Download Susceptibility of coxsackievirus B3 laboratory strains and clinical

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Discovery and development of proton pump inhibitors wikipedia , lookup

Discovery and development of non-nucleoside reverse-transcriptase inhibitors wikipedia , lookup

Discovery and development of direct Xa inhibitors wikipedia , lookup

Discovery and development of integrase inhibitors wikipedia , lookup

Discovery and development of neuraminidase inhibitors wikipedia , lookup

Discovery and development of ACE inhibitors wikipedia , lookup

Transcript
Journal of Antimicrobial Chemotherapy (2005) 56, 648–656
doi:10.1093/jac/dki263
Advance Access publication 8 September 2005
Susceptibility of coxsackievirus B3 laboratory strains and clinical
isolates to the capsid function inhibitor pleconaril: antiviral studies
with virus chimeras demonstrate the crucial role of
amino acid 1092 in treatment
Michaela Schmidtke1*, Elke Hammerschmidt1, Susanne Schüler1, Roland Zell1,
Eckhard Birch-Hirschfeld1, Vadim A. Makarov2, Olga B. Riabova2 and Peter Wutzler1
1
Institute of Virology and Antiviral Therapy, Medical Centre of the Friedrich Schiller University Jena,
Hans Knoell Str. 2, D-07740 Jena, Germany; 2Research Center of Antibiotics, Nagatinskaya str. 3a,
117105, Moscow, Russia
Received 21 April 2005; returned 11 June 2005; revised 23 June 2005; accepted 25 June 2005
Objectives: At present, most promising compounds to treat enterovirus-induced diseases are broadspectrum capsid function inhibitors which bind into a hydrophobic pocket in viral capsid protein 1
(VP1). Coxsackievirus B3 (CVB3) Nancy was the only prototypic enterovirus strain shown to be
pleconaril-resistant. This study was designed to better understand the polymorphism of the hydrophobic
pocket in CVB3 laboratory strains and clinical isolates and its implications for treatment with the capsid
function inhibitor pleconaril.
Methods: Pleconaril susceptibility was determined in cytopathic effect-inhibitory, plaque reduction or
virus yield assays. Sequence analysis of the genome region coding for VP1 and/or subsequent
alignment of amino acids lining the hydrophobic pocket of five CVB3 laboratory strains and 20 clinical
isolates were carried out. Virus chimeras and computational analysis were used to prove the role of amino
acid 1092.
Results and conclusions: Despite high conservation of pocket amino acids, polymorphism was detected
at positions 1092, 1094 and 1180. Neither Pro-1094!Thr nor Val-1180!Ile altered efficacy of pleconaril
treatment. But the amino acid at position 1092 was strongly associated with susceptibility of CVB3 to
the capsid inhibitor. Whereas leucine was involved in resistance, isoleucine and valine were detected in
pleconaril-susceptible CVB3. Results from antiviral assays with hybrid viruses demonstrate the crucial role
of amino acid 1092 in pleconaril susceptibility. A resistant cDNA-generated CVB3 became pleconaril-susceptible after accepting parts from the genome region encoding Ile-1092 into its capsid. Computational
analysis suggests that conformational changes in the hydrophobic pocket occur when leucine is substituted for isoleucine or valine and that this change leads to susceptibility to pleconaril.
Keywords: enterovirus, sequence, capsid inhibitor
Introduction
Coxsackievirus B3 (CVB3) is one of 70 human serotypes of the
genus enterovirus of the Picornaviridae. This virus family includes
small, non-enveloped, single-stranded positive-sense RNA viruses.
The viral RNA is protected by a capsid. Like other enteroviruses,
CVB3 may cause severe acute and chronic diseases, e.g.
encephalitis, meningitis, pancreatitis, gastroenteritis, hand, foot
and mouth syndrome, respiratory diseases and non-specific febrile
illnesses.1–4 Furthermore, it is the most important agent of virusinduced acute and chronic myocarditis.5–7 At present, the treatment
of these diseases is primarily symptomatic.8 There is a considerable
interest in a broad-spectrum antiviral compound for the treatment
of enteroviral disease for two reasons: firstly, because of the large
.............................................................................................................................................................................................................................................................................................................................................................................................................................
*Corresponding author. Tel: +49-3641-657222; Fax: +49-3641-657202; E-mail: [email protected]
.............................................................................................................................................................................................................................................................................................................................................................................................................................
648
The Author 2005. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy. All rights reserved.
For Permissions, please e-mail: [email protected]
Crucial role of amino acid 1092 in anti-CVB3 activity of pleconaril
JAC
antiviral
number of enterovirus serotypes and secondly, because no single
enterovirus serotype is exclusively associated with any particular
disease.
Based on the knowledge of virus capsid structure, broadspectrum antiviral compounds targeting the viral capsid were
developed. The structure of the CVB3 capsid has been solved at
3.5 Å resolution by Muckelbauer et al.9 The CVB3 capsid was
shown to be very similar to that of other members of the Picornaviridae, e.g. poliovirus and rhinovirus 14.10,11 The icosahedral
capsid consists of 60 identical copies each composed of four
structural proteins, designated as VP1, VP2, VP3 and VP4. The
five-fold axis is surrounded by a large depression, the so-called
canyon. At the base of the canyon, a surface-accessible hydrophobic pocket has been found within VP1. This pocket may be
occupied by a pocket factor, which is suggested to be a lipid or
a fatty acid. According to the hypothesis of Rossmann et al.,12
specific cell surface molecules, the receptors, interact with the
canyon floor. Receptor binding induces both the release of the
pocket factor and conformational changes in the capsid which
again allow the viral RNA to enter host cells. This step of the
viral life cycle can be blocked by capsid inhibitors.13,14 Capsid
inhibitors may replace the pocket factor, occupy the hydrophobic
pocket and prevent conformational changes in the viral capsid
necessary for successful virus attachment to cell receptors and/
or uncoating of viral RNA. Various capsid-binding agents were
developed.15 But complex drug metabolism, serious side effects
or a narrow spectrum of activity hindered their clinical use.
Pleconaril, a novel capsid function inhibitor, is considered to be
a potential broad-spectrum anti-picornavirus agent because it
inhibits the replication of most serotypes of rhinoviruses as well
as enteroviruses at nanomolar and micromolar concentrations.16,17
CVB3 Nancy was detected as the only pleconaril-resistant prototypic enterovirus strain.17 However, the molecular base of this
resistance was not studied. Later on, resistant CVB3 were generated from a pleconaril-susceptible wild-type CVB3 grown in the
presence of 1 mg/mL of pleconaril in vitro.18 The resistant mutants
contain amino acid substitution Ile-1092!Leu or Ile-1092!Met
in the hydrophobic pocket in VP1. Therefore, it was suggested
that these amino acid substitutions may be responsible for the
observed resistance of these viruses. This hypothesis was not
proven until now.
This study was conducted to gain a better understanding of the
amino acid polymorphism of the hydrophobic pocket of CVB3
laboratory strains and clinical isolates as well as implications of
amino acid substitutions within the pocket for antiviral therapy
with capsid inhibitors. The nucleotide sequence of the genome
region coding for amino acids lining the hydrophobic pocket
was determined and compared with virus susceptibility to pleconaril. Antiviral investigations with hybrid viruses were applied
to demonstrate the crucial role of amino acid 1092 in pleconaril
susceptibility. Additionally, computational modelling was used to
study conformational changes within the hydrophobic pocket as
the result of amino acid substitutions.
Materials and methods
Cell lines and viruses
HeLa cells and primary human fibroblast line H (HuFi19) were grown
in Eagle minimal essential medium (MEM) supplemented with 10%
neonatal calf serum (NCS). CHO-K1 cells (DSZM no. ACC-10) were
maintained with Dulbecco’s minimal essential medium (DMEM)
supplemented with 10% FBS. Penicillin (100 U/mL) and 100 mg/mL
of streptomycin were added to all three cell lines. The maintenance
medium of HeLa cells (human cervix carcinoma; ATCC no: CCL-2)
contained 2% NCS.
The laboratory virus strains used in this study were CVB3 Nancy
(Institute of Poliomyelitis and Virus Encephalitides, Moscow, Russia),
CVB3 M2 (cDNA generated from plasmid pCVB3-M2 consisting of
cDNA of CVB3 Nancy20), CVB3 HA (ATCC: VR-688), CVB3 H3,21
CVB3 P19 and CVB3 PD.22 Furthermore, CVB3 Bgl/Spe, CVB3 Bgl/
Sal and CVB3 Sal/Spe (all three cDNA generated from a pCVB3-M2
construct containing selected parts of the genome region coding for
VP1 of CVB3 PD; see Figure 323) were included to map pleconaril
resistance. The 20 CVB3 isolates were collected and kindly provided
by Dr W. Melchers (University Nijmegen, Netherlands) and Dr S.
Diedrich (National Reference Laboratory for Polio- and Enteroviruses
at the Robert Koch Institute, Berlin, Germany). These CVB3 were
isolated from patients suffering from encephalitis, meningitis, gastroenteritis, carditis, pneumonia, herpangina, hand, foot and mouth
disease, or morbus bornholm between 1970 and 2000. Virus stocks
were prepared in monolayers of CHO-K1 cells (CVB3 PD and Bgl/
Spe), HuFi H (CVB3 P) or HeLa cells (all other laboratory and clinical
CVB3). Viral titres were determined on HeLa cell monolayers by
end-point titration. Aliquots were stored at –20 C until use.
Pleconaril susceptibility testing
The antiviral activity of pleconaril against the CVB3 laboratory strains
was determined on confluent HeLa cells using a cytopathic effect
(CPE)-inhibitory assay described previously for CVB3 Nancy.24
Briefly, the tests were carried out in 2-day-old confluent HeLa cell
monolayers growing in 96-well flat-bottomed microtitre plates (Falcon
3075). After removal of culture medium, 50 mL of two-fold pleconaril
dilutions and 50 mL of CVB3 suspension containing a certain multiplicity of infection (moi) that guarantees a complete CPE after 24–48 h
were added to the cells. Six wells of non-infected and six wells of
infected cells without the test compound served as cell and virus control, respectively, on each plate. Using a Crystal Violet uptake assay,
the inhibition of viral CPE was scored from 24 to 48 h post-infection
when untreated infected control cells showed maximum cytopathic
effect. The 50% inhibitory concentration (IC50) of pleconaril was
determined from the mean dose–response curves of at least three
separate experiments. Two separate experiments were performed
with pleconaril-resistant CVB3.
Virus yield reduction assays were performed with CVB3 P and
CVB PD in confluent HuFi H monolayers propagated in four-well
plates (Greiner, Frickenhausen, Germany). After the cell culture
medium was removed, three wells in each plate were infected with
the respective virus at an moi of 1 in the absence (virus control plates)
or presence of pleconaril (final concentration in the plate 1 mg/mL).
One mock-infected, untreated well was used as cell control on each
plate. Following an incubation time of 4 h at 37 C, the supernatant
consisting of non-adsorbed virus was removed, the cells were washed
three times with maintenance medium and 1 mL of fresh medium
without (cell and virus controls) or with pleconaril (1 mg/mL) was
added to the wells. Four, 24, 48 and 72 h after virus inoculation,
virus was extracted by triple freezing and thawing. The virus yield
of untreated and pleconaril-treated wells was determined by end-point
titration on HeLa cells. Two independent experiments were performed
each with three parallels.
The susceptibility of CVB3 Nancy and clinical isolates was checked
with plaque reduction assays. Confluent HeLa cells grown in 12-well
plates were inoculated with 0.5 mL of the respective virus suspension
in maintenance medium containing between 30 and 60 pfu in the
649
Schmidtke et al.
absence or presence of serial two-fold dilutions of pleconaril.
Each compound concentration was tested in duplicate. One
uninfected untreated cell control and three untreated virus controls
were included per assay. After virus adsorption at 37 C for 1 h, the
inoculum was aspirated and the cell monolayers were overlaid with
1 mL of maintenance medium containing 0.4% agar. Following
2–4 days of incubation at 37 C, the plates were fixed and stained
with a Crystal Violet formalin solution overnight. Plaques were
counted over a light box after removal of the agar overlay and used
to calculate the compound-induced plaque reduction. The IC50 was
calculated from the mean dose–response curve of at least three
independent plaque reduction assays.
RNA isolation
Total RNA was extracted from HeLa cells infected with clinical
CVB3 isolates using an RNeasy Mini Kit (Qiagen, Hilden, Germany)
according to the manufacturer’s instructions. RNA was eluted from the
spin column in a total volume of 30 or 50 mL of RNAase-free water and
stored frozen at –80 C.
RT–PCR of the hydrophobic pocket-encoding region of
VP1 of clinical CVB3 isolates
The region of viral RNA encoding the complete VP1 was amplified by
RT–PCR. RT was conducted with Omniscript RT (Qiagen, Hilden,
Germany), a CVB3-specific primer pool containing primers designed
for PCR and sequencing, and 3 mg of RNA in a final reaction volume of
20 mL following the manufacturer’s instructions. Two microlitres of
the RT reaction was subjected to PCR amplification using Qbiogene
molecular biology TaqDNAPol (Heidelberg, Germany) according to
the manufacturer’s protocol. The primer pairs utilized for PCR were
50 -ACA TCA GAC GAC TTC CAA TC-30 and 50 -ACG TGT GAC
CCG TCT CAG CAG-30 , 50 -GGA AGA CGC GAT AAC AGC CGC
and 50 -GTG GTC TAG GTA TCC ACG CTT-30 , 50 -TGT CCA TYC
CRT TTT TGA G-30 or 50 -ATG TCC ATA CCR TTT TT-30 and
50 -CTG TTG TAR TCY TCC CAC AC-30 (20 mmol). The PCR cycling
conditions were as follows: 1 cycle of 93 C for 5 min; 35 cycles of
94 C for 1 min, 55 C for 50 s and 72 C for 2 min, and a final cycle of
72 C for 20 min, followed by holding at 10 C.
Because in some cases multiple bands were observed when the
amplification products were subjected to agarose gel electrophoresis,
PCR products of the expected molecular mass were gel-extracted using
QIAquick or PCR purification kit (Qiagen, Hilden, Germany) and
eluted in 30–50 mL of elution buffer. Purified products were stored
at –20 C.
Sequencing and analysis of RT–PCR amplicons
Sequence analysis was performed with fluorescent-labelled
nucleotides on an ABI Prism 310 Genetic Analyser (Applied
Biosystems, Darmstadt, Germany). Sequencing primers included
those used in RT–PCR as well as those derived from genome
walking (50 -ATT ACA TGT TGG TAY CAA AC-30 , 50 -GGC CCA
GTG GAA GAC GCG A-30 , 50 -TGY RCK GCC TGG CGW KGT T-30 ,
50 -GAT CAA ACC TCA TGT ATG TGA-30 ), each 5 mmol, where K =
G and T, R = A and G, W = A and T, and Y = C and T. Twenty-five
cycles of 95 C for 20 s, 50 C for 15 s and 60 C for 1 min were run. To
confirm each nucleotide assignment, sequencing of both strands of
cDNA was performed using the sequencing mix DYEnamic ET
terminator kit obtained from Amersham Biosciences (Freiburg,
Germany). The GenBank accession numbers are DQ093617 to
DQ093636.
Computational modelling
The alignment of three neighbouring amino acid residues at positions
1091–1093 of VP1 was predicted using the computer (2400 MHz)
based Molecular Modelling System HyperChem 7.01 (Hypercube,
Inc., http://www.hyper.com). Additionally, Modified Neglect of
Diatomic Overlap (MNDO), a method for semi-empirical quantum
mechanics calculations, was used as a semi-empirical method of
optimization. MNDO calculates electronic properties, optimized
geometries, total energy and heat of formation.
Results
Amino acid polymorphism within the hydrophobic pocket of
CVB3 laboratory strains and clinical isolates
The hydrophobic pocket of enteroviruses plays an important role in
receptor-mediated attachment and uncoating of enteroviruses.12
The high degree of amino acid conservation of this pocket allowed
the development of capsid inhibitors such as pleconaril which
integrate into this pocket thereby preventing viral attachment
and/or uncoating of a broad spectrum of enterovirus serotypes.17
Because the enterovirus prototype CVB3 Nancy was pleconarilresistant and resistant CVB3 mutants were also detected in
response to drug selection pressure in vitro,17,18 concern was raised
about the amino acid polymorphism of the hydrophobic pocket in
VP1 in CVB3 laboratory strains and clinical isolates. The amino
acid residues chosen for comparative analysis are those as determined by X-ray crystallography in CVB3.9
Prior to this study, amino acid sequences of VP1 of 10 CVB3
laboratory strains were published. Alignment of amino acid residues in the hydrophobic pocket of the five CVB3 laboratory strains
used in this study is shown for example in Table 1. Despite a high
degree of amino acid identity, most of these strains possess a
leucine at position 1092 of VP1.20,23,25–27 Ile-1092 was only found
in CVB3 H3 (so-called Woodruff strain) and CVB3 PD.21,23
Additionally, all strains except CVB3 H3 have Val-1180!Ile.
In this study, sequence data of the VP1 genome region encoding
the amino acids within the hydrophobic pocket were generated for
20 CVB3 isolates. Alignment of the amino acid residues of these
isolates is summarized in Table 1. The data demonstrate strong
amino acid conservation within the hydrophobic pocket. The amino
acid substitution Ile-1092!Val was detected in one of the 20
clinical CVB3 isolates. Furthermore, the amino acid substitution
Pro-1094!Thr was found in all except one (CVB3 99-1000) clinical virus isolate. With the exception of CVB3 H3 which was used in
X-ray crystallography by Muckelbauer et al.,9 all CVB3 laboratory
strains as well as all clinical CVB3 isolates have the same amino
acid at position 1180: Ile-1180. In CVB3 H3, it is replaced by
valine.
Susceptibility of CVB3 laboratory strains to pleconaril
Studying pleconaril-resistant CVB3 mutants isolated from cell culture under drug selection pressure, Leu-1092 and Met-1092 have
been associated with a drug-resistant phenotype.18 The occurrence
of Leu-1092 in most CVB3 laboratory strains (Table 1) suggested a
high incidence of resistance. In order to examine the impact of the
amino acid residue at position 1092 on pleconaril susceptibility,
CPE-inhibitory assays were performed with CVB3 M2, CVB3 HA,
CVB3 P and CVB3 PD using non-cytotoxic concentrations of
pleconaril on HeLa cells. Because CPE-inhibitory assays were
650
JAC
Crucial role of amino acid 1092 in anti-CVB3 activity of pleconaril
antiviral
Table 1. Alignment of amino acids lining the b-sandwich pocket of VP1 according to Muckelbauer et al.9
Amino acid residues of the b-sandwich pocket of viral protein VP1 at position
CVB3
92
I
94
P
95
R
114
L
116
L
143
Y
165
P
166
S
167
V
178
M
180
V
183
L
184
S
189
Y
211
N
213
M
216
L
237
F
H3a
Nancyb
HAc
Pc
PDc
97-927
00-637
99-1000
304037
302784
392323
390557
0450/76
0518/79
0144/81
0412/87
98-1422
0113/79
0141/79
0142/78
0441/84
0445/81
0513/87
1340/70
1569/70
–
L
L
L
–
–
–
V
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
T
T
–
T
T
T
T
T
T
T
T
T
T
T
T
T
T
T
T
T
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
I
I
I
I
I
I
I
I
I
I
I
I
I
I
I
I
I
I
I
I
I
I
I
I
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
–
Sequences of CVB3 laboratory strains used in this study were published by: aKnowlton et al.,21 bKlump et al.25 and cSchmidtke et al.23
also applied in the initial studies to determine the activity of pleconaril against enteroviruses, the results of this study obtained with
pleconaril-resistant and pleconaril-susceptible control viruses
(CVB3 Nancy and CVB3 H3, respectively) are directly comparable to those obtained by Pevear et al.17 The 50% cytotoxic concentration of pleconaril on HeLa cells, determined in our
laboratory, amounts to 12.6 mg/mL and was described recently.28
The results shown in Figure 1(a) indicate that all CVB3 laboratory
strains containing Leu-1092 were pleconaril-resistant like CVB3
Nancy. The resistant phenotype of CVB3 Nancy was also confirmed in plaque reduction assays (n = 4; results not shown). In
addition to CVB3 H3, only CVB3 PD was pleconaril-susceptible
(Figure 1b). Like CVB3 H3, CVB3 PD has an isoleucine at
position 1092.
Because CPE inhibition does not necessarily mean a reduction
in infectious virus, virus yield reduction assays were performed
with CVB3 P and CVB PD in diploid human fibroblasts. As shown
in Figure 1(c), the virus yield of CVB3 P-infected pleconariltreated HuFi was only slightly reduced in comparison with the
untreated virus control. In contrast, replication of CVB3 PD was
nearly completely aborted after treatment with pleconaril. Neither
24 nor 48 nor 72 h after virus inoculation has a rise in viral titres
been found in pleconaril-treated, CVB3 PD-infected HuFi H
whereas the virus yield of untreated virus control increased
about four logarithmic steps (Figure 1d). These results confirm
the resistant phenotype of CVB3 P as well as pleconaril susceptibility of CVB3 PD found in CPE-inhibitory assays.
Relationship between pocket amino acid sequence and
drug susceptibility of clinical CVB3 isolates
Pleconaril susceptibility of clinical CVB3 isolates was studied by
plaque reduction assays in HeLa cells. Based on the mean dose–
plaque reduction curves of three separate experiments, the IC50
values were calculated for individual CVB3 isolates (Figure 2).
Pleconaril inhibited the virus-induced plaque production of
clinical CVB3 isolates by 50% in a concentration range
of 13.0–67.4 ng/mL. The mean IC50 calculated from the individual
values of all 20 isolates amounted to 38.4 – 14.7 ng/mL. All of
the studied clinical CVB3 isolates were pleconaril-susceptible.
The results indicate that none of the observed amino acid substitutions at positions 1092, 1094 and 1180 induced resistance
(Table 1).
Evidence for the crucial role of the amino acid at
position 1092
To demonstrate the impact of amino acid 1092 on pleconaril susceptibility, several virus chimeras were used (Figure 3). These
viruses were originally generated to study the molecular base of
651
Schmidtke et al.
(a)
(b)
100
100
CVB3 Nancy
CPE inhibition (%)
CVB3 M2
Log10 TCID50 /50 µL
80
CVB3 HA
60
60
40
40
20
20
0
(c)
CVB3 P
80
0
6
1
2
4
5
6
3
Concentration (µg/mL)
7
0
8
(d)
Virus control
After treatment with 1 µg/mL of pleconaril
CVB3 H3
CVB3 PD
0
0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 1.0
Concentration (µg/mL)
7
6
5
5
4
4
3
3
2
2
1
0
1
4
24
48
Hours after infection
0
72
4
24
48
Hours after infection
72
Figure 1. Pleconaril susceptibility of CVB3 laboratory strains. Antiviral activity of pleconaril was determined in CPE-inhibitory assays in HeLa cells (a and b). The
mean dose–response curves with SD of resistant viruses were calculated from two (a) and that of susceptible viruses from at least three separate CPE-inhibitory assays
in HeLa cells (b). To confirm the pleconaril-resistant phenotype of CVB3 P (c) and the pleconaril-susceptible phenotype of CVB3 PD (d), virus yield assays were
performed in HuFi infected at an moi of 1 of respective CVB3. At 4, 24, 48 and 72 h after virus infection, the plates were frozen and thawed three times. Virus yield
was determined by end-point titration in HeLa cells. The results are expressed as 50% tissue culture infection dose per 50 mL of virus suspension (TCID50/50 mL). Mean
virus yields – SD of two independent experiments are shown each with three parallels.
70
IC50 values (ng/mL)
60
50
40
30
20
10
97
-9
27
00
-6
3
99 7
-1
00
0
30
40
37
30
27
84
39
23
23
39
05
5
04 7
50
/7
05 6
18
/7
01 9
44
/8
04 1
12
/8
98 7
-1
42
2
13
40
/7
15 0
69
/7
0
01
42
/7
01 8
13
/
01 79
41
/7
9
04
45
/8
1
04
41
/8
4
05
13
/8
7
0
Clinical CVB3 isolates
Figure 2. Pleconaril susceptibility of clinical CVB3 isolates. The antiviral activity of pleconaril was determined by plaque reduction assays in HeLa cell monolayers
grown in 12-well plates. Two-fold dilutions of non-cytotoxic pleconaril concentrations were added to cells during virus adsorption and in the agar overlay. Then,
2–4 days after infection, assays were fixed and stained with Crystal Violet solution, the plaques were counted, and the plaque reduction in compound-treated wells
was calculated in comparison with untreated virus-infected wells. Based on the mean dose–response curve of at least three separate experiments, the IC50 values
were calculated.
the extended cell tropism of CVB3 PD.23 In addition to amino acid
substitutions at the surface of the viral capsid allowing the use of
heparan sulphates as additional receptors,29 CVB3 PD also contains the pocket amino acid Ile-1092. Ile-1092 was suggested to
mediate pleconaril susceptibility.18 All other amino acids lining the
hydrophobic pocket of CVB3 PD were completely the same as
found in the pleconaril-resistant CVB3 Nancy (Table 1). Three
different subgenomic regions of CVB3 PD which included residue
652
JAC
Crucial role of amino acid 1092 in anti-CVB3 activity of pleconaril
antiviral
amino acid residue at position 1092
IC50 (µg/mL)
of pleconaril
L
pCVB3 M2
5'NTR VP4 VP2
Bgl II
Sal I
VP3
VP1
Spe I
2A 2B
2C
3A 3B 3C
3D
3'NTR
>8.0
2C
3A 3B 3C
3D
3'NTR
0.038
2A 2B
2C
3A 3B 3C
3D
3'NTR
0.038
2A 2B
2C
3A 3B 3C
3D
3'NTR
0.014
2C
3A 3B 3C
3D
3'NTR
>8.0
I
Bgl II
CVB3 PD
5'NTR VP4 VP2
VP3
Spe I
VP1
2A 2B
I
Bgl II
CVB3 Bgl/Spe
5'NTR VP4 VP2
VP3
Spe I
VP1
I
Bgl II
CVB3 Bgl/SaI
5'NTR VP4 VP2
Sal I
VP3 VP1
L
Sal I
CVB3 Sal/Spe
5'NTR VP4 VP2
Spe I
VP3 VP1
2A 2B
Figure 3. Structures of the genomes of hybrid viruses used to demonstrate the crucial role of amino acid 1092 in pleconaril susceptibility of CVB3 are shown in
parallel with IC50 values of pleconaril. The IC50 values were calculated from the mean dose–response curves obtained with the respective viruses in at least three
independent CPE-inhibitory assays in HeLa cells. NTR, non-translated region.
Ile-1092 (CVB3 Bgl/Spe and CVB3 Bgl/Sal) or not (CVB3 Sal/
Spe) were amplified, digested with the appropriate restriction
enzymes, and ligated to the plasmid pCVB3-M2. This plasmid
contains the genome encoding CVB3 Nancy. As shown in
Figures 1(a) and 3, infectious viruses generated from pCVB3M2 (Leu-1092) are completely pleconaril-resistant up to a maximum test concentration of 8 mg/mL like CVB3 Nancy. After
replacing the subgenomic regions of the resistant CVB3 M2 encoding Leu-1092 with the respective regions of CVB3 PD encoding
Ile-1092 (CVB3 Bgl/Spe and Bgl/Sal) but not when the region
corresponding to the CVB3 PD Sal/Spe fragment encoding
Leu-1092 like CVB3 M2 was replaced, the replication of the
virus chimera was inhibited by the compound as efficiently as
the pleconaril-susceptible CVB3 PD. The IC50s determined for
CVB PD, CVB3 Bgl/Spe and CVB3 Bgl/Sal were 0.038, 0.038
and 0.014 mg/mL, respectively. In contrast, inhibition of CVB3 Sal/
Spe-induced CPE was not achieved albeit using maximum test
concentrations of pleconaril of 8 mg/mL.
Possible changes in the canyon structure after
substitution of amino acid 1092
The results obtained with virus chimeras in antiviral assays demonstrate that pleconaril susceptibility of CVB3 depends on one
single amino acid in the viral hydrophobic pocket. All of the
detected pleconaril-resistant CVB3 contain a leucine at position
1092 of VP1. In contrast, pleconaril-susceptible CVB3 possess an
isoleucine or a valine at position 1092. Until now, no atomic structure is available for CVB3 possessing a leucine or valine at position
1092 in VP1. The structure of isoleucine, leucine and valine is very
similar. Isoleucine, leucine and valine are non-polar, hydrophobic
amino acids differing in the number of hydrocarbons of side chains
(four in isoleucine and leucine; three in valine) or in the arrangement of these hydrocarbons (Figure 4a). These structural data
suggest that differences in size of hydrophobic side chains alone
do not explain pleconaril resistance. To check the hypothesis that
the amino acid residue at position 1092 may have an effect on the
conformation of the canyon, three-dimensional (3D) conformation
modelling of isolated tri-peptides consisting of amino acids
1091–1093 was applied. The proposed computer model considers
the conformation of the tri-peptides Val–Ile–Thr, Val–Leu–Thr
and Val–Ile–Thr in isolation from the context of the surrounding
capsid residues. In this manner, it was possible to show how a minor
variation like a single amino acid substitution can change the
peptide conformation in a specific position of the hydrophobic
pocket. The obtained biological data together with the detected
high conservation of the amino acid sequence suggest that only
very small changes of the whole peptide conformation may
occur, because otherwise the function of the hydrophobic pocket
would be destroyed. Surprisingly, 3D images of Val–Ile–Thr and
653
Schmidtke et al.
(a)
HOOC
NH2
H3C
CH3
HOOC
HOOC
NH2
NH2
H3C
CH3
CH3
CH3
(b)
VVT
(c)
VIT+
VVT
VIT
VLT
VIT+
VLT
Figure 4. Alignment of amino acid residues at positions 1091–1093 of VP1. The structures of valine, isoleucine and leucine (from left to right) are provided to show
the number of hydrocarbon side chains as well as their arrangement (a). The 3D conformation of Val–Ile–Thr, Val–Val–Thr and Val–Leu–Thr was predicted using the
computer (2400 MHz) based Molecular Modelling System HyperChem 7.01 and semi-empirical optimization with MNDO (b). Additionally, two 3D images (Val–
Ile–Thr and Val–Val–Thr as well as Val–Ile–Thr and Val–Leu–Thr) were laid on top of each other to emphasize the difference in amino acid composition (c).
Val–Val–Thr possess a very similar conformation whereas the
introduction of leucine at position 1092 markedly altered the
arrangement of the amino acid residues of the peptide Val–Leu–
Thr (Figure 4b). To emphasize the difference in amino acid
composition, two 3D images, Val–Ile–Thr and Val–Leu–Thr as
well as Val–Ile–Thr and Val–Val–Thr, are shown in Figure 4(c).
Valine as well as isoleucine contains two alkyl substitutes (methyl
and methyl or methyl and ethyl, respectively) at the third carbon
atom. As shown in Figure 4(b and c), the presence of a second alkyl
substitute at C(3) of the central amino acid residue results in a
conformation problem for oxygen of the respective amino acid.
This oxygen atom contacts the oxygen of the first amino acid
residue which is valine. In contrast to valine as well as isoleucine,
leucine has only one alkyl substitute (isopropyl) at the third carbon
atom. Because sterical inhibition of the oxygen of leucine due to a
second alkyl substitute does not exist, oxygen may remove itself
from the oxygen of valine in Val–Leu–Thr. This induces alterations
in the conformation of the whole molecule studied. These results
indicate that an exchange of isoleucine or valine for leucine may
affect the structure of this part of the hydrophobic pocket and that
may hinder pleconaril binding.
Discussion
CVB3 is one of about 70 distinct human enterovirus serotypes
causing a broad spectrum of human diseases. Because no single
enterovirus serotype is exclusively associated with any particular
disease, diagnostics as a prerequisite of successful treatment were
difficult for a long time. During the last 10 years, PCR methods
were developed for group-specific diagnosis of enterovirus infections.30–32 This rapid group-specific diagnosis now enables the
use of broad-spectrum anti-enteroviral agents for treatment.
Our present knowledge suggests the capsid inhibitor pleconaril
as the most promising candidate. This compound integrates into
a hydrophobic pocket in VP1 and blocks viral attachment
and/or uncoating. Pleconaril inhibited 14 of 15 tested prototypic
enterovirus strains as well as 214 of 215 tested clinical enterovirus
isolates in vitro.17 The only detected resistant prototype enterovirus
was coxsackievirus B3 Nancy. In addition, 10 drug-resistant CVB3
mutants were generated from pleconaril-susceptible wild-type
CVB3 under pleconaril treatment by plaque isolation.18 Because
all these resistant mutants possess an amino acid substitution
Ile-1092!Leu or Ile-1092!Met in the hydrophobic pocket of
VP1, it was suggested that this amino acid position may be critical
to the development of drug resistance. This study reports: (i) the
intraserotypic amino acid polymorphism of the hydrophobic
pocket of 20 clinical isolates in comparison with already known
amino acid sequences of CVB3 laboratory strains; (ii) insight
into the range of pleconaril susceptibility of these viruses; and
(iii) evidence for the crucial role of a single amino acid in pleconaril treatment. Moreover, results from computational modelling
suggest conformational changes within the hydrophobic pocket are
the result of this amino acid substitution.
Taken together, there is a high degree of amino acid identity
within the hydrophobic pocket of the CVB3 studied (Table 1). Of
the 18 amino acid positions, 15 are identical. Val-1180!Ile was
found in all studied CVB3 except CVB H3. Besides CVB3
99-1000, all clinical CVB3 isolates possess Thr-1094 instead of
Pro-1094. Leu-1092 but not Met-1092 was found in CVB3 laboratory strains but not in clinical isolates in this study. In addition,
CVB3 99-1000 contained a valine at that position. Only Leu-1092,
observed in CVB3 Nancy, M2, HA and P, was correlated with
resistance to pleconaril (Figure 1a and c). Because these viruses
were never treated with pleconaril before, they were called naturally pleconaril-resistant. In contrast, replication of CVB3 containing Ile-1092 or Val-1092 (CVB3 H3, CVB3 PD and all clinical
isolates) was strongly inhibited by the drug (Figures 1b and d
and 2). The resistant phenotype of CVB3 Nancy as well as susceptibility of CVB3 H3 described previously by Groarke and
654
Crucial role of amino acid 1092 in anti-CVB3 activity of pleconaril
Pevear18 were confirmed: IC50 values correspond to previously
published data.17 No natural pleconaril-resistant clinical CVB3
isolates were detected. However, neither the existence of resistant
strains nor their emergence as the result of drug selection pressure
can be excluded. Studying sequences submitted to the NCBI GenBank, we found one recent CVB3 isolate from China (AAD50446)
possessing Leu-1092. The majority of the published sequences
(more than 20) contained Ile-1092. Several CVB3 isolates possess
Val-1092. During clinical studies designed to examine the safety
and efficacy of pleconaril for treatment of upper-respiratory-tract
disease of picornavirus aetiology, post-baseline non-susceptible
rhinoviruses were isolated from 2.7% of treated patients.33
Amino acid substitutions of resistant viruses isolated from
pleconaril-treated patients of these clinical studies were different
from those found in natural drug-resistant rhinoviruses.34 In CVB3,
drug selection pressure resulted in the emergence of pleconarilresistant variants containing the same amino acid substitutions18
as those observed in drug-resistant CVB3 (Nancy, M2, HA, P) of
this study which were never treated with pleconaril before.
When assessed in a murine model in which wild-type infection is
lethal, pleconaril-resistant CVB3 that emerged under pleconaril
treatment were attenuated and significantly less virulent than
drug-susceptible wild-type virus.18 These results indicate that
virus–receptor interactions and subsequent viral attachment and
entry are also affected by those amino acid substitutions. However,
results from our previous studies on replication of CVB3 laboratory
strains in cell lines with determined receptor expression indicate no
differences between CVB3 laboratory strains containing Ile-1092
or Leu-1092 in regard to CAR-mediated infection (where CAR
stands for coxsackievirus-adenovirus receptor) and virulence
in vitro.23,29 Moreover, CVB3 with Leu-10927,35 as well as
CVB3 with Ile-1092 (ref. 21 and M. Schmidtke, unpublished
data) were successfully used to establish animal models of acute
and chronic myocarditis. Obviously, amino acid variation at position 1092 is not necessarily associated with attenuated virulence
in vitro or in vivo. Possibly, other mutations in the 50 non-coding
region or in other virus protein coding regions that were not examined in the respective studies have a potential role in the observed
phenotypes.
According to Muckelbauer et al.,9 amino acid residues 1092 and
1180 are situated across the pocket from each other with their side
chains extending into the pocket and both are near the substituted
phenyl ring of capsid binding compounds. These authors suggested
that the change from isoleucine to leucine at 1092, in addition to
Val-1180!Ile, may be responsible for the inability of those CVB3
to bind capsid blocking compounds. In this study, hybrid viruses
constructed on the genetic background of the pleconaril-resistant
CVB3 Nancy (Leu-1092 and Ile-1180) were used to prove the
impact of these amino acids on pleconaril susceptibility
(Figure 3). Three different genome regions of CVB3 PD coding
for parts of VP1 containing Ile-1092 (CVB3 Bgl/Spe and Bgl/Sal)
or not (CVB3 Sal/Spe) in addition to Ile-1180 (all) were transferred
into a plasmid coding for CVB3 Nancy. Viruses obtained after
transfection of HeLa cells with the respective plasmids were tested
for drug susceptibility. Only the two hybrid viruses containing
Ile-1092 resulted in the susceptible phenotype. The CVB3 PD
Bgl/Sal fragment is the minimal sequence that results in the transfer
of susceptibility to pleconaril to the M2 strain. In addition to Ile1092, it contains four other mutations (3237, 1078, 1080 and 1091)
outside of the binding pocket. These are unique for CVB3 PD
(1078, 1080 and 1091) or found also in CVB3 H3 and some of
JAC
antiviral
the isolates (3237). The presence or absence of these amino acid
substitutions did not correlate with drug susceptibility of CVB3
laboratory strains and isolates. Therefore, a potential role of the
other mutations in the observed phenotype can be discarded. Taken
together, these data demonstrate that a single amino acid substitution at position 1092 is indeed associated with susceptibility or
resistance of CVB3 to pleconaril.
Surprisingly, the substitution of non-polar isoleucine by nonpolar leucine but not by non-polar valine induces pleconaril resistance. Both isoleucine and leucine have four hydrocarbon side
chains. The difference between isoleucine and leucine is the
arrangement of these hydrocarbon chains. As shown by 3D conformation modelling of the amino acid at position 1092 together
with its neighbouring amino acids, this small difference has the
potential to alter the arrangement of amino acids of peptides as
shown for Val–Ile–Thr and Val–Leu–Thr (Figure 4b and c). The
results obtained with valine confirm this conclusion. Valine contains three hydrocarbon side chains but their arrangement is
comparable to that of isoleucine (Figure 4a). The 3D conformation
of Val–Val–Thr was very similar to that of Val–Ile–Thr (Figure 4b
and c). These conformational changes may affect the binding of
pleconaril in the pocket. However, the atomic structure of CVB3 is
only known for a strain containing Ile-1092 at present.9 The
correlation between a single amino acid exchange at position
1092 of the hydrophobic pocket and anti-CVB3 activity of pleconaril together with the hypothesis about the structural changes at
this position obtained from computational modelling help us to
understand the molecular basis of anti-CVB3 activity as well as
drug resistance of capsid function inhibitors. Based on these results,
new anti-CVB3 active compounds will be designed, synthesized
and tested.
In summary, data reported here contribute to the CVB3 database
and provide new insight into the polymorphism of the hydrophobic
pocket of these viruses. The observed polymorphism can play an
important role in binding of capsid inhibitors in the pocket and by
this way in treatment. Moreover, these data provide evidence that
the amino acid at position 1092 but not at position 1094 or 1180 can
predict the susceptibility of pleconaril and could form the basis of
rapid resistance monitoring by genotyping.
Acknowledgements
We thank B. Jahn and V. Güntzschel for excellent technical assistance. This study was supported by grants from the DFG (SCHM
1594/1), the German Paul Ehrlich Society for Chemotherapy, and
the Ministry of Culture and Education of Thueringen, Germany.
References
1. Chatterjee NK, Moore DW, Monroe SS et al. Molecular epidemiology of outbreaks of viral gastroenteritis in New York State, 1998–1999.
Clin Infect Dis 2004; 38 Suppl 3: S303–10.
2. Glass RI, Bresee J, Jiang B et al. Gastroenteritis viruses: an overview. Novartis Found Symp 2001; 238: 5–19, discussion 19–25.
3. Monto AS, Fendrick AM, Sarnes MW. Respiratory illness caused by
picornavirus infection: a review of clinical outcomes. Clin Ther 2001; 23:
1615–27.
4. Muir P, van Loon AM. Enterovirus infections of the central nervous
system. Intervirology 1997; 40: 153–66.
655
Schmidtke et al.
5. Kandolf R, Sauter M, Aepinus C et al. Mechanisms and consequences of enterovirus persistence in cardiac myocytes and cells of the
immune system. Virus Res 1999; 62: 149–58.
6. Kim KS, Hufnagel G, Chapman NM et al. The group B coxsackieviruses and myocarditis. Rev Med Virol 2001; 11: 355–68.
7. Klingel K, Selinka HC, Huber M et al. Molecular pathology and
structural features of enteroviral replication. Toward understanding the
pathogenesis of viral heart disease. Herz 2000; 25: 216–20.
8. Rotbart HA. Treatment of picornavirus infections. Antiviral Res
2002; 53: 83–98.
9. Muckelbauer JK, Kremer M, Minor I et al. The structure of coxsackievirus B3 at 3.5 Å resolution. Structure 1995; 3: 653–67.
10. Hogle JM, Chow M, Filman DJ. Three-dimensional structure of
poliovirus at 2.9 Å resolution. Science 1985; 229: 1358–65.
11. Rossmann MG, Arnold E, Erickson JW et al. Structure of a human
common cold virus and functional relationship to other picornaviruses.
Nature 1985; 317: 145–53.
12. Rossmann MG, He Y, Kuhn RJ. Picornavirus-receptor interactions.
Trends Microbiol 2002; 10: 324–31.
13. Bibler-Muckelbauer JK, Kremer MJ, Rossmann MG et al. Human
rhinovirus 14 complexed with fragments of active antiviral compounds.
Virology 1994; 202: 360–9.
14. Rossmann MG. The structure of antiviral agents that inhibit uncoating when complexed with viral capsids. Antiviral Res 1989; 11: 3–13.
15. Diana GD. Inhibitors of picornavirus replication. Curr Med Chem
2003; 2: 1–12.
16. McKinlay MA, Pevear DC, Rossmann MG. Treatment of the picornavirus common cold by inhibitors of viral uncoating and attachment.
Annu Rev Microbiol 1992; 46: 635–54.
17. Pevear DC, Tull TM, Seipel ME et al. Activity of pleconaril against
enteroviruses. Antimicrob Agents Chemother 1999; 43: 2109–15.
18. Groarke JM, Pevear DC. Attenuated virulence of pleconarilresistant coxsackievirus B3 variants. J Infect Dis 1999; 179: 1538–41.
19. Tonew M, Hartmann M, Schmidtke M et al. Replication and persistence of coxsackieviruses B3 in human fibroblasts. Zentralbl Bakteriol
1995; 282: 92–101.
20. Lindberg AM, Crowell RL, Zell R et al. Mapping of the RD phenotype
of the Nancy strain of coxsackievirus B3. Virus Res 1992; 24: 187–96.
21. Knowlton KU, Jeon ES, Berkley N et al. A mutation in the puff region
of VP2 attenuates the myocarditic phenotype of an infectious cDNA of the
Woodruff variant of coxsackievirus B3. J Virol 1996; 70: 7811–8.
22. Tonew M, Wagner B, Wagner M et al. Permissiveness of human
embryonal fibroblasts for coxsackieviruses B3. Investigations on virus
genetic markers in vitro and localization of virus receptor distribution by
immunogold replica technique. Zentralbl Bakteriol 1996; 284: 443–56.
23. Schmidtke M, Selinka HC, Heim A et al. Attachment of coxsackievirus B3 variants to various cell lines: mapping of phenotypic differences
to capsid protein VP1. Virology 2000; 275: 77–88.
24. Schmidtke M, Schnittler U, Jahn B et al. A rapid assay for evaluation
of antiviral activity against coxsackie virus B3, influenza virus A, and
herpes simplex virus type 1. J Virol Methods 2001; 95: 133–43.
25. Klump WM, Bergmann I, Muller BC et al. Complete nucleotide
sequence of infectious coxsackievirus B3 cDNA: two initial 50 uridine
residues are regained during plus-strand RNA synthesis. J Virol 1990;
64: 1573–83.
26. Lindberg AM, Stalhandske PO, Pettersson, U. Genome of coxsackievirus B3. Virology 1987; 156: 50–63.
27. Tracy S, Chapman NM, Tu Z. Coxsackievirus B3 from an infectious
cDNA copy of the genome is cardiovirulent in mice. Arch Virol 1992; 122:
399–409.
28. Makarov VA, Riabova OB, Granik VG et al. Novel [(biphenyloxy)propyl]isoxazole derivatives for inhibition of human rhinovirus 2 and coxsackievirus B3 replication. J Antimicrob Chemother 2005; 55: 483–8.
29. Zautner AE, Korner U, Henke A et al. Heparan sulfates and coxsackievirus-adenovirus receptor: each one mediates coxsackievirus B3
PD infection. J Virol 2003; 77: 10071–7.
30. Rabenau HF, Clarici AM, Muhlbauer G et al. Rapid detection of
enterovirus infection by automated RNA extraction and real-time
fluorescence PCR. J Clin Virol 2002; 25: 155–64.
31. Verstrepen WA, Bruynseels P, Mertens AH. Evaluation of a rapid
real-time RT–PCR assay for detection of enterovirus RNA in cerebrospinal
fluid specimens. J Clin Virol 2002; 25 Suppl 1: S39–43.
32. Watkins-Riedel T, Woegerbauer M, Hollemann D et al. Rapid diagnosis of enterovirus infections by real-time PCR on the LightCycler using
the TaqMan format. Diagn Microbiol Infect Dis 2002; 42: 99–105.
33. Hayden FG, Herrington DT, Coats TL et al. Efficacy and safety of
oral pleconaril for treatment of colds due to picornaviruses in adults: results
of 2 double-blind, randomized, placebo-controlled trials. Clin Infect Dis
2003; 36: 1523–32.
34. Ledford RM, Patel NR, Demenczuk TM et al. VP1 sequencing of all
human rhinovirus serotypes: insights into genus phylogeny and susceptibility to antiviral capsid-binding compounds. J Virol 2004; 78: 3663–74.
35. Merkle I, Tonew M, Gluck B et al. Coxsackievirus B3-induced
chronic myocarditis in outbred NMRI mice. J Hum Virol 1999; 2: 369–79.
656