Skip to main content
  • Research article
  • Open access
  • Published:

Identification of a novel bovine enterovirus possessing highly divergent amino acid sequences in capsid protein

Abstract

Background

Bovine enterovirus (BEV) belongs to the species Enterovirus E or F, genus Enterovirus and family Picornaviridae. Although numerous studies have identified BEVs in the feces of cattle with diarrhea, the pathogenicity of BEVs remains unclear. Previously, we reported the detection of novel kobu-like virus in calf feces, by metagenomics analysis. In the present study, we identified a novel BEV in diarrheal feces collected for that survey. Complete genome sequences were determined by deep sequencing in feces. Secondary RNA structure analysis of the 5′ untranslated region (UTR), phylogenetic tree construction and pairwise identity analysis were conducted.

Results

The complete genome sequences of BEV were genetically distant from other EVs and the VP1 coding region contained novel and unique amino acid sequences. We named this strain as BEV AN12/Bos taurus/JPN/2014 (referred to as BEV-AN12). According to genome analysis, the genome length of this virus is 7414 nucleotides excluding the poly (A) tail and its genome consists of a 5′UTR, open reading frame encoding a single polyprotein, and 3′UTR. The results of secondary RNA structure analysis showed that in the 5′UTR, BEV-AN12 had an additional clover leaf structure and small stem loop structure, similarly to other BEVs. In pairwise identity analysis, BEV-AN12 showed high amino acid (aa) identities to Enterovirus F in the polyprotein, P2 and P3 regions (aa identity ≥82.4%). Therefore, BEV-AN12 is closely related to Enterovirus F. However, aa sequences in the capsid protein regions, particularly the VP1 encoding region, showed significantly low aa identity to other viruses in genus Enterovirus (VP1 aa identity ≤58.6%). In addition, BEV-AN12 branched separately from Enterovirus E and F in phylogenetic trees based on the aa sequences of P1 and VP1, although it clustered with Enterovirus F in trees based on sequences in the P2 and P3 genome region.

Conclusions

We identified novel BEV possessing highly divergent aa sequences in the VP1 coding region in Japan. According to species definition, we proposed naming this strain as “Enterovirus K”, which is a novel species within genus Enterovirus. Further genomic studies are needed to understand the pathogenicity of BEVs.

Background

Bovine enterovirus (BEV) is a single positive-stranded RNA virus belonging to the genus Enterovirus within family Picornaviridae. The viral particle is composed of a small, non-enveloped and icosahedral virion and 7.5 k-base genome containing a single open reading frame (ORF) flanked by untranslated regions (UTRs) at the 5′ and 3′ ends. The ORF encodes a single long polyprotein containing structural proteins (VP1, VP2, VP3 and VP4 encoded in P1) and non-structural proteins (2A, 2B and 2C encoded in P2 as well as 3A, 3B, 3C and 3D encoded in P3) [1, 2].

Genus Enterovirus is divided into 12 species defined as Enterovirus AH and J (EV-A, B, C, D, E, F, G, H and J) and Rhinovirus AC (RV-A, B and C) [2]. BEVs belong to EV-E and EV-F (formerly known as BEV-A and BEV-B, respectively) and can be distinguished from other EVs by the unique secondary structure of their RNA genome: a 5′-cloverleaf and internal ribosome entry site (IRES) linked by additional nucleotide sequences at the 5′UTR [35]. Since the isolation of BEVs from cattle in the late 1950s [68], studies worldwide have detected BEVs not only in cattle but also in other animal species including possums, bottlenose dolphins, camels and alpacas [812]. Although BEVs have been classified based on virus antigenicity determined by cross neutralization testing [1316], the genotype based on the capsid protein (particularly in VP1) amino acid sequences are also used to classify BEVs [4, 1012, 17, 18]. BEVs are classified into 4 sero-/genotypes and 6 sero-/genotypes in EV-E (E1, E2, E3 and E4) and EV-F (F1, F2, F3, F4, F5 and F6), respectively.

Although most EVs cause only mild symptoms, including hand-foot-and-mouth disease, herpangina, pleurodynia and rashes [19, 20], some members belonging to the genus Enterovirus can cause severe diseases. The most well known pathogen is poliovirus affecting humans. Poliovirus and some of other EVs, including coxsackie virus and echovirus, can invade the central nervous system causing neurological diseases, including aseptic meningitis, encephalitis and ataxia [21, 22]. In other animals, although porcine teschovirus, formerly classified as porcine enterovirus, can cause a neurological disorder known as Teschen/Talfan disease [23], the pathogenicity of EVs infecting animals are still unclear. In case of cattle, foot-and-mouth disease virus belonging to the genus Aphthovirus of the family Picornaviridae can cause vesicular diseases leading to a serious economic impact for farmers [24]; the pathogenicity of viruses belonging to the genus Enterorovirus is still unclear. Several reports have claimed that BEVs can cause diarrhea, respiratory diseases, reproductive diseases and infertility in cattle [2527]; however, BEVs have also been widely detected in asymptomatic cattle and their environment, and experimental infection trials of BEV have failed to produce clinical signs [2830]. Therefore, whether BEV infection is clinically important remains unclear.

It is widely known that most viruses belonging to genus Enterovirus utilize “canyon” as their binding site to cells surface receptors, which is formed by outer capsid proteins including VP1, VP2 and VP3 [31]. Several studies of other enteroviruses revealed that sequences of the VP1 coding region are responsible for the phenotype of viruses; some amino acid substitutions in this region altered the pathogenicity and cell tropism of the viruses [3234]. Although the cell surface receptor to BEV has not been identified, it is likely that the capsid proteins, including VP1, may be responsible for the phenotype of BEVs, as their capsid proteins also form a “canyon” on the outer side of the virion, and a strain isolated from cattle with severe symptoms contained specific amino acid substitutions in the capsid regions [27, 35]. To elucidate the determinants of BEV virulence in hosts, genomic information of BEVs must be determined.

Recently, deep sequencing techniques using high-throughput sequencers have been used to evaluate virome including novel viruses in clinical samples without viral isolation to determine total genomic information within samples [36, 37]. We previously identified novel viruses infecting the intestinal tracts of livestock using high-throughput sequencers to study enterovirus, picornavirus and astrovirus in the feces of goat, swine and cattle, respectively [3841].

Previously, we reported the detection of novel kobu-like virus in Japanese Black cattle, using feces of calf, by metagenomics analysis. In the present study, we identified a novel BEV in feces collected for that survey [42]. To characterize the genomic features of this virus, complete genome sequences were determined and phylogenetic trees were constructed. In addition, secondary RNA structures in the 5′UTR and pairwise identity were analyzed.

Methods

Fecal sample and virus isolation

Previously, we reported the detection of a novel kobu-like virus in Japanese black cattle by deep sequencing method [42]. During the metagenomics surveillance, nucleotide sequences with high similarity to BEVs were identified in feces collected from a calf with diarrhea. This feces was collected from a 1-month-old calf with diarrhea in Kagoshima prefecture (Kagoshima sample) in 2014. No other clinical sign was observed except diarrhea. Feces was collected directly from the rectum on the onset day. One gram feces was diluted with 9 mL PBS (−) to prepare a 10% fecal suspension and centrifuged at 10,000 × g for 10 min. The supernatant was collected and stored at −80 °C before RNA extraction and virus isolation.

The supernatant of the Kagoshima sample was subjected to virus isolation. The fecal supernatant was filtered through a 0.45-μm pore size membrane and treated with 10 μg/mL acetylated trypsin (Sigma-Aldrich, St. Louis, MO, USA) for 60 min at room temperature before virus isolation. Treated samples were inoculated into Mardin-Darby bovine kidney cells. Blind passage was subsequently conducted three times. Minimum Essential Medium was used as negative control (Sigma-Aldrich).

Isolated BEV strains

In this study, three BEVs isolated in Japan, BEV IS1/Bos taurus/JPN/1990 (BEV-IS1) and IS2/Bos taurus/JPN/1990 (BEV-IS2), were additionally sequenced and analyzed. These viruses were isolated from a fecal sample collected from one cow at the same time in 1990 in the Ishikawa prefecture (The clinical features of cattle infected with BEV-IS1 and IS2 have not been recorded). In addition, BEV Ho12/Bos taurus/JPN/2009 (BEV-Ho12) was isolated from diarrheic feces collected in Hokkaido in 2009 by as described above [39].

RNA extraction, cDNA library construction and whole genome sequencing

Total RNA was extracted from 0.25-mL supernatants of isolated viruses and 10% fecal samples using TRIzol LS Reagent (Life Technologies, Carlsbad, CA, USA). RNA samples were normalized to 10–100 ng of RNA per reaction, using a Qubit_2.0 Fluorometer (Invitrogen, Carlsbad, CA, USA). The cDNA library of sample RNA was constructed using the NEBNext Ultra RNA Library Prep Kit for Illumina version 2.0 (New England Biolabs, Ipswich, MA, USA) as described previously [40] and sequenced using MiSeq (Illumina, San Diego, CA, USA) with the MiSeq reagent kit V2 (300 cycles) (Illumina). Briefly, all reads were generated as 151 paired end reads. Each sample was multiplexed with other 23 samples prepared from diarrheal feces of other calves (data not shown). 5′-Full RACE Core Set (TaKaRa Bio, Shiga, Japan) and 3′Full RACE Core Set (TaKaRa Bio) were used to complement virus sequences of the 5′ end and 3′ end, respectively.

Analysis of genome sequences

All nucleotide sequences determined by Miseq (referred to as “reads”) were converted to FASTAQ format on MiSeq reporter V2.3 and subsequently analyzed using CLC Genomics Workbench 6.0 (CLC bio, Cambridge, MA, USA). Briefly, the ends of all reads were trimmed to remove adaptor sequences located at both ends of each read. Trimmed reads were assembled into contigs using a de novo assembly algorithm. Contigs generated by de novo assembly algorithm were analyzed using BlastN.

Hypothetical polyprotein cleavage sites of the viruses were predicted by multiple alignments with other BEVs and confirmed by the NetPicoRNA [43]. Nucleotide (nt) sequences or amino acid (aa) sequences were aligned using ClustalW. Phylogenetic trees were constructed by maximum likelihood (ML) methods on MEGA5.2.2 [44]. The mtREV24 + G + F model (5′UTR), rtREV + F model (3′UTR), rtREV + G + F model (P1), rtREV + G + I (P2 and P3), and WAG + G + I (VP1) were employed as evolutionary models for ML method. Pairwise identity was analyzed on CLC Genomics Workbench and the secondary RNA structure of the 5′UTR was predicted by Mfold [45].

VP1 genome sequencing

RT-PCR was performed by using PrimeScript One Step RT-PCR Kit Ver.2 (TaKaRa Bio) to confirm the sequences of the contigs obtained from the Kagoshima sample. Three primer sets were designed based on the contig sequences of this sample. Primer sequences are given in Additional file 1: Table S1. PCR products were sequenced using a 3130xl Genetic analyzer (Applied Biosystems, Foster City, CA, USA).

Detection of other pathogens causing diarrhea

To confirm the presence of other pathogens in the Kagoshima sample, detection of agents causing diarrhea using our real-time PCR system, referred to as “Dembo-PCR,” was performed [46]. This system can identify 19 species of pathogens, including virus, bacteria and protozoa. Briefly, viral DNA and RNA were extracted by high pure viral nucleic acid extraction kit (Roche Diagnostics GmbH, Mannheim, Germany) and bacteria and protozoa DNA were extracted by QIAamp Fast DNA stool mini kit (QIAGEN, Hilden, Germany). Nucleic acids extracted by each kit were subjected to Dembo-PCR, according to a previous report [46].

Results

Virus isolation and determination of viral genome sequences

Although virus isolation using supernatants of the Kagoshima feces was repeated three times, no cytopathic effect could be detected. Therefore, RNA extracted from the Kagoshima sample collected in 2014 and virus stocks of BEV-IS1, BEV-IS2 and BEV-Ho12 were subjected to deep sequencing. The Kagoshima sample was sequenced twice, and all reads obtained from the two runs were used to generate contigs (the first and second deep sequencing yielded 1,304,032 and 929,976 reads, respectively). The results of BlastN analysis revealed that bovine enterovirus F, group A rotavirus (RVA), bovine kobu-like virus and bovine picornavirus were identified with E value = 0. However, RVA was not detected in feces by Dembo-PCR.

The total BEV read counts (percentages indicate BEV reads per total reads of the first and second deep sequencing) of the Kagoshima sample were 1202 reads (0.05%), and an approximately 7400 nt contig was obtained from the integrated result with a 24.24 average sequence read depth (maximum read depth was 46). The complete genome was determined using 5′ and 3′ end RACE methods. Because amino acid sequences of VP1 were not similar to those of other enteroviruses by homology analysis as described below, the VP1 genome sequence was confirmed by directly sequencing the PCR product. As a result, sequences obtained from direct sequencing agreed with the results of deep sequencing. The genome length of BEV from the Kagoshima sample was 7414 nt, excluding the poly (A) tail. We named this BEV as BEV AN12/Bos taurus/JPN/2014 (BEV-AN12).

Viral genomes of isolated viruses including complete ORFs were also determined. The genome lengths of BEV-IS1, BEV-IS2 and BEV-Ho12 were 7413 nt (P1: 2517 nt, P2: 1737 nt, and P3: 2271 nt), 7394 nt (P1: 2496 nt, P2: 1734 nt, and P3: 2271 nt), and 7350 nt (P1: 2496 nt, P2: 1734 nt, and P3: 2271 nt), respectively. The sequences of BEV-AN12, BEV-Ho12, BEV-IS1 and BEV-IS2 were deposited in the DDBJ/EMBL/GenBank database under the accession numbers LC038188, LC150008, LC150009 and LC150010, respectively.

Pairwise identity and genome analysis

Table 1 shows the pairwise aa (polyprotein, 2C + 3CD, P1-P3, VP1-VP4 and 3D) or nt (5′UTR and 3′UTR) identity of BEV-AN12 to representative strains of each species belonging to BEVs and other Japanese BEVs. Deduced aa sequences encoding polyprotein, 2C + 3CD, P1, P2, P3, 3D and four capsid proteins (encoding VP4, VP2, VP3 and VP1) were compared to each EV-E and F. BEV-AN12 possessed showed identity to EV-Fs in polyprotein, 2C + 3CD, P2, P3 and 3D than to those of EV-Es. However, low aa identity (aa identity <70%) was observed in P1 to EV-Es and EV-Fs. Particularly, the VP1 region of BEV-AN12 encoded in P1 showed a significantly low aa identity to other BEVs (54.7% ≤ aa identity ≤ 58.6%). As a result of multiple alignment analysis, several motifs conserved among the genomes of genus Enterovirus were detected in the genome of BEV-AN12. In particular, the [PS] ALXAAXETG motif in VP1, GXCG motif in 2A, GXXGXGKS motif for NTP-binding in 2C, GXCG motif forming part of the catalytic active site in 3C, and KDE [LI] R in 3D were identified [4750]. However, the putative cleavage site at the junction of VP3/VP1 was a glutamine/serine for BEV-AN12. In addition, a 6-aa insertion in the 2A region was identified at position 835–840 aa (PLRTTG) in the BEV-AN12 genome. Multiple alignment using aa sequences encoding polyprotein is supplemented as Additional file 2: Table S2.

Table 1 Pairwise nucleotide (5′UTR and 3′UTR) and amino acid (polyprotein, 2C + 3CD, P1-P3, 3D and VP1-4) identity (%) of BEV-AN12 to Japanese BEVs, EV-Es and EV-Fs

Secondary RNA structure of 5′UTR

The putative secondary 5′UTR RNA structure of BEV-AN12 is shown in Fig. 1. Zell et al. reported that the 5′UTR of BEVs form additional cloverleaf structures and small stem loops between the 5′-cloverleaf structure and IRES, which clearly distinguish BEVs from other EVs [3]. Similarly, our analysis revealed that all Japanese BEVs had BEV-specific structures (domains I* and I**). Domains II, III, IV, V and VI, which are the main domains of type 1 IRES directing cap-independent translation [51], were also observed in all Japanese BEVs.

Fig. 1
figure 1

Secondary RNA structure of 5′UTR of BEV-AN12. Putative secondary RNA structure of 5′UTR of BEV-AN12 was predicted by Mfold. Domains I, I*, I**, II, III, IV, V and VI domain were predicted. Domains I* and I** conserved among BEVs are shown with a bold line. Yn-Xm-AUG motif conserved in domain VI is indicated by a bold line. Kozak consensus sequences with start codon are also indicated by a bold line

Phylogenetic analysis

Phylogenetic trees based on the nt sequences of 5′UTR and 3′UTR and aa sequences of P1, P2, P3 and VP1 are shown in Fig. 2 and Fig. 3, respectively. Phylogenetic analysis showed that all Japanese strains of BEV including BEV-AN12 formed a cluster with other BEVs in all six trees. However, BEV-AN12 was completely separated from other BEVs in the P1 and VP1 trees.

Fig. 2
figure 2

Phylogenetic trees using nucleic acid sequences of 5′UTR and 3′UTR. Phylogenetic trees were constructed using nucleic acid sequences of EV-A to EV-J and Japanese BEVs based on the maximum likelihood method in MEGA5.22 with bootstrap values calculated for 1000 replicates. Scale bar indicates nucleotide substitutions per site. BEVs in Japanese were indicated as ■. Simian sapelovirus (AY064708) was used as outgroup

Fig. 3
figure 3

Phylogenetic trees using amino acid sequences of P1, P2 and P3 and VP1 proteins. Phylogenetic trees were constructed using amino acid sequences of EV-A to EV-J and Japanese BEVs based on the maximum likelihood method in MEGA5.22 with bootstrap values calculated for 1000 replicates. Scale bar indicates amino acid substitution per site. BEVs in Japanese were indicated as ■. Simian sapelovirus (AY064708) was used as outgroup

Detection of agents causing diarrhea by Dembo-PCR

Dembo-PCR was performed to identify agents causing diarrhea in calf. According to the results, only BEV was detected by this test. Genome of other pathogens was not identified in the fecal sample of Kagoshima.

Discussion

According to the species demarcation criteria for the genus Enterovirus defined by the International Committee on Taxonomy of Viruses, members of a species in the genus Enterovirus should share high aa identity (aa >70% in the polyprotein, aa >60% in P1 and >80% aa identity in 2C + 3CD) and compatibility in processing, replication and encapsidation [2]. In addition, EV-E and F can be distinguished from other EVs because of their unique secondary RNA structures in the 5′UTR region (domains I* and I**) [4]. Our genome analysis revealed that BEV-AN12 shared aa sequences and protease cleavage site positions with EV-Fs. In addition, BEV-AN12 contained domains I* and II* in the 5′UTR similarly to other BEVs. Therefore, BEV-AN12 is closely related to EV-Fs. However, pairwise identity analysis revealed that aa sequences in the VP1 region of the BEV-AN12 genome had significantly low identities to other BEVs strains (VP1 aa identity ≤58.6%). Furthermore, BEV-AN12 did not cluster with any other EV-E and EV-F in the VP1 phylogenetic tree, although its P2 and P3 regions were closely related to EV-F. The percentage of aa identity of VP1 is commonly utilized for species and sero-/genotype definition (range from 50 to 55% for heterologous species, 70 to 85 % for heterologous sero-/genotypes/homologous species, and greater than 90 % for homologous sero-/genotypes) [4]. According to the classification definition, our results indicate that BEV-AN12 is taxonomically distant from previously reported BEVs. Therefore, we named this strain as “Enterovirus K”, which is a novel species within the genus Enterovirus. Other Japanese BEVs were classified as typical BEVs (BEV-IS1: EV-E2, BEV-IS2 and BEV-Ho12: EV-F4).

Our recombination analysis could not reveal the source of mutation (data not shown), although several reports suggested that recombinant viruses belonging to the genus Enterovirus were generated by intra/interspecies transmission [52, 53]. Point mutations in the viral genome are common among picornaviruses because their polymerase lacks the proofreading ability and fidelity of amplification [5456]. In addition, VP1 is a capsid protein, which likely influences host immunity in infected animals [35]. Therefore, the accumulation of mutations in the viral genome and subsequent selection by immunity in infected hosts may result in the generation of novel species.

Although the complete ORF and complete or partial UTRs sequences of four Japanese BEVs were determined, the virus could not be isolated from one diarrheal feces of a calf (BEV-AN12). BEV-AN12 has mutations in VP1 and 2A, which are involved in the formation of the capsid protein-host receptor binding site and cell proliferation, respectively [35, 49]. Although critical motifs for their function including [PS] ALXAXETG and GXCG were identified in the BEV-AN12 genome, a short insertion (6 aa) in the 2A protein region and non-synonymous substitution at the junction of VP3/VP1 were observed in the BEV-AN12 genome. Because these mutations may show alter receptor binding or virus replication, further crystal structure analysis of virions should be conducted.

The VP1 proteins in viruses belonging to the genus Enterovirus are widely known to components that form the receptor binding site (this site is referred to as the “canyon”) together with VP2 and VP3 [31]. Reverse genetic analysis of other enteroviruses revealed that amino acid substitution in the VP1 region was responsible for the virus phenotype, such as pathogenicity and cell tropism [3234]. Mutations in capsid protein genes may influence the structure of the “canyon” and receptor-binding capacity. BEVs also form a “canyon” on the outer side of the virion, although the cell surface receptor for BEVs is unknown. Therefore, BEVs may also have specific determinants for their phenotypes based on the aa sequences in the capsid protein encoding region. We also tried to investigate the prevalence of BEV-AN12, using VP1 specific primers, (Additional file 1: Table S1) in 38 diarrheal and 28 non-diarrheal feces samples collected from calves in Kagoshima prefecture during 2014–2015; feces from only one calf was positive, as revealed in the results. Therefore, we could not analyze the relationship between BEV-AN12 and its pathogenicity. According to the results of deep sequencing, we identified bovine picornavirus- and bovine kobu-like virus in the Kagoshima sample. There are reports suggesting that bovine picornavirus and bovine kobuvirus are associated with diarrhea [39, 57]. However, the pathogenicity of these viruses is still unknown. There is a possibility that all viruses can cause diarrhea. To clarify the determinants of the pathogenicity of BEVs, experimental infection based on reverse genetic analysis is necessary.

Conclusions

The present study identified novel BEV possessing highly divergent aa sequences in the VP1 coding region in Japan. We name this strain as “Enterovirus K”, which is a novel species within the genus Enterovirus. To exclude the pathogenicity of BEVs, further genomic information must be accumulated.

Abbreviations

aa:

Amino acid

BEV:

Bovine enterovirus

EV:

Enterovirus

IRES:

Internal ribosome entry site

nt:

Nucleotide

ORF:

Open reading frame

UTR:

Untranslated region

References

  1. Hyypia T, Hovi T, Knowles NJ, Stanway G. Classification of enteroviruses based on molecular and biological properties. J Gen Virol. 1997;78:1–11.

    Article  CAS  PubMed  Google Scholar 

  2. King AM, Adams M, Carsten E, Lefkowitz E. Virus Taxonomy. Ninth Report of the International Committee for the Taxonomy of Viruses. San Diego: Academic; 2012.

    Google Scholar 

  3. Zell R, Sidigi K, Henke A, Schmidt-Brauns J, Hoey E, Martin S, et al. Functional features of the bovine enterovirus 5′-non-translated region. J Gen Virol. 1999;80:2299–309.

    Article  CAS  PubMed  Google Scholar 

  4. Zell R, Krumbholz A, Dauber M, Hoey E, Wutzler P. Molecular-based reclassification of the bovine enteroviruses. J Gen Virol. 2006;87:375–85.

    Article  CAS  PubMed  Google Scholar 

  5. Zell R, Stelzner A. Application of genome sequence information to the classification of bovine enteroviruses: the importance of 5′- and 3′-nontranslated regions. Virus Res. 1997;51:213–29.

    Article  CAS  PubMed  Google Scholar 

  6. Kunin CM, Minuse E. New bovine viral agents isolated in Michigan. Public Health Repts. 1957;72:251.

    Google Scholar 

  7. Kunin CM, Minuse E. The Isolation in Tissue Culture, Chick Embryo and Suckling Mice of Filtrable Agents from Healthy Dairy Cattle. J Immunol. 1958;80:1–11.

    CAS  PubMed  Google Scholar 

  8. Moll T, Finlayson AV. Isolation of cytopathogenic viral agent from feces of cattle. Science. 1957;126:401–2.

    Article  CAS  PubMed  Google Scholar 

  9. Nollens HH, Rivera R, Palacios G, Wellehan JFX, Saliki JT, Caseltine SL, et al. New recognition of Enterovirus infections in bottlenose dolphins (Tursiops truncatus). Vet Microbiol. 2009;139:170–5.

    Article  PubMed  PubMed Central  Google Scholar 

  10. McClenahan SD, Scherba G, Borst L, Fredrickson RL, Krause PR, Uhlenhaut C. Discovery of a bovine enterovirus in alpaca. PLoS One. 2013;8:e68777.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zheng T. Characterisation of two enteroviruses isolated from Australian brushtail possums (Trichosurus vulpecula) in New Zealand. Arch Virol. 2007;152:191–8.

    Article  CAS  PubMed  Google Scholar 

  12. Woo PCY, Lau SKP, Li T, Jose S, Yip CCY, Huang Y, et al. A novel dromedary camel enterovirus in the family Picornaviridae from dromedaries in the Middle East. J Gen Virol. 2015;96:1723–31.

    Article  CAS  PubMed  Google Scholar 

  13. Knowles NJ, Barnett ITR. A serological classification of bovine enteroviruses. Arch Virol. 1985;83:141–55.

    Article  CAS  PubMed  Google Scholar 

  14. Dunne HW, Huang CM, Lin WJ. Bovine enteroviruses in the calf: an attempt at serologic, biologic, and pathologic classification. J. Am. Vet. Med. Assoc. 1974;164(3):290–4.

  15. Huck RA, Cartwright SF. Isolation and Classification of Viruses from Cattle During Outbreaks of Mucosal or Respiratory Disease and from Herds with Reproductive Disorders. J Comp Pathol Ther. 1964;74:346–65.

    Article  CAS  Google Scholar 

  16. Barya MA, Moll T, Mattson DE. Antigenic analysis of bovine enteroviruses through studies of the kinetics of neutralization. Am J Vet Res. 1967;28:1283–94.

    CAS  PubMed  Google Scholar 

  17. Shaukat S, Angez M, Alam MM, Sharif S, Khurshid A, Malik F, et al. Molecular identification and characterization of a new type of bovine enterovirus. Appl Environ Microbiol. 2012;78:4497–500.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Zhang H, Liu H, Bao J, Guo Y, Peng T, Zhou P, et al. Characterization of an Enterovirus species E isolated from naturally infected bovine in China. Virus Res. 2014;191:101–7.

    Article  CAS  PubMed  Google Scholar 

  19. Ang LW, Koh BK, Chan KP, Chua LT, James L, Goh KT. Epidemiology and control of hand, foot and mouth disease in Singapore, 2001–2007. Ann Acad Med Singapore. 2009;38:106–12.

    PubMed  Google Scholar 

  20. Zou X, Zhang X, Wang B, Qiu Y. Etiologic and epidemiologic analysis of hand, foot, and mouth disease in Guangzhou city: a review of 4,753 cases. Braz J Infect Dis Off Publ Braz Soc Infect Dis. 2012;16:457–65.

    Google Scholar 

  21. Gear JH. Nonpolio causes of polio-like paralytic syndromes. Rev Infect Dis. 1984;6 Suppl 2:S379–384.

    Article  PubMed  Google Scholar 

  22. Huang H-I, Shih S-R. Neurotropic Enterovirus Infections in the Central Nervous System. Viruses. 2015;7:6051–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zell R, Dauber M, Krumbholz A, Henke A, Birch-Hirschfeld E, Stelzner A, et al. Porcine Teschoviruses Comprise at Least Eleven Distinct Serotypes: Molecular and Evolutionary Aspects. J Virol. 2001;75:1620–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Knight-Jones TJD, Rushton J. The economic impacts of foot and mouth disease - what are they, how big are they and where do they occur? Prev Vet Med. 2013;112:161–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Dunne HW, Ajinkya SM, Bubash GR, Griel LC. Parainfluenza-3 and bovine enteroviruses as possible important causative factors in bovine abortion. Am J Vet Res. 1973;34:1121–6.

    CAS  PubMed  Google Scholar 

  26. Weldon SL, Blue JL, Wooley RE, Lukert PD. Isolation of picornavirus from feces and semen from an infertile bull. J Am Vet Med Assoc. 1979;174:168–9.

    CAS  PubMed  Google Scholar 

  27. Zhu L, Xing Z, Gai X, Li S, San Z, Wang X. Identification of a novel enterovirus E isolates HY12 from cattle with severe respiratory and enteric diseases. PLoS One. 2014;9:e97730.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Ley V, Higgins J, Fayer R. Bovine Enteroviruses as Indicators of Fecal Contamination. Appl Environ Microbiol. 2002;68:3455–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jiménez-Clavero MA, Escribano-Romero E, Mansilla C, Gómez N, Córdoba L, Roblas N, et al. Survey of Bovine Enterovirus in Biological and Environmental Samples by a Highly Sensitive Real-Time Reverse Transcription-PCR. Appl Environ Microbiol. 2005;71:3536–43.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Blas-Machado U, Saliki JT, Sánchez S, Brown CC, Zhang J, Keys D, et al. Pathogenesis of a Bovine Enterovirus-1 Isolate in Experimentally Infected Calves. Vet Pathol Online. 2011;48:1075–84.

    Article  CAS  Google Scholar 

  31. Hogle JM, Chow M, Filman DJ. Three-dimensional structure of poliovirus at 2.9 A resolution. Science. 1985;229:1358–65.

    Article  CAS  PubMed  Google Scholar 

  32. Kataoka C, Suzuki T, Kotani O, Iwata-Yoshikawa N, Nagata N, Ami Y, et al. The Role of VP1 Amino Acid Residue 145 of Enterovirus 71 in Viral Fitness and Pathogenesis in a Cynomolgus Monkey Model. PLoS Pathog. 2015;11:e1005033.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Caine EA, Moncla LH, Ronderos MD, Friedrich TC, Osorio JE. A Single Mutation in the VP1 of Enterovirus 71 is Responsible for Increased Virulence and Neurotropism in Adult Interferon Deficient Mice. J Virol. 2016;90(19):8592–604.

  34. Lin J-Y, Shih S-R. Cell and tissue tropism of enterovirus 71 and other enteroviruses infections. J Biomed Sci. 2014;21:18.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Goens SD, Botero S, Zemla A, Zhou CE, Perdue ML. Bovine enterovirus 2: complete genomic sequence and molecular modelling of a reference strain and a wild-type isolate from endemically infected US cattle. J Gen Virol. 2004;85:3195–203.

    Article  CAS  PubMed  Google Scholar 

  36. Schlottau K, Schulze C, Bilk S, Hanke D, Höper D, Beer M, et al. Detection of a Novel Bovine Astrovirus in a Cow with Encephalitis. Transbound Emerg Dis. 2016;63:253–9.

    Article  CAS  PubMed  Google Scholar 

  37. Reuter G, Boros Á, Pál J, Kapusinszky B, Delwart E, Pankovics P. Detection and genome analysis of a novel (dima) rhabdovirus (Riverside virus) from Ochlerotatus sp. mosquitoes in Central Europe. Infect Genet Evol J Mol Epidemiol Evol Genet Infect Dis. 2016;39:336–41.

    Article  CAS  Google Scholar 

  38. Omatsu T, Tsuchiaka S, Hirata T, Shiroma Y, Okazaki S, Katayama Y, et al. Detection of enterovirus genome sequence from diarrheal feces of goat. Virus Genes. 2014;48:550–2.

    Article  CAS  PubMed  Google Scholar 

  39. Nagai M, Omatsu T, Aoki H, Kaku Y, Belsham GJ, Haga K, et al. Identification and complete genome analysis of a novel bovine picornavirus in Japan. Virus Res. 2015;210:205–12.

    Article  CAS  PubMed  Google Scholar 

  40. Nagai M, Omatsu T, Aoki H, Otomaru K, Uto T, Koizumi M, et al. Full genome analysis of bovine astrovirus from fecal samples of cattle in Japan: identification of possible interspecies transmission of bovine astrovirus. Arch Virol. 2015;160:2491–501.

    Article  CAS  PubMed  Google Scholar 

  41. Naoi Y, Kishimoto M, Masuda T, Ito M, Tsuchiaka S, Sano K, et al. Characterization and phylogenetic analysis of a novel picornavirus from swine feces in Japan. Arch Virol. 2016;161(6):1685–90.

  42. Otomaru K, Naoi Y, Haga K, Omatsu T, Uto T, Koizumi M, et al. Detection of novel kobu-like viruses in Japanese black cattle in Japan. J Vet Med Sci Jpn Soc Vet Sci. 2016;78(2):321–4.

  43. Blom N, Hansen J, Blaas D, Brunak S. Cleavage site analysis in picornaviral polyproteins: discovering cellular targets by neural networks. Protein Sci. 1996;5:2203–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Tamura K, Peterson D, Peterson N, Stecher G, Nei M, Kumar S. MEGA5: molecular evolutionary genetics analysis using maximum likelihood, evolutionary distance, and maximum parsimony methods. Mol Biol Evol. 2011;28:2731–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zuker M. Mfold web server for nucleic acid folding and hybridization prediction. Nucleic Acids Res. 2003;31:3406–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Tsuchiaka S, Masuda T, Sugimura S, Kobayashi S, Komatsu N, Nagai M, et al. Development of a novel detection system for microbes from bovine diarrhea by real-time PCR. J Vet Med Sci Jpn Soc Vet Sci. 2016;78:383–9.

    Article  Google Scholar 

  47. Kamer G, Argos P. Primary structural comparison of RNA-dependent polymerases from plant, animal and bacterial viruses. Nucleic Acids Res. 1984;12:7269–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chow M, Newman JFE, Filman D, Hogle JM, Rowlands DJ, Brown F. Myristylation of picornavirus capsid protein VP4 and its structural significance. Nature. 1987;327:482–6.

    Article  CAS  PubMed  Google Scholar 

  49. Hughes PJ, Stanway G. The 2A proteins of three diverse picornaviruses are related to each other and to the H-rev107 family of proteins involved in the control of cell proliferation. J Gen Virol. 2000;81:201–7.

    Article  CAS  PubMed  Google Scholar 

  50. Gorbalenya AE, Koonin EV, Donchenko AP, Blinov VM. Two related superfamilies of putative helicases involved in replication, recombination, repair and expression of DNA and RNA genomes. Nucleic Acids Res. 1989;17:4713–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Martínez-Salas E, Francisco-Velilla R, Fernandez-Chamorro J, Lozano G, Diaz-Toledano R. Picornavirus IRES elements: RNA structure and host protein interactions. Virus Res. 2015;206:62–73.

    Article  PubMed  Google Scholar 

  52. Zhang G, Haydon DT, Knowles NJ, McCauley JW. Molecular evolution of swine vesicular disease virus. J Gen Virol. 1999;80:639–51.

    Article  CAS  PubMed  Google Scholar 

  53. Boros Á, Pankovics P, Knowles NJ, Reuter G. Natural interspecies recombinant bovine/porcine enterovirus in sheep. J Gen Virol. 2012;93:1941–51.

    Article  CAS  PubMed  Google Scholar 

  54. Ward CD, Stokes MA, Flanegan JB. Direct measurement of the poliovirus RNA polymerase error frequency in vitro. J Virol. 1988;62:558–62.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Ferrer-Orta C, Arias A, Pérez-Luque R, Escarmís C, Domingo E, Verdaguer N. Sequential structures provide insights into the fidelity of RNA replication. Proc Natl Acad Sci U S A. 2007;104:9463–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Ward CD, Flanegan JB. Determination of the poliovirus RNA polymerase error frequency at eight sites in the viral genome. J Virol. 1992;66:3784–93.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Reuter G, Boros A, Pankovics P. Kobuviruses - a comprehensive review. Rev Med Virol. 2011;21:32–41.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by the Research Project for Improving Food Safety and Animal Health of the Ministry of Agriculture, Forestry and Fisheries of Japan.

Availability of data and materials

Deep sequencing results and Japanese BEVs genomic sequences have been deposited in the DNA Data Bank of Japan (DRA005341, DRA005342, DRA005343, DRA005344, LC038188, LC150008, LC150009 and LC150010). All data related to phylogenetic trees have been deposited in TreeBASE (http://purl.org/phylo/treebase/phylows/study/TB2:S20312).

Authors’ contributions

ST performed almost all in vitro experiments, analyzed and interpreted the data. In addition, ST wrote the manuscript. SS-R performed VP1 genome sequencing. KO contributed to collect the sample from Kagoshima prefecture in Japan, interpreted the data clinically and reviewed the manuscript. HA contributed to collect the sample from Hokkaido in Japan, interpreted the data including phylogenetic analysis and reviewed the manuscript. MK was a contributor to the molecular experiment and interpretation of the data including secondary RNA structure analysis of the 5′ untranslated region. YN was a contributor in revising the manuscript. TO was a major contributor in writing and revising manuscript. KS was a contributor in revising the manuscript. SO-T was a contributor in revising the manuscript. YK was a major contributor to the construction of cDNA library for deep sequencing. MO was a contributor to PCR experiment. MN coordinated and designed this study. In addition, MN was a major contributor to the interpretation of deep sequencing analysis. TM provided the funds necessary for the project, provided guidance to the study design. In addition, TM was a major contributor in writing and reviewing the manuscript, and is the corresponding author. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Consent for publication

Not applicable.

Ethics approval and consent to participate

This section is not applicable because the viral isolates from a cow were not originally obtained for the purpose of this study.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Tetsuya Mizutani.

Additional files

Additional file 1: Table S1.

Primers information for VP1 sequencing. (PDF 84 kb)

Additional file 2: Table S2.

Multiple alignments result using amino acid sequences of polyprotein. (PDF 458 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tsuchiaka, S., Rahpaya, S.S., Otomaru, K. et al. Identification of a novel bovine enterovirus possessing highly divergent amino acid sequences in capsid protein. BMC Microbiol 17, 18 (2017). https://doi.org/10.1186/s12866-016-0923-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12866-016-0923-0

Keywords