Skip to main content

Characterization of porcine sapelovirus prevalent in western Jiangxi, China

Abstract

Background

Porcine sapelovirus (PSV) infection can lead severe polioencephalomyelitis with high morbidity and mortality, which result in significant economic losses. Infection with the PSV is believed to be common yet limited information is available on the prevalence and molecular characterization of PSV in China. Therefore, the objective of this study was to characterize the prevalence and genome of PSV strains identified in the western Jiangxi province of China.

Results

A high specificity and sensitivity SYBR Green I-based RT-PCR method for PSV detection was developed. Two hundred and ninety four fecal samples were collected from December 2018 to March 2019 in 4 farms. An overall PSV-positivity rate of 11.22% (33/294) was detected with the real-time RT-PCR method, and a high infection rate and viral load of PSV were found in nursery pigs. In total, complete VP1 gene sequences of 11 PSV strains (PSV-YCs) were obtained. Homology comparisons of the VP1 gene of the 11 PSV-YCs with previously reported PSVs revealed nucleotide sequence identities ranging from 63% to 96.8%, and deduced amino acid sequence identities from 61.4% to 99.7%. Phylogenetic analyses based on the VP1 gene exhibited 2 main clades corresponding to PSV-1 and PSV-2, and all PSV-YCs prevalent in western Jiangxi belonged to the traditional genotype (PSV-1). In addition, the pairwise distances of VP1 gene sequences between PSV-YCs ranged from 0.009 to 0.198, which indicating that substantial genetic diversity among the PSVs in western Jiangxi.

Conclusions

To the authors’ knowledge, this is the first description of PSV in the Jiangxi province pig herds in China, and it is crucial to understand the epidemiology of the viruses in China. The results also provide an important theoretical foundation for diagnosis and early warning of epidemic diseases caused by PSVs prevailing in this region.

Peer Review reports

Background

Taxonomically, Porcine sapelovirus (PSV), also Sapelovirus A, together with Sapelovirus B belong to the genus Sapelovirus within the family Picornaviridae [1]. The PSV genome consists of a positive-sense, single-stranded RNA of ~7.5 kb, and comprises the typical picornavirus genome organization; 5′-UTRIRES-IV-[L/1A-1B-1C- 1D/2Apro-2B-2C/3A-3B-3C-3D]-3′-UTR. The 1A-1D genes encode four structural proteins (also named VP1-4) to compose virus capsid, and the other genes encode nonstructural proteins [2].

Domestic pigs and wild boars are the only known hosts, and PSV infection can lead severe polioencephalomyelitis with high morbidity and mortality, which result in significant economic losses [3]. However, PSV infections are always subclinical or only involve a series mild symptoms, including spinal cord damage, inappetence, diarrhea and breathless [4, 5]. The disease of PSV infection was first found in Britain in the 1960s [6]. Afterwards, wide infections of PSV in domestic and wild pigs have been reported in many other countries, including Spain, Hungary, Czech Republic, Brazil, China, and South Korea [2, 4, 7,8,9,10]. In the 10th Report of the International Committee on Taxonomy of Viruses (ICTV), PSV only comprised a single genotype. With the recent use of viral metagenomics tool, a novel member of PSV genotypes has been discovered in Hungary [11].

Despite several investigations have documented PSV infection in China, knowledge of the molecular prevalence of PSV in many areas of China is still unclear so far [4, 5, 12, 13]. In the present study, for the first time, a novel SYBR Green I-based real-time RT-PCR assay was developed to investigate the prevalence of PSV in western Jiangxi, China. Moreover, genetic and phylogenetic analyses based on VP1 gene were performed to determine their molecular and evolutionary characterization.

Results

PSV real-time PCR performance and prevalence

A linear regression relationship of the standard curves was observed with a correlation coefficient (R2) of 0.9996, a slope of -5.1538, and an intercept of 50.716 (Fig. S1a). Specificity of the reaction was confirmed by a distinct melting temperature (Tm) of 84.82 (Fig. S1b), and there was no positive signal when testing cross reactivity against common porcine viral pathogens, including CSFV, PRRSV, PRV, PEDV, PTV, and JEV (Fig. S1c). The sensitivity of real-time PCR method was determined to be 5.22×102 copies/μL (Fig. S1d), which 10 times more sensitive than the conventional RT-PCR (Fig. S1e).

PSV was detected in all 4 commercial farms, representing an overall PSV-positivity rate of 11.22% (33/294). Specifically, PSV was identified in 30.36% (17/56) of the nursery pigs, in 8.82% (12/136) of the fattening pigs, and in 7.27% (4/55) of the adult pigs (Table 1). While no positive sample was detected from suckling pigs. Thus, high PSV prevalence was found in nursery pigs, which was significantly (P < 0.01) higher than that in adult pigs, suckling and fattening pigs (Table 1). Fecal samples positive for PSV had CT values of 25.25-38.57, corresponding to the amount of PSV RNA ranged from 6.81×105 to 2.62×108 genomic copies per mL fecal supernatant. Notably, the high PSV viral loads (3.25±0.18, log10 copies/μL) were also identified in the age group of nursery pigs (Table 1).

Table 1 Summary of the history (age, number of farms) and prevalence of PSV in western Jiangxi, China

Molecular characteristics of PSV genome

Eleven complete VP1 gene sequences were obtained from the 33 PSV-positive samples. Subsequently, the obtained PSV sequences (designated as YC1-11) were submitted to the GenBank database (accession nos. MW411420-MW411430). Through alignment of the coding sequences of the 11 PSV-YC strains with those of other known PSVs, the VP1 gene of seven PSV-YC strains (YC1-7) containing 897 nucleotides and encoding a 293 amino acid (AA) protein. The other four PSV-YC strains, YC8-11, contained twelve additional nucleotides, corresponding to four additional AAs (S/T)(T/P)AE inserted between AA 283 and 284 of the VP1 as compared with all other known PSV strains exhibiting the most common genome organization (Fig. 1). Homology comparison of the VP1 genes of the 11 PSV-YC strains with those of other PSV strains from GenBank revealed nucleotide and deduced amino acid sequence identities ranging from 63% to 96.8% and from 61.4% to 99.7%, respectively. In addition, PSV-YCs had sequence identities of 79.9–99% (nucleotides) and 88.3–99.7% (AAs) with each other.

Fig. 1
figure 1

Alignment of the partial amino acid sequences of PSV strains. Alignment of the amino acid sequences near the 3′ end of the VP1 gene. Sequences in boxes show the hypervariable region in the C-terminus of VP1. The strains identified in the present study are indicated by ▲; (-), missing sequences

Phylogenetic analysis of PSV

Phylogenetic analysis was conducted based on the multiple sequence alignment of the complete VP1 gene sequences of different PSV, which including the 11 PSV-YCs characterized in the present study and 101 additional PSV sequences obtained from GenBank database (Table S1). Phylogenetic analysis of VP1 gene sequences showed that all PSVs were separated into two distinct clades, corresponding to two proposed genotypes (designated as PSV-1 and PSV-2) with high bootstrap values within the genus Sapelovirus A (Fig. 2). All the PSV-YC strains fell into PSV-1 along with most of the previously reported PSVs, and two Hungarian strains SZ1M-F/PSV/HUN/2013 and EF9-F/PSV/HUN/2016 were separated into PSV-2 (Fig. 2). Additionally, the pairwise distances of VP1 gene sequences between PSV-YCs ranged from 0.009 to 0.198, revealing the high genetic diversity among the most prevalent PSVs in pig populations of western Jiangxi, China (Table 2).

Fig. 2
figure 2

Evolutionary relationships among PSVs. Phylogenetic tree based on the nucleotide sequences of VP1 gene. The evolutionary history was inferred using the ML method with the GTR model with gamma-distributed rates and proportion of invariant sites (G+I) in PhyML 3.0. The percentage of replicates in which the associated virus clustered together in the bootstrap test (100 replicates) is shown next to the branches (only values >60 % are shown) in the tree. The scale bar indicates nucleotide substitutions per site. Simian sapelovirus (SSV)-1 2383 strain, SSV-2 VRDL strain and SSV-3 WUHARV strain were used as outgroups. The strains isolated in the present study are identified by ▲

Table 2 The estimates of evolutionary divergence over sequence pairs between PSV-YCs identified by the neighbor-joining method, which include VP1 gene sequence of 11 PSVs

Discussion

Although PSV is ubiquitous and is associated with a variety of clinical symptoms in domestic and wild pigs, PSVs infecting swine are often ignored for their subclinical manifestations and co-infections with other pathogens [9, 10]. Therefore, it is necessary to develop a convenient, sensitive and accurate detection method for PSV early warning and rapid diagnosis. The PSV SYBR Green I-based real-time RT-PCR method developed in the present study had the advantages of high specificity and sensitivity. The minimum detection limit of sensitivity test showed 522 copies/μl located at same order of magnitude with the Taqman-based real-time RT-PCR method established by Chen J et al [14]. Specially, the PSV SYBR Green I-based real-time RT-PCR method in this study has high cost-efficient compared to the TaqMan-based method, which can achieving wide application with high standards and low cost. Thus, our method can be applied for clinical monitoring and diagnosis of PSV infection.

The high PSV prevalence was found in nursery pigs compared to sporadically in adult and fattening pigs. Meanwhile, higher viral loads were also found in the age group of nursery pigs. While no positive sample was detected in suckling pigs, which possibly associated with the presence of maternal antibodies in piglets. Thus, nursery pigs become the susceptible populations of PSV infection with decreasing of maternal antibodies of suckling pigs as age increasing. PSV widely prevalent in pigs of all age groups have been reported in previous study, especially high prevalence in nursery and fattening pigs, which is somewhat different from the results in the present study [5]. Therefore, we speculated that multiple factors, including the conformation or structure of farm, the health and neutralizing antibody level of pigs, are likely responsible for the differences.

The capsids of picornaviruses are composed of four structural proteins (VP14), and outer surface structure of virion, as also immunodominant, is mainly determined by the spatial folding and mutual extension of VP1, VP2 and VP3 [15]. Generally, C-terminus of VP1–3 is located on the outer surface of the virion, while the N-terminus lying on the inner surface of the virion [16]. Thus, amino acid mutations in the C-terminus of VP1–3 were suggested to influence the antigenicity of picornaviruses. In the present study, four AAs (S/T)(T/P)AE insertion near the C-terminus of VP1 were found in four PSV-YCs, which was consistent with our previous report. Exposed antigenic sites in the C-terminus of VP1 have been proved induce neutralizing antibodies in many picornaviruses [17,18,19,20,21]. Therefore, mutations of the C-terminus of the PSV VP1 possibly are associated with the escape from host immune response. However, the function and specific biological characteristics of those mutations requires more research. Notably, evolutionary analysis revealed the PSV strains with (S/T)(T/P)AE amino acid residues insertion in the C-terminus of VP1 sharing a close genetic relationship, which indicated those of amino acid residues insertion might be used as molecular marker for a cluster of PSV epidemic strains.

PSV have long been contained only one genotype, recently, a novel PSV strain (SZ1M-F/PSV/HUN2013, MN80775) was reported in pigs of Hungary, which represent a novel genotype of PSV (designed as PSV-2 in the present study) [11] . In this study, phylogenetic analysis of all available VP1 gene sequences of PSVs showed the PSV-YCs prevalent in western Jiangxi belonged to the traditional genotype (PSV-1). Notably, PSV-YCs shared the closest genetic relationship with the strains found in Hunan Province of China, indicating that the strains prevalent in local areas or neighboring provinces likely evolved from common ancestors. Meanwhile, genetic divergence also exists within the PSVs of western Jiangxi, China, even on a same farm, which was consistent with previous report in other regions [5].

Conclusion

In conclusion, we developed a high specificity and sensitivity SYBR Green I-based RT-PCR method for PSV detection. A high infection rate and viral load of PSV were found in nursery pigs in western Jiangxi, China. In addition, PSV strains prevalent in this area showed a close evolutionary relationship with the PSVs previously reported in China. Meanwhile, our data revealed substantial genetic diversity among the PSVs in western Jiangxi, China.

Materials and methods

Samples and viral strains

During December 2018 to March 2019, 294 fecal samples were collected from healthy pigs across 4 farms in the western Jiangxi province, China. The age of the pigs sampled ranged from piglets to adult pigs (Table 1). Samples were transported to the laboratory at 4 оC and stored at -80 оC until required.

PSV (HuN1 strain, GenBank # : KX354740), pseudorabies virus (PRV, XiangA strain, GenBank # : KP710981), Japanese encephalitis virus (JEV, HNML1 strain, GenBank # : KX774636), and porcine teschovirus (PTV, HuN1 strain, GenBank # : MF170905) were stored in our laboratory at -80 оC. Classical swine fever virus (CSFV) and Porcine Reproductive and Respiratory Syndrome Virus (PRRSV) attenuated live vaccine strains were purchased from Pulike Biological Inc., China, and Porcine epidemic diarrhea virus (PEDV) attenuated live vaccine strain was bought from CAHIC Inc., China.

Sample processing, viral RNA/DNA extraction and reverse transcription

Aliquots of 0.5 g of fecal material were re-suspended in 1 mL of ice-cold phosphate-buffered saline (PBS), vortexed for 1 min, and clarified by centrifugation at 12,000 x g for 5 min. The supernatant was transferred into a 2-mL Eppendorf tube used for viral RNA extraction and the remaining was stored at -80 оC until use. For RNA viruses, 0.2 mL of the supernatant of virus strains and clinical samples was used for viral RNA extraction (Tiandz Inc., Beijing, China). The viral cDNA was synthesized using the RevertAid First Strand cDNA synthesis Kit (Thermo Scientific, Waltham, MA, USA) according to the manufacturer’s instructions. All RNA extraction procedures of clinical samples included positive (PSV-HuN1 strain) and negative control (containing only water) in each run. For DNA viruses, viral DNA was carried out using the viral DNA extraction kit (Tiandz) following the manufacturer’s instructions.

Development of a real-time PCR for PSV

To ensure the conservative of primers, the known genomic sequences of PSV were retrieved from GenBank (Table S1, up to July 2020) and analyzed with the Lasergene package (DNAStar Inc., Madison, WI, USA). After multiple alignments, a pair of primers (PSV-rF/PSV-rR), located in the conserved 5’ untranslated region, was designed to amplify a 270-bp fragment of PSV DNA (Table 3). Selected positive PCR products, amplified from cDNA of PSV-HuN1 strain, were separated by electrophoresis on a 0.8% agarose gel, and the target bands were excised and purified using the gel extraction kit (Tiandz). Then, the purified PCR products were cloned into the pMD19-T vector (Takara Bio Inc., Otsu, Japan), and the recombinant plasmids were transformed into Trans1-T1 Escherichia coli bacteria (TransGen, Beijing, China) and propagated according to the instructions of cloning manual. The plasmids were extracted using the plasmid extraction kit (Tiandz) following the manufacturer’s instructions, quantified with spectrophotometer (BioDrop, Cambridge, UK), and then sequenced (BioSune, Shanghai, China).

Table 3 Primers for PSV detection and complete VP1 gene amplification

The plasmids were used as standards for the real-time PCR assay. Each reaction consisted of a total volume of 20 μL, containing 10 μL of the SYBR Green Master Mix (Thermo Scientific, Waltham, MA, USA), 1 μL of the standard plasmids, 0.5 μL of each of the two primers, and 8 μL of distilled water. Amplification and quantification reactions were performed using the ABI StepOnePlusTM Real-Time PCR Systems (Applied Biosystems, Foster City, CA, USA) under universal conditions: 2 min at 50 °C, 2 min at 95 °C, 40 cycles of 15 s at 95 °C and 1 min at 60 °C. A melt curve was performed to verify the specificity of the amplified products under universal conditions: a denaturation step at 95 °C for 15 s, decreased to 60 °C for 1 min, and followed by temperature increase to 95 °C at a rate of 0.3 °C/s. A sample was considered negative if no CT was detected in 40 amplification cycles. The initial plasmid standard with 10-fold serial dilution (5.22×102-5.22×108 copies/μL) was generating the standard curve. Each dilution was run in triplicate.

The sensitivity of the real-time PCR was determined by testing 10-fold serial dilutions of the plasmid standards (5.22×101-5.22×108 copies/μL). Meanwhile, each diluted plasmid standard was used as templates for conventional PCR detection. PCR products were analyzed by agarose gel electrophoresis, and observed under ultraviolet light to determine sensitivity. The specificity of the primers was performed by testing samples positive for other common porcine viral pathogens, including CSFV, PRRSV, PRV, PEDV, PTV, and JEV.

VP1 Gene sequencing

As the sequences of the N-terminal flanking the VP1 gene are hypervariable among PSVs, three different sets of forward primers were designed based on the grouping results of the phylogenetic analysis of these regions. Thus, three different nested-PCR methods, shared the same reverse primers, were used to amplifying the complete VP1 gene sequence of all PSV strains (Table 3). Each method comprised an external and an internal primer pair. PCR amplification of cDNA was carried out using Pfu DNA polymerase (Tiandz) under the following conditions: an initial denaturation step at 94 °C for 4 min; 35 cycles of 94 °C for 30 s, 51 °C for 30 s and 68 °C for 3 min; and a final extension at 68 °C for 10 min. A nested PCR reaction was conducted by setting up a second reaction using 1 μL of the PCR products from the first PCR reaction as template. The PCR products were analyzed by agarose gel electrophoresis and then sequenced (BioSune, Shanghai, China).

Phylogenetic analysis

After the removal of the flanking non-VP1 ends, multiple sequence alignment of the complete VP1 gene sequence was carried out using the MUSCLE method within the MEGA 6.06 software [22]. Phylogenetic analysis was carried out by PhyML 3.0 using maximum-likelihood (ML) methods [23]. The best-fit model of nucleotides for the dataset was determined using jModelTest 2.1.10 [24]. The general time Reversible (GTR) substitution model with the proportion of invariant sites and gamma-distributed rate heterogeneity (GTR+G+I) was used to construct the ML tree with a non-parametric bootstrap analysis with 100 replicates to determine the branch support. To characterize the genetic divergence of PSV, the distances were calculated by the uncorrected p-distance method using MEGA 6.06 software.

Statistical analysis

Differences in PSV prevalence rates among age group were analyzed by chi-squared tests using SPSS software version 19.0 (IBM Inc., Chicago, IL, USA). The results were considered significant at P < 0.05.

Availability of data and materials

The coding sequences of the VP1 gene were submitted to the GenBank database. The virus was named as YC1-11. The accession numbers of the VP1 gene are MW411420-MW411430, respectively

Abbreviations

RT-PCR:

Reverse transcription polymerase chain reaction

PSV:

Porcine sapelovirus

PRV:

Pseudorabies virus

JEV:

Japanese encephalitis virus

PTV:

Porcine teschovirus

CSFV:

Classical swine fever virus

PRRSV:

Porcine Reproductive and Respiratory Syndrome Virus

PEDV:

Porcine epidemic diarrhea virus

ICTV:

International Committee on Taxonomy of Viruses

PBS:

Phosphate-buffered saline

CT:

Threshold cycle

ML:

Maximum-likelihood

GTR:

General time Reversible

Tm:

Melting temperature

AA:

Amino acid

References

  1. Zell R, Delwart E, Gorbalenya AE, Hovi T, King A, Knowles NJ, et al. ICTV Virus Taxonomy Profile: Picornaviridae. J Gen Virol. 2017;98(10):2421–2.

    Article  CAS  Google Scholar 

  2. Son K, Kim D, Kwon J, Choi J, Kang M, Belsham GJ, et al. Full-length genomic analysis of Korean porcine sapelovirus strains. PLoS One. 2014;9(9):e107860.

    Article  Google Scholar 

  3. Arruda P, Arruda BL, Schwartz KJ, Vannucci F, Resende T, Rovira A, et al. Detection of a novel sapelovirus in central nervous tissue of pigs with polioencephalomyelitis in the USA. Transbound Emerg Dis. 2017;64(2):311–5.

    Article  CAS  Google Scholar 

  4. Lan D, Ji W, Yang S, Cui L, Yang Z, Yuan C, et al. Isolation and characterization of the first Chinese porcine sapelovirus strain. Arch Virol. 2011;156(9):1567–74.

    Article  CAS  Google Scholar 

  5. Yang T, Yu X, Yan M, Luo B, Li R, Qu T, et al. Molecular characterization of Porcine sapelovirus in Hunan, China. J Gen Virol. 2017;98(11):2738–47.

    Article  CAS  Google Scholar 

  6. Lamont PH, Betts AO. Studies on enteroviruses of the pig IV. The isolation in tissue culture of a possible enteric cytopathogenic swine orphan (ECSO) virus (V13) from the faeces of a pig. Res Vet Sci. 1960;1:152–9.

    Article  Google Scholar 

  7. Abe M, Ito N, Sakai K, Kaku Y, Oba M, Nishimura M, et al. A novel sapelovirus-like virus isolation from wild boar. Virus Genes. 2011;43(2):243–8.

    Article  CAS  Google Scholar 

  8. Cano-Gómez C, García-Casado MA, Soriguer R, Palero F, Jiménez-Clavero MA. Teschoviruses and sapeloviruses in faecal samples from wild boar in Spain. Vet Microbiol. 2013;165(1):115–22.

    Article  Google Scholar 

  9. Donin DG, de Leme RA, Alfieri AF, Alberton GC, Alfieri AA. Molecular survey of porcine teschovirus, porcine sapelovirus, and enterovirus G in captive wild boars (Sus scrofa scrofa) of Paraná state, Brazil. Pesqui Vet Brasil. 2015;35(5):403–8.

    Article  Google Scholar 

  10. Prodělalová J. The survey of porcine teschoviruses, sapeloviruses and enteroviruses B infecting domestic pigs and wild boars in the Czech Republic between 2005 and 2011. Infect Genet Evol. 2012;12(7):1447–51.

    Article  Google Scholar 

  11. Boros Á, László Z, Pankovics P, Marosi A, Albert M, Cságola A, et al. High prevalence, genetic diversity and a potentially novel genotype of Sapelovirus A (Picornaviridae) in enteric and respiratory samples in Hungarian swine farms. J Gen Virol. 2020;101(6):609–21.

    Article  CAS  Google Scholar 

  12. Li Y, Du L, Jin T, Cheng Y, Zhang X, Jiao S, et al. Characterization and epidemiological survey of porcine sapelovirus in China. Vet Microbiol. 2019;232:13–21.

    Article  Google Scholar 

  13. Yang T, Li R, Peng W, Ge M, Luo B, Qu T, et al. First isolation and genetic characteristics of porcine sapeloviruses in Hunan, China. Arch Virol. 2017;162(6):1589–97.

    Article  CAS  Google Scholar 

  14. Chen J, Chen F, Zhou Q, Li W, Chen Y, Song Y, et al. Development of a minor groove binder assay for real-time PCR detection of porcine Sapelovirus. J Virol Methods. 2014;198:69–74.

    Article  CAS  Google Scholar 

  15. Racaniello VR. Picornaviridae: The viruses and their replication. In: Knipe DM, Howley PM, editors. Fields virology. Philadelphia: Lippincott Williams & Wilkins; 2001. p. 455–7.

    Google Scholar 

  16. James G, Carol H. Picornaviridae. In: Maclachlan NJ, Dubovi EJ, editors. Fenner’s Veterinary Virology. Amsterdam: Elsevier; 2017. p. 478–81.

    Google Scholar 

  17. Mateu MG. Antibody recognition of picornaviruses and escape from neutralization: a structural view. Virus Res. 1995;38(1):1–24.

    Article  CAS  Google Scholar 

  18. Foo DGW, Alonso S, Phoon MC, Ramachandran NP, Chow VTK, Poh CL. Identification of neutralizing linear epitopes from the VP1 capsid protein of Enterovirus 71 using synthetic peptides. Virus Res. 2007;125(1):61–8.

    Article  CAS  Google Scholar 

  19. Pulli T, Roivainen M, Hovi T, Hyypi T. Induction of neutralizing antibodies by synthetic peptides representing the C terminus of coxsackievirus A9 capsid protein VP1. J Gen Virol. 1998;79(9):2249–53.

    Article  CAS  Google Scholar 

  20. Horsington JJ, Gilkerson JR, Hartley CA. Mapping B-cell epitopes in equine rhinitis B viruses and identification of a neutralising site in the VP1 C-terminus. Vet Microbiol. 2012;155(2):128–36.

    Article  CAS  Google Scholar 

  21. Kriegshäuser G, Wutz G, Lea S, Stuart D, Skern T, Kuechler E. Model of the equine rhinitis A virus capsid: identification of a major neutralizing immunogenic site. J Gen Virol. 2003;84(9):2365–73.

    Article  Google Scholar 

  22. Tamura K, Stecher G, Peterson D, Filipski A, Kumar S. MEGA6: Molecular Evolutionary Genetics Analysis version 6.0. Mol Biol Evol. 2013;30(12):2725–9.

    Article  CAS  Google Scholar 

  23. Guindon S, Dufayard JF, Lefort V, Anisimova M, Hordijk W, Gascuel O. New algorithms and methods to estimate maximum-likelihood phylogenies: assessing the performance of PhyML 3.0. Syst Biol. 2010;59(3):307–21.

    Article  CAS  Google Scholar 

  24. Darriba D, Taboada GL, Doallo R, Posada D. jModelTest 2: more models, new heuristics and parallel computing. Nat Methods. 2012;9(8):772.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the Natural Science Foundation of Jiangxi Province (20202BABL205008), and “Xiujiang” Talent Development Program of Yichun, China (grant 2019-4).

Author information

Authors and Affiliations

Authors

Contributions

TY contributed to the conception, designing, writing and revising the manuscript. LZ contributed to the data collection, laboratory work, drafting, and writing the manuscript. YL and MG contributed to the data analyses and revision of the manuscript. ZZ and AL contributed to the data collection and laboratory work. All authors have read and approved the manuscript.

Corresponding author

Correspondence to Taotao Yang.

Ethics declarations

Ethics approval and consent to participate

Fecal samples used in this study had obtained based on informed consent from farm owners. Collection of fecal samples from swines was approved by the Institutional Animal Care and Use Committee (IACUC) of Yichun University (permit number JXSTUDKY2019009), and all methods were performed in accordance with the relevant guidelines and regulations.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Supplementary Fig. 1.

Development of a SYBR Green I-based real-time PCR method for PSV detection. (a) Standard curves generated from the mean cycle threshold (CT) values obtained against the diluted plasmid standards (log 10 copy number). The correlation coefficient (R2) and the equation of the regression curve (Y) were calculated. Equation: y=-5.1538x+50.716; correlation coefficient: R2=0.9996. (b) Melting curve analysis of real-time PCR based on SYBR Green I. The Tm of PSV real-time PCR was 84.82 °C. (c) Specificity of the PSV SYBR Green I real-time PCR. Only the PSV-HuN1 strain showed a high-intensity fluorescent signal. CSFV, PRRSV, PRV, PEDV, PTV, and JEV did not show specific amplification. (d) Sensitivity of the SYBR Green I real-time PCR. The 10-fold serial dilutions of pMD19-T-PSV plasmids ranging from 5.22×108-5.22×101 copies/μL marked as 1-8, respectively. (e) The agarose gel electrophoresis results of conventional PCR. DNA marker of 5000 bp was used. The 10-fold serial dilutions of pMD19-T-PSV plasmids ranging from 5.22×108-5.22 copies/μL in lanes 1-9, respectively; lane 10 contains the negative control.

Additional file 2: Supplementary Table 1.

Sapelovirus strains used in the study.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. 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 in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, T., Zhang, L., Lu, Y. et al. Characterization of porcine sapelovirus prevalent in western Jiangxi, China. BMC Vet Res 17, 273 (2021). https://doi.org/10.1186/s12917-021-02979-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12917-021-02979-7

Keywords