Next Article in Journal
Electrocardiographic Reference Values in Clinically Healthy Lusitano Horses
Previous Article in Journal
How Does Epidemic Prevention Training for Pig Breeding Affect Cleaning and Disinfection Procedures Adoption? Evidence from Chinese Pig Farms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Genetic and In Vitro Characteristics of a Porcine Circovirus Type 3 Isolate from Northeast China

1
Heilongjiang Research Center for Veterinary Biopharmaceutical Technology, State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
2
College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
3
Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
4
Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Veterinary Medicine, Shandong Agricultural University, Tai’an 271002, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Vet. Sci. 2023, 10(8), 517; https://doi.org/10.3390/vetsci10080517
Submission received: 9 June 2023 / Revised: 1 August 2023 / Accepted: 2 August 2023 / Published: 10 August 2023
(This article belongs to the Section Veterinary Microbiology, Parasitology and Immunology)

Abstract

:

Simple Summary

Although several studies of porcine circovirus 3 (PCV3) have been conducted, there is little information regarding the pathogenicity of PCV3, in large part because of the small number of PCV3 isolates available. In this study, a PCV3 strain named PCV3-DB-1, belonging to the PCV3a branch, was isolated from PK-15 cells and with 24 alanine and 27 lysine in the Cap protein of this strain, which is unique. PCV-like particles were subsequently observed using electron microscopy and the RNA replication of PCV3 was confirmed in PK-15 cells using situ hybridization RNA scope. This is the first report on the isolation and in vitro characteristics of PCV3-DB-1 with 24A and 27K of the Cap protein. PCV3 was reported in 2015 on a sow farm experiencing PDNS-like and reproductive failure clinical symptoms in the United States, and was subsequently found in most swine-producing countries in Europe, Asia, and America. Although neurological disorders, as well as respiratory and enteric disease, have been described in association with PCV3 infection, multisystemic inflammation and reproductive failure are the most concerning clinical presentations. Studies reproducing and characterizing PCV3 clinical disease and pathogenesis under experimental conditions are scarce, in large part because of the difficulty of isolating PCV3. In this study, we isolated PCV3-DB-1 in PK-15 cells and characterized them in vitro. By using electron microscopy, PCV-like particles were discovered, and in situ hybridization RNA sequencing showed PCV3 replication in PK-15 cell culture. Based on phylogenetic analysis of PCV3 isolates from the Heilongjiang province of China, PCV3-DB-1 with 24 alanine and 27 lysine in the Cap protein was originally isolated and determined to belong to the clade PCV3a.

Abstract

Porcine circovirus 3 (PCV3) is an emerging virus first discovered in the United States in 2015, and since then, PCV3 has been found in many regions of the world, including America, Asia, and Europe. Although several PCV3 investigations have been carried out, there is a lack of knowledge regarding the pathogenicity of PCV3, mostly due to the limited number of PCV3 isolates that are readily available. In this study, PCV3-DB-1 was isolated in PK-15 cells and characterized in vitro. Electron microscopy revealed the presence of PCV-like particles, and in situ hybridization RNA analysis demonstrated the replication of PCV3 in PK-15 cell culture. Based on phylogenetic analysis of PCV3 isolates from the Heilongjiang province of China, PCV3-DB-1 with 24 alanine and 27 lysine in the Cap protein was originally isolated and determined to belong to the clade PCV3a.

1. Introduction

Circoviruses (CV), the smallest known autonomously replicating viruses, belong to the genus Circovirus in the Circoviridae family [1]. Circoviruses encode three to five open reading frames on opposite strands of a double-stranded DNA replicative intermediate [2,3]. Four porcine circoviruses (PCV) have been recognized to date: PCV1, PCV2, PCV3, and PCV4 [1,4,5,6,7]. PCV1 is not pathogenic in swine but is a common contaminant of the porcine kidney cell line (PK-15) [8]. PCV2 infection causes a range of clinical diseases, including post-weaning multisystemic wasting syndrome (PMWS), porcine dermatitis and nephropathy syndrome (PDNS), and reproductive failure [9]. PCV3 was reported in 2015 on a sow farm experiencing PDNS-like and reproductive failure clinical signs in the United States [5] and was subsequently found in most swine-producing countries in Europe, Asia, and America [10,11,12]. Both PCV2 and PCV3 were detected with high frequency in different tissues of wild boar [13,14], confirming that PCV2 and PCV3 may pose a persistent risk to the swine industry. PCV4 was detected in 2019 in China in association with clinical respiratory and enteric disease and PDNS [7].
More than ten provinces in China have reported the presence of PCV3 [15,16,17,18]. Subclinical PCV3 infection is characterized by viremia or detection of PCV3 in multiple tissues without evident clinical signs or lesions in pigs of all ages [19]. Although neurological disorders, as well as respiratory and enteric disease, have been described in association with PCV3 infection, multisystemic inflammation and reproductive failure are the most concerning clinical presentations. Studies reproducing and characterizing PCV3 clinical disease and pathogenesis under experimental conditions are scarce [20,21], in large part because of the difficulty of isolating PCV3. Hence, two studies have reported successful isolation of PCV3 [20,22], and only one provides further characterization of an isolate in vivo [20].
Significant advances associated with understanding the virus’s genetic structure and evolution have occurred since its first description. A consistent mutation in amino acids of the cap protein has been found through phylogenetic analyses of both recent and historical samples, which suggests that mutations in the amino acids at positions 24 and 27 of the cap protein could serve as molecular markers to divide PCV3 into the three clades PCV3a, PCV3b, and PCV3c [23]. In addition, PCV3a has been further subdivided into three subclades (PCV3a1, PCV3a2, and PCV3a3) based on evolutionary relationships and other molecular characteristics of the cap protein [23]. However, most of these reports were based on molecular detection and not viruses isolated from clinical cases. Thus, the association between the proposed clades and their phenotypic characteristics is incomplete in terms of pathogenicity. This report describes the genetic and in vitro characterization of the first reported PCV3 isolate from the Chinese province of Heilongjiang.

2. Materials and Methods

2.1. Virus Isolation

PCV3 (PCV3-China/DB-1/2017, MH286898) was isolated retrospectively from the lungs of two 10-day-old neonatal piglets in a commercial pig herd in the northeast Chinese province of Heilongjiang. Reported clinical signs were characterized by dyspnea, anorexia, lethargy, and an increment in >50% mortality. The presence of PCV2 [24], PRRSV [25], CSFV [26], PPV [27], swine influenza virus (AIV) [28], and Mycoplasma hyopneumoniae [29] infection was ruled out based on specific PCR detection. Specifically, the isolate was obtained via a single passage in a PCV1- and PCV2-free porcine kidney cell line (PK-15, ATCC® CCL-33™, Hong Kong, China). Lung tissue was processed as previously described [30]. Briefly, PK-15 cells were cultured in a 25 cm2 flask containing DMEM with 10% fetal bovine serum (Gibco, Invitrogen Corporation, Waltham, MA, USA), 100 U of penicillin/mL, and 100 μg streptomycin/mL at 37 °C and 5% CO2. When PK-15 cells reached ~40% confluency, the medium was removed, and the cells were washed three times with sterile phosphate-buffered saline (PBS, pH 7.4). Next, 2 mL of filtered supernatant was added and the sample was incubated at 37 °C and 5% CO2 for 2 h. The inoculum was replaced with a fresh medium with 2% fetal bovine serum, and the cells incubated for 72–96 h at 37 °C and 5% CO2. The virus was recovered through three freeze–thaw cycles and passaged three times in PK-15 cells. The cell lysate was confirmed to be free of PCV2 [24], PCV1 [31], classical swine fever virus [26], porcine parvovirus [27], and pseudorabies virus [32] using real-time PCR.

2.2. Evaluation of Viral Particles in PK-15 Cells Using Electron Microscopy

PK-15 cells were inoculated with ~1.18 × 105 genomic copies/mL of PCV3 DB-1, mock-infected with cell culture lysate in DMEM containing 2% fetal bovine serum (Gibco, Invitrogen Corporation), and incubated for 72–96 h at 37 °C and 5% CO2. Samples for electron microscopy studies were prepared according to the Veterinary Research Institute’s (Harbin, China) standard operating procedures. Briefly, the infected and mock-infected PK-15 cells were harvested and fixed with 2.5% (w/v) glutaraldehyde at 4 °C for 2 h and subsequently fixed with 1% OsO4. After being dehydrated in an acetone gradient, the samples were infiltrated using eponate resin at room temperature and embedded at 70 °C overnight for polymerization in beem capsules. Ultrathin sections 70 nm thick were cut with an Ultracut Eultramicrotome (Leica, Wetzlar, Germany) and stained with 2% uranyl acetate for 15 min and lead citrate for 10 min. After drying overnight, the samples were evaluated using conventional transmission electron microscopy (TEM, H7650, HITACHI, Tokyo, Japan).

2.3. Viral Characterization In Vitro through In Situ Hybridization (RNAscope) in Cell Culture

The PCV3 DB-1 isolate was passaged three times in PK-15 cells under the same conditions described above for virus isolation. After inoculation, the cells were incubated at 37 °C and 5% CO2 for 72–96 h and then fixed with 10% Neutral Buffered Formalin (NBF). According to the manufacturer’s instructions, viral replication was confirmed using a kit from Advanced Cell Diagnostics, Inc., (Hayward, CA, USA), RNAscope® 2.5 HD. The RNAscope® probes (catalog no. 463961 or 530431) targeting PCV3 RNA, along with the control probes, were designed and synthesized by Advanced Cell Diagnostics. Visualization of the samples was carried out with a microscope from Olympus Corporation, Tokyo, Japan.

2.4. Genome Sequence Analysis

Four pairs of primers were synthesized for complete genome amplification through PCR using methods previously reported [5]. Genotype identification of PCV3 was performed using phylogenetic tree deduction based on complete coding sequences (ORF1 + ORF2) [33]. To perform phylogenetic analysis, 58 PCV3 coding sequences originating from Sus scrofa (from NCBI) were downloaded. Alignment was carried out using default parameters in the Clustal W program (Lynnon Co., Zhuhai, China, DNAMAN 6.0 software). A phylogenic tree was constructed through the maximum likelihood method with 1000 bootstrap replicates in the MEGA program (v5.0) [34].

3. Results

The present study describes the retrospective isolation of PCV3 (PCV3 isolate DB-1) from clinically affected animals and its replication in PK-15 cells. Specifically, the PCV3 isolate DB-1 was cultured three times in PK-15 cells, and productive virus replication was confirmed using electron microscopy and ISH RNAscope® on virus obtained from each passage. The evaluation of PK-15 cells infected with PCV3 DB-1 using electron microscopy demonstrated the presence of PCV-like particles after a 72–96 h incubation at 37 °C and 5% CO2. Organized intracytoplasmic inclusion bodies (ICIs) 0.2–0.5 μm in diameter, forming large and complex electron-dense bodies, contained recognizable VLPs (Figure 1). No VLPs were observed in mock-infected cells. Likewise, virus replication in PK-15 cells was confirmed through ISH RNA scope on each passage. At 72–96 h of incubation, ISH revealed intracytoplasmic hybridization signals consistent with genome replication of PCV3 in vitro (Figure 2A,C,E).
Phylogenetic characterization showed that PCV3 DB-1 is a member of the PCV3a clade, but it is the only known PCV3 isolation of the amino acids 24 alanine (A) and 27 lysine (K) of the Cap protein in the NCBI database. Specifically, the nucleotide similarity between PCV3 DB-1 and the PCV3 sequences deposited in GenBank® (containing 58 sample sequences) was 97.3–99.7% (Figure 3). PCV3 DB-1 showed the highest (99.7%) and lowest (97.3%) nucleotide identities with PCV3/CN/Jiangxi-B1/2017 (MF589107) and PCV3/CN/Guangdong-SG1/2016 (MF589105), respectively.

4. Discussion

ICIs, structures that can be observed within the cytoplasm of infected cells, are often associated with viral infection and are the result of the interaction between the virus and host cell. In this study, PCV3-infected cells showed the presence of virus particles with the same size and morphology as porcine circovirus [8], forming large and complex electron-dense bodies. Similarly, PCV3-induced inclusion bodies appear as round or irregular structures within the cytoplasm of infected primary porcine kidney cells [22]. ICI observation in histopathological samples or cell cultures can support the identification and confirmation of PCV3’s presence. The exact composition of the inclusion bodies in PCV3-infected cells has not been fully characterized yet. The understanding of PCV3 and its intracytoplasmic inclusion bodies is still evolving, and further research is needed to fully characterize their composition, formation process, and diagnostic significance.
ISH RNAscope is a molecular technique used to detect and visualize specific RNA molecules within cells or tissue sections, providing insights into the cellular distribution and expression patterns of the target RNA. We confirmed the PCV3 replication in the PK-15 cells using ISH RNAscope and revealed intracytoplasmic hybridization signals consistent with the genome replication of PCV3 in vitro. Similarly, the signals generated by the PCV3 RNA probes indicate the presence and localization of PCV3 RNA within the primary porcine kidney cells [22]. ISH RNA scope allows researchers to directly visualize PCV3 RNA within the cellular context, providing valuable information about viral replication sites, tissue tropism, and cellular response to infection. It is a powerful tool for studying the pathogenesis and molecular biology characteristics of PCV3.
Phylogenetic analysis can identify distinct genotypes, lineages, or subgroups in the classification and clustering of PCV3 strains based on their genetic relatedness, which provides insights into the geographic distribution and evolutionary dynamics of PCV3. The genetic diversity of PCV3 seems to increase as the number of sequences reported from different countries does too; therefore, the current phylogenetic classification remains dynamic and subject to frequent changes. Nevertheless, PCV3 is a recently identified virus, but it has been found retrospectively since 1966 in China [23], 1967 in Brazil [35], 1996 in Spain [36], and 2006 in Thailand [37]. The cap protein of PCV3 exhibits genetic variation, with different strains or variants of the virus having slight differences in their cap protein sequences. These genetic variations may have implications for viral pathogenicity and antigenicity. Studying the cap protein of PCV3 is essential for understanding its structural properties, antigenicity, and potential role in virus–host interactions. Importantly, besides the phylogenetic differences observed in the field, there are no studies that support phenotypic differences amongst different clades. PCV3a has been associated with cases of reproductive failure [38,39], PDNS, and PCVAD [40]. Thus, further research is required to determine if members of the PCV3a clade with 24 alanine and 27 lysine within the Cap might have distinctive phenotypic characteristics that can be related to certain clinical manifestations.
In our study, immortalized cell lines (PK-15, Vero, ST, and DF1) and primary cells (PAM, PBMC, and bronchial epithelial cells) were used to isolate and passage the virus (Tables S1 and S2). The qPCR assay results showed that the PK-15 cell line was more suitable for the proliferation of PCV3-DB-1 isolation for a limited number of passages [41]. The screening of susceptible single-cell clones from the PK-15 cell line may be necessary for in vitro isolation of PCV3. PCV3 has circulated in the global swine population for several decades or more and will undoubtedly continue to do so in the future, bringing with it the potential for future clinical outbreaks. There is a broad spectrum of clinical signs associated with PCV3 infection, ranging from inapparent or subclinical infection to reproductive failure and PDNS-like signs in sows [5] and multisystemic and cardiac inflammation in grower–finisher pigs [42,43]. Interestingly, the experimental inoculation of CDCD piglets did not result in significant clinical disease and produced only mild histological changes associated with myocarditis and systemic perivasculitis [18,19]. It is known that the severity of the clinical disease induced by PCV2 infection is related, amongst other factors, to co-infections with other swine pathogens, including PRRSV, IAV, and PPV [44]. Whether PCV3-associated disease is related to the interactions and synergy of other infectious factors (co-infections), environmental factors related to production systems and husbandry, and pig-related factors like genetics, age, and immune status warrants further investigation.

5. Conclusions

In conclusion, this report describes the retrospective isolation of PCV3 in pigs with respiratory distress and its characterization in vitro. A phylogenic analysis demonstrated that PCV3a isolation with 24 alanine and 27 lysine of the Cap protein in this case is the first reported in the NCBI database of PCV3 so far. Although numerous studies have reported PCV3 in swine production systems worldwide, a complete understanding of the pathogenesis of PCV3 and the risk factors that trigger different clinical presentations in the field requires further investigation.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/vetsci10080517/s1, Table S1: Detection of PCV3-DB-1 passage in cell lines by qPCR; Table S2: Detection of PCV3-DB-1 passage in primary cells by qPCR.

Author Contributions

G.W., X.C. and Y.T. conceived the study. M.W., Y.Y., J.W., S.W., Y.W., H.C., X.H. and G.W. performed the experiments. M.W. and G.W. analyzed the data and wrote the manuscript. G.W., L.G.G.-L., P.P. and J.J.Z. revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant from the State Key Laboratory of Veterinary Biotechnology Foundation (grant number SKLVBP2018002), the Central Public-interest Scientific Institution Basal Research Fund (NO. 1610302022006), and the Ph.D. foundation of Qingdao Agricultural University, China (6631120019).

Institutional Review Board Statement

The sample collected were approved by the Animal Ethics Committee of Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences (CAAS) with approval number SY-2018-SW-003.

Informed Consent Statement

Informed consent was obtained from all subjects involved in this study.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rosario, K.; Breitbart, M.; Harrach, B.; Segales, J.; Delwart, E.; Biagini, P.; Varsani, A. Revisiting the taxonomy of the family Circoviridae: Establishment of the genus Cyclovirus and removal of the genus Gyrovirus. Arch. Virol. 2017, 162, 1447–1463. [Google Scholar] [CrossRef]
  2. Mankertz, A.; Mankertz, J.; Wolf, K.; Buhk, H.J. Identification of a protein essential for replication of porcine circovirus. J. Gen. Virol. 1998, 79 Pt 2, 381–384. [Google Scholar] [CrossRef]
  3. 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]
  4. Allan, G.M.; Mc Neilly, F.; Meehan, B.M.; Kennedy, S.; Mackie, D.P.; Ellis, J.A.; Clark, E.G.; Espuna, E.; Saubi, N.; Riera, P.; et al. Isolation and characterisation of circoviruses from pigs with wasting syndromes in Spain, Denmark and Northern Ireland. Vet. Microbiol. 1999, 66, 115–123. [Google Scholar] [CrossRef]
  5. Palinski, R.; Pineyro, P.; Shang, P.; Yuan, F.; Guo, R.; Fang, Y.; Byers, E.; Hause, B.M. A Novel Porcine Circovirus Distantly Related to Known Circoviruses Is Associated with Porcine Dermatitis and Nephropathy Syndrome and Reproductive Failure. J. Virol. 2017, 91, e01879-16. [Google Scholar] [CrossRef]
  6. Tischer, I.; Rasch, R.; Tochtermann, G. Characterization of papovavirus-and picornavirus-like particles in permanent pig kidney cell lines. Zentralbl. Bakteriol. Orig. A 1974, 226, 153–167. [Google Scholar]
  7. Zhang, H.H.; Hu, W.Q.; Li, J.Y.; Liu, T.N.; Zhou, J.Y.; Opriessnig, T.; Xiao, C.T. Novel circovirus species identified in farmed pigs designated as Porcine circovirus 4, Hunan province, China. Transbound. Emerg. Dis. 2020, 67, 1057–1061. [Google Scholar] [CrossRef]
  8. Tischer, I.; Gelderblom, H.; Vettermann, W.; Koch, M.A. A very small porcine virus with circular single-stranded, D.N.A. Nature 1982, 295, 64–66. [Google Scholar] [CrossRef]
  9. Segales J Porcine circovirus type 2 (PCV2) infections: Clinical signs, pathology and laboratory diagnosis. Virus Res. 2012, 164, 10–19. [CrossRef]
  10. Franzo, G.; Tucciarone, C.M.; Drigo, M.; Cecchinato, M.; Martini, M.; Mondin, A.; Menandro, M.L. First report of wild boar susceptibility to Porcine circovirus type 3: High prevalence in the Colli Euganei Regional Park (Italy) in the absence of clinical signs. Transbound. Emerg. Dis. 2018, 65, 957–962. [Google Scholar] [CrossRef]
  11. Kwon, T.; Yoo, S.J.; Park, C.K.; Lyoo, Y.S. Prevalence of novel porcine circovirus 3 in Korean pig populations. Vet. Microbiol. 2017, 207, 178–180. [Google Scholar] [CrossRef]
  12. Tochetto, C.; Lima, D.A.; Varela, A.P.M.; Loiko, M.R.; Paim, W.P.; Scheffer, C.M.; Herpich, J.I.; Cerva, C.; Schmitd, C.; Cibulski, S.P.; et al. Full-Genome Sequence of Porcine Circovirus type 3 recovered from serum of sows with stillbirths in Brazil. Transbound. Emerg. Dis. 2018, 65, 5–9. [Google Scholar] [CrossRef]
  13. Amoroso, M.G.; Serra, F.; Esposito, C.; D’Alessio, N.; Ferrara, G.; Cioffi, B.; Anzalone, A.; Pagnini, U.; De Carlo, E.; Fusco, G.; et al. Prevalence of Infection with Porcine Circovirus Types 2 and 3 in the Wild Boar Population in the Campania Region (Southern Italy). Animals 2021, 11, 3215. [Google Scholar] [CrossRef]
  14. Fanelli, A.; Pellegrini, F.; Camero, M.; Catella, C.; Buonavoglia, D.; Fusco, G.; Martella, V.; Lanave, G. Genetic Diversity of Porcine Circovirus Types 2 and 3 in Wild Boar in Italy. Animals 2022, 12, 953. [Google Scholar] [CrossRef]
  15. Chen, G.H.; Mai, K.J.; Zhou, L.; Wu, R.T.; Tang, X.Y.; Wu, J.L.; He, L.L.; Lan, T.; Xie, Q.M.; Sun, Y.; et al. Detection and genome sequencing of porcine circovirus 3 in neonatal pigs with congenital tremors in South China. Transbound. Emerg. Dis. 2017, 64, 1650–1654. [Google Scholar] [CrossRef]
  16. Ku, X.; Chen, F.; Li, P.; Wang, Y.; Yu, X.; Fan, S.; Qian, P.; Wu, M.; He, Q. Identification and genetic characterization of porcine circovirus type 3 in China. Transbound. Emerg. Dis. 2017, 64, 703–708. [Google Scholar] [CrossRef]
  17. Wen, S.; Sun, W.; Li, Z.; Zhuang, X.; Zhao, G.; Xie, C.; Zheng, M.; Jing, J.; Xiao, P.; Wang, M.; et al. The detection of porcine circovirus 3 in Guangxi, China. Transbound. Emerg. Dis. 2018, 65, 27–31. [Google Scholar] [CrossRef]
  18. Zhai, S.L.; Zhou, X.; Zhang, H.; Hause, B.M.; Lin, T.; Liu, R.; Chen, Q.L.; Wei, W.K.; Lv, D.H.; Wen, X.H.; et al. Comparative epidemiology of porcine circovirus type 3 in pigs with different clinical presentations. Virol. J. 2017, 14, 222. [Google Scholar] [CrossRef]
  19. Klaumann, F.; Correa-Fiz, F.; Sibila, M.; Nunez, J.I.; Segales, J. Infection dynamics of porcine circovirus type 3 in longitudinally sampled pigs from four Spanish farms. Vet. Rec. 2019, 184, 619. [Google Scholar] [CrossRef]
  20. Mora-Diaz, J.; Pineyro, P.; Shen, H.; Schwartz, K.; Vannucci, F.; Li, G.; Arruda, B.; Gimenez-Lirola, L. Isolation of PCV3 from Perinatal and Reproductive Cases of PCV3-Associated Disease and In Vivo Characterization of PCV3 Replication in CD/CD Growing Pigs. Viruses 2020, 12, 219. [Google Scholar] [CrossRef]
  21. Temeeyasen, G.; Lierman, S.; Arruda, B.L.; Main, R.; Vannucci, F.; Gimenez-Lirola, L.G.; Pineyro, P.E. Pathogenicity and immune response against porcine circovirus type 3 infection in caesarean-derived, colostrum-deprived pigs. J. Gen. Virol. 2021, 102. [Google Scholar] [CrossRef]
  22. Oh, T.; Chae, C. First isolation and genetic characterization of porcine circovirus type 3 using primary porcine kidney cells. Vet. Microbiol. 2020, 241, 108576. [Google Scholar] [CrossRef]
  23. Fu, X.; Fang, B.; Ma, J.; Liu, Y.; Bu, D.; Zhou, P.; Wang, H.; Jia, K.; Zhang, G. Insights into the epidemic characteristics and evolutionary history of the novel porcine circovirus type 3 in southern China. Transbound. Emerg. Dis. 2018, 65, e296–e303. [Google Scholar] [CrossRef] [PubMed]
  24. Caprioli, A.; McNeilly, F.; McNair, I.; Lagan-Tregaskis, P.; Ellis, J.; Krakowka, S.; McKillen, J.; Ostanello, F.; Allan, G. PCR detection of porcine circovirus type 2 (PCV2) DNA in blood, tonsillar and faecal swabs from experimentally infected pigs. Res. Vet. Sci. 2006, 81, 287–292. [Google Scholar] [CrossRef]
  25. Yim-Im, W.; Huang, H.; Zheng, Y.; Li, G.; Rawal, G.; Gauger, P.; Krueger, K.; Main, R.; Zhang, J. Characterization of PRRSV in clinical samples and the corresponding cell culture isolates. Transbound. Emerg. Dis. 2022, 69, e3045–e3059. [Google Scholar] [CrossRef] [PubMed]
  26. Yang, Z.Z.; Fang, W.H.; Habib, M. First results of detection of PRRSV and CSFV RNA by SYBR Green I-based quantitative, P.C.R. J. Vet. Med. B Infect Dis. Vet. Public Health 2006, 53, 461–467. [Google Scholar] [CrossRef]
  27. Prikhod’ko, G.G.; Reyes, H.; Vasilyeva, I.; Busby, T.F. Establishment of a porcine parvovirus (PPV) DNA standard and evaluation of a new lightcycler nested-PCR assay for detection of, P.P.V. J. Virol. Methods 2003, 111, 13–19. [Google Scholar] [CrossRef]
  28. Spackman, E. Avian Influenza Virus Detection and Quantitation by Real-Time RT-PCR. Methods Mol. Biol. 2020, 2123, 137–148. [Google Scholar]
  29. Takeuti, K.L.; Michaelsen, T.R.; Sabedot, C.; Nagae, R.Y.; Forner, R.A.N.; Mazzarollo, A.; de Barcellos, D.E.S.N.; Pieters, M. Mycoplasma hyopneumoniae detection by PCR in naturally infected finishing pigs. J. Microbiol. Methods 2022, 197, 106475. [Google Scholar] [CrossRef]
  30. Wang, H.M.; Liu, Y.G.; Tang, Y.D.; Liu, T.X.; Zheng, L.L.; Wang, T.Y.; Liu, S.G.; Wang, G.; Cai, X.H. A natural recombinant PRRSV between HP-PRRSV JXA1-like and NADC30-like strains. Transbound. Emerg. Dis. 2018, 65, 1078–1086. [Google Scholar] [CrossRef] [PubMed]
  31. Li, J.; Shi, J.L.; Wu, X.Y.; Cong, X.Y.; Xu, S.J.; Yuan, X.Y.; Wu, J.Q.; Sun, W.B.; Du, Y.J.; Peng, Z.; et al. Differentiation of PCV1 and PCV2 by a multiplex real-time PCR assay. Vet. Rec. 2013, 173, 346. [Google Scholar] [CrossRef] [PubMed]
  32. Aytogu, G.; Toker, E.B.; Yavas, O.; Kadiroglu, B.; Ates, O.; Ozyigit, M.O.; Yesilbag, K. First isolation and molecular characterization of pseudorabies virus detected in Turkey. Mol. Biol. Rep. 2022, 49, 1679–1686. [Google Scholar] [CrossRef] [PubMed]
  33. Li, G.; He, W.; Zhu, H.; Bi, Y.; Wang, R.; Xing, G.; Zhang, C.; Zhou, J.; Yuen, K.Y.; Gao, G.F.; et al. Origin, Genetic Diversity, and Evolutionary Dynamics of Novel Porcine Circovirus 3. Adv. Sci. 2018, 5, 1800275. [Google Scholar] [CrossRef]
  34. 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] [PubMed]
  35. Rodrigues, I.L.F.; Cruz, A.C.M.; Souza, A.E.; Knackfuss, F.B.; Costa, C.H.C.; Silveira, R.L.; Castro, T.X. Retrospective study of porcine circovirus 3 (PCV3) in swine tissue from Brazil (1967–2018). Braz. J. Microbiol. 2020, 51, 1391–1397. [Google Scholar] [CrossRef]
  36. Klaumann, F.; Franzo, G.; Sohrmann, M.; Correa-Fiz, F.; Drigo, M.; Nunez, J.I.; Sibila, M.; Segales, J. Retrospective detection of Porcine circovirus 3 (PCV-3) in pig serum samples from Spain. Transbound. Emerg. Dis. 2018, 65, 1290–1296. [Google Scholar] [CrossRef]
  37. Sukmak, M.; Thanantong, N.; Poolperm, P.; Boonsoongnern, A.; Ratanavanichrojn, N.; Jirawattanapong, P.; Woonwong, Y.; Soda, N.; Kaminsonsakul, T.; Phuttapatimok, S. Wajjwalku W The retrospective identification and molecular epidemiology of porcine circovirus type 3 (PCV3) in swine in Thailand from 2006 to 2017. Transbound. Emerg. Dis. 2019, 66, 611–616. [Google Scholar] [CrossRef]
  38. Bera, B.C.; Choudhary, M.; Anand, T.; Virmani, N.; Sundaram, K.; Choudhary, B.; Tripathi, B.N. Detection and genetic characterization of porcine circovirus 3 (PCV3) in pigs in India. Transbound. Emerg. Dis. 2020, 67, 1062–1067. [Google Scholar] [CrossRef]
  39. Serena, M.S.; Cappuccio, J.A.; Barrales, H.; Metz, G.E.; Aspitia, C.G.; Lozada, I.; Perfumo, C.J.; Quiroga, M.A.; Pineyro, P.; Echeverria, M.G. First detection and genetic characterization of porcine circovirus type 3 (PCV3) in Argentina and its association with reproductive failure. Transbound. Emerg. Dis. 2020, 68, 1761–1766. [Google Scholar] [CrossRef]
  40. Vargas-Bermudez, D.S.; Campos, F.S.; Bonil, L.; Mogollon, D.; Jaime, J. First detection of porcine circovirus type 3 in Colombia and the complete genome sequence demonstrates the circulation of PCV3a1 and PCV3a2. Vet. Med. Sci. 2019, 5, 182–188. [Google Scholar] [CrossRef]
  41. Wang, Y.; Feng, Y.; Zheng, W.; Noll, L.; Porter, E.; Potter, M.; Cino, G.; Peddireddi, L.; Liu, X.; Anderson, G.; et al. A multiplex real-time PCR assay for the detection and differentiation of the newly emerged porcine circovirus type 3 and continuously evolving type 2 strains in the United States. J. Virol. Methods 2019, 269, 7–12. [Google Scholar] [CrossRef]
  42. Phan, T.G.; Giannitti, F.; Rossow, S.; Marthaler, D.; Knutson, T.P.; Li, L.; Deng, X.; Resende, T.; Vannucci, F.; Delwart, E. Detection of a novel circovirus PCV3 in pigs with cardiac and multi-systemic inflammation. Virol. J. 2016, 13, 184. [Google Scholar] [CrossRef] [PubMed]
  43. Arruda, B.; Pineyro, P.; Derscheid, R.; Hause, B.; Byers, E.; Dion, K.; Long, D.; Sievers, C.; Tangen, J.; Williams, T.; et al. PCV3-associated disease in the United States swine herd. Emerg. Microbes Infect 2019, 8, 684–698. [Google Scholar] [CrossRef] [PubMed]
  44. Allan, G.M.; McNeilly, F.; Ellis, J.; Krakowka, S.; Meehan, B.; McNair, I.; Walker, I.; Kennedy, S. Experimental infection of colostrum deprived piglets with porcine circovirus 2 (PCV2) and porcine reproductive and respiratory syndrome virus (PRRSV) potentiates PCV2 replication. Arch. Virol. 2000, 145, 2421–2429. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Electron micrographs of PCV3-infected PK-15 cells at 72 h post-inoculation. Viral-like particles are grouped in paracrystalline arrays within a large, complex intracytoplasmic inclusion (ICI) body (insert at higher magnification). Scale bar: 1 μm.
Figure 1. Electron micrographs of PCV3-infected PK-15 cells at 72 h post-inoculation. Viral-like particles are grouped in paracrystalline arrays within a large, complex intracytoplasmic inclusion (ICI) body (insert at higher magnification). Scale bar: 1 μm.
Vetsci 10 00517 g001
Figure 2. Isolation and identification of PCV3 DB-1 in PK-15-infected cells. Brown positive signals were found in the cytoplasm of PK-15-infected cells of PCV3 DB-1 isolates via ISH RNA scope at 10× magnification (A), 20× magnification (C), and 40× magnification (E). No positive signals were detected in the mock-inoculated PK-15 cells (B,D,F).
Figure 2. Isolation and identification of PCV3 DB-1 in PK-15-infected cells. Brown positive signals were found in the cytoplasm of PK-15-infected cells of PCV3 DB-1 isolates via ISH RNA scope at 10× magnification (A), 20× magnification (C), and 40× magnification (E). No positive signals were detected in the mock-inoculated PK-15 cells (B,D,F).
Vetsci 10 00517 g002
Figure 3. Phylogenetic analysis based on the complete coding sequences of the PCV3 isolates. MEGA 5.0 was used to reconstruct the ML tree using 1000 bootstrap trials. Bootstrap values are represented by the values along the branches.
Figure 3. Phylogenetic analysis based on the complete coding sequences of the PCV3 isolates. MEGA 5.0 was used to reconstruct the ML tree using 1000 bootstrap trials. Bootstrap values are represented by the values along the branches.
Vetsci 10 00517 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, M.; Yu, Y.; Wu, J.; Wang, S.; Giménez-Lirola, L.G.; Piñeyro, P.; Wang, Y.; Cui, H.; He, X.; Zimmerman, J.J.; et al. Genetic and In Vitro Characteristics of a Porcine Circovirus Type 3 Isolate from Northeast China. Vet. Sci. 2023, 10, 517. https://doi.org/10.3390/vetsci10080517

AMA Style

Wang M, Yu Y, Wu J, Wang S, Giménez-Lirola LG, Piñeyro P, Wang Y, Cui H, He X, Zimmerman JJ, et al. Genetic and In Vitro Characteristics of a Porcine Circovirus Type 3 Isolate from Northeast China. Veterinary Sciences. 2023; 10(8):517. https://doi.org/10.3390/vetsci10080517

Chicago/Turabian Style

Wang, Menghang, Ying Yu, Jianan Wu, Shujie Wang, Luis G Giménez-Lirola, Pablo Piñeyro, Yu Wang, Hongliang Cui, Xijun He, Jeffrey J. Zimmerman, and et al. 2023. "Genetic and In Vitro Characteristics of a Porcine Circovirus Type 3 Isolate from Northeast China" Veterinary Sciences 10, no. 8: 517. https://doi.org/10.3390/vetsci10080517

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