You are on page 1of 22

© CAB International 2003 Animal Health Research Reviews 4(2); 73–93

ISSN 1466-2523 DOI: 10.1079/AHRR200359

Molecular genetic methods in the veterinary


clinical bacteriology laboratory: current
usage and future applications
Hugh Y. Cai1,2, Marie Archambault1, Carlton L. Gyles2 and
John F. Prescott2*
1Laboratory Services Division, Animal Health Laboratory, and 2Department of Pathobiology,
University of Guelph, Guelph, Ontario N1G 2W1, Canada

Received 22 September, 2003; Accepted 27 October, 2003

Abstract

In the last 5 years, numerous molecular methods have been published for the detection and
characterization of bacteria in the field of veterinary medicine. PCR has been the most com-
monly used technology. Although not currently used for clinical veterinary diagnosis, new
technologies such as liquid-phase hybridization, real-time PCR, pathogen load determination
and DNA/protein microarray have been described and have many possible applications in
the clinical bacteriology laboratory because of their sensitivity and efficiency. This review
describes the basic principles and application of recently published DNA-based molecular
techniques for the purpose of veterinary clinical bacteriological diagnosis. It covers advances
in probe hybridization technology, DNA/RNA amplification techniques and other molecular
detection methods, including 16S rRNA analysis for bacterial characterization and DNA
microarrays for bacterial detection. The review briefly summarizes the application of molecu-
lar methods for the diagnosis of specific important bacterial infections of animals, and for
other animal pathogens that are slow or difficult to isolate in the clinical bacteriology labora-
tory. In addition, the molecular detection of antimicrobial resistance genes and of bovine
mastitis pathogens is briefly described and current commercially available tests are listed.

Keywords: molecular genetics, methods, veterinary, clinical bacteriology, molecular beacons,


displacement hybridization, polymerase chain reaction (PCR), real-time PCR

Introduction The overall purpose is to provide an overview that


speeds the adoption of these powerful and important
Since the late 1980s, many molecular genetic methods molecular techniques in animal health diagnosis.
have been developed for the detection and genotyping
of bacteria. The purpose of this review is to describe
various DNA-based molecular techniques for use in vet- DNA based molecular tests
erinary clinical bacteriology for the purpose of animal
health diagnostics. The principles on which these tech- Advances in probe hybridization technology
niques are based, and their use, will be outlined and
more recently published molecular methods with poten- Hybridization assays using DNA probes were developed
tial for bacterial pathogen diagnosis will be described. in the early 1960s. Originally, the assays were performed
in solution and involved complicated procedures,
requiring either ultracentrifugation or chromatography
*Corresponding author: Department of Pathobiology, University of
Guelph, Guelph, Ontario N1G 2W1, Canada and filtration to separate hybridized from unhybridized
E-mail: prescott@uoguelph.ca probes. In the mid-1960s a solid-based hybridization
74 H. Y. Cai et al.

assay, Southern blotting, overcame the complexity of When the loop hybridizes with a specific target, the
working with solutions. In this well-known procedure, annealed stem is forced to separate and the energy in
DNA is transferred onto a solid membrane or filter, and the fluorophore is converted into fluorescence (Fig. 1).
the hybridization is performed on the membrane. The Molecular beacons can be used to identify specific
technique revolutionized DNA detection and dominated nucleic acids in homogeneous solutions, e.g. PCR prod-
the field for over 20 years (Demidov, 2003). ucts. Amplicons can be detected by adding a PCR
Hybridization on a solid system has the limitation of product in a micrometer-sized well containing the previ-
low sensitivity because of the lack of amplification of ously immobilized probe, and reading the fluorescence
targets and adsorption of the probe. In addition, it is generated. Further transfers or washing steps are not
impossible to perform real-time monitoring because it required. This method has been applied to detect PCR
requires washing to remove unhybridized probes. To products of a number of bacteria, including E. coli
overcome these drawbacks, a new generation of solu- O157:H7 (McKillip and Drake, 2000). Besides detecting
tion-based hybridization approaches has been the PCR end products, molecular beacons can be used
developed since the mid-1990s. These methods elimi- to monitor the synthesis of specific nucleic acids in real
nate the need to remove unhybridized probes. As a time (real-time PCR), which will be discussed later.
result, in the near future the solution-based hybridiza- The presence of the hairpin stem in the molecular
tion approach may become more popular than PCR in beacon significantly enhances its specificity because the
microbial detection because of its high sensitivity, sim- hairpin stem competes with the non-perfect matched
plicity and minimal requirement for specialized targets, which enables a molecular beacon to discrimi-
equipment. nate between DNA sequences differing by only a single
nucleotide (Tyagi et al., 1998). Therefore, the molecular
beacon approach is valuable for detection of single base
Traditional nucleic acid probes mutations in DNA, for example in antimicrobial resist-
ance genes of Mycobacterium tuberculosis (Piatek et al.,
Nucleic acid probes bind to a complementary target 2000). For this, the isoniazid or rifampin resistance-
DNA or RNA with high affinity, and can be used for the related genes and intergenic regions were PCR-amplified
identification of microorganisms. A specific sequence is then probed by nine molecular beacon probes, which
selected and a probe is generated by synthesis, by covered the complete amplicons. An isolate was deter-
cloning or by PCR amplification of a genome carrying mined to be drug resistant if one of the nine beacon
the sequence. Nucleic acids extracted from bacteria are probes did not generate fluorescent signals.
transferred to a nitrocellulose membrane, fixed by UV
cross-linking or baking, then hybridized with the spe-
cific probe. Unbound probe is washed away, and the Tripartite molecular beacon and displacement
bound probe is detected by radioactive or enzymatically hybridization
active moieties incorporated into the probe.
Various techniques are used for hybridization. In the Compared with conventional DNA probes, molecular
dot blot, target DNA is spotted directly on the mem- beacon probes are more expensive and difficult to syn-
brane filter. In the Southern blot a specific DNA thesize. In addition, since both ends of molecular
fragment is separated electrophoretically on a gel before beacon probes are covalently modified with fluorophore
transfer onto the membrane. Its advantage is that the and quencher, they do not offer the flexibility for fluo-
size of the target DNA fragment is known. The northern rophore change and the capability for surface
blot is similar to the Southern blot except that the target immobilization (e.g. microarray spotting) through free
is RNA. DNA ends. To overcome these problems, alternative
molecular beacons designated tripartite molecular bea-
cons (TMBs) have been developed (Nutiu and Li, 2002).
Molecular beacons A tripartite molecular beacon contains an unmodified
oligodeoxyribonucleotide (ODN) that forms a molecular
Molecular beacons, first developed by Tyagi et al. in beacon-like structure with two universal single-stranded
1996, are single-stranded DNA molecules, with a fluo- arms. The arms are made with the sequence to anneal a
rophore attached at the 5 end and a quencher at the 3 universal pair of ODNs modified separately with a fluo-
end. The loop portion of the probe is designed to be rophore and a quencher (Fig. 1). A TMB is simply a
specific to the target sequence. The stem contains five to probe with two standard arms that are interchangeable,
seven complementary nucleotides. Therefore, in the so different probes can be synthesized in a conventional
absence of a specific target, the probe forms a hairpin- way, and then combined with universal fluorophore and
like stem–loop structure, keeping the fluorophore and quencher labeled arms. TMBs are as effective as stan-
the quencher together. The energy from the fluorophore dard molecular beacons in detecting matching or
passes to the quencher and is converted into heat. single-base-mutated nucleic acid targets.
Molecular genetic methods in the veterinary clinical bacteriology laboratory 75

A
Target

Standard molecular beacon Target

Target
B

Target

Tripartite molecular beacon

Fig. 1. Molecular beacon. (A) The standard molecular beacon is a stem-and-loop structure. The sequences at the ends of the
probe match and bind, creating the stem, while the rest of the probe is unmatched and unbound, creating the loop that is
specific to the target sequence. While folded this way, the fluorophore at one end of the probe is next to the quencher at the
other end. When the probe binds to a single-stranded DNA template, the structure unfolds, separating the quencher from the
dye and allowing fluorescence. (B) In a tripartite molecular beacon (TMB), the fluorophore and quencher are covalently
linked to two separate short oligodeoxyribonucleotides, which hybridize strongly with corresponding single-stranded arms
extended beyond the short hairpin stem. The fluorophore and quencher are close to each other, and the fluorescence is
quenched. In the presence of the nucleic acid target, the closed-state TMB is transformed into the open state, emitting a
strong fluorescence signal.

Displacement hybridization Peptide nucleic acid probes and locked nucleic acid

Displacement hybridization is another approach used to Peptide nucleic acid (PNA) molecules are artificially syn-
overcome the shortcomings of conventional molecular thesized nucleic acids. In contrast to natural DNA, the
beacons (Li et al., 2002). Instead of a hairpin structure, negatively charged sugar–phosphate backbone is
two complementary ODNs of different length are replaced by a neutral polyamide backbone formed by
labeled with a fluorophore and a quencher. The probes repetitive units of N-(2-aminoethyl) glycine (Fig. 2;
normally form a double-stranded structure and the fluo- Stender et al., 2002). Similar to DNA, PNA consists of
rescence is quenched, but, in the presence of individual nucleotide bases, which enable PNA to
complementary target DNA, the probes are displaced hybridize to complementary nucleic acid targets accord-
and the longer probe hybridizes with the target, so that ing to the Watson and Crick base-pairing rules. Because
the probes become fluorescent. The presence of the of the lack of electrostatic repulsion, a PNA probe has
complementary DNA strand was also found to signifi- stronger binding ability for complementary targets, and
cantly enhance the specificity of the probes. Although hybridizes independently of the salt concentration. With
we could find no reports of the application of TMBs and an unnatural backbone, a PNA probe resists digestion by
displacement hybridization for the detection of bacteria, nucleases and other enzymes. For these reasons, PNA
it appears that these two techniques will work as well as probes have a better shelf life, are more suitable for
or better than conventional molecular beacons, and flex- antisense therapy, and are more stable as self-reporting
ibility and ease of synthesis should make them more probes for real-time PCR methods. In contrast also to
popular. DNA probes, PNA probes are not degraded during PCR
76 H. Y. Cai et al.

OH OH NH
O O Base
Base Base

N
O
O O O O
O O
P P P
O–
O– O n
n n
LNA DNA PNA

Fig. 2. Chemical structures of DNA, locked nucleic acid (LNA) and peptide nucleic acid (PNA). In PNA, the sugar–phosphate
backbone of DNA is replaced by a polyamide backbone, keeping the spacing between the nucleotide bases the same, and
nucleotide bases are attached to the uncharged protein-like (polyamide) backbone. An LNA comprises a lightly modified
RNA backbone, in which the ribose moiety in the sugar–phosphate backbone is constrained by a methylene bridge between
the 2-oxygen and the 4-carbon atoms.

by the endonuclease activity of Taq DNA polymerase ized molecular biology. The PCR method was devised
(Wilhelm et al., 2001). One very useful feature for micro- and named by Mullis and Faloona (1987). The PCR uses
bial identification is that PNA probes penetrate the the principle of primer extension reaction, which hap-
hydrophobic cell wall of bacteria following preparation pens in vivo and is catalysed by DNA polymerase. The
of a standard bacteriological smear on a glass slide, enzyme synthesizes a complementary strand of DNA in
because of the relatively hydrophobic character of PNA the 5 to 3 direction. Instead of using one primer, the
compared with that of DNA probes. PNA probes have PCR uses two primers, each complementary to opposite
been used to identify bacteria such as M. tuberculosis strands of the region of DNA. Consequently, each primer
and Staphylococcus aureus directly from slide smears, a extension reaction directs the synthesis of DNA towards
process that could be completed within 3 hours (Stender the other and the amount of DNA that is synthesized
et al., 1999; Oliveira et al., 2002). This method may thus increases exponentially. A few copies of the target DNA
revolutionize bacterial direct examination approaches in can be amplified to millions of copies after 30–40 cycles
clinical microbiology because it is rapid and specific, and of PCR. Since PCR became practical in the late 1980s,
works for mixed cultures directly on samples without the many techniques have been developed to improve the
need for DNA extraction or amplification. A PCR–PNA- efficiency and accuracy of PCR reactions.
based enzyme-linked immunosorbent assay (ELISA) In ‘nested’ PCR, two pairs of PCR primers are used for
method was described as able to recognize point muta- a single target DNA sequence. The first pair amplifies the
tions in genes associated with isoniazid and rifampin target in the usual PCR reaction, and the second pair
resistance in M. tuberculosis (Bockstahler et al., 2002). (nested primers) binds within the first PCR product and
Similar to PNA, the locked nucleic acid (LNA) is produces a second PCR product that is shorter than the
another type of nucleic acid mimic. LNA comprises a first. The major purpose is to increase the sensitivity of
lightly modified RNA backbone, in which the ribose PCR reactions by a second amplification. However,
moiety in the sugar–phosphate backbone is constrained nested PCR is so sensitive that DNA contamination within
by a methylene bridge between the 2-oxygen and 4- the PCR laboratory is a major concern. The nested PCR
carbon atoms (Fig. 2). The link fixes the sugar ring in also serves to confirm the specificity of the first PCR.
the 3 endo conformation, favoring the formation of ‘Multiplex’ PCR is a PCR reaction using more than one
hybrids with complementary DNA or RNA sequences, so pair of primers to amplify more than one target
that LNA probes have high binding affinity. Like PNA, sequence in the same PCR reaction. This technique is
LNA is not cleavable by nucleases. Compared with PNA, useful for the detection of multiple pathogens or multi-
however, LNA has less ability to invade duplex DNA ple genes at the same time.
because it does not contain an electrically neutral
pseudopeptide backbone (Demidov, 2003).
Real-time PCR

Polymerase chain reaction Conventionally, the size and purity of PCR products are
detected in a separate gel electrophoresis step after the
The PCR is a widely used technique for the amplification PCR is completed. Real-time PCR is an important and
of specific DNA sequences in vitro that has revolution- relatively recently introduced technology that can
Molecular genetic methods in the veterinary clinical bacteriology laboratory 77

detect PCR products during amplification. In a real-time and non-specific DNA, and primer-dimers. Therefore it
PCR instrument, an optical device is connected to the works well only for highly specific PCRs.
reaction tubes to read the reaction signal during each Real-time PCR probes are labeled with fluorescence
cycle of PCR reaction. Compared with conventional that is quenched before hybridization. For example,
PCR, real-time PCR is faster and the risk of contamina- exonuclease probes (hydrolysis probe or TaqMan probe)
tion is reduced with a closed tube system. In regular produce fluorescence after the probe is hydrolysed by
PCR (sometimes called ‘black PCR’), the amplification Taq polymerase, which has 5–3exonuclease activity.
of different concentrations of templates reaches log The hydrolysis separates fluorescein from a quenching
phase sooner or later so that it is not possible to quan- dye, which allows emission of fluorescence.
titate accurately the initial templates by measuring the Hybridization probes are another example. These are
end PCR products. By monitoring the PCR reaction in double probes designed to hybridize side by side on the
real time, the time (or cycle) in which the log phase PCR product. The 3 end of the upstream probe is
appears can be determined. By comparing the time to labeled with fluorescein, which is the donor of fluores-
log phase of the initial template with that of standard cence resonance energy transfer (FRET). The 5 end of
controls, the initial template concentration can be the downstream probes is labeled with an acceptor dye.
determined. It is only when the two probes hybridize closely that
The current approaches used for monitoring PCR in FRET happens and that fluorescence can be detected.
real time include double-stranded DNA (dsDNA) fluores- Molecular beacons can also be used to perform real-
cence dye, fluorescence-labeled probes or primers (Fig. time PCR monitoring. Compared with conventional
3). SYBR Green I is a commonly used dsDNA-binding probes, some real-time PCR probes are more discrimina-
dye, which shows over a 100-fold enhancement of fluo- tory between perfect and imperfect matches, because
rescence once bound to dsDNA. This dye binds to any double probes are used, or the second structure, e.g. the
dsDNA so that no design or optimization of probes is hairpin of the molecular beacon, competes against an
required, but unfortunately it also binds to both specific imperfectly matched target.

Fluorescent dye that SYBR Green I


binds to double-
stranded DNA Target
single-stranded DNA PCR products

Hybridization probe
(FRET probe)
Target

primer Probe
Hydrolysis probe
(TaqMan probe)
polymerase Target PCR products

Light upon extension Extended primer


(LUX) fluorogenic Hairpin primer
Target
primers

Target

Molecular beacon
Probe

Before amplification After amplification

Fig. 3. Different fluorescence monitoring schemes for real-time PCR. SYBR Green I is a double-stranded DNA-binding dye,
which shows more than 100-fold enhancement of fluorescence once bound to PCR products (double-stranded DNA).
Exonuclease probes (hydrolysis probe or TaqMan probe) produce fluorescence after the probe is hydrolysed by Taq poly-
merase. Hybridization probes are double probes that hybridize side by side on the specific PCR product and introduce
fluorescence resonance energy transfer (FRET), which generates strong fluorescence. LUX (light upon extension) fluorogenic
primers emit light strongly when extended in PCR products. Molecular beacons generate fluorescence when they bind to a
specific DNA fragment.
78 H. Y. Cai et al.

Invitrogen (Carlsbad, CA, USA) recently developed a ple, healthy carrier pigs are estimate to shed 102 to 103
new type of real-time probe called LUXTM (light upon Brachyspira hyodysenteriae cells per gram of feces,
extension) fluorogenic primers. LUX primers have whereas pigs with acute swine dysentery can contain 8
stem–loop structures that do not contain a quencher × 107 B. hyodysenteriae cells per gram of mucosa
moiety. Rather, the fluorescent oligonucleotides are (Kinyon et al., 1977). A quantitative PCR would help
designed to self-quench based on sequence context. accurate diagnosis of the disease, which currently can-
LUX primers quench when free in solution, fluoresce not be made simply based on positive/negative PCR
weakly when denatured, and only emit light strongly results.
when extended in PCR products. LUX fluorogenic
primers are less expensive than dual-labeled probes.
Similar to the SYBR Green methods, if the primers non- Nucleic acid sequence-based amplification (NASBA)
specifically anneal to the target template, false-positive
results may occur. The advantage over SYBR Green is NASBA is an amplification technology that targets RNA
that LUX primers can be labeled with different fluores- based on the simultaneous activity of reverse transcrip-
ceins, so that multiplex real-time PCR is possible. tase, RNase and T7 RNA polymerase with two
After the real-time PCR is completed, the melting tem- oligonucleotide primers. In NASBA reactions, RNA is
perature (Tm) of the double-stranded PCR products can first reverse-transcribed into cDNA that is transcribed
be determined by measuring the fluorescence intensity into antisense RNA by T7 RNA polymerase. The anti-
under different temperatures. Since the Tm of a DNA sense RNA is further reverse-transcribed to cDNA. By
duplex is determined by its nucleotide composition, Tm repeating this cycle, NASBA can amplify a desired frag-
profiles can be used for bacterial strain typing. ment more than 1012-fold in 90–120 minutes (Fig. 4).
Many real-time PCR assays have been described for The amplified RNA product can be detected through the
the detection of pathogenic microorganisms (Table 1). use of a target-specific capture probe bound to magnetic
Although no application has been reported using quanti- particles in conjunction with a ruthenium-labeled detec-
tative real-time PCR to solve veterinary clinical tor probe and an instrument (NucliSens Reader;
diagnostic problems, this certainly will be useful when bioMérieux) capable of measuring electrochemilumines-
the techniques are mature and field-validated. For exam- cence. Alternatively, RNA amplified by NASBA can be

Sense RNA 5
Reverse transcriptase Anti-sense primer with T7 RNA
polymerase promotor overhang (P1)
Sense RNA 5

Rnase H 5 DNA

Sense primer (P2)

Reverse transcriptase 5 DNA

5
DNA
T7 RNA polymerase 5 DNA

5 T7 RNA polymerase
Anti-sense RNA 5
5
5

5
DNA
P2 5 Anti-sense RNA 5 DNA
Reverse transcriptase
Reverse transcriptase

DNA 5
DNA 5 5 Anti-sense RNA
RNase H P1

Fig. 4. Principle of nucleic acid sequence-based amplification (NASBA). RNA is first reverse-transcribed into cDNA that is
transcribed into antisense RNA by T7 RNA polymerase. The antisense RNA is further reverse-transcribed to cDNA. By repeat-
ing this cycle, NASBA can amplify a desired fragment more than 1012-fold in 90–120 minutes. NASBA is a continuous,
isothermal process, which can be carried at a constant temperature, e.g. 41°C.
Table 1. Examples of applications of molecular methods for identification of animal pathogens that are difficult to isolate or identify using conventional phenotypic approaches

Organism Molecular assays Highlights and Advantages Disadvantages References


Anaplasma PCR (16S rRNA gene) and Simultaneously detects and differentiates Ehrlichia Sensitivity was not determined; only Bekker et al., 2002
(and Ehrlichia) reverse line blot assay (probe ruminantium, Anaplasma ovis, A. centrale, A. marginale, positive results are meaningful. May
hybridization with PCR A. bovis, E. ovina, Ehrlichia sp. Strain Omatjenne miss some recently described strains
products) and A. phagocytophila from ruminant blood or of A. centrale
Amblyomma variegatum ticks
A. marginale MSP1a gene Identifies and genotypes A. marginale based on number Size of the specific PCR product Lew et al., 2002
(msp1α ) PCR of repeat units of the gene. Simple to use compared varies;difficult to identify non-specific
with other typing methods amplification
A. marginale MSP5 gene MSP5 gene is the major target for PCR assays. Detect 30 Nested PCR is subject to contamination. Torioni de Echaide
nested PCR and A. marginale infected erythrocytes per ml of blood Hybridization is tedious et al., 1998
hybridization with
PCR products
Bartonella B. henselae, B. quintana, Amplifies citrate synthase gene (gltA) of B. henselae, Validated on pure cultures only Norman et al., 1995
and R. prowazekii B. quintana and R. prowazekii, then uses PCR–RFLP
gltA PCR and PCR–RFLP to differentiate the three species
Bartonella gene sequence- Identifies and differentiates different Bartonella Limited database of these house- La Scola et al., 2003
based species identification species based on sequences of house keeping gene, keeping genes is available currently.
e.g. rpoB and gltA No universal primers available;
may miss some undescribed strains
Borrelia B. burgdorferi flagellin gene Isolation and detection of B. burgdorferi DNA from Analytical validation only. Use Exner and Lewinski, 2003
Molecular genetic methods in the veterinary clinical bacteriology laboratory

(fla) real-time PCR clinical sample. Different DNA extraction systems of robotic DNA extraction is not as
including a robot system were compared, which had effective when dealing with
sensitivity between 25 and 50 cells/ml of fluid samples small number of samples
Brucella B. abortus species-specific Identification and discrimination of B. abortus field PCR may be further optimized Bricker et al., 2003
multiplex PCR strains (wild-type biovars 1, 2 and 4) from B. abortus to sharpen the PCR product
vaccine strains, other Brucella species and non-Brucella bands. Some strains of B. suis,
bacteria B. abortus and B. melitensis
share the same PCR profile
B. abortus IS711 and alkB Syb Green I, TaqMan probe and hybridization probes real SYBR Green assay false positive on Newby et al., 2003
gene and real-time PCR time PCR had same sensitivity: detected as low as two non-abortus Brucella and non-
genocopies of B. abortus Brucella strain; TaqMan probe
and hybridization probes
reacted with some B. canis strains
B. melitensis IS711 PCR When tested with 126 aborted sheep fetuses, diagnostic High threshold of detection Leyla et al., 2003
sensitivity was 97.4% and specificity was 100% using 1.4 × 108 c.f.u./ml (stomach
culture as gold standard content)
B. ovis IS6501 PCR Threshold of detection equivalent to culture in detecting The PCR also amplified other Manterola et al., 2003
B. ovis infection in 192 samples of ram semen. For B. abortus and B. melitensis,
experimentally infected rams, culture had a lower the other Brucella that were tested
threshold in the first 5 weeks and PCR had a lower
79

threshold 6–14 weeks after infection continued


80

Table 1. continued

Organism Molecular assays Highlights and Advantages Disadvantages References


B. abortus, B. melitensis Three separate LightCycler FRET assays specific for Validated on pure cultures only. Redkar et al., 2001
and B. suis IS711 real-time B. abortus, B. melitensis and B. suis with B. abortus assay had relatively
PCR detection limit of 25–50 cells low CP value on positive samples
and was sometimes positive
with B. suis. B. suis assay negative
on B. suis biovars 2, 3, 4 and 5.
E. coli was the only non-Brucella
used for specificity test
Campylobacter C. jejuni ORF C TaqMan C. jejuni specific and as sensitive as culture when Ability to quantify was difficult Sails et al., 2003
real-time PCR validated with food samples to evaluate because some cells
may be dead or injured and the
culture method may have
underestimated the total
number of cells
Campylobacter spp. Genus-specific PCR, followed by three separate Need to develop a multiplex Logan et al., 2001
LightCycler real-time PCR real-time PCR assays to identify C. jejuni, real-time PCR (would reduce
C. coli, C. lari, C. upsaliensis, C. hyointestinalis, the procedures) and software
and C. fetus using probe and melting peak analysis to interpret the complicated
results
Chlamydia and C. suis ompA gene nested Sensitivity and specificity of PCR vs culture as standard No highly acceptable gold Sachse et al., 2003
Chlamydophila PCR were 94.4 and 81.0% respectively in testing of standard available (culture was
109 clinical samples from pigs experimentally 300 times less sensitive)
infected with C. suis
23S rRNA gene and Quantitative detection of C. psittaci and C. pecorum DeGraves et al., 2003
Omp1 FRET real-time PCR in cattle virginal swabs. Highly sensitive in detecting
sub-clinical virginities in virgin heifers (53%
prevalence, most with one genome per specimen)
Ehrlichia HE1/HE3 direct PCR, Four assays were compared. TaqMan real-time Syb Green assay detected E. canis, Loftis et al., 2003
chaffeensis VLPT nested PCR, PCR was more specific than Syb Green PCR and more E. ewingii, E. muris and
16S rRNA gene TagMan sensitive (detects 10 gene copies) than conventional PCR A. phagocytophila, as well
real-time PCR and as E. chaffeensis. Validated in
Syb Green real-time PCR very limited number of biological
samples
Helicobacter 16S rRNA gene sequencing Characterization of mixed infection of spiral-shaped Cannot discriminate Helicobacter Misawa et al., 2002
organism from a puppy with bloody diarrhea and Flexispira at species/genus level
Helicobacter pylori 23S Sensitivity 97.0% (64 of 66 biopsy samples); Gold standard: culture and histology Lascols et al., 2003
rRNA gene hybridization specificity 94.6% (123 of 130 samples)
probe real-time PCR
Pathogenic Leptospira Identifies pathogenic Leptospira. Better than other Needs more validation Gravekamp et al., 1993;
multiplex PCR (DNA PCR assays in detecting L. hardjo from bovine urine on other species of animals Wagenaar et al., 2000
fragments of unknown
H. Y. Cai et al.

function)
Leptospira Leptospira 23S rRNA Validated with 145 dog urine samples. Diagnostic Gold standard: compatible Harkin et al., 2003
gene PCR sensitivity 100%, specificity 88.3%. PCR positive clinical signs and seroconversion
before seroconversion titer ≥1:400

Bovine semen 16S genes PCR Detects 100 bacteria/ml. Differentiates L. interrogans, Restriction patterns were difficult Heinemann et al., 2000
and PCR–RFLP L. borgpetersenii, L. noguchii, L. santarosai and L. biflexa to identify. Polyacrylaminde gels
and silver stain are tedious. An
alternative can be sequencing of the
PCR products
Mycobacterium Multiplex PCR for M. bovis Distinguishes M. bovis from M. tuberculosis with one Validated only on pure cultures Shah et al., 2002
bovis (specific fragment of PCR reaction. Highly specific. Threshold of detection:
unknown function) and 4,000 genomes
M. tuberculosis (pncA)
Multiplex PCR for M. bovis Identifies milk M. bovis (10 c.f.u./ml) and Relatively low sensitivity for B. abortus Sreevatsan et al., 2000;
(hsp65) and Brucella B. abortus (2,000 c.f.u./ml)
abortus (BCSP31K gene)
DR region of M. tuberculosis Both methods had a detection limit of 36 cells. The reduced sensitivity of the Taylor et al., 2001
complex conventional Conventional PCR detected 26 of 28 culture-positive LightCycler PCR was considered
(double run) and real-time cattlelymph node specimens (93%), and the as the perturbation of fluorescent
PCR with magnetic LightCycler PCR detected 20 of 28 culture-positive signals by the red iron oxide
sequence capture method cattle lymph node specimens (71%) present in the sequence capture
magnetic beads, which may be
alleviated by heat treatment and
centrifugation to remove the
magnetic beads
Molecular genetic methods in the veterinary clinical bacteriology laboratory

Mycobacterium IS900 PCR IS900 is widely used as a PCR target IS900-like sequences exist in other Englund et al., 2001,
paratuberculosis Mycobacterium spp. 2002; Garrido et al., 2000
IS900 SYBR Green LightCycler O’Mahony and Hill, 2002
Real-time PCR
IS900 molecular beacon Fang et al., 2002
real-time PCR
IS900 TaqMan real-time Kim et al., 2002
PCR
ISMav2 PCR ISMav2 is unique to M. paratuberculosis. Uses False positive with high cell number Stratmann et al., 2002
peptide-mediated magnetic separation method to of M. marinum and M. scrofulaceum
increase sensitivity to 10 cells/ml milk ISMav2 has not been widely used or
validated as a PCR target
Mycoplasma DGGE of a 16S rRNA Differentiates all 10 avian and 7/10 small ruminant DGGE is tedious McAuliffe et al., 2003
gene PCR Mycoplasma spp. tested
Syb Green LightCycler Validated for detection of M. gallisepticum using 98 Gold standard was serology Carli and Eyigor, 2003
Real-time PCR chicken trachea swabs with sensitivity and specificity test only. No non-Mycoplasma
of 64.2% (18/28 chickens) and 100% respectively strains were tested to evaluate
specificity. Syb Green real-time
has been found to have
false-positive result in general
Multiplex TagMan Validated using 50 feline blood samples for detection Quantitation was based on DNA Tasker et al., 2003
81

real-time PCR and quantitation of M. haemofelis and ‘Candidatus copy number. True correlation continued
82 H. Y. Cai et al.

detected specifically in real time through the use of


molecular beacon probes included in the amplification
reaction. One advantage of NASBA over PCR is that it is

Pusterla et al., 2000


a continuous, isothermal process and does not require

Mott et al., 1997


expensive thermocycling equipment. In addition, since
the target of NASBA is RNA instead of DNA, a positive
References

result indicates the presence of viable organisms (which


produce mRNA). The NASBA technique has already
been successfully applied for the detection of human
and animal pathogens, such as West Nile virus (Lanciotti
Slightly less sensitive than culture and Kerst, 2001), Chlamydia trachomatis (Morré et al.,
1996), Campylobacter spp. (Uyttendaele et al., 1994) and
number of organisms/µl was not
between copy number and the

Mycoplasma pneumoniae (Loens et al., 2002), and the


possible as the organisms are

No comparison with culture

identification of Mycobacterium spp. (Van der Vliet et


al., 1993).
c.f.u., colony-forming unit; DGGE, denaturing gradient gel electrophoresis; RFLP, restriction fragment length polymorphism.
Disadvantages

Strand displacement amplification (SDA)


unculturable

SDA is an isothermal process that uses primers, DNA


polymerase and a restriction enzyme to exponentially
amplify a unique nucleic acid sequence (Walker et al.,
Sensitivity and specificity similar to those of conventional

1992). SDA includes two phases, a target generation


phase and an exponential amplification phase. In target
nested PCR; detected 10 copies of the 16S rRNA gene

generation, dsDNA is heat-denatured (65°C, 10 minutes)


Detects 0.02 pg of N. risticii DNA, field-validated
simultaneously with a detection limit of less than

before primers with an overhang of a restriction enzyme


site anneal to the target. The exponential amplification
M. haemominutum’ in feline blood samples

phase is carried out in an isothermal environment


Validated with 153 horse blood samples.

(37°C). DNA polymerase, lacking 5–3 exonuclease


activity, extends the primers to generate products with a
restriction digestion site in one strand. Because a special
deoxyadenosine, 5-[α-thio]triphosphate (dATP[αS]), is
Highlights and Advantages

used to replace dATP, the restriction site of the newly


synthesized strand is inactivated. In the amplification
4 copies of templates

phase, the restriction enzyme nicks one strand of the


dsDNA products and allows polymerase to extend the
of the bacteria

DNA from the nicked site to displace the nicked strand.


After new dsDNAs are synthesized, the restriction
enzyme nicks them and the DNA extension starts again
(Fig. 5). Commercial SDA kits for detection of M. tuber-
culosis and Neisseria gonorrhoeae are available (BD
Diagnostic Systems, Sparks, MD, USA), and have been
shown to be robust, specific and sensitive (Akduman et
N. risticii 16S rRNA gene

N. risticii 16S rRNA gene

al., 2002; Barrett et al., 2002). SDA methods are simple,


TaqMan real-time PCR

take as little as 20 minutes to complete, and do not


require a thermocycler.
Molecular assays

nested PCR

16S rRNA analysis

Sequencing of the 16S ribosomal RNA (rRNA) gene has


Table 1. continued

been used as an important tool for phylogenetic analysis


and classification of bacteria for about two decades
Neorickettsia

(Lane et al., 1985). The 16S rRNA gene is well conserved


Organism

in all organisms and contains species-specific variable


regions allowing specific species identification. The 16S
rRNA is about 1500 base pairs long and consists of 10
Molecular genetic methods in the veterinary clinical bacteriology laboratory 83

Primer Polymerase
G-T-T-G-A-C
Target
dCTP, dGTP, dTTP, dATPaS

G-T-T-G-A-C
C-AsAsC-T-G
Nicked by restriction enzyme (Hinc II)

G-T-T G-A-C
C-AsAsC-T-G

Polymerization and strand displacement


G-
A-
G-T-T C
C-AsAsC-T-G

G-
A-
C

G-T-T G-A-C
G-T-T-G-A-C
C-AsAsC-T-G
C-AsAsC-T-G

Fig. 5. Principle of strand displacement amplification (SDA). SDA primers that include a HincII site overhang anneal to DNA
targets. DNA replication using dCTP, dGTP, dTTP and dATP[αS] produces a double-stranded DNA with a HincII site in one
strand. HincII nicks the unprotected strand. DNA polymerase extends the 3 end at the nick and displaces the downstream
fragment. The polymerase and displacement regenerate a double-stranded DNA with a HincII site in one strand. Nicking,
polymerization and displacement steps cycle continuously and amplify the DNA target exponentially in an isothermal
environment.

widely conserved regions and 10 divergent regions. The There are some problems with the use of 16S rRNA
divergent regions accumulate mutations with a slow but gene sequencing for bacterial identification. Some closely
constant rate of about 1% per 50 million years and thus related species of bacteria, such as Shigella spp. and E.
can be used for bacterial identification by sequence coli, are difficult to classify by 16S rRNA analysis
comparison (Trotha et al., 2001). It has been proposed (Christensen et al., 1998). For these bacteria, the gyrB
that strains with rRNA similarity less then 97% belong to sequence analysis has been suggested (Fukushima et al.,
different species. 2002). Other housekeeping genes, such as the RNA poly-
Sequencing of 16S rRNA has been used to identify merase -subunit gene (rpoB), have also been
slow-growing, unusual and fastidious bacteria, and bac- investigated as alternatives for speciation of some bacte-
teria that are poorly identified by conventional methods. ria, such as Anaplasma spp., Bartonella spp., Ehrlichia
For example, Bacillus angiomatosis and Tropheryma spp., Mycoplasma spp. and Neorickettsia spp. (Kim et al.,
whippelii are non-culturable bacteria that were first iden- 2003; La Scola et al., 2003; Taillardat-Bisch et al., 2003). In
tified by a 16S rRNA gene sequencing method (Relman addition, some foreign DNA sequences, called intervening
et al., 1990, 1992). 16S rRNA sequencing kits are avail- sequences (IVS), of ~140 base pairs, may exist in 16S
able (PE Applied Biosystems, Forster City, CA, USA) and rRNA genes (Linton et al., 1994; Etoh et al., 1998).
are accurate and rapid for the identification of Therefore, possible IVS should be identified if a 16S rRNA
Mycobacterium spp. (Patel et al., 2000) and clinical gene sequence is larger than the usual size (~1.5 kp).
coryneform bacterial isolates (Tang et al., 2000). This
method has also been used to identify a novel mycobac-
terial species as an agent of canine leproid granuloma DNA microarrays
(Hughes et al., 2000). Cai et al. (2003) reported that 16S
rRNA sequencing was readily applicable to difficult-to- A DNA microarray is essentially a miniaturized form of
identify bacteria in a veterinary clinical bacteriology multiple dot blots. It is composed of DNA probes bound
laboratory, and provided details of the costs and choice to a solid substrate, such as glass. During hybridization,
of primers. a target DNA sample covers the complete array area and
84 H. Y. Cai et al.

hybridizes with the probes that have a complementary bacteria, including newly reclassified members of the
sequence. Hybridized targets are detected using a order Rickettsiaceae (genera Anaplasma, Ehrlichia,
reporter system. Either PCR products or synthesized Neorickettsia and Rickettsia) (Dumler et al., 2001). This
oligonucleotides can be used as probes and spotted on order contains a series of emerging or newly recognized
the supporting matrix. PCR product probes are less pathogens, whose epidemiology is being elucidated
expensive and simpler to generate when limited number largely through molecular diagnostic approaches.
of primers are needed to generate the probes. However, Similarly, as many other bacteria, such as Bartonella,
their lengths make it difficult to discriminate between Borrelia and Mycoplasma, are reclassified based on
minor sequence mismatches. An oligonucleotide probe molecular findings (e.g. 16S rRNA gene sequences)
array is more expensive to construct but can be more (Brenner et al., 1993; Birtles et al., 1995; Neimark et al.,
specific because of the short length of the probe. To 2001), there will be increased reliance on molecular
detect a target DNA or RNA, it usually requires PCR or methods for diagnostic and identification purposes.
reverse transcription–PCR to amplify the targets so as to
achieve adequate sensitivity. Single or multiple reporter
molecules (e.g. biotin and Cy-3) can be incorporated Escherichia coli
directly via 5 primer modifications, nick translation or
chemical incorporation. After hybridization of targets Escherichia coli is part of the normal intestinal
with the microarray, a fluorescence scanner can be used microflora, but small subsets (pathotypes) of the species
to scan a solid (e.g. glass) array, whereas a phosphoim- are capable of causing enteric and extra-intestinal dis-
ager can be used for isotopic reporters and eases. Table 2 lists the common types of pathogenic E.
membrane-based arrays. coli that cause disease in animals. The implication of
Microarrays have been widely used to study bacteria, extraintestinal E. coli in disease is straightforward as it is
but their application in clinical microbiology is hindered typically based on recovery of the organisms from nor-
by low sensitivity when used directly on cultures or mally sterile sites. Pathogenic enteric isolates, however,
samples. PCR is required to amplify the target DNA must be differentiated from normal flora isolates. Gene-
before it can be detected by a DNA array. Therefore a based methods are particularly valuable as they
DNA microarray does not have an advantage over a reg- circumvent the problem of expression of virulence fac-
ular PCR in detecting individual bacteria or individual tors in vitro and they may rapidly provide information
virulence genes. In order to detect multiple bacteria or on the complement of virulence genes present in the
multiple virulence genes, it requires multiplex PCR to isolate.
amplify multiple targets, but multiplex PCR is difficult to Enterotoxigenic E. coli (ETEC) are among the most
optimize and may include a maximum of about 10 common of the pathogenic E. coli implicated in enteric
sets of primers. To circumvent this obstacle, the 16S diseases of animals and the vast majority of samples
rRNA gene has been used as a target, which can be submitted for diagnosis of E. coli enteric disease are
amplified using a set of universal primers and feces or intestine from calves or pigs suspected to be
hybridized with a DNA microarray consisting of species- suffering from ETEC-induced diarrhea. Identification of
specific 16S rRNA gene probes (Zimmermann et al., ETEC is based on detection of the colonization fimbriae
2003). It is likely that microarrays will become common and enterotoxin(s) or the genes that encode them (Table
in clinical microbiology in the future. In particular, they 2). Colonization fimbriae may be detected by immuno-
could be used to identify organisms with complex dis- logical tests, such as agglutination, latex particle
ease-causing abilities, for example by detecting agglutination or ELISA. However, certain fimbriae (such
virulence genes to identify the pathotypes of E. coli as F18) are not readily expressed in vitro, and agglutina-
(Bekal et al., 2003); or they could be used to screen for tion tests are prone to yield false-positive reactions.
multiple antibiotic resistance genes. There are no practical tests for the routine detection of
the enterotoxins in the diagnostic laboratory and sole
reliance on detection of the colonization fimbriae will
Published molecular genetic methods for bacterial falsely identify enterotoxin-negative, fimbriae-positive
pathogens isolates as ETEC.
In the last decade, DNA probes and PCR methods
Table 1 lists the most recent applications of molecular have been developed for the detection of the genes
methods for the diagnosis of specific important bacterial coding for ETEC fimbriae and enterotoxins. The DNA
infections of animals, and for other animal pathogens probes are highly suited to investigations in which large
that are slow or difficult to isolate in the clinical bacteri- numbers of isolates are examined simultaneously. The
ology laboratory. As an example, the molecular PCR is well suited to routine application in the diagnos-
identification of Escherichia coli will be discussed in tic laboratory. A common approach is to use a multiplex
detail. There is clearly tremendous scope for the PCR that targets the genes for enterotoxins and fimbriae.
application of molecular methods to hard-to-isolate We have accumulated some experience with a multiplex
Molecular genetic methods in the veterinary clinical bacteriology laboratory 85

Table 2. Genes for toxins and adhesins that are targeted for identification of E. coli that cause enteric diseases in animals

Animal Pathotype of E. coli Disease Target virulence genes


Pigs
Neonatal Enterotoxigenic E. coli (ETEC) Diarrhea eltA (LT), estA (STa), estB (STb), faeG (F4),
fanA (F5), fim41a (F41), fasA (987P), (AIDA-1)
Weaned ETEC Diarrhea eltA, estA, estB, faeG, fedA (F18)
Shiga toxin-producing E. coli (STEC) Edema disease stx2e (Shiga toxin 2e), fedA
Enteropathogenic E. coli (EPEC) Diarrhea eae (intimin)
Calves and lambs ETEC Diarrhea estA, fanA, fim41a
EPEC Diarrhea eae
STEC Diarrhea/dysentery eae, stx1, stx2
Dogs STEC O157:H7 Bloody diarrhea, rfbE (O157), fliC (H7), eae
skin lesions, HUS stx1, stx2, ehly (EHEC hemolysin)
Rabbits EPEC Diarrhea eae
LT, heat-labile enterotoxin; STa and STb, heat-stable enterotoxins a and b respectively; HUS, hemolytic uremic syndrome.

PCR which targets nine genes, namely genes that icance. Preliminary identification is based on the aggluti-
encode the LT, STa and STb enterotoxins, and the F4 nation of sorbitol-negative suspect colonies in O157
(K88), F5 (K99), F6 (987P), F18 and F41 fimbriae, as antiserum, but false positives occur. Confirmation and
well as the Stx2e Shiga toxin, which is implicated in initial characterization are based on the detection of rfbE
edema disease of pigs (Bosworth and Casey, 1997). We and fliC genes for the O and H antigens respectively,
have found that results are considerably improved with the eae gene, and the stx1 and stx2 genes. The plasmid-
the use of the Qiagen Multiplex Master Mix, which leads encoded ehly gene is sometimes also detected. Several
to few or no non-specific bands. It is not necessary to multiplex PCR protocols have been reported that target
target all nine genes in order to satisfactorily identify these genes (Paton and Paton, 1998; Hu et al., 1999;
ETEC. For example, the PCR may be modified to target Botteldoorn et al., 2003).
only three genes (genes for STa, F5 and F41) for bovine Recently, real-time PCR assays have been developed to
isolates (Franck et al., 1998). detect pathogenic E. coli, mostly using molecular beacon
Edema disease of pigs is usually so characteristic in its probes on TaqMan or LightCycler platforms. Some assays
clinical appearance that laboratory tests that identify the have been applied to animal samples. TaqMan real-time
isolates as Shiga toxin-producing E. coli (STEC) are usu- assays were described for the detection of STEC in milk
ally not necessary. However, the multiplex PCR by detecting stx1 and stx2 genes separately (Burk et al.,
described above can identify edema disease isolates by 2002), by detecting the stx2 gene (McKillip and Drake,
its detection of genes for Stx2e and F18. Many of the 2000), or by detecting the rfbE gene specific for O157
edema disease isolates will also have genes for entero- (Fortin et al., 2001). Sharma (2002) reported two sets of
toxins, and are called ‘STEC/ETEC’. Bosworth et al. TaqMan real-time PCR assays for the simultaneous detec-
(1998) described a single-strand confirmation polymor- tion and quantitation of EHEC in beef and bovine feces
phism assay that differentiates F18ab from F18ac. This by detection of a portion of the eae gene specific to the
differentiation is not necessary for the identification of EHEC O26, O111 and O157 serotypes, and by detection
edema disease strains of E. coli and the test is not suit- of the stx1 and stx2 genes. Amplification and specific
able for application in a clinical diagnostic laboratory. detection of one or two genes of E. coli can be com-
No molecular methods have been developed for the pleted within 60 minutes (Reischl et al., 2002).
specific identification of enteropathogenic E. coli (EPEC) Ge and colleagues (2002) identified enterohemor-
in pigs, cattle or rabbits. Considerable dependence is rhagic E. coli (EHEC) by targeting the EHEC-hlyA gene
placed on the detection of attaching and effacing lesions and amplifying probe–target hybrids by SDA. The assay
in the intestine of affected animals. The PCR may be identified EHEC in 35 minutes with a detection limit of
used to identify EPEC isolates from these animals by the 4.3 bacteria and specificity of 100%. Although SDA is rel-
presence of the eae (E. coli attaching and effacing) gene atively costly and complex to prepare and is susceptible
and the absence of stx genes. STEC, especially those of to minor variations in experimental condition, once opti-
serogroups 5, 26, 111 and 118, have been implicated in mized and commercialized it will be simpler than PCR to
diarrhea and/or dysentery in calves. A multiplex PCR perform, because it uses an isothermal reaction and the
that detects eae, stx1, stx2 and ehly (enterohemorrhagic readout conditions require simple instruments. Most
E. coli hemolysin) may be used for this type of STEC. recently, DNA microarrays have been described for
Typically, these isolates are eae+ and stx1+. detecting and genotyping E. coli (Call et al., 2001;
E. coli O157:H7 is implicated in disease in dogs (albeit Chandler et al., 2001; Bekal et al., 2003).
rarely) and its presence in cattle has public health signif-
86 H. Y. Cai et al.

Detection of antimicrobial resistance genes New technology, including real-time PCR and
microarrays, has been applied to the detection of some
The molecular detection of antimicrobial resistance in bacterial resistance genes (Killgore et al., 2000; Torres et
human medicine and its impact on clinical practice has al., 2000; Westin et al., 2001). DNA arrays and chips are
been reviewed in detail (Bergeron, 2000; Fluit et al., technologies which allow the mass screening of genes.
2001). The simultaneous rapid genotypic identification of If all known resistance genes could be represented on a
bacteria and their antibiotic resistance genes may have a single chip or array, this would provide a complete test.
major impact on the treatment of infectious disease while However, these techniques are still very expensive and
contributing to better control of antimicrobial resistance. new resistance genes will continually emerge, so that
In addition, molecular detection of resistance also offers current genetic tests will miss the new mechanisms.
the potential identification of resistance genes that are Furthermore, new antimicrobial drugs will also be regu-
expressed in vivo but not in vitro (Fluit et al., 2001). larly developed, as illustrated by the continuing
However, phenotypic assays are still the method of appearance of newer quinolones with increased
choice for most resistance determinations because of the potency. Frequent investigation will be needed to
simplicity of the assays and the large numbers of antibi- update genetic tests so as to detect resistance associated
otic resistance genes that may exist for a single antibiotic. with these new drugs and with genetic changes.
Nucleic acid-based assays have the potential to Another problem with the detection of resistance genes
increase the rapidity and accuracy of resistance testing. is that their presence does not lead automatically to treat-
For example, molecular detection of the methicillin ment failure since their expression may be low in vivo.
resistance-encoding mecA gene in staphylococci and This is particularly true for ß-lactamase production by
rifampin resistance in M. tuberculosis has been widely enterobacteria, in which the resistance depends on the
used in human medicine (Fluit et al., 2001). Because of degree of expression (Fluit et al., 2001). The extreme sen-
the emergence of multidrug-resistant microorganisms in sitivity of molecular-based detection tests for resistance
veterinary medicine, such as multidrug-resistant genes also brings a major problem in detecting resistance
Salmonella Newport, rapid testing by molecular meth- genes for non-sterile sites, polymicrobial infections, com-
ods that monitor specific resistance genes may become a plex microflora and contaminated clinical specimens.
useful tool for veterinary teaching hospitals and veteri- In summary, the DNA-based detection of antibiotic
nary clinics. This approach could also be used at the resistant bacteria can potentially be a rapid and sensitive
farm level for the surveillance of antimicrobial resistance alternative to the current phenotypic detection method.
genes in food-producing animals. Molecular methods However, extensive comparison between the two meth-
may also be useful in the rapid detection of resistance ods is required to determine the sensitivity and
from normally sterile clinical specimens, such as blood, specificity before the DNA-based methods can be used
cerebrospinal fluid and joint fluid (Bergeron, 2000). for clinical diagnosis. It is likely that phenotypic meas-
There are limitations in the use of molecular assays urement of antimicrobial susceptibility and resistance
for the detection of antibiotic resistance. For example, will continue to be an important function of clinical bac-
several dozen plasmid-mediated ß-lactamases cause teriology laboratories in the future and an area where
resistance by inactivating ß-lactam antibiotics, and a molecular methods may find less application.
large number of aminoglycoside resistance genes exist
in Gram-negative microorganisms (Fluit et al., 2001).
Problems associated with having many genes that PCR for detection of bovine mastitis pathogens
encode resistance to a single antibiotic are illustrated in
studies by Yang et al. (2001), who described a rapid Early identification of the agents that cause bovine mas-
assay for the detection of multiple antibiotic-resistant titis is important as it facilitates appropriate treatment,
Salmonella Enteritidis and Salmonella Typhimurium iso- minimizes the development of chronic disease, and con-
lates from animals. The assay is a multiplex PCR that tributes to the control of infection in the herd. Bovine
amplifies antibiotic resistance genes and the Salmonella mastitis can be classified into contagious and environ-
spp.-specific gene SipB/C. When this multiplex PCR was mental mastitis. Contagious mastitis is caused by a small
later compared with phenotypic assays, the drug resist- number of pathogens that include S. aureus,
ance patterns did not match well with PCR amplification Streptococcus agalactiae, Mycoplasma bovis and
types (Yang et al., 2002). For example, two ampicillin- Corynebacterium bovis, whereas environmental mastitis
resistant Salmonella isolates were negative for both is caused by a wide range of pathogens that exist in the
blaPSE-1 and blaTEM. The ampicillin resistance may have environment and contaminate the tips of the teats
been conferred by other ß-lactamase genes, such as (Quinn et al., 2002).
blaSHV and oxa-2, which were not investigated in the Routine bacteriological identification of mastitis
experiments. The large number of different resistance pathogens is usually performed by traditional culture
genes for some antibiotics makes full coverage by followed by biochemical tests on bacterial isolates. The
probes or PCR very difficult. advantages of the culture method are that viable
Molecular genetic methods in the veterinary clinical bacteriology laboratory 87

causative bacteria can be identified and antimicrobial for Streptococcus agalactiae and S. dysgalactiae. More
susceptibility can be performed, providing guidance on recently, Phuektes et al. (2003) have used a multiplex
the selection of antimicrobials for treatment. However, PCR to detect the same microorganisms in bulk tank milk
there are disadvantages. The sensitivity of culture may samples, in which correlations with bulk milk somatic cell
not be sufficient to detect intermittent shedders with low counts (BMCC), total bacterial counts and thermoduric
and high shedding cycles during lactation, subclinically bacterial counts were evaluated. There was no relation-
infected glands with low numbers of bacteria, or bacte- ship in their study between the presence of S. aureus, S.
ria inhibited by residual therapeutic antimicrobials or agalactiae or S. uberis and BMCC, total bacterial counts
leukocytes. Environmental contaminants and intracister- or thermoduric bacteria counts, but the presence of S.
nal microorganisms can also represent a major problem agalactiae was associated with high BMCC and total bac-
in the interpretation of culture results. Another problem terial counts. Their results have shown that regular
is that certain bacteria, such as Streptococcus uberis and analysis of bulk milk using this multiplex PCR assay may
S. parauberis (S. uberis type II) cannot be distinguished be a useful tool for monitoring herd status for S. agalac-
by biochemical tests (Jayarao et al., 1991). Moreover, tiae, but it is of less value for monitoring of S. aureus, S.
bacterial culture and identification is time consuming dysgalactiae and S. uberis.
and requires more than 48 hours to complete. Riffon et al. (2001) have developed molecular probes
Because of the limitations of conventional culture, sim- reacting in PCR with bacterial DNA from bovine milk,
plex and multiplex PCR protocols have been developed providing direct and rapid detection of E. coli,
for identification of the various mastitis pathogens Staphylococcus aureus, Streptococcus agalactiae,
(Jayarao et al., 1991; Forsman et al., 1997; Ghadersohi et Streptococcus dysgalactiae, Streptococcus parauberis and
al., 1997; Khan et al., 1998; Hirose et al., 2001; Kim et al., Streptococcus uberis. The two sets of primers appeared
2001; Pinnow et al., 2001; Riffon et al., 2001; Daly et al., to discriminate the close phylogenetic species
2002; Meiri-Bendek et al., 2002; Phuektes et al., 2001, Streptococcus agalactiae and Streptococcus dysgalactiae,
2003). These PCR methods offer the option of identifica- and Streptococcus uberis and Streptococcus parauberis.
tion of bacteria within hours. PCR can also improve the The primers for E. coli, Staphylococcus aureus,
level of detection because of its ability to detect low Streptococcus parauberis and Streptococcus uberis were
numbers of organisms. Mastitis pathogens could proba- designed on the basis of 23S rRNA, and those for
bly be detected in carrier animals or at earlier stages of Streptococcus agalactiae and Streptococcus dysgalactiae
infection in clinical cases. PCR also has the potential to were based on 16S rRNA sequence.
be extremely specific and to discriminate between Several publications have described PCR methods for
closely related microorganisms, such as S. parauberis and the direct detection of Mycoplasma including M. alka-
S. uberis. The disadvantages of PCR methods include lescens, M. bovigenitalium, M. bovirhinis and M. bovis
their very low threshold of detection, since minor con- (Forsman et al., 1997; Hirose et al., 2001; Kim et al.,
taminants and non-viable bacteria in milk samples could 2001; Pinnow et al., 2001). The threshold of detection
lead to misdiagnosis. In addition, PCR does not provide for these PCR tests ranged from 1 to 1500 colony-form-
information on antimicrobial sensitivities. ing units/ml of milk.
To our knowledge, real-time PCR has not yet been In summary, the area of molecular genetic tests for
described for the identification of mastitis pathogens. the identification of mastitis pathogens is underdevel-
These multiplex PCR protocols could be adapted to a real- oped. Because of the enormous economic importance of
time platform PCR in order to increase the ability to detect mastitis to the dairy industry, this is an area in which
low numbers of organisms as well as turn-around time. In rapid, specific and inexpensive tests have the potential
addition, the real-time PCR offers quantitative data, which to be applied extensively in the veterinary clinical
might help in addressing the issue of minor contaminants microbiology laboratory.
found in milk samples. While quantitative PCR has an
obvious place in distinguishing contaminants from gen-
uine infection, further work is needed to define what Commercially available tests
would be regarded as genuinely positive PCR findings.
A species-specific multiplex PCR based on detection of Many laboratories use in-house molecular methods but
the 16S to 23S rRNA spacer region was developed and some tests are available commercially as kits, which
compared with traditional culture by Phuektes et al. have been evaluated analytically or diagnostically to
(2001) for four mastitis pathogens, Staphylococcus aureus, varying degrees. Compared with in-house methods, the
Streptococcus agalactiae, Streptococcus dysgalactiae and commercial kits usually are easier to use because the
Streptococcus uberis. This multiplex PCR was significantly reagents are premixed, and results may be more consis-
more effective than culture for the detection of low num- tent between laboratories as the same reagents are used
bers of Staphylococcus aureus and Streptococcus uberis. in a standardized way. However the price is usually
However, there were no significant differences in detec- higher. Current commercially available molecular test
tion of low numbers of bacteria between PCR and culture kits are listed in Table 3.
88

Table 3. Commercially available kits and systems for molecular tests

Pathogen Kits Test format Company


Actinobacillus pleuropneumoniae Adiavet® APP PCR tests PCR Adiagène, France
Bacillus anthracis Bacillus Anthracis Detection kit LightCycler real-time PCR Roche Diagnostics, Basel, Switzerland
Borrelia burgdorferi and B. afzelli RealArtTMBorrelia PCR kit PCR Artus Biotech USA, USA
BOR B. burgdorferi and B. afzelli detection kit Nested PCR AB Analitica, Italy
Brachyspira hyodysentariae and Adiavet® Brachy PCR test (differentiating PCR Adiagène, France
B. pilosicoli B. hyodysenteriae, B. pilosicoli, B. innocens
and B. intermedia)
Escherichia coli O157:H7 BAX® System PCR assay for screening Automated PCR system DuPont Qualicon, Wilmington, DE, USA
E. coli O157:H7
TaqMan® pathogenic E. coli (0157:H7) TaqMan real-time PCR Applied Biosystem, Foster City, CA, USA
detection kits
Helicobacter pylori HP H. pylori detection kit Nested PCR AB Analitica, Italy
Lawsonia intracellularis Adiavet® law PCR test kit PCR Adiagène, France
Mycobacterium paratuberculosis paratub test kit
Adiavet® PCR Adiagène, France
IDEXX M.pt DNA test kit PCR and dot or Southern blot Idexx, Westbrook, ME, USA
Mycoplasma Adiavet® myco av PCR test kit (for PCR Adiagène, France
M. gallisepticum, M. synoviae and
M. meleagridis)
Adiavet® M.HYOP PCR test (for PCR Adiagène, France
M. hyopneumonaie)
Salmonella RealArtTM Salmonella PCR kit (for all LightCycler real-time PCR or PCR Artus Biotech USA,USA
Salmonella spp.)
BAX® System PCR assay for screening PCR automatic system DuPont Qualicon, Wilmington, DE, USA
Salmonella
TaqMan® Salmonella gold detection kit TaqMan real-time PCR Applied Biosystem, Foster City, CA, USA
Bacteria genotyping system RiboPrinter® microbial characterization Automatic ribotyping system DuPont Qualicon, Wilmington, DE, USA
system (for fingerprinting of any bacteria)
Bacterial 16S and 23S rRNA MicroSeq® microbial identification PCR and DNA sequencing Applied Biosystem, Foster City, CA, USA
identification system (kit)
H. Y. Cai et al.
Molecular genetic methods in the veterinary clinical bacteriology laboratory 89

Concluding remarks to accredit and standardize all molecular methods used


in veterinary bacteriology diagnostic laboratories and
In the last 5 years, numerous molecular methods have provide standard controls, especially for quantitation
been published for the detection and characterization of purposes.
bacteria in the field of veterinary medicine. The PCR has In summary, the door to molecular diagnosis has just
been the most commonly used technology. Although opened. Many molecular methods are waiting to be put
not currently used routinely for clinical veterinary diag- into practical use for more efficient, rapid and sensitive
nosis, new technologies such as liquid-phase laboratory diagnosis of bacterial infections in animals.
hybridization, real-time PCR, pathogen load determina-
tion and DNA/protein microarray have been described
and have many possible applications in the clinical bac- References
teriology laboratory because of their sensitivity and
efficiency. Akduman D, Ehret JM, Messina K, Ragsdale S and Judson FN
(2002). Evaluation of a strand displacement amplification
Veterinary molecular diagnosis has not yet been
assay (BD ProbeTec-SDA) for detection of Neisseria gonor-
applied in the way that it could be in clinical bacteriol- rhoeae in urine specimens. Journal of Clinical
ogy laboratories. Only a limited number of detection Microbiology 40: 281–283.
assays have been developed into commercial diagnostic Barrett A, Magee JG and Freeman R (2002). An evaluation of
kits, and only a few molecular tests are offered by vet- the BD ProbeTec ET system for the direct detection of
Mycobacterium tuberculosis in respiratory samples.
erinary diagnostic laboratories. Most available kits or
Journal of Medical Microbiology 51: 895–898.
laboratory services are for the detection of pathogens or Bekal S, Brousseau R, Masson L, Prefontaine G, Fairbrother J
toxins, and few are for molecular typing. The molecular and Harel J (2003). Rapid identification of Escherichia coli
detection of antimicrobial resistance genes is still a pathotypes by virulence gene detection with DNA
largely unexplored area in veterinary clinical diagnosis. microarrays. Journal of Clinical Microbiology 41:
2113–2125.
There are several obstacles to the application of molecu-
Bekker CP, de Vos S, Taoufik A, Sparagano OA and Jongejan F
lar diagnosis in veterinary bacteriology laboratories. (2002). Simultaneous detection of Anaplasma and
First, most current accredited tests are based on tradi- Ehrlichia species in ruminants and detection of Ehrlichia
tional methods. Without updating of these accreditation ruminantium in Amblyomma variegatum ticks by reverse
standards, the molecular tests are not acceptable to line blot hybridization. Veterinary Microbiology 89:
223–238.
accreditation authorities. Secondly, little information is
Bergeron MG (2000). Genetic tools for the simultaneous identi-
available on method validation. Although a lot of effort fication of bacterial species and their antibiotic resistance
has been devoted to the development of molecular tests, genes: impact on clinical practice. International Journal of
most published research stops at analytical validation Antimicrobial Agents 16: 1–3.
and does not take the additional steps that are required Birtles RJ, Harrison TG, Saunders NA and Molyneux DH (1995).
Proposals to unify the genera Grahamella and Bartonella,
for field validation. The fact that these steps are time-
with descriptions of Bartonella talpae comb. nov.,
consuming and costly is probably responsible for this. Bartonella peromysci comb. nov., and three new species,
An additional problem in the adoption of molecular Bartonella grahamii sp. nov., Bartonella taylorii sp. nov.,
methods is that of the determination of gold standards and Bartonella doshiae sp. nov. International Journal of
against which the sensitivity and specificity of molecular Systematic Bacteriology 45: 1–8.
Bockstahler LE, Li Z, Nguyen NY, Van Houten KA, Brennan MJ,
tests can be measured. For example, it is highly likely
Langone JJ and Morris SL (2002). Peptide nucleic acid
that these may turn out in some cases to be far more probe detection of mutations in Mycobacterium tuberculo-
sensitive than isolation or immunological approaches, sis genes associated with drug resistance. Biotechniques
leading to a tendency to discount the reliability of the 32: 508–510, 512, 514.
newer assays. If these tests are in fact more reliable than Bosworth BT, Dean-Nystrom EA, Casey TA and Neibergs HL
(1998). Differentiation of F18ab+ from F18ac+ Escherichia
conventional tests, they may lead to changed under-
coli by single-strand conformational polymorphism analy-
standing of the epidemiology of some bacterial sis of the major fimbrial subunit gene (fedA). Clinical and
infections of animals, but gaining such acceptance will Diagnostic Laboratory Immunology 5: 299–302.
require careful documentation. However, more robust Bosworth BT and Casey TA (1997). Identification of toxin and
and reliable methods need to be developed and vali- pilus genes in porcine E. coli using multiple primer pairs
via PCR. 97th General Meeting. American Society for
dated in order to reduce the cost of testing and to
Microbiology. B509.
facilitate application in a diagnostic laboratory. Botteldoorn N, Heyndrick M, Rijpens N and Herman L (2003).
Proficiency testing is also urgently needed in molecular Detection and characterization of verotoxigenic
veterinary clinical diagnostic laboratories, as it remains Escherichia coli by a VTEC/EHEC multiplex PCR in
largely unregulated. Most available molecular test labo- porcine faeces and pig carcass swabs. Research in
Microbiology 154: 97–104.
ratory services use procedures developed in-house. It
Brenner DJ, O’Connor SP, Winkler HH and Steigerwalt AG
would be useful for an organization such as the (1993). Proposals to unify the genera Bartonella and
American Association of Veterinary Laboratory Rochalimaea, with descriptions of Bartonella quintana
Diagnosticians or the Office International des Epizooties comb. nov., Bartonella vinsonii comb. nov., Bartonella
90 H. Y. Cai et al.

henselae comb. nov., and Bartonella elizabethae comb. Exner MM and Lewinski MA (2003). Isolation and detection of
nov., and to remove the family Bartonellaceae from the Borrelia burgdorferi DNA from cerebral spinal fluid, syn-
order Rickettsiales. International Journal of Systematic ovial fluid, blood, urine, and ticks using the Roche MagNA
Bacteriology 43: 777–786. Pure system and real-time PCR. Diagnostic Microbiology
Bricker BJ, Ewalt DR, Olsen SC and Jensen AE (2003). and Infectious Disease 46: 235–240.
Evaluation of the Brucella abortus species-specific poly- Fang Y, Wu WH, Pepper JL, Larsen JL, Marras SA, Nelson EA,
merase chain reaction assay, an improved version of the Epperson WB and Christopher-Hennings J (2002).
Brucella AMOS polymerase chain reaction assay for cattle. Comparison of real-time, quantitative PCR with molecular
Journal of Veterinary Diagnostic Investigation 15: beacons to nested PCR and culture methods for detection
374–378. of Mycobacterium avium subsp. paratuberculosis in
Burk C, Braumiller IG, Becker H and Martlbauer E (2002). bovine fecal samples. Journal of Clinical Microbiology 40:
Nuclease fluorescence assay for the detection of verotoxin 287–291.
genes in raw milk. Letters in Applied Microbiology 35: Fluit AC, Visser MR and Schmitz FJ (2001). Molecular detection
153–156. of antimicrobial resistance. Clinical Microbiology Reviews
Cai HY, Archambault M and Prescott JF (2003). 16S ribosomal 14: 836–871.
DNA sequence-based identification of veterinary clinical Forsman P, Tilsala-Timisjarvi A and Alatossava T (1997).
bacteria difficult to identify by conventional phenotypic Identification of staphylococcal and streptococcal causes
methods. Journal of Veterinary Diagnostic Investigation of bovine mastitis using 16S-23S rRNA spacer regions.
15: 465–469. Microbiology 143: 3491–3500.
Call DR, Brockman FJ and Chandler DP (2001). Detecting and Fortin NY, Mulchandani A and Chen W (2001). Use of real-time
genotyping Escherichia coli O157: H7 using multiplexed polymerase chain reaction and molecular beacons for the
PCR and nucleic acid microarrays. International Journal detection of Escherichia coli O157: H7. Analytical
of Food Microbiology 67: 71–80. Biochemistry 289: 281–288.
Carli KT and Eyigor A (2003). Real-time polymerase chain reac- Franck SM, Bosworth BT and Moon HW (1998). Multiplex PCR
tion for Mycoplasma gallisepticum in chicken trachea. for enterotoxigenic, attaching and effacing, and Shiga
Avian Diseases 47: 712–717. toxin-producing Escherichia coli strains from calves.
Chandler DP, Brown J, Call DR, Wunschel S, Grate JW, Holman Journal of Clinical Microbiology 36: 1795–1797.
DA, Olson L, Stottlemyre MS and Bruckner-Lea CJ (2001). Fukushima M, Kakinuma K and Kawaguchi R (2002).
Automated immunomagnetic separation and microarray Phylogenetic analysis of Salmonella, Shigella, and
detection of E. coli O157: H7 from poultry carcass rinse. Escherichia coli strains on the basis of the gyrB gene
International Journal of Food Microbiology 70: 143–154. sequence. Journal of Clinical Microbiology 40: 2779–2785.
Christensen H, Nordentoft S and Olsen JE (1998). Phylogenetic Garrido JM, Cortabarria N, Oguiza JA, Aduriz G and Juste RA
relationships of Salmonella based on rRNA sequences. (2000). Use of a PCR method on fecal samples for diagno-
International Journal of Systematic Bacteriology 48: sis of sheep paratuberculosis. Veterinary Microbiology 77:
605–610. 379–386.
Daly P, Collier T and Doyle S (2002). PCR-ELISA detection of Ge B, Larkin C, Ahn S, Jolley M, Nasir M, Meng J and Hall RH
Escherichia coli in milk. Letters in Applied Microbiology 34: (2002). Identification of Escherichia coli O157: H7 and
222–226. other enterohemorrhagic serotypes by EHEC-hlyA target-
DeGraves FJ, Gao D, Hehnen HR, Schlapp T and Kaltenboeck ing, strand displacement amplification, and fluorescence
B (2003). Quantitative detection of Chlamydia psittaci and polarization. Molecular and Cellular Probes 16: 85–92.
C. pecorum by high-sensitivity real-time PCR reveals high Ghadersohi AR, Coelen J and Hirst RG (1997). Development of
prevalence of vaginal infection in cattle. Journal of a specific DNA probe and PCR for the detection of
Clinical Microbiology 41: 1726–1729. Mycoplasma bovis. Veterinary Microbiology 56: 87–98.
Demidov VV (2003). PNA and LNA throw light on DNA. Trends Gravekamp C, Van de Kemp H, Franzen M, Carrington D,
in Biotechnology 21: 4–7. Schoone GJ, Van Eys GJ, Everard CO, Hartskeerl RA and
Dumler JS, Barbet AF, Bekker CP, Dasch GA, Palmer GH, Ray Terpstra WJ (1993). Detection of seven species of patho-
SC, Rikihisa Y, Rurangirwa FR (2001). Reorganization of genic leptospires by PCR using two sets of primers.
genera in the families Rickettsiaceae and Anaplasmataceae Journal of General Microbiology 139: 1691–1700.
in the order Rickettsiales: unification of some species of Harkin KR, Roshto YM, Sullivan JT (2003). Clinical application
Ehrlichia with Anaplasma, Cowdria with Ehrlichia and of a polymerase chain reaction assay for diagnosis of lep-
Ehrlichia with Neorickettsia, descriptions of six new tospirosis in dogs. American Journal of Veterinary
species combinations and designation of Ehrlichia equi Research 222: 1224–1229.
and ‘HGE agent’ as subjective synonyms of Ehrlichia Heinemann MB, Garcia JF, Nunes CM, Gregori F, Higa ZM,
phagocytophila. International Journal of Systematic and Vasconcellos SA and Richtzenhain LJ (2000). Detection
Evolutionary Microbiology 51: 2145–65. and differentiation of Leptospira spp. serovars in bovine
Englund S, Bolske G, Ballagi-Pordany A and Johansson KE semen by polymerase chain reaction and restriction frag-
(2001). Detection of Mycobacterium avium subsp. paratu- ment length polymorphism. Veterinary Microbiology 73:
berculosis in tissue samples by single, fluorescent and 261–267.
nested PCR based on the IS900 gene. Veterinary Hirose K, Kawasaki Y, Kotani K, Tanaka A, Abiko K and
Microbiology 81: 257–271. Ogawa H (2001). Detection of mycoplasma in mastitic
Englund S, Bolske G and Johansson KE (2002). An IS900-like milk by PCR analysis and culture method. Journal of
sequence found in a Mycobacterium sp. other than Veterinary Medical Science 63: 691–693.
Mycobacterium avium subsp. paratuberculosis. FEMS Hu Y, Zhang Q and Meitzler JC (1999). Rapid and sensitive
Microbiology Letters 209: 267–271. detection of Escherichia coli O157: H7 in bovine faeces by
Etoh Y, Yamamoto A and Goto N (1998). Intervening a multiplex PCR. Journal of Applied Microbiology 87:
sequences in 16S rRNA genes of Campylobacter sp.: diver- 867–876.
sity of nucleotide sequences and uniformity of location. Hughes MS, James G, Ball N, Scally M, Malik R, Wigney DI,
Microbiology and Immunology 42: 241–243. Martin P, Chen S, Mitchell D and Love DN (2000).
Molecular genetic methods in the veterinary clinical bacteriology laboratory 91

Identification by 16S rRNA gene analyses of a potential Linton D, Dewhirst FE, Clewley JP, Owen RJ, Burnens AP and
novel mycobacterial species as an etiological agent of Stanley J (1994). Two types of 16S rRNA gene are found
canine leptroi granuloma syndrome. Journal of Clinical in Campylobacter helveticus: analysis, applications and
Microbiology 38: 953–959. characterization of the intervening sequence found in
Jayarao BM, Dore JJ Jr, Baumbach GA, Matthews KR and Oliver some strains. Microbiology 140: 847–855.
SP (1991). Differentiation of Streptococcus uberis from Loens K, Ursi D, Ieven M, van Aarle P, Sillekens P, Oudshoorn
Streptococcus parauberis by polymerase chain reaction P and Goossens H (2002). Detection of Mycoplasma pneu-
and restriction fragment length polymorphism analysis of moniae in spiked clinical samples by nucleic acid
16S ribosomal DNA. Journal of Clinical Microbiology 29: sequence-based amplification. Journal of Clinical
2774–2778. Microbiology 40: 1339–1345.
Khan MA, Kim CH, Kakoma I, Morin E, Hansen RD, Hurley Loftis AD, Massung RF and Levin ML (2003). Quantitative real-
WL, Tripathy DN and Baek BK (1998). Detection of time PCR assay for detection of Ehrlichia chaffeensis.
Staphylococcus aureus in milk by use of polymerase chain Journal of Clinical Microbiology 41: 3870–3872.
reaction analysis. American Journal of Veterinary Research Logan JM, Edwards KJ, Saunders NA and Stanley J (2001).
59: 807–813. Rapid identification of Campylobacter spp. by melting
Killgore GE, Holloway B and Tenover FC (2000). A 5 nuclease peak analysis of biprobes in real-time PCR. Journal of
PCR (TaqMan) high-throughput assay for detection of the Clinical Microbiology 39: 2227–2232.
mecA gene in staphylococci. Journal of Clinical Manterola L, Tejero-Garces A, Ficapal A, Shopayeva G, Blasco
Microbiology 38: 2516–2519. JM, Marin CM and Lopez-Goni I (2003). Evaluation of a PCR
Kim CH, Khan M, Morin DE, Hurley WL, Tripathy DN, Kehrli M test for the diagnosis of Brucella ovis infection in semen
Jr, Oluoch AO and Kakoma I (2001). Optimization of the samples from rams. Veterinary Microbiology 92: 65–72.
PCR for detection of Staphylococcus aureus nuc gene in McAuliffe L, Ellis RJ, Ayling RD and Nicholas RA (2003).
bovine milk. Journal of Dairy Science 84: 74–83. Differentiation of Mycoplasma species by 16S ribosomal
Kim KS, Ko KS, Chang MW, Hahn TW, Hong SK and Kook YH DNA PCR and denaturing gradient gel electrophoresis fin-
(2003). Use of rpoB sequences for phylogenetic study of gerprinting. Journal of Clinical Microbiology 41:
Mycoplasma species. FEMS Microbiology Letters 226: 4844–4847.
299–305. McKillip JL and Drake M (2000). Molecular beacon polymerase
Kim SG, Shin SJ, Jacobson RH, Miller LJ, Harpending PR, chain reaction detection of Escherichia coli O157: H7 in
Stehman SM, Rossiter CA and Lein DA (2002). milk. Journal of Food Protection 63: 855–859.
Development and application of quantitative polymerase Meiri-Bendek I, Lipkin E, Friedmann A, Leitner G, Saran A,
chain reaction assay based on the ABI 7700 system Friedman S and Kashi Y (2002). A PCR-based method for
(TaqMan) for detection and quantification of the detection of Streptococcus agalactiae in milk. Journal
Mycobacterium avium subsp. paratuberculosis. Journal of of Dairy Science 85: 1717–1723.
Veterinary Diagnostic Investigation 14: 126–131. Misawa N, Kawashima K, Kondo F, Kushima E, Kushima K and
Kinyon JM, Harris DL and Glock RD (1977). Vandamme P (2002). Isolation and characterization of
Enteropathogenicity of various isolates of Treponema hyo- Campylobacter, Helicobacter, and Anaerobiospirillum
dysenteriae. Infection and Immunity 15: 638–646. strains from a puppy with bloody diarrhea. Veterinary
Lanciotti RS and Kerst AJ (2001). Nucleic acid sequence-based Microbiology 87: 353–364.
amplification assays for rapid detection of West Nile and Morré SA, Sillekens P, Jacobs MV, van Aarle P, de Blok S, van
St. Louis encephalitis viruses. Journal of Clinical Gemen B, Walboomers JMM, Meijer CJLM and van den
Microbiology 39: 4506–4513. Brule AJC (1996). RNA amplification by nucleic acid
Lane DJ, Pace B, Olsen GJ, Stahl DA, Sogin ML and Pace NR sequence-based amplification with an internal standard
(1985). Rapid determination of 16S ribosomal RNA enables reliable detection of Chlamydia trachomatis in
sequences for phylogenetic analyses. Proceedings of the cervical scrapings and urine samples. Journal of Clinical
National Academy of Sciences of the United States of Microbiology 34: 3108–3114.
America 82: 6955–6959. Mott J, Rikihisa Y, Zhang Y, Reed SM, Yu CY (1997).
La Scola B, Zeaiter Z, Khamis A and Raoult D (2003). Gene- Comparison of PCR and culture to the indirect fluorescent-
sequence-based criteria for species definition in antibody test for diagnosis of Potomac horse fever.
bacteriology: the Bartonella paradigm. Trends in Journal of Clinical Microbiology 35: 2215–2219.
Microbiology 11: 318–321. Mullis KB and Faloona FA (1987). Specific synthesis of DNA in
Lascols C, Lamarque D, Costa JM, Copie-Bergman C, Le vitro via a polymerase-catalyzed chain reaction. Methods
Glaunec JM, Deforges L, Soussy CJ, Petit JC, Delchier JC in Enzymology 155: 335–350.
and Tankovic J (2003). Fast and accurate quantitative Neimark H, Johansson KE, Rikihisa Y, Tully JG (2001). Proposal
detection of Helicobacter pylori and identification of clar- to transfer some members of the genera Haemobartonella
ithromycin resistance mutations in H. pylori isolates from and Eperythrozoon to the genus Mycoplasma with descrip-
gastric biopsy specimens by real-time PCR. Journal of tions of ‘Candidatus Mycoplasma haemofelis’, ‘Candidatus
Clinical Microbiology 41: 4573–4577. Mycoplasma haemomuris’, ‘Candidatus Mycoplasma
Lew AE, Bock RE, Minchin CM and Masaka S (2002). A haemosuis’ and ‘Candidatus Mycoplasma wenyonii’.
msp1alpha polymerase chain reaction assay for specific International Journal of Systematic and Evolutionary
detection and differentiation of Anaplasma marginale iso- Microbiology 51: 891–899.
lates. Veterinary Microbiology 86: 325–335. Newby DT, Hadfield TL and Roberto FF (2003). Real-time PCR
Leyla G, Kadri G and Umran O (2003). Comparison of poly- detection of Brucella abortus: a comparative study of
merase chain reaction and bacteriological culture for the SYBR green I, 5-exonuclease, and hybridization probe
diagnosis of sheep brucellosis using aborted fetus sam- assays. Applied and Environmental Microbiology 69:
ples. Veterinary Microbiology 93: 53–61. 4753–4759.
Li Q, Luan G, Guo Q and Liang J (2002). A new class of homo- Norman AF, Regnery R, Jameson P, Greene C and Krause DC
geneous nucleic acid probes based on specific (1995). Differentiation of Bartonella-like isolates at the
displacement hybridization. Nucleic Acids Research 30: E5. species level by PCR-restriction fragment length polymor-
92 H. Y. Cai et al.

phism in the citrate synthase gene. Journal of Clinical Riffon R, Sayasith K, Khalil H, Dubreuil P, Drolet M and Lagace
Microbiology 33: 1797–803. J (2001). Development of a rapid and sensitive test for
Nutiu R and Li Y (2002). Tripartite molecular beacons. Nucleic identification of major pathogens in bovine mastitis by
Acids Research 30: e94. PCR. Journal of Clinical Microbiology 39: 2584–2589.
Oliveira K, Procop GW, Wilson D, Coull J and Stender H Sachse K, Grossmann E, Jager C, Diller R and Hotzel H (2003).
(2002). Rapid identification of Staphylococcus aureus Detection of Chlamydia suis from clinical specimens: com-
directly from blood cultures by fluorescence in situ parison of PCR, antigen ELISA, and culture. Journal of
hybridization with peptide nucleic acid probes. Journal of Microbiological Methods 54: 233–238.
Clinical Microbiology 40: 247–251. Sails AD, Fox AJ, Bolton FJ, Wareing DR and Greenway DL
O’Mahony J and Hill C (2002). A real time PCR assay for the (2003). A real-time PCR assay for the detection of
detection and quantitation of Mycobacterium avium Campylobacter jejuni in foods after enrichment culture.
subsp. paratuberculosis using SYBR Green and the Light Applied and Environmental Microbiology 69: 1383–1390.
Cycler. Journal of Microbiological Methods 51: 283–293. Shah DH, Verma R, Bakshi CS and Singh RK (2002). A multi-
Patel JB, Leonard DG, Pan X, Musser JM, Berman RE and plex-PCR for the differentiation of Mycobacterium bovis
Nachamkin I (2000). Sequence-based identification of and Mycobacterium tuberculosis. FEMS Microbiology
Mycobacterium species using the MicroSeq 500 16S rDNA Letters 214: 39–43.
bacterial identification system. Journal of Clinical Sharma VK (2002). Detection and quantitation of enterohemor-
Microbiology 38: 246–251. rhagic Escherichia coli O157, O111, and O26 in beef and
Paton AW and Paton JC (1998). Detection and characterization bovine feces by real-time polymerase chain reaction.
of Shiga toxigenic Escherichia coli by using multiplex PCR Journal of Food Protection 65: 1371–1380.
assays for stx1, stx2, eaeA, enterohemorrhagic E. coli hlyA, Sreevatsan S, Bookout JB, Ringpis F, Perumaalla VS, Ficht TA,
rfbO111, and rfbO157. Journal of Clinical Microbiology Adams LG, Hagius SD, Elzer PH, Bricker BJ, Kumar GK,
36: 598–602. Rajasekhar M, Isloor S and Barathur RR (2000). A multi-
Phuektes P, Mansell PD and Browning GF (2001). Multiplex plex approach to molecular detection of Brucella abortus
polymerase chain reaction assay for simultaneous detec- and/or Mycobacterium bovis infection in cattle. Journal of
tion of Staphylococcus aureus and streptococcal causes of Clinical Microbiology 38: 2602–2610.
bovine mastitis. Journal of Dairy Research 84: 1140–1148. Stender H, Mollerup TA, Lund K, Petersen KH, Hongmanee P
Phuektes P, Browning GF, Anderson G and Mansell PD (2003). and Godtfredsen SE (1999). Direct detection and identifica-
Multiplex polymerase chain reaction as a mastitis screen- tion of Mycobacterium tuberculosis in smear-positive
ing test for Staphylococcus aureus, Streptococcus sputum samples by fluorescence in situ hybridization (FISH)
agalactiae, Streptococcus dysgalactiae and Streptococcus using peptide nucleic acid (PNA) probes. International
uberis in bulk milk samples. Journal of Dairy Research 70: Journal of Tuberculosis and Lung Disease 3: 830–837.
149–155. Stender H, Fiandaca M, Hyldig-Nielsen JJ and Coull J (2002).
Piatek AS, Telenti A, Murray MR, El-Hajj H, Jacobs WR Jr, PNA for rapid microbiology. Journal of Microbiological
Kramer FR and Alland D (2000). Genotypic analysis of Methods 48: 1–17.
Mycobacterium tuberculosis in two distinct populations Stratmann J, Strommenger B, Stevenson K and Gerlach GF
using molecular beacons: implications for rapid suscepti- (2002). Development of a peptide-mediated capture PCR
bility testing. Antimicrobial Agents and Chemotherapy 44: for detection of Mycobacterium avium subsp. paratuber-
103–110. culosis in milk. Journal of Clinical Microbiology 40:
Pinnow CC, Butler JA, Sachse K, Hotzel H, Timms LL and 4244–4250.
Rosenbusch RF (2001). Detection of Mycoplasma bovis in Taillardat-Bisch AV, Raoult D and Drancourt M (2003). RNA
preservative-treated field milk samples. Journal of Dairy polymerase beta-subunit-based phylogeny of Ehrlichia
Science 84: 1640–1645. spp., Anaplasma spp., Neorickettsia spp. and Wolbachia
Pusterla N, Leutenegger CM, Sigrist B, Chae JS, Lutz H and pipientis. International Journal of Systematic and
Madigan JE (2000). Detection and quantitation of Ehrlichia Evolutionary Microbiology 53: 455–458.
risticii genomic DNA in infected horses and snails by real- Tang YW, Von Graevenitz A, Waddington MG, Hopkins MK,
time PCR. Veterinary Parasitology 90: 129–135. Smith DH, Li H, Kolbert CP, Montgomery SO and Persing
Quinn PJ, Markey BK, Donnelly WJ and Leonard FC (2002). DH (2000). Identification of coryneform bacterial isolates
Bacterial causes of bovine mastitis. In: Quinn PJ, Markey by ribosomal DNA sequence analysis. Journal of Clinical
BK, Donnelly WJ and Leonard FC (editors). Veterinary Microbiology 38: 1676–1678.
Microbiology and Microbial Disease. London: Blackwell Tasker S, Helps CR, Day MJ, Gruffydd-Jones TJ and Harbour
Science, pp. 465–475. DA (2003). Use of real-time PCR to detect and quantify
Redkar R, Rose S, Bricker B and DelVecchio V (2001). Real- Mycoplasma haemofelis and ‘Candidatus Mycoplasma
time detection of Brucella abortus, Brucella melitensis and haemominutum’ DNA. Journal of Clinical Microbiology
Brucella suis. Molecular and Cellular Probes 15: 43–52. 41: 439–441.
Reischl U, Youssef MT, Kilwinski J, Lehn N, Zhang WL, Karch Taylor MJ, Hughes MS, Skuce RA and Neill SD (2001).
H and Strockbine NA (2002). Real-time fluorescence PCR Detection of Mycobacterium bovis in bovine clinical speci-
assays for detection and characterization of Shiga toxin, mens using real-time fluorescence and fluorescence
intimin, and enterohemolysin genes from Shiga toxin-pro- resonance energy transfer probe rapid-cycle PCR. Journal
ducing Escherichia coli. Journal of Clinical Microbiology of Clinical Microbiology 39: 1272–1278.
40: 2555–2565. Torres MJ, Criado A, Palomares JC and Aznar J (2000). Use of
Relman DA, Loutit JS, Schmidt TM, Falkow S and Tompkins LS real-time PCR and fluorimetry for rapid detection of
(1990). The agent of bacillary angiomatosis. An approach rifampin and isoniazid resistance-associated mutations in
to the identification of uncultured pathogens. New Mycobacterium tuberculosis. Journal of Clinical
England Journal of Medicine 323: 1573–1580. Microbiology 38: 3194–3149.
Relman DA, Schmidt TM, MacDermott RP and Falkow S (1992). Torioni de Echaide S, Knowles DP, McGuire TC, Palmer GH,
Identification of the uncultured bacillus of Whipple’s dis- Suarez CE and McElwain TF (1998). Detection of cattle
ease. New England Journal of Medicine 327: 293–301. naturally infected with Anaplasma marginale in a region
Molecular genetic methods in the veterinary clinical bacteriology laboratory 93

of endemicity by nested PCR and a competitive enzyme- Walker GT, Little MC, Nadeau JG and Shank DD (1992).
linked immunosorbent assay using recombinant major Isothermal in vitro amplification of DNA by a restriction
surface protein 5. Journal of Clinical Microbiology 36: enzyme/DNA polymerase system. Proceedings of the
777–782. National Academy of Sciences of the United States of
Trotha R, Hanck T, Konig W and Konig B (2001). Rapid ribo- America 89: 392–396.
sequencing—an effective diagnostic tool for detecting Westin L, Miller C, Vollmer D, Canter D, Radtkey R, Nerenberg
microbial infection. Infection 29: 12–16. M and O’Connell JP (2001). Antimicrobial resistance and
Tyagi S and Kramer FR (1996). Molecular beacons: probes that bacterial identification utilizing a microelectronic chip
fluoresce upon hybridization. Nature Biotechnology 14: array. Journal of Clinical Microbiology 39: 1097–104.
303–308. Wilhelm J, Pingoud A and Hahn M (2001). Comparison
Tyagi S, Bratu DP and Kramer FR (1998). Multicolor molecular between Taq DNA polymerase and its Stoffel fragment for
beacons for allele discrimination. Nature Biotechnology quantitative real-time PCR with hybridization probes.
16: 49–53. Biotechniques 30: 1052–1056.
Uyttendaele M, Schukkink R, van Gemen B and Debevere J Yang SJ, Park KY, Seo KS, Besser TE, Yoo HS, Noh KM, Kim
(1994). Identification of Camplylobacter jejuni, SH, Kim SH, Lee BK, Kook YH and Park YH (2001).
Campylobacter coli and Camplylobacter lari by the nucleic Multidrug-resistant Salmonella typhimurium and
acid sequence based amplification system NASBATM. Salmonella enteritidis identified by multiplex PCR from
Journal of Applied Bacteriology 77: 694–701. animals. Journal of Veterinary Science (Suwon-si, Korea)
Van der Vliet GME, Schukkink RAF, van Gemen B, Schepers P 2: 181–188.
and Klatser PR (1993). Nucleic acid sequence based Yang SJ, Park KY, Kim SH, No KM, Besser TE, Yoo HS, Kim
amplification (NASBA) for the identification of SH, Lee BK and Park YH (2002). Antimicrobial resistance
Mycobacteria. Journal of General Microbiology 139: in Salmonella enterica serovars Enteritidis and
2423–2429. Typhimurium isolated from animals in Korea: comparison
Wagenaar J, Zuerner RL, Alt D and Bolin CA (2000). of phenotypic and genotypic resistance characterization.
Comparison of polymerase chain reaction assays with bac- Veterinary Microbiology 86: 295–301.
teriologic culture, immunofluorescence, and nucleic acid Zimmermann K, Eiter T and Scheiflinger F (2003). Consecutive
hybridization for detection of Leptospira borgpetersenii analysis of bacterial PCR samples on a single electronic
serovar hardjo in urine of cattle. American Journal of microarray. Journal of Microbiological Methods 55:
Veterinary Research 61: 316–320. 471–474.

You might also like