You are on page 1of 9

Diagnostic Microbiology and Infectious Disease

49 (2004) 163–171 www.elsevier.com/locate/diagmicrobio

Identification of Bacillus anthracis by multiprobe microarray


hybridization
Dmitriy Volokhova, Andrei Pomerantsevb, Violetta Kivovichb,
Avraham Rasoolyc, Vladimir Chizhikova,*
a
Center for Biologics Evaluation and Research, Food and Drug Administration, Kensington, MD 20895, USA
b
National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
c
Center for Food Safety and Applied Nutrition, Food and Drug Administration, College Park, MD 20740-3835, USA

Received 3 November 2003; accepted 22 March 2004

Abstract
We have developed a rapid assay based on microarray analysis of amplified genetic markers for reliable identification of Bacillus
anthracis and its discrimination from other closely related bacterial species of the Bacillus cereus group. By combining polymerase chain
reaction (PCR) amplification of six B. anthracis-specific genes (plasmid-associated genes encoding virulence factors (cyaA, pagA, lef, and
capA, capB, capC) and one chromosomal marker BA-5449) with analysis of amplicons by microarray hybridization, we were able to
unambiguously identify and discriminate B. anthracis among other closely related species. Bacillus identification relied on hybridization
with multiple individual microarray oligonucleotide probes (oligoprobes) specific to each target B. anthracis gene. Evaluation of the assay
was conducted using several B. anthracis strains (with or without pXO1 and pXO2 plasmids) as well as over 50 other species
phylogenetically related to B. anthracis, including B. cereus, B. thuringiensis, B. mycoides, and B. subtilis. The developed microarray
analysis of amplified genetic markers protocol provides an efficient method for (i) unambiguous identification and discrimination of B.
anthracis from other Bacillus species and (ii) distinguishing between plasmid-containing and plasmid-free Bacillus anthracis strains. © 2004
Elsevier Inc. All rights reserved.

Keywords: Multiplex PCR; pXO1; pXO2; Virulence factors; Anthrax toxins

1. Introduction a group of bacterial toxin genes (pagA, lef, and cyaA) and
pXO2 (93.5 kb), which carries the genes responsible for
Bacillus anthracis is a Gram-positive, rod shaped, spore- capsule synthesis and degradation (capA, capB, capC, and
forming bacterium that belongs to the Bacillus cereus capD) (Leppla, 2000; Makino et al., 2002; Okinaka et al.,
group. This phylogenetically distant group from other Ba- 1999; Pannucci et al., 2002).
cillus species includes B. cereus, B. thuringiensis, B. my- Current methods for B. anthracis identification and char-
coides, and B. weihenstephanensis (Daffonchio et al., 2000; acterization are based on conventional microbiological ap-
Mock and Fouet, 2001). The capsule-forming B. anthracis proaches, which include biochemical testing (Logan et al.,
is the etiological agent of anthrax, the acute and often fatal 1985; Odendaal et al., 1991), immunological testing (Sastry
mammalian disease (Dixon et al., 1999). Although anthrax et al., 2003), antibiotic susceptibility assays (Mohammed et
predominantly occurs among animals (particularly herbi- al., 2002), phage lysing assays (Logan et al., 1985;
vores), occasionally it can also affect humans (Dixon et al., Odendaal et al., 1991; Schuch et al., 2002), and animal
1999). virulence testing (Odendaal et al., 1991). Although shown
Virulent forms of B. anthracis harbor two large patho- to be effective, these approaches are time-consuming and
genicity-related plasmids: pXO1 (181.7 kb), which encodes often yield false-positive results that occur as a consequence
of genetic and phenotypic mimicry of plasmid-free B. an-
* Corresponding author. Tel.: ⫹1-301-827-4041; fax: ⫹1-301-827-
thracis and B. cereus isolates. The utility of molecular
4622. methods based on polymerase chain reaction (PCR) ampli-
E-mail address: chizhikov@cber.fda.gov (V. Chizhikov). fication (including real-time PCR) of unique B. anthracis

0732-8893/04/$ – see front matter © 2004 Elsevier Inc. All rights reserved.
doi:10.1016/j.diagmicrobio.2004.03.015
164 D. Volokhov et al. / Diagnostic Microbiology and Infectious Disease 49 (2004) 163–171

genetic markers could substantially improve B. anthracis based on bacterial cell morphology, colonial morphotype,
identification methodology and facilitate anthrax diagnosis and sensitivity to penicillin. Bacilli that belonged to the
by cutting the time-consuming analysis. The most com- Bacillus cereus group were characterized by the presence of
monly used approach for B. anthracis speciation is based on the cerA gene, the identifying genetic marker for represen-
detection of virulence factor genes associated with pXO1 tatives of this bacterial group (Daffonchio et al., 1999;
and pXO2 plasmids (Bell et al., 2002; Brightwell et al., Schraft and Griffiths, 1995). B. thuringiensis was distin-
1998; Fasanella et al., 2001; Hutson et al., 1993; Jackson et guished using the multiplex PCR assay previously described
al., 1998; Makino et al., 1993; Ramisse et al., 1996; (Ben-Dov et al., 1997).
Timoshin et al., 1994; Turnbull, 1999; Turnbull et al.,
1992). In the case of plasmid loss, however, discriminating 2.2. Total DNA preparation
B. anthracis from other genetically related members of the
Bacillus cereus group poses a significant problem (Helga- Freshly grown bacteria were resuspended in 0.5 mL
son et al., 2000). Thus, the search for genetic markers that water (approximately 108 cells/mL) and treated with ly-
enable unambiguous differentiation of B. anthracis from sozyme (50 mg/mL) for 2 h at 37°C (Chachaty and Saulnier,
other species of the Bacillus cereus group is an important 2000) to disrupt bacterial walls. Proteins were removed
and challenging task. from solution by two sequential phenol-chloroform (1:1)
Several chromosomal gene markers such as 16S rRNA extractions, and bacterial DNA was precipitated by 3 vol-
(Liu et al., 2001; Sacchi et al., 2002), 16S-23S internal umes of anhydrous ethanol. The DNA pellets were dried in
transcribed spacers (Daffonchio et al., 2000), Ba813 (Patra a vacuum and resuspended in 300 ␮L of water. The pres-
et al., 1996; Ramisse et al., 1999), rpoB (Qi et al., 2001), ence and quality of genomic DNA were confirmed by 0.8%
SG749 (Daffonchio et al., 1999), vrrA (Andersen et al., agarose gel electrophoresis followed by gel staining with
1996; Jackson et al., 1997; Keim et al., 2000), and AC-390 ethidium bromide.
(Cherif et al., 2002) have been reported to be potentially
suitable targets for recognition of B. anthracis plasmid- 2.3. Design of the PCR primers and microarray
cured strains by PCR amplification. However, later studies oligoprobes
revealed that a number of these chromosomal markers have
limited specificity (Ramisse et al., 1999), do not provide PCR amplification of target genes (cyaA, pagA, and lef)
sufficient polymorphism to differentiate B. anthracis (Ash encoded on the pXO1 was accomplished using three pairs of
et al., 1991; Bourque et al., 1995; Harrell et al., 1995), or primers previously published (Table 2) (Jackson et al.,
require using complicated and time-consuming experimen- 1998). Three pairs of primers for amplification of B. an-
tal protocols (Daffonchio et al., 1999). thracis capsule genes (capA, capB, and capC) were selected
In this study we describe a novel approach based on based on conserved sequences found within each of the
microarray analysis of amplified genetic markers target genes (Table 2).
(MAAGM), which can be used for unambiguous identifi- B. anthracis-specific chromosomal marker BA-5449 was
cation and characterization of a variety of genetic variants identified by genomic analysis of several recently se-
of B. anthracis strains. The effectiveness of the new ap- quenced B. anthracis strains (GenBank accession numbers
proach was evaluated by its ability to discriminate between NC_003995, NC_003997, NC_004126, and NC_004352).
B. anthracis and other closely related species of the Bacillus Primers for PCR amplification (Table 2) of the BA-5449
genus. ORF were designed using conserved sequences in the re-
gions flanking the BA-5449.
Microarray species-specific oligoprobes for the seven
2. Materials and methods selected genes were designed using following criteria: oli-
goprobes were selected in the regions highly homologous
2.1. Bacterial strains among all known genetic alleles of the target gene (Table 3),
and the melting temperature of complex oligoprobe with
The Bacillus strains (Table 1) were obtained from the template was 50°C and higher (basic algorithm). The se-
American Type Culture Collection (ATCC, Manassas, VA), quences of the target genes found in the GenBank were
the collections of Dr. F. Khambaty of the Center for Food aligned using ClustalX software (Thompson et al., 1994),
Safety and Applied Nutrition, the Food and Drug Admin- and the conserved regions were used to design the gene-
istration (College Park, MD), and Dr. S. Leppla of the specific oligoprobes summarized in Table 4. The 5⬘-end of
National Institute of Allergy and Infectious Diseases, Na- each oligonucleotide was modified during synthesis using
tional Institutes of Health (Bethesda, MD). Some spore- TFA Aminolink CE reagent (PE Applied Biosystems, Fos-
forming, aerobic, Gram-positive Bacillus spp. were isolated ter City, CA) to enable attachment to the silylated glass
from the environment (soil samples) and were characterized slides (CEL Associates, Inc., Houston, TX).
D. Volokhov et al. / Diagnostic Microbiology and Infectious Disease 49 (2004) 163–171 165

Table 1
Origin and description of laboratory and reference strains used in this study

Species Strain (isolate) Relevant characteristic(s) Microarray result Sourcea


⫺ ⫹ ⫺ ⫹ ⫺ ⫹
B. anthracis Ames 35 and Ames 35 (spo0A ) pXO1 /pXO2 pXO1 /pXO2 /BA-5449 LS
B. anthracis Ames 34 and two Ames 34 (spo0A⫺) variants pXO1⫺/pXO2⫹ pXO1⫺/pXO2⫹/BA-5449⫹ LS
B. anthracis Ames 33 and two Ames 33 (spo0A⫺) variants pXO1⫺/pXO2⫺ pXO1⫺/pXO2⫺/BA-5449⫹ LS
B. anthracis Ames 35 (pepM4C⫺) pXO1⫹/pXO2⫺ pXO1⫹/pXO2⫺/BA-5449⫹ LS
B. anthracis Three Ames 35 (spo0A⫺ pepM4C⫺) variants pXO1⫹/pXO2⫺ pXO1⫹/pXO2⫺/BA-5449⫹ LS
B. anthracis Sterne vaccine strain pXO1⫹/pXO2⫺ pXO1⫹/pXO2⫺/BA-5449⫹ LS
B. cereus 569 cerA⫹b; Hem⫹c Negative LS
B. cereus ATCC 49063 cerA⫹; serotype H.26; Hem⫹ Negative ATCC
B. cereus ATCC 33019 cerA⫹; Hem⫹ Negative ATCC
B. cereus 860 cerA⫹; Hem⫹ Negative KF
B. cereus 1241 cerA⫹; Hem⫹ Negative KF
B. cereus 1242 cerA⫹; Hem⫹ Negative KF
B. cereus 1257 cerA⫹; Hem⫹ Negative KF
B. cereus 1259 cerA⫹; Hem⫹ Negative KF
B. cereus 1261 cerA⫹; Hem⫹ Negative KF
B. cereus 1263 cerA⫹; Hem⫹ Negative KF
B. cereus 1265 cerA⫹; Hem⫹ Negative KF
B. thuringiensis ATCC 13366 cerA⫹ Negative ATCC
B. thuringiensis subsp. israelensis ATCC 35646 cerA⫹; produces delta-endotoxin Negative ATCC
B. thuringiensis 1258 cerA⫹ Negative KF
B. thuringiensis 1260 cerA⫹ Negative KF
B. thuringiensis 1262 cerA⫹ Negative KF
B. thuringiensis 1264 cerA⫹ Negative KF
B. thuringiensis 1266 cerA⫹ Negative KF
B. subtilis ATCC 19659 cerA⫺ Negative ATCC
B. subtilis ATCC 21332 cerA⫺; produces surfactin Negative ATCC
B. subtilis 1267 cerA⫺ Negative KF
B. subtilis 1268 cerA⫺ Negative KF
B. subtilis 1269 cerA⫺ Negative KF
B. subtilis 1270 cerA⫺ Negative KF
B. subtilis 1271 cerA⫺ Negative KF
B. mycoides ATCC 6462 cerA⫹; type strain Negative ATCC
B. mycoides CP9 cerA⫹; PRd Negative SS
B. mycoides CP2 cerA⫹; PR Negative SS
B. mycoides CP3 cerA⫹; PR Negative SS
B. mycoides CP13 cerA⫺; PR Negative SS
B. mycoides PR3 cerA⫹; PR Negative SS
B. mycoides TW2 cerA⫹; PR Negative SS
B. mycoides TW4 cerA⫹; PR Negative SS
B. mycoides TW5 cerA⫺; PR Negative SS
B. mycoides TW7 cerA⫹; PR Negative SS
B. mycoides TW8 cerA⫹; PR Negative SS
B. mycoides WM5 cerA⫺; PR Negative SS
B. mycoides WF1 cerA⫹; PR Negative SS
B. mycoides WF14 cerA⫹; PR Negative SS
B. mycoides WFA1 cerA⫺; PR Negative SS
B. mycoides WFA2 cerA⫹ PR Negative SS
B. mycoides SG1 cerA⫹ PR Negative SS
Bacillus spp.e CP1 cerA⫹ PR Negative SS
Bacillus spp. CP5 cerA⫹ PR Negative SS
Bacillus spp. CP8 cerA⫺; PR Negative SS
Bacillus spp. CP10 cerA⫹ PR Negative SS
Bacillus spp. CP12 cerA⫹ PR Negative SS
Bacillus spp. CP16 cerA⫹ PR Negative SS
Bacillus spp. PR2 cerA⫹ PR Negative SS
Bacillus spp. PR4 cerA⫹ PR Negative SS
Bacillus spp. TW1 cerA⫹ PR Negative SS
Bacillus spp. TW3 cerA⫹ PR Negative SS
Bacillus spp. WM1 cerA⫹ PR Negative SS
Bacillus spp. WM2 cerA⫹ PR Negative SS
Bacillus spp. WF7 cerA⫹ PR Negative SS
Bacillus spp. KS14 cerA⫹ PR Negative SS
a
LS, Leppla S; NIH, National Institute of Allergy and Infectious Diseases; ATTC, American Type Culture Collection; KF, Khambaty F; FDA, Food and
Drug Administration; SS, gram-positive Bacillus spp. isolated from environmental (soil samples).
b
Cereolysin A gene positive (cerA⫹) or negative (cerA⫺).
c
Hemolysin positive (Hem⫹).
d
Penicillin G–resistant isolate (PR).
e
Bacillus isolates belonging to Bacillus cereus group.
166 D. Volokhov et al. / Diagnostic Microbiology and Infectious Disease 49 (2004) 163–171

Table 2
Primers used for amplification of Bacillus anthracis genes

Target Primer Nucleotide sequence (5⬘-3⬘) Gene GenBank PCR product Tm (°C)
accession no. size (bp)

pXO1 LEF-1F GGTGCGGATTTAGTTGATTC lef NC_003980 851 50


LEF-1P CGCTTCATTTGTTCTCCC 48
CYA-1F GCGATGAAAACAACGAAGTA cyaA NC_001496 720 48
CYA-1P TCGTCTTTGTCGCCACTATC 52
PA-1F CCAGACCGTGACAATGATG pagA NC_001496 508 51
PA-1R CAAGTTCTTTCCCCTGCTA 49
pXO2 CAPAF TGCGACATGGGTACAACGTACAGAAGC capA NC_003981 525 61
CAPAR TGTTCTTTCGTTGCAATAGCTCCTGCTAC 60
CAPBF CATCGCTTGCTTTAGCGGTAGCAGAG capB NC_003981 432 61
CAPBR GCATACGAGACATAATTTCACTTGTTGACC 59
CAPCF TTGCATTAGTATTAGGAGTTACACTGAGCC capC NC_003981 534 59
CAPCR AGAGGTAATACGATTGCTACATAACGAGG 59
Chromosomal BA-F1 CATGGAATCAAGCTGCAAATTATAAAG BA-5449 NC_003995 1017 54
marker region
BA-R1 CTAAATTGTCATTTGGCAAATTGAAATTC 53

2.4. Microchip design and fabrication sets of primers to ensure the high sensitivity of developed
assay. Thus, the ANX1 primer mixture contained primers
The microchip for identification of B. anthracis, and its for amplification of pXO1-related genes (cyaA, pagA, and
discrimination from closely related Bacillus species, was lef), whereas ANX2 was used for simultaneous amplifica-
designed to contain multiple individual oligonucleotide tion of pXO2 plasmid genes (capA, capB, and capC) and
probes complementary to each target gene (Table 4). The BA-5449 chromosomal intergenic region. The standard
dispensing of oligoprobes onto the microchip surface was PCR mixture (50 ␮L) contained 3 units of HotStarTaq DNA
accomplished using a previously described protocol (Vo- polymerase, 1⫻ reaction buffer supplemented with 2.5 mM
lokhov et al., 2002). MgCl2 (Qiagen, Chatsworth, CA), 300 nM (each) forward
and reverse primers, 300 ␮M deoxynucleoside triphosphate
2.5. Multiplex PCR amplification (dATP, dGTP, dCTP, and dTTP), and 1–2 ␮L DNA tem-
plate (approximately 0.2 ␮g of total bacterial DNA). The
Table 2 summarizes the data on all primers used for B. PCR was carried out using the Gene Amp PCR System
anthracis target gene amplification. Amplification of seven 9700 thermocycler (Applied Biosystems, Foster City, CA)
target B. anthracis genes was performed using two separate with the following cycle conditions: initial activation at
95°C for 15 min, 40 cycles at 94°C for 30 sec, 55°C or 60°C
for 30 sec for pXO1 and pXO2 primer sets respectively,
Table 3 72°C for 1 min, and the final extension at 72°C for 10 min.
Accession numbers of the sequences used for the design of the
microarray oligonucleotide probes and the some cap-specific primers
The synthesis of PCR products was confirmed by electro-
phoresis in 2% agarose gel containing 1⫻ TAE buffer
Gene Organism Accession no. followed by staining with ethidium bromide.
lef Bacillus anthracis NC_003980; AE011190;
AJ413935; AJ413934; 2.6. Synthesis of Cy5-labeled ssDNA
NC_001496; AF065404;
M29081; M30210 Single-stranded DNA (ssDNA) samples for microarray
cyaA Bacillus anthracis AJ413931; AJ413930;
hybridization were synthesized by 40 cycles of the primer
NC_001496; AF065404;
AF003936; A32503; A07289; extension reaction (Volokhov et al., 2002) in the presence of
A02546; M24074; M23179 a mixture of reverse primers (Table 2). The reaction was
pagA Bacillus anthracis AJ413937; AJ413936; conducted in 50 ␮L containing 5 units of Taq DNA poly-
NC_001496; AF306783; merase (Sigma, St. Louis, MO), 1⫻ reaction buffer with 3.0
AF306782; AF306781;
mM MgCl2, 600 nM (each) reverse primer, 200 nM of
AF306780; AF306779;
AF306778; AF065404 dGTP, dATP, and dTTP, 40 nM dCTP, 40 nM Cy5-dCTP
cap (A,B,C) Bacillus anthracis NC_003981; AE011191; and 200 ng of double-stranded DNA template from the first
M24150 round of multiplex PCR and purified using QIAquick PCR
yws (A,B,C) Bacillus subtilis Z92954; AB039950; purification kit (Qiagen, Chatsworth, CA). The protocol
AB046355; Z92953
included the initial activation step at 95°C for 1 min fol-
cap (B,C) Bacillus firmus U60883
lowed by 40 cycles at 94°C for 20 sec, 58°C for 20 sec, and
D. Volokhov et al. / Diagnostic Microbiology and Infectious Disease 49 (2004) 163–171 167

Table 4
Oligonucleotide probes for detection and discrimination of Bacillus anthracis

Type Name Sequence Length (nt) G⫹C (%) Tma (°C)


lef specific Lef1 TTGAAATGGAGAATCCAATTATCAC 25 32 51
Lef2 ATGGAAAGCTTATATTACAAAGAAAC 26 27 50
Lef3 TCAAGAAGCACAGTTAAATATAAATC 26 27 50
Lef4 ATACAAAGCTTATTACATTCAACG 24 29 49
Lef5 ATGCATCCAATATTGTAGAAAGT 23 30 48
Lef6 TAGTTGATGGTAATGGAAGATTTG 24 33 51
cya specific Cya1 TGGAGAAATATAGAAGTGATGG 22 36 49
Cya2 TTGCACTTGCCCCAAGTTTAAC 22 45 53
Cya3 TAACACCCCAAATTCATTAGAAAAG 25 32 51
Cya4 ATTAAATATGGAATTGAGAGGAAAC 25 28 49
Cya5 ACAAATGCTTGATCGTTTGAATG 23 35 50
Cya6 AATAGCTCCTGGTAATAAAGCTTAT 25 32 51
pag specific Pag1 TCACCATGGATTTCTAATATTCATG 25 32 51
Pag2 ATGGAGCACGGCTTCTGATC 20 55 54
Pag3 TTACAGGACGGATTGATAAGAAA 23 35 50
Pag4 ATGTAGATATGGAGAATATTATTCTCT 27 26 51
Pag5 AAGTACATGGAAATGCAGAAGT 22 36 49
Pag6 ATCTGCAGGATTTAGTAATTCG 22 36 49
capA specific CapA1 TCGTGAGAACGAAAAATTGACG 22 41 51
CapA2 ATGGTTGGTGACATTATGATGG 22 41 51
CapA3 AGATTGTTAATCGTTACGGTACA 23 35 50
CapA4 TACGTAAGTGGGAATTTCGAAC 22 41 51
CapA5 AGATAAGAATATTCACTTAAGTGC 24 29 49
CapA6 TGACGGATTATGGTGCTAAGG 21 48 52
capB specific CapB1 TTGCAGCGAATGATCCCTC 19 53 51
CapB2 ACATTACGTATTTGGGAACGT 21 38 49
CapB3 ATGATTTTGGATATAGTAATCTAGC 25 28 49
CapB4 ATTGTAATTATGAATTGCCGCC 22 36 49
CapB5 ACCGCGTTGATCGTACTGAG 20 55 54
CapB6 TGCCATATATTAAAGCGGAAATA 23 30 48
capB specific CapC1 AATCGTTACGTATGGTGTTTCA 22 36 49
CapC2 CGTAGAAAATTTGCGGCAAC 20 45 50
CapC3 TCAGCCCGTATTTATGTTGGT 21 43 50
CapC4 ATGGTGTTTCAAGATTCATGATTTT 25 28 49
CapC5 TATTTGTTTAAAACTTTTATTTGATTATTG 30 13 48
CapC6 ATCAGTATTTTAACATATGTAATCGTT 27 22 49
BA-5449 region BA-1 TTATTAAAGAGCCAGAGGGAT 21 38 49
specific
BA-2 AGCGGTAAAGTAAAGGCACA 20 45 50
BA-3 TGCTTTTAACCGATCGTTTTATAAA 25 28 49
BA-4 TATATAAAACATTAGGATTCGAT 23 22 45
BA-5 AAACAAAGAAGTAGTTGGAGAT 22 32 47
BA-6 ATAATAGATGAATTAAAAAAGAGAA 25 16 45
BA-7 TGAAGAGACGGTAAGAGAAAG 21 43 50
BA-8 TAGTGGATTAACAGAAGAGGA 21 38 49
BA-9 TCAGTTGCAAGTGAATTAAAGA 22 32 47
QC QCprb TGGCAGAAGCTATGAAACGATATGGG 26 46 58
Cy3-QC CCCATATCGTTTCATAGCTTCTGCCA 26 46 58
a
Basic melting temperature (Tm) was calculated with the Oligonucleotide Properties Calculator (http://www.basic.nwu.edu/biotools/oligocalc.html).

72°C for 2 min with the final extension at 72°C for 10 min. 0.1% Tween 20) at 53°C for 30 min. Before hybridization,
The fluorescently labeled ssDNA products were purified 1 ␮L of the Cy5-labeled ssDNA sample was mixed with an
from nonincorporated reagents with the QIAquick PCR equal volume of 2⫻ hybridization buffer containing 0.1 ␮M
purification kit according to the manufacturer’s protocol, Cy3-labeled QC probe (Cy3-QCprb) (Table 4), followed by
dried in vacuum, and solubilized in 10 ␮L of water. a denaturing step at 95°C for 1 min and chilling on ice.
Hybridization mixtures were placed onto separate micro-
2.7. Hybridization conditions chip areas containing oligoprobe microarrays and covered
with a 5 ⫻ 5-mm plastic coverslip to prevent evaporation
Hybridization of fluorescently labeled ssDNA samples to during incubation. Microchips with 10 identical arrays were
the oligonucleotide microarray was performed in 1⫻ hy- used. After the hybridization, the slides were washed for 1
bridization buffer (5⫻ Denhardt’s solution, 6⫻ SSC, and min with 6⫻ SSC containing 0.2% Tween 20, followed by
168 D. Volokhov et al. / Diagnostic Microbiology and Infectious Disease 49 (2004) 163–171

Fig. 1. Microarray analysis of multiple genetic markers of B. anthracis strains using the “Anthrax” microchip. Amplification of seven target B. anthracis genes
was conducted using two separate multiplex PCR mixtures containing primers specific for either (A) cyaA, pagA, and lef or (B) capA, capB, capC and
chromosomal marker BA-5449 genes. Some characteristic hybridization patterns of B. anthracis strains are shown in (C). (A) Lanes: M, MassRuler DNA
Ladder (SM0383, Fermentas); 1, Ames 35; 2, Ames 35(spo0A⫺); 3, Ames 35 (pepM4C⫺); 4, Ames 35 (spo0A⫺pepM4C⫺); 5, Ames 35
(spo0A⫺pepM4C⫺)v2; 6, Ames 35 (spo0A⫺pepM4C⫺)v3; 7, Sterne vaccine strain. (B) Lanes: M, GeneRuler DNA Ladder Mix (SM0333, Fermentas); 1,
Ames 34; 2, Ames 34 (spo0A⫺); 3, Ames 34 (spo0A⫺)v2; 4, Ames 35; 5, Ames 33 (spo0A⫺); 6, Ames 33 (spo0A⫺)v2; 7, Sterne vaccine strain. (C)
Hybridization patterns of B. anthracis strains containing either none of above plasmids or pXO1, or XO2 are shown on panels 1, 2, and 3, respectively.
Microarray rows a and b contain individual oligoprobes for pXO1-specific markers: pagA (columns 1–3), lef (columns 4 – 6), and cyaA (columns 7–9), rows
c and d represent pXO2-specific markers: capA (columns 1–3), capB (columns 4 – 6), and capC (columns 7–9), and row c includes oligoprobes for the
chromosomal marker BA-5449.

three consecutive washes (1 min each) with 6⫻ SSC buffer, 1). Synthesis of detectable amounts of amplicons was observed
2⫻ SSC buffer, and 1⫻ SSC buffer. Traces of remaining with all pXO1-containing B. anthracis isolates, whereas no
buffer were removed by air stream. PCR product associated with the pXO1 plasmid was detected
when DNA from other Bacillus species has been used as PCR
2.8. Microchip scanning and analysis templates (Table 1).
The results of amplification were more complex for the
Fluorescent images of the microarrays were obtained by ANX2 set of primers. We observed the appearance of non-
scanning slides using ScanArray 5000 (PerkinElmer, Bos- specific DNA products having molecular sizes close to
ton, MA). The fluorescent signals from each spot were those of pXO2-derived PCR fragments when the DNA from
measured and compared using QuantArray software some non–B. anthracis was used for PCR amplification.
(PerkinElmer, Boston, MA). Moreover, although some PCR products obtained by am-
plification of B. thuringiensis showed low cross-hybridiza-
tion with B. anthracis cap-specific microarray oligoprobes,
3. Results
the pattern of hybridization signals from oligoprobes differed
3.1. PCR amplification of target B. anthracis genes substantially from that of pXO2-bearing B. anthracis (data not
shown). To improve the specificity of the assay, we designed
The two primer sets, ANX1 and ANX2, were used for PCR a new set of primers for amplification of the capA, capB, and
amplification of the pXO1-associated genes and the pXO2- capC genes. This new set of primers was evaluated using
associated genes in combination with the chromosomal different Bacillus species, summarized in Table 1. We found
marker, respectively. The first set was composed of primers that these primers enabled efficient and highly specific ampli-
previously optimized for analysis of B. anthracis pXO1 genes fication of target pXO2 genes exclusively from pXO2-contain-
pagA, lef, and cyaA (Jackson et al., 1998). We found that these ing B. anthracis strains (Fig. 1B, Table 1).
primers efficiently and specifically amplified all target B. an- The new chromosomal marker, BA-5449 (1017 nts), incor-
thracis genes in both single and multiplex PCR (Fig. 1A, Table porated the sequence of the BA813 marker, which has been
D. Volokhov et al. / Diagnostic Microbiology and Infectious Disease 49 (2004) 163–171 169

previously proposed as a useful discriminatory factor for B. et al., 1997; Turnbull et al., 1992). Potentially, the sensitiv-
anthracis identification. New primers, BA-F1 and BA-R1, ity of PCR amplification performed at optimal conditions
designed to amplify BA-5449 marker, demonstrated high se- enables detection of single copies of target DNA molecules
lectivity in amplification and were able to generate PCR prod- (Tang et al., 1997).
ucts of expected size exclusively with the B. anthracis isolates Several PCR-based approaches that rely on identification
(Fig. 1B). No amplification was observed with any other spe- of different B. anthracis chromosomal and extrachromo-
cies of the Bacillus cereus group (data not shown). somal genetic markers (e.g., 16-23S rRNA intergenic spac-
ers, Ba813, rpoB, GS-749, AC-390 and cyaA, pagA, lef,
3.2. MAAGM-based identification of the B. anthracis capA, capB, capC, respectively) have been developed and
described (Bell et al., 2002; Cherif et al., 2002; Daffonchio
Discrimination of B. anthracis from other bacilli of the et al., 1999; Daffonchio et al., 2000; Fasanella et al., 2001;
Bacillus cereus group was based on PCR amplification of Gryadunov et al., 2001; Jackson et al., 1998; Liu et al.,
target regions of seven B. anthracis–specific genes using 2001; Makino et al., 1993; Patra et al., 1996; Qi et al., 2001;
two primer sets, followed by analysis of the amplified DNA Ramisse et al., 1999; Reif et al., 1994). Although detection
by hybridization with the microchip containing multiple of plasmid-associated genes was demonstrated to be a rel-
sequence-specific oligonucleotide probes. The “Anthrax” atively simple and efficient procedure, the selection of sta-
microchip included a total of 45 oligoprobes placed into ble chromosomal markers unique to B. anthracis was ham-
seven separate panels. Each panel contained oligoprobes pered by the high genetic similarity among different
specific to only one target gene (Fig. 1C). Bacillus species (Ash et al., 1991; Bourque et al., 1995;
The specificity and discriminatory power of the method Daffonchio et al., 2000; Patra et al., 1996; Ramisse et al.,
were evaluated using 56 bacterial isolates closely related 1996; Ramisse et al., 1999; Sacchi et al., 2002; Yamada et
genetically to B. anthracis (Table 1). The results of this al., 1999). We conducted comparative analyses of several
analysis are presented on Fig. 1C, and Table 1. recently sequenced B. anthracis and B. cereus genomes
Only isolates of B. anthracis were found to produce PCR (Ivanova et al., 2003; Read et al., 2003) to identify chro-
products capable of hybridizing with the oligoprobes specific mosomal elements unique to B. anthracis. The analysis
to the B. anthracis chromosomal marker and pXO1/pXO2 allowed us to reveal few genomic regions in the B. anthracis
plasmid-associated genes. The microarray data on the pres- genome that are suitable for discrimination of B. anthracis
ence/absence of pXO1 and pXO2 were in concordance with from other Bacillus species. One of such regions, designated
the phenotypic observation of the B. anthracis isolates. The as BA-5449, was selected for further study. This region
microarray analysis unambiguously identified the presence of incorporated the sequence of genetic marker BA813, previ-
pXO1 in the Ames 35 strain, which produces protective anti- ously proposed for identification of B. anthracis. However,
gen. pXO2 was found in the Ames 34 strain, which produces the utility of the BA813 as a B. anthracis chromosomal
capsule, whereas none of the plasmids was detected in Ames marker was demonstrated to be limited, because of the
33, which produces neither protective antigen nor capsule. presence of sequences homologous to the BA813 in other
No synthesis of the characteristic PCR products or cross- Bacillus species, e.g., B. cereus and B. thuringiensis (Ash et
hybridization of B. anthracis-specific oligoprobes was ob- al., 1991; Bourque et al., 1995; Daffonchio et al., 2000;
served with any other Bacillus species included in this study. Patra et al., 1996; Ramisse et al., 1996; Ramisse et al., 1999;
Sacchi et al., 2002; Yamada et al., 1999). Proposed novel
marker BA-5449 of bigger size (1017 bp) contained addi-
4. Discussion tional multiple sequences unique to B. anthracis and appro-
priate for its identification. Using BA-5449 –specific PCR
In this paper, we describe a development and evaluation primers, we observed the synthesis of amplicon of expected
of a new method for a reliable identification of plasmid- molecular size only when B. anthracis DNA has been used
bearing and plasmid-cured strains of B. anthracis and dis- as a PCR template. In contrast, no PCR products appeared
crimination of anthrax bacilli from other members of the when DNAs of other Bacillus species have been analyzed. The
Bacillus cereus group. simplicity and flexibility of the MAAGM system enable the
All currently available assays for B. anthracis identifi- extending of the number of microarray oligoprobes and PCR
cation suffer, as noted above, from being either labor-con- primers to order to adapt and include in this assay new chro-
suming or having limited specificity (Bell et al., 2002; mosomal markers that might be found suitable for B. anthracis
Ramisse et al., 1996; Ramisse et al., 1999) and call for a identification (Radnedge et al., 2003). Although no cross-spe-
search of new technical approaches enabling a rapid and cies amplification was observed with the Bacillus species used
reliable species identification. The application of PCR- in this study, no one can exclude the possibility that other
based techniques for characterization of species-specific ge- bacilli species can contain genetic regions homologous to BA-
netic markers has substantially improved and facilitated 5449 and may produce some PCR products with physical
detection and identification of many different bacterial characteristics close to those of B. anthracis amplicons, as was
pathogens (Louie et al., 2000; Mock and Fouet 2001; Tang observed with other markers (Ramisse et al., 1996; Ramisse et
170 D. Volokhov et al. / Diagnostic Microbiology and Infectious Disease 49 (2004) 163–171

al., 1999). The main advantage of the MAAGM is that this Bell CA, Uhl JR, Hadfield TL, David JC, Meyer RF, Smith TF, Cockerill
method relies on identification of characteristic sequences in FR 3rd (2002). Detection of Bacillus anthracis DNA by LightCycler
PCR. J Clin Microbiol 40, 2897–2902.
target regions of selected marker genes. The hybridization of Ben-Dov E, Zaritsky A, Dahan E, Barak Z, Sinai R, Manasherob R,
amplicons with multiple short microarray oligoprobes (20-25- Khamraev A, Troitskaya E, Dubitsky A, Berezina N, Margalith Y
mers) provides accurate analysis of such regions and identifi- (1997). Extended screening by PCR for seven cry-group genes from
cation of Bacillus anthracis. Because of oligoprobe redun- field-collected strains of Bacillus thuringiensis. Appl Environ Micro-
dancy, the MAAGM is potentially much more reliable in biol 63, 4883– 4890.
Bourque SN, Valero JR, Lavoie MC, Levesque RC (1995). Comparative
comparison with other PCR-based methods, e.g., real-time analysis of the 16S to 23S ribosomal intergenic spacer sequences of
PCR. For the same reason, the MAAGM is not affected by the Bacillus thuringiensis strains and subspecies and of closely related
occasional appearance of sporadic PCR products that can oc- species. Appl Environ Microbiol 61, 1623–1626.
cur as a result of mispriming during PCR amplification (Elni- Brightwell G, Pearce M, Leslie D (1998). Development of internal controls
fro et al., 2000; Markoulatos et al., 2002). Such nonspecific for PCR detection of Bacillus anthracis. Mol Cell Probes 12, 367–377.
Chachaty E, Saulnier P (2000). Isolation chromosomal DNA from bacteria.
DNA products can often be observed when multiplex PCR is In The Nucleic Acid Protocols: Handbook. Ed, R Rapley. Totowa, NJ:
used for simultaneous amplification of multiple genetic mark- Humana Press Inc., pp 29 –32.
ers. However, such amplicons are unable to hybridize to the Cherif A, Borin S, Rizzi A, Ouzari H, Boudabous A, Daffonchio D (2002).
sequence-specific microarray oligoprobes and therefore cannot Characterization of a repetitive element polymorphism-polymerase
affect microarray-based species identification. chain reaction chromosomal marker that discriminates Bacillus anthra-
cis from related species. J Appl Microbiol 93, 456 – 462.
The current cost of microarray analysis is relatively high, Cherkasova E, Laassri M, Chizhikov V, Korotkova E, Dragunsky E, Agol
but there are several potential approaches to reduce signifi- V, Chumakov K (2003). Microarray analysis of evolution of RNA
cantly microarray expenses and to make this technology af- viruses: New evidence of circulation of virulent highly divergent vac-
fordable for the majority of clinical and public health labora- cine-derived polioviruses. Proc Natl Acad Sci U S A 100, 9398 –9403.
tories. Thus, the implication of simplified variants of scanners Chizhikov V, Rasooly A, Chumakov K, Levy DD (2001). Microarray
analysis of microbial virulence factors. Appl Environ Microbiol 67,
(www.darpa.mil/dso/thrust/biosci/biosensor/argonne.html) and
3258 –3263.
the development of new protocols for preparation of fluores- Chizhikov V, Wagner M, Ivshina A, Hoshino Y, Kapikian AZ, Chumakov
cently-labeled samples for microarray analysis using less ex- K (2002). Detection and genotyping of human group A rotaviruses by
pensive dyes are main ways to reach this goal. oligonucleotide microarray hybridization. J Clin Microbiol 40, 2398 –
We have successfully used this approach in previous 2407.
Daffonchio D, Borin S, Frova G, Gallo R, Mori E, Fani R, Sorlini C (1999).
experiments for the detection and discrimination of different
A randomly amplified polymorphic DNA marker specific for the Ba-
viral and bacterial pathogens from closely related nonpatho- cillus cereus group is diagnostic for Bacillus anthracis. Appl Environ
genic species (Cherkasova et al., 2003; Chizhikov et al., Microbiol 65, 1298 –1303.
2001; Chizhikov et al., 2002; Laassri et al., 2003; Volokhov Daffonchio D, Cherif A, Borin S (2000). Homoduplex and heteroduplex
et al., 2002). The simplicity of the proposed microarray polymorphisms of the amplified ribosomal 16S-23S internal tran-
scribed spacers describe genetic relationships in the “Bacillus cereus
protocol for simultaneous identification of several B. an-
group.” Appl Environ Microbiol 66, 5460 –5468.
thracis–specific genetic markers and the ability to perform Dixon TC, Meselson M, Guillemin J, Hanna PC (1999). Anthrax. N Engl
accurate analysis of multiple samples in a short period (4 –5 J Med 341, 815– 826.
hours) present a great improvement over the existing meth- Elnifro EM, Ashshi AM, Cooper RJ, Klapper PE (2000). Multiplex PCR:
odology for B. anthracis identification. Optimization and application in diagnostic virology. Clin Microbiol
Rev 13, 559 –570.
Fasanella A, Losito S, Trotta T, Adone R, Massa S, Ciuchini F, Chiocco D
(2001). Detection of anthrax vaccine virulence factors by polymerase
Acknowledgments chain reaction. Vaccine 19, 4214 – 4218.
Gryadunov DA, Mikhailovich VM, Noskov AN, Lapa SA, Sobolev A,
This work was supported in part by grants from USDA Pankov SV, Rubina A, Zasedatelev AS, Mirzabekov AD (2001). De-
tection of Bacillus anthracis using multiplex PCR on the oligonuclotide
and funding from FDA office of science awarded to Dr. V. biochip. Dokl Biochem Biophys 381, 384 –386.
Chizhikov and Dr. A. Rasooly and from a grant from the Harrell LJ, Andersen GL, Wilson KH (1995). Genetic variability of Ba-
U.S. Defense Advanced Research Project Agency to Dr. K. cillus anthracis and related species. J Clin Microbiol 33, 1847–1850.
Chumakov. Helgason E, Okstad OA, Caugant DA, Johansen HA, Fouet A, Mock M,
Hegna I, Kolsto AB (2000). Bacillus anthracis, Bacillus cereus, and
Bacillus thuringiensis— one species on the basis of genetic evidence.
Appl Environ Microbiol 66, 2627–2630.
References Hutson RA, Duggleby CJ, Lowe JR, Manchee RJ, Turnbull PC (1993). The
development and assessment of DNA and oligonucleotide probes for the
Andersen GL, Simchock JM, Wilson KH (1996). Identification of a region specific detection of Bacillus anthracis. J Appl Bacteriol 75, 463– 472.
of genetic variability among Bacillus anthracis strains and related Ivanova N, Sorokin A, Anderson I, Galleron N, Candelon B, Kapatral V,
species. J Bacteriol 178, 377–384. Bhattacharyya A, Reznik G, Mikhailova N, Lapidus A, Chu L, Mazur
Ash C, Farrow JA, Dorsch M, Stackebrandt E, Collins MD (1991). Com- M, Goltsman E, Larsen N, D’Souza M, Walunas T, Grechkin Y, Pusch
parative analysis of Bacillus anthracis, Bacillus cereus, and related G, Haselkorn R, Fonstein M, Dusko Ehrlich S, Overbeek R, Kyrpides
species on the basis of reverse transcriptase sequencing of 16S rRNA. N (2003). Genome sequence of Bacillus cereus and comparative anal-
Int J Syst Bacteriol 41, 343–346. ysis with Bacillus anthracis. Nature 423, 87–91.
D. Volokhov et al. / Diagnostic Microbiology and Infectious Disease 49 (2004) 163–171 171

Jackson PJ, Hugh-Jones ME, Adair DM, Green G, Hill KK, Kuske CR, for real-time PCR detection of Bacillus anthracis. Appl Environ Mi-
Grinberg LM, Abramova FA, Keim P (1998). PCR analysis of tissue crobiol 67, 3720 –3727.
samples from the 1979 Sverdlovsk anthrax victims: The presence of Radnedge L, Agron PG, Hill KK, Jackson PJ, Ticknor LO, Keim P,
multiple Bacillus anthracis strains in different victims. Proc Natl Acad Andersen GL (2003). Genome differences that distinguish Bacillus
Sci U S A 95, 1224 –1229. anthracis from Bacillus cereus and Bacillus thuringiensis. Appl Envi-
Jackson PJ, Walthers EA, Kalif AS, Richmond KL, Adair DM, Hill KK, ron Microbiol 69, 2755–2764.
Kuske CR, Andersen GL, Wilson KH, Hugh-Jones M, Keim P (1997). Ramisse V, Patra G, Garrigue H, Guesdon JL, Mock M (1996). Identifi-
Characterization of the variable-number tandem repeats in vrrA from cation and characterization of Bacillus anthracis by multiplex PCR
different Bacillus anthracis isolates. Appl Environ Microbiol 63, 1400 – analysis of sequences on plasmids pXO1 and pXO2 and chromosomal
1405. DNA. FEMS Microbiol Lett 145, 9 –16.
Keim P, Price LB, Klevytska AM, Smith KL, Schupp JM, Okinaka R, Ramisse V, Patra G, Vaissaire J, Mock M (1999). The Ba813 chromosomal
Jackson PJ, Hugh-Jones ME (2000). Multiple-locus variable-number DNA sequence effectively traces the whole Bacillus anthracis com-
tandem repeat analysis reveals genetic relationships within Bacillus munity. J Appl Microbiol 87, 224 –228.
anthracis. J Bacteriol 182, 2928 –2936. Read TD, Peterson SN, Tourasse N, Baillie LW, Paulsen I.T, Nelson KE,
Laassri M, Chizhikov V, Mikheev M, Shchelkunov S, Chumakov K Tettelin H, Fouts DE, Eisen JA, Gill SR, Holtzapple EK, Oa OK,
(2003). Detection and discrimination of orthopoxviruses using microar- Helgason E, Rilstone J, Wu M, Kolonay JF, Beanan MJ, Dodson RJ,
rays of immobilized oligonucleotides. J Virol Methods 112, 67–78. Brinkac LM, Gwinn M, DeBoy RT, Madpu R, Daugherty SC, Durkin
Leppla S (2000). Anthrax toxin. In: Handbook of Experimental Pharma- AS, Haft DH, Nelson WC, Peterson JD, Pop M, Khouri HM, Radune
cology. Eds, K Aktories and I Just. Berlin, Heidelberg: Springer- D, Benton JL, Mahamoud Y, Jiang L, Hance IR, Weidman JF, Berry
Verlag, pp 445– 472. KJ, Plaut RD, Wolf AM, Watkins KL, Nierman WC, Hazen A, Cline
Liu WT, Mirzabekov AD, Stahl DA (2001). Optimization of an oligonu- R, Redmond C, Thwaite JE, White O, Salzberg SL, Thomason B,
cleotide microchip for microbial identification studies: A non-equilib- Friedlander AM, Koehler TM, Hanna PC, Kolsto AB, Fraser CM
(2003). The genome sequence of Bacillus anthracis Ames and com-
rium dissociation approach. Environ Microbiol 3, 619 – 629.
parison to closely related bacteria. Nature 423, 81– 86.
Logan NA, Carman JA, Melling J, Berkeley RC (1985). Identification of
Reif TC, Johns M, Pillai SD, Carl M (1994). Identification of capsule-
Bacillus anthracis by API tests. J Med Microbiol 20, 75– 85.
forming Bacillus anthracis spores with the PCR and a novel dual-probe
Louie M, Louie L, Simor AE (2000). The role of DNA amplification tech-
hybridization format. Appl Environ Microbiol 60, 1622–1625.
nology in the diagnosis of infectious diseases. CMAJ 163, 301–309.
Sacchi CT, Whitney AM, Mayer LW, Morey R, Steigerwalt A, Boras A,
Makino S, Watarai M, Cheun HI, Shirahata T, Uchida I (2002). Effect of
Weyant RS, Popovic T (2002). Sequencing of 16S rRNA gene: A rapid
the lower molecular capsule released from the cell surface of Bacillus
tool for identification of Bacillus anthracis. Emerg Infect Dis 8, 1117–1123.
anthracis on the pathogenesis of anthrax. J Infect Dis 186, 227–233.
Sastry KS, Tuteja U, Santhosh PK, Lalitha MK, Batra HV (2003). Iden-
Makino SI, Iinuma-Okada Y, Maruyama T, Ezaki T, Sasakawa C, Yo-
tification of Bacillus anthracis by a simple protective antigen-specific
shikawa M (1993). Direct detection of Bacillus anthracis DNA in
mAb dot-ELISA. J Med Microbiol 52, 47– 49.
animals by polymerase chain reaction. J Clin Microbiol 31, 547–551.
Schraft H, Griffiths MW (1995). Specific oligonucleotide primers for
Markoulatos P, Siafakas N, Moncany M (2002). Multiplex polymerase
detection of lecithinase-positive Bacillus spp. by PCR. Appl Environ
chain reaction: A practical approach. J Clin Lab Anal 16, 47–51. Microbiol 61, 98 –102.
Mock M, Fouet A (2001). Anthrax. Annu Rev Microbiol 55, 647– 671. Schuch R, Nelson D, Fischetti VA (2002). A bacteriolytic agent that
Mohammed MJ, Marston CK, Popovic T, Weyant RS, Tenover FC (2002). detects and kills Bacillus anthracis. Nature 418, 884 – 889.
Antimicrobial susceptibility testing of Bacillus anthracis: Comparison Tang YW, Procop GW, Persing DH (1997). Molecular diagnostics of
of results obtained by using the National Committee for Clinical Lab- infectious diseases. Clin Chem 43, 2021–2038.
oratory Standards broth microdilution reference and Etest agar gradient Thompson JD, Higgins DG, Gibson TJ (1994). CLUSTAL W: Improving
diffusion methods. J Clin Microbiol 40, 1902–1907. the sensitivity of progressive multiple sequence alignment through
Odendaal MW, Pieterson PM, de Vos V, Botha AD (1991). The biochem- sequence weighting, position-specific gap penalties and weight matrix
ical, morphological and virulence profiles of Bacillus anthracis isolated choice. Nucleic Acids Res 22, 4673– 4680.
in the Kruger National Park. Onderstepoort J Vet Res 58, 21–26. Timoshin VB, Zamaraev VS, Antonov VA, Lipnitskii AV (1994). A
Okinaka RT, Cloud K, Hampton O, Hoffmaster AR, Hill KK, Keim P, species-specific DNA probe for identifying toxic strains of anthrax
Koehler TM, Lamke G, Kumano S, Mahillon J, Manter D, Martinez Y, pathogens. Mol Gen Mikrobiol Virusol 1, 30 –33.
Ricke D, Svensson R, Jackson PJ (1999). Sequence and organization of Turnbull PC (1999). Definitive identification of Bacillus anthracis—a
pXO1, the large Bacillus anthracis plasmid harboring the anthrax toxin review. J Appl Microbiol 87, 237–240.
genes. J Bacteriol 181, 6509 – 6515. Turnbull PC, Hutson RA, Ward MJ, Jones MN, Quinn CP, Finnie NJ,
Pannucci J, Okinaka RT, Williams E, Sabin R, Ticknor LO, Kuske CR Duggleby CJ, Kramer JM, Melling J (1992). Bacillus anthracis but not
(2002). DNA sequence conservation between the Bacillus anthracis always anthrax. J Appl Bacteriol 72, 21–28.
pXO2 plasmid and genomic sequence from closely related bacteria. Volokhov D, Rasooly A, Chumakov K, Chizhikov V (2002). Identification
BMC Genomics 3, 34. of Listeria species by microarray-based assay. J Clin Microbiol 40,
Patra G, Sylvestre P, Ramisse V, Therasse J, Guesdon JL (1996). Isolation 4720 – 4728.
of a specific chromosomic DNA sequence of Bacillus anthracis and its Yamada S, Ohashi E, Agata N, Venkateswaran K (1999). Cloning and
possible use in diagnosis. FEMS Immunol Med Microbiol 15, 223–231. nucleotide sequence analysis of gyrB of Bacillus cereus, B. thuringien-
Qi Y, Patra G, Liang X, Williams LE, Rose S, Redkar RJ, DelVecchio VG sis, B. mycoides, and B. anthracis and their application to the detection
(2001). Utilization of the rpoB gene as a specific chromosomal marker of B. cereus in rice. Appl Environ Microbiol 65, 1483–1490.

You might also like