Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Molecular Epidemiology of Coxsackievirus A16: Intratype and Prevalent Intertype Recombination Identified

  • Xiangpeng Chen ,

    Contributed equally to this work with: Xiangpeng Chen, Xiaojuan Tan

    Affiliation Key Laboratory of Medical Virology Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, P. R. of China

  • Xiaojuan Tan ,

    Contributed equally to this work with: Xiangpeng Chen, Xiaojuan Tan

    Affiliation Key Laboratory of Medical Virology Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, P. R. of China

  • Jing Li,

    Affiliations Key Laboratory of Medical Virology Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, P. R. of China, Yangzhou No.1 People’s Hospital, Yangzhou, P. R. of China

  • Yu Jin,

    Affiliation Nanjing Children’s Hospital, Nanjing Medical University, Nanjing, P. R. of China

  • Liming Gong,

    Affiliation Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, P. R. of China

  • Mei Hong,

    Affiliation Xizang Provincial Center for Disease Control and Prevention, Lasa, P. R. of China

  • Yonglin Shi,

    Affiliation Anhui Provincial Center for Disease Control and Prevention, Hefei, P. R. of China

  • Shuangli Zhu,

    Affiliation Key Laboratory of Medical Virology Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, P. R. of China

  • Baomin Zhang,

    Affiliation Key Laboratory of Medical Virology Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, P. R. of China

  • Shuang Zhang,

    Affiliations Key Laboratory of Medical Virology Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, P. R. of China, Shunyi Center for Disease Control and Prevention, Beijing, P. R. of China

  • Yong Zhang,

    Affiliation Key Laboratory of Medical Virology Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, P. R. of China

  • Naiying Mao,

    Affiliation Key Laboratory of Medical Virology Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, P. R. of China

  • Wenbo Xu

    wenbo_xu1@aliyun.com

    Affiliation Key Laboratory of Medical Virology Ministry of Health, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, P. R. of China

Abstract

Coxsackievirus A16 (CVA16) is responsible for nearly 50% of all the confirmed hand, foot, and mouth disease (HFMD) cases in mainland China, sometimes it could also cause severe complications, and even death. To clarify the genetic characteristics and the epidemic patterns of CVA16 in mainland China, comprehensive bioinfomatics analyses were performed by using 35 CVA16 whole genome sequences from 1998 to 2011, 593 complete CVA16 VP1 sequences from 1981 to 2011, and prototype strains of human enterovirus species A (EV-A). Analysis on complete VP1 sequences revealed that subgenotypes B1a and B1b were prevalent strains and have been co-circulating in many Asian countries since 2000, especially in mainland China for at least 13 years. While the prevalence of subgenotype B1c (totally 20 strains) was much limited, only found in Malaysia from 2005 to 2007 and in France in 2010. Genotype B2 only caused epidemic in Japan and Malaysia from 1981 to 2000. Both subgenotypes B1a and B1b were potential recombinant viruses containing sequences from other EV-A donors in the 5’-untranslated region and P2, P3 non-structural protein encoding regions.

Introduction

In recent years, the hand, foot and mouth disease (HFMD) has been a very common infection for children in Asian countries caused by various enteroviruses, with coxsackievirus A16 (CVA16) and enterovirus 71 (EV-A71) as the two major causative agents [1]. Since EV-A71 was more frequently associated with severe complications including aseptic meningitis, acute flaccid paralysis, brainstem encephalitis, pulmonary edema, and even death, so there were a lot of studies focusing on EV-A71 [2-5], but relatively less on CVA16. Studies on molecular epidemiology of CVA16 were reported elsewhere [6-10], the genetic diversity and evolutionary characteristics of this virus has yet to be explored. Previous studies have shown that CVA16 was responsible for the confirmed HFMD cases in China in recent years [11-13]; besides HFMD and herpangina, infection by CVA16 could also cause serious myocarditis and pericarditis, acute heart failure, and even death [14,15].

The genome of CVA16 is a single stranded, positive sense RNA, containing approximately 7,410 nucleotides with a single open-reading-frame (ORF). The viral genome contains 5'- and 3’- untranslated regions (UTRs) which are essential for viral RNA replication and expression. The 5’-UTR is about 740 nucleotides in length and comprises the internal ribosomal entry site, which is related to the replication and internal initiation of translation of the genomic RNA [16]. The ORF has 6,579 nucleotides, encoding a polyprotein of 2,193 amino acids which is composed of 3 protein precursors: P1, P2, and P3. The P1 polyprotein precursor processed into 4 structural proteins, VP1 to VP4, while P2 and P3 are precursors of the seven nonstructural proteins; 2A, 2B, 2C, 3A, 3B, 3C, and 3D, respectively [17,18].

Since CVA16 was first identified in 1951 in South Africa, it evolved slowly [19]. Complete genomic analysis of Enterovirus A (EV-A) indicated that recombination in the nonstructural region played an important role in the evolution of some EV-A strains [20]. Recombination between CVA16 and other serotypes of EV-A in the nonstructural region had also been documented previously by using partial sequences available earlier [11], however, the recombination analysis was also limited by the lack of complete genome sequences. To clarify the global molecular epidemiology and genetic characteristic of CVA16, we perform an extensive genetic analysis using all of the available genomic sequences of CVA16 dated before December 2012 in the public database, plus 8 complete genome sequences determined in our laboratory.

Results

Genotyping based on complete VP1 sequences of CVA16

A total of 629 sequences with complete VP1 region of CVA 16 were used for analysis, included 621 retrieved from public database dated before December 2012, plus 8 complete genome sequences determined by our laboratory (Table 1, Table S2). Phylogenetic analyses were performed with MEGA software. CVA16 strains could be divided into genotypes A, B1 and B2 as indicated in our previous study [19]. Further, genotype B1 could be divided into subgenotypes B1a, B1b, and B1c. Among 629 VP1 sequences, 607 clustered together with the reference sequence of genotype B1, and 21 showed the nearest relationship with genotype B2, while the prototype CVA16 was the unique member of genotype A (Figure 1). The genotype B2 was first reported from Japan in 1981 (AB465366). Among the 21 B2 strains, 17 were isolated from Japan during 1981-1998, and 4 from Malaysia during 1998-2000. After 2000, there was no genotype B2 reported. The earliest B1 strains were isolated in 1995 from Japan (AB634295-AB634311). Relatively, genotype B1 was more prevalent in the epidemics and had been reported in many countries, including mainland China, Taiwan, Japan, Malaysia, Thailand, Vietnam, Australia, France, Korea, Spain, Sweden and Saudi Arabia since then.

Strain nameSourceGenBank Accession NumberThe place of isolationThe year of isolationgenotype
Kor08/South Korea/2008GenBankJX839965South Korea 2008B1a
PM-12284-99/MAL/1999GenBankJQ746661Malaysia 1999B1a
PM-15765-00/MAL/2000GenBankJQ746666Malaysia 2000B1a
PM-00033-07/MAL/2007GenBankJQ746660Malaysia 2007B1a
THA-CA16-090/Thailand/2010GenBankJF738003Thailand 2010B1a
THA-CA16-069/Thailand/2010GenBankJF738004Thailand 2010B1a
Tainan/5079/98/Taiwan/1998GenBankAF177911Taiwan 1998B1a
BJ/11/11/BJ/CHN/2011GenBankJX068831Beijing, China2011B1a
KMM/08/CHN/2008GenBankHQ423141Yunnan, China2008B1a
TS10/08/CHN/2010GenBankJX068829China 2010B1a
SZ/HK08-3/HK/CHN/2008GenBankGQ279368HongKong, China2008B1a
Ningbo.CHN/028-2/2009/ZJ/CHN/2009GenBankJQ354992Zhejiang, China2009B1a
GZ08/GD/CHN/2008GenBankFJ198212Guangdong, China2008B1b
HQ09011181/YN/CHN/2011GenBankJQ316639Yunnan, China2011B1a
shzh05-1/GD/CHN/2005GenBankEU262658Guangdong, China2005B1b
TS10/07/CHN/2010GenBankJX068827China 2010B1b
BJ09/06/BJ/CHN/2009GenBankJX068832Beijing, China2009B1b
BJCA08/BJ/CHN/2008GenBankJX481738Beijing, China2008B1b
BJ08/07/BJ/CHN/2008GenBankJX068833Beijing, China2008B1b
BJ11/03/BJ/CHN/2011GenBankJX068830Beijing, China2011B1b
BJ11/12/BJ/CHN/2011GenBankJX068828Beijing, China2011B1b
HN1662/HeN/CHN/2010GenBankJN674176Henan, China2010B1b
G20/YN/CHN/2010GenBankJN590244Yunnan, China2010B1b
XM-CA16-3560/FJ/CHN/2009GenBankHQ269389Fujian, China2009B1b
shzh00-1/GD/CHN/2000GenBankAY790926Guangdong, China2000B1b
SZ/HK08-7/HK/CHN/2008GenBankGQ279371HongKong, China2008B1b
SH/CHN/2009/ CHN/2010GenBankJQ034149Shanghai, China2010B1b
ZJ10-48This studyKC755235Zhejiang, China2010B1a
ZJ10-73This studyKC755229Zhejiang, China2010B1a
AH10-2This studyKC755230Anhui, China2010B1a
XZ10-C-1This studyKC755234Tibet, China2010B1a
NJ10-31This studyKC755232Jiangsu, China2010B1a
XZ10-D-1This studyKC755228Tibet, China2010B1b
NJ10-75This studyKC755233Jiangsu, China2010B1b
AH10-12This studyKC755231Anhui, China2010B1b

Table 1. List of the complete genomes sequences of 35 CVA16 strains used for the sequence comparison, phylogenetic and recombination analysis.

CSV
Download CSV
thumbnail
Figure 1. The Neighbor-joining tree of all the 629 CVA16 viruses based on complete VP1 encoding sequences.

The branches showed in red color highlight the sequences from China.

https://doi.org/10.1371/journal.pone.0082861.g001

Among 607 genotype B1 strains, there were 387, 200, and 20 strains could be divided into subgenotypes B1a, B1b, and B1c, respectively, by phylogenetic analysis (Figure 1). In more detail, B1a was the most prevalent one, after its first appearance in 1995 in Japan, it has been circulating in 11 of the 12 countries (regions) involved in this study, except Saudi Arabia. B1b was reported in mainland China, Taiwan, Japan, Malaysia, Australia, and Saudi Arabia during 1998-2011. Circulation of B1c was only detected in Malaysia from 2005-2007, and in France in 2010 (Figure 2).

thumbnail
Figure 2. Global distribution of subgenotypes of CVA16.

The time and spatial distribution of all genotypes, except genotype A, were shown in this chart. All the 628 genotype B strains from all other countries were shown in this chart except CVA16 prototype strain. Bar in blue, red, purple, and green indicate the subgenotype B2, B1a, B1b, and B1c, respectively. The length of the bar correlated with the numbers of the isolates in corresponding year.

https://doi.org/10.1371/journal.pone.0082861.g002

In our study, most of the sequence data (582/629, 92.5%) were reported from Japan, Malaysia, and China. In these three countries, B1a (357/582, 61%) was the most reported genotype, which has been persistently circulating for more than 15 years (Figure 2). In mainland China, B1b was another dominant genotype (169/293, 58%) which has been co-circulating with B1a (124/293, 42%) since 2000. In Malaysia, besides B1a, B1c was also consecutively identified during 2005-2007. While in Japan, although B1b had co-circulated with B1a during 1998-2004, B1a has become the predominant genotype after 2004 (Figure 2).

Analysis of complete genomic sequences of CVA16

For further genomic analysis, a total of 35 complete genome sequences of CVA16 were used, including one prototype, 8 from our laboratory, and 27 retrieved from public database. Among the 35 genome sequences, there were 26 sequences from mainland China, 3 from Malaysia, 2 each from Hong Kong and Thailand, and 1 each from South Korea and Taiwan (Table 1). Phylogenetically, these 35 isolates were grouped into subgenotype B1a (18) and B1b (17) based on complete VP1 region, and all of the B1a sequences were from China (Figure 1, Figure S1a).

Compared to the CVA16 prototype (U05876), there were no deletions or insertions observed in the P1, P2, and P3 encoding regions. Nucleotide substitutions among different CVA16 strains are scattered throughout the genome. Pairwise nucleotide sequence identities are 89.4%-98.9% among the 35 isolates, 91.4%-98.5% and 93.2-98.9% within groups B1a and B1b, respectively. Pairwise nucleotide sequence identities based on each gene were also calculated (Table S4). In the structural capsid protein encoding regions, VP1-VP4, nucleotide sequence identities within subgenotype B1a and B1b are more than 91.3% and 92.2%, respectively. Comparatively, sequences in the non-structural protein encoding regions showed more diversities, especially in 3B (Table S4), the least identity in which is 81.8%. Estimations of mean genetic distance also indicated high homology in the whole genome, which were 0.049 within B1a, 0.043 within B1b (Table 2). The mean nucleotide distance between B1a and B1b is 0.086, which is significantly larger than mean genetic distance within groups. The mean amino acid distance is much smaller, which was 0.009 within both B1a and B1b, and 0.013 between B1a and B1b (Table 2). Both the values in sequence identity and genetic distance suggested a near phylogenetic relationship of sequences within both B1a and B1b.

Regiondistance
B1aB1bB1Between B1a and B1b
nt aant aant aant aa
complete0.049 0.0090.043 0.0090.066 0.0110.086 0.013
5UTR*0.037 --0.030 --0.045 --0 .050 --
P10.048 0.0050.044 0.0050.066 0.0050.086 0.005
VP40.047 0.0050.030 0.0120.054 0.0080.069 0.008
VP20.056 0.0090.045 0.0030.070 0.0060.088 0.006
VP30.045 0.0010.049 0.0050.070 0.0030.092 0.003
VP10.045 0.0040.042 0.0040.063 0.0040.092 0.004
P20.050 0.0080.040 0.0080.066 0.0100.085 0.012
2A0.056 0.0170.039 0.0170.039 0.0200.088 0.022
2B0.048 0.0110.042 0.0110.059 0.0110.072 0.012
2C0.047 0.0030.041 0.0030.066 0.0050.087 0.008
P30.053 0.0160.049 0.0160.074 0.0210.096 0.025
3A0.052 0.0150.048 0.0180.077 0.0310.103 0.045
3B0.053 0.0240.067 0.0240.101 0.0240.140 0.023
3C0.060 0.0190.045 0.0100.077 0.0160.099 0.018
3D0.050 0.0150.050 0.0180.071 0.0200.092 0.024

Table 2. Mean genetic distances based on corresponding regions of 35 complete genomic sequences and amino acid sequences of CVA16.

Note: Kimura-2-parameter nucleotide substitution model was used for estimation of mean genetic distance of the nucleotide sequences, and p-distance, which is the proportion (p) of amino acid sites at which the two sequences to be compared are different, for mean genetic distance of the amino acid sequences. *: Pairwise comparisons not including THA-CA16-090/Thailand/2010, THA-CA16-069/Thailand/2010, and HQ09011181/YN/CHN/2011 for partial missings in 5’UTR. Abbreviation: nt, nucleotide; aa, amino acid.
CSV
Download CSV

Phylogenetic dendrograms based on the whole genomes, protein encoding regions, and each specific gene were performed with MEGA 5.0 software as shown in Figure 3 and Figure S1-2. The grouping of B1a and B1b in phylogenetic trees based on P1, P2, and P3 regions were different (Figure 3), which suggested potential intratypic recombinations between B1a and B1b had occurred. This has also been confirmed by phylogenetic analyses on each specific gene (Figure S1-2). Several sequences of subgenotype B1a clustered with B1b on VP2-VP4 (Figure S1), and the grouping on 2A-2C, and 3A-3D were more diverse (Figure S2).

thumbnail
Figure 3. Phylogenetic dendrogram showing the relationships between the cluster B1a, B1b and EV-A prototypes.

These were constructed by the different genomic regions of 35 complete CVA16 genomic sequences and EV-A prototype sequences. The neighbour-joining trees were reconstructed based on the P1 (a), P2 (b), P3 (c) genomic regions and complete genomes (d), respectively. Triangles indicated the subgenotype B1a. Rhombuses indicated the subgenotype B1b. Squares indicated the CVA16 prototype G-10 strain. The percentage of bootstrap (percentage of 1000 pseudoreplicate datasets) supporting the trees are indicated at the nodes; only values over 80% are shown.

https://doi.org/10.1371/journal.pone.0082861.g003

Moreover, a potential intertypic recombination between the 35 genomic sequences of CVA16 and other serotypes of EV-A was also observed. Based on nucleotide sequences of P1 and its encoding proteins, all of the 35 sequences displayed nearest phylogenic relationship with CVA16 prototype (Figure 3a, Figure S1). However, based on other non-capsid protein encoding regions and 5’ UTR, all sequences exhibited further distance in evolutionary origin with CVA16 than other EV-A serotypes. In dendrograms based on P2 and P3, all 35 sequences clustered with a clade including the prototypes of EV-A71, CVA2, CVA3, CVA6, CVA10 and CVA12 but not CVA16 (Figures 3b, 3c). The different topology of the dendrograms between P1 and P2-P3 regions suggested a potential intertypic recombination of these circulating CVA16 strains with other EV-A serotypes in non-structural protein encoding regions. This deduction could also be supported by the nucleotide sequence identities with every prototype of EV-A. In both P1 and mature proteins VP1-VP4 encoding regions, sequence identities with CVA16 prototype were 76.5%-78.0%, 75.1%-77.1%, 75.1%-78.8%, 76.7%-79.2% and 79.2%-83.5%, respectively (Table 3), these values are consistent with the 75% rule of enterovirus typing [21]. But in P2 and P3 regions, sequences in our study showed significantly higher homology to EV-A71, CVA2, CVA3, CVA6, CVA10 and CVA12, ranging from 81.1%-84.3% and 81.6%-84.1%, than to CVA16 and other prototypes, which were 79.4%-80.3% and 76.9%-78.1%, respectively (Table 3).

RegionsIdentity (%)
G-10EV-A71CVA2CVA3CVA4CVA5CVA6CVA7CVA8CVA10CVA12CVA14EV-A76EV-A89EV-A90EV-A91EV-A92
Genome77.3-79.176.7-79.174.3-76.174.5-76.471.8-73.772.6-74.874.2-76.074.1-75.872.6-74.574.5-76.574.7-76.672.5-74.568.2-69.368.8-70.167.2-68.668.0-69.766.6-68.0
5UTR74.2-86.572.4-83.971.5-82.374.7-85.775.5-87.772.6-83.973.7-86.273.3-84.070.9-82.672.4-84.173.6-85.573.9-86.367.7-78.467.1-78.060.5-70.361.2-71.158.8-69.3
P176.5-78.069.0-69.962.8-64.161.7-62.861.5-62.663.2-64.461.6-62.264.9-66.062.8-63.764.3-65.364.1-65.165.5-66.362.8-6463.8-65.162.8-63.564.8-65.863.1-64.4
VP479.2-83.569.0-72.463.2-67.162.8-65.764.2-66.660.3-63.762.3-65.262.8-65.759.9-62.862.8-66.661.3-64.760.3-63.763.7-66.661.8-64.264.2-67.666.1-70.060.3-63.7
VP275.1-78.870.6-72.764.5-66.464.7-66.565.6-67.365.7-67.565.4-68.368.5-70.765.0-67.466.9-69.268.7-70.868.9-70.564.8-66.564.8-66.663.7-66.166.2-68.466.0-68.3
VP376.7-79.270.3-72.366.3-69.063.3-65.263.3-65.466.7-68.964.3-66.567.7-69.964.7-66.565.2-67.365.6-67.668.1-70.165.9-68.568.0-70.369.2-70.967.7-70.566.0-68.8
VP175.1-77.164.5-66.357.0-59.256.7-58.355.2-56.855.6-57.053.8-56.058.2-59.559.0-60.459.8-61.456.1-57.959.7-62.058.2-59.560.9-62.657.9-59.760.5-62.558.3-60.8
P279.4-80.382.6-84.381.5-83.381.4-83.477.3-78.777.9-79.181.4-83.379.4-80.678.3-80.181.1-82.881.9-83.778.4-79.870.3-72.171.3-73.170.1-71.271.1-72.470.9-72.2
2A 79.1-81.778.2-81.178.4-81.179.3-82.875.1-78.479.5-82.678.2-80.476.0-79.775.7-80.479.3-82.077.7-81.177.3-81.164.2-66.468.0-71.362.0-64.864.6-68.064.6-67.1
2B75.7-78.780.8-85.179.7-83.179.7-82.875.0-78.175.0-78.780.8-84.576.0-78.477.1-79.780.4-84.180.8-83.576.0-79.162.2-67.363.2-67.069.0-72.065.9-70.370.7-73.4
2C 79.6-81.384.4-86.582.7-85.582.7-84.478.0-79.976.6-78.882.0-85.381.0-82.578.9-80.981.1-83.683.2-86.078.7-80.775.2-76.774.8-76.473.4-74.574.7-76.072.8-74.7
P376.9-78.182.2-84.182.2-83.782.5-83.776.4-78.678.7-80.982.2-83.979.2-81.377.7-79.381.7-83.381.6-83.476.4-77.571.2-73.072.2-73.671.8-73.271.1-72.970.1-71.5
3A 79.4-81.783.3-87.980.2-86.481.7-85.277.9-81.375.9-79.481.7-86.477.9-81.076.3-79.881.0-86.081.7-86.877.1-81.367.8-73.268.9-73.668.6-72.067.8-72.062.7-67.0
3B71.2-81.877.2-87.880.3-87.880.3-89.374.2-83.368.1-74.280.3-89.366.6-75.769.6-77.278.7-86.378.7-87.871.2-80.368.1-75.766.6-75.760.6-68.159.0-68.160.6-72.7
3C 73.4-76.682.6-84.881.2-83.281.9-84.675.0-77.976.1-78.881.2-83.276.6-81.075.5-77.280.1-82.681.2-84.374.4-77.070.6-72.672.1-74.169.5-73.071.0-73.769.0-73.0
3D76.9-78.481.3-84.081.7-84.381.8-83.976.1-78.780.3-83.181.6-84.380.1-82.378.4-80.881.6-84.080.7-83.675.9-77.571.3-73.872.1-74.072.7-74.571.6-73.871.2-72.8

Table 3. Pairwise nucleotide sequences identities between 35 genomic sequences and prototype strains of EV-A species.

CSV
Download CSV

Phylogenetic trees from each of mature protein encoding regions of P2 (2A-2C) and P3 (3A-3D), indicated more details of this inconsistency. Based on each specific region of P2 and P3, all the sequences in our study clustered along with the same clade in P2 and P3, except in 2A. In the later case, sequences in our study still showed nearest phylogenetic relationship with the prototype of CVA16, but indicating a low bootstrap value (Figure S2a). These data suggested a heterogenous origin of 2B, 2C, 3A, 3B, 3C and 3D in all 35 sequences (Figure 3, Figure S2). Calculated values of nucleotide sequence identities in the 2B, 2C, 3A, 3B, 3C and 3D regions also showed consistency with phylogenetic dendrograms. While in 2A region, the sequence identities showed no significant difference between CVA16 and others, suggesting a multi-source in 2A of our sequences (Table 3), and possibly the site which intertypic recombination occurred.

To further depict the possible recombination of these strains, similarity plot and bootscanning analyses were performed by using EV-A prototype strains as reference sequences. Four strains, Tainan/5079/98/Taiwan/1998, shzh00-1/GD/CHN/2000, BJ/11/11/BJ/CHN/2011, and BJ/11/12/BJ/CHN/2011, were chosen as the representative strains. These represented the oldest and the latest CVA16 strains of subgenotypes B1a and B1b, respectively (Figure 4). As indicated in the plots, these 4 sequences showed definitely the highest similarity and the nearest phylogenetic relationship with CVA16 prototype in P1 region. However, in P2 and P3 regions, all 4 strains contained some unidentified regions that showed a little higher similarity with prototypes of EV-A71, CVA2, CVA3, CVA6, CVA10 and CVA12, but apparently not CVA16 (Figures 4a, 4c, 4e, and 4g). The bootscanning showed a nearer phylogenetic relationship with EV-A71 (Figures 4b, 4d, 4f, and 4h). An obvious single crossing site in the 2A region suggested a potential recombination occurred which is consistent with deduction above.

thumbnail
Figure 4. Similarity plot and bootscanning analyses of all the two subgenotypes B1a, B1b of CVA16 and other EV-A prototype strains on the basis of full-length genomes.

Two strains were chosen as the representative strains of each subgenotype, respectively. Each of the four viruses was used as the query sequence. A sliding window of 500 nucleotides moving in 20 nucleotides steps was used in this analysis. (a and b) Tainan/5079/98/Taiwan/1998; (c and d) BJ/11/11 /BJ/CHN/2011; (e and f) shzh00-1/GD/CHN/2000; (g and h) BJ11/12/BJ/CHN/2011. The a, c, e, g were the results of similarity plot analyses; the b, d, f, h were the results of bootscanning analyses.

https://doi.org/10.1371/journal.pone.0082861.g004

Discussion

CVA16 was involved in a significantly high transmission among the children with HFMD and herpangina. The symptoms caused by EV-A71 and CVA16 are difficult to differentiate clinically. The co-circulation or alternating circulation of CVA16 and EV-A71 had been proven to have contributed to the outbreaks of HFMD [8,22,23]. CVA16 was responsible for about 50% of all the confirmed enterovirus infections among HFMD cases in mainland China [8,24-26]. The infection by CVA16 was usually mild and benign when compared to EV-A71 infection, however, severe complications such as myocarditis and pericarditis, even death could occur [14,15].

There was lack of systematic epidemiologic analysis on CVA16 in previous studies. We studied the molecular epidemiology of CVA16 by analyzing all the complete VP1 sequences from GenBank database, and clarified the global distribution of the CVA16 genotypes. All CVA16 strains belonged to the genotype B, except the CVA16 prototype, which was the unique strain in genotype A. From 1981 to 2000, majority of the CVA16 strains classified as genotype B2 were only detected in Japan and Malaysia. The subgenotypes B1a and B1b strains were first isolated in 1995 and 1998, respectively, in Japan. These two subgentypes then became the predominant ones circulating globally since 2000. In mainland China, all the CVA16 strains isolated belonged to subgenotypes B1a and B1b. Both subgenotypes could be detected in most of the provinces. However, only single subgenotype B1b was detected in Shanghai, Shaanxi, Jilin, Fujian, Tianjin and Inner Mongolia. There has no subgenotype B1c been detected in China, so the surveillance on this new subgenotype must be continually reinforced.

In this study, high nucleotide sequence identities were observed among all 35 CVA16 complete genome sequences and in their respective encoding regions. Phylogenetic analyses also indicated that these 35 CVA16 strains consistently clustered together in all of the analyzed regions. These result suggested that all 35 CVA16 sequences had high homogeneity with a little internal differences. This revealed that CVA16 strains might have evolved independently, steadily and originated from a same ancestor. It was determined that neutralization epitopes resided mainly on capsid proteins, especially protein VP1 [27]. In our results, the amino acid sequence identities in the P1 and VP1 regions of these 35 CVA16 strains were 99.5% and 99.6%, respectively. Further amino acid sequence comparisons revealed that the field epidemic CVA16 strains also had high sequence identities in protein P1 and detailed VP1-VP4 encoding regions, both within or between the subgenotypes B1a and B1b strains (Table 2). CVA16 immunity might cross-react with EV-A71 [28], however, the amino acid sequence identities in the P1 and VP1-VP4 regions were 79.3%, 71.9%, 84.2%, 85.4%, 78.1% between EV-A71 subgenotype C4 representative strain (JN256060) and prevalent CVA16 strains in our study (data not shown). So, the cross-react neutralization epitopes mighty reside on the proteins VP2 and VP3. The genetic stability in the structural proteins and simplex genotype transmission in China are very important for designing serological diagnostic test and vaccine to the prevalent CVA16 strains.

This study demonstrated that the current CVA16 strains were potential multiple-recombinant viruses containing other EV-A donor sequences. Recombination was frequently detected in the P2 and P3 regions while it was rare in P1 region, probably because of the structural proteins was important for the virus replication and assembling of proper viral capsid [29-31]. Genetic recombination sometimes could result in the virulence change of enteroviruses, which could trigger serious public health problems [32]. Because the multiple intertypic recombinations occurred in the P2 and P3 regions long time before and only few complete sequences of CVA16 are available in GenBank database, so the phylogenetic analyses are limited and hard to determine the donor strains.

These isolated CVA16 strains from HFMD outbreaks in mainland China recently were the sustained circulation of the same B1a and B1b strains from a common ancestor without clearly new genetic recombination and with a slow evolutionary rate. To elucidate further detailed genetic characteristics and evolutionary recombination events in CVA16, continually strengthen on surveillance is necessary.

Materials and Methods

Ethics Statement

This study did not involve human experimentation or human participants; the only human materials used were stool, throat swab, and vesicles samples collected from HFMD patients at the instigation of the Ministry of Health P. R. of China (PRC) for public health purposes. HFMD is a notifiable disease in China, and the pathogenic surveillance of HFMD without private information referred is required by the Law of the PRC on the Prevention and Treatment of Infectious Diseases. According to this law, these data used in this study were obtained from samples as part of this monitoring program. Thus, the requirement for written informed consent was waived by the Ethics Review Committee of the National Institute for Viral Disease Control and Prevention. This study was approved by the Ethics Review Committee of the National Institute for Viral Disease Control and Prevention. The CVA16 isolates involved in this study were isolated from the clinical samples of HFMD patients in 2010. These patients are from different geographical locations in the Tibet Autonomous Region, Zhejiang, Jiangsu and Anhui provinces of China.

Viruses

In this study, eight CVA16 strains were isolated from eight HFMD patients in mainland China in 2010 respectively. Patients were children with the age range from 2 to 14 years old. Viruses were isolated from throat swabs, feces, and vesicle fluid specimens on human rhabdomyosarcoma (RD) cell line.

Determination of the genomes

The complete genome of the 8 CVA16 isolates mentioned above were determined in our laboratory. Viral RNA was extracted using a QIAamp Viral RNA Mini Kit (Qiagen, Valencia, CA, USA). First, the viral RNA was converted to cDNA by a random priming strategy. The cDNA was amplified using the primers designed by multiple alignments of CVA16 genomes available in GenBank database (Table S1). PCR products were purified for sequencing using the QIAquick Gel extraction kit (Qiagen). Then the amplicons were bi-directionally sequenced using fluorescent dideoxy terminators and an ABI PRISM 3100 genetic analyzer (Applied Biosystems Foster City, CA, USA).

Sequences from GenBank

Totally 593 complete VP1 encoding sequences (Table S2) and 28 complete genome sequences (Table 1) of CVA16, which released to public database before December 2012, were downloaded from GenBank. The sequences of prototypes of EV-A group (CVA2, 3, 4, 5, 6, 7, 8, 10, 12, 14, EV-A71, 76, 89, 90, 91 and 92) were used as references in the phylogenetic and recombination analysis (Table S3). These 28 genome sequences of CVA16 that downloaded from GenBank database included 20 from mainland China and 8 from Malaysia (3), Thailand (2), Korea (1), Taiwan (1), and South Africa (1) from 1998 to 2011.

Bioinformatics and Phylogenetic analysis

Trimming of the sequencing chromatogram and sequences assembly were preformed with Sequencher program (version 4.0.5 Gene Codes Corporation, USA). Phylogenetic trees were reconstructed with MEGA program (version 5.0; Sudhir Kumar, Arizona State University, Tempe, Arizona, USA) [33], using the neighbor-joining method and Kimura-2-parameter substitution model. (Note: Maximum likelihood method has also been used to construct the phylogenetic trees to increases the reliability, the results were more or less the same as neighbor-joining method. Data not shown.) Bootstrap testing with 1000 replicates were used to estimate the strength of the phylogenetic trees. Bootstrap values greater than 80% were considered to be a strong support for the tree topology. Completely sequenced 28 CVA16 isolates available in the GenBank, 8 isolates from our laboratory and other EV-A prototype strains were aligned and used for construction of phylogenetic trees and recombiantion analyses. The genetic distance and identity matrices were determined by group mean and pairwise estimation of the sequences divergence with MEGA program [34].

Recombination analysis

Alignment of the complete genome sequences were performed with the ClustalW package in MEGA program (version 5.0; Sudhir Kumar, Arizona State University, Tempe, Arizona, USA) [33]. Potential recombination between CVA16 and other EV-A strains was scanned by Similarity plot and bootscan method (version 3.5.1; Stuart Ray, Johns Hopkins University, Baltimore, Maryland, USA) [35]. The neighbor-joining method and Kimura 2-parameter substitution model were used. A sliding window size of 500bp nucleotides moving in 20bp nucleotides at a time was used in this analysis.

Nucleotide sequence accession numbers

Complete genome sequences of the 8 CVA16 strains have been submitted to the GenBank database with the accession numbers KC755228 to KC755235.

Supporting Information

Table S1.

List of nucleotide sequences of primer for amplification of the whole genome sequences of CVA16.

https://doi.org/10.1371/journal.pone.0082861.s001

(DOCX)

Table S2.

List of the 593 complete VP1 sequences of CVA16 strains available from GenBank which were selected to generate the CVA16 phylogenetic dendrograms.

https://doi.org/10.1371/journal.pone.0082861.s002

(DOCX)

Table S3.

List of the EV-A prototype strains used for the sequence comparison, phylogenetic and recombination analysis.

https://doi.org/10.1371/journal.pone.0082861.s003

(DOCX)

Table S4.

Pairwise nucleotide sequence identities based on corresponding regions of 35 complete genomic sequences of CVA16.

https://doi.org/10.1371/journal.pone.0082861.s004

(DOCX)

Figure S1.

Phylogenetic dendrograms showing the relationships amongst EV-A isolates using the different genomic regions. Phylogenetic dendrogram showing the relationships between the cluster B1a, B1b and EV-A prototypes by using the different genomic regions of 35 complete CVA16 genomic sequences and EV-A prototype sequences. The neighbour-joining trees were constructed from alignment of the VP1 (a), VP2 (b), VP3 (c) and VP4 (d) genomic regions, respectively. Triangles indicated the subgenotype B1a. Rhombuses indicated the subgenotype B1b. Squares indicated the CVA16 prototype G-10 strain. The percentage of bootstrap (percentage of 1000 pseudoreplicate datasets) replicates supporting the trees are indicated at the nodes; for clarity, only values over 80% are shown. The branch lengths are proportional to the genetic distances corrected using Kimura-two-parameter substitution model.

https://doi.org/10.1371/journal.pone.0082861.s005

(EPS)

Figure S2.

Phylogenetic dendrograms showing the relationships amongst EV-A isolates using the different genomic regions. Phylogenetic dendrogram showing the relationships between the cluster B1a, B1b and EV-A prototypes by using the different genomic regions of 35 complete CVA16 genomic sequences and EV-A prototype sequences. The neighbour-joining trees were constructed from alignment of the 2A (a), 2B (b), 2C (c), 3A (d), 3B (e), 3C (f), 3D (g) and 5’UTR (h) genomic regions, respectively. Triangles indicated the subgenotype B1a. Rhombuses indicated the subgenotype B1b. Squares indicated the CVA16 prototype G-10 strain. The percentage of bootstrap (percentage of 1000 pseudoreplicate datasets) replicates supporting the trees are indicated at the nodes; for clarity, only values over 80% are shown. The branch lengths are proportional to the genetic distances corrected using Kimura-two-parameter substitution model.

https://doi.org/10.1371/journal.pone.0082861.s006

(EPS)

Acknowledgments

We would like to acknowledge all the provincial laboratory staffs for collecting clinical specimens from the patients used in this study. And we thank all the reviewers for comments that improved this manuscript.

Author Contributions

Conceived and designed the experiments: XC WX. Performed the experiments: XC XT JL YJ LG MH YS S. Zhu BZ S. Zhang YZ. Analyzed the data: XC XT. Contributed reagents/materials/analysis tools: XC JL YJ LG MH YS NM WX. Wrote the manuscript: XC XT.

References

  1. 1. Zhang Y, Tan XJ, Wang HY, Yan DM, Zhu SL et al. (2009) An outbreak of hand, foot, and mouth disease associated with subgenotype C4 of human enterovirus 71 in Shandong. China. J - Journal of Clin Virol 44: 262-267. doi:https://doi.org/10.1016/j.jcv.2009.02.002.
  2. 2. Huang CC, Liu CC, Chang YC, Chen CY, Wang ST et al. (1999) Neurologic complications in children with enterovirus 71 infection. N Engl J Med 341: 936-942. doi:https://doi.org/10.1056/NEJM199909233411302. PubMed: 10498488.
  3. 3. Solomon T, Lewthwaite P, Perera D, Cardosa MJ, McMinn P et al. (2010) Virology, epidemiology, pathogenesis, and control of enterovirus 71. Lancet Infect Dis 10: 778-790. doi:https://doi.org/10.1016/S1473-3099(10)70194-8. PubMed: 20961813.
  4. 4. Komatsu H, Shimizu Y, Takeuchi Y, Ishiko H, Takada H (1999) Outbreak of severe neurologic involvement associated with Enterovirus 71 infection. Pediatr Neurol 20: 17-23. doi:https://doi.org/10.1016/S0887-8994(98)00087-3. PubMed: 10029254.
  5. 5. Tan X, Huang X, Zhu S, Chen H, Yu Q et al. (2011) The persistent circulation of enterovirus 71 in People's Republic of China: causing emerging nationwide epidemics since 2008. PLOS ONE 6: e25662. doi:https://doi.org/10.1371/journal.pone.0025662. PubMed: 21980521.
  6. 6. Brown BA, Oberste MS, Alexander JP Jr., Kennett ML, Pallansch MA (1999) Molecular epidemiology and evolution of enterovirus 71 strains isolated from 1970 to 1998. J Virol 73: 9969-9975. PubMed: 10559310.
  7. 7. Chang LY, Lin TY, Huang YC, Tsao KC, Shih SR et al. (1999) Comparison of enterovirus 71 and coxsackie-virus A16 clinical illnesses during the Taiwan enterovirus epidemic, 1998. Pediatr Infect Dis J 18: 1092-1096. doi:https://doi.org/10.1097/00006454-199912000-00013. PubMed: 10608631.
  8. 8. Li L, He Y, Yang H, Zhu J, Xu X et al. (2005) Genetic characteristics of human enterovirus 71 and coxsackievirus A16 circulating from 1999 to 2004 in Shenzhen, People's Republic of China. J Clin Microbiol 43: 3835-3839. doi:https://doi.org/10.1128/JCM.43.8.3835-3839.2005. PubMed: 16081920.
  9. 9. McMinn PC (2012) Recent advances in the molecular epidemiology and control of human enterovirus 71 infection. Curr Opin Virol 2: 199-205. doi:https://doi.org/10.1016/j.coviro.2012.02.009. PubMed: 22482716.
  10. 10. Shimizu H, Utama A, Onnimala N, Li C, Li-Bi Z et al. (2004) Molecular epidemiology of enterovirus 71 infection in the Western Pacific. Region - Pediatr Int 46: 231-235. doi:https://doi.org/10.1046/j.1442-200x.2004.01868.x.
  11. 11. Zhao K, Han X, Wang G, Hu W, Zhang W et al. (2011) Circulating coxsackievirus A16 identified as recombinant type A human enterovirus, China. Emerg Infect Dis 17: 1537-1540. PubMed: 21801645.
  12. 12. Chen L, Mou X, Zhang Q, Li Y, Lin J et al. (2012) Detection of human enterovirus 71 and coxsackievirus A16 in children with hand, foot and mouth disease in China. Mol Med Report 5. : 1001-1004.
  13. 13. Ma E, Fung C, Yip SH, Wong C, Chuang SK et al. (2011) Estimation of the basic reproduction number of enterovirus 71 and coxsackievirus A16 in hand, foot, and mouth disease outbreaks. Pediatr Infect Dis J 30: 675-679. doi:https://doi.org/10.1097/INF.0b013e3182116e95. PubMed: 21326133.
  14. 14. Wang CY, Li Lu F, Wu MH, Lee CY, Huang LM (2004) Fatal coxsackievirus A16 infection. Pediatr Infect Dis J 23: 275-276. doi:https://doi.org/10.1097/01.inf.0000115950.63906.78. PubMed: 15014311.
  15. 15. Legay F, Lévêque N, Gacouin A, Tattevin P, Bouet J et al. (2007) Fatal coxsackievirus A-16 pneumonitis in adult. Emerg Infect Dis 13: 1084-1086. doi:https://doi.org/10.3201/eid1307.070295. PubMed: 18214187.
  16. 16. Svitkin YV, Imataka H, Khaleghpour K, Kahvejian A, Liebig HD et al. (2001) Poly(A)-binding protein interaction with elF4G stimulates picornavirus IRES-dependent translation. Rna 7: 1743-1752. PubMed: 11780631.
  17. 17. Palmenberg AC (1990) Proteolytic processing of picornaviral polyprotein. Annu Rev Microbiol 44: 603-623. doi:https://doi.org/10.1146/annurev.mi.44.100190.003131. PubMed: 2252396.
  18. 18. Arnold E, Luo M, Vriend G, Rossmann MG, Palmenberg AC et al. (1987) Implications of the picornavirus capsid structure for polyprotein processing. Proc Natl Acad Sci U S A 84: 21-25. doi:https://doi.org/10.1073/pnas.84.1.21. PubMed: 3467351.
  19. 19. Zhang Y, Wang D, Yan D, Zhu S, Liu J et al. (2010) Molecular evidence of persistent epidemic and evolution of subgenotype B1 coxsackievirus A16-associated hand, foot, and mouth disease in China. J Clin Microbiol 48: 619-622. doi:https://doi.org/10.1128/JCM.02338-09. PubMed: 20018819.
  20. 20. Oberste MS, Peñaranda S, Maher K, Pallansch MA (2004) Complete genome sequences of all members of the species Human enterovirus A. J Gen Virol 85: 1597-1607. doi:https://doi.org/10.1099/vir.0.79789-0. PubMed: 15166444.
  21. 21. Oberste MS, Maher K, Kilpatrick DR, Pallansch MA (1999) Molecular evolution of the human enteroviruses: correlation of serotype with VP1 sequence and application to picornavirus classification. J Virol 73: 1941-1948. PubMed: 9971773.
  22. 22. Hosoya M, Kawasaki Y, Sato M, Honzumi K, Hayashi A et al. (2007) Genetic diversity of coxsackievirus A16 associated with hand, foot, and mouth disease epidemics in Japan from 1983 to 2003. J Clin Microbiol 45: 112-120. doi:https://doi.org/10.1128/JCM.00718-06. PubMed: 17093028.
  23. 23. Ang LW, Koh BK, Chan KP, Chua LT, James L et al. (2009) Epidemiology and control of hand, foot and mouth disease in Singapore, 2001-2007. Ann Acad Med Singapore 38: 106-112. PubMed: 19271036.
  24. 24. De W, Changwen K, Wei L, Monagin C, Jin Y et al. (2011) A large outbreak of hand, foot, and mouth disease caused by EV71 and CAV16 in Guangdong, China, 2009. Arch Virol 156: 945-953. doi:https://doi.org/10.1007/s00705-011-0929-8. PubMed: 21305327.
  25. 25. Zhu Z, Zhu S, Guo X, Wang J, Wang D et al. (2010) Retrospective seroepidemiology indicated that human enterovirus 71 and coxsackievirus A16 circulated wildly in central and southern China before large-scale outbreaks from 2008. Virol J 7: 300. doi:https://doi.org/10.1186/1743-422X-7-300. PubMed: 21050463.
  26. 26. Jia L, Zhao CS, Zhang L, Li S, Zhang DT et al. (2011) Comparisons of epidemiological and clinical characteristics in children with hand-foot-mouth disease caused by Enterovirus 71 and Coxackievirus A16. Zhongguo Dang Dai Er Ke za Zhi 13: 635-637. PubMed: 21849112.
  27. 27. Liu Q, Ku Z, Cai Y, Sun B, Leng Q et al. (2011) Detection, characterization and quantitation of coxsackievirus A16 using polyclonal antibodies against recombinant capsid subunit proteins. J Virol Methods 173: 115-120. doi:https://doi.org/10.1016/j.jviromet.2011.01.016. PubMed: 21295614.
  28. 28. Wu TC, Wang YF, Lee YP, Wang JR, Liu CC et al. (2007) Immunity to avirulent enterovirus 71 and coxsackie A16 virus protects against enterovirus 71 infection in mice. J Virol 81: 10310-10315. doi:https://doi.org/10.1128/JVI.00372-07. PubMed: 17626076.
  29. 29. McWilliam Leitch EC, Cabrerizo M, Cardosa J, Harvala H, Ivanova OE et al. (2012) The association of recombination events in the founding and emergence of subgenogroup evolutionary lineages of human enterovirus 71. J Virol 86: 2676-2685. doi:https://doi.org/10.1128/JVI.06065-11. PubMed: 22205739.
  30. 30. Chen X, Zhang Q, Li J, Cao W, Zhang JX et al. (2010) Analysis of recombination and natural selection in human enterovirus 71. Virology 398: 251-261. doi:https://doi.org/10.1016/j.virol.2009.12.007. PubMed: 20060995.
  31. 31. Wang X, Zhu C, Bao W, Zhao K, Niu J et al. (2012) Characterization of full-length enterovirus 71 strains from severe and mild disease patients in northeastern China. PLOS ONE 7: e32405. doi:https://doi.org/10.1371/journal.pone.0032405. PubMed: 22479324.
  32. 32. Zhang Y, Wang J, Guo W, Wang H, Zhu S et al. (2011) Emergence and transmission pathways of rapidly evolving evolutionary branch C4a strains of human enterovirus 71 in the Central Plain of China. PLOS ONE 6: e27895. doi:https://doi.org/10.1371/journal.pone.0027895. PubMed: 22125635.
  33. 33. Tamura K, Peterson D, Peterson N, Stecher G, Nei M et al. (2011) MEGA5: molecular evolutionary genetics analysis using maximum likelihood, evolutionary distance, and maximum parsimony methods. Mol Biol Evol 28: 2731-2739. doi:https://doi.org/10.1093/molbev/msr121. PubMed: 21546353.
  34. 34. Thompson JD, Gibson TJ, Plewniak F, Jeanmougin F, Higgins DG (1997) The CLUSTAL_X windows interface: flexible strategies for multiple sequence alignment aided by quality analysis tools. Nucleic Acids Res 25: 4876-4882. doi:https://doi.org/10.1093/nar/25.24.4876. PubMed: 9396791.
  35. 35. Lole KS, Bollinger RC, Paranjape RS, Gadkari D, Kulkarni SS et al. (1999) Full-length human immunodeficiency virus type 1 genomes from subtype C-infected seroconverters in India, with evidence of intersubtype recombination. J Virol 73: 152-160. PubMed: 9847317.