Top Banner
i DETECTION AND IDENTIFICATION OF ACTINOBACILLUS PLEUROPNEUMONIAE SEROTYPE 5 BY MULTIPLEX POLYMERASE CHAIN REACTION by Terry M. Lo Thesis submitted to the Faculty of the Virginia Polytechnic Institute and State University in partial fulfillment of the requirements for the degree MASTER OF SCIENCE in Veterinary Medical Sciences APPROVED: ________________________ Thomas J. Inzana, Chair ________________________ _______________________ Nammalwar Sriranganathan Eric A. Wong July 1997 Blacksburg, Virginia
112
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: At

i

DETECTION AND IDENTIFICATION OFACTINOBACILLUS PLEUROPNEUMONIAE SEROTYPE 5

BY MULTIPLEX POLYMERASE CHAIN REACTION

by

Terry M. Lo

Thesis submitted to the Faculty of theVirginia Polytechnic Institute and State University

in partial fulfillment of the requirements for the degree

MASTER OF SCIENCE

in

Veterinary Medical Sciences

APPROVED:

________________________Thomas J. Inzana, Chair

________________________ _______________________Nammalwar Sriranganathan Eric A. Wong

July 1997Blacksburg, Virginia

Page 2: At

ii

DETECTION AND IDENTIFICATION OFACTINOBACILLUS PLEUROPNEUMONIAE SEROTYPE 5

BY MULTIPLEX POLYMERASE CHAIN REACTION

by

Terry M. Lo

Dr. Thomas J. Inzana, Chair

Department of Veterinary Medical Sciences

(ABSTRACT)

Traditional serologic assays of Actinobacillus pleuropneumoniae often have

problems with cross-reactivity. To avoid the complications of antibody-antigen

reactions, a PCR assay was developed to detect Actinobacillus pleuropneumoniae and

identify serotype 5 strains. Primers specific to the conserved capsular export region of A.

pleuropneumoniae amplified a 0.7 kb DNA band in all strains with the exception of

serotype 4. A second set of primers specific to the unique capsular biosynthesis region of

serotype 5 amplified a unique 1.1 kb band for serotype 5 only. The sensitivity of this

assay was determined to be less than 102 colony forming units. This PCR assay enables

us to detect A. pleuropneumoniae and definitively distinguishes serotype 5 strains from

other serotypes.

Page 3: At

iii

This thesis is dedicated to my parents,

Julie and Christopher Lo, a.k.a. Mom and Dad.

Page 4: At

iv

ACKNOWLEDGMENTS

It has been my good fortune to have worked with a number of great people that

have helped make my graduate experience worthwhile and enjoyable. I would like to

thank Dr. Thomas Inzana, my graduate advisor, for all of his guidance, support and

patience. .My committee members, Dr. Eric Wong and Dr. Nammalwar Sriranganathan,

for their excellent suggestions and advice. I greatly appreciate the time and effort they

have put forth. I would also like to thank Christine Ward for helping me get on track

when I first started, Gretchen Glindemann for all of her continuing help, and John

McQuiston for allowing me to pester him with millions of questions. Thanks to Mark

Lawrence, Todd Pack, and Rhonda Wright, who have all showed me great kindness as

well as technical advice in the lab. Thanks to Dr. John Lee and Dr. Lud Eng for their

financial support, and Linda Price, Sherrie Settle, Kim Stowers, and Tracie Sweeny for

their help with all of the administrative details. And let’s not forget about my friends and

co-workers: Mike Howard, Maureen Fallon, Jennifer Hensley, Jane Lee, Sergio Harding,

Noel Hikes, Dave Copeland, everyone at the CMMID, the media lab, and all my fans.

Thanks.

Page 5: At

v

TABLE OF CONTENTSPage

Abstract ..................................................................................................................... ii

Dedication................................................................................................................. iii

Acknowledgments .................................................................................................... iv

List of Figures .......................................................................................................... vii

List of Tables ............................................................................................................viii

Chapter 1 Introduction....................................................................................... 1

Literature Review.................................................................... 1

Actinobacillus pleuropneumoniaeCharacterization and History........................... 1Epidemiology................................................... 2Clinical Signs................................................... 4 Pathology......................................................... 5Virulence Factors............................................. 6

Toxins.................................................... 6Lipopolysaccharide............................... 9Capsule.................................................. 10

Prevention and Control...................................... 11

Identification and Serotyping of Actinobacilluspleuropneumoniae

Introduction......................................................... 14Complement Fixation Test.................................. 16Indirect Hemagglutination Test........................... 17Enzyme Linked Immunosorbent Assay............... 19Agglutination and Coagglutination Tests............. 21Latex Agglutination.............................................. 23Indirect Fluorescent Anitbody Test......................24Immundiffusion Test............................................ 25Ring Precipitation ................................................ 26Biochemical Typing.............................................. 27

Page 6: At

vi

Polymerase Chain Reaction (PCR)...................... 28Summary.............................................................. 30

The Polymerase Chain Reaction and ItsUses in Microbial Detection

History.................................................................. 32Principle of the Polymerase Chain Reaction......... 34PCR and Bacteria.................................................. 41Summary............................................................... 43

Chapter 2 Detection and Identification of Actinobacilluspleuropneumoniae Serotype 5 by MultiplexPolymerase Chain Reaction

Abstract..........................................................................................45

Introduction....................................................................................46

Materials and MethodsStrains and cell cultures.....................................................50Tissue and nasal swab samples..........................................52DNA isolation....................................................................52Multiplex PCR...................................................................52

Sample preparation................................................52Primers ..................................................................53PCR conditions......................................................56Detection................................................................57

Sensitivity..........................................................................58Probes................................................................................58Southern Blotting...............................................................58

Results............................................................................................59Assay of serotype 5 genomic DNA...................................61Assay optimization............................................................77Assay sensitivity...............................................................79Assay specificity...............................................................82Assay of clinical specimens...............................................85

Discussion......................................................................................88

LiteratureCited................................................................................................................92

Page 7: At

vii

List of FiguresChapter 1 Page

Figure 1.1 Schematic diagram of PCR...........................................................36

Chapter 2

Figure 2.1 Map of capsular region and location of primers............................55

Figure 2.2 PCR of serotype 5 genomic DNA.................................................60

Figure 2.3 Serotypes 1-6 at 5 mM MgCl2......................................................63

Figure 2.4 Serotypes 1-6 with reduced cps primers........................................65

Figure 2.5 Serotype 2 single set of primers.....................................................67

Figure 2.6 Serotype 4 single set of primers.....................................................68

Figure 2.7 Serotype 2 at varying Taq polymerase concentrations..................70

Figure 2.8 Serotype 2 at varying MgCl2.........................................................72

Figure 2.9 PCR of serotype 2 sample types...................................................74

Figure 2.10 Serotypes 1-12 at 2 mM MgCl2....................................................76

Figure 2.11 Sensitivity assay............................................................................78

Figure 2.12 Respiratory swine pathogens.........................................................80

Figure 2.13a Gel used for Southern hybridization..............................................82

Figure 2.13b Hybridization of cpx probe............................................................83

Figure 2.14 PCR of lung tissue.........................................................................85

Figure 2.15 PCR of nasal swabs with genomic DNA.......................................87

Page 8: At

viii

List of Tables

PageChapter 2

Table 2.1 Bacterial strains..............................................................................51

Table 2.2 Primer sequences...........................................................................54

Page 9: At

1

Literature Review

Actinobacillus pleuropneumoniae

Characterization and History

Actinobacillus pleuropneumoniae is the etiological agent of swine

pleuropneumonia, a respiratory disease that continues to have a worldwide economic

impact. Swine are the only known natural hosts of the disease, but it is highly contagious

and can be spread from herd to herd by infected carriers (MacInnes et Rosendal, 1988).

Symptoms of the infections caused by A. pleuropneumoniae may vary from death to

chronic illness to subclinical symptoms. Attempts to control and prevent the disease

have been largely ineffective (Sebunya and Saunders, 1983).

The first field isolates of A. pleuropneumoniae were reported in the early 1960's

in Great Britain, California, and Argentina (Matthew and Pattison, 1961; Shope, 1964).

At that time the isolates were typed as one of three species: Haemophilus parainfluenzae,

Haemophilus parahaemolyticus, or Haemophilus pleuropneumoniae. However,

biochemical tests of the organisms' ability to ferment certain sugars established that they

did not belong to the already existing species of H. parainfluenzae. The designations of

H. parahaemolyticus and H. pleuropneumoniae were often used interchangeably. In

1978, Kilian et al. established that the human isolates of H. parahaemolyticus were clearly

distinct from swine isolates and should be classified in separate categories. The swine

Page 10: At

2

isolates were then categorized as H. pleuropneumoniae, a name that had been originally

proposed by Shope (1964).

In 1983, DNA hybridization studies by Pohl et al. indicated that there was no

significant homology between H. pleuropneumoniae and H. influenzae. However, the

homology between H. pleuropneumoniae and Actinobacillus lignieresii showed significant

relatedness. This resulted in a proposal to change the genus name from Haemophilus to

Actinobacillus (Pohl et al. 1983). The official name of the organism is now Actinobacillus

pleuropneumoniae, and it is described as a gram-negative, encapsulated, facultative

anaerobic, non-spore forming, nonmotile pleomorphic coccobacillus (Kilian et Biberstein,

1984). A. pleuropneumoniae that require nicotinamide adenine dinucleotide (NAD) for

growth are designated as biovar 1 while A. pleuropneumoniae isolates that are NAD

independent are designated as biovar 2 (Pohl et al. 1983). In addition, there are 12

serotypes of biovar 1 that are distinguished by their unique capsular polysaccharide

(Perry et al., 1990).

Epidemiology

Transmission of the disease appears to occur directly from an infected pig to a

susceptible pig, since A. pleuropneumoniae is not known to survive long in the

surrounding environment (Willson et al., 1987). However, in acute outbreaks the disease

agent may be transmitted from one pen to another, suggesting that transmission by

Page 11: At

3

aerosol or by farm personnel carrying contaminated exudate of infected pigs is possible

(Nicolet, 1992). Because pigs may carry the bacterium undetected, the disease is often

spread by the introduction of new pigs into the herd (Kume et al., 1986). The probability

of a herd being exposed to A. pleuropneumoniae increases with the number of pigs

purchased at one time (Rosendal and Mitchell, 1983). Although all age groups are

susceptible to the disease, growing pigs seem to be the most susceptible. Factors such as

moving, crowding, and adverse climate conditions play a large role in supporting the onset

and severity of the disease (Nicolet, 1992).

Serotypes of A. pleuropneumoniae are generally distributed by geographic

location. Serotypes 1, 5 and 7 are most commonly found in the United States, while

other serotypes predominate in other parts of the world (Sebunya and Saunders, 1983).

While some serotypes are considered to be of low virulence in some countries and of no

epidemiologic importance, they may be agents of disease in others countries (Desrosiers

et al., 1984). Although there are apparent geographic boundaries, there remains the

possible danger that a new serotype will be introduced into a naive herd (Fedorka-Cray et

al., 1993). A typical outbreak of A. pleuropneumoniae results in about 50% morbidity

and up to 10% mortality (Fenwick et Henry, 1994).

Page 12: At

4

Clinical Signs

An outbreak of A. pleuropneumoniae can result in varying degrees of clinical

disease: peracute, acute, and chronic. In the peracute form, pigs become suddenly ill and

reach fevers of 106.7 F (Nicolet, 1992). Vomiting, diarrhea, and a discharge of blood-

tinged foam from the nostrils and mouth may occur as well as an increase in pulse rate

(Nielsen, 1985; Nicolet, 1992). These symptoms are usually followed by circulatory

failure and cyanosis of the body (Nicolet, 1992). Death occurs within 24-36 hours

(Shope, 1964). Occasionally, an animal may die suddenly without showing the initial

indications of illness (Sebunya and Saunders, 1983).

The acute form of the illness also results in a rise in body temperature, lameness,

stiffness, and lethargy (Fenwick and Henry, 1994). Animals often become depressed and

show a loss of appetite. Respiratory difficulties become evident and circulatory failure

may occur. The acute form of the disease may lead to death or recovery (Nicolet, 1992).

If the pig is able to survive the first four days of the outbreak, it is likely to survive

(Shope, 1964). However, a chronic form of the disease often persists in those animals

that survive the disease (Nielsen, 1985).

Pigs that suffer from a chronic form of pleuropneumonia may exhibit only

subclinical signs (Sebunya and Saunders, 1983). Little or no fever is observed as well as

only occasional coughing. Appetite is decreased resulting in a diminished weight gain

(Nicolet, 1992). In many cases, the only sign of infection is evident at necropsy by

Page 13: At

5

unresolved lung lesions (Shope, 1964). Other respiratory diseases or stress factors may

trigger an increase in pleuropneumonia symptoms (MacInnes and Rosendal, 1988).

Chronic carriers may also transmit the disease to previously unexposed swine (Kume et

al. 1986).

Pathology

Necropsy studies of swine that have been infected by A. pleuropneumoniae

display pneumonic lesions mainly in the respiratory tract (Nicolet, 1992). While the

lesions are found mainly on the caudal lobes, they can also occur in cranial and median

lobes (Sebunya and Saunders, 1983). Lung lesions on acutely affected swine are dark

pink to plum red; there is generally blood stained froth in the trachea, excess bloody fluid

in the thorax, and fibrinous adhesions between lungs, thoracic wall, diaphragm, and

pericardium (Rogers et al., 1990). Most studies have concluded that the characteristic

lesions induced by A. pleuropneumoniae infection are a result of bacterial toxins (Bertram,

1986). Evidence of this is supported by the presentation of similar lesions when

sonicated bacteria and bacterial supernatant were used (Rosendal et al., 1980).

In pigs that suffer from chronic symptoms, determination of A. pleuropneumoniae

infection can be difficult. Severe lung lesions induced by A. pleuropneumoniae can resolve

within a few weeks, and chronic lung lesions can be difficult to differentiate from lesions

caused by various other organisms (Fenwick and Henry, 1994). These lung lesions are

mottled red to yellow and are less extensive with more pronounced fibrosis (Rogers et al.,

Page 14: At

6

1990). The chronic lesions may harbor A. pleuropneumoniae for months, and infected

animals are considered to be carriers of the infection (Fedorka-Cray et al., 1993). In some

cases, the presence of low numbers of A. pleuropneumoniae introduced by subclinical

carriers may provide some herd immunity against the disease (Fenwick and Henry, 1994).

However, the effect of subclinical infection on growth can be substantial (Rohrbach et al.,

1993).

Virulence Factors

There are a number of factors that significantly contribute to the pathogenic

properties of A. pleuropneumoniae. Three of the most characterized of these factors are

exotoxins, lipopolysaccharide, and capsule. These factors may also play key roles in

determining the protective and identifying antigens associated with A. pleuropneumoniae.

The following is a short summary of these three elements.

Toxins

After it had been shown that the supernatant from A. pleuropneumoniae

suspensions could produce the necrotic and hemorrhagic lesions associated with

pleuropneumonia, A. pleuropneumoniae exotoxins have been established as being directly

involved in generating clinical signs (Rosendal et al., 1980). The hemolytic and cytotoxic

characteristics that have been associated with A. pleuropneumoniae have been attributed

Page 15: At

7

to at least three toxin proteins. Early analysis of these proteins have identified them as

members of the RTX-toxin family (Lalonde et al., 1989). These toxins have been named

Apx-toxins for Actinobacillus pleuropneumoniae RTX-toxins and are identified as ApxI,

ApxII, and Apx III (Frey et al., 1993). Each of these toxins vary in their hemolytic and

cytotoxic activity and their presence in A. pleuropneumoniae depends on the serotype of

the strain (Frey, 1995).

ApxI is a strongly hemolytic and cytotoxic protein with a molecular weight of

105-110 kilodaltons (kDa) (Frey and Nicolet, 1988). It is produced and secreted by A.

pleuropneumoniae serotypes 1, 5, 9, 10, and 11 (Frey and Nicolet, 1990; Kamp et al.

1994). The nucleotide sequence that encodes for ApxI has been identified as the operon

apxI, and consists of four genes arranged in the respective order of apxIC, apxIA, apxIB,

and apxID (Gygi et al., 1992; Jansen et al. 1993). The toxin expression is induced by

calcium and is necessary for biological activity (Devenish and Rosendal, 1991). Protein

analysis of the toxin shows that it is 56% homologous to the Escherichia coli hemolysin,

HlyA (Femlee et al., 1985). Strains that produce ApxI tend to be highly virulent,

supporting evidence that toxin activity is related to the virulence of A. pleuropneumoniae

(Kamp et al., 1991; Frey and Nicolet, 1990).

ApxII is weakly hemolytic and weakly cytotoxic and has a molecular mass of

between 103-105 kDa (Frey and Nicolet, 1988). All of the A. pleuropneumoniae

serotypes with the exception of serotype 10 produce and secrete ApxII (Kamp et al.,

Page 16: At

8

1991; Kamp et al., 1994). Protein analysis of ApxII has found it to be 72% homologous

to the leukotoxin of Pasteurella haemolytica (Chang et al., 1989). However, the apxII

operon does not contain any genes that are responsible for secretion (Frey, 1995).

Apparently, the secretion of ApxII is dependent on the apxIBD genes which are found in

all of the A. pleuropneumoniae serotypes with the exception of serotype 3 (Frey, 1995).

ApxIII is not hemolytic but is strongly cytotoxic with a molecular mass of 120

kDa (Kamp et al., 1991). The protein is produced and secreted by A. pleuropneumoniae

serotypes 2, 3, 4, 6, and 8 (Kamp et al., 1991). ApxIII is 50% identical to ApxI and E.

coli HlyI (Jansen et al., 1993). The operon encoding for ApxIII consists of the genes

apxIIICABD and shares an analogous arrangement of the apxI operon (Chang et al., 1993).

The virulence of certain A. pleuropneumoniae serotypes varies from highly

virulent to weakly virulent. These variations are consistent with the type of toxin each

serotype produces. Those serotypes that produce ApxI or those that produce two toxins

are particularly virulent when compared to those serotypes that do not (Kamp et al.,

1991; Frey and Nicolet, 1990). Serotype 5 mutants that do not secrete ApxI or ApxII

have been shown to be non-pathogenic in pigs or mice, indicating that the toxins are

important virulence factors for A. pleuropneumoniae serotype 5 (Inzana, 1991). In

addition this mutant was shown to be non-protective against the wild-type strain

suggesting that the toxins are needed to provide a sufficient immune response against the

pathogenicity of A. pleuropneumoniae serotype 5 (Inzana, 1991).

Page 17: At

9

Lipopolysaccharide

Lipopolysaccharide (LPS) serves as a major component of the outer membrane of

A. pleuropneumoniae and is capable of causing significant tissue damage. Its composition

consists of lipid A, a core region of common sugars, and an O-polysaccharide side chain

(Byrd and Kadis, 1989). The O-side chains of the A. pleuropneumoniae serotypes vary

from smooth, to semi-smooth, to rough (i.e. strains that lack O-side chains). Each of these

serotypes has a particular composition and structure of the LPS O-side chain (Fenwick

and Osburn, 1986; Byrd and Kadis, 1989). Although in some cases, the similarity of the

O-side chain of some serotypes may be responsible for the immunological cross-

reactivity that is observed in A. pleuropneumoniae (Fenwick and Osburn, 1986).

Although pure LPS has the potential to cause damage to lung tissue, the damage

differs from the hemorrhagic and necrotic lung lesions found in typical cases of A.

pleuropneumoniae infection (Udeze et al., 1987). It is likely that LPS and exotoxins are

able to interact to intensify the virulent effects of A. pleuropneumoniae (Inzana, 1991).

In addition, LPS is believed to play a role in the adherence of A. pleuropneumoniae.

Belanger et al. (1990) reported that 83% of serotypes with smooth LPS adhere to the

tracheal rings in large numbers, while 80% of serotypes with semi-smooth LPS adhere

poorly. This suggests that LPS may be a necessary factor in the colonization of A.

pleuropneumoniae to the upper porcine respiratory tract.

Page 18: At

10

Capsule

All 12 A. pleuropneumoniae serotypes are distinguished by capsular

polysaccharide of specific composition and structures that are immunologically unique,

and have been previously characterized (Perry et al., 1990). These capsules are

negatively charged and are composed of oligosaccharide units, techoic acid polymers

joined by phosphate diester bonds, or oligosaccharide polymers joined by phosphate

bonds (Perry et al., 1990). Encapsulated A. pleuropneumoniae can be visualized as an

iridescent hue surrounding the colonies when plated on clear medium (Inzana, 1990).

The A. pleuropneumoniae capsule is poorly immunogenic (Fenwick and Osburn,

1986; Inzana and Mathison, 1987a). Purified capsule fails to activate the complement

cascade and does not demonstrate toxic activity (Ward and Inzana, 1994; Fenwick and

Osburn, 1986). No clinical symptoms or lung lesions were found when purified capsule

was administered endobronchially to pigs (Fenwick and Osburn, 1986). The main role of

the capsule appears to be one of protection for the pathogen. Encapsulated A.

pleuropneumoniae is protected from bactericidal killing by complement in the presence

and absence of capsule specific antibody (Inzana et al., 1988). However, a

nonencapsulated mutant of serotype 5 was shown to be susceptible to complement

killing in the absence of specific antibody (Ward and Inzana, 1994). This suggests that

capsule plays an important part in the resistance of A. pleuropneumoniae to complement

Page 19: At

11

mediated killing. In addition, different serotypes have varying degrees of virulence.

Serotypes possessing larger and more adherent capsule were shown to be more virulent

than serotypes with less capsule (Jensen and Bertram, 1986). This indicates that capsule

may be one of several factors that influence the virulence of different serotypes.

Prevention and Control

There are several methods that can be employed for the prevention and control of

A. pleuropneumoniae in swine herds. In order to choose the most desirable method, it is

important to weigh all factors from financial cost to risk of disease outbreak. Some herd

owners may choose to live with the subclinical symptoms of the less virulent strains of A.

pleuropneumoniae. This reduces the risk of an acute outbreak that may occur in A.

pleuropneumoniae free herds (Fenwick and Henry, 1994). Risk of introducing A.

pleuropneumoniae to the herd can be further reduced by culling seropositive carriers,

purchasing seronegative stock, and quarantining new stock (Rosendal and Mitchell, 1983).

In addition, the herds should be managed between barns and rooms with an all-in and all-

out policy, with thorough cleanings between groups (Fenwick and Henry, 1994).

Subclinically infected carrier pigs are by far the most common means of

transmitting A. pleuropneumoniae (Fenwick and Henry, 1994). Although eradication of

carrier pigs is an ideal method of control, it is often impractical to culture an entire herd.

Page 20: At

12

This may leave the treatment of clinical symptoms as the only recourse (Fedorka-Cray et

al., 1993). However, the use of antibiotics often does not clear the entire infection, and A.

pleuropneumoniae may still be shed (Willson and Osborne, 1985). In addition, the

isolation of antibiotic-resistant strains is becoming increasingly common (Fedorka-Cray et

al., 1993).

Herd depopulation is the most radical alternative to eliminate a disease outbreak.

It consists of removing all animals from the farm site and repopulating with animals from

disease free herds. In cases where there is a high prevalence of seropositive pigs in the

herd, depopulation may be the only effective method of treatment (Nicolet, 1992). This

is, however, a very expensive method and may result in the loss of important bloodlines

(Leman, 1992). This has created an increasing demand for early detection of potential

carriers of the disease by serologic testing. If new policies and management are not

incorporated for improved herd conditions, the risk of pleuropneumonia outbreak will

remain (Fenwick and Henry, 1994).

A variety of vaccines have been developed to provide protection against A.

pleuropneumoniae. However, vaccines have shown some efficacy in experimental

settings, field observations have been less conclusive (Hunneman, 1986). The available

commercial vaccines also do not prevent pigs from becoming subclinical carriers (Nicolet,

1992). The current use of A. pleuropneumoniae vaccines is not a reliable means to

prevent infection and is intended to reduce the severity of illness and death (Fenwick and

Page 21: At

13

Henry, 1994). Recently, a nonencapsulated mutant of serotype 5 has been developed and

shows promise as a possible vaccine candidate (Ward and Inzana, 1995).

Page 22: At

14

Identification and Serotyping of Actinobacillus pleuropneumoniae

Introduction

Actinobacillus pleuropneumoniae is the etiological agent of swine

pleuropneumonia and has resulted in severe economic losses to the swine industry. To

date, there have been at least 12 serotypes identified from field isolates of A.

pleuropneumoniae with specific serotypes predominating in different geographical

regions. Serotypes 1, 5, and 7 are found most frequently in North America (Fenwick and

Henry, 1994), while serotype 2 is found most frequently in Switzerland, Denmark, and

Sweden (Nicolet, 1992).

A. pleuropneumoniae can be isolated from field samples by streaking onto 5%

blood agar plates with a cross-streak of a -toxinogenic, NAD-producing Staphylococcus

aureus strain. Colonies of A. pleuropneumoniae usually produce a -hemolytic zone. A.

pleuropneumoniae is a gram-negative, encapsulated, nonmotile, nonspore forming,

coccobacillary, facultative anaerobe; it is also urease positive and ferments mannitol,

xylose, ribose, and sometimes lactose (Fedorka-Cray et al., 1993).

Serovar classification is based on the capsular polysaccharide antigen (Inzana and

Mathison, 1987a). However, despite the various methods available for serotyping, cross-

reactions often occur unless highly purified reagents are used. Cross-reactions occur most

commonly between serotype 8 and serotypes 3 and 6, between serotype 9 and serotype

Page 23: At

15

1, between serotype 5 and serotype 7, and between serotype 7 and serotype 4 (Rosendal

and Boyd, 1982; Rapp et al., 1985b). Cross-reactions are most likely the result of some

serotypes sharing the same or similar somatic antigens. In addition, since 1990, 3% to

15% of A. pleuropneumoniae isolates typed at Iowa State University have been classified

as untypeable with respect to their serotype (Fedorka-Cray et al., 1993).

Traditional methods of A. pleuropneumoniae detection and serotyping have relied

on antibody based assays. Some of these serotyping methods can be used both for

serotyping and for the species detection of A. pleuropneumoniae. The difficulty in

developing serotyping assays is the preparation of highly purified serotype-specific

antigens or antisera. These preparations can often be very expensive and very time

consuming, but are essential for developing a serotype-specific assay. Speed and

simplicity as well as sensitivity and specificity are all important keys for a good

diagnostic protocol.

Since the 1990's the use of the polymerase chain reaction (PCR) has proven to be

a powerful alternative to traditional serological or immunological based diagnostics.

Several methods of A. pleuropneumoniae detection and serotyping have already employed

PCR as a diagnostic tool (Sirois et al., 1991; Hennessy et al., 1993). However, there have

not been any serotype-specific DNA sequences found as of yet, these methods do not

reach their full potential as a diagnostic method. Once the appropriate specific primer

sequences are identified, PCR should provide a simple method for the identification and

Page 24: At

16

serotyping of A. pleuropneumoniae as well as giving greater sensitivity and specificity

than previous tests.

Complement Fixation Test

The complement fixation test (CFT) was one of the earliest tests used to

serodiagnose A. pleuropneumoniae. It was first described by Nicolet in 1971 as a method

of diagnosing porcine pleuropneumonia and quickly became an early standard for

serotyping A. pleuropneumoniae (Hoffman, 1989). Originally intended as a method for

detecting A. pleuropneumoniae, it was later modified to be serotype specific (Nielsen,

1979). This test relies on having serotype specific antigens to each of the 12 serotypes

and reacting them against the test sera.

The following is a summary of the CFT as described by Lombin et al. (1982).

Antigens for the CFT are prepared by sonicating a suspension of the bacteria and

collecting the supernatant. In addition to the A. pleuropneumoniae antigens, the test

requires the presence of guinea pig complement, calf serum, sheep red blood cells

(SRBC’s) and the test serum. After the test serum has been heat-inactivated, it is

combined together with the complement and calf serum. The prepared antigens are then

added to the mixture. The reactions are incubated overnight to fix the complement.

SRBC’s are added to the mixture the next day. The lysing of SRBC's by the interaction

of complement and bovine serum is used as the indicator for the test. If the swine serum

Page 25: At

17

contains serotype specific antibodies to the antigen, they will interact with the A.

pleuropneumoniae antigen and in turn bind to the complement. This will inactivate or fix

the complement and prevent it from lysing the SRBC's. Thus, there is an inverse

relationship between the presence of A. pleuropneumoniae antibodies and the lysing of

SRBC's.

The CFT had been found to be more immunologically specific than the indirect

hemagglutination test, because it was able to distinguish A. pleuropneumoniae from

Haemophilus parasuis, another swine respiratory pathogen (Lombin et al., 1982; Nielsen,

1974). However, Lombin et al. (1982) reported that they were unable to observe any

serotype specificity by the CFT. Although the complement fixation test has been

frequently used for diagnosis and serotyping in the past, it requires a high level of

standardization and is relatively insensitive when compared to other assays, such as the

enzyme-linked immunosorbent assay (ELISA). In addition, performing this test can be

both laborious and cumbersome.

Indirect Hemagglutination Test

The indirect hemagglutination (IHA) test is another test that detects antibodies in

serum and has been used for many years as a method of bacterial detection (Neter, 1956;

Herbert, 1967). Nielsen (1974) first reported using a modification of the Herbert method

Page 26: At

18

to detect the presence of A. pleuropneumoniae antibodies. Later, Mittal et al. (1983a)

used the IHA test as a method for serotyping A. pleuropneumoniae.

The following is a procedure for the IHA test that has been described by Mittal et

al. (1983a). The antigen is prepared by suspending the bacterial cells in 0.85% saline

overnight and then collection by centrifugation. The supernatant is saved and is referred

to as the saline extract. The saline extract is then incubated together with SRBC’s for an

hour. The bacterial antigens adsorb to the surface of the SRBC’s and are ready to be used

in the IHA test.

The sera is prepared by absorbing it to unsensitized SRBC’s to remove all non-

antigen specific antibodies. The sensitized SRBC’s are placed into the wells of a

microtiter plate. Serial twofold dilutions of the sera are added to the wells. Positive

reactions appear as flat sediment, and negative reactions appear as a smooth dot in the

center of the well. The indirect hemagglutination titer is the reciprocal of the highest

dilution of sera found to give a positive reaction.

Nielsen (1985) reported that the IHA test cross-reacted with serotypes 6 and 8.

However, he also stated that the IHA test and the gel diffusion test are the best methods

for identification of unknown serotypes. The IHA test is also able to distinguish between

the often cross-reactive serotypes 4 and 7 while a variety of other tests are unable to do

so (Mittal, 1990). The IHA test may be limited somewhat by the capacity of specific

antigens to adsorb onto the surface of SRBC’s.

Page 27: At

19

Enzyme -Linked Immunosorbent Assay (ELISA)

The use of an enzyme-linked immunosorbent assay (ELISA) for the serotyping of

A. pleuropneumoniae was first proposed by Nicolet et al. (1981) as an alternative to the

complement fixation test. In an attempt to standardize the procedure Trottier et al.

(1992) developed a standard protocol for identifying serotype 5. Recent attempts to

further optimize this ELISA method have focused on purifying a serotype-specific

antigen. Capsular polysaccharides (Inzana and Mathison, 1987b; Bosse et al., 1990),

lipopolysaccharides (Fenwick and Osburn, 1986), and outer membrane proteins (Rapp

and Ross, 1986) have all been associated as serotype 5 specific antigens. Long-chain

lipopolysaccharides (LC-LPS) were found to be a suitable antigen for the serodiagnosis of

serotype 5 by Gottschalk et al. (1994b). The standardized ELISA procedure was later

adapted to serotype 1. However, highly purified capsular polysaccharides of serotype 1

were cross-reactive with anti-sera to serotypes 9 and 11 (Gottschalk et al., 1994a).

Although LC-LPS of serotype 1 are also cross-reactive to serotypes 9 and 11, Radacovici

et al. (1994) have suggested its use because LC-LPS are easier to purify in large quantities

than capsular polysaccharide.

Performing the ELISA requires selecting and purifying an antigen. The crude

extract of A. pleuropneumoniae can be prepared by boiling A. pleuropneumoniae cells and

collecting the supernatant (Trottier, 1992). Other purification procedures such as phenol

extraction are also often required (Inzana et al., 1992; Radacovici et al., 1994). The

Page 28: At

20

antigen must then be titrated for optimal dilution and adsorbed onto the bottom surface of

the ELISA plates. After washing the plates, the test sera is then added. After an

incubation period, anti-swine IgG conjugated with horse-radish peroxidase is added to the

reactions. Substrate solution is added and the plates are shaken. Positive sera will have

antibodies binding to the immobilized antigen and the anti-swine IgG antibody will then

bind to the antigen-antibody complex. The horse-radish peroxidase creates a color

reaction in the presence of the added substrate and can be measured by the use of a plate

spectrophotometer. The type of plate is also an important factor in performing the

ELISA as the materials can vary considerably. Although most antigens bind to plates

through hydrophilic interaction, capsules are hydrophobic and may bind poorly to most

plates. This will reduce both sensitivity and specificity (Inzana, personal comm.).

An alternative form of the ELISA is the inhibition or blocking ELISA (Nielsen et

al., 1993; Stenbaek and Schirmer, 1994). This method employs a competitive reaction of

antibody detection. Monoclonal antibodies or polyclonal antibodies that have been

absorbed with cross-reacting serotypes are used to bind to a serotype-specific antigen

adsorbed to the plate. These antibodies are conjugated to an enzymatic indicator such as

horse-radish peroxidase. However, the experimental sera is first added to the ELISA and

if it contains antibodies specific to the antigen, it will block the prepared monoclonal or

polyclonal antibodies from binding. Positive sera will result in a negative reaction so that

the absorbance is inversely related to the amount of positive sera.

Page 29: At

21

This type of detection has been used successfully for serotype 8 (Nielsen et al.,

1993) and for serotype 2 (Stenbaek and Schirmer, 1994). Stenbaek et al. (1994) used a

monoclonal blocking antibody, and although the inhibition ELISA for serotype 2 did not

cross-react with other serotypes, it was not able to detect all strains of serotype 2. This

is a common problem that occurs when using monoclonal antibodies. However, in regards

to sensitivity, ELISA’s are more sensitive than most immunologically based assays

because the enzyme associated with each antigen-antibody interaction in an ELISA can

react with a number of substrate molecules.

Agglutination and Coagglutination Tests

Agglutination tests are simple and quick methods for identifying and serotyping A.

pleuropneumoniae isolates. Methods of performing an agglutination test have included

tube agglutination, 2-mercaptoethanol (2-ME) tube agglutination, and rapid slide

agglutination. These tests are very similar with the latter being performed on a slide as

opposed to in a tube. Antiserum for the agglutination test is collected from a rabbit that

has been innoculated with A. pleuropneumoniae antigen. The antiserum is then serially

diluted with saline or with 0.1 M 2-ME, in the case of the 2-mercaptoethanol tube

agglutination test. The antigen from the test isolates is prepared by suspending the

bacteria in formol saline solution and then boiled. An equal volume of test antigen is

thoroughly mixed with the dilutions of antiserum. The reaction is determined for

Page 30: At

22

agglutination by visual inspection. Tube agglutination tests require an 18 hour incubation,

but a positive reaction on the slide agglutination test can usually be seen within a couple

of minutes (Mittal et al., 1984)

The coagglutination test is quite similar to the rapid slide agglutination test. A

protein-A producing Staphylococcus aureus strain is mixed with serotype-specific

antiserum. The Staphylococcus mixture is then added to an equal volume of bacterial

suspension on a glass slide. Protein A binds to the Fc portion of IgG and allows the

complex to become more highly visible during agglutination. A positive reaction usually

occurs within a few seconds (Mittal, 1983b).

Although these tests share similar principles, there are distinct differences in the

results of performing these tests. A comparison of these tests found that the

coagglutination test and the 2-mercaptoethanol tube agglutination test were both more

specific and sensitive than the rapid slide agglutination and tube agglutination tests. The

coagglutination test also has the added advantage of being able to type autoagglutinating

strains (Mittal et al. 1987). However, all of these tests were found to be unable to

differentiate serotype 3 isolates from serotypes 6 and 8. Still, Rapp et al. (1985a) found

that the rapid slide agglutination test was more specific and sensitive than the indirect

fluorescent antibody test. Although the rapid slide agglutination test and the

coagglutination test do not have the same sensitivity as ELISA’s, they are both quick and

Page 31: At

23

simple and have become routine methods for serotyping A. pleuropneumoniae in many

laboratories.

Latex Agglutination

The latex agglutination test was introduced as another simple and quick method

for detection and typing of A. pleuropneumoniae. Serotyping using this method has been

reported for serotypes 1, 2, 3, 5, and 9 (Mitui et al. 1981, Giese et al. 1993, Inzana 1995).

Mitui et al.(1981) used the latex agglutination test to detect A. pleuropneumoniae based

on a method by Suzuki et al (1977). However, use of this method resulted in cross-

reactivity of some serotypes.

A different procedure for the latex agglutination test was proposed by Inzana

(1995) to eliminate cross-reaction. The test uses specific IgG antibody to detect and

serotype isolated A. pleuropneumoniae bacteria or the antigen in tissue samples.

Antiserum to each serotype is collected from a rabbit that have been immunized with

encapsulated A. pleuropneumoniae. The serum is then adsorbed with a non-encapsulated

homologous mutant of the A. pleuropneumoniae strain. This procedure removes all

antibodies to exposed A. pleuropneumoniae antigens that are not capsule specific from the

serum. The specific IgG antibodies are coupled to latex particles and placed on an

agglutination slide. The test sample is mixed on the slide and the reaction is observed for

agglutination. Reaction time is approximately 30 seconds. The occurrence of agglutination

Page 32: At

24

is a positive reaction and indicates the presence of the specific serotype of A.

pleuropneumoniae (Inzana, 1995).

The latex agglutination test described here has been modified from other latex

agglutination tests to allow it to be used for rapid field testing while maintaining both

sensitivity and specificity. This test is still a relatively new procedure and has not yet

been established as an effective diagnostic tool. However, it remains a potentially useful

method for rapid testing without the need for extensive preparation.

Indirect Fluorescent Antibody Test

In 1981, Rosendal et al. reported the use of the indirect fluorescent antibody test

as a method of serotyping and detecting A. pleuropneumoniae. This method can be

performed quickly and relatively easily. Rabbits are inoculated with antigen from each of

the serotypes, and when their sera contains antibody to the homologous antigen, their

antiserum is collected for use. Smears of bacteria or tissue samples are made on glass

microscope slides. One drop of antiserum is placed on a slide and incubated for 45

minutes. One drop of goat-anti-rabbit serum with a fluorescein label is then added to the

slide. After the slide is incubated and washed, it is observed under a fluorescence

microscope at 1000 times magnification. A strong fluorescence on the test slide, when

compared to the background of a negative control, indicates a positive reaction for that

serotype (Rosendal et al., 1981).

Page 33: At

25

The indirect fluorescent antibody test is not time consuming; it takes about 3-4

hours to complete (Rosendal et al., 1981). However, cross- reactivity has been found

from heterologous anti-sera to serotype 6 as well as cross reactions between serotype 4

and 7 and serotypes 4 and 5. In addition the indirect fluorescent antibody test has been

found to be insensitive to serotype 1 (Rapp et al. 1985a). Overall, the indirect

fluorescent antibody test has relatively low sensitivity and specificity.

Immunodiffusion Test

Immunodiffusion is based on the principle of antibodies and antigens diffusing

through agar and visibly precipitating when antibody-antigen complexes are formed.

Gunnarsson (1979) used this method to study the antigenic properties of serotype

specificity. Immunodiffusion has the advantage of forming a visible precipitating band for

the antigen-antibody complexes formed. This makes some of the specific antigens for a

particular strain visible as well as giving the serotype of the strain. The immunodiffusion

test also has the flexibility of detecting either antigen or antibody to A. pleuropneumoniae.

Antisera is prepared from rabbits by immunizing each one with cell-antigen from

one of the A. pleuropneumoniae serotypes. Antisera to all 12 serotypes are then

collected. The test antigen is prepared by phenol-water extraction with the test antigen

remaining in the aqueous phase. A 1% agarose Veronal buffer gel is melted and poured

onto level microscope slides. A well of 2-mm for the test antigen is drilled into the

Page 34: At

26

agarose. One 2 mm well for each of the specific anti-sera is then drilled equidistant

around the center well. The center well is filled with the test antigen, and each of the

surrounding wells is filled with one serotype-specific anti-serum. The reactions are

incubated at room temperature in a moist chamber for several days and then read. If the

test sample is A. pleuropneumoniae, a precipitating band should appear near the center

well between each of the surrounding wells, forming a ring around the antigen well. An

additional precipitation band or bands will appear near the anti-serum well containing the

homologous serotype of the test antigen (Gunnarsson, 1979).

This test, although simple, requires a long incubation period. Precipitation bands

around the wells can be difficult to interpret and may not give clear results. For this

reason Gunnarsson et al. (1978) recommended the use of the tube agglutination test over

the immunodiffusion test for serotyping A. pleuropneumoniae. However, because of the

distinct bands created by each specific antigen-antibody complex, the immunodiffusion

test has been a useful method for studying the additional antigens that are present on a

serotype of A. pleuropneumoniae.

Ring Precipitation

Although the ring precipitation test is not commonly used today, it is another

quick and simple test developed for the serotype-specific detection of A.

pleuropneumoniae antigen. The following method was described by Mittal et al. (1982).

Page 35: At

27

Whole A. pleuropneumoniae cells from each of the serotypes were used to innoculate

rabbits. The antiserum to each serotype is collected and is ready for use in the ring

precipitation test. Test antigen was prepared by removing the bacteria from plates and

suspending them in saline. The cells were then autoclaved and centrifuged. The

supernatant is then collected and ready to use. Serum is aspirated into a pasteur pipette,

and the tip sealed. The prepared test antigen is layered over the serum, and if the antigen

binds to the antibodies in the serum, the complex will precipitate out. Within one minute

a positive reaction will result in a sharp ring of visible precipitation.

Mittal et al. (1982) reported that the ring precipitation test gave consistent and

reproducible results. This test could also type autoagglutinating strains of A.

pleuropneumoniae which could not be typed by agglutination tests. Mittal et al. (1982)

recommended that the ring precipitation test replace the tube agglutination test for routine

laboratory serotyping.

Biochemical Typing

As a possible alternative to traditional serotyping, Sirois and Higgins (1991) have

proposed a method of biochemical typing. By analyzing phenotypic variations,

biochemical tests can type A. pleuropneumoniae strains into selected groups. Sirois and

Higgins (1991) performed 38 biochemical and physiological tests on 67 strains of

serotypes 1 and 5. Of these 38 tests, 17 reactions were uniformly negative, 17 reactions

Page 36: At

28

were uniformly positive, and 1 test was discarded for variable results within a strain.

Three tests were used for biochemical typing and consisted of acid production of glycerol,

lactose, and raffinose.

A. pleuropneumoniae strains were divided into six phenotypic groups by using the

three tests. Forty-three percent of serotype 1 strains were classified into group 1, and

80% of the serotype 5 strains were classified into group 4. Sirois and Higgins (1991)

have suggested that the phenotypic classifications can be used with traditional serotyping

to help identify the epidemiology associated with A. pleuropneumoniae.

Polymerase Chain Reaction (PCR)

In recent years the polymerase chain reaction (PCR) has become a new tool for

the detection and serotyping of A. pleuropneumoniae (see next section on polymerase

chain reaction). Several methods have now been developed to detect or serotype A.

pleuropneumoniae by using the PCR (Sirois et al., 1991; Hennessy, 1993). The use of

PCR allows for the possible detection and typing of A. pleuropneumoniae within several

hours with great sensitivity. However, because very little of the A. pleuropneumoniae

genome has been sequenced, there have been no reports of using serotype-specific

primers on A. pleuropneumoniae. Described below are two methods that use the

polymerase chain reaction for the detection or serotyping of A. pleuropneumoniae.

Page 37: At

29

Hennessy et al. (1993) describe the use of an arbitrarily primed polymerase chain

reaction (AP-PCR) for A. pleuropneumoniae serotyping. By using arbitrarily selected

primers it is possible to amplify regions of genomic DNA without knowing the sequence.

Each of the serotypes should have DNA fragments of different sizes amplified based on

their unique genomic sequences. This results in a specific banding pattern for each

serotype when observed on an agarose gel and is the equivalent of a genomic fingerprint.

By recording the distinctive banding pattern for each serotype it is possible to identify

the A. pleuropneumoniae serotype.

The use of AP-PCR has the advantage of typing bacteria without having the

knowledge of the unique sequences for that serotype. However, AP-PCR has the

disadvantage of requiring a pure culture free from any contaminating DNA, including

plasmid. It is also often difficult and confusing to interpret serotypes that have complex

or similar banding patterns to other serotypes.

Sirois et al. (1991) developed species-specific primers for the detection of A.

pleuropneumoniae. By cloning and sequencing a region responsible for hemolysis from A.

pleuropneumoniae, they were able to target an area that was species specific. The

primers were shown not to amplify DNA from any other bacteria with the exception of

Actinobacillus lignieresii. However, Gram et al. (1996) reported finding non-specific

reactions with this test from bacteria isolated from tonsil cultures. This suggests that

Page 38: At

30

more specific primers are needed in order to detect A. pleuropneumoniae from mixed

bacterial cultures.

Summary

The detection of A. pleuropneumoniae is important for the treatment and control

of the disease. The serotyping of A. pleuropneumoniae is also important for these

reasons: it is essential for epidemiological studies on transmission of the disease; effective

vaccination may be dependent on serotype-specific immunization; serotyping is needed

in serodiagnostic studies (Rosendal et al. 1981).

The effectiveness of A. pleuropneumoniae serotyping is dependent on the type of

test selected and the preparation of antigen or antibody. Although, there is a wide range

of tests to choose from, no single test has proven to be completely satisfactory. The

rapid slide agglutination and coagglutination tests are commonly used by laboratories for

routine detection and serotyping of bacterial samples. The complement fixation test and

the ELISA have been commonly used to detect antibody to A. pleuropneumoniae

serotypes.

The recent advent of the use of PCR as a diagnostic tool has offered an alternative

to the traditional immunologically based assays. Currently, there are methods being

developed to employ PCR as a way to detect and serotype A. pleuropneumoniae. Once

Page 39: At

31

unique DNA sequences for each serotype are found, PCR methods are likely to be the

diagnostic tool of choice because of their sensitivity, specificity, and ease of use.

Page 40: At

32

The Polymerase Chain Reaction and Its Uses in Microbial Detection

History

What is perhaps most intriguing about the origin of the polymerase chain reaction

(PCR) is that all of the basic science and reagents required for the reaction had been

available for at least 10 years prior to the development of the technique. In 1955, Arthur

Kornberg characterized the enzyme DNA polymerase, the cellular enzyme that is

responsible for DNA repair and replication. This would become the key discovery that

would later lead to the invention of both the polymerase chain reaction and the dideoxy

sequencing method.

By the late 1970's radioactive oligonucleotide probes were becoming commercially

available to detect specific fragments of DNA. Oligonucleotide probes are small

sequences of DNA that are manufactured to bind to a specific complementary sequence

of DNA. Because the genomic DNA sequences from living organisms are very large, it is

difficult to study and manipulate a small specific region of DNA. By binding the

oligonucleotide probes to genomic DNA that has been broken into smaller pieces, it is

possible to isolate a much smaller piece of DNA that contains the sequence of interest.

Page 41: At

33

Oligonucleotide probes, however, can also serve an alternative function. By

adding DNA polymerase and nucleotide triphosphates, an entire complementary strand

of DNA can be produced from the oligonucleotide to the end of the DNA fragment. In

1980, Frederick Sanger used this principle to develop a method of DNA sequencing called

enzymatic or dideoxy sequencing. This method allows one to determine the individual

bases that make up a DNA sequence. The discovery eventually triggered the invention of

the polymerase chain reaction.

In 1983, Kary Mullis, a scientist for Cetus Corporation, was trying to develop a

variation of the dideoxy sequencing method while driving to his cabin on a Friday night.

While analyzing the problems associated with using two oligonucleotides simultaneously,

he stumbled across the realization that the sequences would be extended beyond the

oligonucleotide on the opposite end. Repeating the step multiple times would result in

the amplification of DNA. This was the birth of the polymerase chain reaction. The first

publications of this procedure appeared in 1985 (Saiki et al.) and was patented by Perkin-

Elmer Cetus. The widespread use and importance of this procedure became quickly

evident. A Medline search of the polymerase chain reaction found 141 references cited in

1988, 697 references in 1989, and 11,541 references by 1995.

Page 42: At

34

Principles of the Polymerase Chain Reaction

The polymerase chain reaction is an enzymatic reaction that utilizes DNA

polymerase and oligonucleotide primers to amplify fragments of DNA. The discovery of

Taq polymerase, a thermostable polymerase isolated from Thermus aquaticus, was a

main step forward in making PCR a very simple automated process. Prior to the use of

Taq polymerase, fresh DNA polymerase would have to be added after each denaturation

step.

The polymerase chain reaction is based on a simple theoretical process requiring a

three-step cycling process. The three steps involved in PCR are: 1) denaturation; 2)

primer annealing; 3) extension. Double stranded DNA is denatured by heating the

reaction to approximately 95o C for one minute. This separate the DNA from its

complementary strand and gives the primers a target to bind. By reducing the

temperature to between 30-70o C depending on the G+C content, the oligonucleotide

primers can anneal to their target sequence. The temperature is then raised to 72o C, the

optimum temperature for Taq polymerase activity. The Taq polymerase then extends

the primers in the 3' direction. One cycle of these three steps takes approximately 5-10

minutes to complete, and a typical PCR run will need about 30-35 cycles (see Fig. 1).

Oligonucleotide primers are designed to bind at positions flanking the DNA region

of interest. After the DNA strands are separated, the forward primer binds to the sense

strand on one side and the reverse primer binds to the antisense strand on the opposite

Page 43: At

35

end of the DNA region. DNA sequences are elongated in the 5' to 3' direction so that

both primers are extended past each other by the end of the first cycle. When the next

cycle starts the newly replicated DNA separates from the original DNA, and the primer

extension begins again. After the first few cycles the mass of the major amplified DNA

product is equal to the sum of the two primers plus the DNA region between them. The

results of the PCR reaction can easily be verified by determining the size of the DNA

product using electrophoresis.

Page 44: At

36

Figure 1.1. Schematic diagram of the three steps of PCR: denaturation, annealing, and

elongation. After completing these steps, the cycle is repeated.

5’

3’

3’

5’

Denaturation 94o C

Step 1

Step 2

Annealing 52o C

5’

3’

3’

5’

5’

3’

3’

5’

Page 45: At

37

In theory, PCR allows the number of target DNA sequences to double in quantity

after each cycle. The actual amount of amplification can be expressed as (1+X)n where n

is the number of cycles and X is the average efficiency of each cycle (Gingeras et al.,

1990). The efficiency of the reaction has been measured by Mullis and Faloona (1987) to

be between 62-85%. Efficiency of the reactions will decrease in the later cycles because

the increased amount of product will also increase the probability that the DNA strands

will anneal back together again.

Besides the oligonucleotide primers, the template DNA, and the Taq polymerase,

the PCR buffer also contains KCl, Tris-HCl, deoxynucleotide triphosphates, and MgCl2.

The MgCl2 plays a critical role in PCR amplification. Both the specificity and yield of

the products are very sensitive to varying concentrations of MgCl2 (Oste, 1989). The

precise concentration of MgCl2 needed in the PCR buffer must be determined for each

individual reaction. A second parameter that must be individually determined is the

annealing temperature of the primers. By varying the annealing temperature, it is possible

to control the specificity of the primers. The use of annealing temperatures lower than

the optimum annealing temperatures may result in non-specific binding and non-specific

products.

The increasing popularity of PCR has resulted in automation of PCR.

Temperatures are now controlled by programmable thermal cyclers. The thermal cyclers

provide both convenience and increased reliability when running PCR. Commercial kits

Page 46: At

38

are available for optimizing reactions, pre-made buffers, and specialized situations.

However, because of the amplification power of PCR, accidental contamination is always

a concern. Negative controls should always be used, and extra care should be placed in

setting up a pre-PCR work area and a post-PCR work area to prevent cross-

contamination from amplified products via aerosols (Bej et al., 1991). In addition, some

“carry-over” contamination from micropipettes can be avoided by designating one set for

pre-PCR work and one set for post-PCR work.

Applications of PCR

Since its inception, the amplification of DNA by PCR has become a valuable tool

for molecular biology. It has set off a virtual explosion of application ideas. Today, there

are a number of ingenious variations of the original PCR method that scientists use to

develop new applications. Some of the most common PCR variants include the use of

degenerate primers, nested primers, multiplex PCR, inverse PCR, single primer, booster

PCR, and anchored PCR (Cooper and Isola, 1990; Haqqi et al., 1988; Bej et al., 1990;

Ochman et al., 1990; Kalman et al.,1990; Ruano et al., 1989; Loh et al., 1989). PCR has

been used extensively in many fields including, but not limited to, diagnostics, sequencing,

molecular evolution, forensics, and genetic engineering.

Cloning by PCR is a simpler and faster method than standard cloning techniques.

PCR allows restriction sites to be added to DNA fragments and the amplified DNA can

Page 47: At

39

then be cloned directly into a cloning vector. This method bypasses the use of

constructing gene libraries, screening recombinant clones, restriction mapping, and

subcloning (Scharf et al., 1986). PCR methods for constructing cDNA libraries have been

described, as well as methods for screening cDNA recombinants with PCR (Ausch and

Reth, 1990). DNA sequences cloned by PCR can be sequenced by traditional methods.

However because of the error rate of Taq polymerase when incorporating nucleotides,

direct sequencing by PCR is often preferred (Stoflet et al., 1988). This method utilizes a

phage promoter in the primers. The DNA is then transcribed by using a phage

polymerase, and the transcript is sequenced by dideoxy sequencing using reverse

transcriptase. PCR has been used for various other methods of genetic engineering

including site-directed mutagenesis and gene fusion techniques (Hemsley et al, 1989; Yon

and Fied, 1989).

DNA sequence analysis is one of the most informative methods for studying

evolution and the phylogenetic relationships of different species. In the past it was often

difficult to produce enough DNA for analysis, but DNA amplification by PCR has

opened the way for analyzing sequence information by increasing the quantity of DNA

from a limited source. The use of PCR has been applied to mitochondria to study the

origins of the human species (Erlich et al., 1991). It has also allowed DNA analysis of

ancient samples from organisms that have been extinct for thousands of years (Mullis,

1990).

Page 48: At

40

Various PCR methods have also played a key role in the Human Genome Project.

PCR techniques have been developed for physical and genetic mapping of human

chromosomes as well as automated sequencing methods (Erlich et al., 1991). PCR has

also been employed to detect genetic diseases. Amplified regions of target DNA can be

analyzed for mutations or multialleles by sequencing, hybridization, restriction analysis,

or enzymatic cleavage. Some of the genetic diseases that have been characterized and

diagnosed by PCR have included sickle cell anemia, hemophilia A, alpha-thalassemia,

beta-thalassemia, phenylketonuria, and Duchenne’s muscular dystrophy (Saiki et al.,

1985; Kogan et al., 1987; Chehab et al., 1987; Cai et al., 1988; DiLella et al., 1988;

Chamberlain et al., 1988). PCR also allows the identification of an individual from a

single hair or sperm cell (Higuchi et al., 1988). It can also be used in identifying either the

mother or father of a child (Orrego et al., 1988).

PCR is becoming extremely important in detecting viruses that cause infectious

diseases. Many viruses can remain dormant for many years in the human body and can

escape detection by serological assays that do not have the sensitivity of PCR. PCR has

been successfully used to study and detect such viruses as HTLV-I and II, HIV, hepatitis

B, human papillomaviruses, cytomegalovirus, and Epstein-Barr virus (Erlich et al., 1989;

Kwok et al., 1989; Mack and Sninsky, 1988; Ting and Manos, 1990; Demmler et al.,

1988; Saito et al, 1989).

Page 49: At

41

PCR and Bacteria

Bacterial identification by PCR has become an increasingly popular method of

microbial diagnosis. The use of PCR has several distinct advantages over traditional

diagnostic techniques. The genetic specificity provided by species-specific primers gives

a much greater level of specificity than is obtained by immunogenic antigens. This

specificity is critical in avoiding cross-reactivity that can occur from serological methods

of bacterial identification. PCR methods also have the advantage of being effective with

both live and dead organisms. This allows the test to be performed with frozen or

preserved samples that have been stored for indefinite periods of time. In addition, using

dead organisms provides greater safety when testing for dangerous pathogens.

PCR can also provide results more quickly in some cases. Organisms that require

long incubation periods for growth, six weeks in the case of Mycobacterium tuberculosis,

can be identified by PCR without delay (DeWit et al., 1990). The sensitivity of the PCR

assay is also impressive. It is not uncommon for scientists to report the ability to detect

less than 10 organisms in a sample by PCR. This sensitivity is a result of the tremendous

amplification power of PCR and gives innumerable uses in the field of diagnostics and

molecular biology.

There are several options to determine a suitable target for diagnostic purposes.

The arbitrarily primed PCR is a method that takes advantage of PCR without prior

knowledge of specific sequences (Welsh and McClelland, 1990). By selecting arbitrary

Page 50: At

42

primers, it is possible to get a DNA profile of the bacterial genome. This profile should

appear consistently when using the same primers under the same condition. This method

can be effective for both species identification and serotyping. Unfortunately, this

method of identification can be unreliable. The assay is highly subject to contamination

and can only use pure samples of bacterial colonies or DNA. Any type of DNA

contamination, including plasmid, can have a deleterious effect on the test. In addition, all

conditions when performing the test must be highly standardized because even a slight

change in conditions may effect the results.

Another target for bacterial identification by PCR is based on the 16S rRNA

primer selection (Ward et al., 1992). The 16S rRNA contains regions of highly conserved

sequences that are found in all bacteria. Between these highly conserved sequences are

regions that posses moderately to highly variable sequences of DNA. Because primers to

the conserved sequences will work with virtually any bacteria, the variable regions of a

great number of bacteria have already been sequenced. This information was originally

used as a phylogenetic tool to classify the phylum, genus, and species of an organism

(Weisburg et al., 1991). Today, these variable sequences are being used to select PCR

primer targets for the identification of bacterial species.

A third type of primer target for bacterial identification by PCR is a species-

specific gene sequence. However, finding a unique gene sequence that has already been

characterized for a particular bacterium can be difficult. These sequences are often

Page 51: At

43

discovered by determining unique antigens that are presented by the bacterium. PCR

using specific primers has successfully identified organisms such as Bordetella pertussis,

enterotoxigenic Escherichia coli, and Shigella by targeting the genes that encode for the

unique toxins produced by these organisms (Houard et al., 1989; Frankel et al., 1989).

Summary

The use of PCR has become increasingly popular in microbial diagnostics as well

as other areas of molecular biology. The tremendous amplification power of PCR has

resulted in countless uses and applications of the procedures as well as multiple

variations of the technique. Although PCR is not commonly used for routine diagnosis

today, it is becoming increasingly simple and inexpensive to run. The key reagents for a

PCR reaction include: primers, Taq polymerase, MgCl2, and the PCR buffer. Special

attention must also be given to avoiding contamination. PCR has been used extensively in

such areas as diagnostics, sequencing, molecular evolution, forensics, and genetic

engineering.

Identifying bacteria by PCR has several advantages over traditional methods of

identification. PCR can identify both live and dead organisms with little preparation; it is

often far more sensitive than other forms of identification; and the use of specific primers

avoids the possibility of cross-reactivity that is often associated with immunological

Page 52: At

44

assays. With the simplification of performing PCR by the number of commercial kits

already available, PCR diagnostics appears to be a wave of the future.

Page 53: At

45

CHAPTER 2

Detection and Identification of Actinobacillus pleuropneumoniae

Serotype 5 by Multiplex PCR

ABSTRACT

A multiplex polymerase chain reaction (PCR) assay was developed to detect

Actinobacillus pleuropneumoniae and to distinguish serotype 5 strains. Primers specific

to the conserved export and serotype-specific biosynthesis regions of the capsular

polysaccharide of A. pleuropneumoniae serotype 5 were used to amplify 0.7 kb and 1.1

kb DNA fragments, respectively. The 0.7 kb fragment was not amplified by any

heterologous species from swine tested, but was amplified by all A. pleuropneumoniae

strains tested with the exception of serotype 4. In contrast, amplification of the 1.1 kb

fragment was specific for serotype 5 strains. The assay was able to detect A.

pleuropneumoniae and identify serotype 5 strains from lung tissue samples taken from

infected swine. The sensitivity of the assay was less than 102 colony forming units. This

PCR assay enabled us to detect A. pleuropneumoniae rapidly and to distinguish serotype

5 strains from other serotypes. The use of primers specific to the biosynthesis regions of

other A. pleuropneumoniae serotypes would expand the diagnostic and epidemiologic

capabilities of this assay.

Page 54: At

46

INTRODUCTION

Actinobacillus pleuropneumoniae is the etiological agent of swine

pleuropneumonia, a highly contagious and sometimes fatal disease. Current attempts to

control the disease have been unsuccessful and have resulted in large economic losses to

the swine industry (Willson et al., 1987). Response to an infection can vary from sudden

death, to chronic coughing and weight loss, to subclinical symptoms (Nicolet, 1992).

Because the disease can be transmitted quickly throughout the herd, early detection of the

bacteria is important for control and treatment of the disease. Mortality in acute

outbreaks have been reported to be as high as 40% in some cases (Fedorka-Cray et al.,

1993). Chronically infected swine have reduced weight gain, higher medication costs, and

reduced market value (MacInnes and Rosendal, 1988). In addition, animals with

subclinical infections are prevalent in some areas. These animals have decreased growth

gains and increase the risk of an acute outbreak in the herd (Brandreth and Smith, 1985).

A. pleuropneumoniae is a gram-negative, encapsulated, nonmotile, coccobacillus

that can be either NAD-dependent (biovar 1) or NAD-independent (biovar 2) (Shope,

1964; Niven and Levesque, 1988). Serotype specificity is determined by the unique

capsular polysaccharide found on the surface of A. pleuropneumoniae (Inzana and

Mathison, 1987a). A. pleuropneumoniae has been classified into 12 distinct serotypes

that vary in virulence. Specific serotypes predominate in different geographic regions

(Rosendal et al., 1985; Rogers et al., 1990; Sebunya and Saunders, 1983). In the United

Page 55: At

47

States, serotype 5 is one of the most commonly identified A. pleuropneumoniae isolates

(Sebunya and Saunders, 1983). Serotyping is important in understanding how the disease

is spread, for treatment, and for prevention. Several methods of serotyping A.

pleuropneumoniae have been developed based on serologic assays. However, these

methods may have problems with specificity. Cross-reactivity has been reported

between serotypes 1, 9, and 12, between serotypes 3 and 6, and between serotypes 5 and

7 (Perry et al., 1990; Fedorka-Cray et al., 1993) . These cross-reactions are most likely

due to shared species-specific antigens such as lipopolysaccharides or type-specific

shared capsular polysaccharides (Nicolet, 1988).

The polymerase chain reaction (PCR) has become a powerful and increasingly

popular tool in microbial identification. Its ability to detect genetic sequences from

minute quantities of DNA gives it several potential advantages over the traditional

serologic forms of detection. First, the genetic basis of the assay avoids problems with

cross reactivity between antigen-antibody reactions. This may also provide a potential

method for serotyping strains that have been previously characterized as untypeable.

Second, the tremendous amplification power of PCR makes it extremely sensitive. Third,

PCR detection can be performed directly on samples without waiting for culture of the

bacteria.

Recently, a portion of the capsular region of A. pleuropneumonia serotype 5 was

cloned and sequenced by Ward and Inzana (1995). The organization of capsule genes of

Page 56: At

48

A. pleuropneumoniae shows striking similarity to that of other group II capsule genes.

Species in this group include encapsulated Haemophilus influenzae, Escherichia coli, and

Neisseria meningitidis (Frosch et al., 1991). Each of these species contains a central

serotype-specific DNA region responsible for encoding capsular polysaccharide

biosynthesis. Flanking the biosynthesis region are DNA sequences that are necessary for

capsular polysaccharide export. The export regions of each species that produces group

II family capsules has been shown to have substantial homology among the group II

species (Frosch et al., 1991).

A Southern blot was used to identify a portion of the capsular region of A.

pleuropneumoniae serotype 5 strain J45 by using a probe of the H. influenzae type b

capsulation (capb) locus (Ward, 1995). The H. influenzae probe was able to hybridize

with the A. pleuropneumoniae capsule fragment at medium-to-low stringency, indicating

that the two species share DNA homology at this capsule locus. After cloning the

homologous A. pleuropneumoniae fragment, a sequence analysis was performed. The

sequenced portion consisted of a region of export genes (cpx) and a region of biosynthesis

genes (cps). Homology of the cpx region to other A. pleuropneumoniae serotypes was

shown by hybridization of a serotype 5 cpx probe to serotypes 1, 2, 7, and 9. The

ability of the cpx probe to hybridize under stringent conditions with other serotypes is

consistent with the conserved nature of the export genes of group II capsules.

Hybridization studies of the cps region were also repeated with serotypes 1, 2, 7, and 9,

Page 57: At

49

but the serotype 5 cps probe was unable to hybridize to any A. pleuropneumoniae

serotype other than serotype 5. As in other organisms with group II capsules, the

hybridization results indicated that the biosynthesis region was serotype specific (Ward,

1995).

Previous attempts to utilize PCR for detection and serotyping of A.

pleuropneumoniae have been reported. Hennessy et al. (1993) reported the use of an

arbitrarily primed polymerase chain reaction (AP-PCR) for serotyping A.

pleuropneumoniae. This type of PCR method has the disadvantage of requiring pure

bacterial samples for testing and is highly susceptible to contamination. Field samples

must first be plated and incubated overnight. Individual colonies are then picked the next

day and assayed by AP-PCR. This results in the loss of at least a 24 hour period of time

as well as some of the additional difficulties associated with using AP-PCR. Sirois et al.

(1991) described the use of uncharacterized primers that were found to be specific for A.

pleuropneumoniae. Although the method provided an increased level of detection, these

primers were only able to reliably detect A. pleuropneumoniae when pure culture samples

were used. Samples of mixed bacterial cultures from tonsils sometimes showed non-

specific PCR products. This makes the identification of A. pleuropneumoniae from field

samples extremely problematic.

The cloning and sequencing of the capsule region of A. pleuropneumoniae

serotype 5 in combination with PCR provides a unique method for detecting A.

Page 58: At

50

pleuropneumoniae and identifying serotype 5. Because each serotype is distinguished by

its capsule, specific primers to the capsular polysaccharide biosynthesis region should be

able to identify individual serotypes. In addition, the conserved genes in the capsular

polysaccharide export loci may provide a target that is conserved by all 12 serotypes, but

is also specific for A. pleuropneumoniae.

MATERIALS AND METHODS

Strains and cell cultures. All bacterial strains used in this study are described in

Table 2.1. A. pleuropneumoniae strain J45 serotype 5 was provided by B. Fenwick

(Kansas State University, Manhattan), strain 53 serotype 7 was provided by M. Mulks

(Michigan State University, East Lansing), serotype 9 was provided by J. Nicolet

(University of Berne, Switzerland), strain 56153 serotype 11 was provided by M. Rider

(Kansas State University) and strain 405 serotype 8, strain D13039 serotype 10, and

strain 8329 serotype 12 were provided by K. Mittal (University of Montreal, Quebec).

B. bronchipseptica strains were provided by H. Veit. All bacterial strains were grown at

37o C on brain heart infusion (BHI) agar plates (Difco Laboratories, Detroit, MI)

containing 5 µg/ml nicotinamide adenine nucleotide (NAD) or in BHI broth containing 5

µg/ml of NAD, with shaking (BHI/NAD).

Page 59: At

51

Table 2.1. Bacterial strains and their sources.

Species Serotype Strain Source

A. pleuropneumoniae Serotype 1 4074 ATCCA. pleuropneumoniae Serotype 2 27089 ATCCA. pleuropneumoniae Serotype 3 27090 ATCCA. pleuropneumoniae Serotype 4 33378 ATCCA. pleuropneumoniae Serotype 5 K17 ATCCA. pleuropneumoniae Serotype 5 J45 B. FenwickA. pleuropneumoniae Serotype 6 33590 ATCCA. pleuropneumoniae Serotype 7 53 MulksA. pleuropneumoniae Serotype 8 405 K. MittalA. pleuropneumoniae Serotype 9 J. NicoletA. pleuropneumoniae Serotype 10 D13039 K. MittalA. pleuropneumoniae Serotype 11 56153 M. RiderA. pleuropneumoniae Serotype 12 8329 K. MittalA. suis 33415 ATCCH. parasuis field isolate Rollins Diagnostic

Lab, NCS. suis field isolate Rollins Diagnostic

Lab, NCP. multocida field isolate Rollins Diagnostic

Lab, NCB. bronchiseptica PI-53 H. VeitS. choleraesuis field isolate Rollins Diagnostic

Lab, NC

Page 60: At

52

Tissue and nasal swab samples. Lung tissue samples were taken from pigs

challenged intratracheally with 5 X 107 colony forming units (CFU’s) of A.

pleuropneumoniae J45. The lung tissue samples were taken at necropsy in 1991 and

stored at -20o C (Inzana et al., 1993). Nasal swabs were obtained from pigs challenged

with 1 x 108 CFU’s of A. pleuropneumoniae J45 and stored at -20o C.(Inzana, 1995).

DNA Isolation. Purification of genomic A. pleuropneumoniae DNA was

performed by phenol/chloroform extraction as previously described (Ward and Inzana,

1997). Briefly, A. pleuropneumoniae cells were suspended in Tris-EDTA and incubated

at 37o C for 1 hour in 0.66% sodium dodecyl sulfate and 100 µg/ml RNAse. The

suspension was then incubated an additional hour at 56o C with 100 µg/ml of Proteidae K.

The DNA was then purified by repeated phenol/chloroform extractions (Amresco Inc.,

Solon, OH). Genomic DNA was precipitated from the aqueous phase by adding 0.3

volumes of 3 M sodium acetate and 2.5 volumes of 95% ethanol. The DNA precipitate

was then dried and resuspended in sterile water.

Multiplex PCR

Sample Preparation. Rapid preparation of DNA from whole bacterial cells for

PCR was performed by suspending a loopful of colonies in 100 µl of water. The

suspended cells were lysed by heating at 100o C for 10 minutes. The lysed cells were

centrifuged, and the supernatant containing the bacterial DNA was removed and frozen at

-20oC until needed. Rapid preparation of bacterial DNA from lung tissue samples was

performed in a similar manner to the bacterial preparation. The tissue was sectioned into

thin slices of approximately 2 mm in length. The sectioned tissue was then mashed and

Page 61: At

53

vortexed in 100 µl of water. The suspended tissue was boiled at 100o C for 10 minutes.

The cellular extract was then centrifuged, and the supernatant containing the DNA was

removed and frozen at -20o C until ready for use.

Primers. Oligonucleotide primers were selected by using the DNA Star Primer

Select software (Madison, WI). Primers A and B were targeted to the cps region of A.

pleuropneumoniae serotype 5; primers C and D were targeted to the cpx region of A.

pleuropneumoniae serotype 5 (Fig. 2.1). The primers were selected based on the

following parameters: primer length, product length, product location, hairpin formations,

dimer formations, and annealing temperature. The sequences for the oligonucleotide

primers are given in Table 2.2.

Page 62: At

54

Table 2.2 Primer sequences.

Name Sequence PrimerLength

ProductLength

Forward PrimerBiosynthesis Region

A 5'-TTTATCACTATCACCGTCCACACCT-3'

25 bp 1114 bp

Reverse PrimerBiosynthesis Region

B 5'-CATTCGGGTCTTGTGGCTACTAAA-3'

23 bp

Forward PrimerExport Region

C 5'-TGGCGATACCGGAAACAGAGTC-3'

22 bp 715 bp

Reverse PrimerExport Region

D 5'-GCGAAAGGCTATGGTATGGGTATGG-3'

24 bp

Page 63: At

55

1.0 2.0 3.0 4.0 5.0 6.0 7.0 8.0

cpxA cpx B cpx C cpxD cps A cps B cps C

C D A B

Figure 2.1. Map of the sequenced capsular region of A. pleuropneumoniae serotype 5and the location of the cpx and cps primers.

Page 64: At

56

PCR Conditions. PCR reactions were performed in a total volume of 50 µl and

were based on the description given by Saiki et al. (1988). Master mixes for PCR

reactions were made fresh in batches of 750 µl. Each final reaction contained 10 mM

Tris, 50 mM KCl, 2 mM MgCl2, 400 µM each dATP, dCTP, dGTP, and dTTP, and 480

µM of each primer, A, B, C, and D. Five µl of template DNA thawed at room

temperature were added to 45 µl of the master mix for each reaction. The reactions were

then overlaid with 50 µl of mineral oil to prevent evaporation.

The PCR reactions were performed in an Omnigene thermal cycler (Hybaid

Unlimited). The template DNA was denatured at 94o C for 2 minutes, and then 2.5 U of

Taq polymerase (Fisher Scientific, Atlanta, GA) was added. A total of 30 cycles of PCR

was performed with each cycle consisting of 1 minute of denaturation at 94o C, 2 minutes

of annealing at 54o C, and 2 minutes of extension at 72o C.

Detection The amplified PCR products were detected by electrophoresis. Twelve

µl of each PCR reaction was loaded into a 0.7% agarose gel containing 0.5% ethidium

bromide. The DNA was electrophoresed through the gel at 90V using a Biorad

Laboratories (Hercules, CA) electrophoresis unit. The products were visualized by

exposure to UV light.

Page 65: At

57

Sensitivity. One milliliter of BHI/NAD broth that had been innoculated overnight with

J45 was added to 10 milliliters of BHI/NAD broth in a side-arm flask. The culture was

incubated at 37o C for 2.5 hours until a Klett reading of 175 (equivalent to 1 x 109

CFU’s/ml) was reached. Ten µl of culture was added to 990 µl of sterile water and

vortexed. The mixture was then serially diluted by adding 10 µl of the mix to 90 µl of

sterile water giving dilutions of 106 to 102 colonies per milliliter. Twenty µl of the 104

dilution were plated to give approximately 200 colonies. The diluted samples were then

boiled for 10 minutes, and 5 µl of each dilution were used as DNA template for PCR

amplification.

Probes. A cpx probe was manufactured by PCR and labeled with digoxigenin-11-

UTP (DIG-dUTP) labeling (GeniusTM System, Boehringer Mannheim, Indianapolis, IN).

The same cpx primers that were used for the multiplex PCR assay were used to produce

an identical 0.7 kb fragment. The PCR reaction consisted of the following quantities of

reagents: 48.5 µl of sterile H2O, 10 µl of Taq 10X buffer B (Fisher Scientific, Atlanta,

GA), 2 µl of 10mM dATP, 2 µl of 10mM dCTP, 2 µl of 10 mM dGTP, 1.4 µl of 10mM

dTTP, 6 µl of DIG-dUTP, 15 µl of 25 mM MgCl2, 1.5 µl of 20 µM of cpx forward

primer, 1.5 µl of 20 µM of cpx reverse primer, and 10 µl of A. pleuropneumoniae

template DNA. The PCR product was verified by gel electrophoresis. A cps probe was

manufactured by random primed labeling. A PCR fragment was produced with the cps

primer and using similar conditions as the cpx probe with the exception of the Dig-dUTP.

The product was then purified through Promega (Madison, WI) spin columns and labeled

according to the procedure in the GeniusTM System manual.

Page 66: At

58

Southern Blotting. A Southern blot was performed on organisms that produced non-

specific products from the cpx and cps primers. Transfer of DNA from agarose gels to

MagnaGraph nylon membranes (Micron Separations Inc., Westboro, MA) were carried

out by the reverse blot method (ref Current Protocols in Molecular Biology). DNA was

covalently linked to the nylon membranes by ultraviolet irradiation using a UV

Stratalinker (Stratagene, La Jolla, CA). The hybridizations were performed under high

stringency at a temperature of 68o C and at 5X SSC. All other hybridization procedures

were done following the manufacturer’s recommendation. Ten µl of the cps probe were

added to 25 ml of the prehybridization solution to be used as the hybridization solution

for cps products, and 5 µl of the cpx probe was added to 25 ml of prehybridization buffer

for use as the hybridization solution for cpx products. The membranes were washed and

developed as recommended by the manufacturer.

RESULTS

Assay of serotype 5 genomic DNA

Two primers pairs were designed from the sequenced DNA of the A.

pleuropneumoniae serotype 5 capsular region. The four primers are listed in Table 2.2.

Primers A and B were designed to target the serotype-specific cps loci, while primers C

and D targeted the serotype-conserved cpx loci. PCR products that are 0.7 kb in size and

are consistent with expected product of primers C and D will be referred to as the cpx

Page 67: At

59

product; products that are 1.1 kb in size and are consistent with expected product of

primers A and B will be referred to as the cps product.

Amplification of purified A. pleuropneumoniae J45 genomic DNA resulted in a

single product when using the cps primers A and B. The product was detected as a 1.1

kb band by electrophoresis of the DNA through an ethidium-stained agarose gel.

Amplification of purified J45 genomic DNA also resulted in a single product when using

the cpx primers C and D. This product resulted in a band that was determined to be 0.7

kb in size. When both the cpx and cps sets of primers were added to a single PCR tube,

products of 1.1 kb and 0.7 kb could be detected (Fig. 2.2). Both products were consistent

with the sizes predicted from the sequenced data of those regions.

Page 68: At

60

Figure 2.2. Agarose gel electrophoresis of PCR products from A. pleuropneumoniaeserotype 5 genomic DNA. Lane 1, 1-kb DNA ladder; lane 2, cps primers A and B; lane 3,cpx primers C and D; lane 4, primers A, B, C, and D.

Page 69: At

61

Assay optimization

The multiplex PCR assay was optimized using bacterial samples of serotype 5.

The magnesium concentrations used for the assay were varied from 1 mM to 7.5 mM;

annealing temperatures used for the assay were varied from 49o C to 55o C. These

conditions did not seem to affect the amplification of the 1.1 kb cps or 0.7 kb cpx bands

as no differences in the intensity of the bands was observed when the conditions were

altered. Initially, the optimum conditions for assaying serotype 5 samples were

performed at 5 mM MgCl2 and at an annealing temperature of 54o C.

The multiplex PCR assay was then applied to whole cells of the type strains of all

12 A. pleuropneumoniae serotypes. The following parameters were adjusted to optimize

the multiplex PCR assay for all serotypes: annealing temperature, primer concentration,

Taq polymerase concentration, and MgCl2 concentration. Non-specific products were

observed from some serotypes under the conditions used for serotype 5. For serotypes

2, 3, 6, and 7, a band slightly larger than the 1.1 kb cps band was produced.

Amplification of serotype 4 DNA produced a band that was approximately 1.3 kb in

size, but did not amplify the 0.7 kb cpx product. In addition, non-specific bands of

various sizes were often amplified in the background of some serotypes. An example of

this is shown in Figure 2.3 for serotypes 1-6 at a MgCl2 concentration of 5 mM.

Serotypes 5, 9, 11, and 12 appeared to produce more intense bands than the other

serotypes. Lowering the concentration of MgCl2 resulted in progressively fainter to non-

detectable cpx bands in serotypes 1, 2, 3, 6, 7, 8, and 10, whereas the effects on serotypes

Page 70: At

62

9, 11, and 12 were less noticeable. No cpx band was detected with serotype 4 under any

conditions.

Page 71: At

63

Figure 2.3. Agarose gel electrophoresis of PCR products of amplified from whole cells ofserotypes 1-6 at a MgCl2 concentration of 5 mM. Lane 1, 1-kb DNA ladder; lanes 2through 7, the PCR products from serotypes 1-6, respectively, amplified with primers A,B, C, and D.

Page 72: At

64

Annealing temperatures were varied from 49o C to 56o C. Increasing the annealing

temperature to eliminate the non-specific products also resulted in the loss of specific

PCR products, and therefore did not help to optimize the assay.

In order to eliminate the non-specific products similar in size to the cps product

bands observed in serotypes 2, 3, 6, and 7, without eliminating the 0.7 kb cpx band, the

concentration of the cps primers A and B was systematically reduced from a starting

concentration of 480 µM to a final concentration of 25 µM. Figure 2.4 depicts the results

of reducing the cps primers to 25 µM for serotypes 1-6. Although the non-specific bands

in 2, 3, and 6 showed a marked reduction of amplification, the intensity of 1.1 kb band

from serotype 5 was also substantially reduced.

Page 73: At

65

Figure 2.4. Agarose gel electrophoresis of PCR products of bacterial samples ofserotypes 1-6 with primers A and B reduced to 25 µM and primers C and D at 480 µM.Lane 1, 1-kb DNA ladder; lanes 2 through 7 contain the PCR products from serotypes 1-6, respectively.

Page 74: At

66

The inability to eliminate the non-specific bands by reducing the cps primer concentration

suggested that the non-specific bands may have been amplified other primers. To

determine which primers were responsible for generating the non-specific bands, bacterial

samples of serotypes 2 and 4 were amplified with only a single pair of primers at a time.

Figure 2.5 and Figure 2.6 illustrate the results using the following combination of primers

for serotypes 2 and 4, respectively: A and B; C and D; A and D; or C and B. A band

similar in size to the 1.1 kb cpx product was amplilfied from serotype 2 with primers A

and B, C and D, and primers C and B. Similarly, the 1.3 kb band produced from serotype

4 was amplified from one cps primer, primer A, and one cpx primer, primer D. This

indicated that the non-specific band amplified from the DNA of serotypes 2 and 4 could

not be eliminated by reducing the cps primers because the non-specific band was

amplified from cpx primers as well as cps primers.

Page 75: At

67

Figure 2.5. Agarose gel electrophoresis of serotype 2 PCR products amplified fromvarious combinations of primer pairs. Lane 1, 1-kb DNA ladder; lanes 2-5, productsamplified from primers A and B; primers C and D; primers A and D; and primers C andB, respectively.

Page 76: At

68

Figure 2.6. Agarose gel electrophoresis of serotype 4 PCR products amplified fromvarious combinations of primer pairs. Lane 1, 1-kb DNA ladder; lanes 2-5, productsamplified from primers A and B; primers C and D; primers A and D; and primers C andB, respectively.

Page 77: At

69

The effect of the Taq polymerase concentration was examined on PCR products

in volumes of 50 µl and 100 µl. Figure 2.7 depicts serotype 2 products and the effect of

varying the Taq polymerase concentrations when the MgCl2 concentration was 2 mM. At

2.5 U of Taq polymerase in 100 µl reaction volumes, no amplified products were visible.

However, at 7.5 U of Taq polymerase in 100 µl reactions, the cpx band is visible as well

as a faint non-specific band similar in size to the 1.1 kb cps product. In the 50 µl reaction

volumes, 2.5 U of Taq polymerase resulted in the amplification of the cpx band without

the amplification of non-specific products. At 7.5 U of Taq polymerase, both the cpx

band and the non-specific band are clearly visible. This indicated the concentration of Taq

polymerase had a pronounced effect on the amplification of PCR products.

Page 78: At

70

Figure 2.7. Agarose gel electrophoresis of amplified PCR products when theconcentration of Taq polymerase is varied using serotype 2 template DNA. Lane 1, 1-kbDNA ladder; lane 2, 2.5 U Taq polymerase in 50 µl; lane 3, 5 U Taq polymerase in 50 µl;lane 4, 7.5 U Taq polymerase in 50 µl; lane 5, 2.5 U Taq polymerase in 100 µl; lane 6, 5U Taq polymerase in 100 µl; lane 7, 7.5 U Taq polymerase in 100 µl.

Page 79: At

71

Increasing the MgCl2 concentration also resulted in an increase of non-specific

products for serotypes 2 and 4. Figure 2.8 shows the effect of varying the MgCl2

concentration from 2 mM to 7.5 mM for serotype 2. Using 2 mM MgCl2 only the cpx

band was visible. However, as MgCl2 was increased from 2 mM to 5 mM, there was a

noticeable increase in non-specific banding. When the concentration of MgCl2 was greater

than 5mM,the amount of non-specific bands began to decrease. Thus, the optimum

specificity of the multiplex PCR assay with bacterial samples required 2 mM MgCl2,

while maximum sensitivity required 5 mM MgCl2.

Page 80: At

72

Figure 2.8. Agarose gel electrophoresis of DNA products from bacterial samples ofserotype 2 at varying MgCl2 concentrations. Lane 1, 1-kb DNA ladder; lane 2, 2 mMMgCl2; lane 3, 3 mM MgCl2; lane 4, 4 mM MgCl2; lane 5, 5 mM MgCl2; lane 6, 6 mMMgCl2; lane 7, 7.5 mM MgCl2.

Page 81: At

73

The sensitivity and specificity of the multiplex PCR assay was also influenced by

the sample type. In order to visualize the differences in DNA amplification for different

sample types under the same conditions, the multiplex PCR assay was run at 5 mM

MgCl2 for the following serotype 2 sample types: Purified genomic DNA; rapidly

prepared genomic DNA; boiled bacterial cells; boiled lung tissue; and boiled nasal swabs.

Figure 2.14 shows that nonspecific bands at higher molecular weights were produced from

the genomic DNA samples, while products that are more intense at the lower molecular

weights were obtained from the boiled bacterial sample. The boiled lung tissue sample

produced a very faint 0.7 kb cpx band, and the nasal swab sample did not produce any

visible bands. While specificity increased from “pure” samples to “dirty” samples, the

sensitivity was decreased.

Page 82: At

74

Figure 2.9. Agarose gel electrophoresis of PCR products of various serotype 2 sampletypes amplified with primers A-D at a MgCl2 concentration of 5 mM. Lane 1, 1-kb DNAladder, lane 2, purified genomic DNA; lane 3, rapidly prepared genomic DNA; lane 4,bacterial sample; lane 5, lung tissue sample; lane 6, Qiagen prepared nasal swab sample,lane 7, boiled nasal swab sample.

Page 83: At

75

The optimum conditions for the multiplex PCR assay using samples of bacterial

cells were determined to be: 2.5 U of Taq DNA Polymerase, 10 mM Tris HCl, 50 mM

KCl, 2 mM MgCl2, 400 µM dNTP’s, 480 µM of each primer, and 5 µl of bacterial

sample, all mixed in a final volume of 50 µl. Figure 2.10 shows serotypes 1-12 assayed at

these optimum conditions. With the exception of serotype 4, all of the other 11

serotypes produced a band of 0.7 kb in size. In addition, only serotype 5 had a PCR

product of 1.1 kb .

Page 84: At

76

Figure 2.10. Agarose gel electrophoresis of PCR products from bacterial samples ofserotypes 1-12 at a MgCl2 concentration of 2 mM. Lane 1, 1-kb DNA ladder; lanes 2through 14, the PCR products from serotypes 1-12, respectively, amplified with primersA, B, C, and D; lane 15, 1-kb DNA ladder.

Page 85: At

77

Assay sensitivity

The sensitivity of the multiplex PCR assay was determined for strain J45 whole

bacterial cells. The cells were grown in broth to mid-log phase, and harvested at this time

to minimize the number of dead bacteria present in the sample. Both the 0.7 kb cpx and

1.1 kb cps PCR products could be visualized by gel electrophoresis for at least 1 x 102

CFU’s (Fig. 2.11). The use of reagents such as SDS or lysozyme prior to the crude

preparation of cells had no substantial effect. There was no noticeable increase or

decrease in sensitivity or quantity of amplified product when these reagents were used.

Centrifugation of the crude extract after the cells were boiled was important to avoid

decreased sensitivity for PCR preparation. In addition, detection of PCR products was

diminished from cells that had been grown in BHI broth unless they were washed prior to

preparation. Other methods used to improve the specificity of the primers, such as

varying the annealing temperature, were found to be ineffective.

Page 86: At

78

Figure 2.11. Sensitivity of the PCR assay for serotype 5 samples of bacterial cells. Lane1, 1 x 106 cells; lane 2, 1 x 105 cells; lane 3, 1 x 104 cells; lane 4, 1 x 103 cells; lane 5, 1 x102 cells; lane 6, 1 x 10 cells; lane 7, 1-kb DNA ladder.

Page 87: At

79

Assay specificity

PCR with the cps and cpx primers was also performed on six swine respiratory

pathogens (Fig. 2.12). No amplified products were made from three species by the

multiplex PCR assay: A. suis, B. bronchiseptica, or S. choleraesuis. However, PCR

products of various sizes were made from P. multocida, H. parasuis , and S. suis. A band

of approximately 1.5 kb was made from P. multocida. Two sets of double bands were

made from H. parasuis . The first set of bands appeared to have one band slightly larger

than the 0.7 kb cpx band and one band slightly smaller; the second set of bands were of

undetermined smaller molecular size. A band of similar size to the 0.7 kb cpx band was

made from the S. suis genome.

Page 88: At

80

Figure. 2.12. Agarose gel electrophoresis of the PCR products of bacterial samples ofrespiratory swine pathogens amplified with primers A-D. Lane 1, A. pleuropneumoniaeserotype 5; lane 2, A. suis; lane 3, B. bronchiseptica; lane 4, H. parasuis; lane 5, P.multocida; lane 6, S. choleraesuis; lane 7, S. suis; lane 8, 1-kb DNA ladder.

Page 89: At

81

Hybridization analysis was performed on the three non-A. pleuropneumoniae

species from which DNA products were amplified, as well as A. pleuropneumoniae

serotypes 2 and 4. The multiplex PCR assay was performed at a MgCl2 concentration of

5 mM in order to amplify all possible PCR products (Fig. 2.13a). The bands were first

probed with a labeled PCR product generated by the cpx primers C and D from A.

pleuropneumoniae serotype 5. Hybridization of the probe to the other PCR products

was performed at a high stringency of 68o C and 5X SSC. There was no hybridization of

the probe to any of the bands produced by serotype 4 or the three non-A.

pleuropneumoniae species (Fig. 2.13b). The cpx probe only hybridized to the 0.7 kb cpx

bands produced by serotype 2 and the serotype 5 positive control. A second Southern

hybridization was performed with the labeled product of cps primers A and. The cps

probe hybridized to a band slightly less than 1.0 kb in serotypes 2 and 4 as well as the

1.1 cps band of serotype 5. The cps probe did not hybridize to any products from the

other respiratory pathogens.

Page 90: At

82

Figure 2.13a. Agarose gel electrophoresis of the PCR products from bacterial samplesused for Southern hybridizations. Lane 1, serotype 2; lane 2, serotype 4; lane 3, serotype5; lane 4, S. suis; lane 5, P. multocida; lane 6, H. parasuis .

Page 91: At

83

Figure 2.13b. Southern blot of the PCR products from bacterial samples hybridized witha cpx probe. Lane 1, serotype 2; lane 2, serotype 4; lane 3, serotype 5; lane 4, S. suis; lane5, P. multocida; lane 6, H. parasuis .

Page 92: At

84

Assay of clinical specimens

The multiplex PCR was used to determine if A. pleuropneumoniae serotypes 1, 2,

and 5 DNA could be detected from lung tissue of infected swine (Fig. 2.14). The assay

was unable to detect any amplified product from serotype 1 samples. Serotype 2 samples

either produced a very faint 0.7 kb band (lane 4 of Figure 14) or no visible product.

Serotype 5 samples gave two distinct products consistent with the 0.7 kb cpx band and

the 1.1 kb cps band. The PCR assay on lung tissue was performed at a MgCl2

concentration of 5 mM to enhance sensitivity.

Page 93: At

85

Figure 2.14. Agarose gel electrophoresis of PCR products from lung tissue samples takenfrom swine that had been infected with serotypes 1, 2, or 5. Lane 1, 1-kb DNA ladder;lane 2, serotype 1; lane 3, serotype 2, sample 1; lane 4, serotype 2, sample 2; lane 5,serotype 5.

Page 94: At

86

Attempts to detect A. pleuropneumoniae serotypes 2 and 5 using the multiplex

PCR assay were unsuccessful from nasal swabs. No PCR products were detected from

any of the swabs tested when using the boiling protocol for sample preparation or when

prepared using the Qiagen tissue preparation kit. However, serologic assays (latex

agglutination) and culture did not confirm that any bacteria were present in these

specimens.

To determine if the samples themselves were inhibiting the assay, genomic DNA

from serotypes 2 or 4 were added to the corresponding nasal swab sample. Figure 2.15

shows the bands produced from the Qiagen preparations, boiled preparations, and

samples taken directly from the nasal swabs. PCR amplification of the Qiagen prepared

samples appeared to amplify the genomic DNA without noticeable differences from the

standard conditions of genomic amplification. However, the other 2 samples gave no

products for serotype 2 and reduced amplification for serotype 5.

Page 95: At

87

Figure 2.15. Agarose gel electrophoresis of the PCR products from genomic DNA of theappropriate serotype added to nasal swab samples. Lane 1, 1-kb DNA ladder; lane 2,serotype 2 Qiagen sample; lane 3, serotype 5 Qiagen sample; lane 4, serotype 2 boiledsample; lane 5, serotype 5 boiled sample; lane 6, serotype 2 sample; lane 7, serotype 5sample.

Page 96: At

88

DISCUSSION

A. pleuropneumoniae serotypes are distinguished by their unique capsular

polysaccharide (Inzana and Mathison., 1987a). Because cross-reactions often occur from

serotyping with traditional serologic assays, PCR offers a practical alternative that does

not employ antigens or antibodies. This attribute makes capsule genes an ideal target for

typing by PCR. Falla et al. (1994) previously reported using primers from the capsular

DNA region as a reliable method for the typing of H. influenzae by PCR. To date, a

reliable method for serotyping A. pleuropneumoniae by PCR has not yet been

established. Hennessy et al. (1993) have proposed an arbitrarily primed PCR assay for

serotyping A. pleuropneumoniae, but this method can only be used with pure A.

pleuropneumoniae template. The current describes the first use of primers to target

capsular DNA regions to simultaneously identify the species and serotype of A.

pleuropneumoniae.

Both the cps and cpx regions of serotype 5 DNA were successfully amplified with

samples of purified DNA, bacterial colonies, and lung tissue. Combining the primers

together did not require any change in PCR conditions. The use of multiplex PCR

provided the advantage of using multiple primer sets in a single reaction and

simultaneously determining both the species and the serotype: in this case A.

pleuropneumoniae and serotype 5. The detectable limit of the PCR products of serotype

5 by agarose gel electrophoresis was less than 1 x 102 CFU’s.

The presence of non-specific bands amplified by some serotypes was initially

problematic, particularly from serotypes 2, 3, and 6 while maintaining the cpx band in all

Page 97: At

89

serotypes. Although the cpx region appears to be highly conserved, the primers selected

from that region were designed from the sequence of serotype 5 capsular DNA. Because

there is no sequence data available on the capsular region of other serotypes, the amount

of homology of these primers to other serotypes is unknown. The MgCl2 concentration

was the single most important parameter involved in the specificity of PCR amplification,

and was successfully used to control the presence of non-specific bands.

The successful application of the PCR assay to bacterial colonies provided an

effective method of identifying A. pleuropneumoniae. With the exception of the rare

serotype 4, all serotypes amplified a distinct 0.7 kb band. Because both the preparation

and detection of DNA was relatively simple, the entire assay could be performed in under

five hours. Among the common swine respiratory pathogens tested, only S. suis

produced a band of similar size. However, this band was determined to be non-specific as

it did not hybridize with the cpx probe and was distinguished from the A.

pleuropneumoniae cpx band by Southern blotting. In addition, S. suis presents a distinct

clinical picture from that of A. pleuropneumoniae and can, therefore, be distinguished

clinically.

The amplification of the cpx product in all of the serotypes, with the exception

of serotype 4, supports the existing evidence that the capsular export region is highly

conserved among the A. pleuropneumoniae serotypes (Ward, 1995). The results of the

Southern blot also indicated that the 0.7 kb band of serotype 2 contained homology to the

0.7 kb band of serotype 5. Although the 0.7 kb product was not amplified from serotype

4, it is not surprising that even within a highly conserved region there may be some areas

Page 98: At

90

of non-homology. Of interest, however, was that cps primers A and B did not produce a

1.1 kb band from serotypes other than serotype 5, but the cps probe hybridized to a band

from both serotype 2 and serotype 4. This was surprising since the cps region of A.

pleuropneumoniae is antigenically serotype-specific (Ward, 1995). However, it is

possible that the cps loci may contain some genes that are involved with capsular

synthesis that are conserved by all serotypes. For instance, serotypes 1 and 5 both

conatain N-acetylglucosamine (Perry et al., 1990). Sequencing the cps loci of other

serotypes would reveal any homologous genes responsible for encoding similar proteins.

The potential for using this assay with clinical specimens was also investigated.

A. pleuropneumoniae serotype 5 DNA from lung tissue samples of infected swine was

successfully amplified by PCR. PCR assays have been reported to have greater

sensitivity in detecting bacteria than by the culture of clinical samples (Rodu, 1990).

Therefore, this PCR assay may prove to be faster and a more sensitive method for

identifying and serotyping A. pleuropneumoniae. Although this assay was designed to

specifically detect serotype 5 strains, future development of the protocol may result in

the detection of all serotypes.

Although the identification and serotyping of A. pleuropneumoniae from nasal

swabs of live pigs would be advantageous, attempts to develop a reliable method have

been largely unsuccessful (Kume et al., 1984; Sirois et al., 1991). Isolation of A.

pleuropneumoniae is difficult because of the overgrowth of normal flora. Nasal swab

specimens have also been reported to inhibit PCR (Wadowsky et al., 1994). Evidence of

Page 99: At

91

this was shown in Figure 2.15 when genomic DNA added to nasal swab specimens was

not amplified as well as genomic DNA alone.

In conclusion, the PCR assay described was effective in detecting A.

pleuropneumoniae and identifying serotype 5 from whole bacterial cells and infected lung

tissue. This assay can be done both quickly and easily. Once the sequences for the

capsular regions of other serotypes have been determined, the assay can be expanded to

serotype any strain of A. pleuropneumoniae. This work has described a potentially

useful method for detecting and serotyping A. pleuropneumoniae with both high

specificity and sensitivity while avoiding the problems that are associated with serologic

assays.

Page 100: At

92

Literature Cited

Ausch, D., and M. Reth. 1990. Exon trap cloning: using PCR to rapidly detect and cloneexons form genomic DNA fragments. Nucleic Acids. Res. 18:6743.

Bej, A. K., M. H. Mahbubani, and R. M. Atlas. 1991. Amplification of nucleic acids bypolymerase chain reaction (PCR) and other methods and their applications. Critical Rev.Biochem. Mol. Biol. 26:301-334.

Bej, A. K., M. H. Mahbubani, R. Miller, J. L. DiCesare, L. Haff, and R. M. Atlas. 1990.Multiplex PCR amplification and immobilized capture probes for detection of bacterialpathogens and indicators in water. Mol. Cell. Probes. 4:353-365.

Belanger, M., D. Dubreuil, J. Harel, C. Girard, and M. Jacques. 1990. Role oflipopolysaccharides in adherence of Actinobacillus pleuropneumoniae to porcine trachealrings. Infect. Immun. 58:3523-3530.

Bertram, T. 1986. Intravascular macrophages in lungs of pigs infected with Haemophiluspleuropneumoniae. Vet. Pathol. 23:681-691.

Bosse, J.T., R. P. Johnson, and S. Rosendal. 1990. Capsular polysaccharide antigens fordetection of serotype specific antibodies to Actinobacillus pleuropneumoniae. Can. J. Vet.Res. 54:320-325..

Brandreth, S. R. and I. M. Smith. 1985. Prevalence of pig herds by pleuropneumoniaassociated with Haemophilus pleuropneumoniae in eastern England. Vet. Record.117:143-147.

Byrd, W., and S. Kadis. 1989. Structures and sugar compositions of lipopolysaccharidesisolated from seven Actinobacillus pleuropneumoniae serotypes. Infect. Immun. 57:3901-3906.

Cai, S. P., J. P. Zhang, D. H. Huang, Z. X. Wang, and Y. W. Kan. 1988. A simpleapproach to prenatal diagnosis of Beta-thalassemia in geographic area where multiplemutations occur. Blood. 71:1357-1360.

Chamberlain, J., R. A. Gibbs, J. E. Rainer, P. N. Nguyen, and C. T. Caskey. 1988.Deletion screening of the Duchenne muscle dystrophy locus via multiplex DNAamplification. Nucleic Acids Res. 16:141-156.

Chang, Y. F., J. R. Shi, D. P. Ma, S. J. Shin, and D. H. Lein. 1993. Molecular analysis ofthe Actinobacillus pleuropneumoniae RTX Toxin-III gene cluster. DNA Cell Biol. 12:351-362.

Page 101: At

93

Chang, Y. F., R. Young, and D. K. Struck. 1989. Cloning and characterization of ahemolysin gene from Actinobacillus (Haemophilus) pleuropneumoniae. DNA. 8:635-647.

Chehab, F. F., M. Doherty, S. Cai, Y. W. Kan, S. Cooper, and E. W. Rubin. 1987.Detection of sickle cell anaemia and thalassaemias. Nature. 329:293-294.

Cooper, D. L., and N. Isola. 1990. Full length cDNA cloning utilizing the polymerasechain reaction, a degenerate oligonucleotide sequence and a universal mRNA primer.BioTechniqhes. 9:60-65.

Demmler, G. J., G. J. Buffone, C. M. Schimbor, and R. A. May. 1988. Detection ofcytomegalovirus in urine from newborns by using polymerase chain reaction DNAamplification. J. Infect. Dis. 158:1177-1184.

Desrosiers, R., K. R. Mittal, and R. Malo. 1984. Porcine pleuropneumonia associatedwith Haemophilus pleuropneumoniae serotype 3 in Quebec. Vet. Rec. 115:628-629.

Devenish, J., and S. Rosendal. 1991. Calcium binds to and is required for biologicalactivity of the 104 kilodalton hemolysin of Actinobacillus pleuropneumoniae serotype 1.Can. J. Microbiol. 37:317-321.

DeWitt, D., L. Steyn, S. Shoemaker, and M. Sogin. 1990. Direct detection ofMycobacterium tuberculosis in clinical specimens by DNA amplification. J. Clin.Microbiol. 28:2437.

DiLella, A. G., W. M. Huang, and S. L. Woo. 1988. Screening for phenylketonuriamutations by DNA amplification with the polymerase chain reaction. Lancet. 1:497-499.

Erlich, H. A., D. Gefland, and J. J. Sninsky. Recent advances in the polymerase chainreaction. 1991. Science. 252:1643-1651.

Erlich, H. A., H. H. Kazazian Jr., and R. A. Gibbs. 1989. Current Communications inMolecular Biology. Cold Spring Harbor Laboratory Press, New York.

Falla, T. J., D. M. Crook, L. N. Brophy, D. Maskell, J. S. Kroll, E. R. Moxon. 1994.PCR for capsular typing of Haemophilus influenzae. J. Clin. Microbiol. 32:2382-2386.

Fedorka-Cray, P. J., L. Hoffman, W. C. Cray, J. T. Gray, S. A. Breish, and G. A.Anderson. 1993. Actinobacillus (Haemophilus) pleuropneumoniae. Part I. History,epidemiology, serotyping, and treatment. Compend. Contin. Ed. Practic. Vet. 15:1447-1455.

Page 102: At

94

Femlee, T., S. Pellet, and R. A. Welch. 1985. Nucleotide sequence of an Escherichia colichromosomal hemolysin. J. Bacteriol. 163:94-105

Fenwick, B. W. and S. Henry. 1994. Porcine pleuropneumoniae. JAVMA. 204:1334-1340.

Fenwick, B. W., and B. I. Osburn. 1986. Immune responses to the lipopolysaccharidesand capsular polysaccharides of Haemophilus pleuropneumoniae in convalescent andimmunized pigs. Infect. Immun. 54:575-582.

Frankel, G., J. A. Giron, Vallmassoi, and G. K. Schoolnik. 1989. Multi-geneamplification: simultaneous detection of three virulence genes in diarrhoeal stool. Mol.Microbiol. 3:1729-1734.

Frey, J. 1995. Virulence in Actinobacillus pleuropneumoniae and the RTX toxins. Trendsin Micro. 3:257-261.

Frey, J., J. T. Bosse, Y. F. Chang, J. M. Cullen, B. Fenwick, G. F. Gerlach, D. Gygi, F.Haesebrouck, T. J. Inzana, R. Jansen, E. M. Kamp, J. Mcdonald, J. I. MacInnes, K. R.Mittal, J. Nicolet, A. N. Rycroft, R. Segers, M. Smits, E. Stenbaek, D. Struck, J.Vandenbosch, P. Wilson, and R. Young. 1993. Actinobacillus pleuropneumoniae RTX-toxins: uniform designation of haemolysins, cytolysins, pleurotoxin, and their genes. J.Gen. Microbiol. 139:1723-1728.

Frey, J., and J. Nicolet. 1988. Purification and partial characterization of a hemolysinproduced by Actinobacillus pleuropneumoniae type strain 4074. FEMS Microbiol. Lett.55:41-46.

Frey, J., and J. Nicolet. 1990. Hemolysin patterns of Actinobacillus pleuropneumoniae. J.Clin. Microbiol. 28:232-236.

Frosch, M., U. Edwards, K. Bousset, B. Krause, and C. Weisgerber. 1991. Evidence for acommon molecular origin of the capsule gene loci in gram-negative bacteria expressinggroup II capsular polysaccharides. Mol. Microbiol. 5:1251-1263.

Giese, S. B., E. Stebaek, and R. Nielsen. 1993. Identification of Actinobacilluspleuropneumoniae serotype 2 by monoclonal or polyclonal antibodies in latexagglutination tests. Acta Vet. Scand. 34:223-225.

Gingeras, T. R., D. D. Richman, D. Y. Kwoh, and J. C. Guatelli. 1990. Methodologies forin vitro nucleic acid amplification and their applications. Vet. Microbiol. 24:235-251.

Page 103: At

95

Gottschalk, M., E. Altman, N. Charland, F. D. Lasalle, and J. D. Dubreuil. 1994a.Evaluation of a saline boiled extract, capsular polysaccharides and long-chainlipopolysaccharides of Actinobacillus pleuropneumoniae serotype 1 as antigens for theserodiagnosis of swine pleuropneumonia. Vet. Microbiol. 42:91-104.

Gottschalk, M., F. D. Lasalle, S. Radacovici, J. D. Dubreuil. 1994b. Evaluation of longchain lipopolysachharides (LC-LPS) of Actinobacillus pleuropneumoniae serotype 5 forthe serodiagnosis of swine pleuropneumonia. Vet. Microbiol. 38:315-327.

Gram, T., P. Ahrens, J. P. Nielsen. 1996. Evaluation of a PCR for detection ofActinobacillus pleuropneumoniae in mixed bacterial cultures from tonsils. Vet. Microbiol.51:95-104.

Gunnarsson, A. 1979. Serologic studies on porcine strains of Haemphilusparahaemolyticus (pleuropneumoniae): extraction of type-specific antigens. Am. J. Vet.Res. 40:469-472.

Gunnarsson, A. and P. Branefors-Helander. 1979. Antigenic relationship betweenHaemophilus influenzae and Haemophilus isolated from porcine pleuropneumoniae.Current Microbiol. 2:21-24.

Gunnarsson, A., B. Harrel, and E. L. Biberstein. 1978. Serologic studies of porcine strainsof Haemophilus parahaemolyticus (pleuropneumoniae): antigenic specificity andrelationship between serotypes. Am. J. Vet. Res. 39:1286-1292.

Gygi, D., J. Nicolet, C. Hughes, and J. Frey. 1992. Functional analysis of the Ca2+-regulated hemolysin I operon of Actinobacillus pleuropneumoniae serotype 1. Infect.Immun. 60:3059-3064.

Haqqi, T. M., G. Sarkar, C. S. David, and S. S. Sommer. 1988. Specific amplification withPCR of a refractory segment of genomic DNA. Nucleic Acids Res. 16:11844.

Hemsley, A., N. Arnheim, M. D. Toney, G. Cortopassi, and D. J. Galas. 1989. A simplemethod for site-directed mutagenesis using the polymerase chain reaction. Nucleic AcidsRes. 17:6545-6551.

Hennessy, K. J., J. J. Iandolo, and B.W. Fenwick. 1993. Serotype identification ofActinobacillus pleuropneumoniae by arbitrarily primed polymerase chain reaction. J. Clin.Microbiol. 31:1155-1159.

Herbert, W. J. 1967. Passive haemagglutination. Handbook of Immunology. p. 720-741.

Page 104: At

96

Hoffman, L. 1989. Actinobacillus (Haemophilus) pleuropneumoniae: use ofcoagglutination and complement fixation to determine the relationship between presenceof organism and antibody titer in slaughterhouse pigs. J. Vet. Diagn. Invest. 1:12-15.

Houard, S., C. Hackel, A. Herzog, and A. Bollen. 1989. Specific identification ofBordetella pertussis by the polymerase chain reaction. Res. Micobiol. 140:477-487.

Hunneman, W. A. 1986. Incidence, economic effects, and control of Haemophiluspleuropneumoniae infections in pigs. Vet. Q. 8:83-87.

Inzana, T. J. 1991. Virulence properties of Actinobacillus pleuropneumoniae. Microb.Path. 11:305-316.

Inzana, T. J. 1995. Simplified procedure for preparation of sensitized latex particles todetect capsular polysaccharide: application to typing and diagnosis of Actinobacilluspleuropneumoniae. J. Clin. Microbiol. 33:2297-2303.

Inzana, T. J., B. Iritani, R. P. Gogolewski, and P. Anderson. 1988. Virulence propertiesand protective efficacy of the capsular polymer of Haemophilus (Actinobacillus)pleuropneumoniae serotype 5. Infect. Immun. 56:1880-1889.

Inzana, T. J., and B. Mathison. 1987a. Type-specificity and immunogenicity of thecapsular polymer of Haemophilus (Actinobacillus) pleuropneumoniae. Infect. Immun.55:1580-1587.

Inzana, T. J., and B. Mathison. 1987b. Serotype specificity and immunogenicity of thecapsular polymer of Haemophilus pleuropneumoniae serotype 5. Infect. Immun.56:1880-1889.

Inzana, T. J., J. Todd, C. Koch, and J. Nicolet. 1992. Serotype specificity ofimmunological assays for the capsular polymer of Actinobacillus pleuropneumoniaeserotypes 1 and 9. Vet. Microbiol. 31:351-362.

Inzana, T. J., J. Todd, and H. Veit. 1993. Safety, stability, and efficacy of noncapsulatedmutants of Actinobacillus pleuropneumoniae for use in live vaccines. Infect. Immun.61:1682-1686.

Jansen, R., J. Briaire, E. M. Kamp, A. Gielkens, and M. A. Smits. 1993. Structureanalysis of the Actinobacillus pleuropneumoniae RTX-toxin-I (ApxI) operon. Infect.Immun. 61:3688-3695.

Page 105: At

97

Jensen, A. E., and T. A. Bertram. 1986. Morphological and biochemical comparison ofvirulent and avirulent isolates of Haemophilus pleuropneumoniae serotype 5. Infect.Immun. 51:419-424.

Kalman, M., E. T. Kalman, and M. Cashel. 1990. Polymerase chain reaction (PCR)amplification with a single specific primer. Biochem. Biophys. Res. Comm. 167:504-506.

Kamp, E. M., J. K. Popma, J. Anakotta, and M. A. Smits. 1991. Identification ofhemolytic and cytotoxic proteins of Actinobacillus pleuropneumoniae by usingmonoclonal antibodies. Infect. Immun. 59:3079-3085.

Kamp, E. M., T.M. Vermeulen, M. A. Smits, and J. Haagsma. 1994. Production of Apxtoxins by field strains of Actinobacillus pleuropneumoniae and Actinobacillus suis. Infect.Immun. 62:4063-4065.

Kilian, M., E. Biberstein.1984. Haemophilus, in Krieg, N., and J. Holt (eds). Bergey’sManual of Systematic Bacteriology. Vol. 1. Baltimore, MD, William & Wilkins. pp. 558-569.

Kilian, M., J. Nicolet, and E. L. Biberstein. 1978. Biochemical characterization ofHaemophilus pleuropneumoniae (Matthews and Pattison1961) Shope 1964 and proposalof a neotype strain. Int. J. Syst. Bacteriol. 28:20-26.

Kogan, S. C., M. Doherty, J. Gitschler. 1987. An improved method for prenatal diagnosisof genetic diseases by analysis of amplified DNA sequences. New Engl. J. Med. 317:985-990.

Kume, K., I. Nagano, and T. Nakai. 1986. Bacteriological, serological and pathologicalexamination of Haemophilus pleuropneumoniae infection in 200 slaughtered pigs. Jpn. J.Vet. Sci. 48:965-970.

Kume, K., T. Nakai, and A. Sawata. 1984. Isolation of Haemophilus pleuropneumoniaefrom the nasal cavities of healthy pigs. Jpn. J. Vet. Sci. 46:641-647.

Kwok, S., D. H. Mack, J. J. Sninsky, G. D. Ehrlich, B. J. Poiesz, N. L. Dock, H. J. Alter,D. Mildvan, and M. H. Grieco. 1989. Diagnosis of human immunodeficiency virus inseropositive individuals: enzymatic amplification HIV viral sequences in peripheral bloodmononuclear cells, in HIV Detection by Genetic Engineering Methods, Luciw, P. A. and K.S. Steimer, Eds., Marcel Decker, New York. p. 243-252.

Page 106: At

98

Lalonde, G., T. V. McDonald, P. Gardner, and P. D. O’Hanley. 1989. Identification of ahemolysin from Actinobacillus pleuropneumoniae and characterization of its channelproperties in planar phospholipid bilayers. J. Biol. Chem. 264:13559-13564.

Leman, A. D. 1992. The decision to repopulate. In Proceedings. Am. Assoc. Swine Pract.pp. 9-12.

Loh, E. Y., J. F. Elliott, S. Cwirla, L. L. Lanier, and M. M. Davis. 1989. Polymerase chainreaction with single-sided specificity: analysis of T cell receptor delta chain. Science.243:217-220.

Lombin, L. H., S. Rosendal and W. R. Mitchell. (1982). Evaluation of the complementfixation test for the diagnosis of pleuropneumoniae of swine caused by Haemophiluspleuropneumoniae. Can. J. Comp. Med. 46:109-114.

MacInnes, J. I., and S. Rosendal.1988. Prevention and control of Actinobacillus(Haemophilus) pleuropneumoniae infection in swine: A review. Can. Vet. J. 29:572-573.

Mack, D. H. and J. J. Sninsky. 1988. A sensitive method for the identification ofuncharacterized viruses related to known virus groups: hepadnavirus model system. Proc.Natl. Acad. Sci. 85:6977-6981.

Matthew, P. R. and I. H. Pattison. 1961. The identification of a Haemophilus-likeorganism associated with pneumonia and pleurisy in the pig. J. Comp. Pathol. 71:44-52.

Mittal, K. R.. 1990. Cross-reactions between Actinobacillus (Haemophilus)pleuropneumoniae strains of serotypes 1 and 9. J. Clin. Microbiol. 28:535-539.

Mittal K. R., R. Higgins, and S. Lariviere. 1982. Evaluation of slide agglutination and ringprecipitation tests for capsular serotyping of Haemophilus pleuropneumoniae. J. Clin.Microbiol. 15:1019-1023.

Mittal, K. R., R. Higgins, and S. Lariviere. 1983a. Determination of antigenic specificityand relationship among Haemophilus pleuropneumoniae serotypes by an indirecthemagglutination test. J. Clin. Microbiol. 17:787-790.

Mittal, K. R., R. Higgins, and S. Lariviere. 1983b. Identification and serotyping ofHaemophilus pleuropneumoniae by coagglutination test. 18:1351-1354.

Mittal, K. R., R. Higgins, and S. Lariviere. 1984. A 2-mercaptoethanol tube agglutinationtest for diagnosis of Haemophilus pleuropneumoniae infection in pigs. Am. J. Vet. Res.45:715-719.

Page 107: At

99

Mittal, K. R., R. Higgins, and S. Lariviere. 1987. An evaluation of agglutination andcoagglutination techniques for serotyping of Haemophilus pleuropneumoniae isolates.Am. J. Vet. Res. 48:219-226.

Mitui, T., H. Onaga, Y. Nagasawa, Y. Nomura, and S. Kuramasu. 1981. Studies onHaemophilus infection in swine I. Application of the latex agglutination test to thediagnosis of Haemophilus pleuropneumoniae (H. parahaemaelyticus) infection. Vet.Microbiol. 6:339-349.

Mullis, K. B. 1990. The unusual origin of the polymerase chain reaction. Sci. Am. 262:56-65.

Mullis, K. B. and F. A. Falooona. 1987. Specific synthesis of DNA in vitro viapolymerase catalyzed chain reaction. Methods in Enzymology. 155:335-350.Neter, E. 1956. Bacterial hemagglutination and hemolysis. Bacteriol. Rev. 20:166-168.

Nicolet, J. 1988. Taxonomy and serological identification of Actinobacilluspleuropneumoniae. Can. Vet. J. 29:578-579.

Nicolet, J. 1992. Actinobacillus pleuropneumoniae. In, A.D. Leman, B. E. Straw, W.L.Mengeling, S. D’Allaire, and D. J. Taylor (ed), Diseases of swine, Iowa State UniversityPress, Ames. 401-408.

Nicolet, J., M. Krawinkler, and A. Baumgartner. 1981. An enzyme-linked immunosorbentassay, using an EDTA-extractacted antigen for the serology of Haemophiluspleuropneumoniae. Am. J. Vet. Res. 42:2139-2142.

Nielsen, R. 1974. Serological and immunological studies of pleuropneumonia of swinecaused by Haemophilus parahaemolyticus. Acta Vet. Scand. 15:80-89.

Nielsen, R. 1979. Haemophilus parahaemolyticus serotypes serological response. Nord.Vet. Med. 31:401-406.

Nielsen, R. 1985. Serological characterization of Haemophilus pleuropneumonia(Actinobacillus pleuropneumoniae) strains and proposal of a new serotype: serotype 9.Acta Vet. Scand. 26:501-512.

Nielsen, R., T. Plambeck, and N. T. Foged. 1993. Blocking enzyme-linkedimmunosorbent assay for detection of antibodies against Actinobacillus pleuropneumoniaeserotype 8. 34:131-138.

Page 108: At

100

Niven, D. F., and Levesque, M. 1988. V-factor dependent growth of Actinobacilluspleuropneumoniae biotype 2 (Bertschinger 2008/76). Int. J. Syst. Bacteriol. 38:319-320.

Ochman, H., J. W. Ajioka, D. Garza, and D. L. Hartl. 1990. Inverse polymerase chainreaction. Biotechnology. 8:759-760.

Orrego, C., A. C. Wilson, and M. C. King. 1988. Identification of maternally relatedindividuals by amplification and direct sequencing of a highly polymorphic, noncodingregion of mitochondrial. Am. J. Hum. Genet. 43:A219.

Oste, C. 1989. Optimization of magnesium concentration in the PCR reaction,Amplifications - A forum for PCR users. 1:10-13.

Perry, M. B., E. Altman, J.-R. Brisson, L. M. Beynon, and J. C. Richards. 1990.Structural characteristics of the antigenic capsular polysaccharides andlipopolysaccharides involved in the serological classification of Actinobacilluspleuropneumoniae strains. Serodiag. Immun. in Infect. Dis. 4:299-308.

Pohl, S., H. U. Bertschinger, W. Frederiksen, and W. Mannheim.1983. Transfer ofHaemophilus pleuropneumoniae and Pasteurella haemolytica-like organism causingporcine necrotic pleuropneumonia to the genus Actinobacillus (Actinobacilluspleuropneumoniae comb. nov.) On the basis of phenotypic and deoxyribonucleic acidrelatedness. Int. J. Syst. Bacteriol. 33:510-514.

Radacovici, S., M. Gottschalk, and J. D. Dubreuil. 1994. Lipopolysaccharides ofActinobacillus pleuropneumoniae (serotype 1): a readily attainable antigen for ELISAserodiagnosis of pig pleuropneumoniae. Vet. Microbiol. 39:219-230.

Rapp, V. J., R. F. Ross and B. Z. Erickson. 1985. Serotyping of Haemophiluspleuropneumoniae by rapid slide agglutination and indirect fluorescent antibody tests inswine. Am. J. Vet. Res. 46:185-192.

Rapp, V.J., R. F. Ross, amd T. F. Young. 1985. Characterization of Haemophilus spp.isolated from healthy swine and evaluation of cross-reactivity of complement-fixingantibodies to Haemophilus pleuropneumoniae and Haemophilus taxon “minor group”. J.Clin. Microbiol. 22:945-950.

Rodu, B. 1990. Molecular biology in medicine the polymerase chain reaction: therevolution within. Am. J. Med. Sci. 299:210-216.

Rogers, R. J., L. E. Eaves, P. J. Blackall, and K. F. Truman. 1990. The comparativepathogenicity of four serovars of Actinobacillus (Haemophilus) pleuropneumoniae.Australian Vet. J. 67:9-12.

Page 109: At

101

Rohrbach, B. W., R. F. Hall, and J. P. Hitchcock. 1993. Effect of subclinical infectionwith Actinobacillus pleuropneumoniae in commingled feeder swine. JAVMA. 202:1095-1098.

Rosendal, S. and D. Boyd. 1982. Haemophilus pleuropneumoniae serotyping. J. Clin.Microbiol. 16:840-843.

Rosendal, S., D. Boyd, and K. A. Gilbride. 1985. Comparative virulence of porcineHaemophilus bacteria. Can J. Comp. Med. 49:68-74.

Rosendal, S., L. Lombin, and J. DeMoor. 1981. Serotyping and detection ofHaemophilus pleuropneumoniae by indirect fluorescent antibody technique. Can J.Comp. Med. 45:271-274.Rosendal, S. and W. R. Mitchell, 1983. Epidemiology of Haemophilus pleuropneumoniaeinfection in pigs: A survey of Ontario pork producers, 1981. Can J. Comp. Med. 47:1-5.

Rosendal, S., W. R. Mitchell, M. Weber, M. R. Wilson, M. R. Zaman. 1980.Haemophilus pleuropneumoniae lung lesions induced by sonicated bacteria and sterileculture supernatant. Proc. Int. Pig Vet. Soc. 5:211.

Ruano, G., W. Fenton, and K. K. Kidd. 1989. Biphasic amplification of very dilute DNAsamples via “booster” PCR. Nucleic Acids Res. 17:5407.

Saiki, R. K., D. H. Gelfand, S. Stoffel, S. J. Scharf, R. Higuchi,, G. T. Horn, K. B. Mullis,and H. A. Erlich. 1988. Primer-directed enzymatic amplification of DNA with athermostable DNA polymerase. Science. 239:487-491.

Saiki, R. K., S. Scharf, F. Faloona, K. B. Mullis, G. T. Horn, A. Erlich, and N. Arnheim.1985. Enzymatic amplification of Beta-globulin genomic sequences and restriction siteanaysis for diagnosis of sickle cell anemia. Science. 230:1350-1354.

Saito, I., B. Servenius, T. Compton, and R. I. Fox. 1989. Detection of Epstein-Barr virusDNA by polymerase chain reaction in blood and tissue biopsies from patients withSjogren’s syndrome. J. Exp. Med. 169:2191.

Scharf, S. J., G. T. Horn, and H. A. Erlich. 1986. Direct cloning and sequence analysis ofenzymatically amplified genomic sequences. Science. 233:1076-1078.

Sebunya, T. K., and J. R. Saunders.1983. Haemophilus pleuropneumoniae infections inswine: A review. JAVMA. 182:1331-1337.

Page 110: At

102

Shope, R. R. 1964. Porcine contagious pleuropneumonia I. Experimental transmission,etiology and pathology. J. Exp. Med. 1119:357-358.

Shultz, R. A., R. F. Ross, A. Gunnarsson, and R. Nielsen. 1983. Serotyping of 50different isolates of Haemophilus pleuropneumoniae from swine pneumonia in Iowa andsurrounding states. Vet. Med./Small Animal Clin. 78:1451-1453.

Sirois, M., and R. Higgins. 1991. Biochemical typing of Actinobacillus pleuropneumoniae.Vet. Microbiol. 27:397-401.

Sirois, M., E. G. Lemire, and R. C. Levesque. 1991. Construction of a DNA probe anddetection of Actinobacillus pleuropneumoniae by using Polymerase Chain Reaction. J.Clin. Microbiol. 29: 1183-1187.

Stenbaek, E. I., and A. L. Schirmer. 1994. Detection of Actinobacillus pleuropneumoniaeserotype 2 antibodies in pig sera by an inhibition enzyme immunoassay (EIA). Vet.Microbiol. 39:231-244.

Stoflet, E. S., D. D. Koeberl, G. Skar, and S. S. Sommer. 1988. Genomic amplificationwith transcript sequencing. Science. 239: 491-494.

Suzuki, S., H., Sakakibara and S. Hotta. 1977. Latex agglutination test for measurement ofantiplague antibodies. J. Clin.Microbiol. 6:332-336.

Trottier, Y. L., P. F. Wright, and S. Lariviere. 1992. Optimization and standarization of anenzyme-linked immunosorbent assay protocol for serodiagnosis of Actinobacilluspleuropneumoniae serotype 5. J. Clin. Microbiol. 30:46-53.

Udeze, F. A., K. S. Latimer, and S. Kadis. 1987. Role of Haemophilus pleuropneumoniaelipopolysaccharide endotoxin in the pathogenesis of porcine Haemophiluspleuropneumoniae. Am. J. Vet. Res. 48:768-773.

Wadowsky, R. M., S. Laus, T. Libert, S. States, and G. Ehrlich. 1994. Inhibition of PCR-based assay for Bordetella pertussis by using calcium alginate fiber and aluminum shaftcomponents of a nasopharyngeal swab. J. Clin. Microbiol. 32:1054-1057.

Ward, C. K., and T. J. Inzana. 1994. Resistance of Actinobacillus pleuropneumoniae tobactericidal antibody and complement is mediated by capsular polysaccharide andblocking antibody specific for lipopolysaccharide. J. Immunol. 153:2110-2121.

Page 111: At

103

Ward, C. K. 1995. The capsular polysaccharide of Actinobacillus pleuropneumoniaeserotype 5A: Role in serum resistance and characterization of the genetic basis forexpression. Ph.D. dissertation, College of Vet. Med., Virginia Polytechnic Institute andState University, Blacksburg, VA.

Ward, C. K., and T. J. Inzana. 1997. Identification and characterization of a DNA regioninvolved in the export of capsular polysaccharide by Actinobacillus pleuropneumoniaeserotype 5a. Infect. and Immun. 65:2491-2496.

Ward, D. M., M. M. Bateson, R. Weller, and A.L. Ruff-Roberts. 1992. Ribosomal RNAanalysis of microorganisms as they occur in nature. Adv. Microb. Ecol. 12:219-286.

Weisburg, W. G., S. M. Barns, D. A. Pelletier, and D. J. Lane. 1991. 16S ribosomal DNAamplification for phylogenetic study. J. Bacteriol. 173:697-703.

Welsh, J. and M. McClelland. 1990. Fingerprinting genomes using PCR with arbitraryprimers. Nucl. Acids. Res. 18:7213-7218.

Willson, P. J., G. Falk, and S. Klashinsky. 1987. Detection of Actinobacilluspleuropneumoniae infection in pigs. Can. Vet. J. 28:111-116.

Willson, P. J., and A. D. Osborne. 1985. Comparison of common antibiotic therapies ofHaemophilus pleuropneumonia in pigs. Can. Vet. J. 1985. 26:312-316.

Yon, J. and M. Fied. 1989. Precise gene fusion by PCR. Nucleic Acids Res. 17:4895.

Page 112: At

104