short (usually 30-40 nucleotides long) Taq polymerase destroys the probe as it synthesizes a new
synthetically manufactured single-stranded DNA fragments, can be complementary strand of DNA. This releases the fluorophore from
labeled and used as probes. RNA can also be labeled and used as a the quencher and fluorescence is detected by the spectrophotometer.
probe. Increasing amounts of DNA made during each PCR cycle, are
detected by the hybridization of more probes and their subsequent
destruction which leads to an increase in fluorescence.
The FRET technology uses two probes a donor probe conjugated
to fluorescein isothiocyanate (FITC) and an acceptor probe
conjugated to a red dye. Several red dyes are available including
red 610nm, 640nm, 670nm and 705nm, Because the FITC and red
231
Deral J. Jackwood and Mark W. Jackwood

dye must be physically close to each other for FRET to occur, when NUCLEIC ACID SEQUENCING
the donor and acceptor probes are present in solution and not
hybridized to a template, no signal is detected. The probes are Nucleic acid sequencing is a method whereby the order of the
designed to hybridize to the amplified PCR product such that the nucleotides (A, C, G, and T) in a nucleic acid segment is
FITC and red dyes come into close proximity to each other. A determined. Sanger dideoxynucleotide chain termination
470nm light source excites FITC on the donor probe, which emits sequencing is the standard sequencing method used in most
energy that is transferred to the red dye on the acceptor probe. The laboratories (4). Nucleic acid sequencing has been automated.
red dye then emits light at a specific wavelength that is detected by Although the equipment is expensive and a certain level of expertise
the spectrophotometer. Since four different red dyes are available, is required, nucleic acid sequencing is available through numerous
four probe pairs could be used in a multiplex real-time PCR commercial companies.
reaction. During the annealing phase of each PCR cycle the probes Sequencing provides a method of specifically identifying
are annealed to the denatured product and fluorescence is measured. microorganisms based on the actual sequence of their genome.
These probes are not destroyed during the polymerization phase of Nucleic acid sequencing coupled to PCR or RT-PCR can be an
the PCR cycle as they are in the TaqMan® method. Increasing extremely sensitive as well as a specific diagnostic technique.
amounts of DNA produced during each PCR cycle, allows more Sequence data can be compared against the National Center for
probes to hybridize and consequently more FRET signal. This Biotechnology Information sequence database (GenBank,
fluorescence signal is detected at the end of each annealing phase of http://www.ncbi.nlm.nih.gov/) using the BLAST (Basic Local
the PCR cycle. Alignment Search Tool) program. The BLAST program compares
Real time PCR data is generally plotted as fluorescent units per nucleotide or protein sequences to the sequences in the databases
PCR cycle number. A threshold value of fluorescence is set to and calculates a statistical similarity, which can be used to identify
indicate when the sample is considered positive and the number of the infectious agent.
cycles needed for a sample to cross the threshold is reported as the Sequence data can also be used to generate phylogenetic trees,
cycle threshold or CT value. which show the relationship between two or more closely related
After PCR amplification, reactions that utilize SYBR green or organisms presented as a percent similarity between the nucleotides
FRET can be analyzed by melting curve analysis. Melting curve or amino acids. There are rooted and unrooted trees and several
analysis determines when the double stranded amplified product different methods used to identify evolutionary distance
denatures into single strands or when the probe dissociates from the relationships among sequences. Several of the more commonly
amplified product. Melting curve analysis can be added to the end used methods include the unweighted pair-group method using
of the PCR run and, since it is sequence dependent, can be used to arithmetic averages (UPGMA), Neighbor joining method, minimum
verify the PCR product. evolution method and maximum parsimony methods (2). Because
Real time PCR can also be quantitative. Measuring the increase in these methods are statistical estimates of evolutionary relationships,
fluorescence over time provides a mechanism to determine the confidence intervals are often calculated using bootstrap variances
efficiency of the reaction, which in turn can be used to quantify the and covariances.
starting template. Quantitative real-time PCR and quantitative real­
time RT-PCR are important because they can be used to determine REFERENCES
the quantity of nucleic acid present, which gives an indication of the
amount of infectious agent in the sample. 1. Erlich, H. PCR technology. Stockton Press, New York, N.Y. 1989.
2. Nei, M, S. Kumar. Molecular evolution and phylogenetics. Oxford
University Press. New York, N.Y. 2000.
RANDOM AMPLIFIED POLYMORPHIC DNA (RAPD) 3. Sambrook, J., E. F. Fritsch, and T. Maniatis. Molecular cloning a
laboratory manual, 2nd ed. Cold Spring Harbor Laboratory Press, New
The RAPD technique, also known as arbitrarily primed York, N.Y. 1989.
polymerase chain reaction (AP-PCR), was developed by Williams 4. Sanger, F., S. Nicklen, and A R. Coulson. DNA sequencing with chain­
et al. (9) and Welsh and McClelland (7). This PCR technique uses terminating inhibitors. Proc. Natl. Acad Sci. 74:5463-5467. 1977.
short primers with arbitrary sequence to randomly amplify regions 5. Stryer, L. Biochemistry, 2nd ed W. H. Freeman and Co., San Francisco,
of DNA. These randomly amplified DNA fragments are separated Calif. 1981.
6. Tenover, F. C. Diagnostic deoxyribonucleic acid probes for infectious
by electrophoresis and the resulting patterns are markers for regions diseases. Clin. Microbiol. Rev. 1:82-101. 1988.
of the target DNA. 7. Welsh, J., and M McClelland Genomic fingerprinting using arbitrarily
To conduct the RAPD technique, the target genomic DNA is primed PCR and a matrix of pairwise combinations of primers. Nucleic
amplified using a single oligonucleotide primer that has an arbitrary Acids Res. 19:5275-5279. 1991.
sequence. This primer is usually 10-12 nucleotides in length, but 8. Williams, J. G. K., Μ K. Hanafey, J. A. Rafalski, and S. V. Tingey.
longer primers have been used. The PCR used to generate the Genetic analysis using random amplified polymorphic DNA markers.
RAPD is conducted under typical conditions. The reaction products Methods Enzymol. 218:704-740. 1993.
are dependent on the primer sequence, primer length, and reaction 9. Williams, J. G. K., A. R. Kubelik, K. J. Livak, J. A. Rafaiski, and S. V.
Tingey. DNA polymorphisms amplified by arbitrary primers are useful as
conditions of the PCR. When the primer and PCR reaction genetic markers. Nucleic Acids Res. 18:6531-6535. 1990.
conditions are held constant, RAPD markers are very reproducible
and characteristic of the target DNA.
The RAPD technique was initially used to generate markers for
genetic mapping of quantitative trait loci and genetic diagnosis in
plants and animals (8). More recently, RAPD markers have been
used to identify poultry pathogens. The RAPD technique has great
potential for identification of poultry pathogens with complex DNA
genomes because it does not require knowledge of the target DNA
sequence (8, 9). A comprehensive review of the RAPD analysis has
been published (8).
49
ANTIGEN DETECTION SYSTEMS
Mary J. Pantin-Jackwood and Sandra S. Rosenberger

SUMMARY. Infectious agents or their constituent parts (antigens or nucleic acids) can be detected in fresh, frozen, or fixed tissue using a
variety of direct or indirect assays. The assays can be modified to yield the greatest sensitivity and specificity but in most cases a particular
methodology is chosen based on the nature of the sample, availability of equipment, quality of reagents, time, cost, and technical expertise.
Whole virions or bacteria can be visualized directly using electron microscopy. Although this procedure can be performed quickly, it is
insensitive and requires specialized equipment and technical support. Some tests such as agglutination reactions or immunodiffusion assays
are simple and inexpensive but they can only be used to identify a particular etiologic agent possessing agglutinating or soluble antigens,
respectively. The more cumbersome techniques such as antigen-capture enzyme-linked immunosorbent assay, immunohistochemical
staining, or in situ hybridization offer increased sensitivity and specificity but may require better quality reagents, additional pretreatment
procedural steps prior to testing, and a higher level of technical training. To optimize antigen detection systems for diagnostic and research
purposes it is as important to become familiar with the limitations of the assays as it is to understand the pathogenesis of the suspected
etiologic agent in selecting the timing and type of tissue samples for testing.

INTRODUCTION to confirm the presence of the expected virus, but is of even greater
use in situations in which a virus infection is suspected but no CPE
The detection of infectious agent antigens directly in clinical is produced. Also, EM is very useful for monitoring all cell culture
specimens has become an important component of the diagnostic stocks for adventitious viruses and mycoplasmas that can
methodology. Antigen detection methods can provide diagnostic contaminate cultures (48).
information within a few hours of the receipt of die specimen in the The disadvantage of using EM is the availability of the microscope
laboratory; this, and the lack of requirement of the infectious agent and support staff. EM also has drawbacks in sensitivity and
viability, is an important advantage over viral or bacterial culture, specificity that limit application. A fairly high concentration of
especially when the specimen transport time is prolonged or virus is necessary for visualization by negative staining of fluids,
otherwise suboptimal (82). A variety of antigen detection systems although immune electron microscopy (IEM) can increase
for avian pathogens have been developed or modified over the past sensitivity. In thin sections, if the infection is not widespread,
years. The best antigen detection system should be both sensitive virions may be missed because of sampling error. Analysis based
and specific. Sensitivity is measured by the ability to detect very solely on morphology cannot differentiate between viruses in the
small amounts of antigen. Increasing the sensitivity of a test system same family, although IEM can sometimes overcome this problem
will decrease the number of false negative samples. Specificity is as well (48).
measured by the ability to detect a particular antigen while limiting Sample transport between the clinician and the electron
nonspecific reactions or cross reactions with related antigens. microscopist is important to assure that the specimens arrive in
Increasing the specificity of a test system will decrease the number optimal condition. Liquids should not be frozen and tissues should
of false positive samples. However, many antigen detection systems be fixed in 2-5% glutaraldehyde in buffer immediately and stored
are chosen based on technical reasons such as reagent or equipment cold during transport (48). If no samples have been properly
availability, length of time for test procedure, cost per test, collected for EM, formalin fixed, paraffin-embedded tissues can
automation, environmental or safety requirements, and technical still be used.
difficulty.
This chapter will discuss several common techniques used to Negative staining
detect antigens to various pathogens that infect birds. A general Simplicity and rapidity, coupled with high resolution, have
overview of each protocol will be presented, along with some ensured that the negative staining technique is of major importance
current references giving various technical options that may be in virus diagnosis. However direct EM is relatively insensitive with
useful in developing the appropriate test system for an individual a critical lower detection limit of approximately ΙΟ6-107 virus
laboratory. particles per milliliter. The development of airfuge
ultracentrifugation of samples directly onto transmission EM grids
ELECTRON MICROSCOPY has greatly improved antigen detection (30). Negative staining
techniques using phosphotungstic acid and uranyl acetate are used
Electron microscopy (EM) is a valuable tool especially in the to increase contrast, but in some cases viruses or bacteria may lose
diagnosis of viral infections when applied judiciously and used in identifying projections and may be too widely spaced on the grids
conjunction with other diagnostic tests (48). EM has several or among debris to make positive identification. Tissue samples
advantages as a viral diagnostic procedure; in some cases, such as should be fixed, embedded and sectioned for visualization of
for diagnosis of enteric viruses, is the method of choice. It is viruses (48). The best method for virus detection in cultured cells is
relatively simple and rapid. It can be applied to the diagnosis of any thin sectioning, although negative staining can be used to examine
productive viral infection without prior knowledge of which agent the supernatant from the cell culture.
is present to guide the reagent selection, and it does not require the
presence of viable virions (48). In contrast, culturing techniques Immune Electron Microscopy
may be unsuccessful because improper specimen handling may The sensitivity of direct EM can be increased by the addition of
have rendered the sample inviable or because some viruses are highly specific polyclonal or monoclonal antisera following
difficult or impossible to grow in cell culture. Similarly, diagnostic clarification but prior to centrifugation (see Chapter 34 on Enteric
tests that use immunological reagents detect only the agent which viruses). This procedure has been termed immune EM (IEM). The
they are directed and will miss unsuspected viruses. Furthermore, antigen-antibody complexes formed require only low-speed
antibodies are not available for all viruses. EM also has several centrifugation for placement on grids. Alternatively, antibody may
useful applications in the examination of virus-infected cell culture. be bound directly to the film coating of the grids forming a surface
If a distinctive cytopathic effect (CPE) is present, EM can be used to capture antigen (22). An indirect protein A-gold IEM test has
233
Mary J. Pantin-Jackwood and Sandra S. Rosenberger

proven to be a rapid sensitive method for detection of enteric inactivated antigens (32). AC-ELISA has been shown to be more
coronaviruses (20). IEM has been used to identify unknown viruses sensitive than the AGID test (77), but less sensitive than
by the use of convalescent sera, or to specifically serotype viruses conventional methods such as virus isolation in embryonating eggs
(5,48). unless some initial enrichment of antigen is performed (3, 52).
Advantages of AC-ELISA include applicability to diverse
HEMAGGLUTINATION (HA) TEST specimens and potential for automation. Laboratory instruments are
Hemagglutinins are glycoproteins expressed on the surface of now available that can perform AC-ELISA for the detection of
many disease-causing agents. Hemagglutinins can easily be antigens. The sensitivity and specificity of this antigen detection
detected by addition of red blood cells from a species that carries system is directly dependent on the choice of reagents and
complementary receptors (22). However, some infectious agents, procedures.
such as infectious bronchitis virus (IBV), may require pretreatment
with enzymes to expose surface hemagglutinins (71) (see Chapter Primary Capture Antibody
32 on infectious bronchitis virus). The basic procedure involves binding primary capture antibody to
If materials from tracheal swabs are inoculated into specific- specially designed ELISA plates through a series of steps. Coating
pathogen-free embryos the presence of a hemagglutinating agent plates with protein A of Staphylococcus aureus (79) or an avidin­
may be quickly detected by mixing approximately 0.05 ml of 10% biotin complex (32) prior to the addition of capture antibody has
washed chicken red blood cells with approximately 0.5 ml of been suggested to increase the antigen capture capability. The
harvested allantoic fluid on a glass plate (spot HA test). If capture antibody may be polyclonal or monoclonal antibody
agglutination occurs, it can be verified and the particular agent may depending on the purpose of die particular test system. High quality
be identified by the hemagglutination-inhibition (HI) test described polyclonal antiserum is more sensitive in screening samples for
in detail in Chapter 47 on serologic procedures. Because some antigen (33), whereas monoclonal antibody is more useful in
bacteria may agglutinate red blood cells, bacterial contamination identifying the presence of a particular serotypic antigen (52).
should be examined by streaking the sample on microbiological However, very specific polyclonal antiserum can be prepared by
media or repeating the agglutination reaction on the fluids following preabsorbing with low concentrations of related heterologous
filtration through a 0.2-pm syringe-type filter prior to performing antigens (3).
the HA/HI tests.
Antigen Binding
IMMUNODIFFUSION Following various washing steps, freshly collected tissue
homogenates or samples previously inoculated into an appropriate
Soluble viral antigens present in tissue homogenates, egg fluids, culture media for enrichment (3, 52), are added to the test wells and
and tissue culture fluids, can be detected using immunodiffusion in incubated with the appropriate capture antibody bound to the plate
an agar-gel immunodiffusion (AGID) test. This test, described in surface. If the antigen is recognized by the antibody it will be
Chapter 47 on serologic procedures relies on the ability of antigen captured on the plate. Unbound antigen will be washed away.
and antibody to move through an agar matrix forming a precipitin Additional steps may be required in antigen preparation because
line. The AGID test is very simple and inexpensive. For example, some antigens may need to be exposed by enzyme or detergent
infectious bursal disease virus (IBDV)-infected bursae or treatment or precultured prior to screening (52).
chorioallantoic membranes from reovirus-infected embryos can be
prepared as 50% (w/v) homogenates in phosphate-buffered saline Detector Antibody
using a tissue grinder or mortar and pestle. After three freeze-thaw After additional washing steps a secondary or detector antibody is
cycles, the homogenate is centrifuged at low speed and the added. The detector antibody may be labeled directly with enzyme
supernatant used as a test antigen for immunodiffusion. Supernatant such as horseradish peroxidase (89) or biotin (79), but is usually left
fluids from infected cell cultures can also be used in this antigen unlabeled in most procedures. Following incubation with unlabeled
detection system. However, some antigens may not be soluble and detector antibody, enzyme-conjugated antibody is added. Once the
will not diffuse through the agar matrix or the concentration of appropriate substrate is supplied to the test system, captured antigen
antigen will be below the detection limit of the test. Treatments with is detected by a color change recorded as optical density or
detergents or concentration of antigen, respectively, may allow use absorbance values compared to the controls. Specific antigenic
of the immunodiffusion assay, but another antigen detection method types can be detected by using monoclonal antibody in the AC-
may be more appropriate in these cases. AGID has been used for ELISA (52, 79) or the ELISA may be altered into a competition or
detection of several avian viruses including adenovirus, IBDV, ATV inhibition assay (85).
and reovirus. (4, 65, 70) (Chapter 29 on ATV).
Membrane Immunoassays
ANTIGEN CAPTURE ENZYME-LINKED Membrane immunoassay or dot ELISA’s are variants of AC-
IMMUNOSORBENT ASSAY (AC-ELISA) ELISA in which antigens or antigen-antibody complexes are
captured directly on membranes (43, 49, 82). Membrane
Pathogen antigens may be detected in tissue preparations using an immunoassays are also manufactured as self-contained cassettes
adaptation of the ELISA technology discussed in Chapter 47 on that are convenient for testing single specimens. These assays are
serologic procedures. This adaptation, termed antigen-capture simple enough to be used in the field and are routinely used for
ELISA (AC-ELISA), has been used to detect numerous avian diagnosis of avian influenza (14, 19, 72, 82, 90). Large amounts of
pathogens including: avian influenza virus (ATV), IBDV, antibody can be bound to nitrocellulose, nylon membranes, or other
enteroviruses, IBV, avian encephalomyelitis virus (AEV), West modified membranes greatly increasing the sensitivity and reducing
Nile virus, adenoviruses, reticuloendotheliosis virus (REV), total assay time to minutes. Alternatively, the reaction of the
reovirus, turkey coronavirus (ToCV), avian leukosis virus, antibody and the sample can take place on microparticles in a tube
Mycoplasma, and Clostridia, (3, 8, 12, 17-19, 21, 26, 32, 33, 45, 50, or cuvette. These microparticles or beads coated with antibody
52, 54, 73, 74, 77, 85, 88, 91). AC-ELISA is a relatively simple and serve as the solid-phase support.
quick assay providing an alternative to laboratories where histologic Western blot analysis is another antigen detection method where
samples for immunohistologic staining are not readily available. pathogen proteins are separated by electrophoresis in a
The assay can measure live, infectious or noninfectious antigens or polyacrylamide gel and the transferred to a thin, synthetic
234
Chapter 49 Antigen Detection Systems

membrane that has a strong affinity for proteins. Proteins bound to Frozen histological sections can be used by quick freezing tissue in
the membrane are detected by immunostaining. Because proteins isopentane pre-cooled with liquid nitrogen and then cutting on a
bound to the membrane are denatured, antibodies that recognize cryostat.
non-linear epitopes are usually not suitable for their detection. The In most cases, tissues are fixed at room temperature in 10% neutral
main advantage of this technique is that it does not require labeling buffered formalin for routine histological examination. If IHC is to
of proteins and therefore can be applied to tissues and organs as be done, tissues should not be kept in formalin fixatives for more
well as cultured cells (82). This method has been used for detection than 24—48 hr prior to processing because antigenicity can be
of IBDV and Mycoplasma gallisepticum (61, 85). diminished to varying degrees (29). Formalin cross-links
polypeptides by the formation of methylene bridges, which can
IMMUNOHISTOCHEMISTRY mask or alter antigens. Other fixatives containing various
combinations of ethanol, formaldehyde, and glacial acetic acid or
Immunohistochemistry (IHC) is the localization of antigens in zinc, which supposedly inhibits formaldehyde-induced cross
tissue sections by the use of labeled antibodies as specific reagents linking, have been used. Because certain fixatives or length of
through antigen-antibody interactions that are visualized by a fixation may produce variable results for a particular antigen,
marker such as fluorescent dye, enzyme, radioactive element or procedures should be standardized for each agent to have reliable
colloidal gold (1). This antigen detection system has been used to and accurate detection systems.
visualize antigen in cells or tissues infected with several avian Following fixation, tissues are embedded in paraffin and cut into
pathogens including infectious laryngotracheitis virus (ILTV), IBV, sections. To avoid further tissue damage, the paraffin infiltration
IBDV, adenoviruses, AIV, duck viral enteritis, Newcastle disease temperatures should not exceed 60 C (29). Tissue sections should
virus (NDV), AEV, chicken anemia virus (CAV), reovirus, AE be mounted on slides coated with adhesive compounds such as
virus, fowlpox virus, ToCV, ALV, Marek’s disease virus (MDV), poly-L- or poly-D-lysine or 3-aminopropyltriethoxysilane.
REV, Mycoplasma, avian pneumovirus, HEV, and others (9, 11, Following standard deparaffinization, the slides are rehydrated and
15, 16, 23-25, 27, 28, 34-39, 41, 42, 46, 51, 57-60, 64, 67, 75, 78, prepared for staining.
80, 83, 84, 86, 87, 92). A review on the use of Tissue Pretreatments Antigen Retrieval. The demonstration of
immunohistochemical detection of avian infectious agents can be many antigens can be significantly improved by the pretreatment
found in Ramos-Vara et al. (69). IHC allows for the direct with antigen retrieval (AR) reagents and procedures that break the
morphologic localization of an infectious agent and the correlation protein cross-links formed by formalin fixation and thereby uncover
with corresponding pathologic cellular changes or lesion hidden antigenic sites (1). The techniques involve the application of
development if tissue samples are collected at the appropriate time heat or enzyme digestion. Sometimes, multiple antigen retrieval
during infection. For this reason IHC is frequently used in methods are needed to optimize the immunodetection of antigens
pathogenesis studies. With proper controls, positive findings can be (68). The majority of antibodies to bacteria, fungi, and protozoa do
used in diagnosis of specific avian diseases. However, negative not require this treatment (6).
findings should be interpreted with caution (13). A good IHC test Protease-induced antigen retrieval. The use of proteolytic
should be technically simple to minimize errors, inexpensive, and enzymes, such as proteinase K, trypsin, chymotrypsin, pepsin,
allow sufficient signal amplification for easy visualization. Most pronase and various other proteases, has been reported for restoring
importantly, the system must be able to immunogenically detect immunoreactivity to antigens in tissues (1, 29, 68). The effect of the
antigen that has been structurally altered by fixation. An advantage protease-induced AR depends on the concentration and type of
of antigen detection on formalin-fixed tissues is that the same enzyme, incubation parameters (time, temperature, and pH), and the
specimen may be used for both routine histological and IHC duration of fixation (68). The enzyme digestion time is inversely
examination. The ability to detect antigens in fixed specimens also related to the fixation time (68). The disadvantages of protease-
enables retrospective diagnosis in cases when no longer fresh induced antigen retrieval are the rather low number of antigens for
tissues are available (29). The sensitivity and specificity of the IHC which it is the optimal AR method, possible alteration of tissue
test system may be quite variable depending on the procedures, morphology, and possible destruction of epitopes (68). Therefore
reagents, and so on. Consulting several text and review articles on the optimal enzyme concentration and incubation time needs to be
IHC before attempting this assay may be useful (1, 13, 29, 62). determined.
Heat-induced antigen retrieval. The chemical reaction between
Tissue Processing proteins and formalin can be reversed in part by high temperature
Cell or Tissue Fixation To ensure the preservation of tissue coupled to a slightly acidic solution to enhance antigen delectability
architecture and cell morphology, prompt and adequate fixation is (10). Microwave oven, pressure cooker and steamer are the most
essential. However, inappropriate or prolonged fixation may commonly used heating methods. Other tools also include the use of
significantly diminish the antibody binding capability (1). Unfixed an autoclave or a water bath. A heating time of 20 min appears to be
cells or tissue cultures can be used for IHC but this requires that the most satisfactoiy and cooling usually takes about 20 min (1).
preparations be examined quickly (9). Cells or whole tissue samples Citrate buffer at pH6.0 is the most popularly used retrieval solution
are usually fixed in one of several ways to perform IHC. For and is suitable for most antibody applications. The TRIS-EDTA
example, the freshly cut surface of a thymus, a sample of bone buffer at pH9.0 and EDTA at pH8.0 are the second most used
marrow, or tracheal mucosal scrapings, may be pressed onto a retrieval solutions (1).
clean, degreased glass slide with a scalpel blade and removed. The
thin layer of cells left on the slide is air dried and then fixed in Blocking
acetone for 10 min (46). These slides can be stored at -20 C until Background staining. Background is one of the most common
staining. Infected cells grown in tissue culture may be spotted onto problems in immunohistochemistry, and it can seriously affect the
a glass slide and fixed in a similar way (89). interpretation of the immunologic reaction (68). Background
Certain cell antigens do not survive routine fixation and paraffin staining may be specific or non-specific. Inadequate or delayed
embedding. So the use of frozen sections still remains essential for fixation may give rise to false positive results due to the passive
the demonstration of many antigens. However, the disadvantage of uptake of serum protein and diffusion of the antigen. Antibodies,
frozen sections includes poor morphology, poor resolution at higher especially polyclonal antibodies, are sometimes contaminated with
magnifications, special storage, limited retrospective studies and other antibodies due to impure antigen used to immunize the host
tissue section cutting difficulty over paraffin sections (1).
235
Mary J. Pantin-Jackwood and Sandra S. Rosenberger

animal (1). The main cause of non-specific background staining is Direct Staining. Direct staining is a one step method that involves
non-immunological binding of the specific immune sera by a labeled antibody reacting directly with the antigen in tissue
hydrophobic and electrostatic forces to certain sites within tissue sections. This technique is short and quick. However, it is
sections. This form of background staining is usually uniform and insensitive due to little signal amplification (1). Different labels
can be reduced by blocking those sites with normal serum (1). The have been used, including fluorochromes, enzymes, colloidal gold,
increased hydrophobicity of proteins during fixation increases the and biotin (68). Although for diagnostic purposes fluorochromes are
background staining in IHC procedures; therefore, prolonged preferred, enzyme conjugates are favored because slides can be
fixation in formalin or other aldehyde-based fixatives should be stored for several years without loss of staining intensity. Direct
avoided. Background staining from over fixation can be remedied IHC requires that the primary antigen-specific antibody be labeled
by post-fixation with Bouin's, Zenker's, or B5 fixatives (68). In with fluorescein dye or an enzyme and incubated with the tissue
order to prevent non-specific biding, prior to incubation with the sample. The primary antibody must be a high-quality polyclonal or
primary antibody the slides should be treated with 2-5% normal monoclonal antibody to minimize nonspecific binding and the
non-immune serum from an unrelated species, bovine serum highest possible dilution that gives specific staining should be used
albumen, nonfat dry milk, or casein, or, if indirect staining will be (29). Direct fluorescein assays (FA) have been used successfully for
used, serum from the species in which the secondary antibody is many years on fresh or frozen tissues (9, 47). In the direct format, a
prepared. Polyclonal antibodies should be used as highly diluted as fluorescent label, usually fluorescein isothiocyanate (FITC) is
possible to reduce the concentration of proteins that might bind non- conjugated to the antibody that recognizes the antigen. After
specifically (62). staining, the specimen is viewed with epiilumination using
Endogenous enzyme activity. Peroxidase activity naturally ultraviolet light of the wavelength needed to excite the fluorescent
present in red blood cells, granulocytes, and neurons can react with label (82).
the peroxidase staining method to produce a brown product Indirect Staining. Indirect LHC is used more often than direct
indistinguishable from specific immunostaining (68). Although IHC. Unlabeled antigen-specific primary antibody is incubated with
endogenous peroxidase activity is destroyed almost completely the tissue sample and after several washing steps, a secondary
during formalin fixation, pretreatment of tissues with a diluted antibody labeled with fluorescein dye, an enzyme, or biotin is
solution (0.003-3%) of H202 in methanol will further reduce or applied. The secondary antibody is usually a commercially
completely abolish endogenous peroxidase activity (68). available antibody prepared against immunoglobulin G (IgG) of the
Inactivation of endogenous tissue enzymes should be performed animal used to prepare the primary antibody (13, 29). The indirect
prior to proteolytic digestion to minimize the effect of H2O2 on staining method is more complex than the direct method, but the
tissue antigens (29). Many tissues also contain endogenous alkaline indirect method does not require conjugation of primary antibody
phosphatase (AP) activity and should be blocked by the and commercially available fluorescein- or enzyme-conjugated
pretreatment of the tissue section with levamisole if using AP as a secondary antibodies may be used with numerous primary
label (1). Some tissues such as liver and kidney have endogenous antibodies. More importantly, because several secondary antibody
biotin. To avoid unwanted avidin binding to endogenous biotin if molecules may bind with a single primary antibody, the sensitivity
using biotin-avidin detection system, a blocking step is necessary of this test is enhanced by intensifying the detection signal (29).
for these tissues by the pretreatment of unconjugated avidin which Avidin-biotin methods. The sensitivity of the indirect IHC test
is then saturated with biotin (1). may be further increased by using the avidin-biotin-peroxidase
Fc receptors. False positives can occur due to cross-reactions of complex (ABC Methods) or streptavidin-biotin-peroxidase
secondary antibody with endogenous antibody or binding of complex (LSAB methods). The avidin-biotin-peroxidase complex
secondary antibody especially to inflammatory cells that have Fc immunostain increases sensitivity by utilizing the high binding
receptors on their surface (29). Fc receptors of mononuclear blood affinity of the B vitamin biotin for avidin, an egg white
cells can bind IgG, but this is not usually a problem with routinely glycoprotein. Streptavidin also binds biotin, but is produced by
fixed paraffin-embedded tissues because Fc receptors are destroyed bacteria and may be more suitable for detecting avian disease­
during this process. The use of F(ab')2 fragments of the Igs instead causing agents. In this detection system, unlabeled primary antibody
of the whole Ig molecule eliminates nonspecific background from is incubated with the tissue sample. After washing, a biotin-labeled
Fc receptors or the Fc portion of Igs (62, 68). secondary antibody directed against IgG of the animal used to
Autofluorescence. Autofluorescence or natural fluorescence exists prepare the primary antibody is allowed to incubate on the tissue
in some tissues and can cause a background problem when sample. After washing, an avidin-biotin or streptavidin-biotin
fluorescent labels are used in the experiments (62). The simplest complex labeled with the enzyme peroxidase is added and
test is to examine the tissue sections with a fluorescence microscope following a third wash, substrate is added. This system is much
prior to conducting the assay. If autofluorescence is detected in the more time consuming and expensive, but it offers greater
tissue sections, the best solution is to use enzyme or other labeling amplification of scarce antigens. The avidin-biotin-peroxidase
methods (1). complex system in conjunction with monoclonal antibody has been
shown to yield the best results for detection of some antigens (16,
Detection systems 57, 84).
The antigen-antibody reaction cannot be seen with the light Peroxidase anti-peroxidase (PAP) method. This method is a
microscope unless it is labeled. Labels are attached to the primary, further development of the indirect technique and it involves a third
secondary, or tertiary antibodies of a detection system to allow layer, which is a rabbit antibody to peroxidase, coupled with
visualization of the immune reaction. A variety of labels have been peroxidase to make a very stable peroxidase anti-peroxidase
used, including fluorescent compounds, enzymes, and metals (68). complex (1). The sensitivity is higher and also allows for much
The most commonly used labels are enzymes (e.g., peroxidase, higher dilution of the primary antibody, thus eliminating many of
alkaline phosphatase, glucose oxidase). Enzymes in the presence of the unwanted antibodies and reducing non-specific background
a specific substrate and a chromogen will produce a colored staining (1).
precipitate at the site of the antigen-antibody reaction. Selection of a Polymeric labeling two-step method. This method consists of a
detection system is very important, considering that the sensitivity compact polymer to which multiple molecules of enzyme and the
of cm immune reaction will depend mostly on the detection system secondary antibody (specific for the primary antibody) are attached.
used (68). Detection systems are classified as direct or indirect The advantages are: simplicity compared to the three-step methods,
methods. equal or higher sensitivity than ABC or LSAB methods, and lack of
236
Chapter 49 Antigen Detection Systems

background staining because of endogenous biotin or avidin (68). facilitated the use of ISH. Compared to immunohistochemistry, this
One disadvantage is that this method is usually more expensive than system can more easily detect latent infectious and antigenic
ABC or LSAB methods. Numerous companies have subtypes by using highly specific probes, but the required technical
commercialized polymer-based detection systems (1, 68). expertise and cost may be prohibitive in many laboratories. The
Tyramine amplification method. This method is more complex sensitivity of ISH compared to other systems such as IHS for some
and laborious and it involves an initial avidin-biotin procedure avian antigens may be questionable (2).
followed by a tyramine reaction. However, the sensitivity of the
reaction is increased allowing the detection of antigens present in Tissue Preparation
very low amounts in the tissue (1). Paraffin-embedded sections, frozen sections, or tissue smears are
Color of the antigen-antibody reaction. The antigen-antibody prepared in the same way as for IHC. Fixation is critical to preserve
reaction is not visible with the light microscope unless a label is cellular morphology for optimal probe localization and
used. The most commonly used enzymes are horseradish peroxidase identification of labeled cells (6, 44). Prompt fixation or rapid
(HRP) and alkaline phosphatase (AP). Each enzyme has specific freezing will aid in retaining nucleic acids. Aldehyde fixatives, such
substrates and chromogens to produce a colored precipitate. Most as formalin, cross-link DNA with histone proteins that will prevent
common chromogens impart a brown, red, or blue color to the hybridization in a time-dependent manner.
reaction. The choice of enzyme and chromogen will depend on Paraformaldehyde-based fixatives may retain more of the
several factors (e.g., intensity of reaction, location of the antigen, histomorphologic structures. Slides that have been treated with 3-
presence or absence of endogenous pigments, mounting media aminopropyl-triethoxysilane have been suggested for tissue sections
used) but often is a matter of personal preference. For HRP, 3,3’ used with ISH because the high temperatures and enzyme
diaminobenzidine tetrachloride (DAB) is the most commonly used treatments usually detach tissue from untreated slides (6, 44). As
chromogen, giving a brown color, and its insoluble in organic described previously, tissues must be pretreated to increase cell
solvents. 3-amino-9-ethylcarbazole (AEC), another chromogen for permeability and decrease background staining. Proteolytic
peroxidase, produces a red color, but coversliping should be made digestion also aids in separating proteins bound to DNA or RNA to
with a water soluble medium (the precipitate will wash out with allow the probe access for binding. All proteolytic digestions must
organic solvents). For AP, 5-bromo-4-chloro-3- be done with nuclease-free enzymes.
indolylphosphate/nitro blue tetrazoliumchloride (BCIP/NBT; blue
permanent media), fast red (red, aqueous mounting media), and new Probe Selection
fuchsin (fuchsia, permanent media) are the chromogens most The probe is a specific sequence of cloned or synthesized
commonly used. The choice of counterstain will depend on the nucleotides prepared to identify and bind a particular region of
color of the immune reaction. The most frequently used DNA or RNA. The probes are double-stranded DNA, single­
counterstains are hematoxylin (blue), methyl green (green), and stranded complementary RNA, often'referred to as "riboprobes",
nuclear fast red (red) (68). synthetic oligonucleotides or peptide nucleic acids (62). Labeled
nucleotides are incorporated into the probe sequence or added as a
Controls tail. Longer probes will have more incorporated labeled nucleotides
Special controls must be included in order to test the protocols and for better detection. However, the length of the probe affects the
the specificity of the antibody. A positive control tissue is needed to rate of cell penetration. The use of radioactive probes, although
verify the protocol or procedure used (1). Ideally, tissues of a viewed as more sensitive, is impractical for most laboratories
known positive should be used as a control. A negative control because of safety and disposal problems. Non-radioactive probes
tissue is also needed to test for the specificity of the antibody are preferred and are produced by conjugating nucleotides to
involved. Additional controls should include omission of the reporter molecules such as biotin, digoxigenin, or fluorescein. The
primary antibody or the replacement of the specific primary choice of any particular probe is highly dependent upon the desired
antibody by a normal serum (must be the same species as primary application. Regardless of the steps taken towards optimizing probe
antibody)(l). design and performance, specimen processing and sample
pretreatment remain two of the largest sources of variability in
IN SITU HYBRIDIZATION (ISH) assay performance (31).

In situ hybridization (ISH) is a method of localizing unique nucleic Hybridization


acid sequences within a formalin-fixed tissue section or in samples In preparation for hybridization, proteolytic enzyme reactions are
that have been transferred to a nitrocellulose membrane (dot-blot stopped by extensive wash cycles. The probe and its target must be
hybridization). Such detection is made possible by hybridization of single-stranded for successful hybridization. Denaturation of
DNA and/or RNA targets with nucleic acid probes tagged with a double-stranded DNA is usually accomplished by heating in the
signal generating system such as fluorochrome or enzyme (53). This presence of formamide at 92-96 C for 6-10 min. However, each
nucleic acid detection system combines immunohistological laboratory will need to assess the proper temperature that will
staining procedures and some basic molecular biological principles. produce the best signal without changing the cell morphology.
Detailed molecular detection procedures can be located in Chapter Some protocols may include procedures to further permeabilize the
48 on molecular identification procedures. tissue for increased accessibility of probes. This is usually achieved
ISH is used in some diagnostic and research laboratories to by addition of detergents (Triton X-100 or Tween-20, Sigma, St.
identify avian viruses (2, 6, 7, 39-41, 55, 56, 66, 76, 81). The Louis, Mo.) to the various wash solutions (44).
application of ISH techniques can be limited by their inability to For developing successful in situ hybridization procedures, the
detect viral targets with low copies of DNA or RNA. However, melting temperature of the double-stranded hybrids, which is based
during the last few years, several strategies have been developed to on the nucleotide com position, must be known and incubation
improve the sensitivity of ISH by amplification of either target temperature controlled. Once hybridization has occurred the
nucleic acid sequences before ISH by in situ PCR, or signal unbound probe must be removed. The sensitivity and specificity of
detection after hybridization has been completed (e.g. signal the test is a function of how many probe molecules hybridize to the
amplification by the use of tyramine)(63). actual target vs. nonspecific sites and how strongly the probe binds.
The availability of non-radioactive-labeled probes and The set of experimental conditions such as salt concentrations and
commercially available hybridization kits and reagents has temperature that can affect the binding properties of a related, but
237
Mary J. Pantin-Jackwood and Sandra S. Rosenberger

not homologous, probe is termed stringency. Under low stringency 12. Burkhalter, K. L., R Lindsay, R. Anderson, A. Dibemardo, W. Fong,
some mismatch of nucleotides between the target and probe is and R. S. Nasci. Evaluation of commercial assays for detecting West Nile
allowed, whereas under high stringency very few mismatched pairs virus antigen. J. Am. Mosq. Control Assoc. 22:64-69. 2006.
13. Cartum, R. C. Immunohistochemistry of infectious diseases. J
will form (44).
Histotechnology 18:195-202. 1995.
14. Cattoli, G., A. Drago, S. Maneiro, A Toffan, E. Bertoli, S. Fassina, C.
Immunohistologic Staining Terregino, C. Robbi, G. Vicenzoni and I. Capua. Comparison of three rapid
Once hybridization occurs and all unbound probe is washed away, detection systems for type A influenza virus on tracheal swabs of
the antigen is detected by visualizing the labeled probe. This can be experimentally and naturally infected birds. Avian Pathol. 33(4):432-437.
done directly or indirectly as described in ISH. In the direct method, 2004.
biotin-labeled probes are detected by adding streptavidin conjugated 15. Chen, Β. Y, S. Hosi, T. Nunoya, and C.Itakura. Histopathology and
to an enzyme and then incubation with appropriate substrate. More immunohistochemistry of renal lesions due to infectious bronchitis virus in
chicks. Avian Pathol. 25:269-283. 1996.
amplification of the signal can be obtained by using the indirect
16. Cruz-Coy, J. S., J. J. Giambrone, and F. J. Hoerr. Immunohistochemical
method and a monoclonal anti-biotin antibody in the first step (6). detection of infectious bursal disease virus in formalin-fixed, paraffin-
This is followed by biotinylated anti-mouse IgG, which increases embedded chicken tissues using monoclonal antibody. Avian Dis. 37:577-
the amount of biotin associated with the probe. Streptavidin 581. 1993.
conjugated to an enzyme is then added, which binds to all available 17. Cui, Z. Z., L. F. Lee, E. J. Smith, R. L. Witter, and T. S. Chang.
biotin amplifying the antigen detection signal. Probes labeled with Monoclonal-antibody-mediated enzyme-linked immunosorbent assay for
digoxigenin and detected by anti digoxigenin-alkaline-phosphatase detection of reticuloendotheliosis viruses. Avian Dis. 32:32-40. 1988.
conjugate have also been used in the indirect detection system (66). 18. Davidson, I., M Malkinson, C. Strenger, and Y. Becker. An improved
ELISA method, using a streptavidin-biotin complex, for detecting Marek's
Another type of probe has been developed using fluorescein that is
disease virus antigens in feather-tips of infected chickens. J. Virol. Methods
detected by anti-fluorescein-alkaline-phosphatase conjugate. It is 14:237-241. 1986.
thought that the latter method may further reduce nonspecific 19. Davison, S., A. F. Ziegler, and R. J. Eckroade. Comparison of an
reactions because fluorescein is not a naturally occurring antigen-capture enzyme immunoassay with virus isolation for avian
component of the host system (44). influenza from field samples. Avian Dis. 42:791-795. 1998.
20. Dea, S., and S. Garzon. Identification of coronaviruses by the use of
Controls indirect protein A-gold immunoelectron microscopy. J. Vet. Diagn. Invest
ISH has several trouble spots; therefore, several issues must be 3:297-305. 1991.
21. Dhinakar Raj, G., S. Sivakumar, K. Matheswaran, M Chandrasekhar,
addresses in adopting a protocol (53). The design and use of V. Thiagarajan, and K. Nachimuthu. Detection of egg drop syndrome virus
positive and negative controls specific for a particular ISH protocol antigen or genome by enzyme-linked immunbsorbent assay or polymerase
is very important. Some controls relate primarily to the staining and chain reaction. Avian Pathol. 32:545-550. 2003.
signal generating system and others to the hybridization (53). 22. Fenner, F., P. Bachmann, E. Gibbs, F. Murphy, M Studdert, and D.
White. Laboratory Diagnosis of Viral Diseases. In: Veterinary Virology.
REFERENCES Academic Press, Inc., San Diego,CA. pp 237-264. 1987.
23. Fitzgerald, S. D., W. M Reed, and T. Bumstein. Detection of type Π
1. IHC World. Online Information Center for Immunohistochemistry. avian adenoviral antigen in tissue sections using immunohistochemical
http://www.ih cworld. com staining Avian Dis. 36:341-347. 1992.
2. Abbas, F., J. R. Andreasen, Jr., and M W. Jackwood. Development of a 24. Fitzgerald, S. D., W. M Reed, K. A. Langheinrich, A. S. Porter, and L.
polymerase chain reaction and a nonradioactive DNA probe for infectious A. Lumbert. A retrospective immunohistochemical study of type Π avian
laryngotracheitis virus. Avian Dis. 40:56-62. 1996. adenoviral infection in turkey, pheasant, and chicken tissues. Avian Dis.
3. Abdelmoumen, Β. B., and R. S. Roy. An enzyme-linked immunosorbent 38:78-85. 1994.
assay for detection of avian mycoplasmas in culture. Avian Dis. 39:85-93. 25. Fitzgerald, S. D., and A. Richard. Comparison of four fixatives for
1995. routine splenic histology and immunohistochemical staining for group Π
4. Adair, Β. Μ, K. Bums, and E. R. McKillop. Serological studies with avian adenovirus. Avian Dis. 39:425-431. 1995.
reoviruses in chickens, turkeys and ducks. J Comp Pathol 97:495-501. 1987. 26. Gancz, A Y, D. G. Campbell, I. K. Barker, R. Lindsay, and B. Hunter.
5. Alexandrov, M, R. Alexandrova, I. Alexandrov, S. Zacharieva, S. Detecting West Nile virus in owls and raptors by an antigen-capture assay.
Lasarova, L. Doumanova, R. Peshev, and T. Donev. Fluorescent and Emerg. Infect. Dis. 10:2204-2206. 2004.
electron-microscopy immunoassays employing polyclonal and monoclonal 27. Gilka, F., and J. L. Spencer. Immunohistochemical identification of
antibodies for detection of goose parvovirus infection. J. Virol. Methods group specific antigen in avian leukosis virus infected chickens. Can. J.
79:21-32. 1999. Comp. Med. 48:322-326. 1984.
6. Allan, G. M, D. Todd, J. A. Smyth, D. P. Mackie, J. Bums, and M S. 28. Guy, J. S., H.J. Barnes, A.L.G. Smith. Rapid diagnosis of infectious
McNulty. In situ hybridization: an optimized detection protocol for a laryngotracheitis using a monoclonal antibody-based immunoperoxisase
biotinylated DNA probe renders it more sensitive than a comparable 35S- procedure. Avian Pathol. 21:77-86. 1992.
labelled probe. J. Virol. Methods 24:181-190. 1989. 29. Haines, D. M and B. J. Chelack. Technical considerations for
7. Arshad, S. S., L.M Smith, K. Howes, P.H.Russell, K. Venugopal, L.N. developing enzyme immunohistochemical staining procedures on formalin-
Payne. Tropism of subgroup J avian leukosis virus as detected by in situ fixed paraffin-embedded tissues for diagnostic pathology. J. Vet. Diagn.
hybridization. Avian Pathol. 28. 1999. Invest. 3:101-112. 1991.
8. Balamurugan, V., J. M Katari, A. K. Tiwari, K. C. Verma, R. Toroghi, 30. Hammond, G. W., P. R. Hazelton, I. Chuang, and B. Klisko. Improved
and S. J. Jadhao. Development of sandwich ELISA for the detection of fowl detection of viruses by electron microscopy after direct ultracentrifuge
adenovirus 4 associated with hydropericardium syndrome in experimentally preparation of specimens. J. Clin. Microbiol. 14:210-221. 1981.
infected chicken. Acta Virol. 45:95-100. 2001. 31. Harvey, R. In situ hybridization. In: Handbook of Immunochemical
9. Bhattachaijee, P. S., C.S. Naylor,and RC. Jones. A simple method for staining methods. 3rd edition. M S. Boenisch, ed. DakoCytomation,
immunofluorescence staining of tracheal organ cultures for the rapid Carpinteria, CA. pp 41-43. 2001.
identification of infectious bronchitis virus. Avian Pathol. 23:471-480. 1994. 32. Hassan, Μ K., Y. M Saif, and S. Shawky. Comparison between
10. Bratthauer, G. L. Processing of Tissue Specimens. In: Methods in antigen-capture ELISA and conventional methods used for titration of
Molecular Biology: Immunocytochemical Methods and Protocols. L. C. infectious bursal disease virus. Avian Dis. 40:562-566. 1996.
Javois, ed. Humana Press, Totowa, NJ. pp 77-86. 1999. 33. Hayhow, C. S., and Y. M Saif. Development of an antigen-capture
11. Breslin, J. J., L. G. Smith, H. J. Barnes, and J. S. Guy. Comparison of enzyme-linked immunosorbent assay for detection of enterovirus in
virus isolation, immunohistochemistry, and reverse transcriptase-polymerase commercial turkeys. Avian Dis. 37:375-379. 1993.
chain reaction procedures for detection of turkey coronavirus. Avian Dis. 34. Holland, M S., C. D. Mackenzie, R. W. Bull, and R. F. Silva A
44:624-631.2000. comparative study of histological conditions suitable for both
immunofluorescence and in situ hybridization in the detection of

238
Chapter 49 Antigen Detection Systems

Herpesvirus and its antigens in chicken tissues. J. Histochem. Cytochem. 56. Oldoni, I., C. C. Brown, D. J. King, S. Samal, and B. S. Seal. The use of
44:259-265. 1996. in situ hybridization and immunohistochemistry to study the pathogenesis of
35. Hooper, P. T., G. W. Russell, P. W. Selleck, and W. L. Stanislawek. various Newcastle disease virus strains and recombinants in embryonated
Observations on the relationship in chickens between the virulence of some chicken eggs. Microb. Pathog. 39:69-75. 2005.
avian influenza viruses and their pathogenicity for various organs. Avian 57. Oltishi, K., M Senda, H. Yamamoto, H. Nagai, M Norimatsu, and H.
Dis. 39:458-464. 1995. Sasaki. Detection of avian encephalomyelitis viral antigen with monoclonal
36. Hussain, I., and K. V. Nagaraja. A monoclonal antibody-based antibody. Avian Pathol. 23:49-59. 1994.
immunoperoxidase method for rapid detection of haemorrhagic enteritis 58. Pantin-Jackwood, M J., and T. P. Brown. Infectious bursal disease
virus of turkeys. Res. Vet. Sci. 55:98-103. 1993. virus and proventriculitis in broiler chickens. Avian Dis 47:681-690. 2003.
37. Islam, M R., and M Khan. An immunocytochemical study on the 59. Pantin-Jackwood, M J., E. Spackman, and J. M Day. Pathology and
sequential tissue distribution of duck plague virus. Avian Pathol. 24:189- virus tissue distribution of Turkey origin reoviruses in experimentally
194. 1995. infected Turkey poults. Vet Pathol. 44:185-195. 2007.
38. Jiijis, F. E., S. L. Noll, D. A. Halvorson, K. V. Nagaraja, and D. P. 60. Patel, B. L., E. Gonder, and B. S. Pomeroy. Detection of turkey
Shaw. Immunohistochemical detection of avian pneumovirus in formalin- coronaviral enteritis (bluecomb) in field epiomithics, using the direct and
fixed tissues. J. Vet. Diagn. Invest. 13:13-16. 2001. indirect fluorescent antibody tests. Am. J. Vet. Res. 38:1407-1411. 1977.
39. Kapczynski, D. R., H. S. Sellers, G. N. Rowland, and M W. Jackwood. 61. Pereira, S. R., C. E. Travassos, A. Huguenim, A. C. Guimaraes, A. G.
Detection of in ovo-inoculated infectious bronchitis virus by Silva, and M. A. Guimaraes. Western blot detection of infectious bursal
immunohistochemistry and in situ hybridization with a riboprobe in disease virus infection. Braz J Med Biol Res 31:671-674. 1998.
epithelial cells of the lung and cloacal bursa. Avian Dis. 46:679-685. 2002. 62. Polak, J.M and S. Van Noorden. Introduction to Immunocytochemistry.
40. Keam, L., J. J. York, M Sheppard, and K. J. Fahey. Detection of 3rd. edition. Bios Scientific Publishers Limited, Oxford, UK. 2003.
infectious laryngotracheitis virus in chickens using a non-radioactive DNA 63. Qian, X., L. Jin, and R. V. Lloyd. In situ hybridization: Basic approaches
probe. Avian Dis. 35:257-262. 1991. and recent developments. J. of Histotechnology 21(l):53-67. 2004.
41. Liu, X., J. J. Giambrone, and E. J. Hoerr. In situ hybridization, 64. Radi, Z. A., D. W. Trampel, B. S. Smith, R. F. Rosenbusch, and F. Goll.
immunohistochemistry, and in situ reverse transcription-polymerase chain Immunohistochemical detection of Mycoplasma gallisepticum antigens in
reaction for detection of infectious bursal disease virus. Avian Dis. 44:161- turkey respiratory tissues. Avian Dis. 44:399-407. 2000.
169. 2000. 65. Raj, G. D., V. Jayakumar, A. Thangavelu, A. Koteeswaran, and A. T.
42. Lockaby, S. B., F. J. Hoerr, A. C. Ellis, and M S. Yu. Venugopalan. Immunorrheophoresis for the diagnosis of infectious bursal
Immunohistochemical detection of Newcastle disease virus in chickens. disease. Avian Dis. 42:388-392. 1998.
Avian Dis. 37:433-437. 1993. 66. Ramis, A., K.S.Latimer, F.D. Niagro, R.P. Campagnoli, B.W. Ritchie,
43. Lu, H. A longitudinal study of a novel dot-enzyme-linked and D. Pesti. Diagnosis of psittacine beak and feather disease (PBFD) viral
immunosorbent assay for detection of avian influenza virus. Avian Dis. infection, avian polyiomavirus infection, adenovirus infection and
47:361-369. 2003. herpesvirus infection in psittacine tissues using DNA in situ hybridization.
44. Magraff, L. R., and D. Jennings-Siena. In-situ hybridization and Avian Pathol. 23:643-657. 1994.
molecular pathology for the histotechnician. In: Proceedings of the National 67. Ramis, A., J. Tarres, D. Fondevila, and L. Ferrer. Immunocytochemical
Society for Histotechnology. Workshop 71. Albuquerque, NM pp 1-30. study of the pathogenesis of Pacheco's parrot disease in budgerigars. Vet.
1996. Microbiol. 52:49-61. 1996.
45. McCourt, Μ T., D. A. Finlay, C. Laird, J. A. Smyth, C. Bell, and H. J. 68. Ramos-Vara, J. A. Technical aspects of immunohistochemistry. Vet.
Ball. Sandwich ELISA detection of Clostridium perfringens cells and alpha­ Pathol. 42:405-426. 2005.
toxin from field cases of necrotic enteritis of poultry. Vet. Microbiol. 69. Ramos-Vara, J. A., F. De Piero, M Kiupel, S.D. Fitzgerald, A.J.
106:259-264. 2005. Bermudez,G.C. Johnson, M.A. Miller. Diagnostic immunohistochemistry of
46. McNeilly, F., G.M Allan, D.A. Moffett, and MS. McNulty. Detection equine and avian infectiuos diseases. Journal of Histotechnology 25 (4): 185-
of chicken anemia agent in chickens by imunofluorescence and 198. 2002.
immunoperoxidase staining. Avian Pathol. 20:125-132. 1991. 70. Roy, P., and A. T. Venugopalan. Agar-gel-immunodiffusion and
47. McNulty, M S., and G.M Allan. Application of immunofluorescence in counter immunoelectrophoresis for diagnosis of Newcastle disease. Trop.
veterinary viral diagnosis. In: Recent advances in virus diagnosis. M S. Anim. Health Prod. 29:231-234. 1997.
McNulty and J.B. McFerran, eds. Martinus Nijhoff, Dovdrecht, The 71. Ruano, M, J. El-Attrache, and P. Villegas. A rapid-plate
Netherlands, pp 15-26. 1986. hemagglutination assay for the detection of infectious bronchitis virus.
48. Miller, S. E. Diagnosis of Viral Infections by Electron Microscopy. In: Avian Dis. 44:99-104. 2000.
Diagnostic Procedures for Viral, Rickettsial and Chlamydial Infections. 7th 72. Ryan-Poirier, K. A., J. M Katz, R. G. Webster, and Y. Kawaoka.
Edition. Lennette. E.H., D. A. Lennette and E. T. Lennette., eds. American Application of Directigen FLU-A for the detection of influenza A virus in
Public Health Association, Washington, DC. pp 37-93. 1995. human and nonhuman specimens. J. Clin. Microbiol. 30:1072-1075. 1992.
49. Muneer, M A., J. A. Newman, V. Sivanandan, and D. A. Halvorson. A 73. Sapats S., G. G., L. Trinidad, L.H. Parede, C. David, J Ignjatovic. An
dot-immunobinding assay for infectious bronchitis virus. Avian Dis. 32:137- ELISA for detection of infectious bursal disease virus and differentiation of
139. 1988. very virulent strains based on single chain recombinant chicken antibodies.
50. Nagano, Η., M Tsuchimoto, T. Hohdatsu, T. Yamagami, S. Ide, Y. Avian Pathol. 34(6):449-455. 2005.
Eiguchi, Y. Tanaka, and H. Yamagishi. Enzyme-linked immunosorbent 74. Saravanan, P., Satishkumar, J. M Kataria, and T. J. Rasool. Detection
assay for the detection of infectious bronchitis virus (IBV) antigen with of Infectious bursal disease virus by ELISA using an antipeptide antibody
monoclonal antibody. Nippon Juigaku Zasshi 52:657-659. 1990. raised against VP3 region. Acta Virol. 48:39-45. 2004.
51. Naqi, S. A. A monoclonal antibody-based immunoperoxidase procedure 75. Selleck, P. W., S. L. Lowther, G. M Russell, and P. T. Hooper. Rapid
for rapid detection of infectious bronchitis virus in infected tissues. Avian diagnosis of highly pathogenic avian influenza using pancreatic impression
Dis. 34:893-898. 1990. smears. Avian Dis. 47:1190-1195. 2003.
52. Naqi, S. A., K. Karaca, and B.Bauman. A monoclonal base antigen 76. Sellers, H. S., P. N. Villegas, J. El-Attrache, D. R. Kapczynski, and C.
capture enzyme-linked immunosorbent assay for identification of infectious C. Brown. Detection of infectious bursal disease virus in experimentally
bronchitis virus serotypes. Avian Pathol. 22:555-564. 1993. infected chickens by in situ hybridization. Avian Dis. 45:26-33. 2001.
53. Nardone, R. M Overview of In Situ Hybridization. In: 77. Shafren, D. R., and G. A. Tannock An enzyme-linked immunosorbent
Immunocytochemical Methods and Protocols. L. C. Javois, ed. Humana assay for the detection of avian encephalomyelitis virus antigens. Avian Dis.
Press, Totowa, NJ. pp 357-414. 1999. 32:209-214. 1988.
54. Nazerian, K., L. F. Lee, and W. S. Payne. A double-antibody enzyme- 78. Smyth, J. A., D. A. Moffett, M S. McNulty, D. Todd, and D. P.
linked immunosorbent assay for the detection of turkey hemorrhagic Mackie. A sequential histopathologic and immunocytochemical study of
enteritis virus antibody and antigen. Avian Dis. 34:425-432. 1990. chicken anemia virus infection at one day of age. Avian Dis. 37:324-338.
55. Nielsen, O. L., P.H. Jorgensen, M Bisgaard, and S. Alexandersen. In 1993.
situ hybridization for the detection of chicken anemia virus in 79. Snyder, D. B., D. P. Lana, P. K. Savage, F. S. Yancey, S. A. Mengel,
experimentally-induced infections and field outbreaks. Avian Pathol. and W. W. Marquardt. Differentiation of infectious bursal disease viruses
24:149-155. 1995. directly from infected tissues with neutralizing monoclonal antibodies:

239
Mary J. Pantin-Jackwood and Sandra S. Rosenberger

evidence of a major antigenic shift in recent field isolates. Avian Dis. histological changes after eyedrop vaccination. Avian Dis. 40:114-1201
32:535-539. 1988. 1996.
80. Spencer, J. L., and F. Gilka. Lymphoid leukosis: detection of group 87. Tripathy, D. N., L. E. Hanson, and A. H. Killinger. Immunoperoxidee
specific viral antigen in chicken spleens by immunofluorescence and technique for detection of fowlpox antigen. Avian Dis. 17:274-278. 1973.
complement fixation. Can. J. Comp. Med. 46:370-375. 1982. 88. Tsukamoto, K., H. Hihara, and Y. Kono. Detection of avian leukosis
81. Stedman, N. L., T. P. Brown, and C. C. Brown. Localization of avian virus antigens by the ELISA and its use for detecting infectious virus after
leukosis virus subgroup J in naturally infected chickens by RNA in situ cultivation of samples and partial characterization of specific pathogen-free
hybridization. Vet. Pathol. 38:649-656. 2001. chicken lines maintained in this laboratory. J. Vet. Med. Sci. 53:399-4O&.
82. Storch, G. A. Diagnostic Virology. In: Fields Virology. D. A. a. P. M 1991.
H. Knipe, ed., Philadelphia, PA. pp 501-505. 2001. 89. Tsukamoto, K., T. Matsumura, M. Mase, and K. Imai. A highly
83. Swayne, D. E., MD. Ficken, and J.S. Guy. Immunohistochemical sensitive, broad-spectrum infectivity assay for infectious bursal disease
demonstration of influenza A nucleoprotein in lungs of turkeys with natural virus. Avian Dis. 39:575-586. 1995.
and experimental influenza respiratory disease. Avian Pathol. 21:547-557. 90. Wool cock, P. R, and C. J. Cardona. Commercial immunoassay kits for
1992. the detection of influenza virus type A: evaluation of their use with poultry.
84. Tanimura, N., K. Tsukamoto, K. Nakamura, M Narita, and M Maeda. Avian Dis. 49:477-481. 2005.
Association between pathogenicity of infectious bursal disease virus and 91. Yagyu, K., and S. Ohta. Enzyme-linked immunosorbent assay for die
viral antigen distribution detected by immunohistochemistry. Avian Dis. detection of infectious bronchitis virus antigens. Nippon Juigaku Zasshi
39:9-20. 1995. 49:757-763. 1987.
85. Thomas, C. B., and P. Sharp. Detection of antigenic variation among 92. Yilmaz, F., N. Timurkaan, and H. Bulut. Detection of infectious
strains of Mycoplasma gallisepticum by enzyme-linked immunosorbent laryngotracheitis virus in trigeminal ganglia by avidin-biotin complex
inhibition assay (ELISA) and Western blot analysis. Avian Dis. 32:748-756. method in chickens: short communication. Acta Vet. Hung. 52:167-171.
1988. 2004.
86. Toro, Η., V. Godoy, J. Larenas, E. Reyes, and E. F. Kaleta. Avian
infectious bronchitis: viral persistence in the harderian gland and

240
APPENDIX OF ABBREVIATIONS AND ACRONYMS USED IN THE TEXT

A/A Acid slant/acid butt in media tube (avian adenovirus)


AAF Amniotic-allantoic fluid CEO Chicken embryo Origin
AAS Avian adenovirus group Π splenomegaly CET Chick embryo tendon (cells)
AAVLD American Association of Veterinary CE-YS Chick embryo-Yolk sac route
Laboratory Diagnosticians CF Complement fixation
ABC Avidin-biotin-peroxidase complex CGD Cold Gelatin Diluent
AC Anticomplement CK Chicken kidney (cells)
AC-ELISA Antigen-capture ELISA CKC Chicken kidney cell
AE Avian encephalomyelitis CKN Chicken
AEC 3-Amino-9-ethylcarbazole CM Conditioned medium
AEV Avian encephalomyelitis virus CMGS Chopped meat-glucose-starch (media)
AFLP Amplified Fragment Length Polymorphism CNA Colistin Nalidixic Acid
AGID Agar-gel immunodiffusion (test) C/E CELLS CEF resistant to subgroup E ALV
AGP Agar-gel precipitin C/O cells CEFs susceptible to avian leukosis
Al Avian influenza viruses in subtypes A,B,C,D,E,J
AIS Avian intestinal spirochetosis COFAL Complement fixation for avian leukosis
AIV Avian influenza virus COFAR Complement fixation for avian
ALV Avian leukosis virus reticuloendotheliosis
AOAC Association of Official Analytical Chemists CPE Cytopathic effect
AP Alkaline phosphatase CRBC Chicken red blood cell
APEC Avian Pathogenic Escherichia coli CSM Charcoal-selective medium (agar)
APHIS Animal & Plant Health Inspection Service CVA Cefoperazone, vancomycin,
AP-PCR Arbitrarily primed PCR Amphotericin (agar)
APMV-1 Avian paramyxovirus serotype 1 DAB 3,3’ diaminobenzidine tetrachloride
AR Antigen retrieval DAS Diacetoxy scirpenol
ART Avian rhinotracheitis DBH Dot Blot Hybridization
BA Bordetella avium DEAE Diethylaminoethyl
BAM Bacteriological Analytical Manual DEF Duck embryo fibroblast (cells)
BCIP/NBT 5 -bromo-4-chloro-3 -indolylphosphate/ DEK Duck embryo kidney (cells)
Nitro blue tetrazoliumchloride DEL Duck embryo liver (cells)
BD Bactoagar DEPC Dethylpyrocarbonate
BDU 5'-bromo-2'-deoxyuridine DFA Direct fluorescent antibody (test)
BEF Budgerigar embryo fibroblast (cells) DHV Duck hepatitis virus
BEM Broth enrichment medium DID Double immunodiffusion (test)
BG Brilliant green (agar) DMEM Dulbecco’s modified Eagle’s medium
BGN Brilliant green supplemented with DMSO Dimethyl sulfoxide
novobiocin (agar) DNA Deoxyribonucleic acid
BGS Brilliant green supplemented with DON Deoxynivalenol
sulfapyridine or sulfadiazine (agar) DS Double stranded (DNA)
BLAST Basic Local Alignment Search Tool DSA Dextrose starch agar
BLS-3 Biosafety level 3 DSE Delayed secondary enrichment
BPL Beta Propriolactone DSSM Double-strength skim milk
BME Basal medium, Eagle’s DVE Duck virus enteritis
BPW Buffered peptone water EBA Elementary body agglutination (test)
BSA Bovine serum Albumin EDTA Ethylene diaminetetraacetic acid
BSE Bovine Spongiform Encephalopathy EDS Egg drop syndrome
BSK B arbour-Stoenner-Kelly medium EDS 76 Egg drop syndrome 76
BSS Balanced salt solution EEE Eastern equine encephalitis
C Cytosine EEEV Eastern equine encephalitis virus
CAV Chicken anemia virus EFSA European Food safety Authority
CAM Chorioallantoic membrane EID50 Mean embryo infectious dose
CAMP Christie, Atkins, and Muench-Peterson ELD50 Mean embryo lethal dose
(media) ELISA Enzyme-linked immunosorbent assay
CAT Cefoperazone, Amphotericin, ELISA-ALV ELISA for ALV
Teicoplanin agar EM Electron microscopy
CDC Center for Disease Control EMEM Eagle’s minimum essential medium
CE Chick embryo EMJH Ellinghausen-McCullaugh-Johnson-
CE-CAM Chick embryo-Chorioallantoic membrane route Harris (Medium)
CED Chick embryo dermal (cells) ERIC Enterobacterial Repetitive Intergenic
CEF Chick embryo fibroblast (cells) Consensus-based
CEK Chick embryo kidney (cells) ESB Erysipelothrix selective broth
CEL Chick embryo liver (cells) ETOH Ethyl alcohol
CELC Chick embryo liver cells EU European Union
CELO Chick embryo lethal orphan F Fusion protein
241
Appendix of abbreviations and acronyms

FA Fluorescent antibody (test) LL Lymphoid leukosis


FAdV Fowl Adenovirus L/S Leukosis/sarcoma
FBS Fetal bovine serum LPD Lymphoproliferative disease
FC Flow cytometry LPM Lithium chloride-phenyl
FCS Fetal calf serum ethanol-moxalactan (agar)
FDA Food and Drug Administration LPS Lipopolysaccharide
FFU Focus-forming unit LTR Long terminal repeat
FITC Fluorescein isothiocyanate LSAB Streptavidin-Biotin-Peroxidase complex
FRET Fluorescence Resonance Energy Transfer M Matrix protein
G Guanine MA Micro agglutination
GC-MS Mass spectral Gas Chromatography mAbs Monoclonal antibodies
GDPT Gel-diffusion precipitin test MAC MacConkey agar
GMS Gomori mehenamine silver MAC Mycobacterium avium Complex (2 species)
GMT Geometric mean titer MAC-X M. avium Complex (3 species)
GPV Goose parvovirus mCCDA Modified Charcoal Cefoperazone
GS Group-specific Deoxycholate agar
GTS Gene targeted sequencinng MD Marek’s disease
H Hemagglutinin MDEF Muscovy duck embryo fibroblast (cells)
H Flagellar antigens (chapter 10 only) MDPV Muscovy duck parvovirus
H&E Hematoxylin and Eosin MDT Mean death time
HA Hemagglutination MDV Marek’s disease virus
HBSS Hanks’ balanced salt solution MEE Multilocus enzyme electrophoresis
HE Hemorrhagic enteritis MEM Mnimum essential medium
HEV Hemorrhagic enteritis virus MG Mycoplasma gallisepticum
HI Hemagglutination inhibition MLEE Multi locus Enzyme Electrophoresis
HJ Highlands J (virus) MM Miller Mallinson media
HMN Haemophilus maintenance medium MS Mycoplasma synoviae
HNEG Hemorrhagic nephritis and enteritis of geese MSDV Marble Spleen Disease Virus
HN protein Protein with hemagglutination and mMDV Mildly virulent MDV
neuraminidase activities MLIA Modified lysine iron agar
HPAl Highly pathogenic avian influenza MOX Modified Oxford (agar)
HPLC High Pressure Liquid Chromatography MSD Marble spleen disease
HPS Hydropericardium syndrome MW Molecular weight
HRP Horseradish peroxidase N Neuraminidase
HVT Herpes virus of turkey NAD Nicotinamide adenine dinucleotide
HV Herpes virus NBT Nitroblue tetrazolium
IB Infectious bronchitis ND Newcastle disease
IBD Infectious bursal disease NDV Newcastle disease virus
IBH Inclusion body hepatitis N Neuraminidase inhibition (test)
IBDV Infectious bursal disease virus N Neutralization index (chapter 46 only)
IBV Infectious bronchitis virus NGFIS Netherlands Government Food
ICPI Intracerebral pathogenicity index Inspection Service
ICTV International Committee on the NP Non-producer
Taxonomy of Viruses NPIP National Poultry Improvement Plan
ID50 Mean infective dose NVSL National Veterinary Services Laboratory
IDU 5’-iodo-2’-deoxyuridine O Somatic antigens (chapter 10 only)
IEM Immune electron microscopy OIE Office International des Epizooties
IF Immunofluorescence ONPG OrrAo-Nitrophenyl-p-galactoside
IFA Indirect fluorescent antibody (test) PALCAM Polymyxin-acriflavin-lithium chloride-
IgG Immunoglobulin G ceftazidime-esculin-mannitol (agar)
IgM Immunoglobulin M PAS Periodic Acid Schiff
HIS Immunohistochemical staining PBFD Psittacine beak and feather disease
IIF Indirect immunofluorescence PBS Phosphate-buffered saline
ILT Infectious laryngotracheitis PBS-B-G PBS with BSA and gelatin
ILTV Infectious laryngotracheitis virus PCR Polymerase chain reaction
IM Intramuscular PD Proportionate distance
π* Immunoperoxidase PD Protective Dose (Chapter 46)
ISH in situ hybridization pd50 Mean protective dose
ISHH in situ hybridization histochemistry PEA Phenylethyl alcohol agar
ISO International Standards Organization PEMS Poult enteritis mortality syndrome
IV Intravenous PFGE Pulse Field Gel Electrophoresis
IVPI Intravenous pathogenicity index PFU Plaque or pock forming unit
LA Latex agglutination (test) PI Post inoculation
LD50 Mean lethal dose PID50 Mean poultry infectivity dose
LH Lactalbumin hydrolysate PLB Pagano Levin Base media
LI Lysine iron PM Phenotypic mixing (chapter 35 only)
LJ Lowenstein-Jensen agar PM Pasteurella multocida
242
Appendix of abbreviations and acronyms

PRN Plaque reduction neutralization (test) RNP Ribonucleic protein


PSI Pounds per square inch RO Reverse osmosis
PTA Phosphotungstic acid RIF Resistance inducing factor (test)
PVK Pierce-Van Der Kamp (stain) RSV Rous sarcoma virus
QBV Quail Bronchitis Virus RT Reverse transcriptase
RAPD Random amplified polymorphic DNA RT-PCR Reverse transcriptase-polymerase chain
RAV-O Rous-associated virus reaction
RBC Red blood cell RV Rappaport-Vassiliadis (broth)
REs Restriction enzymes SC Subcutaneous
REA Restriction enzyme analysis SDS Sodium dodecyl sulfate
RFLP Restriction fragment length polymorphism SE Salmonella enteritidis
REO Avian reovirus SHS Swollen head syndrome
REV Reticuloendotheliosis virus SIM Sulfide-indole-motility (medium)
RNA Ribonucleic acid SP Sample-positive ratio
rRNA Ribosomal RNA SPA Serum plate agglutination
SPF Specific-pathogen-free
SPG Sucrose-phosphate-glutamate
SPGA Sucrose-phosphate-glutamine albumin
SSA Stunting syndrome agent
T Thymine
TBE Tris borate Buffer
TBS Tri-buffered saline
TBTB Tris-buffered tryptose broth
TC Tissue culture
TCoV Turkey coronavirus
TCID Tissue culture infective doses
TCID50 Mean tissue culture infective dose
TEys Turkey embryo yolk sac
TEM Transmission electron microscopy
TME Turkey meningoencephalitis *
THEV Turkey Hemorrhagic enteritis virus
TLC Thin layer chromatography
TM/S Supplemental test medium without NAD
TM/SN TM/S supplemented with NAD
TPB Tryptone phosphate broth
TSA Tryptic soy agar
TS Triple sugar iron
TT Tetrathionate (broth)
TTC 2,3,5-triphenylterazolium chloride
TOC Tracheal organ culture
TV Trypsin and versene
TVH Turkey viral hepatitis
U Uracil
UPGMA Unweighed paired group method
Using arithmetic averages
USDA United States Department of Agriculture
UV Ultraviolet (light)
UVM University of Vermont
vMDV Very virulent MDV
VN Virus neutralization (test)
wMDV Very, very virulent MDV
w+MDV Greater virulence than wMDV
WBP Whole blood plate
WEE Western equine encephalitis
WN West Nile
XLD Xylose-lysine-desoxycholate (agar)
XLDN XLD supplemented with novobiocin
XLT4 Xylose-lysine-tergitol 4 (agar)

243
QUICK REFERENCE DIAGNOSTIC CHART: BACTERIAL and FUNGAL DISEASES, MYCOTOXINS

Molecular Serogrouping, serotyping,


Pathogen CH Preferred Isolation method or diagnostic test Host Serology
Diagnosis speciation, pathogenicity
A. gallinarum 4 Isol. BA, DSA not useful
Serotyping: PAGE (Aggl. or
A. para­ Isol. BA (with Staph nurse culture), CE-YS, HI), KUME (HI using
6 HP2-PCR HI
gallinarum Ckn inoc. treated bacterial cells and
rbcs), ERIC PCR
B. avium 5 Isol. MAC ELISA and MA
1. Wet smears and microscopy
Borrelia anserina 8 Aggl., FA
2. Isol. BSK, Ckn inoc, CE-YS, TE-YS
AGID, ELISA,
Brachyspira sp. 8 Isol. BA, anaerb. 2S-N-D-PCR
MA, IFA
1. mouse bioassay
C. botulinum 13 PCR not useful
2. Isol. CMGS, CGD
1 Isol. TPA with glucose and yeast (anaerb)
C. colinum 13 CF
2 FA, AGID
1. Isol. BA (anaerb), PEA Toxin typing: Multiplex
C. perfringens 13 not useful
2. FA PCR
PCR, DNA Phenotypic and molecular
Campylobacter 7 Isol. Campy-cefex, CVA, mCCDA not useful
probes based typing
1. Isol. CC confirmed FA;
Chlamydophila 16 2. Isol. CE-YS; confirmed PCR CF, LA, EBA
3. Histo staining
Serogrouping (0 antigen)
E. coli 3 Isol. MAC, and TSI, SIM, BA and serotyping (0 and not useful
Flagellar &/or capsular Ag)
Antigenic typing by mouse
E. rhusopathiae 9 Isol. BA, MAC or TSI (CO2) PCR not useful
protection test
G. anatis b.
4 Isol. BA, DSA not useful
haemolytica
1. Isol. BA, 35 C.
2. USDA method: UVM sei. Enrichment, MOX
DNA probes, Speciation by DNA-DNA
Listeria mono­ and LTM plating. NGFIS method: prim,
10 Real time- hybridization, MLEE, not useful
cytogenes enrichment: PALCAM broth, Sec. Enrichment:
PCR RFLP, PFGE
PALCAM plate, microaerobic.
3. FA
1. Isol. LJ media
Molecular Typing: Aggl. ELISA,
Mycobacterium 14 2. Stained smears: Acid Fast Tuberculin
probes, PCR PFGE, RFLP
3. Fluorochrome stain
PCR, Gene
Mycoplasma 15 Isol. Frey, PPLO medium, plate on Agar, targeted FA, IFA SPA, ELISA, HI
sequencing
Ornithobacterium 17 Isol. BA Serotyping by AGID ELISA
Somatic serotyping: GDPT,
Pasteurella 4 Isol. BA, DSA capsular serogrouping: ELISA
passive hemAggl.
R. anatipestifer 4 Isol. BA or TSB with yeast (CO2) Serotyping: plate Aggl. ELISA
Serogrouping: somatic O-
1. Isol, using selective enrichment (eg TT), non DNA probes,
group antigen Aggl., WBPT, tube
selective pre-enrichment (eg BPW), selective PCR,
Salmonella 2 followed by single factor Aggl., micro
plating (BGN, XLT4, Rambach), and DSE (7 d) magnetic bead
Aggl.; Serotyping: Aggl., ELISA
2. AC-ELISA systems, etc.
microtube Aggl. test
Staphylococcus 11 Isol BA not useful
Streptococcus,
12 Isol BA, MAC FA, Aggl. not useful
Enterococcus
Y pseudo­
4 Isol. TSA or BA serotyping by Aggl. not useful
tuberculosis
Mycotoxins 18 TLC, HPLC, GC-MS, ELISA
1. Isol. SDA or PDA, direct microscopy
Aspergillus 18 not useful
2. Histo (PAS)
Dactylaria 18 Isol. SDA, direct microscopy not useful
Candida 18 Isol. PLB, micropscopic exam not useful
Favus 18 Isol. SDA, microsc. Exam not useful

244
QUICK REFERENCE DIAGNOSTIC CHART: VIRAL DISEASES

Molecular Serogrouping, serotyping,


Pathogen CH Preferred Isolation technique/Diagnostic test Host Serology
Diagnosis speciation, pathogenicity

1. Isol. CEK, TEK, CKC, followed by FA or EM HI (EDS),


Adenovirus 19 2. DEK for EDS, followed by HA, and HI ISH Sub-typing: REA, PCR, SN ELISA, IFA,
3. TCEM or ELISA for Ag detection in tissues AGID

1. Isol, by inoc newborn mice IC


Arbovirus (EEE,
40 2. Isol CEF, DEF, Vero, BHK21, RK13 cells; CF, PCR PRN, HI, ELISA
WEE, HJ, TME)
IFA, PRN, VN

1. See 1. under DHV 1


Astrovirus
37 2. Isol. CE-YS or DE-YS; death, neutralization
(DHVII)
3. TEM
RT-PCR,
multiplex RT-
Astrovirus (enteric) 34 1. Isol. CEL, followed by EM, FA, AGID VN, ELISA
PCR, ISH,
rRT-PCR
Avian
1. Isol. CE-AS, HA activity, VN
Paramyxovirus 30 Serotype ID: HI ELISA
2. Isol. CE-YS for PMV5
2-9

Avian Pathogenicity: IC test in day


RT-PCR,
Paramyxovirus-1 30 1. Inoc CE-AS, HA activity, VN old chicks; Strain variability ELISA, HI
rRT-PCR
(NDV) using Mab, sequencing

RFLP; Cross neutralization in


Bimavirus (IBD) 41 1. Isol. CE-CAM; AGP, FA, AC-ELISA RT-PCR embryos, cross challenge in ELISA, AGP, VN
chickens

DNA Probe,
Circovirus (PBFD) 27 PCR HI
PCR, ISH

IFA, AC-ELISA,
Coronavirus 1. Isol. TE-AS or CE-AS RT-PCR,
34 IFA, sequencing Competitive
(enteric) 2. EM, IEM, IFA, EHC, HA multiplex PCR ELISA, VN, HI

Serotype ID: VN, HI, RRT-


Coronavirus (IBV) 32 1. Isol. CE-AS, VN, HI, IHC PCR ELISA, HI, VN
PCR, RFLP

Coronavirus (TCV) 33 1. Isol. CE-AS, EM, FA, IHC, ELISA RT-PCR ELISA

Entervirus-like 1. Isol. TEF, EM IHC, IFA,


34 not recommended
(enteric) 2. EM on intestinal contents ELISA

1. Isol. MDCC-MSB1, MDCC-CU147, VN ISH, DBH,


Gyrovirus (CAV) 28 ELISA
2. Inoc suscb. Chicks, anemia PCR

1. Isol. MDEF, DEF, or DEL,death, lesions; FA ;


protection in DAZE vaccinated ducks following
challenge with virulent DVE virus
Herpesvirus (DVE) 23 PCR VN
2. Isol in 1 d old Muscovy ducks, FA
3. Isol. DE-CAM, FA
4. Neg. stain TEM (not specific to DVE)

1. Isol. In CEL, CEK, TOC, CE-CAM, CKN inoc.


2. Smears stained with Giemsa PCR/RFLP, reciprocal AGID, ELISA,
Herpesvirus (ILT) 21 PCR
3. EM DNA: DNA hybridization VN
4. Antigen detection: FA, IP, AGID

Herpesvirus PCR, ISH, AGID, VN, IFA,


22 1. Isol in cell culture, FA
(MDV) DBH IHC, ELISA

Herpesvirus of free 1. Isol. CEF, CEK, CEL, VN with known + antisera Pathogenicity in Emb. Ckn
24 PCR VN, ELISA
flying birds 2. Direct IF on tissue/impression smears eggs

245
QUICK REFERENCE DIAGNOSTIC CHART: VIRAL DISEASES

Molecular Serogrouping, serotyping,


Pathogen CH Preferred Isolation technique/Diagnostic test Host Serology
Diagnosis speciation, pathogenicity

1. Isol. CKN or TKY TOC, CE-YS, TE-YS, CEF,


Metapneumovirus 31 VERO, QT-35 PCR VN using monoclonals, ED ELISA, FA, VN
2. IP, IF, Ag detection

Subgroup by VN or flow ELISA, VN or


1. Isol. SPF CEFs resistant to subgroup E ALV and
Oncornavirus PCR cytometry, Pathogenicity in flow cytometry
35 lack endogenous viral genes; Demonstrate ALV gs
(ALV) (Exogenous) SPF genetically susceptible for subgroup
(p27) by ELISA
chicks determination

1. Isol. SPF CEFs and assay for virus by ELISA, IFA,


Subtype determination using
Oncornavirus IP, FC, AGID ELISA, VN, IFA,
35 PCR, RT-PCR Mab, Pathogenicity in SPF
(REV) 2. Direct ELISA or CF for gs (p30) in albumin and AGID
chicks
cloacal and vaginal swabs
Subtyping (HI, NI),
pathogenicity (IV in 4-8w old
Orthomyxovirus RT-PCR, ELISA, HI, NI,
29 1. Inoc. CE-AS, HA activity, AGID chicks), PCR for aminoacid
(Al) rRT-PCR AGID
sequence at the Hemagglutinin
cleavage site for H5 and H7

Parvovirus of 1. Isol. Muscovy DE-AS, EM PCR and


42 VN, AGP, ELISA
waterfowl 2. Isol. Muscovy DEF, EM, IF, IP, AC-ELISA, VN sequencing

AGID, VN,
Picomavirus
36 1. Isol. CE-YS, FA ELISA, Embryo
(AE)
susceptibility
1. Isol. SQ or IM 1-7 d old DVH type 1 susceptible
ducks; death with lesions; isol v from liver; No death
in challenged ducks known to have passive or active
Picomavirus immunity to DVH type 1
37 RT-PCR
(DHV I) 2. Isol. DE-AS, lesions; Hyperimmune serum inhibits
pathogenic effects in embryos.
3. Isol. Ckn Emb
4. Isol. DEL, DVH 1 antisera inhibits CPE
1. Isol. IV or IM 1 day old susceptible ducks;
mortality
Picomavirus
37 2. Isol. DE-CAM; CAM lesions; neutralization by
(DHVni)
antisera;
3. FA DEL cells.

1. Isol. CE-YS; Neg. stain EM


Picomavirus-like
38 2. Isol. By inoc of 1-7 day old poult with liver
(TVH)
suspension or yolk from inf. Embryos

Polyomavirus
26 1. Isol. BEF, CEF, Giemsa PCR, ISH VN, ELISA
(BFD)

1. Isol. CEK, CED, QT-35, LMH cells


Immunoblotting, Sequencing; AGED, VN,
Poxvirus 25 2. Isol. CE-CAM, Pocks on CAM, TEM RFLP, PCR
Host pathogenicity, VN ELISA
3. IP, IFA

ELISA, VN and
Reovirus 39 l.Isol. CE-YS, AGID, FA
AGID

Rotavirus 1. Isol. CEK, TEK, CEL, MA 104 Electropherotyping, FA, IEM,


34
(enteric) 2. EM, IEM PGE

1. Isol, by TKY inoc.


Siadenovirus 2. MDTC cells
20 PCR AGID, ELISA
(HEV, MSDV) 3. AGID for Ag, Ag capture ELISA,EHC, ISH
4. FA, IP, ISH, Ac-ELISA (spleen)
Torovirus (enteric) 34 1. Direct EM, FA, IFA ELISA, VN

246
Index

Acholeplasma, 69, 72 Brain heart infusion (Media), 23, 158


Adeno-associated, 101, 104, 230 Brooder pneumonia, 90
Adenovirus, 33, 98, 100, 101, 102, 103, 104, 105, 107, 109, 136, Budgerigar fledgling disease, 143, 145
146, 153,170,171, 172, 204, 212, 223, 228, 241, 246, 248, Buffered peptone water (Media), 282
253, 260, 274, 278, 279, 280, 282 Calicivirus, 182, 216, 217, 248
Aflatoxicosis, 94, 95, 96,209 Campylobacter coli, 27, 30
Agar gel immunodiffusion test (Procedures),88, 108, 139, 162, Campylobacter jejuni, 27, 30
Agar gel precipitin (Procedures), 109, 224, 229, 268 Campylobacteriosis, vii, 27
Agar overlay (Media), 196, 235 Canarypox, 139, 141, 248
Agglutination (Procedures), 9, 11, 15, 20, 21, 20, 21, 24, 27, 29, Candida, 90, 92, 97, 287
33, 39,45, 46, 51, 64, 67, 68, 70, 73, 74, 82, 85, 87, 89, 162, Candidiasis, 92
187, 188, 228, 260, 261, 265, 273, 274, 283, 284 Catalase (Procedures), 19, 21, 20, 24, 26, 39, 43, 44, 45, 46, 50,
Airsacculitis, 12, 15, 19,22, 68, 71, 87, 89, 112 52
Amniotic Sac Inoculation, 244 Cefoperazone, Amphotericin, Teicoplanin agar, 28
Amplified Fragment Length Polymorphism (Procedures), 70, 74, Cell culture (Media), 43, 77, 78, 79, 80, 81, 85, 99, 100, 101,
282 102, 104, 107,109, 111, 113, 114, 117, 120, 126, 128, 131,
Andrade’s, 39 133, 137, 138,142, 143, 147, 149, 158, 167, 171, 172, 175,
Anemia dermatitis syndrome, 146, 148 180, 182, 187,189, 198, 205, 208, 211, 215, 216, 218, 219,
Antibiotic solution (Media), 78 220, 231, 232,234, 235, 239, 246, 250, 257, 260, 264, 273,
Antigen capture (Procedures), 1, 11, 108, 155, 162, 174, 175, 288
220, 274,279 Cell lines (Procedures), 78, 119, 123, 131, 148, 224, 232, 233,
Antigen capture ELISA (Procedures), 114 238, 240, 246
Antigen preparation (Procedures), 102, 263, 274 Cell-associated virus, 239
Antiserum (Procedures), 9, 20, 25, 39, 69, 79,100, 101, 102, 107, Cellulitis, 12, 58, 59
108, 109, 126,132, 138, 147, 153, 154, 155, 159, 162, 167, Chicken anemia virus, 145, 146, 147, 148,248,282
172,183,184, 190, 196, 197, 198, 199, 224, 228, 262, 274 Chicken embryo fibroblast (Procedures), 142
API 20E (Procedures), 12 Chicken embryo kidney (Procedures), i, 112
Arbitrarily primed PCR (Procedures), 282 Chlamydiosis, 13, 21, 33, 71, 76, 77,.79, 85, 86
Arbovirus, 218,219, 220, 221 Chlamydophila psittaci, 76, 79, 80, 81, 82, 83, 85, 86
Arbovirus infection, 219 Chorioallantoic Membrane Inoculation (Procedures), 243
Arizonosis, 11 Chronic respiratory disease, 26
Aspergillosis, i, 9, 44, 90, 92, 93, 96 Circovirus, 101, 144, 145, 148, 248, 253
Aspergillus, i, 64, 90, 91, 92, 94, 95, 96,287 Clostridium, 36, 52, 53, 54, 55, 56, 58, 59, 60, 133, 221, 279
Avian encephalomyelitis, 44, 54, 203, 205,221,241, 246, 260, CMGS (Media), 53, 282, 286
263, 268, 274, 280 Coagulase test (Procedures), 46
Avian influenza, i, 2, 40, 71,128, 133,150, 151, 152, 153, 155, Colibacillosis, 9,12, 13,14,24, 33, 36,40,46,48, 51, 89, 109
156, 162, 168,173, 190, 209, 244, 253, 260, 262, 263, 265, Columbia (Media), vii, 23, 45, 156
268, 274, 275, 278, 279, 280, 283 Complement fixation (Procedures), 44, 52, 54, 76, 79, 82, 86,
Avian leukosis, 64,193, 200, 201,202,238,241,254,274,278, 132, 193, 222, 239, 246, 280
279, 280, 282 Conditioned medium (Media), 237
Avian pneumovirus, 26, 157, 165, 168, 169, 254, 275,279 Cornmeal agar (Media), 92
Avian pox, 115, 138, 139, 140 Coronaviral enteritis, 175,178, 185, 191,280
Avian rhinotracheitis, 165 Coronavirus, ix, 171, 183, 191, 217, 248, 288
Avibacterium, 15, 17, 18, 21, 22, 23,24, 26, 27 Coryza, 1, 17, 18,21, 22,23, 25, 26,27,173,265
Avipoxvirus, 137, 138, 139, 248 Crop mycosis, 92, 97
Bacillus, 44, 52 Cross-challenge tests (Procedures), 170, 173
Bacitracin (Media), 23 Cryptosporidiosis, 21, 71
Balanced salt solution (Media), 111, 138, 208, 232, 233, 235, Dactylaria, 90, 93, 96, 287
255,283 Dactylariosis, 93, 96
Basal medium, Eagle’s (Media), 282 Decontamination (Procedures), 62, 63, 64, 66
Beak and feather disease, 143, 144, 145, 248, 280, 284 Delayed secondary enrichment (Procedures), 4,283
BEM (Media), 282 Dextrose starch agar, 16, 283
BGN (Media), 6, 7, 8, 11, 282, 286 Dilutions (Procedures), 20, 35, 69, 70, 79, 81, 103, 104, 114,119,
BGS (Media), 8,282 128, 132, 143, 148, 152, 167, 172, 181, 188, 199, 204, 207,
Big liver disease, 192 208, 209, 215,220, 221, 224, 239, 255, 256, 257, 259, 261,
Bile esculin (Media), 50 262, 263, 264
Bimavirus, 224, 288 Dimethyl sulfoxide (Media), 118
Bismuth sulfite (Media), 6 Direct fluorescent antibody test (Procedures), 126
Bluecomb, 175,177, 178, 179, 191, 280 Dot blot hybridization (Procedures), 149
Bordetella avium, 24, 21, 265,282 Drag swabs (Procedures), 4, 5, 6, 11
Bordetellosis, 18, 24 Duck hepatitis, 206, 207, 208, 209, 210, 229, 249, 283
Borrelia, 31, 32, 33, 36, 37,286 Duck hepatitis B virus, 206
Botulism, 52, 53, 54, 59, 60, 133 Duck plague, 17, 125, 126, 127,128, 129, 229, 248, 279
Bovamick’s (Media), 78 Duck septicemia, 19
Brachyspira, 33, 35, 36, 37, 286 Duck virus enteritis, 125,283
247
QUICK REFERENCE DIAGNOSTIC CHART: VIRAL DISEASES

Molecular Serogrouping, serotyping,


Pathogen CH Preferred Isolation techniqne/Diagnostic test
Diagnosis speciation, pathogenicity

1. Isol. CKN or TKY TOC, CE-YS, TE-YS, CEF,


Metapneumovirus 31 VERO, QT-35 PCR VN using monoclonals, ID
2. IP, IF, Ag detection

Subgroup by VN or flow
1. Isol. SPF CEFs resistant to subgroup E ALV and
Oncornavirus PCR cytometry, Pathogenicity in
35 lack endogenous viral genes; Demonstrate ALV gs
(ALV) (Exogenous) SPF genetically susceptible
(p27) by ELISA
chicks

1. Isol. SPF CEFs and assay for virus by ELISA, IFA,


Subtype determination using
Oncornavirus IP, FC, AGID
35 PCR, RT-PCR Mab, Pathogenicity in SPF
(REV) 2. Direct ELISA or CF for gs (p30) in albumin and
chicks
cloacal and vaginal swabs
Subtyping (HI, NI),
pathogenicity (IV in 4-8w old
Orthomyxovirus RT-PCR,
29 1. Inoc. CE-AS, HA activity, AGID chicks), PCR for aminoacid
(Al) rRT-PCR
sequence at the Hemagglutinin
cleavage site for H5 and H7

Parvovirus of 1. Isol. Muscovy DE-AS, EM PCR and


42
waterfowl 2. Isol. Muscovy DEF, EM, IF, IP, AC-ELISA, VN sequencing

Picomavirus
36 1. Isol. CE-YS, FA
(AE)

1. Isol. SQ or IM 1-7 d old DVH type 1 susceptible


ducks; death with lesions; isol v from liver; No death
in challenged ducks known to have passive or active
Picomavirus immunity to DVH type 1
37 RT-PCR
(DHV I) 2. Isol. DE-AS, lesions; Hyperimmune serum inhibits
pathogenic effects in embryos.
3. Isol. Ckn Emb
4. Isol. DEL, DVH 1 antisera inhibits CPE
1. Isol. IV or IM 1 day old susceptible ducks;
mortality
Picomavirus
37 2. Isol. DE-CAM; CAM lesions; neutralization by
(DHVni)
antisera;
3. FA DEL cells.

1. Isol. CE-YS; Neg. stain EM


Picomavirus-1 ike
38 2. Isol. By inoc of 1-7 day old poult with liver
(TVH)
suspension or yolk from inf. Embryos

Polyomavirus
26 1. Isol. BEF, CEF, Giemsa PCR, ISH
(BFD)

1. Isol. CEK, CED, QT-35, LMH cells


Immunoblotting, Sequencing;
Poxvirus 25 2. Isol. CE-CAM, Pocks on CAM, TEM RFLP, PCR
Host pathogenicity, VN
3. IP, IFA

Reovirus 39 1. Isol. CE-YS, AGID, FA

Rotavirus 1. Isol. CEK, TEK, CEL, MAI 04 Electropherotyping, FA, IEM,


34
(enteric) 2. EM, IEM PGE

1. Isol, by TKY inoc.


Siadenovirus 2. MDTC cells
20 PCR
(HEV, MSDV) 3. AGID for Ag, Ag capture ELISA,IHC, ISH
4. FA, IP, ISH, Ac-ELISA (spleen)
Torovirus (enteric) 34 1. Direct EM, FA, IFA

246
Index

Dulbecco's modified Eagle's medium (Media), 187, 283 Immunodiffusion (Procedures), 8, 32, 33, 35, 36, 88, 102,106,
Eagle’s minimum essential medium (Media) 283 108, 111, 112, 137, 150, 153, 156, 159, 162, 167, 171, 214,
Earle’s balanced salt (Media), 235 215, 228, 260, 263, 273, 274, 280, 282, 283
EDTA, 79, 118,119, 121,127, 139,208, 233, 234, 237, 249, Immunoelectron microscopy (Procedures), 101,279
269, 276,283 Immunofluorescence (Procedures), 8, 33, 42, 69, 75, 82, 98, 100,
Egg-drop syndrome (Procedures), 136 101,102, 116, 117, 118, 122, 123, 126,128, 132, 142, 146,
Electron microscopy, 98, 109, 165, 175, 183, 190, 254, 273, 283 147,148,156, 167, 182, 195, 197, 198, 199, 208, 218, 224,
Encephalitis, 4, 36,42, 93, 96, 218, 220, 221, 222, 235, 283, 285 227, 239, 278, 279, 280, 283
Encephalomyelitis Immunohistochemical staining (Procedures), 77, 78, 100,109,
AE, 203, 204, 205, 221, 222, 248, 282 142,233, 273,279
Enrichment (Procedures), 4, 5, 6, 7, 11, 28, 30,42, 43, 53, 79, Immunosuppression, 1, 45, 52,94, 95, 106, 117, 148, 192, 223,
235, 274,282,286 225
Enteric viruses, 101, 179, 180, 182, 190, 273 Inclusion body hepatitis, 98, 101, 104,225, 253
Enterobacter, 12, 13 Indirect fluorescent antibody test (Procedures), 36,102, 175, 177,
Enterococcus, 48, 51, 64, 286 199,215
Enterovirus, 177, 182, 191, 205, 279 Indole test (Procedures), 12, 16
Enzyme linked immunosorbent assay (Procedures), 104, 106, Infectious anemia, 1, 146,148,149, 201, 253
148, 175, 176,218 Infectious bronchitis, i, 1, 24, 71, 114, 165, 168, 170, 173, 174,
Epidemic tremor, 203 175, 177, 178, 181, 183, 184, 191, 225, 241, 246, 253, 254,
Erysipelas, 13, 38, 39, 40, 44,48, 51, 216 260, 263, 268, 274, 278, 279, 280, 281
Erysipelothrix selective (Media), 38, 283 Infectious bronchitis virus
Escherichia coli, 1, 8, 12, 13, 14, 20, 24, 29, 46, 58, 60, 64, 71, IBV, 1,168, 183, 274
110, 133, 165, 173, 191,212, 282 Infectious bursal disease
Favus, 93, 287 IBD, 1, 52, 58, 96, 148, 173, 200, 223, 225, 241, 246, 253,
Filtration (Procedures), 28, 30, 73, 78, 118, 131, 151, 233, 234, 254, 260, 268, 274, 278, 279, 280
235, 236, 250,274 Infectious bursal disease virus
Flaviviruses, 220 IBDV, 223, 224, 248, 280, 283
Fluorescein isothiocyanate (Media), 283 Infectious laryngotracheitis
Fluorescent Antibody Test (Procedures), 121, 167 ILT, 1, 114, 115,116,122, 130, 135, 136, 137, 138,139,140,
Food Safety, v, 27 173, 216, 265, 268, 275, 278, 279, 281
Fowl cholera, 15, 16,17, 21, 22, 26,40,46, 54, 71, 87, 89 Infectious laryngotracheitis virus
Fowl pox, 26, 137, 201, 246, 263, 268 ILTV, 114,115, 173,283
Frey's (Media), 73 Influenza (Al), 150
FTA cards(Media), 269 Intracerebral Pathogenicity Index (Procedures), 160
Fusarium, 90, 94, 95 Laboratory safety, 2, 76
Gallibacterium, 15, 17, 18, 22, 58, 87 Lake Victoria Cormorant, 131
Gangrenous dermatitis, 52, 58, 59, 146,225 Leukosis, 33,107, 192, 200,201,202,241,265,280,282,284
Gel diffusion precipitin test, 22,109 Listeria, 40,42,43, 44, 50, 286
Gentamicin(Media), 13,19, 77, 78, 87, 92,125,151, 193, 208, Listeriosis, 40,42,43,44
234,237,243 LMH, 112, 138, 181, 233, 237, 289
Geometric mean titers (Procedures), 255, 258, 262,266 Lowenstein-Jensen (Media), 284
Glycerol (Media), 17 Lymphoid leukosis, 201
Goose hepatitis, 226,249 Lymphoproliferative disease, 108,198
Goose parvovirus, 226, 227,228, 229, 230, 248, 278 Lysine iron,(Media) 8, 284
Gosling plague, 226 M199 (Media), 130,131,232, 235,237
Gumboro, 223 M20 (Media), 233,238
Hajna (Media), 7 MacConkey (Media) 7,11,12,13,16,17,18, 19,24,20,48, 50,
Hajna and Damon (Media), 7 87, 88,284
Hektoen enteric (Media), 6, 8 Malabsorption syndrome, 179, 214, 217
Hemagglutination (Procedures), 16, 21, 20, 21, 22, 24,25, 27, 44, Malignant edema, 58
68, 70, 73, 74, 98,100,101, 103, 126,129,133,137,139, Mannitol-salt (Media), 45
140, 144, 145, 150, 153, 159, 164, 166, 170, 171, 174, 179, Marble spleen disease, 98,106,109, 248
183, 184, 187, 191, 218, 220, 221, 241, 246, 251, 258, 260, Marek’s disease, 33, 44, 54, 64, 107, 117, 118, 122, 123, 124,
261,262,268,274,280,283 130, 131, 135, 136, 146, 148, 192, 197, 200, 201, 202, 205,
Hemagglutination Inhibition (Procedures), 172 216,224,238, 239, 240, 246,248, 253, 254, 263,268, 275,
Hemorrhagic enteritis, 1, 54,98,108, 109, 110, 179, 219,248, 278, 284
260,265, 279 McClung-Toabe egg yolk agar, 53, 56, 59
Hemorrhagic syndrome, 146 Micrococcus, 46
Herpesvirus, 101, 111, 113, 117, 118, 119,120, 122, 123, 124, Microsporum, 90, 93
125, 126, 131,135, 136, 139, 192, 229, 240, 246, 248, 280 Middlebrook 7H9 (Media), 62, 63
Herpesvirus of turkeys, 123, 135, 240 Minimum essential medium (Media), 131, 182,184, 208, 215,
High pressure liquid chromatography (Procedures) 96 237
Highlands J virus, 221,222, 248 Molecular probes, 64,286
Hybridization with nucleic acid probes (Procedures), 269 Monoclonal antibodies (Media), 42, 43, 64, 79, 85, 114,117,
Hydropericardium syndrome, 98, 104, 278 118, 120, 122, 132, 135, 140,147, 148, 159, 164, 165, 168,
Immunocytochemistry (Procedures), 280 170, 172, 174, 176, 185, 192,194, 197, 198, 200, 201, 202,
225, 227, 229, 246, 278, 280
248
Index

Muscovy duck parvovirus, 226, 229, 230, 248, 284 Quail pox, 137
Mycobacterium, 61, 62, 63, 64, 66, 67, 133, 284, 286 Quality control, 260, 262, 266
Mycoplasma, i, 1, 22, 46, 68, 69, 70, 71, 72, 73, 74, 75,104, 168, Rambach (Media), 8,286
170, 173, 216, 234, 235, 241, 260, 261, 263, 265, 266, 268, Reovirus, 1, 104, 172, 177, 178, 191, 212, 214, 215, 216, 217,
269, 274, 275, 280, 284, 286 223, 240, 241, 246, 248, 265, 274, 275, 284
Mycoplasmosis, 1, 13, 18, 21, 89, 114, 173, 260, 263, 265 Reticuloendotheliosis, 108, 122, 123, 139, 140, 192, 200, 201,
Mynah pox, 140 202,216, 238, 274, 278, 282
Necrotic enteritis, 52, 54, 56, 57, 58, 59, 60, 279 Retrovirus, 197
Neoplasms, 64 Rhinotracheitis, 2,21, 136, 157, 165, 168, 169, 221, 248, 282
Neuraminidase, 39, 150, 152, 154, 156, 159, 161, 166, 168, 172, Ringworm, 93
260, 283 Rotavirus, 136, 148, 177,179, 187, 189, 191, 248, 251
New duck disease, 19 Sabouraud (Media), i, 91, 93
Newcastle disease, 1, 3, 9, 24, 21, 40,44, 54, 71, 122, 128, 133, Salmonella, 8, 9, 11
153, 155, 156, 157, 158, 160, 161, 164, 167, 170, 173, 174, Salmonella pullorum, 260
190, 205, 209, 221, 241, 244, 246, 248, 253, 254, 260, 268, Salmonellae, 6, 7, 8, 9, 11
275, 279, 280, 284 Salmonellosis, 4, 9, 13, 21, 36, 40, 44, 209, 265
Newcastle disease virus, 156, 167, 190 Sarcoma, 192, 196, 198
Nichrome loop (Media), 151 Selenite (Media), 7
Non-producer Cell Activation (Procedures), 196 Sentinel chicken, 220
Notification, 19, 157 Siadenovirus, 98, 106, 108,246,248, 289
Ochratoxin, 90, 95 Silica gel (Media), 48, 151
Oncornavirus, 198, 199, 200, 202 Sodium dodecyl sulfate (Media), 35, 74, 127,139, 184, 189
Ornithosis, 76, 85 SPG buffer (Media), 77
Orthomyxovirus, 246 Spirochetosis, 31, 32, 33, 36, 37, 282
Orthomyxovirus, 289 Splenomegaly, 33,48,206
Osteopetrosis, 192 Staphylococcus, 45, 48, 216
Osteoporosis, 214, 217 Streptococcosis, 40, 44, 48, 51
Pacheco’s disease, 135, 136,248 Stunting syndrome, 179, 190, 191, 214, 217
Packer’s (Media), 38, 39 Stunting syndrome agent, 284
Papovavirus, 143 Supplemental test medium without NAD (Media), 285
Paramyxovirus, 157, 158, 159, 160, 162, 164, 166, 205, 246, 253, Swollen head syndrome, 12,26,157, 165, 169
282 Tellurite glycine (Media), 45
Paratyphoid, 4, 5, 8, 9 Tergitol (Media), 6, 7, 11, 285
Parrot fever, 76 Tetrathionate (Media), 6, 11
Parvovirus, 216, 226, 229, 248, 289 TM/SN, 23, 24, 285
Pasteurella multocida, 15, 16, 21, 22,44, 128, 212, 229, 265, 284 Togaviruses, 133
Penicillium, 90, 94, 95 Torovirus, 179, 190
peptone (Media), 6, 23, 28, 151, 215, 282 Transformation (Focus) Assay, 196
Phenotypic Mixing (Procedures), 196 Tri-buffered saline (Media), 284
Phosphate-buffered saline (Media), 79, 284 Trichophyton, 93
phosphotungstic acid (Media), 183, 189, 246, 251, 273 Triple Sugar Iron (Media), 8
picomavirus, 203, 206, 207, 211 Tris borate Buffer (Media), 284
Picomavirus-like, 289 Tuberculosis, 21,61, 62, 63, 64, 66, 67, 287
Pigeon pox, 135, 137 Tumors, 117, 119, 120, 122, 124, 130, 192, 193, 196, 197,198,
Plague, 17, 126, 127, 129, 150 200, 202,238
Pneumovirus, 1, 21, 169 Turkey herpesvirus, 248
Polyomavirus, 142, 143, 145, 229 Turkey meningoencephalitis, 218
Potato dextrose (Media), 91 Turkey pox, 137
Poult enteritis, 175,177, 178, 179, 191 Turkey viral enteritis, 179
Poult enteritis and mortality syndrome Turkey viral hepatitis, 211, 212, 285
PEMS, 179 Tween 80 Hydrolysis (Media), 63
Pox, 115, 132,137,138, 139 Typhoid, 4,9, 11, 17,46, 265
Poxvirus, 128, 137, 138, 139, 140, 246 Ulcerative enteritis, 52, 54, 58, 59
Proteus, 7, 8, 24 Ureaplasma, 72, 74
Providencia, 7, 8 Vibrionic hepatitis, 27, 30
Pseudomonas, 7, 133 Viral arthritis, 46,214,216, 217,248,260,263
Pseudotuberculosis, 13, 15, 17, 18, 21, 22 Viral enteritis, 129, 134, 229, 275
Psittacine beak and feather disease Virus transport medium (Media), 146, 165
PBFD, 143 Western duck sickness, 53
Psittacosis, 76, 85, 86 XLD supplemented with novobiocin, 285
Pullorum disease (Procedures), 2, 4 XLT4, 6, 7, 8, 11,285,286
Quail bronchitis, 98 Xylose lysine desoxycholate (XLD) (Media), 6
Quail disease, 52, 54, 59 Yersinia, 17, 21
buffer at pH9.0

249
THE AMERICAN ASSOCIATION OF AVIAN PATHOLOGISTS

Fifth Edition 2008

ISBN 978-0-9789163-2-·

You might also like