Sie sind auf Seite 1von 7

Journal of General Virology (1990), 71, 2141-2147.

Printed in Great Britain 2141

Detection and differentiation of picornaviruses in clinical samples


following genomic amplification
D. Michael Olive, l,2* t Siham Al-Mufti, 2 Wahiba Al-Mnlla, 1 M. A. Khan, 3 Alexander Pasca, 1
Glyn Stanway 3 and Widad AI-Nakib 1,2

~Department of Microbiology, Faculty of Medicine, Kuwait University, P.O. Box 24923, 13110 Kuwait, ZPublic Health
Virology Laboratory, 5540 Shaab, 13056 Kuwait and 3Department of Biology, University of Essex, Colchester C04 3SQ,
U.K.

A polymerase chain reaction (PCR) assay was used to virus, influenza virus and rotavirus, as well as human
detect and differentiate picornaviruses (PVs), using and monkey cell DNA, were not amplified. In nasal
primers homologous to the 5' non-coding and VP2 swabs taken from patients with respiratory infections,
regions of the PV genome. The P C R resulted in a the PCR detected 27 of 28 H R V isolation-positive
530 bp PCR product for human rhinoviruses (HRVs) specimens. All specimens from which viruses other
and a 650 bp product for polioviruses, coxsaCkieviruses than HRVS were isolated were negative by PCR. The
(CV) or echoviruses. The P C R assay could detect as P C R definitively identified poliovirus and CVs from
little as 1 p.f.u, of virus in either cerebrospinal fluid the CSF or stool of patients with aseptic meningitis, as
(CSF) or stool, using ethidium bromide-stained gels. well as CV in the pericardial fluid of a patient who had
Standard strains of poliovirus, CV, echovirus and suffered a myocardial infarction. Specimens taken
H R V were detected, with the exception of echovirus from patients with similar pathologies, and from which
type 22. In contrast, heterologous viruses, such as heterologous viruses were isolated, were uniformly
herpes simplex virus, human cytomegalovirus, adeno- negative by PCR.

Introduction infect the lower respiratory tract (Krilov et al., 1986;


Monto et al., 1987). In one study, 6 5 ~ of infected
Picornaviruses (PVs) are a diverse group of human individuals who were 40 or more years of age had lower
pathogens which cause a wide variety of disease respiratory tract infections due to HRVs (Monto et al.,
syndromes. In terms of clinical significance, entero- 1987). HRVs have also been isolated from a high
viruses (EVs) and human rhinoviruses (HRVs) are the percentage of children and infants hospitalized with
most clinically important genera of PVs, causing millions lower respiratory tract infections (Krilov et al., 1986).
of infections each year. Infections due to EVs range in Finally, HRVs are known to exacerbate episodes of
severity from asymptomatic to aseptic meningitis, bronchitis and asthma (Gregg, 1983). With the develop-
encephalitis, paralysis, pneumonia and myocarditis ment of drugs such as R61837 which have been recently
(Bowles et al., 1986, 1987; Easton & Eglin, 1988; Leslie et shown to suppress colds in vivo yet are highly specific for
al., 1989). Although infections of the central nervous HRVs (A1-Nakib et al., 1989), it is imperative to be able
system and heart are of greatest concern, these viruses to diagnose HRV infections rapidly.
are multi-systemic in their tissue tropisms and virulence. The broad antigenic diversity among the nearly 70
For this reason, the clinical diagnosis of EV infection can different serotypes of EVs and over 100 different
be complicated by the non-specific nature of many of the serotypes of HRVs has made immunoassays impractical
syndromes, which often makes them indistinguishable (Yolken & Torsch, 1980, 1981; Dearden & A1-Nakib,
from diseases caused by bacteria and other viruses. 1987). Nucleic acid hybridization assays have been used
HRVs are the major causative agents of the common but they lack sensitivity (Rotbart et al., 1984; A1-Nakib
cold (Couch, 1985). Although infection with HRVs is et al., 1986; Bruce et al., 1989). Despite its suboptimal
usually limited to the upper respiratory tract, they can sensitivity, tissue culture remains the mainstay of the
laboratory diagnosis of EVs and HRVs.
t Present address: GENE-TRAK Systems, 31 New York Avenue, The polymerase chain reaction (PCR; Saiki et al.,
Framingham, Massachusetts 01701, U.S.A. 1988) has been used to detect pathogen-specific nucleic

0000-9581 © 1990 SGM


2142 D. M. Olive and others

acids from viruses such as human immunodeficiency DNAsynthesis-~ ~---DNAsynthesis


virus type 1 (Kwok et al., 1987), human T-cell lympho- OL24: $'CTACTTTGG6TGTeCG-3" 3"-CCI,IACCACCACCTT0/~TGG-5" :OL68-1
tropic virus type 1 (Duggan et al., 1988), human cyto- HRV1B . . . . . *****~***** ********************
megalovirus (HCMV) (Demmler et al., 1988; Shibata et HRVI4 **************** ********c***********
al., 1988; Olive et al., 1989a, b) and PVs (Gama et al., HRV89 *** . . . . . *** . . . . * ****** . . . . . . *****c**
1988, 1989; Torgersen et al., 1989; Hyypifi et at., 1989). CAV9 *** ..... *** .... . *****************c**

However, the PCR assays which have been described for CAV21 **************** **************cG****

PVs have either not successfully differentiated EVs and CBV] **************** ********************

HRVs (Gama et aL, 1989; Hyypi/i et al., 1989) or have CBV3 **************** ***************c.G**

suffered from a lack of specificity (Hyypi/i et al., 1989).


The assays described thus far have not been tested Poliol **************** *****************G**

directly on clinical specimens containing wild-type virus.


We have therefore developed a PCR assay, using Polio3 **************** ********'***'*******

primers which detect and differentiate between EVs and Fig. 1. The sequences of the OL24 and OL68-1 primers used for
HRVs in clinical specimens on the basis of the size of the amplification are shown. The complementary sequences of known EVs
PCR product. In this report, we demonstrate the and HRVs are shown to illustrate the homology. In representing the
specificity of the assay for this group of viruses, as well as primer sequences, N is any nucleotide (A, G, C or T) and Q is either T
its ability to detect wild-type clinical isolates of EVs and or C. The sequences were derived as follows: HRV1B, Hughes et al.
(1988); HRV14, Stanway et al. (1984); HRV89, Deuchler et al. (1987);
HRVs from a variety of clinical specimens. The use of CAV9, Chang et al. (1989); CAV21, Hughes et al. (1989); CBV1,
this PCR assay for detection and differentiation of PVs Iizuka et al. (1987); CBV3, Lindberg et al. (1987), Jenkins et al. (1987);
should greatly aid in the clinical management of disease Polioviruses 1, 2 and 3, Toyoda et al. (1984). Polio 1, 2 and 3,
due to PVs. polioviruses 1, 2 and 3.

nucleic acid extraction. Vanadyl ribonucleoside complex was added to


Methods a final concentration of 10 mM. As a carrier, 1.0 ~tg of phenol-extracted
yeast tRNA was added. The samples were extracted with an equal
Clinical specimens. Either nasal swabs or throat washings were taken volume of phenol saturated with TE buffer (10 mM-Tris-HC1 pH 7-5,
from patients presenting with respiratory infections. Clinical 1 mM-EDTA). The phenol phase was back-extracted with 100 ~tl of
specimens were processed immediately by inoculation onto Ohio HeLa sterile water and the aqueous phases were combined. The aqueous
or human C16 cells (Phillpotts, 1983) for isolation of HRVs and nucleic acid was extracted three more times with phenol-TE, followed
coronaviruses respectively. HRVs were also examined for acid lability by two extractions with equal volumes of chloroform. The nucleic acid
after isolation. Influenza virus isolates were obtained following growth was precipitated by addition of 0.1 volume of sterile 3 M-sodium acetate
in embryonated eggs (Kendal et al., 1985). Cerebrospinal fluids (CSF) (pH 6.5) and 2.5 volumes of 95 ~ ethanol, followed by refrigeration at
and stool swabs were taken from patients presenting with various - 70 °C for 15 min. The nucleic acid was pelleted by centrifugation in a
symptoms possibly due to enterovirus infection. Stool swabs were microfuge for 5 min and the pellet washed once with 70~ ethanol. The
placed in 2 ml of tissue culture medium and, along with CSF pellet was dried under vacuum and dissolved in 50 ktl of sterile water.
specimens, processed for isolation of virus by inoculation onto Vero, The nucleic acid from each sample was extracted in triplicate and
HEp-2 or HeLa cells. EVs were typed using intersecting hyperimmune processed for cDNA synthesis and amplification.
serum pools A to H obtained from the World Health Organization
R e v e r s e transcription. Synthesis of cDNA was performed in a
(WHO). Standard strains of EVs and HRVs were obtained from
Dr D. A. J. Tyrrell at the MRC Common Cold Unit, Salisbury, U.K. reaction mixture containing 25 Ftl of viral nucleic acid, 8 p.l of 5 x
reverse transcriptase buffer (200 mM-Tris-HCI pH 8.3, 375 mM-KC1,
S y n t h e t i c oligonucleotide primers. The mixed primer OL68-1 50 mM-dithiothreitol, 15 m~-MgC12), each dNTP at 0.5 mM, 40 units
(5' GGTAAQTTCCACCACCANCC 3') is complementary to the RNAguard (Pharmacia), 1 pl of OL68-1 primer stock (1-0 A260
genomic sense RNA at positions 1058 to 1077 (Hughes et al., 1988) in units/ml) and 200 units of Moloney murine leukaemia virus reverse
the VP2 region of HRV-1B. The OL68-1 primer contains degeneracies transcriptase (Bethesda Research Laboratories) in a total volume of
at positions 6 and 18 (Q is either T or C; N is A, G, C or T). The primer 40 ~tl. The reaction was incubated at 37 °C for 1 h and was terminated
OL24 (5' CTACTTTGGGTGTCCG 3') is complementary to anti- by the addition of 4 I-tlof 3 ~-sodium acetate pH 6.5 and 100 I.d of 95
genomic sense RNA at positions 542 to 567 (Hughes et al., 1988) in the ethanol. The mixtures were refrigerated at - 7 0 °C for 15 min, pelleted
5'-non-coding region of HRV-1B. The degree of homology exhibited and the cDNA was dissolved in 50 I.d of TE buffer.
by the primers to known EV and HRV sequences is shown in Fig. 1
P o l y m e r a s e chain reaction. For PCR, 15 p.1 of the eDNA was
(M. A. Khan & G. Stanway, unpublished results). The alkaline
combined with 10 I.tlof 10 x Taq salts (500 mM-KC1, 100 mM-Tris-HC1
phosphatase-labelled oligonucleotide probe, PV. 1 (5' AGATTGT-
pH 8.3, 25 mM-MgC12, 0.1 ~ gelatin), 10 ~tl of 8 mM-dNTP stock (2 mM
TATCATAAAGCGA 3'), for detection of poliovirus PCR products,
each dNTP), 2 ~tl each of OL24 and OL68-1 primer stocks (1.0 A z 6 o
was homologous to nucleotides 607 to 626 of poliovirus type I (Toyoda
units/ml each), 60 lal of water and 2.5 units of T h e r m u s a q u a t i c u s DNA
et al., 1984). All oligonucleotides were purchased from Genetic
polymerase (Perkin-Elmer Cetus). The mixture was amplified by 30
Designs.
successive cycles of heating at 94 °C for 2 min, renaturing at 50 °C for
E x t r a c t i o n o f viral nucleic acids. A 100 ~tl portion of either a nasal 2 min and polymerizing at 72 °C for 3 min. A further unit of enzyme
swab in transport medium, CSF or stool suspension was used for was added after 15 cycles. The PCR products were analysed by
Genomic amplification o f picornaviruses 2143

1 2 3 4 5 6 7 8 9 1 2 3 4 5 6 7 8 9 10 11

738 b p - -
615 b p - -

738 b p - -
615 bp--

492 b p - -

Fig. 3. The PCR reaction was tested for cross-reactivity with a variety
of heterologous viruses and cell types. Lane 1, marker DNA; lane 2,
poliovirus type 1; lane 3, HSV; lane 4, HCMV; lane 5, Ad2; lane 6,
Ad5; lane 7, rotavirus; lane 8, HeLa cell DNA; lane 9, HEp-2 cell
DNA; lane 10, Vero cell DNA; lane 11, marker DNA.

electrophoresis on composite agarose gels (3% Nusieve agarose, 1%


HGT agarose). DNA bands were visualized following staining with
ethidium bromide and exposure to u.v. light. Positive controls for the
PCR reaction consisted of cloned cDNAs from poliovirus type 1 or
HRV-14.
Hybridization with an alkaline phosphatase-labelled probe. Portions
(50 ~tl) of the products of the PCR reactions were combined with 50 pl
of 0-6 M-NaOH and incubated at room temperature for 15 min. The
Fig. 2. The PCR products resulting from amplification of some of the denatured DNA was applied to a nylon filter using a filtration manifold
clinical specimens in Tables 2 and 3 are shown. Lane 1, marker DNA; and the samples were neutralized by washing each well with 100 pl of
lane 2, CSF containing vaccine strain poliovirus type 2 (meningitis); 2 M-ammonium aceta~.e. The filters were heated in a vacuum oven for
lane 3, CSF containing vaccine strain poliovirus type 2 (meningitis); 2 h. The filters were prehybridized for 15 min at 45 °C in buffer
lane 4, CSF containing CBV3 (meningitis); lane 5, stool containing containing 5 x SSC (1 × is 0.15 M-NaC1, 0-015 M-sodium citrate), 1%
CBV5; lane 6, pericardial fluid containing CBV2; lane 7, nasal swab SDS and 0-5% bovine serum albumin. The filters were hybridized for
containing wild-type HRV; lane 8, nasal swab containing wild-type 15 min at 45 °C in the same buffer containing 50 pl of a 0.4 ~tMstock of
HRV; lane 9, marker DNA. probe. The filters were washed twice with 100 ml of 1% SDS in 1 x SSC
T a b l e 1. Amplification of standard virus strains at 45 °C, followed by two washes in 100 ml each of 1% Triton X-100 in
1 x SSC at 45 °C. Finally, the filters were washed in 1 × SSC at room
Virus PCR Virus PCR temperature.
strain* result strain* result The washed filters were immersed in 7.5 ml of alkaline phosphatase
buffer (100 mM-Tris HC1 pH 8.5, 100 mM-NaC1, 100 mM-ZnCI2)
HRV2 + Echo2 + containing 33pl of Nitro Blue tetrazolium solution (75mg/ml
HRV9 + Echo3 + in dimethylformamide) and 25~tl of 5-bromo-4-chloro-3-indolyl
HRV14 + Echo4 + phosphate solution (50 mg/ml in dimethylformamide). The colour
Polio 1 + Echo6 + reaction was allowed to proceed for 4 h and terminated by washing in
Polio2 + Echo8 + distilled water.
Polio3 + Echo9 +
CAV1 + Echol l +
CAV9 + Echol6 +
CAV10 + Echol9 + Results
CAV 16 + Echo22 -
CAV21 + Echo25 + Detection o f standard virus strains
CAV24 + HSV -
CBV1 + HCMV - Standard strains of EVs and HRVs were amplified, and
CBV2 + Ad2 - t h e P C R p r o d u c t s w e r e d e t e c t e d b y gel e l e c t r o p h o r e s i s .
CBV3 + Ad5 -
CBV4 + Rotavirus - A s s h o w n i n F i g . 2, t h e E V s t e s t e d g a v e r i s e t o a
CBV5 + HeLa - characteristic PCR product approximately 650 bp in
CBV6 + HEp-2 - length. In contrast, HRV strains gave rise to a PCR
Echol + Vero -
p r o d u c t o f a p p r o x i m a t e l y 530 b p , u s i n g t h e O L 2 4 / 6 8 - 1
* Abbreviations: Poliol, poliovirus type 1 ; Polio2, poliovirus type 2; primers. Table 1 summarizes the results of the PCR assay
Polio3, poliovirus type 3; CAV, coxsackievirus A; Echo, echovirus. on standard strains of HRVs, polioviruses, coxsackie-
2144 D. M. Olive and others

1 2 3 4 5 6 7 8 9 10 Table 2. Detection of HR Vs from nasal swabs taken from


patients with respiratory infections

738 Virus No. of No. positive


615 isolated specimens by PCR
HRV 28 27
CV229E* 7 0
Influenza A 4 0
RSVp" 2 0

* CV229E, Coronavirus 229E.


t The diagnosisof RSV infection was based on a positive result in an
enzyme immunoassay rather than virus isolation.

was applied to 28 specimens taken from patients with


Fig. 4. The sensitivityof the PCR was determined by detecting known respiratory infections from whom H R V s had been
amounts of poliovirusin either stool suspensionor CSF. Seriallydiluted isolated (Table 2). H R V s were detected in 27 of these
virus was added to 100 ~tlof either stool or CSF and processed for virus patients by PCR. In cases where similar respiratory
detection. Lanes 2 to 5 show the results of the titration in stool, whereas infections were caused by coronaviruses, influenza virus
lanes 7 to 10 show the resultsof the CSF titration. The samplesare: lane
type A or respiratory syncytial virus (RSV), the P C R
1, marker DNA; lane 2, 125 p.f.u.; lane 3, 25 p.f.u.; lane 4, 5 p.f.u.;
lane 5, 1 p.f.u.; lane 6, marker DNA; lane 7, 125 p.f.u.; lane 8, assay was uniformly negative.
25 p.f.u.; lane 9, 5 p.f.u.; lane 10, 1 p.f.u.
Detection of enteroviruses from clinical specimens
viruses (CV) and echoviruses, as well as several unrelated
viruses and cell lines. With one exception, the character- EVs are known to be causative agents of aseptic
istic P C R product was seen for all of the PVs tested. meningitis and encephalitis (Melnick, 1985). During the
Echovirus 22, however, did not amplify using this assay. early acute phase of infection, infectious virus can
In contrast to the P C R assay previously reported by sometimes be isolated from the CSF whereas, in the later
Hyypi~i et al. (1989), the OL24 and OL68-1 primers did stages of infection, virus can also be isolated from stool.
not give rise to spurious amplification products when In cases of myocarditis, however, virus is rarely isolated,
tested on herpes simplex virus (HSV), H C M V , adeno- presumably due to low numbers of infectious particles
virus (Ad), rotavirus, H e L a cell D N A , HEp-2 cell D N A (Leslie et al., 1989). Specimens taken from patients
or Vero cell D N A (Fig. 3). Only PVs were amplified and hospitalized for confirmed EV infections were analysed
detected. by P C R along with specimens taken from patients with
similar pathologies which were either verified or
suspected of being caused by infection with viruses other
Detection limits of the PCR assay
than PVs (Table 3). In all cases, the characteristic 650 bp
In order to determine the amount of virus which could be P C R product was detected after amplification of
detected in clinical samples, a titrated stock of poliovirus patients suffering from EV-mediated disease. The
was serially diluted and added to a negative stool or CSF procedure used in this study was suitable for a variety of
specimen. The viral nucleic acid was then extracted, clinical specimens, including stool, CSF and pericardial
reverse-transcribed and amplified. Fig. 4 shows that fluid. Cases of aseptic meningitis caused by either CVs or
1 p.f.u, of virus could be detected on ethidium bromide- the vaccine strain of poliovirus 2 were clearly detected
stained agarose gels regardless of the nature of the (Fig. 2). In two other patients with aseptic meningitis,
clinical specimen. Previous studies have reported com- CVs were detected in stool cultures (Fig. 2). Significant-
parable sensitivities using titrated virus stocks ( G a m a et ly, coxsackievirus B2 (CBV2) isolated from the pericard-
al., 1989; Torgersen et al., 1989). ial fluid of a patient who had suffered a myocardial
infarction was also detected by P C R assay (Fig. 2). The
P C R was unreactive with specimens taken from patients
Detection of HR V in patients with respiratory infections
with similar pathologies containing non-PV viral agents.
Nucleic acids from either nasal swabs immersed in tissue
culture medium or nasal washings were extracted and
Hybridization with a poliovirus-specific probe
subjected to P C R analysis. As shown in Fig. 2, the
characteristic 530 bp P C R product was detected from The identity of poliovirus P C R products was verified by
patients infected with wild-type HRVs. The P C R assay hybridization to an alkaline phosphatase-labelled oligo-
Genomie amplification o f picornaviruses 2145

Table 3. Detection of enteroviruses from clinical specimens 1 2 3 4 5

Virus PCR
Specimen Patient diagnosis isolated result

CSF Meningitis CBV3 + [


CSF Meningitis CBV3 +
PCF* MI~ CBV2 +
Stool Meningitis CBV5 + Fig. 6. Identification of poliovirus PCR products by hybridization
Stool Meningitis CBV 1 + with the PV. 1 probe. The samples are derived from amplification of:
CSF Meningitis CBV5 + 1, poliovirus type 2, aseptic meningitis; 2, CBV3, aseptic meningitis; 3,
CSF Meningitis Polio2:~ +
CSF Meningitis Polio2:~ + poliovirus type 1; 4, poliovirus type 2; 5, poliovirus type 3. The PCR
CSF Encephalitis HSV - products for the aseptic meningitis samples are shown in Fig. 2, lanes 3
CSF Encephalitis HSV - and 4.
CSF SSPE§ None -
CSF SSPE None - shows considerable m i s m a t c h with the sequences o f
CSF SSPE None - other characterized EVs. The identity of the P C R
CSF Multiple sclerosis Nonell -
product obtained by amplification of the C S F material
* Pericardial fluid. from a patient with aseptic meningitis due to poliovirus
~"Myocardial infarction. type 2, was confirmed with the P V . 1 probe (Fig. 6). I n
Vaccine strain. contrast, a similar case of meningitis caused by CBV3
§ The diagnosis of subacute sclerosing panencephalitis (SSPE) was
based on high titres of anti-measles antibody in both serum and CSF. was unreactive with the P V . 1 probe (Fig. 6). The P V . 1
IIThe patient specimen was sent for investigation into possible viral probe did not detect any o f the other EVs tested,
causes. presumably owing to the heterogeneity o f this region
a m o n g EVs (unpublished data).

607 626
Discussion

5" - A C - , ~ T T G T T A T C A T ~ G C ~ - 3 " Efforts to detect and differentiate PVs directly f r o m


clinical samples by using nucleic acid hybridization
Poliol ~ ~ ~ ~ techniques have been largely unsuccessful. Bruce et al.
(1989) have shown that, although radioactively labelled
Polio2 ~ ~ ~ ~ synthetic oligonucleotide probes homologous to the 5'
untranslated region of H R V s can be used to detect most
Polio3 ~ ~ ~ ~ k n o w n serotypes, the sensitivity can vary significantly
depending on the viral isolate. In addition, the radioac-
cBvl **G******C****T**TT* tive label used is unsuitable for routine usage. Alkaline
phosphatase-labelled probes have been used as an
CBV3 -~ -~ ~- * -~ -x.~ .w--~ C -I~-~ -I( -~ -~ ~ ,I~-~ w-T alternative but are even less sensitive (Rotbart et al.,
1988). More recently, Petitjean et al. (1990) have used
CBV4 * * * * * * * * * C * * * * T * * * T * radioactively labelled riboprobes homologous to the 5'
non-coding region o f EVs. A l t h o u g h virus could be
CAV9 * * * * * * * * * C * * * * T * * * T * detected in most stool specimens, the detection o f EVs in
C S F again showed low sensitivity.
CAV21 C-¢~******* C C * * * T * * * T * The P C R assays described thus far have been unable
to give a simple, clear differentiation between EVs and
Fig. 5. The DNA sequence of the poliovirus detection probe, PV. 1.
The sequences of known EVs are shown to illustrale homology. The H R V s , nor have they been applied directly to clinical
sources of the sequence information are those mentioned in Fig. 1. samples ( G a m a et al., 1989; Torgersen et al., 1989;
Nucleotide numbers are indicated above the sequence. Hyypi~i et al., 1989). The specificity of antiviral drugs for
treatment of respiratory disease makes it imperative to
nucleotide probe ( P V . 1 ) specific for a conserved be able to detect and differentiate infections caused by
sequence o f the 5'-non-coding region o f polioviruses (Fig. H R V s and EVs directly on clinical specimens since
5). This sequence is not found in H R V s . The P V . 1 probe severe respiratory infections can be caused by both viral
hybridized specifically to P C R products obtained by groups. In addition, acute diseases such as aseptic
amplification of poliovirus types 1, 2 and 3 (Fig. 6). As meningitis or encephalitis require rapid diagnosis for
can be seen in Fig. 5, however, the P V . 1 probe sequence effective clinical m a n a g e m e n t .
2146 D. M. Olive and others

Therefore, we have developed a PCR assay which products of the expected size for almost every HRV or
detects and differentiates PVs directly from clinical EV tested.
specimens. The OL24 primer sequence is derived from a By applying the PCR assay to respiratory illness, 27 of
highly conserved region of the 5' non-coding region of the 28 wild-type HRVs isolated from nasal swabs were
PV genome. The OL68-1 primer is built around a highly correctly identified. The single HRV not identified
conserved stretch of three tryptophan codons (UGG) probably had a variant nucleic acid sequence at the site
found in the VP2 region of the PV genome. Using these of one of the primers. Similarly, EV isolation-positive
primers, we have been able to exploit the observation CSF fluids and stool specimens taken from patients with
that the 5' non-coding regions of EVs are approximately aseptic meningitis gave rise to the EV-specific 650 bp
120 bp longer than that of HRVs. PCR product. None of the pathologically similar cases
The PCR assay detected and differentiated all from which heterologous viruses had been isolated were
serologically identified EVs or HRVs tested with the amplified.
exception of echovirus type 22. The unreactivity of Significantly, CBV2 was detected in the pericardial
echovirus type 22 in our assay is not surprising, however, fluid of a patient who had suffered a myocardial
since other studies which have tested probes for PV infarction. CVs have long been suspected of playing a
detection have been uniformly unreactive with echovirus significant role in myocarditis, but definitive data have
type 22 (Auvinen et al., 1989; Hyypi~i et al., 1989). been lacking due to the low recovery rates of virus from
Auvinen et al. (1989) have shown that the nucleotide pericardial fluids (Melnick, 1985; Leslie et al., 1989). The
sequence of echovirus type 22 diverges significantlyfrom ability to detect virus in pericardial fluid using the PCR
that of other PVs. Presumably this is the cause of the assay should facilitate a better definition of the role
unreactivity of echovirus type 22 in our and other PCR played by these viruses in cardiac disease.
and hybridization assays. In summary, even in the absence of sequence
Previously, PCR sensitivity has been determined on confirmation, we conclude that the assay is specific for
viral stocks rather than clinical samples. We have shown PVs and, in addition, will differentiate infection by EVs
here, however, that as little as 1 p.f.u, of virus could be and HRVs. The PCR assay described here is also rapid
detected in CSF or stool samples. This level of sensitivity and can be completed in a single day. Indeed, the rapid
is sufficient for clinical application. The conditions detection and differentiation of PVs using the PCR assay
described here for nucleic acid extraction, reverse should allow better clinical management of PV-mediated
transcription and amplification were suitable for all disease.
types of clinical samples tested.
This work was supported by Kuwait University grant number
For detection of PV-mediated infections, we have
MI063.
relied primarily on the electrophoretic detection of
specific sizes of PCR products without further identifi-
cation by hybridization, restriction enzyme digestion or References
DNA sequencing. Although we have been able to use a AL-NAKIB,W., STANWAY,G., FORSYTH,M., HUGHES,P. J., ALMOND,
probe for identity confirmation of poliovirus PCR J. W. & TYRRELL,D. A. J. (1986). Detection of human rhinoviruses
products, this technique is not generally applicable to PV and their molecular relationships using cDNA probes. Journal of
Medical Virology 20, 289-296.
PCR confirmation as much of the region of the viral AL-NAKIB, W., HIGGINS, P. G., BARROW, G. I., TYRRELL, D. A. J.,
genome flanked by the OL24/OL68-1 primers shows ANDRIES,K., VANDENBUSSCHE,G., TAYLOR,N. & JANSSEN,P. A. J.
significant variability among PVs. Torgersen et al. (1989). The suppression of colds in human volunteers challenged
with rhinovirus by a new synthetic drug (R61837). Antimicrobial
(1989) have used restriction enzyme digestion patterns of Agents and Chemotherapy 33, 522-525.
PCR products obtained after amplification as a means of AUVINEN,P., STANWAY,G. & HYYPL~,T. (1989). Genetic diversity of
serotyping HRVs. Owing to the large number of PVs, as enterovirus subgroups. Archives of Virology 104, 175-186.
BOWLES,N. E., OLSEN,E. G. J., RICHARDSON,P. J. & ARCHARD,L. C.
well as the high degree of sequence variability in the (1986). Detection of coxsackie-B-virus specific RNA sequences in
region amplified in our assay, mapping with restriction myocardial biopsy samples from patients with myocarditis and
enzymes would be impractical on unknown virus dilated cardiomyopathy. Lancet i, 1120-1123.
BOWLES, N. E., SEWRY, C. A., DUBOWITZ, V. & ARCHARD, L. C.
isolates. Furthermore, owing to the limited amount of (1987). Dermatomyositis, polymyositis, and coxsackie-B-virus infec-
sequence information available for the more than 170 tion. Lancet i, 1004-1007.
serotypes of PVs, it is likely that not even DNA BRUCE, C. B., AL-NAKIB, W., ALMOND,J. W. & TYRRELL, D. A. J.
(1989). Use of synthetic oligonucleotide probes to detect rhinovirus
sequencing would definitively identify the PCR products RNA. Archives of Virology 105, 179-187.
as PV-derived. Despite this limitation, we have not CHANG, K. H., AUVlNEN,P., HYYPI.~,T. & STANWAY,G. (1989). The
observed spurious amplification of any of the heter- nucleotide sequence of coxsackievirus A9; implications for receptor
binding and enterovirus classification. Journal of General Virology
ologous samples examined and the PCR assay resulted in 70, 3269-3280.
Genomic amplification of picornaviruses 2147

COUCH, R. B. (1985). Rhinoviruses. In Virology,pp. 795-816. Edited by LINDBERG, A. M., STALHANDSKE, P. O. K. & PETTERSON, U. (1987).
B. N. Fields, D. M. Knipe, R. M. Chanock, J. L. Melnick, Genome of coxsackievirus B3. Virology 156, 50-63.
B. Roizman & R. E. Shope. New York: Raven Press. MELNICK, J. L. (1985). Enteroviruses: polioviruses, coxsackieviruses,
DEARDEN, C. & AL-NAK1B, W. (1987). Direct detection of rhinoviruses echoviruses, and newer enteroviruses. In Virology, pp. 739-794.
by an enzyme-linked immunosorbent assay. Journal of Medical Edited by B. N. Fields, D. M. Knipe, R. M. Chanock, J. L. Melnick,
Virology 23, 179-189. B. Roizman & R. E. Shope. New York: Raven Press.
DEMMLER, G. J., BUFFONE, G. J., SCHIMBOR, C. M. & MAY, R. A. MONTO, A. S., BRYAN, E. R. & OHMIT, S. (1987). Rhinovirus infections
(1988). Detection of cytomegalovirus in urine from newborns by in Tecumsah, Michigan: frequency of illness and number of
using the polymerase chain reaction DNA amplification. Journal of serotypes. Journal of Infectious Disease 156, 43 49.
Infectious Disease 158, 1177-1184. OLIVE, D. M., SIMSEK,M. & AL-MUFTI, S. (1989a). Polymerase chain
DEUCHLER, M., SKERN, T., SOMMERGRUBER, W., NEUBAUER, C., reaction assay for detection of human cytomegalovirus. Journal of
GRUENDLER, P., FOGY, I., BLAAS, D. & KUECHLER, E. (1987). Clinical Microbiology 27, 1238 1242.
Evolutionary relationships within the human rhinovirus genus: OLIVE, D. M., AL-MUFTI, S., SIMSEK, M., FAYEZ, H. & AL-NAKIB, W.
comparison of serotypes 89, 2, and 14. Proceedings of the National (1989b). Direct detection of human cytomegalovirus in urine
Academy of Sciences, U.S.A. 84, 2605 2609. specimens from renal transplant patients following polymerase chain
DUGGAN, D. B., EHRLICH, G. D., DAVEY, F. P., KWOK, S., SNINSKY, J., reaction amplification. Journal of Medical Virology 29, 232-237.
GOLDBERG, J., BALTRUCKI, L. & POIESZ, B. J. (1988). HTLV- PETITJEAN, J., QUIBRIAC,M., FREYMUTH,F., FUCHS, F., LACONCHE,N.,
I-induced lymphoma mimicking Hodgkin's disease. Diagnosis by AYMARD, M. & KOPECKA, H. (1990). Specific detection of
polymerase chain reaction amplification of specific HTLV-I enteroviruses in clinical samples by molecular hybridization using
sequences in tumor DNA. Blood, 71, 1027-1032. poliovirus subgenomic riboprobes. Journal of Clinical Microbiology
EASTON, A. J. & EGLIN, R. P. (1988). The detection of coxsackievirus 28, 307 31l.
RNA in cardiac tissue by in situ hybridization. Journal of General PHILLI'OTTS, R. J. (1983). Clones of MRC-C cells may be superior to the
Virology 69, 285 291. parent line for the culture of 229-E-like strains of human respiratory
GAMA, R. E., HUGHES, P. J., BRUCE, C. B. & STANWAY, G. (1988). coronavirus. Journal of Virological Methods 6, 267-269.
Polymerase chain reaction amplification of rhinovirus nucleic acids ROTBART, H. A., LEVIN, M. J. & VILLAREAL, L. P. (1984). Use of
from clinical material. Nucleic Acids Research 16, 9346. subgenomic poliovirus DNA hybridization probes to detect the
GAMA, R. E., HORSNELL, P. R., HUGHES, P. J., NORTH, C., BRUCE, major subgroups of enteroviruses. Journalof Clinical Microbiology 20,
C. B., AL-NAKIB, W. & STANWAY, G. (1989). Amplification of 1105-1108.
rhinovirus specific nucleic acids from clinical material using the ROTBART, H. A., EASTMAN,P. S., RUTH, J. L., HIRATA, K. A. & LEVIN,
polymerase chain reaction. Journal of Medical Virology 28, 73-77. M. J. (1988). Nonisotopic oligomeric probes for the human
GREGG, I. (1983). Provocation of airflow limitation by viral infection: enteroviruses. Journal of" Clinical Microbiology 26, 2669-2671.
implications for treatment. European Journal of Respiratory Disease SAIKI, R. K., GELFAND, D. H., STOFFEL, S., SCHARF,S. J., HIGUCHI, R.,
64, 369-379. HORN, G. T., MULLIS, K. B. & EHRLICH, H. A. (1988). Primer-
HUGHES, P. J., NORTH, C., JELLIS, C. H., MINOR, P. D. & STANWAY,G. directed enzymatic amplification of DNA with a thermostable DNA
(1988). The nucleotide sequence of human rhinovirus 1B: molecular polymerase. Science 239, 487 491.
relationships within the rhinovirus genus. Journalof General Virology SHIBATA, D., MARTIN, W. J., APPLEMAN, M. D., CAUSEY, D. M.,
69, 49-58. LEEDOM, J. M. & ARNHEIM, N. (1988). Detection of cytomegalovirus
HUGHES, P. J., NORTH, C., MINOR, P. D. & STANWAY, G. (1989). The DNA in peripheral blood of patients infected with human
complete nucleotide sequence of coxsackievirus A21. Journal of immunodeficiency virus. Journal of Infectious Disease 158,
General Virology 70, 2943-2952. 1185-1192.
HYYPI.~, T., AUVINEN, P. & MAARONEN, M. (1989). Polymerase chain STANWAY, G., HUGHES, P. J., MOUNTFORD, R. C., MINOR, P. D. &
reaction for human picornaviruses. Journal of General Virology 70, ALMOND, J. W. (1984). The complete sequence of a common cold
3261-3268. virus: human rhinovirus 14. Nucleic Acids Research 12, 7859 7875.
IIZUKA, N., KUGE, S. & NOMOTO, A. (1987). Complete nucleotide TORGERSEN, H., SKERN, T. & BLAAS, D. (1989). Typing of human
sequence of the genome of coxsackievirus B1. Virology 156, 64 73. rhinoviruses based on sequence variations in the 5' non-coding
JENKINS, O., BOOTH, J. D., MINOR, P. D. & ALMOND, J. W. (1987). The region. Journal of General Virology 70, 3111-3116.
complete nucleotide sequence of coxsackievirus B4 and its compari- TOYODA, M., KOHARA, M., KATAOKA,Y., SUGANUMA,T., OMATA,T.,
son to other members of the Picornaviridae. Journal of General IMURA, N. & NOMOTO, A. (1984). Complete nucleotide sequences of
Virology 68, 1835 1848. all three poliovirus serotype genomes: implications for genetic
KENDAL, A. P., DOWDLE, W. R. & NOBLE, G. R. (1985). Influenza relationship, gene function, and antigenic determinants. Journal of
viruses. In Manual of Clinical Microbiology, 4th edn., pp. 755-762. Molecular Biology 174, 561-585.
Edited by E. H. Lennette, A. Batows, W. J. Hausler & H. J. YOLKEN, R. H. & TORSCH, V. M. (1980). Enzyme-linked immuno-
Shadomy. Washington, D.C. : American Society for Microbiology. sorbent assay for detection and identification of coxsackie-B antigen
KRILOV, L., PIERLIK, L., KELLER, E., MAHAN, K., WATSON, D., in tissue culture and clinical specimens. Journalof Medical Virology
HIRSCH, M., HAMPARIAN, V. & MCKINTOSH, K. (1986). The 6, 45 52.
association of rhinoviruses with lower respiratory tract disease in YOLKEN, R. H. & TORSCH, V. M. (198l). Enzyme-linked immuno-
hospitalized patients. Journal of Medical Virology 19, 345-352. sorbent assay for detection of coxsackie-A viruses. Infection and
KWOK, S., MACK, D. H., MULLIS, K. B., POIESZ, B., EHRLICH, H. A., Immunity 31, 742 750.
BLAIR, D., FRIEDMAN-KILN, A. & SNINSKY, J. (1987). Identification
of human immunodeficiency virus sequences by using in vitro
enzymatic amplification and oligomer cleavage detection. Journalof
Virology 61, 1690 1694.
LESLIE, K., BLAY, R., HAISCH, C., LODGE, A., WELLER, A. & HUBER, S.
(1989). Clinical and experimental aspects of viral myocarditis.
Clinical Microbiology Reviews 2, 191-203. (Received 20 March 1990; Accepted 4 June 1990)

Das könnte Ihnen auch gefallen