Next Article in Journal
Poly(rC)-Binding Protein 1 Limits Hepatitis C Virus Virion Assembly and Secretion
Next Article in Special Issue
Recent Progress on Tick-Borne Animal Diseases of Veterinary and Public Health Significance in China
Previous Article in Journal
Stephen Oroszlan and Retroviral Proteins
Previous Article in Special Issue
A Polyvalent Broad-Spectrum Escherichia Phage Tequatrovirus EP01 Capable of Controlling Salmonella and Escherichia coli Contamination in Foods
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Molecular Epidemic Characteristics and Genetic Evolution of Porcine Circovirus Type 2 (PCV2) in Swine Herds of Shanghai, China

1
Shanghai Vocational and Technical College of Agriculture and Forestry, Shanghai 201600, China
2
Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
3
Sub Campus Toba Tek Singh, University of Agriculture, Faisalabad 36050, Pakistan
4
School of Animal Sciences, Yangtze University, Jingzhou 434000, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2022, 14(2), 289; https://doi.org/10.3390/v14020289
Submission received: 20 December 2021 / Revised: 17 January 2022 / Accepted: 26 January 2022 / Published: 29 January 2022
(This article belongs to the Special Issue State-of-the-Art Veterinary Virology Research in China)

Abstract

:
Porcine Circovirus 2 (PCV2) is a crucial swine pathogen and considered a primary causative agent of porcine circovirus-associated diseases (PCVADs), posing a serious economic threat to the swine industry across globe. The world’s biggest agricultural conglomerates have teamed up to create giant commercial pig farms across Shanghai due to the proximity of this region to more affluent lean-pork markets. Since its discovery, PCV2 has displayed extraordinary genetic diversity, and its genome is swiftly evolving through a series of mutations and recombinations. However, limited information on epidemiology, molecular characteristics, vaccine cross-protection, and the co-infection rate of PCV2 with other lethal swine diseases can adversely impact the pig production in the region. To investigate the molecular epidemic characteristics and genetic evolution of PCV2, pigs with doubtful symptoms of PCVADs were sampled from various commercial pig farms with a history of PWMS and/or PDNS across Shanghai from 2014 to 2018. Our results revealed the coexistence of multiple PCV2 genotypes (PCV2b, PCV2e, and PCV2d) among Shanghai pig herds and dominance of PCV2d among them. We also found critical amino acid substitutions in epitope regions of important capsid proteins in PCV2 isolates involved in viral replication and host immune escape. Spotted mutations may favor the prevalence and survival of various PCV2 genotypes despite availability of commercial vaccines. This study also provides insight into the co-infection status of PCV2 with major lethal swine viral diseases such as PPV and PPRSV. Collectively, these investigations will contribute to understanding the molecular epidemiology and evolution of PCV2 across the region.

1. Introduction

Porcine circoviruses (PCVs) are one of the smallest identified animal viruses. They are circular, non-enveloped, single-stranded DNA viruses which belong to the Circovirus genus of the Circoviridae family [1]. PCVs have been categorized into two species so far. Porcine Circovirus type 1 (PCV1), with a genome of 1759 nucleotides, was first detected as a contaminant from the porcine kidney cell line and is classified as non-pathogenic [2]. Porcine Circovirus type 2 (PCV2), with 1767–1768 nucleotides, was initially isolated in Canada in 1991 [3,4,5,6,7] and is considered a primary causative agent of porcine circovirus associated diseases (PCVADs), mainly postweaning multisystemic wasting syndrome, respiratory distress, acute pulmonary edema, reduced growth performance, porcine dermatitis and nephropathy syndrome, and reproductive failures, and is a serious economic threat for the pig industry and wild boar population across the globe [8,9,10].
The PCV2 genome comprises three open reading frames (ORFs). ORF-1 (rep gene) is located on the virus plus-strand and encodes two replicase proteins (Rep and Rep′) that facilitate virus replication [11]. ORF-2 (Cap gene) and ORF-3 are situated on the counter-clockwise strand. ORF-2 encodes the capsid protein [12], and ORF-3 is embedded in ORF-1 and is involved in PCV2-induced apoptotic activities [13,14]. Various genome sequence analyses of PCV2 with phylogenetic clustering analysis, pairwise sequence comparison (PASC), or restriction fragment length polymorphism assays (RFLP) are categorized PCV2 into two distinct genotypes [15,16,17,18] that are defined under different nomenclature in different studies: PCV2 genotypes 1 and 2 [15] groups A and B [19], I and II [20], SG3 and SG1/SG2 [21], 1 and 2 [16], and patterns 321 and 422 [22]. Based on pairwise sequence comparisons, the EU-led consortium on porcine circovirus diseases (www.pcvd.net (accessed 13 December 2021)) proposed a unified and standardized nomenclature for PCV2 genotypes, and later they were characterized into five different genotypes, i.e., PCV2a, PCV2b, PCV2c, PCV2d, and PCV2e [23,24,25,26,27,28,29].
PCV2 ORF-2 genes show diverse genetic variations as compared to ORF-1 and are commonly used as epidemiological and phylogenetic markers [16]. The evolution of PCV2 is the result of genetic combination and mutation, which leads to generation of genetic diversity. Since 2012, the previously predominant PCV2b genotype has started to be replaced by PCV2d in some regions of China, North America, and South Korea, which is the second major genotype shift since the discovery of the virus and may be relevant to changes in vaccine immunity and pathogenicity [29,30]. The potential increase in the virulence of emerged PCV2d and the efficacy of current vaccines derived from PCV2a genotypes against PCV2dhave gained considerable attention.
Virus co-infection plays a crucial role in disease management and control in herds and fields [31,32]. Alongside PCVs, many other viral pathogens impose threats to the swine industry at regional, national, or global levels, including classical swine fever virus (CSFV), pseudorabies virus (PRV), porcine epidemic diarrhea virus (PEDV), and porcine reproductive and respiratory syndrome virus (PRRSV). Previous investigations have revealed that the pathogenesis of PCV2 can be exacerbated by co-infection with additional viral pathogens, such as PEDV and PPRSV [33,34].
PCV2 is encountered as an important challenge to the Chinese pig industry. Shanghai is the biggest eastern coastal province of China with major swine production centers and the largest breeding farms. The prevalence of PCV2′s genotypes, its molecular evolution, and its co-infection with other crucial swine viral diseases in this region are not exactly known. In this study, we have elucidated epidemiological and evolutionary dynamics of PCV2, antigenic variation among PCV2 genotypes, and its co-infection rate with major lethal swine viral diseases in pig herds from Shanghai and surrounding areas from 2014 to 2018.

2. Materials and Methods

2.1. Sample Collection and Viral DNA Extraction

A total of 199 tissue samples (liver, spleen, lymph nodes, and inguinal lymph nodes) were randomly collected from 13 different pig farms across Shanghai, China, with a historical record of post-weaning multisystemic wasting syndrome (PMWS) and/or porcine dermatitis and nephropathy syndrome (PDNS) [35] between the years 2014 and 2018 (details are summarized in Tables 2 and 3). Samples were suspended and homogenized in sterilized phosphate buffered saline (PBS). Homogenized samples were centrifuged for five min at 8000 RPM. Viral DNA/RNA from all tissue samples was extracted using TaKaRa MiniBEST Viral RNA/DNA Extraction Kit (TaKaRa, Beijing, China) according to the manufacturer instructions and stored at −80 °C for further processing.

2.2. Virus Detection

All collected samples were screened through specially designed oligo dT primers (Shanghai Sunny Biotech, Shanghai, China) by Primer Designer v. 2.0 program (Scientific and Educational software) for PCV2, PPV (Porcine Parvo Virus), and PRRSV (Porcine Reproductive and Respiratory Syndrome virus) (primers used are listed in Table 1). Extracted viral DNA/RNA was subjected to PCR and RT-PCR, respectively [32,36]. PCR products were analyzed by gel electrophoresis using 1.5% agarose gel, stained with ethidium bromide, and visualized by UV-based transilluminator. To evade artifacts generated by amplification, the process from PCR to sequencing was repeated and sequences were rechecked for accuracy.

2.3. Virus Amplification and Sequence Determination of Porcine Circovirus Type 2 (PCV2) ORF2

The entire open reading frame 2 (ORF2) of randomly selected PCV2 positive samples (three to five from each farm) was amplified by Primers 5′-AGTTCGTCACCCTTGCGC-3′ (sense primer) and 5′-GCACTTCTTTCGTCGTCAG-3 (antisense primer) using high fidelity LA Taq DNA polymerase (TaKaRa, CA, USA) according to manufacturer protocol. PCR products were subjected to gel electrophoresis, and the PCV2-OFR2 fragment (~705 BP) was recovered by GeneJET Gel Extraction Kit (Thermo Fisher Scientific, Waltham, MA, USA) [37]. The recovered fragment was cloned to pMD 18-T vector (Takara Biomedical Technology, Beijing, China), transformed to DH5 α competent cells, and cultured overnight at 37 °C on Ampicillin agar plates. The clones identified as positive for the respective gene were sequenced in both directions through universal primers (SP6 promoter and T7 primers) with ABI Prism 3730 DNA sequencer (Applied Biosystem, Waltham, MA, USA) at Invitrogen (Guangzhou, China).

2.4. Bioinformatic Analysis

PCV2 genotype group members were classified on the basis of signature motifs [38] and pairwise sequence comparison of ORF2 sequences [16]. Multiple sequence alignment was performed by the Clustal W method. Phylogenetic analysis of recombinants was performed by MEGA5.0 with the kimura 2-parameter model (K2P) [39]. Neighbor Joining (NJ) was used to construct a phylogenetic tree of recombinants, reference strains, available vaccine strains, and other additional PCV2 ORF2 sequences, which were obtained from an NCBI database (http://www.ncbi.nlm.nih.gov (accessed 13 December 2021)). ORF2 amino acid (aa) and nucleotide (nt) comparison of potential recombinants and their parent strain were also analyzed with BioEdit software v7.0.9.0 [40] (details of PCV2 sequences obtained from GenBank are summarized in Supplementary Materials Table S1).

3. Results

3.1. Sample Screening and Co-Infection Status of PCV2

Pigs with doubtful symptoms/clinical history of PCVADs were sampled from various commercial pig farms of Shanghai and surrounding areas. All of these farms (13/13; 100%; farm level) were diagnosed with PCV2 infection with variation in infection and co-infection rates. From the collected tissue samples, 115 out of 199 (57.78%; sample level)screened positive for PCV2 infection. Later, PCV2 positive samples were screened for co-infection status with PPV and PRRSV individually through RT-PCR (PCR data not shown) using specific primers (Table 1). Interestingly, the majority of the PCV2 positive samples were found to be co-infected with PPV at farm (12/13; 92.30%) and sample levels (108/115; 93.91%), whereas co-infection of PCV2 with PRRSV was found to be 46.15% (6/13) and 21.73% (25/115) at farm and sample levels, respectively. The co-existence of PPV2 and PPRV infection in PCV2 positive samples was found to be 38.46% (5/13) at farm level and 15.65% (18/115) at sample level (details are summarized in Table 2).

3.2. PCV2-ORF2 Sequencing and Analysis

ORF2 encodes a viral capsid protein, the major structural PCV2 protein, which is involved in viral replication and host immune responses [41]. Thirteen PCV2 DNA fragments representing the complete ORF2 gene from Shanghai pig farms (one from each farm) were selected and compared with the published sequences deposited in the GenBank database (http://www.ncbi.nlm.nih.gov (accessed 13 December 2021)). Repeated PCR and sequencing of the same samples showed that the methodology used in this study is suitable for accurate amplification of such fragments. The thirteen sequences of the present study and the 28 additional sequences (from China and other countries) retrieved from the GenBank database were analyzed (Supplementary Materials Table S1). The length of the Cap protein coding region varied from 702 to 705 bases. Pairwise similarities of the thirteen new isolates ranged from 90.3 to 100% at nucleotide (nt) level and 87.6 to 99.6% at amino acid (aa) level, respectively.
Examination of typical motifs [5,42,43] and aa substitutions at the Cap region of various PCV2 strains indicated the presence of a typical TNKISI motif for PCV2e, A/TGIE for PCV2b, and SNPLTV and TGID motifs for the majority of PCV2d strains. In addition, various critical amino acid substitutions were spotted at different sites in PCV2 isolates, including antibody epitope regions and an immunodominant decoy epitope. These mutations were found to be associated with antibody recognition, virulence, and an immune escape mechanism of PCV2 in previous studies [44,45,46,47] (Figure 1). As a result of substitution at the stop codon, some strains exhibited an extended lysine (K) residue encoded by AAA or AAG.
The PCV2d genotype harbored eight unique amino acid mutations, i.e., F53→I53, A68→N68, I/R89→L89, S90→T90, T134→N143, S169→R/G169, S/A190-T190, and V215→I215. PCV2b harbored two unique mutations, i.e., 57V→57I and 89I→89R, whereas various PCV2e substitutions were spotted at position 47, 72, 130, 131, 133,185, 187, and 191. All these critical mutations spotted in different PCV2 genotypes are summarized in Figure 2.

3.3. Phylogenetic Analysis and Genotype Determination

Porcine circoviruses’ genotype shifting from PCV2b to PCV2d has been reported in numerous regions of China, indicating dominance of PCV2d [29,30,49,50,51]. To investigate the dominant circulating genotype and emerging strains in the investigation area and to compare them with other native and non-native strains, a phylogenetic tree was established based on a publicly available PCV2-ORF2 sequence assembled from GenBank and newly isolated PCV2-ORF2 sequences which were deposited to the GenBank database (Figure 3).
Phylogenetic analysis revealed that thirteen new isolates from Shanghai belong to three distinct genetic groups. One of thirteen (7.6%) isolates belongs to PCV2b, one of thirteen (7.6%) isolates belongs to PCV2e, and eleven of thirteen (84.6%) isolates belong to PCV2d, indicating that PCV2d was the prominent genotype circulating in Shanghai, China from 2014 to 2018 (Table 3). PCV2d isolated strains, SH150127-1, SH140624-5, SH140718-1, SH140704-1, SH140704-3, SH140408-1, SH140408-3, SH140625-2, SH1601, SH1701, and SH1801 share their ancestor backs with strain HM038031, GQ3590101, JX948771, and other isolates from Shanghai, China, in various years [27,52]. The PCV2b isolated strain shared its ancestor back with commercially available vaccine strain FJ598044, which was detected in Wuhan, Hebei, China in 2008 [46], whereas PCV2e strain KR058357 was related to strains EF524533 and KM360057 isolated from Beijing and Guangdong in 2006 and 2013, respectively [53]. The PCV2a strain contains clusters from China, Canada, and South Korea, PCV2b contains clusters from China (Shanghai, Beijing, and Wuhan) and France, PCV2c contains clusters from Denmark and China, and PCV2e contains clusters from China, whereas PCV2d contains clusters from China and includes the majority of our sequences isolated from Shanghai. Interestingly, the phylogenetic tree showed a high level of divergence among PCV2d isolates from Shanghai (Figure 3).

4. Discussion

PCV2 is accepted as an essential infectious pathogen of PMWS and/or PDNS, economically important diseases that affect both wild and domestic pigs [54,55,56,57]. It is the cause of severe financial losses across the global swine industry and needs to be controlled to improve performance and production. In China, PCVADs have been a major concern for the swine industry since the first domestic outbreak caused by PCV2 in 2002 [58]. Previous investigations have shown that the positive rate of PCV2 infection in swine herds in China was over 50%, and some regions are considered as highly epidemic or endemic for PCV2-associated infections [37,49,59]. As one of the dynamic regions in China regarding breeding of food animals, Shanghai Province raises a dense pig population. This eastern coastal region is highly endemic or epidemic for PCV2 and its associated infections in swine herds, which pose a serious threat to the industry that shoulders the pork food chain across a population of millions.
The genetic diversity of PCV2 is increasing continuously, and a variety of novel variant strains have rapidly emerged in recent years. Based on phylogenetic analysis of its capsid gene, PCV2, is characterized into five genotypes, i.e., PCV2a, PCV2b, PCV2c, PCV2d, and PCV2e [42,44,60]. All currently documented genotypes of PCV2 were identified as circulating in China except for PCV2c, which was only reported in Denmark, Brazil, and Namibia [61,62,63,64]. A shift from PCV2a to PCV2b was seen on a global scale and in China in or prior to 2003, signifying the importance of PCV2b [65,66]. PCV2d, a recently emerged genotype, was spotted in China in 2007 [27], and a massive second genotype shift to PCV2d has occurred on a nationwide scale since the discovery of the virus [42,67]. In this study, 84.6% (11 of 13) of isolated PCV2 strains belonged to genotype PCV2d, 7.6% (1 of 13) were classified under genotype PCV2b, and 7.6% (1 of 13) under genotype PCV2e, signifying the co-circulation of three genotypes in the pig population of the region and dominance of PCV2d among them [30,68].
ORF2 encodes the viral capsid protein, the major structural PCV2 protein that is involved in viral replication and host immune responses. Fewer mutations in this region may influence virus pathogenicity [41]. ORF2 is used as a phylogenetic marker for PCV2 strains because of its competence to reconstruct the same tree as the whole viral genome. Here, several signature motifs consistent with previous studies have been identified on the Cap gene in different genotypes, such as amino acids TNKISI (aa 86 to 91) for PCV2e, S/PNPRSV and A/TGIE (aa 190, 191, 206, and 210) for PCV2b [5,43], and SNPLTV and TGID for PCV2d [42]. Several aa changes are found in reported antibody epitope regions and an immunodominant decoy epitope [45,48,69] of the capsid protein. Furthermore, aa 133–134, which are responsible for the virulence of PCV2, have mutated in some of the strains in this study (Figure 2) [47].
PCV2 vaccination has been found to be highly effectual in inducing protective immunity against PCVAD and PCV2 infection under both field [70] and experimental conditions [71]. The broad use of vaccination may have controlled the disease until now, but the continuous existence of PCVD in swine herds indicates that the virus might have evolved and changed itself in an efficient manner. Selection particles contribute to viral evolution by continuously amending their antigenic properties to escape from the host’s immune defense mechanism. Selective pressure can be detected in the ORFs of the PCV2 genome [16]. The three positive sites (aa 134, 190, and 191) in the epitope region [68] directly affect the pathogenicity, virulence, and neutralizing activities of antibodies [47]. Interestingly, mutations were also seen at these positions among different genotypes of Shanghai isolates, suggesting that the amino acid mutations at these sites may have contributed to the immune escape of PCV2 in the region, leading to a great challenge for the control of PCV2 despite availability of vaccines.
Possible immunoactive regions have been reported previously [69,72,73] in the Cap gene of PCV2, with four antibody recognition domains at amino acid epitopes of 50–81, 113–134, 161–208, and 227–233 [45]. In our study, several amino acid mutations were also spotted at these regions in the isolates, particularly at some epitopes’ associated positions, as mentioned in Figure 2 and Figure 3. Alanine at position 59, threonine at position 190, and glutamic acid at position 191 were previously found to be components of conformational neutralizing epitopes [74,75]. Asparagine at position 77 and isoleucine at position 206 are reported to be key residues essential for antibody recognition [72]. The mutations at these sites on the Cap protein are possibly responsible for antigenic variation among PCV2 genotypes, which may favor survival, virulence, and circulation of PCV2 strains among vaccinated swine herds. However, the connection between virus pathogenicity, dominance, and mutations still needs to be further investigated.
Pigs are commonly identified with co-infection of multiple pathogens in field conditions [30,31,32]. Studies in the past have highlighted that pigs can be co-infected with PCV2 with several pathogens, such as CSFV, PRRSV, PEDV, PRV, and PPV. Some investigations have demonstrated 21.9–52.3% co-infection rates of PCV2 with PRRSV [27,30,76,77] and 33–67% with PPV [78,79,80]. Various patterns of combined infections were assessed in this study with positive rates of 92.30% for PCV2 with PPV and 46.15% for PCV2 with PRRSV at farm levels (Table 2). Our results revealed that there is a high frequency of co-infection of PCV2 and PPV in Shanghai Province. Combined with former investigations, the results of this study indicate that different patterns of dual or multiple infection are commonly observed in the intensive swine breeding system in China. Discussion on refinement of animal disease control strategies was driven by the intricated co-infection status shown [30,31,32,76,77,81]. Therefore, it is of overriding importance to take strict and dynamic measures such as disease surveillance and variant monitoring, analyzing vaccine efficacy, strengthening diagnostic tests, and improving biosecurity and environmental hygiene with a declaration of these measures as an integral part of current PCV2 control protocols.

5. Conclusions

PCV2 is continuously evolving, leading to rapid emergence of new variant strains. Here, we investigated the prevalence and molecular characterization of PCV2 as well its co-infection status in Shanghai Province from 2014 to 2018. Our results revealed the coexistence of multiple genotypes (PCV2b, PCV2e, and PCV2d) among Shanghai vaccinated pig herds and dominance of PCV2d among them. Here, we also found critical amino acid substitutions at the capsid region responsible for antigenic variation, virulence, and immune escape, which may favor the prevalence and survival of various PCV2 strains despite mass vaccination. This study also provides data on and insight into the co-infection status of PCV2 with major lethal swine viral diseases. This study collectively contributes an understanding of the molecular epidemiology and evolution of PCV2 across China and sheds light on the importance of taking forceful measures for prevention and control of emerging swine diseases across the region.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/v14020289/s1, Table S1: Summary of reference sequences of PCV2 obtained from NCBI and analyzed in this study.

Author Contributions

Conceptualization, J.W., Z.M. and D.Z.; methodology, L.K., A.W. and K.S.; software, B.E.M., D.S. and B.L.; validation, Y.Z., K.S. and L.K.; formal analysis, A.W., B.E.M. and J.Z.; investigation, L.K., Y.Z., Z.L. and B.L.; resources, J.W., Z.M., Y.Q. and D.Z.; data curation, K.L., B.L. and D.S.; writing—original draft preparation, A.W. and J.W.; writing—review and editing, A.W., D.Z., Z.M. and J.W.; visualization, Z.M. and J.W.; supervision, Z.M., J.W. and D.Z.; project administration, J.W., Z.M. and D.Z.; funding acquisition, J.W. and Z.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Shanghai Agriculture Applied Technology Development Program, China (Grant No.X2021-02-08-00-12-F00770), the National Basic Research Program (973 Plan) (No.2014CB542703), the National Natural Science Foundation of China (31972693), and the National Key Research and Development Program of China (No.2018YFD0500101).

Institutional Review Board Statement

The study was conducted according to the animal ethics guidelines of China and approved by the Institutional Animal Care and Use Committee of Shanghai Veterinary Research Institute (IACUC No.: Shvri-mo-2019080606, 6 August 2019).

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We are highly thankful to YuHao Li, Di Di, Xin Wang, Anni Ou and Shuang Gao for assistance in sampling.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tischer, I.; Mields, W.; Wolff, D.; Vagt, M.; Griem, W. Studies on epidemiology and pathogenicity of porcine circovirus. Arch. Virol. 1986, 91, 271–276. [Google Scholar] [CrossRef] [PubMed]
  2. Tischer, I.; Rasch, R.; Tochtermann, G. Characterization of papovavirus-and picornavirus-like particles in permanent pig kidney cell lines. Zenibl. Bukt. Orig A 1974, 226, 153–167. [Google Scholar]
  3. Hamel, A.L.; Lin, L.L.; Nayar, G.P. Nucleotide sequence of porcine circovirus associated with postweaning multisystemic wasting syndrome in pigs. J. Virol. 1998, 72, 5262–5267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Meehan, B.M.; McNeilly, F.; Todd, D.; Kennedy, S.; Jewhurst, V.A.; Ellis, J.A.; Hassard, L.E.; Clark, E.G.; Haines, D.M.; Allan, G.M. Characterization of novel circovirus DNAs associated with wasting syndromes in pigs. J. Gen. Virol. 1998, 79, 2171–2179. [Google Scholar] [CrossRef]
  5. Cheung, A.K.; Lager, K.M.; Kohutyuk, O.I.; Vincent, A.L.; Henry, S.C.; Baker, R.B.; Rowland, R.R.; Dunham, A.G. Detection of two porcine circovirus type 2 genotypic groups in United States swine herds. Arch. Virol. 2007, 152, 1035–1044. [Google Scholar] [CrossRef]
  6. Harding, J.C. Postweaning multisystemic wasting syndrome: Epidemiology and clinical presentation. J. Swine Health Prod. 1998, 6, 249–254. [Google Scholar]
  7. Mankertz, A.; Domingo, M.; Folch, J.M.; LeCann, P.; Jestin, A.; Segalés, J.; Chmielewicz, B.; Plana-Durán, J.; Soike, D. Characterisation of PCV-2 isolates from Spain, Germany and France. Virus Res. 2000, 66, 65–77. [Google Scholar] [CrossRef]
  8. Trible, B.R.; Suddith, A.W.; Kerrigan, M.A.; Cino-Ozuna, A.G.; Hesse, R.A.; Rowland, R.R. Recognition of the different structural forms of the capsid protein determines the outcome following infection with porcine circovirus type 2. J. Virol. 2012, 86, 13508–13514. [Google Scholar] [CrossRef] [Green Version]
  9. Chae, C. A review of porcine circovirus 2-associated syndromes and diseases. Vet. J. 2005, 169, 326–336. [Google Scholar] [CrossRef]
  10. Cadar, D.; Cságola, A.; Lőrincz, M.; Tombácz, K.; Spînu, M.; Tuboly, T. Detection of natural inter- and intra-genotype recombination events revealed by cap gene analysis and decreasing prevalence of PCV2 in wild boars. Infect. Genet. Evol. 2012, 12, 420–427. [Google Scholar] [CrossRef]
  11. Mankertz, A.; Mankertz, J.; Wolf, K.; Buhk, H.J. Identification of a protein essential for replication of porcine circovirus. J. Gen. Virol. 1998, 79, 381–384. [Google Scholar] [CrossRef] [PubMed]
  12. Finsterbusch, T.; Mankertz, A. Porcine circoviruses—Small but powerful. Virus Res. 2009, 143, 177–183. [Google Scholar] [CrossRef] [PubMed]
  13. Liu, J.; Chen, I.; Kwang, J. Characterization of a previously unidentified viral protein in porcine circovirus type 2-infected cells and its role in virus-induced apoptosis. J. Virol. 2005, 79, 8262–8274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Liu, J.; Chen, I.; Du, Q.; Chua, H.; Kwang, J. The ORF3 protein of porcine circovirus type 2 is involved in viral pathogenesis in vivo. J. Virol. 2006, 80, 5065–5073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Grau-Roma, L.; Crisci, E.; Sibila, M.; López-Soria, S.; Nofrarias, M.; Cortey, M.; Fraile, L.; Olvera, A.; Segalés, J. A proposal on porcine circovirus type 2 (PCV2) genotype definition and their relation with postweaning multisystemic wasting syndrome (PMWS) occurrence. Vet. Microbiol. 2008, 128, 23–35. [Google Scholar] [CrossRef] [Green Version]
  16. Olvera, A.; Cortey, M.; Segalés, J. Molecular evolution of porcine circovirus type 2 genomes: Phylogeny and clonality. Virology 2007, 357, 175–185. [Google Scholar] [CrossRef] [Green Version]
  17. Wen, L.; Guo, X.; Yang, H. Genotyping of porcine circovirus type 2 from a variety of clinical conditions in China. Vet. Microbiol. 2005, 110, 141–146. [Google Scholar] [CrossRef]
  18. Hamel, A.L.; Lin, L.L.; Sachvie, C.; Grudeski, E.; Nayar, G.P. PCR detection and characterization of type-2 porcine circovirus. Can. J. Vet. Res. 2000, 64, 44–52. [Google Scholar]
  19. Martins Gomes de Castro, A.M.; Cortez, A.; Heinemann, M.B.; Brandão, P.E.; Richtzenhain, L.J. Genetic diversity of Brazilian strains of porcine circovirus type 2 (PCV-2) revealed by analysis of the cap gene (ORF-2). Arch. Virol. 2007, 152, 1435–1445. [Google Scholar] [CrossRef]
  20. De Boisséson, C.; Béven, V.; Bigarré, L.; Thiéry, R.; Rose, N.; Eveno, E.; Madec, F.; Jestin, A. Molecular characterization of Porcine circovirus type 2 isolates from post-weaning multisystemic wasting syndrome-affected and non-affected pigs. J. Gen. Virol. 2004, 85, 293–304. [Google Scholar] [CrossRef]
  21. Timmusk, S.; Wallgren, P.; Belák, K.; Berg, M.; Fossum, C. In Genetic analysis of PCV2 capsid protein sequences reveals two main groups of Swedish isolates. In Proceedings of the International Conference on Animal Circoviruses and Associated Diseases, Belfast, Ireland, 11–13 July 2005. [Google Scholar]
  22. Carman, S.; McEwen, B.; DeLay, J.; van Dreumel, T.; Lusis, P.; Cai, H.; Fairles, J. Porcine circovirus-2 associated disease in swine in Ontario (2004 to 2005). Can. Vet. J. 2006, 47, 761–762. [Google Scholar] [PubMed]
  23. Segalés, J.; Olvera, A.; Grau-Roma, L.; Charreyre, C.; Nauwynck, H.; Larsen, L.; Dupont, K.; McCullough, K.; Ellis, J.; Krakowka, S.; et al. PCV-2 genotype definition and nomenclature. Vet. Rec. 2008, 162, 867–868. [Google Scholar] [CrossRef] [PubMed]
  24. Dupont, K.; Hjulsager, C.K.; Kristensen, C.S.; Baekbo, P.; Larsen, L.E. Transmission of different variants of PCV2 and viral dynamics in a research facility with pigs mingled from PMWS-affected herds and non-affected herds. Vet. Microbiol. 2009, 139, 219–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Dupont, K.; Nielsen, E.O.; Baekbo, P.; Larsen, L.E. Genomic analysis of PCV2 isolates from Danish archives and a current PMWS case-control study supports a shift in genotypes with time. Vet. Microbiol. 2008, 128, 56–64. [Google Scholar] [CrossRef] [Green Version]
  26. Jantafong, T.; Boonsoongnern, A.; Poolperm, P.; Urairong, K.; Lekcharoensuk, C.; Lekcharoensuk, P. Genetic characterization of porcine circovirus type 2 in piglets from PMWS-affected and -negative farms in Thailand. Virol. J. 2011, 8, 88. [Google Scholar] [CrossRef] [Green Version]
  27. Guo, L.J.; Lu, Y.H.; Wei, Y.W.; Huang, L.P.; Liu, C.M. Porcine circovirus type 2 (PCV2): Genetic variation and newly emerging genotypes in China. Virol. J. 2010, 7, 273. [Google Scholar] [CrossRef] [Green Version]
  28. Gagnon, C.A.; Tremblay, D.; Tijssen, P.; Venne, M.H.; Houde, A.; Elahi, S.M. The emergence of porcine circovirus 2b genotype (PCV-2b) in swine in Canada. Can. Vet. J. 2007, 48, 811–819. [Google Scholar]
  29. Karuppannan, A.K.; Opriessnig, T. Porcine Circovirus Type 2 (PCV2) Vaccines in the Context of Current Molecular Epidemiology. Viruses 2017, 9, 99. [Google Scholar] [CrossRef] [Green Version]
  30. Ma, Z.; Liu, M.; Liu, Z.; Meng, F.; Wang, H.; Cao, L.; Li, Y.; Jiao, Q.; Han, Z.; Liu, S. Epidemiological investigation of porcine circovirus type 2 and its coinfection rate in Shandong province in China from 2015 to 2018. BMC Vet. Res. 2021, 17, 17. [Google Scholar] [CrossRef]
  31. Chen, N.; Huang, Y.; Ye, M.; Li, S.; Xiao, Y.; Cui, B.; Zhu, J. Co-infection status of classical swine fever virus (CSFV), porcine reproductive and respiratory syndrome virus (PRRSV) and porcine circoviruses (PCV2 and PCV3) in eight regions of China from 2016 to 2018. Infect. Genet. Evol. 2019, 68, 127–135. [Google Scholar] [CrossRef]
  32. Guo, Z.; Ruan, H.; Qiao, S.; Deng, R.; Zhang, G. Co-infection status of porcine circoviruses (PCV2 and PCV3) and porcine epidemic diarrhea virus (PEDV) in pigs with watery diarrhea in Henan province, central China. Microb. Pathog. 2020, 142, 104047. [Google Scholar] [CrossRef] [PubMed]
  33. Ssemadaali, M.A.; Ilha, M.; Ramamoorthy, S. Genetic diversity of porcine circovirus type 2 and implications for detection and control. Res. Vet. Sci. 2015, 103, 179–186. [Google Scholar] [CrossRef] [PubMed]
  34. Jung, K.; Kim, J.; Ha, Y.; Choi, C.; Chae, C. The effects of transplacental porcine circovirus type 2 infection on porcine epidemic diarrhoea virus-induced enteritis in preweaning piglets. Vet. J. 2006, 171, 445–450. [Google Scholar] [CrossRef] [PubMed]
  35. Allan, G.M.; McNeilly, F.; Kennedy, S.; Daft, B.; Clarke, E.G.; Ellis, J.A.; Haines, D.M.; Meehan, B.M.; Adair, B.M. Isolation of porcine circovirus-like viruses from pigs with a wasting disease in the USA and Europe. J. Vet. Diagn. Investig. 1998, 10, 3–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Li, L.; Yuan, W.; Guo, H.; Ma, Z.; Song, Q.; Wang, X.; Li, H. Prevalence and genetic variation of porcine circovirus type 2 in Hebei, China from 2004 to 2014. Gene 2016, 586, 222–227. [Google Scholar] [CrossRef]
  37. Wei, C.; Zhang, M.; Chen, Y.; Xie, J.; Huang, Z.; Zhu, W.; Xu, T.; Cao, Z.; Zhou, P.; Su, S.; et al. Genetic evolution and phylogenetic analysis of porcine circovirus type 2 infections in southern China from 2011 to 2012. Infect. Genet. Evol. 2013, 17, 87–92. [Google Scholar] [CrossRef]
  38. An, D.J.; Roh, I.S.; Song, D.S.; Park, C.K.; Park, B.K. Phylogenetic characterization of porcine circovirus type 2 in PMWS and PDNS Korean pigs between 1999 and 2006. Virus Res. 2007, 129, 115–122. [Google Scholar] [CrossRef]
  39. 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, 2731–2739. [Google Scholar] [CrossRef] [Green Version]
  40. Hall, T.A. BioEdit: A User-Friendly Biological Sequence Alignment Editor and Analysis Program for Windows 95/98/NT, Nucleic Acids Symposium Series; Information Retrieval Ltd.: London, UK, 1999; pp. 95–98. [Google Scholar]
  41. Nawagitgul, P.; Morozov, I.; Bolin, S.R.; Harms, P.A.; Sorden, S.D.; Paul, P.S. Open reading frame 2 of porcine circovirus type 2 encodes a major capsid protein. J. Gen. Virol. 2000, 81, 2281–2287. [Google Scholar] [CrossRef]
  42. Xiao, C.T.; Halbur, P.G.; Opriessnig, T. Global molecular genetic analysis of porcine circovirus type 2 (PCV2) sequences confirms the presence of four main PCV2 genotypes and reveals a rapid increase of PCV2d. J. Gen. Virol. 2015, 96, 1830–1841. [Google Scholar] [CrossRef]
  43. Cheung, A.K. Homologous recombination within the capsid gene of porcine circovirus type 2 subgroup viruses via natural co-infection. Arch. Virol. 2009, 154, 531–534. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, J.; Wei, C.; Dai, A.; Lin, Z.; Fan, K.; Fan, J.; Liu, J.; Luo, M.; Yang, X. Detection of PCV2e strains in Southeast China. PeerJ 2018, 6, e4476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Trible, B.R.; Kerrigan, M.; Crossland, N.; Potter, M.; Faaberg, K.; Hesse, R.; Rowland, R.R. Antibody recognition of porcine circovirus type 2 capsid protein epitopes after vaccination, infection, and disease. Clin. Vaccine Immunol. 2011, 18, 749–757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Zhai, S.-L.; Chen, S.-N.; Xu, Z.-H.; Tang, M.-H.; Wang, F.-G.; Li, X.-J.; Sun, B.-B.; Deng, S.-F.; Hu, J.; Lv, D.-H.; et al. Porcine circovirus type 2 in China: An update on and insights to its prevalence and control. Virol. J. 2014, 11, 88. [Google Scholar] [CrossRef] [Green Version]
  47. Krakowka, S.; Allan, G.; Ellis, J.; Hamberg, A.; Charreyre, C.; Kaufmann, E.; Brooks, C.; Meehan, B. A nine-base nucleotide sequence in the porcine circovirus type 2 (PCV2) nucleocapsid gene determines viral replication and virulence. Virus Res. 2012, 164, 90–99. [Google Scholar] [CrossRef] [PubMed]
  48. Trible, B.R.; Rowland, R.R. Genetic variation of porcine circovirus type 2 (PCV2) and its relevance to vaccination, pathogenesis and diagnosis. Virus Res. 2012, 164, 68–77. [Google Scholar] [CrossRef]
  49. Zheng, G.; Lu, Q.; Wang, F.; Xing, G.; Feng, H.; Jin, Q.; Guo, Z.; Teng, M.; Hao, H.; Li, D.; et al. Phylogenetic analysis of porcine circovirus type 2 (PCV2) between 2015 and 2018 in Henan Province, China. BMC Vet. Res. 2020, 16, 6. [Google Scholar] [CrossRef]
  50. Xia, D.; Huang, L.; Xie, Y.; Zhang, X.; Wei, Y.; Liu, D.; Zhu, H.; Bian, H.; Feng, L.; Liu, C. The prevalence and genetic diversity of porcine circovirus types 2 and 3 in Northeast China from 2015 to 2018. Arch. Virol. 2019, 164, 2435–2449. [Google Scholar] [CrossRef]
  51. Wang, Y.; Noll, L.; Lu, N.; Porter, E.; Stoy, C.; Zheng, W.; Liu, X.; Peddireddi, L.; Niederwerder, M.; Bai, J. Genetic diversity and prevalence of porcine circovirus type 3 (PCV3) and type 2 (PCV2) in the Midwest of the USA during 2016–2018. Transbound. Emerg. Dis. 2020, 67, 1284–1294. [Google Scholar] [CrossRef]
  52. Zhai, S.-L.; Chen, S.-N.; Zhang, J.-W.; Wei, Z.-Z.; Long, J.-X.; Yuan, S.-S.; Wei, W.-K.; Chen, Q.-L.; Xuan, H.; Wu, D.-C. Dissection of the possible routes on porcine circoviruses infecting human. J. Anim. Vet. Adv. 2012, 11, 1281–1286. [Google Scholar]
  53. Wang, F.; Guo, X.; Ge, X.; Wang, Z.; Chen, Y.; Cha, Z.; Yang, H. Genetic variation analysis of Chinese strains of porcine circovirus type 2. Virus Res. 2009, 145, 151–156. [Google Scholar] [CrossRef]
  54. Segales, J.; Domingo, M.; Latimer, K. In Porcine circovirus is present in cases of porcine dermatitis and nephropathy syndrome (PDNS). In Proceedings of the 15th International Pig Veterinary Society Congress, Birmingham, UK, 5–9 July 1998; p. 215. [Google Scholar]
  55. Segalés, J.; Domingo, M. Postweaning multisystemic wasting syndrome (PMWS) in pigs. A review. Vet. Q. 2002, 24, 109–124. [Google Scholar] [CrossRef] [PubMed]
  56. Lipej, Z.; Segalés, J.; Jemersić, L.; Olvera, A.; Roić, B.; Novosel, D.; Mihaljević, Z.; Manojlović, L. First description of postweaning multisystemic wasting syndrome (PMWS) in wild boar (Sus scrofa) in Croatia and phylogenetic analysis of partial PCV2 sequences. Acta Vet. Hung. 2007, 55, 389–404. [Google Scholar] [CrossRef] [Green Version]
  57. Sofia, M.; Billinis, C.; Psychas, V.; Birtsas, P.; Sofianidis, G.; Leontides, L.; Knowles, N.; Spyrou, V. Detection and genetic characterization of porcine circovirus 2 isolates from the first cases of postweaning multisystemic and wasting syndrome in wild boars in Greece. J. Wildl. Dis. 2008, 44, 864–870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Yang, H. Epidemical characterizations and control strategies of swine immunosuppressive diseases (PRRS and PMWS). Chin. J. Anim. Husband Vet. Med. 2004, 31, 41–43. [Google Scholar]
  59. Sun, J.; Huang, L.; Wei, Y.; Wang, Y.; Chen, D.; Du, W.; Wu, H.; Liu, C. Prevalence of emerging porcine parvoviruses and their co-infections with porcine circovirus type 2 in China. Arch. Virol. 2015, 160, 1339–1344. [Google Scholar] [CrossRef] [PubMed]
  60. Davies, B.; Wang, X.; Dvorak, C.M.T.; Marthaler, D.; Murtaugh, M.P. Diagnostic phylogenetics reveals a new Porcine circovirus 2 cluster. Virus Res. 2016, 217, 32–37. [Google Scholar] [CrossRef] [Green Version]
  61. Liu, X.; Wang, F.X.; Zhu, H.W.; Sun, N.; Wu, H. Phylogenetic analysis of porcine circovirus type 2 (PCV2) isolates from China with high homology to PCV2c. Arch. Virol. 2016, 161, 1591–1599. [Google Scholar] [CrossRef]
  62. Jiang, C.-G.; Wang, G.; Tu, Y.-B.; Liu, Y.-G.; Wang, S.-J.; Cai, X.-H.; An, T.-Q. Genetic analysis of porcine circovirus type 2 in China. Arch. Virol. 2017, 162, 2715–2726. [Google Scholar] [CrossRef]
  63. Franzo, G.; Cortey, M.; de Castro, A.M.; Piovezan, U.; Szabo, M.P.; Drigo, M.; Segalés, J.; Richtzenhain, L.J. Genetic characterisation of Porcine circovirus type 2 (PCV2) strains from feral pigs in the Brazilian Pantanal: An opportunity to reconstruct the history of PCV2 evolution. Vet. Microbiol. 2015, 178, 158–162. [Google Scholar] [CrossRef]
  64. Molini, U.; Franzo, G.; Gous, L.; Moller, S.; Hemberger, Y.M.; Chiwome, B.; Marruchella, G.; Khaiseb, S.; Cattoli, G.; Dundon, W.G. Three different genotypes of porcine circovirus 2 (PCV-2) identified in pigs and warthogs in Namibia. Arch. Virol. 2021, 166, 1723–1728. [Google Scholar] [CrossRef] [PubMed]
  65. Beach, N.M.; Ramamoorthy, S.; Opriessnig, T.; Wu, S.Q.; Meng, X.J. Novel chimeric porcine circovirus (PCV) with the capsid gene of the emerging PCV2b subtype cloned in the genomic backbone of the non-pathogenic PCV1 is attenuated in vivo and induces protective and cross-protective immunity against PCV2b and PCV2a subtypes in pigs. Vaccine 2010, 29, 221–232. [Google Scholar] [CrossRef] [PubMed]
  66. Li, W.; Wang, X.; Ma, T.; Feng, Z.; Li, Y.; Jiang, P. Genetic analysis of porcine circovirus type 2 (PCV2) strains isolated between 2001 and 2009: Genotype PCV2b predominate in postweaning multisystemic wasting syndrome occurrences in eastern China. Virus Genes 2010, 40, 244–251. [Google Scholar] [CrossRef] [PubMed]
  67. Kwon, T.; Lee, D.U.; Yoo, S.J.; Je, S.H.; Shin, J.Y.; Lyoo, Y.S. Genotypic diversity of porcine circovirus type 2 (PCV2) and genotype shift to PCV2d in Korean pig population. Virus Res. 2017, 228, 24–29. [Google Scholar] [CrossRef]
  68. Yang, S.; Yin, S.; Shang, Y.; Liu, B.; Yuan, L.; Zafar Khan, M.U.; Liu, X.; Cai, J. Phylogenetic and genetic variation analyses of porcine circovirus type 2 isolated from China. Transbound. Emerg. Dis. 2018, 65, e383–e392. [Google Scholar] [CrossRef]
  69. Mahé, D.; Blanchard, P.; Truong, C.; Arnauld, C.; Le Cann, P.; Cariolet, R.; Madec, F.; Albina, E.; Jestin, A. Differential recognition of ORF2 protein from type 1 and type 2 porcine circoviruses and identification of immunorelevant epitopes. J. Gen. Virol. 2000, 81, 1815–1824. [Google Scholar] [CrossRef]
  70. Fachinger, V.; Bischoff, R.; Jedidia, S.B.; Saalmüller, A.; Elbers, K. The effect of vaccination against porcine circovirus type 2 in pigs suffering from porcine respiratory disease complex. Vaccine 2008, 26, 1488–1499. [Google Scholar] [CrossRef]
  71. Fort, M.; Sibila, M.; Allepuz, A.; Mateu, E.; Roerink, F.; Segalés, J. Porcine circovirus type 2 (PCV2) vaccination of conventional pigs prevents viremia against PCV2 isolates of different genotypes and geographic origins. Vaccine 2008, 26, 1063–1071. [Google Scholar] [CrossRef]
  72. Khayat, R.; Brunn, N.; Speir, J.A.; Hardham, J.M.; Ankenbauer, R.G.; Schneemann, A.; Johnson, J.E. The 2.3-angstrom structure of porcine circovirus 2. J. Virol. 2011, 85, 7856–7862. [Google Scholar] [CrossRef] [Green Version]
  73. Lekcharoensuk, P.; Morozov, I.; Paul, P.S.; Thangthumniyom, N.; Wajjawalku, W.; Meng, X.J. Epitope mapping of the major capsid protein of type 2 porcine circovirus (PCV2) by using chimeric PCV1 and PCV2. J. Virol. 2004, 78, 8135–8145. [Google Scholar] [CrossRef] [Green Version]
  74. Saha, D.; Lefebvre, D.J.; Ooms, K.; Huang, L.; Delputte, P.L.; Van Doorsselaere, J.; Nauwynck, H.J. Single amino acid mutations in the capsid switch the neutralization phenotype of porcine circovirus 2. J. Gen. Virol. 2012, 93, 1548–1555. [Google Scholar] [CrossRef] [PubMed]
  75. Huang, L.P.; Lu, Y.H.; Wei, Y.W.; Guo, L.J.; Liu, C.M. Identification of one critical amino acid that determines a conformational neutralizing epitope in the capsid protein of porcine circovirus type 2. BMC Microbiol. 2011, 11, 188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Liu, J.K.; Wei, C.H.; Yang, X.Y.; Dai, A.L.; Li, X.H. Multiplex PCR for the simultaneous detection of porcine reproductive and respiratory syndrome virus, classical swine fever virus, and porcine circovirus in pigs. Mol. Cell. Probes 2013, 27, 149–152. [Google Scholar] [CrossRef] [PubMed]
  77. Ge, X.; Wang, F.; Guo, X.; Yang, H. Porcine circovirus type 2 and its associated diseases in China. Virus Res. 2012, 164, 100–106. [Google Scholar] [CrossRef]
  78. Ellis, J.; Hassard, L.; Clark, E.; Harding, J.; Allan, G.; Willson, P.; Strokappe, J.; Martin, K.; McNeilly, F.; Meehan, B.; et al. Isolation of circovirus from lesions of pigs with postweaning multisystemic wasting syndrome. Can. Vet. J. 1998, 39, 44–51. [Google Scholar] [PubMed]
  79. Pescador, C.A.; Bandarra, P.M.; Castro, L.A.; Antoniassi, N.A.B.; Ravazzolo, A.P.; Sonne, L.; Cruz, C.E.F.; Driemeier, D. Co-infection by porcine circovirus type 2 and porcine parvovirus in aborted fetuses and stillborn piglets in southern Brazil. Pesqui. Veterinária Bras. 2007, 27, 425–429. [Google Scholar] [CrossRef]
  80. Saekhow, P.; Kishizuka, S.; Sano, N.; Mitsui, H.; Akasaki, H.; Mawatari, T.; Ikeda, H. Coincidental detection of genomes of porcine parvoviruses and porcine circovirus type 2 infecting pigs in Japan. J. Vet. Med. Sci. 2016, 77, 1581–1586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Qu, T.; Li, R.; Yan, M.; Luo, B.; Yang, T.; Yu, X. High prevalence of PCV2d in Hunan province, China: A retrospective analysis of samples collected from 2006 to 2016. Arch. Virol. 2018, 163, 1897–1906. [Google Scholar] [CrossRef]
Figure 1. Alignment of capsid protein sequences of thirteen PCV2 isolates: (A,B); The grey highlighted area represents antibody recognition domains as described by Trible et al. (2011) [45]. The red solid line boxes show motifs of PCV2 determined by Cheung et al. (2007) and Cheung et al. (2009) [5,43]. The dashed green line represents the location of the immunodominant decoy epitope, which shows reactivity of PCV2 infected pigs’ serum rather than vaccinated pigs’ serum as reported by Trible et al. (2011) and Trible and Rowland et al. (2012) [45,48]. The new PCV2 isolates identified in this study are labeled with red circles, the reference PCV2 clades are represented with blue triangles, and PCV2 available vaccine strains are indicated with the green diamonds.
Figure 1. Alignment of capsid protein sequences of thirteen PCV2 isolates: (A,B); The grey highlighted area represents antibody recognition domains as described by Trible et al. (2011) [45]. The red solid line boxes show motifs of PCV2 determined by Cheung et al. (2007) and Cheung et al. (2009) [5,43]. The dashed green line represents the location of the immunodominant decoy epitope, which shows reactivity of PCV2 infected pigs’ serum rather than vaccinated pigs’ serum as reported by Trible et al. (2011) and Trible and Rowland et al. (2012) [45,48]. The new PCV2 isolates identified in this study are labeled with red circles, the reference PCV2 clades are represented with blue triangles, and PCV2 available vaccine strains are indicated with the green diamonds.
Viruses 14 00289 g001aViruses 14 00289 g001b
Figure 2. Critical amino acid substitutions in ORF2 of various PCV2 genotypes, i.e., PCV2a, PCV2b, PCV2e, and PCV2d.
Figure 2. Critical amino acid substitutions in ORF2 of various PCV2 genotypes, i.e., PCV2a, PCV2b, PCV2e, and PCV2d.
Viruses 14 00289 g002
Figure 3. Phylogenetic analysis: phylogenetic tree was constructed by neighbor-joining (NJ) method for newly isolated 13 ORF2 sequences of PCV2 using MEGA5.0 software. Bootstrap values were calculated on 1000 replicates. The reference sequences of PCV2 clades are indicated with blue triangles. The new PCV2 isolates identified in this study are labeled with red circles and PCV2 available vaccine strains are indicated with green diamonds.
Figure 3. Phylogenetic analysis: phylogenetic tree was constructed by neighbor-joining (NJ) method for newly isolated 13 ORF2 sequences of PCV2 using MEGA5.0 software. Bootstrap values were calculated on 1000 replicates. The reference sequences of PCV2 clades are indicated with blue triangles. The new PCV2 isolates identified in this study are labeled with red circles and PCV2 available vaccine strains are indicated with green diamonds.
Viruses 14 00289 g003
Table 1. Primer sequence used in study for virus screening through PCR.
Table 1. Primer sequence used in study for virus screening through PCR.
Primer PairsPrimer SequenceAmplified LengthAnnealing TemperatureVirus Type
ORF5-F5′-AGGTGGGCAACTGTTTTAGC-3′697 bp53.5 °C PRRSV
ORF5-R5′-TTTGTGGAGCCGTGCTATCA-3′
VP2-F5′-CACGCATCAAGACTCATA-3′472 bp48.0 °CPPV
VP2-R5′-TTGGTGGATTTAGGTTTC-3′
PCV-F5′-CCGCGGGCTGGCTGAACTT-3′1154 bp58.0 °CPCV2
PCV-R5′-ACCCCCGCCACCGCTACC-3′
Table 2. Positive rate of PCV2 and its co-infection status with PRRSV and PPV from samples collected from various pig farms (Shanghai, China) between 2014 and 2018.
Table 2. Positive rate of PCV2 and its co-infection status with PRRSV and PPV from samples collected from various pig farms (Shanghai, China) between 2014 and 2018.
Samples’ IDFarm/AreaHerd
Size
Positive Samples/Total SamplesPCV2 Positive Rate (%)PCV2 + PRRSVPCV2 + PPVFarm
Vaccination
Status
SH150127-1Fengxian>5006/1060.0%0/66/6Yes
SH140624-5Pudong>300013/2161.9%0/1313/13Yes
SH140718-1Pudong>500010/1566.7%0/1010/10Yes
SH140704-1Jiading>200016/2759.3%0/1616/16Yes
SH140704-3Jiading>20009/1560.0%0/99/9Yes
SH140804-1Fengxian>200016/3151.6%0/3116/31Yes
SH140804-3Chongmei>200013/2065.0%0/1313/13Yes
SH140625-2Pudong>20004/757.1%4/44/4Yes
SH140625-1Fengxian>5004/850.0%2/44/4No
SH140625-3Chongmei>20007/1258.3%7/70/7Yes
SH1601Pudong>20008/1553.3%6/88/8Yes
SH1701Jiading>20006/1346.2%3/66/6Yes
SH1801Chongmei>20003/560.0%3/33/3Yes
Table 3. Summary of PCV2 (Cap gene) isolates from Shanghai pig farms during this study and their GenBank accession numbers.
Table 3. Summary of PCV2 (Cap gene) isolates from Shanghai pig farms during this study and their GenBank accession numbers.
Samples’ IDAccession Number (GenBank)YearPCV2 GenotypeHistory of DiseaseORF2 Size (bp)Tissues
SH150127-1KR05835920152dPMWS/PNDS705 Lung, spleen, lymph node
SH140624-5KR05835420142dPMWS/PNDS705 Lung, spleen, lymph nodes
SH140718-1KR05835820142dPMWS/PNDS705 Inguinal lymph nodes
SH140704-1KR05214520142dPMWS/PNDS705 Lung, spleen, lymph nodes
SH140704-3KR05214620142dNo signs705 Lung, spleen, lymph nodes
SH140804-1KR05835220142dPMWS/PNDS705 Inguinal lymph nodes
SH140804-3KR05835320142dPMWS/PNDS705 Inguinal lymph nodes
SH140625-2KR05835620142dPMWS/PNDS705 Lung, spleen, lymph nodes
SH140625-1KR05835520142bPMWS/PNDS702 Inguinal lymph nodes
SH140625-3KR05835720142eNo signs702 Inguinal lymph nodes
SH1601OM03262020162dPMWS/PNDS705Lung, spleen, lymph nodes
SH1701OM03262120172dPMWS/PNDS705Lung, spleen, lymph nodes
SH1801OM03262220182dPMWS/PNDS705Lung, spleen, lymph nodes
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kang, L.; Wahaab, A.; Shi, K.; Mustafa, B.E.; Zhang, Y.; Zhang, J.; Li, Z.; Qiu, Y.; Li, B.; Liu, K.; et al. Molecular Epidemic Characteristics and Genetic Evolution of Porcine Circovirus Type 2 (PCV2) in Swine Herds of Shanghai, China. Viruses 2022, 14, 289. https://doi.org/10.3390/v14020289

AMA Style

Kang L, Wahaab A, Shi K, Mustafa BE, Zhang Y, Zhang J, Li Z, Qiu Y, Li B, Liu K, et al. Molecular Epidemic Characteristics and Genetic Evolution of Porcine Circovirus Type 2 (PCV2) in Swine Herds of Shanghai, China. Viruses. 2022; 14(2):289. https://doi.org/10.3390/v14020289

Chicago/Turabian Style

Kang, Le, Abdul Wahaab, Kun Shi, Bahar E Mustafa, Yan Zhang, Junjie Zhang, Zongjie Li, Yafeng Qiu, Beibei Li, Ke Liu, and et al. 2022. "Molecular Epidemic Characteristics and Genetic Evolution of Porcine Circovirus Type 2 (PCV2) in Swine Herds of Shanghai, China" Viruses 14, no. 2: 289. https://doi.org/10.3390/v14020289

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop