You are on page 1of 647

Essentials of

Microbiology

www.ebook3000.com
Essentials of
Microbiology

Surinder Kumar
MD DNB MNAMS
Director Professor
Department of Microbiology
Maulana Azad Medical College
New Delhi, India

The Health Sciences Publisher


New Delhi | London | Philadelphia | Panama

www.ebook3000.com
Jaypee Brothers Medical Publishers (P) Ltd

Headquarters
Jaypee Brothers Medical Publishers (P) Ltd
4838/24, Ansari Road, Daryaganj
New Delhi 110 002, India
Phone: +91-11-43574357
Fax: +91-11-43574314
Email: jaypee@jaypeebrothers.com

Overseas Offices
J.P. Medical Ltd Jaypee-Highlights Medical Publishers Inc Jaypee Medical Inc
83, Victoria Street, London City of Knowledge, Bld. 237, Clayton The Bourse
SW1H 0HW (UK) Panama City, Panama 111 South Independence Mall East
Phone: +44-2031708910 Phone: +1 507-301-0496 Suite 835, Philadelphia, PA 19106, USA
Fax: +44(0)20 3008 6180 Fax: +1 507-301-0499 Phone: +1 267-519-9789
Email: info@jpmedpub.com Email: cservice@jphmedical.com Email: jpmed.us@gmail.com

Jaypee Brothers Medical Publishers (P) Ltd Jaypee Brothers Medical Publishers (P) Ltd
17/1-B Babar Road, Block-B, Shaymali Bhotahity, Kathmandu
Mohammadpur, Dhaka-1207 Nepal
Bangladesh Phone: +977-9741283608
Mobile: +08801912003485 Email: kathmandu@jaypeebrothers.com
Email: jaypeedhaka@gmail.com

Website: www.jaypeebrothers.com
Website: www.jaypeedigital.com

© 2016, Jaypee Brothers Medical Publishers

The views and opinions expressed in this book are solely those of the original contributor(s)/author(s) and do not necessarily represent those
of editor(s) of the book.

All rights reserved. No part of this publication may be reproduced, stored or transmitted in any form or by any means, electronic, mechanical,
photocopying, recording or otherwise, without the prior permission in writing of the publishers.

All brand names and product names used in this book are trade names, service marks, trademarks or registered trademarks of their respective
owners. The publisher is not associated with any product or vendor mentioned in this book.

Medical knowledge and practice change constantly. This book is designed to provide accurate, authoritative information about the subject
matter in question. However, readers are advised to check the most current information available on procedures included and check
information from the manufacturer of each product to be administered, to verify the recommended dose, formula, method and duration of
administration, adverse effects and contraindications. It is the responsibility of the practitioner to take all appropriate safety precautions.
Neither the publisher nor the author(s)/editor(s) assume any liability for any injury and/or damage to persons or property arising from or
related to use of material in this book.

This book is sold on the understanding that the publisher is not engaged in providing professional medical services. If such advice or services
are required, the services of a competent medical professional should be sought.

Every effort has been made where necessary to contact holders of copyright to obtain permission to reproduce copyright material. If any have
been inadvertently overlooked, the publisher will be pleased to make the necessary arrangements at the first opportunity.

Inquiries for bulk sales may be solicited at: jaypee@jaypeebrothers.com

Essentials of Microbiology
First Edition: 2016
ISBN 978-93-5152-380-2
Printed at
Dedicated to
My father
Late Shri Lachhman Das
My mother
Late Smt Bal Kaur
My wife
Dr (Prof) Savita Kumari
and my sons
Dr Sourabh Kumar and Sanchit Kumar
whose love and affection make everything I do possible.

www.ebook3000.com
Preface

Microbiology is an extremely diverse discipline and can be a bewildering field to the novice. The
traditional books, seem to be exhaustive of microbiologic facts, are too voluminous and detailed to be
read by a typical medical student, who is trying to keep up with simultaneously several classes of other
subjects. One of the ways to fulfill the goals of the book is to concentrate on understanding concepts—
important fundamental ideas or themes that form a framework of the subject. Essentials of Microbiology
is the result of the author’s 28 years of experience in teaching medical students. The book is readable and
complete enough to meet the students’ needs. The overall objective of this book is to provide students
the basics of medical microbiology as well as a complete coverage of the subject. It contains all of the
information that is pertinent to the medical students who are studying microbiology keeping in mind
their examination.
Although the text has been designed to teach undergraduate and postgraguate medical students, it
would also serve as a review tool for the individuals who are taking medical examinations and persons
working in health-related professions, physicians, and infectious disease scientists.
Microbiology has expanded beyond recognition with various medical specialty, and it is not possible
for any one book to cover all aspects of medical microbiology in depth. This book is divided into
the following seven sections, based on the major disciplines included within microbiology: General
Bacteriology, Immunology, Systemic Bacteriology, Virology, Medical Mycology, Miscellaneous, and
Diagnostic Medical Microbiology. The chapters themselves are comprehensive yet free of unnecessary
detail and provide the reader with a framework for understanding. Mycology and parasitology have
continued to flourish and have blossomed into fields of study of their own rights. Therefore, parasitology
has not been included in this book which has a sturdy independence.
I would be thankful for any comments or suggestions from students, teachers, and all the readers of
this book for further improvements in its future editions.

Surinder Kumar

www.ebook3000.com
Acknowledgments

This book took years in writing but a lifetime in preparation. I would like to thank those whose example,
teaching, and prodding helped me develop a thirst for knowledge as well as methods for quenching
that thirst. I am greatly indebted to a number of my mentors, friends and colleagues who encouraged
me and gave valuable suggestions for improving the text. I am always indebted to my late brother Sant
Swaran Dev whose advice, guidance and true life philosophy gave me strength and courage to continue
my work in all odds. I would like to thank my family for patiently enduring the writing of this book,
which seemed at times to be an endless process. I am especially grateful to my wife Dr Savita Kumari,
Professor, Department of Internal Medicine, Post Graduate Institute and Medical Education and Research
(PGIMER), Chandigarh, for her support and encouragement and to my two sons, Dr Sourabh Kumar
and Mr Sanchit Kumar (medical student) who gave me their valuable moments without any complaint
for completing this book, so I could retain my sanity.
My special thanks is to Dr Sanjeev R Saigal, my PhD student and now my Research Associate Indian
Council of Medical Research (ICMR) for his constant help for this book. Sincere appreciation also goes
to all staff of M/s Jaypee Brothers Medical Publishers (P) Ltd., New Delhi, for their guidance and support
in this project.

www.ebook3000.com
Contents

Section 1: General Bacteriology


1. Historical Development of Microbiology 1
∙∙ Infection and Contagion  1
∙∙ Discovery of Microorganisms  1
∙∙ Conflict over Spontaneous Generation  1
∙∙ Role of Microorganisms in Diseases  2
∙∙ Scientific Development of Microbiology  2
∙∙ Louis Pasteur (1822–1895)  2
∙∙ Joseph Lister (1827–1912)  3
∙∙ Robert Koch (1843–1910)  3
∙∙ Paul Ehrlich (1854–1915)  3
∙∙ Discovery of Viruses   4
∙∙ Immunity and Immunization  4
∙∙ Serotherapy and Chemotherapy  5
∙∙ Nobel Prizes Awarded for Research in Microbiology  5
2. Microscopy 8
∙∙ Microscopy: Instruments  8
∙∙ Light Microscopy  8
∙∙ Electron Microscopy  9
∙∙ Scanning-probe Microscopy   10
  3. Morphology of Bacteria 11
∙∙ Comparison between Prokaryotic and Eucaryotic Cells   11
∙∙ Size of Bacteria  11
∙∙ Study of Bacteria  12
∙∙ Arrangement of Bacterial Cells  12
∙∙ Anatomy of the Bacterial Cell   13
∙∙ Pleomorphism and Involution Form  19
∙∙ L-Forms of Bacteria (Cell-wall-defective Organisms)  20
4. Physiology of Bacteria 21
∙∙ Principles of Bacterial Growth  21
∙∙ Bacterial Nutrition  22
∙∙ Bacterial Metabolism  23
5. Sterilization and Disinfection 25
∙∙ Definitions of Frequently Used Terms  25
∙∙ Methods of Sterilization and Disinfection  25
∙∙ Recommended Concentrations of Various Disinfectants  34
∙∙ Testing of Disinfectants  34
6. Culture Media 37
∙∙ Common Ingredients of Culture Media  37
∙∙ Classification of Media  37
7. Culture Methods 43
∙∙ Methods of Bacterial Culture   43
∙∙ Aerobic Culture  44
∙∙ Anaerobic Culture  44
∙∙ Methods of Anaerobiosis  44
∙∙ Methods of Isolating Pure Cultures  46
8. Identification of Bacteria 48
∙∙ Methods Used to Identify Bacteria  48
∙∙ Phenotypic Characteristics  48
∙∙ Genomic Characterization  55
9. Bacterial Taxonomy 56
∙∙ Taxonomy 56
∙∙ Identification 56

www.ebook3000.com
xii  |  Essentials of Microbiology
∙∙ Classification 56
∙∙ Classification Systems   56
10. Bacterial Genetics 58
∙∙ Structure and Functions of the Genetic Material  58
∙∙ Extrachromosomal Genetic Elements  60
∙∙ Genotypic and Phenotypic Variations  61
∙∙ Transmission of Genetic Material (Gene Transfer)  63
∙∙ Genetic Mechanisms of Drug Resistance in Bacteria   68
∙∙ Transposable Genetic Elements   68
∙∙ Molecular Genetics  68
∙∙ Genetic Probes   70
∙∙ Blotting Techniques  71
∙∙ Polymerase Chain Reaction (PCR)  71
∙∙ Gene Therapy  72
11. Infection 75
∙∙ Microorganisms and Host  75
∙∙ Infection and Infectious Disease  75
∙∙ Classification of Infections  75
∙∙ Sources of Infection  76
∙∙ Modes of Transmission of Infection  77
∙∙ Factors Predisposing to Microbial Pathogenicity  78
∙∙ Determinants of Virulence  78
∙∙ Types of Infectious Diseases  80
∙∙ Epidemiological Terminologies  80

Section 2: Immunology
12. Immunity 81
∙∙ Definition 81
∙∙ Classification 81
∙∙ Mechanisms of Innate Immunity  82
∙∙ Local Immunity  86
∙∙ Herd Immunity  86
13. Antigens 87
∙∙ Types of Antigens  87
∙∙ Antigenic Determinant or Epitome  87
∙∙ Determinants of Antigenicity  87
∙∙ Superantigens   89
14. Antibodies—Immunoglobulins 90
∙∙ Antibody Structure  90
∙∙ Immunoglobulin Classes  92
15. Complement System 96
∙∙ Principle Pathways of Complement Activation  96
∙∙ Quantitation of Complement (C) and its Components  99
∙∙ Biosynthesis of Complement  99
∙∙ Complement Deficiencies   99
16. Antigen–Antibody Reactions 101
∙∙ Antigen–Antibody Interactions  101
∙∙ General Characteristics of Antigen–Antibody Reactions  102
∙∙ Antigen and Antibody Measurement  102
∙∙ Parameters of Serological Tests  102
∙∙ Serological Reactions  102
∙∙ Types of Antigen and Antibody Reactions  102
∙∙ Applications of Agglutination Reaction  106
∙∙ ELISA 112
17. Structures and Functions of the Immune System 117
∙∙ Types of Immune Response  117
∙∙ Organs and Tissues of the Immune System  117
∙∙ Cells of the Lymphoreticular System  119
∙∙ Major Histocompatibility Complex  122
∙∙ Mhc Restriction  123
Contents  | xiii
18. Immune Response 125
∙∙ Type of Immune Response  125
∙∙ Humoral Immunity  125
∙∙ Fate of Antigen in Tissues  126
∙∙ Production of Antibodies  127
∙∙ Cell-mediated Immune Responses  130
∙∙ Cytokines 131
∙∙ Immunological Tolerance  134
∙∙ Theories of Antibody formation  135
19. Immunodeficiency Diseases 137
∙∙ Classification of Immunodeficiency Diseases  137
∙∙ Primary Immunodeficiencies  137
∙∙ Disorders of Specific Immunity  137
∙∙ Disorders of Complement  140
∙∙ Disorders of Phagocyte  140
∙∙ Secondary Immunodeficiencies  140
20. Hypersensitivity Reactions 142
∙∙ Classification of Hypersensitivity Reactions  142
∙∙ Type I Hypersensitivity (IgE Dependent)  143
∙∙ Type II Hypersensitivity: Cytolytic and Cytotoxic  146
∙∙ Type III Hypersensitivity: Immune Complex-mediated   147
∙∙ Type IV Hypersensitivity—Delayed Hypersensitivity  148
∙∙ Type V: Hypersensitivity (Stimulatory Type) Jones–Mote Reaction or
Cutaneous Basophil Hypersensitivity  149
∙∙ Schwartzman Reaction  149
21. Autoimmunity 151
∙∙ Mechanisms of Autoimmunity  151
∙∙ Classification of Autoimmune Diseases  152
22. Immunology of Transplantation and Malignancy 154
∙∙ Definition 154
∙∙ Types of Transplants  154
∙∙ Allograft Reaction   154
∙∙ Histocompatibility Testing  155
∙∙ Fetus as an Allograft  156
∙∙ Graft-versus-host Reaction   156
∙∙ Immunology of Malignancy  156
∙∙ Tumor Antigens  156
∙∙ Immune Response in Malignancy  157
∙∙ Immunological Surveillance  157
∙∙ Immunotherapy of Cancer  157
23. Immunohematology 159
∙∙ Other Blood Group Systems  160
∙∙ Medical Applications of Blood Groups  160
∙∙ Complications of Blood Transfusion  160
∙∙ Hemolytic Disease of the Newborn  160

Section 3: Systemic Bacteriology


24. Staphylococcus 163
∙∙ Staphylococcus aureus  163
∙∙ Other Coagulate-positive Staphylococci   168
∙∙ Coagulase-negative Staphylococci  169
∙∙ Micrococci   170
25. Streptococcus and Enterococcus 172
∙∙ Streptococcus pyogenes  173
∙∙ Laboratory Diagnosis  178
∙∙ Other Streptococci Pathogenic for Humans  179
∙∙ Enterococcus 180
∙∙ Viridans Streptococci   180
26. Pneumococcus (Diplococcus pneumoniae: Streptococcus pneumoniae) 183
∙∙ Pneumococcus (Diplococcus pneumoniae, Streptococcus pneumoniae) 183

www.ebook3000.com
xiv  |  Essentials of Microbiology
27. Neisseria and Moraxella 188
∙∙ Neisseria meningitidis (Meningococcus; Diplococcus intracellularis meningitidis)  188
∙∙ Neisseria gonorrhoeae (Gonococcus)  191
∙∙ Nongonococcal (Nonspecific) Urethritis  193
∙∙ Commensal Neisseriae  194
∙∙ Moraxella   194
∙∙ Kingella 195
28. Corynebacterium 197
∙∙ Corynebacterium diphtheriae  197
∙∙ Other Medically Important Corynebacteria  202
∙∙ Diphtheroids 203
∙∙ Other Coryneform Genera  203
29. Bacillus 205
∙∙ General Characterstics of Bacillus  205
∙∙ Bacillus anthracis  205
∙∙ Anthracoid Bacilli  208
30. Clostridium 211
∙∙ General Features of Clostridia  211
∙∙ Classification 212
∙∙ Clostridium tetani   215
∙∙ Clostridium botulinum  218
∙∙ Clostridium difficile  219
31. Nonsporing Anaerobes 221
∙∙ Anaerobic Cocci  221
∙∙ Gram-negative Anaerobic Cocci  222
∙∙ Anaerobic, Nonspore-forming, Gram-positive Bacilli  222
∙∙ Anaerobic Gram-negative Bacilli  223
∙∙ Anaerobic Infections  224
32. Mycobacterium tuberculosis 227
∙∙ M. tuberculosis Complex or MTC  227
∙∙ Mycobacterium tuberculosis  228
33. Mycobacterium leprae 240
∙∙ Mycobacterium leprae   240
∙∙ Mycobacterium lepraemurium  245
34. Nontuberculous Mycobacteria 247
∙∙ Properties of Nontuberculous Mycobacteria  247
∙∙ Classification 248
∙∙ Saprophytic Mycobacteria  249
35. Actinomycetes 252
∙∙ Actinomyces 252
∙∙ Nocardia 253
∙∙ Actinomycotic Mycetoma  254
36. Enterobacteriaceae: Escherichia, Klebsiella, Proteus and Other Genera 256
∙∙ Characteristics of the Family Enterobacteriaceae  256
∙∙ Classification of Enterobacteriaceae  256
∙∙ Classification of Enterobacteriaceae by Tribes  257
∙∙ Escherichia coli  257
∙∙ Infections 263
∙∙ Edwardsiella 263
∙∙ Citrobacter 263
∙∙ Klebsiella 264
∙∙ Enterobacter 265
∙∙ Hafnia 265
∙∙ Serratia 265
37. Tribe Proteeae: Proteus, Morganella and Providencia 267
∙∙ Proteus   267
∙∙ Morganella 269
∙∙ Providencia   269
∙∙ Erwinia 269
38. Shigella 270
Contents  | xv
39. Enterobacteriaceae III: Salmonella 274
∙∙ Salmonella 274
∙∙ Diagnosis of Carriers  283
∙∙ Prophylaxis 283
∙∙ Drug Resistance  284
∙∙ Salmonella Gastroenteritis  284
∙∙ Salmonella Septicemia  285
∙∙ Multiresistant Salmonellae  285
40. Vibrio, Aeromonas and Plesiomonas 287
∙∙ Vibrio 287
∙∙ Vibrio cholerae  287
∙∙ Resistance 288
∙∙ Halophilic Vibrios  294
∙∙ Aeromonas 295
∙∙ Plesiomonas 295
41. Campylobacter and Helicobacter 297
∙∙ Campylobacter 297
∙∙ Helicobacter 298
42. Pseudomonas, Stenotrophomonas, Burkholderia 301
∙∙ Pseudomonas aeruginosa  301
∙∙ Burkholderia cepacia (Formerly Pseudomonas cepacia)  304
∙∙ Burkholderia mallei (Formely Pseudomonas mallei)  304
∙∙ Burkholderia pseudomallei   305
43. Legionella 307
∙∙ Legionella pneumophila  307
44. Yersinia, Pasteurella, Francisella 309
∙∙ Yersinia pestis (Formerly Pasteurella pestis)  309
∙∙ Yersiniosis 313
∙∙ Pasteurella multocida (Formerly Pasteurella septica)  314
∙∙ Francisella tularensis (Pasteurella tularensis, Brucella tularensis)  315
45. Haemophilus 317
∙∙ Haemophilus Influenzae  317
∙∙ Haemophili Other Than H. influenzae  320
46. Bordetella 323
∙∙ Bordetella pertussis  323
∙∙ Bordetella parapertussis  326
∙∙ Bordetella bronchiseptica (Bord. bronchicanis)  326
47. Brucella 327
48. Spirochetes 333
∙∙ Classification 327
∙∙ Treponema 334
∙∙ Nonvenereal Treponematoses  341
∙∙ Nonpathogenic Treponemes  341
∙∙ Borrelia 342
∙∙ Leptospira 344
49. Mycoplasma and Ureaplasma 348
∙∙ Classification 348
∙∙ Morphology 349
∙∙ Cultural Characteristics  349
∙∙ Pathogenicity 350
∙∙ Mycoplasma pneumoniae  350
∙∙ Laboratory Diagnosis  351
∙∙ Mycoplasmas and L Forms of Bacteria  352
∙∙ Ureaplasma urealyticum  353
50. Miscellaneous Bacteria 355
∙∙ Listeria monocytogenes  355
∙∙ Erysipelothrix rhusiopathiae  356
∙∙ Alcaligenes faecalis   357
∙∙ Chromobacterium violaceum  357

www.ebook3000.com
xvi  |  Essentials of Microbiology
∙∙ Flavobacterium meningosepticum  357
∙∙ Donovania granulomatis (Calym­matobacterium granulomatis) or Klebsiella granulomatis  357
∙∙ Acinetobacter (Mima polymorpha; Bacterium anitratum  358
∙∙ Rat-bite fever (Streptobacillus moniliformis and Spirillum minus)  358
∙∙ Spirillum minus  359
∙∙ Eikenella corrodens  359
∙∙ Cardiobacterium hominis  360
∙∙ Capnocytophaga 360
∙∙ Gardnerella vaginalis  360
51. Rickett­siaceae, Bartonellaceae and Coxiella 362
∙∙ Genus Rickettsia  362
∙∙ Ehrlichia, Anaplasma and Neorickettsia   366
∙∙ Genus Coxiella: Q Fever  367
∙∙ Bartonella 368
52. Chlamydia and Chlamydophila 371
∙∙ Classification 371

Section 4: Virology
53. General Properties of Viruses 378
∙∙ Main Properties of Viruses   378
∙∙ Morphology of Viruses  378
∙∙ Structure and Chemical Composition of the Viruses  379
∙∙ Susceptibility to Physical and Chemical Agents  380
∙∙ Viral Hemagglutination  381
∙∙ Viral Replication  381
∙∙ Eclipse Phase  383
∙∙ Abnormal Replicative Cycles  383
∙∙ Cultivation of Viruses  383
∙∙ Detection of Virus Growth in Cell Culture  385
∙∙ Viral Assay  386
∙∙ Viral Genetics  387
∙∙ Classification of Viruses  388
∙∙ Viroids 390
∙∙ Prions 390
54. Virus–Host Interactions: Viral Infections 392
∙∙ Interactions between Viruses and Host Cells  392
∙∙ Pathogenesis of Viral Diseases  393
∙∙ Transmission of Human Virus Infections  393
∙∙ Spread of Virus in the Body  394
∙∙ Significance of the Incubation Period  394
∙∙ Host Response to Virus Infections  395
55. Laboratory Diagnosis, Prophylaxis and Chemotherapy of Viral Diseases 397
∙∙ Laboratory Diagnosis of Viral Infections  397
∙∙ Immunoprophylaxis of Viral Diseases   399
∙∙ Chemoprophylaxis and Chemotherapy of Virus Diseases  400
56. Bacteriophages 402
∙∙ Role of Bacteriophages  402
∙∙ Morphology 402
∙∙ Life Cycle  402
∙∙ Significance of Phages  404
57. Poxviruses 406
∙∙ Morphology 406
∙∙ Physical and Chemical Properties   407
∙∙ Other Poxvirus Diseases  408
58. Herpesviruses 409
∙∙ Structure 409
∙∙ Classification 409
∙∙ Herpes Virus Simiae: B Virus  411
∙∙ Varicella-Zoster Virus  411
Contents  | xvii
∙∙ Herpes Zoster (Shingles, Zona)  412
∙∙ Cytomegalovirus   413
∙∙ Epstein–Barr Virus  413
∙∙ Human Herpesviruses 6 (Hhv6) 415
∙∙ Human Herpesvirus 7 (Hhv7) 415
∙∙ Human Herpesvirus 8 (Hhv8) 416
59. Adenoviruses 418
∙∙ Morphology 418
∙∙ Resistance   418
∙∙ Pathogenesis 418
∙∙ Laboratory Diagnosis  419
∙∙ Treatment, Prevention and Control  420
60. Papovaviruses 421
∙∙ Papillomaviruses 421
∙∙ Polyomaviruses 422
61. Parvoviruses 424
∙∙ Parvovirus 424
∙∙ Dependovirus   424
∙∙ Erythrovirus 424
62. Picornaviruses 426
∙∙ Important Properties of Picornaviruses  426
∙∙ Enteroviruses 426
∙∙ Poliovirus 427
∙∙ Coxsackievirus 429
∙∙ Echoviruses 430
∙∙ Other Enterovirus Types  431
∙∙ Rhinoviruses 432
63. Orthomyxovirus 434
∙∙ Influenza Viruses  434
64. Paramyxoviruses 440
∙∙ Morphology and Structural Proteins of Paramyxoviruses  440
∙∙ Classification 440
∙∙ Parainfluenza Viruses   441
∙∙ Genus Rubulavirus  442
∙∙ Genus Morbillivirus  443
∙∙ Nipah and Hendra Viruses  444
∙∙ Genus Pneumovirus  444
∙∙ Metapnemovirus 445
65. Arboviruses 446
∙∙ Classification 446
∙∙ Properties   446
∙∙ Laboratory Diagnosis  447
∙∙ Pathogenesis   447
∙∙ Families of Arboviruses  447
∙∙ Ungrouped Arboviruses  455
∙∙ Arbovirus Known to Be Prevalent in India  455
66. Rhabdoviruses 457
∙∙ Rabies Virus  457
∙∙ Rabies-related Viruses  463
67. Hepatitis Viruses 465
∙∙ Hepatitis A Virus (Infectious Hepatitis)  465
∙∙ Hepatitis B Virus—Serum Hepatitis  467
∙∙ Hepatitis C Virus (HCV)  471
∙∙ Hepatitis D Virus (HDV) (Delta Agent)  472
∙∙ Hepatitis E Virus (HEV)  473
∙∙ Hepatitis G Virus  474
68. Retroviruses: Human Immunodeficiency Virus (HIV) 476
∙∙ Retroviruses 476
∙∙ Human Immunodeficiency Virus  476

www.ebook3000.com
xviii  |  Essentials of Microbiology
69. Slow Virus and Prion Diseases 488
∙∙ Characteristics of Slow Viruses  488
∙∙ Classification 488
70. Miscellaneous Viruses 492
∙∙ Rubivirus 492
∙∙ Rubella (German Measles)  492
∙∙ Viral Hemorrhagic Fevers  493
∙∙ Arenaviruses 493
∙∙ Filoviruses   494
∙∙ Coronaviruses 494
∙∙ Reoviridae 496
∙∙ Norwalk Virus  497
71. Oncogenic Viruses 499
∙∙ Properties of Cells Transformed by Viruses  499
∙∙ Types of Tumor Viruses  499
∙∙ Oncogenic Viruses  499
∙∙ Viruses Associated with Human Cancer  501
∙∙ Oncogenes 501
∙∙ Antioncogenes 502
∙∙ Mechanisms of Viral Oncogenesis  502

Section 5: Medical Mycology


72. General Properties, Classification and Laboratory Diagnosis of Fungi 503
∙∙ Differences of Fungi from Bacteria  503
∙∙ General Properties of Fungi  503
∙∙ Classification of Fungi  503
∙∙ Reproduction and Sporulation  504
∙∙ Laboratory Diagnosis  505
∙∙ Classification of Mycoses  506
73. Superficial, Cutaneous and Subcutaneous Mycoses 508
∙∙ Superficial Mycoses  508
∙∙ Cutaneous Mycoses  509
∙∙ Subcutaneous Mycoses  511
74. Systemic Mycoses 515
∙∙ Blastomycosis 515
∙∙ Paracoccidioidomycosis 515
∙∙ Coccidioidomycosis 516
∙∙ Histoplasmosis 516
75. Opportunistic Mycoses 519
∙∙ Opportunistic Fungi  519
∙∙ Yeast-like Fungi  519
∙∙ Aspergillosis 522
∙∙ Other Opportunist Fungi  525
76. Mycotoxicosis 528
∙∙ Mycetism 528
∙∙ Mycotoxicosis 528
∙∙ Psychotropic Agents  529

Section 6: Miscellaneous
77. Normal Microbial Flora of the Human Body 530
∙∙ Role of Normal Microbial Flora  530
78. Infective Syndrome 533
∙∙ Bacteremia, Septicemia and Infective Endocarditis  533
∙∙ Bacteremia and Septicemia  533
∙∙ Meningitis 535
∙∙ Purulent Meningitis (Acute Pyogenic Meningitis)  535
∙∙ Aseptic Meningitis  537
∙∙ Tuberculous Meningitis  538
Contents  | xix
∙∙ Urinary Tract Infection  538
∙∙ Types of UTI  538
∙∙ Sore Throat  541
∙∙ Pneumonia 542
∙∙ Diarrhea and Dysentery  544
∙∙ Diarrhea 544
∙∙ Dysentery 546
∙∙ Food Poisoning  547
∙∙ Sexually Transmitted Diseases (STDs)  547
∙∙ Wound Infection  550
∙∙ Pyrexia of Unknown Origin (PUO)  551
79. Hospital-acquired Infection 555
∙∙ Sources of Infections  555
∙∙ Factors Influencing Hospital-associated Infections  555
∙∙ Microorganisms Causing Hospital Infection  555
∙∙ Routes of Transmission  556
∙∙ Common Hospital-acquired Infection  556
∙∙ Diagnosis and Control of Hospital Infection  557
∙∙ Infection Control Policy  557
∙∙ Prevention 557
80. Laboratory Control of Antimicrobial Therapy 559
∙∙ Antibiotic Sensitivity Tests  559
∙∙ Kirby–Bauer Disk Diffusion Method  560
∙∙ Stokes Disk Diffusion Method  561
∙∙ Dilution Methods  563
∙∙ Antibiotic Assays in Body Fluids   564
81. Antimicrobial Chemotherapy 565
∙∙ Antimicrobial Agent  565
∙∙ Antibiotic 565
∙∙ Chemotherapeutic Agents  565
∙∙ Antibacterial Agents  565
∙∙ Mechanisms of Action of Antibacterial Drugs  566
∙∙ Antibiotic Resistance  569
82. Immunoprophylaxis 571
∙∙ Vaccines   571
∙∙ Immunization 572
83. Bacteriology of Water, Milk and Air 575
∙∙ Bacteriology of Water  575
∙∙ Bacterial Flora in Water  575
∙∙ Factors Determining the Number of Bacteria in Water  575
∙∙ Water-borne Pathogens  576
∙∙ Collection of Water Samples  576
∙∙ Bacteriological Examination of Water  576
∙∙ Bacteriology of Milk  577
∙∙ Examination of Air  578
84. Hospital Waste Management 580
∙∙ Universal Precautions  580
∙∙ Definition of Biomedical Waste (BMW)  580
∙∙ Categories of Biomedical Waste  581
∙∙ Waste Segregation   581
∙∙ Treatment and Disposal Techno­logies for Healthcare Waste  581
∙∙ Disposal 582
85. Vehicles and Vectors 584
86. Emerging and Re-emerging Infectious Diseases 586
∙∙ Re-emerging, or Resurging Diseases  586

Section 7: Diagnostic Medical Microbiology


87. Molecular Detection of Microorganisms 589
∙∙ Molecular Methods  589
∙∙ Applications of Molecular Methods in Clinical Laboratory  590

www.ebook3000.com
xx  |  Essentials of Microbiology
88. Staining Methods 592
∙∙ Preparing Film or Smear for Staining  592
∙∙ Types of Stain  592
∙∙ Stained Preparations  592
∙∙ Simple Stains  593
∙∙ Differential Stains  593
∙∙ Special Stains  594
89. Practical Microbiology for MBBS Students 596
∙∙ Spots 596
∙∙ Staining for Practical Examination  598
∙∙ Reagents 600
∙∙ Gram-positive and Gram-negative Bacteria  601
∙∙ Identification of Bacterial Culture  601
∙∙ Staphylococcus 605
∙∙ Bacillus, Proteus sp.  606
∙∙ Lactose Fermenter (LF)  607
∙∙ Stool/Feces Examination  609

Index 615
Section 1: General Bacteriology

1
Chapter

Historical Development of Microbiology

Learning Objectives

∙∙ Contributions of Antony van Leeuwenhoek ∙∙ Koch’s postulates


∙∙ Contributions of Louis Pasteur ∙∙ Contributions of Paul Ehrlich.
∙∙ Contributions of Robert Koch

INTRODUCTION Antony van Leeuwenhoek (1632–1723)


Microbiology is the study of living organisms of The credit for having first observed and reported
microscopic size. Medical microbiology is the bacteria belongs to Antony van Leeuwenhoek.
subdivision concerned with the causative agents Antony van Leeuwenhoek, the Dutchman, was
of infectious disease of man, the response of the a draper and haberdasher in Delft, Holland.
host to infection and various methods of diagnosis, His hobby was grinding lenses and observing
treatment and prevention. diverse materials through them. He was the first
person to observe microorganisms (1673) using
a simple microscope. In 1683, he made accurate
INFECTION AND CONTAGION descriptions of various types of bacteria and
Concept of contagion: Long before microbes communicated them to the Royal Society of
had been seen, observations on communicable London. He recognized them as living crea­tures
diseases had given rise to the concept of contagion: (animalcules) and to Leeuwenhoek the world of
the spread of disease by contact, direct or indirect. ‘little animalcules’ represented only a curiosity
This idea was implicit in the laws enacted in early of nature. Their importance in medicine and in
biblical times to prevent the spread of leprosy. other areas of biology came to be recognized two
centuries later.
Invisible living creatures produced disease:
Varro in the 2nd century BC later recorded the Contributions of Antony van
principle of contagion by invisible creatures. Leeuwenhoek
Roger Bacon, in the 13th century, more than 1. He constructed the first microscope.
a millennium later, postulated that invisible 2. The first person to observe microorganisms
living creatures produced disease. Fracastorius (1673).
(1546), a physician of Verona, concluded that 3. Provided accurate description of bacteria.
communicable diseases were caused by living
agents (germs) ‘seminaria’ or ‘seeds’. Kircher CONFLICT OVER SPONTANEOUS
(1659) reported finding minute worms in the blood GENERATION
of plague victims, but with the equipment available
to him, it is more likely that what he observed were Spontaneous Generation (Abiogenesis)
only blood cells. John Needham (1713–1781), the English priest
in 1745 published experiments purporting
DISCOVERY OF MICROORGANISMS the spontaneous generation (abiogenesis) of
microorganisms in putrescible fluids. Lazzaro
As microbes are invisible to the unaided eye, direct Spallanzani (1729–1799), an Italian priest and
observation of microorganisms had to await the naturalist opposed this view who boiled beef broth
development of the microscope. for an hour, sealed the flasks, and observed no

www.ebook3000.com
Chap-01.indd 1 15-03-2016 11:32:50 AM
2  |  Section 1: General Bacteriology
formation of microbes. Franz Schulze (1815–1873), Louis Pasteur (Fig. 1.1) is known as ‘Father
Theodore Schwann (1810–1882), Georg Friedrich of Microbiology’ because his contribution led to
Schroder and Theodor von Dusch attempted to the development of microbiology as a separate
counter such arguments. Louis Pasteur of proved scientific discipline.
conclusively that all forms of life, even microbes,
arose only from their like and not de novo. Contributions of Louis Pasteur in
John Tyndall (1820–1893), the English Microbiology (Box 1.1)
physicist finally in 1877 proved and was able
∙∙ Coined the term ‘Microbiology’: For the study
to explain satisfactorily the need for prolonged
of living organnisms of microscopic size.
heating to eliminate microbial life from infusions.
∙∙ Proposed germ theory of disease: He
Intermittent heating, now called tyndallization,
established that putrefaction and fermentation
killed both heat-stable form and a heat-sensitive
was the result of microbial activity and that
form of bacteria.
different types of fermentations were associated
with different types of microoganisms (1857).
ROLE OF MICROORGANISMS IN DISEASEs
∙∙ Disapprove d the or y of sp onta­n e ous
Augustino Bassi (1773–1856) demonstrated in generation: He disapproved the theory of
1835 that a silkworm disease called ‘muscardine’ sponta­neous generation in 1860–1861 in public
was due to a fungal infection. MJ Berkeley (1845) controversy with Pouchet who was a proponent
proved that the great potato blight of Ireland was of spontaneous generation. In a series of
caused by a fungus. Following his success with the classic experiments in the swan-necked flasks,
study of fermentation, Pasteur imeestigated the Pasteur proved conclusively by demonstrating
pebrine disease of silueosm. the the ubiquity of microorganisms that all
Indirect transmission was recognized forms of life, even microbes, arose only from
in the 1840s, when American poet-physician their like and not de novo.
Oliver Wendell Holmes (1843) in Boston, USA ∙∙ Developed sterilization techniques and
and Ignaz Semmelweis in Vienna (1846) had developed the steam sterilizer, hot-air oven and
independently concluded that puerperal sepsis was autoclave in the course of these studies.
contagious. Semmelweis also identified its mode ∙∙ Developed methods and techniques for
of transmission by doctors and medical students cultivation of microorganisms.
attending on women in labor in the hospital and ∙∙ Studies on pebrine (silkworm disease),
had prevented it by the simple measure of washing anthrax, chicken cholera and hydrophobia.
hands in an antiseptic solution. Semmelweis was ∙∙ Pasteurization: He deviced the process of
persecuted by medical orthodoxy and driven destroying bacteria, known as pasteurization.
insane for the service to medicine and humanity. ∙∙ Coined the term ‘Vaccine’: It was Pasteur who
coined the term vaccine for such prophylactic
SCIENTIFIC DEVELOPMENT OF preparations.
MICROBIoLOGY ∙∙ Discovery of the process of attenuation
and chicken cholera vaccine: An accidental
The development of microbiology as a scientific
observation that chicken cholera bacillus
discipline dates from Louis Pasteur, perfection
cultures left on the bench for several weeks lost
on microbiological studies by Robert Koch, the
their pathogenic property but retained their
introduction of antiseptic surgery by Lord Lister
ability to protect the birds against subsequent
and contributions of Paul Ehrlich in chemotherapy.
infection by them, led to the discovery of the
process of attenuation and the development
LOUIS PASTEUR (1822–1895)
of live vaccines.
Louis Pasteur (1822– ∙∙ Developed live attenuated anthrax vaccine:
1895) (Fig. 1.1) was born He attenuated cultures of the anthrax bacillus
in the village of Dole, by incubation at high temperature (42–43°C)
France on December 27, and proved that inoculation of such cultures
1822, the son of humble in animals induced specific protection against
p a re nt s. Hi s f at h e r anthrax. The success of such immunization
was a tanner. He was was dramatically demonstrated by a public
originally trained as a experiment on a farm at Pouilly-le-Fort (1881)
chemist, but his studies during which vaccinated sheep, goats and cows
on fermentation led him were challenged with a virulent anthrax bacillus
to take interest in micro- Fig. 1.1: Louis Pasteur culture. All the vaccinated animals survived
organisms. His discoveries revolution­ized medical the challenge, while an equal number of
practice, although he never studied medi­cine. unvaccinated control animals succumbed to it.

Chap-01.indd 2 15-03-2016 11:32:51 AM


Chapter 1: Historical Development of Microbiology  | 3
∙∙ Developed rabies vac­cine in 1885. He did not 5. Koch’s postulates: His criteria for proving the
know that rabies was caused by a virus but he causal relationship between a microorganism
managed to develop a live attenuated vaccine and a specific disease are known as Koch’s
for the disease. postulates.
∙∙ Notic e d pneumo c o c ci: Pneumo co cci
were first noticed by Pasteur and Sternberg Koch’s postulates
independently in 1881. According to Koch’s postulates, a microorganism can be
accepted as the causative agent of an infectious disease
only if the following conditions are satisfied:
JOSEPH LISTER (1827–1912) Postulate 1: The organism should be regularly found in
Joseph Lister was a professor of Surgery in Glasgo the lesions of the disease.
Royal Infirmary. He applied Pasteur’s work and Postulate 2: It should be possible to isolate the organism
in pure culture from the lesions.
introduced antiseptic techniques in surgery (1867).
Postulate 3: Inoculation of the pure culture into suitable
The approach was remarkably successful effecting
laboratory animals should reproduce the lesion of the
a pronounced drop in mortality and morbidity disease.
due to surgical sepsis. He established the guiding Postulate 4: It should be possible to reisolate the
principle of antisepsis for good surgical practice. organism in pure culture from the lesions produced in
Lister antiseptic surgery innvolved carbolic acid the experimental animals.
and was cumbersome and hazardous but was a Subsequently, an additional fifth criterion introduced
milestone in the evolution of surgical practice states that specific antibodies to the organism should
from the era of ‘laudable pus’ to modern aseptic be demonstrable in the serum of patients suffering from
techniques. For this work, he is called the ‘Father the disease.
Limitations of Koch’s postulates: These criteria have
of Modern Surgery’.
proved invaluable in identifying pathogens, but they
cannot always be met, for example, some organisms
Robert Koch (1843–1910) (Fig. 1.2) (including all viruses) cannot be grown on artificial media,
and some are pathogenic only for men. Mycobacterium
Robert Koch (Fig. 1.2) was a German physician. leprae, causative agent of leprosy, has not been cultured
The first direct demonstration of the role of bacteria on artificial medium so far and not fulfilling Koch’s
in carrying the disease came by the study of anthrax postulates.
by Koch. Winner of the Nobel Prize in 1905, Robert
Koch is known as ‘Father of Bacteriology’. 6. Koch’s phenomenon: Koch (1890) observed
that a guinea pig already infected with the
Contributions of Robert Koch tubercle bacillus responded with an exaggerated
1. Staining techniqes: He described methods for expresssion when injected with the tubercle
the easy microscopic examination of bacteria bacillus or its protein. This hypersensitivity
(1877). reaction is known as Koch’s phenomenon.
2. Hanging drop method: He was the first to use Important discoveries by other scientists: Hansen
hanging drop method by studying bacterial (1874) described the leprosy bacillus; Neisser
motility. (1879) discovered the gonococcus; Eberth (1880)
3. He deviced a simple method for isolating observed the typhoid bacillus; Alexander Ogston
pure cultures of bacteria by plating out mixed (1881) described the staphylococci; Loeffler (1884)
material on a solid culture medium. described the diphtheria bacillus; Nicolaier (1884)
4. Discoveries of the causal agents of anthrax observed the tetanus bacillus; Rosenbach (1886)
(1876), tuberculosis (1882) and cholera (1883). demonstrated the tetanus bacillus; Fraenkel
(1886) described the pneumococcus; in 1887,
Weichselbakum described and isolated the
meningococcus; in 1887 Bruce identified the
causative agent of malta fever; in 1905 Schaudin
and Hoffman discovered the syphilis.

PAUL EHRLICH (1854–1915)


Paul Ehrlich was an outstanding German Scientist
and genius of extraordinary activity. He was also
known as the ‘Father of Chemotherapy’.

Contributions of Paul Ehrlich


1. Applied stains to cells and tissues for the
Fig. 1.2: Robert Koch purpose of revealing their functions.

www.ebook3000.com
Chap-01.indd 3 15-03-2016 11:32:51 AM
4  |  Section 1: General Bacteriology
2. Reported acid-fastness of tubercle bacillus difficult. Larger viruses could be seen under
3. Introduced methods of standardizing toxin light microscope after appropriate staining but
and antitoxin their detailed morphology could only be studied
4. Proposed ‘side chain theory’ of anti­b ody by electron microscope by Ruska (1934).
production 4. Cultivation of viruses
5. Salvarsan introduction: He introduced The technique of growing them on chick
salvarsan, an arsenical com­pound, sometimes embryos was developed by Goodpasture in
called the ‘magic bullet’. It was capable of 1930s. The use of living human and animal
destroying the spirochaete of syphilis. Later tissue cells for the in-vitro culture of viruses was
on, he discovered neosalvarsan. Thus, he developed by John Enders (1949) and others.
created a new branch of medicine known as 5. Virus infection and malignancy
chemotherapy. i. Leukemia: Vilhelm Ellerman and Oluf
Bang (1908) in Copenhagen put forth the
DISCOVERY OF VIRUSES possibility that virus infection could lead to
As a science, virology evolved later than malignancy.
bacteriology. Although the physical nature of ii. Sarcoma in fowls: Peyton Rous (1911)
viruses was not fully revealed until the invention three years later isolated a virus causing
of the electron microscope, the infections they sarcoma in fowls. Several viruses have been
cause have been known and feared since the dawn blamed to cause natural and experimental
of history. tumors in birds and animals. Viruses also
1. Infectious Agents Smaller than Bacteria cause malignant transformation of the
The infectious agents of numerous diseases infected cells in tissue culture.
were being isolated and many infectious 6. Viral oncogenesis: The discovery of viral and
diseases had been proved to be caused by cellular oncogenes have put forth the possible
bacteria. But there remained a large number mechanisms of viral oncogenesis. Positive
of diseases for which no bacterial cause could proof of a virus causing human malignancy
be established until it was realized that the was established when the virus of human T-cell
responsible agents were smaller than bacteria. leukemia was isolated in 1980.
2. Various Infections 7. Bacteriophages: Frederick W Twort (1915)
i. Rabies in dogs: Pasteur had suspected and Felix D Herelle (1917) independently
that rabies in dogs could be caused by a discovered a lytic phenomenon in bacterial
microbe too small to be seen under the cultures. The agents responsible were termed
microscope. bacteriophages (viruses that attack bacteria).
ii. Tobacco mosaic disease: Iwanowski
(1892), Russian scientist and Martinus IMMUNITY AND IMMUNIZATION
Beijrinck (1898) in Holland attributed the
cause of tobacco—mosaic disease to the 1. Ancient knowledge
infectious agents in bacteria-free filtrates to The practice of producing a mild form of
be living but fluidcontagium vivum fluidum smallpox intentionally (variolation) was
and introduced the term virus (Latin for prevalent in India, China and other ancient
‘poison’) for such filterable infectious agents. civilizations from time immemorial.
iii. Foot-and-mouth disease of cattle: Friedrich 2. Edward Jenner (1749–1823)
Loeffler and Paul Frosch at the same time in The first scientific attempts at artificial
1898 in Germany found that foot-and-mouth immunizations in the late eighteenth century
disease of cattle was also caused by a similar were made by Edward Jenner (1749–1823)
filter-passing virus. from England. He introduced the technique of
iv. Yellow fever: The discovery of first human vaccination using a related but mild live virus
disease proved to have a viral etiology was of cowpox (1796). Edward Jenner is known as
yellow fever. The US Army Yellow Fever the ‘Father of Immunology’.
Commission under Walter Reed in Cuba 3. Live vaccines
(1902) showed that this human disease Further work on immunization was carried
(yellow fever) was not only a filterable virus out by Louis Pasteur and derived attenuated
but also transmitted through the bite of (reduced in virulence) live vaccines for fowl
infected mosquitoes. cholera, anthrax, swine erysipelas and rabies.
3. Electron microscope: Viruses could not be 4. Vaccine for hydrophobia
visualized under the light microscope or grown Pasteur’s development of a vaccine for hydro­
in culture media. So investigation of viruses phobia made the greatest impact in medicine.
and the disease caused by them were rendered This was acclaimed throughout the world.

Chap-01.indd 4 15-03-2016 11:32:51 AM


Chapter 1: Historical Development of Microbiology  | 5
5. Antibodies and complement the original suggestion of Thomas (1959).
Nuttal (1888) observed that defibrinated Malignancy was thought to be a failure of this
blood had a bactericidal effect and Buchner function.
(1889) noticed that this effect was abolished 10. Transplantation
by heating the sera for one hour at 55°C. This Understanding of the immunological basis
heat-labile bactericidal factor was termed of transplantation, largely due to the work of
‘alexine’. The first step in elucidating the Medawar and Burnet.
mechanisms of acquired immunity was the
discovery of antibodies by Emil von Behring
and Shibasaburo Kitasato (1890) in the sera SEROTHERAPY AND CHEMOTHERAPY
of animals which had received sublethal dose Antisera: The work of Behring and Kitasato led to
of diphtheria or tetanus toxoid. Pfeiffer (1893) the successful use of antisera raised in animals for
demonstrated bactericidal effect in vivo by the treatment of patients with diphtheria, tetanus,
injecting live cholera vibrios intraperitoneally pneumonia and other diseases.
in guinea pigs previously injected with
killed vibrios. Bordet (1895) defined two Magic bullet: Ehrlich (1909) discovered salvarsan
components in this reaction, the first being (arsenaphenamine), sometimes called the ‘magic
heat stable substance ‘antibody’ found in the bul­let’, was capable of destroying the spirochaete
immune sera and the second being heat labile of syphilis. In 1912, he discovered neosalvarsan.
subsequently named ‘complement’. This gave him the title, ‘Father of Chemotherapy’.
6. Cellular concept of immunity
Elie Metchnikoff (1883) discovered the
Antibiotics—A Fotunate Accident
phenomenon of phagocytosis and developed
the cellular concept of immunity. Paul Ehrlich The modern era of antibiotics developed only
hypothesized that immunity could be explained after Gerhard Domagk (1895–1964) found that
by the presence of noncellular components of prontosil (the forerunner of sulfonamides) had
blood. Wright (1903) discovered opsonization. a dramatic effect on streptococcal infection in
Both Metchnikoff and Ehrlich shared a 1935. Sir Alexander Fleming (1881–1955) made
Nobel Prize in 1908 for their contributions accidental discovery that the fungus Penicillium
to the emerging science of immunology. notatum produces a substance which destroys
The pioneering work of Landsteiner laid the staphylococci. In the 1940s, Florey and Chain and
foundation of immunochemistry. their associates demonstrates its clinical value. This
7. Allergy was the beginning of the antibiotics era. Selman
Anaphylaxis—Portier and Richet (1902), Waksman exploited the potential for antibiotic
studying the effect of the toxic extracts of sea production among the soil microorganisms in the
anemones in dogs, made the paradoxical 1940s.
observation that dogs which had prior contact
with the toxin were abnormally sensitive to NOBEL PRIZES AWaRDED FOR
even minute quantities of it subsequently. RESEARCH IN MICROBIOLOGY
This phenomenon was termed anaphylaxis.
8. Selection theory of antibody The number of Nobel laureates in Medicine and
In 1955, Jerne proposed the ‘natural selection Physiology for their contribution in microbiology
theory’ of antibody synthesis. Burnet (1957) is an evidence of the positive contribution made
modified this into clonal selection theory. to human health by the science of microbiology.
9. Immunological surveillance About one-third of these have been awarded to
Burnet (1967) developed the concept of scientists working on microbiological problems
immunological surveillance based on (Table 1.1).

Table 1.1  Nobel laureates for research in microbiology


Year Nobel laureates Contribution
1901 Emil A von Behring Developed a diphtheria antitoxin
1902 Ronald Ross Discovered how malaria is transmitted
1905 Robert Koch Tuberculosis—discovery of causative agent
1907 CLA Laveron Discovery of malaria parasite in an unstained
preparation of fresh blood

Contd...

www.ebook3000.com
Chap-01.indd 5 15-03-2016 11:32:51 AM
6  |  Section 1: General Bacteriology
Contd...

Year Nobel laureates Contribution


1908 Paul Ehrlich and Elie Metchnikoff Developed theories on immunity
Described phagocytosis, the intake of solid materials
by cells
1913 Charles Richet Anaphylaxis
1919 Jules Bordet Discovered roles of complement and antibody in
cytolysis, developed complement fixation test
1928 Charles Nicolle Typhus exanthematicus
1930 Karl Landsteiner Described ABO blood groups; solidified chemical
basis for antigen-antibody reactions
1939 Gerhardt Domagk Antibacterial effect of prontosil
1945 Alexander Fleming, Ernst Chain and Howard Florey Discovered penicillin
1951 Max Theiler Yellow fever vaccine
1952 Selman A Waksman Development of streptomycin. He coined the term
‘antibiotic’
1954 John F Enders, Thomas H Weller and Frederick C Cultured poliovirus in cell cultures
Robbins
1960 Sir Macfarlane Burnet and Sir Peter Brian Medawar Immunological tolerance, clonal selection theory
1962 James D Watson, Frances HC Crick and Maurice AF Double helix structure of deoxyribonucleic acid
Wilkins (DNA)
1966 Peyton Ross Viral oncogenes (avian sarcoma)
1968 Robert Holley, Har Gobind Khorana, and Marshall Genetic code
W Nirenberg
1969 Max Delbruck, AD Hershey and Salvador Luria Mechanism of virus infection in the living cells
1972 Gerald M Edelman and Rodney R Porter Described the nature and structure of antibodies
1975 David Baltimore, Renato Dulbecco and Howard M Interactions between tumor viruses and genetic
Temin material of the cells
1977 Rosalyn Yalow Developed inmmunoassay
1980 Baruj Benacerraf, Jean Dausset and George Snell HLA antigens
1984 Cesar Milstein, Georges Kohler Neils Jerne Developed hybridoma technology for production of
monoclonal antibodies
1987 S Tonegawa Described the genetics of antibody production
1989 J Michael Bishop and Harold E Varmus Discovered cancer-causing genes called oncogenes
1990 Joseph E Murray and E Donnall Thomas Performed the first successful organ transplants by
using immunosuppressive agents
1993 Kary B Mullis Discovered the polymerase chain reaction (PCR) to
amplify DNA
1996 Peter C Doherty and Rolf M Zinkernagel Cell-mediated immune defences
1997 Stanley B Prusiner Prion discovery
2001 Leland H Hartwell, Paul M Nurse, and R Timothy Hunt Discovered genes that encode proteins regulating
cell division
2005 Barry J Marshall and J Robin Warren Helicobacter pylori and its role in gastritis and peptic
ulcer disease
2007 Mario R Capecchi, Oliver Smithies and Sir Martin J Evans Creation of knockout mice for stem cell research
2008 Luc Montagnier and Francoise Barre-Sinoussi Discovery of human immunodeficiency virus
Harald zur Hausen Human papillomavirus causing cervical cancer
2011 Bruce A, Beutler and Jules A Hoffmann Ralph M Discoveries concerning the activation of innate
Steinman immunity
Discovery of the dendritic cell and its role in active
immunity

Chap-01.indd 6 15-03-2016 11:32:51 AM


Chapter 1: Historical Development of Microbiology  | 7

Key Points 2. Who is the father of microbiology?


a. Louis Pasteur
™™ Microbiology is the study of living organisms of b. Robert Koch
microscopic size. c. Paul Ehrlich
™™ Antony van Leeuwenhoek was the first person to d. Joseph Lister
describe micro-organisms. 3. Who is the father of bacteriology?
™™ Louis Pasteur: He is known as the ‘Father of a. Louis Pasteur
Microbiology’. b. Robert Koch
™™ Joseph Lister: Developed a system of antiseptic c. Paul Ehrlich
surgery. For this work he is called the ‘Father of d. Joseph Lister
Modern Surgery’
™™ Robert Koch: Koch’s postulates are used to prove a 4. Which bacteria is the human pathogen but still
direct relationship be­tween a suspected pathogen does not fulfil Koch’s postulates criteria?
and a disease. a. Staphylococcus aureus
™™ Paul Ehrlich is known as ‘Father of Chemotherapy’. b. Bacillus anthracis
™™ Edward Jenner is known as the ‘Father of Immunology’. c. Mycobacterium leprae
™™ Ehrlich is given the title ‘Father of Chemotherapy’. d. Psedomonas aeruginosa
5. Who is the father of chemotherapy?
Important question a. Louis Pasteur
b. Robert Koch
Write short notes on: c. Paul Ehrlich
a. Contributions of Antony van Leeuwenhoek d. Joseph Lister
b. Contributions of Louis Pasteur 6. Who is the father of immunology?
c. Contributions of Robert Koch a. Louis Pasteur
d. Koch’s postulates b. Robert Koch
e. Contributions of Edward Jenner c. Paul Ehrlich
f. Contributions of Paul Ehrlich d. Edward Jenner
g. Name four Nobel laureates in Microbiology
7. Which first human disease proved to be of viral
origin?
Multiple choice questions (MCQs) a. Smallpox
1. Who was the first person to describe micro- b. Dengue
organisms ? c. Yellow fever
a. Louis Pasteur d. Rabies
b. Robert Koch
c. Paul Ehrlich Answers (MCQs)
d. Antony van Leeuwenhoek Ans. 1. d; 2. a; 3. b; 4. c; 5. c; 6. d; 7. c

www.ebook3000.com
Chap-01.indd 7 15-03-2016 11:32:51 AM
2
Chapter

Microscopy

Learning Objectives

After reading and studying this chapter, you should ∙∙ E xplain the principles and describe the uses of the
be able to: following: darkfield microscopy; phase-contrast;
∙∙ Discuss microscopic methods microscopy; fluore­scent microscopy and electron
microscopy.

Introduction 1. Compound Light Microscopy


Microscope is an optical instrument used to A series of finely ground lenses forms a clearly
magnify (enlarge) minute objects or micro- focused image that is many times larger than the
organisms which cannot be seen by naked eye. specimen itself. This magnification is achieved
when light rays from an illuminator, the light
MICROSCOPY: INStRUMENTS source used to illuminate the specimen positioned
on a stage, pass through a condenser, which
Microscopic Methods has lenses that direct the light rays through the
A. Light Microscopy specimen. From here, light rays pass into the
1. Brightfield (light) microscopy objective lenses, the lenses closest to the speci­
2. Darkfield microscopy men. The image of the specimen is magnified again
3. Phase-contrast microscopy by the ocular lens, or eyepiece. Three different
4. Fluorescent microscopy objective lenses are com­monly used: low power
5. Confocal (x10); high dry (x40); and oil immersion (x100).
B. Electron Microscopy
–– Transmission Resolution
–– Scanning The limitation of brightfield microscopy is the reso­
C. Scanning Probe Microscopes lution (also called resolving power) of the image
(i.e. the ability to distinguish that two objects are
A. LIGHT MICROSCOPY separate and not one). The resolving power of a
Light microscopy refers to the use of any kind microscope is determined by the wave­length of
of microscope that uses visible light to observe light used to illuminate the subject and the angle
specimens. A modern compound light microscope of light entering the objective lens (referred to as
(LM) has a se­ries of lenses and uses visible light as the numerical aperture).
its source of illumina­tion. Immersion oil: Immersion oil is placed between the
glass slide and the oil immersion objective lens. The
Principles of Light Microscopy: The immer­sion oil has the same refractive index as glass,
Brightfield Microscopy so the oil becomes part of the optics of the glass of
In light microscopy, light typically passes the microscope. The oil enhances the resolution by
through a specimen and then through a series preventing light rays from dispersing and changing
of magnifying lenses. The most common type of wavelength after passing through the specimens.
light microscope, and the easiest to use, is the A specific objective lens, the oil immersion lens,
brightfield microscopy, which evenly illuminates is designed for use with oil; this lens provides 100x
the field of view. magnification on most light microscopes.
Chapter 2: Microscopy  | 9
2. Darkground (Darkfield) Microscopy Fluorescence Microscopy
Darkfield microscopy is frequently performed on the Fluorescence microscopy takes advantage of
same microscope on which brightfield microscopy fluorescence, the ability of substances to absorb
is performed. Instead of the normal condenser, a short wave­lengths of light (ultraviolet) and give
darkfield microscopy uses a darkfield condenser off light at a longer wavelength (visible). If tissues,
that contains an opaque disc. The disc blocks light cells or bacteria are stained with a fluorescent dye
that would enter the objective lens directly. Only and are examined under the microscope with ultra­
light that is reflected off (turned away from) the violet light instead of ordinary visible light, they
specimen enters the objective lens. Because there become luminous and are seen as bright objects
is no direct background light, the specimen appears against a dark background.
light against a dark background. This creates a ‘dark-
field’ that contrasts against the highlighted edge of Principal Use
the specimens and results when the oblique rays are The principal use of fluorescence microscopy is
reflected from the edge of the specimen upward into a di­agnostic technique called the fluorescent
the objective of the microscope. antibody (FA) technique, or immunofluorescence
employed for detection of antigen (direct
Use fluorescent antibody technique) and antibodies
This technique is particu­larly valuable for observing (indirect fluorescent antibody methods).
organisms such as Trep­o nema pallidum, a
spirochete which cannot be observed with direct Epifluores­cence Microscopy
light. A common variation of the standard fluorescence
microscope is the epifluores­cence microscope,
3. Phase-contrast Microscopy which projects the ultra­violet light through the
Principle: In a phase-contrast microscope, one set objective lens and onto the specimen.
of light rays comes directly from the light source.
The other set comes from light that is reflected Confocal Microscopy
or diffracted from a particular structure in the In confocal microscopy, lenses focus a laser beam
specimen. When the two sets of light rays—direct to illu­minate a given point on one vertical plane of a
rays and reflected or dif­fracted rays—are brought specimen. In effect, this micro­scope is a miniature
together, they form an image of the specimen on CAT scan for cells.
the ocular lens, containing areas that are relatively
light (in phase), through shades of gray to black (out B. Electron Microscopy
of phase). Through the use of annular rings in the
condenser and the objective lens, the differences in ∙∙ Electron microscopy is in some ways
phase are amplified so that in-phase light appears comparable to light microscopy. Rather than
brighter than out-of-phase light. The special phase using glass lenses, visible light, and the eye to
condenser consists of annular diaphragms on a observe the specimen, the electron microscope
rotating disk fitted to the bottom of the condenser. uses electromagnetic lenses, electrons,
and a fluorescent screen to produce the
Uses magnified image. That image can be captured
on photographic film to create an electron
1. To study unstained living cells.
photomi­crograph.
2. Detailed examination of internal structures in
∙∙ The electron beam is focused by circular electro­
living microorganisms.
magnets, which are analogous to the lenses in
3. To study flagellar movements and motility of the light microscope. The object which is held
bacteria and protozoans. in the path of the beam scatters the electrons
4. To study intestinal and other live protozoa such and produces an image which is focused on a
as amoebae and Trichomonas. fluorescent viewing screen.
5. To examine fungi grown in culture. ∙∙ The wavelength of electrons used in an EM is
0.005 nm as compared to 500 nm with visible
Differential Interference Contrast (DIC) light, i.e. about 100,000 times shorter than that
Microscopy of ordi­nary light. Theoretically, the resolv­ing
Differential interference contrast (DIC) microscopy power of the EM should be 100,000 times (reso­
is similar to phase-contrast microscopy in that lution down to 0.0025 nm). In practice, the best
it uses dif­ferences in refractive indices. The res­olution that can be obtained is 0.3–0.5 nm,
resolution of a DIC microscope is higher than that a hun­dred times better than that of the light
of a standard phase-contrast microscope. microscope.

www.ebook3000.com
10  |  Section 1: General Bacteriology
Types of Electron Microscopes ™™ Phase-contrast microscopy: It allows the detailed
There are two types of electron microscopes in observation of the living organisms.
gen­eral use: ™™ Fluorescence microscopy: Fluorescence microscopy
is used primarily in a diagnostic procedure
called fluorescent-antibody (FA) technique, or
(i) Transmission Electron Microscope (TEM) immunofluorescence.
In transmission electron microscope (TEM), ™™ Electron microscopy: Electron microscopes use
electrons like light pass directly through the electromagnetic lenses instead of glass lenses,
electrons and fluorescent screens to produce a
specimen that has been prepared by thin magnified image. There are two types (i) Transmission
sectioning, freeze fracturing, or freeze etching. It electron microscopes (TEMs); and (ii) Scanning
is used to observe fine details of cell structure. electron microscopes
™™ Scanned-probe microscopy: Their resolving power is
(ii) Scanning Electron Microscope (SEM) much greater than the electron microscope.

A scanning electron microscope scans a beam


of electrons back and forth over the surface of a Important question
specimen producing three-dimensional views of Write short notes on:
the surfaces of whole microor­ganisms. a. Darkfield microscopy
b. Fluorescent microscopy
C. SCANNING-PROBE MICROSCOPY c. Phase-contrast microscopy
d. Electron microscopy.
Scanning probe microscopes map the bumps
and valleys of a surface on an atomic scale. Their Multiple choice questions (MCQs)
resolving power is much greater than the electron
microscope, and the samples do not need special 1. Which of the following is not a modification of a
preparation as they do for electron microscopy. compound microscope:
a. Bright microscopy
Among the new scanned-probe microscopes are:
b. Darkfield microscopy
1. Scanning tunneling microscopy (STM): There c. Elecron micrscopy
are used to provide incredibly detailed views of d. Phase-contrast microscopy
mol­ecules, such as DNA. 2. Wavelenth of electrons used in an electron
2. Atomic force microscopy (AFM): These microscopy is:
produce three-dimensional images of the a. 0.001 nm
surface of a molecule. b. 0.005 nm
c. 100 nm
Key Points d. 500 nm
™™ Microscopy: A simple microscope consists of one 3. Theoretically, the resolv­ing power of the electrone
lens; a compound microscope has multiple lenses. micro­scope is:
Compound Light Microscopy a. 10,000 times
™™ The most common microscope used in microbiology b. 100,000 times
is the compound microscope/bright field c. 5,00,000 times
microscope/light microscope. d. 1,000 times
™™ Darkfield microscopy: It is most useful for detecting
the presence of extremely small organisms. Answers (MCQs)
Ans: 1. b; 2. b; 3. b
3
Chapter

Morphology of Bacteria

LEARNING OBJECTIVES

After reading and studying this chapter, you should be ∙∙ Discuss capsule or bacterial capsule
able to: ∙∙ Describe bacterial flagellae
∙∙ Differentiate between prokaryotes and eukaryotes ∙∙ Describe fimbriae or pili
∙∙ Describe anatomy of bacterial cell ∙∙ Discuss bacterial spores or endospores
∙∙ Describe cell envelope ∙∙ Explain L-forms of bacteria.
∙∙ Describe bacterial cell wall

INTRODUCTION Table 3.1  Principle differences between prokar­


yotic and eukaryotic cells
The bacteria are single-celled organisms that
reproduce by simple division, i.e. binary fission. Characteristics Prokaryotic cell Eukaryotic cell
Whittaker’s system recognizes five kingdoms of
1. Size 0.5–3 µm >5 µm
living things—Monera (bacreia), Protista, Fungi, (approximate)
Plantae, and Animalia. Five kingdoms have been
2. Nucleus
modified further by the development of three
Nuclear Absent Present
domains, or Superkingdoms system—the Bacteria, membrane Absent Present
the Archaea (meaning ancient) and the Eucarya. Nucleolus One (circular) More than one
Chromosome (linear)
COMPARISON BETWEEN PROKARYOTIC
Deoxyribonucleo­
AND EUCARYOTIC CELLS protein Absent Present
Division By binary fission By mitosis
All living organisms on earth are composed of
one or the other of two types of cells: prokaryotic 3. Cytoplasm
cells and eukaryotic cells based on differences in Cytoplasmic Absent Present
streaming Absent Present
cellular organization and biochemistry (Table 3.1). Mitochondria Absent Present
1. Prokaryotes: All bacteria and blue-green algae Golgi apparatus Absent Present
are prokaryotes. Lysosomes Absent Present
2. Eukaryotes: Other algae (excluding blue-green Pinocytosis Absent Present
algae), fungi, slime moulds, protozoa, higher Endoplasmic
reticulum
plants and animals are eukaryotic.
4. Chemical
SIZE OF BACTERIA composition
Sterol Absent Present
Bacteria are very small in size. The unit of Muramic acid Present Absent
measurement in bacteriology is the micron or 5. Examples Eubacteria, Fungi, slime
micrometer (mm) 1 micron (m) or micrometer (mm) Archaea moulds,
= a millionth part of a meter or a thousandth part All bacteria and protozoa,
of a millimeter. blue-green algae higher plants
1 millimicron (mm) or nanometer (nm) = and animals
including
one thousandth of a micron or one-millionth of a humans
millimeter.

www.ebook3000.com
Chap-03.indd 11 15-03-2016 11:09:37
12  |  Section 1: General Bacteriology
1 Angstrom unit (A) = one-tenth of a nanometer. 4. Spirilla: Spirilla are rigid spiral or helical forms.
The diameter of the smallest body that can be 5. Spirochaetes: Spirochaetes are flexuous spiral
resolved and seen clearly with naked eye is 200 forms.
µm. Bacteria of medical importance generally 6. Mycoplasma: Mycoplasma are cell wall
measure 0.2–1.5 µm in diameter and about 3–5 deficient bacteria and, hence, do not possess
µm in length. To see bacteria, a light microscope a stable morphology. They occur as round or
must be used. The best light microscope, using the oval bodies and interlacing filaments.
most advanced optics, is capable of magnifications
of 1000–2000 times. The electron microscope ARRANGEMENT OF BACTERIAL CELLS
provides superb resolving power. Following types
of micro­scopes are used for the examination of Pathogenic bacterial species appear as sphere
bacteria: (cocci), rods (bacilli), and spirals. Bacteria sometimes
show characteristic cellular arrangement or
STUDY OF BACTERIA grouping (Fig. 3.2). The type of cellular arrangement
is determined by the plane through which binary
Stained Preparations fission takes place and by the tendency of the
Live bacteria do not show much structural detail daughter cells to remain attached even after division.
under the light microscope due to lack of contrast.
Hence, it is customary to use staining techniques Cocci Arrangement
to produce color contrast. Various staining i. Diplococci: Cocci may be arranged in pairs
techniques are commonly used in bacteriology (diplococci), when cocci divide and remain
(See Chapter 87). together;

Shape of Bacteria
Depending on their shape, bacteria are classified
into several varieties (Fig. 3.1):
1. Cocci: Cocci (from kokkos meaning berry) are
spherical, or nearly spherical.
2. Bacilli: Bacilli (from baculus meaning rod)
are relatively straight, rod-shaped (cylindrical)
cells. In some of the bacilli, the length of the
cells may be equal to the width. Such bacillary
forms are known as coccobacilli and have to be
carefully differentiated from cocci.
3. Vibrios: Vibrios are curved or comma-
shaped rods and derive the name from their
characteristic vibratory motility.

Fig. 3.2: Arrangement of bacteria: A. Cocci: 1. Streptococci;


2. Pneumococci; 3. Gonococci; 4. Meningococci; 5. Neisseria
catarrhalis; 6. Gaffkya tetragena; 7. Sarcina; 8. Staphylococci.
Fig. 3.1: Shapes of bacteria: 1. Coccus; 2. Bacillus; 3. Vibrio; B. Bacilli: 1. Bacilli in cluster; 2. Bacilli in chains (B. anthrax); 3.
4. Spirillum; 5. Spirochete Diplobacilli (K. pneumoniae)

Chap-03.indd 12 15-03-2016 11:09:39


Chapter 3: Morphology of Bacteria  | 13
ii. Long chains: Long chains (Steptococcus, B. Cell interior: Those structures and substances
Enterococcus, and Lactococcus), when cells that are bounded by the c ytoplasmic
adhere after repeated divisions in one plane; membrane compose the cell interior and
iii. Grape-like clusters: Grape-like clusters include cytoplasm, cytoplasmic inclusions
(staphylococci), when cocci divide in random (mesosomes, ribosomes, inclusion granules,
planes; vacuoles) and a single circular chromosome
iv. Tetrads: Square groups of four cells (tetrads) of deoxyribonucleic acid (DNA).
when cocci divide in two planes as in members
of the genus Micrococcus; A. Cell Envelope and its Appendages
v. Cubical packets: Cubical packets of eight of a. The Outer Layer or Cell Envelope
cells (genus Sarcina), when cocci divide in
Cell wall
three planes.
The cell wall is the layer that lies just outside the
plasma membrane. It is strong and relatively rigid,
Bacilli Arrangement and openly porous.
Bacilli split only across their short axes. Therefore,
Functions of the cell wall:
the patterns formed by them are limited. The
1. It imparts shape and rigidity to the cell.
shape of the rod’s end often varies between
2. It supports the weak cytoplasmic membrane
species and may be flat, rounded, cigar-shaped
against the high internal osmotic pressure of
or bifurcated. Some bacilli too may be arranged
the protoplasm.
in chains (streptobacilli). Others are arranged
3. It maintains the characteristic shape of the
at various angles to each other, resembling the
bacterium.
letters ‘V’ presenting a cuneiform or Chinese
4. It takes part in cell division.
letter arrangement and is characteristic of
Corynebacterium diphtheriae. 5. It also functions in interactions (e.g. adhesion)
with other bacteria and with mammalian cells.
6. It provides specific protein and carbohydrate
ANATOMY OF THE BACTERIAL CELL receptors for the attachment of some bacterial
viruses.
The principal structures of the bacterial cell are
shown in Figure 3.3. Chemical structure of bacteria cell wall
Chemically the cell wall is composed of
Bacterial Cell Components mucopeptide (peptidoglycan or murein)
scaffolding formed by N-acetyl glucosamine and
These can be divided into:
N-acetyl muramic acid molecules alternating in
A. Cell envelope and its appendages.
chains, which are crosslinked by peptide bonds
a. The outer layer or cell envelope consists
(Fig. 3.4).
of two components:
1. Cell wall Difference between cell wall of gram-positive
C ytoplasmic or plasma memb­
2.  and gram-negative bacteria
rane—beneath the cell wall In general, the walls of the gram-positive bacteria
b. Cellular appendages: Capsule, fimbriae, have simpler chemical nature than those of gram-
and flagella negative bacteria (Table 3.2).

Fig. 3.3: Anatomy of a bacterial cell

www.ebook3000.com
Chap-03.indd 13 15-03-2016 11:09:40
14  |  Section 1: General Bacteriology

Fig. 3.5: Gram-positive bacterial cell wall

ii. Lipopoprotein: Lipopoprotein, or murein


lipoproteins seemingly attach (both covalently
and noncovalently) to the peptidoglycan
Fig. 3.4: Chemical structure of bacterial cell wall
by their protein portion, and to the outer
Table 3.2  Comparison of cell walls of gram- membrane by their lipid component.
positive and gram-negative bacteria iii. Outer membrane: External to the peptidoglycan,
and attached to it by lipoproteins is the outer
Characteristics Gram positive Gram negative membrane. It is a bilayered structure. Its inner
1. Thickness More Less leaflet is composed of phospholipid while in
2. Peptidoglycan Thick layer (16–80 2 nm (thin layer) phospholipids, the outer leaflet are replaced by
nm ) lipopolysaccharide (LPS) molecules.
3. Teichoic acid Present Absent
Functions
4. Variety of Few Several i. A protective barrier.
aminoacids
ii. Porins or transmembrane proteins: Function
5. Aromatic Absent or scant Present in the selective transport of nutrients into the
and sulfur-
cell.
containing
aminoacids iii. Lipopolysaccharide (LPS)
Lipopolysaccha­r ide (LPS) consists of three
6. Lipids Absent or scant Present
components:
7. Porin proteins Absent Present i. Lipid A: It is the lipid portion of LPS and
8. Periplasmic Absent Present is embedded in the top layer of the outer
region membrane. When gram-negative bacteria
die, they release lipid A, which functions as
an endotoxin. All the toxicity of the endotoxin
Gram-positive bacterial cell wall is due to lipid A which is responsible for the
The gram-positive bacterial cell wall (Fig. 3.5) is endotoxic activities, that is, pyrogenicity, lethal
composed mostly of several layers of peptidoglycan. effect, tissue necrosis, anticomplementary
Peptidoglycans: It is thicker and stronger than that activity, B cell mitogenicity, immunoadjuvant
of gram-negative bacteria. property and antitumor activity.
Teichoic acid: In addition, the cell walls of gram- ii. The core polysaccharide its role is to provide
positive bacteria contain teichoic acids. stability.
iii. O-polysaccharide: It extends outward from
Gram-negative bacterial cell wall
the core polysaccharide and is composed of
The gram-negative bacterial cell wall is structurally
sugar molecules. Polysaccharide represents a
quite different from that of gram-positive cells
major surface antigen of the bacterial cell. It
(Fig. 3.6). It consists of peptidog­lycan, lipoprotein,
is known as O-antigen.
outer membrane, and lipopoly­saccharide.
i. Peptidoglycan layer: Peptidoglycan layer is Demonstration of cell wall
a single-unit thick of gram-negative bacteria. The cell wall cannot be seen by light microscopy
It is bonded to lipoproteins covalently in the and does not stain with simple dyes. It can be dem­
outer membrane and plasma membrane and onstrated by:
is in the periplasm, a gel-like fluid between the i. Plasmolysis: When placed in a hypertonic
outer membrane and plasma membrane. The solution, the cytoplasm loses water by osmosis
periplasm contains a high concentration of and shrinks, while the cell wall retains its
degradative enzymes and transport proteins. original shape and size (bacterial ghost).

Chap-03.indd 14 15-03-2016 11:09:41


Chapter 3: Morphology of Bacteria  | 15

Fig. 3.6: Gram-negative cell wall

ii. Microdissection thick), elastic and can only be seen with electron
iii. Exposure to specific antibody microscope. It is a typical ‘unit membrane’,
iv. Mechanical rupture of the cell composed of phospholipids and proteins.
v. Differential staining procedures Chemically, the membrane consists of
vi. Electron microscopy. lipoprotein with small amounts of carbohydrate.
Enzymes that attack cell walls With the exception of Mycoplasma, bacterial
i. Lysozyme: The enzyme lysozyme, which is cytoplasmic membrane lacks sterols.
found in animal secretions (tears, saliva, nasal Functions of cytoplasmic membrane:
secretions) as well as in egg white, is a natural i. Semipermeable membrane: Controlling the
body defence substance which lyses bacteria inflow and outflow of metabolites to and from
of many species. the protoplasm.
ii. Autolysins: Bacteria themselves possess ii. Housing enzymes : Involved in outer
enzymes, called autolysins, able to hydrolyse membrane synthesis, cell wall synthesis,
their own cell wall substances. and in the assembly and secretion of extra­
Protoplasts and spheroplasts cytoplasmic and extracellular substances.
Protoplasts iii. Housing many sensory and chemotaxis
These are derived from gram-positive bacteria. proteins
The gram-positive cell wall is almost completely iv. Generation of chemical energy (i.e. ATP)
destroyed by lysozyme. They contain cytoplasmic v. Cell motility
membrane and cell wall is totally lacking. Typically, vi. Mediation of chromosomal segregation
a protoplast is spherical and is still capable of carry­ during replication
ing on metabolism. Protoplasts cannot revert to
normal bacterial morphology.
b. Cellular Appendages
Spheroplasts I. Bacterial capsule or slime layer
When lysozyme is applied to gram-negative cells, Structure: Many bacteria synthesize large amount of
usually the wall is not destroyed to the same extent extracellular polymer in their natural environments.
as in gram-positive cells. Spheroplasts retain outer When the polymer forms a condensed, well-defined
membrane and entrapped peptido­glycan from layer closely surrounding the cell, it is called the
gram-negative cell. It differs from the protoplast in capsule as in the Pneumococcus. If the polymer
that some cell wall material is retained. It is also a is easily washed off and does not appear to be
spherical struc­ture. They are capable of reverting associated with the cell in any definite fashion, it
to parent bacterial form when cell wall inhibitor is is referred to as a slime layer as in leuconostoc.
removed from the culture me­dium. A glycocalyx is a network of polysaccharide
2. Cytoplasmic (plasma) membrane extending from the surface of bacteria and other
Structure: The cytoplasmic (plasma) membrane cells. Capsules too thin to be seen under the light
limits the bacterial protoplast. It is thin (5–10 nm microscope are called microcapsules.

www.ebook3000.com
Chap-03.indd 15 15-03-2016 11:09:42
16  |  Section 1: General Bacteriology
Composition of capsules and slime layers: for the typing of pneumococci in the pre­
Capsules and slime layers usually are composed sulfonamide days.
of polysaccharide (for example, Pneumococcus)
Functions of capsule
or of polypeptide in some bacteria (for example,
i. Virulence factor: Capsules often act as a
Bacillus anthracis and Yersinia pestis). Some
virulence factor by protecting the bacterium
bacteria may have both a capsule and a slime layer
from ingestion by phagoc ytosis and
(for example, Streptococcus salivarius). Bacteria
noncapsulate mutant of these bacteria are
secreting large amounts of slime produce mucoid
nonvirurlent.
growth on agar, which is of a stringy consistency
ii. Protection of the cell wall: In protecting
when touched with the loop.
the cell wall attack by various kinds of
Capsulated bacteria: Streptococcus pneumoniae, antibacterial agents.
several groups of streptococci, Neisseria menin­ iii. Identification and typing of bacteria.
gitidis, Klebsiella, Haemophilus influenzae,
II. Flagella
Yersinia and Bacillus.
Motile bacteria, except spirochetes, possess one or
more unbranched, long, sinuous filaments called
Demonstration of Capsule flagella, which are the organs of locomotion.
i. Gram stain: Capsules and slime are usually
not visible in films stained by ordinary Structure: They are long, hollow, helical filaments,
methods except as clear haloes surrounding usually several times the length of the cell. They are
the stained smear. 3–20 μm long and are of uniform diameter (0.01–
ii. Special capsule-staining techniques : 0.013 μm) and terminate in a square tip. It originates
Usually employing copper salts as mordants. in the bacterial protoplasm and is extruded through
iii. India ink staining (negative staining): The the cell wall. Flagella consists of largely or entirely
capsule appears as a clear halo around the a protein, flagellin.
bacterium, against a dark background in the Flagella are highly antigenic and flagellar
film (Fig. 3.7). antigens induce specific antibodies in high titers.
iv. Electron microscope. Flagellar antibodies are not protective but are useful
v. Serological methods: Capsular material in serodiagnosis.
is antigenic and may be demonstrated by Parts and Composition
serological methods. When a suspension Each flagellum consists of three parts (Fig. 3.8):
of a capsulated bacterium is mixed with its i. Filament: The filament is the longest and most
specific anticapsular serum and examined obvious portion which extends from the cell
under the microscope, the capsule becomes surface to the tip.
very prominent and appears ‘swollen’ due to ii. Hook: The hook is a short, curved segment
an increase in its refractivity. It is known as which links the filament to its basal body and
the capsule-swelling reaction or Quellung functions as universal joint between the basal
reaction (Quellung-Ger swelling). It was body and the filament.
described by Neufeld (1902), hence called iii. Basal body: The basal body is embedded
Neufeld reaction. It was widely employed in the cell (cytoplasmic membrane). In the

Fig. 3.7: Pneumococci negatively stained with India ink to Fig. 3.8: The structure of bacterial flagellum
show capsule

Chap-03.indd 16 15-03-2016 11:09:42


Chapter 3: Morphology of Bacteria  | 17
gram-negative bacteria, the basal body has four wall. They originate in the cytoplasmic membrane
rings connected to a central rod (L, P, S and M). and are composed of structural protein subunits
The outer L and P rings are associated with the termed pilins like flagella. Fimbriae are antigenic.
lipopolysaccharide and peptidoglycan layers
Demonstration of fimbriae
respectively. S-ring is located just above the
1. Electron Microscopy.
cytoplasmic membrane and the inner M-ring
2. Hemagglutination: Most fimbriate bacteria
contacts the cytoplasmic membrane.
adhere to guinea pig, fowl, horse and pig-red
Gram-positive bacteria have only two basal
cells very strongly, to human cells moderately
body rings.
strongly, to sheep cells weakly and to ox cells
Arrangement/Types (Fig. 3.9) scarcely at all.
There are four types of flagella arrangement:
Functions of pilli
i. Monotrichous: Single polar flagellum (e.g.
Two classes can be distinguished ordinary
Cholera vibrio)
(common) pili and sex pili.
ii. Amphitrichous: Single flagellum at both
A. Ordinary (common) pili: Fimbriae probably
ends, e.g. Alkaligenes faecalis.
function as organs of adhesions.
iii. Lophotrichous: Tuft of flagella at one or both
B. Sex pili: Sex pili are appear to be involved in
ends, e.g. Spirilla.
the transfer of DNA during conjugation. They
iv. Peritrichous: Flagella surrounding the cell
are found on ‘male’ bacteria.
(e.g. Typhoid bacilli).
Demonstration of flagella B. Cell Interior
Flagella are about 0.02 μm in thickness and,
hence, beyond the resolution limit of the light 1. Cytoplasm
microscope. The following methods are used for The cytoplasm of the bacterial cell is a viscous
its demonstration: watery solution or soft gel, containing a variety
i. Dark ground illumination. of organic or inorganic solutes, and numerous
ii. Special staining methods: In this their ribosomes and polysomes. The cytoplasm of
thickness is increased by mordanting. bacteria differs from that of the higher eukaryotic
iii. Electron microscopy. organisms in not containing an endoplasmic
iv. Indirect methods: Indirect methods by reticulum or membrane-bearing microsomes,
which motility of bacteria can be seen or mitochondria, lysosomes and in showing signs of
demonstrated: internal mobility. The cytoplasm stains uniformly
with basic dyes in young cultures.
a. Microscopically in fluid suspensions (in a
hanging drop or under a coverslip)
b. By spread of bacterial growth as a film over 2. Ribosomes
agar, e.g. swarming growth of Proteus sp. The ribosomes are the location for all bacterial
c. Turbidity spreading through semisolid protein synthesis. Bacterial ribosomes are slightly
agar, e.g. Craigie’s tube method. smaller (10–20 nm) than eukaryotic ribosomes and
they have a sedimentation rate of 70S (S or Svedberg
III. Fimbria or pili unit), being composed of a 30S and 50S subunit.
Structure and synthesis: Many gram-negative
bacteria have short, fine, hair-like surface Types of RNA: Ribosomes are composed of
appendages called fimbriae or pili. They are shorter different proteins associated with three types of
and thinner than flagella and emerge from the cell ribonucleic acid (RNA):
i. Messenger (m) RNA: These molecules are
translated during protein synthesis.
ii. Ribosomal RNA (rRNA)
iii. Transfer RNA (tRNA): Specifically transfers
the genetic information carried in the mRNA
into functional proteins.

3. Mesosomes (Chondroids)
These are convoluted or multilaminated
membranous bodies formed as invaginations of
the plasma membrane into the cytoplasm. These
are of two types:
Fig. 3.9: Arrangement of flagella i. Septal mesosomes; ii. Lateral mesosomes.

www.ebook3000.com
Chap-03.indd 17 15-03-2016 11:09:43
18  |  Section 1: General Bacteriology
Functions of mesosomes: progeny either through conjugation or the agency
i. Compartmenting of DNA; ii. Sites of the of bateriophages.
respiratory enzymes. Functions of plasmids: Plasmids are not essential
for host growth and reproduction they inhibit, but
4. Intracytoplasmic Inclusions may confer on it certain properties such as drug
These are not permanent or essential structures, resistance, and toxigencity which may constitute a
and may be absent under certain conditions of survival advantage.
growth. These bodies are usually sources of storage
of energy. They consist of volutin (polyphosphate), 6. Bacterial Spore
lipid, glycogen, starch or sulfur.
A number of gram-positive bacteria, such as those
of the genera Clostridium and Bacillus can form a
5. Bacterial Nucleus
special resistant dormant structure called an endo­
The genetic material of a bacterial cell is contained spore or, simply, spores. Sporulation in bacteria, is
in a single, long molecule of double-stranded not a method of reproduction but of preservation.
deoxyribonucleic acid (DNA) which can be
extracted in the form of a closed circular thread Sporulation: Spore formation, sporogenesis or
about 1 mm (1000 μm) long. The bacterial sporulation normally commences when growth
chromosome is haploid and replicates by simple ceases due to lack of nutrients, depletion of the
fission instead of by mitosis as in higher cells. nitrogen or carbon source (or both) being the most
significant factor.
Plasmids Stages: It may be divided into several stages
Bacteria may possess extranuclear genetic (Fig. 3.10).
elements in the cytoplasm consisting of DNA ∙∙ Spore septum: In the first observable stage
termed plasmids or episomes (see Chapter 10). of sporulation, a newly replicated bacterial
These can exist and replicte independently of the chromosome and a small portion of cytoplasm
chromosome or may be integrated with it. In either are isolated by an ingrowth of the plasma
case, they normally are inherited or passed on the membrane called a spore septum.

Fig. 3.10: The stages of endospore formation

Chap-03.indd 18 15-03-2016 11:09:43


Chapter 3: Morphology of Bacteria  | 19
∙∙ Forespore: The spore septum becomes a
double-layered membrane that surrounds
the chromosome and cytoplasm. Structure,
entirely enclosed within the original cell, is
called a forespore.
∙∙ Spore coat: The forespore is subsequently
completely encircled by dividing septum as
a dou­ble-layered membrane. The two spore
membranes now engage in active synthesis
of various layers of the spore. The inner layer
becomes the inner mem­brane. Between the
two layers is laid spore cortex and outer layer
is transformed into spore coat which consists
of several layers. In some species from outer
layer also develops exosporium which bears
ridges and folds (Fig. 3.11).
∙∙ Fre e en do sp ore: Finally exosp or ium Fig. 3.11: Bacterial spore (Cross-section)
disintegrates and the endospore is freed.

Properties of Endospores (Fig 3.11)


1. Core; 2. Spore wall; 3. Cortex; 4. Spore coat:
Cortex; 5. Exosporium.

Germination
Germination is the process of conversion of a spore
into vegetative cells under suitable conditions. It
occurs in three stages: activation, initiation and Fig. 3.12: Types of spores. 1. Central, bulging; 2. Central, not
outgrowth. Once activated, a spore will initiate bulging; 3. Subterminal, bulging; 4. Subterminal, not bulging;
ger­mination if the environmental conditions are 5. Terminal, spherical; 6. Terminal, oval
favorable.
ii. Modified Ziehl-Neelsen (ZN) stain­i ng:
Shape and Position of the Spore Spores are slightly acid-fast. Ziehl-Neelsen
The shape and position of the spore and its size staining with 0.25–0.5% sulfuric acid (instead
relative to the parent cell are species’ characteristics. of 20% sulfuric acid as used in conventional
Spores may be central (equatorial), subterminal method) as decoloring agent is used for spore
(close to one end), or terminal (Fig. 3.12). The staining.
appearance may be spherical, ovoid or elongated,
and being narrower that the cell, or broader and Uses of Spores
bulging it. The diameter of spore may be same 1. Importance in food, industrial, and medical
or less than the width of bacteria (Bacillus), or microbiology
may be wider than the bacillary body producing a 2. Sterilization control: For proper sterilization,
distension or bulge in the cell (Clostridium). spores of certain species of bacteria are
e mp l oye d a s i n d i cat o r, e. g. B a cillus
Resistance stearothermophilus which is destroyed at a
These structures are extraordinary resistant to temperature of 121°C for 10–20 minutes. Proper
environ­mental stresses. Spores of all medically sterilization is indicated by the absence of the
important species are destroyed by autoclaving at spores after autoclaving.
120°C for 15 minutes. Endospore heat resistance 3. Research
probably is due to several factors: calcium-
dipicolinate and acid-soluble protein stabilization
PLEOMORPHISM AND INVOLUTION FORM
of DNA, protoplast dehydration, the spore coat,
DNA repair, the greater stability of the cell proteins During growth, bacteria of a single strain may show
in bacteria adapted to growth at high temperatures considerable variation in size and shape. This is
and others. known as pleomorphism and occurs most readily
in certain species. The abnormal cells are generally
Demonstration regarded as degenerate or involution forms.
i. Gram-staining: Spores appear as an unstained Pleomorphism and involution forms are often
refractile body within the cell. due to defective cell wall synthesis. Involution

www.ebook3000.com
Chap-03.indd 19 15-03-2016 11:09:44
20  |  Section 1: General Bacteriology
forms may also develop due to the activity of 3. Write short notes on:
autolytic enzymes. a. Bacterial cell wall
b. Cytoplasmic membrane
c. Plasmids or episomes
L-FORMS OF BACTERIA (CELL-WALL- d. Capsule or bacterial capsule
DEFECTIVE ORGANISMS) e. Bacterial flagellae
f. Bacterial spores or endospores
The first isolation of a naturally occurring L-form g. L-forms of bacteria
took place when Kleineberger-Nobel, studying
Streptobacillus moniliformis in the Lister MULTIPLE CHOICE QUESTIONS (MCQs)
Institute, London, observed abnormal forms of the
bacteria and named them L-forms after the Lister 1. Which of the following is not a distinguishing
Institute, London (hence, the “L”). character­istic of prokaryotic cells?
These are abnormal growth forms that may a. They usually have a single, circular chromo-
arise spontaneously or by the inhibition of cell wall some
b. They lack membrane-enclosed organelles
synthesis in bacteria of normal morphology (in the
c. They have cell walls containing peptidogycan
presence of penicillin or other agents that interfere
d. They lack a plasma membrane
with cell wall synthesis).
2. Peptidoglycan layer of cell wall is thicker in:
L-forms may be unstable. They lack a rigid cell
a. Gram-positive bacteria
wall. They are capable of growing and multiplying b. Gram-negative bacteria
on a suitable nutrient medium unlike protoplasts. c. Fungi
L-forms are difficult to cultivate. Colonies of d. Parasites
L-phase organisms on agar media are small and 3. All the following statements are true for
have a characteristic ‘fried egg’ appearance, rather lipopolysaccharide except:
like mycoplasma, L-forms are nonpathogenic a. It consists of three components: Lipid A, core
to laboratory animals. L-forms in the host may polysaccharide and O polysaccharide.
produce chronic infections and are relatively b. Lipid A functions as an endotoxin.
resistant to antibiotic treatment. They present c. It is an integral part of the cell wall of the gram-
special problems in chemotherapy and explain positive bacteria.
relapses after treatment. d. Polysaccharide represents a major surface
antigen of the bacterial cell.
Key Points 4. A tuft of flagella present at one or both the ends of
™™ Bacteria are unicellular, and most of them multiply
bacterial cell is known as:
by bi­nary fission. They are prokaryotes. a. Monotrichous
™™ The structure of the prokaryotic cell b. Amphitrichous
The bacterial cell consists of (A) cell envelope c. Lophotrichous
outer layer (B) cell interior (i) cell wall and (ii) d. Peritrichous
plasma membrane—beneath cell wall and cellular 5. Which of the following is not true about fimbriae?
appendages—capsule, fimbriae, and flagella, and a. They originate in the cytoplasmic membrane
cell interior (and include cytoplasm, cytoplasmic b. They are composed of protein
inclusions (mesosomes, ribosomes, inclusion
c. They may be used for attachment
granules, vacuoles) and a single circular chromosome
d. They may be used for motility
of deoxyribonucleic acid (DNA).
™™ Gram-positive cell wall: The gram-positive cell wall 6. Which of the following bacteria is cell-wall
contains a relatively thick layer of peptidoglycan. deficient?
Teichoic acids stick out of the peptidoglycan layer. a. Escherichia coli
™™ Gram-negative cell wall: Gram-negative bacteria have b. Streptococcus aureus
a lipopolysaccharide­lipoprotein-phospholipid outer c. Mycoplasma
membrane surrounding a thin peptidoglycan layer. d. Treponema pallidum
™™ Endospores are a dormant stage produced by
7. All of the following are spore-forming bacteria
members of Bacillus and Clostridium for survival
except:
during adverse environmental conditions.
a. Clostridium botulinum
b. Bacillus anthracis
IMPORTANT QUESTIONS c. Bacillus subtilis
d. Vibrio cholerae
1. Describe briefly the anatomy of a bacterial cell.
2. Draw a labelled diagram of a bacterial cell. Write ANSWERS (MCQs)
briefly on the cell wall of bacteria. Ans: 1. d; 2. a; 3. c; 4. c; 5. d; 6. c; 7. d

Chap-03.indd 20 15-03-2016 11:09:44


4
Chapter

Physiology of Bacteria

Learning Objectives

After reading and studying this chapter, you should be ∙∙ D


 efine the atmospheric requirement of microaero­
able to: philic bacteria and capnophilic bacteria
∙∙ Explain generation time of bacteria ∙∙ Explain redox potential.
∙∙ Describe and draw bacterial growth curve

I. Principles of bacterial growth Determination of Viable Counts:


1. Dilution method: In dilution method, the
A. Bacterial Division suspension is diluted to a point beyond which
Bacteria divide by binary fission where individual unit quantities do not yield growth when
cells enlarge and divide to yield two progeny inoculated into suitable liquid media.
of approximately equal size. Nuclear division 2. Plating method: In the plating method,
precedes cell division. The cell division occurs by a appropriate dilutions are inoculated on solid
constrictive or pinching process, or by the ingrowth media by:
of a transverse septum across the cell. The daughter i. Pour-plate method
cells may remain partially attached after division ii. Spread plate method—in which serial
in some species. dilutions are dropped on the surface of
dried plates and colony counts obtained.
Generation Time or Doubling Time
The interval of time between two cell divisions, or B. Bacterial Growth Curve
the time required for a bacterium to give rise to
If a suitable liquid medium is inoculated with
two daughter cells under optimum conditions, is
bacterium and incubated, its growth follows a
known as the generation time or doubling time.
definitive course. Small samples are taken at
Examples: In coliform bacilli and many other regular intervals after inoculation and plotted in
medically important bacteria, it is about 20 relation to time. A plotting of the data will yield a
minutes; in tubercle bacilli, it is about 20 hours and characteristic growth curve (Fig. 4.1).
in lepra bacilli, it is about 20 days.
Phases of bacterial growth curve: The bacterial
Colonies: Bacteria growing on solid media form
growth curve can be divided into four major
colonies. Each colony represents a clone of cells
phases: lag phase, exponential or log (logarithmic)
derived from a single parent cell.
phase, stationary phase and decline phase.
Bacterial Count These phases reflect the physiologic state of the
organisms in the culture at that particular time.
Bacteria in a culture medium or clinical specimen
can be counted by two methods: 1. Lag phase: When microorganisms are introduced
1. Total count: This is total number of bacte­ria into fresh culture medium, usually no immediate
present in a specimen irrespective of whether increase in cell number occurs, and therefore this
they are living or dead. period is called the lag phase. After inoculation,
2. Viable count: This measures only viable there is an increase in cell size at a time when little
(living) cells which are capable of growing and are no cell division is occurring. During this time,
pro­ducing a colony on a suitable medium. however, the cells are not dormant. This initial

www.ebook3000.com
22  |  Section 1: General Bacteriology
of carbon, a source of nitrogen and some inorganic
salts. The water content of bacterial cells can vary
from 75 to 90% of the total weight and is the vehicle
for the entry of all cells and for the elimination of
all waste products. It participates in the metabolic
reactions and also forms an integral part of the
protoplasm.

Categories of Requirements for


Microbial Growth
The requirements for microbial growth can be
divided into two main categories: (A) Chemical
and (B) Physical.
Fig. 4.1: Bacterial growth curve. The viable count shows lag, A. Chemical requirements: Chemical require­
log, stationary and decline phases. In the total count, the ments include sources of carbon, nitro­gen,
phase of decline is not evident sulfur, phosphorus, trace elements, oxygen, and
organic growth factors.
period is the time required for adaptation to the
new environment, during which the necessary
enzymes and metabolic intermediates are built 1. Major Elements (Macroelements or
up in adequate quantities for multiplication to Macronutrients)
proceed. These include carbon, oxygen, hydrogen, nitrogen,
2. Log (logarithmic) or exponential phase: sul­fur, phosphorus, potassium, magnesium,
Following the lag phase, the cells start dividing calcium, and iron. Carbon is the structural back
and their numbers increase exponentially or by bone of living matter; it is needed for all the organic
geometric progression with time. If the logarithm compounds that make up a living cell.
of the viable count is plotted against time, a straight i. Autotrophs: Organisms that can use inorganic
line will be obtained. carbon in the form of carbon dioxide as their
carbon source are called autotrophs (auto
3. Stationary phase: After a varying period of means self ). They are soil are of no medical
exponential growth, cell division stops due to importance.
depletion of nutrients and accumulation of toxic ii. Heterotrophs: Organisms that use organic
products. Eventually growth slows down, and the carbon are called heterotrophs (hetero—
total bacterial cell number reaches a maximum and different; troph—nourishment). They are
stabilizes. The number of progeny cells formed is unable to utilize carbon dioxide as the sole
just enough to replace the number of cells that die. source of carbon and use reduced, preformed
The growth curve becomes horizontal. The viable organic molecules as carbon sources.
count remains stationary as an equilibrium exists
between the dying cells and the newly formed cells.
2. Trace Elements
4. Decline or death phase: The death phase is the Some elements, termed as trace elements or micro­
period when the population decreases due to cell nutrients, are required in very minute quantities
death. Cell death may also be caused by autolysis by all cells. They include cobalt, zinc, copper,
besides nutrient deprivation and build-up of toxic molybdenum and manganese.
wastes.
3. Growth Factors
Batch Culture or Closed System
Some bacteria require certain organic compounds
In the laboratory, bacteria are typically grown in in minute quantities known as growth factors
broth contained in a tube or flask, or on an agar plate. or bacterial vitamins. Growth factors are called
These are consid­ered batch or closed systems. ‘essential’ when growth does not occur in their
Continuous Culture: To maintain cells in a state of absence, or ‘accessory’ when they enhance growth
continuous growth, nutri­ents must be continuously without being absolutely necessary for it.
added and waste products removed. This is called
continuous culture or open system. B. Physical Factors Influencing Microbial
Growth
Bacterial Nutrition 1. Temperature:
The minimum nutritional requirements for growth Optimum temperature: Each bacterial species
and multiplication of bacteria are water, a source has an optimal temperature for growth and
Chapter 4: Physiology of Bacteria  | 23
a temperature range above and below which 6. Light: Darkness provides a favorable condition
growth is blocked. The temperature at which for growth and viability of bacteria. Bacteria
growth occurs best is known as the ‘optimum are sensitive to ultraviolet light and other
temperature’. Bacteria are divided into three radiations.
groups on the basis of temperature ranges 7. Osmotic effect: Tolerance to osmotic variation—
through which they grow: bacteria are more tolerant to osmotic variation.
i. Mesophilic: Bacteria which grow between 8. Mechanical and sonic stresses: In spite of
10°C and 45°C, with optimal growth tough walls of bacteria, they may be ruptured by
between 20 and 40°C. mechanical stress such as grinding or vigorous
Examples: All parasites of warm-blooded shaking with glass beads.
animals are mesophilic.
ii. Psychrophlic: Psychrophilic bacteria BACTERIAL METABOLISM
(cold-loving) are organisms that grow
between 5 and 30°C, optimum at 10 to Metabolism of the substance is defined as the series
20°C. of changes of substance (carbohydrate, protein
They are soil and water saprophytes. or fat) within the bacterial cell from absorption
iii. Thermophilic: Thermophiles (heat-loving) to elimination. Metabolism may be aerobic or
have growth range of 25–80°C, optimum at anerobic and can be divided into two classes of
50–60°C. chemical reactions: those that release energy and
Examples: Some thermophiles (like Bacillus those that require energy.
stearother­m ophiles) form spores that are
exceptionally thermotolerant. Energy Production
2. Oxygen: Based on their O 2 requirements, There are three critical processes of bacterial
prokaryotes can be separated into aerobes and energy production; aerobic respiration, anerobic
anaerobes. respiration and fermentation. There are two
A. Aerobic bacteria: Require oxygen for general aspects of energy production: the concept
growth and may be: of oxidation-reduction and the mechanisms of
i. Obligate aerobes: These have an ATP generation.
absolute or obligate requirement for
oxygen (O2), like the cholera vibrio. Generation of ATP
ii. Facultative anaerobes: These are
Much of the energy released during oxidation-
ordinarily aerobic but can also grow
reduction reactions is trapped within the cell by
in the absence of oxygen, though less
the formation of ATP.
abundantly. Most bacteria of medical
The addition of P (phosphate group) to a
importance are facultative anaerobes.
chemical compound is called phosphorylation.
iii. Microaerophilic organisms: These
grow best at low oxygen tension (~5%), Mechanisms of phosphorylation: There are
e.g. Campylobacter spp. two general mechanisms for ATP production in
B. Anaerobic bacteria: Grow in absence of bacterial cells.
oxygen. i. Substrate-level phosphor ylation: In
Obligate anaerobes: These may even die on substrate-level phosphorylation, high-
exposure to oxygen, e.g. Clostridium tetani. energy phosphate bonds produced by the
3. Carbon dioxide: All bacteria require small central pathways are donated to adenosine
amount of carbon dioxide for growth. Some diphosphate (ADP) to form ATP. The specific
organisms such as Brucella abortus require fermentative pathways used, and, hence,
much higher levels of carbon dioxide (5–10%) end-products produced, vary with dif­ferent
for growth, especially on fresh isolation bacterial species. Fermentation is carried out
(capno­philic bacteria), e.g. pneumococci and by both obligate and facultative anaerobes.
gonococci. ii. Oxidative phosphorylation: In oxidative
4. Moisture and drying: Moisture is very essential phosphorylation, an electron transport
for the growth of the bacteria. However, the system is involved that conducts a series
effect of drying varies in different species. of electron trans­fers from reduced carrier
5. pH: Most pathogenic bacteria grow best at a molecules. The process is known as aerobic
neutral or slightly alkaline pH (7.2–7.6). Some respiration when oxidative phosphorylation
acidophilic bacteria such as lactobacilli grow uses oxygen as the terminal electron ac­ceptor.
under acidic conditions while cholera vibrio Anaerobic respiration refers to processes
grow at high degrees of alkalinity (well above that use final electron acceptors other than
pH 8). oxygen.

www.ebook3000.com
24  |  Section 1: General Bacteriology
Oxidation–Reduction (O–R) Potential 2. What are the heterotrophic bacteria? Discuss the
nutritional and physical requirements for the
(Redox Potential) growth of the bacteria.
Oxidation is the removal of electrons, and reduction 3. Write short notes on:
is the addition of electrons. In other words, each a. Bacterial growth curve/growth phases of bacteria.
time one substance is oxidized, another one is b. Redox potential.
simultaneously reduced. The pairing of these
reactions is called oxidation-reduction or a redox Muliple choice questions (MCQs)
reaction.
The ability of a substance to take up or part 1. Generation time for Mycobacterium tuberculosis
is:
with electrons is known as oxidation-reduction
a. 20 seconds
or redox or Eh-potential.
b. 20 minutes
Toxic derivatives of oxygen: Aerotolerant c. 20 hours
microorganisms may lack catalase but almost have d. 20 days
superoxide dismutase. All strict anerobes lack both 2. Bacteria which can grow at temperature between
enzymes or have them in very low concentrations 20°C and 40°C are known as:
and, therefore, cannot tolerate O2. It is suggested a. Mesophiles
that in the presence of oxygen, hydrogen peroxide b. Psychrophiles
accumulates in the media and inhibits the growth c. Thermophiles
d. None of the above
of anerobes. Another reason might be that anerobes
3. The bacteria which require much higher level of
possess essential enzymes that are active only in the
carbon dioxide for their growth are known as:
reduced state. The enzyme catalase, which splits
a. Microaerophilic bacteria
hydrogen peroxide, is present in aerobic bacteria, b. Capnophilic bacteria
but is absent in the anerobes. c. Aerobic bacteria
Key Points d. Phototrophs
4. Which of the following acidophilic bacteria can
™™ Bacterial growth curve: The bacterial growth curve grow in acidic conditions?
can be divided into four major phases: lag phase,
a. Escherichia coli
exponential or log (logarithmic) phase, stationary
phase, and decline phase. b. Lactobacilli
™™ The requirements for microbial growth: Chemical and c. Pseudomonas aeruginosa
physical. d. Vibrio cholerae
™™ Chemical requirements: All organisms require a 5. The enzyme catalase is present in:
carbon source, nitrogen, and other chemicals a. Aerobic bacteria
required for microbial growth. b. Obligate anerobic bacteria
c. Aerobic and obligate anerobic bacteria
Important Questions d. None of the above
1. Draw a typical bacterial growth curve and describe Answers (MCQs)
it. 1. c; 2. a; 3. b; 4. b; 5. a
5
Chapter

Sterilization and Disinfection

Learning Objectives

After reading and studying this chapter, you should be ∙∙ Describe filtration—uses and types
able to: ∙∙ Discuss types of radiations and their uses
∙∙ Define the following terms—sterilization, dis­ ∙∙ Give the mechanism of action for each type of
infection and antisepsis chemical agent commonly used in antiseptics and
∙∙ Describe various agents used in sterilization disinfectants
∙∙ Describe sterilization by moist heat ∙∙ Describe the following—aldehydes as disinfectants,
∙∙ Describe the different heat methods and their uses of formaldehyde and glutaraldehyde as
respective applications disinfectants
∙∙ Describe the following—pasteurization, tyndaliz­ ∙∙ Explain vapor-phase disinfectants or gaseous
ation or intermittent sterilization or fractional sterilization and discuss the role of ethylene oxide
sterilization, inspissation or serum inspissator, hot in sterilization of disposable items
air oven ∙∙ Describe various tests used for testing of
∙∙ Explain the principle and functioning of autoclave disinfectants.

Definitions of frequently used METHODS OF STERILIZATION AND


terms DISINFECTION (TABLE 5.1)
Sterilization: Sterilization is defined as the process A. Physical agents
by which an article, surface or medium is freed of B. Chemical agents
all living microorganisms either in the vegetative
or spore state.
A. Physical Agents
Disinfection: Disinfection is the killing, inhibition, 1. Sunlight
or removal of microorganisms that may cause the
Sunlight has an appreciable bactericidal activity. Its
disease.
disinfectant action is primarily due to its content
Antiseptics: Antiseptics are chemical agents of ultraviolet rays.
applied to the tissue to prevent infection by killing
or inhibiting pathogen growth; they also reduce the 2. Drying
total microbial population.
Drying in air has a deleterious effect on many
bacteria.
Applications of Sterilization and
Disinfection 3. Heat
1. Aseptic techniques: Used in microbiological Heat is the most reliable and universally applicable
research, the preservation of food and the method of sterilization and, wherever possible,
prevention of the disease. should be methods of choice. Either dry or
2. Sterile apparatus and culture media : moist heat may be applied. Materials that may
Laboratory work with pure cultures requires be damaged by heat can be sterilized at lower
the use of sterile apparatus and culture media. temperature, for longer periods or by repeated
3. The need to avoid infecting the patient. cycles.

www.ebook3000.com
26  |  Section 1: General Bacteriology
Table 5.1  Methods of sterilization and disinfection
A. Physical agents
1. Sunlight
2. Drying
3. Heat
a. Dry heat
b. Moist heat
4. Filtration
5. Radiation
6. Ultrasonic and sonic vibrations
B. Chemical agents
a. Agents that damage the cell membranes
1. Surface-active disinfectants
2. Phenolic compounds
3. Alcohols
b. Agents that damage proteins
  1. Acids and alkalies
  2. Alcohols
c. A
 gents that modify functional groups of proteins and
nucleic acids
1. Heavy metals
2. Oxidizing agents
3. Dyes Fig. 5.1: Hot air oven
4. Alkylating agents

Mechanism of Action heating (Fig. 5.1). It should be fitted with a fan


Dry heat : The lethal effect of dry heat, or to provide forced air circulation throughout
desssication in general, is usually due to protein the oven chamber, a temperature indicator,
denaturation, oxidative damage, and toxic a control thermostat and timer, open mesh
effects of elevated levels of electrolytes. shelving and adequate wall insulation.

Moist heat: It kills microorganisms by coagulation Preparation of Load


and denaturation of their enzymes and structural 1. No overloading
proteins. 2. Articles: There should be thoroughly clean and
dry.
a. Dry Heat Sterilization (Table 5.1) 3. Glassware: These should be perfectly dry
i. Red heat: Inoculating wires, loops and points before being placed in the oven.
of forceps are sterilized by holding them 4. Test tubes and flasks: These should be
almost vertically in a Bunsen flame until red wrapped in paper.
hot. 5. Rubber materials, except silicon rubber, will
ii. Flaming: Scalpel blades, glass slides, mouth not withstand the sterilizing temperature.
of culture tubes and bottles are exposed to a 6. Cotton plugs: These may get charred at 180°C.
flame for a few seconds without heating them 7. Heat-sensitive materials: Dry heat sterilization
to become red hot. is slow and not suitable for heat-sensitive
iii. Incineration: This is an efficient method for materials like many plastic and rubber items.
the sterilization and disposal of contaminated
materials at a high temperature. Such as Sterilizing Cycle
pathological waste materials, surgical i. The sterilization hold time: It is set to 160°C
dressings, contaminated material, animal for 2 hours or 170°C for 1 hour, or 180°C for
carcasses and other clinical waste. 30 minutes.
iv. Hot air oven: Hot air oven is the most widely ii. Cutting instruments such as those used
used method of sterilization by dry heat. in ophthalmic surgery should ideally be
It is used to process materials which can sterilized at 150°C for two hours.
withstand high temperatures for length of iii. Oils, glycerol and dusting powder: The
time needed for sterilization by dry heat, but British Pharmacopoeia recommends a
which are likely to be affected by contact with holding time of one hour at 150°C for oils,
steam. Hot air oven is electrically heated, with glycerol and dusting powder.
Chapter 5: Sterilization and Disinfection  | 27
Cooling: Cooling may take up to several hours and sterile by heating at 80-85°C for half-an-
do not attempt to open the chamber door until hour on three successive days (fractional
the chamber and load have cooled below 80°C. sterilization). This process is called
Glassware is liable to crack if cold air is admitted inspissation and instrument used is called
suddenly while it is still very hot. inspissator.
4. Water bath: Washing or rinsing laundry
Uses of Hot Air Oven or utensils in water bath at 70-80°C for few
It is a method of choice for sterilization of: minutes will kill most nonsporing micro-
1. Glassware such as tubes, flasks, measuring organisms present.
cylinders, all-glass syringes, glass Petri dishes 5. Low temperature steam formaldehyde
and glass pipettes. (LTSF) sterilization: In this method,
2. Metallic instruments such as forceps, scissors steam at subatmospheric pressure at the
and scalpels. temperature of 75°C with formaldehyde
3. Nonaqueous materials and powders, oils and vapor is used. The efficacy of LTSF
greases in sealed containers and swab sticks sterilizers is tested by using Bacillus
packed in test tubes. stearothermophilus as biological control.
B. At temperature of 100°C
Sterilization Controls 1. Boiling: Boiling at 100°C for 10–30 minutes
A. Biological control: An envelope containing a kills all vegetative spores and some bacterial
filter paper strip impregnated with 106 spores spores.
of Bacillus subtilis subsp niger is inserted into 2. Steam at atmospheric pressure at 100°C
suitable packs. No growth of Bacillus subtilis for 90 minutes
subsp niger indicates proper sterilization. This can be provided by the traditional
B. Chemical indicator: A chemical indicator such Koch and Arnold steamer (or by the
as Browne’s tubes No. 3 containing red solution multipurpose autoclave).
is inserted in each load and a color change from Koch and Arnold steamer: Koch and
red to green is observed, which indicates proper Arnold steamer consists of upright metal
sterilization. tank with a removable lid incorporating
C. Thermocouples: These may also be used a chimney. Water is added on the bottom
periodically. and there is a perforated shelf above water
D. Microwave ovens: No reliable sterilization level. Articles to be sterilized are placed on
process using microwaves is presently available. the perforated shelf. Water in the bottom
of the tank is heated by gas or electricity
b. Moist Heat Sterilization (Table 5.1) (Fig. 5.2). They are exposed to steam at
atmospheric pressure for 90 minutes. One
Moist heat is divided into three forms: single exposure to steam for 90 minutes
A. At temperature below 100°C ensures complete sterilization.
B. At a temperature of 100°C 3. Tyndallization: An exposure of steam at
C. At temperature above 100°C 100°C for 20 minutes on three successive
A. At temperature below 100°C: It includes:
1. Pasteurization of milk: Disinfection by
moist heat at temperature below 100°C
is termed pasteurization. Milk can be
pasteurized in two ways. The temperature
is employed either 63°C for 30 minutes
(holder method) or 72°C for 15–20
seconds (flash method) followed by rapid
cooling to 13°C or lower.
All nonspor ing pathogens such as
mycobacteria, brucellae and salmonellae
are destroyed by these processes. Coxiella
burnetii is relatively heat resistant and may
survive the holder method.
2. Vaccine preparatior: Vaccines prepared
from nonsporing bacteria may be inactivated
in a water bath at 60°C for one hour.
3. Inspissation: Media such as Lowenstein-
Jensen and Loeffler’s serum are rendered Fig. 5.2: Steamer

www.ebook3000.com
28  |  Section 1: General Bacteriology
days is called tyndallization or intermittent Principle of Autoclave
sterilization. This is a fractional method of The principle of the autoclave or steam sterilizer is
sterilization. The instrument commonly that water boils when its vapor pressure equals that
used is Koch and Arnold steamer. of the surrounding atmosphere. When pressure
Principle: Vegetative cells and some spores inside a closed vessel increases, the temperature at
are killed during the first heating and that which water boils also increases. Saturated steam
the more resistant spores subsequently has penetrative power and is a better sterilizing
germinate and are killed during either agent than dry heat.
the second or the third heating. Though Steam condenses to water and gives up its latent
generally adequate, this method may heat to that surface when it comes into contact with a
fail with spores of certain anerobes and cooler surface. The energy available from this latent
thermophiles. heat is considerable, e.g. 1600 mL steam at 100°C
Uses: This method is useful in sterilizing and at atmospheric pressure condenses into 1 mL of
heat-sensitive culture media containing water at 100°C and releases 518 calories of heat. The
such materials as carbohydrates, egg or large reduction in volume sucks in more steam to the
serum, which are damaged by higher area and the process continues till the temperature of
temperature of autoclave. that surface is raised to that of the steam. The water
C. At temperature above 100°C of condensation ensures moist conditions for killing
Steam under pressure: Steam above 100°C or of the exposed microorganisms.
saturated steam is a more efficient sterilizing
agent than hot air. Procedure
1. Water: Sufficient water is put in the cylinder.
Autoclave Above this is a perforated shelf on which
Autoclaving is the process of sterilization by articles to be sterilized are placed, and the
saturated steam under high pressure above 100°C. autoclave is heated.
Steam sterilization is carried out in a pressure 2. Lid: The lid is screwed tight with the discharge
chamber called an autoclave (a device somewhat tap open and the safety valve is adjusted to the
like a fancy pressure cooker). required pressure.
3. Air removal: The steam-air mixture is allowed
Various components of autoclave: In its simplest to escape freely till all the air has been displaced.
form, the laboratory autoclave consists of a vertical To know when all the air inside the autoclave
or horizontal cylinder of gunmetal or stainless has escaped the discharge tap is connected with
steel, in a supporting sheet iron case. The lid or one end of a rubber tube and the other end of
door is fastened by screw clamps and made airtight it is placed in water. When the air bubbles stop
by a suitable washer. The autoclave has on its lid coming, it indicates that all the air from inside
or upper side a discharge tap for air and steam, a the autoclave has been removed.
pressure gauge and a safety valve that can be set to 4. The discharge tap is now closed.
blow off at any desired pressure. Heating is done by 5. Holding period: The steam pressure rises
gas or electricity (Fig. 5.3). The domestic pressure inside and when it reaches the desired set level
cooker serves as a miniature autoclave and may (15 psi), the safety valve opens and the excess
be used for sterilizing small articles in clinics and steam escapes. From this point, the holding
similar establishments. period (15 minutes) is calculated.
6. Autoclave cooling: When the holding period
is over, the heater is turned off and the
autoclave allowed to cool till the pressure gauge
indicates that the pressure inside is equal to the
atmospheric pressure.
7. Air entry in the autoclave: The discharge tap
is opened slowly and air is allowed to enter the
autoclave.
8. Removal of articles: The lid is now opened and
the sterilized arti­cles removed.

Precautions
i. Air escape from the chamber : Since
temperature of air-steam mixture is lower than
that of pure steam, the air must be allowed to
Fig. 5.3: A simple autoclave escape from the chamber.
Chapter 5: Sterilization and Disinfection  | 29
ii. Arrangement of the materials: This should been used widely for purification of water for
be done in such a manner which ensures free industrial and drinking purposes. They are of
circulation of steam inside the chamber. two types:
Uses a. Unglazed ceramic filters, e.g. Chamberland,
i. For sterilizing culture media and other and Doulton filters.
laboratory supplies, aqueous solutions, b. Compressed diatomaceous earth filters,
rubber material, dressing materials, gowns, e.g. the Berkefeld and Mandler filters.
dressing, linen, gloves, instruments and ii. Asbestos filters (Seitz filter): They are made
pharmaceutical products. up of a disc of asbestos (magnesium trisilicate).
ii. For all materials that are water-containing, It is supported on a perforated metal disc
permeable or wettable and not liable to be within a metal funnel. It is then fitted on to a
damaged by the process. sterile flask through a silicone rubber bung.
iii. Particularly useful for materials which cannot Sterilized fluid is collected from the flask and
withstand the higher temperature of hot air filter disc is discarded after use. These discs are
oven. available with different grades of porosity.
Examples: Seitz filter, Carlson and Sterimat
Sterilization Controls filters.
A. Biological control (Bacterial spores): iii. Sintered glass filters: They are prepared
An envelope contain­ing a filter paper strip by size grading powdered glass followed by
impregnated with 10 6 spores of Bacillus heating.
stearothermophilus is placed with the iv. Membrane filters: Membrane filters consist
load sterilization is over, the strip is removed of a variety of polymeric materials such
and inoculated into. No growth of B. stea­ as cellulose nitrate, cellulose diacetate,
rothermophilus indicates proper sterilization. polycarbonate and polyester. They are
Spores of this organism withstand 121°C for up manufactured as discs from 13 to 293 mm
to 12 minutes and this has made the organism diameter and with porosities from 0.015 to
ideal for testing autoclaves. 12 mm. They come in a wide range of average
B. Chemical control: A Browne’s tube contai­ning pore diameters (APD), the 0.22 mm size being
red solution changes to green when exposed most widely used for sterilization because the
to temperature of 121°C for 15 minutes in pore size is smaller than that of bacteria.
autoclave. It indicates proper sterilization.
Uses
C. Autoclave tapes
1. They are used routinely in water analysis
D. Thermocouples: These may also be used which
and purification.
record the temperature by a potentiometer.
2. Sterilization and sterility testing.
4. Filtration 3. For preparing sterile solutions for
Filtration is the principal method used in the parenteral use.
laboratory for the sterilization of heat labile 4. Bacterial counts of water:
materials, e.g. sera, solutions of sugars or antibiotics v. Syringe filters: Membrane of different
used for the preparation of culture media. diameters are commonly fitted in syringe-like
holders of stainless steel or polycarbonate.
Uses
For sterilization, the fluid is forced through
1. Heat-sensitive solutions: For sterilization of
the disc (membrane) by pressing the piston
pharmaceuticals, ophthalmic solutions, culture
of the syringe.
media, oils, antibiotics and other heat-sensitive
vi. Vacuum and ‘in-line’ filters: They are
solutions.
suitable for the sterilization or disinfection of
2. For separation of bacteriophages and
large volumes of liquid or air.
bacterial toxins from bacteria.
3. Isolation of organisms which are scanty in vii. Pressure filtration: It may be used for the pro­
fluids. duction of very pure water for laboratory use.
4. Concentration of bacteria from liquids, e.g. viii. Air filters: They are widely used in air
in testing water samples for cholera vibrios or filtration.
typhoid bacilli.
5. For virus isolation. 5. Radiation
Two types of radiations are used:
Types of Filters I. Non-ionizing radiations : Infrared and
i. Earthware filters: These are manufactured in ultraviolet rays are of non-ionizing type.
several different grades of porosity and have The effectiveness of UV light as a lethal and

www.ebook3000.com
30  |  Section 1: General Bacteriology
mutagenic agent is closely correlated with its Factors that determine the potency of dis­
wavelength. The most effective bactericidal infectants
wavelength is in the 240–280 nm range, with the 1. The concentration and stability of the agent
optimum being about 260 nm, the wavelength 2. Nature of the organism
most effectively absorbed by DNA and this 3. Time of action
interfers with DNA replication. 4. pH
Microbial sensitivity to UV radiation 5. Temperature
i. Bacterial spores are generally more resistant 6. The presence of organic (especially protein) or
to UV light than are vegetative cells. other interfering substances
ii. Viruses are also inactivated. 7. Nature of the item to be disinfected.
iii. To disinfect drinking water.
iv. Disinfection of enclosed areas such Categories of Disinfectants
as entryways, hospital wards, operating Disinfection processes have been categorized as
theatres, laboratories and in ventilated high level, intermediate level and low level.
safety cabinets in which dangerous 1. High-level disinfection: High-level disinfections
microorganisms are being handled. can generally approach sterilization in
II. Ionizing radiations: These include X-rays, g effectiveness, whereas spore forms can survive
(gamma) rays and cosmic rays. These have very intermediate-level disinfection, and many
high penetrative power and are highly lethal to mi­crobes can remain viable when exposed to
all cells including bacteria. Ionizing radiations low-level disinfection.
damage the DNA by various mechanisms. High-level disinfectants are used for items
in­volved with invasive procedures that cannot
Applications withstand sterilization procedures (e.g. certain
i. For sterilization in pharmacy and medicine. types of endo­scopes, surgical instruments with
ii. Sterilization of packaged disposable plastic or other components that cannot be
articles such as plastic syringes, intravenous autoclaved).
lines, catheters and gloves that are unable to
withstand heat. Since there is no appreciable Examples:
increase in temperature in this method, it is i. Treatment with moist heat.
known as cold sterilization. ii. Use of liquids such as glutaraldehyde,
iii. Use for antibiotics, hormones, sutures, and hydrogen peroxide, peracetic acid, chlorine
vaccines and to prevent food spoilage. dioxide, and other chlorine compounds.
2. Intermediate-level disinfectants: Intermediate-
B. Chemical Agents level disinfectants are used to clean surfaces
Germicidal chemicals can be used to disinfect and, or instruments in which contamination with
in some cases, sterilize. bacterial spores and other highly resilient
orga­nisms is unlikely. These include flexible
Characteristics of a Disinfectant fi­beroptic endoscopes, laryngoscopes, vaginal
An ideal antiseptic or disinfectant should: specula, anesthesia breathing circuits, and other
∙∙ Have a wide spectrum of activity and must be items. These have been referred to as semi­
effective against a wide variety of infectious critical instruments and devices.
agents (Gram-positive and gram-negative Examples: Alcohols, io­d ophor compounds,
bacteria, acid-fast ; bacteria, bacterial phenolic compounds.
endospores, fungi, and viruses) 3. Low-level disinfectants: Low-level disinfectants
∙∙ Be active at high dilutions and in the presence are used to treat noncritical instruments
of organic matter; and devices such as blood pressure cuffs,
∙∙ Be effective in acid as well as alkaline media; electrocardiogram electrodes and stethoscopes.
∙∙ Have speedy action; They do not penetrate through mucosal surfaces
∙∙ Have high penetrating power; or into sterile tissues, although these items come
∙∙ Be stable; into contact with patients.
∙∙ Be compatible with other antiseptics and
disinfectants; Examples: Quaternary ammonium com­pounds.
∙∙ Not corrode metals;
∙∙ Not cause local irritation or sensitization; Mechanisms of Antimicrobial Action
∙∙ Not interfere with healing; The main modes of action are follows:
∙∙ Not be toxic if absorbed into circulation; A. Agents that damage the cell membrane
∙∙ Be cheap and easily available; 1. Surface active disinfectants
∙∙ Be safe and easy to use. 2. Phenolic compounds
Such an ideal chemical is yet to be found. 3. Alcohols
Chapter 5: Sterilization and Disinfection  | 31
B. Agents that denature proteins active at acid pH and are active against gram-
1. Acids and alkalies positive organisms but are relatively ineffective
2. Alcohols against gram-negative species.
C. Agents that modify functional groups of c. Ampholytic (amphoteric) compounds:
proteins and nucleic acids: Known as ‘Tego’ compounds, these possess
1. Heavy metals and their compounds detergent properties of anionic and antimi­
2. Oxidizing agents—Halogens crobial activity of cationic compounds. They
– Hydrogen peroxide are active over a wide range of pH but organic
3. Dyes—Aniline dyes matter markedly reduces their activity.
– Acridine dyes Uses: They are effective against a wide range
4. Alkylating agents—Aldehydes-formal­ of gram-positive and gram-negative organisms
dehyde, glutaraldehyde and some viruses at a concentration of 1% in
– Ethylene oxide water.
2. Phenols and phenolics
A. Agents that Damage the Cell Membrane Phenol: Phenols are obtained by distillation of coal
1. Surface-active agents tar between temperatures of 170°C and 270°C. It
Substances that alter the energy relationships is now rarely used as an antiseptic or disinfectant
at interfaces, producing a reduction of surface be­cause it irritates the skin and has a disagreeable
or interfacial tension, are referred to as surface- odor. Phenol is bactericidal at a concentration of
active agents or surfactants. They possess both 1%.
hydrophobic (water-repelling) and hydrophilic Phenolics: Derivatives of phenol are called
(water-attracting) groups. phenolics.
Classification: These surfactants are classified Phenol derivatives: Certain phenol derivatives
into anionic, cationic, nonionic and ampholytic like cresol, chlorhexidine, chloroxylenol
(amphoteric). Of these, the cationic and anionic and hexachlorophane are commonly used as
compounds have been the most useful antibacterial antiseptics.
agents. i. Cresols: Cresols, obtained industrially by the
a. Cationic agents: These act on phosphate distillation of coal tar, are emulsified with green
groups of cell membrane phospholipids soap and sold under the trade names of Lysol
and also enter the cell. This leads to loss of and Creolin. ‘White fluids’ such as Lysol are
membrane semi­permeability and leakage from effective but are irritant to the skin. They are
the cell of nitrogen and phosphorus-containing active against a wide range of organisms. They
compounds. The agent which enters the cell are most commonly used for sterilization of
denatures its proteins. Quaternary ammonium infected glasswares, cleaning floors, disinfection
compounds (quats) are the most important of excreta. They are not readily inactivated by
cationic compounds. Examples of quaternary the presence of organic matter.
ammonium compounds include cetrimide ii. Chlorhexidine: Chlorhexidine is a member
(cetavalon), benzal­konium chloride (Zephiran, of the biguanide group with a broad spectrum
a brand name) and cetylpyrimidium chloride of activity. They are more active against
(Cepacol, a brand name). Antimicrobial activity gram-positive than gram-negative bacteria.
is affected greatly by organic matter and by pH, They are biocidal against most vegeta­tive
most active at alkaline pH and acid inactivates bacteria and fungi; mycobacteria are relatively
them. They are inactivated by hard water and resistant; endospores and protozoan cysts
soap. are not affected. The only viruses affected are
Uses certain enveloped (lipo­philic) types.
i. They are primarily active against gram- Uses: Savlon (chlorhexi­dine and cetrimide)
positive, non-sporing bacteria; lethal is widely used in wounds, preoperative
to gram-negative organisms at high disinfection of skin, as bladder irrigant, etc.
concentrations. However, contact with the eyes can cause
ii. They are also fungistatic and active against damage.
viruses with lipid enve­lopes (e.g. herpes iii. Chloroxylenol: It is an active ingredient of
and influenza) and much less against dettol. It is less toxic and less irritant.
nonenveloped viruses (e.g. enteroviruses). iv. H e x a c h l o r o p h a n e : G r a m - p o s i t i v e
b. Anionic agents: These include soap and fatty staphylococci and streptococci, which
acids. Anionic surfactants such as common can cause skin infections in newborns, are
soaps usu­ally have strong detergent but weak particularly susceptible to hexachlorophane.
antimicrobial properties. These agents are most So it is used notably for prophylaxis against

www.ebook3000.com
32  |  Section 1: General Bacteriology
staphylococcal infection in nurseries. However, a. Iodine: Iodine compounds are the
it can cause neurotoxicity (brain damage), most effective halogens available for
especially in infants, and its use is now severely disinfection. It is actively bactericidal, with
restricted. moderate action against spores. It is active
against the tubercle bacteria and viruses.
B. Agents that Denature Proteins Uses
Acids and alkalies: Many aliphatic and aromatic i. Skin disinfectant: Iodine in aqueous and
acids are employed as preservatives, especially alcoholic solution has been used widely
in the food industry, and, to some extent, in as a skin disinfectant. Iodine often has
pharmaceutical and cosmetic products. They are been applied as tincture of iodine, 2% or
not sporicidal. more iodine in a water-ethanol solution of
potassium iodide.
Alcohols ii. Iodophors: Mixtures of iodine with various
Ethyl alcohol (ethanol) and isopropyl alcohol surface-active agents that act as carriers
are the most frequently used. They rapidly kill for iodine are known as iodophors (iodo,
bacteria including tubercle bacilli but they have ‘iodine’; phor ‘carrier’). Povidine-iodine
no action on spores and viruses. However, human (Betadine) for wounds and Wescodyne for
immunodeficiency virus (HIV) is susceptible to skin and laboratory disinfection are some
ethyl alcohol (70%) and isopropyl alcohol (35%) in popular brands.
the absence of organic matter. They must be used at b. Chlorine: In addition to chlorine itself, there
a concentration of 60 to 70% in water to be effective. are three types of chlorine compounds—
They are most frequently used as skin disinfectants hypochlorites and inorganic and organic
and act by denaturing bacterial proteins. chloramines. The disinfectant action
Isopropyl alcohol is preferred. of all chlorine compounds is due to
Methyl alcohol is effective against fungal the liberation of free chlorine. When
spores and is used for treating cabinets and elemental chlorine or hypochlorites are
incubators affected by them. Methyl alcohol added to water, the chlorine reacts with
vapour is toxic and inflammable. water to form hypochlorous acid (HOCL),
which in neutral or acidic solution is a
C. Agents that Modify Functional Groups of strong oxidizing agent and an effective
Proteins and Nucleic Acids disinfectant.
1. Heavy metals: For many years the ions of heavy The activity of chlorine is markedly
metals such as mercury, silver, arsenic, zinc influenced by the presence of organic
and copper were used as germicides. matter.
i. Mercuric chloride: It is very toxic and at Uses: The usual disinfectant for water
present has limited use supplies, swimming pools, dairy product
ii. Silver nitrate: The most commonly and food industries.
employed of the silver salts. c. Hypochlorites: The hypochlorites have
Use a bactericidal, fungicidal, virucidal and
1. Highly bactericidal for the gonococcus. rapidly sporicidal action.
2. Routinely used for the prophylaxis of i. Bleaching powder or hypochlorite
the ophthalmic neonatorum in newborn solution is most widely used for human
infants in a 1% solution. immunodeficiency virus (HIV) infected
3. To prevent infection of burns. material. Hypochlorite solution decays
iii. Copper derivatives are used as algicides, rapidly and should be prepared daily.
fungicides, wood, paint, cellulose and Chloramines are used as antiseptics for
fabric preservation. dressing wounds.
ii. Hydrogen peroxide: It is used to
2. Oxidizing Agents disinfect plastic implants, contact lenses
The most useful antimicrobial agents in this group and surgical prostheses.
are the halogens and hydrogen peroxide. They
inactivate enzymes by converting functional-SH 3. Dyes
groups to the oxidized S-S form. Aniline dyes and acridine dyes are two groups of
i. Halogens: Chlorine and iodine are among dyes which are used extensively as skin and wound
our most useful disinfectants. They are antiseptic. Both are bacteriostatic in high dilution
bactericidal and sporici­dal. They are active but are of low bactericidal activity.
in very high dilutions and their action is very i. Aniline dyes: Of the aniline dyes, derivatives
rapid. of triphenylmethane, especially brilliant
Chapter 5: Sterilization and Disinfection  | 33
green, malachite green and crystal violet Vapor-phase disinfectants
have many uses. 1. Ethylene oxide: This is a colorless liquid
They are highly selective for gram-positive with a boiling point of l0.7°C and is a highly
than against gram-negative organisms and penetrating gas with a sweet ethereal smell. It
have been used in the laboratory in the is highly inflammable and in concentrations
formulation of selective culture media. They in air greater than 3%, highly explosive. It is
have no activity against tubercle bacilli, and, unsuitable for fumigating rooms because of its
hence, the use of malachite green in the explosive property.
Lowenstein-Jenson medium. It is highly lethal to all kinds of microbes
ii. Acridine dyes: They are more active against including spores and tubercle bacilli.
gram-positive organisms than against gram- Uses
negative but are not as selective as the aniline a. Sterilization of articles liable to be
dyes. The more important dyes are proflavine, damaged by heat: It is specially used for
acriflavine, euflavine and aninacrine. They sterilizing heart-lung machines, respirators,
show no significant differences in potency. sutures, dental equipment, books and
clothing.
4. Alkylating Agents
b. Sterilization of a wide range of materials
i. Formaldehyde: Formaldehyde is lethal to
such as glass, metal and paper surfaces,
bacteria and their spores, viruses and fungi.
clothing, plastics, soil, some foods and
It is employed in the liquid and vapor states.
tobacco.
Formaldehyde is commercially available in
Disadvantages of ethylene oxide
aqueous solutions containing 37% formaldehyde
i. It is irritant, and personnel working with it
(formalin) or as paraformaldehyde, a solid
have to take strict precautions.
polymer that contains 91% to 99 % formaldehyde.
ii. Its use as a disinfectant presents a potential
Uses
toxicity to human beings, including
a. Formalin
mutagenicity and carcinogenicity. Baci­
i. Used for preserving fresh tissues for
llus globigi, a red-pigmented variant of
histological examination and is the
B. subtilis, has been used to test ethylene
major component of embalming fluids.
oxide sterilizers.
ii. Used extensively to inactivate viruses in
2. Formaldehyde gas: It is used for fumigation of
the preparation of vaccines.
complex heat-sensitive equipment, including
b. Formaldehyde
anaesthetic machine and baby incubators and
i. Used for preser ving anatomical
for periodic decontamination of laboratory safety
specimens.
cabinets.
ii. Used for destroying anthrax spores in
Fumigation of operation theaters and other
hair.
rooms: This is also widely employed for
iii. Used as an antiseptic mouthwash.
fumigation of operation theaters and other
iv. Used for the disinfection of membranes
rooms (such as isolation rooms). After sealing
in dialysis equipment.
the windows and other outlets, formaldehyde
v. Used as a preservative in hair shampoos.
gas is generated by adding KMnO4 to formalin.
ii. Glutaraldehyde: This has an action similar The reaction produces considerable heat, and
to formaldehyde. It has a broad-spectrum so heat-resistant vessels should be used. After
action against vegetative bacteria including starting generation of formaldehyde vapour, the
mycobacteria, fungi and viruses, but acts more doors should be sealed and left unopened for
slowly against spores. It is more active and 48 hours.
less toxic than formaldehyde. It is used as 2% Formaldehyde has an extremely unpleasant
buffered solution. It can be used for delicate odor and is irritant to mucus membranes.
instruments having lenses. It is available 3. B e ta p r o p i o l a c t o n e ( B P L ) : T h i s i s a
commercially as ‘cidex’. condensation product of ketane and formal­
Uses dehyde with a boiling point of 163°C. It also
i. Cold sterilant: It has been used increasingly destroys microorganisms more readily than
as a cold sterilant for surgical instruments ethylene oxide but does not penetrate materials
and endoscopes such as cystoscopes, well and may be carcinogenic. For sterilization
endoscopes and bronchosope. of biological products, 0.2% BPL is used. It is
ii. Used safely to sterilize corrugated rubber capable of killing all micro-organisms and is
anesthetic tubes and face-masks, plastic very active against viruses.
endotracheal tubes, metal instruments and Use: In the liquid form, it has been used to
polythene tubing. sterilize vaccines and sera.

www.ebook3000.com
34  |  Section 1: General Bacteriology
RECOMMENDED CONCENTRATIONS OF Table 5.3  Various procedures of sterilization/
VARIOUS DISINFECTANTS disinfection of some important materials

The recommended concentrations of various Materials Methods


disinfectants commonly used in the hospitals are 1. Metallic inoculating wires Red heat
given in Table 5.2. Table 5.3 shows various mehods 2. Infective materials like Burning (incineration)
of sterilization/disinfection of some important soiled dressings, bed­
materials. 3. Glasswares-syringes, petri Hot air oven
dishes, test tubes, flasks,
universal containers, oily
Testing of Disinfectants fluids (paraffin)
The following tests are used for testing 4. Metal instruments Autoclaving, hot air
disinfectants: oven, infrared radiation
1. Phenol coefficient test
– Rideal-Walker test 5. Serum, body fluids, Waterbath, at 56°C x 1
– Chick-Martin test bacterial vaccines hour, vaccine bath at x
1 hour
2. Minimum inhibitory concentration (MIC)
6. i. Gloves, aprons, Autoclaving
3. Kelsey-Sykes capacity test
dressings, catheters,
4. In-use test surgical instruments
1. Phenol coeffient test: These tests are irrelevant. except sharp
The test organism is inappropriate and the test instruments
irreproducible. ii. Sharp instruments 5% cresol
i. Rideal-Walker test : The best-known 7. i. Suture materials Autoclaving
disinfectant screening test is the phenol except catgut
ii. Catgut lonizing radiation
coefficient test in which potency of a
8. Milk Pasteurization
disinfectant is compared with that of
phenol. 9. Most of the culture Autoclaving
ii. Chick-Martin test: Chick-Martin test is media
modification of Rideal-Walker test. In this 10. Culture media containing Tyndallization
egg serum or sugar
test, the disinfectant acts in the presence
of organic matter (dried yeast or feces) to 11. Toxin, ascitic fluid, serum, Filtration
sugar and antibi­otic
simulate natural situations. solutions
2. Minimum inhibitory concentration (MIC): This 12 . Rubber, plastic and Gamma radiation,
test measures the lowest concentration of the polythene tubes includ­ing ethylene oxide gas
disinfectant that inhibits the growth of S. typhi in disposable syringes
a nutrient medium. 13. Faeces and urine, vomitus, Bleaching powder,
3. Kelsey-Sykes test (Capacity test): The sputum cresols, formalin
burning, autoclaving
additions are made in increments with or
14. Dispos­able syringes, rubber lonizing radiation
without organic matter and this gives a measure or plastic disposable goods,
of the capacity of the disinfectant to cope with bone and tissue grafts,
successive bacterial invasions. Capacity test adhesive dressings
is designed to simulate the natural conditions 15. Antitoxic sera, serum, urine Merthiolate (1 : 10,00)
16. Operation theatre, wards Formaldehyde gas and
Table 5.2  List and the recommended concentrations and laboratory or floor cresols (Lysol)
of disinfectants commonly used in the hospitals space Sodium hypochlorite
Disinfectant Concentration (1%)
17. Polythene tubing, fabrics, Ethylene oxide
Betadine (Iodophore) 2%
machine
Bleaching powder (calcium 14 g in one liter of water 18. Water Chlorine as
hypochlorite) hypochlorites (0.2%)
Dettol (chloroxylenol) 4% 19. Skin Tincture iodine, spirit
Ethyl alcohol 70% (70% ethanol), savlon
(phenol derivative)
Glutaraldehyde 2%
20. Woollen blankets, wool Formaldehyde gas
Lysol 2.5% and hides
Savlon (chlorhexidine and 2%, 5% 21. Sterilization of operation Formaldehyde gas (50 cc
cetrimide) theatre formalin and 25 g KMnO4
Sodium hypochlorite 1%, 0.1% per 100 cu. ft. space)
Chapter 5: Sterilization and Disinfection  | 35
under which the disinfectants are used in the
Steam under pressure: Autoclaving—Very effective
hospitals. method of sterilization; at about 15 psi of pressure
4. In-use test : The in-use test should only (121°C).
be performed to confirm that the chosen ™™ Filtration: Many types of filters are now available.
disinfectant has been effective under the ™™ Radiation: 1. Ionizing; 2. Nonionizing
conditions and period of use. The liquid ™™ Chemical agents: Although gen­erally less reliable
than heat, these chemicals are suitable for treating
phase of disinfectant solutions is examined large surfaces and many heat-sensitive items.
quantitatively for viable organisms in actual
use hospital practice.
Important questions
Sterilization of Prions 1. Define sterilization and disinfection. Classify the
Prions are infectious proteins without any various agents used in sterilization. Add a note on
detectable nucleic acid. They have properties the principle and functioning of autoclave.
distinct from other infectious agents and are 2. Define the terms sterilization, disinfection and
highly resistant to physical and chemical agents, antisepsis. Name the various agents used for
particularly in their resistance to conventional sterilization and discuss the role of hot air oven in
inactivation methods. They are nonconventional sterilization.
transmissible agents that cause transmissible 3. Write short notes on:
a. Hot air oven
degenerative ence­phalopathies (TDE).
b. Inspissation or serum inspissator
1. Dry heat: Prions are extremely resistant to dry
c. Autoclave
heat. A temperature of 360°C for one hour has
d. Sterilization by radiation and its practical ap-
been reported not to be completely effective. plications.
2. Wet heat: They are more resistant to steam 4. Write briefly about:
sterilization than conventional transmissible a. Sterilization by moist heat
agents (bacteria and their spores, fungi and b. Pasteurisation
viruses). Steam is found to be effective if a c. Tyndallization or intermittent sterilization or
temperature of 134–138°C is maintained for 18 fractional sterilization.
minutes. d. Inspissation or serum inspissator
3. Chemicals: These agents are inactivated by e. Filtration.
sodium hypochlorite (25% available chlorine) 5. Name various types of disinfectants and discuss
treatment at room temperature for one hour. the role of halogens in chemical disinfection.
They are also sensitive to phenol (90%), 6. Write short notes on:
household bleach, ether, acetone, urea (6 a. Vapour-phase disinfectants or gaseous sterili-
mol/L), sodium dodecyl sulfate (10%) and zation.
iodine disinfection. Other chemicals like b. Surface-active disinfectants
c. Quaternary ammonium compounds
aldehydes, potassium permanganate, hydrogen
d. Oxidizing agents
peroxide, ethylene oxide, β-propiolactone,
e. Testing of disinfectants
ethanol, proteases and ionizing radiations have
f. Sterilization of prions
been found to be ineffective.

Key Points Multiple choice questions (MCQs)


™™ Sterilization is the process by which an article, 1. The holder method of pasteurization is not
surface, or medium is freed of all living micro- effective against:
organisms either in the vegetative or spore state. a. Escherichia coli
™™ Disinfection is the killing, inhibition or removal of b. Coxiella burnetii
microorganisms that may cause the disease. c. Staphylococcus aureus
™™ Antisepsis is the prevention of sepsis or putrefaction d. Salmonella typhi
either by killing microorganisms or by preventing 2. The bacterial spore that is most frequently used as
their growth.
indicator of sterilization by hot-air oven is:
™™ Methods of sterilization and disinfection: a. Bacillus subtilis
A. Physical agents: Dry heat: a. Flaming; b. Incineration; b. Clostridium tetani
c. Hot-air Sterilization
c. Bacillus pumilis
™™ Moist heat: Pasteurization: Heat treatment for milk
d. Bacillus globigii
(72°C for about 15 seconds) that kills all pathogens
and most nonpathogens. 3. Which of the following does not kill endospores?
™™ Tyndallization: An exposure of steam at 100°C
a. Autoclaving
for 20 minutes on three successive days is called b. Hot air sterilization
tyndallization or intermittent sterilization c. Pasteurization
d. None of the above

www.ebook3000.com
36  |  Section 1: General Bacteriology
4. Sterilization at 100°C for 20 minutes on three 10. The most widely used disinfectant for human
successive days is known as: immunodeficiency virus (HIV) infected material is:
a. Tyndallization b. Inspissation a. Phenol b. Lysol
c. Pasteurization d. Vaccine bath c. Hypochlorite solution d. Silver nitrate
5. Which bacterial spores are used as sterilization 11. Glutaraldehyde is used as a cold sterilant for
control in autoclave? sterilization of:
a. Bacillus cereus a. Cystoscopes b. Endoscopes
b. Bacillus stearothermophilus c. Bronchosope d. All of the above
c. Clostridium perfingens 12. All the following statements are true for ethylene
d. Pseudomonas aeruginosa oxide except:
6. Autoclave is not useful for sterilization of: a. It diffuses through many types of porous
a. Disposable plastic Petri dishes materials
b. It is used for sterilizing heart-lung machines,
b. Surgical dressings
respirators, books and clothing
c. Metallic instruments
c. It is used for sterilizing glass, metal and paper
d. Liquid paraffin
surfaces, clothing, plastics, some foods
7. Which of following is most effective for sterilizing d. It is suitable for fumigating rooms
mattresses and Petri dishes: 13. Which test is used to simulate the natural conditions
a. Chlorine b. Autoclaving under which the disinfectants are used in the
c. Ethylene oxide d. Glutaraldehyde hospitals?
8. Which of the these disinfectants does not act by a. Kelsey-Sykes capacity test
disrupting the plasma membrane? b. Rideal-Walker test
a. Phenolics b. Ethylene oxide c. In-use test
c. Halogens d. Phenol d. Chick-Martin test
9. Ionizing radiation can be used for sterilization of: Answers (MCQs)
a. Plastic syringes b. Gloves 1. b; 2. a; 3. c; 4. a; 5. b; 6. d; 7. c; 8. c; 9. d; 10. c; 11. d;
c. Catheters d. All of the above 12. d; 13. a
6
Chapter

Culture Media

Learning Objectives

After reading and studying this chapter, you should be ∙∙ Differentiate between the following: enriched
able to: media and enrichment media; indicator media and
∙∙ Describe classification of media differential media; selective media and differential
media with suitable examples.

Introduction 5. Malt extract: It consists mainly of maltose,


starch, dextrins and glucose, and contains
Culture medium: A nutrient material prepared about 5% of proteins and protein breakdown
for the growth of microorganisms in a laboratory products, and a wide range of mineral salts and
is called a culture medium. growth factors.
6. Blood and serum : Thsese are used for
COMMON INGREDIENTS OF CULTURE enriching culture media.
MEDIA
1. Water: Tap water is often suitable for culture Classification of media
media. Media have been classified into many ways
2. Agar: Agar (or agar-agar) is prepared from a (Table 6.1).
variety of seaweeds. The chief component of
agar is a long-chain polysaccharide. It also A. Phases of Growth Media
contains a variety of impurities, including
inorganic salts, a small amount of protein-like Growth media are used in either of the two phases:
material and sometimes traces of long-chain liquid (broth) or solid (agar).
fatty acids.
At the concentrations normally used, most 1. Liquid (Broth) Media
bacteriological agars melt at about 95°C and In broth media, nutrients are dissolved in water,
solidify only when cooled to about 42°C. A and bacterial growth is indicated by a change
concentration of 1–2% usually yields a suitable
gel. New Zealand agar has more jellifying
capac­ity than Japanese agar. Table 6.1  Classification of media
3. Peptone: It is a complex mixture of partially A. Based on phases of C. Special media
digested proteins. The important constituents growth media i. Enriched media
1. Liquid (broth) media ii. Enrichment media
are peptones, proteoses, amino acids, a variety
2. Solid (agar) media iii. Selective media
of inorganic salts, including phosphates, 3. Semisolid media iv. Indicator or dif­
potassium and magnesium, and certain B. Based on nutritional ferential media
accessory growth factors, such as nicotinic acid factors v. Transport media
and riboflavin. 1. Simple media (basal vi. Sugar media
media) D. Reducing media
Commercially available peptones or digest
2. Complex media Based on phases of growth
broth can be used. Meat extract is also available 3. S ynthetic or defined media
commercially and is known as Lab-Lemco. media 1. Liquid (broth) media
4. Yeast extract: It contains a wide range of amino 2. Solid (agar) media
acids, growth factors and inorganic salts. 3. Semisolid media

www.ebook3000.com
38  |  Section 1: General Bacteriology
in broth’s appearance from clear to turbid (i.e. and used as a general purpose media, e.g.
cloudy). peptone water, nutrient broth and nutrient agar
(Tables 6.2 and 6.3).
2. Solid (Agar) Media 2. Complex media: Media that contain some
ingredients of unknown chemical composition
Solid media are made by adding a solidifying
are called complex media. One common
agent to the nutrients and water. Agarose is the
ingredient is peptone. Extracts, which are the
most common solidifying agent. The Petri dish
water-soluble components of a substance, are
containing the agar is referred to as agar.
also used.
3. Semisolid Media Nutrient broth: A commonly used complex
medium, nutrient broth (in liquid form). It is a
For special purposes where agar is added to media simple basal liquid medium, supports growth of
in concentrations that are too low to solidify them. many organisms.

B. Based on Nutritional Factors Types of Nutrient Broth ­


1. Simple media (Basal media): Simple media There are three types of nutrient broth:
are those which contain only basic nutrients 1. Meat infusion broth; 2. Meat extract broth; 3.
required for the growth of ordinary organisms, Digest broth.

Table 6.2  Representative types of liquid media


Medium Composition Characteristics
1. Peptone water Peptone—10 g i. Basis for carbohydrate fermentation media
Sodium chloride (NaCl)- 5 g ii. For testing the formation of indole
Water—1 liter
(pH 7.4–7.5)
2. Nutrient broth Peptone water i. For routine culture
Meat extract ii. As a base to prepare many other culture media
iii. To study growth curve
3. Glucose broth Nutrient broth + Glucose Blood culture
(1% most common) Promotes luxuriant growth of many organisms
Glucose acts as a reducing agent
4. Enichment media
i. Tertrathinate Nutrient broth, Enriches salmonellae and sometimes shigellae
broth Sodium thiosulfate,
Calcium carbonate,
Iodine solution
Phenol red
ii. Selenite F broth Sodium selenite It inhibits coliform bacilli while permitting salmonellae and
Peptone many shigellae to grow
Lactose
iii. Alkaline Peptone—10 g Excellent medium for enriching the number of V. cholerae and
peptone water Sodium chloride other vibrio species in a fecal specimen
(NaCl)—10 g
Distilled water—1 liter
5. Anerobic media
i. Thioglycollate Yeast extract, Supports growth of anaerobes, aerobes, microaerophilic and
broth Casein hydrolysate, fastidious microorganisms
Pancreatic digest
Glucose,
L-cysteine,
Agar,
Sodium chloride, sodium
thioglycollate,
Resazurin sodium solution
Water
ii. Robertson’s Nutrient broth, Culture of anaerobic bacteria
cooked meat Predigested cooked meat Preservaton of stock culture of aerobic bacteria
broth (RCM) of ox heart
Chapter 6: Culture Media  | 39
Table 6.3  Representative types of agar media
Medium Composition Characteristics
A. Simple medium
Nutrient agar Nutrient broth agar (2–3%) Complex medium used for routine laboratory work
B. Enriched media
i. Blood agar Nutrient agar. Sheep blood In addition to being enriched medium, it is an indicator medium
(5–10%) showing the hemolytic properties of bacteria
ii. Chocolate agar Heated blood agar This medium is used to culture fastidious bacteria, such as Hemophilus
(55°C × 2 hours) influenzae, the neisseriae and Pneumococcus
iii. Loeffler’s Nutrient broth Culture of Corynebacterim diphtheriae
serum slope Serum (of ox, sheep or
(LSS) horse)
Glucose
C. Indicator media
MacConkey agar Peptone Isolation and differentiation of lactose fermenting (LF) and nonlactose
Sodium taurocholate, fermenting (NLF) enteric bacilli. It is selective and differential
Agar
Neutral red
Lactose
D. Selective media
i. Deoxycholate Nutrient agar Suitable for the isolation of Salmonella and Shigella
citrate agar Sodium deoxycholate,
(DCA) Sodium citrate,
Lactose,
Neutral red
ii. Bile salt agar Nutrient agar, sodium Culture of Vibrio cholerae
(BSA) taurocholate (0.5%) pH 8.2

Nutrient Agar (Table 6.3 and Fig. 6.1)


Nutrient agar is prepared by adding agar at a
concentration of 2% to the nutrient broth. It
is simplest and most common medium used
routinely in microbiology laboratories, to grow
nonfastidious bacteria nutrient agar is commonly
referred to as “agar medium”.
Semisolid agar: If the concentration of agar is
reduced to 0.2–0.5%, semisolid or sloppy agar
is obtained which enables motile organisms to
spread but not non-motile bacteria.
Firm agar: If the concentration of agar is increased Fig. 6.1: Nutrient agar
to 6%, it is called firm agar.

Examples of Enriched Media


3. Synthetic or Chemically Defined Media
1. Blood agar (Fig. 6.2): Many medically important
They are prepared exclusively from pure chemical bacteria are fastidious, requir­ing a medium that
substances and their exact composition is known. is even richer than nutrient agar commonly
used in clinical laboratories is blood agar.
C. Special Media Blood agar is used for isolation of streptococci,
pneumococci, Haemophilus
i. Enriched Media (Table 6.2)
2. Chocolate agar (Fig. 6.3): A medium used
These are prepared to meet the nutritional to culture even more fastidious bacteria is
requirements of more exacting bacteria by the chocolate agar, (heated blood agar). It is used
addition of substances such as blood, serum or egg for isolation of Neisseria (meningococci and
to a basal medium. gonococci) and Haemophilus.

www.ebook3000.com
40  |  Section 1: General Bacteriology

Fig. 6.2: Blood agar Fig. 6.3: Chocolate agar

3. Loeffeler’s serum slope (Fig. 6.4): Serum is


added for enriching the medium. It is used for
the isolation of Corynebacterium diphtheriae.

ii. Enrichment Media (Table 6.2)


When a substance is added to a liquid medium
which inhibits the growth of unwanted bacteria
and favors the growth of wanted bacteria, it is
known as enrichment media. This medium for an
enrichment culture is usually liquid and provides
nutrients and envi­ronmental conditions that favor
the growth of a particu­lar microbe but not others.
In mixed cultures or in materials containing Fig. 6.4: Loeffler’s serum slope (LSS)
more than one bacterium, the bacterium to be
isolated is often overgrown by the unwanted bacte­
ria. Usually, the nonpathogenic or commensal
bacteria tend to overgrow the pathogenic ones; for
example, S. Typhi being overgrown by Escherichia
coli in cultures from feces.
Examples:
a. Tetrathionate broth: Tetrathionate inhibits
coliforms while allowing typhoid-paraty­phoid
bacilli to grow freely in fecal sample.
b. Selenite F (F for feces) broth: It is used for
dysentery bacilli.
c. Alkaline peptone water: It is used for Vibrio
cholerae from faeces.

iii. Selective Media (Table 6.3)


Fig. 6.5: Lowenstein-Jensen medium
When a substance is added to a solid medium
which inhibits the growth of unwanted bacteria but
favors the growth of wanted bacteria, it is known as
selective media. These media are used to isolate cholerae and inhibits the growth of intestinal
particular bacteria from specimens where mixed organisms.
bacterial flora is expected.
Examples of selective media
iv. Indicator Media
a. Deoxycholate citrate agar (DCA): Addition
of deoxycholate acts as a selective agent for These media contain an indicator which changes
dysentery bacilli (isolation of Shigellae). colour when a bacterium grows in them.
b. Lowenstein-Jensen medium (Fig. 6.5): This Examples:
medium is used for Mycobacterium tuberculosis. a. Wils on an d Blair m e d ium : Th e re i s
c. Bile salt agar (BSA): Bile salt is a selective incorporation of sulfite in Wilson and Blair
agent. It faredurs the growth of only Vibrio medium. S. Typhi reduces sulfite to sulfide
Chapter 6: Culture Media  | 41
in the presence of glucose and the colonies of
S. Typhi have a black metallic sheen.
b. MacConkey agar: MacConkey agar indicates
lactose fermenting property. Lactose fermenter
(LF) produces pink colonies and non-lactose
fermenter (NLF) produces colorless colonies
due to a neutral red indicator.

v. Differential Media
A medium, which has substances incorporated in
it, enabling it to bring out differing characteristics
of bacteria and thus helping to distin­guish between
them, is called a differential medium. Fig. 6.6: MacConkey agar
Example
MacConkey agar (Fig. 6.6): MacConkey agar is
both differential and selective. It contains peptone,
meat extract, NaCl, bile salt, lactose and neutral
red indicator.
Lactose fermenters (LF) form pink or red color
colonies and nonlactose fermenters (NLF) form
colorless or pale colonies.

vi. Sugar Media


For the identification of most of the organisms,
sugar fermentation reactions are carried
out. Carbohydrate fermentation is used ‘for
characterisation and identification of bacteria,
particularly important in the study of Gram-
negative bacilli. Sugar media are used to test
fermentation.
Sugar used for sugar media: The term ‘sugar’ in
microbiology denotes any fermentable substance.
Glucose, lactose, sucrose and mannitol are Fig. 6.7: Sugar media
routinely employed for fermentation tests.
Usual sugar media: The usual sugar media consist
of 1% of the sugar in peptone water along with an specimen to the laboratory or the normal flora may
appropriate indicator (Anrade’s indicator—0.005% over­grow pathogenic flora, such special media are
acid fuchsin in NaOH). A small tube (Durham’s devised to maintain the viability of the pathogen
tube) is kept inverted in the sugar tube to detect gas termed as ‘transport media’.
production (Fig. 6.7). The color of the medium is
light yellow. The test bacterium is inoculated and Examples
incubated. Acid production is indicated by the i. Stuart’s transport medium and Amies
development of pink color. Gas accumulates in the transport medium for gonococci.
inner Durham’s tube. ii. Buffered glycerol saline for enteric bacilli.
Hiss serum sugars: Hiss serum sugars are
used for organisms which are exacting in their Anaerobic Media (Table 6.2)
growth requirements (fastidious organisms) like These media are used to grow anaerobic organisms,
streptococci, pneumococci. and contain reducing substances. These include:
i. Thioglycollate broth; ii. Cooked meat broth.
vii. Transport Media
A transport medium is a holding medium designed i. Thioglycollate Broth
to preserve the viability of microorganisms in the Thioglycollate broth contains reducing agents,
specimen but not allow multiplication. such as sodium thioglycollate, glucose, vitarnin C
Delicate organisms (like gonococci) which (ascorbic acid), cysteine and agar (concentration
may not survive the time taken for transporting the of 0.05%), with methylene blue. Thioglycollate

www.ebook3000.com
42  |  Section 1: General Bacteriology
acts as a reducing agent and creates an anaerobic
™™ Selective media: By inhibiting unwanted organisms
environment deeper in the tube, and allows selective media allow growth of only the desired
anaerobic bacteria to grow. Glucose and microbes, e.g. deoxycholate citrate agar (DCA);
thioglycollate maintain the anerobic condition. Lowenstein Jensen medium (L.J medium)
Agar (0.05%) prevents convection currents of air. ™™ Differential media or indicator media distinguish one
Methylene blue or resazurin act as an oxidation- microorganism from one another growing on the
same media, e.g. MacConkey medium
reduction potential indicator, which should show
™™ Sugar media usually consist of 1% sugar in peptone
that the me­dium is anerobic except in the surface water along with an appropriate indicator
layer in addition to a reducing agent and semi­ ™™ Transport media are used to maintain the viability of
solid agar. certain delicate organisms during their transport to
the laboratory e.g. Stuart’s transport medium
™™ Anaerobic media: Reducing media chemically
ii. Cooked Meat Broth or RCM Broth
remove molecular oxygen that might interfere with
(Robertson’s Cooked Meat Broth) for more the growth of anaerobes.
details, refer to Chapter 7
Important questions
Media to Test Special Properties
1. Distinguish between a selective medium and a
Various media to test special properties like ure­ase differential medium.
production, and composite media for simultane­ 2. Write short notes on:
ous demonstration of different features have been a. Enriched media
devised. They are dealt with in the appropriate b. Enrichment media
chapters. c. Indicator media
d. Differential media
f. Transport media
Process of Media Making g. Anaerobic media
It is essential to monitor the quality of culture h. Cooked meat broth (CMB) or RCM broth.
media at all stages in their preparation and use.
Culture media used to be prepared in laboratories Multiple choice questions (MCQs)
themselves, starting with basic ingredients. Not only
was this laborious, but it also led to considerable 1. The important source of nutrition for bacteria to
batch variation in the quality of media. The process grow is:
a. Agar b. Electrolytes
of media making has become simpler and its
c. Inorganic salts d. Peptone
quality more uniform with the ready avail­ability of
2. All the following are examples of enriched media
commercial dehydrated culture media. except:
Key Points a. Blood agar b. Chocolate agar
c. Loeffler’s serum slope d. Bile salt agar
™™ A culture medium is any material prepared for the
growth of bacteria in a laboratory 3. All the following are examples of selective media
™™ Agar is a common solidifying agent for a culture except:
medium a. Potassium tellur­ite medium
™™ Simple media include the nutrient broth and peptone b. Deoxycholate citrate agar
water, which form the basis of other media (e.g., c. Lowenstein-Jensen medium
nutrient broth, nutrient agar, etc.) d. Nutrient agar
™™ Enriched media are solid media supplemented with
4. Which enrichment medium is preferred to grow
blood, serum, etc. (e.g. blood agar, chocolate agar,
Vibrio cholerae?
Loffler’s serum slope, Lowenstein-Jensen medium)
™™ Enrichment media: An enrichment culture is used a. Tetrathionate broth b. Selenite F broth
to encourage the growth of a particular micro- c. Alkaline peptone water d. All of the above
organism in a mixed culture and are the liquid media Answers (MCQs)
(e.g. selenite F broth or tetrathionate broth)
1. d; 2. d; 3. d; 4. c.
7
Chapter

Culture Methods

Learning Objectives

After reading and studying this chapter, you should be ∙∙ Explain the principle and describe uses of the
able to: following: Mclntosh and Fildes anaerobic jar;
∙∙ Discuss anaerobic culture methods cooked meat broth (CMB).

Introduction
In the clinical laboratory, the indications for
culture are mainly to:
1. Isolate bacteria in pure culture
2. Demonstrate their properties
3. Obtain sufficient growth for preparation of
antigens and for other tests.
4. Type isolates
5. Determine sensitivity to antibiotics
6. Estimate viable counts
7. Maintain stock cultures.

Methods of bacterial culture


The methods of bacterial culture used in the clini­
cal laboratory include streak culture, lawn culture, Fig. 7.1: Streak culture (streak plating) on solid media
stroke culture, stab culture, pour-plate culture, shake
culture and liquid culture.
and cool it on unseeded medium, between each
1. Streak Culture (Surface Plating) sequence. At each step, the inoculum is derived
This method is routinely employed for the isolation from the most distal part of the immediately
of bacteria in pure culture from clinical specimens. preceding strokes.
A platinum loop, No. 23 SWG, 6.5 cm long, is Plates are incubated in the inverted position
charged with the specimen to be cultured. Owing with the lid underneath. On incu­bation, growth
to the high cost of platinum loops for routine work may be confluent at the site of original inoculation
are made of nichrome resistance wire, No. 24 SWG. (well), but becomes progressively thinner, and
The loop is flat, circular and completely closed with well-separated colonies are obtained over the final
2–4 mm internal diameter mounted on a handle. series of streaks.
One loopful of the specimen is smeared
thoroughly over area A (Fig. 7.1), on the surface of 2. Lawn Culture or Carpet Culture
a well-dried plate, to give a well-inoculum or ‘well’. Lawn cultures are prepared by flooding the surface
The loop is resterilized and drawn from the well in of the plate with a liquid culture or suspension of
two or three parallel lines on to the fresh surface of the bacterium, pipetting off the excess inoculum
the medium (B). This process is repeated as shown and incubating the plate. Alternatively, the surface
(C, D, E), care being taken to sterilize the loop, of the plate may be inoculated by applying a swab

www.ebook3000.com
44  |  Section 1: General Bacteriology
soaked in the bacterial culture or suspension. 6. Liquid Culture
After incubation, lawn culture provides a uniform Liquid cultures in tubes, bottles or flasks may be
growth of the bacterium. inoculated by touching with a charged loop or by
add­ing the inoculum with pipettes or syringes.
Uses
i. Antibiotic susceptibility testing Uses
ii. Bacteriophage typing i. Blood culture and for sterility.
iii. For preparation of bacterial antigens and ii. Dilution in the medium: For inocula containing
vaccines—when a large amount of growth is antibiotics and other inhibitory substances.
required on solid media. iii. Large yields.

3. Stroke Culture Disadvantages


Stroke culture is made in tubes containing agar i. It does not provide a pure culture from mixed
slope or slant. Slopes are seeded by lightly smear­ing inocula—the major disadvantage.
the surface of agar with loop in a zig-zag pattern, ii. Identification of bacteria is not possible.
taking care not to cut the agar.
Aerobic culture
It is employed for providing pure growth of
the bacterium for slide agglutination and other For cultivation of aerobes, incubation is done in an
diagnostic tests. incubator under normal atmospheric conditions.
Incubation of cultures at 37°C is standard practice
4. Stab Culture in the culture of bacteria pathogenic to man.
For the preparation of stab cultures, a suita­ble
medium, such as nutrient gelatin or glucose agar
Anaerobic Culture
is punctured with a long, straight, charged wire Anaerobic bacteria require incubation with­
into the center of the medium and withdrawing it out oxygen and differ in their requirement and
in the same line. sensitivity to oxygen. Obligate anaerobes will not
grow from small inocula unless oxygen is absent
Uses and the Eh of the medium is low.
i. Mainly for the demonstration of gelatin
liquefaction. Methods of Anaerobiosis
ii. For demonstration of oxygen requirement of Anaerobiosis can be achieved by a number of
the bacterium under study. methods such as:
iii. For the maintenance of stock cultures. A. Production of vacuum
iv. For studying the motility of bacteria in B. Displacement of oxygen by other gases
semisolid agar. C. Absorption of oxygen by chemical or biological
methods.
5. Pour-plate Culture D. Displacement and combustion of oxygen
E. By reducing agents
A measured amount of the suspension is mixed
with molten agar medium in a Petri dish. Either F. Other anaerobic culture systems.
1.0 mL or 0.1 mL of dilutions of the bacterial
suspension is introduced into a Petri dish. The
A. Production of Vacuum
nutrient medium, in which the agar is kept liquid This was attempted by incubating cultures in a
by holding it in a water bath at 45–50°C, is poured vacuum desiccator, but proved to be unsatisfactory
over the sample, which is then mixed into the because some oxygen is always left behind. This
medium by gentle agitation of the plate. When the method is not in use now.
agar solidifies, the plate is incubated inverted at
37°C for 48 hours. After incubation, colonies will B. Displacement of Oxygen by Other
grow within the nutrient agar as well as on the Gases
surface of the agar plate and can be enumerated Displacement of oxygen with gases, such as hydro­
using colony counters. gen, nitrogen, helium or carbon dioxide is some­times
employed.
Uses
i. To estimate the viable bacterial count in a Candle Jar
suspension. Candle jar is a popular, but ineffective method.
ii. For quantitative urine cultures. Here inoculated plates are placed inside a large
Chapter 7: Culture Methods  | 45
airtight container and a lighted candle kept in it D. Displacement and Combustion of
before the lid is sealed. Although it is expected that Oxygen
the burning candle will use all the available oxygen
inside before it gets extin­guished, but in practice Anaerobic Jars
some amount of oxygen is always left behind. Anaerobic jars provide the method of choice when
an oxygen-free or anaerobic atmosphere is required
C. Absorption of Oxygen by Chemical or for obtaining surface growths of anaerobes.
Biological Methods McIntosh and Fildes’ anaerobic jar: Anaerobiosis
i. Chemical methods obtained by McIntosh and Fildes’ anaerobic jar
(Fig. 7.2) is the most dependable and widely used
a. Pyrogallic acid
method.
b. Mixture of powdered chromium and
The jar (c. 20 × 12.5 cm) should be made
sulfuric acid
of metal or robust plastic with a lid that can be
c. Gaspak clamped down on a gasket to make it airtight. The
ii. Biological methods lid is furnished with two tubes along with valves,
one acting as gas inlet and the other as outlet. The
i. Chemical Methods lid also has two terminals which can be connected
to an electrical supply. On its undersurface, it
a. Pyrogallic acid carries a gauze sachet carrying alumina pellets
Buchner (1888) first introduced alkaline pyrogallol coated with palladium (palladinized alumina).
for anaerobiosis which absorbs oxygen. In a large It acts as a room tempe­rature catalyst for the
tube containing solu­tion of sodium hydroxide, conversion of hydrogen and oxygen into water.
pyrogallic acid is added and the tube is then placed
inside an air-tight jar and provides anaerobiosis
Procedure
b. Mixture of powdered chromium and sulfuric acid
A mixture of chromium and sulfuric acid can Inoculated culture plates are placed inside the jar
also be used for producing anaerobiosis. The two with the medium uppermost and lid downwards
chemicals react in the presence of available oxygen and the lid clamped tight. The outlet tube is con­
and produce chromous sulfate. nected to a vacuum pump and the air inside is eva­
cuated. Approximately 6/7th of the air is evacuated
c. Gaspak (pressure reduced to 100 mm Hg, i.e. 660 mm below
It is commercially available in the form of a atmospheric pressure) and this is monitored on a
disposable packet of aluminum foil containing vacuum gauge. The outlet tap is then closed and
pellets of sodium borohydride and cobalt chloride
and of citric acid and sodium bicarbonate.
After the inoculated plates are kept in the jar,
water is added to the disposable aluminum foil
packet and the packet is immediately put in the
jar and its lid is screwed tight. Reactions then take
place to supply hydrogen and carbon dioxide.
Hydrogen combines with oxy­gen in the presence
of a catalyst present in the undersurface of the lid
of the jar to produce an anaerobic envi­ronment.
The Gaspak is simple and effective, eliminat­ing the
need for drawing vacuum and adding hydrogen.

ii. Biological Methods


With aerobic bacteria, this has been attempted
by incubating aerobic organisms along with
anaerobic bacteria. Two blood agar plates are
taken one is inoculated with aerobic bacteria
(Pseudomonas aeruginosa) and the other with
specimen of anaerobic bacteria. Then these two
plates are placed one over the other and sealed
along the rims and are incubated. Anaerobiosis
produced by such biological methods is slow and
ineffective. Fig. 7.2: Mcintosh and Filde’s anaerobic jar

www.ebook3000.com
46  |  Section 1: General Bacteriology
the inlet tube connected to a hydrogen supply. iii. Sulfhydryl compounds (present in cysteine)
Hydrogen is drawn in rapidly. As soon as this inrush also contribute for a reduced oxidation-
of gas has ceased, the inlet tap is also closed and the reduction (OR) potential.
jar is held on the bench for 10 minutes.
Method
Catalyst Liquid media should be prereduced by holding
After the jar is filled with hydrogen, the electrical in a boiling water bath for 10 minutes to drive off
terminals are connected to a current supply to heat dissolved oxygen, then quickly cooled to 37°C just
the catalyst and if room temperature catalyst is before use. The surface of the CMB medium may
used, heating is not required. The catalyst will help be covered with a layer of sterile liquid paraffin for
to combine hydrogen and residual oxygen to form strict anaerobiosis.
water. The jar is then incubated at 37°C.
Interpretation
Indicator It permits the growth of even strict anaerobes and
An indicator should be employed for verifying the indicates their saccharolytic or proteolytic activities.
anaerobic condition in the jars. Reduced methylene With growth of saccharolytic anaerobes (C. welchi),
blue is generally used as indicator (mixture of color of meat pieces turns red while it becomes
NaOH, methy­lene blue and glucose). It becomes black in case of proteolytic anaerobes (C. tetani).
colorless anaerobically but regains blue color on
expo­sure to oxygen. Uses of CMB
i. CMB is suitable for growing anaerobes in air
Disadvantage ii. Also for the preservation of stock cultures of
Any anaerobic jar system has the major disadvantage aerobic organisms.
that the plates have to be removed from the jar to be The inoculum is introduced deep in the
examined and this, of course, exposes the colonies medium in contact with the meat.
to oxygen, which is especially hazardous to the
anaerobes during their first 48 hours of growth. F. Other Anaerobic Culture Systems
a. Anaerobic cabinets: The advantage of anaerobic
E. By Reducing Agents cabinets is that all of the processing, includ­ing
periodic examination of plates and preparation
Oxygen in culture media can be reduced by various
of subcultures, can be done without exposure
agents and these reducing agents include glucose,
to oxygen.
ascorbic acid, cysteine, sodium mercaptoacetate
or thioglycollate, or the particles of meat in cooked b. Anaerobic bags or pouches: Anaerobic bags are
meat broth. available commercially.
i. Thioglycollate broth: (See Chapter 6: Culture
Methods of isolating pure cultures
Media)
ii. Cooked meat broth (CMB): Cooked meat 1. Surface plating: It is routinely employed in
broth (CMB) original medium known as clinical bacteriology.
‘Robertson’s cooked meat (RCM) medium’) 2. Use of selective, enrichment or indicator media:
has a special place in anaerobic bacteriology. are widely used for the isolation of pathogens
It contains nutrient broth and pieces of fat- from specimens, such as feces, with varied flora.
free minced cooked meat of ox heart. Meat 3. Selective treatment of the specimen before
particles are placed in 30 mL bottles to a depth culture
of about 2.5 cm and covered with about 15 i. Heating at 65°C for 30 minutes or at higher
mL broth. If test tubes are used, the surface temperatures for shorter periods.
medium may be covered with a 1 cm layer of iii. Pretreatment of specimens with appropriate
sterile liquid paraffin, but this is not essential. bactericidal substances: This method is
the standard practice for the isolation of
Principle tubercle bacilli from sputum and other
i. Unsaturated fatty acids present in meat utilize clinical specimens.
oxygen for auto-oxidation, the reaction is 4. Use of selective growth conditions
being catalyzed by hematin in the meat. i. Separation of bacteria with different
ii. Certain redu­cing substances, such as gluta­ temperature optima
thione and cysteine present in meat also ii. Cultivation under aerobic or anaerobic
utilize oxygen. conditions.
Chapter 7: Culture Methods  | 47
5. Separation of motile from nonmotile bacteria 3. Write short notes on:
can be effected using Craigie’s tube. a. Streak culture
6. Animal inoculation: Laboratory animals are b. Lawn culture
highly susceptible to certain organisms; for c. Stroke culture
example, the mouse to the pneumococcus. d. Pour-plate culture
7. Filters: Used for separating viruses from e. Anaerobic culture methods/culture of anaerobic
bacteria. bacteria.
f. McIntosh-Fields’ anaerobic jar
Key Points
g. Cooked meat broth (CMB) or/RCM broth
™™ The methods of bacterial culture used in the clini­cal h. Methods of isolating pure cultures.
laboratory include streak culture, lawn culture, stroke
culture, stab culture, pour-plate culture, shake culture
and liquid culture. Special methods are employed for MulTiple choice questions (MCQs)
culturing anaerobic bacteria.
™™ Streak culture (surface plating): This method is 1. The most useful method for obtaining discrete
routinely employed for the isolation of bacteria in colonies of the bacteria is by:
pure culture from clinical specimens. a. Lawn culture b. Stab culture
™™ Anaerobic culture methods: Anaerobic bacteria c. Pour-plate culture d. Streak culture
require incubation with­out oxygen. 2. The most useful method for obtaining a uniform
™™ Anaerobic jars: Anaerobiosis obtained by Mclntosh
layer of bacterial growth on a solid medium is by:
and Fildes’ anaerobic jar is the most dependable and
widely used method.
a. Lawn culture b. Stab culture
™™ Cooked meat broth (CMB): Original medium known as c. Pour-plate culture d. Streak culture
‘Robertson’s cooked meat (RCM) medium’ has a special 3. All the following are used for anaerobic culture
place in anaerobic bacteriology. except:
a. Robertson’s cooked meat broth
Important questions b. Thioglycollate broth
c. Nutrient broth
1. Discuss in detail anaerobic culture methods. d. McIntosh and Fildes’ anaerobic jar.
2. Enumerate various methods of bacterial culture.
Describe in detail the anaerobic methods of Answers (MCQs)
cultivation. 1. d; 2. a; 3. c

www.ebook3000.com
8
Chapter

Identification of Bacteria

LEARNING OBJECTIVES

After reading and studying this chapter, you should reduction test; 7. Urease test; 8. Catalase production;
be able to: 9. Oxidase test; 10. Phenylalanine deaminase test;
∙∙ Explain principle and discuss interpretation of 11. Hydrogen sulfide production; 12. Triple-sugar
the following biochemical reactions: 1. Sugar Iron (TSI)
fermentation; 2. Indole production; 3. Methyl ∙∙ List various examples of bacteria giving positive
red (MR) test; 4. Voges-Proskauer (VP) test for bio­chemical reactions for mentioned above.
acetoin production; 5. Citrate utilization; 6. Nitrate

METHODS USED TO IDENTIFY BACTERIA nature of the culture medium, temperature and
time of incubation, age of the culture and the
Once a bacterium has been obtained in pure culture, number of subcultures it has undergone. The
it has to be identified. Identification schemes are characteristics noted are shape, size, side ends,
classified into one of the two categories (Table 8.1): arrangement, and irregular forms, motility, flagella
1. Phenotypic characteristics fimbriae, spores, capsule and staining.
2. Genotypic characteristics.
B. Staining Reactions
1. PHENOTYPIC CHARACTERISTICS
A number of staining techniques for the identifi­
A. Microscopic Morphology cation of bacteria, are available. Of these, Gram
The morphology of the bacterium depends on stain and Ziehl-Neelson stain are most important.
a number of factors, such as the strain studied, Gram stain: The Gram stain divides bacteria into
gram-positive and gram-negative.
Table 8.1  Methods used to identify bacteria
Ziehl-Neelsen staining: With Ziehl-Neelsen stain­
Phenotypic characteristics Genotypic characteristics ing divides bacteria into acid fast and non-acid fast.
A. Microscopic Nucleic acid hybridization Numerous other stains are used for special
morphology PCR-amplifying specific purposes, such as demonstration of flagella,
B. Staining reactions DNA sequences capsule, spores, and metachromatic granules. The
C. Metabolism differences Sequencing rRNA genes
fluorescent antibody technique enables one to
i. Macroscopic
morphology identify them according to their surface antigens.
or cultural
characteristics C. Metabolic Differences
ii. Fermentation and
The requirements of oxygen, the need for carbon
other biochemical
reactions dioxide, the capacity to form pigments, and the
D. Serology production of hemolysis help in classification.
E. Antibiotic tolerance
(resistance) tests, dye i. Cultural Characterstics or Macroscopic
tolerance, and other Morphology
inhibition tests
F. Bacteriophage and These provide additional information for the
bacteriocin typing identification of the bacterium. The characteristics
G. Pathogenicity revealed in different types of media are noted.

Chap-08.indd 48 15-03-2016 11:10:56


Chapter 8: Identification of Bacteria  | 49
While studying colonies on solid media, the various
features are noted (Table 8.2).

ii. Biochemical Reactions


A large number of biochemical tests can be emp­
loyed for the identification of different bacteria.
These include:

1. Sugar Fermentation Fig. 8.1: Different elevation of colonies


This is tested in sugar media.
Principle: To determine the ability of an organism
to ferment a specific carbohydrate (sugar)
incorporated in a medium producing acid or acid
with gas.
Method: Test organism is inoculated in a sugar
medium and incubated at 37°C for 18–24 hours.
Glucose, lactose, sucrose and mannitol are widely
used sugars. Sugar media contain 1% sugar. Indicator
used is Andrade’s indicator (a solution of acid fuchsin
to which is added sodium hydroxide).

Interpretation (Figs 8.3 and 8.4)


Positive-pinkish-red (acidic): Acid production Fig. 8.2: Description of edges of colonies
is shown by change in the color of the medium to
pink or red.
Negative: Yellow to colorless (alkaline).

Table 8.2  Description of appearance of growth on


solid or in liquid media
1. Shape: Circular, irregular, or rhizoid
2. Size: In millimeters
3. Elevation: Effuse, elevated, convex; concave, umbonate
or umbilicate (Fig. 8.1).
4. Margins: Bevelled or otherwise
5. Surface: Smooth, wavy, rough, granular, papillate or
glistening.
6. Edges: Entire, undulate, crenated, fimbriate or curled
(Fig. 8.2).
7. Color: Fluorescent, iridescent, opalescent, self-
luminous.
8. Structure: Opaque, translucent or transparent
9. Consistency: Membranous, friable, butyrous or viscid
10. Emulsifiability: Easy or difficult Fig. 8.3: Inverted Durham’s tube showing gas production
11. Differentiation: Differentiated into a central and a
peripheral portion.
Growth in liquid medium
Degree: None, scanty, moderate, abundant or profuse
Turbidity: Present or absent; if present, slight, moderate or
dense; uniform, granular or flocculent
Deposit: Present or absent: If present, slight, moderate or
abundant; powdery, granular, flocculent, membranous
or viscid; disintegrating completely or incompletely on
shaking
Surface growth: Present or absent; if present, ring growth
around wall of tube; or surface pellicle, which is thin or
thick; with a smooth, granular or rough surface and which
disintegrates completely or incomplelely on shaking. All
aerobes have tendency to grow on surface of media due
to more content of oxygen present on the surface, e.g.
Pseudomonas sp. Fig. 8.4: Sugar fermentation tests

www.ebook3000.com
Chap-08.indd 49 15-03-2016 11:10:58
50  |  Section 1: General Bacteriology
Gas production can be seen as bubbles in
Durham’s tube.

Examples of Fermentative Bacteria


Glucose fermenters: All members of the family
Enterobacteriaceae
Glucose and lactose fermenters: Escherichia coli,
Klebsiella sp.
Glucose and mannitol fermenter: Salmonella sp.

2. Indole Production
Principle: This test demonstrates the ability of
certain bacteria to decompose the amino acid
tryptophane to indole, which accumulates in the
medium. Tryptophan is decomposed by an enzyme
tryptophanase produced by certain bacteria.
Fig. 8.5: Indole test
Method: Indole production is detected by
inoculating the test bacterium into peptone water
(tryptophan rich) and incubating it at 37°C for 48–96
hours. Add 0.5 mL Kovac’s reagent and shake gently.
Tryptophan Tryptophanase
→ Indole
Kovac’s reagent consists of:
∙∙ Paradimethylaminobenzaldehyde 10 g
∙∙ Amyl or isoamyl alcohol 150 mL
∙∙ Concentrated hydrochloric acid 50 mL.

Interpretation (Fig. 8.5)


Indole positive: A red color in the alcohol layer
indicates a positive reaction.
Indole negative: Yel­low colored ring (color of
Kovac’s reagent) near the surface of the medium.
Indole positive: E.coli, Shigella, Edwardsiella,
Proteus sp. other than P. mirabilis.
Fig. 8.6: Methyl red (MR) test
Indole negative: Klebsiella sp. Enterbacter sp.,
Serratia, Hafnia spp., P. mirabilis.
MR Positive and Negative Bacteria
3. Methyl Red (MR) Test MR Positive: E. coli, Shigella spp., Edwardsiella
Principle: The methyl red test is employed to spp., Yersinia sp., Listeria monocytogenes.
detect the pro­duction of sufficient acid during the
MR negative: Klebsiella, Enter­obacter sp., Serratia
fermentation of glucose so that pH of the med­ium
spp., Hafnia spp.
falls, and it is maintained below 4.5.
Method: Inoculate the test organism in liquid 4. Voges-Proskauer (VP) Test for Acetoin
medium (glucose phosphate broth) and incubate
Production
at 37°C for 2–5 days. Then add five drops of 0.04%
solution of methyl red. Mix well and read the result Principle: Many bacteria ferment carbohydrates
immediately. with the production of acetyl methyl carbinol
(acetoin) or its reduction product 2, 3 butylene
Interpretation (Fig. 8.6) glycol. In the presence of potassium hydroxide
Positive: Bright red color. and atmospheric oxygen, acetoin is converted to
diacetyl, and a-naphthol serves as a catalyst to
Negative: Yellow color. form a red complex (Fig. 8.7).
Note: If the results after 48 hours are equivocal, Method: Inoculate test organism in glucose
the test should be repeated with cultures that have phosphate broth and incubate at 37°C or 30°C for 48
been incubated for 5 days. hours only. Add 1 mL of 40% potassium hydroxide

Chap-08.indd 50 15-03-2016 11:11:00


Chapter 8: Identification of Bacteria  | 51

Fig. 8.7: Voges-Proskauer (VP) test Fig. 8.8: Citrate utilization test

and 3 mL of a 5% solution of a-naphthol in absolute Citrate Positive and Negative Bacteria


ethanol. Citrate positive: Klebsiella sp., Enterobacter sp.
Serratia spp, Hafnia spp, Salmonella sp. except S.
Interpretation of VP Test (Fig. 8.7) typhi, Citrobacter sp.
Note: Positive reaction: Development of pink color
in 2–5 minutes, becoming crimson in 30 minutes. Citrate negative: E. coli, Edwar­dsiella, Shigella spp,
Negative reaction: Colorless for 30 minutes. Salmonella typhi.
The tube can be shaken at intervals to ensure
maximum aeration. Naphthol is a carcinogen. 6. Nitrate Reduction Test
Principle: This is a test for the presence of the enzyme
VP Positive and Negative Bacteria nitrate reductase which causes the reduction of nitrate
VP positive: Klebsiella sp., Enterobacter sp., Serratia to nitrite which can be tested for by an appropriate
spp. Enterbacter spp., Eltor vibrios, Staphylococcus. colori­metric reagent. Almost all Enterobacteriaceae
reduce nitrate.
VP negative: Esch. coli, Shigella spp., Edwardsiella,
Micrococcus. Method: Inoculate test organism in 5 mL medium
containing potassium nitrate, peptone and distilled
5. Citrate Utilization water.Incubate it at 37°C for 96 hours. Add 0.1 mL
Principle: This is a test for the ability of an of the test reagent to the test culture which consists
organism to utilize citrate as the sole carbon and of equal volumes of 0.8% sulfanilic acid and 0.5%
energy source for growth and an ammonium salt as a-naphthylamine in 5 N acetic acid mixed just
the sole source of nitrogen with resulting alkalinity. before use.

Method: Koser’s liquid citrate medium or solid


Simmons’ citrate agar may be used. Simmon’s
Interpretation (Fig. 8.9) of Nitrate Reduction
citrate medium contains agar, citrate and Test
bromothymol blue as an indicator. Original color Positive: Red color develops within few minutes
of the medium is green. A part of colony is picked (indicates the presence of nitrite and hence the
up with a straight wire and inoculated into either of ability of the organism to reduce nitrate).
these media. Incubate at 37°C for 96 hours. Negative: No color development.

Interpretation (Fig. 8.8) Nitrate Nitrate


 reductase
→ Nitrite
1. Simmons’ Citrate Medium
Positive: Blue color and streak of growth. Nitrate Reduction Positive and Negative
Negative: Original green color and no growth. Bacteria
2. Koser’s Citrate Medium Nitrate reduction positive: All members of
Positive: Turbidity, i.e. growth. Enterobacteriaceae, Branhamella catarr­halis.
Negative: No turbidity. Nitrate reduction negative: Haemophilus ducreyi.

www.ebook3000.com
Chap-08.indd 51 15-03-2016 11:11:01
52  |  Section 1: General Bacteriology

Fig. 8.9: Nitrate reduction test Fig. 8.10: Urease test

7. Urease Test Method


Principle 1. One ml of hydrogen peroxide solution, H2O2 (10
To determine the ability of an organism to produce vol), is poured over a 24 hours nutrient agar slope
an enzyme urease which splits urea to ammonia. culture of the test organism and the tube is held
Ammonia makes the medium alkaline and thus in a slanting position
phenol red indicator changes to pink/red in color. 2. Alternatively, a small amount of the culture to be
tested is picked from a nutrient agar slope with
a clean sterile platinum loop or a clean, thin
Procedure
glass rod and dip it in a drop of 10% hydrogen
The test organism is inocu­lated on the entire slope peroxide on a clean glass slide.
of Christensen’s medium which contains urea and
phenol red indicator in addition to other constituents Interpretation
including agar. It is incubated at 37°C and examined Positive test: Immediate bubbling, easily observed
after 4 hours and after overnight incubation. (O2 formed).
Christensen’s urease medium contains: Note: A false-positive reaction may be obtained if
∙∙ Peptone water the culture medium contains catalase (e.g. blood
∙∙ Urea (20%) agar), or if an iron wire loop is used.
∙∙ Agar Negative test: No bubbling (no O2 formed).
∙∙ Phenol red.
Positive and Negative Bacteria
Interpretation (Fig. 8.10) of Urease Test
Catalase positive: All members of Entero­
Urease positive: Purple-pink color. bacteriaceae except Shigella dysen­triae type 1;
Urease negative: Pale yellow color (Fig. 8.10). Staphylococcus, Micrococcus, Bacillus.
Catalase negative: Shigella dysentriae type 1,
Urease Positive and Negative Bacteria Streptococcus, Clostridium.
Urease positive: Klebsiella sp., Proteus sp., Yersinia
enterocolitica, Helicobacter pylori. 9. Oxidase Test
Principle
Urease negative: E. coli, Providencia sp., Yersinia
pestis. To determine the presence of bacterial cytochrome
oxidase using the oxida­tion of the substrate, a
8. Catalase Production redox dye, tetramethyl-p-­phenylene-diamine
dihydrochloride (oxidase reagent). The dye is
Principle reduced to a deep purple color.
This demonstrates the presence of catalase, an
enzyme that catalyses the release of oxygen from Method
hydrogen peroxide. 1. Plate method: A freshly prepared 1% solution of
H2 O2 
→ H2 O + O(Air bubbles)
Catalase tetramethyl-p-phenylene-diamine dihydro­
chloride is poured on to the plate so as to cover

Chap-08.indd 52 15-03-2016 11:11:02


Chapter 8: Identification of Bacteria  | 53
the surface, and is then decanted. The colonies 11. Hydrogen Sulfide Production
of oxidase-positive organisms rapidly develop Principle
a purple color.
Some organisms decompose sulfur-containing
2. Dry filter paper method: A strip of filter paper
amino acids producing H2S among the products. It
soaked in the oxidase reagent is removed, laid in
detects H2S liberated as a result of the degradation
a petridish and moistened with distilled water.
of sulfur containing amino acids.
The colony to be tested is picked up with a
platinum loop and smeared over the moist area. Procedure
Interpretation: Positive reaction: Is indicated by
The organisms can be grown in culture tubes.
an intense deep-purple hue, appearing within
Between the cotton plug and the tube insert a
5–10 seconds.
filter paper strip soaked in lead acetate solution
Negative reaction: Absence’ of coloration or by
and dried. Browning of the paper indicates H2S
coloration later than 60 seconds.
production. When cultured in media containing
3. Wet filter paper method: Put a drop of freshly lead acetate or ferric ammonium citrate or ferrous
prepared 1% solution of oxidase reagent on a acetate, they turn them black or brown. This
piece of filter paper. Then rub a few colonies method is more sensitive than lead acetate strip
of test organism on it. method.
Interpretation Interpretation
Oxidase-positive bacteria will produce a deep Positive: Black color.
purple color within 10 seconds.
Negative: No change in color.
Oxidase Positive and Negative Bacteria
Oxidase positive: The test is used for screening Positive and Negative Bacteria
species of Neisseria, Alcaligenes, Aeromonas, H2S positive: Proteus mirabilis, Proteus vulgaris,
Vibrio, Campylobacter and Pseudomonas, which Salmonella sp. with some exceptions.
give positive reactions, H2S negative: Morganella sp., Salmonella Paratyphi
Oxidase negative: Family Enterobacteriaceae—All A, S. choleraesuis.
the members of the family Enterobacteriaceae are
oxidase negative. 12. Potassium Cyanide Test
Principle
10. Phenylalanine Deaminase Test
This tests the ability of an organism to grow in the
Phenylalanine deaminase determines whether presence of cyanide.
the organism possesses the enzyme phenylalanine
deaminase that deaminates phenylalanine to Method
phenylpyruvic acid, which reacts with ferric salts
to give a green color. Inoculate buffered peptone water medium,
containing 1 in 13,000 con­centration of potassium
cyanide, with test organ­ism. Incubate at 37°C for
Procedure
24–48 hours.
Agar slants of the medium containing phenylalanine
is inoculated with a fairly heavy inoculum and Interpretation
incubated at 37°C for overnight. A few drops of
10% ferric chloride solution are added directly to Positive: Turbidity due to growth.
the surface of the agar. If the test is positive, a green Negative: Clear (no growth).
color will develop in the fluid and in the slope.
Positive and Negative Bacteria
Interpretation Positive KCN test: Klebsiella sp., Citrobacter
Positive: Green color. freundii, Pseudomonas aeruginosa.
Negative: No color change. Negative KCN test : Salmonella sp., E. coli,
Alkaligenes faecalis.
Positive and Negative Bacteria
PPA positive: Proteus sp., Morganella sp., 13. Triple-sugar Iron (TSI) Agar
Providencia sp. Principle
PPA negative: All members of the rest of Triple-sugar iron agar (TSI) is used to determine
Enterobacteriaceae. whether a gram-negative rod utilizes glucose

www.ebook3000.com
Chap-08.indd 53 15-03-2016 11:11:02
54  |  Section 1: General Bacteriology
and lactose or su­crose fermentatively and forms the slant acidic (yellow), provided the reaction
hydro­gen sulfide (H2S). TSI medium facilitates is read in 18–24 hours. Consequently, when the
preliminary identification of gram-negative bacilli. tube is examined at the end of 18–24 hours, acid
production from fermentation of lactose is still
Media occurring and both the slant and the deep appear
TSI is a composite medium and contains 10 parts yellow, resulting in an acid-slant-acid d
­ eep reaction.
lactose: 10 parts sucrose: 1 part glucose and Reactions in TSI should not be read beyond 24
peptone. Phenol red and ferrous sulfate serve hours of incubation, because aerobic oxidation
as indicators of acidification and H2S formation, of the fermentation products from lactose and/or
re­spectively. The medium is distributed in tubes, sucrose does pro­ceed and the slant will eventually
with a butt and slant. re­vert to the alkaline state.
H2S producing bacteria: Certain bacteria produce
Procedure
H2S which is a colorless gas. H2S combines with
Medium is inoculated with bacterial culture by a ferric ions (from ferric salts) to form ferous
straight wire pierced deep in the butt (stab culture). sulfide as black precipitate and is manifested by
Incu­bate the tube at 35° C in ambient air for 18–24 blackening of the butt of the medium in the tube
hours.
Glucose-fermenting organism: If the tube is Interpretation (Table. 8.3)
inoculated with a glucose-fermenting organism Red color (alkaline): No fermentation.
that cannot utilize lactose, only a relatively small
Yellow color (acidic): Fermentation of carbo­
quantity of acid can be obtained from the 0.1%
hydrate.
concentration of glucose in the medium. Initially
the entire medium becomes acidic (yellow) in Bubbles in the butt: Gas is also produced during
8–12 hours. However, within the next few hours, fermentation of carbohydrate.
the glucose supply is completely exhausted and
Blackening of the medium:­ H2S production.
the bacteria begin oxidative deg­radation of the
To determine the ability of an organism to
amino acids within the slant portion of the tube
attack specific carbohydrates incorporated in a
where oxygen is present. This results in the release
growth medium, with or without the production
of amines that soon counteract the small quantities
of gas, along with the determination of possible
of acid pres­ent in the slant. The entire slant reverts
hydrogen sulfide (H2S) production.
to an alkaline pH (color returns to red) by 18–24
hours. The deep (anaerobic portion) of the tube
remains acidic (yellow) because amino acid degra­ D. Serology
dation is insufficient to counteract the acid formed. By using specific sera we can identify organisms
by agglutination or other suitable serological
Lactose-fermenting organism: If the tube is
reactions.
inoculated with a lactose-fermenting organism,
then fermentation continues as the organism
is able to use lactose (present in 10 times the E. Antibiotic Tolerance Tests, Dye
concentration of glucose), even though the glucose Tolerance, and Other Inhibition Tests
is completely used up after the first 8–12 hours. These range from disk tests of resistance to
Therefore, when, in addition to glucose, lactose antibiotics, such as penicillin, bacitracin,
and/or su­crose are fermented, the large amount gentamicin, novobiocin and metronidazole, to
of fermentation products formed on the slant will special tests that demonstrate tolerance of dyes
more neutralize the alkaline amines and render and other chemicals.

Table 8.3  Expected results of triple-sugar iron (TSI) agar test


Slant/butt Color Utilization and Interpretation
1. Alkaline slant/alkaline butt (K/K) Red/Red No fermentation of glu­cose, lactose or sucrose.
2. Alkaline slant/acid butt (K/A) Red/Yellow Glucose fermented; lactose (or sucrose for TSI medium) not
fermented.
3.Alkaline slant Acid Red/Yellow Glucose fermented; lactose not fermented, hydrogen sulfide
(Black) Deep (K/ A/H2S) H2S produced.
4.Acid slant/Acid Deep (A/A) Yellow/Yellow Glucose and lactose (or sucrose with TSI) fermented.
K-Alkaline; A-Acidic; H2S-Hydrogen sulfide

Chap-08.indd 54 15-03-2016 11:11:02


Chapter 8: Identification of Bacteria  | 55
F. Bacteriophage and Bacteriocin Typing c. Voges-Proskauer (VP) test
d. Citrate utilization test
These enable intraspecies typing of some bacteria. e. Nitrate reduction test
f. Urease test
G. Pathogenicity g. Catalase test
h. Oxidase test
Pathogenicity tests by inoculation of the test
i. Phenylalanine deaminase test
organism into laboratory animals like the j. Triple-sugar Iron (TSI) agars.
guinea pig, rabbit, rat and mouse were common
procedures for identification of isolates in the past.
The animals may be inoculated by intradermal, MULTIPLE CHOICE QUESTIONS (MCQs)
subcutaneous, intramuscular, intraperitoneal, 1. Catalase test is primarily used to differentiate
intracerebral or intravenous, or by oral or nasal Staphylococcus from:
spray. They are rarely used now because simpler a. Pneumococcus
in vitro tests are available. b. Meningococcus
c. Streptococcus
2. GENOMIC CHARACTERIZATION d. Gonococcus
2. All are oxidase positive bacteria except:
Molecular methods, such as polymerase chain a. Neisseria
reaction and other amplification procedures b. Escherichia coli
coupled with nucleic acid probes carrying specific c. Campylobacter
DNA or RNA base sequences are now widely used d. Pseudomonas aeruginosa
for identifying microbes. 3. Phenylpyruvic acid (PPA)-positive bacteria are the
followings except:
a. Proteus spp.
Characterization of Strain Differences b. Providencia spp.
1. Biochemical typing c. Campylobacter spp.
2. Serological typing d. Morganella spp.
3. Antibiogram 4. All of the following bacteria are urease test positive
4. Phage typing except:
5. Genomic typing a. Klebsiella sp.
i. Pulsed-field gel electrophoresis b. Yerrsinia enterocolitica
c. Helicobacter pylori
ii. Ribotyping.
d. Yersinia pestis
Key Points 5. Which of the following tests detects the production
of acetyl methyl carbinol from pyruvic acid in the
Methods used to identify bacteria
media?
A. Phenotypic characteristics
a. Methyl red test
a.  Microscopic morphology; b. Staining reactions;
c. Metabolism differences; d. Serology; e. Antibiotic b. Voges-Proskauer test
tolerance (resistance) tests, dye tolerance, and other c. Urease test
inhibition tests; f. Bacteriophage and bacteriocin d. Citrate utilization
typing; g. Pathogenicity 6. Triple-sugar iron (TSI) agar medium contains all
B. Genomic characterization: Polymerase chain reaction the following carbohydrates except:
(PCR) and other amplification procedures. a. Glucose
b. Lactose
IMPORTANT QUESTION c. Sucrose
d. Mannitol
Write short notes on:
a. Indole production ANSWERS (MCQs)
b. Methyl red test 1. c; 2. b; 3. c; 4. d; 5. b; 6. d

www.ebook3000.com
Chap-08.indd 55 15-03-2016 11:11:02
9
Chapter

Bacterial Taxonomy

Learning Objectives

After reading and studying this chapter, you should be able to:
∙∙ Discuss bacterial classifications.

Taxonomy basic unit. The species designation gives a formal


taxonomic status to a group of related isolates or
Taxonomy is the science that studies organisms in
strains, which in turn, permits their identification.
order to arrange them into groups; those organisms
with similar properties are grouped together and Kingdoms are divided successively into division,
separated from those that are different. Taxonomy class, order, family, genus and species.
can be viewed as three separate but interrelated areas:
1. Identification: The process of characterizing Example
organisms. The full taxonomical position of is the bacterium
2. Classification: The process of arranging organ­ Escherichia coli as follows:
isms into similar or related groups, primarily to Domain Bacteria
provide easy identification and study. Phylum Proteobacteria
3. Nomenclature: The system of assigning of Class Gammaproteobacteria
names to organisms. Family Enterobacteriaceae
Genus Escherichia
1. Identification
Species Coli
To characterize and identify microorganisms,
a wide assortment of technologies, such as Species Concept in Bacteria
microscopic examination, culture characteristics, Species is the standard taxonomical unit in biology. In
biochemical tests and nucleic acid analysis is used. bacteria, the species concept is vague and ill-defined.
2. classification
Classification systems
Three-domain System
The classification scheme currently favored by most Phenetic System
micro­biologists is the three-domain system. This Organisms can be grouped together based on over­
designates all organ­isms as belonging to one of the all similarity to form a phenetic system. Computers
three domains—Bacteria, Archaea, and Eukarya. may be used to analyze data for the pro­duction
of phenetic classifications. The process is called
Five-kingdom System
numerical taxonomy.
The most widely accepted system was the five-
kingdom system, proposed by RH Whittaker in
1. Adansonian or Numerical Classification
1969 before the three-domain classification system
was intro­duced. The five king­doms in this system The Adansonian or numerical classifi­cation, so
are the Plantae, Animalia, Fungi, Protista (mostly called after Michael Adanson who intro­duced it in
single-celled eukaryotes) and Prokaryotae. the eightenth century. It gives equal weight to all
measurable features, and groups organisms on the
Taxonomic Hierarchies basis of similarities of several characteristics.
Taxonomic classification categories are arranged The development of computers has extended
in a hierarchical order, with the species being the the scope of phenetic classification by permitting
Chapter 9: Bacterial Taxonomy  | 57
comparisons of very large numbers of properties of 3. Nomenclature
several organisms at the same time. This is known Nomenclature, the naming of microorganisms
as numerical taxonomy. Information about the according to established rules and guidelines,
proper­ties of organisms is converted into a form provides the accepted labels by which organisms
suitable for numerical analysis and then compared are universally recognized. Bacteria are given
by means of a computer. names according to an official set of inter­nationally
recognized rules, the International Code for the
2. Phylogenetic Classification Nomenclature of Bacteria.
They can be grouped based on probable
evolutionary relationships to produce a phylo­ Casual or Common Name
genetic system. The hierarchical classifi­cation
represents a branching tree-like arrangement, Two kinds of names are given to bacteria. The first
one characteristic being employed for division is the casual or common name which varies from
at each branch or level. These are systems country to country and is in the local language.
based on evo­lutionary relationships rather than
general resemblance. While there is no “official” Scientific or International Name
classification of prokaryotes, microbiologists The second is the scientific or international name
generally rely on the reference text Bergey,’s Manual which is the same throughout the world. The
of Systematic Bacteriology as a guide. scientific name consists usually of two words, the
first being the name of the genus and the second
3. Molecular or Genetic Classification the specific epithet. In this binomial (“two-name”)
This is based on the degree of genetic relatedness sys­tem of nomenclature, every organism is assigned
of different organisms. This classification is said a genus and species name. The generic name is
to be the most natural or fundamental method usually a Latin noun.
since all properties are ultimately based on the Key Points
genes present. DNA relatedness can be tested by
studying the nucleotide sequences of DNA and ™™ Bacterial taxonomy: It comprises three components:
1. Identification of an unknown isolate with a defined
by DNA hybridization or recombination methods. and named unit; 2. Classification of organism; 3.
The nucleotide base composition and base ratio Nomenclature or naming of the microbial isolates
(Adenine- Thymine:Guanine-Cytosine ratio) varies ™™ Bacterial classifications include Adansonian,
widely among different groups of microorganisms, phylogenetic, and genetic classifications
though it is constant for members of the same ™™ Nomenclature of microorganisms – Nomenclature
species. Molecular classification has been employed refers to the naming of microorganisms
more with viruses than with bacteria.
At present no standard classification of bacteria Important Question
is universally accepted and applied, although
Bergey’s Manual of Determinative Bacteriology is Write briefly about bacterial taxonomy.
widely used as an authoritative source.
Multiple choice questions (MCQs)
4. Intraspecies Classification
1. Phylogenetic classification denotes:
It is often necessary to subclassify bacterial species a. Homology of the DNA base sequences of the
for diagnostic or epidemiological purposes on micro­organisms
the basis of biochemical properties (biotypes), b. Evolutionary arrangement of species
antigenic features (serotypes), bacteriophage c. Equal weight to all features and groups of
susceptibility (phage types) or production of bacteria on the basis of similarities of several
bacteriocins (colicin types). A species may be characteristics
divided first into groups and then into types. d. A type of classification which makes an at­
tempt to subclassify species of bacteria
The application of newer techniques from
2. Which of the following methods may be used in
immunology, biochemistry and genetics has led to bacteriology for epidemiological purposes?
much greater discrimination in intraspecies typing. a. Biotyping b. Serotyping
The methods used are of two types: Phenotypic c. Phage typing d. All of the above
and genotypic. Phenotypic methods include 3. Molecular techniques employed for intraspecies
electrophoretic typing of bacterial proteins and typing of bacteria include:
immunoblotting. Genotypic methods include a. Polymerase chain reaction
plasmid profile analysis, restriction endonuclease b. Southern blotting
analysis of chromosomal DNA with Southern c. Nucleotide sequence analysis
d. All of the above.
blotting, PCR and nucleotide sequence analysis.
Some of these techniques are considered in Answers (MCQs)
Chapter 10 (Bacterial Genetics). 1. b; 2. d; 3. d.

www.ebook3000.com
10
Chapter

Bacterial Genetics

Learning Objectives

After reading and studying this chapter, you should and conjugation in the transfer of genetic material
be able to: from one bacterium to another
∙∙ Explain Lac operon ∙∙ Discuss resistance transfer factor (RTF)
∙∙ Discuss regulation or control of gene expression ∙∙ Differentiate between mutational and plasmid-
mediated drug resistance
∙∙ Discuss structure and functions of the plasmids
∙∙ Discuss transposons
∙∙ Describe mutations ∙∙ Describe principle and clinical applications
∙∙ Discuss various methods of gene transfer of the following: Nucleic acid probes; genetic
∙∙ Differentiate among the mechanisms of trans­ engineering; polymerase chain reaction
formation, transduction, lysogenic conversion ∙∙ Describe gene therapy.

Introduction cytosine (C), guanine (G), and uracil (U). A and G


are double-ring structures called purines, whereas
Genetics: Genetics is the study of genes, their T, C, and U are single ring structures referred to as
structure and function, heredity and variation. pyrimidines.
Genomics: The study and analysis of the nucleotide
ii. A pentose (five-carbon) sugar called
sequence of DNA is called genomics.
deoxyribose or ribose
Genome: The complete set of genetic information
iii. A phosphate group (phosphoric acid):
for a cell is referred to as its genome.

STRUCTURE AND FUNCTIONS OF THE A. Deoxyribonucleic Acid (DNA)


GENETIC MATERIAL Structure
Double helix: According to the model proposed by
Nucleic Acid Structure Watson and Crick, a DNA molecule consists of two
A substance called deoxyribonucleic acid (DNA) is long strands wrapped around each other to form a
the substance of which genes are made. DNA, and double helix (Fig. 10.1). The double helix looks-like
another substance called ribonucleic acid (RNA), a twisted ladder, and each strand is composed of
are together referred to as nucleic acids. many nucleotides. The two strands are held together
Nucleo­tides: Nucleotides are the structural units of by weak hydrogen bonds between the nitrogenous
nucleic acids. Nucleotides are named according to bases of the opposing strands (Fig. 10.1).
their nitrogenous base. Sugar-phosphate backbone: Every strand of DNA
composing the double helix has a “backbone”
Parts of Nucleotide consisting of alternating deoxyribose sugar
Each nucleotide has three parts: and phosphate groups. The deoxyribose of one
i. A nitrogen-containing base nucleotide is joined to the phosphate group of the
next. The nitrogen containing bases make up the
Purines and pyrimidines : The nitrogen- rungs of the ladder. Purine A is always paired with
containing bases are cyclic compounds made up the pyrimidine T and that the purine G is always
of carbon, hydrogen, oxygen, and nitrogen atoms. paired with the pyrimidine C. The bases are held
The bases are named adenine (A), thymine (T), together by hydrogen bonds; A and T are held by
Chapter 10: Bacterial Genetics  | 59
cistron or gene. A large number of genes constitute
a locus and a large number of loci constitute cell
genome. DNA can be compared with a book of
infor­mation. Letters represent nucleotides, words
repre­s ent codons, sentences represent genes,
paragraphs represent loci and entire book as DNA
molecule or cell genome.
A DNA mol­ecule consists of a large number
of genes, each of which contains hundreds
of thousands of nucleotides. The bacterial
chromosome consists of a double-strand­e d
molecule of DNA arranged in a circular form. When
straightened, it is about 1,000 μm in length.
Introns and exons: In higher forms of life, several
stretches of DNA that do not appear to function
as codons occur between the coding sequences
of genes. These apparently useless noncoding
intrusions are called introns, while the stretches
of coded genes are called exons.

B. Ribonucleic Acid (RNA) Structure


Three major kinds of RNA have been identified
in cells. These are re­ferred to as messenger RNA
(mRNA), ribosomal RNA (rRNA), and transfer
Fig.10.1: A schematic drawing of the Watson-Crick structure
RNA (tRNA). Each type of RNA has a specific role
of DNA, showing helical sugar-phosphate backbones of the
two strands held together by hydrogen bonding between in protein synthesis.
the bases
Gene Expression
two hydrogen bonds, and G and C are held by three Gene expression involves two separate but
hydrogen bonds. interrelated process­es transcription and translation.
Base pairing: The characteristic bonding of A to T A. Transcription: Transcription is the process of
and G to C is called base­ pairing and is fundamental synthesizing RNA from a DNA template. The
to the remarkable functionality of DNA. DNA acts as a template for the transcription of
RNA by RNA polymerase for subsequent protein
Complementary: Because the sequence of bases production within the cell. RNA polymerase
of one strand is determined by the sequence attaches itself to the beginning of a gene on
of bases of the other, the bases are said to be DNA and synthesizes mRNA, using one of the
complementary. strands in DNA as a template. This process is
known as transcription. The bases in mRNA will
Code and Codon be complementary to one strand of DNA since
Genetic information is stored in DNA as a code. DNA acts as a template for synthesis of mRNA.
The unit of code is known as codon. It consists of a B. Translation: Translation is the process of
sequence of three bases. Therefore, code is triplet. decoding the information carried on the mRNA
Each codon specifies or codes for a single amino to synthesize the specified protein (Fig. 10.2).
acid, but more than one codon may exist for a
Process of translation: The process of translation
single amino acid. Therefore, code is degenerate.
requires three major components—mRNA,
Thus, the triplet AGA codes for arginine but the
ribosomes, and tRNAs, in addition to various
triplets AGG, CGU, CGC, CGA and CGG also code
accessory proteins.
for the same amino acid. Three codons UAA, UAG
and UGA do not code for any amino acid and are Messenger RNA (mRNA): The mRNA is a temporary
known as nonsense codons. They act as punctuation copy of genetic information. It carries the coded
marks, terminating the mes­sage for synthesis of a information for making specific proteins from DNA
polypeptide. to ribosomes, where proteins are synthesized.
Citron or Gene Ribosomes: Serve as the sites of translation, and
A segment of DNA carrying a number of codons their structure facilitates the joining of one amino
specifying for a particular polypeptide is known as acid to another.

www.ebook3000.com
60  |  Section 1: General Bacteriology

Fig. 10.2: Synthesis of polypeptide

Transfer RNA (tRNA): The tRNA molecule


contains a triplet at one end and amino acid at
the other end. The ribosome moves along the
mRNA until the entire mRNA molecule has been
translated into corresponding sequences of amino
acids. Finally, the sequence of amino acids in
the resulting polypeptide chain determines the
configuration into which the polypeptide chain
folds itself, which in many cases determines the
enzymatic properties of the completed protein.
Central dogma of molecular biology: The flow
of information from DNA to RNA to protein is
often referred to as the central dogma of molecular
biology (DNA→RNA→ polypeptide) was once
believed that information flow pro­ceeded only
in this direction. These processes are illustrated
schematically in Figure 10.3.
Fig. 10.3: The lac operon of Escherichia coli
Extrachromosomal genetic
elements forms, though this distinction is not usually made
Plasmids now. These are not essential for the normal life
and functioning of the host bacterium. They
Most bacteria possess extra­chromosomal genetic
may confer on it properties leading to survival
element in addition to chromosomal DNA
advantage under appropriate conditions such as
elements known as plasmid. It con­sists of a circular
resistance to antibiotics, bacteriocin production
piece of double-stranded DNA, can replicate
toxigenicity, etc.
autonomously (independent replicons) and can
maintain in the cyto­p lasm of a bacterium for Conjuga­tive or nonconjugative plasmids: Such
many generations. Plasmids DNA sometimes plas­mid that contains the information for self-
may be integrated with chromosomal DNA. The transfer to another cell by conjugation is known
name episome was employed for such integrated as conjuga­tive or self-transmissible plasmid. Those
Chapter 10: Bacterial Genetics  | 61
plasmids which do not possess information for
self-transfer to another cell are known as non-
conjugative or nonself-transmissible plasmids.

Genotypic and phenotypic


variations
There are two types of variation:
A. Phenotypic variation
B. Genotypic variation.

A. Phenotypic Variation
The phenotype (‘Phaeno’; display) is the physical
expression of various characters by bacterial cells
in a given environment. These properties are
determined not only by its genome (genotype), Fig. 10.4: Frameshift mutation
but also by its environment. Phenotypic variations
are reversible. Also important, but not part of the operon, is a
distant regulatory gene (in this case is LacI) which
codes for a repres­sor protein. It is a protein molecule
Examples of Environmental Influence on
which can combine with either operator region on
Bacteria
the chromosome or with the inducer (lactose).
1. Synthesis of flagella: The typhoid bacillus is
normally flagellated. But the flagella are not Effect of Lactose on the Control of the Lactose
synthesized when grown in phenol agar and is Operon
reversed when subcultured from phenol agar For transcription to occur as the first stage in
into broth. protein synthesis, RNA polymerase has to attach to
2. Synthesis of enzyme: Another example of DNA at a specific promoter region and transcribe
environmental influence is the synthesis the DNA in a fixed direction. In resting stage when
by Escherichia coli of the enzyme beta- lactose (inducer) is not present in the medium,
g a l a c t o s i d a s e, n e c e s s a r y f o r l a c t o s e repressor molecule is bound to the operator,
fermentation but the actual synthesis takes preventing the passage of RNA polymerase from
place only when it is grown in a medium promoter to the structural genes. The repressor
containing lactose. molecule has an affinity for lactose, in the presence
Such enzymes which are synthesized only of which it leaves the operator reg­ion free enabling
when induced by the substrate are called the transcription to take place. When lactose
induced enzymes. The enzymes which are present is completely metabolized, the repressor
synthesized irrespective of the presence or again atta­c hes to the operator, switching off
absence of the substrate are called constitutive transcription. Lactose acts both as an inducer and
enzymes. the substrate for the enzyme.

Regulation of Gene Expression B. Genotypic Variation


Lac Operon It is the hereditary constitution of the cell that is
Originally It was proposed in the early 1960s by transmitted to its progeny.
Jacob and Monod. They suggested that segments Genotypic variations are stable, heritable and
of bacterial DNA are organized into functional not influenced by the environment. They may
units called operons of which the most well-known occur by mutation or by one of the mechanisms of
example is the lactose operon of Esch. coli (Fig. 10.3). genetic transfer or exchange.
Lactose fermentation requires three enzymes:
Beta-galactosi­dase, galactoside permease and Mutation
transacetylase coded by structural genes LacZ, LacY It is a random, undirected, heritable variation
and LacA of Lac operon respectively (Fig. 10.4.). caused by an alteration in the nucleotide sequence
Adjacent to the structural gene is the operator, at some point of the DNA of the cell. It may be due
which is a sequence of bases that controls the to addition, deletion or substitution of one or more
expression (transcription) of the structural genes bases (Fig. 10.5).
and a promoter next to the operator where the RNA The molecular mechanism of mutation is that
polymerase binds that will transcribe the structural during DNA replication, some ‘error’ creeps in while
genes. the progeny strands are copied.

www.ebook3000.com
62  |  Section 1: General Bacteriology
Mutagens
1. Physical agents: (i) UV rays; (ii) lonizing
radiation, e.g. X-rays; (iii) Visible light; (iv) Heat
2. Chemical agents: (i) Alkylating agents; (ii)
Acridine dyes ; (iii) 5-Bromouracil; (iv)
2-aminopurine; (v) Nitrous acid.
C. Point mutations: As the name suggests, point
mutations affect just one point (base pair)
Fig. 10.5: Examples of types of mutation. A portion of the wild in a gene. Such mutations may be a change
type chromosome is shown on the top sequence from which to or substitution of a different base pair.
different mutational rearrangements are derived
Alternatively, a point mutation can result in
the deletion or addition of a base pair. It is, in
Mutation is a natural event, taking place all the general, reversible and is of two classes:
time at its particular frequency in all the dividing 1. Base pair substitution: This comprises those
forms of life. Particular mutations occur at fairly mutants in which a single base pair (nucleotide)
constant rates, normally between once per 104 and has been substituted for another pair, and can
once per 1010 cell divisions. be subdivided into transition (one purine is
Though mutations are taking place all the time, replaced by other purine or a pyrimidine is
most mutants go unrecognized as the mutation replaced by other pyrimidine) and transversion
may involve minor function or it may be lethal. (substitution of a purine for a pyramidine and
Mutation is best appreciated when it involves a vice versa in base pairing).
func­tion which can be readily observed. Mutants
can be detected by selecting or testing for an altered Normal sequence: The Fat Cat Ate The Rat.
phenotype. For example, an E. coli mutant that Substitution: The Fat Can Ate The Rat.
loses its ability to ferment lactose can be readily
Depending on the placement of the substituted
detected on MacConkey agar but is unrecognizable
base, when the mRNA is translated this may cause
on nutrient agar.
no change (silent mutation), lead to the insertion
Lethal mutation: Some mutations involve vital of the wrong amino acid (missense mutation)
functions, and such mutants are nonviable (lethal or generate a stop codon (nonsense mutation),
mutation). An important type of lethal mutation prematurely terminating the polypeptide.
is conditional mutation.
Conditional mutation: A condi­tional lethal mutant Missense mutation: If the triplet code is altered
may be able to live under certain permissive so as to specify an aminoacid different from that
conditions but not under other or non­permissive normally located at a particular position in the
conditions. The most common type of con­ditional protein. This is called missense mutation.
mutant is the temperature sensitive (ts) mutant, Nonsense mutation: Deletion of a nucle­otide within
which is able to live at the permissive temper­ature a gene may cause premature polypeptide chain
(say, 35°C), but not at the restrictive tempera­ture termination by generating a nonsense codon (UAG,
(say, 39°C). UAA or UGA). This is known as nonsense mutation.
Recognition of mutation: Mutation can be best
recognized when it involves a function which can 2. Base pair deletion or insetion
be readily observed by experimental methods like Frameshift mutations: If the number of bases
alteration in colonial morphology, pig­mentation, inserted or deleted is not a multiple of three, there
alteration in cell surface antigens, sensitivity to will be shift in the reading frame, i.e. frameshift
bacteriophages or bacteriocins, loss of abil­ity to mutations. This shifts the normal ‘reading frame’ of
produce capsule or flagella, loss of virulence and the coded message forming newest of triplet codon.
change in biochemical characters. The coded message is read correctly up to the point
of addition or deletion, but the subsequent codons
Types of Mutation will specify the incorrect amino acids (Fig. 10.4).
Mutations can be divided conveniently into:
A. Spontaneous mutation: Many mutations occur Survival advantage of mutation: When mutation
spontaneously in nature in the absence of any confers a survival advantage, it is of vital importance.
mutation-causing agents. For example, if a streptomycin resistant mutant of
B. Induced mutation: The frequency of mutation is the tubercle bacillus develops in a patient under
greatly enhanced by exposure of cells to several treatment with the drug, it multiplies selectively
agents (mutagens) which may be physical or and ultimately replaces the orig­inal drug sensitive
chemical. population of bacteria.
Chapter 10: Bacterial Genetics  | 63
Importance of Bacterial Mutation
1. Drug resistance and develop­m ent of live
vaccines: The practical importance of bacterial
mutation is mainly in the field of drug resistance
and develop­ment of live vaccines.

Mutant Selection in Laboratory


1. Fluctuation test: Luria and Delbruck (1943)
provided the proof that bacteria undergo
spontaneous mutation independent of the
environment by the ‘fluctuation test’. They
found that very wide fluctuations occurred
in the numbers of bacteriophage resistant E.
Fig. 10.6: Fluctuation test
coli colonies when samples were plated from
several separate small volume cultures, as
compared to samples tested from a single large
volume culture. Statistically, this indicated that
mutations occurred randomly in the separate
small volume cultures (Fig. 10.6) some early
and some late, resulting in the wide fluctuation.
In the large volume cultures, fluctuations
were within limits of sampling error. However,
the logic of this experiment was not widely
appreciated by microbiologists, probably due
to the complicated statistical interpretation.
2. Other tests
A. Direct (positive) selection
B. Indirect (negative) selection
C. Conditional lethal mutants
D. The Ames test.
A. Direct selection: Direct selection involves
inoculating cells onto a medium on which the
mutant, but not the parent, can grow.
B. Indirect selection: Indirect selection is required
to isolate an auxotrophic mutant, one that
requires a growth factor which the parent strain
does not. Fig. 10.7: Replica plating method
1. Replica plating: Replica plating, was devised
by the husband-and-wife team of Joshua and D. The Ames test: The test is based on the ability
Esther Lederberg in the early 1950s (Fig.10.7). of a potential mutagen to revert an auxo­trophic
In this technique, a master plate containing mutant to its prototrophic form.
isolated colonies of all cells growing on an
enriched medium is pressed onto sterile velvet Transmission of genetic material
(velvet template). Next, two sterile plates, one (gene transfer)
containing a glucose-salts (minimal) medium
DNA may be transferred between bacteria by the
and the second an enriched, complex medium,
following mechanisms:
are pressed in succession onto the same velvet.
A. Transformation
All cells that do not have a nutritional
B. Transduction
requirement will form colonies on both the
C. Lysogenic conversion
enriched and the glucose-salts medium, but
D. Conjugation.
auxotrophs will only form colonies on the
enriched medium- glucose-salts medium.
2. Penicillin Enrichment. A. Transformation
C. Conditional lethal Mutants Transformation is the transfer of genetic
One class of conditional lethal mutants are information through the agency of free (“naked”)
called temperature-sensitive mutant that can DNA (Fig. 10.8). The initial experiment on
grow only at their lower range of temperature transformation was performed by Frederick Griffith
as a result of defective proteins. in Eng­land in 1928.

www.ebook3000.com
64  |  Section 1: General Bacteriology
Griffith’s Experiment Demonstrating Genetic them genetically so that their progeny were
Transformation encapsulated and therefore virulent.
i. Griffith (1928) found that injections of living The nature of the trans­forming principle was
encapsulated bacteria killed the mouse (Fig. identified as DNA by Oswald T. Avery and
10.9). his associates Colin M MacLeod and Maclyn
ii. Injections of live nonencapsulated bacteria McCarty in the United States in 1944.
or dead encapsulated bacteria did not kill the Transformation and Bacteria: Transformation
mouse. occurs naturally among very few genera of bacteria,
iii. When the dead encapsulated bacteria were including Bacillus, Haemophilus, Neisseria,
mixed with live nonencapsulated bacteria and Acinetobacter, and certain strains of the genera
injected into the mice, many of the mice died. Streptococ­cus and Staphylococcus.
In the blood of the dead mice, Griffith found
living, encapsulated bacteria. Hereditary B. Transduction
material (genes) from the dead bacteria
had entered the live cells and changed The transfer of a portion of the DNA from one
bacterium to another by a bacteriophage is known
as transduction. Bacteriophages are viruses that
parasitise bacteria and consist of a nucleic acid
core and a protein coat. Most bacteriophages carry
their genetic information (the phage genome) as a
length of double-stranded DNA coiled up inside
a protein coat. When bacteriophages multiply
inside an infected bacterial cell, each phage head is
normally filled with a copy of the replicated phage
genome. During the assembly of bacte­riophage
progeny inside infected bacteria, ‘packaging
errors’ may occur occasionally. A phage particle
may have at its core a segment of the host DNA
besides its own nucleic acid. When this particle
infects another bacterium, DNA transfer is affected
and the recipient cell acquires new characteristic
coded by the donor DNA. Bacterial genes have
been transduced by the phage into the second cell
Fig. 10.8: The mechanism of genetic transformation in
bacteria
(Fig.10.10).

Fig. 10.9: Transformation experiment of Griffith


Chapter 10: Bacterial Genetics  | 65

Fig. 10.10: Transduction by a bacteriophage (showing generalized transduction)

Types of Transduction only as a vehicle carry­ing bacterial genes


Two major types of transduction are known to from one cell to another; but in lysogenic
occur in bacteria: Generalized transduction and conversion, the phage DNA itself is the new
specialized transduction. genetic element. Lysogenic conversion
1. Generalized transduction: Since phages of influences susceptibility to bacteriophages
this type pick up any portion of the bacterial (immunity to super­infection with the same or
chromosome at random are termed generalized related phages) and anti­genic characteristics.
transducing phages. Lysogeny is extremely fre­quent in nature.
This is a symbiotic relationship in which the
2. Specialized or restricted transduction: A specific
integrated phage DNA imparts immunity to
bac­teriophage transduces only a particular
the lysogenized cell against superinfection by
genetic trait.
genetically related phages, as well as certain
unrelated phages.
Role of Transduction
1. In episomes and plasmids. Medical Importance
2. Penicillin resistance in staphylococci: The
1. Toxigenicity in diphtheria bacilli: Of great
plasmids determining penicillin resistance in
medical importance is the lysogenic conversion
staphylococci are transferred from cell to cell
in diphtheria bacilli, which acquire toxigenicity
by transduction.
(and therefore virulence) by lysogenization
3. Genetic mapping of bacteria.
with the phage beta.
4. Treatment of some inborn metabolic defects.
2. Production of staphylococci, streptococci
and Clostridia toxins is also dependent
C. Lysogenic Conversion upon lysogenic conversion by specific
Bacteriophages exhibit two types of life cycle: bacteriophages.
Virulent or lytic cycle; ii. Temperate or nonlytic
cycle: D. Conjugation
i. Virulent or lytic cycle: In the virulent or lytic Conjugation is a process in which one cell, the
cycle, large numbers of progeny phages are donor or male cell, makes contact with another,
built up inside the host bacterium, which the recipient or female cell, and DNA is transferred
ruptures to release them. directly from the donor into the recipient.
ii. Temperate or nonlytic cycle: In the temperate or Lederberg and Tatum (1946) first described
nonlytic cycle, the host bacterium is unharmed. bacterial conju­gation in a strain of E. coli called
The phage DNA becomes integrated with K12.
the bacterial chromosome as the prophage
and is replicated stably as part of the host Types of Conjugation
cell chromosome and is transferred to the
Three types of conjugation are described below:
daughter cells. This process is called lysogeny
and bacteria harbouring prophages are called 1. Plasmid Transfer
lysogenic bacteria. In lysogenic bacteria, the Populations of E. coli can be divided into two types
prophage behaves as an additional segment of cells. One, the donor cell, contains an F or fertility
of the bacterial chromosome, coding for new plasmid and is designated F+. The other, the recipient
characteristic. This process by which the cell, does not contain this plasmid and is called
prophage DNA confers genetic information F–. DNA is transferred only in one direction, from
to a bacterium is called lysogenic or phage F+ to F– that is in a polar fashion. Consequently, F+
conversion. In transduction, the phage acts cells are often referred to as males and the F– cells

www.ebook3000.com
66  |  Section 1: General Bacteriology

Fig. 10.11: Conjugation in E. coil

as females. The F plasmid codes for the synthesis of


a struc­ture, the sex or F pilus (Conjugation tube),
the protein appendage that attaches the donor
to the recipient cell (Fig. 10.11) and holds the
two cells together so that DNA can then pass into
the recipient cell. Donor cells can transfer their F
plasmid but not their chromosome into recipi­ent
cells. In this way, a sex factor may spread rapidly
Fig. 10.12: High-frequency recombination (Hfr)
through a whole population of recipient cells. This
process is sometimes described as infectious spread
of a plasmid. The maleness in bacteria is thus a 3. Plasmid and Chromosomal Transfer
transmissible or ‘infectious’ characteristic.
There is an additional mechanism by which
F factor (Fertility factor) chromosomal genes may be mobilized by con­
The F factor is a transfer factor that contains the jugation to a recipient cell. This conversion of
genetic information necessary for the synthesis of an F+ cell into the Hfr state is reversible. When
the sex pilus and for self-transfer. Cells that contain the F factor reverts from the inte­grated state to
the F plasmid free in the cytoplasm (F+ cells) have the free state, the F plasmid is not always excised
no unusual characteristics apart from the ability accurately, and occasionally an F plasmid is
to produce F pili and to transfer the F plasmid excised together with some of the neighboring
to F– cells by conjugation. It was in E. coli K12 chromosomal genes. An F plasmid that has picked
that the role of plasmids in conjugation was first up a small portion of the chromosome in this way
recognized. When other similar plasmids were also is known as an F-prime (F’). When an F’ cell mates
discovered, the name ‘transfer factor’ came to be with a recipient, it transfers along with the F factor,
used for all such plasmids which conferred on their the host genes incorporated with it. This process
host cells the ability to act as donors in conjugation. of transfer of host genes through the F’ factor
resembles transduction and has therefore been
called sexduction (Fig. 10.13)
2. Chromosomal Transfer
High-frequency recombination (Hfr)( Fig. Medically Important Factors Transferred
10.12): The bacterial chromosome is sometimes by Conjugation
transferred to F-cells by a few cells in the F
population. The F factor is, actually an episome Colicinogenic (Col) factor and resistance transfer
and has the ability to exist in some cells in the factor (RTF) are two medically important factors
‘integrated state’ or inserted into the host which can be transferred by conjugation.
chromosome in a very small proportion of F +
cells. Once inserted, the entire chromosome A. Colicinogenic (Col) Factor
behaves like an enormous F plasmid, and hence Several strains of coliform bacteria produce
chromosomal genes can be transferred in the colicins-antibiotic-like sub­s tances which are
normal sex factor manner to a recipient cell specifically and selectively lethal to other
at a relatively high frequency. Cultures of cells enterobacteria. Bacteria other than coliforms
in which the F plasmid has inserted into the also produce similar kind of substances, e.g.
chromosome are consequently termed high- pyocin by Pseudomonas pyocyanea, diphthericin
frequency recombination (Hfr) strains because by Corynebacterium diphtheriae, so the name
such cells are able to transfer chro­mosomal genes bacteriocin has been given to this group of
to recipient cells with high frequency. substances. Colicin production is determined by
Chapter 10: Bacterial Genetics  | 67

Fig. 10.13: Sexduction

a plasmid called the Col factor, which resembles


the F factor in promoting conjugation, leading to
self-transfer and, at times, transfer of chromosomal
segments.

B. Resistance Factors or R Plasmids


Resistance factors (R factors) are plasmids that
have significant medical importance as it leads
to the spread of multiple drug resistance among
bacteria. They were first discovered in Japan in
1959 after several dysentery epidemics by the
Shigella strains, resistant simultaneously to usual
four drugs. In addition, they observed that patients
excreting such Shigella strains also shed in their
feces other normal bacteria from the patients such
as E. coli strains resistant to the same drugs. The Fig. 10.14: Two regions of an R plasmid. The RTF contains
genes needed for plasmid replication and transfer of the
re­sistance is plasmid-mediated and is transferred
plasmid by conjugation to other bacteria; r determinants
by conjugation. This mechanism of drug resistance carry genes for resistance to different antibiotics
is known as transferable, episomal or infectious
drug resistance.
Characteristics of R factor: This R plasmid consists
of two components : RTF+r determinants. The Factors influencing R factor transfer: The transfer
whole plasmid (RTF+r determinants) is known can be effect­ed readily in vitro. It also occurs in
as the R factor. An R factor can have several r vivo but in the normal gut, it is inhibited by several
determinants, and resistance to as many as eight factors, such as anaerobic conditions, bile salts,
or more drugs can be transferred simultaneously alkaline pH and the abundance of anaerobic. In
(Fig. 10.14). the intestines of persons on oral antibiotic therapy,
Resistance transfer factor (RTF): The transfer factor transfer occurs readily due to the destruction of the
called the resistance transfer factor (RTF) is res­ sensitive normal flora and the selection pressure
ponsible for conjugal transfer. produced by the drug.

Resistance determinant (r): A resistance determinant Features of R Factor


(r) code for resistance against various drugs. 1. Transfer to antimicrobial and heavy metal-
sensitive bacteria.
Nonconjugative and conjugative plasmids:
Sometimes the RTF may dissociate from the r 2. Wide host ranges and can multiply in a wide
determinants, the two components existing as variety of different gram­-negative genera, such
separate plasmids. In such cases, though the host as Shigella, Salmonella, Escherichia, Yersinia,
cell remains drug resistant, the resistance is not Klebsiella, Vibrio, and Pseudomonas.
transferable. Those plasmids which lack RTF, i.e. 3. Serious problems for the treatment of infectious
possess only r deter­minants are known as non- diseases with antibiotics.
conjugative or nonself­transmissible plasmids but 4. Transmission from animals to man: Hence,
they still code for drug resistance. Those plasmids indiscriminate use of antibiotics in veter­inary
which possess both RTF and r determinants practice or in animal feeds can also lead to
are known as conjugative or self-transmissible an in­crease of multiple drug resistance in the
plasmids. community.

www.ebook3000.com
68  |  Section 1: General Bacteriology
Genetic mechanisms of drug Table 10.1  Comparison of mutational and trans­
resistance in bacteria ferable drug resistance
Mutational drug Transferable drug
Mechanisms of Drug Resistance in resistance resistance
Bacteria 1. Resistance to one 1. Multiple drug resistance
A. By mutation (chromosomal) drug at a time at one time
B. By genetic exchange. 2.  Low degree resistance 2.  High degree resistance
3. Can overcome by high 3.  High dose ineffective
A. By Mutation (Chromosomal) drug dose
Mutational resistance is mainly of two types: 4. Development of 4. Development of drug
1. Stepwise mutation: The stepwise mutation, drug resistance resistance cannot be
as seen with penicillin, where high levels of can be prevented prevented treatment
by treatment with with combination of
resistance are achieved only by a series of combination of drug drugs
small-step mutations. 5. Resistance is not 5. Resistance is transferable
2. Single large-step mutation or ‘one-step’ mutation: transferable to other organisms
As seen with streptomycin, the drug target is 6. Mutants may 6.  Not defective
altered by mutation so that it is totally unable be metabolically
to bind a drug. defective
7.  Virulence may be low 7.  Virulence not decreased
Clinical importance in tuberculosis: Mutational
resistance is of clinical importance in tuberculosis.
If a patient is treated with streptomycin alone are larger (4–25 Kb) composite elements
initially, the bacilli die in large numbers but soon containing genes for movement as well as
resistant mutants appear and multiply unchecked. genes that encode for various functions, such
If two or more antituberculous, drugs are used for as drug resistance and toxin production. In the
combined treatment, repopulation by resistant 1950s, American geneticist Barbara McClintock
mutants does not occur, as a mutant resistant to discovered transposons in corn during her studies
one drug will be destroyed by the other drug. on maize genetics, for which she was awarded the
Inadequate or inappropriate treat­ment over the Nobel prize for Medicine in 1983.
years has caused extensive resistance in tubercle
bacilli, leading to a pandemic of multidrug resistant
Structure of Transposons
tuberculosis (MDR TB) across the world. The simplest transposable elements are insertion
sequences (IS), or IS elements. It is a short
B. By Genetic Exchange sequence of DNA that contains a gene that
Transferable antibiotic resistance codes for enzyme. The more complex is called a
1. Transformation: Its significance in nature is not composite transposon.All transposons contain the
known. information for their own transposition.
2. Transduction: Acquisition of resistance by A transposon is a segment of DNA with
transduction is common in staphylococci. one or more genes in the center, and the two
The penicillinase plasmids are transmitted by ends carrying ‘inverted repeat’ sequences of
transduction. nucleotides-­nucleotide sequences complementary
3. Conjugation: Plasmids conferring resistance to each other but in the reverse order. Because of
to one or more unrelated groups of antibiotics this feature, each strand of the transposon can
(R plasmids) can be transferred rapidly by form a single-stranded loop car­r ying the gene,
conjugation. and a double-stranded stem formed by hydrogen
Transferable drug resistance mediated by the bonding between the terminal inverted repeat
R factor is the most important method of drug sequences (Fig. 10.15).
resist­ance. Table 10.1 compares mutational and
Significance of Transposons
transferable drug resistance.
1. Mechanism for amplifying ge­netic transfers
Transposable genetic elements 2. Spread from one organism-or species-to another
3. Antibiotic resistance
Certain structurally and genetically discrete
4. Gene manipulations.
seg­ments of DNA which move around between
chromosomal and extrachro­m osomal DNA Molecular Genetics
molecules within cells are called transposons
(‘jumping genes’). This mode of genetic transfer Genetic Engineering
is called transposition. They vary from simple Genetic engineering is a method of inserting foreign
(insertion sequences) to complex. Transposons genes into a bacterium and obtaining chemically
Chapter 10: Bacterial Genetics  | 69
useful products. Genetic engineering, also known of higher forms of life including human beings, and
as recombinant DNA (rDNA) technology, uses the their introduction into suitable microorganisms, in
techniques and tools developed by the bacterial which the genes would be functional, directing the
geneticists to purify, amplify, modify, and express produc­tion of the specific protein. Such cloning of
specific gene sequences. genes in microorganisms enables the preparation
of the desired protein in pure form, in large
Genetic Engineering Procedure (Fig. 10.16) quantities and at a reasonable cost.
This consists of isolation of the genes coding for any
desired protein from microorganisms or from cells Basic Tools of Genetic Engineering
1. Cloning vectors: Cloning vectors can be used
to deliver the DNA sequences into receptive
bacteria and amplify the desired sequence.
Many types of vectors are currently used
such as plasmid vectors, bacteriophages and
other viruses, cosmid vectors, and artificial
chromosomes.
2. Restriction endonucleases (restriction
enzymes): Restriction enzymes are microbial
enzymes which cleave double-stranded DNA at
specific oligonucleotide sequences. Restriction
enzyme recognizes and cuts, or digests, only
one particicular sequence of nucleotide bases
in DNA, and it cuts this sequence in the same
way each time. These enzymes are present in
many prokaryotes organisms, e.g. restriction
endonuclease Eco RI, Hind Ill, obtained from
E. coli, H. influenzae.
3. DNA ligase: The enzyme that links the fragment
Fig. 10.15: Structure of transposon to the cloning vector.

Fig. 10.16: A typical genetic engineering procedure

www.ebook3000.com
70  |  Section 1: General Bacteriology
Applications of Genetic Engineering Hybridization: It is the process whereby two single
1. Production of vaccines: Foot and mouth disease, strands of nucleic acid come together to form a
hepatitis B, ra­bies viruses and DNA vaccine. stable double-stranded molecule.
2. Production of proteins of therapeutic interest: Detection of hybridization: Hybridization assays
Human growth hormones, human insulin, require that one nucleic acid strand (probe)
erythropoietin, blood-clotting factor VIII, originates from an organism of known identity
tissue plasminogen activator, interferons, and the other strand (the target) originates from
tumor necrosis factor, interleukin-1, 2, and unknown organisms to be detected or identified.
3, granulocyte colony stimulating factor, Identification of unknown organism is
epidermal growth factor, fibroblast growth established by positive hybridization (i.e. duplex
factor, somatostatin, growth hormones are formation) between a probe nucleic acid strand
proteins of therapeutic interest. (from unknown organisms) and a target nucleic
C l o n e d h u m a n i n s u l i n , i n t e r f e r o n s, acid strand from the organism to be identified.
somatostatin, growth hormones and many Failure to hybridize indicates lack of homology
other biologicals have already been marketed. between probe and target nucleic acid. Positive
3. Gene therapy. hybridization identifies the unknown organism
4. Others: It has also become essen­t ial to as being the same as the probe-source organisms.
laboratory diagnosis, forensic science, agricul­ With a negative hybridization test, the organism
ture, and many other disciplines. remains undetected or unidentified (Fig. 10.17).

Genetic probes Applications of Nucleic Acid Probes


DNA Probes DNA probes have already been used successfully
to identify a wide variety of pathogens, from simple
DNA probes are pieces of radiolabeled or
viruses to pathogenic bacteria and parasites.
chromogenically labeled pieces of single-stranded
1. Antibiotic resistance: Probes have also been
DNA that will bind to DNA that is complementary
developed which can recognize specific
to the probe using hybridizing technique. The
antibiotic resistance genes.
specificity of the interaction in base pairing during
2. Culture confirmation: DNA probes are being used
DNA or RNA synthesis enables the production of
for culture confirmation as an alternative to con­
specific DNA probes.
ventional, time-consuming or labor-intensive
Development of nucleic acid probe: All meth­o ds in the diagnostic laboratory. For
microorganisms, simple or complex, contain example, DNA hybridization makes it pos­sible
some unique sequences of DNA or RNA within to rapidly identify Mycobacterium tuberculo­sis,
their genome that ‘distinguish them from all other M. kansasii, M. avium complex, and M. gordonae
organ­isms. DNA probes are chemically synthesized isolated in culture, significantly reducing the time
or obtained by cloning specific genomic fragments for reporting of the species of the isolate.
or an entire vi­ral genome into bacterial vectors 3. Direct detection in clinical specimen: For
(plasmids, cosmids). detection of fas­tidious organisms directly in

Fig. 10.17: Principles of nucleic acid hybridization


Chapter 10: Bacterial Genetics  | 71
clinical specimens, probe technology may also (or genes). PCR was invented and patented from
be used. Examples are Neisseria gonorrhoeae the Cetus Corpo­ration. Kary Mulis invented this
and Chlamydia trachomatis. method in 1989. He was awarded Nobel Prize in
1993. It is the most widely used target nucleic acid
Blotting techniques amplification method.
1. Southern blotting: Very specific DNA sequences Principle: PCR is based on repeated cycles
can be detected us­ing hybridization techniques of high temperature template denaturation,
with a technique known as the Southern blot. oligonucleotide primer annealing, and polymerase
This highly sensitive technique for identifying mediated extension. It makes avail­able abundant
DNA frag­ments by DNA: DNA hybridization is quantities of specific DNA sequences starting from
called Southern blotting, after EM Southern sources containing minimal quantities of the same.
who devised it. This tech­nique has very wide Procedure: The reaction consists of three essential
applications in DNA analysis. steps:
2. Northern blotting: An analogous procedure for 1. Dena­turation: Heat at 94°C is applied to the
the analysis of RNA has been called Northern target DNA, breaking the bonds that hold the
blotting (as opposed to southern blotting). strands toge­ther. This is known as denaturation.
Here the RNA mixture is separat­e d by gel 2. Primer annealing: The temperature is reduced
electrophoresis, blotted and identified using to 50–60°C, then oligonucleotide primers
labeled DNA or RNA probes. attach to target DNA. This temperature is
3. Western blotting (Western blot assay): A similar called annealing temperature and the process
technique for the identificaion of proteins is known as annealing of primers.
(antigens) is called immunoblotting (or, in 3. Primerextension: Then polymerase enzyme
conformity with other blotting technique, triggers the forma­t ion of new DNA strand
western blotting). The Western blot detects from the nucleotides. Extension of the primers
antibody instead of DNA. is done by a thermo­stable Taq polymerase
i. The DNA (or RNA) of a particular etiologic (purified from Thermus aquaticus. This
agent is treated with endonucleases, or the is known as primer extension. When, the
protein components of an agent are treated temperature is again raised the new strands
with proteinases to create fragments of separate and the process begins again.
different size. All of the necessary reagents are added
ii. The nucleic acid or protein fragments are to a tube, which is placed in a thermocycler.
separated by agarose gel electrophoresis, These pro­g rammable thermal cycles are
i.e. PAGE (sodium dodecyl sulfate-poly­ used to maintain continuous reaction cycles.
acrylamide gel electrophoresis). As shown in Figure 10.18, for each target
iii. The patterns are then blotted onto nitro­ sequence originally present in the PCR mixture,
cellulose as in the Southern hybridization two double stranded fragments containing
assay. the target sequence are produced after one
iv. The filter paper containing spe­cific nucleic cycle. Therefore, with comple­tion of each cycle
acids or proteins is then allowed to react there is a doubling of target nucleic acid and
with antiserum from a patient or animal after 30–40 cycles 107–108 target copies will
sus­pected of containing antibodies against be present in the reaction mixture, a sharp
the agent. If present, antibodies will bind contrast to the days required by conventional
to the protein or nucleic acid against amplification method (culture).
which they were created, and they can Detection of PCR Products
then be detected by visual methods for
Amplified sequences of target DNA can be det­ected
the detection of antibodies (e.g. enzyme-
by a variety of methods:
labeled probes, fluorescent markers).
1. Gel electrophoresis and ethidium bromide
Use: Diagnosis of human immunodeficiency staining.
virus (HIV) antibody—The Western blot test has 2. Southern blot and dot-blot analysis.
received wide publicity as the confirma­tory test for
the diagnosis of human immunodeficiency virus Applications of PCR
(HIV) antibody in sera. The development of PCR or gene amplification
method is a major meth­odological breakthrough
Polymerase chain reaction (PCR)
in molecular biology. Within a short span, this
Polymerase chain reaction (PCR) or gene method has found its way into nearly every type
amplification method is a rapid automated method of laboratory from forensic to ecology and from
for the amplification of specific DNA sequences diagnosis to pure research.

www.ebook3000.com
72  |  Section 1: General Bacteriology

Fig. 10.18: Polymerase chain reaction

1. In clinical laboratory: Applications of PCR in Gene Therapy


clinical laboratory are given in Table 10.2.
2. In diagnosis of inherited disorders: Such as sickle Gene therapy is inserting a missing gene or replac­
cell anemia, β-thalassemia, cystic fibrosis, etc. ing a defective one in human cells. The benefit of
3. In cancer detection: Identification of muta­ this therapy is to permanently cure the physiological
tions in oncosuppressor genes, such as dysfunction by repairing the genetic defect.
retinoblastoma gene. Adenoviruses and retro­viruses are used most
4. In medicolegal cases: PCR allows DNA in often to deliver genes.
a single cell, hair follicle or sperm to be
amplified enormously and analyzed. The Applications
pattern obtained is then compared with that
1. To treat patients with adenosine deaminase
of various suspects.
(ADA) deficiency, a cause of severe combined
immunodeficiency disease (SCID); Duchenne’s
Derivations/Modifications of the PCR muscular dystrophy, a muscle-destroying
Method disease; cystic fibrosis; and LDL-receptor
Specific examples include multiplex PCR, nested deficiency. Results are still being evaluated.
PCR, quantitative PCR, RT-PCR, arbitrary primed 2. Hemophilia: The first gene therapy to treat
PCR, and PCR for nucleotide sequencing. hemophilia in humans was done in 1999.
Chapter 10: Bacterial Genetics  | 73
Table. 10.2  Applications of PCR in clinical laboratory
Diagnosis of infections due to:
A. Viruses HIV-1, HIV-2, HTLV-1, cytomegalovirus, human papillomavirus, herpes simplex viruses, hepatitis B virus, HCV,
HDV, HEV, rubella virus, Epstein-Barr virus, varicella-zoster virus, human herpes virus-6 and 7, parvovirus B19,
enteroviruses, coxsackieviruses/echoviruses, rhinoviruses, measles virus, rotavirus, I adenovirus, respiratory
syncytial virus.
B. Bacteria Mycobacterium tuberculosis, Mycobacterium avium complex, Legionella pneumophila, Chlamydia trachomatis,
Mycoplasma pneumoniae, Helicobacter pylori, Burkholderia pseudomallei, Campylobacter spp., Corynebacterium
diphtheriae, Leptospira interrogans, Streptococcus pyogenes, Streptococcus pneumoniae, Yersinia enterocolitica.
C. Fungi Candida spp., Cryptococcus neoformans, Aspergillus spp. Pneumocystis carinii.
D. Protozoa Toxoplasma gondii, Trypanosoma cruzi, Enterocytozoon bieneusi, Encephalitozoon hellem, Plasmodiumspp.
HIV-1, human immunodeficiency virus; HTLV-1, human T cell leukemia virus; HCV, hepatitis C virus; HDV, hepatitis D virus; HEV, hepatitis
E virus

3. Other genetic diseases may also be treatable Important questions


by gene therapy in the future, including
1. Describe the structure and functions of the plasmids.
diabetes, sick­le cell disease, and one type of 2. Define mutation. Discuss various types of mutations.
hypercholesterolemia, high blood cholesterol. 3. Name the various methods of gene transfer. Discuss
4. To treat hepatitis, cancers, and one type of anyone of these in detail.
coronary artery disease. 4. Write short notes on:
a. Extrachromosomal genetic elements
Key Points b. Plasmids
c. Lac operon
™™ Genetics is the study of genes, their structure and d. Transformation
function, heredity and variation
e. Transduction
™™ A gene is a segment of DNA, a sequence of
f. Lysogenic conversion
nucleotides, that codes for a functional product,
g. Conjugation
usually a protein
h. Fertility factor (F-factor)
™™ There are three different functional groups of RNA
molecules: messenger RNA (mRNA), ribosomal RNA i. High frequency recombination (Hfr)
(rRNA), and transfer RNA (tRNA) j. R-plasmids
™™ Plasmids: Bacte­ria often have one or more small k. Resistance transfer factor (RTF)
closed loops of DNA called plasmids. These 5. Differentiate between mutational and plasmid-
structures carry infor­mation that can confer selective mediated drug resistance in a tabulated form.
advantages (e.g. antibiotic resistance, protein toxins) 6. Write short notes on:
to the bacteria that contain them a. Transposable genetic elements
™™ Mutation: Mutation is a permanent change in the b. Genetic engineering
cellular DNA. This can occur spontaneously in nature c. Restriction endonucleases
resulting from a replication error or the effects of 7. Write short notes on:
natural radiation –– DNA probes and their applications.
Mechanisms of Gene Transfer –– Blotting techniques.
1. Transformation: DNA-mediated transformation 8. Discuss polymerase chain reaction. What are the
involves the transfer of “naked” DNA applications of this reaction in clinical practice?
2. Transduction: Transduction involves the transfer of
bacterial DNA by a bacteriophage Multiple choice questions (MCQs)
In generalized transduction, any bacterial genes can
be transferred. 1. The following are non-sense codons except:
3. Lysogenic conversion has two types of life cycles. a. UAA b. UAG
4. Conjugation c. UGA d. AGG
2. The process of transfer of genetic information from
™™ R plasmids code for antibiotic resistance; many are
self-transmissible to other bacteria DNA to RNA is known as:
a. Transformation
™™ Mechanisms of drug resistance in bacteria
b. Transduction
(A) By mutation (chromosomal); (B) By genetic
c. Transcription
exchange
d. Translation
™™ Transposons are small segments of DNA that can
3. The process of transfer of DNA from the donor
move from one region to another region of the
bacterium to the recipient bacterium during the
same chromosome, or to a different chromosome
or a plasmid mating of two bacterial cells is known as
a. Transformation
™™ Polymerase chain reaction (PCR): PCRs are amplified
methods. It can be used to increase the amounts of
b. Transposition
DNA in sam­ples to detectable levels. c. Transduction
d. Conjugation

www.ebook3000.com
74  |  Section 1: General Bacteriology
4. Which of the following properties may be plasmid- 10. The transfer of genetic information through the
mediated? agency of free (naked) DNA is called:
a. Antibiotic resistance a. Transformation
b. Enterotoxin production b. Transduction
c. Bacteriocin production c. Conjugation
d. All of the above d. Lysogenic conversion
5. The mutation in which a purine is replaced by 11. The transfer of a portion of DNA from one bacterium
pyrimidine and vice-versa in base pairing, is named: to another by a bacteriophage is known as:
a. Transversion a. Transformation
b. Transition b. Transduction
c. Induced mutation c. Conjugation
d. None of the above d. Lysogenic conversion
6. The mutation in which one purine is replaced 12. Which of the following factors is responsible for
by other purine and a pyrimidine is replaced by transferable drug resistance in bacteria?
another pyrimidine, is named as: a. Resistance transfer factor
a. Transversion b. F factor
b. Transition c. Colicogenic factor
d. All of the above
c. Induced mutation
d. None of the above 13. Resistance transfer factor can be transferred from
7. Frame-shift mutation occurs: one bacterium to another by:
a. When one or more base pairs are added or a. Transformation
deleted in the DNA b. Transduction
c. Conjugation
b. When base substitution in DNA produces a
d. None of the above
terminal codon
c. When one base in the nucleotide sequence is 14. Which of the following statement is true for
inserted in place of another mutational drug resistance?
d. Transposons are integrated into the DNA a. Resistance to one drug at a time
b. Resistance to multiple drugs at one time
8. The DNA is transmitted from one bacterium to
c. High degree resistance
another by all the methods except:
d. Resistance is transferable to other organisms
a. Transformation
b. Transposition 15. Western blotting is done for:
a. Detecting DNA sequesing
c. Transduction
b. Analysis of RNA
d. Conjugation
c. Identification of proteins
9. The process of transfer of DNA from the donor d. All of the above
bacterium to the recipient bacterium during the
mating of two bacterial cells is known as: Answers (MCQs)
a. Transformation b. Transposition 1. d; 2. c; 3. d; 4. d; 5. a; 6. b; 7. a; 8. b; 9. d; 10. a; 11. b; 12.
c. Transduction d. Conjugation a; 13. c; 14. a; 15. c.
11
Chapter

Infection

Learning Objectives

After reading and studying this chapter, you should ∙∙ Name and define various types of carriers
be able to: ∙∙ Discuss modes of spread of infection giving suitable
∙∙ Define the terms saprophytes, parasite, commensal, examples
pathogen ∙∙ List the differences between exotoxins and
∙∙ Describe classification of infections endotoxins.
∙∙ Define and differentiate primary, secondary,
opportunistic and reinfections

Introduction a. Primary (frank) Pathogens


Primary (frank) pathogens are the organisms, which
Infection and immunity involve interaction are capable of producing disease in previously
between the animal body (host) and the infecting healthy individuals with intact immunological
microorganisms. defences.

Microorganisms and host b. Opportunist Pathogens


Opportunist pathogens rarely cause disease
Based on their relationship to their host, they can in individuals with intact immunological and
be divided into saprophytes and parasites. anatomical defences. These bacteria are able
to cause disease only when such defences are
A. Saprophytes impaired or compromised.
Saprophytes (from Greek sapros-decayed; and
phyton-plant) are free-living microbes that live on 2. Commensals
dead or decaying organic matter. They are found Commensals (organisms of normal flora) are the
in soil and water. They are of little relevance in microorganisms that live in complete harmony
infectious disease. with the host without causing any damage to it.

B. Parasites Infection and infectious disease


Parasites are microbes that can establish themselves It is necessary to distinguish between the term
and multiply in the hosts. Parasite microbes may ‘infection’ and ‘infectious disease.’
be either pathogens or commensals:
Infection
1. Pathogens The lodgement and multiplication of a parasite in
Pathogens (from Greek pathos, disease; and gen, to or on the tissues of a host constitute infection. It
produce) are the microorganisms or agents, which does not invariably result in disease.
are capable of producing disesase in the host. Its
ability to cause disease is called pathogenicity. Classification of infections
Types of Pathogens Infections may be classified in various ways:
They are of two types: primary and opportunist 1. Primary infection: Initial infection with a
pathogens. parasite in a host is termed primary infection.

www.ebook3000.com
76  |  Section 1: General Bacteriology
2. Reinfections: Subsequent infections by the same 1. Convalescent carrier: An individual who has
parasite in the host are termed reinfections. recovered from the infectious disease but
3. Secondary infection: When a new parasite sets continues to harbor large numbers of pathogen.
up an infection in a host whose resistance is 2. Healthy carrier: A healthy carrier is an individual
lowered by a preexisting infectious disease, who harbors the pathogen but is not ill.
this is termed secondary infection. 3. Incubatory carrier: An incubatory carrier is an
4. Local infection: The term local infection individual who is incubating the pathogen in
(more appropriately local sepsis) indi­cates large numbers but is not yet ill.
a condition where, due to infection or sepsis
4. Temporary carriers: Convalescent, healthy, and
at localized sites such as appendix or tonsils,
incubatory carriers may harbor the pathogen
generalized effects are produced.
for only a brief period (hours, days, or weeks)
5. Cross-infection: When in a patient already suffer­
and lasts less than six months.
ing from a disease, a new infection is set up from
an­other host or another external source, it is 5. Chronic carriers: They harbor the pathogen for
termed cross-infection. long periods (months, years, or life).
6. Nosocomial infections: Cross-infections 6. Contact carriers: The term contact carrier is
occurring in hospitals are called nosocomial applied to a person who acquires the pathogen
infections (from Greek nosocomion hospital). from a patient.
7. Iatrogenic infection: The term iatrogenic infect­ 7. Paradoxical carrier: This refers to a carrier who
ion refers to physician induced infections acquires the pathogens from another carrier.
resulting from investigative, therapeutic or
other procedures. B. Animals
8. Inapparent infection: Inapparent infection is
Reservoir hosts: Many pathogens are capable
one where clinical effects are not apparent.
of causing infections in both human beings and
9. Subclinical infection: The term subclinical
animals. Therefore, animals may act as a source of
infection is often used as a synonym to
infection of such organisms. These, animals serve to
inapparent infection.
maintain the parasite in nature and act as reservoir
10. Atypical infection: Atypical infection is one
and they are, therefore, called reservoir hosts.
in which the typical or char­acteristic clinical
manifestations of the particular infectious Zoonosis: The diseases and infections, which
disease are not present. are transmissible to man from animals are called
11. Latent infection: Some parasites, fol­lowing zoonosis.
infection, may remain in the tissues in a latent
Examples of zoonotic diseases
or hidden form proliferating and producing
clinical disease when the host resistance is Bacterial: Anthrax, brucellosis, Q fever, leptospirosis,
lowered. This is termed latent infection. bovine tuberculosis, bubonic plague, Salmonella
food poisoning.
Sources of infection
Viral: Rabies, yellow fever, cowpox, monkeypox.
A. Human beings
B. Animals Protozoal: Leishmaniasis, toxoplasmosis, trypano­
C. Insects somiasis, babesiosis.
D. Soil and water
Helminthic: Echinococcosis, taeniasis, trichinellosis.
E. Food.
Fungal: Microsporum canis, Trichophyton verru­
A. Human Beings cosum.
The most common source of infection for human
beings is human beings themselves. The parasite
may originate from a patient or carrier.
C. Insects
Humans serving as the microbial reservoir: Arthropod-borne Diseases
i. Acquisition of “strep” throat through touching Blood-sucking insects, such as mosquitos, ticks,
ii. Hepatitis by blood transfusions mites, flies, and lice may transmit pathogens to
iii. Gonorrhea, syphilis, and AIDS by sexual contact human beings and diseases so caused are called
iv. Tuberculosis by coughing; and the common arthropod borne diseases.
cold through sneezing.
Carrier Vectors
A carrier is person who harbors the microorganisms Insects that transmit infections are called vectors.
without suffering from any ill effect` because of it. Vector-borne transmission can be of two types
There are several types of carriers: either mechanical (external) or biological (internal).
Chapter 11: Infection  | 77
i. Mechanical vector: The disease agent is a. Direct contact: Diseases transmitted by direct
transmitted mechanically by the arthropod. contact include STD (sexually transmitted
Examples : Transmission of diarrhea, diseases), such as syphilis, gonorrhea, lympho­
dysentery, typhoid, food poisoning and granuloma venereum, lymphogranuloma
trachoma by the housely. inguinale, trichomoniasis, herpes simplex type
ii. Biological vectors: Biological vectors are those 2, hepatitis B and acquired immunodeficiency
in whom the pathogens multiply sufficiently syndrome (AIDS).
or has undergone a developmental cycle. b. Indirect contact: Fomites: Indirect contact may
The interval between the time of entry of be through the agency of fomites, which are
the pathogen into the vector and the vector inanimate objects, such as clothing, pencils or
becoming infective is called the extrinsic toys which may be con­taminated by a pathogen
incubation period. from one person and act as a vehicle for its
Examples: Aedes aegypti mosquito in yellow fever, transmission to another.
Anopheles mosquito in malaria.
2. Inhalation
Reservoir hosts: Besides acting as vectors, some
Droplet nuclei: Respiratory infections, such
insects may also act as reservoir hosts (for example,
as common cold, influenza, measles, mumps,
ticks in relapsing fever and spotted fever). Infection
tuberculosis and whooping cough are acquired
is maintained in such insects by transovarial or
by inhalation.
transstadial passage.
3. Ingestion
D. Soil and Water
Intestinal infections are generally acquired by the
i. Soil ingestion of food or drink contaminated by path­
Some pathogens can survive in the soil for long ogens. Infection transmitted by ingestion may be
periods. waterborne (cholera), food borne (food poisoning)
Examples or handborne (dysentery). Diseases transmitted
a. Spores of tetanus and gas gangrene: Spores of by water and food include chiefly infections of
tetanus and gas gangrene remain viable in the the alimentary tract, e.g. acute diarrheas, typhoid
soil for several decades and serve as source of fever, cholera, polio, hepatitis A, food poisoning
infection. and intestinal parasites.
b. Fungi and parasites: Fungi (causing mycetoma,
4. Inoculation
sporotrichosis, histoplasmosis) and parasites
The disease agent may be inoculated directly in
such as roundworms and hookworms also
to the skin or mucosa, e.g. rabies virus depos­
survive in the soil and cause human infection.
ited subcutaneously by dog bite, tetanus spores
implanted in deep wounds, and arboviruses
ii. Water injected by insect vectors.
Water may act as the source of infection Infection by inoculation may be iatrogenic
either due to contamination with pathogenic when unsterile syringes and surgical equipment
microorganisms (Shigella, Salmonella, Vibrio are employed. Hepatitis B and the human
cholerae, poliomyelitis virus, hepatitis virus) immunodeficiency virus (HIV).
or due the presence of aquatic vector (cyclops
containing larvae of guinea worm infection). 5. Insects
Vector-borne
E. Food Vector is defined as an arthropod or any living
Contaminated food may act as source of infection of carrier (e.g. snail) that transports an infectious
organisms causing food poisoning, gastroenteritits, agent to a susceptible individual. In some diseases,
diarrhea and dysentery. blood-sucking insects play an important role in the
spread of infection from one individual to another
Modes of Transmission of Infection (Refer to Chapter 85).

Pathogenic organisms can spread from one host to 6. Congenital


another by a variety of mechanisms. These include Vertical Transmission
as follows: Some pathogens are able to cross the placental
barrier and reach the fetus in utero. This is known
1. Contact as vertical transmission.
Infection may be acquired by contact, which may Examples: So-called TORCH agents (Toxoplasma
be direct or indirect. gondii, rubella virus, cytomegalovirus and

www.ebook3000.com
78  |  Section 1: General Bacteriology
herpes virus), varicella virus, syphilis, hepatitis B, 3. Invasiveness
Coxsackie B and AIDS. Invasiveness signifies the ability of a pathogen to
spread in the host tissues after establishing infec­
7. Iatrogenic and Laboratory Infections tion. Highly invasive pathogens characteristically
If meticulous care in asepsis is not taken, infections produce spreading or generalized lesions (e.g. strep­
like AIDS and hepatitis B may sometimes be tococcal septicemia following wound infection),
transmitted during administration of injections, while less invasive pathogens cause more localized
lumber puncture and catheterization. These lesions (e.g. staphylococcal abscess).
are known as iatrogenic or physician-induced
infections. Modern methods of treatment, such 4. Toxigenicity
as exchange transfusion, dialysis, and heart and Some bacteria cause disease by producing toxins,
transplant surgery have increased the possibilities of which there are two general types: The exotoxins
for iatrogenic infections. and the endotoxins (Table 11.1).

Factors predisposing to microbial a. Exotoxins


pathogenicity Exotoxins are soluble, heat-labile proteins
Pathogenicity and Virulence inactivated at 60°–80°C and diffuse read­ily into
the surrounding medium. These are highly potent
Pathogenicity denotes the ability of a microbial
species to cause disease.
Table 11.1  Differences between exotoxins and
Virulence: The term virulence denotes the ability
endotoxins
of a strain of a species to produce disease. For
example, encapsulated pneumococci are more Exotoxins Endotoxins
virulent than nonencapsulated pneumococci. 1. Proteins 1. Lipopolysaccharide on
outer membrane. Lipid
Exaltation: Enhancement of virulence is known as A portion is toxic
exaltation. This can be induced by serial passage
2. Heat-labile. (inactivated 2. Heat-stable
of a strain in as experimental animal.
at 60°–80°C)
Attenuation: Reduction of virulence is known as 3. Actively secreted by the 3. Form integral part of the
attenuation cells; diffuse into the cell wall; do not diffuse
surrounding medium into surrounding medium
Determinants of virulence 4. Readily separable from 4.  Obtained only by cell lysis
cultures by physical
1. Transmissibility means, such as filtration
The first step of the infectious process is the entry of 5.  Action often enzymatic 5.  No enzymatic action
the microorganism into the host by one of several 6. Specific 6. Nonspecific action of all
ports: the respiratory tract, gastrointestinal tract, pharmacological effect endotoxins
urogenital tract, or through skin that has been cut, for each exotoxin
punctured, or burned. 7.  Specific tissue affinities 7.  No Specific tissue affinities
8. Highly toxic and fatal in 8. Moderate toxicity. Active
2. Adhesion microgram quantities only in very large doses

Adhesins: The initial event in the pathogenesis is 9.  Highly antigenic 9.  Weakly antigenic
the attachment of the bacteria to body surfaces. 10. Action specifically neu­ 10. Neutralization by
This attachment is not a chance event but a specific tralized by antibody antibody ineffective
reaction between surface receptors on host cells 11. Usually do not produce 11. Usually produce
and adhesive structures (ligands) on the sur­face fever fever by release of
interleukin-1
of bacteria. These adhesive structures are called
adhesins. 12. Produced by both 12. Produced by gram-
gram-positive bacteria negative bacteria only
Adhesions may occur as organized structures, and gram-negative
such as fimbriae or fibrillae and pilli, or as bacteria
co­lonization factors. 13. Frequently controlled 13. Synthesized directly by
Adhesins as virulence factors: Adhesins are by extrachromosomal chromosomal genes
usually made of protein and are antigenic in nature. genes (e.g. plasmids)
Specific immunization with adhesins has been 14. Disease examples- 14. Gram-negative
attempted as a method of prophy­laxis in some Botulism diphtheria infections,
tetanus meningococcemia
infections.
Chapter 11: Infection  | 79
in minute amounts and include some of the most d. Antigenic Variation
poisonous substances known. Variation in surface antigen composition during
Treatment with formaldehyde converts the course of infection provides a mechanism of
exotoxin into toxoids, are thus useful in preparing avoidance of specific immune responses directed
vaccines. at those antigens.
They exhibit specific tissue affinity and Examples: (i) Pathogenic Neisseria; (ii) The borreliae
pharmacological activities. generate antigenic variation.
They are associated with specific diseases and
have specific mechanisms of action.
e. Serum Resistance
They are easily inactivated by formaldehyde,
iodine, and other chemicals to form immunogenic To survive in the blood, bacteria must be able to
toxoids. resist lysis.
Exotoxins are generally formed by gram-
positive bacteria but may also be produced by some f. Siderophore and Iron Acquisition
gram-negative organisms such as Shiga’s dysentery Siderophores can acquire iron from the host’s iron
bacillus, cholera vibrio and enterotoxigenic E. coli. binding proteins.

b. Endotoxins 6. Enzymes
These are heat-stable, lipopolysaccharide (LPS) Many species of bacteria produce tissue-degrading
components of the outer membranes of gram- enzymes that play important roles in the infection
negative. Their toxicity depends upon the the process.
component (lipid A). i. Coagulase: Coagulase is produced by
They are released into the host’s circulation Staphylococcus. aureus. This thrombin-like
following bacterial cell lysis. enzyme prevents phagocytosis by forming a
They are toxic only at high doses. fibrin barrier around the bacteria and walling
They cannot be toxoided. off the lesion.
They are poor antigens and weakly immunogenic.
ii. Lecithinase-C and collagenase: Clostridiun.
They do not exhibit specific pharmacological
perfringens produces lecithinase-C and
activi­ties.
collagenase promoting spread of infection in
Intravenous injections of large doses of
tissue.
endotoxin and massive gram-negative septicemias
cause endotoxic shock marked by fever, leukopenia, iii. Hyaluronidases: Hyaluronidases split hyalu­
thrombocytopenia, siignificant fall in blood ronic acid and thus facilitate the spread of
pressure, circulatory collapse and bloody diarrhea infection along tissue spaces, e.g. Streptococcus.
leading to death (Table 11.1). iv. Streptokinase (fibrinolysin): Many haemolytic
streptococci produce streptokinase (fibri­
5. Avoidance of Host Defence nolysin) which promotes the spread of
Mechanisms infections.
v. Cytolysins: These include hemolysins capable
a. Capsules of destroying erythrocytes and leukocidins
Some bacteria such as Streptococcus pneumoniae, damage polymorphonuclear leukocytes.
Neisseria meningitidis, and Haemophilus influenzae vi. IgA 1 proteases: These enzymes specifically
can produce a slippery mucoid capsule that cleave immunoglobulin IgA which protects
prevents the phagocyte from effectively contacting at mucosal surfaces.
the bacterium.

b. Streptococcal M Protein 7. Plasmids


Other bacteria evade phagocytosis by producing Plasmids are extrachromosomal DNA segments
specialized surface proteins such as the M protein that carry genes for antibiotic resistance known as
on Streptococcus pyogenes. R-factors. Multiple drug resistance (R) plasmids
increase the severity of clinical disease by their
c. Resistance to Killing by Phagocytic Cells resist­ance to antibiotic therapy.
Some bacteria have evolved the ability to
survive inside neutrophils, monocytes, and 8. Bacteriophages
macrophages. These pathogens not only survive All the strains of C. diphtheriae produce exotoxin
within macrophages and other phagocytes, only when they are lysogenized with a bacteriophage
but may actually multiply intracellulary, e.g. called betaphage. The elimination of this phage
Mycobacterium tuberculosis. abolishes the toxigenicity of the bacillus.

www.ebook3000.com
80  |  Section 1: General Bacteriology
9. Communicability ™™ Infection: Infections may be classified in various ways.
The ability of a microbe to spread from one host to Sources of infection: A. Human beings; B. Animals; C.
another is known as communicability. Insects; D. Soil and water; E. Food.
Modes of transmission of infection: 1. Contact; 2.
Inhalation; 3. Ingestion; 4. Inoculation; 5. Insects; 6.
10. Infecting Dose Congenital; 7. Iatrogenic and laboratory infections
Adequate number of bacteria is required for ™™ Determinants of virulence: 1. Transmissibility;
successful infections. The dosage may be estimated 2. Adhesion; 3. Invasiveness; 4. Toxigenicity; 5.
as the minimum infecting dose (MID) or minimum Avoidance of host defence mechanisms; Enzymes;
lethal dose (MLD). 7. Plasmids; 8. Bacteriophages; 9. Communicability;
10. Infecting dose; 11. Route of infection
These doses are more correctly estimated as
™™ Types of infectious diseases
statistical expressions, ID50 and LD50 as the dose A. Localized infections may be superficial or deep
required to infect or kill 50 per cent of the animals ­seated
tested under standard conditions. B. Generalized infection: 1. Bacteremia; 2. Septicemia;
3. Pyemia.
11. Route of Infection
Certain bacteria are infective when introduced Important Questions
through optimal route, for example, cholera vibrios
1. Describe in detail the sources of infections to
can produce lesion only when administered by humans beings.
oral route, but unable to cause infection when
2. What are the various modes of spread of infec­tion?
introduced subcutaneously. Describe each in brief giving suitable examples.
3. Describe the factors determining microbial
Types of infectious diseases pathogenicity and virulence.
Infectious diseases may be localized or generalized. 4. Distinguish between exotoxins and endotoxins in
a tabulated form.
A. Localized
Localized infections may be superficial or deep­ Multiple choice questions (MCQs)
seated. 1. The organisms can be transmitted vertically by all
the following ways except:
B. Generalized a. Sexual contact
1. Bacteremia: Circulation of bacteria in the blood b. Through the placenta
c. Within the birth canal
is known as bacteremia.
d. Through breast milk
2. Septicemia: It is the condition where bacteria
2. Which of the following may cause teratogenic
circulate and multiply in the blood, form toxic
infections?
products and cause high, swinging type of fever. a. Toxoplasma
3. Pyemia: It is a condition where pyogenic b. Rubella virus
bacteria produce septicemia with multiple c. Cytomegalovirus
abscesses in internal organs, such as the spleen, d. All of the above
liver and kidney. 3. Which of the following statement is not true for
exotoxins?
Epidemiological terminologies a. They are proteins
b. They are highly antigenic
1. Endemic: The disease which is constantly c. They are heat labile
present in a particular area, e.g. typhoid fever d. They are obtained by cell lysis
is endemic in most parts of India. 4. Which of the statement is not true for endotoxins:
2. Epidemic: The disease that spreads rapidly, a. These are lipopolysacaride components of
involving many persons in a particular area at outer membrane of gram negative bacteria
b. These are heat stable
the same time, is called epidemic disease, e.g.
c. They cannot be toxoided
meningococcal meningitis. d. They are highly antigenic
3. Pandemic: It is an epidemic that spreads through 5. The disease that spreads rapidly, involving many
many areas of the world involving very large persons in a particular area at the same time is
number of persons within a short period, e.g. known as:
cholera, influenza and enteroviral conjunctivitis. a. Sporadic
b. Endemic
Key Points c. Epidemic
™™ Parasites are microbes that can establish themselves d. Pandemic
and multiply in the hosts. Parasite microbes may be
Answers (MCQs)
either pathogens or commensals
1. a; 2. d; 3. d; 4. d; 5. c.
Section 2: Immunology

12
Chapter

Immunity

Learning Objectives

After reading and studying this chapter, you should ∙∙ Differentiate between active and passive immunity.
be able to:
∙∙ Describe innate immunity, artificial active immunity,
natural passive immunity, herd immunity

Definition Innate immunity may be considered at the level of


species, race or individual.
Immunity [Latin immunis, free of burden] refers to
the resistance exhibited by the host towards injury i. Species Immunity
caused by microorganisms and their products. Resistance or susceptibility (lack of resistance) to
infections can vary from one species of animal to
Classification other. It refers to the total or relative refractoriness
to a pathogen, shown by all members of a species.
Immunity against infectious diseases is of different
types: Example
i. Innate (or natural) immunity All human beings are totally unsusceptible to plant
a. Nonspecific Species pathogens and to many animal pathogens, such as
Racial rinderplast or distemper.
Individual ii. Racial Immunity
b Specific Species Within a species, different races may show differences
Racial in susceptibility to infections. This is known as racial
Individual immunity. Such racial differences are known to be
genetic in origin, and by selection and inbreeding.
ii. Acquired (or adaptive) immunity
a. Active Natural Examples
Artificial i. High resistance of Algerian sheep to anthrax is
b. Passive Natural the classic example.
Artificial ii. Susceptiblity to tuberculosis: The people of
Negroid origin in the USA are more susceptible
i. Innate or Natural Immunity than the Caucasians to tuberculosis.
It is the resistance to infections which an individual iii. Genetic resistance to Plasmodium falciparum
possesses by virtue of his genetic or constitutional malaria: It is seen in some parts of Africa and
make up. Repeated exposure to a pathogen does the Mediterranean coast and is attributed to
not enhance the innate immune system. the hereditary abnormality of the red blood
cells (sickling) prevalent in the area.
a. Nonspecific and Specific Immunity iii. Individual Immunity
It may be nonspecific, when it indicates a degree of The difference in innate immunity exhibited
resistance to infections in general, or specific where by different individuals in a race is known as
resistance to a particular pathogen is concerned. individual immunity.

www.ebook3000.com
82  |  Section 2: Immunology
Factors influencing the level of immunity saturated and unsaturated fatty acids that kill many
1. Age bacteria and fungi.
i. Fetus in utero: The two extremes of life
B. Mucous Membrane
carry higher susceptibility to infectious
General protective mechanisms : A major
diseases as compared with adults. The
protective component of mucous membranes is
higher susceptibility of the young appears
the mucus itself.
to associate with immaturity of immune
system. Specific protective characteristics: Besides the
ii. Newborn animals: Newborn animals are general protective properties of mucosal cells,
more susceptible to experimental infection the lining of the different body tracts has other
than adult animals, e.g. coxsackievirus characteristics specific to each anatomic site.
causes fatal infection in suckling mice but
not in adult mice. a. Mouth or Oral Cavity
iii. In the elderly, besides a general waning The mouth or oral cavity is protected by the flow
of the activities of the immune system, of saliva that physically carries microorganisms
physical abnormalities (e.g. prostatic away from the cell surfaces and also contains the
enlargement leading to stasis of urine) lysozyme, which destroys bacterial cell walls and
or long-term exposure to environmental antibodies.
factors (e.g. smoking) are common causes
of increased susceptibility to infection. b. Gastrointestinal Tract
2. Hormonal Influences and Sex i. Stomach: The low pH and proteolytic enzymes
i. Endocrine disorders: There is an increased of the stomach help keep the number of
susceptibility to infection in endocrine micro-organisms low. The pH becomes
disorders, such as diabetes mellitus, progressively alkaline from the duodenum to
hypothyroidism and adrenal dysfunction the ileum.
(increased corticoids secretion).
ii. Small intestine: In the small intestine,
ii. Sex: In general, incidence and death rate protection is provided by the presence of bile
from infectious diseases are greater in males salts.
than in females.
iii. Ileum: The ileum contains a rich and varied
3. Nutrition: In general, both humoral and cell
flora and in the large intestine, the bulk of the
mediated immune processes are reduced
contents is composed of bacteria. Abundant
in malnutrition. Experimental evidence in
resident microflora in the large bowel also
animals has shown that inadequate diet may
contributes significantly to protection.
be correlated with increased susceptibility of
a variety of bacterial diseases, associated with
c. Upper Respiratory Tract
decreased phagocytic activity and leukopenia.
4. Stress: A growing body of evidence has i. Architecture of the nose: In the upper
demonstrated an inverse relation between respiratory tract, nasal hairs keep out large
stress and immune function. The end result is airborne particles that may contain micro-
an increased susceptibility to infection. organisms.
ii. Sticky mucus: The sticky mucus covering the
Mechanisms of Innate Immunity respiratory tract acts as a trapping mechanism
for inhaled particles.
1. Mechanical Barriers and Surface iii. Ciliary motion: Ciliary motion transports the
Secretions trapped organisms back up the respiratory
These surfaces include as follows: tract to the external openings.
A. Skin iv. Cough reflex: Cough reflex is an important
B. Mucous membranes. defence mechanism of the respiratory tract.
A. Skin v. Mucopolysaccharide: Nasal and respiratory
The intact skin and the mucous membranes provide secretions contain mucopolysaccharide
mechanical barriers that prevent the entrance of capable of combining with influenza and
most microbial species. Even though the structure certain other viruses.
of the skin itself undoubtedly gives a great deal of
protection, considerably more important are the d. Genitourinary Tract
fatty acids secreted by the sebaceous glands and i. Normal flow of urine: The normal flow of urine
the propionic acid by the normal flora of the skin. flushes the urinary system, carrying micro-
Secretions from the sebaceous glands contain both organisms away from the body.
Chapter 12: Immunity  | 83
ii. Spermine and zinc: Spermine and zinc present Phagocytosis: Phagocytosis is apart of the innate
in the semen carry out antibacterial activity. immune response, during which microorganisms,
iii. Acidity of the adult vagina: The low pH (acidity) foreign particles, and cellular debris are engulfed
of the adult vagina, due to fermentation of by phagocytic cells, such as neutrophils and
glycogen in the epithelial cells by the resident monocytes in the circulation, and macrophages
aciduric bacilli, provides an inhospitable and neutrophils in interstitial spaces. Phagocytosis
environment for colonization by pathogens. consists of three stages:
A thick mucus plug in the cervical opening is 1. Attachment of the microorganisms or foreign
a substantial barrier. material to the phagocytic cell.
2. Ingestion by the cell and drawn into the cell by
e. Conjunctiva endocytosis.
3. Degradation of the foreign material or micro-
Lachrymal fluid: Conjunctiva is kept moist by the
organism within the phagocytic cells.
continuous flushing action of tears (lachrymal
fluid). Tears contain large amounts of lysozyme,
lactoferrin, and sIgA and thus provide protection.
5. Inflammation
If the surface chemical and physiologic def­
ences of the body are breached by a pathogen,
2. Antibacterial Substances in Blood and inflammation can result, which is an
Tissues important, nonspecific defence mechanism.
Many microbial substances are present in the Sequences of events in acute inflammation in
tissue and body fluids. These are non-specific. The response to an injury will be: (i) vasodilation,
complement system possesses bactericidal activity (ii) increased vascular permeability; (iii) emigration
and plays an important role in the destruction of of leucocytes; (iv) chemotaxis; (v) phagocytosis.
pathogenic bacteria that invade the blood and
tissues. 6. Fever
Other Substances (i) beta lysin; (ii) basic Following infection, a rise of temperature is a
polypeptides, such as leukins extracted from natural defense mechanism. It not merely helps
leucocytes and plakins from platelets; (iii) acidic to accelerate the physiological processes, but
substances, such as lactic acid found in muscle may, in some cases, actually destroy the infecting
tissue and in the inflammatory zones; and pathogens. Fever aids recovery from viral infections
(iv) lactoperoxidase in milk. The production of by stimulating the production of interferon.
interferon is a method of defence against viral
infections. 7. Acute Phase Proteins
A sudden increase in the plasma concentration
3. Microbial Antagonisms of certain proteins, collectively termed as ‘acute
The skin and mucous surfaces have resident phase proteins’ occurs as a result of infection or
bacterial flora which prevent colonization by tissue injury. These include C-reactive protein
pathogens. Invasion by extraneous microbes (CRP), mannose-binding protein, alpha-I-acid
may be due to alteration of normal resident flora, glycoprotein, serum amyloid P component
causing serious diseases, such as staphylococcal and many others. The alternative pathway of
or clostridial enterocolitis or candidiasis following complement is activated by CRP and some other
oral antibiotics. acute phase proteins. They are believed to enhance
host resistance, prevent tissue injury and promote
repair of inflammatory lesions.
4. Cellular Factors in Innate Immunity
Natural defense against the invasion of blood ii. Acquired Immunity
and tissues by microorganisms and other foreign
Acquired immunity refers to the resistance that an
particles is mediated to a large extent by phagocytic
individual acquires during his lifetime. Acquired
cells which ingest and destroy them. Phagocytic
immunity is of two- types: active immunity and
cells, originally discovered by Metchnikoff
passive immunity. (Fig. 12.1 and Table 12.1).
(1883), were classified by him into microphages
(polymorpho­nuclear leucocytes) and macrophages.
a. Active Immunity
Macrophages: Macrophages consist of histiocytes Active immunity is induced after contact with
which are the wandering ameboid cells seen foreign antigens. It is also known as adaptive
in tissues, fixed re­t iculoendothelial cells and immunity. This involves the active functioning
monocytes of blood. of the host’s immune apparatus leading to the

www.ebook3000.com
84  |  Section 2: Immunology
In some infections like syphilis and malaria,
A. Active immunity
the immunity to reinfection lasts only as long as
1. Natural—Clinical
the original infection remains active. Once the
– Subclinical Infection
disease is cured, the patient becomes susceptible to
2. Artificial
the infection again. This special type of immunity
Vaccination—Live
is known as ‘premunition’ or infection-immunity.
– Killed
B. Passive immunity 2. Artificial Active Immunity
1. Natural—Through placenta Artificial active immunity is the resistance induced
– Through breast milk by vaccines. Vaccines are preparations of live or
2. Artificial—Immune serum killed microorganisms or their products used for
– Immune cells immunization. Vaccines are made with either:
Fig. 12.1: Types of immunity
(i) live attenuated microorganisms; (ii) killed
microorganisms; (iii) microbial extract; (iv) vaccine
conjugates; or (v) inactivated toxoids.
synthesis of antibodies and/or the production of
immunologically active cells. Examples of Vaccines
Immune Response I. Bacterial vaccines
a. Live (BCG vaccine for tuberculosis)
A. Primary Response b. Killed (cholera vaccine)
Active immunity sets in only after a latent period. c. Subunit (typhoid Vi antigen)
There is often a negative phase. Once developed, d. Bacterial products (tetanus toxoid)
the active immunity is long-lasting. 2. Viral vaccines
a. Live
B. Secondary Response –– Oral polio vaccine-Sabin
If an individual, who has been actively immunized –– 17D vaccine for yellow fever
against an antigen, experiences the same antigen –– MMR vaccine for measles, mumps, rubella.
subsequently, the immune response occurs more b. Killed
quickly and abundantly than during the first –– Injectable polio vaccine-Salk
encounter. This is known as secondary response. –– Neural and non-neural vaccines for rabies
–– Hepatitis B vaccine
Types of Active Immunity c. Subunit: Hepatitis B vaccine
1. Natural Live Vaccines
2. Artificial In general, live vaccines are more potent immunizing
agents than killed vaccines The immunity lasts for
1. Natural Active Immunity
several years but booster doses may be necessary.
Natural active immunity results from either a
Live vaccines may be administered orally or
clinical or an inapparent infection by a microbe.
parenterally.
Such immunity is usually long-lasting but the
duration varies with the type of pathogen. The Killed Vaccines
immunity is life-long following many viral diseases, Killed vaccines are usually safe and generally less
such as chickenpox or measles. immunogenic than live vaccines, and protection
The immunity appears to be shortlived in some lasts only for a short period. They have, therefore,
viral diseases, such as influenza or common cold. to be administered repeatedly, generally at least

Table 12.1  Comparison of active immunity and passive immunity


Active immunity Passive immunity
1. Produced actively by host’s immune system 1. Received passively. No active host participation
2. Induced by infection or by immunogens 2. Readymade antibody transferred
3.  Durable effective protection 3.  Transient, less effective
4. Immunity effective only after lag period, i.e. time required for 4.  Immediate immunity
generation of antibodies and immunocompetent cells
5. Immunological memory present 5.  No memory
6. Booster effect on subsequent dose 6. Subsequent dose less effective
7.  ’Negative phase’ may occur 7.  No negative phase
8. Not applicable in an immunodeficient 8. Applicable in an immunodeficient
Chapter 12: Immunity  | 85
two doses being required for the production of patients who have recovered recently from an
immunity. The first is known as the primary dose infection or are obtained from individuals who
and the subsequent doses as booster doses. Killed have been immunized against a specific infection.
vaccines are usually administered by subcutaneous Preparations of specific immunoglobulins are
or intramuscular route. available for passive immunization against
tetanus (human tetanus immunoglobulin;
b. Passive Immunity HTIG), hepatitis B (HBIG), rabies (HRIG),
varicella-zoster (ZIG) and vaccinia (AVIG).
The immunity that is transferred to a recipient in Human immune serum does not lead to any
a ‘readymade’ form is known as passive immunity. hypersensitivity reaction. Therefore, there is no
Here the recipient’s immune system plays no active immune elimination and its half-life is more than
role. There is no lag or latent period in passive that of animal sera. It has to be ensured that all
immunity, protection being effective immediately preparations from human sera are free from the
after passive immunization. There is no negative risk of human immunodeficiency virus (HIV),
phase. The immunity is transient. There is no hepatitis B, hepatitis C and other viruses.
secondary type response in passive immunity.
B. Nonhuman (Antisera)
Main Advantages of Passive Immunity The term aniserum is applied to materials
i. The prompt availability of large amount of prepared in animals. Equine hyperimmune sera,
antibody. such as antitetanus serum (ATS) prepared from
ii. It is employed where instant immunity is hyperimmunized horses used to be extensively
required as in case of diphtheria, tetanus, employed. They gave temporary protection but
botulism, rabies, hepatitis A and B following disadvantage is that it may give rise to hypersensitivity
exposure because of its immediate action. and immune elimination.

Passive immunity is of two types Indications of Passive Immunization


1. Natural Passive Immunity 1. To provide immediate protection : To a
This is the resistance passively transferred from nonimmune host exposed to an infection and
mother to baby through the placenta. After birth, lack active immunity to that pathogen.
immunoglobulins are passed to the newborn 2. Treatment of some infections.
through the breast milk. The human colostrum, 3. For the suppression of active immunity when
is rich in IgA antibodies which are resistant to it may be injurious e.g. administration of
intestinal digestion, gives protection to the neonate anti-Rh (D) IgG to Rh-negative mother, bearing
up to three months of age. Rh-positive baby at the time of delivery to
prevent isoimmunization.
2. Artificial Passive Immunity 4. Immunocompromised or immunodeficient
Artificial passive immunity is the resistance passively individuals, e.g. children with hypogamma­
transferred to a recipient by the administration globulin­emia, individuals with AIDS, patients
of antibodies. Although this type of immunity is receiving chemotherapy, organ transplant
immediate, it is short lived and lasts only a few weeks recipients receiving immunosuppressive therapy.
to a few months. The agents used for this purpose
are pooled human gamma globulin, hyperimmune
Combined Immunization
sera of animal or human origin and convalescent
sera . These are used for prophylaxis and therapy. Combined immunization is a combination of
active and passive methods of immunization and
Types of Immunoglobulin Preparations sometimes employed. For example, it is often
Two types of immunoglobulin preparations are undertaken in some diseases, such as tetanus,
available for passive immunization. diphtheria, rabies.
A. Human Immunoglobulins
a. Human normal immunoglobulin: Human
Adoptive Immunity
normal immunoglobulin(HNIG) is used to Injection of immunologically competent
provide temporary protection against hepatitis lymphocytes is known as adoptive immunity
A infection and to prevent measles in highly and does not have general application. Instead of
susceptible individuals. whole lymphocytes, an extract of immunologically
b. Specific (hyperimmune) human immunoglobulin: competent lymphocytes, known as the ‘transfer
These preparations are made from the plasma of factor’, can be used.

www.ebook3000.com
86  |  Section 2: Immunology
Local immunity ™™ Combined immunization is a combination of active
and passive methods of immunization which is
Local immunity is conferred by secretory sometimes employed
immunoglobulin A (secretory IgA) produced ™™ Local immunity is conferred by secretor y
locally by plasma cells present on mucosal surfaces immunoglobulin A (secretory IgA)
or in secretory glands. ™™ Herd immunity is the level of resistance of a
community or a group of people to a particular
disease and is relevant in the control of epidemic
Examples diseases.
1. Poliomyelitis immunization: In poliomyelitis,
active immunization provides systemic Important questions
immunity with the killed vaccine. Natural
1. Define and classify immunity. Discuss mechanisms
infection or immunization with the live oral of innate immunity.
vaccine provides local intestinal immunity. 2. Tabulate the differences between active immunity
2. Influenza, immunization: Natural infection and passive immunity.
or the live influenza vaccine administered 3. Write short notes on:
intranasally provides local immunity. a. Innate immunity
b. Artificial active immunity
c. Natural passive immunity
Herd immunity d. Artificial passive immunity
It is the level of resistance of a community or e. Herd immunity.
a group of people to a particular disease and
is relevant in the control of epidemic diseases. Multiple choice questions (MCQs)
Epidemics are likely to occur on the introduction 1. All are acute phase proteins except:
of a suitable pathogen, when herd immunity is low. a. C-reactive protein
b. Mannose-binding protein
Eradication of communicable diseases depends on
c. Serum amyloid P component
the development of a high level of herd immunity d. Antibody
rather than on the development of a high level of 2. Clinical or inapparent infection leads to:
immunity in individuals. a. Natural active immunity
b. Artificial active immunity
Key Points c. Natural passive immunity
d. Artificial passive immunity
™™ Immunity refers to the resistance exhibited by the 3. Vaccine induces:
host towards injury caused by microorganisms and a. Active natural immunity
their products b. Active artificial immunity
™™ Innate or natural immunity is the resistance to c. Passive natural immunity
infections, which an individual possesses by virtue d. Passive artificial immunity
of his genetic or constitutional make-up. It may be 4. All the following statements are true for artificial
nonspecific, or specific. Innate immunity may be passive immunity except:
considered at the level of species, race or individual. a. Artificial passive immunity is the resistance
™™ Factors influencing the level of immunity are age, passively transferred to a recipient by the ad-
hormonal influences and sex, nutrition and stress ministration of antibodies.
™™ Mechanisms of innate immunity are: 1. Mechanical b. It is shortlived and lasts only a few weeks to a
barriers and surface secretions; 2. Antibacterial few months
substances in blood and tissues; 3. Microbial c. This type of immunity is immediate
antagonisms; 4. Cellular factors in innate immunity; d.  This immunity may be induced by maternal anti-
5. Inflammation; 6. Fever; 7. Acute phase proteins bodies.
™™ Acquired immunity refers to the resistance that an 5. All the following are live vaccines except:
individual acquires during his lifetime. b-Acquired a. BCG b. Sabin vaccine
immunity is of two types: active immunity and c. MMR d. TAB vaccine
passive immunity 6. All the following are killer vaccines except:
™™ Natural active immunity results from either a clinical a. Salk vaccine
or an inapparent infection by a microbe b. Non-neural vaccines for rabies
™™ Artificial active immunity is the resistance induced c. Hepatitis B vaccine
by vaccines d. BCG
™™ The immunity that is transferred to a recipient in
a ‘readymade’ form is known as passive immunity
Answers (MCQs)
1. d; 2. a; 3. b; 4. d; 5. d; 6. d
13
Chapter

Antigens

Learning Objectives

After reading and studying this chapter, you should


be able to:
∙∙ Describe haptens, heterophile antigens and
superantigens.

Antigens: Antigens (antibody generators) are the 2. Complex haptens: Complex haptens can
substances that can stimulate an immune response precipitate with specific antibodies. Complex
and, given the opportunity, react specifically by haptens are polyvalent.
binding with the effector molecules (antibodies)
and effector cells (lymphocytes). Antigenic determinant or epitome
The smallest unit of antigenicity is known as the
Types of antigens antigenic determinant or epitope. Each antigen
can have several antigenic determinant sites or
Based on the ability of antigens to carry out these two
epitopes. The combining area on the antibody
functions, they may be classified into different types:
molecule, corresponding to the epitope, is called
the paratope.
A. Complete Antigen
Complete antigen is able to induce antibody Valence: The number of antigenic determinant
formation and produce a specific and observable sites on the surface of an antigen is its valence. The
reaction with the antibody so produced. antigen is monovalent or multivalent.

B. Haptens (Incomplete Immunogen) Determinants of antigenicity


Haptens are low-molecular- weight molecules which A number of properties have been identified which
cannot induce an immune response when injected by make a substance antigenic but the exact basis of
themselves but can do so when covalently coupled to antigenicity is still not clear.
a large protein molecyle called the carrier molecule.
Example: One example of a hapten is penicillin. By 1. Size
itself, penicillin is not antigenic. However, when it Molecular Weight
combines with certain serum proteins of sensitive
Antigenicity depends upon the molecular weight.
individuals, the resulting molecule does initiate a
Very large molecules are very powerful antigens.
severe and sometimes fatal allergic immune reaction.
Particles with low molecular weight (less than
5000) are nonantigenic or feebly so.
Types of Haptens
Haptens may be simple or complex:
1. S i m p l e h a p t e n s : S i m p l e ha p t e n s a re 2. Chemical Nature
nonprecipitating. They can inhibit precipitation In general, proteins are the best immunogens and
of specific antibodies by the corresponding carbohydrates are weaker immunogens. Lipids and
antigen or complex hapten. nucleic acids are poor immunogenic.

www.ebook3000.com
88  |  Section 2: Immunology
3. Foreignness associated with plasma membrane of tissue
Only antigens which are ‘foreign’ to the individual cells. These antigens are encoded by genes
(nonself ) induce an immune response because known as histocompatibility genes which
host distinguishes self from nonself and normally collectively constitute major histocompatibility
does not respond to self. In general, the greater the complex (MHC). MHC products present on the
difference between the antigen (Ag) and similar surface of leucocytes are known as human
molecules in the host’s body, the greater the leukocyte-associated (HLA) antigens.
immune response that is generated.
8. Autospecificity
4. Susceptibility to Tissue Enzymes Sequestrated Antigens
Only those substances which can be metabolized Autologous or self-antigens are ordinarily
and susceptibility to the tissue enzymes behave as nonantigenic but there are exceptions. Certain
antigens. Substances unsusceptible to the tissue self-antigens are present in closed system and are
enzymes are not antigenic. Substances that are not accessible to the immune apparatus and these
insoluble in body fluid and not metabolized are not are known as sequestrated antigens.
antigenic. Substances very rapidly broken down by
tissue enzymes are also not antigenic. Examples of Sequestrated Antigens
5. Antigenic Specificity i. Lens protein: Sequestrated antigens that are
not normally found free in circulation or tissue
Chemical Groupings fluids (such as lens protein normally confined
Foreignness of a substance to an animal can within its capsule) are not recognized as
depend on the presence of chemical groupings self-antigens. When the antigen leaks out
that are not normally found in the animal’s body. following penetrating injury, it may induce
Antigenic specificity varies with the position of an immune response causing damage to the
antigenic determinant, i.e. whether it is in ortho, lens of the other eye.
meta or para position. ii. Sperm: Similarly, antigens that are absent
In many cases, an antibody specific for one during embryonic life and develop later (such as
antigen may display significant cross-reactivity for sperm), are also not recognized as self-antigens.
an apparently unrelated antigen. When the sperm antigen enters the circulation,
it is immunogenic and is believed to be the
6. Species Specificities pathogenesis of orchitis following mumps.
Tissues of all individuals in a species possess
9. Organ Specificity
species-specific antigens. However, some degree
of cross-reactivity is seen between antigens from Some organs, such as brain, kidney and lens protein
related species. of different species share the same antigens. These
antigens are known as organ-specific antigens,
7. Isospecificities characteristic of an organ or tissue and found in
Isoantigens or alloantigens are antigens found different species. Injection of heterologous organ-
in some but not all members of a species. On the specific antigens may induce an immune response,
basis of isoantigens, a species may be divided into damaging the particular organs or tissue in the host.
different groups.
Example
Examples of Isoantigens Neuroparalytic complications: The neuroparalytic
i. Human erythrocyte antigens: The best complications following antirabic vaccination
example of isoantigens is human erythrocyte using sheep brain vaccines are a consequence of
antigens, on the basis of which all humans can brain-specific antigens shared by sheep and man.
be divided into different blood groups: A, B, The sheep brain antigens induce an immunological
AB and O. Each of these groups may be further response in the vaccines, damaging their nervous
divided into Rh-positive or Rh-negative. tissue due to the cross-reaction between human
and sheep brain antigens.
ii. Histocompatibility antigens: Histocompatibility
antigens are those cellular determinants
specific to each individual of a species. 10. Heterogenetic (Heterophile) Specificity
Histocompatibility typing is essential in organ/ Same or closely related antigens occurring in
tissue transplantation from one individual to different biological species, classes and kingdoms
another within a species. These antigens are are known as heterogenetic or heterophile antigens.
Chapter 13: Antigens  | 89
Examples of Heterophile Antigens 2. Association with diseases: Superantigens should
i. Forssman antigen: It is a lipid carbohydrate be considered possible chronic associates in
complex widely distributed in man, animals, such diseases as rheumatic fever, arthritis,
birds, plants and bacteria. Kawasaki syndrome, atopic dermatitis, and one
ii. Weil-Felix reaction: It is an agglutination test type of psoriasis.
in which patient sera are tested for agglutinins
to the O antigens of certain nonmotile Proteus Key Points
strains OXl9, OX2, and OXK. Cross-reaction
between O antigen of these strains of Proteus ™™ Antigens are the substances that can stimulate an
immune response and, given the opportunity, react
and certain rickettsial antigens is the basis of
specifically by binding with the effector molecules
this test. (antibodies) and effector cells (lymphocytes)
iii. Paul-Bunnell test: During infectious-mono­ ™™ Types of antigen are: A. Complete antigen; B.
nucleosis, heterophile antibodies appear in Haptens (incomplete immunogen)
the serum of the patient. These antibodies ™™ Determinants of antigenicity are: 1. Size; 2.
agglutinate sheep erythrocytes. This test is Chemical nature; 3. Foreignness; 4. Susceptibility to
known as Paul-Bunnell test. tissueenzymes; 5. Antigenic specificity; 6. Species;
iv. Cold agglutinin test: Agglutination of human O specificities; Isospecifities; 8. Autospecicity; 9. Organ
specificity; 10. Heterogenetic (heterophile) specificity.
group erythrocytes at 4°C by the sera of patients
suffering from primary atypical pneumonia.
v. Agglutination of Streptococcus MG: Agglutin­
Important questions
ation of Streptococcus MG by the sera of the
patients of primary atypical pneumonia. 1. What is an antigen? Discuss briefly various deter­
minants of antigenicity.
Superantigens 2. Write short notes on:
a. Haptens
Superantigens are bacterial proteins which can
b. Heterophile antigens
inter­act with antigen-presenting cells (APCs) and
T cells in nonspecific manner. This activity does
not in­volve the endocytic processing required for Multiple choice questions (MCQs)
typical antigen presentation but instead occurs
1. The substances that are least immunogenic are
by concurrent association with MHC class II
a. Proteins
molecules of the APCs and the Vβ domain of the
b. Polysaccharides
T cell receptor. This interaction activates a large
c. Nucleic acids
number of T cells (10%) than conventional anti­
d. None of the above
gens (about 1%), explaining the massive cytokine
expression and immunomodulation. The antigens 2. The test that does not use heterophilic antigen is
which provoke such a drastic immune response are a. TPHA test
termed as superantigens. b. Weil-Felix reaction
c. Paul-Bunnell test
Examples d. Cold agglutination test

1. Staphylococcal enterotoxins, toxic shock syndrome Answers (MCQs)


toxin, exfoliative toxin and some viral proteins. 1. c; 2. a

www.ebook3000.com
14
Chapter

Antibodies—Immunoglobulins

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe structure and functions of IgG, IgA and
be able to: IgM
∙∙ Define antibody and draw labeled diagram of ∙∙ Discuss properties of IgM, IgG, IgA, IgD and IgE
immunoglobulin ∙∙ Draw labeled diagram of IgG, IgM and IgA.

Antibody or immunoglobulin (Ig): An antibody immunoglobulin molecule, which have led to a


or immunoglobulin (Ig) is a glycoprotein that is detailed picture of its structure. Rabbit IgG antibody to
made in response to an antigen, and can recognize egg albumin was digested by papain in the presence
and bind to the antigen that caused its production. of cysteine. Each molecule of immunoglobulin is
Immunoglobulins provide a structural and split by papain into three parts (Fig. 14.1). Two of
chemical concept, while the term ‘antibody’ is a the fragments are identical and are referred to as
biological and functional concept. All antibodies Fab (Fragment antigen-binding) because they retain
are immunoglobulins, but all immunoglobulins the immunoglobulin’s ability to bind specifically
may not be antibodies. to an antigen. The third fragment Fc (Fragment
crystallizable), which can be crystallized, does not
Properties of antibodies bind antigen, but contributes to the biologic activity.
1. Electrophoretic mobility: Serum proteins can be
separated into soluble albumins and insoluble Pepsin Digestion
globulins. Globulins could be separated into When treated with pepsin, a 5S fragment is
water-soluble pseudoglobulins and insoluble obtained, which is composed essentially of two Fab
euglobulins. Most antibodies were found to fragments held together in position. It is bivalent
be euglobulins. Serum glycoproteins can and precipitates with the antigen. This fragment is
be separated according to their charge by called F(ab)2. The Fc portion is digested by pepsin
movement through a gel in an electric field and into smaller fragments (Fig. 14.1).
classified as albumin and globulin (alpha-l,
alpha-2, beta and gamma globulin).
2. Sedimentation and molecular weight: Most
antibodies are sedimented at 7S (MW 150,000–
180,000) based on sedimentation studies. Some
heavier anti­bodies—19S globulins (MW about
900,000) were designated as M or macroglobulins.
3. Physicochemical and antigenic structure: On the
basis of physicochemical and antigenic structure,
Igs can be divided into five distinct classes or iso­
types, namely IgG, IgA, IgM, IgD and IgE.

ANTIBODY STRUCTURE
Porter, Edelman, Nisonoff and their colleagues Fig. 14.1: Basic structure of an immunoglobulin molecule and
poineered studies involving the cleavage of the the fragments obtained by the cleavage by papain and pepsin
Chapter 14: Antibodies—Immunoglobulins  | 91

A B
Fig. 14.2: The four-peptide chain structure of the IgG molecule composed of two identical heavy (H) and two identical light
(L) chains linked by interchain disulfide bonds. Loops formed by intrachain disulfide bonds are domains (shown stippled).
Each chain has one domain in the variable region (VH and VL). Each light chain has one domain in the constant region (CL)
while each heavy chain has three domains in the constant region (CH1, CH2 and CH3). Between CH1 and CH2 is the hinge region

Light and Heavy Chains Table14.1  Immunoglobulin classes and H-chains


All immunoglobulins are composed of the same Immunoglobulin class H-chain
basic units consisting of four chains: two identical IgG gamma (g)
‘light’ (L) chains and two identical heavy chains. The
IgA alpha (α)
L-chain is attached to the H-chain by a disulfide
bond. The two H-chains are joined together by 1–5 IgM mu (μ)
disulfide (S-S) bonds, depending on the class of IgD delta (δ)
immunoglobulins (Fig. 14.2). The smaller chains IgE epsilon (ε)
are called ‘light’ (L) chains and the larger ones
‘heavy’ (H) chains. The variable region contains In humans, there are five classes of heavy chains
the antigen-binding site; the constant region designated by lowercase Greek letters: gamma (g),
encompasses the entire Fc (fragment crystallizable) alpha (a), mu (μ), delta (d) and epsilon (e).
region as well as part of the Fab (fragment antigen-
binding) region. 3. Constant and Variable Regions
All immunoglobulin chains possess a constant (“C”)
1. Classes of L-chains
region that determines the biologic action of an
The L-chains are similar in all classes of immuno­ antibody, and a variable (“V”) region that binds a
globulins. The L-chain has a molecular weight of unique epitope. Both light and heavy chains contain
approximately 25,000 and the H chain of 50,000. two different regions. Of the 220 amino acids, those
There are two classes of L-chains, designated which constitute carboxy-terminal half of L-chain,
kappa (κ) and lambda (l). Both are normally occur in a constant sequence. This part of the chain
expressed in every individual. However, each B-cell is called constant regions (CL).
expresses (and each antibody contains) only one
The H-chain also has constant region (CH) and
type of L-chain—either κ or l but not both. Kappa
variable regions (VH). While in the L-chain, the
(κ) and lambda (l) are named after Korngold and
two regions are of equal length, in the H-chains,
Lapari who originally described them. Kappa (κ)
the variable region constitutes approximately
and lambda (l) chains occur in a ratio of about 2 :
only a fifth of the chain and is located at its
1 in human sera.
aminoterminus. The variable regions (V L and
VH) from different antibodies do have different
2. Classes of H-chains sequences. It is the variable regions (VL, VH) that
The H-chains are structurally and antigenically when folded together form the antigen-binding
distinct for each class and are designated by the Greek sites.The other domains of the heavy chains are
letter corres­ponding to the immunoglobulin class as termed-constant domains and are numbered
shown in Table 14.1. CH1, C H2, C H3, and sometimes C H4, starting with

www.ebook3000.com
92  |  Section 2: Immunology
the domain next to the variable domain. It is the
portion of H-chains present in Fab fragment.
H-chains carry a carbohydrate moiety, which is
distinct for each class of immunoglobulins.

4. Fc Fragment
The Fc fragment is composed of the carboxyterminal
portion of the H-chains. It can be crystallized, and
is, therefore, called Fc (fragment-crystallizable).

Functions of Fc
Fig. 14.3: Secretory IgA. 1. Heavy chain; 2. Light chain; 3. J
Binds complement leading to complement fix­ chain; 4. Secretory component; 5. Disulfide bond
ation.
∙∙ Binds to cell receptors (FcRs) fluid, and it is distributed nearly equally
∙∙ Determines passage of IgG across the placen­tal between extra- and intravascular spaces.
barrier 7. Four subclasses of IgG (Ig1, Ig2, Ig3, Ig4) have
∙∙ Determines skin fixation and catabolic rate. been recognized.
8. Catabolism of IgG is unique in that it varies with
5. Immunoglobulin Domain its serum concentration. When its level is raised,
Each immunoglobulin peptide chain has internal as in chronic malaria, kala-azar or myeloma,
disulfide links in addition to interchain disulfide the IgG synthesized against a particular antigen
bonds which bridge the H- and L-chains. These will be catabolized rapidly and may result in
interchain disulfide bonds form loops in the peptide the particular antibody deficiency. Conversely,
chain, and each of the loops is compactly folded in hypogammaglobulinemia, the IgG given for
to form a globular domain, each domain having treatment will be catabolized only slowly.
a separate function. The variable region domains, Functions of IgG: IgG is a very versatile molecule.
VL and VH, are responsible for the formation of a It may be considered a general-purpose antibody,
specific antigen-binding site. The CH2 region in IgG protective against those infectious agents, which
binds C1q in the classical complement sequence, are active in the blood and tissues.
and the CH3 domain mediates adherence to the
monocyte surface. The areas of the H-chain in the Examples of its Functions
C-region between the first and second C-region i. Transfer from mother to fetus: IgG is the only
domains (CH1 and CH2) is the hinge region. It is class of Igs that can cross the placenta and is
more flexible and is more exposed to enzymes and responsible for the protection of the infant
chemicals. Papain acts here to produce one Fc and during the first few months of life.
two Fab fragments (Figs 14.3). ii. Opsonization: IgG binds to microorganisms
and enhances their phagocytosis.
IMMUNOGLOBULIN CLASSES iii. Fixing to guinea pig skin.
Human serum contains five classes of immuno­ iv. Immunological reactions: IgG participates
globulins—IgG, IgA, IgM, IgD and IgE in the in complement fixation, precipitation and
desending order of the concentration. Table14.2 neutralization of toxins and viruses.
shows their differentiating features. v. Immobilize bacteria.
vi. Suppresses the homologous antibody synthesis:
1. Immunoglobulin G (IgG) ( Fig. 14.3) Passively administered IgG suppresses the
1. This is the major immunoglobulin in human homologous antibody synthesis by a feedback
serum, accounting for about 80% of the total process.
immunoglobulin pool.
2. It has a sedimentation coeffient of 7S and a 2. Immunoglobulin A (IgA)
molecular weight of 150,000. 1. It is the second most abundant class, cons­
3. It contains less carbohydrate than other tituting about 10–13 per cent of serum immuno­
immunoglo­bulins. globulins.
4. The normal serum concentration of IgG is 2. The normal serum level is 0.6–­4.2 mg per mL.
about 8-16 mg per mL. 3. It has a half life of 6–8 days.
5. It has a half-life of 23 days—the longest of all of 4. IgA is the primary immunoglobulin found in
the immunoglobulin isotypes. external secretions, such as mucus, tears, saliva,
6. IgG is the predominant immunoglobulin in gastric fluid, colostrum and sweat. It exists in
blood, lymph, peritoneal fluid and cerebrospinal different forms in these various solutions.
Chapter 14: Antibodies—Immunoglobulins  | 93
Table14.2  Physical, physiologic, and biologic properties of human serum immunoglobulins
Property lgG IgA* lgM lgD IgE
A. Physical properties

1. Sedimentation coefficient(s) 7 7 19 7 8
2. Molecular weight in 150,000 160,000 900,000, 180,000 190,000
kilodaltons
3.  Carbohydrate (%) 3 8 12 13 12
4. Number of four-chain units 1 1–3 5–6 1 1
per molecule
B. Physiologic properties
1. Normal adult serum 12 2 1.2 0.03 0.00004
concentration (mg/mL)
2.  Half-life (in days) 23 6 5 2–8 1–5
4.  Daily production (mg/kg) 34 24 3.3 0.4 0.0023
5. Intravascular distribution (%) 45 42 80 75 50
C. Biologic properties

1. Complement fixation
Classical ++ – +++ – –
Alternative – + – – –
2. Placental transport to fetus + – – – –
3.  Present in milk + + – – –
4. Selective selection by – + – – –
submucus glands
5. Anaphylactic – – – – ++++
hypersensitivity
6.  Heat stability + + + + –
D. Major characteristics Most Protects Very efficienct Mainly Initiates
abundant Ig; mucosal against lymphocyte inflammation;
Longest half surfaces bacteremia receptor; raised in helminth
life: Crosses major surface infection; causes
placenta components of allergy symptoms
opsonizes B cells
antigen
*
IgA may occur in 7S, 9S and 11S forms

5. IgA occurs in two forms. Serum IgA and The secretory piece is believed to protect IgA
secretory IgA (SIgA). from denaturation by bacterial proteases in sites
Serum IgA: Serum IgA is monomeric (one four- such as the intestinal mucosa which have a rich
chain unit) 7S molecule (MW about 160,000). and varied bacterial flora.
Subclasses: There are two subclasses of IgA in
Secretory IgA (SIgA): In contrast, IgA found on
humans: IgA1 and IgA2.
mucosal surfaces and in secretions is a dimer
formed by two monomer units joined together at Functions of IgA
their carboxy terminals by a glycopeptide termed
i. Local immunity: Secretory IgA (SIgA) is
as the J chain (J for joining). This dimeric form
believed to play an important role in local
is more important form, known as secretory IgA
immunity against respiratory and intestinal
(SIgA). Dimeric SIgA is synthesized by plasma
pathogens.
cells situated near the mucosal or glandular
epithelium. J chains are also found in other ii. Prevention of organisms’ entry into body tissues.
polymeric immunoglobulins such as IgM. iii. Newborn protection: IgA present in breast milk
provides the newborn with protection against
Secretory component: Secretory IgA (SIgA)
infection.
contains another glycine-rich polypeptide called
the secretory component or secretory piece. This is iv. Agglutination.
not produced by lymphoid cells but by mucosal or v. Alternative pathways activation.
glandular epithelial cells. vi. Phagocytosis and intracellular killing.

www.ebook3000.com
94  |  Section 2: Immunology
3. Immunoglobulin M (IgM) 4. Immunoglobulin D (IgD)
1. About 10% of normal serum Igs consists of this 1. IgD has a monomer structure similar to IgG.
class. 2. Its molecular weight is 180,000 daltons.
2. It is a heavy molecule (19S; MW 900,000 to 4. IgD is an immunoglobulin found in trace
1,000,000 daltons, hence called ‘millionaire amounts in the blood serum (0.03 mg/mL).
molecule’). 5. Half-life is about 3 days.
3. The normal serum level of IgM is 1.2 mg/mL. 6. IgD antibodies are abundant in combination
4. It has a half-life of about 5 days. with IgM on the surface of B cells and bind
5. IgM is the first immunoglobulin to appear antigens, thus signaling the B cell to start
after exposure to an antigen. antibody production.
6. In the circulation, IgM exists as a pentamer 7. Two subclasses of IgD (IgD1 and IgD2) are known.
of five four-chain units. The five identical IgM
monomers are connected to each other by 5. Immunoglobulin E (IgE)
a polypeptide joining (J) chain (Fig. 14.4). 1. It resembles IgG structurally and also known
Polymerization of the subunits depends upon as reagin antibody.
the presence of the J chain as with IgA (Fig. 14.3). 2. IgE is an 8S molecule (MW 19,000) and half-
7. Most of IgM (80%) is intravascular in life of two days.
distribution. 3. It is present in extremely low amounts in serum.
8. IgM is the first class of antibody produced 4. It exhibits unique properties such as heat
during the primary immune response. It is also lability (inactivated at 56°C in one hour).
the earliest to be synthesized by fetus beginning 5. It is susceptible to mercaptoethanol.
by about 20 weeks of age. As it cannot cross 6. It does not pass the placental barrier.
the placental barrier, the presence of IgM in 7. IgE does not activate complement nor
the fetus or newborn indicates intrauterine agglutinate antigens.
infection. 8. Allergic reactions: IgE may be elevated in allergic
9. They are relatively shortlived, hence their (atopic) individuals, and is responsible for
demonstr­ation in the serum indicates recent many of the symptoms of allergies, bronchial
infection. asthma, and even systemic anaphylaxis.
10. Treatment of serum with 0.12 M 2-mercaptoe­ Allergy mediated by IgE is termed as type I
thanol selectively destroys IgM without hypersensitivity response.
affecting IgG antibodies. 9. Immunity against helminthic parasites:
11. Isohemagglutinins (anti-A and anti-B) and anti­ Children living in insanitary conditions, with
bodies to S. Typhi O antigen and Wassermann a high load of intestinal parasites, have high
reaction antibodies in syphilis are usually IgM. serum levels of IgE.
12. IgM agglutinates bacteria activates complement 10. Extravascular: It is mostly found extra­
by the classical pathway, and enhances the vascularly in the lining of the respiratory and
ingestion of pathogens by phagocytic cells. IgM intestinal tracts.
is normally restricted to the intravascular space 11. Protection against pathogens: The physiological
because of its high molecular weight. role of IgE appears to be protection against
pathogens by mast cell degranulation and
release of inflammatory mediators.

Role of Different Immunoglobulin Classes


IgG: Protects the body fluids
IgA: Protects the body surfaces
IgM: Protects the bloodstream
IgE: Mediates type I hypersensitivity
IgD: Role not known.

Abnormal Immunoglobulins
Other structurally similar proteins are seen in
serum in many pathological processes, and
sometimes even in healthy persons apart from
antibodies in the following pathological conditions:
A. Multiple myeloma
Fig. 14.4: Pentameric IgM molecule, composed of five B. Heavy chain disease
identical monomers C. Cryoglobulinemia
Chapter 14: Antibodies—Immunoglobulins  | 95
A. Multiple Myeloma ™™ An antibody molecule consists of two identical
The abnormal plasma cells are myeloma cells light chains and two identical heavy chains, which
which also collect in the solid part of the bone. are linked by disul­fide bonds. Each heavy chain has
The disease is called “multiple myeloma”. Myeloma an amino-terminal vari­able region followed by a
constant region
is a plasma cell dyscrasia in which there is
™™ Within the amino-terminal variable domain of each
unchecked proliferation of one clone of plasma heavy and light chain are three complementarity-
cells, resulting in the excessive production of the determining re­gions (CDRs). These polypeptide
particular immunoglobulin synthesized by the regions contribute the anti­gen-binding site of an
clone. Such immunoglobulins are, therefore, called antibody, determining its specificity
™™ Abnormal immunoglobulins are multiple myeloma,
monoclonal.
heavy chain disease C and cryoglobulinemia.
Waldenstrom’s macroglobulinemia: Multiple
myeloma may affect plasma cells synthesizing
IgG, IgA, IgD or IgE. Myeloma involving IgM- Important questions
producing cells (lympho-plasmacytoid cells) is
known as Waldenstrom’s macroglobulinemia. In this 1. What is an antibody? Draw a labeled diagram of IgG.
condition, there occurs excessive production of the 2. Name various classes of immunoglobulins and
respective myeloma proteins (M proteins) and of describe structure and functions of IgG, IgA and
their light chains (Bence-Jones proteins). IgM.
3. Write shot notes on:
Bence-Jones proteins: In most patients, the
a. Immunoglobulin G (IgG)
myeloma cells also secrete excessive amounts
b. Immunoglobulin M (IgM)
of light chains. These ex­cess light chains were
first discovered in the urine of myeloma patients c. Immunoglobulin A (IgA)
and were named Bence-Jones proteins, for their
discoverer Bence Jones (1847). Bence Jones proteins Multiple choice questions (MCQs)
can be identified in urine by its characteristic 1. The immunoglobulin that crosses the placenta is:
property of coagulation when heated to 50°C but a. IgG b. IgM
redissolving at 70°C. These proteins are the light c. IgA d. IgE
chains of immunoglobulins and so may occur as 2. The J-chain is present in the immunoglobulin:
the kappa or lambda forms. But in any one patient, a. IgG b. IgM
the chain is either kappa or lambda only, and never c. IgA d. IgE
both, being uniform in all other respects also. 3. All the following immunoglobulins are heat-stable
except:
B. Heavy Chain Disease a. IgG b. IgM
It is a lymphoid neoplasia characterized by the c. IgA d. IgE
overproduction of the Fc parts of the immuno­ 4. Which is the earliest immunoglobulin to be
globulin heavy chains. synthesized by the fetus?
a. IgG b. IgM
C. Cryoglobulinemia c. IgA d. IgE
It is a condition in which there is the formation 5. The immunoglobulin that mediates type I hyper­
of a gel or a precipitate on cooling the serum, sensitivity reaction is:
which redissolves on warming. It may not always a. IgG b. IgM
be associated with disease but is often found in c. IgA d. IgE
myelomas, macroglobulinemias and autoimmune 6. Antibodies that are bound to mast cells and involve
conditions such as systemic lupus erythematosus. in allergic reactions are:
Most cryoglobulins consist of either IgG, IgM or a. IgG b. IgM
mixture of the two. c. IgA d. IgE
7. The first antibodies synthesized, especially against
Key Points microorganisms:
™™ An antibody or immunoglobulin (lg) is a glycoprotein a. IgG b. IgM
that is made in response to an antigen, and can c. IgA d. IgE
recognize and bind to the antigen that caused its
production Answers (MCQs)
1. a; 2. b; 3. d; 4. b; 5. d; 6. d; 7. b.

www.ebook3000.com
15
Chapter

Complement System

Learning Objectives

After reading and studying this chapter, you should ∙∙ Discuss biological effects of complement
be able to: ∙∙ Describe complement deficiencies and associated
∙∙ Describe the sequence of events when the diseases.
classical pathway and the alternative pathway of
the complement system is activated

Complement: The term ‘complement’ (C) refers to 5. Complement fixation, binding or consumption:
a system of factors which occur in normal serum Complement (C) ordinarily does not bind to
and are activated characteristically by antigen- free antigen or antibody but only to antibody
antibody interaction and subsequently mediate a which has combined with its antigen.
number of biologically significant consequences. 6. Site of complement binding: The site of
complement binding is located on the Fc piece
Complement system of the Ig molecule.
The complement system belongs to the group
of biological effector mechanisms (called Complement Activation
triggered enzyme cascades), which also includes There are nine components of complement
coagulation, fibrinolytic and kinin systems. called C1 to C9. The fraction C1 occurs in serum
Pfeiffer phenomenon: It was discovered by as a calcium ion-dependent complex, which on
Pfeiffer (1894) that cholera vibrios were lysed chelation with EDT A yields three protein subunits
when injected intraperitoneally into specifically called C1q, r and s. Thus, C is made up of a total
immunized guinea pigs (bactereolysis in vivo or of 11 different proteins. C fractions are named C1
Pfeiffer phenomenon). to C9 in the sequence of the cascading reaction,
except that C4 comes after C1, before C2. The
General Properties of Complement components of the classical pathway are numbered
1. Present in the sera of all mammals and of most from C1 to C9, although they do not function in
lower animals: numerical order (C4 comes after C1, before C2).
2. Nonspecific serological reagent: Complement Activation of the complement system can be
from one species can react with antibodies from initiated either by antigen–antibody complexes or
other species. by a variety of nonimmunologic molecules.
3. Serum molecules: The complement system Complement components react in a specific
consists of approximately 30 serum molecules sequence as a cascade either through the classical or
constituting nearly 10% of the total serum alternative pathway.
proteins and forming one of the major defence
systems of the body. Principle pathways of complement
4. Heat labile: Complement as a whole is heat activation
labile, and being destroyed in 30 minutes at
56°C. A serum deprived of its complement Three principle pathways are involved in
activity by heating at 56°C for 30 minutes is then complement activation (Classical pathway, alternate
said to be ‘inactivated’. or properdin pathway, and Lectin pathway).
Chapter 15: Complement System  | 97
A. Classical Complement Pathway instance of amplification. Activated Cl cleaves
The chain of events in which C components react in C4 into two pieces C4a and C4b (C4® C4a + C4b).
a specific sequence following activation of C1 and C4a is an anaphylotoxin and C4b which binds
typically culminate in immune cytolysis is known to cell membrane along with C1.
as the classical pathway (Fig. 15.1). It consists of C14b in the presence of magnesium ions
the following steps: cleaves C2 into two pieces (C2® C2a + C2b).
1. Antigen–antibody binding: The first step is the C2a remains linked to cell-bound C4b, and C2b
binding of C1 to the antigen–antibody (AB) which is released into fluid phase. The pieces
complex. The recognition unit of C1 is Clq, recombine, forming C4b2a has enzymatic
which reacts with the Fc piece of bound IgM activity and is referred to as the classical
of IgG. Clq binding in the presence of calcium pathway C3 convertase.
ions leads to sequential activation of Clr and s. 3. Production of C5 convertase: C3 convertase
In the presence of calcium ions, a trimolecular cleaves C3 into two fragments (C3® C3a + C3b).
complex (CI qrs-Ag-Ab) that has esterase C3a is soluble, and is an anahylotoxin, and
activity is rapidly formed. C3b which remains cell-bound along with
­2. Production of C3 convertase: Activated Cls is C4b2a to form a trimolecular complex C4b2a3b,
an esterase (C1s esterase), one molecule of which has enzymatic activity and is called C5
which can cleave several molecules of C4—an convertase of the classic pathway.

Fig. 15.1: Complement cascade—the classical pathway

www.ebook3000.com
98  |  Section 2: Immunology
4. Formation of the membrane attack complex and a C5 convertase. These are factor B, factor D
(MAC): The terminal stage of the classic pathway and properdin (P).
involves creation of membrane attack complex The binding of C3b to an activator is the first
(MAC), which is also called the lytic unit. step in the alternative pathway. Although C3b
Initiation of membrane attack complex (MAC) is present in the circulation but in the free state
assembly begins with cleavage of C5 by C5 convertase it is rapidly inactivated by the serum protein
into C5a and C5b fragments (C5®C5a+C5b). The C5a factors H and I. However, bound C3b is protected
is the most potent anaphylatoxin in the body and from such inactivation. The bound C3b, in the
C5b continues with the cascade. C6 and C7 then presence of Mg++, interacts with plasma protein
join together. A heatstable trimolecular complex factor B forming C3bB which is also known as ‘C3
C567 is formed part of which binds to the cell proactivator convertase’) to form a magnesium-
membrane and prepares it for lysis by C8 and C9 dependent complex ‘C3bB’. Factor B in the complex
which join the reaction subsequently. This complex is cleaved by serum factor D (also called ‘C3
(C5b67) inserts itself into the plasma membrane proactivator convertase’) into two fragments—Ba
of the target cell. Most of C567 escape and serve to and Bb. Fragment Ba is released into the medium.
amplify the reaction by adsorbing onto unsensitized Fragment Bb remains bound to C3b producing
‘bystander cells’ and rendering them susceptible to C3bBb. C3bBb acts as the alternate pathway C3
lysis by C8 and C9. convertase, capable of producing more C3b. This
The unbound C567 has chemotactic activity, enzyme C3bBb is extremely labile. The function of
though the effect is transient due to its rapid properdin (also called Factor P), a serum protein
inactivation. C8 and C9 then bind, forming the is to stabilize the C3 convertase, which hydrolyzes
membrane at­tack complex (C5b6789) that creates a C3, leading to further steps in the cascade, as in the
pore in the plasma membrane of the target cell. The classical pathway (Fig. 15.1).
mechanism of complement-mediated cytolysis is
the production of ‘holes’, approximately 100° A in 2. Production of Alternative Pathway C5
diameter on the cell membrane. This disrupts the Convertase and MAC
osmotic integrity of the membrane, leading to the C3b produced by C3 convertase binds to C3bBb,
release of the cell contents. producing the alterna­tive C5 convertase (C3bBb3b).
C5 convertase may or may not still have factor P
B. Alternative Complement Pathway attached. Properdin enters the reaction sequence
In the complement cascade, the central process is and binds both C3 and C5 convertase to protect
the activation of C3, which is the major component the complex from the action of factor I (a normal
of C. In the classical pathway, activation of C3 is component of serum that is capable of inactivating
achieved by C42 (classical C3 convertase). The C3b). Properdin interaction, therefore, is the
activation of C3 without prior participation of C142 terminal event in the assembly of the activation
is known as the ‘alternative pathway’ (Fig. 15.2). unit for this pathway.
Once the C5 convertase is formed, C5 is
1. Production of Alternative Pathway C3 cleaved to form C5a and C5b, and the spontaneous
formation of the attack complex (C5b-9) quickly
Convertase
follows. C5b is necessary for formation of the mem­
This pathway bypasses both the recognition unit brane attack complex and cell lysis. The formation
and the assembly of the activation unit as described of this attack complex proceeds in the same
for the classic pathway. Instead, there are at least manner as it does in the classic pathway (Fig. 15.1).
three normal serum proteins that, when activated
together with C3, form a functional C3 convertase Biological Effects of Complement (C)
1. Bacteriolysis and cytolysis: Complement
mediates immunological membrane damage.
This results in bacteriolysis and cytolysis.
2. Virus neutralization: Neutralization of certain
viruses requires the participation of C.
3. Anaphylotoxins: C2 kinins are vasoactive amines
and increase capillary permeability. The cleavage
products of both pathways of complement
activation, C3a and C5a are anaphylatoxic
(histamine releasing) and chemotactic. C4a also
has anaphylotoxin activity but is less potent even
than C3a. C567 is chemotactic and also brings
Fig. 15.2: Complement cascade—the alternative pathway about reactive lysis.
Chapter 15: Complement System  | 99
4. Immune adherence and opsonization : 2. Factor H: It has a strong affinity for C3b, and,
Opsonization is the process of making a after binding C3b, it exerts its control.
particulate antigen more easily identified 3. Anaphylatoxin inactivator: Anaphylatoxin
by a phagocyte. If immune complexes have inactivator is an alphaglobulin that
activated the complement system, the C3b enzymatically degrades C3a, C4a and C5a
bound to them stimulate phagocytosis and which are anaphylatoxins released during
removal of immune complexes. This facilitated the C cascade.
phagocytosis is referred to as opsonization. 4. C4 binding protein: It is a normal human
5. Chemotaxis: Factors Ba (the split product serum protein that binds tightly to activated
from the alternate pathway) and C5a are both C4 and enhances C4b degradation.
chemotactic for polymorphonuclears (PMNs)
and macrophages, thus contributing to local Quantitation of complement (c)
inflammation. C5b67, the partially formed and its components
attack complex, has also been implicated as a Measurement of the complement levels in the serum
chemotactic agent. can be accomplished by estimating the highest
6. Hypersensitivity reaction: Complement partici­ dilution of the serum lysing sheep erythrocytes
pates in: sensitized with antierythrocytic antibody. The
i. Type II hypersensitivity (cytotoxic) reactions: hemolytic unit of C (CH50) may be defined as that
ii. Type III (immune complex) hypersensitivity amount of complement that lyses 50% of sensitized
reactions: erythrocytes under defined conditions.
7. Autoimmune diseases: Serum C (complement)
components are decreased in many auto­ Biosynthesis of complement
immune diseases such as systemic lupus Various complement components are synthesized
erythromatosus and rheumatoid arthritis. in different parts of the body, e.g. intestinal
8. Endotoxic shock: Endotoxins can efficiently epithelium (C1), macrophages (C2, C4), spleen
activate the alternative pathway of the comple­ (C5, C8) and liver (C3, C6, C9). The site of synthesis
ment cascade. There is massive C3 fixation and of C7 is not known. The control mechanism
platelets adherence in endotoxic shock. Large that controls the synthesis of the complement
scale platelet lysis and release of large amounts of component is not known.
platelet factor lead to disseminated intravascular
coagulation (DIC) and throbocytopenia. In Complement deficiencies
endotoxic shock with gram-negative septicemia
Complement deficiencies have been associated
or dengue, hemorrhagic fever may have a similar
with recurrent bacterial and fungal infections
pathogenesis. Depletion of C protects against the
as well as with collagen-vascular inflammatory
Schwartzman reaction.
diseases. Human genetic deficiencies of
Regulation of the Complement System complement components and associated diseases
Several inbuilt control mechanisms regulate the are listed in Table 15.1.
complement cascade at different steps. These are
mainly of two kinds: Table 15.1  Human genetic deficiencies of comple­
A. Inhibitors ment components and associated diseases
B. Inactivators
A. Inhibitors: They bind to complement compo­ Complement Association with disease
nents and halt their further functioning. deficiency
1. C1 esterase: Normal serum contains an C1 inhibitor Hereditary angioneurotic edema
inhibitor of C1 esterase (ClsINH). This heat C1r Systemic lupus erythematosus-
like disease, frequently fatal from
labile alpha neuraminoglycoprotein also overwhelming infection
inhibits many other esterases found in
C2 Increased susceptibility to infections
blood, such as plasmin, kininogen and the
C3 Recurrent bacterial infections
Hageman factor.
C4 Systemic lupus erythematosus-like
2. Vitronectin: It is also known as the S protein. disease
The S protein present in normal serum C5 Recurrent infections—lupus like disease
binds to C567 and modulates the cytolytic C6, C7, C8 Recurrent infections—Disseminated
action of the membrane attack complex. gonococcal infections
B. Inactivators: They are enzymes that destroy C9 Not more susceptible to disease
complement proteins. than other individuals in the general
1. Factor 1: Normal serum contains an endo­ population`
peptidase, called Factor 1 which cleaves C3b Factor 1 Low C3 levels with recurrent bacterial
and possibly C4b. infections

www.ebook3000.com
100  |  Section 2: Immunology

Key Points 2. What is the chemical nature of components of


complement?
™™ The complement system comprises a group of serum a. Protein
proteins, many of which exist in inactive forms. It is b. Lipopolysaccharide
present in all normal individuals in their blood c. Lipid
™™ Complement activation occurs by the classical, d. None of the above
alternative or lectin pathways, each of which is 3. The activation of complement takes place through
initiated differently
either of the following pathways except:
™™ The classical pathway is activated with the formation
a. The classical pathway
of soluble antigen-antibody complexes (immune
b. The lectin pathway
complexes) or the binding of antibody to antigen
c. The alternative pathway
on a suitable target
d. The lipid pathway
™™ Activation of the alternative and lectin pathways
is antibody-independent. The lectin pathway is 4. Classical pathway of the complement is activated
activated by lectins by:
™™ Biological effects of complement: 1. Bacteriolysis and a. Antigen
Cytolysis; 2. Virus neutralization; 3. Anaphylotoxins; 4. b. Antibody
Immune adherence and opsonization; 5. Chemotaxis; c. Antigen Antibody complex
6. Hypersenitivity reaction; 7. Autoimmune diseases; d. None of the above
8. Endotoxic shock 5. The alternative pathway of the complement is
™™ Clinical consequences of inherited complement initiated by:
deficien­cies range from increases in susceptibility a. Endotoxins
to infection to tissue damage caused by immune b. Lipopolysaccharides
complexes. c. Yeast cell walls
d. All the above
Important questions 6. First component of complement which binds to
antigen-antibody complex in classical pathway is:
1. Define complement. What is the sequence of events a. C1q b. C1r
when the classical pathway of the complement c. C1s d. C3
system is activated? 7. Which component of complement is present in the
2. Write short notes on: highest concentration in the serum?
a. Alternative pathway of complement. a. C1 b. C2
b. Biological effects of complement. c. C3 d. C5
8. Factor B and Factor D are important components
Multiple choice questions (MCQs) of:
a. Classical pathway of complement
1.
All the following statements are true for b. Alternative pathway of complement
complement except that these are: c. Both of these
a. Glycoproteins d. None of the above
b. Heat labile substances
c. Found in an active state in the plasma Answers (MCQs)
d. Synthesized primarily by liver cells 1. c; 2. a; 3. d; 4. c; 5. d; 6. a; 7. c; 8. b.
16
Chapter

Antigen–Antibody Reactions

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Describe applications of agglutination reactions
be able to: and their uses
∙∙ Differentiate between precipitation and agglutination ∙∙ Discuss prin­ciple and applications of agglutination
∙∙ Describe prozone phenomenon reactions
∙∙ Discuss mechanism and applications of precipit­ ∙∙ Describe principle of complement fixation test
ation reactions giving suitable examples
∙∙ Discuss principle and clinical applications of
∙∙ Describe types of precipitation reactions
immuno­fluorescence technique
∙∙ Describe principle and applications of Immuno­
elec trophoresis, radial immunodiffusion, ∙∙ Discuss principle, various types and clinical
counter­­i mmunoelectrophoresis (CIE)), rocket applications of ELISA technique.
electrophoresis

INTRODUCTION ANTIGEN–ANTIBODY INTERACTIONS


The antigen–antibody interaction is a bimolecular The reactions between antigen and antibody
association that exhibits exquisite specificity. It is occurs in three stages:
similar to an enzyme–substrate interaction, with 1. Primary stage: In primary or initial interaction,
an important distinction. there is no visible effect and the reaction is
rapid.
Uses
2. Secondary stage: The primary interaction in
1. In vivo or in the body
most instances, but not all, is followed by the
i. Protection: The in vivo interaction that
secondary stage, leading to demonstrable
occurs in vertebrate animals are antibody
events such as precipitation, aggluti­nation, lysis
absolutely essential in protecting the
of cells, killing of live antigens, neutralization of
animal.
toxins and other biologically active antigens,
ii. Basis of antibody-mediated immunity:
fixation of complement, immobilization
In the body, they form the basis of
of motile organisms and enhancement of
antibody-mediated immunity in infectious
phagocytosis. A single antibody can cause
diseases, or of tissue injury in some types
precipitation, agglutination and most of the
of hypersensitivity and autoimmune
other serological reactions but it is also true
diseases.
that an antigen can stimulate the production
2. In vitro or in the laboratory
of different classes of immunoglobulins which
i. These assays can be used to detect the are different in their reaction capacities as well
presence of either antibody or antigen as in other properties (Table 16.1).
ii. Vital roles in diagnozing diseases 3. Tertiary stage: Some antigen–antibody reactions
iii. Monitoring the level of the humoral immune occurring in vivo initiate chain reactions
response that lead to neutralization or destruction of
iii. Identifying molecules of biological or injurious antigens, or to tissue damage. These
medical interest. are tertiary reactions and include humoral

www.ebook3000.com
Chap-16.indd 101 15-03-2016 11:12:02
102  |  Section 2: Immunology
Table 16.1  Efficacy of the different Immuno­ Sensitivity
globulin classes in various serological reactions Sensitivity is defined as the ability of a test to
Serological Reaction IgM IgG IgA identify correctly all those who have the disease,
i.e. “true positives”.
1. Precipitation Weak Strong Variable
2. Agglutination Strong Weak Moderate
Specificity
3. Complement Weak Strong Negative
fixation Specificity has been defined as the ability of a test
to identify correctly those who do not have the
disease, i.e. “true negatives”.
immunity against infectious diseaseas well
as clinical allergy and other immunological SEROLOGICAL REACTIONS
diseases. The study of antigen–antibody reactions in vitro
is called serology [serum and ology]. Antigen–
GENERAL CHARACTERISTICS OF antibody reactions in vitro are known as serological
ANTIGEN–ANTIBODY REACTIONS reactions.

1. Highly specific: Antigen–antibody reaction is TYPES OF ANTIGEN AND ANTIBODY


highly specific. REACTIONS
2. Lock and key arrangement: Both antigens
and antibodies participate in the formation of A. Precipitation reactions
agglutinates or precipitates. The molecules are B. Agglutination reactions
held together in lock and key arrangement. C. Complement fixation test (CFT)
3. No denaturation: There is no denaturation of D. Neutralization tests
antigen or antibody during the reaction. E. Opsonization
4. Surface antigens: During combination, only F. Immunofluorescence
surface antigens participate. G. Radioimmunoassay (RIA)
5. Entire molecules react and not fragments. H. Enzyme-linked immunosorbent assay (ELISA)
6. Combination is firm and reversible: The firm­ I. Immunoelectroblot techniques
ness of the union is influenced by the affinity
J. Immunochromatographic tests
and avidity of the reaction.
K. Immunoelectron microscopic tests.
Affinity denotes the intensity of attraction
between the antigen and antibody molecules.
Avidity is the strength of the bond after the A. Precipitation Reactions
formation of the antigen–antibody complexes. Precipitation: When a soluble antigen combines
7. Combination in varying proportions: Antigens with its antibody in the presence of electrolytes
and antibodies can combine in varying (NaCl) at a suitable temperature and pH, the
proportions. Antibodies are generally bivalent. antigen–antibody complex forms an insoluble
Antigens may have valencies upto hundreds. precipitate and is called precipitation. Antibodies
that thus aggregate soluble antigens are called
precipitins. Precipitation is relatively less sensitive
ANTIGEN AND ANTIBODY MEASUREMENT for the detection of antibodies.
Measurement may be in terms of mass (e.g. Flocculation: When instead of sedimenting, the
nitrogen) or more commonly as units or titer. precipitate remains suspended as floccules, the
reaction is known as flocculation. Precipitation can
Titer take place in liquid media or in gels, such as agar,
agarose or polyacrylamide.
Antibody titer of a serum is the highest dilution of the
serum which shows an observable reaction with the Zone phenomenon: A quantitative precipitation
antigen in the particular test. It is usually expressed reaction can be performed by placing a constant
as the reciprocals of the dilution of the serum. amount of antibody in a series of tubes and adding
increasing amounts of antigen to the tubes. This
PARAMETERS OF SEROLOGICAL TESTS plot of the amount of antibody precipitated versus
increasing antigen concentration (at constant total
Two important parameters of serological tests are antibody) reveals three zones (Fig.16.1). This is
sensitivity and specificity: called zone phenomenon.

Chap-16.indd 102 15-03-2016 11:12:02


Chapter 16: Antigen–Antibody Reactions  | 103

Fig. 16.2: Mechanism of precipitation by lattice formation. In


A (antibody excess) and B (antigen excess), lattice formation
does not occur. In C (zone of equivalence), lattice formation and
Fig. 16.1: A quantitative precipitation test showing: A.
precipitation occur optimally
Prozone (zone of antibody excess); B. Zone of equivalence;
C. Zone of antigen excess precipitation is inhibited (Fig 16.2). The lattice
hypothesis holds good for agglutination also.
Three Zones
1. Zone of antibody excess or prozone (ascending Applications of Precipitation Reaction
part):
The prozone is of importance in clinical The precipitation test may be carried out as either a
serology,as sera rich in antibody or may qualitative or quantitative test. It has the following
sometimes give a false-negative precipitation applications:
or agglutination result, unless several dilutions 1. Forensic application in the identification of
are tested. blood and seminal stains.
2. Equivalence zone (peak): Occurs when the ratio 2. In testing for food adul­terants.
of antibody to antigen is optimal. 3. To standardize toxins and antitoxins.
3. Zone of antigen excess or postzone (descending
part): In the tubes containing more antigen, the Types of Precipitation and
amount of precipitate increases up to a point, Flocculation Tests
after which it decreases as a result of smaller A. Ring test: This consists of layering the antigen
complexes being formed in the zone of antigen solution over a col­umn of antiserum in a narrow
excess. tube and is the simplest type of precipitation
test. A precipitate forms at the junction of the
Mechanism of Precipitation two liquids. Ring tests have only a few clinical
The lattice hypothesis was proposed by Marrack applications now.
(1934) to explain the mechanism of precipitation. Examples:
According to this concept, multivalent antigens i. Ascoli’s thermoprecipitin test
combine with bivalent antibodies in varying ii. The grouping of streptococci by the
proportions, depending on the antigen–antibody Lancefield technique.
ratio in the reacting mixture, which is supported B. Slide test (Slide flocculation test): When the
by considerable experimental evidence and is drops each of the antigen and antise­rum are
now widely accepted. Precipitation results when placed on a slide and mixed by shaking, floc­
a large lattice is formed consisting of alternating cules appear.
antigen and antibody molecules. This is possible Example: The VDRL test for syphilis is an
only in the zone of equivalence. Formation of an example of slide flocculation.
antigen–antibody lattice depends on the valency C. Tube test (Tube flocculation test)
of both the antibody and antigen. The antibody i. The Kahn test for syphilis is an example of
must be bivalent; a precipitate will not form with a tube flocculation test.
monovalent antibody fragments. The antigen must ii. Standardization of toxins and toxoids.
be either bivalent or polyvalent. In the zones of D. Immunodiffusion (Precipitation Reactions in Gels)
antigen or antibody excess, the lattice does not Immunodiffusion refers to a precipitation
en­large, as the valencies of the antibody and the reaction that occurs between an antibody and
antigen, respectively, are fully satisfied .In either antigen in an agar gel medium. Immunodiffusion
case, extensive lattices cannot be formed and is usually performed in a soft (1%) agar gel.

www.ebook3000.com
Chap-16.indd 103 15-03-2016 11:12:03
104  |  Section 2: Immunology
Advantages ii. For screening sera for antibodies to influenza
1. The reaction is visible as a distinct band of viruses.
precipitation, which is stable and can be iii. For the laboratory diagnosis of multiple
stained for preservation, if necessary. myeloma or agammaglobulinemia.
2. These immunodiffusion reactions can be used to 4. Double diffusion in two dimensions (Ouch­
determine relative concentrations of antibodies ter­lony Technique
or antigens, to compare antigens, or to determine In the Ouchterlony method, both antigen and
the relative purity of an antigen preparation. antibody diffuse (hence, double diffusion)
3. It also indicates identity, cross-reaction and radially form wells toward each other, thereby
nonidentity between different antigens establishing a concentration gradient. When
soluble antigen and soluble antibody are placed
Types of Immunodiffusion Tests in separate small wells punched into agar that
has solidified on a slide or glass plate, the
1. Single diffusion in one dimension (Oudin
antigen and the antibody will diffuse through
procedure)
the agar. The holes are located only a few
Antibody is incorporated in agar gel in a test
millimeters apart, and antibody and antigen
tube. Antigen solution is then layered over
will interact to form a line of precipitate in the
it. The antigen diffuses downward through
area in which they are in optimal proportions.
the agar gel and wherever it reaches in
In specimens containing several soluble
optimum concentration with antibody, a line
antigens, multiple precipitin lines are observed,
of precipitation is formed (Fig. 16.3). The
each occurring between the wells (Fig. 16.5).
number of bands indicates the number of
The visible line of precipitation permits a
different antigens present.
comparison of antigens for identity (same
2. Double diffusion in one dimension (Oakley-
antigenic determinants), partial identity
Fulthorpe procedure)
(cross-reactivity), or nonidentity against a given
Here the antibody is incorporated in gel in a test
selected antibody.
tube, above which is placed a column of plain
agar and antigen is layered on surface of this.
Antigen and antibody diffuse (double diffusion) Example
towards each other (in one dimension) through Elek test for toxigenicity in diphtheria bacilli:
the intervening column of plain agar and A special variety of double diffusion in two
forms a band of precipitate where they meet at dimensions is the Elek test for toxigenicity in
optimum proportion (Fig. 16.3). diphtheria bacilli.
3. Single diffusion in two dimensions (Radial
immunodiffusion—Mancini method) 5. Immunoelectrophoresis
The assay is carried out by incorporating Immunoelectrophoresis is a procedure that
monospecific antiserum into melted agar and combines electrophoresis and double diffusion
allowing the agar to solidify on a glass plate in a for the separation of antigen–antibody reactions
thin layer. Wells then are punched into the agar, in gels.
and different dilutions of the antigen are placed
into the various wells. As the antigen diffuses
into the agar, a ring of precipitation, a precipitin
ring, forms around the well (Fig. 16.4). The
area of the precipitin ring is proportional to the
concentration of antigen.

Uses
i. To quantitate serum immunoglobulins, Fig. 16.4: Single diffusion in two dimensions (Radial
immunodiffusion—Mancini method)
complement proteins, other substances.

Fig. 16.3: 1. Single diffusion in one dimension (Oudin


procedure) 2. Double diffusion in one dimension (Oakley- Fig. 16.5: Double diffusion in two dimensions (Ouchterlony
Fulthorpe procedure) technique)

Chap-16.indd 104 15-03-2016 11:12:04


Chapter 16: Antigen–Antibody Reactions  | 105

Fig. 16.7: Counterimmunoelectrophoresis (CIE)

i. Counterimmunoelectrophoresis (CIE) or
Countercurrent eletrophoresis (CIEP)
Counterimmunoelectrophoresis can be used only
for antigens and antibody that migrate in opposite
directions in the electric field. The test is set up
similarly to that for double diffusion in agar. Two
wells are punched about 1 cm apart in an agar
slab on a glass plate. The antigen and antibody
Fig. 16.6: Immunoelectrophoresis solutions are placed in these wells in such a way
that when the electric fleld is applied across the
plate, the antigen will migrate toward the antibody,
In this procedure, antigens are separated by and the antibody will migrate toward the antigen.
electrophoresis in an agar gel. A small drop of A precipitin line will form when the antigen and
solution containing the antigens (usually proteins) antibody meet in optimal proportions resulting in
is placed into a small well punched out of solidified precipitation at a point between them (Fig 16.7).
agar on a small glass plate. The plate then is placed Advantages of Counterimmunoelectrophoresis
in an electric field to allow for the electrophoretic (CIE)
migration of the antigens. A trough is then cut 1. More sensitive: It is 10 times more sensitive than
next to the wells and filled with antibody and simple diffusion in agar.
diffusion allowed to proceed for 18–24 hours.The
2. More rapid assay: It is a much more rapid assay
antibody and the antigens diffuse toward each
(as little as 30 minutes for some antigens) than
other, resulting in the formation of precipitin bands
is double diffusion in gel.
or arcs wherever they are in optimal proportions
(Fig. 16.6).
Clinical Applications
Uses i. For detection of various antigens: such as
i. To separate many antigens as well as to hepatitis B surface antigen (HBS antigen) and
indicate the potential purity of an antigen alpha-fetoprotein in serum and meningococcal
ii. To separate the major blood proteins in serum and cryptococcal antigens in CSF.
for certain diagnostic tests. ii. To detect the presence of anti-DNA antibody in
iii. For testing for normal and abnormal proteins the serum of patients with several autoimmune
in serum and urine. disorders.

6. Electroimmunodiffusion ii. One-dimensional Single


Immunodiffusion is a slow process. The Electroimmunodiffusion (Rocket
development of precipitin lines can be speeded Electrophoresis)
up by electrically driving the antigen and antibody As in radial immunodiffusion, small wells are cut
in a gel. Various methods have been described in an agarose gel slab on a glass plate. The agar
combining electrophoresis with diffusion. Of these, contains antibody to one or more antigens of
frequently used methods in the clinical laboratory interest. The antigen, in increasing concentration,
are: is placed in wells, and an electric potential is
1. One-dimensional double electroimmuno­ applied across the plate. A negatively charged
diffusion (counterimmunoelectrophoresis) antigen is electrophoresed in a gel containing
2. One-dimensional single electroimmuno­ antibody. The antigens migrate into the gel and
diffusion (rocket electrophoresis) precipitate with the antibody in the gel at the

www.ebook3000.com
Chap-16.indd 105 15-03-2016 11:12:05
106  |  Section 2: Immunology
appropriate position. The precipitate formed antibody causes a clumping or agglutination of the
between antigen and antibody has the shape of cells. Antibodies that produce such reactions are
a rocket. (Fig. 16.8). The height (distance from called aggglutinin. Agglutination occurs optimally
the antigen well to the top of the precipitin band) when antigen and antibodies react in equivalent
of which is proportional to the concentration proportion.
of antigen in the well. is so named because the Agglutination reactions are more sensitive than
precipitin bands resemble rockets.Thus, this precipitin reactions because of the direct nature of
method is primarily a quantitative technique. the antigen/carrier/antibody interaction.
Prozone phenomenon: The prozone phenomenon
Main Application may be seen when either an antibody or an antigen
i. Quantitative estimation of antigens: For is in excess. Just an excess of antibody inhibits
quantitative estimation of antigens and does precipitation reactions, such excess can also inhibit
permit measurement of antigen levels. reactions. This inhibition is called the prozone
phenomenon. Several mechanisms can cause the
Laurell’s Two-dimensional prozone effect.
Electrophoresis Blocking antibodies: Occasionally antibodies
A variant of rocket electrophoresis is Laurell’s two- are formed that will react with the antigenic
dimensional electrophoresis. In this technique, determinants on a cell but will not cause
the antigen mixture is first electrophoretically agglutination (e.g. anti-Rh and anti-Brucella).
separated in a direction per­pendicular to that of These antibodies are called blocking antibodies
the final rocket stage. By this method, one can because they inhib­it agglutination by the complete
quantitate each of several antigens in a mixture antibody added subsequently.
(Fig 16.9).
APPLICATIONS OF AGGLUTINATION
B. Agglutination Reactions REACTION
When a particulate antigen is mixed with its
antibody in the presence of electrolytes at a 1. Slide Agglutination
suitable tempera­ture and pH, the particles are A drop of sterile saline is placed on one of the
clumped or agglutinated. When antigen is present divisions of the slide or plate and is emulsified in
on the surface of a cell or particle, the addition of it bacterial culture. The diagnostic serum is taken
and is then mixed into the latter. The mixing is
completed by tilting the slide to and fro. Distinct
clumping within 60 seconds is a positive result.
On another area of the slide or plate, in parallel
with the test, a control test is performed, adding
only saline, instead of serum, to the bacterial
suspension. If clumping takes place, the bacterial
suspension is autoagglutinable and the reaction
with the serum must be disregarded.

Uses
Fig. 16.8: Rocket electrophoresis i. For the identification of unknown bacterial
cultures.
ii. Very rapid: It requires only small quantities of
culture and serum.
iii. Also the method for blood grouping and cross-
matching.

2. Tube Agglutination
Fig. 16.9: Laurell’s variant of rocket electrophoresis (two- Serum from a patient thought to be infected with
dimensional electrophoresis) a given bacterium is serially diluted in a series
1. First run—Antigens are separated by electrophoretic of tubes to which the bacteria is added. The last
mobility.
tube showing visible agglutination will reflect the
2. The second run is done at right angles to the first which
drives the antigens into the antiserum containing gel serum antibody titer of the patient. The reciprocal
to form precipitation peaks. The area of the peak is of the greatest serum dilution that elicits a positive
proportional to the concentration of the antigen agglutination is known as the agglutinin titer.

Chap-16.indd 106 15-03-2016 11:12:06


Chapter 16: Antigen–Antibody Reactions  | 107
Uses of Tube Agglutination
Tube agglutination is routinely employed for the
serological diagnosis of typhoid, brucellosis and
typhus fever.
i. Widal test: Used for the diagnosis of enteric
fever.
Fig. 16.10: Antiglobulin (Coombs’) tests. Rh-positive
ii. Tube agglutination test for brucellosis.
erythrocytes 1. are mixed with incomplete antibody; 2.
iii. Weil-Felix reaction: Weil-Felix reaction for The antibody coats the cells; 3. but, being incomplete,
serodiagnosis of typhus fevers is heterophil cannot produce agglutination. On addition of antiglobulin
agglutination test. serum; 4. which is complete antibody to immunoglobulin,
iv. Paul-Bunnel test: It is based on the presence of agglutination takes places
sheep cell agglutinins in the sera of infectious
mononucleosis patients. IgM antibodies bentonite. Such test is more convenient and more
capable of agglutinating human red cells sensitive for detection of anti­bodies. Such tests are
at 0–4°C (cold agglutinins) are sometimes known as passive agglutination tests.
found in certain human diseases including for Reversed passive agglutination: When instead
diagnosis of mycoplasmal (primary atypical of antigen, the antibody is adsorbed to carrier
pneumonia) streptococcal agglutination test. particles in tests for estimation of antigens,
the technique is known as reversed passive
3. Antiglobulin (Coombs’) Test agglutination.
The antiglobulin test for Rh antibodies was
originally devised by British immunologist, RRA Examples of Passive Agglutination
coombs. i. Hemagglutination test
a. Rose-Waaler test: A special type of passive
Principle of the Antiglobulin Test agglutination test is the Rose-Waaler test.
When sera containing incomplete anti-Rh In rheumatoid arthritis,RA factor (an
antibodies are mixed with Rh-positive red cells, antigammaglobulin autoantibody) appears
the antibody coats the surface of the erythrocytes in the serum. It acts as antibody to human
but they are not agglutinated.When such IgG. The RA factor is able to agglutinate
antibody-coated erythrocytes are treated with red cells coated with globulins. The
a rabbit antihuman antiserum against human antigen used for the test is a suspension
gammaglobulin (antiglobulin or Coombs’ serum), of sheep erythrocytes sensitized with a
the cells are agglutinated. This is the principle of subagglutinating dose of rabbit antisheep
the antiglobulin test (Fig. 16.10). erythrocyte antibody (amboceptor).
b. Treponema pallidum haemagglutination
(TPHA): One of the most widely used
Types of Coombs’ Test
passive agglutination tests employing
Coombs test is of two types: direct and indirect. erythrocytes is Treponema pallidum
Direct Coombs’ test: The most direct, Coombs’ hemagglutination (TPHA) for serological
test, the sensitization of the erythrocytes with diagnosis of treponemal infection.
incomplete antibodies takes place in vivo as in ii. Latex agglutination test: Polystyrene latex,
case of hemolytic disease of the newborn due to which can be manufactured as uniform
Rh incompatibility. spherical particles, 0.8 mm in diameter, can
adsorb several types of antigens.
Indirect Coombs’ test: In the indirect Coombs’ Latex agglutination tests (latex fixation tests)
test, sensitization of RBCs with incomplete are widely employed in clinical laboratory
antibodies is performed in vitro (Fig. 16.10). for the detection of antistreptolysin –O(ASO),
C-reactive protein (CRP), RA factor, human
Uses chorionic gonadotropin (HCG) and many
For demonstrating any type of incomplete or other antigens.
nonagglutinating antibody, as e.g. in brucellosis. iii. Coagglutination.
Principle: Similar to latex agglutination,
4. Passive Agglutination Test coagglutination uses anti­body bound to a particle
A precipitation reaction can be converted into to enhance visibility of the ag­glutination reaction
agglu­tination reaction by coating soluble antigen between antigen and antibody Certain strains
on to the surface of carrier particles. The commonly of (the Cowan strain, ATCC 12498) have a high
used carrier particles are red cells, latex particles or content of surface protein A. Protein A on the

www.ebook3000.com
Chap-16.indd 107 15-03-2016 11:12:07
108  |  Section 2: Immunology
highest dilution of the serum that lyses one unit
volume of washed sheep RBCs in the presence of
excess complement within a fixed time (usually
30–60 minutes at 37°C).

Complement Fixation Tests


CFT is a complex procedure consisting of two steps
and five reagents—antigen, antibody, complement,
sheep erythrocytes and amboceptor (rabbit
antibody to sheep red cells). Each of these reagents
has to be sep­arately standardized.

Principle
A known antigen is mixed with test serum lacking
complement. When immune complexes have had
time to form, comple­ment is added to the mixture.
If the patient’s serum contains antibody to the
Fig. 16.11: Coagglutination antigen, the re­sulting antigen–antibody complexes
will bind all of the complement added. In most of
Staph. aureus cell wall binds the Fc portion of the cases, fixation of complement with antigen-
the immunoglobulin molecule, leaving the Fab antibody complex causes in itself no visible effect.
portion free to bind antigen. Visible agglutination (Antigen + Antibody + Complement).
of the staphylococcal cells serves as a positive test This test consists of two sep­arate systems and
to indicate antigen-antibody binding (Fig.16.11). these two systems are tested in sequence (Fig.
16.12). Appropriate controls should be used,
Uses including the following: antigen and serum controls
to ensure that they are not anticomplementary,
i. Coagglutination can be used for detecting the
complement control to ensure that the desired
presence of antigens in serum, urine and CSF.
amount of comple­ment is added, and cell control
ii. Several commercial suppliers have prepared
to see that sensitized erythrocytes do not undergo
coagglutination reagents for identification
lysis in the absence of complement.
of antigens of various streptococcal groups,
including Lancefield groups A, B, C, D, F, G, and N;
Procedure
Streptococcus pneumoniae; Neisseria meningitidis;
N. gonorrhoeae; and Haemophilus influenzae types A. Test system: It consists of (i) Antigen- suspected
A to F grown in cul­ture. of causing the patient’s disease; (ii) Patient’s
serum (antibody); (iii) Complement.
C. Complement Fixation Test (CFT) B. Indicator system: It consists of sheep red cells
(antigen) coated with anti-sheep-red cell
When complement binds to an antigen–antibody
antibody and Complement—An exogenous
complex. it be­c omes “fixed” and “used up.”
source of complement (usually guinea pig
Complement fixation tests are very sensitive and
serum). Afterward, sen­sitized indicator cells,
can be used to detect extremely small amounts of
usually sheep red blood cells previously coated
an an­tibody for a suspect micro-orgamism in an
with complement-fixing antibodies, are added
individual’s serum. This can detect as little as 0.04
to the mixture. Complement lyses antibody-
µg of antibody nitrogen and 0.01 µg of antigen.
coated red cells.
Standardization of Complement and
Interpretation—Positive CF Test—Absence
Amboceptor
of Lysis
Guinea pig serum is first titrated for complement
activity. One unit or minimum haemolytic dose The spe­cific antibodies are present in the test serum
(MHD) of complement is the highest dilution of and complement is con­sumed by the immune
guinea pig serum that lyses one unit volume of complexes. Insufficient amount of comple­ment
washed sheep RBCs in the presence of excess of will be available to lyse the indicator cells.
haemolysin (amboceptor) within a fixed time
(usually 30–60 minutes) at a fixed temperature Negative CF Test Lysis of The Indicator Cells
(37°C). Similarly, amboceptor should also be Lysis of the indicator cells indicates lack of antibody
titrated. One MHD of hemolysin is defined as and a negative CF test. Lysis of the indicator cells

Chap-16.indd 108 15-03-2016 11:12:07


Chapter 16: Antigen–Antibody Reactions  | 109
(Fig. 16.12) results if immune com­plexes do not aggregated and adhere to the cells. This is
form in part of the test because the antibodies are known as immune adherence.
not present in the test serum, complement remains ii. Immobilization test : In the Treponema
and lyses the indicator cells. pallidum immobilization test, a highly specific
test formerly considered the ‘gold standard’
Uses for the serological diagnosis of syphilis,the
test serum is incubated anaerobically
1. Diagnosis of syphilis: Complement fixation
with a suspension of live treponemes and
was once used in the diagnosis of syphilis (the
complement. If antibodies are present, the
Wassermann test).
treponemes will be found to be immobilized.
2. In the diag­n osis of certain viral, fungal, iii. Cytolytic or cytocidal tests: These are also
rickettsial, chlamydial, and proto­zoan diseases, complement-dependent. When V. cholerae
it is currently used. is mixed with its antibody in the presence
3. For diagnosing infection caused by fungi, of com­plement, the bacterium is killed and
respira­tory viruses, and arboviruses, as well as lyzed. This forms the basis of the vibriocidal
to diagnose Q-fever. This test is still probably antibody test for the measurement of
the most common method. anticholera antibodies.

Anticomplementary Effect Indirect Complement Fixation Test


Nonspecific adsorption of complement may give Certain avians (for example, duck, turkey, parrot)
false positive results. Some sera may develop and mammalian (for example, horse, cat) sera
anti­complementary properties on aging, and after do not fix guinea pig com­plement. The indirect
bac­terial contamination. Hemolyzed blood serum complement fixation test may be employed when
also has an anticomplementary effect. such sera are to be tested. Here, the test is set up
Other complement-dependent serological tests: in duplicate and after the first step, the standard
i. Immune adherence: Some bacteria, like antiserum known to fix complement is added to
Vibrio cholerae and Treponema pallidum, one set. If the test serum contained antibody, the
react with spe­cific antibody in the presence an­tigen would have been used up in the first step
of complement and particulate material such and, therefore, the standard antiserum added
as erythrocytes or platelets. The bacteria are subsequently would not be able to fix complement.

Fig. 16.12: Complement fixation test

www.ebook3000.com
Chap-16.indd 109 15-03-2016 11:12:08
110  |  Section 2: Immunology
Therefore, hemolysis indicates a positive result ii. Nagler’s reaction: Cl. perfringens produces
in the indirect test. α-toxin and produces opalescence in serum
or egg yolk media. This reaction is specifically
D. Neutralization Tests neutralized by the antitoxin.
These are of two types:
1. Viral neutralization tests E. Opsonization
2. Toxin neutralization tests. Opsonization is the process in which micro-
organisms or other particles are coated by antibody
1. Viral Neutralization and/or complement, and thus prepared for
Neutralization of viruses by their antibodies “recognition” and ingestion by phagocytic cells.
can be demonstrated in various systems. This Immune opsonization: Phagocytes, such as
antibody-mediated viral inactivation is called viral macrophages, monocytes and neutrophils possess
neutralization. surface receptors (CRI) for C3b and Fc receptors for
Indicator systems: Laboratory animals or tissue antibody. If immune complexes have activated the
culture cells are used as “ indicator systems” in complement system, then Fc and CRI receptors,
these tests. The test serum is diluted serially, present on the phagocyte, bind Fc region of
incubated with a known amount of virus and antibody and C3b bound on immune com­plexes
the mixture is then added to indicator cultures: respectively, thus facilitating their phago­cytosis.
animals, embryonated hen’s egg and tissue culture. This facilitated phagocytosis by antibody and
complement is known as immune opsonization.
2. Toxin Neutralization Nonimmune opsonization: On the contrary,
Bacterial exotoxins are good antigens and nonimmune opsonization requires only C3b
their activity may be com­pletely neutralized by (opsonin) for opsonization. C3b binds to CR1
appropriate concentrations of specific antibody. receptors present on the phagocytes thus
Antibody to bacterial exotoxin is usually referred facilitating their phagocytosis (Fig 16.13).
to as antitoxin which is important clinically, in
protection against and recovery from diseases such F. Immunofluorescence
as diphtheria and tetanus. Toxin neutralization can Immunofluorescence is a process in which dyes
be tested in vivo or in vitro. called fluo­rochromes are exposed to UV, violet or
A. Toxin neutralization in vivo blue light to make them fluoresce or emit visible light.
i. Toxigenicity test: The neutralizing capa­ Fluorescent molecules absorb light of one wavelength
city of an antitoxin can be assayed by (exci­tation) and emit light of another wavelength
neutralization test in which mixture of toxin (emission). Albert Coons and his colleagues (1942)
and antitoxin is injected into a susceptible showed that fluorescent dyes can be conjugated to
animal and the least amount of antitoxin antibodies and that such ‘labelled’ antibodies can
that pre­vents death or disease in the animal be used and identify antigens in tissues. Antibody
is estimated. molecules bound to antigens in cells or tis­sue sections
ii. Schick test: With the diphtheria toxin, which can similarly be visualized. The emitted light can be
in small doses causes cutaneous reaction, viewed with a fluorescence microscope, which is
neutralization test can be carried out on the equipped with a UV light source.
human skin. The Schick test is based on the Fluorescent dyes: Rhodamine B and fluo­rescein
ability of circulating antitoxin to neu­tralize isothiocyanate (FITC) are the most commonly
the diphtheria toxin given intradermally.
Neutralization (no reaction) indicates
immunity and redness and erythema
indicates susceptibility to diphtheria.
B. Toxin neutralization in vitro: If a toxin has a
demonstrable in vitro effect, this effect can be
neutralized by specific anti­toxin.

Examples
i. Antistreptolysin O (ASO) test: Antistreptolysin
O (ASO)-antitoxin, present in the serum of
the patient suffering from Strep. pyogenes
infection, neutralizes the haemolytic activity
of the streptococcal O hemolysin (toxin). Fig. 16.13: Opsonization

Chap-16.indd 110 15-03-2016 11:12:08


Chapter 16: Antigen–Antibody Reactions  | 111
used fluorescent dyes. Fluorescein emits an
intense yellow-green fluorescence. Rhodamine
emits a deep red fluorescence. Phycoerythrin
and Phycobilipro­t eins are other highly fluo­
rescent substances and have also come into use.
The most commonly used fluorescent dyes such
as rhodamine B or fluo­rescein isothiocyanate
A
(FITC) can be coupled to antibody mole­cules
without changing the antibody’s capacity to
bind to a specific antigen. Fluorescent dyes may
also be conjugated with complement. Labeled
complement is a versatile tool and can be employed
for the detection of antigen or antibody.

Sandwich Technique
The dyes can be conjugated to the Fc region of an
antibody molecule without affecting the specificity
of the antibody. For detection of antibod­ies by
immunofluorescence, the sandwich technique
can be employed. The antibody is first allowed to
B
react with unlabeled antigen, which is then treated
with fluorescent labeled antibody. A sandwich, is Fig. 16.14: Direct and indirect immunofluorescence.
thus formed, the antigen being in the middle and fluorochrome-labeled reagent. Indirect immuno­
labeled and unlabeled antibodies on either side. fluorescence is used to detect the presence of
antibodies in serum following an individual’s expo­
Types of Immunofluorescence sure to microorganisms.
There are two main kinds of fluorescent antibody In this technique, a known antigen is fixed
assays: direct and indirect. onto a slide. The test antiserum is then added, and
if the specific antibody is present, it reacts with
1. Direct Immunofluorescence (Fig. 16.14A) antigen to form a complex. When fluorescein-
Principle: In direct staining, the specific antibody labeled anti-immunoglobulin is added, it reacts
(the primary antibody) is directly conjugated with the fixed antibody. The slide is examined with
with fluorescein. It involves fixing the specimen the fluorescence microscope. The occurrence of
containing the antigen of interest onto a slide. fluorescence shows that antibody specific to the
Fluorescein-labeled antibodies are then added test antigen is present in the serum.
to the slide and examined with the fluorescence
microscope for a yellow-green fluorescence. The Uses
pattern of fluorescence reveals the antigen’s location. Diagnosis of syphilis: It is used to identify the
presence of Treponema pallidum antibodies in
Uses the diagnosis of syphilis (fluorescent treponemal
i. It is used to identify antigens, such as those antibody absorption (FTA-ABS).
found on the surface of group A streptococci. Advan­tages
ii. To diagnose enteropathogenic Es­cherichia i. The primary antibody does not need to be
coli, Neisseria meningitidis, Salmonella Typhi, conjugated with a fluorochrome.
Shigella sonnei, Listeria monocytogenes, ii. Increase the sensitivity of staining.
Haemophilus influenzae type b.
iii. To diagnose rabies virus. G. Radioimmunoassay (RIA)
One of the most sensitive techniques for detecting
Disadvantage antigen or antibody is radioimmunoassay
Separate fluorescent conjugates hence to be (RIA). The technique was first developed by two
prepared against each entigen to be tested. endocrinologists, SA Berson and Rosalyn Yalow, in
1960. The technique soon proved its own value for
measuring hormones, serum proteins, drugs, and
2. Indirect Immunofluorescence
vitamins at concentrations of 0.001 micrograms per
(Fig. 16.14B) milliliter or less. The significance of the technique
In indirect staining, the primary antibody is was acknowledged by the award of a Nobel Prize
unlabeled and is detected with an additional to Yalow in 1977, some years after Berson’s death.

www.ebook3000.com
Chap-16.indd 111 15-03-2016 11:12:09
112  |  Section 2: Immunology
Principle of RIA its extreme sensitivity (measuring picograms
The principle of RIA is based on competitive of antigen per milliliter).
binding of radiolabeled antigen (e.g. 125I and ii. To determine the presence of the hepatitis B
unlabeled antigen to a high-affinity antibody. The antigen.
labeled and unlabeled (test) antigens compete
for the limited binding sites on the antibody. This H. Enzyme-linked Immunosorbent Assay
competition is determined by the level of the (ELISA) (Fig. 16.15)
unlabeled (test) antigen present in the reacting Enzyme-linked Immunosorbent Assay, commonly
system. known as ELISA or EIA), is similar in principle to
RIA but the radioactive tag used in RIA techniques
Procedure can be replaced with an enzyme. When this enzyme
1. Measured quantities of labeled antigen is linked to an antibody and used to detect and
(radiolabeled) antigen (of the same kind being measure other antibodies or antigens, the assay is
tested) and antibody (specific to the antigen called the enzyme-linked immunosorbent assay
being tested) are mixed and incubated, one (ELISA). An enzyme conjugated with antibody
mixture with and one without added test reacts with a colorless substrate to generate a
sample. colored reaction product. Such a substrate is called
The labeled antigen is mixed with antibody a chromogenic substrate.
at a concentration that saturates the antigen- Enzyme-linked immunosorbent assay is
binding sites of the antibody molecule. highly sensitive, highly specific and less expensive
technique used in serology to detect antigens or
2. Then increasing amounts of the test sample
antibodies.
(unlabeled antigen) of unknown concentration
are added. The antibody does not distinguish
labeled from unlabeled antigen, and so the two Principle
kinds of antigens compete for available binding ELISA is based on two principles:
sites on the antibody. 1. Solid phase immunoassays are more widely
3. With increasing concentrations of test antigen used. It refers to the binding of either antigen or
(unlabeled antigen), more labeled antigen will antibody to a variety of solid materials, such as
be displaced from the binding sites. polyvinyl or polycarbonate wells or membranes
4. The antigen–antibody complex is washed to (discs) of polyacrylamide, paper or plastic or
remove unbound radiolabeled antigen from the metal beads or some other solid matrix.
mixture. The antigen is separated into ‘free’ and 2. Antigens and antibodies can be covalently
‘bound’ fractions after the reaction and their attached to an active enzyme (such as alkaline
radioactive counts measured. phosphatase or horseradish peroxidase and
5. The radioactivity associated with the antibody β-galactosidase), with the resulting complexes
is then detected by means of radioisotope still fully functional, both immunologically
analyzers and autoradiography. A little amount and enzymatically. Enzyme activity is used to
of bound radioactivity indicates that there is measure the quantity of antigen or antibody
large amount of antigen; and a large amount of present in the test sample. After all the
bound radioactivity indicates that there is little unreacted material is washed away, the
antigen in the sample. substrate for the enzyme is added (usually
6. The standard dose response or reference one that will yield a colored product, such
curve has to be prepared first for any reacting as p-nitrophenol phosphate for alkaline
system. This is plotted on graph by taking the phosphatase), and the conversion of the
ratio of bound radiolabeled antigen to free substrate from colorless to color is a measure of
radiolabeled antigen against varying known antigen-antibody interaction. The test is usually
amounts of unlabeled antigen and the antigen done using microtiter plates (96 w ­ ell) suitable
concentration of the test sample can be read for automation.
from this curve. This curve will allow the
measurement of antigen present by merely Types of ELISA (Fig 16.15)
counting the radioactivity present in the test 1. Indirect ELISA
antigen-antibody precipitated complexes. 2. Sandwich ELISA
3. Competitive ELISA
Uses 1. Indirect ELISA: The indirect immunosorbent
To measure the concentration of certain
i. assay detects antibodies rather than antigens.
hormones: such as insulin, testosterone, Serum or other sample containing primary
growth hormone, and glucagon because of antiboby is added to antigen-coated microtiter

Chap-16.indd 112 15-03-2016 11:12:09


Chapter 16: Antigen–Antibody Reactions  | 113

Fig. 16.15: Enzyme-linked immunosorbent assay (ELISA)

well. Any free antibody is washed away and positive. If the antigen is not recognized by the
the presence of antibody bound to the antigen absorbed antibody, the ELISA test is negative
is detected by adding an enzyme-conjugated because the unat­t ached antigen has been
secondary anti-isotype antibody (antibody 2), washed away, and no antibody-enzyme is
which binds to the primary antibody. Any free bound.
antibody 2 then is washed away, and a substrate 3. Competitive ELISA: In this technique, antibody
for the enzyme is added. The amount of colored is first incubated in solution with a sample
reaction product that forms is measured by containing antigen. The antigen–antibody
specialized spectrophotometric plate readers, mixture is then added to an antigen coated
which can measure the absorbance of all of microtiter well. The more antigen present
the wells of a 96-well plate in less than a few in the sampIe, the less free antibody will be
seconds. available to bind to the antigen-coated well.
2. Sandwich ELISA: The most frequently used Addition of an enzyme-conjugated, secondary
ELISA for detecting microbial antigen is the antibody specific for the isotype of the primary
sandwich solid-phase ELISA. In this technique, antibody can be used to determine the amount
the antibody (rather than the antigen) is of primary antibody bound to the well as in
immobilized on a microtiter well. The test an indirect ELISA. In the competitive assay,
sample is then exposed to the solid-phase however, the higher the concentration of
antibody, to which the antigen, if present, antigen in the original sample, the lower the
will bind. After the well is washed, a second absorbance.
enzyme-linked antibody specific for test
antigen is added and allowed to react with the USES OF ELISA
bound antigen. The conjugated antibody will
react with the antigen held to the solid-phase by ELISA has been used to detect antigens and
the first antibody, forming an antibody-antigen- antibodies of various microorgnisms.
antibody sandwich on the solid phase. After any
free second antibody is removed by washing, Examples
substrate is added, and the colored reaction 1. Parasites
product is measured.
If the antigen has reacted with the absorbed ∙∙ Entamoeba histolytica antigens in feces.
antibodies in the first step, the ELISA test is ∙∙ Toxoplasma antigens in the patient serum.

www.ebook3000.com
Chap-16.indd 113 15-03-2016 11:12:10
114  |  Section 2: Immunology
2. Bacteria and Northern blotting which detects mRNAs. It is a
∙∙ Haemophilus influenzae antigens in spinal variation of an ELISA.
fluid. A specific antibody in a mixture can also be
∙∙ β-hemolytic streptococcal antigen in spinal identified by Western blotting. Known antigens of
fluid. well-defined molec­ular weight are separated by
SDS-polyacrylamide gel (SDS-PAGE), and blotted
∙∙ Labile enterotoxin of E. coli in stools.
onto ni­trocellulose in this case. The separated
To detect antibody specific for Mycoplasmas, bands of known antigens are then probed with
Chlamydiae, Borrelia burgdorferi. the sample (test sample) suspected of containing
antibody specific for one or more of these antigens.
3. Viruses Reaction of an antibody with a band is detected
To detect antibody specific for hepatitis virus; by using either radiolabeled or enzyme-linked
∙∙ Herpes simplex viruses 1 and 2 secondary antibody that is specific for the species
of the antibodies in the test sample. The enzyme
∙∙ Respi­ratory syncytial virus (RSV)
substrate is subsequently added, which indicates
∙∙ Cytomegalovirus positive test. The substrate changes color in the
∙∙ Human im­munodeficiency virus (HIV) presence of enzyme and permanently stains the
∙∙ Rubella virus. nitrocellulose paper. The position of the band on
the paper indicates the antigen with which the
Cassette-based Membrane-bound ELISA antibody has reacted.
Assays
Cassette-based membrane-bound ELISA assays,
Application
designed for testing a single serum, can be i. Confirmatory testing for human immuno­
performed rapidly (often within 10 minutes) as deficiency virus.
compared with the 2–4 hours taken for microplate ii. Antibodies against microbes with numerous
ELISA. There is no need for microplate washers or cross-reacting antibodies.
readers. The result is read visually. Inbuilt positive
and negative controls are usually provided for J. Immunochromatographic Tests
validation of the test procedure.
A one-step qualitative immunochromatographic
Examples of Cassette ELISA tech­n ique has found wide application in
serodiagnosis due to its simplicity, economy
Used for the detection of HIV type 1 and 2 antibodies.
and reliability. A description of its use for Hbs
Antibody-capture ELISAs: Antibody-capture antigen detection illustrates the method. The test
ELISAs are particularly valuable for detecting IgM system is a small cassette containing a membrane
in the presence of IgG. Toxoplasmosis, rubella, impregnated with anti-Hbs antigen antibody
and other infections are diagnosed using this colloidal gold dye conjugate. The membrane is
technology. ex­posed at three windows on the cassette. The
test serum is dropped into the first window. As
Slot-blot and Dot-blot Assays the serum travels upstream by capillary action, a
Slot-blot and dot-blot assays force the target colored band appears at the second window (test
antigen through a membrane filter, causing it to site) if the serum contains Hbs antigen, due the
become affixed in the shape of the hole (a dot or formation of an Hbs antigen–antibody con­jugate
a slot). When test (pa­tient) serum is layered onto complex. This is the positive reaction. Absence of
the membrane, specific anti­bodies, if present, will a colored band at the test site indicates a negative
bind to the corresponding dot or slot of antigen. reaction. Simultaneously, a colored band should
Addition of a labeled second antibody and ap­pear in every case at the third window, which
subsequent development of the label allows visual forms an inbuilt control, in the absence of which
detection of the presence of antibodies based on the test is invalid. The test is claimed to be nearly
the pat­tern of antigen sites. as sensitive and specific as EIA tests.

I. Immunoelectroblot Techniques K. Immunoelectron microscopic Tests


Western Blotting (Immunoblotting): Identifi­ 1. Immunoelectron microscopy: When viral particles
cation of a specific protein in a complex mixture mixed with specific antisera are observed under
of proteins can be accomplished by a technique the electron microscope, they are seen to be
known as West­ern blotting, named for its similarity clumped. This is known as immunoelectron
to Southern blotting, which detects DNA fragments, microscopy. This finds application in the study

Chap-16.indd 114 15-03-2016 11:12:10


Chapter 16: Antigen–Antibody Reactions  | 115
of some viruses such as the hepatitis A virus and
™™ Western blotting is done for the detection of proteins
the viruses causing diarrhea. ™™ Immunoelectron microscopic tests include immuno­
2. Immunoferritin test: Ferritin (an electron- electronmicro­s copy, immunoenzyme test and
dense sub­stance from horse spleen) can be immuno­ferritin test.
conjugated with an­tibody, and such labeled
antibody reacting with an antigen can be
visualized under the electron micro­scope. IMPORTANT QUESTIONS
3. Immunoenzyme test: Some stable enzymes,
1. Name the various antigen–antibody reactions.
such as peroxidase, can be conjugated with
Describe the prin­ ciple and applications of
antibodies. Tissue sections carrying the precipitation reactions, giving suitable examples.
corresponding antigens are treat­e d with
2. Define agglutination reaction. Discuss the prin­
peroxidase labeled antisera. If the tissue section ciple and applications of agglutination reactions,
possesses specific antigen, then peroxidase giving suitable examples.
bound to the antigen can be visualized under the 3. Write short notes on:
elec­tron microscope, by microhistochemical a. Zone phenomenon (or) prozone.
methods. In im­m unoenzyme tests, some b. Immunodiffusion (or) gel diffusion
other enzymes, such as glucose oxidase, phos­ c. Immunoelectrophoresis
phatases and tyrosinase may also be included. d. Counterimmunoelectrophoresis (CIE) or coun-
tercurrenteletrophoresis (CIEP)
Key Points e. Rocket electrophoresis
™™ The antigen–antibody reaction occurs in three f. Coagglutination
stages g. Neutralization tests
™™ Precipitation test is a type of antigen–antibody h. Opsonization
reaction, in which the antigen occurs in a soluble i. Immunofluorescence tests
form. Immunodiffusion procedures are precipitation
reactions carried out in an agar gel medium. j. Radioimmunoassay (RIA)
Electrophoresis can be combined with precipitation k. ELISA-its principle and application.
in gels in a technique called immunoelectrophoresis.
Immunoelectrophoresis combines electrophoresis
with immunodiffusion for the analysis of serum
MULTIPLE CHOICE QUESTIONS (MCQs)
proteins 1. The prozone phenomenon is:
™™ Agglutination reactions: The interaction of particulate a. Zone of antibody excess
antigens with antibodies leads to agglutination b. Zone of antigen excess
reactions. Direct agglutination reactions can be: c. Zone of equivalence of antigens and antibody
slide agglutination test, tube agglutination test,
d. None of the above
heterophile agglutination tests and antiglobulin
(Coombs) test).Passive agglutination reaction 2. Which immunoglobulin class is the most efficient
depends on the carrier particles used to produce agglutination reaction?
™™ Complement-fixation reactions are serological tests a. IgG b. IgM
based on the depletion of a fixed amount of complement c. IgA d. IgE
in the presence of an antigen–antibody reaction, and are 3. Which immunoglobulin class is the most efficient
(i) Complement fixation test; (ii) Immune adherence to produce precipitation reaction?
test; (iii) Immobilization test; (iv) Cytolytic or cytocidal a. IgG b. IgM
reactions c. IgE d. IgA
™™ Neutralization reactions: In neutralization reactions, 4. Ring test is used for:
the harmful effects of a bacterial exotoxin or virus
a. C-reactive protein test
are eliminated by a specific antibody
b. Streptococcal grouping of Lancefield tecnique
™™ Opsonization is Immune opsonization or nonimmune
c. Both the above
opsonization
d. None of the above
™™ Immunofluorescence: Direct immunofluorescence
test is used to detect unknown antigen in a cell or 5. VDRL test is an example of:
tissue. Indirect immunofluorescence test is used for a. Agglutination test b. Flocculation test
detection of specific antibodies in the serum and c. Immunofluorescence d. All the above
other body fluids 6. Radial immunodiffusion can be used to estimate
™™ Radioimmunoassay (RIA) is a highly sensitive and the following immunoglobulin classes:
quanti­tative procedure that utilizes radioactively a. IgG b. IgM
labeled antigen or antibody c. IgA d. All the above
™™ The enzyme-linked immunosorbent assay (ELISA) 7. Counterimmunoelectrophoresis is used for detecting:
de­pends on an enzyme-substrate reaction that a. Hepatitis B antigens
generates a colored reaction product. Types of b. Cryptococcal antigens
ELISA are: 1. Indirect ELISA; 2. Sandwich ELISA; 3.
c. Neisseria meningitidis
Competitive ELISA
d. All the above

www.ebook3000.com
Chap-16.indd 115 15-03-2016 11:12:10
116  |  Section 2: Immunology
8. Tube agglutination test is used for serological 12. Direct immunofluorescence test may be used for
diagnosis of: detection of:
a. Enteric fever a. Rabies virus antigens
b. Infectious mononucleosis b. Antibodies in syphilis
c. Typhus fever c. Both the above
d. All the above d. None of the above
9. Which of the following is/are example/s of 13. Indirect immunofluorescence test may be used for
heterophile agglutination test? detection of:
a. Weil-Felix reaction a. Rabies virus antigens
b. Paul-Bunnel test b. Antibodies in syphilis
c. Streptococcus MG agglutination test c. Both the above
d. All the above d. None of the above
10. Which of the following is/are example/s of passive 14. ELISA can be used for detection of antibodies in:
agglutination test? a. HIV b. Rubella virus
c. Hepatitis B virus d. All the above
a. Latex agglutination test
b. Haemagglutination test 15. The technique of immunoblotting to analyse RNA
c. Coagglutination is named:
d. All the above a. Southern blot
b. Northern blot
11. Which of the following is/are example/s of c. Western blot
neutralization test? d. None of the above
a. Schick test
b. Antistreptolysin ‘0’ test ANSWERS (MCQs)
c. Nagler reaction 1. a; 2. b; 3. a; 4. c; 5. b; 6. d; 7. d; 8. d; 9. d; 10. d; 11. d; 12. a;
d. All the above 13. b; 14. d; 15. b

Chap-16.indd 116 15-03-2016 11:12:10


17
Chapter

Structures and Functions of the Immune System

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe cluster of differentiation (CD)
be able to: ∙∙ Discuss the following; Natural killer cells or NK cells;
∙∙ Differentiate between T and B cells in a tabulated killer cells or K cells or ADCC cells; human leukocyte
form antigen (HLA).

Introduction i. Thymus—primary lymphoid organ


ii. Bone marrow—primary lymphoid tissue.
The lymphoreticular system is responsible
for immunity. Lymphoreticular cells consist of B. Secondary (Peripheral)
lymphoid and reticuloendothelial components.
The lymphoid cells (lymphocytes and plasma cells)
Organs and Tissues
­are primarily concerned with the specific immune The secondary organs and tis­s ues serve as
response. The phagocytic cells (polymorphonuclear areas where lymphocytes may encounter and
leukocytes and macrophages), forming part of bind antigen, whereupon they proliferate and
the reticuloendothelial system, are primarily differentiate into fully mature, antigen-specific
concerned with the ‘scavenger’ functions of effector cells.
eliminating effete cells and foreign particles, thus Examples: Lymph nodes, spleen, various mucosal
contributing to nonspecific immunity by removing ­associated lymphoid tissues (MALT)—such as
microorganisms from blood and tissues. gut-associated lymphoid tissues (GALT), skin
associated lymphoid tissues (SALT)
Types of immune response
The immune response to an antigen, whatever its A. Central (Primary) Lymphoid Organs
nature, can be of two broad types: 1. Thymus: A T-cell Factory
Thymus is a flat, bilobed organ situated above
1. Humoral or Antibody-mediated the heart and is the site of T-cell development
Immunity (HMI or AMI) and maturation.
Humoral immunity is mediated by antibodies Function of the thymus: Immunological
produced by plasma cells. competence on the lymphocytes
The primary function of the thymus is the
2. Cellular or Cell-mediated Immunity (CMI) production of thymic lymphocytes. Precursor
Cellular immunity is mediated directly by cells from the bone marrow migrate into
sensitized lymphocytes. the thymus to the outer cortex where they
proliferate. As they mature and acquire T-cell
Organs and tissues of the immune surface markers, they move to the inner cortex
system where approx­imately 90% die. The other 10%
move into the medulla, become mature T-cells,
A. Primary (Central) Organs or Tissues and enter the bloodstream. The reason for this
The primary organs or tissues are where immature apparently wasteful process is not known. In the
lymphocytes mature and differentiate into antigen- thymus, the lymphocytes acquire new surface
sensitive mature B and T cells. antigens (‘Thy’ antigens).

www.ebook3000.com
118  |  Section 2: Immunology
Thymus (T)-dependent lymphocytes : B-cells and secrete various cytokines that are
Lymphocytes produced in the thymus are called required for development. A selection process
‘thymus (T) dependent lymphocytes’ or ‘T-cells’. within the bone marrow eliminates B-cells with
Lymphocyte proliferation in the thymus is not self-reactive antibody receptors like thymic
dependent on antigenic stimulation,unlike selection during T-cell maturation. Bone
in the peripheral organs. Differentiation marrow is not the site of B-cell development
and maturation are under the influence of in all species.
hormones such as thymopoietin and thymosin,
produced by the epithelial elements in the B. Peripheral (Secondary) Lymphoid Organs
thymus. 1. Lymph nodes: They are encapsulated bean-­
Thymus-dependent antigens: T-lymphocytes shaped structures containing a reticular network
are selectively seeded into certain sites in the packed with lymphocytes, macrophages, and
peripheral lymphatic tissues, being found in dendritic cells. Clus­tered at junctions of the
the white pulp of the spleen, around the central lymphatic vessels, lymph nodes are the first
arterioles, and in the paracortical areas of organized lymphoid structure to encounter
lymph nodes. These regions have been termed antigens that enter the tissue spaces.
‘thymus-dependent’. While thymectomy affects
CMI primarily, it also diminishes antibody Structure of lymph node: Lymph nodes have an
response to many types of antigens (thymus- indentation called the hilus where blood vessels
dependent antigens) such as sheep erythrocytes enter and leave the node. A typical lymph node
and bovine serum albumin. is surrounded by a fibrous capsule from which
Thymus and immune function: The importance trabeculae penetrate into the nodes.
of thy­mus in lymphocyte proliferation and Morphology: Morphologically, a lymph node
development of CMI is evident from the can be divided into three roughly concentric
lymphopenia, deficient graft rejection and regions:,), each of which supports a distinct
the so called ‘runt disease’ seen in neonatal microenvironment.
thymectomized mice. i. Cortex: The outermost layer, the cortex (B-cell
A congenital birth defect in humans (DiGeorge’s area),, contains lymphocytes (mostly B-cells),
syndrome) and in certain mice (nude mice) in macrophages, and follicular dendritic cells
which the thymus fails to develop are other arranged in primary follicles. After antigenic
evidence of the importance of the thymus. In challenge, the primary follicles enlarge into
both the cases, there is an absence of circulating secondary follicles, each containing a germinal
T-cells and of cell-mediated immunity and an center.
increase in infectious disease. ii. Paracortex (T-cell area): Beneath the cortex
2. Bursa of Fabricius: In birds, undifferentiated is the paracortex, which is populated
lymphocytes move from the bone marraw to largely by T-lymphocytes and also contains
the Bursa of Fabricius where B-cells mature. The interdigitating dendritic cells. The paracortex
bursa is also a site of lymphocytic proliferation is sometimes referred to as a thymus-
and differentiation. Stem cells from the yolk sac, dependent area in contrast to the cor­t ex,
fetal liver and bone marrow enter the bursa, which is a thymus-independent area.
proliferate and develop into immunocompetent iii. Medulla: The innermost layer of a lymph
‘bursal dependent’ or B-cells (to designate their node, the medulla (consisting of cellular cords
origin in the bursa). These migrate and seed containing T-cells, B-cells, abundant plasma
selective areas in the peripheral lymphoid cells and macrophages) In the medulla, the
organs—the mantle, germinal follicles and lymphocytes, plasma cells and macrophages
perifollicular regions of the spleen, and the are arranged as elongated branching bands
far cortical areas and medullary cords of (medullary cords). The cortical follicles and
lymphocytes. These are called ‘bursa-dependent’ medullary cords contain B-lymphocytes
or ‘thymus-independent areas’. B-lymphocytes and constitute the bursa dependent areas.
transform into plasma cells and secretes Paracortical area contains T-lymphocytes and
antibodies following antigenic stimulation. constitutes the thymus dependent area.
3. Bone marrow: In humans and other mammals,
Functions of lymph node
the bone marrow acts as the bursa equivalent
and is the site of B-cell origin and development. i. Lymph nodes act as a filter for lymph.
Arising from lymphoid progenitors, im­mature ii. They phagocytose foreign materials, including
B-cells proliferate and differentiate within microorganisms.
the bone marrow, and stromal cells within iii. They help in the proliferation and circulation
the bone marrow interact directly with the of T- and B-cells.
Chapter 17: Structures and Functions of the Immune System  | 119
iv. They enlarge following local antigenic Lymphocytes: Lymphocytes constitute 20%–40%
stimulation. of the body’s white blood cells and 99% of the cells
in the lymph.Many mature lymphoid cells are long-
2. Spleen
lived, and persist as memory cells for many years.
The spleen is the large secondary lymphoid organ
The lymphocytes can be broadly subdivided
located in the abdominal cavity.
into three populations—B-cells, T-cells, and
Structure of spleen: It has a capsule from that natural killer cells-on the basis of function and
extends a number of projections (trabeculae) into cell-membrane components. According to their
the interior to form a compartmentalized structure. size, lymphocytes can be clas­sified into small (5–8
The compartments are of two types, white pulp and μm ), medium (8–12 μm) and large (12–15 μm)
the red pulp. lymphocytes.

White pulp: The splenic white pulp surrounds Lymphatic recirculation: Lymphopoiesis takes
the branches of the splenic artery, forming a place mainly in the central lymphoid organs where
periarteriolar lymphoid sheath (PALS) populated they differentiate and mature before entering the
mainly by T-lymphocytes. Primary lymphoid circulation and then the peripheral lymphoid organs
follicles are attached to the PALS. These follicles and tissues. These populations of lymphocytes do
are rich in B-cells and some of them contain not remain distinct but mix together in a process
germinal centers which develop following antigenic known as ‘lymphocyte recirculation’. There is
stimulation. The lymphatic sheath surrounding the a constant traffic of lymphocytes through the
central arterioles is the thymus dependent area blood, lymph, lymphatic organs and tissues. This
of the spleen. The perifollicular region, germinal re­circulation ensures that following introduction of
centre and mantle layer form the bursa dependent an­tigen into any part of the body, lymphocytes of
(thymus-independent) areas. appropriate specificity would reach the site during
their ceaseless wandering and mount an immune
Red pulp: The splenic red pulp consists of a response. Recirculating lymphocytes are mainly
network of sinusoids. T-cells. B-cells tend to be more sessile. Chronic
Functions of spleen thoracic duct drainage will therefore result in
selective T-cell depletion.
i. Functions as the graveyard for blood cells.
ii. Mounting immune responses to antigens in the
blood stream. Differences between T- and B-Cells
Many tests help in differentiation of T- and B-cells
3. Mucosa-associated lymphoid tissue (MALT) (Table 17.1). These include:
The mucosa lining the alimentary, respiratory,
1. Thymus-specific antigens: Which are absent on
genitouri­nary and other lumina and surfaces are
B-cells.
constantly ex­posed to numerous antigens. These
vulnerable mem­brane surfaces are defended by 2. T-cell receptor (TCR): Which resembles but
a group of organized lymphoid tissues known differs from antibody, and CD2- and CD3-
collectively as mucosal-associated lymphoid tissue associated proteins on their surface.
(MALT). 3. Surface immunoglobulins: B-cells have
immuno­globulin on their surface.
Gut-associated lymphoid tissue (GALT): GALT 4. SRBC rosette: T-cells bind to sheep erythrocytes
includes the tonsils, adenoids, and Peyer’s patches forming rosettes (SRBC) or E rosette, through
in the intestine. CD2 molecule. B-cells do not bind.
Bronchial associated lymphoid tissue (BALT): 5. EAC rosettes: B-cells bind to sheep erythrocytes
Also occurs in the respiratory system. coated with anti­body and complement,forming
EAC rosettes, due to the presence of a C3
Mucosal or secretory immune system: Mucous receptor (CR2) on the B-cell sur­face.
membranes are an effective barrier to the
6. Microvilli: Viewed under the scanning
entrance of most pathogens, which contributes to
microscope, T cells are generally free of
nonspecific immunity. This indi­cates the existence
cytoplasmic surface projections, while B-cells
of a common mucosal or secretory immune system.
have an extensively filamentous surface, with
Cells of the lymphoreticular system numerous microvilli
7. Blast transformation: T-cells undergo blast
Lymphoid Cells: Cells of the Immune System transformation, evidenced by enhanced DNA
The cells responsible for both nonspecific and synthesis, on treatment with mitogens, such as
specific immunity are the white blood cells called phytohemagglutinin (PHA) or Concanavalin
leukocytes. A (Con A), while B-cells undergo sim­ilar

www.ebook3000.com
120  |  Section 2: Immunology
Table 17.1  Comparison of T-cells and B-cells differentiation and functional properties of the
cells. Thus a cell dis­playing CD1 is identified by
Property T cell B cell
the binding of antibodies against CD1. Each class
A. Origin Bone marrow Bone marrow of leucocyte displays a diagnostic pattern of CDs.
B. Maturation Thymus Bursal equivalent: Over 200 CD markers have been identified so far.
bone marrow,
Examples
Payer’s patches
∙∙ CD3 is expressed only by T-cells.
C. Location
∙∙ CD 19 is expressed only by B-cells.
1.  Peripheral blood 65–85% 15–25%
∙∙ CD64 is expressed only by monocytes.
2.  Lymph node 60–75% 30–35% ∙∙ CD66 is expressed only by granulocytes.
3. Spleen 25–45% 55–60% ∙∙ CD68 is expressed only by macrophages.
4.  Thoracic duct 80–90% 10–20% ∙∙ On the other hand, CD18 and CD45 are expr­
5. Thymus 96% Negligible essed by a variety of leucocyte types.
D. Thymus specific + – 2. Antigen recognition receptors
antigens These inc­lude membrane-bound (surface)
E.  CD3 receptor + – immunoglobulins (mlgs or slgs) in B-cells and
F. Surface – + T-cell receptors (TCRs) in T-cells. In contrast to
immunoglobulins CDs, which can serve as diagnostic feature for
G. Receptor for Fc – +
all leukocytes, antigen recog­n ition receptors
piece of IgG are limited to B- and T-lymphocytes only. These
H. SRBC rosette (E + –
receptors are required for B- and T-cells to be
rosette) antigen reactive. Reaction of antigens with mlgs
and TCRs activates B-cells and T-cells respectively,
I. EAC rosette (C3 – +
receptor leading to proliferation and differentiation.
J. Numerous – + b. On the Basis of Functions
microvilli, on
A. Regulatory T-cells
surface
1. Helper /inducer cell(TH): Helper/inducer
K. Blast transform­ + –
cell (TH), with CD 4 surface marker, MHC
ation with:
   a. anti-CD 3 – + class II restriction; generally stimulating
   b. anti-Ig + – and promoting the growth of T-cells and
   c. PHA + – macrophages. They help B-cells make antibody
   d. Concanavalin A – + in response to antigenic challenge; stimulate
   e. Endotoxins
cell mediated immunity. Based on the different
profiles of cytokines produced, two subsets are
transformation with bacterial endotoxins, identified. Th1 and Th2.
Staphylococcus aureus (Cowan 1 strain) or EB Th1 cells: Th1 cells produce mainly the cytokines
virus. interferon gamma (IFN-g) and interleukin-2
(IL2) which activate macrophages and T-cells
T-lymphocytes promoting CMI, destruction of target cells and
Types of T-cells killing of intracellular microbes, such as tubercle
and lepra bacilli.
Based on their surface markers, MHC restriction,
TH2 cells: TH2 cells produce mainly the
target cells and function, the following T-cells sub­
cytokines IL4, 5 and 6 which stimulate B-cells
types are recognised:
to form antibodies.
T-cells may be broadly classified into:
2. Suppressor T-cells (TS cells): These have CD8
surface marker and MHC class I restriction.
a. On the Basis of Surface Markers They can suppress B-cell and T-cell response.
Classification of lymphocytes on the basis of B. Effector cells
surface markers makes use of two important char­
1. Delayed type-hypersensitivity T-cells (T-cells):
acteristics:
They are involved in delayed hypersensitivity
1. Cluster of differentiation or cluster determinant and cell-mediated immune response.
(CD) 2. Cytotoxic T-cells(TC-cells): They are also called
The term cluster of differentiation (CD) refers CD8+ cells with CD8 surface marker and MHC
families of surface glycoprotein antigens that can class I restriction. They can kill and lyse target
be recognized by specific antibodies produed cells carrying new or foreign antigens, including
against them. These markers reflect the stage of tumour, allograft and virus infected cells.
Chapter 17: Structures and Functions of the Immune System  | 121
ii. B-cells and Plasma Cells i. They are not inducible by antigen.
ii. They lack the classic antigen-specificity of
B-cell Maturation
T-cells and B-cells.
The B-lymphocyte derived its letter designation iii. They are not restricted by MHC-encoded
from its site of maturation, in the bursa of Fabricius proteins.
in birds; and the bone marrow of mammals, iv. NK activity is ‘natural’ or ‘nonimmune’ as
including hu­mans and mice. B-cell differentiation it does not require sensitisation by prior
also takes place in the fetal liver and fetal spleen. antigenic contact.
About 5–15% of the circulating lymphoid pool v. They belong to a different lineage from T- and
are B cells defined by the presence of surface B-cells.
immunoglobulin.
Mature B-cell undergoes clonal proliferation on Functions: They are considered to be important
contact with its appropriate antigen. Interaction i. Immune surveillance
between antigen and the membrane-bound ii. Natural defence against virus infected and
antibody on a mature naive B-cell, as well as maliganant mutant cells.
interactions with T-cells and macrophages, iii. NK-cells are capable of nonspecific killing
selectively induces the activation and differentiation of virus-transformed target cells and are
of B-cell clones of corresponding specificity. In involved in allograft and tumour rejection.
this process, the B-cell divides repeatedly and
differentiates, generating a population of plasma b. Antibody-dependent Cell-mediated
cells and memory cells. Plasma cells, synthesize
Cytotoxicity (ADCC)
and secrete antibody. Memory cells circulate until
activated by specific antigen. A subpopulation of LGLs possesses surface
re­ceptors for the Fc part of Ig. They are capable
Plasma cell: They are fully differentiated
of lys­ing or killing target cells sensitised with
antibody-secreting effector cells of the B-cell
IgG antibodies. This is an example of a process
lineage. Antigenically stimulated B-cells undego
known as antibody-dependent cell-mediated
blast transformation, becoming successively
cytotoxicity (ADCC). This antibody dependent
plasmoblasts, intermediate transitional cells and
cellular cytotox­icity is distinct from the action of
plasma cells. Plasma cells are factories of antibody
cytotoxic T-cells, which is independent of antibody.
production. A plasma cell makes an antibody of a
ADCC-cells were formerly called killer (K)-cells but
single specificity, of a single immunoglobulin class
are now classified with NK-cells.
and allotype and a single light chain type only. An
exception is found in primary antibody response,
when a plasma cell producing IgM initiallyand later
c. Lymphokine-activated Killer (LAK)
it may switch to IgG production. Lymphocytes, Cells
lymphoblasts and transitional cells may also Lymphokine activated killer (LAK) cells are
synthesize Ig to some extent, while plasma cell is NK-lymphocytes treated with interleukin-2 (lL-2),
the best antibody producing cell. which are cytotoxic to a wide range of tumour cells
without affecting normal cells. IL-2 also acts as a
iii. Null Cells growth factor for NK-cells.
A small proportion (5-10%) of lymphocytes
that lack distinguishing phenotypic markers Phagocytic Cells
characteristic of T- or B-lymphocytes are called Phagocytic cells are the mononuclear macrophages
null cells. Because of their morphology, they are (of blood and tissues) and the polymorphonuclear
also known as large granular lymphocytes (LGL). microphages.
The member of this group is the
a. Natural killer (NK)-cells Mononuclear cells: The mononuclear phagocytic
b. Antibody dependent cellular cytotoxic (ADCC)- system consists of monocytes circulating in the
cells blood and macrophages in the tissues Both types
c. Lymphokine-activated killer (LAK)-cells. are highly phago­cytic and make up the monocyte-
The term NK-cell is sometimes used as a macrophage system.
common name for all null cells. Macrophages: Monocytes leave the circulation and
reach various tissues to become transformed into
a. Natural Killer (NK) Cells macrophages, with morphological and functional
Natural killer (NK) cells are derived from large features characteristic of the tissues. Macrophages
granular lymphocytes (LGL). They differ from spread throughout the animal body and take up
K-cells in being independent of antibody. NK-cells residence in specific tis­sues where they are given
differ Tc cells in other properties as well: special names, e.g. alveolar macrophages in the

www.ebook3000.com
122  |  Section 2: Immunology
lung, histiocytes in connective tissues, Kupffer cells Major Histocompatibility complex
in the liver, Mesangial cells in the kidney, microglial
cells in the brain and osteoclasts in the bone. The major histocompatibility complex (MHC) is
a remarkable cluster of genes that control T-cell
Functions of Macrophages recognition of self and nonself. MHC proteins
play a pivotal role in “presenting” antigens to
1. Phagoc ytosis: The primary function of T-cells. In fact, T-cells do not respond to foreign
macrophages is phagocytosis. peptides unless the antigen peptides are properly
2. Antigen presentation to T-cells to initiate “presented” (that is, offered in combination with
immune immune responses. a MHC molecule). In humans, the MHC is called
3. Secretion of cytokines to activate and promote the human leuko­c yte antigen (HLA) complex.
innate immune response. These proteins were first detected by their effect on
Macrophages secrete interleukin-1, interleukin-6, transplant rejection (that is, tissue incompatibility).
tumor necrosis factor, and interleukin-12 In 1980, Snell, Dausset and Benacerrof were
in response to bacterial interaction, which awarded the Nobel prize for their work on MHC
stimulate immune and inflammatory responses, and the genetic central of immune response.
including fever. One of the most potent activators
of macrophages is interferon gamma (IFN-g) HLA Complex
se­creted by activated TH cells.
The major an­tigens determining histocompatibility
Polymorphonuclear Microphages in human be­ings are alloantigens, characteristically
found on the surface of leukocytes. Human MHC
Microphages are the polymorphonuclear antigens are therefore synonymous with human
leucocytes of the blood.Because of the irregular- leucocyte antigens (HLA), and the MHC complex
shaped nuclei, granulocytes are also called of genes with the HLA complex.
polymorphonuclear leukocytes or PMNs. Three The HLA complex of genes is located on the
types of granulocytes exist: basophils, eosinophils, short arm of chromosome 6 (Fig. 17.1). It consists
and neutrophils. of three separate clusters of genes:
a. Neutrophils: They are actively phagocytic 1. HLA Class I comprising A, B and C loci: HLA
and form the predominant cell type in acute class I comprising A, Band C loci. Class 1
inflammation. The phagocytic property of proteins are encoded by the HLA-A, HLA-B and
neutrophils is nonspecific, except for its HLA-C genes in humans. HLA Class I antigens
augmentation by opsonins. (A, B and C) are found on the surface of virtually
b. Eosinophils: They possess phagocytic activity all nucleated cells and, in some species (i.e.
but only to a limited degree. They are found mice, but not humans), on red blood cells as
in large numbers in allergic inflammation, well.
parasitic infections and around antigen- They are the principal antigens involved in
antibody complexes. graft rejection and cell mediated cytolysis. Class
c. Basophils: Basophil leukocytes are found in the I molecules may func­tion as components of
blood and tissues (mast cells). Their cytoplasm hormone receptors.
has large numbers of prominent basophilic 2. Class II or the D region consisting of DR, DQ
granules containing heparin, histamine, and DP loci: Class II proteins are encoded
serotonin and other hydrolytic enzymes. by the HLA-D region. There are three main
Degranulation of mast cells, with release of sets: the DP, DQ and DR-encoded molecules.
pharmacologically active agents, constitutes the HLA Class II antigens are more restricted in
effector mechanism in anaphylactic and atopic dis­tribution, being found only on cells of the
allergy. immune system—macrophages, dendritic cells,
Dendritic cells: While macrophages are the activated T-cells, and particularly on B-cells.
major antigen presenting cells, dendritic cell also 3. Class III or the complement region: Class III or
performs this function. Dendritic cells are bone the complement region containing genes for
marrow derived cells of a lineage different from complement components C2 and C4 of the
the macrophages and T- or B-lymphocytes. They
possess MHC class II antigens.
They are more potent antigen-presenting cells
than macrophages and B-cells, both of which need
to be activated before they can function as antigen-
presenting cells (APCs). Dendritic cells are specially
involved in the presentation of antigens to T-cells
during the primary immune response. Fig. 17.1: HLA complex loci on chromosome
Chapter 17: Structures and Functions of the Immune System  | 123
classical pathway, as well as properdin factor B 4. An association between HLA types and diseases:
of the alternative pathway, heat shock proteins For example, strong association has been found
and tumor necrosis factors alpha and beta. between ankylosing spondylitis and HLA-B27,
HLA loci are multiallelic, that is, the gene rheumatoid arthritis and HLA-DR4, and many
occupying the locus can be anyone of several autoimmune conditions and HLA-DR3.
alternative forms (alleles). As each allele
determines a distinct product (antigen), the MHC RESTRICTION
HLA system is very pleomorphic. For example,
at least 24 distinct alleles have been identified Both CD4+ and CD8+ T-cells can recognize antigen
at HLA locus A and 50 at B. only when it is presented by a self-MHC molecule,
an attribute called self-MHC restriction. Both class
I and class II antigens operate in this phenomenon.
HLA Molecules
Cytotoxic T-lymphocytes from immunised mice
HL A antigens are two-chain glycoprotein are able to kill and lyse virus infected target
molecules anchored on the surface membrane cells only when the T-cells and target cells are of
of cells. Class I and class II MHC are membrane- the same MHC type. Helper T-cells can accept
bound glycoproteins that are closely related in both antigen presented by macrophages only when
structure and function. the macrophages bear the same class II MHC
molecules on the surface. For T-cells participating
Role of MHC Diversity in delayed type hypersensitivity the antigen has to
1. Transplantation : The MHC system was be presented along with class II MHC
originally identified in the context of
transplantation, which is an artificial event. Key Points
2. For protecting the species from the broadest
™™ The immune system is organized into several special
possible number of pathogens: The primary aim
tissues which are collectively termed as lymphoid or
of the MHC may be defence against microbes immune tissues
and not against the graft. ™™ The lymphoid organs, based on their functions,
3. Nonimmunological phenomena: Such as are classified into primary (central) and secondary
individual odour, body weight in mice and egg (peripheral) lymphoid organs
laving in chickens. ™™ There are three types of lymphocytes: B-cells, T-cells,
and natural killer cells (NK-cells)
HLA Typing ™™ T-cells perform two important functions: cytotoxicity
and delayed hypersensitivity
Antisera for HLA typing were obtained principally ™™ T-cells play a key role in regulating antibody
from multiparous women as they tend to production and CMI, and in suppression of certain
have antibodies to the HLA antigens of their immune responses
husbands, due to sensitisation during pregnancy. ™™ Bone marrow-derived lymphocytes are known as
Monoclonal antibodies to HLA antigens have B-lymphocytes or B-cells and perform two important
been developed. functions: First, they differentiate themselves into
Typing is done serologically by microcytotoxicity, plasma cells and produce antibodies. Second, they
can present antigen to helper T-cells
which tests for complement mediated lysis of
™™ NK-cells have the ability to kill certain virally infected
peripheral blood lymphocytes with a standard set
cells and tumor cells without prior sensitization
of typing sera. However, serological typing is not
™™ The MHC in humans is known as HLA complex. In
possible for HLA-DR antigens, which are detected humans, the HLA complex of genes is located on
by the mixed leucocyte reaction (MLR) and primed short arm of chromosome 6 containing several genes
lymphocyte typing (PLT), respectively. Genetic that are critical to immune function
methods are being used increasingly for HLA-typing ™™ The genes encode MHC proteins that are classified
in advanced centres. These employ restriction into three groups or classes known as the class I, class
fragment length polymorphism (RFLP) and gene II, and class III molecules
sequence specific oligonucleotide probe typing ™™ HLA typing or tissue typing are usually performed
for 1. Tissue transplantation; 2. Disputed paternity;
3. Anthropological studies; 4. An association between
Uses of HLA Typing HLA types and diseases.
1. Tissue transplantation: HLA typing is used
primarily for testing compatibility between
Important questions
recipients and potential donors before tissue
transplantation. 1. Differentiate between T- and B-cells in a tabulated
2. Disputed paternity. form.
3. Anthropological studies. 2. Give an account of lymphocytes.

www.ebook3000.com
124  |  Section 2: Immunology
3. Write short notes on: 4. All the following statements are true for helper
a. Subsets of T-lymphocytes T-cells except that they:
b. B lymphocytes a. Carry CD4 marker
c. Null cells (or) Large granular lymphocytes (LGL) b. Help or induce immune responses
d. Natural killer cells or NK-cells c. Kill intracellular microorganisms by secreting
cytokines
e. Killer cells or K-cells or ADCC-cells
d. Recognize antigen in association with class I
4. Write briefly on:
MHC
a. Major histocompatibilty complex (MHC)
5. Class II MHC antigens are present on:
b. Human leukocyte antigen (HLA)
a. Macrophages
c. HLA typing b Monocytes
d. MHC restriction c. Activated T lymphocytes (CD4)
d. All the above
Multiple choice questions (MCQs) 6. Class I proteins are encoded by:
a. HLA-A, -B, and -C loci
1. All the following are examples of secondary- b. HLA-DR, HLA-DQ, and HLA-DP loci
lymphoid organs except: c. Complement loci that encode C2 and C4
a. Lymph nodes d. Complement loci that encode factor B
b. Thymus 7. Which of the following HLA types is associated
c. Spleen with ankylosing spondylitis?
d. Mucosa-associated lymphoid tissues a. HLA-B27 b. HLA-DR4
2. The CD4+ T-cells that recruit and activate phagocytic c. HLA-DP d. None of the
cells acting against intracellular microbes are called: above
a. Th-0 cells b. Th-1 cells 8. Which of the following HLA types is associated
c. Th-2 cells d. Antigen-present- with rheumatoid arthritis?
ing cells a. HLA-B27 b. HLA-DR4
3. The molecule expressed on surface of the mature c. HLA-Al d. None of the
T-cells is: above
a. CD 19 b. CD 8 Answers (MCQs)
c. CD 3 d. CD 1 1. b; 2. c; 3. c; 4. d; 5. d; 6. a; 7. b; 8. b
18
Chapter

Immune Response

Learning Objectives

After reading and studying this chapter, you should ∙∙ Discuss monoclonal antibodies—principle, technique
be able to: and applications
∙∙ Differentiate between primary and secondary ∙∙ Describe the following: cytokines; immunological
humoral immune responses tolerance.

Definition iv. It provides immunological surveillance and


immunity against cancer.
The immune response is the specific reactivity
induced in a host by an anti­genic stimulus. For
the generation of immune response, antigen must
Humoral immunity
interact with and activate a number of different cells. Synthesis of Antibody
In addition, these cells must interact with each other. On exposure to antigen, antibody production
follows a characteristic pattern (Fig. 18.1). The
Type of immune response production of antibodies consists of the following
The immune response can be divided into two steps:
types: the humoral (antibody mediated) and the
cellular (cell mediated) types.
1. Lag Phase
A lag phase, the immediate stage following anti­
a. Antibody-mediated Immunity (AMI) genic stimulus during which no antibody is
detectable in circulation.
1. Provides primary defence against most
extracellular bacterial pathogens.
2. Log Phase
2. Helps in defence against viruses that infect
through the respiratory or intestinal tracts. A log phase in which there is steady rise in the titer
3. Prevents recurrence of virus infections. of antibodies.
4. It also partici­p ates in the pathogenesis of
3. A Plateau or Steady State
immediate (types 1, 2 and 3) hypersensitivity
and certain autoimmune diseases. There is an equilibrium between antibody synthesis
and catabolism.
b. Cell-mediated Immunity (CMI)
4. The Phase of Decline
i. Protects against fungi, viruses and facultative
The amount of antibody then declines due to the
intracellular bacterial pathogens like
clearing of antigen–antibody com­plexes and the
Mycobacterium tuberculosis, Mycobacterium
natural catabolism of the immunoglobulin, i.e. the
leprae, Brucella and Salmonella, and parasites
catabolism exceeds the production and the titer
like Leishmania and trypanosomes.
falls (Fig 18.1).
ii. It also participates in the rejection of
homografts and graft-versus-host reaction.
iii. It mediates the pathogenesis of delayed (type Primary and Secondary Responses
4) hypersensitivity and certain autoimmune The kinetics and other characteristics of the
diseases. humoral re­sponse differ considerably depending

www.ebook3000.com
126  |  Section 2: Immunology
produced is 10 or more times greater than during
the primary response.
Priming dose and booster doses: A single
injection of an antigen helps more in sen­sitising or
priming the immunocompetent cells pro­ducing the
particular antibody than in the actual elaboration
of high levels of antibody. Only by subsequent
injections of the antigen effective levels of antibody
are usually induced. The first injection is known as
the ‘priming’ dose and subsequent injections as
‘booster’ doses. With live vaccines, a single dose is
Fig. 18.1: Primary immune response. An antigenic stimulus
sufficient as multiplication of the organism in the
1. Latent period; 2. Long phase (rise in titer of serum antibody);
3. Steady stage of antibody titer; 4. Decline of antibody titer body provides a continuing antigenic stimulus that
acts as both the priming and booster dose.

on whether the hu­moral response results from Negative phase: If the same animal is subsequently
activation of naive lymphocytes (primary response) exposed to the same antigen al­ready carrying the
or memory lymphocytes (secondary re­sponse). specific antibody in circulation, a temporary fall in
In both cases, activation leads to production of the level of circulating antibody oc­curs due to the
se­creted antibodies of various isotypes, which combination of the antigen with the preexisting
differ in their ability to mediate specific effector an­tibody. This is known as the ‘negative phase’. It is
functions. followed by an increase in the titre of the antibody
ex­ceeding the initial level.
a. Primary Humoral Response
The first contact of an exogenous antigen with an
Fate of antigen in tissues
individ­ual generates a primary humoral response, The manner in which an antigen is dealt with in
characterized by the production of antibody- the body depends on factors such as the physical
secreting plasma cells and mem­ory B-cells. and chemical nature of the antigen, its dose and
When first introduced the antigen selects the route of entry, and whether the antigenic stimulus
cells that can react with it. In all cases, however, a is primary or secondary.
primary response to antigen is characterized by a
Physical and chemical nature of the antigen:
lag phase, subsequent clonal expansion, and dif­
Particulate antigens are removed from circulation
ferentiation into memory cells or plasma cells. The
in two phases. The first is the nonimmune phase
duration of the lag phase varies with the nature of
during which the antigen is engulfed by the
the antigen.
phagocytic cells, broken down and eliminated.
During a primary humoral re­sponse, IgM is
The phase of immune elimination begins with the
secreted initially, often followed by a switch to an
appearance of the specific antibody, during which
increasing proportion of IgG. Depending on the
persis­tence of the antigen, a primary response
can last for various periods, from only a few days
to several weeks.

b. Secondary Humoral Re­sponse (Fig. 18.2)


On subsequent exposure, there is a shorter lag
before antibody can be detected: the main isotype
is IgG. The memory B-cells formed during a
primary response stop dividing. These cells have
variable life spans, with some persisting for the life
of the individual.
Activation of memory cells by antigen results
in a sec­ondary antibody response that can be
distinguished from the primary response in several
ways. The secon­dary response has a shorter lag
period, reaches a greater magnitude, lasts longer
Fig. 18.2: Effect of repeated antigenic stimulus. A, B, C
and is also char­acterized by secretion of antibody antigenic stimuli. 1. Primary immune response; 2. Secondary
with a higher affinity for the antigen, and isotypes immune response; 3. Negative phase; 4. High level of antibody
other than IgM predominate. The level of antibody following booster injection
Chapter 18: Immune Response  | 127
antigen–antibody complexes are formed and are present antigens to T-cells, particularly during the
rapidly phagocytozed, resulting in an accelerated secondary response.
disappearance of the antigen from circulation.
2. T-cell and B-cell Activation
Soluble antigens: Three phases can be recognised
with soluble antigens-equilibration, metabolism The activation of resting CD4 (Helper T/T H)
and immune elimination. cells require two signals. The first signal is a
combination of the T-cell receptor (TCR) with the
Route of entry: Antigens introduced intravenously MHC class II complexed antigen. The second is
are rapidly localized in the spleen, liver, bone the costimultory signal, i.e. interleukin-1 (IL-1)
marrow, kidneys and lungs. They are broken down which is produed by the APC. The activated TH
by the reticuloendothelial cells and excreted in the cell forms interleukin-2 and other cytokines such
urine, about 70–80% being thus eliminated within as interleukin-4, IL-5 and IL-6 required for B-cell
one or two days. In contrast, antigens introduced activation. These interleukin-4 (formerly known
subcutaneously are mainly localized in the as B-cell stimulatory factor I), IL-5 (B-cell growth
draining lymph nodes, only small amounts being factor, BCGF) and IL-6 (formerly called B-cell
found in the spleen. stimulatory factor, BCSF-2) activate B-cells which
Speed of elimination: The speed of elimination have combined with their respective antigens to
of an antigen is related to the speed at which it clonally proliferate and differentiate into antibody-
is metabolised. Protein antigens are generally secreting plasma cells.
eliminated within days or weeks, whereas B-cells carry surface receptors which consist of
polysaccharides which are metabolized slowly, IgM or other immunoglobulin classes. A plasma
persist for months or years. cell secretes an antibody of a single specificity of a
single antibody class (IgM, IgG or any other single
class) depending upon these receptors. However,
Production of antibodies primary antibody response is characterized by the
The majority of antigens will stimulate B-cells initial production of IgM, and and later switching
only if they have the assistance of T-lymphocyte over to form IgG. Class switching is influenced by
helper (TH) cells. Antigens can be divided into different combinations of lyniphokines produced
two categories based on their apparent need by helper T (T H)-cells. Under the direction of
for for T H cells for the induction of antibody cytokines produced by effector T-helper (TH) cells,
synthesis (i) those that require Th cells, referred some B-cells become programmed to produce
To as T-dependent antigens (TD-antigens) such antibodies other than IgM. Following antigenic
as proteins and erythrocytes; and (ii) those that stimulus, not all B-lymphocytes are converted into
do not require TH cells, called T-independent plasma cells. A small proportion of activated B-cells
antigens (TI-antigens), such as polysaccharides become long-lived memory cells which produce a
and other structurally simple molecules with secondary type of response to subsequent contact
repeating epitopes. Immune response to an antigen with the antigen. During secondary antigenic
is brought about by three types of cells—antigen stimulus,the increased antibody response is due to
processing cells (APC-principally macrophages the memory cells induced by the primary contact
and dendritic cells), T-cells and B-cells. The with the antigen.
sequence of events is as follows. Cytotoxic T (CD8/Tc)-cells are activated when
they contact antigens presented along with MHC
1. Antigen Processing and Presentation class I molecules. On contact with a target cell
For successful development of antibody response carrying the antigen on its surface, the activated
to a T-dependent antigen, the antigen must be Tc cells release cytotoxins that destroy the target,
associated with MHC class II molecules on the which may be virus infected or tumour cells. Some
surface of an antigen-presenting cell (APC). APC Tc cells also become memory cells.
can ingest antigen, degrade it and present it to
T-cells. T-cell is able to recognise only when the Monoclonal Antibodies
processed antigen is presented on the surface of Monoclonal antibodies : When a clone of
APC, in association with MHC molecules to the lymphocytes or plasma cells undergoes selective
T-cell carrying the receptor (TCR) for the epitope. proliferation, as in multiple myeloma, antibodies
The antigen has to be presented complexed with a single antigenic specificity accumulate.
with MHC Class II in the case of CD4 (Helper T/ Such antibodies produced by a single clone and
TH) cells, and for CD8 (cytotoxic T/Tc) cells with directed against a single antigenic determinant
MHC Class I molecules. B cells, which possess are called monoclonal antibodies e.g. plasma cell
surface Ig and MHC Class II molecules, can also tumour (myeloma). In myeloma, antibodies are

www.ebook3000.com
128  |  Section 2: Immunology
produced by a single clone of plasma cells directed 1. Selection of antigen: Monoclonal antibodies can
against a single antigenic determinant and hence be produced against any substance recognized
the antiboies are homogeneous and monoclonal. as an antigen by the immune system of the
animal being injected.
Hybridoma technology: The discovery of the
2. Animal immunization: Animal (usually mouse)
hybridoma technology for the production of
is immunised with a pure selec­tive antigen ,it
unlimited quantities of identical monoclonal
is killed and B lymphocytes are harvested from
antibodies of the same Ig class, possessing uniform
the spleen or lymph node. A suspension of
specificity, affinity and other properties, created
spleen cells (B cells) is prepared.
a revolution in immunology by opening up
numerous diagnostic, therapeutic and research 3. Fusion of splenic lymphocytes and myeloma
applications. Monoclonal antibodies against cells: A suspension of splenic cells is then fused
several antigens are now available commercially. with a myeloma cell-line by incubating in the
presence of polyethylene glycol.
By laboratory manipulation, Kohler and
Because cells cannot remain viable in cell cul­
Millstein (1975) prepared a hybrid cell line
ture for very long, they must be fused together
(hybridoma) by fusion of a mouse myeloma cell
with cells that are able to survive and multiply
with an antibody producing lymphocyte from
in tissue culture, that is, the continuously
spleen or lymph node of the-same inbred strain
propagating, or immortal cells, of multiple
of mouse. In recognition of the great importance
myeloma (a malignant tumor of antibody-
of this hybridoma technology, the Nobel Prize for
producing plasma cells).
Medicine was awarded to them in 1984.
4. Selection of hybrid lymphocyte-myeloma
cells: Lymphocytes from the spleen of mice
Procedure (Fig. 18.3) immunised with the desired antigen are fused
The production of monoclonal antibodies involves with mouse myeloma cells grown in culture
the following steps listed below: which do not form immunoglobulins and

Fig. 18.3: Production of a monoclonal antibody


Chapter 18: Immune Response  | 129
are deficient in the enzyme hypoxanthine 2. Nutritional Status
phosphoribosyl transferase (HPRT). The fused Malnutrition affects host susceptibility to certain
cells are placed in basal culture medium (HAT microorganisms, especially bacteria, e.g. protein
medium containing hypoxanthine, aminopterin calo­rie malnutrition suppresses both humoral and
and thymidine) which does not permit the cellular immune responses.
growth of the enzyme deficient myeloma cells.
Thus, fused hybridoma cells survive in the 3. Route of Administration
selective medium and can be recognized by There is better humoral immune response
their ability to grow indefinitely in the medium. following parenteral administration of antigen than
Unfused anti­body-producing lymphoid cells die oral or nasal routes.
after several multi­plications in vitro because
they are not immortal, and unfused myeloma
4. Age
cells die in the presence of the toxic enzyme
substrates. The only surviving cells will be true i. Embryonic life: The embryo is immunologically
hybrids. These hybrid cells, called hybridomas immature. The capacity to produce antibodies
(they are hybrids of the two cells). starts only with the development and
5. Cloning the hybridoma cells: The single hybrid differentiation of lymphoid or­gans. When
cells producing the desired antibody must be the potential immunocompetent cell comes
isolated and grown as a clone. Two techniques into contact with its specific antigen during
can be used: (a) limiting dilution and (b) growth embry­onic life, the response is elimination of
in an agar gel medium. the cell or induction of tolerance.
6. Screening for desired antibodies: The growth ii. Infant: The infant has to depend on itself for
medium supernatant from the mi­crodilution antibody production from 3–6 months of age.
tray wells in which the hybridoma cells are However, full competence is acquired only by
growing is then tested for the presence of the about 5–7 years for IgG and 10–15 years for
desired an­tibody. IgA.
7. Mass production of monoclonal antibodies:
Clones producing antibodies against the 5. Multiple Vaccines
desired antigen are selected for continuous
Antigenic competition: The effects may vary
cultivation. Such hybr idomas can be
when two or more antigens are administered
maintained indefinitely in culture and will
simultaneously, Antibodies may be produced
continue to form monoclonal antibodies. They
against the different antigens, or antibody response
may also be injected intraperitoneally in mice.
to one or the other of the antigens may be
Within days, a tumor known as a hybridoma
enhanced, or the response to one or more of them
develops and monoclonal antibodies may
may be diminished (antigenic competition).
be obtained by harvesting the ascitic fluid
produced. Ascitic fluid can be re­moved from Examples
mice many times over the animals’ lifetimes. i. When toxoids are given along with bacterial
vaccines (for example, triple vaccine
Uses of Monoclonal Antibodies containing diphtheria and tetanus toxoids
1. They are routinely used in the typing of tissue. along with Bordetella pertussis vaccine), the
2. Use in the identification and epidemiological response to the toxoid is potentiated.
study of infectious microorganisms. ii. When diphtheria and tetanus toxoids are given
3. Use in the identification of tumor and other together, with one in excess, the response to
surface antigens. the other is inhibited.
4. Use in the classification of leukemias. iii. When triple antigen is given to a person
5. Use in the identification of func­tional popul­ who had earlier received a primary dose of
ations of different types of T-cells. diphtheria toxoid, the response to the tetanus
and pertussis antigens will be diminished.
Factors Influencing Antibody Production
1. Genetic Factors 6. Adjuvant
Adjuvant is any substance that enhances the
Response in different individuals to same antigen
immunogenicity of an antigen.
varies due to genetic factors. Persons capable
of responding to a parti­cular antigen are called Types of adjuvants
responders and those who cannot respond are a. Depot: Repository adjuvants such as aluminium
termed as nonresponders. The IR (immune hydroxide or phosphate, and Freund’s
response) genes control this property. incomplete adjuvant (water in archis oil).

www.ebook3000.com
130  |  Section 2: Immunology
b. Bacterial: Freund’s complete adjuvant is the lymphocytes and has no antimitotic activity.
Freund’s incomplete adjuvant along with a It selectively inhibits helper T-cell activity.
suspension of killed tubercle bacilli. vi. Antilymphocyte serum (ALS): Antilymphocyte
c. Chemical: Silica particles, beryllium sulfate and serum (ALS) is a heterologous antiserum raised
endotoxins activate macrophages. against lymphocytes. Antibody pre­p ared
Action of Adjuvants against thymus cells is called antithymocyte
a. Sustained release of antigen from depot serum (ATS). The corresponding globulin
b. Liberation of lymphocytes activating factor prepara­tions are called ALG and ATG. They
c. Lymphocytes stimulation-B-cell, T-cell or both. were used to prevent graft rejection.
d. Stimulate CMI. ALS acts primarily against T-lymphocytes
and therefore specifically on cell-mediated
Freund’s Complete Adjuvant
immunity. Humoral antibody response to thymus-
The most potent adjuvant is Freund’s complete
independent antigens is unaffected and may even
adjuvant, which is the incomplete adjuvant
be enhanced. ALS acts only against lymphocytes
along with a suspension of killed tubercle bacilli.
in circulation and not cells in lymphoid organs.
Besides increasing the humoral immune response,
As ALS is a foreign protein, its ef­fect is decreased
it induces a high degree of cellular immunity
on repeated administration, which may also lead
(delayed hypersensitivity) as well. It is unsuitable
to serum sickness and other hypersensi­t ivity
for human use as it produces a local granuloma.
reactions.
7. Immunosuppressive Agents
8. Effect of Antibody
These inhibit the im­mune response. They are
Passive administration of the homologous antibody
useful in certain situations like transplantation,
will suppress specifically the humoral immune
when it becomes necessary to prevent graft
response to an antigen. The ac­tion appears to be
rejection.
by a feedback mechanism.
Examples: X-irradiation, radiomimetic drugs,
corticosteroids, antimetabolites and other cytotoxic 9. Superantigens
chemicals, and antilymphocytic serum
i. X-irradiation: Anti­body response is suppressed Superantigens are certain protein molecules that
by sublethal whole body irradiation. activate very large numbers of T-cells irrespective of
ii. Radiomimetic drugs: They belong in general their antigenic spe­cificities such as staphylococcal
to the class of alkylating agents (for example, enterotoxins.
cyclophos­phamide, nitrogen mustard). In
human beings, cyclo­p hosphamide given 10. Mitogens
for three days after the antigen completely Mitogens are certain substances that induce
suppresses the antibody response. It is much division of lymphocytes and other cells.
less effective when given before the antigen.
iii. Corticosteroids: Corticosteroids cause depletion CELL-MEDIATED IMMUNE RESPONSES
of lymphocytes from the blood and lymphoid
organs. Therapeutic doses have little effect on The term ‘cell-mediated immunity’ (CMI) refers
the antibody formation in human beings. They to the specific immune responses which involves
inhibit the induction and manifestations of T-Iymphocyte-mediated functions that do not
delayed hypersensitivity in human beings. involve antibodies. This form of immunity can
iv. Antimetabolites: They include folic acid be transferred from donor to recipient with intact
antagonists (methotrexate), alkylating agents lymphocytes, but not with antisera, hence it is
(cyclophosphamide) and analogues of called cell-mediated immune reaction.
purine (6-mer­captopurine, azathioprine),
cytosine (cytosine arabi­noside) and uracil Scope of CMI
(5-fluorouracil). CMI participates in the following immunological
Antimetabolites are substances that interfere functions:
with the synthesis of DNA, RNA or both and 1. Delayed hypersensitivity (type IV hyper­
thus inhibit cell division and differentiation sensitivity).
necessary for humoral and cellular immune 2. Immunity in infectious diseases caused by
responses. Many antimetabolites find clinical obligate and facultative intracellular parasites.
application in the preven­tion of graft rejection. 3. Transplantation immunity and graft-versus-
v. Cyclosporine: The drug most widely used host reaction.
now for immunosup­pression is cyclosporine, 4. Immunological surveillance and immunity
a cyclic polypeptide. It is not cytotoxic for against cancer.
Chapter 18: Immune Response  | 131
5. Pathogenesis of certain autoimmune diseases Interferons, growth factors and others were
(for example, thyroiditis, encephalomyelitis). found to have similar effects. Therefore, all of them
have been grouped under the term cytokines.
Induction of CMI Recently cy­tokines have been grouped into
Antigen-specific cell-mediated immunity is the following categories or fami­lies: chemokines,
mediated by T-lymphocytes. A second, smaller hematopoietins, interleukins, and members of
population of cells includes natural killer (NK) and the tumor necrosis factor (TNF) family. Some
killer (K) cells. Cell-mediated immune response examples of these cytokine families are provided
can also be divided into primary and secondary in Table 18.1.
cell-mediated immune responses.
Features of Important Cytokines
Primary Cell-mediated Immune Response Various cytokines are shown in Table 18.1.
Foreign antigen is presented by antigen­presenting
celIs (APCs) to T-cells leading to their activation. A. Interleukins
Each T-cell bears on its surface a specific receptor Interleukin-1
(TCR) for one epitope and combines only with It is a stable polypeptide retaining its activity up
antigens carrying that epitope. On contact to 56°C and between pH 3 and pH 11. IL-l occurs
with the appropriate antigen, T-cells undergo in two molecular forms, IL-l alpha and beta.
blast transformation, clonal proliferation and IL-l is principally secreted by macrophages and
differentiation into memory cells and effector cells monocytes but can be produced by most other
providing CMI. T-cells recognise antigens only nucleated cells also. Its production is stimulated
when presented with MHC molecules. by antigens, toxins, injury and inflammatory
The T-cell group is composed of: processes and inhibited by cyc1osporin A,
1. Helper T (Th)-cells which react with antigens corticosteroids and prostaglandins.
presented on the surface of macrophages
or other cells, complexed with MHC class Immunological Effects of IL-1
II molecules. They then release biological i. Stimulation of T-cells for the production of IL2
mediators (lymphokines) which activate and other lymphokines
macrophages, enabling them to kill intracellular ii. B-cell proliferation and antibody synthesis
parasites; and
iii. Neutrophil chemotaxis and phagocytosis
2. T-cytotoxic cells (Tc-cells), which recognize
iv. It mediates a wide range of metabolic, physio­
antigen on the surface of cells, in association
logical, inflammatory and hematological
with MHC class I molecules, secrete lympho­
effects
kines and destroy the target cells.
Together with the tumor necrosis factor (TNF),
Secondary Cell-mediated it is responsible for many of the hematological
changes in septic shock
Immune Response
If the same host is subsequently exposed to the Interleukin-2
same antigen, then the secondary cell-mediated It is the major activator of T- and B-cells and
immune response is usually more pronounced and stimulates cytotoxic T-cells and NK-cells. It
occurs more rapidly. converts some null cells (LGL) into lymphokine-
activated killer (LAK) cells which can destroy NK
CYTOKINES resistant tumor cells. This property has been used
in the treatment of certain types of cancer.
Cytokines (Greek cyto, cell and kinesis, movement)
are biologically active substances produced by Interleukin-3
cells that influence other cells. Biologically active IL-3 is a growth factor for bone marrow stem cells.
substances released by activated T-lymphocytes It is also known as the multicolony-stimulating
were called lymphokines. When released from factor (multi-CSF).
mononu­clear phagocytes, these proteins are called Interleukin-4
monokines. IL-4 activates resting B-cells and acts as a B-cell
The term interleukin was introduced for those differentiating factor. It also acts as a growth factor
products of leucocytes which exert a regulatory for T-cells and mast cells. It enhances the action
influence on other cells; and if their effect is of cytotoxic T-cells. Formerly, it was known as
to stimulate the growth and differentiation of the B-cell growth factor-l (BCGF-1). It may have a
immature leukocytes in the bone marrow, they are role in atopic hypersensitivity as it augments IgE
called colony-stimulating factors (CSFs). synthesis.

www.ebook3000.com
132  |  Section 2: Immunology
Table 18.1  Cytokines: Hormones of the immune system-EDIT
Cytokine Main sources Major functions
A. Interleukins
IL-l (Alpha and b) Macrophages and other cell types Proliferation and differentiation ofT, B and other cells;
pyrogenic; induce acute phase proteins; bone marrow cell
proliferation
IL-2 T-cells Promote growth and differentiation of T- and B-cells,
cytotoxicity ofT and NK cells, secretion of other lymphokines
IL-3 T-cells Multi-CSF
IL-4 TH-cells Proliferation of B- and cytotoxic T-cells; increase IgGI and IgE
production; enhance MHC class II and IgE receptors
IL-5 TH-cells Proliferation of eosinophils, stimulate IgA and IgM production
IL-6 TH-cells Promote B-cell differentiation; IgG production, acute phase
proteins
IL-7 Bone marrow, spleen, stromal cells B- and T-cell growth factor
IL-8 Macrophages, others Neutrophil chemotactic factor
IL-9 T-cell T-cell growth and proliferation
IL-I0 T-, B-cells, macrophages Inhibit IFN production and mononuclear cell functions
IL-11 Bone marrow stromal cells Induce acute phase proteins
IL-12 T-cells Activate NK-cells
IL-13 T-cells Inhibit mononuclear cell functions
B. Colony-stimulating Factors
GM-CSF T-cells, macrophages, fibroblasts T-cell and macrophage growth stimulation
G-CSF Fibroblasts, endothelium Granulocyte growth stimulation
M-CSF roblasts, Fibroblasts, endothelium Macrophages growth stimulation
endothelium
C. Tumor Necrosis Factors
TNF-α Macrophages, monocytes Tumor cytotoxicity, lipolysis, wasting, acute phase proteins,
phagocytic cell activation, antiviral and antiparasitic effects,
endotoxic shock
TNF-β T-cells Induce other cytokines
D. Interferons
IFN-α Leukocytes
IFN-β Fibroblasts Antiviral activity
IFN-γ T-cells Antiviral, macrophage activation; MHC class I and II expression
on cells
E. Others
TGFb T- and B-cells Inhibit T- and B-cell proliferation and hematopoiesis; promote
wound healing
LIF T-cells Proliferation of stem cells; eosinophil chemotaxis

Interleukin-5 B. Colony-stimulating Factors (CSF)


IL-5 causes proliferation of activated B-cells. It also
These cytokines stimulate the growth and
induces maturation of eosinophils. Formerly, it was
differentiation of pluripotent stem cells in the bone
known as the B-cell growth factor-II.
marrow. They have been named after the types of
Interleukin-6 cell colonies they induce in soft agar culture-for
IL-6 is produced by stimulated T- and B-cells, example, granulocyte (G), or mononuclear (M) CSF
macrophages and fibroblasts. It induces (Table 18.1). IL3 which induces growth of all types
immunoglobulin synthesis by activated B cells of hematopoietic cells is known as multi-CSF. They
and formation of IL2 receptors on T cells. are responsible for adjusting the rate of production
Chapter 18: Immune Response  | 133
of blood cells according to requirements, for hypersensitivity (for example, the tuberculin
example, the massive granulocyte response seen test).
in pyogenic infections. 2. Lymphocytes transformation test: Transfor­
mation of cultured sensitized T-lymphocytes
C. Tumor Necrosis Factors (TNF) on contact with the antigen.
The tumor necrosis factor occurs as two types: 3. Target cell destruction: Killing of cultured cells
alpha and beta. by T-lymphocytes sensitized against them.
TNF-a: A serum factor found to induce hemorr­ 4. Migration-inhibiting factor (MIF) test: It is
hagic necrosis in certain tumors was named the commonly employed. As originally described,
tumour necrosis factor. It is formed principally this consisted of incubating in a culture
by activated macrophages and monocytes. It chamber, packed peritoneal macrophages in
resembles IL-l in possessing a very wide spectrum a capillary tube. The macrophages migrate
of biological activities, such as participation in the to form a lacy, fan-like pattern. If the macro­
manifestations of endotoxic and other cytokines. phages are from a guinea pig sensitized to
TNF-b: TNF-b is produced principally by T-helper tuberculoprotein, addition of tuberculin to the
cells and formerly known as lymphotoxin. Its culture chamber will inhibit the migration.
effects are similar to those of TNF-a.
Transfer Factor
D. Interferons (lFNs) Lawrence (1954) reported transfer of CMI in man
Interferons (IFNs) are a group of related low by injection of extract from the leucocytes from
molecular weight, regulatory cytokines produced immunized individual. This extract is known as the
by certain eucaryotic cells in response to a viral ‘transfer factor’ (TF). The transferred immunity is
infection. specific in that CMI can be transferred only to those
antigens to which the donor is sensitive.
There are three classes of IFNs, alpha produced
by leukocytes, beta produced by fibroblasts Properties of transfer factor: TF is a dialyzable,
and gamma by T cells activated by antigens, low molecular weight substance (MW 2000 to
mitogens or exposure to IL-2. IFN-g causes many 4000), resistant to trypsin, DNAase, RNAase and
immunological effects, such as macrophage freeze thawing. It is stable for several years at 20°C
activation, augmentation of neutrophil and and in the lyophilized form at 4°C. It is inactivated at
monocyte functions, and antitumour activity. 56°C in 30 minutes. It is not antigenic. Chemically,
it appears to be a polypeptide-polynucleotide.
E. Other Cytokines TF is highly potent. The transferred CMI
Transforming growth factor beta (TGF b): Besides is systemic and not local at the injected site
acting as a growth factor for fibroblasts and alone. Delayed hypersensitivity and various in
promoting wound healing, it also acts as a down vitro correlates of CMI can be demonstrated in
regulator of some immunological and hemato­ the recipient following TF injection. Humoral
logical processes. immunity is not transmitted by TF; TF transfers
CMI to all the antigens to which the donor is
Leukemia inhibitory factor (LIF): The leukemia
sensitive, en bloc. It is possible to transfer CMI from
inhibitory factor (LIF), produced by T cells, helps
the recipient to another in a serial fashion.
stem cell proliferation and eosinophil chemotaxis.
Mode of action of TF: The mode of action of TF is
Cytokine production is regulated by exogenous
not known. It appears to stimulate the release of
stimuli such as antigens and mitogens, as well as
lymphokines from sensi­tized T-lymphocytes.
by endogenous factors, such as neuroendocrine
hormonal peptides (corticosteroids, endorphins) Applications of Transfer Factor
and products of lipoxygenase and cyclooxygenase
pathways. They also regulate each other by positive 1. It has been used to restore immune capacity in
and negative feedbacks. patients with T-cell deficiency (Wiskott–Aldrich
syndrome).
A number of cytokines (for example, IL-l, 2,
3, colony stimulating factors, interferons) have 2. It has also been used in the treatment of
already found therapeutic application. disseminated infections associated with deficient
CMI (lepromatous leprosy, tuberculosis,
Detection of CMI mucocutaneous candidiasis).
Development of CMI can be detected by following 3. It has been employed in the treatment of
methods: malignant melanoma.
1. Skin test for DH: The original method for 4. Its use has been suggested in some autoimmune
detecting CMI was the skin test for delayed diseases (systemic lupus erythematosus,

www.ebook3000.com
134  |  Section 2: Immunology
rheumatoid arthritis) and diseases of unknown 2. Dose of Antigen
etiology (sarcoidosis, multiple sclerosis). The induction of tolerance is dose-dependent.
With certain antigens, tolerance can be induced by
Immunological tolerance two types of doses, one high and the other low, with
intermediate doses producing immunity instead of
Immunologic tolerance is defined as the absence tolerance. These are known as ‘high zone’ and ‘low
of a specific immune response resulting from a zone’ tolerance respectively. A special type of high
previous expo­sure to the inducing antigen. The zone tolerance is Felton’s immunological paralysis.
most notable example is immunologic tolerance
to self. Any antigen that comes into contact with 3. Route of Tolerogen Administra­tion
the immunological system during embryonic life
would be recognized as a self-antigen and would Certain haptens that are immunogenic in guinea
not induce any immune response. pigs by the intradermal route are tolerogenic orally
Burnet and Fenner (1949) suggested that or intravenously.
the unresponsiveness of individuals to self-
antigens was due to the contact of the immature 4. Nature of Tolerogen
immunological system with self-antigens during Soluble antigens and haptens are more tolerogenic
embryonic life. Any antigen that comes into contact than particulate antigens.
with the immunological system during embryonic
life would be recognized as a self-antigen and would 5. Various Treat­ments
not induce any immune response. Medawar and
his colleagues (1953) proved this experimentally When human gammaglobulin is heat aggregated,
using two strains of syngenetic mice. When a it is highly immunogenic in mice but is tolerogenic
skin graft from one inbred strain of mice (CBA) when deaggregated.
is applied on a mouse of another strain (A), it
is rejected. If CBA cells are injected into fetal or 6. Genetic Background
newborn strain A mice, however, the latter when Rabbits and mice can be rendered tolerant more
they grow up will freely accept skin grafts from rapidly than guinea pigs and chickens.
CBA mice. The content of the self antigen appears
to have been enlarged by contact with a foreign Mechanism of Tolerance
antigen during embryonic life. This phenomenon
is called ‘specific immunological tolerance’. Tolerance can arise through three possible
mechanisms:
1. Clonal deletion
Types of Immunological Tolerance
2. Clonal anergy
Two forms of tolerance are known: 3. Suppression.
1. Natural tolerance: The body develops immune
tolerance towards self-molecules due to natural 1. Clonal Deletion
tolerance during growth of fetus. Autoimmune
disease wi11 develop in the event of breakdown In embryonic life clones of B- and T-cells
of tolerance to self-tissues. possessing receptors that recog­nize self-antigens
2. Acquired tolerance: Tolerance in later life may are selectively deleted or eliminated and, therefore,
occur under certain special circum­stances, e.g. no longer available to respond upon subsequent
ingestion of bovine myelin in high concentrations exposure to that antigen. This is known as clonal
can tolerise an individu­al to myelin. deletion.

2. Clonal Anergy
Parameters that Affect the Induction of
Tolerance Clones of B- and T-cells exp­ressing receptors that
recognize self-antigen might remain but they
These in­clude age, dose of antigen, route of tolerogen cannot be activated. This is known as clonal anergy.
administra­tion, physical nature of the antigen, and
various treat­ments that reduce activation of positive 3. Suppression
regulatory cells of the immune response.
Clones of B- and T-cells exp­ressing receptors that
recognize self-antigens are preserved. Antigen
1. Age
recognition might be capable of causing activation,
The younger the recipient, the easier it is to induce however, exp­ression of immune response might be
toler­ance, and, of course, it is easiest in utero. inhibi­ted or blocked through active suppression.
Chapter 18: Immune Response  | 135
Theories of Antibody formation development in later life by somatic mutation
could lead to autoimmune processes. Each
Theories of immunity fall into two categories: immunocompetent cell was capable of reacting
instructive and selective. with one antigen (or a small number of
antigens) which could recognize and combine
A. Instructive Theories with antigens introduced into the body. The
1. Direct template theories: According to these, result of the contact with the specific antigen
the antigen or the antigenic determinant enters was cellular proliferation to form clones
the antibody-forming cell and serves as a synthesizing the antibody. This theory is more
template against which antibody mole­cules are widely accepted than other theories.
synthesized so that they have combining sites
Key Points
complementary to the antigenic determinants.
These are therefore known as ‘direct template’ ™™ The immune response is the specific reactivity
theories. induced in a host by an anti­genic stimulus and can
be divided into two types—the humoral (antibody-
2. Indirect template theory: This theory was mediated) and the cellular (cell-mediated) types
proposed by Burnet and Fenner (1949). ™™ The production of antibodies consists of three
According to this theory, the entry of the steps: 1. Lag phase; 2. Log phase; 3. A plateau or
antigenic determinant into the antibody steady state
producing cell induced in it a heritable change. ™™ The hu­moral response results from activation of
naive lymphocytes (primary response) or memory
A ‘genocopy’ of the antigenic determinant
lymphocytes (secondary re­sponse)
was thus incorporated in its genome and ™™ Monoclonal antibodies: Such antibodies produced by
transmitted to the progeny cells (indirect a single clone and directed against a single antigenic
template). determinant are called monoclonal antibodies, e.g.
plasma cell tumor (myeloma)
™™ Cell-mediated immunity’ (CMI): The term ‘cell-
B. Selective Theories mediated immunity’ (CMI) refers to the specific
1. Side chain theory: According to side chain immune responses which involves T-Iymphocyte-
theory, immunocompetent cells (ICCs) have mediated functions
™™ Cytokines: Cytokines are biologically active
surface receptors capable of reacting with
substances produced by cells that influence other
antigens which have complementary side cells. Interferons, growth factors and others were
chains. When foreign antigens are introduced found to have similar effects. Therefore, all of them
into the body, they combine with those cell have been grouped under the term ‘cytokines’
receptors which have a complementary ™™ Transfer factor: Transfer of CMI in man by injection of
fit. This inactivates the receptors. There extract from the leukocytes from immunized individual
™™ Immunologic tolerance is defined as the absence of a
is an overproduction of the same type of specific immune response resulting from a previous
receptors which circulate as antibodies as a expo­sure to the inducing antigen. Two forms of
compensatory mechanism. tolerance are known. 1. Natural; 2. Acquired
2. Natural selection theory: This theory was ™™ Tolerance can arise through three possible
mechanisms: 1. Clonal deletion; 2. Clonal anergy;
proposed by Jerne (1955) which postulates that
3. Suppression
about a million globulin (antibody) molecules ™™ Theories of immunity fall into two categories:
were formed in embryonic life, which covered instructive and selective:
the full range of antigenic specificities. These A. Instructive theories: 1. Direct template theories;
globulins were the ‘natural antibodies’. When 2. Indirect template theory
an antigen was introduced, it combined B. Selective theories: 1. Side chain theory; 2. Natural
selection theory; 3. Clonal selection theory.
selectively with the globulin that had the
nearest complementary ‘fit’. The globulin,
with the combined antigen, homed in on the Important questions
antibody-forming cells and stimulated them 1. Discuss the primary and secondary humoral
to synthesize the same kind of antibody. immune responses.
3. Clonal selection theory: This theory was 2. Discuss briefly about:
proposed by Burnet (1957). This theory states Monoclonal antibodies—production and
that during immunological development, cells applications.
Adjuvants
capable of reacting with different antigens
Cytokines
were formed by a process of somatic mutation.
Theories of antibody production
Clones of cells that had immunological 3. Write short notes on:
reactivity with self-antigens were eliminated a. Transfer factor
during embryonic life. Such clones are b. Burnet’s clonal selection theory
called forbidden clones. Their persistence or c. Immunological tolerance

www.ebook3000.com
136  |  Section 2: Immunology
Multiple choice questions (MCQs) 4. Development of cell mediated immunity can be
detected by:
1. The synthesis and production of antibodies a. Skin test for delayed hypersensitivity
typically is dependent on complex interaction of b. Lymphocyte transformation test
all the following cells except: c. Migration inhibiting factor test
a. Macrophages d. All the above
b. Helper T cells 5. Transfer factor shows all the following features
c. Cytotoxic T cells except:
d. B cells a. It is an extract from the leukocytes from the
2. Which animal is used for for monoclonal immnized host
antibodies production: b. Transfers humoral immunity
a. Guinea pig c. Transfers CMI
b. Mouse d. Transferred immunity is systemic
c. Rabbit 6. Clonal selection theory was postulated by:
d. None of the above a. Breinl and Haurowitz
3. Interleukin-l is a protein produced mainly by: b. Burnet and Fenner
c. Ehrlich
a. Macrophages and monocytes
d. Burnet
b. Polymorphonuclear leukocytes
c. Lymphocytes Answers (MCQs)
d. Stem cells 1. c; 2. b; 3. a; 4. d; 5. b; 6. d
19
Chapter

Immunodeficiency Diseases

Learning Objectives

After reading and studying this chapter, you should ∙∙ List primary and secondary immunodeficiency
be able to: syn­dromes.
∙∙ Classify and enumerate immunodeficiency diseases

Introduction This rare disorder, which appears to selectively


involve very early B-lineage cells, was the first
Immunodeficiency diseases are conditions where
recognized of all of the primary immunodeficiencies,
the defence mechanisms of the body are impaired,
and was discovered in 1952 by Colonel Ogden
leading to repeated microbial infections of varying
Bruton. It is seen only in male infants.
severity and sometimes enhanced susceptibility to
malignancies. Manifestations: The disease presents as recurrent
Immunodeficiency disease results from the serious infections with pyogenic bacteria,
absence or failure of normal function, of one or particularly with pneumococci, streptococci,
more elements of the immune system. Specific meningococci, Pseudomonas and H. influenzae.
immunodeficiency diseases involve abnormalities Patients respond normally to viral infections such
of T- or B-cells of the adaptive immune system. as measles and chickenpox.
Nonspecific immunodeficiency diseases involve All classes of immunoglobulins are grossly
abnormalities of elements, such as complement or depleted in the serum. Tonsils and adenoids are
phagocytes, which act nonspecifically in immunity. atrophic. Lymph node biopsy reveals a depletion
of cells of the bursa-dependent areas. Plasma
Classification of cells and germinal centers are absent even after
Immunodeficiency Diseases antigenic stimulation. There is a marked decrease
in the proportion of B-cells in circulation.
Immunodeficiencies may be classified as primary
or secondary. Cell-mediated immunity: Cell-mediated
immunity is not affected. Delayed hypersensitivity
Primary Immunodeficiencies of tuberculin and contact dermatitis types can be
The established types of primary immunodeficiency demonstrated. Allograft rejection is normal.
syndromes are listed in Table 19.1. The lymphoid Management
cell disorders may affect T-cells, B-cells, or both B- Its management consists of the maintenance of an
and T-cells. Deficiencies involving components of adequate level of immunoglobulins.
nonspecific mediators of innate immunity, such as
phagocytes or complement, are impaired. b. Transient Hypogammaglobulinemia of
Infancy
A. Disorders of Specific Immunity This is due to an abnormal delay in the initiation of
I. Humoral Immunodeficiencies (B-cell IgG synthesis in some patients. By three months of
age, normal infants begin to synthesize their own
Defects)
IgG. When there is a delay, immunodeficiency
a. X-linked Agammaglobulinemia (XLA) occurs. Recurrent otitis media and respiratory
The model B-cell deficiency is X-linked agamma­ infections are the common diseases found in these
globulinemia or Bruton’s hypogammaglobulinemia. conditions. It may be found in infants of both sexes.

www.ebook3000.com
138  |  Section 2: Immunology
Table 19.1  Classification of primary immuno­ proper signals from the T-cells. B-cells fail to mature
deficiency syndromes into plasma cells and secrete immunoglobulins.

A. Disorders of specific immunity


Treatment: Patients with CVID should be treated
I. Humoral immunodeficiencies (B-cell defects) with intravenous gamma globulin.
a. X-linked agammaglobulinemia
b. Transient hypogammaglobulinemia of infancy d. Selective Immunoglobulin Deficiencies
c. Common variable immunodeficiency (late onset
hypogammaglobulinemia) In these conditions, there is selective deficiency
d. Selective immunoglobulin deficiencies (lgA, IgM or of one or more immunoglobulin classes, while the
IgG subclasses) others remain normal or elevated.
e. Immunodeficiencies with hyper-IgM i. Selective IgA deficiency: Recurrent respiratory
f. Transcobalamin II deficiency
II. Cellular immunodeficiencies (T-cell defects) and genitourinary tract infections resulting
a. Thymic hypoplasia (Digeorge’s syndrome) from lack of secreted IgA on mucosal surfaces
b. Chronic mucocutaneous candidiasis are common. In addition, problems such as
c. Purine nucleoside phosphorylase (PNP) deficiency. intestinal malabsorption, allergic disease, and
III. Combined immunodeficiencies (B- and T-cell defects) autoimmune disorders may also be associated
a. Cellular immunodeficiency with abnormal
immunoglobulin synthesis (Nezelof syndrome) with low IgA levels.
b. Ataxia telangiectasia ii. Selective IgM deficiency: Selective IgM
c. Wiskott-Aldrich syndrome deficiency is a rare condition and has been
d. Immunodeficiency with thymoma found to be associated with septicemia.
e. Immunodeficiency with short-limbed dwarfism
f. Episodic lymphopenia with lymphocytotoxin
g. Severe combined immunodeficiencies e. X-linked Hyper-IgM Syndrome (XHM)
1. ‘Swiss type’ agammaglobulinemia X-linked hyper-IgM syndrome (XHM) is
2. Reticular dysgenesis of de Vaal
characterized by a deficiency of IgG, IgA and IgE, and
3. Adenosine deaminase (ADA) deficiency
B. Disorders of complement . elevated levels of IgM (hyper M). XHM syndrome is
a. Complement component deficiencies generally inherited as an X-linked recessive disorder.
b. Complement inhibitor deficiencies They are susceptible to pyogenic infections and
C. Disorders of phagocytosis should be treated with intravenous gammaglobulin.
a. Chronic granulomatous disease
Children with XHM suffer recurrent infections,
b. Myeloperoxidase deficiency
c. Chediak-Higashi syndrome especially respiratory infections.
d. Leukocyte G6PD deficiency
e. Job’s syndrome f. Transcobalamin II Deficiency
f. Tuftsin deficiency
g. Lazy leukocyte syndrome In this disorder, patients show metabolic effects
h. Hyper-lgE syndrome of vitamin B12 deficiency including megaloblastic
i. Actin-binding protein deficiency anemia and intestinal villus atrophy and is
j. Shwachman’s disease inherited as autosomal recessive. The associated
immunological defects are depleted plasma cells,
Treatment: Treatment with gammaglobulin may diminished immunoglobulin levels and impaired
be required in some cases but it is not recom­ phagocytosis.
mended prophylactically, as it may contribute to Treatment with vitamin B12 has been reported
prolongation of immunodeficiency by a negative to restore hematopoietic, gastrointestinal and
feedback inhibition of IgG synthesis. B-cell functions but not phagocytic activity.

c. Common Variable Immunodeficiency (CVID) II. Cellular Immunodeficiencies (T-cell


CVID is characharteized by a profound decrease Defects)
in numbers of antibody-producing plasma cells, a. Thymic hypoplasia (DiGeorge’s syndrome):
low levels of most immunoglobulin isotypes Thymic hypoplasia results from dysmorphogenesis
(hypogammaglobulinemia), and recurrent of the third and fourth pharyngeal pouches during
infections. The condition is usually manifested early embryogenesis, leading to hypoplasia or
later in life (in the second or third decade of life) aplasia of the thymus and parathyroid glands.
than other deficiencies and is sometimes called The immunodeficiency primarily involves
late onset hypogammaglobulinemia or incorrectly, cell mediated immunity. The thymus-dependent
acquired hypogammaglobulinemia. Patients with areas of lymph nodes and spleen are depleted of
CVID like males with XLA, are very susceptible to lymphocytes. Delayed hypersensitivity and graft
pyogenic organisms and to the intestinal protozoan, rejection are depressed. Thymic transplantation is
Giardia lamblia, which cause severe diarrhea. The of some value for correcting the T-cell defects, but
B-cells are not defective; instead, they fail to receive many DiGeorge patients have such severe heart
Chapter 19: Immunodeficiency Diseases  | 139
disease that their chances of survival are poor, even thrombocytopenic purpura, and undue susceptibility
if the immune defects are corrected. to infection. Affected boys rarely survive the
first decade of life, death being due to infection,
b. Purine nucleoside phosphorylase (PNP)
hemorrhage or lymphoreticular malignancy.
deficiency: The enzyme purine nucleoside
Cell-mediated immunity undergoes progressive
phosphorylase (PNP) is involved in the sequential
deterioration associated with cellular depletion of
degradation of purines to hypoxanthine and finally
the thymus and the paracortical areas of lymph
to uric acid. The lack of enzyme PNP because of a
nodes. Serum IgM level is low but IgG and IgA
gene defect in chromosome 14 results in impaired
levels are normal or elevated. Isohemagglutinins
metabolism of cytosine and inosine to purine.
are absent in the serum. The humoral defect
These patients show decreased T-cell proliferation
appears to be a specific inability to respond to
leading to decreased T-cell-mediated immunity
polysaccharide antigens
and recurrent or chronic infections.
Bone marrow transplantation and transfer
factor therapy have been found beneficial.
III. Combined Immunodeficiencies (B-
and T-cell Deficiencies) d. Immunodeficiency with Thymoma
a. Cellular Immunodeficiency with Abnormal This syndrome, occurring usually in adults, consists
Immunoglobulin Synthesis (Nezelof of thymoma, impaired cell-mediated immunity
Syndrome) and agammaglobulinemia. This association
There is depressed cell-mediated immunity which of hypogammaglobulinemia with spindle cell
is associated with selectively elevated, decreased thymoma usually occurs relatively late in adult life.
or normal levels of immunoglobulin. Affected It is frequently accompanied by aplastic anemia.
individuals suffer from chronic diarrhea, viral and
fungal infections, and a general failure to thrive. e. Severe Combined Immunodeficiency (SCID)
Abundant plasma cells are seen in the spleen, The family of disorders termed SCID stems from the
lymph nodes, intestines and elsewhere in the body. defects in lymphoid development that affect either
Thymic dysplasia occurs with lymphoid depletion. T cells or both T and B cells. It is usually congenital,
Autoimmune processes, such as hemolytic anemia may be inherited either as an X-linked or autosomal
are common. recessive defect, or may occur sporadically.
i. Swiss type agamma­globulinemia
Treatment
Histocompatible bone marrow transplantation, Agammaglobulinemia with lymphocytopenia
transfer factor and thymus transplantation have a n d s e v e re d e f e c t i n c e l l - m e d i at e d
been used for treatment. Adequate antimicrobial immunity was reported by Swiss workers
therapy is essential for the treatment of microbial in 1958. Referred to as Swiss-type agamma­
infection. globulinemia. The basic defect is presumed
to be at the level of the lymphoid stem cell.
ii. Adenosine deaminase (ADA) deficiency
b. Ataxia Telangiectasia
Adenosine deaminase (ADA) deficiency
Ataxia telangiectasia is a disease syndrome that is the first immunodeficiency disease
includes deficiency of IgA and sometimes of associated with an enzyme deficiency. ADA
IgE. It is inherited as an autosomal-recessive catalyzes the conversion of adenosine to
trait. The most prominent clinical features are inosine, an important step in the purine
progressive cerebellar ataxia, oculocutaneous metabolic pathway. Its deficiency results in
telangiectasias, chronic sinopulmonary disease, accumulation of adenosine, which interferes
a high incidence of malignancy, and a variable with purine metabolism and DNA synthesis.
humoral and cellular immunodeficiency. Death The range of immunodeficiency varies from
occurs due to sinopulmonary infection early in complete absence to mild abnormalities
life, or malignancy in the second or third decade. of B- and T-cell functions. The condition is
The defective cell-mediated immunity results associated with chondrocyte abnormalities
in an impairment of delayed hypersensitivity and which can be discerned radiologically.
graft rejection. Transfer factor therapy and fetal iii. Reticular dysgenesis: This is the most serious
thymus transplants have been tried with some form of SCID. Here the defect is in the
benefit. development of the multipotent bone marrow
stem cell, as a result of which there is a total
c. Wiskott–Aldrich Syndrome (WAS) failure of myelopoiesis leading to lymphopenia,
This is an X-linked recessive syndrome that is neutropenia, thrombocytopenia, anemia and
characterized clinically by the triad of eczema, bone marrow aplasia. A baby with this disorder

www.ebook3000.com
140  |  Section 2: Immunology
usually dies within the first year of life from Bacterial infection: The bacteria involved in
recurrent, intractable infections. the recurrent infections are catalase-positive
iv. Recombinase activating gene (RAG 1/2) pyogenic pathogens such as staphylococci
deficiency. and coliforms. Catalase-negative patho­
v. Interleukin receptor γ chain (γc) deficiency gens such as streptococci and pneumococci
vi. Janus-associated kinase 3 (JAK3) deficiency are han­dled normally. Leukocytes from the
patients are unable to kill catalase positive
B. Disorders of complement bacteria following phagocytosis. The bacteria
multiply in the cells and, being protected from
a. Complement Component Deficiencies antibodies and antibiotics by their intracellular
Genetic defi­ciencies have been detected for almost position, set up chronic suppurative infection.
all the comple­ment components in human beings. Bactericidal defect: The diminished bactericidal
The defects are transmitted as autosomal recessive capacity of the phagocytic cells is associated
traits. Hemolytic and other functional activities with a decrease of some metabolic processes like
are completely restored by supplying the deficient oxygen consumption, hexose monophosphate
factor. Deficiency of C1 and C4 is associated pathway activity and production of hydrogen
with sys­temic lupus erythematosus. Recurrent peroxide. The diminished H2O2 production
pyogenic infections were found associated with C3 appears to be the major reason for the bacteri­
deficiency and neisserial infections with deficiency cidal defect.
of C6, C7 and C8. So far, there is no known disease Leukocytes from the patients fail to reduce nitro­
with deficiency of C9. blue tetrazolium (NBT) during phagocytosis.
This property has been used as a screening
b. Complement Inhibitor Deficiencies method (NBT test) for the diagnosis of chronic
i. C1 inhibitor: Hereditary angioneurotic edema granulomatous disease.
(HAE) is due to a genetic deficiency of C1 b. Myeloperoxidase (MPO) deficiency: In this
inhibitor and is transmit­ted as an autosomal- rare disease, leukocytes are deficient in
dominant condition. It manifests clinically as myeloperoxidase (MPO). Patients are liable to
localized edema of the tissue, often following develop recurrent Candida albicans infec­tion.
trauma, but sometimes with no known cause. c. Chediak-Higashi syndrome: The leukocytes
Management: Androgens, aminocap­roic acid possess diminished phagocytic activity. The
and its analog tranexamic acid have been found individuals with this syn­drome suffer from
useful in the management of this condition. Plasma recurrent infections similar to those seen in
infusions, once recommended for treatment, have persons with CGD
been given up as they were found to worsen the d. Leukocyte G-6-PD deficiency: Leucocytes are
condition in some cases. deficient in glu­cose-6-phosphate dehydrogenase.
ii. Deficiency of C3b inactivator: The rare defici­ These patients show diminished bactericidal
ency of C3b inactivator has been associated activity after phago­cytosis leading to repeated
with chronic recurrent pyogenic lesions. bacterial infections.
e. Job’s syndrome: It is probably a primary defect
in phagocytic function.
C. Disorders of Phagocyte
f. Tuftsin deficiency: A leucokinin capable of
Phagocytosis may be impaired by either intrinsic or stimu­lating phagocytosis, discovered at Tufts
extrinsic defects. Intrinsic disorders may be due to University, Boston, has been designated ‘tuftsin’.
de­fects within the phagocytic cell, such as enzyme Chemically, it is a small tetrapeptide (Thr-Lys-
defi­ciencies. Extrinsic disorders may be due to a Pro-Arg) and patients have been reported to be
deficiency of opsonic antibody, complement or prone to local and systemic bacterial infections.
other factors promoting phagocytosis. Phagocytic g. Lazy leukocyte syndrome: The basic defect here
dysfunction leads to increased susceptibility is in chemotaxis and neutrophil mobility.
to infec­tion, ranging from mild recurrent skin h. Hyper-IgE syndrome: Serum IgE levels are
infections to overwhelming systemic infection. usually more than ten times the normal level.
a. Chronic granulomatous disease (CGD): Chronic i. Actin-binding protein deficiency.
granulomatous disease (CGD) is a group of j. Shwachman’s disease.
disorders, most of which are X-linked recessive, k. Leucocyte adhesion deficiency (LAD).
with some that are autosomal recessive.
Individuals with CGD possess polymorpho­
Secondary immunodeficiencies
nuclear leucocytes that phagocytize invading
bacteria normally but are un­able to kill many Secondary or acquired deficiencies of immuno­
of the ingested microorganisms. logical mecha­n isms can occur secondarily to
Chapter 19: Immunodeficiency Diseases  | 141
a number of disease states, such as metabolic Important questions
disorders, malnutrition, malignancy and infections
1. What are immunodeficiency diseases? Classify
or after exposure to drugs and chemicals. AIDS
and enumerate immunodeficiency diseases.
is a secondary immunodeficiency secondary
2. Write short notes on:
immunodeficiency is far more common than
a. DiGeorge’s syndrome
primary immunodefi­ciency.
b. B-cell defect
c. T-cell defect
Deficiencies of Humoral and Cellular d. Disorders of complement
Immune Response
Deficiencies of humoral and cellular immune Multiple choice questions (MCQs)
response may occur secondarily during the course
of many disease processes. 1. Recurrent infections with certain viruses, protozoa,
and fungi indicate a:
A Humoral immunity depression: Humoral a. T-cell deficiency
deficiency results when B cells are depleted as b. B-cell deficiency
in lymphoid malignancy, particularly in chronic c. Combined T- and B-cell deficiency
lymphatic leukemia; when immunoglobulin d. Complement deficiency
catabolism is increased as in the nephrotic 2. X-linked agammaglobulinemia is:
syndrome; when excessive loss of serum protein a. Prototype of ‘pure’ T-cell deficiency
occurs as in exfoliative skin disease and in b. Prototype of ‘pure’ B-cell deficiency
protein-losing enteropathies; and when excessive c. Prototype of ‘pure’ combined T- and B-cell
production of abnormal immunoglobulins occurs deficiency
as in multiple myeloma. d. Prototype of ‘pure’ complement deficiency
3. All the following are the examples of T-cell
B. Cell-mediated immunity depression: Cell- deficiencies except:
mediated immunity is depressed in lymphoreticular a. DiGeorge’s syndrome
malignancies like Hodgkin’s lymphoma, obstruction b. Chronic mucocutaneous candidiasis
to lymph circulation and infiltration of the thymus- c. Chronic granulomatous disease
dependent area of lymph nodes with non lymphoid d. Purine nucleoside phosphorylase deficiency
cells as in leplromatous leprosy; and, transiently, 4. Which of the following defects occur in Nezelof’s
following certain viral infections, such as measles. syndrome?
a. T cell defects b. B cell defects
C. Humoral and Cell-mediated immunity
c. Both of the above d. None of the
depression: Both types of immune responses
5. The most common severe form of severe combined
are adversely affected in nutritional deprivation.
immunodeficiency is:
Aging also causes waning in the efficiency of
a. Patients with a defect in adenosine deaminase
acquired immunity. Immunodeficiency follows enzyme
the intentional or unintentional administration of b.  Patients with a defect in purine nucleoside
immunosuppressive agents. phosphorylase enzyme
Key Points c. Chediak-Higashi syndrome
d. Swiss type agammaglobulinemia
™™ Immunodeficiency disorders can occur in T-cells,
6. All are disorders of phagocytosis except:
B-cells, comple­ment and phagocytes
™™ These can be classified as primary or secondary
a. Chronic granulomatous disease
immunodefi­ciencies b. Myeloperxidase deficiency
™™ A primary immunodeficiency may affect either c. Chediak-Higashi syndrome
adaptive or innate immune functions d. Digeorge’s syndrome
™™ Secondary immunodeficiencies occur secondary to
numerous diseases or conditions. Answers (MCQs)
1. a; 2. b; 3. c; 4. c; 5. c; 6. d

www.ebook3000.com
20
Chapter

Hypersensitivity Reactions

Learning Objectives

After reading and studying this chapter, you should ∙∙ Discuss type I, type II, type III, type IV hypersensitivity
be able to: reactions: Mechanism and examples.
∙∙ Compare major types of hypersensitivity reactions.
∙∙ Differentiate between immediate and delayed
hypersensitivity.

Hypersensitivity: It is an exaggerated immune Classification of hypersensitivity


response that results in tissue damage and is reactions
manifested in the individual on second or subsequent
contact with an antigen. Hypersensitivity reactions have been classified
Immune responses to foreign antigens are, for the traditionally into ‘immediate’ and ‘delayed’ types,
most part, beneficial to the responding individual. based on the time required for a sensitized host to
Nevertheless, at times, the response to a seemingly develop clinical reactions on re-exposure to the
innocuous antigen can result in tissue damage and antigen.
even death. This inappropriate immune response is The major differences between the immediate
termed as hypersensitivity or allergy. and delayed types of hypersensitivity reactions are
shown in Table 20.1.
Allergy: The term ‘allergy’ was coined by von
Pirquet merely to indicate an altered reactivity on
Gell and Coomb Classification
second contact with an antigen. In time, however,
the term allergy has become synonymous with Peter Gell and Robert Coombs developed a
hypersensitivity. Increased resistance, called classification system for reactions responsible
immunity, and increased susceptibility, called for hypersensitivities in 1963. The Gell–Coombs
hypersensitivity, were regarded as opposite forms classification system divides hypersensitivity into
of allergy. four types: I, II, III, and IV.

Table 20.1  Distinguishing features of immediate and delayed types of hypersensitivity


Characteristic Immediate hypersensitivity Delayed hypersensitivity
1. Time of reaction after 1.  Reaction appears and recedes rapidly 1.  Appears slowly, lasts longer
challenge with antigen
2. Induction 2.  Induced by antigens or haptens 2. Antigen or hapten intradermally or by
any route
3.  Immune response 3. Circulating antibodies present and 3. Circulating antibodies may be absent
responsible for reaction; ‘antibody and are not responsible for reaction;
mediated’ reaction ‘cell-mediated’ reaction
4.  Transfer of hypersensitivity 4.  Passive transfer possible with serum 4. Cannnot be transferred with serum; but
possible with T-cells or transfer factor
5. Desensitization 5.  Desensitization easy, but shortlived 5.  Difficult, but long-lasting
Chapter 20: Hypersensitivity Reactions  | 143
Type I (Anaphylactic, IgE or minutes after a person sensitized to an antigen is
Reagin-dependent) re-exposed to that antigen.
Type I or immediate hypersensitivity is character­
Anaphylaxis
ized by the production of antibodies (‘cytotropic’
IgE antibodies) which bind specifically to a high- Anaphylaxis means “the opposite of protected”,
affinity receptor on mast cells and basophils in from the prefix ana-, against, and the Greek
sensitized individuals. Subsequent exposure to phylaxis, protection. Anaphylaxis is an inclusive
the same antigen will combine with the cell­fixed term for the reactions caused when certain antigens
antibody, leading to release of pharmacologically combine with IgE antibodies. Anaphylactic
active substances (vasoactive amines) which responses can be:
produce the clinical reaction. A. Systemic reactions.
B. Localized reactions: Localized reactions.
Type II (Cytotoxic or Cell-stimulating)
A. Systemic Anaphylaxis (or Anaphylactic
Type II reaction is initiated by IgG (or rarely IgM)
antibodies that react either with cell surface or Shock)
tissue antigens. Cell or tissue damage occurs in Systemic anaphylaxis is a generalized response that
the presence of complement or mononuclear occurs when an individual sensitized to an allergen
cells. Combination with antibody may, in some receives a subsequent exposure to it. Antigen enters
in­stances, cause stimulation instead of damage. the bloodstream and becomes widespread.
An example is the ‘long acting thyroid stimulator’ Systemic anaphylaxis is a shock-like and often
(LATS), an antibody against some determinant on fatal state whose onset occurs within minutes
thyroid cells, which stimulates excessive secretion of a type I hypersensitive reaction. This was the
of thyroid hormone. response observed by Portier and Richet in dogs
after antigenic challenge.
Type III (Immune Complex or
Toxic Complex Disease) Sensitizing Dose and Shocking Dose
Here the damage is caused by antigen antibody Sensitization is most effective when the antigen
complexes. is introduced parenterally but may occur by
any route, including ingestion or inhalation.
Type IV (Delayed or Cell-mediated In susceptible species, very minute doses can
sensitize the host. There should be an interval of at
Hypersensitivity)
least 2-3 weeks between the sensitizing dose and
Type IV or cell-mediated reactions are those in the shocking dose.
which specific T-cells are the primary effector cells.
The antigen activates specifically sensitized T-cells, Target Tissues or ‘Shock Organs’
leading to the secretion of lymphokines, with fluid
and phago­cyte accumulation. The classification Tissues or organs predominantly involved in the
and some of the features of hypersensitivity anaphylactic reaction are known as ‘target tissues’
reactions are shown in Table 20.2. or ‘shock organs’. Other changes seen in anaphylaxis
are edema, decreased coagulability of blood, fall in
blood pressure and temperature, leukopenia and
Type V: Hypersensitivity (Stimulatory Type)
thrombocytopenia.
Jones-Mote Reaction (or)
Cutaneous Basophil Hypersensitivity Experimental animals: Systemic anaphy­laxis can
This is an antibody-mediated hypersensitivity be induced in a variety of experimental animals
and is a modification of type II hypersensitivity and is seen occasionally in humans. Guinea
reaction. Antibodies interact with antigens on pigs are highly susceptible and rats are very
cell surface which leads to cell proliferation and resistant. Rabbits, dogs and human beings are of
differentiation instead of inhibition or killing. intermediate susceptibility.
Antigen-antibody reaction enhances the activity
of affected cell. Systemic Anaphylaxis in Humans
Systemic anaphylaxis in humans is characterized
Type I Hypersensitivity (IgE by a sim­ilar sequence of events that occurs when
an individual sensitized to an allergen receives a
dependent)
subsequent exposure to it. In human beings, fatal
A type I hypersensitive reaction is induced by certain anaphylaxis is fortunately rare. The reaction is
types of antigens referred to as allergens, type I, or immediate due to the large amount of mast cell
anaphylactic, reactions often occur within 2 to 30 mediators released over a short period.

www.ebook3000.com
144  |  Section 2: Immunology
Because of the reac­tions the individual can die with an antigen, the individual produces IgE
within a few minutes from reduced venous return, antibodies. IgE are bound to surface receptors on
asphyxiation, reduced blood pressure, and circula­ mast cells and basophils. IgE molecules attach
tory shock. to these receptors by their Fc end, leaving two
Antigens: A wide range of antigens have been antigen-binding sites free. Mast cells and basophils
shown to trigger this reaction in susceptible coated by IgE are said to be sensitized, making the
humans, includ­ing the venom from bee, wasp, individual allergic to the allergen.
hornet, and ant stings; drugs, such as penicillin, Following exposure to the shocking dose,
insulin, and antitoxins; and seafood and nuts. If the antigen molecules combine with the cell
not treated quickly, these reactions can be fatal. bound IgE, bridging the gap between adjacent
antibody molecules. This cross-linking increases
Treatment: Epinephrine (adrenaline) is the drug
the permeability of the cells to calcium ions and
of choice for systemic anaphylactic reac­tions, a
leads to degranulation, with release of biologically
drug that constricts blood vessels and raises the
active substances contained in the granules. The
blood pressure.
pharmacologically active mediators released
from the granules act on the surrounding tissues.
i. Cutaneous Anaphylaxis The manifestations of anaphylaxis are due
Small amounts of potential allergens are introduced to pharmacological mediators, which can be
at specific skin sites either by intradermal injec­tion classified as two types, either primary or sec­
or by superficial scratching. If a person is allergic ondary. These mediators trigger smooth muscle
to the allergen, there is a wheal and flare (local contractions, vasodilation, increased vascular
anaphylaxis) within 30 minutes. permeability, and mucous secretion (Fig. 20.1)
Use: Cutaneous anaphylaxis is useful in testing A.Primary mediators of anaphylaxis
for hypersensitivity and in identifying the allergen
responsible in atopic diseases. A. Primary Mediators of Anaphylaxis
These are produced before degranulation and are
ii. Passive Cutaneous Anaphylaxis (PCA) stored in the granules. The most significant primary
This test is an extremely sensitive in vivo method mediators are histamine, proteases, eosinophil
for detection of antibodies de­veloped by Zoltan chemotactic factor, neutrophil chemotactic fac­tor,
Ovary (1952). In this pro­cedure, a small vol­ume of and heparin.
the antibody is injected intradermally into a normal 1. Histamine: Histamine, which is formed by
animal. The corresponding antigen is injected decarboxylation of the amino acid histidine
intravenously 4–24 hours afterwards along with and is localized in the granules of mast cells
a dye, such as Evans blue that is strongly bound and basophils and in the platelets of some
to serum albumin. There will be an immediate species. Histamine induces smooth muscle
blueing at the site of intradermal injection due to contraction in diverse tissues and organs,
vasodilatation and in­creased capillary permeability including vasculature, intestines, uterus and
(wheal-and-flare reac­tion). especially the bronchioles. It also stimulates
secretions (secretagogue effect).
Use: PCA can be used to detect human IgG 2. Heparin: It contributes to anaphylaxis in dogs,
antibody which is heterocytotropic (capable of but apparently not in human beings.
fixing to cells of other species) but not IgE which 3. Serotonin (5-hydroxytryptamine): It is found
is homocytotropic (capable of fixing to cells of in the intestinal mucosa, brain tissue and
homologous species only). platelets. It induces contraction of smooth

iii. Anaphylaxis in Vitro


(Schultz–Dale Phenomenon)
Schultz and Dale (1910) demonstrated that isolated
tissues, such as intes­t inal or uterine muscle
strips from sensitised guinea pigs, held in a bath
of Ringer’s solution will contract vigorously on
addition of the specific antigen to the bath. This is
known as the Schultz–Dale phenomenon.

Mechanism of Anaphylaxis
The immunologic basis for hypersensitivity is
cytotropic IgE antibody. After an initial contact Fig. 20.1: Antigen-induced mediator release from mast cell
Chapter 20: Hypersensitivity Reactions  | 145
muscle, increased vascular permeability, and Anaphylactoid Reaction
capillary dilatation. Intravenous injection of peptone, trypsin and
4. Chemotactic Factors certain other substances provokes a clinical reaction
i. Eosinophil chemotactic factor (ECF- resembling anaphylactic shock. This is termed
A): Eosinophil chemotactic factors of ‘anaphylactoid reaction’. The clinical resemblance
anaphylaxis (ECF-A) are acidic tetrapeptides is due to the same chemical me­diators participating
released from mast cell granules which are in both reactions.
strongly chemotactic for eosinophils. These Anaphylactoid shock has no immunological
probably contribute to the eosinophilia basis and is a nonspecific mechanism involving
accompanying many hypersensitivity states. the activation of complement and the release of
ii. Neutrophil chemotactic factors (NCF): anaphylatoxins which is the only dif­ference.
A high molecular weight chemotactic
factor has been identified, which attracts B. Localized Anaphylaxis (Atopy)
neutrophils (NCF).
The term ‘atopy’ (literally meaning out of place
5. Proteases: Enzymatic mediators, such as or strangeness) refers to naturally occurring
proteases and hydrolases are also released familial hypersensitivities of human beings. It was
from mast cell granules. These enzymes most introduced by Coca (1923) and typified by hay fever
probably are involved in the diges­tion of blood and asthma. The antigens commonly involved in
vessel basement membranes, resulting in atopy are inhalants (for example, plant pollen,
increased permeability to a variety of cell types. fungal spores, animal dander, and house dust mites
or other types of fine particles suspended in air) or
B. Secondary Mediators of Anaphylaxis ingestants (for example, milk, milk products, eggs,
1. Platelet-activating factor (PAF): Platelet- meat, fish or cereal). Some of them are contact
activating factor (PAF) is a low molec­u lar allergens, to which the skin and conjunctiva may
weight lipid released from basophils which be exposed. The symptoms depend primarily
caus­es aggregation of platelets and release of on the route by which the antigen enters the
their vasoactive amines. body. These atopens are generally not good
2. Leukotrienes and prostaglandin: They antigens when injected parenterally but induce IgE
are derived by two different pathways from antibodies, formerly termed as ‘reagin’ antibodies.
arachidonic acid, which is formed from Atopic sensitization is developed spontaneously
disrupted cell membranes of mast cells and following natural contact with atopens. It is difficult
other leukocytes. A substance originally to induce atopy artificially.
demonstrated in lungs, producing slow, Predisposition to atopy is genetically deter­
sustained contraction of smooth muscles, and, mined, probably linked to MHC genotypes. Atopy,
therefore, termed as slow reacting substance of therefore, runs in families. What is inherited is not
anaphylaxis (SRS-A) has since been identified sensitivity to a particular antigen, or a particular
a family of leu­kotrienes (LTB4, C4, D4, lM). atopic syndrome but the tendency to produce
In humans, the leukotrienes are thought to IgE antibodies in unusually large quantities. All
contribute to the prolonged bronchospasm individuals are capable of forming IgE antibodies
and buildup of mu­c us seen in asthmatics. in small amounts but in atopics IgE response is
Prostaglandin F2-α and thromboxane A2 are preponderant. About 10% of persons have this
powerful, but transient, bronchoc­onstrictors. tendency to overproduce IgE. It has been reported
3. Cytokines: Human mast cells secrete IL-4, that bottlefed infants tend to develop atopy in later
IL-5, IL-6, and TNF-α. These cytokines alter the life more often than breastfed babies.
local microenviron­ment, eventually leading to
the recruitment of inflammatory cells such as Mechanism of Atopy
neutrophils and eosinophils. The mechanism of development of atopy is essen­
tially the same as that of systemic anaphylaxis. The
Other Mediators of Anaphylaxis symptoms of atopy are caused by the release of
Besides the products of mast cells and other pharmacologically active substances following the
leucocytes, several other biologically substances combination of the antigen and the cell fixed IgE.
are known to induce mast cell degranulation and Atopic sensitivity is due to an overproduction
have been implicated in anaphylaxis. These include of IgE antibodies. This is often associated with
split products from complement activation, C3a a deficiency of IgA. When IgA is deficient, the
and C5a and bradykinin and other kinins formed antigens cause massive stimulation of IgE forming
from plasma kininogens. cells, leading to overproduction of IgE.

www.ebook3000.com
146  |  Section 2: Immunology
Clinical Expression of Atopic Reactions
The clinical expression of atopic reactions is
usually determined by the portal of entry of the
antigen-conjunctivitis, rhinitis, gastrointestinal
symptoms and dermatitis following exposure
through the eyes, respiratory tract, intestine or
skin, respectively. Sometimes the effects may be at
sites remote from the portal of entry, for example,
urticaria following ingestion of the allergen.
Specific desensitisation (hyposensitisation) is
often practised in the treatment of atopy. Atopic
allergies, which afflict at least 20% of the population
in devel­oped countries, include a wide range of Fig. 20.2: Type II hypersensitivity
IgE-mediated disor­ders, including allergic rhinitis
(hay fever), asthma, food allergies and atopic membrane of a foreign cell, or it can mediate
dermatitis (eczema). cell destruction by antibody-dependent cell-
mediated cytotoxicity (ADCC). Type II hyper­
Methods to Detect Type I sensitivity is generally called cytolytic or cytotoxic
Hypersensitivity Reactions reactions because it results in the destruction of
Formerly, measurement of reaginic antibody could host cells, either by lysis or toxic mediators. Type
be done only by an in vivo assay or by skin testing. II hypersensitive reactions involve antibody-
1. Prausnitz–Kustner (PK) reaction mediated destruction of cells (Fig. 20.2).
Prausnitz and Kustner in 1921 demonstrated
transmission of IgE-mediated type I hyper­ Examples
sensitivity by injecting serum containing IgE 1. Transfusion reactions: A transfusion reaction
antibodies from allergic person into the skin can occur if a patient receives erythrocytes
of a normal or nonallergic person. Serum differing antigenically from his or her own
from Kustner, who was hypersensitive to during blood transfusion.
certain species of cooked fish, was injected 2. Hemolytic disease of the newborn: If the child
intracutaneously in Prausnitz (normal) followed is Rh+, Rh– mother can become sensitized to
24 hours later by an intracutaneous injection of this antigen during birth causing the mother’s
cooked fish, to which Kustner was sensitive, into body to produce anti-Rh antibodies of the IgG
the same site in Prausnitz. This led to wheal and type. If the fetus in a subsequent pregnancy
flare reaction within 20 minutes. As IgE antibody is Rh+, her anti-Rh antibodies will cross the
is homocyto­tropic, the test has to be carried placenta and destroy fetal RBCs. The fetal
out on human skin. Therefore, there is risk of body responds to this immune attack by
transmission of hepatitis B virus and human producing large numbers of immature RBCs
immunodeficiency virus. call erythroblasts.
2. Skin testing: It is done by injecting small 3. Drug-induced cytotoxic reactions: Some
amounts of allergen into the skin of an allergic persons develop antibodies against their blood
individual and looking for a wheal and flare elements, resulting in autoimmune hemolytic
reac­tion. anemia, agranulocytosis or thrombocytopenia.
3. Radioimmunosorbent test (RIST): This Blood platelets (thrombocytes) that are
highly sensitive tech­n ique, based on the destroyed by drug-induced cytotoxic reactions
radioimmunoassay, can detect nanomo­lar in the disease called thrombocytopenic
levels of total IgE. purpura (quinine is a familiar example).
4. Radioallergosorbent test (RAST): It detects Drugs may bind similarly to white or red blood
the serum level of IgE specific for a given cells (RBCs), causing lo­cal hemorrhaging and
allergen. yielding symptoms described as “blueberry
muffin” skin mottling. Immune-caused
Type II Hypersensitivity: cytolytic destruc­tion of granulocytic white cells is called
agranulocytosis, and it affects the body’s
and cytotoxic
phagocytic defenses. When RBCs are destroyed
These reactions involve a combination of IgG (or in the same manner, the condition is termed
IgM)an­tibodies with an antigenic determinant hemolytic anemia.
on the surface of cells. Antibody can activate 4. Anemia due to infectious diseases: A variety
the complement system, creating pores in the of infectious diseases due to Salmonella
Chapter 20: Hypersensitivity Reactions  | 147
organisms and mycobacteria are associated of the site with neutrophils. Leukocyte-platelet
with hemolytic anemia. thrombi are formed that reduce the blood supply
and lead to tissue necrosis. The Arthus reaction
Type III Hypersensitivity: immune can be passively transferred with sera containing
complex-mediated precipitating antibodies (IgG, IgM) in high titers.
Clinical syndrome: Arthus reaction forms
Type III reactions involve antibodies against
a pathogenic component of many clinical
soluble anti­gens circulating in the serum. The
syndromes. Examples:
antigen-antibody complexes are de­p osited in
i. Farmer’s lung.
organs and cause inflammatory damage. The
ii. “Pigeon fancier’s disease”
tissue damage that results from the deposition of
immune complexes is caused by the activation of
B. Serum Sickness (Systemic Immune
complement, platelets and phagocytes; in essence,
an acute inflammatory response (Fig. 20.3). Complex Disease)
This is a systemic form of type III hypersensitivity.
Models of immune complex-mediated disease:
This appears 7–12 days following a single injection
Two basic models of immune complex-mediated
of a high concentration of foreign serum, such as the
disease have been well characterized: the Arthus
diphtheria antitoxin. The clinical syndrome consists
reaction and serum sickness. These differ
of fever, weakness, lymphadenopathy, splenomegaly,
somewhat in both their mode of devel­opment and
arthritis, glomerulonephritis, endocarditis, vasculitis,
their clinical manifestations.
urticarial rashes, abdominal pain, nausea and
vomiting.
A. Arthus Reaction (Local Immune Pathogenesis: The pathogenesis is the formation
Complex Disease) of immune complexes (consisting of the foreign
Arthus (1903) observed that when rabbits were serum and antibody to it that reaches high
repeatedly injected subcutaneously with normal enough titers by 7-12 days) and the circulating
horse serum, the initial injections were without immune complexes deposit in the blood vessel
any local effect, but with later injections, there walls and tissues, leading to increased vascular
occurred intense local reaction consisting of permeability and thus to inflammatory diseases
oedema, indura­tion and haemorrhagic necrosis. such as glomerulone­phritis and arthritis. Antigen-
This is known as Arthus reaction and is a local antibody aggregates can fix complement leading
manifestation of generalized hypersensitivity. to inflammation and tissue damage. The plasma
The tissue damage is due to forma­tion of local concentration of complement falls due to massive
precipitating immune complexes which are complement activation and fix­ation by antigen-
deposited on the endothelial lining of the blood antibody complexes. The disease is self-limited.
vessels. Antigen-antibody complexes can then The latent period of 7–12 days is required only
trig­g er and activate complement leading to for serum sickness following a single injection.
release of inflammatory molecules. This leads to Serum sickness differs from other types of
increased vascular permeability and infiltration hypersensitivity reaction in that a single injection

Fig. 20.3: Immune complex-mediated hypersensitivity. (1) Immune complexes on the basement membrane of the wall of
a blood vessel, where they: (2) activate complement and attract inflammatory cells such as neutrophils to the site; (3) The
neutrophilis discharge enzymes as they react with the immune complexes, resulting in damage to tissue cells

www.ebook3000.com
148  |  Section 2: Immunology
can serve as both as sensitizing dose and a shocking tubercu­lin (infection) type and the contact
dose. dermatitis type.
Diseases associated with immune complexes:
Complexes of antibody with various bacterial, 1. Tuberculin (Infection) Type
viral and par­asitic antigens have been shown to This form of hypersensitivity was originally
induce a variety of type III hypersensitive reactions. described by Koch. In tuberculin hypersensitivity
These include systemic lupus erythematosus, tuberculin or purified protein derivative (PPD) is
poststreptococcal glomerulonephritis, endo­ in­jected into the skin of the forearm. intradermally
carditis, dengue haemorrhagic fever, hepatitis B, in an individual sensitized to tuberculoprotein by
malaria, etc. prior infection or immunization. An indurated
(firm and hard) inflammatory reaction, 10 mm or
Type IV Hypersensitivity—delayed more in diameter, develops at the site of injection
hypersensitivity within 48–72 hours. It is characterized by erythema
Type IV hypersensitivity reactions (delayed due to increased blood flow to the damaged
hypersensitivity) constitute one aspect of cell- area and the infiltration with a large number of
mediated immune response and are caused mainly mononuclear cells, mainly T-lymphocytes and
by T-cells. It is named delayed hypersensitivity about 10–20% macrophages into the injection site
because it appears in 24–48 hours after the are responsible for the induration. In unsensitized
presensitized host encoun­ters the antigen, while individuals, the tuberculin injection provokes no
immediate hypersensitivity reactions develop in response.
1/2 to 12 hours. The tuberculin test therefore provides useful
indication of the state of delayed hypersensitivity
Causes of Type IV Reactions (cell-mediated immunity) to the bacilli. The
Type IV reactions occur when antigens, especially tuberculin test differs from the skin test for Type I
those binding to tissue cells, are phagocytosed hypersensitivity not only in the longer interval for
by macrophages and then pre­sented to receptors appearance but also in its morphology and histology.
on the THI cell surface in the context of class I Tuberculin type hypersensitivity develops in
MHC. Contact between the antigen and TH I cell many infections with bacteria, fungi, viruses and
causes the cell to proliferate and release cytokines. parasites, especially when the infection is subacute
Cytokines attract lymphocytes, macrophages, and or chronic and the pathogen intracellular. A similar
basophils to the affected tissue. Extensive tissue hypersensitivity is developed in allograft reaction
damage may result (Fig. 20.4). and in many autoimmune diseases.
Delayed hypersensitivity cannot be passively
transferred by serum but can be transferred by 2. Contact Dermatitis Type
Iymphocytes or the transfer factor. Allergic contact dermatitis is caused by haptens
Types of delayed hypersensitivity: Three types that combine with proteins in the skin to form
of delayed hypersensitivity are recognized—the the allergen that elicits the immune response.
The substances involved are in them­s elves
not antigenic but may acquire antigenicity on
combination with skin proteins. Sub­s equent
contact with allergen in a sensitized indi­vidual
leads to contact dermatitis. As most of the sub­
stances involved are fat soluble, passage along
sebaceous glands may be the method of entry of
the allergens.
Examples: Examples of these haptens include
cosmetics, plant materials (catechol molecules
from poison ivy and poison oak), topical chemo­
therapeutic agents, metals (nickel and chromium),
and chemicals like dyes, picryl chloride and
dinitrochlorobenzene, drugs such as penicillin and
toiletries and jew­elry (especially jewelry containing
nickel).
Mechanism of action: Most of these substances are
Fig. 20.4: Type IV (delayed or cell-mediated) small molecules that can complex with skin proteins.
hypersensitivity This complex is internalized by antigen-presenting
Chapter 20: Hypersensitivity Reactions  | 149
cells in the skin (e.g. Langerhans’ cells), then along with hypersensitivity reac­tions because of
processed and presented together with class II superficial resemblance.
MHC molecules, causing activation of sensitized Shwartzman (1928) observed that when a
TH cells. The sensitized T cells travel to the skin culture filtrate of S. Typhi (endotoxin) is injected
site, where on contacting the antigen they release intradermally in a rabbit, followed by the same
various lymphokines. Approximately 48–72 hours filtrate (endotoxin) intravenously 24 hours later, a
after the second exposure, the secreted cytokines hemorrhagic necrotic lesion develops at the site of
cause macrophages to accumulate at the site. Tissue the intradermal injection.
damage results from lytic enzymes released from
activated macrophages. Contact with the allergen in a Mechanism
sensitised individual leads to ‘contact dermatitis’. The
The first dose is called prepara­tory dose. The
lesions varying from macules and papules to vesicles
second dose is called provoca­t ive dose. The
that break down, leaving behind raw weeping areas
preparatory injection causes accumulation of
typical of acute ec­zematous dermatitis.
leukocytes which condition the site by re­lease
Detection by ‘patch test’: Hypersensitivity is of lysosomal enzymes. These enzymes damage
detected by the ‘patch test’. The allergen is applied capillary walls. Fol­lowing provocative dose, there
to the skin un­der an adherent dressing. Sensitivity occurs intravascu­lar clotting, the thrombi leading
is indicated by itching appearing in 4–5 hours, and to necrosis of ves­sel walls and hemorrhage. This is
local reaction which may vary from erythema to called local form of Shwartzman reaction.
vesicle or blister formation, after 24–28 hours. When both injections are given intravenously,
the animal dies 12–24 hours after the second dose.
3. Granulomatous Hypersensitivity The animal develops disseminated intravascular
Granulomatous hypersensitivity reactions develop coagulation (DIC). Autopsy reveals bilateral
over a period of 21–28 days; the granulomas are cortical necrosis of kid­neys and patchy necrosis of
formed by the aggregation and proliferation of the liver, spleen and other organs. Sanarelli (1924)
macrophages, and may persist for weeks. In terms described an essentially similar phenomenon in
of its clinical consequences, this is by far the most experimental cholera. The reaction is, therefore,
serious type of Type 1V hypersensitivity response. called the Sanarelli–­Shwartzman reaction or the
Examples are leprosy, tuberculosis, leish­ generalized Shwartzman reaction.
maniasis, candidiasis and herpes simplex lesions.
Clinical Conditions
Type V: Hypersensitivity 1. Water­house–Friderichsen syndrome: Pururic
(Stimulatory Type) Jones–Mote rashes of meningococ­cal septicemia and the
acute hemorrhagic adrenal necrosis found
Reaction (or) Cutaneous Basophil in overwhelming infections (Waterhouse–
Hypersensitivity Friderichsen syndrome) mechanisms similar
This is an antibody-mediated hypersensitivity and to the Shwartzman reaction may operate.
is a modification of type II hypersensitivity reaction. 2. Septic shock syndrome.
Antibodies interact with antigens on cell surface
which leads to cell proliferation and differentiation Key Points
instead of inhibition or killing. Antigen-antibody
reaction enhances the activity of affected cell. ™™ Hypersensitivity is an exaggerated immune
response that results in tissue damage and is
Example: Graves’ disease: Thyroid hormones are manifested in the individual on second or subsequent
produced in excess quantity in Graves’ disease. contact with an antigen. Hypersensitivity reactions
are of 5 types: Types I, II, III, IV, and V
Long-acting thyroid stimulating (LATS) antibody
™™ Type I hypersensitive reaction is mediated by IgE
is an autoantibody to thyroid membrane antigen. anti­bodies
It is presumed that LATS combines with a TSH ™™ Clinical manifestations of type I reactions in­clude
receptor on thyroid cell surface and brings about generalized or systemic anaphylaxis or localized
the the same effect as TSH resulting in excessive anaphylactic
secretion of thyroid hormone. ™™ Type II hypersensitive reactions, or cytotoxic
reactions are caused by antibodies that can destroy
normal cells by complement lysis or by antibody-
Schwartzman reaction dependent cellular cyto­toxicity (ADCC). Transfusion
reactions and hemolytic dis­ease of the newborn are
This is not an immune reaction but rather a type II reactions
perturbation in factors affecting intravascular ™™ Type III hypersensitivity reactions are mediated
coagulation. It is, however, traditi­onally described by small antigen–antibody complexes that activate

www.ebook3000.com
150  |  Section 2: Immunology
2. All are primary mediators of anaphylaxis except:
complement and other inflammatory systems,
a. Histamine
attract neutrophils, and contribute to inflammation.
Deposition of im­mune complexes near the site
b. Proteases
of antigen entry can induce an Arthus reaction, c. Eosinophil chemotactic factors of anaphylaxis
(localized reaction) and serum sickness (systems d. Platelet-activating factor
form) 3. Schultz-Dale phenomenon is an example of:
™™ Type IV hypersensitive reactions involve the cell- a. Type I hypersensitivity reaction
mediated branch of the immune system. b. Type II hypersensitivity reaction
Three types of delayed hypersensitivity are: c. Type III hypersensitivity reaction
i. Tuberculin skin test; ii. Contact d. Type IV hypersensitivity reaction
Hypersensitivities; iii. Granulomatous. 4. All the following statements are true for Arthus
reaction except:
a. It is a local manifestation of generalized hyper-
Important questions sensitivity.
b. The tissue damage is due to formation of local
1. What is hypersensitivity? How do you classify precipitating immune complexes.
various types of hypersensitivity reactions? Des­ c. This is systemic form of type III hypersensitivity.
cribe type I hypersensitivity reactions. d. It manifests after a single injection of a high
concentration of foreign serum
2. Write short notes on:
a. Anaphylaxis 5. All the following statements are true for serum
b. Atopy sickness except:
a. It manifests after a single injection of a high
c. Type III hypersensitivity or immune complex
concentration of foreign serum
diseases
b. Disease is self-limited and clears without
d. Arthus reaction
sequelae
e. Serum sickness
c. This is systemic form of type III hypersensitivity.
f. Type IV hypersensitivity or delayed hypersensi- d. It is a localized inflammatory reaction due to
tivity. deposition of immune complexes.
6. Delayed hypersensitivity reaction is mediated by:
Multiple choice questions (MCQs) a. T-lymphocytes b. B-lymphocytes
c. Macrophages d. Basophils
1. All the following statements are true for type I 7. Shwartzman reaction is an example of:
hypersensitivity reaction except: a. Type I hypersensitivity reaction
a. It is called immediate hypersensitivity reaction b. Type II hypersensitivity reaction
b. It always involves IgE-mediated degranulation c. Type III hypersensitivity reaction
of basophils or mast cells d. None of the above.
c. This reaction is always rapid Answers (MCQs)
d. Atopy is one of the manifestations 1. b; 2. d; 3. a; 4. d; 5. d; 6. a; 7. d
21
Chapter

Autoimmunity

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Classify autoimmune diseases
be able to: ∙∙ List autoimmune diseases.
∙∙ Describe the mechanisms of autoimmunity

INTRODUCTION influences. Such altered antigens or ‘neoantigens’


may elicit an immune response.
One of the classically accepted features of the
immune system is the capacity to distinguish self Several chemical agents including drugs can
from non-self. combine with cells and tissues and alter their
antigenic structure. Skin con­tact with a variety of
Definition chemicals may lead to contact dermatitis. Drug-
induced anemia, leukopenia and thrombocytopenia
Autoimmunity is a condition in which structural often have autoimmune basis. Viruses and other
or functional damage is produced by the intracellular pathogens may induce alterations of
action of immunologically competent cells or cell antigens leading to auto­immunity.
antibodies against the normal components of the
body. Autoimmunity is due to copious production
of autoantibodies and autoreactive T-cells. 3. Molecular Mimicry
Autoimmunity literally means ‘protection against A number of viruses and bacteria have been shown
self ’ but it actually implies ‘injury to self ’ and, to possess antigenic determinants that are identical
therefore, it has been criticized as a contradiction or similar to normal host-cell components. The
in terms. fortuitous similarity between some foreign and
self-antigens is the basis of the ‘cross-reacting
MECHANISMS OF AUTOIMMUNITY antigens’ theory of autoimmunity. Molecular
mimicry by cross-reactive microbial antigens can
A variety of mechanisms have been proposed for
stimulate autoreactive B- and T-cells.
induction of autoimmunity.
Organ-specific antigens are present in several
1. Forbidden Clones species. Injections of heterologous organ-specific
Antibody-forming lymphocytes capable of reacting antigens may induce an immune response
with different antigens are formed according to damaging the particular organ or tissue in the
clonal selection theory. During embryonic life, host. One of the best examples of this type of
clones of cells that have immunological reactivity autoimmune reaction is postrabies encephalitis.
with self-antigens are eliminated. Such clones Infection: Streptococcal M proteins and the
are called forbidden clones. Their persistence or heart muscle share antigenic characteristics.
development in later life by somatic mutation can The immune response induced by repeated
lead to autoimmunity. streptococcal infection can, therefore, damage
the heart. Nephritogenic strains of streptococci
2. Neoantigens or Altered Antigens possess antigens found in the renal glomeruli.
Cells or tissues may undergo antigenic alteration Infection with such strains may lead to the
as a result of physical, chemical or biological glomerulonephritis due to antigenic sharing.

www.ebook3000.com
Chap-21.indd 151 15-03-2016 11:12:48
152  |  Section 2: Immunology
4. Polyclonal B-cell Activation the development of autoimmunity in certain
circumstances.
While an antigen generally activates only its
corresponding B-cell, certain stimuli nonspecifically
turn on multiple B clones. Such stimuli include 8. Genetic Factors
chemicals (for example, 2-mercaptoethanol), Their actual role in autoimmunity, if any, has not
bacterial products (PPD, lipopolysaccharides), been established.
enzymes (trypsin), antibiotics (nystatin) and
infection with some bacteria (mycoplasma), viruses CLASSIFICATION OF AUTOIMMUNE
(EB virus, CM virus) and parasites (malaria).
DISEASES

5. Activity of Helper and Suppressor Based on the site of involvement and nature of
lesions, autoimmune diseases may be classified
T-cells as (Table 21.1):
Enhanced helper T-cell and decreased suppressor A. Localized (or organ-specific)
T-cells functions have been suggested as causes of B. Systemic (or nonorgan-specific)
autoimmunity.
A. Localized (Organ-specific)
6. Sequestered Antigens Autoimmune Diseases (Table 21.1)
Certain self-antigens are present in closed systems The immune response is directed to a target
and are not accessible to the immune apparatus. antigen unique to a single organ or gland in an
These are known as sequestered antigens. organ-specific autoimmune disease, so that the
manifestations are largely limited to that organ.
Examples
B. Systemic (or Nonorgan-specific)
i. Lens antigen of the eye: The lens protein is
enclosed in its capsule and does not circulate Autoimmune Disease (Table 21.1)
in the blood. Hence, immunological tolerance In systemic autoimmune diseases, the response is
against this antigen is not established during directed toward a broad range of target antigens
fetal life. The release of lens protein after eye and meshces a number of organs and tissues.
damage has been shown to lead on occasion
to the formation of autoantibodies. When Pathogenesis of Autoimmune Disease
the antigen leaks out, following penetrating Many diseases are considered to be of autoimmune
injury, it may induce an immune response origin, based on their association with cellular or
causing damage to the lens of the other eye. humoral immune responses against self-antigens.
ii. Sperm antigens: Sperms arise late in develop­
ment and sequestered from the circulation. As Humoral and cellular immune processes: The
spermatozoa develop only with puberty, the relative importance of humoral and cellular
antigen cannot induce tolerance during fetal immune processes in the etiology of autoimmune
life. However, after a vasectomy, some sperm diseases is not known. Antibodies may cause dam­
antigens are released into the circulation age by the cytolytic or cytotoxic (type 2) and toxic
and can induce auto-antibody formation in complex (type 3) reactions. They are obviously
some men. This is also believed to be the im­portant in hemocytolytic autoimmune diseases.
pathogenesis of orchitis following mumps. A third mechanism of autoimmune tissue
The virus damages the basement membrane damage is by sensitized T-lymphocytes (type 4
of seminiferous tubules leading to the leakage reaction). It is like­ly that humoral and ceIlular
of sperms and initiation of an immune immune responses may act synergistically in the
response resulting in orchitis. production of some autoim­mune diseases. For
example, experimental orchitis can be induced
only when both types of immune responses are
7. Defects in the Idiotype–Anti-idiotype operative.
Network Autoimmune disease are usually treated with
It is possible that abnormalities in the generation immunosuppressants, or drugs that interfere
of appropriate anti-idiotype antibodies, either with T-cell signaling, steroids and other anti-
at the B- or T-cell level, are responsible for inflammatory drugs.

Chap-21.indd 152 15-03-2016 11:12:48


Chapter 21: Autoimmunity  | 153
Table 21.1  Some autoimmune diseases in humans
Disease Self-antigen Immune response
ORGAN-SPECIFIC AUTOIMMUNE DISEASES
Hashimoto’s thyroiditis Thyroid proteins and cell Autoantibodies
Graves’ disease Thyroid-stimulating hormone receptor Autoantibodies
Goodpasture’s syndrome Renal and lung basement membranes Autoantibodies
Autoimmune hemolytic anemia RBC membrane proteins Autoantibodies
Addison’s disease Adrenal cells Autoantibodies
Idiopathic thrombocyopenia Platelet membrane proteins Autoantibodies
purpura
Insulin-dependent diabetes Pancreatic beta cells T DTH autoantibodies
mellitus
Myasthenia gravis Acetylcholine receptors Autoantibody (blocking)
Myocardial infarction Heart Autoantibodies
Pernicious anemia Gastric parietal cells; intrinsic factor Autoantibodies
Poststreptococcal Kidney Antigen-antibody complexes
glomerulonephritis
Myasthenia gravis Acetylcholine receptors Auto- antibodies (blocking)
Spontaneus infertility Sperm Autoantibodies
SYSTEMIC AUTOIMMUNE DISEASES
Systemic lupus erythematosus DNA, nuclear protein, RBC and platelet Autoantibodies, immune complexes
(SLE ) membranes
Multiple sclerosis Brain or white matter T H1 cells and Tc cells, auto-antibodies
Rheumatoid arthritis Connective tissue, IgG Autoantibodies, immune complexes
Scleroderma Nuclei, heart, lungs, gastrointestinal tract, Autoantibodies
kidney
Sjogren’s syndrome Salivary gland, liver, kidney, thyroid Autoantibodies
Ankylosing sponkylitis Vertebrae Immune complexes

Key Points 2. Write short notes on:


a. Classification of autoimmune diseases
™™ Autoimmunity is a condition in which structural b. Sequestrated antigens or hidden antigens.
or functional damage is produced by the action
of immunologically competent cells or antibodies
against the normal components of the body MULTIPLE CHOICE QUESTIONS (MCQs)
™™ A variety of mechanisms have been proposed for
induction of autoimmunity, such as 1. Forbidden 1. Lens protein of eye is an example of:
clones; 2. Neoantigens or altered antigens; 3.
Molecular mimicry; 4. Polyclonal B-cell activation; a. Sequstered antigen
5. Activity of helper and suppressor T-cells; b. Neoantigen
6. Sequestered antigens 7. Defects in the idiotype- c. Cross-reacting antigen
antiidiotype network d. Molecular mimicary
™™ Autoimmune diseases can be divided into organ
specific or widespread and systemic diseases 2. All of the following diseases are examples of organ-
™™ Autoimmune diseases are usually treated with drugs specific autoimmune diseases except:
that suppress the immune and/or inflammatory a. Goodpasture’s syndrome
responses. b. Graves’ disease
c. Insulin-dependent diabetes mellitus
IMPORTANT QUESTIONS d. Systemic lupus erythematosus

1. What is autoimmunity? What are the mechanisms ANSWERS (MCQs)


of autoimmune diseases, giving suitable examples? 1. a; 2. d.

www.ebook3000.com
Chap-21.indd 153 15-03-2016 11:12:48
22
Chapter
Immunology of
Transplantation and Malignancy

Learning Objectives

After reading and studying this chapter, you should ∙∙ Histocompatibility antigens
be able to: ∙∙ Graft versus host (GVH) reaction
Discuss the following: ∙∙ Tumor antigens
∙∙ Types of transplants ∙∙ Immunological surveillance.

Definition morphologically and functionally healthy


during the first two or three days. By about the
Transplantation refers to the act of transferring fourth day, inflammation becomes evident
cells, tissues or organs from one site to another. and the graft is invaded by lymphocytes and
The tissue or organ transplanted is known as the macrophages. The blood vessels within the
transplant or graft. The individual from whom the graft are occluded by thrombi, the vascularity
transplant is obtained is known as the donor and diminishes and the graft undergoes ischemic
the individual to whom it is applied, the recipient necrosis. The graft assumes a scab-like
or host. appearance with extending necrosis and
sloughs off by the tenth day. This sequence of
Types of transplants (Table 22.1) events resulting in the rejection of the allograft
is known as the first set response (also known
i. Autograft: It is self-tissue transferred from one as the ‘first set rejection or reaction’).
body site to another in the same individual. 2. Second set response: If in an animal which has
ii. Isograft: It is tissue transferred between rejected a graft by the first set response, and
genetically identical individuals. In humans, another graft from the same donor is applied,
an isograft performed between genetically it will be rejected in an accelerated fashion.
identical (monozygomatic) twins and in Vascularization commences but is soon
inbred strains of mice, an isograft performed
from one mouse to another syngeneic mouse Table 22.1  Types of grafts
are examples of isografts.
Donor Name Synonyms
iii. Allograft: It is tissue transferred between
genetically different members of the same Self Autograft Autogenous or
autogenic graft
species.
D i f fe re nt i n d i v i d u a l, Isograft Isologous or
iv. Xenograft: It is tissue transferred between geneti­cally identical with syngeneic graft
different species (e.g. the graft of a baboon recipient. Identical twin or syngraft
heart into a human). or member of same inbred
strain
Allograft reaction Genetically unrelated Allograft Allogeneic
member of same species graft. Formerly
1. First set response: When a skin graft from called
an animal (such as a rabbit) is applied on homograft
a genetically unrelated animal of the same Different species Xenograft Xenogeneic
species, the graft appears to be accepted Formerly called
initially. The graft is vascularized and seems heterograft
Chapter 22: Immunology of Transplantation and Malignancy  | 155
interrupted by the inflammatory response. and recipient are distributed into a series
Thrombosis of vessels is a feature. Necrosis of wells on a microtiter plate, and then
sets in early and the graft sloughs off by the antibodies for various class I and class II
sixth day. The accelerated allograft rejection is MHC alleles are added to different wells.
known as the second set response. After incubation, complement and a dye,
such as trypan blue, are then added. Cells
Mechanism of Allograft Rejection carrying antigens corresponding to the
Graft rejection is caused principally by a cell- HLA antiserum are killed by complement
mediated immune response to alloantigens mediated membrane damage. If antibodies
(primarily, MHC molecules) expressed on cells of in the antiserum have reacted specifically
the graft. with the lymphocyte, the cell is damaged.
The dam­aged cell will take up the dye
(undamaged cells will not), thus indicating
Histocompatibility Antigens that the lymphocyte possesses a certain
Tissues that are antigenically similar are said to antigen. This method is simple and rapid.
be histocompatible; such tissues do not induce ii. Molecular methods: Restriction fragment
an immunologic response that leads to tissue length polymorphism (RFLP) with
rejection. The various antigens that determine southern blotting, and polymerase chain
histocompatibility are encoded by more than 40 reaction(PCR) amplification.
different loci, but the loci responsible for the most
vigorous allograft-rejection reactions are located Tissue Matching
within the major histocompatibility complex A one-way mixed lymphocyte reaction (MLR)
(MHC). In mice the organization of the MHC is can be used to determine identity of class II HLA
called H2 complex, while in humans it is known as antigens between a potential donor and recipient.
human leucocyte antigen (HLA) complex.
Immune response against transplants depends Factors Favoring Allograft Survival
on the presence in the grafted tissue of antigens that 1. Blood group and major histocompatibility
are absent in the recipient and hence recognized as antigens: There are three classes of transplant­
foreign. If the recipient possesses all the antigens ation antigens: (1) ABO group antigens;
present in the graft, there will be no immune (2) MHC antigens; (3) Minor histocompatibility
response, and consequently no graft rejection. antigens.
2. Immunosuppression
Histocompatibility Testing a. General immunosuppressive therapy
For matching of donor and recipient for transplant­ The three nonspecific agents that are most
ation, following procedures are undertaken: widely used in current practice are steroids,
1. ABO grouping: ABO incompatibility is a major cyclosporine and azathioprine.
barrier to transplantation, and matching of b. Specific immunosuppressive therapy
donor and recipient for these antigens is of Specific immunosuppression reduces
utmost importance. antigraft response without increasing
2. Tissue typing (detection of MHC antigens): susceptibility to infection. In humans,
In serological tissue typing, the laboratory procedures, such as total lymphoid
uses standardized antisera or monoclonal irradiation (TLI) and use of antilymphocyte
antibodies that are specific for par­ticular HLAs. serum (ALS) are widely used in heart
Class I antigens are identified by means of transplant recipients to deplete circulating
antisera. Antisera for HLA typing were originally T-cells.
obtained from multiparous women (i.e. women 3. Immune tolerance to allografts: Obviously,
who have had multiple pregnancies), placental in the case of tissues that lack alloantigens,
fluid and from individuals who have received such as cartilage or heart valve, there is no
multiple transfusions, from individuals who immunological barrier to transplantation.
have received and rejected grafts, and from 4. Privileged sites: An allograft can be placed
volunteers who have been immunized with without engendering a rejection reaction, in
cells from another individual with a different immunologically privileged sites. These sites
HLA haplotype. These are being replaced by include the anterior chamber of the eye,
monoclonal antibodies. Following methods are the cornea, the uterus, the testes, and the
used: brain. Each of these sites is characterized by
i. Microcytotoxicity test: In this test, white an absence of lymphatic vessels and in some
blood cells from the potential donors cases by an absence of blood vessels as well.

www.ebook3000.com
156  |  Section 2: Immunology
Fetus as An allograft whole-body irradiation) that he cannot reject
a graft (allograft or xenograft).
Fetus is always a mixture of maternal and paternal
GVHD is a major complication of bone-
genes; therefore, fetal MHC antigens are always
marrow-transplantation and affects 50–70% of
different from those of the mother. The fetus can be
bone-marrow-transplant patients. It develops as
considered an intrauterine allograft, as it contains
donor T-cells recognize alloantigens on the host
antigens which are foreign to the mother. In spite
cells. The activation and proliferation of these
of this, fetus is not treated as foreign transplanted
T-cells and the subsequent production of cytokines
tissue and rejected. The fetus does not reject the
generate inflammatory reactions in the skin,
mother which may be due to the fact that the
gastrointestinal tract and liver.
developing immune system of the fetus cannot
The major clinical features of the GVH in
respond. The reason why the fetus is exempt from
animals are retardation of growth, emaciation,
rejection is not clear, though many explanations
diarrhea, hepatosplenomegaly, lymphoid atrophy
have been offered.
and anemia, terminating fatally. The syndrome has
1. Immunological barrier: The placenta acts
been called runt disease.
as an immunological barrier by generating a
hormone which is locally immunosuppressive.
2. Mucoproteins: They are produced by cells of Immunology of malignancy
the placenta which coat fetal cells, thus masking When a cell undergoes malignant transformation,
histo­c ompatibility antigens and prevent it acquires new surface antigens. It may lose
recognition. some normal antigens and this makes a tumor
3. Soluble inhibitory factor: It can be produced antigenically different from the normal tissues of
by placental giant cells which suppresses T-cell the host. A tumor can, therefore, be considered an
proliferation and antibody production, and allograft and be expected to induce an immune
induces a population of T-cells. Hormones response.
(human chorionic gona­dotropin, produced
by placenta, and the high levels of maternal
Tumor Antigens
progesterone produced during pregnancy) also
cause Immunosuppression. Tumor antigens are antigens that are present in
4. Blocking antibodies: The mother produces malignant cells but absent in the corresponding
specific blocking antibodies to fetal antigens of normal cell of the host. Two types of tumor antigens
the fetal cells thus blocking immune recognition have been identified on tumor cells:
and immune attack by maternal Tc-cells. 1. Tumor-specific transplantation antigens
5. Major histocompatibility complex (MHC) (TSTAs): Tumor-specific antigens are unique
antigens: There antigens are present only in to tumor cells and do not occur on normal
low density on trophoblastic cells and the cell cells in the body. Different tumors possess
membranes are relatively resistant to attack by different TSTA, even though induced by the
T- or K-cells. same carcinogen. In contrast, TSTA of virus
6. Alphafetoprotein: The high concentration of induced tumors is virus specific in that all
alphafetoprotein in fetal blood also may be a tumors produced by one virus will possess
factor, as it has immunosuppressive properties. the same antigen, even if the tumor occur in
different animal strains or species.
Graft-versus-host reaction 2. Tumor-associated transplantation antigens
(TATAs): These are present on tumor cells
Graft rejection is due to the reaction of the host to
and also on some normal cells. Since they are
the grafted tissue (host-versus-graft response). The
also present on some normal cells, therefore,
contrary situation, in which the grafted tissue may
they do not evoke an im­mune response and
react to and reject the host, is known as the graft-
are of little significance in tumor rejection.
versus-host (GVH) reaction.
The more common tumor-associated antigens
The GVH reaction occurs when the following
are oncofetal antigens and increased levels of
conditions are present:
normal oncogene products.TATAs fall into three
1. The graft (bone marrow, lymphoid tissue,
categories:
splenic tissue, etc.) contains immunocompetent
T-cells. i. Tumor associated carbohydrate antigens
2. The recipient possesses transplantation anti­ (TACAs), such as mucin-associated antigen
gens that are absent in the graft. detected in pancreatic and breast cancers.
3. The recipient must not reject the graft. The ii. Oncofetal tumor antigens: Oncofetal
host’s immunological responsiveness must tumor antigens, as the name implies,
be either destroyed or so impaired (following are found not only on cancerous cells but
Chapter 22: Immunology of Transplantation and Malignancy  | 157
also on normal fetal cells. These antigens It is evident that tumor cells must develop
appear early in embryonic development, mechanisms to escape or evade the immune
before the immune system acquires system in immuno­competent hosts. Several such
immunocompetece. If these antigens mechanisms may be operative:
appear later on cancer cells, they are 1. Weak immunogenicity: Some tumors are
recognized as nonself and induce an weakly immunogenic, so in small numbers they
immunologic response. Two well-studied do not elicit an immune response. But when
oncofetal antigens are alpha-fetoprotein their numbers increase enough to provoke
(AFP) in hepatoma and cacinoembryonic immune response the tumor load may be too
antigen (CEA) found in colonic cancer. great for the host’s immune system to mount
an effective response.
iii. Differentiation antigens: They are peculiar
2. Modulation of surface antigens: Certain
to the differentiation state at which cancer
tumor-specific antigens disappear from the
cell are arrested. For example, CD10, an
surface of tumor cells in the presence of serum
antigen expressed in early B lymphocytes, antibody and then to appear after the antibody
is present in B-cell leukaemias. Similarly, is no longer present.
prostate specific antigen (PSA) is expreseed 3. Masking tumor antigens: Certain cancers
on the normal as well as cancerous produce copious amounts of a mucoprotein
prostatic epithelium. Both serve as useful called sialomucin. It binds to the surface of the
differentiation markers in the diagnosisi of tumor cells. Immune system does not reco­gnize
lymphoid and prostatic cancer. these tumor cells as foreign since sialomucin is
a normal component.
Immune response in malignancy 4. Induction of immune tolerance: Some tumor
cells can synthesize various immunosup­
Tumors can induce potent immune responses. In pressants.
experimental animals, tumor antigens can induce 5. Production of blocking antibodies: Antitumor
both humoral and cell-mediated immune responses antibody itself acts as a blocking factor.
that result in the destruction of the tumor cells. In 6. Low levels of HLA class I molecules: This
general, the cell-mediated response appears to play impairs presentation of antigenic peptides to
the major role. The immune response to tumor cytotoxic T-cells.
includes cytotoxic T lymphocytes (CTL) mediated
lysis, natural killer (NK)-cell activity, macrophage-
Immunotherapy of cancer
mediated tumor destruction, and destruction
mediated by antibody-dependent cell mediated Different approaches have been attempted in
cytotoxicity (ADCC). Several cytotoxic factors, the immunotherapy of cancer-active or passive,
including, TNF-α and TNF-b, help to mediate specific or nonspecific.
tumor-cell killing. A. Active
∙∙ Nonspecific
Immunological surveillance ∙∙ Specific
B. Passive
The immune surveillance theory was first
∙∙ Nonspecific
conceptualized in the early 1900s (1906) by Paul
∙∙ Specific
Ehrlich. He suggested that cancer cells frequently
arise in the body but are recognized as foreign ∙∙ Combined
and eliminated by the immune system. Some 50
years later, Lewis Thomas revived it in 1950s and A. Active Immunotherapy
was developed by Burnet. It postulates that the 1. Nonspecific active immunotherapy: Biological
primary function of cell mediated immunity is response modifiers (BRMs) are used to enhance
to ‘seek and destroy’ malignant cells that arise by immune responses to tumors and fall into four
somatic mutation. Such malignant mutations are major groups—(i) Bacterial products; (ii) Synthetic
believed to occur frequently and would develop molecules; (iii) Cytokines; (iv) Hormones.
into tumors but for the constant vigilance of the i. Bacterial products: Broadly speaking,
immune system. Inefficiency of the surveillance bacterial products such as BCG and nonliving
mechanism, either as a result of ageing or in Corynebacterium parvum have adjuvant
congenital or acquired immunodeficiencies, leads effects on macrophages. Intralesional BCG
to an increased incidence of cancer. can cause regression of melanoma and
The development of tumors represents a lapse nonspecific local immunization with BCG is
in surveillance. effective against bladder tumors.

www.ebook3000.com
158  |  Section 2: Immunology
ii. Synthetic molecules: Dinitrochlorobenzene ™™ Graft-versus-host (GVH) reaction to occur requires
has been tried in the treatment of squamous three important components:
and basal cell carcinoma of the skin. Glucan, a The donor graft must contain immunocompetent
pyran polymer derived from micro-organisms, T-cells. The host must be immunocompromised.
and levamisole, originally introduced as an The recipient should express antigens which will be
antihelmintic, have been tried for stimulating identified as foreign to the donor
™™ Tumor antigens can be classified as tumor-
cell mediated immunity and macrophage specific antigens (TSAs) and tumor-associated
functions. transplantation antigens (TATAs). The TSAs are
iii. Cytokines: Immunotherapy with cytokine can unique to tumors and not found on other cells of
cause tumor regression. the body
™™ Cancer should not occur if immunological
iv. Hormones: Thymic hormones can be used to surveillance is effective.
enhance T-cell function.
2. Specific active immunotherapy: This immuno­ Important questions
therapy includes therapeutic vaccines of tumor 1. What is a transplant or graft? Define various types
cells, cell extracts, purified or recombinant of grafts.Describe allograft reaction.
antigens, peptides, heat shock proteins or DNA 2. What are histocompatibility antigens? Describe
antigen-pulsed dendritic cells. Specific active various procedures for histocompatibility testing.
immunotherapy by the injection of tumor cell 3. Write short notes on:
‘vaccines’ was tried early in this century but was a. Mechanism of allograft rejection
given up unprofitable. b. Graft versus host (GVH) reaction
c. Tumor antigens
d. Immunological surveillance
B. Passive Immunotherapy
i. Nonspecific (lymphokine-activated killer Multiple choice questions (mcqs)
(LAK) cells)
ii. Specific (antibodies alone or coupled to 1. Tissue transferred between two genetically different
drugs, pro drugs, toxins or radioisotope, members of the same species are known as:
a. Autografts b. Isografts
bi-specific antibodies, T-cells)
c. Allografts d. Xenografts
iii. Combined (LAK cells and bispecific antibody). 2. Acute rejection shows all the following features
except:
Key Points a. It takes days or weeks after transplantation.
b. It is due to the primary activation of T-cells
™™ Transplantation can be defined as the transfer of c. Acute rejection is the typical first-set rejection
cells, tissues, or organs from one site in an individual d. Vascular endothelial injury is the most common
to another, or between two individuals feature
™™ There are four different basic types of transplants: 3. The following cell may provide the first line of
autograft, isograft, allograft and xenograft defence against many tumors:
™™ The immune response to tissue antigens encoded a. Eosinophils
within the major histocompatibility complex is the b. Natural killer cells
strongest force in rejection c. Monocytes
™™ The match between a recipient and potential graft d. None of the above
donors is assessed by typing MHC class I and class
II tissue antigens Answers (MCQs)
1. c; 2. d; 3. b
23
Chapter

Immunohematology

Learning Objectives

After reading and studying this chapter, you should ∙∙ List various infectious agents transmitted via blood
be able to: transfusion.
∙∙ Describ Rh blood groups
∙∙ Discuss complications of blood transfusion

History H Antigen
The ABO system is the most important of all Red cells of all ABO groups possess a common
the blood group systems and its discovery made antigen, the H antigen or H substance which is the
blood transfusion possible. No other blood group precursor for the formation of A and B antigens. H
antigens were discovered for the next 25 years. antigen is not ordinarily important in grouping or
Subsequently, other blood groups (MN, P, Rh, blood transfusion due to its universal distribution.
Luthe­ran, Lewis, Kell, Duffy, Kidd, Diego, Yt, Kg, However, Bhende et al. (1952) from Bombay
Dom­broc and Colton) were reported. reported a very rare instance in which A and B
antigens as well as H antigens were absent from the
red cells. This is known as Bombay or OH blood
ABO Blood Group System group. Sera of these individuals have anti-A, anti-B
The ABO blood group system was originally and anti-H antibodies. Therefore, they can accept
described by Landsteiner (1900) and now contains the blood only from the same rare blood group.
four blood groups. The blood group is determined A, B and H antigens are glycoproteins. They
by the presence or abse­nce of two distinct antigens are also present in almost all the tissues and fluids
A and B on the surface of the erythrocytes. It is of the body in addi­tion to erythrocytes. They are
these antigens (also called agglunotigens) that found in secretions (saliva, gastric juice, sweat) of
cause blood transfusion reactions. Red cells of only about 75% of all persons while these antigens
group A carry antigen A, cells of group B antigen B are always present in tissues. Such persons are
and cells of group AB have both A and B antigens, called ‘secretors’ and those who lack blood group
while group O cells have neither A nor B antigen. antigens in secretions are called ‘nonsecretors’.
The serum contains the isoantibodies specific
for the antigen that is absent on the red cell. The Table 23.1  Distribution of ABO antigens on the
serum of a group A individual has anti-B antibody, red blood cells and antibodies in the serum
group B has anti-A and group O both anti-A and Blood Antigens on Antibodies Occurrence
anti-B, while in group AB, both anti-A and anti-B group red blood cells in serum (%) in India
are absent (Table 23.1). A A Anti-B 22
The frequency of ABO distribution antigens B B Anti-A 33
differs in different people. Group O is the most
AB A and B None 5
common group and AB the rarest. In India, the
distribution is approximately O–40%, A—22%, O None Anti-A and 40
Anti-B
B—33% AB—5%.

www.ebook3000.com
160  |  Section 2: Immunology
Rh (Rhesus) Blood Group System possessed neither A nor B antigen. Hence
It was discovered by Landsteiner and Weiner in the O group was designated as the ‘universal
1940. Their experiment was to produce an antibody donor’. The anti-A and anti-B antibodies in
to the red cells of the Rhesus monkey in rabbits the transfused O blood group do not ordinarily
and guinea pigs, but they discovered that not only cause any damage to the red cells of the A or B
did the antibody in the rodents’ serum agglutinate group recipients because they will be rendered
the Rhesus monkey red cells, it also agglutinated ineffective by dilution in the recipient’s plasma.
the red cells of 85% of the human population. If an But some O group plasma may contain
individual’s red cells were clumped together by this isoantibodies in high titers (1:200 or above)
antiserum, they were said to have the Rhesus factor so that damage to recipient cells may result.
on their red cells (i.e. Rh positive). If an individual’s This is known as the ‘dangerous O group’. The
cells were not agglutinated by the antiserum, they AB group persons were designated ‘universal
were said to lack the Rhesus factor (i.e. Rh negative). recipients’ due to the absence of isoantibodies
Rh antigens—“Rh-positive” and “Rh-negative” in plasma.
people. 2. Rh compatibility: An Rh positive person may
There are six common types of Rh antigens, safely receive either Rh positive or negative
each of which is called an Rh factor. These types are blood. But an Rh-negative individual receiving
designated as C, D, E, c, d, and e. The designations Rh-positive blood may form antibodies against
employed by the two system for the different Rh the Rh-antigen. A subsequent transfusion with
types are as follows: Rh positive blood may then cause a rapid,
serious hemolytic reaction.
The type D antigen (Rho) is widely present in the
population and considerably more antigenic than
the other Rh antigens. Rh-positive or Rh-negative Complications of blood
blood depends on the presence or absence of transfusion
D-antigen on the surface of red cells respectively. The complications of blood transfusion may be divi­
It can be accomplished by testing with anti-D ded into immunological and nonimmunological.
(anti-Rh) serum. About 15% of the population have
A. Immunological complications: Immunological
no RhD antigens and thus are “Rh negative”. Among
complications may be caused by red cell,
Indians, approximately 93% are Rh positive and
leukocyte or platelet incompatibility or allergic
about 7% negative. The Rh factor can be detected
reaction to plasma components. Red cell
by testing the blood with anti-D (anti-Rh) serum.
incompati­bility leads to acute intravascular
hemolysis or the red cells may be coated
Other blood group systems by antibodies and engulfed by phagocytes,
Blood group systems other than ABO and Rh are removed from the circulation and subjected to
of little clinical importance as they do not usually extravascular lysis. Hemolysis may also be due
cause transfusion reactions or hemolytic disease. to transfusion of group O whole blood or plasma
They have applications in genetics, anthropology, to group A or group B or group AB recipients.
tissue typing and forensic medicine. As blood group B. Nonimmunological complications: Non-
antigens are inherited from the parents, they are immunological complications of blood trans­
often useful in settling cases of disputed paternity. fusion include transmission of infectious
agents and circulatory overload. Infectious
Lewis blood group system: It differs from other
agents which may be transmitted during
blood group systems in that the antigens are
blood transfusion may be viruses, bacteria and
present primarily in the plasma and saliva and
protozoa (Table 23.2). Massive transfusion may
antigens do not form an integral part of the red
lead to circulatory overload.
cell membrane.
MN system: The antigens are M, N, S and s. Hemolytic disease of the newborn
This system has expanded to include at least 28
antigens. When an Rh– woman and an Rh+ man produce
a child, there is a 50% chance that the child will
Medical applications of blood be Rh+. If the child is Rh +, the Rh– mother can
become sensitized to this antigen during birth,
groups when the placental membranes tear and the fetal
1. Blood transfusion: Ideally, the donor and Rh+ RBCs enter the maternal circulation, causing
recipient should belong to the same ABO group. the mother’s body to produce anti-Rh antibodies
It used to be held that O group cells could be of the IgG type. During subsequent pregnancy,
transfused to recipients of any group as they Rh antibodies of the IgG class pass from the
Chapter 23: Immunohematology  | 161
Table 23.2  Nonimmunological complications of Detection of Rh-antibodies
blood transfusion Most Rh-antibodies are of the IgG class, and they
A. Transmission of Infectious Agents do not agglutinate Rh-positive cells in saline being
Viruses ‘incomplete antibodies’. IgG anti-D antibodies
•  Hepatitis B virus* may be detected by the following techniques:
•  Hepatitis C virus** 1. Using a colloid medium such as 20% bovine
•  Human immunodeficiency virus 1 and 2*
•  Human T-cell Iymphotrophic virus 1 and 2
serum albumin.
•  Cytomegalovirus 2. Using red cells treated with enzymes such as
Bacteria trypsin, pepsin, ficin or bromelin, and
•  Treponema pallidum* 3. By the indirect Coombs test: This is the most
•  Leptospira interrogans sensitive method.
•  Borrelia burgdorferi
Parasites
Plasmodium spp.* Prevention of Rh-isoimmunization
•  Babesia spp.
•  Trypanosoma cruzi HDNB is usually prevented today by passive immu­
•  Leishmania donovanii nization of the Rh– mother at the time of delivery
•  Toxoplasma gondii (within 24–48 hours) of any Rh+ infant with anti-
B. Circulatory Overload Rh-antibodies, which are available commercially
* Mandatory tests in India
(Rhogam). To be effective, this should be employed
** Mandatory tests in India since June 2001 from the first delivery onwards.

ABO Hemolytic Disease


mother to the fetus and damage its erythrocytes.
The fetal body responds to this immune attack The majority of cases (65%) of hemolytic disease
by producing large numbers of immature RBCs of the newborn have minor consequences and
called erythroblasts. Thus, the term erythroblastosis are caused by ABO blood-group incompatibility
fetalis was once used to describe what is now called between the mother and fetus. Maternofetal
hemolytic dis­ease of the newborn (HDNB). It is ABO incompatibility is very common and in a
an example of an antibody-mediated cytotoxicity proportion of these, hemolytic disease occurs in
disorder. the newborn. Natural antibodies are IgM in nature
in persons of blood group A or B, and so do not
Factors Influencing the Incidence of cross the placenta to harm the fetus. However, in
persons of blood group O, the isoantibodies are
Hemolytic Disease predominantly IgG in nature either through natural
The following factors influence the incidence of exposure or through expo­sure to fetal blood-group
hemolytic disease due to Rh-incompatibility: A or B antigens in successive pregnancies. Hence,
1. Immunolo gical unresp onsiveness to ABO hemolytic disease is seen largely in O group
th e Rh-antig en: Fo l l ow i ng a nt ig e n i c mothers, bearing A or B group fetus.
stimulation not every Rh-negative individual ABO hemolytic disease is much milder than Rh
forms Rh-antibodies. Some Rh-negative disease. The direct Coombs test is, often negative
individuals fail to do so even after repeated in this condition, while the indirect Coombs
injection of Rh-positive cells. They are called test (neonatal serum with type-specific adult
nonresponders. erythrocytes) is more commonly positive. Peripheral
2. Fetomaternal ABO incompatibility: Rh blood smear characteristically shows spherocytosis.
immunization is more likely to result when the
mother and fetus possess the same ABO group. Blood Group and Diseases
Rh sensitisation in the mother is rare when Rh It has been shown that some diseases may
and ABO incompatibility coexist. influence blood group antigens. Blood group
3. Number of pregnancies: The risk to the infant antigens have been reported to become weak in
increases with each successive pregnancy. The leukemia.
first child usually escapes disease because Red cell suspensions contaminated with certain
sensitisation occurs only during its delivery. bacteria, such as Pseudomonas aeruginosa, become
4. Zygosity of the father: An individual may be agglutinable by all blood group sera and even by
homozygous or heterozygous with respect to normal human sera. This phenomenon is known
D antigen. When the father. is homozygous as the Thomsen-Friedenreich phenomenon.
all his children will be Rh-positive. When It is due to the unmasking of a hidden antigen
he is heterozygous, half his children will be normally present on all human erythrocytes called
Rh-positive. the T-antigen.

www.ebook3000.com
162  |  Section 2: Immunology
It has been shown that duodenal ulcer is more Multiple Choice Questions (Mcqs)
frequent in persons of blood group O than in
1. The ABO blood group system was described by:
others. An association has also been established
a. William Harvey
between group A and cancer of the stomach.
b. Ehrlich and Morgenroth
Key Points c. Philip Levine
™™ Immunohematology is the study of blood group d. Karl Landsteiner
antigens and anti­bodies and their interactions in 2. All the following statements are true for H antigen
health and disease except:
™™ ABO blood group system: The ABO blood group a. It is a glycoprotein
substances are glycopeptides
b. It is structurally an L-sucrose
™™ Rh antigens: There are six common types of Rh
antigens designated as C, D, E, c, d, and e. Of all the c. It is a precursor for the production of A and B
Rh antigens, antigen D (Rho) is most important antigens
™™ Transmission of infectious agents especially HIV I and d. It is present on red blood cells of all ABO groups
II, HBV, and HCV through blood is the most important 3. Hemolytic disease in newborn may occur when:
complication a. An Rh negative mother carries an Rh positive
™™ Hemolytic disease of the newborn. Rh antibodies fetus
of the IgG class pass from the mother to the fetus
through the placenta and damage its erythrocytes. b. An Rh positive mother carries an Rh negative
This disease usually occurs even in the second or fetus
successive child. c. Both of the above
d. All of the above
Important questions 4. All the following infectious agents may be trans­
mitted by blood transfusion except:
1. Name various blood group systems and discuss a. Human immunodeficiency viruses I and II
Rh blood group system. Add a note on hazards of b. Hepatitis A virus
incompatible blood transfusion.
c. Hepatitis B virus
2. Write short notes on: d. Hepatitis C virus
a. Complications of blood transfusion.
b. Hemolytic disease of the newborn (or) erythro­ Answers (MCQs)
blastosis fetalis. 1. d; 2. b; 3. a; 4. b
Section 3: Systemic Bacteriology

24
Chapter

Staphylococcus

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe staphylococcal diseases
be able to: ∙∙ Discuss laboratory diagnosis of infections caused
∙∙ Describe species of Staphylococcus by Staphylococcus aureus
∙∙ Describe morphology and culture characteristics ∙∙ Explain methicillin-resistant staphylococci and its
of Staphylococcus aureus clinical problem
∙∙ List characteristics of Staph. aureus strains ∙∙ D escribe the following: Coagulase-negative
∙∙ Explain coagulase test staphylococci (CNS); micrococci
∙∙ List and describe toxins and enzymes of Staphylo­ ∙∙ Distinguish characteristics of Staph. aureus, Staph.
coccus aureus epidemidis and Staph. saprophyticus.

Introduction Staphylococcus aureus


Staphylococci are gram-positive cocci that occur Morphology
in grape-like clusters. Staphylococci were first They are spherical cocci, approximately 1 μm in
observed in human pyo­g enic lesions by von diameter, arranged characteristically in grape-like
Recklinghausen in 1871. It was Sir Alexander Ogston, clusters (Fig. 24.1). Cluster formation is due to cell
a Scottish surgeon, who established conclusively the division occurring in three planes, with daughter
causative role of the coccus in abscesses and other cells tending to remain in close proximity. They
suppurative lesions (1880). He also gave it the name may also be found singly, in pairs and in short
Staphylococcus (Staphyle, in Greek, meaning ‘bunch chains of three or four cells, especially when
of grapes’: kokkos, meaning a berry) due to the typical examined from liquid culture. Long chains never
occurrence of the cocci “in grape-like clusters in pus occur.
and in cultures.
They are nonsporing, nonmotile and usually
noncapsulate with the exception of rare strains.
Species
The genus Staphylococcus contains 33 defined
species and 20 species found in man. Species
of staphylococci are initially differentiated by
the coagulase test and are classified into two
groups: coagulase-positive and coagulase-negative
staphylococci.
Coagulase-positive staphylococci: Staphylococcus
aureus (formerly also called Staph. pyogenes) is
coagulase ­positive.
Coagulase-negative staphylo co cci (CNS):
S. epidermidis and S. saprophyticus are the most
clinically significant species in this group. Fig. 24.1: Staphylococcus in a smear of pus

www.ebook3000.com
Chap-24.indd 163 15-03-2016 11:33:06 AM
164  |  Section 3: Systemic Bacteriology
They stain readily with aniline dyes and are food and dust, likely to contain a predominance
uniformly gram-positive. of other kinds of bacteria. Therefore, 7–10% of
sodium chloride may be added to nutr­ient agar
Cultural Characteristics (Salt agar) or milk agar (salt milk agar); mannitol
They are aerobes and facultative anaerobes. Opti­ salt agar containing 1% mannitol, 7.5% NaCl,
mum temperature for growth is 37°C ( range being and phenol red in nutrient agar; and Ludlam’s
12–44°C). Optimum pH is 7.5. They can grow medium containing lithium chloride and tellurite;
readily on ordinary media. and salt cooked meat broth (10% NaCl.).
1. Nutrient agar: After aerobic incubation for 24
hours at 37°C, colonies are 1–3 mm in diameter Biochemical Reactions
and have a smooth glistening surface, an 1. Sugar fermentation: S. aureus ferments a range
entire edge, a soft butyrous consistency and an of sugars producing acid but no gas. Sugar
opaque, pigmented appearance. Most strains fermentation is of no diagnostic value except
produce golden-yellow (aureus) pigment. These for mannitol, which is usually fermented
white-colonied strains of S. aureus are fully anaerobically by Staph. aureus but not by other
virulent. Pigmentation is characteristic of this species.
species when grown aerobically. 2. Catalase: Catalase positive (un­like streptococci).
Pigmentation is enhanced on fatty media, such 3. Lipolytic: When grown on media containing
as tween agar, by prolonged incuba­tion, and by egg-yolk, produce a dense opacity because
leaving plates at room temperature. The pigment most strains are lipolytic.
is believed to be lipoprotein allied to carotene. 4. Phosphatase test: This is a useful screening
2. Blood agar: The colonies have the same procedure for dif­ferentiating Staph. aureus
appearances as on nutrient agar, but may from Staph. epidermidis in mixed cultures, as
be surrounded by a zone of β-hemolysis the former gives prompt phos­phatase reaction,
(Fig. 24.2). Hemolysis is more likely to be while the latter is usually negative or only
present if sheep, human or rabbit blood is used. weakly positive. All strains of S. aureus produce
3. MacConkey agar: Colonies are smaller and are phosphatase which liber­ates phenolphthalein
pink due to lactose fermentation. from sodium phenolph­thalein diphosphate
4. Milk agar: On this medium, after overnight The culture plate, with the culture, is inverted
incubation, the colonies of S. aureus are larger over the ammonia for a minute or so. Colonies
than those on nutrient agar and pigmentation of S. aureus become bright pink because
is well-developed and easily recognized against phenolphthalein is pink in alkaline pH. Most
the opaque white background. other staphy­lococci form colonies that remain
5. Phenolphthalein phosphate agar: This is an uncolored.
indicator medium and assists in the iden­ 5. Deoxyribonulease (DNAase) test: It produces a
tification of S. aureus in mixed cultures. deoxyribonulease (DNAase), and a heat-stable
Appearance of the colonies of S. aureus on this nuclease (thermonuclease, TNAase).
medium is similar to those on nutrient agar. 6. Other biochemical tests: Indole negative, MR
Colonies of S. aureus become bright pink when positive, VP positive, urease positive, hydrolyzes
culture plate is inverted over the ammonia for gelatin and reduces nitrates to nitrites.
a minute or so. Table 24.1 shows the characteristics of
6. Selective salt media: Selective medium may Staphylococcus aureus.
be useful for the isolation and enumeration of
staphylococci from materials, such as feces,
Table 24.1  Characteristics of Staph. aureus strains
Staph. aureus strains usually exhibit the following
characteristics:
1. Beta hemolysis—produce clear hemolysis on blood
agar
2. Golden yellow pigment
3. Coagulase positive
4. Greater biochemical activity, ferment mannite
5. Liquefy gelatin
6. Produce phosphatase
7. Black colonies on potassium tellurite blood agar—
produce black colonies in a medium containing
potassium tellurite by reducing tellurite to metallic
tellurium
8. Produce thermostable nucleases which can be
demonstrated by the ability of boiled cultures to
Fig. 24.2: Structure of staphylococcal cell wall degrade DNA in an agar diffusion test

Chap-24.indd 164 15-03-2016 11:33:07 AM


Chapter 24: Staphylococcus  | 165
Resistance to staphylococcal cells. This is known as
coagglutination and has led to numerous
S. aureus and the other Micrococcaceae are among
applications (Chapter 16).
the hardiest of the nonsporing bacteria. Dried on
threads, they retain their viability for 3–6 months. 2. Cytoplasmic membrane: It serves as an osmotic
They have been isolated from dried pus after 2–3 barrier for the cell and provides an anchorage
months. It withstands moist heat at 60°C for 30 for the cellular biosynthetic and respiratory
min but is killed after 60 min. Most strains grow enzymes.
in the presence of 10% NaCl. These fea­tures are of 3. Clumping factor (bound coagulase): The
significance in food preservation. component on the cell wall of S. aureus that
It is readily killed by phenolic and hypochlorite results in the clumping of whole staphylococci
disinfectants, and by antiseptic preparations such in the presence of plasma is referred to as the
as hexachlorophane, chlorhexidine and povidone- clumping factor (also called bound coagulase).
iodine. They are very sensitive to aniline dyes; This factor reacts directly with fibrinogen in
thus they are inhibited on blood agar medium plasma, converts it to insoluble fibrin, causing
containing 1 in 500,000 crystal violet, which the staphylococci to clump or aggregate.
permits the growth of streptococci. Staphylococci Slide coagulase test: Since this factor is detected
are uniformly resistant to lysozyme but some by perform­ing the test on a slide, therefore, the
micrococci are sensitive to it. Staphylococci are test is known as slide coagulase test.
generally sensitive to lysostaphin—a mixture of
enzymes produced by a particular strain of Staph. Toxins and Enzymes
epidermidis. S. aureus produces a number of toxins and
enzymes. They are the virulence factors.
Antigenic Structure of
Staphylococcus aureus A. Toxins
The antigenic structure of S. aureus (Fig. 24.2) is 1. Cytolytic Toxins
complex. It is as follows. At least five cytolytic or membrane-dam­aging toxins
(alpha, beta, delta, gamma, and Panton-Valentine
A. Cell-associated Polymers [P-V] leukocidin are produced by S. aureus.
1. Capsule: Capsular polysaccharide surrounding i. Alpha (a) hemolysin: Alpha (a) lysin is the
the cell wall inhibits opsonization. most important among them. It is a protein
2. Peptidoglycan: The cell wall polysaccharide and is inactivated at 60°C; however, its activity
peptidoglycan confers rigidity and structural is reg­ained paradoxically, if it is further heated
integrity to the bacterial cell. It activates to bet­ween 80° and 100°C. This is due to the
complement and induces release of inflamm­ fact that the toxin combines with a heat-labile
atory cytokines. inhibitor at 60°C but at higher temperature
3. Teichoic acids: Teichoic acid, an antigenic (80–100°C) the inhibitor is inactivated thus
component of the cell wall, facilitates adhesion toxin regains its activity. Above 100°C toxin
of the cocci to the host cell surface and itself gets inactivated.
protects them from complement-mediated The toxin lyses rabbit and sheep erythro­
opsonization. cytes. It is leucocidal, dermonecrotic and
lethal.
B. Cell Surface Proteins ii. Beta (b) hemolysin: Beta (b) toxin, also called
1. Protein A: Protein A is a group-specific antigen sphingomyelinase C, is a heat-labile protein
unique to S. aureus strains. produced by most strains of S. aureus. b lysin
Biologic properties: Protein A has many is strongly active on sheep and weakly active
biologic properties including chemotactic, on rabbit and human red blood cells. It does
anticomplementary, and antiphagocytic and not lyse horse red blood cells. It exhibits a
elic­its hypersensitivity reactions and platelet ‘hot-cold phenomenon’, the hemolysis being
injury. It is mitogenic and potentiates natural initiated at 37°C, but becoming evident only
killer activity of human lymphocytes. after chilling.
Protein A binds IgG molecules, nonspecifically, It is not dermonecrotic and kills experi­
through Fc region leaving specific Fab sites mental animals only when injected in large
free to combine with specific antigen. When doses.
suspension of such sensitized cells is treated iii. Gamma (γ) hemolysin: γ lysin acts on sheep,
with homologous (test) antigen, the antigen rabbit and human red blood cells but not on
combines with free Fab sites of IgG attached those of horse.

www.ebook3000.com
Chap-24.indd 165 15-03-2016 11:33:07 AM
166  |  Section 3: Systemic Bacteriology
iv. Delta (d) hemolysin: It lyses red blood cells of syndrome’ (SSSS). Two distinct forms of exfoliative
sheep, rabbit, horse and man. toxin (ETA and ETB) have been identified, and
v. Leukocidin: Leukocidin (called the Panton- either can produce disease. ETA is heat-stable and
Valentine toxin af­ter its discoverers) is also a the gene is chromosomal, whereas ETB is heat-
two component toxin, like the gamma lysin, labile and plasmid-mediated. SSSS is seen mostly
S (slow) and F (Fast). These components act in young children and only rarely in older children
synergistically to damage polymor­phonuclear and adults.
leukocytes and macrophages and to pro­duce
dermonecrosis. B. Extracellular Enzymes
2. Enterotoxins Staph. aureus produces a number of enzymes such
This toxin is responsible for the manifestations as coagulase catalase, hyaluronidase, fibrinolysin,
of staphylococcal food poisoning n ­ ausea, lipases, nucleases and penicillinase.
vomiting and diarrhea 2–6 hours after consuming Coagulase: S. aureus produces an extracel­lular
contaminated food containing preformed toxin. enzyme called coagulase which brings about
Enterotoxins are commonly produced by about clotting of human or rabbit plasma. It acts along
two-thirds of Staph. aureus strains, growing in with a ‘coagulase reacting factor’ (CRF) present
carbohydrate and protein foods. The toxin is in plasma, binding to prothrombin and converting
relatively heat stable, resisting 100°C for 10–40 fibrinogen to fibrin. Coag­ulase does not clot plasma
minutes depending on the concentration of the of guinea pigs because they lack CRF. Coagulase
toxin and nature of the medium. test is the standard criterion for the identification
Eight serologically distinct staphylococcal of Staph. aureus isolates.
enterotox­ins (A-E, G-I) and three subtypes of Eight antigenic types (A-H) have been des­
enterotoxin C have been identified. Enterotoxin cribed. Most human strains form coagulase type A.
A is most commonly associated with disease. Coagulase Fibrin (Clotting)
Fibrinogen →
Enterotoxins C and D are found in contaminated CRF
milk products, and enterotoxin B causes staphylo­ Coagulase test: Coagulase test is done by two
coccal pseudomembran­ous enterocolitis. methods—slide and tube coagulase test. The slide
The precise mechanism of toxin activity is not or tube coagulase test is performed to distinguish
understood. The toxin is believed to act directly on Staph. aureus from coagulase-negative species.
the autonomic nervous system to cause the illness, a. Slide coagulase test: The slide test detecting
rather than on the gastrointestinal mucosa. The bound coagulase is much simpler. It can be
toxin is antigenic and neutralized by the specific detected by emulsifying a few colonies of the
antitoxin. Type A toxin is responsible for most cases. bacteria in a drop of normal saline on a clean
For detection of the toxin sensitive serological tests, glass slide and mixing it with a drop of rabbit
such as latex agglutination and ELISA are available. plasma. Prompt clumping of the organisms
These toxins are super­a ntigens, however, indicates the presence of clumping factor
capable of inducing nonspecific activation of T (bound coagulase). Positive and negative
cells and cytokine release. The toxin is potent, controls also are set up.
microgram amounts being ca­pable of causing the Almost all the clumping factor producing
illness. strains of S. aureus produce coagulase.
b. Tube coagulase test: The tube coagulase test
3. Toxic Shock Syndrome Toxin-I (TSTT-1) detects free coagulase. 0.1 ml of an overnight
S. aureus is associated with toxic shock syndrome broth culture or broth suspension from an agar
(TSS), a severe and often fatal disorder characterized plate culture made up to the same density is
by multiple organ dysfunction. TSST-I (formerly mixed with 0.5 mL of a 1 in 10 dilution of human
called pyrogenic exotoxin C and enterotoxin F) or rabbit plasma. The mixture is incubated in
is heat and proteolysis resistant, chromosomally a water bath at 37°C for three to six hours. If
mediated exotoxin. It is antigenic and most persons positive, the plasma clots and does not flow when
over 30 years of age have circulating antibodies. the tube is inverted. If clot does not appear it is left
The enterotoxins and TSST-1 belong to a class overnight at room temperature and re-examined.
of polypeptides known as superantigens which On continued incubation, the clot may be
are potent activators of T lymphocytes leading to lysed by fibrinoly­sin produced by some strains.
the release of cytokines such as interleukins and Controls with plasma alone, known coagulase-
tumour necrosis factor. This results in clinical positive and coagulase-negative cultures must
condition of TSS. be set up with each batch of tests.
4. Epidermolytic Toxins (Exfoliative Toxins) False-pos­itive reaction: Citrated plasma should not
This toxin, also known as ET or ‘exfoliatin’ is be used because contaminating gram-negative
responsible for the ‘staphylococcal scalded skin bacilli (e.g. Pseudomonas) may utilize the citrate

Chap-24.indd 166 15-03-2016 11:33:07 AM


Chapter 24: Staphylococcus  | 167
and produce false-positive reaction. Oxalate, EDTA nonmenstruating women, children, and
or heparin are suitable anticoagulants. men with boils or staphylococcal infections
of wounds can also have TSS. Though
Epidemiology tampon-related TSS is now rare, the
Staphylococci are ubiquitous. Staphylococcus is a syndrome occurs in other infections of the
normal component of man’s indigenous microflora skin, mucosa and other sites and also in
and is carried asymptomati­cally in a number of some surgical wounds.
body sites. About 10–30% healthy persons carry iii. Exfoliative diseases: These lesions are pro­
staphylococci in the nose and about 10% in the duced by the strains of S. aureus which
perineum and also on the hair. Vaginal carriage is produce epidermolytic toxins. This toxin
about 5–10%, which rises greatly during menses, a is responsible for the ‘staphylococcal
factor relevant in the pathogenesis of TSS related scalded skin syndrome’ (SSSS), exfoliative
to menstruation. Cross-infection is an important skin diseases in which the outer layer
method of spread of staphylococcal disease, of epidermis gets separated from the
particularly in hospi­t als. Staphylococci are a underlying tissues. SSSS is seen mostly in
common cause of postopera­tive wound infection young children and only rarely in older
and other hospital infections. Most of these are children and adults. The severe form of
due to certain strains of staphylococci the so-called SSSS is known as Ritter’s disease in the
‘hospital strains’ that are present in the hospital newborn and toxic epidermal necrolysis in
environ­ment. They belong to a limited number older patients. Milder forms are pemphigus
of phage types and are commonly resistant to neonatorum and bullous impetigo. Bullous
penicillin and other antibiotics routinely used in impetigo is a localized form of SSSS.
hospitals. Epidemics of hospital cross infections
are caused by some of them, the ‘epidemic strains’. Bacteriophage Typing
Phage type 80/81 was the first to be recognized Staphylococci may be typed, based on their
for most of staphylococcal infections in hospitals suscepti­bility to bacteriophages. An internationally
throughout the world. recogni­zed set of 23 standard typing phages is used
for epidemiological studies and tracing the source
Staphylococcal Diseases of infection. (Table 24.2).
Staphylococcal infections are among the most The strain to be typed is inoculated on a plate of
common of bacterial infections and range from nutrient agar to form a lawn culture. After drying.
the trivial to the fatal. Staphylococcal infections the phages are applied over marked squares in
are characteristically localized pyogenic lesions, a fixed dose (routine test dose). After overnight
in contrast to the spreading nature of streptococcal incubation, the culture will be observed to be lysed
infections. S. aureus causes disease through the by some phages but not by others (Fig. 24.3). The
direct invasion and destruction of tissue or through phage type of a strain is expressed by designation
the production of toxin. of the phages that lyse it, and there is interna­
tional agreement on the interpretation of results.
A. Cutaneous infections: These include wound
Thus, if a strain is lysed only by phages 3C, 55
and burn infection, pustules, furuncles or boils
and 71, it is called phage type 3C/55/71. Phage
carbuncles, styes, impetigo and pemphigus
typing is important in epidemiological studies of
neonatorum.
staphylococcal infections.
B. Deep infections: These include osteomyelitis,
The reference center for staphylococcal phage
periostitis, tonsillitis, pharyngitis, sinusitis,
typing in India is located in the Department of
broncho­pneumonia, empyema, septicemia,
meningitis, endocarditis, breast abscess, renal
abscess and abscesses in other organs. Table 24.2  International basic set of phages for
C. Toxin-mediated diseases typing Staph. aureus of human origin
i. Food poisoning: Staphylococcal food
Lytic group
poisoning may follow 2–6 hours after the Designation of phages
of phages
ingestion of food in which S. aureus has
I 29 52 52A 79 80
multiplied and formed enterotoxin.
II 3A 3C 55 71 95
ii. Toxic shock syndrome (TSS): Toxin-
producing strains of S. aureus have been III
{ 6 42E 47 53 54
implicated in most cases of TSS, a multi­ 75 77 83A 84 85
system disease that primarily afflicts young V 94 96
women. Most cases occur in menstruating Unclassified 81
women who use tampons. However,

www.ebook3000.com
Chap-24.indd 167 15-03-2016 11:33:07 AM
168  |  Section 3: Systemic Bacteriology
performed appropriate to the clinical situation.
This is important as staphylo­c occi readily
develop resistance to drugs.
6. Bacteriophage typing: Bacteriophage typing
may be done if the informa­t ion is desired
for epidemiological purposes. Other typing
methods include antibiogram pattern, plasmid
profile, DNA fingerprinting, ribotyping and
PCR-based analysis for genetic pleomorphism.
7. Serological tests : Serological tests may
sometimes be of help in the diagnosis of
hidden deep infections. Antistaphy­lolysin
(anti-alphalysin) titers of more than two units
per mL, especially when the titer is rising, may
be of value in the diagnosis of deep seated
Fig. 24.3: Bacteriophage typing of staphylococci
infections, such as bone abscesses.
Micro­biology, Maulana Azad Medical College,
New Delhi. Treatment
Benzyl penicillin is the most effective antibiotic,
Laboratory Diagnosis if the strain is sensitive but most clinical isolates
1. Specimens: The specimens to be collected of S.aureus are resistant to benzyl penicillin due to
depend on the type of lesion, for example: production of beta lactamase. Cloxacillin, oxacillin,
pus from suppurative lesions; sputum from flucloxacillin and methicillin are penicillinase-
respiratory infec­t ions; food remains and resistant penicillins.
vomit from cases of food poisoning; nasal and Methicillin-resistant Staphylococcus aureus
perineal swabs from suspected carriers. (MRSA) are also resistant to other penicillins
Swabs of the perineum, pieces of hair and and cephalosporins. Glycopeptides (vancomycin
umbilical stump—may be necessary in special or teicoplanin) are the agents of choice in the
situa­tions. treatment of systemic infec­tion, but these agents
2. Direct microscopy: Direct microscopy with are expensive and may be toxic.
Gram stained smears is useful in the case of For mild superficial lesions, topical appli­cations
pus, where cocci in clusters may be seen. This of drugs as bacitracin, chlorhexidine or mupirocin
is of no value for specimens like sputum where may be sufficient.
mixed bacterial flora are normally present. The treatment of carriers is by local application
3. Culture: The specimens are cultured on a blood of antibiotics such as bacitracin and antiseptics
agar plate. Staphylococcal colonies appear such as chlorhexidine. In resistant cases, rifampicin
after overnight incubation specimens, where along with another oral antibiotic may be effective.
staphylococci are expected to be outnumbered
by other bacteria (e.g. wound swab and Control
feces), are inoculated on selective media like 1. The focus of infection (e.g. abscess) must be
Ludlam’s or salt-milk agar or Robertson’s cooked identified and drained.
meat medium containing 10% sodium. The 2. Treatment is symptomatic for patients with
inoculated media is incubated at 37°C for 18–24 food poisoning.
hours. On blood agar plate, look for hemolysis 3. Proper cleansing of wounds and use of
around the colonies. The plates are inspected disinfectant.
for golden-yellow or white colonies. Smears are 4. Thorough hand washing and covering of
examined from the culture and coagulase test exposed skin helps medical personnel prevent
done when stahylococci are isolated. infection or spread to other patients.
4. Identification: Relatively simple biochemical
5. Asymptomatic nasopharyngeal carriage—
tests (e.g. positive reactions for coagulase
the use of chemoprophylaxis consist­ing of
(clumping factor), heat-stable nuclease, alkaline
vancomycin and rifampin to prevent spread of
phosphatase, and mannitol fermentation) can
oxacillin-resistant organisms
be used to differentiate S. aureus and the other
staphylococci. Other Coagulate-positive
Coagulase test: Coagulase test is done by two
staphylococci
methods-slide and tube coagulase test.
5. Antibiotic sensitivity tests: As a guide to Other staphylocoagulase producing (coagulase
treatment antibiotic sensitivity tests should be ­p ositive) staphylococci are S. intermedius,

Chap-24.indd 168 15-03-2016 11:33:08 AM


Chapter 24: Staphylococcus  | 169
Table 24.3  Characteristics distinguishing three species of the genus Staphylococcus
Character S. aureus S. epidermis S. saprophyticus
Anaerobic growth and fermentation of glucose + + –
Mannitol
Acid aerobically + V V
Acid anaerobically + – –
Coagulase + – –
DNAase + – –
Phosphatase + –/weak+ –
α-Toxin + – –
Protein A in cell wall + – –
Novobiocin sensitivity Sensitive Sensitive Resistant
V, variable

S. delphini, S. lutrae, and some strains of S. hyicus. to novobiocin and by its failure to ferment glucose
These are often animal-associated species and are anaerobically. It is nonhemolytic and does not
infrequently iso­lated from human samples. contain protein A. Table 24.3 lists the features
useful for distinguishing the major species of
Coagulase-negative staphylococci.
Staphylococci
Other Coagulase-negative Staphylococci
Coagulase-negative staphylococci (CONS) are
commonly found on the surface of healthy persons. S. haemolyticus has been reported in wounds,
They are opportunistic pathogens that cause bacteremia, endocarditis, and UTIs. Other species
infection in debilitated or compro­mised patients. include S. lugdunensis, S. warneri, S. capitis,
Various species are Staph. epidermidis, Staph. S. simulans, and S. schleiferi.
saprophyticus, Staph. hemolyticus, Staph. hominis,
and Staph. capitis. Sensitivity to Antibiotics
Before the introduction of penicillin, most of the
Staphylococcus epidermidis strains of S. aureus were sensitive to this antibiotic.
Staph. epidermidis is invariably present on normal Staphylococci quickly developed drug resistance
hu­man skin. It is nonpathogenic ordinarily but after penicillin was introduced.
can cause disease when the host defences are Penicillin resistance is of three types:
breached. S. epidermidis has a distinct predilection 1. Production of beta lactamase (penicillinase):
for foreign bodies, such as artificial heart valves, Production of beta lactamase (penicillinase)
indwelling intravascular catheters, central nervous which inactivates penicillin by splitting the beta
system shunts, and hip prostheses. Their etiological lactam ring. Staphylococci produce four types
role is proved by repeated isolation. of penicillinases, A to D. Penicillinase is an
inducible en­zyme and its production is usually
controlled by plasmids which are transmitted
Clinical Infection
by transduction or conjugation.
∙∙ Stitch abscesses; endocarditis of native Penicillinase plasmids are transmitted to
and prosth­etic valves; intravenous catheter the sen­sitive staphylococci by transduction and
infections; CSF shunt infections; bacteremia; also possibly by conjugation.
osteomyelitis; wound infections, vascular graft 2. Changes in bacterial surface receptors:
infections, prosthetic joint infection, etc. Changes in bacterial surface receptors, reduc­
ing binding of beta-lactam antibiotics to cells.
Staphylococcus saprophyticus This change is normally chromosomal in nature
S. saprophyticus is a common cause of urinary tract and is expressed more at 30°C than at 37°C. This
infections in sexually active young women. It may resistance also extends to cover beta lactamase-
also cause ure­thritis in men and women, catheter- resistant penicil­lins, such as methicillin and
associated uri­nary tract infections, prostatitis in cloxacillins. Some of these strains may show
elderly men, and rarely bacteremia, sepsis and resistance to other antibiotics and heavy metals
endocarditis. also and cause outbreaks of hospital infection.
This coagulase-negative Staphylococcus can be These strains have been called ‘epidemic
distinguished from S. epidermidis by its resistance methicillin-resistant Staphylococcus aureus’

www.ebook3000.com
Chap-24.indd 169 15-03-2016 11:33:08 AM
170  |  Section 3: Systemic Bacteriology
Table 24.4  Differentiation between staphylococci and micrococci
Property Staphylococcus Micrococcus
Gram staining Gram-positive Gram-positive, darkly stained
Grape-like clusters In groups of four (tetrad) or eight
Uniform staining Often staining is not uniform
Colony characters Colonies are golden Colonies are white in color generally
yellow Size 1 mm Size Larger than staphylococcus
Anaerobic acid production from glucose + –
Aerobic acid production form glycerol + –
in the presence of erythromycin
Modified oxidase – +
Bacitracin sensitivity (0.04 unit disk) Resistant Sensitive
Lysostaphin sensitivity Sensitive Resistance
Furazolidone susceptibility (100 µg) of furazolidone disk Sensitive Resistance

or EMR­M. (as methicillin is an unstable drug, samples. A high salt concen­tration (5.5% NaCl)
cloxacillin is used for sensitivity testing instead). and polymyxin B make the medium selective
3. Development of tolerance: Development of for staphylococci.
tolerance to penicillin, by which the bacterium 2. The gold standard for MRSA detection is the
is only inhibited but not killed. detection of the mecA gene by using nucleic
acid probes or poly­merase chain reaction (PCR)
Methicillin-resistant Staphylococcus amplification.
aureus (MRSA) Control of MRSA: Control of MRSA requires strict
Methicillin was the first compound deve­loped adherence to infection-control practices such as
to combat resistance due to penicillinase (beta barrier.
lactamase) production by staphylococci. Due
to the limitations in clinical use of methicillin, Micrococci
cloxacillins are used instead against penicillinase-
producing strains. But methicillin resistant strains Micrococci are catalase-positive, gram-positive,
of Staph. aureus (MRSA) became common, which coagulase-negative and usually oxidase-positive.
were resist­ant not merely to penicillin, but also They may ocassionally colonize the skin or mucous
to all other beta lactam antibiotics and many membrane of humans, but they are only rarely
others besides. Isolates that are resistant have associated with infections.
been traditionally termed methicillin-resistant Only two species, Micrococcus luteus and
staphylococci, with S. aureus being called MRSA Micrococcus lylae, remain in the genus. Micrococci,
and S. epidermidis referred to as MRSE. When especially M. luteus, have a tendency to produce
any staphylococcus isolated is identified as being a yellow pigmented colony. Nine species of genus
resistant to meth­icillin, this implies that it is Micrococcus have been described.
also resistant to nafcillin and oxacillin and to all Table 24.4 gives some differentiating features
β-lactam antibiotics, including the cepha­losporins. of Staphylococcus and Micrococcus.
MRSA is also becoming more common in the
community, especially in long-stay insti­tutions. Key Points
Staphylococcus
Treatment-glycopeptides (vancomycin or
™™ Staphylococcus is gram-positive cocci arranged in
teicoplanin) are the agents of choice in the
clusters
treatment of systemic infec­tion, but these agents
™™ Species of staphylococci are classified by the
are expensive and may be toxic. Concerns with coagulase test into two groups: the coagulase-
rising resistance to glycopep­t ides call for the positive (Staphylococcus aureus) and coagulase-
restrictive use of these drugs. negative staphylococci. S. epidermidis and S.
saprophyticus
Laboratory Diagnosis Staphylococcus aureus
™™ Staph. aureus strains usually exhibit following
1. For laboratory purposes, oxacillin is generally characteristics: (1) Beta hemolysis; (2) Golden
used for detection of methicillin resistance. yellow pigment; (3) Coagulase positive; (4) Greater
The use of an oxacillin-salt agar plate, such as biochemical activity, ferment mannite; (5) Liquefy
the oxacillin resistance screening agar can be gelatin; (6) Produce phosphatase; (7) Black colonies
used as a screening test for MRSA in clinical on potassium tellurite blood

Chap-24.indd 170 15-03-2016 11:33:08 AM


Chapter 24: Staphylococcus  | 171

™™ S. aureus produces many virulence factors including: f. Epidermolytic toxins of Staphyloco­ccus aureus
(1) Cytolytic or membrane-damaging toxins (alpha, g. Drug resistance in staphylococci.
beta, delta, gamma, and Panton-Valentine [P-V] h. Methicillin-resistant Staphylococcus aureus
leukocidin); (2) Exfoliative toxins: (3) Enterotoxins (MRSA)
(A-E, G-I); (4) Toxic shock syndrome toxin-1 (TSST-1) i. Coagulase-negative staphylococci (CNS)
™™ Diseases: S. aureus causes cutaneous infections j. Micrococci.
such as folliculitis, boils, carbuncles, impetigo, and
purulent abscesses. These cutaneous infections
can progress to deeper abscesses involving other
Multiple choice questions (MCQs)
organ systems and progress to septicemia and 1. Staphylococcus aureus shows the following
bacteremia. Toxin-induced diseases, such as food characters except
poisoning, scalded skin syndrome (SSS), and toxic
a. It produces golden brown pigment on the blood
shock syndrome (TSS), are also associated with this
agar
organism
Other systemic diseases (frequently associated with b. It ferments mannitol
bacteremia) include pneumonia, empyema, septic c. It is novobiocin-sensitive
arthritis, osteomyelitis, acute endocarditis, and d. It has protein A
catheter-related bacteremia 2. Protein A is a cell wall components of
™™ Diagnosis: It is done by microscopy, culture, antibiotic a. Staphylococcus aureus
sensitivity tests and serological tests b. Staphylococcus epidermidis
™™ Bacteriophage typing may be done if the information
c. All of the above
is desired for epidemiological purposes
™™ Treatment: The antibiotics of choice are oxacillin
d. None of the above
(or other penicillinase-resistant penicillin) or 3. The enzyme coagulase shows all the following
vancomycin for oxacillin-resistant strains features except
™™ Methicillin-resistant strains (MRSA). Glycopeptides a. It has eight serotypes
(vancomycin or teicoplanin) are the agents of choice b. It is extracellular
in the treatment of systemic infection c. It is detected by tube coagulase test
™™ Coagulase-negative staphylococci: Coagulase- d. Undiluted serum is used in the test
negative staphylococci are opportunistic pathogens
4. Scalded skin syndrome is caused by the
that cause infection in debilitated or compromised
following toxin of Staphylococcus aureus
patients. Staph. epidermidis accounts for about 75%
of all clinical isolates. Other species include Staph. a. Enterotoxins
haemolyticus, Staph. hominis, Staph. capitis and b. Toxin shock syndrome
Staph. saprophyticus c. Exfoliative toxin
™™ Staph. saprophyticus can be distinguished from S. d. Leukocidin
epidermidis by its resistance to novobiocin and by 5. Which of the of the following Staphylococcus is
its failure to ferment glucose anaerobically novobiocin resistant?
™™ Micrococci may occasionally colonize the skin or a. Staphylococcus aureus
mucous membrane of humans, but they are only
b. Staph. epidermidis
rarely associated with infections.
c. Staph. saprophyticus
d. None of the above
Important Questions 6. The most common cause of cystitis in a young
1. Describe the morphoplogy, cultural characteristics healthy sexually active women is:
and antigenic structure of Staphylococ­cus aureus. a. Staphylococcus aureus
2. Name various virulence factors of Staphylococcus b. Staphylococcus epidermidis
aureus. c. Staphylococcus saprophyticus
3. Classify staphylococci. Discuss pathogenicity and d. Staphylococcus saccharolyticus
laboratory diagnosis of Staph. aureus. 7. All are coagulase-negative staphylococci except:
4. Write short notes on: a. Staphylococcus epidermidis
a. Coagulase or staphylocoagulase b. Staphylococcus saprophyticus
b. Clumping factor c. Staphylococcus haemolyticus
c. Toxins and enzymes of produced by Staphylo­ d. Staphylococcus aureus
coccus aureus
d. Staphylococcal food poisoning. Answers (MCQs)
e. Toxic shock syndrome 1. a; 2. a; 3. d; 4. c; 5. c; 6. c; 7. d

www.ebook3000.com
Chap-24.indd 171 15-03-2016 11:33:08 AM
25
Chapter

Streptococcus and Enterococcus

Learning Objectives

After reading and studying this chapter, you should ∙∙ List and describe toxins and enzymes of Strepto­
be able to: coccus pyogenes
∙∙ Classify streptococci ∙∙ Describe non-suppurative complications of Str.
∙∙ Describe antigenic structure of Str. pyogenes pyogenes infections
∙∙ List and describe toxins and enzymes of Strepto­ ∙∙ Discuss laboratory diagnosis of streptococcal
coccus pyogenes infections
∙∙ Discuss pathogenicity of streptococci ∙∙ Discuss group B streptococci, Group D streptococci
and viridans group.

Introduction Classification
The genus Streptococcus comprises a large and Streptococci are first divided into obligate anaerobe
biologically diverse group of gram-positive cocci and facultative anaerobes. Obligate anaerobe are
that grow in pairs or chains (Fig. 25.1). They are designated as peptostreptococci.
normal flora of humans and animals. They inhabit Three different schemes are used to classify the
various sites, notably the upper respiratory tract, organism, as shown in Figure 25.2.
and live harmlessly as commensels.
Streptococci were first described by Billroth
(1874) in exudates from erysipelas and wound
infections, who called them streptococci (streptos,
meaning twisted or coiled; coccus, a grain or
berry). Pasteur (1879) found similar organisms
in the blood of a patient with puerperal sepsis.
Ogston (1881) isolated them in acute abscesses,
distinguished them staphylococci and by animal
inoculation established their pathogenicity.

Fig. 25.1: Streptococci Fig. 25.2: Classification of streptococci


Chapter 25: Streptococcus and Enterococcus  | 173
1. Hemolytic Activity Biolochemical (Physiologic) Properties
The aerobic and facultative anaerobic streptococci Biochemical and other criteria are also used in
are classified on the basis of their hemolytic defining various species within a single serogroup,
properties. The type of hemolytic reaction and some species contain strains of more than one
displayed on blood agar has long been used to serogroup.
classify the streptococci. Brown (1919) categorized Table 25.1 shows characteristics and clinical
them into three varieties based on their growth in significance of important streptococci and
5% horse blood agar pour plate culture. enterococci.

A. Alpha-hemolytic (α) Streptococci Streptococcus pyogenes


They produce a zone of partial hemolysis with a Morphology
greenish discoloration around the colonies on S. pyogenes are gram-positive, spherical to ovoid
blood agar. The zone of lysis is small (1–2 mm wide) organisms 0.5–1.0 mm in diameter. The organism
with indefinite margin, with in this zone unlysed grows in short or moderately long chains, the chain
erythrocytes can be made out microscopically. length being dependent on the strain and culture
The streptococci producing a-hemolysis are also medium. Chain formation is due to the cocci
known as viridans streptococci. dividing in one plane only and the daughter cells
failing to separate completely. Chains are longer in
B. Beta (β) Hemolytic Streptococci liquid media than in solid media.
They produce a sharply defined, clear, colorless Streptococci are nonmotile and nonsporing.
zone of hemolysis (2–4 mm wide) around the Some strains of S. pyogenes and some group C
colony, caused by complete lysis of red blood strains produce a capsule of hyaluronic acid,
cells in the agar medium induced by bacterial while polysaccharide capsules are encountered
hemolysins. No red blood cell is visible on in members of group B and D which may be
microscopic examination in clear zone of complete demonstrable in very young cultures.
hemolysis.
The term ‘hemolytic streptococci’ strictly Cultural Characteristics
applies only to beta lytic strains. β-hemolysis They are aerobe and facultative anaerobes, growing
constitutes the principal marker for potentially best at a temperature of 37°C (range 22–42°C). The
pathogenic streptococci in cultures of throat swabs optimal pH for growth is 7.4–7.6. It is exacting in
or other clinical samples. nutritive requirements, growth occurring only in
media containing fermentable carbohydrates or
C. Gamma (γ) or Non-hemolytic Streptococci enriched with blood or serum.
They produce no hemolysis on blood agar. On blood agar, S. pyogenes colonies are small
Enterococcus faecalis is an important organism of (0.5–1 mm in diameter), circular, semitransparent,
this group. low convex disks surrounded by a wide zone
of β-hemolysis, several times greater than the
2. Serological Properties diameter of the colony after incubation for 24
hours. An enhancement of growth and hemolysis
Lancefield Grouping are promoted by 10% CO 2 . The matt (finely
The work of Rebecca Lancefield in 1933 laid the granular) colonies contain M antigen, which are
groundwork for the serological classification of virulent strains, while avirulent strains form glossy
b- hemolytic streptococci. On the basis of group- colonies. Mucoid colonies may occur when a strain
specific carbohydrate (C) antigens in the cell is heavily capsulate. Very rarely, nonhemoloytic
wall, b-hemolytic streptococci are divided into 21 group A streptococci are encountered, which are
serological groups from A to W (without I and J). typical of S. pyogenes in other respects.
These are known as Lancefield groups. Groups Crystal violet blood agar and PNF medium
A, B, C, D, and G are most commonly found (blood agar containing polymyxin-B, neomycin
associated with human infections. and fusidic acid) are selective for beta hemolytic
streptococci. Pike’s medium is a transport
Griffith Typing medium for clinical specimens containing Group A
Hemolytic streptococci of group A are known as streptococci. Pike’s medium is prepared by adding
Str. pyogenes. These are further divided into types crystal violet (1 in 1,000,000) and sodium azide (1
based on the protein (M, T and R) antigens present in 16,000) to blood agar.
on the cell surface (Griffith typing). About eighty In liquid media, such as glucose or serum
types of Str. pyogenes have been recognized so far broth, growth occurs as a granular turbidity with a
(types 1, 2, 3 and so on). powdery deposit. No pellicle is formed.

www.ebook3000.com
174  |  Section 3: Systemic Bacteriology
Table 25.1  Characteristics and clinical significance of important streptococci and enterococci
Species Lancefield Hemolysis Natural habitat Associated diseases Laboratory tests
group
Str. pyogenes A Beta Throat, skin Pharyngitis, scarlet fever, Bacitracin sensitive;
pyo­derma, erysipelas, PYR test positive;
cellulitis, nec­rotizing fasciitis, Ribose not
streptococcal toxic shock fermented
syndrome, bactere­mia,
rheumatic fever, glomerulo­
nephritis
Str. agalactiae B Beta Female genital Neonatal sepsis, meningitis, Hippurate
tract, rectum puerperal fever, pyogenic hydrolysis, CAMP
infections test,
S. equisimilis C Beta Throat Pharyngitis, endocarditis Ribose and
trehalose
fermentation
Enterococcus sp. Group D Variable Gastrointestinal Urinary tract infections, Growth in 6.5%
(Enterococcus hemolysis tract, oral cavity, endocarditis, bacteremia, NaCI; PYR positive
fecalis and other gallbladder, abdominal infections
enterococci) urethra, and
vagina
Nonenterococcal Group D Alpha- Gastrointestinal Neonatal meningitis No growth in 6.5%
Group D species hemolytic or tract NaCI
(Streptococcus nonhemolytic
bovis)
Str. anginosus A, C, F, G, Beta (alpha, Throat, colon, Pyogenic infections Group A strains
group untypable gamma) female genital bacitracin-resistant
tract PYR negative
colony variants of
other groups
Viridans Not typed Alpha Mouth, throat, Dental caries; endocarditis Optochin
streptococci (Str. (gamma) colon, female resistant, species
mitis, Str. mutans, genital tract classification
Str. salivarious, on biochemical
and many other properties
species)

Biochemical Reactions Antigenic Structure (Fig. 25.3)


i. Str. pyogenes is Catalase negative. 1. Inner layer of Peptidoglycan
ii. Insoluble in 10% bile unlike S. pneumoniae.
iii. It ferments several sugars producing acid and Peptidoglycan: Peptidoglycan (mucoprotein) is
no gas. responsible for cell wall rigidity, pyrogenic and
iv. Hydrolysis of pyrrolidonyl naphthylamine thrombolytic activity.
(PYR test) is positive and failure to ferment
ribose distinguishes it from non-group A 2. Group-specific Polysaccharide Antigen
hemolytic streptococci.
Serologic classification of b-hemolytic streptococci
Resistance is based on their cell wall polysaccharide antigen.
Strep. pyogenes is a delicate organism, can be killed As this antigen is integral part of the cell wall, it has
by heating at 54°C for 30 minutes. It can, however, to be extracted for grouping by a precipitation test
survive in dust for several weeks, if protected from with group antisera.
sunlight. It is more resistant to crystal violet than Techniques for the extraction of the group
many other bacteria, including Staphylococcus antigens:
aureus; hence, it is used for preparation of selective i. Lancefield’s acid extraction method: For the
media. It is sensitive to benzylpenicillin and a test, streptococci are grown in Todd-Hewitt
wide range of antimicrobial drugs. It is susceptible broth and extracted with hydrochloric acid.
to sulfonamides, but unlike Staph. aureus does
ii. Formamide (Fuller’s method).
not develop resistance to drugs. It is sensitive to
bacitracin, and this property is employed as a iii. By enzyme produced by Streptococcus albus
convenient method for differentiating Str. pyogenes (Maxted’s method).
from other hemolytic streptococci. iv. By autoclaving (Rantz and Randall’s method).
Chapter 25: Streptococcus and Enterococcus  | 175
iv. M associated protein (MAP)
A nontype-specific protein, associated with the
M protein. This is known as M-associated protein
(MAP).
b. Other cell surface components
Other important components in the cell wall of
S. pyogenes include M-like proteins, lipoteichoic
acid, and F protein. Lipoteichoic acid, and
F protein facilitate binding of host cells by
complexing with fibronectin, which is present on
the host cell surface.
c. Capsule
The outermost layer of the cell wall is the capsule,
which is composed of hyaluronic acid and is not
Fig. 25.3: Antigenic structure of Str. pyogenes: 1. Hyaluronic immunogenic. It has an antiphagocytic effect like
acid capsule; 2. Cell wall comprising; 2A, peptidoglycan, 2B.
group-specific carbohydrate, and 2C. protein lipoteichoic
other bacterial capsule.
acid fimbria; 3. Cytoplasmic membrane; 4. Cytoplasm; 5. Pili
covered with lipoteichoic acid Toxins and Enzymes
Test performance Str. pyogenes forms several exotoxins and enzymes
The extract and the specific antisera are allowed to which contribute to its virulence.
react in capillary tubes. A white disk of precipitation
may be seen within five minutes. Tests may be A. Toxins
performed in the narrowing neck of small Pasteur 1. Hemolysins
pipette. A variety of agglutination techniques using Two hemolytic and cytolytic toxins—streptolysin
extracts or whole cells may also be used. O (SLO) and streptolysin S (SLS)—are produced
by most strains of group A streptococci and many
3. Outer layer strains of groups C and G.
a. Type-specific Antigens
Several proteins antigens have been identified i. Streptolysin O
in the outer layer of the cell wall. Streptococcus Streptolysin O is so called because it is oxygen-
pyogenes can be typed, based on the surface labile hemolysin. It is inactivated in the oxidized
proteins M, T and R. form but may be reactivated by treatment with
mild reducing agents. SLO is heat-labile protein,
i. M protein cytolytic and capable of lysing erythrocytes,
The M protein is the most important of these three. leukocytes, platelets and cultured cells. It is lethal
It acts as virulence factor by inhibiting phagocytosis. on intravenous injection into animals and has a
It is antigenic and specific anti-M antibody develops specific cardiotoxic activity.
after infection. M protein is resistant to heat and
acid but susceptible to trypsin. It can be extracted ASO Test
by the Lancefield acid extraction method and M Streptolysin O is strongly antigenic and anti-
typing is performed by capillary tube precipitation streptolysin O (ASO) antibodies appears in sera
test using type-specific antisera and acid extract. following streptococcal infection. An ASO titer
About 100 M protein types have been recognized. in excess of 200 Todd units/mL is considered
significant and suggests either recent or recurrent
ii. T protein infection with streptococci.
T (Trypsin-resistant) protein is heat and acid-lable
but resistant to trypsin present in many serotypes of ii. Streptolysin S (SLS)
S. pyogenes. It may be specific but many different Streptolysin S is an oxygen-stable and causes the
M types possess the same antigen. T typing is hemolysis around the colonies on aerobic blood
performed by a slide agglutination test that uses culture. It is protein but nonantigenic. It is cell-
trypsin-treated whole streptococci. bound hemolysin that can lyse erythrocytes, leuko­
cytes, and platelets.
iii. R protein
Some strains of S. pyogenes (types 2, 3, 28 and 48) 2. Pyrogenic Exotoxins (Erythrogenic, Dick,
and some strains of groups B, C and G contain Scarlatinal Toxin)
third antigen R protein. R protein has no relation The primary effect of the toxin is induction of fever
to virulence. and so it was renamed streptococcal pyrogenic

www.ebook3000.com
176  |  Section 3: Systemic Bacteriology
exotoxin (SPE). This toxin was originally called 5. Serum opacity factor (SOF)
‘erythrogenic’ toxins because its intradermal The exact biological significance is not known it is
injection into susceptible individuals produced an produced mainly by strains causing skin infections.
erythematous reaction (Dick test, 1924). Antitoxin
6. Nicotinamide adenine dinucleotidase (NADase)
injected into the skin of a patient with scarlet fever
It is believed to be leukotoxic.
causes localized blanching as a result of neutral­
ization of erythrogenic toxin (Schultz-Charlton 7. Other enzymes
reaction). Many strains of streptococci also produce ATPase,
Three immunologically distinct types heat- phosphatase, esterases, amylase, N-acetylglucos­
labile toxins (SPE A, B, and C) have been described in aminidase, neuraminidase. and other toxins or
S. pyogenes. Type A and C are encoded by enzymes.
lysogenic phages; the gene for B is located on the
bacterial chromosome. SPEs act as ‘superantigens’ Epidemiology
interacting with both macrophages and helper T Group A streptococci commonly colonize the
cells with the release of inflammatery cytokines. oropharynx of healthy children and young adults.
These cytokines cause important effects, including Person-to-person spread is by respiratory droplets
the fever, shock, the organ failure and the rash (pharyngitis) or through breaks in skin after direct
observed in patients with scarlet fever. contact with infected person, fomite, or arthropod
vector. Although the organism is ubiquitous, there
B. Enzymes are seasonal incidences of specific diseases.
Pharyngitis due to S. pyogenes is primarily a
1. Deoxyribonucleases (streptodornase DNAase) disease of childhood between the age of 5 and 15
These enzymes are capable of depolymerizing the years, but. Crowding, such as in classrooms and
highly viscous DNA which accummlates in thick pus daycare facilities, increase the opportunity for the
as a result of disintegration of polymorphonuclear organism to spread. No seasonal distribution has
leukocytes. Streptodornase helps to liquefy the been identified in the tropics. Immunity is type
thick pus and may be responsible for the thin serous specific and appears to be associated with antibody
character of streptococcal exudates. This property to the M protein. Reinfections occur because of the
has been applied therapeutically for breaking down multiplicity of the serotypes.
blood clots, thick pus and fibrinous exudates in
closed spaces such as joints or pleural cavity. Pathogenesis
There are four antigenically distinct nucleases
Acute diseases associated with Streptococcus
(A, B, C and D), of which B is the most antigenic in
pyogenes occur chiefly in the respiratory tract,
human beings. Antibody titers to DNase B are of
bloodstream, or the skin. Two post streptococcal
great value in the serodiagnosis of pharyngeal or
sequelae (rheumatic fever following respiratory
skin infection, especially the latter, where the ASO
infection and glomerulonephritis following
titer may be low.
respiratory or skin infection), occur in 1–3% of
2. Streptokinase (Fibrinolysin) untreated infections.
Streptokinase, also known as fibrinolysin, is
another spreading factor. This acts on plasminogen, A. Suppurative Streptococcal Disease
a factor present in normal plasma, which is 1. Respiratory Infections
converted into plasmin, an active proteolytic i. Sore throat is the most common of
enzyme that lyses fibrin. Fibrinolysin appears to streptococcal disease. It may be localized as
play a biological role in streptococcal infections by tonsillitis or may involve the pharynx more
breaking down the fibrin barrier around the lesions diffusely (pharyngitis). Tonsillitis is more
and facilitates the spread of infection. common in older children and adults than in
An antigenic protein and neutralizing anti­ younger children.
bodies appear in convalescent sera which provide ii. Scarlet fever : It is a complication of
retrospective evidence of streptococcal infection. streptococcal pharyngitis that occurs when
the infecting strain is ly­s ogenized by a
3. Hyaluronidase
temperate bacteriophage that stimulates
Hyaluronidase splits hyaluronic acid, and might
production of a pyrogenic exotoxin.
favor the spread of streptococcal infection along
iii. Suppurative complications: The infection
the intercellular spaces.
may spread to the surrounding tissues which
4. Proteinase may cause suppurative complications of
It destroys several proteins formed by the Strepto­ streptococcal pharyngitis, such as periton­
coccus itself. sillar or retropharyngeal abscess, otitis
Chapter 25: Streptococcus and Enterococcus  | 177
media, mastoitidis, quinsy, Ludwig’s angina, Table 25.2  Distinguishing features of rheumatic
suppurative adenitis and disseminated fever and glomerulonephritis
infections to brain, heart, bone, and joints.
Feature Acute Acute
2. Skin and Soft Tissue Infections rheumatic glomerulonephritis
Str. pyogenes causes a variety of suppurative fever
infections of the skin, including infection of Primary site of Throat Throat or skin
wounds or burns, with a predilection to produce infection
lymphangitis and cellulitis. Infection of minor Latent period Longer Shorter (1–3 weeks)
abrasions may at times lead to fatal septicemia. (2–5 weeks)
The two typical streptococcal skin infections are Prior Essential Not necessary
erysipelas and impetigo. sensitization
i. Erysipelas: Erysipelas is an acute infection of Repeated Common Absent
the skin. It is a diffuse infection involving the attacks
superficial lymphatics. Erysipelas occurs most Genetic Present Not known
commonly in young children or older adults. susceptibility
ii. Pyoderma (impetigo): Pyoderma (impetigo) Serotype of Any Pyodermal types 49,
is seen primarily in young children. Group Str. pyogenes 53–55, 59–61 and
A streptococci causing impetigo are pharyngitis strains 1
frequently nephritogenic, that leads to acute and 12
glomerulonephritis. Immune Marked Moderate
For retrospective diagnosis of pyoderma response
antecendent to acute glomerulonephritis, Complement Unaffected Lowered
antibody to DNAase B and hyaluronidase are level
more useful. Course Progressive or Not indicated
iii. Cellulitis: Typically involves the skin and static
deeper subcutaneous tissues. Prognosis Variable Good
iv. Necrotizing fascitis (Streptococcal gangrene). Penicillin Essential Not indicated
v. Streptococcal Toxic Shock Syndrome. prophylaxis

3. Other Suppurative Infections


and is believed to result from the production of
a. Puerperal sepsis: Streptococcal puerperal
antibodies and T lymphocytes induced by cross-
sepsis used to take a heavy toll of life before
reactive components of the bacteria and host
antibiotics became available.
tissues.
b. Abscesses in internal organs, such as the
A common cross-reacting antigen may
brain, lungs, liver and kidneys.
exist in some group a streptococci and heart,
c. Septicemia and pyemia.
therefore, antibodies produced in response to
B. Nonsuppurative Sequelae the streptococcal infection could cross-react with
myocardial and heart value tissues, causing cellular
Str. pyogenes infections lead to two important destruction.
nonsuppurative sequelae: acute rheumatic
fever (ARF) and acute glomerulonephritis 2. Acute Poststreptococcal Glomerulonephritis
(AGN). Both are caused by immune reactions (AGN)
induced by the streptococcal infection. These In contrast to rheumatic fever, which occurs only
complications ensue 1–5 weeks after the acute after pharyngitis, AGN may be seen after either
infection. They differ in their natural history in a pharyngeal or a cutaneous infection. Specific
a number of respects (Table 25.2). nephritogenic strains of group A streptococci are
associated with this disease. AGN is most often
1. Acute Rheumatic Fever (ARF) seen in children
Rheumatic fever is a non­suppurative inflammatory
reaction that is epidemiologically and serologically Pathogenesis
related to antecedent group A streptococcal
1. Immune complex deposition in the glomeruli.
infection. Typically, rheumatic fever follows
2. Reaction of antibodies cross-reactive with
persitent or repeated streptococcal throat infection
streptococcal and glomerular antigen.
with a srong antibody response. The disease is
3. Alterations of glomerular tissues by strepto­
caused by specific M types.
coccal products such as streptokinase.
Pathogenesis 4. Direct complement activation by streptococcal
The pathogenesis of rheumatic fever is poorly components that have a direct affinity for
understood. The disease is autoimmune in nature glomerular tissues.

www.ebook3000.com
178  |  Section 3: Systemic Bacteriology
5. Circulating immune complexes have been D. Identification
found in the serum of patients with acute Colonies of S. pyogenes on SBA are small,
poststreptococcal glomerulonephritis. transparent, and smooth with a well-defined area
of β-hemolysis. A Gram stain will reveal gram-
Laboratory diagnosis positive cocci with some short chains. Hemolytic
streptococci are grouped by the Lancefield
In acute infections, diagnosis is established by
technique using serologic methods, or biochemical
the isolation and identification of β-streptococci
tests can be performed. The fluorescent antibody
from the patient, while in non-suppurative
technique has been employed for the rapid
complications, diagnosis is based mainly on the
identification of group A streptococci. A key test
examination of the patient’s serum for a rising titer
that should be done is bacitracin susceptibility
of antibody to one or more streptococcal antigens.
or PYR hydrolysis.
Bacitracin susceptibility: S. pyogenes is more
1. Acute Suppurative Infections
sensitive to bacitracin than most other streptococci,
A. Specimens A disk containing 0.04 units of bacitracin is applied
Throat and nose swabs, high vaginal swabs, pus on the surface of an inoculated blood culture plate.
or pus swabs are the usual specimens collected. S. pyogenes should show a large inhi­bition zone
Serum is obtained for antibody demonstration. (e.g. >15 mm diameter) and most other streptococci
should show little or no inhibition. S. pyogenes is
susceptible to bacitracin and hydrolyzes PYR,
B. Microscopy
whereas the other β-hemolytic groups are resistant
Presumptive information may be obtained by an to bacitracin and are PYR negative.
examination of Gram stained films from pus and Typing of Str. pyogenes is required for epidemio­
CSF. The observation of typical gram-positive logical in spe­cialized reference laboratories.
cocci in chains may indicate the likelihood of the
presence of streptococcal infection. In contrast, E. Antigen Detection
smears are of no value in infections of throat or
genitalia, where streptococci may form part of part Detection of Str. pyogenes is done directly in throat
of the resident flora and has a poor predictive value. swabs without cultivation. Enzyme immunoassay
(ElA) or agglutination tests are used to demonstrate
the presence of the antigen.
C. Culture
For culture, swabs should be collected under vision 2. Non-suppurative Complications
from the affected site and either plated immediately Serological Tests
or if there is likely to be delay, swab should be sent
to the laboratory in Pike’s medium (blood agar Detection of antibodies against antigens of Str.
containing 1 in 1,000,000 crystal violet and in 1 in pyogenes is an important means of establishing
16,000 sodium azide). The specimen is plated on the diagnosis of poststreptococcal rheumatic
blood agar and incubated at 37°C anaerobically or fever and glomerulonephritis. Some immunologic
under 5–10% CO2, as hemolysis develops better tests used to detect past infection with S. pyogenes
un­d er these conditions. Sheep blood agar is include ASO, anti-DNase, anti­streptokinase, and
recommend­ed for primary isolation because it is antihyaluronidase titers.
inhibitory for Haemophilus haemolyticus, colonies Antistreptolysin O (ASO) test is used most
of which may be confused with those of hemolytic frequently. ASO titers higher than 200 Todd units/
streptococci but their hemolysis is stronger on mL are indicative of prior streptococ­cal infection.
aerobic than on anaerobic plates. High levels are usually found in acute rheumatic
Crystal violet blood agar and PNF medium fever but in glomerulonephritis, titers are often low.
are selective media that inhibit many throat Antideoxyribonuclease B (anti­D Nase B)
commensal bacteria and may facilitate the estimation is also commonly employed. Titers
detection of small numbers of S. pyogenes in throat higher than 300 or 350 are taken as significant.
swabs.They are rarely used for routine culture. S. Anti­DNase B and antihyaluronidase (ASH) tests
pyogenes does not grow on MacConkey’s bile-salt are very useful for the retrospective diagnosis of
medium. streptococcal pyoderma, for which ASO is of much
For isolating group A streptococci from throat less value.
swabs the most common medium is blood agar Commercial products are available for detection
supplemented with antibiotics trimethoprim­/ of antistreptococcal antibodies. Streptozyme
sulfamethoxazole to suppress the groth of normal test detects a mixture of antibodies.It is a passive
flora. slide he­magglutination test using erythrocytes
Chapter 25: Streptococcus and Enterococcus  | 179
sensitized with a crude preparation of extracellular Other group B infections in neonates: Osteomy­
antigens of streptococci, is a convenient, sensitive elitis, conjunctivitis, sinusitis, otitis media,
and speci­fic screening test. It becomes positive endocarditis and peritonitis may also occur.
after nearly all types of streptococcal infections,
2. Infections in the adult: Puerperal sepsis,
whether of the throat or the skin.
pneumonia endo­metritis, urinary tract infection,
wound infection, and bac­teremia.
Treatment
S. pyogenes is very sensitive to penicillin and Identification
penicillin resistance has not yet been observed in Pre sumptive identification method is based on
S. pyogenes. Erythromycin or an oral cephalosporin their ability to hydrolyze hippurate. They may be
can be used in patients with a history of penicillin identified by the CAMP reaction (Christie, Atkins
allergy. Adequate treatment during acute infec­tion and Munch-Peterson), which can be demonstrated
prevents the complications of acute rhematic fever. as an accentuated zone of hemolysis (arrowhead-
shaped area of enhanced hemolysis) when Str.
agalactiae is inoculated perpendicular to a streak
Prophylaxis of Staph. aureus grown on blood agar (Fig. 25.4). S.
Patients with a history of rheumatic fever require agalactiae produces a CAMP factor that enhances
long-term antibiotic prophylaxis to prevent the lysis of sheep red cells by staphylococcal β-lysin.
recurrence of the disease. Those persons who have Group B streptococci are identified definitively by the
recovered from ARF are given oral peni­cillin for many demonstration of the group-specific carbohydrate
years to prevent recurrence. Antimicrobial drugs or the use of commercially prepared molecular
have no effect on established cases of AGN and ARF. probes. Occasional strains are bacitracin sensitive.

Other streptococci pathogenic Group C Streptococci


for humans Streptococci of this group are predominantly
Besides Str. pyogenes, streptococci belonging to animal pathogens and comprise four species: S.
groups B, C, D, F, G and rarely H, K, O and R may equi, S. equisimilis, S. dysgalactiae and S. zooep­
also cause human infections. idemicus. Group C strains isolated from human
sources usually belong to S. equisimilis. It can
Group B Streptococci: Streptococcus cause upper respiratory infections, as well as deep
infections, such as endocarditis, osteomyelitis,
agalactiae
brain abscess, pneumonia and puerperal sepsis.
Streptococcus agalactiae belongs to Lancefield All species of group C are β-hemolytic, with
group B. Human pathogenic Group B strains the exception of S. dysgalactiae, which may be
possess a polysaccharide capsule. Nine capsular a-hemolytic or nonhemolytic. It resembles Str.
serotypes have been identified. pyogenes in fermenting treha­lose but differs in
Previously, Streptococcus agalactiae was fermenting ribose. It produces streptolysin O,
recognized primarily as a cause of bovine mastitis streptokinase and other extracellu­lar substances.
(agalactia, want of milk). It has become the leading
cause of neonatal infections in industrialized Treatment: Treatment is similar to that used for
countries and is also important cause of morbidity group A streptococcal infections.
among peripartum women and nonpregnant
adults with chronic medical conditions. Group G Streptococci
Strept. agalactiae is found in the vaginocervical These are commensals in the throats of hu­man
tract of female carriers. Transmission occurs from beings, monkeys or dogs. These may cause sore
an infected mother to her infant at birth.

Clinical Diseases
1. Infection in the neonate
A. Early-onset disease : It develops during the
first week of life and disease is characterized by
bacteremia, pneumonia, or meninigitis and is
often fatal.
B. Late-onset disease: It develops between
second and twelfth weeks of life. The predominant
manifestation is bacteremia with meningitis, but
septic arthritis. Fig. 25.4: CAMP reaction

www.ebook3000.com
180  |  Section 3: Systemic Bacteriology
throat, pneumo­nia, septicemia, endocarditis, and media and on MacConkey agar, on which it forms
bone, joint, skin and wound infections. small (0.5–1 mm), usually magenta-colored
colonies.
Group D Streptococci
Distinctive Features of Enterococci
Until the mid-1980s, the group D strepto­cocci were
The enterococci possess several distinctive
divided into the two groups:
features separting them from sreptocooci: The
1. Enterococcus group (enterococci or fecal
enterococci grow in the presence of 6.5% NaCl,
streptococci) which have been reclassified as
40% bile, at ph 9.6, at 45°C and in 0.1% methelyne
a separate genus called Enterococcus.
blue. It survives heating at 60°C for 30 min, a
2. Nonenterococcal group, for example, Str. bovis,
feature distinguishing it from streptococci, and
Str. equinus.
also grows within a wider range of temperatures
(10–45°C). On MacConkey medium, they produce
Enterococcus deep pink colonies. Enterococci are PYRase test
The enterococci (“enteric cocci”) were previously positive. They do not hydrolyze hippurate.
clas­sified as group D streptococci (Table 25.3). The
enterococci were reclassified into the new genus Identification
Enterococcus, and there are currently 16 species The identification of Enterococcus species is made
in this genus. on biochemical characteristics. E. aecalis can
be identified by its ability to ferment mannitol,
Species sucrose, sorbitol and aesculin, and to grow on
Entero­coccus faecalis (“pertaining to feces”) is tellurite blood agar producing black colonies with
the Ente­rococcus most often isolated from human gas production. It is VP positive.
sources.
Enterococcus fae­cium (“of feces”). Clinical Infections
Other species: E. durans, E. avium, E. casseliflavus, The enterococci inhabit the gasterointestinal
E. gallinarum, and E. raffinosus are observed tract and the genitourinary tract in humans and
occasionally. other animals. Enterococci are frequent causes of
nosocomial infec­tions and may cause urinary tract
Characteristics of Enterococci infection, bacteremia, infective endocarditis,
The enterococci are gram-positive cocci typically biliary tract infection, intra-abdominal abscess
ar­ranged in pairs and short chains, and is nonmotile complicating diverticulitis, peritonitis and
and noncapsulate. The cocci are facultatively wound infection.
anaerobic and grow optimally at 35°C, although
most isolates can grow in the temperature range Treatment
10°C to 45°C. They grow read­ily on blood agar Most strains of enterococci are resistant to
media, with large, white colonies appearing penicillin. They are also resistant to sulfona­mides.
after 24 hours of incubation; the colonies are Recently they have developed resist­ance to newer
typically nonhemolytic but can be α-hemolytic or penicillins and cephalosporins, streptomycin and
β-­hemolytic. It grows readily on ordinary nutrient gentamicin.

Nonenterococcal Species of Group D


Table 25.3  Differences between group D
streptococci and Enterococcus spp Nonenterococcal species of group D (Str. bovis,
Str. equinus) are generally susceptible to penicillin
Characteristics Group D Enterococcus spp and are inhibited by 6.5% sodium chloride or bile.
streptococci
They may cause urinary infection or endocarditis
1. Hemolysis type α , none α, β, none rarely.
2. G
 rowth in 6.5% – +
sodium chloride Viridans streptococci
3. G
 rowth in – +
presence of 40% The viridans streptococci are commensals of
bile mouth and upper respiratory tract. The viridans
4. Growth at 45°C – + group of streptococci are a heterogeneous
collection of α-hemolytic and nonhemolytic
5. PYRase test – +
strepto­c occi. The term viridis means “green.”
6. S usceptibility to + – (Latin for “green”) because many of these bacteria
penicillin
produce a green pigment on blood agar media.
Chapter 25: Streptococcus and Enterococcus  | 181
Classification Key Points
The current clas­sification assigns streptococci ™™ Streptococci are gram-positive cocci arranged in
species in the viridans group to one of the four long chains
groups: ™™ Three different schemes are used to classify the
1. Anginosus group (S. anginosus, S. constellatus, organism:
Streptococcus pyogenes
and S, intermedius): Organisms of the angi­ ™™ Strep. pyogenes are gram-positive, spherical to ovoid
nosus group may possess the Lancefield group organisms
A, C, F, G, or N antigen and in some instances ™™ Catalase-negative; PYR-positive; bacitracin-
may not be groupable. susceptible are important identification tests
™™ Crystal violet blood agar and PNF medium are
2. Mitis group (S. sanguis, S. mitis, etc.)
selective for beta hemolytic streptococci
3. Mutans group (S. mutans, etc.) ™™ Antigenic structure: Many streptococci can be
4. Salivarius groups (S. salivarius). categorized based on Lancefield group antigens
Diseases:
A. Respiratory infection 1. Streptococcal pharyngitis;
Clinical Infections 2. Scarlet fever (complication of pharyngitis)
The viridans streptococci are opportunistic B. Pyogenic cutaneous infections: Impetigo,
pathogens but can, on occasion, cause disease. erysipelas, cellulitis; necrotizing fasciitis involving
deep subcutaneous tissues; streptococcal toxic
Two clinically important phenomena are associ­ shock syndrome
ated with viridans streptococci. dental caries and ™™ Nonsuppurative sequelae: Rheumatic fever and
subacute endocarditis. Streptococcus anginosus is acute glomerulonephritis
responsible for causing pyogenic infections, They ™™ Laboratory diagnosis is done by microscopy, culture,
have also been implicated in meningitis, abscesses, antigen detection and serological tests such as
osteomyelitis, and empyema. ASO, anti-DNase, antistreptokinase, and anti-
hyaluronidase titers to detect past infection with
S. pyogenes
1. Dental Caries ™™ Streptococcus agalactiae is a significant cause of
invasive disease in newborns. Positive CAMP and
S. mutans is the principal cause of dental caries
hippurate hydrolysis reactions are important
(tooth decay). S. mutans adheres to dental surfaces identification tests
via extracellular carbohydrates (dextran) and ™™ Enterococci: Enterococcus faecalis is the Enterococcus
erodes the teeth by con­verting sucrose to acetic most often isolated from human sources. Enterococci
acid and lactate. It breaks down dietary sucrose, are frequent causes of nosocomial infections and
producing acid and a tough adhesive dextran. may cause urinary tract infection, bacteremia,
infective endocarditis, biliary tract infection, intra-
The acid damages dentine and the dextrans bind abdominal abscess complicating diverticulitis,
together food debris, epithelial cells, mucus and peritonitis and wound infection
bacteria to form dental plaques, which lead ™™ Viridans streptococci: The viridans streptococci are
to caries. Experimental caries in monkeys has commensals in mouth and upper respiratory tract
been prevented by a Str. mutans vaccine, but its that are regarded as opportunistic pathogens. Two
extention to human use is fraught with problems. clinically important phenomena are associated with
viridans streptococci: dental caries (tooth decay)
and infective endocarditis.
2. Subacute Endocar­ditis
Viridans streptococci are the most common cause Important Questions
of subacute bacterial endocarditis. About two-
thirds of the viridans-associated cases are due to 1. Classify streptococci. Describe the laboratory
S. sanguis and S. mutans. Transient bacteremia is diagnosis of streptococcal sore throat.
associated with endocarditis. Most patients have 2. Write short notes on:
underlying valvular heart disease, and the course a. Antigenic structure of Str.pyogenes
of the endocarditis is generally subacute. Dental b. Lancefield grouping
procedures, vigorous tooth brushing or chew­ing, c. Toxins and enzymes of Streptococcus pyogenes
and use of a water pick to clean teeth can cause d. Pathogenicity of streptococci
a transient bacteremia that is enough to initiate e. Nonsuppurative complications of Str. pyogenes
valvu­lar infection in a person with damaged infections
valvular tissue. While viridans streptococci 3. Write briefly about:
are generally penicillin-sensitive, some strains a. Group B streptococci
may be resistant. It is, therefore, essential that b. CAMP reaction
in endocarditis, the causative strain is isolated c. Group D streptococci
and its antibiotic sensitivity determined so that d. Enterococci (or) fecal streptococci
appropriate antibiotics in adequate bactericidal e. Viridans streptococci
concentration can be employed for treatment. f. Heat test.

www.ebook3000.com
182  |  Section 3: Systemic Bacteriology
Multiple choice questions (MCQs) 4. Which of the following is selective medium for
Streptococcus pyogenes?
1. Which type of hemolysis is produced by Strepto­ a. Blood agar
coccus pyogenes on blood agar?
b. Crystal violet blood agar
a. Alpha hemolysis b. Beta hemolysis
c. Gamma hemolysis d. None of the above c. Potassium tellurite blood agar
2. Group-specific antigen extraction of Streptococcus d. Chocolate agar
pyogenes by treating with hydrochloric acid 5. Susceptibility to bacitracin can be used to identify:
method is known as
a. Streptococcus pyogenes
a. Lancefield’s method b. Fuller’s method
b. Streptococcus viridans
c. Maxted’s method d. Randall’s method
3. Erythrogenic toxin is responsible for: c. Streptococcus mitis
a. Pyoderma d. Streptococcus agalactiae
b. Schultz-Charlton reaction
c. Necrotising facititis Answers (MCQs)
d. All of the above 1. b; 2. a; 3. b; 4. b; 5; a
26
Chapter
Pneumococcus (Diplococcus pneumoniae:
Streptococcus pneumoniae)

Learning Objectives

After reading and studying this chapter, you should ∙∙ Explain C-reactive protein
be able to: ∙∙ Discuss laboratory diagnosis of pneumococcal
∙∙ Describe morphology and cultural characters of infections
pneumococci ∙∙ Differentiate between Str. pneumoniae and Str.
∙∙ Describe Quellung reaction viridans.

Introduction for which some strains have a strict requirement.


Optimum temperature being 37°C (range 25–40°C)
Pneumococcus, a gram-positive lanceolate and pH 7.8 (range 6.5–8.3). Pneumococcus has
Diplococcus, formerly classified as Diplococcus complex nutritional requirements and grow only
pneumomiae, has been reclassified as Str.
pneumoniae because of its genetic relatedness to
streptococcus. Pneumococcus differs from other
streptococci chiefly in its morphology, bile solubility,
optochin sensitivity and possession of a specific
polysaccharide capsule. Pneumococci are normal
inhabitants of the human upper respiratory tract.

Pneumococcus
(Diplococcus Pneumoniae,
Streptococcus Pneumoniae)
Morphology
Pneumococci are gram-positive cocci in pairs Fig. 26.1: Str. pneumoniae in pus
(diplococci). The cocci are about 1 μm, slightly
elongated cocci, with one end broad or rounded
and the other pointed, presenting a flame-shaped
or lanceolate appearance (Fig. 26.1). They are
nonmotile and nonsporing.
All freshly isolated strains are capsulate. The
capsule encloses each pair. The capsule may
be demonstrated as a clear halo in Indian ink
preparations (Fig. 26.2) or may be stained directly
by special techniques or by use of homologous
type-specific antibody in the Quellung reaction.

Cultural Characteristics
They are aerobes and facultative anaerobes. It Fig. 26.2: Pneumococci. Indian ink preparation to show
grows best in air or hydrogen with 5–10% CO2 , capsules

www.ebook3000.com
184  |  Section 3: Systemic Bacteriology
in enriched me­dia. It grows on ordinary media, but Optochin Sensitivity: Pneumococci are highly
better on media with serum, blood or heated blood. sensitive to killing by optochin (ethyl hydrocuprein
On blood agar, after incubation for 18 hours, hydro­chloride), and is useful in distinguishing
the colonies are small (0.5–1 mm), dome-shaped on an area of a blood agar plate inoculated with
and glistening, with an area of green discoloration pneumo­coccus-like colonies from the primary
(alpha hemolysis) around them. On further diagnostic plate. A growth of Pneumococcus will
incubation, the colonies become flat with raised be inhibited in a zone extending radially for at least
edges and depressed centrally, so that concentric 5 mm from the margin of the disc on incubation.
rings are seen on the surface when viewed from Viridans streptococci will grow right up to the disc.
above (draughtsman or carrom coin appear­ance)
which is due to autolysis of bacteria within the flat Antigenic Structure
pneumococcal colonies.
1. Capsular Antigens
Under anaerobic conditions, however, a zone of
beta hemolysis is produced around the colony by The most important antigen of the Pneumococcus
an oxygen­labile pneumolysin O. In liquid media is the type specific capsular polysaccharide. It is
such as glucose broth, growth occurs as uniform also called the ‘specific soluble substance’ (SSS)
turbidity. The cocci readily undergo autolysis in as this polysaccharide diffuses into the culture
cultures due to the activity of intracellular enzymes. medium or infective exudates and tissues. These
Autolysis is en­hanced by bile salts, sodium lauryl polysaccharides are antigenic and form the basis
sulfate and other surface active agents. for the separation of pneumococci into different
serotypes. A total of 90 different capsular serotypes
have been identified. The serotypes are designated
Biochemical Reactions
by numbers, and those that are structurally related
1. Inulin fermentation: Pneumococci ferment are grouped together (I, 2, 3. 4, 5, 6A, 68, etc.).
sever­al sugars with the production of acid and The tests may be done by :
no gas. Fermentation of inulin by pneumococci 1. Agglutination of washed capsulate cocci.
is a useful test for dif­ferentiating them from 2. Precipi­tation of SSS from culture supernates.
streptococci as the latter do not ferment it. 3. Quellung reaction or capsule swelling reaction
Fermentation is tested in Hiss’s serum water It was described by Neufeld (1902). In the
or serum agar slopes. capsule swelling or ‘quellung’ reaction (quellung
2. Bile solubility test: = swelling), a suspension of pneumococci is mixed
Basis: Bile solubility test for identifying on a slide with a drop of the type specific antiserum
pneu­m ococci is based on the presence in and a loopful of methylene blue solution and then
pneumococci, of an autolytic amidase that examined using the oil-immersion objective. The
cleaves the bond between alanine and muramic capsule becomes apparently swollen, sharply
acid in the peptido­g lycan. The amidase is delineated and refractile in the presence of the
activated by surface-active agents such as bile homologous antiserum. This reaction can be used
or bile salts, resulting in lysis of the organisms. to identify the organism directly from ‘sputum, CSF,
Procedure: When sodium deoxy­c holate and other sources. It used to be a routine bedside
solution is added to broth culture, the culture procedure in the past.
clears due to lysis of cocci. Pneumococci are
soluble in bile; viridans and other streptococci 2. Somatic Antigen
are not.
a. C polysaccharide: The cell wall of S. pneumoniae
Alternatively, touch a suspected pneumo­
contains a species specific carbohydrate
coccal colony with a loopful of 2% sodium
antigen, referred to as C substance. C-reactive
deoxycholate solution, it disappears, leaving
protein (CRP) is an abnormal protein (beta
an area of α-hemolysis on the blood agar.
globulin) that precipi­tates with the somatic ‘C’
3. Pneumococci are catalase and oxidase
antigen of pneumococci. It appears in acute
negative.
phase sera of cases of pneumonia but dis­
appears during convalescence. It is known as
Resistance C-reactive protein because it precipitates with
Pneumococci are delicate organisms and are killed C antigen of pneumococci. CRP is present in
by moist heat at 55°C in 10 min, and readily by most low concentrations in healthy people but in
disinfectants. Most strains are highly sensitive to elevated concentrations in patients with acute
benzylpenicillin, other penicillins. A drug resistant inflammatory diseases.
Strep. pneumoniae (DRSP) strain originating in It is an ‘acute phase’ substance, produced
Spain has spread to most parts of the world posing in hepatocytes. Its production is stimulated by
problems in treament. bacterial infections, inflammation, malignancies
Chapter 26: Pneumococcus (Diplococcus pneumoniae: Streptococcus Pneumoniae)  | 185
and tissue destruction. It disappears when the pneumococcal pneumonia. In children, types
inflammatory reactions subside. It is used as 6, 14, 19 and 23 are frequent causes.
an index of response to treatment in rheumatic ii. Bronchopneumonia: It is almost always a
fever and certain other conditions. secondary infection. This may be caused by
CRP testing, by passive ag­glutination using any serotype of Pneumococcus. Other causative
latex particles coated with anti-CRP antibody agents responsible for bronchopneumonia
is a routine diagnostic procedure. include Staph. aureus, K. pneumoniae, Str.
b. F antigen: The lipid ­bound teichoic acid in the pyogenes, H. influenzae, Fusobacterium species
bacterial cytoplasmic mem­brane is called the and Bacteroides.
F or Forssman antigen because it can cross-
react with the Forssman surface antigens on 2. Acute Exacerbations in Chronic Bronchitis
mammalian cells. Pneumococci are commonly associated with
c. M protein: Type-specific protein antigens the acute exacerbations in chronic bronchitis.
analogous to the M protein of Streptococcus Another bacterium commonly associated with this
pyogenes. condition is Haemophilus influenzae.

Genetic Variation 3. Meningitis


1. Smooth-to-rough (S–R) variation: On repeated S. pneumoniae is among the leading causes of
subculture, pneumococci undergo a smooth- bacterial meningitis. It can spread into the central
to-rough (S–R) variation. nervous sys­tem after bacteremia, infections of the
ear or sinuses, or head trauma.
2. Transformation: Rough pneumococci derived
from capsulated cells of one serotype may 4. Bacteremia
be made to produce capsules of the same or
different serotypes, on treatment with DNA Bacteremia occurs in 25–30% of patients with
from the respective serotypes of pneumococci. pneumococcal pneumonia and in more than 80%
of patients with meningitis.
Virulence Factors 5. Other Infections
1. Capsule: The capsular polysaccharide is a crucial Pneumococci may also produce suppurative
virulence factor. The capsule is antiphagocytic, lesions in other parts of the body—empyema,
inhibiting complement deposition and pericarditis, otitis media, sinusitis, conjunctivitis,
phagocytosis. Noncapsulated strains are avirulent. suppurative arthritis and peritonitis, usually as
The antibody to the capsular polysaccharide complications of pneumonia.
affords protection against in­fection.
2. IgAI protease: Pneumococci produce an Epidemiology
extra­cellular protease that specifically cleaves S. pneumoniae is a common inhabitant of the throat
human IgA I in the hinge region. and nasopharynx in healthy people. Pneumonia
3. Pneumolysin: A cytotoxin similar to the occurs when the endogenous oral orga­nisms are
streptolysin O in S. pyogenes and so may be a aspirated into the lower airways. Cases are more
virulence factor. common in winter and affect the two extreme age
4. Autolysin: When activated, the pneumococcal groups more often.
auto­lysin breaks the peptide cross-linking of the
cell wall peptidoglycan, leading to lysis of the Laboratory Diagnosis
bacteria. 1. Specimens
Sputum, lung aspirate, pleural fluid, cerebrospinal
Pathogenesis
fluid, urine or blood are collected according
Pneumococci are one of the most common to the site of lesion. Blood culture is useful in
bacteria causing pneumonia, both lobar and pnemococcal septicemia.
bronchopneumonia. They also cause acute
tracheobronchitis and empyema. Bacteremia 2. Collection and Transport
may complicate pneumococcal pneumonia. All the specimens should be collected in sterile
This can result in metastatic involvement of the containers under all aseptic conditions. They
meninges, joints and, rarely, the endocardium. should be processed immediately. CSF specimen
should never be refrigerated in case of delay and
1. Pneumonia should be kept at 37°C (H.influenzae, another
i. Lobar pneumonia: In adults, types 1–8 causative agent of pyogenic meningitis may die at
are responsible for about 75% of cases of cold temperature).

www.ebook3000.com
186  |  Section 3: Systemic Bacteriology
3. Microscopy and Antigen Detection Table 26.1  Differential characters of pneumococci
A centrifuged deposit of the CSF should be and viridans streptococci
examined immediately in a Gram film in case of Character Pneumococcus Viridans
meningitis and presumptive diagnosis may be streptococci
made by finding gram-positive diplococci both Morphology Ovoid or Short or long
inside the poly­morphs and extracellularly. lanceolate chains of
Pneumococcal antigen is often detectable by diplococci; some rounded cocci
short chains
coagglutination (COA), latex agglutination (LA) or
counterimmuno­electrophoresis (CIE) and ELISA. Capsule Present Usually absent
In addition to CSF, capsular polysaccharide Colonies Become Convex
can be demonstrated in the blood and urine by flattened
or draughtsman
counterim­munoelectrophoresis
Effect on Narrow zones of Wide or narrow
blood agar a-hemolysis zone of
4. Capsule Swelling Tests a-hemolysis
If typing sera are available, the most simple, rapid, Optochin Sensitive Resistant
and accurate method for the identification of sensitivity
pneumococci by direct examination is the quellung Bile solubility + –
reaction. Inulin + –
fermentation
5. Culture Virulence in mice + –
Specimen is inoculated on plates of blood agar and
heated-blood agar incubated in air with 5–10%
CO2 for 18–24 hours. Typical colonies develop Prophylaxis
with α-hemolysis. The colonies are small (0.5–1 Immunity is type-specific and associated
mm), dome-shaped and glistening, with an area with antibody to the capsular polysaccharide.
of green discoloration (alpha hemolysis) around The existence of some 90 serotypes makes a
them. On further incubation, the colonies have complete polyvalent vaccine impracticable. The
draughtsman or carrom coin appear­ance. current vaccine contains 23 different capsular
polysaccharides which is stated to give 80–90%
6. Identification protection. The vaccine is immunogenic in normal
Procedures commonly used to distinguish S. adults, and the immunity is long-lived. It is not
pneumoniae from the viridans strep­tococci are meant for only in persons at enhanced risk of
optochin susceptibility, bile solubility, and the pneumococcal infection, such as patients with
quellung reaction. S. pneumoniae is susceptible to absent or dysfunctional spleen, sickle cell dis­ease,
optochin, whereas other α-hemolytic species are celiac disease, chronic renal, lung, heart and liver
resistant (Table 26.1). diseases, diabetes mellitus and immunodeficien­
cies, including human immunodeficiency virus
7. Intraperitoneal Injection into Mice (HIV) infection and renal transplant.
Vaccination is contraindicated in young
Isolation may be obtained by intra­p eritoneal
children and elderly with lymphoreticular malig­
inoculation in mice from specimens where
nancies and immunosuppressive therapy.
pneumococci are expect­ed to be scanty. Inoculated
mice die in 1–3 days, and pneu­mococci may be
demonstrated in the peritoneal exu­date and heart Treatment
blood. Penicillin is the drug of choice for susceptible
strains, al­t hough resistance is increasingly
8. Blood Culture common. Cephalosporins, er ythromycin,
chloramphenicol, or van­comycin are used for
In the acute stage of pneumonia, the organism may patients allergic to penicillin or for treatment of
be obtained from blood culture in glucose broth. penicillin-resistant strains.
The finding of pneumococci in the blood is much
better evidence of their pathogenic role in the Key Points
lung than is their finding in sputum. Isolation of ™™ Streptococcus pneumoniae are elongated or
pneumococci from blood indicates bad prognosis. “lancet-shaped,” gram-positive cocci arranged in
pairs (diplococci). They are capsulated
™™ The Pneumococcus has complex nutritional
9. Antibiotic Sensitivity Test
requirements. On blood agar, the colonies are small
It is especially useful in strains which are resistant.
Chapter 26: Pneumococcus (Diplococcus pneumoniae: Streptococcus Pneumoniae)  | 187

(0.5–1 mm), dome-shaped and glistening, with an


2. Draughtsman appearance colony is a characteristic
area of green discoloration (alpha hemolysis) around feature of:
them. On further incubation, the colonies become a. Streptococcus pyogenes
flat with raised edges and depressed centrally, so b. Strep. pneumoniae
that concentric rings are seen on the surface when c. Enterococcus jacecalis
viewed from above (draughtsman or carrom coin
appear­ance) d. Viridans streptococci
™™ Pnemococci differs from viridans streptococci in 3. Streptococcus pneumoniae causes following
their morphology (lanceolate diplococci), optochin infections except:
sensitivity, bile solubility, inulin fermentation and a. Otitis media
virulence in mice
™™ Bacteria are inhibited by optochin (useful identi­
b. Urinary tract infections
fication test) c. Meningitis
™™ Diseases: Pneumonia, meningitis, sinusitis and otitis d. Sinusitis
media. It can cause a variety of systemic infections,
4. Streptococcus pneumoniae shows following
including bacteremia and endocarditis.
characteristics except:
a. Optochin sensitivity test positive
Important questions b. Bile solubility test positive
1. Describe the laboratory diagnosis of pneumococcal c. Inulin fermentation test positive
infections d. Bacitracin test positive
2. Differentiate between Str. pneumoniae and Str.
viridans in a tabulated form. 5. Pneumococcal 23-valent polysaccharide is given
to following persons except:
Multiple choice questions (MCQs) a. Child younger than 2 years
b. Persons older than 65 years
1. Which of the following bacteria produce alpha c. Persons with HIV infections
hemolysis on blood agar?
d. Persons with diabetes mellitus
a. Staphylococcus aureus
b. Streptococcus pyogenes
c. Streptococcus pneumoniae Answers (MCQs)
d. All of the above 1. c; 2. b; 3. b; 4. d; 5. b

www.ebook3000.com
27
Chapter

Neisseria and Moraxella

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe morphology, culture characteristics,
be able to: biochemical reactions of Neisseria gonorrhoeae
∙∙ Describe morphology, culture characteristics, ∙∙ Discuss pathogenicity of N. gonorrhoeae
biochemical reactions and antigenic structure of ∙∙ Discuss laboratory diagnosis of gonorrhoea
Neisseria meningitidis ∙∙ Explain nongonococcal urethritis (or) nonspecific
∙∙ Discuss pathogenicity and lab diagnosis of urethritis
meningococcal meningitis ∙∙ Describe Morexella (Branhamella) catarrhalis.

Introduction Cultural Characteristics


Meningococci have exact­ing growth requirements
Members of the genus Neis­s eria are aerobic,
and do not grow on ordinary media. Growth occurs
gram-nega­tive cocci typically arranged in pairs
on media enriched with blood, serum or ascitic
(diplococci) with adjacent sides flattened together
fluid. Strains will grow on Mueller–Hinton me­dium
(resembling coffee beans). All species are oxidase-
without the addition of blood or serum.
positive, and most produce catalase-properties.
They are strict aerobes, no growth occurring
Species anaerobically. The optimum temperature for
growth is 35–36°C. No growth takes place below
Important spe­c ies of the genus Neisseria are:
30°C. Opti­mum pH is 7.0–7.4. Growth is facilitated
N. meningitidis, N. gonorrhoeae, N. flavescens, N.
by 5–10% CO2 and high humidity. Blood agar,
subflava, N. sicca, N. mucosa, N. lactamica and
chocolate agar and Mueller–Hinton starch casein
N. polysac­chareae. N. gonorrhoeae and N. menin­
hydrolysate agar are the media common­ly used for
gitidis are the primary human pathogens of the
genus.

Neisseria meningitidis
(Meningococcus; Diplococcus
intracellularis meningitidis)
Morphology
Meningococci are gram-negative oval or spherical
cocci (0.6–0.8 μm in size), typically arranged
in pairs, with the adjacent sides flattened or
concave opposing edges and the long axes parallel.
They are typically seen in large numbers inside
polymorphonuclear leukocytes (Fig. 27.1).
Most fresh isolates are capsulated. They are Fig. 27.1: Neisseria meningitidis in cerebrospinal fluid.
nonsporing and nonmotile. Inset—enlarged view showing flat adjacent side of cocci
Chapter 27: Neisseria and Moraxella  | 189
culturing meningococci. Modified Thayer -Martin Stages of Meningococcal Infections
(with vancomycin, colistin and nystatin) is a useful There are three stages.
selective medium.
On blood agar after 24 hours incubation, First Stage—Nasopharyngeal Infection
colonies are 1–2 mm in diameter, round, convex, The organisms appear in naso­pharynx leading
gray, translucent and nonhemolytic. Heated blood to nasopharyngeal infection, which is usually
(chocolate) agar-colonies are slightly larger on asymptomatic.
heated blood (chocolate) agar than on ordinary
blood agar. Growth is poor in liquid media, producing Second Stage—Meningococcal Septicemia
a granular turbidity with little or no surface growth. In a small percentage of cases, the meningococci enter
the bloodstream from the posterior nasopharynx.
This stage is called meningococcemia. The patient
Biochemical Reactions
develops fever, malaise and petechial skin lesions.
1. They are catalase and oxidase positive. The organisms may also cause lesions in the
Oxidase test: When 1% solution of oxidase joints and lungs and rarely cause massive bilateral
reagent (tetramethyl­paraphenylene-diamine- hemorrhages in the adrenals (Waterhouse–
dihydrochloride) is poured on culture media, Friderichsen syndrome). It is an overwhelming
Neisseria colonies quickly turn deep-purple. and usually fatal condition, characterized by shock,
This prompt oxidase reaction helps in the disseminated intravascular coagulation (DIC)
identification of meningococci and gonococci and multisystem failure. Meningococcal dis­ease is
in mixed cultures. favored by deficiency of the terminal comple­ment
The test may also be performed by rubbing components (C5–C9).
a little of the growth with a loop on a strip of
filter paper moistened with the oxidase reagent Third Stage—Meningitis
(Kovacs’ method). A deep purple color appears In the third stage of meningococcal infection, the
immediately. or­ganisms can cross the blood–brain barrier and
2. Sugar fermentation: Meningococci ferment infect the meninges. The route of spread from the
glucose and maltose with the production of nasopharynx to the meninges is controversial. The
acid but no gas, but not lactose or sucrose spread may be directly along the perineural sheath
(gonococci ferment glucose but not mal­tose). of the olfactory nerve, through the cribriform plate
to the subarachnoid space, or more probably,
3. Indole and hydrogen sulfide are not produced
through the bloodstream. In certain cases, the site of
and nitrates are not reduced.
entry of the Meningococcus may be the conjunctiva.
On reaching the central nervous system, a
Antigenic Classification suppurative le­sion of the meninges is set up. Case
fatality is variable but in untreated cases may be as
Based on their capsular polysaccharide antigens,
high as 80%. Survivors may have sequelae such as
meningococci are classified into at least 13
blind­ness and deafness.
serogroups: A, B, C, X, Y, Z, Zl (29E) and W135.
The pathogenic agent in meningococcal disease
Further serogroups H, I, K and L have also been
appears to be the endotoxin (LPS) released by
described. A group D was described but no
auto­lysis. The vascular endothelium is particularly
capsular polysaccharide specific for this group has
sensi­tive to the endotoxin.
yet been demonstrated. Groups A, B and C are the
most important. Serogroups are further classified Epidemiology
into serotypes and subtypes.
Humans are the only natural carriers for N. menin­
gitidis. The oral and nasopharyngeal carriage rates are
Resistance highest for school-aged children and young adults,
are higher in lower socioeconomic populations.
Meningococci are very delicate organ­isms, being
The carrier rate is higher in the members of the
highly susceptible to heat, dessication, alterations
household of a pa­tient with meningococcal disease.
in pH and to disinfectants. They die within a few
Natural infection is limited to human beings.
days at room temperature. They are killed by
heating at 55°C in 5 minutes. Laboratory Diagnosis
1. Specimens
Pathogenicity
i. Cerebrospinal fluid (CSF),
Meningococci are strict human parasites ii. Blood for culture (which may come from a
inhabiting the nasopharynx. Infection is usu­ally patient with meningitis, a hemorrhagic rash
asymptomatic. or pyrexia of uncertain origin)

www.ebook3000.com
190  |  Section 3: Systemic Bacteriology
iii. Aspirate from skin lesions or pus from an The oxidase test is performed on colonies on
infected joint, solid medium.
vi. Throat or nasopharyngeal swabs from vi. Antibiotic sensitivity tests
suspected cases. Swabs should be transported Set up antibiotic sensitivity tests.
in Stuart’s transport medium. All speci­ vii. Serogrouping is performed by slide agglutina­
mens where meningococcal infection is tion with hyperimmune sera that provide
suspected must be submitted to the laboratory important epidemiological information.
immediately.
3. Blood Cultures
2. Examination of CSF Blood culture is often positive in meningococcemia
If meningitis is suspected, a lumbar puncture and in early cases of meningitis. Cultures should
should be performed as soon as possible unless be incubated for 4–7 days, with daily subcultures.
there are signs of raised intracranial pressure. Subculture to blood agar and heated blood agar.
i. Perform a cell count. The exudate in mening­ Incubate cultures in 5–10% CO 2 for 24 hours
ococcal meningitis is typically polymor­ and examine oxidase-positive colonies of gram-
phonuclear. negative diplococci as above.
ii. Centrifuge the remaining CSF. Make a
smear of the centrifuged deposit and stain 4. Pus, Aspirates and Swabs
with Gram-stain. CSF from a typical case of Gram-stained films are examined. In addition
meningococcal meningitis will show gram- to blood agar and heated blood agar, Thayer–
negative diplococci inside a limited proportion Martin selec­tive medium is used for the culture of
of the pus cells; many are extracellular. materials expected to yield a mixture of organisms
Stain a second film with methylene blue to such as pus, aspirates, and throat, nasopharyngeal
determine the cell type; occasionally, diplococci and genital swabs.
may be seen more easily with this stain.
If fluorescein isothiocyanate­ coupled
5. Petechial Lesions
antiserum is available, a smear of the de­posit
may be examined for the direct identification Meningococci may sometimes be demonstrated in
of the meningococcal serogroup responsible petechial lesions by microscopy and culture.
for infection.
iii. Divide the supernatant CSF into two aliquots­: 6. Serological Diagnosis
One to be kept if necessary for biochemical Paired sera may be tested for the presence of
examina­tion, the other to be examined for the complement-fixing antibodies.
presence of meningococcal polysaccharide Specific antibodies to capsular polysaccha­ride
antigen by counter­immunoelectrophoresis, may be demonstrated by a (i) hemagglutination
latex agglutination or coagglutination using test (ii) ELISA tests.
meningococcal antisera. Similar tests are also
available for pneumococcus, H. influenzae 7. Polymerase Chain Reaction (PCR)
type b and group B Streptococcus antigens. Group specific diagnosis of infection can be made
Antigen detection is particularly useful in by detection of meningococcal DNA sequence in
partially treated pa­tients in whom smear and CSF or blood by PCR amplification.
culture tests may be nega­tive.
iv. Cuture: Plate out the centrifuged deposit on Treatment
both blood and heated blood agar (chocolate
agar) and incubate at 37°C in 5–10% CO2. Penicillin is currently the antibiotic of choice.
Colonies appear after 18–24 hours which Chloramphenicol is effective, but risks of blood
may be identified by morphology and dyscrasia have limited its use. Either chloramphenicol
biochemical reactions. or a third-generation cephalosporin, such as
Subculture: Add Robertson’s cooked meat cefotaxime or ceftriaxone is used in persons allergic
broth to the remaining deposit, incubate to penicillin allergy.
overnight and subculture in the same way. In At the end of a course of therapy with penicillin,
the absence of visible meningo­cocci, glucose it is important to give eradicative treatment with
broth may be added to the remaining sed­ rifampicin or ciprofloxacin.
iment of the centrifuged deposit to facilitate
the isolation. Prophylaxis
v. Biochemical reactions 1. Chemoprophylaxis: Minocycline and rifampin
Sugar utilization tests or commercial kits are have been used effectively for antibiotic-
used to identify any gram-negative diplococci. mediated chemoprophylaxis. Ciprofloxacin is
Chapter 27: Neisseria and Moraxella  | 191
widely used as a prophylactic for adolescents round, translucent, convex or slightly umbonate,
and adults as a single, oral dose. with finely granular surface and lobate margins
2. Immunoprophylaxis: A polyvalent vaccine after incubation for 24 hours..They are soft and
effective against serogroups A, C, Y, and W135, easily emulsifiable. After 48 hours, the colonies are
which can be administered to children older larger (1.5–2.5 mm), sometimes with a crenated
than 2 years, has been developed. The vaccine margin and an opaque raised center.
cannot be administered to children in younger Types of gonococci: Kellogg divided gonococci
age groups because they do not respond to into four types (T1–T4) on the basis of colonial
polysaccharide antigens. The immunity is appearance, auto-agglutinability and virulence.
group specific. There is no Group B vaccine
available at present. Types 1 and 2 form small brown colonies and bear
nu­merous fimbriae (piliated types P1 and P2). They
are auto-agglutinable and virulent.
Neisseria gonorrhoeae
(gonococcus) Types T3 and T4 are nonpiliated (P–), form smooth
suspensions and are avirulent.
N. gonorrhoeae causes the venereal disease Tl and T2 types are also known as P+ and P++,
gonorrhea. Gonococci resemble meningococ­ci respectively, while T3 and T4 are known as P–.
very closely in many properties.
Biochemical Reactions
Morphology
Gonococcus is oxidase positive and resembles
Morphology and staining of N. gonorrhoeae are menin­g ococci except in the fermentation of
identical to those of N. meningitidis. In smears maltose. Gono­cocci ferment only glucose and
from the urethral discharge in acute gonorrhea, not maltose and neither species ferments lactose
the organism appears as a Diplococcus with the or sucrose. This can be remembered by G for
adjacent sides concave (Fig. 27.2), being typically Gonococcus and M+G for Meningococcus.
kidney-shaped. It is found predominantly within
the polymorphs, some cells containing as many
Antigenic Structure
as a hundred cocci.
The structure of N. gonorrhoeae is typical of gram-
Cultural Characterstics negative bacteria. The surface struc­tures include
Gonococci are more diffi­c ult to grow than the following:
meningococci They are aerobic but may grow 1. Pili: Pili are hair-like appendages that extend
anaerobically also. Growth occurs best at pH 7.0– up to several mi­crometers from the gonococcal
7.4 and at a temperature of 35–36°C. It is es­sential surface. They act as virulence factors by
to provide 5–10% CO2. promoting attachment to host cells and
They grow well on chocolate agar and Mueller– inhibiting phagocytosis. The pili are composed
Hinton agar. A popu­lar selective medium is of repeating protein subunits (pilins). Pili
the Thayer–Martin medium (chocolate agar undergo antigenic and phase variation.
containing vancomycin, colistin and nystatin) 2. Por proteins (protein I): The Por proteins
which inhibits most contaminants, including (formerly pro­tein I) are porin proteins that form
nonpathogenic Neisseria. Colonies are small, pores or channels in the outer membrane. Two
classes of Por proteins (PorA and PorB), have
been identified. Anyone strain carries only
either IA or IB but not both.
3. Opa protein (protein II): These proteins facili­
tate bacterial adherence to each other and to
eukaryotic cells and also for the clumping of
cocci seen in urethral exudate smears.
4. Rmp (protein III): These proteins stimulate
antibodies that block serum bactericidal
activity against N. gonorrhoeae.
5. Lipooligosaccharide (LOS): This antigen
possesses endotoxic activity.
6. Other proteins: Other important gonococcal
proteins are IgA1 protease, β-lactamase,
Fig. 27.2: N. gonorrhoeae in urethral pus. Inset— enlarged which degrades penicillin and Fbp (iron-
view showing diplococci with adjacent surfaces concave binding protein).

www.ebook3000.com
192  |  Section 3: Systemic Bacteriology
Resistance Once ver y common, this has been
Gonococcus is a very delicate organ­ism, readily controlled by the practice of instilling 1%
killed by drying, soap and water, and many other silver nitrate solution into the eyes of all
cleansing or antiseptic agents at their correct use- newborn babies (Crede’s method).
dilution. Organisms may remain viable for a day or ii. Vulvovaginitis: In prepubertal girls,
so in pus contaminating linen or other fabrics. In vulvovaginitis may be caused by gonococci.
cultures, the coccus dies in 3–4 days at room tem­ This occurs either in conditions of poor
perature. Freeze-drying is the most reli­able method hygiene or by sexual abuse.
for long-term storage of gonococci but storage at
–70°C or in liquid nitrogen may be more convenient Epidemiology
for intermediate storage. Gonorrhea occurs only in humans. It has no other
known reservoir. N. gonorrhoeae is transmitted
Pathogenesis primarily by sexual contact. A higher incidence of
Gonorrhea gonorrhea has been observed in persons belonging
to blood group B. The basis for this is not known.
Gonorrhea is a venereal disease. The disease is
acquired by sexual contact. The incubation period
is 2–8 days. Laboratory Diagnosis
A. Disease in men: The most common clinical Diagnosis can be established readily in the acute
presentation is acute urethritis in the male. stage but chronic cases some­times present great
Dysuria and a purulent penile discharge difficulties.
make most sufferers seek treatment rapidly.
The infection extends along the urethra to the 1. Specimens
prostate, seminal vesicles and epididymis. A. Specimens in Men
Chronic urethritis may lead to stricture 1. Urethra: In men, urethral samples usually suffice
formation. The infection may spread to the (with rectal cultures in homosexual males).
periurethral tissues, causing abscesses and In acute gonorrhea, the urethral discharge
multiple discharging sinuses (‘watercan contains gonococci in large num­b ers. The
perineum’). meatus is cleaned with a gauze soaked in saline
B. Disease in women: Asymptomatic carriage and a sample of the discharge collected with a
in women is common, espe­c ially in the platinum loop for culture, or directly on slide for
endocervical canal. The infection may extend smears. Purulent discharge may be exp­ressed at
to Bartholin’s glands, endometrium and the anterior urethra and collected with a swab.
fallopian tubes to give rise to acute salpingitis, In chronic infections, there may not be any
which may be followed by pelvic inflammatory ure­t hral discharge. The morning drop of
disease and a high probability of sterility. secretion may be examined or some exudate
Peritoneal spread occasionally occurs and may may be obtained after prostatic massage. It may
produce a perihepatic inflammation (Fitz– also be possible to demon­strate gonococci in
Hugh–Curtis syndrome). the centrifuged deposits of urine in cases where
Proctitis occurs in both sexes. Gonococcal no urethral discharge is available.
pharyngitis may follow oro­genital contact in 2. Anal canal: In homosexual males.
either sex. Conjunctivitis may occur usually by
B. Specimens in Women
autoinoculation with fingers.
1. Endocervical swab: In women, ure­t hral,
C. Disseminated gonococcal disease: Blood cervical and rectal specimens should always be
invasion may occur from the primary site of examined. A single well taken endocervical swab
infection and may lead to metastatic lesions will detect approximately 90% of gonococcal
such as arthritis, ulcerative endocarditis and infec­tions in women. A high vaginal swab is
very rarely meningitis. not suitable. Throat infection also occurs and
D. Disease in childern should be sought where appropriate.
i. Ophthalmic neonatorum: A nonvenereal 2. Urethral
infection is ophthalmia neonatorum in 3. Anal canal and
the newborn, in which the eyes are coated 4. Throat.
with gonococci as the baby passes down
the birth canal. A severe purulent eye C. Blood, Swabs of skin lesions, or pus aspirated
from a joint.
discharge with periorbital edema occurs
within a few days of birth. If untreated, D. Conjunctival Swab: Particularly in neonatal
ophthalmia leads rapidly to blindness. ophthalmia.
Chapter 27: Neisseria and Moraxella  | 193
E. Urine Specimen: Any urine specimen showing 6. Genetic Probes
gram-negative diplococci in a Gram stain should Probes specific for the nucleic acids of N. gonorr­
be cultured on an appropriate selective medium. hoeae have been developed for the direct detec­tion
of bacteria in clinical specimens.
2. Transport
For culture, specimens should be inoculated on 7. Serological Diagnosis
prewarmed plates, immediately on collection. If No serological test has been found useful for
this is not possible, specimens should be collected routine diagnostic purposes.
with char­coal impregnated swabs and sent to the
laboratory in Stuart’s transport medium. Treatment
Penicillin is no longer the antibiotic of choice for
3. Direct Microscopy treatment of gonorrhea since the development and
Do the Gram staining shows characteristic kidney- widespread use of penicillin, gonococcal resistance
shaped gram-negative diplococci lying within to penicillin has gradually risen, owing to the selec-
polymorphonuclear leukocytes with a few extra­ tion of chromosomal mutants, so that many strains
cellular. Approximately 95% of infected men will now require high concentrations of peni­cillin G for
yield a positive smear. It has to be emphasized that inhibition (MIC 2 µg/mL).
diagnosis of gonorrhea by smear examination is Penicillinase ­p roducing gonococci (PPNG):
unreliable in women as some of the normal genital In 1976, gonococci producing β-lactamase
flora have an essentially similar morphology. (penicillinase) have appeared, rendering penicillin
Immunologic methods employing monoclonal treatment ineffective. Penicillinase production, in
anti­bodies may be used for the identification of gonococci, is plasmid-mediated.
N. g onorrho eae. These methods include
Chromosomally-mediated resistance (CMRNG):
coagglutination and fluorescent antibody testing.
This chromosomally-mediated resistance
(CMRNG) is not limited only to penicillin but
4. Culture extends to tetracyclines, erythromycin, and
In acute gonorrhea, cul­tures can be obtained aminoglycosides.
readily on chocolate agar or Mueller–Hinton agar Empirical therapy: Currently, the Centers for Dis­
incubated at 35–36°C under 5–10% CO2. In chronic ease Control and Prevention (CDC) recommends
cases, where mixed infection is usual and in the that ceftriaxone, cefixime, ciprofloxacin, or
examination of lesions, such as proctitis; however, ofloxacin be used as the initial therapy for cases
it is better to use a selective medi­um such as the of uncomplicated gonorrhea. Doxycycline or
Thayer–Martin medium. Examine plates after 24 azithromycin should be added for infections
hours incubation and the growth is identified by complicated by dual infections with Chlamydia.
morphology and biochemical reactions. Incubation
of primary isolation plates is continued for 48 hours. Prophylaxis
Colonies are small, round, translucent, convex
Control of gonorrhea consists of early detection of
or slightly umbonate, with finely granular surface
cases, contact tracing, health education and other
and lobate margins. They are soft and easily
general measures.
emulsifiable. After 48 hours, the colonies are larger
As even clinical disease does not confer any
(1.5–2.5 mm), sometimes with a crenated margin
immunity, vaccination has no place in prophylaxis.
and an opaque raised center.
Smear is made from the colony and Gram Nongonococcal (nonspecific)
staining is done. Gonococci are gram-negative
cocci arranged in pairs (diplococci) with adjacent
urethritis
sides concave (pear or bean shaped). Nongonococcal urethritis (NGU), also known
as nonspecific urethritis (NSU), refers to chronic
5. Identification urethritis where gonococci cannot be demonstrated.
N. gonorrhoeae is identified preliminarily on the
basis of the isolation of oxidase-positive, gram-
Causative Agents
negative diplo­cocci that grow on chocolate blood The most important causative agents are as follows:
agar or on media that are selective for pathogenic
Neisseria species.N gonorrhoeae is oxidase positive. A. Bacterial
It ferments glucose with acid only. It does not ∙∙ Chlamydia trachomatis
ferment maltose unlike meningococci. ∙∙ Ureaplasma urealyticum

www.ebook3000.com
194  |  Section 3: Systemic Bacteriology
∙∙ Mycoplasma hominis N. flavescens, N. catarrhalis) have been reported
∙∙ Gardnerella vaginalis occasionally as having caused meningitis. N.
∙∙ Acinetobacter wolfii sicca, N. subflava, N.cinera, N. mucosa, and N.
∙∙ Acinetobacter calcoaceticus. flavescens are also members of the normal flora of
the respiratory tract, because of its carbohydrate
B. Viral fermentation pattern.
∙∙ Herpes virus
∙∙ Cytomegalovirus. Moraxella
C. Fungi The genus Moraxella is a member of the family
Neisseriaceae.
∙∙ Candida albicans.
Species: The Moraxella spp. of medical importance
D. Protozoa are M. lacunata, M. catarrh­alis, M. osloensis, M.
∙∙ Trichomonas vaginalis. phenylpyruvica, M. atlantae and M. nonliquefaciens.

E. By Mechanical or chemical irritation Moraxella (Branhamella) Catarrhalis


Diagnosis Morphology
1. Demonstration of a leukocyte exudate They are oval gram-negative cocci. They are non-
2. Exclusion of urethral gonorrhea by Gram stain capsulate and non-motile.
and culture.
3. Several rapid tests for detecting Chlamydia in Cultural Characteristics
urine spec­imens are also available. They are aerobes. Most strains grow on nutrient
agar, blood agar or chocolate agar.
Treatment
Treatment is with tetracycline, doxycy­c line, Biochemical Reactions
erythromycin, or sulfisoxazole. ∙∙ It is oxidase positive and catalase positive
∙∙ It does not produce acid from glu­cose, maltose,
Commensal neisseriae sucrose, lactose or fructose
Several species of Neisseriae inhabit the normal ∙∙ It reduces nitrate to nitrite.
respi­ratory tract. They are regularly found in the
throat, nose and mouth and, less frequently, on Pathogenesis
the genital mucosae. The characteristic features They form part of the normal pharyngeal flora
of some of the common species are listed in but can cause respiratory infections, including
Table 27.1. Their patho­g enic significance is otitis media, sinusitis, tracheobronchitis and
uncertain though some of them (for example, pneumonia.

Table 27. 1  Differential characteristics of commonly isolated Neisseriae


Species Colonies Growth Fermentation Serological
classification
On At 22°C Glucose Maltose Sucrose
nutrient
agar
N. menigitidis Round, smooth, shiny, – – A A – Thirteen antigenic
creamy consistency Groups
N. gonorrhoeae Same as above, but – – A – – Antigenically
smaller and more heterogeneous
opalescent
N. flavescens Resemble meningococ- + + – – – Antigenically distinct
cus but pigmented homogeneous group
yellow
N. sicca Small, dry, opaque, + + A A A Autoagglutinable
wrinkled, brittle
N. catarrhalis Variable, smooth and + + – – – Autoagglutinable
(Branhamella translucent or adherent
catarrhalis and opaque, not easily
emulsifiable
Chapter 27: Neisseria and Moraxella  | 195
Treatment HACEK group of oral bacteria: Kingella species
As many strains produce beta-lactamases, are included in the so-called HACEK group of
penicillins are not useful in treatment unless given oral bacteria (Haemophilus spp, Actinobacillus
in combination with clavulanate or sulbactam. actinomycetemcomitans, Cardiobacterium
hominis, Eikenella corrodens and Kingella
spp). These organisms are known to colonize
Moraxella lacunata (Morax–Axenfeld
endovascular tissue and produce vegetations on
Bacillus) heart valves. In common with some other Gram-
Formerly this was included in the genus Haemo­ negative rods, such as Cardiobacterium hominis,
philus, but it has been separated into the genus Eikenella corrodens and Actinobacillus actino­
Moraxella as it does not require X or V factor. M. mycetemcomitans, Kingella species (usually K.
lacunata was first reported as the cause of angular kingae) are sometimes found in endocarditis. They
conjunctivitis by Morax (1896) and Axenfeld. have also been implicated in joint infections.
Hence, it is also known as the Morax–Axenfeld
bacillus.
Key Points
Morphology Neisseria
These are short, plump, gram-negative bacilli ™™ Important spe­c ies of the genus Neisseria are N.
meningitides and N. gonorrhoeae
usually arranged in pairs. They are nonflagellated
but have been reported to be sluggishly motile. Neisseria meningitidis
™™ Gram-negative diplococci with fastidious growth
require­ments
Culture ™™ Growth is facilitated by 5–10% CO 2 and high
They are strictly aerobes, grow on ordinary humidity and grows best at 35°C to 37°C. Blood agar,
media but require blood or serum for growth. On chocolate agar and Mueller–Hinton starch casein
Loeffler’s serum slope, the colonies form pit or hydrolysate agar are the media common­ly used for
culturing meningococci. Modified Thayer–Martin is
lacunae (hence the name lacunata). a useful selective medium
™™ Oxidase and catalase positive; acid produced from
Biochemical Reactions glucose and maltose oxidatively
They do not ferment sugars and oxidase and ™™ Antigenic classification: Based on their capsular
catalase positive. polysaccaride antigens, meningococci are classified
into at least 13 serogroups.
™™ Diseases: Meningitis, meningoencephalitis, bactere­
Pathogenesis
mia, pneumonia, arthritis, urethritis
The moraxellas occur as components of the normal ™™ For immunoprophylaxis, vaccination is used only for
flora of the upper respiratory tract, the conjunctiva, serogroups A, C, Y, and W135.
the skin and the genital tract. N. gonorrhoeae
1. Moraxella lacunata causes a form of purulent ™™ N. gonorrhoeae appears as a Diplococcus with the
conjunctivitis classically presenting as an adjacent sides concave, being typically kidney
shaped
angular blepharoconjunc­tivitis. A few species
™™ It has fastidious growth require­ments. They grow
of Moraxella cause endophthalmitis, sinusitis, well on chocolate agar and Mueller–Hinton agar.
conjunctivitis, bronchitis, endocarditis, A popular selective medium is the Thayer–Martin
meningitis septicemia and sytemic infections. medium
™™ Diseases: It causes urethritis, cervicitis, salpingitis,
Treatment pelvic inflammatory disease, proctitis, bacteremia,
arthritis, conjunctivitis, pharyngitis
M. lacunata is very sensitive to zinc salts. They are ™™ Treatment: Gonococci producing β-lactamase
sensitive to penicillin and most other antibiotics. (penicillinase) have appeared called penicillinase
­p roducing gonococci (PPNG) and is plasmid
Kingella mediated. Chromosomally mediated resistance
(CMRNG) have also been iso­lated
The genus Kingella, comprising some species of ™™ Nongonococcal urethritis (NGU), refers to chronic
oxi­dase positive, nonmotile, gram-negative rods, urethritis where gonococci cannot be demonstrated
with a tendency to occur as coccobacillary and ™™ Commensal Neisseriae: They are regularly found
in the throat, nose and mouth. Their patho­genic
diplococcal forms was formerly grouped under the
significance is uncertain (for example, N. flavescens,
genus Moraxella. The genus con­tains three species N. catarrhalis)
(K. kingae, K. indologenes and K. denitrificans). ™™ Morexella (Branhamella) catarrhalis: They are now
K. kingae is the most commonly isolated recognized as an important respiratory pathogen
species. It is part of the normal oral flora and has ™™ Kingella: K. kingae is part of the normal oral flora
been associated with endocarditis and infections and has been associated with endocarditis and
infections of bones, joints and tendons.
of bones, joints and tendons.

www.ebook3000.com
196  |  Section 3: Systemic Bacteriology
Important questions 4. Neisseria meningitidis shows all the following
characteristics except:
1. Describe the morphology, pathogenicity and a. Oxidase test positive
laboratory diagnosis of meningococcal meningitis. b. Catalase test positive
2. Discuss laboratory diagnosis of gonorrhea. c. Ferments maltose with production of acid
3. Write short notes on: d. Ferments sucrose with production of acid
a. Antigenic structure of Neisseria gonorrhoeae. 5. Waterhouse–Friderichsen syndrome is caused by:
b. Nongonococcal urethritis (or) nonspecific ure- a. Naieisseria meningitidis
thritis b. Leptospira
c. Moraxella (Branhamella) catarrhalis. c. Streptococcus pyogenes
d. Neisseria gonorrhoeae
Multiple choice questions (MCQs) 6. Causative agent of nongonococcal urethritis is
caused by:
1. All of the following bacteria are oxidase positive a. Chlamydia trachomatis
except: b. Ureaplasma urealytium
a. Neisseria gonorrhoeae c. Mycoplasma hominis
b. Neisseria meningitidis d. All of the above
c. Vibrio cholerae 7. Following statements are true for quadrivalent
d. Enterobacter meningococcal polysaccharide vaccine (MPSV4)
2. The most common infective cause of vaginal except:
discharge in a sexually promiscuous female is: a. The vaccine is given intramuscularly
a. Trichomonas vaginalis b. Prevents disease caused by A, C, Y, and W135
b. Gardnerella vaginalis serogroups of meningococci
c. Neisseria gonorrhoeae c. Indicated for at risk population during out-
break of meningococcal infection
d. Candida albicans
d. Produce good antibody response in children
3. The specimen of choice for isolation of gonococci
below 2 years of age
from women with gonorrhea is:
a. Vaginal swab b. Cervical swab Answers (MCQs)
c. Urethral swab d. Urine 1. d; 2. c; 3. a; 4. d; 5. a; 6. d; 7. d
28
Chapter

Corynebacterium

Learning Objectives

After reading and studying this chapter, you should ∙∙ Discuss diphtheria toxin
be able to: ∙∙ Discuss laboratory diagnosis of diphtheria
∙∙ Describe morphology, cultural characteristics, ∙∙ Toxigenicity tests/virulence tests of C. diphtheriae.
biochemical reactions, toxin production and ∙∙ Describe the following: Schick test; DPT vaccine or
pathogenesis of diphtheria triple vaccine; diphtheroids
∙∙ Differentiate three biotypes: Gravis, intermedius ∙∙ Differentiate between C. diphtheriae and diphthe­
and mitis of C. diphtheriae roids.

Introduction groups or as individual cells lying at sharp angles


to another, resembling the letters V or L. This
Corynebacteria are gram-positive, nonacid fast, particular arrangement with C. diphtheriae has
nonmotile rods with irregularly stained segments, been called the Chinese letter or cuneiform
and sometimes granules. They frequently show arrangement (Fig. 28.1). This is due to the
club-shaped swellings and hence the name incomplete separation of the daughter cells after
Corynebacteria (from coryne, meaning club). binary fission.
The diphtheria bacillus was first observed and This organism has granular and uneven
described by Klebs (1883) but was first cultivated staining. When stained with methylene blue
by Loeffler (1884). It is hence known as the or toluidine blue, the granules in the cell stain
Klebs–Loeffler bacillus. Roux and Yersin (1888) metachromatically reddish–purple. These granules
discovered the diphtheria exotoxin and established are known as metachromatic granules, volutin
its pathogenic effect. The antitoxin was described granules or Babes Ernst granules. They are
by von Behring (1890) who was awarded nobel often situated at the poles of the bacilli and
prize for this work. are called polar bodies. Special stains, such as
Diseases: The major disease caused by C. Albert’s, Neisser’s and Ponder’s have been devised
diphtheriae is diphthe­ria (greek, diphtheria, for demonstrating the granules clearly. With
“leathery skin,” referring to the pseudomembrane
that initially forms on the pharynx).

Corynebacterium diphtheriae
Morphology
They are, thin, slender gram-positive bacilli but
are decolorized easily, particularly in old cultures,
measuring approximately 3–6 μm × 0.6–0.8 μm.
They have a tendency to clubbing at one or both
ends. They are highly pleomorphic. They are non-
motile, non-spore forming, and nonacid fast.
The bacilli are arranged in a characteristic
fashion in smears. They are usually seen in pairs, Fig. 28.1: Corynebacterium diphtheriae showing
palisades (resembling stakes of a fence) or small metachromatic granules and Chinese letter arrangement

www.ebook3000.com
198  |  Section 3: Systemic Bacteriology
Albert’s stain, the granules stain bluish black and of cystine to a tellurite ­containing medium
the protoplasm green. The granules represent (Tinsdale’s medium) has greatly helped the
accumulation of polymerized polyphosphates. isolation of diphtheria bacilli. The growth
The granules formation is best seen on Loeffler’s of diphtheria bacilli may be delayed on the
serum slope. tellurite medium and colonies may take two
days to appear.
Cultural Characteristics Based on colo­nial morphology on the tellurite
medium and other properties, Mc Leod and
C. diphtheriae is an aerobe and facultative anaerobe;
Anderson described three different biotypes:
the optimum temperature for growth is 37°C (range
Gravis, intermedius and mitis (Table 28.1). The
15–40°C) and optimum pH 7.2. It can grow on
names were originally proposed to relate to the
ordinary nutrient agar, but its growth is improved
clinical severity of the disease produced by the
by the pres­ence of animal proteins such as blood
three types—gravis, causing the most serious, and
or serum. Two media are useful for this purpose:
mitis the mildest variety, with intermedius being
1. Loeffler’s serum slope: Diphtheria bacilli
responsible for disease of intermediate severity.
grow on LoeffIer’s serum slope very rapidly
and colonies can be seen in 6–8 hours, long
before other bacteria grow. Colonies are at first Biochemical Reactions
small, circular white opaque disks but enlarge C. diphtheriae ferments glucose and maltose
on continued incubation and may acquire a with the production of acid (but no gas) but not
distinct yellow tint. lactose, mannitol, trehalose or sucrose. Starch and
2. Tellurite blood agar: The addition of potassium glyco­gen are used for biochemical differentiation
tellurite (0.03–0.04%) makes the medium of three biotypes of C. diphtheriae (Table 28.1).
selec­tive for Corynebacteria by inhibiting most Gravis strains utilize glycogen and starch, while
other pathogenic and commensal bacteria. On mitis and inter­medius do not. Fermentation of
this medium, C. diphtheriae give grey/black, sugars are usually done in Hiss’s serum peptone
shiny or dull black colonies. The addition water medium.
Table 28.1  Differentiating features of Corynebacterium diphtheriae
Gravis Intermedius Mitis
1. Morphology i. Usually short rods, with i. Long barred forms with i. Long, curved, rods
uniform staining clubbed ends; ii. Prominent granules
ii. Few or no granules. ii. Poor granulation iii. Pleomorphic
iii. P leomorphism (some iii. Very pleomorphic
degree), with irregularly
barred, snow-shoe and
tear­drop forms
2. Colony on tellurite i. In 18 hours: Colony is i. 18 hour: Colony small, I i. Size variable, shiny black.
blood agar 1–2 mm in size, greyish mm in size, misty.
black centre, paler,
semitranslucent periphery
and commencing ii. i n 48 hours Does not ii. In 2–3 days: Colonies
crenation of edge. enlarge, dull granular become flat, with
ii. In 2–3 days: 3–5 mm in centre with smoother, more a central elevation
size, flat colony with raised glistening periphery and a ‘poached egg’ colony
dark centre and crenated lighter ring near the edge -
edge with radial striation - ‘frog’s egg’ colony
‘daisy head’ colony
3. Consistency of colonies i. Brittle, moves as a whole Intermediate between (i) Soft, buttery, (ii) Easily
on the plate like ‘cold gravis and mitis emulsifiable
margarine’.
ii. Not easily picked out or
emulsifiable
4. Hemolysis Variable Nonhemolytic Usually hemolytic
5. Growth in broth Surface pellicle. Deposit Turbidity in 24 hours, Turbidity diffuse with soft
granular. clearing in 48 hours, with pellicle later
Turbidity little or no fine granular sediment
6. Glycogen and starch Positive Negative Negative
fermentation
7. Toxigenic strains Almost 100% 95–99% 80–85%
8. Virulence Severe Moderate Mild
9. Predominant strains Epidemic areas Epidemic areas Endemic areas
Chapter 28: Corynebacterium  | 199
C. diphtheriae is H2S positive and reduces and mediates the entry of fragment A into the
nitrate to nitrite. It does not liquefy gelatin or cytoplasm. The antibody to fragment B prevents the
hydrolyze urea or form phosphatase. binding of toxin to cells and is thus protective. The
Pyrazinamidase (PYZ) test: In pyrazinamidase toxin is heat labile. It is extremely potent (0.0001 mg
(PYZ) test, pyrazinamide is converted into pyrazinoic kills a guinea pig of 250 g weight). It is converted into
acid by the organisms which produce pyrazinamidase toxoid by heat (at 37°C for 4–6 weeks), treatment
(PYZ). This test is helpful to distinguish ‘C. diphtheriae’ with 0.2–0.4% formalin or by acidic pH. The toxoid
(PYZ-negative) from other Corynebacterium species is a toxin that has lost its toxicity but has retained the
(mostly PYZ-positive). antigenicity. It is capable of producing antitoxin. It
has a special affinity for certain tissues such as the
myocardium, adrenals and nerve endings.
Toxin
Toxigenic strains of C. diphtheriae produce a a Mode of Action
very powerful exotoxin. The toxicity observed
The diphtheria toxin acts by inhibiting protein
in diphtheria is directly attributed to the toxin
synthesis. It inhibits polypeptide chain elongation
secreted by the bacteria at the site of infection.
in the presence of nicotinamide adenosine
dinucleotide (NAD) by inactivating elongation
Synthesis factor 2 (EF-2), an enzyme required for elongation
Almost all strains of gravis, 95-99% of intermedius of polypeptide chains on ribosomes. Inhibition of
and 80-85% of mitis produce this toxin. Strains of protein synthesis is probably responsible for both
all three types are invariably virulent when isolated the necrotic and neurotoxic effects of the toxin.
from acute cases. Avirulent strains are common NAD+ + EF–2 = ADPR–EF–2 + nicotinamide + H+
among convalescents, contacts and carriers. The Active Inactive
strain almost universally used for toxin production
is the ‘Park Williams 8’ strain, which has been Resistance
variously described as a mitis (Topley and Wilson)
and an inter­medius strain (Cruickshank). They are readily killed, however, by a 1-minute
exposure to100°C or a 10 minute exposure to 58°C.
They are susceptible to most of the routinely used
Lysogeny and toxin Production
disinfectants. It remains alive for weeks in dust and
The toxigenicity of the diphtheria bacillus de­pends on fomites when dry and protected from sunlight.
on the presence in it of corynephages (tox +), It is susceptible to penicillin, erythromycin and
which act as the genetic determinant controlling broad spectrum antibiotics.
toxin production. Nontoxigenic strains can be
converted to tox+ by infection with the appropriate Antigenic Structure
bacteriophage. This is known as lysogenic or phage
con­version. The bacillus loses the toxigenicity Diphtheria bacilli possess three distinct antigens:
when it is cured of its phage, as by growing it in the 1. A deep-seated antigen found in all Coryne­
presence of antiphage serum. bacterial species
2. A heat-labile protein (K-antigen)
3. A heat-stable polysaccharide (O-antigen).
Iron for toxin Production
Toxin production is also influenced by the Serotypes: On the basis of agglutination reaction,
concentration of iron in the medium. The optimum biotypes gravis, intermedius and mitis have been
level of iron for toxin production is 0.1 mg per liter, divided into 13, 4 and 40 serotypes, respectively.
while a concentration of 0.5 mg per liter inhibits
the forma­tion of toxin. The toxin is released in Bacteriophage Typing
significant amounts only when the available iron The susceptibility of C. diphtheriae to bacterio­
in the culture medium is exhausted. phage strains has been most comprehensively
studied; 22 phages were used to type the gravis
Properties of toxin strains into 14 types, the intermedius into 3 and the
Diphtheria toxin is an iron-free, crystalline, heat mitis into 4. An additional set of 33 phages has also
labile protein. The diphtheria toxin is a protein and been used. The only other corynebacteria re­ported
has a molecular weight of about 62000. It consists as susceptible to the diphtheria typing phages are
of two fragments A (active) and B (binding) of C. ulcerans and C. pseudotuberculosis.
molecular weights 24000 and 38000 respectively.
Both fragments are required for toxicity. Fragment Pathogenesis
A has all the enzymatic activity whereas fragment The organism is carried in the upper respiratory tract
B is responsible for binding the toxin to the cells and spread by droplet infection or hand-to-mouth

www.ebook3000.com
200  |  Section 3: Systemic Bacteriology
contact. The incubation period of diphtheria is 2–5 sys­temic complications are less common than from
days, with a range of 1–10 days. Diphtheria, which upper respiratory infections with C. diphtheriae.
occurs in two forms (respiratory and cutaneous),
is found worldwide. Laboratory Diagnosis
Diagnostic laboratory tests serve to confirm the
A. Respiratory Diphtheria clinical impression and are of epidemiologic
The illness begins gradually and is characterized significance but not for the treatment of individual
by low-grade fever, malaise, and a mild sore throat. cases. Specific treatment should be instituted
The most common site of infection is the tonsils immediately on suspicion of diph­theria without
or pharynx. The organisms rapidly multiply on waiting for laboratory tests. Any delay may be
the epithelial cells, and the toxigenic strains of C. fatal. Laboratory diagnosis consists of isolation
diphtheriae pro­duce toxin locally, causing tissue of the diphtheria bacillus and demonstration of
necrosis and exudate formation triggering an its toxicity.
inflammatory reaction. This combination of cell 1. Specimens
necrosis and exudate forms a tough gray to white
Swabs from the nose, throat, or other suspected
pseudomembrane, which attaches to the tissues-
le­sions must be obtained before antimicrobial
commonly over the tonsils, pharynx, or lar­ynx. Any
drugs are administered. In suspected cases,
attempt to remove the pseudomembrane results
whether of faucial or nasal diph­t heria, swabs
in bleeding. In nasopharyngeal infection, the
should be taken both from the throat and from the
pseudomembrane may involve nasal mucosa, the
nose, and preferably two swabs from the site most
pharyngeal wall and the soft palate. In this form,
affected. Swabs should also be taken from skin
oedema involving the cervical lymph glands may
lesions and wounds where diphtheritic infection is
occur in the anterior tissues of the neck, a condition
suspected, and both throat and nose swabs should
known as bullneck diphtheria. Laryngeal
be taken from suspected carriers.
involvement leads to obstruction of the larynx and
lower airways. 2. Microscopy
Systemic effects Direct microscopy of a smear is unreliable since
The toxin also is absorbed and can produce a vari­ C. diphtheriae is morphologically similar to
ety of systemic effects involving the kidneys, heart, other coryneforms. Smears stained with alkaline
and nervous system, although all tissues possess methylene blue or Gram’s stain show beaded rods
the receptor for the toxin and may be affected. in typical arrangement. Hence smear examination
Intoxication takes the form of myocarditis and alone is not sufficient for diagnosing diphtheria
peripheral neuritis, and may be associated with but is important in identifying Vincent’s angina.
thrombocytopenia. Visual disturbance, difficulty in For this, a Gram or Leishman stained smear is
swallow­ing and paralysis of the arms and legs also examined for Vincent’s spirochetes and fusiform
occur but usually resolve spontaneously. Complete bacilli. Toxigen­ic diphtheria bacilli may be
heart block may result from myocarditis. Death is identified in smears by immunofluorescence.
most commonly due to congestive heart failure and
cardiac arrhythmias. 3. Culture
The swab should be in­oculated on Loff­ler’s serum
Complications slope, tellurite blood agar, and blood agar. The
The common complications are as follows: cultures should be incubated aerobically at 37°C.
1. Asphyxia due to mechanical obstruction of the Unless the swab can be inoculated promptly, it
res­piratory passage by the pseudomembrane should be kept moistened with sterile horse serum
for which an emergency tracheostomy may so the bacilli will remain vi­able.
become necessary. i. Loeffler’s serum slope: After incubation for 6
2. Acute circulatorv failure, which may be hours or overnight, make a smear of growth from
peripheral or cardiac. all parts of the slope mixed in the condensation
3. Postdiphtheritic paralysis, which typically water, stain by the Albert-Laybourn method and
occurs in the third or fourth week of the disease; look for the presence of slender green-stained
palatine and ciliary but not pupillary paralysis bacilli containing the purple-black granules
is characteristic, and spontaneous recovery is characteristic of C. diphtheriae.
the rule. ii. Tellurite blood agar: Blood tellurite agar is
4. Septic, such as pneumonia and otitis media. examined after 24 hours and after 48 hours,
as growth may sometimes be delayed.
B. Cutaneous Diphtheria iii. Blood agar: It is used for differentiating
In cutaneous diphtheria, which is prevalent in the streptococcal or staphylococcal pharyngitis,
tropics, the toxin also is absorbed systemically, but which may simulate diphtheria.
Chapter 28: Corynebacterium  | 201
4. Identification Tests Advantages of the intracutaneous test:
Identification is based on carbohydrate ferment­ a. The animals do not die
ation reactions and enzymatic activities. C. b. As many as ten strains can be tested at a time
diphtheriae ferments glucose and maltose, on a rabbit.
producing acid but not gas, and is catalase positive. B. In vitro Test
It reduces nitrate to nitrite and is nonmotile. i. Elek’s gel precipitation test: The in vitro
Commercial kits such as the API Coryne strip diphtheria toxin detection procedure is an
provide a reliable identification. immunodiffusion test first described by Elek.
Procedure: A rectangular strip of filter paper
5. Virulence Tests impregnated with diphtheria antitoxin (1000 units/
Such tests are re­ally tests for toxigenicity of an ml) is placed on the surface of a 20% normal horse
isolated diphtheria-like organism. Diagnosis of serum agar in a Petri dish while the medium is still
diphtheria depends on showing that the isolate fluid. When the agar has set, the surface is dried.
produces diphtheria toxin. Virulence testing may The plate should be streaked with the test strain as
be by in vivo or in vitro methods. well as the control positive and negative strains at
A. In vivo tests right angles to the strip in a single straight line and
i. Subcutaneous test parallel to each other. The plate is incubated at 37°C
and examined after 24 and 48 hours.
ii. Intracutaneous test
B. In vitro test Interpretation: Toxins produced by the bacterial
growth will diffuse in the agar and where it meets the
i. Precipitation test
antitoxin at optimum concentration will produce a
ii. Tissue culture test line of precipitation (Fig. 28.2). A negative control
iii. Enzyme-linked immunosor­bent assays should be free of any line. No precipitate will form
iv. Polymerase chain reaction (PCR). in the case of nontoxigenic strains.
A. In vivo Tests ii. Tissue culture test : The toxigenicity of
i. Subcutaneous test: The growth from an diphtheria bacilli can be demonstrated by
overnight culture on Loeffler’s slope is incorporating the strains in the agar overlay of
emulsified in 2–4 mL broth and 1 mL of the cell culture monolayers. The toxin produced
emulsion injected subcutaneously into diffuses into the cells below and kills them.
two guinea pigs or rabbits, one of which has iii. Enzyme-linked immunosor­b ent assays
been protected with the diphtheria antitoxin (ELISA): Rapid, enzyme-linked immunosor­
(500–1000 units) 18–24 hours previously and bent assays and immunochromatographic
was used as control. If the strain is virulent, strip assays are also available for the detection
the unprotected animal will die within of diphtheria toxin.
four days. Postmortem examination would iv. Polymerase chain reaction (PCR): In addition,
show hemorrhage at the site of injection procedures for detecting the C. diphtheriae tox
and injected blood vessels, with typically gene by the polymerase chain reaction (PCR)
hemorrhagic adrenal necrosis. have been developed. The PCR assay can also
Simple and reliable subcutaneous be applied directly to clinical specimens.
toxigenicity tests in rabbits or larger guinea- Schick Test
pigs were used in the past, at a time when
laboratories had many isolates each day. Schick (1913) introduced an intradermal test
The method is not usually employed as it is (Schick test) for distinguishing between susceptible
wasteful of animals. and immune persons.
ii. Intracutaneous (Intradermal) test: The
broth emulsion of the culture is inoculated
intracutaneously into two guinea pigs (or
rabbits) so that each receives 0.1 mL in two
different sites. One animal acts as the control
and should receive antitoxin (500 units)
the previous day. After four hours the skin
test, the other is given 50 units of antitoxin
intraperitoneally in order to prevent death.
In the test animal, toxigenicity is indicated by
inflammatory reaction at the site of injection,
progressing to necrosis in 48–72 hours and in
the control animal no change. Fig. 28.2: Elek’s test

www.ebook3000.com
202  |  Section 3: Systemic Bacteriology
Principle DPT Vaccine: Diphtheria toxoid is usually given
This test depends upon the principle of toxin- in children as a trivalent preparation containing
antitoxin neutralization, in vivo, and the test is tetanus toxoid and pertussis vaccine also as the
carried out by injecting one Schick test dose of DPT or triple vaccine.
diph­theria toxin (0.2 mL containing 1/50 MLD)
intradermally on the anterior surface of the left Schedule of primary immunization: The schedule
forearm and a control injection in the right forearm of primary immunization of infants and children
contains a heat-inactivated dose (70°C for 30 consists of DPT given at the age of 6 weeks, 10
minutes) or preferably, purified diphtheria toxoid. weeks, 14 weeks and 16–24 months followed by
booster dose DT at the age of 5–6 years (school
Results entry).
Readings are taken after 1, 4 and 7 days. Four types
of reactions may occur:
B. Passive Immunization
1. Negative reaction: There is no reaction of any
kind in either arm. This indicates that the toxin This is an emergency measure to be employed
has been neutralized by the circulating antitoxin where susceptibles (nonimmunized) are exposed
and the person is immune to diphtheria and to in­fection, as when a case of diphtheria is
does not need immunization. admitted to general pediatric wards. It consists of
2. Positive reaction: In the test arm, there appears the subcutaneous administration of 500–1000 units
erythema and swelling at the site of inoculation in of antitoxin (antidiphtheritic serum, ADS). As this is
24–36 hours, reaching its maximum (1–5 cm) by a horse serum, precaution against hypersensitivity
the 4th to 7th days and then fading with superficial should be ­observed.
scaling and persistent brownish pigmentation. On
the control arm, there is no reaction. C. Combined Immunization
A positive Schick test indicates that the This consists of adminis­tration of the first dose
individual is susceptible to diphtheria and of adsorbed toxoid on, while ADS is given on the
little or no antitoxin (less than 0.01 unit/mL) is other arm, to be continued by the full course of
present. The subject is not immune and should active immunization since protection conferred
be immunized. by passive immunization is of short duration.
3. Pseudoreaction: There is erythema occurring Ideally, all cases that receive ADS prophylactically
within 6–24 hours and disappearing within four should receive combined immunization.
days. The reaction is the same on both arms.
This indicates that the individual is immune to
Treatment
diphtheria and also that he is hypersensitive to
one or more antigens in the toxin preparation. Specific treatment of diphtheria consists of
This individual does not need immunization. antitoxic and antibiotic therapy. Antitoxin
4. Combined reaction: Here the initial picture is should be given immediately as soon as clinical
that of pseudoreaction, but while the erythema diagnosis is made to neutralize the toxin being
in the control arm fades, within four days, it produced. The dosage recommen­ded is 20,000
pro­gresses in the test arm to a typical positive units intramuscularly for moderate cases and
reaction. This indicates that the individual is 50,000 to 100,000 units for serious cases, half the
susceptible to diphtheria and is sensitive to dose being given intravenously.
one or more antigens in the toxin preparation C. diphtheriae is sensitive to most antibiotics,
making immunization nec­essary but likely to including penicillin and erythro­mycin for the
induce reaction. treatment of patients as well as carriers. The
antibiotics do not neutralize circulating toxin.
Prophylaxis Penicillin-sensitive individuals can be given
The meth­ods of immunization available are active, erythromycin. Erythromycin is more active than
passive or combined. penicillin in the treatment of carriers.

A. Active Immunization OTHER MEDICALLY IMPORTANT


The preparations used for active immunization
are as follows:
CORYNEBACTERIA
1. Toxin-antitoxin mixture: It is not without hazards. The non-diphtheria corynebacteria are diverse
2. Single vaccines are less frequently used usually isolated from the environment and
3. Combined preparations: commensals of the skin and mucous membranes.
– DPT (diphtheria-pertussis-tetanus) vaccine The principal species involved and the main
– DT (diphtheria-tetanus toxoid) clinical syndromes associated with infection are
– dT (diphtheria-tetanus, adult type). shown in Table 28.2.
Chapter 28: Corynebacterium  | 203
Table 28.2  Medically important nondiphtheria corynebacteria and disease associations of these
corynebacteria
Organism Major habitat Disease association
C. ulcerans Human throat and Man: diphtheria (toxigenic strains), pharyngitis and wound
skin; animals; raw milk infection; cattle: mastitis
C. pseudotuberculosis Sheep, horses, goats Man: lymphadenitis
Animals: abscesses and abortion
C. jeikeium Skin Bacteremia, endocarditis; infection of foreign bodies and CSF
shunts
C. urealyticum Skin, urinary tract Urinary tract infection, pyelonephritis, endocarditis
C. amycolatum Man and animals Man: Bacteremia, endocarditis, peritonitis and wound
infection; cattle: mastitis
C. glucuranalyticum Urinary tract of man Urogenital tract infection
and animals
C. minutissimum Skin, urinary tract Erythrasma, bacteremia
C. striatum Respiratory tract, skin Respiratory tract infection, wound infection, bacteremia
C. pseudodiphtheriticum Respiratory tract Respiratory tract infection, endocarditis
Arcanobacterium haemolyticum Throat Pharyngitis, skin ulcers, endocarditis
Rhodococcus equi Animals, soil Pulmonary infection and soft tissue infection

1. Corynebacterium ulcerans history of intravenous drug abuse. It is the most


C. ulcerans has been isolated from patients with common cause of diphtheroid prosthetic valve
diphtheria-like illness. It resembles gravis type endocarditis in adults.
of C. diphtheriae but it liquefies gelatin, ferments
trehalose slowly and does not reduce nitrate to
5. Corynebacterium xerosis
nitrite. It is PYZ negative and urease positive. It C. xerosis is commonly found on skin and mucocut­
commonly produces diphtheria toxin as well as the a­neous sites. Human infection with C. xerosis is
separate toxin produced by C. pseudotuberculosis. rare, and affected patients are invariably immuno­
It is pathogenic to guinea pigs. The lesions suppressed.
produced resembling those caused by C. diphtheriae.
It can cause mastitis in cattle and man exposed to 6. Corynebacterium bovis
infected animals or milk may develop infection. C. bovis, commensal of cow’s udder, which may
cause bovine mastitis. Many of them cause
2. Corynebacterium pseudotuberculosis infections in immunocompromised patients.
(C. ovis)
Like C. ulcerans, C. pseudotuberculosis (Preisz-
Diphtheroids
Nocard bacillus) is primarily an animal pathogen Corynebacteria resembling C. diphtheriae, occur
and rarely infects man. Human infections mainly as normal commensals in the throat, skin and
occur in patients with animal (sheep) contact. other areas. These may be mistaken for diphtheria
It causes caseous lymphadenitis in sheep and bacilli and are known as diphtheroids. They
goats and abscesses or ulcerative lymphangitis in stain more uniformly than diphtheria bacilli,
horses. Rarely, it may cause subacute and chronic are arranged in V forms or palisades rather
lymphadenitis involving axillary or cervical lymph than Chinese letter arran­g ement and possess
nodes in those with prolonged exposure to sheep few or no metachromatic gra­nules. They can be
and horses. differentiated from C. diphthe­riae on the basis
of biochemical characters and toxigenicity tests
3. Corynebacterium minutissimum (Table 28.3). The common diphtheroids are C.
It is believed to be the causative agent of erythrasma. pseudodiphtheriticum and C. xerosis.
It is a localized infection of the stratum corneum.
Other Coryneform Genera
4. Corynebacterium jeikeium Other genera of irregularly shaped, gram-positive
Cornybacterium jeikeium, infections have been ba­c illi are Arcanobacterium, Brevibacterium,
limited to patients who are immune compromised, Oerskovia and Turicell. They have been found to
have undergone invasive proce­dures, or have a colonize humans and cause disease.

www.ebook3000.com
204  |  Section 3: Systemic Bacteriology
Table 28.3  Differences between C. diphtheriae and diphtheroids
Feature C. diphtheriae Diphtheroids
1. Morphology i. Weakly gram-positive and thin bacilli Strongly gram-positive, short and thick bacilli
ii. Metachromatic granules present Few or absent
iii. Arranged in Chinese letter pattern Pallisade arrangement
iv. Pleomorphism present Very little pleomorphism present
2. Culture Grow on enriched media Can grow on ordinary media
3. Biochemical tests Ferments glucose only and does not Ferments both glucose and sucrose
ferment sucrose
4. Toxin production Toxic Nontoxic
5. Virulence tests Positve Negative

f. Nondiphtheria corynebacteria
Key Points g. Diphtheroids.
™™ Corynebacterium is gram-positive bacilli with an
irregular shape, tendency to clubbing at one or both,
highly pleomorphic with Chinese letter or cuneiform
Multiple choice questions (MCQs)
arrangement. The granules in the cell are known as 1. Corynebacterium diphtheriae is classified into three
metachromatic granules volutin granules or Babes distinct biotypes (mitis, intermedius, and gravis)
Ernst granules. With Albert’s stain, the granules stain based on the morphologies of the colonies on:
bluish black and the protoplasm green a. Tellurite blood agar
™™ Two media are useful. 1. Loeffler’s serum slope- b. Loeffler’s serum slope
tellurite blood agar c. Blood agar
™™ Fermentations of sugars are usually done in Hiss’s
d. All the above media
serum peptone water medium
2. Diphtheria toxin shows following features except:
™™ C. diphtheriae is H2S positive and reduces nitrate
a. It is a protein with a molecular weight of 58,300
to nitrite
™™ Toxin: Toxigenic strains of C. diphtheriae produce
Da
a very powerful exotoxin. The toxigenicity of the b. It consists of two functionally distinct polypep-
diphtheria bacillus de­pends on the presence in it of tide chain fragments, A and B protein
corynephages (tox+). Diphtheria toxin is, heat labile c. It inhibits synthesis of fatty acids
protein, and consists of two fragments. Inhibition of d. It is a very potent toxin
protein synthesis is probably responsible for both 3. Following statements are true for diphtheria
the necrotic and neurotoxic effects of the toxin antitoxin except:
™™ Clinical diseases: Diphtheria occurs in two forms a. It is the mainstay of therapy in diphtheria
(respiratory and cutaneous) b. It is of immense value in cutaneous diphtheria
™™ Laboratory diagnosis: Depends upon micros­ c. It is of no value in treatment of asymptomatic
copy,culture and virulence tests. Virulence testing carriers
may be by in vivo or in vitro methods. In vivo tests d. It is ineffective after toxin has entered into the cell
are i. Subcutaneous test; ii. Intracutaneous test. 4. Diphtheria toxin has a special affinity for which of
In vitro test include i) Precip­itation test; ii) Tissue the following tissue/s?
culture test; iii) Enzyme-linked immunosorbent a. Heart muscles b. Nerve endings
assays (ELISA); iv) Polymerase chain reaction (PCR) c. Adrenal glands d. All of the above
™™ Prophylaxis: DPT given at the age of 6 weeks, 10 5. Which of the following sites is most commonly
weeks, 14 weeks and 16–24 months followed by
affected by diphtheria bacilli?
booster dose DT at the age of 5–6 years (school entry)
a. Upper respiratory tract b. Skin
™™ Diphtheroids: Cor ynebacteria resembling
c. Cornea d. Conjunctiva
C. diphtheriae occur as normal commensals in the
throat, skin and other areas. These may be mistaken
6. Diphtheroids show following features except:
for diphtheria bacilli and are known as diphtheroids. a. They possess few or no metachromatic granules
The common diphtheroids are C. pseudodiphth­ b. They are usually arranged in parallel rows
eriticum and C. xerosis. c. Most of them do not produce toxins
d. They do not ferment sucrose
7. A positive Schick test implies that the person is:
Important questions a. Immune and nonhypersensitive.
b. Susceptible and nonhypersensitive
1. Discuss morphology, cultural characteristics c. Immune and hypersensitive
and biochemical characters of Corynebacterium d. Non-immune and hypersensitive
diphtheriae. 8. Which of the following bacteria can cause infection
2. Name different species of genus Corynebacterium. in immunocompetent patients?
Discuss in detail laboratory diagnosis of diphtheria. a. Corynebacterium ulcerans
3. Write short notes on: b. Corynebacterium haemolyticum
a. Diphtheria toxin c. Corynebacterium pseudotuberculosis
b. Pathogenicity of C. diphtheriae d. All of the above
c. Toxigenicity tests/Virulence tests of C. diphtheriae
d. Schick test Answers (MCQs)
e. Prophylaxis of diphtheria 1. a; 2. c; 3. b; 4. d; 5. a; 6. d; 7. b; 8. d
29
Chapter

Bacillus

Learning Objectives

After reading and studying this chapter, you should ∙∙ Discuss laboratory diagnosis of anthrax
be able to: ∙∙ Descrbethe following: Anthracoid bacilli; Bacillus
∙∙ Describe the morphology, cultural characteristics cereus food poisoning.
and pathogenicity of Bacillus anthracis

Introduction 1. It was the first pathogenic bacterium to be


observed under the microscope (Pollender,
The family Bacillaceae has two clinically important 1849).
genera are Bacillus (the aerobic and facultative
2. The first communicable disease shown to be
anaerobic spore-formers) and Clostridium (the
transmit­ted by inoculation of infected blood
strict anaerobic spore-formers ). In the past
(Davaine, 1850) was anthrax.
few years, Bacillus has been subdivided into six
genera. 3. B. anthracis was first bacillus to be isolated
in pure culture and shown to possess spores
(Koch, 1876).
General Characterstics of Bacillus
4. It was in studies on anthrax that Koch demon­
1. The genus Bacillus consists of aerobic ba­cilli strated for the first time a set of criteria or
forming heat resistant spores, are gram-positive, postulates.
and are generally motile with peritrichate 5. The first bacterium used for the preparation of
flagella, the an­thrax bacillus being a notable an attenuated vaccine by Pasteur.
exception. Their spores are ubiquitous, being 6. Nobel Prize winner Metchnikoff studied
found in soil, dust, water and air and constitute virulent and attenuated strains of B. anthracis,
the commonest con­taminants in bacteriological in his pioneering work on phagocytosis.
culture media.
Species: The species that are of medical interest Morphology
are as follows: B. anthracis is one of the largest of pathogenic
1. Bacillus anthracis: The organism responsible bacteria. 3–8 by 1–1.3 µm and is gram-positive
for anthrax, is the most important member of nonacid fast, non-motile straight, sporing
this genus. bacillus. It is rec­tangular in shape and arranged
2. Bacillus cereus: May contaminate food, in filamentous chains in culture. In cultures, the
especially rice, in large numbers and is bacilli are arranged end to end in long chains. The
commonly implicated in episode of food ends of the bacilli are truncated or often concave
poisoning. and somewhat swollen so that a chain of bacilli
presents a ‘bamboo stick’ appearance.
The spore is oval (ellipsoidal), refractile, central
Bacillus anthracis in position and of the same diameter as the Bacillus
Historically, considerable attention was early and not swelling the mother cell (Fig. 29.1). Spores
focused on the genus Bacillus because of the are formed in culture, in the soil, and in the tissue
economic importance of anthrax, the disease and exudates of dead animals but never in the
caused by B. anthracis. blood or tissues of living animals. Spores seen as

www.ebook3000.com
206  |  Section 3: Systemic Bacteriology

Fig. 29.1: Anthrax bacilli Fig. 29.2: Medusa head appearance of anthrax bacilli

unstained spaces in Gram-stained bacilli and, when


free, faintly outlined with Gram counterstain. The
anthrax bacillus is nonmotile, unlike most other
members of this genus.
It is found singly, in pairs or in short chains
in tissues. The entire chain being surrounded by
a capsule which is polypeptide in nature, being
composed of a polymer of d (–) glutamic acid.
Capsules are formed in the animal body but in
culture only if the media contain added bicarbonate
or are incubated under 10–25% CO2.
They are gram-positive. When blood films
con­t aining anthrax bacilli are stained with
polychrome methylene blue for a few seconds and
examined under the microscope, an amorphous
purplish material is no­t iced around the blue
bacilli. This represents the capsular material and Fig. 29.3: Anthrax bacillus in gelatin stab culture showing
is characteristic of the anthrax bacillus. This is inverted fir tree appearance
called the M’Fadyean’s reaction and is em­ployed 3. In broth: Growth develops as silky strands, a
for the presumptive diagnosis of anthrax in surface pellicle and a floccular deposit.
an­imals. Purple bacillus with red capsule is seen 4. In a gelatin stab, there is growth down the stab
with Giemsa’s stain. Fat globules may be made line with lateral spikes, longer near the surface,
out within the bacilli when stained with Sudan giving an ‘inverted fir tree’ appearance (Fig.
black B. Spores seen as unstained spaces in Gram- 29.3).
stained bacilli and, when free, faintly outlined with 5. Selective medium: A selective medium (PLET
Gram counterstain. medium), consisting of polymyxin, lysozyme,
ethylene diamine tetra acetic acid (EDTA) and
Cultural Characteristics thallous acetate added to heart infusion agar,
It is aerobe and facultative anaerobe. Temperature has been devised to isolate B. antracis from
range for growth is 12–45°C (optimum 37°C). Good mixtures containing other spore-bearing bacilli.
growth oc­curs on ordinary media. 6. Solid medium containing penicillin: When
1. Nutrient agar: On nutrient agar, colonies are B. anthracis is grown on a solid medium
irregularly round, 2–3 mm in diameter, raised, containing 0.05–0.5 units penicillin per mL,
dull, opaque, greyish white, with a frosted in 3–4 hours the cells become large, spherical,
glass appearance. The edge of the colony is and occurs in chains on the surface of the
composed of long, interlacing chains of bacilli, agar, resembling a string of pearls. This string
resem­bling locks of matted hair under the low of pearls reaction useful in differentiation of
power microscope. This is called the ‘Medusa B. anthracis from B. cereus and other aerobic
head appearance’ (Fig. 29.2). spore bearers. B. anthracis is susceptible to
2. Blood agar: Colonies on horse or sheep blood gamma phage which is another characteristic
agar are virtually nonhemolytic. that differentiates it from B. cereus.
Chapter 29: Bacillus  | 207
Biochemical Reactions be localized, re­sembling the cutaneous disease
B. anthracis ferments glucose, maltose, sucrose, in human beings. Infected animals shed in the
trehalose and dextrin with the production of discharges from the mouth, nose and rectum, large
acid and no gas. Nitrates are reduced to nitrites. numbers of bacilli, which sporulate in soil and
Catalase is formed. remain as the source of infection.

B. Human infection
Resistance
Based on the mode of infection, human Anthrax
The spores are resistant to chemical disinfec­tants
presents in one of three ways: (1) cutaneous (2)
and heat. With moist heat, the vegetative bacilli are
pulmonary, or (3) intestinal. All types leading to
killed at 60°C in 30 minutes and the spores at 100°C
fatal septicemia or meningitis.
in 10 minutes. With dry heat the spores are killed at
150°C in 60 minutes. The spores are also killed by 1. Cutaneous Anthrax
4% formaldehyde or 4% potassium permanganate Cutaneous anthrax used to be caused by shaving
in a few minutes. brushes made with animal hair. It begins 2–5 days
The bacilli are sensitive to benzylpenicillin, after infection as a small papule that develops
streptomycin, tetracyclines, chloramphenicol, within a few days into a vesicle filled with dark
ciprofloxacin, the cephalosporins and sulfonamides. bluish black fluid. Rupture of the vesicle reveals
a black eschar at the base, with a very prominent
Antigenic Structure inflammatory ring of reaction around the eschar.
(The name anthrax, which means coal, comes
Three main antigens have been characterized: from the black color of the es­c har). This is
1. Capsular polypeptide: It is polypeptide con­ sometimes referred to as a malignant pustule.
sisting exclusively of D-glutamic acid. The lesion is classically found on the hands,
2. Somatic polysaccharide: It is a component of forearms, or head and is painless. The disease used
the cell wall. to be common in dock workers carrying loads of
3. Complex protein toxin (anthrax toxin): hides and skins on their bare backs and hence was
Anthrax toxin is a complex exotoxin consisting of known as the ‘hide porter’s disease.’ Cutaneous
three protein components: Protective antigen anthrax generally resolves spontaneously, but
(PA), lethal factor (LF), and edema factor (EF). 10–20% of untreated patients may develop fatal
Each of the separate components is serol­ogically septi­cemia or meningitis.
active and distinct and is also immunogenic.
2. Pulmonary Anthrax
Pulmonary anthrax, known as ‘wool-sorter’s
Virulence Factors disease, because it used to be common in workers
The pathogenesis of B. anthracis depends on two in wool factories, due to inhalation of dust from
important virulence factors: A poly (D-glutamic infected wool. It occurs in patients who handle raw
acid) capsule and a three-component protein wool, hides, or horsehair and acquire the disease
exotoxin. Fully virulent organisms have two large by the inhalation of spores. This is a hemorrhagic
plasmids that code for these products. pneumonia with a high fatality rate. Hemorrhagic
1. Capsule: The capsule interferes with phagocytosis. meningitis may occur as a complication.
2. Anthrax toxin: Anthrax toxin consists of three 3. Intestinal Anthrax
proteins called protective antigen (PA), edema Intestinal anthrax, is rare and occurs mainly in
factor (EF), and lethal factor (LF), each of primitive communities who eat the carcasses of
which individually is nontoxic but together act ani­mals dying of anthrax. An individual may suffer
synergistically to produce damaging effects. after a day or so from hemorrhagic diarrhea, and
The three fac­tors have been characterized and dies rapidly from septicemia.
cloned.
Laboratory Diagnosis of Human Anthrax
Pathegenesis
In laboratories unfamiliar with the disease,
Cattle, sheep, goats and other herbivores are
additional precautions for staff safety need to be
naturally infected.
organized. All procedures connected with the
handling of B. anthracis should be carried out with
A. Animal Infection greatest care in a safety cabinet.
Anthrax is a zoonosis. Animals are infected by the 1. Specimens: Material from a malignant pustule,
inges­tion of the spores present in the soil. Direct sputum from pulmonary anthrax, gastric
spread from animal to animal is rare. The disease aspirates, feces or food in intestinal anthrax and
is generally a fatal septicemia but may sometimes in the blood in the septicemic stage of all forms

www.ebook3000.com
208  |  Section 3: Systemic Bacteriology
of the infection. Specimens should be taken antigen, a ring of precipitation will appear at the
before antibiotic therapy has been instituted. junction of the two liquids within 5 minutes at
2. Microscopy: Prepare smears of each specimen room temperature.
and stain with Gram’s method and McFadyean’s With the availability of purified anthrax toxin
method or with Giemsa stain. Gram’s stain antigen, Ascoli’s test has been replaced by highly
may show typical large gram-positive bacilli. sensitive and specific immunoassays. EIA can also
Capsule appears as a clear halo around the detect antibody in the serum of animals surviving
bacterium by India-ink staining. anthrax infection.
Direct fluorescent antibody test (DFA) for
capsule specific staining and for polysaccharide 6. Polymerase Chain Reaction (PCR)
(cell wall) antigen confirms the identification. A sensitive and specific PCR technique has been
3. Culture: Culture the exudate on nutrient agar, developed for the detection of anthrax contami­
blood agar, PLET medium and nutrient broth. nation of animal and agricultural products.
Incubate at 37°C for 18 hours. Examine plates
for the medusa-head colonies characteristic Treatment
of B. anthracis, nonhemolytic on the blood Ciprofloxacin is the drug of choice. Penicillin,
agar plate. Prepare a smear, stain it by Gram’s doxycycline, erythromycin, or chloramphenicol
method and look for tan­gled chains of large can be used (if suscepti­ble).
gram-positive bacilli some of which have
central, oval, non-bulging spores. In nutrient Prophylaxis
broth, look for a pellicle and a deposit.
Control of human anthrax ultimately depends on
4. Confirmatory tests
control of the disease in animals. Animals with
i. Biochemical and physiological reactions:
known or suspected anthrax should be handled
Demonstration of non-motility, gelatin
with care and carcasses of animals suspected to have
liquefaction, growth in straight chains and died of anthrax are incinerated or buried deep in
enhanced growth aerobically, as seen in the quicklime. Wool, horsehair, and hides coming from
characteristic inverted fir tree appearance areas where epidemic anthrax is present should be
in a gelatin stab, will generally iden­tify B. gas sterilized. A vaccine is available for control of
anthracis completely. outbreaks of human anthrax in an ‘industrial setting’.
Toxin production can be demonstrated by
immunological or gene probe methods in Immunization
reference laboratories. Live-attenuated bacilli were first used by Louis
ii. Animal inoculation: Inoculate intraperi­ Pasteur in May 1881. Pasteur’s vaccine was the
toneally in mice a 24 hours broth culture. anthrax bacillus attenuated by growth at 42–43°C.
The animal dies in 48–72 hours. Make The original Pasteur’s anthrax vaccine is of great
smears from heart blood and spleen, stain historical importance.
by Gram’s and McFadyean’s methods, and
Sterne strain of live spore vaccine: Subsequently,
look for typical anthrax bacilli. the Sterne strain of live spore vaccine has been
The anthrax bacillus can often be isolated used for animal immunization. The Sterne vaccine
from contaminated tissues by applying contained spores of a noncapsulated avirulent
them over the shaven skin of a guinea mutant strain. Live bacterial vaccines are not
pig. It is able to pen­etrate through minute considered safe for man.
abrasions and produce fatal infection.
Alum-precipitated toxoid prepared from the
5. Serological diagnosis: Serological diagnosis by
protective antigen has been shown to be a safe
enzyme-linked immunosorbent assay (ELlSA)
and effective vaccine for human use. It has been
is seldom used diagnostically.
used in persons occupationally exposed to
Ascoli’s thermoprecipitin test: If the sample anthrax in­fection. Three doses intramuscularly at
received is putrid so that viable bacilli are intervals of six weeks between first and second,
unlikely, diagnosis may be established by Ascoli’s and six months be­tween second and third doses
thermoprecipitin test by demonstration of the induce good immunity, which can be reinforced if
anthrax antigen in tissue extracts.The original necessary with annual booster injections. Frequent
thermoprecipitin test devised by Ascoli (191l) booster doses are necessary.
was a ring precipitation by letting the boiled
tissue extract in a test tube react with the anthrax
ANTHRACOID BACILLI
antiserum. The tissue is ground up in saline, boiled
for 5 minutes, filtered and layered over anti anthrax Nonpathogen­ic aerobic spore bearing bacilli having
serum in a narrow tube. If tissue contains anthrax a general resemblance to anthrax bacilli have been
Chapter 29: Bacillus  | 209
collectively called pseudoanthrax or anthracoid There is a longer incubation period occurring
bacilli. The important species include B. cereus, B. 8–24 hours after ingestion, during which the
subtilis, B. stearothermophilus B. licheniformis, B. organism multiplies in the patient’s intestinal
pumilus. B. cereus has been recognized as a frequent tract and produces the heat-labile entero­toxin.
cause of foodborne gastroenteritis. Table 29.1 lists Then the diarrhea, nausea, and abdominal
the main differentiating features between Anthracoid cramps develop.
bacilli and B. anthracis. The diarrheal disease is mostly caused
by serotypes 2, 6, 8, 9, 10 or 12 of B. cereus
Bacillus cereus strains. Isolates from the diarrheal type of
B. cereus has recently assumed importance as a disease produce entero­toxin which causes
cause of food poisoning. It is widely distributed in fluid accumulation in ligated rab­bit ileal loop,
nature, may be readily isolated from soil, vegetables resembling the heat labile enterotoxin of
and a wide variety of foods including milk, cereals, Escherichia coli and Vibrio cholerae.
spices, meat and poultary. 3. Ocular infection: Common cause of post-
traumatic ophthalmitis.
4. Other opportunistic infections: Intravenous
Pathogenesis catheter and central nervous system shunt
1. Emetic form (the short incubation type): The infections and endocarditis as well as
emetic form results from the consumption of pneumonitis, bactere­mia, and meningitis in
contami­nated rice. The heat-stable enterotoxin severely immunosuppressed pa­tients.
that is released is not destroyed when the rice
is reheated. After ingestion of the enterotoxin Laboratory Diagnosis
and a 1–6-hour incubation period, a disease of
If food is available for testing, confirmation is easy.
short duration (less than 24 hours) develops.
High numbers of B. cereus, in the absence of other
Symptoms consist of vomiting, nausea, and
food poi­soning bacteria are sufficient to make the
abdominal cramps. Fever and diarrhea are
diagnosis.
generally absent.
Large facultatively anaerobic Gram-positive
Two mechanisms of action have been
bacilIi that produce anthracoid colonies on blood
described for the enterotoxin of B. cereus, one
agar after overnight incubation at 37°C are almost
involving stimulation of CAMP system and the
certain to be B. cereus.
other independent of it.
2. Diarrheal form: The diarrheal form of B. cereus Treatment : Both the emetic and diarrheal
food poisoning re­sults from the consumption syndromes are shortlived and no specific treatment
of contaminated meat, veg­etables or sauces. is needed.

Table 29.1  Differentiating features between B. anthracis and Anthracoid bacilli


Features B. anthracis Anthracoid bacilli
1. Motiliy Nonmotile Generally motile
2. Capsule Capsulated Noncapsulated
3. Chain formation Grow in long chains Grow in short chains
4. Colony on nutrient agar Medusa head colony Not present
5. Growth in penicillin agar (10 units/mL) No growth Grow usually
6. Hemolysis on blood agar Hemolysis absent or weak Usually well-marked
7. Gelatin stab culture Inverted fir tree growth and slow Rapid liquefaction
gelatin liquefaction
8. Turbidity in broth No turbidity Turbidity usual
9. Salicin fermentation Negative Usually positive
10. Growth at 45°C No growth Grows usually
11. Growth inhibition by chloral hydrate Growth inhibited Not inhibited
12. Susceptible to gamma phage Susceptible Not susceptible
13. Pathogenic to laboratory animals Pathogenic Not pathogenic
14. Mc Fadyean’s reaction Positive Negative
15. Ascoli’s precepitin test Positive Negative
16. Fluorescent antibody test with anthrax Positive Negative
antiserum

www.ebook3000.com
210  |  Section 3: Systemic Bacteriology
Prevention Important questions
Prevention is best accomplished by the prompt 1. Discuss laboratory diagnosis of anthrax.
refri­geration of boiled rice and other foods because 2. Write short notes on Bacillus cereus food poisoning.
it is nearly impossible to eliminate B. cereus spores
from food. Multiple choice questions (Mcqs)
Key Points 1. McFadyean’ s reaction is employed for the presump­­­
™™ The genus Bacillus consists of Gram positive, aerobic tive diagnosis of:
ba­cilli forming heat resistant spores a. Anthrax b. Tetanus
™™ Bacillus has two important species—1. Bacillus c. Diphtheria d. Typhoid
anthracis—the organism responsible for anthrax 2. ‘Medusa head’ appearance of the colonies is
and 2. Bacillus cereuus—can cause food poisoning.
characteristic of:
™™ Bacillus anthracis
a. Proteus mirabilis
Spore-forming gram-positive, capsulated bacilli.
B anthracis is a M’Fadyean’s reaction positive. It b. Clostridium tetani
grows as ‘Medusa head appearance’ on nutrient c. Bacillus anthracis
agar medium d. Pseudomonas aeruginosa
™™ Diseases: Cutaneous anthrax, inhalation anthrax 3. Malignant pustule is characteristic of:
and gastrointestinal anthrax a. Cutaneous anthrax
™™ Diagnosis: Isolation of the organism from clinical b. Pulmonary anthrax
specimens (e.g. papule or ulcer, blood)
c. Intestinal anthrax
™™ Animal vaccination is effective, but human vaccines
d. All of the above
have limited usefulness
™™ Aerobic spore bearing bacilli having a general resem­ 4. Ascoli’s thermoprecipitin test helps in confirming
blance to anthrax bacilli which have been collectively the laboratory diagnosis of:
called pseudoanthrax or anthracoid bacilli a. Anthrax b. Tetanus
™™ Bacillus cereus Infections c. Typhoid d. Cholera
People at risk include those who consume food 5. Which of the following foods is most often
contaminated with the bacterium (e.g. rice, meat, associated with emetic type of food poisoning
vegetables, sauces), those with penetrating injuries
caused by Bacillus cereus?
(e.g. to eye), and those who receive intravenous
a. Meat b. Milk
injections
™™ Diseases: Emetic (vomiting) and diarrheal forms c. Eggs d. Rice
of gastroenteritis; ocular infection; and other
opportunistic infections Answers (MCQs)
1. a; 2. c; 3. a; 4. a; 5. d
30
Chapter

Clostridium

Learning Objectives

After reading and studying this chapter, you should ∙∙ Discuss morphology, cultural characteristics of
be able to: C. tetani
∙∙ Describe classification of clostridia and diseases ∙∙ Describe toxins produced by C. tetani
produced by different clostridia ∙∙ Describe the following: Pathogenesis of tetanus;
∙∙ Discuss morphology, cultural characteristics of prophylaxis of tetanus
C. welchii ∙∙ Discuss morphology and cultural characteristics
∙∙ Discuss Nagler reaction of C. botulinum
∙∙ Discuss lab. diagnosis and prophylaxis of gas ∙∙ Discuss lab. diagnosis of botulinum
gangrene ∙∙ Describe the following: gas gangrene; botulinum
toxin; Clostridium difficile.

Introduction terminally. The position of the developing


spores within the vegetative cell is useful in
The genus Clostridium includes all anaerobic,
identifying the species.
gram-positive bacilli capable of forming
2. Culture: Most species are obligate anaerobes.
endospores. Spores of clostridia are usually wider
Clostridia grow on enriched media in the
than the diameter of the rods in which they are
presence of reducing agent such as cysteine
formed, giving the bacillus a swollen appearance
or thioglycollate (to maintain a low oxidation-
resembling a spindle (Clostridium is Latin for ‘little
reduction potential),or in an O2 free gaseous
spindle’). The name Clostridium is derived from
atmosphere.
the word ‘Kloster’ (meaning a spindle).
A very useful medium is Robertson’s cooked
Most of the species are saprophytes that
meat broth (CMB). It contains unsaturated fatty
normally occur in soil, water and decomposing
acids which take up oxygen the reaction being
plant and animal matter. The genus contains bacteria
catalyzed by hematin in the meat. Clostridia
responsible for three major diseases of human
grow in the medium, render­ing the broth
beings—gas gangrene, food poisoning and tetanus.
turbid. Most species produce gas.
3. Biochemical reactions: On the basis of
General features of clostridia
biochemical reactions many clo­stridia can be
1. Morphology: The clostridia are gram-positive divided into: (A) Predominantly saccharolytic
typically large, straight or slightly curved rods, clostridia, (B) Predominantly proteolytic
3–8 × 0.6–1 μm with slightly rounded ends. clostridia, (C) Slightly proteolytic clostridia
Most species of Clostridia are motile with Table 30.1.
peritrichous flagella except C. perfringens 4. Resistance: Spores of C. botulinum may
and C. tetani type VI which are non­motile. All withstand boil­ing after 3–4 hours and even at
clostridia are noncapsulated with the exception 105°C may not killed be completely in less than
of C. perfringens and C. butyricum. 100 minutes. Spores of most strains of
All produce endospores. Spores of clostridia C. perfringens are destroyed by boiling for less
are usually wider than the diameter of the rods than five minutes, but those of some type A
in which they are formed. In the various species, strains that cause food poisoning survive for
the spore is placed centrally, subterminally, or several hours. C.terani spores persist for years

www.ebook3000.com
212  |  Section 3: Systemic Bacteriology
in dried earth or dust. All species are killed by Table 30.1  Clostridia as human pathogens
autoclaving at 121°C within 20 minutes. Among
A. The gas gangrene group
hospital disinfectants the greatest sporicidal
1. Established •  C. perfringens
activity is shown by alcoholic hypochlorite and pathogens •  C. septicum
glutaraldehyde. •  C. novyi
In general, clostridia are suscepti­ble to 2. Less pathogenic •  C. histolyticum
metronidazole, penicillin chloramphenicol •  C. fallax
3.  Doubtful pathogens •  C. bifermentans
and erythromycin; less so to tetracyclines, and
•  C. sporogenes
resistant to aminoglycosides and quinolones.
5. Diseases produced: Clostridia are more B. Tetanus C. tetani
commonly associ­ated with skin and soft tissue C. Food poisoning
infections, food poison­ing, and antibiotic- 1. Gastroenteritis C. perfringens (Type A)
associated diarrhea and colitis (Table 30.1). 2. Necrotizing enteritis C. perfringens (Type C)
3. Botulism C. botulinum
D.  Acute colitis C. difficile
Classification
The traditional method for classifying an isolate in
the ge­nus Clostridium was based on a combination
of diag­nostic tests, including the demonstration of
spores, op­timal growth in anaerobic conditions, a
complex pattern of biochemical reactivity such as
saccharolytic and proteolytic capacities and the
findings yielded by gas chromatography analysis
of the meta­bolic byproducts. With these methods,
more than 130 species have been defined.
Fortunately, most of the clinically important
isolates fall within a few species. Fig. 30.1: Reverse CAMP test

hu­man blood agar. It results from a narrow zone of


Clostridium perfringens (Clostridium welchii) complete hemolysis due to theta toxin and a much
C. perfringens is a normal inhabitant of the large wider darker zone of incomplete hemolysis due to
intestines of human beings and animals. The the α-toxin. On longer incubation this double zone
spores are commonly found in soil, dust and air. pattern of hemolysis may fade.
C.perfrigens also produces a characteristic
Morphology pattern of synergistic b-hemolysis when streaked
It is a relatively large gram-positive bacillus (about alongside Streptococcus agalactiae (the reverse
4–6 × 1μm) . It is pleomorphic, and filamentous. It CAMP test) (Fig 30.1).
is capsulated and nonmotile.
Spores are typically oval, central or sub terminal Biochemical Reactions
and not bulging but are rarely seen in artificial culture
It is actively saccharolytic. Glucose, maltose, lactose
or in material from pathological lesions, and their and sucrose are fermented with the production
absence is one of the characteristic morphological of acid and gas. It is indole negative, MR positive
features of C. perfringens. Special media normally and VP negative. Hydrogen sulphide is produced
must be used to demonstrate sporulation. abundantly; sulphite is actively reduced; most
strains reduce nitrates to nitrites.
Cultural Characteristics In litmus milk medium, fermentation of
It is an anaerobe. It grows over a pH range of 5.5–­ lactose leads to formation of acid, which is indicated
8.0 and temperature range of 20°C–50°C (optimum by the change in the color of litmus from blue to red.
temperature range 37°C–45°C). It grows on blood The acid clots the milk – casein-(acid clot) and the
agar, cooked meat broth(CMB) and thioglycollate clotted milk is dis­rupted due to the vigorous gas
broth within 24–48 hours. Good growth occurs in production. This is known as ‘stormy fermentation
Robertson’s cooked meat medium. The meat is or ‘stormy clot’ reaction but is not specific for this
turned pink but is not di­gested. The culture has an organism.
acid reaction and a sour odor.
Surface colonies are large, smooth, regular, Resistance
convex, slightly opaque discs. Colonies of most Spores are usually destroyed within five minutes
strains demonstrate a ‘tar­get hemolysis’ after by boiling but those of the ‘food poisoning’ strains
overnight incubation on rabbit, sheep, ox, or of type A and certain type C strains resist boiling
Chapter 30: Clostridium  | 213
for 1–3 hours. Autoclaving of 121°C for 15 minutes
is lethal. Spores generally resist routinely used
antiseptics and disinfectants.

Toxins
C. perfringens is one of the most prolific of toxin-
producing bacteria, forming at least 12 distinct
soluble substances or toxins, all of which are of
protein in nature and antigenic. Major lethal toxins
include: α (alpha), b (beta), e (epsilon) and i (iota)
and minor lethal toxins include: g (gamma), d
(delta), k (kappa), l (lambda), m (mu), n (nu), q
(theta) and h (eta). The four 'major toxins', alpha,
Fig. 30.2: Nagler’s reaction. C perfringens colonies on the
beta, epsilon and iota, are predominantly respon­ right half of the plate are surrounded by haloes, while colonies
sible for pathogenicity. on the left half (containing antiserum to alpha toxin) have no
haloes around them
Classification Interpretation: On the section containing no
C. perfrin­gens can be divided into five types, A to antitoxin, C. perfringens colonies show surrounding
E on the basis of four major toxins. Strains of C. zone of opalescence, i.e. Nagler reaction. There
perfringens type A that produce enterotoxin are will be no opacity around the colonies on the half
associated with a mild form of food poisoning. of the plate with the antitoxin, due to the specific
neutralization of the alpha toxin (Fig. 30.2).
Alpha Toxin This reaction, however, is not totally specific
The alpha (a) toxin is produced by all types of for C. perfringens since the opalescence in the egg
C. perfringens and most abundantly by type A yolk media may be produced by other lecithinase
strains, is a lecithinase (phospho­lipase C) that lyses forming bacte­ria (C. novyi, C. bifermentans, some
erythrocytes, platelets, leukocytes, and endothelial vibrios, some aerobic spore bearers). The reaction
cells. It is lethal, dermonecrotic and hemolytic for produced by C. perfringens is speci­fically neutralized
the red cells of most species, except horse and by C. perfringens antitoxin, but serologically related
goat. The lysis is of the hot-cold variety, being best phospholipases of C. bifermen­tans and ‘C. sordellii
seen after incubation at 37°C followed by chilling and some other phospholi­pases are also inhibited.
at 4°C. This toxin increases vascular permeability, These organisms can be separated by other tests.
resulting in massive hemolysis and bleeding,
tissue destruction, hepatic toxicity, and myocardial Other Major Toxins
dysfunction. It is relatively heat stable. Beta (β), Epsi­lon (e) and iota (i) toxins have lethal
and necrotizing properties.
Nagler’s Reaction
Minor Toxins
Basis: The alpha (a) toxin is lecithinase C (or
phospholipidase C) splits lecithin into phosphoryl Gamma and eta toxins have minor lethal toxins.
choline and diglyceride, in the presence of Ca++ and Delta toxin has a lethal effect and is hemolytic for
Mg++ ions because the toxin is activated by Ca++ and the red cells of even- goat, pigs and cattle. Theta
Mg++ ions. This reaction is seen as opalescence in toxin is oxygen-labile hemolysin antigeni­cally
serum or egg yolk media and is specifically neutral­ related to streptolysin O. It is also lethal and a
ized by the antitoxin. This is the basis of Nagler general cytolytic toxin.
reaction.
Enterotoxin
Procedure: For rapid detection of C. perfringens, a
C. perfringens type A strains produce a potent
culture plate containing 6% agar, 5% peptic digest
enterotoxin which causes diarrhaea and other
of sheep blood and 20% human serum or 5% egg-
symptoms of food poisoning.
yolk is prepared. The in­corporation of neomycin
sulphate in the medium makes it more selective,
Pathogenesis
inhibiting coliforms and aer­obic spore bearers.
On one half of the plate, 2–3 drops of C. 1. Soft Tissue Infections
perfringens antitoxin are spread and allowed to dry. Soft tissue infections caused by C. perfringens
The plate is then inocu­lated with the test organisms are subdi­vided into (1) cellulitis, (2) fasciitis or
or the exudate under investigation and incubated suppurative myositis, and (3) myonecrosis or gas
anaerobically at 37°C for 18 hours. gangrene.

www.ebook3000.com
214  |  Section 3: Systemic Bacteriology
Clostridial myonecrosis or gas gangrene: The 5. C. perfringens Colitis
disease is characterized by rapidly spreading edema, A sporadic diarrheal syndrome, usually occurring in
myositis, necrosis of tissues, gas production and elderly patients during treatment with antibiotics,
profound toxemia occurring as a complication of has been described.
wound infection. The disease has been referred to in
the past as ‘malignant edema’. Other descriptive terms 6. Clostridial Endometritis
that have been used are ‘anaerobic (clostridial)
This condition is a grave complication of incom­
myositis’ and ‘clostridial myonecrosis.
plete abortion, or the use of inadequately sterilized
Etiology: Amongst the pathogenic clostridia, instruments.
C. perfringens is the most frequently encountered
(ap­p rox imately 60%), and C . n ov y i and Laboratory Diagnosis
C. septicum being the next common (20–40%), and
Gas gangrene is a medical emergency. The diagnosis
C. histolyticum less often. Other clostridia usually
of gas gangrene must be made primarily on clinical
found are C. sporogenes, C. fallax, C. bifermentans,
grounds, and the function of the laboratory is only
C. sordelli, C. aerofoetidum and C. tertium.
to provide confirma­tion of the clinical diagnosis
Mechanism of infection: Clostridial spores are as well as identification and enumeration of the
introduced into tissue, e.g. by contamination with infecting organisms.
dirt, or by endogenous transfer from the intestinal
tract. After injury there is incubation period may A. Specimens
be as short as seven hours or as long as six weeks,
(1) Edge of the affected muscles; (2) Exudates from
usually of 12–48 hours.
the wound; and (3) Necrotic tissue and mus­cle
When there is considerable cell injury or
fragments.
compromise of circulation, germination and
outgrowth of clostria spores occurs. Alpha toxin and
B. Microscopy
other exotoxins are secreted and extensive cell killing
ensues. The production of enzymes that break down Gram stained films give presumptive information
ground substance facilitates the spread of infection. about the species of clostridia present and their
Fermentation of tissue carbohydrates yields gas, and rela­t ive numbers. If gas gangrene is present,
an accu­mulation of gas bubbles in the subcutaneous Gram-positive rods may predominate. Thick,
spaces produces a crinkling sensation on palpation stubby, Gram-positive rods suggest C. perfringens
(crepitation), hence the name gas gangrene. or C. sordellii, ‘citron bodies’, boat- or leaf­shaped
pleomorphic bacilli with irregular staining, may
2. Septicemia indicate C. septicum; slender rods with round
terminal spores suggest C. tetani and large rods with
Invasion of the bloodstream may occur in associ­ oval sub terminal spores indicate C. novyi.
ation with malignancy and may involve a localized
myonecrosis in addition to a fulminating clostridial C. Culture
septicemia.
Fresh and heated blood agar are used for aerobic
and anaerobic cultures. To prevent swarming
3. Food Poisoning by some species of clostridia, the use of plates
The organisms usually involved are strains of type containing in­creased agar (5-6%) are considered. A
A. Meat, chicken, fish and their by-products are the plate of serum or egg yolk agar, with C. perfringens
most common vehicles for clostrial food poisoning. antitoxin spread on one half is used for the ‘Nagler
Clostridial food poisoning, is characterized by (i) a reaction’.
short incubation period (8–24 hours), (ii) a clinical Four tubes of cooked meat broth are inoculated
presentation that includes abdominal cramps and and heated at 100°C for 5, 10, 15 and 20 minutes,
watery diarrhea but no fever, nausea, or vomiting, incubated and sub­cultured on blood agar plates
and (iii) a clinical course lasting less than 24 hours. after 24–48 hours, to dif­ferentiate the organisms
The illness is self-limited and recovery occurs in with heat resistant spores. Blood cultures are
24–48 hours. often positive, especially in C. perfringens and
C. septicum infections. However, C. per­fringens
4. Enteritis Necroticans (Necrotizing bacteremia may occur without gas gangrene.
Jejunitis, Necrotic Enteritis)
This is severe and often fatal enteritis known by D. Identification
different names in different countries: β-toxin- Examine plates for typical colonies. The isolates are
producing C. perfringens type C is responsible for identified based on their morphological, cultural,
this disease. biochemical and toxigenic characters.
Chapter 30: Clostridium  | 215
Animal Pathogenicity
0.1 mL 24 hours growth in cooked meat broth is
injected into a healthy guinea pig by intramuscular
route. The animal dies within 24 hours. A control
animal protected with antiserum prior to test is also
included. On autopsy, bacteria can be recovered
from heart and spleen of the test animal.

Laboratory Diagnosis of Food Poisoning


Prophylaxis and Therapy
1. Surgery: All damaged tissues should be
removed promptly and the wounds irrigated
to remove blood clots, necrotic tissue and Fig. 30.3: C. tetani, some with spores and
foreign materials. In established gas gangrene, some without spores
uncompromising excision of all affected parts
may be life-saving. The meat is not digested but becomes black on
2. Antibiotics: Penicillin, metronidazole and prolonged incubation. On blood agar the bacilli
an aminoglycoside may be given in combin­ produce a swarming (thin spreading film) growth.
ation. Alternatively, clindamycin plus an On horse blood agar, the colonies of C. tetani
aminoglycoside or a broad-spectrum antibiotic, are surrounded by a zone of a-hemolysis, which
such as meropenem or imipenem, may be subsequently develops into b-hemolysis, due to
considered. the production of an oxygen-labile hemolysin
3. Passive immunization: A polyvalent antiserum known as tetanolysin. On egg-yolk agar, it does
used to be available but it has now been not produce opalescence or pearly layer.
replaced by intensive antimicrobial therapy. In deep agar shake cultures, the colonies are
4. Hyperbaric oxygen: Hyperbaric oxygen may spherical fluffy balls, 1–3 mm in diameter, made up
be beneficial in treatment and is introduced in of filaments with a radial arrangement. In gelatin
the depth of wound to reduce anaerobiosis. stab cultures a fir tree type of growth occurs, with
5. Active immunization: Toxoids have been slow liq­uefaction.
found to induce antitoxic response but it is not
of practical use. Biochemical Reactions
C. tetani has feeble proteo­lytic but no saccharolytic
Clostridium tetani property. It does not attack any sugar. Gelatin
liquefaction occurs very slowly. It is indole positive
Clostridium telani is the causative agent of tetanus,
and MR, VP, H2S and nitrate reduction negative.
a disease that is now relatively rare in well-
A greenish fluores­cence is produced on media
developed countries.
containing neutral red (as on MacConkey’s medium).
Morphology Resistance: The spores are killed by boiling for
It is a gram-positive, slender bacillus, 2–5 × 0.4–1 mm 10–15 minutes but some resist boiling for up
with rounded ends. The spores are spherical, ter­ to three hours. They can, however, be killed by
minal and twice the diameter of vegetative cells autoclaving at 121°C for 15 minutes.
giving them typical drumstick appearance (Fig. Spores are able to survive in soil for years.
30.3). It is non-capsulated and motile by peritrichate They are killed by exposure to iodine (1% aqueous
flagella (except C. tetani type VI) with peritrichate solution), hydrogen peroxide (10 volumes) and
flagella. Young cultures of the organism usually stain glu­taraldehyde (2%) within a few hours.
Gram-positive, but in older cultures and in smears
made from wounds, they are gram-variable and even Antigenic Structure
are gram-negative. Flagella (H), somatic (O), and spore antigens
have been demonstrated in C. tetani. The spore
Cultural Characteristics antigens are different from the H and 0 antigens of
C. tetani is an obligate anaerobe. The optimal the somatic cell.
temperature for growth is 37° C, and the optimal pH Flagella (H) antigen: Ten serological types have
is 7.4. It can grow well in cooked meat broth (CMB), been rec­ognized based on agglutination (types I
thioglycollate broth, nutrient agar and blood agar. to X) of which type I and III are the most common.
In cooked meat broth (CMB), growth occurs as This typing is on the basis of their flagellar (H)
turbidity and there is also some gas formation. antigens. Type VI contains nonflagellated strains.

www.ebook3000.com
216  |  Section 3: Systemic Bacteriology
Somatic (O) antigen: There is a single somatic tetanus’, are demonstrable in experimental animals,
agglutin­ation group for all strains that permits but the stages tend to merge in their clinical present­
identification of the organism. ation in man.

Tetanus Toxin Epidemiology


C. tetani produces at least two distinct toxins—an Tetanus is more common in the developing coun­
oxygen-labile hemolysin (tetanolysin) and a tries. In rural In­dia, tetanus was a common cause
powerful plasmid-encoded, heat-labile neurotoxin of death, parti­cularly in the newborn.
(tetanospasmin). A third toxin, a nonspasmogenic, Tetanus was a serious disease with a high rate
peripherally active neurotoxin, has been identified. of mortality (80–90%), before specific treatment
It is not known whether this plays any role in the became available. Tetanus neonatorum and
patho­genesis of tetanus. uterine tetanus have very high fatali­ty rates (70–
100%), while otogenic tetanus is much less serious.
Tetanospasmin
All of the symptoms in tetanus are attrib­utable to an Laboratory Diagnosis
extremely toxic neurotoxin, tetanospasmin. The toxin
The diagnosis of tetanus is made on clinical
is a heat-labile, oxygen stable, powerful plasmid-
grounds. Laboratory tests only help in confirmation.
encoded neurotoxin and has been crystallized. It
A. Specimen: Wound exudate or tissue removed
gets toxoided spontaneously or in the presence of low
from the wound.
concentrations of formaldehyde. It is a good antigen
B. Microscopy: Microscopy is unreliable and the
and is specifically neutralized by the antitoxin.
demonstration of the typical ‘drumstick’ bacilli
On release from the bacillus, it is autolyzed to
in wounds in itself is not diagnostic of tetanus.
form a heterodimer consisting of a heavy chain
It may not also be possible to distinguish by
(93,000 MW) and a light chain (52,000 MW) joined
microscopy between C. tetani and morphologi­
by a disulphide bond. The purified toxin is active in
cally similar bacilli such as C. tetanomorphum
extremely small amounts and has an minimal lethal
and C. sphenoides. Hence, microscopy is
dose (MLD) for mice of about 50–75 × 10-6 mg.
unreliable. Simple light microscopy is often
unsuc­cessful. Immunofluorescence microscopy
Tetanolysin with a specific stain is possible but not generally
Tetanolysin is a heat labile, oxygen labile hemo­ available.
lysin, antigenically related to the oxygen labile C. Culture: The material is inoculated on one
hemolysins produced by C. perfringens, C. novyi and half of a blood agar plate. C. tetani produces
Str. pyogens. It is not relevant in the pathogenesis of a swarming growth which may be detected
tetanus. on the opposite half of the plate after 1–2 days
incubation anaerobically. The incorporation of
Pathogenicity polymyxin B, to which clostridia are resistant,
Tetanus develops following the contami­nation makes the medium more selective.
of wound with C. tetani spores. The most typical The material is also inoculated into three
focus of infection in tetanus is a puncture wound. tubes of cooked meat broth, one of which is
Introduced for­eign bodies or small areas of cell heated to 80°C for 15 minutes, the second
killing create a nidus of devital­ized material in for five minutes, and the third left unheated.
which tetanus spores can germinate and grow. The purpose of heating for different pe­riods
Germination of spores is dependent upon the is to kill vegetative bacteria, while leaving
reduced oxygen tension occurring in devitalized un­damaged tetanus spores, which vary widely
tissue. After germination of the spores, toxin in heat resistance. The cooked meat tubes are
is elaborated and gains entrance to the central incubated at 37°C and subcultured on one half
nervous system. of blood agar plates daily for up to four days.
C. tetani has little invasive power. Infection D. Toxigenicity test: Toxigenicity is best tested
strictly remains localized in the wound and the in animals. Control mice are protected with
disease is due to the effect of a potent diffusible tetanus antitoxin. Two mice, one unprotected
exotoxin (tetanospasmin). The toxin exerts its effects and other protected are used for each test.
on the spinal cord, the brainstem, peripheral nerves, One animal is protected by giving 1,000 units of
at neuromuscular junctions and directly on muscles. tetanus antitoxin intraperi­toneally 1 hour before
Toxin diffuses to affect the relevant level of the the test. Inject 0.1 ml of a 48 hours CMB culture
spinal cord (local tetanus) and then to affect the supernate of the organism intramuscularly into
entire system (generalized tetanus). These stages, the hind limb of one mouse (the test) and the
including the intermediate one of ‘ascending same amount in the another animal (control
Chapter 30: Clostridium  | 217
animals). The protected mouse remains well. starting at age as early as 6 weeks. Booster
Signs of ascen­d ing tetanus develop in the doses are given at age of 18 months and then
unprotected animal after several hours, they at five years. Thereafter, booster doses of TT
begin in the inoculated leg and extend to the (tetanus toxoid) are given at the age of 10 and
tail, then the other hind limb is affec­ted and 16 years. Subsequently, immunity to tetanus
then generalized signs appear. The animal dies can be maintained by booster doses of tox­oid
within 2 days but may be killed earlier as the every 10 years.
appearance of ascending teta­nus is diagnostic. ii. Passive immunization:
a. Antitetanus serum (ATS): ATS from
Prophylaxis hyperim­mune horses was the preparation
The available methods of prophylaxis are: originally used. The dose employed was 1500
1. Surgical attention IU given subcutaneously or intramuscularly
2. Antibiotics in nonimmune persons soon after re­ceiving
3. Immuniza­tion—passive, active or combined. any tetanus prone injury. ATS gives passive
1. Surgical prophylaxis: This includes prompt and protection for about 7–10 days.
adequate wound toilet and proper surgical debri­ Disadvantages of ATS: Equine ATS carried
dement of wound, removal of foreign material, two disadvantages implicit in the use of any
necrotic tis­sue and blood clots to prevent an heterologous serum - ‘immune elimination
anaero­bic environment for the growth of C. lelani. and hypersensitivity reactions. It is,
2. Antibiotic prophylaxis: Antibiotics destroy therefore, mandatory to test for hypersen­
or inhibit C. tetani and pyogenic bacteria sitivity before administration of ATS.
in the wound, so that the production of the b. Human antitetanus immunoglobulin
toxin can be prevented. Long-acting penicillin (HTIG): Passive immunity without risk of
injection is the drug of choice. An alternative hypersensitivity can be obtained by the use
is erythromycin. Antibiotic prophylaxis does of human antitetanus immunoglobulin
not replace immunoprophylaxis but serves as (HTIG). This is effective in smaller doses.
a useful adjunct. The prophylac­tic dose of hTIG is 250 units
3. Immunoprophylaxis by intramuscular injection.
It includes 3 types of immunization: iii. Combined immunization: It consists of
i. Active immunization administering to a nonimmune person ATS or
ii. Passive immunization hTIG at one site, along with the first dose of a
iii. Combined immunization course of active immunization with adsorbed
i. Active immunization: Tetanus is best toxoid at the same time at another site,
prevented by active immunization with tetanus followed by second and third doses of TT at
toxoid. Two preparations are available for active appropriate intervals. The active immunization
immunization are: course must be subsequently completed.
a. Combined vaccine (DPT)
Treatment
b. Monovalent vaccines
∙∙ Plain or fluid (formol) toxoid 1. The patient remains conscious and requires
skilled seda­tion and constant nursing.
∙∙ Tetanus vaccine, adsorbed (PTAP, APT)
Tetanus toxoid (formol toxoid), which is 2. Treatment of tetanus requires debridement
available either as ‘plain toxoid’, or adsorbed of the primary wound, use of metronidazole,
on aluminum hydroxide or phosphate (APT), passive immunization with human teta­nus
is commonly used for active immunization. immunoglobulin, and vaccination with tetanus
Three doses of 0.5 ml tetanus toxoid tox­oid.
(APT) each are given intramuscularly, 3. Full wound exploration and debridement is
with an interval of 4–6 weeks between first arranged, and the wound is cleansed and left
two doses and 6–12 months between the open with a loose pack.
second and third dose. A full course of three 4. The patient is given 10,000 units of human
doses confers immunity for a period of at tetanus immunoglobulin (HTIG) in saline by
least 10 years. A ‘booster dose’ of toxoid is slow intravenous infusion.
recommended after 10 years. 6. Penicillin or metronidazole is given for as long
Tetanus toxoid is given along with diphtheria as considered necessary to ensure that bacterial
toxoid and pertusis vaccine called DPT in growth and toxin production are stopped.
children as tri­ple vaccine. Pertusis vaccine 7. Vaccination with a series of three doses of
acts as adjuvant. Three doses are given tetanus toxoid followed by booster doses every
intramuscularly at interval of 4–6 weeks, 10 years is highly effective in preventing tetanus.

www.ebook3000.com
218  |  Section 3: Systemic Bacteriology
Clostridium botulinum The toxin is relatively stable being inactivated
at 80°C for 30­–40 minutes and at 100°C for 10
C. botulinum (from the Latin botulus, “sausage”) minutes. Food suspected to be contaminated with
causes botulism. Botulism is a severe, often fatal, bot­ulinum toxin can be rendered completely safe
form of food poisoning characterized by pronounced by pres­sure cooking or boiling for 20 minutes. It can
neurotoxic effects. The disease has been caused be toxoi­ded. It is a good antigen and is specifically
by a wide range of foods, the disease can be a pure neutral­ized by the antitoxin.
intoxication. Botulinum toxin gains access to the peripheral
nervous system, where it acts preferentially on
Morphology cholinergic nerve endings to block the release of
C. botulinum is a strictly anaerobic gram-positive the neurotransmitter, acetylcholine, from the nerve
bacillus (about 5 × 1 mm). It is non-capsulated, ter­minals of neuromuscular junctions. A symmetric
motile with peritrichous flagella and produces descending paralysis is the characteristic pattern,
spores which are oval, subterminal and bulging. ending in death bv respiratory paralysis.

Cultural Characteristics Pathogenicity


It is a strict anaerobe. Opti­mum temperature is It is noninvasive and its pathogenicity is entirely
35°C but some strains may grow even at 1–5°C. due to the toxin produced by it. The dis­ease caused
Good growth occurs on ordinary media. Surface by this organism is known as botulism. It is of 3
colonies are large, irregular, semi-transparent, types—foodborne botulism, wound botulism and
with fimbriate border. On horse blood agar, all infant botulism.
strains except those of type G are β-hemolytic. On
egg-yolk agar (EYA) all types except G produce 1. Food-borne Botulism
opalescence and a pearly effect. It is due to the inges­tion of preformed toxin. The
Resistance: Spores are heat and radiation resistant, source of botulism is usually preserved food such
surviving several hours at 100°C and for up to 10 as meat and meat products, fish, and vegeta­bles.
minutes at 120°C. Spores of nonproteolytic types Food responsible for botulism is usually abnor­mal
of B, E and F are much less resistant to heat. The in appearance and odour.
resistance of the spores to radiation is of special Symptoms usually begin 18–36 hours after inge­
relevance to food processing. stion of food and may include nausea, vomiting,
thirst, constipation, double vision, difficulty in
swal­lowing, speaking and breathing. This may be
Classification
fol­lowed by muscular weakness, blurred vision,
The species C. botulinum has been divided into and death as a result of respiratory failure. Case
eight serologically distinct types—A, B, C1, and fatality varies from 25–70%.
C2, D, E, F, and G-on the basis of the type of toxin
produced. The toxins produced by different types
2. Wound Botulism
are antigenically dis­tinct but pharmacologically
similar. Wound botulism is a very rare condition resulting
from wound infection with C. botulinum. Toxin is
pro­duced at the site of infection and is absorbed.
Botulinum Toxin
The symptoms are those of foodborne botulism
A powerful exotoxin produced by C. botulinum is except for the gastrointestinal components which
responsible for its pathogenicity. It differs, however, are absent. Type A has been responsible for most
from a classic exotoxin in that it is not released of the cases studied.
during the life of the organism but appears in
the medium only after death and autolysis of the 3. Infant Botulism
organism. It is believed to be synthesised in­itially
This is a toxico-infection. C. botulinum spores are
as a nontoxic protoxin or progenitor toxin. Trypsin
ingested in food, get established in the gut and
and other proteolytic enzymes activate pro­genitor
there produce the toxin. The disease typically
toxin to active toxin. The production of botulinum
affects infants younger than 1 year (most between
toxin is governed by specific bacteriophages.
me and 6 months). The most common food source
Properties of toxin: Botulinum toxin is one of the in infant botulism is honey contaminated with
most potent toxins known. It is isolated as a pure botulinum spores.
crystalline protein with MW 70,000. It has a lethal Clinically, infant botulism is an acute flaccid
dose for mice of 0.000,000,033 mg and lethal dose for paralysis. After a period of normal develop­ment;
human be­ings is probably 1–2 mg. It is a neurotoxin the infant develops constipation, listlessness,
and acts slowly, taking several hours to kill. difficulty in sucking and swallowing, weak or
Chapter 30: Clostridium  | 219
alte­red cry, muscle weakness, ptosis, and loss of Treatment
head control. Eventually the baby appears ‘floppy’ 1. Elimination of the organism from the gastro­
(floppy child syndrome) and develops respiratory intestinal tract through the judicious use of
insuffi­ciency or respiratory arrest. Fulminant forms gastric lavage and metronidazole or penicillin
may resemble the sudden infant death syndrome therapy.
(SIDS or crib death). 2. The use of polyvalent antitoxin to neutralize
Management consists of supportive care and unfixed toxin.
assisted feeding. 3. Adequate ventilatory support.

Laboratory Diagnosis Clostridium difficile


Botulism confirmed by isolating the organism or C. difficile was first isolated in 1935 from the feces
detect­ing the toxin in food products or the patient’s of newborn infants. It was so named because of the
feces or serum. unu­sual difficulty in isolating.
1. Specimens: Fe­ces, food, vomitus, gastric fluid,
serum, environmental samples and occasionally Morphology
wound exudate.
C. difficile is a motile Gram-positive rod with oval
2. Culture: For the isolation of C. botulinum,
subter­minal spores. Spores are large, oval and
the specimen is inoculated on Egg-yolk agar
terminal. It is nonhemolytic, saccharolytic and
(EYA), blood agar and CMB. The strains of C.
weakly’ proteolytic.
botulinum associated with human botulism are
characterized by lipase production (appears Pathogenesis
as an iri­descent film on colonies grown on
Toxins: The organism produces an enterotoxin
egg-yolk agar) as well as the ability to digest
(toxin A) and a cytotoxin (toxin B).
milk proteins, hydrolyze gelatin, and ferment
1. Toxin A is an enterotoxin that is primarily
glucose.
responsible for diarrhea. It is capable of pro­
Presence of bacilli in food or feces in absence
ducing fluid accumulation in ligated rabbit ileal
of toxin is of no significance. Hence, toxin
loop assay.
in culture fluid must be demonstrated by
2. Toxin B is a potent cytotoxin capable of
toxigenicity test in mice.
producing cytopathogenic effects in several
3. Demonstration of toxin: Demonstration
tissue culture cell lines.
of toxin production must be done with a
mouse bioassay. This procedure consists of Pathogenesis: It is a proven cause of antibiotic
the preparation of two aliquots of the isolate, associated diarrhea (AAD), and pseudomembranous
mixing of one aliquot with antitoxin, and colitis (PMC) a life-threatening condition. The
intraperitoneal inocula­tion of each aliquot into disease develops in people taking antibiotics.
mice. Toxin activity is confirmed if the antitoxin The three drugs most commonly implicated are
treat­ment protects the mice. Control animals clindamycin, ampicillin and the cephalosporins.
protected by polyvalent antitoxin re­m ain The severity of disease varies widely from mild
healthy. Typing is done by passive protection diarrhea through varying degrees of inflammation
with type specific antitoxin.Samples of the of the large intestine to a fulminant PMC.
implicated food, stool specimen, and patient’s
serum should also be tested for toxin activity. Laboratory Diagnosis
A retrospective diagnosis may be made by 1. Isolation of bacilli: C. difficile can be isolated
detec­tion of antitoxin in the patient’s serum but it from the feces by enrich­ment and selective
may not be seen in all cases. culture procedures.
2. Demonstration of toxin: Toxin B can be
Prophylaxis demonstrated in the feces of patients by its
Control can be achieved by proper canning and characteristic effect on Hep-2 and human
preservation. Children younger than 1 year should diploid cell cultures or both toxins may be
not eat honey. demonstrated by immunological methods, e.g.
A prophylactic dose of polyvalent antitoxin enzyme-linked immunosorbent assay (ELISA).
should be given intramuscularly to all persons who
have eaten food suspected of causing botulism. Treatment and Prophylaxis
Active immunization should be considered The disease is treated by discontinuing the
for laboratory staff. Two injections of aluminum antibiotic that is pre­sumed to have precipitated
sulfate adsorbed toxoid may be given at an interval the disease and by giving oral metronidazole or
of ten weeks, followed by a booster a year later. vancomycin.

www.ebook3000.com
220  |  Section 3: Systemic Bacteriology

Key Points i. Botulism


j. Botulinum toxin
™™ The genus Clostridium includes all anaerobic, gram- k. Laboratory diagnosis of botulism
positive bacilli capable of forming endospores. l. Clostridium difficile.
Clostridia are more commonly associ­ated with m. Pseudomembranous colitis.
skin and soft tissue infections, food poison­ing, and n. Antibiotic associated colitis (diarrhea).
antibiotic-associated diarrhea and colitis
Clostridium perfringens
™™ Large, rectangular, Gram-positive bacillus. Forms
Multiple choice questions (MCQs)
spores but they are rarely seen in clinical spec­imens 1.  Which of the following types of Clostridium
or culture perfringens produces alpha toxin most abundantly?
™™ Blood agar and Robertson’s cooked meat medium a. Type A b. Type B
are used. Large spreading colonies are seen within c. Type C d. Type D
first day of culture; “double zone” of hemolysis on
2. Nagler’s reaction is useful for the identification of:
blood agar (due to a and 8 toxins)
a. Clostridium tetani
™™ Toxins: C. perfringens is forming at least 12 distinct
b. Clostridium perfringens.
soluble substances or toxins. The four major toxins,
c. Clostridium botulinum.
al­pha, beta, epsilon and iota, are predominantly
d. Clostridium difficile.
respon­sible for pathogenicity
™™ Diseases: 1. Soft tissue infections (cellulitis, suppurative
3. All the following species cause gas gangrene except:
myositis, my­onecrosis). 2. Food poisoning. 3. Septi­ a. Clostridium perfringens
cemia. b. Clostridium novyi
™™ Nagler reaction and reverse CAMP tests are useful in c. Clostridium sordellii
identification of C. perfringens d. Clostridium histolyticum
™™ Systemic infections require surgical debridement 4. Food poisoning is caused by Clostridium perfringens:
and high-dose penicillin therapy; antiserum against a. Type A b. Type B
a toxin not used now c. Type C d. Type D
Clostridium tetani 5. Stormy clot reaction is useful in identification of:
™™ Clostridium tetani is the causative agent of tetanus. a. Clostridium perfringens
™™ Tetanus toxin: Two distinct toxins–an oxygen– b. Clostridium tetani
labile hemolysin (tetanolysin) and a neurotoxin c. Clostridium botulinum
(tetanospasmin) d. Clostridium difficile
™™ Prevention through use of vaccination, consisting of 6. Typical drumstick appearance of bacilli is observed
three doses of tetanus toxoid followed by boosters in:
every 10 years a. Clostridium perfringens
Clostridium botulinum b. Clostridium tetani
™™ Clostridium botulinum forms a powerful which is c. Clostridium botulinum
responsible for botulinum d. Clostridium histolyticum.
™™ Diseases: The dis­ease caused by this organism 7. The most potent naturally occurring toxin known
is known as botulism. It is of 3 types: foodborne to human­kind is:
botulism, wound botulism and infant botulism a. Botulinum toxin b. Tetanus toxin
Clostridium difficile
c. Cholera toxin d. Diphtheria toxin
™™ The three drugs most commonly implicated are
8. Antibiotics-associated diarrhea is caused by:
clindamycin, ampicillin and the cephalosporins.
a. Clostridium perfringens
™™ Diseases: 1. Asymptomatic colonization; 2. Antibiotic-
b. Clostridium tetani
associated diarrhea (AAD); 3. Pseudomembranous
colitis. c. Clostridium botulinum
d. Clostridium difficile
9. Clostridium botulinum food poisoning is due to:
Important questions a. Preformed toxin
1. Classify clostridia. Discuss the laboratory diagnosis b. Invasion of bacteria in the intestine
c. Both of the above
of gas gangrene.
d. None of the above
2. Discuss the pathogenicity and prophylaxis of gas
10. Floppy child syndrome is associated with:
gangrene.
a. Clostridium botulinum infection
3. Define gas gangrene and discuss its bacteriology
b. Clostridium tetani infection
and pathogenesis
c. Clostridium perfringens infection
4. Enumerate various pathogenic clostridia. Describe d All of the above
the pathogenesis and laboratory diagnosis of 11. Which of the following bacteria is responsible for
tetanus. pseudomembranous enterocolitis?
5. Write short notes on: a. Clostridium perfringens b. C. tetani
a. Alpha toxin c. C. botulinum d. C. difficile
b. Stormy clot reaction 12. Antitoxic therapy is useful in:
c. Nagler reaction a. Gas gangrene b. Diphtheria.
d. Gas gangrene. c. Both of the above d. None of the
e. Toxins of C. tetani above
f. Tetanospasmin
g. Prophylaxis of tetanus. Answers (MCQs)
h. Clostridium botulinum 1. a; 2. b; 3. d; 4. a; 5. a; 6. b; 7. a; 8. d; 9. a; 10. a; 11. d; 12. c
31
Chapter

Nonsporing Anaerobes

Learning Objectives

After reading and studying this chapter, you should ∙∙ List infections caused by nonsporing anaerobes
be able to: ∙∙ Discuss laboratory diagnosis of infections caused
∙∙ Describe classification of nonsporing anaerobes by nonsporing anaerobes.

Introduction Table 31.1  Classification of nonsporing anaerobes


The anaerobic bacteria are widespread in nature. I. Cocci
A. Gram-positive
A bewildering range of anaerobes is found in the a. Peptostreptococcus
mouth and oropharynx, gastrointestinal tract b. Peptococcus
and female genital tract of healthy individuals as B. Gram-negative
part of the commensal flora. They constitute the Veillonella
predominant part of our normal indigenous flora II. Bacilli
1. Endosoore forming
on skin and membrane surfaces and outnumber Clostridia
facul­tatively anaerobic bacteria in the gut by a 2. Nonsporing
factor of 1000:1. The numbers of anaerobes present A. Gram-positive
have been estimated to be 104 to 105/mL in the small a. Eubacterium
intestine, 108/mL in saliva and 1011/g in the colon. b. Propionibacterium
c. Lactobacillus
On the skin, in the mouth, in the upper respiratory d. Mobiluncus
tract, and often in the female lower genital tract, e. Bifidobacterium
they outnumber facultatively anaerobic bacteria f. Actinomyces
by a factor of 5: 1 to 10: 1. Many of these anaerobic B. Gram-negative
organisms. Previously considered to be harmless a. Bacteroides
b. Prevotellla
commensals of our indigenous flora, are now c. Porphyromonas
recognized as opportunistic pathogens that may d. Fusobacterium
produce disease when the host’s resistance is e. Leptotrichia
reduced. III. Spirochetes
a. Treponema
b. Borrelia
Classification
Medically important anaerobes may be broadly
Gram-positive anaerobic cocci comprise
classified as shown in Table 31.1.
part of the normal microbial flora of the mouth,
gastrointestinal tract, genitourinary tract and skin.
Anaerobic cocci
Strictly anaerobic gram-positive cocci have Peptococcus
been assigned to the genera Peptococcus, Most of the peptococci have now been reclassified
Peptostreptococcus, Coprococcus, Ruminococcus as peptostreptococci. Peptococcus niger is the
and Sarcina. Strictly anaerobic gram-negative only surviving member of the genus Peptococcus.
cocci are included in Veillonella, Megasphera and They are gram-positive, nonsporing, anaerobic
Acidaminococcus. cocci, that occur singly or in pairs or in clusters.

www.ebook3000.com
222  |  Section 3: Systemic Bacteriology
They produce black colonies on blood agar on They are found as part of the normal flora of
prolonged incubation due to the production of the mouth, stomach, intestines, and genitourinary
H2S. They occur as normal flora of skin, intestine tract. Lactobacilli are normally present in the mouth
and genitourinary tract. They may cause pyogenic and have been incriminated in the pathogenesis
infections of wounds, puerperal sepsis and urinary of dental caries. It is believed that lactobacilli
tract infections. ferment sucrose to produce lactic acid, which
dissolves the mineral components of enamel and
Peptostreptococcus dentine causing dental caries.
Several species of lactobacilli are present in the
They are cocci of small size (0.2­–2.5 µm). Many of
intestine, the most common being L. acidophilus.
them are aerotolerant and grow well under 10%
Intestinal lactobacilli are beneficial in synthesizing
CO2 in an aerobic atmosphere.
vitamins, such as biotin, vitamin B12 and vitamin K,
Peptostreptococcus anaerobius is most often
which may be absorbed by the host.
responsible for puerperal sepsis and Pst. magnus
for abscesses. Lactobacillus species are major members of
the normal flora of the vagina and, typically, in
the adult vagina and these are collectively known
Gram-negative anAerobic cocci as Doderlein’s bacilli. They ferment the glycogen
Veillonella deposited in the vaginal epithelial cell and form
lactic acid, which accounts for the highly acidic
Veillonellae are gram-negative cocci of varying
pH of vaginal mucus epithelia. They protect
sizes, and measure 0.3–2.5 μm in diameter.
adult vagina from infections. In prepubertal and
Occurring as diplococci, short chains or groups.
postmenopausal vagina, lactobacilli are scanty.
They are normal inhabitants of the mouth,
It is generally nonpathogenic though L. cat­
intestinal and genital tracts. Veillonella parvula
enaforme has been associated with bronchopulmo­
has been reported from clinical specimens but its
nary infections. Lactobacilli are acidophilic
pathogenic role is uncertain.
and grow best at acidic pH. Some species have
strict growth require­ments and are used for the
Anaerobic, nonspore-forming, microbiological assay of growth factors (vitamins).
gram-positive bacilli
This group contains many genera, of which medically 4. Mobiluncus
relevant are Eubacterium, Propionibacterium, Members of the genus Mobiluncus are obligate
Lactobacillus, Mobiluncus, Bifidobacterium and anaero­bic, gram-variable or gram-negative, curved
Actinomyces. bacilli with tapered ends. Two species, Mobiluncus
curtisii and Mobiluncus mu­lieris, have been identified
1. Eubacterium­ in humans.
Mobiluncus curtisii and Mobiluncus mu­lieris
Members of the genus Eubacterium are strictly
have been isolated from the vagina in bacterial
anaerobic and grow very slowly. They are members
vaginosis, along with Gardnerella vaginalis. The
of the normal mouth and intestinal flora. Some
organisms colonize the genital tract in low numbers
species (E. brachy, E. timidum and E. nodatum)
but are abun­d ant in women with bacterial
are commonly seen in periodontitis. E. lentum is
vaginosis. Their microscopic appearance is a useful
commonly isolated from nonoral clinical specimens.
marker for this disease, but the precise role of these
organisms in the pathogenesis of bacterial vaginosis
2. Bifidobacterium is unclear.
Bifidobacterium is gram-positive bacilli nonmotile,
nonsporing and pleomorphic, showing true and 5. Propionibacterium
false branching. They occur as normal flora of Propionibacterium species are members of the
mouth, gastrointestinal tract (GI) tract and genito­ normal flora of the skin and cause dis­ease when
urinary tract. They are usually nonpathogenic. they infect plastic shunts and appliances. Their
metabolic products include propionic acid, from
3. Lactobacillus which the genus name derives.
Lactobacilli are gram-positive bacilli, straight or Species: The two most commonly isolated species
slightly curved which frequently show bipolar and are Propionibacterium acnes and Propioni­
barred staining. They are nonsporing and most bacterium propionicus.
strains are nonmotile. They form considerable P. acnes are responsible for acne and
amount of lactic acid from carbohydrates and grow oportunistic infections in patients with prosthetic
best at pH of 5 or less. devices or intravascular lines.
Chapter 31: Nonsporing Anaerobes  | 223
Propionibacterium propionicus mouth and upper alimentary and respiratory tracts.
Causes lacrimal canaliculitis and abscesses. Its cell wall contains a strong endotoxin. Prevotella
infections are usually associated with the upper
6. Actinomyces—See Chapter 35. respiratory tract, causing, for example, dental
and sinus infections, pulmonary infections and
Anaerobic gram-negative bacilli abscesses, brain abscesses, and infections caused
Gram-negative, anaerobic,nonsporeforming, by a human bite.
non motile rods were previously classified in the Laboratory identification is similar to that of B.
the family Bacteroidaceae within three genera, tragilis. In addition, P. melaninogenica forms black
Bacteroides, Fusobacterium and Leptotrichia. colonies on blood agar—a characteristic from which
its name was derived. The color is not due to the
1. Bacteroides melanin pigment as was once thought. Cultures of
The genus Bacteroides can be divided. P. melaninogenica and even dressings from wounds
infected with the bacillus give a characteristic red
a. Bile-­tolerant fluorescence when exposed to ultraviolet light. P.
i. Members of the B. fragilis group—B. fragilis, B. melaninogenica exhibits less drug resistance than
ovatus, B. distasonis, B. vulgatus, B. thetaiotao­ do the bac­teroides, and are usually susceptible to
micron, and others. penicillin.
ii. Two other species—Bacteroides eggerthii and
Bacteroides splanchnicus. 2. Porphyromonas
b. Bile Sensitive species Porphyromonas species can be cultured from
i. Pigmented: Have been reclas­sified into—the gingival and periapical tooth infections and, more
genera Porphyromonas, Prevotella and commonly, breast, axillary, perianal, and male
porphyromonas. genital infections.
ii. Nonpigmented species: Many nonpigmented
species of Bacteroides also have been transferred ii. Nonpigmented Species
to the genus Prevotella. Many nonpigmented species of bacteraies have
been transferred to the genus prevotella.
2. Fusobacterium (Bacilli with pointed ends) Nonpigmented Prevotella spp. includes
Fusobacterium necrophorum, Fusobacterium Prevotella bivia, Prevotella buccae, etc.
nucleatum, Fusobacterium necrogenes.

3. Leptotrichia (Large bacilli) 2. Fusobacterium


Leptotrichia buccalis—the only species. Fusobacteria are spindle-shaped gram-negative
bacilli with pointed ends a morphology character­
1. Bacteroides
istically referred to as fusiform. They are found as
a. Bile-tolerant Species part of the normal flora of the mouth, female genital
Members of the B. fragilis group, which contains tract, and colon. They grow slowly in vivo, and are
about 10 related species, are especially pathogenic. therefore of limited virulence.
B. fragilis is the most common species of anaerobic Species: i. F. nucleatum: It is frequently recovered
bacteria isolated from infectious processes of soft from mixed infections of the head and neck region,
tissue and anaerobic bacteremia. including dental abscesses and the central nervous
B. fragilis is the most frequent of the nonsporing system, from transtracheal aspirates and pleural
anaerobes isolated from clinical specimens. It is fluid.
often recovered from blood, pleural and peritoneal
ii. F. necrophorum: Is an important animal
fluids, CSF, brain abscesses, wounds and urogenital
pathogen.
infec­tions. Their polysaccharide capsule is an
important virulence factor, conveying resistance
3. Leptotrichia
to phagocytosis.
They are long, straight or slightly curved rods, often
b. Bile-sensitive Species with pointed ends. This species was originally
i. Pigmented species classified in the genus Fusobacterium, and was
formerly known as Vin­cent’s fusiform bacillus or
1. Prevotella Fusobacterium fusiforme. The genus Leptotrichia
Prevotella spp. appears as gram-negative coccobacilli contains the single spe­c ies, L. buccalis. The
or bacilli, very similar to Bacteroides spp. Prevotella association of L. buccalis with disease is not clearcut,
melaninogenica is part of the normal flora of the although it has been reported in acute necrotizing

www.ebook3000.com
224  |  Section 3: Systemic Bacteriology
ulcerative gingivitis (Vincent’s gingivitis) or A. Specimen Collection and Transport
Vincent’s angina, together with Treponema, Specimens should be collected in such a manner as
Porphyromonas and Fusobacterium species. It is to avoid resident flora. For example, from a suspected
seen in patients with malnutrition, debility and case of lung abscess the sputum is unsatisfactory for
poor oral hygiene. It is characterized by pain, culture and only material collected by aspiration
hemorrhage, foul odor, destruction of interdental. would be acceptable. In general, material for
Vincent’s angina may resemble diphtheria, anaerobic culture is best ob­tained by tissue biopsy or
with the inflamed pharyn­geal mucosa showing a by aspiration using a needle and syringe. Aspirated or
grayish membrane which peels easily. l. buccalis tissue specimens are preferable to swabs whenever
and T. vincentii are present in the exudate and feasible. Swabs are generally un­satisfactory but
pseudomembrane, and diagnosis is made by direct where they are to be used, they should be sent in
microscopy. Stained smears show large fusiform Stuart’s transport medium.
and spiral bacilli. Ideally, specimens should be placed in an
anaerobic transport device that consists of a
Anaerobic infections tube or vial containing an anaerobic gas mixture
substituted for air. Specimens should be delivered
Most of the anaerobic bacteria that cause infection
immediately (within 20 minutes) for culture.
are members of our normal indigenous flora.
Recapping a sy­ringe and transporting the needle
Anaerobic infections are usually endogenous and
and syringe to the lab­oratory is no longer acceptable
are caused by tissue invasion by bacteria nor­mally
because of safety concerns involving needle stick
resident on the respective body surfaces. Anaerobic
injuries. Therefore, even aspirates must be injected
bacteria are normally present on the skin, mouth,
into some type of oxygen-free transport tube or vial.
nasopharynx and upper respiratory tract, intestines
In some laboratories, gas-liquid chro­matography
and vagina (Table 31.2).
is carried out directly on pus and other clini­cal
Anaerobic infec­tions generally follow some specimens in order to detect metabolic products,
precipitating factor, such as trauma, tissue necrosis, such as butyric and propionic acids, that are
impaired circulation, he­matoma formation or the characteristic of certain anaerobes.
presence of foreign bodies. Diabetes, malnutrition,
malignancy or prolonged treatment with aminoglyco­ B. Direct Microscopy
side antibiotics, corticosteroids and cytotoxic agents
Examination of a gram stained smear is very useful.
may act as predisposing factors. Anaerobic infections
Pus in anaerobic infection usually shows a large
are typically polymicrobial.
variety of different organisms and numerous pus
Table 31.3 lists the common sites and type of cells.
anaerobic infections and the bacteria responsible. Examination of the specimen under ultraviolet
light may show the bright red fluorescence of P.
Laboratory Diagnosis melaninogenica.
As anaerobes form part of the normal flora of the
skin and mucous surfaces, their isolation from C. Culture
specimens has to be interpreted cautiously. The Several special media have been described for
mere presence of an anaerobe does not prove its anaerobes but for routine diagnostic work, freshly
causal role. prepared blood agar with neomycin, yeast extract,

Table 31.2  Normal anaerobic flora of the human body


Anaerobe Skin Mouth–Nasopharynx Intestine Vagina
Clostridium ++
Actinomyces +
Bifidobacterium + ++ +
Propionibacterium ++
Bacteroides fragilis ++
P. melaninogenica ++ + ++
Fusobacterium ++ +
Gram-positive cocci ++ ++ ++
Gram-negative cocci ++ + ++
Spirochetes +
Chapter 31: Nonsporing Anaerobes  | 225
Table 31.3  Selected infections typically involving nonsporing anaerobes
Site Type of infection Bacteria commonly responsible
A. Central nervous system Brain abscess B. fragilis; Peptostreptococcus
B. Ear, nose, throat Chronic sinusitis, otitis media, mastoiditis, Fusobacteria (aerobes frequently
orbital cellulitis responsible)
C. Mouth and jaw Ulcerative gingivitis (Vincent’s) Fusobacteria spirochetes, mouth
Dental abscess, cellulitis; anaerobes,
Abscess and sinus of jaw Actinomyces, other mouth anaerobes.
D. Respiratory Aspiration pneumonia, lung abscess, Fusobacteria, P. melaninogenica, anaerobic
bronchiectasis, empyema cocci; B. fragilis rarely
E. Abdominal Subphrenic abscess, hepatic abscess; B. fragilis
appendicitis, peritonitis;
ischiorectal abscess;
wound infection after
colorectal surgery
F. Female genitalia Wound infection following P. melaninogenica,
genital surgery; anaerobic occi; B. fragilis
Puerperal sepsis; Genital anaerobes and Cl. perfrinfens
tubo-ovarian abscess,
Bartholin’s abscess,
septic abortion
G. Skin and soft tissue Infected sebacious cyst. Anaerobic cocci.
Breast abscess, axillary abscess Anaerobic cocci; P. melaninogenica
(Staphylococcus aureus commonest cause)
Cellulitis, diabetic ulcer, gangrene B. fragilis and others

hemin and vitamin K is adequate. Plates are incu­ 3. An indication of antimicrobial agents likely to
bat­ed at 37°C in an anaerobic jar, with 10% CO2. The be used.
Gas-Pak system provides a convenient method of
routine anaerobic cultures. Plates are examined after E. Antibiotic Sensitivity Tests
24 or 48 hours. Some anaerobes, such as fusobacte­ Antibiotic sensitivity tests can be done by disc
ria, require longer periods of incubation. Since many diffusion or dilution methods.
anaerobes are relatively slow-growing, it is essential
that cultures are incubated for several days before F. Other Anaerobic Techniques
being discarded. In mixed infec­tions, fast-growing Gloved anaerobic chambers with continuous gas
aerobic or facultatively anaerobic organisms are flow may be used for culture of specimens. Pre-
often detected within 24 hours, whereas some reduced anaerobically sterilised media (PRAS) can
anaerobes may require incubation for 7–10 days also be employed but are not essential for rountine
before their colonies can be recognized. diagnostic procedures.
Other anaerobic media, such as cooked meat
broth (CMB) and thioglycollate broth, may also Treatment of Anaerobic Infections
be used for incoculating the specimens. Parallel Treatment of mixed anaerobic infections is by
aerobic cultures (such as Pseudomonas aeruginosa surgical drainage (under most circumstances) plus
should always be set up. This is nec­essary as a antimicrobial therapy.
control for the growth on anaerobic plates and The most active drugs for treatment of anaerobic
also because in most anaerobic infections aero­bic infections are clindamycin and metronidazole.
bacteria are also involved. Clin­damycin is preferred for infections above the
D. Identification diaphragm. Penicillin G remains the drug of choice
Colony morphology, pigmentation, and fluorescence for treatment of anaerobic infections that do not
are helpful in identifying anaerobes. Biochemical involve β-lactamase-producing Bacteroides and
activi­ties and production of short-chain fatty acids Prevotella species.
as mea­sured by gas-liquid chromatography are used
Key Points
for labora­tory confirmation. It takes time and is
difficult, but it is possible to report on the following: ™™ Many anaerobic bacteria are pathogenic for human
1. Whether the infection is solely aerobic, anaerobic beings, and they outnumber aerobic bacteria in
or mixed. many habitats
™™ Anaerobic gram-positive cocci: Most of important
2. The identification of the more common anaero­ anaerobic cocci belong to the genus Peptostreptococcus
bes, particularly of B. fragilis.

www.ebook3000.com
226  |  Section 3: Systemic Bacteriology
b. Lactobacillus
™™ Anaerobic gram-negative cocci: Veillonella parvula c. Bacteroides
is the species frequently reported from clinical d. Fusobacterium
specimens e. Anaerobic gram-positive bacilli.
™™ Anaerobic nonspore-forming gram-positive
bacilli: Include Eubacterium, Propionibacterium,
Lactobacillus, Bifidobacterium and Mobiluncus Multiple choice questions (MCQs)
™™ Anaerobic gram-negative bacilli: Medically
important anaerobic gram-negative bacilli belong 1. The following is an example of gram-negative
to the family Bacteroidaceae and are classified into anaerobic cocci
the genera Bacteroides, Prevotella, Porphyromonas, a. Veillonella
Fusobacterium and Leptotrichia b. Peptococcus
™™ Laboratory diagnosis: Specimens should be placed c. Peptostreptococcus
in an anaerobic transport device. Examination of d. Bacteroides
a Gram stained smear is very useful. The Gas-Pak 2. Lactobacilli constitute the normal flora of:
system provides a convenient method of routine a. Adult vagina
anaerobic cultures. Other anaerobic media, such b. Prepubertal vagina
as cooked meat broth (CMB) and thioglycollate c. Post-menopausal vagina
broth, may also be used. Colony morphology, d. None of the above
pigmentation, and fluorescence are helpful in
3. All the following anaerobic bacteria cause head
identifying anaerobes. Biochemical activi­ties and
production of short-chain fatty acids as mea­sured
and neckinfections except.
by gas-liquid chromatography are used for labora­ a. Bacteroides thetaiotaomicron
tory confirmation. b. Fusobacterium nucleatum
c. Fusobacterium necrophorum
d. Porphyromonas asaccharolytica
Important questions 4. Which of the following bacterial colonies fluoresce
brick-red in UV light?
1. Classify nonsporing anaerobes. Discuss the a. Bacteroides fragilis
laboratory diagnosis of infections caused by b. Bacteroides melaninogenicus
nonsporing anaerobes. c. Bacteroides gingiva/is
2. Give an account of infections caused by nonsporing d. Bacteroides levii.
anaerobes.
3. Write short notes on: Answers (MCQs)
a. Anaerobic cocci 1. (a); 2. (a); 3. (a); 4. (b)
32
Chapter

Mycobacterium tuberculosis

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Differentiate between Mycobacterium tubercu­losis
be able to: and M. bovis
∙∙ Classify mycobacteria ∙∙ Describe pathogenesis of Mycobacterium tuberculosis
∙∙ Discuss morphology and culture characteristics ∙∙ Discuss the laboratory diagnosis of pulmonary
and biochemical characteristics of Mycobacterium tuberculosis
tuberculosis ∙∙ Describe the following: Koch’s phenomenon;
Tuberculin test; BCG vaccine.

INTRODUCTION M. butyricum from butter, M. phlei from grass,


M. stercoris from dung and M. smegmatis from
The genus Mycobacterium belongs to the family smegma (Table 32.1).
Mycobacteriaceae. There are over 80 named
species of mycobacteria. The most familiar of
the species are Mycobacterium tuberculosis M. TUBERCULOSIS COMPLEX OR MTC
(MTB) and Mycobacterium leprae, the causative
M. tuberculosis is a member of the Mycobacterium
agents of tuberculosis (TB) and Hansen’s disease
tuberculosis complex, which also includes M.
(leprosy), respectively. The name mycobacterium,
bovis, M. africanum, M. canettii, and M. microti.
meaning ‘fungus like bacterium’ is derived from
Mycobac­t erium spp. produce a spectrum of
the mould-like appearance of Mycobacterium
infections in humans and animals in addi­tion to
tuberculosis when grow­ing in liquid media.
They are aerobic, nonmotile, noncap­sulated Table 32.1  Classification of mycobacteria
and nonsporing. Growth is generally slow. The
Tubercle bacilli
genus includes obligate parasites, opportunistic 1. Human - M. tuberculosis
pathogens and saprophytes (Table 32.1). They 2. Bovine - M. bovis
do not stain readily, but once stained with hot 3. Murine - M. microti
carbol fuchsin or other aryl methane dyes, they 4. Avian - M. avium
5. Cold blooded - M. marinum
resist decolorization with dilute mineral acids (or
Lepra bacilli
alcohol). Mycobacteria are, there­fore, known as 1. Human - M. leprae
acid-fast bacilli (AFB). 2. Murine - M.lepraemurium
The first member of this genus to be identified Mycobacteria causing skin ulcers
was the lepra bacillus discovered by Armal1er 1. M. ulcerans
2. M. balnei
Hansen in 1868. Robert Koch (1882) isolated
Atypical mycobacteria
the mammalian tubercle bacillus and proved 1. Photochromogens
its causative role in tuberculosis by satisfying 2. Scotochromogens
Koch’s postulates. Tuberculosis in hu­mans was 3. Nonphotochromogens
subsequently shown to be caused by two types 4. Rapid growers
of the bacillus—the human and bovine types, Johne’s bacillus
1. M. paratuberculosis
designated Mycobacterium tuberculosis and M.
Saprophytic mycobacteria
bo­vis respectively. Saprophytic mycobacteria were 2. M. butyricum. M. phlei, M. stercoris. M. smegmatis
isolated from a number of sources. These included

www.ebook3000.com
Chap-32.indd 227 15-03-2016 11:13:50
228  |  Section 3: Systemic Bacteriology
tuberculosis and Hansen’s disease. A large group
of mycobacteria, excluding the M. tuberculosis
complex and M. leprae, normally inhabit the
environment and can cause disease that often
resembles tuberculosis in humans. These organ­
isms are sometimes referred to as atypical
mycobacteria or mycobacteria other than the
tubercle bacillus (MOTI). Classification of mycobac­
teria are shown in Table 32.1.

MYCOBACTERIUM TUBERCULOSIS
Morphology
M. tuberculosis is a slender, straight, or slightly
curved rod with rounded ends, about 3 µm × 0.3 µm, Fig. 32.1: Mycobacterium tuberculosis in Ziehl–Neelsen
in pairs or as small clumps. The bacilli are nonmotile, stained smear
nonsporing, noncapsulated and acid-fast.
When stained with carbol fuchsin by the Ziehl–
Neelsen method or by fluorescent dyes (auramine
O, rhodamine), they resist decolorization by 20%
sulphuric acid and absolute alcohol for 10 minutes
(acid and alcohol fast). With Ziehl–Neelsen acid-
fast stain, the tubercle bacilli stain bright red, while
the tissue cells and other organisms are stained
blue (Fig. 32.1).
Acid fastness has been ascribed variously to
the presence in the bacillus of an unsaponifiable
wax (mycoloic acid) or to a sem­i permeable
membrane around the cell. It is related to the
integrity of the cell and appears to be a property
of the lipid-rich waxy cell wall. Staining may be
uniform or granular. In M. tuberculosis beaded or
barred forms are frequently seen, but M. bovis stains
more uni­formly. M. bovis appear straighter, stouter Figs 32.2: L–J media without growth (A) and with growth (B)
and shorter with uniform stainin (Table 32.2).
other bacteria and to provide a contrasting color
Cultural Characteristics against which colonies of mycobacteria are easily
seen. In this medium egg acts as a solidifying agent.
M. tuberculosis is an obligate aerobe while M. The addition of 0.5% glycerol improves the growth
bovis is microaerophilic on primary isolation, of M. tuberculosis, but has no effect on or may even
beco­ming aerobic on subculture. The optimal impair the growth of M. bovis. Sodium pyruvate
growth temperature of tubercle bacilli is 35–37°C. helps the growth of both types (Figs 32.2A and B).
Optimum pH is 6.4–7.0. The bacilli grow slowly, Human tubercle bacilli produce visible growth
the generation time in vitro being 14–15 hours. on LJ medium in about 2 weeks, although on
Colo­nies appear in about two weeks and may primary isolation from clinical material colonies
sometimes take up to eight weeks. may take up to 8 weeks to appear. On solid media,
M. tuberculosis forms dry, rough, raised, irregular
Solid Medium colonies with a wrinkled surface. They are creamy
Tubercle bacilli are able to grow on a wide range of white, becoming yellowish or buff colored on
enriched culture media and do not have exacting further incubation. They are tenacious and not
growth re­quirements. The solid media contain egg easily emulsified. Mycobacterium tuberculosis has
(Lowenstein Jensen, Petragnini, Dorset), blood a luxuriant growth (eugenic growth) as compared
(Tarshis), serum (Loeffler) or potato (Pawlowsky). to Mycobacterium bovis which grows poor­ly on LJ
The solid medium most widely employed glycerol medium (dysgonic growth) and colonies,
for routine culture is Lowen­stein–Jensen (L–J) in compari­son are flat, smooth, moist, white and
medium. This consists of coagulated hens’ egg, break up easily when touched. The growth of M.
mineral salt solution, asparagine and malachite bovis is much better on LJ pyruvate medium (media
green, the last acting as a selective agent inhib­iting containing sodium pyruvate in place of glycerol).

Chap-32.indd 228 15-03-2016 11:13:51


Chapter 32: Mycobacterium tuberculosis  | 229
Table 32.2  Distinguishing features of Mycobacterium tuberculosis and M. bovis
Character Mycobacterium tuberculosis M. bovis
1. Morphology Slender, straight or slightly curved Straight, stout, short uniformly
rods with barred or beaded
appearance
2. Growth on LJ medium Growth is eugonic (luxuriant) Growth is dysgonic
3. Effect of glycerol In the concentration of 0.75% In the concentration of 0.75% growth is
enhances the growth inhibited.
4. Oxygen requirement Obligate aerobe Microaerophilic on primary isolation but
becomes aerobic on subculture
5. Colony morphology Dry, rough, raised, wrinkled, off- Moist, smooth, flat, friable and white
white to buff colored and not easily
emulsifiable (rough, buff and tough)
6. Biochemical reactions
• Nitrate reduction + –
• Niacin production + –
• Tween 80 hydrolysis Variable –
7. Susceptibility to pyrazinamide + –
(50 µg/mL)
8. Susceptibility to thiophen­ – +
2-carboxylic acid
hydrazide (10 µg/mL )
9. Animal pathogenicity
• Guinea-pig, rabbit + +
– +

Liquid Media by heat (60°C for 15–20 min, or by autoclaving).


Among the several liquid media described,Dubos’, Cultures may be killed by exposure to direct
Middlebrook’s, Proskauer and Beck’s, Sula’s and sunlight for two hours, but bacilli in sputum may
Sauton’s media are the more com­mon. Dubos’ remain alive for 20–30 hours.
medium and Middlebrook 7H9 are two commonly Tubercle bacilli are relatively resistant to
used liquid media. Liquid media are not generally chemi­cal disinfectants, surviving exposure to 5%
employed for routine cultivation, but are used phenol, 15% sulphuric acid, 3% nitric acid, 5%
for sensitivity testing, chemical analyses and oxalic acid and 4% sodium hydroxide. They are
preparation of antigens and vaccines. sensitive to formaldehyde and gluteraldehyde.
In liquid media without dispersing agents the They are destroyed by tincture of iodine in five
growth begins at the bottom, creeps up the sides and minutes and by 80% eth­anol in 2–10 minutes.
forms a prominent surface pellicle which may extend
along the sides above the medium. Ordinarily, Biochemical Reactions
mycobacteria grow in clumps or masses because of
the hydrophobic character of the cell surface. Diffuse Several biochemical tests are used in identifying
growth is obtained in Dubos’ medium containing and differentiation of mycobacterial species.
Tween-80 (sorbitan monooleate). In liquid cultures 1. Niacin test: Most mycobacteria possess the
they often grow as twisted rope-like colonies termed enzyme that converts free niacin to niacin
serpentine cords. Virulent strains tend to form long ribonucleotide. M. tuberculosis lacks this
serpentine cords in liquid media, while avirulent enzyme and accumulates niacin (nicotinic
strains grow in a more dispersed manner. The cord acid) in the culture medium. The test is positive
factor itself is not a virulence factor, as it is present with hu­man type and negative with bovine type
in some avirulent strains as well. of bacilli.
2. Nitrate reduction test: M. tuberculosis, M.
Resistance kansasii, M. fortuitum, M. chelonei, M szulgai,
Although mycobacteria can survive for several M. flavescens, M. terrae and M. triviale produce
weeks in the dark, especially under moist enzyme nitroreductase. Therefore, all these
conditions, and for many days in dried sputum reduce nitrate to nitrite. M. avium do not do so.
on clothing and in dust, they are rapidly killed 3. Catalase activity: Catalase is an enzyme that
by ultraviolet light (including the component in can split hydrogen perox­ide into water and
daylight and sunlight), even through glass, and oxygen. Mycobacteria are catalase positive.

www.ebook3000.com
Chap-32.indd 229 15-03-2016 11:13:51
230  |  Section 3: Systemic Bacteriology
However, not all srains produce a positive
reaction after the culture is heated to 68° C for
20 minutes.
4. Tween 80 hydrolysis: Some mycobacteria
possess a lipase that can split detergent Tween
80 into oleic acid and polyoxyethylated sorbitol
which modifies the optical characteristics of
the test solution from a straw yellow to pink
whereas pathogenic species do not. This test is
useful in distinguishing scotochromogenic and
Fig. 32.3: Cell wall of Mycobacterium tuberculosis
nonchro­mogenic mycobacteria. M. tub­erculosis
gives variable result.
5. Arylsulfatase test: The enzyme is present in of the cell wall. The agglutinogen antigens
most mycobacteria. This test is positive only have been identified as the sugar moieties on
with atypical mycobacteria. mycosides.
6. Neutral red test: M. tuber­culosis, M. bovis, M. The cell wall antigens include arabi­nomanan,
avium and M. ulcerans give positive tests. arabinogalactan and lipoarabinomannan.
7. Amidase tests: Atypical mycobacteria can be
differentiated by their ability to split amides. 2. Cytoplasmic Antigens
A useful pattern is provided by testing five Cytoplasmic antigens are protein antigens which
amides, viz. acetamide, benzamide, carbamide, are employed to type the mycobacteria. These
nicotinamide and pyrazina­mide. include antigen 5, antigen 6, antigen 14, antigen
M. tuberculosis produces nicotinamidase and 19, antigen 32, antigen 38 and antigen 60. All are
pyrazinamidase which splits nicotinamide and protein in nature except antigen 60 which is a
pyrazina­mide. lipopolysaccharide protein complex.
8. Susceptibility to pyrazinamide: M. tuber­
culosis is sensitive to pyrazinamide, while M.
bovis and other mycobacteria are resistant. Mycobacteriophages
9. Growth inhibition by thiophen-2-carboxylic Numerous phages with activities on many
acid hydrazide (TCH): This test is used to mycobacterial species, including M. tuberculosis, have
distinguish M. bovis from M. tuberculosis, been isolated from both clinical and environmental
because only M. bovis is usually susceptible sources. Many mycobacteriophages have been
while M. tuberculosis is usually not susceptible isolated from soil, water and other environmen­
to this chemical. tal sources as well as from lysogenic strains. Many
my­cobacteria infected with phage are not truly lyso­
Antigenic Structure genic. Instead of being integrated with the bacterial
Mycobacteria possess two types of antigens, chromosome, the phage genome appears free, like
cell wall (insoluble) and cytoplasmic (soluble) a plasmid. This is called pseudolysogeny.
antigens. M. tuberculosis has been classified into four
phage types - A,B,C, and I (a type intermediate
1. Cell Wall Antigens between A and B). Phage type A is the most common
and is present worldwide. Type B is common in
The cell wall consists of lipids, proteins and
Europe, the Middle East and North America. Type
polysaccharides. The lipid content accounts for
C is seen rarely. Type I is common in India and
60% of the cell wall weight. The cell wall is made
neighboring countries. Phage 33D can lyse M.
up of four distinct layers (Fig. 32.3).
tuberculosis, M. bovis, M. afri­canum and M. microti.
i. Peptidoglycan (murein) layer: It is the
innermost layer which maintains the shape
and rigidity of the cell. Pathogenesis
ii. Arabinogalactan layer: It lies external to the The source of infection is usually an open case of
peptidoglycan layer. pulmonary tuberculosis. The mode of infection is
iii. Mycolic acid layer: It is the principal constituent by direct inhalation of aerosolized bacilli contained
of cell wall and is a dense band on long chain in droplet nuclei of expectorated sputum tubercle
a-alkyl and β-­hydroxy fatty acids attached by bacilli are acquired from persons with active
ester bonds to the terminal arabinose units of disease who are excreting viable bacilli by means
arabinogalactan. of coughing, sneezing, or talking. Infection also
iv. Mycosides (peptidoglycolipids or phenolic occurs infrequently by ingestion, for example,
glycolipids): These form the outermost layer through infected milk, and rarely by inoculation.

Chap-32.indd 230 15-03-2016 11:13:52


Chapter 32: Mycobacterium tuberculosis  | 231
The initial infection with M. tuberculosis is The reactivation type almost always begins at the
referred to as a primary infection. Subsequent apex of the lung, where the oxygen tension (PO2)
disease in a previously sensitized person, either from is highest.
an exogenous source or by reactivation of a primary Two special features of secondary tuber­
infection, is known as postprimary(secondary culosis are the presence of caseous necrosis and
or reinfection) tuberculosis with quite different of cavities, which may rupture into blood vessels,
pathological features. spreading mycobacteria throughout the body, and
break into airways, releasing infectious mycobac­
1. Primary Tuberculosis teria in aerosols and sputum (open tuberculosis).
The site of the initial infection is usually the lung,
following the inhalation of bacilli. These bacilli Epidemiology
engulfed by alveolar macrophages multiply and give Tuberculosis is an ancient disease. Tuberculosis
rise to a subpleural focus of tuberculous pneumonia, also remains the leading cause of death among
commonly located in the lower lobe or the lower part notifiable infectious diseases. It has been called
of the upper lobe to form the initial lesion or Ghon the ‘white plague’ and ‘the captain of all the men
focus. The Ghon focus, together with the enlarged of death’.
hilar lymph nodes, form the primary complex. In The most frequent source of infection is
addition, bacilli are seeded by further lymphatic and the human who excretes, particularly from the
hematogenous dissem­ination in many organs and respiratory tract, large numbers of tubercle bacilli.
tissues, including other parts of the lung. The large majority of the cases and deaths are from
Within about 10 days of infe­ction, clones of the poor nations. India accounts for nearly one-
antigen-specific T lymphocytes are produced third of global burden of tuberculosis.
interacting with macrophages. These release Poverty and tuberculosis go together. Tuber­
cytokines, notably interferon-gam­ma, which activates culosis had declined rapidly in the affluent nations
macrophages and cause them to form a compact with im­provements in standards of living. But with
cluster or granuloma, around the foci of infection. the progress of the AIDS pandemic, tuberculosis
These activated mainphages are called epitheloid became a problem for the rich nations also.
cells. M. bovis infection in humans is zoonotic
If there is little antigen and a great deal of acquired through direct contact or by ingestion
hypersensitivity reaction, a hard tubercle or of raw milk from animals. Bovine tuberculosis is
granuloma may be formed. When fully developed, spread from animal to animal.
this lesion, a chronic granuloma, consists of three
zones: Laboratory Diagnosis
1. A cen­tral area of large, multinucleated giant The definitive diagnosis of tuberculosis is based on:
cells containing tubercle bacilli. Microscopy, culture, by transmitting the infection
2. A mid zone of pale epithelioid cells, often to experimen­t al animals, demonstration of
arranged radially, and hypersensitivity to tuberculoprotein and mole­cular
3. A peripheral zone of fibroblasts, lymphocytes, diagnostic methods.
and monocytes.
Later, periph­eral fibrous tissue develops, and the Pulmonary Tuberculosis
central area under­goes caseation necrosis. Such a 1. Specimens
lesion is called a tubercle. A caseous tubercle may
a. Sputum: The most usual specimen for diagnosis
break into a bronchus, empty its contents there, and
of pulmonary tuberculosis is sputum. Patient is
form a cavity. It may subsequently heal by fibrosis
instructed to cough up the sputum into a clean
or calcification.
wide-mouthed container. Disposable waxed
cardboard containers are ideal. If sputum is
2. Postprimary (Secondary) Tuberculosis scanty, a 24-hour sample may be tested. Three or
The postprimary (secondary or adult) type more con­secutive samples should be examined,
of tuberculosis is due to reactivation of latent collected first thing in the morning if possible.
infection (postprimary progression, endogenous b. Laryngeal swabs or bronchial washings:
reactivation) or exogenous reinfection and differs Laryngeal swabs or bronchial washings may
from the primary type in many respects. be collected where sputum is not available.
Reacti­vation tuberculosis is characterized by c. Gastric lavage: Gastric lavage can be ex­amined in
chronic tissue lesions, the formation of tubercles, small children who tend to swallow the sputum.
caseation, and fibro­sis. Regional lymph nodes are Tissue biopsies are homogenized by
only slightly involved, and they do not caseate. grinding for microscopy and culture.

www.ebook3000.com
Chap-32.indd 231 15-03-2016 11:13:52
232  |  Section 3: Systemic Bacteriology
2. Microscopy acid fast bacilli provides only presumptive evidence
Direct or concentration smears of spu­tum are of tuberculous infection, as even saprophytic
examined. Smears should be prepared from the mycobacteria may present a similar appearance.
thick purulent part of the sputum. Smears are
dried, heat fixed and stained by the Ziehl–Neelsen 3. Concentration Methods
technique (hot stain procedure). Several methods have been described for the
In the Ziehl–Neelsen (Z–N) staining technique, homogenization and concentration of sputum
heat-fixed smears of the specimens are flooded and other specimens. Con­centration methods
with a solution of carbol fuchsin (a mixture of basic that do not kill the bacilli and so can be used for
fuchsin and phenol) and heated until steam rises culture and animal inoculation. Several meth­ods
but without boiling. Allow the preparation to stain are in use:
for 5 minutes, applying heat at intervals to keep
i. Petroff’s method: This simple method is widely
the stain hot. The slide is then washed with water
used. Equal volumes of sputum and 4% sodium
and decolorized with 20% sul­phuric acid till no
hydroxide are mixed and incubated at 37°C with
more stain comes off and then with 95% alcohol
frequent shaking till it gets liquefied and beco­
(ethanol) for two minutes. Decolorization may be
mes clear. It is then centrifuged at 3,000 r.p.m.
carried out as a single step with acid alcohol (3%
for 30 minutes. The supernatant fluid is pipetted
HCl in 95% ethanol). After washing, the smear is
off and the deposit is neutralized by adding 8%
counter­stained with Loeffler’s methylene blue, I%
hydrochloric acid in the presence of a drop of
picric acid or 0.2% malachite green for one minute.
phenol red indicator and used for smear, culture
Under the oil immersion objective, acid fast
and animal inoculation.
bacilli are seen as bright red rods while the
background is blue, yellow or green depending on ii. Other methods: Instead of alkali, homogenization
the counterstain used. At least 10,000 acid fast bacilli can be achieved by treatment with dilute acids (6%
should be present per mL of sputum for them to be sulphuric acid, 3% hydrochloric acid or 5% oxalic
readily demonstrable in direct smears. acid), N acetyl cysteine with NaOH, pancreatin,
Smears should be examined carefully by desogen, zephiran and cetrimide.
scanning at least 300 oil immersion fields before
reporting a smear as negative (Table 32.3). 4. Culture
The classic carbolfuchsin (Ziehl­–Neelsen) stain It is a very sensitive diagnostic technique for tu­bercle
requires heating the slide for better pene­tration of bacilli, detecting as few as 10–100 bacilli per mL. The
stain into the mycobacterial cell wall. Hence it is concentrated material is inoculated onto at least
also known as the hot stain procedure. Kinyoun two bottles of L–J medium. A direct drug sensitivity
acid-fast stain is similar to the Ziehl–Neelsen stain test may also be set-up if the specimen is positive
but without heat ; hence the term cold stain. by microscopy.
It is more convenient to use fluorescent
Inoculated media are incubated at 35–37°C.
microscopy. Smears are stained with auramine
Growth of most strains of M tuberculosis may
phenol or auramine rhodamine fluorescent
appear in 2–8 weeks.
dyes. Screening of specimens with rho­damine or
Cultures are examined for growth after
rhodamine-auramine will result in a higher yield
incubation at 37°C for four days (for rapid grow­ing
of positive smears and will substantially reduce
mycobacteria, fungi and contaminant bacteria)
the amount of time needed for examining smears.
and at least twice weekly thereafter. Any bacterial
In general, specificity of acid-fast smear exami­
growth is stained by the ZN method and, if acid-fast,
nation is very high. Microscopic demonstration of
it is subcultured for further identification. The first
Table 32.3  Methods for reporting numbers of acid step in identification is to determine whether an
fast bacilli observed in stained smears isolate is a member of the M. tuberculosis complex
(Fig. 32.4).
No. of bacilli Observed in (oil Report
immersion field)
For routine purposes, a slow growing, nonpig­
mented, ni­acin positive acid fast bacillus is taken
0 300 fields Negative
as M. tubercu­losis. Confirmation is by detailed
1–2 100 fields ± biochemical studies. When the isolate is niacin
1–9 100 fields 1+ negative, a battery of tests may be needed for
1–9 10 fields 2+ identification, including growth at 25°C and
1–9 1 field 3+
45°C, animal pathogenicity and biochemi­cal tests
(Tables 32.2 and 32.4).
More than 9 1 field 4+
In recent years, isolation of mycobacteria from
Fields—Oil immersion field clinical samples has been improved by newer

Chap-32.indd 232 15-03-2016 11:13:52


Chapter 32: Mycobacterium tuberculosis  | 233

Fig. 32.4: Identification of tubercle bacilli and related mycobacteria

Table 32.4  Some differential characteristics of tubercle bacilli causing human disease
Species Oxygen Glycerol Niacin Nitrate TCH Pyrazinamide Pathogenicity
preference enhanced reduction

M. Aerobic Yes Positive Positive Resistantb Sensitive Pathogen


tuberculosisa
M. bovis Micro- No Negative Negative Sensitive Resistant Pathogen
aerophilic
M. bovis BCG Aerobic Yes Negative Negative Sensitive Resistant Opportunistic
pathogen
M. africanum Micro- No Variable Variable Sensitive Sensitive Pathogen
aerophilic
a
Includes the rare M. canetti variant.
b
Strains from south India may be sensitive.
TCH = thiophen-2-carboxylic acid hydrazide.

tech­niques, notably radiometric respirometry. 5. Animal Inoculation


This is usually by means of the Bactec radiometric Sputum is decontaminated and concentrated by
tech­nique, using the Bactec 460 TB instrument. The Petroff’s method and 0.5 mL each of the neutralized
procedure utilizes vials containing a 14C-labeled and centrifuged deposit is inoculated intramuscu­
palmitic acid substrate which during microbial larly into the thigh of two healthy, tuberculin-nega­
growth releases 14CO2 into the atmosphere above tive guinea-pigs about 12 weeks old. The animals
the medium. The 14CO2 is detected by the instrument are weighed be­fore inoculation and at intervals
and converted proportionally to a quan­titative thereafter at weekly intervals and tuberculin test
Growth Index (GI) on a scale of 0–999. A vial giving is done after 3–4 weeks. One animal is killed after
a positive GI (> 50) is checked by AP and Z–N smears four weeks and autop­sied. If it shows no evidence
to determine the presence of acid-fast bacilli. The of tuberculosis, the other is autopsied after eight
sensitivity of radiometric method is slightly more weeks.
than that of traditional culture method. This method
allows a mean isolation time for all mycobac­teria of Necropsy shows the following:
about 12 days as opposed to 26 days or more. i. Caseous lesion at the site of inoculation.
Mycobacterial growth indicator tube (MGIT) is ii. The draining and internal lymph nodes are
another rapid method for detection of mycobacterial enlarged and caseous. The infection may
growth. Bact/Alert 3D system is a non-radimetric, spread to lumbar, portal, media­stinal and
rapid and fully automated. cervical lymph nodes.

www.ebook3000.com
Chap-32.indd 233 15-03-2016 11:13:52
234  |  Section 3: Systemic Bacteriology
iii. The spleen is en­larged with irregular necrotic methods, such as col­umn chromatography and
areas. Tubercles are seen in the peritoneum thin-layer chromatography, have been replaced
and sometimes in the lung, but the kidneys by gas–liquid chromatography and, most recently,
are unaffected. high-pressure liquid chromatography (HPLC).
The autopsy lesions have to be confirmed as Chromatography is rapid and highly reproducible,
tuberculous by acid fast staining of smears, to but the initial cost of equipment is high.
exclude Y. pseudotuberculosis, Brucella, Salmone­lla
and several fungi which may resemble the lesions of Extrapulmonary Tuberculosis
tuberculosis but will be smear negative. M. tubercu­ For diagnosis of extrapulmonary tubercu­losis,
losis is highly pathogenic for guinea-pigs and virtu­ extrapulmonary tuberculosis microscopy, culture
ally nonpathogenic for rabbits, while M. bovis is and occasionally animal inocula­tion are also used,
highly pathogenic for both guinea-pigs and rabbits. though it is difficult to get conclusive results as the
Guinea pig inoculation, is now seldom resorted to bacilli are present in far fewer numbers in these
because it is cumbersome, costly and less sensitive lesions than in pulmonary disease.
than culture, particularly with catalase negative, CSF from tuberculous meningitis often develops
INH resistant strains isolated from south India. a spider web clot on standing, examination of
The animal inoculated with strains of low virulence which may be more successful than of the fluid.
may have to be observed for 12 weeks or longer and The use of PCR and DNA probes may help detect
sometimes the only lesion demon­strable may be an the bacilli speedily and more often.
enlarged lymph node. Bone marrow and liver biopsy specimens from
Guinea-pig inoculation is very rarely used miliary tuberculosis and blood from those with
nowa­days. HIV coinfection are useful for culture. Pus from
6. Immunodiagnosis tubercu­lous abscesses often yields positive results
in smear and culture.
i. Serology: Various serological tests like enzy­
Pleural effusion and other exudates may be
me-linked immunosorbent assay (ELISA),
col­lected with citrate to prevent coagulation. If free
radioimmunoassay (RIA) and latex agglutination
from other bacteria, they may be used for culture
have been tried for the serodiagnosis of
after cen­trifugation. If other bacteria are present,
tuberculosis. ELISA is considered to be the
prior concen­tration is necessary.
most sensitive and specific. ELISA test has been
Urinary excretion of bacilli in renal tuberculosis is
attempted using several antigenic materials
intermittent. Hence it is advisable to test 3–6 morning
such as anti­gen 5, antigen 6, antigen 60, purified
samples of urine. Each sample is centrifuged for 3000
mycobacterial glycolipids, unheated sterile
rpm for 30 minutes and the sediment used for culture
culture filtrate of M. tuberculosis and purified
after concentration.
protein derivative derived from M. tuberculosis.
ii. Tuberculin testing: Demonstration of hyper­
sensitivity to tuberculo­protein (tuberculin Sensitivity Testing
testing) is a standard procedure. Its scope and Drug-resistant mutants continuously arise at a low
limitations are discussed below. rate in any mycobacterial population. The purpose
of sensitivity testing is to determine whether the
7. Hybridization and Nucleic Acid Technology great majority of the bacilli in the cul­ture are
i. Nucleic acid probes: are available for the sensitive to the antitubercular drugs cur­rently in
identification of the M. tuberculosis complex use. Several methods have been described:
and specifically, M. tubercu­losis. 1. Absolute concentration method: Resistance is
ii. Polymerase chain reaction (PCR) and ligase expressed as the lowest concentration of drug
chain reaction (LCR) are used as diagnostic that inhibits all or almost all of the growth,
techniques. that is, the minimum inhibitor concentration
iii. Transcription mediated amplification, (MIC). This method is inferior to resistance
targeting ribosomal RNA has been introduced ratio method because known sensitive strain
as an improvement on PCR based DNA is not tested for MIC.
amplification. 2. Resistance ratio method: The resistance-ratio
iv. DNA ‘fingerprinting: Methods for epidemio­ method in which test strains and susceptible
logical purposes. controls, i.e. a known sensitive strain (H37Rv)
of M. tuberculosis are inoculated on sets of LJ
8. Chromatography medium containing doubling dilutions of drug.
The cell walls of Mycobacterium organisms contain The results are expressed as the ratio of the drug
long chain fatty acids called mycolic acids, which concentration inhibiting the test strain to that
may be detected chromographically. Earlier inhibiting the control strains.

Chap-32.indd 234 15-03-2016 11:13:53


Chapter 32: Mycobacterium tuberculosis  | 235
Susceptible strains have ratios of 1 or 2, while simultane­ously in the naturally infected host.
higher ratios indicate resistance. Both these are mediated by T-cells sensitized to
3. Proportion method: For each drug tested, bacterial antigen. Although humoral antibodies
several dilutions of standardized inoculum are are produced in response to naturally occurring
inoculated onto control and drug-containing tuberculous infection, they appear to play
agar medium. The extent of growth in the no beneficial role in host defense. Acquired
absence or presence of drug is compared and antituberculous immunity is the prototype of
expressed as a percentage. If growth at the cell-mediated immunity invoked by facultative
critical concentration of a drug is >1%, the intracellular bacteria. In the nonimmune host, the
isolate is considered clinically resistant. bacilli are able to multiply inside phagocytes and
4. Radiometric method: Employing the principles lyse the host cells, while in immune host CD4+
of the proportion method, this rapid method uses helper T cells and CD8+ suppressor T cells are
liquid medium containing 14C-labeled growth produced. The former secrete interferon-gamma
substrate. Growth is indicated by the amount of which acti­vates macrophages to kill intracellular
14
C-labeled-carbon dioxide (CO2) released, as mycobacte­ria and the latter kill the macrophages
measured by the BACTEC 460 instrument. For that are infec­ted with mycobacteria.
each drug tested, a standardized inoculum is
inoculated into a drug-free and drug-containing Koch Phenomenon
vial. The rate and amount of CO2 to determine
Robert Koch (1890, 1891) originally described the
susceptible or resistance produced in the absence
response of a tuberculous animal to reinfec­tion.
or presence of drug is then compared.
In a normal guinea pig subcutaneous injection of
5. Nonradiometric method: Mycobacterial virulent tubercle bacillus produces no immediate
growth indicator tube (MGIT) can also be used response, but after 10–14 days a nodule develops
for sensitivity testing of M. tuberculosis. at the site, which breaks down to form an ulcer
6. New approaches that persists till the animal dies of progressive
i. Luciferase reporter mycobacteriophage: tuberculosis. The draining lymph nodes are
Luciferase reporter mycobacteriophage enlarged and caseous. If on the contrary, virulent
(containing the firefly luciferase gene) has tubercle bacillus is injected in a guinea pig which
been used for susceptibility testing of M. had received a prior injection of the tubercle
tuberculosis. The isolate of M. tuberculosis bacillus 4–6 weeks earlier, an indurated lesion
to be tested is grown in the presence ap­pears at the site in a day or two. This undergoes
and absence of drug and the reporter necro­sis in another day or so to form a shallow
mycobacteriophage is added. Then a ulcer, followed by rapid healing and no lymph
substrate of luciferase, luciferin is added node involvement or other tissues. This is known
following infection. If bacteria are viable, the as the Koch phenomenon and is a combination of
luciferin is broken down and light is emitted hypersensitivity and immuni­ty.
which can be measured. This method is
called chemiluminescence. If the isolate is Components of Koch Phenomenon
resistant to drug, light will be emitted, while Koch phenomenon has three components:
bacteria susceptible to the drug will not 1. A local reaction of induration and necrosis.
emit any light. The amount of light emitted 2. A focal response in which there occurs acute
is directly proportional to the number of congestion and even hemorrhage around
viable bacteria. tuerculous foci in tissues.
ii. Epsilometer test (E-test): It has also been 3. A systemic response of fever which may some­
applied for susceptibility testing of M. times be fatal.
tuberculosis.
iii. Detection of resistance gene: Demonstration Tuberculin Test
of mutation in specific genes for different Principle: The principle of this test is delayed
drugs is a useful indicator of drug resistance. (Type IV) hypersensitivity reaction.
Mutations to rifampicin resistance caused by
mutations in the rpoB gene are commercially Reagents
available.
i. ‘Original’ or ‘old tuberculin’ (OT): Koch pre­
pared a protein extract of tu­bercle bacillus by
Allergy and Immunity concentrating tenfold by evapora­tion, a 6–8
Two immunologic responses, antituberculous week culture filtrate of the bacillus grown in
im­munity and tuberculin hypersensitivity, develop 5% glycerol broth. This was called ‘original’ or

www.ebook3000.com
Chap-32.indd 235 15-03-2016 11:13:53
236  |  Section 3: Systemic Bacteriology
‘old tuberculin’ (OT). Initially Koch employed and other exanthema­tous reactions; 5. Occasionally
OT in the treatment of tuberculosis but it was after chemotherapy and removal of lung lesion; 6.
soon given up. Advanced age; 7. Immunosuppressive therapy
ii. Purified Protein Derivative (PPD): OT was and defective cell mediated immunity (CMI); 8.
first used for allergic (tuberculin) test­ing by von Lymphoreticular malignancy; 9. Sarcoidosis; 10.
Pirquet (1906). It was replaced by a partially Severe malnutrition; 11. False-negative results
purified protein antigen introduced by Seibert may also be due to inactive PPD preparations and
as OT was a crude product. This is known as the improper injection technique.
purified protein derivative (PPD).
Dose of PPD: Purified protein derivative (PPD) Uses of Tuberculin Test
is the skin test reagent that is primarily used to 1. To diagnose active infection in infants and
detect hypersensitivity in these persons. One large young children.
batch of PPD made by Seibert (PPD-S) in 1939 was 2. To measure prevalence of infection in an area .
recognized as the international standard PPD- 3. To select susceptibles for BCG vaccination.
tuberculin and arbitrarily designated to contain of 4. Indication of successful BCG vaccination.
OT or 0.00002 mg of PPD-S. In recent years in vitro interferon-γ release
Another PPD is RT-23 with tween 80. In India, assay (IGRA) have been introduced sensitive and
PPDRT-23 of strength 1TU and 2TU are available more specific to tuberculin test. This test is used in
1TU of PPD RT-23 is equivalent to 5TU of PPD-S. blood specimens which contains lymphocytes. It
uses ELISA for measuring interferon-γ production
Method by sensitized T lymphocytes.
1. Mantoux test: In the Mantoux test, 0.1 mL of
PPD containing 5 TU is injected intradermally Prophylaxis
on the flex­o r aspect of the forearm with a In the prevention of tuberculosis general measures
tuberculin syringe. such as adequate nutrition, good housing and
A PPD dose of 1 TU is used when extreme health education are as important as specific anti­
hypersensitivity is suspected; and doses of 10 bacterial measures. The latter consists of early
or 100 when 5 TU test is negative. detection and treatment of cases, BCG vaccination
Interpretation: Tuberculin tests should be and by chemoprophylaxis.
read 48–72 hours after injection. Induration of
diameter 10 mm or more is considered positive, BCG Vaccination
5 mm or less negative and 6–9 mm is of doubtful
Immunoprophylaxis is by intradermal injection of
significance because it may be due to other
the live attenuated vaccine developed by Calmette
myco­bacterial infections.
and Guerin (1921), the Bacille Calmette Guerin
2. Heaf test: Multiple puncture testing as Heaf test
or BCG. This is a strain of M. bovis attenuated by
is done with a spring-loaded gun which fires
239 serial subcultures in a glycerine-bile-potato
six prongs into the skin through a drop of PPD
medium over a period of 13 years between the
(Heaf method). Single-test disposable devices
years 1908 and 1920 which was avirulent for man
with PPD dried onto prongs (tine tests) are also
while retaining its capacity to induce an immune
available for individual testing.
response. The foller widely used BCG strains
include pasteur 1173 P2, Tokyo-172, copenhagen
Result 1331 and Glaxo-1077.
I. Positive Test
Aim: The aim of BCG vaccination is to induce
A positive tuberculin test indicates hypersensitiv­ity a benign, artificial primary infection which will
to tuberculoprotein denoting infection with tuber­ stimulate an acquired resistance to possible
cle bacillus or BCG immunization, recent or past, subsequent infection with virulent tubercle bacilli.
with or without clinical disease. The test becomes
positive 4–6 weeks after infection or immunization. Dose and administration: BCG vaccine is available
in liquid form and freeze-dried (lyophilized) form.
Freeze-dried (lyophilized) form is more stable
II. False-positive reactions
preparation and com­monly used. The lyophilized
False–positive reactions may be seen in infections vaccine is reconstituted by sterile physiological
with relat­ed mycobacteria (‘atypical’ mycobacteria). saline to make a final con­centration of 0.1 mg
(moist weight) in 0.1 mL of the vaccine and it is
III. False-negative Tests (Tuberculin Anergy) supplied by BCG vaccine laboratory,Chennai.
1. Early tuberculosis; 2. Advanced tuberculosis; Vaccine should be uti­lized within 3–6 hours once
3. Miliary tuberculosis; 4. In patients with measles reconstituted. The organisms grow to a limited

Chap-32.indd 236 15-03-2016 11:13:53


Chapter 32: Mycobacterium tuberculosis  | 237
extent in the tissues following injection of 0.1 mL Chemoprophylaxis
of vaccine intradermally. BCG vaccine should be
Chemoprophylaxis or preventive chemotherapy
administered soon after birth failing which it may
is the administration of antituberculous drugs
be given at any time during the first year of life.
(usually only isoniazid) to persons with latent
Phenomena after vaccination: Two to three weeks tuberculosis (asymptomatic tuberculin positive)
after injection a small nodule develops at the and a high risk of developing active tuberculosis, or
site of inoculation. It increases slowly in size and to the uninfected exposed to high risk of infection.
reaches a diameter of 4–8 mm by about 5 weeks. It is particularly in­dicated in infants of mothers
It then subsides or breaks into a shallow ulcer with active tuberculosis and in children living with
which heals by scarring. Normally the individual a case of active tuberculo­sis in the house. Isoniazid
becomes Mantoux-positive after a period of 8 5 mg/kg daily for 6–12 months is the usual course.
weeks has elapsed, but sometimes about 14 weeks Trials have shown that this reduces the risk of
are needed. developing active disease by 90%. HIV infected
contacts of active tuberculosis also benefit from
Protective Efficacy this prophylaxis.
The duration of protection is from 15 to 20 years.
Several field trials have been conducted to assess the Treatment
efficacy of the BCG vaccine. Studies have shown that The bactericidal drugs, along with the bacteri­
the range of protection offered by BCG varied from static drug ethambutol (E) constitute the first-line
0 to 80% in different parts of the world (Table 32.5). drugs in antituberculous therapy. The old practice
The consensus opinion is that BCG may not of daily administration of drugs for two years or
pro­tect from the risk of tuberculosis infection, but so has been replaced by short course regimens of
gives protection to infants and young children 6–7 months, which are effective and convenient. A
against the more serious types of the disease, such typical example of such a schedule for a new smear
as meningitis and disseminated tuberculosis. positive case is a combination of four drugs (HRZE)
BCG induces a nonspecific stimulation of the given three times a week during an initial intensive
im­mune system providing some protection against phase of two months, followed by 4–5 months of
lep­rosy and leukemia. Multiple injection of BCG con­tinuing phase with only two drugs (HR) three
has been tried as adjunctive therapy in some times a week.
malignan­cies.
Multidrug Resistant Mycobacterium
Complica­tions of BCG Vaccine
Tuberculosis (MDR-TB)
A. Local: Abscess, indolent ulcer, keloid.
A very serious consequence of unchecked drug
B. Regional: Enlargement and suppuration of
resistance has been the emergence and spread
draining lymph nodes.
of ‘multidrug resistant tuberculosis’(MDR-TB).
C. General: Fever, mediastinal adenitis, erythema
WHO defines a multi-drug resistance (MDR) strain
nodo­sum.
as one that is at least resistant to rifampicin(R) and
Contraindications: BCG should not be given isoniazid (H). This is because R and H form the
to patients suffering from generalized eczema, sheet anchor of short-term chemotherapy and any
infective dermatosis, hypogammaglobulinemia strain resistant to both these drugs is unlikely to
and to those with a history of deficient immunity. respond to treatment.

Table 32.5  Protective efficacy of BCG vaccinations in nine major trials


Immunization period Country or population Age range of vaccinees Efficacy (%)
1935–38 North American Indian 0–20 80
1937–48 Chicago, USA Neonates 75
1947 Georgia, USA 6–17 0
1949–51 Puerto Rico 1–18 31
1950 Georgia and Alabama, USA >5 14
1950–52 UK 14–15 78
Madanapalle, South India All ages 60
(Rural)
1968–71 Chingleput, All ages 0
South India (Rural)

www.ebook3000.com
Chap-32.indd 237 15-03-2016 11:13:53
238  |  Section 3: Systemic Bacteriology
Risk factors for drug resistance may include ™™ Liquid media are Dubos’, Middlebrook’s, Proskauer
previous treatment for TB, residence in an area and Beck’s, Sula’s and Sauton’s media are the more
endemic for drug resistance, or close contact with com­mon
an individual who is infected with MDR-TB. Drug ™™ They are weakly catalase positive, neutral-red positive,
resist­ance is usually acquired by spontaneous amidase positive, nitrate reduction test positive, niacin
mutations as a result of the inappropriate use of test negative, and arylsulfatase negative
antimicrobial agents to treat M. tuberculosis and ™™ Pathogenesis and immunity: The source of infection
is usually an open case of pulmonary tuberculosis.
the lack of patient com­pliance. Another serious
The initial infection with M. tuberculosis is referred
condition extensively drug resistant-tuberculosis to as a primary infection. Subsequent disease in a
(XDR-TB) has emerged recently. XDR-TB is due to previously sensitized person, known as postprimary
M. tuberculosis strains which are resistant to any (secondary or reinfection) tuberculosis
fluoroquinolone and at least one of three injectable ™™ Diseases: M. tuberculosis causes primarily pulmonary
second line drugs (capreomycin, kanamycin and tuberculosis
amikacin) in addition to isoniazid and rifampicin. ™™ Diagnosis: Bacteriological diagnosis of tuberculosis
Between 50 and 100 million people worldwide can be established by direct microscopy, culture
examination or by animal inoculation test
are thought to be infected with strains of drug
1. Sputum is the specimen of choice for pulmonary
resistant tuberculosis. MDR-TB requires an
tuberculosis
extended treatment period compared with drug-
2. Microscopy: Z–N staining and auramine-
susceptible isolates. For cases of resistance to rhodamine staining for demonstration of AFB in
isoniazid or rifampin, second-line. anti­tuberculosis stained smears is most
drugs may include aminoglycosides (kanamycin, 3. Culture is the definite method to detect and
amikacin, capreomycin), and fluoro­quinolones. identify M. tuberculosis and is sensitive and specific
With the numbers of cases of multidrug-resistant 4. Serology is of limited value in the diagnosis of
M. tuberculosis increasing, newer agents are pulmonary tuberculosis
being tested in vitro to determine their efficacy. If 5. D irect detection by molecular probes is
compliance is an issue, a single daily dose of all first relatively insensitive
line antitubercular drugs is preferred, and ‘directly 6. The recent rapid and automated methods include
observed treatment strategy’ (DOTS) has been automated radiometric culture methods (e.g.
BACTEC) , SEPTICHEK, MGITs, etc.
recommended by WHO. Otherwise resistance may
™™ Resistance ratio method, absolute concentration
be assumed and tested for in vitro. method and proportion method are used to
determine the sensitivity testing of M. tuberculosis.
Key Points Other methods for sensitivity testing include BACTEC
radiometric method, MGIT, chemiluminescence and
™™ Mycobacteria are aerobic, nonmotile, noncap­sulated
Epsilometer test (E-test)
and nonsporing. Growth is generally slow
™™ Tuberculin test: This test is delayed (Type IV) hyper­
™™ Mycobacteria are, known as acid-fast bacilli (AFB).
sensitivity reaction. Many methods such as 1. Mantoux
Acid fastness has been ascribed variously to the
test and 2. Heaf test had been described for tuberculin
presence in the bacillus of mycolic acid or to a sem­
testing
ipermeable membrane around the cell
™™ Treatment, Prevention, and Control
Mycobacterium tuberculosis
Chemotherapy forms the mainstay of treatment of
™™ Mycobacterium tubercu­losis is weakly gram-positive,
tuberculosis . Drug resistance in M. tuberculosis is due
strongly acid-fast, aerobic bacilli
to mutation. The emergence of multidrug resistance-
™™ Ziehl-Neelsen acid-fast stain is useful in staining tuberculosis (MDR-TB) is a very serious problem. DOT
organisms. With this stain, the tubercle bacilli stain is a method now being widely followed for treatment
bright red. Tubercle bacilli may also be stained with of cases of tuberculosis. Another serious condition
the fluorescent dyes (auramine 0, rhodamine) and extensively drug resistant-tuberculosis (XDR- TB) has
appear yellow luminous bacilli under the fluorescent emerged recently
microscope
™™ Preventive measures against tuberculosis include
™™ They are aerobes, slow growers, produce luxuriant chemoproph­ylaxis, vaccination, and general health
eugonic growth after 2–8 weeks measures. Immunoprophylaxis with BCG in endemic
™™ The solid medium most widely employed for routine countries.
culture is Lowen­stein–Jensen (L–J) medium without
starch
™™ On L–J media, M. tuberculosis forms dry, rough, raised, IMPORTANT QUESTIONS
irregular colonies with a wrinkled surface. They are
creamy white, becoming yellowish or buff colored 1. Describe the morphology, cultural characteristics
on further incubation. They are tenacious and not and pathogenicity of M. tuberculosis.
easily emulsified. Mycobacterium tuberculosis has 2. Classify mycobacteria. Describe the laboratory
a luxuriant growth (eugenic growth) as compared diagnosis of pulmonary tuberculosis.
to Mycobacterium bovis, which grows poor­ly on LJ
3. Differentiate between Mycobacterium tubercu­losis
glycerol medium (dysgonic growth)
and Mycobacterium bovis in a tabulated form.

Chap-32.indd 238 15-03-2016 11:13:53


Chapter 32: Mycobacterium tuberculosis  | 239
4. Write short notes on: 5. A positive Mantoux test is indicated by an area of
a. Antigenic structure of Mycobacterium tuberculosis induration of:
b. Pulmonary tuberculosis a. 10 mm or more in diameter
c. Koch’s phenomenon b. 5–9 mm in diameter
d. Tuberculin test c. 3–4 mm in diameter
e. Mantoux test d. None of the above
6. Mantoux test shows false negative reaction in all
f. BCG vaccine
the following patients except:
g. Sensitivity testing of Mycobacterium tuberculosis
a. Military tuberculosis b. Malignancies
h. Multidrug-resistant Mycobacterium tuberculosis c. Leprosy d. Malnutrition
(MDR-TB).
7. Culture of tubercle bacilli maybe positive if
number of bacteria in the specimen is:
MULTIPLE CHOICE QUESTIONS (MCQs) a. As few as 1–4 per mL
1. Eugonic growth on Lowenstein-Jensen medium is b. As few as 5–9 per mL
produced by: c. As few as 10–100 per mL
a. Mycobacterium tuberculosis d. as few as 150–200 per mL
b. M. bovis 8. Mycobacterium. tuberculosis is pathogenic for:
c. Both of the above a Rabbits
b. Guinea pigs
d. None of the above
c. Both of the above
2. LJ medium consists of the following ingredients
d. None of the above
except:
9. Multidrug resistance tuberculosis (MDR-TB) is
a. Egg yolk b. Agar
due to M. tuberculosis strain as one that is:
c. Mineral salts d. Malachite green
a. Resistant to rifampicin only
3. Nitrate reduction test is positive in:
b. Resistant to isoniazid only
a. Mycobacterium tuberculosis
c. Resistant to at least rifampicin and isoniazid
b. M. jortuitum
d. None of the above
c. M. chelonei
10. Extensively drug resistant tuberculosis (XDR-TB) is
d. All of the above due to M. tuberculosis strains which are resistant to:
4. All the following statements are true regarding a. Any fluoroquinolone
Mycobacterium tuberculosis except: b. Isoniazid and rifampicin
a. They are straight or slightly curved acid-fast ba- c. At least one of three injectable second line
cilli drugs (capreomycin, kanamycin and amikacin)
b. They produce luxuriant eugonic growth after d. All of the above
4–6 weeks
c. They are weakly Gram-positive ANSWERS (MCQs)
d. They are niacin test positive 1. a; 2. b; 3. d; 4. d; 5. a; 6. c; 7. c; 8. b; 9. c; 10. d

www.ebook3000.com
Chap-32.indd 239 15-03-2016 11:13:53
33
Chapter

Mycobacterium leprae

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe the following:
be able to: – Lepromatous leprosy; tuberculoid leprosy;
∙∙ Describe morphology of M. leprae lepromin test
∙∙ Discuss cultivation of lepra bacilli – Differentiate between lepromatous and tuber­
∙∙ Explain animal models in leprosy culoid leprosy
∙∙ Describe pathogenesis of leprosy –  Discuss laboratory diagnosis of leprosy.

Introduction bacilli are arranged in clumps resembling cigarette


ends. The globi are present in Virchow’s lepra cells
Leprosy is probably the oldest disease known to or foamy cells which are large undifferentiated
mankind. Leprosy was described as ‘kushta’ in histiocytes.
Sushruta Samhita written in India in 600 B.C. It
is caused by Mycobacterium leprae which was Generation time of the lepra bacillus: The
discovered by Hansen in 1874 in Norway. generation time of the lepra bacillus has been found
to be exceptionally long in animal experiments,
12–13 days on the average but may vary from 8 to 42
Mycobacterium leprae
days, in comparison with about 14 hours in the case
Morphology of the tubercle bacillus and about 20 minutes in the
M. leprae is a straight or slightly curved rod, 1–8 case of coliform bacilli.
mm x 0.2–0.5 mm in size, showing consider­able
morphological variations. The rods may stain Cultivation
uniformly or show granules and beads that are In spite of the efforts of many workers it has not so
slightly larger than the average diameter of the far been possible to cultivate lepra bacilli either in
cell. Polar bodies and other intracellular elements bacteriological media or in tissue culture. There
may be present. Clubbed forms, lateral buds or have been several reports of successful cultivation
branching may be observed. They are nonmotile but none has been confirmed. One of the best
and non­sporing. known of such reports (1962) came from the In­dian
They are gram-positive and stain more readily Cancer Research Center (ICRC) Mumbai, where
than M. tuberculosis. With Ziehl–Neelsen stain, an acid-fast bacillus was isolated from leprosy
they are less acid-fast than tubercle bacilli, so 5% patients, em­ploying human fetal spinal ganglion
sulfuric acid is employed for decoloriza­tion after cell culture. This is known as ICRC bacillus. This
staining with carbol fuchsin. The bacilli are seen ICRC bacillus has been adapted for growth on
singly and in groups, intracellularly or lying free Lowenstein-Jensen medium. Its relation to the
outside the cells. Inside the cells they are present lepra bacillus is uncertain.
as bundles of organisms bound together by a lipid- There have been many attempts to transmit
like substance, the glia. These masses are known as lepro­sy to experimental animals. However, the
globi. Large numbers of bacilli may be packed in real break through was in 1960, when Shepard
the cells in an arrangement that suggests packets of discovered that M. leprae could multiply in the
cigars. The parallel rows of bacilli in the globi give footpads of mice kept at a low temperature (20°C).
appearance of a cigar bundle. In tissue sections, the This observation has been confirmed and has
Chapter 33: Mycobacterium leprae  | 241
become the standard procedure for experimental The cell wall of M. leprae is made up of four
work with the bacillus. Following animals have been layers like other Mycobacteria. The innermost is a
used for experimental infection with M. leprae. peptidoglycan layer which gives the cell its shape and
1. Mouse (footpad) rigidity. External to this is lipoarabinomannan-B
2. Nine-banded armadillos (LAM-B) layer, attached to which is a dense
3. Chimpanzees, palisade of characteristic long chain fatty acids
4. Monkeys known as mycolic acid. The outermost layer is
5. Slender loris composed of mycosides. A major component of this
6. Indian pangolin layer is phenolic glycolipid-l (PGL-l).
7. Chipmunks LAM-B is a dominant antigen of M. leprae,
8. Golden hamsters is highly immunogenic and is used in the
9. European hedgehog. serodiagnosis of leprosy. Patient, infected with M.
leprae, develops antibodies against polysaccharide
Mouse: In the mouse, infections can be initiated with
constituent of PGL-I which has been used for the
as few as 1–10 bacilli. A granuloma develops at the
serodiagnosis of leprosy.
site in 1–6 months following intradermal inoculation
In addition, M. leprae possesses a large number
into the footpads of mice. If cell-mediated immunity
of protein antigens namely 18 kDa, 28 kDa, 35 kDa,
is suppressed by thymectomy or the administration
36 kDa, 65 kDa and so on, according to molecular
of antilymphocyte serum, a generalized infection is
weight of antigens in kilodaltons (kDa).
produced, simulating lepromatous leprosy.
Nine-banded Armadillo: The nine-banded Resistance
armadillo (Dasypus novemcinctus) is highly
In a warm humid environment, lepra bacilli have
susceptible to infection with lepra bacilli. This is
been found to remain viable for 9–16 days and in
presum­ably due to low body temperature. Following
moist soil for 46 days. They survive exposure to
inoculation into armadilles, a generalized infection
direct sunlight for two hours and ultraviolet light
occurs with ex­tensive multiplication of the bacilli and
for 30 minutes.
production of lesions typical of lepromatous leprosy.

Antigenic Structure Classification


Mycobacterial antigens may be classified on the Classification System of Ridley and Jopling
basis of : The spectrum of disease activity in leprosy is very
1. Solubility as (a) soluble (cytoplasmic) and (b) broad, characterized by pronounced variations in
insoluble (cell wall lipid-bound). clinical, histopathologic, and immunologic findings.
2. Chemi­cal structure as (a) carbohydrate and (b) On the basis of these properties, Ridley and Jopling
protein (1966) have established a clas­sification scheme
3. Distribution within the species. Up to 90 consisting of five forms of leprosy (Table 33.1):
soluble antigens have been demon­strated by ∙∙ Tuber­culoid (TT)
counterimmunoelectrophoresis in Mycobac­ ∙∙ Borderline tuberculoid (BT)
teria. They can be divided into 4 groups: ∙∙ Borderline (BB)
Group I: Common to all Mycobacteria. ∙∙ Borderline lepromatous (BL)
Group II: Occur in slowly growing species. ∙∙ Lepromatous (LL).
Group III: Occur in rapidly growing species. Hyperreactive tuberculoid (TT) leprosy is at
Group IV: Unique to each individual species. one pole and anergic lepromatous (LL) leprosy
M. leprae possesses antigens of groups I and IV. at the other. The type of disease is a reflection of

Table 33.1  Characteristics of lepromatous and tuberculoid leprosy


Feature Lepromatous leprosy Tuberculoid (TT) leprosy
1. Resistance Seen in persons whose resistance is low Seen in persons whose resistance is high
2. Infectivity High Low
3.  Bacilli in skin +++ –
4.  Bacilli in nasal secretions +++ –
5.  Granuloma formation +++ –
6.  Lepromin test – +++
7.  Antibodies to M. leprae Hypergammaglobulinemia Normal
8. Prognosis Poor Good

www.ebook3000.com
242  |  Section 3: Systemic Bacteriology
the immune status of the host. It is therefore not 2. Tuberculoid (TT) Leprosy
permanent and varies with chemotherapy and The skin lesions are few and sharply de­marcated,
alterations in host resistance. TT and LL are stable. consisting of macular anesthetic patches. In
The others are unstable, especially BT, which in tuberculoid leprosy, skin biopsies show mature
the absence of treatment can regress to BB or BL. granuloma formation in the dermis consisting of
epithelioid cells, giant cells, and rather extensive
Pathogenesis infiltration of lymphocytes.
Leprosy (Hansen’s disease) is a chronic granulo­ There are very few acid-fast bacilli (AFB) so
matous disease of humans primarily involving that they are generally not seen microscopically
the skin, peripheral nerves and nasal mucosa but (paucibacillary disease) and infectivity is minimal.
capable of affecting any tissue or organ. The organisms invade the nerves and selectively
M. leprae is an obligate intracellular parasite colonize the Schwann cells. The local nerves are
that multiplies very slowly within the mononuclear involved in the early stage and gradually the infec­
phagocytes, especially the histiocytes of the skin tion extends into the bigger nerve trunks which are
and Schwann cells of the nerves. M. leprae has an thickened, hard and tender. This leads to deformi­
especially strong predilection for nerves and is the ties of hand and feet.
only known human pathogen that preferentially
attacks the peripheral nerves. The two extreme or 3. Borderline (BT) Leprosy
polar forms of the disease are the lepromatous and
Borderline leprosy (BB), sometimes called
tuberculoid types TT) types (Table 33.2).
dimorphous or intermediate leprosy, has features
of both tuberculoid and lepromatous forms. This is
1. Lepromatous Leprosy an unstable form of the disease and may shift to the
Patient develops numerous nodular skin lesions lepromatous or tuberculoid part of the spectrum
(lepromata) on face, ear lobes, hands, feet and depending on chemotherapy or alterations in host
less commonly trunk. Skin lesions contain many resistance.
macrophages, often seen as large foamy cells
packed with AFB. Skin biopsy specimens may 4. Indeterminate Type
contain up to 109 bacilli per gram of tissue. This is
There is an early unstable tissue reaction with mild
known as ‘multi­bacillary disease’. Nodular skin
transient tissue lesion, often resembling maculo-
lesions ulcerate due to repeated trau­ma as a result
anesthetic patches which is not characteristic of
of loss of sensation. The ulcerated nodules become
either the lepro­matous or the tuberculoid type. In
secondarily infected that leads to distortion and
many persons, the indeterminate lesions undergo
mutilation of extremities.
healing spontaneously. In others, the lesions may
Bacilli invade the mucosa of the nose, mouth
progress to the tuberculoid or lepromatous types.
and up­per respiratory tract and are shed in large
numbers in nasal and oral secretions. Bacillemia
is common. Eyes, testes, kidneys and bones are Epidemiology
also involved. Leprosy is an exclusively human dis­ease and the
Lepromatous leprosy is more infectious than only source of infection is the patient. Disease is
other types and has a poor prognosis. Cell-mediated spread by person-to-person contact. The mode
immunity is deficient and the lepromin test is of entry may be either through the respiratory
negative in lepromatous leprosy. Humoral antibodies tract or through the skin. Asymptomatic infection
agai­nst mycobacterial antigens are produced in high appears to be quite common in endemic areas.
concentrations which play no protective role. Most Bacilli may also be transmitted via breast milk
cases show biological false positive reaction in from lepromatous mothers, by insect vectors, or
standard serologi­cal tests for syphilis. by tattooing needles.

Table 33.2  Characteristics of the five forms of leprosy


TT BT BB BL LL
Bacilli in skin − +/ − + ++ +++
Bacilli in nasal secretions − − − + +++
Granuloma formation +++ ++ + − −
Lepromin test +++ + +/ − − −
Antibodies to M. leprae +/ − +/ − + ++ +++
TT, tuberculoid; BT, borderline tuberculoid; BB, borderline; BL, borderline lepromatous; LL, lepromatous
Chapter 33: Mycobacterium leprae  | 243
Leprosy has a long incubation period, an institution of chemotherapy. Clinically crops
average of 3–5 years or more for lepromatous cases. of red nodules appear in the skin, lasting for 1
It is rare in children aged less than 5 years. It has or 2 days. The histological picture is that of an
been estimated to vary from a few months to as Arthus reaction or immune complex disease.
long as 30 years. It is generally held that intimate The immunological basis of type 2 is vasculitis
and prolonged contact is necessary for infection associ­ated with the deposition of antigen-
to take place. The disease is more likely if contact antibody complexes.
occurs during childhood.
Once worldwide in distribution, leprosy is now Lepromin Test
confined mainly, but not exclusively, to the under­ Till recently, the only method for studying
developed areas of the tropics and the southern immunity in leprosy was a skin test for delayed
hemi­sphere. Leprosy is widely prevalent in India. hypersensitivity, the lepromin test first described
Although the disease is present throughout the by Mitsuda in 1919.
country, the distribution is uneven.
Lepromins: The lepromins used as antigen in
lepromin test may be of human origin (lepromin-H)
Immunity or of armadillo origin (lepromin-A) and are of two
Leprosy is a disease of low infectivity. A high degree types:
of innate immunity against lepra bacilli seems to 1. Integral lepromin (Mitsuda lepromin): The
exist in human beings so that only a minority of original antigen (lepromin) was developed by
those infected develop clinical disease. Mitsuda in 1919. The original crude Mitsuda
Infection with lepra bacilli induces both antigens extracted from skin lesions of
humoral and cellular immune responses. Humoral lepromatous patients (integral lepromins) were
antibodies do not have deleterious effect on the standardized on the basis of tissue content.
bacilli, while cellular immune mechanisms are Standard lepromins are being prepared
a­ble of destroying them. Also, in leprosy there is increas­ingly from armadillo-derived lepra
a close correlation between the various clinical bacilli (lepromin A ­ ).
forms and the cell-mediated immunity (CMI) 2. Bacillary lepromin: This contains more of
response of the host. When it is adequate, the bacillary components and less of tissue. An
lesions are of the tuberculoid type. Patients with impor­tant example of bacillary lepromin is
tuber­culoid leprosy exhibit a strong delayed-type Dharmendra antigen which is prepared by
hypersensitivity to lepromin. floating out the bacilli from finely ground
The lepromatous type of disease develops lepromatous tissue with chloro­form, evapo­
when cell mediated immunity is deficient. Delayed rating it dry and removing the lipids by washing
hyper­sensitivity to the lepra bacillus protein is with ether. The antigen is made up in phe­nol
absent. A number of abnormal serologic activities saline for use.
are also associated with lepromatous leprosy,
including a biologic false positive reaction in Procedure
routine serologic tests for syphilis. The test is performed by injecting intradermally 0.1
ml of lepromin into the inner aspect of the forearm of
Reactions the individual. As a routine, the reaction is read at 48
Though leprosy is a chronic disease, its course is hours and 21 days. The response to the intradermal
sometimes interspersed with acute exacerbations injection of lepromin is typically biphasic:
due to immune reactions. These are of two types: 1. Early or Fernandez reaction: The early
1. Type 1 (Reversal reaction or the ‘lepra re­action’) reaction is also known as Fernandez reaction.
2. Type 2 (Erythema nodosum leprosum, (ENL)) It consists of erythema and induration at the site
1. Type 1 (Reversal reaction or the ‘lepra of inoculation developing in 24–48 hours and
re­action’): This occurs in borderline cases, usually remaining for 3–5 days. This reaction
occurring spontaneously or more often during indicates that the patient has been infected at
chemotherapy. It is a cell-mediated immune some time in the past and is a measure of pre-
reaction, with an influx of lymphocytes into existing delayed hypersensitivity.
lesions, and a shift to tuberculoid morphology. 2. Late or Mitsuda reaction: This is the classical
It may rapidly cause severe and permanent Mitsuda reaction. The reaction develops late,
nerve damage. becomes apparent in 7–10 days following the
2. Type 2 (Erythema nodosum leprosum, injection and reaching its maximum in 3 or 4
(ENL)): This is an immune-complex reaction weeks. At the end of 21 days, if there is a nodule
seen only in lepromatous and borderline more than 5 mm in diameter at the site of
lepromatous cases, usually a few months after inoculation, the reaction is said to be positive.

www.ebook3000.com
244  |  Section 3: Systemic Bacteriology
The nodule may even ulcerate and heal with dries, it is fixed by passing the slide twice or thrice
scarring if the antigen is crude. It takes several over a flame with the surface carrying the smear
weeks to heal. Histologically, there is infiltration uppermost. About 5–6 different areas of the skin
with lymphocytes, epithelioid cells and giant should be sampled, including the skin over the but­
cells. tocks, forehead, chin, cheek and ears.

Uses of Lepromin Reaction ii. Nasal Scrapings


The test is employed for the following purposes: A blunt, narrow scalpel is introduced into the
1. To classify the lesions of leprosy patients: The nose and the internal septum scraped sufficiently
reaction is positive in tuberculoid and negative to remove a piece of mu­cus membrane, which is
in lepromatous leprosy patients. transferred to a slide and teased out into a uniform
2. To assess the prognosis and response to smear.
treat­ment: A positive reaction indicates good The skin or nasal smear is immediately fixed by
prognosis and a negative one bad prognosis. lightly passing the underslide of the slide over the
3. To assess the resistance of individuals to spirit lamp flame and transported to the laboratory
leprosy: It is desirable to recruit only lepromin for staining with Ziehl­–Neelsen method.
positive persons for work in leprosaria as
lepromin negative persons are more prone to 2. Microscopy
develop the disease. Smears are stained by Ziehl–Neelsen method using
4. To verify the identity of candidate lepra 5% in­stead of 20% sulfuric acid for decolourization.
bacilli: Cul­tivable acid-fast bacilli, claimed to Acid­fast bacilli (AFB) arranged in parallel bundles
be lepra bacilli, should give matching results within macrophages (Lepra-cell) confirm the
when tested in parallel with standard lepromin. diagnosis of lepromatous leprosy. The viable
bacilli stain uniformly and the dead bacilli are
Laboratory Diagnosis fragmented, irregular or granular.
M. leprae cannot grow in cell-free cultures. Thus, The smears are graded, based on the number of
laboratory confirmation of leprosy requires bacilli as follows:
histopatho­logic findings consistent with the 1–10 bacilli in 100 fields = 1+
clinical disease and either skin test reactivity to 1–10 bacilli in 10 fields = 2+
lepromin or the presence acid-fast bacilli in the 1–10 bacilli per field = 3+
lesions. The diagnosis consists of demonstra­tion 10–100 bacilli per field = 4+
of acid-fast bacilli in the lesions. 100–1,000 bacilli per field = 5+
More than 1,000 bacilli,
1. Specimens clumps and globi in every field = 6+
For routine examination, specimens are collected Differentiation of live and dead bacilli: Live bacilli
from the nasal mucosa, skin lesions and ear lobules. in the smear appear solid and uni­formly stained,
Biop­sy of the nodular lesions and thickened nerves, while dead or dying forms appear fragmented,
and lymph node puncture may be necessary in beaded and granular.
some cases. i. Bacteriological index (BI): The bacteriological
index (the number of bacilli in tissues) is
i. Skin Smears calculated by totalling the grades (number of
Slit and scrape method: Material from the skin pluses, +s scored in all the smears and divided
is obtained from an active lesion, and also from by number of smears. Thus if 7 (seven) smears
both the ear lobes by the “slit and scrape” method. examined have a total of fourteen pluses (14+),
Samples from the skin should be obtained from BI will be 2. For calculating BI, a minimum of
the edges of the lesion rather than from the center. four skin lesions, a nasal swab and both the ear
The skin is pinched up tight to minimize bleeding. lobes are to be examined.
A cut about 5 mm long is made with a scalpel, ii. Morphological index (MI): It is defined as
deep enough to get into the infiltrated layers. After the percentage of uniformly stained bacilli
wiping off blood or lymph that may have exuded, out of the total number of bacilli counted. This
the blade of the scalpel is then turned at right angle provides a method of assessing the progress
to the cut (slit) and the bottom and the sides of the of patients on chemotherapy.
slit are scraped with the point of the blade, sev­eral
times in the same direction so that tissue fluid and 3. Animal Inoculation
pulp (not blood) collects on one side of the blade Injection of ground tissue from lepromatous
which is smeared uniformly on a slide. When smear nodules or nasal scrapings from leprosy patient into
Chapter 33: Mycobacterium leprae  | 245
the foot pad of mouse produces typical granuloma 2. Intravenous injection of transfer factor, an extract
at the site of inoculation within 6 months. Nine- of lymphocytes, from patients suffering from
banded armadillo is another animal used for tuberculoid leprosy. Sensitized lympho­cytes
inoculation of material. The lesions which develop secrete certain substances called lym­phokines
in these animals can be identified by histological which stimulate the macrophages to ingest and
examination and Ziehl–Neelsen staining. kill the bacilli.
3. Intradermal injection of vaccine.
4. Lepromin Test
It is not a diagnostic test but is used to assess the Prophylaxis
resistance of patient to M. leprae infection. The test Case finding and adequate ther­apy have been the
can be used to assess the prognosis and response methods employed for prophylaxis. Long-term
to treatment. chemoprophylaxis has given encouraging results
in child contacts of infectious cases in India and
5. Serological Test the Philippines.
Detection of antibody against M. leprae phenolic
glycolipid antigen has been claimed to be a Immunoprophylaxis
specific diagnostic test. Various serological tests
At present no effective vaccine against leprosy
like latex agglutination, Mycobacterium leprae
exists. A number of candidate vaccines have been
particle agglutination (MLPA) and ELISA have been
tried and are still under trial. However, none of
described. Anti-PGL-1 antibody titers are higher in
these have reached the stage for universal use.
lepromatous patients but the tuberculoid patients
show low titers. The antibody titers decrease BCG vaccine: There is now considerable evidence
following effective chemotherapy. that BCG vaccine can provide some protection
against clinical leprosy. The results of controlled
6. Molecular Diagnostic Methods trials with BCG vaccine have demonstrated
significant but varying levels of protection in
The PCR is increasingly used to detect M. leprae in
four different countries—Uganda, Myanmar,
clinical specimens.
Papua New Guinea and in India, the Chingleput
(Chennai), ranging from 23–80%. Field trials in
Treatment Venezuela with killed M. leprae and BCG provided
Dapsone (4, 4’-diaminodiphenyl sulfone; DDS) no better protection than BCG alone.
was the first effective chemo­therapeutic agent
Candidate vaccines: All the reported “candidate”
against leprosy. Due to emergence of dapsone
vaccines have shown a similar degree of lepromin
resistance, WHO recommended multiple drug
conversions in lepromatous patients (50­–70%)
therapy (MDT) for all leprosy cases based on
and lepromin negative healthy individuals (90%).
dapsone, rifampicin and clofazimine. Patients
Field trials with different leprosy vaccines (BCG +
with paucibacillary (I, TT, BT) leprosy are given
killed lepra bacilli; ICRC bacillus) have not given
rifampicin 600 mg once a month (supervised)
conclusive results so far.
and dapsone 100 mg daily (unsupervised) for six
months. For multibacillary (BB, BL, LL leprosy,
rifampicin 600 mg once a month (supervised, Mycobacterium lepraemurium
dapsone 100 mg daily (unsupervised), clofazimine M. lepraemurium, a causative agent of rat leprosy,
300 mg once monthly supervised and 50 mg daily, was first described by Stefan sky in 1901 at Ode­
unsupervised are given for two years or until skin ssa. The disease is probably transmitted naturally
smears are negative. from rat to rat by fleas. Rat leprosy characterized
by subcutaneous indurations, swelling of lymph
Immunotherapy nodes, emaciation, and sometimes ulceration arid
Since lepromatous leprosy patients do not possess loss of hair.
CMI, efforts are being made to induce effective CMI DNA studies have revealed that M. leprae
to M. leprae in these patients. Procedures for trying murium and M. leprae are not related spe­cies but
to achieve this objective include: that there is a relatedness between M. lep­raemurium
1. Intravenous injection of peripheral blood lym­ and M. avium. M. lepraemurium can be maintained
phocytes obtained from patients with tuber­ for months in tissue cultures of monocytes, where
culoid leprosy or from healthy donors possess­ing it has a generation time of about 7 days. It has also
vigorous CMI and showing a strongly posi­tive been cultured in rat fibroblasts, and in vitro in a
lepromin (Mitsuda) reaction. cysteine-containing medium.

www.ebook3000.com
246  |  Section 3: Systemic Bacteriology

Key Points Multiple choice questions (MCQs)


™™ Mycobacterium leprae are weakly gram-positive, less 1. All the following are used as animal models for
acid-fast than tubercle bacilli, so 5% sulfuric acid leprosy except:
is employed for decoloriza­tion after staining with a. Rhesus monkey
carbol fuchsin b. Nine-banded armadillo
™™ They fail to grow in cell-free culture media. Animal c. Slender loris
models for culture include footpads of mice, and d. Indian pangolin
nine-banded armadillo (Dasypus novemcinctus) 2. The generation time of Mycobacterium leprae is:
™™ Leprosy is classified into five types (tuberculoid a. 20 minutes b. 20 hours
leprosy, border­line tuberculoid leprosy, mid- c. 12–13 days d. 12–13 weeks
borderline leprosy, borderline lepromatous leprosy, 3. The most infectious form of leprosy is:
and lepromatous leprosy) a. Tuberculoid leprosy
™™ Lepromin test: It is a skin test for delayed hyper­ b. Borderline tuberculoid leprosy
sensitivity for studying immunity in leprosy. c. Mid-borderline leprosy
™™ Laboratory diagnosis consists of demonstra­tion of d. Lepromatous leprosy
acid-fast bacilli in the lesions, animal inoculation, 4. Tuberculoid form of leprosy is seen in patients with:
lepromin test, serology and polymerase chain a. Good cell-mediated immunity
reaction (PCR) b. Good humoral immunity
™™ Dapsone with or without rifampin is used to treat c. Deficient cell-mediated immunity
the tuberculoid form of disease; clofazimine is d. Poor humoral immunity
added for the treatment of the lepromatous form. 5. Lepromin test is used for all the following except:
The vaccines have been evaluated against leprosy a. Determine the type of leprosy
with limited success. b. Confirm diagnosis of leprosy
c.  Monitor leprosy patients to treatment with
chemotherapy
Important Questions d. Evaluate host resistance to leprosy
6. All the following statements are true for Mitsuda
1. Describe mycobacteria. Discuss the etiology, reaction except:
pathogenesis and laboratory diagnosis of leprosy.
a. A nodule develops after 3–4 weeks of injection
2. Write short notes on: of lepromin antigen
a. Cultivation of leprae bacilli b. Manifestation of cell-mediated immunity
b. Animal models in leprosy c. Negative in tuberculoid leprosy
c. Lepromatous leprosy d. Negative in lepromatous leprosy
d. Tuberculoid leprosy 7. Fernandez reaction in lepromin test appean in:
e. Differences between lepromatous and tubercu- a. 24–48 hours b. 3 days
loid leprosy c. 2 weeks d. 3–4 weeks
f. Lepromin test Answers (MCQs)
g. Mycobacterium lepraemurium. 1. a; 2. c; 3. d; 4. a; 5. b; 6. c; 7. a
34
Chapter

Nontuberculous Mycobacteria

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Name the diseases caused by nontuberculous
be able to: mycobacteria
∙∙ Classify nontuberculous mycobacteria and ∙∙ Discuss the clinical significance of nontuberculous
examples of different groups of nontuberculous mycobacteria.
mycobacteria

INTRODUCTION Differentiating characterstics of M. tuberculosis


and ‘nontuberculous mycobacteria (NTM) are
Mycobacteria other than human or bovine tubercle shown in Table 34.1.
bacilli, may occasionally cause human disease
resembling tuberculosis. This large group of PROPERTIES OF NONTUBERCULOUS
mycobac­teria have been known by several names;
atypical,anonymous, unclassified, paratubercle,
MYCOBACTERIA (TABLE 34.1)
tuberculoid, environmental or nontuberculous 1. Temperature: They can grow at 25°C, 37°C, and
mycobacteria (NTM), opportunistic MOTT even at 44°C.
(mycobacteria other than tubercle bacilli). 2. Rate of growth: Some of them are rapid
The names ‘environmental’ or ‘opportunistic growers. They produce colonies within 1–2
mycobacteria’ are better suited as their natural weeks of incubation in the Lowenstein-Jensen
habitat appears to be soil or water and they cause (LJ) medium.
opportunistic infec­tions in human beings. The 3. Colony characters: Some of them may produce
name ‘nontuberculous mycobacteria (NTM)’ has bright yellow or orange pigments during their
gained wide acceptance in recent years. growth on the L–J medium.
Table 34.1  Differentiating characterstics of M. tuberculosis and ‘nontuberculous mycobacteria (NTM)
Characteristics M. tuberculosis Nontuberculous mycobacteria (NTM)
1. Cuture
Temperature 37° 25–45°C
Rate of growth Slow Slow or rapid
Growth on L–J medium Eugonic Dysgonic
Colony characters Dry, rough, tough, buff colored, Dry, yellow, orange or creamy and easily
difficult to emulsify emulsifiable
Growth in the presence of – +
p-nitrobenzoic acid (PNB) 500 µg/mL
2. Biochemical reactions
Niacin test + –
Nitrate reduction test + –
3. Animal pathogenicity Pathogenic to guinea pig Pathogenic to guinea pig
4. Transmission Person to person Soil or water

www.ebook3000.com
Chap-34.indd 247 15-03-2016 11:14:38
248  |  Section 3: Systemic Bacteriology
4. Staining: They are acid fast as well as alcohol is principally isolated from cases of pulmonary
fast. They may differ or resemble in their disease. Natural reservoir is tap water. Pulmonary
morphology from those of tubercle bacilli. disease is the most common clinical form of M.
5. Biochemical reactions: They are arylsulfatase kansasii infection. It occurs pri­marily in middle-
test positive, but are niacin and neutral red aged or elderly white men, most of whom have
reactions negative. some pre-existing form of lung disease.
6. Animal pathogenicity: They are nonpathogenic M. kansasii may also occasionally cause
for guinea pigs but pathogenic for mouse. infections of the cervical lymph nodes, penetrating
7. Treatment: They are usually resistant to anti­ wound infections, and granulomatous synovitis. It
tubercular drugs such as streptomycin, INH, can produce generalized infection in HIV-positive
and para-aminosalicylic acid. patients.
8. Transmission: Soil or water Mycobacterium marinum (M. marinum): M.
9. Classification: They are classified by Runyon marinum (previously termed the fish tubercle
into four groups on the basis of their rate of bacillus) the cause of a warty skin infection known
growth and their ability to produce pigments as swimming pool granuloma. Microscopically, it
in the presence or absence of light. resembles M. kansasii. but can be differentiated by
its poor growth at 37°C, negative nitratase, positive
CLASSIFICATION pyrazinamide hy­drolase and L-fucosidase activities.
Runyon(1959) classified NTM into four groups Mycobacterium simiae (M. simiae): M. simiae,
(Table 34.2) based on phenotypic char­acteristics of which, like M. kansasii, grows at 37°C and is
the various species, most notably pigment (yellow occasionally involved in pulmonary disease. Several
or orange) production and rate of growth. These photochromogenic mycobacteria were isolated
include: from monkeys exported from India. They have
∙∙ Group I: Photochro­mogens subsequently been associated with pulmo­nary
∙∙ Group II: Scotochromogens disease in human beings.
∙∙ Group III: Nonphotochromogens
∙∙ Group IV: Rapid growers. Group II: Scotochromogens
Species identification depends on several
additional characteristics (Table 34.2). The scotochromogens are slow-growing NTM
whose colonies are pigmented (yellow-orange-red)
when grown in the dark or the light. They are widely
Group I: Photochromogens distributed in the environment and sometimes
Photochromogens develop a bright yellow or contaminate cultures of tubercle bacilli.
orange coloration if young cultures are exposed to a i. Mycobacterium scrofulaceum (M. scrofulaceum):
light source. They are slow growing, though growth M. scrofulaceum is principally associated with
is faster than that of tubercle bacil­li. The important scrofula or cervical lymphadenitis in children,
species in this group are M. kansasii, M. marinum but also causes pulmonary disease.
and M. simiae.
ii. Mycobacterium gordonae (M. gordonae):
Mycobacterium kansasii (M. kansasii): Myco­ M. gordonae (formerly M. aquae), frequently
bacterium kansasii, which grows well at 37°C and found in water and a common contaminant of
clinical material, is a rare cause of pulmonary
Table 34.2  Runyon classification scheme of non- disease.
tuberculous mycobacteria iii. Mycobacterium szulgai (M. szulgai): M.
szulgai, an uncommon cause of pulmonary
Runyon group Name Species
disease and bursitis.It is a scotochromogen
I Photochromogens M. kansasii when incubated at 37°C but a photochromogen
M. marinum
M. simiae at 25°C.
II Scotochromogens M. scrofulaceum
M. gordonae
Group III: Nonphotochromogens
M. szulgai The nonphotochromogens are slow-growing NTM
III Non- M. auium whose colonies produce no pigment whether they
photochromogens M. intracellulare are grown in the dark or the light. Colonies may
M. xenopi resemble those of tubercle bacilli.
M. ulcerans Of the organisms classified in this group, those
M. malmoense
belonging to nonpathogenic for humans are M.
IV Rapid growers M. chelonei terrae complex and M. gastri. Medically important
M. fortuitum
species are M. avium. M. intracellulare, M. xenopi

Chap-34.indd 248 15-03-2016 11:14:38


Chapter 34: Nontuberculous Mycobacteria  | 249
and the skin pathogen M. ulcerans. The most M. fortuitum: M. fortuitum (the frog tubercle
prevalent and important opportunistic pathogens of bacillus), some isolates of which are sometimes
man are: M. avium and M. intracellulare. classified as M. peregrinum. M. fortuitum can
Mycobacterium avium: M. avium (the avian further be differentiated from M. chelonei in
tubercle bacillus) which causes natural tuberculosis reducing nitrate and assimilation of iron from ferric
in birds and lymphadenopathy in pigs. The closely ammonium citrate.
related M. intracellulare is commonoly known as Disease: Both species occasionally cause
the ‘Battey bacillus’ because it was first identified pulmonary or disseminated disease but are princi­
as a human pathogen at the Battey State Hospital pally responsible for postinjection abscesses and
for Tuberculosis, Georgia, USA. M. avium and M. wound infections. Outbreaks of abscesses following
intracellulare are so closely similar that these two injection of vaccines and other preparations
species are usually grouped together as the M. contaminated by these mycobacteria have been
avium complex (MAC). reported on a number of occasions.
Organisms of the M. avium complex cause
tuberculosis in birds and lymphadenitis in pigs as SAPROPHYTIC MYCOBACTERIA
well as occasional disease in various other wild and
domestic animals. In man, they are responsible All the chromogenic rapid growers are sapro­
for lymphadenopathy, pulmonary lesions and phytes (for example, M. smegmatis, M. phlei).
disseminated disease, notably in patients with the Mycobacterium gor­donae and Mycobacterium terrae
acquired immune deficiency syndrome (AIDS). are among the saprophytic species that have been
associated with human disease on rare occasions.
M. xenopi: M. xenopi was first isolated from a skin
lesion in a South African toad (Xenopus laevis). It Mycobacterium smegmatis: Since M. smegmatis
grows poorly at 37°C, is a thermophile and grows is normally present in smegma around the orifice
well at 45°C. It has been isolated from water, from of urethra and is a frequent contam­inant of urine
both hot and cold taps, and from granulomatous specimens. Some strains of M. smeg­matis are acid-
lesions in swine. M. xenopi produces a chronic fast but not alcohol-fast. Therefore, they are not
slowly progressive pulmonary disease, which is seen in a Ziehl–Neelsen smear if acid alcohol is
clinically and radio­logically similar to tuberculosis. used as decolorizer.
M. malmoense: M. malmoense grows very slowly, Mycobacterium phlei: M. phlei is rarely encountered
often taking as long as 10 weeks to appear on and is nonpatho­genic. It can be differentiated from
primary culture. It causes pulmonary disease and M. smegmatis by its ability to grow at 52°C and
lymphadenitis. It was first isolated from patients survive heating at 60°C for 4 hours.
from Malmo in Sweden. It is resist­ant to isoniazid
and rifampicin and sometimes also to streptomycin Pathogenesis
and ethambutol. Four main types of opportunist mycobacterial
M. ulcerans: The bacillus grows on Lowenstein- disease have been described in man (Table 34.3).
Jensen medi­u m slowly, in 4–8 weeks. The
temperature of incuba­tion is critical; growth occurs Table 34.3  Principal types of opportunist myco­
between 30°C and 33 °C. A toxin is produced by M. bacterial disease in man and the usual causative agents
ulcerans that causes inflammation and necrosis
when injected into the skin of guinea pigs. This is Disease Usual causative agent
the only known instance of toxin produced by any A. Lymphadenopathy M. avium complex
Mycobacterium species. M. scrofulaceum
B. Skin lesions M. chelanae
Group IV: Rapid Growers 1. Post-trauma abscesses M. fortuitum
This is a heterogeneous group of mycobacteria M. terrae
capable of rapid growth, colonies appearing within 2. Swimming pool granuloma M. marinum
seven days of incubation at 37°C or 25 °C. Within 3. Buruli ulcer M. ulcerons
the group, photochromogenic, scotochromogenic, C. Pulmonary disease M. avium complex
and non-chromogenic species oc­cur. Most of these M. kansasii
are purely environmental saprophytes. Only two M. xenopi
of the rapidly growing species are well recog­nized M. malmoense
human pathogens: M. chelonae and M. fortuitum. D. Disseminated disease
1. AIDS-related M. avium complex
Mycobacterium chelonae: M. chelonae (the turtle
M. genevense
tubercle bacillus), some isolates of which are
sometimes classified as M. abscessus, M. chelonae 2. Non-AIDS-related M. avium complex
M. chelonae
grows better at 25°C than at 37°C.

www.ebook3000.com
Chap-34.indd 249 15-03-2016 11:14:38
250  |  Section 3: Systemic Bacteriology
A. Lymphadenitis: Most patients are chil­dren aged forming indolent ulcers which slowly extend under
less than 5 years. The M. avium complex is the the skin. Initially, smears from the edge of the ulcer
predominant cause worldwide. Some reports claim show large clumps of bacilli which are acid fast and
a high incidence of M. scrofulaceum. alcohol fast. Later, the immunoreactive phase sets in
and the bacilli disappear. The ulcers then heal with
B. Skin lesions: Three main types have been
disfigur­ing scars.
described:
C. Pulmonary disease: These infections resemble
1. Postinjection (and posttraumatic) abscesses
tuberculosis. In most but not all cases, there is some
2. Swimming pool granu­loma
predisposoing lung disese. This is most frequently
3. Buruli ulcer.
seen in middle-aged or elderly men with lung
1. Postinjection abscesses: These are usually
damage. The disease may be caused by many
caused by the rapidly growing pathogens M.
species, but the most frequent are the M. avium
chelonae and M. fortuitum. Abscesses occur
complex and M. kansasii.
when batches of injectable materials are con­
Organism must be isolated repeatedly from the
taminated by these bacteria. Abscesses are
sputum to diffrentiate true disease from transient
painful and last for many months.
colonization.
2. Swimming pool granuloma: It is caused by
D. Disseminated disease: Up to a half of all
M. marinum and is also known as fish tank
persons dying of AIDS in the USA had disseminated
granuloma and fish fancier’s finger. M. marinum
myco­bacterial disease in the 1980s and early 1990s,
is a natural pathogen of cold-blooded animals,
almost always due to the M. avium complex.
causing tuberculosis in fish and amphibia.
Human infection originates from contaminated
swim­m ing pools or fish tanks. The lesion, Laboratory Diagnosis
beginning as a pa­pule and breaking down to A. Specimen: Sputum, pus or exudates.
form an indolent ulear, usually follows abrasions. B. Microscopy: Ziehl–Neelsen staining of smear
It was first described from Sweden under shows acid fast bacilli. Reapeted smear
the name ‘swimming pool granuloma’, examination is necessary.
and the bacillus was named M. balnei (from C. Culture: They grow well on LJ medium.
balneum, meaning bath). The disease is usually Several LJ media should be inoculated with
self-limiting although chemother­a py with the specimen. These are incubated in the dark
minocycline, co-trimoxazole or rifampicin with and in the light at different temperatures for
ethambutol hastens its resolution. distinguishing the species.
3. Buruli ulcer: This disease, caused by M. D. Identification: There is no universally recognized
ulcerans, was first described from human skin identification scheme, although reliance is
lesions in Australia (1948). The name Buruli is usually placed on cultural characteristics (rate
derived from the Buruli dis­trict of Uganda where and temperature of growth and pigmentation),
a large outbreak was extensively investigated. various biochemical reac­tions and resistance
Ulcers are usually seen on the legs or arms. to antimicrobial agents (Table 34.4). The
Indurated nodules appear, which break down most discriminative methods are based on

Table34.4  Differentiation between tubercle bacilli and some species of atypical mycobacteria
Test M. M. M. M M M. M. avium M. M. M. M.
tubercu bovis microti kansasii marinum scrofula­ intra­­ fortuitum chelonei phlei smegmatis
losis ceum cellulare
complex
Growth in – – – – – – – + + + +
7 days
Growth at – – – + + + ± + + + +
25°C
Growth at + + + + ± + + + + + +
37ºC
Growth at – – – – – ± – – – + +
45°C
Pigment – – – + + + – – – + –
in light
Pigment – – – – – + – – – + –
in dark
Niacin + – ± – – – – – – – –
Nitrate + – – + – – – + – + +
reduction
Urease + + + – + + – + + + +

Chap-34.indd 250 15-03-2016 11:14:38


Chapter 34: Nontuberculous Mycobacteria  | 251
the detection of sequence differences in 16S 2. Write short notes on:
ribosomal RNA. a. Atypical mycobacteria.
b. Differentiation of typical mycobacteria from
atypical mycobacteria
Epidemiology c. Photochromogenic atypical mycobacteria
d. Scotochromogenic atypical mycobacteria
Environmetal mycobacteria are widely distributed
e. Mycobacterium ulcerans
in nature. Infection with them is quite common, f. Mycobacteria causing skin ulcers
from soil, water and air. Infec­t ion is mainly g. Swimming pool granuloma.
asymptomatic. In countries in which tuberculosis is
‘uncommon, opportunist mycobacterial infections
MULTIPLE CHOICE QUESTIONS (MCQs)
are relatively common. In addition, the absolute
incidence is increasing as a result of the growing 1. Nontuberculous mycobacteria or atypical
number of immunocompromised individuals, mycobacteria show following features except:
notably patients with AIDS.Some opportunist a. They are acid fast as well as alcohol fast
species may colonise tap water. When staining b. They are arylsulfatase test positive
c. They are niacin test positive
reagents were prepared from contaminated water,
d.  They are usually resistant to antitubercular
false-positive sputum smear examinations for acid- drugs such as streptomycin, INH, and p-ami-
fast bacilli have occurred. nosalicylic acid
2. All the following are scotochromogens except:
Treatment a. Mycobacterium kansasii
b. Mycobacterium scrofulaceum
Most environmental mycobacteria are resistant c. Mycobacterium gordonae
to the usual antituberculosis drugs although d. Mycobacterium szulgai
infections often respond to various combinations 3. All the following species are rapid growers except:
of these drugs. a. Mycobacterium fortuitum
b. Mycobacterium chelonae
c. Mycobacterium abscessus
Key Points
d. Mycobacterium ulcerans
™™ Mycobacteria other than the tubercle and lepra 4. Which of the following bacteria cause/s pulmonary
bacilli are known as nontuberculous mycobacteria disease?
™™ Classification: Runyon(1959) classified NTM into a. Mycobacterium avium-intracellulare
four groups: Group I Photochro­mogens; Group II b. M. kansasii
Scotochromogens; Group III Non­photochromogens c. M. xenopi
and Group IV Rapid Growers d. All of the above
™™ Disease: A. Localized lymphadenitis; B. Skin lesions
5. Swimming pool granuloma is caused by:
following traumatic inoculation of bacteria; C.
a. M. ulcerans
Tuberculosis-like pulmonary lesions; D. Disseminated
disease b. Mycobacterium marinum
c. M. chelonei
™™ Laboratory diagnosis: Ziehl–Neelsen staining of
d. M. ortuitum
smear shows acid fast bacilli. They grow well on
L–J medium.There is no universally recognized 6. Buruli ulcer is caused by:
identification scheme a. Mycobacterium marinum
b. M. fortuitum
c. Mycobacterium ulcerans
IMPORTANT QUESTIONS d. Mycobacterium xenopi
1. Discuss the classification of atypical mycobacteria ANSWERS (MCQs)
and name the diseases caused by these bacteria. 1. c; 2. a; 3. d; 4. d; 5. b; 6. c

www.ebook3000.com
Chap-34.indd 251 15-03-2016 11:14:38
35
Chapter

Actinomycetes

Learning Objectives

After reading and studying this chapter, you should ∙∙ Discuss laboratory diagnosis of actinomycosis
be able to: ∙∙ Differentiate between the genera Actinomyces and
∙∙ Describe morphology of Actinomyces Nocardia
∙∙ Explain clinical forms of Actinomyces ∙∙ Describe the following: nocardiosis, mycetoma.

Introduction Morphology
Actinomyces are Gram-positive, nonmotile, non­
Actinomycetes are traditionally considered to be
sporing, nonacid-fast. They often grow in mycelial
tran­sitional forms between bacteria and fungi.
forms and break-up into coccoid and bacillary
They form a mycelial network of branching
forms. Most show true branch­ing.
filaments like fungi but, like bacteria, they are
thin, possess cell walls con­taining muramic acid,
have prokaryotic nuclei and are susceptible to
Cultural Characteristics
antibacterial antibiotics. They are there­fore true They are facultative anaerobes. They grow best
bacteria, bearing a superficial resemblance to under anaerobic or micro-aerophilic conditions
fungi. Actinomycetes are related to mycobacteria with the addition of 5–10% CO2. The optimum tem­
and corynebacteria. perature for growth is 35–37°C. They can be grown
They are gram-positive, nonmotile, nonsporing, on brain heart infusion agar, heart infusion agar
noncapsulated filaments that break up into supplemented with 5% defibrinated horse, rabbit
bacillary and coccoid elements. Although mostly or sheep blood. Suitable liquid media include brain
soil saprophytes, occa­s ionally cause chronic heart infusion broth and thioglycollate broth which
granulomatous infections in animals and man. may be supplemented with 0.1–0.2% sterile rabbit
serum. Most species show good growth after 3–4
Important genera: The family Actinomycetes days incubation however, A. israelii may take 7­–14
contains three major medically important genera, days.
Actinomyces, Nocardia and Actinomadura. Another
genus, Streptomyces rarely causes disease in man.
Pathogenesis
Actinomyces is anaerobic or microaerophilic
and nonacid-fast, while Nocardia is acid-fast and Actinomycosis: The Actinomyces causes the
aerobic. Streptomyces is nonacid-fast and aerobic. disease known as actinomycosis. Actinomycosis is a
chronic disease characterized by multiple abscesses
and granulomata, tissue destruction, extensive
Actinomyces
fibrosis and the formation of sinuses. Within
A mould-like organism in the lesion of ‘lumpy jaw’ diseased tissues the actinomycetes form large
(actinomycosis) in cattle was found by Bollinger masses of mycelia embedded in an amorphous
(1877).The name Actinomyces was coined by protein–polysac­charide matrix and surrounded by
Harz to refer to the raylike appearance of the a zone of gram-nega­tive, weakly acid-fast, club-like
organism in the granules that characterize the structures (Fig. 35.1). The mycelial masses may
lesions (Actinomyces, meaning ray fungus). This be visible to the naked eye and are called sulfur
was named Actinomyces israelii. It causes human granules, as they are often light yellow in color. The
actinomycosis. sulfur granules may be dark brown and very hard in
Chapter 35: Actinomycetes  | 253
using 1% sulfuric acid for decolorization. Gram
staining shows a dense network of thin gram-positive
filaments, surrounded by a peripheral zone of
swollen radiating club shaped structures, presenting
a sun-ray appearance (Fig. 35.1). The ‘clubs’ are
believed to be antigen-antibody complexes.Acid
fast staining shows central part as non-acid­fast
surrounded by acid-fast ‘clubs’. In absence of sulfur
granules, Gram’s staining of pus shows gram-positive
branching filaments.
Sulfur granules and mycelia in tissue sections
Fig. 35.1: Sulfur granule. Section of tissue showing an
actinomycotic clolony, the clubs at the periphery giving a can also be identified by direct fluorescence
‘sun-ray’ appearance microscopy.
older lesions because of the deposition of calcium
3. Culture
phosphate in the matrix.
In man, actinomycosis.is usually caused by Sulfur granules or pus containing actinomycetes are
Actinomyces israelii. Less common causes include washed and inoculated into thioglycollate liquid
A. gerencseriae, A. naeslundii, A. odontolyticus, A. medium or streaked on brain h ­ eart infusion agar
viscosus, A. meyeri, Propionibacterium propionicum (BHI agar) blood agar and incubated anaerobically
and members of the genus Bifidobacterium. at 37°C. On solid media, A. israelii may form
so-called spider colonies that resemble molar teeth
in 48–72 hours that become heaped up, white and
Human Actinomycosis
irregular or smooth, large colonies in 10 days. Other
Human actinomycosis may take several forms: species have different types of colonies.
1. Cervicofacial: This is the most common type
and it occur mainly in cheek and submaxillary 4. Identification
regions. The disease is endogenous in origin. The identity may be confirmed by direct fluo­
Dental caries is a predisposing factor, and rescence microscopy and biochemical tests or
infection may follow tooth extractions or other by gas chromatography of metabolic products of
dental procedures. carbohydrate fermentation.
2. Thoracic: Thoracic actinomycosis commences
in the lung, probably as a result of aspiration of 5. Biopsy
actinomyces from the mouth. In hematoxylin and eosin stained sections, the
3. Abdominal: The lesion is usually around the sulfur granules are deeply stained with hematoxy­
cecum, with the involvement of the neighboring lin except in the periphery which is stained by
tissues and the abdominal wall. eosin, which shows short, radiate, club-like
4. Pelvic: Pelvic actinomycosis occasionally structures. On Gram staining, the fila­ments are
occurs in women fitted with plastic intra- gram-positive and periphery gram-nega­tive.
uterine contraceptive devices.
5. Punch actinomycosis: It is a rare infection of Epidemiology
the hand acquired by injury of the knuckles on
Actinomycosis is an endogenous infection.
an adversary’s teeth.
Actinomycosis is more common in rural areas and
Laboratory Diagnosis in agricultural workers. Young male persons (10–30
years old) are most commonly affected. About
1. Specimens 60% of the cases are cervicofacial and some 20%
Pus, sinus discharge, bronchial secretions, sputum abdominal. Pelvic actinomyces is seen mainly in
or infected tissues are collected aseptically. women using intrauterine devices.

2. Microscopy Treatment
‘Sulfur granules’ may be demonstrated in pus Treatment for actinomycosis involves the combin­
by shaking it up in a test tube with some saline. ation of surgical debridement of the involved
On standing, the granules sediment may be tissues and prolonged treatment with penicillin or
withdrawn with a capillary pipette. Granules may tetracycline.
also be obtained by applying gauze pads over the
discharging sinuses. Nocardia
Granules are crushed between two slides and Nocardia resemble Actinomycetes morphologically
stained with Gram and Ziehl–Neelsen staining but are aerobic. Most species (such as N. asetroides

www.ebook3000.com
254  |  Section 3: Systemic Bacteriology
Table 35.1  Differences between the genera Cutaneous infection: Primary or secondary
Actinomyces and Nocardia cutaneous infection may lead to mycetoma, lymph­
ocutaneous infections, cellulites, subcutane­ous
Actinomyces spp. Nocardia spp.
abscesses.
•  Facultative anaerobes •  Strict aerobes
•  Grow at 35–37°C •  W
 ide temperature Laboratory Diagnosis
range of growth
Diagnosis is by demonstration of branching fila­
•  Oral commensals •  E nvironmental
ments microscopically and by isolation in culture.
saprophytes
•  Nonacid-fast mycelia •  Usually weakly acid-fast
1. Specimens
•  E ndogenous cause of •  Exogenous cause of
disease disease
Pus or purulent sputum.

and N. brasiliensis) are acid-fast when decolorized 2. Microscopy


with 1% sulfuric acid. Nocardia are frequently The smears are stained with Gram staining and
found in soil and infection may be exogenous. Ziehl–Neelsen (Z–N) technique using decolorization
Differentiating features of Actinomyces and with 1% sulfuric acid. Gram-positive filamentous
Nocardia are shown in Table 35.1. bacteria can be seen on Gram staining. Acid-fast
bacilli are detected on Z–N technique though some
Species: The species most commonly associated
species are nonacid-fast.
with human disease are N. asteroides, N. brasiliensis,
N. farcinica, N. otitidiscaviarum, N. nova and N.
transvalensis. 3. Culture
The specimens are inoculated on nutrient agar,
Morphology Sabouraud’s dextrose agar (SDA) and brain ­heart
Nocardiae are gram-positive bacteria and form infusion agar (BHI agar) and incubated at 36°C for
a mycelium that fragments into rod shaped 3 weeks. Colony morphology is seen and bacteria
and coccoid elements. Nocardia resembles are identified by staining.
Actinomyces, but some species are acid-fast, and a Nocardia can be isolated from sputum by para­
few are nonacid-fast. ffin bait technique. The specimen is homogenized
with sterile glass beads and 2 ml of it is inoculated
Cultural Characteristics into carbon-free broth containing paraffin coated
They are strict aerobes. Nocardiae readily grow in glass rod. The organisms grow on the rod at the
ordinary media. They are slow growing (require air-liquid surface which may be subcultured onto
5–14 days). Nocardiae readily grow on nutrient agar media.
agar, Sab­ouraud dextrose agar, brain heart infusion
agar and yeast extract-malt extract agar. The Treatment
inoculated plates should be incubated at 36°C Sulphonamide are the antibiotics of choice. They
for up to 3 weeks. They can grow at wide range are also susceptible to ami­k acin, imipenem,
of temperature. Selective growth is favored by minocycline, tobramycin and van­comycin.
incubation at 45°C. In addition, the technique of
paraffin baiting may be used. Colonies of nocardiae
Actinomycotic mycetoma
are cream, orange or pink c­ olored.
Mycetoma is a localized chronic, granulomatous
Pathogenesis involvement of the subcutaneous and deeper
Nocardiae produce opportunistic pulmonary disease tissues, commonly affecting the foot and less often
known as nocardiosis in immunocompromised the hand and other parts. It presents as a tumor
individuals including those with AIDS. Soil is with multiple sinuses. This clinical syndrome was
known to be natural habitat of Nocardia. Man first described from Madura by Gill (1842) and
acquires infection by inhalation of the bacteria from came to be known as Maduramycosis. See chapter
environmental sources. The infection is exogenous, 73 for “Mycetoma” detail.
resulting from inhalation of the bacilli. Etiology: It can be divided into three types,
Bronchopulmonary disease: Systemic nocardiosis eumycetomas, actinomycetomas and botryo­
usually caused by N. asteroides manifests primarily mycosis. Bacterial mycetomas are usually caused
as pulmonary disease, pneumonia, lung abscess by actinomycetes­—Actinomyces (A. israelii, A.
or other lesions resembling tuberculosis. Systemic bovis), Nocardia (N. asteroides, N. brasiliensis, N.
nocardiosis occurs more often in immunodeficient caviae), Actinomadura (A. madurae, A. pelletierii),
persons. Streptomyces (S. somaliensis).
Chapter 35: Actinomycetes  | 255
Diagnosis: Etiological diagnosis of mycetoma
™™ Actinomycotic mycetoma: Mycetoma is a
is important in choosing appropriate treatment. localized chronic, granulomatous involvement of
The color of the granules gives some indication. In the subcutaneous and deeper tissues, commonly
actinomycotic mycetoma, the granules are white to affecting the foot and less often the hand and other
yellow, while in eumycotic mycetomas, the granules parts. Mycetomas are usually caused by fungi but
are generally black. Examination of crushed smears may be caused by bacteria as well.
of the granules helps to differentiate actinomycotic
from mycotic mycetomas. In the former, the Important question
filaments are thin (about 1 μm), while in the latter
they are stout (about 4–5 μm). lsolation of the agent Write short notes on:
in culture establishes the diagnosis. a. Actinomycosis
b. Laboratory diagnosis of actinomycosis
Key Points c. Nocardiosis
d. Laboratory diagnosis of nocardiosis
™™ Actinomycetes are traditionally considered to be
transitional forms between bacteria and fungi
e. Mycetoma.
™™ Actinomycetes are gram-positive, nonmotile, non­
sporing, noncapsulated filaments Multiple choice questions (MCQs)
™™ The family Actinomycetes contains important
genera, Actinomyces, Nocardia, Actinomadura, 1. Which of the following bacteria is/are acid fast?
Streptomyces a. Actinomyces b. Nocardia
™™ Actinomyces is anaerobic or microaerophilic and c. Streptomyces d. All of the above
nonacid-fast 2. Cervicofacial actinomycosis is caused most
™™ Actinomyces causes the disease k nown as commonly by
actinomycosis. The disease occurs in five clinical a. Actinomyces radingae
forms: cervicofacial, thoracic, abdominal, pelvic and b. Actinomyces turicensis
punch actinomycosis c. Actinomyces gerencseriae
™™ Laboratory diagnosis: The specimens include d. Actinomyces israelii
sputum, bronchial secretions, and discharges, 3. Molar-teeth-like colonies are produced by
and infected tissues which may contain sulfur a. Nocardia asteroides
granules. Gramstained smear of the granules shows b. Actinomyces israeli
gram-negative club shaped structures (sun-ray c. Actinomadura madurae
appearance)
d. Tropheryma whippelii
™™ Nocardia: Nocardia resemble Actinomycetes
4. Paraffin bait technique is used for the isolation of
morphologically but are aerobic. The nocardiae are
branched, strictly aerobic, gram-posi­tive bacteria,
Nocardia from:
are acid-fast when decolorized with 1% sulfuric acid a. Soil b. Sputum
™™ Nocardia species cause primary cutaneous nocardiosis, c. Both of the above d. None of the above
bronchopulmonary infection, and secondary CNS Answers (MCQs)
infection 1. b; 2. d; 3. b; 4. c

www.ebook3000.com
36
Chapter
Enterobacteriaceae: Escherichia,
Klebsiella, Proteus and Other Genera

LEARNING OBJECTIVES

After reading and studying this chapter, you should –  Discuss various groups of E. coli producing diarrhea
be able to: – Differentiate between heat labile toxin (LT) and
∙∙ Describe general characters of the family Enter­ heat stable toxin (ST) of E. coli
obacteriaceae – Discuss laboratory diagno­sis of urinary tract
–  Classify the family Enterobacteriaceae infections caused by E. coli
– Describe morphology, culture characteristics and – Discuss morphology, culture characteristics and
biochemical reactions of E. coli biochemical reactions of Klebsiella sp
–  Discuss pathogenicity of E. coli –  Differentiate between E. coli and Klebsiella sp.

INTRODUCTION CLASSIFICATION OF
The family Enterobacteriaceae is the largest, most ENTEROBACTERIACEAE
heterogeneous collection of medically important
Gram-negative bacilli. Because of their normal
Lactose Fermentation
habitat in humans, these organisms are referred The oldest method was to classify these bacteria
to as the “enteric bacilli” or “enterics” into three groups based on their action on
lactose. Lactose fermenters (LF), Late lactose
CHARACTERISTICS OF THE FAMILY fermenter and non­lactose-fermenting (NLF).
ENTEROBACTERIACEAE The specimen is plated on a medium containing
lactose and neutral red indicator. (MacConkey
Members of the family Enterobacteriaceae have agar). The organisms fermenting the lactose form
the following characteristics: acid and in acidic pH, neutral red is red in colour,
i. They are gram-negative bacilli. therefore, the colonies of lactose-fermenting
ii. They are aerobes or and facultative anaerobes bacteria are red or pink and those of nonlactose-
and grow readily on ordinary laboratory media fermenting (NLF) bacteria are pale. All lactose-
including MacConkey’s lactose bile-salt agar. fer­m enting enterobacteria, e.g. Escherichia,
iii. All species ferment glucose with the production Klebsiel­l a, Enterobacter and Citrobacter are
of acid or acid and gas. popularly known as ‘coliform bacilli’ as the
iv. They are either non-motile or motile with most common mem­ber of this group is the colon
peritrichous flagella. bacillus or Escherichia coli. The major intestinal
pathogens, Salmonella and Shigella are non-
v. They are catalase positive (except for Shigella
Iactose-fermenters (NLF). There remained a
dysenteriae type 1 which is catalase-negative)
small group which showed delayed fermentation
vi. They are oxidase-negative. of lac­tose (2–8 days) and with the exception of
vii. They reduce nitrate to nitrites Shigella sonnei, they were all commensals. This
viii. They are typically intestinal parasites of heterogenous group of late lactose fermenters was
humans and animals. called paracolon bacilli.

Chap-36.indd 256 15-03-2016 11:15:18


Chapter 36: Enterobacteriaceae: Escherichia, Klebsiella, Proteus and Other Genera  | 257
Table 36.1  Classification of the family Entero­ It can grow on ordinary media such as nutrient
bacteriaceae agar. Colonies are large, thick, grayish white, moist,
smooth opaque or partially translucent disks. This
Tribe Genus
description applies to the smooth (S) form seen on
i. Escherichiae 1. Escherichia fresh isolation. On blood agar, many strains, are
2. Shigella
hemolytic. On MacConkey’s medium, colonies
ii. Edwardsiella Edwardsiella are red or pink due to lactose fermentation. On
iii. Salmonellae Salmonella selective media such as DCA or SS agar growth
iv. Enterobacteriaceae Citrobacter is largely inhibited used for the isolation of
v. Klebsiellae 1. Klebsiella salmonellae and shigellae. In broth, growth occurs
2. Enterobacter as general turbidity and a heavy deposit.
3. Serratia
4. Hafnia Biochemical Reactions
vi. Proteeae 1. Proteus i. E.coli ferments glucose, lactose, mannitol,
2. Morganella maltose and many other sugars with the
3. Providencia production of acid and gas. Typical strains do
vii. Yersiniae Yersinia not ferment sucrose.
ii. Indole and MR posi­tive, and VP and citrate
negative (IMViC + + – –)
CLASSIFICATION OF iii. It is negative for phenylalanine deaminase
ENTEROBACTERIACEAE BY TRIBES test, urease test, H 2S production, gelatin
Three systems of nomenclature have been liquefication, growth in the presence of KCN,
proposed (Bergey’s manual, Kauffmann, Edwards­ and malonate utilization.
-Ewing) have certain differences, the general
ap­proach is the same. Antigenic Structure
The family is first classified into its major Serotyping of E. coli is based on three antigens—
subdivision—group or tribe. Each tribe consists the flagellar antigen H, somatic antigen O and the
of one or more genera and each genus one or capsular antigen K as detected in agglutination
more subgenera and species. The species are assays with specific rabbit antibodies.
clas­s ified into types—biotypes, serotypes, 1. H antigens: These are thermolbile. So far 75
bacteri­ophage types, colicin types. Table 36.1 antigens have been identified. There are only a
lists the in the family Enterobacteriaceae and their few significant cross-reactions between them
respective tribes and genera. Table 36.2 shows the and with the H antigens of other members of the
biochemical features that differ­entiate different Enterobacteriaceae. Strains may need to be grown
genera of Enterobacteriaceae. in semi-solid agar to induce flagella expression.
2. Somatic antigen (O antigen): These are
ESCHERICHIA COLI heat-stable, lipopolysaccharide antigens of
cell walls. Over 173 different O antigens have
Introduction been described. Serotyping may detect cross-
This genus is named after the German pediatrician reactions because of shared epitopes on the LPS
Theodar Escherich who first identified Escherichia expressed by strains of E. coli and organisms
coli (1885). The genus Escherichia consists of five belonging to the genera Brucella, Citrobacter,
species of which E. coli is the most common and Providencia, Salmonella, Shigella and Yersinia.
clinically most impor­tant. Unlike other coliforms, The normal colon strains belong to the
E. coli is a obligate parasite living only in the human ‘early’ O groups (1, 2, 3, 4 etc.), while the
or animal intestine that cannot live free in nature. enteropathogenic strains belong to the ‘later’ O
groups (26, 55, 86, 111, etc.).
Morphology 3. Capsular antigen (K antigen): K antigen’ refers
to the acidic polysaccharide anti­gen located in
E. coli is a gram-negative, straight, rod measuring the ‘envelope’ or microcapsule. (K for Kapsel,
1–3 × 0.4–0.7 µm arranged singly or in pairs. It German for capsule). It encloses the O anti­gen
is motile by peritrichate flagella, though some and renders the strain in agglutinable by the O
strains may be nonmotile. It is nonsporing and antiserum. It may also contribute to virulence
noncapsulated. by inhibiting phagocytosis.
In the past, these antigens were divided into
Cultural Characteristics three classes—L, A and B (the thermolabile
It is an aerobe and a faculta­tive anaerobe. The L anti­g ens, the thermostable A and B
temperature range is 10–40°C (op­timum 37°C). antigens) according to the effect of heat on

www.ebook3000.com
Chap-36.indd 257 15-03-2016 11:15:18
Chap-36.indd 258
Table 36.2  Important distinguishing features of the different genera of Enterobacteriaceae
Test Escherichia Shigella1 Edwardsiella Klebsiella Enteobacter Serratia Hafnia Citrobacter Salmonella2 Proteus Morganella Providencia
Motility + – + – + + + + + + + +
Gas from glucose + – + + + d + + + d + +
Acid from lactose + – – + + – – + – – – –
Acid from sucrose d – – + + + – d – d – d
Growth in KCN – – – + + + + + d + + +
Indole + d + – – – – d – d + +
MR + + + – – – – + + + + +
258  |  Section 3: Systemic Bacteriology

VP – – – + + + + – – – – –
Citrate – – – + + + + + + d d d
H2S – – + + – – – + + + – –
Urease – – – + d – – – – + + d
Phenylalanine – – – – – – – – – + + +
deaminase (PPA)
Arginine d – – – d – – d + – – –
dehydrolase
Lysine + – + d d + + – + – – –
decarboxylase
Ornithine d d + – + + + d + d + –
decarboxylase
(d = results different in different species or strains)
Important exceptions:
1. Sh. sonnei ferments lactose and sucrose late
2. S. Typhi does not produce gas from sugars

15-03-2016 11:15:18
Chapter 36: Enterobacteriaceae: Escherichia, Klebsiella, Proteus and Other Genera  | 259
Table 36.3  K antigens (group I and group II) of colonization factor antigens (CFAs) (CFAI,
E. coli CFAII, CFA/III) expressed by enterotoxigenic E.
coli (ETEC) causing diarrheal disease in humans.
Properties Group I Group II
1. Molecular weight >100 000 <50 000 B. Toxins
2. O groups 08.09 Many 1. Exotoxins: E. coli produces two kinds of
3. Acidic component Hexuronic acid Glucuronic acid, exotoxins hemolysins and enterotoxins.
phosphate, a. Hemolysins: Hemolysins do not appear to
KDO, NeuNAc be relevant in pathogenesis.
4. Electrophoretic Low High b. Enterotoxins: Three distinct types of E. coli
mobility enterotoxins have been identified:
5. Expressed at Yes No i. Heat-labile toxin (LT): Mechanism
17-20°C of action of LT: E. coli LT is heat labile
6. Chromosome site His SerA protein and is closely related to the
KDO, ketodeoxyoctonate; NeuNAc, N-acetylneuraminic toxin produced by strains of Vibrio
acid cholerae. There are two main forms,
termed LT-I and LT-II. Different forms
of LT-1 have been described. Similarly,
the agglutinability, antigenicity and antibody two forms of LT-II (LT-IIa and LT- lIb)
binding power of bacterial strains that express have been detected.
them. Later it was shown that the B antigen was LT is a complex of polypeptide subunits-
not a separate entity. K antigens are therefore each unit of the toxin consisting of
cur­rently classified into two groups, I and II, one subunit A (A for active) and five
(Table 36.3). subunits B (B for binding). The toxin
4. Fimbrial antigen (F antigen): These are binds to the Gm1 ganglioside receptor
thermolabile proteins. Heating the organisms on intestinal epithelial cells by means of
at 100°C leads to detachment of fimbriae. The subunit B, following which the subunit
F antigen has no role in antigenic classification A is activated to yield two fragments—A1
of E. coli. and A2. The A1 fragment activates adenyl
cyclase in the enterocyte to form cyclic
Virulence Factors
adenosine 5’ monophosphate (cAMP),
Two types of virulence factors have been recognized leading to increased outflow of water
in E. coli—surface antigens and toxins. and electrolytes into the gut lumen, with
consequent diarrhea.
A. Surface Antigens LT is a powerful antigen and can
1. Somatic antigen (O antigen): The somatic therefore, be detected by a number of
lipopolysaccharide surface O antigen, besides serological as well as biolog­ical tests.
exerting endotoxic activity, also protects the ii. Heat-stable toxin (ST): The heat stable
bacillus from phagocytosis and the bactericidal toxins of E. coli (ST) have a low molecular
effects of complement. weight which is probably responsible for
2. K antigen: Most strains of E. coli responsible their heat stability and poor antigenicity.
for neonatal meningitis and septicemia carry There are two major classes, designated
the KI envelope antigen which is a virulence ST-I (or STa) and ST-II (or STb). STa is
factor resembling the group B antigen of associated with human disease.
meningococcci. a. ST-I (or STa): STa is a small, monomeric
3. Fimbriae: Like many other members of the toxin that binds to guanyl­ate cyclase,
Enterobacteriaceae, strains of E. coli exhibit leading to an increase in the level of
common fimbriae which are chromosomally cyclic guanosine monophosphate and
determined, present in large numbers and subsequent hypersecre­tion of fluids. ST-I
causing mannose sensitive hemagglutination is methanol soluble, is plasmid encoded.
and probably not relevant in pathogenesis. b. ST-II (STb)—ST-II (STb) is distinguished
Filamentous protein structures resembling from ST-I (STa) by its biological activity
fimbriae cause mannose-resistant hemagglutin­ and by its insolubility in methanol. It
ation and play an important part in the stimulates fluid accumulation in ligated
pathogenesis of diarrheal disease and in intestinal loops of young piglets (upto
urinary tract infection. They include the nine weeks) but not in the infant mouse
K88 antigen found in strains causing enteritis test. The mechanism of action is not
of pigs, the K99 antigen found in strains known but it appears not to act via cAMP
causing enteritis of calves and lambs, and the or cGMP.

www.ebook3000.com
Chap-36.indd 259 15-03-2016 11:15:18
260  |  Section 3: Systemic Bacteriology
Table. 36.4  Differentiating proper ties of age groups in the local population. In developing
verocytotoxins by E. coli countries. ETEC are a major cause of mortality in
children under the age of 5 years.Persons from
VT1 VT2 VT2v1
developed countries visiting endemic areas often
• Synonyms SLT1 SLT2 SLT2v suf­fer from ETEC diarrhea—a condition known as
• Cytotoxicity ‘trav­eller’s diarrhea’.
Vero cells + + + ETEC produce a heat-stable toxin (ST) or a
HeLa cells + + – heat-labile toxin (LT) or both (See under virulence
factors above). The organism must initially be able
Genes phage-encoded + + –
to adhere to the mucosal surface of the epithelial
1
Human and porcine variants cells of the small intestine. This adhesion is usually
mediated by fimbriae that bind to specific receptors
iii. Verocytotoxin or Verotoxin (VT): The in the intestinal cell membrane. These adhesins
biological properties, physical char­ have been termed colonization factors antigens
acteristics and antigenicity of VT are (CFAs) of which a number of have been identified
very similar to those of Shiga toxin (Stx), (CFAI, II, III,IV). Plasmids that simultaneously
produced by strains of Sh. dysenteriae carry genes for both CFAs and enterotoxin
type 1 so it is also known as ‘Shiga-like production have been described.
toxin’ (SLT). Though plasmids with enterotoxin genes may
2. VT1 and VT2 form: Serological tests have be present in any strain of E. coli, in practice only a
revealed two antigenically distinct forms, small number of serotypes become enterotoxigenic
termed VTl and VT2. Antibodies prepared to (for ex­ample, 06, 08, 015, 025, 027, 0167).
VT1 neutralize Shiga toxin, while antibodies
specific for VT2 do not. Variant forms of VT2 C. Enteroinvasive E. coli (EIEC)
(VT2v) have been described in strains of human These are closely related by phenotypic and
and porcine origin (Table 36.4). pathogenic properties to Shigella. Many of these,
VT1 and VT2, like Shiga toxin, are cytotoxic for strains are non­motile, do not ferment lactose or
Vero and HeLa cells, although VT2 variant toxins ferment it late with acid, but without producing any
do not bind to HeLa cells. Enterotoxicity in ligated gas and do not form lysine decarboxylase. Many of
rabbit gut loops and mouse paraliytic lethality can these show 0 antigen cross- reaction with shigellae.
also be used to detect VTs. These ‘atypical’ E. coli strains had earlier been
grouped under the Alkales­cens-Dispar Group
Clinical Infections and given names such as ‘Shigel­la alkalescens’
Four main types of clinical syndromes are caused (resembling Sh. flexneri except in fermenting
by E. coli. dulcitol and forming alkali in litmus milk) and ‘Sh.
dispar’ (late lactose fermenter like Sh. sonnei but
1. Diarrhea indole positive). Besides these atypical strains
At least five different types of diarrheagenic E. many typical E. coli strains can also cause clinical
coli are now recognized, each associated with illness resembling shigellosis. These have been
specific serotypes and with different pathogenic termed enteroinvasive E. coli because they have
mechanisms. the capacity to invade interstitial epithelial cells
in vivo and pene­trate HeLa cells in tissue culture.
A. Enteropathogenic E. coli (EPEC) EIEC strains usually belong to serogroups 028ac,
These have been associated mainly with diarrhea 0112ac, 0124, 0136, 0143, 0114, 0152, 0154. The
in infants and chil­d ren usually occurring as most common serogroup is 0124.
institutional outbreaks. Certain strains belonging to
characteristically EPEC serogroups, such as O26 and Pathogenesis
O111, were later shown to express verocytotoxins. EIEC, like those of Shigella species, can penetrate
Pathogenesis of EPEC diarrhea: EPEC neither the epithelial cells of the large intestine and
ordinarily produce entero­t oxins, nor are they multiply intracellularly, giving rise to blood and
invasive. In infantile enteritis, the bacilli are seen mucus in the stool. Infection is by ingestion.
to be adherent to the mucosa of the up­per small Clinically, EIEC infection resembles shigellosis,
intestine, intimately attached to cup-like projections ranging from mild diarrhea to frank dysentery, and
(pedestals) of the enterocyte membrane, causing occurs, in children as well as adults.
disruption of the brush border microvilli.
D. E. coli Verocytotoxin or Verotoxin (VT)
B. Enterotoxigenic E. coli (ETEC) E. coli verocytotoxin or verotoxin (VT) causes
Diarrhea caused by ETEC is endemic in the hemorrhagic colitis and hemolytic uremic
developing countries in the tropics, among all syndrome. Outbreaks were first recognized in

Chap-36.indd 260 15-03-2016 11:15:18


Chapter 36: Enterobacteriaceae: Escherichia, Klebsiella, Proteus and Other Genera  | 261
the USA in 1982 and strains of VTEC belonging to Etiology of UTI
serogroup 0157 emerged as the major cause.
1. Other members of family Enterobacteriaceae
Outbreaks of infection with VTEC have occurred
that usually cause UTI are Kiebsiella, Proteus,
in the community, in nursing homes for the elderly
Citrobacter, and those which rarely produce UTI
and in daycare centres for young children. The
are salmonellae, edwarosiellae and Enterobacter.
most severe clinical manifestations are usually seen
in the young and the elderly. 2. The gram-positive organisms, which can cause
UTI are Staphylococcus aureus, coagulase-
E. Enteroaggregative E. coli (EAEC) negative staphylococci, Streptococcus faecalis,
S. pyo­g enes, S. agalactiae, S. milleri, other
These strains are so named because they are
streptococci and anaerobic streptococci.
characterized by their ability to adhere to particular
laboratory-cultured cells, such as HEp-2, in an 3. Rarely, Gardnerella vaginalis may cause UTI.
aggregative or ‘stacked brick’ pattern. They have 4. Candida albicans may cause UTI in diabetic
been associated with persistent diarrhea, especially and immunocompromised patients.
in developing countries. 5. The hospital-associated infection follow­ing
Diagnosis: The only methods currently available for instrumentation and catheterization is mostly
detecting these bacteria are the HEp-2 cell test for caused by Pseudomonas and Proteus.
determining the aggregative phenotype, and DNA
probes. The HEp-2 cell test involves allowing strains 3. Pyogenic Infections
of E. coli to adhere to cell monolayers in vitro and
E. coli form the most common cause of intra-
observing the pattern of adhesion by microscopy.
abdominal infections, such as peritoni­t is and
2. Urinary Tract Infection abscesses resulting from spillage of bowel con­tents.
They also cause pyogenic infections in the perianal
E. coli and coliforms account for the large majority area. They are an important cause of neonatal
of naturally acquired urinary tract infections. Those meningitis, but is much less so in older patients.
acquired in the hospi­tal, following instrumentation,
are more often caused by other bacteria, such as
Pseudomonas and Proteus. 4. Septicemia
Most frequently encountered O serotypes of E. Bloodstream invasion by E. coli may lead to
coli in UTI include 01, 02, 04, 06,07, 018 and 075. fatal conditions like septic shock and ‘systemic
These are also known as nephritogenic strains. inflammatory response syndrome’ (SIDS). As E.
Special nephropathogenic potential of these strains coli commonly show multiple drug resist­ance,
appears to be due to: antibiotic sensitivity testing of strains is impor­tant
a. The polysaccharides of the O and K antigens in treatment.
protect the organism from the bactericidal
effect of complement and phagocytes in Laboratory Diagnosis
the absence of specific antibodies. Strains
possessing K1- or K5 antigen appear to be more a. Laboratory Diagnosis of EPEC
virulent. Fresh diarrheal feces is plated on blood agar and
b. Fimbriae mediate the adherence of the Mac­Conkey media. After overnight incubation,
organism to the uroepithelial cells. The E. coli colonies are emulsified in saline on a slide
receptor is part of the P blood group antigen and are examined by slide agglutination with
and therefore the fim­briae have been termed polyvalent antisera belonging to EPEC-associated
P fimbriae. serogroups. Bacteria agglutinated by these sera
E. coli that cause UTI often originate in the gut are then identified with monovalent antisera to
of the patient. The bacteria may gain access to the individual serogroups. At least ten colonies per
urinary tract by the ascending or the hematogenous plate should be tested as many serotypes are
route. The ascending route of infection is belie­ved present in a single culture.
to be usual one. The bacteria from the fecal flora When the outbreak is caused by a strain with
spread to the perineum and from there they ascend some readily demonstrable feature such as failure
into the bladder. to fer­ment sorbitol, rapid identification is possible
UTI occurs more often in females than in by using appropriate culture media.
males. This is due to short urethra, pregnancy,
infrequent voiding and sexual intercourse which
b. Laboratory Diagnosis of ETEC
may lead to ‘honeymoon’ cystitis. About 5–7
percent of pregnant women have been reported Diagnosis of ETEC diarrhea depends on the dem­
to have urinary infection without any symp­toms. onstration of enterotoxin in E. coli isolates by any
(asymptomatic bacteriuria). of the methods listed in Table 36.5.

www.ebook3000.com
Chap-36.indd 261 15-03-2016 11:15:19
262  |  Section 3: Systemic Bacteriology
Table 36.5:  Methods for detection of ETEC enterotoxins
Assay LT ST
In vivo tests
Ligated rabbit ileal loop
Read at 6 hours ± +
Read at 18 hours + −
Infant rabbit bowel + +
Infant mouse intragastric (4 hours) − +
Adult rabbit skin (vascular permeability factor) + −
In vitro tests
i. Tissue culture tests
ii. Rounding of Y1 mouse adrenal cells
iii. Elongation of Chinese hamster ovary (CHO) cells + −
iv. Serological tests
ELISA + (ST-ELISA with monocolonial antibody)
Passive agglutination tests, passive immune hemolysis, + −
precipitin (Biken’s) test
v. Genetic tests
DNA probes + +

c. Laboratory Diagnosis of EIEC Laboratory Diagnosis of UTI


i. Sereny test: For laboratory diagnosis of EIEC, A. Collection of Specimen
the Sereny test used to be employed (that is, i. Catheter specimen
instillation of a suspension of fresh­ly isolated ii. Midstream urine specimen
EIEC or Shigella into the eyes of guinea pigs iii. Suprapubic stab
leads to mucopurulent conjunctivitis and i. Catheter specimen: Normal urine is sterile,
severe keratitis). but during voiding may become contaminated
ii. Tissue culture and DNA hybridization with genital commensals. In order to avoid
methods: Cell penetration of HeLa or HEP-2 such contamination urine used to be collected
cells in culture is a more humane diagnostic test. by catheterization for culture. Catheterization
iii. ELISA (VMA ELISA) test: The plasmid codes for this purpose is no longer considered
for outer membrane antigens called the justifiable because even under ideal conditions,
virulence marker antigens (VMA) which can catheterization leads to urinary infection in at
be detected by the ELISA (VMA ELISA) test. least two per cent and when precautions are
inadequate, the risk is much higher.
d. Laboratory Diagnosis of VTEC ii. Midstream urine specimen: In, male patients,
i. Demonstration of the bacilli or VT in feces: retract the prepuce and clean the glans penis
Laboratory diagnosis of VTEC diarrhea can with wet cotton. In case of female, anogenital
be made by demonstration of the bacilli or toilet is more important and should consist of
VT in feces directly or in culture. Most strains careful cleaning with soap and water. Separate
of 0157 VTEC produce colorless colonies after labia majora with fingers of one hand and
overnight incubation and these can be tested collect midstream urine in a sterile wide-
with an O157 LPS-specific antiserum in a mouthed container. The first portion of urine
simple agglutination assay. that flushes out commensal bacteria from the
ii. Sorbitol MacConkey medium: Most VTEC anterior urethra is discarded. The next portion
strains belong to the serotype O157 H7 does of the urine (midstream sample) is collected
not ferment sorbitol. directly into a sterile wide-mouthed container
iii. Confirmation: Toxigenicity is confirmed by and transported to the laboratory.
gene probes, by PCR, by testing strains for a iii. Suprapubic stab: In children and young infants,
cytotoxic effect on Vero cells or by a specific urine may be aspirated from the bladder.
ELISA.
iv. Serology: Demonstration of VT neutralizing B. Transport
antibodies in convalescent sera may help in Urine is a good medium for the growth of coliforms
retrospective diagnosis. and other urinary pathogens. If delay of more than

Chap-36.indd 262 15-03-2016 11:15:19


Chapter 36: Enterobacteriaceae: Escherichia, Klebsiella, Proteus and Other Genera  | 263
1–2 hours is unavoidable, the specimen should be Identification: The isolate is identified by colony
refrigerated at 4°C, morphology, staining, motility and biochemical
C. Microscopy of urine: The deposit of the centrifuged reactions.
urine can be examined under microscope to find out
the presence of pus cells, red blood cells and bacteria EDWARDSIELLA
in it. Genus Edwardsiella is separated from Escherichia
D. Semiquantitative culture: For quantitative by its ability to produce hydrogen sulfide in triple
culture, serial tenfold dilutions of urine are tested sugar iron agar. The genus contains the species
by the pour plate or surface culture methods. This, Edwardsiella tarda. The name tarda refers to slow
however, is too complicated for rou­tine diagnostic or weak fermentation of sugars by the organism. It
work, for which semiquantitative tech­niques are is the only recognized human pathogen.
more convenient. It is a gram-negative bacillus, motile, non-
Standard loop method capsulated.
The most widely used technique employs a It ferments only glucose and maltose with weak
standard loop.Measured quantity of urine with the fermentative powers. It is indole and MR positive,
help of standarized loop is inoculated on blood and VP and citrate negative (I MVi C + + – –). It
agar and another loopful on MacConkey agar and produces H2S, decarboxylates lysine and ornithine.
incubated overnight at 37°C. Blood agar medium Clinical infection: E. tarda is a normal intestinal
gives a quantitative meas­urement of bacteriuria, inhabitant of snakes and other cold-blooded animals.
while MacConkey agar enables a presumptive It has been cultured from normal and diarrheaic
diagnosis of the bacterium. human feces. It mainly causes wound infection, but
Other methods of semiquantitative estimation meningitis and septicaemia have also been reported.
of bacterial counts are filter paper method, dip
spoon, dip slide methods, etc. CITROBACTER
Interpretation of results Members of the genus Citrobacter are motile
Kass (1956) gave a criterion for active bacterial enterobacteria confused with both Escherichia
infection of urinary tract as follows: and Salmonella. They are motile, indole positive
Significant bacteriuria: When bacterial count is or negative, MR positive, VP negative, citrate
more than 105/mL of a single species. positive (I, M Vi C – + – +), urease weak positive
Doubtful significance: Between 104 to 105 bacteria and may or may not ferment lactose but they nearly
per mL. Specimen should be repeated for culture. always produce β-galactosidase (ONPG positive).
They do not decarboxylate lysine but most strains
No significant growth: <103 bacteria per mL and decarboxylate ornithine.
are regarded as contaminant.
Species: Three species are recognised, Citro. freundii
E. Identification: The organisms are identified
which gives typical reactions and Citro. koseri
by colony characters, Gram’s staining, motility,
(formerly Citro. diversus) and Citro. amalonaticus
biochemical reactions and slide agglutination test.
which do not form H2S (Table 36.5).
F. Antibiotic sensitivity test: E. coli and other
Ballerup-Bethesda group: The genus Citrobacter
common urinary pathogens develop drug
was first proposed for a group of lactose-negative or
resistance that no antibacterial therapy can be
late lactose-fermenting coliform bacteria that share
instituted meaningfully without testing individual
certain somatic antigens with salmonellae and
strains. Re­sistance is often to multiple drugs and is
were also known as the Ballerup–Bethesda group.
of the trans­ferable variety. Antibiotic sensitivity is
These organisms are now known as Citrobacter
necessary to administer proper drugs.
freundii. They exhibit extensive antigenic sharing
INFECTIONS with salmo­nellae and may cause confusion in the
diagnostic laboratory. Some strains (for exmaple,
Laboratory Diagnosis of Septicemia the Bhatnagar strain) have a Vi antigen serologically
Diagnosis depends on the isolation of the organism identical to the antigen of S. typhi and S. paratyphi
by blood culture and its identification by colony C. These may be used for the estimation of Vi
morphology, staining, motility and biochemical antibodies or for raising Vi antisera. However,
reactions. they can be distinguished by their negative lysine
decarboxylase and positive KCN reactions.
Laboratory Diagnosis of Pyogenic Infection
Clinical infection: Citrobacter may cause infections
Specimens: The specimens are usually pus and of the urinary tract, gallbladder, middle ear and
wound swab. meninges. C. koseri occasionally causes neonatal
Culture: Cultures are made on MacConkey’s agar. meningitis.

www.ebook3000.com
Chap-36.indd 263 15-03-2016 11:15:19
264  |  Section 3: Systemic Bacteriology
KLEBSIELLA Table 36.6  Distinguishing reactions of Citrobacter
Introduction Species Test/substratea
ind mal H2S KCN adon arbtl mel
Members of the genus Klebsiella are gram-negative,
nonsporing, non-motile bacilli that grow well on C. freundii − − ± + − − ±
ordinary media, produce pink mucoid colonies C. koseri + + − − + + −
on MacConkey’s agar. They are usually found in C. + − − + − − −
the intestinal tract of humans and animals or free- amalonaticus
living in soil, water, and on plants. a
Ind, indole production; mal, utilization of malonate; H2S,
H2S produced in TSI agar; KCN, growth in KCN medium;
Classification adon, arbtl, mel, fermentation of adonitol, D-arabitol,
melibiose.
Their classification has undergone var­ious
modifications. The name K. pneumoniae is used
for the species as a whole. It is further divided into 1. Capsular (K) antigen: On the basis of capsular (K)
4 subspecies. The most frequently encountered, antigens, the klebsiellae have been differentiated,
biochemically typical form of it is known as K. into 80 serotypes. Members of capsular types 1–6
pneumoniae subsp. aerogenes, K. pneumoniae occur most frequently in the human respiratory
subsp. ozaenae, K. pneumoniae subsp. pneumoniae tract.
K. pneumoniae subsp. rhinoscleromatis (Table Capsular antigens are usually detected by means
36.6). Indole-producing strains that resemble of the capsular ‘swelling’ reaction, countercurrent
K. pneumoniae subsp. aerogenes biochemically are immunoelectrophoresis and enzyme-linked
classified in a separate species, K. oxytoca. immunosorbent-assay (ELISA).
2. Somatic (O) antigen: Five different somatic or O
Morphology antigens (01–05) occur in various combinations
They are short,plump, gram-negative, non-sporing, with the capsular antigens. Four of the five
capsulated, non-motile bacilli, 1–2 µm long and Klebsiella O antigens are identical to or related
0.5–0.8 µm wide with parallel or bulging sides and to E. coli O antigens.
slightly pointed or rounded ends.
Typing Methods
Cultural Characteristics 1. Bacteriocin (Klebocin or pneumocin) typing;
Klebsiellae grow well on ordinary media at a 2. Phage typing; 3. Biotyping; 4. Resistotyping; 5.
temperatures between 12°C and 43°C (optimum, Molecular typing methods: Plasmid analysis,
37°C) in 18–24 hours. On MacConkey agar, the DNA profiling by random amplified polymorphic
colonies typically appear large, mucoid and red in DNA (RAPD) and pulsed-field gel electrophoresis.
colour. Mucoid nature of colonies is due to capsular
Pathogenicity: Klebsiella pneumoniae can cause
material produced by the organism.
a primary community-acquired pnuemonia,
nosocomial infections, urinary tract infections,
Biochemical Reactions
wound infections, bacteremia and meningitis and
They ferment sugars (glucose, lactose, sucrose, rarely diarrhea. In some hospital, K. pneumoniae
mannitol) with production of acid and gas. They has replaced E. coli as the leading blood culture
are urease positive, indole negative, MR negative, isolate. As most strains are resistant to antibiotics,
VP positive and citrate positive (IMViC – – ++). treatment poses serious problems.
These reactions are typical of K. pneumoniae subsp.
aerogenes. Pneumonia: Klebsiella pneumonia is a serious
disease with high case fatality. The typical patient
Glucose Lactose Sucrose Mannitol is a middle- or older-aged man who have medical
+ (AG) + + +
problems. Positive blood cultures can be obtained
in about 25% of the cases.
Urease Indole MR VP Citrate
+ – – + + Diarrhea: Some strains of K. pneumoniae isolated
from cas­es of diarrhea have been shown to produce
Biochemical reactions of different subspecies of K. an entero­toxin very similar to the heat stable toxin
pneumoniae and K. oxytoca are given in Table 36.6. of E. coli.
K. ozaenae is a bacillus associated with ozena,
Antigenic Structure an uncommon, chronic disease in which there is
Klebsiella possess capsular (K) and somatic(O) atrophy of the nasal mucosa characterized by foul
antigens. smelling nasal discharge.

Chap-36.indd 264 15-03-2016 11:15:19


Chapter 36: Enterobacteriaceae: Escherichia, Klebsiella, Proteus and Other Genera  | 265
Table 36.7  Distinguishing reactions of Klebsiella species
Tests K. pneumoniae subspecies K oxytoca
aerogenes pneumoniae ozaenae rhinoscleromatis
Gas from glucose + + V – +
Acid from lactose + + V – +
Urease + + V – +
Citrate + + V – +
Malonate + + – + +
MR – + + + V
VP + – – – V
Lysine + + V – +
decarboxylase
KCN + + + ± +
V, variable

K. rhinoscleromatis causes rhinoscleroma, a urine, pus and other pathological materials. They
chronic granulomatous hypertrophy of the nose. may cause urinary tract infections and hospital
K. oxytoca may be rarely isolated from clinical infections. They are occasionally associated with
specimens (Table 36.7). meningitis and septicemia.
Treatment: Aminoglycosides are often effective in
Laboratory Diagnosis the treatment of Enterobacter infections.
Diagnosis is made by culturing appropriate spec­
imens on blood agar and Mac Conkey agar and HAFNIA
identifying the isolate by biochemical reactions.
Antibiotic sensitivity should invariably be done. These organisms are probably best regarded
Many strains carry plasmids determining multiple as non-lactose fermenter (NLF) member of
drug resistance. genus Enterobacter. This is a motile, nonlactose-
fermenting bacillus which is indole and MR
Treatment negative and VP and citrate positive. Biochemical
They are normally susceptible to cephaloporins, reactions are evident best at 22°C but at 37°C they
especially β-lactamase stable derivatives may be negative or irregular. Hafnia alvei is the
such as cefuroxime and cefotaxime, and to only species.
fluoroquinolones. They are often sensitive to Strains are isolated from feces of man and
gentamicin and other aminoglycosides., but other animals and are also found in sewage, soil,
transferable enzymic resistance to aminoglycosides water and dairy products. They are occasionally
and other antimicrobial agents has become encountered as opportunistic pathogens that has
common in strains found in some hospitals. been recovered from infected wounds, abscesses,
Klebsiella infection of the urine often responds sputum urine, blood and other sites.
to trimethoprim, nitrofurantoin, co-amoxiclav
or oral cephalosporins. Pneumonia and other SERRATIA
serious infections require vigorous treatment S. marcescens is the one most commonly
with aminoglycoside or a cephalosporin, such as encountered species in clinical specimens. It is
cefotaxime. small, motile, gram-negative bacillus. This differs
from Hafnia in forming a pink, red or magenta,
ENTEROBACTER nondiffusible pigment called prodigiosin which
Enterobacter is a motile, capsulated, lactose is formed optimally at room temperature.
fermenting bacillus which is indole and MR negative
and VP and citrate positive (IMViC – - + +). These Pathogenesis
characteristics are similar to those of Klebsiella but It is a saprophyte found in water, soil and food.
can be differentiated from Klebsiella because it is It may grow in sputum after collection and may
motile and ornithine positive. Two clinically relevant suggest hemoptysis because of the pigment formed
species are E. cloacae and E. aerogenes. (pseudo­hemoptysis).Nosocomial infections due to
Clinical Infections: They are normally found S. marc­escens are being reported with increasing
in feces, sewage, soil and water and rarely in frequency. The bacillus has been associated with

www.ebook3000.com
Chap-36.indd 265 15-03-2016 11:15:19
266  |  Section 3: Systemic Bacteriology
infections of the urinary and respiratory tracts, 5. Write short notes on:
meningitis, wound infections, septicemia and a. Antigenic structure of Escherichia coli
endocarditis. Multiple drug resistance is common b. Enterotoxins of Escherichia coli
in hospital strains. c. Verotoxin (VT) or Shiga-like toxin (SLT)
6. Write briefly about:
a. Citrobacter
Key Points
b. Klebsiella pneumoniae
Escherichia coli c. What are the properties which differentiate E.
™™ Gram-negative bacilli aerobe and a faculta­t ive coli from Klebsiella
anaerobe. On MacConkey’s medium, colonies are d. Enterobacter
red or pink due (lactose fermenter). Selective media e. Serratia
such as DCA or SS agar growth
™™ B. Toxins: 1. Exotoxins: Hemolysins and enterotoxins.
™™ Enterotoxins: (e.g. heat-stabile and heat-labile MULTIPLE CHOICE QUESTIONS (MCQs)
enterotoxins, and Verotoxin (VT) also known as
1. Which of the following properties is/are shown
Shiga-like toxin (SLT)-
by the organisms belonging to the family Enter­
™™ Clinical infections: 1. Urinary tract infection; 2.
Diarrhea, 3. Pyogenic infections, 4. Bacteremia obacteriaceae?
™™ Diagnosis: Organisms grow rapidly on most culture a. They are catalase positive
media b. They are oxidase negative
™™ Edwardsiella: Edwardsiella tarda mainly causes c. They ferment glucose
wound infection, but meningitis and septicemia d. All of the above
have also been reported 2. Which of the following bacteria is/are known as
™™ Citrobacter: It may cause infections of the urinary coliform bacilli?
tract, gallbladder, middle ear and meninges. C. koseri a. Escherichia b. Klebsiella
occasionally causes neonatal meningitis
c. Enterobacter d. All of the above
™™ Klebsiella: Members of the genus Klebsiella are
gram-negative, nonsporing, nonmotile bacilli that 3. Vero cytotoxin of Escherichia coli is similar to:
grow well on ordinary media, produce pink mucoid a. Shiga toxin
colonies on MacConkey’s agar b. Cholera toxin
™™ The name K. pneumoniae is used for the species as a c. Heat-labile enterotoxin of Escherichia coli
whole. It is further divided into 4 subspecies d. Heat-stable enterotoxin of Escherichia coli
™™ Klebsiella pneumoniae can cause a primary 4. Traveler’s diarrhea is caused by
communiy-acquired pnuemonia, nosocomial a. Enteropathogenic Escherichia coli:
infections, urinary tract infections, wound infections,
b. Enterotoxigenic Escherichia coli
bacteremia and meningitis and rarely diarrhea
™™ K. ozaenae- is associated with ozena
c. Enteroinvasive Escherichia coli
™™ K. rhinoscleromatis causes rhinoscleroma, a chronic d. Verotoxigenic Escherichia coli
granulomatous hypertrophy of the nose 5. Haemolytic uraemic syndrome is caused by:
™™ K. oxytoca may be rarely isolated from clinical a. Enteropathogenic E. coli
specimens b. Enterotoxigenic E. coli
™™ Enterobacter: They may cause urinary tract c. Enteroinvasive E. coli
infections and hospital infections, occasionally d. Vero cytotoxin producing E. coli:
associated with meningitis and septicemia
6. Sereny test is used for detection of
™™ Hafnia: They are has been recovered from infected
wounds, abscesses, sputum urine, blood and other
a. Enteropathogenic Escherichia coli
sites b. Enterotoxigenic Escherichia coli
™™ Serratia: S. marcescens is associated with infections c. Enteroinvasive Escherichia coli
of the urinary and respiratory tracts, meningitis, d. Verotoxigenic Escherichia coli
wound infections, septicemia and endocarditis. 7. Prodigiosin (red pigment) is produced by members
of genus:
a. Serratia b. Enterobacter
IMPORTANT QUESTIONS c. Hafnia d. Citrobacter
8. Which of the following species/subspecies of
1. Discuss various mechanisms by which Escheri­chia Klebsiella produces indole?
coli produces diarrhea. Describe the laboratory a. K. pneumoniae subspecies aerogenes
diagnosis of bacterial diarrheas. b. K. oxytoca
2. Discuss the pathogenicity of Escherichia coli. c. K. pneumoniae subspecies pneumoniae
3. Discuss the laboratory diagnosis of various d. K. pneumoniae subspecies rhinoscleromatis
infections caused by Escherichia coli.
4. Discuss the pathogenesis and laboratory diagnosis ANSWERS (MCQs)
of urinary tract infections caused by Escheri­chia coli. 1. d; 2. d; 3. a; 4. b; 5. d; 6. c; 7. a; 8. b

Chap-36.indd 266 15-03-2016 11:15:19


37
Chapter
Tribe Proteeae: Proteus,
Morganella and Providencia

Learning Objectives

After reading and studying this chapter, you should ∙∙ List various differences among Proteus, Morganella
be able to: and Providentia.
∙∙ Describe morphology, culture characteristics and
biochemical reactions of Proteus sp

Classification name ‘Proteus’ refers to their pleomorphism, after


the Greek god Proteus who could assume any shape.
The tribe Proteeae is classified into three genera
Genus Proteus has four species: P. mirabilis,
­Proteus, Morganella and Providencia. Most of
P. vulgaris, P. myxofaciens and P. penneri. P.
them except for some Providencia strains, produce
mirabilis, P. vulgaris are widely recognized as
a pow­erful urease which rapidly hydrolyses urea
human pathogens. These are motile, gram-negative
to ammonia and carbon dioxide. A characteristic
bacilli, characterized by swarming growth on agar.
feature which dis­tinguishes tribe Proteeae from
other enterobacteria is the presence, in all Morphology
members of the tribe, of the enzyme phenyl alanine
They are gram-negative coccobacilli, 1–3 mm long and
deaminase which converts phenyl alanine to
0.6 mm wide. Pleomorphism is frequent—short
phenyl pyruvic acid (PPA reaction). All members
coccobacilli to long filaments. In young swarming
of this tribe fail to ferment lactose.
cultures, many of the bacteria are long, curved and
The major differentiating features of medically
filamentous. They may be arranged singly, in pairs
important species of proteus bacilli are shown in
or in short chains. They are actively motile with
Table 37.1.
peritrichous flagella. They also have more type of
Proteus fimbriae and are noncapsulated.

Proteus Bacilli Cultural Characteristics


Proteus bacilli are nor­mal intestinal commensals They are aerobe and facultative anaerobes. All grow
and opportunistic pathogens like coliforms. The well on laboratory nutrient media. Proteus organisms

Table 37.1  Biochemcal features of species of Proteus, Morganella and Providencia


Test Pr. vulgaris Pr. mirabilis Morg. morganii Prov. alcalifaciens Prov. stuarti Prov. rettgeri
Swarming + + – – – –
Gas from glucose + + + + – –
Indole + – + + + +
Phenyl pyruvic acid (PPA) test + + + + + +
Urease + + + – ± +
H2S production + + – – – –
Ornithine decarboxylase – + + – – –
Fermentation of adonitol – – – + ± ±
Fermentation of trehalose ± + ± – + –

www.ebook3000.com
268  |  Section 3: Systemic Bacteriology
are usually first recognized by their characteristic interest. Weil and Felix (1916) studying Proteus
pu­trefactive odor described as ‘fishy’ or ‘seminal’ bacilli ob­served that flagellated strains growing
and swarming appearance on noninhibitory on agar formed a thin surface film resembling the
solid media, such as nutrient agar and blood agar. mist produced by breathing on glass and named
Swarming is a striking feature of Pr. mirabilis and Pr. this variety the ‘Hauch’ form (from Hauch, meaning
vulgaris. Swarming of Proteus appears to be due to film of breath). Nonflag­ellated variants grew as
vigorous motility of the organisim although the exact isolated colonies without the surface film and
cause is not yet established. were called ‘Ohne Hauch’ (meaning without film
Swarming growth is a problem in the laboratory of breath). These names came to be ab­breviated
when mixed growth is obtained in which Pro­ as the Hand 0 forms. Subsequently, the H and O
teus bacilli are present with other bacteria. A were extended to refer to the flagellar and so­matic
number of methods have been devised to inhibit antigens of other bacilli as well.
swarming. Swarming of Proteus can be inhibited Weil and Felix also observed that certain
by (i) increasing concentration of agar (6%) and nonmotile strains of Pr. vulgaris, called the ‘X
by (ii) incorporation of chloral hy­drale (1 : 500), strains’, were agglutinated by sera from typhus fever
sodium azide (1:500), alcohol (5–6%), sulfonamide, patients. This heterophilic agglutination due to the
surface. active agents or boric acid (1:1000). sharing of an alkali stable carbo­hydrate antigen
Swarming does not occur on MacConkey’s by certain strains of Proteus (OX2, OX19 and OXK)
medium, on which smooth colorless (NLF) formed. and rickett­siae forms the basis of the Weil-Felix
Proteus produces uniform turbidity with a slight reaction for the diagnosis of some rickettsial
powdery deposit and an ammonical odor in liquid infections. Three nonmo­tile Proteus strains OX2,
medium (peptone water). OX19 and OXK are used in the agglutination test.
Dienes phenomenon: When two identical strains OX19, OX2 are the strains of P. vulgaris serotype 01
of Proteus are inoculated at different places of the and serotype 02 and OXK is the strain of P. mirabilis
same culture plate, the resulting swarms of growth serotype 03.
coalesce and no line is formed between swarming Typing methods: Phage typing, bacteriocin
culture of the same strain. When, however, two (proticin) typing and serotyping schemes have been
different strains of Proteus species are inoculated, developed for Proteus and Providencia species.
the spreading films of growth fail to coalesce and Swarming Proteus strains exhibit the Dienes
remain separated by a narrow but easily visib1e phenomenon and this forms the basis for a precise
furrow. This is known as Dienes phenomenon. method of differentiation among such strains.
It has been used to determine the identity or
non-identity of various strains of Proteus. Pathogenesis
Proteus bacilli are widely distributed in nature as
Biochemical Reactions saprophytes, being found in decomposing animal
The distinctive characters of this genus are: mat­ter, in sewage, in manured soil and in human and
i. PPA test—Deamination of phenyl alanine ani­mal feces. They are frequently present on the moist
to phenyl pyruvic acid (PPA test) is always areas of the skin. They are opportunistic pathogens,
positive. commonly responsible for urinary and septic infec­
ii. Urea hydrolysis—Urea hydrolysis by enzyme tions, often nosocomial.
urease is another characteristic of Proteus, but P. mirabilis accounts for the majority of human
is negative in some Providencia strains. infections seen with this group of organisms. All
iii. All species of Proteus produce acid from members of the tribe can cause urinary tract
glucose. infections (UTI), wound infections, pneumonia,
iv. Lactose is not fermented. infection of the ear, respiratory tract infection,
v. They are malonate utilization negative. septicemia and nosocomial infections. Strains of
vi. Indole is formed by Pr. vulgaris but is negative Pr. mirabilis are a prominent cause of urinary tract
in Pr. mirabilis. infection in children and in domiciliary practice.
vii. They are MR positive and VP negative. UTI caused by Proteus tends to be more serious
viii. H2S is produced by Pr. vulgaris and Pr. mirabilis. than that caused by E. coli and other coliforms.
ix. Nitrate reduction positive. It produces urease which splits urea into carbon
Other biochemical characters of species of dioxide and ammonia. Ammonia inactivates
Proteus are given in Table 37.1. complement, damages renal epithelium and
makes the urine alkaline.This increase in pH causes
Antigenic Structure precipitation of calcium and magnesium salts from
Proteus bacilli possess somatic O and flagellar the urine and results in the formation of urinary
H antigens, which are of considerable historical calculi.
Chapter 37: Tribe Proteeae: Proteus, Morganella and Providencia  | 269
Labortory Diagnosis isolated from respiratory and urinary infections in
Culture: Laboratory diagnosis of the infections predisposed or hospitalized patients.
caused by species Proteus can be carried out by
culture of the specimen on MacConkey agar or Key Points
DCA. ™™ The tribe Proteeae is classified into three genera
­Proteus, Morganella and Providencia
Identification: The isolate is identified by its
™™ Genus Proteus has four species: P. mirabilis, P. vulgaris,
morphological, biochemical and agglutination P. myxofaciens and P. penneri
reactions. ™™ Cultural characteristics: Proteus organisms are
usually first recognized by their characteristic pu­
Treatment trefactive odor described as ‘fishy’ or ‘seminal’ and
swarming appearance on noninhibitory solid media
Proteus bacilli are resistant to many of the common such as nutrient agar and blood agar
antibiotics. An exception is P. mirabilis which is ™™ Pathogenesis: All members of the tribe can cause
sensitive to ampicillin and cephalosporins. urinary tract infections (UTI), wound infections,
pneumonia
™™ Morganella morganii causes urinary infection infre­
Morganella quently. Nosocomial wound infections also occur
™™ Five Providencia species may be recovered from feces
Morganella morganii is the only species in the
™™ Erwinia: E. herbicola has occasionally been
genus Morganella. It is motile and lactose-non- isolated from respiratory and urinary infections in
fermenting, and do not swarm on solid media. predisposed or hospitalized patients.
M. morganii is commonly found in human
and animal feces and causes urinary infection
infrequently. Nosocomial wound infections also Important question
occur. Write short notes on:
a. Classification of tribe Proteeae
Providencia b. Pathogenicity of Proteus species
c. Swarming growth
Five Providencia species include P. alcalifaciens, d. Dienes phenomenon
P. rettgeri, P. rustigianii and P. stuartii and P. e. Weil-Felix rection
heimbachae.All species may be recovered from f. Genus Morganella
feces. However, only P. alcalifaciens may be g. Genus Providencia.
associated with diarrhea. P. rettgeri and P. stuartii
have been associated with hospital-acquired Multiple choice questions (MCQs)
urinary tract, wound and other infections, 1. The bacterium that shows swarming on blood agar
particularly in immunocompromised patients. is:
P. stuartii may occasionally cause hospital- a. Proteus stuartii
associated urinary tract infection, infection of b. Proteus mirabilis
c. Providencia rettgeri
burns and pneumonia and septicemia in elderly
d. Morganella morganii
and immunodeficient patients. 2. Phenylalanine deaminase test is positive in:
P. rettgeri is part of the normal fecal flora of a. Proteus b. Morganella
reptiles and amphibians, and sometimes causes c. Providencia d. All of the above
nosocomial infection of the urinary tract, wounds, 3. Diene’s phenomenon is used to detect identity of
burns and blood. strains of:
a. Klebsiella b. Salmonella
c. Shigella d. Proteus
Laboratory Diagnosis
4. Swarming of Proteus strains on solid media can be
Culture: Laboratory diagnosis of the infections inhibited by:
caused by species Proteus, Morganella and a. Increasing the concentration of agar in the me-
Providencia can be carried out by culture of the dium
specimen on MacConkey agar or DCA. b. Incorporation of chloral hydrate in the medium
c. Incorporation of sodium azide in the medium
Identification: The isolate is identified by its d. All of the above methods
morphological, biochemical and agglutination 5. Which of the following Proteus strains is/are used
reactions. in Weil-Felix reaction?
a. Proteus vulgaris OX2
b. Proteus vulgaris OX19
Erwinia c. Proteus mirabilis OXK
These are anaerogenic bacilli forming a yellowish d. All of the above
pigment, usually found in soil and causing plant Answers (MCQs)
infections. E. herbicola has occasionally been 1. b; 2. d; 3. d; 4. d; 5. d.

www.ebook3000.com
38
Chapter

Shigella

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Discuss the antigenic structure and toxins of
be able to: Shigella
∙∙ Describe morphology and various culture media ∙∙ Discuss laboratory diagnosis of bacillary dysentery.
for the isolation of Shigella

INTRODUCTION 5. Salmonella–Shigella (SS) agar: Salmonella–


Shigella (SS) agar is a highly sel­ective medium
The genus Shigella is named after the Japanese
for the isolation of Salmonella and Shigella.
microbiologist Kiyoshi Shiga, who first isolated the
Colonies of Shigella on this medium are
organism in 1896. It was then called S. shigae and is
colorless with no blackening.
now known as S. dysenter­iae serotype 1. S. flexneri
6. Hektoen enteric (HE) agar: Hektoen enteric
by Flexner (1900), S. sonnei by Sonne (1915)and S.
(HE) agar is useful as a direct plating medium for
boydii Boyd (1931) were subsequently described.
fecal specimens for the isolation of Shigella and
Morphology Salmonella. Colonies of Shigella on this medium
are green while those of Sal­monella are blue green
Shigellae are nonsporing, non­capsulate, Gram-
with black centers due to H2S production.
negative rods, 2–4 × 0.6 µm, nonmotile and non­
7. Peptone water and nutrient broth: Good
flagellate.
growth with uniform turbidity on incubation
Cultural Characteristics overnight at 37°C.
They are aerobes and facul­tative anaerobes, with a 8. Enrichment broths
growth temperature range of 10–40 °C and optima a. Selenite F broth-Selenite F broth will grow
of 37°C and pH 7.4. They grow well on conventional and enrich S. sonnei and S. flexneri serotype
media. 6, but is inhibitory to other shigellae.
1. Nutrient agar and blood agar: Colonies are b. Gram-negative (GN) broth: Most strains
smooth, grayish or colorless, translucent, of Shigella grow well.
often 2–3 mm in diameter, circular, convex,
Resistance
resembling those of salmonellae.
2. MacConkey agar: Colonies are colorless, Shigellae are killed by moist heat at 55°C in 1 hour
i.e.pale and yellowish (non-lactose-fermenting) and fairly readily by strong disinfectants, e.g. by
due to the absence of lactose fermentation. 1% phenol in 15 min. S. sonnei is more resistant to
An exception is S. sonnei, a late fermenter adverse environmental conditions as compared
of lactose, become pink when incubation is to other species.
prolonged beyond 24 hours.
3. Deoxycholate citrate agar (DCA): Deoxycholate Biochemical Reactions
citrate agar (DCA) is an excellent selective The shigellae are divided into four groups, or
plating medium. Colonies are pale and similar species, by their biochemical reactions and
to, though usually slightly smaller, and more antigenic structure (Table 38.1 ). The groups A, B,
translucent than those of salmonellae. C and D correspond to the species S. dysenteriae,
4. Xylose lysine deoxycholate (XLD): XLD is S. flexneri, S. boydii and S. sonnei.
probably the best selective medium for shigellae. Glucose is fermented with the production
Colonies are red, without black centers. of acid, without gas, except for two serotypes, S.

Chap-38.indd 270 15-03-2016 11:16:01


Chapter 38: Shigella  | 271
flexneri serotype 6 and S. boydii serotype 14, which Large-Sachs group. Three further serotypes have
form gas. Fermentation of mannitol is of importance been described making a total of ten.
in classification and shigellae have tradition­ Exotoxin: S. dysenteriae type 1 produces a powerful
ally been divided into mannitol fermenting and exotoxin (Shiga toxin). It acts as enterotoxin as
nonfermenting species. S. flexneri, S. boydii and S. well as neurotoxin. As enterotoxin, it acts on the
sonnei ferment mannitol, while S. dysenteriae does intes­tinal mucosa causing transudation of fluid in
not. Lactose and sucrose are not fermented, except the lumen. As neurotoxin, it damages endothelial
by S. sonnei which ferments them late. Dulcitol is cells of small blood vessels of the central nervous
not fermented by the majority of shigellae. system which results in neurological complications
S. dysenteriae serotype 1, S. flexneri serotype 6 like polyneuritis, coma and meningism.
and S. sonnei are always indole negative. Shigellae Cytotoxin: S. dysenteriae produces also an cytotoxin
are MR positive and negative for Voges Proskauer which is active on vero cells and is known as
reaction, citrate and malonate utilization. They Verotoxin. This appears to be the same as Verotoxin
reduce nitrates to nitrites, do not form H2S and are 1 (or Shiga-like toxin) produced by certain strains of
inhibited by KCN. Catalase is produced, except by E. coli (VTEC).
Sh. dysenteriae type 1 which are catalase negative.
Members of groups A, B and C fail to decarbox­ Group B (Shigella flexneri)
ylate lysine and ornithine. S. sonnei decarboxylates This group is named af­ter Flexner, who described
ornithine but not lysine. the first of the mannitol fermenting shigellae from
Philippines (1900).
Antigenic Structure S. flexneri is biochemically heterogeneous and
The shigellae are divided into four ‘major’ O antigenic antigeni­cally the most complex among shigellae.
groups, designated A, B, C, and D. In addition to the Based on type specific and group specific antigens,
major O antigens, groups A, B and C contain ‘minor’ they have been classified into six serotypes (1-6) and
O antigens, which allow for subgrouping. Some several subtypes (1a; 1b; 2a, 2b; 3a, 3b, 3c, 4a, 4b, 5a,
strains possess K or envelope antigens. 5b). In addition, two antigenic ‘variants’ called X and Y
are recognized, which have lost their type antigens and
Classification are distinguished by their different group antigens.
Shigellae are classified into four species or sub­ Serotype 6 is always indole negative and occurs in
groups (A,B,C,D) based on a combination of three biotypes: Boyd 88, Manchester and Newcastle,
biochemical and serological characteristics. some of which form gas from sugars (Table 38.2).
Serotypes are distin­guished within the species.
S. sonnei is serological­ly homogeneous and is Group C (Sh. boydii)
classified by colicin typing (Table 38.1). The group is named after Boyd, who first described
these strains from India (1931). This group consists
Group A (S. dysenteriae) of dys­e ntery bacilli that resemble S. flexneri
This species of man­nitol nonfermenting bacilli biochemical­ly but not antigenically. Eighteen
consists of 12 serotypes. Serotypes 1 and 2 are different serotypes of S. boydii are recognized.
the organisms formerly called S. shigae and S.
schmitzii. S. shigae is indole negative and is the Group D (S. sonnei)
only member of the family that is always catalase This bacillus, first de­scribed by Sonne (1915) in
negative. S. dysenteriae type 2 (S. schmitzi) Denmark, ferments lac­tose and sucrose late. It is
forms in­dole and ferments sorbitol and rhamnose. indole negative. It is antigenically homo­geneous.
Serotypes 3–7 were described by Large and It may, however undergo an antigenic vari­ation
Sachs in India and hence used to be known as the and may occur in two forms, phase 1 and phase II.

Table 38.1  Distinguishing features of Shigella species


Subgroup A B C D
Species Sh. dysenteriae Sh. flexneri Sh. boydii Sh. sonnei
Mannitol – A A A
Lactose – – – A (Late)
Sucrose – – – A (Late)
Dulcitol – – d
Indole d d d
Ornithine decarboxylase – – – +
Serotypes 10 6 + variants 15 Only one
A, acid; d, delayed

www.ebook3000.com
Chap-38.indd 271 15-03-2016 11:16:01
272  |  Section 3: Systemic Bacteriology
Table 38.2  Biotypes of Sh. flexneri type 6 illness may also be caused by members of S. flexneri
and S. boydii groups. In contrast dysentery associated
Fermentation of
with S. sonnei (Sonne dysentery) in an otherwise
Glucose Mannitol healthy person may be confined to the passage of a
Boyd 88 A A few loose stools with vague abdonimal discomfort
Manchester AG AG and the patient often continues at school or work.
Newcastle A or AG − Complications: Complications are most often seen
A, acid; AG, acid and gas in patients with S. dysenteriae serotype 1 infection.
These include arthritis, toxic neuritis, conjunctivitis,
paro­t itis, and, in children, intussusception.
S. sonnei causes the mildest form of bacillary Hemolytic uremic syn­drome (HUS) may occur as
dysentery. In many cases the disease may only be a a complication in severe cases.
mild diarrhea. However, S. sonnei infection persists The severity of the disease may vary from acute
as the most common shigellosis in the advanced fulminating dysentery to mild diarrhea. As the term
countries. For epidemiological purposes, S. sonnei bacillary dysentery refers only to the more severe
has been classified into 26 colicin types. cases, the term ‘shigellosis’ has been employed
to include the whole spec­trum of disease caused
Pathogenic Mechanisms by shigellae.
1. Surface properties: Lipopolysaccharide (LPS)
has been implicated to entering intestinal cells. Epidemiology
2. Invasiveness: Invasive property is related to Bacillary dysentery has a global distribution. It is
the presence in the bacillus of large plasmids mostly associated with the overcrowding and bad
coding for the outer membrane protein hygienic conditions. Humans are the only known
responsible for cell penetration. These proteins reservoir of Shigella organisms. Transmission may
are called ‘virulence marker antigens’ (VMA). occur by direct person-to person contact, and spread
3. Toxins: S. dysenteriae type 1 forms a produces may take place via the fecal. oral route, with carriers
a powerful exotoxin (Shiga tox­in) (details see as the source. From the carries the organisms can
under heading group A (S. dysenteriae). be spread by flies, fingers, food and feces. Young
children in daycare centers, people living in crowded
Pathogenicity and less-than-adequate housing, and young male
Shigellae cause bacillary dysentery. Humans are homosexuals as part of the gay bowel syndrome who
the only known reservoir of Shigella organisms participate in anal-oral sex are most likely affected.
infection occurs by ingestion. The minimum Sh. sonnei is the predominant infecting agent.
infective dose is low, as few as 10–100 bacilli being S. sonnei is the main type in the north in the USA,
capable of initiating the disease. while S. flexneri is more common in the south. In
Shigella cause disease by invading and India, S. flexneri has been the predominant species,
replicating in cells lining the colonic mucosa. After having formed 50–85% of isolates in different series.
reaching the large intestine, the shigellae multiply S. dysenteriae (8–25%) and S. sonnei (2–24%) are the
in the gut lumen. The shigellae multiply within the next common species. S. boydii (0–8%) has been
epithelial cells and spread laterally into adjacent isolated least frequently.
cells, where cell-to-cell passage occurs, and deep
into the lamina propria. The infected epithelial Laboratory Diagnosis
cells are killed and the lamina propria and Diagnosis depends on isolating the bacillus from
submucosa develop an inflammatory reaction with feces.
capillary thrombosis. Patches of nerotic epithelium
are sloughed and ulcer form. The cellular response A. Specimens
is mainly by polymorphonu­clear leukocytes.
i. Feces: A specimen of feces is always preferable
The ulcers of the bacillary dysentery are much
to a rectal swab
shallower than amoebic ulcers. Bacteremia may
ii. Rectal swabs: A direct swab may be taken from
occur in severe infections.
the ulcer by sigmoidoscopic examination.
Bacillary dysentery has a short incubation
period (1–7 days, usually 48 hours). The main clinical
features are frequent passage of loose, scanty feces B. Transport
containing blood and mucus, along with abdominal Fresh feces should be inoculat­ed without delay or
cramps and tenesmus. Fever and vomiting may be transported in a suitable medium such as Sachs’
present. Infection is usually self-limited. buffered glycerol saline. pH 7.0–7.4, which prevents
In dysentery caused by S. dysenteria type 1, the dysentery bacilli from being destroyed by the
patients experience more severe symptoms. Severe acid produced during the growth of other organisms.

Chap-38.indd 272 15-03-2016 11:16:01


Chapter 38: Shigella  | 273
C. Microscopy Key Points
Make a wet film of a suspension of the feces in ™™ The genus Shigella gram-negative bacilli, non-motile.
saline. This will show numerous erythro­cytes and They are facultative anaerobe and grow well on
polymorphs and some macrophages. conventional media
™™ The shigellae are divided into four groups, or
D. Culture species. The groups A, B, C and D correspond to
the species S. dysenteriae, S. flexneri, S. boydii and
For inoculation it is best to use mucus flakes if they S. sonnei
are present in the sample. The feces are inoculated ™™ Shigella causes bacillary dysentery. Bacteria are
on Mac­C onkey agar and DCA. SS agar and acquired by fecal-oral spread. A severe form
XLD medium can also be used. After overnight of disease is caused by S. dysenteriae (bacterial
dysentery). Hemolytic colitis and hemolytic uremic
incubation at 37°C, the plates are inspected for pale syndrome (HUS) associated with Shigella
(non-lactose-fer­menting) colonies on MacConkey ™™ Diagnosis: Culture of feces reveals nonmotile bacillus
agar and DCA, and red and colorless colonies with that is urease, citrate, H2S and KCN negative and
no blackening on XLD and SS agar, respectively. should be further investigated by biochemical tests.

Identification IMPORTANT QUESTIONS


i. Biochemical reactions: These are tested
1. Discuss the antigenic structure and toxins of
for motility and biochemical reactions. Any
Shigella.
nonmotile bacillus that is urease, citrate, 2. Enumerate the various causes of bacillary dysentery.
H 2S and KCN negative should be further Discuss laboratory diagnosis of Shigella dysentery.
investigated by biochemical tests (Table 38.1). 3. Write short notes on:
ii. Slide agglutination: Identification is confirmed Classification of shigella
by slide agglutination with polyvalent and Pathogenesis of shigella.
monovalent sera and then with type-specific
sera (belonging to subgroups A, B, C) unless MULTIPLE CHOICE QUESTIONS (MCQs)
the strain is S. sonnei.
1. All the following media are employed for isolation
of Shigella except:
E. Colicine Typing a. Hektoen enteric agar
Plasmid pattern analysis and colicine typing may b. Salmonella-Shigella agar
help elucidate patterns of spread of S.sonnei. c. Thiosulfate citrate bile salt agar
d. Xylose lysine deoxycholate agar
2. Enrichment medium of choice for isolation of
F. Serology Shigella is:
Demonstration of antibodies in sera is not useful a. Alkaline peptone water
have no place in diagnosis of the disease. b. Selenite F broth
c. Taurocholate peptone enrichment medium
d. Tetrathionate broth
Treatment 3. All Shigella species ferment mannitol except:
Most of the cases of bacillary dysentery especially a. Shigella dysenteriae b. Shigella boydii
those due to S. sonnei, are mild and self-limiting c. Shigella flexneri d. Shigella sonnei
and do not require antibiotic therapy. Replacement 4. Which of the following bacteria is more resistant to
of fluids and electrolytes by oral rehydration salt adverse environmental conditions?
a. S. dysenteriae b. S. flexneri
solution is all that is required. c. S. boydii d. S. sonnei
Routine antibacterial treatment is not indicated 5. Verocytoxin is produced by:
in dysentery. Treatment with a suitable antibiotic a. Shigella dysenteriae serotype 1
is necessary in the very young, the aged or the b. Shigella dysenteriae serotype 2
debilitated, and in severe infections. Ampicillin, c. Shigella dysenteriae serotype 8
cotrimoxazole, tetracycline, the quinolone anti­ d. Shigella dysenteriae serotype 10
biotics, such as nalidixic acid and ciprofloxacin are 6. Shigella species most widely found in India is:
a. Shigella dysenteriae b. Shigella boydii
appropriate choices.
c. Shigella flexneri d. Shigella sonnei
Multiple drug resistance plasmids are widely 7. All of the following complications can be associated
prevalent in shigellae. The choice of antibiotic should with S. dysenteriae type I infection except:
be based on the sensitivity of the prevailing strain. a. Arthritis
b. Toxic neuritis
Control c. Hemolytic-uremic syndrome
d. Intestinal perforation
Control consists essentially improving personal an
environmental sanitation. Antibiotics have no place ANSWERS (MCQs)
in prophylaxis. No effective vaccine is available. 1. c; 2. b; 3. a; 4. d; 5. a; 6. c; 7. d

www.ebook3000.com
Chap-38.indd 273 15-03-2016 11:16:01
39
Chapter

Enterobacteriaceae III: Salmonella

LEARNING OBJECTIVES

After reading and studying this chapter, you should Salmonella; antigenic variation of Salmonellae;
be able to: Kauffmann-White scheme
∙∙ Describe morphology, culture charactrestics and ∙∙ Discuss laboratory diagnosis of enteric fever
biochemical reactions of Salmonella sp ∙∙ Describe vaccination against enteric fever
∙∙ Describe the following: Antigenic structure of ∙∙ Discuss Salmonella gastroenteritis.

INTRODUCTION temperature 15–­45°C (optimum 37°C). They can


grow on simple laboratory media. Various media
Genus Salmonella consists of bacilli that parasi­ are as follows:
tise the intestines of a large number of vertebrate A. Nutrient agar and blood agar: Colonies of
species and infect human beings, leading to enteric most strains are moderately large (e.g. 2–3 mm
fever, gastroenteritis, septicemia with or without in diameter), grey-white, moist, circular discs
focal suppuration, and the carrier state. with a smooth convex surface and entire edge
The most important member of the genus is after 24 hours at 37°C.
Salmonella Typhi, the causative agent of typhoid B. Peptone water and nutrient broth: In liquid
fever. Eberth (1880) first observed the typhoid media most strains give abundant growth with
bacillus. Salmon and Smith (1885) described a uniform turbidity.
bacillus which was believed to cause hog cholera. C. Differential and selective solid media: They
include:
SALMONELLA 1. MacConkey agar: The colonies are 1–3 mm
For practical purposes, they may be divided into in diameter pale yellow or nearly colourless
two groups: due to the absence of lactose fermentation.
1. The enteric fever group: It consists of the typhoid 2. Brilliant green MacConkey agar: Salm­
and paratyphoid bacilli that are exclusively or onellae appear as low convex, pale-green
primarily human parasites. trans­lucent colonies 1–3 mm in diameter.
2. The food poisoning group: These are essentially 3. Deoxycholate-citrate agar (DCA): The
animal parasites but which can also infect colonies of salmonellae on DCA are similar
human beings, pro­d ucing gastroenteritis, to or slightly smaller in size than those on
septicemia or localized infections. MacConkey agar.
4. Wilson and Blair’s brilliant-green bismuth
Morphology sulfite agar (BBSA): Jet black colonies
with a metallic sheen are formed due
Salmonellae are gram-negative bacilli, 2–4 × 0.6 µm in
to production of H2S. This is due to the
size. They are motile with peritrichous flagella except
reduction of sulfite to sulfide. This medium
S. Gallinarum­-Pullorum which is nonmotile. They
is particularly valuable for the isolation of S.
are non-acid-fast, noncapsulate and nonsporing
Typhi. S. Paratyphi A and other species that
do not form H2S produce green colonies.
Cultural Characteristics 5. Xylose lysine deoxycholate (XLD) agar:
Salmonellae are aerobes and facultatively anaer­ Most salmonellae produce hydrogen sulfide
obes, growing readily over a range of pH 6–8 and which reacts with ferric ammo­nium citrate

Chap-39.indd 274 15-03-2016 11:16:22


Chapter 39: Enterobacteriaceae III: Salmonella  | 275
in the medium to produce black centers
in their red colonies. The H 2S-negative
salmonellae (e.g. Paratyphi A) form red
colonies without black centers.
Enrichment media: Tetrathionate broth and
Selenite F broth are commonly used enrichment
media for inoculation of specimens escepially feces.

Biochemical Reactions
1. Salmonellae ferment glucose, mannitol, arabi­
nose, maltose, dulcitol and sorbitol, forming
acid and gas except S. Typhi, Gallinarum and
rare anaerogenic variants in other serotypes form
only acid and no gas.
2. Lactose, sucrose, salicin or adonitol are not
fermented.
3. They are, indole positive, MR positive, VP
negative and citrate positive (IMViC – + – +) Fig. 39.1: Antigenic structure of salmonellae
except by S. Typhi and S. Paratyphi A which are Several strains carry fimbriae. Fimbrial antigens
citrate negative as they need tryptophan as the are not important in identification but may cause
growth factor. confusion due to their nonspecific nature and
4. Hydrogen sulfide is produced except by S. widespread shar­ing among enterobacteria.
Paratyphi A, S. Choleraesuis, S. Typhisuis and 1. H antigen: These antigens represent deter­
S. Sendai. minant groups on the flagellar protein. They
5. Urease is not hydrolyzed. are heat-labile and alco­hol-labile, but are well
6. Salmonellae decar­boxylate the amino acids preserved in 0.04–0.2% formaldehyde. In most
lysine, ornithine and arginine. salmonellae, H antigen exists in two alternative
The enteric fever group may be separated phases called phase 1, and phase 2.
biochemically (Table 39.1). When mixed with antisera, H suspensions
agglutinate rapidly, producing large, loose, fluffy
Resistance clumps. The H antigen is strongly immunogenic
Salmonellae are readily killed by moist heat, at 55°C and induces antibody formation rapidly and in
in 1 hour or at 60°C in 15 minutes and most strong high titers follow­ing infection or immunization.
disinfectants. Boiling or chlorination of water and 2. O antigens: The somatic O antigen is a
pasteurization of milk destroy the bacilli. They are phospholipidprotein-polysaccharide complex
killed within five minutes by mercuric chloride (1: which forms an integral part of the cell wall.
500) or 5% phenol. The O antigen is unaffected by boiling (heat-
stable), alcohol (alcohol-stable) or weak acids.
Antigenic Structure The O antigen is less immunogenic than the H
antigen and the titer of the O antibody induced
Antigenic structure of salmonellae is shown in after infection or immunization is generally lower
Figure 39.1. Salmonellae possess the follow­ing than that of the H antibody. O agglutination takes
antigens based on which they are classified and place more slowly and at a higher temperature
identified: optimum (50–55°C) than H agglutination (37°C).
1. Flagellar antigen H. 3. Vi antigen: Almost all recently isolated strains of
2. Somatic antigen O. S. Typhi form Vi antigen as a covering layer
3. Surface antigen Vi, found in some species. outside their cell wall. This antigen is an acidic
Table 39.1  Biochemical characters of typhoid and polysaccharide.When fully developed it renders
paratyphoid bacilli the bacteria agglutinable by Vi antibody and
inagglutinable by O antibody. Felix and Pitt,
Glucose Xylose d-Tartrate Mucate who first described this antigen, believed that
S. Typhi A d A d it was related to virulence and gave it the name
S. Paratyphi A AG − − − ‘Vi antigen’ (Vi for virulence). It is analogous to
S. Paratyphi B AG AG − AG
the K antigens of coliforms.
The Vi antigen is heat-labile. It can be removed
S. Paratyphi C AG AG AG −
from the bacteria by heating a suspension for 1
A = Acid; AG = Acid and Gas; d = Delayed hour at 100°C and centrifuging the bacteria from

www.ebook3000.com
Chap-39.indd 275 15-03-2016 11:16:23
276  |  Section 3: Systemic Bacteriology
the Vi-containing fluid. It is also destroyed by N at both ends, in such a way that it projects above
HCl and 0.5 N NaOH. It is unaffected by alcohol the agar. The strain is inoculated carefully into
or 0.2% formol. the inner tube. After shortest period (8–16 hours)
Originally observed in S. Typhi, the Vi antigen incubation, subcultures are withdrawn from
with similar antigenic specificity is present in S. the top of the agar outside the central tube. This
Para­typhi C and S. Dublin, as well as in certain yields a population of motile cells (Fig. 39.2).
strains of Citrobacter (the Ballerup-Bethesda A U-tube of soft agar may be employed
group). The Vi antigen tends to be lost on serial instead of Craigie’s tube. Inoculation is made into
subculture. The Vi polysaccharide acts as a one limb and subculture taken from the other.
virulence factor by inhibiting phagocytosis, 2. Phase variation: The flagellar antigens of most
resisting complement activation and bacterial sal­monellae occur in one of two phases. Phase
lysis by the alternative pathway and peroxi­ 1 antigens are either specific for a species or
dase mediated killing. Strains possessing the shared by a few species only. Hence phase I is
Vi antigen were found to cause clinical disease called the ‘specific’ phase. Phase 2 antigens are
more consistently than those lacking it in human widely shared and hence phase 2 is called the
volunteer experiments. ‘nonspecific’ or ‘group’ phase. The presump­
The Vi antigen is poorly immunogenic and tive identification of serotypes, therefore, mainly
following infec­tion only low titers of antibody are depends on the identification of the H antigens
produced. Vi antigen is not employed in the Widal in phase I, which are relatively ‘specific’.
test because detection of the Vi antibody is not Phase 1 antigens are designated a, b, c, d,
helpful for the diagnosis of cases. The total absence etc., and after z, as z 1, z2, etc. Phase 2 antigens
of the Vi antibody in a proven case of typhoid fever are far fewer and are termed I, 2, etc. In some
indicates poor prognosis. The antibody disappears species, antigens belonging to phase 1 may
early in con­valescence. Its persistence indicates the occur as the phase 2 antigens (for example, e, n,
development of the carrier state. The Vi antigen x, z 15). The strains that possess both phases are
affords a method of epidemiological typing of the S. known as ‘diphasic’ and the strains that possess
Typhi strains based on specific Vi bacteriophages. only one phase are known as monophasic (e.g.
Phenolized vaccine induces no Vi antibody S. Typhi, S. Paratyphi A, S. Enteritidis, etc.).
though low titers are produced by the alcohol­ized A culture will contain cells with the flagellar
vaccine. The protective efficacy of the Vi antigen an­tigens of both phases but generally one or
is demonstrated by the success of the purified Vi the other phase will predominate so that the
vac­cine for typhoid now in routine use. culture is aggluti­nated only by one of the phase
antisera. It may be necessary to identify the
Antigenic Variations flagellar antigens of both phases for serotyping
The antigens of salmonellae undergo phenotypic of salmonella isolates. A culture in phase 1 can
and genotypic variations. be converted to phase 2 by passing it through
1. H–O variation: This variation is associated a Craigie’s tube containing specific phase 1
with the loss of flagella. When salmonellae antiserum, and the reverse conversion achieved
are grown on agar containing phenol (1 : 800), by using phase 2 antiserum (Fig. 39.2).
flagella are inhibited. This change is phenotypic 3. V–W variation: Almost all recently isolated
and temporary. Flagella reap­p ear when strains of S. Typhi form Vi antigen as a covering
the strain is subcultured on media without
phenol. Salmonellae may rarely lose flagella by
mutation resulting in the development of stable
or irreversible mutant. For example, a stable
nonmotile mutant of S. Typhi is the 901-O strain
which is widely employed for the prepa­ration
of O-agglutinable bacterial suspensions.
Craigie’s tube: Gener­ally, the loss of flagella is
not total and there occurs only a diminution
in the number of flagella and the quantity of
the H antigen. Flagellated cells are found in
small numbers in such cultures. To obtain a
population of motile cells, rich in H antigen,
from such cultures, selection may be carried out
by using Craigie’s tube. This consists of a wide
tube containing soft agar (0.2%) at the center of
which is embedded a short, narrow tube open Fig. 39.2: Craigie’s tube

Chap-39.indd 276 15-03-2016 11:16:23


Chapter 39: Enterobacteriaceae III: Salmonella  | 277
layer outside their cell wall. When fully Table 39.2  Biochemical reactions of Salmonella
developed it renders the bacteria agglutinable subgenera
by Vi antibody and inagglutinable by O antibody.
Test Subgenera
This is called the ‘V form’. The Vi antigen is either
partially or completely lost after a number of 1 II III IV
subcul­tures. Such cultures are inaggluttinable Dulcitol + + – –
with the Vi antiserum but readily agglutinable fermentation
with the O antiserum. This is called the ‘W’form. Lactose fermentation – – + –
With partial loss of Vi antigen, the organisms Malonate utilization – + + –
are agglutinable with both O and Vi antisera d-Tartrate + – – –
and these intermediate forms are known as VW
Salicin fermentation – – – +
forms.
Culture in KCN – – – +
Other Vi-containing bacilli, such as S.
Paratyphi C and S. Dublin do not completely
mask the O antigen.
4. S–R variation: The smooth(S) to rough(R) Subgenus III: It contains bacilli, formerly designated
variation is associated with the change in Arizona, originally isolated from lizards but sub­
colony morphology and loss of the O antigen sequently found in reptiles, birds, domestic animals
and of virulence. Conversion into R forms and human beings. Many of them are prompt
occurs by mutation. Suspensions in saline lactose fermenters.
are autoagglutinable The colonies become Subgenus IV: Subgenus IV strains are rarely
large, rough and irregular. Rough variation encountered and may be considered atypical
is common in laboratory strains maintained members of subgenus II.
by serial subcultivation. S–R variation may Modern taxonomical techniques, have shown
be prevented to some extent by maintaining that all the members of the genus Salmonellae
cultures on Dorset’s egg media in the cold, or and of the former genus Arizona are so closely
ideally by lyophilization. related that they should all be considered as
5. Variation in O antigen: Changes in the structural belonging to a single species. Thus, a new species
formulae of the O antigen may be induced by name S. enterica has been coined to include all
lysogenization with some converting phages, salmonellae. S. enterica is classified into seven
resulting in the alteration of the serotypes. Thus, subspecies named enterica, salamae, arizonae,
S. Anatum (serotype 3,10:e,h:1,6) is converted by diarizonae, houtenae, bongori and indica. Most of
phage 15 into S. Newington (serotype 3,15:e,h:1, the serotypes that infect mammals are found in a
6) and S. Newington into S. Minneapolis subspecies also designated enterica. Subspecies
(serotype 3,15, 34:e, h:1,6 ) by phage 34. It is likely enterica corresponds to the former subgenus 1.
that such changes occurring in nature contribute Such classification and nomenclature would
to the abundance of Salmonella seroyypes. be too complicated for use in clinical bacteriology,
while being taxonomically correct. For example,
Classification and Nomenclature the taxonomically correct name for the typhoid
The classification and nomenclature of salmonellae bacillus would be ‘Salmonella enterica, subspecies
have undergone several modifications over the enterica, serotype Typhi’, but this is abbreviated
years. Inclusion in the genus is based on common to S. serotype Typhi, or simply S. Typhi. Serotype
biochemical properties. Classification within the name should be given in Roman and not in italics.
genus is on antigenic characterization based on the Therefore, the old practice of referring to clinically
Kauffmann-White scheme. important salmonellae serotypes by the species
name continues in clinical bacteriology.
A. Classification Based on Biochemical
Reactions B. Kauffmann–White Scheme of Classification
On the basis of biochemical reactions, the genus Classification within the genus is on antigenic
salmonellae is classified into four subgenera (Table characterization based on the Kauffmann–White
39.2): scheme. This scheme depends on the identification,
by agglutination, of the structural formulae of the O
Subgenus 1: It is the largest and medically the most and H antigens of the strains (Table 39.3).
important group contains all the species which This scheme, first developed in 1934, classifies
commonly cause human and animal infections. the salmonellae into different O groups, or O
Subgenus II: It contains mostly species isolated serogroups, each of which contains a number
from reptiles. of serotypes pos­s essing a common O antigen

www.ebook3000.com
Chap-39.indd 277 15-03-2016 11:16:23
278  |  Section 3: Systemic Bacteriology
Table 39.3  Kauffmann–White classification–Antigenic formulae of some representative serotypes of
Salmonella
Serogroup Oantigen Serotype name O antigens and Vi H antigens
group
Phase 1 Phase 2
2 A Paratyphi A 1, 2, 12 a [1, 5]
4 B Paratyphi B 1, 4, [5], 12 b 1, 2
Stanley 1.4, [5], 12, 27 d 1, 2
Typhimurium 1.4, [5], 12 i 1, 2
Heidelberg 1.4, [5], 12 r 1, 2
7 C1 Choleraesuis 6, 7 c 1, 5
Paratyphi C 6, 7 [Vi] c 1, 5
Typhisuis 6, 7 c 1, 5
Virchow 6, 7 r 1, 2
8 C2-C3 Muenchen 6, 8 d 1, 2
Newport 6, 8, 20 e, h 1, 2
Hadar 6, 8 z10 e, n, x
Miami 1, 9, 12 a 1, 5
- Sendai 1, 9, 12 a 1, 5
9 D1 Typhi 9, 12 [Vi] d –
Enteritidis 1, 9, 12 g, m [1, 7]
Dublin 1, 9, 12, [Vi] g, p –
Panama 1, 9, 12 I, v 1, 5
Gallinarum 1, 9, 12 – –
3,10 E1 Anatum 3, 10, [12], [1Q,; H) e, h 1, 6

not found in other O groups. The O groups first more than 2400 serotypes of salmonellae, giving
defined were designated by capital letters A to Z individual names is not realistic.
and those discovered later by the number (51–67)
of the’ characteristic O anti­gen. It would be more Differentiation of Antigenically Similar
logical to name the serogroups according to their Strains
characteristic O antigen factor number rather than
Serotypes that share the same antigenic formula
by letters, i.e. to abandon the letters A–Z used to
may be distinguished from one another by bio­
designate early O groups. Hence, O groups become:
chemical tests, e.g. Paratyphi C has the same O and
O2 (A), O4 (B), O7 (Cl), O8 (C2–C3), 09, 12 (Dl),
H antigens as Choleraesuis, and Typhisuis. Thus,
09, 46 (D2), 03, 10 (EI) etc. Some serogroups were
Paratyphi C ferments d-tartrate and trehalose,
subdivideed into subserogroups (C1–C4; E1–E4).
but not Stern’s glycerol. Choleraesuis ferments
Serogroups A–Z and 51–67 represented O antigens
d-tartrate, but not trehalose or Stern’s glycerol, and
2–50 and 51–67 respectively. Groups 02 to 03, 10
Typhisuis only trehalose.
(A–EI) contain nearly all the salmonellae that are
important pathogens in man and animals.
Within each O group the different serotypes are Bacteriophage Typing
distinguished by their particular H antigen or com­ Strains within a particular serotype may be differen­
bination of H antigens (Table 39.3). Kauffmann– tiated into a number of phage types by their patterns
White classification gave species status to each of susceptibility to lysis by members of a series of
serotype. The species were named according to phages with different specificities. Intraspecies
the disease caused (S. Typhi), the animal source (S. classification of S. Typhi for epidemiolocal
Gallinarium), the discoverer (S. Schottmulleri), the purpose was made possible by bacteriophage
name of the patient from whom the first strain was typing, first developed by Craige and Yen (1937).
isolated (S. Thompson), or the place of isolation They observed that a bacteriophage acting on the
(S. Poona). This was satisfactory so long as the Vi antigen of the typhoid bacillus (Vi phage II) is
serotypes were not too many but now with some highly adaptable. The parent phage is called phage

Chap-39.indd 278 15-03-2016 11:16:24


Chapter 39: Enterobacteriaceae III: Salmonella  | 279
A. It could be made specific for a particular strain of by Paratyphi A, B or C. The clinical features tend
typhoid bacillus by serial propagation in the strain. to be more severe’ with S. Typhi (typhoid fever).
Such adaptation was obtained by phenotypic or
genotypic variation.Phage type E1, O and A are the a. Typhoid Fever
most common in India. The infection is acquired by ingestion. On reaching
Serotypes that are subdivided in existing the gut, the bacilli attach themselves to the microvilli
systems of phage typing include Typhi, Paratyphi of the ileal mucosa on the epithelium. They then
A, Paratyphi B, Enteritidis, Hadar, Thompson, pene­trate to the lamina propria and submucosa,
Typhimurium and Virchow. Thus, over 100 different where they are phagocytized by neutrophils and
phage types of Typhi and over 230 phage types macro­p hages. They resist intracellular killing
of Typhimurium are distinguished. Among the and multi­ply within these cells. They enter the
Paratyphi A isolated from India, phage types 1 and mesenteric lymph nodes, where after a period of
2 are the most common. multiplication they invade the bloodstream via
the thoracic duct. During this period the bacilli
Phage typing is carried out at the National
are seeded in the liver, gall bladder, spleen,
Phage Typing Center and is coordinated by the
bone marrow, lymph nodes, lungs and kidneys,
International Reference center. The National
where further multiplication takes place. After
Salmonella Phage Typing Center for India is located
multiplication in these organs, bacilli pass into the
at the Lady Hardinge Medical College, New Delhi.
blood, causing a second and heavier bacteremia,
Phage types A and E1 are the most common and
the onset of which approximately coincides with
present throughout India.However, the relative
that of fever and other signs of clinical illness.
prevalence in different regions is subject to change
The salmonellae multiply abundantly in the
from time to time.
gall bladder as bile is a good culture medium for
the bacillus and are discharged continuously into
Biotyping the intestine where Peyer’s patches and other
Biotyping is a useful adjunct to phage typing for gut lym­phoid tissues of ileum become involved.
it can subdivide a large group of untypale strains These become inflamed and infiltration with
or members of common phage types. Similarily, mononuclear cells, followed by necrosis, sloughing
strains of the same biotype may be subdivided into and the formation of characteristic typhoid ulcers
different phage types. occurs. Ulceration of the bowel leads to two
major complications of the disease—intestinal
Pathogenesis perforation and hemorrhage. ­
Clinical course: The incubation period is usually
Salmonellae are strict parasites of animals or
7–14 days. The onset is usually gradual and early
humans. All vertbrates appear capable of harboring
symptoms are often vague: headache, mal­aise,
these bacteria in their gut and salmonellae can
anorexia, a coated tongue and abdominal dis­
colonize virtually all animals.
comfort with either constipation or diarrhea.
In the untreated case the temperature shows
Host-adapted Serotypes a step ladder rise over the first week of the illness,
Some serotypes exhibit host specificity. S. Typhi, remains high for 7–10 days and then falls by lysis
S. Paratyphi A and usually,but not invariably S. during the third or fourth week. Physical signs
Paratyphi B are confined to human beings. Other include a relative bradycardia at the height of the
salmonellae are parasitic in various animals— fever, hepatomegaly, splenomegaly and often a
domestic animals, rodents, reptiles and birds. rash of rose spots, found on the front of the chest
during the second or third week and fade on
Clinical Syndromes pressure. The white blood cell count is normal or
low. Apparent recovery can be followed by relapse
Salmonellae cause the following four major in 5–10% of untreated cases. Classic typhoid fever
syndromes: is a serious infection which, when untreated, has
1. Enteric fever. a mortality approaching 20%.
2. Bacteremia with focal lesion. Complications: The most important complications
3. Gastroenteritis or food poisoning. are intestinal perforation, hemorrhage and
4. Asymptomatic carrier state. circulatory collapse. Some degree of bronchitis or
bronchopneumonia is always found.
1. Enteric Fever b. Paratyphoid Fever
The term enteric fever includes typhoid fever S. Paratyphi A and B cause paratyphoid fever which
caused by S. Typhi and paratyphoid fever caused resembles typhoid fever but is generally milder.

www.ebook3000.com
Chap-39.indd 279 15-03-2016 11:16:24
280  |  Section 3: Systemic Bacteriology
S. Paratyphi C may also cause paratyphoid fever A. Isolation and Identification of the Bacilli
but more often it leads to a frank septicemia with It may be done by culture of specimens such
suppurative complications. as patient’s blood, feces, urine, bone marrow,
duodenal drainage rose spots etc. For the
2. Bacteremia with Focal Lesion leborotatory diagnosis of enteric fever, selection
This is associated with S. Cholerae-suis but may of relevant specimens depends upon duration
be caused by any Salmonella serotype. Infection of illness, e.g. blood for culture must be taken
occurs by oral route and there is early invasion of repeatedly. Urine culture may be positive after
the bloodstream with possible focal lesions and second week and stool second or third week.
in particular may cause septicemic disease (with
focal lesions, in lungs, bones, meninges, etc.) but Blood Culture
intestinal manifectations are often absent. Blood
Blood cultures are positive in approximately 90%
culture are positive.
cases in the first week of fever, in approximately
75% of cases in the second week, 60% in the third
3. Gasteroenteritis or Food Poisoning
week and 25% thereafter till the subsidence of
This is the most common manifestation of pyrexia (Fig. 39.3, Table 39.4).
Salmonella infection. In the USA, Salmonella With all aseptic precautions, about 5–10 mL of
Typhimurium and Salmonella enteritis are blood is collected by venipuncture and inoculated
prominent (For detail see under the heading, into a culture bottle containing 50–100 mL of 0.5%
“Salmonella gastroenteritis”.) bile broth. Large quantity (5–10 mL) of blood is
required because the number of organisms are not
4. Asymptomatic Carrier State numerous and may be quite small. The blood is
Most people infected with salmonella continue
to excrete the organism in their stools for days
or weeks after complete clinical recovery, but
eventual clearance of the bacteria from the body
is usual. A few patients continue to excrete the
salmonellae for prolonged periods.

Epidemiology
The typhoid and paratyphoid bacilli are essentially
human parasites. Man is the reservoir host and
most infections can be traced to a human source,
or at least to a source of human sewage. All other
salmonellae have animal hosts.
S. Paratyphi A is prevalent in India and other
Asian countries, Eastern Europe and South
America, S. Paratyphi B in Western Europe, Britain
and North America; and S. Paratyphi C in Eastern Fig. 39.3: Laboratory diagnosis of typhoid fever. The appro­
Europe and Guyana. Enteric fever is endemic in all ximate percentages of tests found positive during different
parts of India. Paratyphoid B is rare and C very rare. stages of the disease (from 1st to 5th week). A. Widal aggluti­
nation. B. Feces culture. C. Blood culture
The disease occurs at all ages but is probably most
common in the 5–20 year age group.
The source of infection is a patient, or far more
Table 39.4  Positivity of various specimens at
different phases of enteric fever
frequently, a carrier. Food handlers or cooks who
become carriers are particularly dangerous. The Duration Specimen % positivity
best known of such typhoid carriers was Mary 1st week Blood culture 90
Mallon (‘Typhoid Mary’), a New York cook who Feces culture –
caused at least seven outbreaks affecting over 200
Widal test –
persons over a period of 15 years.
2nd week Blood culture 75
Laboratory Diagnosis Feces culture 50
Bactriological dignosis of enteric fever consists of: Widal test Low titer
A. Isolation and identification of the bacilli. 3rd week Blood culture 60
B. Demonstration of circulating antigen.
Feces culture 80
C. Demonstration of antibodies in patient’s serum.
Widal test 80–100
D. Other laboratory tests.

Chap-39.indd 280 15-03-2016 11:16:24


Chapter 39: Enterobacteriaceae III: Salmonella  | 281
diluted between 1 in 5 and 1 in 10 in culture medium well as in patients. The use of enrichment and
to reduce the concentration of natural antimicrobial selective media and repeated sampling increase
constituents to a sub-effective level. The addition of the rate of isolation. Fecal culture is particularly
liqoid (sodium polyanethol sulphonate) counteracts valuable in patients on antibiotics as the drug does
the bactericidal action of blood. not eliminate the bacilli from the gut as rapidly as
After overnight incubation at 37°C, the bile it does from the blood.
broth is subcultured on MacConkey agar and Fecal samples are plated diretly on MacConkey
blood agar media. These plates are incubated at agar, DCA and Wilson and Blair’s brilliant-
37°C for 24 hours. Pale nonlactose fermenting green bismuth sulfite agar media. The last is
(NLF) colonies that may appear on MacConkey highly selective and should be plated heavily.
agar medium are picked out for biochemical tests On MacConkey and DCA media, salmonellae
and motility. Salmonellae will be gram-negative appear as pale colonies. On Wilson and Blair’s
and motile bacilli. brilliant-green bismuth sulfite agar media,
Subcultures repetition: If salmenellae are not S. Typhi forms large black colonies, with a metallic
obtained from the first subculture from bile broth, sheen. S. Paratyphi A produces green colonies due
then subcultures should be repeated every other the absense of H2 S production.
day till growth is obtained. If no growth is obtained For enrichment, one tube each of selenite F
after 5–7 days, then the culture is declared negative. and tetrathionate broth are also inoculated. These
are also incubated at 37°C for 8–12 hours with
Castaneda’s method of culture: Castaneda’s subsequent subculture on MacConkey agar and
method of culture may be adopted to eliminate the DCA media.
risk of introducing contamination during repeated
subculture, and also for economy and safety. In this
Urine Culture
method, a double medium is used. The bottle of bile
broth has an agar slant on one side. After inoculation Urine culture is less useful than the culture of
of blood, the bottle is incubated in the upright blood and feces because salmonellae are shed in
position. For subculture, the bottle is merely tilted the urine irregularily and infrequently. Generally
so that the broth runs over the surface of the agar. It cultures are positive only in the second and third
is reincubated in the upright position. If salmonellae weeks and then only in about 25% of cases. The
are present, colonies will appear on the slant. rate of isolation is improved by repeated sampling.
Clean voided urine samples are centrifuged and
the deposit inoculated into enrichment and
Clot Culture
selective media as for fecal culture.
An alternative to blood culture is the clot culture.
Here, with strict aseptic precautions, 5 mL of blood
Other Materials for Culture
is withdrawn from the patient into a sterile test
tube and allowed to clot. The serum is pipetted Bone marrow culture is valuable and it is positive
off and used for Widal test. The clot is broken up in most cases even when blood culture is negative.
with a sterile glass rod and added to a bottle of bile Culture of bile obtained by duodenal aspiration is
broth incorportating streptokinase (100 units/mL). usually positive and this may be employed for the
Streptokinase in the broth facilitates lysis of the clot detection of carrirs. Other matrials, such as rose
with release of bacteria trapped in the clot. spots, pus from suppurative lesions, CSF and
sputum may yield isolation at times. At autopsy,
Advantage of clot culture: i. Clot cultures yields a cultures may be obtained from the gallbladder,
higher rate of isolation than blood cultures. liver, spleen and mesenteric lymph nodes.
ii. A sample of serum also becomes available for
Widal test. Even though agglutinins may be absent Colony morphology: Pale nonlactose fermenting
in the early stages of the disease, a Widal test (NLF)colonies that may appear on MacConkey
provides a baseline titer against which the results agar or DCA medium after incubating at 37°C for
of tests performed later may be evaluated. 24 hours are picked out for biochemical tests and
motility. Salmonellae will be gram-negative and
Feces Culture motile bacilli.
Salmonellae are shed in the feces throughout the Biochemical reactions: Salmonellae will be
course of the disease and even in convalescence. indole and urease negative, catalase positive,
Hence, fecal cultures are almost as valuable as oxidase negative, nitrate reduction positive and
blood cultures in diagnosis. Feces culture are ferment glucose, mannitol and maltose but not
generally positive after second week of illness. Iso­ lactose or sucrose. S. Typhi will be anerogenic
lation from the feces is of less certain significance. and ferments glucose and mannitol with
A positive fecal culture may occur in carriers as production of acid only, while paratyphoid bacilli

www.ebook3000.com
Chap-39.indd 281 15-03-2016 11:16:24
282  |  Section 3: Systemic Bacteriology
(S. Paratyphi A, B and C) will form acid and gas i. Widal Test
from sugars. Testing the patient’s serum for Salmonella antibodies
Identification: Identification of the isolate is by is useful only in the diagnosis of enteric fever (Widal
slide agglutination. reaction). Tests for the presence of Salmonella
antibodies in the patient’s serum may be of value
Slide agglutination test: A loopful of the growth in the diagnosis of enteric fever. This is a test for
from an agar slope is emulsified in two drops of measurement of H and O agglutinins for typhoid
saline on a slide. One emulsion acts as a control to and paratyphoid bacilli in the patient’s serum. The
show that the strain is not autoagglutinable. If S. patient’s serum is tested by tube agglutination for its
Typhi is suspected (that is, when no gas is formed titers of antibodies against H, O and Vi suspensions
from glucose), a loopful of typhoid O antiserum of the enteric fever bacteria likely to be encountered,
(factor 9/group D) is added to one drop of e.g. S. Typhi and S. Paratyphi A in India. Two types
bacterial emulsion on the slide, and agglutination of tubes are generally used for the test:
looked for after rocking the slide gently. Prompt 1. Dreyer’s agglutination tube (narrow tubes with
agglutination indicates that the isolate belongs a conical bottom) for the H agglutination.
to Salmonella group D. Its identity as S. Typhi is 2. Felix tube (a short round bottomed tube) for
established by agglutination with the flagellar the O agglutination.
antiserum (anti-d serum). Quite often, fresh Equal volumes (0.4 mL) of serial dilutions of
isolates of S. Typhi are in the V form and do not the serum (from 1/10 to 1/640) and the H antigens
agglutinate with the O antiserum. Such strains may of S. Typhi (TH) and S. Paratyphi A (AH) and O
be tested for agglutination against anti-Vi serum. antigen of S. Typhi (TO) are mixed in Dreyer’s and
Alternatively, the growth is scraped off in a small Felix’s agglutination tubes respectively. Incubate H
amount of saline, boiled for 20 minutes and tested agglutinations for 2–4 hours in water bath at 37°C
for agglutination with the O antiserum. and read after standing on the bench for half an
Where the isolate is a nontyphoid Salmonella hour. Incubate O agglutinations for 4–6 hours at
(producing gas from sugars), it is tested for 37°C and read after overnight refrigeration at 4°C.
agglutination with O and H antisera for groups A, Controls tubes containing the antigen and normal
B and C. For identification of unusual serotypes, saline are set to check for autoagglutination. The
the help of the ‘National Salmonella Reference agglutination titers of the serum are read.
Center’ should be sought. The National Salmonella H agglutination leads to the formation of loose,
Reference Center in India is located at the Central cotton woolly clumps, while O agglutination is
Research Institute, Kasauli. The reference center seen as a disc like pattern at the bottom of the
for salmonellae of animal origin is at the Indian tube. Control (Felix) tube shows a compact
Veterinary Research Institute, Izatnagar. deposit. In both, the supernantant fluid is rendered
clear. The highest dilution of the serum showing
B. Demonstration of Circulating Antigen agglutination indicates the titer of the antibody.
In the early phase of the disease, typhoid bacillus The antigens used in the test are the H and O
antigens are consistently present in the blood antigens of S. Typhi and S. Paratyphi A and B. The
and also in the urine of patients. The antigen can paratyphoid O antigens are not employed as they
be demonstrated by sensitized staphylococcal cross react with the typhoid O antigen due to their
coagglutination. Staphylococcus aureus containing sharing of factor 12. Although suspensions may be
protein A (Cowan 1 strain) is stabilized with prepared from suitable stock laboratory cultures,
formaldehyde and then coated with S. Typhi anibody. there is little need for that nowadays because
When a 1% suspension of such sensitized cells is ready made Widal kits of stained antigens available
mixed on a slide with serum from patients in the first commertially are now widely used.
week of typhoid fever, the typhoid antigen present in
the serum combines specifically with the antibody
Interpretation of Widal Test
attached to the staphylococcal cells producing visible 1. Stage of the disease: The agglutinins titer will
agglutination within two minutes. The test is rapid, depend on the stage of the disease. Usually
sensitive and specific but is not positive after first week agglutinins appear by the end of first week
of the disease. ELISA test has also been employed to (seventh to tenth day) of the illness, so that
detect typhoid antigen in blood and urine. blood taken earlier may give a negative result
and is inconclusive. The titer then increases
C. Demonstration of Antibodies in steadily till the third or the fourth week, after
which it declines gradually.
Patient’s Serum 2. Rising titer: Demonstration of a rise in titer of
i. Widal test. antibodies by testing two or more samples is
ii. Other serological tests. more meaningful than a single test.

Chap-39.indd 282 15-03-2016 11:16:24


Chapter 39: Enterobacteriaceae III: Salmonella  | 283
3. Single Widal test: The results of a single Widal 1. Isolation of the bacillus: The identification of
test should be interpretated with caution. Levels the fecal carriers is by isolation of the bacillus
of significance is difficult to lay down though it from feces or from bile. The frequency and
is generally stated that titers of 1/100 or more intensity of bacillary shedding vary widely
for O agglutinins and 1/200 or more for H and it is essential, therefore, to test repeated
agglutinins are significant. It is necessray to obtain samples. Chance of isolation is increased by
information on the distribution of agglutinin cholagogue purgatives. For the detection of
levels in ‘normal sera’ in different areas. urinary carriers, repeated urine culture should
4. Immunization: Serum from an individual be carried out.
immunized with TAB vaccine will generally 2. Widal test: Widal test is of no value in the
have antibodies to S. Typhi, S. Paratyphi A and detection of carriers in endemic countries like
B. However, in case of infection antibodies will India. The demonstration of Vi agglutinins
be seen only against the infecting species. (1:10 or more) has been claimed to indicate the
5. O and H agglutinis: H agglutinis persist longer carrier state. Whlie this is useful as a screening
(for many months) than O agglutinins. Therefore, test but confirmation should be made culture.
rise in O agglutinins indicate recent infection. 3. Sewer-swab technique: The tracing of carriers
6. Anamnestic response: Persons who have in cities may be accomplished by the ‘sewer-
prior infection or immunization may develop swab’ technique. Gauze pads left in sewers
anamnestic response during an unrelated and drains are cultured, and by tracing positive
fever, such as malaria, influenza, etc. This may swabs, one may be led to the house harboring
be differentiated by repetition of the test after a a carrier.
week. The anamnestic shows only a transient
4. Filtration through millipore membrane:
rise, while in enteric fever the rise is sustained.
Another technique of isolating salmonellae
7. Fimbrial antigen: Bacterial suspension used
from sewage is filtration through millipore
as antigens should be free from fimbria which
membrane and culturing the membranes on
may produce false positive results.
highly selective media such as Wison and Blair
8. Effect of treatment: Patients treated early with
media.
chroramphenicol may show a poor agglutinin
response.
PROPHYLAXIS
ii. Other Serological Tests
Control depends essentially on safe water
Enzyme-linked immunosorbent assay (ELISA), supply, proper sewage disposal, handling of food
indirect hemagglutination test and CIEP are other hygienically, and periodic examination of food
serological methods of diagnosis. handlers to ascer­tain that they are not carriers.
D. Other Laboratory Tests
Vaccines against Typhoid Fever
i. Total leukocyte count (TLC): Leucopenia
with a relative lymphocytosis is seen. Killed whole Cell Vaccine: TAB Vaccine
ii. Diazo test in urine: This test becomes positive Heat-killed, phenol-preserved whole-cell vaccines
generally between 5th and 14th day of fever con­taining a mixture of cultures of Typhi, Paratyphi
and remains positive till the fever subsides. A and Paratyphi B (TAB) have been used for many
Procedure: Equal volumes of patient’s urine and years in countries with a high endemic level of
the diazo reagent are mixed and a few drops of 30% typhoid fever. The TAB vaccine which came into
ammonium hydroxide are added. If the test is positive, general use contained S. Typhi 1,000 million and
a red or pink froth develops on shaking the mixture S. Paratyphi A and B, 750 million each per mL
killed by heating at 50–60°C and preserved in 0.5%
Diazo Reagent phenol. Acetone-killed vac­cine preserved in the
∙∙ Solution A­—Sulfanilic acid; Conc. H 2SO 4; dry state also provides similar protection.
distilled water; Dose schedule: The vaccine is given in 2 doses of
∙∙ Solution B—Sodium nitrite; distilled water; for 0.5 mL subcutaneously at an interval of 4–6 weeks
use, 40 parts of solution A are added to one part followed by a booster dose every 3 years.
of solution B.
Protection: Field trials have shown that such
DIAGNOSIS OF CARRIERS preparations confer protection against typhoid
fever with an effi­cacy of 70–90% for 3–7 years.
The detection of carriers is important for
epidemilogical and public health purposes. It is Side effects: Local and general reactions lasting for
also useful in screening food handlers and cooks. one or two days are quite frequent.

www.ebook3000.com
Chap-39.indd 283 15-03-2016 11:16:24
284  |  Section 3: Systemic Bacteriology
Note: In India a divalent vaccine containing S. Typhi chloramphenicol did not pose any problem
and S. Paratyphi A are now in use instead of the TAB in typhoid fever till 1972, when resistant
vaccine, eliminating S. paratyphi B which is rare in strains emerged in Mexico and Kerala in India.
the country or the monovalent typhoid vaccine is Chloramphenicol resistant typhoid fever appeared
preferred. In Europe and USA, monovalent vaccine in epidemic form first in Calicut (Kerala) in early
containing S. Typhi is employed as paratyphoid A 1972. It became endemic and was confined to
and B infections are rare in these countries. Kerala till 1978. Subsequently such strains carrying
drug resistance plasmids appeared in many
Oral Vaccine–Live Oral (Ty21) Typhoid Vaccine other parts of India. Resistance was originally
The live oral vaccine (typhoral) is a stable mutant of confined to phage type D1-N, but later to types
S. Typhi strain (Ty21a), lacking the enzyme UDP- C5, A and O. This multidrug resistance was due
galactose-4-epimerase (Gal E mutant). On inges­tion, to a transmissible plasmid carrying resistance
it initiates infection but ‘self destructs’ after four or determinants to chlora­mphenicol, streptomycin,
five cell divisions, and therefore, it cannot induce sulfadiazine and tetra­cycline (CSSuT).
any illness. This oral vaccine is available in enteric A ciprofloxacin-resistant S. Typhi isolate was
coated capsule containing 109 viable lyophilized first reported from the United Kingdom in 1992. At
mutant bacilli. present, the drugs useful in treatment of such multire­
sistant typhoid cases are the later fluoroquinolones
Dose schedule: Three doses of the vaccine are (such as ciprofloxacin, pefloxacin, ofloxacin) and
given on alternate days to children. The course the third generation celphalosporins (such as
consists of one capsule orally, taken an hour before ceftazidime, ceftrioxone, cefotaxime).
food, with a glass of water or milk, on days 1, 3, and
5. No antibiotic should be taken during this period. SALMONELLA GASTROENTERITIS
Protection: It is safe and confers 65–96% protection Salmonella gastroenteritis (more appropriately
for 3–5 years. enterocolitis) or food poisoning is generally a
zoonotic disease. The source of infection being ani­
Vaccine of Purified Vi Antigen (Typhim-Vi) mal products. It may be caused by any salmonella
This injectable vaccine (typhim-Vi) contains except S. Typhi. In most parts of the world, S.
purified Vi polysaccharide antigen (25 µg per dose) Typhimurium is the commonest (30–40%) species.
from S. Typhi strain Ty2. Some other common species have been S. Enteri­
It is given as a single subcutaneous or intra­ tidis, S. Haldar, S. Heidelberg, S. Agona, S. Virchow,
muscular injection. This vaccine is not recom­ S. Seftenberg, S. Indiana, S. Newport and S. Anatum
mended for children younger than 2 years. S. Dublin.
In the trials conducted the protection conferred
Source of Infection
by the vaccine was 64% and 72%.
The most frequent sources of Sal­monella food
Treatment poisoning are poultry, meat, milk and milk products.
Specific antibacterial therapy for enteric fever Of great concern are eggs and egg products.
became available only in 1948 with the introduction Food contamination may also result from the
of chloram­phenicol, which continued as the sheet droppings of rats, lizards or other small animals.
anchor against the disease till the 1970s when Human carriers do occur but their role is minimal.
resistance became common. Ampicillin, amoxicillin,
and trimethoprim-sulfamethoxazole have been used Clinical Features
successfully. Clinically, the disease develops after a short
In the past decade, third-generation cephalo­ incubation period of 24 hours or less, with diarrhea,
sporins, particularly ceftri­axone and cefoperazone, vom­iting, abdominal pain and fever. It usually
and fluoroquinolones including norfloxacin, subsides in 2­–4 days but in some cases a more
ciprofloxacin, ofloxacin, and pefloxacin all are at prolonged enteritis develops.
least as effective as chloramphenicol in treating
typhoid fever. Ciprofloxacin has emerged as the Laboratory Diagnosis
drug of choice for the treatment of adult typhoid,
The laboratory diagnosis depends on the isolation
and is proving equally effective and free from side-
of the causal organism from samples of feces or
effects in children.
suspected foodstuffs.

DRUG RESISTANCE Treatment


S. Typhi resistant to chloramphenicol was first Treatment of uncomplicated, noninvasive salmo­
reported in England in 1950. Resistance to nellosis is symptomatic. Antibiotics should not be

Chap-39.indd 284 15-03-2016 11:16:24


Chapter 39: Enterobacteriaceae III: Salmonella  | 285
used. Not only do they not hasten recovery but they
™™ Infections with S. Typhi and S. Paratyphi or
may actually increase the period of fecal shedding disseminated infections with other organisms
of the bacilli. But for the serious invasive cases, should be treated with an effective antibiotic
antibiot­ic treatment is needed.­ such as fluoroquinolones (e.g., ciprofloxacin),
chloramphenicol, trimethoprim/sulfamethoxazole,
SALMONELLA SEPTICEMIA or a broad-spectrum cephalosporin can be used
™™ TAB vaccine, Vi capsular polysaccharide antigen
S. Cholerae-suis, in particular may cause septicemic vaccine, and acetone-inactivated parenteral vaccine
disease with focal suppurative lesions, such as are the killed vaccines and Ty21 a vaccine is the oral
osteomyelitis, deep abscesses, endocarditis, vaccine used for immunization against typhoid fever
™™ Salmonella gastroenteritis is generally a zoonotic
pneumonia and meningitis. Focal lesions may
disease. It may be caused by any salmonella except S.
develop in any tissue. Typhi. The most frequent sources of Sal­monella food
Salmonella septicemia is prolonged and poisoning are poultry, meat, milk and milk products
characterized by fever, chills, anorexia, and anemia. ™™ Salmonella septicemia: S. Cholerae-suis, in
Gastroenteritis is minor or even absent, and the particular may cause septicemic disease with focal
organism is rarely cultured from feces. Infection suppurative lesions, such as osteomyelitis, deep
abscesses, endocarditis, pneumonia and meningitis.
occurs by oral route and the incubation period is
short. The case fataliy may be as high as 25%.
Salmonelle may be isolated from the blood IMPORTANT QUESTIONS
or from the pus from the suppurative lesions.
1. Name the salmonellae causing enteric fever.
Feces culture may sometimes may be positive.
Describe in detail the laboratory diagnosis of
Septicemic samonellosis should be treated with enteric fever.
chroramphenicol or other appropriate antibiotics 2. Describe the pathogenesis and laboratory diagnosis
as determined by sensitivity tests. of enteric fever.
3. Write short notes on:
MULTIRESISTANT SALMONELLAE a. Antigenic structure of Salmonella
In India, several hospital outbreaks of neonatal b. Vi-antigen or surface antigen
septicemia caused by multiresistant salmonellae c. Kauffmann-White scheme
have occurred in recent years. Mortality in d. Clot culture
neonates is very high unless early treatment is e. Widal test
started with antibiotics to which the infecting strain f. Vaccination against enteric fever
is sensitive. g. Salmonella gastroenteritis
h. Salmonella septicemia.
Key Points
Salmonella MULTIPLE CHOICE QUESTIONS (MCQs)
™™ Gram-negative bacilli, nonsporing, and noncapsu­ 1. All the following enrichment media are used for
lated, motile bacilli isolation of Salmonella except:
™™ Aerobic and facultative anaerobic grow easily on a. Alkaline peptone water
a variety of nonselective media (e.g. nutrient agar) b. Selenite F broth
and selective (e.g. Wilson and Blair’s bismuth sulfite c. Brilliant green tetrathionate broth
medium, xylose, lysine deoxycholate agar, etc).. d. Tetrathionate broth
Selenite F and tetrathionate broth are enrichment
2. The selective medium used for isolation of
media
Salmonella is:
™™ Salmonellae possess the (1) Flagellar antigen H, (2)
a. Wilson and Blair’s brilliant-green bismuth sulfite
Somatic antigen O (3) Surface antigen Vi, found in
agar
some species
b. Cary-Blair medium
™™ Salmonella are classified by Kauffman–White scheme
c. Thiosulfate citrate bile salt agar
based on structural formulae of the O and H antigens
d. Xylose, lysine deoxycholate agar
of the strains
3. All the following Salmonella serotypes are motile
™™ S. Typhi and S. Paratyphi are strict human pathogens
except
(no alternative reservoir)
a. Salmonella Typhi
™™ Diseases: Asymptomatic colonization; enteric fever;
b. Salmonella Typhimurium
enteritis; bacteremia
c. Salmonella Choleraesuis
™™ Diagnosis: Isolation of the S. Typhi or S. Paratyphi
d. Salmonella Gallinarum
from blood, feces, urine, bone marrow, duodenal
drainage rose spots, etc. 4. Hydrogen sulfide is not produced by the following
serotypes of Salmonella:
™™ Blood culture is positive in 90% cases in first week
of fever a. S. Paratyphi A
b. S. Choleraesuis
™™ Widal test is the traditional serologic test used for
the diagnosis of typhoid fever c. Both of the above
d. None of the above

www.ebook3000.com
Chap-39.indd 285 15-03-2016 11:16:25
286  |  Section 3: Systemic Bacteriology
5. Which of the following serotypes of Salmonella is/ 9. Most important complications of enteric fever is/
are anerogenic? are:
a. S. Typhi b. S. Paratyphi A a. Intestinal perforation b. Hemorrhage
c. S. Enteritidis d. All of the above c. Circulatory collapse d. All of the above
6. Citrate is not used by the following Salmonella: 10. Most important specimen for detection of carriers
a. Salmonella Typhi in enteric fever is:
b. Salmonella Paratyphi A
a. Feces b. Urine
c. Both of the above
d. None of the above c. Blood d. Sputum
7. Which of the following Salmonella is/are primarily 11. All the following are the examples of killed vaccines
human pathogens? used against typhoid fever except:
a. S. Typhi b. S. Paratyphi A a. TAB vaccine
c. S. Paratyphi B d. All of the above b. Vi capsular polysaccharide antigen vaccine
8. The most important specimen for isolation of c. Acetone-inactivated parenteral vaccine
Salmonella Typhi culture in first week of enteric d. Ty21a vaccine
fever?
a. Blood b. Urine ANSWERS (MCQs)
c. Feces d. CSF 1. a; 2. a; 3. d; 4. c; 5. a; 6. c; 7. d; 8. a; 9. d; 10. a; 11. d

Chap-39.indd 286 15-03-2016 11:16:25


40
Chapter

Vibrio, Aeromonas and Plesiomonas

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Differentiate between classical and El Tor vibrios
be able to: ∙∙ Discuss laboratory diagnosis of cholera
∙∙ Describe morphology, cuture characteristics and ∙∙ Describe the following: cholera vaccine; nonagglut­
biochemical reactions of Vibrio cholerae inating vibrios; halophilic vibrios
∙∙ Discuss antigenic structure of Vibrio cholerae
∙∙ Describe the following: pathogenesis of cholera;
mechanism of action of cholera toxin

INTRODUCTION It is actively motile, by means of a single, polar


sheathed flag­ellum. The motility is of the darting
The second major group of gram-negative, type. They are nonsporing, noncapsulated and
facultatively anaerobic, fermentative bacilli are the non­acid-fast (Fig. 40.1).
genera Vibrio, Aeromonas, and Plesiomonas. Vibrio
and Aeromonas are now classified in the families Cultural Characteristics
Vibrionaceae and Aeromonadaceae, respectively.
The cholera vibrio is strongly aerobic, growth being
Plesiomonas is closely related to Proteus and has now
scanty and slow anaerobically. It grows within a
been placed in the Enterobacteriaceae family.
tem­perature range of 16–40°C (optimum 37°C).
VIBRIO Growth is better in an alkaline medium and it
occurs freely between pH 7.4 and 9.6 (optimum pH
Of the 35 Vibrio species recognized,12 have been 8.2). V. cholerae is a nonhalophilic vib­rio. It grows
implicated in gastrointestinal and extraintestinal well on ordinary media.
infections in man. The genus can be divided into
nonhalophilic vibrios, includ­ing V. cholerae and
other species and halophilic species. The species
most frequently isolated from clinical specimens
are strains of V. cholerae, V. parahaemolyticus, V.
vulnificus, V. mimicus and V. alginolyticus.

VIBRIO CHOLERAE
Morphology
These are gram-negative, short, curved, cylindrical
rods, about 1.5 μm × 0.2–0.4 μm in size. The cell
is typically comma-shaped. S-shaped or spiral
forms may be seen due to two or more cells lying
end-to-end. In old cultures, they are frequently
highly pleomorphic. The vibrios are seen arranged
in parallel rows, described by Koch as the ‘fish in
stream’ appearance in stained films of mucous
flakes from acute cholera cases. Fig. 40.1: Cholera vibrios

www.ebook3000.com
Chap-40.indd 287 15-03-2016 11:17:33
288  |  Section 3: Systemic Bacteriology
A. Ordinary Media C. Plating Media
i. Nutrient agar: After overnight growth, 1. Alkaline bile salt agar (BSA); pH 8.2
colonies are moist, translu­cent, round disks, This is modified nutrient agar medium containing
about 1–2 mm in diameter, with a bluish tinge 0.5% sodium taurocholate. The colonies on BSA are
in transmitted light. The growth has a dis­ similar to those on nutrient agar medium.
tinctive odor. 2. Monsur’s gelatin taurocholate trypticase
ii. MacConkey agar: The colonies are color­less, tellurite agar (GTTA) medium; pH 8.5
but become reddish on prolonged incubation After 24 hours incubation, vibrios produce small
due to the late fermentation of lactose. (1–2 mm) translucent colonies with grayish­black
iii. Blood agar: Colonies are initially surrounded center and a turbid halo, due to hydrolysis and
by a zone of greening, which later becomes denaturation of gelatin. After 48 hours incuba­tion,
clear due to hemodigestion. colonies increase in size to 3–4 mm.
iv. Gelatin stab culture: At first a white line 3. Thiosulfate-citrate-bile-sucrose (TC­BS) agar—
of growth appears along the track of the pH 8.6
inoculating wire. Liq­u efaction of gelatin This is most used selective plating medi­um for
begins at the top, which spreads downwards in vibrios. Constituents of this medium are sodium
infundibuli­form (funnel shaped) or napiform thiosulphate, sodium citrate, ox bile, sucrose, yeast
(turnip-shaped) in 3 days at 22°C. extract, peptone, sodium chloride, ferric cit­rate,
v. Peptone water: When incubated at 37°C in thymol blue, bromothymol blue (indicator) and
liquid media, such as peptone water, it forms water. On this differential medium, the colonies
a fine surface pellicle because of its affinity for of sucrose-fermenting vibrios, e.g. V. cholerae, are
oxygen. yellow, those of sucrose-non-fermenting vibrios,
e.g. V. parahaemolyticus, are green.
B. Special Media Biochemical Reactions
a. Holding or Transport Media 1. Sugar fermentation: It ferments glucose,
1. Venkatraman–Rama­krishnan (VR) medium mannitol, maltose, mannose and sucrose and
A simple modified form of this medium is prepared ferments lactose only after several days (late
by dissolving 20 g crude sea salt and 5 g peptone lactose-fermenter).
in one liter of distilled water and adjusting the pH 2. Cholera red reaction: V. cholerae is strongly
to 8.6–8.8. It is dispensed in screw capped bottles indole positive and reduces nitrates to nitrites.
in 10–15 mL amounts. About 1–3 mL stool is to be These two properties contribute to the ‘cholera
added to each bottle. Vibrios do not multiply in red reaction’ which is tested by adding a few
this medium but remain viable for several weeks. drops of concentrated sulfuric acid to a 24-hour
pep­tone water culture at 37°C. A reddish pink
2. Cary–Blair medium color is developed due to the formation of
This is a buffered solution of disodium hydrogen ni­troso-indole with cholera vibrios.
phosphate, sodium thioglycollate, sodium chloride 3. It is catalase and oxidase-positive, methyl red
and agar to distilled water and pH 8.4. It is a suitable and urease negative.
transport medium for Salmonella and Shigella as 4. It decarbo­xylates lysine and ornithine but does
well as for vibrios. not utilize arginine.
3. Autoclaved sea water: Autoclaved sea water also 5. Gelatin is liquefied.
serves as a holding medium. 6. Voges-Proskauer reaction and hemolysis of
sheep RBCs are positive in El Tor biotype and
b. Enrichment Media both these reac­tions are negative in classical
biotype.
The rapid growth and tolerance of vibrios for alkaline 7. String test: Vibrio colonies may be identified
conditions is exploited in the formulation of media by the ‘string test’. A loopful of the growth is
used for their isolation. mixed with a drop of 0.5% sodium deoxycholate
1. Alkaline peptone water: Alkaline peptone water in saline on a slide. If the test is positive, the
at pH 8.6 is useful for preliminary enrichment suspension loses its turbidity, becomes mucoid
of vibrios from feces or other contaminated and forms a ‘string’ when the loop is drawn
materials. slowly away from the suspension.
2. Monsur’s taurocholate tellurite peptone
water at pH 9.2. RESISTANCE
Both these are good transport as well as enrich­ Cholera vibrios are susceptible to heat, drying and
ment media. acids, but resist high alkalinity. Vibrios are killed

Chap-40.indd 288 15-03-2016 11:17:33


Chapter 40: Vibrio, Aeromonas and Plesiomonas  | 289
by heating at 56°C for 30 minutes or within a few Serological Classification
seconds by boiling. In general, the El Tor vibrio A serological classification was introduced by
sur­vives longer than the classical cholera vibrio. Gardner and Venkatraman (1935). Cholera vibrios
In grossly contaminated water, such as the and biochemically similar vibrios, possessing a
Ganges water of India, the vibrios do not survive com­mon flagellar (H) antigen were classified as
for any length of time, due to the appar­ently large Group A vibrios, and the rest as Group B vibrios
amounts of vibriophages present. They survive in comprising a heterogeneous collection.
clean tap water for 30 days. On fruits, they survive The somatic (O) antigen structure is of
for 1–5 days at room temperature and for a week fundamental importance in the identification
in the refrigerator. of this organism. Based on the major somatic
(O) antigen, Group A vibrios were classified
Glucose Mannitol Maltose Mannose Sucrose Lactose into ‘subgroups’ (now called O serogroups or
A + + + + – serovars), more than 200 of which are currently
Indole NO3 Catalase Oxidase MR VP* known. (Flowchart 40.1). All isolates from
+ reduction + + – – epidemic cholera (till 1992) belonged to serogroup
+ O-1. Therefore, in the diagnostic laboratory
Lysine Ornithine Arginine Gelatin Sheep group O-1 antiserum (commonly called ‘cholera
+ + – + RBCs non­d ifferential serum’) came to be used for
hemo­
identifying pathogenic cholera vibrios (which are
lysis*
+ referred to as ‘agglutinable vibrios’). Other vibrio
*In case of El Tor biotypes VP and sheep RBCs hemolysis are
isolates which were not agglutinated by the O–1
positive and all biochemical reactions are similar. antiserum came to be called nonagglutinable or
NAG vibrios. They were considered nonpathogenic
and hence also called non­cholera vibrios (NCV).
Antigenic Structure
1. Flagellar antigen (H antigen): Many vibrios Biotypes of V. cholerae 01
share a single heat-labile flagellar antigen.
There are two biotypes of V. cholerae 01: classical
2. O antigen (LPS, endotoxin): V. cholerae has
and El Tor biotypes. The differ­e nces between
O lipopolysaccharides (LPS, endotoxin) that
classical and El Tor biotypes V. cholerae are
confer serologic specificity.
shown in Table 40.3. El Tor biotype produces
There are at least more than 200 O antigen
acetoin in the Voges-Proskauer test, agglutinates
groups. V. cholerae strains of O group 1 and O group
fowl erythrocytes, lyses sheep erythrocytes in a
139 cause classic cholera. Occasionally, non-Ol/
heart infusion broth with glycerol, grows in the
non­0139 V cholerae causes cholera-like disease.
presence of polymyxin (50 unit disk), is resistant
to Mukerjee’s group IV phage and sensitive to the
Classification group V phage of Basu and Mukerjee. The classical
In the past, many oxidase positive, motile, curved biotype has the opposite pro­perties.
rods were rather loosely grouped as vibrios. Precise 1. Chick red cell agglutination test: A loopful
criteria have been laid down for differentiating of the organisms from an agar cultures is
vibrios from related genera (Table 40.1). emulsified in a drop of saline on a slide and
Vibrios were classified by Heiberg (1934) into a drop of 2.5% chick erythrocyte suspension
six groups based on the fermentation of mannose, is added. Clumping of erythrocytes within a
sucrose and arabinose. Two more groups were minute indicates a positive test. The test is
added later. Chol­era vibrios belong to Group I positive with all EI Tor strains but classical
(Table 40.2). cholera vibrios are negative.

Table 40.1  Differentiation of vibrios from allied genera


Genus Oxidation–Fermentation Utilization of amino acids String test
(Hugh-Leifson Test)
Oxidation Fermentation Lysine Arginine Ornithine
Vibrio + +1 + − + +
Aeromonas + +2 − + − V
Pseudomonas + − V V V −
Plesiomonas + + + + + −
1 = No gas produced; 2 = Gas may or may not be produced; V = Variable reaction

www.ebook3000.com
Chap-40.indd 289 15-03-2016 11:17:33
290  |  Section 3: Systemic Bacteriology
Table 40.2  Heiberg grouping of vibrios Table 40.4  Serotypes of cholera vibrios
Group Fermentation Sucrose Arabinose Serotype O antigens
of mannose Ogawa AB
I A A – Inaba AC
II – A – Hikojima ABC
III A A A
IV – A A 3. Sensitivity to cholera phage IV
V A – – All strains of classical cholera vibrio are lysed
VI – – – by Mukherjee’s group IV phage routine test
dilution (RTD), while all EI Tor strains are
VII A – A
not lysed. This is considered to be the most
VIII – – A dependable test for differen­tiating between EI
Tor and classical strains.

Flowchart 40.1: Antigen classification of vibrios (Gardner Subtypes of V. cholerae 01


and Venkatraman, updated) Strains of V. cholerae 01 may be further subdivided
on the basis of their 0 antigens into three subtypes,
Ogawa, Inaba and Hikojima. This is on the basis
of differences in minor 0 antigens (A, B and C).
Antigen A is present in all the three subtypes. 0
antigens present in Ogawa, Inaba and Hikojima
are A and B, A and C, and A, Band C respectively
(Table 40.4).
Nonagglutinating (NAG) vibrios: Though NAG
vibrios are not agglutinable by the O–1 antiserum,
they are readily agglutinated by their own antisera.
The non–Ol vibrios (the so called NAG vibrios) have
been classified into many serogroups, currently up
to more than 200. V. cholerae O1 and O139 are
responsible for causing classic cholera, which
can occur in epidemics or worldwide pandemics.
Occasionally, non-O1/non O139 V. cholerae causes
cholera-like disease.

V. cholerae 0139
Table 40.3  Differeces between classical cholera In 1992 cases of cholera indistinguishable from that
and El Tor vibrios caused by V. cholerae 01 were reported in Madras
Test Classical cholera El. Tor (Chennai), India. By mid-January 1993 similar
isolates were found in neighboring Bangladesh,
Hemolysis − +*
and these rapidly spread north, following the
Voges-Proskauer − +* course of the major rivers and raising fears of a new
Chick erythrocyte − + pandemic.
agglutination V. cholerae 0139 may have evolved from V.
Polymyxin B sensitivity† + − cholerae 01, but with modified lipopolysaccharide
Group IV phage + − structure. V. cholerae 0139 makes a polysaccharide
susceptibility capsule like other non-O 1 V. cholerae strains, while
El Tor phage 5 − + V. cholerae 01 does not make a capsule. In the
susceptibility affected areas, this strain replaced the El Tor vibrios
* Strain isolated after 1961 give variable results. as the epidemic and environmental serov­ar. It also

50 i. u. disk. showed a tendency to be more invasive, causing
bacteremic illness in some. Both O-1 El Tor and
2. Sensitivity to polymyxin B: The organism is O-139 strains began to coexist in endemic areas.
tested by the disc diffusion method using discs
containing 50 units of polymyxin B. All strains Phage Typing
of classical cholera vibrio are sensitive and all Phage typing schemes have been standardized
strains of EI Tor vibrio are resistant. for classical and El Tor biotypes. New molecular

Chap-40.indd 290 15-03-2016 11:17:34


Chapter 40: Vibrio, Aeromonas and Plesiomonas  | 291
Table 40.5  Phase types of strains of classical cholera toxin, CT, or CTX). CT production is
biotype of Vibrio cholerae 01 (Mukerjee et al. 1957) determined by a filamentous phage integrated with
the bacterial chromosome.
Phage type Sensitivity to phase group
I II III IV Mechanism of Action
1 + + + +
The toxin molecule, of approximately 84,000 MW
2 − + + + consists of one A and 5 B subunits. The B (binding)
3 + − + + subunit of cholera toxin binds to the ganglio­side
4 − − + + GM1 receptors on the intestinal epithelial cells,
5 + + − +
which promotes entry of subunit A into the cell.
The A (active) subunit, on being transported into
the enterocyte dissociates into two fragments A1
Table 40.6  Phase types of strains of El Tor biotype and A2. The A2 frag­ment only links the biologically
of Vibrio cholerae 01 (Basu and Mukerjee, 1968) active A1 to the B subunit. The active portion (A1) of
Phage type Sensitivity to phase group the A subunit enters the cell and activates adenyl
cyclase. The cholera enterotoxin causes the transfer
I II III IV V
of adenosine diphosphoribose (ADP ribose) from
1 + + + + + nicotinamide adenine dinucleotide (NAD) to a
2 + + + − + regu­latory protein, which is part of the adenylate
3 + + − + + cyclase enzyme responsible for the generation of
4 + + − − + intracellular cyclic adenosine monophosphate
(cAMP). The result is irreversible activation of
5 + − − − +
adenylate cyclase and overpro­duction of cAMP.
6 − + − − + This in turn causes inhibition of uptake of Na+

and Cl ions by cells lining the villi, together with

meth­o ds like ribotyping have added further hypersecretion of Cl and HCO3 ions. This blocks
refinements to strain typing. the uptake of water which normally accompanies

Strains of the classical biotype of V. cholerae 01 Na+ and Cl absorption, and there is a passive net
can be divided into 5 types by means of 3 phages outflow of water across mucosal cells, leading to
(I–III) and a fourth phage (IV) lyses all classical serious loss of water and electrolytes.
but not El Tor strains (Table 40.5). On the basis of
lysis by 4, phages, El Tor strains can be divided into Cholera
6 types. All these strains are lysed by a fifth phage
The incubation period varies from less than 24
(V) (Table 40.6).
hours to about five days. The clinical illness may
be­gin slowly with mild diarrhea and vomiting in
Pathogenesis 1–3 days or abruptly with sudden massive diarrhea.
In human infection, the vibrios enter orally through The cholera stool is typically a colorless watery
contaminated water or food. Vibrios are high­ly fluid with flecks of mucus, said to resemble water
susceptible to acids, and gastric acidity provides in which rice has been washed (hence called ‘rice
an effective barrier against small doses of cholera water stools’). It has a characteristic inoffensive
vibrios. sweetish odor. In compo­sition it is a bicarbonate-
The cholera vibrios are ingested in drink or food rich isotonic electrolyte solu­tion, with little protein.
and, in natural infections, the dose must often be Its outpouring leads to diminution of extracellular
small. After passing the acid barrier of the stomach fluid volume, hemocon­centration, hypokalemia,
the organ­isms begin to multiply in the alkaline base-deficit acidosis and shock. There is rapid loss
environment of the small intestine. of fluid and elec­trolytes, which leads to profound
Once in the small intestine, strains of V. cholerae dehydration, circula­tory collapse, and anuria.
O1 migrate towards epithelial cells. A hemagglutinin- The common complications are muscular
protease (former­ly known as ‘cholera lectin’) cleaves cramps, renal failure, pulmonary edema, cardiac
mucus and fibronectin. Adhe­sion to the epithelial ar­rhythmias and paralytic ileus. The mortality rate
surface and colonization may be facilitated by without treatment is between 25% and 50%. The El
special fimbria, such as the ‘toxin co­regulated pilus’ Tor biotype tends to cause milder dis­ease than the
(TCP). classic biotype.

Cholera Toxin (CT) Epidemiology


Vibrios multiplying on the intestinal epithelium Cholera is both an epidemic and endemic disease.
produce a toxin (choleragen, cholera enterotoxin, Seven major pandemics of cholera have occurred

www.ebook3000.com
Chap-40.indd 291 15-03-2016 11:17:34
292  |  Section 3: Systemic Bacteriology
since 1817, resulting in thousands of deaths and by 1994 the El Tor strain regained its dominance
major socioeconomic changes. and the threat of an 0­139 pandemic diminished.
Cholera is an exclusively human disease.
Infection is generally spread by contaminated Laboratory Diagnosis
water or foods. The source of the contamination A. Specimen
is usually the feces of car­riers or patients with
∙∙ Watery stool
cholera.
∙∙ Rectal swabs.
All pandemics of cholera caused by serotype 01,
al­though 0139 can cause similar diseases and may B. Collection of Specimen
cause a pandemic.
a. Stool: A fresh specimen of stool should be
India, more specifically the large deltaic area
collected for laboratory examination. Sample
of the Ganges and Brahmaputra in Bengal, is its
should be collected before the person is treated
homeland, where it has been known from very
with antibiotics. Collection may be made
ancient times. Till early in the nineteenth century,
generally in one of the following ways:
cholera was virtually confined to India, periodically
i. Rubber catheter: Fecal specimens from early
causing large epidemics in different parts of the
acute cases should be collected into a sterile
country. From 1817 to 1923, cholera vibrios had
container, preferably through a soft sterile
spread from Bengal, in six separate pandemic waves,
rubber catheter inserted into the rectum.
involv­ing most parts of the world. After the end of the
ii. Rectal swab: Rectal swabs are useful in
6th pandemic in 1923, till 1961 the disease remained
collecting specimens from convalescents.
con­fined to its endemic areas, except for an isolated
Collection from a bedpan should be avoided
epi­demic in Egypt in 1947. It was largely due to the
because of the risk of contamination or the
threat of pandemic cholera that international health
presence of disinfectant.
organizations came into being.
The seventh pandemic occurred in 1961 and was C. Transportation
first to be caused by the El Tor biotype. It originated
If possible, specimens should be processed without
from Sulawesi (Celebes), Indonesia. V. cholerae 0 I
delay but, if there is to be a delay of more than 6
biotype El Tor was first isola­ted by Gotschlich at the
hours in their reaching the laboratory, feces or
El Tor Quarantine Station in Egypt. After spreading
rectal swabs should be placed in a liquid alkaline
to Hongkong and the Philippines, it spread steadily
transport medium. Stool samples may be preserved
westwards, invading India in 1964. By 1966, it had
in VR fluid or Cary­Blair medium for long periods.
spread throughout the Indian subcontinent and
If the specimen can reach the laboratory in a
West Asia. In the 1970s the pandemic extended to
few hours, it may be trans­ported in enrichment
Africa and parts of Southern Europe.
media, such as alkaline peptone water or Monsur’s
Wherever the El Tor vibrio has caused long­
medium.
standing infection, in all those areas it has
Strips of blotting paper may be soaked in the
displaced the classical vibrio. It supplanted the
watery stool and sent to the laboratory packed
classical bio­type in India during the 1960s and
in plas­tic envelopes if transport media are not
spread into other parts of the country previously
availa­ble.
free from this infec­tion. Similarly, in Bangladesh
Whenever possible, specimens should be
it entirely displaced classical biotype by 1973, but
plated at the bedside and the inoculated plates sent
the latter staged a comeback in 1982 and replaced
to the laboratory.
El Tor vibrio in many areas.
In January, 1991, the pandemic reached Peru, D. Microscopy
thus encircling the globe in 30-year time. By 1994
most parts of Cen­tral and South America had been Direct microscopic examination of feces is not rec­
involved and rendered endemic. ommended. For rapid diagnosis, the characteristic
In 1992, cases of cholera indistinguishable motili­ty of the vibrio and its inhibition by antiserum
from that caused by V. cholerae 01 were reported can be demonstrated under the dark field or phase
in Madras (Chennai), India. By mid-January contrast microscope.
1993 similar isolates were found in neighboring
Bangladesh, and these rapidly spread north, E. Culture
following the course of the major rivers and raising The specimens sent in enrichment media should
fears of a new pandemic. The new strain was be incubated for 6–8 hours, including transit time.
assigned to a new serogroup, 0139 Bengal. The new The specimens sent in holding media should be
strain continued spreading, eastwards to the South inoculated into enrichment media, to be incubated
East Asian coun­tries, and westwards to Pakistan, for 6–8 hours before being streaked on a selective
China and some parts of Europe. But surprisingly, and a nonselective medium.

Chap-40.indd 292 15-03-2016 11:17:34


Chapter 40: Vibrio, Aeromonas and Plesiomonas  | 293
Selective Media non-Ol cholera vibrio. Specific antiserum against
The plating media employed usually are bile salt 0-139 is available.
agar (BSA), MacConkey agar for nonselective and
GTTA and TCBS agar for selective plates. Generally,
F. Serological Diagnosis
the plates are examined after overnight incubation In patients who have not been immunized, a retro­
at 37°C. spective diagnosis of infection can be made by the
demonstration of a rising titer of agglutinins or
Colony morphology: On MacConkey medium, vibriocidal antibodies in paired sera; or antibody
they form translucent colonies, on GTTA medium against cholera toxin (CT) may be detected in an
they form translucent colonies with grayish-black ELISA test.
center and a turbid halo and on TCBS, they form
yellow colonies.
Detection of Carriers
Identification: Do the Gram staining from the For isolation of vibrios from carriers, essentially
suspected colonies and look for gram-­negative the same techniques are to be followed, except
curved or comma-shaped rods. Perform motility that more than one cycle of enrichment may be
and oxidase tests. Cholera vibrios show characteristic necessary. As vibrio excretion is intermittent,
motility and are oxidase ­positive. repeated stool ex­amination will yield better results.
Slide agglutination: Pick up oxidase-positive Serological examination is of little use in the
colonies with a straight wire and test by slide diagnosis of cases though it may be helpful in
agglutination with V. cholerae 0 1 antiserum. If assessing the prevalence of cholera in an area. The
positive, agglutination may be repeated using tests availa­ble are agglutination using live or killed
specific Ogawa and Inaba antisera. Hikojima vibrio sus­pensions, indirect hemagglutination,
strains will aggluti­nate well with both Ogawa and vibriocidal test and antitoxin assay. Of these, the
Inaba antisera. If agglutination is negative with complement de­pendent vibriocidal antibody test
one colony, repeat the test with at least five more is the most useful.
colonies as 01 and non-01 vibrios may co-exist in
the same specimen. Examination of Water Samples
If slide agglutination is positive, the isolate For examination of water samples for vibrios,
is tested for chick red cell agglutination. This is enrichment or filtration methods may be employed.
employed for presumptive differentiation between A. Enrichment method: In this method, 900 mL of
El Tor and clas­sical cholera vibrios. If no vi­brios are water are added to 100 mL ten­fold concentrated
isolated, a second cycle of enrichment and plating peptone water at pH 9.2, incubated at 37°C for
may succeed in some cases. 6–8 hours and a second enrichment done before
plating on selective media.
Biochemical Reactions B. Filtration method: For the filtration technique
The identity of the organism should be con­firmed the water to be tested should be filtered through
biochemically in a set of conventional tests. The use the Millipore membrane filter, which is then
of a Hugh and Leifson O/F test, Moller’s arginine placed directly on the surface of a selective
dihydrolase, lysine and ornithine decarboxylases, medium and incubated. Colonies appear after
arabinose, inosi­tol, mannose and sucrose peptone overnight incu­bation. Sewage should be diluted
water sugars and a test for growth in the absence in saline, filtered through gauze and treated as
of NaCI by inoculating either a 1% tryptone water for water.
with no added NaCl or a CLED plate.
For further characterization of the biotype of Treatment
the V. cholerae 01 isolate, do VP test, agglutination 1. Oral rehydration therapy (ORT): In cholera,
of fowl RBCs, haemolysis of sheep RBCs, and absolute priority must be given to the life­saving
sensitivity to polymyxin B, Mukerjee phages IV replacement of fluid and electrolytes. Oral
and V (Table 40.3). The strain may be sent to rehy­dration therapy (ORT) is often sufficient,
the International Reference Centre for vibrio but severe cases may require intravenous
phage typing at the National Institute of Cholera rehydration.
and Enteric Disease (NICED) at Kolkata for 2. Antibacterial therapy: Oral tetracycline was
confirmation of identity and for O-serotyping. recom­mended for reducing the period of vibrio
When the isolated strain is not agglutinated excretion and the need for parenteral fluids. This
by V. cholerae 01 antiserum, it should be tested dramatically reduces infectivity. Alternatively,
for agglutination with V. cholerae H antiserum. the long-acting tetracycline (doxycycline) may
Any vibrio which is agglutinated by H antiserum be used for chemoprophylaxis, if the prevailing
and not by 01antiserum is considered to be to be strains are not resistant.

www.ebook3000.com
Chap-40.indd 293 15-03-2016 11:17:34
294  |  Section 3: Systemic Bacteriology
Immunity ii. Live oral vaccine
Gastric acid provides some protection against Recombinant DNA vac­cine with expression of V.
cholera vibrios. In cholera, the vibrios remain cholerae 01 in attenuated strain of S. Typhi Ty21 as
confined to the intestine, where they multiply and a carrier bacterium has been developed. The live
elaborate the enterotoxin which is responsible salmonellae colonize the Peyer’s patches of the
for the disease. Immunity, therefore, may be small intestine and induce IgA response by local
directed against the bacterium or against the toxin- imm­une system of the gut.
antibacterial or antitoxic. Natural infection confers
some amount of immunity but it does not seem to HALOPHILIC VIBRIOS
last for more than 6-12 months and reinfections are Vibrios that have a high requirement of sodium
known after this period. chloride are known as halophilic vibrios. Their
An attack of cholera is followed by immunity natural habitat is sea water and marine life. Some
to reinfection, but the duration and degree of halo­philic vibrios have been shown to cause human
immunity are not known. The presence of antitoxin disease—V. parahaemolyticus, V. alginolyticus and
antibodies has not been associated with protection. V. vulnificus.
Immunity may be local, in the intestine, or
systemic. The appearance of local antibodies in the Vibrio parahaemolyticus
intestine has been known for a long time. These are Morphologically, it resembles V. cholerae except
known as ‘coproantibodies as they appear in the that it is capsulated, shows bipolar staining and
faeces. They consist of IgG, IgM and IgA. has a tendency to pleomorphism, especially when
grown on 3% salt agar and in old cultures. However,
Prophylaxis being a halophilic species, it grows well in pep­tone
1. General Measures water with 8% (but not 10%) sodium chloride.
It grows well on blood agar. On MacConkey agar
Most important are the provision of safe drinking
it forms pale, nonIactose­fermenting colonies and
water supplies and the proper disposal of human
on sheep blood agar it produces β-hemolysis. On
feces. Control rests on education and on improvement
TCBS agar, the colonies are green (non-sucrose-
of sanitation, particularly of food and water.
fermenting). Unlike other vibrios, it produces
peritrichous flagella when grown on solid media.
2. Specific Measures—Vaccines
Polar flagella are formed in liquid cultures.
a. Killed whole organism vaccine
It is killed suspension containing 8000 million V. Biochemical Reactions
cholerae per mL, composed of equal numbers of It is oxidase, catalase, indole and citrate positive.
Ogawa and Inaba serotypes. The concentration of It reduces nitrate to nitrite. It ferments glucose,
the vaccine has been increased to 12,000 million maltose, mannitol, mannose and arabinose with
per mL, in order to improve the anti­genic stimulus. the production of acid only. Lactose, sucrose
Primary immunization consists of 2 equal doses, salicin, dulcitol or inositol are not fermented.
injected subcutaneously, at an interval of 4–6 weeks. It is VP positive and decarboxylates lysine and
This vaccine offers about 60% protection for 3–6 ornithine but not arginine.
months. A single dose of vaccine is ineffective in
children below five years of age while two doses at Pathogenesis
1–4 week intervals are protective. However, it confers Not all strains of V. parahaemolyticus are patho­
protection in adults due to its action as a booster genic for human beings. Strains isolated from
because of prior natural infection. environmental sources (such as water, fish,
Cell free somatic antigen preparations are as crabs or oysters) are nearly always nonhemolytic
effective as whole cell vaccine. when grown on a special high salt blood agar
(Wagatsuma’s agar), while strains from human
b. Oral vaccine: Two types of oral cholera vaccines
patients are al­m ost always hemolytic. This
are available in some countries.
hemolysis is known as the Kanagawa phenomenon
i. Nonliving oral B subunit-whole cell (BS-WC) and is due to a heat stable hemolysin.
vaccine: Kana­gawa positive strains being considered
This vaccine consists of killed whole-cell V. cholerae pathogenic for human beings and negative strains
01 in combination with a recombinant B-subunit nonpathogenic.
of cholera toxin (WC/rBS). Vibrio parahaemolyticus causes acute gastro­
It is given orally in two dose schedule, 10–14 enteritis following ingestion of conta­m inated
days apart. On an average, the vaccine confers seafood, such as raw fish or shellfish. After an
50–60% protection for at least 3 years. incubation period of 12–24 hours, nausea and

Chap-40.indd 294 15-03-2016 11:17:34


Chapter 40: Vibrio, Aeromonas and Plesiomonas  | 295
vomit­ing, abdominal cramps, fever, and watery Table 40.7  Some characteristics of V. parahae­
to bloody diarrhea occur. Fecal leukocytes are molyticus and V. alginolyticus
often observed. The enteritis tends to subside
V. V. alginolyticus
spontaneously in 1–4 days with no treatment other
parahaemolyticus
than restoration of water and electrolyte balance.
Indole + +
In Calcutta V. parahaemolyticus could be
isolated from 5–10% of diarrhea cases admitted to VP − +
the Infectious Dis­eases Hospital. Nitrate + +
reduction
Laboratory Diagnosis Urease − −
The feces of patients with a history of recent Sucrose − +
consump­tion of seafood may be examined by the fermentation
methods used for V. cholerae. In the examination of Swarming − +
seafood and sea or estuarine waters for halophilic − −
Growth in 0%
species, including V. parahaemolyticus, enrichment NaCl
culture in alkaline peptone water containing 1%
7% NaCl + +
sodium chloride is used.
10% NaCl − +
Vibrio Vulnificus
V. vulnificus, previously known as L+ vibrio or and V. carchariae (wound in­fection) have been
Beneckea vulnifica, is a marine Vibrio of medical documented.
importance.
AEROMONAS
This halophilic vibrio resembles V. para­
haemolyticus in forming green, nonsucrose-fer­ Until recently, Aeromonas was classified in the
menting colonies on TCBS medium. It is VP family Vibrionaceae. The genus has been placed in
negative and ferments lactose but not sucrose. It has the new family Aeromonadaceae from the family
a salt tolerance of less than eight percent. The ability Vibrionaceae.
of. V. vulnificus to ferment lactose (‘lactose positive Aeromonas is a gram-negative, facultative
vibrios’) a key identifying characteristic.It can grow anaerobic ba­cillus that morphologically resembles
in the presence of a 300 unit disk of polymyxin. members of the Enterobacteriaceae. The most
Vibrio vulnificus can cause severe wound important pathogens are Aero­monas hydrophila,
infections, bacteremia, and probably gastroenteritis. Aeromonas caviae, and Aeromonas ve­ran; biovar
It is responsible for rapidly progressive wound soma. The organisms are ubiquitous in fresh and
infections after exposure to contaminated seawater. brackish water.
Following ingestion of the Vibrio, usually in oysters, The two major diseases associated with Aero­
it penetrates the gut mucosa without causing monas are gastroenteritis and wound infections
gastrointestinal manifestations and enters the (with or with­out bacteremia).
bloodstream, rapidly leading to septicemia with Gastroenteritis typically occurs after the ingestion
high mortality. of contaminated water or food, whereas wound
infections result from exposure to contaminated
Vibrio alginolyticus water. They are opportunistic pathogens.
Vibrio alginolyticus is a halophilic organism formerly Aeromonas strains are susceptible to tetra­
regarded as biotype 2 of V. parahaemolyticus. It cyclines, aminoglycosides, and cephalosporins.
resembles V. parahaemolyti­cus in many respects
and. It has a higher salt tolerance, is VP positive PLESIOMONAS
and ferments sucrose (Table 40.7). It forms large, Plesiomonas are closely related to Proteus and
yellow (sucrose ­fermenting) colonies on TCBS. have now been placed in the Enterobacteriaceae
There is pronounced swarming on nonselective family. It has only one species, P. shigelloides which
solid media. It has been associated with infections is taxonomically related to Proteus species and
of eyes, ears and wounds in human beings exposed serologically related to Shigella sonnei.
to sea water. They are oxidase positive, motile, have multiple
polar flagella gram-negative bacilli and may be
Other Vibrio Species mistaken for vibrios. The organism is found in
V. hollisae, V. mimicus, V. fluvialis, V. furnissii, and V. fresh water and estuarine waters and is ac­quired
damsela are responsible for causing gas­troenteritis, through contact with fresh water, the con­sumption
wound infections, and bacteremia. of seafood, or exposure to amphibians or reptiles.
Single cases of infections with V. metschnikovii P. shigelloides is ubiquitous in surface waters
(bacteremia), V. cincinnatiensis (meningitis), and in soil. It has also been isolated from a variety

www.ebook3000.com
Chap-40.indd 295 15-03-2016 11:17:34
296  |  Section 3: Systemic Bacteriology
of mammals, including dogs, cats, goats, sheep h. Kanagawa phenomenon
and monkey. It is rare1y recovered from human i. Aeromonas
fe­ces. It commonly infects various cold-blooded j. Plesiomonas.
ani­mals like frogs, snakes, turtles and lizards. Man
is infected primarily by ingesting contaminated MULTIPLE CHOICE QUESTIONS (MCQs)
water or food. It causes: 1. All the following Vibrio species require sodium
1. Gastroenteritis, which manifests as a mild, chloride as a growth factor except:
watery diarrhea in which stools are free of blood a. Vibrio cholerae
and mucin. b. Vibrio parahaemolyticus
2. Extraintestinal lesions, including septicemia, c. Vibrio vulnificus
endophthalamitis, septic arthritis, meningitis, d. Vibrio alginolyticus
cellulitis, and acute cholecystitis. 2. Which of the following media can serve a transport
medium for Vibrio cholerae?
Key Points a. Selenite-F broth
b. Tetrathionate broth
™™ Vibrio and Aeromonas are classified in the families
Vibrionaceae and Aeromonadaceae, respectively. c. Venkatraman–Ramakrishnan medium
Plesiomonas are closely related to Proteus and have d. Nutrient broth
now been placed in the Enterobacteriaceae family 3. Classical and El Tor biotypes of Vibrio cholerae can
™™ Vibrio cholerae is short, typically comma-shaped, show be differentiated by which of the following tests?
typical darting type motility. It is strongly aerobic a. Sensitivity to polymyxin B
™™ It grows well on a wide variety of media including b. Agglutination of fowl RBCs
ordinary and special media c. Sensitivity to Mukerjee group IV phage
™™ Transport media such as VR medium and Cary–Blair d. All of the above
medium, o Enrichment media such as alkaline
4. The DNA expressing for production of cholera
peptone water and Monsur’s taurocholate tellurite
peptone, Selective media such as TCBS medium, toxin of Vibrio cholerae is located in:
Monsur’s GTTA medium, and alkaline BSA a. Chromosome b. Plasmid
™™ Two biotypes of V. cholerae 01 strains-EI Tor and c. Transposon d. Phage
classical 5. Cholera toxin resembles which of the following
™™ Classical and El Tor biotype vibrios can be differentiated toxins?
by acetoin in the Voges-Proskauer test, agglutination a. Labile toxin of E. coli
of fowl erythrocytes, lysis of sheep erythrocytes, b. Stable toxin of E. coli.
polymyxin B senisitivity, susceptibility to Mukerjee’s c. Diphtheria toxin
group IV phage and sensitivity to the group V phage
d. Tetanus toxin
™™ Disease: Cholera. Spread is by consumption of
contaminated food or water 6. Stools from suspected cholera cases can be trans­
™™ Laboratory diagnosis: Fresh stool specimen is used for ported to the laboratory in:
dark-field microscopy and direct immunofluorescence. a. Venkatraman–Ramakrishnan medium
The specimen inoculated on a selective and b. Cary–Blair medium
nonselective media. Suspected V. cholerae are tested c. Thick blotting paper
by slide agglutination using specific V. cholerae 01 d. All of the above
antisera. If the colony is identified as V. cholerae 01 7. Which of the following vaccines is/are available for
then it is tested by various tests to determine whether prophylaxis against cholera?
isolated V. cholerae 01 is classical or El tor a. Killed parenteral vaccine
™™ Halophilic vibrios
b. Killed parenteral vaccine
™™ Vibrio parahaemolyticus, Vibrio alginolyticus, and
Vibrio vulnificus are three important halophilic vibrios c. Live oral vaccine
species known to cause infection in humans. V. d. All of the above
parahaemolyticus in humans causes gastroenteritis 8. Which of the following bacteria is/are associated
™™ Aeromonas: Aeromonas species in humans cause with food poisoning due to consumption of sea fish?
gastroenteritis and wound infections a. Vibrio alginolyticus
™™ Plesiomonas: P. shigelloides (the only species) in b. Vibrio parahaemolyticus
humans cause gastroenteritis, celullitis, septic c. Vibrio vulnificus
arthritis, septicemia, neonatal meningitis, etc. d. All of the above
9. Acute diarrhea resembling cholera can be caused by:
IMPORTANT QUESTIONS a. Vibrio parahaemolyticus
b. Vibrio vulnificus
1. Discuss laboratory diagnosis of cholera. c. Vibrio alginolyticus
2. Write short notes on: d. Aeromonas hydrophila
a. Classification of vibrios 10. Which of the following conditions can be caused
b. Noncholera vibrios by Plesiomonas?
c. Differences between classical and El Tor vibrios a. Gastroenteritis b. Septicemia
d. Pathogenesis of cholera
c. Cellulitis d. All of the above
e. Cholera toxin
f. Prophylaxis against cholera ANSWERS (MCQs)
g. Halophilic vibrios 1. a; 2. c; 3. d; 4. b; 5. a; 6. d; 7. d; 8. b; 9. d; 10. d

Chap-40.indd 296 15-03-2016 11:17:34


4141
Chapter

Campylobacter and Helicobacter

Learning Objectives

After reading and studying this chapter, you should ∙∙ Discuss morphology, culture characteristics and
be able to: biochemical reactions of Helicobacter
∙∙ Describe morphology, culture characteristics and ∙∙ Discuss laboratory diagnosis of Helicobacter pylori
biochemical reactions of Campylobacter infections.

Campylobacter and reduce nitrates to nitrites. C. jejuni has the


ability to hydrolyze sodium hippurate.
Introduction: Campylobacter and arcobacter are
grouped into the family Campylobacteriaceae. Pathogenesis
A total of 18 species and subspecies are now
Infection is acquired by ingestion. The jejunum and
recognized; 13 have been associated with hu­man
ileum are the first sites to become colonized, but the
disease. Campylobacter jejuni is the prototype
infection extends distally to affect the terminal ileum
organism in the group and is very common cause
and usually the colon and rectum. The organisms are
of diarrhea in humans.
invasive and may involve mesenteric lymph nodes
Morphology and cause bacteremia. Heat-labile entero­toxin
along with the invasive property of this organism
The genus Campylobacter (Greek, meaning curved
may contribute to the production of the damage.
rod) consists of small, comma-shaped, gram-
negative bacilli that microscopically resemble Mechanism of Producing Diarrhea
vibrios. They are motile by means of a single polar
It can produce diarrhea by following mecha­nisms:
flagellum. Old cultures are coccoid and pleomorphic.
1. It produces a heat-labile enterotoxin resem­
They are nonsporing.
bling CT that raises intracellular levels of cAMP
Cultural Characteristics leading to watery diarrhea.
2. Like Shigella and Salmonella, it penetrates gut
Most species are microaerobic, requiring an
epithelium leading to edematous exuda­tive
atmosphere with decreased oxygen (5% oxygen)
enteritis of jejunum, ileum and colon, with
and increased hydrogen and carbon dioxide (CO2)
infiltration by polymorphonuclear leukocytes
level for aerobic growth. Many pathogenic species
and ulceration of the mucosa.
are thermophilic, growing well at 42°C.
Selective media for isolation of C. jejuni are But­ Clinical manifestations: Clinical manifestations
zler’s selective medium, Skirrow’s Campylobacter are acute onset of crampy abdominal pain, profuse
selective medium, Preston Campylobacter selec­tive diarrhea that may be grossly bloody, headache,
medium and Blaser’s medium (Campy-BAP). Plates malaise, and fever. Usually the illness is self-limited
are incubated for 48 hours. Colonies are circular and to a period of 5–8 days.
convex but those of thermophilic group, particularly Complications: The complications are reactive
C. jejuni, are flat and tend to swarm on moist agar. (aseptic) arthritis and Guillain–Barre syndrome, a
form of peripheral polyneuropathy.
Biochemical Reactions
Campylobacters do not ferment carbohydrates and Epidemiology
utilize a respiratory (oxidative) pathway. They are C. fetus is a major cause of abortion in sheep
strongly oxidase positive. They are catalase positive and cattle worldwide. Campylobacter infections

www.ebook3000.com
298  |  Section 3: Systemic Bacteriology
are zoonotic, with a variety of animals serving lost fluids and electrolytes. Antimicrobial treatment
as reservoirs. Hu­m ans acquire the infections should be reserved for patients with severe or
with C. jejuni and C. coli after consumption of complicated infections. Severe gastroenteritis and
contaminated food, milk, or wa­ter. septicemia are treated with erythromycin (drug of
choice), tetracyclines and quinolones.
Laboratory Diagnosis Control: Gastroenteritis is prevented by proper
Laboratory diagnosis depends on isolation of the preparation of food (particularly poultry), and
Campylobacter from feces. consumption of pasteurized milk; prevention of
contaminated water supplies also controls infection.
A. Specimens
Diarrheal stool is the usual specimen. Other Campylobacters
Campylobacter species other than C jejuni are
B. Microscopy encoun­tered infrequently.
Gram-stained smears of stool may show the
typical “gull wing”—shaped rods. Dark-field or 1. Campylobacter fetus
phase contrast microscopy may show the typical Campylobacter fetus subspecies fetus is a very
darting or tumbling motility of the spiral rods. important veterinary pathogen. It causes infective
abortion in cattle and sheep.It is an opportunistic
C. Culture pathogen that causes systemic infections in
Feces or rectal swabs are plated on selective media. man in immuno­compromized patients. It may
A transport medium has to be employed in case of occasionally cause diar­rhea.
delay in culturing. Campylobacters survive for 1–2
weeks at 4°C in Cary–Blair transport medium but 2. C. concisus
glycerol-saline is not satisfactory. Selective media It has been isolated from cases of gingivitis and
for isolation of C. jejuni are But­zler’s selective periodontal disease. It has also been isolated from
medium, Skirrow’s Campylobacter selective faeces.
medium, Preston Campylobacter selec­tive medium C. fetus subsp. venerealis: It causes enzootic
and Blaser’s medium (Campy-BAP). Skirrow’s sterility (infectious infertility) of cattle but has not
medium contains vancomycin, polymyxin B, and been associated with human infection.
trimethoprim to inhibit growth of other bacteria.
Inoculated plates are incubated at 42°C to favor 3. C. jejuni subsp. doylei
growth of the ther­mophilic Campylobacters (C. This organism can be distinguished from other
jejuni, C. coli, C. lari and C. hyointestinalis) over that campylobacters because it does not reduce nitrate to
of other faecal bacteria. If, however, the presence of nitrite and hydrolyzes hippurate. The pathogenicity
C. fetus (nonthermophile) is suspected, additional of this organism is unknown. It has been isolated
plates should be incubated at 37°C to allow growth from human gastric epithelium biopsy and from
of this nonthermophile. Incubation must be done faeces of children with diarrhea.
in an atmosphere of 5% O2 10% CO2 and 85% N2.
Plates are incubated for 48 hours. 4. C. coli
Colonies are typically flat and effuse, with a It causes an infection clinically indistinguishable from
tendency to spread on moist agar. They are nonhe­ that of C. jejuni. It is commonly found in healthy pigs.
molytic, gray or colorless, moist, and flat or convex.
5. C. lari
D. Identification C. lari (formerly known as e. laridis). It causes
Suggestive colonies are screened by Gram staining, enteritis simulating C. jejuni infections in humans.
motility and oxidase tests. Confirmation is by
further biochemical tests, including positive 6. C. sputorum subsp. sputorum
catalase and nitrate reduction tests. It constitutes a part of normal flora of respiratory
tract and gingival crevices of man. It has also been
E. Serology isolated from feces of 2% of healthy people. It may
Complement fixation test and enzyme-linked occasionally cause diarrhea, abscess and septicemia.
immunosorbent assay (ELISA) are group-specific
tests that can detect recent infection with C. jejuni Helicobacter
or C. coli.
These are strict microaerophiles with a spiral or
Treatment helical morphology. They possess sheathed flagella.
Campylobacter gastroenteritis is typically a self- Species: Various species included in this genus
limited infection managed by the replacement of are: H. pyl­ori, H. cinaedi, H. fennelliae, H. canis,
Chapter 41: Campylobacter and Helicobacter  | 299
H pullorum H.rappininae and H. canadensis. Of vicinity of the organism, thus favoring bacterial
these, first three are medically important. multiplicatfun.
3. The bacterial antigens cross-react with antran
Helicobacter pylori gastric antigens stimulating an autoimmune
Warren and Marshall in Australia in 1983 observed response.
spiral, Campylobacter-like bacteria in close 4. Protease produced by the organism degrades
apposition to the gastric mucosa in several cases of gastric mucosa.
gastritis and peptic ulcer. They were originally named 5. H. pylori infected patients show hypergast­
Campylobacter pyloridis then C. pylori and now rinemia that upsets gastrin-HCI homeostasis.
redesignated as Helicobacter pylori. Features that Pathogenesis
distinguish this organism from Campylobacters are
its multiple sheathed flagella, its strong hydrolysis of H. pylori colonize the surface of the gastric
urea and its unique fatty acid profile. mu­cosa, especially of the antrum but any part of
the stomach may be colonized. Colonization often
extends into gastric glands, but the mucosa is not
Morphology
invaded by the bacteria.
H. pylori is a gram-negative spirally shaped Although gastric acid is potentially destructive
bacterium, 0.5–0.9 mm wide by 2–4 mm long. It is to H. pylori, protection is provided by its powerful
motile by means of a tuft of sheathed unipolar flagella, urease. H. pylori produce potent urease activity,
unlike the unsheathed flagella of campylobac­ters. It which yields production of ammonia and further
is non-sporing. buffering of acid and neutralizes acid around the
bacteria. Where they are numerous, the underlying
Cultural Characteristics mucosa usually shows a superficial gastritis of the
Like campylobacters, H. pylori is microaerophic. type known as chronic active or type B gastritis.
The optimum temperature for its growth is 35–37°C, H. pylori are associated with antral gastritis,
some grow poorly at 42°C but none grows at 25°C. It duodenal (peptic) ulcer disease, gastric ulcers.
can grow in an atmosphere of 5% O2, 10% CO2 and It is also recognized as a risk factor for gastric
85% N2. It does not grow anaerobically or in air. It malignancies, namely, ‘adenocarcinoma’ and
can be grown on moist freshly prepared chocolate ‘mucosa associated lymphoid tissue’ (MALT)
agar and Skirrow’s Campylobacter selective lymphomas.
medium. H. pylori produce circular, convex and
translucent colonies after incubation at 35–37°C in Laboratory Diagnosis
a microaerophilic atmosphere for 3–5 days. Diagnostic tests are of two kinds:

Biochemical Reactions A. Noninvasive tests


In practice, noninvasive tests are used for initial
H. pylori givs the following reactions.
screening.
i. Abundant urease production: A distinctive
1. Serology: Antibodies to H. pylori or its products
feature is the production of abundant urease,
can be detected in the patient serum by ELISA
and this property has been used as a rapid
test.
diagnostic test in gastric biopsy samples. The
2. Urea breath test: Urea tagged with an isotope
urease enzyme produced by H. pylori is almost
of carbon (carbon-14 or -13) is fed to the
100 times more active than that of Proteus
patient. If the patient’s stomach is colonized
vulgaris.
with H. pylori, urea is converted into ammonia
ii. It produces oxidase, catalase, phosphatase
and tagged CO2. The latter appears in the breath
and H2S.
where it can be measured. Patients infected
iii. It does not metabolize carbohydrates or
with H. pylori give high read­ings of the isotope.
reduce nitrate.
3. Fecal antigen test: In this polyclonal antibodies
are used to detect H. pylori antigens in feces.
Virulence Factors 4. Polymerase chain reaction (PCR): Various
1. H. pylori produce a cytotoxin causing DNA probes have been developed for the direct
vacuolation of gastric mucosa. Its production detection of H. pylori by PCR in gastric juice,
is determined by Cag A Gene. Virulence has feces, dental plaque and water supplies.
been associated with certain alleles in genes,
such as cag (cytotoxin associated gene) and B. Invasive Tests
vac (vacuolating cytotoxin gene). Specimens: Endoscopic biopsy of gastric mucosa
2. Urease released by H. pylori produces ammonia for examination by microscopy, culture and urease
ions that neutralize stomach acid in the tests.

www.ebook3000.com
300  |  Section 3: Systemic Bacteriology
1. Microscopy: The biopsy specimen can be
™™ Helicobacter pylori: Curved gram-negative bacilli.
examined by microscopic examination of Urease production at very high levels is typical of
Gram’s staining, silver staining, hematoxylin gastric helicobacters (e.g., H. pylori)
and eosin (Hand E) staining, Giemsa staining or ™™ Diseases–H. pylori causes gastritis, peptic ulcers,
immunofluorescence for the presence of bacteria. gastric adenocarcinoma
Warthin-Starry silver stain is the most sensitive. ™™ Diagnosis: A. Noninvasive tests: 1. Serology 2. Urea
2. Culture: Culture is done on nonselective breath test. 3. Faecal antigen test 4. Polymerase chain
medium such as chocolate agar and a selective reaction (PCR) B. Invasive tests: 1. Microscopy 2.
medium Skirrow’s campylobacter selective Culture 3. Biopsy urease test.
medium. Plates are incubated for 2–7 days in a
moist, microaerophilic atmosphere at 35–37°C Important questions
in the presence of 5–10% CO2.High humidity
is essential. The organism is identi­fied on the 1. Discuss pathogenesis and laboratory diagnosis of
basis of its colonial morphology, Gram staining, diarrhea caused by Campylobacter.
biochemical properties and positive urease tests. 2. Write short notes on:
3. Biopsy urease test a. Campylobaeter infections
The biopsy urease test can be performed by b. Helicobacter pylori
c. Helicobacter pylori infections.
crushing biopsy tissue in 0.5 mL urea solution with an
d. Laboratory diagnosis of Helicobacter pylori in-
indicator and incubated at 37°C. If H. pylori is present,
fections
the pH changes within a few minutes to 2 hours due e. Urea breath test.
to the production of ammonia. The abundance of
urease produced by the or­ganism permits detection
of the alkaline byproduct in less than 2 hours.
Multiple choice questions
1. Most common Campylobacter species known to
Treatment cause diarrhea in humans is:
The standard treatment is a combination of a. Campylobacter lari
bismuth subsalicylate, tetracycline (or amoxicillin) b. Campylobacter fetus
c. Campylobacter jejuni
and metronidazole for two weeks. An alternative
d. Campylobacter coli
schedule employs a proton pump inhibitor like
2. All the following media are commonly used for
omeprazole and clarithromycin. cultivation of Campylobacter except:
Prevention and control: Prophylactic treatment a. Butzler medium
of colonized individuals has not been useful b. Skirrow’s medium
and potentially has adverse effects, such as c. Preston’s Campylobacter selective medium
predisposing patients to adenocarcinomas of d. TCBS medium
the lower esophagus. Human vaccines are not 3. Which of the following bacteria is/are microaerophilic?
currently available. a. Campylobacter jejuni
b. Helicobacter pylori
Helicobacter cinaedi c. Mycobacterium bovis
d. All of the above
H. cinaedi (formerly known as Campylobacter 4. Campylobacter and Helicobacter can be different­
cinaedi) has been associated with proctitis in iated on the basis of:
homosexual men. a. Multiple sheathed flagella
b. Strong hydrolysis of urea
Helicobacter fennelliae c. Both of the above
H.fennelliae (formerly known as Campylobacter d. None of the above
fennelliae) like H. cinaedi has been associated with 5. Helicobacter pylori have been implicated in all of
proctitis in homosexual men. the following clinical conditions except
a. Chronic gastritis
b. Peptic ulcer disease
Key Points
c. Septicemia
™™ Campylobacters are thin, curved gram-negative d. Idiopathic thrombocytopenic purpura
bacilli. They are are micro­a erobic and strongly 6. Which of the following tests is/are useful in
oxidase positive identification of Helicobacter pylori?
™™ Campylobacter jejuni and Campylobacter coli have a. Warthin-starry silver staining
emerged as common human pathogens b. Biopsy urease test
™™ Diseasess—Zoonotic infection; improperly prepared c. Urea breath test
poultry is a common source of human infections d. All of the above
™™ Diagnosis—Microscopy is insensitive. Culture
Answers (MCQs)
requires use of specialized media incubated
1. c; 2. d; 3. d; 4. c; 5. c; 6. d
42
Chapter
Pseudomonas,
Stenotrophomonas, Burkholderia

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe the following: Pigments of Pseudomonas;
be able to: pathogenicity of Pseudomonas aeruginosa;
∙∙ Describe morphology, cultural characteristics, Burkholderia mallei.
biochemical reactions and laboratory diagnosis of
Pseudomonas aeruginosa

Introduction of temperatures, 6–­42°C, the optimum being 37°C.


Optimum pH 7.4–7.6. It grows well on ordinary
The term Pseudomonads describes a large group media and in the laboratory, it can be isolated on
of aerobic, nonfermentative, nonspor­ing gram- virtually any medium.
negative bacilli, motile by polar flagella. They 1. Nutrient agar: After aerobic incubation on
belong to over 100 species that were originally nutrient agar at 37°C for 24 hours, the colonies are
con­tained within the genus Pseudomonas. Most large, 2–3 mm in diameter, smooth, translucent,
are sapro­phytes found widely in soil, water and irregularly round and emit a characteristic fruity
other moist environments. odor. (Fig. 42.1). This grape-like smell is due to
Pseudomonas aeruginosa is the most common the produc­tion of aminoacetophenone from
pseudomonad and this is the species most com­ tryptophan.
monly associated with human disease. Burkholderia 2. It grows on MacConkey and DCA media,
(previously Pseudomonas) pseudomallei is an forming nonlactose-fermenting colonies.
important pathogen in tropical areas of the 3. Blood agar: Colonies on blood agar may be
world. Burkholderia (previously Pseudomonas) surrounded by a zone of hemolysis.
cepacia, which is now encompassed within the 4. In broth, it forms a dense turbidity with a surface
B. cepacia complex, has emerged as an important pellicle.
pathogen in immunocom­p romised patients,
particularly in individuals with cystic fibrosis or
chronic granulomatous disease. Stenotrophomonas
maltophilia also infects immuno­compromised
patients.

Pseudomonas aeruginosa
Morphology
It is a slender gram-negative bacillus, 1.5–3 μm ×
0.5 μm, actively motile usually with a single polar
flagel­lum. Occasional strains have two or three
flagella. It is nonsporing, noncapsulated but many
strains have a mucoid slime layer.

Cultural Characteristics
It is a strict aerobe but can grow anaerobically if Fig. 42.1: Pseudomonas aeruginosa on nutrient
nitrate is available. Growth occurs at a wide range agar

www.ebook3000.com
302  |  Section 3: Systemic Bacteriology
5. Cetrimide agar is selective medium for Ps. Epidemiological Typing Methods
aeruginosa 1. Serotyping: Identification of group-specific
heat-stable lipopolysaccharide antigens by
Pigment Production agglutination forms the basis of O serotyping.
P. aeruginosa produces at least 4 distinct pigments: Typically, nine serotypes ac­count for over 90%
i. Pyocyanin: It is a bluish-green phenazine of isolates.
pigment soluble in chloroform and water. This 2. Bacteriocin (pyocin) typing: Three types
pigment is not produced by other species of of bacteriocins (pyocins) are produced by P.
this genus. Demonstration of the presence aeruginosa which are known as R, F and S.
of the blue phenazine pigment pyocyanin Depending upon the growth inhibition of these
is absolute confirmation of a strain as P. 13 indicator strains, 105 types are recognized.
aeruginosa. Pyocin typing is easy to perform, results are
ii. Pyoverdin (fluorescein): The yellow/green available by the third day and has a reasonable
pigment pyoverdin (fluorescein) is also reproducibility and good discrimination.
produced by most strains, giving the charac­ 3. Phage typing: In bacteriophage typing
teristic blue-green appearance of infected pus considerable difficulties have been encounterd.
or cultures. It is insoluble in chloroform but 4. Molecular methods: Restriction endonuclease
soluble in water. typing wih pulsed field gel electrophoresis
iii. Pyorubrin: It is a bright red water soluble (PFGE) is the most reliable typing method
pigment and is insoluble in chloroform. and discriminatory of the present DNA-based
iv. Pyomelanin: It is a brown to black pigment typing methods and is considered to be the gold
and its production is uncommon. standard.

Biochemical Reactions Virulence Factors


Most strains produce two exotoxins, exotoxin A and
Ps. aeruginosa derives energy from carbohy­
exoenzyme S, and a variety of cytotoxic substances
drates by an oxidative rather than a fermentative
including proteases, phospholipases; rhamnolipids
metabolism. Special media, such as the O–F
and the blue-green pigment pyocyanin; an
medium of Hugh and Leifson must be used for
alginate-like exopolysaccharide.
diagnostic tests.
1. Capsule: P. aeruginosa produces a polysacch­
It utilizes glucose oxidatively with the pro­
aride capsule (also known as mucoid exopoly­
duction of acid only. Lactose and maltose are not
saccharide, alginate coat, or glycocalyx) that
utilized. Indole, MR, VP and H2S tests are negative.
has multiple functions and is responsible for
However, all strains give a rapid positive
the mucoid pheno­type.
oxidase reaction (within 30 seconds) and utilize
2. Pili: Adherence of P. aeruginosa to host cells is
citrate as sole source of carbon.
mediated by pili and nonpilus adhesins.
It reduces nitrates to nit­rites and further to
3. Lipopolysaccharide (LPS): It has endotoxin
gaseous nitrogen.
activity.
It is catalase, arginine dihydrolase and
4. Pyocyanin: It mediates tissue damage through
gelatinase positive and lysine decarboxylase and
and also stimulates the inflam­matory response
aesculin hydrolysis negative.
and impairs ciliary function.
5. Exotoxin A and Exotoxin S: Mechanism
Antigenic Characteristics of action of exotoxin A is iden­tical to that of
O antigens: P. aeruginosa possesses 19 distinct, diphtheria toxin. Exotoxin S Inhibits protein
group-specific 0 antigens. O antigens are heat- synthesis and is immunosuppressive.
stable. 6. Extracellular enzymes and hemolysins:
H antigens: On the basis of slide agglutina­tion, at P. aeruginosa produces proteases (general
least two heat-labile H antigens have been recog­ pro­t ease, alkaline protease and elastase),
nized. hemolysins (phospholipase C and heat-stable

O–F medium Glucose Lactose Maltose Mannitol Sucrose


of Hugh and A – – – –
Leifson—
Oxidative
Citrate Indole, MR, H2S NO3
+ VP – reduction
– – – –
O–F, oxidative–fermentative
Chapter 42: Pseudomonas, Stenotrophomonas, Burkholderia  | 303
rhamnolipid) and lipase. These play a key role following catheterization infection in burns
in the formation of local lesions. iatrogenic meningi­t is following lumbar
7. Antibiotic resistance: P. aeruginosa is inherently puncture and post-tracheostomy pulmonary
resistant to many antibiotics and can mutate to infection.
even more resistant strains during therapy. 2. Septicemia and endocarditis.
3. Ecthyma gangreno­sum and many other types
Resistance of skin lesions.
The bacillus is not particularly heat resist­ant, being 4. Infection of the nail bed.
killed at 55°C in one hour but exhibits a high degree 5. Infantile diarrhea and sepsis.
of resistance to chemical agents. It is resistant to 6. Shanghai fever: Ps. aeruginosa has been
the common antiseptics and disinfectants such reported to cause a self-limited febrile illness
as quaternary ammonium compounds, chloroxy­ (Shanghai fever) re­sembling typhoid fever in
lenol and hexachlorophene. It is sensitive to a some tropical areas.
2% aqueous alkaline solution of glutaraldehyde
7. Other infections: Including those localized
(cidex), acids, silver salts and strong phenolic
in the gastrointes­tinal tract, central nervous
disinfectants.
system, and musculoskeletal system.
Ps. aeruginosa possesses a considerable degree
of natural resistance to antibiotics.
Laboratory Diagnosis
Epidemiology 1. Specimens: Pus, wound swab, urine, sputum,
Pseudomonads are opportunistic pathogens. CSF or blood.
Its ability to persist and multiply, particularly in 2. Microscopy: Gram-negative rods are often seen
moist envi­ronments and on moist equipment in smears.
(e.g. humidifiers) in hospital wards, bathrooms 3. Culture: They grow easily on common isolation
and kitchens, is of particular importance in cross- media such as blood agar and MacConkey
infection control. Pseudomonads are resistant to agar. It may be necessary to use selective
many antibiotics and disin­fectants. media, such as cetrimide agar for isolation
of P. aeruginosa from feces or other samples
Pathogenesis with mixed flora, such as wound swab. As Ps
P. aeruginosa has many virulence factors, including aeruginosa is a frequent contaminant, isolation
structural components, toxins, and enzymes (See of the bacillus from a specimen should not
under Virulence factors). P. aeruginosa produces always be taken as proof of its etiological
numerous toxins and extracellular products that role. Repeated isolations help to confirm the
promote local invasion and dissemination of the diagnosis.
organism. 4. Identification: The isolates are identified by
Individuals most at risk include those with their colonial morphology and biochemical
impaired immune defenses. A prior antibiotic characters. The colonial morphology (e.g.
therapy that eliminate normal flora can also colony size, hemolytic activity, pigmentation,
provide P. aeruginosa with increased access for odor) combined with the results of selected
colonizing tissue. rapid biochemical tests (e.g. positive oxidase
reaction) is sufficient for the preliminary
A. Community Infections identification of these isolates.
1. Otitis externa and varicose ulcers. 5. Typing method: Bio­chemical profiles, antibiotic
2. Corneal infec­tions resulting from contaminated susceptibility patterns, phage typing, production
contact lenses or other sourcesl. of pyocins, serologic typing, and the molecular
3. Jacuzzi rash or whirlpool rash: Recreational charac­terization of DNA or ribosomal RNA are
and occupational conditions associated with used for the specific classification of isolates for
pseudomonas infections include Jacuzzi epidemiologic pur­poses.
rash or whirlpool rash (an acute self-limiting 6. Antibiotic sensitivity tests: It is useful to select
folliculitis). out proper antibiotic as multiple reistance to
antibiotics is quite common in Ps. aeruginosa
4. Industrial eye injuries, which may lead to
panophthalmitis.
Treatment
B. Hospital Infections P. aeruginosa is intrinsically resistant to most
1. Localized lesions: Localized lesions are com­monly employed antimicrobial agents. Many
commonly infections of wounds and bed­ strains are, however, susceptible to carbenicillin,
sores, eye infections and urinary infections azlocillin, ticarcillin, cefotaxime, ceftazidime,

www.ebook3000.com
304  |  Section 3: Systemic Bacteriology
gentamicin and tobramycin. Ciprofloxacin exhibits 3. Septicemia, particularly in patients with
good activity against P. aeruginosa and penetrates contaminated intravascular catheters.
well into most tissues. 4. Endocarditis, especially in drug addicts,
pneumonitis, osteomyelitis, dermatitis and
Control wound infections
Prevention of Ps. aeruginosa cross infection in
Treatment
hospitals requires constant vigilance and strict
attention to asepsis. The inappro­priate use of B. cepacia is susceptible to trimethoprim-
broad-spectrum antibiotics should also be avoided. sulfamethoxazole.
Immunotherapy in human burns cases with
antiserum to Ps. aeruginosa may be useful. Vaccine Burkholderia mallei (formely
from appropriate types administered to high-risk Pseudomonas mallei)
patients provides protection against Pseudomonas
The bacillus was discovered by Loeffler and Schutz
sepsis.
(1882) from a horse dying of glanders. It is the
causative agent of glanders (malleus,in Latin),
Stenotrophomonas maltophilia a disease primarily of equine animals, such as
(Formerly Pseudo­monas maltophilia) horses, mules and asses—but capa­ble of being
It is responsible for infections in debili­tated patients transmitted to other animals and to human beings.
with impaired host defense mechanisms. The
spectrum of nosocomial infections with S. mal­tophilia Morphology
includes bacteremia, pneumonia, meningitis, Ps. mallei is a slender, nonmotile, gram-negative
wound infections, and urinary tract infections. bacillus, 2–5 μm × 0.5 μm staining irregularly and
of­ten giving a beaded appearance.
Burkholderia cepacia (formerly
Pseudomonas cepacia) Culture
Morphology It is an aerobe and facultative anaerobe, growing
on ordinary media. Colonies which are small and
B. cepacia is a slender, motile, gram-negative rod.
transluscent initially become yellowish and opaque
The bacillus accumulates poly-β-hydroxybutyrate
on aging. On potato, a characteristic am­ber, honey-
as granules, so stains irregularly.
like growth appears, becoming greenish yellow
resembling Ps. aeruginosa.
Culture
It can grow in many common disinfectants and can Biochemical Reactions
even use penicillin G as a sole source of carbon It is It is quite inactive biochemically, attacking only
aerobic and grows well on nutrient agar optimally glucose. It is the only nonmotile species in the
at 25–30°C. On prolonged incubation, colonies genus Pseudomonas.
become reddish-purple due to the formation of a
nondiffusible phen­azine. Animal Pathogenicity
The natural disease in equines occurs in two
Biochemical Reactions
forms—glanders and farcy.
lt is weak oxidase-positive. It utilizes glucose, 1. Glanders: In glanders, respiratory system is
maltose, lactose, and mannitol and is lysine affected. The infected animal develops profuse
decarboxylase positive, ornithine decarboxylase catarrhal discharge from the nose and the nasal
positive, and arginine dihydrolase negative. Isolates septum shows nodule formation. Later the
are motile by means of polar tuft of flagella. nodules break down with the production of
irregular ulcers.
Pathogenicity 2. Farcy: This follows infection through skin with
Like P. aeruginosa, B. cepacia can colonize a variety involvement of superficial lymph vessels and
of moist environmental surfaces and is commonly lymph nodes. The lymph vessels are thick­ened
asso­ciated with nosocomial infections. and stand out as hard cords under the skin which
Infections caused by this organism include the are called ‘farcy pipes’
following: Strauss reaction: Guinea-pigs are susceptible
1. Respiratory tract infections in patients with and intraperitoneal injection into male guinea
cystic fibrosis or chronic granulomatous disease. pigs induces the Strauss reaction. This consists of
2. Urinary tract infections in catheterized patients. swelling of the testes, inflammation of tunica vaginal
Chapter 42: Pseudomonas, Stenotrophomonas, Burkholderia  | 305
and ulceration of the scrotal skin. The Strauss Toxins
reaction is not diagnostic of glanders, as it may Two thermolabile exotoxins, one lethal and the
also be produced by inoculation of other bacteria other necrotizing have been identified in culture
such as Brucella species, Preisz–Nocard bacillus filtrates.
Actinobacillus lignieresii and Ps. pseudomallei.

Human Pathogenicity Pathogenesis


Humans may become infected via skin abra­sions or Human infection is mainly acquired cutaneously
wounds which come into contact with the discharges through skin abrasions or by inhalation of
of a sick animal. Human disease may take the form of contaminated particles. It may also get transmitted
an acute fulminant febrile illness or a chronic indo­ from the animals by the bite of hematophagous
lent infection producing abscesses in the respiratory insects. Agriculture work­e rs, especially those
tract or skin. The fatality rate is high. Laboratory who work in moist soil (e.g. paddy fields), are
cultures are highly infectious and Ps. mallei is one of particularly prone to infection.
the most dangerous bacteria to work with. The disease may take one of four forms: acute,
subacute, chronic, or latent. It may be an acute
Laboratory Diagnosis septicemia with involvement of many organs, a
The diagnosis is based on: subacute typhoid like disease, or pneumonia and
1. Culture of the organism from local lesions of hemoptysis resembling tuberculosis. In chronic
humans or horses. form, there may be multiple caseous or suppurative
2. Rising agglutin titers. foci, with abscess formation in the skin and
subcutaneous tissues, bones and internal organs.
Mallein Test Acute melioidosis has a high case fatality rate.
Suppurative parotitis is a characteristic
Animals suffering from glanders develop a delayed
presentation of melioidosis in children.
hypersensitivity to the bacterial protein. This is
the basis of the mallein test used for diagnosing In India, cases of melioidosis have been repor­
glanders. This is analogous to the tuberculin test ted from Maharashtra, Kerala, Tamil Nadu, Orissa,
and may be performed by the subcutaneous, West Bengal and Tripura.
intracutaneous or conjunctival methods.
Laboratory Diagnosis
Burkholderia pseudomallei 1. Microscopy: A Gram stain of an appropriate
Burkholderia pseudomallei (formerly Pseudomonas specimen will show small gram-negative bacilli;
pseudomallei, also known as W hitmore’s bipolar staining (safety pin appearance) is
bacillus, Actinobacillus whitmori, Malleomyces seen with Wright’ stain or methylene blue stain.
pseudomallei, Loefflerella pseudomallei). 2. Culture: The organism may be observed may
B. pseudomallei is a saprophyte found in soil, be cultured from sputum, urine, pus or blood
water, and vegetation. It is endemic in Southeast on selective media. The organism is highly
Asia, India, Africa, and Australia. This is the causative infectious.
agent of melioidosis, a glanders-like disease, (The 3. Serology: Enzyme-linked immunosorbent
name is derived from ‘melis’, a disease of asses— assay (ELlSA) for detection of bacterial antigen,
glanders, and eidos, meaning resemblance). This specific IgG and IgM antibody to B. pseudoma­llei,
organism was isolated by Whitmore and Krish­ as well as an indirect hemagglutination test.
naswami (1912) from a glanders-like disease of man 4. Polymerase chain reaction (PCR).
in Rangoon.
Glucose Nonfermenters
Morphology The heterogeneous group of aerobic gram-negative
It resembles Ps. mallei but differs in being motile. bacilli commonly referred to as glucose nonfermenters
is taxonomically distinct from the carbohydrate
Culture fermenting Enterobacteriaceae and the oxidative
B. pseudomallei is easily cultured and produces pseu­domonads. They grow easily on common
charac­teristic wrinkled colonies after several days culture media, but unequivocal identification may
of growth on nutrient agar. Fresh cultures emit a be difficult as most species are relatively inert in the
characteristic pungent odor of putrefaction. biochemical tests used in identification of gram-
negative bacteria. Their clinical relevance is based
Biochemical Reactions on their role as opportunistic pathogens in hospital-
It resembles Ps. mallei but differs in liquefying acquired infec­tions and their intrinsic resistance
gelatin and forming acid from several sugars. to many antimicrobial agents. Susceptibility

www.ebook3000.com
306  |  Section 3: Systemic Bacteriology
Table 42.1  Some characteristics of nonfermenters
Organisms Oxidase Habitat Pathogenicity
test
Acinetobacter spp. _ Saprophytes found in soil, water Opportunist pathogens, serious infections include
and sewage, and occasionally meningitis, pneumonia and septicemia
as commensals of moist areas of
human skin.
Alcaligenes spp. + Human feces UTI, wound infection
Achromobacter spp. + − CSOM, Postoperative meningitis
Flavobacterium spp. + Saprophyte of soil and mo­ist Opportunistic nosocomial infections, particularly in
envi­ronments infants and associated with meningitis
Eikenella spp. + Commensal of mucosal sur­faces Endocarditis, meningitis, pneumonia, and infec­t­
ions of wounds and various soft tissues.

to antibiotics of glucose nonfermenters is very e. Bacteriocins produced by Pseudomonas


variable, and treatment should be based on the aer­uginosa
results of laboratory tests. These are Acinetobacter, f. Burkholderia mallei
Alcaligenes and Achromobacter, Eikenella g. Pyocin typing
corrodens, Flavobacterium meningosepticum. h. Burkholderia pseudomallei
Some characteristics of these organisms are shown i. Stenotrophomonas maltophilia.
in Table 42.1.
Multiple Choice Questions (MCQs)
Key Points 1. All the following statements are true for Pseudo­
monas aeruginosa except that they:
Pseudomonas aeruginosa
a. Are catalase positive
™™ Small gram-negative bacilli. Nonfermenter
™™ It is a strict aerobe. It grows well on ordinary media
b. Are oxidase positive
and in the laboratory. Cetrimide agar is selective c. Are fermenters
medium for Ps. aeruginosa d. Reduce nitrates to nitrogen gas
™™ P. aeruginosa produces at least 4 distinct pigments: (i) 2. Which of the following pigments is diagnostic of
Pyocyanin, (ii) Pyoverdin (fluorescein), (iii) Pyorubrin, Pseudomonas aeruginosa?
(iv) Pyomelanin a. Pyocyanin b. Pyoverdin
™™ Diseases: Burn wound infections and other skin c. Pyomelanin d. Pyorubin
and soft tissue infections tract infections pulmonary 3. Which is the most popular method for typing of
infections external otitis eye infections, etc. Pseudomonas aeruginosa?
™™ Stenotrophomonas maltophilia. The spectrum of
a Pyocin typing
nosocomial infections with S. mal­tophilia includes
bacteremia, pneumonia, meningitis, wound b. Serotyping
infections, and urinary tract infections c. Bacteriophage typing
™™ Burkholderia cepacia: Diseases caused are respiratory d. Antibiogram typing
tract infections, particularly in patients with cystic 4. Which of the following infections can be caused by
fibrosis; uri­nary tract infections; septic arthritis; Pseudomonas aeruginosa?
peritonitis; septicemia; opportunistic infections a. Urinary tract infection
™™ Burkholderia pseudomallei: It causes asymptomatic b. Wound and bum infection
colonization; cutaneous infection with regional c. Respiratory tract infection
lymphadenitis, fever, and malaise; pulmonary disease d. All of the above
ranging from bronchitis to necrotiz­ing pneumonia
5. All the following statements are true for Burkholderia
™™ Burkholderia mallei: Diseases caused is glanders in
livestock.
mallei except that they:
a. Cause glanders in horses
b. Cause farcy in horses
Important questions c. Are motile Gram-negative bacilli
d. Are usually sensitive to chloramphenicol
1. Describe the morphology, cultural characteristics, 6. Human melioidosis manifests as:
pathogenicity and laboratory diagnosis of a. Benign pulmonary infection
Pseudomonas aeruginosa. b. Multiple abscesses in various organs and tissues
2. Write short notes on: c. A fulminating septicemia
a. Pseudomonas aeruginosa. d. All of the above
b. Pigments of Pseudomonas.
c. Pathogenicity of Pseudomonas aeruginosa Answers (MCQs)
d. Enzymes and toxins of Pseudomonas aeru­ginosa 1. c; 2. a; 3. a; 4. d; 5. d; 6. d
4343
Chapter

Legionella

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe the following: Legionella pneumophilia;
be able to: diseases caused by Legionella.
∙∙ Describe morphology, culture characteristics and
biochemical reactions of Legionella

Introduction Culture
In the summer of 1976, public attention was Legionellae are nutritionally fastidious. Their
focused on an outbreak of severe pneumonia that growth is enhanced with iron salts and depends
caused many deaths in members of the American on the supple­mentation of media the L-cysteine.
Legion convention in Philadelphia. The disease They have fastidious requirements and grow on
was characterized by fever, cough and chest pain, complex media such as buffered charcoal, yeast
leading on to pneumonia and often ending fatally. extract (BCYE) agar, with L-cysteine and antibiotic
The causative agent has been called Legionella supplements, with 5% CO2, at pH 6.9, 35°C and 90%
pneumophila. Subsequent studies found this humidity. Growth is slow and colonies take 3–6
organism to be the cause of multi­ple epidemic and days to appear.
sporadic infections. It is now recognized to be a
ubiquitous aquatic saprophyte. Biochemical Reactions
The organisms are nonfermentative. Most species
Species: Taxonomic studies have shown that are motile and catalase-positive, liquefy gelatin,
the family Le­gionellaceae consists of one genus, and do not reduce nitrate or hydrolyze urea.
LegionelIa, with 40 species and more than 60
serogroups. The original isolate in this genus is Epidemiology
designated L. pneumophila serogroup 1 (SG1),
The bacteria are commonly present in natural
which accounts for nearly all severe infections.
bodies of water, such as lakes and streams, as well
Examples of other species that cause human
as in air conditioning cooling towers and condens­
infection less often are L. micdadei, L. bozemanii,
ers and in water systems (e.g. showers, hot tubs).
L. dumoffii and L. gormanii.
Legionellae survive and multiply inside free-living
amebae and other protozoa. They also multiply in
LEGIONELLA PNEUMOPHILA some artificial aquatic environments, which serve
Morphology as amplifiers.
Legionellae are thin, noncapsulated bacilli, 25 mm Human infection is typically by inhalation of
× 0.3–0.1 mm coccobacillary in clinical material aerosols produced by cooling towers, air conditioners
and assuming longer forms in culture. Most are and shower heads which act as disseminators. Man-
motile with polar or subpolar flagella. They are to-man transmission does not occur.
gram-negative but stain poorly, particularly in
smears from clinical specimens. They stain better Pathogenesis
by silver impregnation, but are best visualized by Respiratory tract disease caused by Legionella
direct fluorescent antibody (DFA) staining with species develops in susceptible people who inhale
monoclonal or polyclonal sera. infectious aerosols.

www.ebook3000.com
308  |  Section 3: Systemic Bacteriology
Clinical Diseases Treatment
Asymptomatic Legionella infections are relatively For treatment, the newer macrolides,ciprofloxacin,
com­mon. Symptomatic infections primarily affect and tetracyclines are effective. Rifampicin is
the lungs and present in one of two forms: employed in severe cases.
1. Pontiac fever: An influenza-like illness
2. Legionnaires’ disease: A severe form of Prevention
pneumonia Prevention of legionellosis requires identification of
1. Pontiac fever: Pontiac fever is a milder, nonfatal the environmental source of the organism and reduc­
‘influenza-like’ illness with fever, chills, myalgia tion of the microbial burden. Hyperchlorination of
malaise, and headache but no clinical evidence the water supply and the maintenance of elevated
of pneumonia. Outbreaks with high attack rates water temperatures have proved moderately
may occur. successful. How­e ver, complete elimination of
2. Legionnaire’s disease: The incubation period Legionella organisms from a water supply is often
is 2–10 days. ‘The disease presents with fever, difficult or impossible. Because the organism
nonproductive cough and dyspnea, rapidly has a low potential for causing disease, reducing
progressing, if untreated, to pneumonia. Multi­ the number of organisms in the water supply is
or­gan disease involving the gastrointestinal frequently an adequate control measure. Hospitals
tract, central nervous system, liver and kidneys with patients at high risk for disease should monitor
is common. their water supply on a regular basis for the presence
Case fatality may be 15–20%. All age groups are of Legionella and their hospital population for
susceptible, though more cases have occurred in disease.
the elderly. Key Points
™™ Legionella pneumophilia serogroup 1 is most
Laboratory Diagnosis common human pathogen
1. Microscopy ™™ L. pneumophilia are small, slender, pleomorphic,
Gram-negative bacilli
Legionellae in clinical specimens stain poorly ™™ Human infection is typically by inhalation of aerosols
with Gram stain. Nonspecific staining methods, produced by cooling towers, air conditioners and
such as those using Dieterle’s silver or Gimenez’s shower heads
stain, can be used to visualize the organisms. ™™ L. pneumophilia causes Legionnaire’s disease
The most sensitive way of detecting legionellae ™™ Laboratory diagnosis depends on direct fluorescent
antibody (DFA) test, culture, detection of antigen and
microscopically in clinical specimens is to use the demonstration of serum antibodies
di­rect fluorescent antibody (DFA) test, in which ™™ Hyperchlorination of the water and superheating of
fluores­cein-labeled monoclonal or polyclonal water is of value in preventing the disease.
antibodies directed against Legionella species are
used.
Important questions
2. Culture 1. Discuss pathogenicity and laboratory diagnosis of
The medium most commonly used for the isolation Legionnaire’s disease.
2. Write short notes on:
of legionellae is buffered charcoal-yeast extract
a. Legionella pneumophilia
(BCYE) agar. Anti­biotics can be added to suppress b. Legionnaire’s disease
the growth of rapidly growing contaminating c. Pontiac fever.
bacteria. Legionellae grow in air or 3% to 5% carbon
dioxide at 35°C after 3–5 days. Their small (1–3 mm) Multiple choice question (mCQs)
colonies have a ground­glass appearance. 1. The causative agent of Pontiac fever is:
a. Legionella pneumophila
3. Antigen Detection b. Pseudomonas putida
c. Yersinia pseudotuberculosis
Enzyme-linked immunoassays, radioimmuno­
d. Francisella tularensis
assays, the agglutination of antibody-coated latex 2. The natural habitat of Legionella pneumophila is:
particles, and nucleic acid analysis studies have a. Water b. Sputum
all been used to detect legionellae in respiratory c. Faeces d. Urine
specimens and urine. 3. The antibiotic of choice in Legionella infections is:
a. Erythromycin b. Cephalosporins
4. Serology c. Penicillins d. All of the above
Detection of serum antibody is done by ELISA or Answers (MCQs)
indirect immunofluorescent assay. 1. a; 2. a; 3. a
44
Chapter

Yersinia, Pasteurella, Francisella

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe laboratory diagnosis of plague
be able to: ∙∙ Describe the following: prophylaxis against plague;
∙∙ Describe morphology, culture characteristics and Yersinia enterocolitica; Yersinia pseudotuberculosis;
biochemical reactions of Yersinia pestis Pasteurella multocida; Francisella tularensis.

Introduction Yersinia pestis (formerly


The organisms within these three genera (Yersinia, Pasteurella pestis)
Pasteurella, Francisella) are animal pathogens Morphology
that, under certain conditions, are transmissible to
Y. pestis is a gram-negative, short, oval cocco­bacilli
man either directly, or indirectly through food and
with rounded ends and convex sides, about 1.5
water or via insect vectors. They are gram-negative
× 0.7 µm, occurring singly, in short chains or in
coccobacilli, formerly contained within one genus,
small groups. In smears stained with Giemsa or
Pasteurella. Molecular genetics has indicated
methylene blue, it shows bipolar staining (safety
a com­p letely separate identity for the three
pin appearance) with the two ends densely
genera—Yersinia, Pasteurella and Francisella.
stained and the central area clear (Fig. 44.1).
Genus Yersinia: It was so named after Alexandre Pleo­morphism is marked in culture, especially
Yersin, who had isolated the plague bacillus in 1894. in old cultures, involution forms are seen-coccoid,
Yersinia belongs to the family Enterobacteriaceae club saped, filamentous and giant forms. This
and the tribe Yersinieae The genus Yersinia involution in culture can be hastened by the
currently consists of 11 named species. addition of 3% NaCI.
Medically important species are:
Y. pes­tis (the causative agent of plague);
Y. pseudotuber­culosis (a primary pathogen of
rodents);
Y. enterocolitica (which causes enteric and
systemic disease in animals and human beings).
Genus Pasteurella: The genus Pasteurella is
now restricted to a number of animal pathogens
and contains several related bacteria causing
hemorrhagic septicemia in different species of
ani­mals and occasionally producing local and
systemic infections in human beings, grouped
under a com­mon species named P. multocida.
Genus Francisella: The third new genus Francisella,
consisting of F. tularensis, is named after Francis for Fig. 44.1: Smear from gland puncture in a case of plague
his pioneering studies on tularemia caused by this showing Y. pestis with bipolar staining (safety pin appearance),
bacillus. a few red blood cell and leukocytes

www.ebook3000.com
310  |  Section 3: Systemic Bacteriology
The bacillus is surrounded by a slime layer Biochemical Reactions
(enve­lope or capsule). It is nonmotile, nonsporing It ferments glucose, mannitol and maltose with the
and non­acid fast. production of acid but no gas. Lactose and sucrose
or rhamnose are not fer­mented.
Cultural Characteristics It is catalase positive, indole negative, MR
The plague bacillus is aerobe and facultative positive, VP and citrate nega­tive (IMViC – + – –),
anaerobe. Growth occurs over a wide range of pH nitrate reduction positive, aesculin positive and
5–9.6 (optimum pH 7.2). Optimum temperature oxidase and urease negative. Gelatin is not liquefied.
for primary culture is 27°C (range 14–37°C). Several
phenotypic characteristics are best expressed at room Physiological varieties of Y. pestis: Devignat has
temperature but the envelope develops at 37o C. distinguished three physiological varieties of Y.
1. Nutrient agar: Colonies are small (as small as pestis based on the fermentation of glycerol and
0.1–0.2 mm), delicate, transparent disks and reduction of nitrate. These biotypes have been
become opaque on continued incubation. designated orientalis, medieval and antigua, and
2. Blood agar: Colonies are dark brown due to are characterized by differences in their geo­
absorption of the haemin pigment. graphic distribution. This typing appears to be of
3. DCA and MacConkey agar: On DCA and epidemiological significance because of the different
MacConkey agar it grows poorly, producing pin- geographical distribution of the types (Table 44.1).
point, reddish colonies after 24 hours incubation.
4. In broth, a flocculent growth occurs at the Resistance
bottom and along the sides of the tube, with little
The plague bacillus is easily destroyed by exposure
or no turbidity. A delicate pellicle may form later.
to heat, sunlight, drying and chemical disin­
5. Ghee broth: If grown in a flask of broth with
fectants. It is killed by moist heat at 55°C in 5
sterile oil or ghee (clarified butter) floated
min and by 0.5% phenol in 15 minutes. It is very
on top (ghee broth) a charac­teristic growth
susceptible to drying. It remains viable for long
occurs which hangs down into the broth from
peri­ods in cold, moist environments.
the surface, resembling stalactites (stalac­tite
growth) (Fig. 44.2).
Antigens, Toxins and
Other Virulence Factors
At least 20 different antigens have been detected
in Y. pestis.
1. F-l antigen or envelope antigen: The heat-
labile Fraction I (FI) protein capsular antigen
is a soluble antigen contained within the
bacterial envelope. It helps the organism to
resist phagocytosis.
2. V and W antigens: These antigens have been
considered to be the virulence factors as they
inhibit phagocytosis. Production of V and W
antigens is plasmid mediated.
3. Pigment binding and iron-regulated surface
proteins: In yersiniae, avirulent and low-
pathogenicity strains require iron overload to
produce septicemia in humans or lethality in
Fig. 44.2: Y. pestis in ghee broth culture. Stalactite growth mice.

Table 44.1  Biotypes of Yersinia pestis


Variety Glycerol Nitrate Geographical distribution
fermentation reduction
Y. pestis var. orientalis − + Primary foci in India, Myanmar, and China. Causative agent of 1894
pandemic. Responsible for wild plague in Western USA, South
America, South Africa
Y. pestis var. antiqua + + Transbaikalia, Mongolia, Manchuria, perhaps responsible for
Justinian plague
Y. pestis var. medievalis + − Southeast Russia
Chapter 44: Yersinia, Pasteurella, Francisella  | 311
4. Pesticin I, coagulase, and plasminogen proventriculus. When such a ‘blocked flea’ bites
activator: Pesticin I is a bacteriocin produced another rodent, it cannot suck in blood because the
by Y. pestis. It is thought that these enzymes, bacterial mass blocks the passage mechanically.
especially the plasminogen activator, may The blood, mixed with the bacteria is regurgitated
contribute to the highly invasive fulminant into the bite, transmitting the infection. Infection
character of the disease. may also be trans­ferred by contamination of the
5. Other virulence-associated factors bite wound with the feces of infected fleas.
a. Endotoxin: The role of Y. pestis endotoxin Forms of human plague: Traditionally, three
in the disease process is ill-defined. severe forms of human plague are described. All
b. Murine toxins: They are thermolabile and of these may occur at different stages in the same
may be toxoided but do not diffuse freely patient.
into the medium and are released only by
the lysis of the cell. They are called ‘murine 1. Bubonic Plague
toxins’ as they are active in rats and mice.
The role of plague toxins in natu­ral disease Incubation period is 2–5 days. As the plague
in human beings is not known. bacillus usually enters through the bite of infected
c. pH 6 Ag: The pH 6 Ag is synthesized by flea on the legs, the inguinal lymph nodes are invol­
Y. pestis at a temperature of 37°C and at ved, hence the name bubonic plague (bubo means
pH levels similar to those in macrophage enlarged gland in groin). The glands become enlar­
phagolysosomes or abscesses. ged and suppurate. The bubo may be preceded
6. Purine synthesis: Virulence has also been by prodromata of chills, fever, malaise, confusion,
associated with the ability for purine synthesis. nausea, and pains in the limbs and back. The bacilli
enter the bloodstream and produce septicemia.
The case fatality in unteated cases may be 30–90%.
Plague
Plague is one of the oldest recorded infectious 2. Pneumonic Plague
diseases and is an ancient scourge of mankind.
This can develop in patients presenting with
Central Asia is believed to have been the original
bubonic or septicaemic plague. It may also be
home of plague. More than 150 epidemics, most of
acquired as a primary infection by inhalation
them associated with three main pandemics, have
of droplets infected with Y. pestis. The bacilli
been reponed.
spread through the lungs producing hemorrhagic
The second pandemic, known as the Black
pneumonia. The sputum becomes thin and blood-
Death started in the fourteenth century. The third
stained.
pandemic originated in Burma, spread to China
This type of plague is highly contagious and
in 1894, and from Hong Kong was carried to other
is almost invariably fatal unless treated very early
continents. India was one of the countries worst hit
almost 100% and with treatment it is 5–30%.
by this pandemic. Plague reached Bombay in 1896
and spread all over the country during the next few
years, causing more than 10 million deaths by 1918. 3. Septicemic Plague
It grad­ually receded thereafter, though occasional This may occur as a primary infection or as a
cases con­tinued to occur in endemic foci till 1967. complication of bubonic or pneumonic plague.
No further plague cases were seen in India till 1994, Purpura may develop in the skin, giving the skin
when in August a nonfatal outbreak of bubonic a blackish coloration which, in the past, led to the
plague was re­p orted from Maharashtra (Beed name “black death”. Disseminated
district). In Septem­ber pneumonic plague was intravascular coagulation is usually present.
reported in Surat and adjoining areas of Gujarat Meningitic involvement may occur rarely.
and Maharashtra. More recently as of 19th Feb.
2002, the Ministry of Health reported a total of 16 Epidemiology
cases of pneumonic plague (including 4 deaths) in Several species of fleas may act as vectors, the
Hat Koti village, Shimla Dist. Himachal Pradesh. most important being Xenopsylla cheopis. X.
astia and Ceratophyllus fasciatus. X. cheopis, the
Pathogenesis predominant species in north India is a more
Plague is a zoonotic disease. The plague bacillus efficient vector than the south Indian species X.
is naturally parasitic in rodents. Infec­t ion is astia.
transmitted among them by rat fleas. When a rat Plague is perpetuated by three cycles: (i) natural
flea, commonly X cheopsis, bites a diseased rat, foci among commensal rodents with transmission
it sucks blood. In the flea, the bacilli multiply in by fleas (sylvatic plague, wild plague), (ii) urban
the stomach to such an extent that they block the rat plague, which is transmitted by the rat flea

www.ebook3000.com
312  |  Section 3: Systemic Bacteriology
(domestic plague, urban plague), and (iii) human Postmortem examination shows a marked local
plague, which may be acquired by contact with inflammatory condition at the site of inocula­tion
either of the former cycles and which may be with necrosis and edema. The regional lymph
transmitted by pneumonic spread or, rarely, by the nodes are enlarged, the spleen is enlarged and
bite of a human flea. con­gested and may show grayish-white patches
In the 1990s, there has been a re-emergence in the tissue. Prepare films from the local lesions,
of plague in countries where it had ceased to be lymph nodes, spleen pulp and heart blood; stain
noticed for many years. Y. pestis has been employed and examine for characteristic plague bacilli.
as a biological warfare agent.
5. Antigen Detection
Laboratory Diagnosis Demonstration of the FI capsular antigen by
Plague is confirmed by demonstrating the bacilli immunospecific staining and ELISA test will
in fluid from buboes or local skin lesions, in the confirm the presence of Y. pestis.
sputum and in blood films. Blood culture may be Dipstick test: FI glycoprotein can be detected by
intermittently positive in all forms of the disease. dipstick test using monoclonal antibodies. It is
Postmortem, the bacilli can usually be isolated rapid test and produces reliable result within 15
from a wide range of tissues, especially spleen, lung minutes.
and lymph nodes.
6. Serology
1. Specimens
The antibodies to F-I antigen can be demonstrated
i. Bubonic plague—pus or fluid aspirated. in patient’s serum by complement fixation test
ii. Pneumonic plague—sputum and blood. (CFT), hemagglutination test and enzyme linked
iii. Septicemic plague—blood. immunosorbent assay (ELISA).
iv. Meningeal plague—cerebrospinal fluid (CSF).
v. On postmortem—splenic tissue.
7. Polymerase Chain Reaction (PCR)
2. Microscopy A polymerase chain reaction (PCR), with primers
based on FI gene sequences, offers a rapid and less
Smears of exudate or sputum are stained with hazardous means of diagnosis than culture.
methy­lene blue or Giemsa stain. Characteristic
gram-negative coccobacilli and bacilli showing
Diagnosis of Plague in Wild Rats
bipolar staining with methylene blue suggest
plague bacilli. The smears are also stained by Before examining rats that may have died as a result
Gram’s method and observed for Gram negative, of plague, immerse them in disinfectant to kill any
ovoid coccobacilli with bipolar bodies. The infected fleas. Necropsy has the following features
fluorescent anti­body technique may be of use in in plague:
identifying plaguqe bacilli. i. Enlargement of the lymphatic nodes with
periglandular inflamma­t ion and edema; ii.
3. Culture Pleural effusion; iii. Enlarge­ment of the spleen; iv.
Liver congested and mottled; v. Congestion and
Culture the samples on blood agar plates,
hemorrhages under the skin and in internal organs.
MacConkey agar, nutrient agar and ghee broth
and incu­bated at 27°C. Colonies on blood agar Make films and cultures of heart blood, lymph
are dark brown. Colonies on MacConkey agar are nodes and spleen. Inoculate material from lesions
colorless. In ghee broth, a characteristic stalactite on to the nasal mucosa of guinea pigs or white
growth is produced. The growth is identified by rats as with sputum. Examine stained films and
biochemical tests and slide agglutination tests. pure cultures for characteristic morphology and
Demonstration of the FI capsular antigen by biochemical reactions of Y. pestis.
immunospecific staining will confirm the presence
of Y. pestis. Prophylaxis
Plague is one of the internationally quarantinable
4. Animal Inoculation diseases, and reporting of cases is mandatory.
If exudate is inoculated subcutaneously into Prophylaxis can be carried out by:
guinea-pigs or white rats, or on to their nasal
A. General Measures
mucosa, infection follows and the animals die
within 2–5 days. The bacilli may then be isolated 1. Control of fleas and rodents.
from the blood or from smears of spleen tissue 2. Spray insecticide (DDT) inside the rodent,
taken postmortem. burrows and houses to kill the fleas.
Chapter 44: Yersinia, Pasteurella, Francisella  | 313
3. After the fleas have been killed, kill the rat with pseudotuberculosis and Y. enter­ocolitica. They are
rat-poison. found in the intestinal tract of a variety of animals,
in which they cause diseases, and are transmissible
B. Vaccination to humans, in which they produce a variety of
Two types of vaccine have been in use-killed and clinical syndromes. These are zoonotic diseases.
live attenuated vaccines. These are nonlactose fermenting gram-
negative rods that are urease-positive and oxidase-
1. Killed vaccine: The k­ illed vaccine used in India
negative. They resemble Y. pestis because they are
(prepared at the Haffkine Institute, Bombay) is
small, gram-negative rods with bipolar staining
a whole culture antigen. A virulent strain of the
and rodents, wild and domestic animals are
plague bacillus is grown in casein hydrolysate
reservoirs of infection. They differ from Y. pestis by
broth for 2–4 weeks at 32°C and killed by 0.05%
motility when grown at 22°C (nonmotile at 37°C),
formaldehyde and preserved with phenyl
non­capsulated, urease positive, oxidase-negative
mercuric nitrate (Sokhey’s modification of
and insusceptible to Y. pestis bacteriophage.
Haffkine’s vaccine). In Asia, Haffkine Institute,
Mumbai is the only vaccine producing center.
Yersinia pseudotuberculosis
Vaccine dose schedule: The vaccine is given
subcutaneously, two doses at an interval of 1–3 Y. pseudotuberculosis is a small, oval, Gram-
months, followed by a third six months later. negative,bipolar-stained bacillus. It is non-sporing,
Vaccination can confer significant protection non-capsulated and slightly acid-fast.
against bubonic but not pneumonic plague. The The organisms may be differentiated from
protection does not last for more than six months. Y. pestis by: by its relatively poor growth on
MacConkey agar.
Side effects: Fever, headache, malaise, lym­ ∙∙ Motility when grown at 22°C (but not at 37°C)
phadenopathy, and erythema and induration at ∙∙ Ability to produce urease
the site of inoculation. It may cause fetal damage ∙∙ Fermentation of rhamnose and melibiose
and abortion in pregnant women. Vaccination ∙∙ Failure to be lysed by the antiplague bac­
is not very effective, therefore, even vaccinated teriophage at 22°C.
individuals should also be given chemoprophylaxis ∙∙ Lack of the FI antigen as shown by immunospecific
when exposed to plague. staining or PCR.
2. Live attenuated vaccines: Live vaccines are There are eight major O serotypes, several of
prepared from two avirulent strains of Y. pestis, which can be separated into subtypes. The majority
Otten’s Tjiwidej strain from Jawa and Girard’s of human cases of Y. pseudotuberculosis infection
EV 76 strain from Malagasey. Killed vaccine are due to serotype 1.
is recommended for general use because
live vaccines are difficult to prepare and may Pathogenesis
provoke unacceptable reactions. Live vaccines
Animal infection: It causes pseudotuberculosis
are not in use now.
which is a zoonotic diseses. In animals, the natural
mode of infection is by gastrointestinal tract. In
Chemoprophylaxis infected guinea pigs, the liver, spleen and lungs
A person exposed to definite risk of infec­tion, show multiple nodules resembling tuberculosis
whether vaccinated or not, should be given lesions (hence the name pseudotuberculosis).
chemoprophylaxis-cotrimoxazole or tetracycline
Human infection: Human infection probably
orally for at least five days. Close contacts of
results from ingestion of materials contaminated
patients with plague should be given a course
with animal feces. Infection may be subclinical, but
of tetracycline (500 mg 6 hourly for one week).
occasionally results in a severe typhoid-like illness
with fever, purpura and enlargement of the liver
Treatment and spleen, which is usually fatal. More frequently
Streptomycin is the drug of choice. Chloramphenicol it causes mesenteric lymphade­nitis and terminal
is recommended in patients with meningitic ileitis simulating acute or subacute appen­dicitis.
symptoms. Tetracycline may be adequate in It has also been reported to cause immunological
uncompli­cated bubonic plague. Gentamicin and sequelae such as erythema nodosum or reactive
ciprofloxacin are also effective. arthritis in some patients.

Yersiniosis Laboratory Diagnosis


The term yersiniosis denotes infection with Laboratory diagnosis may be made by isolation
Yersinia species other than Y. pestis, namely, Y. of the organ­ism in culture from blood, local

www.ebook3000.com
314  |  Section 3: Systemic Bacteriology
lesions or mesenteric nodes, or demonstration by demonstration of antbodies in the patient
of antibodies in patient serum during the acute serum. Isolation of the organisms can be done
phase of the illness by tube agglutination tests, from feces, blood or from mesenteric lymph
hemag­glutination of red cells sensitized with nodes. Blood agar and MacConkey agar are used
LPS, or ELISA­. for growing this organism. Selective medium for
recovery of Y. enterocolitica from faeces, food
Treatment soil etc. is cefsulodin-irgasan novobiocin (CIN)
Ileitis and mesenteric lymphadenitis are usually Yersinia selective agar. Colonies of Y. enterocolitica
self-limiting but septice­m ia may be treated have a bull’s eye appearance, colored dark red
with parenteral ampicillin, chloramphenicol, and surrounded by a transparent border on CIN
gentamicin or tetracycline. agar. Identify the strain by biochemical tests and
serotype determination.
Yesinia enterocolitica
It is a gram-negative coccobacillus showing Pasteurella multocida (formerly
pleomorphism in older cultures. This bacillus Pasteurella septica)­
resembles Y. pseudotuberculosis in being motile at
22°C but differs from it in fermenting sucrose and A group of related bacteria isolated from hemor­
cellobiose and decarboxylating ornithine. It does rhagic septicemia in a variety of animals and birds
not ferment rhamnose or melibiose. Many strains had, in the past, been named according to their
are indole and VP positive. spe­cies of origin—Pasterella boviseptica. Pasterella
lepiseptica, Pasterella avisepti­ca, etc. Though they
Cultural Characters show some degree of host specificity, they are so
It is aerobe and facultative anaerobe. Optimum alike in other respects that they are now considered
temperature for growth is 22–29°C. On blood strains of a single species designated P. multocida.
agar, it forms nonhemolytic, smooth, translucent
colonies, 2–3 mm in diameter in 48 hours. On Morphology
MacConkey medium, it forms pinpoint non- They are gram-negative, nonmotile, nonsporing
lactose fermenting (NLF) colonies. coccobacilli which show bipolar staining. Some
strains are encapsulated.
Antigenic Structure
Based on O and H antigens, more than 60 different Cultural Characteristics
O antigens and 19 H factors have been identified. Pasteurellae are aerobes and facultative anaerobes.
Serotypes O3, O8 and O9 account for most human They grow on blood agar and chocolate agar.
infections.

Pathogenesis Pathogenesis
The bacillus is usually normal inhabitant of the
Y. enterocolitica has been isolated from a wide
upper respira­tory tract of a variety of animals
range of domestic and wild animals. Human
such as dogs, cats, cattle and sheep. Pasteurella
disease usuaIly results from ingestion of contam­
infections are considered zoonoses. The most
inated food or from contact with the environment.
common presentation is a history of animal bite.
Blood transfusion is a significant hazard.
Human infections usually present as:
Types of Disease in Humans 1. A local abscess at the site of a cat or dog bite;
2. Infections of the respiratory system.
1. Gastroenteritis or enterocolitis, which is self-
3. Meningitis or cerebral abscess (usually follow­
limited and occurs in young children.
ing head injury), endo­carditis, pericarditis or
2. Mesenteric lymphadenitis and terminal ileitis
septicemia.
in older children that may mimic appendicitis
3. Septicemia, which is often fatal. Animals and birds: P. multocida can be extremely
4. Pneumonia and meningitis are rare present­ virulent to many species of animals and birds,
ations. causing fowl cholera; hemorrhagic septicemia. It
5. Post­infectious complications include erythema respiratory infections and atrophic rhinitis in pigs.
nodosum, polyarthritis, Reiter’s syndrome and
thyroiditis. Laboratory Diagnosis
1. Culture: Swabs from bite wounds, from blood,
Laboratory Diagnosis from CSF in cases of meningitis and from
The laboratory diagnosis of Y. enterocolitica secretions in suppurative conditions of the
consists of isolation of the organism and indirectly respiratory tract are cultured on blood agar
Chapter 44: Yersinia, Pasteurella, Francisella  | 315
plates incubated at 37°C for 24 hours. The Prophylaxis
organisms are identified by various cultural and A vaccine based on the live-attenuated LVS strain
biochemical tests. confers some protection. It can be administerd by
2. Serology: Serology is of no value in diagnosis. scarification to persons who are subject to high
3. Polymerasrs chain reaction (PCR): PCR is risk of infection.
potentially useful but rarely available. F. tularensis has been developed as a biological
warfare agent and has potential application in
Francisella tularensis bioterrorism.
(Pasteurella tularensis, Brucella Key Points
tularensis) ™™ Yersinia are gram-negative bacilli that are facultative
Francisella tularensis produces tularaemia in man anaerobes, positive by the catalase test and negative
by the oxidase test
and certain small mammals, notably rabbits, hares,
™™ The genus Yersinia consists of 10 species, with Y. pestis,
beavers and various rodent species. Tularemia was Yersinia enterocolitica, and Yersinia pseudotuberculosis
originally described in Tulare county, California. It the well-known human pathogens
can be transmitted by direct contact, by biting flies, ™™ Y. pestis is a gram-negative, shows bipolar staining
mosquitoes and ticks, by contaminated water or (safety pin appearance), pleo­morphic, nonmotile,
meat, or aerosols. and is capsulated
™™ F-l antigen or envelope antigen has been considered
a virulence determinant
Morphology ™™ Plague is a zoonotic infection in humans. Disease is
It is a very small, nonmotile, non-sporing, capsulate, spread by flea bites or direct contact with infected
tissues or person-to-person by inhala­tion of infectious
Gram- negative coccobacillus, about 0.3–0.7 µm ×
aerosols from a patient with pulmonary disease
0.2 µm in size. ™™ Diseases: Yersinia pestis causes plague, which
manifests in one of three forms: Bubonic plague;
Cultural Characters pneumonic plague; septicemic plague
™™ Diagnosis: For diagnosis, material is aspirated
F. tularensis is strictly aerobic. It will not grow on from the bubo, for demonstration of bipolar
ordinary nutrient media, but grows well on blood staining short rods (coccobacilli) using Wright’s or
agar containing 2.5% glucose and 0.1% cysteine Gram stain, for demonstration of the bacteria by
hydrochlo­r ide. Minute droplet-like colonies immunofluorescence. Culture of the bacteria on
blood agar. Serology can be used to detect antibody
develop in 72 hours. to capsular antigen
™™ Yersinia pseudotuberculosis: It causes pseudotuber­
Pathogenesis culosis, a zoonotic disease
Yesinia enterocolitica
The infection, which is a typical zoonosis, is mainly
™™ It produces in humans-gastroenteritis or enterocol­
spread by insects or ticks among lagomorphs and itis, mesenteric lymphadenitis and terminal ileitis
rodents. It is transmitted to man through handling in older children, septicemia, pneumonia and
of infected animals. meningitis, erythema nodosum, polyarthritis, Reiter’s
In human beings, tularemia may present as a syndrome and thyroiditis
local ulceration with lymphadenitis, a typhoid like Pasteurella multocida
fever with glandular enlargement or an influenza ™™ Pasteurella infections are considered zoonoses
like respiratory infection. Francisella tularensis
™™ Francisella tularensis produces tularemia in man and
certain small mammals. The infection, is a typical
Laboratory Diagnosis zoonosis.
Diagnosis may be made by culture or by inoculation
into guinea pigs or mice. A PCR has been Important questions
described, but is not widely available. Serology is
most likely to be positive after 3 weeks. Rising titers 1. Describe the pathogenesis and laboratory diagnosis
of plague.
of agglutinins to F. tularensis or individual titers
2. Write short notes on:
of 160 are diagnostic. An intradermal delayed
a. Prophylaxis against plague
hypersensitivity test has been used in the past but
b. Yersinia pseudouberculosis
the antigen is not readily available.
c. Yersinia enterocolitica
d. Pasteurellosis
Treatment e. Pasteurella multocida
Streptomycin or gentamicin are the antibiotics of f. Francisella tularensis
choice in tularemia and are usually curative. g. Tularemia

www.ebook3000.com
316  |  Section 3: Systemic Bacteriology
Multiple choice questions (Mcqs) 5. Which of the following vaccines is/are recommended
for immunization against plague?
1. Bipolar staining is characteristic of: a. Live b. Killed
a. Yersinia pestis c. Subunit d. All of the above
b. Yersinia enterocolitica 6. Yersinia enterocolitica shows all the following
c. Yersinia pseudotuberculosis characteristics except:
d. Proteus mirabilis a. They are motile at 22°C
2. Bubonic plague is transmitted by: b. They produce hemolytic colonies on blood agar
a. Inoculation b. Inhalation c. They ferment sucrose and cellobiose
c. Ingestion d. All of the above d. They decarboxylate ornithine
routes 7. Which of the following diseases is/are caused by Y.
3. Pneumonic plague is transmitted to humans by: enterocolitica in man?
a. Rat flea b. Droplet infection a. Gastroenteritis
c. Ingestion d. Inoculation b. Mesenteric lymphadenitis
c. Septicemia
4. The name black death is given to which of the d. All of the above
following diseases?
a. Tuberculosis b. Diphtheria Answers (MCQs)
c. Plague d. AIDS 1. a; 2. a; 3. b; 4. c; 5. b; 6. c; 7. d
45
Chapter

Haemophilus

Learning Objectives

After reading and studying this chapter, you should ∙∙ Discuss laboratory diagnosis of infections caused
be able to: by Haemophilus influenzae
∙∙ Describe morphology and culture characteristics ∙∙ Discuss Haemophilus influenzae biogroup aegyptius
of Haemophilus influenzae ∙∙ Describe morphology and culture characteristics of
∙∙ Describe the following: X and V factors; Satellitism; Haemophilus ducreyi
Antigenic structure of Haemophilus influenzae ∙∙ Discuss Haemophilus ducreyi or chancroid or soft
∙∙ Describe pathogenicity of H. influenzae sore and its laboratory diagnosis.

Introduction Table 45.1  Growth characteristics of Haemophilus


species
The genus Hemophilus comprises a group of
small, nonmotile, nonsporing, non-acid-fast, Growth Hemolysis on
gram-negative coccobacilli or rods, often markedly Species requirements horse blood
pleo­morphic and sometimes filamentous that are X V CO2 agar
parasitic on human beings or animals. They are H. influenzae + + − −
characterized by their requirement of one or both H. aegyptius + + − −
of two accessory growth factors (X and V) present
H. ducreyi + − Variable Variable
in blood. The name of the genus comes from the
requirement by these organisms for accessory H. parainfluenzae − + − −
growth factors found in blood, that is, haemo H. haemolyticus + + − −
(Greek for blood) and philos (Greek for loving). H. parahaemolyticus − + − −
Species: Haemophilus influenzae-most commonly H. aphrophilus + − − −
associated with disease H. paraphrophilus − + − −
Haemophilus ducreyi
Haemophilus aphrophilus: The other members
of the genus are commonly isolated in clinical
specimens but are rarely pathogenic, being Cultural Characteristics
re­sponsible primarily for opportunistic infections H.influenzae is an aerobe and facultative anaerobe.
(Table 45.1). Growth is enhanced by a moist atmosphere
supplemented with 5–10% CO2. The optimum
Haemophilus influenzae
temperature is 37°C. The accessory growth factors;
Morphology named X and V, present in blood are essential for
H. influenzae is a small, gram-negative rods or growth.
coccobacilli (0.3–0.5 × 1–2 mm size), exhibiting X factor: X factor (hemin) is a heat-stable protopor­
considerable pleomorphism. Cells from young phyrin IX, haemin or some other iron-containing
cultures (18–24 hours) are usually cocco­bacillary, porphyrin. It is required for the synthesis of the
while older cultures are distinctly pleomor­ iron-containing respiratory enzymes cytochrome
phic. Virulent strains are often capsulated. It is c, cytochrome oxidase, peroxidase and catalase.
nonmotile, nonsporing and non-acid-fast. The X factor is not required for anaerobic growth.

www.ebook3000.com
318  |  Section 3: Systemic Bacteriology
V factor: V factor is coenzyme, nicotinamide This is a routine test in clinical bac­teriology for the
adenine dinucleotide (NAD), NAD phosphate identification of H. influenzae.
(NADP) which acts as a hydrogen acceptor in
the metabolism of the cell. It is heat labile being Biochemical Reactions
destroyed at 120°C in a few minutes. It is present in H. influenzae ferments glucose and galactose but
red blood cells and in many other animal and plant does not ferment sucrose, lactose and mannitol.
cells. It is synthesized by some fungi and bacteria Catalase and oxidase reactions are positive. It
such as Staph. aureus.The differential requirement reduces nitrates to nitrites. H. influenzae can
of X and V factors help to differentiate various be divided into eight biotypes on the basis of
species of haemophilus (Table 45.1). indole pro­duction, urease activity and ornithine
The growth is scanty on blood agar because V decarboxylase reactions (Table 45.2). Biotypes
factor is present inside the RBC. Chocolate agar I-III are the most common, and biotype I is most
(heated blood agar) is superior to plain blood frequently responsible for meningitis.
agar for the growth of H. influenzae, because V
factor is released from within the erythrocytes
and inactivates serum NADase. Clear transparent Antigenic Structure
media may be prepared by boiling and filtering a There are three major surface antigens:
mixture of blood and nutrient broth (Levinthal’s 1. Capsular antigens: The major antigenic
medium) or by adding a peptic digest of blood to determinant of capsulated strains is the capsular
nutrient agar (Fildes agar). polysaccharide based on which H. influenzae
Colonies: The colonies are small, translucent and strains have been classified by Pittman into six
nonhemolytic on blood agar. Capsulated strains capsular types—types a to f. Typing was orig­inally
produce larger, distinctive iridescent colonies. done by agglutination but other methods such
Fildes agar is best for primary isolation of H. as Quellung reaction (swelling of the capsule)
influenzae and gives a copious growth. Capsulated with type-specific antisera, precipitation,
strains produce translucent colonies with a coagglutination, counterimmunoelectrophoresis
distinctive iridescence on Levinthal’s agar. (CIE) and enzyme-linked immunosorbent assay
(ELISA) may also be used. Diagnostic kits for the
Satellitism: The V factor is intracellular and
identification of H. influenzae type b (Hib) are
is present inside the RBC. It is synthesized by
com­mercially available.
some fungi and bacteria such as Staph. aureus
The type b capsular polysaccharide has
in excess of their requirements and released into
a unique chemical structure, containing the
the surrounding medium. When Staph. aureus
pentose sugars ribose and ribitol. The capsular
is streaked across a plate of blood agar on which
polyribosyl ribitol phosphate (PRP) antigen of
a spec­imen containing H. influenzae has been
Hib in­duces IgG, IgM and IgA antibodies which
inoculated, af­ter overnight incubation, the colonies
are bacteri­cidal, opsonic and protective. Hib
of H. influenzae will be large and well developed
PRP is, therefore, employed for immunization.
alongside the streak of Staphylococcus, and
H. influenzae strains lacking a capsule cannot
smaller farther away. This phenomenon is called
be typed and are called ‘nontypable strains’.
satellitism and demonstrates the dependence of H.
influenzae on the V factor, which is available in high 2. Somatic antigen: The cell envelope of H. influenzae
concentrations near the staphylococcal growth and consists of an outer and inner membrane
only in smaller quantities away from it (Fig. 45.1). containing protein and lipooligosaccharide
(LOS) antigens. Outer membrane protein (OMP)
antigens of Hib have been classified into at least
13 subtypes. OMP and LOS subtyping may be of
epidemiological value.

Table 45.2  Characters of six biotypes of Haemo­


philus influenzae
Biotype
Character I II III IV V VI VII VIII
Indole + + − − + − + −
production
Urease activity + + + + − − − −
Ornithine + − − + + + − −
Fig. 45.1: Satellitism. H. influenzae colonies are large near decarboxylase
growth of Staphylococcus, and smaller away from it
Chapter 45: Haemophilus  | 319
Resistance always asso­ciated with bacteremia and blood
H. influenzae is a delicate organism. It is readily cultures are usually positive.
killed by moist heat (at 55°C in 30 minutes), refrige­ 3. Pneumonia : Haemophilus pneumonia
ration (4°C), drying and disinfectants. It also dies typically occurs in infants and is accompanied
in 1–48 hours in dried secretions and airborne by empyema and sometimes meningitis as
drop­let-nuclei. In culture, the cells die within two well. While these are primary infections due
or three days due to autolysis. to capsulated strains, bronchop­n eumonia
may occur as a secondary infection with the
noncapsulated strains.
Variation
4. Suppurtive lesions: Suppurative lesions such
Colonies of H. influenzae show a smooth to rough as arthritis, endocarditis and pericarditis may
(S–R) variation associated with loss of capsule and result from hematogenous dissemination.
virulence. As in case of Strepto­coccus pneumoniae, 5. Cellulitis: Cellulitis partic­ularly in the buccal
genetic transformation has been demonstrated in and periorbital areas is seen in young children.
H. influenzae also. The char­acters transformed are
the capsular antigens and antibiotic resistance. B. Noninvasive Disease
1. Acute sinusitis and otitis media
Virulence Factors 2. Acute exacerbations of chronic obstructive
The virulence of H. influenzae is determined by the airway disease
following factors: 3. Conjunctivitis
1. Capsular polysaccharide: This confers on the
bacterium ability to resist phagocytosis. Epidemiology
2. Adherence: Most noncapsulate strains are Infection is transmitted by the respiratory route.
adherent to human epithelial cells and may Carriage in the upper respiratory tract is common
explain the tendency of these strains to cause particularly in young children. The frequency of
more localized infections. Most serotype b strains invasive infections is inversely related to age; only
are not adherent and explain the tendency for a small percentage occurs in adults and older
type b strains to cause systemic infections. children. Infections in the first 2 months of life are
3. Outer membrane proteins: They also contribute rare, probably because of transplacental transfer
in adhesion and invasion of host tissues. of maternal antibody.
4. IgAl protease: Production of IgA1 protease
allows pathogens to inactivate the primary Laboratory Diagnosis
antibody found on mucosal surfaces and 1. Specimens
thereby elimi­nate protection of the host by the The following specimens may be collected
antibody. depending upon the type of lesion:
i. Blood culture; ii. cerebrospinal fluid; iii. throat
Pathogenesis swabs; iv. sputum; v. pus; vi. aspirates from joints,
H. influenzae is exclusively a human parasite, middle ears or sinuses, etc.
which resides principally in the upper respiratory
tract. Capsulate strains are present in 5–10%.
2. Collection and Transport
H. influenzae causes invasive and non- For optimal yield, specimens should be transported to
invasive infections. the laboratory and seeded on to appropriate cuIture
media without delay. As haemophili are poorly viable
A. Invasive Infections in clinical specimens particularly at 4°C, therefore, the
specimens should never be refrigerated.
1. Meningitis: The most serious of the diseases
produced by H. influenzae is an acute bacterial 3. Direct Microscopy
meningitis. The bacilli reach the meninges
i. Gram-stained smear
from the nasopharynx, apparently through the
Gram-stained smear of clinical material show­ing
bloodstream. The disease is more common in
poorly stained gram-negative coccobacilli and
children between two ‘months and three years
occasionally slender filamentous forms.
of age. Case fatality rates are up to 90% in the
untreated. Majority of the cases are due to type b ii. Immunofluorescence and Quellung reaction
strains. Immunofluorescence and Quellung reaction
2. Epiglottitis: This is an acute inflam­mation of can be employed for direct demonstration of
the epiglottis, with obstructive laryngitis, seen H. influenzae after mixing with specific rabbit
in children above two years. This condition is antiserum type b.

www.ebook3000.com
320  |  Section 3: Systemic Bacteriology
iii. Antigen detection due to the production of β-lactamase. Amoxycillin-
Type b capsular antigen can be detected in patient clavulanate or clarithromycin is more effective.
serum, urine, CSF or pus by several methods.
These include latex agglutination, coagglutination, Prophylaxis
countercurrent immunoelectrophoresis radio­
Active Immunization
immunoassay (RIA) enzyme linked immonosorbent
assay (ELISA). i. A purified type b capsular polysaccharide
vaccine
4. Culture It is used in children of 18–24 months. Vaccine
is administered in two doses at an interval of
i. CSF culture: CSF should be plated promptly
two months.
on blood agar or chocolate agar. A strain of S.
ii. Conjugate vaccines: Hib PRP vaccine in which
aureus should be streaked across the blood
the polysaccharide is cova­lently coupled
agar plate on which the specimen has already
to various proteins (e.g. tetanus toxoid,
been inoculated. Plate is then incubated
Neisseria meningi­t idis outer membrane
at 37°C with 5–10% CO2 and high humidity
protein, and diphtheria toxoid) produce a
overnight. It may also be cultured on Levinthal
lasting anamnestic response. Such Hib PRP
and Fildes blood-digest agar The growth
are available for use in young children.
is identified by colony morphology, Gram
staining, satellite phenomenon, biochemical iii. Household contacts of patients
reactions and serotyping. Rifampicin given for four days pre­v ents
secondary infection in contacts and also
ii. Blood culture: Blood cultures are often
eradi­cates carrier state.
positive in cases of epiglottitis and pneumonia.
iii. Sputum culture: Sputum should be homo­
genized by treatment with pancreatin or by
Haemophili other than H. influenzae
shak­ing with sterile water and glass beads Haemophilus influenzae Biogroup Aegyptius
for 15–30 minutes. The rate of isolation is (Koch-Weeks Bacillus, Formerly H. Aegypticus)
increased by culturing several samples of
sputum from the patient. Koch (1883) observed a small bacillus in conjunc­
tivitis cases in Egypt. It was first cultivated by Weeks
(1887) in New York.This organism, formerly known
5. Identification
as the Koch-Weeks bacillus and H. aegyptius, is
H. influenzae colonies have a characteristic seminal now thought to be a subgroup of H. influenzae.
odour. Confirmation of the identity depends on It is indistinguishable from H. influenzae
demonstrating a requirement for one or both of the biotype III in routine tests, but can be identified
growth factors, X and V H. influenzae requires both. by a PCR method.

6. Molecular Techniques Diseases: It causes purulent conjunctivitis and


Brazilian purpuric fever (BPF), a clinical syn­
Polymerase chain reaction (PCR) is used to
drome which is characterized by conjunctivitis,
identify Haemophilus species in clinical specimens
high fever, vomiting, petechiae, purpura, septice­
and as confirmatory tests on isolates.
mia, and shock. It occurs in infants and children
with high fatality.
7. Antibiotic Sensitivity Tests
Accurate determination of the antibiotic sus­ Treatment
ceptibility of H influenzae requires careful
standardization of the methodology because of Ampicillin in combination with chlorampheni­
the fastidious nature of the organism. col has been successful when treatment has been
started sufficiently early.
Treatment
Haemophilus ducreyi
H. influenzae is susceptible to sulphonamides,
trimetho­prim, ampicillin, chloramphenicol, tetra­ Ducreyi (1890) demonstrated this bacillus in
cycline, co­amoxiclav, ciprofloxacin, cefuroxime, chancroid lesions.
cefotaxime and ceftazidime. Ceftriaxone and
related cephalosporins such as cefo­taxime are Morphology
the antibiotics of first choice for the treat­ment H. ducreyi is a gram-negative short, ovoid bacillus
of meningitis. Cefotaxime, cef­u roxime and (1–1.5 um × 0.6 mm) with a tendency to occur in
ceftazidime are highly effective for the treatment of end to end pairs or short chains and frequently
H. influenzae infection. Ampicillin resistance is shows bipolar staining. The bacilli may be
Chapter 45: Haemophilus  | 321
arranged in small groups or whorls or in parallel iv. Agglutination: H. ducreyi is antigenically
chains giving a ‘school-of fish; or ‘rail road track’ homo­geneous and cultures are identified by
appearance. agglutination with the antiserum.

Cultural Characteristics Treatment


It requires X factor but not V factor for its growth. It Chancroid may be treated with azithromycin, ery­
can be grown on rabbit-blood agar, fresh clotted thromycin, ciprofloxacin or ceftriaxone.
rabbit blood or chocolate agar enriched with
1% Iso Vitalex, and containing vancomycin as Haemophilus parainfluenzae
a selective agent. It requires 10% CO2 and high It can be distinguished from H. influenzae by its
humidity for primary isolation. Cultures should be non-requirement of X factor, ability to ferment
incubated for up to 5 days at 35–37°C in a humid sucrose but not D-xylose, and low pathogenicity.
atmosphere with additional CO2. It may also be grown
H parainfluenzae is normally present as a com­
on chorioallantoic membrane of the chick embryo.
mensal in the mouth and throat.
The colonies of H. ducreyi are small, pinpoint
to 0.5 mm in diameter, nonmucoid, grey, yellow It may occasionally cause conjunctivitis, acute
or tan, translucent or semi­opaque after 24 hours pharyngitis, infective endocarditis, uretritis and
incubation. After 48–72 hours, the colonies are 1–­2 bronchopulmonary infections in patients with cystic
mm in diameter and semiopaque. fibrosis.

Identification Haemophilus haemolyticus


The species is antigenically homogeneous and This haemophilus has also been regarded as a
cultures may be identified by agglutination with the variety of H. influenzae, from which it differs in
antiserum. Intradermal inoculation of the culture forming a zone of β-hemolysis around its colonies
into rabbits produces a local ulcerative lesion on blood agar. The haemolyis is strongest on sheep
or ox blood, but is also seen on horse or human
Biochemical reactions: H. ducreyi is biochemically blood. Colonies on blood agar may be mistaken for
inert with the exception of positive nitrate reduction those of hemolytic strepto­cocci. It requires both X
test. and V factors.
It is found as a commensal in the throat, but not
Pathogenicity mouth, and appears to be non-pathogenic.
H. ducreyi is the etiologic agent of a highly com­ Strains that do not require the X factor have
municable sexually transmitted disease (STD) been designated H. parahaemolyticus.
chancroid or soft sore or soft chancre character­
ized by tender nonindurated irregular ulcers on
HACEK Group Bacteria
the genitalia. The lesions are generally on the penis
in male and they may be present on the labia and HACEK is an acronym consisting of the first initial
within the vagina in females. The infection remains of each genus represented in the group:
localized, spreading only to the inguinal lymph 1. Haemophilus species (parainfluenzae, aphro­
nodes which are enlarged and painful. philus, pamphrophilus)
There is no immunity following infection 2. Actinobacillus actinomycetemcomitans
but a hypersensitivity develops, which can be 3. Cardiobacterium hominis
demonstrated by the intradermal inoculation of 4. Eikenella corrodens
killed bacilli. 5. Kingella kingae.
The acronym HACEK refers to a group of
Laboratory Diagnosis fastidious slow growing bacteria, normally resident
i. Specimens: Take specimens from the base of in the mouth, which can sometimes cause severe
an ulcer or aspirate material from bubo. infections, particularly endocarditis. Members
ii. Direct microscopy: Typical small gram- of the HACEK group include both fermentative
negative bacilli can be seen in material from and nonfermentative, gram-negative bacilli.
the ulcers or in pus from lymph node aspirates. Members of this group of gram-negative bacilli
iii. Culture: Inoculate heated blood agar with 1% have in common the need for an environment with
Iso Vita­lex. Vancomy­cin 3 mg/L may be added increased CO2 (capnophilic).
to make the medium selective. Cultures should Blood cultures from HACEK patients take 7–30
be incubated for up to 5 days at 35–37°C in a days to become positive. Antibi­otic sensitivity tests
humid atmosphere with additional CO2 and are essential for effective therapy as drug resistance
look for characteristic colonies. is very common.

www.ebook3000.com
322  |  Section 3: Systemic Bacteriology

Key Points f. Haemophilus influenzae biogroup aegyptius


g. Haemophilus ducreyi or Chancroid or Soft sore.
™™ The genus Haemophilus comprises a group of small,
pleomorphic, gram-negative bacilli or coccobacilli.
Most species require X and/or V factor for growth.
Multiple choice questions (Mcqs)
Haemophilus influenzae and Haemophilus ducreyi are 1. Following statements are true for morphology of
important pahogens Haemophilus influenzae except:
Haemophillus influenzae a. It is a Gram-negative coccobacillus
™™ Satellitism: This phenomenon demonstrates the b. It rarely shows pleomorphism
dependence of H. influenzae on the V factor c. It is nonmotile
™™ Diseases: H. influenzae is responsible for meningitis, d. The capsule may be present
epiglottitis, cellulitis, arthritis, otitis, sinusitis, lower 2. All the following Haemophilus species require
respiratory tract disease, conjunctivitis both X and V factors for their growth except:
™™ Diagnosis: Microscopy is a sensitive test for detecting a. Haemophilus influenzae
H. infiuenzae in CSF, synovial fluid, and lower b. Haemophilus aegyptius
respiratory specimens. Culture is performed using c. Haemophilus haemolyticus
chocolate agar. Antigen detection: Type b capsular d. Haemophilus ducreyi
antigen can be detected several methods such as 3. Haemophilus influenzae shows the phenomenon
latex agglutination, coagglutination, countercurrent of satellitism on:
immunoelectrophoresis, RIA and ELISA a. Blood agar b. Chocolate agar
™™ Haemophilus influenzae biogroup aegyptius: c. Nutrient agar d. All of the above
causes purulent con­junctivitis and Brazilian purpuric 4. Chancroid is caused by:
feve (BPF) a. Haemophi/us injluenzae
™™ Haemophillus ducreyi: is the etiologic agent of a b. Haemophilus aegyptius
highly communicable sexually transmitted disease c. Haemophilus ducreyi
(STD chancroid or soft sore d. Haemophilus parainfluenzae
™™ Haemophilus haemolyticus: nonpathogenic. 5. Most common Pasteurella species causing human
infections is:
a. Pasteurella canis
Important questions b. Pasteurella multocida
c. Pasteurella bettyae
1. Discuss pathogenesis and laboratory diagnosis of
d. Pasteurella dagmatis
infections caused by Haemophilus influenzae.
6. Conjunctivitis is caused by:
2. Write short notes on: a. Haemophilus influenzae
a. Hemophilus influenzae b. Haemophilus aegyptius
b. X and V factors c. Haemophilus ducreyi
c. Satellitism. d. Haemophilus parainjluenzae
d. Antigenic structure of Haemophilus influenzae Answers (MCQs)
e. Virulence factors of Haemophilus influenzae 1. b; 2. d; 3. a; 4. c; 5. b; 6. b
46
Chapter

Bordetella

Learning Objectives

After reading and studying this chapter, you should ∙∙ Discuss pathogenesis of pertussis
be able to: ∙∙ Discuss laboratory diagnosis of pertussis
∙∙ Describe various factors which determine the ∙∙ Describe the following: Cough plate method;
virulence of Bordetella pertussis vaccination against pertussis
∙∙ Describe culture media for Bordetella pertussis

Introduction Cultural Characteristics


The genus Bordetella con­stitutes a group of minute, It is an obligate aerobe. The optimum temperature
gram-negative, non-acid-fast, non-sporing, for growth is 35–36°C. Complex media are necessary
coccobacilli, often described as parvobacteria. for primary isola­tion. The medium in common use
Species: The genus contains four species. is the Bordet–Gengou medium (potato-blood-
1. Bordetella pertussis: Causes whooping cough glycerol agar). Primary isolation of B. pertussis
(pertussis). requires the addition of charcoal, ion exchange
2. Bord. parapertussis can cause a similar res­ins, or 15–20% blood to neutralize growth-
disease and was isolated from mild cases of inhibit­ing effects.
whooping cough. The plates are incubated at 35–36°C in a moist
3. Bord. bronchiseptica: It may occasionally environment. After incubation for 48–72 hours,
infect human beings, producing a condi­tion colonies on Bordet–Gengou medium are small,
resembling pertussis dome shaped, smooth, opaque, viscid, greyish
4. Bard. avium causes respiratory disease in white, refractile and glistening, resembling
turkeys. ‘bisected pearls’ or ‘mercury drops’. Colonies are
5. Bord holmesii. surrounded by a hazy zone of hemolysis. Confluent
6. Bord hinzii. growth presents an ‘aluminium paint’ appearance.
7. Bord. trematum.
8. Bord. ansorpic. Biochemical Reactions
9. Bord. petrii. It is biochemically inactive. It does not ferment
sugars, form indole, reduce nitrates, utilize citrate
Bordetella pertussis or split urea (Table 46.1). It produces oxidase and
Morphology usually catalase also.
The bacteria are small, gram-negative coccobacilli Resistance
with slight pleomorphism measuring 0.2–0.3 µm by
B. pertussis is a delicate organism. It can be killed
0.5–1.0 µm. They appear singly, in pairs, and in small
by heating at 55°C for 30 minutes, drying and dis­
clusters. It is nonmo­tile and nonsporing. Bipolar
metachromatic staining may be demonstrated infectants. Outside the body it can survive for five days
with toluidine blue. Capsules may be demonstrable on glass, three days on cloth and a few hours on paper.
in young, freshly isolated cultures only by special
stains. In culture films, the bacilli tend to be arranged Antigenic Constituents and
in loose clumps, with clear spaces in between giving Virulence Factors
a ‘thumb print’ appearance. Freshly isolated strains B. pertussis produces a number of factors that are
of Bord. pertussis have fimbriae. involved in the pathogenesis of disease.

www.ebook3000.com
324  |  Section 3: Systemic Bacteriology
Table 46.1  Differentiatial characterstics of Bordetella species
Characteristics Bord. pertussis Bord. parapertussis Bord. bronchiseptica Bord. avium
1. Motility – – + +
2. Growth on:
Nutrient agar – + + +
Growth on Bordet- 3–6 1–2 1 1
Gengou medium (days)
MacConkey agar – + + +
3. Urease – – + +
4. Citrate utilization – V + +
5. Nitrate reduction – – + –
6. Toxins
Pertussis toxin + – – –
Adenylate cyclase toxin + + + –
Heat labile toxin + + + +
Tracheal cytotoxin + + + +
Lipopolysaccharide + + + +
7. Agglutinogens 1–7,13 7–10,14 7–13 Not known

Adhesins 5% of the cases and by Bord. bronchiseptica very


1. Agglutinogens: Bordetellae possess genus- infrequently (0.1%).
specific and species-specific surface 14 Stages of disease: In human beings, after an
agglutinogens associated with the capsular K incubation period of about 1–2 weeks, the disease
antigens or fimbriae. Agglutinogens promote takes a protracted course comprising three stages—
virul­e nce by helping bacteria to attach to the catarrhal, paroxysmal and convalescent—each
respiratory epithelial cells. They are useful lasting approximately two weeks.
in serotyping strains and in epidemiological 1. Prodromal or catarrhal stage: Clinical diagnosis
studies. Bordetellae are classified into various in the catarrhal stage is difficult.
types based on the agglutinogens they carry. 2. Paroxysmal stage: After 1–2 weeks, the
2. Pertussis toxin (PT): Pertussis toxin promotes paroxysmal stage begins. During the paroxysm,
lymphocytosis, sensitization to histamine, and the patient is subjected to violent spasms
enhanced insulin secretion. of continuous coughing, followed by a long
It can be toxoided. Pertussis toxoid is the major inrush of air into the almost empty lungs, with
component of acellular pertussis vaccines. a characteristic whoop (hence the name).
3. Filamentous hemagglutinin adhesin (FHA): 3. Convalescent stage: After 2–4 weeks, the
It is a protein that acquired its name through its paroxysmal stage is fol­lowed by convalescence
ability to agglutinate erythrocytes. It mediates stage, during which the frequency and severity
adhesion to ciliated epithelial cells. of coughing gradually decrease but secondary
4. Adenylate cyclase (AC): At least two types of complications can occur.
AC are known, only one of which has the ability
to enter target cells and act as a toxin. This is Complications
known as AC toxin (ACT).
5. Heat-labile toxin (HLT) or dermonecrotic 1. Subconjunctival hemorrhage, subcutane­ous
toxin: It is a heat-labile toxin its role in disease emphysema, inguinal hernia or rectal prolapse
is unknown. due to pressure effects during the violent bouts
6. Tracheal cytotoxin (TCT): Tracheal cytotoxin of coughing
has a specific affinity for ciliated epithelial cells. 2. Respiratory (bronchopneumonia, lung collapse).
It induces ciliary damage leading to severe 3. Neurological (convulsions, coma).
cough.
7. Lipopolysaccharide (Heat-stable toxin): Their Epidemiology
role in the disease process is unknown. Whooping cough is predominantly a pediatric
disease, the incidence and mortality being highest
Pathogenesis in the first year of life. The source of infection is the
Whooping cough is predominantly a pediatric patient in the early stage of the disease. Infection is
disease. Whooping cough in 95% of cases is caused transmitted by droplets and fomites contaminated
by Bord. pertussis. Bord. parapertussis causes about with oropharyngeal secretions. Natural infection
Chapter 46: Bordetella  | 325
confers protection though it may not be permanent, 5. Detection of Bacterial Antigens
and second attacks have been reported. Bordetella antigens may be detected in serum and
urine in tests with specific antiserum. Alternatively,
Laboratory Diagnosis bacteria in nasopharyngeal secretions are labeled
1. Microscopy with fluorescein-conjugated antiserum and
Microscopic diagnosis depends on demonstration examined by ultraviolet microscopy.
of the bacilli in respiratory secretions by the
fluores­cent antibody technique. 6. Polymerase Chain Reaction (PCR)
Polymerase chain reaction (PCR) is used for the
2. Specimen Collection and Transport detection of bordetella DNA in nasopharyngeal
Though the disease is mainly in the lower specimens.
respiratory tract, the organism can be recovered
7. Serology
readily from the nasopharynx. ‘Cough plates’ and
postnasal swabs are unsatisfactory because of Rise in antibody titer may be demonstrated in paired
overgrowth by commensal bacteria. serum samples by ELISA, agglutination, complement
i. Cough plate method: Here, a culture plate is fixation, immunoblotting, indirect hemagglutination,
held about 10–15 cm in front of the patient’s and toxin neutralization. Demonstration of specific
mouth during a bout of spontaneous or secretory IgA antibody in nasopharyngeal secretions
induced coughing so that droplets of respiratory by ELISA has been proposed as a diagnostic method
exudates impinge directly on the medium. This in culture negative cases.
has the advantage that specimen is directly
Treatment
inoculated at the bedside.
ii. Postnasal (peroral) swab: Secretions from Bord. pertussis is susceptible to several antibiotics
the posterior pharyngeal wall are collected (except penicillin). The drug of choice is erythro­
with a cotton swab on a bent wire passed mycin (or one of the newer macrolides such as
through the mouth. A West’s post­nasal swab darithromycin). Chloram­phenicol and cotrimoxazole
may be conveniently employed. Cotton swabs are also useful.
should not be used because they contain
Prophylaxis
fatty acids that are toxic to B. pertussis so it is
preferable to use dacron or calcium alginate Vaccination
swabs for specimen collection. Specific immunization with killed Bord. pertussis
iii. Pernasal swab: A sterile swab on a flexible vaccine has been found very effective. It is of
wire is passed gently along the floor of the utmost importance to use a smooth phase I strain
nose until it meets resistance. The swab, for vaccine production. The vaccines in general use
which will collect muco-pus, is withdrawn are suspensions of whole bacterial cells, killed by
and either plated immediately on charcoal heat or chemicals.
blood agar, or placed in transport medium. In India, National policy is to immunize against
diphtheria, whooping cough and tetanus (DPT)
3. Culture simultaneously, by administering 3 doses (each
The swab is inoculated imm­ediately on charcoal- dose 0.5 mL) of DPT vaccine intramuscularly, at 1–2
horse blood agar and Bordet–Gengou medium months interval, starting when the infant is about
both with and without methicillin or cephalexin 6 weeks old. A booster dose of DPT is indicated at
and incubated for at least seven days before being the age of 18–24 months. Bord. pertussis acts as an
discarded as neg­a tive. The speci­m en may be adjuvant for the toxoids (diphtheria and tetanus
transported in Regan–Lowe semiso­lid medium if toxoid) producing better antibody response.
delay in transport is unavoidable. Adverse reactions: Pertussis vaccination may
Plates are incu­bated in high humidity at 35–36°C induce reactions ranging from local soreness and
and colonies appear in 48–72 hours. Typical fever to shock and neurological complications like
‘bisected pearl’ colonies appearing after 3–5 days convulsions and encephalopathy. Provocation
must be investigated further. poliomyelitis is a rare complication.

4. Identification Acellular Pertussis Vaccine


Identification is confirmed by microscopy and Acellular vaccines containing the protective
slide agglutination with specific antisera. lmmuno­ components of the pertussis bacillus (PT FHA.
fluorescence is useful in identifying the bacillus in agglutinogens 1, 2, 3) first developed in Japan, cause
direct smears of clinical specimens and of cultures. far fewer reactions, particularly in older children.
The differentiating features of bordetellae are listed Both whole cell and acellular vaccines have a
in Table 46.1. protection rate of about 90%. Even.

www.ebook3000.com
326  |  Section 3: Systemic Bacteriology
Bordetella parapertussis Important questions
This organism is readily distinguished from B. 1. Discuss various factors which determine the
pertussis by its ability to grow on nutrient agar, with virulence of Bordetella pertussis.
the production of a brown diffusible pigment after 2. Write short notes on:
a. Culture media for Bordetella pertussis
2 days (Table 46.1).
b. Pathogenesis of pertussis
This is an infrequent cause of whooping
c. Laboratory diagnosis of pertussis
cough (5% cases) and disease is mild. The
d. Cough plate method
pertussis vaccine does not protect against Bord.
e. Vaccination against pertussis
parapertussis infection. It usually causes less severe f. Acellular pertussis vaccine.
illness than B. pertussis.
Multiple choice questions (MCQs)
Bordetella bronchiseptica (Bord.
1. Which of the following species of Bordetella is the
bronchicanis) most important human pathogen?
It differs from the other species by also being a. Bordetella pertussis
motile by peritrichate flagella and by producing an b. Bordetella parapertussis
obvious alkaline reaction in the Hugh and Leifson c. Bordetella bronhiseptica
medium. d. Bordetella avium
2. Bordet–Gengou medium contains all the following
It can grow on nutrient agar and is antigenically ingredients except:
related to Bord. pertussis and Brucella abortus a. Potato b. Blood
(Table 46.1). It has been found to cause a very small c. Glycerine d. Agar
proportion (0.1%) of cases of whooping cough. 3. Pertussis toxin is produced by:
a. Bordetella pertussis
Bord. avium: It causes respiratory disease in
b. Bordetella parapertussis
Turkeys (rhinotracheitis).
c. Bordetella bronchiseptica
Key Points d. All of the above
4. The most infective stage in whooping cough is:
™™ The genus Bordetella con­s titutes a group of
minute, gram-negative, non-acid-fast, non-sporing, a. Catarrhal stage
coccobacilli b. Paroxysmal stage
™™ Three important species of Bordetella include Bordetella c. Convalescent stage
pertussis, B. parapertussis and B. bronchiseptica d. None of the above
™™ B. pertussis are extremely small, ovoid, gram-negative 5. Which of the following methods is most suitable
coccobacilli showing pleomorphism; nonmotile and for culture of Bordetella pertussis?
nonsporing a. Pernasal swab culture
™™ Bordetella pertussis are strict aerobes and nutritionally
b. Postnasal swab culture
fastidious, grow on specialized media such as Bordet–
Gengou agar and charcoal agar with 10% blood c. Cough plate method
™™ B. pertussis produces a number of factors that are d. Sputum culture
involved in the pathogenesis of disease, such as 6. The following Bordetella species cause infections
pertussis toxin (PT) in humans except:
™™ Diseases: Pertussis and is characterized by three a. Bordetella pertussis
stages: catarrhal, paroxysmal, and convalescent b. Bordetella parapertussis
stages. Children younger than 1 year at greatest risk c. Bordetella bronchiseptica
for infection
d. Bordetella avium
™™ Laboratory diagnosis: Depends on microscopy,
culture and polymerase chain reaction (PCR) 7. Vaccines available for prophylaxis of whooping
™™ Treatment with macrolide (i.e. erythromycin, cough is/are:
azithromycin) is effective in. Erythromycin has been a. Whole cell pertussis vaccine
used for prophylaxis. Vaccination with whole-cell b. Acellular pertussis vaccine
vaccines is effective but associated with side effects. c. Both the of the above
Acellular vaccines are effective and associated with d. None of the above
fewer adverse effects 8. The most common clinical manifestation of
™™ Bordetella parapertussis is responsible for only tularemia is:
about 5% of cases of whooping cough and relatively
a. Ulceroglandular tularemia
causes a mild form
™™ Bordetella bronchiseptica: It is responsible to cause b. Glandular tularemia
in very small proportion (0.1%) of cases of whooping c. Oculoglandular tularemia
cough d. Oropharyngeal tularemia
™™ Bord. avium: It causes respiratory disease in Turkeys
Answers (MCQs)
(rhinotracheitis).
1. a; 2. c; 3. a; 4. a; 5. a; 6. c; 7. c; 8. a
47
Chapter

Brucella

Learning Objectives

After reading and studying this chapter, you should ∙∙ Discuss pathogenesis of brucellosis
be able to: ∙∙ Discuss laboratory diagnosis of brucellosis
∙∙ Discuss classification of Brucella ∙∙ Describe Castaneda method of blood culture.
∙∙ Describe culture characteristics and biochemical
reactions of Brucella sp.

Introduction Biochemical Reactions


The genus Brucella consists of very small, nonmotile, The metabolism is oxidative and not fermentative.
aerobic, gram-negative coccobacilli. Brucellae are They are catalase-positive, urease positive (variable
essentially pathogens of goats, sheep, cattle and in B. melitensis) and usually oxidase-positive
pigs. Man acquires infection by direct or indirect (except B. neotomae, B. ovis and some strains of
contact with infected animals. Human infection is B. abortus).
a zoonosis that is acquired from animals or animal Indole, MR and VP tests are negative. Citrate
products. is not utilized.
Species: Six species are currently recognized: B. Most Brucella strains reduce nitrates to nitrites
melitensis, B. abortus, B. suis, B. ovis, B. neotomae (except B. ovis).
and B. canis. H 2S is produced from sul­p hur-containing
amino acids by B. neotomae, B. abortus (many
Morphology strains) and B. suis (some strains).
Brucellae are coccobacilli or short rods 0.5–0.7 µm
× 0.6–1.5 µm. They are gram-negative nonacid Resistance
fast, nonmotile, noncapsulated and nonsporing. Brucellae are destroyed by heat at 60°C in 10
Bipolar staining is usually not ob­served. minutes; therefore, they are killed in milk by
pasteurization. They remain viable for 10 days in
Cultural Characteristics refrigerated milk, one month in ice cream, four
Brucellae are strict aerobes. Many strains of B. months in butter and for varying periods in cheese
abortus and nearly all of B. ovis are cap­nophilic, depending on its pH. They can be killed by 1.0%
requiring 5–10% CO2 for growth. The optimum phenol in 15 minutes.
temperature is 37°C (range 20–40°C) and pH 6.6– They are sensitive to many disinfectants and
7.4. They may grow on simple me­dia, though growth antibiotics, including ampicillin, co-amoxiclav,
tends to be slow and scanty. Growth is improved by cephalosporins, aminoglycosides, tetracyclines,
serum or blood. The media employed currently are chloramphenicol, ciprofloxacin, sulfonamides and
serum dextrose agar, serum potato infusion agar, cotrimoxazole.
trypticase soy agar, or tryptose agar. The addition
of bacitracin, polymyxin and cycloheximide to the Antigenic Structure
above media makes them selective. On solid culture There are at least two antigenic determinants—A
media, colonies may take 2–3 days to develop. They (abortus) and M (melitensis)—present on
are small, smooth, transparent, low convex with an the lipopolysaccharide (LPS) protein complex.
entire margin. In liquid media, growth is uniform. B. abortus contains about 20 times A as M, B.

www.ebook3000.com
328  |  Section 3: Systemic Bacteriology
melitensis about 20 times M as A. B. suis has an ∙∙ CO2 requirement
intermediate antigenic pattern. Absorption of the ∙∙ H2S production
minor antigenic component from an antiserum ∙∙ Sensitivity to dyes (basic fuchsin and thionin).
will leave most of the major antibody component ∙∙ Agglutination with monospecific antisera.
and such absorbed A and M monospecific sera are ∙∙ Lysis by specific bacteriophage.
useful for species identification by the agglutination
test. Species and biotype identification depends on Biotypes
a variety of other factors besides antigenic structure The three major species are B. melitensis, B.
(Table 47.1). abortus, B suis infecting primarily goats or sheep,
Antigenic cross reactions exist between brucelIae cattle and swine, respectively. Many biotypes have
and V. cholerae, with E. coli 0: 116; 0: 157, SalmonelIa been recognized in these species (Table 47.1).
serotypes group N (0:30 antigen), Ps. maltophi­la, Y. i. B. melitensis: three biotypes.
enterocolitica and F. tularensis. ii. B. abortus: 7 biotypes (1-6 and 9, biotypes 7
and 8 have been discarded as invalid).
Brucella Bacteriophage iii. B. suis: 5 biotypes.
B. suis strains that produce H 2S are known
The Tblisi (Tb) phage has been designated as the as ‘American’ strains and those that do not as
reference phage and at routine test dose (RTD). It ‘Danish’ strains.
lyses B. abortus at both RTD and 10,000 TTD. B.
suis is lysed at 10,000 RTD, while B. melitensis is Pathogenesis
not lysed at all. They have been classified into six All three major species of brucellae are pathogenic
groups based on their host specificity. to human beings. B. melitensis is the most
pathogenic, B. abortus and B. suis of interme­diate
Classification of Brucellae pathogenicity.
Brucellae may be categorized into species and Mode of infection: The modes of infec­tion are
biovars by the following tests (Table 47.1): by ingestion, contact, inhalation or accidental

Table 47.1  Differential characteristics of the species and biotypes of Brucella


Lysis by phage Growth on dye media Agglutination by mono-
specific serum
Basic Fuchsin 1:50,000

Most common host


Thionin 1:25,000

Thionin 1:50,000
CO2 requirement

H2S production
RTD x 104
Biotypes
Species

RTD

A M R
B. melitensis 1 − − − − + − + − + Sheep,
2 − − − − + − + + − − goats
3 − − − − + − + + + −
B. abortus 1 + + ± + + − − + − − Cattle
2 + + + + − − − + − −
3 + + ± + + + + + − −
4 + + ± + + − − − + −
5 + + − − + + + − + −
6 + + − − + + + + − −
9 + + ± ± + + + − + −

B. suis 1 − + − + − − + + − − Pigs
2 − + − − − − + + − − Pigs, hare
3 − + − − + + + + − − Pigs
4 − + − − + + + + + − Reindeer
B. neotoma'e − + − + − − − + − − Wood rat
B. ovis − − + − + + + − − + Sheep
B. canis − − − − − + + − − + Dogs
RTD: Routine test dilution
A—abortus; M—melitensis; R—rough
Chapter 47: Brucella  | 329
inoculation. Person-to-person spread does not goat, sheep, cattle, buffaloes, and swine. Infection
ordinar­ily occur, but very rarely transmission has is transmitted among animals directly or through
been reported through the placenta, breast feeding blood-sucking arthropods, particularly ticks.
and sex. Brucellosis is a zoonosis of worldwide impor­
i. Ingestion: The most important vehicle of tance, particu­larly in developing countries.
infection is raw milk. Brucellosis may also be potentially transmitted
ii. Contact: Contact infection is especially in association with biowarfare, bioterrorism, or
important as an occupational hazard in agri­ biocriminal activities.
cultural workers, veterinarians, butchers, Almost all human infections in various parts of
animal handlers, and others in occupations that India are due to B. melitensis acquired from goats
involve handling of animals or uncooked animal and sheep. Animal brucellosis is reported from
tissues are at higher risk for direct inoculation. practically every state in India.
iii. Inhalation: Infection is transmitted by inhal­
ation of dried material of animal origin such Laboratory Diagnosis
as dust from wool. The clinical manifestations of human brucellosis are
Course of disease: Brucellosis is primarily an variable, so clinical diagnosis is almost impossible
intracellular pathogen affecting reticuloendothelial and laboratory aid is therefore essential. Laboratory
system. Brucellae have a special predilection for methods include culture of brucellae, serology,
intracellular growth and may be demonstrated polymerse chain reaction and hypersensitivity type
inside phagocytic cells. The brucellae spread skin tests.
from the initial site of infec­tion through lymphatic
channels to the local lymph glands, in the cells of 1. Specimens
which they multiply. They then spill over into the
Blood culture is most important. Material from
bloodstream and are disseminated throughout the
bone marrow or liver biopsy, is also cultured, lymph
body. Once liberated into the bloodstream, they
nodes, cerebrospinal fluid, urine and abscesses, and
may infect a variety of organs but are most often
on occa­sion, also from sputum, breast milk, vaginal
localized in the reticuloendothe­lial system, where
discharges and seminal fluid.
they reside within phagocytic cells. Granulomas or
abscesses most often develop in the bone marrow,
2. Culture
liver, spleen, lymph nodes, or lungs. Other sites
of infection may include subcutaneous tissue, a. Blood culture
testes, epididymis, ovary, gallbladder, kidneys, and Blood culture is the most definitive method for
brain. Meningitis and endocarditis are commonly the diagnosis of brucellosis. When brucellosis is
reported complications. suspected, blood culture should be attempted
repeatedly, not only during the febrile phase.
Types of Human Infection Because the organisms may be scanty, at least
10 mL of blood should be withdrawn on each
The incubation period is usually about 10–30 days.
occasion, 5 mL being added to each of two blood
Human infection may be of three types:
culture bottles containing serum dextrose (SD)
1. Latent or subclinical infection
broth. One of these bottles should be incubated in
There is no clini­cal evidence of disease but is
an atmosphere containing 10% carbon dioxide at
detectable only by serological tests.
37°C. Subcultures are made on solid media (serum
2. Acute brucellosis: Acute brucellosis is mostly
dextrose agar) every 3–­5 days, beginning on the
due to B. melitensis. It is associated with
fourth day. Growth may often be delayed and blood
prolonged bacteremia and irregular fever. It is
cultures should be retained for 6–8 weeks before
also known as undulant fever or Malta fever
being discarded as negative. Preliminary lysis and
because of the periodic noctural fever that may
centrifugation of the blood improves the isolation
occur over weeks, months or years particu­larly
rate. Automated blood culture systems may also
in untreated cases.
be used.
3. Chronic brucellosis: Chronic brucellosis is a low
grade infection with periodic exacerbations. It is b. Castaneda’s method of blood culture (Fig.
usually nonbacteremic. The illness lasts for years. 47.1)
Advantages: The Castaneda method of blood
Epidemiology culture has sev­eral advantages and is recommended.
Brucella organisms are distributed throughout i. A two-phase Castaneda culture system, in
the world. Human brucellosis is acquired from which the broth is periodi­cally allowed to flow
animals, directly or indirectly. The animals that over agar contained within the blood culture
com­monly act as sources of human infection are bottle, may be used. The blood is inoculated

www.ebook3000.com
330  |  Section 3: Systemic Bacteriology
usually persist throughout the active phase of the
disease and in some cases long thereafter. As the
disease progresses, IgM antibodies decline, while
the IgG antibodies persist or increase in titer.
In chronic infections, IgM may often be absent
and only IgG can be demonstrated.
The agglutination test iden­tifies mainly the
IgM antibody, while both IgM and IgG can fix
complement. However, as IgG and IgA antibodies
are formed during the course of the infection
some of them bind with antigen, thus preventing
its aggluti­nation by larger IgM molecule. These
IgG and IgA antibodies are known as blocking
or non-agglutinating antibodies which may
prevent agglutination. It is thus evident that the
agglutination test is usually posi­t ive in acute
infection but may be only weakly positive or even
negative in chronic cases.
Fig. 47.1: Castaneda's method of blood culture
Brucella antibodies can be detected by a variety
into the broth and the bottle incubated in the of serological tests.
upright position. For subculture, it is sufficient i. Standard agglutination test (SAT): This is a
if a bottle is tilted so that the broth flows tube agglutination test in which equal volumes
over the surface of the agar slant. It is again of serial dilutions of the patient’s serum and the
incubated in an upright position. Colonies standardized antigen (a killed suspension of a
appear on the slant. standard strain of Br. abortus) are mixed and
ii. This method minimizes materials and incubated at 37°C for 24 hours or 50°C for 18
manipulation. hours. It detects antibodies against B. abortus,
iii. It reduces chances of contamination during B. suis, and B. melitensis but not B. canis. A titer
the period of incu­bation and risk of infection of 160 or more is considered significant.
to laboratory workers. Prozone phenomenon: Sera often contains
Blood cultures are positive only in about 30–50 ‘blocking’ or ‘nonaggluti­nating’ antibodies. A
per cent cases, even when repeated samples blocking factor may interfere with agglutination at
are tested. B. melitensis and B. suis are more low serum dilutions (the prozone phenomenon)
frequently isolated from blood than are B. although positive in higher dilutions.
abortus or B. canis. The blocking effect may some­times be removed
c. Bone marrow or liver biopsy by prior heating of the serum at 55°C for 30 minutes
Isolation rates can be markedly improved if material or by using 4% saline as the dilu­ent for the test. The
from bone marrow or liver biopsy is also cultured. most reliable method for obviating the blocking
Identification: Depends upon biochemical effect and detecting the ‘incomplete’ an­tibodies is
tests and may be classified into dif­ferent species the antiglobulin (Coombs) test.
and biovars, based on CO 2 requirements, H 2S Cross-reactions: Cross-reactions may be observed
production, sensitivity to dyes (basic fuchsin and with antibodies directed against Francisella
thionin), agglutination with monospecific sera, tularensis, Vibrio cholerae, or Yersinia enterocolitica
lysis by specific phage and oxidative metabolic tests or immunization. Cholera induced aggluti­nins may
with amino acids and carbohydrates. be differentiated by the agglutinin absorption test
and also as they are removed by treatment with
3. Serological Tests 2-mercapto-ethanol. In order that results from,
In the absence of positive cultures, the diagnosis different laboratories are comparable; it is the
of bru­cellosis usually depends on serological tests, practice to express agglutinin titers in International
the results of which tend to vary with the stage of Units. This is done by using a standard reference
the infection. serum for com­parison.
Antibodies appear within 7–10 days of onset ii. Mercaptoethanol test: The mercaptoethanol
of the disease. IgM antibodies appear first which test is carried out simultaneously and in
are rapidly followed and superseded by IgG and the same manner as the standard aggluti­
to lesser extent IgA antibodies. These antibodies nation test except that the saline diluent
reach their maximum titers in the third or fourth contains 0.05 M 2-mercaptoethanol. The
week of disease and then slowly decline but they agglutinating ability of IgM, and sometimes
Chapter 47: Brucella  | 331
IgA, is destroyed by 2-mercaptoethanol, and Result
therefore agglutination in this test is indicative Positive test: The bacilli are agglutinated and rise
of the continuing presence of IgG and the with the cream to form a blue ring at the top, leav­
likelihood of persisting infection. ing the milk unstained.
iii. Complement fixation test: The complement
fixation test is more useful in chronic cases as Negative: No colored ring is formed and the milk
it detects IgG and IgM antibody also. remains uniformly blue.
iv. Enzyme-linked immunosorbentassay
(ELISA) and radio immunoassay (RIA): These ii. Whey Agglutination Test
tests are very sensitive useful for differentiation It is another useful method for detecting the
between the acute and chronic phases of antibodies in milk.
brucellosis. ELISA is sensitive, specific and can
detect IgM and IgG antibody separately. Prophylaxis
v. Rose Bengal plate test: This is a rapid slide
1. Prevention consists of checking brucellosis in
agglutination test. It is widely used as a
dairy animals.
screening test in farm animals.
2. Control of this disease in cattle has been
vi. Rapid dipstick assay: The rapid dipstick
achieved by serologic surveillance, vaccination
assay, a relatively simple screening procedure
(B. abortus strain 19), and elimination of
for Brucella­ specific IgM, shows promise as a
reactor cattle.
field test in areas without direct access to a
reference laboratory. 3. Pasteurization of infected milk or milk products.
4. Vaccines have been developed for use in animals.
Polymerase Chain Reaction (PCR) The live-attenuated B. abortus strain S19 vaccine
has been is now being replaced by the rough
Promising results have been obtained in clinical
strain B. abortus RB51, which gives comparable
studies.
protection, but does not induce interfering
Hypersensitivity Test (Brucellin Skin Test) antibodies and is less hazardous to man.
The live-attenuated smooth strain B. melitensis
Delayed hypersensitivity type skin tests with
Rev I is used to protect sheep and goats from B.
brucella antigens (‘brucellins’) are not useful in
melitensis infection. Strain 2 has been used in
diagnos­ing acute brucellosis. This test, similar to
China.
the tuberculin test, is no longer recommended.
They parallel the tuberculin test in indicating only 5. Human vaccination is not recommended.
prior sensitization with the antigens, and may
remain positive for years. Treatment
Brucella infections respond to a combination of
Detection of Animal Infection strepto­mycin or gentamicin and tetracycline or to
The methods used for the laboratory diagnosis rifampicin and doxycycline. Tetracycline alone is
of human brucellosis may also be employed for often adequate in mild cases. Treatment should be
the diagnosis of animal infections. In addition, continued for at least 6 weeks. Cotrimoxazole and
brucellae may be demonstrated microscopically rifampicin can be used in children.
in pathological specimens by suitable staining or
Key Points
by immunofluores­cence.
Several rapid methods have been employed for ™™ The genus Brucella consists of very small, nonmotile,
the detection of brucellosis in herds of cattle. These aerobic, Gram-negative coccobacilli. They are
essentially patho­gens of goats, sheep, cattle and pigs
include the rapid plate agglutination test and the
™™ Six species are currently recognized and three major
Rose Bengal card test. For the detection of infected species of the genus Brucella are B. melitensis, B.
animals in dairies, pooled milk samples may be abortus, B. suis
tested for ba­cilli by culture and for antibodies by ™™ They grow best on media employed currently is
several tech­niques such as milk ring test. serum dextrose agar, serum potato infusion agar,
tryp­ticase soy agar, or tryptose agar. They are
i. Milk Ring Test catalase and oxidase positive
™™ Animal reservoirs are goats and sheep, cattle, swine,
1. In the milk ring test a sample of whole milk is and dogs. Individuals at greatest risk for disease are
mixed well with a drop of the stained brucella people who consume unpasteurized dairy products,
antigen (a concentrated suspension of killed Br. people in direct contact with infected animals, and
abortus stained with hematoxylin). laboratory workers
™™ Human brucellosis is primarily a zoonotic bacterial
2. It is incubated in a water bath at 70°C for 40–50 infection
minutes.

www.ebook3000.com
332  |  Section 3: Systemic Bacteriology
3. Brucellae are transmitted to humans by:
™™ Laboratory diagnosis: Depends on the culture of
brucellae serology. Detection of Brucella species. Milk a. Direct contact with animal tissues
ring test is a screening test used for demonstration b. Ingestion of contaminated meats
of anti­bodies in the milk of animals c. Ingestion of raw infected milk
™™ Human disease is controlled by eradication of the d. All of the above
disease in the animal reservoir; pasteurization of 4. The most common Brucella species causing human
dairy products; and use of proper safety techniques infection in India is:
in clinical laboratories work­ing with this organism. a. Brucella melitensis b. Brucella abortus
c. Brucella suis d. Brucella canis
Important questions 5. The specimen of choice for isolation of Brucella is:
a. Blood b. Bone marrow
1. Discuss laboratory diagnosis of brucellosis. c. Urine d. Stool
2. Write short notes on: 6. Rose Bengal Card’ test is employed for which of the
a. Castaneda method of blood culture following infections?
b. Serodiagnosis of brucellosis a. Brucellosis
c. Diagnosis of brucellosis in animals. b. Salmonellosis
c. Tularensis
Multiple choice questions (Mcqs) d. None of the above
7. Milk ring test is used for diagnosis of:
1. All the following statements are true regarding
a. Bovine tuberculosis b. Brucellosis
cultural characteristics of Brucella except:
c. Salmonellosis d. Anthrax
a. They are strict aerobes
b. They require 5–10% of CO2 for better growth 8. Vaccination of cattle against brucellosis is done
c. They produce detectable colonies within 24–48 with:
hours a. Attenuated B. abortus vaccine
d. Erythritol has stimulatory effect on growth of the b. Attenuated B. melitensis vaccine
bacteria. c. Attenuated B. ovis vaccine
2. Humans acquire Brucella melitensis infection from: d. Attenuated B. neotome vaccine
a. Sheep b. Cow Answers (MCQs)
c. Pig d. Wood rat 1. c; 2. a; 3. d; 4. a; 5. a; 6. a; 7. b; 8. a
4848
Chapter

Spirochetes

LEARNING OBJECTIVES

∙∙ Describe diseses caused by different spirochetes ∙∙ Describe the following: fluorescent treponemal
∙∙ Name various pathogenic treponemes antibody-absorption (FTA-ABS) test; TPHA (or) T.
∙∙ Describe morphology of Treponema pallidum pallidum hemag­glutination test
∙∙ Discuss BFP (Biological false-positive) reactions
∙∙ Discuss pathogenesis of syphilis
∙∙ Describe the following: i. Endemic syphilis (or)
∙∙ Describe diseses caused by Treponema pallidum
Bejel; ii. Yaws; iii. Treponema pertenue; iv. Pinta; v.
their laboratory diagnosis
Borrelia recurrentis; vi. Borrelia vincentii; vii. Borrelia
∙∙ Explain serological tests for syphilis burgdorferi or Lyme disease; viii. Leptospirosis; ix.
∙∙ Discuss standard tests for syphilis (STS) Weil’s disease.

INTRODUCTION Larger spirochetes like Borrelia are Gram-


The spirochetes (from Speira, meaning coil and negative but other spirochetes cannot be stained
chaite, meaning hair) are elongated, motile, slender, by routine methods. However, the spirochetes
helically coiled, flexible organisms with one or more can be seen by dark ground microscopy, silver
complete turns in the helix. Multiplication is by impregnation method and immunofluorescence.
transverse fission. Many are free-living saprophytes,
while some are obligate parasites. They may be CLASSIFICATION
aero­bic, anaerobic or facultative.
Spirochetes are slender unicellular helical Spirochetes belong to the order Spirochetales. It
or spiral rods with a number of distinctive has two families:
ultrastructural features. They are gram-negative and 1. Spirochetaceae: Spirochaetes are anaerobic,
are 0.1–3.0 µm wide and 5–250 µm in length. They facultative anaerobic or microaerophilic. They
are structurally more complex than other bacteria. are not hooked.
The spirochetes have gram-negative type cell wall
Family Spirochaetaceae has genera: Borrelia,
composed of an outer membrane, a peptidoglycan
Cristispira, Serpulina, Spirocheta and Trepo­
layer and a cytoplasmic membrane. They are
nema;
structurally more complex than other bacteria.
Their most distinctive morphologic property is 2. Family Leptospiraceae: These spirochetes are
the presence of varying num­bers of endoflagella. obligate aerobes and are hooked. It contains
These endoflagella are polar flagella wound along three genera: Leptospira, Leptonema and
the helical protoplasmic cylinder, and situated Tumeria. Only Leptospira spp. are considered
between the outer membrane and cell wall. to be pathogenic for animals or man.
The spiral shape and serpentine motility of the The spirochetes also fall into genera based
spirochaetes depend upon the integrity of these loosely on their morphology (Fig. 48.1). Treponema
endoflagella. Motility is of three types: (i) flexion are slender with tight coils; Borrelia are somewhat
and extension of cells, (ii) corkscrew-like rotatory thicker with fewer and looser coils; and Leptospira
move­ment around the long axis, and (iii) translatory resemble Borrelia except for their hooked ends.
motion, i.e. from one site to another. Some are very Diseased caused by Treponema, Borrelia, and
actively motile’while others are sluggish. Leptospira are given in Table 48.1.

www.ebook3000.com
Chap-48.indd 333 15-03-2016 11:18:34
334  |  Section 3: Systemic Bacteriology
Morphology
It is a very delicate, spiral filament 6–14 µm (average
10 µm) by 0.2 µm, with 6–12 coils which are com­
paratively small, sharp and regular. The length of
the coils is about 1 µm and the depth 1–1.5 µm. The
ends are pointed and tapering. Spirochaetes show
rotary corkscrew-like motility and also movements
of flexion; angulation, with the organism bending
almost to 90° near its centre, is highly characteristic
of T. pallidum. During motion, secondary curves
Fig. 48.1: Species designation of spirochetes based on
appear and disappear in succession but the
morphology primary spirals are unchanged (Fig. 48.2).
T. pallidum cannot be seen under the light
micro­scope in wet films but can be made out by
TREPONEMA negative staining with Indian ink. Its morphology
and motility can be seen under the dark ground
The name Treponena is derived from the Greek
or phase contrast microscope (Fig. 48.2). It does
words trepo :to turn and nema, meaning thread) are
not take ordinary bacterial stains but stains light
relatively short slender spiro­chetes with fine spirals
rose red with prolonged Giemsa stain­ing. It can
and pointed or rounded ends. Some of them are
be stained by silver impregnation methods.
pathogenic, while others occur as commensals in
Fontana’s method is useful for staining films
the mouth, intestines, and genitalia.
and Le­v aditi’s method for tissue sections.
The two treponemal species that cause human Immunofluorescence methods can now be used
disease are Treponema pallidum (with three to detect treponemes in tissues and body fluids.
subspecies) and Treponema carateum. The species Ultrastructurally, the cytoplasm of T. pallidum
T. pallidum is now considered to include three is surrounded by a trilaminar cytoplasmic
subspecies—subspecies pallidum, endemicum membrane, enclosed by a cell wall containing
and pertenue. peptidoglycan. External to this is the rigid rich outer
membrane layer. Usually three, but occasionally
Treponemes Cause the Following four, endoflagella lie inside the outer membrane
Diseases in Humans and are inserted at the tapering portion at each
1. T. pallidum subspecies pallidum causes end of the cell. In contrast to other motile bacteria,
Venereal syphilis these flagella do not protrude into the sur­rounding
2. T. pallidum, subspecies endemicum (T. endem­ medium but are enclosed within the bacterial outer
icum) causes endemic syphilis. membrane. A capsular or slime layer has been
observed occasionally on the surface of T. pallidum.
3. T. pallidum subspecies pertenue causes yaws
4. T. carateum causes pinta. Cultivation
It is possible to maintain T. pallidum in motile and
Treponema pallidum Subspecies pallidum virulent form for 10–12 days in complex media
Treponema pallidum is the causative agent of syphilis. under anaerobic conditions. Virulent T. pallidum
The name pallidum refers to its pale staining. strains have been maintained by serial testicular

Table 48.1  Diseases caused by spirochetes


Genus Species Diseases Transmission
Teponema T. pallidum Syphilis Sexual contact or congenital
T. pertenue Yaws Traumatized skin comes in contact with an infected lesion
T. carateum Pinta Traumatized skin comes in contact with an infected lesion
T. endemicum Endemic syphilis Mouth to mouth by utensils
Borrelia B. recurrentis Epidemic Body louse
Relapsing fever Soft-shelled tick
Endemic Relapsing
fever
B. vincentii Vincent’s angina
B. burgdorferi Lyme disease Tick bites
Leptospira L. interrogans Leptospirosis
L. biflexa Saprophytes

Chap-48.indd 334 15-03-2016 11:18:35


Chapter 48: Spirochetes  | 335
nonpatho­genic treponemes, such as Reiter
treponeme.
2. Species-specific treponemal antigen: It appears
to be polysaccharide in nature. T. pallidum is
used as antigen for detection of species-specific
antibody.

Pathogenesis
Treponema pallidum subsp. pallidum causes
syphilis. Venereal syphilis is acquired by sexual
contact. Syphilis can also be acquired by nongeni­
tal contact with a lesion (e.g. on the lip) or
transplacen­tal transmission to a fetus, resulting in
congenital syphilis. T. pallidum enters tissues by
penetration of intact mucosae or through abraded
skin. Clinical disease sets in after an incubation
period of about a month (range 10–90 days). The
Fig. 48.2: Treponema pallidum-dark ground illumination natural course of syphilis can be divided into
primary, secondary, and tertiary stages based
passage in rabbits for many decades. One such
on the clinical manifestations.
strain (Nichol’s strain) was isolated in 1912 from a
patient of general paralysis of the insane. i. Primary Disease
Cultivable treponemes such as T. phagedenis
The bacte­ria multiply at the initial entry site and
(Reiter’s treponeme) and T. refringens (Noguchi
the primary lesion in syphilis is the chancre.
strain) are nonpathogenic. They can be grown
The chan­cre is a painless, relatively avascular,
under strict anaerobic conditions.
circumscribed, indurated, superficially ulcerated
lesion. It is covered by a thick, glairy exudate very
Resistance
rich in spiro­chetes. It is known as ‘hard chancre’.
T. pallidum is very delicate, being readily inactivated The chancre is most frequently on the external
by drying or by heat (41–42°C in one hour). It is genitalia.
inactivated by contact with oxygen, distilled water, The regional lymph nodes are swollen,
soap, arsenicals,mercurials, bismuth, common dis­c rete, rubbery and nontender. The chan­
antiseptic agents and antibiotics. It is killed in 1–3 cre invariably heals in about 10–40 days, even
days at 0–4°C, so that transfusion syphilis can be without treatment, leaving a thin scar. Chancres
prevented by storing blood for at least four days in usually occur singly but multiple or persistent
the refrigerator before transfu­sion. Stored frozen chancres may develop in immunocompromised
at –70°C in 10% glycerol, or in liquid nitrogen individuals, such as those infected with the human
(–130°C), it remains viable for 10–15 years. immunodeficiency virus (HIV).
Antigenic Structure ii. Secondary Syphilis
The antigenic structure of T. pal­lidum is complex. Secondary syphilis sets in 1–3 months after healing
Treponemal infection induces at least three types of primary lesion. The secondary lesions are due
of antibodies. On the basis of these antibodies, to wide­spread multiplication of the spirochetes
the treponemal antigens may be divided into and their dissemination through the blood. In this
nonspecific and specific antigens. stage, patients typically experience a “flu-like”
syndrome, lymphadenopathy, and a generalized
A. Nonspecific Antigen mucocutaneous rash. Characteristic lesions
The first is the reagin antibody in which a hapten are roseolar or papular skin rashes, mucous
extracted from the beef heart is used as the antigen. patches in the oropharynx and condylomata at
This lipid hapten is known as cardioliom and is the mucocutaneous junctions. Condylomata lata
chemically a diphosphatidyl glycerol. This lipid occur around moist areas, such as the anus and
has been detected in T. pallidum but it is not vagina. The patient is most infectious as with the
known whether the reagin antibody is in­duced primary chancre. There may also be ophthalmic,
by cardiolipin that is present in the spirochete or osseous and meningeal involvement.
released from damaged host tissues.
iii. Latent Syphilis
B. Specific Antigens After the secondary lesions disappear there is a
1. Group-specific antigen: It is a protein anti­ period of quiescence known as ‘latent syphilis’. No
gen present in T. pallidum as well as in clinical manifestations are evident and diagnosis

www.ebook3000.com
Chap-48.indd 335 15-03-2016 11:18:35
336  |  Section 3: Systemic Bacteriology
during this period is possible only by serological applying thin coverslips, examined under the dark
tests. In many cases, this is followed by natural cure ground microscope.
but in others, after several years, manifes­tations of
tertiary syphilis appear. A. Demonstration of Treponemes
a. Demonstration of Treponemes in the
iv. Tertiary Syphilis or Late Syphilis Exudate
Tertiary or late syphilis, which may develop decades 1. Dark Ground Microscopy
after the primary infection, is a slowly progressive, Dark field microscopy: Diagnosis by microscopy
destructive inflammatory disease that may affect is applicable in primary and secondary stages and
any organ. The three most common forms of late in cases of congenital syphilis with superficial
syphilis are cardiovascular syphilis, gummatous lesions. This bacterium is too thin to be visualized
syphilis and neurosyphilis. with a standard Gram stain so two techniques
a. Cardiovascular syphilis: These consist of to visualize it with a light microscope are dark
cardiovascular lesions including aneurysms, field microscopy and immunofluorescence. T.
chronic granulomata (gummata) and meningo­ pallidum is recognized by its slender struc­ture,
vascular manifestations. regularity of its spirals and slightly pointed ends.
b. Gummatous syphilis: It is a rare granulomatous A treponemal concen­tration of 104 per mL in the
lesion of the skeleton, skin or mucocutaneous exudates is required for the test to be positive.
tissues. 2. Direct Fluorescent-antibody Staining for
c. Neurosyphilis: Neurological manifestations, Treponema pallidum (DFA- Tp)
such as tabes dorsalis or general paralysis of the For direct fluorescent-antibody staining for T.
insane develop several decades after the initial pallidum (DFA-TP) test whereby a smear of exudate
infection in a few cases. These are known as late is made on a slide, fixed in acetone, and DFA-TP test
tertiary or quaternary syphilis. is done using fluorescent tagged anti T. pallidum
antiserum. The use of specific monoclonal
Congenital Syphilis antibody has made the test more reliable. The
In conn­genital syphilis infection is transmitted treponemes appear distinct, sharply outlined and
from mother to fetus transplacentally. The lesions exhibit an apple green fluorescence. It is a better
of congenital syphilis usually develop only after and safer method for microscopic examination.
the fourth month of gestation. Congenital syphilis
can be prevented if the mother is given adequate b. Demonstration of Treponemes in Tissues
treatment before the fourth month of pregnancy. Treponemes in the tissues can be demonstrated by
immunofluorescence staining or silver impregn­
Syphilis Acquired Nonvenereally ation method of staining (Levaditis stain).
In syphilis acquired nonvenereally (as occupa­ c. Demonstration of Treponemal Antigen in
tionally in doctors or nurses), the natural evolution
the Lesion
is as in venereal syphilis except that the primary
chan­cre is extragenital, usually on the fingers. In T. pallidum antigen in the lesion can be detected
the rare instances where syphilis is transmitted by enzyme immunoassay and polymerase chain
by blood trans­fusion, the primary chancre does reaction (PCR).
not occur.
B. Serological Tests
Laboratory Diagnosis These tests form the mainstay of laboratory
diagnosis. (Table 48.2). Two major types of serologic
Labo­ratory diagnosis consists of demonstration tests exist: nontreponemal tests and treponemal
of the spi­rochetes under the microscope and of tests. In nontreponemal tests or standard tests
antibodies in serum or CSF. for syphilis (STS) cardiolipin or lipoidal antigen
is used, while in treponemal tests treponemes are
Specimen Collection and Handling used as the antigen.
Specimens should be collected with care as
the lesions are highly infectious. The lesion is a. Nontreponemal Tests or Standard Tests for
cleaned with a gauze soaked in warm saline and Syphilis
the margins gently scraped so that the superficial Reagin antibodies are detected by cardiolipin
epithelium is abraded. Gentle pressure is applied antigen in standard tests for syphilis (STS).
to the base of the lesion and the serum that exudes The antigen used in these tests is an alcoholic
is collected preventing admixture with blood. extract of beef heart tissue (cardiolipin) to which
Wet films are prepared with the exudate and after lecithin and cholesterol are added. The STS

Chap-48.indd 336 15-03-2016 11:18:35


Chapter 48: Spirochetes  | 337
Table 48.2  Diagnostic tests for syphilis
A. Demonstration of treponemes
   a. D emonstration of treponemes in the 1. Dark ground microscopy
exudate 2. Direct fluorescent-antibody staining for Treponema pallidum (DFA- Tp)
   b. D
 emonstration of treponemes in tissues 1. By immunofluorescence staining
2. Silver impregnation method ( Levaditis stain)
   c. Demonstration of treponemes antigen in 1. Enzyme immunoassay
the lesion 2. Polymerase chain reac­tion (PCR).
B. Serological tests a. Non-treponemal tests
Nonspecific (reagin antibody) tests using the cardiolipin antigen
(standard tests for
syphilis or STS)
1. Wassermann CF reaction
2. Kahn flocculation test
3. Venereal Disease Research Laboratory (VDRL) test
4. Rapid Plasma Reagin (RPR) test
5. Toluidine Red Unheated Serum Test (TRUST)
b. Treponemal tests
a. Group specific test using cultivable treponemal (Reiter strain) antigen
I. Reiter Protein CF (RPCF) test (1953)
b. Specific tests using pathogenic treponemes (T. paliidum)
I. Test using live T. pallidum
Treponema pallidum Immobilization (TPI) test
II. Tests using killed T. pallidum
a. Treponema pallidum agglutination (TPA) test
b. Treponema pallidum immune adherence(TPIA) test
c. Fluorescent Treponemal antibody absorption (FTA-ABS) test
III. Tests using T. pallidum extract
a. Treponema pallidum Hemagglutination Assay (TPHA)
Microhemagglutination test for Treponema pallidum (MHA-TP)
b. Treponema pallidum Enzyme Immunoassays (TP-EIA)

includes Wassermann, Kahn, Venereal Diseases Serum is inactivate heated to it prior to the test,
Research Laboratory (VDRL) and the rapid plasma whereas CSF need not be heated.
reagin (RPR) tests. All these tests are flocculation 2. Inactivated patient serum (0.05 mL) is pipetted
tests except Wassermann reaction which is a into the paraffin ring on the glass slide. Each
complement fixation test (CFT). The Wassermann (0.05 mL) of positive and negative control sera
reaction is no longer in use. Similarly Kahn test is are pipetted into other paraffin rings.
rarely done these days. 3. One drop of working antigen suspension is
The two nontreponemal tests widely used today added to each of these paraffin rings from a
are the Venereal Disease Research Laboratory syringe delivering 60 drops in 1 mL.
(VDRL) and rapid plasma reagin (RPR) tests.
4. Mix with wooden sticks and rotate slide at 180
Veneral Disease Research Laboratory revolutions per minute for four minutes on a
(VDRL) Test mechanical VDRL or manually. Flocculation
occurs in a positive reaction and is observed
It is more rapid test which gives more quantitative
microscopically.
results (VDRL, for Veneral Disease Research
Laboratory, USPHS, New York, where the test was
developed). These tests are cheaper, more rapid Interpretation
and simpler to perform and control. The results of qualitative test are reported as
VDRL is the most widely used simple and rapid ‘reactive’, ‘weak reactive’ or ‘nonreactive’.
test which requires only a small quantity of serum. Reactive’ means positive (large clumps of antigen
The VDRL test uses a cardiolipin antigen that is with marked background clearing are obtai­ned.)
mixed with the patient’s serum or CSF. Flocculation while ‘nonreactive’ is negative.The reciprocal of
occurs in a positive reaction and is observed the end point is given as the titer for reporting
microscopically. of quantitative test, e.g. reactive in 1:4 dilution is
reported as ‘reactive 4 dilution’ or R4.
Method Sometimes VDRL test may give false-negative
1. The test is done in a specially prepared slide, reaction due to high titers of antibody in patient’s
with depressions of 14 mm diameter each. serum (prozone phenomenon). The test is

www.ebook3000.com
Chap-48.indd 337 15-03-2016 11:18:35
338  |  Section 3: Systemic Bacteriology
performed with diluted serum in such cases and damage and are typically seen in: 1. SLE and
it becomes positive. other collagen diseases; 2. Leprosy; 3. Malaria;
4. Relapsing fever; 5. Infectious mononu­cleosis;
Rapid Plasma Reagin Test 6. Hepatitis; 7. Tropical eosinophilia
The black carbon particles are bound to cardiolipin;
when mixed with a positive serum on a disposable b. Treponemal Tests (Table 48.2)
card, the particles clump together. Agglutination is Treponemal tests in which treponemes are
easily observed without a micro­scope. used as the antigen. These are of two types:
RPR test employs a stabilized VDRL carbon 1. Those using cultivable treponemes, such
antigen which make the result more clear cut and as Reiter treponemes (T. phagedenis) as the
is read macroscopically. antigen-Reiter protein comple­ment fixation
(RPCF) test
Advantages of RPR Test 2. Species tests using pathogenic T. pallidum
(Nichol’s strain)
i. It enables the result to be read by eye instead
of microscopically. i. Using Live T. Pallidum
ii. Useful in field studies in developing countries. Treponema pallidum immobilisation (TPI)
test
iii. RPR test can be done with unheated serum or
plasma. ii. Using Killed T. Pallidum
iv. A fingerprick sample of blood is sufficient. a. Treponema pallidum immune
adherence (TPIA) test.
b. Treponema pallidum agglutination
Disadvantage
(TPA) test.
It is not suitable for testing cerebrospinal fluid c. Fluorescent treponemal antibody-
(CSF). absorption (FTA-Abs) test.
iii. Using an extract of T. pallidum
Toluidine Red Unheated Serum Test (TRUST)
a. Treponema pallidum
Instead of carbon particles it uses paint pigment hemagglutination assay (TPHA) test.
toner toludine red particles. b. Enzyme immunoassay (EIA).
Automated RPR test (ART): Automated RPR test
is available for large scale tests. 1. Group-specific Tests
Using Reiter Treponeme
VDRL–ELISA test: An automated VDRL–ELISA test
has been developed which can measure IgG and Reiter Protein Comple­ment Fixation (RPCF)
IgM antibodies separately and is suitable for large Test
scale testing of sera. The principle of this test is the same as that of
Wassermann test. These employed the cutivable
Disadvantages of STS Reiter treponemes for prepation of antigen. Its
Since cardiolipin antigen tests detect antibodies sensitivity and specificity were lower than those
against a nonspecific antigen a positive result of tests using T. pallidum. RPCF and other Reiter
is some­times obtained with sera from healthy treponeme tests are not now in general use.
individuals or patients without clinical evidence
of syphilis. These reactions are termed biological 2. Species Tests Using Pathogenic
false-positives (BFP) rections. These are not T. Pallidum (Nichol’s Strain)
caused by technical faults. They represent i. Tests Using Live T. pallidum
nontreponemal cardiolipin antibody responses. Treponema pallidum immobilization (TPI) test
BFP antibody is usually IgM, while reagin The test serum is incubated with complement and
antibody in syphilis is mainly IgG. Clinically, BFP T. pallidum maintained in a complex medium
reactions may be classified as acute or chronic. anaerobically. If an­t ibodies are present, the
a. Acute BFP reactions: Acute BFP reactions last treponemes are immobilized, that is, rendered
only for a few weeks or months and are usually nonmotile, when examined under dark ground
associated with acute infec­tions, injuries or illumination. The test is considered posi­tive if the
inflammatory conditions. percentage of treponemes immobilized is 50 or
b. Chronic BFP reactions: Chronic BFP reactions more, negative if 20 or less, and inconclusive if in
persist for longer than six months. These between.
may occur in a wide variety of infectious and TPI was the most specific test availa­ble for
noninfectious conditions associ­ated with tissue diagnosis of syphilis and was considered the gold

Chap-48.indd 338 15-03-2016 11:18:35


Chapter 48: Spirochetes  | 339
standard in syphilis serology. Because the TPI test of fluorescein-labeled, anti-human globulin and
employs live treponemes it is time-consuming, examination of the slide with an ultraviolet (UV)
expensive and technically de­manding. microscope. Positive results are indicated by
fluorescence of the T. pallidum organisms. An
ii. Tests Using Killed T. pallidum FTA-ABS 19S-lgM test has also been developed for
The Treponema pallidum immune adherence evaluation of congenital syphilis.
(TPIA) test and Treponema pallidum agglutination FTA-ABS is as specific as the TPI test and is
(TPA) test were used for some time but have now now accepted as a standard refer­ence test and
been given up. is highly specific and sensitive at all stages of
syphilitic infection although a small percentage
a. Treponema pallidum Immune Adherence of false positive reactions occurs. The FTA-Abs
(TPIA) Test test is positive in approximately 80, 100 and
95% of primary, secondary and late syphilitics,
A suspension of T. pallidum, inactivated by
respectively, and, unlike the VDRL test, remains
formalin, is mixed with the test serum and
positive following successful therapy. However,
examined under dark ground microscopy. The
as it can be done only in suitably equipped
treponemes are found agglutinated in the presence
laboratories, it is not available for routine testing.
of antibodies. The test is not very specific and false-
positive reactions are common. iii. Tests Using an Extract of T. pallidum
b. Treponema pallidum Agglutination (TPA) a. T. Pallidum Hemagglutination Assay
Test (TPHA)
A suspension of inactivated T. pallidum is mixed The Treponema pallidum hemagglutination as­say
with the test serum, complement and fresh (TPHA) uses tanned erythrocytes sensitized with
heparinized whole blood from a normal individual a sonicated extract of T. pallidum as antigen.
and incubated. The treponemes will be found When these sensitized erythrocytes are mixed with
to adhere to the erythrocytes in the presence of patiet’s serum (test sera) containing antireponemal
antibodies. Both TPA and TPIA are not used in antibodies it causes hemagglutination. As in the
diagnostic laboratories. FTA-Abs assay, sera are pre­absorbed with a non-
pathogenic treponeme to remove antibody against
Fluorescent Treponemal Antibody (FTA) Test commensal spirochetes.
It is an indirect immunofluorescence test using as The test sera for TPHA are absorbed with a dilu­
antigen, smears prepared on slides with Nichol’s ent containing components of the Reiter treponeme,
strain of T. pallidum. The slides can be stored for rabbit testis and sheep erythrocytes. Sera are
sev­eral months in deep freeze. The currently used screened in an initial dilution of 1 : 80 but titers of
modi­fication of the test is the FT A-Absorption (FT 5120 or more are common in the secondary stage.
A-ABS) ~ in which the test serum is preabsorbed Advantages
with a son­icate of the Reiter treponemes (sorbent) i. TPHA is just as specific as FTA-ABS and almost
to eliminate group specific reactions. FTA-ABS is as sensitive, except in the primary stage.
as specific as the TPI test and is now accepted as ii. This assay can be used to detect localized
a standard refer­ence test. However, as it can be production of antitreponemal antibodies in
done only in suitably equipped laboratories, it is cerebrospinal fluid.
not available for routine testing. iii. It is also much simpler and more economical.
iv. No special equipment is needed. Kits are
c. Fluorescent Treponemal Antibody- available commercially.
absorption (FTA-ABS) Test Microhemagglutination test (MHA-TP): TPHA
It is an indirect immunofluorescence test. Smears may be performed in microtiter plates and is
are prepared on slides with Nichol’s strain of T. called microhemagglutination test (MHA- TP).
pallidum as antigen. The slides can be stored for Hemagglutination teponemal test for syphilis
sev­eral months in deep freeze. The currently used (HATTS) is an automated conversion of TPHA test.
modi­fication of the test is the FTA-Absorption (FTA- The only disadvantage of MHA-TP and HATTS is that
ABS) test in which the test serum is preabsorbed they lack sensitivity in primary syphilis.
with a sonicate of the Reiter treponemes (sorbent)
to eliminate group specific reactions. After specific b. Enzyme Immunoassays (EIA)
antibody from the patient is allowed to react with Enzyme immunoassays (EIA) have been devel­
the organisms, unbound antibodies in the serum oped using T. pallidum antigens and are available
are removed by washing. The presence of anti-T commercially (Bio-Enza Bead test ; Captia
pallidum antibody is then detected by application Syphilis-G test). Assays that detect either IgM or

www.ebook3000.com
Chap-48.indd 339 15-03-2016 11:18:35
340  |  Section 3: Systemic Bacteriology
IgG are available and are increasingly replacing 4. Diagnosis of Congenital Syphilis
the TPHA and VDRL tests for routine screening. In the diagnosis of congenital syphilis it
is necessary to differentiate between passive
Treponema pallidum Particle Agglutination
transplacental trans­fer of maternal antibody to the
(TP-PA) fetus and production by the fetus of endogenous
The TP-PA test has largely replaced the antitreponemal antibody. As IgM does not cross the
microhemag­glutination assay for antibodies to placenta, its presence in neonatal serum confirms
T. pallidum (MHA-TP) test. The TP-PA test uses con­g enital syphilis. If IgM-FTA-ABS test gives
gelatin particles sensitized with T. pallidum a reactive test result with infant blood then it is
antigens instead of the sensitized red blood cells strong evidence of active congenital disease. Serial
used in the MHA-TP assay. The TP-PA test is testing is also useful because the titer of passively
simpler to perform than the FTA-ABS test, does transferred antibody decreases rapidly, the VDRL
not require an absorption step or expensive UV test becoming negative by three months.
microscope, and is more specific than the MHA-TP
test. Table 48.3 shows the relative sensitivities of Syphilis Associated with Human
the serological tests in common use.
Immunodeficiency Virus (HIV) Infections
Interpretation of Various Serological Tests HIV-infected patients who acquire syphilis may fail
to produce antitreponemal antibodies. This has
1. Serological Screening
extremely serious implications for the diagnosis
Because of their simplicity and accuracy, the and control of syphilis.
cardiolipin antigen tests are used as screening or
first ­line procedures for both routine diagnosis
Epidemiology
and mass screening programs. When used together,
the VDRL and TPHA tests provide a highly efficient Syphilis is found worldwide in distribution.
screen for the detection or exclusion of treponemal Natural syphilis is exclusive to humans and has
infection. no other known natural hosts. The most common
route of spread is by direct sexual contact. The
2. Response to Treatment disease can also be acquired congenitally or by
Reagin tests usually become negative 6–18 months transfusion with contaminated blood. Syphilis is
after effective treatment of syphilis, depending not highly contagious. High-risk sexual behavior
on the stage at which treatment is given. Serial and coinfection with HIV continue to complicate
quantitative VDRL testing provides the best means syphilis control efforts.
of measuring response to treatment in most stages
of treponemal infection. Treatment in the primary Immunity
stage leads to serore­versal in about four months; in The immune mechanisms in syphilis are not
the secondary and early latent stages, it takes 12–18 adequately understood. Humoral immune
months; in later stages, it may take five years or response against the treponeme does not appear
more. In some cases low titer reactivity may persist to be effective but Cell-mediated immunity may
indefinitely, in spite of effective treatment. Specific be more rele­vant.
treponemal tests tend to remain positive in spite of In a person already having active infection
treatment so they are of little value as indicators of reinfections do not appear to occur. It was believed
clinical cure. TPHA titers may fall rapidly following that premunition or infection immunity, as seen
treatment in sec­ondary syphilis but remain positive in some parasitic infections, holds good in syphilis
for life in low titers. also and that a patient becomes susceptible to
reinfection only when his original infection is cured.
3. Biological False-positive (BFP) Reactions
TPHA and FTA-ABS are helpful in excluding
Prophylaxis
or confirming the diagnosis of syphilis and for
identify­ing BFP reactions. 1. As transmission is by direct contact, it is possible
to protect against syphilis.
Table 48.3  Frequency of reactive serological tests 2. When this is not complied with, the use of
in untreated syphilis (percentage) in common use physical barriers (such as condoms), antiseptics
Stage VDR/RPR FTA-ABS TPHA (potassium permanganate) or antibiotics may
minimize the risk.
Primary 70–80 85–100 65–85
3. Educating people about sexually transmitted
Secondary 100 100 100
dis­eases.
Latent/late 60–70 95–100 95–100
4. Protective vaccines are not available.

Chap-48.indd 340 15-03-2016 11:18:35


Chapter 48: Spirochetes  | 341
Treatment 2. Yaws (Frambesia)
Penicillin is the drug of choice for treating Yaws is a disease that is endemic among rural
infections with T. pallidum. So far there have been populations in tropical and sub­tropical countries.
no reports of penicillin-resistance. In early cases, In India, cases of yaws have been identified in
a single injection of 2.4 million units of benzathine Andhra Pradesh, Orissa and Madhya Pradesh.
penicillin is adequate. For late syphilis, this may be
Causative agent : T. palli­d um subspecies
repeated weekly for three weeks. In pat­ients allergic
pertenue (still known as T. pertenue) which is
to penicillin, erythromycin or tetracy­cline may be
morphologically and antigenically indistin­
used. Only penicillin can be used for the treatment
guishable from T. pallidum.
of neurosyphilis.
Jarisch–Herxheimer reaction: Antibiotic Clinical Manifestations: The primary lesion
therapy of syphilitics, particularly with penicillin, (mother yaw) is an extragenital papule. It enlarges
characteristically induces a systemic response and breaks down to form an ulcerating granuloma.
called the Jarisch–Herxheimer reaction. This is As in syphilis, secondary and tertiary manifesta­
characterized by the rapid onset (within 2 hours tions follow but neurological and cardiovascular
fever, chills, myalgia, tachycardia, hyperventilation, involvement is rare. In the late stages of yaws,
vasodilatation and hypotension. It is believed to be ulcerative skin lesions and bone lesions develop.
due to the liberation of toxic products like tumor Transmission: Infection is acquired by direct
necrosis factors from the massive destruction of contact with open ulcers. Flies may act as
treponemes or due to hyper­sensitivity. mechanical vectors.
Laboratory diagnosis and treatment are similar to
NONVENEREAL TREPONEMATOSES
those of venereal syphilis. There appears to be some
Nonvenereal treponemal diseases are found in cross immunity be­between yaws and syphilis, in
developing countries where hygiene is poor, little that venereal syphilis is rare in communities where
clothing is worn, and direct skin contact is common yaws is endemic.
because of overcrowding. Infection is usually
transmitted by direct body to body contact. The 3. Pinta
three nonvenereal treponemal diseases are:
Pinta (carate, mal del pinto) is a contagious
1. Endemic syphilis inoculable disease endemic in Central and South
2. Yaws America and the neighboring islands. It is caused
3. Pinta by T. carateum which is morphologically and
antigen­ically indistinguishable from T. pallidum
1. Endemic Syphilis (Bejel) so cross immunity between pinta and syphi­lis is
Endemic syphilis or bejel sphilis, transmitted only partial.
nonvenereally, was endemic in several foci and Spread of infection is by direct contact with
mainly affects children in rural populations where infectious lesions. Transmission appears to require
living conditions and personal hygiene are poor. direct person to person contact. Incubation
ranges from 7 to 21 days.The pri­mary lesion is
The etiologic agent is T. pallidum subspecies an extragenital papule, which does not ulcerate
endemicum and closely resembles yaws in clinical but develops into a lichenoid or psoriaform.
manifestations.Transmission is by direct person- Secondary skin lesions are characterized by
to-person contact and by sharing of contaminated hyperpigmentation or hypo­pigmentation. The
eating or drinking utensils. laboratory diagnosis and treatment are similar
Clinical Manifestations: The primary chancre is to those of venereal syphilis.
not usually seen, except sometimes on the nipples
of mothers infected by their children. The disease NONPATHOGENIC TREPONEMES
is usually seen with mani­festations of secondary
syphilis, such as mucous patches in the mouth Several commensal treponemes occur on the
and skin eruptions. The dis­ease progresses to buccal and genital mucosa and may cause
tertiary lesions particularly syphi­litic gummata confusion in the di­agnosis of syphilis by dark field
on the skin, bone and nasopharynx. As in yaws, examination. Best known among them is the oral
the cardiovascular and central nervous systems spirochete, T. dentium, which can be readily cul­
are not involved, and congenital infection is tivated.
rare because of the early age of infection. The T. refringens and T.gracilis may be found as
laboratory diagnosis and treatment are as for normal commensal in genitalia. In experimental
venereal syphilis. work on T. pallidum in rabbits, T. paraluiscuniculi

www.ebook3000.com
Chap-48.indd 341 15-03-2016 11:18:35
342  |  Section 3: Systemic Bacteriology
(formerly, T. cuniculi), which has a very similar the development of immunity to all the anti­genic
appearance and causes natural venereal infection variants.
in rabbits, may pose problems. Agglutinating, complement fixing and lytic
antibodies develop during infection but their dem­
BORRELIA onstration is not possible as a routine diagnostic
Species of Borrelia test due to the difficulty in preparing satisfactory
Borreliae of medical importance are: antigens.
1. B. recurrentis causing relapsing fever
2. B. vin­centi sometimes causes fusospirochetosis Pathogenicity
3. B. burgdorferi—causative agent of Lyme Relapsing Fever
disease.
Relapsing (RF) is an arthropod-borne infection,
Morphology and two types of which occur ­louse-borne and
tick-borne. The borreliae causing them are
Borrelia are helical organisms 0.2–0.5 mm wide indistinguishable in morphology and many other
and 8–20 mm in length. They are gram-negative, features but differ in their arthropod hosts.
actively motile and possess 5–8 irregular spirals at
intervals of 2 mm with pointed ends (Fig. 48.3).
1. Epidemic or Louse-borne Relapsing Fever
Spirals are coarser and more irregular than those of
the treponemes or leptospires and usually can be The causative agent of louse-borne or epidemic RF
seen with light microscopy in preparations stained is B. recurrentis. It is an exclusive human pathogen,
with aniline dyes, such as Wright’s or Giemsa stains. being transmitted from person to person through
body lice (Pediculus humanus corporis). Humans
Cultural Characteristics are the only reser­voir for B. recurrentis.
Borrelia are microaerophilic. Optimum temperature
for growth is 28–30°C. The organism can be grown 2. Endemic or Tick-borne Relapsing Fever
on Noguchi’s medium (ascitic fluid containing The second form of relapsing fever is endemic and
rabbit kidney), chorioallantoic membrane (CAM) of tick-borne. It is caused by as many as 15 species
chick embryos and in mice or rats intraperitoneally. of borreliae and cause RF (B. dut­tonni, B. hermsii,
B. parkeri B. turicatae, etc.) and is spread by
Antigenic Properties infected soft ticks of the genus Ornithodoros that
An­tigenic variations: The most striking property vary according to the country where the infection
of relapsing fever is the capacity of Borrelia to occurs.
undergo several antigenically distinct variations
within a given host during the course of a single Pathogenicity
infection. This is believed to be the reason for the
After an incubation period of 2–10 days relapsing
occurrence of relapses in the disease. Ultimate
fever sets in as fever of sudden onset. During this
recovery after a number of relapses may be due to
period, borreliae are abundant in the patient’s
blood. The fever subsides in 3–5 days. Another bout
of fever sets in after an afebrile period of 4–10 days
during which borreliae are not demonstrable in
blood. The borreliae reappear in blood during the
re­lapses of fever. The disease ultimately subsides
after 3–10 relapses. Subsequent relapses are usually
milder and of shorter duration.

Epidemiology
The etiologic agent of louse-borne epidemic
relapsing fever is B. recurrentis. The vector is
the human bodylouse, and humans are the only
reservoir. The infection is transmitted not by the
bite of lice but by their being crushed and rubbed
into abraded skin.
Louse-borne relapsing fever tends to occur as
epidemics whenever poverty, overcrowding and
lack of personal hygiene encourage louse infesta­
Fig. 48.3: Borrelia recurrentis in peripheral blood smear tion.

Chap-48.indd 342 15-03-2016 11:18:36


Chapter 48: Spirochetes  | 343
Tick-borne endemic relapsing fever is a wide, with 3–8 coils of variable size. It is easily
zoonotic disease, with rodents, small mammals, stained with dilute carbol fuchsin and is gram-
and soft ticks (Ornithodoros species) the main res­ negative.
ervoirs and many species of Borrelia responsible for T. vincentii is a normal commensal of mouth.
the disease. The infection is transmitted to humans It may give rise to ulcerative gingivostomatitis
through the bite of ticks or their discharges. or oropharyngitis (Vincent’s angina) under
Several species of soft ticks belonging to the predisposing conditions such as malnutrition or
genus Ornithodorus act as vec­t ors. In India, viral infections, In Vincent’s angina, T. vincentii
the vector species are O. tholozoni, O. crossi., O. is often associ­ated with anaerobic gram-negative
lahorensis and the fowl tick, Argas ver­sicus. ‘fusiform bacillus known as Fusobacterium fusiforme
(now known as Leptotrichia buccalis). This symbiotic
Laboratory Diagnosis infection is known as fusospirochetosis.
Routine laboratory diagnosis of relapsing fever
Diagnosis
depends on demonstrating the spirochetes in
peripheral blood samples, either in the living state 1. Microscopic examination: Diagnosis may
or after staining. be made by demonstrating spiro­chetes and
fusiform bacilli in stained smears of exudates
i. Dark Ground Microscopy from the lesions.
2. Culture: B. vincentii may be cultivated with
During the pyrexial phase of the illness a drop of
difficulty in enriched media anaerobically.
blood may be examined as a wet film under the
dark ground or phase contrast microscope and Treatment: Penicillin and metronidazole are
borreliae detected by their lashing move­ments. effective in treatment.

ii. Giemsa or Leishman Stain Lyme Disease: Borrelia burgdorferi


Blood smears are stained with Giemsa or Leishman Lyme disease was identified in 1975. Lyme disease
stain and examined for borreliae. or Lyme borreliosis (originally Lyme arthritis), as
it was first observed in Lyme, Connecticut., USA.
iii. Animal Inoculation The disease is widespread in the USA. It is caused
Inoculate intraperitoneally 1–2 mL blood into by Borrelia burgdorferi, transmitted by the bite of
six young white mice. Examine drops of blood Ixodid ticks.
obtained by clipping the tip of the tail 48 hours later Morphology: It measures 4–30 mm × 0.2–0.25 mm.
and daily thereafter for up to 1 week. Examine by It is flexible, helical and gram-negative.
darkfield microscopy for living spirochetes or after Culture: It is a microaerophilic spirochete. It
staining by Giemsa. is fastidious bacterium and can be grown in a
modified Kelley’s (BSK-Barbour, Stonner Kelly)
iv. Culture and Serology medium, after incubation for two weeks or more.
Cultivation of the borreliae and demonstration of Optimum temperature for growth is 33°C.
antibodies are too difficult and unreliable to be
used in diagnosis. Patients with relapsing fever Pathogenesis
sometimes develop false-positive serological tests The natural hosts for B. burgdorferi are wild and
for syphilis. domesticated animals, including mice and other
rodents, deer, sheep, cattle, horses and dogs. B.
Prophylaxis burgdorferi is transmitted to man by ixodid ticks
Prevention of louse-borne relapsing fever consists that become infected while feeding on infected
of prevention of louse infestation along with the use animals. The bacterium grows primarily in the
of insecticides whenever necessary. midgut of the tick, and trans­mission to man occurs
Prevention of tick-borne disease is less easy and during regurgitation of the gut contents during the
consists of identification of tick infested places and blood meal.
their avoidance, or eradication of the vectors. No Stages of Lyme disease: Lyme disease may be a
vaccine is available. progressive illness, and is divided into three stages:
Treatment: Tetracyclines, chloramphenicol, Stage 1: The first stage of ‘localised infection’
penicil­lin, and erythromycin are effective. appears as a small red macule or papule at the site
of bite (erythema migrans or EM) after an incu­
Borrelia vincentii (Treponema vincentii) bation period of 3–30 days.
T. vincentii (old name Borrelia vincentii) is a motile Stage 2: The second stage of ‘disseminated
spirochete, about 5–20 mm long and 0.2–0.6 mm infection’ develops with headache, fever, myalgia

www.ebook3000.com
Chap-48.indd 343 15-03-2016 11:18:36
344  |  Section 3: Systemic Bacteriology
and lymphadenopathy. Some develop meningeal Morphology
or cardiac involvement. Leptospires are delicate spirochetes about 6–20
Stage 3: The third stage of ‘persistent infection’ mm long and 0.1 mm thick. They have numerous
sets in months or years later with chronic closely wound primary coils, so closely set together
arthritis, polyneuropathy, encephalopathy and that they are difficult to demonstrate in stained
acrodermatitis. preparations although they are quite obvious in the
living state by darkfield microscopy or by electron
Laboratory Diagnosis microscopy. Their ends are hooked and resemble
A. Isolation of the borrelia: The borrelia has been umbrella handles (Fig. 48.4). They are actively
iso­lated from ticks as well as from skin lesions, motile, Gram-negative, but take up conventional
CSF and the blood of patients, but culture is too stains poorly. They can be visualized by Giemsa or
slow and diffi­cult to be of use in diagnosis. silver deposition methods or by use of fluorescent
B. Serology: Serological tests such as ELISA and antibody.
immunofluorescence (IF) have been described
and immuno­b lotting recommended for Cultural Characteristics
confirmation. Antibodies take 1–2 months to They are aerobic and microaerophilic. Optimum
appear. temperature is 25–30°C and optimum pH 7.2–7.5.
False-positive syphilis serology may be seen, Leprospires can be grown in media enriched with
with FTA-ABS being positive and VDRL test rabbit serum.
negative. Liquid medium consists of Stuart’s or Korthof’s
medium. Semisynthetic media, such as EMJH
Treatment (Ellinghausen, McCullough, Johnson, Harris)
Penicillins, the newer macrolides, cephalosporins medium are now commonly used. A simple
and tetracyclines have all been used successfully in semisolid medium is Fletcher’s medium which
Lyme disease. consists of nutrient agar and rabbit serum. In
semisolid media, growth occurs characteristically
a few millimeters below the surface.
LEPTOSPIRA
Leptospires may be grown on the chorioallantoic
Introduction: Leptospires are actively motile, membrane (CAM) of chick embryos.
delicate spirochetes, possessing a large number of
closely wound spirals and characteristic hooked Resistance
ends. They are too thin to be seen under the light Leptospires are susceptible to heat; 10 min at
microscope (leptos, meaning fine or thin). They 50°C or 10 seconds at 60°C kills them. They are
may be visualized under dark ground illumination. also sensitive to acid and are destroyed by gastric
They do not stain readily. juice in 30 minutes. They are rapidly killed by bile
or trypsin.
Classification They are also readily destroyed by chlo­rine and
The family Leptospiraceae belongs to the order most other antiseptics and disinfectants. Their
Spirochetales and can be subdivided into three
morphologically indistinguishable genera:
Leptospira, Leptonema and Tumeria. Only
Leptospira spp. are considered to be pathogenic
for animals or man.
General characteristics: The genus Leptospira is
now classified into two species L. interrogans, and
L. biflexa. Pathogenic species are called Leptospira
interro­gans, and most saprophytic leptospires are
called L. bifiexa.
1. Leptospira interrogans: It comprises the
parasitic and pathogenic leptospires. L.
interrogans is classified into 23 serogroups
(Icterohemorrhagiae, Canicola, Pyrogen­es,
Autumnalis, Australis, Pomona, Hebdomadis,
Grippotyphosa, etc.). Within each serogroup
over 200 serovars are recognized. Fig. 48.4: Dark ground microscopy shows the appearance
2. L. bifiexa: It contains saprophytic leptospires. of living leptospires

Chap-48.indd 344 15-03-2016 11:18:36


Chapter 48: Spirochetes  | 345
Table 48.4  Important leptospiral infections
Serotype Disease Clinical picture Animal reservoir Distribution
Icterohemorrhagiae Weil's disease Fever, jaundice, Rat Worldwide
Canicola Canicola fever hemorrhages influenza like, Dog Worldwide
aseptic meningitis
Grippotyphosa Swamp or marsh Fever, prostration, aseptic- Field mice Europe, Africa,
fever men­i­ngitis SE Asia, USA
Pomona Swineherd’s disease Fever Pig America, Europe, Middle
East, Indonesia, Australia
Hebdomadis Seven day fever Fever, lymphadenopathy Field mice Japan, Europe, USA
Fortbragg Pretibial fever, Fever, rash over tibia Not known Japan, SE
Fort Bragg fever Asia, USA
Pyrogenes Febrile Fever Pig SE Asia, Europe, USA
spirochetosis
Bataviae Indonesian Weil’s Fever Rat SE Asia, Africa, Europe
disease
Hardjo Dairy farm fever Fever Cattle UK, USA, New Zealand

survival in water or soil depends on tempera­ Duration of the illness varies from less than
ture, acidity, salinity and nature and amount of 1 week to 3 weeks. Late manifestations may be
pollu­tion, dying rapidly in acid urine, nonaerated caused by the host immunologic response to the
sewage, saltish or brackish water. They can survive infection (Table 48.4).
for days in moist conditions at pH 6.8–8. Serious cases of leptospirosis are caused most
often by serotype icterohemorrhagiae, though
Antigenic Properties they m­ ay also be due to coenhageni and less
Leptospires exhibit considera­ble antigenic cross often batavitae, gripotyphosahosa, pyrogenes
reaction. A genus specific somatic antigen is and some others. Asep­tic meningitis is common
present in all members of the genus. Classi­fication in canicola infection and abdominal symptoms in
into serogroups and serotypes (now referred to grippotyphosa infections.
as sero-varieties or serovars) is based on sur­face
antigens. Laboratory Diagnosis
Diagnosis may be made by 1. Demonstration of
Pathogenesis the leptospires microscopically in blood or urine;
In natural reservoir hosts, leptospiral infection is 2. Isolation in culture; 3. Animal inoculation; 4.
asymptomatic. It is transmitted to humans when Serological tests.
the leptospires in water contaminated by the urine
of carrier animals enters the body through cuts or 1. Demonstration of Leptospiras in Blood
abrasions on the skin or through intact mucosa of
or Urine
mouth, nose or conjuctiva. During the acute phase
of the disease, leptospires are seen in the blood but Microscopy
can seldom be demonstrated after 8–10 days. They As leptospires disappear from the blood after the
persist in the internal organs, and most abundantly first week, blood examination is helpful only in
in the kidneys, so that they may be demonstrated in the early stages of the disease. Leptospires may be
the urine in the later stages of the disease. demonstrated by examination of the blood under the
Diseases: Mild virus-like syndrome. The incubation dark field microscope or by immunofluorescence
period is usually about 10 days (range 2–26 days). but this is of little practical value.
The onset of clinical illness is usually abrupt, Leptospires may be found in the urine during
with nonspecific, influenza-like constitutional the second week and intermittently for 4–6 weeks
symptoms such as fever, chills, headache, severe or even longer. Centrifuged deposit of the urine
myalgia, and malaise. may be examined under dark ground illumination.
Systemic leptospirosis with aseptic meningitis.
The patient may develop a more advanced disease- 2. Culture
including aseptic meningitis. Three or four drops of blood are inoculated into
Severe systemic disease (Well’s disease) each of several bijou bottles containing EMJH
includes renal failure, hepatic failure, and intra­ or similar medium. The bottles are incubated
vascular disease, and may result in death. at 37°C for two days and left thereafter at room

www.ebook3000.com
Chap-48.indd 345 15-03-2016 11:18:36
346  |  Section 3: Systemic Bacteriology
temperature in the dark for two weeks. Samples Pathogenic lepto­spires survive for long periods in
from the cultures are examined every third day the convoluted tubules of the kidneys in natural
for the presence of leptospires under dark ground hosts, multiply and are shed in the urine. Humans:
illu­mination. accidental end-stage host. People at risk are those
Direct culture of urine is seldom successful but exposed to urine-contaminated streams, rivers,
isolation is usually possible by inoculation into and standing water; occupational exposure to
guinea pigs. infected animals for farmers, meat handlers,
veterinarians.
3. Animal Inoculation When human beings come into contact with
such water, the leptospires enter the body through
The blood or urine from the patient is inoculated
abraded skin or mucosa and initiate infection.
intraperitoneally into young guinea pigs. The
Several animals act as carriers. Rats are particu­
animals develop fever and die within 8–12 days
larly important as they are ubiquitous and carry
with jaundice and hemorrhage into the lungs
the most pathogenic serotype icterohemorrhagiae
and serous cavities with virulent serotypes like
in the convoluted tubules of the kidney long-term,
icterohemorrhagiae.
resulting in chronic excretion of viable leptospires
in their urine. Field mice carry grippotyphosa, pigs
4. Serological Diagnosis pomona and dogs can­icola serotypes.
Tests for detection of leptospiral antibodies in sera
are of two kinds: i.Genus-specific tests; ii. Sero- Prophylaxis
group­specific tests General measures of prevention such as:
i. Rodent control
A. Genus-specific Tests ii. Disin­fection of water
iii. Wearing of protective clothing.
The antigens for these tests are prepared from the
nonpathogenic L. biflexa Patoc 1 strain. The tests Vaccination: Vaccination has been at­tempted with
employed include sensitized erythro­cyte lysis some success in dogs, cattle and pigs. Immunity
(SEL), complement fixation, agglutination and following vaccination or infection is sero­type
indirect immunofluorescence. ELISA has been specific. Vaccination has also been tried in per­sons
used to detect IgM and IgG antibodies separately, at high risk such as agricultural workers.
in order to indicate the stage of infection. A simple Treatment
and rapid dip-stick assay has been developed for
Penicillin is given intravenously (IV), 1–2 million
the assay of leptospira-specific IgM antibody in
units 6 hourly for 7 days in serious cases. For milder
human sera.
infections a 7–10-day course of oral amoxicillin is
appropriate. Patients allergic to penicillins can be
B. Serogroup-specific Tests
treated with erythromycin. Doxycycline 200 mg
Serogroup-specific tests are reactive mainly with orally once a week is effective in prophylaxis.
strains of the same serogroup as the infecting
strain. They comprise agglutination tests that Key Points
are either macroscopic, or microscopic. In ™™ Spirochetes are slender unicellular, motile, helical or
macroscopic agglutination tests, formalinized spiral rods. They are motile because of endoflagella.
suspensions of prevalent leptospira serovars are ™™ The members of genera Treponema, Borrelia and
Leptospira are pathogenic to man
tested for macro­scopic agglutination with serial
Treponema pallidum
dilutions of the test serum. ™™ Treponema pallidum is the causative agent of syphilis.
The micro­s copic agglutination test (MAT) ™™ T. pallidum is thin, coiled spirochete and is observed
generally uses live cultures of different serotypes by dark field or phase contrast microscopy
and agglutination is observed under the low power ™™ Diseases: Venereal syphilis. Syphilis is a sexually
transmitted disease
dark field microscope. This test is more specific and ™™ Lab diagnosis: Dark field microscopy is useful for
is generally accepted as “gold standard”. demonstration of treponemes in the clinical specimen.
Direct fluorescent antibody T. pallidum (DFA-TP) is a
Examination of Water for sensitive method for direct detection of treponemal
antigen in the exu­dates for diagnosis of syphilis
Pathogenic Leptospires Serological tests: Two major types of serologic tests
If a shaved and scarified area of the skin of a young exist: nontreponemal tests and treponemal tests.
guinea pig is immersed in water for an hour, In nontreponemal tests or Standard tests for
infection takes place through the abrasions. syphilis (STS) cardiolipin or lipoidal antigen is
used. Nontreponemal tests are Venereal Diseases
Epidemiology: Leptospirosis is most common Research Laboratory (VDRL) test and rapid plasma
zoonotic bacterial disease throughout the reagin (RPR). These are flocculation tests
world. Rodents are most important reservoirs.

Chap-48.indd 346 15-03-2016 11:18:36


Chapter 48: Spirochetes  | 347
4. Name the diseases spread by rats. Describe
™™ VDRL or RPR tests are used for screening or for
the pathogenicity and laboratory diagnosis of
diagnostic purposes of large number of sera
™™ Biological false positive (BFP) reactions may occur
leptospirosis.
in certain conditions because of use of nonspecific
antigen (cardiolipin) in STS MULTIPLE CHOICE QUESTIONS (MCQs)
™™ Treponemal tests in which treponemes are used 1. Spirochaetes exhibit:
as the antigen. These tests use live T. pallidum a. Flexion and extension motility
strains (e.g., T. pallidum immobilization test), killed b. Corkscrew like rotatory movement
T. pallidum (e.g., T. pallidum aggluti­nation test, T. c. Translatory motion
pallidum immune adherence test, and fluorescent d. All of the above
treponema antibody test), or T. pallidum extracts as
2. Treponema pallidum can be cultivated in:
antigens(e.g., Treponema pallidium hemagglutination
a. Thayer-Martin medium
(TPHA test) and enzyme immunoassay (EIA)]
b. Blood agar medium
™™ IgM-FTA-ABS test can detect IgM antibodies in
c. Chocolate agar medium
congenital syphilis and helps to differentiate it from
seropositivity due to passively transferred maternal
d. Rabbit testes
antibodies 3. Hard chancre is characteristic of:
™™ Endemic syphilis or bejel is caused by T. pallidum a. Primary syphilis
subspecies endemi­c um;Yaws by T. pallidum b. Secondary syphilis
subspecies pertenue and Pinta caused by Treponema c. Latent syphilis
carateum d. Tertiary syphilis
Borrelia 4. Which of the following serological tests is employed
™™ The important pathogenic borreliae of medical for diagnosis of congenital syphilis?
importance include B. recurrentis, B. vincentii and a. FTA-ABS test
B. burgdorferi. B. recurrentis is the causative agent b. IgM FTA-ABS test
of relapsing fever while B. vincentii causes vincent’s c. TPHA test
angina. Lyme disease is caused by B. burgdorferi d. Reiter protein complement fixation test
™™ Diseses: Epidemic relapsing fever: transmitted 5. The spirochete that can easily be cultured is:
person to person; vector-human body louse. a. Treponema pallidum
Endemic relapsing fever: transmitted rodents to b. Borrelia recurrentis
humans; vector-soft ticks c. Borrelia burgdorferi
™™ Lyme disease: Transmitted by hard ticks from mice d. Leptospira interrogans
to hu­mans; vectors ixodid ticks. 6. Jarisch-Herxheimer reaction is a complication
Leptospira observed following therapy with antibiotics in:
™™ They stain poorly with aniline dyes (e.g. Gram stain) a. Syphilis b. Relapsing fever
but stain well with silver impregnation methods c. Lyme disease d. Leptospirosis
(e.g., Levaditi’s and Fontana’s staining). They are 7. The causative agent of louse-borne relapsing fever
obligate aerobes
is:
™™ Diseases: Leptospirosis is most common zoonotic
a. Borrelia recurrentis b. B. duttoni
bacterial disease. Rodents are most important
c. B. Vincentii d. B. Burgdorferi
reservoirs. It causes-Mild virus-like syndrome;
Systemic leptospirosis with aseptic meningitis. 8. Erythema chronicum migrans is seen in:
Overwhelming disease (Weil’s disease) with vascular a. A. Syphilis b. Relapsing fever
collapse, thrombocytopenia, hemorrhage, and c. Lyme disease d. Leptospirosis
hepatic and renal dysfunction. 9. All the following diseases are transmitted non­
venereally except:
a. Syphilis
IMPORTANT QUESTIONS b. Pinta
c. Yaws
1. Classify spirochetes. Name different spirochetes d. Congenital syphilis
and diseases caused by them. Discuss the 10. Which serogroup of Leptospira interrogans is
laboratory diagnosis of syphilis. responsible for causing Weil’s disease?
2. Discuss pathogenesis of syphilis. a. Icterohemorrhagiae b. Hebdomadis
3. Write short notes on: c. Australis d. Canicola
a. Standard tests for syphilis (STS) 11. Which of the following serological tests is accepted
as gold standard for diagnosis of leptospirosis?
b. VDRL test
a. Microscopic agglutination test
c. Treponemal tests for serodiagnosis of syphilis b. Macroscopic agglutination test
d. Biological false positive (BFP) reactions. c. Both of the above
e. Fluorescent treponemal antibody-absorption 12. Which of the following tests can be performed for
(FTA-ABS) test. the serodiagnosis of leptospirosis?
f. TPHA (or) T. pallidum hemagglutination test. a. Macroscopic agglutination test
g. Nonvenereal treponematoses b. Microscopic agglutination test
h. Borrelia recurrentis c. Complement fixation test
i. Lyme disease d. All of the above
j. Leptospirosis ANSWERS (MCQs)
k. Weil’s disease 1. d; 2. d; 3. a; 4. b; 5. a; 6. a; 7. a; 8. c; 9. a; 10. a; 11. a; 12 d.

www.ebook3000.com
Chap-48.indd 347 15-03-2016 11:18:36
4949
Chapter

Mycoplasma and Ureaplasma

LEARNING OBJECTIVES

After reading and studying this chapter, you should pneumoniae, Mycoplasma hominis, and Ureaplasma
be able to: urealyticum
∙∙ Describe the general characteristics of the ∙∙ Analyze the diagnostic methods appropriate for
Mycoplasma spp. and how they differ from other detection of mycoplasmal and ureaplasmal infections
bacterial species ∙∙ Discuss the use of serologic tests for diagnosing M.
∙∙ Describe morphology and characteristies of pneumoniae infections
Mycoplasma spp ∙∙ Compare mycoplasmas and L forms
∙∙ Compare the clinical diseases caused by Mycoplasma ∙∙ Describe Ureaplasma urealyticum.

INTRODUCTION 14 species, of which Acholeplasma laidlawii


was the first to be named.
Mycoplasmas are the smallest prokaryotes capable
3. Family Spiroplasmataceae: It contains
of self replication. The first to be recognized,
the Genus Spiroplasma, which parasitize
Mycoplasma mycoides ssp. mycoides was
arthropods and plants.
isolated by Nocard and Roux in 1898 from cattle
with pleuropneumonia. As other pathogenic 4. Family Anaeroplasmataceae: Genus Anaero­
and saprophytic isolates accumu­lated from plasma, found in the rumen of cattle and sheep.
veterinary and human sources they were known 5. Family Entomoplasmataceae: It contains the
as pleuropneumonia-like organisms (PPLO). genera Entomoplasma and Mesoplasma found
‘Mycoplasma’ (Greek: mykes = fungus; plasma = in insects and plants.
something moulded) refers to the filamentous More than 60 species of Mycoplasma are known
(fungal-like) nature of the organisms of some to cause disease in a variety of mammalian, in­sect
species and to the plasticity of the outer membrane and plant hosts. About sixteen species, belonging
resulting in pleomorphism. to three families are found in human beings.

CLASSIFICATION General Characteristics


Mycoplasmas are members of the class 1. Mycoplasma and Ureaplasma, organisms are
Mollicutes (mollis, soft; kutis, skin), and the order the smallest free-living bacteria.
Mycoplasmatales. The class Mollicutes contains 2. Mycoplasmas differ from other bacteria in that
five families. they lack a rigid cell wall and due to absence
1. Family Mycoplasmataceae: It is subdivided of a cell wall are highly pleomorphic, with no
into two genera: fixed shape or size.
a. Genus Mycoplasma: It utilizes glucose or 3. These organisms are bounded by a soft
arginine but do not split urea. It has more trilaminar unit membrane containing sterols.
than 110 named species. 4. They lack even cell wall precursors like
b. Genus Ureaplasma: It hydrolyzes urea. It muramic acid or diaminopimelic acid.
has six species. 5. The mycoplasmas form pleomorphic filaments
2. Family Acholeplasmataceae: It contains a and many can pass through the 0.45 µm, filters
single genus. Acholeplasma. comprising at least used to remove bacte­ria from solutions.

Chap-49.indd 348 15-03-2016 11:19:13


Chapter 49: Mycoplasma and Ureaplasma  | 349
6. The organisms divide by binary fission (typical as a source of cholesterol and other lipids.
of all bacteria), grow on artificial cell—free Mycoplasmas may be cultivated in liquid or solid
media, and contain both RNA and DNA. media. A medium widely used pleuropneumonia
7. Mycoplasmas are generally slow-growing, high­ly like organisms (PPLO) broth for the isolation of
fastidious, facultative anaerobes requiring mycoplasmas consists of bovine heart to which
complex media containing cholesterol and fatty are added 20% horse serum and 10% fresh yeast
acids for growth. extract along with glucose and phenol red as a
8. Mycoplasma species grow embedded beneath pH indicator. This medium can be solidified by
the surface of solid media. On solid media, the addition of agar. Penicillin, ampicillin and
some species, (e.g. M. hominis) form colonies polymyxin B may be added in the medium to
with slightly raised centers giving the classic inhibit contaminating bacteria and amphotericin
“fried egg” appearance” . B to inhibit fungi. A diphasic medium in screw-
9. The mycoplasmas adhere to the epithelium capped bottle containing an agar phase that is
of mucosal surfaces in the respiratory and overlain with broth medium of similar composition
urogenital tracts and are not eliminated by may also be used. Colonies appear after incubation
mucous secretions or urine flow. for 2–6 days and are about 10–600 µm in size. On
10. The human mycoplasmas are susceptible to agar, colonies are typically biphasic that have a
ad­verse environmental conditions, such as ‘fried egg’ appearance, with an opaque central
heat and drying. zone of growth within the agar and a translucent
11. Transmission of mycoplasmas and ureaplas­ peripheral zone on the surface (Fig. 49.1).
mas in humans may occur via direct sexual Colonies may be seen with a hand lens but are
contact, from mother to child during delivery best studied after staining by Dienes method. For
or in utero, and by respiratory secretions or this, a block of agar containing the colony is cut
fomites in cases of M. pneumoniae infections. and placed on a slide. It is covered with a cover slip
on which has been dried an alcoholic solution of
Morphology methylene blue and azure.
Mycoplasmas are the smallest free living micro­ Colonies cannot be picked with platinum loops.
organisms. They can pass through bacterial Subculture is done by cutting out an agar block
filters. They lack cell wall but are bounded by a with colonies and rubbing it on fresh plates. Most
trilaminar membrane 8–10 nm thick which is rich Mycoplasma colonies are hemolytic.
in cholesterol and other lipids. They are very small
pleomorphic cells and range in size from 0.2 to 0.8 Biochemical Reactions
mm in diameter which may range from spherical 1. Mycoplasmas are mainly fermentative. Most
through coccoid, coccobacillary, ring and dumb- mycoplasmas use glucose (or other carbohy­
bell forms, to short and long branching , beaded drates) or arginine as a major source of energy.
or segmented filaments. The filaments are slender,
2. Urea is not hydrolyzed, except by ureaplasmas.
of varying lengths and show true branching. Myc­
oplasmas are gram-negative but are better stained 3. They are generally not prote­olytic.
by Giemsa stain. Replication is basically by binary
fission.
Mycoplasmas do not possess spores, flagella
or fimbria. Some mycoplasmas, including M.
pneumoniae, exhibit a gliding motility on liquid-
covered surfaces.

Cultural Characteristics
Most mycoplasmas are facultatively anaerobic but,
since organisms from primary tissue specimens
fre­quently grow only under anaerobic conditions,
an atmo­sphere of 95% nitrogen and 5% carbon
dioxide is preferred for primary isolation. They
grow within a temperature range of 22–41°C, the
parasitic species growing optimally at 35–37 °C and
the saprophytes at lower temperatures.
Media for cultivating Mycoplasma are enriched
with 20% horse or human serum and yeast extract. Fig. 49.1: Typical large Mycoplasma colony showing ‘fried
The high concentration of serum is necessary egg” appearance

www.ebook3000.com
Chap-49.indd 349 15-03-2016 11:19:14
350  |  Section 3: Systemic Bacteriology
Antigenic Structure children and young adults are especially susceptible
The surface antigens presented to the infected host to infection. Clinical disease is uncommon in very
are components of the cell membrane and consist young children and older adults. Other groups at
of glycolipids and proteins. risk include closed-in populations such as prisoners,
college students and military personnel. Epidemics
A. Glycolipid antigen: The glycolipid antigens of
are known to occur in these populations. Infection
M. pneumoniae are major membrane antigens
is not considered seasonal, but many cases occur
found in the lipid fraction. The purified
in autumn and early winter. Transmission is proba­
glycolipids act as antigens in the complement-
bly through aerosol droplet spray produced while
fixation test and other in vitro antigen–antibody
coughing.
reactions.
Pneumonias not attributable to any of the
B. Protein antigens: During the course of
common bac­terial causes were labeled historically
infection a number of protein antigens are
as primary atypical pneumonias. Eaton (944) was
recognized. Among them are two major surface
the first to isolate the causative agent of the disease
antigens, one of which is the PI protein that
and because it was filterable, it was considered
mediates attach­ment. Antibody to these surface
to be a virus (Eaton agent). The most important
proteins is found consistently in convalescent
species is Mycoplasma pneumoniae (also called
human sera and in respiratory secretions by
Eaton’s agent after the investigator. Who originally
radioimmunoprecipitation, gel electrophoresis,
isolated it.
and Western blot analysis.
The cold agglutinins that are induced in The disease has an incubation period of 1–3
patients with M. pneumoniae infection are directed weeks, and early symptoms are nonspecific,
against the I antigen deter­minant of erythrocytes, consist­ing of headache, low-grade fever, malaise,
which is also present on the surface of other blood and anorexia. Sore throat, dry cough, and earache
cells. are accompanying symptoms. However, patients
usually do not appear seriously ill and few warrant
Resistance: Mycoplasmas are killed by heating at admission to hospital.
56°C for 30 minutes. For long-term preservation, The disease is characterized by paucity of
lyophilization or freezing broth cultures at-70°C respi­ratory signs on physical examination but
(or in liquid nitrogen) are the preferred storage radiological evidence of consolidation, which is
methods. usually patchy involving one of the lower lobes,
M. pneumoniae can grow in the presence of starting at the hilum, and fanning out to the
0.002% methylene blue in agar, while many other periphery. About 20% of patients suffer bilateral
species are inhibited. Antiseptic solutions, such as pneumonia, but pleurisy and pleural effusions
chlorhexidine and cetrimide inhibit the growth of are unusual. The disease is usual­ly self-limited,
mycoplasmas.­ recovery occurring in 1–2 weeks, but can be
They are resistant to penicillin and cepha­ prolonged.
losporin as well as to lysozymes that act on the Extrapulmonary complications, including
bacte­rial cell walls but are sensitive to tetracycline cardio­vascular, central nervous system, dermato­
and many other antibiotics that inhibit protein logic, and gastrointestinal problems, are rare
synthesis. occurrences. Secondary complications include otitis
media, erythema multi­forme (Stevens–Johnson
Pathogenicity syndrome), hemolytic ane­mia, myocarditis, peri­
Parasitic mycoplasmas exhibit host specificity. They carditis, and neurologic abnor­malities.
generally produce surface infections by adhering
to the mucosa of the respiratory, gastrointestinal
Epidemiology
and genitourinary tracts. Mycoplasma causes
two types of diseases in humans—peumonia and M. pneumoniae is worldwide in its distribu­
genital infections. tion and is found at all ages. Transmission is by
droplets of nasopharyngeal secretion. Disease is
A. Mycoplasma pneumoniae most common in school aged children and young
Infection with M. pneumoniae typically produces adults (5–15 years). Close contact is necessary for
mild upper respiratory tract disease. More transmission, and the infection usually occurs
severe disease with lower respi­r atory tract among classmates or family members. Spread is
symptoms occurs in less than 10% of pa­tients. favored by close contact, as in families and most
Tracheobronchitis can occur. Pneumonia typically among military recruits. Mycoplasma may
(referred to as primary atypical pneumonia or remain in the throat for two or more months after
walking pneumonia) can also develop. School age recovery from the disease.

Chap-49.indd 350 15-03-2016 11:19:14


Chapter 49: Mycoplasma and Ureaplasma  | 351
Laboratory Diagnosis
Laboratory diagnosis Mycoplasma pneumoniae
infections may be established either by isolation of
the Mycoplasma or by serological methods.

1. Specimens
M. pneumoniae may be recovered from throat
swabs, nasopharyngeal swabs, sputum, throat
washings, bronchoalveolar lavage, tracheal
aspirate and lung tissue specimens.

2. Culture
In the laboratory, if inoculation is not possible
immediately, then the specimen may be held up
to 24 hours at 4°C. If delay more than 24 hours is
expected, then the specimen should be frozen at
Fig. 49.2: Typical large Mycoplasma pneumoniae colony
– 70°C. A widely used isolation medium contains showing “mullberry shaped” appearance. (Courtesy
bovine heart infusion (PPLO broth) with fresh Dr Surinder Kumar, Director Professor, Department of
yeast extract and horse serum supplemented with Microbiology, Maulana Azad Medical College, New Delhi, India)
penicillin (to inhibit other bacteria), thallium
ii. Species identification: These colonies may
acetate, glucose and with phenol red as a pH
be identified by:
indicator because mycoplasmas do not produce
turbidity in broth media. For genital Mycoplasma, a. Hemadsorption test: The characteristic
polymymin B and amphotericin B and lincomycin of guinea pig red blood cells is adhering to
are also added to mycoplamal broth. colonies of M. pneumoniae.
b. Tetrazolium reduction test: Colonies of
1. Isolation and Identification M. pneumoniae appear red when these
In general, the growth of M. pneumoniae from are flooded with solution oftetrazolium
clinical specimens is detected by the ability of compound which is colourless. M.
these organisms to produce acid from glucose. pneumoniae reduces tetrazolium (colour­
Most broth media are dipha­sic such as methylene less) to red coloured compound.
blue-glucose dipha­sic medium. Broth cultures are c. Serological techniques: Identification of
incubated at 35°C with the caps tightened. Tubes isolates can be con­firmed by inhibition of
are inspected daily for color changes (from salmon their growth with specific antisera.
to yellow) in the medium. A slight, gradual shift in
the pH indicator over an 8–15 day period without 2. Polymerase Chain Reaction (PCR)
gross turbidity suggests a true-positive culture. Amplification
The broth must be subcultured to appropriate agar
M. pneumoniae in respiratory exudates or secretions
me­dium as soon as color changes in the medium
is detected by polymerase chain reaction (PCR)
are appar­ent. Inspection of the agar surface under
amplification of a chosen sequence in its genome.
the low power of the microscope will reveal small
colonies of the organisms. In the absence of
obvious color change in diphasic media, a blind 3. Antigen Detection Techniques
subculture to agar media should be performed after Detection of antigen in respiratory exudates by
1 and 3 weeks of incubation. direct immunofluorescence and counterimmuno­
i. Colonies: M. pneumoniae may take 21 days or electrophoresis techniques, immunoblotting
more, while M. hominis colonies may appear with monoclonal antibodies and antigen capture
within 2–4 days. Colonies of M. pneumoniae are enzyme immunoassay (EIA).
small, betahemolytic and have a homogeneous
granular appearance (“mulberry shaped”), 4. DNA Probes
unlike the fried-egg morphology of other
DNA probes can be used to detect M. pneumoniae
mycoplasmas. Because the organisms do not
RNA in sputum specimens.
stain well with gram or acridine orange stains,
Mycoplasma ­like colonies are stained with the
Dienes or methylene blue stains. M. hominis 5. Serological Tests
colonies have a typical large “fried egg” Serologic tests are available only for M. pneumoniae
appearance (Fig. 49.2 ). Serological diagnosis may be made by:

www.ebook3000.com
Chap-49.indd 351 15-03-2016 11:19:14
352  |  Section 3: Systemic Bacteriology
A. Specific tests using mycoplasmal antigens rubella, adenovirus infections, psittacosis, tropical
B. Nonspecific methods. eosinophilia, trypanosomiasis, cirrhosis of liver,
paroxysmal hemo­g lobinuria and haemolytic
A. Specific Tests Using Mycoplasmal Antigens anemia.
The development of antibody to M. pneumoniae by
infected subjects may be measured by a range of Treatment
techniques of widely differing sensitivity, viz. com­ Tetracyclines and erythromycin are drugs of choice
plement fixation, metabolic inhibition, inhibition for the treatment of Mycoplasma and Ureaplasma
of tetrazolium reduction, immunofluorescence on infections. Patients with NGU should receive one
sections of chick embryo lung, direct or antibody of the tetracy­clines and ureaplasmas resistant to
capture enzyme- immunosorbent assays (EIA), or tetracyclines, patients should then be treated with
agglutination of antigen-coated erythrocytes, latex erythromycin, to which most tetracycline-resistant
or gelatin particles. ureaplasmas are sensitive.
i. Complement fixation test : Detection of
antibodies directed against M. pneumoniae by Infections in Immunocompromised Patients
complement fixation is a useful. Titers peak in 4 M. pneumoniae may cause severe pneumonia
weeks and persist for 6–12 months; diagnostic titer in immunodeficient patients and may persist
is ≥ 1: 32, or a fourfold increase. for many months in the respiratory tract of
hypogammaglobulinemia patients, despite
ii. Enzyme-linked immunosorbent assays: The
apparently adequate treatment.
tests are easier to perform and are highly sensitive
if both immunoglobulin M (IgM) and IgG are mea­ Mycoplasma and HIV Infection
sured.
Mycoplasmas tend to cause more severe and
prolonged infections in the human immuno­
B. Nonspecific Serological Tests deficiency virus (HIV) infected and other
The nonspecific serological tests are Streptococ­cus immunodeficient subjects. A synergetic effect
MG and cold agglutination tests. had been proposed between HIV and Mycoplasma
i. Sterptococcus MG test (particularly M. fermentans).
It is done by mixing serial dilutions of the patient’s
un­heated serum and a heat killed suspension of MYCOPLASMA AS CELL CULTURE
Strepto­coccus MG, and observing agglutination after CONTAMINANTS
overnight incubation at 37°C. A titer of 1 : 20 or over
is considered suggestive. Few primary cell cultures become infected with
mycoplasmas, but continuous cell lines do so
ii. Cold agglutination test
frequently. The contamination may originate from
The cold agglutination test is based on the appear­
the worker or from animal sera or trypsin used in cell
ance in a high proportion of cases with primary
culture. The mycoplasmas most responsible are M.
atypical pneumonia, of macroglobulin antibodies
arginini. M. fermentans, M. hyorhinis, M. orale, M.
that agglutinate human group O cells at low
salivarium and A. laidlawii.
temperature. Cold agglutinins appear about one
week after infection with a peak at 4–5 weeks. Contamination generally does not pro­duce
Thereafter, titer declines rapidly and the test beco­ cytopathic effects but may interfere with the
mes negative after about 5 months. growth of viruses in such cell cultures and may
This test is easily performed by mixing serial also produce misleading results in serological
dilutions of the patient’s serum with an equal volume tests. Eradication of mycoplasmas from infected
of a 0.2% washed human O group erythrocytes and cells is difficult.
clumping observed after incubating the mixture
overnight. The test is based on the development MYCOPLASMAS AND L FORMS OF
of hemagglutination, which can be reversed by
placing the tubes at 37oC. A titer of 1 : 32 or over
BACTERIA
is suggestive but demonstration of rise in titer in Kleineberger (1935) found pleuropneumonia-like
paired serum samples is more reliable. Because forms in a culture Streptobacillus moniliformis
paired sera are not always available, a single and termed them L forms, after Lister Institute,
antibody titer of 64–128 or more with either test, in London, where the observation was made. It was
a suggestive clinical setting, should be sufficient to subsequently shown that many bacteria, either
institute therapy. spontaneously or induced by certain substances
Cold agglutinins are occasionally induced in like penicillin, lost part or all of their cell wall
other diseases such as infectious mononucleosis, and develop into L forms. Such L forms may

Chap-49.indd 352 15-03-2016 11:19:14


Chapter 49: Mycoplasma and Ureaplasma  | 353
be ‘unstable’, when they revert to their normal B. Urogenital Infections
morphology, or ‘stable’ when they continue in the M. hominis, M. genitalium, M. fermentans, M.
cell wall deficient state permanently. penetrans, M. salivarium, M. spermatophilum and
Cell wall deficient forms (L forms, protoplasts, ureaplasmas have been isolated from urogenital tract.
spheroplasts) may not initiate disease but may be Genital infections are caused by U. urealyticum
important in bacterial per­sistence during antibiotic and M. hominis. They are transmitted by sexual
therapy and subsequent recurrence of the infection. contact, and may cause urethritis, proctitis,
balanoposthitis and Reiter’s syndrome in men,
Differenecs between L-forms and and acute salphingitis, pelvic inflammatory
Mycoplasma disease, cervicitis and vaginitis in women. They
It has been suggested that mycoplasmas may have also been associ­ated with infertility, abortion,
represent stable L forms of bacteria. L-forms also postpartum fever, chor­ioamnionitis and low birth
produce ‘fried egg’ colonies like Mycoplasma, but weight of infants.
they differ in the following respects:
1. L-forms are not filterable. Mycoplasma hominis
2. Do not require sterol for growth. M. hominis is found in the lower genitourinary
3. The remnants of cell wall components can be tracts of approximately 50% of healthy adults and
demonstrated in L-forms though they lack cell has not been reported as a cause of nongonococcal
walls. urethritis (NGU). The organism may, however,
invade the upper genitourinary tract and cause
4. The stable L-forms continue in the cell wall
salpingitis, pyelonephritis, pelvic inflammatory
deficient state permanently but resemble
disease (PID), or postpartum fevers.
parent bacteria both biochemically and
antigenically.
Mycoplasma genitalium
5. L-forms play an important role in persistence
of chronic infection during antibiotic therapy M. genitalium has been associated with some
and subsequent recurrence of the infection but cases of non-gonococcal urethritis and pelvic infla­
may not initiate disease. mmatory disease.
6. Nonpathogenic to laboratory animals.
As agglutinins to Streptococcus MG are
Ureaplasma urealyticum
frequently detected following infection with M. Some strains of mycoplasma frequently isolated
pneumoniae, it is thought that the latter is an from the urogenital tract of human beings and
L-form for the former, but several investigations animals form very tiny colonies, generally 15–50
of postulated L-phase-bacterial relationships (e.g. µm in size. They were called T strain or T
Strep­tococcus MG and M. pneumoniae) have failed form mycoplasmas (T for tiny) because of the
to show the same G + C ratio or sequence homology small colonies they produce. They are peculiar
between the genomes of the pairs of organisms. in their ability to hydrolyze urea, with the
production of ammonia, which is an essential
Atypical Pneumonia growth factor in addition to cholesterol. Human
T strain mycoplasmas have been reclassified as
Atypical pneumonia was defined as lower
Ureaplasma urealyticum.
respiratory infection that did not resemble classic
lesions that had been described. Around the
turn of century, any patient who pre­sented with Clinical Manifetations
a sudden onset of fever with shaking chills, 1. U. urealyticum may cause nonchlamydial,non-
pleuritic pain and the production of rust-colored gonococcal urethritis (NGU), epididymitis,
sputum was thought to have typical pneu­monia vaginitis and cervicitis.
attributable to Streptococcus pneumoniae. All 2. They may cause chorioamnionitis, prematurity,
other patients who did not show this characteristic postpartum endometritis, chronic lung disease
picture were referred to as patients with ‘atypical of the premature infant and infection of wounds
pneumonia or walking pneumonia. The primary and soft tissues.
pathogen responsible for atypical pneumonia 3. Ureaplasmas are the most common organisms
are Mycoplsasma pneumoniae, chlamydophia isolated from the central nervous system (CNS)
pneumoniae and Legionella pneu­mophila. or lower respiratory tract of sick premature or
Atypical pathogens do not respond to β-lactam newborn infants.
antibiotics and their presence is often overlooked 4. Ureaplasmas have also been blamed to cause
until patients fail to respond to standard penicillin male and female infertility and low birth weight
or cephalosporin therapy. but there are conflicting reports.

www.ebook3000.com
Chap-49.indd 353 15-03-2016 11:19:14
354  |  Section 3: Systemic Bacteriology

Key Points b. Mycoplasma pneumoniae or PPLO or Eaton’s


agent.
™™ Mycoplasmas are the smallest free-living bacterium. c. Streptococcus MG agglutination test.
They were previously named as pleuropneumonia like d. Cold agglutinin test for Mycoplasma infection
organisms (PPLO) e. Ureaplasma urealyticum.
™™ Absence of cell wall and a cell membrane containing f. Atypical pneumonia.
sterols are unique among bacteria. Due to lack of
rigid cell wall, they are extremely pleomorphic. MULTIPLE CHOICE QUESTIONS (MCQs)
Three-layer (trilaminar) cell membrane contains
sterols 1. Which of the following media is/are used for
™™ Mycoplasmas may be cultivated in liquid or solid isolation of Mycoplasma?
media A medium widely used for the isolation of a. PPLO broth
mycoplasmas is PPLO broth. This medium can be b. PPLO agar
made solid by the addition of agar. On agar, colonies c. Both of the above
are typically biphasic that have a ‘fried egg’ ppearance d. None of the above
™™ Pathogenesis 2. The following Mycoplasma species produce fried-
Mycoplasma pneumoniae: Primarily infects children egg-like colonies except:
aged 5–15 years. Transmitted by inhalation of a. Mycoplasma hominis
aerosolized droplet. Strict human pathogen
b. Mycoplasma pneumoniae
™™ Diseases: Upper respiratory infections
c. Mycoplasma fermentans
Lower respiratory infections-, including tracheobron­
d. Mycoplasma genitalis
chitis and pneumonia (referred to as primary
atypical pneumonia or walking pneumonia) 3. Which of the following statements are true for
™™ Laboratory diagnosis: Laboratory diagnosis of Ureaplasma urealyticum except:
Mycoplasmal infections may be carried out by a. They do not hydrolyze urea
isolation of the organism or by serological tests b. They require supplementation with urea for their
growth
Culture: Test is slow and insensitive
c. They utilize arginine as an energy source
DNA probes can be used to detect M. pneumoniae
d. They are nonproteolytic
RNA in sputum specimens
4. Which of the following pathogens cause nongonoco­
Serology-nonspecific test: Streptococcus MG and
ccal urethritis except
cold agglutination tests are non-specific test used
a. Mycoplasma hominis
for detection of antibody in serum. Complement
b. Chlamydia trachomatis
fixation fixation test and enzyme immunoassay (EIA)
c. Ureaplasma urealyticum
are specific tests to detect antibody against M.
d. Mycoplasma genitalium
pneumoniae
5. Atypical pneumonia is caused by all the following
Ureaplasma urealyticum bacteria except:
™™ They were called T strain or T form mycoplasmas a. Streptococcus pneumoniae
(T for tiny) because of the small colonies, peculiar b. Chlamydia pneumoniae
in their ability to hydrolysis urea. Human T strain c. Legionella pneumophila
mycoplasmas have been reclassified as Ureaplasma d. Mycoplasma pneumoniae
urealyticum. 6. Which of the following tests can help in the laboratory
diagnosis of primary atypical pneumonia?
a. Cold agglutination test
IMPORTANT QUESTIONS b. Streptococcus MG agglutination test
c. Culture on Mycoplasma broth medium
1. Classify mycoplasmas. Describe the morphology, d. All of the above
cultivation pathogenicity and Laboratory diagnosis 7. Which of the following drugs is most suitable for
of Mycoplasma infections. the treatment of Mycoplasma and Ureaplasma
2. Discuss laboratory diagnosis of Mycoplasma and infections?
Ureaplasma infections. a. Penicillins b. Cephalosporins
3. Write short notes on: c. Tetracyclines d. Nalidixic acid
a. Morphology and general characters of Myco­ ANSWERS (MCQs)
plasma and Ureaplasma. 1. c; 2. b; 3. a; 4. b; 5. a; 6. d; 7. c

Chap-49.indd 354 15-03-2016 11:19:14


50
Chapter

Miscellaneous Bacteria

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Discuss rat-bite fever
be able to: ∙∙ Describe Klebsiella granulomatis and disease caused
∙∙ Describe morphology and culture characteristics by it
of Listeria ∙∙ Acinetobacter spp.
∙∙ Describe infections caused by Listeria monocytogenes
and their laboratory diagnosis

LISTERIA MONOCYTOGENES Biochemical Reactions


Organisms of the genus Listeria are nonsporing L. monocytogenes ferments glucose, maltose,
gram-positive bacilli. The genus contains eight L. rhamnose and alpha methyl D-mannoside,
species, but almost all cases of human listeriosis producing acid without gas. It is catalase positive.
are caused by L. monocytogenes. It grows in the presence of 0.1 % potassium tellurite,
10% salt and at pH 9.6.
Morphology
Various Species
Listeria monocytogenes is a small, coccoid, Gram-
positive bacillus measuring approximately 0.5 On the basis of a few tests the genus Listeria can
× 2–3 µm. They occur singly or in pairs which be divided into 8 spe­c ies (Table 50.1). Many
are often angled at the point of contact and may serovars have been rec­ognised. Two major tests of
resemble diphtheroids or diplococci. It exhib­its a differentiation of various species include D-xylose
characteristic, slow; tumbling motility when grown fermentation and CAMP test with Staph. aureus and
at 25°C but at 37°C is nonmotile. This is be­cause Rhodococcus equi. L. monocytogenes gives a positive
peritrichous flagella are produced by the bacil­lus CAMP test with Staph. aureus.
optimally at 20–30°C but only scantily or not at all
at 37°C. They are noncapsulate, nonsporing and Pathogenicity
nonacid-fast. Listeria monocytogenes is commonly ingested
in food. L. ivanovii and L. seeligeri have been
Cultural Characters associated with a very small number of human
Listeriae are aerobes and facultative anaerobes. infections.
They can grow over a temperature range of Experimental inoculation in rabbits causes
2–43°C, the optimum temperature for the growth a marked monoc ytosis (hence the name
is 35–37°C. They can grow on ordinary media monocytogenes). Monocytosis is a feature of human
containing fermentable carbohydrate, but growth listeriosis also. In­stillation into the eyes of rabbits
is better on blood agar or tryptose phos­phate agar. produces keratocon­junctivitis (Anton test).
After 24 hours incubation at 37°C, colonies are Human infection is believed to result from
0.5–1.5 mm in diameter, smooth, translucent and contact with infected animals, inhalation of
emulsifiable and non-pigmented. contaminated dust or ingestion of contaminated
On blood agar, L. monocytogenes develops milk or food. Hospital acquired infections have
zones of slightly hazy β-hemolysis. also been reported. L.monocytogenes produces

www.ebook3000.com
Chap-50.indd 355 15-03-2016 11:20:04
356  |  Section 3: Systemic Bacteriology
Table 50.1  Distinguishing characters of Listeria spp
Species Hemolysis CAMP test with Acid production from Nitrate
reduction
S t a p h . Rhodococcus D-Mannitol L-Rhamnose D-Xylose a-Methyl
aureus equi D-mannoside
L. monocytogenes + + −a − + − + −
L. innocua − − − − +/− − + −
L. ivanovii ++ − + − − + − −
L. seeligeri ± + − − − + +/− −
L. welshimeri − − − − +/− + + −
L. grayi − − − + +/− − NK +/−
L. murayi − − − + V − NK +
L. denitrificans − − − − − + NK +
a
Regarded as ±; + = Positive reaction; – = Negative reaction; V = Variable; NK = Not known

a hemolysin known as listeriolysin-O, which 3. Culture


is a virulence factor anti­g enically related to Specimens should be inoculated on blood agar,
streptolysin-O and pneumolysin. chocolate agar and tryptose phosphate agar, and
incu­bated at 35–37°C for 1–3 days. Greater success
Clinical Features in isolation is achieved if the materi­als are stored
1. Intrauterine and neonatal infection: Intra­ in tryptose phosphate or thioglycollate broth at 4°C
uterine infection of the fetus may result in and subcultures are done at weekly inter­vals for 1–6
abortion, stillbirth, premature delivery, or months (cold enrichment ).
acute-onset disseminated infection in the Blood agar shows small colonies surrounded
newborn infant (including the form known as by a narrow zone of b-hemolysis. The bacteria are
granulomatosis infantiseptica). Asymptomatic actively motile when grown at 25°C. The isolate is
infection of the female genital tract may cause identified by its morphology and biochemical tests
infertility. Meningitis or septicemia may occur (Table 50.1).
in neonates.
2. Adult and juvenile infection: It may cause Treatment
meningitis or meningoencephalitis, particularly Currently, penicillin or ampicillin, either alone
in neonates and in the elderly. or with gentamicin, is the treatment of choice for
3. Disease in healthy adults: Most Listeria infections infections with L. monocytogenes. Erythromycin
in healthy adults are asympto­matic or occur in the can be used in patients allergic to penicillin.
form of a mild influenza-like illness. Several food-
borne outbreaks of acute gastroenteritis with fever
have been described.
ERYSIPELOTHRIX RHUSIOPATHIAE
4. Other infections: Listeriosis may also present as The genus Erysipelothrix contains two species, of
abscesses, conjunctivitis, pharyngitis, urethritis, which Erysipelothrix rhusiopathiae is responsible
pneumonia, infectious mononucleosis like for human disease.
syndrome, endocarditis or septicemia. Morphology: E. rhusiopathiae is a slender, non-
motile, nonspor­ing, noncapsulated, straight or
Laboratory Diagnosis slightly curved, gram-positive rod, measuring 1–2
1. Specimens × 0.2-0.4 mm with tendency towards formation of
Blood, CSF, amniotic fluid, placenta, pus and long filaments.
biopsy material from the organs involved may be
collected. Specimens may also be collected from Cultural Characteristics
neonate, stillbirth or products of conception. It is microaerophilic on primary isolation but
on subcul­ture, grows as an aerobe or facultative
2. Microscopy anaerobe and growth is improved by 5–10% CO2.
If the Gram stain shows organisms, they are It can grow on nutrient agar but the growth is
intracellular and extracellular gram-positive cocco­ improved by added glucose, serum or blood. Black
bacilli. colonies are devel­oped in tellurite media.

Chap-50.indd 356 15-03-2016 11:20:04


Chapter 50: Miscellaneous Bacteria  | 357
Biochemical Reactions man and animals. They have been considered
E. rhusiopathiae ferments glucose without gas pro­ responsible for a typhoid-like fever, urinary
duction, and forms acid from fructose, galactose infections, infantile gastroenteritis and suppuration
and lactose. It is catalase negative, with negative in various parts of the body.
reactions for oxi­dase, indole production, urease,
nitrate reduction, Voges-Proskauer and methyl red CHROMOBACTERIUM VIOLACEUM
tests. H2S is produced in triple sugar-iron agar (TSI). Chromobacterium violaceum is a gram-negative,
nonsporing bacillus, motile by means of single
Pathogenicity polar and scanty lateral flagella.
Erysipelothrix is a natural parasite of many animals. It is a facultative anaerobe. It grow­s readily
Disease is common in swine but rare in humans. It on simple nutrient agar at 35–37°C including
causes various diseases and syn­dromes in animals, MacConkey agar producing violet pigment soluble
notably a form of erysipelas in pigs and arthritis in in ethanol and insoluble in water and chloroform.
pigs and sheep. It is catalase and oxidase positive and indole and
urease negative.
Human infection: Human infection is an occupat­
ional hazard and usually occurs on the hand or It occurs as a saprophyte in soil and water in
fingers of persons handling animals fish or animal the tropical and subtropical regions. It causes rare
products. but dangerous infection and consist of skin lesions
with pyemia and multiple abscesses.
Three forms of human infection with E. rhusi­
opathiae have been described: It is sensitive to aminoglycosides chloramphenicol
and tetracycline.
1. Localized skin infection (erysipeloid): Inocul­
ation of the organism through the skin causes
erysipeloid. FLAVOBACTERIUM MENINGOSEPTICUM
2. Generalized cutaneous form Flavobacterium meningosepticum is a gram-
3. Septicemia: Septicemia and/or endocarditis negative nonmotile rod. It can grow on nutrient
may occur. agar. It forms a yellow nondiffusible pigment after
incubation at room temperature for 48 hours. It
Laboratory Diagnosis is catalase- and oxidase-positive, proteolytic and
weakly fermentative.
1. Specimen: A full-thickness skin biopsy tissue
fluid from the periphery of the lesions. F. meningosepticum is a saprophyte whose
natural habitat is soil and moist environments,
2. Culture: Specimen is inoculated in for primary
including nebulizers. It may cause opportunistic
culture in glucose or serum broth. Subcultures
nosocomial infections, particularly in infants. It
care made on blood agar. Blood cultures
has been responsible for outbreaks of meningitis
are diagnostic in cases of septicaemia or
in newborn infants. Infection in adults leads to a
endocarditis. E. rhusiopathiae is identified by
mild febrile illness.
its microscopic and colonial appearances and
biochemical reactions confirm the diagnosis. It is usually sensitive to co­t rimoxazole,
novobiocin, rifampicin, clindamycin and cefoxitin.
Treatment: Penicillin G is the drug of choice.
E. rhusiopathiae is also highly susceptible to
DONOVANIA GRANULOMATIS (CALYM­
ampicillin, methicillin, piperacillin, cephalothin,
cefotaxime, ciprofloxacin and clindamycin. MATOBACTERIUM GRANULOMATIS) OR
KLEBSIELLA GRANULOMATIS
ALCALIGENES FAECALIS Morphology
Alcaligenes faecalis refers to gram-negative, short, K. granulomatis is a small, capsulate, gram-nega­
nonsporing bacilli, which are strict aerobes and tive, coccobacillus. Diagnosis can be made by
do not ferment sugars. They are motile by means demonstration of Donovan bodies in Wright-
of peritrichous flagella. They are usually oxidase Giemsa stained impression smears from the lesions.
positive. Nitrate reduction is variable. It does They appear as rounded cocobacilli, 1–2 µm, within
not ferment any of the sugars in peptone water. cystic spaces in large mononuclear cells. They
It produces alkaline reaction in litmus milk and show bipolar condensation of chromatin, giving a
sugar media. closed safety pin appearance in stained smears.
Alc. faecalis is a saprophyte found in water and Capsules are usually seen as dense acidophilic
soil contaminated with decaying organic matter. areas around the bacilli. They are gram-negative,
They are also commensals in the intestines of nonmotile, nonsporing and nonacid-fast.

www.ebook3000.com
Chap-50.indd 357 15-03-2016 11:20:04
358  |  Section 3: Systemic Bacteriology
Culture Pathogenesis
It can be cultured readily in the egg-yolk medium The organisms survive well in the hospital environ­
and on a modified Levinthal agar. ment, and are increasing in importance as opportunist
pathogens. Serious infections, including meningitis,
Pathogenicity pneumonia and septicemia, are most commonly
associated with Acinetobacter baumannii.
Donovanosis is a venereal disease, first described
by McLeod in India in 1882 and seen mainly in the Treatment
tropics. It can be transmitted after repeated exposure
through sexual intercourse or non­sexual trauma to Most strains are resistant to sulphonamides,
penicillins, erythromycin, the tetracyclines and
the genitalia. The incubation period ranges from
chloramphenicol. But empirical therapy for seri­ous
1 to 12 weeks. It begins as a painless papule on
infections should consist of a b-Iactam antibiotic (e.g.,
the genitalia, which leads to a slowly progressive,
ceftazidime, imipenem) and an aminoglycoside.
autoinoculable ulcers. The disease runs a chronic
course.It causes a chronic granulomatous disease RAT-BITE FEVER (STREPTOBACILLUS
known as granuloma inguinale, granulomatous
MONILIFORMIS AND SPIRILLUM MINUS)
venereum or donovanosis.
Streptobacillus moniliformis and Spirillum minus
Laboratory Diagnosis are the causative agents of two distinct diseases
referred to collectively as rat-bite fever. Rat-bite fever
Laboratory confirmation of granuloma inguinale (RBF) is characterized by relapsing fever, rash and
is made by scraping the border of the lesion, by arthralgia occurring days or weeks after a rat-bite.
spread­ing the collected tissue on a slide, and Two different bacteria can cause this condition—
by staining it with Giemsa or Wright’s stain. Streptobacillus moniliformis and Spirillum minus,
Pathognomonic Dono­van bodies are observed both of which are natural parasites of rodents.
within mononuclear phago­cytes.
Streptobacillus moniliformis
Treatment Morphology
Cases of donovanosis respond well to tetracycline, S. monili­formis is a long, thin (0.1–0.5 × 1–5 µm),
chloramphenicol, erythromycin, clindamycin, gram- negative bacillus that tends to stain poorly
cotrimoxazole, streptomycin and other aminogly­ and to be more pleomorphic in older cultures.
cosides. ­ Granules and bul­b ous swellings resembling a
string of beads may be seen hence the species
ACINETOBACTER (MIMA POLYMORPHA; name ‘moniliformis’. It may lose its cell wall and
BACTERIUM ANITRATUM exists as L-form. In fact, L-forms were originally
discovered during the study of this bacillus.
The genus Acinetobacter contains strictly aerobic
short, stout, often capsulate, nonmotile gram- Culture
negative bacilli or coccobacilli that grow well on S. moniliformis is a nutritionally exacting aerobe and
simple media. They are oxidase negative. facultative anaerobe. Growth requires the presence
The genus contains only one species, Acinet­ of blood or other body fluids. It grows well on moist
obacter calcoaceticus, which embraces two variants: Loef­fler’s serum slope or moist plate of nutrient
A. calcoaceticus var. anitratus produces acid agar containing 20% horse serum and in 20% serum
oxidatively from glucose whereas A. calcoaceticus broth. Colonies appear on the surface after 48 hours
var. lwoffii does not. incubation and are discrete granular and greyish
yellow. L phase variants show minute colonies
A. baumannii (0.1–0.2 mm diameter) with a fried egg appearance.
They con­sist mainly of small coccoid bodies.
These form pinkish colonies on MacConkey medium.
Acid without gas is formed in glucose, arabinose, Biochemical Reactions
xylose, and occasionally in rhamnose. A characteristic
It attacks sugars fermentatively and produces
reaction is the formation of acid in 10%, but not 1%
acid without gas from glucose, galactose, dextrin,
lactose. Several serotypes have been identified by
raffinose and starch. It is catalase, oxidase, indole
capsule swelling and immunofluorescence.
production, urease and nitrate reduc­tion negative.

A. lowffi Pathogenesis
This forms yellow colonies on MacConkey medium In humans, it causes streptobacillary rat-bite fever.
and does not acidify sugars. Some strains are Another type of clinically indistinguishable, rat-bite
oxidase positive. fever is caused by Spirillum minus.

Chap-50.indd 358 15-03-2016 11:20:04


Chapter 50: Miscellaneous Bacteria  | 359
In man, the organisms enter the body through S. minus is a natural parasite of wild rats and other
the wound caused by the bite of rat or other rodents. The organism is inoculated into humans
animals. The disease can also occur as outbreaks, through the bite of a rat. Local lymphadenopathy
in the absence of rat-bite. This condition, first and lymphangitis develop with the onset of fever and
observed in Haverhill, USA, is called Haverhill systemic disease. A generalized rash with large brown
fever or erythema arthriticum epi­demicum. It is to purple macules is usually observed, but some
believed to be caused also by consumption of raw patients present with urticarial lesions. A roseolar
milk or water contaminated by rat excrement. rash may spread from the area of the original bite.

Laboratory Diagnosis Laboratory Diagnosis


1. Specimens 1. Microscopic Examination
i. Blood—during acute phase of disease. Darkfield microscopy examination of blood, ulcer
ii. Joint fluid—when arthritis develops. exu­dates, or lymph node aspirates is used to detect
2. Culture: Specimen is inoculated on blood agar S. minus. Blood smears can be stained with Giemsa’s
or Loeffier’s serum slope. or Wright’s stain.
3. Animal pathogenicity test: Mice are highly
susceptible to intraperitoneal inoculation of 2. Animal Inoculation
infected material. The animals develop a rapidly S. minus can be isolated by intraperitoneal
fatal generalized condition or a more progressive inoculation of blood or material from lymph
disease with swelling of feet and legs. node into mice and guinea pigs. Spirilla may be
4. Serology: Diagnosis may also be established demonstrated by dark field microscopy in blood
by serological tests including agglutination, and peritoneal fluid of animal 1–3 weeks after the
complement fixation and fluorescent antibody tests. intraperitoneal inoculation.
Treatment Treatment
Penicillin is the antibiotic of choice for treating rat- Infections with S. minus respond to treatment with
­bite fever. S. moniliformis is also sensitive to cep­ peni­cillin and tetracyclines.
halosporins, erythromycin, clindamycin, tetracycline
and aminoglycosides. The L phase variant is peni­
EIKENELLA CORRODENS
cillin-resistant but sensitive to tetracycline.
Morphology
SPIRILLUM MINUS E. corrodens is a fastidious , small, non­motile,
The organism commonly known as Spirillum non-spore-forming, facultatively anaerobic
minus, one of the causes of rat-bite fever in man Gram-negative bacillus. It lacks flagella and shows
is of uncer­tain taxonomic position. It was once twitching motility which is due to contractile
regarded as a spirochaete but was later placed in fimbria-like filamentous appendages
the genus Spirillum.
Culture
Morphology It is an aerobe and facultative anaerobe. A slow-
S. minus is a short, spiral, gram-negative organism growing, fastidious organism, E. corrodens re­quires
about 3–5 μm × 0.2–0.5 μm in size, with two or 5–10% carbon dioxide to grow. It can grow on blood
three regular spirals and 1–7 amphitrichous agar or chocolate agar. After 48 hours incubation on
flagella. It is very actively motile, showing darting blood agar or chocolate agar, the colonies are small
movements like those of a vibrio.The organisms can (0.5–1 mm in diameter) with characteristic pitting
be demonstrated with the dark ground microscopy or corroding of blood agar, hence the species name
or by staining with Leishman or Giemsa stain. corrodens. The organism also produces a charac­
teristic bleach like odor.
Culture
The organism has not been cultivated on artificial Biochemical Characters
media. S. minus is best isolated by intra­peritoneal It is oxidase positive and catalase negative. It does
inoculation of specimens (infected tissue or blood) not produce acid from carbohydrates. It is indole
into mice and guinea pigs. and urease negative but lysine and ornithine
decarboxylase positive.
Pathogenesis
It was first observed in a rat by Carter (1888) Pathogenesis
in India. Japanese workers identified it as the Eikenella corrodens is normally present in the mouth,
causative agent of one type of RBF, called Sodoku. upper respiratory tract and gastrointestinal tract of

www.ebook3000.com
Chap-50.indd 359 15-03-2016 11:20:04
360  |  Section 3: Systemic Bacteriology
human beings. It is an opportunistic pathogen that are hemolytic on human or rabbit blood agar. It is
causes infections a human bite wound or fist fight catalase, oxidase, indole and urease negative.
injury, endocarditis, sinusitis, meningitis, brain G. vaginalis is considered responsible for
ab­scesses, pneumonia, and lung abscesses. bacterial vaginosis, a mild but common condition
characterized by raised vaginal pH > 4.5, foul smelling
Treatment discharge and the presence of ‘clue cells’, which are
E. corrodens is susceptible to penicillin, ampicillin, vaginal epithelial cells with their surface studded
extended-spectrum cephalosporins, tetracyclines, with numerous small bacteria. Bacterial vaginosis is
and fluoroquinolone. also associated with anaerobic bacteria, particularly
Mobiluncus. Metronidazole is effective in treatment.
CARDIOBACTERIUM HOMINIS
Key Points
This gram-negative bacilli are nonmotile, char­
™™ Listeria monocytogenes is a small, coccoid, Gram-
acteristically small (1 × 1–2 mm) but some­times
positive bacillus and exhib­its a characteristic, slow,
pleomorphic. The organism grows slowly in culture. tumbling motility when grown at 25 °C but at 37 °C
On blood agar the colonies are small, 1–2 mm in is nonmotile
diameter, smooth, glistening, circular and opaque ™™ The disease chiefly affects pregnant women, unborn
after 48 hours incubation at 35°C. The organism or newly delivered infants, the immunosuppressed
does not grow on MacConkey agar or other selective and elderly. It is predominantly transmitted by the
media commonly used for gram-negative bacilli. consumption of contaminated food
The bacteria are fermentative, indole- and oxidase- ™™ Erysipelothrix rhusiopathiae: Disease is common in
swine but rare in humans. Three forms of human
positive, and catalase and nitrate negative.
infection-erysipeloid, generalized cutaneous form
C. hominis occurs commonly as a commensal and septicemia
in the human nose and throat may cause endo­ ™™ Alcaligenes faecalis: It causes urinary tract infection,
carditis, particularly in those with pre-existing infantile gastroenteritis, and typhoid-like fever in
cardiovascular disease. humans
It can be diagnosed by isolation of the causative ™™ Chromobacterium violaceum: It is associated
with intestinal and genitourinary infections and
agent in blood culture.
septicemic illnesses with pneumonia
C. hominis is susceptible to multiple antibiotics, ™™ Flavobacterium meningosepticum: causes oppor­
and most infections are successfully treated with tunistic infections, neonatal meningitis in prema­ture
penicillin or ampicillin for 2–6 weeks. infants and pneumonia in immuno­compromised
hosts
CAPNOCYTOPHAGA ™™ Calymmatobacterium (Donovania) granulomatis
is the causative agent of granuloma inguinale, a
The Capnocytophaga species are gram-negative, sexually transmitted disease
slow-growing, capnophilic, fusiform or filamentous ™™ Acinetobacter species Acinetobacter species may
bacilli. They are fermentative and facultative cause nosocomial infections
anaerobes that require CO2 for aerobic growth. They ™™ Rat bite fever is caused by two different bacilli:
may show gliding motility which can be seen as Streptobacillus moniliformis and Spirillum minus
outgro­wths of colonies. ™™ Eikenella corrodens: Causes a human bite wound
or fist fight injury, endocarditis, sinusitis, meningitis,
C. ochracea, C. gingivalis and C. sputigena are
brain abscesses, pneumonia, and lung abscesses
members of the normal oral flora of humans. They
™™ Cardiobacterium hominis: C. hominis may cause
have been associated with severe periodontal disease endocarditis, particularly in those with preexisting
in juveniles. They occasionally cause bacteremia and cardiovascular disease
severe systemic disease in immunocompromised ™™ Capnocytophaga species have been associated
patients, especially granulocytopenic patients with with severe periodontal disease in juveniles,
oral ulcerations. bacteremia and severe systemic disease in immuno­
Most Capnocytophaga infections can be compromised patients
treated with broad-spectrum cephalosporins, ™™ Gardnerella vaginalis is considered responsible for
bacterial vaginosis, a mild but common condition
fluoroquinolones, or penicillin. characterized by raised vaginal pH >4.5, foul
smelling discharge and the presence of clue cells’.
GARDNERELLA VAGINALIS Metronidazole is effective in treatment.
Gardnerella vaginalis is a small, gram-negative,
nonmotile, pleomorphic rod which shows IMPORTANT QUESTION
metachromatic granules. It was formerly known as Write short notes on:
Haemophilus vaginalis or Corynebacterium vaginale. a. Listeria monocytogenes
It grows on blood or chocolate agar aerobically under b. Elysipelothrix rhusiopathiae
5% CO2, minute colonies appear in 24–48 hours and c. Donovan bodies

Chap-50.indd 360 15-03-2016 11:20:04


Chapter 50: Miscellaneous Bacteria  | 361
d. Rat-bite fever 3. Which of the following bacteria can cause rat bite
e. Eikenella corrodens fever?
f. Acinetbacter spp. a. Streptobacillus moniliformis
g. Alkaligenes faecalis b. Listeria monocytogenes
c. Chromobacterium violaceum
d. Flavobacterium meningosepticum
MULTIPLE CHOICE QUESTIONS (MCQs) 4. Causative agent of donovanosis is:
a. Klebsiella pneumonia
1. Tumbling motility is seen in: b. Klebsiella oxytoca
a. Listeria monocytogenes c. Klebsiella granulomatis
b. Enterobacter cloacae d. None of the above
c. Proteus vulgaris 5. Which of the following bacteria does not produce
pigment?
d. Salmonella Typhi
a. Staphylococcus aureus
2. In adults, listeriosis may lead to: b. Chromobacterium violaceum
a. Meningitis c. Flavobacterium meningosepticum
b. Meningoencephalitis d Listeria monocytogenes
c. Encephalitis ANSWERS (MCQs)
d. All of the above 1. a; 2. d; 3. a; 4. c; 5. d

www.ebook3000.com
Chap-50.indd 361 15-03-2016 11:20:04
51
Chapter

Rickett­siaceae, Bartonellaceae and Coxiella

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Discuss the laboratory diagnosis of rickettsial
be able to: infections
∙∙ Describe diseases caused by different rickettsiae ∙∙ Describe Weil–Felix reaction
∙∙ Describe Brill–Zinsser disease ∙∙ Discuss Q fever, Trench fever and Oroya fever.

INTRODUCTION Table 51.1  The family Rickettsiaceae causing


human diseases
Rickettsiae are small. Gram-negative bacilli
adapted to obligate intracelIular parasitism, and Genus Species
transmitted by arthropod vectors. A. Rickettsia R. prowazekii
R. typhi
The family Rickettsiaceae is named after R. rickettsii
Howard Taylor Ricketts who discovered the spotted R. conorii
fever rickettsia (1906) and died of typhus fever R. australis
contracted during his studies. These bacteria R. sibirica
were originally thought to be viruses because R. akari
they are small, stain poorly with the Gram stain, B. Orientia O. tsutsugamushi
and grow only in the cytoplasm of eukaryotic
cells. Nevertheless, these organisms have the GENUS RICKETTSIA
characteristics of bacteria:
Morphology
Characteristics Similar to Bacteria In smears from infected tissues, rickett­siae appear as
pleomorphic coccobacilli, 0.3–0.6 mm × 0.8–2 mm
1. They are structurally similar to gram-negative in size. They are nonmotile and noncap­sulated.
bacilli. They are gram-negative, though they do not take
2. Their cell wall contains muramic acid. the stain well. They stain bluish purple with Giemsa
3. They contain both DNA and RNA. and Castaneda stains and deep red with Machiavello
4. They contain enzymes for the Krebs cycle and and Gimenez stains.
ribosomes for protein synthesis.
5. They multiply by binary fission. Cultivation
6. They are inhibited by antibiotics (e.g. Rickettsiae are unable to grow in cell­free media.
tetracycline, chloramphenicol). The optimum temperature for growth is 32–35°C.
1. Yolk sac: They are readily cultivated in the yolk
sac of developing chick embryos.
CLASSIFICATION
2. Cell culture: They grow on mouse fibroblast,
The family Rickett­siacae currently comprises two HeLa, HEp-2, Detroit 6 and other continuous
genera­ Rickettsia and Orientia (Table 51.1). cell lines but tissue cul­tures are not satisfactory
Coxiella is now included in the family Coxiellaceae. for primary isolation.
Ehrlichia and Anaplasma are included in family 3. Labo­ratory animals, such as guinea pigs and
Anaplasmataceae. mice are useful for the isolation of rickettsiae

Chap-51.indd 362 15-03-2016 11:20:55


Chapter 51: Rickett­siaceae, Bartonellaceae and Coxiella  | 363
from patients. They may also be propagated in also transmit the infection but not the pubic louse.
arthropods. Unlike with most other rickettsial diseases, humans
are the primary reservoir of typhus.
Resistance The lice become infected by feeding on
They are rapidly de­stroyed at 56°C and at room rickettsiaemic patients. The rickettsiae multiply
temperature when sepa­rated from host components, in the gut of the lice and appear in the feces in
unless preserved in skimmed milk or a suspending 3–5 days. Lice defecate while feeding. Infection is
medium containing su­crose, potassium phosphate transmitted when the contaminat­ed louse feces
and glutamate (SPG me­dium). Rickettsiae are is rubbed through the minute abrasions caused
susceptible to tetracycline, chlo­ramphenicol and by scratching. Occasionally, infection may also be
ciprofloxacin. transmitted by aerosols of dried louse feces through
inhalation or through the conjunctiva.
Antigenic Structure The incubation period typically ranges from
Rickettsiae possess at least three types of antigens: 10 to 14 days. Usually the onset is abrupt, with
1. Group specific soluble antigen: It is present generalized myalgias, chills, fever and headache.
on the surface of rickettsiae. Other symptoms, such as gastrointestinal comp­
2. Species specific antigen: It is associated laints, weakness, cough and meningismus may also
with the bodies of rickettsiae. In case of scrub be present and lead to diagnostic confu­sion. One of
typhus, it is strain specific. the hallmarks of epidemic typhus, skin rash, usually
3. Alkali stable polysaccharide: It is an alkali appears from 4 to 7 days after onset characteristically
stable polysaccharide found in some rickettsiae first on the trunk and later spreads to the extremities.
and in some strains of Proteus bacilli. This The pa­tient becomes stuporous and delirious
sharing of antigens between rickettsiae and towards the second week and develops the cloudy
pro­teus is the basis for the Weil–Felix reaction state of consciousness in the disease (from typhos,
used for the diagnosis of rickettsial infections meaning cloud or smoke). The case fatality may
by the demon­stration of agglutinins to Proteus reach 40% and increases with age.
strains OX 19, OK 2 and OK.
2. Brill–Zinsser Disease (Recrudescent Typhus)
Pathogenesis The rickettsiae may remain latent in the lymphoid
Rickettsiae normally enter the body through tissues or organs for years in some who recover
the bite or feces of an infected arthropod vector. from epidemic typhus. Such latent infection may, at
On entry into the human body, the rickettsiae times, be reactivated leading to recrudescent typhus
multiply lo­cally and enter the blood. They become (Brill–Zinsser disease). Brill (1898) first recognized
localized chiefly in the vascular endothelial cells, and Zinsser (1934) isolated R. prow­azekii from such
which en­large, degenerate and cause thrombus cases and proved that they were recrudescences of
formation, with partial or complete occlusion of infections acquired many years previously.
the vascular lumen. Brill–Zinsser disease is a milder illness than that
of epidemic typhus and the duration of the disease
A. Typhus Fever Group is shorter. Case fatality is lower.
Typhus group rickettsiae cause:
1. Epidemic typhus 3. Endemic Typhus (Murine Typhus, Flea-
2. Brill–Zinsser disease borne Typhus, Rat Typhus)
3. Endemic typhus (Murine typhus). Endemic or murine typhus is a milder disease than
epidemic typhus.
1. Epidemic Typhus (Louse-borne Typhus, It is caused by Rickettsia typhi (R. mooseri).
Classical Typhus) The primary reservoir is rodent, and the
Epidemic typhus is a louse-borne disease and principal vector is the rat flea (Xenopsylla cheopis).
had a tremendous impact on the history of man. Most cases occur dur­ing the warm months.
The disease has been reported from all parts of The rickettsia multiplies in the gut of the flea
the world but has been particularly common in and is shed in its feces. The flea is unaffected but
Russia and Eastern Europe. In India, the endemic remains infectious for the rest of its natural span
spot is Kashmir. of life. Man is infected by the contamination of
The etiologic agent of epidemic typhus is-R. abraded skin, respiratory tract or conjunctiva
prowazekii. with infective flea feces. Ingestion of food recently
The principal vector is the human body louse contaminated with infected rat urine or flea feces
Pediculus humanus cor­poris. The head louse may may also cause infection. Human infection is a

www.ebook3000.com
Chap-51.indd 363 15-03-2016 11:20:55
364  |  Section 3: Systemic Bacteriology
dead end. Man to man transmission does not fever group is widespread vasculitis involving the
occur. In Kashmir and China, lice have been known skin (with production of a rash) and internal organs
to transmit endemic typhus in human beings, (producing dysfunctions of the brain, heart, lungs,
producing smouldering outbreaks. Endemic and kidneys).
typhus is worldwide in prevalence.
Differences between R.typhi (R. mooseri) and Tick Typhus
R. prowazekii Rocky Mountain Spotted Fever
R.typhi and R. prowazekii are closely similar Rocky mountain spotted fever (RMSF) is a potent­
but may be differentiated by biological and ially life-threatening infection. R. rickettsii is the
immunologi­cal tests. etiologic agent of Rocky Mountain spotted fever.
i. Neill–Mooser or the tunica reaction
When male guinea pigs are inoculated Vector: Ixodid (hard) ticks are the vectors. It is
intra­peritoneally with blood from a case of transmitted by Dermacentor andersoni and related
endemic typhus or with a culture of R. typhi, species of ticks. The ecology and epidemiology of
they develop fever and a characteristic scrotal spot­ted fever are directly related to the life cycle of
inflammation. The scrotum be­comes enlarged four species of ixodid (hard) ticks vectors. Humans
and the testes cannot be pushed back into the are only accidentally infected.
abdomen because of inflammatory adhesions Clinical diseases: The incubation period is about
between the layers of the tunica vaginalis. This one week. Clinical picture is similar to that of
is known as the Neill–Mooser or the tunica typhus fever but the rash appears earlier and is
reaction. The Neill-Mooser reaction is negative more pronounced. It is prevalent in many parts of
with R. prowazekii. North and South America.
ii. Other methods used include-IFA, ELISA and
PCR ­based DNA tests. Other Spotted Fever Rickettsiae (Table 51.2)
R. siberica causes the Siberian tick typhus which
B. Spotted Fever Group is mild rickettsial disease. Boutonneuse fever is
They are all transmitted by ticks, except R. akari, caused by R. conorii. R. conori strains are isolated
which is mite-borne. Rickettsiae of this group from the Mediterranean littoral; Kenya, South
possess a common soluble antigen and multiply Africa and India are indistinguishable. Australian
in the nucleus as well as in the cytoplasm of host tick typhus is caused by R. australis. All these
cells. The basic pathologic process in the spotted three rickettsiae are maintained in nature in ixodid

Table 51.2  Human disease caused by Rickettsia and Orientia species


Group Species Diseases Vector Vertebrate Geographical
reservoir distribution
Typhus group R. prowazekii Epidemic typhus Louse Human beings Worldwide group
Brill–Zinsser disease Louse Human beings America, Europe
R. typhi Endemic typhus Rat flea Rat Worldwide
R. felis Endemic typhus Cat flea Opossum USA
Spotted fever group R. rickettsii Rocky mountain Tick Rabbit, dog N. America
spotted fever
R. siberica Siberian tick typhus Tick Wtld Animals cattle Russia, Mongolia
R. conorii Boutonneuse fever Tick Dog, rodents Mediterranean
S. African tick typhus Tick Dog, rodents S. Africa
Kenyan tick typhus Tick Rodents Kenya
Indian tick typhus Tick ? Rodents India
R. australis Queensland tick Tick Bush rodents N. Australia
typhus
R. japonica Oriental spotted Tick ? Japan
fever
R. akari Rickettsial pox Gamasid mite Mouse USA, Russia
Scrub typhus group: O. tsutsugamushi Scrub typhus Trombiculid mite Small rodents, East Asia, Pacific

Chap-51.indd 364 15-03-2016 11:20:55


Chapter 51: Rickett­siaceae, Bartonellaceae and Coxiella  | 365
ticks. Humans accidentally enter the natural cycle. in­fectious and have caused several serious and
Diseases produced by these rickettsiae resemble fatal in­fections among laboratory workers, their
RMSF but are of milder form. A black spot having a isolation should be attempted with utmost care
necrotic center (Eschar) frequently occurs at the site and only in lab­oratories equipped with appropriate
of the tick bite in all spotted fever group infections safety provisions. Rickettsiae can be isolated in
except Rocky Mountain spotted fever. The tick labora­tory animals such as mice or guinea-pigs,
Rhipicephalus sanguineus is the most important in embry­onated chicken eggs and in cell culture.
vector. Haemaphysalis leachi, Amblyomma and
Hyalomma ticks can also transmit the infection. i. Labora­tory animals
Rickettsiae may be isolated in male guinea pigs
Rickettsial pox or mice from patients in the early phase of the
disease. Blood clot ground in skimmed milk or
Rickettsial pox is a relatively mild infection transmitted
any suitable suspending medium is inoculated
by mites. Rickettsial pox is characterized by a local
intraperitoneally. The animals have to be observed
eschar, a papuloves­icular rash, and a benign clinical
for 3–4 weeks and their temperature recorded daily.
course. It is also called vesicular or varicelliform
rick­ettsiosis. 1. In Rocky Mountain spot­ fever (RMSF):
The causative agent is R. akari (from akari, Guinea-pigs develop fever, scrotal necrosis and
meaning mite). The reservoir of infection is the may even die due to overwhelm­ing infection of
domestic mouse, Mus musculus. The vector is R. rickettsii.
the mite, Liponyssoides sanguineus, in which 2. In R. typhi, R. conorii and R. akari infection:
transovarial transmission occurs. Guinea-pigs develop fever and tunica reaction.
3. In R. prowazekii infection: The animals deve­
C. Scrub Typhus Group lop fever without any testicular inflammation.
Genus Orientia [Scrub Typhus (Chigger- For isolation of O. tsutsugamushi, mice are
borne Typhus), Tsutsugamushi Disease] preferred over guinea-pigs. The infected animals
become ill and develop ascites.
Scrub typhus is caused by Orientia tsutsugamushi.
Smears from peritoneum, tunica and spleen
It was first observed in Japan where it was found to
of infected animals may be stained by Giemsa or
be transmit­ted by mites. The disease was therefore
Gimenez methods to demonstrate the rick­ettsiae.
called tsu­g amushi (from tsutsuga, meaning
dangerous, and mushi meaning insect or mite). It ii. Embry­onated chicken egg: Rickettsiae can also
occurs all along East Asia, from Korea to Indonesia, be grown in the yolk sac of chick embryo.
and in the Pacific Islands including Australia.
iii. Cell culture: Rickettsiae grow well in vero
The vectors are tromiculid mites belonging to
cell MRC 5 cell cultures and can be identified
the genus Lep­totrombidium. O. tsutsugamushi is
by immunofluroscence using group and strain
transmitted to man by the larval stages of mites of
specific monoclonal antibodies.
the genus Leptotrombidium- L. akamushi in Japan
and L. deliensis in India.
2. Direct Detection of the
Clinical diseases: The incubation period is 1–3 Organisms and Their Antigens
weeks. Patients typ­ically develop a characteristic
a. Detection of rickettsiae in tissue
eschar at the site of the mite bite, with regional
Skin biopsies from the centre of petechial lesions can
lymphadenopathy and mac­ulopapular rash. The
be examined for rickettsiae by immunofluorescence
disease sets in with fever, head­ache and conjunctival
or immuno-enzyme methods. Biopsy specimens
injection. Death may result from encephalitis,
may be stained with Giemsa, Macchiavello or
respira­tory failure and circulatory failure.
Gimenez stains and with direct and indirect
immuno­fluorescence techniques. In tissue smears,
Laboratory Diagnosis
rickett­siae are usually seen as bipolar rods occurring
Rickettsial diseases may be di­a gnosed in the near cells or free in the cytoplasm. R. rickettsii may
laboratory either also be seen within the nuclei of infected cells.
1. Isolation of rickettsiae.
2. Direct detection of the organisms and their b. Polymerase chain reaction (PCR): Detection of
anti­gens. rickettsial DNA by PCR is more rapid than isolation.
3. Serology.
3. Serology
1. Isolation of Rickettsiae Serological diagnosis may be by the heterophile
Isolation of the organism provides conclusive proof Weil-Felix reaction or by specific tests using rickett­
of rickettsial infection. As rickettsiae are highly sial antigens.

www.ebook3000.com
Chap-51.indd 365 15-03-2016 11:20:55
366  |  Section 3: Systemic Bacteriology
i. Weil–Felix (WF) reaction Prophylaxis
The Weil–Felix reaction is an agglutina­tion test
1. General measures: General measures, such
in which sera are tested for agglutinins to the
as control of vectors and animal reservoirs are
a-antigens of certain nonmotile Proteus strains OX
useful to prevent rickettsial diseases.
19, OX 2 and OX K.
2. Vaccination: Immunization is useful in special
Basis of the test: The basis of the test is the sharing situations. There is no safe, effective vaccine for
of an alkali-stable car­bohydrate antigen by some any of the rickettsial diseases.
rickettsiae and by certain strains of Proteus, P.
i. Weigl’s vaccine: In earlier days, phenolized
vulgaris OX 19, and OX 2 and P. mirabilis OX K.
intestinal contents of lice infected per
Procedure: The test may be performed as a micro- rectum with R. prowazekii (Weigl’s vaccine)
agglutination reac­tion in microtiter plates with was developed. It was too complicated for
round bottomed wells with hematoxylin-stained mass production.
antigen or as a tube agglutination test, though rapid
ii. A live vaccine: Containing attenuated E.
slide agglutination meth­ods have been employed
strain of R. prowazekii grown in yolk sac have
for screening.
been found to be effective but a proportion
of vaccines develop mild disease.
Interpretation
iii. Formalin inactivated R. rickettsii has
Sera from epidemic and endemic typhus aggluti­
been used but does not prevent the disease
nate OX 19 and sometimes OX 2 also. The test is
completely.
neg­ative or only weakly positive in Brill–Zinsser
disease. In tick-borne spotted fever both OX 19 and iv. Recombinant vaccines may be more
OX 2 are agglutinated. OX K agglutinins are found successful.
only in scrub typhus. (Table 51.3).
The Weil–Felix reaction is a simple and useful EHRLICHIA, ANAPLASMA AND
test for the diagnosis of some rickettsial diseases. NEORICKETTSIA
The antibody appears rapidly during the course of
the dis­ease, reaches peak titers of up to 1 : 1000 or Ehrlichia and Anaplasma are small, obligate
1 : 5000 by the second week and declines rapidly intracellular, gram-negative bacteria. They infect
during conva­lescence. False-positive reaction may circulating leukocytes, erythrocytes, and platelets,
occur in some cases of urinary or other infections where they multiply within phagocytic vacuoles,
by Proteus and at times in typhoid fever and liver forming clusters with inclusion-like appearance.
diseases. Hence it is desirable to demonstrate a rise These clusters of ehrlichiae resemble mulberries
in titer of antibodies for the diagnosis of rickettsial and are called morulae (Latin morum = mulberry).
infection.
Species
ii. Specific tests using rickettsial antigens
Serological methods using rickettsial antigens are The ehrlichiae that cause disease in humans have
specific, which include complement fixation test, been classified in a limited number of species
latex agglutination test and enzyme immunoassay. which are as follows: (i) Ehrlichia chaffeensis,
causes human monocyte ehrlichiosis (HME); (ii)
Ehrlichia ewingii causes E ewingii ehrlichiosis;
Treatment (iii) Anaplasma phagocytophilum causes human
Rickettsial infections may be treated with tetracyc­ granulocyte anaplasmosis (HGE) (Table 51.4). The
lines or chloramphenicol. Both these drugs are same genera contain additional species that infect
rick­ettsiostatic. animals but apparently not humans. The human
pathogens in the group have animal reservoirs and
Table 51.3  Weil–Felix reaction in rickettsial diseases can cause disease in animals as well.
Disease Agglutination pattern with
1. Ehrlichia chaffeensis
OX 19 OX2 OXK
It is tick-borne ehrlichiosis. Deer and rodents are
1. Epidemic typhus +++ + –
believed to be reservoir hosts. Ehrlichia chaffeensis
2. Brill–Zinsser disease usually or weak causes human monocyte ehrlichiosis (HME). E
negative positive
chaffeensis frequently causes severe or fatal illness.
3. Endemic typhus +++ +/- –
4. Tickborne spotted ++ ++ – 2. Ehrlichia ewingii
fever
E. ewingii has a geographic distribution similar to
5. Scrub typhus – – +++
E. chaffeensis. Ehrlichia ewingii, causes E. ewingii

Chap-51.indd 366 15-03-2016 11:20:55


Chapter 51: Rickett­siaceae, Bartonellaceae and Coxiella  | 367
Table 51.4  Ehrlichia, Anaplasmaa and Neorickettsia species responsible for human disease
Species Disesase Vector or Source
E. chaffeensis Human monocycte ehrlichiosis (HME) Amblyomma americnnum (Lone Star tick)
E. ewingii Ewingii ehrlichiosis Amblyomma americnum
Anaplasma. Human granulocyte ehrlichiosis Ixodes (including 1. scapularis,1. pacificus, 1.
phagocytophilum (HCG) ricinus)
Neorickettsia sennetu Sennestu fever Ingestion of raw fish infested flukes

ehrlichiosis; It is transmitted by ticks. Leukopenia year or more at 4°C and in meat at least for one
and thrombocytopenia are seen in patients. month. It is not completely inactivated at 60°C or
by 1% phenol in one hour. In milk it may survive
3. Anaplasma phagocytophilum pasteurization by the holding method, but the
It infects human granulocytic cells. Anaplasma flash method is effective. It can survive in dust
phagocytophilum, causes human granulocyte and aerosols, therefore, can be transmitted as an
anaplasmosis (HGE). The human pathogens in airborne infection. It can be inactivated by 2%
the group have animal reservoirs and can cause formaldehyde, 5% hydrogen peroxide and 1% lysol.
disease in animals as well
Antigenic Variation
4. Neorickettsia sennestu An important characteristic of Coxiella infections
It was previously called Ehrlichia sennestu, an is the ability to undergo antigenic variation in
intraleukocytic bacterium. It causes sennestu expres­sion of the cell wall LPS antigen. Cox. bumetii
ehrlichiosis, an illness resebling glandular fever shows phase variation. Fresh isolates are in Phase I.
(sennetsu being the Japanese name for glandular It becomes Phase II on repeated passage in yolk sac,
fever). No insect vector has yet been implicated in its but reverts to Phase I by passag­ing in guinea pigs.
transmission. It is associated with ingestion of raw fish
infested with infected flukes. Similar cases have been Pathogenesis
reported in Malaysia and Philippines. Coxiella burnetii causes Q fever which has a world­
wide distribution, as a zoonosis solidly established
Laboratory Diagnosis in domestic livestock.
1. Identification of characteristic morulae: Typical Cycles of infection: In nature there are two cycles
morulae in white blood cells is diagnostic in of infection of C. burnetii. One involves arthropods
Giemsa stained peripheral blood smear. (especially ticks) and a variety of vertebrates. The
2. Specific antibodies can be demonstrated by other cycle is maintained among domestic animals
indirect immunofluorescence methods. (cattle, sheep and goats).
3. Polymerase chain reaction (PCR) for ehrlichial
DNA. Reservoirs of the disease: The primary reservoirs
of the disease are wild and domestic ungulates,
including cattle, sheep, goats, rabbits, cats and dogs.
GENUS COXIELLA: Q FEVER It is transmitted among them and to cattle, sheep
The name Q fever (Q for “query”) was first used when and poultry by ixodid ticks.
Derrick (1935) was investigating an outbreak of Animal infection: Domestic animals have
typhoid-like fever in abattoir workers in Australia. Q inapparent infections but may shed large quantities
fever is caused by Coxiella burnetit. Coxiella burnetii, of infec­tious organisms in their urine, milk, feces,
is an obligately intracellular prokaryote. and especially, their placental products.

Morphology Human infection: Human infection may occur


occupationally through consumption of infected
Cox. burnetii is pleomorphic, occurring as small milk, handling of contaminated wool or hides,
rods 0.2–0.4 μm × 0.4–1.0 μm or as spheres 0.3–0.4 soil contaminated by infected animal feces,
μm in diameter. It is filterable. Generally regarded infected straw, and even to dusty clothing. Coxiella
as gram-negative. It is better stained with Giminez proliferate in the respiratory tract and then
and other rickettsiae stains. disseminate to other organs. Person-to-person
transmission is a rarity. Ticks do not seem to be
Resistance important in human infection.
C. burnetii may be the most infectious of all Incubation period is 2–4 weeks. Acute diseases
bacteria. In dried feces or wool it survives for a include influenza-like syndrome, atypical

www.ebook3000.com
Chap-51.indd 367 15-03-2016 11:20:56
368  |  Section 3: Systemic Bacteriology
pneumonia, hepatitis, pericarditis, myocarditis, Pathogenesis
meningo­encephalitis. Chronic diseases include Oroya fever: Bartonella bacilliformis is responsible
endocarditis, hepatitis, pul­monary disease, and for bartonellosis, an acute febrile illness consisting
infection of pregnant. of severe anemia (Oroya fever) fol­lowed by a
chronic cutaneous form (verruga). Oroya fever
Laboratory Diagnosis presents as fever and progressive anemia due to
1. Isolation: Isolation of C. burnetii from patient bacterial invasion of erythrocytes. Mortality is high
specimens is a specialized procedure and is in untreated cases.
not generally recommended because of the
Verruga peruana: Some of the survivors developed
extremely infectious nature of the organism.
nodular ulcerating skin lesions, called verruga
2. Serology: Using complement fixation test
peruana. It is a late sequel in survivors or in those
(CFT) or indirect immunofluorescence assay.
with asymptomatic infection.
Treatment: Tetracyclines are the drugs of choice
for acute infections. Rifampin combined with either Carrion’s disease: Oroya fever and verruga
doxycycline or trimetho­prim-sulfamethoxazole is peruana are also known as Carrion’s disease.
used to treat chronic infections. Etiology: It is spread by the sandfly vector Phlebo­
tomus.
Prophylaxis
Prevention of disease is by observing basic hygienic Laboratory Diagnosis
precautions while dealing with infected animals.
i. B. bacilliformis can be demonstrated in blood
Pasteurization of raw milk helps prevent milk-
smears stained by Giemsa. Organisms are
borne transmission. Vaccines have been developed
seen in the cytoplasm as well as adhering to
from formalin killed whole cells.
the cell surfaces.
BARTONELLA ii. Organisms may be isolated from the blood,
in semi-solid medium containing rabbit
Family Bartonellaceae contains two genera: serum and he­moglobin. Identification can be
Bartonella and Grahamella. achieved by PCR or by cultural and serological
Members of genus Bar­tonella are very small tests.
gram-negative bacilli. The genus Bartonella, which iii. Guinea pig inoculation leads to verruga
now consists of 11 species, including 5 that cause peruana but not Oroya fever.
human disease (Table 51.5). Members of the iv. Other tests
genus Grahamella preferentially grow within the a. Polymers chain reaction (PCR)
erythrocytes of vertebrates, but do not infect humans.
b. Serology: Indirect hemag­g lutination,
indirect immunofluorescence and ELISA.
1. Bartonella bacilliformis
It is a pleomorphic gram-negative rod, which is Treatment: B. bacilliformis is susceptible to
motile by a tuft of polar flagella. It can be cultivated in penicillin, strepto­mycin, tetracycline and chloram­
semisolid agar with rabbit or human blood. Growth phenicol.
is slow and becomes visible after 10 days incubation. Prophylaxis: Insecticides such as DDT should be
used to eliminate the sandfly inside and outside
Table 51.5  Bartonella species associated with the houses.
human infections
Species Diseases
2. Bartonella quintana
1. B. bacilliformis Oroya fever (bartonellosis, Carrion’s B. quintana is a small, gram-negative bacillus. It
disease) does not possess flagella, although it may exhibit
2. B. qintana Trench fever; cutaneous, subcutaneous,
twitching movement caused by fim­briae. It grows
and osseous manifestations of bacillary slowly on rabbit or sheep blood agar at 35°C in
angiomatosis; endocarditis 5% CO2 in air. Colonies appear after two weeks
3. B. henselae Cat-scratch disease; bacteremia; in primary culture and 3–5 days in sub­sequent
cutaneous, lymphatic, and passages.
hepatosplenic (peliosis hepatis)
manifestations of bacillary
angiomatosis; endocarditis
Pathogenesis
4. B. clarridgeiae Endocarditis, cat-scratch disease (rare)
Trench fever or five-day fever: B. quintana
causes trench fever or five-day fever occurred
5. B. elizabethae Endocarditis (rare)
among soldiers fighting in the trenches in Europe

Chap-51.indd 368 15-03-2016 11:20:56


Chapter 51: Rickett­siaceae, Bartonellaceae and Coxiella  | 369
during the First World War. The disease was not Afipia felis has also been implicated in a small
fatal but because of its slow course and prolonged proportion of cases of cat scratch disease.
convalescence, it caused very considerable loss of 2. Bacillary angioma­tosis: B. henselae is also
manpower. responsible for bacillary angiomatosis, but
Trench fever is an exclusively human disease primarily involving the skin, lymph nodes, or
and no animal reservoir is known. It is transmitted liver and spleen (peliosis hepatis).
by the body louse. Infec­tion is acquired when 3. Subacute bacterial endocarditis.
infected lice feces are scratched into the skin. The
lice are unharmed and remain infectious for life. Laboratory Diagnosis
Vertical transmission does not occur in lice.
1. It can be demonstrated in lymph node biopsy
Clinical diseases: Clinically, after an incubation smears and sections by Warthin–Starry staining
period of 14–30 days patient develops headache, show clusters of bacilli.
malaise, fever, chills, severe pain in the back and 2. B. henselae has been isolated from the blood
legs and a rose­olar rash on the chest, abdomen or of patients, in blood media after prolonged
back. Recov­ery is frequently followed by relapses incubation and is now considered as its
occurring at five day intervals (hence the species etiological agent.
name, quin­tana). The infection is mild, recovery is
slow and fatalities are rare. Treatment: Cat scratch dis­ease is usually self
More recently, B. quintana has been associ­ limiting disease and does not appear to respond
ated with bacillary angiomatosis, a vascular to antimicrobial therapy.
proliferative disor­der seen primarily in immuno­
compromised patients. Key Points
The disease frequently leads to a chronic or ™™ The family rickettsiaceae comprises two genera­-
Rickettsia and Orientia
latent infection. Relapses have been reported as
™™ Coxiella and Ehrlichia are now included in the family
long as 20 years after the primary disease. The coxiellaceae and family Anaplasmataceae
chronic infection and late relapses help to maintain ™™ Genus Rickettsia: Rickettsiae are gram-negative,
the bartonella in the absence of animal reservoirs. small, intracellular bacteria. Replication occurs in
cytoplasm of infected cells. They grow in various
tissue cultures and in yolk sac of embry­onated egg
Laboratory Diagnosis
™™ Pathogenesis: A. Typhus fever group: 1. Epidemic
i. B. quintana can be isolated by allowing typhus; 2. Brill–Zinsser disease; 3. Endemic ty­phus
healthy lice to feed upon the patient and ™™ Rickettsia prowazekii
the bartonellae may be detected in the gut ™™ Diseases: 1. Epidemic typhus-caused by R. prow­
azekii. The principal vector is the human body louse.
of these lice. It can also be cultivated from 2. Brill-zinsser disease (Recrudescent typhus)-caused
patient’s blood on rabbit or sheep blood agar. by R. prowazekii
ii. Polymerase chain reaction (PCR): B. 3. Endemic typhus is caused by R. mooseri (R. typhi).
quintana has been detected in the tissues by Vector is rat flea (Xenopsyella cheopis)
™™ R. mooseri can be differentiated from R. prowazekii
PCR.
by Neil-Mooser or tunica reaction
™™ R. rickettsii causes Rocky Mountain spotted fever
3. Bartonella henselae ™™ R. conori is responsible for boutonneuse fever and
transmitted by ixodid ticks
Bart. henselae is responsible for cat­scratch disease. ™™ Rickettsia akari causes rickettsial pox. Infection is
It is small, slightly curved gram-negative bacillus. transmitted by the bite of mites
It shows twitching motility. It can be grown on ™™ O tsutsugamushi causes scrub typhus. The disease
chocolate agar, Columbia agar with 5% blood is transmitted by trombiculid mite
and trypticase agar. The optimum temperature ™™ Laboratory diagnosis of rickettsial diseases may
for growth is 35–37°C in 5% CO2, Growth is slow be carried out by isolation of rickettsiae and serology.
Rickettsiae can be isolated in cell culture, in labora­
and takes 5–15 days. Colonies are white, dry, tory animals such as mice or guinea-pigs, or in
cauliflower-like and embedded in the agar. embry­onated chicken eggs
a. Detection of rickettsiae in tissue by immuno­
Pathogenesis fluorescence or immuno-enzyme methods and
polymerase chain reaction (PCR)
1. Cat scratch disease: The disease is acquired The Weil–Felix reaction is an agglutina­tion test
after exposure to cats (e.g. scratches, bites, and which detects anti-rickettsial antibodies in which sera
contact with cat fleas). Cat­scratch disease is a are tested for agglutinins to the antigens of certain
severe condition of regional lymphadenopa­thy nonmotile Proteus strains OX 19, OX 2 and OX K
b. Specific tests using rickettsial antigens: Include
and fever resulting from the scratch or bite of
complement fixation test, latex agglutination test
an infected cat. Bart. clarridgeiae, can cause and enzyme immunoassay
an identi­cal syndrome. An organism known as

www.ebook3000.com
Chap-51.indd 369 15-03-2016 11:20:56
370  |  Section 3: Systemic Bacteriology
3. Human body louse is responsible for transmission
Ehrlichia of which of the following diseases?
™™ These tick-borne bacteria cause three human a. Epidemic typhus b. Murine typhus
infections: E sennetsu causes a type of glandular fever; c. Rickettsial pox d. Q fever
E chaffeensis causes human monocytic ehrlichiosis,
4. Mites are responsible for transmission of diseases:
E phagocytophila causes human granulocytic
ehrlichiosis a. Epidemic typhus b. Murine typhus
c. Scrub typhus d. Trench fever
Coxiella burnetii
5. The rickettsial disease transmitted by ixodid ticks
™™ It is small, pleomorphic coccobacillary bacterium is:
with a gram-negative cell wall, intracellular bacteria
a. Rickettsial pox:
™™ Disease: Q fever is a worldwide zoonosis. Most
b. Rocky Mountain spotted fever
disease acquired through inhalation; possible
c. Trench fever
disease from consumption of contaminated milk
d. Scrub typhus
Bartonella 6. Weil-Felix reaction is positive in all the following
™™ The genus Bartonella contains B. bacilliformis, diseases except:
B. quintana and B. henselae which cause Oroya a. Epidemic typhus b. Endemic typhus
fever, trench fever and cat scratch disease in man
c. Brill–Zinsser’s disease d. Q fever
respectively.
7. Coxiella differs from rickettsial pathogens as it:
a. Has typical Gram-negative cell walls
IMPORTANT QUESTIONS b. Contains both DNA and RNA
c. Is susceptible to antibiotics
1. Write short notes on: d. Is transmitted by inhalation or ingestion
a. Typhus fevers 8. Human infections due to Coxiella can be acquired
b. Brill-zinsser disease (Recrudescent typhus) by:
c. Rocky mountain spotted fever a. Consumption of infected milk.
d. Scrub typhus b. Handling of infected wool or hides.
e. Trench fever c. Soil contaminated with feces of infected ani-
f. Coxiella burnettii or Q fever mals
g. Weil-Felix reaction d. All of the above
h. Cat scratch disease 9. Oroya fever is caused by:
i. Neil–Mooser reaction or Tunica reaction a. Capnocytophages ochracea
j. Ehrlichia b. Legionella pneumophilia
k. Oroya fever c. Streptobacillus moniliformis
2. Discuss laboratory diagnosis of rickettsial infections. d. Bartonella bacilliformis
10. The causative agent of cat scratch disease is:
a. Bartonella henselae
MULTIPLE CHOICE QUESTIONS (MCQs) b. Bartonella quintana
1. Which of the following bacteria does not require c. Coxiella burnettii
an arthropod vector for its transmission? d. Rickettsia prowazekii
a. Coxiella burnetii 11. All the statements are true for Bartonella quintana
b. Rickettsia akari except:
c. Bartonella quintana a. It is nonmotile
d. Rickettsia prowazekii b. It requires 1–2 weeks to produce demonstrable
2. The causative agent of epidemic typhus is: colonies on rabbit blood agar
a. Rickettsia prowazekii c. It is transmitted by hard ticks
b. Rickettsia rickettsii d. It is the causative agent of trench fever
c. Coxiella burnettii ANSWERS (MCQs)
d. Rickettsia akari 1. a; 2. a; 3. a; 4. c; 5. b; 6. d; 7. d; 8. d; 9. d; 10. a; 11. c

Chap-51.indd 370 15-03-2016 11:20:56


52
Chapter

Chlamydia and Chlamydophila

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Discuss morphology and the unique growth cycle
be able to: of Chlamydia, describing elementary body (EB) and
∙∙ Describe differences between chlamydiae and viruses reticulate body (RB) bodies
∙∙ Describe serotypes of various chlamydiae and ∙∙ Discuss laboratory diagnosis of chlamydial infections
diseases produced by them ∙∙ Describe the following: TRIC agents, inclusion
∙∙ Associate each of the major diseases with the three conjunctivitis lymphogranuloma venereum (LGV),
most important human species of Chlamydia and Frei’s test.
Chlamydophila

INTRODUCTION CLASSIFICATION
Chlamydiaceae is a family of obligate intracellular Genus Chlamydia is in the order Chlamydiales
bacterial parasites, small, nonmotile and gram- and the family Chlamydiaceae. The proposed new
negative with a tropism for columnar epithelial taxonomic classification for the family Chlamy­
cells lining the mucous membranes. diaceae consists of two genera: (1) Chlamydia to
Based on the human diseases they were then include C. trachomatis and (2) Chlamydophila to
known to cause, they were called psittacosis- include C. pneumoniae, C. psittaci, and C. pecorum.
lymphogranuloma-trachoma (PLT) viruses, or Other species have been placed into the two genera,
noncommittally as ‘PLT agents’ or TRIC (trachoma- but they are uncommon human pathogens and are
inclusion conjunctivitis) organisms. not discussed in this chapter (Table 52.1).

Chlamydia Species
Differences between Chlamydiae and Chlamydia trachomatis: C. trachomatis is divi­
Viruses ded into two biovars—those causing trachoma,
The Chlamydiaceae were once considered viruses. inclusion conjunctivitis (the so-called TRIC agents)
However, they differ from viruses in many respects and oculogenital infection.
and the organisms have the following properties
Chlamydophila Species
of bacteria:
1. Chlamydophila psittaci: It may lead to severe
1. They possess inner and outer membranes and sometimes fatal pneumonia in man (psittacosis
similar to those of gram-negative bacteria. or ornithosis).
2. They contain both DNA and RNA.
Table 52.1  Revised classification of the family
3. They possess prokaryotic ribosomes.
Chlamydiaceae
4. They synthesize their own proteins, nucleic
acids, and lipids. Genus Chlamydia Genus Chlamydophila
5. They multiply by binary fission. C. trachomatis C. pneumoniae
C. muridarum C. psittaci
6. They do not have ‘eclipse phase’ following C. suis C. pecorum
cellular infection. C. abortus
7. They are susceptible to numerous antibacterial C. caviae
antibiotics. C. felis

www.ebook3000.com
Chap-52.indd 371 15-03-2016 11:21:32
372  |  Section 3: Systemic Bacteriology
2. Chlamydophila pneumoniae: It is a common Reticulate body: The reticulate body is the intra­
cause of acute respiratory disease worldwide (Table cellular growing and replicative form, 500–1000 nm
52.2). in size (larger than the EB).
The chlamydiae are nonmotile, lacking flagella,
Morphology and non­piliated.

Chlamydiae are small, nonmotile bacteria. Although Growth Cycle


they stain poorly with gram’s stain they have the The Chlamydia growth cycle is initiated by the
typical LPS of gram-negative bacteria. There are attachment of an infectious elementary body to
two morphologically distinct forms of chlamydiae: the surface of a susceptible epithelial cell, followed
elementary body (EB) and reticulate body (RB). by its endocytosis (Fig. 52.1). Inside the host cell,
There are two morphologically distinct forms of the elementary body lies within the endosome,
chlamydiae: elementary body (EB) and reticulate being separated from the host cell cytoplasm by
body (RB). the endosomal membrane throughout its active
growth cycle.
Elementary body: It is a spherical particle, 200–300 By about eight hours, the elementary body
nm in diameter, with a rigid trilaminar cell wall. within the endosome undergoes spheroplast-
like transformation to the large reticulate body,
Table 52.2  Human diseases caused by Chlamydiae which be­gins to divide by binary fission and are
and Chlamydophila converted to elementary bodies.The developing
Species Serotype* Disease
chlamydial microcolony within the host cell is
called the inclusion body. The mature inclusion
C. trachomatis A, B, Ba, C Endemic blinding
body contains 100–500 elementary bodies which
trachoma
are ultimately released from the host cell. The
C. trachomatis D, E, F, G, H, I, J, Inclusion
host cell is severely damaged and release of the
K (D to K) conjunctivitis
(neonatal and adult) elementary bodies occurs within 48 hours by host
Genital chlamydiasis cell lysis in C. psittaci in­fections (Fig. 52.1).
Infant pneumonia
Laboratory Propagation
C. trachomatis L1, L2, L2a, L3 Lymphogranuloma
venereum Chlamydiae can be propagated in the mouse,
C. psittaci Many Psittacosis
chick embryo or in cell culture though they
serotypes show in­dividual variations in susceptibility. The
presence of chlamydial inclusions is determined by
Chlamydophila Only one Acute respiratory
pneumoniae serotype disease microscopy in conjunction with a suitable staining
*Predominant types associated with the disease
method, prefer­ably fluorescence microscopy with
labeled monoclonal antibody.

Fig. 52.1: The Chlamydia growth cycle

Chap-52.indd 372 15-03-2016 11:21:33


Chapter 52: Chlamydia and Chlamydophila  | 373
Antigenic Structure 1. Ocular Infections
Chlamydiae possess three major groups of antigens: i. Trachoma
genus-specific antigens, species-specific anti­gens, Trachoma is a chronic kerato­conjunctivitis. It is
and serotype-specific antigens. characterized by follicular hypertrophy, papillary
1. Genus-specific antigens: The heat-stable genus- hyperplasia, pannus formation and in the late
specific antigen common to all Chlamydia is LPS, stages, cicatrisation. caused by C. trachomatis
with an acidic polysaccharide as the antigenic serotypes A, B, or C. Trachoma is transmitted eye-
determinant. The antigen, can be detected by the to-eye by droplet, hands, contaminated clothing,
complement fixation test, SDS polyacrylamide and eye-seeking flies. The pathogen may also be
gel electrophoresis, and by polyclonal or epitope transmitted by respiratory droplet or through fecal
specific monoclonal antibodies. contamination. Trachoma generally is endemic
2. Species-specific antigens: Several species- in communities where the living conditions are
specific antigens have been detected on or near crowded, sani­tation is poor, and the personal
the envelope surface.They help in classifying hygiene of the people is poor.
chlamy­diae into the species—trachomatis,
Laboratory Diagnosis
psittaci, pneumoniae and pecorum.
3. Serotype-specific antigens: Serotype-specific 1. Direct Cytopathologic Examination
determinants are common only to cer­t ain The characteristic inclusion (Halberstaedter-
isolates within a species and help in intraspecies Prowazek or HP bodies) may be dem­onstrated in
typing. They are located on the major outer conjunctival scrapings, after staining by Giemsa,
membrane proteins (MOMP) and can be Castaneda or Macchiavello methods. They may
demon­strated by microimmunofluorescence. By be stained with iodine solution also because they
micro-IF, chlamydiae have been classified into possess a glycogen matrix. The fluorescent antibody
many serological variants (serovars, serotypes). method enhances the sensitivity of smear diagnosis.

C. trachomatis: Chlamydia trachomatis has been 2. Culture


divided into three biovars (biological variants) i. Embryonated egg: The chlamydia may be
which cause trachoma, inclusion conjunctivitis grown in the yolk sac of 6–8 days old eggs.
(the so-called TRIC agents) and lymphogranuloma ii. Cell culture: Tissue culture using stationary
venereum (LGV), and mouse pneumonitis phase cells (nonreplicating cells) is the method
(renamed C. muridarum). Both the trachoma and of choice for isolation.The bacteria infect a
the LGV biovars can be divided into serotypes restricted range of cell lines in vitro (e.g. HeLa-
(serovars) on the basis of epitopes carried on their 229, McCoy BHK-21, Buffalo green monkey
major outer membrane protein (MOMP). kidney cells), similar to the narrow range of
Trachoma biovar: There are 15 serovars in the cells they infect in vivo. McCoy cells rendered
trachoma biovar (with variants within them) which nonreplicating by irradiation or antimetabolites
are given the letters serovars A through K (A, B, Ba, are used. HeLa or HL cells treated with DEAE
C, D, Da, E, F, G, Ga, H, I, la, J, and K). Serovars A, dextran may also be used. The inoculum has to
B, Ba and C cause blinding trachoma in endemic be driven into the cells by centrifugation up to
areas, and serovars D to K associated with the less 15,000 g to get a good growth.
serious ocular in­fection, inclusion conjunctivitis Treatment and control: Local application and oral
and with various genital infections. administration of erythromycin and tetracycline or
other suitable an­tibiotics should be continued for
LGV biovar: Four serovars L1, L2 and L3, in the several weeks. Single-dose azitromycin treatment
LGV biovar. Serovars L1’ L2, L2a’ and L3 are associated has been used with good results.
with LGV, an invasive urogenital tract disease and Vaccines that are efficacious and safe are not
hemorrhagic proctitis. available Basic. however, to the ultimate control
C. psittaci: The serological classification of C. psittaci of trachoma, are good standards of hygiene that
is complex, many serotypes having been identified. accompany improvement in standards of living.

C. pneumoniae: C. pneumoniae has not been ii. Inclusion conjunctivitis


subclassified yet (Table 52.2). The natural habitat of C. trahomatis types D to K is
the genital tract in both sexes.
a. Inclusion blennorrhea: Inclusion blennorrhea,
Chlamydia trachomatis
is the neonatal form of inclusion conjunctivitis.
C. trachomatis is a leading cause of ocular and The disease in the newborn usually becomes
genital infections worldwide. clinically apparent 5–12 days after birth. The

www.ebook3000.com
Chap-52.indd 373 15-03-2016 11:21:33
374  |  Section 3: Systemic Bacteriology
organisms are acquired from the mother during sole natural hosts of this infection caused by the
birth. The disease can be prevented by local LG V biovar of C. trachomatis, Ll, L2 and L3 most
application of antibiotics. commonly L2. (Table 52.2).
b. Inclusion conjunctivitis: Inclusion con­
junctivitis (paratrachoma) is most prevalent in Clinical Manifestations
sexually active young people, being spread from The usual incubation period of LGV is 1–4 weeks.
genitalia to the eye and occurs worldwide. It
1. Primary lesion: The primary lesion is
was known as ‘swimming pool conjunctivitis’
painless, small, inconspicuous, and vesicular
as infection was associated with bathing in
and often escapes notice. Characteristically the
community swimming pools which presumably
presenting complaint concerns the enlarged
get contaminated with chlamydia from the
matted inguinal and femoral lymph nodes
genital secretions of bathers. Contamination of
which are moderately painful and firm and may
the eye with the patient’s own genital secretion
become fluctuant.
may be the cause more often. The disease is
much milder, is usually self-limiting and rarely 2. Secondary stage: The secondary stage results
causes visual loss, presumably because, unlike from lym­phatic spread to the draining lymph
trachoma, repeated infection is less common. nodes. In men the inguinal lymph nodes
are involved most often and in women the
2. Genital Infections intrapelvic and pararectal nodes. Women and
homosexual men may develop hemorrhagic
C. trachomatis infections of the genital tract are of proctitis with regional lymphadenitis. The
two types and are sexually transmitted: nodes enlarge, suppurate, become adherent
1. Those caused by the oculogenital serotypes to the skin and breakdown to form sinuses
D through K collectively referred to as ‘genital discharging pus. Metastatic complications may
chlamydiasis’ sometimes occur, with involvement of joints,
2. LGV caused by serotypes L1, L2, L2a and L3. eyes, and meninges.
3. Tertiary stage: The tertiary stage is chronic,
I. Genital chlamydiasis lasting for several years, representing the
Ch. trachomatis D to K are called genital chlamy­ sequelae of scarring and lymphatic blockage.
diasis. Rectal stricture and rectal perforation are
recognized late sequelae to LGV proctitis. The
i. Infection in men: In men, they cause urethritis
course of the disease is variable. Late sequelae
(nongonococcal urethritis), epididymitis,
are more distressing in women and lymphatic
proctitis, conjunctivitis and Reiter’s syndrome.
obstruction in women can lead to elephantiasis
Reiter’s syndrome is a triad of recurrent
of the vulva, called esthiomene.
conjunctivitis, polyarthritis and urethritis
or cervicitis. Anal intercourse may cause
chlamydial proctitis in either sex. Associated Laboratory Diagnosis
symptoms include rectal pain and bleeding, The primary lesion usually goes unnoticed and
mucopurulent discharge and diarrhea. the disease is seen commonly first in the stage of
ii. Infection in women: Most genital tract inguinal adenitis (bubo).
infections in women are asymptomatic (as
many as 80%) but can nevertheless become A. Microscopy
symptomatic. The clinical manifestations
Smears of material aspirated from the bubos
include acute urethral syndrome, bartholinitis,
may show the elementary bodies (Miyagawa’s
mucopurulent cervicitis, endometritis,
granulocorpuscles). The sensitivity of microscopic
salpingitis, pelvic inflamma­tory disease (PID),
diagnosis is very low.
conjunctivitis, perihepatitis (Fitz–Hugh–
Curtis syndrome) and Reiter’s syndrome.
Genital chlamydiasis may cause infertility, B. Culture
ectopic pregnangy, premature deliveries, Isolation of the Chlamydia by intracerebral
perinatal morbidity and postpartum fever. inocula­tion into mice and into yolk sac of eggs has
been replaced by cell cultures.
II. Lymphogranuloma Venereum
Lymphogranuloma inguinale, climatic bubo, C. Serology
tropical bubo, and esthiomene are synonyms of LGV patients develop high titers of circulating
this sexually transmissible disease. It characterised antibodies, with titers of 1:64 or more in CF test
by suppurative inguinal adenitis. Humans are the and 1:512 or more in micro-IF.

Chap-52.indd 374 15-03-2016 11:21:33


Chapter 52: Chlamydia and Chlamydophila  | 375
D. Intradermal Test (Frei’s Test) Psittacosis can be prevented only through the
An intradermal test (Frei’s test) originally described control of infections in domestic and imported
by Frei in 1925 was commonly used formerly. The pet birds.
crude chlamydial antigen originally obtained
from the bubo pus, and later from mouse brain
Chlamydophila pneumoniae
or yolk sac cul­tures (lygranum), was inoculated Chlamydophila pneumoniae was formerly known
intradermally in the forearm, with a control on as Chlamydia sp., strain TWAR, and it was originally
the other arm. A positive reaction is indicated by identified in 1965 from a conjunctival culture of a
indura­tion of 7 mm or more in 2–5 days. This test child (TW) enrolled in a Taiwan trachoma vaccine
beco­mes positive 2–6 weeks after infection and study. In 1983 at the University of Washington, a
remains positive for several years. similar organism was isolated in HeLa cells from
Frei’s test is now not in use due to the frequent a pharyngeal specimen of a college student (AR).
occurrence of false positive reactions. At present, Grayston and colleagues (1986) isolated a chlamydial
the test has few diagnostic indications. strain from acute respiratory disease in adults in Tai­
wan and designated it as C. psittaci strain TWAR
Treatment {from Taiwan Acute Respiratory). It possessed the
Treatment is with tetracycline, which should be group-specific antigen in common with. C. psittaci
giv­en for at least three weeks. and C. trachomatis but could be distinguished from
both of them by species-specific antigens, DNA
Chlamydophila psittaci hy­bridisation and restriction endonuclease analysis.
Chlamydophila psittaci is the cause of psittacosis This new organism was initially called TWAR, then
(psittacos means parrot) among psittacine birds, classified as Chlamydia pneumoniae, and finally
also known as ornithosis (derived from the Greek placed in the new genus Chlamydophila. Infection
word ornithos for “bird”) or parrot fever. Human is transmitted by respiratory secretions; no animal
infections are mostly occupational. The respiratory reservoir has been identified.
tract is the main portal of entry, and infection
usually is acquired by inhalation of organisms from Spectrum of Disease
infected birds and their droppings. The incubation 1. C. pneumoniae has been associated with
period is about 10 days. The illness ranges from an pneumonia, bronchitis, pharyngitis, sinusitis,
‘influenza-like’ syndrome, to a severe illness with and a flu-like illness.
de­lirium and pneumonia. Psittacosis is a septicemia 2. It has been implicated in other chronic afflictions
and there may be meningoencephalitis, arthritis, such as asthma and cardiovascular diseases.
pericarditis or myocarditis, or a predominantly 3. It has also been linked to chronic illnesses such
typhoidal state with enlarged liver and spleen. as atherosclerosis, coronary heart disease, and
stroke.
Laboratory Diagnosis 4. Infection with C. pneumoniae has been established
A. Culture as a risk factor for Guillain-Barré syndrome
Chlamydia can be isolat­ed from blood during the CGBS) and also appears to be a relationship
early stages of the disease and from sputum later between sarcoidosis and C. pneumoniae
on. Infected cells, including alveolar macrophages
from patients, and mouse brain, yolk sac and cell Laboratory diagnosis
cultures show inclusion bod­ies (Levinthal-Cole- Diagnosis of C. pneumoniae infections is difficult.
Lillie or LCL bodies).
1. Cultivation
B. Antigen Detection C. pneumoniae will grow in the HEp-2 cell line.
Antigen detection is done by direct fluorescent C. pneumoniae species-specific monoclonal
antibody staining or by immunoassay or molecular antibodies can detect the organism in cell culture.
diagnosis by polymerase chain reaction.
2. Serology
C. Serology Complement fixation (CF) or microimmuno­
A four-fold increase in titer, shown by the group fluorescence (MIF) tests can be used to make a
specific CF testing of paired acute and convalescent serologic diagnosis. More recently, some partially
phase sera, is suggestive of C. psittaci infection, but automated enzymelinked immunosorbent assays
the species-specific MIF (micro-IF) test must be (ELISAs) have become commercially available.
performed to confirm the diagnosis.
3. Direct Detection Methods
Treatment and Prevention Detection of C. pneumoniae by nucleic acid
Infections can be treated successfully with tetracy­ amplification techniques has been successful.
clines or macrolides. Several C. pneumoniae-specific primers have been

www.ebook3000.com
Chap-52.indd 375 15-03-2016 11:21:33
376  |  Section 3: Systemic Bacteriology
used in PCR assays to detect organisms and appear characteristic inclusions can be found in yolk sac
to be more sensitive than cell culture. material from infected 6–8 day embryos. However,
this procedure is impractical for use by clinical
Treatment laboratories since it is tedious, time-consuming,
Macrolides (erythromycin, azithromycin, clarithro­ and less sensitive than tissue culture.
mycin), tetracyclines (tetracycline, doxycycline), or b. Animal inoculation
lev­ofloxacin administered for 10–14 days has been Chlamydiae can be propagated in experimental
used to treat infections. animals such as mice. C. psittaci strains infect
Control of exposure to C. pneumoniae is likely mice by intracere­bral, intranasal, intraperitoneal
to be difficult because the bacterium is ubiquitous. and subcutaneous routes. Among C. trachomatis
strains, only the LGV serovars (Ll, L2, L3) infect
Laboratory Diagnosis of Chlamydia mice when injected intracerebrally, but other C.
Infections trachomalis strains (The TRIC serovars) will not
The laboratory diagnosis of Chlamydia infections infect mice by any route of infection.
can be ac­c omplished by various approaches: c. Tissue culture
(1) microscopic demonstration of inclusion or The most widely used method of cultivation is by
elementary bodies, (2) isolation of the organisms, the use of tissue cultures. Monkey kidney cell lines,
(3) demonstration of chlamydial antigen and McCoy and HeLa cell lines and, more recently, the
(4) detection of specific antibodies (5) Skin test- heteroploid HL cell line are commonly used.
Hypersensitivity against these bacteria.
Cell cultures used for isolation are pretreated
The specimens collected are conjunctival by irradiation or chemicals such as 5-iodo-2-
scrapings, sputum, throat swab, bubo pus, genital deoxyuridine, cytochalasin B, or cycloheximide to
swabs and blood. enhance chlamydial replication and to allow easier
1. Demonstration of Inclusion or Elementary recognition of inclusions. Pretreatment of the cells
with diethylaminoethyl­dextran (DEAE-dextran)
Bodies
or centrifugation of the chlamydiae onto the host
i. Light microscopy: Smears may be prepared cells increases contact between the infectious
from conjunctival scrapings or bubo pus and chlamydial particles and the host cell monolayer,
stained by Giemsa, Castaneda, Machiavello or with a subsequent increase in infectivity.
Giminez stains to demonstrate characteristic
LGV strains grow well, while TRIC strains are
inclusion bodies under light microscope.
less in­fective. C. psittaci can be isolated from
Chlamydia are gram-negative but are stained
infected material by methods similar to those used
better by, Castaneda, Machiavello or Giminea
for other Chlamydia.
stains. Chlamydial elementary bodies and
Propagation of C. pneumoniae in cell culture
inclusions are large enough to be seen under
has proven to be difficult. HeLa 229 cells and, more
the light microscope.
recently, the heteroploid HL cell line have been
Glycogen-containing inclusions of C. tracho­
described as being more sensitive than McCoy cells
matis can be stained with Lugol’s iodine. The
for the isolation of this organism.
inclusion bodies of trachoma and inclusion
conjunctivitis are named Halberstaedter 3. Demonstration of Chlamydial Antigen
Prowazek or HP bodies whereas the elementary a. Immunofluorescence (IF): For diagnosis by
bodies of C. psittaci are called Levinthal Cole demonstration of chlamydial antigens, the
Lillie or LCL bodies. method commonly used is micro-IF. This test
ii. Immunofluorescence (IF): Immunofluore­ approaches cultures in sensitivity.
scence (IF) can iden­tify not only inclusions b. ELISA : The ELISA method is preferred for
but also extracellular elementary bodies. screening as it enables rapid testing for LPS
Besides ocular infections, IF is useful also in antigen in large numbers of specimens.
examination of cervical or urethral spec­imens, c. Molecular methods: Molecular methods like
which may contain elementary bodies. DNA probes and amplification techniques
(polymerase chain reaction, ligase chain
2. Isolation of the Organisms reaction) have greatly increased the sensitivity
Traditionally the isolation of Chla­mydia has been and specificity of antigen detection.
accomplished by the inoculation of infected material
into either embryonated eggs, experimental 4. Detection of Specific Antibodies or
animals, or selected tissue culture cell lines. Hypersensitivity Response
a. Yolk sac inoculation The diagnosis of chlamydial infections can
All known strains of Chlamydia will infect the be accomplished by either the group-specific
chick embryo, and group-specific antigen and complement-fixation (CF) test or type-specific

Chap-52.indd 376 15-03-2016 11:21:33


Chapter 52: Chlamydia and Chlamydophila  | 377
microimmunofluorescence (micro-IF) technique.
of organism and serology for antibody detection.
Like the CF test, EIAs are genus-specific, and Frei’s test is a skin test previously employed for
measurements of IgG and IgM can be accomplished. diagnosis of LGV.
i. Complement-fixation (CF) test: The CF test
uses the genus-specific antigen (LPS) and is
applicable for the diagnosis of infections caused IMPORTANT QUESTIONS
by C. psittaci, C. pneumoniae, and the LGV 1. Discuss laboratory diagnosis of chlamydial infec­
strains of C. trachomtis. In the majority of patients tions.
for whom the identity of the infecting serovar of 2. Discuss the pathogenicity of chlamydiae.
C. trachomatis is known, there is a type-specific 3. Write short notes on:
antibody response with a titer of 1:8 or greater. a. Developmental cycle of chlamydiae
ii. Microimmunofluoroscence (Micro-IF): b. Antigenic structure of chlamydiae
Micro-IF can test IgG and IgM antibody separately. c. TRIC agents
Type-specific antibodies are demonstrated by this d. Inclusion conjunctivitis
method. e. Frei’s test
iii. ELISA test: Measurements of IgG and IgM f. Lymphogranuloma venereum
can be accomplished like the CF test. They are g. Chlamydia pneumonia.
comparable in sensitivity to the micro-IF test
but may have limited specificity. The assay has MULTIPLE CHOICE QUESTIONS (MCQs)
been used successfully to detect chlamydial 1. Chlamydia organisms are not viruses because of
IgM in infants with pneumonia. all the following properties except:
a. They contain both DNA and RNA
5. Skin Test b. They contain prokaryotic ribosomes
The Frei test is an intradermal skin test that detects c. They can grow on cell-free media
d. They are susceptible to a wide range of anti­
a delayed hypersensitivity response to chlamydial
biotics
antigen was widely used formerly for diagnosis of 2. Trachoma is caused by C. trachomatis serovars:
LGV but has been given up because false-positive a. A. A, B, Ba, C b. D-K
results are very frequent. c. Ll-L3 d. All the serovars
Key Points 3. Man acquires psittacosis from:
a. Parrots b. Ducks
™™ Chlamydiae are obligate intracellular parasites which c. Pigeons d. All of the above
are small, nonmotile and gram-negative 4. Tests which can aid in the laboratory diagnosis of
™™ The family Chlamydiaceae has been divided into chlamydial infections is/are:
two genera, Chlamydia and Chlamydophila: a. Light microscopy
(1) Chlamydia to include C. trachomatis and (2) b. Immunofluorescence test
Chlamydophila to include C. pneumoniae, C. psittaci,
c. ELISA for detection of antigen
and C. pecorum
d. All of the above
™™ Chlamydia trachomatis infections: Two human
biovars: trachoma (with 15 serovars) and lympho­ 5. Which of the following serovars of Chlamydia are
granuloma venereum (LGV; 4 serovars) responsible for inclusion conjunctivitis?
™™ Diseases: C. trachomatis is the most common a. A, B, Ba, C b. D-K
sexually transmitted bacterial pathogen. Trachoma c. Ll-L3 d. None of the above
biovar responsible for ocular trachoma, adult 6. All the following statements are true for chlamy­
inclusion conjunctivitis, neonatal conjunctivitis, infant dophila pneumoniae except:
pneumonia, and urogenital infections. LGV biovar a. It is an important cause of respiratory disease in
responsible for lymphogranuloma venereum (LGV) olderchildren and adults
™™ Diagnosis: Culture is highly specific but is relatively b. Infection is transmitted by respiratory secretions
insensitive. Antigen tests (DFA, ELISA) are relatively from animal reservoirs
insensitive. The molecular amplification tests are the c.  Direct fluorescent antibody test is used to
most sensitive and specific tests currently available detect C. pneumoniaea antigen in specimens
™™ C. psittaci: C. psittaci is the cause of psittacosis, also d.  Tetracycline, doxycycline, erythromycin, etc.
known as parrot fever or ornithosis are effective against C. pneumoniae infection
™™ Diagnosis is often made by serologic assays
7. Frei’s test is an intradermal skin test used for the
™™ C. pneumoniae: C. pneumoniae, recognized as
diagnosis of:
a significant community acquired respiratory
pathogen, has been implicated in other chronic a. Lymphogranuloma venereum
afflictions such as asthma and cardiovascular dis­ b. Adult inclusion conjunctivitis
eases. It has also been linked to chronic illnesses, such c. Neonatal conjunctivitis
as atherosclerosis, coronary heart disease, and stroke d. Infant pneumonia
™™ Laboratory diagnosis of chlamydial infections
ANSWERS (MCQs)
depends on direct detection of antigens, isolation
1. c; 2. a; 3. a; 4. d; 5. b; 6. b; 7. a

www.ebook3000.com
Chap-52.indd 377 15-03-2016 11:21:33
Section 4: Virology

5353
Chapter

General Properties of Viruses

Learning Objectives

After reading and studying this chapter, you should ∙∙ List various cell cultures
be able to: ∙∙ Discuss detection of virus growth in cell cultures
∙∙ Describe size, shape and symmetry of viruses ∙∙ List DNA and RNA viruses
∙∙ Describe cultivation of viruses ∙∙ Describe prions and viroids.

Introduction Morphology of viruses


Viruses are the smallest known infective agents Size
and are perhaps the simplest form of life known. Viruses are much smaller than bac­t eria. The
Viruses do not possess a cellular organization extracellular infectious virus particle is called the
and they do not fall strictly into the category of virion. It was their small size and ‘filterability’
unicellular micro­organisms. (ability to pass through filters that can hold back
bacteria) that led to their recognition as a separate
Main properties of viruses class of infectious agents. Hence they were for
1. Viruses do not have a cellular organization a time known as ‘filterable viruses.’ They were
2. They contain only one type of nucleic acid, called ‘ultramicro­scopic’ as they were too small
either DNA or RNA but never both to be seen under the light microscope. Some of
the larger viruses, such as poxvirus­es can be seen
3. They are obligate intracellular parasites
under the light microscope when suitably stained.
4. They lack the enzymes necessary for protein The virus particles seen in this manner are known
and nucleic acid synthesis and are dependent as ‘elementary bodies’.
for replication on the synthetic machinery of The unit for measurement of virion size is
host cells nanometers (nm). Viruses vary widely in size from
5. They multiply by a complex process and not by 20 nm to 300 nm. The largest among them is pox
binary fission virus (300 nm) and is as large as the smallest bacteria
6. They are unaffected by antibacterial antibiotics. (myc­oplasma). The smallest virus is the parvovirus
The major differences between viruses and (about 20 nm) and are nearly as small as the largest
microorganisms are shown in Table 53.1. protein molecules such as hemocyanin.

Table 53.1  Properties of prokaryotes and viruses—Ananthnarayan


Cellular Growth on Binary Both DNA Ribosome s Sensitivity to Sensitivity
organization inanimate fission and RNA antibacterial to interferon
media antibiotics
Bacteria + + + + + + –
Mycoplasmas + + + + + + –
Rickettsiae + + + + + + –
Chlamydiae + - + + + + +
Viruses – – – – – +

Chap-53.indd 378 15-03-2016 11:33:18 AM


Chapter 53: General Properties of Viruses  | 379
Measuring the Size of Viruses Functions of Capsid
A. Passage them through Collodion membrane. i. Protection: It protects the viral genome
B. Electron microscopy. from physical des­truction and enzymatic
C. Sedimentation in the ultracentrifuge. inactivation by nucleases in biological material.
D. Comparative measurements: It can be done ii. Binding sites
with reference to: Staphylococcus, bacterial iii. It facilitates the assembly and packaging of
viruses (bacteriophages) and representative viral genetic information.
protein molecules. iv. Vehicle of transmission: From one host to
another.
Shape of the Virus v. Antigenic
The overall shape of the virus particle varies in vi. Host’s defence
different groups of viruses. Most of the animal vii. It provides the structural symmetry to the
viruses are roughly spherical, some are irregular virus particle.
and pleo­morphic. Poxviruses are brick-shaped,
rabies virus is bullet-shaped, tobacco mosaic virus B. Virus Symmetry
is rod-shaped. Bacteriophages have a complex Viral architecture can be grouped into three
morphology. The extracellular infectious virus types based on the arrangement of morphologic
particle is known as virion. sub­units: (1) Icosahedral symmetry (2) helical
symmetry (3) complex struc­tures.
Structure and chemical
1. Icosahedral Symmetry
composition of the viruses
An icosahedral ( icosa, meaning 20 in Greek) is a
A. Viral Capsid polygon with 12 vertices or corners and 20 facets
Viruses consist of nucleic acid core surrounded by or sides. Each facet is in the shape of an equilateral
a protein coat called capsid. The capsid with the trian­g le. There are always 12 pen­tons but the
enclosed nucleic acid is known as nucleocapsid. number of hexons varies with the virus group, e.g.
The capsid is composed of a large number of adenoviruses (Fig. 53.2).
capsomers, which form its morphological units.
The chemical units of the capsid are polypeptide 2. Helical Symmetry
molecules which are arranged symmetrically to The nucleic acid and the capsomers are wound
form molecules to form an impenetrable shell together in the form of a helix or spiral.
around the nucleic acid core (Fig. 53.1).
Examples: Single-stranded RNA viruses such as
influenza (Fig. 53.2), the parainfluenza viruses,
and rabies.

3. Complex Symmetry
Viruses (e.g. poxviru­ses) which do not show either
icosahedral or helical symmetry due to complexity
of their structure are referred to have complex
symmetry.
A B
C. Viral Envelope
Virions may be enveloped or nonenveloped
(naked). Enveloped Virus: The envelope or
outer covering of virus containing lipid is derived
from the plasma membrane of the host cell during
their release by budding from the cell surface. The
envelope is glycoprotein in nature. The lipid is largely
C D of host cell origin while the protein is virus-encoded.
Fig. 53.1: Schematic diagram illustrating the components of Peplomers: In mature virus particle, the glyco­
the complete virus particle (the virion). (A) Naked icosahedral proteins often appear as projecting spikes on the
virus, consisting of an inner core of nucleic acid enclosed by a outer surface of the envelope. These are known as
capsid, which is made of capsomers; (B) Enveloped virus; (C)
Naked helical virus composed of capsomers wound round the
peplomers. A virus may have more than one type
nucleic acid to form a tubular structure; (D) Envelope helical of peplomers, e.g. the influenza virus carries two
virus in which the tubular capsid is pliable and is enclosed kinds of peplomers, the hemagglutinin which is
within an envelope a triangular spike and the neuraminidase which

www.ebook3000.com
Chap-53.indd 379 15-03-2016 11:33:19 AM
380  |  Section 4: Virology

Fig. 53.2: Shapes and relative sizes of animal viruses of families that infect vertebrates
is a mushroom-shaped structure. Envelope confer information necessary for replication of the virus.
chemical, antigenic and biological properties on The genome may be single-stranded or double-
viruses. stranded, circular or linear, and segmented or
nonsegmented. The type of nucleic acid, its
Functions of Peplomers strandedness, and its size are major characteristics
i. Mediate attachment of the virus to the host- used for classifying viruses into families.
cell receptors.
ii. Attach to receptors on red blood cells.
Susceptibility to physical and
iii. Enzymatic activity chemical agents
iv. Major antigens: For protective immunity. 1. Heat and Cold
i. Heat-labile: With few exceptions, viruses are
D. Viral Nucleic Acids very heat-labile. They are inactivated within
Viruses contain a single kind of nucleic acid— seconds at 56°C, minutes at 37 °C and days at
either DNA or RNA—that encodes the genetic 4°C.

Chap-53.indd 380 15-03-2016 11:33:20 AM


Chapter 53: General Properties of Viruses  | 381
ii. Stable at low temperatures: They are stable the viruses collide in the suspension and adhere
at low temperatures. For long-term storage, to each other resulting in hemagglutination.
they are kept fro­zen at – 70°C. A better method The hemagglutination reac­tion is important in
for prolonged storage is lyophilization or laboratory work. Hemagglutina­tion inhibition
freeze drying (drying the frozen virus under (HI) provides a convenient test for the anti­viral
vacuum). antibody, as hemagglutination is specifically
inhibited by the antibody to the virus. This test can
2. pH be used for detection and quantitation of antibody
Viruses are usually stable between pH values of to virus.
5.0 and 9.0. The influenza virus also carries on its surface
another peplomer, the enzyme neuraminidase
which acts on the receptor and destroys it.
3. Stabilization of Viruses by Salts Neuraminidase is, therefore, called the ‘receptor
Molar concentrations of certain salts (MgCl 2 destroying enzyme’ (RDE). RDE is pro­duced by
Na2 SO4) also protect some viruses (for example, many viruses including cholera vibrios, and is also
poliovirus) against heat inactivation. present in many vertebrate cells. Destruction of the
receptor leads to the reversal of hemagglutination
4. Radiation and the release of the virus from the red cell surface.
Ultraviolet, X-ray, and high-energy particles This is known as elution. Hemagglutination and
inactivate viruses. elution also help in purifying and concentrating
the virus.
5. Disinfectants
i. Bacteria are killed in 50% glycerol saline but
Viral Hemaggluti­nation Test
this acts as a preservative for many viruses (for The hemagglutination test can be carried out in
example, vaccinia, rabies). test tubes or special plastic trays. When RBCs are
ii. Phenolic disinfectants are only weakly added to serial dilutions of viral suspension, the
virucidal. highest dilution that produces hemagglutination
iii. Oxidizing agents: The most active antiviral provides the hemagglutination titer which is
disinfectants are oxidizing agents, such as defined in the form of HA units. Red cells which
hydrogen peroxide, potassium permanganate are not agglutinated settle at the bottom in the
and hypochlorites. Chlorination of drinking form of a ‘button,’ while the agglutinated cells are
water kills most viruses. Some viruses (such as seen spread into a shield-like pattern (Fig. 53.3).
hepatitis virus, polio viruses) are relatively The test serves to titrate killed influ­enza vaccines
resistant to chlorination. Formaldehyde and as the inactivated virus can also hemagglutinate.
beta-propiolactone are actively virucidal and Hemagglutination is a convenient method of
are commonly employed for the preparation detection and assay of the influenza virus.
of killed viral vaccines. Hemagglutination is stable with other viruses.
Hemagglutination appears to be a reversible state
6. Lipid Solvents of equilibrium between the virus and erythrocytes
in the case of arboviruses, being influenced by
Enveloped viruses which possess lipid-containing slight variations in pH and temperature.
envelope are sensitive to lipid solvents, such as
ether, chloroform and bile salts and the naked
viruses are resistant to them.
Viral replication
The genetic information necessary for viral replica­
7. Antibiotics tion is contained in the viral nucleic acid but lacking
Antibiotics active against bacteria are completely biosynthetic enzymes, the virus depends on the
ineffective against viruses.

Viral hemagglutination
A large number of viruses contain hemagglutinin
spikes (peplomers) on the capsid or envelope
which can agglutinate red cells of different Fig. 53.3: Viral hemagglutination. Virus containing fluid is
diluted in doubling dilutions and 0.5% suspension of chick
species. Hemagglutinin of influenza virus is due
red cells added. There is diffuse widespread even pattern
to the presence of hemaggluti­nin spikes on the on the bottom of the wells in the plastic plate where virus is
surface of the virus. When RBCs are added to present. The cells settle down to a button like aggregate with
serial dilutions of viral suspension, the RBCs and sharp edges where no virus is present

www.ebook3000.com
Chap-53.indd 381 15-03-2016 11:33:21 AM
382  |  Section 4: Virology
synthetic machinery of the host cell for replication. ii. Translation of the mRNA into ‘early
The viral multiplication cycle can be divided proteins’. These are enzymes which initiate
into six sequential phases, though the phases and maintain synthesis of virus components.
may sometimes be overlapping: (1) adsorption They may also induce shut down of host
or attachment, (2) penetration, (3) uncoating, protein and nucleic acid synthesis.
(4) biosynthesis, (5) maturation, and (6) release. iii. Replication of viral nucleic acid.
iv. Synthesis of ‘late’ or structural proteins,
1. Adsorption and Absorption which are the components of daughter virion
Virions come in contact with cells by random capsids.
collision but adsorption or attachment is specific Viruses have been categorized into six classes
and is mediated by the binding of virion surface by Baltimore (1970) based on thier replication
structures, known as ligands, to receptors on mechanisms (Table 53.2).
cell surface. In case of influenza virus, a surface
glycoprotein (the hemagglutinin) binds specifically 5. Maturation
to sialic acid residue of glycoprotein receptor sites Newly synthesized viral genomes and capsid
on the surface of respiratory epithelium. In case polypep­tides assemble together to form progeny
of human immunodeficiency virus-1 (HIV1), viruses. Assembly of the various viral components
surface glycoprotein gp120 acts as a ligand. It binds into vir­ions occurs shortly after the replication
to the CD4 60 kDa glycoprotein on the surface of of the viral nucleic acid and may take place in
mature T lymphocytes. either the nuc­leus (herpes and adenoviruses) or
cytoplasm (pic­orna and poxviruses). In case of
2. Penetration enveloped viruses, the envelopes are derived from
After binding, the virus particle is taken up inside Table 53.2  Baltimore classification of viruses
the cell.This is accomplished by receptor-medi­ based on replication
ated endocytosis (viropexis), with uptake of
the ingested virus particles within endosomes. Class
Enveloped viruses fuse their membranes with 1. Double stranded DNA viruses: In the case of fully
cellu­lar membranes to deliver the nucleocapsid double stranded DNA viruses, the DNA enters the
or genome directly into the cytoplasm. host cell nucleus and uses the host cell enzymes
for transcription.
Examples: (i) Hepadnaviruses, (ii) Poxviruses
3. Uncoating 2. Single stranded DNA viruses: The DNA molecule
This is the process of stripping the virus of its moves into the host cell nucleus and is converted
outer layers and capsid so that the nucleic acid is into the duplex form. Transcription is achieved by
host enzymes, for example, parvovirus
released into the cell. With most viruses, uncoating
is affected by the action of lysosomal enzymes of 3. Double stranded RNA viruses: The double
the host cell. stranded RNA is transcribed to mRNA by viral
polymerases, e.g. reoviruses
4. Single stranded RNA viruses: are classified into
4. Biosynthesis two categories
This phase includes synthesis not merely of the (i) Positive strand (plus strand, positive sense):
viral nucleic acid and capsid protein but also of The viral RNA itself act as the mRNA. Viral RNA
is infectious by itself and is translated directly
enzymes necessary in the various stages of vi­ral
into viral proteins in the host cell cytoplasm e.g.
synthesis, assembly and release. In addition, picorna-, togaviruses
cer­tain ‘regulator proteins’ are also synthesized (ii) The negative strand (minus sense) RNA
which serve to shut down the normal cellular viruses: The RNA is ‘antisense’, with polarity
metabolism and direct the sequential production opposite to mRNA. They possess their own RNA
polymerases for, mRNA transcription. Extracted
of viral compo­nents. In general, most DNA viruses
nucleic acids from these viruses are not infectious
synthesize their nucleic acid in the host cell for example rhabdo-, orthomyxo-, paramyxo­viridae
nucleus. The excep­tions are the poxviruses. Most
5. Retroviridae exhibit a unique replicative strategy.
RNA vi­ruses synthesize all their components in Their single stranded RNA genome is con­verted
the cyto­plasm, except for orthomyxoviruses, some into an RNA: DNA hybrid by the viral reverse
paramyxoviruses and retroviruses. Viral protein is transcriptase (RNA directed DNA polymerase)
synthesized only in the cytoplasm. enzyme. Double stranded DNA is then synthesized
from the RNA: DNA hybrid. The double stranded
DNA form of the virus (provirus) is integrated into
Steps of Biosynthesis the host cell chromosome. This integration may
Transcription of messenger RNA (mRNA)
i. lead to transformation of the cell and development
of neoplasia
from the viral nucleic acid.

Chap-53.indd 382 15-03-2016 11:33:21 AM


Chapter 53: General Properties of Viruses  | 383
the host cell nuclear membrane (herpes virus) and Yield of progeny virions occurs only if the cells are
from plasma membrane when the assembly occurs simultaneously infected with a helper virus, which
in the cytoplasm of the host cell (orthomyxoviruses can supplement the genetic defi­ciency.
and paramyxoviruses). Example: (i) Hepatitis D virus and adeno-
associated satellite viruses which replicate only
6. Release in the presence of their helper viruses - hepatitis
Viruses can be released from cells after lysis of B and adenoviruses respectively.
the cell, by exocytosis, or by budding from the
plasma mem­brane. Viruses that exist as naked Cultivation of viruses
nucleocapsids may be released by the lysis of the
Because viruses are obligate intracellular
host cell (polioviruses) or they may be extruded by
parasites,their growth requires susceptible host
a process which may be called reverse phagocyto­
cells capable of replicating them. They cannot be
sis. Release of many enveloped viruses occurs
grown on any inanimate culture medium. Three
after budding from the plasma membrane with­out
methods are employed for the cultivation of viruses:
killing the cell.
A. Animal inoculation
B. Embyronated eggs
Eclipse phase C. Cell culture.
The virus cannot be dem­o nstrated inside the
host cell from the stage of penetration till the A. Animal Inoculation
appearance of mature daughter virions. This period
Uses of Animal Inoculation
is known as the ‘eclipse phase.’ The time tak­en for
a single cycle of replication is about 15–30 minutes i. Primary isolation of certain viruses
for bacteriophages and about 15–30 hours for ii. For the study of pathogenesis, immune
animal viruses. response and epi­demiology of viral diseases
iii. For the study of oncogenesis.
Abnormal replicative cycles
Animals
1. Incomplete Viruses 1. Monkeys: Monkeys find only limited appli­
A proportion of daughter virions that are produced cation in virology.
may not be infective. This is due to defective 2. Mice: Infant (suckling) mice are very suscep­
assembly. Such ‘incomplete viruses’ are seen in tible to coxsackie and arboviruses. Mice may
large pro­portions when cells are infected with a be inoculated by several routes—intracerebral,
high-dose of the influenza virus. The virus yield will subcutaneous, intraperitoneal or intranasal.
have a high hemagglutinin titer but low infectivity. The growth of the virus in inoculated animals
This is known as the von Magnus phenomenon. may be indicated by death, disease or visible
lesions. The viruses are identified by testing
2. Abortive Infections for neutralization of their patho­genicity for
animals, by standard antiviral sera.
Abortive infections fail to produce infectious
progeny, either because the cell may be
nonpermissive and unable to support the expression B. Embyronated Eggs
of all viral genes or because the infecting virus may The embryonated hen’s egg was first used for the
be defective, lacking some functional viral gene. cultivation of viruses by Goodpasture (1931) and
the method was further developed by Burnet. The
3. Latent Infection embryonated eggs (8–11 days old) are inoculation
by several routes for the cultivation of viruses such
A latent infection may ensue, with the persistence
as chorioallantoic membrane (CAM), allantoic
of viral genomes, the expression of none or a few
cavity, amniotic cavity and yolk sac (Fig. 53.4). After
viral genes, and the survival of the infected cell.
inoculation, eggs are incubated for 2–9 days.
1. Chorioallantoic membrane (CAM): Inocul­
4. Defective Viruses ation on the chorioallantoic membrane
Viruses which are genetically deficient and (CAM) produces visible lesions (pocks). Each
therefore incapable of producing infectious infectious virus particle can form one pock.
daughter virions without the helper activity of Pock counting, there­fore, can be used for
another virus are known as’defective viruses’. the assay of pock-forming viruses. Different
Some viruses are genetically defective and they viruses have different pock morphology, e.g.
are unable to give rise to fully formed progeny. herpesvirus, smallpox virus (variola), vaccinia.

www.ebook3000.com
Chap-53.indd 383 15-03-2016 11:33:21 AM
384  |  Section 4: Virology
and a buffering system generally consisting of
bicarbonate in equilibrium with atmosphere
containing about 5% carbon dioxide. This is
supplemented with upto 5% calf or fetal calf serum.
Antibiotics are added to prevent bacterial con­
taminants and phenol red as indicator.

Classification of Cell Cultures


Cell cultures are classified into three types based
on their origin, chromosomal characters and the
number of generations through which they can be
maintained, (Table 53.3).
Fig. 53.4: Cross-section of an embryonated hen’s egg
1. Primary cell cultures: These are normal cells
fresh­ly taken from the body and cultured. They
2. Allantoic cavity: Inoculation into the allanto­ic
are capable of only limited growth in culture (5–10
cavity provides a rich yield of influenza and
passages). Primary cell cultures are useful for the
some paramyxoviruses for vaccine production.
isolation of viruses and their cultivation for vaccine
Other chick embryo vaccines are yellow fever
production. Important examples are monkey
(17D strain) and rabies (Flury strain) vaccines.
kidney; human embryonic kidney, human amnion
Duck eggs are bigger and provide a better
and chick embryo cell cultures.
yield of rabies vi­r us and were used for the
preparation of the inactivated, non-neural 2. Diploid cell strains: These are cells of a single
rabies vaccine. type that retain the original diploid chromosome
3. Amniotic sac: Inoculation into the amniotic number and karyotype during serial subcultivation
sac is employed for the primary isolation of the for a limited number of times. They undergo
influenza virus. ‘senescence’ after about fifty serial passages.
4. Yolk sac: Yolk sac inoculation is used for the They are useful for isolation of some fastidious
cultivation of some viruses, chlamydiae, pathogens and for the production of viral vaccines,
Coxiella burnetti and rickettsiae. e.g. rabies vaccine is produced by cultivation of the
fixed rabies virus in WI-38 human embryonic lung
C. Tissue Culture cell strain.
Three types of tissue cultures are available:
Table 53.3  Some cell cultures in common use
1. Organ culture: Small bits of organs can be
main­tained in vitro, preserving their original Type Name of the cell culture
architecture and function. Organ cultures are A. Primary cell 1. Rhesus monkey kidney cell
useful for the isolation of some viruses which cultures culture
appear to be highly specialized parasites of 2. Human amnion cell culture
3. Chick embryo fibroblast cell
certain organs. For example, the tracheal ring culture
organ culture for the isolation of coronavirus,
B. Diploid cell 1. WI-38 (Human embryonic lung
a respiratory pathogen.
strains cell strain)
2. Explant culture: Fragments of minced tissue 2. HL-8 (Rhesus embryo cell strain)
can be grown as ‘explant’ embedded in plasma C. Continuous cell 1. HeLa (Human carcinoma of
clots. This method is now seldom employed in lines cervix cell line)
virology. 2. HEP-2 (Human epithelioma of
3. Cell cultures: This is the type of culture larynx cell line)
3. KB (Human carcinoma of
routinely employed for growing viruses. Tissues
nasopharynx cell line)
are dissociated into the component cells by the 4. McCoy (Human synovial
action of proteolytic enzymes, such as trypsin carcinoma cell line)
and mechanical shaking. The cells are washed, 5. Detroit-6 (Sternal marrow cell
counted and suspended in a growth medium. line)
6. Chang C/I/L/K (Human
The cell suspension is dispensed to the glass
conjunctiva (C),
surface and on incubation, divide to form a Intestine (I), Liver (L) and Kidney
con­fluent monolayer sheet of cells covering the (K) cell lines)
surface within about a week. 7. Vero (Vervet monkey kidney
Growth me­dium: The essential constituents of cell line)
8. BHK-21 (Baby hamster kidney
the growth me­dium are physiologic amounts of
cell line)
essential amino acids and vitamins, salts, glucose,

Chap-53.indd 384 15-03-2016 11:33:22 AM


Chapter 53: General Properties of Viruses  | 385
3. Continuous cell lines: These are cells of a Main Types of CPE
single type, usually derived from cancer cells, i. Rounding of cells: Viral replication may lead
that are capa­ble of continuous serial cultivation to nuclear pyknosis, rounding, refractility,
indefinitely. Standard cell lines derived from degenera­tion. This is seen in pic­ornaviruses.
human cancers, such as HeLa, Hep-2 and KB cell ii. Cell necrosis and lysis: Enteroviruses pro­
lines have been used in laboratories throughout duce rapid CPE with crenation of cells and
the world for many years. These cell lines may be degeneration of the entire cell sheet.
maintained by serial subculti­vation or stored in
iii. Syncytium formation: Some viruses (measles,
the cold (–70°C) for use when nec­essary. Some
respiratory syncytial virus, human immuno­
cell lines are now permitted to be used for vaccine
deficiency virus (HIV) lead to syncytium
manufacture, for example, Vero cell for rabies
formation in which infected cells fuse with
vaccine. Other cell lines (and their sources) are in
neighboring infected or uninfected cells to
common use (Table 53.4).
form giant cells containing several nuclei.
iv. Discrete focal degeneration: In Herpes virus.
Detection of virus growth in cell v. Rounding and aggregation: Adenovirus
culture produces large granular clumps resembling
Virus growth in cell cultures can be detected by the bunches of grapes.
follow­ing methods:
2. Metabolic Inhibition
1. Cytopathic Effect When viruses grow in cell cultures, cell metabolism
Many viruses cause morphologi­c al changes is inhibited and there is no acid production. This
in cultured cells in which they grow. These can be made out by the color of the indicator
changes can be readily observed by microscop­ic (phenol red) incorporated in the medium.
examination of the cultures. These changes are
known as ‘cytopathic effects’ (CPE) and the 3. Hemadsorption
viruses causing CPE are called ‘cytopathogenic When hemagglutinating viruses (such as influenza
viruses.’ Most viruses produce some obvious and parainfluenza viruses) grow in cell cultures,
cytopathic effect in infected cells that is generally their presence can be indicated by the addition
characteristic of the viral group (Figs 53.5A to D). of guinea pig erythrocytes to the cultures. The

A B

C D
Figs 53.5A to D: A. Normal vero cell monolayer. B. Vero cell monolayer infected with Coxsackie B virus C. HeLa cell
monolayer. D. HeLa cell monolayer infected with Coxsackie virus B3, stained after 48 hours

www.ebook3000.com
Chap-53.indd 385 15-03-2016 11:33:22 AM
386  |  Section 4: Virology
erythro­cytes will adsorb onto the surface of cells 1. Total Virus Particles
if the viruses are multiplying in the culture. This is
A. Electron Microscopy
known as ‘hemadsorption.’ This reaction becomes
positive before cyto­pathic changes are visible and By simple negative staining, the virus particles in
in some cases occurs in the absence of cytopathic a suspension can be counted directly under the
effects. electron microscope. The virus suspension can
be mixed with a known latex particles. The ratio
4. Interference between particles under the electron microscope
gives an indication of the virus count.
The growth of a noncytopathogenic virus in cell
culture can be tested by the subsequent challenge
with a known cytopathogenic virus. The growth of B. Hemagglutination
the first will inhibit infection by the sec­ond virus A convenient method of quantitation is the
by interference. determination of hemagglutination titers with
Example: Rubella virus does not produce cytopathic hemagglutinating viruses. Hemagglutination is not
changes, although they multiply within the cell. a very sensitive indicator of the presence of small
A known cytopathogenic challenge virus is then amount of virus particles. Thus, approximately
introduced into the cells. No CPE will be seen in the 10 7 influenza virions are required to produce
cell culture as replication of challenge virus will be macroscopic agglutination.
prevented because of interference by rubella virus.
2. Infectious Virion Assay
5. Transformation Two types of infectivity assays can be carried out—
Tumor forming (oncogenic) viruses induce cell ­quantitative and quantal assays.
‘transformation’ and loss of contact inhibition,
so that growth appears in a piled-up fashion A. Quantal Assays
producing microtumors. Some herpes viruses,
adeno­viruses, hepadnaviruses, papovaviruses and Quantal assays only indicate the pres­e nce or
retro­viruses (human T cell lymphotropic virus type absence of infectious viruses. Quantal assays of
I) can transform cells. infectivity can be carried out in animals, eggs
or tissue culture for those viruses that do not
6. Immunofluorescence form plaques, and the effects of the virus can
be observed. Examples of endpoints used for
Cells from virus infected cultures can be stained by
infectivity titration are the death of the animal,
fluorescent conjugated antiserum and examined
production of hemagglutinin in allantoic fluid or
under the UV microscope for the presence of
the appearance of CPE in cell cultures. The titer is
virus antigen. This gives posi­tive results earlier
usually expressed as the 50 percent infectious dose’
than other methods and, there­fore, finds wide
(1D50) per mL, which indicates the highest dilution
application in diagnostic virology.
of the inoculum that would produce an effect in
50% of animals, eggs or cell cultures inoculated.
7. Detection of Virus-specific Nucleic Acid
Molecular-based assays, such as polymerase chain B. Quantitative Infectivity Assay
reaction, provide rapid, sensitive, and specific
Quantitative assays measure the actual number of
methods of detection.
infectious particles in the inoculum. Two methods
are available—plaque assay in monolayer cell
8. Detection of Enzymes culture and pock assay on chick em­bryo CAM.
The virus isolate can be identified by detection
of viral enzymes, such as reverse transcriptase in i. Plaque Assay
retroviruses, in the culture fluid. A viral suspension is add­ed to a monolayer of
cultured cells in a bottle or Petri dish, and after
9. Electron Microscopy allowing time for absorption, the medi­u m is
removed and replaced with a solid agar gel to
Viruses have distinctive appearances and can be
prevent virus spreading throughout the culture,
detected by electron microscopy of ultra thin sec­tions
to ensure that the spread of progeny virions is
of infected cells.
confined to the immediate vicinity of infected cells.
In this system, each infectious viral particle gives
Viral assay
rise to a localized focus of infected cells that can
The virus content of a specimen can be assayed in be seen with the naked eye. Such foci are known as
two ways: Either with reference to the total virus ‘plaques’ and each plaque indicates an infectious
particles or with reference to infectious virion only. virus (Fig. 53.6).

Chap-53.indd 386 15-03-2016 11:33:22 AM


Chapter 53: General Properties of Viruses  | 387
importance in laboratory studies is the conditional
lethal mutant. These are mutants which are able to
grow under certain conditions (called permissive
conditions), but are lethal, that cannot grow
under certain other specified conditions (called
nonpermis­sive or restrictive conditions). There
are different types of conditional lethal mutants
but the types most widely employed in genetic
studies are the ‘tempera­t ure sensitive’ (ts)
mutants. These can grow at low (permissive)
temperature (28–31°C), but not at a higher
(restrictive) temperature (37°C). Because of their
low virulence, these mutants have been used
extensively in att­empts to produce attenuated live
virus vaccine.

B. Recombination
When two different, but related, viruses infect a
Fig. 53.6: Plaque formation in monkey kidney cells by cell si­multaneously, genetic recombination may
poliovirus take place. The two viruses exchange segments
of nucleic acid between them so that a hybrid
ii. Pock Assay is formed possessing genes from both parents.
Certain viruses, e.g. herpes and vaccinia, form Recombinants may occur between (1) two active
pocks when inoculated onto the chorioallantoic (infectious) viruses, (2) one active and one inactive
membrane of an embryonated egg. Such viruses virus, and (3) two inactive viruses.
can be assayed by counting the number of pocks i. Recombinants between two active (infect­
formed on CAM by appropriate inocula of vi­rus. ious) viruses: When two inactive viruses
This is known as pock assay. markers (such as pock morphology or antigenic
properties) are grown together, recombinants
Viral genetics may be derived that possess the distinctive
Viruses obey the laws of genetics like all other ‘living properties of both parents. Thus, if a human
beings’. Several properties of viruses, such as virulence and an avian strain of influenza virus (whose
and antigenicity are under genetic control. Main hemagglutinin and neuraminidase antigens
mechanisms for genetic modification in viruses are: are different and easily identifiable) are grown
A. Mutation. together, a hybrid may be obtained with
B. Recombination. the he­magglutinin of one parent and the
C. Nonheritable variations: In addition, viruses neuraminidase of the other. This may be one
may exhibit many nonheritable vari­ations due of the ways by which the pandemic strains of
to gene product interactions. the influenza virus originate in nature.
ii. Recombinants between one active and one
A. Mutation inactive virus: Cross-reactivation or marker
rescue
It is a random, undirected and heritable variation. When a cell is ‘infected’ with an active virus
The frequency of mutation in viruses is about 10-4 and a different but related inactivated virus,
to 10-8, approximately the same as in bac­teria. progeny pos­sessing one or more genetic traits
Mutations, therefore, occur during every viral of the inactivated vi­rus may be produced. This
infection. Many mutations are lethal, because phenomenon is known as cross-reactivation
the mut­ated virus is unable to replicate. A mutant or marker rescue.
becomes evident only if the mutation confers some iii. Recombinants between two inactive viru­
readily observable property or affords the mutant ses: Multiplicity reactivation: When a cell is
virus some selection or survival advantage. Mutation ‘infected’ with a large dose (high multiplicity)
may occur spontaneously or may be induced by of a single virus inactivated by UV irradiation,
mutagens, physical agents, such as UV light or live virus may be produced. The different
irradiation or chemical agents such as 5-fluorouracil. virions that cause multiple infection of a
cell may have suffered damage to different
Conditional lethal Mutant genes. Thus from the total genetic pool it may
Mutations in essential genes are termed lethal be possible to obtain a full complement of
mu­tations. A class of mutants that are of great undamaged genes. This phenomenon is called

www.ebook3000.com
Chap-53.indd 387 15-03-2016 11:33:22 AM
388  |  Section 4: Virology
multiplicity reactivation. There is the potential Classification of viruses
danger of multiplicity reactivation taking place
following the administration of UV irradiated Main Criteria Used for the Classification
vac­cines. of Viruses
Pseudovirion: As a general rule, virus capsids 1. Type of nucleic acid: Viruses are classified
enclose viral nucleic acids. Sometimes segments into two main divisions depending on the type
of host nucleic acid become encapsidated instead. of nucleic acid they possess: Riboviruses are
This is known as pseudovirion. For example, in those containing RNA and deoxyriboviruses
a papovavirus capsid, a linear piece of host DNA are those containing DNA.
rough­ly the same size as the papovavirus genome 2. Number of strands of nucleic acid: Single-or
may be found. double-stranded, linear, circular, circular with
breaks, segmented;
C. Nongenetic interactions 3. Polarity of the viral genome: RNA viruses in
1. Phenotypic Mixing which the viral genome can be used directly
as messenger RNA are by convention termed
When two different viruses infect the same cell, some
‘positive-stranded’ and those for which a
‘mix up’ may take place during assembly so that
transcript has first to be made are termed
progeny genome of one virus may be surrounded by
‘negative-stranded’
the capsid belonging entirely or partly to the other
4. The symmetry of the nucleocapsid
virus. This is known as phenotypic mixing. This is
not a stable variation. On subsequent passage, the 5. The presence or absence of a lipid envelope.
capsid will be found to be of the original type only. If
the genome is encased in a completely heterologous Short Descriptions of the Major Groups
protein coat, this extreme example of phenotypic of Viruses
mixing may be called “pheno­typic masking” or DNA Viruses (Fig. 53.7)
“transcapsidation.” 1. Herpesviridae family: Human herpesviruses
include herpes simplex types 1 and 2 (oral
2. Genotypic Mixing or Heterozygosis
and genital lesions), vari­c ella-zoster virus
It results from the incorporation of more than (chickenpox and shingles), cytomegalovirus,
one complete genome into a single virus particle. Epstein-Barr virus (infectious mononucleosis
There is no recombination be­tween the different and association with human neoplasms),
genomes so that the two kinds of viral progeny are human herpesviruses 6 and 7 (T Iymphotropic),
formed on passage. and human herpesvirus 8 (associated with
Kaposi’s sarcoma). Other herpesviruses occur
3. Complementation in many animals.
The basis for complementation is that one virus 2. Hepadnaviridae family: This consists of
provides a gene product in which the second is the human hepatitis type B virus and related
defective, allowing the second virus to grow. The viruses of animals and birds. (The name comes
geno­types of the two viruses remain unchanged. from hepa = liver, and dna for DNA core.)
If both mutants are defective in the same gene 3. Papovaviridae family: Two genera have been
product, they will not be able to complement each recognized. Papillomavirus and Polvomavirus.
other’s growth. Papillomavirus is also a former member of the
Papovaviridae family. Polyomaviruses were
4. Interference formerly a part of the Papovaviridae family
Infection of either cell cultures or whole animals before it was split into two families.
with two viruses often leads to an inhibition of 4. Adenoviridae family: Members have been
multiplica­tion of one of the viruses, an effect called classified into two gen­era: Mastadenovirus
interference. (mammalian adenoviruses) and Aviadenovirus
Viral interference has been applied in the field in (adenoviruses of birds).
controlling poliomyelitis outbreaks by introducing 5. Parvoviridae family: Three genera have been
into the population, the live attenuated poliovirus described: Parvovirus, Adenosatellovirus and
vaccine. The vaccine virus interferes with the spread Densovirus.
of wild poliovirus and halts the outbreak. 6. Poxviridae family: The family is divided into
several genera. All poxviruses tend to produce
5. Enhancement skin lesions. Some are patho­genic for humans
Mixed infection of cells may some­times lead to (smallpox, vaccinia, molluscum conta­giosum);
increased virus yield or greater CPE. This is known others that are pathogenic for animals and can
as ‘enhancement’. infect humans (cowpox, monkeypox).

Chap-53.indd 388 15-03-2016 11:33:22 AM


Chapter 53: General Properties of Viruses  | 389

Fig. 53.7: Summary of the classification of DNA viruses

Fig. 53.8: Summary of the classification of RNA viruses

RNA Viruses (Fig. 53.8) 4. Bunyaviridae Family


1. Picornaviridae Family Five genera are established—the large genus
Three genera are of medical importance: Bunyavirus containing about 150 species-and four
i. Enterovirus, including polio, coxsackie, echo other genera—Hantavirus, Nairovirus, Phlebovirus,
and several other related viruses. Ukuvirus, and many unas­signed viruses.
ii. Rhinovirus, including human, bovine and 5. Arenaviridae Family
equine rhinoviruses.
iii. Hepatovirus: Hepatitis A virus. Only one genus Arenavirus has been recognized.
Species include lym­phocytic choriomeningitis
virus, Lassa and members of the Tacaribe complex.
2. Orthomyxoviridae Family
Only one genus lnfluenzavirus has been recognised. 6. Rhabdoviridae Family
Influenzavirus type C possesses several distinctive Two genera have been recognized:
features and may have to be sepa­rated into a new i. Vesiculovirus, containing vesicular stomatitis
genus. virus, Chandipura virus (isolated from human
in India) and re­lated species.
3. Paramyxoviridae Family ii. Lyssavirus, containing the rabies virus and
Three genera have been recognized: related viruses such as Lagos bat, Mokola,
i. Paramyxovirus which consists of the Duvenhage and oth­ers.
Newcastle disease virus, mumps virus and
parainfluenza viruses of humans, other 7. Togaviridae Family
mammals and birds. Three genera have been described:
ii. Morbillivirus, containing measles, canine i. Alpha virus, consisting of viruses formerly
distem­per, rinderpest and related viruses. classi­fied as Group A arboviruses.
iii. Pneumovirus, containing respiratory syncytial ii. Rubivirus, consisting of the rubella virus and
vi­rus of humans and related viruses. has no arthropod vector.

www.ebook3000.com
Chap-53.indd 389 15-03-2016 11:33:24 AM
390  |  Section 4: Virology
iii. Pestivirus, consisting of the mucosal disease without a protein coat. They cause disease of plants.
virus, hog cholera vi­rus and related viruses. Viroids are agents that do not fit the definition of
classic viruses.
8. Coronaviridae Family
Only one genus Coronavirus has been recognized. PRIONS
Toroviruses, which cause gastroenteritis, form a Prions (proteinaceous infectious particles) are
distinct genus. infectious particles composed solely of protein
with no detectable nucleic acid. Unlike viruses, the
9. Retroviridae Family: (Re = reverse, tr = agents are resistant to a wide range of chemical and
transcriptase) physical treatments. They are highly resistant to
These are RNA tumor viruses and related agents. inactivation by heat, formaldehyde, and ultraviolet
The characteristic biochemical feature is the light that inacti­vate viruses. The prion protein is
presence of RNA-dependent DNA polymerase encoded by a single cellular gene.
(reverse transcriptase) within the virus that Prion diseases: Prion diseases, called “transmissible
produces a DNA copy of the RNA genome. This spongiform encephalopathies,’’ include scrapie in
DNA becomes circularized and integrated into host sheep, mad cow disease in cattle, and kuru and
chromosomal DNA. The virus is then replicated Creutzfeldt–­Jakob disease in humans. Prions do
from the integrated “provirus” DNA copy. Three not appear to be viruses. It has been suggested that
subfamilies are recognized: they may also be responsible for some other chronic
1. Oncovirinae, the RNA tumor virus group. neuro­logical degenerative diseases of humans.
2. Spumivirinae, the foamy virus group (Spuma =
foam) Key Points
3. Lentivirinae, (Lenti = slow) visna and maedi
™™ Viruses are the smallest known infective agents
viruses of sheep belonging to the slow virus ™™ The viruses are obligate intracellular parasites
group. ™™ They do not grow in inanimate media. They are
resistant to antibiotics
10. Flaviviridae Family ™™ The extracellular infectious virus particle is the virion
™™ The viruses vary widely in their size: Range from 20
This group of arboviruses includes yellow fever nm virus (arboviruses) to 300 nm virus (poxvirus)
virus and dengue viruses. ™™ The virion consists of a nucleic acid core, the genome,
surrounded by a protein coat, the capsid
11. Caliciviridae Family ™™ Viruses may have icosahedral (cubical) symmetry,
helical symmetry, or complex symmetry
An important human pathogen is Norwalk virus, ™™ The capsid together with the enclosed nucleic acid
the cause of epidemic acute gastroenteritis. is known as the nucleocapsid. Some viruses are
surrounded by envelopes
12. Filoviridae Family ™™ Three methods are employed for cultivation of
viruses namely animal inoculation, embryonated egg
This contains the Marburg and Ebola viruses inoculation and tissue culture
causing human hemorrhagic fevers. ™™ Embryonated egg inoculation is done by one of the
several routes such as chorioallantoic membrane
13. Reoviridae Family (CAM), allantoic cavity, amniotic sac and yolk sac
™™ Tissue culture are of three types: Organ culture;
Three genera have been recognized: explant culture and cell culture
1. Reovirus, containing reoviruses from humans, ™™ The viruses show variation in their genomic structure
other mammals and birds. by mutations and recombination
2. Orbivirus, containing several species of ™™ Depending on the type of nucleic acids viruses
possess, they are classified into two groups:
arboviruses such as blue tongue virus, African Deoxyriboviruses that contain DNA (DNA virus) and
horse sickness virus. riboviruses that contain RNA (RNA virus)
3. Coltivirus includes Colorado tick fever virus of ™™ Prions (proteinaceous infectious particles) are
humans. infectious particles composed solely of protein with
no detectable nucleic acid
4. Rotavirus including human rotaviruses, calf
™™ Prion diseases, called “transmissible spongiform
di­arrhea virus and related agents. Other genera encephalopathies,’’ include scrapie in sheep, mad
may have to be defined to include plant and cow disease in cattle, and kuru and Creutzfeldt­Jakob
insect viruses belong­ing to this family. disease in humans.

Viroids Important questions


Viroids are small infectious agents which are 1. Describe the various methods of isolation of
circular single stranded RNAs (MW 70,000–120,000) viruses in the laboratory.

Chap-53.indd 390 15-03-2016 11:33:24 AM


Chapter 53: General Properties of Viruses  | 391
2. Describe the classification of viruses. 5. All the following are the examples of enveloped
3. Write short notes on: viruses except:
a. Morphology of viruses a. Polio virus b. Rubivirus
b. Determination of size of viruses c. Adenovirus d. Herpesvirus
c. Viral hemagglutination 6. Which of the following disinfectants is/are effective
d. Recombination in viruses against viruses?
e. Cultivation of viruses
a. Hypochlorite b. Formaldehyde
f. Detection of virus growth in cell cultures
g. Prions c. Hydrogen peroxide d. All of the above
h. Viroids 7. Suckling mice are used for the isolation of:
a. Poxviruses b. Herpesviruses
c. Arboviruses d. All of the above
Multiple choice questions (Mcqs)
8. All are continuous cell lines for growing viruses
1. The viruses show all the following features except: except:
a. They are filterable agents a. HeLa
b. They are obligate intracellular parasites b. HEp-2
c. They contain either DNA or RNA, but not both
c. KB
d. They multiply inside the living cells by using
their synthe­sizing machinery d. Rhesus monkey kidney cell culture
2. The symmetry of poxviruses is: 9. The following routes of inoculating embryonated
a. Icosahedral hen’s egg is/are used for cultivation of viruses:
b. Helical a. Chorioallantoic membrane
c. Complex b. Amniotic cavity
d. None of the above c. Allantoic cavity
3. Which of the following is the largest virus? d. All of the above
a. Smallpox virus b. Adenovirus 10. Which of the following agents is/are prions?
c. Coronavirus d. Parvovirus a. Agent causing kuru
4. Which of the following viruses is/are relatively b. Agent causing scrapie
thermostable? c. Agent causing Creutzfeldt-Jakob disease
a. Hepatitis A virus d. All of the above
b. Human immunodeficiency virus
c. Rubella virus Answers (MCQs)
d. All of the above 1. d; 2 c; 3. d; 4. a; 5. b; 6. d; 7. c; 8. d; 9. d; 10. d

www.ebook3000.com
Chap-53.indd 391 15-03-2016 11:33:24 AM
5454
Chapter

Virus–Host Interactions: Viral Infections

Learning Objectives

After reading and studying this chapter, you should ∙∙ D


 escribe the following: Immunity in virus infections;
be able to: interferons.
∙∙ Describe inclusion bodies
∙∙ Describe routes of transmission of human virus
infections

Interactions between viruses and syncytium formation by paramyxoviruses.


host cells Herpesviruses and some retroviruses also
give rise to syncytia.
Virus–host interactions may be considered at c. Inclusion bodies : Inclusion bodies
different levels—the cell, the individual and are structures with distinct size, shape,
the community. The cellular response to viral location and staining properties that can be
infection may range from no apparent effect to demonstrated in virus infected cells under
cytopathology with accompanying cell death to the light microscope. The appearance of
hyperplasia or cancer. inclusion bodies is the most characteristic
Some vi­ruses, like poliovirus cause cell death histological feature in virus-infected cells.
(cytocidal ef­fect) or even lysis (cytolysis). Others They may be acidophilic (stained by eosin)
may cause cellular proliferation (as molluscum or baso­philic (stained by hematoxylin),
contagiosum) or malignant transformation (as single or multiple, large or small and round
oncogenic viruses). In some instances, the virus or irregular. The presence of inclusion bodies
and host cell enter into a peaceful coexistence, may be of consider­able diagnostic aid. The
both replicating independently without any intracytoplasmic inclusion in nerve cells,
cellular injury, a condition known as ‘steady state the Negri body, is pathognomonic for rabies.
infection.’ They may be situated in the nucleus, in the
1. Cellular factors: The presence of appropriate cytoplasm or in both.
receptors on the surface of the cell determines Intracytoplasmic inclusion bodies: Those
whether virus can adsorb to it and the virus viruses that have cytoplasmic assembly (mainly
gets into the cell. It must replicate in order to RNA vir­uses) yield cytoplasmic inclusions.
establish infection, which may take several These are found in cells infected with rabies
forms. virus (Negri bodies), vac­c inia (Guarnieri
2. Cytopathic effects: Many viruses kill the bodies), fowlpox (Bollinger bodies), molluscum
cells in which they replicate, sometimes with contagiosum (molluscum bodies), para­
characteristic appearances or cytopathic effects myxoviruses and reoviruses.
(CPEs). These are often distinctive enough to Intranuclear inclusion bodies: In general,
give an idea of the virus concerned, a property those viruses that are assembled in the nucleus
that is useful in the diagnostic laboratory. (usu­ally DNA viruses) produce intranuclear
a. Cell lysis: Death of the cell is followed by inclusions. They are found in cells infected with
lysis and release of large numbers of virions. herpesviru­ses, adenoviruses and parvoviruses.
b. Cell fusion: Some cause fusion of Intranuclear inclusion bodies were classi­fied
adjacent cell membranes, leading to into two types by Cowdry (1934). Cowdry type
Chapter 54: Virus–Host Interactions: Viral Infections  | 393
A inclusions are of variable size and granular diseases in man are human immunodeficiency
appear­a nce (as with herpesvirus, yellow virus (HIV) which causes acquired immuno­
fever virus), while type B inclusions are more deficiency syndrome; kuru agent and
circumscribed and often multiple (as with Creutzfeldt-Jakob agent which cause slowly
adenovirus, poliovirus). progressive encephalo­pathy.
Intranuclear and intracytoplasmic inclusion
bodies: Some viruses, such as measles virus and Pathogenesis of viral diseases
cytomegalovirus may produce both intranuclear
To produce disease, viruses must enter a host,
and intracytoplasmic.
come in contact with susceptible cells, replicate,
Nature of the inclusion bodies: The nature of
and produce cell injury. Understanding of viral
the inclusions varies with the virus concerned.
pathogenesis at the mole­cular level is necessary
In many viral infections, the inclusion bodies are
to design effective and specific antiviral strategies.
the site of development of the virions (the viral
Much of our knowledge of viral pathogenesis is
factories) or made up of virus antigens present at
based on animal models, because such systems
the site of virus synthesis, or simply degenerative
can be readily manipulated and studied.
changes produced by viral infection which
confer al­tered staining properties on the cell.
Varia­t ions in the appearance of inclusion Transmission of human virus
material depend largely upon the tissue fixative infections
used. Viruses enter the body through the following routes
3. New cell-surface antigens: These antigens (Table 54.1).
may confer new properties on the cells. This is
particularly important in the case of enveloped 1. Respiratory Tract
viruses that bud from the cell surface (e.g.
Many viruses enter the body through inhaled drop­
herpes-, myxo-, paramyxo-, and retroviruses).
lets expelled from the nose or mouth of infected
Virus coded anti­gens also appear on the surface
persons during talking, coughing or sneezing. This
of cells transformed by oncogenic viruses.
4. Dam­age to the chromosomes of host cells:
Certain viruses such as measles, mumps,
adenovi­ruses, cytomegalovirus and varicella Table 54.1  Transmission of human virus infection
virus cause dam­a ge to the chromosomes Route of Viruses
of host cells. Chromatid gaps and breaks in transmission
chromosome 17 occur frequently in cul­tured 1. Respiratory tract Respiratory viruses: Influenza A, B
cells infected with adenovirus types 12 and 31. and C, parainfluenza types 1–4, RSV,
5. Latent and persistent infections: Viruses have rhinovirus, coronavirus, adenovirus
and coxsackievirus A
evolved mechanisms to continue to survive in Systemic viruses: Measles, mumps,
the face of a strong host immune respo­nse. rubella, varicella-zoster, CMV and EBV
Examples 2. Alimentary tract Many enteroviruses including
i. Herpes simplex types 1 and 2 and varicella- poliovirus types 1–3, HAV, HEV,
zoster viruses: The viruses remain latent in adenoviruses, rotaviruses, Norwalk
and related viruses, coronavirus,
the ganglia, to be reactivated periodically in astrovirus, coxsackievirus A and B
some indi­viduals causing recurrent lesions. and echovirus
ii. Cytome­galovirus (CMV) and Epstein-Barr 3. Skin Minor abrasions: Papillomaviruses,
virus (EBV): are also known to establish molluscum contagiosum, cowpox,
latent infection. orf, milker’s nodes viruses, herpes
iii. Hepatitis B virus (HBV): Some vir­uses like simplex viruses and HBV
Insect bite: Arboviruses
hepatitis B virus (HBV) may cause chronic Animal bite: Rabies virus, herpes
infections which may remain clinically B virus
inapparent for many years. Injection: HBV, HCV, HIV-1, HIV-2,
iv. Subacute sclerosing panencephali­t is HTLV, CMV, EBV and ebola virus
(SSPE): An important example of this type 4. Genital tract Papillomaviruses, herpes simplex
of persist­ent infection is subacute sclerosing viruses, HIV, HTLV, HBV, HCV
panencephali­tis (SSPE). This disease develops 5. Conjunctiva Some adenoviruses and few
between 1 and 10 years after recovery from enteroviruses
measles virus infection. RSV, respiratory syncytial virus; CMV, cytomegalovirus; EBV, Epstein-
v. Human immunodeficiency virus (HIV): Barr virus; HAV, hepatitis A virus; HEV, hepatitis E virus; HIV, human
immunodeficiency virus; HTLV, human T cell leukemia virus
Viruses that produce slowly progressive fatal

www.ebook3000.com
394  |  Section 4: Virology
is the most frequent means of viral entry into the the primary agents responsible for congenital
host. Some viruses, such as influenza A, B and C, defects in humans and produce maldevelopment
parainfluenza types 1–4, respiratory syncytial virus or severe neonatal disease.
(RSV), Rhinovirus, Coronavirus, adenovirus and
coxsackievirus are restricted to respiratory tract Spread of virus in the body
where they multiply and produce local disease.
Other viruses, multiply locally to initiate a silent
Generalized Infections
local infection which is followed by lymphatic or Sequence of events: The sequence of events,
hematogenous spread to other parts of the body summarized as follows:
where more extensive multiplication takes place 1. Entry: Virions enter through an epithelial
before producing genera­lized disease. surface, where they undergo limited replication.
2. Migration: They then migrate to the regional
2. Alimentary Tract lymph nodes where some are taken up by
The alimentary tract is the next most important macrophages and inactivated, but others enter
route of entry for viruses. The viruses that initiate the bloodstream.
infection of humans via the alimentary tract are 3. Primary viremia: Virions which enter the
many enteroviruses inc­luding poliovirus types 1–3, bloodstream. This is the primary viremia
hepatitis A virus (HAV), hepatitis E virus (HEV), 4. Secondary viremia: From the blood, the virus
adenoviruses, rota­viruses, Norwalk and related gains access to the large reticuloendothelial
viruses, Coronavirus, Astrovirus, coxsackie A and organs—liver, spleen, and bone marrow—in
B and echovirus. Polioviruses, HAV and HEV do which it again multiplies, and a large amount
not produce any intestinal symptoms, but cause of virus is pro­duced which again spills over into
generalized inf­ection. the bloodstream, causing a secondary viremia.
This heralds the onset of clinical symptoms (the
3. Skin prodromal phase in eruptive fevers).
Of the viruses that enter through the skin, only a few 5. Target organ: The virus reaches the target
produce local lesions. Some obtain entry through organ through the bloodstream. Multiplication
small abra­sions of the skin (papillomaviruses, in the target sites produces the distinctive
molluscum contagiosum, cowpox, orf, milker’s lesions.
nodes viruses, herpes simplex and HBV), insect
bite (arboviruses), animal bite (ra­b ies virus, Significance of the
herpes B virus) or injection (HBV, HCV, HIV, HTLV,
incubation period
CMV, EBV and Ebola virus) (Table 54.1).
Viruses that are introduced deeper into the The incubation period represents the time taken
dermis have access to blood vessels, lymphatics, for the virus to spread from the site of entry to
dendritic cells, and macrophages and usually the organs of viral multiplication and hence to
spread and cause systemic infections. Rabies virus the target organs for the production of lesions. Its
travels along the nerves to the spinal cord or brain. duration is therefore influenced by the relation
between the sites of entry, multiplication and
4. Conjunctiva lesion. They are classifed into four main groups:
The conjuctiva also may act as a portal of entry for short, medium, long, and very long.
viruses. This may lead to local disease (adenovirus) 1. Short means less than a week and primarily
or to systemic spread (measles). applies to viruses causing localized infections
that spread rapidly on mucous sur­faces.
5. Genital Tract 2. Medium incubation periods range from about
Papillomaviruses and herpes simplex viruses are 7–21 days.
sexually transmitted and produce lesions on the 3. Long refers to periods measured in weeks or
genitalia and perineum. HIV, HTLV, HBV and HCV months (e.g. 2–6 weeks for hepatitis A and 6–20
are also sexually transmitted but do not produce weeks for hepatitis B). Rabies may also have
local lesions. incubation periods extending for many months.
4. Very long incubation periods are measured
6. Congenital Viral Infections in years, which is why the agents involved were
Congenital infection may occur at any stage from originally termed ‘slow’ viruses. This group
the development of the ovum up to birth. If the comprises the prions and a few’con­ventional’
virus crosses the pla­centa and infection occurs in viruses, such as papovaviruses and measles,
utero, serious damage may be done to the fetus. which very occasionally cause delayed disease
Rubella virus and cytomegalovirus are presently of the central nervous system.
Chapter 54: Virus–Host Interactions: Viral Infections  | 395
Host response to virus infections against viral infections. On the other hand,
macrophages phagocytose viruses and
The outcome of a virus infection is influenced are important in clearing viruses from the
by the virulence of the infecting strain and the bloodstream.
resistance offered by the host. Mechanisms of host
2. Fever: Fever may act as a natural defence
resistance may be immunological or nonspecific.
mechanism against viral infections as most
viruses are inhibited by temperatures above
A. Immunological Response 39°C.
Virions in general are good antigens and induce 3. Hormones: Corticosteroid administration
both antibody- and cell-mediated immunity (CMI). enhances most viral infections. Injudicious
For some diseases such as poxviruses, measles, use of steroids in the treatment of herpetic
herpes sim­plex virus and CMV infections, CMI keratoconjunctivitis may cause blindness.
appears to be the main immunospecific defence,
4. Malnutrition: Malnutrition can exacerbate viral
for others, such as enterovirus and arbovirus
infections, for example, mea­sles produces a much
infections, antibody production appears to be the
higher case fatality in mal­nourished than in well-
main immunospecific defence. However, some
fed children.
viruses have evolved strategies to evade detection
and persist in their host. 5. Age: Most viral infections are commoner and
more dangerous at the two extremes of age.
1. Antibody-mediated Immunity 6. Interferon: Interferons are a family of host
coded proteins produced by cells on induction
In mediating humoral antiviral immunity, the
by viral or nonviral inducers. Isaacs and
important classes of antibodies are IgG, IgM
Lindenmann in 1957 discovered that virus
and IgA. IgG and IgM play a major role in blood
­infected cells produce a soluble factor that
and tissue spaces, while secretory IgA antibody
protects other cells from infection and they gave
is important in protecting against infection by
the name interferon to this antiviral substance.
viruses through the respiratory or gastrointestinal
tracts. Humoral immu­nity protects the host against Mode of action of interferon: On exposure to
reinfection by the same virus. Mechanisms of interferon, cells produce a protein (translation
antibodies effecting virus neutralization inhibiting protein, TIP) which selectively inhibits
Antibodies effect virus neutralization by several translation of viral mRNA, without affecting cellular
mechanisms: mRNA. What has been called TIP is “actually
a mixture of at least three different enzymes (a
i. They may prevent adsorption of the virus to cell
protein kinase, an oligonucleotide synthetase and
receptors, cause enhanced virus degradation
an RNAase) which together block translation of
or prevent release of the progeny virus from
viral mRNA into viral proteins. It has also been
infected cells.
suggested that inhibition of viral transcription
ii. Opsonization of virions for phagocytosis and
may also be responsible for the antiviral activity
killing by macrophages.
of interferon.
iii. Complement acts in conjunction with
antibodies in causing surface damage to
enveloped virions and in producing cytolysis
Synthesis of Interferons
of virus infected cells. Interferons are produced by all vertebrate species.
Nor­mal cells do not generally synthesize interferon
2. Cell-mediated Immunity until they are induced to do so. Viruses also vary
in their capacity to induce interferon, cytocidal
Cell-mediated immunity prevents infection of
and virulent viruses being poor inducers and
target organs and pro­motes recovery from disease
avirulent viruses being good inducers. Infection
by destroying virus and virus-infected cells by any
with viruses is a potent insult leading to induction;
of the following four different processes:
RNA viruses are stronger inducers of interferon
1. Cytolysis by cytotoxic T cells(Tc) cells. than DNA viruses.
2. Cytolysis by natural killer (NK) cells.
3. Antibody-complement-mediated cytotoxicity. Types of Interferon
4. Antibody-dependent cell-mediated cytotoxicity
There are multiple species of interferons that fall
(ADCC).
into three general groups, designated IFN-a, IFN-
b, and IFN-g.
B. Nonimmunological Responses 1. Alpha-interferon (IFN-a): It is produced by
1. Phagocytosis: Polymorphonuclear leukocytes leukocytes following induction by suitable
do not play any significant role in the defence viruses.

www.ebook3000.com
396  |  Section 4: Virology
2. Beta-interferon (IFN-b): It is produced by fibro­ ™™ Interferons are a family of host coded proteins
blasts and epithelial cells following stimulation produced by cells on induction by viral or nonviral
by viruses or polynucleotides. It is a glycoprotein. inducers. Interferons fall into three general groups,
3. Gamma-interferon (IFN-g): It is produced by designated IFN-a, IFN-b, and IFN-g
T lymphocytes, on stimulation by antigens or
mitogens. Important Questions
1. Discuss various methods by which viruses can be
Main Biological Effects of Interferons transmitted to human beings.
1. Antiviral effects: Induction of resistance to 2. Write short notes on:
infections. a. Inclusion bodies b. Interferons
2. Antimicrobial effects: Resistance to intracellular
infections, for example, toxoplasma, chlamydia, Multiple choice questions (MCQs)
malaria. 1. Syncytium formation is the typical cytopathic
3. Cellular effects: Inhibition of cell growth and effect produced by:
proliferation; and of DNA and protein synthesis; a. Adenovirus b. Herpes virus
c. Measles virus d. SV40
increased expression of MHC antigens on cell
2. Intracytoplasmic inclusion bodies are found in
surfaces.
cells infected with:
4. Immunoregulatory effects: Enhanced cytotoxic a. Rabies virus
activity of NK, K and T cells; activation of b. Vaccinia virus
macrophage cytocidal activity; modulation of c. Molluscum contagiosum virus
antibody formation; activation of suppressor T d. All of the above
cells; suppression of DTH. 3. All of the following viruses are transmitted by the
respiratory route except:
a. Mumps virus b. Measles virus
Key Points c. Rubella virus d. Norwalk virus
4. All of the following viruses may be transmitted
™™ Virus-host interactions may be considered at the
level of the cell, individual and community through genital tract except:
™™ The cellular response to viral infection may range a. Papillomavirus
from no apparent effect to cytopathology with b. Herpes simplex virus
accompanying cell death to hyperplasia or cancer c. Hepatitis C virus
™™ Inclusion bodies are structures with distinct size, d. Coronavirus
shape, location and staining properties that can be 5. Which type/s of interferon is/are produced by T
demonstrated in virus infected cells under the light lymphocytes?
microscope a. a b. b
™™ Viruses enter the body through the respiratory tract, c. g d. g and b
the alimentary tract, the skin, conjunctiva and the 6. Which type/s of interferon are produced by virus
genital tract; many viruses (cytomegalovirus and infected cells?
rubella) can also be transmitted vertically from a. a
parent to progeny b. b
™™ The immune response is the best and most c. Both of the above
important means of controlling virus infections. d. None of the above
™™ Nonimmunological responses include age;
phagocytosis, fever, hormones, age and interferon Answers (MCQs)
1. c; 2. d; 3. d; 4. d; 5. c; 6. c.
55
Chapter
Laboratory Diagnosis, Prophylaxis and
Chemotherapy of Viral Diseases

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe immunoprophylaxis of viral diseases
be able to: ∙∙ Describe the following: Live viral vaccines; killed
∙∙ Describe laboratory diagnosis of viral infections viral vaccines.

Laboratory diagnosis of A. Direct detection of virus


viral infections B. Virus isolation and growth
C. Detection of viral proteins
1. Indications for Labora­tory Diagnosis D. Detection of viral genetic material
of Viral Infections E. Serology.
i. Diagnosis of diseases caused by viruses
for which antivi­ral chemotherapy is already A. Direct Detection of Virus
available (herpes­viruses, human immuno­ 1. Electron Microscopy (EM)
deficiency virus (HIV). Clinical applications of electron microscopy include
ii. For proper management of the patient detection of rotavirus and hepatitis A virus in fecal
Example: speci­mens, poxviruses in vesicle fluid and herpes
– Rubella, rabies diagnosis virus in brain biopsy tissue.
– Rabies in the animal and postexposure
immunization of the patient prevents the 2. Immunoelectron Microscopy
development of rabies The addition of virus-specific antibody to a
– Primary genital herpes sample can cause viral particles to clump, thereby
– Baby born to an HBsAg positive mother. facilitating the detection and simultaneous
iii. Screening of blood donors: For the prevention identification of the virus (immunoelectron
of some diseases (such as screening for HBV microscopy). This method is useful for the
and HIV in blood donors). detection of enteric viruses, such as rotavirus.
iv. Early detection of dangerous epidemics:
Such as yellow fever, encephalitis, influenza, 3. Fluorescence Microscopy
poliomyelitis, etc. Direct or indirect fluorescent antibody technique
v. To discover new viruses: HIV was discovered can be used to detect virions or viral antigens in
in 1983. frozen tissue sections, acetone-fixed cell smears,
cells from virus infected cultures or vesicles fluid.
2. Specimens Fluorescence microscopy of brain biopsy can
The appropriate specimens should be collected be used for the verification of rabies in the brain
from patients, preserved and transported to of animals suspected to be rabid. This method is
the laboratory in the proper manner along with also useful for the rapid diagnosis of respiratory
pertinent clinical and epidemiological information. infections caused by paramyxoviruses, orthomyxo­
viruses, adenoviruses and herpes viruses.
3. Methods for Laboratory Diagnosis of
Viral Infections 4. Light Microscopy
Laboratory diagnosis of viral infections can be Demonstration of the inclusion body is a routine
carried out by the following methods: diagnostic method. Rabies may be detected

www.ebook3000.com
398  |  Section 4: Virology
through the finding of Negri bodies (rabies virus DNA mole­cule in the clinical specimens are
inclu­sions) in brain cells of animals. first separated, then, allowed to hybridize with
a labeled single-stranded DNA or RNA probe
B. Virus Isolation present in excess. Depending on the type
For virus isolation it is imperative that the specimen of label attached to the probe, hybridized-
be collected properly and transported with least labeled probe can be detected by radiography,
delay to the laboratory. The reasons are that many gamma-counting or a simple colorimetric
viruses are labile and that the samples are sus­ceptible evaluation (dot-blot hybridiza­tion). The DNA
to bacterial and fungal overgrowth. Viruses are best probes are detected with autoradiography
transported and stored on ice and in special media. or with fluorescent or EIA-like methods. By
In general, the methods used for isolation use of nucleic acid probes cytomegalovi­rus,
consist of inoculation into animals, eggs or tissue papillomavirus and Epstein-Barr virus have
culture, after the specimen is processed to remove been identified.
bacterial contaminants. iii. Southern, Northern andd Dot blot analy­sis:
The isolates are identified by neutralization Viral genomes can also be detected in clinical
or other suitable serological procedures. Many sam­ples with the use of dot blot or Southern
viruses (for example adenoviruses, enteroviruses) blot analy­sis. Electrophoretically sepa­rated
are frequently found in normal individuals. The viral RNA (Northern blot-RNA: DNA probe
results of isolation should always be interpreted hybridization) blotted onto a nitrocellulose
in the light of the clinical data. filter can be detected in a similar manner.
iv. Polymerase chain reaction: The polymerase
C. Detection of Viral Proteins chain reaction (PCR) for DNA, reverse
The viral proteins can be assayed by the following transcriptase polymerase chain reaction(RT-
methods: PCR) for RNA, and branched-chain DNA
i. Protein patterns (by electrophoresis)—HSV. (DNA, RNA) assays are becoming very
ii. Enzyme activities (e.g. reverse transcriptase)— important for viral detection.
Retrovirus. Use of the appropriate primers for PCR can
iii. Hemagglutination and hemadsorption— promote a million-fold amplification of a
Influenza virus. target se­quence in a few hours. This technique
iv. Antigen detection: Viral antigens on the cell is especially useful for detecting latent and
surface or within the cell can be detected by integrated sequences of viruses, such as
immunofluorescence and enzyme immuno­ retroviruses, herpes viruses, papillomavi­
assay (EIA). Virus or antigen released from ruses, and other papovaviruses as well as
infected cells can be detected by enzyme- the sequences of viruses present in low
linked immunosorbent assay (ELISA), radio­ concentrations.
immunoassay (RIA), and latex agglutination v. Reverse transcriptase polymerase chain
(LA). reaction (RT-PCR) for RNA: This appro­ach
was very useful for identifying and disting­
D. Detection of Viral Genetic Material uishing the Hantaviruses.
i. The electrophoretic patterns of RNA (influ­ vi. Branched-chain DNA assays: Quantitate
enza, reovirus) or restriction endonuclease viral DNA or RNA much like ELISA.
fragment lengths from DNA viral genomes
are like genetic fingerprints for these viruses. E. Serological Diagnosis
ii). Different strains of HSV-l and HSV-2 can The demonstration of a rise in titer of antibodies
be distinguished in this way by restriction to a virus during the course of a disease is strong
fragment length polymorphism. evidence that it is the etiological agent. For this,
ii. DNA probes: DNA probes with sequences it is essential to examine paired sera, the ‘acute’
complementary to spe­c ific regions of a sample collected early in the course of the disease
viral genome can be used, like anti­bodies, and the ‘convalescent’ sample collected 10–14
as sensitive and specific tools for detecting days later. Examination of a single sample of serum
a virus. Enzyme-labelled or radiolabeled for antibodies may not be meaningful except
nucleic acid (DNA or RNA) sequences when IgM specific tests are done which is present
complementary to unique reg­ions in nucleic during the first 2 or 3 weeks of a primary infection,
acid sequences of most viruses are now generally indicates a recent primary infection.
manufactured commercially. These labeled The serological techniques employed would
complementary sequences are known as depend on the virus but those in general uses
nucleic acid probes. Two strands of the target are neutralization, complement fixation, ELISA,
Chapter 55: Laboratory Diagnosis, Prophylaxis and Chemotherapy of Viral Diseases  | 399
hemagglutination inhibition tests, indirect fluore­ ∙∙ Recombinant live viral or bacterial vectors, e.g.
scent antibody test, latex agglutination test. influenza.

Immunoprophylaxis of Advantages of live-virus Vaccines


viral diseases 1. Single dose: A single dose is usually sufficient
because they multiply in the human host and
A. Acive Immunization provide conti­nuous antigenic stimulation over
Viral vaccines are of the following types (Table 55.1). a period of time resulting in durable immunity.
2. Local immunity: They can be administered
I. Live-virus Vaccines by the route of natural infection so that local
These are prepared from: immunity is induced.
∙∙ Attenuated strains, e.g. yellow fever 3. Wide spectrum of immunoglobulins: Including
∙∙ Temperature sensitive (ts) and cold-adapted secretory IgA to whole range of viral antigens.
(ca) mutants, e.g. influenza 4. Cell-mediated immunity
5. long-lasting immunity
Table 55.1  Viral vaccines in common use 6. Primary vaccine failures are uncommon.
Disease Type of Mode of preparation 7. More economical.
vaccine
Poliomyelitis Live Avirulent strains grown Disadvantages of live-virus Vaccines
Killed in monkey kidney cell 1. Reversion of virulence
culture
2. Heat-labile: and have to be preserved under
Virulent strains grown
in monkey kidney cell strict refrigeration.
culture, formalin-killed 3. Interference: Interference by coinfection with a
Rabies Killed (sample Fixed virus grown naturally occurring, wild-type virus may inhibit
type) in sheep brain and replication of the vaccine virus and decrease
inactivated by phenol or its effectiveness, e.g. with the vaccine strains of
beta propiolactone poliovirus inhibited by concurrent infections by
Killed Virus grown in cell various enteroviruses.
culture and inactivated 4. Contamination: with poten­tially dangerous
with beta propiolactone viruses, such as oncogenic viruses or other
Yellow fever Live (17D) Attenuated virus grown infectious agents.
in chick embryos and 5. Local remote complications
lyophilized
6. Virus may spread from the vaccines to contacts.
Japanese Killed Virus grown in mouse
encephalitis brain and inactivated by
formalin II. Killed Viral Vaccines
Varicella Live Attenuated virus Killed vaccines have been prepared by inactivating
grown in chick embryo viruses with heat, phenol and formalin or beta
fibroblast culture propiolactone. Adverse reactions may also be
Mumps Live Attenuated virus grown reduced by the use of ‘subunit vaccines’ in which
in human diploid cell the virus is split by detergents or other chemicals
culture and only the relevant antigens incorporated in
Influenza Killed Virus disintegrated with the vaccine. Vaccine production by cloning the
(subunit) sodium deoxycholate desired antigen in bacteria or yeast is becoming
Live Virus attenuated by
(attenuated) serial passage in eggs
increasingly common, as in hepatitis B vaccine.
Live (mutant) (ts) mutants which are
Live avirulent Advantages
(recombinant) Recombinants with
surface antigens
1. Stability and safety.
of new strains and 2. They can be given in combination as polyvalent
growth characters of vaccines.
established strains 3. There is also no danger of the spread of the virus
Measles Live Attenuated virus grown from the vaccinee.
in tissue culture
Rubella Live Attenuated virus grown Disadvantages
in tissue culture
1. Extreme care is required in their manufacture
Hepatitis B Cloned HBsAg cloned in yeast to make certain that no residual live virulent
subunit
virus is pres­ent in the vaccine.

www.ebook3000.com
400  |  Section 4: Virology
2. The immunity conferred is often brief and must Table 55.2  Major antiviral compounds used for
be boosted. treatment of viral infections
3. Parenteral administration of killed-virus
Drug Viral spectrum
vaccine, even when it stimulates circulating
Viral polymerase inhibitors
antibody (IgM, IgG) to satisfactory levels, has
sometimes given limited pro­tection because Acyclovir Herpes simplex, varicella-
zoster
local resistance (IgA) is not induced ade­quately.
4. Cell-mediated response to inactivated vac­cines Cidofovir Cytomegalovirus, herpes
simplex
is generally poor.
Foscarnet Herperviruses, HIV-1, HBV
5. Some killed-virus vaccines have induced hyper­
sensitivity to subsequent infection. Ganciclovir Cytomegalovirus
Valacyclovir Herpesviruses
B. Passive Immunization Vidarabine Herpesviruses, vaccinia,
HBV
Passive immunization with human gammaglobulin,
convalescent serum or specific immune globulin Blocking of viral uncoating
gives temporary protection against many viral Amantadine Influenzavirus A
diseases such at. measles, mumps and infectious HIV protease inhibitors
hepatitis. These are indicated only when Indinavir HIV-1, HIV-2
nonimmune individuals who are as special risk
Ritonavir HIV-1, HIV-2
are exposed to infection. Combined active and
passive immunization is an established method Saquinavir HIV-1, HIV-2
for the prevention of rabies. Reverse transcriptase inhibitors
Lamivudine HIV-1, HIV-2, HBV
Chemoprophylaxis and (dideoxythiacytidine)
chemotherapy of virus diseases Nevirapine HIV-1
Stavudine HIV-1, HIV-2
Because viruses are obligate intracellular parasites, (didehydrodexoythymidine)
antiviral agents must be capable of selectively
Zidovudine HIV-1, HIV-2, HBV
inhibiting viral functions without damaging the
(azidothymidine, AZT)
host, making the development of such drugs very
Zatcitabine HIV-1, HIV-2, HBV
difficult. Viral infection may be checked at the
(dideoxycytidine, ddC)
level of attachment, transcription of viral nucleic
acid, translation of viral mRNA, replication of viral Didanosine HIV-1, HIV-2
(dideoxyinosine, ddl)
nucleic acid and’ assembly and release of viral
progeny. Blocking of capping of mRNA

Available antiviral agents can be considered Ribavirin Respiratory synchtial virus,


Influenzaviruses A and B
under the following categories (Table 55.2). Lassa fever

1. Nucleoside Analogs 2. Protease Inhibitors


Examples of nucleoside analogs include acyclovir Saquinavir was the first protease inhibitor to be
(acycloguanosine), lamivudine (3TC), ribavirin, approved for treatment of HIV infection. Other
vidarabine (adenine arabinoside), and zidovudine protease inhibitors include indinavir and ritonavir.
(azi­dothymidine; AZT).
Azidothymidine (Zidovudine, AZT)
Acyclovir (Acycloguanosine) The widely publicized drug azidothymidine
Acyclovir (acycloguanosine) is acting against (zidovudine, AZT) used against HIV infection is
herpes viruses. The related drug ganciclovir is more a thymidine analogue. AZT is used widely in HIV
active against CMV. infection, but is toxic and costly.
A series of did eoxynucleosides (didanosine,
Amantadine and Rimantadine zalcitabine, stavudine and lamivudine) have been
These synthetic amines specifically inhibit synthesized and found to possess anti-HIV activity
influenza A viruses by block­ing viral uncoating. by blocking reverse transcriptase.
Amantadine (adamantanamine hydrochloride,
symmetrol) blocks host cell penetration by Interferons
influenza A virus but not B or C. A derivative Beneficial effect of interferons has been obtained
rimantadine is less toxic and equally effective. in persistent infections such as hepatitis B and C,
Chapter 55: Laboratory Diagnosis, Prophylaxis and Chemotherapy of Viral Diseases  | 401
laryngeal papilloma and against CMV infections 3. If Polymerase chain reaction is useful in the
in transplant recipients. High doses of interferon diagnosis of which of the following infec­tions?
lead to toxic effects. a. Hepatitis B virus
b. HIV
Key Points c. Human papilloma virus
d. All of the above
™™ Laboratory diagnosis of viral infections can be carried 4. Fluorescence microscopy can be used for the
out by the methods, such as I. Direct detection of laboratory diagnosis of:
virus; II. Virus isolation; III. Detection of viral protins; a. Herpes simplex encephalitis
IV. Detection of viral genetic material and serology b. Subacute sclerosing panencephalitis
™™ Immunoprophylaxis of viral diseases is by active and c. Rabies
passive immunization d. All of the above
™™ Viral vaccines are used for active immunization 5. Which of the following vaccines is/are live?
which may be may be live or killed a. Measles vaccine
™™ Passive immunization with human gamma globulin, b. Mumps vaccine
convalescent serum or specific immune globulin c. Rubella vaccine
gives temporary protection against many viral d. All of the above
diseases. 6. Antiviral drugs are available against all the
following viral infections except:
a. Herpes simplex virus
Important Questions b. Human immunodeficiency virus
c. Rubella vaccine
1. Discuss laboratory diagnosis of viral infections. d. Measles virus
2. Discuss viral vaccines. 7. All the following are the examples of protease
3. Write short notes on: inhibitors antiviral agents except:
a. Live viral vaccines a. Saquinavir
b. Acquired immunity in viral infections b. Amantadine
c. Antiviral therapies. c. Indinavir
d. Nelfinavir
Multiple choice questions (MCQs) 8. Acyclovir is used for treatment of which of the
following infections?
1. Electron microscopy can be used for the laboratory a. Herpes simplex b. Hepatitis B
diagnosis of: c. AIDS d. Influenza
a. Rotavirus infections 9. Amantadine is used for the treatment of:
b. Hepatitis A virus infections a. Varicella-zoster b. Hepatitis B
c. Adenovirus infections c. Influenza d. AIDS
d. All of the above 10. Which of the following drugs is/are effective against
2. Fluorescence microscopy can be used for the HIV?
laboratory diagnosis of: a. Zidovudine b. Ganciclor
a. Rabies c. Acyclovir d. All of the above
b. subacute sclerosing panencephalitis
c. Herpes simplex encephalitis Answers (MCQs)
d. all of the above 1. d; 2. d; 3. d; 4. d; 5. d; 6. d; 7. b; 8. a; 9. c; 10. a

www.ebook3000.com
56
Chapter

Bacteriophages

Learning Objectives

After reading and studying this chapter, you should ∙∙ Discuss lytic and lysogenic cycle of bacteriophage
be able to: ∙∙ Describe phage typing.
∙∙ Describe morphology of bacteriophage

Introduction 2. Tail: The tail is cylindrical and is composed of a


central hollow core or tube, a con­tractile sheath
Viruses that infect bacteria are called bacteriophages, surrounding the core and a terminal base plate
or simply phages. Phages can readily be isolated from which has attached to it prongs or tail fibers
a wide range of environments such as feces, sewage (usually six in number) or both (Fig. 56.1) that
and other natural sources of mixed bacterial growth. bind to specific receptor sites on the bacterial
surface.
Role of bacteriophages The type of nucleic acid present in the phage
1. They play an important role in the transmission particle varies, but no phage has more than one
of genetic information from one bacterium to type. Although double-stranded DNA is the most
another by the process of transduc­tion. common, single­stranded RNA or DNA is present
2. They also play a role in the evolution of in some phages.
bacterial types and in the transmission of some
virulence characters. Life cycle
3. Phages may indeed be effective in treating
bacterial infections, including those caused by Phages exhibit two different types of life cycle.
antibiotic-resistant bacteria. In the virulent or lytic cycle, intracellular mul­
tiplication of the phage culminates in the lysis
4. Phages have been used as cloning vectors in
of the host bacterium and the release of progeny
genetic manipulations.
virions. In the temperate or lysogenic cycle the
5. They may have a role in the control of bacterial
populations in natural waters.

Morphology
Certain bacteriophages that infect E. coli, called the
T even phages (T2, T4, T6), have been studied in
great detail and traditionally serve as the prototypes
in describing the properties of bacterio­phages.
Most phages are tadpole-shaped, possess a
hexagonal head and a cylindrical tail.
1. Head: The head consists of a tightly packed
core of nucleic acid (double stranded DNA) Fig. 56.1: Morphology of bacteriophage. A. hexagonal head,
surrounded by a protein coat or capsid. The B. DNA core, C. demarcation between head and tail, D. tail,
head of phage T4 has a diameter of 65 nm and E. base plate, F. tail fibers, G. prongs; right, process of injection
is 100 nm long. of phage DNA into host cell
Chapter 56: Bacteriophages  | 403
phage DNA becomes integrated with the bacterial protein capsid outside. The process of penetration
genome, repli­cating synchronously with it, causing resembles injection through a syringe. Following
no harm to the host cell (Fig. 56.2). adsorption, six tail pins make contact with the host
cell surface and firmly attach the phage plate to it.
1. Lytic Cycle The contractile tail sheath then contracts forcing
Replication of a virulent phage can be considered the hollow interior tail tube into the bacterial cell
in the following stages adsorption, pene­tration wall. The phage DNA then passes through the
and synthesis of phage components, assembly, hollow interior tail tube. The empty head and tail
maturation and release of progeny phage particles. of the phage remain outside the bacterium as the
shell or ‘ghost’ after penetration.
When bacteria are mixed with phage particles at
i. Adsorption
high multiplicity (that is very large number of phages
By random collision, phage particles attach to per bacterial cell), multiple holes are produced on
virus-specific receptors on the host cell by its tail. the cell with the consequent leakage of cell contents.
Adsorption is a specific process and depends on Bac­terial lysis occurs without viral multiplication.
the presence of complementary chemical groups This is known as ‘lysis from without’.
on the receptor sites on the bacterial surface and on
the terminal base plate of the phage. The infection iii. Synthesis of Phage Nucleic Acid and Proteins
of a bacterium by the naked phage nucleic acid is The synthesis of the phage components is ini­tiated
known as transfec­tion. immediately after penetration of the phage nucle­ic
acid. The first products to be synthesized (called
ii. Penetration early proteins) are the enzymes necessary for the
After adsorption, most phages inject their nucleic building of the complex molecules peculiar to the
acid into the bacterial cytoplasm and leave their phage. Subsequently, late proteins appear which

Fig. 56.2:  Lytic and lysogenic life of bacteriophage. 1. Adsorption, 2. Injection of phage DNA, 3. Circularization of phage
DNA, 4. Replication of phage DNA, 5. Production of phage components, 6. Assenbly of phage, 7. Release of progeny phage,
8. Integration of phage DNA with host chromosome, 9. Binary fission of lysogenic bacterium, 10. Daughter bacteria carrying
prophage, 11. Excision of prophage, 12. Same stage as 3 (1 to 3 injection; 4 to 7 lytic; 8 to 10 lysogenic cycle; 11 and 12 induction)

www.ebook3000.com
404  |  Section 4: Virology
in­clude the protein subunits of the phage head and bacillus and nontoxigenic strains can be made
tail. During this period, the synthesis of bacterial toxigenic by lysogenization.
protein, DNA and RNA ceases and the cell is forced ii. Clostridium botulinum types C and D produce
to make viral constituents. toxin only if these are infected with phage CE
b and DE b, respectively.
iv. Assembly and Maturation iii. A wide variety of temperate phages of Salmo­
Phage DNA, head protein and tail protein are nella can modify the antigenic properties of
syn­thesized separately in the bacterial cell. The somatic O antigen. When Salmonella is infected
DNA is condensed into a compact polyhedron by an epsilon phage, the structure of its outer
and ‘packaged’ into the head and, finally, the tail lipopolysaccharide layer may be modified.
structures are added. This assembly of the phage The prophage may become ‘excised’ from
components into the ma­t ure infective phage occasional cells during the multiplication of
particle is known as maturation. lysogenic bacteria. The excised prophage initiates
lytic replication and the daughter phage particles
v. Release are released, which infect other bacterial cells
Release of the mature progeny phages typically and render them lysogenic. This is known as
occurs by lysis of the bacterial cell. Phage enzymes act spontaneous induction of prophage. While this is
on the weakened cell wall causing it to burst or lyse a rare event, all lysogenic bacteria in a population
resulting in the release of mature daughter phages. can be induced to shift to the lytic cycle by
exposure to certain physical and chemical agents.
Such inducing agents include UV rays, hydrogen
Eclipse Phase
per­o xide and nitrogen mustard. A lysogenic
The interval between the entry of the phage nucle­ic bacterium is resistant to reinfection by the same
acid into the bacterial cell and the appearance of or related phages. This is known as super­infection
the first infectious intracellular phage particle is immunity.
known as the eclipse phase. The interval be­tween
the infection of a bacterial cell and the first release Significance of phages
of infectious phage particles is known as the latent
period (20–40 minutes). Immediately following the 1. Virulent Phage
latent peri­od, the number of phage particles released i. Phage Assay
increases for a few minutes till the maximum number
of daugh­ter phages is attained. This period, during When a phage is applied on the lawn culture of
which the number of infectious phages released a susceptible bacterium, areas of clearing occur
rises, is known as the rise period. The average yield after in­cubation. These zones of lysis are called
of progeny phag­es per infected bacterial cell is plaques. The size, shape and nature of plaques are
known as the burst size (100–300 phages). characteristic for different phages. Plaque assay
can be employed for titrating the number of viable
phages in a preparation.
2. Lysogenic Cycle
Unlike virulent phages which pro­duce lysis of the
host cell, temperate phages enter into a symbiotic ii. Phage Typing
relationship with their host cells without destroying Bacteriophage (phage) typing is based on the
them. Following entry into the host cell, the specificity of phage surface receptors for cell surface
temperate phage nucleic acid becomes integrated receptors. The limited host range of many phages
with the bacterial chromosome. The integrated enables them to be used as an epidemiological
phage nucleic acid is known as the prophage. marker to discriminate between bacterial strains
The prophage behaves like a segment of the that are bio­chemically or serologically indisting­
host chromosome and replicates synchronously uishable.
with it. This phenomenon is called lysogeny The strain to be typed is ino­culated on a plate
and a bacterium that carries a prophage within of nutrient agar to produce a lawn culture. After
its genome is called a lysogenic bacterium or drying, the phages are applied over marked squares
lysogen. in a fixed dose (routine test dose). The highest
The prophage confers certain new properties dilution of the phage preparation that just produces
on the lysogenic bacterium. This is known as lyso­ confluent lysis known as the routine test dose
genic or phage conversion. (RTD). After overnight incubation, the culture will
i. Toxin production in Corynebacterium be observed to be lysed by some phages but not by
diphthe­riae is determined by the presence in others. The phage type of the strain is expressed by
it of the pro­phage b. The elimination of this the designation of the phage/phages that lyse it.
prophage abolishes the toxigenicity of the Area of lysis caused by phage is known as plaque.
Chapter 56: Bacteriophages  | 405
The most important application of phage typing
(virulent) cycle, intracellular multiplication of the
is for intraspecies typing of bacteria, as in the phage phage ends in lysis of the host bacterium and release
typing of Salmonella Typhi and staphylococi. of progeny virions
Phages are available that lyse all members of ™™ In the lysogenic (temperate) cycle, phage DNA
a bacterial genus (for example genus-specific becomes incorporated within the bacterial genome
bacteri­ophage for Salmonella), all members of and then replicates synchronous with it, causing no
harm to the host cell
a species (for example specific bacteriophage
™™ Phage typing has been used for Staphylococcus
for B. anthracis), and all members of a biotype aureus, Vibrio cholerae, Salmonella, and many other
or subspecies (for example Mukerjee’s phage IV bacteria.
which lyses -all strains of classical V. cholerae but
not V. cholerae biotype EI Tor).
Important questions
1. Draw a labeled diagram of a T-even phage.
2. Temperate Phage 2. Discuss the life cycle of bacteriophages.
i. Transduction: Bacteriophages may act 3. Write short notes on:
as carriers of genes from one bacterium a. Lysogenic cycle of phages
to another. This is known as transduc­tion. b. Lysogenic conversion
Two types of transduc­tion are recognized: c. Phage typing
d. Significance of phages.
generalized transduction, in which any
portion of the donor DNA can be transferred,
and specialized transduction, in which only Multiple choice questions (Mcqs)
a specific set of genes can be carried to a 1. T-even bacteriophages possess
recipient cell. a. Single-stranded RNA
ii. Toxin production: Toxin production in b. Double-stranded RNA
Coryne­bacterium diphthe­riae and Clostridium c. Single-stranded DNA
botulinum are determined by genes carried in d. Double-stranded DNA
prohage DNA. 2. All the following statements for stage of adsorption
iii. Antigenic property: A wide variety of in lytic cycle of bacteriophage are true accept
a. It is a specific process
temperate phages of Salmo­nella can modify
b. It depends upon the susceptibility of the bacte-
the antigenic properties of somatic O antigen. rium to the specific phages
Such acquisition of new properties by c. It occurs by random collision
bacterial cells following phage infection is d. It is a very slow process
called “phage conversion”. 3. All the following are the examples of phage
iv. Cloning vector: Bacteriophages have been used conversions except
as cloning vectors in genetic manipulations. a. Phage-mediated conversion of somatic anti-
gens of Salmonella spp.
Key Points b. Toxigenicity of Corynebacterium diphtheriae
™™ Bacteriophages are bacterial viruses. They are c. Toxicity of Clostridium botulinum
associated with transmission of genetic material d. Toxigenicity of Vibrio cholerae
from one bacterium to another 4. Which of the following bacteria can be typed by
™™ Morphology: Most phages are tadpole-shaped, phage typing method?
possess a hexagonal head and a cylindrical tail. Most a. Staph. aureus
of the phages usually consist of single, linear, and b. Salmonella typhi
double­stranded DNA molecule genome. The phages c. Vibrio cholerae
show high host specificity d. All of the above
™™ Life cycle: Phages exhibit two different types of
life cycle: lytic cycle and lysogenic cycle. In the lytic Answers (MCQs)
1. d; 2. d; 3. d; 4. d.

www.ebook3000.com
57
Chapter

Poxviruses

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe molluscum contagiosum.
be able to:
∙∙ Describe structure of vaccinia virus

Introduction Table 57.1  Poxviruses that infect humans


Poxviruses are the largest and most complex of Genus Virus Primary Clinical
host(s) features
viruses that infect verte­brates, large enough to be
in humans
seen under the light micro­scope.
Orthopoxvirus Variola Man Smallpox
Smallpox, the great success story in the fight
against infectious disease and one that provides Vaccinia Man Vesicular
many valuable lessons. This epic provides at least vaccination
lesion
three ‘firsts’: the first vaccine, the first disease to be
totally eradicated by immuniza­tion, and the first Cowpox Cattle, Lesions on
virus infection against which chemotherapy was cats, hands
rodents
clinically effective.
Monkeypox Monkeys, Resembles
squirrels smallpox
Classification
Parapoxvirus Pseudocowpox Cattle Localized
The family Poxviridae contains two subfamilies: the nodular
Chordo­poxvirinae, the poxviruses of vertebrates, lesions
(‘milkers’
and Entomopoxvirinae, the poxviruses of insects.
nodules’)
Chordopoxvirinae are placed in eight genera-
Orthopoxvirus, Para poxvirus, Molluscipoxvirus, Yatapoxvirus Orf Sheep, Localized
Yatapoxvirus, Capri poxvirus, Leporipoxvirus, goats vesiculo.
Granulo­
Avipoxvirus (fowlipox, pigeonpox, canarypox, matous
turkeypox) and Sui poxvirus. Poxviruses causing lesions
diseases are enumerated in Table 57.1.
Tanapox Monkeys Vesicular skin
lesions and
Morphology febrile illness
Yabapox Monkeys Human
These are the largest viruses of all. The orthopox­ infections
viruses are brick-shaped. They measure about very rare and
230 × 270 nm and when suit­ably stained can just accidental;
be seen with an ordinary light microscope. The localized skin
poxviruses is referred to as complex. Inside there tumors
is a core structure shaped like a dumb­bell, and two Molluscipoxvirus Molluscum Man Multiple
accompanying lateral bodies, so named after their contagiosum small
location in the virion (Fig. 57.1). skin nodules
Chapter 57: Poxviruses  | 407

A B
Fig. 57.1: Structure of vaccinia virus. The nucleic acid is Fig. 57.2: Variola and vaccinia pocks on cam. A. Variola,
contained within a dumb-bell shaped core. Fitting into the showing small, uniform pocks; B. Vaccinia, showing large,
concavities of the core are two lateral bodies. The virion is irregular pocks
enclosed within a protein shell which has an irregular surface

Physical and Chemical Properties Variola and Vaccinia Viruses


Poxviruses survive for years in the cold or when Variola viruses: The variola virus is the causative
freeze dried. They are susceptible to ultraviolet agent of smallpox. Variola has a narrow host range
light and other irradiations. They are resistant (only humans and monkeys). Smallpox used to
to 50% glycerol and 1% phenol but are readily occur in two distinct clinical varieties.
inactivated by formalin and oxidizing disinfectants. a. Variola major or classical smallpox: The
florid, highly fatal disease typically seen in Asia.
Antigenic Structure b. Variola minor or alastrim: The mild, nonfatal
disease (alastrim) typically seen in Latin
All pox viruses share a common nucleoprotein (NP) America.
antigen. Some twenty different antigens have been Variola major and mi­nor was antigenically
identified by immunodiffusion. These include the LS identical but they differed in certain biological
antigens (a complex of two antigens, the heat labile characteristics. They were stable variants as
L and the heat stable S anti­gens), agglutinogen, and the disease produced by each always bred true.
hemagglutinin. Alastrim did not lead to smallpox and vice versa
Vaccinia virus: Vaccinia virus, the agent used
Cultivation and Host Range for smallpox vaccination, is a distinct species
A. Chick Embryo of Ortho­p oxvirus. At some time after Jen­n er’s
Virus isolation is carried out by inoculation of original use of “cowpox” virus, the vaccine virus
vesic­ular fluid onto the chorioallantoic membrane became “vaccinia virus”. Vaccinia virus is unique
(CAM) of chick embryos. Both vaccinia and variola in that it is an ‘artificial virus’ and does not occur
viruses grow on the CAM of 11–13-day-old chick in nature as such. It may be the prod­uct of genetic
embryo producing pocks in 48­–72 hours. Variola recombination, a new species derived from cowpox
pocks are small, shiny, white,.convex, non- virus or variola virus by serial passage, or the
necrotic, non-hemorrhagic lesions. Vaccin­ia pocks descendant of a now extinct viral genus.
are larger, irregular, flat, greyish, necrotic lesions, Variola has a narrow host range (only humans
some of which are hemorrhagic (Fig. 57.2). and monkeys), whereas vaccinia has a broad
host range that includes rabbits and mice. It
is safer to work with vaccinia virus. For the
B. Tissue Culture
development of recom­binant vaccines, vaccinia
Variola and vaccinia viruses can be grown in virus is being em­ployed as a vector. Many genes
tissue cultures of monkey kidney, HeLa and chick have been inserted, including those coding for
embryo cells. Cytopathic effects are produced by the antigens of hepatitis B virus, HIV, rabies and
vaccinia in 24–48 hours and more slowly by vari­ola. for pharmaco­logically important products such
Eosinophilic inclusion bodies—Guarnieri bodies— as neuropeptides.
can be demonstrated in stained preparations.
Vaccinia virus produces plaques in chick embryo Control of Smallpox
tissue cultures but not variola virus. The story of Edward Jenner’s discovery in 1796 that
inocula­tion with cowpox would prevent smallpox
C. Animals is well known. To Jenner, however, goes the credit
Monkeys, calves, sheep and rabbits can be infected for showing that, following the inoculation of young
by scarification leading to vesicular lesions. James Phipps with cowpox virus on May 14, 1796,

www.ebook3000.com
408  |  Section 4: Virology
deliberate inoculation with smallpox material 7. Molluscum Contagiosum
failed to induce the disease. Molluscum contagiosum is a benign epidermal tumor
For thousands of years, smallpox raged as a that occurs only in humans. The causative agent is
scourge of mankind, causing death and disfigure­ molluscum contagiosum virus. It is a contangious
ment. The global eradication of smallpox, achieved disease. The lesions of this disease are small, pink,
after 10 years of concerted campaigns under the wart-like tumors on the face, arms, back, and buttocks
auspices of the WHO, has been a most impres­ are more frequent in children than in adults. It is
sive medical achievement. Naturally occurring spread by direct contact [e.g. sexual contact, wrestling
smallpox came to an end in 1977. On May 8, or fomites (e.g. towels)]. The disease is more common
1980, the WHO formally announced the global in children than adults but its incidence is increasing
eradication of smallpox. in sexually active individuals.
The diagnosis of molluscum contagiosum can
OTHER POXVIRUS DISEASES usu­ally be made clinically. Sections of skin lesions
show large, eosinophilic intracytoplasmic inclusions,
1. Cowpox which displace the nuclei to the margin, known as
Cowpox is, a zoonosis. Cowpox lesions are seen on molluscum bodies under light microscope. It can
the udder and teats of cows and may be transmitted be detected under electron microscope and PCR
to humans during milking. The lesions in humans can detect viral DNA sequences. The virus cannot
usually appear on the hands or fingers. The natural be grown in animals or tissue cultures.
reservoir of cowpox seems to be a rodent.
Key Points
2. Monkeypox ™™ The family Poxviridae is a large family of ovoid or
brick-shaped viruses, and large enough to be just
This orthopoxvirus zoonosis causes an illness in visible under a light microscope
humans very similar to smallpox, squirrels seem to ™™ The genus Orthopoxvirus includes the viruses
be the main reservoir of infection, and the infection causing cowpox, vaccinia and variola. The variola
virus is an orthopoxvirus and the cause of smallpox
is seen mainly in chil­dren who may acquire it from
™™ They can grow in chorioallantoic membrane CAM)
playing with captive animals. of chick embryos and tissue culture
™™ Smallpox was the first viral disease to be eradicated
3. Buffalopox from the world
™™ Molluscum contagiosum: The virus causing mollus­
Buffalopox was identified in cattle in India in 1934 cum contagiosum is a poxvirus. The virus is spread by
and was considered an outbreak of vaccinia in close contact, often through sexual contact. It causes
them. Epizootics had occurred in buffaloes and small, pink, papular, pearl-like benign tumors of the
lesions had been observed on the hands of persons skin or mucous membranes.
in contact with infected animals.
Important question
4. Milker’s Node Write short notes on:
a. Variola virus. b. Vaccinia virus
Milker’s node (paravaccinia) is a trivial occupa­ c. Molluscum contagiosum
tional disease that humans get by milking infected
cows. The lesions are small ulcerating nodules. Multiple choice questions (mcqs)
1. All the statements are true for vaccinia virus except:
5. Orf (Contagious Pustular Dermatitis or a. It is an artificial virus
Sore Mouth) b. It is used for preparation of smallpox vaccine
c. It produces small, shiny, white, convex colonies
Orf is a disease of sheep and goats transmitted to on the chorioallantoic membrane
human beings by contact. It is an occupational d. It can be cultured in vitro on chorioallantoic
disease of sheep handlers. In humans, the disease membrane of ape
occurs as a single papulovesicular lesion with a 2. The last case of naturally occurring smallpox was
central ulcer usually on the hand, forearm, or face. detected in:
a. India in 1980
b. Bangladesh in 1979
6. Tanapox c. England in 1978
This virus takes its name from the Tana River in d. Somalia in 1977
Kenya, where it was first diagnosed. It is prevalent 3. What is/are the route/s of transmission of Molluscum
in monkeys and appears to be spread by insect contagiosum virus?
bites. Monkeys are the only animals susceptible. a. Direct contact. b. Sexual contact.
c. None of the above. d. Both of the above
There is usually only one vesicular lesion but its
appear­ance is preceded by fever and quite severe Answers (MCQs)
malaise. Recovery is uneventful. 1. c; 2. d; 3. d
58
Chapter

Herpesviruses

Learning Objectives

After reading and studying this chapter, you should ∙∙ D


 escribe the following: Varicella-zoster virus;
be able to: cytomegalovirus
∙∙ Describe morphology of herpesvirus ∙∙ Discuss clinical manifestations of Epstein-Barr virus
∙∙ Classify of human herpesviruses (EBV)
∙∙ Describe infections caused by herpes simplex virus ∙∙ Describe Paul-Bunnell test.
type 1 and herpes simplex virus type 2

Introduction Properties of the Viruses


The herpesvirus family contains over a hundred There are two distinct herpes simplex viruses:
spe­c ies of enveloped DNA viruses that affect type 1 and type 2 (HSV-1, HSV-2) (Table 58.1).
humans and animals. The outstanding property They differ in their mode of transmission. HSV
of herpesviruses is their ability to establish latent type 1 (Hu­man herpes virus type 1 or HHV type
infections, lifelong persistent infections in their 1) is usually isolated from lesions in and around
hosts and to undergo periodic reactivation. the mouth and is transmitted by direct contact
or droplet spread from cases or carriers. HSV
type 2 (HHV type 2) is transmitted sexually or from
Structure
a maternal genital infection to a new­born.
The herpesvirus capsid is icosahedral, composed of In­tracerebral inoculation in rabbits and mice
162 capsomers, and enclosing the core containing leads to encephalitis, and corneal scarification
the linear double stranded DNA genome. The produces keratoconjunctivitis in rabbits. The virus
nucleo­capsid is surrounded by the lipid envelope. grows in a variety of primary and continuous cell
The envelope carries surface spikes, about 8 nm long. cultures (monkey or rabbit kidney, human amnion,
Between the envelope and the capsid is an amor­ HeLa) producing cytopathic changes, well defined
phous structure called the tegument, containing foci with heaped up cells and syncytial or giant cell
sever­al proteins (Fig. 58.1). Herpesviruses replicate formation. On chick embryo CAM, small (diameter
in the host cell nucleus. They form Cowdry type A less than 0.5 mm) white shiny non-necrotic pocks
intranuclear (Lipschutz) inclusion bodies. are produced. The two types of the virus cross react

Classification
The family Herpesviridae is divided into three
subfamilies based on biological, physical and
genetic properties (Table 58.1).
Eight different types of herpesviruses are known
whose primary hosts are human (Table 58.1).

Herpes Simplex Virus (HSV)


Herpes simplex virus (HSV) was the first human
herpesvirus to be recognized. Fig. 58.1: Typical structure of the herpesviruses

www.ebook3000.com
410  |  Section 4: Virology
Table 58.1:  Classification of human herpesviruses
Subfamily Virus Primary target cell Site of latency Mode of spread
Alphaherpesvirinae
Human herpesvirus 1 Herpes simplex type 1 Mucoepithelial cells Neuron Close contact
Human herpesvirus 2 Herpes simples type 2 Mucoepithelial cells Neuron Close contact (sexually
transmitted disease)
Human herpesvirus 3 Varicella-zoster virus Mucoepithelial cells Neuron Respiratory and close
contact
Gammaherpesvirinae
Human herpesvirus 4 Epstein-Barr virus B cells and epithelial cells B cell Saliva (kissing disease)
Human herpesvirus 8 Kaposi’s sarcoma- Lymphocyte and ? Close contatct (sexual),
related virus other cells saliva?
Betaherpesvirinae
Human herpesvirus 5 Cytomegalovirus Monocyte, lymphocyte, Monocyte, Close contact,
and epithelial cells lymphocyte, transfusions, tissue
and ? transplant, and
congenital
Human herpesvirus 6 Herpes lymphotropic T cells and ? T cells and ? Respiratory and close
virus contact?
Human herpesvirus 7 Human herpesvirus 7 T cells and ? T cells and ? ?

serologically. They can be differentiated by the Once HSV has established itself in a ganglion in
following features: vivo, reactivations are liable to take place at inter­
1. Antigenic differences can be made out using vals of weeks or months thereafter trig­gered by a
type specific monoclonal antibodies. variety of stimuli. The virus follows axons back to
2. On chick embryo CAM type 2 strains form the peripheral site, and replication proceeds at the
larger pocks resembling variola. skin or mucous membranes.
3. Types 2 strains replicate well in chick embryo HSV-1 is usually associated with infections
fibroblast cells, while type 1 strains do so above the waist, and HSV-2 with infections below
poorly. the waist, consistent with the means of spread for
4. The infectivity of type 2 is more temperature these viruses.
sensitive than that of type 1.
5. Type 2 strains are more neurovirulent in labora­ Clinical Features
tory animals than type 1. 1. Cutaneous infections: (i) The most common
6. Type 2 strains are more resistant to antiviral site is the face—on the cheeks, chin, around
agents like IUDR and cytarab­ine in culture. the mouth or on the forepead; (ii) Napkin
7. Restriction endonuclease analysis of viral DNA rash; (iii) ‘Fever blister’ or herpis febrilis. In
enables differentiation between the two types some sensitive persons, very minor stimuli, like
as well as between strains within the same type. common cold, exposure to sun or even mental
strain or menses may bring on such reactivation;
Pathogenesis (iii) Herpetic whitlow- an infection of the
finger; (iv) Herpes gladiatorum an infection
The mechanisms involved in the pathogenesis of of the body; (v) Eczema herpeticum.
HSV-1 and HSV-2 are very similar. Both viruses 2. Oral infection: Acute gingivostomatitis,
initially infect and replicate in mucoepithelial cells herpetic stomatitis, pharyngitis, tonsillitis and
and then establish latent infection of the innervating localized lymphadenopathy may occur.
neu­rons. Skin and mucous membranes are the 3. Ophthalmic: Severe keratoconjunctivitis,
portals of entry in which the virus also multiplies, follicular conjunctivitis with vesicle formation
causing lysis of cells and formation of vesicles. on the lids, dendritic keratitis or corneal
Soon after replication is under way in the skin ulcers or as vesicles on the eyelids, corneal
or a mucous membrane, virions travel to the root scarring and impairment of vision.
ganglia via the sensory nerves supplying the area. 4. Nervous system: (i) HSV encephalitis; (ii)
Primary infections of the orofacial and genital Sporadic encephalitis; (iii) HSV meningitis;
areas involve respectively the trigeminal and (iv) Sacral autonomic dysfunction; (v) rarely
lumbosacral dorsal root ganglia. The virus then transverse myelitis or the Guillain–Barre
becomes latent in the ganglia. syndrome and Bell’s palsy.
Chapter 58: Herpesviruses  | 411
5. Visceral: (i) HSV esophagitis; (ii) tracheo­ be used. Typical cytopathic changes may appear
bronchitis and pneumonitis; (iii) hepatitis; (iv) as early as in 24–48 hours but cultures should be
Erythema multiforme; (v) disseminated HSV observed for two weeks before being declared
infection. negative.
6. Genital infections: Genital disease is usually HSV isolates can be typed by biochemical,
caused by HSV-2. Genital herpetic ulcers are biologic, nucleic acid, or immunologic methods.
known to increase the risk of transmission of The restriction endonuclease cleavage patterns
infection with human immunodeiciecy virus of the DNA of HSV-1 and HSV-2 are unique allow
(HIV). unequivocal typing of the isolates. HSV type-
(i) In male patients: The lesions typically specific DNA probes, spe­cific DNA primers for PCR
develop on the glans or shaft of the penis and and antibodies are also available for detecting and
occasionally in the urethra; (ii) In female differentiating HSV-1 and HSV-2.
patients: The lesions may be seen on the vulva,
vagina, cervix, perianal area, or inner thigh; C. Serology
(iii) Inguinal lymphadenopathy: In patients
of both sexes; (iv) Herpetic proctitis: in Rise in titer of antibodies may be demonstrated by
homosexuals; (v) Association between HSV-2 ELISA, neutralization or complement fixation tests.
and carcinoma of the cervix uteri: There have
been several reports of an association between D. Polymerase Chain Reaction
HSV -2 and carcinoma of the cervix uteri but a
causal relationship has not been established. Polymerase chain reaction is useful for detecting
viral DNA in cerebrospinal fluid when herpetic
7. Neonatal Herpes: Transplacental infection with
HSV1 or 2 can lead to congenital malformations. infection of the CNS is suspected.
8. Infections in immunocompromised hosts.
Treatment
Laboratory Diagnosis Idoxuridine used topically in eye and skin infections
was one of the first clinically successful antiviral
1. Specimens
agents. Acyclovir and vi­darabine enabled the effective
2. Microscopy management of deep and systemic infections.
i. Tzanck smear: Characteristic cytopathologic Intravenous, oral, and topical preparations are
effects (CPEs) can be identified in a Tzanck available. Drug-resistant virus strains may emerge.
smear, Papanicolaou smear, or biopsy Early treatment with intravenous acyclovir has
specimen. CPEs include syncytia, “ballooning” improved the outcome of encephalitis.Valaciclovir
cytoplasm, and Cowdry type A intranu­clear and famciclovir are more effective oral agents.
inclusions. When resistance to these drugs develop, drugs like
The Tzanck smear is a rapid, fairly sen­sitive foscarnet which are independent of viral thy­midine
and inexpensive diagnostic method. Smears kinase action may be useful.
are prepared from the lesions, preferably
from the base of vesicles and stained with Herpes virus simiae: B virus
1% aqueous solution of tolu­idine blue ‘0’ for
15 seconds. Multinucleated giant cells with Herpes B virus of Old World monkeys is highly
faceted nuclei and homogeneously stained patho­genic for humans. This virus was isolated
‘ground glass’ chromatin (Tzanck cells) by Sabin and Wright (1934) from the brain of a
constitute a positive smears. laboratory worker who developed fatal ascending
myelitis after being bitten by an appar­ently healthy
ii. Electron mi­croscopy: The virus par­ticle may
monkey. It came to be known as the ‘B’ virus from
also be demonstrated under the electron
the initials of this patient.
mi­croscope.
The virus is transmitted to humans by monkey
iii. Fluoroscent an­t ibody technique: The
bites or saliva or even by tissues and cells widely
fluorescent antibody test on brain biopsy
used in virology labo­ratories.
specimens provides reliable and speedy
diagnosis in encephalitis. Virus isolation or serologic tests can be used to
establish the diagnosis of B-virus infections.
B. Virus Isolation
Varicella-zoster virus
Primary human embryonic kidney, human amnion
and many other cells are susceptible, but human Varicella-zoster virus (VZV) causes chickenpox
diploid fibroblasts are preferred. Specimen such (varicella) and, with recur­rence, causes herpes
as vesicle fluid, spinal fluid, saliva and swab may zoster, or shingles. Vari­cella (chickenpox) and

www.ebook3000.com
412  |  Section 4: Virology
herpes zoster are different manifestations of the residual immunity. It is believed that years after the
same virus infection. Thus, chickenpox is ‘caught’ initial infection, when the immunity has waned,
but not zoster. the virus may be reactivated, and triggered by
some precipitating stimulus, travel along the sen­
Properties of the Virus sory nerve to produce zoster lesions on the area of
Varicella-zoster virus is morphologically identical the skin or mucosa supplied by it. It usually starts
to her­pes simplex virus. It can be grown in cultures with severe pain in the area of skin or mucosa
of human fibroblasts, human amni­on or HeLa cells. supplied by one or more groups of sensory nerves
Cytopathic changes are more focal and spread and ganglia. The most common sites are the areas
much more slowly than those induced by HSV. Only innervated by spinal cord segments D3 to L2 and
one antigenic type of VZV is known. the trigeminal nerve, particularly, its ophthalmic
branch. Within a few days after onset, a crop of
vesicles appears over the skin supplied by the
Varicella (Chick­enpox) affected nerves.
Varicella (chick­enpox) is one of the mildest highly
communicable and most common of childhood Complications
infections. 1. Postherpetic pain: In the affected area is
It is usually a mild disease of childhood and is frequent, particularly in the elderly.
normally symptomatic, although asymptomatic 2. Ophthalmic zoster.
infection may occur. The portal of entry of the 3. Generalized zoster
virus is the respiratory tract or conjunctiva. After an 4. Ramsay Hunt syndrome: It is a rare form of
incubation period of about two weeks (7–23 days) zoster affecting the facial nerve, with eruption
the lesions begin to ap­pear. on the tympanic membrane and external
In children, there is little prodromal illness and auditory canal, and often a facial palsy.
the disease is first noticed when skin lesions ap­pear.
Initially macular, the rash rapidly evolves through
papules to the characteristic clear vesicles. The
Immunity
rash is characteristically cen­tripetal in distribu­tion, Previous infection with varicella is believed to
affecting mainly the trunk, and is very superficial confer lifelong immunity to varicella.
without in­volving the deeper layers of the skin. The The development of varicella-zoster virus-
rash appears in successive crops, so that all stages specific cell-mediated immunity is important
of the eruption can be seen at the same time. It in recovery from both varicella and zoster.
matures very quickly, beginning to crust within 48 Appearance of local interferon may also contribute
hours.Recovery is usually uneventful. to recovery.
Complications: Primary infection is usually more
severe in adults than in children. The rash may Laboratory Diagnosis
become hemorrhagic. Varicella pneumonia is Diagnosis is usually clinical.
more common in adults. A variety of organs may 1. Microscopy: Multinucleated giant cells and type
be affected with complications like myocarditis, A intranuclear in­clusion bodies may be seen
nephritis, acute cere­bellar ataxia, meningitis in smears prepared by scraping the base of the
and encephalitis. Secondary bacterial infections, early vesicles (Tzanck smear) and stained with
usually due to staphy­lococci or streptococci, may toluidine blue, Giemsa or Papanicolou stain.
occur. Reyes’ syndrome may follow varicella in Direct examination by electron microscopy will
some cases with a history of administration of reveal herpes particles.
salicylates. 2. Virus isolation: Virus isolation can be
attempted by inoculating human amnion,
Herpes zoster (Shingles, Zona) human fibroblast, HeLa or Vero cells.
3. Virus antigen: The virus antigen can be
The name is derived from Heroein, meaning to detected in scrapings from skin lesions by
creep; Zoster, meaning girdle. immunofluorescence, and in vesicle fluid by
While varicella is typically a disease of child­ counterimmunoelectrophoresis with zoster
hood, herpes zoster is one of old age. immune serum. ELISA and PCR tech­niques
are also in use.
Pathogenesis 4. Serological diagnosis: A rise in specific
Herpes zoster usually occurs in persons who antibody titer can be detected in the patient’s
had chickenpox several year earlier. The virus serum by various tests, including fluorescent
remaining latent in the sensory ganglia, may leak antibody, latex agglutination, and enzyme
out at times but is usually held in check by the immunoas­say.
Chapter 58: Herpesviruses  | 413
Prophylaxis and Treatment and the reticuloendothelial system. Clinical
features—include intrauterine growth retar­
Active Immunization
dation, jaundice, hepatosplenomegaly, throm­
A live-attenuated varicella vaccine was developed bocytopenia, microcephaly, chorio­retinitis and
by Takahashi in Japan in 1974 by attenuating a cerebral calcification resembling congenital
strain of varicella virus (Oka strain, so named after toxoplasmosis.
the patient) by serial passage in tissue culture.
Perinatal infection may be acquired from the
This vaccine, given by subcutaneous injection,
infected mother through genital secretions or
has been found to be immunogenic with good
breast milk.
antibody response but it was very labile and had
to be stored frozen. A modified lyophilized form of D. Postnatal infection: This can be acquired in
the vaccine is now available, which can be stored many ways-such as saliva, sexual transmission,
between 2°C and 8°C. It is recommended as a single blood transfusion and donated organs
subcutaneuos dose for childern 1–12 years old, and
for those older as 2 doses 6–10 weeks apart. It is safe Laboratory Diagnosis
and effective. It is not considered safe in pregnancy. A. Specimens: CMV can be isolated from the
urine, saliva, breast milk, semen, cervical
Passive Immunization secretions and blood leucocytes.
Varicella-zoster immunoglobulin (VZIG) prepared B. Demonsration of cyto­m egalic cell: The
from the patients convalescing from herpes zoster histologic hallmark of CMV infection is the
seems to be of some use in preventing or modifying cyto­megalic cell, which is an enlarged cell
severe disease in immunodeficient patients. that contains a dense, central, “owl’s-eye,”
basophilic intranuclear inclusion body. The
Laboratory Diagnosis inclusions are readily seen with Pa­panicolaou
or hematoxylin-eosin staining
Diagnosis is easily made clinical. Laboratory
C. lsolation of virus: Human cytomegaloviruses
diagnosis is as for chickenpox.
can be grown in human fibroblast cultures.
Cultures have to be incubated for prolonged
Treatment periods as the cytopathic effects (swollen
It is as for chickenpox. refractile cells with cytoplasmic granules) are
slow in appearance.
Cytomegalovirus D. DNA probes: DNA probes are used to directly
detect the CMV antigens in tissues or fluids.
Cytomegaloviruses (CMV), formerly known as
salivary gland viruses, are a group of ubiquitous E. Polymerase chain reaction (PCR): To directly
herpes­viruses of humans and animals. detect the genome in tissues or fluids.
F. Serology: IgM antibodies suggests a current
infection and can be detected in serum by
Pathogenesis
ELISA.
The congenital, oral, and sexual routes, blood
transfusion, and tissue transplantation are the major Treatment and Prevention
means by which CMV is transmitted. Congenitally
infected infants have viruria for upto 4–5 years. They Ganciclovir (dihydroxypropoxymethyl guanine)
are highly infectious in early infancy. and foscarnet (phosphonoformic acid) have been
approved for the treatment of CMV infections.
A. Normal hosts: Primary cytomegalovirus
infection of older children and adults is Prevention is indicated only in high risk
usually asymptomatic but occasionally causes cases. Screening of blood and organ donors and
a spontaneous infectious mononucleosis administration of CMV immunoglobulins have
syndrome. been employed in prevention.
B. Immunocompromised hosts: This occurs No vaccine is available.
in transplant recipients, cancer patients on
chemotherapy, and more particularly in the
Epstein–Barr virus
HIV infected. Epstein–Barr virus (EBV) is in some respects the
C. Congenital and perinatal infections: Intra­ most sinister herpesvirus, for its association with
uterine infection leads to fetal death or malignant disease is now well established. Only
cytomegalic inclusion disease of the newborn human and some subhuman primate B cells have
which is often fatal. Cytomegalic inclusion receptors for the virus. EBV infected B cells are
disease of newborns is characterized by transformed so that they become capable of con­
involvement of the central nervous system tinuous growth in vitro.

www.ebook3000.com
414  |  Section 4: Virology
Pathogenesis A. Infectious Mononudeosis (Glandular Fever)
Epstein–barr virus (EBV) is commonly transmitted This is an acute self-limited illness usually seen
by infected saliva and initiates infection in the in nonimmune young adults. The incubation
oropharynx. The virus enters the pharyngeal period is 4–8 weeks.Infectious mononucleosis is
epithe­lial cells through CR2 (or CD21) receptors. charac­terized by high fever, malaise, pharyngitis,
It multiplies locally, invades the bloodstream lymphade­nopathy (swollen glands), and, often,
and infects B lymphocytes in which two types hepatosplenomeg­aly. A mild transient rash may
of changes are produced. In most cases, the virus be present. Some patients treated with ampicillin
be­comes latent inside the lymphocytes, which may develop a maculopapular rash due to immune
become transformed or ‘immortalized so that complex reaction to the drug. In most patients
they become ca­pable of indefinite growth in vitro. the spleen is pal­pable and there is some liver
Lytic infection is a second type of effect, shown by dysfunction, occasionally with frank jaundice. The
a few infected B cells with cell death and release of typical illness is self-limited and lasts 2–4 weeks.
mature progency virions. Complications are rare but some are serious:
The classic lymphocytosis associated with i. Acute airway obstruction.
infectious mononucleosis results mainly from the ii. Splenic rupture.
activation and proliferation of T cells. These appear iii. Neurological complications include meningitis,
as atypical lymphocytes (also called Downey encephalitis and the Guillain-Barre syndrome.
cells).
Intermittent reactivation of the latent EB virus B. Oral Hairy leukoplakia
leads to clonal proliferation of infected B cells. In This lesion is a wart-like growth that develops
immunocompetent subjects, this is kept in check on the tongue in some HIV-infected persons and
by activated T cells. In the immunodeficient, B transplant patients. It is an epithelial focus of EBV
cell clones may replicate unchecked, resulting in replication.
lympho­mas. Hyperendemic malaria prevalent in
Africa is believed to be responsible for the immune C. Chronic Disease
impairment in children with Burkitt’ lymphoma. Epstein-barr virus (EBV) can cause cyclic recurrent
The frequency of lymphomas seen in many types of disease in some people. This disorder is different
immunodefi­ciencies, most typically in AIDS, may from chronic fatigue syn­drome, which has another
have a similar pathogenesis. etiology.
Nasopharyngeal carcinoma seen in men of
Chinese origin. Genetic and environmental factors D. Burkitt’s lymphoma
are said to be important in the and EB virus DNA
Epstein–barr virus (EBV) is associated with the
is regularly found in the tumour cells.
development of Burkitt’s lymphoma (a tumor of
the jaw in African children and young adults).
Epidemiology Most African tumors (>90%) contain EBV DNA
Epstein–Barr (EB) virus is ubiq­uitous in all human and express EBNA1 anti­gen. Malaria, a recognized
populations. Infection with EBV is transmitted by cofactor.
saliva, and requires intimate oral contact.
The source of infection is usually the saliva E. Nasopharyngeal Carcinoma
of in­fected persons who shed the virus in This cancer of epithelial cells is common in males
oropharyngeal secretions. Saliva sharing between of Chinese origin. It mainly affects people aged
adolescents and young adults often occurs during 20–50 years, males preponderating. EBV DNA is
kissing; thus, the nickname “kissing disease” for regularly found in nasopharyngeal carcinoma
infectious mononucleosis. Chil­dren can acquire (NPC) cells. Genetic and environmental factors
the virus at an early age by sharing contaminated are believed to be important in the development
drinking glasses. Infection may also follow blood of nasopharyngeal carcinoma.
or marrow transfusion but these are rare events.
F. Lymphoproliferative Diseases in
Clinical conditions
Immunodeficient Hosts
1. Infectious mononucleosis.
Immunodeficient patients are susceptible to EBV
2. EBV associated malignancies:
­induced lymphoproliferative diseases that may
a. Burkitt’s lymphoma. be fatal. AIDS patients are susceptible to EBV-
b. Lymphomas in immunodeficient persons. associated lymphomas and hairy oral leukoplakia
c. Nasopharyngeal carcinoma. of the tongue.
Chapter 58: Herpesviruses  | 415
Laboratory Diagnosis IgG anti-VCA antibody indicates past or recent
infection and persists throughout life.
1. Differential White Cell Count
Early antigen (EA) antibodies are often found in
Blood examination during the initial phase may patients with Burkitt’s lymphoma or nasopharyngeal
show leucopenia. Later there is a prominent carcinoma.
leu­cocytosis with the appearance of abnormal Antibodies to the EB nuclear antigen (EBNA)
mononuclear cells. These atypical mononuclear reveal past infection with EBV, though detection
cells are lymphoblasts derived from T cells reactive of a rise in anti-EBNA antibody would suggest a
to the virus infection. The blood picture may primary infection.
sometimes resemble lymphocytic leukemia.
4. Antigen Detection
2. Paul–Bunnell Test
Epstein-barr virus (EBV) antigen can be detected
B cells transformed by EBV undergo polyclonal by immunofluorescence using monodonal
expansion. These anti­b odies include an IgM antibodies.
heterophile antibody. Agglutination of horse or
sheep red cells by serum absorbed to exclude a 5. Nucleic Acid Hybridization
natural antibody is the basis of this test. It is the most sensitive means of detecting EBV in
patient materials.
Procedure
Inactivated serum. 56°C for 30 minutes in doubling 6. Virus Isolation
dilutions is mixed with equal volumes of a 1% Epstein-barr virus (EBV) can be isolated from
suspension of sheep erythrocytes. After incubation saliva, peripheral blood, or lymphoid tissue by
at 37°C for four hours the tubes are examined for immortalization of normal human lymphocytes,
agglutination. An aggluti­nation titer of 100 or above usually obtained from umbilical cord blood.
is suggestive of infectious mononucleosis.
7. Polymerase Chain Reaction.
Confirmation
For confirmation, differential absorp­t ion of Human Herpesviruses 6 (HHV6)
agglutinins with guineapig kidney and ox red cells Human herpesviruses 6 was first isolated from the
is necessary. The Forssman antibody induced blood of patients with AIDS and grown in T-cell
by injection of horse serum is removed by cultures. HHV6 is lymphotropic and ubiquitous. It
treatment with guineapig kidney and ox red cells. is present in the saliva of most adults and is spread
Normally occur­ring agglutinins are removed by by oral secretions. Two variants are recognized,
guineapig kidney, but not by ox red cells. Infectious A and B. Variant B is the cause of the mild but
mononucleosis anti­body is removed by ox red cells common childhood illness ‘exanthem subitum’
but not guinea pig kid­ney (Table 58.2). (roseola infantum or ‘sixth disease’). In older age
groups, it has been associated with infectious
3. Epstein–Barr Virus-specific Antibodies mononucleosis syndrome, focal en­cephalitis and,
Tests are also available for the demonstration of in the immunodeficient, with pneumonia and
specific EB virus antibodies. Immunofluorescence disseminated disease.
and ELISA are commonly employed. Laboratory dignosis: HHV6 can be isolated
from peripheral blood mononuclear cells in early
The IgM antibody to VCA (virus capsid antigen)
febrile stage of the illness by co-cultivation with
appears soon after primary infection and disappears
lymphocytes.
in 1–2 weeks and indicative of current infection. The
Virus antigen-can be detected by immuno­
fluroscence using monoclonal antibodies. ELISA
Table 58.2:  Differential absorption test for Paul- is used for detecting both antigen and antibidies
Bunnell antibody in patient serum.
Result of absorption Ox red cells
on guinea pig kidney Human Herpesvirus 7 (HHV7)
Normal serum Absorbed Absorbed
Like HHV6, HHV7 also appears to be widely distri­
Antibody after Absorbed Not absorbed buted and transmitted through saliva. However,
serum therapy
HHV7 remains an orphan virus with no disease
Infectious Not absorbed Absorbed association. It shares with HIV the same CD4
mononucleosis
receptor on T cells.

www.ebook3000.com
416  |  Section 4: Virology
It may cause an illness resem­bling infectious
™™ Laboratory diagnosis—Atypical lymphocytes are
mononucleosis, some cases of exanthem subitum. probably the earliest de­tectable indication of an
EBV infection
Human Herpesvirus 8 (HHV8) ™™ Paul-Bunnell test is most frequently used test
to detect hetero­p hile antibodies in infectious
A new herpesvirus, also called Kaposi’s sarcoma- mononucleosis patients
associ­a ted herpesvirus (KSHV) is the cause Human Herpesvirus 8 (HHV8)
of Kaposi’s sarcomas, vascular tumors of mixed ™™ It is the cause of Kaposi’s sarcomas, vascular tumors
cellular composition, and is involved in the of mixed cellular composition, body cavity-based
lymphomas occurring in AIDS patients and of
pathogenesis of body cavity-based lymphomas multicentric Castle­man’s disease.
occurring in AIDS patients and of multicentric
Castle­man’s disease.
It appears to be sexually transmitted among Important questions
men who have sex with men. Infections are
common in Africa with infections acquired early in 1. Name various viruses of the family Herpesviridae.
life by nonsexual routes, possibly through contact Discuss the various infections caused by her­pes
with oral secre­tions. simplex virus types 1 and 2.
Diagnosis depends mainly on detection of viral 2. Classify human herpesviruses. Discuss briefly
DNA by PCR. their pathogenesis and laboratory diagnosis.
3. Describe the lesions caused by herpes simplex
virus and their laboratory diagnosis.
Key Points
4. Write short notes on:
™™ Herpesviruses are DNA viruses a. Varicella-zoster virus
™™ Human herpesviruses include human herpesvirus b. Varicella or chickenpox
1 (HV1) to human herpesvirus 8 (HV-8). HHV 3, HHV c. Cytomegalovirus
4 and HHV 5 are varicella-zoster virus, Epstein-Barr d. Epstein-Barr virus (or) EB virus
(EB) and cytomegalovirus (CMV) respectively. Herpes e. Infectious mononucleosis
simplex virus type 1 and 2 are designated as HHV 1 f. Paul-Bunnel test
and HHV 2
g. Human herpesvirus 6 (HHV6)
Clinical syndromes
™™ HSV-l is generally transmitted orally; HSV-2 is gener­
ally transmitted sexually Multiple choice questions (mcqs)
™™ HSV-1 causes acute herpetic gingivostomatitis, acute
herpetic pharyngotonsillitis, herpes labial is, herpes 1. Which of the following infection/s is/are caused by
encephalitis, eczema herpeticum, and herpetic herpes simplex virus type I?
whitlow. HSV-2 causes genital herpes, neonatal a. Acute gingivostomatitis
infection, and aseptic meningitis b. Keratoconjunctivitis
Laboratory diagnosis: (A) Direct microscopic examin­ c. Encephalitis
ation of cells from base of lesion; (B) Cell culture; d. All of the above
(C) Assay of tissue biopsy, smear, or vesicular fluid 2. All the following infections are caused by HSV-2
for HSV antigen; (D) HSV type distinction (HSV-l vs. except:
HSV-2) type a. Neonatal infection
™™ Varicella Zoster Virus (VZV)—causes chickenpox b. Aseptic meningitis
(varicella) and herpes zoster or shingles, two distinct
c. Herpetic whitlow
clinical entities in humans
d. Genital herpes
™™ Virus causes lifelong infection
3. All the following infections are caused by HSV-2
Cytomegalovirus
except:
™™ Virus is transmitted orally and sexually, in blood
a. Neonatal infection
trans­fusions, in tissue transplants, in utero, at birth,
and by nursing b. Aseptic meningitis
™™ Clinical syndromes—Congenital CMV infection,
c. Herpetic whitlow
acquired CMV infection, CMV infec ­t ion in d. Genital herpes
immunocompromised patients, and CMV infection 4. Ramsay–Hunt syndrome can be caused by:
in immunocompetent adult hosts. CMV generally a. Herpes simplex virus
causes subclinical infection b. Herpes-zoster virus
Epstein–Barr Virus c. Cytomegalovirus
™™ EBV causes heterophile antibody-positive infec­tious d. Epstein–Barr virus
mononucleosis and has been causally associated
5. Shingles is caused by:
with Burkitt’s lymphoma, Hodg­k in’s disease, and
a. Varicella–zoster virus
nasopharyngeal carcinoma
b. Cytomegalovirus
™™ Transmission occurs via saliva, close oral contact
(“kiss­ing disease”), or sharing of items c. Epstein–Barr virus
d. Herpes simplex virus type 1
Chapter 58: Herpesviruses  | 417
6. Owl’s eye is the characteristic feature of the cell 8. Paul–Bunnell test is used for serodiagnosis of:
infected by: a. Infectious mononucleosis
a. Herpes simplex virus b. Genital herpes
b. Epstein-Barr virus c. Neonatal infection
d. Aseptic meningitis
c. Cytomegalovirus
9. Which of the following viruses is associated in
d. Human herpesvirus 8
causation of Kaposi’s sarcoma?
7. Which of the following malignancies is/are associated a. Herpes simplex virus
with Epstein–Barr virus? b. Herpesvirus simiae
a. Burkitt’s lymphoma c. Human herpesvirus 8
b. Nasopharyngeal carcinoma d. Human herpesvirus 6
c. B cell lymphoma Answers (MCQs)
d. All of the above 1. d; 2. c; 3. c; 4. b; 5. a; 6. c; 7. d; 8. a; 9. c.

www.ebook3000.com
59
Chapter

Adenoviruses

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe diseases associated with adenovirus
be able to: ∙∙ Describe adenovirus-associated viruses (AAV).
∙∙ Describe morphology of adenovirus

Introduction capsomers which consists of hexons, while the 12


capsomers at the vertices have five neighbors and
Adenoviruses are a group of medium sized, are called pentons.
nonenvel­oped, double stranded DNA viruses that
Each penton unit consists of a penton base
share a com­mon complement fixing antigen. The
anchored in the capsid and a projection or fiber
name “adenovirus” (adeno, from adenoid) reflects
consisting of a rod like portion with a knob
the recovery of the initial isolate from explants of
attached at the distal end. Thus, the virion has the
human adenoids. All human serotypes are included
appearance of a space vehicle. The DNA is linear
in a single genus within the family Adeno­viridae.
and double­stranded.
Classification
Resistance
The family Adenoviridae comprises two genera.
A. Mastadenovirus: Infects mammals. Human Adnoviruses are relatively stable, remain­ing
adenoviruses are further subdivided into viable for about a week at 37°C. They are readily
six species A–F (also called subgroups or inactivated at 50°C. They resist ether and bile salts.
subgenera) (Table 59.1).
B. Aviadenovirus: Infects birds. Pathogenesis
Adenoviruses infect and replicate in epithelial cells
Morphology of the respiratory tract, eye, gastrointestinal tract,
Adenoviruses are 70–90 nm in diameter and urinary blad­der, and liver.
display icosahedral symmetry (Fig. 59.1). There Several distinct clinical syndromes are associ­
is no envelope. The capsid is composed of 252 ated with adenovirus infection (Table 59.2)
Table 59.1:  Classification of human adenoviruses
Group Serotype (Species) Total Hemagglutination pattern with Oncogenicity in
(Subgenus) red cells of newborn hamsters
A 12, 18, 21 3 Rat (Partial) High
B 3, 7, 11, 14, 16, 21, 34, 35 8 Monkey (Complete) Low
C 1, 2, 5, 6 4 Rat (Partial) None
D 8–10, 13, 15, 17, 19, 20, 29 Rat (Complete ) None
22–30, 32, 33, 36–39, 42–47
E 4 1 Rat (Partial) None
F 40, 41 2 Rat (Partial) None
Total 47
Chapter 59: Adenoviruses  | 419
B. Eye Infections
1. Phar yngoconjunctival fever : Phar yn­
goconjunctival fever tends to occur in outbreaks,
such as at children’s summer camps (swimming
pool conjunc­tivitis), and is associated with
serotypes 3, 7 and 14.
2. Epidemic keratoconjunctivitis (EKC): This
is a seri­ous condition which may appear as an
epidemics, usually caused by type 8 and less
often by types 19 and 37. This disease occurs
mainly in adults and is highly contagious.
3. Acute follicular conjunctivitis: Types 3, 4 and
11 are commonly re­sponsible. Adenoviral and
chlamydial conjunctivitis are clinically similar.

C. Gastrointestinal Disease
Diarrhea: Some fastid­ious adenoviruses can cause
Fig. 59.1: Morphology of advenovirus diarrheal disease in children (for example, types
40 and 41).
A. Respiratory Diseases
1. Pharyngitis: Adenoviruses are the major cause D. Other Diseases
of nonbacterial pharyngitis and tonsillitis,
Mesenteric adenitis and intussusception in children,
presenting as febrile common cold. Types 1–7
pertussis­-like illness, acute hemorrhagic cystitis with
are commonly re­sponsible.
dysuria and hematuria in young boys, musculoskeletal
2. Pneumonia: Adenovirus types 3 and 7 are
disorders, genital and skin infections.
associated with pneumonia in adults resembling
primary atypical pneumonia. Adenoviruses,
E. Systemic Infection in Immunocompromised
particularly types 3, 7, and 21 are thought to be
responsible for about 10–20% of pneu­monias Patients Include Pneumo­nia and Hepatitis
in childhood. In infants and young children
type 7 may lead to more serious and even fatal Laboratory Diagnosis
pneumonia.
3. Acute respiratory diseases: Adenoviruses are A. Specimens
the cause of an acute respiratory disease (ARD) Depending on the clinical disease, virus may be
syndrome among military recruits. Serotypes 4, recovered from stool or urine or from a throat,
7 and 21 are the agents commonly isolated. conjunctival, or rectal swab.

Table 59.2:  Disease associated with adenovirus serotypes


Disease Those at risk Associated serotypes
1. Acute febrile pharyngitis
Endemic Infants, young children 1, 2, 5, 6
Epidemic Infants, young children 3, 4, 7
2. Pneumonia Infants 1, 2, 3, 7
3. Acute respiratory disease Military recruits 4, 7, 14, 21
4. Pharyngoconjunctival fever Older school-age children 3, 7
5. Epidemic keratoconjunctivitis (ship yard Adults 8, 19, 37
eye)
6. Follicular (swimming pool) conjunctivitis Any age 3, 4, 11

7. Diarrhea and vomiting Infants, young children 40, 41


8. Intussusception Infants 1, 2, 5
9. Hemorrhagic cystitis Infants, young children 11, 21
10. Disseminated infection Immunocompromised, e.g. AIDS, renal, bone 5, 11, 34, 35, 43–47
marrow and heart-lung transplant recipients

www.ebook3000.com
420  |  Section 4: Virology
B. Direct Demonstration of Virus deaminase deficiency), cystic fibrosis, lysosomal
i. Electron microscopy: Virus particles may storage diseases, and even cancer.
be seen directly in stool extracts by electron
Adenovarus-associated Viruses
microscopy.
The adenovirus-associated viruses (AAVs) are
ii. Virus antigen: The presence of viral antigen
members of the parvoviridae. They are about 22
in the nasopharynx may be identified by
nm in diameter, appear to be more hexagonal
immunofluorescence with group-specific
than circular in outline and contain insufficient
antibodies (polyclonal or monoclonal) directly
single-stranded DNA to replicate on their own.
on aspi­rates. Alternatively, viral antigen may be
They form a genus, dependoviruses, indicating their
detected by enzyme immunoassays.
dependence on adenoviruses (or herpes simplex
iii. Viral DNA: It is also possible to detect viral
virus) to provide the missing functions.
DNA directly from feces by polyacrylamide gel
They can be detected by electron microscopy
electrophoresis.
and complement fixation or immun­ofluorescence
iv. La­tex agglutination method: It uses latex
with specific antisera. Types 1, 2 and 3 are of human
particles coated with specific antibody to each
origin and cause natural infection, while type 4 is of
virus.
simian origin. Their pathogenic role is uncertain.
True AAV (also known as adenovirus satellite
C. Virus Isolation
virus) has not been implicated in clinical disease
The clinical specimens are inoculated in tissue and its pathogenic role is uncertain.
culture such as HeLa, Hep., KB and human embryo
kidney cells. The development of char­acteristic Key Points
cytopathic effects-rounding and clustering of ™™ Adenoviruses are nonenveloped, icosahedral, DNA
swollen cells—indicates the presence of adenovirus viruses. The virion has the appearance of a space
in inoculated cultures. vehicle
Isolates can be identi­fied as adenoviruses by ™™ Adenoviruses infect and replicate in epithelial cells
of the respiratory tract, eye, gastrointestinal tract,
immunofluorescence tests using an antihexon urinary blad­der, and liver.
antibody and infected cells hemagglutination ™™ Adenovirus-associated viruses (AAVs) are members of
inhibition and neutralization tests. the parvoviridae. They form a genus, dependoviruses,
Characterization of viral DNA by hybridization indicating their dependence on adenoviruses
or by restriction endonuclease digestion patterns (or herpes simplex virus) to provide the missing
can identify an isolate as an adenovirus and group it. functions.

D. Polymerase Chain Reaction Important questions


Polymerase chain reaction (PCR) assays can be 1. Discuss laboratory diagnosis of infections caused
used for diagnosis of adenovirus infections. by adenoviruses.
2. Write short notes on:
E. Serology a. Adenoviruses.
b. Adenovirus-associated viruses.
For serological diagnosis, rise in titer of antibodies
should be demonstrated in paired sera. Comple­ Multiple choice questions (mcqs)
ment fixation and neutralization tests may be used
1. What is the symmetry of adenoviruses?
by reference laboratories or in research. a. Icosahedral b. Helical
c. Complex d. None of the above
Treatment, Prevention and Control 2. Which of the following viruses morphology appear
There is no known treatment for adenovirus as a space vehicle?
a. Rabies virus
infection. Live oral vaccines have been used to
b. Adenoviruses
prevent infections with adenovirus types 4 and c. Cytomegalovirus
7 in military recruits. The widespread use of d. Herpes simplex virus
live adenovirus vaccines is unlikely. However, 3. Which of the following cell lines can be used for
genetically engineered subunit vaccines could be the growth of adenoviruses?
prepared and used in the future. a. HeLa. b. HEp-2.
c. KB. d. All of the above.
Gene Therapy 4. All the following infections are caused by adeno­
viruses except:
There is growing interest in the potential use of aden­ a. Pharyngoconjunctival fever
o­viruses as gene delivery vehicles for gene therapy or b. Epidemic keratoconjunctivitis
DNA vaccination. Adenoviruses have been used and c. Acute hemorrhagic cystitis
are being considered for more applications of gene d. Cancer
delivery for correction of several human diseases, Answers (MCQs)
including immune deficiencies (e.g. adenosine 1. a; 2. b; 3. a; 4. d
60
Chapter

Papovaviruses

Learning Objectives

After reading and studying this chapter, you should be able to:
∙∙ Describe the following: Papillomaviruses; Polyomaviruses.

Introduction adults. In women, they are found on the vulva,


within the vagina and on the cervix. In men, the
The term ‘Papova’ is a sigla indicating the names most common sites for lesions are the shaft of the
of viruses included in this group: (Pa, papilloma; penis, perianal skin and the anal canal.
po, polyoma; va, vacuolating virus) belong to Human papillomavirus types 6 and 11 are
the family Papovaviri­dae and has two genera— commonly found in benign vulval or penile warts.
Papillomavirus and Polyomavirus. Cervical and anogenital warts may be due to HPV
types 16 or I8.
Papillomaviruses
Papillomaviridae family includes papillomaviruses. 3. Recurrent Respiratory Papillomatosis
Papillomaviruses are slightly larger in diameter This is a rare condition characterized by the presence
(55 nm) than the polyomaviruses (45 nm) and of benign squamous papillomata on the mucosa of
contain a larger genomea. All papil­lomaviruses the respiratory tract, most commonly on the larynx.
induce hyperplastic epithelial lesions in their host It has peaks of incidence in children under 5 years
species. Over seventy types of human papilloma­ of age and adults after the age of 15 years. Children
viruses (HPVs) are now recognized. acquire the disease by passage through an infected
birth canal, while adults acquire the disease from
Pathogenesis orogenital contact with an infected sexual partner.
Papillomaviruses cause several different kinds It is caused by infection of the respiratory mucosa
of warts in humans, including cutaneous warts, with HPV types 6 and 11.
genital warts, respiratory papillomatosis, oral
papillomas and cancer. 4. Oral Papillomatosis
A variety of papillomata and benign lesions
1. Cutaneous Warts associated with HPV occur on the oral mucosa and
These are frequently seen in young children and tongue. Multiple lesions may develop on the buccal
adolescents. The viruses associated with such mucosa, a condition known as oral florid papillo­
lesions are HPV types 1 and 4 (plantar warts); 3 and matosis. Infection is acquired during orogenital
10 (flat warts); 2, 4 and 7 (common warts). contact with an infected sexual partner. Several
HPV types, including types 2, 7, 13 and 32, have
Epidermodysplasia Verruciformis been found.
Multiple warts that do not regress, but instead
spread to many body sites. 5. Cancer
HPV DNA can be detected in all grades of the
2. Anogenital Warts premalignant lesions of the female and male genital
These lesions (also known as condylomata tract. HPV types 6 and 11 are most commonly
acuminata) are commonest in sexually active found in low-grade disease whereas. HPV types 16

www.ebook3000.com
422  |  Section 4: Virology
and 18 are more commonly associated with lesions 4. BK Polyomaviruses
of greater severity and invasive cancer. The human polyomaviruses (BK and JC) have been
isolated from immunocompromised patients. It
Laboratory Diagnosis was named after the initials of the person from
A. Morphological identification: HPV infection whom it was first isolated. BK virus isolated from
may be readily diagnosed when there are the urine of a patient with kidney transplant.
typical clinical lesions.
Subclinical infection requires lab­o ratory Laboratory Diagnosis
confirmation using:
1. Electron Microscopy
1. Cytological and histological detection.
2. Immunocytochemical detection: HPV capsid Human polyomavirurses can be detected by
antigen in sections of tissues or in cell smears electron microscopy from brain tissue in a case
can be detected by immunoperoxidase test of PML (JC virus) and from the urine of a renal
using antiserum. It detects all the genital transplant case (BK virus).
HPV types.
3. Molecular methods: Amplification of HPV 2. Virus Isolation
DNA by polymerase chain reaction (PCR). Human fetal glial cell culture and human diploid
B. Serology: Are appropriate for epidemiological fibroblasts are used for the isolaton of JC polyomavirus
studies. BK virus respectively. Hemaggltination inhibition is
used to differentiate these two viruses.
Polyomaviruses
These are small viruses (diameter 45 nm) that 3. Viral Antigen Detection
possess a circular genome of double-stranded DNA The brain biopsy or autopsy material can be examined
enclosed within a nonenveloped capsid exhibiting directly for JCV antigen by immunofluorescence or
icosahedral symmetry. immunoperoxidase staining.
The name is derived from ‘poly’ (many) and
‘oma’ (tumor). The viruses are species-specific. 4. Viral nucleic Acid Detection
Recognized members of this group include:
Viral nucleic acid can be detected by nucleic acid
1. Mouse polyomavirus
hybridization and polymerase reaction (PCR).
2. Simian vacuolating virus 40 (SV 40)
3. JC virus (JCV) 5. Cytopathology
4. BK virus (BKV)
Exfoliated urinary epithelial cells show the
1. Mouse Polyomavirus presence of enlarged deeply stained basophilic
nuclei with a single inclusion.
It causes harmless infections in mice by natural
routes. However, it induces different types of
Key Points
malignant tumors when injected into infant rodents.
™™ Papovaviruses are double-stranded DNA viruses and
has two genera, Papillomavirus and Polyomavirus
2. Simian Vacuolating Virus 40 (SV 40) ™™ Papillomaviruses are species-specific DNA viruses
The simian vacuolating virus (SV 40) was isolat­ed that infect the squamous epithelia and mucous
from uninoculated rhesus and cynomolgus membranes of vertebrates, including man
monkey kidney tissue cultures. SV40 is oncogenic ™™ Over seventy types of human papillomaviruses
in newborn hamsters. Its only medical importance (HPVs) are now recognized
™™ Papillomaviruses cause several different kinds of
is that because of its oncogenic potential, live viral
warts in humans, including cutaneous warts, genital
vaccines should be manufactured only in monkey warts, respiratory papillomatosis, oral papillomas
kidney tissue cultures tested and found free from and cancer
SV 40 infection. ™™ Polyomaviruses include: Mouse polyomavirus, simian
virus 40 (SV 40) of monkeys, JC virus (JCV) and BK
3. JC Virus (JCV) polyomavirus.

The JC virus was isolated from the brain of a patient


with Hodgkin’s disease and progressive multifocal Important question
leukoencephalopathy (PML). It was named Write short notes on:
after the initials of the person from whom it was a. SV40
first isolated. JC virus is the cause of progressive b. Papovavirus
multifocal leukoencephalopathy (PML). c. Polyomavirusess
Chapter 60: Papovaviruses  | 423
Multiple choice questions (Mcqs) c. SV40 virus
d. Polyomavirus of mice
1. Cervical papillomas and dysplasia in human is 3. BK polyomavirus is isolated from urine by culture
associated with human papillomaviruses: in:
a. HPV-l b. HPV-16 a. Human diploid fibroblasts
c. HPV-26 d. HPV-28 b. Human fetal glial cell culture
2. The polyoma virus that causes progressive multifocal c. Vero cell culture
leukoencephalopathy (PML) in humans is: d. HeLa cell culture
a. BK virus Answers (MCQs)
b. JC virus 1. b; 2. b; 3. a

www.ebook3000.com
61
Chapter

Parvoviruses

Learning Objectives

After reading and studying this chapter, you should be able to:
∙∙ Describe Parvoviruses.

Introduction Clinical Diseases


The parvoviruses are the smallest of the DNA 1. Minor illness: Nonspecific respiratory tract
viruses (about 20 nm) and have been isolated illness is the next most common illness, at least
from a wide range of organisms, from arthropods in boys.
to humans. 2. Erythema infectiosum (Fifth disease): B19
The family Parvoviridae is divided into two virus causes an erythematous maculopapular
subfamilies: the Parvovirinae and the Densovirinae. rash, which in its most clinically distinct form
The Parvovirinae contains three genera: Parvovirus, is called erythema infectiosum. It is common
Dependovirus and Erythrovirus. in children aged 4–11 years, and is sometimes
called fifth disease. Classically, it starts with an
A. Parvovirus intense erythema of the cheeks, hence another
of its names—‘slapped-cheek disease’. The rash
Parvovirus genus contains the autonomous par­
then proceeds to involve the trunk and limbs.
voviruses, which are widespread in nature and
There may be associated lymphadenopathy
capable of autonomous replication. The group
and joint symptoms.
includes feline and canine parvoviruses (FPV
3. Joint disease: In addition to a rash, a compli­
and CPV) which are so important in veterinary
cation accompanying B19 infection is an acute
medicine that immunization against them is a
arthritis.
routine practice in developed countries.
4. Aplastic crisis: Parvovirus B19 induces in
B. Dependovirus childern with aplastic crisis with chronic
hemolytic anemia (e.g. sickle cell anemia or
In contrast to par­voviruses, dependoviruses require thalassemia).
helper virus functions for replication. The most
5. Infection during pregnancy: It may result in
studied are the human adeno-­associated viruses
nonimmmune fetal hydrops.
(AAV), to assist in replication.
6. Infection in immunosuppressed: Persistent
infections have been described in patients with
C. Erythrovirus
underlying immunodeficiency states.
There is only one parvovirus (B19) known to cause
disease in humans.
Laboratory Diagnosis
Parvovirus (B19): The cell receptor for parvovirus
B19 is the P antigen. This is present on red blood Diagnosis may be made by detection of virus in the
cells, erythroid progenitors, vascular endothelium blood in early cases, and of antibody later.
and fetal myocytes. Infection is acquired in 1. Virus isolation: Parvovirus B19 may be cultured
childhood and is often asymptomatic. Transmission in cells from human bone marrow or fetal liver.
of parvoviruses appears to be by the respiratory It can be detected in patient’s blood by electron
route. Parvovirus 19 is highly contagious. microscopy.
Chapter 61: Parvovirus  | 425
2. Detection of viral nucleic acid: Nucleic acid Multiple choice questions (Mcqs)
can be deteted by nucleic acid dot blot hybridiz­
1. Which of the following viruses is also known as
ation or by polymerase chain reaction-based “adeno-associated virus”?
assays. a. Parvovirus
3. Antigen detection: Detection of viral antigen b. Erythrovirus
is done by counterimmunoelectrophoresis, c. Dependovirus
ELISA, RIA or indirect immunofluoescence. d. JC polyoma virus.
4. Antibody detection: IgM antibodies or a 2. B19 virus infection is associated with all the
significant rise in IgG antibodies can be deted by following infections except:
ELISA or RIA. It is the most successful technique. a. Erythema infectiosum
b. Fulminant hepatitis
Key Points c. Aplastic crisis
d. Hydrops fetalis
™™ Parvoviruses are the smallest of the DNA viruses
(about 20 nm) 3. B19 virus infection is transmitted by all the
™™ The family Parvoviridae is divided into two following routes except:
subfamilies: the Parvovirinae and the Densivirinae a. Sexual transmission
™™ The Parvovirinae contains three genera: Parvovirus, b. Aerosol droplets
Dependovirus and Erythrovirus c. Feco-oral route
™™ Erythrovirus: It is only one parvovirus (B19) known
d. Transfusion of blood and blood products
to cause disease in humans.
4. Which of the following viruses is associated with
“slapped cheek” syndrome?
Important question a. Parvovirus
b. Erythrovirus
Write short notes on: c. Dependovirus
a. Parvovirus d. JC polyoma virus
b. Dependovirus
c. Erythrovirus or Parvovirus (B19) Answers (MCQs)
d. Erythema infectiosum or fifth disease. 1. c; 2. b; 3. c; 4. b

www.ebook3000.com
6262
Chapter

Picornaviruses

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Differentiate coxsackie A and coxsackie B viruses
be able to: ∙∙ Describe diseases caused by coxsackie viruses
∙∙ Describe enteroviruses ∙∙ D escribe the following: acute hemorrhagic
∙∙ Discuss prophylaxis against poliomyelitis conjunctivitis; Rhinoviruses.
∙∙ Differentiate live and killed polio vaccines

INTRODUCTION Genome: Single-stranded RNA, linear, positive-


sense.
The family Picornaviridae comprises a large
number of very small RNA (pico, meaning small, Proteins: Four major polypeptides cleaved from a
RNA: rna) viruses with a diameter of 27–30 nm large pre­cursor polyprotein. Surface proteins VPl
and containing single-stranded RNA. They are and VP3 are major antibody-binding sites. Internal
nonenveloped viruses, re­sistant to ether and other protein VP4 is associated with viral RNA.
lipid solvents. Envelope: None
Replication: Cytoplasm
CLASSIFICATION Culture: Many enteroviruses (polioviruses,
The Picornaviridae family has more than 230 echoviruses, some coxsackieviruses) can be grown
members that are divided into six genera: at 37°C in human and monkey cells. Most rhinovirus
Enterovirus, Rhinovirus, Cardiovirus, Aphthovirus, strains can be recovered only in human cells at 33°C.
Hepatovirus and Parechovirus (Table 62.1). Coxsackieviruses are pathogenic for newborn mice.
Important properties of picornaviruses are
IMPORTANT PROPERTIES OF summerized in Table 62.2.
PICORNAVIRUSES
ENTEROVIRUSES
Size: 28–30 nm in diameter.
Enteroviruses of medical importance include
Virion: Icosahedral, contains 60 sub­units. polioviruses, coxsackieviruses and echoviruses
Table 62.1:  Picornaviridae Table 62.2:  Some properties of picornaviruses
1. Enterovirus Property Enteroviruses Rhinoviruses
Poliovirus types 1, 2 and 3
Size (nm) 22–30 30
Coxsackie A virus types 1–22 and 24
Coxsackie B virus types 1–6 Capsid
Echovirus (ECHO virus) types 1–9, 11–27, and 29–34
Form Icosahedral Icosahedral
Enterovirus 68–72
Polypeptides VP1, VP2, VP3, VP4 VP1, VP2, VP3, VP4
2. Rhinovirus types 1–100+ most important cause of
RNA type Single-stranded, Single-stranded,
the common cold positive-sense positive-sense
3. Cardiovirus of mice, including the
encephalomyocarditis virus Optimal 37°C 33-34°C
4. Aphthovirus causing the foot and mouth disease of cat­tle temperature for
5. Heparnavirus—Hepatitis A virus growth
6. Parechovirus (pare­choviruses)—Echovirus 22 and 23. Acid Stable (pH 3–9) Labile (pH 3–5)

Chap-62.indd 426 15-03-2016 11:22:28


Chapter 62: Picornaviruses  | 427
because they are all found in the intestines and The infected cells round up and become refra­
are excreted in the faeces. At least 72 serotypes ctile and py­k notic. Eosinophilic intranuclear
of human enteroviruses exist, including the inclusion bodies may be demonstrated in stained
polioviruses types 1–3, coxsackieviruses A types preparations. Well-formed plaques develop in
1–24, coxsackievirus B types 1–6, echovirus types infected monolayers with agar overlay.
1–34 and enterovirus types 68–72.
Enterovirus 72 is the virus causing infectious Pathogenesis
hepatitis (Hepatitis type A), which has been The virus is transmitted by the fecal-oral route
reclassified as a separate genus Hepatovirus. through ingestion. Inhalation or entry through
Because of its special status, it is considered in the conjunctiva of droplets of respiratory secre­tions
chapter on Hepatitis Viruses. may also be possible modes of entry in close
contacts of patients in the early stage of the disease.
POLIOVIRUS Virus is ingested and multiplies initially in the
lym­phoid tissue of the tonsil or Peyer’s patches in
Morphology the small intestine. It then spreads to the regional
Size: The virion is a spherical particle, about 27 lymph nodes and enters the bloodstream (mi­nor or
nm in diameter. primary viremia). Primary vire­mia spreads the virus
Capsid: It consists of a capsid shell of 60 subunits, to receptor-bearing target tissues, where a second
each consisting of four viral proteins (VP1–VP4), phase of viral replication may occur, resulting in
arranged in icosahedral symmetry. symptoms and a secondary viremia.
Genome: The genome is a single strand of positive
sense RNA. Clinical Features
The incubarion period is usually 7–14 days, but
Resistance it may range from 3 days to 35 days. Following
exposure to poliovirus, 90–95% of susceptible
1. Poliovirus is resistant to ether, chloroform, bile,
individuals develop only inapparent infection,
proteolytic enzymes of the intestinal con­tents
which causes seroconver­sion alone. It is only in
and detergents.
5–10% that any sort of clinical illness results.
2. It is stable at pH 3.
1. Asymptomatic illness : At least 90% of
3. In feces, virus can survive for months at 4°C, for
poliovirus infections are asymptomatic.
years at –20 or –70°C and at room temperature
2. Abortive poliomyelitis, the minor illness:
for several weeks.
is a nonspecific febrile illness occurring in
4. They are inactivated when heated at 55°C for
approximately 5 % of infected people.
30 min­u tes, but molar MgCl 2 prevents this
3. Nonparalytic poliomyelitis or aseptic
inactivation.
meningitis: It occurs in 1–2% of patients with
5. Drying rapidly inactivates enteroviruses by
poliovirus infec­tions. In this disease, the virus
ultraviolet light, and usually by drying.
progresses into the cen­tral nervous system and
6. Formaldehyde and oxidizing disinfectants
the meninges, causing back pain and muscle
destroy the virus.
spasms in addition to the symptoms of the
7. Chlorination destroys the virus in water but
minor illness.
organic matter delays inactivation.
4. Paralytic poliomyelitis, the major illness:
8. Poliovirus does not survive lyophilization well.
Paralytic polio, the major illness, occurs in
0.1–2.0% of persons with poliovirus infections.
Antigenic Properties The predominating complaint is flaccid paralysis
Two antigens C and D (C = coreless or capsid; D = resulting from lower motor neuron damage.
dense) can be recognized by complement fixation, Depending on the distribution of paralysis, cases
enzyme-linked immunosobent assay (ELISA) or are classified as spinal, bulbar or bulbospinal.
precipitation tests. Mortality ranges from 5–10% and is mainly due
Types: There are three types (1, 2 and 3) of to res­piratory failure.
poliovirus, identified by neu­tralization tests. The 5. Progressive postpoliomyelitis muscle atrophy
three types are antigenically distinct, but overlap That may occur much later in life of the original
occurs in neutralization tests. The prototype strains victims.
are as follow.
Laboratory Diagnosis
Host Range and Cultivation A. Specimens
Primary monkey kidney cultures are used for Many speci­mens can be used, including blood,
diagnostic cultures and vaccine production. CSF, throat swabs and feces.

www.ebook3000.com
Chap-62.indd 427 15-03-2016 11:22:28
428  |  Section 4: Virology
B. Culture 2. Oral polio vaccine (OPV) (Sabin vaccine)
Primary monkey kidney cells are usually employed, Oral polio vaccine (OPV) was described by Sabin
though any other human or simian cell culture may in 1957. It contains live attenuated virus (types
be used. The virus growth is indicated by typical 1, 2 and 3) grown in primary monkey kidney or
cytopathic effects in 2–3 days. An isolated virus is human diploid cell. cultures. The use of molar
identified and typed by neutralization with specific MgCl2 or sucrose stabilizes the vaccine against heat
antiserum. inactivation, partic­ularly under tropical conditions.
It can be given to young infants, as the maternal
The mere isolation of poliovirus from feces does
antibody has little effect on in­testinal infection.
not constitute a diagnosis of poliomyelitis. Virus
isolation must be inter­preted along with clinical The shelf life of the vaccine at 4–8°C is four
and serological evidence. months and at –20°C is two years. Improper storage
conditions and ‘cold chain’ failure may be partly
responsible for the apparent failure of OPV to
C. Serological Tests
control poliomyelitis in the developing countries.
Serodiagnosis is less often employed. Antibody rise
can be demonstrated in paired sera by neutralisa­ Criteria of attenuated strains for live vaccine
tion or complement fixation tests. 1. They should not be neurovirulent.
2. They should be able to set up intestinal infect­
Immunity ion following feeding and should induce an
Immunity is permanent to the type causing immune response.
the infec­tion. Humoral immunity provided by 3. They should not acquire neurovirulence after
circulating and secre­tory antibody is responsible serial enteric passage.
for protection against po­liomyelitis. 4. They should possess stable genetic character­
The virus also induces cell mediated immunity, istics (markers) by which they can be differenti­
but its impor­tance appears to be uncertain. ated from the wild virulent strains.
Markers for differ­entiating the wild from the
Prophylaxis attenuated strains
Immunization 1. d marker: Wild strains will grow well in low
Both killed and live attenuated vaccines are levels of bicar­bonate but avirulent strains will
available. Two types of vaccines are used throughout not.
the world; they are: 2. rct 40: Wild strains grow well at 40°C, while
1. Inactivated (Salk) polio vaccine (lPV) avirulent strains grow poorly.
2. Oral (Sabin) polio vaccine (OPV). 3. MS: Wild strains grow well in a sta­ble cell line
of monkey kidney, while avirulent strains grow
1. Inactivated polio vaccine(IPV)—Salk’s killed poorly.
polio vaccine
4. McBride’s intratypic an­tigenic marker shown
By 1953, Salk had developed a killed vaccine.
by the rate of inactivation by specific antiserum.
Salk’s killed polio vaccine is a formalin inactivat­ed
preparation of the three types of poliovirus grown in
monkey kidney tissue culture. Killed vaccine is given Immunization Schedule
by injection and is therefore called inactivated or The WHO Program on Immunization (EPI) and
injectable poliovaccine (lPV). The primary or initial the National Immunization Program in India
course of immunization consists of 4 inoculations. recommend a primary course of 3 doses of OPV
The first 3 doses are given at intervals of 1–2 months at one-month intervals, commencing the first
and 4th dose 6–12 months after the third dose. First dose when the infant is 6 weeks old. One booster
dose is usually given when the infant is 6 weeks old dose of OPV is recommended 12–18 months later.
to ensure that immune response is not impaired by It has been recommended that in the tropics, the
residual maternal antibod­ies. Additional doses are number of doses of vaccine be increased to five, in
recommended prior to school entry and then every. order to enhance seroconversion in the vaccinees.
5 years until the age of 18. It is very important to complete vaccination of all
IPV induces humoral antibodies (lgM, IgG infants before 6 months of age. This is because most
and IgA serum antibodies) but does not induce polio cases occur between the ages of 6 months
intestinal or local immunity. The circulating and 3 years.
antibodies protect the individual against paralytic There has been much controversy about the
polio, but do not prevent reinfection of the gut by rel­ative merits of killed and live vaccines. The
wild viruses. In the case of an epidemic, IPV is differences between IPV and OPV are given in
unsuitable. Table 62.3.

Chap-62.indd 428 15-03-2016 11:22:28


Chapter 62: Picornaviruses  | 429
Table 62.3:  Differences between killed (IPV) and live polio vaccines (OPV)
Killed polio vaccine (Salk type) Live polio vaccine (Sabin type)
Virus Killed formolized virus Live attenuated virus
Route of administration Given subcutaneously or 1M Given orally
Nature of immunity Induces circulating antibody, but Immunity is both humoral and intestinal.
no local (intestinal) immunity Induces antibody quickly
Prevents paralysis, but does not Prevents not only paralysis,
prevent reinfection by wild polio but also intestinal reinfection
viruses
Useful in controlling epidemics Not useful in controlling Can be effectively used in controlling epi­demics.
epidemics Even a single dose elicits substantial immunity
(except in tropical countries)
Manufacture More difficult to manufacture Easy to manufacture
Duration of immunity Life long Booster vaccine needed for lifelong immunity
Cost The virus content is 10,000 times Cheaper
more than OPV; hence costlier
Storage Does not require stringent Requires to be stored and transported at
conditions during storage, and subzero temperatures, unless stabilized
transportation
Has a longer shelf-life.
Contraindicated in No Yes
immunodeficiency states or
pregnancy

Global Eradication COXSACKIEVIRUS


By global immuniza­tion with OPV, it is possible The prototype strain was isolated by Dalldorf and
to eradicate the disease. A major campaign by Sickles (1948) from the village of Coxsackie in
the World Health Organization is underway to New York. Coxsackieviruses are classified into two
eradicate poliovirus from the world as was done groups, A and B based on the path­ological changes
for smallpox virus. The Americans were certified produced in suckling mice (Table 62.4).
as free from wild poliovirus in 1994, the Western
Pacific Region in 2000, and Europe in 2002. Progress Properties of the Virus
is being made globally, but several thousand cases
Coxsackieviruses are highly infective for newborn
of polio still occur each year, principally in Africa
mice. Following inoculation in suckling mice,
and the Indian subcontinent.
Group A viruses produce widespread myositis in
Epidemiology the skeletal muscles of newborn mice, resulting
in flaccid paralysis without other observable
Poliomyelitis is an exclusively hu­man disease and
lesions leading to death within a week.Group
the only source of virus is humans. The virus is
B viruses may produce a patchy fecal my­ositis,
transmitted by fecal oral route through ingestion.
spastic paralysis, necrosis of the brown fat and,
The disease occurs in all age groups, but
often, pancreatitis, hepatitis, myocarditis and
children are usually more susceptible than adults.
encephalitis (Table 62.4).
In India, polio is essentially a disease of infancy
and childhood. In developed countries, before the
advent of vaccination, the age dis­tribution shifted Antigenic Characters
so that most patients were over age 5 and 25% were Thirty antigenic types have been defined by cross-
over age 15. The case fatality rate is variable. It is neutralization tests in mice or cell culture, and
highest in the oldest patients. cross-complement fixation reactions. Twenty-three
Most infections are subclinical. It is estimated have the features of group A and six have those
that for every clinical case, there may be 1000 of group B. Coxsackie A 23 is the same as echo 9
subclinical cases in children and 75 in adults. (Coxsackievirus A23 has now been reclassified
Poliovirus type I is responsible for most as echovirus 9) and Coxsackie A24 the same as
epidemics of paralytic poliomyelitis. Type 3 also ECHO 34.
causes epidemics to a lesser extent. Type 2 usually
causes inapparent infections in the western coun­ Clinical Features
tries but in India paralysis due to type 2 is quite Like other enteroviruses, coxsackie­viruses inhabit
common. Immunity is type-specific. the alimentary canal primarily and are spread

www.ebook3000.com
Chap-62.indd 429 15-03-2016 11:22:28
430  |  Section 4: Virology
Table 62.4:  Features of coxsackieviruses A and B children and adults but are most threatening in
infection in the laboratory newborns.
Features Coxsackievirus A Coxsackievirus B 3. Aseptic meningitis with paralyisis.
1. Growth in + + 4. Juvenile diabetes
monkey Juvenile diabetes has been claimed to be associated
kidney with coxsackie B4 infection.
2. Effect in Generalized Patchy focal
suckling myositis my­ositis 5. Neonatal infections
mice Flaccid paralysis Spastic paralysis, Transplacental and neonatal transmission has
Death within a Necrosis of the been demonstrated with coxsackie B viruses,
week brown fat and, resulting in a serious disseminated disease that
often, pancreatitis,
may include hepatitis, meninoencephaIitis and
hepatitis,
myocarditis and adrenocortical involvement.
encephalitis
6. Chronic fatigue syndrome
3. Types 23 (1–24*) 6 (1–6) There is some evidence of a possible association
*Coxsackievirus A23 now classified as echovirus 9. between chronic fatigue syndrome and infection
with enteroviruses, particularly coxsackie B viruses.
by the fecal–oral route. Young infants are most
commonly affected. The incubation period of Laboratory Diagnosis
coxsackievirus infection ranges from 2 days to 9 A. Virus Isolation
days. The clinical manifestations of infection with
yarious coxsackieviruses are diverse and may The virus is isolated readily from throat washings
present as distinct disease entities (Table 62.4). conjunctival swabs, throat swabs and feces.
i. Inoculation into suckling mice: Specimens
Group A Viruses are inoculated into suckling mice. In suckling
mice, signs of illness appear usually within 3–8
1. Herpangina (vesicular pharyngitis)
days with group A strains and 5–14 days with
Herpangina is a severe febrile pharyngitis and is a
group B strains. Identification is by studying
common clinical manifestation of coxsackie group
the histopathology in infected mice and by
A infec­tion in children.
neutralization tests.
2. Aseptic meningitis ii. Tissue culture: Specimens are inoculated into
Aseptic meningitis is caused by all types of tissue cultures. Of group A coxsackieviruses,
group B cox­sackieviruses and by many group A only A7 and A9 grow in monkey kidney cells,
coxsackieviruses, most commonly A7 and A9. and coxsackievirus A21 can be grown in
3. Hand-foot-and-mouth disease HeLa, HEp2 or human embryonic kidney cell
Hand-foot-and-mouth disease is a vesicular exan­ cultures. All group B coxsackieviruses grow
them and is characterized by oral and pharyngeal readily in monkey kidney cell cultures.
ulcerations and a vesicular rash of the palms and
soles. This disease has been associated particularly B. Serology
with coxsackievirus A16, but A5 and Al0 have also Serologic tests are difficult to evaluate (because of
been implicated. the multiplicity of types).
4. Respiratory infections
A number of the enteroviruses have been asso­ C. Nucleic Acid Detection
ciated with common colds; among these are Reverse transcrip­tion–polymerase chain reaction
coxsackieviruses A21, A24, B1, and B3–5. tests can be broadly reactive.
Group B Viruses Prevention: Vaccination is not practi­cable as there
1. Epidemic myalgia or Born­holm disease are several serotypes and immunity is type-specific.
It is so called because it was first described on the
Danish island of Bornholm. It is an acute illness ECHOVIRUSES
in which patients have a sudden onset of fever Echoviruses (enteric cytopathogenic human
and unilateral low tho­racic, pleuritic chest pain orphan viruses) are grouped together because
that may be excruciating. Coxsackie B virus is the they infect the human enteric tract. They were
causative agent. called orphans as they could not be associated
2. Myocardial and pericardial infections with any particular clinical disease then. There is
Myocardial and pericardial infections caused still no clear association of some types with specific
by coxsackie B virus occur sporadically in older diseases.

Chap-62.indd 430 15-03-2016 11:22:28


Chapter 62: Picornaviruses  | 431
Clinical Features 3. Serology
Most echovirus infections are asymptomatic but Serological diagnosis is impractical.
some have been associated with clinical syndromes 4. Polymerase chain reaction (PCR)
(Table 62.5). Nucleic acid detection assays, such as polymerase
1. Aseptic meningitis chain reaction, are more rapid than virus isolation
2. Respiratory illnesses in children for diagnosis.
3. Infantile diarrhea has not been established.
4. Occasionally, there is con­junctivitis, muscle OTHER ENTEROVIRUS TYPES
weakness and spasm. Four enteroviruses (types 68–71) grow in monkey
kid­ney cultures, and three of them cause human
Laboratory Diagnosis disease (Table 62.5).
1. Specimen
Acute Hemorrhagic Conjunctivitis
The procedure of choice is isolation of virus from
A pandemic of acute hemorrhagic conjunctivitis,
throat swabs, stools, rectal swabs, and, in aseptic
apparently arising in West Africa in 1969 spread
meningitis, cerebrospinal fluid.
widely involving several parts of Africa, the Mid­dle
2. Virus isolation East, India, South-East Asia, Japan, England and
Specimens may be inoculated into monkey Europe. The disease is most common in adults,
kidney tissue cul­tures and virus growth detected with an incubation period of 1 day. The symptoms
by cytopathic changes. The large number of are sudden swelling, congestion, watering and
serotypes makes identification by neutralization pain in the eyes. Sub­conjunctival hemorrhage is
tests laborious. a characteristic feature. Complete recovery is the
rule. There is transient corneal involvement but
Table 62.5:  Illness associated with recently recovery is usually complete in 3–7 days.
identified enteroviruses Enterovirus 70 is the chief cause of acute
Enterovirus type Clinical illness
hemor­r hagic conjunctivitis. It grows only on
cultured human cells (human embryonic kidney or
68 Pneumonia and bronchiolitis
HeLa) on primary isolation, but can be adapted to
69 Isolated from an ill person in Mexico grow on monkey kidney cells. Coxsackievirus type
70 Acute hemorrhagic conjuctivitis A 24 also produces the same disease.
70, 71 Paralysis, meningoencephalitis Enterovirus 71 has been isolated from patients
71 Hand-foot-and-mouth disease
with meningitis, encephalitis and paralysis res­
embling poliomyelitis.
72* Hepatitis A
Various clinical syndromes associated with
*Reclassified as Hepatavirus enteroviruses are given in Table 62.6.

Table 62.6  Summary of clinical syndromes caused by major enterovirus groups


Syndrome Polioviruses Coxsackie A Coxsackie B Echoviruses Enterovirus
virus virus types 68–71
Aseptic meningitis 1–3 Many 1–6 Many 71
Paralysis 1–3 7, 9 2–5 2, 4, 6, 9, 11, 30 70, 71
Paralysis
Encephalitis 71 1–3 2, 5–7, 9 1–5 2, 6, 9, 19 70, 71
Fever with rash – 9, 16, 23 – 4, 6, 9, 16 –
Herpangina – 1–6, 8, 10 – – –
Hand, foot and – 5, 10, 16 – – 71
mouth disease
Upper respiratory – 21 – 11, 20 –
infection
Pneumonitis, – – – – 68
bronchiolitis
Bornholm disease – – 1, 5 – –
Myocarditis, 1, 5 – –
pericarditis
Acute hemorrhagic – 24 – – 70
conjunctivitis

www.ebook3000.com
Chap-62.indd 431 15-03-2016 11:22:28
432  |  Section 4: Virology
RHINOVIRUSES Treatment and Prophylaxis
Rhinoviruses are the common cold viruses and Antiviral drugs are thought to be a more likely
are the most important cause of the com­mon cold con­trol measure for rhinoviruses. Pleconaril is one
and upper respiratory tract infections. They are such drug showing activity against rhinoviruses
usually iso­lated from nasal secretions but may also and enteroviruses.
be found in throat and oral secretions. Hand washing and the disinfection of contamin­
ated objects are the best means of preventing the
spread of the virus.
Properties of the Virus
Key Points
They differ from enteroviruses in being more
acid labile, but more heat stable. Inactivation of ™™ The Picornaviridae family has divided into six genera:
Enterovirus, Rhinovirus, Cardiovirus, Aphthovirus,
rhinoviruses occurs below pH 6.0 and is more rapid
Hepatovirus and Parechovirus
the lower the pH. They are relatively stable in the
™™ Enteroviruses include the polioviruses, echoviruses,
range from 20 to 37°C (Table 62.2).
coxsackieviruses
By neutralization tests, they have been classified Polioviruses
into over 113 serotypes. Immunity is type-specific. ™™ Poliovirus is the causative agent of poliomyelitis
™™ Prevention of poliomyelitis is accomplished by an
inactivated (killed) injectable vaccine (Salk vaccine)
Host Range and Growth or an attenuated live, orally administered vaccine
(Sabin vaccine), which confers immunity by raising
Rhinoviruses can be grown in tissue cultures of
neutralising antibody
human or simian origin with cytopathic changes.
Coxsackieviruses
Rhinoviruses were classified into three groups, H,
™™ Based on the pathological changes produced in
M and O, depending upon growth in tissue culture,. suckling mice, they are classified into two group
H strains grew only in human cells, while M strains A and B
grew equally well in human and monkey cells. Zero ™™ The clinical manifestations of infection with yarious
strains could be grown only in nasal or tracheal coxsackieviruses are diverse and may present as
ciliated epi­thelium. distinct disease entities
Echoviruses
™™ Echoviruses are found in the intestinal tract of the
Pathogenesis infected humans
™™ Most echovirus infections are asymptomatic but
The virus enters via the upper respiratory tract. some have been associated with clinical syndromes
Local inflammation and cytokines may be Rhinoviruses
responsible for the symptoms of common cold. ™™ Rhinoviruses are the most important causative
agents of the common cold and upper respiratory
tract infections.
Clinical Syndromes
The incubation period is brief from 2 days to 4 IMPORTANT QUESTIONS
days. The acute illness usually lasts for 7 days.
Usual symptoms in adults include sneezing, nasal 1. Name the picornaviruses. Discuss pathogenesis
and laboratory diagnosis of poliomyelitis.
obstruction, nasal discharge, and sore throat; other
2. Write short notes on:
symptoms may include headache, mild cough, a. Prophylaxis against poliomyelitis
malaise and a chilly sensation. There is little or b. Coxsackie­viruses
no fever. c. Echoviruses
d. Enterovirus 70
e. Acute hemorrhagic conjunctivitis
Laboratory Diagnosis f. Rhinoviruses.
The clinical syndrome of the common cold is
usually so characteristic that laboratory diagnosis MULTIPLE CHOICE QUESTIONS (MCQs)
is unneces­sary.
1. The following statement is not true for polioviruses:
A. Specimens: Nose, throat swabs and nasophary­ a. Poliovirus was the first animal virus to be ob-
ngeal aspirates. tained in crystalline form
B. Culture: Cell cultures of human origin such as b. They are enveloped viruses
MRC5 or WI38 are preferred for the isolation c. They have single-stranded RNA genome
d. They have three serotypes
of rhinoviruses. Cultures are incubated at 33°C
and observed microscopically for a CPE. 2. The prototype strains for type 1 poliovirus are:
a. Brunhilde and Mahoney strains
C. Serology: Serology is not feasible. b. Lansing and MEFI strains
D. Nucleic acid detection: Likely to become a c. Leon and Saukett strains
significant tool in diagnosis. d. None of the above

Chap-62.indd 432 15-03-2016 11:22:29


Chapter 62: Picornaviruses  | 433
3. How much percentage of poliovirus infections 6. Which of the following statement is true for coxsa­
may develop into paralytic poliomyelitis? ckie A virus?
a. 90–95% b. 4–8% a. It has six serotypes
c. 1–2% d. 0.1–2% b. It causes herpangioma
4. Which of the following vaccines induces
c. It shows predilection for visceral organs
production of local secretory IgA antibodies?
a. Salk polio vaccine d. It produces spastic paralysis
b. Sabin polio vaccine 7. All the following enteroviruses are associated with
c. Both the above human infections except:
d. None of the above a. Enterovirus 68
5. Which of the following statements is true for rhino­
b. Enterovirus 69
viruses:
a. It has three antigenic types c. Enterovirus 70
b. Amantadine is effective against the virus d. Enterovirus 71
c. It is the most common cause of common cold
d. It is transmitted by transfusion of blood and ANSWERS (MCQs)
blood products 1. b; 2. a; 3. d; 4. b; 5. c; 6. a; 7. a

www.ebook3000.com
Chap-62.indd 433 15-03-2016 11:22:29
63
Chapter

Orthomyxovirus

Learning Objectives

After reading and studying this chapter, you should ∙∙ D escrbe the following: Hemagglutinin (H) and
be able to: neuramini­dase (NA); Antigenic variation in Influenza
∙∙ Differentiate between orthomyxoviruses and virus; Antigenic shift and antigenic drift
paramyxo­viruses ∙∙ Discuss laboratory diagnosis of influenza
∙∙ Describe morphology of influenza virus ∙∙ D escribe the following: Influenza pandemics;
∙∙ Discuss types and subtypes of orthomyxoviruses prophylaxis against influenza; influenza vaccines.

Introduction occur within the type A group of influenza viruses


and to a lesser degree in the type B group, whereas
The name ‘Myxovirus’ was used originally for a type C appears to be antigenically stable.
group of enveloped RNA viruses characterized by
their abil­ity to adsorb into mucoprotein receptors
on erythrocytes, causing hemagglutination. The Influenza viruses
name referred to the affinity of the viruses to
mucins (from myxa, meaning mucus). Despite Morphology
having certain similarities, the orthomyx­oviruses
Virus: The influenza virus is typically spheri­cal,
and the paramyxoviruses are separated into two
with a diameter of 80–120 nm but pleomorphism
distinct groups because of fundamental differences
is common.
in their structures and their patterns of replication.
Table 63.1 lists the important differences between Genome: The virus core consists of ribonucleoprotein
orthomyxovirus and paramyxovirus. in helical symmetr y. The negative sense
singlestranded RNA genome is segmented and
Properties of Orthomyxovirus exists as eight pieces. Also present within the virion
The family Orthomyxoviridae comprises four is the viral RNA-dependent RNA polymerase; this
genera : influenza A , B and C viruses and is essential for infectivity as the virion RNA is of
thogotoviruses. Antigenic changes con­tinually negative sense.
Table 63.1:  Differences between orthomyxovirus and paramyxovirus
Property Orthomyxovirus Paramyxovirus
Size of virion 80–120 nm 100–300 nm
Shape Spherical; filaments in fresh isolates Pleomorphic
Genome Segmented; eight pieces of RNA Single linear molecule of RNA
Diameter of nucleocapsid 9 nm 18 nm
Site of synthesis of ribonucleoprotein Nucleus Cytoplasm
DNA-dependent RNA synthesis Required for multiplication Not required
Effect of actinomycin D Inhibits multiplication Does not inhibit
Antigenic stability Variable Stable
Hemolysin Absent Present
Chapter 63: Orthomyxovirus  | 435
Envelope: The nucleocapsid is surrounded by an It is type-specific and based on its nature,
M1 protein shell, immediately exterior to which influenza vi­ruses are classified into types A, B
is a lipid envelope derived from the host cell. The and C.
M2 protein projects through the envelope to form 2. Matrix (M) protein: M protein antigen is also
ion channels. The protein part of the envelope is type-specific. The M1 proteins line the inside
virus coded but the lipid layer is derived from the of the virion and promote assembly. The M2
modified host cell membrane, during the process protein forms a proton channel in membranes
of replication by budding. and promotes uncoating and viral release.
Peplomers: Projecting from the envelope are
two types of spikes (peplomers): hemagglutinin B. Surface Antigens
(HA) spikes which are triangular in cross-sec­tion The term ‘viral’ or V antigen was formerly used to
and the mushroom-shaped neuraminidase (NA) describe the surface antigen of the influenza virus.
peplomers which are less numerous (Fig 63.1). The V antigen is actually com­posed of at least two
These two surface glycoproteins are the important viruscoded proteins, the haemagglutinin and the
antigens that determine antigenic variation of neuraminidase.
influenza viruses and host immunity. 1. Hemagglutinin (HA): Hemagglutinin (HA) is
a glycoprotein composed of two polypeptides
Resistance —HA1 and HA2. The HA has several functions.
The influenza virus withstands slow drying at room It is the viral attachment protein responsible for
tem­perature on articles such as blankets and glass. hemaglutination and hemadsorotion. It enables
It can be preserved for long periods at –70°C, and the virus to adsorb to mucoprotein receptors on
remains viable indefinitely when freeze-dried. red cells as well as on respiratory epithelial cells.
Influenza viruses are relatively hardy in vitro Anti­hemagglutinin antibodies are produced
and may be stored at 0–4°C for weeks without loss following infection and immunization.
of via­bility. Exposure to heat for 30 minutes at 56°C Mutation-derived changes in HA are responsi­
is sufficient to inactivate most strains. The viruses ble for the minor (drift) and major (shift)
are inactivated by a variety of sub­stances, such as changes in antigenicity. Fifteen distinct HA
20% ether in the cold, phenol, formaldehyde, salts subtypes, named H1 to H15 have been identified
of heavy metals, detergents, soaps, halogens and in avi­an influenza viruses, but only four of them
many others. Iodine is particularly effective. have been found in human isolates so far. Shifts
occur only with influenza A virus.
Antigenic Structure 2. Neuraminidase (NA): Neuraminidase is
a glycoprotein enzyme which destroys cell
The antigens of the influenza virus can be classified
receptors by hydrolytic cleavage. Neura­mini­
as the internal antigens and the surface antigens.
dase activity is also thought to be important in
the final stages of release of new virus particles
A. Internal Antigens from the infected cells. The antineuraminidase
1. Ribo­n ucleoprotein (RNP) antigen: The antibody is formed following infection and
internal antigen is the ribo­nucleoprotein and immunization. It is not as effective in protection
is hence called the RNP antigen. It was also as the antihemagglutinin antibody. It does not
called the ‘soluble’ (S) antigens because it is prevent the adsorption of virus onto cells but
found free in infected tissues and occurs in the can inhibit the release and spread of progeny
supernatant when the virus containing fluid is virions and may thus contribute to limiting
centrifuged. the infection. It is a strain-specific antigen and
exhibits variation. Major differences acquire the
designations NI, N2, and so on. Nine different
subtypes have been identified (Nl–N9).

Antigenic Variation
Influenza viruses are remarkable because of the
frequent ‘antigenic changes that occur in HA and
NA. This is of great importance in the epidemiology
of the disease. Antigenic variability is highest in
influ­enza virus type A and less in type B, while it
has not been demonstrated in type C.
The internal RNP antigen and M protein
Fig. 63.1: Diagrammatic representation of influenza virus antigen are stable but both the surface antigens,

www.ebook3000.com
436  |  Section 4: Virology
hemaggluti­n in and neuraminidase, undergo hemagglutinin titers, but low infectiv­ity. This has
independent anti­genic variations, which may be of been called the von Magnus phenomenon­and is
two types a
­ ntgenic drift (minor anti­genic changes) due to the formation of incomplete virus particles
and antigenic drift (major antigenic changes) in HA lacking nucleic acid.
or NA result in the appearance of a new subtype.
Antigenic drift: Antigenic drift is the gradual
Pathogenesis
se­quential change in antigenic structure occurring Influenza virus spreads from person to person
reg­u larly at frequent intervals. Here the new by air­borne droplets. The viral neuraminidase
antigens, though different from the previous facilitates infection by reducing the viscosity
antigens, are yet related to them. Antigenic drift of the mucous film lining the res­piratory tract
is due to mutation and selection. Antigenic drift and exposing the cell surface receptors for virus
accounts for the periodical epidemics of influenza. adsorption. The ciliated cells of the respiratory
tract are the main sites of viral infection. Within a
Antigenic shift: Antigenic shift is an abrupt, short time, many cells in the respiratory tract are
drastic, discontinuous variation in the antigenic infected and eventually killed. Influenza infections
structure, resulting in a novel virus strain unrelated cause cellular destruction and desquamation of
antigenically to predecessor strains. Such changes superficial mucosa of the respiratory tract. This
may involve hemagglutinin, neuraminidase or both. renders the respirato­r y tract highly vulnerable
The mechanism for shift is genetic reassortment to bacterial invasion, especially staphylococci,
between human and avian influenza viruses. streptococci, and Haemophilus influenzae. Viral
Antibodies to predecessor viruses do not neutralise pneumonia is seen only in more severe cases.
the new variants and can, therefore, spread widely
in the population causing major epidemics or Clinical Features
pandemic. Influenza B and C viruses do not exhibit
A. Uncomplicated Influenza
antigenic shift because few related viruses exist in
animals. The incubation period is 1–3 days. The disease
varies in severity from a mild coryza to fulminating
and rapidly fatal pneumonia. Most in­fections are
Antigenic Classification
subclinical.
Antigenic differences exhibited by two of the inter­ Symptoms of classic influenza usually appear
nal structural proteins, the nucleocapsid (NP) and abruptly and include chills, headache and dry
matrix (M) proteins, are used to divide influenza cough, followed closely by high fever, generalized
viruses into types A, B and C. These proteins muscular aches, malaise and anorexia. The fever
possess no cross reactivity among the three types. usually lasts 3–5 days, as do the systemic symptoms.
Antigenic variations in the surface glycoproteins,
HA and NA, are used to sub­type the viruses. Only
Complications
type A has designated subtypes. Influenza virus
type A strains can be classified into subtypes based Complications of influenza include primary viral
on variations in their surface antigens. pneumonia, secondary bac­terial pneumonia,
myositis and cardiac complica­t ions, such as
congestive failure or myocarditis and, neurological
Host Range
involvement, such as Guillain–Barre syndrome,
1. Animals: Intranasal inoculation in ferrets encephalopathy, encephalitis and Reye’s syndrome
produces an acute respiratory disease. may occur.
2. Egg inoculation: The virus grows well in
the amniotic cavity of chick embryos. After Reye’s Syndrome
a few egg passages, the virus grows well in
Influenza, particularly infection with type B, has
the allantoic cavity also, except for the type
been associated with Reye’s syndrome. It is an
C virus which does not generally grow in the
acute encephalopathy of children and adolescents,
al­lantoic cavity. Virus growth is detected by the
usually between 2 and 16 years of age and is
appearance of hemaggluti­nin in the allantoic
characterized by acute degenerative changes in
and amniotic fluids.
the brain, liver and kidneys. Type B infections
3. Cell culture: The virus grows in primary may sometimes cause gastrointes­tinal symptoms
monkey kidney cell cultures, as well as in some (gastric flu).
continuous cell lines.
von Magnus phenonomenon: When passaged Laboratory Diagnosis
serially in eggs, using as inocula undiluted infected Diagnosis of influenza relies on isolation of the
allantoic fluid, the progeny virus will show high virus, identification of viral antigens or viral nucleic
Chapter 63: Orthomyxovirus  | 437
acid in the patient’s cells, or demonstration of a and convalescent sera are necessary. A fourfold
specific immuno­logic response by the patient. or greater increase in titer must occur to indi­cate
influenza infection.
1. Demonstration of the Virus Antigen Neutralization tests are the most specific and
Rapid diagnosis of influenza may be made by the best predictor of susceptibility to infection.
demonstration of the virus antigen on the surface of ELISA test is more sensitive than other assays.
the nasopharyngeal cells by immunofluorescence. It is possible to estimate the neuraminidase
This test is rapid but is not as sensitive as viral anti­body by enzyme neutralization tests.
isola­tion.
Detection of influenza RNA by reverse trans­ Immunity
criptase polymerase chain reaction may be more Immunity to influenza is long-lived and subtype­-
sensitive than antigen detection but is not widely specific. Antibodies against HA and NA are
available in diagnostic laboratories. important in immunity to influenza, whereas
antibodies against the other virus-encoded proteins
2. Isolation of the Virus are not protective.
Nasal washings, gargles and throat swabs are the An attack of influenza confers protection
best specimens for viral isolation and should be effective for about one or two years. The apparent
obtained within 3 days after the onset of symptoms short duration of immunity is due to the antigenic
but less often in later stages. Throat gar­glings var­iation that the antigenic variation that the virus
are collected using broth saline or other suita­ble undergoes frequently. Following infection and
buffered salt solution. The sample should be held at immunization, circulating antibodies are formed
4°C until inoculation into cell cul­ture, or if the delay against the various antigens of the virus. However,
is long, at –70°C. The specimen should be treated it the local concentration of anthaemagglutinin
with antibiotics to destroy bacteria. Isolation may and, to a smaller extent, of anineuraminadase
be made in eggs or in monkey kidney cell culture. antibodies (mainly IgA) in the respiratory tract that
The material is inoculated into the amniotic is more relevant in protection.
cavi­ty of 11–13 day-old eggs, using at least six eggs When an individual experiences repeated
per specimen. After incubation at 35°C for three infections with different antigenic variants of
days, the eggs are chilled and the amniotic and influenza virus type A, he responds by forming
allantoic fluids harvested separately. The fluids antibodies not only against each infecting strain
are tested for hemag­glutination using guinea pig but also against the strain that he first comes into
and fowl cells in parallel, at room temperature and contact with. The dominant antibody response
at 4°C. Some strains of the influenza virus type A will be against the strain that caused the earliest
agglutinate only guinea pig cells on initial isolation. infection. This phenome­non called the doctrine
The type B virus agglutinates both cells, while type of “original antigenic sin”.
C strains agglutinate only fowl cells at 4°C. Subtype Influenza virus infection induces cell-mediated
identification is made by hemagglutination immunity also but its role in protection has not
inhibition test. Some of the recent type A strains been clarified.
can be isolated by direct allantoic inoc­ulation of the
clinical specimen into 9–11-day-old eggs. However, Epidemiology
type Band C viruses will be missed if only allantoic Influenza viruses occur worldwide and occurs
inoculation is used. sporadically, as epidemics or in pandemic form.
For primary isolation the most suitable cells are The source of infection is an infected individual.
primary monkey kidney or human embryo kidney Influenza infection is spread readily via small
cells, but most labora­tories now use secondary airborne droplets expelled during talking,
baboon kidney cells or Madin–Darby canine breathing, and coughing. Influenza C is least
kidney cells. Incubation at 33°C in roller drum significant; it causes mild, sporadic respiratory
is recommended. The presence of virus may be disease but not epidemic influenza. Influenza B
detected by hemadsorption with human O group, sometimes causes epidemics, but influenza type A
fowl or guinea­pig red blood cells. Rapid results can sweep across continents and around the world
can be obtained by demonstrating virus antigen in massive epidemics called pandemics.
in infected cell cultures by immunofluoroscence. What makes influenza an important and
challenging disease is its propensity for causing
3. Serology pandemics. Domestic birds like ducks can get
Complement fixation tests (CFTs) and hemaggluti­ infected from wild birds and carry the infection to
nation inhibition (HI) tests are employed for the pigs.The genetic reassortment may take place in
serological diagnosis of influenza. Paired acute pigs that have receptors for both human and avian

www.ebook3000.com
438  |  Section 4: Virology
and rimantadine which block the viral M2 protein
which functions as an ion channel. These act only
with type A virus and not with type B, which lacks
the M2 components.

B. Influenza Vaccines
Influenza vaccines have been in use for many
decades. The aim of immunization is to produce
hemagglutina­t ion inhibiting or neutralizing
antibody in all vaccinees. However, cer­t ain
characteristics of influenza viruses make prevention
and control of the disease by immunization
especially difficult. Existing vaccines are continually
being ren­dered obsolete as the viruses undergo
antigenic drift and shift. Vaccines are of two types
as follow.

a. Inactivated Viral Vaccines


Fig. 63.2: Sequential changes in influenza A antigens Vaccines are either whole virus (WV), subvirion
associated with antigenic shift. Antigenic drift occurs after (SV), or surface antigen preparations.
the appearance of a new subtype
Whole virus (WV) vaccine: Inactivated vaccines
strains, and may act as a mixing vessel from which are prepared from appropriate strains of influenza
certain subtypes may transmit to humans and such A and B grown in the chick embryo allantoic cavity.
hybrid strains may lead to human infection with Subunit vaccines: The H and N antigens(purified
potential pandemic spread. HA and NA) may be separated from the whole virus
Influenza pandemics have been recorded at by treatment with ether and detergent, and these
irregular intervals from 1173. Pandemics of modern subunit or split-virus vaccines are better tolerated,
times date from 1889. The most severe pandemic in especially in young children.
recorded history occurred in 1918–1919 (‘Spanish
flu’). India suffered the most, with some 10 million
deaths. The next pandemic occurred in 1957 when Indications
the ‘Asian strain’ H2N2 originated in China and 1. Annual influenza vaccination is recommended
spread throughout the world. The Hong Kong H3N2 for high-risk groups: These include individuals
strain appearing in 1968 also caused a pandemic. at increased risk of complications associated
Figure 63.2 lists the sequence of appearance of with influenza infection.
these various subtypes. 2. Pandemic threat : The most important
Cases of influenza have appeared in Mexico indication for immunoproph­ylaxis is when a
and then in USA in April 2009. These were first pandemic is threatened by a new virus.
thought to be due to swine influenza A virus (H1N1) Contraindication: The only contraindication
but it had been identified as a new HINI virus which to vaccination is a history of allergy to egg protein.
was different from swine influenza virus. It can be
transmitted from human to human and spread to b. Live-virus Vaccines
many countries including India across the world. 1. The earliest live vaccine was the virus attenuated
WHO declared this situation as pandemic. On by repeated egg passage. It was admin­istered by
August 10, 2010, WHO announced the end of HINI intranasal instillation. However, it sometimes
pandemic and also declared that it has moved gave rise to clinical disease, especially in
into post pandemic period. However, localized children. These vac­cines have been generally
outbreaks of various magnitudes are likely to occur. effective in provoking a good local (IgA) antibody
More than 214 countries have reported laboratory response but are not at presently widely used.
confirmed cases including over 18,449 deaths as of 2. Use of temperature-sensitive mutants: A cold-
1st August 2010. This pandemic had occured 41 adapted donor virus, able to grow at 25°C but not
years after the last pandemic in 1968. at 37°C—the temperature of the lower respiratory
tract—should replicate in the nasopharynx,
Prophylaxis which has a cooler temperature (33°C). A live
A. Chemoprophylaxis attenu­ated, cold-adapted, trivalent influenza
Chemoprophylaxis has been reported to be virus vaccine administered by nasal spray has
successful with the antiviral drugs amantadine proved effective in clin­ical trials in children.
Chapter 63: Orthomyxovirus  | 439
Treatment 3. Write short notes on:
a. Hemagglutination
The antiviral drug amantadine and its analogue
rimantadine inhibit an uncoating step of the b. Antigenic shift
influenza A virus but do not affect the influenza c. Antigenic drift
B or C virus. The target for their action is the M2 d. Laboratory diagnosis of inflenza
protein. Resistance to these drugs develops rapidly. e. Prophylaxis against influenza.
Zanamivir and oseltamivir inhibit both influ­
enza A and B as enzyme inhibitors of the neuramini­ Multiple choice questions (mcqs)
dase. Zanami­vir is inhaled, whereas oseltamivir is 1. The influenza virus that is not responsible for
taken orally as a pill. These drugs are effective pandemics or epidemics is:
for prophylaxis and for treatment during the first a. Influenza virus A
24–48 hours after the onset of influenza A illness. b. Influenza virus B
Treatment cannot prevent the later host-induced c. Influenza virus C
immunopathogenic stages of the disease. d. None of the above
2. All the following statements are true for influenza
Key Points viruses except that:
a. They are spherical or filamentous, enveloped
™™ The family Orthomyxoviridae comprises four genera:
influenza A, B and C viruses and thogotoviruses
particles, 80–120 nm in diameter
™™ The influenza viruses are of three types: A, B and C b. The virion contains an RNA-dependent RNA
™™ The antigens of the influenza virus can be classified polymerase
as the internal antigens and the surface antigens c. Viral genome is a single-stranded unsegmented
(Hemagglutinin (HA) and Neuraminidase (NA) RNA
™™ Antigenic drift is the gradual se­quential change in d. The virion has hemagglutinin and neuramini-
antigenic structure occurring reg­ularly at frequent dase peplomers
intervals and is due to mutation and selection. 3. Antigenic drift in influenza viruses results from
Antigenic drift accounts for the periodical epidemics mutations in:
of influenza
a. Haemagglutinin genes
Antigenic shift is an abrupt, drastic, discontinuous
variation in the antigenic structure, resulting in b. Neuraminidase genes
a novel virus strain unrelated antigenically to c. Both the above
predecessor strains d. None of the above
™™ Influenza virus is transmitted from person to person, 4. Which of the following can be used for isolation of
primarily by air­borne respiratory droplets influenza virus from clinical specimens:
Adults: classic flu syndrome a. Madin-Darby canine kidney cell lines
™™ Laboratory diagnosis depends on demonstration b. Monkey kidney cell cultures
of virus antigens, isolation of the virus and serology
c. Amniotic cavity of chick embryo
™™ Influenza virus vaccines have been used to prevent
influenza, primarily influenza A and B. Vaccines are d. All the above
either inactivated viral vaccines or live virus vaccines 5. All the following statements are true for the
™™ Amantadine, rimantadine, zanamivir, and oseltamivir treatment of influenza by amantadine except:
(Tamiflu) have been approved for prophylaxis or a. It is effective against both influenza A and B
early treatment. b. It reduces severity of the disease
c. It hastens the disappearance of fever and other
Important questions symptoms
d. Emergence of viral resistance can occur during
1. Tabulate the differences between orthomyxoviruses treatment
and paramyxoviruses.
2. Discuss the morphology and pathogenesis of Answers (MCQs)
influenza virus infection. 1. c; 2. c; 3. c; 4. d; 5. a

www.ebook3000.com
64
Chapter

Paramyxoviruses

Learning Objectives

After reading and studying this chapter, you should ∙∙ D


 escribe the following: Para influenza viruses;
be able to: Measles virus; Respiratory syncytial virus (RSV)
∙∙ Classify paramyxoviruses ∙∙ Discuss pathogenicity and complications of measles
∙∙ Describe morphology of paramyxoviruses ∙∙ Discuss measles, mumps and rubella (MMR) vaccine.

Introduction associated with the nucleoprotein (NP), polymerase


phosphoprotein (P), and large (L) protein. Unlike
Family Paramyxoviridae causes a variety of the or­thomyxoviruses, the par­amyxoviruses with
diseases, predominantly involving the respiratory their unsegmented genome do not undergo genetic
tract, in humans, birds, and other animals. recombination’s or antigenic varia­tions. Hence, all
In humans, they include measles, respira­t ory paramyxoviruses are antigenically stable.
infections caused by respiratory syncytial virus The nucleocapsid associates with the matrix
(RSV) and parainfluenza viruses, and the more (M) protein at the base of the lipid envelope. The
innocuous salivary gland infection of mumps. virion envelope contains two glycoproteins, a fusion
(F) protein, which promotes fusion of the viral
Morphology and structural and host cell membranes, and a viral attachment
proteins of Paramyxoviruses protein (hemagglutinin–neuraminidase [HN],
Paramyxoviruses resemble orthomyxoviruses in hemagglutinin [H], or G protein). The F or fusion
morphology but are larger and more pleomorphic. protein, mediates the fusion of infected cells which
(Fig. 64.1). They are roughly spherical in shape and then form the syncytia so characteristic of infections
range in size from 100 to 300 nm, sometimes with with this group of viruses.
long filaments and giant forms of up to 800 nm.
The helical nucleo­capsid is much wider than in Classification
orthomyxoviruses, with a diameter of 18 nm (except Within the family Paramyxoviridae two subfamilies
in Pneumovirus where it is 13 nm). The nucleocapsid (Table 64.1). Paramyxoviridae and Pneumovirinae
consists of the negative-sense; sin­gle-stranded RNA are recog­nized.

Subfamily Paramyxoviridae
1. Respirovirus (para-influenza viruses 1, 3).
2. Rubulavirus (mumps virus, para-influenza
viruses 2, 4a, 4b).
3. Morbillivirus (Measles).
4. Henipavirus—Nipah virus and Hendra virus.

Subfamily Pneumovirinae
1. Pneumovirus (respiratory syncytial virus (RSV)
Fig. 64.1: Schematic diagram of a paramyxovirus showing 2. Metapneumovirus—Human metapneumo­
major components virus.
Chapter 64: Paramyxoviruses  | 441
Table 64.1:  Characteristics of genera in the subfamilies of the family Paramyxoviridae
Paramyxovirinae Pneumovirinae
Property Respirovirus Rubulavirus Morbillivirus Henipavirus 1
Pneumovirus Metapneumovirus
Human Parainfluenza Mumps, Measles Hendra, Respiratory Human
viruses 1,3 parainfluenza Nipah syncytial metapneumovirus
2, 4a, 4b virus
Serotypes 1 each 1 each 1 ? 2 ?
Diameter of 18 18 18 ? 13 13
nucleocapsid
(nm)
Membrane + + + + + +
fusion
(F protein)
Hemolysin2 + + + ? 0 0
Hemaglutinin +3 +3 +4 0 0 0
Hemadsorption + + + 0 0 0
Neuraminidase +3 +3 0 0 0 0
Inclusions 5
C C N, C ? C ?
1
Zoonotic paramyxoviruses
2
Hemolysin activity carried by F glycoprotein
3
Hemagglutination and neuraminidase activities carried by HN glycoprotein
4
Hemagglutination of monkey erythrocytes only, by H glycoprotein that lacks neuraminidase activity
5
C, cytoplasm; N, nucleus

Differentiation of Genera Primary infections in young children usually


Para­influenza viruses, NDV and mumps virus result in rhinitis and pharyngitis, often with fever and
have a surface hemagglutinin and neuraminidase, some bronchitis. However, children with primary
while measles virus has a hemagglutinin but no in­fections caused by parainfluenza virus type 1, 2, or
neuraminidase, and pneu­movirus has neither. 3 may have serious illness, ranging from laryngotra­
In addition, measles virus has a haemolysin not cheitis and croup (particularly with types 1 and 2)
possessed by the others, while RS virus has a large to bronchiolitis and pneumonia (particularly with
surface glycoprotein, G, which has a similar cell- type 3). The severe illness associated with type 3
attaching function as a hemagglutinin (Table 64.1). occurs mainly in infants under the age of 6 months.
Unlike the orthomyxoviruses, the paramyxoviruses Croup or laryngotracheobronchitis is more likely to
with their unsegmented genome cannot exchange occur in older children.
genetic information by recombination and variation Parainfluenza virus type 4 does not cause
depends on mutational change. serious disease, even on first infection.

Parainfluenza viruses Laboratory Diagnosis


Parainfluenza viruses are respiratory viruses that A. Direct Identification
usually cause mild cold like symptoms but can
Immunofluorescence or ELISA: Antigens may
also cause serious respi­ratory tract disease. There
be detected in exfoli­ated nasopharyngeal cells by
are four types of para-influenza viruses (1–4) with
immunofluorescence or ELISA.
antigenically distinct epitopes.

Pathogenesis B. Virus Isolation


Paramyxoviruses are acquired by droplets and Throat and nasal swabs are inoculated in primary
contact with respiratory secretions. Incubation monkey kidney cell cultures or continuous
period varies from 2–6 days. monkey kidney cell lines (LLC-MK2) with trypsin.
Parainfluenza viruses 1, 2, and 3 may cause Parainfluenza viruses grow slowly and produce very
respiratory tract syndromes ranging from a mild little cytopathic effect. Virus growth is detected by
coldlike upper respiratory tract infection (coryza, hemadsorption. Typing is by immunofluorescence,
pharyngitis, mild bronchitis, wheezing, and fever) to he­madsorption inhibition, or hemagglutination
bronchiolitis and pneumonia. inhibition.

www.ebook3000.com
442  |  Section 4: Virology
C. Serology variation. Immunity is permanent after a single
Serodiagnosis should be based on paired sera and infection.
can be tested by neu­tralization, enzyme linked
immunosorbent assay (ELISA) or Hemagglutination Laboratory Diagnosis
inhibition test (HI) or complement fixation test for Laboratory studies are not usually required to
rise in titre of antibodies. establish the diagnosis of typical cases.
The diagnosis may be established by virus
Genus Rubulavirus isolation and serological tests.
Mumps Virus A. Direct Demonstration
Mumps is an acute contagious disease commonly Direct demonstration by immunofluorescence on
affecting children character­ized by nonsuppurative secretions is very rarely successful.
enlargement of one or both parotid glands.
Morphology: Mumps virus is a typical paramyxo­ B. Virus Isolation and Identification
virus; possess both hemagglutinin and neuramini­ Virus can be recovered from the saliva and CSF.
dase (HN) or a fusion (F) protein. The envelope Monkey kidney cells are preferred for viral isola­
also contains a matrix (M) protein. It agglutinates tion. For rapid di­agnosis, immunofluorescence
the erythrocytes of fowl, guinea pig, humans and using mumps-specific antiserum can detect
many other species. Hemagglutination is followed mumps virus antigens as early as 2–3 days after the
by hemo­lysis and elution at 37°C. inoculation of cell cultures in shell vials.
Isolation can also be made by inoculation into
Pathogenesis six to eight day old chick embryos by the amniotic
Transmission is from person to person by large route and testing the amniotic fluid after 5–6 days
droplets. Primary replication occurs in nasal or for hemagglutinins. The virus can be identified
upper respiratory tract epithelial cells. Virernia by hemagglutination inhibition using specific
then dissem­inates the virus to the salivary glands antisera.
and other major organ systems and infects the
parotid gland. The virus is spread by the viremia C. Serology
throughout the body to the testes, ovary, pancreas,
Enzyme linked immunosorbent assay (ELISA) or
thyroid, and other organs. Infection of the central
hemagglutination inhibition test is commonly used.
nervous system, especially the meninges, immunity
Mumps specific IgM antibodies can be detected in
is life­long.
serum by enzyme linked immunosorbent assay
Clinical features: Infection is acquired by inhalation, (ELISA) for rapid diagnosis.
and probably also through the conjunctiva. The
incubation period may range from 7 days to 25 days Prophylaxis
but is typically about 16–18 days. The generalized
1. Vaccination
phase is the usual ‘flu-like’ illness with fever
and malaise, followed by developing pain in the An effective attenuated live Mumps virus vaccine
parotid glands, which then swell rapidly. Parotitis is based on the Jeryl Lynn or Urabe strains. Mumps
nonsuppura­tive and usually resolves within a week. vaccine is available in monovalent form (mumps
However, involvement of the extraparotid sites may only) or as combined vaccine, viz. combined
be more serious. mumps-rubella (MR) or into a triple vaccine
against measles, mumps and rubella (MMR) live-
Complications: Epididymo-orchitis, aseptic
virus vaccines. The vaccine is recommended for
meningitis, post­infection encephalitis, mumps
children over age 1 year.
meningitis. Other less common complications
are arthritis, oophoritis, nephritis, pancreatitis, The vaccine is given as a single dose (0.5 mL)
thyroiditis and myocarditis. intramuscularly. It provides effective protection for
at least ten years. Contraindications are pregnancy,
Epididymo-orchitis: Is a complication seen in immunodeficiency, severely ill and hypersensitivity
about a third of postpubertal male patients and to neomycin or egg protein.
rarely causes sterility.
2. Immunoglobulin
Immunity A specific immunoglobulin (mumps immuno­
There is only one antigenic type of mumps virus, globulin) is available and is of no value either for
and it does not exhibit significant antigenic postexposure prophylaxis or treatment.
Chapter 64: Paramyxoviruses  | 443
Complications
1. Complications may be due to the virus
(croup, bronchitis) or to secondary bacterial
infection (pneumonia, otitis media). Giant
cell pneumonia, particularly in children with
immunodeficiencie’s or severe malnutrition.
2. Post-measles encephalitis.
3. Subacute sclerosing panencephalitis (SSPE)
Is an extremely serious, very late neurologic
sequel of mea­sles. It occurs in children or
early adolesents who have measles early in life,
usually less than 2 years of age. This disease
Fig. 64.2: Schematic diagram of measles virus occurs when a defective measles virus persists
Genus morbillivirus in the brain and acts as a slow virus. The virus
can replicate and spread directly from cell to
Measles (Rubeola) cell but is not released. Within infected cells
Measles is one of the five classic childhood exant­ is a defective form of measles virus, which,
hems, along with rubella, roseola, fifth disease, and because it is unable to induce the production
chicken­pox. of a functional M protein, is not released as
complete virus from the cell.
Morphology 4. Protracted diarrhea in children in the poor
nations.
The virus has the general morphology of paramyxo­
viruses (Fig. 64.2). It is a roughly spherical but often
Laboratory Diagnosis
pleomorphic particle, 120–250 nm in diameter. The
tighdy coiled helical nucleocapsid is surrounded Typical measles is reliably diagnosed on clinical
by the lipoprotein envelope carrying on its surface grounds, usually in the patient’s home. Laboratory
hemagglutinin (H) spikes. The envelope also diagnosis may be necessary in cases of modified or
has the F protein which mediates cell fusion and atypical measles.
hemolytic activities. The measles virus agglutinates
monkey erythrocytes but there is no elution as the A. Demonsration of Virus Antigen
virus does not possess neuraminidase activity. A simple diagnostic test, which can be used even
before the rash appears, is the demonstration of
Pathogenesis multinucleated giant cells in Giemsa stained smears
The virus gains access to the human body via the of nasal secretions. The measles virus antigen can
respiratory tract, or the conjunctiva where it multiplies be detected in these cells by immunofluorescence.
locally. The in­fection then spreads to the regional
lymphoid tissue, where further multiplication occurs. B. Virus Isolation
Primary viremia disseminates the virus, which then The virus can be isolated from the nose, throat,
replicates in the reticuloendothelial system. Finally, conjunctiva and blood during the prodromal phase
a secondary viremia seeds the epithelial surfaces and upto about two days after the appearance of the
of the body, in­cluding the skin, respiratory tract, and rash. The virus may be obtained from the urine for
conjunctiva, where focal replication occurs. a few more days. Primary human or monkey kidney
and amnion cells are most useful. Cytopathic
Clinical Features changes may take up to 7–10 days to develop
Measles is a serious febrile illness. Incubation but earlier diagnosis of viral growth is possible
pe­riod is 9–11 from the time of exposure to infection by immunofluorescence. Typical cytopathic
for the first signs of clinical disease to appear. After effects (multinucleated giant cellscontaining
2 days of illness, the typical mucous mem­brane both intranuclear and intracytoplasmic inclusion
lesions, known as Koplik’s spots, appear most bodies) suggests the presence of measles virus.
commonly on the buccal mucosa across from the
molars. Their appearance in the mouth establishes C. Serological Diagnosis
with cer­tainty the diagnosis of measles. Demonstration of measles-specific IgM in a single
Before the rash there is a prodromal stage specimen of serum drawn between one and two
lasting 2 or 3 days, with high fever and CCC and weeks after the onset of the rash is confirmatory.
P-cough, co­ryza, and conjunctivitis, in addition to Enzyme linked immunosorbent assay (ELISA),
photophobia. hemagglutination inhibition (HI), and neutralization

www.ebook3000.com
444  |  Section 4: Virology
(Nt) tests all may be used to measure measles resulting in at least one fatal infection in human
antibodies, though ELISA is the most practical in Australia.
method. A four fold rise in titer is diagnostic. The taxonomic position of these new viruses
High titer measles antibody in the CSF is has yet to be established, but they will most
diagnostic of SSPE. probably be put in a sep­arate genus within the
Paramyxoviridae. Fruit bats appear to be the
Epidemiology natural reservoir of both viruses, with transmis­sion
The key epidemiologic features of measles are as to mammals (including man) an exceptional event.
follows: the virus is highly contagious, there is a
sin­gle serotype, there is no animal reservoir, in Genus pneumovirus
apparent infections are rare, and infection confers Respiratory Syncytial Virus (RSV)
lifelong im­munity. Transmission is person-to-
person, probably by respir­atory droplets, but the Respiratory syncytial virus (RSV) first isolated from
associated conjunctivitis may also be a source. In a chimpanzee was named respiratory syncytial
gen­eral, epidemics recur regularly every 2–3 years. virus (RSV) because it caused cell fusion and the
formation of multinucleated syncytia in cell cultures.
Prophylaxis
Description
A. Passive immunization
RSV is pleomorphic and ranges in size from 150­–
Passive protection with normal immunoglobulin 300 nm. The viral envelope has two glycoproteins—
(NHIG) given within six days of exposure can the G protein by which the virus attaches to
prevent or modify the disease, depending on the cell surfaces, and the fusion (F) protein which
dose. Passive immunization is recommended in brings about fusion between viral and host cell
children with immunodeficiency, pregnant women membranes. The F protein is also responsible for
and others at risk. cell-to-cell fusion, which leads to the characteristic
syncytial cytopathic changes in RSV infection.
B. Active Immunization
It is placed in a separate genus—Pneumovirus
A highly effective, safe, attenuated live measles
because of these minor physical differences (Table
virus vaccine is available. The original live vaccines
64.1).
used the Edmonston strain developed by multiple
RSV does not grow in eggs but can be propagated
passage through human kidney, amnion and chick
on heteroploid human cell cultures, such as HeLa
embryo cultures. The Schwartz and Moraten
and Hep-2. It is highly labile and is inactivated
strains so developed were safe but effective only
rapidly at room temperature. It is antigenically stable
in children older than 15 months. In the tropics,
and for most purposes there is only one serotype.
measles is common and serious in children below 12
months. Recent observations have suggested that the
Edmonston–Zagreb strain, attenuated by passage in Clinical Features
human diploid cells, may protect children from 4 to The spectrum of respiratory illness ranges from the
6 months of age. The recommended age for measles common cold in adults, through febrile bronchi­tis
vaccination in the developing countries is 9 months, in infants and older children and pneumonia in
while in the advanced countries it remains 15 months. in­fants, to bronchiolitis in very young babies.
The vaccine is given either by itself, or in 1. Common cold: In adults RSV infection may
combination as the MMR vaccine. A single present as a febrile common cold. It can cause
subcutaneous injection of the measles vaccine pneumonia in the elderly.
provides protection beginning in about 12 days 2. Febrile rhinitis and pharyngitis
and lasting for over 20 years, probably for life. 3. Bronchiolitis, pneumonia, or both: The most
Contraindications are pregnancy, acute illnesses, serious illness caused by RS virus are bronchi­
immunodeficiency and untreated tuberculosis. olitis in young babies.
A live attenuated vaccine has been developed 4. Sudden infant death syn­drome: A relation
which can be given by intranasal aerosol in young between RSV and the sudden death syndrome
babies and gives good protection irrespective of the in infants has been proposed but not proven.
presence of maternal antibodies.
Epidemiology
Nipah and Hendra viruses Respiratory syncytial virus is distributed world­wide
Nipah virus is distinct genetically from all the other and is recognized as the major pediatric respira­tory
paramyxoviruses and is most closely related to tract pathogen. It is the most com­mon cause of viral
Hendra, causing epidemic fatal res­piratory disease pneumonia in children under age 5 years. RSV is
in horses and which can be transmitted to man, highly contagious.
Chapter 64: Paramyxoviruses  | 445
RSV infections almost always occur in the ™™ Within the family Paramyxoviridae two subfamilies-
winter.It causes nosocomial in­fections in nurseries Parmyxoviridae and Pneumovirinae are recog­nized
and on pediatric hospital wards. ™™ Parainfluenza viruses 1, 2, and 3 may cause respiratory
tract syndromes ranging from a mild coldlike upper
Laboratory Diagnosis respiratory tract infection to bronchiolitis and
pneumonia. Parainfluenza virus type 4 does not
A. Demonstration of Virus Antigen cause serious disease, even on first infection
Immunofluorescence and enzyme immunoas­say ™™ Mumps virus causes mumps
tests are available for direct detection of the viral ™™ Measles virus causes measles, atypical measles, and
subacute sclerosing panencephalitis (SSPE)
antigen in infected cells and nasal washings.
™™ Measles vaccine is a live attenuated vaccine which
now uses Schwartz and Moraten attenuated strain
B. Virus Isolation of the original Edmonston B strain
Human heteroploid cell lines HeLa and HEp-2 are ™™ Measles vaccine along with mumps and rubella
the most sensitive for viral isolation. The presence of (MMR) vaccine is currently used for universal
immunization of the children
respiratory syncytial virus can usu­ally be recognized
™™ Respiratory syncytial virus (RSV) causes infection
by development of giant cells and syncytia in of the respiratory tract, ranging from the common
inoculated cultures but cytopathic effects may take cold in adults, through febrile bronchi­tis in infants
as long as 10 days for to appear. Definitive di­agnosis and older children and pneumonia in in­fants, to
can be established by detecting viral antigen in bronchiolitis in very young babies.
infected cells using a defined antiserum and the
im­munofluorescence test. Important questions
C. Serology 1. Classify and discuss the morphology of paramyxo­
Serological diagnosis is by demonstration of viruses.
rising antibody titers in paired serum samples by 2. Write short notes on:
enzyme linked immunosorbent assay (ELISA), a. Parainfluenza viruses
b. Mumps virus
complement fixation (CF), neutralization or
c. Measles virus
immunofluorescence tests.
d. Respiratory syncytial virus (RSV)
e. Measles, mumps and rubella (MMR) vaccine.
Prophylaxis
f. Subacute sclerosing panencephalitis (SSPE)
No vaccine is currently available for RSV prophy­ g. Atypical measles.
laxis. The use of recombinant DNA technology for
making RSV vaccine is now being studied.
Multiple choice questions (mcqs)
Treatment 1. The paramyxovirus virus that possesses a fusion
In otherwise healthy infants, treatment is protein but lacks both hemagglutinin and neura­
supportive care. Ribavirin is administered by minidase activities is:
in­halation (nebulization) and has been found a. Mumps virus
b. Measles virus
beneficial in hospitalized patients, decreasing the
c. Parainfluenza virus
duration of illness and of virus shedding. d. Respiratory syncytial virus
2. Which of the following conditions can be caused
Metapnemovirus by infection with mumps virus?
Human metapneumovirus is a respiratory a. Swelling of parotid glands
pathogen and is an important cause of respiratory b. Orchitis
tract infection in children. It can also cause disease c. Meningoencephalitis
in adults. It causes a disease similar to that of d. All of the above
human respiratory syncytial virus. 3. Koplik’s spots are characteristic of:
It is a single stranded RNA virus like other a. Mumps
paramyoviruses. Respiratory secretions are clinical b. Measles
specimen for test. The virus is difficult to grow. c. Herpes
Polymerase chain reaction (PCR) can be used d. Rubella
for diagnosis. No specific antiviral treatment or 4. MMR vaccine is:
vaccine is available. a. a live attenuated vaccine
b. a killed vaccine
Key Points c. a subunit vaccine
d. a synthetic peptide vaccine
™™ Unlike the or­thomyxoviruses, the par­amyxoviruses
with their unsegmented genome do not undergo
Answers (MCQs)
genetic recombinations or antigenic varia­tions
1. d; 2. d; 3. b; 4. a

www.ebook3000.com
65
Chapter

Arboviruses

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ List the arboviruses prevalent in India
be able to: ∙∙ Describe the following: Chikungunya; Japanese B
∙∙ Classify arboviruses encephalitis; Dengue fever (or) Break bone fever;
∙∙ Describe laboratory diagnosis of arboviruses Kyasanur forest disease (KFD).
∙∙ List mosquito-borne and tick-borne arboviruses

INTRODUCTION Table 65.1:  Taxonomy of some important arbo­


Arboviruses (Arthropod-borne viruses) are viruses
define as viruses “which are maintained in nature Family Genus Important species
principally, or to an important extent through Togaviridae Alphavirus Eastern, Western and
biological transmission between susceptible Venezuelan equine
vertebrate hosts by hematophagous arthropods. encephalitis viruses,
They multiply in the tissues of arthropods, and Chikungunya, O’nyong-
are passed onto new vertebrates by the bites of nyong, Mayaro, Semliki
Forest, Sindbis, Ross River.
arthropod after a period of extrinsic incubation.
Certain viruses within the six families contain­ Flaviviridae Flavivirus St. Louis encephalitis,
ing arboviruses are not transmitted by arthropods, tiheus, West Nile, Murray
Valley encephalitis,
but are maintained in nature within rodent Japanese encephalitis,
reservoirs that may transmit infection directly to Yellow Fever, Dengue
humans. These include the Hantavirus genus of types 1, 2, 3, 4,
the family Bunyaviridae Kyasanur forest disease
Russian spring summer
CLASSIFICATION encephalitis complex,
Louping ill, Powassan.
Arboviruses are classified within five families (Table Omsk hemorrhagic fever
65.1). Most are members of the families Togaviridae, Bunyaviridae Bunyavirus California encephalitis,
Flaviviridae and Bunyaviridae. Within each family, Phlebovirus Oropouche, Turlock
they are­classified into genera, and antigenic Nairovirus Sandfly fever viruses, Rift
groups, based on serological relationships. valley fever virus
Hantavirus Crimean Congo hemor­
PROPERTIES rhagic fever viruses,
Nairobi sheep disease
Arboviruses share common biological attributes virus, Ganjam virus
(Table 65.2). Hantan, Seoul, Puumala,
1. Intracerebral inoculation in suckling mice is the Prospect Hill, Sin Nombre
viruses
most sensitive method for their isolation in the
laboratory. Reoviridae Orbivirus African horse sickness,
Coltivirus Blue tongue viruses
2. Most arboviruses agglutinate the red cells of
Colorado tick fever
goose or day-old chicks.
3. They can be grown in the yolk sac or chorio­ Rhabdoviridae Vesiculovirus Vesicular stomatitis virus,
Chandipura virus
allantoic membrane of chick embryo, in tissue

Chap-65.indd 446 15-03-2016 11:23:04


Chapter 65: Arboviruses  | 447
Table 65.2:  Properties of Arboviruses
Properties Alphavirus Flavivirus Bunyavirus Rhabdovirus Reovirus
1. Symmetry Cubic Cubic Helical Bullet-shaped Cubic
2. Size (Diameter in nm) 60–65 40–60 80–100 180 x 85 60–80
3. Nucleic acid Single stranded Single stranded Single stranded Single stranded Double
positive sense positive sense negative sense negative sense stranded RNA
RNA RNA RNA RNA
4. Inactivation by + + + + -
diethyl ether/sodium
deoxycholate

cultures of primary cells and in cul­tures of reverse transcriptase polymerase chain reaction
appropriate insect tissues. (RTPCR).
4. Many arboviruses multiply in continuous tissue
cultures of mosquito cells when incubated at D. Serology
34°C or lower tem­peratures. Diagnosis may also be made serologically by
5. Mosquito-borne arboviruses multiply after oral demonstrating rise in antibody titer in paired
feeding or intrathoracic injection of several serum samples by hemagglutination inhibition,
Aedes and Culex mosquito species. complement fixation or neutralization tests. Virus-
6. Tick-borne arboviruses multiply after oral specific IgM antibody may be detected within I day
feeding to larval or nymphal ixodes ticks. of onset of clinical symptoms using an IgM capture
7. In general, arboviruses are labile, being readily ELISA test.
inactivated at room temperature and by bile
salts, ether and other lipid solvents. PATHOGENESIS
The virus enters the body through the bite of the
LABORATORY DIAGNOSIS insect vector. After multiplication in the re­ticulo­
A. Specimen endothelial system, viremia of varying duration
ensues and, in some cases, the virus is transported
As all arbovirus infec­t ions are viremic, blood
to the target organs, such as the central nervous
collected during the acute phase of the disease may
system in encephalitides, the liver in yellow fever
yield the virus. Isolation may also be made from
and the capillary endothelium in hemorrhagic
the CSF in some encephalitic cases but the best
fevers. Arboviruses cause the following clinical
specimen for virus isolation is the brain.
syndromes (Table 65.3).
∙∙ Fever with or without rash and arthralgia
B. Virus Isolation ∙∙ Encephalitis
i. Suckling Mice ∙∙ Hemorrhagic fever
Specimens are inoculated intracerebrally into suck­ ∙∙ The characteristic systemic disease, yellow
ling mice. The animals develop fatal encephalitis. This fever
is the most sensitive method for isolation of viruses. Arboviruses are maintained in natural trans­
mission cycles involving reservoir hosts and
ii. Tis­sue Cultures arthropod vectors, typically: ticks, mosquitoes and
Some viruses may also be isolated in tis­s ue other biting flies.
cultures or, less readily, in eggs. Specimens are
inoculated into Vero, BHK-21 and mosquito cello FAMILIES OF ARBOVIRUSES
lines. Isolates are iden­tified by hemaglutination A. Family Togaviridae
inhibition, complement fixation, gel precipitation,
Togaviruses
immunofluorescence, im­munochromatography,
ELISA or neutralization with appropriate antisera. Morphology
Togaviruses are spherical enveloped viruses with
iii. Virus Isolation from Insect Vec­tors and a diameter of 50–70 nm. The genome is single
Reservoir Animal stranded RNA. The name Togavirus is derived from
‘toga’, meaning the Roman mantle or cloak, and
C. Arbovirus-specific RNA Detection refers to the viral envelope.
Viral RNA is extracted from serum or suspensions
of tissues from patients, or from tissue culture cells Classification
or mosquito homogenates. This is amplified by The family Togaviridae contains two genera:

www.ebook3000.com
Chap-65.indd 447 15-03-2016 11:23:04
448  |  Section 4: Virology
Table 65.3:  Arboviruses associated with different clinical syndromes
Family Genus Virus Vector Geographic Reservoir
distribution
Fever with or without rash and arthralgia
Togaviridae Alphavirus Chikungunya Mosquito Africa, Asia Not known (? Monkeys)
Alphavirus Onyong-nyong Mosquito Africa Not known
Alphavirus Ross River Mosquito Australia Small animals
Alphavirus Sindbis Mosquito Africa, Asia Birds, mammals
Alphavirus Mayaro Mosquito South America Monkeys, marsupials
Flaviviridae Flavivirus Dengue, types 1–4 Mosquito Widespread, Not known
especially (? Monkeys)
Asia Pacific,
Caribbean
Flavivirus West Nile Mosquito Asia, Africa, USA Birds
Bunyaviridae Phlebovirus Sandfly fever Sandfly Mediterranean, Asia, not known
Tropical America (? Small mammals)
Phlebovirus Rift valley fever Mosquito Americas Sheep, cattle
Bunyavirus Oropouche Mosquito South America Not known
Reoviridae Orbivirus Colorado tick fever Tick USA Rodents
Encephalitis
Togaviridae Alphavirus Eastern equine Mosquito Americas Birds
encephalitis
Alphavirus Western equine Mosquito Americas Reptiles (? Birds)
encephalitis
Alphavirus Venezuelan equine Mosquito Americas Rodents
Flaviviridae Flavivirus St. Louis encephalitis Mosquito Americas Birds
Flavivirus West Nile Mosquito Africa, Europe, USA, Birds
West Asia
Flavivirus Japanese encephalitis Mosquito East and South East Birds
Asia
Flavivirus Murray valley Mosquito Australia Birds
encephalitis
Flavivirus RSSE complex Tick East Europe, USSR Rodents, other
mammals, birds, ticks
Flavivirus Louping ill Tick Britain Sheep
Flavivirus Powassan Tick North America Rodents
Bunyaviridae Bunyavirus California Mosquito North America Rodents
Hemorrhagic fever
Togaviridae Alphavirus Chikungunya Mosquito Africa, Asia Not known
(? Monkeys)
Flaviviridae Flavivirus Dengue types 1–4 Mosquito Tropics Not known
(? Monkeys)
Flavivirus Yellow fever Mosquito Africa, South America Monkeys, man
Flavivirus Kyasanur forest Tick Southwest India Rodents (? Ticks)
disease
Flavivirus Omsk hemorrhagic Tick USSR Small mammals
fever
Flavivirus Crimean Congo Tick USSR, Central Asia, Small mammals
hemorrhagic fever Africa

Alphavirus Rubivirus
The Alphavirus genus consists of about 32 viruses Rubivirus, which is not arthropod-borne and which
of which at least 13 are known to infect humans. causes rubella.
All of them are mosquitoborne.

Chap-65.indd 448 15-03-2016 11:23:04


Chapter 65: Arboviruses  | 449
Viruses of Togaviridae 3. Semliki Forest Virus
1. Alphavirus (Mosquito -borne) This virus has not been associated with clinical
A. Encephalitis viruses illness in humans though neutralizing antibodies
i. Eastern equine encephalitis (EEE) to the virus have been demonstrated in Aricans.
ii. Western equine encephalitis (WEE)
iii. Venezuelan equine encephalitis (VEE) 4. Sindbis Virus
B. Viruses causing febrile illness The Sindbis virus, originally’ isolated from Culex
mosquitoes in the Sindbis district of Egypt in 1952,
i. Chikungunya virus (CHIKV)
has subsequently been recovered from other parts
ii. Onyong-nyong virus (ONNV) of Africa, India, Philippines and Australia. In Africa,
iii. Semliki Forestvirus it is known to be associated with febrile illness
iv. Sindbis virus in human beings. In India, antibodies have been
v. Ross river virus detected in human sera but no association has
been established with human disease.
Rubivirus
Rubella virus.
5. Ross River Virus
1. Alphavirus
The closely related Ross River virus has been
Encephalitis viruses: Three members of this
associated with epidemic polyarthritis in Australia.
group, Eastern, Western and Venezuelan equine
encephalitis viruses, cause encephalitis in horses
and humans. B. Family Flaviviridae
Reservoirs: Several species of Culex and Anopheles The family Flaviviridae contains three genera:
mosquitoes are the vectors, and wild birds the 1. Flavivirus; 2. Pestivirus; 3. Hepacivirus.
reservoirs.
Flavivirus
Vaccine: Formalinized vaccines have been
developed for EEE and WEE and a live attenuated The name Flavivirus refers to the type species,
vaccine for VEE. the yellow fever virus (Flavus, L = yellow). The
Flaviviridae family consists of about 70 viruses.
B. Viruses Causing Febrile Illness
Morphology
1. Chikungunya Virus
In Swahili, ‘chikungunya’ means ‘that which bends They are 40 nm in diameter. They contain a single
up’, and refers to the posture assumed by patients stranded positive sense RNA. Inner viral core is
suffering from severe joint pains. surrounded by a lipid envelope which is covered
with glycoprotein peplomers and matrix or
The incubation period is 1–12 days with an aver­
membrane protein.
age of 2–3 days. The disease presents as a sudden
onset of fever, crippling joint pains, lymphadenopathy They may be considered under two sections,
and conjunctivitis. A maculopapular rash is common the mosquito-borne and the tick-borne viruses
and some show hemorrhagic manifestations. (Table 65.3).

Vector: The vector is Aedes aegypti. No animal A. Mosquito-borne Group


reservoir has been identified.
a. Encephalitis viruses: St. Louis encephalitis,
In India the virus first appeared in 1963 and
Ilheus, West Nile, Murray Valley encephalitis,
caused very extensive epidemics in Calcutta,
Japanese encephalitis.
Madras and other areas. Chikungunya outbreaks
b. Yellow Fever
occurred at irregular intervals along the east coast
c. Dengue types 1, 2, 3, 4.
of India and in Maharashtra till 1973.
B. Tick-borne Group
2. Onyong-nyong Virus
These viruses produce—two clinical syndromes,
Onyong-nyong virus (ONNV), is confined to
encephalitis and hemorrhagic fevers.
Africa, is closely related to the chikungunya virus
antigenically and causes a similar disease.
This is transmitted by the Anopheles species A. Mosquito-borne Group
(Anopheles funestus and Anopheles gambiae). The a. Encephalitis viruses: Five members of this
Mayaro virus causes a similar disease in the West group cause encephalitis, each of them limited to
Indies and South America. a geographic zone.

www.ebook3000.com
Chap-65.indd 449 15-03-2016 11:23:04
450  |  Section 4: Virology
1. St. Louis encephalitis virus (SLEV): This is of the disease in man may be divided into three
prevalent in North and Central America and is the stages:
most important mosquito-borne disease in the a. Prodromal stage: The onset of illness is usually
USA. acute and is heralded by fever, headache, and
Wild birds act as the reservoir and Culex tarsalis malaise.
as the vector. No vaccine is yet available. b. Acute encephalitic stage: The prominent
2. Ilheus virus: This occurs in South and Central features are fever, nuchal rigidity, focal central
America, maintained in forests by a cycle involving nervous system (CNS) signs, convulsions and
mosquitoes, wild birds and monkeys. Human altered sensorium progressing in many cases
infection is largely subclinical or leads to febrile to coma.
illness. Encephalitis is rare. c. Late stage and sequelae: Convalescence may
be prolonged and residual neurological deficits
3. West Nile virus (WNV): WNV was first isolated may not be uncommon. The case fatality rate
from a febrile human in the West Nile district of varies between 20 and 40%.
Uganda in 1937, has since been reported from many The large majority of infections are, however,
African countries, Israel, Cyprus, France and India. asymptomatic.
Vector—both mosquitoes and ticks have been
reported as vectors. The principal vectors are Epidemiology: Unlike the dengue viruses, JE virus
considered to be mosquitoes of the Culex genus. infects several extrahuman hosts, e.g. animals and
birds. The disease is transmitted to man by the bite
4. Murray valley encephalitis virus (MVEV): This of infected mosquitoes. Available evidence indicates
is confined to Australia and New Guinea. Vector- that the basic cycles of transmission are:
Natural cycles of transmission of MVEV involve Cx.
a. Pig → Mosquito → Pig
annulirostris as the principal mosquito vector and
water birds as reservoirs. b. The Ardeid bird → Mosquito → Ardeid bird

5. Japanese encephalitis: Japanese encephalitis a. Animal host: Pigs considered as “amplifiers” of


(JE) is a mosquito-borne encephalitis caused by a the virus.
group B arbovirus (Flavivirus) and transmitted by b. Birds: Natural infection has been demonstrated in
culicine mosquitoes. It is a zoonotic disease, i.e., Ardeid birds (herons and egrets), as well as bird-to-
infecting mainly animals and incidentally man. bird transmission through Culex tritaeniorhynchus.
Culex tritaeniorhynchus, a rural mosquito that
breeds in rice fields, is the principal vector. Vectors of JE: Culicine mosquitoes, notably C.
tritaenior­hynchus, C. vishnui and C. gelidus have
Geographical Distribution been incriminated as the vectors of JE.

The disease has been recognised in Japan since Control of Japanese Encephalitis
1871 and was named Japanese ‘B’ encephalitis to
distinguish it from ‘encephalitis A (encephalitis Preventive measures include mosquito control and
lethargica, von Economo’s disease) which was locating piggeries away from human dwellings.
then prevalent. The virus was first isolated in Japan
during an epidemic in 1935. i. Formalin Inactivated Mouse Brain Vaccine
A formalin inactivated mouse brain vaccine
Problem in India using the Nakayama strain has been employed
Recognition of JE was first made in 1955 when the successfully for human immunization in Japan
virus was isolated from mosquitoes of the Culex and, in a small scale, in India also. For primary
vishnui complex from Vellore. From 1976, there have immunization, 2 doses of 1 ml each (0.5 mL for
been periodical outbreaks of the disease in various children under the age of 3 years) should be
parts of India—Dibrugarh (Assam), Gorakhpur (Uttar administered subcutaneously at an interval of
Pradesh) and Haryana and Goa and Maharashtra, 7–14 days. A booster injection of 1 mL should be
Kolar in Karnataka, Andhra Pradesh, in Tamil given after a few months (before one year) in order
Nadu, in Pondicherry and lately in Kerala. Japanese to develop full protection. Protective immunity
encephalitis has become a major public health develops in about a month’s time after the second
problem of national importance in India. dose. Revaccinations may be given after 3 years.

Clinical Features ii. Live Attenuated Vaccine


The incubation period in man probably varies from A live attenuated vaccine has been developed in
5–15 days, following mosquito bite. The course China from JE strain SA 14-14-2, passed through

Chap-65.indd 450 15-03-2016 11:23:05


Chapter 65: Arboviruses  | 451
weanling mice. The vaccine is produced in forest or when the monkeys raid villages near
primary baby hamster kidney cells. Administered the forest.
in two doses, one year apart, the vaccine has been Yellow fever is largely confined to Central and
reportedly effective in preventing clinical disease. South America and Africa. Yellow fever has never
Vaccination of pigs: Vaccination of pigs has been invaded Asia, even though the vector. A. aegypti, is
proposed in view of their importance as amplifier widely distributed there. It is likely that stray virus
hosts. During major epidemics, slaughter of pigs introduced may have been kept out, due to the
have been employed as a measure of containment. prevalence in the local Aedes aegypti of Dengue virus,
and of antibodies to a wide range of Arboviruses in
the local population. Another reason could have
b. Yellow Fever
been that in Africa, yellow fever was mainly in the
Yellow fever virus is the prototype member of the west, and in India, Aedes mosquitoes were along the
Fla­viviridae family. lt causes yellow fever, an acute, east coast, so that even stray importations of virus
febrile, mosquito-borne illness that occurs only in by sea may not have found suitable vectors. Yellow
Africa and Central and South America. It does not fever does not exist in India and it is important to us
exist in India. for this paradoxical reason.

Pathogenesis and Pathology Control


The virus is introduced by a mosquito through the 1. Vector Control
skin and spreads to the local lymph nodes where 2. Vaccination:
it multi­plies. From the lymph nodes, it enters the
circulating blood and becomes localized in the a. French Neurotropic Vaccine (Dakar)
liver, spleen, kidney, bone marrow, and lymph
The infected mouse brain was used as a vaccine in
glands, where it may persist for days. The lesions
former French West Africa (Dakar vaccine) was
of yellow fever are due to the localization and
thermostable and administered by scarification
propagation of the virus in a particular organ.
and hence convenient for use under tropical field
Infec­tions may result in necrotic lesions in the liver
conditions. However, the vaccine carries a high
and kid­ney.
risk of producing encephalitis in the vaccines,
Histologically,the liver shows cloudy and especially in children. It was later replaced by a
fatty degeneration and necrosis which is typically non-­neurotropic (17D) vaccine.
midzonal. The necrosed cells coalesce and become
hyalinized leading to the formation of characteristic b. 17D Vaccine
eosinophilic masses known as Councilman bodies.
A safe and equally effective vaccine, the 17D
Acidophilic intranuclear inclusion bodies (Torres
vaccine was developed by Theiler in 1937 by
bodies) may be seen in the infected liver cells in
passaging the Asibi strain serially in mouse embryo
the early stages.
and whole chick embryo tissues and then in chick
embryo tissue from which the central nervous
Clinical Findings tissue has been removed and has been used as a
The incubation period is 3–6 days. The disease vaccine for over 40 years.
starts as a fever of acute onset with chills, headache, The 17D vaccine is thermolabile and is
nausea and vomiting. Jaundice, albuminuria, and administered by subcutaneous inoculation.
hemorrhagic manifestations develop and the Immunity develops within 10 days of vaccination.
patient may die of hepatic or renal failure. Vaccination which is mandatory for travel to or
from endemic areas is valid for 10 years beginning
Epidemiology 10 days after vaccination. Serious side effects are
Two major epidemiologic. Cycles of transmission rare with the 17D vaccine. In India, the 17D vaccine
of yel­low fever are recognized: is manufactured at the Central Research Institute,
1. Urban yellow fever: In the urban cycle, Kasauli.
humans at both as the natural reservoir, and as
the definitive case, the virus being transmitted c. Dengue
by the domestic Aedes aegypti mosquito. Dengue virus is widely distributed throughout
2. Forest or sylvatic cycle: in the forest or sylvatic the tropics and subtropics. The term ‘break-
cycle, wild monkeys act as the reservoirs and bone fever’ was coined during the Philadelphia
forest mosquitoes (Haemagogus spegazzinii epidemic in 1780.
in South America and Aedes africanus and A. Four types of dengue virus exist : DEN 1,
simpsoni in Africa) as the vectors. Human cases DEN 2, and DEN 3 and 4. Immunity is type
occur only when humans trespass into the specific. However, the febrile clinical symptoms

www.ebook3000.com
Chap-65.indd 451 15-03-2016 11:23:05
452  |  Section 4: Virology
associated with dengue are similar to those of other suckling mice. Further identification is done by
Arboviruses from other families. using fluorescent antibody test. Live mosquito
inoculation is the most sensitive technique for
Clinical Findings isolation of virus.
Classic dengue usually affects older children and B. Polymerase chain reaction (PCR): PCR ­based
adults.After incubation period of 2–7 days, pat­ient methods are available for rapid identification
develops fever of sudden onset and often bip­hasic and subtyping.
with severe headache, chilies, retrobulbar pain, C. Serology: Demonstration of circulating
conjunctivitis and severe pain in the back, muscles IgM antibody provides early diagnosis, as it
and joints (breakbone fever). A maculopapular appears within two to five days of the onset of
rash generally appears on the trunk in 3–5 days of illness and persists for one to three months.
ill­ness and spreads later to the face and extremities. IgM ELISA test offers reliable diagnosis. A
Lymph nodes are frequently enlarged. Leukopenia strip immunochromatographic test for IgM is
with a relative lymphocytosis is a regular occurrence. available for rapid diagnosis.
Complications and death are rare. IgG antibody appears later than IgM antibody.
ELISA is used for detection of IgG antibody.
Complications of Dengue: Dengue may also Detection of four fold rise in IgG titer in paired
occur in more serious forms, with hemorrhagic sera taken at an interval of ten days or more is
manifestations (dengue hemorrhagic fever) or confirmatory.
with shock (dengue shock syndrome). They are
more common in previously healthy children in the Hematological diagnosis: Thrombocytopenia
indigenous populations of endemic areas. (100,000 cells or less per mm3); Hemoconcentration
(720% in hematocrit).
Pathogenesis: The pathogenesis of the severe
syndrome involves preexisting dengue antibody.
Epidemiology
It is postulated that virus–antibody complexes
are formed within a few days of the second Dengue virus is transmitted from person to person
dengue infection and that the non-neutralizing by Aedes aegypti mosquitoes. Most subtropical and
enhancing antibodies promote infection of tropical regions around the world where Aedes
higher num­bers of mononuclear cells, followed vecrors exist are endemic areas. Dengue was
by the release of cytokines, vasoactive mediators, initially confined to the east coast of India and has
and procoagulants, lead­ing to the disseminated caused epidemics. All four types of dengue virus
intravascular coagulation seen in the hemorrhagic are present in this country.
fever syndrome.
Control
Dengue hemorrhagic fever: Dengue hemorrhagic
Control depends upon antimosquito measures, as
fever (DHF) may occur in individuals (usually
no vaccine is currently available.
children) with passively acquired (as maternal
A live attenuated vaccine containing all four
antibody) or preexisting non-neu­t ralizing
dengue serotypes is under clinical trial in order to
heterologous dengue antibody due to a previous
avoid DHF/DSS in immunized persons.
infection with a different serotype of virus.
Dengue shock syndrome (DSS), a more severe
form of the disease characterized by shock and B. Tick-borne Group
hemoconcentration, may ensue. Circumstantial These viruses produce two clinical syndromes,
evi­dence suggests that secondary infection with encephalitis hemorrhagic fevers.
dengue type 2 following a type 1 infection is a
particular risk factor for severe disease. a. Tick-borne Encephalitis Viruses (TBEV)
Russian spring summer encephalitis (RSSE)
Laboratory Diagnosis
complex: A number of viruses belonging to the
(a) Specimens Russian spring summer encephalitis (RSSE)
For antibody detection—Serum complex cause encephalitis along a wide area of
For isolation of virus and PCR. Serum, plasma, the northern landmass from Scotland to Siberia.
whole blood (washed buffy coat), autopsy tissues The names given to the disease vary from one area
and, mosquitoes collected in nature. to another. Thus, in Scotland, it is called ‘louping ill’
A. Virus detection: Isolation of the virus is as the disease occurs primarily in sheep in which
difficult. Virus isolation can be done by it causes a curious ‘leaping’ gait. Human cases that
inoculating clinical specimen into mosquitoes, result from contact with these sheep are mild and
mosquitoes cell lines (C6/36 or AP-61 cells), or present as aseptic meningitis. It is called Central

Chap-65.indd 452 15-03-2016 11:23:05


Chapter 65: Arboviruses  | 453
European Encephalitis in Central Europe, biphasic c. Vectors: Infection is transmitted by the bite of
meningoencephalitis in Eastern Europe and RSSE ticks, the principal vector being Haemaphysalis
in USSR. spinigera.
Human infections by TBEV may range in
severity from mild biphasic meningoencephalitis Clinical Features
to a severe form of polioencephalomyelitis. Incubation period is between 3 and 8 days. The
Biphasic meningoencephalitis may be trans­ disease appears with a sudden onset of fever,
mitted to human beings by drinking the milk of headache and severe myalgia, with prostration in
infected goats. Rare erosol transmission may occur. some patients. The acute phase lasts about 2 weeks.
Gastrointestinal disturbances and hemorrhages
Russian spring-summer encephalitis (RSSE): from nose, gums, stomach and intestine may occur
Infection is transmitted by the bite of lxodid ticks. in severe cases.
The virus is transmitted transovarially in ticks so
that they can act as vectors as well as reservoir Laboratory Diagnosis
hosts. Wild rodents and migrating birds are other A. Virus isolation: Virus can be isolated from
reservoirs. the blood up until the12th day in suckling mice,
hamster or monkey kidney cells or HeLa cells with
Control cytopathic effect.
The control of infection depends on the avoidance B. Serology: Serological diagnosis can be made
of tick bites. A formalin inactivated vaccine is with rising antibody (IFA, HI and N) titers in acute
commercially available for the Western subtype and convalescent sera, as well as by EIA tests.
and for the Eastern subtype.
Control
ii. Powassan virus: Powassan virus causes
Control is essentially a breaking of the tick-human
encephalitis in Canada and Northern USA.
contact. Personal protection involves regular
de-ticking of the body and the use of repellents and
b. Tick-borne Hemorrhagic Fevers protective clothing.
1. Kyasanur Forest Disease (KFD) Vaccine: A killed KFD virus vaccine was used in
Kyasanur forest disease (KFD) is a febrile disease a small field, trial and appeared to provide some
associated with hemorrhages caused by an degree of protection.
Arbovirus flavivirus and transmitted to man by bite
of infective ticks. A new Arbovirus antigenically 2. Omsk Hemorrhagic Fever
related to the RSSE complex, was isolated by This occurs in Russia and Romania. It is clinically
investigators from the National Institute of Virology similar to KFD and is caused by a related virus.
(then Virus Research Centre), Pune, from the Dermacentor ticks are the vectors.
patients and dead monkeys. The disease was later
named after the locality ­Kyasanur Forest from Family Bunyaviridae
where the virus was first isolated. The Bunyaviridae family contains more than 300
viruses is the largest group of arboviruses, mostly
History arrhropod-rransmitted.
KFD was first recognized in 1957 in Shimoga district Morphology: The virus is about 100 nm in diameter
of Karnataka state in South India. The situation and has a complex structure, with a triple segmented
changed suddenly in 1982 with the appearance genome of single stranded RNA.
of an epizootic and epidemic. The outbreak,
known locally as monkey fever’, started with dead Classification
monkeys. The ecological disturbance caused by
The family Bunyaviridae contains four genera of
clear felling of the virgin forest is believed to have
medical importance:
activated a silent enzootic focus of the virus.
A. Bunyavirus—Mosquito-borne
B. Phlebovirus—Sandfly
Epidemiology C. Nairovirus—Tick borne
a. Agent-Flavivirus: The agent KFD virus is a D. Hantavirus—Non-arthropord borne.
member of group B Togaviruses (Flaviviruses). A number of viruses are yet ungrouped.
b. Natural hosts and reservoirs—Main reservoirs
of the virus are small mammals particulairly A. Bunyavirus
rats and squirrels and forest birds. Monkeys are The genus contains over 150 species, of which only
only amplifier hosts. a few cause human infections. This genus includes

www.ebook3000.com
Chap-65.indd 453 15-03-2016 11:23:05
454  |  Section 4: Virology
California encephalitis virus, La Crosse virus, and (HFRS). Principal vertebrate reservoirs com­
Chittor virus. California encephalitis virus and La prise Apodemus agrariusrodents in Asia and
Crosse virus are isolated from the United States Clethrionomys glareolus (bank vole) in Europe.
and Chittor virus from India. The clinical disease Species: The genus contains at least four’
caused is encephalitis, aseptic meningitis and fever. species:
All are mosquito-borne infections. (i) Hantaan virus; (ii) Seoul virus; (iii) Puumala
virus; (iv) Prospect hill virus. Hantavirus
B. Phlebovirus species are natural pathogens of rodents-field
a. Sandfly fever (Phlebotomus fever): Sandfly mice (Apodemus agrarius) being the major host
fever (Phlebotomus fever) also known as for Hantaan. Transmission to humans occurs by
Pappataci fever and three-day fever, is a inhaling Aerosols of rodent excreta. The disease
self­, nonfatal fever transmitted by the bite of occurs in two forms—the milder epidemic
sandfly Phlebotomus papatasii. It occurs along nephritis (EN) common in Scandinavia and the
the mediterranean Coast and Central Asia, more serious epidemic hemorrhagic fever (EHF)
extendIng as far east as Pakistan and North in the far east. The clinical picture resembles
West India. Cases have also been reported from typhoid, leptospirosis and scrub typhus.
South and Central America. Domestic rats appear to be the source of
Natural vectors are Phlebotomus papatasi and infection in urban cases of HFRS. HFRS should
other phlebotomine sandflies. be considered a robovirus and not stricly an
Twenty antigenic types of the virus exist, of arbovirus infection in the absence of proved
which only five cause human disease—Naples, arthropod transmission.
Sicilian, Punta Toro, Chagres, Candiru. Its 2. Sin nombre virus (SNV): In 1993 an outbreak
occurrence in India was thought to be doubtful. of severe respiratory illness in the United States,
b. Rift valley fever: The agent of this disease, now designated the hantavirus pul­monary
a bunyavirus of the Phle­b ovirus genus, is a syndrome (HPS), was found to be caused by a
mosquito-borne zoonotic virus path­ogenic novel hantavirus.
primarily for sheep, cattle, and goats. Humans The disease is caused by a newly identified
are secondarily infected. hantavirus, the Sin nombre (meaning nameless)
virus, which is associated with the deer mouse and
C. Nairovirus other rodents.
Members of the Crimean Congo hemorrhagic Infection appears to be caused by inhalation
group are the major human pathogens in this of the virus aerosols in dried rodent feces. Person-
genus. to-person trans­mission of hantaviruses seldom
Crimean Congo Hemorrhagic Fever (CCHF) occurs.
virus: is distributed widely. Cattle, sheep, goats
and other domesticated animals act as natural Laboratory Diagnosis
reservoirs. It is transmitted by Hyalomma ticks. Laboratory diagnosis depends on detection
During the acute phase of the disease, the blood of viral nucleic acid by reverse transcription-
of the patients is highly infectious and direct polymerase chain reaction or detection of specific
transmission may occur through contact. antibodies using recom­binant proteins of different
hantaviruses. Hantaviruses can be isolated in
Hazara virus: A related virus, Hazara, has been cultured cells.
isolated in Pakistan.
Nairobi sheep disease: It is an acute, hemorrhagic Reoviridae
gastroenteritis caused by a Nairovirus in sheep and Family Reoviridae contains four genera-Orbivirus,
goats in East Africa. It is transmitted by Rhipice­ Coltivirus, Orthoreovirus, and Rotavirus
phalus ticks. The genus Orbivirus contains arthropod
Ganjam virus: Ganjam virus, isolated from ticks borne viruses. Rotaviruses and reoviruses have no
collected from sheep and goats in Orissa, India, is arthropod vectors.
closely related to the Nairobi sheep disease virus. A. Genus Orbivirus: The genus Orbivirus contains
arthropod borne viruses which infect animals
D. Hantavirus and humans.
1. Hantaviruses: Hantaan viruses are classified i. African horse sickness virus: African horse
in the Hantavirus genus of the Bunyaviridae sickness virus, transmitted by Culicoides.
family. The viruses are found world­wide and It caused extensive disease among army
cause hemorrhagic fever with renal syndrome horses and mules in India.

Chap-65.indd 454 15-03-2016 11:23:05


Chapter 65: Arboviruses  | 455
ii. Palyam, Kasba and Vellore viruses: Palyam,
™™ Arboviruses are classified within six families. Most
Kasba and Vellore viruses belonging to the are members of the families Togaviridae, Flaviviridae
Orbivirus group have been isolated from and Bunyaviridae
mosquitoes in India but their pathogenic ™™ Arboviruses cause the clinical syndromes such
significance is not known. as fever with or without rash and arthralgia
B. Genus Coltivirus: Colorado tick fever is encephalitis, hemorrhagic fever, the characteristic
systemic disease, yellow fever
classified in the genus Coltivirus.
™™ The family Togaviridae contains two genera:
Colorado tick fever: Colorado tick fever, is
Alphavirus and rubivirus
transmitted by a tick. It is spread by the wood
1. Alphavirus (Mosquito-borne)-A. Encephalitis
tick Dermacentor andersoni. viruses: (i) Eastern equine encephalitis (EEE); (ii)
C. Orthoreovirus: Cause asymptomatic infections Western equine encephalitis (WEE); (iii) Venezuelan
in humans. equine encephalitis (VEE) B. Viruses Causing Febrile
D. Rotavirus: Causes human infantile gastroemeritis. Illness; 1. Chikungunya virus (CHIKV); (ii) Onyong-
nyong virus (ONNV); (iii) Semliki Forestvirus; (iv)
Rhabdoviridae Sindbis virus; (v) Ross River virus
2. Rubivirus; Rubella virus
Chandipura virus: Chandipura virus, belonging
™™ The chikungunya virus has been implicated in
to the vesiculovirus genus of Rhabdoviridae, was
epidemics in India. Humans are the host, and Aedes
isolated in 1967 from the blood of patients during aegypti mosquitoes the vectors
an epidemic of dengue­ chikungunya fever in ™™ Japanese encephalitis (JE) is a mosquito-borne
Nagpur. The virus appears to multiply in sandflies encephalitis caused by a group B arbovirus
and Aedes mosquitoes. Antibodies are common in (Flavivirus) and transmitted by culicine mosquitoes.
human sera from different parts of India, as well as Culex tritaeniorhynchus, a rural mosquito that breeds
in animal sera. The pathogenic significance of this in rice fields, is the principal vector.
virus has not been established. ™™ Yellow fever virus is the prototype member of the
Fla­viviridae family. lt causes yellow fever, an acute,
febrile, mosquito-borne illness that occurs only in
UNGROUPED ARBOVIRUSES Africa and Central and South America. The disease
yellow fever does not occur in India
Wanowri virus: This was isolated from Hyalomma ™™ The dengue virus causes classic dengue or
ticks in India. breakdown fever, dengue hemorrhagic fever (DHF),
Bhanja virus: This was isolated from hemophysalis and dengue shock syndrome (DSS). Both dengue
virus and chikungunya virus are transmitted by Aedes
ticks from goats in Ganjam, Orissa.
aegypti mosquito
™™ Kyasanur Forest Disease (KFD) is a febrile disease
ARBOVIRUS KNOWN TO BE PREVALENT associated with hemorrhages caused by an arbovirus
IN INDIA flavivirus and transmitted to man by bite of infective
ticks
Some of the arboviruses known to be prevalent in ™™ Hantaviruses are classified in the Hantavirus
India are as shown in Table 65.4. genus of the Bunyaviridae family. The viruses cause
hemorrhagic fever with renal syndrome (HFRS). The
Table 65.4:  Some of the arboviruses known to be genus contains at least four species:
prevalent in India
A.Group A (Alphaviruses) C.Others
Sindbis Umbre
Chikungunya Sathuperi
IMPORTANT QUESTIONS
Chandipura 1. Classify arboviruses. Discuss various methods
B.Group B ( Flaviruses) Chittoor used for laboratory diagnosis of arboviruses.
Dengue Ganjam
KFD Minnal
2. Name the arboviruses which cause encephalitis.
JE Venkatapuram Describe briefly Japanese B encephalitis.
West Nile Dhori 3. List the arboviruses prevalent in India. Describe
Kaisodi the pathogenicity and laboratory diagnosis of
Sandfly fever dengue virus.
African horse sickness 4. Write short notes on:
Vellore
a. Chikungunya
b. Yellow fever
Key Points c. Dengue fever (or) Break bone fever
™™ Arboviruses are viruses “which are maintained in d. Kyasanur forest disease (KFD)
nature principally, or to an important extent through e. Bunyaviruses
biological transmission between susceptible f. Sandfly fever (phlebotomus fever)
vertebrate hosts by hematophagous arthropods
g. Hantavirusess.

www.ebook3000.com
Chap-65.indd 455 15-03-2016 11:23:05
456  |  Section 4: Virology
MULTIPLE CHOICE QUESTIONS (MCQs) 5. All the following statements are true for dengue
hemorrhagic fever except:
1. Which of the following viral genera is/are not a. This occurs in children with passively acquired
arthropod-borne? maternal antibodies
a. Alphavirus b. Flavivirus b. It is a most severe manifestation of the disease
c. Rubivirus d. All of the above c. This condition shows a fatality rate as high as
2. All the following viruses are transmitted by 10%
arthropod vectors except: d. Immunization is carried out by, a heat-killed
a. Hantavirus vaccine
b. Chandipura virus
6. The vaccine is not available for:
c. Rift Valley fever virus
a. Dengue fever
d. Sandfly fever virus
b. Japanese encephalitis
3. Mosquitoes act as the vector in all of the following
c. Yellow fever
viruses except:
a. Chikungunya virus d. Russian spring-summer encephalitis
b. Dengue virus 7. Hantavirus pulmonary syndrome is caused by:
c. Semiliki Forest virus a. Nairobi sheep disease virus
d. Omsk hemorrhagic fever virus b. Chittor virus
4. Arbovirus transmitted by tick is: c. Sin Nombre virus
a. Western equine encephalitis d. West Nile virus
b. B. Kyasanur Forest disease
c. Russian spring-summer encephalitis ANSWERS (MCQs)
d. Omsk hemorrhagic fever 1. c; 2 a; 3. d; 4. a; 5. d; 6. a; 7. c

Chap-65.indd 456 15-03-2016 11:23:05


66
Chapter

Rhabdoviruses

Learning Objectives

After reading and studying this chapter, you should ∙∙ Discuss laboratory diagnosis of rabies
be able to: ∙∙ Discuss prophylaxis against rabies
∙∙ Describe morphology of rabies virus ∙∙ Describe neural and non-neural vaccines against
∙∙ Describe the following: Street virus vs. fixed virus; rabies.
pathogenesis and clinical features of rabies; Negri
bodies

Introduction of the virus which may be invaginated at the


planar end. (Fig. 66.1).
Bullet shaped, enveloped viruses with single stranded 4. Genome: The core of the virion consists of
RNA genome are classified as rhabdoviruses (from hel­ically arranged ribonucleoprotein. The
rhabdos, meaning rod) in the family Rhabdoviridae. genome is single-stranded RNA, linear, non-
segmented, negative-sense. RNA-dependent
Classification RNA polymerase enzyme which is essential
Rhabdoviruses infecting mammals belong to two for the initiation of replication of the virus, is
genera in Rhabdoviridae family enclosed within the virion in associa­tion with
the ribonucleoprotein core.
A. Lyssavirus
It contains rabies virus and related viruses
(Lagos bat virus, Mokola, Duvenhage).
B. Genus vesiculovirus: It con­tains vesicular
stomatitis virus (VSV) and related viruses like
Chandipur (arbelius).

Rabies virus
Morphology
1. Virion: The rabies virion consists of a helical
nucleocapsid contained in a bullet-shaped
lipoprotein envelope 180 × 75 nm, with one
end rounded or conical and the other plane or
concave (Fig. 66.1).
2. Proteins: Protruding from the lipid envelope
are approximately 200 glycoprotein (G) spikes
of the virus, hemagglutinin activity. Spikes do
not cover the planar end of the virion.
3. Membrane or matrix (M) protein: Beneath Fig. 66.1: Rabies virus: Bullet-shaped virion, showing tightly
the envelope is the membrane or matrix (M) wound helix of ribonucleoprotein in the core, and bilayered
protein and is the major structural protein membranous envelope carrying glycoprotein spikes

www.ebook3000.com
458  |  Section 4: Virology
Resistance Street Virus
Rabies virus is sensitive to ethanol, iodine prepar­ The rabies virus isolated from natural human
ations, quaternary ammonium compounds, or animal infection is termed the street virus.
soap, detergents and lipid solvents such as ether, Following inoculation by any route, it can
chloroform and acetone. It is inactivated by phenol, cause fatal encephalitis in laboratory animals
formalin, beta propiolactone, ultraviolet irradiation, after a long and variable incubation period of
sunlight and heat at 50°C for 1 hour or 60°C for 5 about 1–12 weeks (usually 21–60 days in dogs).
minutes. Rabies virus survives storage at 4°C for Intracytoplasmic inclusion bodies (Negri bodies)
weeks and at –70°C for years or by lyophilization It can be demonstrated in the brain of animals dying
is inactivated by CO2 so on dry ice it must be stored of street virus infection.
in glass-sealed vials.
Fixed Virus
Antigenic Properties After several serial intracerebral passages in rabbits,
There is a single serotype of rabies virus. the virus undergoes certain changes and becomes
A. Glycoprotein G: The surface spikes are what is called the fixed virus that no longer multiplies
composed of glycoprotein G, which is important in extraneural tis­sues. The fixed (or murant) virus
in pathogenesis, virulence and immunity. is more neurotropic, though it is much less infective
Purified spikes containing the viral glycoprotein by other routes. After intracerebral inoculation, it
elicit neutralizing antibody in animals. The produces fatal encephalitis after a short and fixed
purified glycoprotein may therefore provide a incubation period of 6–7 days. Negri bodies are
safe and effective subunit vaccine. usually not demonstrable in the brain of animals
Hemagglutinating activity: Rabies virus dying of fixed virus infection. The fixed virus is used
possesses hemagglutinating activity, optimally for vaccine production.
seen with goose erythrocytes at 0–4 °C and
pH 6.2. Hemagglutination is a property of B. Chick Embryos
the glycoprotein spikes. The hemagglutinin The rabies virus can be grown in chick embryos.
antigen is species specific and distinct from the The usual mode of inoculation is into the yolk sac.
antigens on rabies related viruses. Serial propagation in chick embryos has led to the
B. Nucleocapsid protein: Complement fixing development of attenuated vaccine strains like
antibodies are induced by the nucleocapsid Flury and Kelev.
protein and are not protective. This antigen
is group specific and cross-reactions are seen C. Tissue Culture
with some rabies related viruses. Antiserum pre­
The rabies virus can grow in chick embryo
pared against the purified nucleocapsid is used
fibroblast, porcine or hamster kidney cells human
in diag­nostic immunofluorescence for rabies.
diploid cell and vero cell cultures.
C. Other antigens: Other antigens identified
include two membrane proteins, glycolipid
and RNA dependent RNA polymerase.
Pathogenesis
Rabies infection usually results from the bite of
Host Range and Growth Characteristics rabid dogs or other animals. The virus can also be
transmitted following nonbite exposures through
A. Animals the inhalation of aerosolized virus (as may be found
Rabies virus has a wide host range. All warm- in bat caves), in transplanted infected tissue (e.g.
blooded animals, including humans, can be cornea), and by inoc­ulation through intact mucosal
infected. All mammals are susceptible to rabies membranes. The virus present in the saliva of the
infection, though differences in susceptibility animal is deposited in the wound.
exist between species. Suscepti­bility varies among The virus appears to multiply in the muscles,
mammalian species, ranging from very high connective tissue or nerves at the site of deposition.
(foxes, coyotes, wolves) to low (opossums); those The virus remains at the site for days to months
with intermediate susceptibility include skunks, (Fig. 66.2) before progressing to the central
raccoons, and bats. Humans and dogs occupy an nervous system (CNS). Rabies virus travels by
intermediate position. Pups are more susceptible retrograde axoplasmic transport to the dorsal root
than adult dogs. Experimental infection can be ganglia and to the spinal cord. Once the virus gains
produced in any laboratory animal but mice are access to the spinal cord, the brain becomes rapidly
the animals of choice. They can be infected by any infected. The virus then disseminates from the CNS
route. After intracerebral inoculation, they develop via afferent neurons to highly innervated sites, such
encephalitis and die within 5–30 days. as the skin of the head and neck, salivary glands,
Chapter 66: Rhabdoviruses  | 459
b. Acute neurologic phase: During the acute
neurologic phase, which lasts 2–7 days, patients
show signs of nervous system dysfunction such
as nervousness, apprehension, hallucinations,
and bizarre behavior.
Hydrophobia: The pathognomonic feature
is difficulty in drinking, together with intense
thirst. Patients may be able to swallow dry solids
but not liquids. Attempts to drink bring on
such painful spasms of the pharynx and larynx
producing choking or gagging that patients
develop a dread of even the sight or sound of
water (hydrophobia). Generalized convulsions
follow.
c. Coma: Patients who survive the stage of acute
neurological involvement lapse into coma.
Death usually occurs within 1–6 days due to
respiratory arrest during convulsions.

B. Rabies in Dogs
Clinical Picture
Fig. 66.2: Pathogenesis of rabies virus infection Rabies in dogs may manifest itself in two forms—
Furious rabies and Dumb rabies.
retina, cornea, nasal mucosa, adrenal medulla, a. Furious rabies: This is the typical “mad dog
renal paren­chyma, and pancreatic acinar cells. syndrome”, characterized by (i) A change in
The presence of the virus in the saliva and behavior; (ii) Running amuck; (iii) Change in
the irritability and aggression brought on by the voice; (iv) Excessive salivation and foaming at
encephalitis ensure the transmission and survival the angle of the mouth; (v) Paralytic stage—
of the virus in nature. The virus ultimately reaches Paralysis, convulsions and death follow.
virtually every tissue in the body, though the
b. Dumb rabies: In this type, the excitative or
centrifugal dissemination may be interrupted at
irritative stage is lacking. The dumb form is as
any stage by death. The virus is almost invariably
infectious as the furious type.
present in the cornea and the facial skin of patients
because of their proximity to the brain. The virus
may also be shed in milk and urine. Laboratory Diagnosis
With rare exception (three known cases), rabies 1. Diagnosis of Human Rabies
is fatal once clinical disease is apparent.
Tests are performed on samples of saliva, serum,
spinal fluid, and skin biopsies of hair follicles at the
Clinical Features nape of the neck.
A. Humans
Rabies is primarily a disease of lower animals and A. Rabies Antigens by Immunofluorescence
is spread to humans by bites of rabid animals or
The method most commonly used for diagnosis
by contact with saliva from rabid animals. The
is the demonstration of rabies virus antigens by
infection has also been acquired from aerosols in
immunofluorescence. The specimens tested are
bats’ caves.
corneal smears and skin biopsy (from face or neck)
The incubation period in humans is typically
or saliva antemortem, and brain postmorterm.
1–2 months but may be as short as 1 week or as long
Direct immunofluorescence is done using antirabies
as many years (up to 19 years). It is usu­ally shorter
serum tagged with fluorescein isothiocyanate. The
in children than in adults.
use of monoclonal antibody instead of crude
antiserum makes the test more specific.
Phases of Clinical Spectrum
The clinical spec­trum can be divided into three B. Virus Isolation
phases:
a. Prodro­mal phase: The prodrome, lasting a. Mouse Inoculation
2–10 days, may show any of the nonspecific Samples of brain tissue, saliva, CSF, or urine may
symptoms. be injected intracerebrally into newborn mice for

www.ebook3000.com
460  |  Section 4: Virology
isolation of the virus. Infection in mice results in Italian physician who first discovered them. This is
encephalitis and death. The inoculated mice are still the method most commonly used as facilities
examined for signs of illness and their brains are for immunofluorescence and biological tests are
examined at death or at 28 days postinoculation not available in many laboratories.
for Negri bodies, or by immunofluorescence rabies Impression smears of the brain are stained by
antigen. Seller’s technique (basic fuchsin and methylene
blue in methanol), which has the advantage that
b. Isolation in Cell Culture fixation and staining are done simultaneously.
Negri bodies are seen as intracytoplasmic, round
A more rapid and sensitive method is isolation
or oval, purplish pink structures with characteristic
of the virus in tissue culture cell lines (WI38,
basophilic inner granules. Negri bodies vary in size
BHK21,CER). Virus isolations is identified by
from 3–27 µm. Other types of inclusion bodies may
immunofluorescence. A positive IF test can be
sometimes be seen in the brain in diseases such
obtained as early as 2–4 days after inoculation.
as canine distemper but the presence of inner
The identity of the isolate can be established by the
structures in the Negri bodies makes differentiation
neutralization test with specific antirabies antibody.
easy. Failure to find Negri bodies does not
exclude the diagnosis of rabies. The microscopic
C. Serology examination for Negri bodies identifies 75–90% of
Rabies antibodies can be detected in the serum and cases of rabies in dogs. Failure to find Negri bodies
CSF of the patient by ELISA. High titer antibodies does not exclude the diagnosis of rabies.
are present in the CSF in rabies but not after Negri bodies contain rabies virus antigens and
immunization. can be demonstrated by immunofluores­cence.
Both Negri bodies and rabies antigen can usually
D. Detection of Nucleic Acid be found in animals or humans infected with
rabies, but they are rarely found in bats.
Reverse transcription-polymerase chain reaction
(RT-PCR) test­ing can be used to amplify parts If impression smears are negative, the tissue
of a rabies virus genome from fixed or unfixed should be sectioned and stained by Giemsa or
brain tissue for detection of rabies virus RNA. This Mann’s method.
technique can confirm dFA results and can detect 3. Isolation of the rabies virus (biological test)
rabies virus in saliva and skin biopsy samples. This is done as described above, for human rabies
diagnosis.
2. Animal Rabies 4. Corneal test
The head of the animal is cut off and sent to the Rabies virus antigen can be detected in live animals
nearest testing laboratory, duly packed in ice in an in corneal impressions or in frozen sections of skin
air-tight container. Alternatively, the brain may be biopsies by the flueroscent antibody test.
removed with antiseptic precautions and sent in
50% glycerol-saline for examination and the other Prophylaxis
in Zenker’s fixative, sent for biological test and
microscopy, respectively. The portion of brain sent This may be considered under:
should include the hippocampus and cerebellum
as Negri bodies are most abundant there. The A. Postexposure Prophylaxis
following tests are done in the laboratory: B. Preexposure Prophylaxis
1. Demonstration of rabies virus antigen by A. Post-exposure Prophylaxis
immuno­fluorescence This consists of: a. Local treatment , b. Antirabic
This is a highly reliable and the best single test vaccines, c. Hyperimmune serum.
currently available for the rapid diagnosis of rabies a. Local treatment of wound:
viral antigen in infected specimens. This test i. Cleansing: Immediate flushing and
can establish a highly specific diagnosis within washing the wound(s), scratches and the
a few hours. Examination of salivary glands by
adjoining areas with plenty of soap and
immunofluorescence is useful.
water, preferably under a running tap, for
2. Demonstration of inclusion bodies (Negri at least 5 minutes as soap inactivates virus
bodies) by destroying its envelope.
A definitive pathologic diagnosis of rabies can be ii. Chemical treatment: After cleansing
based on the finding of Negri bodies in the brain wound should be inactivated by irrigation
or the spinal cord. Negri bodies, named after the with virucidal agents - either alcohol
Chapter 66: Rhabdoviruses  | 461
(400–700 ml/liter), tincture or 0.01% 3. Infant brain vaccines
aqueous solution of iodine or povidone The encephalitogenic factor in brain tissue is a
iodine. basic protein associated with myelin. It is scanty
iii. Suturing: Bite wounds should not be or absent in the nonmyelinated neural tissue of
immediately sutured. newborn animals. So vaccines were developed
iv. Antirabies Serum: The local application of using infant mouse, rat or rabbit brain.
antirabies serum or its infiltration around
the wound has been shown to be highly II. Non-neural Vaccines
effective in preventing rabies. 1. Egg vaccines
v. Antibiotics and antitetanus measure:
a. Duck embryo vaccine (DEV): It was disconti­
When indicated should follow the local
nued because of its poor immunogenicity. (b)
treatment recommended above.
Live attenuated chick embryo vaccines—These
b. Antirabic vaccines: Antirabic vaccines fall into vaccines were used for vaccination of animals.
two main categories: neural and non-neural Two types of vaccines were developed with the
(Table 66.2). The former are associated with Flury strain.
serious risk of neurological complications and
i. Low Egg Passage (LEP) vaccine: at 40–50 egg
have been replaced by the latter.
passage level for immunization of dogs.
I. Neural Vaccines ii. High Egg Passage (HEP) vaccine: at 180
passage level for cattle and cats. These are not
These are suspensions of nervous tissues of
in use now.
animals infected with the fixed rabies virus.
2. Cell culture vaccines
1. Semple Vaccine
Semple (1911) developed this vaccine at the Central The cell culture vaccines are of two types:
Research Institute, Kasauli (India), had been the
i. Human origin
most widely used vaccine for over half a century.
It is a 5% suspension of sheep brain infected with Human diploid cell vaccine (HDCV)
fixed virus and inactivated with phenol at 37°C, The first cell culture vaccine was the human diploid
leaving no residual live virus. cell (HDC) vaccine developed by Koprowsky,
Wiktor and Plotkin. It is a purified and concentrated
2. Beta propiolactone (BPL) vaccine preparation of fixed rabies virus (Pitman–Moore
This is a modification of the Semple vaccine, strain) grown on human diploid cells (WI 38 or
in which beta propiolactone is used as the MRC 5) and inactivated with beta propiolactone
inactivating agent instead of phenol. It is prepared or tri-n-butyl phosphate. It is highly antigenic and
from fixed virus grown in the brains of adult sheep free from serious side effects. Its only disadvantage
(Semple type) or other animals. It is believed to be is its high cost. HDC vaccine is now licensed for use
more antigenic. in a number of countries including India for both
pre- and postexposure immunization.

Table 66.2:  Rabies vaccines ii. Nonhuman origin


I. Neural vaccines iii. Second generation tissue culture vaccines
1. Pasteur vaccine (Table 66.2): Second generation tissue culture
2. Fermi vaccine vaccines are cheaper than HDC vaccines.
3. Semple vaccine
Cell culture vaccines in India: In India the
4. Beta-propiolactone (BPL) vaccine
following cell culture vaccines are available:
5. Suckling mouse brain vaccine
II. Non-neural vaccines i. Human diploid cell (HDC) vaccine
A. Duck egg vaccine ii. Purified chick embryo cell (PCEC) vaccine
B. Cell culture vaccines iii. Purified vero cell (PVC) vaccine.
a. First-generation cell culture vaccine All three of them are equally safe and effective.
– Human diploid cell vaccine (HDCV)
b. Second-generation cell culture vaccines
– Purified chick embryo cell vaccine (PCEC) 3. Subunit Vaccine
– Purified Vero cell rabies vaccine (PVRV) The glycoprotein subunit on the virus surface,
III. Third-generation rabies vaccine which is the protective antigen, has been cloned
Poxvirus-rabies glycoprotein recombinant vaccine and recombinant vaccines produced. They are still
(undergoing clinical trials in humans) in the experimental stage.

www.ebook3000.com
462  |  Section 4: Virology
Indications for Antirabies Treatment Dosage Schedules
Antirabies treatment should be started immediately: Two dosage schedules (one recommended by
i. If the animal shows signs of rabies or dies the Central Research Institute, Kasauli and the
within 10 days of observation. other recommended by the Pasteur Institute of
ii. If the biting animal cannot be traced or Southern India, Coonoor) are followed in India.
identified. A full schedule consists of 7–10 daily inoculations
iii. Unprovoked bites. followed by 1–2 boosters.
iv. Laboratory tests (e.g. fluorescent rabies anti­
body test or test for Negri bodies) of the brain Site for Vaccination
of the biting animal are positive for rabies.
v. All bites by wild animals. The ideal site for vaccination is the anterior
abdominal wall, for this area offers enough space
to accommodate the large quantity of vaccine
Vaccination Schedules to be injected. The injections are given deep
Neural Vaccines subcutaneously.
The dosage of the vaccine depends on the degree
of risk to which the patient has been exposed. Adverse Reactions
Accordingly, patients are classified as follows.
1. General: Headache, insomnia, giddiness,
palpitation, diarrhea.
Classification of Exposures
2. Local: Itching irritation, pain, tenderness,
One of the factors determining the dose of anti-
redness and swelling at the site of injection.
rabies vaccine is the degree of risk of rabies to
which the person is exposed. Accordingly, patients 3. Allergic: Urticaria, syncope, angioneurotic
are classified as follows. edema, anaphylactic reaction.
4. Neuroparalysis: Post-vaccinial paralysis due to
Class I (Slight risk) sensitization.
a. Licks on healthy unbroken skin.
b. Licks on intact mucous membrane or conjunctiva.
Cell Culture Vaccines
c. Bites or scratches which have raised the
epidermis but have not drawn blood, on all All three cell culture vaccines available in India
parts of the body except the head, face, neck or (HDC, PCEC and PVC) have the same dosage
fingers. schedule, which is the same for both adults and
d. Consumption of unboiled milk or handling raw children.
flesh of rabid animals.
Class II (Moderate risk) i. Preexposure Prophylaxis
a. Licks on definitely remembered fresh cuts or This is indicated for persons at high risk of contact
abrasions on the fingers. with rabies virus such as laboratory workers
b. Scratches with oozing of blood. handling rabies virus or with rabid animals
c. All bites except those on head, neck, face, palms (veterinarians). Preexposure prophylaxis requires
and fingers. three doses of the vaccine injected on day 0, 7, 21 or
d. Minor wounds less than 5 in number. 0, 28 and 56. A booster dose is recommended after
one year and then one every five years.
Class III (Severe risk)
a. Licks on definitely remembered fresh cuts or
abrasions on head, face or neck. ii. Postexposure Prophylaxis
b. All bites or scratches on the head, face or neck. Postexposure prophylaxis requires five or six doses,
c. All bites or scratches on the fingers which are on days 0, 3, 7, 14, 30 and optionally 90. The vaccine
lacerated, more than half centimeter long or is to be given intra muscular (1M) or subcutaneous
have penetrated the true skin. (SC) in the deltoid region, or in children on the
d. All bites penetrating the true skin and drawing anterolateral aspect of the thigh. Gluteal injections
blood, when there are five teeth marks or more. are to be avoided as they are found to be less
e. All bites on any part of the body causing immunogenic. This course is expected to give
extensive laceration. protection for at least five years, during which
f. Bites from wild animals—All jackal and wolf period any further exposure may need only one or
bites. two booster doses (on days 0, 3) depending on the
g. Any Class II patient who has not received degree of risk. After five years, it is advisable to give
treatment within 14 days of exposure. a full five injection course if exposed to infection.
Chapter 66: Rhabdoviruses  | 463
C. Passive Immunization Table 66.3:  Lyssavirus sera/genotypes
Passive immunization is an important adjunct to Genotype/ Virus Isolated Disribution
vaccination and should be invariably employed Serotype from
whenever the exposure is considered of high risk. Virus
Two preparations of anti rabies serum are available 1. Rabies Warm Worldwide
for passive immunization: blooded with few
animals exceptions
i. Horse Antirabies Serum: It should be given on 2. Lagos bat/ Bat/cat Nigeria/
day 0 in a single dose of 40 IU/kg of body weight Natal bat Central and
subject to a maximum of 3000 Units. South Africa
3. Mokola Shrew/ Nigeria/
ii. Human Rabies Immune Globulin cat/dog/ other African
human countries
Human rabies immune globulin (HRlG) has
now replaced equine antirabies serum in many 4. Duvenhage Human/ South Africa
countries. The dose recommended is a single bat
administration of 20 IU per kg of body weight. 5. European Bat/human Europe
bat
Vaccine for Animals lyssavirus:
Type I
Concentrated cell culture vaccines containing 6. European Bat/human Europe
inactivated virus are now available, which give bat
good protection after a single IM injection. lyssavirus:
Injections are given at 12 weeks of age and repeated Type II
at 1–3 year intervals. 7. Australian Bat/human Australia
bat
lyssavirus:
B. Preexposure Prophylaxis
This is indicated for persons at high risk of contact
with rabies virus or with rabid animals. It is rec­
Rabies in India: Rabies is endemic in India.
ommended that antibody titers of vaccinated
Rabies is not a notifiable disease and the 30,000
individuals be monitored periodically and that
deaths reported by national authorities may not be
boosters be given when required.
complete picture. Every year approximately 1.1–1.5
million people receive post-exposure treatment with
Epidemiology either nerve tissue or cell culture rabies vaccine.
Rabies is the classic zoonotic infection, spread More than 95% of these cases are bitten by dogs.
from animals to humans. Rabies virus is present in
terrestrial animals in all parts of the world except Control
Australasia and Antartica, and some islands like Human rabies can be checked by control of rabies
Britain. in domestic animals, by registration, licensing and
All warm blooded animals including man are vaccination of pets and destruction of stray animals.
susceptible to rabies. Rabies in man is a dead-end
infection, and has no survival value for the virus. Rabies related viruses
Major source of virus is saliva in bite of rabid
animal. Direct person-to-person transmission of The genus Lyssavirus consists of the rabies virus and
rabies has not been recorded. An unusual mode other serologically related viruses. The relevance of
of transmission of rabies has occurred in some rabies related viruses in human diseases is not clear,
recipients of corneal grafts. Minor source is aerosols though some of them have caused illness and death
in bat caves containing rabid bats. in humans. They are considered to represent a
biological bridge between the rabies virus and other
Epidemiological forms of rabies: Rabies exists
rhabdoviruses. Lyssaviruses have been classified
in two epidemiological forms: a. Urban rabies: b.
into seven serotypes (Table 66.3).
Wild-life or sylvatic rabies:
In certain Latin American countries and parts Key Points
of U.S.A. the vampire bat is an important host
and vector of rabies. Vampire bats have not been ™™ Rhabdoviruses are bullet or rod shaped, enveloped
viruses with single stranded RNA genome
reported in India.
™™ The rabies virus causes rabies in humans and a wide
Reservoir of rabies—Reservoir of rabies are variety of animals
wild animals.

www.ebook3000.com
464  |  Section 4: Virology
™™ The rabies virus isolated from natural human or 3. Describe the prophylaxis of rabies.
animal infection is termed the street virus 4. Write short notes on:
™™ After several serial intracerebral passages in rabbits, a. Street virus vs. fixed virus or Differences between
the virus undergoes certain changes and becomes street virus and fixed virus.
what is called the fixed virus (or murant) virus b. Negri bodies.
™™ Rabies virus is transmitted to humans by primarily c. Neural vaccines against rabies.
a bite of a rabid dog or by other infected animals.
The infection has also been acquired from aerosols d. Non-neural vaccines used against rabies.
in bats’ caves e. Cell culture vaccines.
™™ Laboratory tests for human rabies—Viral antigens f. Rabies related viruses.
can be demonstrated in the corneal smear, skin
biopsy, and saliva (antemortem) or in the brain
tissue (postmortem) directly by direct fluorescent
Multiple choice questions
antibody (DFA) test 1. Which of the following species of animals is most
™™ Isolation of rabies virus can be done by mouse susceptible to rabies infection?
inoculation and isolation in cell culture. Reverse a. Dog b. Man
transcription-polymerase chain reaction (RT-PCR) c. Cat d. Fowl
test­ing can be used for detection of rabies virus RNA
2. The inclusion bodies found in rabies are called:
™™ Demonstration of Negri bodies in neural tissues by a. Bollinger bodies
microscopy is diagnostic of rabies
b. Councilman bodies
™™ Postexposure prophylaxis is started in the persons c. Cowdry type a bodies
immediately after exposure to infection and consists d. Negri bodies
of (a) Local treatment of wound; (b) Antirabic
vaccines and (c) Hyperimmune serum 3. Negri bodies can be demonstrated in infection with:
a. Fixed rabies virus
™™ Cell culture vaccines such as human diploid cell
b. Street rabies virus
vaccine (HDCS), puri­fied chick embryo cell (PCEC)
c. Both of the above
vaccine, and purified vero cell (PVC) vaccines are now
d. None of the above
increasingly used. These are non-neural vaccines
™™ Postexposure prophylaxis requires five or six doses, 4. Which of the following clinical specimens can be
on days 0, 3, 7, 14, 30 and optionally 90 used for the demonstration of rabies antigen by
™™ Human rabies can be checked by control of rabies direct immunofluorescence antemortem?
in domestic animals, by registration, licensing and a. Salivary smears b. Corneal smears
vaccination of pets and destruction of stray animals. c. Conjunctival smears d. All of the above
5. All of the following antirabies vaccine are inactivated
Important questions vaccines except:
a. Human diploid cell strain vaccine
1. Describe the morphology and draw a labeled b. Purified vero cell vaccine
diagram of rabies virus. Discuss the laboratory c. Purified chick embryo cell culture vaccine
diagnosis of rabies. d. Chick embryo vaccine
2. Discuss the pathogenesis and clinical features of Answers (MCQs)
rabies. 1. c; 2. d; 3. b; 4. d; 5. d
6767
Chapter

Hepatitis Viruses

Learning Objectives

After reading and studying this chapter, you should modes of transmission of hepatitis B virus; hepatits
be able to: B carriers
∙∙ Classify various hepatitis viruses ∙∙ Describe laboratory diagnosis of hepatitis B virus
∙∙ Tabulate differences between various hepatitis ∙∙ Discuss prophylaxis of hepatitis B infections or
viruses hepatitis B vaccine
∙∙ Compare various features of hepatitis A virus (HAV) ∙∙ Describe the following:Hepatitis C virus or Type C
and hepatitis B virus (HBV) hepatitis; Hepatitis D virus or Delta agent; Hepatitis
∙∙ Describe the following: Morphology of hepatitis E virus; Hepatitis G virus.
B virus; antigenic structure of hepatitis B virus;

Introduction The virus is destroyed by autoclaving (121°C for 20


minutes), by boiling in water for 5 minutes, by dry
Viral hepatitis is a systemic disease primarily heat (180°C for 1 hour), by ultraviolet irradiation
involving the liver. At least six viruses, A through E (1 minute at 1.1 watts), by treat­ment with formalin
and a newly discovered G, are considered hepatitis (1:4000 for 3 days at 37°C, or by treatment with
viruses. Hepatitis A virus (HAV) and Hepatitis B chlorine (10–15 ppm for 30 minutes). It survives
vi­rus (HBV) are the best known, but three non- pro­longed storage at a temperature of 4°C or below.
A, non­-B hepatitis (NANBH) viruses (C, G, and
E) have been described, as has hepatitis D virus
(HDV), the delta agent (Table 67.1).
Pathogenesis
Hepatitis A virus is ingested and probably enters
Hepatitis A Virus the bloodstream through the oropharynx or the
epithelial lining of the intestines to reach its target,
(Infectious hepatitis)
the parenchymal cells of the liver. The virus can be
Hepatitis A virus (HAV) causes infectious hepatitis localized by immunofluorescence in hepatocytes
and is spread by the fecal–oral route. It is a subacute and Kupffer’s cells. Virus is produced in these cells
disease of global distribution, affecting mainly chil­ and is released into the bile and from there into
dren and young adults. the stool. Virus is shed in large quantity into the
stool approximately 10 days before symptoms of
Morphology jaundice appear or antibody can be detected.
HAV is a 27 nm nonenveloped RNA virus be­longing
to the Picornavirus family (Fig. 67.1). Although it Epidemiology
was first provisionally classified as enterovirus 72, The HAV transmission is by the fecal–oral route in
but it has been placed into a new genus, Heparna­ con­taminated water, in food, and by dirty hands.
virus, on the basis of its unique genome. Only one HAV outbreaks usually originate from a common
serotype is known. source (e.g. water supply, restaurant, daycare center).
Under crowded conditions and poor sanitation,
Resistance HAV infections occur at an early age. In India, type A
HAV is stable to treatment with 20% ether, acid hepatitis is the most common cause of acute hepatitis
(pH 1.0 for 2 hours), and heat (60°C for 1 hour). in children, but is much less frequent in adults.

www.ebook3000.com
466  |  Section 4: Virology
Table 67.1:  Comparative features of hepatitis viruses
A B C D E
Virus structure HAV, 27 nm RNA, HBV, 47 nm DNA HCV, 30–60 nm HDV, 35–37 nm HEV, 32–34 nm RNA
Picornavirus (Hepadnavirus) RNA, Flavivirus Defective RNA Herpes virus
(Hepatovirus) (hepacivirus) Deltavirus
Modes of Fecal-oral Parenteral Parenteral Parenteral Fecal–oral
infection Vertical, sexual
Age affected Children Any age Adults Any age Young adults
Incubation 15–45 30–180 15–160 30–180 15–60
period (days)
Onset Acute Insidious Insidious Insidious Acute
Illness Mild Occasionally Moderate Occasionally severe Mild, except in preg-
severe nancy
Carrier state Nil Common Present Nil (only with HBV) Nil
Oncogenicity Nil Present specially Present Nil Nil
after neonatal
infection
Prevalence Worldwide Worldwide Probably Endemic areas Only developing
worldwide (Mediterranean, N, countriest lndia,
Europe, Central and Asia, Africa, Central,
N. America) America)

Laboratory Ig and vaccine Ig and vaccine Nil HBV vaccine Nil


diagnosis
Specific
prophylaxis

vomiting and liver tenderness. These usually


subside with the onset of jaundice. The patient
starts to feel better within the next week or so and
the jaundice disappears within a month. Re­covery
is slow, over a period of 4–6 weeks.
Hepatitis A is nearly always self-limiting, but
relapses have been reported. The disease is milder
in children, in whom many infec­tions may be
anicteric.

Fig. 67.1: The picornavirus structure of hepatitis A virus. The Laboratory Diagnosis
icosahedral capsid is made up of four viral polypeptides (VP1– Etiological diagnosis of type A hepatitis may be
VP4). Inside the capsid is a single-stranded, positive-sense RNA made by demonstration of the virus or its antibody.
single stranded RNA (ssRNA) that has a genomic viral protein
(VPg) on the 5’ end A. Demonstration of the virus: The virus can be
visualized by immune electron microscopy
(IEM) in fecal extracts during the late incubation
Natural infection with HAV is seen only in period and the preicteric phase.
humans. Though primates, such as chimpanzees B. Demonstration of antibody: The best way
have been shown to acquire the infection from to demonstrate an acute HAV in­fection is by
humans and transmit it to human contacts, there finding anti-HAV immunoglobulin M (IgM), as
is no evidence of any extrahuman source of the measured by an enzyme-linked immunosorbent
virus in nature. assay (ELISA) or radioimmunoassay. Antibody
IgM anti-HAV antibody appears during the
Clinical Features late incubation period, reaches peak levels in
The incubation period is 2–6 weeks. Disease in 2–3 weeks and disappears after 3–4 months.
children is generally milder than that in adults IgG antibody appears at about the same time,
and is usually asymptomatic. The clinical disease peaks in 3–4 months and persists much longer,
consists of two stages: the prodromal or preicteric perhaps for life (Fig. 67.2). Demonstration of
and the icteric stages. The onset may be acute or IgM antibody in serum indicates current or
insidious, with fever, malaise, anorexia, nausea, recent infection, while IgG antibody denotes
Chapter 67: Hepatitis Viruses  | 467

Fig. 67.2: Typical course of hepatitis type A Fig. 67.3: Hepatitis B virus structure

recent or remote infection and immunity. ELISA virus (HBV) infects the liver and, to a lesser extent,
kits for detection of IgM and IgG antibodies are the kidneys and pancreas of only humans and
available. chim­panzees.
C. Virus isolation: It is not routinely performed.
Classification
Prophylaxis
HBV is assigned to a separate family Hepadnaviridae
A. General Measures (Hepatitis DNA viruses) which consists of two
The spread of HAV is reduced by interrupting the genera:
fecal–oral spread of the virus by avoiding potentially i. Orthohepadnavirus: It containing HBV as
contaminated water or food, water. well as the woodchuck and ground squirrel
hepatitis viruses. HBV is Hepadnavirus type 1.
B. Immunization ii. Avihepadnavirus: It containing the Pekin
1. Passive protection: Specific passive prophylaxis duck and gray heron hepatitis viruses.
by pooled normal human immunoglobulin
(16% solution in a dose of 0.2–0.12 mL/kg body Structure
weight) intramuscular (IM), before exposure
The HBV is a 42 nm DNA virus with an outer
or in early incubation period, can prevent or
envelope and an inner core, 27 nm in diameter,
attenuate clinical illness, while not necessarily
enclosing the viral genome and a DNA polymerase
preventing infection and virus excretion.
(Fig. 67.3) which is circular double stranded DNA.
2. Hepatitis A vaccine
i. Formalin inactivated, alum conjugaged Australia antigen: In 1965, Blumberg, studying
vaccine: A safe and effective formalin human serum lipoprotein allotypes, observed in
inactivated, alum conjugated vaccine the serum of an Australian aborigine, a new antigen
containing HAV grown in human diploid which gave a clearly defined line of precipitation
cell culture is available for use in children with sera from two hemophiliacs who had received
and adults at high risk for infection, multiple blood transfusions. This was named the
especially travelers to endemic regions. A Australia antigen. By 1968 the ‘Australia antigen’
full course consists of two intramuscular was found to be associated with serum hepatitis.
injections of the vaccine. Protection begins It was subsequently shown to be the surface
4 weeks after injection and lasts for 10–20 component of HBV. Therefore, the name Australia
antigen was changed to hepatitis B surface antigen
years.
(HBsAg).
ii. Live HAV vaccine: It has been developed
in China. Types of particles: Under the electron microscope,
sera from type B hepatitis patients show three types
Treatment of particles (Fig. 67.4).
Treatment is symptomatic. No specific antiviral i. Spherical particle: The predominant form is
drug is available. a small, spherical particle with a diameter of
22 nm.
ii. Tubular particle: The second type of particle
Hepatitis B Virus—serum hepatitis
is filamentous or tubular with a diameter of 22
Type B hepatitis is the most widespread and the nm and of varying length. The particles carry
most important type of viral hepatitis. Hepatitis B the hepatitis B surface antigen (HBsAg).

www.ebook3000.com
468  |  Section 4: Virology
iii. Dane particle: The third type of particle, far 2. Hepatitis B core antigen (HBcAg): The antigen
fewer in number, is a double walled spherical expressed on the core is called the hepatitis B
structure, 42 nm in diameter. This particle core antigen (HBcAg).
is the complete hepatitis B virus. It was first 3. Hepatitis B e antigen (HBeAg): Hepatitis Be
described by Dane in 1970 and so is known antigen (HBeAg) is a soluble nonparticulate
as the Dane particle.The outer surface, or nucleocapsid protein. The HBeAg and HBcAg
envelope, contains HBsAg and surrounds a proteins share most of their protein sequence.
27-nm inner nucleocapsid core that contains 4. Viral genes and antigens: The genome has
HBcAg. The variable length of a single- a compact structure with four overlapping
stranded region of the circular DNA genome genes. These include structural proteins of the
results in genetically heterogeneous parti­cles virion surface and core, a small transcriptional
with a wide range of buoyant densities. trans­activator (X), and a large polymerase (P)
Genome: The nucleocapsid encloses the viral protein that includes DNA polymerase, reverse
genome consisting of two linear strands of DNA held transcriptase, and RNase H activities (Table
in a circular configuration. One of the strands (the 67.2, Fig. 67.5).
plus strand) is incomplete, so that the DNA appears
partially double stranded and partially single HBV Subtypes
stranded. Associated with the plus strand is a viral The particles containing HBsAg are antigenically
DNA polymerase, which has both DNA-dependent complex. It contains two different antigenic
DNA polymerase and RNA-dependent reverse components—the common group reactive antigen
transcriptase functions. Although a DNA virus, a, and two pairs of type specific antigens d-y and
it encodes a reverse transcriptase and replicates w-r, only one member of each pair being present at
through an RNA intermediate. This polymerase a time. HBsAg can thus be divided into four major
can repair the gap in the plus strand and render the antigenic subtypes: adw, adr, ayw and ayr.
genome fully double stranded (Fig. 67.3). They show a distinct geographical distribution.
(Table 67.3).
Antigenic Structure
1. Hepatitis B surface antigen (HBsAg): The Cultivation
envelope proteins expressed on the surface
HBV does not grow in any conventional culture
of the virion and the surplus 22 nm diameter
system. However, limited production of the virus
spherical and filamentous particles constitute
and its proteins can be obtained from several cell
the hepatitis B surface antigen (HBsAg).
lines transfected with HBV DNA. HBV proteins have
been cloned in bacteria and yeast. The chimpanzee
is susceptible to experimental infection and can be
used as a laboratory model.

Stability
The HBV is a relatively heat-stable virus. It remains
viable at room temperature for long periods. Heating
to 60°C for 10 hours inactivates virus by a factor of
100–1000-fold. Exposure to hypochlorite (10,000
ppm available chlorine) or 2% glutaraldehyde for 10
Fig. 67.4: Different types of particles of HBV: Spherical 22 minutes will inactivate virus 100,000-fold, though
nm particle, double shelled 42 nm particle (Dane particle), HBsAg may not be destroyed by such treatment.
tubular 22 nm particle

Table 67.2:  Genes coding for antigens of HBV


Gene Regions Antigen
S
(Having three regions S,
Pre-S1 and Pre-S2)
S
S + Pre-S2
S + Pre-S1 and S2
Major protein (S) Surface
Middle protein (M) antigen (HBsAg) }
Large protein (L)—Present only in virion
C C Core antigen (HBcAg)
(Having two regions C and C + Pre-C HBeAg
Pre-C)
P DNA polymerase
X HBx Ag (Nonparticulate antigen which leads to enhanced replication
of HBV)
Chapter 67: Hepatitis Viruses  | 469
Hepatocytes carry viral antigens and are subject
to antibody-dependent NK cell and cytotoxic T
cell attack. In the absence of adequate immune
response, HBV infection may not cause hepatitis,
but may lead to carrier state.

Epidemiology
The HBV is worldwide in distribution. Natural
infection occurs only in humans. There is no animal
reservoir. The virus is maintained in the large
pool of carriers whose blood contains circulating
virus for long periods, in some even lifelong.
The prevalence of HBV infection varies widely in
different parts of the world.
India falls in the intermediate group, with
Fig. 67.5: The HBV genes and gene products higher carrier rates in the southern part of the
country and lower rates in the northern part. The
Table 67.3:  Antigenic types of HBsAg rich and the poor countries also differ in the age
and modes of infection.
Antigenic types Distribution
adw Worldwide Mode of Transmission
adr Asia
The HBV is a blood borne virus and there are three
ayw India, Africa, Russia important modes of transmission:
ayr India, Africa, Russia 1. Parenteral transmission
2. Perinatal transmission
Clinical Syndromes 3. Sexual transmission.
Acute Infection 1. Parenteral transmission: HBV is transmitted
The clinical presentation of HBV in children is less only in blood and other body fluids, including
severe than that in adults, and infection may even cervical secretions, semen, and breast milk. Many
be asymptomatic. Clinically apparent illness occurs other therap­eutic, diagnostic, prophylactic and
in as many as 25% of those infected with HBV. even nonmedical procedures are now the main
1. Preicteric phase: HBV infection is characterized modes of infection. HBV is very highly infectious
by a long incubation period (about 1–6 months) far more that HIV.
and an insidious onset. Symptoms during the 2. Perinatal transmission: Vertical transmission
prodromal period may include fever, malaise, from mother to child is one of the most
and anorexia, followed by nausea, vomiting, important routes. HBV can be transmit­ted to
abdominal dis­comfort, and chills. babies through contact with the mother’s blood
2. Icteric phase: The classic icteric symptoms of at birth and in mother’s milk.
liver damage (e.g. jaundice, dark urine, pale 3. Sexual transmission: Since HBV is present in
stools) follow soon thereafter. semen and vaginal secretions, therefore, it can
3. Convalescent phase: About 90–95% of adults be transmitted by sexual contact. The risk of
with acute hepatitis B infection recover within transmission by heterosexual and homosexual
1–2 months of onset and eliminate the virus from contact increases with the number of partners
the body within about six months, remaining and the duration of such relationships.
immune thereafter. Mortality is about 0.5–2%.
Chronic infection: A proportion of cases (1–10%) Hepatitis B Carriers
remain chronically infected. They may be asympto­ Carriers are of two types:
matic carriers or may progress to recurrent or chronic 1. Super carriers: They have HBeAg, high titters
liver disease or cirrhosis. A few of them may develop of HBsAg and DNA polymerase in their blood.
hepatocellular carcinoma after many decades. HBV may also be demonstrable in their blood.
Very minute amount of serum or blood from
Pathogenesis such carriers can transmit the infection. About a
The pathogenesis of hepatitis appears to be immune quarter of the carriers in India are HBeAg positive.
mediated. HBV replicates in the hepatocytes, 2. Simple carriers: These are more common
reflected in the detection of viral DNA and HBcAg types of carriers who have low titer of HBsAg
in the nucleus and HBsAg in the cytoplasm and at in blood, with negative HBeAg, HBV and DNA
the hepatocyte membrane. polymerase. They transmit the infec­tion only

www.ebook3000.com
470  |  Section 4: Virology
when large volumes of blood are transferred indicative of viral replication. HBcAg is not
as in blood transfusion. Many super carriers in demonstrable in circulation. It is the earliest
time become simple carriers. antibody marker to be seen in blood, long
before anti-HBe or anti-HBs. As anti-HBc
HBV Markers remains lifelong, it serves as a useful indicator
The main antigens HBsAg, HBcAg, and HBeAg of prior infection with HBV; even after all the
each induce corresponding antibodies. With other viral markers become undetectable.
the exception of HBcAg, all these antigens and Initially, anti-­H Bc is predominantly IgM,
antibodies, together with the viral DNA polymerase, but after about 6 months, it is mainly IgG.
can be detected in the blood at various times after Selective tests for IgM or IgG anti-HBc
infection and are referred to as ‘markers’ (Table therefore enable distinction between recent
67.4). HBcAg is readily detectable only in the or remote infection respectively.
hepatocyte nuclei. iii. HBeAg: HBeAg presence denotes high
infectivity and its absence, along with the
Laboratory Diagnosis presence of anti-HBe, indicates low infectivity.
HBeAg appears in blood concurrently with
Specific Diagnosis HBsAg, or soon afterwards. Circulating HBeAg is
Specific diagnosis of hepatitis B tests on serological an indicator of active intrahepatic viral replication,
demonstration of the viral markers and can be and the presence in blood of DNA polymerase,
carried out by detection of HBsAg, anti-HBs, HBV DNA and virions, reflecting high infectivity.
HBeAg, anti-HBe, IgM anti-HBc, IgG anti-HBc Before HBsAg disap­p ears, HBeAg is replaced
and HBV DNA in the serum. The sequence of by anti-HBe, signaling the start of resolution of
appearance of viral markers in the blood is shown the disease. Anti-HBe levels often are no longer
in Figure 67.6. These can be detected by sensitive detectable after 6 months.
and specific tests like ELISA and RIA.
2. Viral DNA Polymerase
1. Detection of Viral Markers
DNA polymerase activity, HBV DNA, and HBeAg,
i. HBsAg: HBsAg is the first marker to appear which are representative of the viremic stage of
in blood after infection. HBsAg is usually hepatitis B, occur early in the incubation period,
detectable 2–6 weeks in advance of clinical con­currency or shortly after the first appearance
and biochemical evidence of hepatitis and of HBsAg.
persists throughout the clinical course of
the disease. In the typical case, it disappears
3. Polymerase Chain Reaction (PCR)
within about 2 months of the start of clinical
disease, but may sometimes last for 6 months Like HBeAg, HBV DNA is also an indicator of viral
and even beyond but typically disappears by replication and infectivity. Molecular methods,
the sixth month after exposure. such as DNA:DNA hybridization and PCR, at
ii. HBcAg: High levels of IgM-specific anti-HBc present used for HBV DNA testing are highly
are frequently detected at the onset of clinical sensitive and quantitative. HBV DNA level in serum
illness. Because this anti­b ody is directed reflects the degree of viral, replication in the liver
against the 27 nm internal core compo­ and so helps assess the progress of patients with
nent of HBV, its appearance in the serum is chronic hepatitis under antiviral chemotherapy.

Table 67.4:  Interpretation of serological markers in HBV infection


Clinical condition Serological tests
HBsAg HBeAg Anti-HBS Anti-HBe Anti-HBc
IgM IgG
Late incubation period or early hepatitis + + – – – –
Acute hepatitis + + – – + –
Late/chronic HBV infection + ±* – – – +
Simple carrier + – – – – +
Super carrier + + – – – +
Past infection – – + + _ +
Immunity following vaccination – – + – _ _
*When +, it indicates high infectivity while – indicates low infectivity.
Chapter 67: Hepatitis Viruses  | 471
source was human plasma, limited in
availability and not totally free from
possible risk of unknown pathogens.
2. Recombinant yeast hepatitis B vaccine:
The currently preferred vaccine is
genetically engineered by cloning the S
gene of HBV in baker’s yeast. It consists
of nonglycosylated HBsAg particles
alone. This vaccine is safe, antigenic, free
from side effects and as immunogenic
Fig. 67.6: Hepatitis antigens, antibodies and DNA in a
patient recovering from acute HBV infection as plasma-derived vaccine. It is given
with alum adjuvant, IM into the deltoid
or, in infants into the anterolateral
4. Biochemical Tests
aspect of the thigh. Gluteal injection is
In acute viral hepatitis caused by various hepatitis not recommended as it may result in
viruses, levels of serum transaminases (amino­ poor immune response. Three doses
transferases) are increased 5–100-fold. Both given at 0, 1 and 6 months constitute
alanine and aminotransferase and aspartate the full course. Seroconversion occurs
aminotransferase, rise together late in the incubation in about 90% of the vaccinees.
period. Peak level is obtained about the time jaundice 3. Recombinant Chinese hamster ovary
appears and reverts to normal in next 2 months. (CHO) cell hepatitis vaccine: Expression
Serum bilirubin levels may rise up to 25-fold. system of CHO cells has been successfully
used and the product is commercially
Prophylaxis available. This is the first vaccine using
Measures for the control of HBV infection are the mammalian cell expression system.
same as those for HIV infection. 4. Synthetic peptide vaccines: These are
A. General prophylaxis: consists in avoiding chemically synthesized polypep­t ide
risky practices like promiscuous sex, injectable vaccines and are safe and cheap. These
drug abuse and direct or indirect contact with are still under experimental stage.
blood, semen or other body fluids of patients 5. Hybrid virus vaccine: Potential live
and carriers. vacc­ines using recombinant vaccinia
B. Immunization: virus have been prepared for hepatitis
I. Passive immunization: Hyperimmune B, influenza,rabies, Epstein–Barr and
hepatitis B immune globulin (HBIG)- human immunodeficiency viruses.
Hyperimmune hepatitis B immune globulin
(HBIG) prepared from human volunteers Treatment
with high titer anti-HBs, administered 1M in No specific antiviral treatment is available for acute
a dose of 300–500 iu soon after exposure to HBV infection. Hepatitis B immune globulin may
infection constitutes passive immunisation. be ad­ministered within a week of exposure and
It may not prevent infection, but protects to newborn infants of HBsAg-positive mothers.
against illness and the carrier state. Interferon alpha, alone or in combination, with
HBIG must be given as soon as possible other antiviral agents, such as lamivudine and
after an accident and preferably within 48 famcyclovir, has been beneficial in some cases of
hours. A second dose is given 4 weeks later to chronic hepatitis.
those who do not respond to current vaccines.
If the victim has not been vaccinated, HBIG
Hepatitis C Virus (HCV)
should be used and a course of active
immunization started, injecting the two Hepatitis C virus (HCV) resembles flaviviruses
materials into different body sites. in structure and organization, and has been
II. Active immunization: classified as a new genus Hepacivirus in the family
1. Plasma-derived hepatitis B vaccine: Flaviviridae. HCV is a 50–60 nm virus with a linear
The initial vaccine was prepared by single stranded RNA genome, enclosed within
purifying HBsAg associated with the a core and surrounded by an envelope, carrying
22 nm particles from healthy HBsAg- glycoprotein spikes.
positive carriers and treating the particles The virus shows considerable genetic and
with virus-inactivating agents (formalin, antigenic diversity. Various viruses can be
urea, heat). This was immunogenic, differentiated by RNA sequence analysis into at
but became unacceptable because its least six major genotypes (clades) and more than

www.ebook3000.com
472  |  Section 4: Virology
70 subtypes. Because of this diversity there is little HEPATITIS D virus (HDV) (DELTA agent)
heterologous or even homologous postinfection
immunity in hepatitis C. This curious little agent was first detected in
people undergoing exacerbations of chronic HBV
The virus has not been grown in culture, but
infections. In 1977, Rizzetto et al. in Italy identified
has been cloned in Escherichia coli
a new viral antigen in the liver cell nuclei of patients
Mode of infection: Infection is mainly by blood infected with hepatitis B virus. This has been
transfusion and other modes of contact with infected shown to be due to the hepatotropic virus delta or
blood or blood products. Injectable drug abusers, hepatitis D. HDV is unique in that it uses HBV and
transplant recipients and immunocompromised target cell proteins to replicate and produce its one
persons are at high risk. Sexual transmission protein. Delta is a defective RNA virus dependent
is probably less important. The virus can be on the helper function of HBV for its replication
transmitted from mother to infant. and expression. It acquires an HBsAg coat for trans­
Infections by HCV are extensive throughout the mission. Therefore, it has no independent existence
world. HCV infection is seen only in humans. The and can survive and replicate only as long as HBV
groups at risk are broadly similar to those listed infection persists in the host. It is often associated
for hepatitis B but their relative proportions are with the most severe forms of hepatitis in HBsAg-
different. positive patients. It is a viral parasite, proving that
“even fleas have fleas.”
Clinical Features The HDV is enclosed within the hepatitis B
The incubation period is long, 15–160 days, with a surface antigen, HBsAg, and has no recognizable
mean of 50 days. morphology of its own.
HCV causes three types of disease: The HDV is a defective satellite virus requiring
i. Acute hepatitis with resolution of the infect­ HBV as helper virus. HDV is a spherical, 36 nm
ion and recovery in 15% of cases. particle with an outer coat composed of the
ii. Chronic persistent infection with pos­sible hepatitis B surface antigen surrounding the circular
progression to disease much later in life for 70%. single stranded RNA genome. The HDV RNA
iii. Severe rapid progression to cirrhosis in 15% of genome is very small, single­stranded RNA and
patients—Many patients develop cirrhosis and circular. Delta agent is thus an incomplete virus,
are at high risk for hepatocellular carcinoma reminiscent of the Dependoviruses (Fig. 67.7).
(5–25%) decades later. It has been classified in a new genus Deltavirus,
because of its special features.
Laboratory Diagnosis
Pathogenesis
A. Antobody detection: The diagnosis and
detection of HCV infection are based on ELISA Its mode of transmission is the same as for HBV.
recognition of antibody. The antigens used are Similar to HBV, the delta agent is spread in blood,
various structural and nonstructural proteins semen, and vaginal secretions. However, it can
cloned in E. coli. Confirmation by immunoblot repli­cate and cause disease only in people with
assay is therefore recommended. In HCV active HBV infections.
infection antibodies appear irregularly and late,
limiting their diagnostic utility.
B. HCV RNA identification: Reverse transcriptase-
polymerase chain reaction, branched-chain
DNA, and other genetic techniques can detect
HCV RNA in seronegative people and have
become key tools in the diagnosis of HCV
infection.

Prophylaxis
Only general prophylaxis, such as screening of
blood and blood products prior to transfusion, is
possible. No specific active or passive immunizing
agent is available.

Treatment
Recombinant interferon-alpha, alone or with
ribavarin is the only known treatment for HCV. Fig. 67.7: Delta hepatitis virion
Chapter 67: Hepatitis Viruses  | 473
Types of infection: Two types of infection are young to middle aged adults (15–40 years old). The
recognized: symptoms and course of HEV disease are simi­lar
1. Coinfection: In coinfection, delta and HBV are to those of HAV disease; it causes only acute dis­
transmitted together at the same time. ease. However, the symptoms for HEV may occur
2. Superinfection: In superinfection, delta later than those of HAV disease, and response
infection occurs in a person already harboring to se­rum immunoglobulin G may be poor. The
HBV. More rapid, severe progression occurs in mortality rate associated with HEV disease is 1–2%.
HBV carriers super­infected with HDV than in HEV infection is especially serious in pregnant
people coinfected with HBV and the delta agent. women and during pregnancy may cause a high
No association has been noted between HDV rate of abortion, intrauterine death and increased
and hepatocellular carcinoma. In simultaneous perinatal mortality in babies born to women with
acute HBV and HDV infec­tions, IgM anti-HBc fulminant hepatitis.
will be detectable, while in acute HDV infection
superimposed on chronic HBV infections, anti- Epidemiology
HBc will be of IgG class. HEV is predominantly spread by the feco-oral
route, especially in contaminated water. Hepatitis
Laboratory Diagnosis E has been shown to occur in epidemics, endemics
The only way to determine the presence of the and sporadic forms almost exclusively in the
agent is by detecting the delta antigen or antibodies. less developed parts of the world. A substantial
ELISA and radioimmunoassay procedures are proportion of cases of acute viral hepatitis occurring
available for do­ing this. Anti-delta antibodies in young to middle-aged adults in Asia and the
appear in serum and can be identified by ELISA. Indian subcontinent appear to be caused by HEV.
The IgM antibody appears 2–3 weeks after infection The largest such epidemic occurred in Delhi
and is soon replaced by the IgG antibody in acute during the winter of 1955–56, following a breakdown
delta infection. in the water supply and sewage systems of Delhi,
For the rapid identification of delta particles in affecting over 30,000 persons. The cause was
circulation, RNA sequences have been cloned and eventually identified as a new agent, HEV. A similar
DNA probes have been developed. The woodchuck epidemic of hepati­tis E occurred between December
has been found to be a suitable experimental 1975 and Janu­ary 1976 in Ahmedabad city, India.
model for the study of HDV infection.
Laboratory Diagnosis
Treatment Several antibody assays are being developed,
There is no known specific treatment for HDV mostly based on ELISA methods.
hepati­tis. 1. Exclusion of hepatitis A and hepatitis B:
Exclusion of hepatitis A by IgM serology and
Prophylaxis hepatitis B by absence of HBsAg and IgM anti-
Because the delta agent depends on HBV for repli­ HBc.
cation and is spread by the same routes, prevention 2. Immunoelectron microscopy: Immunoelectron
of infection with HBV prevents HDV infection. microscopic examination of patient feces for
Immuni­zation with HBV vaccine protects against aggregated calicivirus-like particles using
subsequent deltavirus infection. monoclonal antibodies.
3. ELISA tests: for IgM and IgG anti-HEV.
Hepatitis E Virus (HEV) 4. Western blot assay: A Western blot assay for
IgM and/or IgG anti­HEV.
Hepatitis E virus (HEV) has been provisionally
5. Polymerase chain reaction (PCR): Polymerase
classified in the genus Hepevirus under the family
chain reaction (PCR) assay for the detection of
Caliciviridae. HEV is a spherical nonenveloped
HEV RNA (as cDNA) in patient feces or in acute-
virus, 32–34 nm in diameter, with a single
phase sera.
stranded RNA genome. The surface of the virion
shows indentation and spikes. HEV (E-NANBH)
(The E stands for “Enteric” or “epidemic”) is Prophylaxis
predominantly spread by the feco-oral route, General measures: These depend on the mainten­
especially in contaminated water. ance of a clean water supply, and generally
resemble those used to control HAV.
Clinical Features Immunization: Vaccines based on recombinant
The incubation period ranges from 2–9 weeks with antigens are under develop­ment, and show some
an average of six weeks. Most cases occur in the promise.

www.ebook3000.com
474  |  Section 4: Virology
Hepatitis G Virus encloses an inner icosahedral 27 nm nucleocapsid
(core), which contains hepatitis B core antigen
Two flavivirus-like isolates were’ obtained in 1995
(HBcAg). Inside the core is the genome, a circular
from Tamarin monkeys inoculated with blood from double stranded DNA and a DNA polymerase
a young surgeon (GB) with acute hepatitis. It was ™™ Hepatitis B surface antigen (H BsAg) is also named
termed GB, the patient’s initials. A similar virus was as Australia antigen
isolated from another human specimen the same ™™ Mode of transmission: HBV is a blood borne
year. These isolates were called GB viruses A, B virus and there are three important modes of
and C respectively. transmission: 1. Parenteral transmission; 2.
Perinatal transmission; 3. Sexual transmission
In 1996, an isolate closely resembling GBV-C ™™ Laboratory diagnosis: Specific diagnosis of hepatitis
was obtained from a patient with chronic hepatitis. B rests on serological demonstration of the viral
This has been called hepatitis G virus (HGV). markers and can be carried out by detection of
Hepatitis G virus resembles HCV in many ways. HBsAg, anti-HBs, HBeAg, anti-HBe, IgM anti-HBc, IgG
HGV is a flavivirus, is transmitted in blood, and has anti-HBc and HBV DNA in the serum. These can be
detected by sensitive and specific tests like ELISA
a predi­lection for chronic hepatitis disease. It has not
and RIA. Molecular methods such as DNA: DNA
been grown, but its RNA genome has been cloned. hybridization and PCR are used for HBV DNA testing
The HGV RNA has been found in patients with are highly sensitive and quantitative
acute, chronic and fulminant hepatitis, hemophiliacs, Prophylaxis: General prophylaxis consists in
patients with multiple transfusions and hemodialysis, avoiding risky practices
intravenous drug addicts and blood donors. HGV Immunization: I. Passive immunization: Hyper­
appears to be a blood borne virus resembling HCV. immune hepatitis B immune globulin (HBIG)
administered 1 M in a dose of 300–500 iu soon after
Its role in hepatitis is yet to be clarified. exposure to infection
The virus is present worldwide. Majority of the II. Active immunization: such as plasma-derived
individuals with HGV infection have no detectable hepatitis B vaccine, recombinant yeast hepatitis
evidence of liver disease. There have been, how­ever, B vaccine (Three doses given at 0, 1 and 6 months
cases of acute, fulminant and chronic hepatitis constitute the full course), recombinant Chinese
hamster ovary (CHO) cell hepatitis vaccine,
where HGV is presently the only explanation for
synthetic peptide vaccines and hybrid virus vaccine
their liver disease. There is no evidence of a causal
Hepatitis C Virus
relationship between HGV infection and hepato­
™™ HCV can cause acute HCV infection, chronic HCV
cellular carcinoma. HGV infection results frequently infection, and cirrhosis and other complications
in chronic viraemia. It often subsides after several induced by hepatitis
years and anti-HGenv antibody develops. ™™ Blood or blood products and also organs of infected
patients are the major sources of infection
™™ For diagnosis, antibody to HCV antigen can be
Laboratory Diagnosis detected by enzyme immunoassay
1. HGV is identified by detection of the genome by ™™ The HCV RNA can be amplified by RT-PCR
reverse transcriptase ­polymerase chain reaction Hepatitis D Virus
(RT-PCR) or other RNA detection methods. ™™ The hepatitis D virus (HDV) is unique in being an
2. An immunoassay has been developed to detect incomplete virus, that requires hepadnavirus helper
anti-HG env. Serum HGV RNA indicates viremia, functions for propagation in hepatocytes
™™ Transmission of HDV occurs parenterally
whereas anti-HGenv is associated with recovery.
™™ HDV superinfections in patie nts with HBV results in
Key Points chronic HDV infection
™™ Vaccination with HBV vaccine protects against
™™ At least six viruses, A through G (A, B, C, D, E and G.) subsequent HDV infection
are hepatitis viruses Hepatitis E virus (HEV)
™™ All the hepatitis viruses are RNA viruses, except ™™ Hepatitis E virus is the primary cause of enterically
the hepatitis B virus (HBV), which is a DNA virus transmitted non-A, non-B hepatitis
belonging to the family Hepadnaviridae ™™ It usually causes an acute, self-limiting disease similar
™™ Hepatitis A virus (HAV) to HAV. Pregnant women—high mortality associated
™™ Hepatitis A virus (HAV) can be demonstrated in the with HEV
stool by immunoelectron microscopy (IEM). ELISA is ™™ Hepatitis G virus (HGV) is a flavivirus, is transmitted
the method for detection of IgM and IgG antibodies in blood, and has a predi­lection for chronic hepatitis
in the serum disease.
™™ Prevention of HAV infection depends on: vaccines
containing formalin-inactivated HAV
Hepatitis B Virus
™™ HBV is associated with hepatocellular carcinoma Important questions
™™ Hepatitis B virus (HBV) is a double-walled spherical
1. Name the hepatitis viruses. Describe the morphology
structure and measures 42 nm in diameter (Dane
and antigenic structure of hepatitis B virus.
particle). The outer surface or envelope of virus
2. Classify hepatitis viruses. Discuss the laboratory
contains hepatitis B surface antigen (HBsAg). It
diagnosis of infections caused by hepatitis B virus.
Chapter 67: Hepatitis Viruses  | 475
3. Write short notes on: 8. Which of the following viral markers is first marker to
a. Hepatitis A virus (HAV) or type A hepatitis or appear in biood after infection with hepatitis B virus?
infectious hepatitis. a. HBsAg
b. Hepatitis B virus or Dane’s particle. b. HBcAg
c. Hepatitis B surface antigen (HBsAg) or Austral­ c. HBeAg
ia antigen. d. Antibody to HBsAg
d. Draw a neat labeled diagram of hepatitis B virus. 9. Which of the following viral markers is/are positive
e. Hepatitis B virus markers. in simple carriers of hepatitis B?
f. Hepatitis C virus or type C hepatitis. a. HBsAg
g. Hepatitis D virus or Delta agent. b. HBeAg
h. Non-A, Non-B hepatits c. Both of the above
i. Hepatitis E virus d. None of the above
j. Hepatitis G virus 10. In super carriers of hepatitis B which of the following
k. Pathogenesis of HBV or type B hepatitis or viral markers is/are positive?
serum hepatitis or serum jaundice or transfu­ a. HBsAg
sion hepatitis. b. HBeAg
l. Laboratory diagnosis of type B hepatitis. c. Both of the above
m. Prophylaxis of hepatitis B or hepatitis B vaccine. d. None of the above
11. High infectivity of hepatitis B virus is indicated by
which of the following markers when positive?
Multiple choice questions (mcqs) a. HBsAg
1. Which of the following hepatitis viruses is DNA b. HBeAg
c. HBcAg
virus?
d. None of the above
a. Hepatitis A virus
12. For preparation of recombinant hepatitis B vaccine
b. Hepatitis B virus
which of the following hepatitis B genes are used?
c. Hepatitis C virus
a. HBsAg gene b. HBeAg gene
d. Hepatitis E virus
c. HBcAg gene d. All of the above
2. Which of the following hepatitis viruses can be
13. The carrier state in hepatitis B virus infection is
transmitted by sexual route?
demonstrated by:
a. Hepatitis A virus a.  Persistent presence of HBsAg in blood for at
b. Hepatitis C virus’ least 6 months
c. Hepatitis E virus b.  Persistent presence of H and Ab in blood for at
d. All of the above least 6 months
3. Which of the following hepatitis viruses may cause c.  Persistent presence of HBeAg in blood for at
hepatic carcinoma? least 6 months
a. Hepatitis A virus d.  Persistent presence of H and Ag in blood for at
b. Hepatitis C virus least 6 months
c. Hepatitis E virus 14. All the following statements are true for hepatitis C
d. Hepatitis G virus virus except:
4. Which of the following hepatitis viruses is non­ a. It is closely related to hepatitis D virus
enveloped? b. It is a leading cause of cirrhosis
a. Hepatitis A virus c.  Blood transfusion is the most important mode
b. Hepatitis B virus of trans­mission of HCV
c. Hepatitis C virus d. A live vaccine is available against HCV
d. Hepatitis D virus 15. Which statement is true for hepatitis D virus:
5. All the following statements are true for hepatitis A a. An incomplete virus
virus (HA V) except: b. Related to hepatitis B
a. It is most commonly transmitted by fecal-oral c. A single-stranded DNA
route d. Transmitted by fecal-oral route
b. It causes chronic disease or fatal disease 16. Hepatitis E virus is:
c. t has a short incubation period of 3–4 weeks a. Related to hepatitis C virus
d. There is only one serotype of HAV b. Yet to be cultured
6. Which of the following methods is preferred for c. Associated with chronic liver infection
diagnosis of hepatitis A virus (HAV) infection? d. A major cause of hepatitis transmitted by blood
a. Alanine aminotransferase levels 17. Which of the following statement is true for hepatitis
b. Detection of IgM antibody to HAV by ELISA G virus (HGV):
c. Detection of IgG antibody to HAV by ELISA a. HGV is a flavivirus
d. Cell cultures for growing HAV b. Hepatitis G virus resembles hepatitis C virus
(HCV)
7. Which of the following antigens is present in
c. Hepatitis G virus is transmitted in blood
the envelope of hepatitis B virus?
d. All of the above
a. HBsAg
b. HBcAg Answers (MCQs)
c. HBeAg 1. b; 2. b; 3. b; 4. a; 5. b; 6. b; 7. a; 8. a; 9. a; 10. c; 11. b; 12.
d. None of the above a; 13. a; 14. d; 15. a; 16. b; 17. d

www.ebook3000.com
68
Chapter
Retroviruses: Human
Immunodeficiency Virus (HIV)

Learning Objectives

After reading and studying this chapter, you should ∙∙ D iscuss opportunistic infections associated with
be able to: human immunodeficiency virus infection
∙∙ Classify retroviruses. ∙∙ D escribe the laboratory diagnosis of human
∙∙ Describe morphology of human immunodeficiency immuno­deficiency virus infection
virus ∙∙ D escribe the laboratory tests for detection of
∙∙ Describe the antigenic structure of human immuno­ specific antibodies in human immunodeficiency
deficiency virus (HIV) and draw labeled diagram of virus infection
HIV-I ∙∙ Describe strategies of human immunodeficiency
∙∙ Describe modes of transmission of human immuno­ virus testing
deficiency virus ∙∙ Describe the following: Antiviral therapy for HIV;
∙∙ Describe types of exposure and their relative risks postexposure prophylaxis.

Retroviruses In 1983, Luc Montagnier and colleagues from


the Pasteur Institute, Paris, isolated a retrovirus
These are RNA viruses that belong to family and called it lymphadenopathy associated virus
Retroviridae (Re = Reverse, tr = transcriptase). (LAV) In 1984, Robert Gallo and colleagues from
Members of this family possess an RNA genome the National Institutes of Health, USA, reported
and the characteristic biochemical feature is the isolation of a retrovirus from AIDS patients and
presence of RNA­dependent DNA polymerase called it human T cell lymphotropic virus-III
(reverse transcriptase) within the virus. (HTLV-III). The International Committee on Virus
Nomen­clature in 1986 decided on the generic
Classification
name—human immunodeficiency virus (HIV)
The family Retroviridae is classified into three for these viruses. HIV occurs in two types: HIV-1
subfamilies (Table 68.1). and HIV-2. Luc Montagnier was awarded Nobel
prize in 2009 for the discovery of HIV.
Human immunodeficiency virus
Human immunodeficiency virus-2 (HIV-2): A
Human immunodeficiency virus (HIV) types, fourth human retrovirus, HIV-2, was isolated from
derived from primate lentiviruses, are the etiologic mildly immunosuppressed patients in West Africa
agents of acquired immunodeficiency syndrome and appears to be less pathogenic.
(AIDS). The illness was first described in 1981,
and HIV-l was iso­lated by the end of 1983. The Structure
first indication of this new syndrome came in the 1. Enveloped virus: HIV is a spherical enveloped
summer of 1981, with reports from New York and virus, about 90–120 nm in size (Fig. 68.1).
Los Angeles (USA), of a sudden unexplained out­ 2. Nucleocapsid: The nucleocapsid has an outer
break of two very rare diseases—Kaposi’s sarcoma icosahedral shell and an inner coneshaped
and Pneumocystis carinii pneumonia (PCP) in core, enclosing the ribonucleoproteins.
young homosexuals or addicted to injected narcot­ 3. Genome: There are two identi­cal copies of the
ics. They appeared to have lost their immune positive sense, single-stranded RNA genome in
compe­tence. This condition was given the name the capsid (retroviruses are diploid). Also found
acquired immunodeficiency syndrome (AIDS). within the capsid are the enzymes reverse
Chapter 68: Retroviruses: Human Immunodeficiency Virus  | 477
Table 68.1:  Human retroviruses Viral Genes and Antigens
Subfamily Genus Virus Disease The genome organization is similar for all
Oncovirinae Retrovirus HTLV-1 Adult T cell leu­k­­ retroviruses in that their genomes contain in the
emia/lym­phoma same order the genes gag, pol and env, which code
HTLV-2 Prevalent in for the three groups of structural proteins (Figs 68.2
intravenous and 68.3). The genome of HIV contains the three
drug users, not structural genes (gag, pol and env) characteristic of
associated with
disease all retroviruses, as well as other nonstructural and
regulatory genes specific for the virus (Fig. 68.3).
Lentivirinae Lentivirus HIV-1 AIDS
HIV-2 AIDS
The products of these genes, both structural and-
nonstructural, act as antigens (Table 68.2). Sera
Spumavirinae Spumavirus Human Nil
foamy
of infected persons contain antibodies to them.
virus Detection of these antigens and antibodies is useful
in the diagnosis and prognosis of HIV infections
HTLV: Human T-cell lymphotropic virus
HIV: Human immunodeficiency virus (Table 68.3).
AIDS: Acquired immunodeficiency syndrome

Table 68.2:  Major antigens of HIV


A. Envelope antigens:
1. Spike antigen—gp120
(Principal envelope antigen)
2. Transmembrane pedicle protein—gp 41
B. Shell antigen
1. Nucleocapsid protein—p18
C. Core antigens
1. Principal core antigen—p24
2. Other core antigens—p15, p55
D. Polymerase antigens—p31, p51, p66

Table 68.3:  Retrovirus genes and their function


Gene Virus Function
gag All Group-specific antigen—core and
capsid proteins
Fig. 68.1: Structure of HIV (diagrammatic representation):
pol All Polymerase—reverse transcriptase,
1. Envelope glycoprotein spike (gp120), 2. Transmembrane
protease, integrase
pedicle glycoprotein (gp41), 3. Outer icosahedral shell of
nucleocapsid (p18), 4. Cone shaped core of nucleocapsid env All Envelope—glycoproteins
(p24), 5. Inner core, 6. Viral proteins associated with RNA, tax HTLV Transactivation of viral and cellular
7. Viral RNA, 8. Reverse transcriptase, 9. Envelope lipid bilayer genes
tat HIV-1 Transactivation of viral and cellular
genes
transcriptase, (which is a characteristic feature
of retroviruses). rex HTLV Regulation of RNA splicing and
promotion of export to cytoplasm
4. Lipoprotein envelope: During viral replication,
when the naked virus buds out through the rev HIV-1 Regulation of RNA splicing and
promotion of export to cytoplasm
host cell surface membrane, it ac­q uires a
lipoprotein envelope, which consists of lipid nef HIV-1 Alteration of cell activation signals;
derived from the host cell membrane and progression to AIDS (essential)
glycoproteins which are virus coded. The vif HIV-1 Virus infectivity, promotion of
major virus coded envelope proteins are the assembly
projecting knob-like spikes on the surface and vpu HIV-1 Facilitation of release of virus,
the anchoring transmembrane pedicles. The decrease of cell surface CD4
env polypeptide is composed of two subunits, vpr (vpx*) HIV-1 Transport of complementary DNA to
the outer glycoprotein knob (gp120) and a nucleus, arresting of cell growth
trans­membrane portion (gp41) which joins the LTR All Promoter, enhancer elements
knob to the virus lipid envelope. The receptor *In HIV-2
binding site for CD4 is present on gp120. HIV, human immunodeficiency virus; HTLV, human
Transmembrane pedicles cause cell fusion. T-lymphotropic virus; LTR, long-terminal repeat (sequence)

www.ebook3000.com
478  |  Section 4: Virology
A. Genes Coding for Structural Proteins primate species. There are two distinct types of
(Table 68.2) human AIDS viruses: HIV-l and HIV-2.
1. The gag (group-specific antigen) gene: The HIV-1 groups: Based on env gene sequences, HIV-1
gag gene determines the core and shell of the comprises three distinct virus groups—M (for
virus. It is expressed as a precursor protein, ‘major’), O (for ‘outlier’) and N (for new).
P55. This precursor protein is cleaved into M group: The predominant M group, which cause
three proteins, P15, p18 and p24. The four the large majority of HIV-1 infections worldwide,
proteins coded for by the gag gene of HIV are all contains nine subtypes or “clades” (A-K, omit­ting
found in the virion , including p24. The ma­jor E and I).
core antigen is p24 which can be detected in
se­rum during the early stages of HIV infection O group: A few HIV-l strains isolated from West
before antibodies appear. Late in the course of Africa (Cameroon, Gabon) do not fall within the
infection, the decline of free anti-p24 antibody Group M and have been designated group O.
and reappearance of p24 antigen in circulation N group: Some recent iso­lates of HIV-1 from
point to exacerbation of the illness. Cameroon, distinct from M and O groups have
2. The env: The env determines the synthesis of been called group N.
envelope glycoprotein gp160, which is cleaved
into the two envelope components—gp120 Human immunodeficency virus-2 (HIV-2): HIV
which forms the surface spikes and gp41, the strains, first isolated from West Africa in 1986,
transmembrane anchoring protein. The spike which react with HIV type 1 antiserum very weakly
glycoprotein gp120 is the major envelope or not at all have been termed HIV type 2. The
antigen, and antibodies to gp120 are present envelope antigens of the two types are different,
in circulation till the terminal stage of the though their core polypeptides show some cross
infection. reactivity. HIV-1 and the chimpanzee virus carry
3. The pol gene: The pol gene codes for the a vpu gene, whereas HIV-2 and most SIVs have a
protease, endonuclease, integrase and reverse vpx gene. The sequences of the gag and pol genes
tran­scriptase. It is expressed as a precursor are highly conserved. It is much less virulent than
protein, which is cleaved into proteins p31, p51 HIV-1. It is large­ly confined to West Africa, though
and p66. isolations have been reported from some other
areas, including west­ern and southern India.
B. Nonstructural and Regulatory Genes HIV-2 subtypes: Six subtypes of HIV-2 (A-F) have
There are at least six regulatory genes in HIV and been identified. HIV-2 has only 40% genetic identity
at least two in HTLV-I (Fig. 68.3). with HIV-I. It is more closely related to simian
1. tat (transactivating gene) enhancing the expres­ immunodeficiency virus than to HIV-1.
sion of all viral genes. HIV-1 subtypes show a geographical distribu­
2. nef (negative factor gene) down regulating viral tion. All known HIV virus groups and subtypes are
replication. present in Cameroon, West Africa. Subtype A is the
3. rev (regulator of virus gene) enhancing most prev­alent, being found worldwide, while B
expression of structural proteins. is the most common in the Americas and Europe.
4. vif (viral infectivity factor gene) influencing The common subtypes in Africa are A, C and D,
infec­tivity of viral particles. while in Asia the common subtypes are E, C and
5. vpu (only in HIV-1) and vpx (only in HIV-2) B. Subtype E is the most common in Thailand. In
enhancing maturation and release of progeny India and China, subtype C is the most prevalent.
virus from cells. (Detection of the type-specific
sequences vpu and vpx is useful in distinguishing Resistance
between infection by HIV-1 and 2). 1. Temperature: HIV is inactivated by heat, in the
6. vpr stimulating promoter region of the virus. autoclave or hot air oven. HIV is thermolabile,
The Vpr protein increases transport of the viral being inactivated in 10 minutes at 60°C and in
preintegration complex into the nucleus and seconds at 100°C.
also arrests cells in the G2 phase of the cell cycle. 2. Lyophilization: It withstands lyophilization.
7. LTR (long terminal repeat) sequences, one 3. Disinfectants: HIV is completely inactivated by
at either end, containing sequences giving treatment for 10 minutes at room temperature
promoter, enhancer and integration signals. with any of the following: 10% household bleach,
50% ethanol, 35% isopropanol, 1% Nonidet
Classification P40, 0.5% Lysol, 0.5% paraformaldehyde, or
Lentiviruses have been isolated from many species, 0.3% hydrogen per­oxide. The virus is also
including at least 26 different African nonhuman inactivated by extremes of pH (pH 1.0, pH
Chapter 68: Retroviruses: Human Immunodeficiency Virus  | 479
13.0). For treatment of contaminated medical The presence of other sexually transmitted
instruments, a 2% solution of glutaraldehyde is diseases such as syphilis, gonorrhea, or herpes
useful. simplex type 2 increases the risk of sexual HIV
The virus is not inactivated by 2.5% Tween 20. transmission. Transmission may be more likely
from male to female. Asymptomatic virus­positive
Routes of Transmission individuals can transmit the virus.
Virus is present in the blood, semen, and cervical
and vaginal secretions, and these sources are 2. Blood and Blood Products
important in transmission. HIV is spread only by Transfusion of infectious blood or blood products
three modes (Table 68.4): is an effective route for viral transmission.
1. Sexual contact with infected persons (hetero­
sexual or homosexual). Contaminated needle: It can transmits the
infection. This is particularly relevant in drug
2. By blood and blood products.
addicts. The use of unsterile syringes and needles
3. From infected mother to babies (intrapartum, by qualified and unqualified health workers makes
perinatal, postnatal). iatrogenic infection likely.
The modes of transmission of HIV and their
relative risks are shown in Table 68.5. 3. Mother-to-child Transmission
1. Sexual Intercourse Transmission of infection from mother to baby can
HIV is primarily a sexually transmitted infection. take place before, during or after birth. Mother-to-
Both sexes are affected equally. Transmission in the infant transmission rates vary from 13 to 40% in
developing countries is almost always heterosexual untreated women. Infants can become infected in
and can take place in both directions. utero, during the birth process, or, more commonly,
through breastfeeding. Transmission during
breastfeeding usually occurs early (by 6 months).
Table 68.4:  Transmission of HIV infection
Routes Specific transmission Pathogenesis
Known routes of transmission Infection is transmitted when the virus enters
1. Inoculation in blood Transfusion of blood and the blood or tissues of a person and comes into
blood products contact with a suitable host cell, principally the
Needle sharing among
CD4 lymphocyte. The major determinant in the
intrave­nous drug abusers
Needlestick, open wound, pathogenesis and disease caused by HIV is the
and mucous membrane virus tropism for CD4-ex­pressing T cells and cells
exposure in healthcare of the macrophage lineage.
workers HIV genome is uncoated and internalized into
Tattoo needles
the cell after fusion of the virus with the host cell
2. Sexual transmission Anal and vaginal intercourse membrane Viral reverse transcriptase mediates
3. Mother to baby Intrauterine transmission transcription of its RNA into double stranded
Peripartum transmission DNA, which is integrated into the genome of
Breast milk
the infected cell through the action of the viral
Routes not involved in transmission enzyme integrase, causing a latent infection. HIV
Close personal contact Household members causes lytic and latent infection of CD4 T cells
Healthcare workers not and persistent infection of cells of the monocyte
exposed to blood
macrophage family and disrupts neurons. The
outcomes of these actions are immunodeficiency
and acquired immuno­d eficiency syndrome
(AIDS) dementia. From time to time, lytic
Table 68.5:  Efficiency of different routes of trans­
infection is initiated releasing progeny virions
mission of HIV
which infect other cells. The long and variable
Route Efficiency incubation period of HIV infection is because of
Blood transfusion >90% the latency. HIV can be isolated from the blood,
Perinatal 13−40%
lymphocytes, cell-free plasma, semen, cervical
secretions, saliva, tears, urine and breast milk in
Sexual intercourse
an infected individual.
• Anal intercourse 1% per episode The primary pathogenic mechanism in HIV
• Vaginal intercourse 0.1% per episode
infection is the damage caused to the CD4+ T
Intravenous drug use 0.5−1% lymphocyte. The T4 cells decrease in numbers

www.ebook3000.com
480  |  Section 4: Virology
and the T4:T8 (helper: suppressor) cell ratio is Table 68.7:  Summary of classification system for
reversed. An important feature in HIV infection is HIV infection (Centers for Disease Control, USA)
the polyclonal activation of B lymphocytes leading
Group I Acute HIV syndrome
to hypergam­maglobulinemia.
In peripheral blood, lymphoid tissue and other Group II Asymptomatic infection
tissues such as brain where HIV replication occurs, Group III Persistent generalized
HIV targets CD4 positive (CD4+) cells and cells of lymphadenopathy
the monocyte­-macrophage lineage. The principal Group IV Other diseases:
immunological abnormalities seen in HIV infection Subgroup A Constitutional disease—ARC
are listed in Table 68.6.
Subgroup B Neurologic diseases
Clinical manifestations in HIV infections are
Subgroup C Secondary infectious diseases
due not primarily to viral cytopathology but are:
secondary to the failure of immune responses. This Category C1 Specified infectious
renders the patient susceptible to opportunistic diseases listed in the CDC
surveillance definition
infections and malignancies. for AIDS, such as P. carinii
pneumonia, cryptosporidiosis,
Clinical Features of HIV Infection toxoplasmosis, generalized
strongyloidiasis,
The Centers for Disease Control (USA) have classified cryptococcosis CMV or herpes
the clinical course of HIV infection under various diseases.
groups (Table 68.7). The natural evolution of HIV Category C2 Other specified secondary
infection can be considered in the following stages. diseases, such as oral hairy
leukoplakia, salmonella
Group I—Acute HIV Infection bacteremia, nocardiosis,
tuberculosis, thrush
The acute seroconversion illness: It resembles
Subgroup D Secondary cancers, such as
glandular fever, with adenopathy and flu-like Kaposi’s sarcoma, lymphomas
symptoms.Within 3–6 weeks of infection with HIV,
Subgroup E Other conditions.
about 50% of persons experience low grade fever,
malaise, headache, lymphadenopathy, sometimes
with rash and arthropathy resembling glandular fever. Group III—Persistent Generalized
Tests for HIV antibodies are usually negative at lymphadenopathy
the onset of the illness but become positive during This has been defined as the presence of enlarged
its course. HIV antigenemia (p24 antigen) can be lymph nodes, at 1east 1 cm in diameter, in two
demonstrated at the beginning of this phase. or more noncontiguous extrainguinal sites, that
persist for at least three months, in the absence of
Group II—Asymptomatic or latent infection any current ill­ness or medication that may cause
A clinically asymptomatic or “latent” period follows lymphadenopathy.
the acute infection. They show positive HIV anti­
body tests during this phase and are infectious. Group IV—AIDS Related Complex
This group includes patients with considerable
Table 68.6:  Immunological abnormalities in HIV immunodeficiency, suffer­ing from various
infection constitutional symptoms or minor opportunistic
I. Features that characterize AIDS
infections. The typical constitutional symptoms
1. Lymphopenia. are fatigue, unexplained fever, persistent diarrhea
2. Selective T cell deficiency—­Reduction in number and marked weight loss of more than 10% of body
of T4 (CD4) cells, inversion of T4:T8 ratio. weight. The common opportunistic in­fections
3. Decreased delayed hypersensitivity on skin are oral candidiasis, herpes zoster, hairy cell
testing.
4. Hypergammaglobulinemia—­predominantly IgG
leukoplakia, salmonellosis or tuberculosis.
and IgA; and IgM also in children. General­ized lymphadenopathy and splenomegaly
5. Polyclonal activation of B cells and increased are usually present. ARC patients are usually
spontaneous secretion of Ig. severely ill and many of them progress to AIDS
II. Other consistently observed features: in a few months. With no treatment, the interval
1. Decreased in vitro lymphocyte proliferative
response to mitogens and antigens.
between primary infec­tion with HIV and the first
2. Decreased cytotoxic responses by T cells and NK appearance of clinical dis­ease is usually long in
cells. adults, averaging about 8–10 years. Death occurs
3. Decreased antibody response to new antigens. about 2 years later.
4. Altered monocyte/macrophage function. The acquired immunodeficiency syndrome
5. Elevated levels of immune complexes in serum.
(AIDS) presents in many ways, all due to the
Chapter 68: Retroviruses: Human Immunodeficiency Virus  | 481
underlying severe loss of the ability to respond is probable that transmission can also take place
to infectious agents and to control tumors. The during delivery or from breast milk.
features classified as group IV include what was
known as the AIDS-related complex or ARC. Laboratory Diagnosis
Laboratory procedures for the diagnosis of HIV
AIDS: This is the end-stage disease representing
infec­tion include specific tests for HIV and tests for
the irreversible breakdown of immune defence
immunodeficiency as well as. Evidence of infection
mechanisms, leaving the patient prey to progressive
by HIV can be detected in three ways:
oppor­t unistic infections and malignancies.
1. Specific tests for HIV infection.
AIDS may be manifested in several different
2. Nonspecific tests.
ways, including lymphadenopathy and fever,
3. Tests for opportunistic infections and tumor.
opportunistic in­fections, malignancies, and AIDS-
related dementia (Table 68.8).
A. Specific Tests for HIV Infection (Table 68.9)
Pediatric AIDS: About one-third to half the 1. Antigen Detection
number of babies born to infected mothers are
The virus antigens may be detectable in blood
infected with HIV. In most cases, infection is
after about two weeks following a single massive
transmitted to the baby in the perinatal period. It
in­fection. The major core antigen p24 is the earliest
virus marker to appear in blood and is the one
Table 68.8:   Opportunistic infections and malign­ tested for IgM antibod­ies appear in about 4–6
ancies commonly associated with HIV infection weeks, to be followed by IgG antibodies (Fig. 68.2).
After­wards, free p24 antigen disappears from
I. BACTERIAL circulation and remains absent during the long
1. M. avium complex
asymptomatic phase, to reappear only when severe
2.  Mycobacterium tuberculosis—disseminated or
extrapulmonary clinical disease sets in. The p24 antigen capture
3. Salmonella—recurrent septicemia assay (ELISA) which uses anti-p24 antibody as the
II. VIRAL solid phase can be used for this antigen.
1. Cytomegalovirus
2. Herpes simplex virus
3. Varicella-zoster virus 2. Virus Isolation
4. Epstein-Barr virus The virus is present in circulation and body fluids,
5. Human herpesvirus 6 within lymphocytes or cell-free. It can be isolated
6. Human herpesvirus 8
III. FUNGAL
from CD4 lymphocytes of peripheral blood, bone
1. Candidiasis marrow and serum. The technique of isolation
2. Cryptococcosis is by cocultivation of the patient’s lymphocytes
3. Aspergillosis with uninfected lymphocytes in the presence of
4. Pneumocystis carinii pneumonia interleukin-2. Virus presence is detected by assays
5. Histoplasmosis
6. Coccidioidomycosis
for reverse transcriptase and p24 antigen in the
IV. PARASITIC culture fluids.
1. Toxoplasomosis Virus titers parallel p24 titers, being high soon
2. Cryptosporidiosis after infection, low and antibody ­bound during
3. lsosporiasis the asymptomatic period, and again high towards
4. Microsporidiosis
5. Generalized strongyloidiasis
the end.
V. MALIGNANCIES
1. Kaposi’s sarcoma 3. Detection of Viral Nucleic Acid
2. B cell lymphoma or non-Hodgkin’s lymphoma
VI. SLIM DISEASE
As the most sensitive and specific test, PCR has
become the gold standard for diagnosis in all stages

Table 68.9:  Evolution of serological markers during HIV infection


Markers
State of infection p24 Ag (free) Anti-HIV IgG Anti-HIV IgM Western blot pattern
Early infection − − − −
Acute (seroconversion) illness +→− −→+ + Partial: p24 and or gp120
Carrier asymptomatic − + − Full pattern
PGL + + − Loss of p24/p55
AIDS + Absence of p24: loss of other
reactivities

www.ebook3000.com
482  |  Section 4: Virology
HIV-1 ELISA tests were the earliest approved
serologic tests for HIV infection and remain the
most sensitive approved commercial assays for
infection. Direct solid phase antiglobulin ELISA
is the method most commonly used. The antigen
is obtained from HIV grown in continuous T
lymphocyte cell line or by recombinant techniques
and should represent all groups and subtypes
of HIV-1 and HIV-2. The antigen is coated on
Fig. 68.2: Sequence of appearance of p24 antigen and
antibodies after a massive HIV infection (as by blood
microtiter wells or other suitable solid surface.
transfusion) The test serum is added, and if the antibody is
↑ exposure: A = p24 antigen; B = IgM antibody; C = IgG present, it binds to the antigen. After washing away
antibody; 2, 4, 8, 12 = week after exposure. the unbound serum, antihuman immunoglobulin
↓ n ↓ = virus readily isolated from blood. linked to a suitable enzyme is added, followed by a
color-forming substrate. If the test serum contains
of HIV infection. Two forms of PCR have been used, anti-HIV antibody, a photometrically detectable
DNA PCR and RNA PCR. color is formed which can be read by special ELISA
i. DNA PCR: In the DNA PCR, peripheral readers. Conversion of substrate to product is
lymphocytes from the subject are lysed and quantitated by spectrophotometry.
the proviral DNA amplified using primer Modifications of ELISA in which the antibody
pairs from relatively constant regions of HIV in test serum either competes with enzyme
genome. The test is highly sensitive and conjugated anti-HIV antibody, or is captured by
specific when done with proper controls. antihuman immunoglobulin onto solid phase are
ii. RNA PCR: A related test, HIV RNA PCR can more specific. Third­generation assays employ
be used for diagnosis as well as for monitoring a sandwich technique using enzyme-cou­p led
the level of viremia. HIV antigens and take advantage of the bi- or
multivalent nature of antibodies to improve
4. Antibody Detection specificity.
Demonstration of antibodies is the simplest and ELISA specific for IgM antibody is also available.
most widely employed technique for the diagnosis Immunometric assays are highly sensitive and
of HIV infection. The mean time to seroconversion specific.
after HIV infection is 3–4 weeks. Most individuals b. Rapid tests: These tests take less than 30
will have detectable anti­bodies within 6–12 weeks minutes and do not require expensive equipment.
after infection, whereas virtually all will be positive A number of ‘rapid tests’ have been introduced for
within 6 months. this purpose:
Serological tests for anti-HIV antibodies are of
∙∙ Dot blot assays
two type-screening and confirmatory tests (Table
∙∙ Particle agglutination (gelatin, RBC, latex,
68.10).
microbeads)
∙∙ HIV spot and comb tests
A. Screening Tests (Table 68.10)
∙∙ Fluorometric microparticle technologies
a. Enzyme-linked immunosorbent assays (ELISA) Tests using finger-prick blood, saliva and urine
tests have also been developed.
Table 68.10  Laboratory tests for detection of c. Simple tests
specific antibodies in HIV infection They take 1–2 hours. They also do not require
expensive equipment.
1. Screening (E/R/S) tests
a. ELISA B. Confirmatory Tests
b. Rapid tests
– Dot blot assays a. Western blot test: In this test, HIV proteins
– Particle agglutination (gelatin, RBC, latex, separated by polyacrylamide gel electrophoresis
microbeads) are blotted onto strips of nitrocellulose paper. The
– HIV spot and Comb tests
antigen impregnated nitrocellulose is then cut into
– Fluorometric microparticle technologies
c. Simple tests strips. Each strip is then incubated with a dilution
These are also based on ELISA principle but take 1–2 of patient serum. Antibodies which attach to the
hours separated viral antigens on the strip are detected
2. Supplemental tests by antihuman immunoglobulin antibody to which
a. Western blot assay
enzyme has been attached followed by application
b. Immunofluorescence test
of a substrate. The substrate changes color in
Chapter 68: Retroviruses: Human Immunodeficiency Virus  | 483
the presence of enzyme and permanently stains detection of true reactivity (window period)
the strip. The location or position on the strip at or false reactivity with principally single-band
which a patient’s antibodies attach to viral antigens reactivity. In such cases the Western blot may
indicates whether antibody is specific for viral be repeated a later. If no definitive result can
antigens or directed against nonviral material from be given even then, it may be necessary to
the cells in which the virus was grown. have p24 assay done.
b. Immunofluorescence test: In this test, HIV
Interpretation of Western Blot Test Results
infected cells are acetone fixed on to glass slides and
Western blot results are scored as negative, positive, then reacted with test serum follo­wed by fluorescein
or indeterminate (Fig. 68.3). conjugated antihuman gammaglobulin. A positive
i. Negative result: The WHO has suggested that reaction appears as apple-green fluorescence of
a weakly reactive p17 band may be consid­ered cell membrane under fluorescence microscope.
negative. Antibodies to viral core protein p24
or envelope glycoproteins gp41, gp120, or Strategies of HIV Testing
gp160 are most commonly detected.
There are three strategies of HIV testing:
ii. Positive result: In a positive serum, bands will
be seen with multiple proteins, typically with Strategy I: The serum is tested with one E/R/S
p24 (gag gene, core protein), p31 (pol gene, test and if reactive, sample is considered positive
reverse transcriptase) and gp41, gp120 or and if non-reactive it is considered negative. This
gp160 (env gene, surface antigens). A positive strategy is used for transfusion safety. For this pur­
reaction with proteins representing the three pose, a highly sensitive and very reliable test kit
genes gag, pol, env is conclusive evidence of must be used.
HIV infection. The test may be considered
positive even if it shows bands against at Strategy II: The serum reactive with one E/R/S test
least two of the following gene products: p24, is retested with a second E/R/S test with higher
gp41, gp120/160. However, interpretation specificity, based on a different antigen prepara­tion
becomes difficult when bands that appear do and/or different test principle. If found reac­tive
not satisfy these criteria. This may happen in on second E/R/S test also, it is reported as posi­
early infection but may also be nonspecific. tive, otherwise as negative. This strategy is used
for HIV surveillance. The majority of individuals
iii. Indeterminate result : Indeterminate
seroconvert within 2 months after viral exposure.
results are not uncommon. Indeterminate
HIV infection for longer than 6 months without a
results may arise from either insensitive
detectable antibody response is very uncommon
Strategy III: The serum reactive with two E/R/S
tests is retested with a third E/R/S test. The third
test should again be based on different anti­gen
preparation or test principle. A serum testing
reactive with all three E/R/S tests is reported
posi­tive. A serum sample nonreactive in third
E/R/S is considered equivocal/borderline. Such
individuals should be retested after three weeks. If
this sample also provides an equivocal result, the
person is consi­dered to be HIV antibody negative.
For asympto­matic HIV infection, it is necessary to
confirm the diagnosis with three tests. Symptomatic
infections with opportunistic infections, however,
may be sub­jected to two tests.
The first test selected for any of the strategies
should be of highest sensitivity and second and
third E/R/S test selected should be of highest
specificity.

B. Nonspecific Tests
Immunological Tests
Fig. 68.3: Diagrammatic representation of Western blot The following parameters help to establish the
test for HIV immunodeficiency in HIV infec­tion.

www.ebook3000.com
484  |  Section 4: Virology
a. Total leukocyte and lymphocyte count to tests are negative during the early stage of HIV
demon­strate leukopenia and a lymphocyte infection when the individual is infectious,
count usually below 2000/mm3. screening may not detect all dangerous donors
b. T cell subset assays. Absolute CD4+ T cell count but can still eliminate a large majority of them.
will be usually less than 200/mm3. T4:T8 cell Screening for p24 antigen can detect those in
ratio is reversed. the window period also.
c. Platelet count will show thrombocytopenia. A person found positive for HIV antigen or
d. Raised IgG and IgA levels. antibody should never donate blood or other
e. Diminished CMI as indicated by skin tests. biological materials. As the infection can be
f. Lymph node biopsy showing profound transmitted from mother to baby before, during
abnormali­ties. or after birth, antenatal screening is useful.
2. Seroepidemiology: Antibody surveys have been
C. Tests for Opportunistic Infections and most useful in identifying the geographical extent
Tumors of HIV infection and in other epidemiological
Apart from diagnosing HIV infection, the laboratory studies.
would be called upon to identify the opportunistic 3. Diagnosis: Serology is almost always positive
infections that are a feature of AIDS. Routine in persons with clinical features of AIDS. It
microbiological methods would suffice for this. may, however, be negative in acute illness
However, serological diagnosis of infections may and sometimes in the very late cases where
not always be reliable in AIDS as antibody formation the immune system is nonreactive. Routine
may be affected by the immune deficiency. serology may also be negative when the
infection is with a different AIDS virus. For
Applications of Serological Tests (Table example, HIV-2 infections are likely to be
68.11) missed if antibody testing is done with the HIV-l
antigen alone.
Serological tests for HIV infection are employed in
4. Prognosis: In a person infected with HIV,
the following situations:
loss of detectable anti-p24 antibody indicates
1. Screening: Screening is defined as the systematic
clinical deterioration. This is also associated
application of HIV testing, whether voluntary or
with HIV antigenemia and increased virus titer
mandatory, to entire populations or selected
in circulation.
target groups. It should be mandatory that all
donors of blood, blood products, semen, cells,
tissues and organs be screened. As antibody Laboratory Monitoring of HIV Infection
Some laboratory tests are important in monitoring
Table 68.11:  Potential antiviral therapies the course of HIV infection.
for HIV infection 1. CD4+ T cell count: When the count falls below
500, it is an indication of disease progression
Nucleoside analog reverse transcriptase inhibitors
Azidothymidine (AZT) (Zidovudine) and the need for antiretroviral therapy. Counts
Dideoxycytidine (ddC) (Zalcitabine) below 200 denote risk of serious infections.
Dideoxyinosine (ddI) (Didanosine) 2. Direct measurement of HIV RNA: Direct
d4T (Stavudine) measurement of HIV RNA becomes necessary,
3TC (Lamivudine)
ABC (Abacavir) particularly in the course of treatment. This
is done usually by two methods, the reverse
Non-nucleoside reverse transcriptase inhibitors
Nevirapine (Viranune)
transcriptase PCR (RT-PCR) assay and the
Delavirdine (Rescriptor) branched DNA (bDNA) assay.
Efavirenz (Sustiva) 3. Beta-2-microglobulin and Neopterin: Beta-2-
Protease inhibitors microglobulin and Neopterin can be measured
Saquinavir (Invirase/Fortovase) in serum or urine. Their concentrations are
Ritonavir (Norvir) low in asymptomatic infection and rise with
Indinavir (Crixivan)
advancing disease.
Nelfinavir (Viracept)
Amprenavir (Agenerase)
Highly active antiretroviral therapy (HAART) Epidemiology
(combination) Routes of Transmission
Indinavir/AZT/3TC
Ritonavir/AZT/3TC HIV is spread only by three modes—sexual
Nelfinavir/AZT/3TC contact with infected persons; by blood and blood
Nevirapine/AZT/ddI products; and from infected mother to babies
Nevirapine/indinavir/3TC (intrapartum, perinatal, postnatal).
Chapter 68: Retroviruses: Human Immunodeficiency Virus  | 485
Geographic distribution: HIV -1 infections are non-nucleoside reverse transcrip­tilse inhibitors, or
spreading worldwide, with the largest number protease inhibitors (Table 68.11). Other anti-HIV
of AIDS cases in sub-Saharan Africa but with drugs being developed
a growing number of cases in Asia, the United In the current guidelines, AZT is recommended
States, and the rest of the world. HIV-2 is more for the treatment of asymptomatic or mildly
prevalent in Africa (especially West Africa) than symptomatic people with CD4 counts of less than
in the United States. Heterosexual trans­mission is 500/µL and for the treatment of infected pregnant
the major means of spread of HIV-1 and HIV-2 in women to reduce the likelihood of transmission
Africa, HIV-2 produces a disease similar to but less of the virus to the fetus. Unfortunately, the high
severe than AIDS. mutation rate of HIV promotes the development
AIDS in the developing countries differs from of resistance to these drugs.
the disease in the western countries clinically too. A cocktail of several antiviral drugs (e.g. AZT,
In Africa, the major manifestation is pronounced 3TC, protease inhibitor) termed highly active
wasting so that it has been called the ‘slim disease’. antiretroviral treatment (HAART), each with
The high prevalence of tuberculosis and parasitic different mechanisms of action, has less potential to
infections complicate the clinical picture. breed resistance and has become a recommended
HIV infection in India: HIV infection was detected therapy. Multidrug therapy can reduce blood levels
rather late in India, the first cases having been of virus to nearly zero and reduce morbidity and
found in female sex workers in Madras (Chennai) mortality in many patients with advanced AIDS.
in 1986 and the first AIDS patient the same year Although HAART is a difficult drug regimen, many
from Bombay (Mumbai). Since then in every high patients return to nearly normal on this therapy.
risk group, the rate of infection has been mounting. Apart from specific antiretroviral therapy,
By the end of 2003, India is believed to have about other measures in the treatment of AIDS include
5 million HIV-infected people, the second largest (i) treatment and prophylaxis of opportunistic
such population after South Africa. infections and tumors (ii) general management
and (iii) immunorestorative measures.
Control
i. Sexual transmission: The best method of Postexposure Prophylaxis (Pep)
checking sexual transmission of infection is If an accidental exposure occurs, any wound
health education The use of condoms and should be washed with soap and water, or mucous
vaginal antiseptics could have an impact. membranes flushed with water. The accident must
ii. Exposure to blood: Drug injectors can avoid be reported so that, if necessary, prophylaxis can
risk by not injecting, or can reduce risk by be started as soon as possible. Knowledge of the
using only clean equipment. status of the source patient is essential.
iii. Mother to child transmission: This can be Exposure to blood, body fluid, other potentially
reduced by identifying infected mothers and infected material or an instrument contaminated
giving specific therapy in the later stages of with one of these materials may lead to risk of
pregnancy and to the baby after birth. All acquiring HIV infection. The risk of infection varies
women who have been potentially exposed with the type of exposure and other factors. Most
should seek HIV antibody testing before exposures do not result in infection. Health workers
becoming pregnant and, if the test is pos­itive, are normally at very low risk of acquiring infection
should consider avoiding pregnancy. HIV during management of infected patients. Following
­infected mothers should avoid breastfeeding exposure, postexposure prophylaxis (PEP) may be
to reduce transmission of the virus to their required depending upon the category of exposure
children. and HIV status of exposure source (Table 68.12).
Prophylaxis Basic PEP regimen consists of two drug
combination while expanded PEP regimen is a
The prevention of AIDS rests at present on general
combination of three drugs. Zidovudine 300 mg
measures, such as health education, identification
BD and Lamivudine 150 mg BD are used in basic
of sources and elimination of high risk activities. No
two drug regimen. In expanded three drug PEP
specific vaccine is available.
regimen, a protease inhibitor is added to this
Vaccine development: No vaccine against HIV is combination of drugs. Among protease inhibitors,
available despite several trials. lopinavir 400 mg BD or 800 mg OD or ritonavir 100
mg BD or 200 mg OD are preferred as third drug.
Treatment To be effective these drugs must be started within
The anti-HIV drugs approved can be classified as the first 72 hours and ideally within 2 hours. The
nucleoside analog reverse transcrip­tase inhibitors, PEP should be continued for a period of four weeks.

www.ebook3000.com
486  |  Section 4: Virology
Table 68.12:  PEP regimen according to exposure and status of source
Category of exposure HIV positive and HIV positive HIV staus not known
asymptomatic and clinically
symptomatic
i. Mild exposure (Mucous Consider two drug PEP regimen Start two drug PEP Usually no PEP or
membrane/non-intact skin with regimen consider two drug
small volumes). PEP regimen
ii. Moderate exposure (Mucous Start two drug PEP regimen Start three drug PEP Usually no PEP or
membrane/ nonintact skin with regimen consider two drug
large volume or percutaneous PEP regimen
superficial exposure with solid
needle)
iii. Severe exposure (Percutaneous Start two drug PEP regimen Start three drug PEP Usually no PEP or
with large volume) regimen consider two drug
PEP regimen

Both risk of infection and possible side-effects of ™™ Laboratory diagnosis of HIV infection includes specific
antiretroviral drugs should be carefully considered tests for HIV as well as tests for immunodeficiency.
when deciding to start PEP. Besides PEP, injured Specific tests include antigen (P24) detection, virus
isolation, detection of viral nucleic add and antibody
site on the wound should be thoroughly washed
detection
with soap and water. Antiseptics may also be used.
™™ The p24 antigen is the earliest virus marker to appear
Therapy should be continued for 4 weeks and in the blood. HIV infected persons remain negative
the victim followed with testing for virus for the for antibodies during window period. Viral isolation,
next 6 months. Exposed persons should have post- detection of viral nucleic acid by polymerase chain
PEP HIV testing, at three months and at 6 months. If reaction (PCR) and p24 antigen detection are useful
for diagnosis in window period
the test at six months is negative, no further testing
™™ HIV can be cultured by co-cultivation of lymphocytes
is required. with poten­tially infected and uninfected mono­
Key Points nuclear cells
™™ The diagnosis of HIV infection is made by detecting
™™ Human immunodeficiency virus (HIV) causes AIDS, serum antibodies to viral proteins. There are two
belongs to retroviruses types of serological tests for anti-HIVantibodies are of
™™ HIV is a spherical enveloped virus measuring up to two types—screening tests and supplementary tests
120 nm in diameter and consists of two identical ™™ Screening tests—ELISA, rapid test, and simple test
copies of single-stranded positive sense RNA (E/R/S) are usually highly sensitive tests
genome ™™ There are three strategies (strategy I to III) for HIV
™™ The three important enzymes contained in the virion testing in India
are reverse transcriptase, protease and integrase.In ™™ Prevention and control include health education,
association with viral RNA is the reverse transcriptase screening of blood and blood products for HIV
enzyme antibodies or antigens, and infection control
™™ Important structural components of the virus include ™™ Antiretroviral treatment (ART) is the mainstay in HIV
the surface antigen gp120, the transmembrane treat­ment
antigen gp41, the matrix protein p17 and the capsid
™™ Postexposure prophylaxis (PEP) may be required
antigen p25
when there is exposure to blood, body fluid, other
™™ The genome of HIV contains the three structural potentially infected material or an instrument
genes (gag, pol and env) characteristic of all contaminated with HIV.
retroviruses, as well as other nonstructural and
regulatory genes specific for the virus (tat, rev, nef,
vif, vpr and vpu). both Important questions
™™ HIV shows two distinct antigenic types—HIV-1 and
HIV-2 1.
Describe the structure and laboratory
™™ There are three modes of transmission of HIV diagnosis of human immunodeficiency virus.
infection. Sexual contact, parenteral and perinatal 2. Describe the antigenic structure of human
™™ HIV infects principally the CD4 lymphocytes. immunodeficiency virus and draw labeled
The infection causes damage to T helper (T4)
lymphocytes. T4 cells are depleted in numbers and
diagram of HIV-1.
the T4:TB (helper: suppressor) ratio is reversed 3. Discuss the modes of transmission and
™™ HIV causes acute infection, AIDS related complex pathogenesis of human immunodeficiency
(ARC), and AIDS virus.
™™ When CD4+ cells fall below 200 per mm3, the titer 4. Write short notes on:
of virus increases markedly and there is irreversible a. Human immunodeficiency virus.
breakdown of immune defence mechanisms
b. Antigens of human immunodeficiency virus.
Chapter 68: Retroviruses: Human Immunodeficiency Virus  | 487
c. Opportunistic infect­ions associated with hu- 7. Which type of cells are most often infected by HIV?
man immunodeficiency virus infection. a. CD4+T lymphocytes
d. Strategies of human immunodeficiency virus b. CD8+T lymphocytes
testing. c. Null cells
e. Control HIV d. None of the above
f. Antiviral therapy for HIV 8. Which of the following opportunistic parasitic
infections is/are associated with HIV infection?
g. Postexposure prophylaxis
a. Toxoplasmosis
b. Cryptosporidiosis
Multiple choice questions (mcqs) c. Isosporiasis
d. All of the above
1. All of the following statements are true for the 9. Which of the following tests may be used for
viral genome in HIV except that they: diagnosis of HIV infection?
a. Are diploid a. P24 antigen detection
b. Consist of DNA-dependent DNA polymerase b. Virus nucleic acid detection
activity c. Antibody detection
Consist of three major genes gag, pol, and env;
c.  d. All of the above
character­istic of all retroviruses 10. All of the following are the examples of screening
d. Are most complex of human retroviruses tests in HIV except:
2. What is the characteristic feature of viruses a. Western blot
b. ELISA
belonging to family Retroviridae?
c. HIV spot test
a. Presence of enzyme reverse transcriptase d. Latex agglutination test
b. Presence of envelope
11. Which of the following tests is/are confirmatory
c. Presence of nucleic acid
test for HIV infection?
d. Presence of lipid
a. Virus isolation.
3. Which structural genes are present in the genome b. Detection of p24 antigen.
of human immunodeficiency virus-I and not in c. Detection of viral nucleic acid.
immunodeficiency virus-2? d. All of the above.
a. gag gene 12. In HIV testing in India, the strategy I is used for:
b. pol gene a. Diagnosis of HIV infection in asymptomatic
c. env gene persons
d. All of the above b. Diagnosis of HIV infection in symptomatic per-
4. Which is the principal envelope spike antigen of sons
HIV-1? c. HIV surveillance
a. gp120 d. Ensuring safety of blood, organ, and tissue do-
b. gp140 nations
c. gp41 13. Which of the following antiviral agents has been
d. gp38 most widely used to inhibit HIV replication?
5. Which is the transmembrane pedicle antigen of a. Azidothymidine b. Zintevir
HIV-1? c. Nevirapine d. Indinavir
a. gp120 14. All of the following statements are true for highly
b. gp140 active antiretroviral therapy (HAART) except:
c. gp41 a. Inhibition of HIV replication
d. gp38 b. Improved efficacy of the therapy
c. Prevents emergence of drug resistance
6. HIV cannot be transmitted by: d. Minimize toxicity following thearpy)
a. Sexual contact
b. Kissing Answers (MCQs)
c. Intravenous drug abuse 1. b; 2. a; 3. d; 4 a; 5. c; 6. b; 7. a; 8. d; 9; d; 10. a; 11. d; 12.
d. Blood transfusion d; 13. a; 14. c

www.ebook3000.com
69
Chapter

Slow Virus and Prion Diseases

Learning Objectives

After reading and studying this chapter, you should (CJD); Kuru; Bovine spongi­form encephalopathy
be able to: (BSE) or mad cow disease; subacute sclerosing
∙∙ Describe slow virus and prion diseases panencephalitis (SSPE); progressive multifocal
∙∙ Describe the following: Creutzfeldt-Jakob Disease leukoencephalopathy (PML).

Introduction Group B: Group B comprising prion diseases of the


central nervous system (CNS) collectively known as
The term ‘slow virus disease’ is applied to a group the subacute spongiform viral encephalopathies.
of infections in animals and human beings,
characterized by a very long incubation period Animal Diseases
and a slow but relent­less course, terminating i. Scrapie.
fatally. This usually ends months later in disability ii. Bovine spongiform encephalopathy (BSE)
or death. (mad cow disease).
The concept of ‘slow infection’ was originally iii. Chronic wasting disease (in mule, deer, and
proposed by Sigurdsson (1954), a veterinary elk).
pathologist for slowly progress­ing infections of iv. Transmissible mink encephalopathy.
sheep, such as scrapie, visna and maedi.
Human Diseases
Characteristics of slow viruses i. kuru.
ii. Creutzfeldt–Jakob disease (CJD).
1. Incubation periods: Long incubation period iii. Gerstmann–Straussler–Scheinker (GSS) dis­
ranging from months to years. ease.
2. Course of illness: Lasting for months or years.
iv. Fatal familial insomnia (FFI).
3. Predilec­tion for involvement of the central
nervous system. Group C: Some chronic degenerative diseases of
4. Absence of immune response or an immune the central ner­vous system in humans are caused
response that does not arrest the disease. by “slow” or chronic, persistent infections by
5. A genetic predisposition. classic viruses. Group C consists of two unrelated
6. Invariable fatal termination. CNS diseases of human beings:
i. Subacute sclerosing panencephalitis
Classification ii. Progressive multifocal leukoencephalopathy

Slow virus diseases may be classified into three Group A


groups: Group A, group B, group C (Table 69.1).
Visna
Group A: Group A consisting of slowly progressive
infections of sheep, caused by serologically related Visna, a demyelinating disease of sheep and has
nononcogenic retroviruses called lentiviruses an incubation period of about two years. It has an
(from Latin. lentus, meaning slow). Examples: insidious onset with pareses, progressing to total
Visna; Maedi (progressive pneumonia). pa­ralysis and death.
Chapter 69: Slow Virus and Prion Diseases  | 489
Table 69.1:  Slow virus and prion diseases
Disease group Agent Hosts Incubation period Nature of disease
Group A
Visna Retrovirus Sheep Months to years Central nervous system
demyelination
Group B
(a) Animal diseases
i. Scrapie Prion Sheep, goat, Months to years Spongiform encephalopathy
mice
ii. Bovine spongiform Prion Cattle Months to years Spongiform encephalopathy
encephalopathy (BSE)
iii. Chronic wasting disease Prion Mule, deer, elk Months to years Spongiform encephalopathy
iv. Transmissible mink Prion Mink, other Months Spongiform encephalopathy
encephalopathy animals
(b) Human diseases
i. kuru Prion Humans, Months to years Spongiform encephalopathy
chimpanzees,
monkeys
ii. Creutzfeldt–Jakob disease Prion Humans, Months to years Spongiform encephalopathy
(CJD) chimpanzees,
monkeys
iii. Gerstmann–Straussler– Prion
Scheinker (GSS) dis­ease
iv. Fatal familial insomnia (FFI) Prion
Group C
(a) Subacute sclerosing Measles virus Humans 2–20 years Chronic sclerosing
panencephalitis variant panencephalitis
(b) Progressive multifocal Polyomavirus Humans Years Central nervous system
Leukoencephalopathy JCV demyeli­nation

The virus can be grown in sheep choroid plexus ii. Transmissible mink encephalopathy: Trans­
tissue cultures, from the CSF, blood and saliva of missible mink encephalopathy is a scrapie-
affected animals. like disease of farm-raised mink. It is believed
to have spread to mink by feeding them on
Maedi (Progressive Pneumonia) scrapie infected sheep meat.
Maedi (progressive pneumonia) is a slowly pro­ iii. Bovine spongiform encephalopathy (BSE)
gressive fatal hemorrhagic pneumonia of sheep, or mad cow disease:
with an incubation period of 2–3 years. A disease similar to scrapie, designated bovine
Visna and maedi (progressive pneumonia) spongi­form encephalopathy (BSE), or “mad
viruses are closely related and are classified as cow disease,” emerged in cattle in Great
retroviruses (genus Lentivirus). Britain in 1986. This out­break was traced to the
use of cattle feed that contained contaminated
Group B (Prion Diseases) bone meal from scrapie-infected sheep and
BSE-infected cattle carcasses.
Transmissible Spongiform Encephalopathies The epidemic of “mad cow disease” peaked
(Prion Diseases) in Great Britain in 1993.
Prion diseases of animals It is now accepted that the new variant forms
i. Scrapie: Scrapie is the prototype prion disease of CJD and BSE are caused by a common
and shows marked differences in susceptibility agent, indicating that the BSE agent had
of different breeds of animal. The incubation infected humans. Through 2002, of 129 people
period is about two years. The affected animals who had been diagnosed with new variant CJD
are irritable and develop in­tense pruritus, in England, 121 had died.
scraping themselves against trees and rocks, iv. Chronic wasting disease: A scrapie-like
hence the name scrapie. Emaciation and disease, designated chronic wasting disease, is
paralysis set in, leading to death. found in mule deer and elk in the United States.
The causative agent has been maintained in Human prion diseases
brain tissue explant cultures through several 1. Kuru: Kuru was a fatal neurologic disease
serial passages. with cerebellar signs exclusive to the Fore

www.ebook3000.com
490  |  Section 4: Virology
tribes in Papua New Guinea. Kuru (means 3. Gerstmann–Straus­s ler–Scheinker (GSS)
“shivering” or “trembling,”) was first described disease: It is rare neurologic diseases of humans
by Gajdusek and Zigas in 1957 as a mysterious due to prion-type agent.
disease seen only in the Fore tribe inhab­iting 4. Fatal familial insomnia (FFI): It is also a rare
the eastern highlands of New Guinea. The dis­ neurologic diseases of humans are due to prion-
ease had an incubation period is 5–10 years type agent. In fatal familial insomnia there is loss
and led to progressive cerebellar ataxia and of ability to sleep and death within 1 to 2 years.
tremors, ending fa­tally in 3 to 6 months. A 5. Alzheimer’s disease: There are some neuro­
total of 3700 cases occurred in a population of pathologic similarities between CJD and
35,000. Alzheimer’s disease. However, the disease has
Transmission from human to human was not been transmitted experimentally to primates
associated with ritual cannibalism involving or rodents, and the amyloid material in the
the consumption of the body of a dead member brains of Alzheimer’s patients does not contain
of the family after ritual nonsterilizing cooking. Prpsc protein.
When Gajdusek began his study, he noted
that women and children in particular were
the most susceptible to the disease, and he
Group C
deduced that the reasons were that the women i. Subacute Sclerosing Panencephalitis (SSPE)
and children pre­pared the food and they were
given the less desirable viscera and brains to Subacute sclerosing panencephalitis (SSPE) is an
eat. Their risk for infection was higher because extremely serious, very late neurologic sequela
they handled the contaminated tissue, making of mea­sles. This disease occurs when a defective
it possible for the agent to be introduced measles virus persists in the brain and acts as a
through the conjunctiva or cuts in the skin, and slow virus. It has been reported that SSPE may
they ingested the neural tissue, which contains also develop as a very rare late complication of
the highest concentrations of the kuru agent. live measles virus vaccination. A similar picture
No cases have been seen in those born since has also been described as a rare complication of
1957, when cannibalism ceased. rubella infection.
Carlton Gajdusek was awarded the Nobel The SSPE is most prevalent in children who
Prize for Medicine in 1976 for his important were initially infected when younger than 2 years.
contributions on Kuru. The disease begins insidiously 5 to 15 years after a
case of measles. It is characterized by progressive
2. Creutzfeldt–Jakob disease(CJD): Creutzfeldt-
mental deterioration, involuntary movements,
Jakob disease is a rare chronic encephalopathy
muscular rigidity, and coma. Death occurs 1 to 3
of humans with associated dementia. CJD in
years after onset of symptoms.
humans develops gradually, with progressive
dementia, ataxia, and myoclonus, and leads to
death in 5–12 months. The disease was known ii. Progressive Multifocal
as both sporadic and inherited forms. leukoencephalopathy (PML)
The natural mode of transmission of Progressive multifocal leukoencephalopathy (PML)
Creutzfeldt–Jakob disease (GJD) is unknown, is a rare, degenerative CNS infection that usually
and it does not appear to be acquired from occurs in persons with severe immunodeficiency
sheep. Iatrogenic CJD has been transmitted disorders. The disease is characterized by
accidentally by contaminated growth hormone, memory loss, difficulty in speaking, and a loss of
by corneal transplant, by contaminated surgical coordination. Death occurs in 3–4 months.
instruments, and by cadaveric human dura
mater grafts. Causative agent: The causative agent is a human
A protein very similar to scrapie PrPsc is polyomavirus (JC virus), a member of the family
present in brain tissue infected with classic CJD. Polyomaviridae. PML has been seen in several
It has been spec­ulated that the agent of CJD pa­tients with AIDS-associated neurologic compli­
was derived originally from scrapie-infected cations.
sheep and transmitted to humans by ingestion
of poorly cooked sheep brains. Key Points
An epidemic of mad cow disease in the United
Kingdom and the unusual incidence of CJD ™™ The term ‘slow virus disease’ is applied to a group
of infections in animals and human beings,
in younger people (younger than 45 years) characterized by a very long incubation period and
prompted con­cern that contaminated beef was a slow but relent­less course, terminating fatally
the source of this new variant of CJD.
Chapter 69: Slow Virus and Prion Diseases  | 491
d. Subacute sclerosing panencephalitis (SSPE)
™™ Slow virus diseases are classified into: e. Progressive multifocal leukoencephalopathy
–  Group A; such as visna and medi. (PML)
–  Group B; prion diseases of the CNS (subacute
spongiform viral encephalopathies). Human
diseases include kuru, Creutzfeldt–Jakob disease Multiple choice questions (mcqs)
(CJD), Gerstmann–Straussler–Scheinker (GSS)
1. The prion-mediated disease in humans is
dis­ease and fatal familial insomnia (FFI) Scrapie,
bovine spongiform encephalopathy (BSE) (mad cow a. Progressive multifocal leukoencephalopathy
disease), chronic wasting disease, and transmissible b. Subacute sclerosing panencephalitis
mink encephalopathy occur in animals. c. Gerstmann–Straussler–Scheinker syndrome
–  Group C diseases include subacute sclerosing d. Bovine spongiform encephalopathy
panencephalitis and progressive multifocal 2. Variant Creutzfeldt–Jakob disease is transmitted by
leukoencephalopathy. a. Cuts in the skin
™™ SSPE is a very rare delayed sequel to infection with
b. Transplantation of contaminated cornea
measles virus, many years after the initial infection.
Papova virus is responsible for PML. c. Ingestion of infected tissue
d. Blood transfusion
3. All the following are slow diseases transmitted by
Important Questions conventional viruses in humans except
a. Subacute sclerosing panencephalitis
1. Write briefly on slow virus diseases. b Progressive multifocal leukoencephalopathy
2. Write short notes on: c. Acquired immunodeficiency syndrome
a. Creutzfeldt–Jakob disease d. Creutzfeldt–Jakob disease
b. Kuru
c. Bovine spongi­form encephalopathy (BSE) or Answers (MCQs):
mad cow disease 1. c; 2. d; 3. d

www.ebook3000.com
70
Chapter

Miscellaneous Viruses

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Describe structure of rotavirus
be able to: ∙∙ Discuss laboratory diagnosis of rotavirus infections
∙∙ Describe Rubella virus ∙∙ List the viruses causing diarrhea.
∙∙ Describe the following: Lassa fever; severe acute
respiratory syndrome (SARS)

RUBIVIRUS 4. Rare complications: Include thrombocy­


topenic purpura and encephalitis.
Rubella virus has been classified in the family
Togaviridae as the only member of the genus Congenital rubella: Based on the results of in
Rubivirus. It resembles togaviruses structurally and vitro studies, rubella infection is presumed to
in many other features. cause chromosomal breakages and inhibition of
mitoses in infected embryonic cells. Fetal damage
caused by maternal rubella is related to the stage
RUBELLA (GERMAN MEASLES) of pregnancy.
Rubella or German measles is a mild exanthematous Classical congenital rubella syndrome: The virus
fever characterized by transient macular rash and may spread to the fetus through the bloodstream,
lymphadenopathy. causing death due to infection in early pregnancy,
Morphology: Rubella virus is a pleomorphic, congenital malformations in infection during the
roughly spherical particle, 50–70 nm in diameter, first trimester and more subtle damage in later
with single stranded RNA genome and surrounded infections. The most common malformations
by an envelope carrying hemagglutinin peplomers. caused by rubella are cardiac defects, cataract and
deafness, which constitute the classical congenital
Properties: It aggluti­nates goose, pigeon, day-
rubella syndrome.
old chick and human erythrocytes at 4°C, a
characteristic which is utilized in the hemag­
glutination inhibition test for specific antibodies. Epidemiology
Resistance: The virus is inactivated by ether, Rubella has a worldwide distribution. Humans are
chloroform, for­maldehyde, beta propiolactone and the only host for rubella infection is transmitted by
desoxycholate. It is destroyed by heating at 56°C, the respiratory route. Currently, only 2–3% of young
but survives for sev­eral years at –60°C. adult females are susceptible.

Clinical features: Humans are the only host for


Laboratory Diagnosis
rubella infection is transmitted by the respiratory
route. Incubatian period is 2–3 weeks. The A. Virus Isolation
characteristic clinical features are: The virus may be isolated from blood during the
1. Rash: A generalized rash develops. early stage or more successfully from throat swabs
2. Lymphadenopaty of posterior cervical glands. in rabbit kidney or vero cells. Various tissue culture
3. Common complications: Are arthralgia cell lines of monkey (BSC-I, Vero) or rabbit (RK-
and arthritis, commoner in women and with 13, SIRC) origin, as well as primary African green
increasing age. monkey kidney cultures, may be used. Rubella

Chap-70.indd 492 15-03-2016 11:23:52


Chapter 70: Miscellaneous Viruses  | 493
produces a rather inconspicuous cytopathic effect surrounded by an envelope with large, club-shaped
in most of the cell lines. Therefore, rubella virus, peplomers; and measure 50-300 nm in diameter
in cell culture, is detected by interference with the (mean, 110–130 nm). Electron microscopy of
CPE of a challenge virus (coxsackievirus A19) and thin sections shows characteristic electron-dense
by immunofluorescence or immunoperoxidase granules resem­bling grains of sand within virus
staining for detection of antigen. The virus particles. Hence, the name arena (L), meaning
grows better if cultures are incubated at a lower sand. These particles are cellular ribosomes picked
temperature, 33–35°C. Rubella virus isolation is up by the virus presumably during maturation by
not commonly employed for diagnosis because of budding from host cells.
the dif­ficulties and delay involved. Arenaviruses establish chronic infections in
rodents. Each virus is generally associated with a
B. Serology single rodent species. Humans are infected when
ELISA for IgM and IgG antibodies gives valuable they come in contact with rodent excreta. Some
information. IgM antibody alone, without IgG viruses cause severe hemor­rhagic fever.
means current acute infection. In case of rubella
IgG antibody, four fold or more rise in titer in paired Arenaviruses Causing Human Diseases
sera has a diagnostic value. IgG antibody alone, At least seven arenaviruses cause human disease-­
without IgM means past infection or vaccination lymphocytic choriomeningitis (LCM), Lassa, Junin,
and denotes immunity, as there is only one Machupo, Guanarito, Sabia, Whitewater Arroyo.
serotype of rubella virus.

Prophylaxis 1. Lymphocytic Choriomeningitis Virus (LCM)


Passive prophylaxis: There is little evidence that Lymphocytic choriomeningitis virus (LCM) virus
administ­ering normal human immunoglobulin after has a worldwide distribution, It is endemic in
contact reduces the risk of mater­nal rubella and fetal mice and other animals (dogs, monkeys, guinea
infection. pigs, hamsters) and is occasionally transmitted to
humans. It may chronically infect mouse or ham­
Active Prophylaxis ster colonies.
Rubella vaccines: Live attenuated vaccine in use Human infections: Most human infections are
now is the RA 27/3 strain grown in human diploid cell acquired by contact with laboratory mice or
culture and administeted by subcutaneous injection hamsters, presumably via mouse droppings.
in a single dose of 0.5 mL. The vaccine is available Lymphocytic choriomeningitis in humans is an
as a single antigen or combined with measles and acute disease manifested by aseptic meningitis
mumps components as MMR vaccine. Vaccine- or a mild sys­temic influenza-like illness. Rarely
induced immunity persists in most vaccinees for at there is a severe encephalomyelitis or a fatal
least 14–16 years and probably is lifelong. systemic disease. Many infections are subclinical.
LCM has been reported to account for 5–10% of
The vaccine virus is apparently not teratogenic.
sporadic viral meningitis in human beings. The
Inadvertent administration of the vaccine to
incubation period is usu­ally 1–2 weeks.
a pregnant women may not therefore lead to
congenital defects in the baby.
2. Lassa Fever
VIRAL HEMORRHAGIC FEVERS The first recognized cases of Lassa fever occurred
in 1969 among Americans stationed in the Nigerian
Hemorrhagic manifestations are sometimes seen
village of Lassa.
in many viral fevers, such as exanthematous fevers
Natural reservoir is the multimammate rat,
-­smallpox, chickenpox and measles; mosquito borne
Mastomys natalensis. Rodent excreta probably act
diseases—yellow fever, dengue and chikungun­ya;
as the source of infection. The incubation period is
tick-borne fevers—Kyasanur forest dis­e ase,
3–16 days. The virus is present in the throat, urine
Omsk hemorrhagic fever and the Crimean­Congo
and blood of patients. Nosocomial infection has
hemorrhagic fever. However, the term hemorrhagic
occurred frequently.
viral fevers is not applied to them, but only to a group
The antiviral drug ribavirin is the drug of choice
of diseases, apparently zoonotic in nature, with typical
for Lassa fever and is most effective if given early
hemorrhage features caused by viruses belonging to
in the dis­ease process. A potential vaccine is under
two families: Arenavirus and Filovirus.
study.

ARENAVIRUSES 3. South American Hemorrhagic Fevers


Arenaviruses are typified by pleomorphic particles The South American arenaviruses are all con­sidered
that contain a segmented RNA genome; are to be members of the Tacaribe group of arenaviruses.

www.ebook3000.com
Chap-70.indd 493 15-03-2016 11:23:52
494  |  Section 4: Virology
Rodents act as reservoirs and transmission is in Marburg, Frankfurt (Germany) and Belgrade
believed to occur through rodent excreta. Serious (Yugoslavia) in 1967.In each case the infection
human pathogens are the closely related Junin, arose from tissues of African green monkeys to
Machupo, Guanariro, and Sabia viruses. which the laboratory workers had been exposed
a. Junin hemorrhagic fever (Argentine hemo­ imported to laboratories from Uganda via London.
rrhagic fever).
b. Machupo hemorrhagic fever (Bolivian hemo­ Ebola Virus
rrhagic fever). In 1976, two severe epidemics of hemorrhagic fever
c. Guanarito virus (the agent of Venezuelan occurred in Sudan and Zaire with high fatality. The
hemor­rhagic fever). causative virus was morphologically identical to
the Marburg virus but antigenically distinct. The
Laboratory Diagnosis of Arenaviruses virus responsible was named Ebola virus after the
1. Virus Isolation: Blood, CSF, throat washings, Ebola river. In 1979 Ebola re-emerged in Sudan,
pleural fluid and urine are the specimens used with serial person­t o-person spread. Another
for diagnosis. Arenaviruses can be isolated from epidemic occurred in Kikwit, Zaire, in 1995.
specimens by inoculation of suckling mice, Three distinct strains of Ebola virus have been
hamsters and guinea pigs. Virus can also be recognized.
isolated by inoculation of Vero E6 and BHK-21 1. Zaire strain (EBO- Z)
cells. 2. Sudan strain (EBO-S)
2. Antigen Detection: Viral antigen can be 3. Reston strain (EBO-R)
detected by Indirect immunofluorescence is
used for detection of viral antigen in the blood. Pathogenesis
3. Serology: For detection of IgM antibody and/or
It is probable that Marburg and Ebola viruses have
a rising titre of antibody in paired sera indirect
a reservoir host, perhaps a rodent or a bat, and
immunofluorescence test may be used.
become transmitted to humans only accidentally.
FILOVIRUSES Trans­mission among humans generally requires
direct contact with blood or body fluids, although
They have been classified as filoviridae.These are droplet infections can occur.
long threadlike viruses, hence the name (filum
means thread). They range in size from 80 to Laboratory Diagnosis
800–1000 nm. Marburg and Ebolaviruses causing
1. Electron microscopy: In the blood and in the
hemorrhagic fever belong to the genus Filovirus.
cytoplasm of the affected cells filamentous
Morphology: Marburg and Ebola viruses are viruses can be seen by electron microscopy.
enveloped nega­tive sense single stranded RNA 2. Isolation of virus: Virus can be cultured in
viruses with long tu­bular or filamentous forms vero cells from the blood during the febrile
(Fig. 70.1). phase. Virus isolate is identified by electron
microscopy and direct immunofluorescence.
Marburg Virus Virus culture must only be attempted in
Marburg disease is a hemorrhagic fever that laboratories with required biosafety level.
occurred simultaneously in laboratory workers 3. Serology: Indirect immunofluorescence can be
used for detection of antibodies in the serum.

CORONAVIRUSES
A group of spherical or pleomorphic enveloped RNA
viruses, carrying petal or club shaped peplomers on
their surface has been classified as coronaviruses
(Corona, meaning crown) resembling the solar
corona (Fig. 70.2). It has been classified in the
family coronaviridae with two genera: Torovirus
and Coronavirus.
Torovirus: The toroviruses are wide­spread in ungul­
ates and appear to be associated with diarrheas.
Coronavirus: There seem to be two serogroups of
human coro­naviruses. Coronaviruses of domestic
animals and rodents are included in these two
Fig. 70.1: Ebola virus (long thin filamentous form) groups. There is a third distinct antigenic group

Chap-70.indd 494 15-03-2016 11:23:53


Chapter 70: Miscellaneous Viruses  | 495
when a physician from Guangdong visited Hong
Kong, fell ill and died there, after infecting twelve
persons who had stayed in the same hotel. They in
turn, went to their separate countries to fall ill and
initiate outbreaks there.
In February in Hanoi, Vietnam, a private hospital
sought the help of a WHO local office about an
unusual case of pneumonia. Dr Carlo Urbani,
the nearest WHO infectious disease specialist
volunteered. Sensing the danger, he immediately
arranged for isolation and quarantine, but by then
outbreaks had begun in many countries: Canada,
USA, Ireland and some European countries, Hong
Fig. 70.2: Coronavirus Kong, China, Taiwan and most countries in South
East Asia. The new disease was named severe acute
which contains the avian infectious bronchitis respiratory syndrome (SARS). It had affected
virus of chickens. over 30 countries, with many thousand cases and
The novel coronavirus recovered in 2003 from over 800 deaths by July, when the pandemic was
patients with severe acute respiratory syndrome controlled. India escaped the SARS epidemic;
(SARS) appears to represent a new (fourth) group though a few suspect cases were detected and
of viruses. quarantined.
Human coronavuiruses: Coronavirus infections A coronavirus was found in the respiratory
in humans usually remain limited to the upper secretions of patients, identified by electron
respiratory tract. The human coronaviruses cause microscopy, and confirmed by growth in Vero cell
common colds and have been implicated in culture, animal inoculation, cloning, sequencing
gastroenteritis in infants. Inoculation in human and histology. This appears to be a new virus
volunteers induces common cold after an incubation distinct from other coronaviruses, which had been
period of 2–5 days. The resulting immunity is poor classified into three types: Mammalian viruses in
and reinfections can occur even with the same types 1 and 2 and avian viruses in type 3. The new
serotype. They appear to be the second most SARS virus becomes coronavirus type 4. It may be
common cause of common cold, par­ticularly in a recombinant of some animal and human virus.
winter, next only to rhinoviruses. Coronaviruses
are suspected of causing some gas­troenteritis in Causative agent: It has been suggested that the
humans, but the agents have not been iso­lated. causative agent isolated be designated ‘Urbani
SARS associated coronavirus’ in memory of Dr
Carlo Urbani who identified the new epidemic,
Laboratory Diagnosis
initiated early steps for its control and died of it
i. Isolation of virus: Nasopharyngeal washings within a month.
are used for the isolation of virus. Human
coronaviruses can be cultured in human fetal Mode of infection: SARS spreads by inhalation
tracheal organ culture. Some strains may grow of the virus present in droplets or aerosols of
on monolayers of diploid human embryonic respiratory secretions of patients. Fecal aerosols
fibroblasts, with minimal cytopathic effects. also may be infectious.
ii. Demonstration of antigen: ELISA test is Clinical features: The incubation period is under
used for detection of coronavirus antigens in 10 days. The disease starts as fever with cough or
respiratory secretions. other respiratory symptoms. The outbreak of SARS
iii. Antibody detection: Antibodies in serum may was char­acterized by serious respiratory illness,
be detected by ELISA. including pneu­monia and progressive respiratory
Animal coronaviruses: Coronaviruses are esta­ failure. Diarrhea is sometimes seen. The chest
blished agents of diarrhea in calves, piglets and radiograph shows pneumonic changes. Death is
dogs. due to respiratory failure.

Severe Acute Respiratory Syndrome Laboratory Diagnosis


In November 2002, Guangdong province in South Specimen: Nasopharyngeal swab or aspirate,
China experienced an outbreak of an unusual throat swab or stool specimens may be collected
respiratory infection, with many deaths. The for laboratory diagnosis. Serum is used for antibody
world outside knew about it only in February 2003, detection.

www.ebook3000.com
Chap-70.indd 495 15-03-2016 11:23:54
496  |  Section 4: Virology
1. Reverse transcription PCR—Reverse trans­ without the RNA core are also seen. The genome
cription PCR (RT-PCR has been used for early of rotaviruses is located inside the inner core and
diagnosis). consists of 11 segments of double-stranded RNA.
2. Virus culture: Virus in clinical specimens can All segments, except one, code for only one virus-
be cultured on Vero cell lines. specific protein (VP).
3. Serology: Demonstration of rise in titer of
antibodies by “ELISA or indirect immuno­ Classification
fluorescent test in paired serum samples are Rota­viruses share a common group antigen situated
useful later. However, for early diagnosis PCR is in the inner capsid layer. So far, rotaviruses have
preferred. been classified into at least seven antigenic groups
(A to G) based on antigenic epitopes on the internal
Treatment and Prophylaxis structural protein VP6. For groups A-E complete
lack of serological cross-reactivity has been proven.
No specific therapy or prophylaxis has been
These can be detected by immunofluores­cence,
identified. The virus is highly mutable and so
ELISA, and immune electron microscopy (IEM).
vaccine prophylaxis may not be easy. Control has
Multiple serotypes have been identified among
been achieved by strict isolation and quarantine.
human and animal rotaviruses.
However, ribavirin and steroids have been shown
to be useful in treatment of critical patients. Group A strains: Cause the majority of human
infections have been classified into subgroups
REOVIRIDAE (I and II) by ELI­SA, CF or immune adherence
agglutination, and into many serotvpes (1, 2, 3, etc.)
Reoviridae constitutes a diverse family of viruses that by neutralisation tests.
infect humans, many mammals, other vertebrates Group B strain: The Chinese ADRV is of group B.
(including birds), plants and insects. The family
Reovi­ridae derives its name from the pro­totype Group C rotaviruses: Cause occasional outbreaks
virus which was known as the Respiratory enteric in humans.
orphan virus, because it could be isolated fre­
Animal Susceptibility
quently from the respiratory and enteric tracts,
and it was considered ‘orphan’ because it was not Rotaviruses have a wide host range. Human
associated with any disease. rotavirus infection has been transferred to piglets,
calves and monkeys. Swine rotavirus infects both
Classification newborn and weanling piglets.
The family Reoviridae is divided into nine genera. Propagation in Cell Culture
Four of the genera are able to infect humans and
Rotaviruses are fastidious agents to culture. As calf
animals: Orthoreovirus, Rotavirus, Coltivirus, and
and simian viruses grow readily in cell cultures, they
Orbivirus. Four other genera infect only plants and
have been used as antigens for serological studies.
insects, and one infects fish.
Morphology: All reoviruses have a double- Epidemiology
shelled capsid, no envel­ope and measure 75–80 Rotaviruses are the most common cause of
nm in diameter. The genome consists of double diarrhea in infants and children the world over.
stranded RNA in 10–12 pieces, a feature unique Rotavirus infections usually predominate during
among animal viruses. They are nonenveloped and the winter season. In tropical areas infections occur
re­sistant to lipid solvents. evenly throughout the year.
Rotavirus diarrhea is usu­ally seen in children
Rotaviruses below the age of five years but symptomatic
Morphology: Morphologically, rotaviruses are infections are most com­mon in children between
polyhedrons of 75 nm diameter displaying char­ ages 6 months and 2 years. By the age of five years,
acteristic sharp-edged double ­shelled capsids, most children have had clinical or subclinical
which in electron micrographs look like spokes infection. Transmission appears to be by the fecal–
grouped around the hub of a wheel. The name is oral route. Nosocomial infections are frequent.
derived from rota, in Latin, meaning wheel. Both
‘complete’ and ‘incomplete’ particles are seen. Clinical Features
The complete or ‘double shelled’ virus measures Infection is by the fecal-oral route. The incubation
about 70 nm in diameter and has a smooth surface. period is 2–3 days. Vomiting and diarrhea occur
The incomplete or ‘single shelled” virus is smaller, with little or no fever. Stools are usually greenish
about 60 nm, with a rough surface and is rota yellow or pale, with no blood or mucus. The disease
virus that has lost the outer shell. ‘Empty’ particles is self-limited and recovery occurs within 5–10 days.

Chap-70.indd 496 15-03-2016 11:23:54


Chapter 70: Miscellaneous Viruses  | 497
Laboratory Diagnosis Laboratory Diagnosis
A. Demonstration of virus: At the peak of the 1. Electron microscopy: The virus can be demon­
disease as many as 1011 virus particles per strated in feces by electron microscopy.
milliliter of feces are present. Virus in stool is 2. Antigen detection: Radioimmunoassay (RIA)
demonstrated by immune electron microscopy and ELISA can detect the virus and viral antigen.
(IEM), latex agglutination tests, or ELISA. 3. Serology: Radioimmunoassay (RIA) and ELISA
B. Genotyping: Genotyping of rotavirus nucleic acid can detect antibody to norwalk agent.
from stool specimens by the polymerase chain
reaction is the most sensitive detection method. Adenoviruses
C. Virus isolation: Rotaviruses can be propagated Several outbreaks of diarrhea in children have been
in secondary or continuous cultures of monkey associated with the presence of large numbers of
kidney cells. Cell culture, however, is not used adenoviruses in feces. Serotypes 40 and 41 are
for routine diagnosis. most commonly associated with acute diarrhea
D. Serologic tests: Can be used to detect after in infants.
antibody titer rise, particularly ELISA.
Astrovirus
Treatment These star shaped (Greek, astron = a star, 28 nm
Management consists of replacement of fluids isometric particles which have been associated
and restoration of electrolyte balance either intra­ with some epidemics of diarrhea in children.
venously or orally, as feasible. They are recognized as pathogens for infants and
children, elderly institu­tionalized patients, and
Control immunocompromized persons.Similar viruses
In view of the fecal–oral route of transmission, have also been identified in lamb and calf diarrhea.
waste­water treatment and sanitation are significant Coronavirus
control measures. These are well established causes of acute diarrhea
in calves, piglets and dogs. They have been observed
Rotavirus Vaccine in human feces also but their relation to diarrhea is
An oral live attenuated rhesus-based rotavirus uncertain.
vaccine was licensed in the United States in 1998
for vaccination of infants. It was withdrawn a year Key Points
later. A number of different candidate vaccines of ™™ Rubella virus has been classified in the family
live ­attenuated rotavirus of bovine or human origin. Togaviridae as the only member of the genus Rubivirus
™™ Rubella or German measles is a mild exanthematous
fever characterized by transient macular rash and
Other Viruses Causing Diarrhea lymphadenopathy. Infection is acquired by inhalation
In addition to rotaviruses and noncultivable aden­ ™™ Diagnosis of rubella infection can be established by
oviruses, members of the family Caliciviridae are isolation of virus from blood or throat swabs in RK13
important agents of viral gastroenteritis in humans. or Vero cells or by serology (commonly done by ELISA)
™™ Prophylaxis is relevant only in women of child-
The most significant member is Norwalk virus and bearing age is best carried out by immunization
others are adenoviruses, astrovirus and coronavirus. with a live attenuated vaccine. (RA 27/3 strain grown
in human diploid cell culture) is administered by
subcutaneous injection to children 15 months of
NORWALK VIRUS age as such or in combination (mumps-measles-
Norwalk virus has been included in the family rubella vaccine)
™™ Hemorrhagic viral fevers is applied only to a
Calicivi­ridae. Caliciviruses are similar to picorna­ group of diseases, apparently zoonotic in nature,
viruses but are slightly larger (27–40 nm). with typical hemorrhage features caused by
Norwalk virus is the most important cause of viruses belonging to two families: Arenavirus and
epidemic viral gastroenteritis in adults. A 27 nm Filoviruses
virus was shown to be responsible for an epidemic Coronaviruses: There are two genera in the
of gastroenteritis affect­ing school children and Coronaviridae family: Torovirus and Coronavirus
Torovirus: Coronavirus: There seem to be two
teachers in Norwalk, Ohio, in 1972. The virus serogroups of human coro­naviruses- of domestic
induced diarrhea in human volun­teers. Serological animals and rodents. There is a third distinct
surveys have shown that infection with Norwalk antigenic group which contains the avian infectious
virus is widespread in many countries. The viruses bronchitis virus of chickens. The novel coronavirus
are most often associated with epidemic outbreaks recovered in 2003 from patients with severe acute
respiratory syndrome (SARS) appears to represent
of waterborne, food-borne, and shellfish-asso­
a new (fourth) group of viruses
ciated gastroenteritis.

www.ebook3000.com
Chap-70.indd 497 15-03-2016 11:23:54
498  |  Section 4: Virology
2. Which of the following can cause congenital
™™ Severe acute respiratory syndrome (SARS) refers
infections?
to a severe atypical pneumonia that assumed
pandemic proportions in 2003 a. Toxoplasma gondii
™™ Reoviridae: There are three genera in this family: b. Rubella virus
Reovirus, Orbivirus and Rotavirus: c. Cytomegalovirus
– Orbivirus: The only known orbivirus infection of d. All of the above
human beings is Colorado tick fever, a minor febrile
3. All of the following viruses can cause haemorrhagic
illness.
fevers except:
–  Rotaviruses are the most common cause of
diarrhea in infants and children. The human a. Junin virus
rotavirus is related to the viruses of epidemic b. Machupo virus
diarrhea of infant mice (EDIM), Nebraska c. Ebola virus
calf diarrhea and the simian virus SAIl. Swine
d. Norwalk virus
rotavirus infects both newborn and weanling
piglets. 4. Severe acute respiratory syndrome (SARS) can be
™™ Laboratory diagnosis is by demonstrating of virus by diagnosed by which of the following tests?
IEM, latex agglutination tests, or ELISA. On clarified a. Electron microscopy
fecal samples. Polymerase chain reaction is the b. Growth in Vero cell culture
most sensitive detection method. Development of
c. Cloning
a rotavirus vaccine has been difficult.
Viruses causing diarrhea
d. All of the above
™™ Viruses that are important causes of diarrhea include 5. Which of the following viral agents can cause
rotaviruses, the Norwalk virus, certain adenoviruses diarrhea?
and coronaviruses, and astroviruses. a. Norwalk agent
b. Astroviruses
c. Rotaviruses
IMPORTANT QUESTION
d. All of the above
Write short notes on:
6. All the followings are the causative agents of South
a. Rubella or German measles
American hemorrhagic fever except:
b. Viral hemorrhagic fevers
c. Lymphocytic choriomeningitis virus (LCM) a. Lassa virus
d. Ebola virus b. Junin virus
e. Coronaviruses c. C. Machupo virus
f. Severe acute respiratory syndrome (SARS) d. Ebola virus
g. Viruses causing diarrhea
h. Rotaviruses. 7. Which of the following techniques can be used for
detecting rotavirus infection?
MULTIPLE CHOICE QUESTIONS (MCQs) a. Latex agglutination test.
b. ELISA.
1. Which of the following viruses can cause cervical
c. Immune electron microscopy.
cancers?
a. Norwalk virus d. All of the above.
b. Herpes simplex virus
ANSWERS (MCQs)
c. Epstein-Barr virus
d. Human papilloma virus 1. d; 2. d; 3. a; 4. d; 5. d; 6. d; 7. d

Chap-70.indd 498 15-03-2016 11:23:54


71
Chapter

Oncogenic Viruses

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Describe oncogenes and mechanism of viral
be able to: oncogenesis
∙∙ Describe oncogenic viruses ∙∙ List of viruses associated with human cancers.
∙∙ Classify oncogenic viruses

INTRODUCTION Table 71.1:  Properties of cells transformed by


viruses
The association of viruses with malignancy dates
from the observation by Ellerman and Bang I. Altered cell morphology
(1908). Peyton Rous (1911) showed that a virus II. Altered cell metabolism
causes fowl sarcoma, a discovery for which he was III. Altered growth characteristics—Transformed cells
awarded the Nobel prize belatedly in 1966. Shope are altered in shape and lose the property of ‘contact
inhibition’ so that, instead of growing as monolayer,
isolated the rabbit fibroma virus in 1932 and the they grow piled up, one over another, forming
papilloma virus in 1933. Bittner (1936) proposed ‘microtumors’
that breast cancer in mice could be caused by a IV. Antigenic alterations—Appearance of new virus-
virus transmitted from mother to offspring through specified antigens (T-antigen ­TSTA)
breast milk. It is now acknowl­edged that at least V. Tumourogenicity—Capacity to induce tumors in
15% of all human tumors worldwide have a viral susceptible animals
cause.
are classified among the papil­loma-, polyoma-,
Oncogenic Viruses adeno-, herpes-, hepadna-, and poxvirus groups
Oncogenic viruses are viruses that produce tumors (Table 71.2).
in their natural hosts or in experimental animals, RNA tumor viruses: Most RNA tumor viruses
or induce malignant trans­formation of cells on belong to the retrovirus family. Retroviruses are
culture. re­sponsible for naturally occurring leukemia and
sarco­ma in several species of animals.
PROPERTIES OF CELLS TRANSFORMED
BY VIRUSES ONCOGENIC VIRUSES (TABLE 71.2)
Transformation from nor­mal to malignant cell is a A. Oncogenic DNA Viruses
multistep process and may be partial or complete.
Table 71.1 shows properties of cells transformed I. Papovaviruses (See Chapter 60)
by viruses. 1. Papilloma viruses: Papilloma viruses cause
benign tumors in their natu­r al hosts but
TYPES OF TUMOR VIRUSES some of them (e.g. condyloma acumina­urn in
humans, rabbit papilloma) may turn malignant.
Both DNA and RNA viruses are oncogenic.
The association between human papilloma
DNA tumor Viruses: All known tumor viruses virus (HPV) infection and cancer of cervix uteri,
either have a DNA genome or generate a DNA particular­ly HPV types 16 and 18 have been
provirus after infection of cells. DNA tumor viruses established.

www.ebook3000.com
Chap-71.indd 499 15-03-2016 11:24:18
500  |  Section 4: Virology
Table 71.2:  List of oncogenic viruses Marek’s disease can be pre­vented by vaccination
with an attenuated strain of Marek’s disease
A. DNA viruses
I. Papovavirus
virus.
1. Papillomaviruses of human beings, rabbits and 2. Lucke’s tumor of frogs: A herpesvirus is consi­
other animals dered to be the etiological agent of a renal
2. Polyomavirus adenocarcinoma in frogs.
3. Simian virus 40 3. Herpesvirus saimiri: It causes fatal lymphoma
4. BK and JC viruses or reticulum cell sarcoma when injected into
II. Poxvirus owl monkeys or rabbits.
1. Yaba virus (Yaba monkey tumor virus)
4. Epstein–Barr virus: Epstein–Barr (EB) herpes­
2. Shope fibroma virus
3. Molluscum contagiosum virus virus causes acute infectious mononucleosis
III. Adenovirus when it infects B lymphocytes of susceptible
Many human and nonhuman types humans. EB virus is etiologically linked to Burkitt’s
IV. Herpes virus lymphoma; to nasopharyngeal carcinoma
I. Marek’s disease virus (NPC); to post-transplant lymphomas; and to
2. Lucke’s frog tumour virus Hodgkin’s disease. Malaria may be a cofactor of
3. Herpes virus pan, papio, ateles and saimiri African Burkitt’s lymphoma.
4. Epstein–Barr virus 5. Herpes simplex and cancer cervix: An associ­
5. Herpes simplex Virus types 1 and 2
ation has been proposed between herpes
6. Cytomegalovirus
simplex type 2 infection and cancer of the
V. Hepatitis Band C viruses
B. RNA viruses
uterine cervix, though not proved.
I. Retroviruses Kaposi’s sarcoma-associated herpesvirus is also
1. Avian leukosis viruses known as human herpesvirus 8 (KSHV/HHV8).
2. Murine leukosis viruses It is suspected of being the cause of Kaposi’s
3. Murine mammary tumor virus sarcoma, primary effusion lymphoma, and a
4. Leukosis-sarcoma viruses of various animals particular lymphoproliferative disorder.
5. Human T-cell leukemia viruses 6. Cytomegalovirus: Cytomegalovirus infection
has been associated with carcinoma of the
prostate and Kaposi’s sarcoma.
2. Polyoma virus: The polyoma virus causes
natural latent infection in laboratory and
domestic mice. However, it induces a wide V. Hepatitis B Virus
variety of histologically diverse tumors, when Hepatocellular carcinoma (HCC) following
injected into infant mice or other rodents. chronic infection with hepatitis B virus is one of
3. Simian virus 40 (SV 40): See Chapter 60. the most prevalent forms of cancer. HBV has been
4. BK and JC virus: The papovaviruses BK and claimed to be directly or indirectly in­volved in the
JC, which cause wide­spread asymptomatic etiology of hepatocellular carcinoma.It is also the
human infection, can induce tumors in only malignant disease of humans preventable by
immunodeficient subjects (see Chapter 60). immunization against the causal agent.

II. Poxvirus B. Oncogenic RNA Viruses


Yaba virus produces benign tumors (histiocytomas) Retroviruses: Retroviruses contain an RNA genome
in its natural host, monkeys. Shope fibroma virus and an RN-A­directed DNA polymerase (reverse
produces fibromas in some rabbits and is able to transcriptase). RNA tumor viruses in this family
alter cells in culture. mainly cause tumors of the reticuloendothelial and
Molluscum contagiosum virus produces small hematopoietic systems (leukemias, lymphomas) or
benign growths in human. of connective tissue (sarco­mas).
Oncogenic retroviruses:
III. Adenovirus 1. Avian leukosis complex: A group of antigen­
Though some types (12, 19,21) of human ically related viruses which induce avian
adenovirus may produce sarcomas in newborn leukosis (lymphomatosis, myeloblastosis and
rodents after ex­perimental inoculation, they do not erythroblastosis viruses) or sarcoma in fowls
appear to have any association with human cancer. (Rous sarcoma virus, RSV).
2. Murine leukosis viruses: This group consists of
IV. Herpesvirus several strains of murine leukemia and sarcoma
1. Marek’s disease: Marek’s disease is a highly viruses.
contagious lymphoproliferative disease of 3. Mammary tumor virus of mice: This virus
chickens. occurs in certain strains of mice having a high

Chap-71.indd 500 15-03-2016 11:24:18


Chapter 71: Oncogenic Viruses  | 501
natural inci­dence of breast cancer. It used to related human virus, HTLV-2, has not been
be known as the ‘milk factor’ or ‘Bittner virus’. conclusively associated with a specific disease.
It multiplies in the mammary gland and is 6. Hepatitis C virus: Chronic infection with
transmitted from mother to offspring through hepatitis C virus is also consid­ered to be a
breast milk. causative factor in hepatocellular carcinoma.
4. Leukosis-sarcoma viruses of other animals:
A number of viruses have been isolated from VIRUSES ASSOCIATED WITH HUMAN
leukosis and sarcomas in various species of CANCER
animals—cat, ham­ster, rat, guinea pig and
monkey. Viruses associated with cancer in human beings
are shown in Table 71.3.
5. Human retroviruses: Human T-cell leukemia
(lymphotropic) viruses (HTLV)
Retroviruses named human T-cell leukemia ONCOGENES
viruses were isolated. HTLV-l has been establi­
“Oncogene” is the general term given to genes
shed as the causative agent of adult T cell
that cause cancer. Viral oncogenes (V-onc),
leukemia-lymphomas (ATL) as well as a ner­vous
commonly known as ‘can­cer genes’ are genes
system degenerative disorder called tropical
which encode proteins triggering transformation
spastic paraparesis. The virus preferentially
of normal cells into cancer cells. Normal versions
infects T4 (CD4) cells. Infect­ed T-cells express
of these transforming genes are present in normal
large quantities of interleukin-2 re­ceptors. A
cells and have been desig­nated proto-oncogenes
Table 71.3:  Viruses implicated in human cancers or c-onc. They are not of viral origin. Oncogenes
are not essential for the replication of the virus
Virus family Virus Tumor
DNA viruses and mutants lacking them occur, which replicate
normally without being oncogenic.
Papovaviridae Human Cervical, vulvar, penile
papilloma cancers Apparently viral oncogenes originated at
virus (HPV) Squamous cell some distant past from proto-oncogenes by
carcinoma recombination between retroviral and cellular
Herpesviridae Epstein–Barr Nasopharyngeal genes. Transduction of the cellular genes was
virus (EBV) carcinoma probably an accident, as the presence of the
African Burkitt’s cellular sequences is of no benefit to the viruses.
HSV type 2 lymphoma
Human B-cell lymphoma
Proto-oncogenes are widespread in vertebrates
herpesvirus 8 Cervical carcinoma and metazoa—from human beings to fruitflies.
Kaposi’s sarcoma They have been found to code for proteins involved
Hepadnaviridae Hepatitis B Hepatocellular in regu­lating cell growth and differentiation.
virus carcinoma Incorrect expression of any component might
RNA viruses: interrupt that regulation, resulting in uncontrolled
growth of cells (cancer). The presumed functions of
Flaviviridae Hepatitis C Primary liver cancer
virus many oncogenes have been identified (Table 71.4).
Retroviridae Human T-cell Adult T-cell leukemia/ Transfection: Transfection is a useful method for
lymphotropic lymphoma the study of oncogenes. Certain mouse fibroblast
virus (HTLV-1)
cell lines, such as NIH 3T3, can take up foreign
Table 71.4:  Some oncogenes* and their chromosomal locations in humans
Viral oncogene Origin Natural tumor Human gene Chromosomal location in human
beings
V-src Chicken Sarcoma C-src 20
V-ras Rat Sarcoma C-ras 11
V-myc Chicken Leukemia C-myc 8
V-fes Cat Sarcoma C-fes 15
V-sis Monkey Sarcoma C-sis 22
V-mos Mouse Sarcoma C-mos 8
*Oncogenes are given three-letter codes from the animal or tumor from which they are derived, preceded by either V- or C-, for viral or
cellular genes, respectively
src, sarcoma of chicken; ras, rat sarcoma; sis, simian sarcoma

www.ebook3000.com
Chap-71.indd 501 15-03-2016 11:24:18
502  |  Section 4: Virology
DNA, incorporate them into their genome and DNA of the infected host cell, and it is from this
express them. This method of gene transfer is called integrated DNA copy that all proteins of the virus
transfection. are translated.

ANTIONCOGENES Key Points


A class of genes has been identified in normal human ™™ Oncogenic viruses are viruses that produce tumors
in their natural hosts or in experimental animals, or
cells, whose function appears to be inhibition of induce malignant trans­formation of cells on culture
malignant transformation. They have been termed ™™ DNA tumor viruses are classified among the papil­
antioncogenes, tumor-suppressor or growth- loma-, polyoma-, adeno-, herpes-, hepadna- and
suppressor genes. poxvirus groups
™™ Most RNA tumor viruses belong to the retrovirus
Examples family. Retroviruses are re­sponsible for naturally
occurring leukemia and sarco­ma in several species
i. Retinoblastoma (Rb) gene: The loss of of animals
Rb gene function is causally related to the ™™ Viral oncogenes (V-onc), commonly known as
development of retinoblastoma-a rare ocular ‘can­cer genes’ are genes which encode proteins
tumor of children-and other human tumors. triggering transformation of normal cells into cancer
cells
ii. p53 gene: The p53 gene is mutated in over half ™™ The main oncogenic DNA viruses are papillo­
of all human cancers. Specific chromosomal maviruses, Epstein-Barr virus, and hepatitis B. Most
deletions, recognized in associ­ation with of the oncogenic RNA viruses are retroviruses.
certain types of human cancers may reflect
the loss of tumor-suppressor genes. IMPORTANT QUESTIONS
MECHANISMS OF VIRAL ONCOGENESIS 1. Define and classify oncogenic viruses.
2. Enumerate RNA and DNA oncogenic viruses.
DNA genome: Some viruses that possess a DNA Describe mechanism of viral carcinogenesis.
genome, or that synthesize DNA during repli­ 3. Write short notes on:
cation (the retroviruses), are able to induce malig­ a. Viral oncogenes.
nant changes in cells, both in vivo and in vitro
b. Viruses associated with human cancer.
(transformation). The viral DNA (or a portion of
it) is integrated with the host cell genome in the
case of oncogenic DNA viruses. The viral DNA MULTIPLE CHOICE QUESTIONS (MCQs)
being incomplete or ‘defec­tive’, no infectious virus 1. Which of the following viruses can be oncogenic?
is produced. However, under its influence, the host a. Polyomavirus b. Simian virus 40
cell undergoes malignant trans­formation. A virus- c. Marek’s disease virus d. All the above
transformed cancer cell is in many ways analogous 2. All of the following viruses are associated with
to a bacterium lysogenized by a de­fective phage. human cancercepy:
a. Human papilloma viruses b. Epsten Barr virus
Retroviruses: Retroviruses carry an RNA-directed c. Hepatitis C virus d. All the above
polymerase (reverse transcriptase) that constructs
a DNA copy of the RNA genome of the virus. The ANSWERS (MCQs)
DNA copy (provirus) becomes integrated into the 1. d; 2. d

Chap-71.indd 502 15-03-2016 11:24:18


Section 5: Medical Mycology

72
Chapter
General Properties, Classification and
Laboratory Diagnosis of Fungi

Learning Objectives

After reading and studying this chapter, you should be ∙∙ Classify fungi
able to: ∙∙ Describe laboratory diagnosis of fungal infections
∙∙ Differntiate between fungi and bacteria ∙∙ Discuss diseases caused by fungi.

Introduction CLASSIFICATION OF FUNGI


Mycology is the study of fungi. Fungi are placed in the phylum Thallophyta. It
is divisible into two groups, algae and fungi. See
Differences of fungi from bacteria Flowchart 72.1.
1. They pos­sess rigid cell walls containing chitin, Algae: Algae produce their own food by means of
mannan and other polysaccharides. chlorophyll possessed by them.
2. The cytoplasmic membrane contains sterols.
Fungi: Fungi do not possess chloro­phyll and are,
3. Cytoplasmic contents include mitochondria
therefore, saprophytes or parasites.
and endoplasmic reticulum.
A. Morphological classification
4. They possess true nuclei with nuclear membrane
and paired chromosomes. B. Systematic classification.
5. They may be unicellular or multicellular.
6. They divide asexually, sexually or by both the A. Morphological Classification
processes. On the basis of morphology, there are four groups
7. Most fungi are obligate or facultative aerobes. of fungi:
8. They are chemotrophic. 1. Yeasts: Yeasts are round, oval or elongated
9. The cells show various degrees of specialization. unicellular fungi. Most reproduce by an asexual
process called budding. (Fig. 72.1). Some
General properties of fungi reproduce by fission. On culture, they form
Fungi grow in two basic forms, as yeasts and molds. smooth, creamy colonies.
Examples: Cryptococcus neoformans.
Yeast: The simplest type of fungus is the unicellular
2. Yeast-like fungi: Yeast-like fungi grow partly as
bud­ding yeast.
yeast and partly as elongated cells resembling
Hypha: Elongation of the cell produces a tubular, hyphae. The latter form a pseudomycelium.
thread like structure called hypha. Example: Candida albicans.
Hyphae may be septate or nonseptate. 3. Molds or filamentous fungi: Molds or filament­
ous fungi form true mycelia and reproduce by
Mycelium: A tangled mass of hyphae constitutes
the formation of different types of spores.
the mycelium. Fungi which form mycelia are called
moulds or filamentous fungi. Examples of molds: Dermatophytes, Aspegillus,
In a growing colony of filamentous fungus, Penicillium, Mucor and Rhizopus.
the mycelium can be divided into the vegetative 4. Dimorphic fungi: Many fungi pathogenic to
mycelium and the aerial mycelium. man have a yeast form in the host tissue and

www.ebook3000.com
504  |  Section 5: Medical Mycology
Flowchart 72.1: Classification of Fungi

4. Deute­romycetes (Fungi Imperfecti): Most


fungi of medical importance belong to this class.
Examples: Coccidioides immitis, Paracocci­
dioides brasiliensis, Candida albicans.

Reproduction and sporulation


Types of fungal spores: Fungal spores are of two
types- sexual and asexual spores.

A. Sexual spores: Sexual spore is formed by


fusion of cells and meiosis as in all forms of higher
life. Sexual spores are of four types—oospore,
ascospore, zygospore and basidiospore (Fig. 72.2).

B. Asexual spores: These spores are produced by


mitosis. These may be vegetative spores or aerial
Fig. 72.1: Basic fungal morphology spores
a. Vegetative spores (Fig. 72.3)
in vitro at 37°C on enriched media and hyphal i. Blastospores: These are formed by budding
(mycelial) form in vitro at 25°C. from parent cell, as in yeasts.
Examples: Histoplasma capsulatum, Sporothrix ii. Arthrospores: These are formed by the pro­
schenckii, Blastomyces dermatitidis, Coccidioi­ duction of cross-septa into hyphae resulting
des immitis, Paracoccidioides brasiliensis, and in rectangular thick-walled spores.
Penicillium marneffei. iii. Chlamydospores: These are thick-walled
resting spores developed by rounding up and
B. Systematic Classification thickening of hyphal segments.
On the basis of formation of sexual spores, fungi b. Aerial spores (Fig. 72.4)
have been divided into four classes (See Flowchart
i. Conidiospores: Spores borne externally on
72.1).
sides or tips of hyphae are called conidiospores
1. Zygomycetes: Examples: Rhizopus, absidia,
or simply conidia.
mucor, pilobolus.
Other three classes, Ascomycetes, Basi­diomy­ ii. Microconidia: When conidia are small and
cetes and Deuteromycetes or Fungi Imperfecti single, these are called microconidia.
possess septate hyphae. iii. Macroconidia: These are large and septate
2. Ascomycetes: Ascomycetes include both yeasts conidia and are often multicellular.
and filamentous fungi. iv. Sporangiospores: These are spores formed
3. Basidiomycetes: Examples: Mushrooms, Filo­ within the sporangium. They develop on
basidiella neoformans (anamorph, Cryptococcus the ends of hyphae called sporangiophores.
neo­formans. Examples: Mucor and Rhizopus.
Chapter 72: General Properties, Classification and Laboratory Dignosis of Fungi  | 505
of mycoses can be identified by the following
methods.

B. Direct Microscopy
I. Potassium Hydroxide (KOH) Preparation
A portion of the treated specimen should be
examined microscopically to determine whether
hyphal elements are present. Most specimens
can be exam­ined satisfactorily in wet mounts
after partial digestion of the tissue with 10–20%
potassium hydroxide. The alkali digests cells
and other tissue materials, enabling the fungus
elements to be seen clearly. Newer preparations
for the KOH test may provide easier and more
reliable results.

II. Potassium Hydroxide (KOH) with Calcofluor


Fig. 72.2: Sexual spores White
Addition of Calcofluor white and subsequent
examina­tion by fluorescence microscopy enhances
the detection of most fungi.

III. Gram-staining
It is used for the diagnosis of yeast infections of
mucous membranes.

IV. India Ink Preparations


It may be used for detecting encapsulated yeast
Cryptococcus neoformans in cerebrospinal fluid
(CSF).
Fig. 72.3: Vegetaive spores
V. Histology
Common tissue stains used for detection of fungal
elements are the periodic acid-Schiff (PAS), Grocott–
Gomori methenamine-silver (GMS), hematoxylin
and eosin (H and E), Giemsa, and the Fontana–
Masson stains, are based on the presence of chitin
and polysaccharides in their cell wall.

C. Culture
1. Culture Media: The commonest culture media
used in mycology are Sab­ouraud’s dextrose agar
(SDA, pH 5.4), SDA with antibiotics, potato
dextrose or the slightly modified potato flakes
agar (PFA), and brain heart infusion (BHI) agar
with blood and antibiotics. The antimicrobials
usually included in SDA with antibiotics are
Fig. 72.4: Aerial spores
chloramphenicol and gentamicin to inhibit
bacterial growth and cycloheximide (actidione)
Laboratory Diagnosis to inhibit saprophytic fungi. The pH of Emmons’
modification of SDA is close to neutral and is
A. Collection and Processing of more efficient medium for primary isolation
Specimens than the original for­mulation.
The sampling proce­dures vary according to the 2. Incubation: Cultures are routinely incubated
area and type of tissue involved. Causative agents in parallel at room temperature 25°C (room

www.ebook3000.com
506  |  Section 5: Medical Mycology
temperature for weeks) and at 37°C for days.
Cultures should be retained for at least 2-3
weeks before being discarded.

D. Identification of Fungi
Once an organism has grown, it is examined for
characteristic gross and microscopic structures,
so that identification can be made (Fig. 72.5).
1. Gross or macroscopic examination of
cultures: Growth characteristics useful for
identification are the rapidity of growth, colour
and morphology of the colony on the obverse
and pigmentation on the reverse.
2. Microscopic examination
i. Tease mount: For microscopic examination
slide mounts should be made in lactophenol Fig. 72.5: Asexual forms and asexual spores of fungi
cotton blue (LCB) from fungal colony (in
teased mounts or slide cultures) to study
the morphology of hyphae, spores and other Mycoses (Fungus Infections)
structures. Teased mounts are prepared Infection caused by fungus is known as mycosis
in lactophenol cotton blue (LCB) which (plural mycoses).
contains lactic acid, phenol and cotton blue.
Microscopic characteristics that should be Classification of mycoses
obtained as the following:
a. Septate versus sparsely septate hyphae Classification of fungal disease according to
b. Spores or conidia primary sites of infections is as follows (Table 72.1):
ii. Cellophane tape preparation: It should be A. Superficial mycoses: These infections are
read within 30 minutes and then dis­carded. limited to the outer­most layers of the skin and
iii. Slide culture: Slide culture provides a hair. These include:
useful technique for demonstrating the a. Infection of skin caused by Malassezia
natural morphology of fungal structures. furfur (pityriasis versicolor) and Exophiala
3. Tests for the identification of molds: werneckii (tinea nigra), and
i. Hair perforation test: It is done to differtiate b. Infection of hair caused by Piedraia hortae
T. rubrum, from T. mentagrophytes,. (black piedra) and Trichosporon beigelii
ii. Urease test: It is done to help differentiate T. (white piedra).
menta­gro­phytes from T. rubrum B. Cutaneous mycoses: Infections that extend
iii. Thiamine requirement deeper into the epidermis as well as invasive
iv. Trichophyton agars hair and nail diseases.
v. Growth on rice grains.
4. Tests for the identification of yeasts Examples
Germ tube production, carbohydrate assimilation, a. Infection of skin, hair and nail caused by
chromogenic substrates, cornmeal agar, potassium der­matophytes.
nitrate assimilation, temperature studies and urease. b. Infection of skin, nail and mucous membrane
caused by C. albicans and other Candida
E. Serologic Tests specicies.
The most common tests for C. Subcutaneous mycoses: These infections
a. Fungal anti­bodies: involve the dermis, subcutaneous tissues,
1. Immunodiffusion; 2. countercurrent immuno- muscle, and fascia.
electrophoresis (CIE); 3. whole cell agglutination; Examples: Mycetoma, chromomycosis, sporotri­
4. complement fixation; 5. Enzyme-linked chosis and rhinosporidiosis.
immunosorbent assay (ELISA). D. Systemic mycoses: Systemic mycoses-infections
b. Antigen detection: that originate primarily in the lung but that
1. Latex particle agglutination; 2. ELISA. may spread to many organ systems. Systemic
mycoses are caused by inhalation of airborne
F. Polymerase Chain Reaction (PCR) spores.
Polymerase chain reaction (PCR) for detection of Examples: Blastomycosis, histoplasmosis, cocci­
fungal DNA in clinical material. dioidomycosis and paracoccidioidomycosis.
Chapter 72: General Properties, Classification and Laboratory Dignosis of Fungi  | 507
Table 72.1  The major mycoses and causative fungi
Type of mycosis Mycosis Causative Fungal Agents
A. Superficial Pityriasis versicolor Malassezia species
Tinea nigra Hortaea werneckii
White piedra Trichosporon species
Black piedra Piedraia hortae
B. Cutaneous Dermatophytosis Microsporum species, Trichophyton species, and
Epidermophyton floccosum
Candidiasis of skin, mucosa, or nails Candida albicans and other candida species
C. Subcutaneous Sporotrichosis Sporothrix schencki
Chromoblastomycosis Phialophora verrucosa, Fonsecaea pedrosoi, others
Mycetoma Pseudallescheria boydii, Madurella mycetomatis, others
Phaeohyphomycosis Exophiala, bipolaris, exserohilum, and others
D. Systemic (prlmary, Coccidioidomycosis Coccidioides immitis, C. posadasii
endemic) Histoplasmosis Histoplasma capsulatum
Blastomycosis Blastomyces dermatitidis
Paracoccidioidomycosis Paracoccidioides brasiliensis
E. Opportunistic Systemic candidiasis Candida albicans and other candida species
Cryptococcosis Cryptococcus neoformans
Aspergillosis Aspergillus fumigatus and other aspergillus species
Mucormycosis (zygomycosis) Species of Rhizopus, Absidia, Mucor, and other
zygomycetes
Penicilliosis Penicillium marneffei

E. Opportunistic mycoses: Opportunistic infection Important Questions


occurrs in patients with debilitating dis­eases.
1. Describe the laboratory diagnosis of fungal diseases.
Opportunistic infections are caused mainly by 2. Write short notes on classification of fungi.
fungi that are normally avirulent.
Examples: Aspergillosis, penicilliosis, zy­gomycosis Multiple choice questions (MCQs)
or mucormycosis, candidiasis and cryp­tococcosis.
1. All the following fungi are mold except:
F. Miscellaneous mycoses: Penicilliosis, otomy­ a. Aspergillus fumigates
cosis and keratomycosis. b. Rhizopus
c. Absidia
Key Points d. Cryptococcus neoformans
2. All the following are examples of dimorphic fungi
™™ Mycology is the study of fungi except:
™™ The cell wall of fungi possesses two characteristic a. Coccidioides immitis
cell structures: chitin and ergosterol b. Cryptococcus neoformans
™™ Fungi grow in two basic forms, as yeasts and molds.
c. Sporothrix schenkii
Elongation of the cell produces a tubular, thread like
d. Blastomyces dermatitidis
structure called hypha (septate or nonseptate). A
tangled mass of hyphae constitutes the mycelium. 3. Which of the following methods can be used for
Fungi which form mycelia are called molds or laboratory diagnosis of fungal infections?
filamentous fungi a. Direct microscopy of specimens
™™ The fungi are classified in the phylum Thallophyta. b. Culture
The phylum consists of four classes of fungi c. Histology
Zygomycetes, Ascomycetes, Basidiomycetes, and d. All the above
Deuteromycetes or Fungi Imperfecti 4. Which of the following culture media can be used
™™ The fungi can also be classified as yeast, yeast-like for growing fungi?
fungi, molds, and dimorphic fungi depending on a. Sabouraud’s dextrose agar
their morphology b. Brain heart infusion agar
™™ Laboratory diagnosis of fungal infections depends
c. Both the above
on: Direct microscopy, culture, serological tests,
nonculture methods and molecular methods
d. None of the above
™™ Mycoses (fungus infections): Infection caused by Answers (MCQs)
fungus is known as mycosis (plural mycoses). 1. d; 2. d; 3. d; 4. c

www.ebook3000.com
73
Chapter
Superficial, Cutaneous and
Subcutaneous Mycoses

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Describe causative agets of ectothrix and endothrix
be able to: ∙∙ Describe the following: mycetoma; chromoblasto-
∙∙ List superficial, cutaneous and subcutaneous mycosis; sporotrichosis; rhinosporidiosis.
mycoses

A. SUPERFICIAL MYCOSES
These infections are limited to the outer­most layers
of the skin and hair. These include:
a. Infection of skin: Caused by Malassezia furfur
(pityriasis versicolor) and Exophiala werne­ckii
(tinea nigra)
b. Infection of hair: Caused by Piedraia hortae
(black piedra) and Trichosporon beigelii (white
piedra).

a. Infection of Skin
Pityriasis Versicolor (Tinea Versicolor) Fig. 73.1: Morphology of pityriasis versicolor
Pityriasis versicolor (Tinea versicolor) is a chronic, Creamy colony develop within 5–7 days at 37°C.
usually asymptomatic, involvement of the stratum Lactophenol cotton blue mount of the colonies
corneum, characterized by discrete or confluent show budding yeast cells along with a number
macular areas of discoloration or depigmentation of bottleshaped cells. Hyphae are occasionally
of the skin. The areas involved are mainly the chest, seen in culture.
abdomen, up­per limbs and back. The disease is
worldwide in distribution. Tinea Nigra
Causative agent: A lipophilic, yeast-like fungus Tinea nigra (or tinea nigra palmaris) is a localized
Pityro­sporum orbiculare (Malas­sezia furfur). infection of the stratum corneum, particularly of
the palms, producing black or brownish macular
Laboratory Diagnosis lesions. It is caused by the dematiaceous fungus
A. Direct microscopy: The diagnosis is confirmed Hortaea (Exophiala) werneckii.
by direct microscopic examination of scrap­ings
of infected skin, treated with 10–20% KOH or Laboratory Diagnosis
stained with calcofluor white. Demonstration of A. Direct microscopy: Microscopic examination
clusters of the characteristic round yeast cells with of skin scrapings from the periphery of the
short, stout hyphae, which may be curved and lesion will reveal brownish, branched, septate
occasionally branched, is diagnostic (Fig. 73.1). hyphae and budding cells (Fig. 73.2).
B. Culture: The fungus can be grown on Sabour­ B. Culture: On Sabouraud agar the fungus
aud’s agar, covered with a layer of olive oil. develops as grey, yeast-like colonies, which

Chap-73.indd 508 15-03-2016 11:25:05


Chapter 73: Superficial, Cutaneous and Subcutaneous Mycoses  | 509
Classification
Dermatophytes have been classified into three
genera:
A. Trichophyton: Tri­chophyton species infect hair,
skin or nails.
B. Microsporum: Microsporum species infect only
hair and skin.
C. Epidermo­phyton: Epidermophyton attacks the
skin and nails but not the hair.
Fig. 73.2: Hortaea werneckii About 40 species of dermatophytes are known
to cause infection in humans and animals.
gradually become, more mycelial and darker
Dermatophytes are probably restricted to the non­
coloured with age.
viable skin because most are unable to grow at 37°C
b. Infection of Hair or in the presence of serum.
Piedra Classification depeding on habitat: Dermato­
phytes are classified as anthropophilic, geophilic,
Piedra is a fungal infection of the hair, characterised zoophilic, or depending on whether their usual
by the appearance of firm, irregular nodules habitat is soil, animals or humans.
along the hair shaft. Two varieties of piedra are 1. Anthropophilic species: Human beings are
recognised–black piedra caused by Pie­draia hortae the main or only hosts for anthropophilic
and white piedra caused by Trichospo­ron beigelii. dermatophytes. Exam­ples are T. rubrum, M
Black Piedra audouinii and Epidermo­phyton floccosum.
2. Zoophilic spe­cies: These are natural parasites
This condition, caused by Piedraia hortae. It
of animals. Examples are T. verrucosum in cattle
is characterized by the presence of black, hard
and M. canis in dogs and cats.
nodules up to 1 mm in diameter, mainly on the
3. Geophilic species: They occur naturally in soil
hairs of the scalp.
and are relatively less pathogenic for human
Laboratory Diagnosis beings. Ex­amples are M. gypseum and T. ajelloi.
A. Direct microscopy: Crushing the nodules Identification
reveals the sexual reproductive phase, club-
In skin, they are diagnosed by the presence of
shaped asci, each with eight ascospores.
hyaline, septate, branching hyphae or chains of
B. Culture: Culture is not necessary.
arthroconidia. In cultures on Sabouraud’s agar,
White Piedra they form characteristic colonies consisting of
septate hy­phae and two types of asexual spores,
White piedra, an infection of the hair, is caused by the
microconidia and macroconidia. Differentiation
yeast-like organism Trichosporon beigelii. Axillary,
into the three genera is based mainly on the nature
pubic, beard, and scalp hair may be infected.
of macro­conidia (Fig 73.3). In culture, the many
B. CUTANEOUS MYCOSES species are closely related and often difficult to
identify.
Infections that extend deeper into the epidermis as
well as invasive hair and nail disease. Morphology and Identification
Examples: Dermatophytes are identified by their colonial
a. Infection of skin, hair and nail caused by der­ appear­ance and microscopic morphology after
matophytes. growth for 2 weeks at 25°C on Sabouraud’s dextrose
b. Infection of skin, nail and mucous membrane agar.
caused by C. albicans and other Candida
specicies. Trichophyton
Colonies may be powdery, vel­vety or waxy, with
Dermatophytes pigmentation characteristic of dif­ferent species.
The dermatophytes are a group of closely related Microconidia are abundant and are arranged in
filamen­t ous fungi that infect only superficial clusters along the hyphae or borne on co­nidiophores.
keratinized tis­sues—the skin, hair and nails. Macroconidia are relatively scanty. They are
Dermatophytoses are among the most prevalent generally elongated, with blunt ends (Figs 73.3
infections in the world. Being superfi­cial, dermatophyte and 73.4). Macroconidia have distinctive shapes in
(ringworm) infections have been rec­ognized since different spe­cies and are of importance in species
antiquity. identification. Some species possess special hyphal

www.ebook3000.com
Chap-73.indd 509 15-03-2016 11:25:05
510  |  Section 5: Medical Mycology

Fig. 73.4: Trichophyton species. Macroconidium, spiral


hypha and typical microconidia

M. canis
M. canis forms numerous thick-walled, 8–15 celled
macroconidia with curved or hooked spiny tips. A
Fig. 73.3: Characteristic macroconidia of dermatophytes. (1) lemon-yellow or yellow-orange pigment develops
Cylindrical in Trichophyton; (2) Fusiform in Microsporum; and on the reverse of the colony. It is zoophilic.
(3) Club-shaped in Epidermophyton
M. gypseum
characters, such as spiral hyphae, racquet mycelium M. gypseum colonies grow rapidly producing a flat,
and favic chandeliers (Fig. 73.4). spreading, powdery surface that is cinnamonbuff to
brown which consists of abundant macroconidia.
Important Species They are boat-shaped, rough walled with 4 to 6
T. rubrum, T. mentagrophyte, T. tonsurans, T. septa. It is geophilic.
schoenleinii, T. violaceum.
Pathogenicity
T. rubrum: The typical colony of T. rubrum has a Dermatophytes grow only on the keratinised
white, cottony surface and a deep red, nondiffusible layers of the skin and its appendages and do not
pigment when viewed from the reverse side of the ordinarily penetrate the living tissues. Lesions vary
colony. The microconidia are small and piriform considerably according to the site of the infection
(pear-shaped) and clubshaped thinwalled macro­ and the species of fungus involved.
conidia (scanty).
Dermatophytids (or ‘id’ reaction): Hypersensitivity
T. mentagrophyte: T. mentagrophyte has grape- to fungus antigens may play a role in pathogenesis
like clusters of microconidia (Fig. 73.4) and cigar and is probably responsible for the sterile
shaped macroconidia.It has no red pigment and vesicular lesions, sometimes seen in sites distant
urease positive. Hair perforation test is positive. from the ringworm. These lesions are called
T. tonsurans: T. tonsurans produces a flat, dermatophytids (or ‘id’ reaction). Hypersensitivity
powdery to velvety colony on the obverse surface can be demonstrated by skin testing with the fungus
that becomes reddish-brown on reverse. The antigen, trichophytin.
microconidia are mostly elongate (Fig. 73.4). Types of hair infections: Three types of hair
infection can be seen in 10% KOH wet mounts:
Microsporum 1. Ectothrix: In this, the hyphae are sparsely
Colonies are cotton-like, velvety or powdery, with distributed within hair shaft with a sheath
white to brown pigmentation. Micro­conidia are of arthrospores on the surface of hair shaft.
relatively scanty and are not distinctive. Macroconidia It is caused by M. audouinii, M. canis and T.
are the predominant spore form. They are large, mentagrophytes (Fig. 73.5A).
multicellular, spindle shaped structures, borne singly 2. Endothrix: In this, the arthrospore forma­tion
on the ends of hyphae. Both types of conidia are occurs entirely within the hair completely filling
borne singly in these genera. Microsporum species the hair shaft. This is caused by T. tons­urans and
infect only hair and skin but usually not the nail. T. violaceum (Fig. 73.5B).
Important species are M. audouinii, M. canis, 3. Favus: In this, there is sparse hyphal growth and
M. gypseum formation of air spaces within the hair shaft.
This is caused by T. schoenleinii (Fig. 73.5C).
M. audouinii
M. audouinii rarely forms conidia in the culture but Clinical Findings
many thick-walled chlamydospores (chlamydo­ Dermatophyte infections were mistakenly termed
conidia) may be present. It is anthropophilic. ring­worm or tinea. Table 73.1 lists the clinical

Chap-73.indd 510 15-03-2016 11:25:06


Chapter 73: Superficial, Cutaneous and Subcutaneous Mycoses  | 511
Table 73.1:  Clinical types of dermatophytoses and
their common causative agents
Disease Common causative agents
Tinea capitis Microsporum any species,
Trichophyton most species
Favus T. schoenleinii, T. violaceum,
M. gypseum
Tinea barbae T. rubnun, T. mentagrophytes,
T. verrucosum
Tinea imbricata T. concentricum
Tinea corporis T. rubrum and any other
dermatophyte
T. cruris E. floccosum, T. rubrum
T. pedis T. rubrum, E. Boccosum
Ectothrix hair infection Microsporum species,
A B C
T. rubrum, T.menrngrophytes
Figs 73.5A to C: Various forms of hair invasion by dermato­
Endothrix hair infection T. schoenieinii, T. tonsurans,
phytes as seen in longitudinal and transverse sections of hair
T. violaceum
shaft: (A) Ectothrix with hyphae sparsely distributed within
hair shaft and a sheath of arthroconidia on the outside; (B)
Endothrix with heavy arthroconidia formation completely with or without calcofluor white. In skin or nails,
filling hair shaft; (C) Favus showing sparse hyphal growth and branched septate hyphae or chains of arthroconidia
formation of air spaces (arthrospores) are seen.
Selection of infected hair for examination is
types of dermatophytoses and their common
facilitated by exposure to UV light (Wood’s lamp).
causative agents.
Infected hair will be fluorescent. Two types of hair
The clinical forms are based on the site of
infection may be distinguished in wet mounts,
involvement.
‘ectothrix’ (Figs 73.5A to C) and ‘endothrix’.
1. Tinea corporis (Tinea glabrosa): It is ringworm
of the smooth or nonhairy skin of the body. A C. Culture
special type Tinea imbricata is characterized by
Specimens are inoculated onto inhibitory mold
extensive con­centric rings of papulosquamous
agar or Sabouraud’s agar slants containing cyclo­
scaly patches.
heximide and chloramphenicol to suppress mold
2. Tinea cruris or jock itch: When the infection
and bacte­rial growth, incubated for 1–3 weeks at
occurs in the groin and the perineum.
room temperature, and further examined in slide
3. Tinea manus: Ringworm of the hands or fingers.
cultures if necessary. Species are identified on the
4. Tinea barbae or barber’s itch: It is involvement
basis of colonial morphol­ogy (growth rate, surface
of the bearded areas of the face and neck.
texture, and any pigmenta­t ion), microscopic
5. Tinea pedis or ath­letes’ foot: It is ringworm of
morphology (macroconidia, micro­conidia), and,
the foot.
in some cases, nutritional requirements.
6. Tinea unguium (onychomycosis): Nail
infection may follow prolonged tinea pedis. Epidemiology
7. Tinea capitis: Ringworm of the scalp and hair.
Dermatophytosis occurs throughout the world
8. Favus: It is a chronic type of ringworm in which
Tinea pedis is rare in the tropics where most walk
dense crusts (scutula) develop in the hair
barefoot. In India, tinea capitis occurs more often
follicles, which lead to alopecia and scarring.
in the native children than in Europeans.
9. Kerion: Scalp infection sometimes produces
severe boggy le­sions with marked inflammatory Treatment and Prevention
reaction called kerion. Topical therapy is satisfactory for most skin
infections. Topical agents include azole compounds,
Laboratory Diagnosis
terbinafine, amorolfine and ciclopirox olamine.
A. Specimens Oral griseofulvin is useful for scalp, skin and
Specimens consist of scrapings from both the skin fingernail infections. Relatively little has been done
and the nails plus hairs plucked from involved areas. to control the spread of ringworm.
B. Microscopic Examination
C. SUBCUTANEOUS MYCOSES
The routine method of diagno­sis is by the examin­
ation of KOH mounts. Specimens are placed on The principal subcutaneous mycoses are mycetoma,
a slide in a drop of 10–20% potassium hydroxide, chromomycosis, sporotrichosis and rhinosporidiosis.

www.ebook3000.com
Chap-73.indd 511 15-03-2016 11:25:07
512  |  Section 5: Medical Mycology
1. Mycetoma Table 73.2  Important causative agents and color
Mycetoma is a chronic, granulomatous infection of the grains of various types of mycetoma
of the skin, subcutaneous tissues, fascia and bone, Causative agent Color of the grains
which most often affects the foot or the hand. A. Eumycetoma
The disease was originally reported by Gill (1842) •  Madurella mycetomatis Black
from Madurai, South India, and Carter (1860)
•  M. grisea Black
established its fungal etiology. It is, therefore,
•  Exophiala jeanselmei Black
commonly known as Maduramycosis or Madura
foot. •  Pseudallescheria boydii White-yellow
Mycetoma occurs worldwide. The disease is •  Acremonium falciforme White-yellow
most prevalent in tropical and subtropical regions •  Aspergillus nidulans White
of Africa, Asia and Central America. In India, it is B. Actinomycetoma
quite common in Tamil Nadu but rare in Kerala. •  Actinomadura madurae White-yellow
•  A. pellitieri Red-yellow
Etiology
•  Nocardia asteroids White-yellow
It can be divided into three types, eumycetomas,
•  N. brasiliensis White
actinomycetomas and botryomycosis. A similar
condition called ‘botryomycosis’ is caused by •  Streptomyces somaliensis Yellow
Staphylococcus aureus and some other bacteria. C. Botryomycosis
Important causative agents of these are given in •  Staphylococcus species White
Table 73.2. •  Streptococcus species White
•  Escherichia coli White
Pathogenesis •  Proteus species White
Triad of symptoms: Infection follows traumatic •  Pseudomonas aeruginosa . White
inoculation of the organism into the subcutaneous Actinobacillus lignieresi Yellow
tissue from soil or vegetable sources, usually on
thorns or splinters and results in tume­factions,
Treatment
deformities and draining sinuses discharg­ing
fungal colonies called grains or granules (triad of The management of eumycetoma is difficult,
symptoms). Subcutaneous tissues of the feet, lower involving surgical debridement or excision and
extremities, hands, and exposed areas are most chemotherapy. Actinomycetoma responds well to
often involved. rifampicin in combi­nation with sulfonamides or
cotrimoxazole.
Grains: Within host tissues the organisms develop
to form compacted colonies (grains) 0.5–2 mm 2. Chromoblastomycosis
in diameter, the color of which depends on the This disease, also known as chromomycosis, is a
organism responsible. The color and consistency chronic, localized disease of the skin and subcutan­
of the grains vary with the different agents causing eous tissues. The disease is mainly encountered in
the disease (Table 73.2). the tropics.
Laboratory Diagnosis Etiological agents: The etiological agents are soil
inhabiting fungi of the family Dematiaceae.
A. Direct Examination All are dematiaceous fungi which are darkly
The presence of grains in pus collected from pigmented fungi. These include: Phialophora
draining sinuses or in biopsy material is diagnostic. verrucosa, Fonse­caea pedrosoi, Rhinocladiella
The grains are visible to the naked eye and their aquaspersa, Fonsecaea com­pacta, and Cladophial­
color may help to identify the causal agent. Grains ophora carrionii.
should be examined microscopically to differentiate They enter the skin by traumatic implantation.
between actinomycetoma and eumycetoma. The lesion develops slowly around the site
Material from actinomycetoma grains may be Gram of implantation. The infection is chronic and
stained to demonstrate the gram-positive filaments. characterized by the slow development of progres­
sive granulomatous lesions that in time induce
B. Culture hyperplasia of the epidermal tissue.
Samples should also be cultured, at both 25–30°C
and 37°C, on brain-heart infusion agar or blood Laboratory Diagnosis
agar for acti­nomycetes and on Sabouraud agar A. Microscopic examination: Histologically, the
(without cyclohex­imide) for fungi. The fungi that lesions show the presence of the fungus as
cause eumycetoma are all septate molds that round or irregular, dark brown, yeast­like bodies
appear in culture within 1–4 weeks. with septae, called sclerotic cells (Fig. 73.6).

Chap-73.indd 512 15-03-2016 11:25:07


Chapter 73: Superficial, Cutaneous and Subcutaneous Mycoses  | 513

Fig. 73.7: Sporothrix (Sporotrichum) schenckii: culture mount


showing fine branching hyphae and pear shaped conidle
borne in rosette clusters at tips of lateral branches and singly
Fig. 73.6: Chromoblastomycosis: KOH mount of lesion along sides of hyphae
large septate ‘sclerotic bodies’

Diagnosis can be established by demonstration on delicate sterigmata (Fig. 73.7). Conidia are
of these sclerotic bodies in KOH mounts or in also produced along the sides of the hyphae.
sec­tions, and by culture on Sabouraud’s agar. C. Serology: A latex agglutination test is of value
B. Culture: Culture on Sabouraud agar at 25–30°C for the diagnosis of the extracutaneous forms of
yields slow-growing, greenish grey to black, sporotrichosis.
compact, folded colonies. Cultures should be D. Skin test: A skin test with sporotrichin antigen
incubated for 4–6 weeks. is positive in almost all patients with cutaneous
sporotrichosis.
Phaeohyphomycosis E. Animal inoculation: Rats are highly susceptible
Phaeohyphomycosis is a term applied to infections and can be infected by intraperitoneal or
char­acterized by the presence of darkly pigmented intratesticular inoculation.
septate hyphae in tissue. The sites of lesions may be
cutaneous, subcutaneous, deeper tissues, or organs 4. Rhinosporidiosis
like the brain or lung. Sclerotic cells or granules are Rhinosporidiosis is a chronic granulomatous
not found. The fungi appear in lesions as distorted disease characterized by the development of large
hyphal strands. polyps or wart-like lesions in the nose, conjunctiva
and occ­asionally in ears, larynx, bronchus, penile
3. Sporotrichosis urethra, vagina, rectum and skin. More than 90% of
Sporotrichosis is a chronic, pyogenic granulomatous the cases have been reported from India, Sri Lanka
infection of the skin and subcutaneous tissues and South America.
which may remain localized or show lymphatic
Etiology: The causative fungus Rhinosporidium
spread. The disease is worldwide and is rare in
seeberi.
Europe.
Causative agent: Sporothrix schenckii, a saprophyte Mode of infection: The mode of infection is not
in nature. known, though infection is believed to originate
from stagnant water or aquatic life. It is believed
Pathogenesis that fish may be the natural hosts of R. seeberi.
The fungus is a sapro­phyte found widely on plants,
thorns and timber. Infec­tion is acquired through Laboratory Diagnosis
thorn pricks or other minor injuries. It has not been cultured and animal inoculation is
also not successful.
Laboratory Diagnosis
Diagnosis is made by culture as frequently the Demonstration of Sporangia
fungus may not be demonstrable in pus or tissues. Diagnosis depends on the demonstration of
A. Microscopic examination: Direct microscopy sporangia. Direct examination of the surface of
is of little value. the polypoid growth and histologic examination
B. Culture: SDA or blood agar are the media used. are the only ways to make a diagnosis. R. seeberi
S. schenckii is a dimorphic fungus occurring can be identi­fied in hematoxylin and eosin-stained
in the yeast phase in tissues and in cultures at sections, but sometimes one may need special
37°C, and in the mycelial phase in nature and stains. Histologically, the lesion is composed of
in cultures at room temperature. The septate large numbers of fungal spherules embedded in
hyphae are very thin (1-2 mm diameter) and a stroma of connective tissue and capillaries. The
carry flower-like clusters of small conidia borne spherules are 10–200 mm in diameter and contain

www.ebook3000.com
Chap-73.indd 513 15-03-2016 11:25:08
514  |  Section 5: Medical Mycology

™™ Rhinosporidiosis is a chronic granulomatous


disease characterized by the development of large
polyps or wart-like lesions in the nose, conjunctiva
and occ­asionally in ears, larynx, bronchus, penile
urethra, vagina, rectum and skin. The causative
fungus is Rhinosporidium seeberi
™™ Subcutaneous phycomycosis: The causative agent
is Basio­diobolus haptosporus.

IMPORTANT QUESTIONS
1. Discuss the morphology, cultural characters and
Fig. 73.8: Rhinosporidiosis: Sporangium with numerous pathogenicity of dermatophytes.
endospores 2. Describe the etiology, pathogenesis and laboratory
diagnosis of mycetoma.
thousands of endospores (6–7 μm in diameter) 3. Discuss different kinds of subcutaneous mycoses.
(Fig. 73.8). These spores develop into new 4. Write short riotes on:
sporangia when released. a. Superficial mycoses
b. Dermatophytes
5. Subcutaneous Phycomycosis c. Mycetoma or Madura foot or maduramycosis
In this condition, a painless subcutaneous nodule d. Chromoblastomycosis or verrucous dermatitis
develops which enlarges to involve a whole limb e. Sporotrichosis
or large areas of the body. f. Rhinosporidiosis.
Causative agent: is Basio­diobolus haptosporus,
a saprophytic phycomycete. MULTIPLE CHOICE QUESTIONS (MCQs)
Key Points 1. Black piedra is a superficial infection of the hair
caused by:
Superficial Mycoses a. Malassezia furfur
™™ Pityriasis versicolor or tinea versicolor is caused by
b. Cladosporium werneckiib
Malassezia furfur (Pityrosporum orbiculare)
™™ Tinea nigra is an infection of keratinized layer of skin
c. Trichosporon beigelli
caused by Hortaea (Exophiala) werneckii d. Piedraia hortae
™™ Black piedra is a superficial infection of the hair 2. All the following are anthropophilic dermatophytes
caused by Piedroiohortae, a dematiaceous fungus except:
™™ White piedra is an infection of the hair caused by a. Trichophyton violaceum
yeast-like organism, Trichosporon beigelii b. Trichophyton rubrum
Cutaneous Mycoses c. Epidermophyton floccosum
™™ The dermatophytes infect only superficial keratinized d. Microsporum audouinii
structure such as skin, hair and nail but not deeper 3. Urease test is positive by:
tissues
a. Trichophyton violaceum
™™ Dermatophytes belong to three genera: Trichophyton,
Microsporum and Epidermophyton
b. Trichophyton rubrum
™™ Clinical findings: Dermatophyte infections were c. Trichophyton mentagrophytes
mistakenly termed ring­worm or tinea because of d. Epidermophyton floccosum
the raised circular lesions 4. The major causative agent of mycetoma in India is:
™™ Laboratory diagnosis is based on microscopy and a. Actinomadura madurae
culture. The differentiation of the three genera is b. Streptomyces somaliensis
based mainly on nature of macroconidia c. Nocardia brasiliensis
Subcutaneous Mycosis d. Actinomyces pelletieri
™™ Mycetoma is a chronic, granulomatous infection
5. The sclerotic bodies are useful for diagnosis of:
of the skin, subcutaneous tissues, fascia and bone,
a. Sporotrichosis b. Mycetoma
which most often affects the foot or the hand
™™ It can be divided into three types, eumycetomas,
c. Chromoblastomycosis d. Rhinosporidiosis
actinomycetomas and botryomycosis 6. Which of the following fungi has not been cultured:
™™ Chromoblastomycosis or chromomycosis is caused a. Sporothrix b. Rhinosporidium
by fungi collectively called dematiaceous fungi c. Blastomyces d. Acremonium
™™ Sporotrichosis is caused by Sporothrix schenckii, a
saprophyte in nature
ANSWERS (MCQs)
1. d; 2 a; 3. c; 4. a; 5. c; 6. b

Chap-73.indd 514 15-03-2016 11:25:09


74
Chapter

Systemic Mycoses

Learning Objectives

After reading and studying this chapter, you should ∙∙ Descrbe histoplasmosis
be able to:
∙∙ List of fungi causing systemic infections

Introduction
These are the include blastomycosis, histoplasmosis,
coccidioidomycosis and paracocci­dioidomycosis.

1. Blastomycosis
Blastomycosis is a chronic infection of the lungs
which may spread to other tissues, particularly
skin, bone and genitourinary tract.
It is caused by Blastomyces dermatitidis, a
dimorphic fungus. The disease has been called Fig 74.1: Blastomyces dermatitidis: yeast and mycelial
North American blastomycosis, because it is forms
endemic and most cases occur in the United States fila­mentous with septate hyphae and many round or
and Canada. oval conidia, and in older cultures, chlamydospores
also.
Pathogenesis
Laboratory Diagnosis
Soil is considered to be the source of infection,
which is acquired by inhalation. Human infection A. Specimens: Sputum or pus, exudates, urine
is initiated in the lungs. and biopsy lesions.
B. Microcopic examination: Potassium hydro­xide
1. Asymptomatic
(10%) mount of specimens may show char­
2. Chronic pneumonia acteristic thick-walled yeast cells with a single
3. Disseminated diseases: They form multiple broadbased bud.
abscesses in various parts of the body. C. Culture: Colonies usually develop on Sabouraud’s
4. Cutaneous blastomycosis: The cutaneous dextrose agar or enriched blood agar at 30°C
disease is usually on the skin of the face or other within 2 weeks.
exposed parts of the body. D. Serology: Antibodies can be measured by
complement fixation (CF) test, immuno­diffusion
Morphology (ID) tests and enzyme immunoassay (EIA).
Blastomyces dermatitidis is a dimorphic fungus. In Treatment: Severe cases of blastomycosis are
tissue and in cultures at 37°C, the fungus appears treated with amphotericin B.
as budding yeast cells, which are large (7–20 μm)
and spherical, with thick, double-contoured walls. 2. Paracoccidioidomycosis
Each cell carries only a single broad-based bud This is a chronic granulomatous disease of the skin,
(Fig. 74.1). At room temperature, the culture is mucosa, lymph nodes and internal organs. As the

www.ebook3000.com
516  |  Section 5: Medical Mycology

A B
Figs 74.3A and B: Coccidioides immitis: (A) Arthrospores
formation; (B) Spherule formation with endospores

3. Disseminated disease: Chronic progressive


disseminated disease (coccidioidal granuloma)
A B
which is highly fatal.
Figs 74.2A and B: Paracoccidioides brasiliensis. (A) yeast
phase; (B) mycelial phase Morphology
disease is con­fined to South America, it is called The fungus is dimorphic. The mycelial phase consists
‘South American blastomycosis’. of hyphae which fragment into arthrospores which
are highly infectious. In culture and in soil C. immitis
Causative fungus: It is caused by Paracoccidioides grows as a mould, producing large numbers of barrel-
brasiliensis, a dimorphic fungus. shaped arthrospores (4 × 6 μm diameter) which are
highly infectious. They characteristically alter­nate
Pathogenesis
with smaller intervening empty cells (Fig. 74.3A).
It is charac­terized by primary pulmonary infection The yeast form is a spherule (15–75 μm diameter)
that spreads by hematogenous route to mucosa with a thick, doubly refractile wall and filled with
of the nose, mouth and the gastrointestinal tract, endospores. Endospores develop to form new
skin, lymphatic system, and the internal organs spherules in adjacent tissue or elsewhere in the
producing chronic granulomatous reaction. body (Fig. 74.3B).
Laboratory Diagnosis Laboratory Diagnosis
1. Specimens: Sputum or pus, crusts and biopsies 1. Specimens
from granulomatous lesions.
Sputum, pus and biopsy material.
2. Direct microscopy: Microscopy of sputum or
pus, crusts and biopsies from granulomatous 2. Microscopic Exami­nation
lesions usually reveals numerous yeast cells (10–
Diagnosis may be made by microscopic exami­
60 μm) with multiple buds, which is diagnostic
nation of specimens.
(Figs 74.2A and B). Tissue sections should be
stained with H and E, PAS and GMS. 3. Culture
3. Culture: P. brasiliensis grows in the mycelial Specimen is inoculated on SDA medium in the test
phase in culture at 25–30°C, and in the yeast tube and incubated at 25–30°C. The arthroconidia
phase in tissue or at 37°C. Blood agar (without are highly infectious. Consequently, Petri dishes
cyclohexi­m ide) and incubation at 37°C is should never be used for isolation of the organ­ism.
recommended for isolation of the yeast phase
(tissue form). Mycelial (mold) phase of the 4. Serological Tests
fungus devel­ops on SDA incubated at 25–30°C. Serological tests, such as precipitin test, latex
agglutina­tion test and complement fixation test
3. Coccidioidomycosis play an important part in diagnosis.
This is primarily an infection of the lungs caused by
Coccidioides immitis, a dimorphic fungus found in
5. Skin Tests
the soil of semi-arid areas, mainly in the south-west Skin test is done with coccidioidin, a culture filtrate
USA and northern Mexico. antigen from fungus. The test becomes positive
between 3 and 21 days of symptoms.
Clinical Features
4. HISTOPLASMOSIS
1. Asymptomatic
2. Primary pulmonary disease: This condition is Histoplasmosis is an intracellular infection of
called valley fever, San Joaquin Valley fever or the reticuloendothelial system caused by the
desert rheumatism. dimorphic fungus Histoplasma capsulatum.
Chapter 74: Systemic mycoses  | 517
Pathogenesis 25–30°C. On SDA, it forms white to tan fluffy colony
Infection is acquired by inhalation. Most infections with septate branching hyphae with two types
are asymptomatic. Some infected persons develop of unicellular, asexual spores. (1) Large round,
pulmonary disease which resembles tuberculosis. tuberculate macroconidia (8–14 μm) are most
prominent and are diagnostic. (2) Small spores or
1. Pulmonary Infection microconidia are sessile or stalked, smooth-walled,
round to pyriform, 2–4 μm in diameter (Fig. 74.5).
A chronic form of histoplasmosis occurs mainly
in adults. The clinical picture- closely resembles 4. Serological Tests
tuberculosis.
Serological tests like latex agglutination, precipita­
2. Disseminated Histoplasmosis tion and complement fixation become positive two
weeks after infection. Tests for antigen detection
Disseminated infection occurs most often in old
by radioimmunoassay or ELISA are useful, but are
age and infancy, or in individuals with impaired
not widely available.
immune responses. The reticuloendothelial system
is involved with resultant lymphadenopathy, 5. Histoplasmin Skin Test
hepatosplenomegaly, fever, anemia and a high
rate of fatality. Delayed hypersensitivity to the fungus can be
demonstrated by histoplasmin skin test. The test
3. Skin and Mucosa is similar to tuberculin test but antigen used is
histoplasmin. Histoplasmin is a culture filtrate
Granulomatous and ulcerative lesions may develop
antigen of mycelial phase of H. capsulatum.
on the skin and mucosa.
The histoplasmin skin test becomes positive
soon after infection and remains positive for years.
Laboratory Diagnosis A positive ‘histoplasmin skin test’ indicates past or
1. Specimens present infection, but does not differentiate active
Blood films, bone marrow slides, and biopsy and past infections.
specimens may be examined microscopically.
Specimens for culture include sputum, urine, Epidemiology
scrapings from superficial lesions, bone marrow The disease has a worldwide distribution but is
aspirates, and buffy coat blood cells. most common in the USA. Twenty-five authentic
cases of his­toplasmosis have been reported from
2. Microscopic Examination India. The etiologic agent of histoplasmosis,
Microscopy of smears of sputum or pus should be H. capsulatum var. capsulatum, grows in soil
stained by the Wright or Giemsa procedure. Liver with a high nitrogen con­tent, especially in areas
or lung biopsies stained with PAS or methenamine- contaminated with the excreta of bats and birds.
silver may provide a rapid diag­nosis of disseminated
histoplasmosis in some patients. H. capsulatum Treatment
is seen as small, oval yeast cells (2–4 μm in Amphotericin B is the treatment of choice for
diameter), typically packed within the cytoplasm most forms of disseminated histoplasmosis;
of macrophages or monocytes (Fig. 74.4). this is followed by oral itraconazole in immuno­
compromised patients.
3. Culture
Specimens are cultured in rich media, such African Histoplasmosis
as glucose­cysteine blood agar at 37°C and on This disease is caused by H. capsulatum val. dub­
Sabburaud’s agar or inhibitory mold agar at oisii. H. capsulatum val’. duboisii is morphologically

Fig. 74.4: H. capsulatum: yeast cells in macrophage Fig. 74.5: H. capsulatum: mycelial form

www.ebook3000.com
518  |  Section 5: Medical Mycology
identical to H. capsulatum in its mycelial phase but Important Questions
differs in the yeast phase both in vivo and in vitro 1. Enumerate dimorphic fungi. Describe the morphology,
and H. duboisii is morphologically identical to H. pathogenicity and lab diagnosis of dimorphic fungi.
capsulatum in its mycelial phase but the yeast phase 2. Write short notes on:
has larger cells (12–15 μm diameter). a. Blastomycosis
African histoplasmosis is mainly restricted to b. Coccidioidomycosis or Coccidioides immitis
c. Histoplasmosis or Darling’s disease
the continent of Africa. It involves mainly the skin,
d. Paracoccidioidomycosis or Paracoccidioides
subcutaneous tissues and bones. The lungs are not brasiliensis
commonly affected and the disseminated disease
is infrequent. Multiple choice questions (MCQs)
1. All the following statements are true for spherule
of Coccidioides immitis except:
Key Points
a. It is infective stage of the fungus
™™ Blastomycosis: Blastomycosis is caused by Blasto­ b. It reproduces byendosporulation
myces dermatitidis, a dimorphic fungus. It is also c. It contains endospores
known as North American blastomycosis. d. It is not found in culture
™™ Coccidioidomycosis is primarily an infection of the
2. Which of the following fungi infects reticuloendo­
lungs caused by Coccidioides immitis, a dimorphic thelial system?
fungus. Infection is acquired by inhalation of dust a. Aspergillus fumigatus
containing arthrospores of the fungus. b. Histoplasma capsula tum
c. Trichophyton rubrum
™™ Histoplasmosis: It is primarily a disease of reticulo­
d. All the above
endothelial system caused by an intracellular fungus
3. The diagnostic form of Histoplasma capsulatum is:
Histoplasma capsulatum, a dimorphic fungus.
a. Arthrospore b. Spherule
™™ H. capsulatum causes acute pulmonary histoplasmosis, c. Macroconidia d. Microconidia
chronic pulmonary histoplasmosis, and progressive
disseminated histoplasmosis. Answers (MCQs)
1. a; 2. b; 3. c
75
Chapter

Opportunistic Mycoses

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Discuss cryptococcosis
be able to: ∙∙ Discuss laboratory diagnosis of Cryptococcus
∙∙ Describe diseases caused by Candida albicans neoformans
∙∙ Describe the following: thrush or oral thrush; Germ ∙∙ Describe species of Asperillus
tube test or Reynaulds–Braude phenomenon ∙∙ Describe the following: Aspergillosis; mucormycosis;
∙∙ Discuss laboratory diagnosis of candidiasis pneumocystosis; otomycosis; mycotic keratitis.
∙∙ List opportunistic fungi

OPPORTUNISTIC FUNGI pseudohy­phae when the buds continue to grow but


fail to detach, producing chains of elongated cells
Patients with compromised host defenses, who are that are pinched or constricted at the septations
sus­ceptible to ubiquitous fungi, are referred to as between cells (Figs 75.1 and 75.2).
opportunistic fungi. Healthy people, if exposed to
ubiquitous fungi are usually resistant. Species of Candida
Important species of Candida found in man are:
Causative Fungal Agents (i) C. albicans; (ii) C. stellatoidea; (iii) C. tropicalis;
A. Yeast and yeast-like fungi: Cryptococcus, (iv) C. krusei; (v) C. guilliermondii; (vi) C. parapsilosis;
Candida spp., Torulopsis. (vii) C. glabrata (viii) C. viswanathii
B. Filamentous fungi: Aspergillus, Mucor,
Absidia, Rhizopus, Cephalosporium, Fusarium, Pathogensis
Penicil­lium, Geotrichum, Scopulariopsis. A. Superficial (cutaneous or mucosal) candidiasis
C. Others: Pnuemocystis jiroveci. B. Systemic can­didiasis
A. Superficial (cutaneous or mucosal) candidiasis
A. YEAST AND YEAST-LIKE FUNGI
Candidiasis
Candisois (candidiasis, moniliasis) is an infection
of the skin, mucosa, and rarely of the internal
organs, caused by a yeast-like fungus Candida
albicans, and occasionally by other Candida
species. Several species of the yeast genus Candida
are capable of causing candidiasis. Candido­sis
is an opportunistic endogenous infection, the
com­monest predisposing factor being diabetes.
They are members of the normal flora of the skin,
mucous membranes, and gastrointestinal tract.

Morphology
In culture or tissue, Candida species grow as oval, Fig. 75.1: Sputum specimen illustrating budding yeast cells
budding yeast cells (3–6 μm in size). They also form and pseudohyphae of Candida species

www.ebook3000.com
Chap-75.indd 519 15-03-2016 11:25:32
520  |  Section 5: Medical Mycology

Fig. 75.2: Candida albicans: yeast form and pseudohyphae Fig. 75.3: Gram stained smear of Candida albicans

a. Mucocutaneous Lesions A. Specimens: Specimens include swabs and


1. Oral thrush: Oral thrush can occur on scrapings from superficial lesions, blood, spinal
the tongue, lips, gums or palate. It is found fluid, tissue biopsies, urine, exu­dates, and material
commonly in bottle-fed infants and the from removed intravenous catheters.
aged and debilitated. Creamy white patches B. Direct microscopy: Tissue biopsies, centrifuged
appear on the tongue or buccal mucosa, spinal fluid, and other spec­imens may be examined
that leave a red oozing surface on removal. in Gram-stained smears for pseudohyphae
2. Vulvovaginitis and budding cells. Wet films or Gram-stained
3. Balanitis smears from lesions or exudates show budding
4. Conjunctivitis gram-positive cells. As Candida can be seen on
5. Keratitis. normal skin or mucosa as well, only its abundant
presence is of significance. Demonstration of
b. Skin and Nail Infections mycelial forms indicates coloniza­tion and tissue
i. Intertriginous infection: Intertriginous invasion.
infection occurs in moist, warm parts of
C. Culture: Cultures are obtained on Sabouraud’s
the body such as the axillae, groin, and
dextrose agar (SDA) and on ordinary bacteriological
intergluteal or inflamammary folds.
culture media, e.g. blood agar at room temperature
ii. Interdigital involvement: Inter­d igital
or at 37°C. Colonies are creamy white, smooth
involvement between the fingers follows
and with a yeasty odor. Gram-stained smear from
repeated prolonged immersion in water.
colonies shows gram-positive budding yeast cells
iii. Onychomycosis: Candidal invasion of
(Fig. 75.3).
the nails and around the nail plate causes
onychomycosis. D. Identification: The following tests are done to
iv. Napkin dermatitis in infants. differentiate C. albicans from other species.
B. Systemic candidiasis i. Germ tube test: C. albicans has an ability
1. Intestinal candidosis: It is a frequent sequel to form germ tubes within two hours when
to oral antibiotic therapy and may present as incubated in human serum at 37°C (Reynolds
diarrhea not re­sponding to treatment. Braude phenomenon) (Figs 75.4).
2. Bronchopulmonary candidosis ii. Chlamydospores: Chlamydospores develop
3. Septicemia in a nutritionally deficient medium, such as
4. Endocarditis cornmeal agar at 20°C. They can be seen at
5. Meningitis the end of pseudohyphae (Fig. 75.5).
6. Kidney infections iii. Carbohydrate fermentation and carbohydrate
7. Urinary tract infections assimilation tests.
C. Chronic mucocutaneous candidiasis: Most These are used in identification of C. albicans
forms of this disease have onset in early childhood, and other species of Candida.
are associated with cellular immunodeficiencies E. Serology: The detection of circulating cell wall
and endocrinopathies, and result in chronic mannan, using a latex agglutina­tion test or an
superficial dis­figuring infections of any or all areas enzyme immunoassay, is much more spe­cific.
of skin or mucosa.
F. Skin test: Delayed hypersensitivity to Candida is
Laboratory Tests so universal that skin testing with Candida ex­tracts
Diagnosis can be established by microscopy and is used as an indicator of the functional integrity of
culture. cell-mediated immunity.

Chap-75.indd 520 15-03-2016 11:25:33


Chapter 75: Opportunistic Mycoses  | 521

Fig. 75.4: Candida albicans showing germ tubes Fig. 75.5: Formation of chlamydospores by Candida
albicans when cultured on cornmeal agar at 25°C
Treatment
Management of candidosis is mainly by removing the
predisposing causes. All Candida strains are sensitive
to Nystatin. Ampho­tericin B, 5-fluorocytosine
and clotrimazole may be used for disseminated
candidosis.

Cryptococcosis
Cryptococcosis (torulosis, European blastmycosis,
Busse–Buschke disease) is subacute or chronic
infection caused by the capsulate yeast Cryptococcus
neoformans. It is most frequently recog­nized as a
disease of the central nervous system (CNS), although
the primary site of infection is the lungs. The disease
Fig. 75.6: Cryptococcus neoformans: India ink preparation of
occurs sporadically throughout the world but it is now spinal fluid showing yeast cells surrounded by a large capsule
seen most often in patients with AIDS.
do not appear to become infected, probably
Morphology because of their high body temperature. In addition
In culture, C. neojormans produces a whitish mucoid to patients with AIDS or hematologic malignancies,
colony in 2–3 days. Microscopically, in culture patients being maintained on corticosteroids are
or clinical material, C. neoformans is a spherical highly susceptible to cryptococcosis.
budding yeast (5–10 μm in diameter), surrounded
by a thick polysaccharide capsule (Fig. 75.6). Pathogenesis
Infection is usually acquired by inhalation but may
Serotypes sometimes be through skin or mucosa. The primary
Adsorbed antisera have defined five serotypes pulmonary infection may be asymptomatic or may
(A–D and AD) and three varieties—C. neofor­mans mimic an influenza-like respira­tory infection, often
vargrubii (serotype A), C. neoformans var neofor­ resolving spontaneously. Pulmonary cryptoccoc­
mans (serotype D), and C. neoformans var gattii osisis may lead to a mild pneumonitis. In patients
(serotype B or C). Most infections are caused by C. who are compromised, the yeasts may multiply
neoformans var. neoformans, which is commonly and disseminate to other parts of the body but
found in the excreta of wild and domesti­cated birds preferen­t ially to the central nervous system,
throughout the world. causing cryptococ­cal meningoencephalitis. Other
common sites of dis­semination include the skin,
Epidemiology eye, and prostate gland.
Cryptococcosis is worldwide in distribution.
Bird droppings (particularly pigeon droppings) Cryptococcal meningitis
enrich for the growth of C. neoformans and serve Cryptococcal meningitis is the most serious type
as a reservoir of infection. The organism grows of infection and can resemble tuberculous or other
luxuriantly in pigeon excreta. The birds them­selves chronic types of meningitis.

www.ebook3000.com
Chap-75.indd 521 15-03-2016 11:25:34
522  |  Section 5: Medical Mycology
Other Sites of Dis­semination 3. Produce phenol oxidase—produce black colo­
Although predominantly a disease of the CNS, nies on niger seed agar, bird seed agar and
lesions of the skin, mucosa, viscera and bones may caffeic acid agar.
also occur. Visceral forms simulate tuberculosis 4. Produce disease in mice on intracerebral and
and cancer clinically. Bones and joints may be intraperitoneal inoculation (animal inoculation
involved. Cutaneous crypto­coccosis varies from test positive). Capsulated budding yeast cells can
small ulcers to large granulomas. be demonstrated in the brain of infected mice.

Laboratory Diagnosis Treatment


A. Specimens Combination therapy of amphotericin B and flucyto­
Specimens include spinal fluid, tissue, exudates, sine has been considered the standard treatment
spu­tum, blood and urine. for cryptococcal meningitis. Fluconazole offers
excellent penetration of the central nervous system.
B. Micrscopic Examination
India ink or nigrosine preparation: In unstained, B. FILAMENTOUS FUNGI
wet preparations of CSF mixed with a drop of
India ink or nigrosine, the capsule can be seen Aspergillosis
as a clear halo around the yeast cells (Fig. 75.6). Aspergillus species are ubiquitous saprophytes in
The yeast cells of C. neoformans are round, nature, and aspergillosis occurs worldwide. The
5–10 μm in diameter, and are surrounded by a most important species are A. fumigatus, A. niger,
mucopolysaccharide capsule. A. flavus, A. terreus and A. nidulans.
Tissue sections: For examination of tissue sections,
it is best to use a specific fungal stain such as PAS. Pathogenesis
Alciari blue and mucicarmine which stain the Following inhalation of conidia, atopic individuals
capsular material. often develop severe allergic reactions to the
C. Culture conidial antigens. In im­m unocompromized
On Sabouraud agar (without cycloheximide) patients, the conidia may germinate to produce
cultured at 25–30°C and 37°C, colonies normally hyphae that invade the lungs and other tissues.
appear within 2–3 days. In culture, C. neoformans A. Localized infections
appears as smooth, mucoid, cream colored
colonies. Cultures can be identified by growth at Sinusitis: A. flavus and A. fumigatus.
37°C and detection of urease. Mycotic keratits: A. flavus and A. fumigatus.
Niger seed (bird seed) agar is a differential
medium for presumptive identification of C. Otomycosis: A niger.
neoformans. It produces brown colonies on this B. Systemic aspergillosis
medium within one week when incubated at 1. Pulmonary aspergillosis
30°C. C. neoformans produces phenoloxidase,
a. Allergic asthma: In some atopic individuals,
which oxidises the caffeic acid in the niger seed
de­v elopment of IgE antibodies to the
into melanin.
surface antigens of aspergillus conidia
D. Serological Tests elicits an immediate asthmatic reac­tion
Cryptococcal capsular polysaccharide antigen can upon subsequent exposure.
be detected in CSF and blood by latex aggluti­nation b. Bronchopulmonary as­p ergillosis: The
and ELISA test. A whole-cell agglutination test conidia germinate and hyphae colonize
for serum antibody is positive in less than 50% of the bronchial tree with­out invading the
proven cases of cryptococcal meningitis lung parenchyma. This phenomenon
E. Animal Inoculation Test is characteristic of allergic broncho­
Intracerebral or intraperitoneal inoculation into mice pulmonary as­pergillosis.
leads to a fatal infection in case of C. neoformans. c. Colonizing aspergillosis (aspergilloma):
Capsulated budding yeast cells can be demonstrated Colo­nizing aspergillosis usually develops
in the brain of the infected mice. in preexisting pulmonary cavities, such as
Differentiation of pathogenic (C. neoformans) in tuberculosis or cystic disease. It is also
from other nonpathogenic crypto­cocci referred to as fungus ball. The fungus grows
Pathogenic C. neoformans can be differentia­ted into large ‘balls’ (as­pergilloma). Cases of
from nonpathogenic species by its ability to: as­pergilloma rarely become invasive.
1. Grow at 37°C. 2. Invasive aspergillosis: This form occurs in
2. Hydrolyze urea. severely immunocompromised individuals.

Chap-75.indd 522 15-03-2016 11:25:34


Chapter 75: Opportunistic Mycoses  | 523
Disseminated aspergillosis involving the brain, Mucormycoses (Zygomycosis)
kidney and other organs is a fatal complication. Mucormycosis is an opportunistic mycosis caused
3. Endocarditis by a number of molds classified in the order
4. Paranasal granuloma. Muco­rales of the class Zygomycetes. The leading
pathogens among this group of fungi are species
Laboratory Diagnosis of the genera Rhizopus, Rhizomucor, Ab­s idia,
A. Specimens: Sputum, other respiratory speci­ Cunninghamella and Mucor. These fungi are ubiq­
mens, or lung biopsy tissue provide good specimens. uitous, thermotolerant saprophytes.
B. Microscopic examination: On direct exami­ The conditions that place patients at risk include
nation of sputum with KOH or calcofluor white acidosis—especially that associated with diabetes
or in histologic sections, the fungus appears meIlitus-leukemias, lym­phoma, corticosteroid
as nonpigmented septate mycelium, 3–5 μm treatment, severe burns, immunodeficiencies, and
in diameter, with characteris­t ic dichotomous other debilitating diseases as well as dialysis with
branching and an irregular outline. the iron chelator deferoxamine.
In tissue sections, Aspergillus species are best
seen after staining with PAS or methenamine-silver. Clinical Manifestations
There are a number of different clinical varieties
C. Culture: Aspergillus species grow readily on
of mucormycosis
Sabouraud agar without cycloheximide at 25–37°C.
Colonies appear after 1–2 days. 1. Rhinocerebral mucormycosis: The major
Species are identified according to the clinical form is rhinocerebral mucormy­cosis,
morphology of their coni­dial structures. Asexual a rapidly fulminating infection. It results from
conidia are arranged in chains, carried on elongated germination of the sporan­giospores in the nasal
cells called ‘sterigmata’, borne on the expanded passages and invasion of the hy­phae into the
ends (vesicles) of conidio­phores (Fig. 75.7). blood vessels. The disease can progress rapidly
with invasion of the sinuses, eyes, cranial bones
D. Skin tests: Skin tests with A. fumigatus antigen and brain.
are useful for the diagnosis of allergic aspergillosis. 2. Thoracic mucormycosis: Thoracic mucor­
E. Serological tests: Immunodiffusion, CIE and mycosis follows inhalation of the sporangio­
ELISA are widely used for the detection of antibodies spores with invasion of the lung parenchyma
in the diagnosis of all forms of aspergillosis. and vasculature.
For diagnosis of invasive aspergillosis, antigen 3. Other sites of invasion: Primary cutaneous
detection has also been used successfully by infections have also been reported, but these
techniques such as ELISA and latex agglutination. are extremely rare. Subcutaneous forms of
zygomycosis are less serious.
F. Polymerase chain reaction (PCR)-1: This is
now increasingly used for diagnosis of invasive
aspergillosis.
Laboratory Diagnosis
1. Specimens: Material, such as nasal discharge
or sputum seldom contains much fungal material
and examination of a biopsy is usually necessary
for a firm diagnosis.
2. Microscopy: Direct examina­tion of curetted or
biopsy material in potassium hydroxide (KOH) may
reveal the characteristic broad, aseptate, branched
mycelium and sometimes distorted hyphae.
A B
However, they are seen much more clearly when
stained with methenamine-silver. The hyphae of
these fungi do not stain with PAS.
3. Culture: The fungi are readily isolated on
Sabouraud agar without cycloheximide at 37°C
producing abundant cottony colonies.
4. Identification: Identification is based on the
sporangial structures (Fig. 75.8).
C D i. Mucor shows nonseptate myc­elium without
Figs 75.7A to D: Aspergillus spp. (A) A. fumigatus; (B) A. rhizoids (root-like structures). Sporangiophores,
flavus, (C) A. niger, (D) A. terreus which may be branched, terminate in large

www.ebook3000.com
Chap-75.indd 523 15-03-2016 11:25:35
524  |  Section 5: Medical Mycology

Fig. 75.8: Zygomycetes: (A) Mucor; (B) Rhizopus; (C) Absidia Fig. 75.9: Penicillium

globose spor­a ngia containing numerous C. OTHER FUNGAL AGENTS


spores.
ii. Rhizopus shows nonseptate mycelium with Pneumocystis jiroveci
rhizoids . Unbranched spor­angiophores arise Until recently, P. jeroveci was thought to be a proto­
in groups directly above the rhiz­oids. zoan. Molecular studies indicate that Pneumocystis
iii. Absidia has also rhizoids but sporangiophores carinii is a fungus with a close relationship to
arise from the aerial mycelium in between the ascomycetes. It has yet to be fully embraced by
rhizoids. mycologists.

Morphology
Treatment
P. carinii has three stages:
Treatment consists of aggressive surgical debride­ Trophozoites thin­walled (1–4 μm).
ment, rapid administration of amphotericin B, and Precyst: It is 5–8 μm.
control of the underlying disease.
Cysts: Cysts are thick-walled and spherical (4–6 μm)
and contains 4 to 8 nuclei.
Penicilliosis
Pathogenesis
There are more than 150 known species of the
genus Penicillium. Infections are caused by P. jiroveci is normally a commensal in the lung, spread
Penicillium marnefei. by respiratory droplets. In immunocompetent
individuals, infection is asymp­tomatic. However,
Pathogenesis in inimunocompromised patients, serious life-
threatening pneumonia can develop (Fig. 75.10).
It causes penicillosis, keratitis, otomycosis and Until the AIDS epidemic, human disease was
rarely deep infections. Penicillium marneffei causes confined to inter­stitial plasma cell pneumonia
serious disseminated disease with characteristic in malnourished or premature infants and
papular skin lesions in AIDS patients in South- immunosuppressed patients. Since the early 1980s,
East Asia. Cutaneous lesions and subcutaneous it has remained one of the primary opportunistic
abscesses have been reported. in­fections found in patients with AIDS.
The multiplication of the parasite in the lungs
Identification induces a hyaline or foamy alveolar exudate. In
Fungi belonging to this genus are characterized by stained sections, the exudate filling the alveoli
producing conidiophores at the tips of branching shows a characteristic honeycomb pattern. Chest
sep­tate hyphae, which in turn may produce radiographs may be normal or show a diffuse
secondary structures termed metulae, from which interstitial infiltrate.
flask-shaped structures called phialides bearing
smooth or rough ­shaped conidia are produced in Laboratory Diagnosis
chains, giving the en­tire structure a brush-like or 1. Specimens: Traditionally, diagnosis was made
broom-like appearance (Fig. 75.9). by finding the cyst or trophozoite in tissue

Chap-75.indd 524 15-03-2016 11:25:36


Chapter 75: Opportunistic Mycoses  | 525
Otomycosis
Otomycosis is a fungal infection of the external ear.
It is a very common disease and is usually caused
by species of A. niger, A. fumigatus, Penicillium,
Candida albicans, C. tro­picalis and C. krusei.
The symptoms are itching, pain and deafness.
Secondary bacterial infection, commonly due to
Pseu­domonas and Proteus, causes suppuration.
Diagnosis can be made by demonstration of the
fungi in scrap­ings and by culture.

Keratomycosis (Mycotic Keratitis or


Fig. 75.10: Life cycle of Pneumocystis jiroveci. The parasite Fungal Keratitis)
enters the lung in respiratory droplets and gets attached to
Keratomycosis or mycotic or fungal keratitis is an
alveolar epithelium. It divides by binary fission. Some form
a thick-walled cyst within which sporozoites develop. When invasive fungal infection of the cornea, secondary
mature cyst ruptures, sporozoites are released, which initiate to injury, bacterial infection and treatment with
fresh cycles of infection antibacterial agents and steroids. They occur most
often in hot cli­mates and are caused by common
saprophytic molds.
obtained through open lung biopsy. To establish
the diagnosis of P. jiroveci pneumonia, specimens Causes
of bronchoalveolar lavage, lung biopsy, or It is most frequently caused by A. fumigatus, A.
induced sputum are stained and examined for flavus, A. glaucus and A. niger. In addition, species
the presence of cysts or trophozoites. of Fusar­ium, Curvularia, Candida, Acremonium,
2. Staining: Appropriate stains include Giemsa, Paecilomyces, Penicillium, Alternaria, Fonsecea,
toluidine blue, methenamine silver and Pseu­dallescheria, Drechslera and Aureobasidium
calcofluor white. Cyst wall stains black with may also cause keratomycosis.
methenamine silver staining. With the Giemsa
stain, the organism appears round, and the Pathogenesis
cyst wall is barely visible. Intracystic bodies It usually follows trau­ma. Fungal spores colonize the
are seen around the interior of the organism. injured tissue and initiate an inflammatory reaction
Fluorescent monoclonal antibody staining leading to hypop­yon, ulcer and endophthalmitis.
shows ‘honeycomb’ appearance of the cyst. P. Increased incidence of keratomycosis is due to
jiroveci cannot be cultured. widespread use of cor­ticosteroids.
3. Serology: Complement fixation titres of 1:4
or more is indicative of active disease. Latex Laboratory Diagnosis
agglutination test is also used. Diagnosis can be made by microscopic examination
4. Polymerase chain reaction (PCR): It is a rapid and culture of scrapings taken from the base or edge
method for detection of early infection. of the ulcer. Local application of amphotericin B,
Nystatin and Pimaricin (Natamycin) may be useful.
Treatment
Key Points
Acute cases of pneumocystis pneumonia are treated
with trimethoprim-sulfamethoxazole (TMP-SMZ) ™™ Patients with compromised host defenses, which
are sus­ceptible to ubiquitous fungi, are referred to
or pentamidine isethionate. Prophylaxis can be as opportunistic fungi. Candida albicans, Aspergillus
achieved with daily TMP-SMZ or aerosolized fumigatus, Aspergillus niger, Penicillium sp., Rhizopus
pentamidine. and Mucor are some examples of opportunistic fungi.
Candidiasis
OTHER OPPORTUNIST FUNGI ™™ Candidosis (candidiasis, moniliasis) is an infection of
the skin, mucosa, and rarely of the internal organs,
Almost any fungus may invade a severely immuno­ caused by a yeast-like fungus Candida albicans,
compromized host and infections with many and occasionally by other Candida species. It
common fungi, including Fusarium species, causes (a) Mucocutaneous lesions (Oral thrush); 2.
Trichosporon beigelii and Pseudallescheria boydii Vulvovaginitis, conjunctivitis keratitis; Skin and nail
have been reported. Diagnosis is made by culture infections
™™ Gram-stained smear of the exudates or tissue
of the causative organism from clinical specimens
shows gram­- positive, oval, budding yeast and
and serological tests play little part.

www.ebook3000.com
Chap-75.indd 525 15-03-2016 11:25:37
526  |  Section 5: Medical Mycology

pseudohyphae. Germ tube is a rapid method for e. Opportunistic fungi


identification of C. albicans f. Mucormycosis or zygomycosis
Cryptococcosis g. Pneumocystosis
™™ Cryptococcosis is caused by the capsulate yeast h. Pneumocystis carinii (jiroveci)
Cryotococcus neoformans
™™ C. neoformans causes: Pulmonary cryptococcosis in
immunocompromised hosts. Central nervous system MULTIPLE CHOICE QUESTIONS (MCQs)
(CNS) cryptococcosis. Disseminated nonpulmonary
non-CNS cryptococcosis 1. Which of the following species of Candida is most
™™ Laboratory diagnosis: In unstained, wet preparations frequently responsible for human infections?
of CSF mixed with a drop of India ink or nigrosine, a. Candida albicans b. C. krusei
the capsule can be seen as a clear halo around the c. C. glabrata d. C. stellatoidea
yeast cells 2. Candida albicans can be differentiated from other
™™ A specific fungal stain, such as PAS, Alciari blue species of candida by:
and mucicarmine stain the capsular material of a. Germ tube test
C. neoformans in tissue specimens. The culture of b. Chlamydospore formation on corn meal agar
centrifuged CSF specimens confirms diagnosis of c. Carbohydrate fermentation and assimilation
the condition tests
™™ Cryptococcal capsular polysaccharide antigen can d. All the above
be detected in CSF and blood by latex aggluti­nation 3. Which of the following fungi is a capsulated?
and ELISA test a. Aspergillus fumigatus
Aspergillosis b. Cryptococcus neoformans
™™ Aspergillus species most frequently involved in human
c. Candida albicans
infections are A. fumigatus, A. flovus and A. niger d. None of the above
™™ In immunocompetent hosts, Aspergillus species
may primarily affect the lungs causing four main 4. Cryptococcus neoformans shows all the following
syndromes including allergic bronchopulmonary features except that:
aspergillosis, chronic necrotizing aspergillus a. It assimilates inositol b. It assimilates
pneumonia, aspergilloma and invasive aspergillosis lactose
™™ In immunocompromised host, Aspergillus cause a c. It produces urease d. It produces
disseminated disease melanin
Zygomycosis 5. Which animal is used for pathogenicity test in
™™ Zygomycosis (mucormycosis or phycomycosis) is an Cryptococcus neoformans:
infection caused by saprophytic molds of the class a. Rabbit b. Mice
Zygomycetes (mainly Mucor, Rhizopus and Absidia) c. Guinea pig d. None of the
™™ Zygomycetes can cause rhinocerebral zygomycosis, above
pulmonary zygomycosis and gastrointestinal 6. Differentiation of pathogenic Cryptococcus neofor­
zygomycosis mans from nonpathogenic cryptococci by:
™™ The fungi appear in biopsy material as broad- a. Growth at 37°C
branched hyphae with no cross-walls. They grow as b. Urea hydrolysis
molds on ordinary culture media c. Production of brown colonies on niger seed
Pneumocystis jiroveci agar
™™ Pneumocystis jiroveci, is the causative agent of d. All the above
Pneumocystis carinii pneumonia (PCP). Transmission
of infection occurs by inhalation
7. The diagnosis of allergic bronchopulmonary
™™ PCP is the most common opportunistic infection in
aspergillosis is established by the following tests
HIV-patients except:
™™ Otomycosis is a fungal infection of the external
a. Demonstration of Aspergillus hyphae in the
ear. and is usually caused by species of A. niger, A. sputum
fumigatus, Penicillium, Candida albicans, C. tro­picalis b. Positive skin test for Aspergillus fumigatus
and C. krusei c. Highly increased level of serum IgE
™™ Keratomycosis or mycotic or fungal keratitis: It is d. Positive serology for Aspergillus precipitation or
most frequently caused by A. fumigatus, A. flavus, A. Aspergillus-specific IgG
glaucus and A. niger. 8. Which of the following species of Aspergillus
is most often oportunistic pathogen in human
disease?
IMPORTANT QUESTIONS a. Aspergillus fumigates b. A. niger
1. Describe the morphology, pathogenicity and c. A. flavus d. None of the
laboratory diagnosis of Candida albicans. above
2. Describe the morphology, cultural characters and 9. Which of the following fungi is/are associated with
laboratory diagnosis of Cryptococcus neoformans zygomycosis:
3. Write short Notes on: a. Mucor b. Rhizopus
a. Candidiasis, candidosis or moniliasis c. Absidia d. All the above
b. Cryptococcosis 10. The most useful test for diagnosis of zygomycosis is:
c. Opportunistic systemic mycoses a. Serology b. Fungal culture
d. Aspergillosis c. Histopathology d. Blood cultures

Chap-75.indd 526 15-03-2016 11:25:37


Chapter 75: Opportunistic Mycoses  | 527
11. All the following statements are true for Penicillium 13. Which of the following fungi can cause otomycosis?
marneffei except: a. Aspergillus niger
a. It is a mold b. A. fumigatus
b. It is an important opportunistic pathogen in
AIDS patients c. Penicillium species
c. It causes tuberculosis-like disease in patients d. All the above
with AIDS 14. Keratomycosis may be caused by:
d. Few case reports have also been documented a. A. fumigatus
from India
b. Aspergillus niger
12. The method that is useful to demonstrate a foamy-
appearing eosinophilic material surrounding c. Fusarium
Pneumocystis for diagnosis of PCP is: d. All the above
a. Methenamine silver stain
b. Gram-Weigert stain ANSWERS (MCQs)
c. Cresyl violet stain 1. a; 2. d; 3. b; 4. c; 5. b; 6. d; 7. a; 8. a; 9. d; 10. c; 11. a;
d. Papanicolaou smear 12. d; 13. d; 14. d

www.ebook3000.com
Chap-75.indd 527 15-03-2016 11:25:37
76
Chapter

Mycotoxicosis

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe the following: my­cotoxicosis; aflatoxin
be able to:
∙∙ Describe mycotic poisoning

Many fungi form poisonous substances. Mycotic Other Mycotoxicoses


poi­soning is of two types.
Classic examples of human diseases caused
by Fusarium mycotoxins include alimentary
Mycetism toxic aleukia, Urov or Kashin–Beck disease and
A fungus which is eaten for itself causes toxic Akakabibye (scabby grain intoxication). Two
effects. Mycetism may cause gastrointestinal of these are yellow rice toxicosis in Japan and
disease, dermatitis or death. alimentary toxic aleukia in the former Soviet Union.
There are also other fungi responsible for
mycotoxicosis (Table 76.1).
Mycotoxicosis
Table 76.1  List of some mycotoxins and myco­
The diseases that result from ingestion of food toxin-producing fungi
or feed that contains myco­toxins are known as
mycotoxicoses. Mycotoxin Mycotoxin-producing fungi

A variety of mycotoxins are produced by Aflatoxin A. flavus, A. parsiticus, etc.


mushrooms (e.g. Amanita species), and their Ascladiol A. clavatus
ingestion results in a dose-related disease called Butenolide F. tricinctum, F. nivale, F. equiseti
mycetismus. Ergot alkaloid Claviceps sp.
Fumigatin A. fumigatus
Aflatoxin Chlorine-containing P. islandicum
One of the most potent is aflatoxin, which is peptide
elaborated by Aspergillus flavus and is a frequent Muscarine, etc. Amanita muscaria, etc
contaminant of peanuts, corn, grains and other Ochratoxin A A. ochraceus
foods.
Patulin P. urticae, A. clavatus, P.
Effect of aflatoxin: It is highly toxic to animals and claviforme, P. expansum, A.
giganteus, etc.
birds, and probably to human beings as well. It
can cause hepatomas in ducklings and rats, and its Penicillic acid P. puberculum, P. cyclopium, P.
thomii, etc
possible carcinogenic effect in human beings has
caused great concern. Phalloidine Amanita phalloides
Psilocybine Psilocybe sp.

Ergot Alkaloids Psoralens Sclerotinia sclerotiorum

Ergotoxicosis (ergotism) is due to the toxic alkaloids Rubratoxin B P. rubrum, P. purpurogenum


produced by the fungus Claviceps purpurea, while Scirpenols (nivalenol, F. nivale, F. tricinctum
growing on the fruiting heads of rye. fusarenon)
Chapter 76: Mycotoxicosis  | 529
Psychotropic agents ™™ Psychotropic agents: Toxic metabolites produced by
fungi such as psilocybin and psilocin, as well as the
Toxic metabolites produced by fungi have been semisynthetic derivative lysergic acid diethylamide
used by primitive tribes for religious, magical (LSD).
and social purposes. The hallucinogenic agents
(d-lysergic acid, psilocybin) are produced by the Important Question
Psilocybe species and other fungi. In the 20th
rite short notes on:
W
century, problems involving toxins of fungi were
a. Mycotoxicosis
seen with the recreational use of psychotropic b. Aflatoxin
agents such as psilocybin and psilocin, as well
as the semisynthetic derivative lysergic acid die­ Multiple choice questions (mcqs)
thylamide (LSD). 1. Which mycotoxin is produced by Aspergillus flavus:
a. Aflatoxin
Key Points b. Ergot alkaloids
c. Penicillic acid
™™ Mycotic poi­soning is of two types: d. Muscarine
1. Mycetism 2. Ergot alkaloids are toxins produced by:
2.  My­cotoxicosis a. Fusarium nivale
™™ Aflatoxin is elaborated by Aspergillus flavus and b. Aspergillus flavus
related molds c. Claviceps purpurea
™™ Ergot alkaloids: Ergotoxicosis (ergotism) is due to d. Penicillium rubrum
the toxic alkaloids produced by the fungus Claviceps
purpurea Answers (MCQs)
1. a; 2. c

www.ebook3000.com
Section 6: Miscellaneous

77
Chapter

Normal Microbial Flora of the Human Body

Learning Objectives

After reading and studying this chapter, you should ∙∙ L ist organisms present as normal microbial flora
be able to: in normal flora of upper respiratory tract and
∙∙ Describe the role of normal flora in a human body gastrointeinal tract

Introduction 3. Stimulus for the development of the immune


system: Bacterial colonization of the newborn
The term ‘normal microbial flora’ denotes the infant acts as a powerful stimulus for the
popula­tion of microorganisms that inhabit the development of the immune system.
skin and mucous membranes of healthy normal
4. The microflora of the intestinal tract synthesize
persons. The skin and mucous membranes always
vitamin K and several B vitamins.
harbor a variety of micro-organisms that can be
arranged into two groups: 5. Normal flora may liberate endotoxins, which
may activate alternate complement pathway
and help the defense mechanisms of the body.
1. Resident Flora
The resident flora consists of relatively fixed types B. Harmful Effects of Normal Flora
of microorganisms regularly found in a given area 1. When the organisms are displaced from
at a given age. their normal site in the body to an abnormal
site, e.g. the introduction of the normal
2. Transient Flora skin bacterium, Staphylococcus epidermidis,
The transient flora consists of nonpathogenic into the bloodstream, where it can colonize
or potentially pathogenic microorganisms that catheters and heart valves, resulting in bacterial
inhabit the skin or mucous membranes for hours, endocarditis.
days or weeks. Members of the transient flora are 2. When potential pathogens gain a competitive
gener­ally of little significance so long as the normal advantage due to diminished populations of
resident flora remains intact. harmless competitors, e.g. when normal bowel
flora is depleted by antibiotic therapy leading to
over­growth by the resistant Clostridium difficile,
Role of normal microbial flora which can cause a severe colitis.
The microorganisms that are constantly present on 3. When some harmless, commonly ingested food
body surfaces are commensals. Their presence is substances are converted into carcinogenic
not essential to life. derivatives by bacteria in the colon. A well-
known example is the conversion by bacterial
A. Beneficial Functions of Normal Flora sulfa­tases of the sweetener cyclamate into the
bladder carcinogen cyclo­hexamine.
1. Prevent colonization by pathogens: On 4. Immunocompromized individuals: Normal
mucous membranes and skin. flora can overgrow and become pathogenic
2. Antimicrobial substances production: For when individuals are immunocompromized.
example, colicins, have a harmful effect on 5. Drug resistance: Penicillinase-producing
pathogens. micro­organisms can aggravate infection by
Chapter 77: Normal Microbial Flora of the Human Body  | 531
conferring drug resistance against antibiotics Normal Flora of the Mouth
for example, increase in carriage of antibiotic The mouth contains a plethora of organisms—
resistant staphylococci. pig­mented and nonpigmented micrococci, some
Certain alpha hemolytic streptococci, that of which aerobic, gram-positive aerobic spore-
live as commensals in the mouth may cause bearing bacilli, coliforms, Proteus and lactobacilli.
dental caries. Within 4–12 hours after birth, viridans streptococci
6. Confusion in diagnosis: They cause confusion become established as the most prominent
in diagnosis due to their ubiquitous presence members of the resident flora. Early in life, aerobic
in the body, and their morphological similarity and anerobic staphylococci, gram-negative
with some pathogens. diplococci (neisseriae, Moraxella catarrhalis),
diph­t heroids, and occasional lactobacilli are
Normal Flora of the Skin added. When teeth begin to erupt, the anerobic
The predominant resident microorganisms of spirochetes, pre­v otella species (especially P
the skin are aerobic and anerobic diphtheroid melaninogenica), Fusobac­terium species, Rothia
bacilli (e.g. Corynebacterium, Propionibacterium); species, and Capnocytophaga species establish
nonhemolytic aerobic and anerobic staphylococci themselves, along with some anerobic vibrios and
(Staphylococcus epi­d ermidis, occasionally S. lactobacilli. Yeasts (Candida species) occur in the
aureus, and Peptostreptococcus species); gram- mouth.
positive, aerobic, spore-forming bacilli; alpha­
hemolytic streptococci (viridans streptococci) Normal Flora of the Upper Respiratory
and ente­rococci (Enterococcus species); and gram- Tract
negative col­iform bacilli and Acinetobacter. Within 12 hours after birth, alpha hemolytic strep­
Hair frequently harbors Staph aureus. Often tococci are found in the upper respiratory tract and
the skin of the face, neck, hands and but­tocks be­come the dominant organisms of the oropharynx
carries pathogenic hemolytic streptococci and and remain so for life. Small bronchi and alveoli are
staphylococci. Penicillin-resistant staphylococci normally sterile. In the pharynx and trachea, flora
are seen in individuals working in hospitals. similar to that of the mouth establish themselves.
Among the factors that may be important in
elimi­nating nonresident microorganisms from the Normal Flora of the Gastrointestinal Tract
skin are the low pH, the fatty acids in sebaceous At birth: At birth the intestine is sterile. In all cases,
secretions, and the presence of lysozyme. within 4–24 hours of birth, an intestinal flora is
established partly from below and partly by invasion
Normal Flora of the Conjunctiva from above.
The predominant organisms of the conjunctiva
are diphtheroids (Corynebacterium xerosis.), S. Breastfed children: In breastfed children, the
epidermidis, and nonhemolytic streptococci. intestine con­tains lactobacilli, enterococci, colon
Neisseriae and gram­-negative bacilli resembling bacil­li and staphylococci. In bottlefed children,
hemophili (Moraxella species) are also frequently a more mixed flora exists in the bowel, and.
present. The conjunctival flora is normally held lactobacilli are less prominent. In artificilly fed
in check by the flow of tears, which contain anti­ (bottlefed) chil­dren intertine contains lactabaulli
bacterial lysozyme. enterococci, colon bacilli and staphylococci. With
the change of food to the adult pattern, the flora
Normal Flora of the Nose, Nasopharynx change. Diet has a marked influence on the rela­
and Accessory Sinuses tive composition of the intestinal and fecal flora.
The floor of the nose harbors corynebacteria, Normal adult: In the normal adult, the micro-
staphy­lococci and streptococci. Haemophilus organisms on the sur­face of the esophageal wall
species and Moraxella lacunata may also be seen. are those swallowed with saliva and food. Because
The nasopharynx of the infant is sterile at of the low pH of the stomach, it is virtually sterile
birth but, within 2–3 days after birth, acquires the except soon after eating. The stomach’s acidity
common commensal flora and the pathogenic keeps the number of microorganisms at a mini­
flora. The nasopharynx can be considered the mum (l03–105/g of contents) unless obstruction at
natural habitat of the common path­ogenic bacteria the pylorus favors the proliferation of gram-positive
which cause infections of the nose, throat, bronchi cocci and bacilli.
and lungs. Certain gram-negative or­g anisms As the pH of intestinal contents becomes
from the intestinal tract such as Pseu­domonas alkaline, the resident flora gradually increases. The
aeruginosa, E. coli, paracolons and Proteus are also number of bacteria increases progressively be­yond
occasionally found in normal persons. the duodenum to the colon, being comparatively

www.ebook3000.com
532  |  Section 6: Miscellaneous
low in the small intestine. In the adult duodenum, activity of Doderl­ien’s bacilli, E. coli and yeasts.
there are 103–106 bacteria per gram of contents; in This appears to be an important mechanism in
the jejunum and ileum, 105–108 bacteria per gram; preventing the establish­ment of other, possibly
and in the cecum and transverse colon, 108–1010 harmful, microorganisms in the vagina. During
bacteria per gram. In the upper intestine, lac­tobacilli pregnancy, there is an in­crease in Staphylococcus
and enterococci predominate, but in the lower ileum epidermidis, Doderlien’s bacilli and yeasts.
and cecum, the flora is fecal. In the sigmoid colon Occasionally, other members of the in­testinal
and rectum, there are about 1011 bacteria per gram flora may be present. After menopause, lactobacilli
of contents, constituting 10–30% of the fecal mass. again diminish in number and a mixed flora returns
Aner­obes outnumber facultative organisms by 1000- and the flora resembles that found before puberty.
fold. In diarrhea, the bacterial content may diminish
greatly, whereas, in intestinal stasis, the count rises. Key Points
In the normal adult colon, 96–99% of the resident ™™ The term ‘normal microbial flora’ denotes the
bacterial flora consists of anerobes: Bacteroides population of microorganisms that inhabit the
species, especially B. fragilis; Fusobacterium skin and mucous membrane of normal healthy
species; anerobic lac­tobacilli, e.g. bifidobacteria; individuals
clostridia (C perfringens, 103–105/g); and anerobic ™™ The resident flora consists of relatively fixed types
of micro-organisms regularly found in a given area
gram-positive cocci (Pep­tostreptococcus species).
at a given age. and cannot be removed permanently
Only 1–4% are facultative aer­obes (gram-negative ™™ The transient flora inhabit the skin or mucous
coliform bacteria, enterococci, and small numbers membranes and does not pro­duce disease, and
of Proteus, pseudomonads, lactobacilli, candidae, does not establish itself permanently on the surface.
and other organisms). More than 100 distinct types The normal flora is composed principally of bacteria
of organisms occur regularly in normal fecal flora. ™™ A resident, actively proliferating, microbial flora
occurs in the skin, nose and oropharynx, the mouth
and large intestine, the anterior parts of the urethra
Normal Flora of the Genitourinary Tract and the vagina.
Mycobacterium smegmatis, a harmless commensal,
is found in the smegma of the genitalia of both
men and women. This may, by its presence in the Important questions
voided speci­mens of urine, cause confusion with 1. What is normal microbial flora of the human body?
tubercle bacilli. From apparently normal men, Write briefly on beneficial and harmful effects of
aerobic and anerobic bacteria can be cultured normal flora.
including, lactobacilli, Gard. vaginalis, alpha 2. Write short notes on:
hemolytic streptococci and Bacteroids species. Chlam. a. Normal bacterial flora.
trachomatis and Ureaplasma urealyti­cum may also
b. Difference between resident and transient flora.
be present. The female urethra is either sterile or
c. Normal flora of the skin.
contains a few gram-positive cocci.
d. Normal flora of the intestine.
At birth: At birth, the vagina is sterile. In the e. Normal flora in the mouth and upper respiratory
first 24 hours, it is invaded by micrococci, tract.
enterococci and diphtheroids. In 2–3 days, the f. Normal flora of the genitourinary tract.
maternal estrin induc­es glycogen deposition in
the vaginal epithelium. This facilitates the growth Multiple choice questions (mcqs)
of a Lactobacillus (Doderlien’s bacillus), which
produces acid from glycogen, and the flora for a 1. The majority of normal bacteria flora are present in:
few weeks is similar to that of the adult. After the a. Conjunctiva b. Skin
passively transfected estrin has been eliminated c. Nasopharynx d. Large intestine
2. Which bacteria is responsible for producing acidic
in the urine, the glycogen disappears, along with
pH in adult vagina:
Doderlien’s bacillus and the pH of the vagina
a. Lactobacillus
becomes alkaline. This brings about a change in b. Bacteroides species
the flora to micrococci, alpha and nonhemolytic c. Diphtheroids
streptococci, coliforms and diph­theroids. d. Gardnerella vaginalis
At puberty: At puberty, the glycogen reappears Answers (MCQs)
and the pH changes to acid due to the metabolic 1. d; 2. a
78
Chapter

Infective Syndromes*

Learning Objectives

After reading and studying this chapter, you should ∙∙ List microbial pathogens that cause pneumonia.
be able to: ∙∙ Define diarrhea and dysentery
∙∙ Define bacteremia, septicemia, pyemia and ∙∙ List causative organisms of diarrhea and dysentery
endotoxemia ∙∙ Discuss laboratory diagnosis of dirrhea
∙∙ List causative organisms of septicemia, infective ∙∙ Discuss laboratory diagnosis of dysentery
endo­carditis and subacute endocarditis ∙∙ List causative organisms of food poisoning
∙∙ Discuss laboratory diagnosis of subacute endocarditis ∙∙ Discuss the laboratory diagnosis of food poisoning
∙∙ List causative organisms of meningitis ∙∙ List microbial pathogens that cause pneumonia
∙∙ Discuss the laboratory diagnosis of acute pyogenic ∙∙ List causative organisms of (i) sexually transmitted
meningitis diseases (STDs); (ii) painless genital ulcer; painful
∙∙ Describe characterstics features of cerebrospinal genital ulcer; urethral discharge
fluid (CSF) in different forms of meningitis ∙∙ Discuss the laboratory diagnosis of gonorrhea
∙∙ List causative organisms of urinary tract infection ∙∙ Describe the following; Non-gonococcal urethritis
∙∙ Discuss the laboratory diagnosis of urinary tract (NGU)
infection ∙∙ Discuss the laboratory diagnosis of syphilis
∙∙ Describe characterstics features of cerebrospinal ∙∙ List causative organisms of wound infection
fluid (CSF) in different forms of meningitis ∙∙ Discuss the laboratory diagnosis of wound infection
∙∙ Describe the following: aseptic meningitis; tuber­ ∙∙ List causative organisms of pyrexia of unknown origin
culous meningitis (PUO)
∙∙ List causative organisms of sore throat ∙∙ Discuss the laboratory diagnosis of pyrexia of
∙∙ Discuss the laboratory diagnosis of sore throat unknown origin (PUO)

1. Bacteremia, Septicemia and Infective Endocarditis


Bacteremia and Septicemia 4. Endotoxemia: Endotoxemia is a condition
in which bacterial endotoxin circulates in the
1. Bacteremia: Bacteremia may be defined as blood.
presence of bacte­r ia in blood without any
multiplication. Infective Endocarditis (Table 78.2)
2. Septicemia: Septicemia is a condition in which Infective endocarditis denotes a condition of
bacteria circulate and actively multiply within proliferation of microorganisms on the endothelium
the blood stream. Microorganisms causing of the heart. Damaged endocardium provides a
septicaemia are given in Table 78.1. site for the aggregation of platelets and fibrin is
3. Pyemia: Pyemia is essentially septicemia with deposited to build up a sterile vegetation. This
metastatic infection. is then liable to be colonized by blood-borne

*This chapter was contributed by Dr Savita Kumari, Professor, Department of Internal Medicine, Postgraduate
Institute and Medical Research (PGIMER), Chandigarh-160 012, India.

www.ebook3000.com
534  |  Section 6: Miscellaneous
Table 78.1  Causative organisms of septicemia Table 78.3  Causative agents of subacute endocar­
ditis
A. Gram-negative bacilli (60–70% cases)
Salmonella Typhi (A) Bacteria
S. Paratyphi A Viridans group of streptococci- responsible for 60–80%
S. Paratyphi B of cases?
S. Paratyphi C Streptococcus sanguis
Brucella spp. Str. mutans
Haemophilus influenzae Str. mitis
Escherichia coli Enterococcus faecalis
Klebsiella pneumoniae Staph. epidermidis
Proteus spp. Coxiella burnetii
Enterobacter spp. Chlamydia psittaci
Bacteroides spp. (B) Fungi
Pseudomonas spp. Candida albicans
B. Gram-positive bacilli Aspergillus spp.
Listeria monocytogenes
C. Gram-negative cocci
Neisseria meningitidis vegetations comprising of dense fibrin, platelet
D. Gram-positive cocci (20–40% cases) aggregates with bacterial colonies are formed.
Staphylococcus aureus It runs a chronic course. It is more common
Staph. epidermidis and comprises of almost 70% cases of bacterial
Streptococcus pyogenes
Str. pneumoniae
endocarditis. Causative agents of subacute
bacterial endocarditis are shown in Table 78.3.

Table 78.2  Causative agents of infective endocarditis Pathogenesis


A. Bacteria Endocarditis is an endogenous infection acquired
• Viridans group of streptococci when organisms entering the bloodstream
– Streptococcus sanguis
– S. mutans
establish themselves on the heart valves. About
– S. intermedius 70% patients have one or the other predisposing
– S. mitis cardiac lesion and other abnormalities that
• Groups F and G streptococci . predispose to endocarditis. These include:
• Staphylococcus epidermidis 1. Rheumatic valvular disease
• Enterococcus faecalis
• S. aureus
2. Congenital valve deformities
• Diphtheroids 3. Cardiac valve prosthesis
• Haemophilus spp. 4. Degenerative cardiac disease
• Coliforms 5. Drug abuse.
• Pseudomonas
• Coxiella burnetii
• Chlamydia psittaci Laboratory Diagnosis
B. Fungi Diagnosis of infective endocarditis depends on
• Candida spp. isolation of the causative agent from blood. Blood
• Aspergillus spp.
culture is the most important test for diagnosing
infective endocarditis
organisms. It may occur as an acute rapidly
progressive disease or in a subacute form. 1. Specimen
Three to six samples of blood, 10 mL each should
1. Acute Endocarditis be collected from antecubital vein over 24 hours.
Virulent species such as Staphylococcus aureus and Samples should be collected before antimicrobial
Streptococcus pneumoniae are usually the cause, therapy is administered. Each sample should be
and they infect both normal and abnormal heart inoculated into 50­–100 mL of glucose broth.
valves. They can often produce a rapidly progressive Large amount of blood is required because the
disease, often with valve destruction and formation number of organisms in the blood may be very few.
of abscesses in the heart muscle, lead­ing to heart In addition, blood provides nutrition for the growth
failure. of organisms. Repeated blood cultures are made
because the bacteremia is intermittent.
2. Subacute Endocarditis
Subacute bacterial endocardi­tis is usually caused 2. Cultures
by organisms with little virulence. These organisms Cultures are incubated at 37°C for at least 3 weeks.
of relatively low virulence cause infection on Subcultures are made on solid media such as blood
damaged or defective valve cusps, and large firm agar and MacConkey agar after 24 hours, 48 hours
Chapter 78: Infective Syndromes  | 535
and once a week thereafter. These solid media are These organisms and entero­cocci, which are always
incubated at 37°C for 24 hours. more resistant to penicillin, should be treated with
a combination of penicillin (or ampicillin) and an
3. Identification aminoglycoside.
The isolated organism is identified by colony Staphylococcal endocarditis-A β-lactamase
morphology, Gram staining, biochemical reactions stable penicillin, such as cloxacilIin is often suitable
and serological tests. Refer to the corresponding and may be given in combination with an amino­
chapters for details of these agents. glycoside, rifampicin or fusidic acid. Vancomycin
or teicoplanin should be used for penicilIin-
4. Antibiotic Sensitivity Tests alIergic patients and for treating methicillin-
Minimum inhibitory concentration (MIC) and resistant staphylococci.
minimum bactericidal concentration (MBC) of
the antimicrobial agent for the isolated organism Prevention
must be determined. The measurement of MIC and No scientifically proven preventive methods are
MBC helps to determine the adequate dose of the available.
antibiotic to be used for ensuring the serum levels i. Importance of maintaining good dental hygiene.
that can penetrate the valves and kill the organisms. ii. Prophylactic antibiotic—the accepted practice
is to give prophylactic antibiotic before dental
5. Culture Negative Endocarditis extraction and other surgical procedures.
Blood cultures are persistently negative in about
10-20% cases. It may be due to following reasons: Key Points
i. Recent antibiotic therapy ™™ Bacteremia may be defined as presence of bacte­ria
ii. Inadequate number of specimens in blood without any multiplication
iii. Infection with Coxiella burnetii or Chlamydia ™™ Septicemia is a condition in which bacteria circulate
spp. and acti­vely multiply within the bloodstream
Blood cultures may be negative because ™™ Pyemia is essentially septicemia with metastatic
infection
Coxiella burnetii and Chlamydia spp. cannot
™™ Infective endocarditis denotes a condition of prolifer­
grow on cell-free media.
ation of micro-organisms on the endothelium of
the heart
6. Other Tests for Diagnosis
™™ Acute endocarditis—Virulent species, such as
i. Erythocyte sedimentation rate (ESR): It is Staphylo­coccus aureus and Streptococcus pneumoniae
elevated. are usually the cause
ii. Normocytic normochromic anemia ™™ Subacute endocarditis with viridans group of
iii. Total leukocyte count : Leukocytosis is streptococci-responsible for 60–80% of cases
common. ™™ Diagnosis of infective endocarditis depends on
iv. C-reactive protein (CRP): More reliable than isolation of the causative agent from blood. Blood
culture is the most important test for diagnosing
ESR in assessing progress. infective endocarditis.
v. Proteinuria
vi. Microscopic hematuria: Usually present.
vii. Echocardiography. Important Questions
1. Define bacteremia, septicemia, pyaemia and
Treatment
endotoxemia. Name various organisms causing
The antibiotic treatment regimen for infective septicemia. How will you diagnose it in the
endocarditis depends upon the infecting organism laboratory?
For penicillin-susceptible streptococci, high- 2. Enumerate causative. agents of infective endocar­
dose penicillin is the treatment of choice. Patients ditis. How will you proceed to diagnose it in the
with a good history of penicillin allergy can be laboratory?
treated with clindamycin or with a macrolide. 3. Write short notes on subacute endocarditis.

2. Meningitis
Meningitis is an inflammation of the membranes 1. Purulent meningitis (acute
surrounding the brain and spinal cord. Most cases pyogenic meningitis)
of meningitis fall into one of the two cat­egories:
purulent meningitis and aseptic meningitis. The In purulent meningitis, the CSF is typically turbid
causative agents of these types are given in Table due to the presence of large numbers of leukocytes,
78.4. from 100 to several thousands/mm3, most of which

www.ebook3000.com
536  |  Section 6: Miscellaneous
Table 78.4  Causative agents of purulent and B. Laboratory Examination of CSF for Cells
aseptic meningitis and Microorganisms
Purulent meningitis Aseptic meningitis 1. Naked eye examination: Naked eye examin­
•  Neisseria meningitidis A. Viruses ation is done for the presence of turbidity and
• Streptococcus • Enteroviruses any sign of contamination with blood from the
pneumoniae (echoviruses, polioviruses, puncture wound.
•  Haemophilus coxsackieviruses)
2. Cell count: The leucocytes in the CSF are
influenzae •  Mumps
•  Escherichia coli •  Herpes simplex counted by microscopical observation. The
•  Group B streptococci •  Varicella-zoster relative numbers of polymorphs and lympho­
•  Pseudomonas •  Measles cytes should be noted, and the number of
•  Salmonellae •  Adenoviruses erythrocytes in specimens contaminated with
• Staphylococcus aureus •  Arboviruses
blood (Table 78.5).
•  S. epidermidis B. Spiral bacteria
• Listeria • Syphilis (Treponema A wet film of centrifuged CSF deposit mixed
monocytogenes pallidum) with India ink will, when examined under oil-
•  Klebsiella • Leptospira immersion, demonstrate the characteristic
•  Anaerobic cocci Interrogans serovars capsulate yeast cells of Cryptococcus neofor­
•  Bacteroides Icterohaemorrhagiae and
mans and a wet film examined on a warm
In neonates and canicola
infants C. Other bacteria stage will show the slowly motile trophozoites
E. coli Tuberculosis (Mycobacterium of Acanthamoeba (Hartmanella) or Naegleria.
Group B streptococci tuberculosis)
Pseudomonads, Partially treated with Dilution
salmonellae antibiotics
Staph. aureus Brain abscess
CSF that is clear or only slightly turbid should be
H. influenzae D. Fungi examined undiluted but when the speci­men is
Listeria monocytogenes Cryptococcus neoformans highly turbid and its cell count very high, it may
Streptococcus Candida albicans be necessary to dilute it 1 in 10 or 1 in 100 before
pneumoniae E. Protozoa examination.
Klebsiella spp. •  Acanthameba
•  Naegleria
•  Toxoplasma gondli Gram Film of CSF
F. Noninfective After taking some CSF for the cell count, the
Lymphoma remain­d er should be centrifuged to deposit
Leukemias
Metastatic and primary
any cells and bacteria and a film of the deposit
neoplasms should be stained by Gram’s method. The finding
Collagen-vascular disease of bacterial forms resem­b ling meningococci,
pneumococci, haemophili, co­liform bacilli,
streptococci or listeriae be reported.
The supernatant from the centrifuged CSF should
are polymorphs. The majority of cases are caused be tested for its content of glucose and protein.
by one or other of three bacteria: Meningo­coccus,
Pneumococcus and Haemophilus influenzae. In Culture
neonates and infants, coliform bacilli, group B i. CSF Culture
streptococci and, less commonly, pseudomonads, Immediately after centrifugation of the CSF and the
salmonellae and Listeria monocytogenes may be the removal of some of the deposit for the Gram film, the
cause (Table 78.4). remainder of the deposit should be seeded heavily
on to culture media, blood agar and chocolate agar
Laboratory Diagnosis for incubation in humid air plus 5–10%, CO2 and
A. Collection of Specimens a-tube of cooked-meat broth. A further blood agar
The principal specimen to be examined is of CSF plate should be seeded for incubation for 2–5 days
collected by lumbar puncture under strict aseptic in an anerobic atmosphere with 5-10% CO2.
conditions. Only 3–5 mL of fluid should be collected The cultures should be inspected after overnight
in a fresh sterile screw-capped containers. The incubation. The isolated organisms are identified
specimen must be dispatched to the laboratory as by colony morphology, Gram staining from
quickly as possible, for delay may result in the death colonies, biochemical reactions and/or serological
of delicate pathogens, such as meningococci, and the tests. Tests with appropriate antibiotics should be
disintegration of leukocytes. It should not be kept in a done on the isolate.
refrigerator, which tends to kill H. influenzae. If delay If no growth is apparent after overnight
for a few hours is unavoidable, the specimen is best incubation, the plates should at once be reincubated
kept in an incubator at 37°C. for another day and then again inspected for growth.
Chapter 78: Infective Syndromes  | 537
Table 78.5  Typical CSF findings in different types of meningitis
CSF in
Characteristic Normal CSF Acute pyogenic Tuberculous meningitis Viral meningitis
meningitis
I. Pressure Normal Highly increased Moderately increased Slightly increased
II. Direct examination
A. Cell count
1. Total cell 1–3 1,000–20,000 50–500 10–500
(per cu. mm
2. Predominant Lymphocytes Neutrophils (90–95%) Lymphocytes (90%) Lymphocytes
B. Biochemical analysis
1. Total 30–45 100–600 (Highly 80–120 (moderately 60–80 (slightly
proteins increased) increased) increased)
(mg%)
2. Sugars 40–80 Diminished or absent Diminished (30–50) Normal
(mg%) (10–20)
III. Bacteriological examination
A. Microsocopy
1. Gram Nil Gram-negative cocci, — —
staining Gram-positive cocci,
Gram-negative bacilli
or gram-positive bacilli
may be found depending
upon the causative agent
responsible.
2. Ziehl– Nil Nil Acid-fast bacilli (AFB) —
Neelsen may be found
staining
B. Culture Nil According to the M. tuberculosis may grow Viruses may be
causative agent, specific on L–J media grown on cell
organism may grow on cultures
appropriate media.

If the plate cultures remain free from growth, tests are used for rapid diagnosis of meningitis and
and turbidity develops in the cooked-meat broth, are particularly useful in partially treated patients
the broth should be filmed and subcultured in whom smear and culture may be negative. These
on to blood agar and heated-blood agar plates, are available for N. meningitidis, Str. pneumoniae,
incubated aerobically and anerobically. H. influenzae type b and Group B streptococcus.

ii. Blood Culture Agglutination


Blood culture is particularly useful in meningitis The isolated organisms may be grouped by
due to N. meningitidis, H. influenzae and Str. agglutination with appropriate antisera.
pneumoniae. About 50% of these cases have
positive blood culture. Demonstration of Bacterial Endotoxin
This is especially useful when a patient has been
Biochemical Tests partially treated and culture shows no growth.
The supernate from the centrifuged CSF should Bacterial endotoxin in blood can be detected by
be tested for its content of glucose and protein the limulus lysate test.
(Table 78.5). For detailed identification of different
organisms, refer to the respective chapters.
Antigen Detection
The supernatant part of CSF contains antigen, 2. Aseptic Meningitis
which may be demonstrated by the performance In aseptic meningitis, the CSF is clear or only
of a latex agglutination test or co­agglutination or slightly turbid and contains only moderate
counter-immunoelectrophoresis (CIEP) test. These numbers of leucocytes, e.g. 10–500/mm3, most of

www.ebook3000.com
538  |  Section 6: Miscellaneous
which are lymphocytes, except in the earliest stage. 2. Microscopy
The great majority of cases are due to viruses (viral The cen­trifuged deposit of the CSF should be
meningitis), particularly enteroviruses of the echo, examined in an auramine or Ziehl–Neelsen-stained
coxsackie and polio groups. (Table 78.4). film for acid­fast bacilli.
A few cases with CSF findings resembling
those of viral meningitis are caused by leptospires 3. Culture
(serovars canicola and icterohemorrhagiae), fungi
(Crypto­coccus neoformans or Candida albicans) Centrifuged deposit of CSF is inoculated on Lowen­
and amoebae (Naegleria or Hartmanella) and an stein–Jensen (L–J) media and incubated at 37°C
underlying viral encephalitis may give a moderate for 6–8 weeks. Identification of M. tuberculosis
lymphocytic exu­date in the CSF. depends on colony morphology, Z–N staining from
colonies and biochemical reactions.
It should also be noted that when antibiotic
If facilities are available, some of the CSF should
therapy is started at an early stage in a bacterial
be inoculated into a guinea-pig. As in purulent
meningitis, the CSF findings may be like those of
meningitis, the glucose con­t ent of the CSF is
aseptic meningitis (Tables 78.4 and 78.5).
reduced and the protein content increased.
Laboratory Diagnosis Key Points
CSF is used for: ™™ Meningitis is an inflammation of the membranes
Cell count and biochemical tests, microscopy, surrounding the brain and spinal cord
culture and other tests according to suspected ™™ Most cases of meningitis fall into one of the two cat­
causative agents (viruses, fungi or protozoa). egories: purulent meningitis and aseptic meningitis
™™ Acute pyogenic or purulent meningitis. The majority
of cases are caused by one or the other of three
3. Tuberculous meningitis bacteria: Meningo­c occus, Pneumococcus and
Haemophilus influenzae
The CSF findings often resemble those of aseptic ™™ Laboratory examination: CSF is the specimen of
meningitis, but the cell count is usually slightly choice. Direct microscopy by Gram staining, antigen
higher, e.g. 100–500 leucocytes/mm 3 , mostly detection and culture are key methods in laboratory
lymphocytes, and a veil clot (fibrin web) often diagnosis of acute pyogenic meningitis
develops when the CSF is allowed to stand ™™ Aseptic meningitis of the great majority of cases
are due to viruses (viral meningitis), a few cases are
undisturbed (Table 78.5).
caused by leptospires, fungi and amebae
When a tuberculous infection is suspected, the ™™ Tuberculous meningitis-The CSF findings often
cen­trifuged deposit of the CSF should be examined resemble those of aseptic meningitis
in an auramine or Ziehl–Neelsen-stained film for ™™ When a tuberculous infection is suspected, the
acid­fast bacilli and cultured on one or two slopes CSF should be examined in an auramine or Ziehl–
Neelsen-stained film for acid­fast bacilli and cultured
of Lowenstein–Jensen medium.
Lowenstein–Jensen medium.

Laboratory Diagnosis
Important Questions
1. Specimen
1. Name the various organisms causing meningitis.
CSF is collected by lumbar puncture in a sterile
Discuss the laboratory diagnosis of acute pyogenic
container under aseptic conditions. When CSF meningitis.
is allowed to stand, a fibrin web (cobweb) often 2. Write short notes on:
develops. Cell count and biochemical analysis can a. Aseptic meningitis
be done as described earlier. b. Tuberculous meningitis.

3. Urinary Tract Infection


Urinary tract infection (UTI) may be defined as the 1. Lower UTI
presence of bacteria undergoing multiplication
i. Urethritis
in urine within the urinary drainage system.
ii. Cystitis
Urinary tract infection is the second most common
infection after respiratory tract for bacterial iii. Prostatitis
infection. Microorganisms causing UTI are shown Lower UTI is due to ascending infection.
in Table 78.6.
2. Upper UTI
Types of UTI i. Acute pyelitis—infection of pelvis of kidney
Acute infections of UTI can be subdivided into two ii. Acute pyelonephritis—infection of parenchyma
anatomic categories: of kidney.
Chapter 78: Infective Syndromes  | 539
Table 78.6  Causes of urinary tract infection In males: From male patients, a midstream
specimen of urine (MSU) is collected. Before
A. Gram-negative Bacilli
collecting a sample, retract the prepuce,
A. Gram-negative bacilli are by far the most common
infecting agents. clean it with sterile normal saline and collect
1. Escherichia coli causes approximately 80% of acute midstream specimen.
infections in patients without catheters In females: In case of females, anogenital
2.  Proteus mirabilis toilet is more important. Wash perineum and
3. Klebsiella periurethral area with soap and water. Then
4.  Enterobacter (occasionally) clean with nonirritant antiseptic, such as
5. Serratia
chlorhexidine. Separate apart labia with fingers
6.  Pseudomonas aeru­ginosa
B. Gram-positive cocci—lesser role in UTIs
of one hand and collect midstream urine.
1.  Staphylo­coccus saprophyticus (10–15%), 2. Catheter specimen of urine (CSU): Catheter
2.  S. epidermidis (1–5%), specimen of urine (CSU) was com­m only
3.  S. aureus (1–5%), collected in the past from females, but
4.  Enterococcus spp. (1­5%) catheterization for this purpose is no longer
C. Other organisms which may occa­sionally cause UTI considered justifiable because it carries a 2–6%
1.  Mycobacterium tuber­culosis risk of introducing and initiating infection.
2. Enterobacter
3. Suprapubic stab: from children and young
3. Citrobacter
4.  Salmonellae
infants.
5.  Streptococcus pyogenes 4. Noninvasive method: A noninvasive method
6.  S. agalactiae of stimulating urine flow in a baby is by tapping
7.  Gard­nerella vaginalis just above the pubis with two fingers at 1 hour
D. Fungus after a feed.
Candida albicans may cause UTI in diabetics and 5. Early morning urine (EMU): If tuberculosis of
immunocompromized patients the urinary tract is suspected.
6. Initial flow of urine: In the investigation of
Pyelonephritis is probably due to hematogenous urethritis and prostatitis.
infection.
B. Transport of Specimen
Predisposing Factors
1. Gender and sexual activity: The females are Once collected, a specimen of urine must be
more frequently affected by UTI. transported to the laboratory with­out delay, for
Sexual intercourse causes the introduction of urine is an excellent culture medium. If a delay
bac­teria into the bladder. of more than 1–2 hours storage is done in a
2. Pregnancy: Pregnancy UTI is are detected in refrigerator at 4°C.
2– 8% of pregnant women.
3. Obstruction: Any impediment to the free flow C. Laboratory Methods
of urine—tumor, stricture, stone, or prostatic Part of the specimen is used for bacteriological
hypertrophy-results in hydronephrosis and a culture and the rest is examined immediately
greatly increased frequency of UTI. under the microscope.
4. Neurogenic bladder dysfunction.
5. Vesicoureteral reflux. 1. Microscopy
6. Genetic factors. The deposit of the centrifuged urine can be examined
under microscope to find out the presence of pus
Clinical Features cells, red blood cells and bacteria in it. Presence
1. Asymptomatic bacteuria: Symptomless of more than 3 pus cells per high- power field is
urinary tract infection is not uncommon and suggestive of infection.
can be detected only by urine culture. Nowadays centrifugation is not recommended.
2. Symptomatic: The common symptoms of Wet film examination: The finding of 1 leucocyte
urinary tract infection are urgency and frequency per 7 high-power fields corresponds with 10 4
of micturition, with asso­ciated discomfort or leukocytes per mL and the larger number than
pain. this indicates significant pyuria. Therefore,
centrifugation is not recommen­ded.
Laboratory Diagnosis
A. Specimen Collection 2. Culture
1. Midstream specimen of urine (MSU): Speci­ A. Quantitative
mens of urine are generally collected in plastic It is too laborious for routine use and thus not used
universal containers. routinely.

www.ebook3000.com
540  |  Section 6: Miscellaneous
B. Semiquantitative methods iii. Triphenyltetrazolium chloride (TTC) test:
These are quicker mehods: It is ­based on the production of a pink-red
precipitate in the reagent caused by the
I. Standard loop Method respiratory activity of the growing bacteria.
Measured quantity of uncentricuged urine with iv. Glucose test paper: It is based on the
the help of standardized loop is inoculated on utilisation of the minute amounts of glucose
blood agar and MacConkey media and incubated present in normal urine, by bac­teria causing
overnight at 37°C. The number of colonies is the infection. Hence, it indicates bacteriuria.
counted to get the bacterial count per mL of urine. v. Polymorphonuclear neutrophils (PMNs):
PMNs are counted in uncentrifuged urine
Interpretation of Results specimen with the help of hemacytometer. 8
Kass (1957) gave a criterion of active bacterial PMN/mm3 is indicative of infection.
infection of urinary tract as follows: vi. Leukocyte esterase: Presence of this enzyme
i. Count more than 10 5 bacteria of single in urine is indication of bacteriuria.
species per mL: significant bacteriuria which vii. Gram staining: Presence of at least one
indicates active UTI. bacteria per oil immersion field (examining 20
ii. Less than 104 organisms/mL and usually less fields) correlates with significant bacteriuria
than 103/mL accounts contamina­tion. (>105 bacteria/mL).
viii. Dip-slide culture methods: Agar-coated
Identification and Sensitivity Tests slides are immersed in urine or even exposed
If similar colonies are found in numbers suggesting to the stream of urine during voiding,
significant bacteriuria, a separate colony or a incubated and the growth estimated by colony
portion of apparently pure growth should be counting or by color change of indicators.
subcultured for identification and testing of its None of the screening methods is as sensitive
sensitivity to antibiotics. or reliable as a culture.

II. Filter Paper Method Differentiation of Upper UTI and Lower UTI
This method of semiquantitative culture is rapid The antibody-coated bacteria test has been
and very economical in the use of culture medium. employed for the localization of the site of urinary
infection. This is based on the assumption that
bac­teria coated with specific antibodies are present
III. Dip-slide Method
in the urine only when the kidneys are infected
The dip-slide is small plastic tray carrying a layer (upper UTI) and not when the infection is confined
of an appropriate agar culture medium. Opposite to the bladder (lower UTI). Anti­b ody-coated
sides of the tray may carry different media, e.g. bacteria are detected by immunofluo­rescence
cysteine lactose electrolyte-deficient (CLED) agar using fluorescent-tagged antihuman globulin or
medium on one side and MacConkey, brainheart by staphylococcal coagglutination.
infusion or pseudomonas selective agar on
the other. The dip-slide is withdrawn from the Tuberculosis of Kidney and Urinary Tract
container and briefly immersed in the urine. It is Tuberculosis of kidney is a blood-borne infection.
then incubated at 37°C overnight and examined The patient presents with frequency and painless
for a growth of colonies. This method is relatively hematuria and routine urine culture does not show
expensive. any pathogen. Tuberculosis must be considered in
cases where pyuria is present without bacteriuria.
Screening Techniques
Laboratory Diagnosis
Several screening techniques have been introduced
for the presumptive diagnosis of significant 1. Specimen
bacteriuria. These include the following: Excretion of M. tuberculosis from kidney is
i. Griess nitrite test: It is based on nitrite- intermittent. Hence, midstream urine specimen
reducing enzymes produced by bacteria is not useful. Early morning urine specimens
present in urine. The presence of nitrite, should be collected in sterile container on three
detectable by a simple test, indicates the consecutive days.
presence of nitrite-reducing bacteria in urine.
ii. Catalase test: The pres­ence of catalase as 2. Direct Ziehl–Neelsen Staining
evidenced by frothing on addi­tion of hydrogen Smear made from centrifuged deposit of urine
peroxide indicates bacteriuria, though a is stained with Ziehl–Neelsen staining and may
positive result is obtained also in hematu­ria. reveal acid-fast bacilli. Saprophytic mycobacteria
Chapter 78: Infective Syndromes  | 541
(e.g. M. smegmatis) may be present in normal urine
™™ Laboratory diagnosis: For specimen collection,
which may be excluded by using acid-alcohol various methods used are: 1. Midstream specimen
as decolorizing agent in staining procedure. M. of urine (MSU); 2. Catheter specimen of urine (CSU);
tuberculosis is acid-alcohol-fast while M. smegmatis 3. Suprapubic stab; 4. Noninvasive method; 5. Early
is only acid-fast. morning urine (EMU); 6. Initial flow of urine
™™ Part of the specimen is used for bacteriological
culture and the rest is examined immediately under
3. Culture the microscope
Culture is performed on Lowenstein–Jensen ™™ Culture: Semi-quantitative methods (Standard loop
medium and incubated for 6–8 weeks. Growth technique) are used for culture
is identified by Ziehl–Neelsen staining and ™™ Count more than 105 bacteria of single species per ml
is called significant bacteriuria. It indicates active UTI
biochemical tests. ™™ None of the screening methods is as sensitive or
reliable as a culture.
Key Points
™™ Urinary tract infection (UTI) may be defined as the
presence of bacteria undergoing multiplication in Important Questions
urine within the urinary drainage system
1. Name the various organisms causing urinary tract
™™ Lower UTI includes) Urethritis, cystitis and prostatitis infection. Discuss the laboratory diagnosis of this
and due to ascending infection condition.
™™ Upper UTI includes acute pyelitis and acute pyelone­
2. Write short notes on:
phritis.
Midstream specimen of urine (MSU).

4. Sore throat
Sore throat is essentially an acute tonsillitis Viral pharyngitis or other causes of pharyngitis/
or pharyngitis. It is characterized by redness tonsillitis must be differentiated from that caused
and edema of mucosa, exudation of tonsils, by Streptococcus pyogenes since it is treatable with
pseudomembrane formation, edema of uvula, penicillin whereas viral infections are not.
grey coating of tongue and enlar­gement of cervical
lymph nodes. Causative agents of sore throat are A. Specimen
given in Table 78.7. For bacteriological sampling, a plain, albumen-
Pseudomembrane formation: Corynebacterium coated or charcoal-coated cotton-wool swab should
diphtheriae, Can­dida albicans, β-hemolytic group A be used to collect as much exudate as possible from
Streptococ­cus, Treponema vincentii and Leptotrichia the tonsils, posterior pha­ryngeal wall and any other
buc­calis may lead to pseudomembrane formation. area that is inflamed or bears exudate. Two throat
swabs are collected. If it cannot be delivered to the
Laboratory Diagnosis laboratory within about 1 hour, it should be placed
in a refrigerator at 4°C until delivery.
The signs and symptoms of sore throat (pharyngitis)
caused by streptococci and viruses are similar.
B. Direct Microscopy
From one swab, make two smears for Gram
Table 78.7  Causative agents of sore throat staining and Albert-staining.
A. Bacteria 1. Gram Staining
• Streptococcus β-hemolytic group A and occasionally
groups C and G
It is not helpful unless Vincent’s organisms or
•  Corynebacterium diphtheriae
Candida albicans are suspected. Vincent’s infection
•  Haemophilus influenzae
shows gram-negative spirochaetes (Borrelia
•  Bordetella pertussis
vincentii) and gram-negative fusiform bacilli
•  Neisseria gonorrheae
(Fusobacterium spp.). When Candida albicans is
•  Treponema vincentii
suspected, it appears as gram-positive oval budding
•  Leptotrichia buccalis
yeast cells.
B. Fungi 2. Albert Staining
•  Candida albicans It shows green-colored, V and L-shaped (Chinese
C. Viruses letter pattern) bacilli with bluish-black metachromatic
•  Epstein–Barr virus granules in infection due to C. diphtheriae. Albert-
•  Adenoviruses staining is helpful for presumptive diagnosis of C.
•  Coxsackievirus A diphtheriae.

www.ebook3000.com
542  |  Section 6: Miscellaneous
C. Culture Elek’s gel precipitation test .
Culture media are selected according to the Animal inculation test
organism suspected to be the causative agent iii. For Candida albicans
of sore throat. Following media may be used for a. Germ tube test
culture. b. Carbohydrate fermentation and assimil­
ation tests.
Blood agar: All the organisms will grow on this iv. For other causative agents
medium. Special culture media and different biochemical
reactions or serological tests may be required
Crystal violet blood agar: It is selective for Str.
as described in respective chapters. For
pyogenes especially when incubated anaerobically.
details of tests mentioned above, refer to the
Loeffler’s serum slope: For isolation of C. corresponding chapters.
diphtheriae, grow very rapidly (in 6–8 hours).
Potassium tellurite blood agar: Selective media E. Antibiotic Sensitivity
for isolation of C. diphtheriae. All β-hemolytic group A streptococci are sensitive to
Sabouraud’s dextrose agar (SDA): When penicillin G, and most are sensitive to erythromycin.
suspecting Candida albicans, SDA should be C. diphtheriae is sensitive to penicillin.
included.
These culture media should be incubated at 37°C Pneumonia
for overnight. In case of potassium tellurite blood
agar, it should be incubated for 48 hours. After 6–8 Pneumonia may be defined as inflammation and
hours, a subculture should be made from Loeffier’s consolidation of the lung substance. Bacterial
serum slope onto potassium tellurite blood agar, causes for pneumonia are listed in Table 78.8.
which is then incubated at 37°C for 48 hours.
Table 78.8  Microbial pathogens that cause pneu­
D. Identification monia
1. Morphology A. Community-acquired pneumonia
i. Str. pyogenes: Colnies are small (pin-point), Common organisms
circular, semitransparent, low convex discs Streptococcus pneumoniae
having β-hemolysis. Chlamydophila pneumoniae
ii. C. diphtheriae: On potassium tellurite blood Mycoplasma pneumoniae
agar, gray or black coloured round colonies Legionella pneumophila
Uncommon organisms
are seen.
Haemophilus influenzae
iii. Candida albicans: White or cream colored
Staphylococcus aureus
colonies may be seen.
Chlamydophila psittaci
Coxiella burnetii
2. Gram Staining or Albert Staining Kleb. pneumoniae
i. Gram Staining Actinomyces israillae
Primary viral pneumonia
∙∙ Small Gram: Positive spherical cocci occurring Influenza, parainfluenza virus and measles
in chains is characteristic of Str. pyogenes. Respiratory syncytial virus in infancy
∙∙ Candida albicans reveals gram-positive budding Varicella (chickenpox)
yeast cells. B. Hospital-acquired pneumonia
C. Diphtheriae is seen as gram-positive bacilli. Gram-negative bacilli (60%)
Escherichia, Pseudomonas, Klebsiella species
ii. Albert Staining Gram-positive organisms (16%)
Staph. aureus
Diphtheriae shows green-colored V-or L-shaped
Anaerobes
bacilli with bluish-black metachromatic granules.
Legionella spp.
C. Pneumonia in immuno­compromised patients
3. Other Tests for Confirmation Pnemocystis carinii
i. For β-hemolytic streptococci Gram-negative bacteria—Ps. aeruginosa
a. Bacitracin sensitivity test—for Str. pyogenes Mycobacteruim tuberculosis
b. Lancefield grouping—for all β-hemolytic Streptococcus pneumoniae
streptococci Haemophilus influenzae
ii. For C. Diphtheriae Candida albicans
Aspergillus fumigatus
a. Biochemical tests
Viruses–Cytomegalovirus
b. Toxigenicity tests
Chapter 78: Infective Syndromes  | 543
1. Classification 3. Giemsa staining: Giemsa stain of sputum.
is useful to detect cysts and trophozoites of
A. Community-acquired Pneumonia Pneumocystis carinii.
Community-acquired pneumonia has been thought
to present as either of the two syndromes—typical C. Culture
or atypical. Sputum, blood and pleural fluid can be cultured
on blood agar and chocolate agar. Purulent portion
Typical Pneumonia Syndrome of sputum is best for culture. If the sputum is too
Typical pneumonia syndrome is usually caused viscid, it may be homogenized.
by the most bacterial pathogen in community- 1. Blood agar: It is incubated aerobically at 37°C
acquired pneumonia, Streptococcus pneumoniae, under 5–10% CO2.
but can also be due to other bacterial pathogens. 2. Chocolate agar: It is also incubated at 37°C
with 5–10 percent CO2.
Atypical Pneumonia Syndrome 3. Lowenstein–Jensen (L–J) medium: Three
Atypical pneumonia is classically produced by specimens of sputum are collected on three
Mycoplasma pneumoniae, Legionella pneumophila. successive days for culture on L–J media which
Chlamydophila pneumoniae. There is patchy are then incubated at 37°C for 6–8 weeks.
consoli­dation of lungs. 4. Selective media are required for culture of L.
pneumophila (see Chapter 37).
Lobar Pneumonia 5. Isolation of chlamydia can be done on cell-lines.
It is an acute inflammation characterized by homo­
geneous consolidation of one or more lobes. It is D. Detection of Bacterial Antigens
caused by Streptococcus pneumoniae.
Direct Immunofluorescence Test
Bronchopneumonia Direct immunofluorescence examination of sputum
It is almost always a secondary infection and is done to detect antigens in L. pneumophila.
generally follows viral infections of the respiratory
tract. It is an acute inflammation of bronchi and E. Serology
the consolidation is scattered. It is caused by 1. Mycoplasma pneumoniae
Streptococcus pneumoniae Haemophilus influenzae –– Complement fixation test (CFT)
(rarely Kleb. pneumoniae, Staph. aureus). –– Cold agglutinin test
Respiratory syncytial virus, Mycoplasma 2. Chlamydia pneumoniae
pneumoniae, Chlamydophila pneumoniae and
–– Microimmunofluorescence
Bordetella pertussis cause bronchitis and bronchiolitis.
–– TWAR antigen
B. Hospital-acquired Pneumonia –– CFT
–– Immunofluorescent antibody test
Hospital-acquired or nosocomial pneumonia
4. Coxiella burnetii
refers to a new episode of pneumonia occurring
–– CFT
at least two days after admission in the hospital.
Key Points
C. Pneumonia in lmmuno­compromized
™™ Sore throat is essentially an acute tonsillitis or
Patients pharyngitis
The common causative agents include Pneumocystis ™™ Laboratory diagnosis of sore throat caused by
carinii, Staph. aureus, Ps. aeruginosa, viral bacteria depends on direct microscopy and culture.
infections (CMV and herpes) and M. tuberculosis. For bacteriological sampling, two throat swabs are
collected
™™ Pneumonia may be defined as inflammation and
Laboratory Diagnosis consolidation of the lung substance, which may
A. Specimen be classified as community-acquired pneumonia,
hospital-acquired pneumonia and pneumonia in
Sputum; blood; pleural fluid; blood for serological immuno­compromized patients
tests. ™™ Laboratory diagnosis of pneumonia depends on
direct microscopy and culture.
B. Direct Microscopy
1. Gram Staining: Adequate number of pus Important Questions
cells alongwith the presence of predominant 1. Name the various organisms causing sore throat.
organisms gives a clue to the probable pathogen. How will you diagnose it in the laboratory?
2. Ziehl–Neelsen staining: Presence of acid-fast 2. Name the various bacterial causes of pneumonia.
bacilli (AFB) gives a presumptive diagnosis of Discuss the laboratory diagnosis of pneumococcal
tuberculosis. pneumonia.

www.ebook3000.com
544  |  Section 6: Miscellaneous

5. Diarrhea and Dysentery


A. Diarrhea Table 78.9  Causative agents of infective diarrhea
Diarrhea is defined as the passage of loose, liquid A. Bacteria C. Protozoa
or watery stools. These liquid stools are usually •  Vibrio cholerae •  Entamoeba histolytica
passed more than three times a day. Infective •  V. parahaemolyticus •  Giardia lamblia
diarrhoea may be caused by viruses, bacteria, • Escherichia coli (ETEC, •  Cryptosporidium parvum
EPEC) •  Isospora belli
protozoa and Fungi (Table 78.9). •  Salmonella enteritidis D. Cestodes
•  S. Typhimurium •  Hymenolepis nana
B. Gastroenteritis •  Other Salmonella spp. E. Nematodes
Gastroenteritis may be defined as inflammation of •  Campylobacter spp. •  Trichuris trichiura
the mucous membrane of stomach and intestine •  Yersinia enterocolitica •  Strongyloides stercoralis
•  Shigella spp. •  Ascaris lumbricoides
resulting in frequent loose motions with or without • Clostridium •  Hookworms
mucus and with or without blood, pain abdomen perfringens F. Trematodes
and with or without fever. It is often used as a •  C. difficile Schistosoma mansoni
synonym for acute diarrhea, especially when • Staphylococcus
associated with vomiting. aureus
•  Bacillus cereus
C. Dysentery • Aeromonas
hydrophila
Dysentery is a disease marked by frequent • Plesiomonas
watery stools, often with blood and mucus, and shigelloid es
characterized clinically by cramping abdominal B. Viruses
•  Rotavirus
pain, tenesmus, fever and dehydration. •  Astrovirus
For all practical purposes, the terms diarrhea, •  Calicivirus
gastroenteritis and dysentery are collectively •  Norwalk virus
included as diarrheal diseases. •  Adenovirus

D. Traveller’s Diarrhea iii. EIEC


Persons from the developed countries visiting iv. EAEC
endemic areas may acquire various exotic intestinal a. Specimen: Feces
pathogens and cause diarrheal illness soon after b. Culture: On blood agar and MacConkey’s agar.
the traveller has returned by air—a condition These media are incubated at 37°C for 24 hours.
known as “Traveller’s diarrhea”. c. Identification
Colony morphology, biochemical reactions, slide
Diarrhea agglutination with antisera.
Causative agents of diarrhea are shown in Table Sereny test is used for the identification of
78.9. EIEC strains. Invasion of cultured HeLa cells is
another method for identification of these strains.
Laboratory Diagnosis Production of verocytotoxin (VT) is confirmed by
A. Bacteria testing the strains on Vero cells, in which they cause
cytopathic effects.
1. Vibrios
i. Vibrio cholerae 3. Salmonellae
ii. Vibrio parahaemolyticus S. Typhimurium, S. Enteritidis, S. Dublin, S.
iii. Other halophilic vibrios Cholerae-suis, S. Heidelberg and S. Thompson.
V. mimicus and V. vulnificus cause sporadic
cases of diarrheal disease. Laboratory diagnosis
a. Specimen: Feces i. Specimen: Feces or any suspected foodstuff.
b. Culture: Selective media, such as TCBS or bile ii. Culture: Specimen is inoculated in enrichment
salt agar are used. Plates are incubated at 37°C medium and on the selective media, such as
for 24–48 hours. Vibrio parahaemolyticus is a MacConkey’s agar, deoxycholate agar (DCA)
halophilic vibrio, in media containing sodium and XLD agar. Wilson and Blair’s medium may
chloride. also be used.
c. Identification: Colony morphology, biochemical Selective media are incubated at 37°C for 24
reactions and slide agglutination test. hours. After 6 hours incubation of enrichment
2. E. coli medium, subculture is made onto the selective
i. ETEC medium. Pale nonlactose fermenting colonies
ii. EPEC develop on MacConkey’s agar and DCA or
Chapter 78: Infective Syndromes  | 545
black shiny colonies on Wilson and Blair’s following ingestion of contaminated food (meat,
medium. On XLD medium, colonies are red poultry products). Most cases of human clostridial
in color. gastro­enteritis are caused by type A strains. The
iii. Identification: Colony morphology and incubation period is 6–12 hours. Type A strains
biochemical reactions. produce a heat-labile enterotoxin which acts on
The species is identified by agglutination test the membrane permeability of the small intestine.
with polyvalent antisera using that of H and O. Enterotoxin production has also been reported for
type C and type D strains.
4. Shigellae
All four serogroups of shigellae (Shigella dysenteriae, Laboratory Diagnosis
S. fiexneri, S. boydii and S. sonnei) cause bacillary Since Cl. perfringens type A forms a part of normal
dysentery. flora in 5–30% of the population, it is difficult to
interpret its causative role. When the same serotype
Laboratory Diagnosis is isolated from large number of the victims of
i. Specimens: Feces an outbreak and from the suspected food, a
ii. Culture: on MacConkey’s agar and DCA presumptive diagnosis can be made.
medium. S. sonnei is a late lactose i. Specimens: Feces, food
iii. Identification ii. Culture: Culture is done on blood agar and
It depends on Colony morphology, biochemical incubated anaerobically at 37°C for 24 hours.
reactions and agglutination test by group specific iii. Identification: Colonies are either beta-
polyvalent antisera. hemolytic or non­hemolytic.
Gram staining: Gram-positive bacillus with
5. Campylobacter jejuni subterminal spore.
C. jejuni occurs in intestinal flora of many animals, Further identified by Naegler reaction and
especially poultry which probably serves as serotyped by agglutination test.
the major source of human infection. Milk and
waterborne outbreaks of diarrhea have been 8. Clostridium difficile
reported. Incubation period varies from 3 to 10 Cl. difficile is associated with antibiotic­-associated
days. The disease is sometimes associated with diarrhea and pseudomem.
vomiting and bloody mucoid stools.
Laboratory Diagnosis
Laboratory Diagnosis i. Specimen: Feces
i. Specimen: Feces ii. Culture: Feces is cultured on selective media
ii. Culture with subsequent toxigenicity test.
Selective medium containing vancomycin and iii. Demonstration of toxin:
polymyxin. The inoculated culture medium is Toxin-characteristic effects on HEp-­2 and
Incubated at 43°C under microaerophilic human diploid cell cultures, or by ELISA.
conditions. C. jejuni and C. coli selectively Toxin is specifically neutralized by antiserum.
grow against other faecal bacteria.
iii. Identification 9. Staphylococcus aureus
Curved, Gram-negative bacilli exhibiting Laboratory Diagnosis
darting motility oxidase positive. i. Specimens: Vomit, feces, suspected food.
ii. Culture: On selective medium (mannitol salt
6. Yersinia enterocolitica agar) or on ordinary media.
Y. enterocolitica has been identified as an important iii. Identification:
cause of diarrhea. Sources of infection are birds and Colony morphology, Gram staining, catalase
animals. Foodborne outbreaks have been reported. test and coagulase test. Phage-typing may be
done for epidemiological purposes.
Laboratory Diagnosis iv. Demonstration of enterotoxin: Reverse passive
latex agglutination assay (RPLA) or ELISA.
i. Specimens: Feces, blood
ii. Culture: Isolated from faeces or blood 10. Bacillus cereus
cultures. Non-lactose fermenting (NLF). Laboratory Diagnosis
i. Specimen: Vomit, feces, suspected food
7. Clostridium perfringens ii. Culture: Ordinary media (nutrient agar or
Cl. perfringens gastroenteritis or food poisoning blood agar) and incubated at 37°C for 24
is characterised by diarrhea and abdominal pain hours. A special mannitol-egg yolk-phenol

www.ebook3000.com
546  |  Section 6: Miscellaneous
red-polymyxin agar (MYPA) medium may also D. Fungus
be used. There have been reports of diarrhea associated
iii. Identification: Colonies have curled hair with Candida albicans.
appearance. Gram-staining shows gram-
positive spore bearing bacilli. Dysentery
Dysentery is a disease marked by frequent
11. Other Bacteria watery stools, often with blood and mucus, and
Aeromonas hydrophila and Plesiomonas shigelloides characterized clinically by cramping abdominal
have been reported to cause diarrheal diseases. pain, tenesmus (painful straining when passing the
stools), fever and dehydration. Dysentery results
from ‘enteroinvasive’ microorganisms (Table 78.10).
B. Viruses
that penetrate through the mucosa and cause
1. Rotavirus inflammation of the intestinal wall. The differences
2. Norwalk virus between proto­zoal (amebic) and bacterial (bacillary)
3. Adenoviruses dysentery are given in Table 78.10. For laboratory
4. Other viruses: diagnosis, see under “Diarrhea”.
Astroviruses and caliciviruses. Table 78.10  Microorganisms causing dysentery
A. Bacteria
Laboratory Diagnosis •  Shigella dysenteriae
•  S. fIexneri
i. Specimen: Feces •  S. boydii
ii. Electron microscopy •  S. sonnei
iii. Fluorescent antibody test and ELISA. •  Escherichia coli (EIEC, EPEC, EHEC)
B. Protozoa
•  Entamoeba histolytica
C. Protozoa •  Balantidium coli

1. Entamoeba histolytica
Key Points
2. Giardia lamblia
3. Cryptosporidium parvum ™™ Diarrhea is defined as the passage of loose, liquid or
watery stools. These liquid stools are usually passed
4. Balantidium coli: This is a rare cause of chronic more than three times a day
recurrent diarrhea, with dysenteric episodes in ™™ Gastroenteritis may be defined as inflammation of
some. the mucous membrane of stomach and intestine,
resulting in frequent loose motions with or without
Laboratory Diagnosis mucus and with or without blood, pain abdomen
and with or without fever. It is often used as a
i. Specimen: Feces synonym for acute diarrhea, especially when
associated with vomiting
ii. Microscopy
™™ Bacterial diarrhea Vibrio cholerae, E. coli, Salmonellae
are some important bacterial causes of diarrheal
a. Saline Preparation and Iodine Mount diseases. Rotavirus is the most important viral
etiology of diarrhea
In saline preparation, motility of trophozoites ™™ Dysentery is a disease marked by frequent
can be observed while cysts take up iodine and watery stools, often with blood and mucus, and
appear distinct in iodine mount. Cysts and motile characterized clini­cally by cramping abdominal pain,
trophozoites of E. histolytica can be observed in feces tenesmus, fever and dehydration
of amebic dysentery. Giardia lamblia cysts in formed ™™ Laboratory diagnosis of diarrhea, and dysentery
stools, or trophozoites in fresh diarrheal stools can be depends on isolation of organism from the relevant
specimen.
seen in giardiasis. Trophozoites of Balantidium coli
are found in liquid stool of this parasitic infection.
Rarely cysts may be seen in formed stools. Important questions
1. Enumerate the different causes of diarrhea. How will
b. Acid-fast Staining you diagnose a case of diarrhea in the laboratory.
2. Enumerate the etiological agents of dysentery.
Feces smear shows acid-fast oocyst of Crypto­
Discuss in detail the laboratory diagnosis of
sporidium parvum.
dysentery.
iii. Serological tests: Indirect hemagglutination 3. Write short notes on:
assay (lHA) and ELISA are used to detect a.  Traveller’s diarrhea
antibody titer in sera of patients with amebiasis. b.  Viral diarrhea
Chapter 78: Infective Syndromes  | 547

6. Food poisoning
The term bacterial food poisoning is restricted to 8–24 hours. The typical example of this type of food
acute gastroenteritis due to the presence of bacteria, poisoning is by salmonellae.
usually in large numbers, or their products in food.
It is of three types (Table 78.11). B. Toxic Type
In this type, the disease follows ingestion of food
A. Infective Type with preformed toxin. Incubation period is short
In this type, multiplication of bacteria occurs in (2 to 6 hours).Example is staphylococcal food
vivo when infective doses of microorganisms are poisoning.
ingested with food. Incubation period is generally
C. Infective-toxic Type
Table 78.11  Causative agents of food poisoning In this type, bacteria re­lease the toxin in the bowel.
The incubation period is 6–12 hours. The typical
1. Infectlve type example is Cl. perfringens food poisoning.
• Salmonella Typhimurium For the laboratory diagnosis, refer to the corres­
• S. Enteritidis ponding chapters. It has also been described under
• S. Heidelberg “Laboratory diagnosis of diarehea”.
• S. Indiana
Key Points
• S. Newport
™™ The term ‘bacterial food poisoning’ acute in is of
• S. Dublin
three types A. Infective type, B. Toxic type, and
• Vibrio parahaemolyticus Infective-toxic type.
• Campylobacter jejuni ™™ Laboratory diagnosis of food poisoning depends on
isolation of organism from the relevant specimen.
2. Toxic type
• Staphylococcus aureus
Bacillus cereus Important questions
• Clostridium botulinum 1. Name the various organisms causing food
poisoning. Describe briefly the laboratory diagnosis
3. Infectlve-toxic type
of this condition.
Clostridium perfringens
2. Write short notes on food-borne botulism.

7. Sexually transmitted Diseases (STDs)


The sexually transmitted diseases (STDs) are A. Syphilis
a group of communicable diseases which are Syphilis is caused by Treponema pallidum.
transmitted predominantly or entirely by sexual
contact. The causative organisms include a wide 1. Specimens
range of bacterial, viral. protozoal and fungal (i) Fluid from chancre; (ii) Scrapings from ulcerated
agents. STDs may present as genital ulcers, genital secondary lesions; (iii) Blood for serology.
discharge without any genital lesion or only as
2. Microscopy
systemic manifestations.
Dark-ground microscopy or phase-contrast micro­
scopy is used for demonstration of T. pallidum in
Etiology exudate. The direct fluorescent antibody test for T.
pallidum (DF ATP) is a better and safer method for
Organisms causing sexually transmitted diseases
microscopic diagnosis.
(STDs) and their clinical presentations are shown
in Table 78.12. 3. Serological tests
i. Nonspecific tests:
Laboratory Diagnoses a. VDRL test
Laboratory diagnoses of these diseases has already b. Rapid plasma reagin (RPR) test
been described in the corresponding chapters. ii. Specific tests:
However, the salient features of laboratory a. TPHA (Treponema pallidum hemagglutin­
diagnoses of important STDs are mentioned here. ation assay).

www.ebook3000.com
548  |  Section 6: Miscellaneous
Table 78.12  Organisms causing sexually trans­ 2. Isolation: Isolation of the Chlamydia by
mitted diseases and their clinical presentations intracerebral inoculation into mice and into yolk
sac of eggs has been replaced by cell cultures.
Sexually transmitted Organisms
diseases (STDs) 3. Serological tests: LGV patients develop high
titers of circulating antibodies, with titers of 1:64
A. Painless genital ulcers
or more in CF test and 1:512 or more in micro-IF.
• Syphilis Treponema pallidum
• Lymphogranuloma Chlamydia trachomatis Serological diagnosis is, therefore, feasible.
venereum (LGV) 4. Frei test: It is a skin test using LGV antigen and
• Donovanosis Calymmatobacterium shows delayed type of hypersensitivity.
granulomatis
B. Painful genital ulcers C. Donovanosis
• Chancroid Haemophilus ducreyi
• Herpes genitalis Herpes simplex viruses Donovanosis is caused by Calymmatobacterium
(HSV) type 2 and 1 granulomatis.
1. Specimen: Tissue smear from the ulcer
C. Urethral discharge
• Gonorrhea Neisseria gonorrhoeae 2. Staining: Diagnosis can be made by demon­
• Nongonococcal Chlamydia trachomatis stration of Donovan bodies in Wright-Giemsa-
urethritis (NGU) (types D-K) stained impression smears from the lesions.
Ureaplasma urealyticum They show bipolar condensation of chromatin,
Mycoplasma genitalium
giving a closed safety pin appearance in stained
M. hominis
smears. Capsules are usually seen as dense
D. Vaginal discharge acidophilic areas around the bacilli.
• Gonorrhea N. gonorrhoeae
• NGU C. trachomatis
M. hominis D. Chancroid
• Trichomoniasis Trichomonas vaginalis Chancroid or soft chancre is caused by H. ducreyi.
• Vaginitis Gardnerella vaginalis
Mobiluncus sp.
1. Specimen: Exudate
• Vulva-vaginal Candida albicans 2. Gram staining: Gram-negative coccobacilli are
candidiasis seen in Gram staining.
E. Genital warts Human papilloma viruses 3. Culture: Exudate is cultured onto chocolate agar
enriched with isovitalex and fetal calf serum,
F. No genital lesions HIV-1 and HIV-2
but only systemic Hepatitis B virus (HBV)
and containing vancomycin as a selective agent.
manifestations Hepatitis C virus (HCV) Culture plates are incubated at 35°C under 10
percent CO2 and in high humidity in 2–8 days.
G. Miscellaneous Group B streptococci
Molluscum contagiosum 4. Identification: For identification of the organism,
virus colony morphology and Gram staining are
Cytomegalovirus useful.
Phthirus pubis i. Colony morphology: H. ducreyi forms small,
Sarcoptes scabei
Shigella sp. gray, translucent colonies.
Campylobacter sp. ii. Gram-staining: It shows gram-negative
Giardia lamblia coccobacilli.
Entamoeba histolytica

E. Herpes genitalis
Herpes simplex virus (HSV), types 1 and 2, is the
b. FTA-ABS (fluorescent treponemal antibody etiological agent but type 2 strains are more com­
absorption test). monly associated.
c. TPI (Treponema pallidum immobilization 1. Specimens
test) i. Scrapings from base of the lesions
VDRL and TPHA are two most commonly
ii. Blood for serology
used tests.
2. Microscopy: Intranuclear type A inclusion
bodies may be seen in Giemsa-stained smears.
B. Lymphogranuloma Venereum (LGV) The virus particle may also be demonstrated
It is caused by C. trachomatis serotypes L1, L2 and L3. under the electron microscope.
1. Direct microscopy: Smears of material aspirated 3. Virus isolation: Diagnosis is confirmed by
from the bubos may show the elementary tissue culture in human diploid fibroblast cells.
bodies (Miyagawa’s granulocorpuscles). The Typical cytopathic changes may appear as early
sensitivity of microscopy is very low. as in 24–48 hours.
Chapter 78: Infective Syndromes  | 549
4. Serology: ELISA, neutralization or complement immunofluorescence test with a mono­
fixation tests are used for antibody detection clonal antibody or by ELISA.
and are useful in the diagnosis of primary 3. Culture: The exudate is inoculated on McCoy or
infection. HeLa cell cultures treated with cycloheximide.
Intracytoplasmic glycogen-rich inclusions
F. Gonorrhea are detected by Giemsa stain or by immuno­
It is caused by Neisseria gonorrhoeae. fluorescence. These are suggestive of C.
1. Specimens: Specimens used are: (i) Urethral trachomatis.
discharge; (ii) An endocervical swab; (iii) In 4. Serology
chronic cases; (iv) Rectal swab. i. Complement fixation test (CFT)
2. Direct Gram-staining: Gram-stain of the penile ii. Microimmunofluorescence or ELISA is useful
exudate shows characteristic and diagnostic for detection of serovar-specific antibody.
gram-negative intracellular diplococci (GNID)
in granulocytes.
H. Trichomoniasis
3. Culture: Commonly used selective media are
modified Thayer–Martin and Martin–Lewis It is caused by Trichomonas vaginalis.
plates. Inoculated plates should immediately 1. Specimen: Swab of vaginal discharge is exami­
be placed in an incubator at 37°C for 24–48 ned freshly. Specimen should be collected in
hours in the presence of CO2. Cultures with no stuart’s transport medium if delay in transport
visible growth must be held for 72 hours before is inevitable.
discarding. 2. Direct microscopy: Direct wet film shows
4. Identification: Identification of an organism motile trichomonads. Direct microscopy is at
is based on colony morphology, Gram staining least 80 percent as positive as culture.
from colonies and biochemical reactions. 3. Culture: Fineberg’s medium is used for culture
i. Colony morpholo g y: Small, gray, of specimen and it is incubated for 5 days and
translucent, raised colonies. examined for motile protozoa.
ii. Gram-staining: Gram-negative diplococci
iii. Biochemical reactions: They are oxidase I. Bacterial Vaginosis-associated Organisms
positive and produce acid from glucose but The diagnosis of bacterial vaginosis does not depend
not lactose, maltose or sucrose. on the isolation of a particular microorganism, e.g.
iv. Direct detection methods, such as Gardnerella vaginalis or Mycoplasma hominis, but on
fluorescent antibody and agglutination the replacement of the predominantly lactobacillary
tests, are also available. flora with a mixture of aerobes; and anaerobic
v. Nucleic acid probes for the direct detection organisms, a shift of pH to neutral or alkaline, and
of N. gonorrhoeae from clinical specimens. the presence of ‘clue’ cells. These changes are most
easily assessed from the Gram stained film.
G. Nongonococcal Genital Infection For diagnosis of Shigella sp, Campylobacter
Symptoms of discharge and dysuria clinically sp, Group B streptococci refer to corresponding
indistinguishable from gonorrhea caused by chapters.
organisms other than N. gonorrhoeae is called
nongonococcal urethritis (NGU). Causative agents J. Vulvovaginal Candidiasis
are shown in Table 78.13. A significant proportion It is caused by various species of Candida but C.
of nongonococcal genital infection in women is albicans accounts for 80% of cases.
generally due to chlamydia trachomatis. 1. Specimen: Swab from vaginal secretions
2. Direct microscopy
Laboratory Diagnosis
i. KOH mount: It shows yeast cells.
1. Specimens
(i) Swabs from exudate of urethra; (ii) Cervical ii. Gram-staining: Gram staining shows
discharge. characteristic gram-positive budding yeast
2. Direct examination cells and pseudohyphae.
i. Giemsa stain: It shows intracytoplasmic 3. Culture: Sabouraud’s dextrose agar (SDA) is
inclusion bodies suggestive of C. tracho­ inoculated with the specimen and incubated
matis. at 37°C for 48 hours.
ii. Antigen detection: For detection of ele­ 4. Identification
mentary bodies of C. trachomatis, smears i. Colony morphology: Colonies are creamy
are made from exudate and examined by white and smooth.

www.ebook3000.com
550  |  Section 6: Miscellaneous
ii. Gram staining: Gram-stained smear shows Key Points
budding gram-positive yeast cells.
™™ The sexually transmitted diseases (STDs) are a group
Germ tube formation: C. albicans forms
iii.
of communicable diseases which are transmitted
germ tube within two hours when incubated predominantly or entirely by sexual contact. The
in human serum at 37°C. causative organisms include a wide range of
iv. Chlamydospores formation: On cornmeal bacterial, viral. protozoal and fungal agents
™™ For laboratory diagnosis and treatment according
agar C. albicans forms chlamydospores.
to the suspected organism responsible for the
manifestations of that particular STD.
K. Genital Warts
Genital warts, also known as condyloma acuminata, Important Questions
are common in sexually active adults. These are 1. Name the various organisms causing sexually
usually due to human papillomavirus (HPV) types transmitted diseases. Discuss the laboratory
6 and 11. diagnosis of syphilis.
For detection of inclusion bodies of HPV, 2. Write short notes on:
cytological or histological examination of cells in a. Laboratory diagnosis of gonorrhea
b. Nongonococcal urethritis (NGU)
urine is used.
c. Chancroid
For diagnosis of Shigella sp, Campylobacter spp, d. Lymphogranulum venereum (LGV)
Group B streptococci, refer to the corresponding e. Donovanosis
chapters. f. Vulvovaginal candidiasis.

8. Wound Infection
Wound infections may be endogenous or exogenous. Gas chromatography may be performed directly
It may be caused by a variety of aerobic and on liquid specimens to indicate the presence of
anerobic species of bacteria (Table 78.13). anerobes.

Laboratory Diagnosis A. Naked-eye Examination


Pus or exudate is often submitted on a swab for
laboratory investigation. If possible, send two The pus of a staphylococcal lesion is typically
swabs taken from the depths of the wound or creamy and thick in consistency.
lesion, so that one can be used for the preparation The pus of a Streptococcus pyogenes infection
of a smear for microscopy and the other for the is generally straw-coloured and watery.
seeding of cultures The pus of proteus infection has a fishy smell.
The basic procedures usually include a The pus of pseudomonas infection a sweet,
naked-eye examination of the specimen, the musty odor and often a blue pigmentation.
microscopical examination of a Gram film, and Pus containing anerobic organisms often has
culture on aerobic and anerobic blood agar plates, an offensive putrid smell, and that of actinomycosis
on MacConkey agar and in cooked-meat broth. may contain small microcolonies that appear as
‘sulfur granules’. In some fungal infec­tions such
Table 78.13  Microorganisms causing wound as mycetoma, black or brown granules may be
infection present. The pus of an amoebic abscess is said to
resemble anchovy sauce.
A. Aerobes
Staphylococcus aureus
β-haemolytic streptococci B. Microscopy
•  Enterococcus spp
Smear Stained by Gram’s Method
•  Streptococcus pneumoniae
•  Escherichia coli Gram-positive cocci in typical clusters may
•  Other coliform bacilli suggest a staphylo­coccal infection,
•  Proteus spp In chains, streptococcal infection.
•  Pseudomonas aeruginosa Gram-positive diplococci may be given by either
•  Mycobacterium marinum
pneumococci or enterococci.
•  Nocardia spp
B. Anerobes Gram-variable filaments of Actinomyces may
•  Peptostreptococci appear like chains of cocci and their fragments as
•  Bacteroides spp diphtheroid bacilli.
•  Clostridium perfringens and other clostridia. Examination of a wet film may reveal the presence
•  Actinomyces israelii of fungi or motile bacteria.
Chapter 78: Infective Syndromes  | 551
Darkground microscopy of a wet film is useful in for their identification should be done, e.g. the
the diagnosis of primary syphilis. coagulase test on staphylococci, Lancefield’s
A smear stained by the auramine or Ziehl–Neel­ grouping of β-hemolytic streptococci, and
sen method—for tubercle bacillus, another biochemical tests on coliform bacilli and anerobes.
Mycobacterium or a Nocardia. For detailed laboratory diagnosis, refer to the
corresponding chapters.
C. Culture
The specimen should be inoculated onto two
E. Antibiotic Sensitivity
plates of blood agar, the one for incubation at At the same time, the pure cultures should be tested
37°C aerobically, preferably in air plus 5–10% CO2, for sensitivity to an extended range of antibiotics
the other for incubation anaerobically in nitrogen useful in therapy.
hydrogen plus 5–10% CO2, It should also be plated
Key Points
for aerobic incubation on MacConkey agar or
CLED agar and be inoculated into a tube of cooked- ™™ Wound infections may be caused by a variety of
meat broth for the enrichment of exacting aerobes aerobic and anerobic species of bacteria
™™ Laboratory Diagnosis: Pus or exudate is often
and anerobes. The culture plates are examined
submitted on a swab for laboratory investigation.
after overnight incubation at 37°C for 18–24 hours. The basic procedures usually include a naked-
If at 24 or 48 hours there is growth in the eye examination of the specimen, microscopical
cooked-meat broth, but no growth on the plates, examination of a Gram film, and culture on aerobic
the broth should be filmed and subcultured, both and anerobic blood agar plates, on MacConkey agar
aerobically and anerobically. If tuberculous or and in cooked-meat broth. Gas chromatography
may be performed directly on liquid specimens to
fungal infection is suspected, the specimen should indicate the presence of anerobes.
be cul­tured by the appropriate methods on the
appropriate special media.
Important questions
D. Identification of Isolates
1. Name the various microorganisms causing wound
After the bacteria cultured have been obtained infection. Write briefly on laboratory diagnosis of
in pure subcultures, any further necessary tests wound infection.

9. Pyrexia of Unknown Origin (PUO)


Pyrexia of unknown origin (PUO) may be defined 2. Collection
as (a) any febrile illness (body temperature greater
These specimens must be collected in sterile
than 38°C) on several occasions, (b) duration of
containers under aseptic conditions. Blood is
fever of more than 3 weeks, and (c) failure to reach a
collected in blood culture bottles for culture and in
diagnosis despite 1 week of inpatient investigation.
a sterile vial for serology. Blood culture should be
It is also known as ‘fever of unknown origin’
collected before antibiotics are given. Midstream
(FUO).
urine specimen of urine (MSU) should be collected
in a sterile universal container.
Causes of PUO
Infection is the most common cause of PUO. 3. Culture
However, there are important non-infectious causes
of fever. The causes of PUO are given in Table 78.14. i. Blood culture
For blood culture, 5 mL of blood is collected in
Laboratory Diagnosis of PUO each bottle of 50 mL glucose broth and 50 mL
taurocholate broth. These broths are incubated at
Tests should first be done for the more likely infec­ 37°C for 24 hours and then subcultures are made on
tions and then, if these are negative, tests for the blood agar (from glucose broth) and MacConkey
less likely should be done. agar (from taurocholate broth). Blood agar and
MacConkey agar plates are incubated at 37°C for
A. Bacterial Infections 24 hours.
1. Specimens
ii. Urine culture
Blood: for blood culture, peripheral blood smear,
A calibrated volume of midstream urine specimen
haematology, serology and other tests.
is inoculated on blood agar and MacConkey agar.
Urine, Sputum, Pus. These media are incubated at 37°C for 24 hours.

www.ebook3000.com
552  |  Section 6: Miscellaneous
Table 78.14  Causes of pyrexia of unknown origin (PUO)
A. Infective causes B. Noninfective causes
a. Bacterial a. Neoplasms
• Urinary tract infections • Hodgkin’s lymphoma
• Lung, subdiaphragmatic, appendix and other deep • Non-Hodgkin’s lymphoma
abscesses • Leukemia
• Septicemia associated with cryptic abscesses, • Hypernephroma
pneumonia, pyelonephritis, biliary tract infection, • Hepatoma
infective endocarditis and immuno­deficiencies • Disseminated malignancy
• Enteric fever b. Connective tissue disorders
• Tuberculosis • Systemic lupus erythematosus (SLE)
• Brucellosis • Polyarteritis nodosa
• Syphilis • Temporal arteritis
• Relapsing fever c. Granulomatous diseases
• Rheumatic fever • Sarcoidosis
• Leptospirosis without jaundice or meningitis • Crohn’s disease
• Typhus fever • Granulomatous hepatitis
• Nonmeningitic meningococcal infection d. Drug reactions
• Q fever Drug-induced fevers
b. Parasitic
• Malaria
• Hepatic amebiasis
• Leishmaniasis
• Trypanosomiasis
• Toxoplasmosis
• Filariasis
c. Viral
• EBV infection
• CMV infection
• HIV infection
• Rubella and other infectious fevers without typical
rash

Culture should be performed on Lowenstein– 5. Serology


Jensen (L–J) medium in case of renal tuberculosis. Paired sera should be collected for serological
tests for antibody responses to a range of possible
iii. Sputum culture
pathogens, e.g. cytomegalovirus, hepatitis B
Specimen is inoculated on blood agar and virus, Coxiella, Salmo­nella, Brucella, Leptospira,
MacConkey agar plates and incubated at 37°C Toxoplasma, Aspergillus and Entamoeba. HIV
for 24 hours. Specimen should be cultured on infection should also be con­sidered. The anti­
Lowenstein–­J ensen (L–J) medium in case of streptolysin-O (ASO) test should be done for
tuberculosis and incubated at 37°C for 6 weeks. cryptic Streptococcus pyogenes infection.
iv. Pus culture
B. Parasitic Infections
Pus is inoculated in glucose broth, blood agar and
MacConkey agar. These media are incubated at Stained peripheral blood films smears (thin
37°C for 24 hours. Pus should be cultured on L–J and thick) will help in diagnosis of malaria,
media for M. tuberculosis. Culture of pus should leishmaniasis, trypanosomiasis and filariasis.
be performed under anaerobic conditions when Wet blood film may show microfilaria in cases of
suspecting anaerobic organisms. filariasis. Serology is useful in amebiasis.

C. Viral Infections
4. Identification
Peripheral blood smear may be helpful in infectious
Organisms may be identified on the basis of
mononucleosis. Viral infections may be detected
colony morphology, Gram-staining, biochemical
either by tissue culture or by serology. Paul–Bunnel
reactions and agglutination, etc. Ziehl–Neelsen
test is useful in infectious mononucleosis.
(Z–N) staining is performed to detect acid-fast
bacilli (AFB) for M. tuberculosis. This is further
confirmed by culture and biochemical reactions. D. Fungal Infections
For details of individual organisms, refer to Specimens may be cultured on Sabouraud’s
corresponding chapters. dextrose agar or brain–heart infusion agar.
Chapter 78: Infective Syndromes  | 553
E. Other Tests for Diagnosis 4. The most common viral agent causing aseptic
meningitis is:
1. Skin Tests a. Enteroviruses
Mantoux test: A tuberculin test and a chest X-ray b. Adenoviruses
should be done to detect tuberculosis. c. Cytomegalovirus
Skin tests for histoplasmosis, coccidioido­ d. Parainfluenza virus
mycosis, sarcoidosis should also be done. 5. Which of the following bacteria is the most
comrnon cause of UTI:
2. Hematological Investigations a. E. coli
Hematological investigations should be done b. Ps. aeruginosa
to detect leukocytosis, suggestive of a cryptic c. Staph. aureus
abscess; eosinophilia, suggestive of helminthiasis; d. Staph. saprophyticus
and atypical lymphocytes, suggestive of infectious 6. All the following are causative agents of sore throat
mononucleosis. except:
a. Streptococcus pyogenes
3. Immunologic Tests b. Haemophilus influenzae
LE cell phenomenon and antinuclear antibody c. Borrelia vincenti
test in SLE. d. Staphylococcus aureus
7. Which of the following organisms may lead to
4. Biopsy pseudomembrane formation in throat:
a. C. diphtheriae b. Strep. pyogenes
Biopsy of liver and bone marrow and other tissues
c. Candida albicans d. All the above
such as skin, lymph nodes and kidney.
8. Which of the following agents can cause diarrhea?
Key Points a. Rotavirus b. Vibrio cholerae
c. S. Typhimurium d. All the above
™™ Pyrexia of unknown origin (PUO) may be defined
as (a) any febrile illness (body temperature greater 9. Which of the following protozoa can cause diarrhea:
than 38°C) on several occasions, (b) duration of a. Entamoeba histolytica
fever of more than 3 weeks, and (c) failure to reach a b. Cryptosporidium parvum
diagnosis despite 1 week of inpatient investigation.
c. Giardia lamblia
It is also known as ‘fever of unknown origin’ (FUO)
d. All the above
™™ The causes of PUO include infections bacterial, parasitic
and viral) neoplasms, connective tissue disorders, 10. Which of the following agents can cause dysentery:
granulomatous diseases and drug reactions a. Escherichia coli (EIEC)
™™ Tests should first be done for the more likely infec­ b. Shigella dysenteriae
tions and then, if these are negative, tests for the less
c. Entamoeba histolytica
likely should be done.
d. All the above
11. Which of the following bacteria can cause infective
Important Question type of food poisoning:
Define and enumerate the causes of pyrexia of unknown a. S Enteritidis
origin (PUO). Discuss the laboratory diagnosis of PUO. b. Clostridium botulinum
c. Staph. aureus
Multiple choice questions (MCQs) d. All the above
12. Infective-toxic type of food poisoning is caused by:
1. Presence of bacteria in blood without multiplication
a. Staph. aureus
is known as:
b. Salomonella Enteritidis
a. Bacteremia b. Septicemia
c. Clostridium perfringens
c. Pyemia d. Endotoxemi
d. Salmonella Dublin
2. Which of the following bacteria can cause purulent
meningitidis: 13. Ulcer/s is/are painless I n which of the following
a. H. influenzae sexually transmitted diseases:
b. Strep. pneumoniae a. Syphilis b. Chancroid
c. N. meningitis c. Herpes genitalis d. All of the above
d. All the above 14. Genital ulcer is painful in:
3. Which of the following agents cause/s aseptic a. Syphilis
meningitis? b. Chancroid
a. Herpes simplex b. Enteroviruses c. lymphogranuloma venereum
c. Arboviruses d. All of the above d. Donovanosis

www.ebook3000.com
554  |  Section 6: Miscellaneous
15. Which of the following bacteria is/are implicated 17. Which of the following anerobes can cause wound
as causative agents of nongonococcal urethritis: infection:
a. C. trachomatis a. Bacteroides spp
b. Peptostreptococci
b. Ureaplasma urealyticum
c. Clostridium perjringens
c. Mycoplasma hominis. d. All the above
d. All of the above 18. Which of the following conditions can result in PUO:
16. Urethral discharge is present in: a. Urinary tract infection b. Enteric fever
a. Chancroid c. Malaria d. All the above
b. Gonorrhea Answers (MCQs)
c. Herpes genitalis 1. a; 2. d; 3. d; 4. a; 5. a; 6. d; 7. d; 8. d; 9. d; 10. d; 11. d;
d. Lymphogranuloma venereum 12. c; 13. a; 14. b; 15. d; 16. b; 17. d; 18. d.
79
Chapter

Hospital-acquired Infection

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe common hospital-associated infections
be able to: and causative organisms responsible for these
∙∙ Define hospital-associated infection conditions
∙∙ List of routes of transmission of hospital-associa­ted ∙∙ Describe diagnosis and control of hospital-
infections associated infections.

Introduction 2. Susceptibility to infection: Preexisting disease,


such as diabetes, or other conditions, and
The terms hospital infection, hospital-acquired the medical or surgical treatment, including
infection or nosocomial infection (from nosoco­ immunosuppressive drugs, radiotherapy or
meion, meaning hospital) are applied to infections splenectomy, may also reduce the patient’s
developing in hospitalized patients, not present or natural resistance to disease.
in incubation at the time of their admission. 3. Hospital environment: Patients shed them
from their bodies; hospital personnel spread
Sources of infections them through their hands and clothes. Bedding,
Hospital infection may be exogenous or endogen­ linen and utensils act as fomites. Equipment
ous in origin. may be contaminated. Patho­gens are present
in the hospital dust and air, and sometimes
A. Exoge­nous even in antiseptic lotions and ointments.
Contamination of hospital food or water may
Exogenous source may be another person in the cause outbreaks of infections.
hospital (cross­-infection) or a contaminated item 4. Diagnostic or therapeutic procedures.
of equipment or building service (environmental 5. Drug-resistance: The hos­pital microbial flora
infection). is usually multidrug resistant due to injudicious
1. Contact with other patients and staff. use of antibiotics, thus limiting the choice of
2. Environmental sources: These include inani­ therapy.
mate objects, air, water and food in the hospital.
6. Transfusion: Blood, blood products and
intravenous fluids used for transfusion, if not
B. Endogenous properly screened, can transmit many infections.
A high proportion of clinically apparent hospital 7. Advances in medical progress: Advances in
infections are endogenous (self-infection), the treatment of cancer, organ transplanta­tion,
infecting organism being derived from the patient’s implanted prostheses and other sophisticated
own skin, gastrointestinal or upper respiratory flora. medical technologies enhance the risk of
infection to patients.
FACTORS INFLUENCING HOSPITAL-
ASSOCIATED INFECTIONS Microorganisms causing hospital
infection
1. Age: Natural resistance to infection is lower in
infants and the elderly, who often constitute the Almost any microbe can cause a hospital-acquired
majority of hospital patients. infection, but those that are able to survive in

www.ebook3000.com
556  |  Section 6: Miscellaneous
the hos­pital environment for long periods and skin scales, spread to the susceptible site,
develop resist­ance to antibiotics and disinfectants e.g. Ps aeruginosa, Staph. aureus.
are particularly important in this respect. Though iii. Aerosols: Aerosols produced by nebulizers,
protozoal infections are rare. Strep. pyogenes was, humidifiers and air condi­tioning apparatus
per­haps, the most important cause of hospital transmit certain pathogens to the
infection formerly but is hardly ever encountered respiratory tract. Occurrence of legionellae
now as it is highly susceptible to antibiotics. in hos­pital water supply and a number of
1. Staph. aureus: Staph. aureus strains, resistant persons with an impaired immune system
to multiple antibiotics and belonging to has led to outbreaks of infection mainly
phage type 80/81, spread globally in the 1950s
with Legionella pneumophila.
and 1960s, colonizing hospitals and causing
nosocomial infection. Subsequently, epidemic 3. Oral route: Hospital food contains gram-­
or pandemic strains characterized by resistance negative bacilli which are most often antibiotic
to methi­cillin-resistant Staphylococcus aureus resistant (P. aeruginosa, E. coli, Klebsiella spp.
(MRSA) have been found in many hospitals and others), which may colonize the gut and
world­w ide.Staph. epidemidis and Group D later cause infection in susceptible patients.
streptococci also are sometimes respon­sible 4. Parenteral route (inoculation): Certain
for hospital infections. infections may be transmitted by blood
2. Pseudomonas species: Ps. aeruginosa and transfusion or tissue donation, contaminated
other Pseudomonas species have always been blood-products (factor VIII), contaminated
important causes of hospital infec­tion. infusion fluids and from accidental injury
with contamina­ted sharp instruments (HIV,
3. Tetanus spores: Hospital tetanus is usually due
hepatitis B and C).
to faulty sterilization techniques or other lapses
in asepsis. 5. Self-infection and cross-infection: Self-
4. Viral infections: Viral infections probably infection may occur due to transfer into the
account for more hospital-acquired infections wound of staphylococci (or occasion­a lly
than previously realized. streptococci) carried in the patient’s nose and
dis­tributed over the skin, or of coliform bacilli
i. HIV and hepatitis B and C viruses are
and anaerobes released from the bowel during
transmitted by contaminated blood or
surgery. Alternatively, cross-infection may result
blood products. Screening of blood donors from staphylo­cocci or coliform bacilli derived
has reduced the risk to a large extent. from other patients or healthy staff carriers.
However, HIV escapes detection during
the window period. Common hospital-acquired
ii. Viral diarrhea and chickenpox are other infection
viral infections that spread in hospitals.
iii. Cytomegalovirus, herpesvirus, influenza, 1. Urinary Tract Infection
enteroviruses and arenaviruses may also Most hospital-acquired infections of the urinary tract
cause hospital infection. are associated with urethral catheterization. Urinary
5. Fungus and parasites: The range of hospital tract infection is caused by Esch. coli, Klebsiella,
pathogens also includes yeasts (Candida Proteus, Serratia, Pseudomonas, Providencia, coagu­
albicans), moulds, (Aspergillus, Mucor) and lase negative staphylococci, enterococci and Candida
protozoa (Entamoeba histolytica, plas­modia, albicans.
Pneumocystis carinii, Toxoplasma gondii).
2. Respiratory Infections
ROUTES OF TRANSMISSION Aspiration in unconscious patients and pulmonary
1. Contact ventilation or instrumentation may lead to
Direct contact: Spread from person to person nosocomial pneumonia, particularly in those
(staphylococcal and streptococcal sepsis). with preexisting cardiopulmonary disease. The
Indirect contact: Spread via contaminated major pathogens include Staph. aureus, Klebsiella
hands or equipment (enterobacterial diarrhoea, spp., Enterobacter, Serratia, Proteus, Esch. coli,
Pseudomonas aeruginosa sepsis). Pseudomonas aeruginosa, Acinetobacter, Legionella
2. Airborne spread pneumophila and respiratory viruses.
i. Droplets
ii. Dust: Dust from bedding, floors; exudate 3. Wound and Skin Sepsis
dispersed from a wound during dressing The incidence of postoperative infection is higher
and from the skin by natural shedding of in elderly patients. Most wound infections manifest
Chapter 79: Hospital-aquired Infection  | 557
within a week of surgery. Staph. aureus is the medical microbiologist who will usually serve
predominant pathogen, followed by Pseudomonas as chairman, a physician, a surgeon, nurse
aeruginosa and then Esch. coli, Proteus, enterococci teachers, nurse repre­sentatives for surgery,
and coagulase negative staphylococci. obstetrics, gynecology and medicine, and
sterile service manager. The ICC should meet
4. Gastrointestinal Infections regularly to formulate and update policy for the
Food poisoning and neonatal septicemia in hospital whole hospital matters having implications for
have been reported. These infections are mainly infection control and to manage outbreaks of
associated with salmonella and Shigella sonnei. no noso­comial infection.
Roles of the infection control committee
5. Burns i. The surveillance of hospital infection.
Staph. aureus, Pseudomonas aeruginosa , ii. The establishment and monitoring of policies
Acinetobacter and Strept. pyogenes are responsible and proce­dures designed to prevent infection,
for hospital-acquired infections in cases of burns. e.g. catheter care policy, antibiotic policy,
disinfectant policy.
iii. The investigation of outbreaks.
6. Bacteremia and Septicemia
2. Infection control team: An infection control
These may be conse­q uences of infections at team of workers, headed by the infection
any site but are commonly caused by infected control doctor (usually the microbiologists).
intravenous cannulae. Gram-negative bacilli are The functions of this team include surveillance
the common pathogens. and control of infection and monitoring of
Staph. epidermidis bacteremia is seen hygiene practices. The infection control nurse
commonly in patients with artificial heart valves. is a key member of this team.
Bacteremia in those with valvular defects may lead
to endocarditis. Prevention
Diagnosis and control of The hospital-acquired infections can be prevented
by following means:
hospital infection
1. Sterilization: The provision of sterile
The most important steps in’ preventing instruments, dressings, surgical gloves, face-
nosocomial infections are to first recognize their masks, theater clothing and fluids.
occurrence and then establish policies to prevent 2. Cleaning and disinfection: The general
their development. Hospital infection may occur hospital environment can be kept in good
sporadically or as out­breaks. order by attention to basic cleaning, waste
Etiological diagnosis is by the routine bacterio­ disposal and laundry. The use of chemical
logical methods of smear, culture, identification disinfectants for walls, floors and furniture is
and sensitivity testing. When an outbreak occurs, necessary only in special instances.
the source should be identified and eliminated. 3. Skin disinfection and antiseptics: Procedures
This requires the sampling of possible sources of for preoperative disinfection of the patient’s
infection. Typing of isolate—phage, bacteriocin, skin and for surgical scrubs are mandatory
antibiogram or biotyping—from cases and sites within the operating theater.
may indicate a causal connection. 4. Rational antibiotic prophylaxis
The provision of sterile instruments, dressings 5. Protective clothing
and fluids is of fundamental importance in hospital 6. Isolation source isolation and protective
practice. The cause of infection may be a defective isolation.
autoclave or im­proper techniques, such as boiling 7. Hospital building and design: The routine
infusion sets in ward sterilizers. A careful analysis of maintenance.
the pattern of infection may often reveal the source 8. Equipment
but sometimes it eludes the most diligent search. 9. Personnel: Hepatitis B vaccine should be
given to all health care workers.
Infection control policy 10. Monitoring: Monitoring of the physical
The establishment of an effective infection performance of air-conditioning plants
control orga­nization is the responsibility of good and machinery used for disinfection and
management of any hospital. There will normally sterilization is essential.
be two parts: 11. Surveillance and the role of the laboratory:
1. Infection control com­mittee (ICC): Every The detection and characterization of hospital
hospital should have an infection control com­ infection incidents or outbreaks rely on
mittee (ICC). The committee will consist of a laboratory data.

www.ebook3000.com
558  |  Section 6: Miscellaneous

Key Points c. Prevention of hospital-associated infections


d. Disinfection policy
™™ Hospital-acquired infections or nosocomial e. Infection control committee
infections are infec­tions occurring in hospitalized
patients who were neither infected nor were in Multiple choice questions (MCQs)
incubation at the time of their admission
™™ The sources of hospital-acquired infection may be 1. Bacteria that are most commonly transmitted by
exogenous or endogenous direct hand contact, causing nosocomial infection
™™ Nosocomial infections are transmitted by air, are
direct contact, oral route, and parenteral route. a. Escherichia coli
UTls, nosocomial pneumonia, and surgical wound
b. Enterococcus species
infections are the common examples of hospital
infections
c. C. Staphylococcus aureus
™™ Diagnosis of hospital-acquired infections is made by d. Clostridium perfringens
routine bacteriological methods 2. Hospital-associated respiratory infections are
™™ The source of infection may be traced by performing caused by which of the following bacteria?
phage typing, bacteriocin typing, biotyping, or a. Staphylococcus aureus b. Klebsiella
molecular typing c. Enterobacter d. All of the above
™™ Active hospital surveillance is the key for successful 3. The pathogen that is transmitted by air in a hospital
hospital infection control
is
™™ Every hospital must have an effective hospital-
a. Hepatitis A virus
acquired infection control committee (ICC).
b. Legionella species
c. Shigella species
Important questions d. Human immunodeficiency virus
4. The control of hospital-acquired infection is the
1. Define hospital-associated infection. Enumerate main responsibility of:
organisms causing it. What are the factors which a. Medical superintendent of hospital
influence development of this infection? b. Head of department of medicine
2. Write short notes on: c. Head of department of microbiology
a. Routes of transmission of hospital-associa­ted d. Hospital infection control committee
infections
b. Diagnosis and control of hospital-associated Answers (MCQs)
infections 1. c; 2. d; 3. b; 4. d
80
Chapter

Laboratory Control of Antimicrobial Therapy

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Explain the meaning of sensitive, intermediate and
be able to: resistant applied to antimicrobial susceptibility
∙∙ List different methods of antibiotic sensitivity test results
testing ∙∙ Describe the following: Epsilometer or E-Test;
∙∙ Describe disk diffusion methods Minimum inhibitory concentration of antimi­crobial
∙∙ Explain Stokes disk diffusion method and its reporting agents;Minimum bactericidal concentration of
∙∙ Differentiate between Stokes disk diffusion and antimicrobial agents.
modified Stokes disk diffusion method

INTRODUCTION Medium
It is essential to determine the susceptibility of Mueller–Hinton broth and agar may be used for
isolates of pathogenic bacteria to antibiotics that testing aerobic and facul­tative anaerobic isolates.
are likely to be used in treatment. As strains of most The medium is and poured to a depth of 4 mm
pathogenic organisms differ from one another (25 mL medium) in flat-bottomed 9 cm Petri dishes
within their species in their antibiotic sensitivities, on a level surface. When set, the plates may be
sensitivity tests are required as a routine. stored for up to a week at 4°C and their surfaces
should be dried with their lids ajar before use. The
ANTIBIOTIC SENSITIVITY TESTS pH of the medium must be close to 7.3. A more acid
Antibiotic sensitivity tests are of two types: reaction decreases the activity of amino­glycoside
A. Diffusion methods and macrolide antibiotics. A more alkaline pH
1. Kirby–Bauer disk diffusion method favors the action of tetracyclines, novobiocin
2. Stokes disk diffusion method and fusidic acid, but interferes seriously with the
B. Dilution methods activity of nitrofurantoin.
1. Broth dilution method The addition of 5% lysed horse blood is needed
2. Agar dilution method to support the growth of fastidious species such
Diffusion methods: Here the drug is allowed as Haemophilus influenzae. Lysed horse blood
to diffuse through a solid medium so that a should also be added for tests with sulfonamides
gradient is estab­lished, the concentration and trimethoprim; its content of thymidine
being highest near the site of application of phosphorylase is needed to neutralize the inhibitory
the drug and decreasing with distance. The effect of thymidine in the medium on the action of
test bacterium is seeded on the medium and these drugs. Low levels of free Ca2+ and Mg2+ ions in
its sensitivity to the drug determined from the the medium increase the action of aminoglycoside
inhibition of its growth. Several methods have antibiotics against Pseudomonas aeruginosa.
been used for the application of the drug. The The addition of 5% NaCl to the medium
method most commonly em­ployed is to use is needed in one of the methods for detecting
filter paper disks, impregnated with antibiotics. resistance to methicillin in strains of staphylococci .
Disk Diffusion Methods Inoculum: Prepare the inoculum from material
These methods are suitable for organisms that picked up with a loop from five to ten colonies
grow rapidly overnight at 35°C. of the species to be tested. Inoculate them in a

www.ebook3000.com
Chap-80.indd 559 15-03-2016 11:26:48
560  |  Section 6: Miscellaneous
Table 80.1  Control strains for Kirby–Bauer and
Stokes disk diffusion methods
Test bacteria Control strain
Kirby–Bauer Stokes
Coliform E. coli ATCC 25922 E. coli NCTC
organisms 10418
Pseudomonas P. aeruginosa P. aeruginosa
ATCC 27853 NCTC 10662
Haemophilus H. influenzae H. influenzae
spp ATCC 49247 NCTC 11931
Gonococci N. gonorrhoeae N. gonorrhoeae
Fig. 80.1: Principle of antibiotic diffusion in agar. The ATCC 49226 (sensitive strain)
concentration of antibiotic decreases as the distance from Enterococci E. faecalis ATCC E. faecalis
the disk increases 29212 NCTC 12697
Other organisms S. aureus ATCC S.aureus
that can grow 25923 NCTC 6571
suitable broth medium. Incubate at 35–37°C for
aerobically
4–6 hours when the growth is considered to be in
logarithmic phase. The density of the organisms is
adjusted to approximately 108 colony-­forming units Table 80.2  Diluents used for various antibiotics
(cfu)/mL by comparing its turbidity with that of 0.5
Antibiotic Diluent
McFarland opacity standard (Fig. 80.1).
Chloramphenicol Ethanol

Control Strains Rifampicin Ethanol


Erythromycin Ethanol
Control strains for Kirby–Bauer and Stokes disk
diffusion methods are given in Table 80.1. Nitrofurantoin NaOH solution
Sulfonamides NaOH solution
Antibiotic Disks Trimethoprim Acetic acid
Commercially, prepared disks 6 mm in diameter Amoxicillin NaHCO3 (saturated)
should be used. Manufac­turers produce disks with Ceftazidime NaHCO3 (saturated)
accurate antibiotic content. If disks are prepared
locally in the laboratory, then pure antimicrobial
agents obtained from the manufacturers and KIRBY–BAUER DISK DIFFUSION METHOD
not the ones for clinical use should be used. Procedure
Proper diluents should be used. Distilled water A. Preparation of Inoculum (Growth Method)
serves to dissolve most antibiotic powders, but
chloramphe­nicol, rifampicin and erythromycin Dip a sterile nontoxic cotton swab into the
must first be dis­s olved in a small amount of inoculum suspension and rotate the swab several
ethanol, nitrofurantoin and sulfonamides in times with firm pressure on he inside wall of the
small volume of NaOH solution, trimethoprim in tube to remove excess fluid. Inoculate the dried
weak acid (acetic or lac­tic), and amoxicillin and surface of a Mueller–Hinton agar plate that has been
ceftazidime in a small vol­ume of saturated NaHC03 ‘brought to room temperature by streaking the swab
(Table 80.2). three times over the entire agar surface. Replace the
Disks and disk dis­pensers should be stored in lid of the dish. Allow at least 3–5 minutes but no
sealed containers with a desiccant. Before they are longer than 15 minutes for the surface of the agar
opened for use, the containers should be allowed to to dry before adding the antibiotic disks.
warm up slowly at room temperature to minimize
condensa­t ion of moisture, which may lead to B. Testing of Antibiotics
hydrolysis of the antibiotic. Therefore, they should The appropriate antimicrobial-impregnated disks
be taken out from refrigerator 1–2 hours before are placed on the surface of the agar, using either
applying on the culture medium. Disk contents of sterile forceps or multidisk dispenser. Disks must
antimicrobial agents are given in Table 80.3. be evenly distributed on the agar so that they are
Drugs to be tested against each species of bac­ no closer than 24 mm from center to center. On
teria should be grouped in sets of six or seven, the a plate of 100 mm diameter, seven disks may be
maximum number that can be accommodated applied, one in the center and six in the periphery
on a single 100 mm diameter plate by Stokes and (Fig. 80.2). The plates are then incubated at
Kirby–­Bauer methods, respectively. 35°C for 16–18 hours (24 hours when testing

Chap-80.indd 560 15-03-2016 11:26:49


Chapter 80: Laboratory Control of Antimicrobial Therapy  | 561
Table 80.3  Disk contents of various antimicrobial
agents for the comparative methods
Antimicrobial agent (and test option) Disk content
Benzylpenicillin
staphylococci 2 IU (1.2 mg)
pneumococci and meningococci 0.25 IU (0.15 mg)
Ampicillin
Enterobacteriaceae and 10 mg
enterococci 2 mg
Haemophilus and Moraxella spp
Amoxycillin/clavulanate
Enterobacteriaceae 20 mg/10 mg
Haemophilus spp, Moraxella spp Fig. 80.2: Kirby–Bauer disk diffusion method
and staphylococci 2 mg/1 mg
Piperacillin 30 mg staphylococci against methicillin or oxacillin or
Mezlocillin 30 mg enterococci against vancomycin).
Azlocillin 30 mg C. Interpretation
Cephalothin 30 mg Examine the plates after overnight incubation. With
Cephalexin 30 mg the use of sliding calipers, a ruler, or a template,
Cephadroxil 30 mg the zones of complete growth inhibition around
Cephradine 30 mg each of the disks are carefully measured to within
Cefuroxime 30 mg the nearest millimeter. The diameter of the disk is
Ceftazidime 10 mg included in this measurement.
Cefotaxime 10 mg The interpretation of zone size into susceptible
Cefsulodin 30 mg (infection treatable with nonnal dosage), moderately
Methicillin 5 mg susceptible (infection that may respond to therapy
Carbenicillin 100 mg with higher dosage) or resistant (not treatable with
Ticarcillin 75 mg this agent) is based on the inter­pretation chart
Imipenem 10 mg
(Table 80.4). Reference strains of S. aureus, E. coli,
P. aeruginosa, etc. should be tes­ted each time a new
Gentamicin 10 mg
batch of disks or agar is used.
Amikacin 30 mg
Kirby–Bauer test results are interpreted using
Tobramycin 10 mg a table that re­lates zone diameter to the degree of
Neomycin 30 mg microbial resistance.
Netilmicin 10 mg
Erythromycin 5 mg STOKES DISK DIFFUSION METHOD
Clindamycin 2 mg For Stokes disk diffusion method, the plate is divided
Tetracycline 10 mg into three parts. The control inoculum should be
Fusidic acid 10 mg spread in two bands on either side of the plate,
Chloramphenicol leaving a central band uninoculated. The test orga­
Enterobacteriaceae 30 mg nism is inoculated on central one-third and control
haemophili, pneumococci and 10 mg on upper and lower thirds of the plate. However, in
meningococci modified Stokes disk diffusion method, the test org­
Colistin 10 mg anism is inoculated in the upper and lower thirds
Nalidixic acid 30 mg and control on central one-third. An uninoculated
Nitrofurantoin 50 mg gap 2–3 mm wide should separate the test and
Sulfafurazole control areas on which antibiotic disks are applied
Enterobacteriaceae and 100 mg (Fig. 80.3). A maximum of six antibiotic disks can
enterococci 25 mg be accommodated on a single 100 mm diameter
meningococci
plate. Plates should be incubated in air at 35-37°C
Trimethoprim 2.5 mg overnight (ideally for 16–18 hours). Tests should not
Trimethoprim/sulfamethoxazole 1.2 mg/23.8 mg be read earlier.
Spectinomycin 100 mg
Vancomycin 30 mg Reading and Reporting Results
Rifampicin 5 mg Measure the inhi­bition zones of the control strain,
i.e. the distance in millimeters from the edge of the
Ciprofloxacin 1 mg
disk to the zone edge if that is obvious.
Mupirocin 5 mg
Each zone size is interpreted as follows:

www.ebook3000.com
Chap-80.indd 561 15-03-2016 11:26:49
562  |  Section 6: Miscellaneous
Table 80.4  Interpretation chart used in Kirby–Bauer disk diffusion method
Antibiotic* Diameter of zone inhibition (in mm)
Resistant Intermediate sensitive Sensitive
Benzylpenicillin ≤28 — ≥29
Ampicillin
Methicillin ≤9 10–13 ≥14
Carbenicillin
Gentamicin ≤12 — ≥13
Amikacin ≤14 15–16 ≥17
Erythromycin ≤13 14–17 ≥18
Tetracycline ≤14 15–18 ≥19
Chloramphenicol ≤12 13–17 ≥18
Nalidixic acid ≤13 14–18 ≥19
Trimethoprim ≤10 11–15 ≥16
Ciprofloxacin ≤15 16–20 ≥21
* Only limited antibiotics have been shown in the table.

should be reported as resistant to penicillin irre­


spective of zone size.
2. Motile organisms: Such as Proteus mirabilis
and P. vulgaris may swarm when growing on
agar surface resulting in a thin veil that may
penetrate into the zones of inhibition around
antibiotic disks. The zones of swarm­ing should
be ignored and the outer margin, which is
usually clearly outlined, should be measured.
3. Polymyxins: Diffuse poorly in agar so that
zones are small. In this case, by Stokes method,
report as:
Sensitive: Zone radius equal to, wider than, or
not more than 3 mm smaller than the control.
Resistant: Zone radius more than 3 mm smaller
Fig. 80.3: Stokes disk diffusion method than the control.
Categories of sensitivity: Three categories of 4. Zones around ciprofloxacin: Zones around
sensitivity can be recognized: ciprofloxacin disks are large with some control
1. Sensitive: The zone size of the test strain is strains. These are interpreted as follows:
larger than, equal to or not more than 3 mm a. When sensitive control used is Staphylo­
smaller than that of the control strain. coccus aureus or Pseudomonas aeruginosa.
2. Intermediate: The zone size of the test strain is Sensitive: Inhibition zone of the test
at least 2 mm, but also 3 mm smaller than that bacterium is equal to, greater than, or not
of the control strain. more than 7 mm smaller than that of control.
3. Resistant: The zone size of the test strain is Intermediate sensitive: Inhibition zone of
smaller than 2 mm. the test bacterium is more than 2 mm but
is smaller than that of the control by more
Exceptions than 7 mm.
1. Penicillinase-producing strain of Staphylo­ Resistant : Inhibition zone of the test
coccus: It will show an inhibition zone but, it will bacterium is 2 mm or less.
be smaller and colonies at the edge are large and b. When sensitive control used is Esch. coli or
well developed and there is no gradual fading of H. influenzae.
growth towards the disk. These penicillinase- Sensitive: Inhibition zone of the test bacterium
producing staphylococci show heaped­up zone is equal to, greater than, or not more than 10
edges in tests of penicillins; accordingly they mm smaller than that of the control.

Chap-80.indd 562 15-03-2016 11:26:50


Chapter 80: Laboratory Control of Antimicrobial Therapy  | 563
Intermediate sensitive: Inhibition zone of growth of an organism after overnight incubation.
the test bacterium is more than 2 mm but The minimum bactericidal concentration (MBC) is
is smaller than that of the control by more the amount of agent that will prevent growth after
than 10 mm. sub­culture of the organism to antibiotic-free medium.
Resistant : Inhibition zone of the test
bacterium is 2 mm or less. DILUTION METHODS
5. Methicillin-resistant Staphylococcus aureus Dilution tests may be done by the tube dilution or
(MRSA): Will often appear fully sensitive when agar dilution methods.
tested in ordinary way. Many of these organisms
grow more slowly in the presence of methicillin
Broth Dilution Method
and growth will only appear within the zone when
the incubation is continued for 48 hours. This Serial dilutions of the drug in Mueller–Hinton broth
difficulty can be overcome either by incubat­ing are taken in tubes and a standardized suspension
the culture at 30°C or test may be reliable when of the test bacterium inoculated. The inoculum is
incubated at 37°C if 5% NaCl has been added to prepared as in case of disk diffusion methods by
the medium. comparing with 0.5 McFar­land opacity standard.
An organism of known sensitivity should also be
6. Trimethoprim and sulfamethoxazole
titrated. Incubate at 35–37°C for 16–18 hours and
disks: For sen­sitivity tests trimethoprim and
read the results. Incubate at 30°C for determination
sulfamethoxazole disks containing both drugs
of MIC of methi­cillin (Fig. 80.5).
are widely used. Such disks may be misleading,
MIC is the lowest concentration of antimicrobial
it is impossible to know whether the organism
agent at which there is no visible growth. For deter­
is sensitive to both or only to one of them
mination of MBC, subculture from each tube show­
when both drugs are present. To overcome this
ing no growth over a quarter of a nutrient medium
problem, each drug should, therefore, be tested
free from antimicrobial agent. Incubate and exam­
separately.
ine them for growth. The tube containing lowest
concentration of the antimicrobial agent that fails
Primary Sensitivity Tests to yield growth, on subculture, is the MBC of the
In primary sensitivity tests the specimen serves as antimicrobial agent for the test strain. MIC inhibits
the inoculum. A portion of it is spread uniformly the bacterial growth while MBC kills the bacterium.
over part or whole of the primary culture plate and
antibiotic disks are applied. Principal Uses of MIC
In practice, it appears that primary sensitivity
1. In the determination of antibiotic sensitivities
tests are of most value for specimens of urine,
of organisms from patients with serious
of some value for swabs or pus from patients
infections, e.g. infective endocarditis.
attending acci­dent and emergency departments.
2. In measuring the antimicrobial sensitivities of
slow-growing organisms, e.g. Mycobacterium
Epsilometer or E-test
tubercu­losis).
The E-test, a modification of the disk diffusion
test, utilizes a strip impregnated with a gradient of
concentrations of an antimicrobial drug. Multiple
strips, each containing a different drug, are placed
on the surface of an agar medium that has been
uniformly inoculated with the test organism so that
they extend out radially from the center (Fig. 80.4).
Each strip contains a gradient of an antibiotic
and is labeled with a scale of minimal inhibitory
concentration values. The lowest concen­tration in
the strip lies at the center of the plate. After 24–48
hours of incubation, an elliptical zone of inhibition
appears. The MIC is determined by reading a
number off the numerical scale printed on the strip
at the point where the bacterial growth intersects it.

Minimum Inhibitory and Bactericidal


Concentrations Fig. 80.4: E-test (Courtesy Dr Krishna Prakash, Ex-Director
The minimum inhibitory concentration (MIC) is the Professor, Department of Microbiology, Maulana Azad Medical
least amount of antimicrobial that will inhibit visible College, New Delhi, India)

www.ebook3000.com
Chap-80.indd 563 15-03-2016 11:26:50
564  |  Section 6: Miscellaneous

Fig. 80.5: Broth dilution methods showing MIC and MBC

3. Reference points in the evaluation and com­ in controls against many variables and therefore
parison of new and existing antimicrobial provides dependable results.
agents. ™™ Dilution tests: There are two types of dilution tests:
Broth dilution method and agar dilution method.
Agar Dilution Method
Here, serial dilutions of the drug are prepared in IMPORTANT QUESTIONS
agar (Mueller–Hinton agar) and poured into plates.
1. Name different methods of antibiotic sensitivity
The ‘agar dilution’ method is more convenient when testing. Discuss in detail Kirby–Bauer disk diffusion
several strains are to be tested at the same time. The method for antimicrobial sensitivity testing.
advantage is that many strains can be inoculated on 2. Write short notes on:
each plate containing an antibiotic dilution. a. Kirby–Bauer disk diffusion method
Automated versions of sensitivity tests are b. Stokes disk diffusion method
available and are in use in large laboratories. c. Minimum inhibitory concentration of antimi­
crobial agents.
ANTIBIOTIC ASSAYS IN BODY FLUIDS d. Minimum bactericidal concentration of antimi-
crobial agents.
These are required to verify whether adequate drug
concentrations are achieved in blood and other MULTIPLE CHOICE QUESTIONS (MCQs)
body fluids. Likewise, new drugs must be tested to
determine achievable levels in the blood, urine, or 1. Which of the following media is most suitable for
antibiotic sensitivity testing?
other body fluids.
The assays are generally done by making serial a. Mueller–Hinton medium b. Nutrient agar
dilutions of the specimen and inoculating standard c. Blood agar d. MacConkey agar
suspensions of bacteria of known MIC. Assays 2. The pH of the medium for antibiotic sensitivity
by the agar diffusion method can also be done. testing should be:
A technique called the diffusion assay is used to a. 6.0–6.2 b. 6.8–7.0
measure the concentration of an antimicrobial c. 7.2–7.4 d. 7.6–7.8
in a fluid specimen. The test relies on the same 3. In Stokes disk diffusion method, if the zone size
principle as the Kirby–­Bauer test, except in this case is larger than, equal to or not more than 3 mm
it is the concentration of drug, not the sensitivity of smaller than the control a strain is considered:
organism, being assayed. This depends on the direct a. Sensitive b. Intermediate
relationship between antibiotic concentration c. Resistant d. None of the above
and the diameter of the zone of inhibition with a 4. Addition of 5% salt in the medium is done when
standard sensitive strain of bacterium. testing antibiotic susceptibility of :
a. Methicillin resistant staphylococci
Key Points b. Pneumococci
™™ Antibiotic susceptibility tests are of two types: c. Coliforms
Diffusion tests and dilution tests d. Non fermenting gram-negative bacilli
™™ Diffusion tests consists of Kirby–Bauer and Stokes
disk method. Stokes disk method incorporates built- ANSWERS (MCQs)
1. a; 2. c; 3. a; 4. a

Chap-80.indd 564 15-03-2016 11:26:51


81
Chapter

Antimicrobial Chemotherapy

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Describe mechanism of drug resistance
be able to: ∙∙ List cephalosporins of first, second, third and fourth
∙∙ Describe mechanism of action of antibacterial generation.
drugs

ANTIMICROBIAL AGENT Table 81.1  Mechanisms of antibacterial drug action


Antimicrobial agent is a chemical substance 1. Inhibitors of bacterial cell wall synthesis
inhibiting the growth or causing the death of a Penicillins
mi­croorganism. Cephalosporins
Vancomycin
ANTIBIOTIC Bacitracin
Antibiotic as origin­ally defined was a chemical Cycloserine
substance produced by various species of micro­ Fosfomycin
organisms that was capable of inhibiting the
2. Inhibitors of bacterial cytoplasmic membrane
growth or causing death of other microorganisms function
in low concentration. Polymyxins
Gramicidin
CHEMOTHERAPEUTIC AGENTS Tyrocidine
Chemotherapeutic agents are the chemical
3. Inhibition of bacterial nucleic acid synthesis
substances used to kill or inhibit the growth of
Quinolones
microorganisms already estab­lished in the tissues
Rifamycins
of the body. Nowadays the term antibiotic is
Nitroimidazoles
used loosely to describe agents (mainly, but not
Nitrofurans
exclusively, antibacterial agents) used to treat
Novobiocin
systemic infection.
Antimicrobial agent (AMA): The term Anti­ 4. Inhibition of bacterial protein synthesis
microbial agent (AMA) is to designate synthetic Aminoglycosides
Chloramphenicol
as well as naturally obtained drugs that attenuate
Tetracyclines
microorganisms.
Macrolides
Antiseptics or disinfectants: Antimicrobial Lincosamides
substances that are too toxic to be used other Fusidic acid
than in topical therapy or for environmental Streptogramins
decon­tamination are referred to as antiseptics or Mupirocins
disinfectants.
5. Metabolic antagonism
ANTIBACTERIAL AGENTS Sulfonamides
Trimethoprim
The principal types of antibacterial agents are
Dapsone
listed in Table 81.1. These have been grouped
Isoniazid
according to their site of action.

www.ebook3000.com
Chap-81.indd 565 15-03-2016 11:28:09
566  |  Section 6: Miscellaneous
MECHANISMS OF ACTION OF 2. Those with high activity against both gram-­
ANTIBACTERIAL DRUGS positive and gram-negative organisms
but des­troyed by b-lactamases: Ampicillin;
Mechanisms of action of antibacterial agents can amoxicillin; carbenicillin; ticarcillin; piperacillin.
be placed under the headings: 3. Those stable in gastric acid and suitable for
1. Inhibition of bacterial cell wall synthesis. oral administration: Penicillin-V; cloxacillin;
2. Inhibition of bacterial cytoplasmic membrane ampicillin.
function. 4. b-Iactamase resistant penicillins: Methicillin;
3. Inhibition of bacterial nucleic acid synthesis. nafcillin; oxacillin; cloxacillin; dicloxacillin;
flucloxacillin.
4. Inhibition of bacterial protein synthesis.
5. Penicillins active against Pseudomonas:
Apalcillin, carbenicillin; ticarcillin; azlocillin;
1. Inhibititon of Bacterial Cell Wall mezlocillin; piperacillin.
Synthesis
Inhibitors of bacterial cell wall synthesis act on Cephalosporins
the formation of the peptidoglycan layer. Bacteria Cephalosporins are a family of antibiotics and
that lack peptidoglycan, such as mycoplasmas, are their b-lactam structure is very similar to that of
resistant to these agents. the penicillins (Figs 81.1A and B). In place of
The anti­b iotics which inhibit cell wall 6-aminopenicillanic acid, they have a nucleus of
synthesis are b-Iactam antibiotics (penicillins and 7-aminocephalosporanic acid. They are broad-
cephalosporins), glyco­peptides, bacitracin, cyclo­ spectrum drugs frequently given to patients with
serine, fosfomycin and isoniazid. penicillin allergies.
They are grouped as the first, second, third, and
A. b-Iactam Agents fourth ­generation cephalosporins. These include
This group includes penicillins, cephalosporins cephalexin and cephradine (first generation),
and other compounds that feature a b-lactam ring cefaclor and cefprozil (second gen­e ration),
in their structure. All these compounds bind to cefixime and ceftbuten (third generation), and
proteins situated at the cell wall–cell membrane cefepime (fourth generation) (Table 81.2).
interface. These penicillin-binding proteins (PBPs) First-generation cephalosporins are more
are involved in cell wall con­struction, including effective against Gram-positive than gram-negative
the cross-linking of the peptidoglycan strands that
gives the wall its strength. Opening of the b-lactam
ring by hydrolytic enzymes, collectively called
b-lactamases, abolishes antibacterial activity. Many
such enzymes are found in bacteria.

Penicillins
Penicillins are a group of antimicrobial substances,
all of which possess a common chemical nucleus
(6-aminopenicillanic acid) which contains a
b-­lactam ring essential to their biologic activity. A
side chain is attached to the b-lactam ring which
determines many of the antibacterial and pharma­
cological characteristics of a particular type of
peni­cillin.
All b-lactam antibiotics inhibit formation of
bacterial cell wall. They particularly block the
final transpeptidation reac­tion in the synthesis of
cell wall peptidoglycan and also activate autolytic
enzymes in the cell wall.
Penicillins can be divided into several groups:
1. Those with highest activity against gram-posi­
tive organisms but susceptible to hydrolysis by Figs 81.1A and B: The β-lactam ring of penicillins and
b-lactamases: cephalosporins the core chemical structure of (A) a penicillin
(B) a cephalosporin. The β-lactam rings are marked by an
• Penicillin-G orange circle. The R groups vary among different penicillins
• Penicillin-V and cephalosporins

Chap-81.indd 566 15-03-2016 11:28:09


Chapter 81: Antimicrobial Chemotherapy  | 567
Table 81.2  Important cephalosporins and their antibacterial spectrum
Class Compounds Antibacterial spectrum
First generation Cephalothin S. aureus, streptococci (other than enterococci), E. coli, Klebsiella
Cephalexin H. influenzae and P. mirabilis
Second generation Cefamandole First generation spectrum expanded to indole positive Proteus, Enterobacter,
Cefotaxime Citrobacter, Serratia and many gram-negative anaerobes
Third generation Cefotaxime Spectrum of second generation expanded to give high activity against
Cefoperazone H. influenzae, gonococci including b-lactamase producing strains and activity
against P. aeruginosa and many gram-negative anaerobes
Fourth generation Cefepime Spectrum similar to that of third generation compounds, but highly resistant to
Cefpirome b-lactamases, hence active against many bacteria resistant to the earlier drugs. P.
aeruginosa is also inhibited by cefepime

pathogens. Second ­generation drugs act against subunit of DNA gyrase, an enzyme that engineers
many gram-negative as well as Gram-positive the breaking and rejoin­ing of super coiled DNA.
pathogens. Third-generation drugs are particularly Nalidixic acid and its early congeners are narrow-
effective against gram-negative pathogens, and spectrum agents active only against gram-negative
often also reach the central nervous system. bacteria.
Newer quinolones like ciprofloxacin, norflox­
Other b-Lactam Antibiotics acin, ofloxacin, pefloxacin and lomefloxacin are
Two other groups of β-lactam drugs, carbapenems broad spectrum quinolones. These have been
and monobactams, are very resistant to β-lactamases. success­fully used in a wide variety of infections, but
resist­ance is becoming more prevalent.
Carbapenems
The carbapenems are effective against a wide range Rifamycins
of gram-negative and gram-positive bacteria. Two Rifamycins inhibit bacterial growth by binding
types are available, imipenem and meropenem. strongly to the DNA-dependent RNA polymerase of
Glycopeptides bacteria thus inhibiting transcription of RNA from
DNA. This group of antibiotics is characterized
Two glycopeptides, vancomycin and teicoplanin,
by excellent activity against mycobacteria.
are in clinical use. Their chief importance resides
Staphylococci in particular are exquisitely sensitive.
in their action against gram-positive cocci with
Rifampicin and rifabutin are most widely used.
multiple resistance to other drugs. They are mainly
used in serious infections with staphylococci and
Nitroimidazoles
enterococci that are resistant to other drugs.
Azole derivatives have wide-ranging antimicrobial
Other Inhibitors of Bacterial Cell Wall activity against fungi, protozoa, helminths, as well
Synthesis as bacteria. Those that exhibit antibacterial activity
These include bacitracin, cycloserine, fosfomycin are 5-nitroimidazoles which are active only against
and isoniazid. anaerobic bacteria and anaerobic protozoa. The
representative of the group most commonly used
2. Inhibition of Bacterial Cytoplasmic clini­cally is metronidazole, but similar derivatives
Membrane Function include tinidazole, ornidazole and nimorazole.
They are primarily anti­protozoal agents, but they
Only polymyxins have been regularly used
exhibit potent activity against anaerobic bacteria.
systemically among membrane active agents used
in human medicine. Two members of the family Nitrofurans
are in therapeutic use: poly­myxin B and colistin
(polymyxin E). Various nitrofuran derivatives are in use around
They exhibit potent antipseudomonal activity, the world as antibacterial agents. These include
but toxicity has limited their usefulness, except in nitro­furantoin and furazolidone.
topical preparations and bowel decontamination Nitrofuran­toin: It is bacteri­cidal to most urinary
regimens. pathogens at concentrations achievable in urine.
Furazolidone: Furazolidone is used in enteric
3. Inhibitors of Nucleic Acid Synthesis
infections. The mode of action of nitrofurans
Quinolones has not been elucidated, but it is probable that
The quinolones are syn­thetic drugs that contain a reduced metabolite acts on DNA in a manner
the 4-quinolone ring. Quinolones act on the α analogous to that of the nitroimidazoles

www.ebook3000.com
Chap-81.indd 567 15-03-2016 11:28:10
568  |  Section 6: Miscellaneous
Novobiocin Macrolides
Novobiocin acts on the β subunit of bacterial DNA The macrolides reversibly bind to the 50S ribosomal
gyrase. It is quite active against staphylococci and subunit and prevent the continuation of protein
streptococci, but is no longer favored because of synthesis. Macrolides include erythromycin,
problems of resistance and toxicity. Staphylococ­cus azithromycin, clarithromycin, dirithromycin and
saprophyticus is novobiocin resistant. spiramycin.
Macrolides as a group are effective against a
4. Inhibition of Bacterial Protein Synthesis variety of bacteria, including many gram-positive
organisms as well as the most common causes of
The major classes of antibiotics that inhibit atypical pneumonia (walking pneumonia). They
protein syn­thesis are the aminoglycosides, the often serve as the drug of choice for patients who
tetracyclines, and the macrolides. Others include are allergic to penicillin.
the lincosamides and chlorampheni­col. Of these,
only the aminoglycosides are bactericidal; the Lincosamides
oth­ers are all bacteriostatic. Two classes of drugs
Lincosamides bind to the 50S ribosomal subunit
that have recently been approved for use are
and resemble macrolides in binding site,
the oxazolidi­nones and the streptogramins. A
antibacterial activity, and mode of action.
synergistic combination of two streptogramins is
bactericidal against some organisms.
Fusidic Acid
It blocks factor G, which is involved in peptide
Aminoglycosides
elongation. It has excellent activity against
The aminoglycosides irreversibly bind to the staphylococci, good activity against Corynebacterium
30S ribosomal sub­unit, causing it to distort and diphtheriae and modest activity against streptococci,
malfunction. This blocks the ini­tiation of translation Gram-nega­tive anaerobes, Nocardia asteroides, and
and causes misreading of mRNA by ribosomes that Myco­bacterium tuberculosis.
have already passed the initiation step. Examples of
aminoglycosides include streptomycin, kanamycin, Streptogramins
neo­mycin, gentamicin, tobramycin, amikacin, etc.
Derivatives suit­able for parenteral administration,
Use: They are bactericidal compounds, and some, quinupristin and dalfopristin, have been developed
notably gentamicin and tobramycin, exhibit good as a combination product. The combination is
activity against Pseudomonas aeruginosa. The effective against a variety of gram-positive bacteria,
group also has in common a ten­dency to damage including some of those that are resistant to
the eighth cranial nerve (ototoxicity) and the b-lactam drugs and vancomycin.
kidney (nephrotoxicity).
Mupirocin
Chloramphenicol This is an antibiotic, produced by Pseudomonas
fluorescens. Its useful activity is restricted to
Chloramphenicol binds to the 50S ribosomal
staphylococci and streptococci and used only in
subunit. It is mainly bacteriostatic.
topical preparations.
Use: Use of chlo­ramphenicol has been limited to
typhoid fever, meningi­tis and a few other clinical 5. Metabolic Antagonism
indications because of the occurrence of a rare but Antibacterial medications that inhibit metabolic
fatal side-effect, aplastic anemia. pathways.

Tetracyclines Sulfonamides and Diaminopyrimidines


The tetracyclines reversibly bind to the 30S These agents affect DNA synthesis because of their
ribosomal subunit, blocking the attachment of tRNA role in folic acid metabolism.
to the ribosome and prevent­ing the continuation of
protein synthesis. Doxycycline and minocycline are Mechanism of Action of
in most common use. Sulfonamides (Fig. 81.2)
Tetracyclines are broad-spectrum agents with Sulfonamides are ana­logs of para-aminobenzoic
important activity against chlamydiae, rickettsiae, acid. Be­c ause of their similar structures
mycoplasmas and, sur­prisingly, malaria parasites, (Fig. 81.3), there occurs competition between the
as well as most conventional gram-positive and sulfonamides and the PABA for the active site on
Gram-negative bacteria. Tetracyclines can cause the surface of the enzyme initiating the conversion
discoloration in teeth when used by young children. of PABA to dihy­drofolic acid. When sulfonamides

Chap-81.indd 568 15-03-2016 11:28:10


Chapter 81: Antimicrobial Chemotherapy  | 569
spp. and Toxoplasma gondii. Sulfa­d oxine or
sulfadiazine combined with pyrimetha­mine are
used in malaria and toxoplasmosis respectively.

ANTIBIOTIC RESISTANCE
Understanding the mechanisms and the spread
of antimicrobial resistance is an important step in
curtailing the problem.

Mechanisms of Drug Resistance


A. Acquired Antimicrobial Resistance
1. Drug-inactivating Enzymes
Fig. 81.2: Mechanism of action of sulfonamides Some organisms produce enzymes that chemically
modify a spe­cific drug in such a way as to render it
ineffective.
i. Penicillinase: The best-known example is
the hydrolysis of the β-lactam ring of many
penicillins by the enzyme penicillinase.
ii. Chloramphenicol acetyl transferase: The
enzyme chloramphenicol acetyl trans­ferase
chemically alters the antibiotic chloram­
phenicol.
Fig. 81.3: Structure of p-aminobenzoic acid and basic ring 2. Alteration in the Target Molecule
of sulfonamides Examples
i. Alterations in the penicillin-binding proteins
enter into reac­tion in place of PABA, nonfunctional prevent b-lactam drugs from binding to them.
analogs of folic acid are formed, preventing ii. Similarly, a change in the ribosomal RNA, the
further growth of bacterial cell. Human tissue cells target for the macrolides, prevents those drugs
and sulfonamide ­resistant bacteria also require from interfering with ribosome function.
folic acid for synthe­sis of nucleic acids but are
capable of taking up preformed folic acid from the 3. Decreased Uptake of the Drug
environment and their growth is independent of Many gram-negative bacteria are unaffected
the conversion of PABA to folic acid. Other analogs by penicillin G because it cannot penetrate
of PABA are diami­nodiphenylsulfone (dapsone) the envelope’s outer mem­brane. A decrease in
and p-aminosalicy­lic acid (PAS) which are active permeability can lead to sulfonamide resistance.
against lepra and tubercle bacilli, respectively. Mycobacteria resist many drugs because of the high
Diaminopyrimidines, which include the broad- content of mycolic acids in a complex lipid layer out­
spectrum antibacterial agent trimethoprim and side their peptidoglycan.
the antimalarial compounds pyrime­thamine and 4. Increased Elimination of the Drug
cycloguanil (the metabolic product of proguanil), The systems that bacteria use to transport detrimental
prevent the reduction of dihydrofolate to com­pounds out of a cell are called efflux pumps. This
tetrahydrofolate. Sulfonamides and diaminopyrim­ resistance strategy is to pump the drug out of the cell
idines thus act at sequential stages of the same after it has entered.
metabolic pathway and interact synergically, but
in bac­terial infections trimethoprim is generally B. Genetic Basis of Antibiotic Resistance
sufficiently effective, and less toxic, when used
Antimicrobial resistance can be due to either
alone.
spontaneous mutation, which alters existing genes,
Sulfonamides are broad-spectrum antibacterial
or acquisition of new genes.
agents, predomi­n antly bacteriostatic, but
resistance is common and the group also suffers 1. Spontaneous Mutation
from problems of toxicity. They are now lit­tle used Drugs such as streptomycin used in tuberculosis
alone, but the combination of sulfametho­xazole treatment to which single point muta­t ions
with trimethoprim (cotrimoxazole) is still widely can confer resistance are sometimes used in
used, notably in the prophylaxis and treat­ment of combination with another drug to prevent survival
Pneumocystis carinii pneumonia. They have some of resistant mutants. The chance of an organism
activity against protozoa, including Plas­modium simultaneously developing mutational resistance

www.ebook3000.com
Chap-81.indd 569 15-03-2016 11:28:11
570  |  Section 6: Miscellaneous
to each drug is extremely low. If any organism
™™ Antimicrobial resistance can be due to either
spontaneously devel­ops resistance to one drug, spontaneous muta­tion, or acquisition of new genes
the other drug will still kill it. ™™ The most common mechanism of transfer of resis­
tance is through the conjugative transfer of R
2. Gene Transfer plasmids (resistance plasmids).
i. Conjugation: R plas­mids frequently carry
several different resistance genes, each one
mediating resistance to a specific antimicrobial
IMPORTANT QUESTIONS
drug. Thus, when an organism acquires an R 1. Define the terms antimicrobial agent, chemothera­
plasmid, it acquires resistance to several different peutic agent and antibiotic. Describe the various
medications simultaneously. mecha­nisms of action of antibiotics with examples.
ii. Transduction: Acquisition of resistance by 2. Describe the structure and functions of bacterial
cell wall. Name various antibiotics which affect cell
transduction is common in staphylococci.
wall synthesis.
The penicillin plasmids (carrying gene for 3. Write short notes on:
β-lactamase production) enclosed in a a.  Antibiotics inhibiting bacterial cytoplasmic
bacteriophage is transferred from a penicillin- membrane function.
resistant staphylococcus to a susceptible b. Antibiotics inhibiting bacterial nucleic acid
staphylococcus. synthesis.
iii. Transformation: Resistance transfer can c. Antibiotics that act as metabolic antagonists.
be demonst­r ated experimentally but its 4. Discuss genetic basis of drug resistance in bacteria.
significance is not known.
iv. Transposons: Many of the resistance genes MULTIPLE CHOICE QUESTIONS (MCQs)
on R plasmids are carried on transposons that 1. Which of the following antibiotics act/s by
can move from a plasmid to the chromosome, inhibiting cell wall synthesis?
from one plasmid to another, or from the a. Penicillins b. Vancomycin
chromosome to a plasmid. Thus, if one organism c. Bacitracin d. All of the above
has two dif­ferent plasmids, an antibiotic- 2. Which of the following antibiotics acts by inhibiting
resistance gene can move from one to the other. cytoplasmic membrane:
a. Polymyxin b Thyrocidine
Key Points c. Gramicidin d. All of the above
™™ Antimicrobial agent is a chemical substance 3. Which antibiotic/s act/s by inhibiting bacterial
inhibiting the growth or causing the death of a mi­ nucleic acid synthesis?
croorganism a. Quinolones b. Novobiocin
™™ Antibiotic as origin­ally defined was a chemical c. Nitrofurans d. All of the above
substance produced by various species of 4. Which of the following antibiotics act/s by
microorganisms that was capable of inhibiting the inhibiting bacterial protein synthesis?
growth or causing death of other microorganisms a. Aminogycosides b. Tetracycline
in low concentration c. Macrolides d. All of the above
™™ Chemotherapeutic agents are the chemical 5. Which antibiotic/s act/s as metabolic antagonists
substances used to kill or inhibit the growth of
for their action?
microorganisms already estab­lished in the tissues
a. Sulphones b. Trimethoprim
of the body
c. Isoniazid d. All of the above
™™ Mechanisms of action’ of antibacterial agents can
be: 1. Inhibition of bacterial cell wall synthesis; 6. What is the genetic basis of drug resistance in
2. Inhibition of bacterial cytoplasmic membrane Mycobacterium tuberculosis?
function; 3. Inhibition of bacterial nucleic acid a. Transformation b. Transduction
synthesis; 4. Inhibition of bacterial protein synthesis c. Mutation d. Conjugation
and 5. Metabolic Antagonism 7. Staphylococcus aureus acquire drug resistance by:
™™ Mechanisms of drug resistance are: 1. Drug- a. Transformation b. Transduction
inactivating enzymes; 2. Alteration in the target c. Mutation d. Conjugation
molecule; 3. Decreased uptake of the drug; 4.
Increased elimination of the drug ANSWERS (MCQs)
1. d; 2. d; 3. d; 4. d; 5. d; 6. c; 7. b

Chap-81.indd 570 15-03-2016 11:28:11


82
Chapter

Immunoprophylaxis

Learning Objectives

After reading and studying this chapter, you should ∙∙ Explain immunization schedule
be able to: ∙∙ Discuss national immunization schedule
∙∙ List of immunizing agents ∙∙ Describe passive immunization.
∙∙ Describe the following: live attenuated vaccines;
killed vaccines; toxoids

Introduction Live vaccines should not be administered


to persons with immune deficiency diseases
An important contribution of microbiology to
or to persons whose immune response may be
medi­cine has been immunization, which is one of
suppressed.
the most effective methods of controlling infectious
In the case of live vaccines, immunization
diseases.
is generally achieved with a single dose. The
exception is polio vaccine. Live vaccines usually
Immunizing Agents produce a durable immunity.
The immunizing agents may be classified as:
A. Vaccines. 2. Killed (Inactivated) Vaccines
B. Immunoglobulins.
Organisms killed by heat or chemicals, when
infected into the body stimulate active immunity.
A. VACCINES They are usually safe but generally less efficacious
A vaccine (Latin vacca, cow) is a preparation than live vaccines. Killed vaccines usually require a
from an infectious agent that is administered to primary series of 2 or 3 doses of vaccine to produce
humans and other animals to induce protective an adequate antibody response, and in most cases
immunity against a given disease. It stimulates “booster” injections are required. Killed vaccines
the production of protective antibody and other are usually administered by subcutaneous or
immune mechanisms. Vaccines may be prepared intramuscular route. Examples of killed vaccumes
from live modified organisms, inactivated or killed are typhoid, cholera, pertussis, pneumococcal,
organisms, extracted cellular fractions, toxoids or rabies, hepatitis B and influenza vaccines.
combination of these. More recent preparations
are subunit vaccines and recombinant vaccines. 3. Toxoids
Certain organisms produce exotoxins, e.g.
Types of Vaccines diphtheria and tetanus bacilli. The toxins produced
1. Live Vaccines by these organisms are detoxicated and used in
Live vaccines are prepared from live (generally the preparation of vaccines. In general, toxoid
attenuated) organisms. In general, live vaccines preparations are highly efficacious and safe
are more potent immunizing agents than killed immunizing agents.
vaccines. Live attenuated example are BCG,
smallpox vaccine, oral polio vaccine (OPV), 4. Cellular Fractions
mumps, measles and rubella (MMR) vaccine and Vaccines, in certain instances, are prepared from
yellow feller vaccine. extracted cellular fractions, e.g. meningococcal

www.ebook3000.com
572  |  Section 6: Miscellaneous
vaccine from the polysaccharide antigen of the based on active immunization, e.g. polio,
cell wall, the pneumococcal vaccine from the tetanus, diphtheria and measles. Vaccination
polysaccharide contained in the capsule of the against these diseases is given as a routine
organism and hepatitis B polypeptide vaccines. during infancy and early childhood, with
periodic boosters to maintain adequate levels
5. Mixed or Combined Vaccine of immunity.
If more than one kind of immunizing agent is 2. Immunization of individuals or selected
included in the vaccine, it is called a mixed or groups: There are immunizations against certain
combined vaccine. The aim of combined vaccines diseases which are offered to high-risk groups or
is to simplify administration, reduce costs and restricted to definite geographic areas where the
minimize the number of contacts of the patient disease is endemic or a public health problem
with the health system. The following are some of (e.g. yellow fever).
the well-known combinations:
∙∙ DPT (Diphtheria-pertussis-tetanus) Immunization Schedules
∙∙ DT (Diphtheria-tetanus) National Immunization Schedule
The National Immunization Schedule is given in
∙∙ DP (Diphtheria-pertussis) .
Table 82.1. The first visit may be made when the
∙∙ DPT and typhoid vaccine infant is 6 weeks old; the second and third visits,
∙∙ MMR (Measles, mumps and rubella) at intervals of 1–2 months. Oral polio vaccine may
∙∙ DPTP (DPT plus inactivated polio) be given concurrently with OPT. BCG can be given
Plain vaccines are less efficacious than adjuvant with any of the three doses but the site for the
vaccines. Freeze-dried vaccines (e.g. BCG, yellow injection should be different. The schedule also
fever measles) are more stable preparations than covers immunization of women during pregnancy
liquid vaccines. against tetanus.

6. Recombinant-vector Vaccines Expanded Program on Immunization


Recently several microorganisms have been used In May 1974, the WHO officially launched a global
in the pro­duction of these recombinant-vector immunization program, known as Expanded
vaccines. Program on Immunization (EPI) to protect all
children of the world against six vaccine-preventable
Examples: Adenovirus, vaccinia virus, canarypox
diseases, namely diphtheria, whooping cough,
virus, attenuated poliovirus, and attenuated strains
tetanus, polio, tuberculosis and measles by the year
of Salmonella and Mycobacterium.
2000. EPI was launched in India in January 1978. This
is given in Table 82.2. The Program is now called
7. DNA Vaccines Universal Child Immunization Program, 1990.
A DNA vaccine elicits protective immunity against The Indian version is the Universal Immuniz­
a microbial pathogen by activating both branches ation Program.
of the immune system: humoral and cellular.
Examples: At present, there are human trials B. Passive Immunization
under way with several dif­ferent DNA vaccines Passive immunization is used when it is considered
against malaria, AIDS, influenza, hepatitis B, and necessary to protect a patient at short notice and
herpesvirus. Vaccines against a number of cancers for a limited period. Antitoxic, antibacterial or
(lym­phomas, prostate, colon) are also being tested. antiviral antibodies in human (homologous) or
animal (heterologous) se­rum are injected to give
IMMUNIZATION temporary protection.
Immunization is of three types: active immunization, Preparations for passive immunization: Three
passive immunization and combined passive and types of preparations are available for passive
active immunization. immunization:

A. Active Immunization 1. Normal Human Immunoglobulin


Active immunization is the protection of susceptible Normal human immunoglobulin is used to
humans from communicable diseases by the prevent measles in highly susceptible individuals
administration of vaccines (vaccination). and to provide temporary protection (up to 12
Immunoprophylaxis may be in the form of: weeks) against hepatitis A infection for travellers
1. Routine immunization: There are some to endemic areas and to control institutional and
infectious diseases whose control is solely household outbreaks of hepatitis A infection.
Chapter 82: Immunoprophylaxis  | 573
Table 82.1  National immunization schedule 3. Antisera
a. For infants The term antiserum is applied to materials prepared
At birth – BCG and OPV-O dose in animals. Originally passive immunization
(for institutional – BCG (if not given at birth) was achieved by the administration of antisera
deliveries – DPT-1 and OPV-1 or antitoxins prepared from non­human sources
At 6 weeks – DPT-2 and OPV-2
At 10 weeks – DPT-3 and OPV-3
such as horses. Since human immunoglobulin
At 14 weeks – Measles preparations exist only for a small number of
At 9 months diseases, antitoxins prepared from nonhuman
b. At 16–24 months – DPT and OPV sources (against tetanus, diphtheria, botulism, gas
gangrene and snake bite) are still the mainstay of
c. At 5–6 years – DT—the second dose of DT
should be given at an interval passive immunization.
of one month if there is no Administration of antisera may occasionally
clear history or documented give rise to serum sickness and anaphylactic shock
evidence of previous due to abnormal sensitivity of the recipient. The
immuanization with DPT
current trend is in favor of using immunoglobulins
d. At 10 and 16 years – Tetanus toxoid—The second wherever possible.
dose of TT vaccine should be
given at an interval of one
month if there is no clear history C. Combined Passive and
or documented evidence of Active Immunization
previous immunization with
DPT, DT or TT vaccines In some diseases (e.g. tetanus, diphtheria, rabies)
passive immunization is often undertaken in
e. For pregnant
women conjunction with inactivated vaccine products, to
provide both immediate (but temporary) passive
Early in – TT-1 or Booster
pregnancy immunity and slowly developing active immunity.
One month after Individual Immunization
TT-1–TT-2
Vaccines offered under national programs are
Note: i. Interval between 2 dose should not be less
than one month.
limited by economic considerations and so some
II. Minor cough, cold and mild fever are not a important vac­c ines may be omitted because
contraindication to vaccination. they are costly. These may be supplemented by
iii. In some states, hepatitis B vaccine is given as individual initiative, whenever pos­sible.
routine immunization.
Hepatitis B vaccine: Three doses of killed vaccine
are given at 0,1 and 6 months intramuscularly
Table 82.2  World health organization (WHO) EPI into the deltoid or, in infants into the anterolateral
immunization schedule (when early protection is aspect of thigh.
a must) Typhoid vaccine: Two re­cent typhoid vaccines,
the live oral galactose epimeraseless (Gal-E)
Age Vaccine
mutant vaccine and the injectable purified Vi
Birth BCG, oral polio polysaccharide vaccine are recommended for
6 weeks DPT, oral polio immunization of those five years old or above and
10 weeks DPT, oral polio so may be employed at school entry.
14 weeks DPT, oral polio
Key Points
9 months Measles
™™ Immunoprophylaxis is the prevention of disease by
the production of active or passive immunity
™™ The immunizing agents are vaccines and immunoglo­
2. Specific Human Immunoglobulin bulins
These preparations are made from the plasma ™™ Active immunization can be achieved by natural
of patients who have recently recovered from an infection with a microorganism administration of
a vaccine
infection or are obtained from individuals who
™™ Vaccines may be live attenuated, killed, or in the form
have been immunized against a specific infection. of toxoids
They therefore have a high antibody content against ™™ Live attenuated vaccines—BCG, smallpox vaccine,
an individual infection and provide immediate oral polio vaccine (OPV), mumps, measles, and
protection, e.g. for passive immunization against rubella (MMR) vaccine, and yellow fever vaccine are
tetanus (human tetanus immu­noglobulin, HTIG), some of the examples of live vaccines
™™ Killed inactivated vaccines—Typhoid, cholera,
hepatitis B immunoglobulin (HBIG), rabies (HRIG),
pertussis, pneumococcal, rabies, hepatitis B, and
varicella-zoster (ZIG) and vaccinia (AVIG).

www.ebook3000.com
574  |  Section 6: Miscellaneous
2. An example of killed inactivated vaccine is:
influenza vaccines are the examples of killed inacti­
vated vaccines a. MMR vaccine
™™ Toxoids: Tetanus toxoid and diphtheria toxoids are b. Influenza vaccines
two most widely used toxoids for immunization c. Oral polio vaccine
™™ Subunit vaccines: Hepatitis B subunit vaccine d. BCG vaccine
™™ Passive immunization is carried out by administration 3. An example of live attenuated vaccine is:
of human and animal sera a. Hepatitis B vaccine
™™ Combined active and passive immunization is often b. Rocky Mountain spotted fever vaccine
carried out to confer slowly developing immunity
c. Yellow fever vaccine
and immediate passive immunity, respectively,
against certain diseases, such as diphtheria, tetanus,
d. Rabies vaccine
and rabies. 4. Which of the following vaccines is/are subunit
vaccine/s?
a. Hepatitis B vaccine (plasma derived)
Important question b. Vi typhoid fever vaccine
rite short notes on:
W c. Meningococcal vaccine
a. Vaccines d. All of the above
b. Live attenuated vaccines 5. Toxoid is used for active immunization against:
c. Killed vaccines a. Pertussis b. Diphtheria
d. Toxoids c. Typhoid fever d. Tuberculosis
e. National Immunization Schedule 6. Specific immunoglobulins are available for passive
f. WHO EPI Immunization Schedule. immunization against:
a. Tetanus b. Rabies
Multiple choice questions (MCQs) c. Hepatitis B d. All of the above
7. Combined passive and active immunization is
1. Which of the following vaccines is/are killed available against all the diseases except:
vaccines? a. Poliomyelitis b. Tetanus
a. Cholera vaccine c. Rabies d. Diphtheria
b. Pertussis vaccine
c. Japanese encephalitis vaccine Answers (MCQs)
d. All of the above 1. d; 2. b; 3. c; 4. d; 5. b; 6. d; 7. a
8383
Chapter

Bacteriology of Water, Milk and Air

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe the following: Water-borne pathogens;
be able to: presumptive coliform count; Eijkman test
∙∙ Describe bacteriological examination of water ∙∙ Describe settle plate method and slit sampler
∙∙ Discuss bacteriological examination of milk method for bacteriology of air.

BACTERIOLOGY OF WATER 2. Salinity: In saline water number of bacteria


is less as compared to fresh water.
Introduction 3. Mineral springs: These are usually pure and
Safe and wholesome water is defined as water that most of the organisms found in water derived
is free from pathogenic agents, free from harmful from them come from imperfectly sterilized
chemical substances, pleasant to the taste and usable bottles.
for domestic purposes. The aim of microbiological 4. Nutrition: When organic matter is plentiful,
examination of water supplies is to detect whether organisms are abundant, when it is scarce
pollution of the water by pathogenic organisms has they are few, and tend to die out.
occurred or not. Reliance is placed on testing the 5. Temperature: When nutrition is available,
supply for microorganisms which indicate that fecal rise in temperature leads to multiplication,
pollu­tion has taken place. oth­e rwise, the number decreases. Low
temper­ature favors survival of bacteria.
BACTERIAL FLORA IN WATER 6. Light: Day light is bactericidal.
7. Acidity: Acidity of water has a bactericidal
Bacterial flora can be divided into three groups:
action.
1. Natural water bacteria.
8. Dissolved oxygen: It is essential for survival
2. Soil bacteria.
of aerobes.
3. Sewage bacteria: Bacterial flora in water is
given in Table 83.1. 9. Protozoal content: Certain flagellates exter­
minate bacteria in water and bring down their
FACTORS DETERMINING THE NUMBER number.
10. Rain: Early rain washes large number of
OF BACTERIA IN WATER
bacteria from the soil which may contaminate
1. Surface or deep water: Surface water is more water sources. Subsequent rains dilute the
likely to be contaminated. bacterial population.

Table 83.1  Bacterial flora in water


Natural water bacteria Micrococcus, Pseudomonas, Serratia, Flavobacterium, Chromobacterium, Acinetobacter and
Alcaligenes
Soil bacteria Bacillus subtilis, B. megaterium, B. mycoides, Enterobacter aerogenes and E. cloacae
Sewage bacteria Escherichia coli, Enterococcus faecalis, Clostridium perfringens, Salmonella Typhi and Vibrio
• Intestinal bacteria cholerae
• Sewage bacteria Proteus vulgaris, Clostridium sporogenes, Zoogloea ramigera, Sphaerotilus natans, Haliscomeno­
proper bacter hydrossis, Nostocoida limicola, Microthrix parvicella, Hexiaecter, Micro­scilla and Nocardia

www.ebook3000.com
576  |  Section 6: Miscellaneous
Table 83.2  Water-borne diseases BACTERIOLOGICAL EXAMINATION OF
1. Those caused by the presence of an infective agent: WATER
a.  Viral: Viral hepatitis A, hepatitis E, poliomyelitis,
The following tests are generally done for routine
rotavirus diarrhea in infants
b.  Bacterial: Typhoid and paratyphoid fever, bacteriological analysis of water.
bacillary dysentery, Escherichia coli diarrhea,
cholera a. Multiple Tube Test
c. Protozoal: Amebiasis, giardiasis 1. Total coliform count or presumptive coliform
d.  Helminthic: Roundworm, threadworm, hydatid
count.
disease.
e. Leptospiral: Weil’s disease 2. Fecal coliform and confirmed Escherichia coli
2. Those due to the presence of an aquatic host count: Eijkman test.
a. Snail: Schistosomiasis 3. Count of fecal streptococci.
b. Cyclops: Guinea worm, fish tape worm 4. Count of Clostridium perfringens.

11. Storage: Storage of water decreases bacterial b. Membrane Filtration Tests


count due to sedimentation and devitalization. A. Plate Count
The plate count expresses the number of all colony
Water-borne Pathogens ­forming bacteria in 1 ml water. It is of limited value
Water-borne diseases are give in Table 83.2. by itself.

COLLECTION OF WATER SAMPLES B. Counting of Indicator Organisms


Two methods are available for this purpose, the
For collection, use heat-sterilized bottles containing
multiple tube method and the membrane filtration
a sufficient volume of sodium thiosulfate to
method.
neutralize the bactericidal effect of any chlorine
or chloramine in the water. Each bottle of 100 ml a. Multiple tube test
capacity should contain 0.1 ml of a fresh 1.8%
(w/v) aqueous solution of sodium thiosulfate. 1. Total Coliform Count or Presumptive
Coliform Count
1. Sampling from a Tap or Pump Putlet Indicator medium: The indicator medium used
When collecting the sample from taps, exercise most has been MacConkey broth containing
extreme care to avoid contaminating it with bacteria bromocresol purple as indicates the for­mation
from the environment. Allow water to run to waste of acid from the lactose in the broth. An inverted
for 2–3 min before running it into the bottle. Durham tube is placed in each bottle or tube of the
medium for indication of gas production.
The following range is put up:
2. Sampling from Reservoir (Streams,
∙∙ One 50 ml quantity of water added to 50 ml
Rivers, Lakes and Tanks) dou­ble strength medium
When sampling from streams or lakes, open the ∙∙ Five 10 mL quantities each to 10 ml double
bottle at a depth of about 30 cm with its mouth strength medium
facing the current. Collect at least 100 ml in each ∙∙ Five 1 ml quantities each to 5 ml single
bottle. strength medium
∙∙ Five 0.1 ml quantities each to 5 ml single
3. Sampling from a Dug Well strength medium.
A stone of suit­able size is tied with the bottle. Incubate the seeded media aerobically at 37°C.
Then a clean cord of suitable length is tied with After 24 hours and 48 hours of incubation, Note
the bottle and lowered into the well. Immerse the the number of cultures of each volume of water
bottle completely in the water. When the bottle is that show the production of acid and gas. These
filled, pull it out, stopper it and wrap it in a kraft acid- and gas-producing cultures are con­sidered
paper. The bottle should not touch the sides of the ‘presumptive positive’ growths of coliform bacilli.
well any time. By reference to tables of most probable number
of coliforms with the combination of positive
Transport and negative results observed, read off the most
Stopper the bottle, label it with full details of the probable number (MPN) of presumptive coliform
source of the water and time and date of collec­ bacilli present in 100 ml of the sampled water (Table
tion, and deliver it to the laboratory as quickly as 83.3). The presumptive coliform count of 0, 1-3,
possible, at least within 6 hours, keeping it in a cool 4-10 and > 10 per 100 mL is interpreted as excellent,
container and protected from light. satisfactory, suspicious and unsatisfactory.
Chapter 83: Bacteriology of Water, Milk and Air  | 577
Table 83.3  Most probable number (MPN) values From the combination of positive and nega­tive
results for gas and indole production at 44°C, read
No. of tubes giving a positive reaction
off the most probable number of E. coli per 100 ml
1 × 50 mL 5 × 10 mL 5 × 1 mL MPN/100mL
of water. This latter value is known as the confirmed
0 0 0 <1 E. coli count.
0 0 1 1
0 0 2 2
0 1 0 1 3. Count of Fecal Streptococci
0 1 1 2
0 1 2 3 If there is difficulty in interpreting the results of
0 2 0 2 the presumptive coliform and confirmed E. coli
0 2 1 3 tests, a demonstration of the presence of fecal
0 2 2 4 streptococci will confirm the fecal origin of the
0 3 0 3
coliform bacilli. Subcultures are made from all the
0 3 1 5
0 4 0 5 positive bottles in the presumptive coliform test
1 0 0 1 into tubes containing 5 ml of glucose azide broth.
1 0 1 3 The presence of Enterococcus. faecalis is indicated
1 0 2 4 by the production of acid in the medium within 18
1 0 3 6
hours at 45°C. The positive tubes should be plated
1 1 0 3
1 1 1 5 onto MacConkey’s agar for confirmation.
1 1 2 7
1 1 3 9 4. Count of Clostridium perfringens
1 2 0 5
1 2 1 7 This is tested by incubating varying quantities of the
1 2 2 10 water in litmus milk medium (anaerobically) at 37°C
1 2 3 12 for five days and looking for stormy fermentation.
1 3 0 8
1 3 1 11
1 3 2 14 b. Membrane Filtration Tests
1 3 3 18
1 3 4 21 In this method, a measured volume of the water
1 4 0 13 sample is filtered through a membrane which retains
1 4 1 17 the indicator bacteria on its surface. The membrane
1 4 2 22 is then placed and incubated on a selective indicator
1 4 3 28
1 4 4 35 medium at the appropriate temperature, so that
1 4 5 43 the indicator bacteria grow into colonies on its
1 5 0 24 upper surface. These colonies are counted and the
1 5 1 35 bacteriological content of water calculated.
1 5 2 54
1 5 3 92
1 5 4 161 Tests for Pathogenic Bacteria
1 5 5 > 180
Specific pathogens such as typhoid bacilli or
cholera vibrios may have to be looked for in water
by employing enrichment and selective media.
2. Eijkman Test Isolation of Salmonella Typhi: For iso­lation of
Some spore-bearing bacteria give false-positive Salmonella Typhi, equal volume of water is added to
reac­tions in the presumptive coliform test. It is double strength selenite broth followed by incu­bation
necessary, therefore, to confirm the presence of and subculture on Wilson and Blair’s med­ium.
true (‘fecal’) coliform bacilli. Isolation of vibrio cholerae: For isolation of
The Eijkman test is usually employed to vibrio cholerae, alkaline peptone water is mixed
find out whether the coliform bacilli detected in with nine times its volume of water, incubated
the presumptive test are E. coli. After the usual and subcultured on bile salt agar. A sim­pler and
presumptive test, subcultures are made from all more sensitive method is to filter the water sample
the tubes/bottles showing acid and gas to fresh through membrane filters and incubate the fil­ters
tubes of single strength MacConkey’s medium on appropriate solid media.
al­ready warmed to 44°C. Incubation at 44°C should
be carried out in thermostatically controlled
BACTERIOLOGY OF MILK
water baths. Those showing gas in Durham’s
tubes contain E. coli. Further con­firmation of the Human infections may be caused by the ingestion of
presence of E. coli can be obtained by testing for animal milk which contains microorganisms de­rived
indole production and citrate utilization. either from the animal, e.g. by contamination with

www.ebook3000.com
578  |  Section 6: Miscellaneous
its feces, or from the environment or from milk i. Methylene Blue Reduction Test
handlers such as dairy workers. It depends on the reduction of methylene blue
by bacteria in milk when incubated at 37°C in
Milk-borne Diseases complete darkness.
Milk is a goodmedium for bacteria and good Procedure: The test is performed by adding 1 ml of
vehicle for many pathogens. Milk-borne diseases standard me­thylene blue solution to 10 ml of milk
are shown in Table 83.4. in a test tube. The tube is closed with a sterile rubber
stopper, inverted once or twice and incubated in the
Bacteriological Examination of Milk dark at 37°C. The milk is considered sat­isfactory, if
1. Viable Count it fails to reduce the dye in 30 minutes.
This is estimated by doing plate counts with serial Resazurin test: The Resazurin test is similar but
dilutions of the milk sample. Raw milk always the dye resazurin, on reduction, passes through a
contains bacteria, varying in number from about series of color changes.
500 to several million per ml.
ii. Phosphatase Test
The enzyme phosphatase normally present in milk
2. Test for Coliform Bacilli
is inactivated if pasteurization has been carried
This is tested by in­oculating varying dilutions out properly. The test depends upon the ahility of
of milk into three tubes of MacCo­n key’s fluid the enzyme to liberate p-nitrophenol from diso­
medium with Durham tube and noting the dium p-nitrophenyl phosphate and thereby pro­
production of acid and gas after incubation at 37°C duce a yellow color that can be quantitated by a
for 48 hours. All coliforms are killed by adequate colorimeter. Residual phosphatase activity indicates
pasteurization and their presence in pasteur­ that pasteurization has not been adequate.
ized milk indicates faults in pasteurizer or post­
pasteurization contamination. iii. Turbidity Test
This is the definitive test for sterilized milk. If milk
3. Chemical Tests has been boiled or heated to the tem­perature
prescribed for ‘sterilization’, all heat coag­ulable
Table 83.4  Milk-borne diseases proteins are precipitated. If ammonium sulfate
1. Infections of animals that can be transmitted to is then added to the milk, no turbidity results.
man Absence of turbidity indicates that the milk has
Primary importance been heated to at least 100°C for at least 5 min.
• Tuberculosis
• Brucellosis 4. Examination for Specific Pathogens
• Streptococcal infections
a. Tubercle bacillus
• Staphylococcal enterotoxin poisoning
Centrifuge 100 ml of milk at 3,000 rpm for 30
• Salmonellosis
minutes and ino­culate two guinea pigs. Keep the
• Q fever
Lesser importance animals under observation for signs of tuberculous
• Cowpox lesions. Tubercle bacilli may also be isolated in
• Foot and mouth disease culture. Microscopic examination for tubercle
• Anthrax bacilli is unsatisfactory.
• Leptospirosis b. Brucella
• Tick-borne encephalitis—transmitted through goat Brucella may be isolated by inoculating cream
milk
from the milk sample on serum dextrose agar or
2. Infections primary to man that can be transmitted
by injecting centrifuged deposit of the milk sample
through milk
intramuscularly into guinea pigs. The animals are
• Typhoid and paratyphoid fevers
sacrificed after six weeks and the serum tested for
• Shigellosis
agglutinins and the spleen inoculated in culture
• Cholera
media for brucellae. Brucellosis in animals can be
• Enteropathogenic Escherichi coli (EEC)
detected also by demonstrating the antibodies in
• Nondiarrheal diseases
milk, by the milk-ring or the whey agglutination tests.
a. Streptococcal infections
b. Staphylococcal food poisoning
c. Diphtheria EXAMINATION OF AIR
d. Tuberculosis
A person inhales over 15 cubic meters of air in the
e. Enteroviruses
course of a day. Hence the bacterial content of the
f. Viral hepatitis
air one breathes is important, particularly when it
Chapter 83: Bacteriology of Water, Milk and Air  | 579
contains pathogens. The bacterial content of air Important question
depends on the location, i.e. whether it is outdoor
rite short notes on:
W
air or indoor air.
a. Bacterial flora in water
Essential conditions for bacteriological examin­ b. Water-borne pathogens
ation of air c. Bacteriological analysis of water
1. Surgical operation theater. d. Bacteriology of milk
e. Presumptive coliform count
2. In hospital wards in which there is an outbreak
f. Eijkman test
of cross-infection.
g. Bacteriology of air.
3. Premises where food articles are prepared and
packed.
Multiple choice questions (MCQs)
4. Premises where pharmaceutical materials are
prepared. 1. Which of the following organisms can serve as
indicators of fecal pollution of drinking water?
a. E. coli
Measurement of Air Contamination b. Bacillus subtilis
The methods for bacteriological examination of air c. Corynebacterium diphtheriae
d. All of the, above
are of two types.
2. All the following bacteria are indicators of bacterial
contamination of water except:
1. Settle Plate Method a. Escherichia coli
Petri dishes containing an agar medium of known b. Enterococcus species
c. Staphylococcus aureus
surface area are left open for a measured period d. Clostridium perfringens
of time. Large bacteria-carrying dust particles
3. Drinking water is considered satisfactory if the
settle on to the medium. The plates are incubated presumptive coliform count of water is:
at 37°C for 24 hours and a count of the colonies a. 0/100 mL b. 1–3/100 mL
formed shows the number of settled pastulle. Blood c. 4–10/100 mL d. > 10/100 mL
agar medium may be used for the count of the 4. The test done for the routine bacteriological
pathogenic, com­mensal and saprophytic bacteria analysis of water is:
in the air. The method is specially used for testing A. Slit sampler method b. Turbidity test
the air in surgical theatres and hospital wards. C. Phosphatase test d. Eijkman test
5. All the following bacteria can be normally present
2. Slit Sampler Method in the milk except:
a. Streptococcus lactis
By this method, the number of bacteria in a meas­ b. Enterococcus species
ured volume of air is determined. c. Lactobacillus bulgaricus
d. Streptococcus cremoris
Procedure: In this, a known volume of air is 6. All the following tests are used for detection of
directed onto a plate through a slit 0.33 mm wide bacterialcontamination of milk except:
and 27.5 mm long with vertical parallel sides about a. Eijkman test
3 mm deep. At the correct negative pressure, air will b. Methylene blue reduction test
enter through a slit of these dimensions. The culture c. Phosphatase test
medium is incubated and colonies counted which d. Turbidity test
gives the number of bacteria present in the air. 7. The method used for estimation of number of
bacteria in a measured volume of air is:
a. Settle plate method
Key Points b. Phosphatase test
c. Turbidity method
™™ Methods of water analysis include presumptive
d. Methylene blue reduction test method
coliform count, differential coliform count,
membrane filtration method, and detection of fecal 8. In operation theaters bacterial count of air should
streptococci and C. perfringens not exceed:
™™ Pathogenic microorganisms in milk can transmit
a. 50 per cubic foot
milk-borne dis­e ases such as tuberculosis, and b. 10 per cubic foot
typhoid fever c. 5 per cubic foot
d. 1 per cubic foot
™™ Bacteriological examination of milk can be carried
out by colony counts, coliform counts; chemical tests 9. What is the acceptable limit of bacterial count in
such as methylene blue reduction test, phosphatase air in operation theatre for neurosurgery?
test, and turbidity test; and detection of specific a. 50 per cubic feet b. 10 per cubic feet
pathogens c. 4 per cubic feet d. 1 per cubic feet
™™ Settle plate method and slit sampler methods are Answers (MCQs)
used for bacteriological examination of air. 1. a; 2. c; 3. b; 4. d; 5. b; 6. a; 7. a; 8. b; 9. d

www.ebook3000.com
84
Chapter

Hospital Waste Management

LEARNING OBJECTIVES

After reading and studying this chapter, you should waste; Waste egregation; Waste treatment and
be able to: disposal
∙∙ Describe universal precautions ∙∙ Describe treatment and technologies for health
∙∙ Describe the following: Categories of biomedical care waste.

Let the wastes of “the sick” not contaminate the lives of the healthy”

INTRODUCTION 6. Decontaminate the laboratory work surfaces


with an appropriate disinfectant after the
Hospitals regularly generate waste which may be spillage of blood or other body fluids and
a potential health hazard to health care workers, when the procedures are completed.
the general public and the environment. Therefore, 7. Limit use of needles and syringes to situations
adequate management and disposal of waste is for which there are no other alternatives.
essential. 8. Biological safety hoods should be used for
laboratory work.
UNIVERSAL PRECAUTIONS 9. All the potentially contaminated materials
of the laboratory should be decontaminated
Concerns about transmission of the hepatitis
before disposal or reprocessing.
B virus (HBV) and human immunodeficiency
10. Always wash hands after completing
virus (HIV) led to the introduction of ‘universal
laboratory work and remove all protective
precautions’, to minimize the infections in medical
clothings before leaving the laboratory.
laboratory workers and health care personnel.
These universal precautions include: Agents which are associated with laboratory
1. Assume that all specimens/patients are acquired infections: Most common agents which
potentially infectious for HIV and other blood are associated with laboratory acquired infections
borne pathogens. include hepatitis B virus, Coccidioides immitis,
2. All blood specimens or body fluids should Bacillus anthracis, Brucella species, Mycobacterium
be placed in a leak-proof impervious bags for tuberculosis, Francisella tularensis and Shigella
transporation to the laboratory. species.
3. Use gloves while handling blood and body
fluid specimens and other objects exposed DEFINITION OF BIOMEDICAL WASTE
to them. If there is a likelihood of spattering, (BMW)
use face masks with glasses or goggles. According to Biomedical Waste (Management
4. Wear laboratory coats or gowns while working and Handling) Rules, 1998 of India, “Biomedical
in the laboratory. Wrap-around gowns should waste” means any waste, which is generated
be preferred. These should not be taken during the diagnosis, treatment or immunization
outside. of human beings or animals or in research activities
5. Never pipette by mouth. Mechanical pipetting pertaining thereto or in the production or testing
devices should be used. of biologicals.

Chap-84.indd 580 15-03-2016 11:28:48


Chapter 84: Hospital Waste Management  | 581
CATEGORIES OF BIOMEDICAL WASTE translucent bags for shredding/mace­ration and
disinfection before disposal (recy­cling or landfill).
The detail categories of biomedical waste as given
in schedule 1 of BMW’98 annexture A (Box 84.1).
Black
WASTE SEGREGATION Incineration ash and solid chemical waste such as
It is important that hazardous waste component is discarded medicines should be collected in black
separated from non hazardous waste and to collect bags for disposal in secured landfill.
these in appropriate receptacles. Mixing of waste Proper segregation will minimise the waste
will render the entire waste potentially hazardous. stream needing special treatment i.e. infectious
The wastes are segregated preferably at the point waste. This practice also helps in safe handling and
of generation. This is the most important step to transporation of waste.
safeguard the occupational health of healthcare
personnel. Waste should be segregated in bags of TREATMENT AND DISPOSAL TECHNO­
different colors to facili­tate appropriate treatment
LOGIES FOR HEALTHCARE WASTE
and disposal (Table 84.1).
Waste Treatment
Yellow Bags
Following techniques are in use for treatment of
Infectious non-sharp waste should be put in yellow infected material.
bags. 1. Incineration
2. Autoclaving
Red Bags 3. Microwaving
Red bags may be used for non-sharp waste (except 4. Hydroclaving
anatomical tissues) if micro­waving/ autoclaving/ 5. Plasma torch
chemical treatment fol­lowed by landfill is the 6. Chemical treatment.
disposal option.

Blue/White Translucent 1. Incineration


Plastic disposable items such as used gloves, Incineration is a high temperature dry oxidation
catheters and IV sets should be put into blue/white process, that reduces organic and combustible
waste to inorganic incombustible matter and
Box 84.1  Schedule 1: Category of biomedical
results in a very significant reduction of waste.
waste in India
Category No. 1 Human anatomical waste
Advantage of incinerator
The incinerator has an advantage of dealing with
Category No. 2 Animal waste
all pathological and cytotoxic wastes. Body parts,
Category No. 3 Microbiology and biotechnology
waste animal waste, microbiological waste and soiled
Category No. 4 Waste sharps dressings can be treated with this technique.
Category No. 5 Discarded medicines and cytotoxic Disadvantage of incinerator
drugs 1. It generates highly toxic gases (e.g. dioxins and
Category No. 6 Solid waste furans, if PVC plastics are present).
Category No. 7 Infectious solid waste 2. It adversely affects the health of the community.
Categroy No. 8 Liquid waste 3. Recycling and reprocessing of materials cannot
Categroy No. 9 Incineration ash be done.
Categroy No. 10 Chemicals used in production 4. Burning of plastic waste or sharps is also not
of biologicals, chemicals used in
disinfection, as insecticide, etc.
recommended.

Table 84.1  Color coding and type of container for disposal of biomedical wastes
Color coding Type of container Waste category Treatment option as per
schedule 1
Yellow Plastic bag 1, 2, and 5, Cat. 6 Incineration/deep burial
Red Disinfected container/plastic 3, 4, 7 Autoclaving/microwaving/
bag chemical treatment
Blue/white translucent Plastic bag/puncture proof Autoclaving/microwaving/
container chemical treatment and
destruction/shredding
Black Plastic bag Cat. 5 and Cat. 9 and Cat. Disposal in secured landfill
10 (solid)

www.ebook3000.com
Chap-84.indd 581 15-03-2016 11:28:48
582  |  Section 6: Miscellaneous
Types of incinerators chlo­rine required is 0.1% and for dirty conditions
Three basic kinds of incineration technology are of available chlorine should be 1.0%.
interest for treating health-care waste:
i. Double-chamber pyrolytic incinerators which 4. Wet and Dry Thermal Treatment
may be especially designed to burn infectious Wet thermal treatment
healthcare waste. Wet thermal treatment or steam disinfection is
ii. Single-chamber furnaces with static grate, based on exposure of shredded infectious waste
which should be used only if pyrolytic to high temperature, high pressure steam, and is
incinerators are not affordable, and similar to the autoclave sterilization process.
iii. Rotary kilns operating at high temperatures, Screw-feed technology
capable of causing decomposition of genotoxic Screw-feed technology is the basis of a non-burn,
substances and heat-resistant chemicals. dry thermal disinfection process in which waste is
Double chambered incineration shredded and heated in a rotating auger.
An incinerator should consist of 2 cham­b ers,
5. Microwave Irradiation
primary and secondary. Waste is burnt in one
chamber (primary chamber) at 800°C. Combustion Most microorganisms are destroyed by the
of gases emitted from the first chamber, occurs in action of microwave of a frequency of about 2450
the second or secondary chamber which has a MHz and a wave length of 12.24 cm. The water
high temperature of 1000°C. The negative pressure contained within the waste is rapidly heated by
is maintained inside the incinerator by the system, the microwaves and the infectious components
thereby forcing the end-gases out of the chimney. are destroyed by heat conduction. This cannot be
used for animal or human body parts, metal items
2. Autoclaving or toxic or radioactive material.
Autoclaving, at 121°C for 60 minutes, is an effec­tive 6. Inertization
method for treating infectious waste before disposal. The process of “inertization” involves mixing waste
A separate autoclave dedicated for waste treatment with cement and other substances before disposal,
should be used. Waste arising from microbiology in order to minimize the risk of toxic substances
and biotechnology laboratories includ­ing cultures contained in the wastes migrating into the surface
and stocks must be autoclaved before disposal by water or ground water.
incineration. Plastic disposables includ­ing blood
bags, urine bags, etc. should be autoclaved followed DISPOSAL
by shredding. It is not recommended for pathological
Land filling, deep burial and sewage are used for
waste. Autoclaved material is typically land-filled,
disposal. Infectious waste after treatment can be
therefore, it has a large strain on land fill capacity.
disposed of by land-filling or deep burial. Liquid
Types of autoclaves: There are two kinds of waste can be disposed in sewage drains. Besides
autoclaves: treatment, incineration is also a method of disposal.
i. Prevacuum type: In the prevacuum type, Treatment methods used for different types of
steam is created outside the chamber loaded infectious wastes are shown in Table 81.1.
with waste. Air in the chamber is then gradually
removed and steam is injected in. This type of Waste Management Program
autoclave eliminate ‘cold spots’ and ‘air pockets’ All laboratories should develop waste management
(where the steam is unable to penetrate) by program according to the specific needs of the
creating this vacuum. This ensures quicker individual laboratory. The policies and procedures
heating. A temperature of 121°C and pressure should be incorporated in the laboratory’s
of 15 pounds per square inch is used. operating manuals. Emphasis should be on waste
ii. Gravity autoclave: For gravity autoclave, minimization (by reducing waste, reuse and
the waste material should be subjected to recycling), proper segregation, and health and
autoclave residence time of not less than 60 safety of the workers. All personnel generating,
minutes, while in a vacuum autoclave time collecting, transporting and storing infectious
period should not be less than 45 minutes. waste must be trained under the programme.
Biological (Bacillus stearothermophilus
spores) or chemical indicators (strips/tapes) Key Points
should be used for validation test of autoclave. ™™ Biomedical waste (BMW) means any waste, which
is generated during the diagnosis, treatment or
3. Chemical Disinfection immunization of human beings or animals or in
The most economical and effective disinfectant research activities pertaining there to or in the
production or testing of biologicals
is hypochlorite. For clean conditions available

Chap-84.indd 582 15-03-2016 11:28:48


Chapter 84: Hospital Waste Management  | 583
2. Waste sharps belong to:
™™ Categories of biomedical wastes are eight waste; a. Category No 1 b. Category No 2
animal waste; microbiology and biotechnology c. Category No 3 d. Category No 4
waste; waste sharps; cytotoxic waste; solid waste;
3. Which color plastic bag is used for non-infectious
liquid waste, incineration ash; chemicals used in
waste?
production of biologicals
a. Yellow b. Red
™™ Waste segregation: Waste should be segregated
in bags of different colours to facili­tate appropriate c. Black d. Blue
treatment and disposal 4. Steam autoclaving is recommended for treatment
™™ Treatment and disposal technologies for health care all the followings except:
waste include incineration, autoclaving, microwaving, a. Laboratory waste
hydroclaving, plasma torch and chemical treatment. b. Waste sharps
c. Human anatomical waste
d. Human blood
IMPORTANT QUESTIONS 5. The chemical decontamination is not
recommended for treatment of:
1. Describe various techniques used for the treatment a. Laboratory waste
and disposal of hospital waste. b. Anatomical waste
c. Human blood
2. Write short notes on:
d. Body fluid waste
a. Universal precautions.
b. Segregation of waste. 6. The only disposal technology proven to be capable
c. Hospital waste management of biomedical of handling all components of the biomedical
waste. waste stream is:
a. Incineration
b. Steam autoclaving
MULTIPLE CHOICE QUESTIONS (MCQs) c. Microwave treatment
d. Chemical decontamination
1. Animal waste does not include:
a. Body parts b. Hair and nails ANSWERS (MCQs)
c. Carcasses d. Body fluids 1. b; 2. d; 3. c; 4. c; 5. b; 6. a

www.ebook3000.com
Chap-84.indd 583 15-03-2016 11:28:48
85
Chapter

Vehicles and Vectors

Learning Objectives

After reading and studying this chapter, you should


be able to:
∙∙ Role of various vehicles and vectors in disease spread.

Introduction ∙∙ Cholera
∙∙ Poliomyelitis
To maintain an active infectious disease in a human ∙∙ Hepatitis A virus infection
population, the pathogen must be transmitted ∙∙ Food poisoning
from one host or source to another. Transmission ∙∙ Intestinal parasitic infestation.
is the third link in the infectious disease cycle and
occurs by four main routes: airborne, contact, B. Diseases Transmitted by Blood
vehicle, and vector-borne.
1. Viruses
Vehicles and vectors ∙∙ Hepatitis B
∙∙ Human immunodeficiency viruses (HIV)
The agents of transmission that bring the ∙∙ Human T cell lymphotropic viruses
microorganism from the reservoir to the host may ∙∙ Infectious mononucleosis
be a living entity, in which case they are called ∙∙ Cytomegalovirus
vectors, or they may be a nonliving entity referred
to as a vehicle or fomite. 2. Bacteria
Modes of transmission ∙∙ Syphilis
A. Direct: Transmitted by direct contact between ∙∙ Brucellosis
reservoir and host.
B. Indirect: Transmitted to host (human host) 3. Parasites
via intervening agent(s) such as vectors ∙∙ Malaria
(animals, insects, other humans and vehicles ∙∙ Trypansomiasis (Chaga’s disease)
(water, food, air, medical devices, various other ∙∙ Trypanosoma cruzi.
inanimate objects).
2. Vector-borne
1. Vehicle-borne In infectious disease epidemiology, vector is
Vehicle-borne transmission implies transmission defined as an arthropod or any living carrier (e.g.
of the infectious agent through the agency of water, snail) that transports an infectious agent to a
food, ice, blood, serum plasma or other biological susceptible individual. Transmission by a vector
products such as tissues and organs. Of these may be mechanical or biological. In the latter case,
water and food are the most frequent vehicles of the disease agent passes through a developmental
transmission, because they are used by everyone. cycle multiplication in the vector.

A. Diseases Transmitted by Water and Food A. Mechanical Transmission


∙∙ Acute diarrheas A crawling or flying arthropod mechanically
∙∙ Typhoid fever transports the infectious agent. There is no
Chapter 85: Vehicles and Vectors  | 585
development or multiplication of the infectious c. Cyclodevelopmental: When the disease
agent on or within the vector. agent undergoes cyclical change but does
not multiply in the body of the arthropod, e.g.
B. Biological Transmission filarial parasite in culex mosquito and guinea
The infectious agent undergoing replication worm embryo in Cyclops.
or development or both in vector and require Transovarial transmission
incubation period before vector can transmit, When the infectious agent is transmitted vertically
biological transmission is of three types: from infected female to her progeny in the vector,
a. Propagative: When the disease agent it is know transovarial transmission.
undergoes no cyclical change, but multiplies
in the body of the vector, transmission is said Medical Entomology
to be propagative, e.g. plague bacilli in rat fleas.
A study of the arthropods of medical importance
b. Cyclopropagative: The disease agent under­
is known as medical entomology which is an
goes cyclical change, and multiplies in the
important branch of preventive medicine.
body of the arthropod, e.g. malaria parasite in
Arthropods act as vectors or carriers of diseases
anopheles mosquito.
(Table 85.1).
Table 85.1  Arthropod-borne diseases Key Points
Arthropod Diseases transmitted ™™ Transmission of the infectious disease occurs by
1. Mosquito Malaria, filaria, viral encephalitis (e.g. four main routes: airborne, contact, vehicle, and
Japanese encephalitis), viral fevers (e.g. vector-borne
dengue, West Nile, viral hemorrhagic ™™ The agents of transmission that bring the
fevers (e.g. yellow fever, dengue microorganism from the reservoir to the host may
hemorrhagic fever) be a living entity, in which case they are called
2. Housefly Typhoid and paratyphoid fever, diar­ vectors, or they may be a nonliving entity referred
rhea, dysentery, cholera, gastroenteritis, to as a vehicle or fomite
amoebiasis, helminthic infestations, ™™ Modes of transmission: The mode of transmission
poliomyelitis, conjunctivitis, trachoma, is A. Direct; B. Indirect.
anthrax, yaws, etc.
3. Sandfly Kala-azar, oriental sore, sandfly fever,
oroya fever Important questions
4. Tsetse fly Sleeping sickness 1. Discuss the role of vehicles and vectors in
5. Louse Epidemic typhus, relapsing fever, transmission of infectious agents.
trench fever, pediculosis 2. Write short notes on:
a. Water-borne diseases
6. Rat flea Bubonic plague, endemic typhus, b. Diseases transmitted by blood and blood prod-
chiggerosis, hymenolepis diminuta ucts
7. Blackfly Onchocerciasis c. Diseases transmitted arthropods as vectors.
8. Reduviid Chagas disease
bug Multiple choice questions (MCQs)
9. Hard tick Tick typhus, viral encephalitis, viral
1. All diseases can be transmitted by food and water
fevers, viral hemorrhagic fever, (e.g.
except:
Kyasanur forest disease), tularemia,
tick paralysis, human babesiosis a. Cholera
b. Polimyelitis
10. Soft tick Q fever, relapsing fever c. Hepatitis A virus infection
11. Trombiculid Scrub typhus, Rickettsialpox d. Hepatitis C virus infection
mite 2. Which of the following diseases can be transmitted
12. Itch-mite Scabies by blood?
a. Hepatitis b. HIV infection
13. Cyclops Guinea-worm disease, fish tapeworm c. Syphilis d. All of the above
(D. latus)
Answers (MCQs)
14. Cockroaches Enteric pathogens
1. d; 2. d

www.ebook3000.com
86
Chapter
Emerging and Re-emerging
Infectious Diseases

Learning Objectives

After reading and studying this chapter, you should be able to:
∙∙ Describe the following: Emerging infectious diseases; re-emerging infectious diseases.

Introduction 2. A new breed of deadly hemorrhagic fevers, of


which Ebola is the most notorious, has struck
Sometimes infectious agents that have not in Africa, Asia, the United States and Latin
been pre­viously recognized appear. Emerging America.
infectious diseases are those whose incidence in
3. Hantavirus pulmonary syndrome: The United
humans has increased during the last two decades
States has seen the emergence of hantavirus
or which threaten to increase in the near future.
pulmonary syndrome, other hantaviruses
The term also refers to newly-appearing infectious
have been recognized for many years in Asia,
diseases, or diseases that are spreading to new
where they cause hemorrhagic fever with renal
geographical areas—such as cholera in South
involvement in humans.
America and yellow fever in Kenya. During the past
4. Foodborne and waterborne diseases: Epide­
20 years, at least 30 new diseases have emerged
mics of foodborne and waterborne diseases due
to threaten the health of hundreds of millions of
to new organisms such as cryptosporidium or
people. For many of these diseases there is no
new strains of bacteria such as Escherichia coli
treatment, cure or vaccine and the possibility of
(0157:H7 strain of E.coli) have hit industrialized
preventing or controlling them is limited.
and developing countries alike. A completely
new strain of cholera, 0139, appeared in south­
Examples of Emerging Pathogens eastern India in 1992 and has since spread north
Table 86.1 summarizes the etiological agents and and west to other areas of India, into western
infectious diseases in humans and/or animals China, Thailand and other parts of South-East
recognized since 1973. The year may differ from Asia.
first appearance and first identification of cases. 5. Influenza pandemic: The threat of a new global
Approximately 75% of emerging pathogens influenza pandemic is increasing. Epidemic
are zoonotic, that is, communicated by animals to strains of influenza viruses originate from China.
humans. HIV/AIDS, avian influenza, monkeypox, The influenza virus is carried by ducks, chickens
Nipah, SARS, and Ebola are all the result, to a and pigs raised in close proximity to one another
greater or lesser extent, of interactions with animals on farms. The exchange of genetic material
that led to the emergence and re-emergence of between these viruses produces new strains,
deadly diseases. leading to epidemics of human influenza, each
The diseases in question involve all the major epidemic being due to a different strain.
modes of transmission—they are spread either
from person to person, by insects or animals, or Re-emerging, or resurging
through contaminated water or food.
diseases
1. Human immunodeficiency virus (HIV): The
most dramatic example of a new disease is Reemerging, or resurging, diseases are those that
AIDS, caused by the human immunodeficiency have been around for decades or centuries, but
virus (HIV). have come back in a different form or a different
Chapter 86: Emerging and Re-emerging Infectious Diseases  | 587
Table 86.1  New infectious diseases recognized since 1973
Year Agent Type Disease/comments
1973 Rotavirus Virus Major cause of infantile diarrhea worldwide
1975 Parvovirus B19 Virus Aplastic crisis in chronic hemolytic anemia
1976 Cryptosporidium parvum Parasite Acute and chronic diarrhea
1977 Ebola virus Virus Ebola hemorrhagic fever
1977 Legionella pneumophila Bacterium Legionnaires' disease
1977 Hantaan virus Virus Hemorrhagic fever with renal syndrome (HRFS)
1977 Campylobacter jejuni Bacterium Enteric pathogen distributed globally
1980 Human T-Iymphotropic virus 1 Virus T-cell lymphoma-leukemia
(HTLV-1)
1981 Toxin-producing strains of Bacterium Toxic shock syndrome
Staphylococcus aureus
1982 Escherichia coli 0157:H7 Bacterium Hemorrhagic colitis; hemolytic uremic syndrome
1982 Borrelia burgdorferi Bacterium Lyme disease
1982 HTLV-2 Virus Hairy cell leukemia
1983 Human immunodeficiency virus (HIV) Virus Acquired immunodeficiency syndrome (AIDS)
1983 Helicobacter pylori Bacterium Peptic ulcer disease
1985 Enterocytozoon bieneusi Parasite Persistent diarrhea
1986 Cyclospora cayetanensis Parasite Persistent diarrhea
1986 BSE agent? Non- Bovine spongiform encephalopathy in cattle (Mad
conventional cow disease)
agent
1988 Human herpes virus 6 (HHV-6) Virus Exanthem subitum
1988 Hepatitis E virus Virus Enterically transmitted non-A, non-B hepatitis
1989 Ehrlichia chaffeensis Bacterium Human ehrlichiosis
1989 Hepatitis C virus Virus Parenterally transmitted non-A, non-B liver hepatitis
1991 Guanarito virus Virus Venezuelan hemorrhagic fever
1991 Encephalitozoon hellem Parasite Conjunctivitis, disseminated disease
1991 New species of Babesia Parasite Atypical babesiosis
1992 Vibrio cholerae 0139 Bacterium New strain associated with epidemic cholera
1992 Bartonella henselae Bacterium Cat-scratch disease; bacillary angiomatosis
1993 Sin Nambre virus Virus Hantavirus pulmonary syndrome
1993 Encephalitozoon cuniculi Parasite Disseminated disease
1994 Sabia virus Virus Brazilian hemorrhagic fever
1995 Human herpesvirus 8 Virus Associated with Kaposi's sarcoma in AIDS patients
1996 nvCJD Australian bat lyssavirus Virus −
1997 H5N1 Virus Avian flu (Bird flu)
1999 Nipah virus Virus −
2003 Corona virus Virus SARS

cholera in 1995 and dengue hem­orrhagic fever


location. Re-emerging Pathogens are anthrax
in 1996. New and previously unknown diseases
(Bacillus anthracis), Botulism (Clostridium
continue to emerge (Table 86.2).
botulinum), Plague (Yersinia pestis), smallpox
The emergence of drug-resistant strains of
virus, tularemia (Francisella tularensis).
microorganisms or parasites is promoted by
treatments that do not result in cure. The reasons
Important Examples of Re-emerging for outbreaks of new diseases, or sharp increases
Infections in India in those once believed to be under control, are
Appearance of plague in an explosive form in 1994 complex and still not fully understood. The fact
after a period-of quiescence of almost 27 years, is however, that national health has become an

www.ebook3000.com
588  |  Section 6: Miscellaneous
Table 86.2  Re-emerging infectious diseases and vaccines; and above all by strengthening
epidemiological surveillance and drug-resistance
Disease Causative agent
surveillance mechanisms and procedures with
A. Bacterial appropriate laboratory support for early detection,
tuberculosis Multidrug resistant
confirmation and communication. The laboratory
Mycobacterium tuberculosis
technologist must relearn information once
Typhoid fever Multidrug resistant
thought to be out of date. Media selection,
Salmonella Typhi
identification tech­niques, and safety precautions
Leptospirosis Leptospira interrogans must all be re-examined and implemented.
Melioidosis Burkholderia Interaction with the infection control program
(Pseudomonas) strengthens the establishment of prevention and
pseudomallei
control strategies.
Anthrax Bacillus anthracis The category of diseases—“new diseases—
Plague Yersinia pestis new problems”—such as Ebola and other viral
B. Parasitic hemorrhagic fevers, is probably the most frightening.
Malaria Drug resistant Much of this already applies to HIV/AIDS, one of the
Plasmodium falciparum most serious diseases to emerge in recent decades.
Leishmaniasis Leishmania donovani
Lymphatic filariasis Wuchereria bancrofti, Key Points
Brugia malayi and B. timori
™™ Emerging infectious diseases are those whose
incidence in humans has increased during the last
international challenge. An outbreak anywhere two decades or which threaten to increase in the
must now be seen as a threat to virtually all near future
™™ Re-emerging, or resurging, diseases are those that
countries, especially those that serve as m have been around for decades or centuries, but have
come back in a different form or a different location.
Antimicrobial Resistance ™™ Resistance by disease-causing organisms to
Resistance by disease-causing organisms to antimicrobial drugs and other agents is a major
antimicrobial drugs and other agents is a major public health problem world­wide.
public health problem world­w ide. It is having
a deadly impact on the control of diseases Important Questions
such as tuberculosis, malaria, enterococci,
staphylococci, streptococci, pneumococci and 1. Write an essay on emerging and re-emerging
infections.
Haemophilus influenzae, Neisseria gonorrhoeae,
2. Discuss various factors responsible for the emer­
Shigella dysenteriae, Salmonella Typhi. gence and re-emergence of infectious diseases.
Factors responsible for emergence and
re-emergence of infectious diseases Multiple Choice Questions (MCQs)
1. Economic development and land use, 1. Factors responsible for the emergence and
unplanned and under-planned urbanization. re-emergence of communicable diseases is/are:
2. Overcrowding and rapid population growth. a. Unhygienic living conditions
b. Encroachment by humans to areas so far unin-
3. Poor sanitation.
habited leading to ecological changes.
4. Inadequate public health infrastructure. c. Development of insecticide resistance in vec-
5. Resistance to antibiotics. tors.
6. Increased exposure of humans to disease d. All of the above
vectors and reservoirs of infection in nature, 2. The problem of drug resistance has assumed
and alarming proportions in:
a. Tubercle bacilli
7. Rapid and intense international travel.
b. Pseudomonas aeruginosa
c. Klebsiella pneumoniae
Responding to Epidemics d. All of the above
The strategy for controlling re-emerging diseases is 3. Following are the examples of re-emerging
through available cost-effective interventions such infections except:
as early diagnosis and prompt treatment, vector a. Plague
control measures and the prevention of epidemics, b. Malaria
for malaria ; and DOTS—directly observed c. AIDS
treatment, short-course—for tuberculosis; by d. Ebala hemorrhagic fever
launching research initiatives for treatment Answers (MCQs)
regimens and improved diagnostics, drugs 1. d; 2. d; 3. c
Section 7: Diagnostic Medical Microbiology

87
Chapter

Molecular Detection of Microorganisms

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe the following: Nucleic acid probes,
be able to: polymerase chain reaction (PCR); Transcription
∙∙ Describe molecular methods for microbial mediated amplification ( TMA); Nucleic acid
identification sequence based amplification (NASBA); Ligase
chain reaction (LCR).

Introduction specimens and for identification of organisms after


isolation of culture. Applications of DNA probe
The deoxyribonucleic acid (DNA), ribonucleic
technology in microbiology are:
acid (RNA), or proteins of an infectious agent in
i. Nucleic acid probes for direct detection of
a clinical sample can be used to help identify the
group A streptococci, Chlamydia trachomatis
agent. Molecular methods have been found to be
and Neisseria gonorrhoeae are available.
advantageous in situations in which conventional
ii. Probes for identification of group A strepto­cocci,
methods are slow, insensitive, expensive or not
group B streptococci, enterococci, Haemophilus
available. Additionally, nonnucleic acid-based
influenzae, mycobacteria, N. gonorrhoeae,
analytic methods that detect phenotypic traits
Staphylococcus aureus, Streptococcus pneumo­
not detectable by con­ventional strategies (e.g.
niae, Campylobacter sp., Histoplasma cap­
cell wall components) have been developed to
sulatum, Blastomyces dermatitidis and
enhance bacterial detection, identification, and
Coccidioides immitis isolated in culture are
characterization.
also available.
Molecular Methods iii. DNA probes for detection of LT and ST toxins
of Esch. coli are also available.
Molecular methods are classified into three
categories: B. Amplified Methods
A. Hybridization
1. Polymerase chain reaction (PCR)
B. Amplification
2. Transcription mediated amplification (TMA)
C. Sequencing and enzymatic digestion of nucleic
3. Nucleic acid sequence based amplification
acids.
(NASBA)
4. Ligase chain reaction (LCR)
A. Hybridization (See Chapter 10 for detail)
1. Polymerase chain reaction: It is the target
DNA probes can be used like antibodies as sensitive amplification system. The polymerase chain
and specific tools to detect, locate, and quantitate reaction (PCR) can detect single copies of viral
spe­c ific nucleic acid sequences in clinical DNA by amplifying the DNA many million-fold
specimens. Nucleic acid probes are segments of and is one of the newest techniques of genetic
DNA or RNA labeled with radioisotopes or enzymes analysis. The basic principle of polymerase chain
that can hybridize to complementary nucleic acid reaction (PCR) and its application in clinical
with high degree of specificity. laboratory for diagnosis of various infectious
Principle of nucleic acid probes is described in agents has been described in Chapter 11.
Chapter 10 under heading ‘DNA probes’. Besides originally described PCR, other
A number of DNA probes have been developed types of PCR include reverse-transcriptase PCR
for direct detection of microorganisms in clinical (RT-PCR), nested PCR and multiplex PCR.

www.ebook3000.com
590  |  Section 7: Diagnostic Medical Microbiology
i. Reverse-transcriptase PCR (RT-PCR): B o t h T M A a n d NA S BA a re e x a mp l e s
Reverse transcriptase PCR (RT-PCR) of transcription-mediated amplification.
amplifies an RNA target. The unique step These isothermal assays use three enzymes:
to this procedure is the use of the enzyme transcriptase (RT), RNAase H, and T7 DNA
reverse transcriptase that directs synthesis dependent RNA polymerase. Like TMA, it is also
of DNA from the viral RNA tem­plate. Once an isothermal RNA amplification method. The
the DNA has been produced, relatively rou­ method is similar to TMA. RNA target is reverse
tine PCR technology is applied to obtain transcribed into cDNA and then RNA copies are
amplification. synthesized with the help of RNA polymerase.
ii. Nested PCR: Nested PCR involves the It also does not require thermal cycler. NASBA
based kits for detection and quantitation of
sequential use of two primer sets. The first
HIV-1 RNA and CMV RNA are available.
set is used to amplify a target se­quence.
4. Ligase chain reaction (LCR): Ligase chain
The amplicon obtained is then used as the
reaction (LCR) is an amplification of probe
target sequence for a second amplification
nucleic acid rather than target nucleic acid. By
using primers inter­nal to those of the first this approach an amplified probe is the final
amplicon. Essentially this is an amplification reaction product to be detected, while the target
of a sequence internal to an amplicon. sequence is neither amplified nor incorporated
The advantage of this approach is extreme into this product.
sensitivity and confirmed specificity The LCR uses two pairs of probes that span
without the need for using probes. the target sequence of interest, Once annealed
iii. Multiplex PCR: Multiplex PCR is a method to the target se­quence, a space remains between
by which more than one primer pair is the probes that is en­zymatically closed using a
included in the PCR mixture. This will help ligase (i.e. a ligation reaction). On heating, the
in amplification of more than one target joined probes are released as a single strand that
sequence in a clinical specimen. The control is complementary to the target nucleic acid. These
amplicon should always be detectable after newly synthesized strands are then used as the
PCR. Mutliplex PCRs are usually less sensi- tem­plate for subsequent cycles of probe annealing
tive than PCRs with single set of primers. and liga­tions. Through the process, probe DNA is
iv. Arbitrary primed PCR: Arbitrary primed amplified to a level readily detectable using assays
PCR uses short primers that are not similar to those de­scribed for the biotin-avidin
specifically complementary to a particular system. Like PCR, LCR also requires thermal cycler.
sequence of a target DNA. By comparing frag­ The LCR based amplification has been used
to detect Chlamydia trachomatis and Neisseria
ment migration patterns following agarose
gonorrhoeae.
gel elec­trophoresis, strains or isolates can be
judged to be the same, similar, or unrelated. C. Sequencing and Enzymatic Digestion
v. Real-time PCR: Real-time PCR combines
of Nucleic Acids
rapid thermocycling with the ability to detect
target by fluorescently labeled probes as the 1. Nucleic acid sequences.
hy­brids are formed, i.e. in real time. This 2. High density DNA probes.
3. Enzyme digestion and electrophoresis of
technology al­lows for high throughput of
nucleic acids.
samples, multiplexing reac­tions, quantitation
of target, and on-line monitoring. Applications of molecular
2. Transcription mediated amplification
methods in clinical laboratory
(TMA): Transcription mediated amplification
(TMA) is an isothermal RNA amplification Molecular methods have a significant role in
method and use three enzymes; reverse the following situations in clinical microbiology
transcriptase (RT), RNAase H, and T7 DNA laboratory.
dependent RNA polymerase. RNA target is 1. Detection of uncultivable growing micro­
reverse transcribed into cDNA and then RNA organisms.
copies are synthesized with the help of RNA 2. Role in clinical virology.
polymerase. A 109 fold amplification of the 3. Disease prognosis.
target RNA can be achieved in about 2 hours. 4. Response to treatment.
TMA based assays are available for detection of
M. tuberculosis, C. trachomatis, N. gonorrhoeae, 1. Detection of Uncultivable and Slow
HCV and HIV-l. Growing Microorganisms
3. Nucleic acid sequence-based amplification Molecular methods have been used to detect
(NASBA) or self-sustained sequence replicat­ previously unknown agents di­rectly in clinical
ions (3SR). specimens by using broad-range primers for a
Chapter 87: Molecular Detection of Microorganisms  | 591
number of microorganisms. HCV, Sin nombre virus genes, which mediate van­c omycin resistance
and Human herpes virus 8 (HHV-8), Bartonella among enterococci, and the mec gene, which
henselae are some examples of human pathogens encodes resistance among staphylococci and
first identified from clinical specimens using rifampicin resistance in Mycobacterium tuberculosis.
molecular methods. Molecular techniques have a significant role
These methods are also useful for fastidious in predicting and monitoring patient responses to
microorganisms which may die in transit or may antiviral therapy. HIV-1 viral load assays have been
be overgrown by contaminants when cultured. N. developed to monitor the response of antiretroviral
gonorrhoeae is one such example whose nucleic therapy. Viral load assays have also been used in
acid can be detected under circumstances in which monitoring the response to therapy in patients who
it cannot be cultured. The use of improper collection, are chronically infected with HBV and HCV.
inappropriate transport conditions or delay in Molecular methods can be used to detect drug
transport can reduce the viability of the organism but resistance mutations in RT and protease genes
do not affect the nucleic acid detection. of HIV-1. These mutations lead to lower levels of
These can detect and identify organisms sensitivity to antiretroviral drugs and are important
that can­not be grown in culture or are extremely causes of treatment failure. This helps to determine
difficult to grow (e.g. hepatitis B virus and the an appropriate treatment in patients who do not
agent of Whip­ple’s disease) and also more rapid respond to therapy.
detection and identification of organ­isms that grow
slowly (e.g. mycobacteria, certain fungi). Key Points
™™ Molecular methods are classified into three
2. Role in Clinical Virology categories: A. Hybridization; B. Amplification; C.
Sequencing and enzymatic digestion of nucleic acids
Molecular methods are limited to replace culture ™™ Amplified methods: 1. Polymerase chain reaction
for detection of bacteria in routine practice (PCR); 2. Transcription mediated amplification (TMA);
because of need to isolate the organisms for 3. Nucleic acid sequence amplification (NASBA); 4.
antibiotic sensitivity testing. The culture can Ligase chain reaction (LCR).
™™ Molecular methods have a significant role in 1.
actually be replaced by these methods only in
Detection of uncultivable growing microorganisms;
those microorganisms which have predictable 2. Role in clinical virology; 3. Disease prognosis; 4.
antibiotic susceptibility, and consequently, routine Response to treatment
susceptibility testing is not performed.
Molecular approaches are often faster, more
sensitive, and more cost-effective than the Important Question
conventional approaches in clinical virology. rite short notes on:
W
Enteroviral meningitis, HSV encephalitis and CMV a. Detection of microorganisms by molecular methods
infections in immunocompromised patients are b. Nucleic acid probes
examples for which nucleic acid-based tests are c. Polymerase chain reaction (PCR) in diagnosis of
infectious agents.
relevant and cost effective for diagnosis.
d. RT-PCR
e. Applications of molecular methods in clinical
3. Disease Prognosis microbiology
Molecular methods are able to quantitate infectious
agent burden di­rectly in patient specimens, an Multiple choice questions (MCQs)
application that has particular importance for 1. DNA probes can be applied for:
managing human immunodeficiency virus (HIV a. Detection of microbes in specimens
infections). Thus, it provides important information b. Identification of resistant markers
which may predict disease progression. c. Identification of culture isolates
Molecular methods can be used for subtyping d. All of the above
of certain viruses which may provide information 2. All of the following molecular methods is/are
about the severity of infection. HPV causes amplified methods for detection of microorganisms
except:
dysplasia, neoplasia and carcinoma of cervix in
a. Polymerase chain reaction (PCR)
women. HPV types 16 and 18 are associated with a b. Ligase chain reaction (LCR)
high risk of progression to neoplasia, whereas HPV c. RT-PCR
types 6 and 11 have a low risk. d. ELISA
3. How many pairs of amplification primers are used
4. Response toTreatment and Drug Resistance in nested PCR?
Molecular methods have been developed to detect a. One b. Two
the genes responsible for drug resistance that c. Three d. Five
may not always readily be detected by phenotypic Answers (MCQs)
methods. Examples include detection of the van 1. d; 2. d; 3. b

www.ebook3000.com
88
Chapter

Staining Methods

Learning Objectives

After reading and studying this chapter, you should ∙∙ Describe the following: Simple stains; Differential
be able to: stains; Gram stain; Acid fast stain (Ziehl–Neelsen
∙∙ Describe common staining techniques staining of acid­fast bacilli); Albert’s stain.

Introduction microbes off the slide. The stained microorganisms


are now ready for microscopic examination.
Because most microorganisms appear almost
colorless when viewed through a standard light Types of Stain
microscope, we often must prepare them for
observation. Live bacteria do not show much Basic dyes: Stains are salts composed of a positive
structural detail under the light microscope due and a negative ion, one of which is colored and is
to lack of contrast. Hence, it is customary to use known as the chromophore. The color of so-called
staining techniques to produce color contrast. basic dyes is in the positive ion; in acidic dyes, it is
in the negative ion. Bacteria are slightly negatively
charged at pH 7. Thus, the colored positive ion in
Preparing film or smear for staining
a basic dye is attracted to the negatively charged
Slides bacterial cell.
Film preparations are made either on cover-slips or Basic dyes, which include crystal violet,
on 3 × 1 in glass slides, usually the latter. It is essential methylene blue, malachite green, and safranin, are
that the cover-slips or slides be perfectly clean and more commonly used than acidic dyes.
free from grease, otherwise films will be uneven. Acidic dyes: Acidic dyes are not attracted to most
types of bacteria because the dye’s negative ions are
Smear preparation: A thin film of material repelled by the negatively charged bacterial surface,
containing the microorganisms is spread over the so the stain colors the background instead. Examples
surface of the slide. This film, called a smear, is of acidic dyes are eosin, acid fuchsin, and nigrosin.
allowed to air dry.
Negative staining: Preparing colorless bacteria
Fixation: Before the microorganisms can be against a colored background is called negative
stained, however, they must be fixed (attached) to staining. It is valuable in the observation of overall
the microscope slide. In most staining procedures cell shapes, sizes, and capsules because the cells
the slide is fixed by passing it through the flame of are made highly visible against a contrasting dark
a Bunsen burner several times, smear side up, or by background. Distortions of cell size and shape are
covering the slide with methyl alcohol for 1 minute. minimized because heat fixing is not necessary and
Air drying and flaming fix the microorganisms the cells do not pick up the stain.
to the slide. Fixing simultaneously kills the To apply acidic or basic dyes, microbiologists
microorganisms and attaches them to the slide. It use three kinds of staining techniques: simple,
also preserves various parts of microbes in their differential, and special.
natural state with only minimal distortion.
A. Stained preparations
Staining: Stain is applied and then washed off
with water; then the slide is blotted with absorbent Staining simply means coloring the microorganisms
paper. Without fixing, the stain might wash the with a dye that emphasizes certain structures.
Chapter 88: Staining Methods  | 593
Bacteria may be stained in the living state, but devised by the histologist Hans Christian Gram
this type of staining is employed only for special (1884) as a method of staining bacteria in tissues.
purposes. Routine methods for staining of bacteria It is one of the most useful staining procedures
involve drying and fixing smears, procedures because it classifies bacteria into two large groups:
that kill them. Fixing simultaneously kills the gram-positive and gram-negative.
microorganisms and attaches them to the slide. It
also preserves various parts of microbes in their Method
natural state with only minimal distortion. The staining technique consists of four steps:
1. Primary staining with a basic pararosaniline
Common Staining Techniques (triphenylmethane) violet dye.
2. Application of a dilute solution of iodine.
A. Simple stains
3. Decolorization.
B. Differential stains 4. Counterstaining.
a. Gram stain
b. Acid-fast stain (Ziehl–Neelsen staining of Procedure (see chapter 89)
acid fast bacilli)
b. Acid Fast Stain (Ziehl–Neelsen Staining
C. Special stains
of Acid Fast Bacilli)
a. Negative staining for capsules
Acid fast stain was discovered by Ehrlich (1882),
b. Endospore (spore) staining
who found that after staining with aniline dyes,
c. Flagella staining. tubercle bacilli resist decolorization with acids.
The method, as modified by Ziehl and Neelsen, is
A. Simple stains in common use now.
A single stain is used in simple staining. A simple stain
Principle
is an aqueous or alcohol solution of a single basic dye.
Some of the simple stains commonly used in Some bacteria, such as mycobacteria are resistant
the laboratory are methylene blue, carbol fuchsin, to aniline dyes and do not readily penetrate the
crystal violet, and safranin. substance of the tubercle bacillus and are therefore
1. Loeffler’s methylene blue: The films are unsuitable for staining it. The dye can be made
stained for 3 min, then are washed with water. to penetrate the bacillus by the use of a powerful
This preparation does not readily over-stain. staining solution that contains phenol, and the
Sections are stained for 5 min or longer. application of heat. Once stained the tubercle
bacillus cannot be decolorized even with powerful
Loeffler’s methylene blue is generally the
decolorizing agents for a considerable time and
most useful of the many preparations of this
thus still retains the stain when everything else in
dye. It shows the characteristic morphology of
the microscopic preparation has been decolorized.
polymorphs, lymphocytes and other cells more
Hence, they are called Acid–fast bacilli (AFB).
clearly than do stronger stains such as the Gram
The stain used consists of basic fuchsin, with
stain or Z–N staining (dilute carbol fuchsin.)
phenol (acts as a-mordant) added. The dye is basic
2. Polychrome methylene blue: The preparation is
and its combination with a mineral acid used as
used in a manner similar to Loeffler’s methylene
decolorizer produces a compound that is yellowish
blue. It is also employed in McFadyean’s reaction
brown in color which is readily dissolved out of
for the identification of anthrax bacilli in blood
all structures except acid-fast bacteria. Any strong
films.
acid can be used as a decolorizing agent, but 20%
3. Dilute carbol fuchsin: Dilute carbol fuchsin sulfuric acid (by volume) is usually employed. In
is made by diluting Ziehl–Neelsen’s stain with order to show structures and cells, including non-
10–20 times its volume of water. Stain for 10–25 acid-fast bacteria, that have been decolorized, and
seconds and wash well with water. to form a contrast with the red-stained bacilli, the
preparation is counterstained with methylene blue
B. Differential stains or malachite green.
These stains impart different colors to different Acid fastness has been ascribed to the high
bacteria or bacterial structures. Gram stain and the content and variety of lipids, fatty acids and higher
acid fast stain are two most widely used differential alcohols found in tubercle bacilli. A lipid peculiar
stains. to acid fast bacilli, a high molecular weight hydroxy
acid wax containing carboxyl groups (mycolic
a. Gram Stain acid) is acid fast in the free state. Acid-fastness
Gram stain is the principal stain used for depends also on the integrity of the cell wall
microscopic examination of bacteria. It was first besides lipid contents.

www.ebook3000.com
594  |  Section 7: Diagnostic Medical Microbiology
Procedure-See chapter 89 page 598. the stain carbol fuchsin to build up the diameters
of the flagella until they become visible under the
Fluorochrome Staining for
light microscope.
Acid Fast Bacteria
A stain containing the fluorescent dye, auramine Staining of Volutin-containing Organisms
O, is substituted for the hot carbol fuchsin in
Certain bacilli possess volutin granules in the
the Ziehl–Neelsen method. Tubercle bacilli
protoplasm which aid in the identification of
are rendered fluorescent and becomes easy to
diphtheria bacilli. Well developed granules of
detect by fluorescence microscopy. Heating is
volutin (polyphosphate) may be seen in unstained
unnecessary.
wet preparations as round refractile bodies within
Tubercle bacilli are seen as yellow luminous the bacterial cytoplasm. They tend to stain more
rods in a dark field. When detected under low strongly than the rest of the bacterium with basic
power, the morphology of the bacilli is confirmed dyes, and with toluidine blue or methylene blue
with an oil-immersion objective. they stain metachromatically, a reddish-purple
color. They are demonstrated most clearly by
C. Special Stains special methods, such as Albert’s and Neisser’s,
Special stains are used to color and isolate specific which stain them dark purple but the remainder
parts of microorganisms, such as endospores and of the bacterium with a contrasting counterstain.
flagella, and to reveal the presence of capsules.
Special Stains for Corynebacterium
1. Negative Staining for Capsules diphtheriae (Stains to Demonstrate
Here, bacteria are mixed with dyes such as Indian Metachromatic Granules)
ink or nigrosin that provide a uniformly colored Certain bacilli possess volutin granules in the
background against which the unstained bacteria protoplasm which aid in the identification of
stand out in contrast. This is particularly useful in diphtheria bacilli. Several special and differential
the demonstration of bacterial capsules which do stains are used to stain diphtheria bacilli and to
not take simple stains. demonstrate their metachromatic granules.
Capsules stain poorly, a characteristic exploited 1. Albert’s stain
with a capsule stain, an example of a negative stain. 2. Neisser’s stain
It colors the background, allowing the capsule to 3. Ponder’s stain
stand out as a halo around an organism. 4. Pugh’s stain.
To observe capsules, a liquid specimen is
placed on a slide next to a drop of India ink. A thin 1. Albert’s Stain
glass coverslip is then placed over the two drops, It is used for routine use (See chapter 89, page 599).
causing them to flow together. At the optimum
concentration of India ink, the fine dark particles
of the stain color the background enough to allow
2. Neisser’s Stain
the capsule to be visible. By this method the bacilli exhibit deep blue
granules and the bacterial protoplasm takes pink
2. Endospore (Spore) Staining color.
Endospores cannot be stained by ordinary methods,
such as simple staining and Gram staining, because 3. Ponder’s Stain
the dyes do not penetrate the wall of the endospore. The volutin granules stain dark blue, whereas the
A spore stain is used to make endospores more Bacillus pale blue.
readily noticeable. Generally, malachite green is
used as a primary stain. Its uptake by the endospore
4. Pugh’s Stain
is facilitated by gentle heat. When water is then
used to rinse the smear, only endospores retain the The volutin granules stain reddish purple and the
malachite green. The smear is then counterstained, remainder of the organism light blue.
most often with the red dye safranin. The spores
appear green amid a background of pink cells. Key Points

3. Flagella Staining ™™ Staining means coloring a microorganism with a dye


to make some structures more visible.
Bacterial flagella are too small to be seen with a ™™ Fixing uses heat or alcohol to kill and attach
light microscope without staining. A tedious and microorganisms to a slide.
delicate staining procedure uses a mordant and
Chapter 88: Staining Methods  | 595
2. Give an account of differential stains.
™™ Differential stains, such as the Gram stain and acid-
fast stain, differentiate bacteria according to their 3. Write short notes:
reactions to the stains. a. Simple staining.
™™ The Gram stain procedure uses a purple stain (crystal b. Acid-fast stain or Ziehl-Neelsen’s stain.
violet), iodine as a mordant, an alcohol decolorizer, c. Negative staining.
and a red counterstain. Gram-positive bacteria stain d. Impregnation methods.
purple and Gram-negative bacteria stain pink. e. Albert’s stain.
™™ The acid-fast stain is used to stain organisms
such as Mycobacteria, which do not take up stains
Multiple choice questions (mcqs)
readily; acid-fast organisms stain pink and all other
organisms stain blue. 1. All include basic dyes except:
™™ Special stains: 1. The endospore stain and flagella a. Crystal violet b. Methylene blue,
stain are special stains that color only certain parts c. Malachite green d. Nigrosin
of bacteria. 2. Negative staining is used to make
2. Special stains are used to color and isolate various
microbial capsules visible.
structures such as:
a. Capsule b. Endospore
Important questions c. Flagella d. All of the above

1. Describe in detail the Gram’s staining. Descibe Answers (MCQs)


Gram staining mechanism. 1. d; 2. d

www.ebook3000.com
89
Chapter

Practical Microbiology for MBBS Students

LEARNING OBJECTIVES

After reading and studying this chapter, you should ∙∙ Describe the following: Gram staining; Ziehl–
be able to: Neelsen staing; Alber staining
∙∙ Describe various spots for practical examination ∙∙ Discuss identification of bacterial culture
with identification with relevant questions to answer ∙∙ Describe stool/feces examination for isolation and
∙∙ Prepare exercises for practical in microbiology for identification of pathogenic findings.
undergraduate students

INTRODUCTION Table 89.1 shows specimen copy of practical


question paper of microbiology for MBBS (2nd
Practical examination in microbiology for MBBS Prof.) examination.
(2nd Prof.) may include the following exercises:
1. Spots 1. SPOTS
2. Staining
3. Identification of bacterial culture Question for spots—Identify the spots displayed.
In general, five spots are displayed for identification
4. Stool/feces examination
in the MBBS 2nd Professional microbiology
1. Spots: Five different spots in the form of practical examination. At times one question
specimens-slides, glassware, media, etc. are pertaining to the spot is also asked. There is
kept for identification with relevant questions no specific pattern for the spots.Therfore, the
to answer in one or two sentences. spots may be from bacteriology (in the form of
2. Staining: Two smears are provided for staining culture media without growth and with growth,
i.e. one for Ziehl–Neelsen (Z–N) staining and biochemical reactions, animal pathogenicity
other for Albert’s staining. tests), parasitology (specimens of parasites,
3. Identification of bacterial culture: Bacterial wet preparation of stool specimen Microscopic
growth is provided for identification on a slides), mycology (fungal growth and, microscopic
culture plate as well as in liquid medium. slides), glassware, equipments, swabs and some
4. Stool/feces examination: Feces specimen is miscellaneous items such as anaerobic jar, chrome
given for isolation and identification of two wire or loop, microtiter plate, VDRL slide, agar-agar
pathogenic findings i.e. ova and/or cysts. shreds, etc. It is not possible to give a complete list

Table 89.1  Specimen Practical question paper of Microbiology for MBBS 2nd Professional examination
Total Marks:25
Ques. No. 1 Identify the spots displayed. (2 × 2 + 3 × 1) = 7
Ques. No. 2 (a) Stain smears “A” by Ziehl–Neelsen technique. Draw and comment on this smear and show your findings
to the examiner. (3)
(b) Stain smear “B” by Albert’s technique. Draw and comment on this smear and show your findings tto the examiner. (2)
Ques. No. 3 You have been provided with a pure bacterial culture on plate and in broth. Do Gram’s staining from plate
and hanging drop preparation for motility from broth. Comment on the growth and mention other tests needed to
identify this bacterium. Show your findings to the examiner. 9(3+3+3)
Ques. No. 4 Identify and draw two abnormal findings (ova and cysts) in the given specimen of feces. Show your findings
to the examiner. 4(2+2)

Chap-89.indd 596 15-03-2016 11:29:17


Chapter 89: Practical Microbiology for MBBS Students  | 597
of spots for examination because spots may be possible questions asked are not given here due to
given from any component of Microbiology. space constraint and students are advised to read
The spots which may be generally selected the respective chapters for these spots which will be
in the examination are shown in Table 89.2. The useful for answering the questions related to a spot.

Table 89.2  List of important spots for MBBS practical examination


I. Bacteriology
a. Culture media without growth 4. Nutrient agar with Pseudomonas sp.
1. Nutrient agar 5. Potassium tellurite blood-agar with growth of
2. Blood agar Corynebacterium diphtheriae
3. Chocolate agar c. Biochemical reactions for bacteria
4. MacConkey's agar 1. Glucose with acid and gas (with Durham’s tube)
5. Loeffler's serum slope 2. Carbohydrate fermentation tests
6. Lowenstein-Jensen (LJ) medium 3. CAMP reaction
7. Blood culture set containing glucose broth and 4. Indole test
taurocholate broth 5. Methyl red test
8. Potassium telluride blood agar 6. Vogus Proskauer (VP) test
9. Castaneda blood culture medium 7. Citrate utilization test
10. Glucose broth 8. Urease test
11. Peptone water 9. Nitrate reduction test
12. Robertson cooked meat (RCM) broth 10. Phenylpyruvic acid (PPA) test
13. Selenite F broth 11. Triple sugar iron (TSI) agar
b. Culture media with growth d. Microscopic slides
1. Blood agar with growth of Streptococcus, 1. Gram-positive cocci
Staphylococcus or Proteus sp. 2. Gram-negative bacilli
2. Lowenstein-Jensen (LJ) medium with growth of M. 3. Corynebacterium diphtheriae
tuberculosis or atypical mycobacteria. 4. Intracellular diplococci
3. MacConkey's agar with growth of Escherichia coli, 5. Mycobacterium tuberculosis
Klebsiella sp.—lactose fermenter(LF) or nonlactose 6. Mycobacterium leprae
fermenter (NLF) colonies
II. Parsitology
a. Specimens of adult parasites 4. Egg of Taenia
1. Hookworm 5. Trichuris trichiura
2. Roundworm 6. Egg of Enterobius vermicularis
3. Hydatid cyst 7. Egg of hookworm
4. Tapeworm 8. Egg of Hymenolepsis nana
b. Wet preparation of stool specimen c. Microscopic slides
1. Cyst of Entamoeba histolytica 1. Different stages of Plasmodium vivax or P. falciparum
2. Cyst of Giarddia lamblia 2. LD bodies
3. Egg of roundworm 3. Microfilaria bancrofti
III. Mycology
a. Fungal Growth b. Microscopic slides
1. Sabouraud's dextrose agar (SDA) 1. Candida albicans,
2. Sabouraud's dextrose agar (SDA) with growth of 2. Germ tube of Candida albicans
Candida sp. 3. Cryptococcus neofrmans
3. Sabouraud's dextrose agar (SDA) with growth of 4. Histoplasmosis
Cryptococcus neofrmans 5. Rhinosporidiosis
4. Sabouraud's dextrose agar (SDA) with growth of
Aspergillus, sp (Aspergillus niger, Aspergillus flavus
Aspergillus fumigatus)
5. Slide culture test used in mycology
IV. Virology
1. Hemagglutination plate Microscopic slides
2. Specimen of pocks on chorioallantoic membrane Negri bodies
(CAM)
V. Glassware
1. Bijou bottle 6. Petri dish
2. Desiccator 7. Tuberculin syringe
3. Glass syringe 8. Universal container
4. Graduated pipette 9. Medically flat bottle or McCartney bottle
5. Pasteur pipette

Contd...

www.ebook3000.com
Chap-89.indd 597 15-03-2016 11:29:17
598  |  Section 7: Diagnostic Medical Microbiology
Contd...
VI. Immunology
1. Latex agglutination tile 5. Counter immunoelectrophoresis
2. Microtitre plate 6. HIV-Comb test
3. VDRL slide 7. HIV-Tri-dot test
4. Radial immunodiffusion
VII. Disposable items
1. Presterilized disposable container 3. Presterilized disposable swab
2. Presterilized disposable syringe
VIII. Equipments/instruments
1. Incubator 5. Water bath
2. Hot air oven 6. VDRL rotator
3. Autoclave 7. Lovibond comaparator
4. Centrifuge
IX. Miscellaneous spots
1. Agar-Agar shreds 8. West’s postnasal swab
2. Gas-Pak 9. Stoke’s method of antibiotic sensitivity testing
3. Craigie's tube 10. Kirby-Bauer’s method of antibiotic sensitivity
4. McIntosh-Filde's jar testing
5. Nichrome wire loop 11. Tissue culture bottle
6. Seitz filter 12. Candle jar
7. NIH swab 13. Epsilometer or E-test
XI. Medical entomology
1. Mosquito 6. Ticks
i. Anopheles i. Hard tick
ii. Aedes ii. Soft tick
iii. Culex 7. Mite
2. Housefly i. Trombiculid mite
3. Sandfly ii. Itch mite
4. Louse 8. Cyclops
5. Fleas

2. STAINING FOR PRACTICAL 3. The stained smear is decolorized with 20%


EXAMINATION sulfuric acid. The red color of the preparation is
changed to yellow brown. After about 1 min in
In general, two smears are provided for two staining the acid, wash the slide with water, and pour on
to students-Albert’s staining and Ziehl–Neelsen fresh acid. Repeat this procedure several times.
staining.Gram staining is done for identification When it is complete, the film, after washing, is
of bacteria. only very faintly pink.
(In case of lepra bacilli 5% sulfuric acid is
A. Ziehl–Neelsen (Z–N) Staining or Acid used as Mycobacterium leprae is less acid-fast.
Fast Staining Another alternative for decolorization is acid-
A fixed smear is provided for Ziehl–Neelsen (Z–N) alcohol (3 mL HCI and 97 mL ethanol).
staining. If the smear is unfixed, it is fixed by passing 4. The smear is counterstained with a contrasting
the dried slide, film downwards, three times slowly dye such as methylene blue for 1–2 minutes.
through the flame, or by heating through the glass Malachite green can also be used as counterstain
slide (the slide is held, film upwards) in the top of instead of methylene blue.
the Bunsen flame for a few seconds so that the slide 5. Wash with water, blot with clean paper, dry and
becomes hot. mount.
6. Examine under oil immersion (× 100) objective.
Procedure Acid-fast bacilli appear red (color of carbol
1. The slide containing fixed smear is covered with fuchsin) while other organisms, tissue cells and
carbol fuchsin. The carbol fuchsin is left on the debris are stained blue or green according to the
slide for 5–10 minutes with intermittent heating counterstain used (Fig. 89.1).
during that period. Heat the slide until the steam
rises, but without boiling. (Do not allow the stain
Important Points in Observation
to dry, to counteract drying more solution of stain
is added to the slide and the slide reheated). 1. Acid-fast bacilli such as Mycobacterium
2. Wash in tap water. tuberculosis appear red while other organisms,

Chap-89.indd 598 15-03-2016 11:29:17


Chapter 89: Practical Microbiology for MBBS Students  | 599
tissue cells and debris are stained blue or green 3. Wash in water and blot dry.
according to the counterstain used (Fig. 89.1). 4. Cover slide with Albert’s iodine (Albert’s
2. Show your observations to the examiner by solution B) for 1 min.
focusing a good stained field of your smear as 5. Wash with water and blot dry. Observe under
such and by drawing a well labeled diagram the oil-immersion objective (× 100).
using colored pencils. The metachromatic (volutin) granules of
3. Study in detail Chapters 32–34 for all possible Corynebacterium diphtheriae stain bluish black,
questions asked by examiner. and the bacterial protoplasm green and other
organisms mostly light green (Fig. 89.2).
Acid-fast Organisms Important points in observation
1. All mycobacteria are acid-fast e.g. Mycobacterium 1. In case of Corynebacterium diphtheriae, green
tuberculosis, Mycobacterium bovis, Mycobacter­ colored bacilli with bluish black metachromatic
ium leprae, atypical mycobacteria. (volutin) granules are observed.
2. Nocardia asteroides, Nocardia braziliensis. 2. Bacilli are arranged in Chinese letter or
3. Cryptosporidium-a protozoan coccidian cuneiform arrangement (Fig. 89.2).
parasite, which causes opportunistic infections 3. Show your observations to the examiner by
in AIDS, is acid-fast. focusing a good stained field of your smear as
4. Bacterial spores are weakly acid-fast. such and by drawing a well labeled diagram
using colored pencils.
4. Study in detail Chapter 28 for all possible
questions asked by examiner.

Fig. 89.1: Acid-fast bacteria

B. Albert’s stain
A fixed smear is provided for Albert’s staining. If the Fig. 89.2: Albert’s stain
smear is unfixed, it is fixed by passing the dried slide,
film downwards, three times slowly through the Gram Staining (See Also Chapter 88)
flame, or by heating through the glass slide (the slide Gram staining is done for bacterial culture (solid
is held, film upwards) in the top of the Bunsen flame material, such as cultures on agar) which is provided
for a few seconds so that the slide becomes hot. to the student along with liquid culture of the same
material in a test tube which is used for hanging
Staining Solution drop preparation to observe the motility of bacteria.
A. Albert 1 or Albert stain Preparing film or Smear for Staining
1. Toluidine blue 1.5 g 1. Film preparations are made on clean and free
2. Malachite green 2g from grease glass slides.
3. Glacial acetic acid 10 mL 2. Place a loopful of clean water on the slide and
4. Alcohol (95% ethanol) 20 mL the loop is then sterilized.
5. Distilled water 1 liter 3. A minute quantity of material, obtained by
just touching the growth (with solid material,
B. Albert II or Albert’s iodine
such as cultures on agar), is transferred to the
Iodine 6g
drop, thoroughly emulsified to make a smooth
Potassium iodide 9g suspension, and the mixture is spread evenly
Distilled water. 900 mL on the slide. A thin film of material containing
the microorganisms is spread over the surface
Procedure of the slide with the help of wire loop. This film,
1. Make film, dry in air, and fix by heat. called a smear, is allowed to air dry.
2. Cover slide with Albert’s stain (Albert’s solution 4. The slide is then held in the palm of the hand
A) and allow to act for 3–5 min. high over a Bunsen flame and dried. The

www.ebook3000.com
Chap-89.indd 599 15-03-2016 11:29:18
600  |  Section 7: Diagnostic Medical Microbiology
film is fixed by passing the dried slide, film Interpretation of Gram Staining
downwards, three times slowly through the Two broad groups
flame, or by heating through the glass slide. ∙∙ Gram-positive
In the latter method the slide is held, film ∙∙ Gram-negative
upwards, in the top of the Bunsen flame for a
few seconds so that the slide becomes hot. Care Gram-positive: Gram-positive bacteria are those
must be taken not to char the film, and when that resist decolorization and retain the primary
the slide is just too hot to be borne on the back stain, appearing violet.
of the hand, fixation is complete. Air drying and Gram-negative: Gram-negative bacteria are
flaming fix the microorgan­isms to the slide. decolorized by organic solvents (acetone/alcohol)
and, therefore, take the counterstain, appearing red.
Reagents Gram reactivity is of considerable importance
as the gram-positive and -negative bacteria
A. Violet Dye differ not merely in staining characteristics and
Crystal violet or methyl violet is used at concentra­ in structure but also in several other properties
tions of 0.5–2%. Solution is facilitated if the dye is first such as growth requirements, susceptibility to
dissolved in alcohol and then added to the water. antibiotics and pathogenicity.
1. Crystal violet or methyl violet 6B 10 g
2. Absolute alcohol (100% ethanol) 100 mL Gram Staining Mechanism
3. Distilled water 1L The exact mechanism is not understood. It may,
however, be attributed to following:
B. Gram’s Iodine 1. Protoplasm: There is a more acidic protoplasm
1. Iodine 10 g (pH 2–3) in the gram-positive cells, which is
2. Potassium iodide 20 g responsible for retaining the basic dye more
3. Distilled water 1L strongly than the Gram-negative bacteria.
2. Cell wall structure: Different kinds of bacteria
C. Decolorizer react differently to the Gram stain, because
structural differences in their cell walls affect
i. Acetone.
the retention or escape of a combination of
ii. Absolute alcohol (100% ethanol).
crystal violet and iodine, called the crystal
iii. Acetone-alcohol. This is a mixture of 1 volume
violet-iodine (CV-I) complex.
of acetone with 1 volume of 95% ethanol. It
Iodine makes the protoplasm more acidic and
requires application for about 10 seconds.
serves as mordant, i.e. iodine combines with dye
to form a dye-iodine complex and fixes the dye in
D. Safranine Counterstain bacterial cell. When applied to both gram-positive
Safranine 0.5% in distilled water. and gram-negative cells, crystal violet and then
iodine readily enter the cells. Inside the cells, the
Procedure crystal violet and iodine combine to form crystal
1. Heat-fixed smear is covered with a basic purple violet-iodine (CV-I) complex.
dye, usually crystal violet (primary stain) for
one minute. (Other dyes such as gentian violet or Gram-positive Cells
methyl violet may also be used as primary stain). Gram-positive bacteria have a thicker peptido­
2. Wash the smear thoroughly with water. glycan cell wall than Gram-negative bacteria. The
3. Cover the smear with Gram’s iodine for one complex crystal violet-iodine (CV-I) complex is
minute. larger than the crystal violet molecule that entered
4. Wash again with water. the cells, and because of its size, it cannot be washed
5. Decolorize the smear with acetone for 10 out of the intact peptido­glycan layer of gram-positive
seconds or less taking care not to overdecolorise. cells by acetone/alcohol. Conse­quently, gram-
(Alcohol can be substituted for acetone). positive cells retain the color of the crystal violet dye.
6. Immediately wash with water to remove the
decoloriser. Gram-negative Cells
7. Cover the slide with dye safranin (counterstain) Gram-negative bacterial cell walls are thinner, have
for 1 minute. (Dilute carbol fuchsin or neutral a smaller amount of peptidoglycan, less exten­
red may also be used as counterstain). sively cross-linked and contain a high percentage
8. Wash off the smear with water, blot and dry. of lipids. There is a layer of lipopolysaccharide
9. Examine the stained smear under the 100 × as part of their cell wall. They dissolve during
(oil) immersion objective of the microscope treatment with acetone alcohol, forming larger
(Fig. 89.3). pores in the cell wall, and the CV-I complex is

Chap-89.indd 600 15-03-2016 11:29:18


Chapter 89: Practical Microbiology for MBBS Students  | 601

Fig. 89.3: Gram staining

washed out through the thin layer of peptidoglycan –  Clostridium


causing out-flow of dye-iodine complex and take –  Lactobacillus
up counter stain, thus appearing red/pink (gram- –  Mycobacterium
negative). As a result, gram-negative cells are –  Actinomyces
color­less until counterstained with Safranin, after –  Nocardia
which they are pink. B. Gram-negative bacteria
This is not all-or-none phenomenon. The gram- 1. Cocci—Neisseria
positive cells may be decolorized by prolonged 2. Bacilli
treatment with acetone/alcohol. In contrast, a. Enterobacteria
inadequate decolorization may cause cells to – Escherichia coli
appear gram-positive. – Klebsiella
– Salmonella
Cell Wall Integrity – Shigella
It has been found that Gram positivity depends on – Proteus
the integrity of cell wall and presence of specific – Yersinia
magnesium–ribonucleate–protein complex. The b. Vibrio
gram-positive bacteria become gram-negative c. Pseudomonas
when cell wall is damaged. Thus, gram-positive d. Parvobacteria
bacteria may become gram-negative, if the cell is – Hemophilus
aging, mechanically ruptured or by the action of – Bordetella
ribonuclease. – Brucella
e. Bacteroides.
GRAM-POSITIVE AND GRAM-NEGATIVE
BACTERIA 3. IDENTIFICATION OF BACTERIAL CULTURE
A. Gram-positive bacteria A. Identification of Bacterial Culture:
1. Cocci—Staphylococcus; Streptococcus; An Overview
Enterococcus: Streptococcus pneumoniae Bacterial growth on a culture plate and in a liquid
2. Bacilli—Corynebacterium culture medium will be provided to student for
–  Bacillus identification of bacterial culture.

www.ebook3000.com
Chap-89.indd 601 15-03-2016 11:29:19
602  |  Section 7: Diagnostic Medical Microbiology
Table 89.3  Description of appearance of growth Table 89. 4  Description of appearance of growth
on solid media in liquid media
1. Shape—circular, irregular, or rhizoid. 1.  Degree: None, scanty, moderate abundant or profuse
2. Size—in millimeters. Pin-head size is characteristic 2. Turbidity: Present or absent; if present, slight,
of staphylococci and pin-point size is a feature of moderate or dense; uniform, granular or flocculent.
streptococcal colonies. Most of the gram-negative bacteria grow in uniform
3. Elevation—effuse, elevated, convex; concave, turbidity form.
umbonate or umbilicate. 3. Deposit: Present or absent: if present, slight,
4. Margins—bevelled or otherwise. moderate or abundant; powdery, granular, flocculent,
5. Surface—smooth, wavy, rough, granular, papillate or membranous or viscid; disintegrating completely or
glistening. incompletely on shaking, e.g. Strepto­cocci growth
6. Edges—entire, undulate, crenated, fimbriate or occurs as a granular turbidity with a powdry deposit.
curled. 4. Surface growth: Present or absent; All aerobes
7. Color—pigment production have tendency to grow on surface of media due to
8. Structure—opaque, translucent or transparent more content of oxygen present on the surface, e.g.
9. Consistency—membranous, friable, butyrous or Pseudomonas sp.
viscid.
10. Emulsifiability—easy or difficult.
11. Differentiation—Differentiated into a central and a
peripheral portion

Identification Scheme
The following scheme of identification should be
adopted.

A. Culture Plate
Culture medium is identified and cultural
characteristics of bacterial growth are noted.
1. Culture Medium
Culture media provided may be nutrient agar,
blood agar or MacConkey’s agar. A
i. Cultural characteristics: Then the cultural
characteristics (Table 89.3) of bacterial
growth is described. This may help in some
provisional identification of the bacteria.
ii. Gram staining: Gram staining is done from
culture plate and notes the following:
i. Gram reaction—Gram-positive or gram- B
negative Figs 89.4: (A) Hanging drop preparation;
ii. Morphology—Cocci, bacilli or coccobacilli, (B) Hanging drop examination
approxi­mate size. If bacilli, then describe
Various characterstics are noted such as degree,
whether these are thin, stout, long or short
turbidity, deposit and surface growth (Table 89.4).
bacilli.
Hanging drop preparation—is done to find out.
i. Motility—Motile or non-motile.
B. Liquid Medium ii. Morphology—Cocci, bacilli or coccobacilli.
Liquid medium is prepared from the same culture
plate which is given for examination and has the same Hanging Drop Preparation
bacteria. Therefore, first identify the liquid culture
This experiment is done to demonstrate motility
medium and describe the characteristics of growth
and to study morphology of bacteria (Fig. 89.4). It
(Table 89.4). Hanging drop preparation is made
is prepared from the bacterial growth provided in
from this liquid media for motility and morphology
liquid culture medium in a test tube.
of the bacteria.
Requirements: 1. A clean cavity (depression) slide;
i. Liquid Culture Medium 2. A clean coverslip; 3. Young broth culture; 4. Wire
Liquid culture medium may be in peptone water loop; 5. Bunsen burner/spirit lamp; 6. Microscope.
or glucose broth. Glucose broth is darker and more
yellowish in color as compared to peptone water. Procedure
Generally, streptococci are provided in glucose broth 1. A ‘hollow ground’ slide, (a glass slide with a
whiles all other bacteria are given in peptone water. shallow, circular concavity in its center) is used

Chap-89.indd 602 15-03-2016 11:29:20


Chapter 89: Practical Microbiology for MBBS Students  | 603
for hanging drop preparation (a plasticin ring E. Interpretation and Practical Examination
can also be used). Record your findings on answer sheet as above and
2. Encircle the concavity with a line streak of soft show to the examiner. Examiner may ask a number
petroleum jelly applied with a glass rod to the of questions related to culture medium, Gram’s
surface of the slide just outside the concavity. staining, hanging drop prepara­tion and bacterial
3. Place drop of culture using the sterile wire loop growth. Student should prepare questions related
on the center of the coverslip. to individual bacterium and is instructed to read
4. Invert the cavity slide over the coverslip so in detail the related chapters of important bacteria
that the drop of the culture is in the center of for practical examination.
the cavity; press it lightly, so that the coverslip
adheres to the vaseline ring. Points to Remember for Identification of
5. Then quickly turn round the slide so that the Bacteria in Practical Examination
cover-slip is uppermost. The drop will then be 1. Culture medium should be identified correctly.
hanging from the cover-slip in the center of the 2. Colony characteristics—especially the size,
concavity, that is why it is called hanging drop. shape, elevation, opacity, color, hemolysis and
6. Proceed to examine first with a low-power consistency should be described.
objective and then with a high-power one. 3. Gram reaction should be drawn and labeled
7. First focus the edge of a hanging drop along with the morphology of bacteria before
preparation under low power objective (× 10) showing finding to the examiner.
of microscope, after reducing the light intensity 4. Liquid medium is from the given solid culture
(by partial closing of iris diaphragm). which is used for growth characteristics and
hanging drop preparion to differentiate certain
8. Turn high power objective (× 40) into position
bacteria.
and focus the edge of the hanging drop and
5. In hanging drop preparation, show motility as
observe for the motility of bacteria.
well as morphology of bacteria, e.g. cocci or
Note: Instead of slide with a shallow, circular bacilli. Draw a labeled diagram before you show
concavity, in many institutions, plasticin is the examiner.
provided to students. Make a ring of plasticin and
place it in the center of a clean glass slide. Invert the B. Bacterial Culture Identification ­
glass slide containing the plasticin ring and place it Question No. 3—You have been provided with a pure
on the coverslip. Rest procedure is the same. bacterial culture on plate and in broth. Do Gram’s
staining from plate and hanging drop preparation
Important Points for Observing Motility for motility from broth. Comment on the growth
of Organisms and mention other tests needed to identify this
When examining living organisms for the property bacterium. Show your findings to the examiner.
of active locomotion, it is essential to distinguish Answer—First, the ability to determine which
true motility, whereby the organisms move in organisms grow on selective and nonselective
different directions and change their positions media aids in making an initial distinction between
in the field, from either (1) Passive drifting Gram-positive and Gram-negative isolates.
of the organisms in the same direction in a One of the following media with bacterial
convectional current in the fluid or (2) Brownian growth may be provided for identification:
movement, which is an oscillatory movement 1. Nutrient agar: Pseudomonas aeruginosa
about a nearly fixed point possessed by all small 2. Blood agar: Staphylococcus, aureus, Streptococcus
bodies suspended in fluid and due to irregularities pyogenes and Proteus species.
in their bombardment by molecules of water. 3. MacConkey’s agar
A. Lactose fermenter-
– Escherichia coli
C. Provisional Diagnosis – Klebsiella sp.
A provisional diagnosis can be made on the basis of B. Nonlactose fermenter—Salmonella sp.- Shigella
culture media and liquid medium findings. Further sp.
tests such as fermentation and other biochemical
reactions, serology, etc. should be used to confirm 1. Nutrient Agar
the identification of bacteria. It is often difficult to identify the organism by
examining only the morphology of colony on
D. Final Identification of Bacteria nutrient agar plate. In general, bacterial growth
This depends on a number of tests which may vary with green pigment on nutrient agar is provided
from one bacterium to another. for examination along with liquid broth.

www.ebook3000.com
Chap-89.indd 603 15-03-2016 11:29:20
604  |  Section 7: Diagnostic Medical Microbiology
Confirmatory Biochemical Reactions for
Pseudomonas aeruginosa
1. Oxidative–fermentation
medium of Hugh
and Leifson
(OF medium) Oxidative
2. Glucose Oxidatively with the
pro­duction of acid
only
3. Lactose Negative
4. Maltose Negative
5. Indole Negative
6. MR Negative
Fig. 89.5: Green pigmented growth of Pseudomonas 7. VP Negative
aeruginosa on the nutrient agar
H2S Negative
8. Oxidase reaction (within 30 seconds)
Observations (Fig. 89.5) 9. It reduces nitrates to nit­rites and further to
A. Solid Media gaseous nitrogen.
i. Medium: Nutrient agar. 10. It is catalase, arginine dihydrolase and gelatinase
ii. Colony morphology: The colonies are large, positive
2–3 mm in diameter, smooth, translucent, 11. Lysine decarboxylase Negative
irregularly round and emit a characteristic 12. Aesculian hydrolysis Negative
fruity or grape-like smell. Note your observations on your answer sheet
iii. Pigment : It is a bluish-green diffusible with well labeled diagrams of your observations
pigment. This pigment diffuses into the with color pencils and show to the examiner.
surrounding medium. Demonstration of
the presence of the pigment is absolute 2. Blood Agar
confirmation of a strain as Pseudomonas
aeruginosa and thus the major diagnostic test. Blood agar supports the growth of a variety of
B. Liquid media (Peptone water) fastidious and nonfastidious organisms, gram-
Growth in broth: Dense turbidity with a surface positive bacteria and gram-negative bacteria. Gram-
pellicle green pigment. Surface pellicle gives’ a positive cocci (Staphylococcus aureus or Streptococcus
clue that bacteria are aerobic nature because pyogenes) or gram-negative bacilli such as Proteus
bacteria have tendency to accumulate at the species may also be provided on blood agar plate
top due to more oxygen content available at
surface. A. Gram-positive cocci Observations
C. Gram staining and Hanging Drop Preparation A. Solid Media
Proceed for gram staining (from solid media) i. Medium: Blood agar
and hanging drop preparation (from liquid ii. Colony morphology: In case of a bacterial
media). Note the following finding in case of growth with hemolysis on blood agar,
Pseudomonas aeruginosa proceed further as under Staphylococcus and
Gram staining—gram-negative bacilli Streptococcus below.
Hanging drop preparation (motiliy): Actively
Staphylococcus and Streptococcus are further
motile.
processed as follows.
D. Presumptive diagnosis-Pseudomonas sp.
E. Confirmatory test: P. aeruginosa is identified by B. Liquid media-Growth in broth.
colonial characteristics and simple biochemical C. Gram staining and Hanging Drop Preparation
tests (e.g. positive oxidase reaction). All strains Proceed for gram staining (from solid media) and
Pseudomonas euruginosa give a rapid positive hanging drop preparation (from liquid media). Note
oxidase reaction (within 30 seconds) and this the following finding.
is a useful preliminary test for non-pigmented Gram staining: Gram-positive cocci in clusters
strains. Pseudomonas are nonfermenter and (Staphylococcus)-gram-positive cocci in chains
break down glucose oxidatively with acid (Streptococcus).
production. They reduce nitrates to nitrites and Hanging drop: Non-motile cocci
further to gaseous nitrogen and utilize citrate D. Presumptive diagnosis: Staphylococcus and
as sole source of carbon. Streptococcus are further processed as follows.

Chap-89.indd 604 15-03-2016 11:29:20


Chapter 89: Practical Microbiology for MBBS Students  | 605

Fig. 89.6: Growth of Staphylococcus aureus showing beta Fig. 89.7: Growth of Streptococcus pyogenes showing beta
hemolysis and golden pigment on blood agar hemolysis on blood agar

Staphylococcus 8. Other biochemical tests: Indole negative,


MR positive, VP positive, urease positive and
Observations reduces nitrates to nitrites.
A. Solid Media All the above tests confirm the isolate as a
1. Medium: Blood agar pathogenic strain of Staphylococcus, i.e. Staphylo­
2. Colony morphology: Colonies are 2–4 coccus aureus.
mm in diameter (pin-head size), surface F. Draw well labeled diagrams of your obser­vations
smooth glistening, edge entire, a soft butyrous with color pencils to show Gram reaction.
consistency and an opaque and easily emulsi­
fiable, pigmented appearance surrounded by a Streptococcus pyogenes
zone of β-hemolysis (Fig. 89.6). After observing the colony morpho­logy on solid
medium, growth in liquid medium, Gram’s staining
B. Liquid Media and hanging drop preparation, findings should be
recorded as follows:
Growth in broth: (in peptone water): Uniform
turbidity. Observations
A. Solid Media
C. Gram Staining and Hanging Drop
Preparation 1. Culture Medium: Blood agar
2. Colony morphology: Colonies are small,
C. Proceed for gram staining (from solid media) and
0.5–1.0 mm in diameter (pin-point in contrast
hanging drop preparation (from liquid media). Note
to pin-head size of Staphylococcus), circular,
the following finding.
semi-transparent, low convex discs surrounded
i. Gram’s staining: Gram-positive cocci (1 mm
by a wide zone of b-hemolysis) (Fig. 89.7).
diameter) arranged in grape like clusters.
ii. Hanging drop prepation: Non-motile cocci B. Liquid Media
in clusters. i. Growth in broth: (in glucose or serum):
Granular turbidity with powdery deposite.
D. Presumptive Diagnosis- Staphylococcus sp
C. Gram Staining and Hanging Drop
E. Confirmatory Tests
Preparation
1. Catalase: Positive
i. Gram’s staining: Cocci are Gram-positive,
2. Mannitol fermentation: Positive
spherical to ovoid organisms 0.5–1.0 mm in
3. Oxidative–Fermentation
diameter, in short or moderately long chains.
medium of Hugh and
ii. Hanging drop prepation: Non-motile cocci
Leifson (OF medium): Fermetative in chains.
4. Phosphatase test: Positive
5. Deoxyribonulease D. Presumptive Diagnosis
(DNAase) test: Positive Streptococcus sp.
6. Tellurite reduction: On potassium
tellurite blood agar- E. Confirmatory Tests
black colonies i. Catalase test-Negative. (It differentiates
7. Gelatin liquefaction: Positive Streptococcus from Staphylococcus).

www.ebook3000.com
Chap-89.indd 605 15-03-2016 11:29:21
606  |  Section 7: Diagnostic Medical Microbiology
ii. Insoluble in 10% bile unlike Streptococcus. suspected because swarming does not occur
pneumoniae. To differentiate Streptococcus on MacConkey’s medium, on which smooth
at species level, following tests are to be colorless (NLF) formed.
performed: the
1. Lancefield technique: Hemolytic strepto­ B. Liquid Media
cocci are grouped by the Lancefield Growth in broth (in peptone water): Uniform
technique using serologic methods, or turbidity’ with slight powdery deposit and an
biochemical tests can be performed. ammonical odor in liquid medium (peptone water).
2. Fluorescent antibody technique: For the
rapid identification of group A streptococci.
C. Gram Staining and Hanging Drop
3. Str. pyogenes (Group A) Sensitive to
Preparation
bacitracin (0.04 unit disk).
A key test that should be done is bacitracin i. Gram staining: Gram-negative coccobacilli,
susceptibility or PYR hydrolysis. 1–3 mm long and 0.6 mm wide. Pleomorphism is
4. Pyrrolidonyl naphthylamine (PYR) frequent – short coccobacilli to long filaments.
hydrolysis –Positive. ii. Hanging drop preparation: Actively motile.
iii. Group ‘B’ streptococci-CAMP reaction and
hippurate hydrolysis is positive. D. Presumptive Diagnosis
iv. Group ‘D’ streptococci-Heat resistant test is Proteus sp.
positive. The enterococci grow in the presence
of 6.5% NaCl, 40% bile, at ph 9.6, at 45°C and in E. Confirmatory Tests
0.1% methelyne blue and survives heating at Keeping in view all the observations your
60°C for 30 min. On MacConkey medium they presumptive diagnosis is Proteus sp. It is confirmed
produce deep pink colonies. Enterococci are with the following tests.
PYRase test positive. They do not hydrolyze i. Phenyl pyruvic acid (PPA) test: Positive
hippurate.
ii. Urease test: Positive
E. Draw well labeled diagrams of your observations
iii. Glucose: A/G (acid/gas)
with color pencils to show Gram reaction.
iv. Lactose: Negative
v. Malonate utilization: Negative
BACILLUS PROTEUS SP.
vi. Indole test : Positive in P. vulgaris but is
If the student is provided with a culture media with negative in P. mirabilis.
swarming growth on noninhibitory solid media vii. MR: Positive
such as nutrient agar and blood agar, the same viii. VP: Negative.
procedure is adopted for identification of bacteria. ix. H2S: Positive in P. vulgaris and P. mirabilis.
x. Ornithine decarboxylase: Positive in P.
A. Solid Media mirabilis but negative in P. vulgaris
1. Culture medium: Blood agar (Fig. 89.8)
2. Colony morphology: Swarming appearance on F. Final Diagnosis
noninhibitory solid media, such as nutrient agar The given bacterium may be identified as P. vulgaris
and blood agar with characteristic putrefactive or P. mirabilis depending on biochemical reac­tions.
odor (‘fishy’ or ‘seminal’) then genus Proteus is Typing methods (phage typing, bacteriocin typing
and serotyping schemes) have been developed for
Proteus species. Swarming Proteus strains exhibit the
Dienes phenomenon and this forms the basis for a
precise method of differentiation among such strains.

3. MacConkey’s Agar
1. Gram-negative rods are better described on
MacConkey agar because these organisms
produce similar-looking colonies on blood agar
plate and chocolate agar media. MacConkey
is best used, however, to differentiate lactose
fermenters (LF) from nonlactose ferment­
ers (NLF). Therefore, on MacConkey’s agar,
Fig. 89.8: Growth of Proteus sp. showing swarning on colonies may be either pink (lactose fermen­
blood agar ter) or pale (non-lactose fermenter).

Chap-89.indd 606 15-03-2016 11:29:22


Chapter 89: Practical Microbiology for MBBS Students  | 607

Fig. 89.9: MacConkey agar with smooth, lactose fermenter Fig. 89.10: MacConkey‘s agar with lactose fermenter
(non-mucoid) colonies of Escherichia coli (mucoid) colonies of Klebsiella sp.

2. The LF colonies may be E. coli or Klebsiella Escherichia coli: Red or pink, nonmucoid colony,
sp., while NLF colonies generally given are gram-negative straight, rod and motile.
Salmonella sp. or Shigella sp. Klebsiella sp: 2 Red or pink, mucoid colonies, short
3. Examine the colony characters on solid and plump bacilli on Gram staining and nonmotile.
medium and growth in liquid medium.
4. Gram staining from culture plate and hanging F. Confirmatory Tests by Various
drop preparation from liquid broth are to be Biochemical Reactions (Table 89.5)
made.
Escherchia coli: Biochemical reactions
5. The following features may be observed on
i. E. coli ferments glucose, lactose, mannitol,
Gram’s staining and hanging drop preparation.
maltose and many other sugars with the
production of acid and gas. Typical strains do
Lactose Fermenter (LF)
not ferment sucrose.
A. Solid Media ii. Indole and MR posi­tive, and VP and citrate
1. Culture medium: MacConkey’s agar (Figs negative (IMViC + + – –)
89.9 and 89.10). iii. It is negative for phenylalanine deaminase
2. Colony morphology: red or pink, nonmucoid test, urease test, H 2S production, gelatin
colony—Escherichia coli. liquefaction, growth in the presence of KCN,
Large, mucoid pink colonies—Klebsiellaj sp. and malonate utilization.
B. Klebsiella sp: Biochemical reactions (Table 89.3)
B. Liquid Broth They ferment sugars (glucose, lactose, sucrose,
Growth in broth: General turbidity and a
i. mannitol) with production of acid and gas, urease
heavy deposit or uniform turbidity. positive, indole negative, MR negative, VP positive
and citrate positive (IMViC – – ++). These reactions
C. Gram Staining and Hanging Drop are typical of K. pneumoniae subsp. aerogenes.
Preparation Final diagnosis of Esch. coli or Klebsiella sp. can be
made based on the above-mentioned biochemical
i. Gram staining: Gram-negative straight rod- reactions. Further tests, such as serotyping should
Escherichia coli; gram-negative short and be done for confirmation. Typing mehods, such as
plump bacilli Klebsiellaj sp. bacteriocin typing, phage typing, resistotyping and
ii. Hanging drop preparation: Motile or non­ molecular typing methods (Plasmid analysis, DNA
motile. profiling by random amplified polymorphic DNA
(RAPD) and pulsed-field gel electrophoresis) can
D. Identification be used for Klebsiella strains.
The bacteria may be identified by correlating the
colony characters, Gram staining and hanging B. Nonlactose Fermenter (NLF) Colonies
drop preparation and then by various biochemical 1. For undergraduate practical examination the
reactions. students are provided either of two important
NLF bacteria—Salmonella sp. or Shigella sp
E. Presumptive Diagnosis (non­lactose-fermenting bacteria are pale).
Presumptive diagnosis of Esch. coli or Klebsiella sp. Liquid broth of the same bacteria is also given
can be made on the basis of above features. alongwith it.

www.ebook3000.com
Chap-89.indd 607 15-03-2016 11:29:23
608  |  Section 7: Diagnostic Medical Microbiology
Table 89.5  Differentiating features of Escherichia coli and Klebsiella sp
Characteristics Escherichia coli Klebsiella sp
1. Colony morphology on MacConkey’s Colonies are red or pink due to lactose Colonies are red or pink in color due
agar fermentation (LF or lactose fermenter to lactose fermentation (LF or lac­tose
colonies), nonmucoid, circular moist, fermenter colonies), large and mucoid.
smooth with entire margin (Large, mucoid and red in color)
2. Growth in broth (peptone water) Growth occurs as general turbidity and Uniform turbidity.
a heavy deposit.
3. Gram staining Gram-negative, straight, rod Gram-negative, short, plump,
measuring 1–3 x 0.4–0.7 µm nonsporing, capsulated, 1–2 µm long
arranged singly or in pairs. and 0.5–0.8 µm wide.
4. Motility (Hanging drop preparation) Motile bacilli Nonmotile bacilli
5. Biochemical reactions
1. Acid from glucose Acid and gas (AG) Acid and gas (AG)
2. Lactose + +
Mannitol + +
Sucrose Variable +
Indole + –
Methyl red (MR) + –
Voges–Proskauer (VP) – +
Citrate – +
Urease – +
Growth in KCN – +

Fig. 89.11: Nonlactose fermenter colonies of Salmonella sp Fig. 89.12: Nonlactose fermenter colonies of Shigella sp
on MacConkey‘s agar on MacConkey‘s agar

A. Solid Media D. Presumptive Diagnosis


1. Culture medium: MacConkey’s agar. (Figs The bacteria may be identified by correlating the
89.11 and 89.12) colony characters, Gram staining and hanging
drop preparation and then by various biochemical
2. Colony morphology: See Table 89.6.
reactions.
B. Liquid Broth Presumptive diagnosis of Salmonella sp. or
Shigella sp. can be made on the basis of above
Growth in broth: Abundant growth with uniform features.Differentiating feature of these two bacteria
turbidity. is motility (Shigella sp. is nonmotile and Salmonella
sp. is motile). Differentiating features of Salmonella
C. Gram Staining and Hanging Drop sp. and Shigella sp. are shown in Table 89.6.
Preparation
i. Gram staining: Gram-negative bacilli. E. Confirmatory Tests
ii. Hanging drop preparation: Motile or non- Confirmatory tests of Salmonella sp. and Shigella
motile. sp: The fermentation of carbo­hydrates and other

Chap-89.indd 608 15-03-2016 11:29:24


Chapter 89: Practical Microbiology for MBBS Students  | 609
Table 89.6  Differentiating features of Shigella sp. and Salmonella sp.
Characteristics Shigella sp Salmonella sp.
1. C
 olony morphology on MacConkey’s Colonies are 2–3 mm in diameter, Colonies are 1–3 mm in diameter
agar circular, translucent, convex, circular, translucent, moist, pale
colorless, i.e. pale and yellowish yellow or nearly colorless (NLF),
(nonlactose-fermenting-NLF) entire edge
An exception is S. sonnei, a late
fermenter of lactose (LLF)
2. Growth in liquid media (peptone water) Good growth with uniform Abundant growth with uniform
turbidity turbidity
3. Gram staining Gram-negative bacilli, 2–4 x 0.6 Gram-negative bacilli, 2–4 × 0.6 µm
mm in size
4. Motility (Hanging drop preparation Nonmotile bacilli Motile bacilli

Table 89.7  Distinguishing features of Shigella vi. They reduce nitrates to nitrites, do not form
species H2S and are inhibited by KCN.
Subgroup A B C D vii. Catalase is produced, except by S. dysenteriae
type 1 which are catalase negative.
Species Sh. Sh. Sh. Sh.
dysenteriae flexneri boydii sonnei viii. Members of groups A, B and C fail to decar­
Mannitol – A A A boxylate lysine and ornithine. S. sonnei decar­
boxylates ornithine but not lysine.
Lactose – – – A (Late)
Salmonella sp. Biochemical Reactions (Table
Sucrose – – – A (Late)
89.8)
Dulcitol – – d
Salmonellae will be indole and urease negative,
Indole d d d
catalase positive, oxidase negative and ferment
Ornithine – – – + glucose, mannitol and maltose but not lactose or
decarboxylase
sucrose. S. Typhi will be anaerogenic and ferments
Serotypes 10 6+ 15 Only glucose and mannitol with production of acid only,
variants one
while paratyphoid bacilli (S. Paratyphi A, B and C)
A = Acid; d = Variable will form acid and gas from sugars. Identification
of the isolate is by slide agglutination.
biochemical reactions are very important to
F. Show your findings to the examiner after
differentiate these Salmonella sp. and Shigella sp.
drawing well labeled diagrams of your observations
up to species level (Tables 89.6 to 89.8).
of Salmonella sp. or Shigella sp.
Shigella sp: Biochemical reactions (Table 89.7)
i. The shigellae are divided into four groups, or IV. Bacterial Growth on any Other Medium
species, by their biochemical reactions and
antigenic structure (Table 89.7). The groups Usually bacterial growth is given on the medium
A, B, C and D correspond to the species S. such as nutrient agar, blood agar and MacConkey’s
dysenteriae, S. flexneri, S. boydii and S. sonnei. agar. If the media other than nutrient agar, blood
agar and MacConkey’s agar is provided for
ii. Glucose is fermented with the production of
examination, then proceed using the same basic
acid, without gas, except for two serotypes:
method of processing, i.e. colony morphology,
Sh. flexneri serotype 6 (Newcastle and
liquid broth, Gram staining, hanging drop
Manchester varieties) and S. boydii serotype
preparation, presumptive diagnosis and further
14. Fermentation of mannitol in Sh. flexneri,
confirmatory tests. In vivo examiner may ask any
Sh. boydii and Sh. sonnei
question related to the specific bacteria along with
Mannitol nonfermenting—Sh. dysenteriae questions related to culture media, Gram staining,
iii. Lactose and sucrose are not fermented, except hanging drop preparation.
by Sh. sonnei which ferments them late.
iv. Dulcitol is not fermented by the majority of
shigellae. Adonitol, inositol and salicin are 4. STOOL/FECES EXAMINATION
not fermented. In this exercise, stool specimen is provided to
v. IMV, C––– + +–– (S. dysenteriae serotype 1, S. isolate two pathogenic parasitic findings. Stool
flexneri serotype 6 and S. sonnei are always examination is done to find out ova and cysts of
indole negative). different parasites.

www.ebook3000.com
Chap-89.indd 609 15-03-2016 11:29:24
610  |  Section 7: Diagnostic Medical Microbiology
Table 89.8  Biochemical reactions of Salmonella species
Subgroup S. Typhi S. Paratyphi A S. Paratyphi B S. Paratyphi C
Catalase + + + +
Oxidase – – – –
Urease – – – –
Glucose A AG AG AG
Mannitol A AG AG AG
Maltose A AG AG AG
Lactose – – – –
Sucrose – – – –
Indole – – – –
Methyl red + + + +
Voges- Proskauer(VP) – – – –
Citrate – – + +
H2 S production + – + +
+, positive; negative; A, acid; AG, acid and gas

Fig. 89.14: Stool examination: It should be done in the


direction of arrows starting from one end
Fig. 89.13: Stool preparation

Method any suspicious structures for identification/


Wet preparations of stool specimen are prepared confirmation of the ova, tropho­zoites or cysts.
as follows (Fig. 89.13): ii. Show your findings to the examiner under
1. Saline preparation as well as iodine preparation high power objective (40X) after drawing a
of stool are made on the same glass slide. labeled diagram.
2. A drop of physiologic saline (0.9%) is placed at Note: Student may isolate cysts or eggs of the
one end and a drop of iodine at the other end following parasites:
the same slide.
i. Eggs: Roundworm, hookworm, Enterobius
3. A small amount (2 mg) of feces is added to each
vermicularis, Trichuris trichiura, Hymenolepis
drop and mixed well with the help of a small
nana, Taenia species.
wooden stick to make a uniform suspension.
4. Each preparation (saline and iodine) should ii. Cysts: Entamoeba histolytica, Giardia lamblia.
be covered with a square coverslip, taking care Saline preparation: It is useful for detection of
that no air bubble forms. helminth eggs or larvae, motile trophozoites and
5. The film should not be too thick. A convenient refractile protozoan cysts.
rule of thumb is to prepare the film just thin Iodine preparation: Iodine stains the cysts of
enough so that ordinary newsprint can easily be amebae (e.g. Entamoeba histolytica, Giardia
read through it and should not overflow beyond lamblia) and other protozoa, revealing some details
the edges of the coverslip. that cannot be seen in the unstained preparation
6. Examination (Fig. 89.14): Examine the but kills trophozoites.
preparation under low power objective lens of
microscope (10X). Caution
i. Examine both the preparations (saline and 1. It is easier to identify eggs of cestodes (Taenia
iodine) under low power objective (10X), sp.), helminthes (roundworm, hookworm,
starting at one corner and following a Enterobius vermicularis, Hymenolepsis nana)
systematic ver­tical or horizontal pattern until and protozoa (cyst of Giardia lamblia, Therefore,
the entire preparation has been examined. always prefer to search for these abnormal
A high-power objective is used to identify findings.

Chap-89.indd 610 15-03-2016 11:29:25


Chapter 89: Practical Microbiology for MBBS Students  | 611
2. Cyst of Entamoeba coli is nonpathogenic and A. Eggs—Bile stained
may also be present in the stool specimen. 1. Roundworm (Ascaris lumbricoides)
Never show Entamoeba coli to the examiner Eggs are of two types; fertilized eggs and unfertilized
because it is normally present in the feces and eggs.
is nonpathogenic and student is asked to show The characteristics of a fertilized egg are as follows:
abnormal findings.
3. Always report your findings under high power Fertilized egg (Fig. 89.15A)
objective (40X). Draw well labeled diagrams of It is round or oval in shape measuring 60–75 µm in
your observations. length and 40–50 µm in breadth.
i. It is bile stained and thus brownish in (golden
Precautions brown) color.
ii. It is surrounded by a thick, smooth translucent
i. Before adding feces either to saline or iodine shell with an outer albuminous coat in
drops, feces in container should be mixed the form of rugosities. This outer coat is
properly with wooden stick. Sometimes ova sometimes lost (decorticated egg).
or cysts may settle down in the container, iii. It contains a very large unsegmented ovum
mixing ensures the uniform distribution of with a clear crescentric area at each pole.
these within the feces.
ii. If preparation has dried up, prepare another Unfertilized Egg (Fig. 89.15B)
slide for examination purposes. i. It is narrower and longer measuring 80 µm in
length and 55 µm in breadth.
ii. It is bile stained (brownish in color)
Identification of Eggs and Cysts
iii. It has a thinner shell with an irregular coating
A. Eggs—Bile stained: Roundworm, Trichuris of albumin.
trichiura, Taenia. iv. It contains a small atrophied ovum with
B. Nonbile stained (colorless): Hookworm, a mass of disorganized, highly refractile
Enterobius vermicularis, Hymenolepis nana. granules of various sizes.

2. Trichuris trichiura (Fig. 89.16)


i. Size about 50 µm in length by 25 µm in breadth.
ii. Color-brown (bile-stained).
iii. It has a double shell, the outer one is bile-
stained.
iv. Barrel-shaped with a mucous plug at each
pole.
v. Contains an unsegmented ovum.

B
Figs 89.15A and B: (A) Fertilized egg of Ascaris lumbricoides; Fig. 89.16: Egg of Trichuris trichiura
(B) Unfertilized egg of Ascaris lumbricoides (Roundworm)

www.ebook3000.com
Chap-89.indd 611 15-03-2016 11:29:26
612  |  Section 7: Diagnostic Medical Microbiology

Fig. 89.17: Egg of Taenia

Fig. 89.19: Egg of Enterobius vermicularis (Threadworm)

Fig. 89.18: Egg of Ankylostoma duodenale (Hookworm)

3. Taenia sp. (Fig. 89.17)


i. Spherical and brown in color (bile-stained).
ii. 31 to 43 µm in diameter.
iii. The inner embryophore is brown in color, Fig. 89.20: Egg of Hymenolepsis nana
thick­walled and radially striated.
iv. It contains an oncosphere (measur­ing 14–20 iv. Surrounded by a transparent shell.
µm in diameter) with 3 pairs of hooklets. v. Contains a coiled tadpole-like larva.

B. Eggs—Nonbile Stained (colorless) 3. Hymenolepis nana (Fig. 89.20)


1. Hookworm (Fig. 89.18) i. Spherical or oval in shape.
i. The egg is oval or elliptical in shape, measuring ii. Measuring 30–45 µm in diameter.
65 µm in length and 40 µm in breadth. iii. It has two distinct membranes: Outer mem­
ii. Colorless (not bile-stained). brane and inner embryophore
iii. It is surrounded by a transparent hyaline shell- a. The outer membrane is thin and colorless
membrane. b. The inner embryophore encloses an
iv. It contains a segmented ovum with four oncosphere with three pairs of lancet-
blastomeres. shaped hooklets.
v. It has a clear space between the egg shell and iii. The space between the two membranes is
the segmented ovum. filled with yolk granules and polar filaments
emanating from little knobs at either end of
2. Enterobius vermicularis (Fig. 89.19) embryophore.
i. Colorless, i.e. not bile-stained.
ii. Shape is asymmetrical, being planoconvex, Cysts
i.e. flattened on one side (the ventral side) and Entamoeba histolytica (Fig. 89.21)
convex on the other (dorsal side). i. It is spherical, 10 to 15 µm in diameter and is
iii. Mesures: 50–60 µm in length and 30 µm in surrounded by a highly refractile membrane,
breadth. called the cyst wall.

Chap-89.indd 612 15-03-2016 11:29:29


Chapter 89: Practical Microbiology for MBBS Students  | 613

parasitoloy, mycology, glassware, equipments, swabs


and some miscellaneous items.
2. Staining:
Differential stains, such as the Gram stain and acid-fast
stain, differentiate bacteria according to their reactions
to the stains.
a. Ziehl-Neelsen (ZN) staining or acid fast staining
The acid-fast stain is used to stain organisms, such
as mycobacteria, which do not take up stains readily;
acid-fast organisms stain pink and all other organisms
stain blue.
Fig. 89.21: Cyst of Entamoeba histolytica
Procedure
i. The slide containing fixed smear is covered with carbol
fuchsin. The carbol fuchsin is left on the slide for 5–10
minutes with intermittent heating during that period.
ii. Wash in tap water.
iii. The stained smear is decolourised with 20% sulphuric
acid. The red colour of the preparation is changed
to yellow brown. When it is complete, the film, after
washing, is only very faintly pink.
iv. The smear is counterstained with methylene blue for
1–2 minutes.
v. Wash with water, blot with clean paper, dry and mount.
vi. Examine under oil immersion (100 X) objective.
Acid-fast bacilli appear red while other organisms,
tissue cells and debris are stained blue or green
Fig. 89.22: Cyst of Giardia lamblia according to the counterstain used.
b. Gram staining
ii. It starts as a uninucleate body, but later the The Gram stain procedure uses a purple stain (crystal
nucleus divides to form two and then four violet), iodine as a mordant, an alcohol decolorizer,
nuclei (quadri­nucleate). and a red counterstain.Gram-positive bacteria stain
iii. Uninucleate and binucleate cysts also possess purple and gram-negative bacteria stain pink.
a glycogen mass, which stains brown with Procedure
iodine and 1 – 4 chromidial or chromatoid i. Heat-fixed smear is covered with crystal violet for one
minute.
bars and do not stain with iodine but appear
ii. Wash the smear thoroughly with water.
as refractile bars with rounded ends in normal
iii. Cover with Gram’s iodine for one minute.
saline preparations.
iv. Wash again with water.
v. Decolourise the smear with acetone for 10 seconds or
Giardia lamblia (Fig. 89.22) less (alcohol can be substituted for acetone)
i. The cyst is oval in shape and measures 12 µm vi. Wash with water .
long and 7 µm broad. vii. Cover the slide with dye safranin for 1 minute.
ii. The axostyles lie more or less diagonally, viii. Wash off the smear with water, blot and dry.
ix. Examine the stained smear under the 100X(oil)
form­ing a dividing line within the cyst­wall.
immersion objective of the microscope.
iii. It contains four nuclei. Which may remain
c. Albert’s stain
clustered at one end or lie in pairs at opposite Procedure
poles. i. Make film, dry in air, and fix by heat.
Trophozoites of Entamoeba histolytica or ii. Cover slide with Albert’s stain((Albert’s solution A )and
Giardia lamblia may also be present in the stool allow to act for 3–5 minutes.
specimens. However, fresh stool specimens are iii. Wash in water and blot dry.
required to examine trophozoites to appreciate the iv. Cover slide with Albert’s iodine ((Albert’s solution B)
motility. Trophozoites should always be examined for 1 minutes.
in normal saline preparation. v. Wash with water and blot dry. Observe under the oil-
immersion objective(X100).
The metachromatic (volutin) granules of Coryne­
Key Points bacterium diphtheriae stain bluish black, and the
Practical examination in microbiology for MBBS (2nd bacterial protoplasm green and other organisms
Prof.) examination may include the exercises: 1. Spots; mostly light green
2. Staining; 3. Identification of bacterial culture; 4. Stool/ 3. Identification of Bacterial Culture
feces examination Bacterial growth on a culture plate and in a liquid
1. Spots: There is no specific pattern for the spots. culture medium will be provided to student for
Therfore, the spots may be from bacteriology, identification of bacterial culture.

www.ebook3000.com
Chap-89.indd 613 15-03-2016 11:29:30
614  |  Section 7: Diagnostic Medical Microbiology

Hanging drop preparation Method


Procedure: i. A drop of physiologic saline is placed at one end and
i. A ‘hollow ground’ slide is used for hanging drop a drop of iodine at the other end the same slide.
preparation. ii. A small amount of feces is added to each drop and
mixed well with the help of a small wooden stick to
ii. Encircle the concavity with a line streak. make a uniform suspension.
iii. Place drop of culture using the sterile wire loop on the iii. Each preparation (saline and iodine) should be covered
centre of the cover slip. with a square coverslip.
iv. Invert the cavity slide over the coverslip. iv. The film should not be too thick.
v. Then quickly turn round the slide so that the cover-slip v. Examination
is uppermost. a. Examine both the preparations (saline and iodine)
vi. First focus the edge of a hanging drop preparation under low power objective (IOX). A high-power object­
under low power objective (10 X) of microscope. ive is used to identify any suspicious structures for
vii. With high power objective (40 X), focus the edge of the identification/ confirmation of the ova, trophozoites
hanging drop and observe for the motility of bacteria. or cysts.
b. Saline preparation—is useful for detection of
It is essential to distinguish true motility and and
helminth eggs or larvae, motile trophozoites and
(1) Passive drifting of the organisms or (2) Brownian
refractile protozoan cysts.
movement. c. Iodine preparation-Iodine stains the cysts of amebae
4. Stool/feces examination (e.g. Entamoeba histolytica, Giardia lamblia.) and other
Stool examination is done to find out ova and cysts of
 protozoa, revealing some details that cannot be seen
different parasites in the unstained preparation but kills trophozoites.

Chap-89.indd 614 15-03-2016 11:29:30


Index
Page numbers followed by f and t refer to figure and table, respectively.

A Alpha (a) hemolysin 165 Forssman 89


Alzheimer’s disease 490 histocompatibility 88
ABO antigens 159 Amantadine 400 human erythrocyte 88
ABO hemolytic disease 161 Amidase tests 230 sequestrated 88
Acid fast stain 593 Aminoglycosides 568 Antigen–antibody interactions 101
Acinetobacter 358 Amphitrichous 17 primary stage 101
treatment 358 Ampicillin, for Koch-Weeks secondary stage 101
Acinetobacter baumannii 358 bacillus 320 tertiary stage 101
Acinetobacter lowffi 358 Anaerobic culture 44 Antigen–antibody reactions,
Acquired immunodeficiency Anaerobic Jar 45 general characteristics 102
syndrome 476 McIntosh and Filde’s 45f Antigen-binding site 91
Actinomyces 223 Anaerobiosis, methods of 44 Antigenicity, determinants of 87
Actinomyces israillae 542t Anaphylactic shock 143 Antigen-presenting cells 89
Acute poststreptococcal Anaphylactoid shock 145 Antiglobulin (Coombs’) test 107
glomerulonephritis/AGN 177 Anaphylaxis 143 Antioncogenes 502
pathogenesis 177 cutaneous 144 Antirabies treatment,
Acute rheumatic fever/ARF 177 in humans 143
indications for 462
Acyclovir 400 in vitro 144
Antiseptics 25
Adenosine deaminase (ADA) passive cutaneous 144
deficiency 72 Antisera 5, 573
primary mediators 144
Adenoviruses 418, 497, 500 Antistreptolysin O/ASO test 107, 178
secondary mediators 145
associated viruses 420 Antiviral compounds 400t
systemic 143
classification of 418t Arboviruses 446
Anaplasma 366
disease associated with 419t Anaplasma phagocytophilum 367 associated clinical
Adhesins 78 Andrade’s indicator 49 syndromes 448t
Adjuvant 129 Anthracoid bacilli 208 classification 446
Aedes aegypti 77, 449, 451, 452, 455 Anthrax 208 families of 447
Aerobic culture 44 cutaneous 207 properties of 446t
Aeromonas 295 intestinal 207 Arenaviruses 493
Aflatoxin 528 laboratory diagnosis of 207 Arthritis 89
African histoplasmosis 517 prophylaxis 208 Arthropod-borne diseases 76
Agar pulmonary 207 Arthus reaction 147
bile salt 40 vaccine 208 Arylsulfatase test 230
blood 39t, 39, 40f Antibacterial drugs, mechanisms of Ascoli’s thermoprecipitin test 208
chocolate 39t, 39, 40f action of 566 Aspergillosis 522, 526
deoxycholate citrate 40 Antibiotic sensitivity tests 559 Astrovirus 497
dilution method 564 Antibiotic tolerance tests 54 Ataxia telangiectasia 139
firm 39 Antibiotics 5 Atopy 145
MacConkey 41 Antibody/antibodies 90 ATP, generation of 23
media 39t formation theories 135 Attenuated anthrax vaccine 2
nutrient 39, 39f, 39t monoclonal 127, 135 Attenuation 78
semisolid 39 production of 127, 128f Autoclave 28, 28f
Agglutination properties of 90 sterilization controls 29
passive 107 structure of 90 Autograft 154, 158
reactions 106 synthesis of 125 Autoimmune diseases 152, 153t
slide 106 uses of 129 localized 152
tube 106 Antibody-dependent cell-mediated systemic 152
Albert’s stain 594, 599, 599f cytotoxicity 121 Autoimmunity 151
Alcaligenes faecalis 17, 357, 360 Antigen and antibody reactions, mechanisms of 151
Allergy 5, 142 types of 102 Autolysins 15
Allograft 154, 158 Antigen/antigens 87 Autotrophs 22
fetus 156 complete 87, 89 Azidothymidine 400

www.ebook3000.com
616  |  Essentials of Microbiology
B Bacteriophages 4, 64, 65f, 79, 402 Bordetella species, differentiatial
lysogenic cycle 403, 404f characterstics of 324
Bacillary dysentery 272 lytic cycle 403, 403f Borrelia 342, 347
Bacilli/bacillus 12f, 12, 205 morphology 402, 402f laboratory diagnosis 344
arrangement 13 role of 402 morphology 342
general characterstics of 205 significance of 404 Borrelia burgdorferi 342, 343
Bacillus anthracis 205, 210 Bacteroides spp 223, 534t Borrelia recurrentis 342; 342
antigenic structure 207 Bartonella 368 Borrelia vincenti 342
differentiating features between Bartonella bacilliformis 368 Botulinum toxin 218
B. anthracis and anthracoid Bartonella henselae 369 Botulism 218
bacilli 209 Bartonella quintana 368 food-borne 218
morphology 205 Batch culture 22 infant 218
resistance 207 B-cell 119, 121
wound 218
Bacillus cereus 205, 209 activation 127
Bovine spongiform
infections 210 maturation 121
encephalopathy 489
Bacteremia 80, 533, 535 BCG vaccination 236, 245
Brazilian purpuric fever/BPF 320
Bacteria 11-20 Bence–Jones proteins 95
Brill–Zinsser disease 363, 369
aerobic 23 Benzyl penicillin 168
Beta (β) hemolysin 165 Broth dilution method 563, 564f
anaerobic 23, 221
Betapropiolactone/BPL 33 Broth, nutrient 38t
capsulated 16
fermentative 50 Bifidobacterium 222 Brucella 327, 534
genotypic characteristics of 48, 48t Biomedical waste /BMW 580 antigenic structure 327
gram-positive and gram- black bags 581 classification of 328
negative 14t blue/white translucent bags 581 differential characteristics of 328
methods used to identify 48t disposal 582 laboratory diagnosis 329
phenotypic characteristics 48t, 48 red bags 581 morphology 327
shape of 12, 12f segregation 581 pathogenesis 328
transformation in 64 treatment of 581 resistance 327
mechanism of genetic 64f yellow bags 581 Brucella abortus 23, 326
Bacteria capsule 15 BK polyomaviruses 422 Brucella bacteriophage 328
demonstration of 16 Blastomyces dermatitidis 515 Brucella melitensis 328
Bacteria cell 11 Blastomycosis 515, 518 Brucella suis 328
anatomy of 13, 13f Bleaching powder 32 Brucella tularensis 315
cellular appendages 13, 15 Blood group systems 160 Brucellin skin test 331
chemical structure of 13 ABO 159 Brucellosis 329
components 13 Lewis 160 blood culture in 329
cytoplasm 17 MN system 160 course of disease 329
gram-positive and gram- Rh (rhesus) 160 prophylaxis 331
Blood transfusion 160 treatment 331
negative 13
complications of 160 types of infection 329
mesosomes 17
nonimmunological
ribosomes 17 Bruton’s hypogammaglobulinemia
complications of 161
RNA 17 137
Blotting 71
Bacteria cell wall Burkholderia cepacia 304, 306
northern 71
chemical structure 13, 14f pathogenicity 304
southern 71
comparison of treatment 304
western 71
gram-positive bacteria 13, 14t Burkholderia mallei 304
Bone marrow 117, 123
gram-negative bacteria 13, 14t Burkholderia pseudomallei 305
Bordetella ansorpic 323
functions 13, 14 Burkitt’s lymphoma 414, 500
Bordetella avium 323, 326
gram-negative 14, 15f Bordetella bronchicanis 326 Buruli ulcer 250
gram-positive 14, 14f Bordetella bronchiseptica 323, 326 Busse–Buschke disease 521
Bacterial count 21 Bordetella holmesii 323 C
Bacterial cultures Bordetella petrii 323
identification of 601, 603 Bordetella trematum 323 California encephalitis virus 454
methods of isolating 46 Bordetella hinzii 323 Calymmatobacterium
Bacterial division 21 Bordetella parapertussis 323, 326 granulomatis 360
Bacterial flora, in water 575 Bordetella pertussis 323, 326, 541 Camp test 355
Bacterial growth curve 21, 22f adhesins 324 Campylobacter 297
Bacterial metabolism 23 antigenic constituents 323 Campylobacter jejuni 545
Bacterial nucleus 18 morphology 323 Candida 519
Bacterial nutrition 22 prophylaxis 325 C. albicans 519, 520, 525
Bacterial spore 18, 19f resistance 323 C. glabrata 519
Bacteriology of milk 577 treatment 325 C. guilliermondii 519
Bacteriolysis 98, 100 virulence factors 323 C. krusei 519
Index  | 617
C. parapsilosis 519 Cholera toxin 291 fixation test 102, 108, 109f
C. stellatoidea 519 Christensen’s urease medium 52 indirect 109
C. tropicalis 519 Chromobacterium uses 109
C. viswanathii 519 violaceum 357, 360 inhibitor deficiencies 140
Candidiasis 519, 525 Chromoblastomycosis 512 system 96
superficial 519 Chronic granulomatous disease 140 Conjugation 65, 68
systemic 519, 520 Chronic mucocutaneous chromosomal transfer 66
treatment 521 candidiasis 520 in E.coil 66f
Candle Jar 44 Chronic wasting disease 489 plasmid and chromosomal
Capnocytophaga 360 Ciprofloxacin, for Legionella transfer 66
Capsule-swelling reaction 16 pneumophila 308 Contagious pustular dermatitis 408
Carbapenems 567 Citrate utilization test 51, 51f Continuous culture 22
Cardiobacterium hominis 360 Koser’s citrate medium 51 Cooked meat broth/CMB 42, 46
Carrier 76 Simmons’ citrate medium 51 Coombs’ test
Cellulitis, due to H. inflenzae 319 C1 esterase 99 direct 107
Central dogma of molecular Clostridial endometritis 214 indirect 107
biology 60 Clostridial myonecrosis 214 types of 107
Cephalosporins Clostridium 211 Coronaviruses 494, 495f
antibacterial spectrum 567t classification 212 severe acute respiratory
for lyme disease 344 diseases produced 212 syndrome 495
for Pneumococcus 186 general features of 211 Counter immunoelectrophoresis 105
Chlamydophila psittaci 542t Clostridium botulinum 218, 219, Cowpox 408
220, 404 Coxiella 367
Chediak-Higashi syndrome 140
treatment 219 antigenic variation 367
Chemotactic factors 145
Clostridium difficile 219, 220
eosinophil 145 Coxiella burnetii 367, 370, 542t
antibiotic associated
neutrophil 145 prophylaxis 368
diarrhea 219
Chemotherapy 5 Coxsackievirus 429
pseudomembranous colitis 219
Chicken cholera vaccine 2 antigenic characters 429
toxins 219
Chickenpox 412 clinical syndromes associated
treatment and prophylaxis 219
Chick-Martin test 34 with 431t
Clostridium perfringens 212, 214,
Chigger-borne typhus 365 features of 429, 430t
220, 545, 577
Chikungunya virus 449, 455 group a viruses 430
colitis 214
Chlamydia 371 group B viruses 430
food poisoning 214
antigenic structure 373 morphology 212 C-reactive protein/CRP 83
classification 371 prophylaxis 215 Cresols 31
C. pecorum 371 resistance 212 Creutzfeldt–Jakob disease/CJD 490
C. pneumoniae 371 soft tissue infections 213 Cryoglobulinemia 95
C. psittaci 371 toxins 213 Cryotococcus neoformans 526
differences between treatment 215 Cryptococcal meningitis 521
chlamydiae and viruses 371 Clostridium tetani 215, 220 Cryptococcosis 521, 526
growth cycle 372, 372f Cluster of differentiation 120 Culex tritaeniorhynchus 451, 455
Chlamydia and chlamydophila; Coagglutination 108f Culture media
diseases caused by 372t Coagulase 79 anaerobic media 41
Chlamydia pneumonia 375, 543 Cocci 12, 12f classification of 37, 37t
Chlamydia trachomatis 371, 373 anaerobic 221 complex media 38
biovars of 373 arrangement 12 differential media 41
genital infections 374 Coccidioides immitis 516, 518 enrichment media 37t, 39, 40
inclusion conjunctivitis 373 Coccidioidomycosis 515, 516, 518 indicator media 37t, 40
infections 377 Code 59 ingredients of 37
trachoma 373 Codon 59 liquid media 37, 37t, 38t
Chlamydophila pneumoniae 372, Cold agglutinin test 89 selective media 40
375, 377 Colicinogenic/Col factor 66 semisolid media 38
Chlamydophila psittaci 371, 375, 377 Collagenase 79 simple media 38
Chloramphenicol Collagen-vascular inflammatory solid media 38
for Neisseria meningitidis 190 diseases, complement sugar media 41
for plague 313 deficiencies in 99 transport media 41
for Pneumococcus 186 Colony-stimulating factors 132t, 132 Cystic fibrosis 72
Chlorhexidine 31 Commensals 75 Cytokines 131, 132t, 135
Chlorine 32 Complement 96 Cytolysins 79
Chloroxylenol 31 activation 96, 100 Cytolysis 100
Cholera 291 principle pathways 96 Cytolytic/cytocidal tests 109
prophylaxis 294 biological effects of 98 Cytomegalovirus 393, 413, 416, 500
treatment 293 complement deficiencies 99t, 140 Cytopathic effects 385

www.ebook3000.com
618  |  Essentials of Microbiology
D Ectothrix 510 Epidermolytic toxins/exfoliative
Edward jenner 7 toxins 166
Dapsone; for leprosy 245 Ehrlichia 366 Epiglottitis, due to H. influenzae 319
Delta (δ) hemolysin 166 Ehrlichia chaffeensis 366 Epitome 87
Dengue 451 Ehrlichia ewingii 366 Epsilometer or E-test 563
clinical findings 452 Eijkman test 577 Epstein–barr virus 413, 416, 500
Dengue hemorrhagic fever 99, 452, Eikenella corrodens 359 360 associated malignancies 414
453, 455, 585 treatment 360 infectious mononucleosis 414
Dental caries 181 Electroimmunodiffusion 105 specific antibodies 415
Deoxyribonucleic acid (DNA) 58 countercurrent eletrophoresis/ Erwinia 269
structure 58 CIEP 105 Erysipelas 177
Deoxyribonulease (DNAase) counterimmuno­- Erysipelothrix 357
test 164 electrophoresis/CIE 105 Erysipelothrix rhusiopathiae 356,
Dependo virus 425 one-dimensional double 105 360
Dermatophytes 509 one-dimensional single 105 human infection 357
classification 509 Electrophoresis Erythro virus 424
Diarrhea 544 Laurell’s two-dimensional 106, Erythromycin, for Pneumococcus 186
causative agents of 544t Escherichia coli 534t
106f
traveller’s 544 differentiating features of
Elisa 112f
Dienes phenomenon 268 Escherichia coli and
cassette-based
Digeorge’s syndrome 138 Klebsiella sp 608t
membrane-bound 114
Dilute carbol fuchsin 593 Ethylene oxide 33
competitive 112
Dimorphic fungi 503 Eubacterium 222
indirect 112, 113
Diphthericin 66 Eukaryotic cells 11
sandwich 112, 113
Diplococci 12 differentition from
uses of 113
Diplococcus pneumoniae 183 prokaryotes 11t
Endemic 80
Disinfectants Exaltation 78
Endemic typhus 363, 369
categories of 30 Exons 59
Endocarditis
characteristics of 30 Exotoxins 78t, 78
acute 534
low-level 30
subacute 534
skin 32
used in hospitals 34t
Endospore (spore) staining 594 F
Endospores 19
Disinfection 25, 35 Fab fragments 90
Endothrix 510
high-level 30 Farmer’s lung 147
Endotoxemia 533
methods of 25, 26t Fatal familial insomnia/FFI 490
Endotoxins 78t, 79
Disseminated histoplasmosis 517 Favus 510, 511
Entamoeba histolytica, cysts 612
DNA probes 70 FC fragment 90, 91
Donovania granulomatis 357 Enteric fever 279
functions of 92
Donovanosis 358, 548 Enterobacter spp 534
Fertility factor 66
treatment 358 Enterobius vermicularis 597,
Fever 83
Double helix 58 611,612
Filamentous fungi 503, 519
DPT vaccine 325 Enterococcus/enterococci 174,
Filamentous hemagglutinin
Drug resistance 180,181
adhesin/FHA 324
in bacteria 68 characteristics of 180
Filoviruses 494
mutational CE 68, 68t clinical infections 180
Filters 29
transferable 68, 68t differences between group d
asbestos 29
Duchenne’s muscular streptococci and earthware 29
dystrophy 72 Enterococcus spp 180 membrane 29
Dyes identification 180 sintered glass 29
acridine 33 treatment 180 Filtration 29
aniline 32 Enterococcus avium 180 Fimbria 17
Dysentery 546 Enterococcus casseliflavus 180 Fixed virus 458, 460, 464
microorganisms causing 546t Enterococcus durans 180 Flagella 16
Enterococcus faecalis 174, 180 demonstration of 17
Enterococcus gallinarum 180 Flagellin 16
E Enterococcus raffinosus 180 Flagellum 16f
Eastern equine encephalitis/EEE Enterotoxins, of Staphylococcus Flavobacterium meningosepticum
448t, 455 aureus 166 357, 360
Ebola virus 494 Enteroviruses 394, 398, 426 Flocculation 102
Sudan strain 494 Enzyme immunoassay/EIA 338, 339 Flocculation tests, types of 103
Zaire strain 494 Enzymes 79 Fluctuation test 63, 63f
Echoviruses 430 Epidemic 80 Fluorescent treponemal antibody/
clinical features 430 Epidemic typhus 363 FTA test 339
Index  | 619
Fluorochrome staining 594 Grafts 154 Hepatitis A virus 465, 474
Food poisoning 547 Graft-versus-host reaction 156 antigenic structure 468
Foot-and-mouth disease of cattle 4 Gram stain 48, 593 immunization 467
Formaldehyde 33 Gram staining 599, 601f prophylaxis 467
Formalin 33 interpretation of 600 treatment 467
Fort bragg fever 345t mechanism 600 Hepatitis B carriers 469
F-prime 66 Gram-negative bacteria 601 Hepatitis B virus 467, 474
Fragment-crystallizable See FC active immunization 471
Gram-positive bacteria 601
Frambesia 341 acute infection 469
Granuloma inguinale 358
Francisella tularensis 315 avihepadnavirus 467
Granulomatosis infantiseptica 356
morphology 315 chronic infection 469
Griffith typing 173
prophylaxis 315 hepatocellular carcinoma 500
Gut-associated lymphoid tissue 119 interpretation of serological
Frei’s test 375, 377
French neurotropic vaccine 451 markers 470t
Fungal keratitis 525, 526 H markers 470
Fungi 503 orthohepadnavirus 467
H antigen 159, 302
classification of 503, 503fc passive immunization 471
Hacek group bacteria 321
general properties of 503 prophylaxis 471
Haemophilus aphrophilus 317 structure 467
identification of 506 Haemophilus ducreyi 317, 320, 322 subtypes 468
Fusidic acid 568 Haemophilus haemolyticus 321, 322 treatment 471
Fusobacterium 223 Haemophilus influenzae 317, 318t, Hepatitis c virus 471, 474
322, 534 clinical features 472
G antigenic structure 318 prophylaxis 472
characters of six biotypes of 318t treatment 472
Gamma (γ) hemolysin 165
cultural characteristics 317 Hepatitis D virus/HDV 472, 474
Gardnerella vaginalis 360
growth characteristics of 317 Hepatitis E virus/HEV 473, 474
Gas gangrene 214
invasive infections 319 clinical features 473
Gaspak 45
Gastroenteritis 544 laboratory diagnosis 319 prophylaxis 473
due to Salmonella infection 280 morphology 317 Hepatitis G virus 474
Gastrointestinal tract 82 noninvasive disease 319 Hepatitis viruses, comparative
prophylaxis 320 features of 466t
Gene expression 59
resistance 318 Herpangina 430, 431t
Gene therapy 72
treatment 320 Herpes genitalis 548
applications 72
Haemophilus parainfluenzae 321 Herpes gladiatorum 410
Genetic engineering 68
Halberstaedter-Prowazek/HP Herpes simplex virus 393, 409
basic tools of 69
bodies 373 association with cervical
procedure 69, 69f
Halophilic vibrios 294 cancer 500
Genetics 58
Hand-foot-and-mouth clinical features 410
Genital chlamydiasis 374
disease 430, 431t encephalitis 410
Genitourinary tract, role in
Hansen’s disease 228, 242 See also sporadic encephalitis 410
immunity 82
leprosy treatment 411
Genome 58, 59 Herpes virus 409, 411, 500
Genomics 58 Hantavirus 454
classification of 410
Gentamicin, for plague 313 Haptens 87, 89
structure 409
German measles 492 complex 87
typical structure of 409f
Gerstmann–Straussler–Scheinker/ simple 87
Herpes zoster 412
GSS disease 490 HbcAg 470
Herpis febrilis 410
Giardia lamblia 613 HbeAg 470
Heterotrophs 22
Glomerulonephritis 177 HbsAg 470, 474 Hexachlorophane 31
distinguishing features Heavy chain disease 95 Hib PRP vaccine 320
of rheumatic fever and Hemolysins 175 Histamine 144
glomerulonephritis 177t Hemolytic disease of newborn 146, Histocompatibility antigens 155
Glomerulonephritis, due to Str. 160, 162 Histocompatibility complex 122
pyogene 176 Hemophilia 72 Histoplasma capsulatum 516, 518
Glucose nonfermenters 305 Hemorrhagic fever with renal Histoplasmin skin test 517
characteristics of 306t syndrome 454, 455 Histoplasmosis 515, 516, 518
Glutaraldehyde 33 Hemorrhagic viral fevers 497 HIV infection
Glycopeptides 567 Hendra viruses 444 clinical features of 480
Gonorrhea 192 Henipavirus 440 confirmatory tests 482
prophylaxis 193 Hepadnaviruses 382 infections and malignancies
treatment 193 Heparin 144 associated 481t

www.ebook3000.com
620  |  Essentials of Microbiology
postexposure prophylaxis 485 Immune response 125 Immunodiffusion 103
prophylaxis 485 cell-mediated 130 Immunodiffusion tests, types of 104
treatment 485 deficiencies of humoral and Immunoelectron microscopic
screening tests 482 cellular 141 tests 102
serological markers 481 primary 131 Immunoelectrophoresis 104
HIV virus 393, 476 secondary 131 Immunoenzyme test 115
major antigens of 477t type of 117, 125 Immunofluorescence 110
resistance 478 Immunity 4, 5, 81, 125 direct 111, 111f
routes of transmission 479, 479t acquired 81, 83, 86 indirect 111, 111f
HIV-1 476, 486 active 83, 84, 84t Immunoglobulin A/IgA 92
HIV-2 476, 486 artificial 84, 86 Immunoglobulin classes 92, 92t
HLA complex 122 natural 84, 86 Immunoglobulin D/IgD 94
HLA molecules 123 adoptive 85 Immunoglobulin deficiencies,
HLA typing 123 antibody-mediated 125 selective 138
Hookworm 544t, 597t, 610, 611, 613 cell-mediated 117, 125, 135 Immunoglobulin domain 92
Hortaea werneckii 509f humoral 125 Immunoglobulin E/IgE 94
Hospital-acquired infection 555 primary (central) organs 117 Immunoglobulin G/IgG 92
factors influencing 555 secondary (peripheral) Immunoglobulin M/IgM 94, 94f
Hot air oven 26, 26f, 27 organs 117 Immunoglobulin preparations 85
sterilization controls 27 cell-mediated 137 Immunoglobulin/Ig 90, 91
Human herpesvirus 6 415 cellular concept of 5 abnormal 94
Human herpesvirus 7 415 HERD 86 H-chains 91
Human herpesvirus 8 416 humoral or antibody- human 85
mediated 117 human serum 93t
Human T cell lymphotropic
individual 81
virus-III/HTLV-III 476 L-chains 91
innate 86
Human T-cell leukemia nonhuman 85
cellular factors in 83
(lymphotropic) Immunological response 395
mechanisms of 82
viruses/HTLV 501 Immunological tolerance 134; 135
local 86
Human virus infection, acquired tolerance 134
mechanical barriers 82
transmission of 393 natural tolerance 134
natural 81, 86
Humoral response Immunosuppressive agents 130
nonspecific 81
primary 126 Immunotherapy
passive 84t, 85
secondary 126 active 157
passive artificial 85
Hyaluronidase 79, 176 of cancer 157
passive natural 85
Hybridization 70, 589 racial 81 passive 158
Hydrogen peroxide 32 species 81 Impetigo 177
Hymenolepis nana 613 specific 81 Inactivated polio vaccine/IPV 428
Hyperimmune hepatitis B immune Immunization 4, 572 Inactivators 99
globulin/HBIG 471 active 572 Incineration 26
Hypersensitivity 142 combined 85 Incomplete immunogen 87
cell-mediated 143 influenza 86 Indole test 50f
classification of 142 National Immunization Kovac’s reagent 50
delayed 142, 148 Schedule 572, 573t Indonesian Weil’s disease 345t
granulomatous 149 passive 572 Infant death syndrome 444
immediate 142 indications of 85 Infections 75
type I 143, 146 Immunochromatographic tests atypical 76
type II 146 102, 114 cross 76
type III 147 Immunodeficiencies 137; 138 iatrogenic 76
type IV 148 cellular 138 inapparent 76
type V 149 combined 139 latent 76
Hypochlorites 32 common variable local 76
immunodeficiency 138 modes of transmission of 77
I humoral 137 nosocomial 76
primary 137, 141 primary 75
IgA, functions of 93 secondary 140, 141 route of 80
IgA, secretory 93 severe combined secondary 76
IgA, serum 93 immunodeficiency/SCID 139 subclinical 76
IgG, functions of 92 with thymoma 139 Infectious disease 75
Ilheus virus 450 Immunodeficiency diseases 137 types of 80
Immune complex disease classification of 137 Infective endocarditis 533, 535
local 147 Immunodeficiency syndromes, treatment 535
systemic 147 primary 138 Inflammation 83
Index  | 621
Influenza Lachrymal fluid, role in Listeriosis 355
chemoprophylaxis 438 immunity 83 clinical features 356
complications 436 Lactobacillus 222 Live-virus vaccines 399
diagnosis of; immunity Lactoperoxidase 83 advantages of 399
against 437 Lactose fermenter 607 disadvantages 399
pandemic 586 Lamivudine 485 Loeffler’s methylene blue 593
vaccines 438 Lassa fever 493 Lopinavir 485
uncomplicated 436 Latex agglutination test 107 Louis Pasteur 2, 7
Inhibitors 99 Lawn culture 43 contributions in microbiology 2
Inspissation 27 Lazy leukocyte syndrome 140 Louse-borne typhus 363
Interferons 132t, 133, 395 LDL-receptor deficiency 72 Lowenstein-Jensen medium 40, 40f
biological effects of 396 Lecithinase-C 79 Lyme disease 343
synthesis of 395 Lectin pathway 96, 100 stages of 343
types of 395 Legionella pneumophila 307 treatment 344
Interleukins 131, 132t clinical diseases 308 Lymphadenitis 250
Intestinal candidosis 520 morphology 307 Lymphatic recirculation 119
Introns 59 treatment 308 Lymphocytes 119
Iodophors 32 Legionnaire’s disease 308 Lymphocytic choriomeningitis
Isoantigens 88 Lens protein 88, 152 virus 493
Isograft 154, 158 Lepromin test 243, 246 Lymphogranuloma venereum 374,
Isolation, virus 398 Leprosy 241 548
Isospecificities 88 borderline 242 clinical manifestations 374
classification system of Ridley Lymphoid cells 119
and Jopling 241
J forms of 241
Lymphokine-activated killer/LAK
cells 121
Japanese ‘B’ encephalitis 450 immunoprophylaxis 245
Lysogenic conversion 65
Japanese encephalitis 446t, 448t, indeterminate type 242
Lysozyme 15
449, 450 lepromatous 242
Lyssavirus sera 463t
Jarisch–Herxheimer reaction 341 prophylaxis 245
JC virus 422,490, 500t tuberculoid (TT) 242
Job’s syndrome 140 Leptospira 344, 347 M
Joseph Lister 3 antigenic properties 345
MacConkey’s agar 606
classification 344
Macrolides 568
resistance 344
K Leptospiral infections 345
Macrophages 83, 121
functions of 122
Kaposi’s sarcoma 476 Leptospirosis 345
polymorphonuclear 122
Kaposi’s sarcoma-associated treatment 346
Maedi 489
herpesvirus 500 Leptotrichia 223
Magic bullet 5
Kawasaki syndrome 89 Leptotrichia buccalis 541
Major histocompatibility
Kelsey-Sykes test 34 Leukemia 4
complex 88
Keratomycosis 525, 526 Leukemia inhibitory factor 133
Leukocidin 166 Malignancy
Kerion 511
Leukocyte G-6-PD deficiency 140 immune response in 157
Killed viral vaccines 399
Leukocyte-associated antigen 88 immunology of 156
Kingella 195
Leukosis-sarcoma viruses 501 Mallein test 305
Kirby–Bauer disk diffusion
Leukotrienes 145 MALT See mucosa-associated
method 559, 560, 561f
LFNS See interferons 132t lymphoid tissue
interpretation chart 562t
Kissing disease 414, 416 LGV biovar 373 Marburg virus 494
Klebsiella granulomatis 357 LIF See leukemia inhibitory Marek’s disease 500
Klebsiella pneumoniae 534, 542t factor McCrady probability table 577t
Koch and Arnold steamer 27 Ligase chain reaction/LCR 589 Measles 443
Koch’s phenomenon 3, 235 Lincosamides 568 clinical features 443
Koch’s postulates 3 Liquid culture 44 complications 443
Koch-Weeks bacillus 320 Listeria ivanovii 355 morphology; prophylaxis 444
treatment 320 Listeria monocytogenes 355, Membrane attack complex 98
Kuru 489 360, 534 Meningitis 535
Kyasanur forest disease 453 laboratory diagnosis 357 aseptic 537
morphology 355 causative agent 536t, 537t
treatment 356 CSF findings in 537
L Listeria seeligeri 355 due to H. influenzae 319
LA crosse virus 454 Listeria, distinguishing examination of CSF 536
Lac operon 61 characters of 356t purulent 535, 538
of Escherichia coli 60f Listeriolysin-O 356 tuberculous 538

www.ebook3000.com
622  |  Essentials of Microbiology
Meningococcal septicemia 189 Morbillivirus 440, 441t Nasopharyngeal carcinoma 414
Mercuric chloride 32 Morganella 269 Negative staining for capsules 594
Messenger RNA 59 Morganella morganii 269 Neisser’s stain 594
Metapneumovirus 445 Mouth or oral cavity, role in Neisseria 188
Methyl red/MR test 50, 50f immunity 82 commensal 194, 195
Metronidazole; for Clostridium Mucormycoses 523 differential characteristics of 194r
difficle 219 clinical manifestations 523 Neisseria gonorrhoeae 191, 195
Microbial growth, requirements rhinocerebral 523 cultural characterstics 191
for 22 thoracic 523 morphology 191
Microbiology 2 treatment 524 opa protein 191
contributions of Louis Mucosa-associated lymphoid pili 191
Pasteur in 2 tissue 119 por proteins 191
scientific development of 2 Multiple myeloma 95 proctitis 192
Micrococci 170 Multiresistant salmonellae 285 resistance 192
Microorganisms 2 Mumps 441t, 442 rmp 191
role in diseases 2 Murray valley encephalitis types of 191
Microphages 122 virus 450 Neisseria meningitidis 188, 195, 534
Microscope 8 Mutagens 62 cultural characteristics 188
electron 10 Mutation 62 laboratory diagnosis 189
scanning 10 conditional 62 prophylaxis 190
transmission 10 frameshift 61f, 62 resistance 189
Microscopy 8 induced 62 stages of infections 189
lethal 62 treatment 190
atomic force 10
missense 62 Neorickettsia 366
compound light 8
nonsense 62 Neorickettsia sennestu 367
confocal 9
recognition of 62 Neosalvarsan 4, 5
darkground 9
spontaneous 62 Neutralization tests 110
differential interference
types of 62 toxin neutralization 110
contrast 9
Mycetism 512, 528 viral neutralization 110
electron 9, 10, 397
causative agents 512 Nichol’s strain 338
epifluorescence 9
Mycobacteria 227 Nicotinamide adenine
fluorescence 9, 397
classification of 227t dinucleotidase 176
immunoelectron 397
Mycobacteriophages 230 Nipah virus 440, 444
light 8, 397 Mycobacterium gordonae 248 Nitrate reduction test 51, 52f
phase-contrast 9 Mycobacterium leprae 240, 246 Nitrofurans 567
scanned-probe 10 morphology 240 Nitroimidazoles 567
scanning tunneling 10 Mycobacterium lepraemurium 245 NK-cells 121, 123
scanning-probe 10 Mycobacterium marinum 248 Nonsporing anaerobes
Microsporum 510 Mycobacterium szulgai 248 classification of 221t
M. audouinii 510 Mycobacterium tuberculosis 70, Nongonococcal urethritis 193, 195
M. canis 510 228, 238 Nonlactose fermenter/NLF
M. gypseum 510 cell wall of 230f colonies 607
Mild virus-like syndrome 345 cultural characteristics 228 Nonphotochromogens 248
Milk ring test 331 distinguishing features of Nonsporing anaerobes 221
Milk mycobacterium tuberculosis Nontuberculous mycobacteria 247
bacteriological and M Bovis 229t differentiating characterstics
examination of 578 multidrug resistant 237 of M. tuberculosis and
pasteurization of 27 Mycoplasma 12, 12f nontuberculous
Milk-borne diseases 578, 578t pneumoniae 542, 542t, 543 mycobacteria 247
Milker’s node 408 Mycoses 506 Runyon classification
Minimum infecting dose 80 classification of 506 scheme 248
Minimum inhibitory cutaneous 509 treatment 251
concentration/MIC 34 subcutaneous 511 Normal flora 530
Minimum lethal dose 80 superficial 508 harmful effects of 530
Mitogens 130 Mycotoxicosis 528 of conjunctiva 531
MMR vaccine 84, 444, 445, 493, 571 Mycotoxins 528t of gastrointestinal tract 531
Mobiluncus 222 Myeloperoxidase/MPO of genitourinary tract 532
Molluscipoxvirus 406t deficiency 140 of nose, nasopharynx and
Molluscum contagiosum 408 accessory sinuses 531
Monkeypox 408 of upper respiratory tract 531
N
Monocytosis 355 role of 530
Mononuclear cells 121 Nagler’s reaction 220, 213f Novobiocin 568
Moraxella lacunata 195 Nairovirus 389, 453, 454 Nucleic acid probes 70
Index  | 623
Nucleic acid sequence based Paul Ehrlich 3, 6t, 7 differential characters of
amplification/NASBA 589 Paul-Bunnel test 89, 107, 416 pneumococci and viridans
Nucleic acid structure 58 PCR, applications of 71, 73t streptococci 186t
Nucleotides 58 Penicillin laboratory diagnosis 185
Null cells 121 for Clostridium perfringens 215 meningitis 185
Nutrient agar 38 for lyme disease 344 pathogenesis 185
Nutrient broth 38 for Pneumococcus 186 pneumonia 185
for syphilis 341 prophylaxis 186
mechanisms of action 566 resistance 184
O neisseria meningitidis 190 treatment 186
Oncogenic viruses 499, 500t, 502 Penicilliosis 524 virulence factors 185
DNA viruses 499 Peplomers 380 Pneumocystis carinii
RNA viruses 500 Pepsin digestion 90 pneumonia 476
Onychomycosis 520 Peptococcus 221 Pneumocystis jiroveci 519, 524, 526
Onyong-nyong virus 449 Peptone water 38 life cycle of 525f
Ophthalmic neonatorum 192 Peptostreptococcus 222 Pneumonia 542
Opportunist mycobacterial Pertussis toxin/PT 324, 326 community-acquired 543
disease 249t Pertussis vaccine, acellular 325 due to H influenzae 319
Opportunistic fungi 519 Pfeiffer phenomenon 96 hospital-acquired 542
Opsonization 99, 102, 100, 110 Phaeohyphomycosis 513 in immunocompromized
Oral hairy leukoplakia 414 Phagocyte, disorders of 140 patients 543
Oral polio vaccine/OPV 428 Phagocytic cells 121 Pneumonia syndrome
differences between killed/IPV Phagocytosis 83 atypical 543
and live polio Phenylalanine deaminase test 53 typical 543
vaccines/OPV 429 Phlebovirus 389, 446t, 453, 454 Pneumovirus 440, 444
Orbivirus 498 Phosphorylation 23 Poliomyelitis
Orthomyxovirus 434 oxidative 23 abortive 427
differences between substrate-level 23 global eradication 429
orthomyxovirus and Photochromogens 248 nonparalytic 427
paramyxovirus 434t Picornaviridae 389, 426, 432 paralytic 427
properties of434 classification 426 progressive
Orthopoxvirus 406t properties of 426t postpoliomyelitis 427
Orthoreovirus 454, 496 Piedra 509 Poliovirus 427, 432
Oseltamivir, for influenza 438 Piedraia hortae 508, 509 antigenic properties 427
Otomycosis 525, 526 Pigeon fancier’s disease 147 clinical features 427
Oxidase test 52 Pili 17 resistance 427
Pinta 334, 341 Polychrome methylene blue 593
Pityriasis versicolor 508 Polymerase chain reaction 71, 72f,
P morphology of 508f 589
Pandemic 80 Plague 311, 315 Polyomaviruses 388, 421, 422
Papillomatosis bubonic 311 Ponder’s stain 594
oral 421 chemoprophylaxis 313 Pontiac fever 308
recurrent respiratory 421 laboratory diagnosis 312 Porphyromonas 223
Papillomaviruses 421 pathogenesis 311 Potassium cyanide test 53
Papovaviruses 499 pneumonic 311 Pour-plate culture 44
Paracoccidioides brasiliensis 516 prophylaxis 312 Poxvirus 382, 406, 500
Paracoccidioidomycosis 515 septicemic 311 antigenic structure 407
Parainfluenza viruses 441 treatment 313 morphology 406
Paramyxoviruses 440 vaccine 313 Prausnitz–Kustner/PK reaction 146
morphology 440 Plasma cells 121 Precipitation 102
Parapoxvirus 406t Plasmid transfer 65 mechanism of 103
Parasites 75 Plasmids 18, 60, 79 reaction 103
Paratyphoid bacilli 275 conjugative 60, 67 types of 103
Paratyphoid fever 279 nonconjugative 60 Primary sensitivity tests 563
Parvovirus 424, 587t Platelet-activating factor 145 Primary tuberculosis 231
Pasteurella multocida 314, 315 Pleomorphism 19 Prion diseases 489
morphology 314 Plesiomonas 295 Prions 35, 390
Pasteurization 2 Pneumococcus 183 Progressive multifocal
Pathogens bacteremia 185 leukoencephalopathy/
opportunist 75 biochemical reactions 184 PML 490
primary 75 bronchopneumonia 185 Prokaryotes 11
types of 75 cultural characteristics 183 differentiation from eukaryotic 11t

www.ebook3000.com
624  |  Essentials of Microbiology
Properdin pathway 96 Reoviridae 496 Rose-Waaler test 107
Propionibacterium 222 classification 496 Ross river virus 449
Prostaglandin 145 Reovirus 498 Rotavirus 496, 498
Proteases 145 Replica plating 63 Rotavirus diarrhea 496
Proteinase 176 Resistance factors/R factors 67 treatment 497
Proteus bacilli 267 characteristics of 67 Rubella 492
Proteus sp 267, 534t, 606 features of 67 clinical features 492
Providencia 269 Resistance transfer factor /RTF 67 prophylaxis 493
Prozone phenomenon 330 Resolution 8 Rubella vaccines 493
Pseudomonas aeruginosa 301, 306 Respiration Rubella virus 449, 492, 497
antigenic characteristics 302 aerobic 23 Rubivirus 492
pathogenesis 303 anaerobic 23
pigment production 302 Respiratory syncytial virus 440, S
resistance 303 441, 444
treatment 303 clinical features 444 S. delphini 169
Psoriasis 89 treatment 445 S. dysenteriae 271
Psychotropic agents 529 Respirovirus 440, 441t S. epidermidis 163
Pugh’s stain 594 Retroviruses 476 clinical infection 169
Purines 58 genes 477, 478t S. haemolyticus 169
Pyemia 80, 533, 535 structure 476 S. hyicus 169
Pyocin 66 Retroviruses 477t, 501 S. intermedius 168
Pyocyanin 302 Reye’s syndrome 436 S. lutrae 169
Pyomelanin 302 Rh (rhesus) blood group S. saprophyticus 163
Pyorubrin 302 system 160 S. sonnei 271
Pyoverdin 302 Rh antigens 162 Salmonella 274, 285
Pyrimidines 58 Rh compatibility 160 antigenic structure of 275, 275f
Pyrogallic acid 45 Rh factor 160 morphology 274
Rhabdoviridae 455 syndromes due to 279
Rhabdoviruses 457 Salmonella gastroenteritis 285
Q
Rheumatic fever 89 Salmonella septicemia 285
Q fever 367 due to STR Pyogene 176 Salmonella Typhi 534t
Quaternary ammonium Rhinosporidiosis 513 Salvarsan 4, 5
compounds/QUATS 31 Rhinoviruses 432 Saprophytes 75
Quellung-Ger swelling 16 clinical syndromes 432 Saprophytic mycobacteria 249
Rh-isoimmunization 161 Sarcoma, in fowls 4
R prevention of 161 SARS See Severe acute
Ribavirin 496 respiratory syndrome
R plasmids 67, 79, 570 Ribonucleic acid 58 Satellitism 318, 318f, 322
Rabies 4, 459 structure 59 Scarlet fever 176
clinical features 459 Ribosomes 59 Schultz-Dale phenomenon 144
postexposure prophylaxis 460, Rickettsia 362 Schwartzman reaction 149
462, 463 antigenic structure 363 Scotochromogens 248
preexposure prophylaxis 460, morphology 362 Scrapie 489
462, 463 resistance 363 Semliki forest virus 449
vaccines 3, 461 Rickettsia akari 365, 369 Sensitivity 102
Rabies virus 457, 459f Rickettsia conorii 365, 369 Septic shock syndrome 149
antigenic properties 458 Rickettsia mooseri 369 Septicemia 80, 533
resistance 458 Rickettsia prowazekii 369 Serotherapy 5
Radiations 29 Rickettsia rickettsii 369 Serotonin 144
ionizing 30 Rickettsia typhi 363 Serum opacity factor 176
non-ionizing 29 differences between R. typhi Serum sickness 147
Radioimmunoassay 111 (R. mooseri) and Severe acute respiratory
Rapid plasma reagin/RPR R. prowazekii 364 syndrome 495, 497, 498
tests 337, 338 Rickettsial diseases 365 Severe combined
Rat-bite fever 358 treatment 366 immunodeficiency disease/
Reagents 600 Rickettsial pox 365 SCID 72
Redox potential 24 Rideal-Walker test 34 Sexduction 66
Reiter protein complement Rifamycins 567 Sexually transmitted diseases/
fixation/RPCF test 338 Rimantadine 400 STDs 547
Reiter’s syndrome 374 Ring test 103 organisms causing 548t
Relapsing fever 342 Ritonavir 485 Shanghai fever 303
endemic Robert Koch 3, 5t, 7 Shigella 270
or louse-borne 342 Rocky mountain spotted cultural characteristics 270
or tick-borne 342 fever 364, 365 laboratory diagnosis 272
Index  | 625
Shigella boydii 271 Staphylococcus aureus 534t Stroke culture 44
Shigella flexneri 271 Staphylococcus epidermidis 169 Subacute endocarditis 181
Shock organs 143 Staphylococcus saprophyticus 169 causative agents of 534t
Shwachman’s disease 140 Stenotrophomonas maltophilia 304 Subacute sclerosing
Silver nitrate 32 Sterilization 25, 35 panencephalitis/SSPE 393, 490
Sindbis virus 449 dry heat 26, 26f Subcutaneous phycomycosis 514
Skin, role in immunity 82 intermittent 28 Sugar fermentation 49, 49f
Slide test 103 low temperature steam Sulfonamides, mechanism of
Slides 592 formaldehyde/LTSF 27 action of 568
Slot-blot and dot-blot assays 114 methods of 25, 26t Superantigens 89, 130
Slow virus disease 488, 490 moist heat 27 Superinfection immunity 404
classification 488 procedures of 34t Suppurative streptococcal disease 176
Smallpox 406 Sterilizing cycle 26 Surface-active agents 31
control of 407 Stokes disk diffusion Swimming pool granuloma 250
Sore mouth 408 method 559, 561 Swineherd’s disease 345t
Sore throat 541 Streak culture 43, 43f Syphilis 334, 335, 547
causative agents of 541t Street virus 458 acquired nonvenereally 336
South American hemorrhagic Streptobacillus moniliformis 358 associated with human
fevers 493 morphology 358 immunodeficiency virus/
Specificity 102 treatment 359 HIV infections 340
antigenic 88 Streptococcal gangrene 177 congenital 336
autospecificity 88 Streptococcal toxic shock diagnosis 109
heterogenetic 88 syndrome 177 diagnostic tests for 337t
organ 88 Streptococci 172, 174 endemic 334
species 88 immunity in 340
Alpha (α) hemolytic 173
Spirilla 12, 12f latent 335
beta (β) hemolytic 173
Spirillum minus 359
classification 172 primary 335
morphology 359
gamma (γ) or prophylaxis in 340
treatment 359
non-hemolytic 173 secondary 335
Spirochetes 12, 12f, 333
group B 179 tertiary 336
classification 333
group C 179 treatment 341
diseases caused by 334
group D 180 venereal 334
Spleen 119
group G 179
functions of 119
Spontaneous generation 1
Streptococcus agalactiae 174, 179, 181 T
Sporotrichosis 513 Streptococcus bovis 174
Streptococcus equisimilis 174 Taenia sp. 612
Spotted fever group 364
Streptococcus mitis 174 Taxonomy 56
Stab culture 44
Streptococcus mutans 174 T-cell activation 127
Stain
Streptococcus pneumoniae 183, T-cells 119, 120
differential 593
534t, 535 basis of functions 120
special 594
Streptococcus pyogenes 173, 174t, basis of surface markers 120
types of 592, 593
181, 534t, 605 blast transformation 119
Staphylococcal diseases 167
acute suppurative infections 178 cytotoxic 120
cutaneous infections 167
deep infections 167 antigenic structure 174, 175f delayed type-
toxin-mediated diseases 167 cultural characteristics 173 hypersensitivity 120
Staphylococcus 163, 605 enzymes 176 differentiation from B-cells 119
bacteriophage typing 167 laboratory diagnosis 178 regulatory 120
differentiation between morphology 173 suppressor 120
staphylococci and non-suppurative types of 120
micrococci 170 complications 178 TEGO compounds 31
epidemiology 167 nonsuppurative sequelae 177 Tetanolysin 216
Staphylococcus aureus 163, 170, 535 pathogenesis 176 Tetanospasmin 216
biochemical reactions 164 prophylaxis 179 Tetanus 216
cell surface proteins 165 pyrogenic exotoxins 175 laboratory diagnosis 216
cell-associated polymers 165 resistance 174 prophylaxis 217
cultural characteristics 164 toxins 175 treatment 217
enzymes 165 treatment 179 Tetanus toxin 216, 220
methicillin-resistant 170 Streptococcus salivarious 174 Tetracycline, for plague 313
morphology 163, 163f Streptokinase 79, 176 TGF See tumor necrosis factors
resistance 165 Streptolysin O 175 Th1 cells 120
sensitivity to antibiotics 169 Streptolysin S /SLS 175 Th2 120
toxins 165 Streptomycin, for plague 313 Thioglycollate broth 41, 46

www.ebook3000.com
626  |  Essentials of Microbiology
Thymus 117 Treponema pallidum immune Vancomycin
Tick typhus 364 adherence/TPIA test 339 for Clostridium difficle 219
Tick-borne encephalitis viruses/ Treponema pallidum particle for Pneumococcus 186
TBEV 452 agglutination/TP-PA 340 Varicella 412
Tick-borne hemorrhagic fevers 453 Treponema vincentii 541 Varicella-zoster
Tinea barbae 511 Treponemal tests 338, 347 immunoglobulin 413
Trichophyton 509 Varicella-zoster virus 393, 411, 416
Tinea corporis 511
Trichosporon beigelii 508 properties of 412
Tinea cruris 511
Trichuris trichiura 597t, 610, 611 prophylaxis and treatment 413
Tinea nigra 508 Triple-sugar iron/TSI agar 53, 54t Vectors 76, 77, 584
Tinea pedis 511 Tsutsugamushi disease 365 Vectors-borne diseases 584
Tinea unguium 511 Tube flocculation test 103 Vehicles 584
Tissue culture 384, 390 Tuberculin test 235, 238 Veillonella 222
Tobacco mosaic disease 4 Tuberculosis 231 Veneral disease research
Togaviruses 382, 447, 453, 492 extrapulmonary 234 laboratory/VDRL test 337
Toluidine red unheated serum test/ postprimary (secondary) 231 Venezuelan equine encephalitis/
TRUST 338 pulmonary 231 VEE 446t, 449, 455
Torch agents 77 Tuberculosis complex 227 Vertical transmission 77
Toxic shock syndrome toxin-I/ Tumor antigens 156 Vesicoureteral reflux 539
TSTT-1 166 oncofetal 156 Vibrio cholerae 287, 290, 544
Tumor associated carbohydrate morphology 287
Toxoids 79
antigens 156 pathogenesis 291
Trachoma 373
Tumor necrosis factors 132t, 133 resistance 288
Trachoma biovar 373, 377
Tumor-associated transplantation subtypes of 290
Transcobalamin II deficiency 138 antigens/TATAs 156 Vibrio parahaemolyticus 294, 544
Transcription 59 Tyndallization 28. 35 Vibrio vulnificus 295
Transcription mediated Typhoid fever 279 Vibrios 12, 12f
amplification/TMA 589, 590 laboratory diagnosis of 280 Vincent’s angina 343
Transduction 64, 68 prophylaxis 283 Viral assay 386
generalized 65 vaccines against 283 Viral capsid 379
role of 65 Typhus fever 369 Viral diseases
specialized 65 immunoprophylaxis of 399
types of 65 U pathogenesis of 393
Transfer factor 133, 135 Viral envelope 379
Ulcerative gingivostomatitis 343 Viral genetics 387
Transfer RNA/TRNA 60
Urease test 52, 52f Viral hemagglutination 381
Transformation 68
Urinary tract infection 538 Viral hemorrhagic fevers 493
Transforming growth factor causes of 539t Viral hepatitis 465
beta 132t, 133 clinical features 539 Viral infections
Transient differentiation of upper UTI and indications for 397
hypogammaglobulinemia of lower UTI 540 laboratory diagnosis of 397
infancy 137 lower 538 Viral nucleic acids 380
Transmissible spongiform tuberculosis of kidney and Viral oncogenes 502
encephalopathies 489 urinary tract 540 Viral oncogenesis 4
Transplantation 5, 158 upper 538 Viral proteins, detection of 398
Transplants 154 Viral replication 381
types of 154 V Viral vaccines, in common use 399t
Transposons 68 Viridans streptococci 180
structure of 68, 69f V factor 318 classification 181
Treponema 334 334 Vaccines 2, 84, 571 clinical infections 181
attenuated anthrax 2 Viroids 390
nonpathogenic 341
bacterial 84 Virulence 78
Treponema carateum 334
cellular fractions 571 determinants of 78
Treponema endemicum 334
chicken cholera 2 Virus diseases
Treponema pallidum 9, 334 killed 84, 85, 571 chemoprophylaxis and
agglutination/TPA test 338, 339 live 4, 84, 571 chemotherapy of 400
antigenic structure 335 mixed or combined 572 Virus infections 395
hemagglutination/TPHA multiple 129 antibody-mediated
107, 338, 339 recombinant-vector 572 immunity 395
immobilisation/TPI test 338 subunit 84 cell-mediated immunity 395
morphology 334 toxoids 571 immunological response 395
pathogenesis 335 viral 84 nonimmunological
resistance 335 Vaccinia virus 407 responses 395
Index  | 627
Virus neutralization 98 Water, peptone 38t Yaws 334
Virus symmetry 379 Water-borne diseases 576t Yeast 519, 503
complex symmetry 379 Waterhouse–Friderichsen Yeast-like fungi 503, 519
helical symmetry 379 syndrome 149 Yeasts 503
icosahedral symmetry 379 Watson-Crick structure, of Yellow fever 4
Viruses 4, 390, 378 DNA 59f Yellow fever virus 449, 451, 455
baltimore classification of 382 Weil-Felix reaction 89, 107, 366, 369 Yersinia 309
cultivation of 383 West nile virus 450 Yersinia enterocolitica 314, 315, 545
direct detection of 397 Western blot test 482 Yersinia pestis 309
discovery of 4 Western equine encephalitis/WEE biotypes of 310f
DNA, double stranded 382 448t, 449, 455 morphology 309
double stranded 382 White piedra 509 resistance 310
incubation period of 394 Whooping cough 324 toxins 310
morphology of 378 complications 324 virulence factors 310
properties of 378 stages of 324 Yersinia pseudotuberculosis 313
RNA 389, 389f Widal test 107, 282 treatment 314
single stranded 382 Wilson and Blair medium 40 Yersiniosis 313
Visna 488 Wiskott–Aldrich syndrome 139
Vitronectin 99
Voges-Proskauer/VP test 50, 51f Z
X
Zaire strain 494
W Xenograft 154, 158
Zanamivir, for influenza 439
X-linked agammaglobulinemia 137
Waldenstrom’s X-linked hyper-IgM syndrome 138 Zidovudine 400, 485
macroglobulinemia 95 Ziehl-Neelsen staining 48, 593, 598
Warts 421 Zone phenomenon 102
Y Zoonosis 76
anogenital 421
cutaneous 421 Yatapoxvirus 406t Zygomycosis 523, 526

www.ebook3000.com

You might also like